1
|
Blondeau JM, Fitch SD. Effects of Comparative Killing by Pradofloxacin and Seven Other Antimicrobials Against Varying Bacterial Densities of Swine Isolates of Pasteurella multocida. Microorganisms 2025; 13:221. [PMID: 40005588 PMCID: PMC11857592 DOI: 10.3390/microorganisms13020221] [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: 11/28/2024] [Revised: 12/27/2024] [Accepted: 01/17/2025] [Indexed: 02/27/2025] Open
Abstract
Bacterial killing is important for recovering from infection. Pasteurella multocida is a key bacterial pathogen causing swine respiratory disease and is associated with substantial mortality. Antimicrobial therapy remains an important therapeutic intervention for treating infected animals. Pradofloxacin (fluoroquinolone) is the most recently approved antimicrobial agent for treating pigs with swine respiratory disease. We compared in vitro killing of swine P. multocida strains by pradofloxacin in comparison to ceftiofur, enrofloxacin, florfenicol, marbofloxacin, tildipirosin, tilmicosin, and tulathromycin over a range of bacterial densities and four clinically relevant drug concentrations. Pradofloxacin killed 92-96.9% of cells across 106-108 cfu/mL densities at the mutant prevention drug concentration following 2-24 h of drug exposure, 96.9-98.9% of cells across 106-109 cfu/mL at the maximum serum drug concentration following 30 min of drug exposure, increasing to 99.9-100% kill following 12-24 h of drug exposure. At the maximum tissue drug concentration and against bacterial densities of 106-109 cfu/mL, pradofloxacin killed 91.3-99.8% of cells following 2 h of drug exposure, which increased to 99.9-100% kill following 12-24 h of drug exposure. Pradofloxacin was rapidly bactericidal across a range of bacterial densities and at clinically relevant drug concentrations. Pradofloxacin will be an important antibiotic for treating pigs with swine respiratory disease and where clinically indicated.
Collapse
Affiliation(s)
- Joseph M. Blondeau
- Department of Clinical Microbiology, Royal University Hospital and Saskatchewan Health Authority, Saskatoon, SK S7N 0W8, Canada;
- Pathology and Laboratory Medicine and Ophthalmology, Departments of Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada
| | - Shantelle D. Fitch
- Department of Clinical Microbiology, Royal University Hospital and Saskatchewan Health Authority, Saskatoon, SK S7N 0W8, Canada;
| |
Collapse
|
2
|
Blondeau JM, Fitch SD. Comparison of the Minimum Inhibitory and Mutant Prevention Drug Concentrations for Pradofloxacin and 7 Other Antimicrobial Agents Tested Against Swine Isolates of Actinobacillus pleuropneumoniae and Pasteurella multocida. Molecules 2024; 29:5448. [PMID: 39598838 PMCID: PMC11597606 DOI: 10.3390/molecules29225448] [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: 09/12/2024] [Revised: 11/12/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
Pradofloxacin is a dual targeting, bactericidal fluoroquinolone recently approved for treating bacteria causing swine respiratory disease. Currently, an abundance of in vitro data does not exist for pradofloxacin. We determined the minimum inhibitory concentration (MIC) and mutant prevention concentrations (MPC) of pradofloxacin compared to ceftiofur, enrofloxacin, florfenicol, marbofloxacin, tildipirosin, tilmicosin and tulathromycin against swine isolates of Actinobacillus pleuropneumoniae and Pasteurella multocida. Overall, pradofloxacin had the lowest MIC and MPC values as compared to the other agents tested. For example, pradofloxacin MIC values for 50%, 90% and 100% of A. pleuropneumoniae strains were ≤0.016 µg/mL, ≤0.016 µg/mL and ≤0.016 µg/mL and for P. multocida were ≤0.016 µg/mL, ≤0.016 µg/mL and 0.031 µg/mL, respectively. The MPC values for 50%, 90% and 100% of A. pleuropneumoniae strains were 0.031 µg/mL, 0.063 µg/mL and 0.125 µg/mL and for P. multocida were ≤0.016 µg/mL, 0.031 µg/mL and 0.0.063 µg/mL, respectively. By MPC testing, all strains were at or below the susceptibility breakpoint. Based on MPC testing, pradofloxacin appears to have a low likelihood for resistance selection. This study represents the most comprehensive in vitro comparison of the above noted drugs and the first report for pradofloxacin and tildipirosin.
Collapse
Affiliation(s)
- Joseph M. Blondeau
- Departments of Biochemistry, Microbiology and Immunology, Pathology and Laboratory Medicine and Ophthalmology, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada
- Department of Clinical Microbiology, Royal University Hospital and Saskatchewan Health Authority, Saskatoon, SK S7N 0W8, Canada;
| | - Shantelle D. Fitch
- Department of Clinical Microbiology, Royal University Hospital and Saskatchewan Health Authority, Saskatoon, SK S7N 0W8, Canada;
| |
Collapse
|
3
|
Blondeau JM, Blondeau LD, Fitch SD. In vitro killing of drug susceptible and multidrug resistant bacteria by amikacin considering pulmonary drug concentrations based on an inhaled formulation. J Chemother 2024; 36:389-397. [PMID: 38339845 DOI: 10.1080/1120009x.2024.2313908] [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: 06/08/2023] [Revised: 12/18/2023] [Accepted: 01/25/2024] [Indexed: 02/12/2024]
Abstract
Nosocomial infections with drug resistant bacteria impact morbidity and mortality, length of therapy and stay and the overall cost of treatment. Key pathogens with ventilator associated pneumonia may be drug-susceptible or multi-drug resistant and inhaled amikacin has been investigated as an adjunctive therapy option. High pulmonary drug concentrations (epithelial lining fluid [ELF]) along with minimal systemic toxicity is seen as an advantage to inhaled formulations. In vitro killing of bacteria using clinically relevant drug concentrations provide insight on bug-drug interactions. The aim of this study was to measure killing of clinical isolates of Acinetobacter baumannii, Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa, methicillin-resistant Staphylococcus aureus and methicillin-susceptible S. aureus using the minimum inhibitory concentration (MIC), mutant prevention concentration (MPC) and median (976 µg/ml) ELF drug concentration for amikacin. Overall killing took longer at the MIC drug concentration and was inconsistent amongst the pathogens tested with the percentage of bacteria killed following 180 min of drug exposure ranging from growth in the presence of the drug to 95% kill. At the MPC drug concentrations, killing ranged from 55-88% for all pathogens following 30 min of drug exposure and increased to 99-100% following 180 min of drug exposure. At the ELF amikacin tested, killing was 81-100% following 20 min and 94-100% by 30 min of drug exposure. Rapid killing against MDR respiratory pathogens by amikacin ELF drug concentrations is encouraging.
Collapse
Affiliation(s)
- Joseph M Blondeau
- Division of Clinical Microbiology, Royal University Hospital and Saskatchewan Health Authority, Saskatoon, Saskatchewan, Canada
- Departments of Microbiology and Immunology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Pathology and Ophthalmology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Leah D Blondeau
- Division of Clinical Microbiology, Royal University Hospital and Saskatchewan Health Authority, Saskatoon, Saskatchewan, Canada
| | - Shantelle D Fitch
- Division of Clinical Microbiology, Royal University Hospital and Saskatchewan Health Authority, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
4
|
Krajewska J, Tyski S, Laudy AE. In Vitro Resistance-Predicting Studies and In Vitro Resistance-Related Parameters-A Hit-to-Lead Perspective. Pharmaceuticals (Basel) 2024; 17:1068. [PMID: 39204172 PMCID: PMC11357384 DOI: 10.3390/ph17081068] [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: 07/26/2024] [Revised: 08/10/2024] [Accepted: 08/12/2024] [Indexed: 09/03/2024] Open
Abstract
Despite the urgent need for new antibiotics, very few innovative antibiotics have recently entered clinics or clinical trials. To provide a constant supply of new drug candidates optimized in terms of their potential to select for resistance in natural settings, in vitro resistance-predicting studies need to be improved and scaled up. In this review, the following in vitro parameters are presented: frequency of spontaneous mutant selection (FSMS), mutant prevention concentration (MPC), dominant mutant prevention concentration (MPC-D), inferior-mutant prevention concentration (MPC-F), and minimal selective concentration (MSC). The utility of various adaptive laboratory evolution (ALE) approaches (serial transfer, continuous culture, and evolution in spatiotemporal microenvironments) for comparing hits in terms of the level and time required for multistep resistance to emerge is discussed. We also consider how the hit-to-lead stage can benefit from high-throughput ALE setups based on robotic workstations, do-it-yourself (DIY) continuous cultivation systems, microbial evolution and growth arena (MEGA) plates, soft agar gradient evolution (SAGE) plates, microfluidic chips, or microdroplet technology. Finally, approaches for evaluating the fitness of in vitro-generated resistant mutants are presented. This review aims to draw attention to newly emerged ideas on how to improve the in vitro forecasting of the potential of compounds to select for resistance in natural settings.
Collapse
Affiliation(s)
- Joanna Krajewska
- Department of Environmental Health and Safety, National Institute of Public Health NIH—National Research Institute, 00-791 Warsaw, Poland;
| | - Stefan Tyski
- Department of Pharmaceutical Microbiology and Laboratory Diagnostic, National Medicines Institute, 00-725 Warsaw, Poland;
| | - Agnieszka E. Laudy
- Department of Pharmaceutical Microbiology and Bioanalysis, Medical University of Warsaw, 02-097 Warsaw, Poland
| |
Collapse
|
5
|
Jariyapamornkoon N, Nuanualsuwan S, Suanpairintr N. In Vitro Antibacterial Activities of Fosfomycin against Escherichia coli Isolates from Canine Urinary Tract Infection. Animals (Basel) 2024; 14:1916. [PMID: 38998027 PMCID: PMC11240368 DOI: 10.3390/ani14131916] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 06/23/2024] [Accepted: 06/27/2024] [Indexed: 07/14/2024] Open
Abstract
Fosfomycin is a bactericidal drug recommended as an alternative treatment for canine bacterial cystitis, particularly in cases involving multidrug-resistant (MDR) infections when no other options are available. In this study, minimum inhibitory concentration (MIC) and mutant prevention concentration (MPC) of fosfomycin were determined against 79 clinical E. coli isolates using the agar dilution method. The susceptibility rate of E. coli to fosfomycin was 86.06%, with MIC50 and MIC90 values of 4 mg/L and 96 mg/L, respectively. MPC50 and MPC90 values were 64 mg/L and 192 mg/L. Using pharmacokinetic (PK) data from dogs given a single 80 mg/kg oral dose of fosfomycin, the area under the curve per MIC50 (AUC0-24/MIC50) was 85.79 with time above MIC50 (T > MIC50) exceeding 50%. In urine, the AUC0-24/MIC50 was 10,694.78, and the AUC0-24/MPC90 was 222.81, with T > MPC90 extending beyond 24 h. Therefore, fosfomycin exhibited significant antibacterial activity against canine uropathogenic E. coli, including MDR strains, at concentrations below the susceptible MIC breakpoint. However, the high MPC values, especially the MPC90, indicate the critical importance of performing susceptibility testing for fosfomycin and maintaining ongoing resistance monitoring.
Collapse
Affiliation(s)
- Nattha Jariyapamornkoon
- Graduate Program in Veterinary Bioscience, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Suphachai Nuanualsuwan
- Department of Veterinary Public Health, Faculty of Veterinary Sciences, Chulalongkorn University, Bangkok 10330, Thailand;
- Center of Excellence for Food and Water Risk Analysis (FAWRA), Department of Veterinary Public Health, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Nipattra Suanpairintr
- Center of Excellence for Food and Water Risk Analysis (FAWRA), Department of Veterinary Public Health, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Pharmacology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
6
|
Blondeau JM, Fitch SD. Comparative In Vitro Killing by Pradofloxacin in Comparison to Ceftiofur, Enrofloxacin, Florfenicol, Marbofloxacin, Tildipirosin, Tilmicosin and Tulathromycin against Bovine Respiratory Bacterial Pathogens. Microorganisms 2024; 12:996. [PMID: 38792823 PMCID: PMC11123926 DOI: 10.3390/microorganisms12050996] [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: 04/16/2024] [Revised: 05/03/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Pradofloxacin is the newest of the veterinary fluoroquinolones to be approved for use in animals-initially companion animals and most recently food animals. It has a broad spectrum of in vitro activity, working actively against Gram-positive/negative, atypical and some anaerobic microorganisms. It simultaneously targets DNA gyrase (topoisomerase type II) and topoisomerase type IV, suggesting a lower propensity to select for antimicrobial resistance. The purpose of this study was to determine the rate and extent of bacterial killing by pradofloxacin against bovine strains of Mannheimia haemolytica and Pasteurella multocida, in comparison with several other agents (ceftiofur, enrofloxacin, florfenicol, marbofloxacin, tildipirosin, tilmicosin and tulathromycin) using four clinically relevant drug concentrations: minimum inhibitory and mutant prevention drug concentration, maximum serum and maximum tissue drug concentrations. At the maximum serum and tissue drug concentrations, pradofloxacin killed 99.99% of M. haemolytica cells following 5 min of drug exposure (versus growth to 76% kill rate for the other agents) and 94.1-98.6% of P. multocida following 60-120 min of drug exposure (versus growth to 98.6% kill rate for the other agents). Statistically significant differences in kill rates were seen between the various drugs tested depending on drug concentration and time of sampling after drug exposure.
Collapse
Affiliation(s)
- Joseph M. Blondeau
- Department of Clinical Microbiology, Royal University Hospital and Saskatchewan Health Authority, Saskatoon, SK S7N 0W8, Canada;
- Pathology and Laboratory Medicine and Ophthalmology, Departments Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada
| | - Shantelle D. Fitch
- Department of Clinical Microbiology, Royal University Hospital and Saskatchewan Health Authority, Saskatoon, SK S7N 0W8, Canada;
| |
Collapse
|
7
|
Blondeau JM, Fitch SD. Comparative Minimum Inhibitory and Mutant Prevention Drug Concentrations for Pradofloxacin and Seven Other Antimicrobial Agents Tested against Bovine Isolates of Mannheimia haemolytica and Pasteurella multocida. Pathogens 2024; 13:399. [PMID: 38787251 PMCID: PMC11123865 DOI: 10.3390/pathogens13050399] [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: 04/16/2024] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024] Open
Abstract
Pradofloxacin-a dual-targeting fluoroquinolone-is the most recent approved for use in food animals. Minimum inhibitory and mutant prevention concentration values were determined for pradofloxacin, ceftiofur, enrofloxacin, florfenicol, marbofloxacin, tildipirosin, tilmicosin, and tulathromycin. For M. haemolytica strains, MIC50/90/100 values were ≤0.016/≤0.016/≤0.016 and MPC50/90/100 values were 0.031/0.063/0.063; for P. multocida strains, the MIC50/90/100 values ≤0.016/≤0.016/0.031 and MPC50/90/100 ≤ 0.016/0.031/0.063 for pradofloxacin. The pradofloxacin Cmax/MIC90 and Cmax/MPC90 values for M. haemolytica and P. multocida strains, respectively, were 212.5 and 53.9 and 212.5 and 109.7. Similarly, AUC24/MIC90 and AUC24/MPC90 for M. haemolytica were 825 and 209.5, and for P. multocida, they were 825 and 425.8. Pradofloxacin would exceed the mutant selection window for >12-16 h. Pradofloxacin appears to have a low likelihood for resistance selection against key bovine respiratory disease bacterial pathogens based on low MIC and MPC values.
