1
|
Ashraf KU, Bunoro-Batista M, Ansell TB, Punetha A, Rosario-Garrido S, Firlar E, Kaelber JT, Stansfeld PJ, Petrou VI. Structural basis of undecaprenyl phosphate glycosylation leading to polymyxin resistance in Gram-negative bacteria. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.29.634835. [PMID: 39974898 PMCID: PMC11838356 DOI: 10.1101/2025.01.29.634835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
In Gram-negative bacteria, the enzymatic modification of Lipid A with aminoarabinose (L-Ara4N) leads to resistance against polymyxin antibiotics and cationic antimicrobial peptides. ArnC, an integral membrane glycosyltransferase, attaches a formylated form of aminoarabinose to the lipid undecaprenyl phosphate, enabling its association with the bacterial inner membrane. Here, we present cryo-electron microscopy structures of ArnC from S. enterica in apo and nucleotide-bound conformations. These structures reveal a conformational transition that takes place upon binding of the partial donor substrate. Using coarse-grained and atomistic simulations, we provide insights into substrate coordination before and during catalysis, and we propose a catalytic mechanism that may operate on all similar metal-dependent polyprenyl phosphate glycosyltransferases. The reported structures provide a new target for drug design aiming to combat polymyxin resistance.
Collapse
Affiliation(s)
- Khuram U Ashraf
- Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers Biomedical Health Sciences, Newark, NJ, 07103, USA
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers Biomedical Health Sciences, Newark, NJ, 07103, USA
| | - Mariana Bunoro-Batista
- School of Life Sciences and Department of Chemistry, University of Warwick, Coventry, United Kingdom
| | - T. Bertie Ansell
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Ankita Punetha
- Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers Biomedical Health Sciences, Newark, NJ, 07103, USA
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers Biomedical Health Sciences, Newark, NJ, 07103, USA
| | - Stephannie Rosario-Garrido
- Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers Biomedical Health Sciences, Newark, NJ, 07103, USA
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers Biomedical Health Sciences, Newark, NJ, 07103, USA
| | - Emre Firlar
- Rutgers Cryo-EM & Nanoimaging Facility, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA
| | - Jason T. Kaelber
- Rutgers Cryo-EM & Nanoimaging Facility, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA
| | - Phillip J. Stansfeld
- School of Life Sciences and Department of Chemistry, University of Warwick, Coventry, United Kingdom
| | - Vasileios I. Petrou
- Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers Biomedical Health Sciences, Newark, NJ, 07103, USA
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers Biomedical Health Sciences, Newark, NJ, 07103, USA
| |
Collapse
|
2
|
Liu Q, Zhou J, Zheng Y, Xu B, Li D, Liu M, Zhang X, Wu X. Three methods to optimise polymyxin B dosing using estimated AUC after first dose: validation with the data generated by Monte Carlo simulation. Xenobiotica 2024; 54:615-623. [PMID: 38884560 DOI: 10.1080/00498254.2024.2370051] [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: 04/06/2024] [Revised: 06/16/2024] [Accepted: 06/16/2024] [Indexed: 06/18/2024]
Abstract
To achieve the AUC-guided dosing, we proposed three methods to estimate polymyxin B AUC across 24 h at steady state (AUCSS,24h) using limited concentrations after its first dose.Monte Carlo simulation based on a well-established population PK model was performed to generate the PK profiles of 1000 patients with normal or abnormal renal function. Polymyxin B AUCSS,24h was estimated for each subject using three methods (two-point PK approach, three-point PK approach, and four-point PK approach) based on limited concentration data in its first dose and compared with the actual AUC at steady state calculated using the linear-trapezoidal formula.In patients with normal renal function, the mean bias of two-point PK approach, three-point PK approach, and four-point PK approach was -8.73%, 1.37%, and -0.48%, respectively. The corresponding value was -11.15%, 1.99%, and -0.28% in patients with renal impairment, respectively. The largest mean bias of two-point PK approach, three-point PK approach, and four-point PK approach was -12.63%, -6.47%, and -0.54% when the sampling time shifted.The Excel calculators designed based on the three methods can be potentially used to optimise the dosing regimen of polymyxin B in the clinic.
Collapse
Affiliation(s)
- Qingxia Liu
- School of Pharmacy, Fujian Medical University, Fuzhou, China
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jianxing Zhou
- School of Pharmacy, Fujian Medical University, Fuzhou, China
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
| | - You Zheng
- School of Pharmacy, Fujian Medical University, Fuzhou, China
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
| | - Baohua Xu
- School of Pharmacy, Fujian Medical University, Fuzhou, China
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
| | - Dandan Li
- School of Pharmacy, Fujian Medical University, Fuzhou, China
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
| | - Maobai Liu
- School of Pharmacy, Fujian Medical University, Fuzhou, China
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaohan Zhang
- College of Arts and Sciences, University of Virginia, Charlottesville, VA, USA
| | - Xuemei Wu
- School of Pharmacy, Fujian Medical University, Fuzhou, China
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
3
|
Jalil AT, Alrawe RTA, Al-Saffar MA, Shaghnab ML, Merza MS, Abosaooda M, Latef R. The use of combination therapy for the improvement of colistin activity against bacterial biofilm. Braz J Microbiol 2024; 55:411-427. [PMID: 38030866 PMCID: PMC10920569 DOI: 10.1007/s42770-023-01189-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023] Open
Abstract
Colistin is used as a last resort for the management of infections caused by multi-drug resistant (MDR) bacteria. However, the use of this antibiotic could lead to different side effects, such as nephrotoxicity, in most patients, and the high prevalence of colistin-resistant strains restricts the use of colistin in the clinical setting. Additionally, colistin could induce resistance through the increased formation of biofilm; biofilm-embedded cells are highly resistant to antibiotics, and as with other antibiotics, colistin is impaired by bacteria in the biofilm community. In this regard, the researchers used combination therapy for the enhancement of colistin activity against bacterial biofilm, especially MDR bacteria. Different antibacterial agents, such as antimicrobial peptides, bacteriophages, natural compounds, antibiotics from different families, N-acetylcysteine, and quorum-sensing inhibitors, showed promising results when combined with colistin. Additionally, the use of different drug platforms could also boost the efficacy of this antibiotic against biofilm. The mentioned colistin-based combination therapy not only could suppress the formation of biofilm but also could destroy the established biofilm. These kinds of treatments also avoided the emergence of colistin-resistant subpopulations, reduced the required dosage of colistin for inhibition of biofilm, and finally enhanced the dosage of this antibiotic at the site of infection. However, the exact interaction of colistin with other antibacterial agents has not been elucidated yet; therefore, further studies are required to identify the precise mechanism underlying the efficient removal of biofilms by colistin-based combination therapy.
Collapse
Affiliation(s)
| | | | - Montaha A Al-Saffar
- Community Health Department, Institute of Medical Technology/Baghdad, Middle Technical University, Baghdad, Iraq
| | | | - Muna S Merza
- Prosthetic Dental Techniques Department, Al-Mustaqbal University College, Babylon, 51001, Iraq
| | - Munther Abosaooda
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Rahim Latef
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| |
Collapse
|
4
|
de Souza GH, Vaz MS, Dos Santos Radai JA, Fraga TL, Rossato L, Simionatto S. Synergistic interaction of polymyxin B with carvacrol: antimicrobial strategy against polymyxin-resistant Klebsiella pneumoniae. Future Microbiol 2024; 19:181-193. [PMID: 38329374 DOI: 10.2217/fmb-2023-0070] [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: 03/23/2023] [Accepted: 10/09/2023] [Indexed: 02/09/2024] Open
Abstract
Objective: The antimicrobial activities of the synergistic combination of carvacrol and polymyxin B against polymyxin-resistant Klebsiella pneumoniae were evaluated. Methods: The methods employed checkerboard assays to investigate synergism, biofilm inhibition assessment and membrane integrity assay. In addition, the study included in vivo evaluation using a mouse infection model. Results: The checkerboard method evaluated 48 combinations, with 23 indicating synergistic action. Among these, carvacrol 10 mg/kg plus polymyxin B 2 mg/kg exhibited in vivo antimicrobial activity in a mouse model of infection, resulting in increased survival and a significant decrease in bacterial load in the blood. Conclusion: Polymyxin in synergy with carvacrol represents a promising alternative to be explored in the development of new antimicrobials.
Collapse
Affiliation(s)
- Gleyce Ha de Souza
- Laboratório de Pesquisa em Ciências da Saúde, Universidade Federal da Grande Dourados - UFGD, Dourados, Mato Grosso do Sul, 79825-900, Brazil
| | - Marcia Sm Vaz
- Laboratório de Pesquisa em Ciências da Saúde, Universidade Federal da Grande Dourados - UFGD, Dourados, Mato Grosso do Sul, 79825-900, Brazil
| | - Joyce A Dos Santos Radai
- Laboratório de Pesquisa em Ciências da Saúde, Universidade Federal da Grande Dourados - UFGD, Dourados, Mato Grosso do Sul, 79825-900, Brazil
| | - Thiago L Fraga
- Centro Universitário da Grande Dourados - UNIGRAN, Dourados, Mato Grosso do Sul, 79824-900, Brazil
| | - Luana Rossato
- Laboratório de Pesquisa em Ciências da Saúde, Universidade Federal da Grande Dourados - UFGD, Dourados, Mato Grosso do Sul, 79825-900, Brazil
| | - Simone Simionatto
- Laboratório de Pesquisa em Ciências da Saúde, Universidade Federal da Grande Dourados - UFGD, Dourados, Mato Grosso do Sul, 79825-900, Brazil
| |
Collapse
|
5
|
Kim KY, Kim BH, Kwack WG, Kwon HJ, Cho SH, Kim CW. Simple and robust LC-MS/MS method for quantification of colistin methanesulfonate and colistin in human plasma for therapeutic drug monitoring. J Pharm Biomed Anal 2023; 236:115734. [PMID: 37776629 DOI: 10.1016/j.jpba.2023.115734] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/12/2023] [Accepted: 09/18/2023] [Indexed: 10/02/2023]
Abstract
A rapid, simple, and robust LC-MS/MS method was developed and validated for the quantitation of colistin and colistin methanesulfonate (CMS) in human plasma. The method also prevented overestimation of colistin concentration by establishing the stability of CMS under sample preparation conditions, including blood and plasma storage conditions. Polymyxin B1 was used as an internal standard, and positive-ion electrospray ionization in multiple reaction monitoring mode was used for quantification. Chromatographic separation was achieved using a Zorbax eclipse C18 column (3.5 µm, 2.1 mm i.d. × 100 mm), with a flow rate of 0.5 mL/min, 5 μL injection volume, and gradient elution with a mixture of acetonitrile-water (containing 0.1 % trifluoroacetic acid). The method had a quantifiable range of 0.043-8.61 and 0.057-11.39 μg/mL for colistin A and B in human plasma, respectively, under a total runtime of 6.0 min. Further, it demonstrated appropriate extraction efficiency, no significant interference from co-eluting endogenous compounds, and satisfactory intraday and interday precision and accuracy. The proposed procedure for sample preparation successfully addressed the issue of CMS instability, consequently diminishing the probability of overestimating the concentration of colistin. Therefore, this simple and robust LC-MS/MS method for CMS and colistin quantification in human plasma is a valuable tool for clinicians to accurately monitor colistin treatment in patients with infections caused by multidrug-resistant (MDR) Gram-negative bacteria.
