1
|
Farooq A, Drotleff B, Kroemer N, Han ML, Li J, Decousser JW, Schrey D, Buyck J, Grégoire N, Nordmann P, Wicha SG. Evaluation of in vitro pharmacodynamic drug interactions of ceftazidime-avibactam with tigecycline in ESBL- and carbapenemase producing Escherichia coli and Klebsiella pneumoniae. Int J Antimicrob Agents 2025; 65:107457. [PMID: 39894062 DOI: 10.1016/j.ijantimicag.2025.107457] [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: 10/09/2024] [Revised: 01/09/2025] [Accepted: 01/23/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND Combination therapy offers a promising option to enhance efficacy and prevent resistance. A comprehensive and quantitative assessment of the last-resort combination of ceftazidime/avibactam and tigecycline is not available. OBJECTIVE This study systematically investigated the pharmacodynamic interaction between ceftazidime/avibactam and tigecycline in clinical and isogenic Escherichia coli and Klebsiella pneumoniae strains harbouring genes that encode various carbapenemases or ESBLs. METHODS An adaptive in vitro 'dynamic' checkerboard design and pharmacometric modelling were employed for the evaluation of pharmacodynamic interactions in fifteen bacterial isolates. Additionally, time-kill assays and metabolomic analyses were used to provide mechanistic insights. RESULTS Antagonistic drug interactions between ceftazidime/avibactam and tigecycline were identified in the majority of tested strains. Time-kill assays confirmed antagonistic interactions, with tigecycline limiting ceftazidime/avibactam total killing. Metabolomic analyses of mono and combined drug exposure to bacteria revealed matching metabolomes in tigecycline alone and the combination with ceftazidime/avibactam, corroborating the identified antagonism between these drugs. CONCLUSIONS Our study reveals that the antagonistic interaction between ceftazidime/avibactam and tigecycline can undermine ceftazidime/avibactam's efficacy, suggesting limited clinical benefit in combining these antibiotics. Therefore, further research is encouraged to explore this and alternative combinations or approaches that may offer better clinical outcomes.
Collapse
Affiliation(s)
- Aneeq Farooq
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| | - Bernhard Drotleff
- Metabolomics Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Niklas Kroemer
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| | - Mei-Ling Han
- Department of Microbiology, Biomedicine Discovery Institute, Infection and Immunity Program, Monash University, Clayton, Victoria, Australia
| | - Jian Li
- Department of Microbiology, Biomedicine Discovery Institute, Infection and Immunity Program, Monash University, Clayton, Victoria, Australia
| | - Jean Winoc Decousser
- Dynamic Team - EA 7380, Faculté de Santé, Université Paris-Est-Créteil Val-De-Marne, Créteil, France
| | - David Schrey
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| | - Julien Buyck
- Inserm U1070, University of Poitiers, Poitiers, France
| | | | - Patrice Nordmann
- Medical and Molecular Microbiology, University of Fribourg, Fribourg, Switzerland
| | - Sebastian G Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Hamburg, Germany.
| |
Collapse
|
2
|
Maiga M, Dembele L, Courlet P, Khandelwal A, Dara A, Sogore F, Diakité O, Maiga FO, Dao F, Sissoko S, Barre Y, Goita S, Diakite M, Diakite SAS, Djimde AA, Oeuvray C, Spangenberg T, Wicha SG, Demarta-Gatsi C. Towards clinically relevant dose ratios for Cabamiquine and Pyronaridine combination using P. falciparum field isolate data. Nat Commun 2024; 15:7659. [PMID: 39227370 PMCID: PMC11372057 DOI: 10.1038/s41467-024-51994-3] [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: 02/15/2024] [Accepted: 08/22/2024] [Indexed: 09/05/2024] Open
Abstract
The selection and combination of dose regimens for antimalarials involve complex considerations including pharmacokinetic and pharmacodynamic interactions. In this study, we use immediate ex vivo P. falciparum field isolates to evaluate the effect of cabamiquine and pyronaridine as standalone treatments and in combination therapy. We feed the data into a pharmacometrics model to generate an interaction map and simulate meaningful clinical dose ratios. We demonstrate that the pharmacometrics model of parasite growth and killing provides a detailed description of parasite kinetics against cabamiquine-susceptible and resistant parasites. Pyronaridine monotherapy provides suboptimal killing rates at doses as high as 720 mg. In contrast, the combination of a single dose of 330 mg cabamiquine and 360 mg pyronaridine provides over 90% parasite killing in most of the simulated patients. The described methodology that combines a rapid, 3R-compliant in vitro method and modelling to set meaningful doses for new antimalarials could contribute to clinical drug development.
Collapse
Affiliation(s)
- Mohamed Maiga
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Laurent Dembele
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali.
| | - Perrine Courlet
- Merck Institute of Pharmacometrics (an affiliate of Merck KGaA), Lausanne, Switzerland
| | - Akash Khandelwal
- The Healthcare Business of Merck KGaA, Darmstadt, Germany
- UCB Biosciences GmbH, Rolf-Schwarz-Schütte-Platz 1, Monheim am Rhein, Germany
| | - Antoine Dara
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Fanta Sogore
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Ousmaila Diakité
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Fatoumata O Maiga
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - François Dao
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Sekou Sissoko
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Yacouba Barre
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Siaka Goita
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Mahamadou Diakite
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Seidina A S Diakite
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Abdoulaye A Djimde
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
- Pathogens genomic Diversity Network Africa, Sotuba, Bamako, Mali
| | - Claude Oeuvray
- Global Health R&D of the healthcare business of Merck KGaA, Darmstadt, Germany, Ares Trading S.A. (an affiliate of Merck KGaA, Darmstadt, Germany), Eysins, Switzerland
| | - Thomas Spangenberg
- Global Health R&D of the healthcare business of Merck KGaA, Darmstadt, Germany, Ares Trading S.A. (an affiliate of Merck KGaA, Darmstadt, Germany), Eysins, Switzerland
| | - Sebastian G Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Bundesstr. 45, Hamburg, Germany.
| | - Claudia Demarta-Gatsi
- Pathogens genomic Diversity Network Africa, Sotuba, Bamako, Mali.
- Global Health R&D of the healthcare business of Merck KGaA, Darmstadt, Germany, Ares Trading S.A. (an affiliate of Merck KGaA, Darmstadt, Germany), Eysins, Switzerland.
| |
Collapse
|
3
|
Steyl SK, Jeyapalina S, Griffin A, Krishnamoorthi V, Beck JP, Agarwal J, Shea J. Efficacy of sintered Zinc-doped fluorapatite scaffold as an antimicrobial regenerative bone filler for dental applications. J Dent 2024; 146:105070. [PMID: 38740251 PMCID: PMC11180563 DOI: 10.1016/j.jdent.2024.105070] [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: 02/15/2024] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024] Open
Abstract
OBJECTIVES The objective of this study was to assess whether zinc-doped fluorapatite (ZnFA) could serve as an effective antimicrobial dental bone filler for bone regeneration compared to autografts. METHODS FA and 2 % zinc-doped FA (2ZnFA) were synthesized and characterized in-house. Compressed and sintered FA and 2ZnFA disks were incubated with bacteria to assess antimicrobial properties. Adipose-derived stem cells were cultured on these discs to evaluate the surfaces' ability to support cell growth and promote osteogenic differentiation. Surfaces exhibiting the highest expressions of the bone markers osteopontin and osteocalcin were selected for an in vivo study in a rat mandibular defect model. Twenty rats were divided into 5 groups, equally, and a 5 mm surgical defect of the jaw was left untreated or filled with 2ZnFA, FA, autograft, or demineralized bone matrix (DBM). At 12 weeks, the defects and surrounding tissues were harvested and subjected to microCT and histological evaluations. RESULTS Standard techniques such as FTIR, ICP-MS, fluoride probe, and XRD revealed the sintered FA and ZnFA's chemical compositions and structures. Bacterial studies revealed no significant differences in surface bacterial adhesion properties between FA and 2ZnFA, but significantly fewer bacterial loads than control titanium discs (p < 0.05). Cell culture data confirmed that both surfaces could support cell growth and promote the osteogenic differentiation of stem cells. MicroCT analysis confirmed statistical similarities in bone regeneration within FA, 2ZnFA, and autograft groups. CONCLUSION The data suggests that both FA and 2ZnFA could serve as alternatives to autograft materials, which are the current gold standard. Moreover, these bone fillers outperformed DBM, an allograft material commonly used as a dental bone void filler. CLINICAL SIGNIFICANCE The use of FA or 2ZnFA for treating mandibular defects led to bone regeneration statistically similar to autograft repair and significantly outperformed the widely used dental bone filler, DBM. Additional translational research may confirm FA-based materials as superior substitutes for existing synthetic bone fillers, ultimately enhancing patient outcomes.