Collapse
Affiliation(s)
- Joseph M. Blondeau
- Department of Clinical Microbiology, Royal University Hospital and Saskatchewan Health Authority, Saskatoon, SK S7N 0W8, Canada;
- Department of Biochemistry, Microbiology and Immunology, Pathology and Laboratory Medicine and Ophthalmology, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada
| | - Shantelle D. Fitch
- Department of Clinical Microbiology, Royal University Hospital and Saskatchewan Health Authority, Saskatoon, SK S7N 0W8, Canada;
| |
Collapse
|
8
|
Chukwu KB, Abafe OA, Amoako DG, Ismail A, Essack SY, Abia ALK. Impact of Environmental Sub-Inhibitory Concentrations of Antibiotics, Heavy Metals, and Biocides on the Emergence of Tolerance and Effects on the Mutant Selection Window in E. coli. Microorganisms 2023; 11:2265. [PMID: 37764108 PMCID: PMC10535725 DOI: 10.3390/microorganisms11092265] [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: 07/16/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Bacteria's ability to withstand the detrimental effects of antimicrobials could occur as resistance or tolerance with the minimum inhibitory concentration, the mutant prevention concentration, and the mutant selection window as salient concepts. Thus, this study assessed the impact of exposure to extremely high doses of ampicillin on the level of persistence and tolerance development in isolates previously exposed to different concentrations of selected antibiotics, biocides, and heavy metals. These isolates were previously exposed to oxytetracycline (OXYTET), amoxicillin (AMX), copper (Cu), zinc (Zn), benzalkonium chloride (BAC) 10, dimethylammonium chloride (DADMAC) 12 and a combination of all the individual pollutants (ALL). The isolates were exposed to very high concentrations (25 × MIC) of ampicillin, and their tolerance was calculated as the time required to kill 99.9% of the bacterial population (MDK99.9). The MDK99.9 increased by 30 to 50% in test isolates (DADMAC, OXYTET, Zinc = 28 h; BAC, Copper = 30 h; amoxycillin, ALL = 26 h) compared to the untreated control. BAC-exposed isolates decreased from 2.5 × 108 CFU/mL to 2.5 × 104 CFU/mL on the second day, displaying the highest tolerance increase. The tolerance appeared to originate from two sources, i.e., stochastic persistence and genetic-induced persistence, involving multiple genes with diverse mechanisms. The mutant selection window of the isolates to ampicillin, amoxicillin, and oxytetracycline also slightly increased compared to the control, indicating the selective survival of persister cells during the 30-day exposure. These findings indicate that bacterial exposure to sub-inhibitory concentrations of environmental chemical stressors may not always result in the development of antimicrobial resistance but could initiate this process by selecting persisters that could evolve into resistant isolates.
Collapse
Affiliation(s)
- Kelechi B. Chukwu
- Antimicrobial Research Unit, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa; (K.B.C.); (O.A.A.); (D.G.A.)
| | - Ovokeroye A. Abafe
- Antimicrobial Research Unit, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa; (K.B.C.); (O.A.A.); (D.G.A.)
- Residue Laboratory, Agricultural Research Council, Onderstepoort Veterinary Research Campus, Onderstepoort 0110, South Africa
- School of Geography, Earth and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Daniel G. Amoako
- Antimicrobial Research Unit, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa; (K.B.C.); (O.A.A.); (D.G.A.)
- Department of Integrative Biology and Bioinformatics, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Arshad Ismail
- Sequencing Core Facility, National Institute for Communicable Diseases, National Health Laboratory Service, Johannesburg 2192, South Africa;
- Department of Biochemistry and Microbiology, University of Venda, Thohoyandou 0950, South Africa
| | - Sabiha Y. Essack
- Antimicrobial Research Unit, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa; (K.B.C.); (O.A.A.); (D.G.A.)
| | - Akebe L. K. Abia
- Antimicrobial Research Unit, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa; (K.B.C.); (O.A.A.); (D.G.A.)
- Environmental Research Foundation, Westville 3630, South Africa
| |
Collapse
|
9
|
Bergmann F, Nussbaumer-Pröll A, Wulkersdorfer B, Eberl S, Ruppitsch W, Lepuschitz S, Zeitlinger M. Antimicrobial activity and pathogen mutation prevention of originator and generics of cefepime, linezolid and piperacillin/tazobactam against clinical isolates of Staphylococcus aureus. J Glob Antimicrob Resist 2023; 34:179-185. [PMID: 37473915 DOI: 10.1016/j.jgar.2023.07.010] [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: 03/31/2023] [Revised: 07/02/2023] [Accepted: 07/12/2023] [Indexed: 07/22/2023] Open
Abstract
OBJECTIVES Although generic medicinal products are required to have the same qualitative and quantitative composition of the active substance as their reference originator product, patients and health care professionals express concerns about their interchangeability and safety. Therefore, the present study investigated the antimicrobial activity and pathogen mutation prevention of original and generic cefepime, linezolid and piperacillin/tazobactam against Staphylococcus aureus. METHODS Two generic formulations of cefepime, linezolid and piperacillin/tazobactam were tested against their respective originator products. Susceptibility testing was performed with twenty-one clinical isolates of S. aureus and ATCC-29213 using broth microdilution. Time kill curves (TKC) were performed with ATCC-29213 at drug concentrations above and below the respective minimum inhibitory concentrations (MIC). Mutation prevention concentration was determined for each drug formulation against ATCC-29213. All experiments were performed in triplicate. Mutant colonies from mutation prevention concentration (MPC) experiments were genotypically tested by sequence analysis. RESULTS MIC ratios between contiguous originator and generic drugs were similar for each isolate. No visual differences were observed in TKCs between originator and generic substances. The MPC did not differ between different formulations of the same substance. Although sequence analysis of mutant colonies revealed genomic differences compared with the original ATCC-29213, no differences in mutation frequencies were observed between clinical isolates and ATCC-29213 treated with originator or generic substances. CONCLUSIONS Similar antimicrobial activity and pathogen mutation prevention was observed between contiguous substances. These results support the interchangeability of generic and originator drug formulations with the same active ingredient.
Collapse
Affiliation(s)
- Felix Bergmann
- Medical University of Vienna, Department of Clinical Pharmacology, Vienna, Austria; Medical University of Vienna, Clinical Division of Plastic and Reconstructive Surgery, Department of Surgery, Vienna, Austria
| | | | | | - Sabine Eberl
- Medical University of Vienna, Department of Clinical Pharmacology, Vienna, Austria
| | - Werner Ruppitsch
- AGES - Austrian Agency for Health and Food Safety, Institute of Medical Microbiology and Hygiene, Vienna, Austria
| | - Sarah Lepuschitz
- AGES - Austrian Agency for Health and Food Safety, Institute of Medical Microbiology and Hygiene, Vienna, Austria
| | - Markus Zeitlinger
- Medical University of Vienna, Department of Clinical Pharmacology, Vienna, Austria.
| |
Collapse
|
10
|
Mutant Prevention Concentrations of Ciprofloxacin and Levofloxacin and Target Gene Mutations of Fluoroquinolones in Elizabethkingia anophelis. Antimicrob Agents Chemother 2022; 66:e0030122. [PMID: 35708332 DOI: 10.1128/aac.00301-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Fluoroquinolones are potentially effective against Elizabethkingia anophelis. We investigated the MIC, mutant prevention concentration (MPC), and target gene mutations of fluoroquinolones in E. anophelis. Eighty-five E. anophelis isolates were collected from five hospitals in Taiwan. The MIC and MPC of ciprofloxacin and levofloxacin were examined for all E. anophelis except 17 isolates, in which ciprofloxacin MPC could not be determined due to drug precipitation caused by overly high drug concentration. Mutations in the quinolone resistance-determining regions of DNA gyrase (GyrA and GyrB) and topoisomerase IV (ParC and ParE) in the clinical isolates and fluoroquinolone-selected mutants were examined. Overall, 23.5% and 71.8% of the isolates tested were susceptible to ciprofloxacin and levofloxacin, respectively. The MPC50 of ciprofloxacin was 128 mg/L, and the MPC50 of levofloxacin was 51.2 mg/L. The MPC50/MIC50 ratio for ciprofloxacin was 64, whereas that for levofloxacin was 25.6. The coefficient of determination between the MPC and MIC for ciprofloxacin and levofloxacin was 0.72 and 0.56, respectively, in the linear regression analysis. Preexisting mutations in GyrA (S83I, S83R, and D87Y) were identified in 18 clinical isolates, all of which were resistant to both ciprofloxacin and levofloxacin. Additional amino acid substitutions in GyrA were identified in all ciprofloxacin- and levofloxacin-selected mutants. Furthermore, GyrB alterations (D431N or D431H) were found in nine levofloxacin-treated isolates. Given that maintaining the serum concentrations of fluoroquinolones above MPCs is impossible under presently recommended doses, the selection of mutant E. anophelis strains seems inevitable.
Collapse
|
11
|
Cengiz M, Sahinturk P, Hepbostanci G, Akalin H, Sonal S. The in vitro activity of danofloxacin plus ceftiofur combination: implications for antimicrobial efficacy and resistance prevention. VETERINARY RESEARCH FORUM : AN INTERNATIONAL QUARTERLY JOURNAL 2022; 13:149-153. [PMID: 35919857 PMCID: PMC9340295 DOI: 10.30466/vrf.2020.113272.2696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 02/12/2020] [Indexed: 11/13/2022]
Abstract
Due to the high prevalence of multi-drug resistant bacteria, combination therapy is an efficient choice for treatment of infections caused by highly resistant strains. In this study, the efficacy of ceftiofur plus danofloxacin combination was investigated against resistant Escherichia coli. The interaction between the two drugs was determined by checkerboard tests and time-kill assays. The combination was defined as bactericidal or bacteriostatic based on the minimum bactericidal concentration test results. Mutant prevention concentration test was used to evaluate the resistance tendency suppression potential of the combination. The combination had a synergistic effect against 83.00% of the isolates as verified by the checkerboard and time-kill assays. The combination was defined as bactericidal against all E. coli strains, since minimum bactericidal concentration: minimum inhibitory concentration ratios were below four thresholds and also markedly reduced mutant prevention concentration values of ceftiofur up to 4000-fold compared to its single use. Ceftiofur plus danofloxacin combination inhibited growth of E. coli strains which were resistant to ceftiofur or newer generation of fluoroquinolones. Our results suggest that ceftiofur plus danofloxacin combination has a bactericidal characteristic and can be an important alternative for the treatment of infections caused by resistant E. coli.
Collapse
Affiliation(s)
- Murat Cengiz
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Uludag University, Nilufer, Turkiye; ,Correspondence Murat Cengiz. PhD, Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Uludag University, Nilufer, Turkiye. E-mail:
| | - Pinar Sahinturk
- Institute of Health Science, Uludag University, Nilufer, Turkiye;
| | | | - Halis Akalin
- Department of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, Uludag University, Nilufer, Turkiye.
| | - Songul Sonal
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Uludag University, Nilufer, Turkiye;
| |
Collapse
|
12
|
Wentzel JM, Biggs LJ, Van Vuuren M. Comparing the minimum inhibitory and mutant prevention concentrations of selected antibiotics against animal isolates of Pasteurella multocida and Salmonella typhimurium. Onderstepoort J Vet Res 2022; 89:e1-e7. [PMID: 35144445 PMCID: PMC8831991 DOI: 10.4102/ojvr.v89i1.1955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 10/18/2021] [Indexed: 11/18/2022] Open
Abstract
Historically, the use of antibiotics was not well regulated in veterinary medicine. The emergence of antibiotic resistance (ABR) in pathogenic bacteria in human and veterinary medicine has driven the need for greater antibiotic stewardship. The preservation of certain antibiotic classes for use exclusively in humans, especially in cases of multidrug resistance, has highlighted the need for veterinarians to reduce its use and redefine dosage regimens of antibiotics to ensure efficacy and guard against the development of ABR pathogens. The minimum inhibitory concentration (MIC), the lowest concentration of an antibiotic drug that will prevent the growth of a bacterium, is recognised as a method to assist in antibiotic dosage determination. Minimum inhibitory concentrations sometimes fail to deal with first-step mutants in bacterial populations; therefore dosing regimens based solely on MIC can lead to the development of ABR. The mutant prevention concentration (MPC) is the minimum inhibitory antibiotic concentration of the most resistant first-step mutant. Mutant prevention concentration determination as a complementary and sometimes preferable alternative to MIC determination for veterinarians when managing bacterial pathogens. The results of this study focused on livestock pathogens and antibiotics used to treat them, which had a MIC value of 0.25 µg/mL for enrofloxacin against all 27 isolates of Salmonella typhimurium. The MPC values were 0.50 µg/mL, with the exception of five isolates that had MPC values of 4.00 µg/mL. The MPC test yielded 65.52% (18 isolates) Salmonella isolates with florfenicol MICs in the sensitive range, while 11 isolates were in the resistant range. Seventeen isolates (58.62%) of Pasteurella multocida had MIC values in the susceptible range and 41.38% (12 isolates) had an intermediate MIC value. Mutant prevention concentration determinations as done in this study is effective for the antibiotic treatment of bacterial infections and minimising the development of resistance. The MPC method can be used to better control to prevent the development of antibiotic drug resistance used in animals.