Collapse
Affiliation(s)
- Kwang-Youl Kim
- Department of Clinical Pharmacology, Inha University Hospital, Inha University School of Medicine, Incheon, the Republic of Korea
| | - Bo-Hyung Kim
- Department of Clinical Pharmacology and Therapeutics, Kyung Hee University Hospital, Seoul, the Republic of Korea; East-West Medical Research Institute, Kyung Hee University, Seoul, the Republic of Korea
| | - Won Gun Kwack
- Department of Internal Medicine, Kyung Hee University Hospital, Seoul, the Republic of Korea
| | - Hyun-Jung Kwon
- Department of Clinical Pharmacology, Inha University Hospital, Inha University School of Medicine, Incheon, the Republic of Korea
| | - Sang-Heon Cho
- Department of Clinical Pharmacology, Inha University Hospital, Inha University School of Medicine, Incheon, the Republic of Korea
| | - Cheol-Woo Kim
- Department of Clinical Pharmacology, Inha University Hospital, Inha University School of Medicine, Incheon, the Republic of Korea; Department of Internal Medicine, Inha University Hospital, Inha University School of Medicine, Incheon, the Republic of Korea.
| |
Collapse
|
6
|
Hanafin PO, Kwa A, Zavascki AP, Sandri AM, Scheetz MH, Kubin CJ, Shah J, Cherng BPZ, Yin MT, Wang J, Wang L, Calfee DP, Bolon M, Pogue JM, Purcell AW, Nation RL, Li J, Kaye KS, Rao GG. A population pharmacokinetic model of polymyxin B based on prospective clinical data to inform dosing in hospitalized patients. Clin Microbiol Infect 2023; 29:1174-1181. [PMID: 37217076 DOI: 10.1016/j.cmi.2023.05.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 05/03/2023] [Accepted: 05/14/2023] [Indexed: 05/24/2023]
Abstract
OBJECTIVES To develop a population pharmacokinetic (PK) model with data from the largest polymyxin B-treated patient population studied to date to optimize its dosing in hospitalized patients. METHODS Hospitalized patients receiving intravenous polymyxin B for ≥48 hours were enrolled. Blood samples were collected at steady state and drug concentrations were analysed by liquid chromotography tandem mass spectrometry (LC-MS/MS). Population PK analysis and Monte Carlo simulations were performed to determine the probability of target attainment (PTA). RESULTS One hundred and forty-two patients received intravenous polymyxin B (1.33-6 mg/kg/day), providing 681 plasma samples. Twenty-four patients were on renal replacement therapy, including 13 on continuous veno-venous hemodiafiltration (CVVHDF). A 2-compartment model adequately described the PK with body weight as a covariate on the volume of distribution that affected Cmax, but it did not impact clearance or exposure. Creatinine clearance was a statistically significant covariate on clearance, although clinically relevant variations of dose-normalized drug exposure were not observed across a wide creatinine clearance range. The model described higher clearance in CVVHDF patients than in non-CVVHDF patients. Maintenance doses of ≥2.5 mg/kg/day or ≥150 mg/day had a PTA ≥90% (for non-pulmonary infections target) at a steady state for minimum inhibitory concentrations ≤2 mg/L. The PTA at a steady state for CVVHDF patients was lower. DISCUSSION Fixed loading and maintenance doses of polymyxin B seemed to be more appropriate than weight-based dosing regimens in patients weighing 45-90 kg. Higher doses may be needed in patients on CVVHDF. Substantial variability in polymyxin B clearance and volume of distribution was found, suggesting that therapeutic drug monitoring may be indicated.
Collapse
Affiliation(s)
- Patrick O Hanafin
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Andrea Kwa
- Department of Pharmacy, Singapore General Hospital, Singapore, Singapore; Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Alexandre P Zavascki
- Infectious Diseases Service, Hospital Moinhos de Vento, Porto Alegre, Brazil; Department of Internal Medicine, Medical School, Universidade Federal, Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Ana Maria Sandri
- Infection Control Department, Hospital São Lucas da Pontifícia Universidade Católica Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Marc H Scheetz
- Department of Pharmacy Practice, Midwestern University Chicago College of Pharmacy, Downers Grove, IL, USA
| | - Christine J Kubin
- New York-Presbyterian Hospital/Columbia University Irving Medical Center, New York, NY, USA
| | - Jayesh Shah
- Division of Infectious Diseases, Department of Internal Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Benjamin P Z Cherng
- Department of Infectious Diseases, Singapore General Hospital, Singapore, Singapore
| | - Michael T Yin
- Division of Infectious Diseases, Department of Internal Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Jiping Wang
- Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Lu Wang
- Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - David P Calfee
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Maureen Bolon
- Department of Healthcare Epidemiology and Infection Prevention, Northwestern Memorial Hospital, Chicago, IL, USA; Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jason M Pogue
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Anthony W Purcell
- Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Roger L Nation
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Jian Li
- Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Keith S Kaye
- Division of Allergy, Immunology and Infectious Diseases, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA.
| | - Gauri G Rao
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
7
|
Transcriptomic Responses to Polymyxin B and Analogues in Human Kidney Tubular Cells. Antibiotics (Basel) 2023; 12:antibiotics12020415. [PMID: 36830325 PMCID: PMC9952791 DOI: 10.3390/antibiotics12020415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Polymyxins are last-line antibiotics for the treatment of Gram-negative 'superbugs'. However, nephrotoxicity can occur in up to 60% of patients administered intravenous polymyxins. The mechanisms underpinning nephrotoxicity remain unclear. To understand polymyxin-induced nephrotoxicity, human renal proximal tubule cells were treated for 24 h with 0.1 mM polymyxin B or two new analogues, FADDI-251 or FADDI-287. Transcriptomic analysis was performed, and differentially expressed genes (DEGs) were identified using ANOVA (FDR < 0.2). Cell viability following treatment with polymyxin B, FADDI-251 or FADDI-287 was 66.0 ± 5.33%, 89.3 ± 3.96% and 90.4 ± 1.18%, respectively. Transcriptomics identified 430, 193 and 150 DEGs with polymyxin B, FADDI-251 and FADDI-287, respectively. Genes involved with metallothioneins and Toll-like receptor pathways were significantly perturbed by all polymyxins. Only polymyxin B induced perturbations in signal transduction, including FGFR2 and MAPK signaling. SIGNOR network analysis showed all treatments affected essential regulators in the immune system, autophagy, cell cycle, oxidative stress and apoptosis. All polymyxins caused significant perturbations of metal homeostasis and TLR signaling, while polymyxin B caused the most dramatic perturbations of the transcriptome. This study reveals the impact of polymyxin structure modifications on transcriptomic responses in human renal tubular cells and provides important information for designing safer new-generation polymyxins.
Collapse
|
8
|
The Impact of Colistin Resistance on the Activation of Innate Immunity by Lipopolysaccharide Modification. Infect Immun 2023; 91:e0001223. [PMID: 36722977 PMCID: PMC9933656 DOI: 10.1128/iai.00012-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Colistin resistance is acquired by different lipopolysaccharide (LPS) modifications. We proposed to evaluate the of effect in vivo colistin resistance acquisition on the innate immune response. We used a pair of ST11 clone Klebsiella pneumoniae strains: an OXA-48, CTX-M-15 K. pneumoniae strain susceptible to colistin (CS-Kp) isolated from a urinary infection and its colistin-resistant variant (CR-Kp) from the same patient after prolonged treatment with colistin. No mutation of previously described genes for colistin resistance (pmrA, pmrB, mgrB, phoP/Q, arnA, arnC, arnT, ugdH, and crrAB) was found in the CR-Kp genome; however, LPS modifications were characterized by negative-ion matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) mass spectrometry. The strains were cocultured with human monocytes to determine their survival after phagocytosis and induction to apoptosis. Also, monocytes were stimulated with bacterial LPS to study cytokine and immune checkpoint production. The addition of 4-amino-4-deoxy-l-arabinose (Ara4N) to lipid A of CR-Kp accounted for the colistin resistance. CR-Kp survived significantly longer inside human monocytes after being phagocytosed than did the CS-Kp strain. In addition, LPS from CR-Kp induced both higher apoptosis in monocytes and higher levels of cytokine and immune checkpoint production than LPS from CS-Kp. Our data reveal a variable impact of colistin resistance on the innate immune system, depending on the responsible mechanism. Adding Ara4N to LPS in K. pneumoniae increases bacterial survival after phagocytosis and elicits a higher inflammatory response than its colistin-susceptible counterpart.
Collapse
|
9
|
Haddad N, Carr M, Balian S, Lannin J, Kim Y, Toth C, Jarvis J. The Blood-Brain Barrier and Pharmacokinetic/Pharmacodynamic Optimization of Antibiotics for the Treatment of Central Nervous System Infections in Adults. Antibiotics (Basel) 2022; 11:antibiotics11121843. [PMID: 36551500 PMCID: PMC9774927 DOI: 10.3390/antibiotics11121843] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/08/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
Bacterial central nervous system (CNS) infections are serious and carry significant morbidity and mortality. They encompass many syndromes, the most common being meningitis, which may occur spontaneously or as a consequence of neurosurgical procedures. Many classes of antimicrobials are in clinical use for therapy of CNS infections, some with established roles and indications, others with experimental reporting based on case studies or small series. This review delves into the specifics of the commonly utilized antibacterial agents, updating their therapeutic use in CNS infections from the pharmacokinetic and pharmacodynamic perspectives, with a focus on the optimization of dosing and route of administration that have been described to achieve good clinical outcomes. We also provide a concise synopsis regarding the most focused, clinically relevant information as pertains to each class and subclass of antimicrobial therapeutics. CNS infection morbidity and mortality remain high, and aggressive management is critical in ensuring favorable patient outcomes while averting toxicity and upholding patient safety.
Collapse
Affiliation(s)
- Nicholas Haddad
- College of Medicine, Central Michigan University (CMU), Mt Pleasant, MI 48859, USA
- Correspondence: ; Tel.: +1-(989)-746-7860
| | | | - Steve Balian
- CMU Medical Education Partners, Saginaw, MI 48602, USA
| | | | - Yuri Kim
- CMU Medical Education Partners, Saginaw, MI 48602, USA
| | - Courtney Toth
- Ascension St. Mary’s Hospital, Saginaw, MI 48601, USA
| | | |
Collapse
|
10
|
Next-Generation Polymyxin Class of Antibiotics: A Ray of Hope Illuminating a Dark Road. Antibiotics (Basel) 2022; 11:antibiotics11121711. [PMID: 36551367 PMCID: PMC9774142 DOI: 10.3390/antibiotics11121711] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/21/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022] Open
Abstract
Although new-generation antimicrobials, in particular β-lactam/β-lactamase inhibitors, have largely replaced polymyxins in carbapenem-resistant Gram-negative bacterial infections, polymyxins are still needed for carbapanem-resistant Acinetobacter baumannii infections and in settings where novel agents are not readily available. Despite their potent in vitro activity, the clinical utility of polymyxins is significantly limited by their pharmacokinetic properties and nephrotoxicity risk. There is significant interest, therefore, in developing next-generation polymyxins with activity against colistin-resistant strains and lower toxicity than existing polymyxins. In this review, we aim to present the antibacterial activity mechanisms, in vitro and in vivo efficacy data, and toxicity profiles of new-generation polymyxins, including SPR206, MRX-8, and QPX9003, as well as the general characteristics of old polymyxins. Considering the emergence of colistin-resistant strains particularly in endemic regions, the restoration of the antimicrobial activity of polymyxins via PBT2 is also described in this review.
Collapse
|
11
|
Zhang J, Diao S, Liu Y, Wang H, Liu Y, Zhu S, Feng K, Tang X, Oo C, Zhu P, Lv Z, Yu M, Sy SKB, Zhu Y. The combination effect of meropenem/sulbactam/polymyxin-B on the pharmacodynamic parameters for mutant selection windows against carbapenem-resistant Acinetobacter baumannii. Front Microbiol 2022; 13:1024702. [PMID: 36483204 PMCID: PMC9723340 DOI: 10.3389/fmicb.2022.1024702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/07/2022] [Indexed: 01/25/2023] Open
Abstract
The objective of this study was to evaluate whether combinations of sulbactam, meropenem, and polymyxin-B could reduce or close the gap of mutant selection window (MSW) of individual antibiotics against Acinetobacter baumannii harboring OXA-23. MICs of three antimicrobials used alone and in combination (meropenem/polymyxin-B or meropenem/polymyxin-B/sulbactam) were obtained in 11 clinical isolates and mutant prevention concentrations were determined in 4 of the 11 isolates. All isolates were resistant to meropenem or polymyxin-B. Combining meropenem and polymyxin-B with or without sulbactam resulted in synergistic bactericidal activities. Pharmacokinetic (PK) simulations of drug concentrations in the blood and epithelial lining fluid coupled with pharmacodynamic (PD) evaluations revealed that the fractions of time over the 24-h in terms of free drug concentration within the MSW (fTMSW) and above the MPC (fT>MPC) were optimized by combination therapy. The resultant clinical regimens of meropenem, polymyxin-B, and sulbactam evaluated in the PK-PD analysis were 2 g q8h, 2.5 mg/kg loading dose followed by 1.5 mg/kg q12h, and 3 g q8h, respectively, in patients with normal renal function. Subsequent corresponding equivalent exposure regimens would depend on the extent of renal failure. The overall results indicate that combination antibiotics consisting of sulbactam/meropenem/polymyxin-B can confer potential efficacy against A. baumannii harboring OXA-23, and reduce the opportunity for bacteria to develop further resistance. This study provides a framework for pharmacodynamic evaluation of drug-resistant mutant suppression in an antimicrobial co-administration setting. The results thereby lay the groundwork for additional studies and future clinical confirmation is warranted.
