1
|
Yaliniz A, Blouin M, Métras MÉ, Boulanger MC, Cloutier K, Dubé MH, Autmizguine J, Marsot A. Vancomycin in Pediatric Patients with Cystic Fibrosis: Dose Optimization Using Population Pharmacokinetic Approach. Eur J Drug Metab Pharmacokinet 2024; 49:677-687. [PMID: 39283469 DOI: 10.1007/s13318-024-00913-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2024] [Indexed: 11/09/2024]
Abstract
BACKGROUND An increase in Staphylococcus aureus infections has been reported in pediatric patients with cystic fibrosis (CF) over the last few years. This pathogen is commonly treated with vancomycin, an antibiotic for which therapeutic drug monitoring (TDM) is recommended. Updated guidelines were recently published regarding new targets of exposure for the TDM of vancomycin through a Bayesian approach, using population pharmacokinetic (popPK) models. OBJECTIVES This study aims to assess the predictive performance of vancomycin popPK models in pediatric patients with CF and to recommend optimal initial dosing regimens based on simulations. METHODS Patient data were collected from two centers in Canada, and a literature review was conducted to identify all published vancomycin popPK models for pediatric CF patients. External evaluation and simulations were performed according to patient and occasion of treatment. RESULTS A total of 53 vancomycin concentrations were collected from six pediatric CF patients. Only two popPK models of vancomycin for pediatric CF patients were identified through the literature review. The external evaluation results for both centers combined revealed a population bias of 28.1% and an imprecision of 33.7%. A re-estimation of parameters was performed to improve predictive performance. The optimal initial dosing regimen was 15 mg/kg/dose administered every 6 hours according to the per occasion remodel. CONCLUSION The predictive performance and identified optimal initial dosing regimens associated with the model were different depending on the data used, showing external evaluation's importance before implementing a model in clinical practice.
Collapse
Affiliation(s)
- Aysenur Yaliniz
- STP2 Laboratory, Faculty of Pharmacy, Université de Montréal, 2940 Chemin de Polytechnique, Montreal, QC, H3T 1J4, Canada.
- Faculty of Pharmacy, Université de Montréal, Montreal, Canada.
| | - Mathieu Blouin
- STP2 Laboratory, Faculty of Pharmacy, Université de Montréal, 2940 Chemin de Polytechnique, Montreal, QC, H3T 1J4, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, Canada
| | - Marie-Élaine Métras
- Faculty of Pharmacy, Université de Montréal, Montreal, Canada
- Department of Pharmacy, Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
| | - Marie-Christine Boulanger
- Faculty of Pharmacy, Université Laval, Quebec, Canada
- Department of Pharmacy, CHU de Québec-Université Laval, Quebec, Canada
| | - Karine Cloutier
- Faculty of Pharmacy, Université Laval, Quebec, Canada
- Department of Pharmacy, CHU de Québec-Université Laval, Quebec, Canada
| | - Marie-Hélène Dubé
- Faculty of Pharmacy, Université Laval, Quebec, Canada
- Department of Pharmacy, CHU de Québec-Université Laval, Quebec, Canada
| | - Julie Autmizguine
- Department of Pharmacology, Faculty of Medicine, Université de Montréal, Montreal, Canada
- Clinical Pharmacology Unit, Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
- Research center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
| | - Amélie Marsot
- STP2 Laboratory, Faculty of Pharmacy, Université de Montréal, 2940 Chemin de Polytechnique, Montreal, QC, H3T 1J4, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, Canada
- Research center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
| |
Collapse
|
2
|
Struiken STL, Lobée D, van Tuinen EL, Touw DJ, van der Vaart H, Bourgonje AR, Rottier BL, Koppelman GH, Mian P. Evaluation of Target Attainment for Tobramycin in Children and Adults with Cystic Fibrosis. J Clin Med 2024; 13:2641. [PMID: 38731170 PMCID: PMC11084493 DOI: 10.3390/jcm13092641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Introduction: Patients with cystic fibrosis (CF) commonly experience pulmonary exacerbations, and it is recommended by the TOPIC study to treat this with tobramycin at a dose of 10 mg/kg once daily. The aim of this study was to evaluate the target attainment of the current dosing regimen. Methods: A single-center retrospective cohort study of child and adult patients with CF who received tobramycin between 2019 and 2022 was conducted. Descriptive statistics and linear mixed models were used to assess target attainment for tobramycin. Results: In total, 25 patients (53 courses), of which 10 were children (12 courses) and 15 were adults (41 courses), were included. Those 25 patients all received 10 mg/kg/day. The tobramycin peak concentrations were supratherapeutic in 82.9% and therapeutic in 100.0% of adults and children, respectively. The trough concentrations were outside the target range in 0% and 5.1% of children and adults, respectively. We found lower tobramycin concentrations with the same dose in children compared to adults. Conclusions: This study illustrates the need to validate dosing advice in a real-world setting, as supratherapeutic concentrations of tobramycin were prevalent in adults with CF.
Collapse
Affiliation(s)
- Sheseira T. L. Struiken
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, 9712 GZ Groningen, The Netherlands; (S.T.L.S.); (D.L.); (E.L.v.T.); (D.J.T.)
| | - Danique Lobée
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, 9712 GZ Groningen, The Netherlands; (S.T.L.S.); (D.L.); (E.L.v.T.); (D.J.T.)
| | - Eline L. van Tuinen
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, 9712 GZ Groningen, The Netherlands; (S.T.L.S.); (D.L.); (E.L.v.T.); (D.J.T.)
| | - Daniel J. Touw
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, 9712 GZ Groningen, The Netherlands; (S.T.L.S.); (D.L.); (E.L.v.T.); (D.J.T.)
- Department of Pharmaceutical Analysis, Groningen Research Institute for Pharmacy, University of Groningen, 9712 GZ Groningen, The Netherlands
| | - Hester van der Vaart
- Department of Pulmonary Diseases and Tuberculosis, University Medical Center Groningen, University of Groningen, 9712 GZ Groningen, The Netherlands;
| | - Arno R. Bourgonje
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9712 GZ Groningen, The Netherlands;
- Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bart L. Rottier
- Department of Pediatric Pulmonology and Pediatric Allergology, University Medical Center Groningen, Beatrix Children’s Hospital, University of Groningen, 9712 GZ Groningen, The Netherlands; (B.L.R.); (G.H.K.)
- Department of Pediatric Pulmonology, University Medical Center Groningen, University of Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), 9712 GZ Groningen, The Netherlands
| | - Gerard H. Koppelman
- Department of Pediatric Pulmonology and Pediatric Allergology, University Medical Center Groningen, Beatrix Children’s Hospital, University of Groningen, 9712 GZ Groningen, The Netherlands; (B.L.R.); (G.H.K.)
- Department of Pediatric Pulmonology, University Medical Center Groningen, University of Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), 9712 GZ Groningen, The Netherlands
| | - Paola Mian
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, 9712 GZ Groningen, The Netherlands; (S.T.L.S.); (D.L.); (E.L.v.T.); (D.J.T.)