Collapse
Affiliation(s)
- Samantha K Steyl
- Orthopaedic and Plastic Surgery Research Laboratory, George E. Wahlen Department of Veterans Affairs Medical Center, 500 Foothill Drive Salt Lake City, UT 84148, USA; Division of Plastic Surgery, Department of Surgery, University of Utah School of Medicine, 30 North 1900 East, Salt Lake City, UT 84132, USA; Department of Biomedical Engineering, University of Utah, 36 S. Wasatch Drive, Rm. 3100. Salt Lake City, UT 84112, USA
| | - Sujee Jeyapalina
- Orthopaedic and Plastic Surgery Research Laboratory, George E. Wahlen Department of Veterans Affairs Medical Center, 500 Foothill Drive Salt Lake City, UT 84148, USA; Division of Plastic Surgery, Department of Surgery, University of Utah School of Medicine, 30 North 1900 East, Salt Lake City, UT 84132, USA; Department of Biomedical Engineering, University of Utah, 36 S. Wasatch Drive, Rm. 3100. Salt Lake City, UT 84112, USA
| | - Alec Griffin
- Orthopaedic and Plastic Surgery Research Laboratory, George E. Wahlen Department of Veterans Affairs Medical Center, 500 Foothill Drive Salt Lake City, UT 84148, USA
| | - Vishnu Krishnamoorthi
- Orthopaedic and Plastic Surgery Research Laboratory, George E. Wahlen Department of Veterans Affairs Medical Center, 500 Foothill Drive Salt Lake City, UT 84148, USA
| | - James Peter Beck
- Orthopaedic and Plastic Surgery Research Laboratory, George E. Wahlen Department of Veterans Affairs Medical Center, 500 Foothill Drive Salt Lake City, UT 84148, USA; Department of Orthopaedics, University of Utah School of Medicine, 590 Wakara Way Salt Lake City, UT 84108, USA
| | - Jay Agarwal
- Orthopaedic and Plastic Surgery Research Laboratory, George E. Wahlen Department of Veterans Affairs Medical Center, 500 Foothill Drive Salt Lake City, UT 84148, USA; Division of Plastic Surgery, Department of Surgery, University of Utah School of Medicine, 30 North 1900 East, Salt Lake City, UT 84132, USA
| | - Jill Shea
- Orthopaedic and Plastic Surgery Research Laboratory, George E. Wahlen Department of Veterans Affairs Medical Center, 500 Foothill Drive Salt Lake City, UT 84148, USA; Division of Plastic Surgery, Department of Surgery, University of Utah School of Medicine, 30 North 1900 East, Salt Lake City, UT 84132, USA; Department of Biomedical Engineering, University of Utah, 36 S. Wasatch Drive, Rm. 3100. Salt Lake City, UT 84112, USA.
| |
Collapse
|
4
|
Taha AM, Elmasry MS, Hassan WS, Sayed RA. Spider chart, greenness and whiteness assessment of experimentally designed multivariate models for simultaneous determination of three drugs used as a combinatory antibiotic regimen in critical care units: Comparative study. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 313:124115. [PMID: 38484641 DOI: 10.1016/j.saa.2024.124115] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/24/2024] [Accepted: 03/02/2024] [Indexed: 04/02/2024]
Abstract
In this study, five earth-friendly spectrophotometric methods using multivariate techniques were developed to analyze levofloxacin, linezolid, and meropenem, which are utilized in critical care units as combination therapies. These techniques were used to determine the mentioned medications in laboratory-prepared mixtures, pharmaceutical products and spiked human plasma that had not been separated before handling. These methods were named classical least squares (CLS), principal component regression (PCR), partial least squares (PLS), genetic algorithm partial least squares (GA-PLS), and artificial neural network (ANN). The methods used a five-level, three-factor experimental design to make different concentrations of the antibiotics mentioned (based on how much of them are found in the plasma of critical care patients and their linearity ranges). The approaches used for levofloxacin, linezolid, and meropenem were in the ranges of 3-15, 8-20, and 5-25 µg/mL, respectively. Several analytical tools were used to test the proposed methods' performance. These included the root mean square error of prediction, the root mean square error of cross-validation, percentage recoveries, standard deviations, and correlation coefficients. The outcome was highly satisfactory. The study found that the root mean square errors of prediction for levofloxacin were 0.090, 0.079, 0.065, 0.027, and 0.001 for the CLS, PCR, PLS, GA-PLS, and ANN models, respectively. The corresponding values for linezolid were 0.127, 0.122, 0.108, 0.05, and 0.114, respectively. For meropenem, the values were 0.230, 0.222, 0.179, 0.097, and 0.099 for the same models, respectively. These results indicate that the developed models were highly accurate and precise. This study compared the efficiency of artificial neural networks and classical chemometric models in enhancing spectral data selectivity for quickly identifying three antimicrobials. The results from these five models were subjected to statistical analysis and compared with each other and with the previously published ones. Finally, the whiteness of the methods was assessed by the recently published white analytical chemistry (WAC) RGB 12, and the greenness of the proposed methods was assessed using AGREE, GAPI, NEMI, Raynie and Driver, and eco-scale, which showed that the suggested approaches had the least negative environmental impact. Furthermore, to demonstrate solvent sustainability, a greenness index using a spider chart methodology was employed.
Collapse
Affiliation(s)
- Asmaa M Taha
- Analytical Chemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt.
| | - Manal S Elmasry
- Analytical Chemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Wafaa S Hassan
- Analytical Chemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Rania A Sayed
- Analytical Chemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
5
|
Xu S, Esmaeili S, Cardozo-Ojeda EF, Goyal A, White JM, Polyak SJ, Schiffer JT. Two-way pharmacodynamic modeling of drug combinations and its application to pairs of repurposed Ebola and SARS-CoV-2 agents. Antimicrob Agents Chemother 2024; 68:e0101523. [PMID: 38470112 PMCID: PMC10989026 DOI: 10.1128/aac.01015-23] [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: 08/16/2023] [Accepted: 02/20/2024] [Indexed: 03/13/2024] Open
Abstract
Existing pharmacodynamic (PD) mathematical models for drug combinations discriminate antagonistic, additive, multiplicative, and synergistic effects, but fail to consider how concentration-dependent drug interaction effects may vary across an entire dose-response matrix. We developed a two-way pharmacodynamic (TWPD) model to capture the PD of two-drug combinations. TWPD captures interactions between upstream and downstream drugs that act on different stages of viral replication, by quantifying upstream drug efficacy and concentration-dependent effects on downstream drug pharmacodynamic parameters. We applied TWPD to previously published in vitro drug matrixes for repurposed potential anti-Ebola and anti-SARS-CoV-2 drug pairs. Depending on the drug pairing, the model recapitulated combined efficacies as or more accurately than existing models and can be used to infer efficacy at untested drug concentrations. TWPD fits the data slightly better in one direction for all drug pairs, meaning that we can tentatively infer the upstream drug. Based on its high accuracy, TWPD could be used in concert with PK models to estimate the therapeutic effects of drug pairs in vivo.
Collapse
Affiliation(s)
- Shuang Xu
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
| | - Shadisadat Esmaeili
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
| | - E. Fabian Cardozo-Ojeda
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
| | - Ashish Goyal
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
| | - Judith M. White
- Department of Microbiology, University of Virginia, Charlottesville, Virginia, USA
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Stephen J. Polyak
- Virology Division, Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Joshua T. Schiffer
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| |
Collapse
|
6
|
Berska J, Bugajska J, Sztefko K. A Liquid Chromatography-Tandem Mass Spectrometry Method for Simultaneously Determining Meropenem and Linezolid in Blood and Cerebrospinal Fluid. Ann Lab Med 2024; 44:174-178. [PMID: 37869779 PMCID: PMC10628749 DOI: 10.3343/alm.2023.0250] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/03/2023] [Accepted: 09/14/2023] [Indexed: 10/24/2023] Open
Abstract
Antibiotic therapy requires appropriate dosage of drugs for effective treatment. Too low antibiotic concentrations may lead to treatment failure and the development of resistant pathogens, whereas overdosing may cause neurological side effects or hemolytic diseases. Meropenem and linezolid are used only in the treatment of serious infections or when other antibiotics are no longer effective as well as for treating central nervous system infections. It is difficult or sometimes even impossible to predict the relation between dosing of antibiotics and its cerebrospinal fluid (CSF) concentration; thus, a method of determining antibiotics not only in the blood but also in the CSF is needed. Analytical method validation is an integral part of good laboratory practice and ensures high accuracy of the results. We performed complete validation process according to the Food and Drug Administration and European Medicine Agency, covering the aspects precision, specificity, accuracy, recovery, limit of detection, limit of quantification, stability, carry-over, and matrix effects. Our liquid chromatography-tandem mass spectrometry method for the simultaneous measurement of meropenem and linezolid in different matrix meets all the acceptance criteria. The method was successfully applied to determine meropenem and linezolid concentrations in serum and CSF samples obtained from children treated with these antibiotics.
Collapse
Affiliation(s)
- Joanna Berska
- Department of Clinical Biochemistry, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Jolanta Bugajska
- Department of Clinical Biochemistry, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Krystyna Sztefko
- Department of Clinical Biochemistry, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
7
|
Yekani M, Azargun R, Sharifi S, Sadri Nahand J, Hasani A, Ghanbari H, Sadat Seyyedi Z, Memar MY, Maleki Dizaj S. Preparation, Physicochemical Characterization, Antimicrobial Effects, Biocompatibility and Cytotoxicity of Co-Loaded Meropenem and Vancomycin in Mesoporous Silica Nanoparticles. Biomedicines 2023; 11:3075. [PMID: 38002075 PMCID: PMC10669092 DOI: 10.3390/biomedicines11113075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/05/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Mesoporous silica nanoparticles (MSNPs) have been reported as an effective system to co-deliver a variety of different agents to enhance efficiency and improve biocompatibility. This study was aimed at the preparation, physicochemical characterization, antimicrobial effects, biocompatibility, and cytotoxicity of vancomycin and meropenem co-loaded in the mesoporous silica nanoparticles (Van/Mrp-MSNPs). The prepared nanoparticles were explored for their physicochemical features, antibacterial and antibiofilm effects, biocompatibility, and cytotoxicity. The minimum inhibitory concentrations (MICs) of the Van/Mrp-MSNPs (0.12-1 µg/mL) against Staphylococcus aureus isolates were observed to be lower than those of the same concentrations of vancomycin and meropenem. The minimum biofilm inhibitory concentration (MBIC) range of the Van/Mrp-MSNPs was 8-64 μg/mL, which was lower than the meropenem and vancomycin MBICs. The bacterial adherence was not significantly decreased upon exposure to levels lower than the MICs of the MSNPs and Van/Mrp-MSNPs. The viability of NIH/3T3 cells treated with serial concentrations of the MSNPs and Van/Mrp-MSNPs were 73-88% and 74-90%, respectively. The Van/Mrp-MSNPs displayed considerable inhibitory effects against MRSA, favorable biocompatibility, and low cytotoxicity. The Van/Mrp-MSNPs could be a potential system for the treatment of infections.