Collapse
Affiliation(s)
- Jiayuan Zhang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Shuo Diao
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Yanfei Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hongxiang Wang
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
| | - Yuwei Liu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Shixing Zhu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Kun Feng
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Xiaoqian Tang
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
| | - Charles Oo
- SunLife Biopharma, Morris Plains, NJ, United States
| | - Peijuan Zhu
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, United States
| | - Zhihua Lv
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China,Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, China,*Correspondence: Zhihua Lv, ; Mingming Yu,
| | - Mingming Yu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China,Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, China,*Correspondence: Zhihua Lv, ; Mingming Yu,
| | - Sherwin K. B. Sy
- Department of Statistics, State University of Maringá, Maringá, Brazil
| | - Yuanqi Zhu
- Department of Laboratory Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
12
|
Kyriakoudi A, Pontikis K, Valsami G, Avgeropoulou S, Neroutsos E, Christodoulou E, Moraitou E, Markantonis SL, Dokoumetzidis A, Rello J, Koutsoukou A. Pharmacokinetic Characteristics of Nebulized Colistimethate Sodium Using Two Different Types of Nebulizers in Critically Ill Patients with Ventilator-Associated Respiratory Infections. Antibiotics (Basel) 2022; 11:1528. [PMID: 36358184 PMCID: PMC9686516 DOI: 10.3390/antibiotics11111528] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 05/25/2024] Open
Abstract
Background: Rising antimicrobial resistance has led to a revived interest in inhaled colistin treatment in the critically ill patient with ventilator-associated respiratory infection (VARI). Nebulization via vibrating mesh nebulizers (VMNs) is considered the current standard-of-care, yet the use of generic jet nebulizers (JNs) is more widespread. Few data exist on the intrapulmonary pharmacokinetics of colistin when administered through VMNs, while there is a complete paucity regarding the use of JNs. Methods: In this study, 18 VARI patients who received 2 million international units of inhaled colistimethate sodium (CMS) through a VMN were pharmacokinetically compared with six VARI patients who received the same drug dose through a JN, in the absence of systemic CMS administration. Results: Surprisingly, VMN and JN led to comparable formed colistin exposures in the epithelial lining fluid (ELF) (median (IQR) AUC0-24: 86.2 (46.0-185.9) mg/L∙h with VMN and 91.5 (78.1-110.3) mg/L∙h with JN). The maximum ELF concentration was 10.4 (4.7-22.6) mg/L and 7.4 (6.2-10.3) mg/L, respectively. Conclusions: Based on our results, JN might be considered a viable alternative to the theoretically superior VMN. Therapeutic drug monitoring in the ELF can be advised due to the observed low exposure, high variability, and appreciable systemic absorption.
Collapse
Affiliation(s)
- Anna Kyriakoudi
- Intensive Care Unit, 1st Department of Pulmonology, Medical School, National & Kapodistrian University of Athens, General Hospital for the Diseases of the Chest “I Sotiria”, 11527 Athens, Greece
| | - Konstantinos Pontikis
- Intensive Care Unit, 1st Department of Pulmonology, Medical School, National & Kapodistrian University of Athens, General Hospital for the Diseases of the Chest “I Sotiria”, 11527 Athens, Greece
| | - Georgia Valsami
- Department of Pharmacy, School of Health Sciences, National & Kapodistrian University of Athens, 15784 Athens, Greece
| | - Stavrina Avgeropoulou
- Intensive Care Unit, 1st Department of Pulmonology, Medical School, National & Kapodistrian University of Athens, General Hospital for the Diseases of the Chest “I Sotiria”, 11527 Athens, Greece
| | - Efthymios Neroutsos
- Department of Pharmacy, School of Health Sciences, National & Kapodistrian University of Athens, 15784 Athens, Greece
| | - Eirini Christodoulou
- Department of Pharmacy, School of Health Sciences, National & Kapodistrian University of Athens, 15784 Athens, Greece
| | - Eleni Moraitou
- Microbiology Department, General Hospital for the Diseases of the Chest “I Sotiria”, 11527 Athens, Greece
| | - Sophia L. Markantonis
- Department of Pharmacy, School of Health Sciences, National & Kapodistrian University of Athens, 15784 Athens, Greece
| | - Aristides Dokoumetzidis
- Department of Pharmacy, School of Health Sciences, National & Kapodistrian University of Athens, 15784 Athens, Greece
| | - Jordi Rello
- Clinical Research in Pneumonia (CRIPS), Vall d’Hebron Institute of Research, 08035 Barcelona, Spain
- Clinical Research, CHU Nîmes, 30900 Nîmes, France
| | - Antonia Koutsoukou
- Intensive Care Unit, 1st Department of Pulmonology, Medical School, National & Kapodistrian University of Athens, General Hospital for the Diseases of the Chest “I Sotiria”, 11527 Athens, Greece
| |
Collapse
|
13
|
Zhu S, Song C, Zhang J, Diao S, Heinrichs TM, Martins FS, Lv Z, Zhu Y, Yu M, Sy SKB. Effects of amikacin, polymyxin-B, and sulbactam combination on the pharmacodynamic indices of mutant selection against multi-drug resistant Acinetobacter baumannii. Front Microbiol 2022; 13:1013939. [PMID: 36338049 PMCID: PMC9632654 DOI: 10.3389/fmicb.2022.1013939] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/29/2022] [Indexed: 12/01/2022] Open
Abstract
Amikacin and polymyxins as monotherapies are ineffective against multidrug-resistant Acinetobacter baumannii at the clinical dose. When polymyxins, aminoglycosides, and sulbactam are co-administered, the combinations exhibit in vitro synergistic activities. The minimum inhibitory concentration (MIC) and mutant prevention concentration (MPC) were determined in 11 and 5 clinical resistant isolates of A. baumannii harboring OXA-23, respectively, in order to derive the fraction of time over the 24-h wherein the free drug concentration was within the mutant selection window (fTMSW) and the fraction of time that the free drug concentration was above the MPC (fT>MPC) from simulated pharmacokinetic profiles. The combination of these three antibiotics can confer susceptibility in multi-drug resistant A. baumannii and reduce the opportunity for bacteria to develop further resistance. Clinical intravenous dosing regimens of amikacin, polymyxin-B, and sulbactam were predicted to optimize fTMSW and fT>MPC from drug exposures in the blood. Mean fT>MPC were ≥ 60% and ≥ 80% for amikacin and polymyxin-B, whereas mean fTMSW was reduced to <30% and <15%, respectively, in the triple antibiotic combination. Due to the low free drug concentration of amikacin and polymyxin-B simulated in the epithelial lining fluid, the two predicted pharmacodynamic parameters in the lung after intravenous administration were not optimal even in the combination therapy setting.
Collapse
Affiliation(s)
- Shixing Zhu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Chu Song
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Jiayuan Zhang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Shuo Diao
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Tobias M. Heinrichs
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Frederico S. Martins
- Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Zhihua Lv
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
- *Correspondence: Zhihua Lv,
| | - Yuanqi Zhu
- Department of Laboratory Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Mingming Yu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
- Mingming Yu,
| | - Sherwin K. B. Sy
- Department of Statistics, State University of Maringá, Paraná, Brazil
- Sherwin K. B. Sy,
| |
Collapse
|
14
|
Samarkos M, Papanikolaou K, Sourdi A, Paisios N, Mainas E, Paramythiotou E, Antoniadou A, Sambatakou H, Gargalianos-Kakolyris P, Skoutelis A, Daikos GL. The Effect of Different Colistin Dosing Regimens on Nephrotoxicity: A Cohort Study. Antibiotics (Basel) 2022; 11:antibiotics11081066. [PMID: 36009935 PMCID: PMC9405298 DOI: 10.3390/antibiotics11081066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/03/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
(1) Background: It is not known whether different daily dosing schemes have different effects on colistin nephrotoxicity. We examined the effect of once- versus twice- or thrice-daily doses of colistin on renal function. (2) Methods: We performed a multicenter retrospective cohort study of hospitalized patients with a baseline glomerular filtration rate ≥ 50 mL/min who received intravenously the same colistin dose once (regimen A), twice (regimen B) or thrice daily (regimen C). The primary endpoint was acute kidney injury (AKI), defined as fulfilment of any of the RIFLE (Risk-Injury-Failure-Loss-End stage renal disease) criteria. (3) Results: We included 306 patients; 132 (43.1%) received regimen A, 151 (49.3%) regimen B, and 23 (7.5%) regimen C. Ninety-nine (32.4%) patients developed AKI; there was no difference between regimen A vs. B and C [45 (34.1%) vs. 54 (31.0%), p = 0.57]. In a propensity score−matched cohort, AKI was similar in patients receiving Regimen A, Regimen B, and Regimen C (31.6% vs. 33.3%, p = 0.78). On logistic regression analysis, diabetes was an independent predictor of AKI (OR = 4.59, 95% CI 2.03−10.39, p = 0.001) while eGFR > 80 mL/min (OR = 0.50, 95% CI 0.25−0.99, p = 0.048) was inversely associated with AKI. (4) Conclusions: Colistin once daily is not more nephrotoxic than the standard colistin regimens. The only independent predictor of nephrotoxicity was diabetes mellitus, while eGFR > 80 mL/min had a protective effect.
Collapse
Affiliation(s)
- Michael Samarkos
- 1st Department of Medicine, Laikon General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | | | - Athena Sourdi
- 1st Propaedeutic Department of Medicine, Laikon General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Nikolaos Paisios
- 1st Department of Medicine, G. Gennimatas General Hospital, 11527 Athens, Greece
| | - Efstratios Mainas
- 2nd Department of Medicine, Ippokrateion General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | | | - Anastasia Antoniadou
- 4th Department of Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Helen Sambatakou
- 2nd Department of Medicine, Ippokrateion General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | | | | | - George L. Daikos
- 1st Department of Medicine, Laikon General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Correspondence:
| |
Collapse
|
15
|
Ciloglu FU, Hora M, Gundogdu A, Kahraman M, Tokmakci M, Aydin O. SERS-based sensor with a machine learning based effective feature extraction technique for fast detection of colistin-resistant Klebsiella pneumoniae. Anal Chim Acta 2022; 1221:340094. [DOI: 10.1016/j.aca.2022.340094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 06/12/2022] [Accepted: 06/13/2022] [Indexed: 11/01/2022]
|
16
|
Ahmad A, Nii T, Mori T, Katayama Y, Toyofuku M, Kishimura A. Nanostructure Control of an Antibiotic-based Polyion Complex Using a Series of Polycations with Different Side-chain Modification Rates. Macromol Rapid Commun 2022; 43:e2200316. [PMID: 35661316 DOI: 10.1002/marc.202200316] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/07/2022] [Indexed: 11/08/2022]
Abstract
Developing nanovehicles for delivering antibiotics is a promising approach to overcome the issue of antibiotic resistance. This study aims to utilize a polyion complex (PICs) system for developing novel nanovehicles for polymyxin-type antibiotics, which are known as last resort drugs. The formation of antibiotic-based PIC nanostructures was investigated using colistimethate sodium (CMS), an anionic cyclic short peptide, and a series of block catiomers bearing different amounts of guanidinium moieties on their side chains. In addition, only the modified catiomer, and not the unmodified catiomer, self-assembles with CMS, implying the importance of the guanidine moieties for enhancing the interaction between the catiomer and CMS via the formation of multivalent hydrogen bonding. Moreover, micellar and vesicular PIC nanostructures are selectively formed depending on the ratio of the guanidine residues. Size-exclusion chromatography revealed that the encapsulation efficiency of CMS is dependent on the guanidinium modification ratio. The antimicrobial activity of the PIC nanostructures is also confirmed, indicating that the complexation of CMS in the PICs and further release from the PICs successfully occurs. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Asmariah Ahmad
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Teruki Nii
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Takeshi Mori
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka, 819-0395, Japan.,Center for Future Chemistry, Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Yoshiki Katayama
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka, 819-0395, Japan.,Center for Future Chemistry, Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka, 819-0395, Japan.,Center for Molecular Systems, Kyushu University 744 Moto-oka, Nishi-ku, Fukuoka, 819-0395, Japan.,Center for Advanced Medical Innovation, Kyushu University 3-1-1 Maedashi, Higashi-ku, Fukuoka, 812-8582, Japan.,Department of Biomedical Engineering, Chung Yuan Christian University, 200 Chung Pei Rd., Chung Li, 32023, Taiwan, ROC
| | - Masanori Toyofuku
- Faculty of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Japan.,Microbiology Research Center for Sustainability (MiCS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Japan
| | - Akihiro Kishimura
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka, 819-0395, Japan.,Center for Future Chemistry, Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka, 819-0395, Japan.,Center for Molecular Systems, Kyushu University 744 Moto-oka, Nishi-ku, Fukuoka, 819-0395, Japan.,RIKEN Center for Emergent Matter Science, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| |
Collapse
|
17
|
Bian X, Qu X, Zhang J, Nang SC, Bergen PJ, Tony Zhou Q, Chan HK, Feng M, Li J. Pharmacokinetics and pharmacodynamics of peptide antibiotics. Adv Drug Deliv Rev 2022; 183:114171. [PMID: 35189264 PMCID: PMC10019944 DOI: 10.1016/j.addr.2022.114171] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 01/23/2022] [Accepted: 02/16/2022] [Indexed: 01/05/2023]
Abstract
Antimicrobial resistance is a major global health challenge. As few new efficacious antibiotics will become available in the near future, peptide antibiotics continue to be major therapeutic options for treating infections caused by multidrug-resistant pathogens. Rational use of antibiotics requires optimisation of the pharmacokinetics and pharmacodynamics for the treatment of different types of infections. Toxicodynamics must also be considered to improve the safety of antibiotic use and, where appropriate, to guide therapeutic drug monitoring. This review focuses on the pharmacokinetics/pharmacodynamics/toxicodynamics of peptide antibiotics against multidrug-resistant Gram-negative and Gram-positive pathogens. Optimising antibiotic exposure at the infection site is essential for improving their efficacy and minimising emergence of resistance.
