1
|
Wu M, Feng K, Wu X, Liu C, Zhu S, Martins FS, Yu M, Lv Z, Yan M, Sy SKB. Prediction of tissue exposures of polymyxin-B, amikacin and sulbactam using physiologically-based pharmacokinetic modeling. Front Microbiol 2024; 15:1435906. [PMID: 39435440 PMCID: PMC11491386 DOI: 10.3389/fmicb.2024.1435906] [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: 05/21/2024] [Accepted: 09/19/2024] [Indexed: 10/23/2024] Open
Abstract
Background The combination antimicrobial therapy consisting of amikacin, polymyxin-B, and sulbactam demonstrated in vitro synergy against multi-drug resistant Acinetobacter baumannii. Objectives The objectives were to predict drug disposition and extrapolate their efficacy in the blood, lung, heart, muscle and skin tissues using a physiologically-based pharmacokinetic (PBPK) modeling approach and to evaluate achievement of target pharmacodynamic (PD) indices against A. baumannii. Methods A PBPK model was initially developed for amikacin, polymyxin-B, and sulbactam in adult subjects, and then scaled to pediatrics, accounting for both renal and non-renal clearances. The simulated plasma and tissue drug exposures were compared to the observed data from humans and rats. Efficacy was inferred using joint probability of target attainment of target PD indices. Results The simulated plasma drug exposures in adults and pediatrics were within the 0.5 to 2 boundary of the mean fold error for the ratio between simulated and observed means. Simulated drug exposures in blood, skin, lung, and heart were consistent with reported penetration ratio between tissue and plasma drug exposure. In a virtual pediatric population from 2 to <18 years of age using pediatric dosing regimens, the interpretive breakpoints were achieved in 85-90% of the population. Conclusion The utility of PBPK to predict and simulate the amount of antibacterial drug exposure in tissue is a practical approach to overcome the difficulty of obtaining tissue drug concentrations in pediatric population. As combination therapy, amikacin/polymyxin-B/sulbactam drug concentrations in the tissues exhibited sufficient penetration to combat extremely drug resistant A. baumannii clinical isolates.
Collapse
Affiliation(s)
- Mengyuan Wu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Kun Feng
- Women and Children Hospital, Qingdao University, Qingdao, China
| | - Xiao Wu
- Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Chang Liu
- Women and Children Hospital, Qingdao University, Qingdao, China
| | - Shixing Zhu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Frederico S. Martins
- Department of Clinical and Toxicological Analysis, Faculty of Pharmaceutical Sciences, Universidade de São Paulo, São Paulo, Brazil
| | - Mingming Yu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Zhihua Lv
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Meixing Yan
- Women and Children Hospital, Qingdao University, Qingdao, China
| | - Sherwin K. B. Sy
- Department of Statistics, Universidade Estadual de Maringá, Maringá, Paraná, Brazil
| |
Collapse
|
2
|
Deshpande D, Magombedze G, Boorgula GD, Chapagain M, Srivastava S, Gumbo T. Ceftriaxone Efficacy for Mycobacterium avium Complex Lung Disease in the Hollow Fiber and Translation to Sustained Sputum Culture Conversion in Patients. J Infect Dis 2024; 230:e230-e240. [PMID: 38036299 PMCID: PMC11326821 DOI: 10.1093/infdis/jiad545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/17/2023] [Accepted: 11/28/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND Only 35.6%-50.8% of patients with Mycobacterium avium complex (MAC) pulmonary disease achieve sustained sputum culture conversion (SSCC) on treatment with the azithromycin-ethambutol-rifabutin standard of care (SOC). We tested the efficacy of ceftriaxone, a β-lactam with a lung-to-serum penetration ratio of 12.18-fold. METHODS We mimicked lung concentration-time profiles of 7 ceftriaxone once-daily doses for 28 days in the hollow fiber system model of intracellular MAC (HFS-MAC). Monte Carlo experiments were used for dose selection. We also compared once-daily ceftriaxone monotherapy to 3-drug SOC against 5 MAC clinical isolates in HFS-MAC using γ (kill) slopes, and translated to SSCC rates. RESULTS Ceftriaxone killed 1.02-3.82 log10 colony-forming units (CFU)/mL, at optimal dose of 2 g once-daily. Ceftriaxone killed all 5 strains below day 0 versus 2 of 5 for SOC. The median γ (95% confidence interval [CI]) was 0.49 (.47-.52) log10 CFU/mL/day for ceftriaxone and 0.38 (.34-.43) log10 CFU/mL/day for SOC. In patients, the SOC was predicted to achieve SSCC rates (CI) of 39.3% (36%-42%) at 6 months. The SOC SSCC was 50% at 8.18 (3.64-27.66) months versus 3.58 (2.20-7.23) months for ceftriaxone, shortening time to SSCC 2.35-fold. CONCLUSIONS Ceftriaxone is a promising agent for creation of short-course chemotherapy.
Collapse
Affiliation(s)
| | | | | | - Moti Chapagain
- Department of Cellular and Molecular Biology, School of Medicine, University of Texas Health Science Center at Tyler
| | - Shashikant Srivastava
- Baylor University Medical Center, Dallas
- Department of Medicine, School of Medicine, University of Texas at Tyler
- Department of Cellular and Molecular Biology, School of Medicine, University of Texas Health Science Center at Tyler
| | - Tawanda Gumbo
- Mathematical Modeling and AI Department, Praedicare Inc, Dallas
- Hollow Fiber System and Experimental Therapeutics Laboratories, Praedicare Inc, Dallas, Texas
| |
Collapse
|
3
|
Tessier B, Moine L, Peramo A, Tsapis N, Fattal E. Poly(malic acid)-budesonide nanoconjugates embedded in microparticles for lung administration. Drug Deliv Transl Res 2024; 14:2062-2078. [PMID: 38517568 DOI: 10.1007/s13346-024-01571-4] [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] [Accepted: 03/04/2024] [Indexed: 03/24/2024]
Abstract
To improve the therapeutic activity of inhaled glucocorticoids and reduce potential side effects, we designed a formulation combining the advantages of nanoparticles, which have an enhanced uptake by alveolar cells, allow targeted delivery and sustained drug release, as well as limited drug systemic passage, with those of microparticles, which display good alveolar deposition. Herein, a polymer-drug conjugate, poly(malic acid)-budesonide (PMAB), was first synthesized with either 11, 20, 33, or 43 mol% budesonide (drug:polymer from 1:8 to 3:4), the drug creating hydrophobic domains. The obtained conjugates self-assemble into nanoconjugates in water, yielding excellent drug loading of up to 73 wt%, with 80-100 nm diameters. In vitro assays showed that budesonide could be steadily released from the nanoconjugates, and the anti-inflammatory activity was preserved, as evidenced by reduced cytokine production in LPS-activated RAW 264.7 macrophages. Nanoconjugates were then embedded into microparticles through spray-drying with L-leucine, forming nano-embedded microparticles (NEMs). NEMs were produced with an aerodynamic diameter close to 1 µm and a density below 0.1 g.cm-3, indicative of a high alveolar deposition. NEMs spray-dried with the less hydrophobic nanoconjugates, PMAB 1:4, were readily dissolved in simulated lung fluid and were chosen for in vivo experiments to study pharmacokinetics in healthy rats. As it was released in vivo from NEMs, sustained distribution of budesonide was obtained for 48 h in lung tissue, cells, and lining fluid. With high loading rates, modulable release kinetics, and low cytotoxicity, these nanoconjugates delivered by NEMs are promising for the more efficient treatment of pulmonary inflammatory diseases.
Collapse
Affiliation(s)
- Barbara Tessier
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France
| | - Laurence Moine
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France
| | - Arnaud Peramo
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France
| | - Nicolas Tsapis
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France
| | - Elias Fattal
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France.
| |
Collapse
|
4
|
Geilen J, Kainz M, Zapletal B, Schweiger T, Jäger W, Maier-Salamon A, Zeitlinger M, Stamm T, Ritschl V, Geleff S, Schultz MJ, Tschernko E. Effects of lung inflammation and injury on pulmonary tissue penetration of meropenem and vancomycin in a model of unilateral lung injury. Int J Antimicrob Agents 2024; 64:107180. [PMID: 38649034 DOI: 10.1016/j.ijantimicag.2024.107180] [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: 01/16/2024] [Revised: 03/23/2024] [Accepted: 04/18/2024] [Indexed: 04/25/2024]
Abstract
OBJECTIVE The timing and dosing of antimicrobial therapy are key in the treatment of pneumonia in critically ill patients. It is uncertain whether the presence of lung inflammation and injury affects tissue penetration of intravenously administered antimicrobial drugs. The effects of lung inflammation and injury on tissue penetration of two antimicrobial drugs commonly used for pneumonia were determined in an established model of unilateral lung injury. METHODS Unilateral lung injury was induced in the left lung of 13 healthy pigs through cyclic rinsing; the right healthy lung served as control. Infusions of meropenem and vancomycin were administered and concentrations of these drugs in lung tissue, blood, and epithelial lining fluid (ELF) were compared over a period of 6 h. RESULTS Median vancomycin lung tissue concentrations and penetration ratio were higher in inflamed and injured lungs compared with uninflamed and uninjured lungs (AUC0-6h: P = 0.003 and AUCdialysate/AUCplasma ratio: P = 0.003), resulting in higher AUC0-24/MIC. Median meropenem lung tissue concentrations and penetration ratio in inflamed and injured lungs did not differ from that in uninflamed and uninjured lungs (AUC0-6: P = 0.094 and AUCdialysate/AUCplasma ratio: P = 0.173). The penetration ratio for both vancomycin and meropenem into ELF was similar in injured and uninjured lungs. CONCLUSION Vancomycin penetration into lung tissue is enhanced by acute inflammation and injury, a phenomenon barely evident with meropenem. Therefore, inflammation in lung tissue influences the penetration into interstitial lung tissue, depending on the chosen antimicrobial drug. Measurement of ELF levels alone might not identify the impact of inflammation and injury.
Collapse
Affiliation(s)
- Johannes Geilen
- Department of Anaesthesia, General Intensive Care and Pain Management, Division of Cardiothoracic and Vascular Anaesthesia and Intensive Care Medicine, Medical University of Vienna, Vienna, Austria
| | - Matthias Kainz
- Department of Anaesthesia, General Intensive Care and Pain Management, Division of Cardiothoracic and Vascular Anaesthesia and Intensive Care Medicine, Medical University of Vienna, Vienna, Austria
| | - Bernhard Zapletal
- Department of Anaesthesia, General Intensive Care and Pain Management, Division of Cardiothoracic and Vascular Anaesthesia and Intensive Care Medicine, Medical University of Vienna, Vienna, Austria
| | - Thomas Schweiger
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Walter Jäger
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | | | - Markus Zeitlinger
- Department of Clinical Pharmacology, Clinical Pharmacokinetics/Pharmacogenetics and Imaging, Medical University of Vienna, Vienna, Austria
| | - Tanja Stamm
- Institute of Outcomes Research, Centre for Medical Data Science, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna, Austria
| | - Valentin Ritschl
- Institute of Outcomes Research, Centre for Medical Data Science, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna, Austria
| | - Silvana Geleff
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Marcus J Schultz
- Department of Anaesthesia, General Intensive Care and Pain Management, Division of Cardiothoracic and Vascular Anaesthesia and Intensive Care Medicine, Medical University of Vienna, Vienna, Austria; Department of Intensive Care and Laboratory of Experimental Intensive Care and Anaesthesiology (L·E·I·C·A), Amsterdam University Medical Centres, location 'AMC', Amsterdam, The Netherlands
| | - Edda Tschernko
- Department of Anaesthesia, General Intensive Care and Pain Management, Division of Cardiothoracic and Vascular Anaesthesia and Intensive Care Medicine, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
5
|
Rebholz D, Liebchen U, Paal M, Vogeser M, Starp J, Gräfe C, Brozat CI, Happich FL, Habler K, Scharf C. Can linezolid be validly measured in endotracheal aspiration in critically ill patients? A proof-of-concept trial. Intensive Care Med Exp 2024; 12:47. [PMID: 38717627 PMCID: PMC11078899 DOI: 10.1186/s40635-024-00630-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/03/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Therapeutic drug monitoring (TDM) of anti-infectives such as linezolid is routinely performed in blood of intensive care unit (ICU) patients to optimize target attainment. However, the concentration at the site of infection is considered more important for a successful therapy. Until now, bronchoalveolar lavage (BAL) is the gold standard to measure intrapulmonary concentrations of anti-infective agents. However, it is an invasive method and unsuitable for regular TDM. The aim of this proof-of-concept study was to investigate whether it is possible to reliably determine the intrapulmonary concentration of linezolid from endotracheal aspiration (ENTA). METHODS Intubated ICU patients receiving 600 mg intravenous linezolid twice daily were examined in steady state. First, preliminary experiments were performed in six patients to investigate which patients are suitable for linezolid measurement in ENTA. In a second step, trough and peak linezolid concentrations of plasma and ENTA were determined in nine suitable patients. RESULTS Linezolid can validly be detected in ENTA with viscous texture and > 0.5 mL volume. The mean (SD) linezolid trough concentration was 2.02 (1.27) mg/L in plasma and 1.60 (1.36) mg/L in ENTA, resulting in a median lung penetration rate of 104%. The mean (SD) peak concentration in plasma and ENTA was 10.77 (5.93) and 4.74 (2.66) mg/L. CONCLUSIONS Linezolid can validly be determined in ENTA with an adequate texture and volume. The penetration rate is comparable to already published BAL concentrations. This method might offer a simple and non-invasive method for TDM at the site of infection "lung". Due to promising results of the feasibility study, comparison of ENTA and BAL in the same patient should be investigated in a further trial.
Collapse
Affiliation(s)
- Diana Rebholz
- Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Uwe Liebchen
- Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Michael Paal
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Michael Vogeser
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Johannes Starp
- Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Caroline Gräfe
- Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Clara I Brozat
- Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Felix L Happich
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Katharina Habler
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Christina Scharf
- Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
6
|
Day NJ, Santucci P, Gutierrez MG. Host cell environments and antibiotic efficacy in tuberculosis. Trends Microbiol 2024; 32:270-279. [PMID: 37709598 DOI: 10.1016/j.tim.2023.08.009] [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: 05/08/2023] [Revised: 08/18/2023] [Accepted: 08/23/2023] [Indexed: 09/16/2023]
Abstract
The aetiologic agent of tuberculosis (TB), Mycobacterium tuberculosis (Mtb), can survive, persist, and proliferate in a variety of heterogeneous subcellular compartments. Therefore, TB chemotherapy requires antibiotics crossing multiple biological membranes to reach distinct subcellular compartments and target these bacterial populations. These compartments are also dynamic, and our understanding of intracellular pharmacokinetics (PK) often represents a challenge for antitubercular drug development. In recent years, the development of high-resolution imaging approaches in the context of host-pathogen interactions has revealed the intracellular distribution of antibiotics at a new level, yielding discoveries with important clinical implications. In this review, we describe the current knowledge regarding cellular PK of antibiotics and the complexity of drug distribution within the context of TB. We also discuss the recent advances in quantitative imaging and highlight their applications for drug development in the context of how intracellular environments and microbial localisation affect TB treatment efficacy.