Collapse
Affiliation(s)
- Mina Yekani
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz 51548-53431, Iran
- Department of Microbiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan 87137-83976, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan 87137-83976, Iran
| | - Robab Azargun
- Medicinal Plants Research Center, Maragheh University of Medical Science, Maragheh 55158-78151, Iran
| | - Simin Sharifi
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz 51548-53431, Iran;
| | - Javid Sadri Nahand
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz 51548-53431, Iran
| | - Alka Hasani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51548-53431, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz 51548-53431, Iran
| | - Hadi Ghanbari
- Department of Pharmacognosy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz 51548-53431, Iran;
| | - Zahra Sadat Seyyedi
- Department of Microbiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan 87137-83976, Iran
| | - Mohammad Yousef Memar
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz 51548-53431, Iran
| | - Solmaz Maleki Dizaj
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz 51548-53431, Iran;
| |
Collapse
|
8
|
Kroemer N, Martens M, Decousser JW, Grégoire N, Nordmann P, Wicha SG. Evaluation of in vitro pharmacodynamic drug interactions of ceftazidime/avibactam and fosfomycin in Escherichia coli. J Antimicrob Chemother 2023; 78:2524-2534. [PMID: 37624929 DOI: 10.1093/jac/dkad264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 07/31/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Combination therapy can increase efficacy of antibiotics and prevent emergence of resistance. Ceftazidime/avibactam and fosfomycin may be empirically combined for this purpose, but a systematic and quantitative evaluation of this combination is needed. OBJECTIVES In this study, a systematic analysis of the pharmacodynamic interactions of ceftazidime/avibactam and fosfomycin in clinical and isogenic Escherichia coli strains carrying genes coding for several carbapenemases or ESBLs was performed and pharmacodynamic interactions were quantified by modelling and simulations. METHODS Pharmacodynamic interactions were evaluated in 'dynamic' chequerboard experiments with quantification of viable bacteria in eight isogenic and six clinical E. coli strains. Additionally, supplemental time-kill experiments were performed and genomic analyses were conducted on representative fosfomycin-resistant subpopulations. Models were fitted to all data using R and NONMEM®. RESULTS Synergistic drug interactions were identified for 67% of the clinical and 75% of the isogenic isolates with a mean EC50 reduction of >50%. Time-kill experiments confirmed the interactions and modelling quantified EC50 reductions up to 97% in combination and synergy prevented regrowth of bacteria by enhanced killing effects. In 9 out of 12 fosfomycin-resistant mutants, genomic analyses identified previously reported mutations. CONCLUSIONS The broad synergistic in vitro activity of ceftazidime/avibactam and fosfomycin confirms the potential of the application of this drug combination in clinics. The substantial reduction of the EC50 in combination may allow use of lower doses or treatment of organisms with higher MIC values and encourage further research translating these findings into the clinical setting.
Collapse
Affiliation(s)
- Niklas Kroemer
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| | - Miklas Martens
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| | - Jean-Winoc Decousser
- Dynamic Team-EA 7380, Faculté de santé, Université Paris-Est-Créteil Val-De-Marne, Créteil, France
| | - Nicolas Grégoire
- Inserm U1070, Pharmacologie des Anti-infectieux et Antibiorésistance, Poitiers, France
- Université de Poitiers, UFR de Médecine Pharmacie, Poitiers, France
- CHU de Poitiers, Laboratoire de Toxicologie-Pharmacologie, Poitiers, France
| | - Patrice Nordmann
- Medical and Molecular Microbiology, University of Fribourg, Fribourg, Switzerland
| | - Sebastian G Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| |
Collapse
|
9
|
Peris-Vicente J, Albiol-Chiva J, Bose D, Durgbanshi A, Carda-Broch S. A method to determine two antibiotics prescribed to treat nosocomial infections in plasma and urine by micellar liquid chromatography. J Chromatogr B Analyt Technol Biomed Life Sci 2023; 1225:123777. [PMID: 37290211 DOI: 10.1016/j.jchromb.2023.123777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/19/2023] [Accepted: 05/30/2023] [Indexed: 06/10/2023]
Abstract
Combined prescription of the antimicrobial drugs linezolid and meropenem is a common strategy to treat multidrug-resistant nosocomial infections. We propose an innovative method to determine these two drugs in plasma and urine, based on micellar liquid chromatography. Both biological fluids were diluted in mobile phase, filtered and directly injected, without any extraction step. Using a C18 column and a mobile phase of 0.1 M sodium dodecyl sulfate - 10 % methanol, phosphate buffered at pH 3, running under isocratic mode, both antibiotics were eluted without overlapping in<15 min. Detection was by absorbance: 255 nm for linezolid and 310 nm for meropenem. The influence of sodium dodecyl sulfate and methanol concentration on retention factor was established for both drugs using an interpretative approach assisted by chemometrics. The procedure was successfully validated following the guidelines of 2018 Bioanalytical Method Validation Guidance for Industry in terms of: linearity (determination coefficients over 0.99990), calibration range (1 - 50 mg/L), instrumental and method sensitivity, trueness (bias of -10.8 to + 2.4%), precision (relative standard deviation of < 10.2%), dilution integrity, carry-over effect, robustness and stability. It should be emphasized that the method uses low volumes of toxic and volatile solvents and can be achieved in a short period. The procedure was found useful for routine analysis, as it was cost-affordable, more eco-friendly and safer than hydroorganic HPLC, easy-to-handle and highly sample-throughput. Finally, it was applied to incurred samples of patients taking this medication.
Collapse
Affiliation(s)
- Juan Peris-Vicente
- Department of Analytical Chemistry, Faculty of Chemistry, Universitat de València, 46100 Burjassot, Spain.
| | - Jaume Albiol-Chiva
- Department of Physical and Analytical Chemistry, ESTCE, Universitat Jaume I, 12071 Castelló, Spain
| | - Devasish Bose
- Department of Criminology and Forensic Science, Doctor Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh 470003, India
| | - Abhilasha Durgbanshi
- Department of Chemistry, Doctor Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh 470003, India
| | - Samuel Carda-Broch
- Department of Physical and Analytical Chemistry, ESTCE, Universitat Jaume I, 12071 Castelló, Spain
| |
Collapse
|
10
|
In silico assessment on TdP risks of drug combinations under CiPA paradigm. Sci Rep 2023; 13:2924. [PMID: 36807374 PMCID: PMC9940090 DOI: 10.1038/s41598-023-29208-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 01/31/2023] [Indexed: 02/22/2023] Open
Abstract
Researchers have recently proposed the Comprehensive In-vitro Proarrhythmia Assay (CiPA) to analyze medicines' TdP risks. Using the TdP metric known as qNet, numerous single-drug effects have been studied to classify the medications as low, intermediate, and high-risk. Furthermore, multiple medication therapies are recognized as a potential method for curing patients, mainly when limited drugs are available. This work expands the TdP risk assessment of drugs by introducing a CiPA-based in silico analysis of the TdP risk of combined drugs. The cardiac cell model was simulated using the population of models approach incorporating drug-drug interactions (DDIs) models on several ion channels for various drug pairs. Action potential duration (APD90), qNet, and calcium duration (CaD90) were computed and analyzed as biomarker features. The drug combination maps were also used to illustrate combined medicines' TdP risk. We found that the combined drugs alter cell responses in terms of biomarkers such as APD90, qNet, and CaD90 in a highly nonlinear manner. The results also revealed that combinations of high-risk with low-risk and intermediate-risk with low-risk drugs could result in compounds with varying TdP risks depending on the drug concentrations.
Collapse
|
11
|
Pearson RA, Wicha SG, Okour M. Drug Combination Modeling: Methods and Applications in Drug Development. J Clin Pharmacol 2023; 63:151-165. [PMID: 36088583 DOI: 10.1002/jcph.2128] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/22/2022] [Indexed: 01/18/2023]
Abstract
Combination therapies have become increasingly researched and used in the treatment and management of complex diseases due to their ability to increase the chances for better efficacy and decreased toxicity. To evaluate drug combinations in drug development, pharmacokinetic and pharmacodynamic interactions between drugs in combination can be quantified using mathematical models; however, it can be difficult to deduce which models to use and how to use them to aid in clinical trial simulations to simulate the effect of a drug combination. This review paper aims to provide an overview of the various methods used to evaluate combination drug interaction for use in clinical trial development and a practical guideline on how combination modeling can be used in the settings of clinical trials.
Collapse
Affiliation(s)
- Rachael A Pearson
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sebastian G Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| | - Malek Okour
- Clinical Pharmacology Modeling and Simulation (CPMS), GlaxoSmithKline, Upper Providence, Pennsylvania, USA
| |
Collapse
|
12
|
Liu G, Liu A, Yang C, Zhou C, Zhou Q, Li H, Yang H, Mo J, Zhang Z, Li G, Si H, Ou C. Portulaca oleracea L. organic acid extract inhibits persistent methicillin-resistant Staphylococcus aureus in vitro and in vivo. Front Microbiol 2023; 13:1076154. [PMID: 36713183 PMCID: PMC9874160 DOI: 10.3389/fmicb.2022.1076154] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 12/13/2022] [Indexed: 01/12/2023] Open
Abstract
Staphylococcus aureus continues to be one of the most important pathogens capable of causing a wide range of infections in different sites of the body in humans and livestock. With the emergence of methicillin-resistant strains and the introduction of strict laws on antibiotic usage in animals, antibiotic replacement therapy has become increasingly popular. Previous studies have shown that Portulaca oleracea L. extract exerts a certain degree of bacteriostatic effect, although the active ingredients are unknown. In the present study, the antibacterial activity of the organic acid of P. oleracea (OAPO) against S. aureus was examined using a series of experiments, including the minimum inhibitory concentration, growth curve, and bacteriostasis curve. In vitro antibacterial mechanisms were evaluated based on the integrity and permeability of the cell wall and membrane, scanning electron microscopy, and soluble protein content. A mouse skin wound recovery model was used to verify the antibacterial effects of OAPO on S. aureus in vivo. The results showed that OAPO not only improved skin wound recovery but also decreased the bacterial load in skin wounds. Moreover, the number of inflammatory cells and cytokines decreased in the OAPO-treated groups. In summary, this study reports a botanical extract that can inhibit S. aureus in vitro and in vivo, indicating the potential use of OAPO to prevent and control S. aureus infection in the near future.