Collapse
Affiliation(s)
- Xingchen Bian
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China; Key Laboratory of Clinical Pharmacology of Antibiotics, Shanghai, China; National Health Commission & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China; School of Pharmacy, Fudan University, Shanghai, China
| | - Xingyi Qu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China; Key Laboratory of Clinical Pharmacology of Antibiotics, Shanghai, China; National Health Commission & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China; School of Pharmacy, Fudan University, Shanghai, China; Phase I Unit, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jing Zhang
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China; Key Laboratory of Clinical Pharmacology of Antibiotics, Shanghai, China; National Health Commission & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China; Phase I Unit, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Sue C Nang
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Melbourne, Australia
| | - Phillip J Bergen
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Melbourne, Australia
| | - Qi Tony Zhou
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, IN, USA
| | - Hak-Kim Chan
- Advanced Drug Delivery Group, School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Meiqing Feng
- School of Pharmacy, Fudan University, Shanghai, China
| | - Jian Li
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Melbourne, Australia.
| |
Collapse
|
18
|
The Role of Colistin in the Era of New β-Lactam/β-Lactamase Inhibitor Combinations. Antibiotics (Basel) 2022; 11:antibiotics11020277. [PMID: 35203879 PMCID: PMC8868358 DOI: 10.3390/antibiotics11020277] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/17/2022] [Accepted: 02/17/2022] [Indexed: 02/06/2023] Open
Abstract
With the current crisis related to the emergence of carbapenem-resistant Gram-negative bacteria (CR-GNB), classical treatment approaches with so-called “old-fashion antibiotics” are generally unsatisfactory. Newly approved β-lactam/β-lactamase inhibitors (BLBLIs) should be considered as the first-line treatment options for carbapenem-resistant Enterobacterales (CRE) and carbapenem-resistant Pseudomonas aeruginosa (CRPA) infections. However, colistin can be prescribed for uncomplicated lower urinary tract infections caused by CR-GNB by relying on its pharmacokinetic and pharmacodynamic properties. Similarly, colistin can still be regarded as an alternative therapy for infections caused by carbapenem-resistant Acinetobacter baumannii (CRAB) until new and effective agents are approved. Using colistin in combination regimens (i.e., including at least two in vitro active agents) can be considered in CRAB infections, and CRE infections with high risk of mortality. In conclusion, new BLBLIs have largely replaced colistin for the treatment of CR-GNB infections. Nevertheless, colistin may be needed for the treatment of CRAB infections and in the setting where the new BLBLIs are currently unavailable. In addition, with the advent of rapid diagnostic methods and novel antimicrobials, the application of personalized medicine has gained significant importance in the treatment of CRE infections.
Collapse
|
19
|
Tan X, Qiao J, Li H, Huang D, Hu X, Wang X. Global metabolic regulation in Vibrio parahaemolyticus under polymyxin B stimulation. Microb Pathog 2021; 161:105260. [PMID: 34688850 DOI: 10.1016/j.micpath.2021.105260] [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: 06/29/2021] [Revised: 09/25/2021] [Accepted: 10/13/2021] [Indexed: 11/17/2022]
Abstract
Vibrio parahaemolyticus is responsible for infection diseases of people who consume the contaminated seafood, but its metabolic regulation profile in response to colistin, the last treatment option for multidrug-resistant Gram-negative bacteria, remains unclear. In this study, the metabolic regulation profile of V. parahaemolyticus ATCC33846 under polymyxin B stimulation has been investigated. V. parahaemolyticus exposed to polymyxin B resulted in 4597 differentially transcribed genes, including 673 significantly up-regulated genes and 569 significantly down-regulated genes. In V. parahaemolyticus under polymyxin B stimulation, the cellular antioxidant systems to prevent bacteria from oxidant stress was activated, the synthesis of some nonessential macromolecules was reduced, and the assembly and modification of lipopolysaccharide and peptidoglycan to resist the attack from other antibiotics were promoted. These findings provide new insights into polymyxin B-related stress response in V. parahaemolyticus which should be useful for developing novel drugs for infection.
Collapse
Affiliation(s)
- Xin Tan
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Jun Qiao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Hedan Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Danyang Huang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Xiaoqing Hu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Xiaoyuan Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China; International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
20
|
Nebulized Colistin in Ventilator-Associated Pneumonia and Tracheobronchitis: Historical Background, Pharmacokinetics and Perspectives. Microorganisms 2021; 9:microorganisms9061154. [PMID: 34072189 PMCID: PMC8227626 DOI: 10.3390/microorganisms9061154] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 05/14/2021] [Accepted: 05/20/2021] [Indexed: 11/17/2022] Open
Abstract
Clinical evidence suggests that nebulized colistimethate sodium (CMS) has benefits for treating lower respiratory tract infections caused by multidrug-resistant Gram-negative bacteria (GNB). Colistin is positively charged, while CMS is negatively charged, and both have a high molecular mass and are hydrophilic. These physico-chemical characteristics impair crossing of the alveolo-capillary membrane but enable the disruption of the bacterial wall of GNB and the aggregation of the circulating lipopolysaccharide. Intravenous CMS is rapidly cleared by glomerular filtration and tubular excretion, and 20-25% is spontaneously hydrolyzed to colistin. Urine colistin is substantially reabsorbed by tubular cells and eliminated by biliary excretion. Colistin is a concentration-dependent antibiotic with post-antibiotic and inoculum effects. As CMS conversion to colistin is slower than its renal clearance, intravenous administration can lead to low plasma and lung colistin concentrations that risk treatment failure. Following nebulization of high doses, colistin (200,000 international units/24h) lung tissue concentrations are > five times minimum inhibitory concentration (MIC) of GNB in regions with multiple foci of bronchopneumonia and in the range of MIC breakpoints in regions with confluent pneumonia. Future research should include: (1) experimental studies using lung microdialysis to assess the PK/PD in the interstitial fluid of the lung following nebulization of high doses of colistin; (2) superiority multicenter randomized controlled trials comparing nebulized and intravenous CMS in patients with pandrug-resistant GNB ventilator-associated pneumonia and ventilator-associated tracheobronchitis; (3) non-inferiority multicenter randomized controlled trials comparing nebulized CMS to intravenous new cephalosporines/ß-lactamase inhibitors in patients with extensive drug-resistant GNB ventilator-associated pneumonia and ventilator-associated tracheobronchitis.
Collapse
|
21
|
Abstract
Antibiotic resistance is a major global health challenge and, worryingly, several key Gram negative pathogens can become resistant to most currently available antibiotics. Polymyxins have been revived as a last-line therapeutic option for the treatment of infections caused by multidrug-resistant Gram negative bacteria, in particular Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacterales. Polymyxins were first discovered in the late 1940s but were abandoned soon after their approval in the late 1950s as a result of toxicities (e.g., nephrotoxicity) and the availability of "safer" antibiotics approved at that time. Therefore, knowledge on polymyxins had been scarce until recently, when enormous efforts have been made by several research teams around the world to elucidate the chemical, microbiological, pharmacokinetic/pharmacodynamic, and toxicological properties of polymyxins. One of the major achievements is the development of the first scientifically based dosage regimens for colistin that are crucial to ensure its safe and effective use in patients. Although the guideline has not been developed for polymyxin B, a large clinical trial is currently being conducted to optimize its clinical use. Importantly, several novel, safer polymyxin-like lipopeptides are developed to overcome the nephrotoxicity, poor efficacy against pulmonary infections, and narrow therapeutic windows of the currently used polymyxin B and colistin. This review discusses the latest achievements on polymyxins and highlights the major challenges ahead in optimizing their clinical use and discovering new-generation polymyxins. To save lives from the deadly infections caused by Gram negative "superbugs," every effort must be made to improve the clinical utility of the last-line polymyxins. SIGNIFICANCE STATEMENT: Antimicrobial resistance poses a significant threat to global health. The increasing prevalence of multidrug-resistant (MDR) bacterial infections has been highlighted by leading global health organizations and authorities. Polymyxins are a last-line defense against difficult-to-treat MDR Gram negative pathogens. Unfortunately, the pharmacological information on polymyxins was very limited until recently. This review provides a comprehensive overview on the major achievements and challenges in polymyxin pharmacology and clinical use and how the recent findings have been employed to improve clinical practice worldwide.
Collapse
Affiliation(s)
- Sue C Nang
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Melbourne, Victoria, Australia (S.C.N., M.A.K.A., J.L.); Department of Pharmacology and Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia (T.V.); and Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana (Q.T.Z.)
| | - Mohammad A K Azad
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Melbourne, Victoria, Australia (S.C.N., M.A.K.A., J.L.); Department of Pharmacology and Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia (T.V.); and Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana (Q.T.Z.)
| | - Tony Velkov
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Melbourne, Victoria, Australia (S.C.N., M.A.K.A., J.L.); Department of Pharmacology and Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia (T.V.); and Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana (Q.T.Z.)
| | - Qi Tony Zhou
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Melbourne, Victoria, Australia (S.C.N., M.A.K.A., J.L.); Department of Pharmacology and Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia (T.V.); and Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana (Q.T.Z.)
| | - Jian Li
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Melbourne, Victoria, Australia (S.C.N., M.A.K.A., J.L.); Department of Pharmacology and Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia (T.V.); and Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana (Q.T.Z.)
| |
Collapse
|
22
|
Ram K, Sheikh S, Bhati RK, Tripathi CD, Suri JC, Meshram GG. Steady-state pharmacokinetic and pharmacodynamic profiling of colistin in critically ill patients with multi-drug-resistant gram-negative bacterial infections, along with differences in clinical, microbiological and safety outcome. Basic Clin Pharmacol Toxicol 2020; 128:128-140. [PMID: 33245629 DOI: 10.1111/bcpt.13482] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 07/11/2020] [Accepted: 08/11/2020] [Indexed: 12/29/2022]
Abstract
Limited data are present regarding the steady-state pharmacokinetics and pharmacodynamics of colistin in critically ill patients suffering from multi-drug-resistant gram-negative bacterial (MDR-GNB) infections. We aimed to profile the steady-state pharmacokinetics and pharmacodynamics of colistin in critically ill patients with MDR-GNB infections, along with determining the predictors that could influence the clinical, microbiological and safety outcome. We recruited 30 critically ill patients suffering from MDR-GNB infections in our prospective open-label study. Intravenous colistimethate sodium (CMS) 2 million IU was administered concurrently with inhalational CMS 1 million IU every 8 hours. Steady-state plasma colistin levels were measured. Logistic regression analysis was used to identify various predictors of clinical, microbiological and safety outcome. A large variability was observed in the steady-state colistin pharmacokinetic/pharmacodynamic parameters, along with the factors that influenced the clinical, microbiological and safety outcome. In conclusion, steady-state colistin pharmacokinetic and pharmacodynamic parameters observed in our study were largely consistent with those reported in previous studies. High acute physiology and chronic health evaluation II scores were associated with poor clinical outcome. Log-transformed colistin maximum concentration, area under the plasma concentration curve for 8 hours, apparent total body clearance and apparent volume of distribution were significantly associated with the safety outcome.
Collapse
Affiliation(s)
- Kishna Ram
- Department of Pharmacology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Salim Sheikh
- Department of Pharmacology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.,Department of Pharmacology, Employees' State Insurance Corporation Medical College and Hospital, Faridabad, India
| | - Rahul Kumar Bhati
- Department of Pharmacology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Chakra Dhar Tripathi
- Department of Pharmacology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.,Department of Pharmacology, Employees' State Insurance Corporation Medical College and Hospital, Faridabad, India
| | - Jagdish Chander Suri
- Department of Pulmonary Critical Care and Sleep Medicine, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Girish Gulab Meshram
- Department of Pharmacology, Employees' State Insurance Corporation Medical College and Hospital, Faridabad, India
| |
Collapse
|
23
|
Ayoub Moubareck C. Polymyxins and Bacterial Membranes: A Review of Antibacterial Activity and Mechanisms of Resistance. MEMBRANES 2020; 10:membranes10080181. [PMID: 32784516 PMCID: PMC7463838 DOI: 10.3390/membranes10080181] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 12/19/2022]
Abstract
Following their initial discovery in the 1940s, polymyxin antibiotics fell into disfavor due to their potential clinical toxicity, especially nephrotoxicity. However, the dry antibiotic development pipeline, together with the rising global prevalence of infections caused by multidrug-resistant (MDR) Gram-negative bacteria have both rejuvenated clinical interest in these polypeptide antibiotics. Parallel to the revival of their use, investigations into the mechanisms of action and resistance to polymyxins have intensified. With an initial known effect on biological membranes, research has uncovered the detailed molecular and chemical interactions that polymyxins have with Gram-negative outer membranes and lipopolysaccharide structure. In addition, genetic and epidemiological studies have revealed the basis of resistance to these agents. Nowadays, resistance to polymyxins in MDR Gram-negative pathogens is well elucidated, with chromosomal as well as plasmid-encoded, transferrable pathways. The aims of the current review are to highlight the important chemical, microbiological, and pharmacological properties of polymyxins, to discuss their mechanistic effects on bacterial membranes, and to revise the current knowledge about Gram-negative acquired resistance to these agents. Finally, recent research, directed towards new perspectives for improving these old agents utilized in the 21st century, to combat drug-resistant pathogens, is summarized.