| |
Collapse
|
3
|
Rakhshan A, Farahbakhsh N, Khanbabaee G, Tabatabaii SA, Sadr S, Hassanzad M, Sistanizad M, Dastan F, Hajipour M, Bahadori AR, Mirrahimi B. Evaluating the efficacy of inhaled amikacin as an adjunct to intravenous combination therapy (ceftazidime and amikacin) in pediatric cystic fibrosis pulmonary exacerbation. Front Pharmacol 2023; 14:1130374. [PMID: 36969859 PMCID: PMC10034196 DOI: 10.3389/fphar.2023.1130374] [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: 12/23/2022] [Accepted: 02/22/2023] [Indexed: 03/29/2023] Open
Abstract
Background: Pseudomonas aeruginosa is the most common microorganism found in the sputum culture of Cystic fibrosis (CF) patients causing the pulmonary destruction. Aminoglycosides have a low diffuse rate from lipid membranes, and respiratory system secretions. Regarding the burden of pulmonary exacerbation caused by the pseudomonas aeruginosa in cystic fibrosis patients in the long term and the limited number of clinical trials focused on appropriate treatment strategies, the present study evaluated the concurrent inhaled and intravenous aminoglycoside antibiotics for pulmonary exacerbation caused by the pseudomonas aeruginosa as a safe and effective treatment in children. Method: This study was a blinded, randomized clinical trial phase conducted in a tertiary referral pediatric teaching hospital from May 2021 to May 2022. The patients were randomly allocated to receive intravenously administered ceftazidime and Amikacin alone or with inhaled Amikacin. Forced expiratory volume (FEV1), Amikacin via the level, kidney function tests, audiometry, inflammatory markers (erythrocyte sedimentation rate and C-reactive protein), hospital stay, and bacterial eradication rate were compared in two therapy groups. Results: the average FEV1 has increased by 47% in Neb + group compared to Neb- group following treatment. Hospital stay was lower in Neb + group. No renal toxicity or ototoxicity was observed in both therapy groups. Pseudomonas aeruginosa eradication rate Neb- and Neb + groups were 44% and 69%, respectively (p-value = 0.15). Conclusion: Concurrent inhaled and intravenous Amikacin is safe and effective to treat Pseudomonas aeruginosa exacerbation in CF patients. Moreover, co-delivery antibiotics' route treatment increased the eradication rate. Although not statistically significant, never the less, it is clinically relevant. The intervention reduced the length of hospitalization in this group. Clinical Trial Registration: clinicaltrials.gov, identifier [IRCT20120415009475N10].
Collapse
Affiliation(s)
- Amin Rakhshan
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nazanin Farahbakhsh
- Department of Pediatric Pulmonology, Mofid Children’s Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ghamartaj Khanbabaee
- Department of Pediatric Pulmonology, Mofid Children’s Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Ahmad Tabatabaii
- Department of Pediatric Pulmonology, Mofid Children’s Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeed Sadr
- Department of Pediatric Pulmonology, Mofid Children’s Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Hassanzad
- Pediatric Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Sistanizad
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farzaneh Dastan
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahmoud Hajipour
- Children’s Gastroenterology, Liver and Nutrition Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Reza Bahadori
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bahador Mirrahimi
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- *Correspondence: Bahador Mirrahimi,
| |
Collapse
|
4
|
Imburgia TA, Seagren RM, Christensen H, Lasarev MR, Bogenschutz MC. Review of Tobramycin Dosing in Pediatric Patients With Cystic Fibrosis. J Pediatr Pharmacol Ther 2023; 28:63-70. [PMID: 36777977 PMCID: PMC9901314 DOI: 10.5863/1551-6776-28.1.63] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 02/08/2022] [Indexed: 02/05/2023]
Abstract
OBJECTIVE An institution's tobramycin pharmacokinetics (PK) database was reviewed to evaluate the efficacy and safety of empiric tobramycin dosing and monitoring strategies used in pediatric patients with cystic fibrosis (CF). The relationship between patient age and tobramycin dosing needed to achieve the area under the curve (AUC) goal was investigated. METHODS Retrospective chart review was performed for patients who received tobramycin during a CF exacerbation from 2009 to 2019 who received PK monitoring by pediatric pharmacists. Tobramycin dosing needed to achieve an AUC of 100 mg·hr/L was calculated for each patient. Serum creatinine and concomitant nephrotoxin use were collected as surrogate nephrotoxicity endpoints to evaluate safety. RESULTS Goal AUC (100 ± 15 mg·hr/L) was achieved based on initial or repeat PK calculations in 43.5% (95% CI, 37.7-49.3) of 85 unique patients across 326 encounters. Patients with calculated recommended doses of 9.5 to 11.9 mg/kg every 24 hours empirically achieved goal AUC in 77% (78/101) of encounters. The odds of achieving goal AUC were 56% higher for children aged 10 vs 5 years (OR = 1.56; 95% CI, 1.04-2.34; p = 0.033) and 32% higher for children aged 15 vs 10 years (OR = 1.32; 95% CI, 1.07-1.61; p = 0.008). Overall rates of acute kidney injury and concomitant nephrotoxin use were 10.8% (95% CI, 6.2-15.5) and 80.7% (95% CI, 74.3-87.1), respectively. CONCLUSIONS Desired AUC was achieved by 43.5% of pediatric patients with CF using tobramycin 10 mg/kg every 24 hours. Older patient age was associated with higher initial AUC attainment and fewer dose modifications. Younger children may require higher weight-based dosing to meet AUC goals.
Collapse
Affiliation(s)
- Taylor A. Imburgia
- Department of Pharmacy (TAI, RMS, HC, MCB), American Family Children's Hospital at University of Wisconsin Health, Madison, WI
| | - Ryan M. Seagren
- Department of Pharmacy (TAI, RMS, HC, MCB), American Family Children's Hospital at University of Wisconsin Health, Madison, WI
| | - Hanna Christensen
- Department of Pharmacy (TAI, RMS, HC, MCB), American Family Children's Hospital at University of Wisconsin Health, Madison, WI
| | - Michael R. Lasarev
- Department of Biostatistics and Medical Informatics (MRL), University of Wisconsin–Madison, Madison, WI
| | - Monica C. Bogenschutz
- Department of Pharmacy (TAI, RMS, HC, MCB), American Family Children's Hospital at University of Wisconsin Health, Madison, WI
| |
Collapse
|
5
|
Fogliano C, Motta CM, Avallone B. Salicylate attenuates gentamicin-induced ototoxicity and facilitates the recovery in the basilar papilla of the lizard Podarcis siculus. Neurotoxicology 2022; 93:301-310. [DOI: 10.1016/j.neuro.2022.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/24/2022] [Accepted: 10/24/2022] [Indexed: 11/15/2022]
|
6
|
Lloyd EC, Cogen JD, Maples H, Bell SC, Saiman L. Antimicrobial Stewardship in Cystic Fibrosis. J Pediatric Infect Dis Soc 2022; 11:S53-S61. [PMID: 36069899 DOI: 10.1093/jpids/piac071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 07/06/2022] [Indexed: 12/14/2022]
Abstract
The chronic airway infection and inflammation characteristic of cystic fibrosis (CF) ultimately leads to progressive lung disease, the primary cause of death in persons with CF (pwCF). Despite many recent advances in CF clinical care, efforts to preserve lung function in many pwCF still necessitate frequent antimicrobial use. Incorporating antimicrobial stewardship (AMS) principles into management of pulmonary exacerbations (PEx) would facilitate development of best practices for antimicrobial utilization at CF care centers. However, AMS can be challenging in CF given the unique aspects of chronic, polymicrobial infection in the CF airways, lack of evidence-based guidelines for managing PEx, limited utility for antimicrobial susceptibility testing, and increased frequency of adverse drug events in pwCF. This article describes current evidence-based antimicrobial treatment strategies for pwCF, highlights the potential for AMS to beneficially impact CF care, and provides practical strategies for integrating AMS programs into the management of PEx in pwCF.