Collapse
Affiliation(s)
- Jeanette M Wentzel
- Hans Hoheisen Research Station, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa; and, Department of Veterinary Tropical Disease, Faculty of Veterinary Science, University of Pretoria, Pretoria.
| | | | | |
Collapse
|
13
|
Nemonoxacin enhances antibacterial activity and anti-resistant mutation ability of vancomycin against methicillin-resistant Staphylococcus aureus in an in vitro dynamic pharmacokinetic/pharmacodynamic model. Antimicrob Agents Chemother 2021; 66:e0180021. [PMID: 34902266 DOI: 10.1128/aac.01800-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Reduced susceptibility and emergence of resistance to vancomycin in methicillin-resistant Staphylococcus aureus (MRSA) have led to the development of various vancomycin based combinations. Nemonoxacin is a novel nonfluorinated quinolone with antibacterial activity against MRSA. The present study aimed to investigate the effects of nemonoxacin on antibacterial activity and the anti-resistant mutation ability of vancomycin for MRSA and explore whether quinolone resistance genes are associated with a reduction in the vancomycin minimal inhibitory concentration (MIC) and mutant prevention concentration (MPC) when combined with nemonoxacin. Four isolates, all with a vancomycin MIC of 2 μg/mL, were used in a modified in vitro dynamic pharmacokinetic/pharmacodynamic model to investigate the effects of nemonoxacin on antibacterial activity (M04, M23 and M24) and anti-resistant mutation ability (M04, M23 and M25, all with MPC ≥19.2 μg/mL) of vancomycin. The mutation sites of gyrA, gyrB, parC, and parE of 55 clinical MRSA isolates were sequenced. We observed that in M04 and M23, the combination of vancomycin (1g q12h) and nemonoxacin (0.5g qd) showed a synergistic bactericidal activity and resistance enrichment suppression. All clinical isolates resistant to nemonoxacin harbored gyrA (S84→L) mutation; gyrA (S84→L) and parC (E84→K) mutations were the two independent risk factors for the unchanged vancomycin MPC in combination. Nemonoxacin enhances the bactericidal activity and suppresses resistance enrichment ability of vancomycin against MRSA with a MIC of 2 μg/mL. Our in vitro data support the combination of nemonoxacin and vancomycin for the treatment of MRSA infection with a high MIC.
Collapse
|
14
|
Yang F, Chen P, Wang H, Xing X, Wang S, Ishaq HM, Liao W. Comparative Minimal Inhibitory and Mutant Prevention Concentration of Eight Antimicrobial Agents Against Klebsiella pneumoniae. Microb Drug Resist 2021; 28:229-235. [PMID: 34851749 DOI: 10.1089/mdr.2021.0228] [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: 01/08/2023] Open
Abstract
Purpose: With the emergence of multidrug-resistant and pan-resistant strains, Klebsiella pneumoniae (K. pneumoniae) shows higher treatment failure rates and mortality in clinics. It is more important to develop an effective method for treating K. pneumonia infections. The main objectives of this study were to determine the minimal inhibitory concentration (MIC) and the mutant prevention concentration (MPC) for eight antimicrobial agents against K. pneumoniae isolated from different hosts and compare the emergence of resistant mutants between animal strains and human strains. Materials and Methods: A total of 72 nonduplicate K. pneumoniae isolates and 8 antimicrobial agents (amikacin, azithromycin, levofloxacin, doxycycline, nitrofurantoin, colistin, tigecycline, and imipenem) were used. The MIC and MPC values were determined using agar plate assays. The values of the selection index (SI) were calculated with MPC90/MIC90. Pharmacodynamic parameters were calculated using published plasma pharmacokinetic variables. Results: For human isolate strains, the MPC50/90 (μg/mL) values were as follows: amikacin, 32/128; azithromycin, 64/128; levofloxacin, 4/16; doxycycline, 32/32; nitrofurantoin, 128/512; colistin, 4/8; tigecycline, 8/16; and imipenem, 4/8. The value of SI was 8 for azithromycin, doxycycline, and tigecycline; 16 for amikacin, levofloxacin, and nitrofurantoin; 4 for imipenem; and 2 for colistin. For animal isolate strains, the MPC90 values were 128 μg/mL for azithromycin and doxycycline, 64 μg/mL for amikacin, 32 μg/mL for levofloxacin, 512 μg/mL for nitrofurantoin, 8 μg/mL for colistin and tigecycline, 4 μg/mL for imipenem. The value of SI was 2 for colistin and imipenem, 8 for tigecycline, 16 for amikacin, and 32 for the other four agents. In combination with pharmacokinetic parameters, these findings indicated that the plasma concentrations of the seven antibiotics except imipenem were below the MPC for the entire dosing interval. Conclusion: The ability of eight antibiotics to prevent resistant mutants of K. pneumoniae was different between animal strains and human strains. Higher doses than those currently approved should be required to prevent the enrichment of mutants of drug-resistant bacteria in the clinics.
Collapse
Affiliation(s)
- Fan Yang
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China.,Xinxiang Key Laboratory of Pathogenic Biology, Xinxiang Medical University, Xinxiang, China
| | - Ping Chen
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China
| | - Huiyuan Wang
- Department of Pathogenic Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xiaoyu Xing
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China
| | - Sisi Wang
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China
| | - Hafiz Muhammad Ishaq
- Faculty of Veterinary and Animal Sciences, Muhammad Nawaz Shareef University of Agriculture, Multan, Pakistan
| | - Wei Liao
- Department of Clinical Laboratory, The Affiliated People's Hospital of Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
15
|
Blondeau JM, Fitch SD. In Vitro Killing of Canine Urinary Tract Infection Pathogens by Ampicillin, Cephalexin, Marbofloxacin, Pradofloxacin, and Trimethoprim/Sulfamethoxazole. Microorganisms 2021; 9:2279. [PMID: 34835405 PMCID: PMC8619264 DOI: 10.3390/microorganisms9112279] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 12/24/2022] Open
Abstract
Urinary tract infections are common in dogs, necessitating antimicrobial therapy. We determined the speed and extent of in vitro killing of canine urinary tract infection pathogens by five antimicrobial agents (ampicillin, cephalexin, marbofloxacin, pradofloxacin, and trimethoprim/sulfamethoxazole) following the first 3 h of drug exposure. Minimum inhibitory and mutant prevention drug concentrations were determined for each strain. In vitro killing was determined by exposing bacteria to clinically relevant drug concentrations and recording the log10 reduction and percent kill in viable cells at timed intervals. Marbofloxacin and pradofloxacin killed more bacterial cells, and faster than other agents, depending on the time of sampling and drug concentration. Significant differences were seen between drugs for killing Escherichia coli, Proteus mirabilis, Enterococcus faecalis, and Staphylococcus pseudintermedius strains. At the maximum urine drug concentrations, significantly more E. coli cells were killed by marbofloxacin than by ampicillin (p < 0.0001), cephalexin (p < 0.0001), and TMP/SMX (p < 0.0001) and by pradofloxacin than by cephalexin (p < 0.0001) and TMP/SMX (p < 0.0001), following 5 min of drug exposure. Rapid killing of bacteria should inform thinking on drug selection for short course therapy for uncomplicated UTIs, without compromising patient care, and is consistent with appropriate antimicrobial use and stewardship principles.
Collapse
Affiliation(s)
- Joseph M. Blondeau
- Departments of Microbiology and Immunology, Pathology and Laboratory Medicine and Ophthalmology, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada
- Department of Clinical Microbiology, Royal University Hospital and Saskatchewan Health Authority, Saskatoon, SK S7N 0W8, Canada;
| | - Shantelle D. Fitch
- Department of Clinical Microbiology, Royal University Hospital and Saskatchewan Health Authority, Saskatoon, SK S7N 0W8, Canada;
| |
Collapse
|
16
|
Seabra G, Ventura Mendes RF, Dos Santos Amorim LFV, Peregrino IV, Branquinha MH, Dos Santos ALS, Nunes APF. Azithromycin Use in COVID-19 Patients: Implications on the Antimicrobial Resistance. Curr Top Med Chem 2021; 21:677-683. [PMID: 34028347 DOI: 10.2174/156802662108210319145317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Gabriela Seabra
- Laboratorio de Resistencia Bacteriana (RESBAC), Departamento de Microbiologia, Centro de Ciencias da Saude (CCS), Universidade Federal do Espirito Santo (UFES), Vitoria, Brazil
| | - Roberta Ferreira Ventura Mendes
- Laboratorio de Resistencia Bacteriana (RESBAC), Departamento de Microbiologia, Centro de Ciencias da Saude (CCS), Universidade Federal do Espirito Santo (UFES), Vitoria, Brazil
| | - Luiz Felipe Vieira Dos Santos Amorim
- Laboratorio de Resistencia Bacteriana (RESBAC), Departamento de Microbiologia, Centro de Ciencias da Saude (CCS), Universidade Federal do Espirito Santo (UFES), Vitoria, Brazil
| | - Ingrid Vianez Peregrino
- Laboratorio de Resistencia Bacteriana (RESBAC), Departamento de Microbiologia, Centro de Ciencias da Saude (CCS), Universidade Federal do Espirito Santo (UFES), Vitoria, Brazil
| | - Marta Helena Branquinha
- Laboratorio de Estudos Avancados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Goes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - André Luis Souza Dos Santos
- Laboratorio de Estudos Avancados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Goes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Ana Paula Ferreira Nunes
- Laboratorio de Resistencia Bacteriana (RESBAC), Departamento de Microbiologia, Centro de Ciencias da Saude (CCS), Universidade Federal do Espirito Santo (UFES), Vitoria, Brazil
| |
Collapse
|
17
|
The Role of PK/PD Analysis in the Development and Evaluation of Antimicrobials. Pharmaceutics 2021; 13:pharmaceutics13060833. [PMID: 34205113 PMCID: PMC8230268 DOI: 10.3390/pharmaceutics13060833] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 05/27/2021] [Accepted: 05/31/2021] [Indexed: 12/13/2022] Open
Abstract
Pharmacokinetic/pharmacodynamic (PK/PD) analysis has proved to be very useful to establish rational dosage regimens of antimicrobial agents in human and veterinary medicine. Actually, PK/PD studies are included in the European Medicines Agency (EMA) guidelines for the evaluation of medicinal products. The PK/PD approach implies the use of in vitro, ex vivo, and in vivo models, as well as mathematical models to describe the relationship between the kinetics and the dynamic to determine the optimal dosing regimens of antimicrobials, but also to establish susceptibility breakpoints, and prevention of resistance. The final goal is to optimize therapy in order to maximize efficacy and minimize side effects and emergence of resistance. In this review, we revise the PK/PD principles and the models to investigate the relationship between the PK and the PD of antibiotics. Additionally, we highlight the outstanding role of the PK/PD analysis at different levels, from the development and evaluation of new antibiotics to the optimization of the dosage regimens of currently available drugs, both for human and animal use.
Collapse
|
18
|
Evaluation of Fluoroquinolone Resistance in Clinical Avian Pathogenic Escherichia coli Isolates from Flanders (Belgium). Antibiotics (Basel) 2020; 9:antibiotics9110800. [PMID: 33198103 PMCID: PMC7696922 DOI: 10.3390/antibiotics9110800] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/06/2020] [Accepted: 11/11/2020] [Indexed: 11/16/2022] Open
Abstract
Fluoroquinolones are frequently used antimicrobials for the treatment of avian pathogenic Escherichia coli (APEC) infections. However, rapid development and selection of resistance to this class of antimicrobial drugs is a significant problem. The aim of this study was to investigate the occurrence and mechanisms of antimicrobial resistance against enrofloxacin (ENRO) in APEC strains in Flanders, Belgium. One hundred and twenty-five APEC strains from broilers with clinical colibacillosis were collected in Flanders from November 2017 to June 2018. The minimum inhibitory concentration (MIC) of all strains and the mutant prevention concentration (MPC) of a sample of sensitive isolates were determined using a commercial gradient strip test and via the agar dilution method, respectively. Non-wild type (NWT) isolates were further characterized using polymerase chain reaction (PCR), gel electrophoresis and gene sequencing. Forty percent of the APEC strains were NWT according to the epidemiological cut-off (ECOFF) measure (MIC > 0.125 μg/mL). With respect to clinical breakpoints, 21% were clinically intermediate (0.5 ≤ MIC ≤ 1 μg/mL) and 10% were clinically resistant (MIC ≥ 2). The MPC values of the tested strains ranged from 0.064 to 1 μg/mL, resulting in MPC/MIC ratios varying from 4 to 32. The majority (92%) of the NWT strains carried one or two mutations in gyrA. Less than a quarter (22%) manifested amino acid substitutions in the topoisomerase IV parC subunit. Only three of the NWT strains carried a mutation in parE. Plasmid mediated quinolone resistance (PMQR) associated genes were detected in 18% of the NWT strains. In contrast to the relatively large number of NWT strains, only a small percentage of APEC isolates was considered clinically resistant. The most common MPC value for sensitive strains was 0.125 μg/mL. Some isolates showed higher values, producing wide mutant selection windows (MSW). Chromosomal mutations in DNA gyrase and topoisomerase IV were confirmed as the main source of decreased antimicrobial fluoroquinolone susceptibility, de-emphasizing the role of PMQR mechanisms.
Collapse
|
19
|
Charoo NA, Abdallah DB, Parveen T, Abrahamsson B, Cristofoletti R, Groot DW, Langguth P, Parr A, Polli JE, Mehta M, Shah VP, Tajiri T, Dressman J. Biowaiver Monograph for Immediate-Release Solid Oral Dosage Forms: Moxifloxacin Hydrochloride. J Pharm Sci 2020; 109:2654-2675. [DOI: 10.1016/j.xphs.2020.06.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/05/2020] [Accepted: 06/03/2020] [Indexed: 01/31/2023]
|
20
|
Coetzee JF, Cernicchiaro N, Sidhu PK, Kleinhenz MD. Association between antimicrobial drug class selection for treatment and retreatment of bovine respiratory disease and health, performance, and carcass quality outcomes in feedlot cattle. J Anim Sci 2020; 98:5816850. [PMID: 32255182 DOI: 10.1093/jas/skaa109] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 04/04/2020] [Indexed: 11/14/2022] Open
Abstract
Treatment and control of bovine respiratory disease (BRD) is predicated on the use of two categories of antimicrobials, namely bacteriostatic drugs that inhibit bacterial growth and replication (STATIC), and bactericidal drugs that kill bacteria in in vitro culture systems (CIDAL). Recently, we reported that initial BRD treatment with a STATIC antimicrobial followed by retreatment with a CIDAL antimicrobial was associated with a higher frequency of multidrug-resistant bacteria isolated from field cases of BRD submitted to a veterinary diagnostic laboratory. The present study was conducted to test the hypothesis that calves administered the same class of antimicrobial for first and second BRD treatment (i.e., CIDAL-CIDAL or STATIC-STATIC) would have improved health and performance outcomes at the feedlot compared to calves that received a different antimicrobial class for retreatment (i.e., STATIC-CIDAL or CIDAL-STATIC). The association between antimicrobial treatments and health, performance, and carcass quality outcomes were determined by a retrospective analysis of 4,252 BRD treatment records from a commercial feedlot operation collected from 2001 to 2005. Data were compared using generalized linear mixed statistical models that included gender, season, and arrival weight as covariates. The mean (±SE) probability of BRD cases identified as requiring four or more treatments compared to three treatments was greater in calves that received STATIC-CIDAL (73.58 ± 2.38%) or STATIC-STATIC (71.32 ± 2.52%) first and second antimicrobial treatments compared to calves receiving CIDAL-CIDAL (50.35 ± 3.46%) first and second treatments (P < 0.001). Calves receiving CIDAL-CIDAL first and second treatments also had an increased average daily gain (1.11 ± 0.03 kg/d) compared to calves receiving STATIC-CIDAL (0.95 ± 0.03 kg/d) and STATIC-STATIC (0.84 ± 0.02 kg/d) treatments (P < 0.001). Furthermore, CIDAL-CIDAL-treated calves had a higher probability of a choice quality grade at slaughter (36.44 ± 4.80%) compared to STATIC-CIDAL calves (28.09 ± 3.88%) (P = 0.037). There was no effect of antimicrobial treatment combination on BRD mortality (P = 0.855) or yield grade (P = 0.240) outcomes. These observations suggest that consideration should be given to antimicrobial pharmacodynamics when selecting drugs for retreatment of BRD. These findings have implications for developing BRD treatment protocols that address both post-treatment production and antimicrobial stewardship concerns.