Collapse
Affiliation(s)
- Nathan J Day
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Pierre Santucci
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Maximiliano G Gutierrez
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.
| |
Collapse
|
7
|
Lock GDA, Helfer VE, Dias BB, Torres BGS, De Araújo BV, Dalla Costa T. Population pharmacokinetic modeling of the influence of chronic and acute biofilm-forming Pseudomonas aeruginosa lung infection on ciprofloxacin free pulmonary and epithelial lining fluid concentrations. Eur J Pharm Sci 2023; 189:106546. [PMID: 37517670 DOI: 10.1016/j.ejps.2023.106546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 08/01/2023]
Abstract
We previously reported that ciprofloxacin (CIP) free lung interstitial concentrations are decreased by biofilm-forming Pseudomonas aeruginosa pulmonary chronic (14 d) infection. To get a better understanding on the influence of infection on CIP lung distribution, in the present study free lung interstitial fluid and epithelial lining fluid (ELF) concentrations were determined by microdialysis in biofilm-forming P. aeruginosa acutely (2 d) and chronically infected (14 d) Wistar rats following CIP 20 mg/kg i.v. bolus dosing. A popPK model was developed, using NONMEM® (version 7.4.3) with FOCE+I, with plasma data described as a three-compartment model with first-order elimination. For lung data inclusion, the model was expanded to four compartments and ELF concentrations were described as a fraction of lung levels estimated as a distribution factor (ƒD). Acute infection had a minor impact on plasma and lung CIP distribution and both infection stages did not alter ELF drug penetration. Probability of target attainment of ƒAUC0-24/MIC ≥ 90 using 20 mg q8h, equivalent to 400 mg q8h in humans, showed that CIP free concentrations in plasma are adequate to successfully treat lung infections. However, lung and ELF free interstitial concentrations might be insufficient to result in efficacious treatment of biofilm-forming P. aeruginosa chronic infection. However, lung and ELF free interstitial concentrations might be insufficient to result in efficacious treatment of biofilm-forming P. aeruginosa chronic infection.
Collapse
Affiliation(s)
- Graziela De Araujo Lock
- Pharmacokinetics and PK/PD Modeling Laboratory, Pharmaceutical Sciences Graduate Program, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Victória Etges Helfer
- Pharmacokinetics and PK/PD Modeling Laboratory, Pharmaceutical Sciences Graduate Program, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Bruna Bernar Dias
- Pharmacokinetics and PK/PD Modeling Laboratory, Pharmaceutical Sciences Graduate Program, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Bruna Gaelzer Silva Torres
- Pharmacokinetics and PK/PD Modeling Laboratory, Pharmaceutical Sciences Graduate Program, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Bibiana Verlindo De Araújo
- Pharmacokinetics and PK/PD Modeling Laboratory, Pharmaceutical Sciences Graduate Program, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Teresa Dalla Costa
- Pharmacokinetics and PK/PD Modeling Laboratory, Pharmaceutical Sciences Graduate Program, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
8
|
Alsmadi MM. Evaluating the Pharmacokinetics of Fentanyl in the Brain Extracellular Fluid, Saliva, Urine, and Plasma of Newborns from Transplacental Exposure from Parturient Mothers Dosed with Epidural Fentanyl Utilizing PBPK Modeling. Eur J Drug Metab Pharmacokinet 2023; 48:567-586. [PMID: 37563443 DOI: 10.1007/s13318-023-00842-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND AND OBJECTIVE Fentanyl can mitigate the mother and newborn complications resulting from labor pain. However, fentanyl shows a narrow therapeutic index between its respiratory depressive and analgesic effects. Thus, prenatally acquired high fentanyl levels in the newborn brain extracellular fluid (bECF) may induce respiratory depression which requires therapeutic drug monitoring (TDM). TDM using saliva and urine in newborns can reduce the possibility of infections and distress associated with TDM using blood. The objective of this study was to develop a physiologically based pharmacokinetic (PBPK) model to predict fentanyl concentrations in different newborn tissues due to intrauterine exposure. METHODS A fentanyl PBPK model in adults after intravenous and epidural administration was built, validated, and scaled to pregnancy and newborn populations. The dose that the newborn received transplacentally at birth was calculated using the pregnancy model. Then, the newborn bECF, saliva, plasma, and urine concentrations after such a dose were predicted using the newborn PBPK model. RESULTS After a maternal epidural dose of fentanyl 245 µg, the predicted newborn plasma and bECF levels were below the toxicity thresholds. Furthermore, the salivary threshold levels in newborns for fentanyl analgesic and respiratory depression effects were estimated to be 0.39 and 14.7-18.2 ng/ml, respectively. CONCLUSION The salivary TDM of fentanyl in newborns can be useful in newborns exposed to intrauterine exposure from parturient females dosed with epidural fentanyl. However, newborn-specific values of µ-opioid receptors IC50 for respiratory depression are needed.
Collapse
Affiliation(s)
- Mo'tasem M Alsmadi
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan.
- Nanotechnology Institute, Jordan University of Science and Technology, Irbid, Jordan.
| |
Collapse
|
9
|
Shi R, Fu Y, Gan Y, Wu D, Zhou S, Huang M. Use of polymyxin B with different administration methods in the critically ill patients with ventilation associated pneumonia: a single-center experience. Front Pharmacol 2023; 14:1222044. [PMID: 37719858 PMCID: PMC10502420 DOI: 10.3389/fphar.2023.1222044] [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: 05/16/2023] [Accepted: 08/22/2023] [Indexed: 09/19/2023] Open
Abstract
Background: Whether nebulized polymyxin B should be used as an adjunctive therapy or substitution strategy to intravenous polymyxin B for the treatment of ventilator-associated pneumonia (VAP) remains controversial. This study's aim is to evaluate the efficacy and safety of different administration ways of polymyxin B in the treatment of ventilator-associated pneumonia caused by extensively drug-resistant Gram-negative bacteria(XDR-GNB). Methods: This retrospective cohort study enrolled ventilator-associated pneumonia patients caused by XDR-GNB treated with polymyxin B in the intensive care unit. Patients were categorized by the administration methods as intravenous (IV) group, inhaled (IH) group, and the intravenous combined with inhaled (IV + IH) group. Microbiological outcome and clinical outcome were compared in each group. The side effects were also explored. Results: A total of 111 patients were enrolled and there was no difference in demographic and clinical characteristics among the three groups. In terms of efficacy, clinical cure or improvement was achieved in 21 patients (55.3%) in the intravenous group, 19 patients (50%) in the IH group, and 20 patients (57.1%) in IV + IH group (p = 0.815). All three groups showed high success rates in microbiological eradication, as 29 patients with negative cultures after medication in inhaled group. Among all the patients who had negative bacterial cultures after polymyxin B, the inhaled group had significantly shorter clearance time than the intravenous group (p = 0.002), but with no significant difference in 28-day mortality. Compared with intravenous group, a trend towards a lower risk of acute kidney injury was observed in inhaled group (p = 0.025). Conclusion: From the perspective of minimal systemic renal toxicity, nebulized polymyxin B as a substitution strategy to intravenous polymyxin B for the treatment of ventilator-associated pneumonia caused by XDR-GNB is feasible.
Collapse
Affiliation(s)
- Rupeng Shi
- Department of Geriatric ICU, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuanyuan Fu
- Department of Pharmacy, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yujing Gan
- Department of Geriatric ICU, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Danying Wu
- Department of Geriatric ICU, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Suming Zhou
- Department of Geriatric ICU, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Min Huang
- Department of Geriatric ICU, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
10
|
Li X, Chang P, Liu X, Zhao Z, Li W, Kang Y, Duan Y, Zhang W. Calibration and validation of ultraviolet time-of-flight mass spectrometry for online measurement of exhaled ciprofol. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:4179-4186. [PMID: 37578256 DOI: 10.1039/d3ay01168b] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Ciprofol (HSK 3486, C14H20O), a novel 2,6-disubstituted phenol derivative similar to propofol, is a new type of intravenous general anaesthetic. We found that the exhaled ciprofol concentration could be measured online by ultraviolet time-of-flight mass spectrometry (UV-TOFMS), which could be used to predict the plasma concentration and anaesthetic effects of ciprofol. In this study, we present the calibration method and validation results of UV-TOFMS for the quantification of ciprofol gas. Using a self-developed gas generator to prepare different concentrations of ciprofol calibration gas, we found a linear correlation between the concentration and intensity of ciprofol from 0 parts per trillion by level (pptv) to 485.85 pptv (R2 = 0.9987). The limit of quantification was 48.59 pptv and the limit of detection was 7.83 pptv. The imprecision was 12.44% at 97.17 pptv and was 8.96% at 485.85 pptv. The carry-over duration was 120 seconds. In addition, we performed a continuous infusion of ciprofol in beagles, measured the exhaled concentration of ciprofol by UV-TOFMS, determined the plasma concentration by high-performance liquid chromatography, and monitored the anaesthetic effects as reflected by the bispectral index value. The results showed that the exhaled and plasma concentrations of ciprofol were linearly correlated. The exhaled ciprofol concentration correlated well with the anaesthetic effect. The study showed that we could use UV-TOFMS to provide a continuous measurement of gaseous ciprofol concentration at 20 second intervals.
Collapse
Affiliation(s)
- Xiaoxiao Li
- Department of Anaesthesiology, West China Hospital, Sichuan University, China.
- Laboratory of Anaesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anaesthesiology, West China Hospital, Sichuan University, China
| | - Pan Chang
- Department of Anaesthesiology, West China Hospital, Sichuan University, China.
- Laboratory of Anaesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anaesthesiology, West China Hospital, Sichuan University, China
| | - Xing Liu
- Department of Anaesthesiology, West China Hospital, Sichuan University, China.
- Laboratory of Anaesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anaesthesiology, West China Hospital, Sichuan University, China
| | - Zhongjun Zhao
- School of Mechanical Engineering, Sichuan University, China
| | - Wenwen Li
- School of Mechanical Engineering, Sichuan University, China
| | - Yi Kang
- Department of Anaesthesiology, West China Hospital, Sichuan University, China.
- Laboratory of Anaesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anaesthesiology, West China Hospital, Sichuan University, China
| | - Yixiang Duan
- School of Mechanical Engineering, Sichuan University, China
| | - Wensheng Zhang
- Department of Anaesthesiology, West China Hospital, Sichuan University, China.
- Laboratory of Anaesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anaesthesiology, West China Hospital, Sichuan University, China
| |
Collapse
|
11
|
Alsmadi MM, Jaradat MM, Obaidat RM, Alnaief M, Tayyem R, Idkaidek N. The In Vitro, In Vivo, and PBPK Evaluation of a Novel Lung-Targeted Cardiac-Safe Hydroxychloroquine Inhalation Aerogel. AAPS PharmSciTech 2023; 24:172. [PMID: 37566183 DOI: 10.1208/s12249-023-02627-3] [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: 04/20/2023] [Revised: 07/21/2023] [Accepted: 07/25/2023] [Indexed: 08/12/2023] Open
Abstract
Hydroxychloroquine (HCQ) was repurposed for COVID-19 treatment. Subtherapeutic HCQ lung levels and cardiac toxicity of oral HCQ were overcome by intratracheal (IT) administration of lower HCQ doses. The crosslinker-free supercritical fluid technology (SFT) produces aerogels and impregnates them with drugs in their amorphous form with efficient controlled release. Mechanistic physiologically based pharmacokinetic (PBPK) modeling can predict the lung's epithelial lining fluid (ELF) drug levels. This study aimed to develop a novel HCQ SFT formulation for IT administration to achieve maximal ELF levels and minimal cardiac toxicity. HCQ SFT formulation was prepared and evaluated for physicochemical, in vitro release, pharmacokinetics, and cardiac toxicity. Finally, the rat HCQ ELF concentrations were predicted using PBPK modeling. HCQ was amorphous after loading into the chitosan-alginate nanoporous microparticles (22.7±7.6 μm). The formulation showed a zero-order release, with only 40% released over 30 min compared to 94% for raw HCQ. The formulation had a tapped density of 0.28 g/cm3 and a loading efficiency of 35.3±1.3%. The IT administration of SFT HCQ at 1 mg/kg resulted in 23.7-fold higher bioavailability, fourfold longer MRT, and eightfold faster absorption but lower CK-MB and LDH levels than oral raw HCQ at 4 mg/kg. The PBPK model predicted 6 h of therapeutic ELF levels for IT SFT HCQ and a 100-fold higher ELF-to-heart concentration ratio than oral HCQ. Our findings support the feasibility of lung-targeted and more effective SFT HCQ IT administration for COVID-19 compared to oral HCQ with less cardiac toxicity. Graphical abstract.
Collapse
Affiliation(s)
- Mo'tasem M Alsmadi
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan.
- Nanotechnology Institute, Jordan University of Science and Technology, Irbid, Jordan.
| | - Mays M Jaradat
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan
| | - Rana M Obaidat
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, The University of Jordan, Amman, Jordan
| | - Mohammad Alnaief
- Department of Pharmaceutical and Chemical Engineering, Faculty of Applied Medical Sciences, German Jordanian University, Amman, Jordan
| | | | | |
Collapse
|
12
|
Escalona J, Soto D, Oviedo V, Rivas E, Severino N, Kattan E, Andresen M, Bravo S, Basoalto R, Bachmann MC, Wong KY, Pavez N, Bruhn A, Bugedo G, Retamal J. Beta-Lactam Antibiotics Can Be Measured in the Exhaled Breath Condensate in Mechanically Ventilated Patients: A Pilot Study. J Pers Med 2023; 13:1146. [PMID: 37511759 PMCID: PMC10381781 DOI: 10.3390/jpm13071146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Different techniques have been proposed to measure antibiotic levels within the lung parenchyma; however, their use is limited because they are invasive and associated with adverse effects. We explore whether beta-lactam antibiotics could be measured in exhaled breath condensate collected from heat and moisture exchange filters (HMEFs) and correlated with the concentration of antibiotics measured from bronchoalveolar lavage (BAL). We designed an observational study in patients undergoing mechanical ventilation, which required a BAL to confirm or discard the diagnosis of pneumonia. We measured and correlated the concentration of beta-lactam antibiotics in plasma, epithelial lining fluid (ELF), and exhaled breath condensate collected from HMEFs. We studied 12 patients, and we detected the presence of antibiotics in plasma, ELF, and HMEFs from every patient studied. The concentrations of antibiotics were very heterogeneous over the population studied. The mean antibiotic concentration was 293.5 (715) ng/mL in plasma, 12.3 (31) ng/mL in ELF, and 0.5 (0.9) ng/mL in HMEF. We found no significant correlation between the concentration of antibiotics in plasma and ELF (R2 = 0.02, p = 0.64), between plasma and HMEF (R2 = 0.02, p = 0.63), or between ELF and HMEF (R2 = 0.02, p = 0.66). We conclude that beta-lactam antibiotics can be detected and measured from the exhaled breath condensate accumulated in the HMEF from mechanically ventilated patients. However, no correlations were observed between the antibiotic concentrations in HMEF with either plasma or ELF.