Collapse
Affiliation(s)
- Gengsong Liu
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Aijing Liu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Cheng Yang
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Congcong Zhou
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Qiaoyan Zhou
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Haizhu Li
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Hongchun Yang
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Jiahao Mo
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Zhidan Zhang
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Gonghe Li
- College of Animal Science and Technology, Guangxi University, Nanning, China,Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning, China,Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning, China
| | - Hongbin Si
- College of Animal Science and Technology, Guangxi University, Nanning, China,Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning, China,Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning, China
| | - Changbo Ou
- College of Animal Science and Technology, Guangxi University, Nanning, China,Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning, China,Guangxi Key Laboratory of Animal Reproduction, Breeding and Disease Control, Nanning, China,*Correspondence: Changbo Ou, ✉
| |
Collapse
|
13
|
New In Vitro Interaction-Parasite Reduction Ratio Assay for Early Derisk in Clinical Development of Antimalarial Combinations. Antimicrob Agents Chemother 2022; 66:e0055622. [PMID: 36197116 PMCID: PMC9664866 DOI: 10.1128/aac.00556-22] [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] [Indexed: 11/24/2022] Open
Abstract
The development and spread of drug-resistant phenotypes substantially threaten malaria control efforts. Combination therapies have the potential to minimize the risk of resistance development but require intensive preclinical studies to determine optimal combination and dosing regimens. To support the selection of new combinations, we developed a novel in vitro-in silico combination approach to help identify the pharmacodynamic interactions of the two antimalarial drugs in a combination which can be plugged into a pharmacokinetic/pharmacodynamic model built with human monotherapy parasitological data to predict the parasitological endpoints of the combination. This makes it possible to optimally select drug combinations and doses for the clinical development of antimalarials. With this assay, we successfully predicted the endpoints of two phase 2 clinical trials in patients with the artefenomel-piperaquine and artefenomel-ferroquine drug combinations. In addition, the predictive performance of our novel in vitro model was equivalent to that of the humanized mouse model outcome. Last, our more informative in vitro combination assay provided additional insights into the pharmacodynamic drug interactions compared to the in vivo systems, e.g., a concentration-dependent change in the maximum killing effect (Emax) and the concentration producing 50% of the killing maximum effect (EC50) of piperaquine or artefenomel or a directional reduction of the EC50 of ferroquine by artefenomel and a directional reduction of Emax of ferroquine by artefenomel. Overall, this novel in vitro-in silico-based technology will significantly improve and streamline the economic development of new drug combinations for malaria and potentially also in other therapeutic areas.
Collapse
|
14
|
Iqbal K, Rohde H, Huang J, Tikiso T, Amann LF, Zeitlinger M, Wicha SG. A pharmacokinetic-pharmacodynamic (PKPD) model-based analysis of tedizolid against enterococci using the hollow-fibre infection model. J Antimicrob Chemother 2022; 77:2470-2478. [PMID: 35696407 DOI: 10.1093/jac/dkac183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 05/14/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Tedizolid is a novel oxazolidinone antibiotic. Considering the higher antibacterial effect in immunocompetent compared with immunosuppressed animals, it is not recommended in immunocompromised patients. OBJECTIVES In this study, we assessed the 'pure' pharmacokinetic-pharmacodynamic (PKPD) relationship for tedizolid against Enterococcus in the hollow-fibre infection model (HFIM). METHODS Unbound plasma concentration time profiles (200-5000 mg/day IV) were simulated in the HFIM over 120 h against an Enterococcus faecalis strain and two clinical isolates of Enterococcus faecium (VRE-vanB and VRE-vanA). Next, a PKPD model describing tedizolid efficacy against bacterial isolates was developed. A population PK model was linked to the developed PKPD model and utilized to predict the bacterial kinetics in plasma and in target tissues [adipose, muscle, epithelial lining fluid (ELF) and sputum] over 120 h of therapy. RESULTS The PKPD model adequately described the bacterial kill kinetics for all bacterial populations. At the human recommended dose of 200 mg/day, bacterial growth was predicted in plasma and all tissues, except for ELF. Bacteriostasis was observed only at a higher dose of 1200 mg/day over 120 h. An fAUC/MIC of 80 related to stasis over 120 h. Subpopulations resistant to 3 × MIC were amplified in plasma and target tissues, except for ELF, at doses of 200-800 mg/day. CONCLUSIONS The human dose of 200 mg/day was insufficient to suppress bacterial growth in the HFIM, indicating that further components contribute to the clinical effect of tedizolid. This study supports the warning/precaution for tedizolid to limit its use in immunocompromised patients.
Collapse
Affiliation(s)
- K Iqbal
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| | - H Rohde
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - J Huang
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - T Tikiso
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| | - L F Amann
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| | - M Zeitlinger
- Department of Clinical Pharmacology, General Hospital (AKH), Medical University of Vienna, Vienna, Austria
| | - S G Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| |
Collapse
|
15
|
Lill D, Kümmel A, Mitov V, Kaschek D, Gobeau N, Schmidt H, Timmer J. Efficient simulation of clinical target response surfaces. CPT Pharmacometrics Syst Pharmacol 2022; 11:512-523. [PMID: 35199969 PMCID: PMC9007598 DOI: 10.1002/psp4.12779] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/18/2022] [Accepted: 02/14/2022] [Indexed: 11/08/2022] Open
Abstract
Simulation of combination therapies is challenging due to computational complexity. Either a simple model is used to simulate the response for many combinations of concentration to generate a response surface but parameter variability and uncertainty are neglected and the concentrations are constant—the link to the doses to be administered is difficult to make—or a population pharmacokinetic/pharmacodynamic model is used to predict the response to combination therapy in a clinical trial taking into account the time‐varying concentration profile, interindividual variability (IIV), and parameter uncertainty but simulations are limited to only a few selected doses. We devised new algorithms to efficiently search for the combination doses that achieve a predefined efficacy target while taking into account the IIV and parameter uncertainty. The result of this method is a response surface of confidence levels, indicating for all dose combinations the likelihood of reaching the specified efficacy target. We highlight the importance to simulate across a population rather than focus on an individual. Finally, we provide examples of potential applications, such as informing experimental design.
Collapse
Affiliation(s)
- Daniel Lill
- IntiQuan GmbH Basel Switzerland
- Institute of Physics University of Freiburg Freiburg Germany
| | | | | | | | | | | | - Jens Timmer
- Institute of Physics University of Freiburg Freiburg Germany
- Centre for Integrative Biological Signalling Studies (CIBSS) University of Freiburg Freiburg Germany
- Freiburg Center for Data Analysis and Modelling (FDM) University of Freiburg Freiburg Germany
| |
Collapse
|
16
|
Vittrup SØ, Hanberg P, Knudsen MB, Tøstesen SK, Kipp JO, Hansen J, Jørgensen NP, Stilling M, Bue M. Tibial bone and soft-tissue concentrations following combination therapy with vancomycin and meropenem - evaluated by microdialysis in a porcine model : should patients with open fractures have higher doses of antibiotics? Bone Joint Res 2022; 11:112-120. [PMID: 35176868 PMCID: PMC8882321 DOI: 10.1302/2046-3758.112.bjr-2021-0321.r1] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Aims Prompt and sufficient broad-spectrum empirical antibiotic treatment is key to preventing infection following open tibial fractures. Succeeding co-administration, we dynamically assessed the time for which vancomycin and meropenem concentrations were above relevant epidemiological cut-off (ECOFF) minimal inhibitory concentrations (T > MIC) in tibial compartments for the bacteria most frequently encountered in open fractures. Low and high MIC targets were applied: 1 and 4 µg/ml for vancomycin, and 0.125 and 2 µg/ml for meropenem. Methods Eight pigs received a single dose of 1,000 mg vancomycin and 1,000 mg meropenem simultaneously over 100 minutes and 10 minutes, respectively. Microdialysis catheters were placed for sampling over eight hours in tibial cancellous bone, cortical bone, and adjacent subcutaneous adipose tissue. Venous blood samples were collected as references. Results Across the targeted ECOFF values, vancomycin displayed longer T > MIC in all the investigated compartments in comparison to meropenem. For both drugs, cortical bone exhibited the shortest T > MIC. For the low MIC targets and across compartments, mean T > MIC ranged between 208 and 449 minutes (46% to 100%) for vancomycin and between 189 and 406 minutes (42% to 90%) for meropenem. For the high MIC targets, mean T > MIC ranged between 30 and 446 minutes (7% to 99%) for vancomycin and between 45 and 181 minutes (10% to 40%) for meropenem. Conclusion The differences in the T > MIC between the low and high targets illustrate how the interpretation of these results is highly susceptible to the defined MIC target. To encompass any trauma, contamination, or individual tissue differences, a more aggressive dosing approach may be considered to achieve longer T > MIC in all the exposed tissues, and thereby lower the risk of acquiring an infection after open tibial fractures. Cite this article: Bone Joint Res 2022;11(2):112–120.