Collapse
|
24
|
Wang P, Zhang Q, Zhu Z, Feng M, Sun T, Yang J, Zhang X. Population Pharmacokinetics and Limited Sampling Strategy for Therapeutic Drug Monitoring of Polymyxin B in Chinese Patients With Multidrug-Resistant Gram-Negative Bacterial Infections. Front Pharmacol 2020; 11:829. [PMID: 32581795 PMCID: PMC7289991 DOI: 10.3389/fphar.2020.00829] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 05/20/2020] [Indexed: 12/17/2022] Open
Abstract
Polymyxin B is used as a last therapeutic option for the treatment of multidrug-resistant Gram-negative bacterial infections. This study aimed to develop a population pharmacokinetic model and limited sampling strategy, a method to estimate the area under the concentration curve (AUC) by using a limited number of samples, to assist therapeutic drug monitoring of polymyxin B in Chinese patients. Population pharmacokinetic analysis was performed using Phoenix® NLME with data obtained from 46 adult patients at steady state. Various demographic variables were investigated as potential covariates for population pharmacokinetic modeling. The limited sampling strategies based on the Bayesian approach and multiple linear regression were validated using the intraclass correlation coefficient and Bland-Altman analysis. As a result, the data was described by a two-compartment population pharmacokinetic model. Through the modeling, creatinine clearance was found to be a statistically significant covariate influencing polymyxin B clearance. The limited sampling strategies showed the two-point model (C0h and C2h) could predict polymyxin B exposure with good linear relativity (r2 > 0.98), and the four-point model (C1h, C1.5h, C4h, and C8h) performed best in predicting polymyxin B AUC (r2 > 0.99). In conclusion, this study successfully developed a population pharmacokinetic model and limited sampling strategies that could be applied in clinical practice to assist in therapeutic drug monitoring of polymyxin B in Chinese patients.
Collapse
Affiliation(s)
- Peile Wang
- Department of Pharmacy, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Qiwen Zhang
- Department of Pharmacy, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Zhenfeng Zhu
- Department of Pharmacy, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Min Feng
- Department of ICU, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tongwen Sun
- Department of General ICU, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Yang
- Department of Pharmacy, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Xiaojian Zhang
- Department of Pharmacy, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
25
|
Comparison of the recommended colistin susceptibility testing methods with colistin gradient strips and semi-automated method for antimicrobial-resistant non-fermenting rods. J Microbiol Methods 2020; 172:105905. [PMID: 32229263 DOI: 10.1016/j.mimet.2020.105905] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/25/2020] [Accepted: 03/25/2020] [Indexed: 11/21/2022]
Abstract
An increased frequency of multidrug-resistant non-fermenting rods isolation has resulted in the excessive use of colistin - often the last chance antimicrobial. However, determination of colistin susceptibility is difficult, mainly because of its structure and limited diffusion properties. This study was performed to compare colistin susceptibility testing among Pseudomonas aeruginosa (n = 49) and Acinetobacter baumannii (n = 49) strains. Four methods were applied: colistin gradient strips (Liofilchem, Italy), semi-automated method Phoenix BD (Becton Dickinson, USA) and two broth microdilution methods: SensiTest Colistin (Liofilchem, Italy) and MICRONAUT MIC-Strip (MERLIN Diagnostika GmbH, Germany). Data were analyzed by comparison of MIC values and strains susceptibility interpretation criteria (resistant and sensitive, respectively). The same interpretation results were obtained for 46 (93.9%) P. aeruginosa and 37 (75.5%) A. baumannii isolates in all of the applied methods. Using broth microdilution methods, the same interpretation was obtained for 48 (98.0%) P. aeruginosa and 42 (85.7%) A. baumannii isolates. The results obtained by colistin gradient strips usually confirm the results of broth microdilution tests for P. aeruginosa isolates, the automated method is in turn less labor-intensive. However, MIC values, obtained with their use, are less precise because of the antibiotic dilutions limited to only several concentrations. The results underline the importance of choosing of the appropriate type of method, also among those recommended and commercially available.
Collapse
|
26
|
Extensive Therapeutic Drug Monitoring of Colistin in Critically Ill Patients Reveals Undetected Risks. Microorganisms 2020; 8:microorganisms8030415. [PMID: 32183443 PMCID: PMC7143967 DOI: 10.3390/microorganisms8030415] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/05/2020] [Accepted: 03/12/2020] [Indexed: 01/16/2023] Open
Abstract
(1) Background: With the rise of multi-/pan-drug resistant (MDR/PDR) pathogens, the less utilized antibiotic Colistin has made a comeback. Colistin fell out of favor due to its small therapeutic range and high potential for toxicity. Today, it is used again as a last resort substance in treating MDR/PDR pathogens. Although new guidelines with detailed recommendations for Colistin dosing are available, finding the right dose in critically ill patients with renal failure remains difficult. Here, we evaluate the efficiency of the current guidelines' recommendations by using high resolution therapeutic drug monitoring of Colistin. (2) Methods: We analyzed plasma levels of Colistin and its prodrug colisthimethate sodium (CMS) in 779 samples, drawn from eight PDR-infected ICU patients, using a HPLC-MS/MS approach. The impact of renal function on proper Colistin target levels was assessed. (3) Results: CMS levels did not correlate with Colistin levels. Over-/Underdosing occurred regardless of renal function and mode of renal replacement therapy. Colistin elimination half-time appeared to be longer than previously reported. (4) Conclusion: Following dose recommendations from the most current guidelines does not necessarily lead to adequate Colistin plasma levels. Use of Colistin without therapeutic drug monitoring might be unsafe and guideline adherence does not warrant efficient target levels in critically ill patients.
Collapse
|
27
|
Tang H, Zhang Y, Ma J, Dong Y, Gao Q, Feng J. Design, synthesis and antimicrobial studies of some polymyxin analogues. J Antibiot (Tokyo) 2019; 73:158-166. [DOI: 10.1038/s41429-019-0262-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/27/2019] [Accepted: 11/27/2019] [Indexed: 01/14/2023]
|
28
|
Su M, Wang M, Hong Y, Nimmagadda A, Shen N, Shi Y, Gao R, Zhang E, Cao C, Cai J. Polymyxin derivatives as broad-spectrum antibiotic agents. Chem Commun (Camb) 2019; 55:13104-13107. [PMID: 31612170 PMCID: PMC10484568 DOI: 10.1039/c9cc06908a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
We designed a few polymyxin derivatives which exhibit broad-spectrum antimicrobial activity. Lead compound P1 could disrupt bacterial membranes rapidly without developing resistance, inhibit biofilms formed by E. coli, and exhibit excellent in vivo activity in an MRSA-infected thigh burden mouse model.
Collapse
Affiliation(s)
- Ma Su
- Department of Chemistry, University of South Florida, Tampa, FL 33620, USA.
| | - Minghui Wang
- Department of Chemistry, University of South Florida, Tampa, FL 33620, USA.
| | - Yuzhu Hong
- College of Pharmacy, University of South Florida, Tampa, FL 33620, USA.
| | - Alekhya Nimmagadda
- Department of Chemistry, University of South Florida, Tampa, FL 33620, USA.
| | - Ning Shen
- College of Pharmacy, University of South Florida, Tampa, FL 33620, USA.
| | - Yan Shi
- Department of Chemistry, University of South Florida, Tampa, FL 33620, USA.
| | - Ruixuan Gao
- Department of Chemistry, University of South Florida, Tampa, FL 33620, USA.
| | - En Zhang
- Department of Chemistry, University of South Florida, Tampa, FL 33620, USA. and School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, P. R. China
| | - Chuanhai Cao
- College of Pharmacy, University of South Florida, Tampa, FL 33620, USA.
| | - Jianfeng Cai
- Department of Chemistry, University of South Florida, Tampa, FL 33620, USA.
| |
Collapse
|
29
|
Mesini A, Loy A, Gattorno M, Moscatelli A, Bandettini R, Faraci M, Cangemi G, Castagnola E. Colistin Area Under the Time-Concentration in Children Treated With Intravenous Loading Dose and Maintenance Therapy. Clin Infect Dis 2019; 66:808-809. [PMID: 29020309 DOI: 10.1093/cid/cix757] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Alessio Mesini
- Infectious Diseases Unit, University of Genoa, S. Martino Hospital
| | - Anna Loy
- Istituto Giannina Gaslini, Children's Hospital Genoa, Italy
| | - Marco Gattorno
- Istituto Giannina Gaslini, Children's Hospital Genoa, Italy
| | | | | | - Maura Faraci
- Istituto Giannina Gaslini, Children's Hospital Genoa, Italy
| | | | | |
Collapse
|
30
|
Labelling Conventions and Product Package Insert of Parenteral Polymyxins: Factors Causing Potential Medication Errors and Impeding Optimal Clinical Use. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1145:133-141. [DOI: 10.1007/978-3-030-16373-0_10] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
|
31
|
Scott A, Pottenger S, Timofte D, Moore M, Wright L, Kukavica-Ibrulj I, Jeukens J, Levesque RC, Freschi L, Pinchbeck GL, Schmidt VM, McEwan N, Radford AD, Fothergill JL. Reservoirs of resistance: polymyxin resistance in veterinary-associated companion animal isolates of Pseudomonas aeruginosa. Vet Rec 2019; 185:206. [PMID: 31239295 DOI: 10.1136/vr.105075] [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] [Received: 06/22/2018] [Revised: 05/16/2019] [Accepted: 05/27/2019] [Indexed: 11/03/2022]
Abstract
BACKGROUND Pseudomonas aeruginosa is an opportunistic pathogen and a major cause of infections. Widespread resistance in human infections are increasing the use of last resort antimicrobials such as polymyxins. However, these have been used for decades in veterinary medicine. Companion animals are an understudied source of antimicrobial resistant P. aeruginosa isolates. This study evaluated the susceptibility of P. aeruginosa veterinary isolates to polymyxins to determine whether the veterinary niche represents a potential reservoir of resistance genes for pathogenic bacteria in both animals and humans. METHODS AND RESULTS Clinical P. aeruginosa isolates (n=24) from UK companion animals were compared for antimicrobial susceptibility to a panel of human-associated isolates (n=37). Minimum inhibitory concentration (MIC) values for polymyxin B and colistin in the companion animals was significantly higher than in human isolates (P=0.033 and P=0.013, respectively). Genotyping revealed that the veterinary isolates were spread throughout the P. aeruginosa population, with shared array types from human infections such as keratitis and respiratory infections, suggesting the potential for zoonotic transmission. Whole genome sequencing revealed mutations in genes associated with polymyxin resistance and other antimicrobial resistance-related genes. CONCLUSION The high levels of resistance to polymyxin shown here, along with genetic similarities between some human and animal isolates, together suggest a need for sustained surveillance of this veterinary niche as a potential reservoir for resistant, clinically relevant bacteria in both animals and humans.
Collapse
Affiliation(s)
- Andrea Scott
- Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| | - Sian Pottenger
- Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| | - Dorina Timofte
- Institute of Veterinary Science, University of Liverpool, Neston, Wirral, UK
| | - Matthew Moore
- Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| | - Laura Wright
- Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| | | | | | | | | | - Gina L Pinchbeck
- Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| | - Vanessa M Schmidt
- Institute of Infection and Global Health, University of Liverpool, Liverpool, UK.,Institute of Veterinary Science, University of Liverpool, Neston, Wirral, UK
| | - Neil McEwan
- Institute of Veterinary Science, University of Liverpool, Neston, Wirral, UK
| | - Alan D Radford
- Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| | - Joanne L Fothergill
- Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| |
Collapse
|
32
|
Abstract
Pneumonia, including community-acquired bacterial pneumonia, hospital-acquired bacterial pneumonia, and ventilator-acquired bacterial pneumonia, carries unacceptably high morbidity and mortality. Despite advances in antimicrobial therapy, emergence of multidrug resistance and high rates of treatment failure have made optimization of antibiotic efficacy a priority. This review focuses on pharmacokinetic and pharmacodynamic approaches to antibacterial optimization within the lung environment and in the setting of critical illness. Strategies for including these approaches in drug development programs as well as clinical practice are described and reviewed.