Collapse
Affiliation(s)
- José Escalona
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Unidad de Paciente Crítico, Hospital El Salvador, Santiago 8331150, Chile
| | - Dagoberto Soto
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Vanessa Oviedo
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Elizabeth Rivas
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Nicolás Severino
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Programa de Farmacología y Toxicología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Eduardo Kattan
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Max Andresen
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Sebastián Bravo
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Roque Basoalto
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Programa de Medicina Física y Rehabilitación, Red Salud UC-CHRISTUS, Santiago 8331150, Chile
| | - María Consuelo Bachmann
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Kwok-Yin Wong
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Kowloon 999077, Hong Kong
| | - Nicolás Pavez
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Departamento de Medicina Interna, Facultad de Medicina, Universidad de Concepción, Concepción 4030000, Chile
| | - Alejandro Bruhn
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Guillermo Bugedo
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Jaime Retamal
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| |
Collapse
|
13
|
Lepak AJ, Trang M, Hammel JP, Sader HS, Bhavnani SM, VanScoy BD, Pogue JM, Ambrose PG, Andes DR. Development of Modernized Acinetobacter baumannii Susceptibility Test Interpretive Criteria for Recommended Antimicrobial Agents Using Pharmacometric Approaches. Antimicrob Agents Chemother 2023; 67:e0145222. [PMID: 36946729 PMCID: PMC10112158 DOI: 10.1128/aac.01452-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 02/13/2023] [Indexed: 03/23/2023] Open
Abstract
Acinetobacter baumannii-Acinetobacter calcoaceticus complex (referred to herein as A. baumannii) treatment guidelines contain numerous older antimicrobial agents with susceptibility test interpretive criteria (STIC, also known as susceptibility breakpoints) set using only epidemiological data. We utilized a combination of in vitro surveillance data, preclinical murine thigh and lung infection models, population pharmacokinetics, simulation, and pharmacokinetic/pharmacodynamic (PK/PD) target attainment analyses to evaluate A. baumannii STIC for four commonly recommended antimicrobials from different classes (amikacin, ceftazidime, ciprofloxacin, and minocycline). Antimicrobial in vitro surveillance data were based on 1,647 clinical A. baumannii isolates obtained from 109 centers in the United States and Europe. Among these isolates, 5 were selected for evaluation in murine infection models based on fitness and MIC variability. PK and dose-ranging studies were conducted using neutropenic murine thigh and lung infection models The MIC ranges for the 5 isolates evaluated were as follows: amikacin, 2 to 32 μg/mL; ceftazidime, 4 to 16 μg/mL; ciprofloxacin, 0.12 to 2 μg/mL; minocycline, 0.25 to 4 μg/mL. All organisms grew ≥1.5 log10 CFU in both models in untreated controls. Plasma and epithelial lining fluid (ELF) pharmacokinetics for all drugs were determined in mice using liquid chromatography-tandem mass spectrometry (LC-MS/MS) methods. For each isolate, 5 dose levels of each drug were tested individually in the thigh and lung infection model. The inoculum ranged from 7.9 to 8.4 and 6.8 to 7.7 log10 CFU/mL for the lung and thigh models, respectively. PK/PD targets associated with net bacterial stasis and 1- and 2-log10 CFU reductions from baseline were identified for each organism/infection model using Hill-type models. Population pharmacokinetic models for each agent were identified from the literature. Using demographic variables for simulated patients with hospital-acquired or ventilator-associated bacterial pneumonia or urinary tract infections (including acute pyelonephritis) who were administered maximal dosing regimens of each agent, estimates of protein binding, and ELF penetration ratios based on data from the literature, free-drug plasma and total-drug concentration-time profiles were generated, and PK/PD indices by MIC were calculated. Percent probabilities of attaining median and randomly assigned PK/PD targets associated with the above-described endpoints were determined. Recommended susceptible breakpoints for each agent were those representing the highest MIC at which the percent probabilities of achieving PK/PD targets associated with a 1-log10 CFU reduction from baseline approached or were ≥90%. The following susceptible breakpoints for A. baumannii were identified: amikacin, ≤8 μg/mL for pneumonia; ceftazidime, ≤32 and ≤8 μg/mL for pneumonia; ciprofloxacin, ≤1 μg/mL; and minocycline, ≤0.5/≤1 μg/mL which correspond to the standard and high minocycline dosing regimens of 200 mg per day and 200 mg every 12 h, respectively. Implementation of appropriate STIC will help clinicians optimally use the above-described agents and improve the likelihood of successful patient outcomes.
Collapse
Affiliation(s)
- A. J. Lepak
- University of Wisconsin—Madison, Department of Medicine, Madison, Wisconsin, USA
| | - M. Trang
- Institute for Clinical Pharmacodynamics, Schenectady, New York, USA
| | - J. P. Hammel
- Institute for Clinical Pharmacodynamics, Schenectady, New York, USA
| | | | - S. M. Bhavnani
- Institute for Clinical Pharmacodynamics, Schenectady, New York, USA
| | - B. D. VanScoy
- Institute for Clinical Pharmacodynamics, Schenectady, New York, USA
| | - J. M. Pogue
- University of Michigan, Ann Arbor, Michigan, USA
| | - P. G. Ambrose
- Institute for Clinical Pharmacodynamics, Schenectady, New York, USA
| | - D. R. Andes
- University of Wisconsin—Madison, Department of Medicine, Madison, Wisconsin, USA
| | - United States Committee on Antimicrobial Susceptibility Testing
- University of Wisconsin—Madison, Department of Medicine, Madison, Wisconsin, USA
- Institute for Clinical Pharmacodynamics, Schenectady, New York, USA
- JMI Laboratories, North Liberty, Iowa, USA
- University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
14
|
Alsmadi MM, Idkaidek N. The Analysis of Pethidine Pharmacokinetics in Newborn Saliva, Plasma, and Brain Extracellular Fluid After Prenatal Intrauterine Exposure from Pregnant Mothers Receiving Intramuscular Dose Using PBPK Modeling. Eur J Drug Metab Pharmacokinet 2023; 48:281-300. [PMID: 37017867 DOI: 10.1007/s13318-023-00823-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2023] [Indexed: 04/06/2023]
Abstract
BACKGROUND AND OBJECTIVE Pethidine (meperidine) can decrease labor pain-associated mother's hyperventilation and high cortisol-induced newborn complications. However, prenatal transplacentally acquired pethidine can cause side effects in newborns. High pethidine concentrations in the newborn brain extracellular fluid (bECF) can cause a serotonin crisis. Therapeutic drug monitoring (TDM) in newborns' blood distresses them and increases infection incidence, which can be overcome by using salivary TDM. Physiologically based pharmacokinetic (PBPK) modeling can predict drug concentrations in newborn plasma, saliva, and bECF after intrauterine pethidine exposure. METHODS A healthy adult PBPK model was constructed, verified, and scaled to newborn and pregnant populations after intravenous and intramuscular pethidine administration. The pregnancy PBPK model was used to predict the newborn dose received transplacentally at birth, which was used as input to the newborn PBPK model to predict newborn plasma, saliva, and bECF pethidine concentrations and set correlation equations between them. RESULTS Pethidine can be classified as a Salivary Excretion Classification System class II drug. The developed PBPK model predicted that, after maternal pethidine intramuscular doses of 100 mg and 150 mg, the newborn plasma and bECF concentrations were below the toxicity thresholds. Moreover, it was estimated that newborn saliva concentrations of 4.7 µM, 11.4 µM, and 57.7 µM can be used as salivary threshold concentrations for pethidine analgesic effects, side effects, and the risk for serotonin crisis, respectively, in newborns. CONCLUSION It was shown that saliva can be used for pethidine TDM in newborns during the first few days after delivery to mothers receiving pethidine.
Collapse
Affiliation(s)
- Mo'tasem M Alsmadi
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Jordan University of Science and Technology, P.O.Box 3030, Irbid, 22110, Jordan.
- Nanotechnology Institute, Jordan University of Science and Technology, P.O.Box 3030, Irbid, 22110, Jordan.
| | | |
Collapse
|
15
|
Alsmadi MM. The investigation of the complex population-drug-drug interaction between ritonavir-boosted lopinavir and chloroquine or ivermectin using physiologically-based pharmacokinetic modeling. Drug Metab Pers Ther 2023; 38:87-105. [PMID: 36205215 DOI: 10.1515/dmpt-2022-0130] [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: 04/29/2022] [Accepted: 08/19/2022] [Indexed: 11/07/2022]
Abstract
OBJECTIVES Therapy failure caused by complex population-drug-drug (PDDI) interactions including CYP3A4 can be predicted using mechanistic physiologically-based pharmacokinetic (PBPK) modeling. A synergy between ritonavir-boosted lopinavir (LPVr), ivermectin, and chloroquine was suggested to improve COVID-19 treatment. This work aimed to study the PDDI of the two CYP3A4 substrates (ivermectin and chloroquine) with LPVr in mild-to-moderate COVID-19 adults, geriatrics, and pregnancy populations. METHODS The PDDI of LPVr with ivermectin or chloroquine was investigated. Pearson's correlations between plasma, saliva, and lung interstitial fluid (ISF) levels were evaluated. Target site (lung epithelial lining fluid [ELF]) levels of ivermectin and chloroquine were estimated. RESULTS Upon LPVr coadministration, while the chloroquine plasma levels were reduced by 30, 40, and 20%, the ivermectin plasma levels were increased by a minimum of 425, 234, and 453% in adults, geriatrics, and pregnancy populations, respectively. The established correlation equations can be useful in therapeutic drug monitoring (TDM) and dosing regimen optimization. CONCLUSIONS Neither chloroquine nor ivermectin reached therapeutic ELF levels in the presence of LPVr despite reaching toxic ivermectin plasma levels. PBPK modeling, guided with TDM in saliva, can be advantageous to evaluate the probability of reaching therapeutic ELF levels in the presence of PDDI, especially in home-treated patients.
Collapse
Affiliation(s)
- Mo'tasem M Alsmadi
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
16
|
Shen Y, Kuti JL. Optimizing antibiotic dosing regimens for nosocomial pneumonia: a window of opportunity for pharmacokinetic and pharmacodynamic modeling. Expert Opin Drug Metab Toxicol 2023; 19:13-25. [PMID: 36786064 DOI: 10.1080/17425255.2023.2178896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
INTRODUCTION Determining antibiotic exposure in the lung and the threshold(s) needed for effective antibacterial killing is paramount during development of new antibiotics for the treatment of nosocomial pneumonia, as these exposures directly affect clinical outcomes and resistance development. The use of pharmacokinetic and pharmacodynamic modeling is recommended by regulatory agencies to evaluate antibiotic pulmonary exposure and optimize dosage regimen selection. This process has been implemented in newer antibiotic development. AREAS COVERED This review will discuss the basis for conducting pharmacokinetic and pharmacodynamic studies to support dosage regimen selection and optimization for the treatment of nosocomial pneumonia. Pharmacokinetic/pharmacodynamic data that supported recent hospital-acquired bacterial pneumonia/ventilator-associated bacterial pneumonia indications for ceftolozane/tazobactam, ceftazidime/avibactam, imipenem/cilastatin/relebactam, and cefiderocol will be reviewed. EXPERT OPINION Optimal drug development requires the integration of preclinical pharmacodynamic studies, healthy volunteers and ideally patient bronchoalveolar lavage pharmacokinetic studies, Monte-Carlo simulation, and clinical trials. Currently, plasma exposure has been successfully used as a surrogate for lung exposure threshold. Future studies are needed to identify the value of lung pharmacodynamic thresholds in nosocomial pneumonia antibiotic dosage optimization.
Collapse
Affiliation(s)
- Yuwei Shen
- Center for Anti-Infective Research and Development, Hartford Hospital, Hartford, CT USA
| | - Joseph L Kuti
- Center for Anti-Infective Research and Development, Hartford Hospital, Hartford, CT USA
| |
Collapse
|
17
|
Pharmacodynamics of Temocillin in Neutropenic Murine Infection Models. Antimicrob Agents Chemother 2023; 67:e0143322. [PMID: 36692307 PMCID: PMC9933716 DOI: 10.1128/aac.01433-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Temocillin is used for the treatment of various infections caused by Enterobacterales. The pharmacokinetic (PK)/pharmacodynamic (PD) index that is best correlated with the activity of beta-lactams is the percentage of time that the unbound concentration exceeds the MIC (%fT>MIC). However, the %fT>MIC needed for a bacteriostatic or killing effect of temocillin is unknown in thigh and lung infection models. In the present study, we studied the temocillin PK in plasma and epithelial lining fluid (ELF) of infected neutropenic mice and determined the plasma exposure-response relationships for Escherichia coli and Klebsiella pneumoniae. Neutropenic murine thigh and lung infection models were used. The bacterial loads in the thighs or lungs were determined. A sigmoid maximum-effect model was used to fit the plasma exposure-response relationship. A one-compartment model with first-order absorption best described temocillin PK (clearance [CL], 1.03 L/h/kg; volume of distribution [V], 0.457 L/kg). Protein binding was 78.2% ± 1.3% across different plasma concentrations. A static effect was achieved for all strains in both the thigh and lung infection models. However, the median %fT>MIC needed for a static effect was much lower in the lung infection model (27.8% for E. coli and 38.2% for K. pneumoniae) than in the thigh infection model (65.2% for E. coli and 64.9% for K. pneumoniae). A 1-log kill was reached for all strains in the lung infection model (median %fT>MIC values of 42.1% for E. coli and 44.1% for K. pneumoniae) and 7 out of 8 strains in the thigh infection model (median %fT>MIC values of 85.4% for E. coli and 74.5% for K. pneumoniae). These data support the use of temocillin in patients with pneumonia.
Collapse
|
18
|
Nguyen PTN, Le NV, Dinh HMN, Nguyen BQP, Nguyen TVA. Lung penetration and pneumococcal target binding of antibiotics in lower respiratory tract infection. Curr Med Res Opin 2022; 38:2085-2095. [PMID: 36189961 DOI: 10.1080/03007995.2022.2131304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
OBJECTIVES To achieve the therapeutic effects, antibiotics must penetrate rapidly into infection sites and bind to targets. This study reviewed updated knowledge on the ability of antibiotics to penetrate into the lung, their physicochemical properties influencing the pulmonary penetration and their ability to bind to targets on pneumococci. METHODS A search strategy was developed using PubMED, Web of Science, and ChEMBL. Data on serum protein binding, drug concentration, target binding ability, drug transporters, lung penetration, physicochemical properties of antibiotics in low respiratory tract infection (LRTI) were collected. RESULTS It was seen that infection site-to-serum concentration ratios of most antibiotics are >1 at different time points except for ceftriaxone, clindamycin and vancomycin. Most agents have proper physicochemical properties that facilitate antibiotic penetration. In antimicrobial-resistant Streptococcus pneumoniae, the binding affinity of antibiotics to targets mostly decreases compared to that in susceptible strains. The data on binding affinity of linezolid, clindamycin and vancomycin were insufficient. The higher drug concentration at the infection sites compared to that in the blood can be associated with inflammation conditions. Little evidence showed the effect of drug transporters on the clinical efficacy of antibiotics against LRTI. CONCLUSIONS Data on antibiotic penetration into the lung in LRTI patients and binding affinity of antibiotics for pneumococcal targets are still limited. Further studies are required to clarify the associations of the lung penetration and target binding ability of antibitotics with therapeutic efficacy to help propose the right antibiotics for LRTI.