Collapse
Affiliation(s)
- Sofus Ørbæk Vittrup
- Aarhus Denmark Microdialysis Research (ADMIRE), Orthopaedic Research Laboratory, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Pelle Hanberg
- Aarhus Denmark Microdialysis Research (ADMIRE), Orthopaedic Research Laboratory, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Martin Bruun Knudsen
- Aarhus Denmark Microdialysis Research (ADMIRE), Orthopaedic Research Laboratory, Aarhus University Hospital, Aarhus, Denmark
| | - Sara Kousgaard Tøstesen
- Aarhus Denmark Microdialysis Research (ADMIRE), Orthopaedic Research Laboratory, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Josephine Olsen Kipp
- Aarhus Denmark Microdialysis Research (ADMIRE), Orthopaedic Research Laboratory, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jakob Hansen
- Department of Forensic Medicine, Aarhus University Hospital, Aarhus, Denmark
| | | | - Maiken Stilling
- Aarhus Denmark Microdialysis Research (ADMIRE), Orthopaedic Research Laboratory, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Orthopaedic Surgery, Aarhus University Hospital, Aarhus, Denmark
| | - Mats Bue
- Aarhus Denmark Microdialysis Research (ADMIRE), Orthopaedic Research Laboratory, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Orthopaedic Surgery, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
17
|
Kroemer N, Aubry R, Couet W, Grégoire N, Wicha SG. Optimized Rhombic Experimental Dynamic Checkerboard Designs to Elucidate Pharmacodynamic Drug Interactions of Antibiotics. Pharm Res 2022; 39:3267-3277. [PMID: 36163408 PMCID: PMC9780134 DOI: 10.1007/s11095-022-03396-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 09/09/2022] [Indexed: 12/27/2022]
Abstract
PURPOSE Quantification of pharmacodynamic interactions is key in combination therapies, yet conventional checkerboard experiments with up to 10 by 10 combinations are labor-intensive. Therefore, this study provides optimized experimental rhombic checkerboard designs to enable an efficient interaction screening with significantly reduced experimental workload. METHODS Based on the general pharmacodynamic interaction (GPDI) model implemented in Bliss Independence, a novel rhombic 'dynamic' checkerboard design with quantification of bacteria instead of turbidity as endpoint was developed. In stochastic simulations and estimations (SSE), the precision and accuracy of interaction parameter estimations and classification rates of conventional reference designs and the newly proposed rhombic designs based on effective concentrations (EC) were compared. RESULTS Although a conventional rich design with 20-times as many combination scenarios provided estimates of interaction parameters with higher accuracy, precision and classification rates, the optimized rhombic designs with one natural growth scenario, three monotherapy scenarios per combination partner and only four combination scenarios were still superior to conventional reduced designs with twice as many combination scenarios. Additionally, the rhombic designs were able to identify whether an interaction occurred as a shift on maximum effect or EC50 with > 98%. Overall, effective concentration-based designs were found to be superior to traditional standard concentrations, but were more challenged by strong interaction sizes exceeding their adaptive concentration ranges. CONCLUSION The rhombic designs proposed in this study enable a reduction of resources and labor and can be a tool to streamline higher throughput in drug interaction screening.
Collapse
Affiliation(s)
- Niklas Kroemer
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| | - Romain Aubry
- Inserm U1070, Poitiers, France ,Université de Poitiers, UFR de Médecine Pharmacie, Poitiers, France
| | - William Couet
- Inserm U1070, Poitiers, France ,Université de Poitiers, UFR de Médecine Pharmacie, Poitiers, France ,CHU de Poitiers, Laboratoire de Toxicologie-Pharmacologie, Poitiers, France
| | - Nicolas Grégoire
- Inserm U1070, Poitiers, France ,Université de Poitiers, UFR de Médecine Pharmacie, Poitiers, France ,CHU de Poitiers, Laboratoire de Toxicologie-Pharmacologie, Poitiers, France
| | - Sebastian G. Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| |
Collapse
|
18
|
A personalised approach to antibiotic pharmacokinetics and pharmacodynamics in critically ill patients. Anaesth Crit Care Pain Med 2021; 40:100970. [PMID: 34728411 DOI: 10.1016/j.accpm.2021.100970] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 07/26/2021] [Accepted: 08/14/2021] [Indexed: 01/01/2023]
Abstract
Critically ill patients admitted to intensive care unit (ICU) with severe infections, or those who develop nosocomial infections, have poor outcomes with substantial morbidity and mortality. Such patients commonly have suboptimal antibiotic exposures at routinely used antibiotic doses related to an increased volume of distribution and altered clearance due to their underlying altered physiology. Furthermore, the use of extracorporeal devices such as renal replacement therapy and extracorporeal membrane oxygenation in these group of patients also has the potential to alter in vivo drug concentrations. Moreover, ICU patients are likely to be infected with less-susceptible pathogens. Therefore, one potential contributing cause to the poor outcomes observed in critically ill patients may be related to subtherapeutic antibiotic exposures. Newer concepts include the clinician considering optimised dosing based on a blood antibiotic exposure defined by pharmacokinetic modelling and therapeutic drug monitoring, combined with a knowledge of the antibiotic penetration into the site of infection, thereby achieving optimal bacterial killing. Such optimised dosing is likely to improve patient outcomes. The aim of this review is to highlight key aspects of antibiotic pharmacokinetics and pharmacodynamics (PK/PD) in critically ill patients and provide a PK/PD approach to tailor antibiotic dosing to the individual patient.
Collapse
|
19
|
Seeger J, Michelet R, Kloft C. Quantification of persister formation of Escherichia coli leveraging electronic cell counting and semi-mechanistic pharmacokinetic/pharmacodynamic modelling. J Antimicrob Chemother 2021; 76:2088-2096. [PMID: 33997902 DOI: 10.1093/jac/dkab146] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 04/07/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Persister formation of Escherichia coli under fluoroquinolone exposure causes treatment failure and promotes emergence of resistant strains. Semi-mechanistic pharmacokinetic/pharmacodynamic modelling of data obtained from in vitro infection model experiments comprehensively characterizes exposure-effect relationships, providing mechanistic insights. OBJECTIVES To quantify persister formation of E. coli under levofloxacin exposure and to explain the observed growth-kill behaviour, leveraging electronic cell counting and pharmacokinetic/pharmacodynamic modelling. METHODS Three fluoroquinolone-resistant clinical E. coli isolates were exposed to levofloxacin in static and dynamic in vitro infection model experiments. Complementary to plate counting, bacterial concentrations over time were quantified by electronic cell counting and amalgamated in a semi-mechanistic pharmacokinetic/pharmacodynamic model (1281 bacterial and 394 levofloxacin observations). RESULTS Bacterial regrowth was observed under exposure to clinically relevant dosing regimens in the dynamic in vitro infection model. Electronic cell counting facilitated identification of three bacterial subpopulations: persisters, viable cells and dead cells. Two strain-specific manifestations of the levofloxacin effect were identified: a killing effect, characterized as a sigmoidal Emax model, and an additive increase in persister formation under levofloxacin exposure. Significantly different EC50 values quantitatively discerned levofloxacin potency for two isolates displaying the same MIC value: 8 mg/L [EC50 = 17.2 (95% CI = 12.6-23.8) mg/L and 8.46 (95% CI = 6.86-10.3) mg/L, respectively]. Persister formation was most pronounced for the isolate with the lowest MIC value (2 mg/L). CONCLUSIONS The developed pharmacokinetic/pharmacodynamic model adequately characterized growth-kill behaviour of three E. coli isolates and unveiled strain-specific levofloxacin potencies and persister formation. The mimicked dosing regimens did not eradicate the resistant isolates and enhanced persister formation to a strain-specific extent.
Collapse
Affiliation(s)
- Johanna Seeger
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Kelchstr. 31, 12169, Berlin, Germany
| | - Robin Michelet
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Kelchstr. 31, 12169, Berlin, Germany
| | - Charlotte Kloft
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Kelchstr. 31, 12169, Berlin, Germany
| |
Collapse
|
20
|
An In Vitro Perspective on What Individual Antimicrobials Add to Mycobacterium avium Complex Therapies. Antimicrob Agents Chemother 2021; 65:e0273020. [PMID: 33972258 DOI: 10.1128/aac.02730-20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
For Mycobacterium avium complex pulmonary disease (MAC-PD), current treatment regimens yield low cure rates. To obtain an evidence-based combination therapy, we assessed the in vitro activity of six drugs, namely, clarithromycin (CLR), rifampin (RIF), ethambutol (EMB), amikacin (AMK), clofazimine (CLO), and minocycline (MIN), alone and in combination, against Mycobacterium avium and studied the contributions of individual antibiotics to efficacy. The MICs of all antibiotics against M. avium ATCC 700898 were determined by broth microdilution. We performed kinetic time-kill assays of all single drugs and clinically relevant two-, three-, four-, and five-drug combinations against M. avium. Pharmacodynamic interactions of these combinations were assessed using area under the time-kill curve-derived effect size and Bliss independence. Adding a second drug yielded an average increase of the effect size (E) of 18.7% ± 32.9%, although antagonism was seen in some combinations. Adding a third drug showed a smaller increase in effect size (+12.2% ± 11.5%). The RIF-CLO-CLR (E of 102 log10 CFU/ml · day), RIF-AMK-CLR (E of 101 log10 CFU/ml · day), and AMK-MIN-EMB (E of 97.8 log10 CFU/ml · day) regimens proved more active than the recommended RIF-EMB-CLR regimen (E of 89.1 log10 CFU/ml · day). The addition of a fourth drug had little impact on effect size (+4.54% ± 3.08%). In vitro, several two- and three-drug regimens are as effective as the currently recommended regimen for MAC-PD. Adding a fourth drug to any regimen had little additional effect. In vitro, the most promising regimen would be RIF-AMK-macrolide or RIF-CLO-macrolide.