Collapse
Affiliation(s)
- Johann F Coetzee
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS
| | - Natalia Cernicchiaro
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, College of Veterinary Medicine, Kansas State University, Manhattan, KS
| | - Pritam K Sidhu
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS
| | - Michael D Kleinhenz
- Department of Clinical Sciences, College of Veterinary Medicine, College of Veterinary Medicine, Kansas State University, Manhattan, KS
| |
Collapse
|
21
|
Ahirrao P, Tambat R, Chandal N, Mahey N, Kamboj A, Jain UK, Singh IP, Jachak SM, Nandanwar HS. MsrA Efflux Pump Inhibitory Activity of
Piper cubeba
L.f. and its Phytoconstituents against
Staphylococcus aureus
RN4220. Chem Biodivers 2020; 17:e2000144. [DOI: 10.1002/cbdv.202000144] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/16/2020] [Indexed: 02/01/2023]
Affiliation(s)
- Pallavi Ahirrao
- Department of Pharmaceutical ChemistryChandigarh College of Pharmacy Landran-Mohali 140307 Punjab India
- Research scholar of IK Gujral Punjab Technical UniversityKapurthalaDepartment of Pharmaceutical ChemistryChandigarh College of Pharmacy CGC Landran, Mohali 140307 Punjab India
| | - Rushikesh Tambat
- Clinical Microbiology and Bioactive Screening LaboratoryCSIR-Institute of Microbial Technology, Sector 39-A Chandigarh 160036 India
| | - Nishtha Chandal
- Clinical Microbiology and Bioactive Screening LaboratoryCSIR-Institute of Microbial Technology, Sector 39-A Chandigarh 160036 India
| | - Nisha Mahey
- Clinical Microbiology and Bioactive Screening LaboratoryCSIR-Institute of Microbial Technology, Sector 39-A Chandigarh 160036 India
| | - Anjoo Kamboj
- Department of Pharmaceutical ChemistryChandigarh College of Pharmacy Landran-Mohali 140307 Punjab India
| | - Upendra K. Jain
- Department of Pharmaceutical ChemistryChandigarh College of Pharmacy Landran-Mohali 140307 Punjab India
| | - Inder Pal Singh
- Department of Natural ProductsNational Institute of Pharmaceutical Education and Research (NIPER), Sector-67 SAS Nagar, Mohali 160062 Punjab India
| | - Sanjay M. Jachak
- Department of Natural ProductsNational Institute of Pharmaceutical Education and Research (NIPER), Sector-67 SAS Nagar, Mohali 160062 Punjab India
| | - Hemraj S. Nandanwar
- Clinical Microbiology and Bioactive Screening LaboratoryCSIR-Institute of Microbial Technology, Sector 39-A Chandigarh 160036 India
| |
Collapse
|
22
|
Mataracı-Kara E, Yılmaz M, Özbek-Çelik B. In Vitro Synergistic Effect and Mutant Prevention Concentrations of Cefepime Alone or in Combination with Sulbactam Against OXA-48-positive Klebsiella pneumoniae Isolates. Curr Microbiol 2020; 77:2137-2142. [PMID: 32572550 DOI: 10.1007/s00284-020-02094-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 06/15/2020] [Indexed: 10/24/2022]
Abstract
The aim of this study is to investigate the combination of cefepime and sulbactam. Sulbactam, when administered , will effectively inhibit all Extended-spectrum beta lactamases (ESBLs) of the microorganism, while cefepime will inhibit the growth of the resistant microorganisms since it will not be hydrolyzed by OXA-48. Forty OXA-48-producing K. pneumoniae strains were investigated for their Minimum inhibitory concentrations (MICs) for carbapenems, cefepime, and cefepime + sulbactam by broth microdilution method. Also, the mutant prevention concentration (MPC)s of cefepime alone or in combination with sulbactam was determined. Additionally, the bactericidal activities of cefepime and cefepime + sulbactam were evaluated by the time-kill curve (TKC) assay against selected strains. Also, the in vitro synergistic activity of cefepime + sulbactam combination was determined by TKC. Based on MIC results, up to 35/40 and 34/40 of the strains were resistant to carbapenems and cefepime, respectively. Cefepime + sulbactam MIC range was lower than those for cefepime alone against all the studied isolates. Moreover, cefepime + sulbactam combination presented lower MPC values than cefepime alone. The synergistic interactions of cefepime + sulbactam were also achieved against studied strains at 24 h. No antagonism was observed against studied K. pneumoniae strains. The findings of this study displayed that cefepime + sulbactam combination had synergistic or additive effect against OXA-48-producing K. pneumoniae strains. Additionally, it was first observed that this combination could display a lower MPC than cefepime alone. Further investigations may be helpful for understanding the effectiveness of cefepime + sulbactam combinations for OXA-48-positive carbapenem-resistant K. pneumoniae isolates.
Collapse
Affiliation(s)
- Emel Mataracı-Kara
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Istanbul University, 34116, Beyazit-Istanbul, Turkey.
| | - Mesut Yılmaz
- Department of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, Istanbul Medipol University, 34214, Istanbul, Turkey
| | - Berna Özbek-Çelik
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Istanbul University, 34116, Beyazit-Istanbul, Turkey
| |
Collapse
|
23
|
Alsaeed A, Wright G, Deneer H, Rubin JE, Sanche SE, Blondeau JM. Methicillin-resistant Staphylococcus aureus replication in the presence of high (≥32 µg/ml) drug concentration of vancomycin as seen by electron microscopy. J Chemother 2020; 32:179-187. [PMID: 32393118 DOI: 10.1080/1120009x.2020.1761191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) has unfortunately become a common pathogen in many healthcare facilities. In many institutions, vancomycin remains the preferred agent for treating serious MRSA infections including bacteraemia with or without endocarditis. The mutant prevention concentration (MPC) testing ≥109 colony forming units of bacteria, describes the antimicrobial drug concentration blocking the growth of the least susceptible cell from high density bacterial populations. With blood culture isolates of MRSA, we discovered strains with MPC values ≥32 µg/ml and viable cells could be readily recovered from agar plates containing 32 µg/ml of vancomycin. To investigate MRSA strains surviving in high concentrations of vancomycin on drug containing agar plates, we utilized electron microscopy to measure cell wall thickness as this has been previously reported as a potential mechanism of resistance1 along with septum thickening. Our data shows MRSA replication from high density bacterial populations in the presence of ≥32 µg/ml of vancomycin. Such observations may explain vancomycin failure in some patients and/or persistent bacteraemia and could potentially question the use of this drug in some critically ill patients in favour of an alternative agent.
Collapse
Affiliation(s)
- Amal Alsaeed
- University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Glenda Wright
- Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Harry Deneer
- Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.,Division of Clinical Microbiology, Royal University Hospital and Saskatchewan Health Authority, Saskatoon, Saskatchewan, Canada
| | - Joseph E Rubin
- Veterinary Microbiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Stephen E Sanche
- Division of Clinical Microbiology, Royal University Hospital and Saskatchewan Health Authority, Saskatoon, Saskatchewan, Canada.,Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Joseph M Blondeau
- Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.,Division of Clinical Microbiology, Royal University Hospital and Saskatchewan Health Authority, Saskatoon, Saskatchewan, Canada.,Departments of Microbiology and Immunology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.,Ophthalmology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
24
|
Lozano‐Huntelman NA, Singh N, Valencia A, Mira P, Sakayan M, Boucher I, Tang S, Brennan K, Gianvecchio C, Fitz‐Gibbon S, Yeh P. Evolution of antibiotic cross-resistance and collateral sensitivity in Staphylococcus epidermidis using the mutant prevention concentration and the mutant selection window. Evol Appl 2020; 13:808-823. [PMID: 32211069 PMCID: PMC7086048 DOI: 10.1111/eva.12903] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/14/2019] [Indexed: 01/09/2023] Open
Abstract
In bacteria, evolution of resistance to one antibiotic is frequently associated with increased resistance (cross-resistance) or increased susceptibility (collateral sensitivity) to other antibiotics. Cross-resistance and collateral sensitivity are typically evaluated at the minimum inhibitory concentration (MIC). However, these susceptibility changes are not well characterized with respect to the mutant prevention concentration (MPC), the antibiotic concentration that prevents a single-step mutation from occurring. We measured the MIC and the MPC for Staphylococcus epidermidis and 14 single-drug resistant strains against seven antibiotics. We found that the MIC and the MPC were positively correlated but that this correlation weakened if cross-resistance did not evolve. If any type of resistance did evolve, the range of concentrations between the MIC and the MPC tended to shift right and widen. Similar patterns of cross-resistance and collateral sensitivity were observed at the MIC and MPC levels, though more symmetry was observed at the MIC level. Whole-genome sequencing revealed mutations in both known-target and nontarget genes. Moving forward, examining both the MIC and the MPC may lead to better predictions of evolutionary trajectories in antibiotic-resistant bacteria.
Collapse
Affiliation(s)
| | - Nina Singh
- Department of Ecology and Evolutionary BiologyUniversity of CaliforniaLos AngelesCAUSA
| | - Alondra Valencia
- Department of Ecology and Evolutionary BiologyUniversity of CaliforniaLos AngelesCAUSA
| | - Portia Mira
- Department of Ecology and Evolutionary BiologyUniversity of CaliforniaLos AngelesCAUSA
| | - Maral Sakayan
- Department of Ecology and Evolutionary BiologyUniversity of CaliforniaLos AngelesCAUSA
| | - Ian Boucher
- Department of Ecology and Evolutionary BiologyUniversity of CaliforniaLos AngelesCAUSA
| | - Sharon Tang
- Department of Ecology and Evolutionary BiologyUniversity of CaliforniaLos AngelesCAUSA
| | - Kelley Brennan
- Department of Ecology and Evolutionary BiologyUniversity of CaliforniaLos AngelesCAUSA
| | - Crystal Gianvecchio
- Department of Ecology and Evolutionary BiologyUniversity of CaliforniaLos AngelesCAUSA
| | - Sorel Fitz‐Gibbon
- Department of Molecular, Cell, Developmental BiologyUniversity of CaliforniaLos AngelesCAUSA
| | - Pamela Yeh
- Department of Ecology and Evolutionary BiologyUniversity of CaliforniaLos AngelesCAUSA
- Santa Fe InstituteSanta FeNMUSA
| |
Collapse
|
25
|
Susceptibility of Cutibacterium acnes to topical minocycline foam. Anaerobe 2020; 62:102169. [DOI: 10.1016/j.anaerobe.2020.102169] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/13/2020] [Accepted: 01/27/2020] [Indexed: 11/19/2022]
|
26
|
Boby N, Abbas MA, Lee EB, Park SC. Pharmacodynamics of Ceftiofur Selected by Genomic and Proteomic Approaches of Streptococcus parauberis Isolated from the Flounder, Paralichthys olivaceus. Int J Genomics 2020; 2020:4850290. [PMID: 32318593 PMCID: PMC7150728 DOI: 10.1155/2020/4850290] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/14/2020] [Accepted: 03/13/2020] [Indexed: 11/17/2022] Open
Abstract
We employed an integrative strategy to present subtractive and comparative metabolic and genomic-based findings of therapeutic targets against Streptococcus parauberis. For the first time, we not only identified potential targets based on genomic and proteomic database analyses but also recommend a new antimicrobial drug for the treatment of olive flounder (Paralichthys olivaceus) infected with S. parauberis. To do that, 102 total annotated metabolic pathways of this bacterial strain were extracted from computational comparative metabolic and genomic databases. Six druggable proteins were identified from these metabolic pathways from the DrugBank database with their respective genes as mtnN, penA, pbp2, murB, murA, coaA, and fni out of 112 essential nonhomologous proteins. Among these hits, 26 transmembrane proteins and 77 cytoplasmic proteins were extracted as potential vaccines and drug targets, respectively. From the FDA DrugBank, ceftiofur was selected to prevent antibiotic resistance as it inhibited our selected identified target. Florfenicol is used for treatment of S. parauberis infection in flounder and was chosen as a comparator drug. All tested strains of fish isolates with S. parauberis were susceptible to ceftiofur and florfenicol with minimum inhibitory concentrations (MIC) of 0.0039-1 μg/mL and 0.5-8 μg/mL, IC50 of 0.001-0.5 μg/mL and 0.7-2.7 μg/mL, and minimum biofilm eradication concentrations (MBEC) of 2-256 μg/mL and 4-64 μg/mL, respectively. Similar susceptibility profiles for ceftiofur and florfenicol were found, with ceftiofur observed as an effective and potent antimicrobial drug against both planktonic and biofilm-forming strains of the fish pathogen Streptococcus parauberis, and it can be applied in the aquaculture industry. Thus, our predictive approach not only showed novel therapeutic agents but also indicated that marketed drugs should also be tested for efficacy against newly identified targets of this important fish pathogen.