Collapse
Affiliation(s)
| | - Nho Van Le
- Danang University of Medical Technology and Pharmacy, Da Nang, Vietnam
| | | | | | - Thi Van Anh Nguyen
- Department of Life Sciences, University of Science and Technology of Hanoi (USTH), Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| |
Collapse
|
19
|
Tetz G, Tetz V. Overcoming Antibiotic Resistance with Novel Paradigms of Antibiotic Selection. Microorganisms 2022; 10:2383. [PMID: 36557636 PMCID: PMC9781420 DOI: 10.3390/microorganisms10122383] [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: 11/05/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/04/2022] Open
Abstract
Conventional antimicrobial susceptibility tests, including phenotypic and genotypic methods, are insufficiently accurate and frequently fail to identify effective antibiotics. These methods predominantly select therapies based on the antibiotic response of only the lead bacterial pathogen within pure bacterial culture. However, this neglects the fact that, in the majority of human infections, the lead bacterial pathogens are present as a part of multispecies communities that modulate the response of these lead pathogens to antibiotics and that multiple pathogens can contribute to the infection simultaneously. This discrepancy is a major cause of the failure of antimicrobial susceptibility tests to detect antibiotics that are effective in vivo. This review article provides a comprehensive overview of the factors that are missed by conventional antimicrobial susceptibility tests and it explains how accounting for these methods can aid the development of novel diagnostic approaches.
Collapse
Affiliation(s)
- George Tetz
- Human Microbiology Institute, New York, NY 100141, USA
| | | |
Collapse
|
20
|
Cooper MS, Zhang L, Ibrahim M, Zhang K, Sun X, Röske J, Göhl M, Brönstrup M, Cowell JK, Sauerhering L, Becker S, Vangeel L, Jochmans D, Neyts J, Rox K, Marsh GP, Maple HJ, Hilgenfeld R. Diastereomeric Resolution Yields Highly Potent Inhibitor of SARS-CoV-2 Main Protease. J Med Chem 2022; 65:13328-13342. [PMID: 36179320 PMCID: PMC9574927 DOI: 10.1021/acs.jmedchem.2c01131] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Indexed: 12/02/2022]
Abstract
SARS-CoV-2 is the causative agent behind the COVID-19 pandemic. The main protease (Mpro, 3CLpro) of SARS-CoV-2 is a key enzyme that processes polyproteins translated from the viral RNA. Mpro is therefore an attractive target for the design of inhibitors that block viral replication. We report the diastereomeric resolution of the previously designed SARS-CoV-2 Mpro α-ketoamide inhibitor 13b. The pure (S,S,S)-diastereomer, 13b-K, displays an IC50 of 120 nM against the Mpro and EC50 values of 0.8-3.4 μM for antiviral activity in different cell types. Crystal structures have been elucidated for the Mpro complexes with each of the major diastereomers, the active (S,S,S)-13b (13b-K), and the nearly inactive (R,S,S)-13b (13b-H); results for the latter reveal a novel binding mode. Pharmacokinetic studies show good levels of 13b-K after inhalative as well as after peroral administration. The active inhibitor (13b-K) is a promising candidate for further development as an antiviral treatment for COVID-19.
Collapse
Affiliation(s)
- Mark S. Cooper
- Bio-Techne
(Tocris), The Watkins
Building, Atlantic Road, Bristol, BS11 9QD, U.K.
| | - Linlin Zhang
- Institute
of Molecular Medicine, University of Lübeck, 23562 Lübeck, Germany
| | - Mohamed Ibrahim
- Institute
of Molecular Medicine, University of Lübeck, 23562 Lübeck, Germany
| | - Kaixuan Zhang
- Institute
of Molecular Medicine, University of Lübeck, 23562 Lübeck, Germany
| | - Xinyuanyuan Sun
- Institute
of Molecular Medicine, University of Lübeck, 23562 Lübeck, Germany
| | - Judith Röske
- Institute
of Molecular Medicine, University of Lübeck, 23562 Lübeck, Germany
| | - Matthias Göhl
- Department
of Chemical Biology, Helmholtz Centre for
Infection Research (HZI), 38124 Braunschweig, Germany
| | - Mark Brönstrup
- Department
of Chemical Biology, Helmholtz Centre for
Infection Research (HZI), 38124 Braunschweig, Germany
- German
Center for Infection Research (DZIF), Partner
Site Braunschweig-Hannover, 38124 Braunschweig, Germany
| | - Justin K. Cowell
- Bio-Techne
(Tocris), The Watkins
Building, Atlantic Road, Bristol, BS11 9QD, U.K.
| | - Lucie Sauerhering
- Institute
of Virology, University of Marburg, 35043 Marburg, Germany
| | - Stephan Becker
- Institute
of Virology, University of Marburg, 35043 Marburg, Germany
- German Center
for Infection Research (DZIF), Marburg-Gießen-Langen
Site, 35043 Marburg, Germany
| | - Laura Vangeel
- Rega
Institute, Department of Microbiology, Immunology and Transplantation, KU Leuven, B-3000 Leuven, Belgium
| | - Dirk Jochmans
- Rega
Institute, Department of Microbiology, Immunology and Transplantation, KU Leuven, B-3000 Leuven, Belgium
| | - Johan Neyts
- Rega
Institute, Department of Microbiology, Immunology and Transplantation, KU Leuven, B-3000 Leuven, Belgium
| | - Katharina Rox
- Department
of Chemical Biology, Helmholtz Centre for
Infection Research (HZI), 38124 Braunschweig, Germany
- German
Center for Infection Research (DZIF), Partner
Site Braunschweig-Hannover, 38124 Braunschweig, Germany
| | - Graham P. Marsh
- Bio-Techne
(Tocris), The Watkins
Building, Atlantic Road, Bristol, BS11 9QD, U.K.
| | - Hannah J. Maple
- Bio-Techne
(Tocris), The Watkins
Building, Atlantic Road, Bristol, BS11 9QD, U.K.
| | - Rolf Hilgenfeld
- Institute
of Molecular Medicine, University of Lübeck, 23562 Lübeck, Germany
- German
Center for Infection Research (DZIF), Hamburg-Lübeck-Borstel-Riems
Site, University of Lübeck, 23562 Lübeck, Germany
| |
Collapse
|
21
|
Delgado CP, Rocha JBT, Orian L, Bortoli M, Nogara PA. In silico studies of M pro and PL pro from SARS-CoV-2 and a new class of cephalosporin drugs containing 1,2,4-thiadiazole. Struct Chem 2022; 33:2205-2220. [PMID: 36106095 PMCID: PMC9463509 DOI: 10.1007/s11224-022-02036-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 08/09/2022] [Indexed: 11/29/2022]
Abstract
The SARS-CoV-2 proteases Mpro and PLpro are important targets for the development of antivirals against COVID-19. The functional group 1,2,4-thiadiazole has been indicated to inhibit cysteinyl proteases, such as papain and cathepsins. Of note, the 1,2,4-thiadiazole moiety is found in a new class of cephalosporin FDA-approved antibiotics: ceftaroline fosamil, ceftobiprole, and ceftobiprole medocaril. Here we investigated the interaction of these new antibiotics and their main metabolites with the SARS-CoV-2 proteases by molecular docking, molecular dynamics (MD), and density functional theory (DFT) calculations. Our results indicated the PLpro enzyme as a better in silico target for the new antibacterial cephalosporins. The results with ceftaroline fosamil and the dephosphorylate metabolite compounds should be tested as potential inhibitor of PLpro, Mpro, and SARS-CoV-2 replication in vitro. In addition, the data here reported can help in the design of new potential drugs against COVID-19 by exploiting the S atom reactivity in the 1,2,4-thiadiazole moiety. Supplementary Information The online version contains supplementary material available at 10.1007/s11224-022-02036-5.
Collapse
Affiliation(s)
- Cássia Pereira Delgado
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS 97105-900 Brazil
| | - João Batista Teixeira Rocha
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS 97105-900 Brazil
| | - Laura Orian
- Dipartimento di Scuenze Chimiche, Università degli Studi di Padova, Via Marzolo 1, 35131 Padua, Italy
| | - Marco Bortoli
- Institut de Química Computacionali Catàlisi (IQCC), Departament de Química, Facultat de Ciències, Universitat de Girona, C/M. A. Capmany 69, 17003 Girona, Spain
| | - Pablo Andrei Nogara
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria (UFSM), Santa Maria, RS 97105-900 Brazil
| |
Collapse
|
22
|
Kawaguchi N, Katsube T, Echols R, Wajima T, Nicolau DP. Intrapulmonary Pharmacokinetic Modeling and Simulation of Cefiderocol, a Parenteral Siderophore Cephalosporin, in Patients With Pneumonia and Healthy Subjects. J Clin Pharmacol 2022; 62:670-680. [PMID: 34648652 PMCID: PMC9306831 DOI: 10.1002/jcph.1986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/07/2021] [Indexed: 11/16/2022]
Abstract
Cefiderocol is a siderophore cephalosporin for the treatment of infections caused by gram-negative bacteria including carbapenem-resistant strains. The aim of this study was to develop an intrapulmonary pharmacokinetic (PK) model of cefiderocol and assess the PK profile in lungs. An intrapulmonary PK model of cefiderocol was developed using the concentration data in plasma and epithelial lining fluid (ELF) from 7 patients with pneumonia requiring mechanical ventilation and 20 healthy subjects. Subsequently, the model was applied to assess the ELF exposure of 125 patients with nosocomial pneumonia. Monte Carlo simulations were performed to calculate the probability of target attainment for the percentage of time for which free ELF concentrations exceed the minimum inhibitory concentration (MIC) over the dosing interval (%fT>MIC,ELF ). The developed model adequately described ELF concentrations and suggested the delayed distribution in ELF for patients with pneumonia compared to healthy subjects. Lung penetration ratio of cefiderocol in patients with pneumonia was calculated to be 34%, which was 1.4-fold that in healthy subjects. The estimated %fT>MIC,ELF was 100% in most of patients with nosocomial pneumonia, and no PK/pharmacodynamic relationship with %fT>MIC,ELF was found for microbiological or clinical outcome. The probability of target attainment for 100% fT>MIC,ELF was ≥ 99.5% against MICs ≤2 μg/mL and ≥87.0% against MICs ≤4 μg/mL regardless of renal function. The median of simulated ELF trough concentrations at steady state was >4 μg/mL regardless of renal function. These results reveal the adequacy of cefiderocol exposure in plasma and ELF at the recommended dosing regimens adjusted on the basis of renal function in critically ill patients with pneumonia.
Collapse
Affiliation(s)
- Nao Kawaguchi
- Clinical Pharmacology & PharmacokineticsShionogi & Co., Ltd.OsakaJapan
| | - Takayuki Katsube
- Clinical Pharmacology & PharmacokineticsShionogi & Co., Ltd.OsakaJapan
| | - Roger Echols
- Infectious Disease Drug Development Consulting, LLCEastonConnecticutUSA
| | - Toshihiro Wajima
- Clinical Pharmacology & PharmacokineticsShionogi & Co., Ltd.OsakaJapan
- Clinical PharmacologyIDEC IncShinjuku‐kuTokyoJapan
| | - David P. Nicolau
- Center for Anti‐Infective Research and DevelopmentHartford HospitalHartfordConnecticutUSA
| |
Collapse
|
23
|
Kosinsky Y, Peskov K, Stanski DR, Wetmore D, Vinetz J. Semi-Mechanistic Pharmacokinetic-Pharmacodynamic Model of Camostat Mesylate-Predicted Efficacy against SARS-CoV-2 in COVID-19. Microbiol Spectr 2022; 10:e0216721. [PMID: 35412356 PMCID: PMC9047529 DOI: 10.1128/spectrum.02167-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 03/28/2022] [Indexed: 12/15/2022] Open
Abstract
The SARS-CoV-2 coronavirus, which causes COVID-19, uses a viral surface spike protein for host cell entry and the human cell-surface transmembrane serine protease, TMPRSS2, to process the spike protein. Camostat mesylate, an orally available and clinically used serine protease inhibitor, inhibits TMPRSS2, supporting clinical trials to investigate its use in COVID-19. A one-compartment pharmacokinetic (PK)/pharmacodynamic (PD) model for camostat and the active metabolite FOY-251 was developed, incorporating TMPRSS2 reversible covalent inhibition by FOY-251, and empirical equations linking TMPRSS2 inhibition of SARS-CoV-2 cell entry. The model predicts that 95% inhibition of TMPRSS2 is required for 50% inhibition of viral entry efficiency. For camostat 200 mg dosed four times daily, 90% inhibition of TMPRSS2 is predicted to occur but with only about 40% viral entry inhibition. For 3-fold higher camostat dosing, marginal improvement of viral entry rate inhibition, up to 54%, is predicted. Because respiratory tract viral load may be associated with negative outcome, even modestly reducing viral entry and respiratory tract viral load may reduce disease progression. This modeling also supports medicinal chemistry approaches to enhancing PK/PD and potency of the camostat molecule. IMPORTANCE Strategies to repurpose already-approved drugs for the treatment of COVID-19 has been attractive since the beginning of the pandemic. Camostat mesylate, a serine protease inhibitor approved in Japan for the treatment of acute exacerbations of chronic pancreatitis, inhibits TMPRSS1, a host cell surface serine protease essential for SARS-CoV-2 viral entry. In vitro experiments provided data suggesting that camostat might be effective in the treatment of COVID-19. Multiple clinical trials were planned to test the hypothesis that camostat would be beneficial for treating COVID-19 (for example, clinicaltrials.gov, NCT04353284). The present work used a one-compartment pharmacokinetic (PK)/pharmacodynamic (PD) mathematical model for camostat and the active metabolite FOY-251, incorporating TMPRSS2 reversible covalent inhibition by FOY-251, and empirical equations linking TMPRSS2 inhibition of SARS-CoV-2 cell entry. This work is valuable to guide further development of camostat mesylate and possible medicinal chemistry derivatives for the treatment of COVID-19.
Collapse
Affiliation(s)
| | - Kirill Peskov
- M&S Decisions LLC, Moscow, Russia
- Sechenov First Moscow State Medical University, Moscow, Russia
- STU “Sirius,” Sochi, Russia
| | | | - Diana Wetmore
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Joseph Vinetz
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
24
|
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: 5.5] [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
|
25
|
Prediction of lung exposure to anti-tubercular drugs using plasma pharmacokinetic data: implications for dose selection. Eur J Pharm Sci 2022; 173:106163. [DOI: 10.1016/j.ejps.2022.106163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 12/28/2021] [Accepted: 03/02/2022] [Indexed: 01/08/2023]
|
26
|
PK-PD Modeling and Optimal Dosing Regimen of Acetylkitasamycin against Streptococcus suis in Piglets. Antibiotics (Basel) 2022; 11:antibiotics11020283. [PMID: 35203885 PMCID: PMC8868236 DOI: 10.3390/antibiotics11020283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/16/2022] [Accepted: 02/19/2022] [Indexed: 02/04/2023] Open
Abstract
Streptococcus suis (S. suis) causes severe respiratory diseases in pigs and is also an important pathogen causing hidden dangers to public health and safety. Acetylkitasamycin is a new macrolide agent that has shown good activity to Gram-positive cocci such as Streptococcus. The purpose of this study was to perform pharmacokinetic–pharmacodynamic (PK-PD) modeling to formulate a dosing regimen of acetylkitasamycin for treatment of S. suis and to decrease the emergence of acetylkitasamycin-resistant S. suis. The minimal inhibitory concentration (MIC) of 110 S. suis isolates was determined by broth micro dilution method. The MIC50 of the 55 sensitive S. suis isolates was 1.21 μg/mL. The strain HB1607 with MIC close to MIC50 and high pathogenicity was used for the PK-PD experiments. The MIC and MBC of HB1607 in both MH broth and pulmonary epithelial lining fluid (PELF) was 1 and 2 μg/mL, respectively. The liquid chromatography–tandem mass spectrometry (LC-MS/MS) method was used to determine the concentration change of acetylkitasamycin in piglet plasma and PELF after intragastric administration of a single dose of 50 mg/kg b.w. acetylkitasamycin. The PK parameters were calculated by WinNolin software. The PK data showed that the maximum concentration (Cmax), peak time (Tmax), and area under the concentration–time curve (AUC) were 9.84 ± 0.39 μg/mL, 4.27 ± 0.19 h and 248.58 ± 21.17 h·μg/mL, respectively. Integration of the in vivo PK data and ex vivo PD data, an inhibition sigmoid Emax equation was established. The dosing regimen of acetylkitasamycin for the treatment S. suis infection established as 33.12 mg/kg b.w. every 12 h for 3 days. This study provided a reasonable dosing regimen for a new drug used in clinical treatment, which can effectively be used to treat S. suis infection and slow down the generation of drug resistance.