Collapse
Affiliation(s)
- Ana Motos
- Center for Anti-Infective Research and Development, Hartford Hospital, 80 Seymour Street, Hartford, CT 06102, USA; Division of Animal Experimentation, Department of Pulmonary and Critical Care, Hospital Clinic, 170 Villarroel Street, Barcelona 08036, Spain
| | - James M Kidd
- Center for Anti-Infective Research and Development, Hartford Hospital, 80 Seymour Street, Hartford, CT 06102, USA
| | - David P Nicolau
- Center for Anti-Infective Research and Development, Hartford Hospital, 80 Seymour Street, Hartford, CT 06102, USA; Division of Infectious Diseases, Hartford Hospital, 80 Seymour Street, Hartford, CT 06102, USA.
| |
Collapse
|
33
|
Lima MR, Ferreira GF, Nunes Neto WR, Monteiro JDM, Santos ÁRC, Tavares PB, Denadai ÂML, Bomfim MRQ, dos Santos VL, Marques SG, de Souza Monteiro A. Evaluation of the interaction between polymyxin B and Pseudomonas aeruginosa biofilm and planktonic cells: reactive oxygen species induction and zeta potential. BMC Microbiol 2019; 19:115. [PMID: 31142260 PMCID: PMC6542102 DOI: 10.1186/s12866-019-1485-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 05/10/2019] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Although the most widely accepted mechanism of action for polymyxins is related to bacterial lysis via disruption, we hypothesized that this antimicrobial drug class could have other effects on Pseudomonas aeruginosa planktonic and sessile cells. Little is known regarding oxidative burst and zeta potential (ZP) data associated with the interaction between polymyxin B and P. aeruginosa cells. The present study evaluated endogenous reactive oxygen species (ROS) production and changes in the net charges of biofilm and planktonic cells in response to polymyxin B. RESULTS Polymyxin B induced concentration-dependent killing at all concentrations tested in planktonic and sessile cells from P. aeruginosa strains. Sublethal concentrations of polymyxin B induced oxidative burst. ROS production was higher in resistant planktonic cells than in biofilm cells but this was not observed for susceptible cells. Moreover, no net surface charge alterations were observed in planktonic cells from a susceptible strain treated with polymyxin B, but a significant increase of ZP was noted in planktonic cells from a resistant strain. CONCLUSION Oxidative burst generated by planktonic and sessile cells from P. aeruginosa strains against polymyxin B indicates that ROS may have an important role in the mechanism of action of this drug. ZP data revealed that electrostatic interactions of the cationic peptide with the anionic surface of the cells are strain-dependent. Therefore, we suggested that the intracellular effects of polymyxin B should be further investigated to understand polymyxin B-induced stress in P. aeruginosa.
Collapse
Affiliation(s)
- Marlucy Rodrigues Lima
- Faculdade de Ciências da Saúde, Universidade Vale do Rio Doce, Governador Valadares, MG Brazil
| | - Gabriella Freitas Ferreira
- Departamento de Farmácia, Programa Multicêntrico de Pós-Graduação em Bioquímica e Biologia Molecular, Universidade Federal de Juiz de Fora, UFJF, Campus Governador Valadares - MG. R. Manoel Byrro, 241 - Vila Bretas, Governador Valadares, MG 35032-620 Brazil
| | | | | | - Áquila Rodrigues Costa Santos
- Departamento de Farmácia, Programa Multicêntrico de Pós-Graduação em Bioquímica e Biologia Molecular, Universidade Federal de Juiz de Fora, UFJF, Campus Governador Valadares - MG. R. Manoel Byrro, 241 - Vila Bretas, Governador Valadares, MG 35032-620 Brazil
| | | | - Ângelo Márcio Leite Denadai
- Departamento de Farmácia, Programa Multicêntrico de Pós-Graduação em Bioquímica e Biologia Molecular, Universidade Federal de Juiz de Fora, UFJF, Campus Governador Valadares - MG. R. Manoel Byrro, 241 - Vila Bretas, Governador Valadares, MG 35032-620 Brazil
| | | | - Vera Lúcia dos Santos
- Departamento de Microbiologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG Brazil
| | - Sirlei Garcia Marques
- Hospital Universitário da Universidade Federal do Maranhão, São Luís, MA Brazil
- Laboratório Cedro, São Luís, MA Brazil
| | | |
Collapse
|
34
|
Tsala M, Vourli S, Georgiou PC, Pournaras S, Tsakris A, Daikos GL, Mouton JW, Meletiadis J. Exploring colistin pharmacodynamics against Klebsiella pneumoniae: a need to revise current susceptibility breakpoints. J Antimicrob Chemother 2019; 73:953-961. [PMID: 29377998 DOI: 10.1093/jac/dkx522] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 12/13/2017] [Indexed: 11/12/2022] Open
Abstract
Objectives Because the pharmacokinetic/pharmacodynamic (PK/PD) characteristics of colistin against Enterobacteriaceae are not well explored, we studied the activity of colistin against K. pneumoniae in an in vitro PK/PD model simulating different dosing regimens. Methods Three clinical isolates of K. pneumoniae with MICs of 0.5, 1 and 4 mg/L were tested in an in vitro PK/PD model following a dose-fractionation design over a period of 24 h. A high and low inoculum of 107 and 104 cfu/mL with and without a heteroresistant subpopulation, respectively, were used. PK/PD indices associated with colistin activity were explored and Monte Carlo analysis was performed in order to determine the PTA for achieving a bactericidal effect (2 log kill). Results The fAUC/MIC (R2 = 0.64-0.68) followed by fCmax/MIC (R2 = 0.55-0.63) best described colistin's 24 h log10 cfu/mL reduction for both low and high inocula. Dosing regimens with fCmax/MIC ≥6 were always associated with a bactericidal effect (P = 0.0025). However, at clinically achievable concentrations, usually below fCmax/MIC = 6, an fAUC/MIC ≥25 was more predictive of a bactericidal effect. Using a dosing regimen of 9 MU/day, the PTA for this pharmacodynamic target was 100%, 5%-70% and 0%, for isolates with MICs of ≤0.5, 1 and ≥2 mg/L, respectively. Dosing regimens that aim for a trough level of 1 mg/L achieve coverage of strains up to 0.5 mg/L (target trough/MIC = 2 mg/L). Conclusions Characterization of the pharmacodynamics of colistin against Enterobacteriaceae in an in vitro model of infection indicates that a revision of current susceptibility breakpoints is needed. Therapeutic drug monitoring of colistin to achieve pharmacodynamic targets in individual patients is highly recommended.
Collapse
Affiliation(s)
- Marilena Tsala
- Clinical Microbiology Laboratory, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Sophia Vourli
- Clinical Microbiology Laboratory, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiota-Christina Georgiou
- Clinical Microbiology Laboratory, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Spyros Pournaras
- Clinical Microbiology Laboratory, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece.,Department of Microbiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasios Tsakris
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - George L Daikos
- First Department of Propaedeutic Medicine, Laikon Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Johan W Mouton
- Department of Medical Microbiology and Infectious Diseases, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Joseph Meletiadis
- Clinical Microbiology Laboratory, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece.,Department of Medical Microbiology and Infectious Diseases, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
35
|
Diekjürgen D, Grainger DW. A murine ex vivo 3D kidney proximal tubule model predicts clinical drug-induced nephrotoxicity. Arch Toxicol 2019; 93:1349-1364. [PMID: 30863989 DOI: 10.1007/s00204-019-02430-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 03/05/2019] [Indexed: 12/18/2022]
Abstract
Drug attrition and clinical product withdrawals due to nephrotoxicity remain major challenges for pharmaceutical drug development pipelines. Currently, no reliable high-throughput in vitro screening models are available that provide reliable, predictive toxicology data for clinical nephrotoxicity. Drug screens to predict toxicity and pharmacology assessments are compromised by standard two-dimensional (2D) cell monoculture models. Here we extend a previously reported murine three-dimensional (3D) kidney-derived intact proximal tubule model to provide ex vivo drug toxicity data that reliably compare to clinical experiences and improve nephrotoxicity predictions over current 2D cell assays. Proximal tubule cytotoxicity was monitored by ATP depletion for 12 compounds (acarbose, acetylsalicylic acid, captopril, cimetidine, cidofovir, cisplatin, doxorubicin, gentamicin, polymyxin B, polymyxin B nonapeptide, probenecid and vancomycin) in 3D proximal tubule ex vivo assays. Drug concentration-response curves (1-1000 µM) and IC50, lowest effective concentration (LEC) and AUC values were compared to clinical therapeutic exposure levels (Cmax). The 100-fold Cmax threshold demonstrated best sensitivity (96.9%) and specificity (87.5%) for this assay, with high positive (93.9%) and negative (93.3%) predictive values for nephrotoxicity. IC50 values were superior to LEC. Results also support the model's capability to predict substrate-inhibitor/competitor interactions, yielding toxicity results similar to those reported in vivo. These 3D proximal tubule-based drug screens provide more reliable nephrotoxicity predictions, and more insight into complex mechanisms implicated in nephrotoxicity than current standard 2D cell assays. This new approach for rapid drug toxicity testing produces more reliable clinical comparisons than current 2D cell culture screening techniques.
Collapse
Affiliation(s)
- Dorina Diekjürgen
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, 84112-5820, USA
| | - David W Grainger
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, 84112-5820, USA. .,Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, 84112-5820, USA.
| |
Collapse
|
36
|
Gajdács M. The Concept of an Ideal Antibiotic: Implications for Drug Design. Molecules 2019; 24:E892. [PMID: 30832456 PMCID: PMC6429336 DOI: 10.3390/molecules24050892] [Citation(s) in RCA: 185] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 02/21/2019] [Accepted: 02/27/2019] [Indexed: 02/06/2023] Open
Abstract
The emergence and spread of antibiotic-resistant pathogens is a major public health issue, which requires global action of an intersectoral nature. Multidrug-resistant (MDR) pathogens-especially "ESKAPE" bacteria-can withstand lethal doses of antibiotics with various chemical structures and mechanisms of action. Pharmaceutical companies are increasingly turning away from participating in the development of new antibiotics, due to the regulatory environment and the financial risks. There is an urgent need for innovation in antibiotic research, as classical discovery platforms (e.g., mining soil Streptomycetes) are no longer viable options. In addition to discovery platforms, a concept of an ideal antibiotic should be postulated, to act as a blueprint for future drugs, and to aid researchers, pharmaceutical companies, and relevant stakeholders in selecting lead compounds. Based on 150 references, the aim of this review is to summarize current advances regarding the challenges of antibiotic drug discovery and the specific attributes of an ideal antibacterial drug (a prodrug or generally reactive compound with no specific target, broad-spectrum antibacterial activity, adequate penetration through the Gram-negative cell wall, activity in biofilms and in hard-to-treat infections, accumulation in macrophages, availability for oral administration, and for use in sensitive patient groups).
Collapse
Affiliation(s)
- Márió Gajdács
- Department of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, 6720 Szeged, Hungary.
| |
Collapse
|
37
|
|
38
|
Dupuy FG, Pagano I, Andenoro K, Peralta MF, Elhady Y, Heinrich F, Tristram-Nagle S. Selective Interaction of Colistin with Lipid Model Membranes. Biophys J 2019; 114:919-928. [PMID: 29490251 DOI: 10.1016/j.bpj.2017.12.027] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 12/19/2017] [Accepted: 12/21/2017] [Indexed: 11/24/2022] Open
Abstract
Although colistin's clinical use is limited due to its nephrotoxicity, colistin is considered to be an antibiotic of last resort because it is used to treat patients infected with multidrug-resistant bacteria. In an effort to provide molecular details about colistin's ability to kill Gram-negative (G(-)) but not Gram-positive (G(+)) bacteria, we investigated the biophysics of the interaction between colistin and lipid mixtures mimicking the cytoplasmic membrane of G(+), G(-) bacteria as well as eukaryotic cells. Two different models of the G(-) outer membrane (OM) were assayed: lipid A with two deoxy-manno-octulosonyl sugar residues, and Escherichia coli lipopolysaccharide mixed with dilaurylphosphatidylglycerol. We used circular dichroism and x-ray diffuse scattering at low and wide angle in stacked multilayered samples, and neutron reflectivity of single, tethered bilayers mixed with colistin. We found no differences in secondary structure when colistin was bound to G(-) versus G(+) membrane mimics, ruling out a protein conformational change as the cause of this difference. However, bending modulus KC perturbation was quite irregular for the G(-) inner membrane, where colistin produced a softening of the membranes at an intermediate lipid/peptide molar ratio but stiffening at lower and higher peptide concentrations, whereas in G(+) and eukaryotic mimics there was only a slight softening. Acyl chain order in G(-) was perturbed similarly to KC. In G(+), there was only a slight softening and disordering effect, whereas in OM mimics, there was a slight stiffening and ordering of both membranes with increasing colistin. X-ray and neutron reflectivity structural results reveal colistin partitions deepest to reach the hydrocarbon interior in G(-) membranes, but remains in the headgroup region in G(+), OM, and eukaryotic mimics. It is possible that domain formation is responsible for the erratic response of G(-) inner membranes to colistin and for its deeper penetration, which could increase membrane permeability.