Collapse
Affiliation(s)
- Elizabeth C Lloyd
- Department of Pediatrics, Division of Pediatric Infectious Diseases, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jonathan D Cogen
- Department of Pediatrics, Division of Pulmonary and Sleep Medicine, Seattle Children's Hospital, Seattle, Washington, USA
| | - Holly Maples
- Department of Pharmacy Practice, University of Arkansas for Medical Sciences College of Pharmacy, Little Rock, Arkansas, USA.,Quality and Safety Division, Arkansas Children's, Little Rock, Arkansas, USA
| | - Scott C Bell
- Department of Thoracic Medicine, The Prince Charles Hospital, Brisbane, Queensland, Australia.,Children's Health Research Centre, The University of Queensland, Brisbane, Queensland, Australia.,Translational Research Institute, Brisbane, Queensland, Australia
| | - Lisa Saiman
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA.,Department of Infection Prevention and Control, NewYork-Presbyterian Hospital, New York, New York, USA
| |
Collapse
|
7
|
Grant JJ, McDade EJ, Zobell JT, Young DC. The indispensable role of pharmacy services and medication therapy management in cystic fibrosis. Pediatr Pulmonol 2022; 57 Suppl 1:S17-S39. [PMID: 34347382 DOI: 10.1002/ppul.25613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/24/2021] [Accepted: 08/02/2021] [Indexed: 11/06/2022]
Abstract
Care for people with cystic fibrosis (PWCF) is highly complex and requires a multidisciplinary approach where the pharmacist plays a vital role. The purpose of this manuscript is to serve as a guideline for pharmacists and pharmacy technicians who provide care for PWCF by providing background and current recommendations for the use of cystic fibrosis (CF)-specific medications in both the acute and ambulatory care settings. The article explores current literature surrounding the role of pharmacists and pharmacy technicians, proven pharmacy models to emulate, and pharmacokinetic idiosyncrasies unique to the CF population while also identifying areas of future research. Clinical recommendations for the use of CF-specific medications are broken down by organ system including mechanism of action, adverse events, dosages, and monitoring parameters. The article also includes quick reference tables essential to the acute and chronic medication therapy management of PWCF.
Collapse
Affiliation(s)
- Jonathan J Grant
- Department of Outpatient Pharmacy-Specialty Services, The John's Hopkins Hospital, Baltimore, Maryland, USA
| | - Erin J McDade
- Pharmacy Department, Texas Children's Hospital, Houston, Texas, USA
| | - Jeffery T Zobell
- Pharmacy Department, Intermountain Primary Children's Hospital, Salt Lake City, Utah, USA
| | - David C Young
- Department of Pharmacotherapy, University of Utah College of Pharmacy, Salt Lake City, Utah, USA
| |
Collapse
|
8
|
Ochs MA, Dillman NO, Caverly LJ, Chaffee VD. Aminoglycoside dosing and monitoring for Pseudomonas aeruginosa during acute pulmonary exacerbations in cystic fibrosis. Pediatr Pulmonol 2021; 56:3634-3643. [PMID: 33983680 DOI: 10.1002/ppul.25441] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/28/2021] [Accepted: 04/14/2021] [Indexed: 11/11/2022]
Abstract
Aminoglycosides are commonly used for the treatment of Pseudomonas aeruginosa (PsA) in the setting of acute pulmonary exacerbations (PEx) in pediatric patients with cystic fibrosis (CF). There are controversies and practice differences between institutions related to aminoglycoside dosing and monitoring strategies. The purpose of this review article is to summarize the currently available literature and identify gaps in the literature related to pharmacokinetic parameter goals, aminoglycoside dosing strategies, and methods for monitoring serum aminoglycoside concentrations for treatment of PsA in CF PEx, and throughout will discuss anticipated changes with the increasing availability of highly effective CF transmembrane conductance regulator modulators. This review focuses on tobramycin, as it is the most commonly used aminoglycoside in CF PEx, and will briefly discuss special circumstances surrounding use of amikacin and gentamicin.
Collapse
Affiliation(s)
- Madeleine A Ochs
- Department of Pharmacy Services and Clinical Pharmacy, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Nicholas O Dillman
- Department of Pharmacy Services and Clinical Pharmacy, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Lindsay J Caverly
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Valerie D Chaffee
- Department of Pharmacy, Cancer and Hematology Centers of West Michigan, Grand Rapids, Michigan, USA
| |
Collapse
|
9
|
Landmesser KB, Autry EB, Gardner BM, Bosko KA, Schadler A, Kuhn RJ. Comparison of the predictive value of area under the curve versus maximum serum concentration of intravenous tobramycin in cystic fibrosis patients treated for an acute pulmonary exacerbation. Pediatr Pulmonol 2021; 56:3209-3216. [PMID: 34241975 DOI: 10.1002/ppul.25569] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 04/19/2021] [Accepted: 06/21/2021] [Indexed: 12/29/2022]
Abstract
OBJECTIVES The primary objective of this study was to compare the therapeutic predictive value of area under the curve (AUC24 ) versus maximum concentration (Cmax ) in cystic fibrosis (CF) patients receiving intravenous (IV) tobramycin for a Pseudomonas aeruginosa (PsA) acute pulmonary exacerbation (APE). Acute kidney injury (AKI) incidence and the relationship between time undetectable and efficacy were also assessed. METHODS A retrospective review was conducted in patients aged at least 1 month with a diagnosis of CF receiving IV tobramycin for treatment of a PsA APE and admitted to the University of Kentucky between August 2015 and August 2019. Patients were excluded if they had no growth of PsA on sputum culture or if two postdose tobramycin levels were not obtained following a dose adjustment of ≥20%. RESULTS A total of 44 pediatric and 107 adult patient encounters met inclusion criteria. In patients with therapeutic success (n = 91), 75.8% had an AUC24 ≥80% and 80.3% had a Cmax ≥8 times the highest PsA minimal inhibitory concentration. There was a significant correlation between AUC24 and Cmax (r[149] = 0.727; p < 0.001). AKI incidence was significantly higher in patients receiving IV tobramycin dosed multiple times daily versus at least every 24 h (χ2 [1, 151] = 3.9; p = 0.047). CONCLUSIONS The results of this study indicate that both AUC24 and Cmax serve as relatively accurate predictors of tobramycin efficacy. Additionally, given the significant increase in incidence of AKI, multidaily dosing of IV tobramycin should be avoided in pediatric and adult patients with CF.
Collapse
Affiliation(s)
| | - Elizabeth B Autry
- Department of Pharmacy, Kentucky Children's Hospital, Lexington, Kentucky, USA.,Department of Pharmacy, The University of Kentucky College of Pharmacy, Lexington, Kentucky, USA
| | - Brian M Gardner
- Department of Pharmacy, Kentucky Children's Hospital, Lexington, Kentucky, USA.,Department of Pharmacy, The University of Kentucky College of Pharmacy, Lexington, Kentucky, USA
| | - Katherine A Bosko
- Department of Pharmacy, The University of Kentucky College of Pharmacy, Lexington, Kentucky, USA
| | - Aric Schadler
- Department of Pharmacy, The University of Kentucky College of Pharmacy, Lexington, Kentucky, USA.,Department of Pediatrics, Kentucky Children's Hospital, Lexington, Kentucky, USA
| | - Robert J Kuhn
- Department of Pharmacy, Kentucky Children's Hospital, Lexington, Kentucky, USA.,Department of Pharmacy, The University of Kentucky College of Pharmacy, Lexington, Kentucky, USA
| |
Collapse
|
10
|
Woillard JB, Bouchet S, Fayon M, Marquet P, Monchaud C, Bui S. A Population Pharmacokinetic Modeling Approach to Determine the Efficacy of Intravenous Amikacin in Children with Cystic Fibrosis. Ther Drug Monit 2021; 43:499-504. [PMID: 33346630 DOI: 10.1097/ftd.0000000000000855] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 11/25/2020] [Indexed: 11/26/2022]
Abstract
BACKGROUND In children with cystic fibrosis (CF), the currently recommended amikacin dose ranges between 30 and 35 mg/kg/d; however, data supporting this dosing efficacy are lacking. In this article, the objectives were to develop a nonparametric pharmacokinetic population model for amikacin in children with CF and investigate the efficacy and toxicity at different dose rates for distinct minimum inhibitory concentration (MIC) clinical breakpoints using Monte Carlo simulations. METHODS Data from 94 children with CF (613 serum concentrations) from the Bordeaux University Hospital's CF-centre were analyzed. After determination of nonparametric pharmacokinetic population model parameters and associated influent covariates in Pmetrics, 1000 Monte Carlo simulations were performed for 7 different dose rates between 30 and 60 mg/kg/d, to predict the probability of obtaining peak serum amikacin ≥10 × MIC and trough level ≤2.5 mg/L, for MIC values between 1 and 16 mg/L. RESULTS The median (min-max) age and weight were 10 (0.3-17) years and 29 (6-71) kg, respectively, with only 2 children younger than 1 year of age. Body weight and creatinine clearance significantly impacted the amikacin volume of distribution and clearance. The mean relative bias/root mean squared error between observed and individual predicted concentrations was -0.68%/8.1%. Monte Carlo simulations showed that for sensitive bacteria with MICs ≤ 4, 30 mg/kg/d was most appropriate for a 100% success rate; for bacteria with MICs ≥ 8 [eg, Pseudomonas aeruginosa (MICamikacin = 8)], a dose of at least 40 mg/kg/d allowed a high success probability (90%), with a trough level below 2.5 mg/L. CONCLUSIONS For intermediate pathogens, a dose of at least 40 mg/kg/d can improve efficacy, with an acceptable calculated residual trough level in cases of normal or hyperfiltration. Because amikacin undergoes renal clearance, which is immature until 1 year of age, dosing recommendations for this age group may be markedly high, warranting cautious interpretation.