Collapse
Affiliation(s)
- Naila Boby
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu 41569, Republic of Korea
| | - Muhammad Aleem Abbas
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu 41569, Republic of Korea
| | - Eon-Bee Lee
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu 41569, Republic of Korea
| | - Seung-Chun Park
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu 41569, Republic of Korea
| |
Collapse
|
27
|
Blondeau JM, Fitch SD. In vitro killing of canine strains of Staphylococcus pseudintermedius and Escherichia coli by cefazolin, cefovecin, doxycycline and pradofloxacin over a range of bacterial densities. Vet Dermatol 2020; 31:187-e39. [PMID: 31825131 PMCID: PMC7317566 DOI: 10.1111/vde.12835] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/20/2019] [Indexed: 01/21/2023]
Abstract
Background Bacterial densities likely fluctuate during infection and may exceed the bacterial density used in susceptibility testing. As such, investigation of bacterial killing by antibiotics over a range of varying bacterial densities may provide important differences between compounds and could impact drug selection for therapy. Hypothesis/Objectives To measure killing of clinical isolates of Staphylococcus pseudintermedius and Escherichia coli by cefazolin, cefovecin, doxycycline and pradofloxacin at clinically relevant (minimum inhibitory, mutant prevention, maximum serum and maximum tissue) drug concentrations against varying densities of bacteria. Animals/Materials Bacterial strains collected from dogs with urinary tract infections were studied. Methods and materials High bacterial densities ranging from 106 to 109 colony forming units (cfu)/mL were exposed to minimum inhibitory, mutant prevention, blood and tissue drug concentrations, and the percentages (log10) of viable cells killed following 30 min, 1, 2, 4, 6, 12 and 24 h of drug exposure were quantified. Results Doxycycline exhibited bacteriostatic properties with less killing than the other three agents. For example, at a 107 cfu/mL density of S. pseudintermedius, more cells were killed by pradofloxacin (P < 0.0001) and cefovecin (P = 0.0014) but not cefazolin when compared to doxycycline at the maximum serum drug concentration following 12 h of drug exposure. Conclusions and clinical importance Differences were seen between some drugs in the speed and extent of bacterial killing; this could be clinically important and may impact drug selection and length of therapy. Background – Bacterial densities likely fluctuate during infection and may exceed the bacterial density used in susceptibility testing. As such, investigation of bacterial killing by antibiotics over a range of varying bacterial densities may provide important differences between compounds and could impact drug selection for therapy. Hypothesis – To measure killing of clinical isolates of Staphylococcus pseudintermedius and Escherichia coli by cefazolin, cefovecin, doxycycline and pradofloxacin at clinically relevant (minimum inhibitory, mutant prevention, maximum serum and maximum tissue) drug concentrations against varying densities of bacteria. Conclusions and clinical importance – Differences were seen between some drugs in the speed and extent of bacterial killing; this could be clinically important and may impact drug selection and length of therapy.
Collapse
Affiliation(s)
- Joseph M Blondeau
- Division of Clinical Microbiology, Royal University Hospital and Saskatchewan Health Authority, 103 Hospital Drive, Saskatoon, Saskatchewan, Canada, S7N 0W8.,Departments of Microbiology and Immunology, Pathology and Ophthalmology, University of Saskatchewan, 103 Hospital Drive, Saskatoon, Saskatchewan, Canada, S7N 0W8
| | - Shantelle D Fitch
- Division of Clinical Microbiology, Royal University Hospital and Saskatchewan Health Authority, 103 Hospital Drive, Saskatoon, Saskatchewan, Canada, S7N 0W8
| |
Collapse
|
28
|
Gostev V, Sopova J, Kalinogorskaya O, Tsvetkova I, Lobzin Y, Klotchenko S, Sidorenko S. In Vitro Ceftaroline Resistance Selection of Methicillin-Resistant Staphylococcus aureus Involves Different Genetic Pathways. Microb Drug Resist 2019; 25:1401-1409. [PMID: 31329022 DOI: 10.1089/mdr.2019.0130] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The pathways in the development of ceftaroline resistance of methicillin-resistant Staphylococcus aureus (MRSA) isolates belonging to the ST8, ST239, and ST228 were evaluated. Ceftaroline-resistant derivatives were isolated through selection during 40 passages. Ceftaroline MIC measurements and whole-genome sequencing were performed after 5, 20, and 40 passages. In two ST8 derivative isolates, ceftaroline MIC increased up to 128 mg/L. Mutations were acquired in gdpP and graS in one isolate after 20 passages and in gdpP in another after 40 passages. MIC for two ST239 derivatives increased to 128 mg/L. Substitutions in Pbp4 and polymorphisms in the upstream region of pbp4 were identified in both derivatives after 40 passages. In one isolate, additional mutation in gdpP and deletion in graR were detected. In an ST228 derivative, MIC increased to 32 mg/L with one mutation in penicillin-binding protein 2a (Y446N) detected after five passages and a second (E447K) after 20 passages. Three pathways in the development of ceftaroline resistance were identified. For ST8 and ST239 derivatives mutations were detected in gdpP and pbp4, respectively, whereas in ST228 - in mecA. Most derivatives harbored additional mutations whose potential role in the development of resistance has not been determined.
Collapse
Affiliation(s)
- Vladimir Gostev
- Department of Medical Microbiology and Molecular Epidemiology, Pediatric Research and Clinical Center for Infectious Diseases, Saint Petersburg, Russia
- Department of Medical Microbiology, North-Western State Medical University named after I.I. Mechnikov, Saint Petersburg, Russia
| | - Julia Sopova
- Laboratory of Genetic Models of Human Diseases, Saint Petersburg Branch of Vavilov Institute of General Genetics, Saint Petersburg, Russia
- Department of Genetics and Biotechnology, Saint Petersburg State University, Saint Petersburg, Russia
| | - Olga Kalinogorskaya
- Department of Medical Microbiology and Molecular Epidemiology, Pediatric Research and Clinical Center for Infectious Diseases, Saint Petersburg, Russia
| | - Irina Tsvetkova
- Department of Medical Microbiology and Molecular Epidemiology, Pediatric Research and Clinical Center for Infectious Diseases, Saint Petersburg, Russia
| | - Yuri Lobzin
- Department of Medical Microbiology and Molecular Epidemiology, Pediatric Research and Clinical Center for Infectious Diseases, Saint Petersburg, Russia
- Department of Medical Microbiology, North-Western State Medical University named after I.I. Mechnikov, Saint Petersburg, Russia
| | - Sergey Klotchenko
- Division of Viral Molecular Biology, Smorodintsev Research Institute of Influenza, Saint Petersburg, Russia
| | - Sergey Sidorenko
- Department of Medical Microbiology and Molecular Epidemiology, Pediatric Research and Clinical Center for Infectious Diseases, Saint Petersburg, Russia
- Department of Medical Microbiology, North-Western State Medical University named after I.I. Mechnikov, Saint Petersburg, Russia
| |
Collapse
|
29
|
Mutant Prevention Concentration of Ciprofloxacin against Klebsiella pneumoniae Clinical Isolates: An Ideal Prognosticator in Treating Multidrug-Resistant Strains. Int J Microbiol 2019; 2019:6850108. [PMID: 31772582 PMCID: PMC6855012 DOI: 10.1155/2019/6850108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 09/11/2019] [Indexed: 11/17/2022] Open
Abstract
Background Fluoroquinolone-resistant Klebsiella pneumoniae poses a therapeutic challenge when implicated in urinary tract infections, pyelonephritis, pneumonia, skin infections, osteomyelitis, and respiratory infections. The mutant prevention concentration (MPC) represents a concentration threshold above which increase of resistant mutants occurs rarely. The aim of the present study is to determine the MPC among ciprofloxacin-resistant K. pneumoniae clinical isolates. Materials and Methods A total of 240 clinical isolates of K. pneumoniae were collected from a tertiary care hospital. The MPCs were determined for 24 selected strains using an inoculum of 1010 CFU/ml in Müller-Hinton agar plates with serial/various concentrations (0.003-100 μg) of ciprofloxacin. In addition to the MPC, phenotypic screening for ESBL, AmpC, and carbapenemase was performed. The detection of qnr genes for 24 isolates and DNA sequencing for six isolates were performed. Results Ciprofloxacin resistance was observed in 19.6% of the K. pneumoniae clinical isolates. Among the ciprofloxacin-resistant isolates, 14 isolates showed an MPC value of more than 100 μg. The MPC ranged between 100 μg and 20 μg for ciprofloxacin-resistant isolates. ESBL producers and qnr gene-producing strains had a high MPC. 11 isolates showed the presence of either qnrB or qnrS genes. None of the samples showed the presence of the qnrA gene. Conclusion From our study, we infer that ESBL producers and qnr gene-possessing strains are frequently resistant to ciprofloxacin. Estimation of the MPC in the case of multidrug-resistant isolates in the clinical setup may help in treating these drug-resistant strains.
Collapse
|
30
|
Implications for dosing regimen of enrofloxacin administered concurrently with dexamethasone in febrile buffalo calves. Trop Anim Health Prod 2019; 52:1093-1102. [PMID: 31701397 DOI: 10.1007/s11250-019-02103-w] [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: 02/25/2019] [Accepted: 09/26/2019] [Indexed: 10/25/2022]
Abstract
The objective of the study was to determine the influence of dexamethasone (DXM) on pharmacokinetics (PK) and pharmacodynamics (PD) of enrofloxacin (ENR) for dosage optimization following concurrent administration of ENR and DXM in febrile buffalo calves. A 2 μg/kg intravenous dosage of lipopolysaccharide derived from Escherichia coli was used to induce fever in calves. After inducing fever, ENR was administered at the dose rate of 12 mg/kg, IM followed by IM injection of DXM (0.05 mg/kg) in calves. Minor alterations in PK of ENR were observed following the administration of ENR + DXM. The PK parameters were t1/2K10 = 6.34 h, Cl/F = 0.729 L/kg/h, and MRT0-∞ = 10.5 h. Antibacterial activity (MIC, MBC, ex vivo time-kill kinetics) of ENR for P. multocida was not affected by DXM. But MPC of ENR against P. multocida was lessened in presence of DXM. Using PK-PD-modeled AUC0-24h/MIC values for bactericidal effect against P. multocida, daily dosages of ENR administered in combination with DXM were 4.02 mg/kg and 16.1 mg/kg, respectively, for MIC90s of 0.125 μg/ml and 0.50 μg/ml. A dose of 5.38 mg/kg was determined for ENR for frequently occurring P. multocida infections having ≤ MIC90 of 0.125 μg/ml and PK-PD modeled dose was comparable with the recommended ENR dose of 5 mg/kg for bovines for mild infections. It is suggested that a recommended dosage of 5-12.5 mg/kg of ENR can be used effectively in combination with DXM to treat P. multocida associated infections in buffalo calves without any risk of resistance amplification.
Collapse
|
31
|
Golikova MV, Strukova EN, Portnoy YA, Dovzhenko SA, Kobrin MB, Zinner SH, Firsov AA. A novel parameter to predict the effects of antibiotic combinations on the development of Staphylococcus aureus resistance: in vitro model studies at subtherapeutic daptomycin and rifampicin exposures. J Chemother 2019; 31:1-9. [PMID: 31314704 DOI: 10.1080/1120009x.2019.1640924] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/01/2019] [Indexed: 10/26/2022]
Abstract
The search for optimal predictors of anti-mutant effects remains a pressing problem in studies of antibiotic-associated bacterial resistance. To relate the emergence of bacterial resistance with the antibiotic mutant prevention concentration (MPC), a novel integral parameter - the area around the resistance threshold, i.e. MPC level (AAMPC) is proposed. The AAMPC is the algebraic sum of the area under the antibiotic concentration-time curve that is above the MPC (positive area) and the area above the concentration-time curve that is under the MPC (negative area). To assess the predictive performance of AAMPC, the enrichment of resistant Staphylococcus aureus was studied by simulating treatment with daptomycin and rifampicin alone and in combination in an in vitro dynamic model. The enhanced anti-mutant effects of the antibiotic combinations were due to lowering the negative 24-h AAMPCs. These findings suggest that a novel MPC-related parameter is a reliable predictor of mutant enrichment.
Collapse
Affiliation(s)
- Maria V Golikova
- a Department of Pharmacokinetics & Pharmacodynamics , Gause Institute of New Antibiotics , Moscow , Russia
| | - Elena N Strukova
- a Department of Pharmacokinetics & Pharmacodynamics , Gause Institute of New Antibiotics , Moscow , Russia
| | - Yury A Portnoy
- a Department of Pharmacokinetics & Pharmacodynamics , Gause Institute of New Antibiotics , Moscow , Russia
| | - Svetlana A Dovzhenko
- a Department of Pharmacokinetics & Pharmacodynamics , Gause Institute of New Antibiotics , Moscow , Russia
| | - Mikhail B Kobrin
- a Department of Pharmacokinetics & Pharmacodynamics , Gause Institute of New Antibiotics , Moscow , Russia
| | - Stephen H Zinner
- b Department of Medicine , Mount Auburn Hospital, Harvard Medical School , Cambridge , MA , USA
| | - Alexander A Firsov
- a Department of Pharmacokinetics & Pharmacodynamics , Gause Institute of New Antibiotics , Moscow , Russia
| |
Collapse
|
32
|
Levin-Reisman I, Brauner A, Ronin I, Balaban NQ. Epistasis between antibiotic tolerance, persistence, and resistance mutations. Proc Natl Acad Sci U S A 2019; 116:14734-14739. [PMID: 31262806 PMCID: PMC6642377 DOI: 10.1073/pnas.1906169116] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Understanding the evolution of microorganisms under antibiotic treatments is a burning issue. Typically, several resistance mutations can accumulate under antibiotic treatment, and the way in which resistance mutations interact, i.e., epistasis, has been extensively studied. We recently showed that the evolution of antibiotic resistance in Escherichia coli is facilitated by the early appearance of tolerance mutations. In contrast to resistance, which reduces the effectiveness of the drug concentration, tolerance increases resilience to antibiotic treatment duration in a nonspecific way, for example when bacteria transiently arrest their growth. Both result in increased survival under antibiotics, but the interaction between resistance and tolerance mutations has not been studied. Here, we extend our analysis to include the evolution of a different type of tolerance and a different antibiotic class and measure experimentally the epistasis between tolerance and resistance mutations. We derive the expected model for the effect of tolerance and resistance mutations on the dynamics of survival under antibiotic treatment. We find that the interaction between resistance and tolerance mutations is synergistic in strains evolved under intermittent antibiotic treatment. We extend our analysis to mutations that result in antibiotic persistence, i.e., to tolerance that is conferred only on a subpopulation of cells. We show that even when this population heterogeneity is included in our analysis, a synergistic interaction between antibiotic persistence and resistance mutations remains. We expect our general framework for the epistasis in killing conditions to be relevant for other systems as well, such as bacteria exposed to phages or cancer cells under treatment.