Collapse
|
21
|
Ruth MM, Koeken VACM, Pennings LJ, Svensson EM, Wertheim HFL, Hoefsloot W, van Ingen J. Is there a role for tedizolid in the treatment of non-tuberculous mycobacterial disease? J Antimicrob Chemother 2021; 75:609-617. [PMID: 31886864 PMCID: PMC7021090 DOI: 10.1093/jac/dkz511] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/06/2019] [Accepted: 11/08/2019] [Indexed: 12/29/2022] Open
Abstract
Background Pulmonary infections caused by non-tuberculous mycobacteria (NTM) are hard to treat and have low cure rates despite intensive multidrug therapy. Objectives To assess the feasibility of tedizolid, a new oxazolidinone, for the treatment of Mycobacterium avium and Mycobacterium abscessus. Methods We determined MICs of tedizolid for 113 isolates of NTM. Synergy with key antimycobacterial drugs was assessed using the chequerboard method and calculation of the FIC index (FICI). We performed time–kill kinetics assays of tedizolid alone and combined with amikacin for M. abscessus and with ethambutol for M. avium. Human macrophages were infected with M. abscessus and M. avium and subsequently treated with tedizolid; intracellular and extracellular cfu were quantified over time. Results NTM isolates generally had a lower MIC of tedizolid than of linezolid. FICIs were lowest between tedizolid and amikacin for M. abscessus (FICI = 0.75) and between tedizolid and ethambutol for M. avium (FICI = 0.72). Clarithromycin and tedizolid showed initial synergy, which was abrogated by erm(41)-induced macrolide resistance (FICI = 0.53). Tedizolid had a weak bacteriostatic effect on M. abscessus and combination with amikacin slightly prolonged its effect. Tedizolid had concentration-dependent activity against M. avium and its efficacy was enhanced by ethambutol. Both combinations had a concentration-dependent synergistic effect. Tedizolid could inhibit the intracellular bacterial population of both M. avium and M. abscessus. Conclusions Tedizolid should be further investigated in pharmacodynamic studies and clinical trials for M. avium complex pulmonary disease. It is less active against M. abscessus, but still promising.
Collapse
Affiliation(s)
- Mike Marvin Ruth
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Valerie A C M Koeken
- Radboudumc Center for Infectious Diseases, Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lian J Pennings
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Elin M Svensson
- Radboudumc Center for Infectious Diseases, Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Heiman F L Wertheim
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Wouter Hoefsloot
- Radboudumc Center for Infectious Diseases, Department of Pulmonary Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jakko van Ingen
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
22
|
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
|
23
|
Franck S, Michelet R, Casilag F, Sirard JC, Wicha SG, Kloft C. A Model-Based Pharmacokinetic/Pharmacodynamic Analysis of the Combination of Amoxicillin and Monophosphoryl Lipid A Against S. pneumoniae in Mice. Pharmaceutics 2021; 13:469. [PMID: 33808396 PMCID: PMC8065677 DOI: 10.3390/pharmaceutics13040469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 11/16/2022] Open
Abstract
Combining amoxicillin with the immunostimulatory toll-like receptor 4 agonist monophosphoryl lipid A (MPLA) represents an innovative approach for enhancing antibacterial treatment success. Exploiting pharmacokinetic and pharmacodynamic data from an infection model of Streptococcus pneumoniae infected mice, we aimed to evaluate the preclinical exposure-response relationship of amoxicillin with MPLA coadministration and establish a link to survival. Antibiotic serum concentrations, bacterial numbers in lung and spleen and survival data of mice being untreated or treated with amoxicillin (four dose levels), MPLA, or their combination were analyzed by nonlinear mixed-effects modelling and time-to-event analysis using NONMEM® to characterize these treatment regimens. On top of a pharmacokinetic interaction, regarding the pharmacodynamic effects the combined treatment was superior to both monotherapies: The amoxicillin efficacy at highest dose was increased by a bacterial reduction of 1.74 log10 CFU/lung after 36 h and survival was increased 1.35-fold to 90.3% after 14 days both compared to amoxicillin alone. The developed pharmacometric pharmacokinetic/pharmacodynamic disease-treatment-survival models provided quantitative insights into a novel treatment option against pneumonia revealing a pharmacokinetic interaction and enhanced activity of amoxicillin and the immune system stimulator MPLA in combination. Further development of this drug combination flanked with pharmacometrics towards the clinical setting seems promising.
Collapse
Affiliation(s)
- Sebastian Franck
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, 12169 Berlin, Germany; (S.F.); (R.M.)
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, 20146 Hamburg, Germany;
| | - Robin Michelet
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, 12169 Berlin, Germany; (S.F.); (R.M.)
| | - Fiordiligie Casilag
- CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR8204-CIIL-Center of Infection and Immunity of Lille, University Lille, 59019 Lille, France; (F.C.); (J.-C.S.)
| | - Jean-Claude Sirard
- CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR8204-CIIL-Center of Infection and Immunity of Lille, University Lille, 59019 Lille, France; (F.C.); (J.-C.S.)
| | - Sebastian G. Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, 20146 Hamburg, Germany;
| | - Charlotte Kloft
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, 12169 Berlin, Germany; (S.F.); (R.M.)
| |
Collapse
|
24
|
Iqbal K, Broeker A, Nowak H, Rahmel T, Nussbaumer-Pröll A, Österreicher Z, Zeitlinger M, Wicha S. A pharmacometric approach to define target site-specific breakpoints for bacterial killing and resistance suppression integrating microdialysis, time–kill curves and heteroresistance data: a case study with moxifloxacin. Clin Microbiol Infect 2020; 26:1255.e1-1255.e8. [DOI: 10.1016/j.cmi.2020.02.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 01/18/2020] [Accepted: 02/11/2020] [Indexed: 02/07/2023]
|
25
|
Ruth MM, Sangen JJN, Remmers K, Pennings LJ, Svensson E, Aarnoutse RE, Zweijpfenning SMH, Hoefsloot W, Kuipers S, Magis-Escurra C, Wertheim HFL, van Ingen J. A bedaquiline/clofazimine combination regimen might add activity to the treatment of clinically relevant non-tuberculous mycobacteria. J Antimicrob Chemother 2020; 74:935-943. [PMID: 30649327 DOI: 10.1093/jac/dky526] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 11/07/2018] [Accepted: 11/13/2018] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Non-tuberculous mycobacteria (NTM) infections are hard to treat. New antimicrobial drugs and smarter combination regimens are needed. OBJECTIVES Our aim was to determine the in vitro activity of bedaquiline against NTM and assess its synergy with established antimycobacterials. METHODS We determined MICs of bedaquiline for clinically relevant NTM species and Mycobacterium tuberculosis by broth microdilution for 30 isolates. Synergy testing was performed using the chequerboard method for 22 reference strains and clinical isolates of Mycobacterium abscessus (MAB) and Mycobacterium avium complex (MAC). Time-kill kinetics (TK) assays with resistance monitoring of bedaquiline alone and combined with clofazimine were performed for MAB CIP 104536 and M. avium ATCC 700898; bedaquiline/clarithromycin combinations were evaluated against M. avium ATCC 700898. Interactions were assessed for TK experiments based on Bliss independence. RESULTS Bedaquiline had modest activity against tested NTM, with MICs between <0.007 and 1 mg/L. Bedaquiline showed no interaction with tested drugs against MAB or MAC. Lowest mean fractional inhibitory concentration index (FICI) values were 0.79 with clofazimine for MAB and 0.97 with clofazimine and 0.82 with clarithromycin for MAC. In TK assays, bedaquiline showed a bacteriostatic effect. Clofazimine extended the bacteriostatic activity of bedaquiline against MAB and yielded a slight bactericidal effect against M. avium. The bedaquiline/clofazimine combination slowed emergence of bedaquiline resistance for M. avium but promoted it for MAB. Relative to Bliss independence, bedaquiline/clofazimine showed synergistic interaction over time for MAB and no interaction for M. avium and bedaquiline/clarithromycin showed antagonistic interaction for M. avium. CONCLUSIONS Following these in vitro data, a bedaquiline/clofazimine combination might add activity to MAB and MAC treatment. The bedaquiline/clarithromycin combination might have lower activity compared with bedaquiline alone for MAC treatment.
Collapse
Affiliation(s)
- Mike Marvin Ruth
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jasper J N Sangen
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Karlijn Remmers
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lian J Pennings
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Elin Svensson
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Rob E Aarnoutse
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sanne M H Zweijpfenning
- Department of Pulmonary Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Wouter Hoefsloot
- Department of Pulmonary Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Saskia Kuipers
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Cecile Magis-Escurra
- Department of Pulmonary Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Heiman F L Wertheim
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jakko van Ingen
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
26
|
Yang C, Wang J, Yin Z, Wang Q, Zhang X, Jiang Y, Shen H. A sophisticated antibiotic-loading protocol in articulating cement spacers for the treatment of prosthetic joint infection. Bone Joint Res 2019. [DOI: 10.1302/2046-3758.811.bjr-2019-0339.r3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Objectives The optimal protocol for antibiotic loading in the articulating cement spacers for the treatment of prosthetic joint infection (PJI) remains controversial. The objective of the present study was to investigate the effectiveness of articulating cement spacers loaded with a new combination of antibiotics. Methods A retrospective cohort study involving 114 PJI cases treated with implantation of an articulating cement spacer between 2005 and 2016 was performed. The treatment outcomes of the conventional protocol (i.e. gentamicin and vancomycin (GV protocol)) were compared with those reported using the sophisticated antibiotic-loading protocol (i.e. vancomycin, meropenem, and amphotericin (VMA protocol)). Results There were 62 and 52 PJI cases treated with the GV and VMA protocols, respectively. Antimicrobial susceptibility testing revealed that 22/78 of all isolates (28.2%) in this series were resistant to gentamicin, whereas there were no vancomycin-, meropenem-, or amphotericin-resistant strains. The overall infection recurrence rates were 17.7% (11/62) and 1.9% (1/52), respectively (p = 0.006). In patients with a negative preoperative culture, there was no infection recurrence reported in the VMA cohort (0/45 (0%) vs 10/54 (18.5%) in the GV cohort; p = 0.002). Multivariate analysis indicated that the VMA protocol correlated with a decreased risk of infection recurrence compared with the GV protocol (p = 0.025). Conclusion The sophisticated VMA protocol for the loading of antibiotics in articulating cement spacers, as part of a two-stage exchange, was associated with a reduced rate of infection recurrence. This proposed protocol appears to be safe and effective, especially in patients with negative culture results prior to the first-stage operation. Cite this article: Bone Joint Res 2019;8:526–534.