Collapse
Affiliation(s)
- Jean-Baptiste Woillard
- Department of Pharmacology and Toxicology, CHU Limoges
- IPPRITT, Université de Limoges
- INSERM, IPPRITT, U1248, Limoges
| | - Stéphane Bouchet
- CHU de Bordeaux, Hôpital Pellegrin, Service de Pharmacologie et Toxicologie
| | - Michael Fayon
- CHU de Bordeaux, Hôpital Pellegrin-Enfants, CRCM Pédiatrique and
- Université de Bordeaux, INSERM, Centre de Recherche Cardio-thoracique de Bordeaux (U1045), Centre d'Investigation Clinique (CIC1401), Bordeaux, France
| | - Pierre Marquet
- Department of Pharmacology and Toxicology, CHU Limoges
- IPPRITT, Université de Limoges
- INSERM, IPPRITT, U1248, Limoges
| | - Caroline Monchaud
- Department of Pharmacology and Toxicology, CHU Limoges
- IPPRITT, Université de Limoges
- INSERM, IPPRITT, U1248, Limoges
| | - Stéphanie Bui
- CHU de Bordeaux, Hôpital Pellegrin-Enfants, CRCM Pédiatrique and
| |
Collapse
|
11
|
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: 3.5] [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
|
12
|
Soren O, Rineh A, Silva DG, Cai Y, Howlin RP, Allan RN, Feelisch M, Davies JC, Connett GJ, Faust SN, Kelso MJ, Webb JS. Cephalosporin nitric oxide-donor prodrug DEA-C3D disperses biofilms formed by clinical cystic fibrosis isolates of Pseudomonas aeruginosa. J Antimicrob Chemother 2021; 75:117-125. [PMID: 31682251 PMCID: PMC6910178 DOI: 10.1093/jac/dkz378] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/16/2019] [Accepted: 08/02/2019] [Indexed: 11/30/2022] Open
Abstract
Objectives The cephalosporin nitric oxide (NO)-donor prodrug DEA-C3D (‘DiEthylAmin-Cephalosporin-3′-Diazeniumdiolate’) has been shown to initiate the dispersal of biofilms formed by the Pseudomonas aeruginosa laboratory strain PAO1. In this study, we investigated whether DEA-C3D disperses biofilms formed by clinical cystic fibrosis (CF) isolates of P. aeruginosa and its effect in combination with two antipseudomonal antibiotics, tobramycin and colistin, in vitro. Methods β-Lactamase-triggered release of NO from DEA-C3D was confirmed using a gas-phase chemiluminescence detector. MICs for P. aeruginosa clinical isolates were determined using the broth microdilution method. A crystal violet staining technique and confocal laser scanning microscopy were used to evaluate the effects of DEA-C3D on P. aeruginosa biofilms alone and in combination with tobramycin and colistin. Results DEA-C3D was confirmed to selectively release NO in response to contact with bacterial β-lactamase. Despite lacking direct, cephalosporin/β-lactam-based antibacterial activity, DEA-C3D was able to disperse biofilms formed by three P. aeruginosa clinical isolates. Confocal microscopy revealed that DEA-C3D in combination with tobramycin produces similar reductions in biofilm to DEA-C3D alone, whereas the combination with colistin causes near complete eradication of P. aeruginosa biofilms in vitro. Conclusions DEA-C3D is effective in dispersing biofilms formed by multiple clinical isolates of P. aeruginosa and could hold promise as a new adjunctive therapy to patients with CF.
Collapse
Affiliation(s)
- Odel Soren
- National Biofilms Innovation Centre, University of Southampton, Southampton SO17 1BJ, UK.,Biological Sciences and Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Ardeshir Rineh
- Molecular Horizons and School of Chemistry & Molecular Bioscience, University of Wollongong, NSW, 2522, Australia.,Illawarra Health & Medical Research Institute, Wollongong, NSW, 2522, Australia
| | - Diogo G Silva
- National Biofilms Innovation Centre, University of Southampton, Southampton SO17 1BJ, UK.,Faculty of Medicine and Institute for Life Sciences, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Yuming Cai
- National Biofilms Innovation Centre, University of Southampton, Southampton SO17 1BJ, UK.,Biological Sciences and Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Robert P Howlin
- Biological Sciences and Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK.,NIHR Southampton Clinical Research Facility and NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation, Southampton SO16 6YD, UK
| | - Raymond N Allan
- National Biofilms Innovation Centre, University of Southampton, Southampton SO17 1BJ, UK.,Biological Sciences and Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK.,NIHR Southampton Clinical Research Facility and NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation, Southampton SO16 6YD, UK
| | - Martin Feelisch
- Faculty of Medicine and Institute for Life Sciences, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Jane C Davies
- Cystic Fibrosis Trust Strategic Research Centre and National Heart and Lung Institute, Imperial College London, London SW3 6LY, UK
| | - Gary J Connett
- National Biofilms Innovation Centre, University of Southampton, Southampton SO17 1BJ, UK.,Faculty of Medicine and Institute for Life Sciences, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK.,NIHR Southampton Clinical Research Facility and NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation, Southampton SO16 6YD, UK
| | - Saul N Faust
- National Biofilms Innovation Centre, University of Southampton, Southampton SO17 1BJ, UK.,Faculty of Medicine and Institute for Life Sciences, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK.,NIHR Southampton Clinical Research Facility and NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation, Southampton SO16 6YD, UK
| | - Michael J Kelso
- Molecular Horizons and School of Chemistry & Molecular Bioscience, University of Wollongong, NSW, 2522, Australia.,Illawarra Health & Medical Research Institute, Wollongong, NSW, 2522, Australia
| | - Jeremy S Webb
- National Biofilms Innovation Centre, University of Southampton, Southampton SO17 1BJ, UK.,Biological Sciences and Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK.,NIHR Southampton Clinical Research Facility and NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation, Southampton SO16 6YD, UK
| |
Collapse
|
13
|
Miller T, Pastuch C, Garavaglia L, Gannon K, Parravani A. Unknown Renal Impairment: A Rare Case of Inhaled Tobramycin Induced Acute Kidney Injury in a Cystic Fibrosis Patient. Antibiotics (Basel) 2021; 10:antibiotics10040424. [PMID: 33921466 PMCID: PMC8070657 DOI: 10.3390/antibiotics10040424] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/09/2021] [Accepted: 04/10/2021] [Indexed: 11/16/2022] Open
Abstract
Acute kidney injury is a reversible medical condition commonly caused by nephrotoxic agents. The infrequency that a nebulized medication elicits a renal insult presents a rare diagnostic challenge. Within this case, we report a 57-year-old cystic fibrosis patient with chronic kidney disease (CKD) Stage G3b (baseline 1.5–1.6 mg/dL) who developed an acute kidney injury (AKI) with a serum creatinine elevation to 4.08 mg/dL and associated worsening vestibular dysfunction related to twice-daily nebulized tobramycin inhalation solution (TIS). The patient was found to have a tobramycin serum level of 4.2 μg/mL 2.5 h after TIS dosing, with elevation remaining present at 1.1 μg/mL 24 h after discontinuation of therapy. Laboratory values at one month continued to show elevated creatinine levels at 2.1 mg/dL, suggesting progression of his baseline CKD. This case supports the benefit of obtaining tobramycin serum levels and vestibular/audiology function testing when evaluating patients on chronic nebulized TIS who present with acute or chronic renal dysfunction. From these serum levels, adjustments to daily dosing, regular monitoring of tobramycin serum levels, or discontinuation of treatment should be made to prevent permanent renal damage in patients with CKD. Calculated Naranjo ADR Probability Scale: 9; Definite.