Collapse
Affiliation(s)
- Irit Levin-Reisman
- Racah Institute of Physics, The Hebrew University of Jerusalem, 91904 Jerusalem, Israel
- The Harvey M. Kruger Family Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, 91904 Jerusalem, Israel
| | - Asher Brauner
- Racah Institute of Physics, The Hebrew University of Jerusalem, 91904 Jerusalem, Israel
- The Harvey M. Kruger Family Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, 91904 Jerusalem, Israel
| | - Irine Ronin
- Racah Institute of Physics, The Hebrew University of Jerusalem, 91904 Jerusalem, Israel
- The Harvey M. Kruger Family Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, 91904 Jerusalem, Israel
| | - Nathalie Q Balaban
- Racah Institute of Physics, The Hebrew University of Jerusalem, 91904 Jerusalem, Israel;
- The Harvey M. Kruger Family Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, 91904 Jerusalem, Israel
| |
Collapse
|
33
|
Alieva KN, Golikova MV, Portnoy YA, Dovzhenko SA, Kobrin MB, Zinner SH, Firsov AA. Concentration-dependent enrichment of resistant Enterococcus faecium exposed to linezolid in an in vitro dynamic model. J Chemother 2019; 30:364-370. [PMID: 30663551 DOI: 10.1080/1120009x.2018.1533267] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
To explore the relationship between pharmacokinetic variables and enterococcal resistance to linezolid, a vancomycin-resistant strain whose mutant prevention concentration (MPC) exceeded the MIC by two fold was selected among six clinical isolates of Enterococcus faecium. The selected strain was exposed to simulated pharmacokinetics of twice-daily linezolid for five days. Mutants resistant to 2 × MIC of the antibiotic were enriched at ratios of the 24-h area under the concentration-time curve (AUC24) to the MIC of 15 and 30 h but not at 60 and 120 h. These observations could be explained by the different times when antibiotic concentrations exceed the MPC (T>MPC): 0 to 14, 63 and 100% of the dosing interval. Using the area under the bacterial mutant concentration-time curve (AUBCM) determined in this study and in previous work with other E. faecium strains (MPC/MIC 4), a strain-independent T>MPC relationship with mutant enrichment was established.
Collapse
Affiliation(s)
- Kamilla N Alieva
- a Department of Pharmacokinetics and Pharmacodynamics , Gause Institute of New Antibiotics , Moscow , Russia
| | - Maria V Golikova
- a Department of Pharmacokinetics and Pharmacodynamics , Gause Institute of New Antibiotics , Moscow , Russia
| | - Yury A Portnoy
- a Department of Pharmacokinetics and Pharmacodynamics , Gause Institute of New Antibiotics , Moscow , Russia
| | - Svetlana A Dovzhenko
- a Department of Pharmacokinetics and Pharmacodynamics , Gause Institute of New Antibiotics , Moscow , Russia
| | - Mikhail B Kobrin
- a Department of Pharmacokinetics and Pharmacodynamics , Gause Institute of New Antibiotics , Moscow , Russia
| | - Stephen H Zinner
- b Department of Medicine, Mount Auburn Hospital, Harvard Medical School , Cambridge , Massachusetts , USA
| | - Alexander A Firsov
- a Department of Pharmacokinetics and Pharmacodynamics , Gause Institute of New Antibiotics , Moscow , Russia
| |
Collapse
|
34
|
Gianvecchio C, Lozano NA, Henderson C, Kalhori P, Bullivant A, Valencia A, Su L, Bello G, Wong M, Cook E, Fuller L, Neal JB, Yeh PJ. Variation in Mutant Prevention Concentrations. Front Microbiol 2019; 10:42. [PMID: 30766517 PMCID: PMC6365975 DOI: 10.3389/fmicb.2019.00042] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/11/2019] [Indexed: 12/12/2022] Open
Abstract
Objectives:Understanding how phenotypic traits vary has been a longstanding goal of evolutionary biologists. When examining antibiotic-resistance in bacteria, it is generally understood that the minimum inhibitory concentration (MIC) has minimal variation specific to each bacterial strain-antibiotic combination. However, there is a less studied resistance trait, the mutant prevention concentration (MPC), which measures the MIC of the most resistant sub-population. Whether and how MPC varies has been poorly understood. Here, we ask a simple, yet important question: How much does the MPC vary, within a single strain-antibiotic association? Using a Staphylococcus species and five antibiotics from five different antibiotic classes—ciprofloxacin, doxycycline, gentamicin, nitrofurantoin, and oxacillin—we examined the frequency of resistance for a wide range of concentrations per antibiotic, and measured the repeatability of the MPC, the lowest amount of antibiotic that would ensure no surviving cells in a 1010 population of bacteria. Results: We found a wide variation within the MPC and distributions that were rarely normal. When antibiotic resistance evolved, the distribution of the MPC changed, with all distributions becoming wider and some multi-modal. Conclusion: Unlike the MIC, there is high variability in the MPC for a given bacterial strain-antibiotic combination.
Collapse
Affiliation(s)
- Crystal Gianvecchio
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Natalie Ann Lozano
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Claire Henderson
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Pooneh Kalhori
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Austin Bullivant
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Alondra Valencia
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Lauren Su
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Gladys Bello
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Michele Wong
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Emoni Cook
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Lakhia Fuller
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Jerome B Neal
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Pamela J Yeh
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, Los Angeles, CA, United States.,Santa Fe Institute, Santa Fe, NM, United States
| |
Collapse
|
35
|
Shinohara DR, Menegucci TC, Fedrigo NH, Migliorini LB, Carrara-Marroni FE, Maria Dos Anjos M, Cardoso CL, Nishiyama SAB, Tognim MCB. Synergistic activity of polymyxin B combined with vancomycin against carbapenem-resistant and polymyxin-resistant Acinetobacter baumannii: first in vitro study. J Med Microbiol 2019; 68:309-315. [PMID: 30663954 DOI: 10.1099/jmm.0.000920] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
PURPOSE The effect of a combination of polymyxin B (PMB) and vancomycin (VAN) was assessed against six Acinetobacter baumannii clinical isolates belonging to six different clusters (three PMB-susceptible and three PMB-resistant). METHODOLOGY The synergistic effect of the PMB-VAN combination was determined with the checkerboard, time-kill, disk-diffusion and M.I.C.Evaluator assays. PMB-resistance was investigated with mcr-1 gene amplification and a mutant frequency assay. RESULTS In the checkerboard assay, all PMB-resistant isolates showed a synergistic effect. The time-kill assay demonstrated that the PMB-VAN combination had a bactericidal effect at 24 h against isolates with a high mutant rate for PMB, suggesting that this combination may block the hypermutation of some isolates. No antagonism was detected. All PMB-resistant isolates also showed synergism in the disk-diffusion test, and a significant decrease in VAN MICs in the M.I.C.Evaluator assay. CONCLUSION Our findings indicate that the PMB-VAN combination has a synergistic effect on A. baumannii, especially against PMB-resistant isolates.
Collapse
Affiliation(s)
| | | | | | | | | | - Márcia Maria Dos Anjos
- 1 Universidade Estadual de Maringá, Avenida Colombo 5790, CEP 87.020-900 Maringá, Paraná, Brazil
| | - Celso Luiz Cardoso
- 1 Universidade Estadual de Maringá, Avenida Colombo 5790, CEP 87.020-900 Maringá, Paraná, Brazil
| | | | | |
Collapse
|
36
|
Blondeau JM, Fitch SD. Mutant prevention and minimum inhibitory concentration drug values for enrofloxacin, ceftiofur, florfenicol, tilmicosin and tulathromycin tested against swine pathogens Actinobacillus pleuropneumoniae, Pasteurella multocida and Streptococcus suis. PLoS One 2019; 14:e0210154. [PMID: 30629633 PMCID: PMC6328246 DOI: 10.1371/journal.pone.0210154] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 12/18/2018] [Indexed: 11/18/2022] Open
Abstract
Actinobacillus pleuropneumoniae, Pasteurella multocida and Streptococcus suis are prevalent bacterial causes of swine infections. Morbidity, mortality and positively impacting the financial burden of infection occurs with appropriate antimicrobial therapy. Increasing antimicrobial resistance complicates drug therapy and resistance prevention is now a necessity to optimize therapy and prolong drug life. Mutant bacterial cells are said to arise spontaneously in bacterial densities of 107-109 or greater colony forming units/ml. Antibiotic drug concentration inhibiting growth of the least susceptible cell in these high density populations has been termed the mutant prevention concentration (MPC). In this study MPC and minimum inhibitory concentration (MIC) values of ceftiofur, enrofloxacin, florfenicol, tilmicosin and tulathromycin were determined against the swine pathogens A. pleuropneumoniae, P.multocida and S. suis. The following MIC90/MPC90 values (mg/L) for 67 A. pleuropneumoniae and 73 P. multocida strains respectively were as follows: A. pleuropneumoniae 0.031/0.5, ≤0.016/0.5, 0.5/2, 4/32, 2/32; P. multocida 0.004/0.25, 0.016/0.125, 0.5/0.5, 8/16, 0.5/1. For 33 S. suis strains, MIC90 values (mg/L) respectively were as follows: 1, 0.25, 4, ≥8 and ≥8. A total of 16 S. suis strains with MIC values of 0.063-0.5 mg/L to ceftiofur and 0.25-0.5 mg/L to enrofloxacin were tested by MPC; MPC values respectively were 0.5 and 1 mg/L respectively. MPC concentrations provide a dosing target which may serve to reduce amplification of bacterial subpopulations with reduced antimicrobial susceptibility. Drug potency based on MIC90 values was ceftiofur > enrofloxacin >florfenicol = tulathromycin > tilmicosin; based on MPC90 values was enrofloxacin > ceftiofur > tulathromycin > florfenicol ≥ tilmicosin.
Collapse
Affiliation(s)
- Joseph M. Blondeau
- Department of Clinical Microbiology, Royal University Hospital and the Saskatchewan Health Authority, Saskatoon, Saskatchewan, Canada
- Departments of Microbiology and Immunology, Pathology and Ophthalmology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- * E-mail:
| | - Shantelle D. Fitch
- Department of Clinical Microbiology, Royal University Hospital and the Saskatchewan Health Authority, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
37
|
Xiao X, Lan W, Wang Y, Jiang L, Jiang Y, Wang Z. Comparative pharmacokinetics of danofloxacin in healthy and Pasteurella multocida infected ducks. J Vet Pharmacol Ther 2018; 41:912-918. [PMID: 30182430 DOI: 10.1111/jvp.12712] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/24/2018] [Accepted: 07/12/2018] [Indexed: 11/30/2022]
Abstract
Pasteurella multocida (P. multocida) infection causes substantial economic loss in the duck industry. Danofloxacin, a fluoroquinolone solely used in animals, shows good antibacterial activity against P. multocida. In this study, the in vitro pharmacodynamics of danofloxacin against P. multocida was studied. The serum and lung tissue pharmacokinetics of danofloxacin were studied in healthy and P. multocida infected ducks following oral administration of a single dose of 5 mg/kg body weight (b.w.). The MIC, MBC and MPC of danofloxacin against P. multocida (C48-1 ) were 0.25, 1 and 3.2 μg/ml, respectively. The Cmax was 0.34 μg/ml, attained at 2.03 hr in healthy ducks, and was 0.35 μg/ml, attained at 2.87 hr in diseased ducks. Compared to the serum pharmacokinetics of danofloxacin in healthy ducks, the absorption rate and extent were similar in healthy and diseased animals. In contrast, the elimination rate was slower, with an elimination half-life (T1/2β ) of 13.17 and 16.18 hr for healthy and infected animals, respectively; the AUCs in the two groups were 5.70 and 7.68 μg hr/ml, respectively, which means the total amount of drug in the circulation was increased in the infected ducks. The maximum concentration in lung tissues between healthy and infected animals was not significantly different (8.96 vs. 8.93 μg/g). However, the Tmax in healthy ducks was longer than that in infected ducks (4 hr vs. 1.75 hr), which means that the distribution rate of danofloxacin was slower in healthy ducks. The concentration of danofloxacin in lung tissues was approximately 24-fold higher than that in the serum. In the serum pharmacokinetic profiles, the ƒAUC0-24 hr /MIC was 18.19 in healthy ducks and was 25.04 in P. multocida infected ducks at the clinical recommended dose, which is far from the PK/PD target (125 hr) of fluoroquinolones. Danofloxacin, at a dose of 5 mg/kg b.w., seems to be insufficient for ducks infected with P. multocida, with an MIC equal to 0.25 μg/ml.
Collapse
Affiliation(s)
- Xia Xiao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China
- Institutes of Agricultural Science and Technology Development, Yangzhou, Jiangsu, China
| | - Weixuan Lan
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Ying Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Lijie Jiang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Yongjia Jiang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Zhiqiang Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China
- Institutes of Agricultural Science and Technology Development, Yangzhou, Jiangsu, China
| |
Collapse
|
38
|
Xiao X, Pei L, Jiang LJ, Lan WX, Xiao JY, Jiang YJ, Wang ZQ. In Vivo Pharmacokinetic/Pharmacodynamic Profiles of Danofloxacin in Rabbits Infected With Salmonella typhimurium After Oral Administration. Front Pharmacol 2018; 9:391. [PMID: 29719510 PMCID: PMC5913287 DOI: 10.3389/fphar.2018.00391] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 04/04/2018] [Indexed: 12/17/2022] Open
Abstract
Salmonella typhimurium is a highly transmissible pathogen in rabbits that causes significant losses. Danofloxacin shows excellent efficacy against S. typhimurium infections. However, there are few reports of the pharmacokinetic/pharmacodynamic (PK/PD) modeling of danofloxacin against this pathogen. The aim of this study was to evaluate the in vivo PK/PD relationship of danofloxacin in rabbits infected with S. typhimurium. We used the reduction of bacterial burden in the blood, liver, spleen, and lung as the target PD endpoints, and determined the PK/PD indexes that best correlated with the efficacy and its corresponding magnitude. Danofloxacin was administrated orally to experimentally S. typhimurium-infected rabbits once daily for three successive days. The concentrations of danofloxacin in the serum and the bacterial burden in the blood, liver, spleen, and lung were determined. The PK/PD relationships of danofloxacin against S. typhimurium were evaluated using a Sigmoid Emax model. The results showed that the area under the concentration-time curve from 0 to 24 h/minimum inhibitory concentration (AUC24 h/MIC) ratio correlated well with the in vivo antibacterial effectiveness in different organs, with an r2 of 0.8971, 0.9186, 0.9581, and 0.8708 in the blood, liver, spleen, and lung, respectively. The AUC24 h/MIC ratios for the bactericidal effect (3 × Log10 colony forming units/mL reductions) were 121.30, 354.28, 216.64, and 228.66 in the blood, liver, spleen, and lung, respectively, indicating that the in vivo effectiveness of danofloxacin against S. typhimurium using bacterial reduction in different organs as PD endpoints was not identical. This study illustrated that the selection of the target organ for bacterial reduction determination had little effect on best PK/PD parameter determination, but is critical for parameter magnitude calculation in antimicrobial PK/PD modeling, and furthermore, has an impact on the rational dosage optimization process.