Collapse
|
27
|
Palmer LB, Smaldone GC. The Unfulfilled Promise of Inhaled Therapy in Ventilator-Associated Infections: Where Do We Go from Here? J Aerosol Med Pulm Drug Deliv 2022; 35:11-24. [PMID: 35099284 PMCID: PMC8867107 DOI: 10.1089/jamp.2021.0023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Respiratory infection is common in intubated/tracheotomized patients and systemic antibiotic therapy is often unrewarding. In 1967, the difficulty in treating Gram-negative respiratory infections led to the use of inhaled gentamicin, targeting therapy directly to the lungs. Fifty-three years later, the effects of topical therapy in the intubated patient remain undefined. Clinical failures with intravenous antibiotics persist and instrumented patients are now infected by many more multidrug-resistant Gram-negative species as well as methicillin-resistant Staphylococcus aureus. Multiple systematic reviews and meta-analyses suggest that there may be a role for inhaled delivery but “more research is needed.” Yet there is still no Food and Drug Administration (FDA) approved inhaled antibiotic for the treatment of ventilator-associated infection, the hallmark of which is the foreign body in the upper airway. Current pulmonary and infectious disease guidelines suggest using aerosols only in the setting of Gram-negative infections that are resistant to all systemic antibiotics or not to use them at all. Recently two seemingly well-designed large randomized placebo-controlled Phase 2 and Phase 3 clinical trials of adjunctive inhaled therapy for the treatment of ventilator-associated pneumonia failed to show more rapid resolution of pneumonia symptoms or effect on mortality. Despite evolving technology of delivery devices and more detailed understanding of the factors affecting delivery, treatment effects were no better than placebo. What is wrong with our approach to ventilator- associated infection? Is there a message from the large meta-analyses and these two large recent multisite trials? This review will suggest why current therapies are unpredictable and have not fulfilled the promise of better outcomes. Data suggest that future studies of inhaled therapy, in the milieu of worsening bacterial resistance, require new approaches with completely different indications and endpoints to determine whether inhaled therapy indeed has an important role in the treatment of ventilated patients.
Collapse
Affiliation(s)
- Lucy B Palmer
- Pulmonary, Critical Care and Sleep Division, Department of Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Gerald C Smaldone
- Pulmonary, Critical Care and Sleep Division, Department of Medicine, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
28
|
Abouelhassan YP, Nicolau D. Pharmacokinetic/Pharmacodynamic Optimization of Hospital-Acquired and Ventilator-Associated Pneumonia: Challenges and Strategies. Semin Respir Crit Care Med 2022; 43:175-182. [PMID: 35088402 DOI: 10.1055/s-0041-1742105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Hospital-acquired pneumonia (HAP) and ventilator-associated pneumonia (VAP) are correlated with high mortality rates worldwide. Thus, the administration of antibiotic therapy with appropriate dosing regimen is critical. An efficient antibiotic is needed to maintain an adequate concentration at the infection site, for a sufficient period of time, to achieve the best therapeutic outcome. It can, however, be challenging for antibiotics to penetrate the pulmonary system due to the complexity of its structure. Crossing the blood alveolar barrier is a difficult process determined by multiple factors that are either drug related or infection related. Thus, the understanding of pharmacokinetics/pharmacodynamics (PK/PD) of antibiotics identifies the optimum dosing regimens to achieve drug penetration into the epithelial lining fluid at adequate therapeutic concentrations. Critically ill patients in the ICU can express augmented renal clearance (ARC), characterized by enhanced renal function, or may have renal dysfunction necessitating supportive care such as continuous renal replacement therapy (CRRT). Both ARC and CRRT can alter drug elimination, thus affecting drug concentrations. PK of critically ill patients is less clear due to the multiple variabilities associated with their condition. Therefore, conventional dosing regimens often lead to therapeutic failure. Another major hurdle faced in optimizing treatment for HAP/VAP is the reduction of the in vitro potency. Therapeutic drug monitoring (TDM), if available, may allow health care providers to personalize treatment to maximize efficacy of the drug exposures while minimizing toxicity. TDM can be of significant importance in populations whom PK are less defined and for resistant infections to achieve the best therapeutic outcome.
Collapse
Affiliation(s)
- Yasmeen P Abouelhassan
- Center for Anti-Infective Research and Development, Hartford Hospital, Hartford, Connecticut
| | - David Nicolau
- Center for Anti-Infective Research and Development, Hartford Hospital, Hartford, Connecticut.,Division of Infectious Diseases, Hartford Hospital, Hartford, Connecticut
| |
Collapse
|
29
|
Wu C, Zhang X, Xie J, Li Q, He J, Hu L, Wang H, Liu A, Xu J, Yang C, Yang Y, Qiu H, Huang Y. PK/PD parameters of linezolid in the epithelial lining fluid of patients with sepsis. J Clin Pharmacol 2022; 62:891-897. [PMID: 35049077 DOI: 10.1002/jcph.2031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 01/17/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Changde Wu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine Department of Critical Care Medicine Zhongda Hospital School of Medicine Southeast University Nanjing China
| | - Xiwen Zhang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine Department of Critical Care Medicine Zhongda Hospital School of Medicine Southeast University Nanjing China
| | - Jianfeng Xie
- Jiangsu Provincial Key Laboratory of Critical Care Medicine Department of Critical Care Medicine Zhongda Hospital School of Medicine Southeast University Nanjing China
| | - Qing Li
- Jiangsu Provincial Key Laboratory of Critical Care Medicine Department of Critical Care Medicine Zhongda Hospital School of Medicine Southeast University Nanjing China
| | - Jie He
- Department of Pharmacy Zhongda Hospital School of Medicine Southeast University Nanjing China
| | - Linlin Hu
- Department of Pharmacy Zhongda Hospital School of Medicine Southeast University Nanjing China
| | - Haofei Wang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine Department of Critical Care Medicine Zhongda Hospital School of Medicine Southeast University Nanjing China
| | - Airan Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine Department of Critical Care Medicine Zhongda Hospital School of Medicine Southeast University Nanjing China
| | - Jingyuan Xu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine Department of Critical Care Medicine Zhongda Hospital School of Medicine Southeast University Nanjing China
| | - Congshan Yang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine Department of Critical Care Medicine Zhongda Hospital School of Medicine Southeast University Nanjing China
| | - Yi Yang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine Department of Critical Care Medicine Zhongda Hospital School of Medicine Southeast University Nanjing China
| | - Haibo Qiu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine Department of Critical Care Medicine Zhongda Hospital School of Medicine Southeast University Nanjing China
| | - Yingzi Huang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine Department of Critical Care Medicine Zhongda Hospital School of Medicine Southeast University Nanjing China
| |
Collapse
|
30
|
Boisson M, Bouglé A, Sole-Lleonart C, Dhanani J, Arvaniti K, Rello J, Rouby JJ, Mimoz O. Nebulized Antibiotics for Healthcare- and Ventilator-Associated Pneumonia. Semin Respir Crit Care Med 2022; 43:255-270. [PMID: 35042259 DOI: 10.1055/s-0041-1740340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Global emergence of multidrug-resistant and extensive drug-resistant gram-negative bacteria has increased the risk of treatment failure, especially for healthcare- or ventilator-associated pneumonia (HAP/VAP). Nebulization of antibiotics, by providing high intrapulmonary antibiotic concentrations, represents a promising approach to optimize the treatment of HAP/VAP due to multidrug-resistant and extensive drug-resistant gram-negative bacteria, while limiting systemic antibiotic exposure. Aminoglycosides and colistin methanesulfonate are the most common nebulized antibiotics. Although optimal nebulized drug dosing regimen is not clearly established, high antibiotic doses should be administered using vibrating-mesh nebulizer with optimized ventilator settings to ensure safe and effective intrapulmonary concentrations. When used preventively, nebulized antibiotics reduced the incidence of VAP without any effect on mortality. This approach is not yet recommended and large randomized controlled trials should be conducted to confirm its benefit and explore the impact on antibiotic selection pressure. Compared with high-dose intravenous administration, high-dose nebulized colistin methanesulfonate seems to be more effective and safer in the treatment of ventilator-associated tracheobronchitis and VAP caused by multidrug resistant and extensive-drug resistant gram-negative bacteria. Adjunctive nebulized aminoglycosides could increase the clinical cure rate and bacteriological eradication in patients suffering from HAP/VAP due to multidrug-resistant and extensive drug-resistant gram-negative bacteria. As nebulized aminoglycosides broadly diffuse in the systemic circulation of patients with extensive bronchopneumonia, monitoring of plasma trough concentrations is recommended during the period of nebulization. Large randomized controlled trials comparing high dose of nebulized colistin methanesulfonate to high dose of intravenous colistin methanesulfonate or to intravenous new β-lactams in HAP/VAP due to multidrug-resistant and extensive drug-resistant gram-negative bacteria are urgently needed.
Collapse
Affiliation(s)
- Matthieu Boisson
- INSERM U1070, Université de Poitiers, UFR de Médecine Pharmacie, Poitiers, France.,Service de Prévention et de Contrôle de l'Infection, Hôpitaux Universitaires de Genève, Genève, Suisse
| | - Adrien Bouglé
- Medicine Sorbonne University, Anaesthesiology and Critical Care, Cardiology Institute, Paris, France.,Department of Anaesthesiology and Critical Care, La Pitié-Salpêtrière Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Candela Sole-Lleonart
- Intensive Care Unit, Consorci Hospitalari de Vic (CHV), The University of Vic - Central University of Catalonia (UVic-UCC), Vic, Barcelona, Spain
| | - Jayesh Dhanani
- Department of Intensive care medicine, Centre for Clinical Research, The University of Queensland, The Royal Brisbane and Women's Hospital Herston, Brisbane, Australia
| | - Kostoula Arvaniti
- Intensive Care Unit Department, Papageorgiou Hospital of Thessaloniki, Thessaloniki, Greece
| | - Jordi Rello
- Centro de Investigación Biomédica en Red (CIBERES), Instituto de Salud Carlos III, Madrid, Spain.,Clinical Research and Innovation in Pneumonia and Sepsis, Vall d'Hebron Institute of Research (VHIR), Barcelona, Spain.,Clinical Research, CHU Nîmes, Université Montpellier-Nîmes, Nîmes, France
| | - Jean-Jacques Rouby
- Department of Anaesthesiology and Critical Care, Medicine Sorbonne University, Multidisciplinary Intensive Care Unit, La Pitié Salpêtrière Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Olivier Mimoz
- INSERM U1070 Université de Poitiers, UFR de Médecine Pharmacie and Service des Urgences Adultes & SAMU 86, Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| | | |
Collapse
|
31
|
Aulin LBS, Tandar ST, van Zijp T, van Ballegooie E, van der Graaf PH, Saleh MAA, Välitalo P, van Hasselt JGC. Physiologically Based Modelling Framework for Prediction of Pulmonary Pharmacokinetics of Antimicrobial Target Site Concentrations. Clin Pharmacokinet 2022; 61:1735-1748. [PMID: 36401151 PMCID: PMC9676785 DOI: 10.1007/s40262-022-01186-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2022] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND OBJECTIVES Prediction of antimicrobial target-site pharmacokinetics is of relevance to optimize treatment with antimicrobial agents. A physiologically based pharmacokinetic (PBPK) model framework was developed for prediction of pulmonary pharmacokinetics, including key pulmonary infection sites (i.e. the alveolar macrophages and the epithelial lining fluid). METHODS The modelling framework incorporated three lung PBPK models: a general passive permeability-limited model, a drug-specific permeability-limited model and a quantitative structure-property relationship (QSPR)-informed perfusion-limited model. We applied the modelling framework to three fluoroquinolone antibiotics. Incorporation of experimental drug-specific permeability data was found essential for accurate prediction. RESULTS In the absence of drug-specific transport data, our QSPR-based model has generic applicability. Furthermore, we evaluated the impact of drug properties and pathophysiologically related changes on pulmonary pharmacokinetics. Pulmonary pharmacokinetics were highly affected by physiological changes, causing a shift in the main route of diffusion (i.e. paracellular or transcellular). Finally, we show that lysosomal trapping can cause an overestimation of cytosolic concentrations for basic compounds when measuring drug concentrations in cell homogenate. CONCLUSION The developed lung PBPK model framework constitutes a promising tool for characterization of pulmonary exposure of systemically administrated antimicrobials.
Collapse
Affiliation(s)
- Linda B. S. Aulin
- grid.5132.50000 0001 2312 1970Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Sebastian T. Tandar
- grid.5132.50000 0001 2312 1970Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Torben van Zijp
- grid.5132.50000 0001 2312 1970Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Etienne van Ballegooie
- grid.5132.50000 0001 2312 1970Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Piet H. van der Graaf
- grid.5132.50000 0001 2312 1970Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands ,Certara QSP, Canterbury, UK
| | - Mohammed A. A. Saleh
- grid.5132.50000 0001 2312 1970Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Pyry Välitalo
- grid.9668.10000 0001 0726 2490University of Eastern Finland, Kuopio, Finland ,grid.490668.50000 0004 0495 5912Finnish Medicines Agency, Kuopio, Finland
| | - J. G. Coen van Hasselt
- grid.5132.50000 0001 2312 1970Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| |
Collapse
|
32
|
Target Site Pharmacokinetics of Meropenem: Measurement in Human Explanted Lung Tissue by Bronchoalveolar Lavage, Microdialysis, and Homogenized Lung Tissue. Antimicrob Agents Chemother 2021; 65:e0156421. [PMID: 34570645 DOI: 10.1128/aac.01564-21] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pneumonia is one of the most common infections in intensive care patients, and it is often treated with beta-lactam antibiotics. Even if therapeutic drug monitoring in blood is available, it is unclear whether sufficient concentrations are reached at the target site: the lung. The present study was initiated to fill this knowledge gap. Various compartments from 10 patients' explanted lungs were subjected to laboratory analysis. Meropenem was quantified in serum, bronchoalveolar lavage (BAL) fluid, microdialysate, and homogenized lung tissue with isotope dilution liquid chromatography tandem mass spectrometry (ID-LC-MS/MS). BAL fluid represents diluted epithelial lining fluid (ELF), and microdialysate represents interstitial fluid (IF). Differences between target site and blood concentrations were investigated. The median meropenem concentration in blood, ELF, IF, and tissue were 26.8, 18.0, 12.1, and 9.1 mg/liter, respectively. A total of 37.5% of the target site ELF and IF meropenem concentrations were below the clinical EUCAST breakpoint of 8 mg/liter. The median ELF/serum quotient was 61.8% (interquartile range [IQR], 24.8% to 87.6%), the median IF/serum quotient was 35.4% (IQR, 23.8% to 54.3%), and the median tissue/serum quotient was 34.2% (IQR, 28.3% to 38.2%). We observed a substantial interindividual variability between the blood and the compartments (ELF and IF), whereas the intraindividual variability was relatively low. Target site measurement in different lung compartments was feasible and successfully applied in a clinical setting. A relevant amount of 37.5% of the target site concentrations were below the clinical EUCAST breakpoint, indicating subtherapeutic dosing in high-risk patients receiving perioperative antibiotic prophylaxis in lung transplantation. (This study has been registered at ClinicalTrials.gov under identifier NCT03970265.).