Collapse
Affiliation(s)
- Tyler Miller
- Department of Medicine, West Virginia University, Morgantown, WV 26506, USA
- Correspondence:
| | - Cristina Pastuch
- Department of Pediatrics and Medicine, West Virginia University, Morgantown, WV 26506, USA;
- Adult Cystic Fibrosis, Mountain State Cystic Fibrosis Center, West Virginia University, Morgantown, WV 26506, USA
| | - Lisa Garavaglia
- Department of Pharmaceutical Services, West Virginia University, Morgantown, WV 26506, USA;
| | - Kelley Gannon
- Section of General Internal Medicine, Department of Medicine, West Virginia University, Morgantown, WV 26506, USA;
| | - Anthony Parravani
- Section of Nephrology, Department of Medicine, West Virginia University, Morgantown, WV 26506, USA;
| |
Collapse
|
14
|
Pharmacokinetic and Pharmacodynamic Optimization of Antibiotic Therapy in Cystic Fibrosis Patients: Current Evidences, Gaps in Knowledge and Future Directions. Clin Pharmacokinet 2021; 60:409-445. [PMID: 33486720 DOI: 10.1007/s40262-020-00981-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2020] [Indexed: 10/22/2022]
Abstract
Antibiotic therapy is one of the main treatments for cystic fibrosis (CF). It aims to eradicate bacteria during early infection, calms down the inflammatory process, and leads to symptom resolution of pulmonary exacerbations. CF can modify both the pharmacokinetic (PK) and pharmacodynamic (PD) profiles of antibiotics, therefore specific PK/PD endpoints should be determined in the context of CF. Currently available data suggest that optimal PK/PD targets cannot be attained in sputum with intravenous aminoglycosides. Continuous infusion appears preferable for β-lactam antibiotics, but optimal concentrations in sputum are unlikely to be reached, with some possible exceptions such as meropenem and ceftolozane. Usual doses are likely suboptimal for fluoroquinolones and linezolid, whereas daily doses of 45-60 mg/kg and 200 mg could be convenient for vancomycin and doxycycline, respectively. Weekly azithromycin doses of 22-30 mg/kg could also be appropriate for its anti-inflammatory effect. The difficulty with achieving optimal concentrations supports the use of combined treatments and the inhaled administration route, as very high local concentrations, concomitantly with low systemic exposure, can be obtained with the inhaled route for aminoglycosides, colistin, and fluoroquinolones, thus minimizing the risk of toxicity.
Collapse
|
15
|
Antibiotics in Adult Cystic Fibrosis Patients: A Review of Population Pharmacokinetic Analyses. Clin Pharmacokinet 2021; 60:447-470. [PMID: 33447944 DOI: 10.1007/s40262-020-00970-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2020] [Indexed: 12/26/2022]
Abstract
BACKGROUND Lower respiratory tract infections are common in adult patients with cystic fibrosis (CF) and are frequently caused by Pseudomonas aeruginosa, resulting in chronic lung inflammation and fibrosis. The progression of multidrug-resistant strains of P. aeruginosa and alterations in the pharmacokinetics of many antibiotics in CF make optimal antimicrobial therapy a challenge, as reflected by high between- and inter-individual variability (IIV). OBJECTIVES This review provides a synthesis of population pharmacokinetic models for various antibiotics prescribed in adult CF patients, and aims at identifying the most reported structural models, covariates and sources of variability influencing the dose-concentration relationship. METHODS A literature search was conducted using the PubMed database, from inception to August 2020, and articles were retained if they met the inclusion/exclusion criteria. RESULTS A total of 19 articles were included in this review. One-, two- and three-compartment models were reported to best describe the pharmacokinetics of various antibiotics. The most common covariates were lean body mass and creatinine clearance. After covariate inclusion, the IIV (range) in total body clearance was 27.2% (10.40-59.7%) and 25.9% (18.0-33.9%) for β-lactams and aminoglycosides, respectively. IIV in total body clearance was estimated at 36.3% for linezolid and 22.4% for telavancin. The IIV (range) in volume of distribution was 29.4% (8.8-45.9%) and 15.2 (11.6-18.0%) for β-lactams and aminoglycosides, respectively, and 26.9% for telavancin. The median (range) of residual variability for all studies, using a combined (proportional and additive) model, was 12.7% (0.384-30.80%) and 0.126 mg/L (0.007-1.88 mg/L), respectively. CONCLUSION This is the first review that highlights key aspects of different population pharmacokinetic models of antibiotics prescribed in adult CF patients, effectively proposing relevant information for clinicians and researchers to optimize antibiotic therapy in CF.
Collapse
|
16
|
Expression of the MexXY Aminoglycoside Efflux Pump and Presence of an Aminoglycoside-Modifying Enzyme in Clinical Pseudomonas aeruginosa Isolates Are Highly Correlated. Antimicrob Agents Chemother 2020; 65:AAC.01166-20. [PMID: 33046496 PMCID: PMC7927871 DOI: 10.1128/aac.01166-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/15/2020] [Indexed: 12/26/2022] Open
Abstract
The impact of MexXY efflux pump expression on aminoglycoside resistance in clinical Pseudomonas aeruginosa isolates has been debated. In this study, we found that, in general, elevated mexXY gene expression levels in clinical P. aeruginosa isolates confer to slight increases in aminoglycoside MIC levels; however, those levels rarely lead to clinically relevant resistance phenotypes. The main driver of resistance in the clinical isolates studied here was the acquisition of aminoglycoside-modifying enzymes (AMEs). The impact of MexXY efflux pump expression on aminoglycoside resistance in clinical Pseudomonas aeruginosa isolates has been debated. In this study, we found that, in general, elevated mexXY gene expression levels in clinical P. aeruginosa isolates confer to slight increases in aminoglycoside MIC levels; however, those levels rarely lead to clinically relevant resistance phenotypes. The main driver of resistance in the clinical isolates studied here was the acquisition of aminoglycoside-modifying enzymes (AMEs). Nevertheless, acquisition of an AME was strongly associated with mexY overexpression. In line with this observation, we demonstrate that the introduction of a gentamicin acetyltransferase confers to full gentamicin resistance levels in a P. aeruginosa type strain only if the MexXY efflux pump was active. We discuss that increased mexXY activity in clinical AME-harboring P. aeruginosa isolates might affect ion fluxes at the bacterial cell membrane and thus might play a role in the establishment of enhanced fitness that extends beyond aminoglycoside resistance.