Collapse
Affiliation(s)
- Chuang Yang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jin Wang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhifei Yin
- Department of Orthopaedic Surgery, Kunshan Traditional Chinese Medicine Hospital, Kunshan, China
| | - Qiaojie Wang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xianlong Zhang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yao Jiang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Hao Shen
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
27
|
Yang C, Wang J, Yin Z, Wang Q, Zhang X, Jiang Y, Shen H. A sophisticated antibiotic-loading protocol in articulating cement spacers for the treatment of prosthetic joint infection: A retrospective cohort study. Bone Joint Res 2019; 8:526-534. [PMID: 31832172 PMCID: PMC6888737 DOI: 10.1302/2046-3758.811.bjr-2018-0339.r3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
OBJECTIVES The optimal protocol for antibiotic loading in the articulating cement spacers for the treatment of prosthetic joint infection (PJI) remains controversial. The objective of the present study was to investigate the effectiveness of articulating cement spacers loaded with a new combination of antibiotics. METHODS A retrospective cohort study involving 114 PJI cases treated with implantation of an articulating cement spacer between 2005 and 2016 was performed. The treatment outcomes of the conventional protocol (i.e. gentamicin and vancomycin (GV protocol)) were compared with those reported using the sophisticated antibiotic-loading protocol (i.e. vancomycin, meropenem, and amphotericin (VMA protocol)). RESULTS There were 62 and 52 PJI cases treated with the GV and VMA protocols, respectively. Antimicrobial susceptibility testing revealed that 22/78 of all isolates (28.2%) in this series were resistant to gentamicin, whereas there were no vancomycin-, meropenem-, or amphotericin-resistant strains. The overall infection recurrence rates were 17.7% (11/62) and 1.9% (1/52), respectively (p = 0.006). In patients with a negative preoperative culture, there was no infection recurrence reported in the VMA cohort (0/45 (0%) vs 10/54 (18.5%) in the GV cohort; p = 0.002). Multivariate analysis indicated that the VMA protocol correlated with a decreased risk of infection recurrence compared with the GV protocol (p = 0.025). CONCLUSION The sophisticated VMA protocol for the loading of antibiotics in articulating cement spacers, as part of a two-stage exchange, was associated with a reduced rate of infection recurrence. This proposed protocol appears to be safe and effective, especially in patients with negative culture results prior to the first-stage operation.Cite this article: Bone Joint Res 2019;8:526-534.
Collapse
Affiliation(s)
- Chuang Yang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jin Wang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhifei Yin
- Department of Orthopaedic Surgery, Kunshan Traditional Chinese Medicine Hospital, Kunshan, China
| | - Qiaojie Wang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xianlong Zhang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yao Jiang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Hao Shen
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
- H. Shen;
| |
Collapse
|
28
|
Auranofin Activity Exposes Thioredoxin Reductase as a Viable Drug Target in Mycobacterium abscessus. Antimicrob Agents Chemother 2019; 63:AAC.00449-19. [PMID: 31262763 DOI: 10.1128/aac.00449-19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 06/23/2019] [Indexed: 02/01/2023] Open
Abstract
Nontuberculous mycobacteria (NTM) are highly drug-resistant, opportunistic pathogens that can cause pulmonary disease. The outcomes of the currently recommended treatment regimens are poor, especially for Mycobacterium abscessus New or repurposed drugs are direly needed. Auranofin, a gold-based antirheumatic agent, was investigated for Mycobacterium tuberculosis Here, we test auranofin against NTM in vitro and ex vivo We tested the susceptibility of 63 NTM isolates to auranofin using broth microdilution. Next, we assessed synergy between auranofin and antimycobacterial drugs using the checkerboard method and calculated the fractional inhibition concentration index (FICI). Using time-kill kinetics assays (TK), we assessed pharmacodynamics of auranofin alone and in combination with drug combinations showing the lowest FICIs for M. abscessus CIP 104536. A response surface analysis was used to assess synergistic interactions over time in TKs. Primary isolated macrophages were infected with M. abscessus and treated with auranofin. Finally, using KEGG Orthology, we looked for orthologues to auranofins drug target in M. tuberculosis M. abscessus had the lowest auranofin MIC50 (2 μg/ml) among the tested NTM. The lowest average FICIs were observed between auranofin and amikacin (0.45) and linezolid (0.50). Auranofin exhibited concentration-dependent killing of M. abscessus, with >1-log killing at concentrations of >2× MIC. Only amikacin was synergistic with auranofin according to Bliss independence. Auranofin could not lower the intracellular bacterial load in macrophages. Auranofin itself may not be feasible for M. abscessus treatment, but these data point toward a promising, unutilized drug target.
Collapse
|
29
|
Clewe O, Wicha SG, de Vogel CP, de Steenwinkel JEM, Simonsson USH. A model-informed preclinical approach for prediction of clinical pharmacodynamic interactions of anti-TB drug combinations. J Antimicrob Chemother 2019; 73:437-447. [PMID: 29136155 PMCID: PMC5890720 DOI: 10.1093/jac/dkx380] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 09/16/2017] [Indexed: 12/27/2022] Open
Abstract
Background Identification of pharmacodynamic interactions is not reasonable to carry out in a clinical setting for many reasons. The aim of this work was to develop a model-informed preclinical approach for prediction of clinical pharmacodynamic drug interactions in order to inform early anti-TB drug development. Methods In vitro time–kill experiments were performed with Mycobacterium tuberculosis using rifampicin, isoniazid or ethambutol alone as well as in different combinations at clinically relevant concentrations. The multistate TB pharmacometric (MTP) model was used to characterize the natural growth and exposure–response relationships of each drug after mono exposure. Pharmacodynamic interactions during combination exposure were characterized by linking the MTP model to the general pharmacodynamic interaction (GPDI) model with successful separation of the potential effect on each drug’s potency (EC50) by the combining drug(s). Results All combinations showed pharmacodynamic interactions at cfu level, where all combinations, except isoniazid plus ethambutol, showed more effect (synergy) than any of the drugs alone. Using preclinical information, the MTP-GPDI modelling approach was shown to correctly predict clinically observed pharmacodynamic interactions, as deviations from expected additivity. Conclusions With the ability to predict clinical pharmacodynamic interactions, using preclinical information, the MTP-GPDI model approach outlined in this study constitutes groundwork for model-informed input to the development of new and enhancement of existing anti-TB combination regimens.
Collapse
Affiliation(s)
- Oskar Clewe
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Sebastian G Wicha
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Corné P de Vogel
- Department of Medical Microbiology and Infectious Diseases, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Jurriaan E M de Steenwinkel
- Department of Medical Microbiology and Infectious Diseases, Erasmus Medical Centre, Rotterdam, The Netherlands
| | | |
Collapse
|
30
|
Wicha SG, Chen C, Clewe O, Simonsson USH. A general pharmacodynamic interaction model identifies perpetrators and victims in drug interactions. Nat Commun 2017; 8:2129. [PMID: 29242552 PMCID: PMC5730559 DOI: 10.1038/s41467-017-01929-y] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 10/25/2017] [Indexed: 12/20/2022] Open
Abstract
Assessment of pharmacodynamic (PD) drug interactions is a cornerstone of the development of combination drug therapies. To guide this venture, we derive a general pharmacodynamic interaction (GPDI) model for ≥2 interacting drugs that is compatible with common additivity criteria. We propose a PD interaction to be quantifiable as multidirectional shifts in drug efficacy or potency and explicate the drugs’ role as victim, perpetrator or even both at the same time. We evaluate the GPDI model against conventional approaches in a data set of 200 combination experiments in Saccharomyces cerevisiae: 22% interact additively, a minority of the interactions (11%) are bidirectional antagonistic or synergistic, whereas the majority (67%) are monodirectional, i.e., asymmetric with distinct perpetrators and victims, which is not classifiable by conventional methods. The GPDI model excellently reflects the observed interaction data, and hence represents an attractive approach for quantitative assessment of novel combination therapies along the drug development process. Assessment of pharmacodynamic interactions is at the heart of combination therapy development. Here the authors introduce a general drug interaction scoring model that enables quantification of synergistic and antagonistic interactions and determination of the directionality of the interactions.
Collapse
Affiliation(s)
- Sebastian G Wicha
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, 75124, Sweden.
| | - Chunli Chen
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, 75124, Sweden
| | - Oskar Clewe
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, 75124, Sweden
| | - Ulrika S H Simonsson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, 75124, Sweden
| |
Collapse
|
31
|
Chen C, Wicha SG, de Knegt GJ, Ortega F, Alameda L, Sousa V, de Steenwinkel JEM, Simonsson USH. Assessing Pharmacodynamic Interactions in Mice Using the Multistate Tuberculosis Pharmacometric and General Pharmacodynamic Interaction Models. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2017; 6:787-797. [PMID: 28657202 PMCID: PMC5702905 DOI: 10.1002/psp4.12226] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 06/07/2017] [Accepted: 06/11/2017] [Indexed: 02/04/2023]
Abstract
The aim of this study was to investigate pharmacodynamic (PD) interactions in mice infected with Mycobacterium tuberculosis using population pharmacokinetics (PKs), the Multistate Tuberculosis Pharmacometric (MTP) model, and the General Pharmacodynamic Interaction (GPDI) model. Rifampicin, isoniazid, ethambutol, or pyrazinamide were administered in monotherapy for 4 weeks. Rifampicin and isoniazid showed effects in monotherapy, whereas the animals became moribund after 7 days with ethambutol or pyrazinamide alone. No PD interactions were observed against fast‐multiplying bacteria. Interactions between rifampicin and isoniazid on killing slow and non‐multiplying bacteria were identified, which led to an increase of 0.86 log10 colony‐forming unit (CFU)/lungs at 28 days after treatment compared to expected additivity (i.e., antagonism). An interaction between rifampicin and ethambutol on killing non‐multiplying bacteria was quantified, which led to a decrease of 2.84 log10 CFU/lungs at 28 days after treatment (i.e., synergism). These results show the value of pharmacometrics to quantitatively assess PD interactions in preclinical tuberculosis drug development.