Collapse
Affiliation(s)
- Fernando G Dupuy
- Biological Physics Group, Physics Department, Carnegie Mellon University, Pittsburgh, Pennsylvania; Instituto Superior de Investigaciones Biológicas (INSIBIO) CONICET-UNT and Instituto de Química Biológica "Dr Bernabé Bloj", Facultad de Bioquímica, Química y Farmacia, UNT, San Miguel de Tucumán, Argentina
| | - Isabella Pagano
- Biological Physics Group, Physics Department, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Kathryn Andenoro
- Biological Physics Group, Physics Department, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Maria F Peralta
- Biological Physics Group, Physics Department, Carnegie Mellon University, Pittsburgh, Pennsylvania; Instituto de Investigación Médica M y M Ferreyra, CONICET-National University of Córdoba, Córdoba, Argentina
| | - Yasmene Elhady
- Biological Physics Group, Physics Department, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Frank Heinrich
- Biological Physics Group, Physics Department, Carnegie Mellon University, Pittsburgh, Pennsylvania; National Institute of Standards and Technology Center for Neutron Research, Gaithersburg, Maryland
| | - Stephanie Tristram-Nagle
- Biological Physics Group, Physics Department, Carnegie Mellon University, Pittsburgh, Pennsylvania.
| |
Collapse
|
39
|
Mangal S, Huang J, Shetty N, Park H, Lin YW, Yu HH, Zemlyanov D, Velkov T, Li J, Zhou QT. Effects of the antibiotic component on in-vitro bacterial killing, physico-chemical properties, aerosolization and dissolution of a ternary-combinational inhalation powder formulation of antibiotics for pan-drug resistant Gram-negative lung infections. Int J Pharm 2019; 561:102-113. [PMID: 30797863 DOI: 10.1016/j.ijpharm.2019.02.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 02/02/2019] [Accepted: 02/08/2019] [Indexed: 12/12/2022]
Abstract
Combinational antibiotic formulations have emerged as an important strategy to combat antibiotic resistance. The main objective of this study was to examine effects of individual components on the antimicrobial activity, physico-chemical properties, aerosolization and dissolution of powder aerosol formulations when three synergistic drugs were co-spray dried. A ternary dry powder formulation consisting of meropenem (75.5 %w/w), colistin (15.1 %w/w) and rifampicin (9.4 %w/w) at the selected ratio was produced by spray drying. The ternary formulation was characterized for in-vitro antibacterial activity, physico-chemical properties, surface composition, aerosol performance and dissolution. All of the formulations demonstrated excellent aerosolization behavior achieving a fine particle fraction of >70%, which was substantially higher than those for the Meropenem-SD and Colistin-Meropenem formulations. The results indicated that rifampicin controlled the surface morphology of the ternary and binary combination formulations resulting in the formation of highly corrugated particles. Advanced characterization of surface composition by XPS supported the hypothesis that rifampicin was enriched on the surface of the combination powder formulations. All spray-dried formulations were amorphous and absorbed substantial amount of water at the elevated humidity. Storage at the elevated humidity caused a substantial decline in aerosolization performance for the Meropenem-SD and Colistin-Meropenem, which was attributed to increased inter-particulate capillary forces or particle fusion. In contrast, the ternary combination and binary Meropenem-Rifampicin formulations showed no change in aerosol performance at the elevated storage humidity conditions; attributable to the enriched hydrophobicity of rifampicin on the particle surface that acted as a barrier against moisture condensation and particle fusion. Interestingly, in the ternary formulation rifampicin enrichment on the surface did not interfere with the dissolution of other two components (i.e. meropenem and colistin). Our study provides an insight on the impact of each component on the performance of co-spray dried combinational formulations.
Collapse
Affiliation(s)
- Sharad Mangal
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Jiayang Huang
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Nivedita Shetty
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Heejun Park
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Yu-Wei Lin
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria 3800, Australia
| | - Heidi H Yu
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria 3800, Australia
| | - Dmitry Zemlyanov
- Birck Nanotechnology Center, Purdue University, West Lafayette, IN 47907, USA
| | - Tony Velkov
- Department of Pharmacology & Therapeutics, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jian Li
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria 3800, Australia
| | - Qi Tony Zhou
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA.
| |
Collapse
|
40
|
Rigatto MH, Falci DR, Zavascki AP. Clinical Use of Polymyxin B. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1145:197-218. [PMID: 31364080 DOI: 10.1007/978-3-030-16373-0_14] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Polymyxin B is another clinically available polymyxin that has re-emerged in clinical practice to treat infections caused by multi-drug (MDR) or extensively-drug-resistant (XDR) Gram-negative bacteria (GNB). Its chemical structure is very similar to the structure of polymyxin E (colistin). However, since the latter is administered as a prodrug, there are major pharmacokinetic differences between both polymyxins that may potentially determine different clinical and microbiological outcomes. Studies addressing clinical or microbiological outcomes in patients treated with polymyxin B for MDR or XDR GNB are reviewed in this chapter.
Collapse
Affiliation(s)
- Maria Helena Rigatto
- Infectious Diseases Service, Hospital São Lucas da Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
- Medical School, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Diego R Falci
- Post-Graduate Program in Health and Human Development, Universidade La Salle, Canoas, Brazil
- Infectious Diseases Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Alexandre P Zavascki
- Infectious Diseases Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.
- Department of Internal Medicine, School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| |
Collapse
|
41
|
Marchaim D, Kaye D, Kaye KS. Use of Colistin in Critically Ill Patients. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1145:155-179. [PMID: 31364078 DOI: 10.1007/978-3-030-16373-0_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Due to lack of better therapeutic options, colistin use for extensively drug-resistant Gram-negative organisms was revived in the past two decades, including in patients in intensive-care units (ICU). There are multiple knowledge gaps pertaining to the clinical use and utility of colistin in critically-ill patients, but due to lack of options, it is used in these high risk patients. In this chapter, we critically review the various topics pertaining to colistin use in critically-ill patients, while highlighting the (lack of) controlled evidence supporting common current practices pertaining to colistin use by clinicians.
Collapse
Affiliation(s)
- Dror Marchaim
- Unit of Infection Control, Shamir (Assaf Harofeh) Medical Center, Zerifin, Israel. .,Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| | - Donald Kaye
- Drexel University College of Medicine, Philadelphia, PA, USA
| | - Keith S Kaye
- Division of Infectious Diseases, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
42
|
Thomas R, Velaphi S, Ellis S, Walker AS, Standing JF, Heath P, Sharland M, Dona' D. The use of polymyxins to treat carbapenem resistant infections in neonates and children. Expert Opin Pharmacother 2018; 20:415-422. [PMID: 30576264 DOI: 10.1080/14656566.2018.1559817] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION The incidence of healthcare-associated multidrug resistant bacterial infections, particularly due to carbapenem resistant organisms, has been on the rise globally. Among these are the carbapenem resistant Acinetobacter baumannii and Enterobacteriaceae, which have been responsible for numerous outbreaks in neonatal units. The polymyxins (colistin and polymyxin B) are considered to be the last resort antibiotics for treating such infections. However, pharmacokinetic and pharmacodynamic data on the use of polymyxins in neonates and children are very limited, and there are safety concerns. AREAS COVERED In this review, the authors summarize the global burden of multidrug resistance, particularly carbapenem resistance, in the neonatal and paediatric population, and the potential wider use of polymyxins in treating these infections. EXPERT OPINION Both colistin and polymyxin B have similar efficacy in treating multidrug resistant infections but have safety concerns. However, polymyxin B appears to be a better therapeutic option, with more rapid and higher steady state concentrations achieved compared to colistin and less reported nephrotoxicity. There is virtually no data in neonates and children currently; there is therefore an urgent need for pharmacokinetic and safety trials in these populations to determine the optimal drug and dosing regimens and provide recommendations for their use against carbapenem resistant infections.
Collapse
Affiliation(s)
- Reenu Thomas
- a Department of Paediatrics, School of Clinical Medicine, Faculty of Health Sciences , University of the Witwatersrand , Johannesburg , South Africa
| | - Sithembiso Velaphi
- a Department of Paediatrics, School of Clinical Medicine, Faculty of Health Sciences , University of the Witwatersrand , Johannesburg , South Africa
| | - Sally Ellis
- b Global Antibiotic Research and Development Partnership (GARDP) , Drugs for Neglected Diseases initiative , Geneva , Switzerland
| | - A Sarah Walker
- c MRC Clinical Trials Unit at University College London , University College London , London , UK
| | - Joseph F Standing
- d Great Ormond Street Institute of Child Health , University College London , London , UK
| | - Paul Heath
- e Paediatric Infectious Disease Research Group, Institute for Infection and Immunity , St George's University of London , London , UK
| | - Mike Sharland
- e Paediatric Infectious Disease Research Group, Institute for Infection and Immunity , St George's University of London , London , UK
| | - Daniele Dona'
- e Paediatric Infectious Disease Research Group, Institute for Infection and Immunity , St George's University of London , London , UK
| |
Collapse
|
43
|
Li ZD, Luo J, Jia LH, Wang XY, Xun ZK, Liu M. Cytochrome C suppresses renal accumulation and nephrotoxicity of polymyxin B. Hum Exp Toxicol 2018; 38:193-200. [PMID: 30016892 DOI: 10.1177/0960327118783543] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The receptor megalin plays an important role in the accumulation of polymyxin B (PMB) in renal cells in vitro. This study aimed to examine the effects of cytochrome c (cyto c), a typical megalin ligand, on renal accumulation and nephrotoxicity of PMB in vivo. Thirty Sprague-Dawley rats were randomly divided into the vehicle control group, PMB group, PMB + cyto c 50, 100, or 200 mg/kg group, respectively, and were treated with intravenous cyto c 30 min before the administration of PMB 4.0 mg/kg once a day for consecutive 5 days. On the 4th day after administration, 24 h urine was collected to determine N-acetyl-β-D-glucosaminidase excretion. Six hours after the last injection on the 5th day, kidneys were harvested to assay PMB concentration and observe pathological alterations, and blood samples were collected to assay serum creatinine (SCr), blood urea nitrogen (BUN), and blood β2-microglobulin (β2-MG) levels. Cyto c 50, 100, and 200 mg/kg decreased the accumulation of PMB in the kidney by 18.5%, 39.1% ( p < 0.01), and 36.8% ( p < 0.01), respectively, and reduced 24 h N-acetyl-β-D- glucosaminidase excretion by 22.5% ( p < 0.05), 40.4% ( p < 0.01), and 40.4% ( p < 0.01), respectively. Kidney pathological damage induced by PMB was markedly reduced by cyto c 100 mg/kg and 200 mg/kg. However, there were no significant differences in SCr, BUN, and blood β2-MG levels among the groups. These results indicated that cyto c may inhibit the renal accumulation and nephrotoxicity of PMB in a rat model, further proving the role of megalin in the accumulation of PMB.
Collapse
Affiliation(s)
- Z-D Li
- 1 Department of Pharmacy, Electric Power Teaching Hospital, Capital Medical University, Beijing, China.,2 Department of Pharmacy, Clinical School of Air Force, Anhui Medical University, Hefei, China
| | - J Luo
- 2 Department of Pharmacy, Clinical School of Air Force, Anhui Medical University, Hefei, China
| | - L-H Jia
- 1 Department of Pharmacy, Electric Power Teaching Hospital, Capital Medical University, Beijing, China
| | - X-Y Wang
- 1 Department of Pharmacy, Electric Power Teaching Hospital, Capital Medical University, Beijing, China
| | - Z-K Xun
- 1 Department of Pharmacy, Electric Power Teaching Hospital, Capital Medical University, Beijing, China
| | - M Liu
- 3 Department of pharmacy, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
44
|
Borowiak M, Fischer J, Hammerl JA, Hendriksen RS, Szabo I, Malorny B. Identification of a novel transposon-associated phosphoethanolamine transferase gene, mcr-5, conferring colistin resistance in d-tartrate fermenting Salmonella enterica subsp. enterica serovar Paratyphi B. J Antimicrob Chemother 2018; 72:3317-3324. [PMID: 28962028 DOI: 10.1093/jac/dkx327] [Citation(s) in RCA: 372] [Impact Index Per Article: 53.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 08/04/2017] [Indexed: 01/17/2023] Open
Abstract
Objectives Plasmid-mediated mobilized colistin resistance is currently known to be caused by phosphoethanolamine transferases termed MCR-1, MCR-2, MCR-3 and MCR-4. However, this study focuses on the dissection of a novel resistance mechanism in mcr-1-, mcr-2- and mcr-3-negative d-tartrate fermenting Salmonella enterica subsp. enterica serovar Paratyphi B (Salmonella Paratyphi B dTa+) isolates with colistin MIC values >2 mg/L. Methods A selected isolate from the strain collection of the German National Reference Laboratory for Salmonella was investigated by WGS and bioinformatical analysis to identify novel phosphoethanolamine transferase genes involved in colistin resistance. Subsequently PCR screening, S1-PFGE and DNA-DNA hybridization were performed to analyse the prevalence and location of the identified mcr-5 gene. Cloning and transformation experiments in Escherichia coli DH5α and Salmonella Paratyphi B dTa+ control strains were carried out and the activity of MCR-5 was determined in vitro by MIC testing. Results In this study, we identified a novel phosphoethanolamine transferase in 14 mcr-1-, mcr-2- and mcr-3-negative Salmonella Paratyphi B dTa+ isolates with colistin MIC values >2 mg/L that were received during 2011-13. The respective gene, further termed as mcr-5 (1644 bp), is part of a 7337 bp transposon of the Tn3 family and usually located on related multi-copy ColE-type plasmids. Interestingly, in one isolate an additional subclone with a chromosomal location of the mcr-5 transposon was observed. Conclusions Our findings suggest that the transfer of colistin-resistance-mediating phosphoethanolamine transferase genes from bacterial chromosomes to mobile genetic elements has occurred in multiple independent events raising concern regarding their variety, prevalence and impact on public health.