Collapse
|
17
|
Velino C, Carella F, Adamiano A, Sanguinetti M, Vitali A, Catalucci D, Bugli F, Iafisco M. Nanomedicine Approaches for the Pulmonary Treatment of Cystic Fibrosis. Front Bioeng Biotechnol 2019; 7:406. [PMID: 31921811 PMCID: PMC6927921 DOI: 10.3389/fbioe.2019.00406] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 11/27/2019] [Indexed: 12/24/2022] Open
Abstract
Cystic fibrosis (CF) is a genetic disease affecting today nearly 70,000 patients worldwide and characterized by a hypersecretion of thick mucus difficult to clear arising from the defective CFTR protein. The over-production of the mucus secreted in the lungs, along with its altered composition and consistency, results in airway obstruction that makes the lungs susceptible to recurrent and persistent bacterial infections and endobronchial chronic inflammation, which are considered the primary cause of bronchiectasis, respiratory failure, and consequent death of patients. Despite the difficulty of treating the continuous infections caused by pathogens in CF patients, various strategies focused on the symptomatic therapy have been developed during the last few decades, showing significant positive impact on prognosis. Moreover, nowadays, the discovery of CFTR modulators as well as the development of gene therapy have provided new opportunity to treat CF. However, the lack of effective methods for delivery and especially targeted delivery of therapeutics specifically to lung tissues and cells limits the efficiency of the treatments. Nanomedicine represents an extraordinary opportunity for the improvement of current therapies and for the development of innovative treatment options for CF previously considered hard or impossible to treat. Due to the peculiar environment in which the therapies have to operate characterized by several biological barriers (pulmonary tract, mucus, epithelia, bacterial biofilm) the use of nanotechnologies to improve and enhance drug delivery or gene therapies is an extremely promising way to be pursued. The aim of this review is to revise the currently used treatments and to outline the most recent progresses about the use of nanotechnology for the management of CF.
Collapse
Affiliation(s)
- Cecilia Velino
- Institute of Science and Technology for Ceramics (ISTEC), National Research Council (CNR), Faenza, Italy
| | - Francesca Carella
- Institute of Science and Technology for Ceramics (ISTEC), National Research Council (CNR), Faenza, Italy
| | - Alessio Adamiano
- Institute of Science and Technology for Ceramics (ISTEC), National Research Council (CNR), Faenza, Italy
| | - Maurizio Sanguinetti
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Dipartimento di Scienze di Laboratorio e Infettivologiche, Rome, Italy
- Istituto di Microbiologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alberto Vitali
- Institute for the Chemistry of Molecular Recognition (ICRM), National Research Council (CNR), c/o Institute of Biochemistry and Clinical Biochemistry, Catholic University, Rome, Italy
| | - Daniele Catalucci
- Humanitas Clinical and Research Center, Rozzano, Italy
- Institute of Genetic and Biomedical Research (IRGB) - UOS Milan, National Research Council (CNR), Milan, Italy
| | - Francesca Bugli
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Dipartimento di Scienze di Laboratorio e Infettivologiche, Rome, Italy
- Istituto di Microbiologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Michele Iafisco
- Institute of Science and Technology for Ceramics (ISTEC), National Research Council (CNR), Faenza, Italy
| |
Collapse
|
18
|
Thirion DJG, Pasche V, Marsot A. What is the recommended amikacin dosing for cystic fibrosis patients with acute pulmonary exacerbations? Pediatr Pulmonol 2019; 54:1652-1653. [PMID: 31313524 DOI: 10.1002/ppul.24453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 06/30/2019] [Indexed: 11/09/2022]
Affiliation(s)
- Daniel J G Thirion
- Faculté de Pharmacie, Université de Montréal, Montréal, Québec, Canada.,Pharmacy Department, McGill University Health Center, Montréal, Québec, Canada
| | - Valérian Pasche
- Laboratoire de suivi thérapeutique pharmacologique et pharmacocinétique, Faculté de Pharmacie, Université de Montréal, Montréal, Québec, Canada
| | - Amélie Marsot
- Laboratoire de suivi thérapeutique pharmacologique et pharmacocinétique, Faculté de Pharmacie, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
19
|
Le J, Bradley JS. Optimizing Antibiotic Drug Therapy in Pediatrics: Current State and Future Needs. J Clin Pharmacol 2019; 58 Suppl 10:S108-S122. [PMID: 30248202 DOI: 10.1002/jcph.1128] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 03/01/2018] [Indexed: 12/19/2022]
Abstract
The selection of the right antibiotic and right dose necessitates clinicians understand the contribution of pharmacokinetic variability stemming from age-related physiologic maturation and the pharmacodynamics to optimize drug exposure for clinical response. The complexity of selecting the right dose arises from the multiplicity of pediatric age groups, from premature neonates to adolescents. Body size and age (which relate to organ function) must be incorporated to optimize antibiotic dosing in this vulnerable population. In the effort to optimize and individualize drug dosing regimens, clinical pharmacometrics that incorporate population-based pharmacokinetic modeling, Bayesian estimation, and Monte Carlo simulations are utilized as a quantitative approach to understanding and predicting the pharmacology and clinical and microbiologic efficacy of antibiotics. In addition, opportunistic study designs and alternative blood sampling strategies can serve as practical approaches to ensure successful conduct of pediatric studies. This review article examines relevant literature on optimization of antibiotic pharmacotherapy in pediatric populations published within the last decade. Specific pediatric antibiotic data, including beta-lactam antibiotics, aminoglycosides, and vancomycin, are critically evaluated.
Collapse
Affiliation(s)
- Jennifer Le
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California at San Diego, La Jolla, CA, USA
| | - John S Bradley
- Department of Pediatrics, Division of Infectious Diseases, University of California at San Diego, La Jolla, CA, USA.,Rady Children's Hospital San Diego, San Diego, CA, USA
| |
Collapse
|
20
|
Abstract
Abstract
Purpose
The changes in physiological functions as children grow and organ systems mature result in pharmacokinetic alterations throughout childhood. These alterations in children result in absorption, distribution, metabolism, and excretion of drugs that are different from those seen in the typical adult diseased population.
Summary
Changes in gastrointestinal motility and gastric pH in neonates and infants affect the absorption rate and bioavailability of drugs. Skin absorption rate and extent can be altered by different skin structures and perfusion in young children. Intramuscular and rectal absorption become less predictable in children due to erratic absorption site perfusion and other factors. Children’s body compositions also differ greatly from that in adults. Water-soluble drugs distribute more extensively in newborns due to larger water content than in older children and adults. Drug elimination and excretion are also affected in pediatric population due to differences in liver and renal function. Immature enzyme development and renal function result in reduced clearance of drugs in young children. There are limited pharmacokinetic data available for many drugs used in children.
Conclusion
Considering the changes in pharmacokinetics in children can help pharmacists optimize the dosing and monitoring of drugs and do the best they can to help this vulnerable population.
Collapse
|
21
|
Poole K, Gilmour C, Farha MA, Parkins MD, Klinoski R, Brown ED. Meropenem potentiation of aminoglycoside activity against Pseudomonas aeruginosa: involvement of the MexXY-OprM multidrug efflux system. J Antimicrob Chemother 2019; 73:1247-1255. [PMID: 29420743 DOI: 10.1093/jac/dkx539] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 12/20/2017] [Indexed: 12/25/2022] Open
Abstract
Objectives To assess the ability of meropenem to potentiate aminoglycoside (AG) activity against laboratory and AG-resistant cystic fibrosis (CF) isolates of Pseudomonas aeruginosa and to elucidate its mechanism of action. Methods AG resistance gene deletions were engineered into P. aeruginosa laboratory and CF isolates using standard gene replacement technology. Susceptibility to AGs ± meropenem (at ½ MIC) was assessed using a serial 2-fold dilution assay. mexXY expression and MexXY-OprM efflux activity were quantified using quantitative PCR and an ethidium bromide accumulation assay, respectively. Results A screen for agents that rendered WT P. aeruginosa susceptible to a sub-MIC concentration of the AG paromomycin identified the carbapenem meropenem, which potentiated several additional AGs. Meropenem potentiation of AG activity was largely lost in a mutant lacking the MexXY-OprM multidrug efflux system, an indication that it was targeting this efflux system in enhancing P. aeruginosa susceptibility to AGs. Meropenem failed to block AG induction of mexXY expression or MexXY-OprM efflux activity, suggesting that it may be interfering with some MexXY-dependent process linked to AG susceptibility. Meropenem potentiated AG activity versus AG-resistant CF isolates, enhancing susceptibility to at least one AG in all isolates and susceptibility to all tested AGs in 50% of the isolates. Notably, meropenem potentiation of AG activity was linked to MexXY in some but not all CF isolates in which this was examined. Conclusions Meropenem potentiates AG activity against laboratory and CF strains of P. aeruginosa, both dependent on and independent of MexXY, highlighting the complexity of AG resistance in this organism.