Collapse
Affiliation(s)
- Chunli Chen
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden.,College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin, 150030, P. R. China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, 600 Changjiang Road, Xiangfang District, Harbin, 150030, P. R. China
| | - Sebastian G Wicha
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Gerjo J de Knegt
- Erasmus Medical Center, Department of Medical Microbiology and Infectious Disease, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Fatima Ortega
- Diseases of Developing World Medicines Development Campus, GlaxoSmithKline, Tres Cantos, Madrid, Spain
| | - Laura Alameda
- Diseases of Developing World Medicines Development Campus, GlaxoSmithKline, Tres Cantos, Madrid, Spain
| | - Veronica Sousa
- Diseases of Developing World Medicines Development Campus, GlaxoSmithKline, Tres Cantos, Madrid, Spain
| | - Jurriaan E M de Steenwinkel
- Erasmus Medical Center, Department of Medical Microbiology and Infectious Disease, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | | |
Collapse
|
32
|
Suppressive drug combinations and their potential to combat antibiotic resistance. J Antibiot (Tokyo) 2017; 70:1033-1042. [PMID: 28874848 DOI: 10.1038/ja.2017.102] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 07/26/2017] [Accepted: 07/28/2017] [Indexed: 12/25/2022]
Abstract
Antibiotic effectiveness often changes when two or more such drugs are administered simultaneously and unearthing antibiotic combinations with enhanced efficacy (synergy) has been a longstanding clinical goal. However, antibiotic resistance, which undermines individual drugs, threatens such combined treatments. Remarkably, it has emerged that antibiotic combinations whose combined effect is lower than that of at least one of the individual drugs can slow or even reverse the evolution of resistance. We synthesize and review studies of such so-called 'suppressive interactions' in the literature. We examine why these interactions have been largely disregarded in the past, the strategies used to identify them, their mechanistic basis, demonstrations of their potential to reverse the evolution of resistance and arguments for and against using them in clinical treatment. We suggest future directions for research on these interactions, aiming to expand the basic body of knowledge on suppression and to determine the applicability of suppressive interactions in the clinic.
Collapse
|
33
|
Wicha SG, Huisinga W, Kloft C. Translational Pharmacometric Evaluation of Typical Antibiotic Broad-Spectrum Combination Therapies Against Staphylococcus Aureus Exploiting In Vitro Information. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2017; 6:512-522. [PMID: 28378945 PMCID: PMC5572409 DOI: 10.1002/psp4.12197] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 03/24/2017] [Accepted: 03/29/2017] [Indexed: 11/15/2022]
Abstract
Broad‐spectrum antibiotic combination therapy is frequently applied due to increasing resistance development of infective pathogens. The objective of the present study was to evaluate two common empiric broad‐spectrum combination therapies consisting of either linezolid (LZD) or vancomycin (VAN) combined with meropenem (MER) against Staphylococcus aureus (S. aureus) as the most frequent causative pathogen of severe infections. A semimechanistic pharmacokinetic‐pharmacodynamic (PK‐PD) model mimicking a simplified bacterial life‐cycle of S. aureus was developed upon time‐kill curve data to describe the effects of LZD, VAN, and MER alone and in dual combinations. The PK‐PD model was successfully (i) evaluated with external data from two clinical S. aureus isolates and further drug combinations and (ii) challenged to predict common clinical PK‐PD indices and breakpoints. Finally, clinical trial simulations were performed that revealed that the combination of VAN‐MER might be favorable over LZD‐MER due to an unfavorable antagonistic interaction between LZD and MER.
Collapse
Affiliation(s)
- S G Wicha
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Berlin, Germany
| | - W Huisinga
- Institute of Mathematics, University of Potsdam, Potsdam-Golm, Germany
| | - C Kloft
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Berlin, Germany
| |
Collapse
|
34
|
Ferrone V, Carlucci M, Cotellese R, Raimondi P, Cichella A, Marco LD, Carlucci G. Development and validation of a fast micro-extraction by packed sorbent UHPLC-PDA method for the simultaneous determination of linezolid and ciprofloxacin in human plasma from patients with hospital-acquired pneumonia. Talanta 2016; 164:64-68. [PMID: 28107984 DOI: 10.1016/j.talanta.2016.11.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 11/07/2016] [Accepted: 11/08/2016] [Indexed: 10/20/2022]
Abstract
An ultra high-performance liquid chromatographic (UHPLC) method with PDA detection was developed and validated for the simultaneous quantification of linezolid and ciprofloxacin in human plasma and applied in hospital acquired pneumonia patients (HAP). The method uses a semi-automated microextraction by packed sorbent for sample preparation. All parameters in the extraction step (pH, sample volume, sample dilution and number of aspiration - ejection cycles) and in the desorption step (percentage of acetonitrile in the solvent of elution and number of aspirations of elution solvent through the device) were statistically significant when the recovery was used as response. The method showed good linearity with correlation coefficients, r2>0.9995 for the two drugs, as well as high precision (RSD%<9.77% in each case), accuracy ranged from -6.2% to +8.2. The limit of quantification of the two drugs was established at 0.01 and 0.02μg/mL for ciprofloxacin and linezolid, respectively. Linezolid, ciprofloxacin and internal standard were extracted from human plasma with a mean recovery ranging from 92.4% to 97.4%. During validation, the concentrations of linezolid and ciprofloxacin were found to be stable after 3 freeze-thaw cycles and for at least 24h after extraction. This method will subsequently be used to quantify the drugs dosage in patients with HAP to establish if the dosage regimen given is sufficient to eradicate the infection at the target site.
Collapse
Affiliation(s)
- Vincenzo Ferrone
- Dipartimento di Farmacia, Università degli Studi "G. d'Annunzio" Chieti-Pescara, via dei Vestini, 66100 Chieti, Italy
| | - Maura Carlucci
- Dipartimento di Scienze Mediche Orali e Biotecnologiche, Università degli Studi "G. d'Annunzio" Chieti-Pescara, via dei Vestini, 66100 Chieti, Italy
| | - Roberto Cotellese
- Dipartimento di Scienze Mediche Orali e Biotecnologiche, Università degli Studi "G. d'Annunzio" Chieti-Pescara, via dei Vestini, 66100 Chieti, Italy
| | - Paolo Raimondi
- Dipartimento di Scienze Mediche Orali e Biotecnologiche, Università degli Studi "G. d'Annunzio" Chieti-Pescara, via dei Vestini, 66100 Chieti, Italy
| | - Annadomenica Cichella
- Dipartimento di Scienze Mediche Orali e Biotecnologiche, Università degli Studi "G. d'Annunzio" Chieti-Pescara, via dei Vestini, 66100 Chieti, Italy
| | - Lorenzo Di Marco
- Dipartimento di Farmacia, Università degli Studi "G. d'Annunzio" Chieti-Pescara, via dei Vestini, 66100 Chieti, Italy
| | - Giuseppe Carlucci
- Dipartimento di Farmacia, Università degli Studi "G. d'Annunzio" Chieti-Pescara, via dei Vestini, 66100 Chieti, Italy.
| |
Collapse
|
35
|
Wicha SG, Kloft C. Simultaneous determination and stability studies of linezolid, meropenem and vancomycin in bacterial growth medium by high-performance liquid chromatography. J Chromatogr B Analyt Technol Biomed Life Sci 2016; 1028:242-248. [DOI: 10.1016/j.jchromb.2016.06.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 06/16/2016] [Accepted: 06/19/2016] [Indexed: 10/21/2022]
|
36
|
In vitro time-kill curves study of three antituberculous combinations against Mycobacterium tuberculosis clinical isolates. Int J Antimicrob Agents 2015; 47:97-100. [PMID: 26691020 DOI: 10.1016/j.ijantimicag.2015.10.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 09/30/2015] [Accepted: 10/02/2015] [Indexed: 11/22/2022]
Abstract
The objective of this study was to examine the in vitro synergism of three-drug combinations against Mycobacterium tuberculosis (levofloxacin/linezolid/ethambutol, levofloxacin/amikacin/ethambutol and levofloxacin/linezolid/amikacin) using the time-kill curves method. In total, 8 multidrug-resistant and 12 drug-susceptible M. tuberculosis isolates were used. Minimum inhibitory concentrations (MICs) of the isolates for each drug were determined by the proportions method. Time-kill curves were studied for the three combinations proposed over 14 days using two different protocols. In protocol 1, 0.5× MIC for each drug was used. In protocol 2, 0.5× MIC for levofloxacin and linezolid and 0.25× MIC for amikacin and ethambutol were used. The MICs for all of the isolates studied were 0.5 mg/L for levofloxacin and linezolid and 2.5 mg/L for ethambutol and amikacin. All of the combinations displayed an additive activity compared with the most active individual drug. In conclusion, these results demonstrate that the three combinations tested were equally effective against M. tuberculosis isolates. The study of antituberculous combinations using in vitro methods is an excellent first step to predict their effect in clinical development phases as well as to test new regimens of the antituberculous drugs currently available.
Collapse
|
37
|
Douros A, Grabowski K, Stahlmann R. Drug–drug interactions and safety of linezolid, tedizolid, and other oxazolidinones. Expert Opin Drug Metab Toxicol 2015; 11:1849-59. [DOI: 10.1517/17425255.2015.1098617] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|