Collapse
|
33
|
Pasipanodya JG, Gumbo T. The Relationship Between Drug Concentration in Tuberculosis Lesions, Epithelial Lining Fluid, and Clinical Outcomes. Clin Infect Dis 2021; 73:e3374-e3376. [PMID: 32857152 DOI: 10.1093/cid/ciaa1271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 08/24/2020] [Indexed: 11/12/2022] Open
Affiliation(s)
| | - Tawanda Gumbo
- Praedicare Inc., Dallas, Texas, USA.,Department of Medicine, University of Cape Town, Observatory, South Africa
| |
Collapse
|
34
|
McCallum AD, Pertinez HE, Else LJ, Dilly-Penchala S, Chirambo AP, Sheha I, Chasweka M, Chitani A, Malamba RD, Meghji JZ, Gordon SB, Davies GR, Khoo SH, Sloan DJ, Mwandumba HC. Intrapulmonary Pharmacokinetics of First-line Anti-tuberculosis Drugs in Malawian Patients With Tuberculosis. Clin Infect Dis 2021; 73:e3365-e3373. [PMID: 32856694 PMCID: PMC8563277 DOI: 10.1093/cid/ciaa1265] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Further work is required to understand the intrapulmonary pharmacokinetics of first-line anti-tuberculosis drugs. This study aimed to describe the plasma and intrapulmonary pharmacokinetics of rifampicin, isoniazid, pyrazinamide, and ethambutol, and explore relationships with clinical treatment outcomes in patients with pulmonary tuberculosis. METHODS Malawian adults with a first presentation of microbiologically confirmed pulmonary tuberculosis received standard 6-month first-line therapy. Plasma and intrapulmonary samples were collected 8 and 16 weeks into treatment and drug concentrations measured in plasma, lung/airway epithelial lining fluid (ELF), and alveolar cells. Population pharmacokinetic modeling generated estimates of drug exposure (Cmax and AUC) from individual-level post hoc Bayesian estimates of plasma and intrapulmonary pharmacokinetics. RESULTS One-hundred fifty-seven patients (58% HIV coinfected) participated. Despite standard weight-based dosing, peak plasma concentrations of first-line drugs were below therapeutic drug-monitoring targets. Rifampicin concentrations were low in all 3 compartments. Isoniazid, pyrazinamide, and ethambutol achieved higher concentrations in ELF and alveolar cells than plasma. Isoniazid and pyrazinamide concentrations were 14.6-fold (95% CI, 11.2-18.0-fold) and 49.8-fold (95% CI, 34.2-65.3-fold) higher in ELF than plasma, respectively. Ethambutol concentrations were highest in alveolar cells (alveolar cell-plasma ratio, 15.0; 95% CI, 11.4-18.6). Plasma or intrapulmonary pharmacokinetics did not predict clinical treatment response. CONCLUSIONS We report differential drug concentrations between plasma and the lung. While plasma concentrations were below therapeutic monitoring targets, accumulation of drugs at the site of disease may explain the success of the first-line regimen. The low rifampicin concentrations observed in all compartments lend strong support for ongoing clinical trials of high-dose rifampicin regimens.
Collapse
Affiliation(s)
- Andrew D McCallum
- Malawi-Liverpool-Wellcome Clinical Research Programme, University of Malawi College of Medicine, Blantyre, Malawi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Henry E Pertinez
- Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Laura J Else
- Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Sujan Dilly-Penchala
- Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Aaron P Chirambo
- Malawi-Liverpool-Wellcome Clinical Research Programme, University of Malawi College of Medicine, Blantyre, Malawi
| | - Irene Sheha
- Malawi-Liverpool-Wellcome Clinical Research Programme, University of Malawi College of Medicine, Blantyre, Malawi
| | - Madalitso Chasweka
- Malawi-Liverpool-Wellcome Clinical Research Programme, University of Malawi College of Medicine, Blantyre, Malawi
| | - Alex Chitani
- Malawi-Liverpool-Wellcome Clinical Research Programme, University of Malawi College of Medicine, Blantyre, Malawi
| | - Rose D Malamba
- Malawi-Liverpool-Wellcome Clinical Research Programme, University of Malawi College of Medicine, Blantyre, Malawi
| | - Jamilah Z Meghji
- Malawi-Liverpool-Wellcome Clinical Research Programme, University of Malawi College of Medicine, Blantyre, Malawi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Stephen B Gordon
- Malawi-Liverpool-Wellcome Clinical Research Programme, University of Malawi College of Medicine, Blantyre, Malawi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Geraint R Davies
- Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Saye H Khoo
- Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Derek J Sloan
- School of Medicine, University of St Andrews, St Andrews, United Kingdom
| | - Henry C Mwandumba
- Malawi-Liverpool-Wellcome Clinical Research Programme, University of Malawi College of Medicine, Blantyre, Malawi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| |
Collapse
|
35
|
Awad M, Yosri M, Abdel-Aziz MM, Younis AM, Sidkey NM. Assessment of the Antibacterial Potential of Biosynthesized Silver Nanoparticles Combined with Vancomycin Against Methicillin-Resistant Staphylococcus aureus-Induced Infection in Rats. Biol Trace Elem Res 2021; 199:4225-4236. [PMID: 33389618 DOI: 10.1007/s12011-020-02561-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 12/21/2020] [Indexed: 10/22/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is considered one of the most serious multidrug-resistant bacteria worldwide. MRSA resistance to methicillin antibiotics made vancomycin, the acceptable treatment option. Silver nanoparticles (Ag-NPs) are among the well-known antibacterial substances showing multimode antibacterial action. Therefore, Ag-NPs are appropriate applicants for use in combination with vancomycin in order to augment its antibacterial action. This study aimed to biosynthesize silver nanoparticles and to evaluate its antibacterial activity against MRSA alone and when combined with vancomycin both in vitro and in vivo. Agaricus bisporus is used to reduce the silver nitrate salts in solution to yield silver nanoparticles which was characterized by UV-visible spectrophotometric analysis that shows maximum absorption at 420 nm as a preliminary confirmation for nanoparticles synthesis, Energy-Dispersive Analysis of X-ray (EDX) which confirms the crystalline nature of silver nanoparticles and transmission electron microscopy (TEM) image shows the particles in spherical form with mean size 27.45 nm. The synthesized silver nanoparticles were tested for antibacterial activity against MRSA, and the synergetic effects of the combination of silver nanoparticles and vancomycin were evaluated. The results showed a strong synergistic antibacterial effect between Ag-NPs and vancomycin in vitro with fractional inhibitory concentration 0.37 and in vivo against MRSA strain. The result revealed that mycosynthesized silver nanoparticles (NPs) enhance the in vitro and in vivo antibacterial activity of vancomycin against MRSA. These results suggested that sliver nanoparticles have an effective antibacterial activity against MRSA count, histopathology, and liver enzymes as well as protective immune response specially when combined with vancomycin in the lungs of infected rats with MRSA.
Collapse
Affiliation(s)
- Mohammed Awad
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo, 11841, Egypt
| | - Mohamed Yosri
- The Regional Center for Mycology and Biotechnology, Al-Azhar University, Nasr City, Cairo, 11787, Egypt.
| | - Marwa M Abdel-Aziz
- The Regional Center for Mycology and Biotechnology, Al-Azhar University, Nasr City, Cairo, 11787, Egypt
| | - Ahmed M Younis
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo, 11841, Egypt
| | - Nagwa M Sidkey
- Botany and Microbiology Department, Faculty of Science (Girls), Al-Azhar University, Nasr City, Cairo, Egypt
| |
Collapse
|
36
|
Tanner L, Mashabela GT, Omollo CC, de Wet TJ, Parkinson CJ, Warner DF, Haynes RK, Wiesner L. Intracellular Accumulation of Novel and Clinically Used TB Drugs Potentiates Intracellular Synergy. Microbiol Spectr 2021; 9:e0043421. [PMID: 34585951 PMCID: PMC8557888 DOI: 10.1128/spectrum.00434-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 08/19/2021] [Indexed: 12/13/2022] Open
Abstract
The therapeutic repertoire for tuberculosis (TB) remains limited despite the existence of many TB drugs that are highly active in in vitro models and possess clinical utility. Underlying the lack of efficacy in vivo is the inability of TB drugs to penetrate microenvironments inhabited by the causative agent, Mycobacterium tuberculosis, including host alveolar macrophages. Here, we determined the ability of the phenoxazine PhX1 previously shown to be active against M. tuberculosis in vitro to differentially penetrate murine compartments, including plasma, epithelial lining fluid, and isolated epithelial lining fluid cells. We also investigated the extent of permeation into uninfected and M. tuberculosis-infected human macrophage-like Tamm-Horsfall protein 1 (THP-1) cells directly and by comparing to results obtained in vitro in synergy assays. Our data indicate that PhX1 (4,750 ± 127.2 ng/ml) penetrates more effectively into THP-1 cells than do the clinically used anti-TB agents, rifampin (3,050 ± 62.9 ng/ml), moxifloxacin (3,374 ± 48.7 ng/ml), bedaquiline (4,410 ± 190.9 ng/ml), and linezolid (770 ± 14.1 ng/ml). Compound efficacy in infected cells correlated with intracellular accumulation, reinforcing the perceived importance of intracellular penetration as a key drug property. Moreover, we detected synergies deriving from redox-stimulatory combinations of PhX1 or clofazimine with the novel prenylated amino-artemisinin WHN296. Finally, we used compound synergies to elucidate the relationship between compound intracellular accumulation and efficacy, with PhX1/WHN296 synergy levels shown to predict drug efficacy. Collectively, our data support the utility of the applied assays in identifying in vitro active compounds with the potential for clinical development. IMPORTANCE This study addresses the development of novel therapeutic compounds for the eventual treatment of drug-resistant tuberculosis. Tuberculosis continues to progress, with cases of Mycobacterium tuberculosis (M. tuberculosis) resistance to first-line medications increasing. We assess new combinations of drugs with both oxidant and redox properties coupled with a third partner drug, with the focus here being on the potentiation of M. tuberculosis-active combinations of compounds in the intracellular macrophage environment. Thus, we determined the ability of the phenoxazine PhX1, previously shown to be active against M. tuberculosis in vitro, to differentially penetrate murine compartments, including plasma, epithelial lining fluid, and isolated epithelial lining fluid cells. In addition, the extent of permeation into human macrophage-like THP-1 cells and H37Rv-infected THP-1 cells was measured via mass spectrometry and compared to in vitro two-dimensional synergy and subsequent intracellular efficacy. Collectively, our data indicate that development of new drugs will be facilitated using the methods described herein.
Collapse
Affiliation(s)
- Lloyd Tanner
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Gabriel T. Mashabela
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Charles C. Omollo
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Timothy J. de Wet
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | | | - Digby F. Warner
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa
| | - Richard K. Haynes
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
| | - Lubbe Wiesner
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
37
|
Katsube T, Nicolau DP, Rodvold KA, Wunderink RG, Echols R, Matsunaga Y, Menon A, Portsmouth S, Wajima T. Intrapulmonary pharmacokinetic profile of cefiderocol in mechanically ventilated patients with pneumonia. J Antimicrob Chemother 2021; 76:2902-2905. [PMID: 34383901 PMCID: PMC8521398 DOI: 10.1093/jac/dkab280] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/08/2021] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES Lung penetration of cefiderocol, a novel siderophore cephalosporin approved for treatment of nosocomial pneumonia, has previously been evaluated in healthy subjects. This study assessed the intrapulmonary pharmacokinetic profile of cefiderocol at steady state in hospitalized, mechanically ventilated pneumonia patients. METHODS Patients received cefiderocol 2 g (or ≤1.5 g if renally impaired), administered IV q8h as a 3 h infusion, or 2 g q6h if patients had augmented renal function (estimated CLCR > 120 mL/min). After multiple doses, each patient underwent a single bronchoalveolar lavage (BAL) procedure either at the end of the infusion or at 2 h after the end of infusion. Plasma samples were collected at 1, 3, 5 and 7 h after the start of infusion. After correcting for BAL dilution, cefiderocol concentrations in epithelial lining fluid (ELF) for each patient and the ELF/unbound plasma concentration ratio (RC, E/P) were calculated. Safety was assessed up to 7 days after the last cefiderocol dose. RESULTS Seven patients received cefiderocol. Geometric mean ELF concentration of cefiderocol was 7.63 mg/L at the end of infusion and 10.40 mg/L at 2 h after the end of infusion. RC, E/P was 0.212 at the end of infusion and 0.547 at 2 h after the end of infusion, suggesting delayed lung distribution. There were no adverse drug reactions. CONCLUSIONS The results suggest that cefiderocol penetrates the ELF in critically ill pneumonia patients with concentrations that are sufficient to treat Gram-negative bacteria with an MIC of ≤4 mg/L.
Collapse
Affiliation(s)
| | - David P Nicolau
- Centre for Anti-Infective Research & Development, Hartford
Hospital, Hartford, CT, USA
| | - Keith A Rodvold
- College of Pharmacy, University of Illinois at Chicago, Chicago,
IL, USA
| | - Richard G Wunderink
- Division of Pulmonary and Critical Care Medicine, Northwestern University
Feinberg School of Medicine, Chicago, IL, USA
| | - Roger Echols
- Infectious Disease Drug Development Consulting, LLC, Easton, CT,
USA
| | | | | | | | | |
Collapse
|
38
|
Humphries H, Almond L, Berg A, Gardner I, Hatley O, Pan X, Small B, Zhang M, Jamei M, Romero K. Development of physiologically-based pharmacokinetic models for standard of care and newer tuberculosis drugs. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2021; 10:1382-1395. [PMID: 34623770 PMCID: PMC8592506 DOI: 10.1002/psp4.12707] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/12/2021] [Accepted: 08/22/2021] [Indexed: 12/19/2022]
Abstract
Tuberculosis (TB) remains a global health problem and there is an ongoing effort to develop more effective therapies and new combination regimes that can reduce duration of treatment. The purpose of this study was to demonstrate utility of a physiologically‐based pharmacokinetic modeling approach to predict plasma and lung concentrations of 11 compounds used or under development as TB therapies (bedaquiline [and N‐desmethyl bedaquiline], clofazimine, cycloserine, ethambutol, ethionamide, isoniazid, kanamycin, linezolid, pyrazinamide, rifampicin, and rifapentine). Model accuracy was assessed by comparison of simulated plasma pharmacokinetic parameters with healthy volunteer data for compounds administered alone or in combination. Eighty‐four percent (area under the curve [AUC]) and 91% (maximum concentration [Cmax]) of simulated mean values were within 1.5‐fold of the observed data and the simulated drug‐drug interaction ratios were within 1.5‐fold (AUC) and twofold (Cmax) of the observed data for nine (AUC) and eight (Cmax) of the 10 cases. Following satisfactory recovery of plasma concentrations in healthy volunteers, model accuracy was assessed further (where patients’ with TB data were available) by comparing clinical data with simulated lung concentrations (9 compounds) and simulated lung: plasma concentration ratios (7 compounds). The 5th–95th percentiles for the simulated lung concentration data recovered between 13% (isoniazid and pyrazinamide) and 88% (pyrazinamide) of the observed data points (Am J Respir Crit Care Med, 198, 2018, 1208; Nat Med, 21, 2015, 1223; PLoS Med, 16, 2019, e1002773). The impact of uncertain model parameters, such as the fraction of drug unbound in lung tissue mass (fumass), is discussed. Additionally, the variability associated with the patient lung concentration data, which was sparse and included extensive within‐subject, interlaboratory, and experimental variability (as well interindividual variability) is reviewed. All presented models are transparently documented and are available as open‐source to aid further research.