Collapse
Affiliation(s)
- Xia Xiao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Institute of Agricultural Science and Technology Development, Yangzhou, China
| | - Lin Pei
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Institute of Agricultural Science and Technology Development, Yangzhou, China
| | - Li-Jie Jiang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Institute of Agricultural Science and Technology Development, Yangzhou, China
| | - Wei-Xuan Lan
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Institute of Agricultural Science and Technology Development, Yangzhou, China
| | - Jia-Yu Xiao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Institute of Agricultural Science and Technology Development, Yangzhou, China
| | - Yon-Jia Jiang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Institute of Agricultural Science and Technology Development, Yangzhou, China
| | - Zhi-Qiang Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Institute of Agricultural Science and Technology Development, Yangzhou, China
| |
Collapse
|
39
|
Bordallo-Cardona MÁ, Marcos-Zambrano LJ, Sánchez-Carrillo C, de la Pedrosa EGG, Cantón R, Bouza E, Escribano P, Guinea J. Mutant Prevention Concentration and Mutant Selection Window of Micafungin and Anidulafungin in Clinical Candida glabrata Isolates. Antimicrob Agents Chemother 2018; 62:e01982-17. [PMID: 29311063 PMCID: PMC5826129 DOI: 10.1128/aac.01982-17] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Accepted: 12/29/2017] [Indexed: 11/20/2022] Open
Abstract
We report the mutant prevention concentration (MPC) and mutant selection window (MSW) for micafungin and anidulafungin administered to treat Candida glabrata We also determine the mutation frequency. We studied 20 echinocandin-susceptible, fluconazole-intermediate, and FKS wild-type C. glabrata isolates. Adjusted inocula were stroked directly onto Sabouraud agar plates containing different concentrations of micafungin or anidulafungin and visually inspected daily for up to 5 days of incubation. Individual colonies growing on the plates containing echinocandins at 1 mg/liter were selected for antifungal susceptibility testing. The FKS genes of the resulting individual phenotypically resistant colonies were sequenced, and the MPC, MSW, and mutation frequency were determined. Biofilm was quantified, and the growth kinetics and virulence (Galleria mellonella model) of the resulting individual FKS mutant colonies were studied. For micafungin and anidulafungin, we found similar results for the MPC (0.06 to 2 mg/liter and 0.25 to 2 mg/liter, respectively), MSW (0.015 to 2 mg/liter for both echinocandins), and mutation frequency (3.7 × 10-8 and 2.8 × 10-8, respectively). A total of 12 isolates were able to grow at 1 mg/liter on echinocandin-containing plates, yielding a total of 32 phenotypically resistant colonies; however, FKS2 mutations (ΔF658, S663P, W715L, and E655A) were observed only in 21 colonies. We did not find differences in biofilm formation, the kinetic parameters studied, or the median survival of larvae infected by wild-type isolates and the resulting individual FKS2 mutant colonies. Echinocandin concentrations lower than 2 mg/liter can lead to selection of resistance mutations in C. glabrata isolates in vitro.
Collapse
Affiliation(s)
- María Ángeles Bordallo-Cardona
- Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Laura Judith Marcos-Zambrano
- Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Carlos Sánchez-Carrillo
- Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Elia Gómez G de la Pedrosa
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Biomédica, Madrid, Spain
- Red Española de Investigación en Patología Infecciosa (REIPI), Madrid, Spain
| | - Rafael Cantón
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Biomédica, Madrid, Spain
- Red Española de Investigación en Patología Infecciosa (REIPI), Madrid, Spain
| | - Emilio Bouza
- Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- CIBER Enfermedades Respiratorias-CIBERES (CB06/06/0058), Madrid, Spain
- Medicine Department, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Pilar Escribano
- Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Jesús Guinea
- Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- CIBER Enfermedades Respiratorias-CIBERES (CB06/06/0058), Madrid, Spain
- Medicine Department, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
40
|
Sidhu PK, Waraich GS, Kaur G, Daundkar PS, Sharma SK, Gehring R. Difference in the PK of ceftiofur in the presence and absence of nimesulide and implications for dose determination through PK/PD integration. Small Rumin Res 2018. [DOI: 10.1016/j.smallrumres.2017.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
41
|
Lei Z, Liu Q, Yang S, Yang B, Khaliq H, Li K, Ahmed S, Sajid A, Zhang B, Chen P, Qiu Y, Cao J, He Q. PK-PD Integration Modeling and Cutoff Value of Florfenicol against Streptococcus suis in Pigs. Front Pharmacol 2018; 9:2. [PMID: 29387013 PMCID: PMC5776115 DOI: 10.3389/fphar.2018.00002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 01/03/2018] [Indexed: 01/22/2023] Open
Abstract
The aims of the present study were to establish optimal doses and provide an alternate COPD for florfenicol against Streptococcus suis based on pharmacokinetic-pharmacodynamic integration modeling. The recommended dose (30 mg/kg b.w.) were administered in healthy pigs through intramuscular and intravenous routes for pharmacokinetic studies. The main pharmacokinetic parameters of Cmax, AUC0-24h, AUC, Ke, t1/2ke, MRT, Tmax, and Clb, were estimated as 4.44 μg/ml, 88.85 μg⋅h/ml, 158.56 μg⋅h/ml, 0.048 h-1, 14.46 h, 26.11 h, 4 h and 0.185 L/h⋅kg, respectively. The bioavailability of florfenicol was calculated to be 99.14% after I.M administration. A total of 124 Streptococcus suis from most cities of China were isolated to determine the minimum inhibitory concentration (MIC) of florfenicol. The MIC50 and MIC90 were calculated as 1 and 2 μg/ml. A serotype 2 Streptococcus suis (WH-2), with MIC value similar to MIC90, was selected as a representative for an in vitro and ex vivo pharmacodynamics study. The MIC values of WH-2 in TSB and plasma were 2 μg/ml, and the MBC/MIC ratios were 2 in TSB and plasma. The MPC was detected to be 3.2 μg/ml. According to inhibitory sigmoid Emax model, plasma AUC0-24h/MIC values of florfenicol versus Streptococcus suis were 37.89, 44.02, and 46.42 h for the bactericidal, bacteriostatic, and elimination activity, respectively. Monte Carlo simulations the optimal doses for bactericidal, bacteriostatic, and elimination effects were calculated as 16.5, 19.17, and 20.14 mg/kg b.w. for 50% target attainment rates (TAR), and 21.55, 25.02, and 26.85 mg/kg b.w. for 90% TAR, respectively. The PK-PD cutoff value (COPD) analyzed from MCS for florfenicol against Streptococcus suis was 1 μg/ml which could provide a sensitivity cutoff value. These results contributed an optimized alternative to clinical veterinary medicine and showed that the dose of 25.02 mg/kg florfenicol for 24 h could have a bactericidal action against Streptococcus suis after I.M administration. However, it should be validated in clinical practice in the future investigations.
Collapse
Affiliation(s)
- Zhixin Lei
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- National Reference Laboratory of Veterinary Drug Residues and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, China
- Department of Veterinary Pharmacology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Qianying Liu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- National Reference Laboratory of Veterinary Drug Residues and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, China
| | - Shuaike Yang
- National Reference Laboratory of Veterinary Drug Residues and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, China
- Department of Veterinary Pharmacology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Bing Yang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- National Reference Laboratory of Veterinary Drug Residues and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, China
| | - Haseeb Khaliq
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Kun Li
- Department of Veterinary Pharmacology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, University of lllinois at Urbana – Champaign, Champaign, IL, United States
| | - Saeed Ahmed
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- National Reference Laboratory of Veterinary Drug Residues and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, China
| | - Abdul Sajid
- National Reference Laboratory of Veterinary Drug Residues and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Sciences and Animal Husbandry, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Bingzhou Zhang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Pin Chen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- National Reference Laboratory of Veterinary Drug Residues and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, China
| | - Yinsheng Qiu
- School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Jiyue Cao
- National Reference Laboratory of Veterinary Drug Residues and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, China
- Department of Veterinary Pharmacology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Qigai He
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
42
|
Lei Z, Liu Q, Yang B, Khaliq H, Cao J, He Q. PK-PD Analysis of Marbofloxacin against Streptococcus suis in Pigs. Front Pharmacol 2017; 8:856. [PMID: 29209222 PMCID: PMC5701813 DOI: 10.3389/fphar.2017.00856] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 11/08/2017] [Indexed: 12/18/2022] Open
Abstract
Marbofloxacin is a fluoroquinolone antibiotic and highly effective treatment for respiratory diseases. Here we aimed to evaluate the ex vivo activity of marbofloxacin against Streptococcus suis in pig serum, as well as the optimal dosages scheme for avoiding the fluoroquinolone resistance development. A single dose of 8 mg/kg body weight (bw) was administrated orally to healthy pigs and serum samples were collected during the next 72 h. Serum marbofloxacin content was determined by high-performance liquid chromatography. We estimated the Cmax (6.28 μg/ml), AUC0-24 h (60.30 μg.h/ml), AUC0-∞ (88.94 μg.h/ml), T1/2ke, (12.48 h), Tmax (0.75 h) and Clb (0.104 L/h) of marbofloxacin in pigs, as well as the bioavailability of marbofloxacin (94.21%) after a single 8 mg/kg oral administration. We also determined the pharmacodynamic of marbofloxacin against 134 Streptococcus suis strains isolated from Chinese cities in TSB and serum. These isolated strains had a MIC90 of 1 μg/ml. HB2, a virulent, serotype 2 isolate of SS, was selected for having antibacterial activity in TSB and serum to marbofloxacin. We determined the minimum inhibitory concentration (MIC, 1 μg/ml in TSB, 2 μg/ml in serum), minimum bactericidal concentration (MBC, 4 μg/ml in TSB, 4 μg/ml in serum), and mutant prevention concentration (2.56 μg/ml in TSB) for marbofloxacin against Streptococcus suis (HB2). In serum, by inhibitory sigmoid Emax modeling, the AUC0-24h/MIC values for marbofloxacin against HB2 were 25.23 (bacteriostatic), 35.64 (bactericidal), and 39.71 (elimination) h. Based on Monte Carlo simulations, the predicted optimal oral doses of marbofloxacin curing Streptococcus suis were 5.88 (bacteriostatic), 8.34 (bactericidal), and 9.36 (elimination) mg/kg.bw for a 50% target attainment ratio, and 8.16 (bacteriostatic), 11.31 (bactericidal), and 12.35 (elimination) mg/kg.bw for a 90% target attainment ratio. The data presented here provides optimized dosage information for clinical use; however, these predicted dosages should also be validated in clinical practice.
Collapse
Affiliation(s)
- Zhixin Lei
- Department of Veterinary Pharmacology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,National Reference Laboratory of Veterinary Drug Residues and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, China
| | - Qianying Liu
- Department of Veterinary Pharmacology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,National Reference Laboratory of Veterinary Drug Residues and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, China
| | - Bing Yang
- Department of Veterinary Pharmacology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,National Reference Laboratory of Veterinary Drug Residues and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, China
| | - Haseeb Khaliq
- Department of Veterinary Pharmacology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jiyue Cao
- Department of Veterinary Pharmacology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,National Reference Laboratory of Veterinary Drug Residues and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, China
| | - Qigai He
- State Key Laboratory of Agriculture Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
43
|
Lei Z, Liu Q, Xiong J, Yang B, Yang S, Zhu Q, Li K, Zhang S, Cao J, He Q. Pharmacokinetic and Pharmacodynamic Evaluation of Marbofloxacin and PK/PD Modeling against Escherichia coli in Pigs. Front Pharmacol 2017; 8:542. [PMID: 28871226 PMCID: PMC5566571 DOI: 10.3389/fphar.2017.00542] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 08/03/2017] [Indexed: 12/15/2022] Open
Abstract
The aim of this study was to evaluate the activity of marbofloxacin and establish the optimal dose regimens for decreasing the development of fluoroquinolone resistance in pigs against Escherichia coli with ex vivo pharmacokinetic/pharmacodynamic (PK/PD) modeling. The recommended dose (2 mg/kg body weight) of marbofloxacin was orally administered in healthy pigs. The ileum content and plasma were both collected for the determination of marbofloxacin. The main parameters of Cmax, AUC0-24 h, AUC, Ke, t1/2ke, MRT and Clb were 11.28 μg/g, 46.15, 77.81 μg⋅h/g, 0.001 h-1, 69.97 h, 52.45 h, 0.026 kg/h in ileum content, and 0.55 μg/ml, 8.15, 14.67 μg⋅h/ml, 0.023 h-1, 30.67 h, 34.83 h, 0.14 L/h in plasma, respectively In total, 218 E. coli strains were isolated from most cities of China. The antibacterial activity in vitro and ex vivo of marbofloxacin against E. coli was determined following CLSI guidance. The MIC90 of sensitive strains (142) was calculated as 2 μg/ml. The minimum inhibitory concentration (MIC) of HB197 was 2 and 4 μg/ml in broth and ileum fluids, respectively. In vitro mutant prevention concentration, growth and killing-time in vitro and ex vivo of marbofloxacin against selected HB197 were assayed for pharmacodynamic studies. According to the inhibitory sigmoid Emax modeling, the value of AUC0-24 h/MIC produced in ileum content was achieved, and bacteriostatic, bactericidal activity, and elimination were calculated as 16.26, 23.54, and 27.18 h, respectively. Based on Monte Carlo simulations to obtain 90% target attainment rate, the optimal doses to achieve bacteriostatic, bactericidal, and elimination effects were 0.85, 1.22, and 1.41 mg/kg.bw for 50% target, respectively, and 0.92, 1.33, and 1.53 mg/kg.bw for 90% target, respectively, after oral administration. The results in this study provided a more optimized alternative for clinical use and demonstrated that the dosage 2 mg/kg of marbofloxacin by oral administration could have an effect on bactericidal activity against E. coli.