Collapse
Affiliation(s)
- Maria Borowiak
- German Federal Institute for Risk Assessment, BfR, Department for Biological Safety, Berlin, Germany
| | - Jennie Fischer
- German Federal Institute for Risk Assessment, BfR, Department for Biological Safety, Berlin, Germany
| | - Jens A Hammerl
- German Federal Institute for Risk Assessment, BfR, Department for Biological Safety, Berlin, Germany
| | - Rene S Hendriksen
- National Food Institute, Technical University of Denmark, WHO Collaborating Center for Antimicrobial Resistance in Foodborne Pathogens and European Union Reference Laboratory for Antimicrobial Resistance, Kgs Lyngby, Denmark
| | - Istvan Szabo
- German Federal Institute for Risk Assessment, BfR, Department for Biological Safety, Berlin, Germany
| | - Burkhard Malorny
- German Federal Institute for Risk Assessment, BfR, Department for Biological Safety, Berlin, Germany
| |
Collapse
|
45
|
Sarıca S, Yurttutan S. An evaluation of hearing in infants administered with colistin in the premature neonatal intensive care unit. J Matern Fetal Neonatal Med 2018; 31:2918-2922. [PMID: 29779419 DOI: 10.1080/14767058.2018.1479388] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
AIM The aim of this study was to investigate the effects of colistin on hearing by evaluating the otoacoustic emission tests and clinical auditory brainstem responses in preterm infants treated with colistin in the neonatal intensive care unit. METHOD The study included 30 neonates (male: n = 16, female: n = 14) born before 37 weeks who were admitted to the Neonatal Intensive Care Unit at the Kahramanmaras Sutcu Imam Medical Faculty between January 2014 and January 2015 and who were treated with colistin during their time in intensive care because of infection. A control group was formed consisting of 30 preterm infants (male: n = 18, female: n = 12) with no additional disease born in the hospital during the same period. Following an ear, nose and throat examination the distortion product otoacoustic emission test, transient evoked otoacoustic emission and clinical auditory brainstem response tests were applied to all 60 patients. RESULTS The otoacoustic emission responses obtained from the control group were positive and clinical auditory brainstem responses up to 15 dB were obtained. In the colistin group negative otoacoustic emission responses were obtained in two patients unilaterally and in one patient bilaterally and loss was observed at the thresholds in the clinical auditory brainstem response test. Significantly prolonged fifth wave latency was observed in the colistin group compared to the control group for the clinical auditory brainstem response at 15 dB. CONCLUSIONS Given that the study results showed unilateral hearing loss in two patients and bilateral hearing loss in one as well as latency at 15 dB, hearing tests to check for ototoxicity are recommended for patients given colistin.
Collapse
Affiliation(s)
- Selman Sarıca
- a Kahramanmaras Sutcu Imam Universitesi Tip Fakultesi , Kahramanmaras , Turkey
| | - Sadık Yurttutan
- a Kahramanmaras Sutcu Imam Universitesi Tip Fakultesi , Kahramanmaras , Turkey
| |
Collapse
|
46
|
Porreca AM, Sullivan KV, Gallagher JC. The Epidemiology, Evolution, and Treatment of KPC-Producing Organisms. Curr Infect Dis Rep 2018; 20:13. [PMID: 29730830 DOI: 10.1007/s11908-018-0617-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
PURPOSE OF REVIEW The purpose of this review is to investigate the evolution and epidemiology of Klebsiella pneumoniae carbapenemase (KPC)-producing organisms and the current and future treatment options for infections caused by KPC-producing isolates. RECENT FINDINGS The emergence of resistance in Enterobacteriaceae producing carbapenemases globally has increased the challenges in treating infections caused by these organisms. One of the prominent mechanisms of resistance is the production of KPC enzymes. Infections caused by organisms producing KPCs have limited treatment options and are associated with poor clinical outcomes. The rapid rise of KPC-producing organisms necessitated the use of drugs with pharmacokinetic and toxicity limitations, including polymyxins, tigecycline, fosfomycin, and aminoglycosides. The availability of new beta-lactamase inhibitor combinations that are effective against KPC-producing organisms represent an advance in safety and efficacy. Several agents are currently being studied that have activity against KPC-producing organisms and appear to represent promising additions to our armamentarium. KPC-producing organisms cause infections with high morbidity and mortality. Limited treatment options are available, though new therapies have been developed. Pipeline agents are likely to have a place in therapy for the treatment of infections caused by KPC-producing isolates.
Collapse
Affiliation(s)
- Ann Marie Porreca
- Temple University School of Pharmacy, 3307 North Broad Street, Philadelphia, PA, USA
| | - Kaede V Sullivan
- Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Jason C Gallagher
- Temple University School of Pharmacy, 3307 North Broad Street, Philadelphia, PA, USA.
| |
Collapse
|
47
|
Pharmacokinetics of colistin methanesulfonate (CMS) in healthy Chinese subjects after single and multiple intravenous doses. Int J Antimicrob Agents 2018; 51:714-720. [DOI: 10.1016/j.ijantimicag.2017.12.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 12/17/2017] [Accepted: 12/24/2017] [Indexed: 12/13/2022]
|
48
|
Richter SS, Karichu J, Otiso J, Van Heule H, Keller G, Cober E, Rojas LJ, Hujer AM, Hujer KM, Marshall S, Perez F, Rudin SD, Domitrovic TN, Kaye KS, Salata R, van Duin D, Bonomo RA. Evaluation of Sensititre Broth Microdilution Plate for determining the susceptibility of carbapenem-resistant Klebsiella pneumoniae to polymyxins. Diagn Microbiol Infect Dis 2018; 91:89-92. [PMID: 29456071 DOI: 10.1016/j.diagmicrobio.2018.01.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 12/20/2017] [Accepted: 01/03/2018] [Indexed: 10/18/2022]
Abstract
Colistin and polymyxin B MICs were determined for 106 carbapenem-resistant Klebsiella pneumoniae (CR-Kp) isolates using Sensititre Research Use Only GNX2F plates (Thermo Fisher) and compared to CLSI broth macrodilution (BMD) as the reference method. For colistin, EUCAST breakpoints were applied and testing of isolates with very major (VM) errors was repeated in duplicate by both methods to determine a majority result. Essential agreement (MIC ± one dilution) of GNX2F with the reference method was 97.1% for polymyxin B and 92.5% for colistin (7 VM errors, 22.6%). After discrepancy testing, there were 28 colistin resistant isolates by BMD and essential agreement was 94.3% with 4 VM errors (14.3%). Colistin and polymyxin B GNX2F results showed acceptable essential agreement with BMD for MICS without interpretation. Colistin VM errors with EUCAST breakpoints were due to MIC variability in the 2 to 4 μg/mL range that could be addressed by establishing an intermediate category.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Laura J Rojas
- Department of Veteran Affairs Medical Center, Cleveland, OH,; Department of Molecular Biology and Microbiology, Case Western Reserve Univ. School of Medicine, Cleveland, OH
| | - Andrea M Hujer
- Department of Veteran Affairs Medical Center, Cleveland, OH,; Case-VA CARES, Case Western Reserve Univ., Cleveland, OH
| | - Kristine M Hujer
- Department of Veteran Affairs Medical Center, Cleveland, OH,; Department of Medicine, Case Western Reserve Univ. School of Medicine, Cleveland, OH
| | - Steve Marshall
- Department of Veteran Affairs Medical Center, Cleveland, OH
| | - Frederico Perez
- Department of Veteran Affairs Medical Center, Cleveland, OH,; Case-VA CARES, Case Western Reserve Univ., Cleveland, OH,; Department of Medicine, Case Western Reserve Univ. School of Medicine, Cleveland, OH
| | - Susan D Rudin
- Department of Veteran Affairs Medical Center, Cleveland, OH,; Department of Medicine, Case Western Reserve Univ. School of Medicine, Cleveland, OH
| | - T Nicholas Domitrovic
- Department of Veteran Affairs Medical Center, Cleveland, OH,; Department of Medicine, Case Western Reserve Univ. School of Medicine, Cleveland, OH
| | | | - Robert Salata
- Case-VA CARES, Case Western Reserve Univ., Cleveland, OH
| | | | - Robert A Bonomo
- Department of Veteran Affairs Medical Center, Cleveland, OH,; Case-VA CARES, Case Western Reserve Univ., Cleveland, OH,; Department of Medicine, Case Western Reserve Univ. School of Medicine, Cleveland, OH; Department of Molecular Biology and Microbiology, Case Western Reserve Univ. School of Medicine, Cleveland, OH; Department of Biochemistry, Case Western Reserve Univ. School of Medicine, Cleveland, OH; Department of Pharmacology, Case Western Reserve Univ. School of Medicine, Cleveland, OH; Department of Proteomics and Bioinformatics, Case Western Reserve Univ. School of Medicine, Cleveland, OH
| |
Collapse
|
49
|
Polymyxins: Antibacterial Activity, Susceptibility Testing, and Resistance Mechanisms Encoded by Plasmids or Chromosomes. Clin Microbiol Rev 2017; 30:557-596. [PMID: 28275006 DOI: 10.1128/cmr.00064-16] [Citation(s) in RCA: 994] [Impact Index Per Article: 124.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Polymyxins are well-established antibiotics that have recently regained significant interest as a consequence of the increasing incidence of infections due to multidrug-resistant Gram-negative bacteria. Colistin and polymyxin B are being seriously reconsidered as last-resort antibiotics in many areas where multidrug resistance is observed in clinical medicine. In parallel, the heavy use of polymyxins in veterinary medicine is currently being reconsidered due to increased reports of polymyxin-resistant bacteria. Susceptibility testing is challenging with polymyxins, and currently available techniques are presented here. Genotypic and phenotypic methods that provide relevant information for diagnostic laboratories are presented. This review also presents recent works in relation to recently identified mechanisms of polymyxin resistance, including chromosomally encoded resistance traits as well as the recently identified plasmid-encoded polymyxin resistance determinant MCR-1. Epidemiological features summarizing the current knowledge in that field are presented.
Collapse
|
50
|
Gustinetti G, Cangemi G, Bandettini R, Castagnola E. Pharmacokinetic/pharmacodynamic parameters for treatment optimization of infection due to antibiotic resistant bacteria: a summary for practical purposes in children and adults. J Chemother 2017; 30:65-81. [PMID: 29025364 DOI: 10.1080/1120009x.2017.1377909] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
In the last years, there has been a tremendous increase in the incidence of bacterial infections due to resistant strains, especially multi-drug resistant Gram-negative bacilli. In Europe, a north to south and a west to east gradient was noticed, with more than one third of the K. pneumonia isolates being resistant to carbapenems in few countries. New antibiotics are lacking and, as a consequence, pharmacokinetic/pharmacodynamic parameters, normalized to pathogen minimal inhibitory concentration, are used with increased frequency to treat infections due to difficult-to-treat pathogens. These parameters are available at least for the adult population, but sparse in many different publications. This review wants to provide a comprehensive and 'easy to read' text for everyday practice, briefly summarizing the presently available knowledge on pharmacokinetic/pharmacodynamic parameters (normalized for minimal inhibitory concentration values) of different class drugs, that can be applied for an effective antibacterial treatment infections due to antibiotic-resistant pathogens.
Collapse
Affiliation(s)
- Giulia Gustinetti
- a Department of Infectious Diseases , University of Genoa , Genoa , Italy
| | - Giuliana Cangemi
- b Istituto Giannina Gaslini, Children's Hospital , Genoa , Italy
| | | | - Elio Castagnola
- b Istituto Giannina Gaslini, Children's Hospital , Genoa , Italy
| |
Collapse
|