Collapse
Affiliation(s)
| | - Lisa Almond
- Certara UK Limited, Simcyp Division, Sheffield, UK
| | | | - Iain Gardner
- Certara UK Limited, Simcyp Division, Sheffield, UK
| | | | - Xian Pan
- Certara UK Limited, Simcyp Division, Sheffield, UK
| | - Ben Small
- Certara UK Limited, Simcyp Division, Sheffield, UK
| | - Mian Zhang
- Certara UK Limited, Simcyp Division, Sheffield, UK
| | - Masoud Jamei
- Certara UK Limited, Simcyp Division, Sheffield, UK
| | | |
Collapse
|
39
|
Echeverria-Esnal D, Martín-Ontiyuelo C, Navarrete-Rouco ME, Barcelo-Vidal J, Conde-Estévez D, Carballo N, De-Antonio Cuscó M, Ferrández O, Horcajada JP, Grau S. Pharmacological management of antifungal agents in pulmonary aspergillosis: an updated review. Expert Rev Anti Infect Ther 2021; 20:179-197. [PMID: 34328373 DOI: 10.1080/14787210.2021.1962292] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Aspergillus may cause different types of lung infections: invasive, chronic pulmonary or allergic bronchopulmonary aspergillosis. Pharmacological management with antifungals poses as a challenge. Patients diagnosed with pulmonary aspergillosis are complex, as well as the problems associated with antifungal agents. AREAS COVERED This article reviews the pharmacology of antifungal agents in development and currently used to treat pulmonary aspergillosis, including the mechanisms of action, pharmacokinetics, pharmacodynamics, dosing, therapeutic drug monitoring and safety. Recommendations to manage situations that arise in daily clinical practice are provided. A literature search of PubMed was conducted on November 15th, 2020 and updated on March 30th, 2021. EXPERT OPINION Recent and relevant developments in the treatment of pulmonary aspergillosis have taken place. Novel antifungals with new mechanisms of action that extend antifungal spectrum and improve pharmacokinetic-related aspects, drug-drug interactions and safety are under current study. For those antifungals already marketed, new data related to pharmacokinetics, pharmacodynamics, dose adjustments in special situations, therapeutic drug monitoring and safety are available. To maximize efficacy and reduce the risk of associated toxicities, it is essential to choose the most appropriate antifungal; optimize its dose, interval, route of administration and length of treatment; and prevent side effects.
Collapse
Affiliation(s)
- Daniel Echeverria-Esnal
- Pharmacy Department, Hospital Del Mar, Parc De Salut Mar, Barcelona, Spain.,Infectious Pathology and Antimicrobials Research Group (IPAR), Institut Hospital Del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain
| | | | | | | | - David Conde-Estévez
- Pharmacy Department, Hospital Del Mar, Parc De Salut Mar, Barcelona, Spain.,Department Of Pharmacology, Universitat Autònoma De Barcelona, Barcelona, Spain
| | - Nuria Carballo
- Pharmacy Department, Hospital Del Mar, Parc De Salut Mar, Barcelona, Spain
| | | | - Olivia Ferrández
- Pharmacy Department, Hospital Del Mar, Parc De Salut Mar, Barcelona, Spain
| | - Juan Pablo Horcajada
- Infectious Pathology and Antimicrobials Research Group (IPAR), Institut Hospital Del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain.,Department Of Pharmacology, Universitat Autònoma De Barcelona, Barcelona, Spain.,Infectious Diseases Department, Hospital Del Mar, Parc De Salut Mar, Barcelona, Spain
| | - Santiago Grau
- Pharmacy Department, Hospital Del Mar, Parc De Salut Mar, Barcelona, Spain.,Infectious Pathology and Antimicrobials Research Group (IPAR), Institut Hospital Del Mar d'Investigacions Mèdiques (IMIM), Barcelona, Spain.,Department Of Pharmacology, Universitat Autònoma De Barcelona, Barcelona, Spain
| |
Collapse
|
40
|
López-Lorente CI, Awchi M, Sinues P, García-Gómez D. Real-time pharmacokinetics via online analysis of exhaled breath. J Pharm Biomed Anal 2021; 205:114311. [PMID: 34403867 DOI: 10.1016/j.jpba.2021.114311] [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: 05/19/2021] [Revised: 07/24/2021] [Accepted: 08/04/2021] [Indexed: 11/26/2022]
Abstract
The advantages that on-line breath analysis has shown in different fields have already made it stand as an interesting tool for pharmacokinetic studies. This review summarizes recent progress in the field, diving into the different analytical methods and the different advantages and hurdles encountered. We conclude that there is a wealth of limitations in the application of this technique, and key aspects like standardization are still outstanding. Nevertheless, this is an experimental field that has not yet been fully explored; and the advantages it offers for animal welfare, decrease in the amount of drug needed in experimental studies, and complementary insights to current pharmacological studies, warrant further exploration. Further studies are needed to overcome current limitations and incorporate this technique into the toolbox of pharmacological studies, both at an industrial and academic level.
Collapse
Affiliation(s)
| | - Mo Awchi
- University Children's Hospital Basel, University of Basel, Basel, Switzerland; Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Pablo Sinues
- University Children's Hospital Basel, University of Basel, Basel, Switzerland; Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Diego García-Gómez
- Department of Analytical Chemistry, University of Salamanca, Salamanca, Spain.
| |
Collapse
|
41
|
Abstract
Many adverse reactions to therapeutic drugs appear to be allergic in nature, and are thought to be triggered by patient-specific Immunoglobulin E (IgE) antibodies that recognize the drug molecules and form complexes with them that activate mast cells. However, in recent years another mechanism has been proposed, in which some drugs closely associated with allergic-type events can bypass the antibody-mediated pathway and trigger mast cell degranulation directly by activating a mast cell-specific receptor called Mas-related G protein-coupled receptor X2 (MRGPRX2). This would result in symptoms similar to IgE-mediated events, but would not require immune priming. This review will cover the frequency, severity, and dose-responsiveness of allergic-type events for several drugs shown to have MRGPRX2 agonist activity. Surprisingly, the analysis shows that mild-to-moderate events are far more common than currently appreciated. A comparison with plasma drug levels suggests that MRGPRX2 mediates many of these mild-to-moderate events. For some of these drugs, then, MRGPRX2 activation may be considered a regular and predictable feature after administration of high doses.
Collapse
Affiliation(s)
- Benjamin D. McNeil
- Division of Allergy and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
42
|
Guo Y, Bera H, Shi C, Zhang L, Cun D, Yang M. Pharmaceutical strategies to extend pulmonary exposure of inhaled medicines. Acta Pharm Sin B 2021; 11:2565-2584. [PMID: 34522598 PMCID: PMC8424368 DOI: 10.1016/j.apsb.2021.05.015] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/19/2021] [Accepted: 04/26/2021] [Indexed: 12/13/2022] Open
Abstract
Pulmonary administration route has been extensively exploited for the treatment of local lung diseases such as asthma, chronic obstructive pulmonary diseases and respiratory infections, and systemic diseases such as diabetes. Most inhaled medicines could be cleared rapidly from the lungs and their therapeutic effects are transit. The inhaled medicines with extended pulmonary exposure may not only improve the patient compliance by reducing the frequency of drug administration, but also enhance the clinical benefits to the patients with improved therapeutic outcomes. This article systematically reviews the physical and chemical strategies to extend the pulmonary exposure of the inhaled medicines. It starts with an introduction of various physiological and pathophysiological barriers for designing inhaled medicines with extended lung exposure, which is followed by recent advances in various strategies to overcome these barriers. Finally, the applications of the inhaled medicines with extended lung exposure for the treatment of various diseases and the safety concerns associated to various strategies to extend the pulmonary exposure of the inhaled medicines are summarized.
Collapse
Key Words
- ALIS, amikacin liposomal inhalation suspension
- API, active pharmaceutical ingredient
- BALF, bronchoalveolar lavage fluid
- COPD, chronic obstructive pulmonary diseases
- CS, chitosan
- DPIs, dry powder inhalers
- DPPC, dipalmitoylphosphatidylcholine
- DSPC, 1,2-distearoyl-sn-glycero-3-phosphocholine
- Da, aerodynamic diameters
- ELF, epithelial lining fluid
- FDA, US food and drug administration
- FDKP, fumaryl diketopiperazine
- HA, hyaluronic acid
- IL-4, interleukin-4
- IL-5, interleukin-5
- Inhaled sustained release formulations
- LABA, long-acting β2-adrenoceptor agonist
- LPPs, large porous particles
- Local lung diseases
- MCE, mucociliary escalator
- MDIs, metered dose inhalers
- MP, mucoadhesive particles
- MPP, mucus-penetrating particles
- MW, molecular weight
- Mn, number-average molecular weight
- NLCs, nanostructured lipid carriers
- PCL, poly-ε-caprolactone
- PDD, pulmonary drug delivery
- PEG, polyethylene glycol
- PK, pharmacokinetics
- PLA, polylactic acid
- PLGA, poly(lactic-co-glycolic acid)
- PVA, polyvinyl alcohol
- Pharmaceutical strategies
- Pulmonary clearance pathways
- Pulmonary drug delivery
- Pulmonary exposure
- Pulmonary safety
- SLNs, solid lipid nanoparticles
- Systemic diseases
- Tmax, time of maximum concentration
Collapse
Affiliation(s)
- Yi Guo
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hriday Bera
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Changzhi Shi
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Li Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Dongmei Cun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Corresponding author. Tel./fax: +86 24 23986165.
| | - Mingshi Yang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| |
Collapse
|
43
|
Ruth MM, Raaijmakers J, van den Hombergh E, Aarnoutse R, Svensson EM, Susanto BO, Simonsson USH, Wertheim H, Hoefsloot W, van Ingen J. Standard therapy of Mycobacterium avium complex pulmonary disease shows limited efficacy in an open source hollow fibre system that simulates human plasma and epithelial lining fluid pharmacokinetics. Clin Microbiol Infect 2021; 28:448.e1-448.e7. [PMID: 34332109 DOI: 10.1016/j.cmi.2021.07.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 07/04/2021] [Accepted: 07/08/2021] [Indexed: 11/03/2022]
Abstract
OBJECTIVES Mycobacterium avium complex (MAC) bacteria can cause chronic pulmonary disease (PD). Current treatment regimens of azithromycin, ethambutol and rifampicin have culture conversion rates of around 65%. Dynamic, preclinical models to assess the efficacy of treatment regimens are important to guide clinical trial development. The hollow fibre system (HFS) has been applied but reports lack experimental details. METHODS We simulated the human pharmacokinetics of azithromycin, ethambutol and rifampicin both in plasma and epithelial lining fluid (ELF) in a HFS, exposing THP-1 cells infected with M. avium to the triple-drug regimen for 3 weeks. We accounted for drug-drug interactions and protein-binding and provide all laboratory protocols. We differentiated the effects on the intracellular and extracellular mycobacterial population. RESULTS The antibiotic concentrations in the HFS accurately reflected the time to peak concentration (Tmax), the peak concentration (Cmax) and half-life of azithromycin, rifampicin and ethambutol in plasma and ELF reported in literature. We find that plasma drug concentrations fail to hold the MAC bacterial load static (ΔLog10 CFU/mLControl:Regimen = 0.66 ± 0.76 and 0.45 ± 0.28 at 3 and 21 days); ELF concentrations do hold the bacterial load static for 3 days and inhibit bacterial growth for the duration of the experiment (ΔLog10 CFU/mLControl:Regimen = 1.1 ± 0.1 and 1.64 ± 0.59 at 3 and 21 days). DISCUSSION In our model, the current therapy against MAC is ineffective, even when accounting for antibiotic accumulation at the site of infection and intracellularly. New treatment regimens need to be developed and be compared with currently recommended regimens in dynamic models prior to clinical evaluation. With the publication of all protocols we aim to open this technology to new users.