Collapse
Affiliation(s)
- Zhixin Lei
- Department of Veterinary Pharmacology, College of Veterinary Medicine, Huazhong Agricultural UniversityWuhan, China.,National Reference Laboratory of Veterinary Drug Residues and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agriculture UniversityWuhan, China
| | - Qianying Liu
- Department of Veterinary Pharmacology, College of Veterinary Medicine, Huazhong Agricultural UniversityWuhan, China.,National Reference Laboratory of Veterinary Drug Residues and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agriculture UniversityWuhan, China
| | - Jincheng Xiong
- Department of Veterinary Pharmacology, College of Veterinary Medicine, Huazhong Agricultural UniversityWuhan, China
| | - Bing Yang
- Department of Veterinary Pharmacology, College of Veterinary Medicine, Huazhong Agricultural UniversityWuhan, China
| | - Shuaike Yang
- Department of Veterinary Pharmacology, College of Veterinary Medicine, Huazhong Agricultural UniversityWuhan, China
| | - Qianqian Zhu
- National Reference Laboratory of Veterinary Drug Residues and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agriculture UniversityWuhan, China
| | - Kun Li
- Department of Veterinary Pharmacology, College of Veterinary Medicine, Huazhong Agricultural UniversityWuhan, China
| | - Shishuo Zhang
- State Key Laboratory of Agriculture Microbiology, College of Veterinary Medicine, Huazhong Agriculture UniversityWuhan, China
| | - Jiyue Cao
- Department of Veterinary Pharmacology, College of Veterinary Medicine, Huazhong Agricultural UniversityWuhan, China.,National Reference Laboratory of Veterinary Drug Residues and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agriculture UniversityWuhan, China
| | - Qigai He
- State Key Laboratory of Agriculture Microbiology, College of Veterinary Medicine, Huazhong Agriculture UniversityWuhan, China
| |
Collapse
|
44
|
In Vitro Resistance Selection in Shigella flexneri by Azithromycin, Ceftriaxone, Ciprofloxacin, Levofloxacin, and Moxifloxacin. Antimicrob Agents Chemother 2017; 61:AAC.00086-17. [PMID: 28483960 DOI: 10.1128/aac.00086-17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 05/04/2017] [Indexed: 01/02/2023] Open
Abstract
Shigella flexneri continues to be a major cause of diarrhea-associated illness, and increasing resistance to first-line antimicrobials complicates the treatment of infections caused by this pathogen. We investigated the pharmacodynamics of current antimicrobial treatments for shigellosis to determine the likelihood of resistance promotion with continued global antimicrobial use. The mutant prevention concentration (MPC) and mutant selection window (MSW) were determined for azithromycin, ceftriaxone, ciprofloxacin, levofloxacin, and moxifloxacin against a wild-type strain of S. flexneri (ATCC 12022) and an isogenic gyrA mutant (m-12022). Time-kill assays were performed to determine antimicrobial killing. Concentrations of approved doses of ciprofloxacin, levofloxacin, and moxifloxacin are predicted to surpass the MPC for a majority of the dosage interval against ATCC 12022. However, against m-12022, concentrations of all fluoroquinolones are predicted to fall below the MPC and remain in the MSW for a majority of the dosage interval. Concentrations of ceftriaxone fall within the MSW for the majority of the dosage interval for both strains. All agents other than azithromycin displayed bactericidal activity in time-kill assays. Results of pharmacodynamic analyses suggest that all tested fluoroquinolones would achieve a favorable area under the concentration-time curve (AUC)/MPC ratio for ATCC 12022 and would restrict selective enrichment of mutants but that mutant selection in m-12022 would be likely if ciprofloxacin were used. Based on pharmacodynamic analyses, azithromycin and ceftriaxone are predicted to promote mutant selection in both strains. Confirmation of these findings and examination of novel treatment regimens using in vivo studies are warranted.
Collapse
|
45
|
Margaritis A, Galani I, Chatzikonstantinou M, Petrikkos G, Souli M. Plasmid-mediated quinolone resistance determinants among Gram-negative bacteraemia isolates: a hidden threat. J Med Microbiol 2017; 66:266-275. [DOI: 10.1099/jmm.0.000397] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Athanasios Margaritis
- 4th Department of Internal Medicine, Infectious Diseases Laboratory, Molecular Biology Section, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Irene Galani
- Present address: 4th Department of Internal Medicine, National and Kapodistrian University of Athens, School of Mecicine, Athens, Greece
- 4th Department of Internal Medicine, Infectious Diseases Laboratory, Molecular Biology Section, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Marianthi Chatzikonstantinou
- 4th Department of Internal Medicine, Infectious Diseases Laboratory, Molecular Biology Section, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - George Petrikkos
- 4th Department of Internal Medicine, Infectious Diseases Laboratory, Molecular Biology Section, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Maria Souli
- 4th Department of Internal Medicine, Infectious Diseases Laboratory, Molecular Biology Section, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
- Present address: 4th Department of Internal Medicine, National and Kapodistrian University of Athens, School of Mecicine, Athens, Greece
| |
Collapse
|
46
|
Hao H, Li F, Han J, Foley SL, Dai M, Wang X, Wang Y, Huang L, Sun Y, Liu Z, Yuan Z. Cj1199 Affect the Development of Erythromycin Resistance in Campylobacter jejuni through Regulation of Leucine Biosynthesis. Front Microbiol 2017; 8:16. [PMID: 28144238 PMCID: PMC5239772 DOI: 10.3389/fmicb.2017.00016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 01/04/2017] [Indexed: 01/08/2023] Open
Abstract
The aim of this study was to reveal the biological function of Cj1199 which was overexpressed in the laboratory induced erythromycin resistant strains. The Cj1199 deletion mutant (ΦCj1199) was constructed via insertional inactivation from its parent strain Campylobacter jejuni NCTC11168. The ΦCj1199 and NCTC11168 were then subjected to microarray and real-time PCR to find gene pathway of Cj1199. The antimicrobial susceptibility, antimicrobial resistance development, growth characteristics and leucine metabolism were examined to confirm the biological function of Cj1199. Our result showed that a total of 20 genes were down-regulated in ΦCj1199. These genes were mainly involved in leucine biosynthesis, amino acid transport and periplasmic/membrane structure. Compared to NCTC11168, ΦCj1199 was difficult to acquire higher-level erythromycin resistance during the in vitro step-wise selection. The competition growth and leucine-dependent growth assays demonstrated that ΦCj1199 imposed a growth disadvantage under pressure of erythromycin and in the leucine-free medium. In conclusion, Cj1199 gene may directly regulate the leucine biosynthesis and transport and indirectly affect the development of erythromycin resistance in C. jejuni.
Collapse
Affiliation(s)
- Haihong Hao
- National Reference Laboratory of Veterinary Drug Residues and MOA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural UniversityWuhan, China; MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural UniversityWuhan, China; Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Huazhong Agricultural UniversityWuhan, China
| | - Fei Li
- National Reference Laboratory of Veterinary Drug Residues and MOA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural UniversityWuhan, China; MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural UniversityWuhan, China
| | - Jing Han
- Division of Microbiology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson AR, USA
| | - Steven L Foley
- Division of Microbiology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson AR, USA
| | - Menghong Dai
- National Reference Laboratory of Veterinary Drug Residues and MOA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural UniversityWuhan, China; MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural UniversityWuhan, China
| | - Xu Wang
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural UniversityWuhan, China; Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Huazhong Agricultural UniversityWuhan, China
| | - Yulian Wang
- National Reference Laboratory of Veterinary Drug Residues and MOA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural UniversityWuhan, China; Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Huazhong Agricultural UniversityWuhan, China
| | - Lingli Huang
- National Reference Laboratory of Veterinary Drug Residues and MOA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural UniversityWuhan, China; Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Huazhong Agricultural UniversityWuhan, China
| | - Yawei Sun
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural UniversityWuhan, China; Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Huazhong Agricultural UniversityWuhan, China
| | - Zhenli Liu
- National Reference Laboratory of Veterinary Drug Residues and MOA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural UniversityWuhan, China; MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural UniversityWuhan, China; Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Huazhong Agricultural UniversityWuhan, China
| | - Zonghui Yuan
- National Reference Laboratory of Veterinary Drug Residues and MOA Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural UniversityWuhan, China; MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural UniversityWuhan, China; Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Huazhong Agricultural UniversityWuhan, China
| |
Collapse
|
47
|
Waraich GS, Sidhu PK, Daundkar PS, Kaur G, Sharma SK. Pharmacokinetic and pharmacodynamic characterization of ceftiofur crystalline-free acid following subcutaneous administration in domestic goats. J Vet Pharmacol Ther 2016; 40:429-438. [DOI: 10.1111/jvp.12373] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 09/19/2016] [Indexed: 11/30/2022]
Affiliation(s)
- G. S. Waraich
- Department of Veterinary Pharmacology and Toxicology; College of Veterinary Science; Guru Angad Dev Veterinary and Animal Sciences University; Ludhiana India
| | - P. K. Sidhu
- Animal Disease Research Centre; College of Veterinary Science; Guru Angad Dev Veterinary and Animal Sciences University; Ludhiana India
| | - P. S. Daundkar
- Department of Veterinary Pharmacology and Toxicology; College of Veterinary Science; Guru Angad Dev Veterinary and Animal Sciences University; Ludhiana India
| | - G. Kaur
- Department of Veterinary Microbiology; College of Veterinary Science; Guru Angad Dev Veterinary and Animal Sciences University; Ludhiana India
| | - S. K. Sharma
- Department of Veterinary Pharmacology and Toxicology; College of Veterinary Science; Guru Angad Dev Veterinary and Animal Sciences University; Ludhiana India
| |
Collapse
|
48
|
Zhang X, Jiang A, Yu H, Xiong Y, Zhou G, Qin M, Dou J, Wang J. Human Lysozyme Synergistically Enhances Bactericidal Dynamics and Lowers the Resistant Mutant Prevention Concentration for Metronidazole to Helicobacter pylori by Increasing Cell Permeability. Molecules 2016; 21:molecules21111435. [PMID: 27801837 PMCID: PMC6273225 DOI: 10.3390/molecules21111435] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 10/25/2016] [Accepted: 10/25/2016] [Indexed: 02/06/2023] Open
Abstract
Metronidazole (MNZ) is an effective agent that has been employed to eradicate Helicobacter pylori (H. pylori). The emergence of broad MNZ resistance in H. pylori has affected the efficacy of this therapeutic agent. The concentration of MNZ, especially the mutant prevention concentration (MPC), plays an important role in selecting or enriching resistant mutants and regulating therapeutic effects. A strategy to reduce the MPC that can not only effectively treat H. pylori but also prevent resistance mutations is needed. H. pylori is highly resistant to lysozyme. Lysozyme possesses a hydrolytic bacterial cell wall peptidoglycan and a cationic dependent mode. These effects can increase the permeability of bacterial cells and promote antibiotic absorption into bacterial cells. In this study, human lysozyme (hLYS) was used to probe its effects on the integrity of the H. pylori outer and inner membranes using as fluorescent probe hydrophobic 1-N-phenyl-naphthylamine (NPN) and the release of aspartate aminotransferase. Further studies using a propidium iodide staining method assessed whether hLYS could increase cell permeability and promote cell absorption. Finally, we determined the effects of hLYS on the bactericidal dynamics and MPC of MNZ in H. pylori. Our findings indicate that hLYS could dramatically increase cell permeability, reduce the MPC of MNZ for H. pylori, and enhance its bactericidal dynamic activity, demonstrating that hLYS could reduce the probability of MNZ inducing resistance mutations.
Collapse
Affiliation(s)
- Xiaolin Zhang
- The Department of Pharmacy, Food and Drug School, Anhui Science and Technology University, Fengyang 233100, China.
| | - Anmin Jiang
- The School of Life Science, University of Science and Technology of China, Hefei 230032, China.
| | - Hao Yu
- The Department of Pharmacy, Food and Drug School, Anhui Science and Technology University, Fengyang 233100, China.
| | - Youyi Xiong
- The Department of Pharmacy, Food and Drug School, Anhui Science and Technology University, Fengyang 233100, China.
| | - Guoliang Zhou
- The Department of Pharmacy, Food and Drug School, Anhui Science and Technology University, Fengyang 233100, China.
| | - Meisong Qin
- The Department of Pharmacy, Food and Drug School, Anhui Science and Technology University, Fengyang 233100, China.
| | - Jinfeng Dou
- The Department of Pharmacy, Food and Drug School, Anhui Science and Technology University, Fengyang 233100, China.
| | - Jianfei Wang
- The Department of Pharmacy, Food and Drug School, Anhui Science and Technology University, Fengyang 233100, China.
- The Ministry of Agriculture Key Laboratory of Microbial Organic Fertilizer, Bengbu 233030, China.
| |
Collapse
|
49
|
Hariharan P, Paul-Satyaseela M, Gnanamani A. In vitro profiling of antimethicillin-resistant Staphylococcus aureus activity of thymoquinone against selected type and clinical strains. Lett Appl Microbiol 2016; 62:283-9. [PMID: 26743923 DOI: 10.1111/lam.12544] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 01/01/2016] [Accepted: 01/02/2016] [Indexed: 11/24/2022]
Abstract
UNLABELLED This study explores antimethicillin-resistant Staphylococcus aureus (MRSA) activity of a bioactive phytochemical constituent, thymoquinone obtained from the medicinal herb, Nigella sativa Linn. Based on initial assessment on crude extract of seeds of Nigella sativa Linn, the pure active constituent was employed in the study. A total of 99 MRSA strains which comprised of 40 types and 59 clinical strains were selected for the study. Minimum inhibitory concentration (MIC), bactericidal activity, postantibiotic effect (PAE) and propensity to select resistant mutants were determined using standard protocols. Results revealed that thymoquinone exhibited MIC in the range of 8-16 μg ml(-1) and MIC90 of 16 μg ml(-1) against MRSA strains. It was bactericidal to MRSA by demonstrating >3 log kill. It showed a longer PAE of 3·2 ± 0·2 h. Upon exposure to high-density inoculum of MRSA, it did not select resistant mutants. Transmission electron microscopy of thymoquinone-treated MRSA showed no lysis but damage to cell wall and cell membrane which corroborated well with the salt tolerance and bacteriolysis assays. In conclusion, MIC90 , bactericidal property, longer PAE, absence of resistant mutant selection and damages in cell membrane and cell wall imply a promising anti-MRSA activity of thymoquinone. SIGNIFICANCE AND IMPACT OF THE STUDY This is the first detailed report on anti-MRSA activity of thymoquinone. The assessment was made with both type and clinical strains. Thymoquinone may be a potential lead compound which can be further optimized to discover novel anti-MRSA agents.
Collapse
Affiliation(s)
- P Hariharan
- Orchid Chemicals and Pharmaceuticals Ltd., Chennai, India
| | | | - A Gnanamani
- Microbiology Division, CSIR-CLRI, Chennai, India
| |
Collapse
|
50
|
Firsov AA, Golikova MV, Strukova EN, Portnoy YA, Dovzhenko SA, Kobrin MB, Zinner SH. Pharmacokinetically-based prediction of the effects of antibiotic combinations on resistant Staphylococcus aureus mutants: in vitro model studies with linezolid and rifampicin. J Chemother 2016; 29:220-226. [DOI: 10.1080/1120009x.2016.1245174] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Alexander A. Firsov
- Department of Pharmacokinetics & Pharmacodynamics, Gause Institute of New Antibiotics, Moscow 119021, Russia
| | - Maria V. Golikova
- Department of Pharmacokinetics & Pharmacodynamics, Gause Institute of New Antibiotics, Moscow 119021, Russia
| | - Elena N. Strukova
- Department of Pharmacokinetics & Pharmacodynamics, Gause Institute of New Antibiotics, Moscow 119021, Russia
| | - Yury A. Portnoy
- Department of Pharmacokinetics & Pharmacodynamics, Gause Institute of New Antibiotics, Moscow 119021, Russia
| | - Svetlana A. Dovzhenko
- Department of Pharmacokinetics & Pharmacodynamics, Gause Institute of New Antibiotics, Moscow 119021, Russia
| | - Mikhail B. Kobrin
- Department of Pharmacokinetics & Pharmacodynamics, Gause Institute of New Antibiotics, Moscow 119021, Russia
| | - Stephen H. Zinner
- Department of Medicine, Mount Auburn Hospital, Harvard Medical School, Cambridge, MA, USA
| |
Collapse
|