Collapse
Affiliation(s)
- Keith Poole
- Department of Biomedical and Molecular Sciences, Botterell Hall, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Christie Gilmour
- Department of Biomedical and Molecular Sciences, Botterell Hall, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Maya A Farha
- M.G. DeGroote Institute for Infectious Disease Research and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Michael D Parkins
- Department of Microbiology Immunology and Infectious Diseases and Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Rachael Klinoski
- Department of Biomedical and Molecular Sciences, Botterell Hall, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Eric D Brown
- M.G. DeGroote Institute for Infectious Disease Research and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| |
Collapse
|
22
|
Crass RL, Pai MP, Lodise TP. Individualizing piperacillin/tazobactam dosing in adult patients with cystic fibrosis: can tobramycin measurements help? J Antimicrob Chemother 2019; 74:126-129. [PMID: 30252050 DOI: 10.1093/jac/dky388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/27/2018] [Indexed: 11/14/2022] Open
Abstract
Background Empirical models to predict β-lactam pharmacokinetics (PK) using information from routine aminoglycoside therapeutic drug monitoring (TDM) have been proposed for critically ill patients; however, no such models exist for patients with cystic fibrosis (CF). Objectives To investigate whether PK parameters of tobramycin could be used to predict those of piperacillin. Methods A non-interventional, open-label PK study was conducted in hospitalized adults treated with piperacillin/tazobactam and tobramycin for acute pulmonary exacerbations of CF. Six serum samples per patient were collected and analysed. One- and two-compartment population PK models with linear, Michaelis-Menten or mixed elimination were evaluated for both drugs within the PmetricsTM package for R. Models were developed and compared iteratively using the log-likelihood and Akaike information criterion (AIC) objective functions. Results Nine primarily female (n = 8) and Caucasian (100%) adult CF patients were enrolled. The median (IQR) age, height, weight and serum creatinine of included patients was 31 (27-32) years, 51.4 (49.9-55.8) kg, 162.6 (160.0-165.1) cm and 0.6 (0.5-0.6) mg/dL, respectively. The final model with the lowest objective function values consisted of one compartment with first-order elimination for tobramycin and two compartments with mixed-order elimination for piperacillin with the elimination rate constant of piperacillin modelled as a linear function of the elimination rate constant of tobramycin. Conclusions A relationship was identified between the elimination rate constants of tobramycin and piperacillin. Validation of this relationship in larger studies of adult patients with CF is needed before application to the precision dosing of piperacillin/tazobactam in this patient population.
Collapse
Affiliation(s)
- Ryan L Crass
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Manjunath P Pai
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Thomas P Lodise
- Department of Pharmacy Practice, Albany College of Pharmacy and Health Sciences, Albany, NY, USA
| |
Collapse
|
23
|
Nolt VD, Pijut KD, Autry EB, Williams WC, Burgess DS, Burgess DR, Arora V, Kuhn RJ. Amikacin target achievement in adult cystic fibrosis patients utilizing Monte Carlo simulation. Pediatr Pulmonol 2019; 54:33-39. [PMID: 30507069 DOI: 10.1002/ppul.24194] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 09/27/2018] [Indexed: 11/12/2022]
Abstract
AIM Pseudomonas aeruginosa (PsA) is a common pathogen in cystic fibrosis (CF). Management of an acute pulmonary exacerbation (APE) caused by PsA is dual anti-pseudomonal antibiotics, a beta-lactam plus aminoglycoside. Aminoglycoside dosing in CF differs from the general population due to altered pharmacokinetics. The primary objective of this study was to utilize pharmacokinetic data from adult CF patients that received amikacin to determine the probability of target attainment for APEs caused by PsA. METHODS This was a single-center, non-randomized, retrospective cohort study of patients >18 years with CF that received intravenous amikacin between January 2010 and July 2016. Amikacin dose, frequency, and serum concentrations were used to calculate pharmacokinetic parameters assuming a one-compartment model. Monte Carlo simulation was conducted with MIC values from CF patients with a PsA positive sputum culture between January 2014 and September 2016 to predict concentration-time profiles for different doses of amikacin. RESULTS This study included pharmacokinetic parameters for 14 amikacin courses administered to six unique patients. The average empiric dose of amikacin was 24.3 ± 14.6 mg/kg, achieving a peak:MIC ratio ≥8 at a rate of 37% (median 5.87; IQR 3.05-10.96). A dose of 45 mg/kg/day was needed to achieve target peak:MIC ratios 90% of the time for a PsA MIC of 8 mg/L. CONCLUSION Our data suggests it may not be clinically feasible to utilize amikacin for PsA isolates with a MIC of 16 mg/L. Current guideline dosing recommendations of amikacin 30-35 mg/kg/day are only adequate for PsA with a MIC ≤4 mg/L.
Collapse
Affiliation(s)
- Valerie D Nolt
- Department of Pharmacy Services, University of Kentucky HealthCare, Lexington, Kentucky
| | - Kyle D Pijut
- Department of Pharmacy Services, University of Kentucky HealthCare, Lexington, Kentucky
| | - Elizabeth B Autry
- Department of Pharmacy Services, University of Kentucky HealthCare, Lexington, Kentucky.,Department of Pharmacy Practice and Science, University of Kentucky College of Pharmacy, Lexington, Kentucky
| | - Whitney C Williams
- Department of Pharmacy Services, University of Kentucky HealthCare, Lexington, Kentucky
| | - David S Burgess
- Department of Pharmacy Practice and Science, University of Kentucky College of Pharmacy, Lexington, Kentucky
| | - Donna R Burgess
- Department of Pharmacy Services, University of Kentucky HealthCare, Lexington, Kentucky.,Department of Pharmacy Practice and Science, University of Kentucky College of Pharmacy, Lexington, Kentucky
| | - Vaneet Arora
- Clinical Microbiology, University of Kentucky HealthCare, Lexington, Kentucky
| | - Robert J Kuhn
- Department of Pharmacy Practice and Science, University of Kentucky College of Pharmacy, Lexington, Kentucky
| |
Collapse
|
24
|
Hong LT, Liou TG, Deka R, King JB, Stevens V, Young DC. Pharmacokinetics of Continuous Infusion Beta-lactams in the Treatment of Acute Pulmonary Exacerbations in Adult Patients With Cystic Fibrosis. Chest 2018; 154:1108-1114. [PMID: 29908155 PMCID: PMC6689083 DOI: 10.1016/j.chest.2018.06.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 06/01/2018] [Accepted: 06/04/2018] [Indexed: 10/14/2022] Open
Abstract
BACKGROUND Several clinical trials have shown the efficacy of continuous infusion beta-lactam (BL) antibiotics in patients with cystic fibrosis (CF); however, little is known about pharmacokinetic changes during the treatment of an acute pulmonary exacerbation (APE). Identifying and understanding these changes may assist in optimizing antibiotic dosing during APE treatment. METHODS This study was a retrospective cohort study of 162 adult patients with CF admitted to the University of Utah Hospital between January 1, 2008, and May 15, 2014, for treatment of an APE with both a continuous infusion BL and IV tobramycin. We extracted the administered doses of continuous infusion BLs and tobramycin along with serum drug concentrations and calculated medication clearance rates. The primary outcome was change in clearance rates of continuous infusion BLs between day 2 and day 7 of APE treatment. RESULTS The BL clearance rate increased 20.7% (95% CI, 11.42 to 32.49; P < .001), whereas the tobramycin clearance rate decreased 6.3% (95% CI, -12.29 to -4.45; P < .001). The mean percent predicted FEV1 increased between admission and discharge by 12.2% (95% CI, -13.81 to -10.55; P < .001). CONCLUSIONS Clinicians should monitor BL levels along with aminoglycoside levels and make dose adjustments to maximize the chance of optimal antibiotic treatment. Continuous infusion BL and tobramycin clearance can change dramatically during the treatment of an APE, which may necessitate significant changes in dosing to achieve optimal antibiotic levels. Clearance rates of these antibiotics may change in opposite directions, requiring specific monitoring of each medication.