Collapse
Affiliation(s)
- Mike Marvin Ruth
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Jelmer Raaijmakers
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Erik van den Hombergh
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Rob Aarnoutse
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Elin M Svensson
- Radboudumc Center for Infectious Diseases, Department of Pharmacy, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Pharmacy, Uppsala University, Sweden
| | - Budi O Susanto
- Department of Pharmaceutical Biosciences, Uppsala University, Sweden
| | | | - Heiman Wertheim
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Wouter Hoefsloot
- Radboudumc Center for Infectious Diseases, Department of Pulmonary Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jakko van Ingen
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
44
|
Chalmers JD, van Ingen J, van der Laan R, Herrmann JL. Liposomal drug delivery to manage nontuberculous mycobacterial pulmonary disease and other chronic lung infections. Eur Respir Rev 2021; 30:30/161/210010. [PMID: 34289985 PMCID: PMC9488898 DOI: 10.1183/16000617.0010-2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 05/01/2021] [Indexed: 12/20/2022] Open
Abstract
Nontuberculous mycobacterial (NTM) pulmonary disease is a chronic respiratory infection associated with declining lung function, radiological deterioration and significantly increased morbidity and mortality. Patients often have underlying lung conditions, particularly bronchiectasis and COPD. NTM pulmonary disease is difficult to treat because mycobacteria can evade host defences and antimicrobial therapy through extracellular persistence in biofilms and sequestration into macrophages. Management of NTM pulmonary disease remains challenging and outcomes are often poor, partly due to limited penetration of antibiotics into intracellular spaces and biofilms. Efficient drug delivery to the site of infection is therefore a key objective of treatment, but there is high variability in lung penetration by antibiotics. Inhalation is the most direct route of delivery and has demonstrated increased efficacy of antibiotics like amikacin compared with systemic administration. Liposomes are small, artificial, enclosed spherical vesicles, in which drug molecules can be encapsulated to provide controlled release, with potentially improved pharmacokinetics and reduced toxicity. They are especially useful for drugs where penetration of cell membranes is essential. Inhaled delivery of liposomal drug solutions can therefore facilitate direct access to macrophages in the lung where the infecting NTM may reside. A range of liposomal drugs are currently being evaluated in respiratory diseases. Liposome-encapsulated antibiotics can optimise respiratory disease treatment. Amikacin liposomal inhalation suspension is effective in nontuberculous mycobacterial pulmonary disease that has failed to convert following oral guideline-based therapy.https://bit.ly/3f3ixIu
Collapse
Affiliation(s)
- James D Chalmers
- Scottish Centre for Respiratory Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Jakko van Ingen
- Dept of Medical Microbiology, Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Jean-Louis Herrmann
- Université Paris-Saclay, UVSQ, INSERM, Infection and Inflammation, Montigny-le-Bretonneux, France.,APHP, Groupe Hospitalo-Universitaire Paris-Saclay, Hôpital Raymond Poincaré, Garches, France
| |
Collapse
|
45
|
Exploration of Clinical Breakpoint of Danofloxacin for Glaesserella parasuis in Plasma and in PELF. Antibiotics (Basel) 2021; 10:antibiotics10070808. [PMID: 34356730 PMCID: PMC8300709 DOI: 10.3390/antibiotics10070808] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/26/2021] [Accepted: 06/29/2021] [Indexed: 02/02/2023] Open
Abstract
Background: In order to establish the clinical breakpoint (CBP) of danofloxacin against G. parasuis, three cutoff values, including epidemiological cutoff value (ECV), pharmacokinetic-pharmacodynamic (PK-PD) cutoff value (COPD) and clinical cutoff value (COCL), were obtained in the present study. Methods: The ECV was calculated using ECOFFinder base on the MIC distribution of danfloxacin against 347 G. parasuis collected from disease pigs. The COPD was established based on in vivo and ex vivo PK-PD modeling of danofloxacin both in plasma and pulmonary epithelial lining fluid (PELF) using Hill formula and Monte Carlo analysis. The COCL was established based on the relationship between the possibility of cure (POC) and MIC in the clinical trials using the "WindoW" approach, nonlinear regression and CART analysis. Results: The MIC50 and MIC90 of danofloxacin against 347 G. parasuis were 2 μg/mL and 8 μg/mL, respectively. The ECV value was set to 8 μg/mL using ECOFFinder. Concentration-time curves of danofloxacin were fitted with a two-compartment PK model. The PK parameters of the maximum concentration (Cmax) and area under concentration-time curves (AUC) in PELF were 3.67 ± 0.25 μg/mL and 24.28 ± 2.70 h·μg/mL, higher than those in plasma (0.67 ± 0.01 μg/mL and 4.47 ± 0.51 h·μg/mL). The peak time (Tmax) in plasma was 0.23 ± 0.07 h, shorter than that in PELF (1.61 ± 0.15 h). The COPD in plasma and PELF were 0.125 μg/mL and 0.5 μg/mL, respectively. The COCL calculated by WindoW approach, nonlinear regression and CART analysis were 0.125-4 μg/mL, 0.428 μg/mL and 0.56 μg/mL, respectively. The 0.5 μg/mL was selected as eligible COCL. The ECV is much higher than the COPD and COCL, and the clinical breakpoint based on data in plasma was largely different from that of PELF. Conclusions: Our study firstly established three cutoff values of danofloxacin against G. parasuis. It suggested that non-wild-type danofloxacin-resistant G. parasuis may lead to ineffective treatment by danofloxacin.
Collapse
|
46
|
Pharmacokinetics and pharmacodynamics of antibiotics in cystic fibrosis: a narrative review. Int J Antimicrob Agents 2021; 58:106381. [PMID: 34157401 DOI: 10.1016/j.ijantimicag.2021.106381] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/21/2021] [Accepted: 06/10/2021] [Indexed: 12/22/2022]
Abstract
Cystic fibrosis affects several organs, predisposing patients to severe bacterial respiratory infections, including those caused by methicillin-resistant Staphylococcus aureus. Cystic fibrosis is also associated with a wide spectrum of pathological changes that can significantly affect the absorption, distribution, metabolism, and/or elimination of several drugs, including antibacterial agents. Therefore, awareness of the pharmacokinetic derangements in patients with cystic fibrosis is mandatory for the optimisation of antibiotic therapy. This review discusses the basic principles of pharmacokinetics and the pathophysiology of the pharmacokinetics changes associated with cystic fibrosis; it also provides an update of available data for the most widely used antibiotics. Evidence accumulated in the last few years has clearly shown that a significant number of cystic fibrosis patients treated with conventional dosing schemes have sub-therapeutic antibiotic concentrations, increasing their risk of therapeutic failure and/or the emergence of resistant pathogens. Some proposals to optimise antibiotic therapies in this clinical setting based on therapeutic drug monitoring are also discussed.
Collapse
|
47
|
Schütz C, Ho D, Hamed MM, Abdelsamie AS, Röhrig T, Herr C, Kany AM, Rox K, Schmelz S, Siebenbürger L, Wirth M, Börger C, Yahiaoui S, Bals R, Scrima A, Blankenfeldt W, Horstmann JC, Christmann R, Murgia X, Koch M, Berwanger A, Loretz B, Hirsch AKH, Hartmann RW, Lehr C, Empting M. A New PqsR Inverse Agonist Potentiates Tobramycin Efficacy to Eradicate Pseudomonas aeruginosa Biofilms. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2004369. [PMID: 34165899 PMCID: PMC8224453 DOI: 10.1002/advs.202004369] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/21/2021] [Indexed: 05/21/2023]
Abstract
Pseudomonas aeruginosa (PA) infections can be notoriously difficult to treat and are often accompanied by the development of antimicrobial resistance (AMR). Quorum sensing inhibitors (QSI) acting on PqsR (MvfR) - a crucial transcriptional regulator serving major functions in PA virulence - can enhance antibiotic efficacy and eventually prevent the AMR. An integrated drug discovery campaign including design, medicinal chemistry-driven hit-to-lead optimization and in-depth biological profiling of a new QSI generation is reported. The QSI possess excellent activity in inhibiting pyocyanin production and PqsR reporter-gene with IC50 values as low as 200 and 11 × 10-9 m, respectively. Drug metabolism and pharmacokinetics (DMPK) as well as safety pharmacology studies especially highlight the promising translational properties of the lead QSI for pulmonary applications. Moreover, target engagement of the lead QSI is shown in a PA mucoid lung infection mouse model. Beyond that, a significant synergistic effect of a QSI-tobramycin (Tob) combination against PA biofilms using a tailor-made squalene-derived nanoparticle (NP) formulation, which enhance the minimum biofilm eradicating concentration (MBEC) of Tob more than 32-fold is demonstrated. The novel lead QSI and the accompanying NP formulation highlight the potential of adjunctive pathoblocker-mediated therapy against PA infections opening up avenues for preclinical development.
Collapse
|
48
|
Tanaka K, Vu H, Hayashi M. In vitro activities and spectrum of lascufloxacin(KRP-AM1977)against anaerobes. J Infect Chemother 2021; 27:1265-1269. [PMID: 33867268 DOI: 10.1016/j.jiac.2021.03.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/26/2021] [Accepted: 03/30/2021] [Indexed: 10/21/2022]
Abstract
The in vitro antibacterial spectra and activities of five antimicrobial agents, including lascufloxacin (LSFX) and two quinolones, were investigated against 69 species of anaerobes in 31 genera and 188 strains in 9 genera, respectively. In this study, minimum inhibitory concentrations (MICs) of lascufloxacin against the reference strains associated with respiratory and head and neck infections. LSFX inhibited the growth of 33 gram-positive and gram-negative reference strains at ≤0.015-2 μg/mL, except for Leptotrichia buccalis. MICs ranges of LSFX against the clinical isolates of 44 Porphyromonas spp., 45 Prevotella spp., 25 Fusobacterium spp., 7 Leptotrichia spp., 25 Parvimonas micra, 25 other gram-positive anaerobic cocci, and 17 Veillonella spp., were ≤0.015-4, 0.125-4, 0.06-0.5, 2, 0.25-16, ≤0.015-2, ≤0.015-16 μg/mL, respectively. LSFX demonstrated potent antibacterial efficacy against a wide range of species isolated from specimens involved in respiratory as well as head and neck infections.
Collapse
Affiliation(s)
- Kaori Tanaka
- Division of Anaerobe Research, Life Science Research Center, Gifu University, Gifu, Japan; United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu, Japan.
| | - Hanh Vu
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu, Japan
| | - Masahiro Hayashi
- Division of Anaerobe Research, Life Science Research Center, Gifu University, Gifu, Japan
| |
Collapse
|
49
|
Heffernan AJ, Sime FB, Lim SMS, Naicker S, Andrews KT, Ellwood D, Lipman J, Grimwood K, Roberts JA. Impact of the Epithelial Lining Fluid Milieu on Amikacin Pharmacodynamics Against Pseudomonas aeruginosa. Drugs R D 2021; 21:203-215. [PMID: 33797739 PMCID: PMC8017437 DOI: 10.1007/s40268-021-00344-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2021] [Indexed: 12/14/2022] Open
Abstract
Background Even though nebulised administration of amikacin can achieve high epithelial lining fluid concentrations, this has not translated into improved patient outcomes in clinical trials. One possible reason is that the cellular and chemical composition of the epithelial lining fluid may inhibit amikacin-mediated bacterial killing. Objective The objective of this study was to identify whether the epithelial lining fluid components inhibit amikacin-mediated bacterial killing. Methods Two amikacin-susceptible (minimum inhibitory concentrations of 2 and 8 mg/L) Pseudomonas aeruginosa isolates were exposed in vitro to amikacin concentrations up to 976 mg/L in the presence of an acidic pH, mucin and/or surfactant as a means of simulating the epithelial lining fluid, the site of bacterial infection in pneumonia. Pharmacodynamic modelling was used to describe associations between amikacin concentrations, bacterial killing and emergence of resistance. Results In the presence of broth alone, there was rapid and extensive (> 6 − log10) bacterial killing, with emergence of resistance identified in amikacin concentrations < 976 mg/L. In contrast, the rate and extent of bacterial killing was reduced (≤ 5 − log10) when exposed to an acidic pH and mucin. Surfactant did not appreciably impact the bacterial killing or resistance emergence when compared with broth alone for either isolate. The combination of mucin and an acidic pH further reduced the rate of bacterial killing, with the maximal bacterial killing occurring 24 h following initial exposure compared with approximately 4–8 h for either mucin or an acidic pH alone. Conclusions Our findings indicate that simulating the epithelial lining fluid antagonises amikacin-mediated killing of P. aeruginosa, even at the high concentrations achieved following nebulised administration. Supplementary Information The online version contains supplementary material available at 10.1007/s40268-021-00344-5.
Collapse
Affiliation(s)
- Aaron J Heffernan
- School of Medicine, Griffith University, Gold Coast, QLD, Australia. .,Centre for Translational Anti-infective Pharmacodynamics, School of Pharmacy, The University of Queensland, Cornwall St, Woolloongabba, QLD, 4102, Australia.
| | - Fekade B Sime
- Centre for Translational Anti-infective Pharmacodynamics, School of Pharmacy, The University of Queensland, Cornwall St, Woolloongabba, QLD, 4102, Australia.,Faculty of Medicine, University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia
| | - Sazlyna Mohd Sazlly Lim
- Centre for Translational Anti-infective Pharmacodynamics, School of Pharmacy, The University of Queensland, Cornwall St, Woolloongabba, QLD, 4102, Australia.,Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Saiyuri Naicker
- Centre for Translational Anti-infective Pharmacodynamics, School of Pharmacy, The University of Queensland, Cornwall St, Woolloongabba, QLD, 4102, Australia.,Faculty of Medicine, University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia
| | - Katherine T Andrews
- Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - David Ellwood
- School of Medicine, Griffith University, Gold Coast, QLD, Australia.,Gold Coast Health, Southport, QLD, Australia
| | - Jeffrey Lipman
- Department of Intensive Care Medicine, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia.,Division of Anaesthesiology Critical Care Emergency and Pain Medicine, Nîmes University Hospital, University of Montpellier, Nîmes, France
| | - Keith Grimwood
- School of Medicine, Griffith University, Gold Coast, QLD, Australia.,Gold Coast Health, Southport, QLD, Australia
| | - Jason A Roberts
- Centre for Translational Anti-infective Pharmacodynamics, School of Pharmacy, The University of Queensland, Cornwall St, Woolloongabba, QLD, 4102, Australia.,Faculty of Medicine, University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia.,Department of Intensive Care Medicine, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia.,Division of Anaesthesiology Critical Care Emergency and Pain Medicine, Nîmes University Hospital, University of Montpellier, Nîmes, France
| |
Collapse
|
50
|
Almangour TA, Garcia E, Zhou Q, Forrest A, Kaye KS, Li J, Velkov T, Rao GG. Polymyxins for the treatment of lower respiratory tract infections: lessons learned from the integration of clinical pharmacokinetic studies and clinical outcomes. Int J Antimicrob Agents 2021; 57:106328. [PMID: 33785362 DOI: 10.1016/j.ijantimicag.2021.106328] [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: 11/13/2020] [Revised: 02/18/2021] [Accepted: 03/20/2021] [Indexed: 11/26/2022]
Abstract
The global rise in nosocomial pneumonia caused by multidrug-resistant (MDR) Gram-negative pathogens and the increasingly limited antibiotic treatment options are growing threats to modern medicine. As a result, older antibiotics such as polymyxins are being used as last-resort drugs for MDR nosocomial pneumonia. Polymyxins are bactericidal against most aerobic Gram-negative bacilli. High-dose intravenous (IV) adminsitration of polymyxins, however, results in subtherapeutic concentrations at the site of infection making treatment challenging. Alternative forms of polymyxin delivery have been considered in order to better achieve the necessary concentrations at the site of infection. Several studies have evaluated the effectiveness of aerosolised polymyxins in patients with nosocomial pneumonia caused by MDR Gram-negative pathogens such as Pseudomonas aeruginosa, Acinetobacter baumannii and Klebsiella pneumoniae. Here we evaluated the pharmacokinetic data supporting the use of inhaled polymyxins in nosocomial pneumonia and provide insight into the limitations and challenges that future studies should address. We have also reviewed the literature published between 2006 and 2020 on the use of aerosolised polymyxins for the treatment of nosocomial pneumonia, including ventilator-associated pneumonia, in patients without cystic fibrosis to evaluate their safety and efficacy as monotherapy or as an adjunct to IV antimicrobials. This review highlights the need for well-designed multicentre studies with standardised methodologies to further evaluate the effectiveness of inhaled polymyxins and to provide reliable pharmacokinetic/pharmacodynamic data in order to redefine appropriate dosing strategies.
Collapse
Affiliation(s)
- Thamer A Almangour
- Department of Pharmacotherapy and Experimental Therapeutics, University of North Carolina at Chapel Hill, Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA; Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Estefany Garcia
- Department of Pharmacotherapy and Experimental Therapeutics, University of North Carolina at Chapel Hill, Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA
| | - Qi Zhou
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana, USA
| | - Alan Forrest
- Department of Pharmacotherapy and Experimental Therapeutics, University of North Carolina at Chapel Hill, Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA
| | - Keith S Kaye
- Department of Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jian Li
- Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Melbourne, Victoria, Australia
| | - Tony Velkov
- Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Gauri G Rao
- Department of Pharmacotherapy and Experimental Therapeutics, University of North Carolina at Chapel Hill, Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA.
| |
Collapse
|