Collapse
Affiliation(s)
- Lisa T Hong
- Loma Linda University School of Pharmacy, Loma Linda, CA.
| | | | - Rishi Deka
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, San Diego, CA
| | | | | | - David C Young
- University of Utah College of Pharmacy, Salt Lake City, UT; University of Utah Health Care, Salt Lake City, UT
| |
Collapse
|
25
|
Lafoeste H, Regard L, Martin C, Chassagnon G, Burgel PR. [Acute pulmonary and non-pulmonary complications in adults with cystic fibrosis]. REVUE DE PNEUMOLOGIE CLINIQUE 2018; 74:267-278. [PMID: 30343944 DOI: 10.1016/j.pneumo.2018.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
INTRODUCTION Cystic fibrosis (CF) is a genetic disease primarily affecting the lungs, which could lead to chronic respiratory failure and premature death. CF patients are usually followed in specialized centers, but may present outside of these centers when they seek care for acute pulmonary and/or non-pulmonary complications. The aim of this paper is to provide appropriate knowledge necessary for managing respiratory and non-respiratory emergencies in CF adults. METHODS The review is based on international guidelines, extensive search of the available literature using Pubmed, and experience of the CF reference center at Cochin hospital (Paris, France). Complications occurring after solid organ transplantation (e.g., lung and/or liver) are excluded from this review. RESULTS Main acute respiratory complications are pulmonary exacerbations, hemoptysis, pneumothorax and allergic bronchopulmonary aspergillosis. Acute non-respiratory complications include hyponatremic dehydration, acute pancreatitis, acute complications of gallstones, distal intestinal obstruction syndrome, symptomatic nephrolithiasis, acute kidney injury, drug intolerances and catheter-related acute complications. CONCLUSION This review summarizes acute pulmonary and non-pulmonary complications occurring in adults with CF, focusing on diagnosis and principles of treatment, with the aim of providing a reference that can be used in clinical practice.
Collapse
Affiliation(s)
- H Lafoeste
- Université Paris Descartes, Sorbonne Paris cité, 75005 Paris, France; Service de pneumologie, centre de référence maladies rares : mucoviscidose et affections liées à une anomalie de CFTR (Site coordonnateur national), hôpital Cochin, AP-HP, 75014 Paris, France
| | - L Regard
- Université Paris Descartes, Sorbonne Paris cité, 75005 Paris, France; Service de pneumologie, centre de référence maladies rares : mucoviscidose et affections liées à une anomalie de CFTR (Site coordonnateur national), hôpital Cochin, AP-HP, 75014 Paris, France
| | - C Martin
- Université Paris Descartes, Sorbonne Paris cité, 75005 Paris, France; Service de pneumologie, centre de référence maladies rares : mucoviscidose et affections liées à une anomalie de CFTR (Site coordonnateur national), hôpital Cochin, AP-HP, 75014 Paris, France
| | - G Chassagnon
- Université Paris Descartes, Sorbonne Paris cité, 75005 Paris, France; Service de radiologie, hôpital Cochin, AP-HP, 75014 Paris, France
| | - P-R Burgel
- Université Paris Descartes, Sorbonne Paris cité, 75005 Paris, France; Service de pneumologie, centre de référence maladies rares : mucoviscidose et affections liées à une anomalie de CFTR (Site coordonnateur national), hôpital Cochin, AP-HP, 75014 Paris, France.
| |
Collapse
|
26
|
Mikalauskas A, Parkins MD, Poole K. Rifampicin potentiation of aminoglycoside activity against cystic fibrosis isolates of Pseudomonas aeruginosa. J Antimicrob Chemother 2018; 72:3349-3352. [PMID: 28961705 DOI: 10.1093/jac/dkx296] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 07/21/2017] [Indexed: 01/30/2023] Open
Abstract
Objectives Rifampicin potentiates the activity of aminoglycosides (AGs) versus Pseudomonas aeruginosa by targeting the AmgRS two-component system. In this study we examine the impact of rifampicin on the AG susceptibility of cystic fibrosis (CF) lung isolates of P. aeruginosa and the contribution of AmgRS to AG resistance in these isolates. Methods amgR deletion derivatives of clinical isolates were constructed using standard gene replacement technology. Susceptibility to AGs ± rifampicin (at ½ MIC) was assessed using a serial 2-fold dilution assay. Results Rifampicin showed a variable ability to potentiate AG activity versus the CF isolates, enhancing AG susceptibility between 2- and 128-fold. Most strains showed potentiation for at least two AGs, with only a few strains showing no AG potentiation by rifampicin. Notably, loss of amgR increased AG susceptibility although rifampicin potentiation of AG activity was still observed in the ΔamgR derivatives. Conclusions AmgRS contributes to AG resistance in CF isolates of P. aeruginosa and rifampicin shows a variable ability to potentiate AG activity against these, highlighting the complexity of AG resistance in such isolates.
Collapse
Affiliation(s)
- Alaya Mikalauskas
- Department of Biomedical and Molecular Sciences, Botterell Hall, Queen's University, Kingston, Ontario, K7L 3N6, Canada
| | - Michael D Parkins
- Department of Microbiology, Immunology and Infectious Diseases and Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Keith Poole
- Department of Biomedical and Molecular Sciences, Botterell Hall, Queen's University, Kingston, Ontario, K7L 3N6, Canada
| |
Collapse
|
27
|
Illamola SM, Sherwin CM, van Hasselt JGC. Clinical Pharmacokinetics of Amikacin in Pediatric Patients: A Comprehensive Review of Population Pharmacokinetic Analyses. Clin Pharmacokinet 2018; 57:1217-1228. [DOI: 10.1007/s40262-018-0641-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
28
|
Deschamp AR, Pettit RS, Donaldson JA, Slaven JE, Davis SD. Safety of intravenous tobramycin in combination with a variety of anti-pseudomonal antibiotics in children with cystic fibrosis. SAGE Open Med 2017; 5:2050312117736694. [PMID: 29085640 PMCID: PMC5648079 DOI: 10.1177/2050312117736694] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 09/20/2017] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVES Previous studies have examined renal safety of once daily intravenous tobramycin in individuals with cystic fibrosis. This has been mainly in combination with ceftazidime in an adolescent or adult population. In this report, we describe our institutional experience of once daily intravenous tobramycin in combination with a variety of second anti-pseudomonal antibiotics in children with cystic fibrosis. METHODS We present a retrospective review including children with cystic fibrosis, who were admitted for a pulmonary exacerbation from January 2009 to December 2011, and treated using intravenous tobramycin. A literature review of once daily intravenous aminoglycoside dosing in cystic fibrosis was performed to compare our results to existing literature. RESULTS A total of 35 subjects were divided into once daily dosing (n = 20) versus multiple daily dosing (n = 15) groups. Mean age was 11.3 years (± 5.7) for the once daily dosing group and 13.1 years (± 4.4) for the multiple daily dosing group (p = 0.34). All subjects had normal baseline serum creatinine at admission (once daily dosing 0.49 ± 0.14 mg/dL vs multiple daily dosing 0.62 ± 0.23 mg/dL, p = 0.07). All subjects received intravenous tobramycin, and most received piperacillin-tazobactam as their second anti-pseudomonal antibiotic (once daily dosing 45% and multiple daily dosing 40%). There was no significant change in serum creatinine in either group during antibiotic treatment (once daily dosing 0.08 ± 0.12 mg/dL vs. multiple daily dosing 0.06 ± 0.10 mg/dL, p = 0.43). All subjects had significant improvement in lung function following intravenous antibiotic therapy. CONCLUSION We show that both once daily dosing and multiple daily dosing of intravenous tobramycin in combination with a variety of second anti-pseudomonal antibiotics were safe in terms of nephrotoxicity in children with cystic fibrosis. These findings are important given existing literature mainly examines once daily tobramycin in combination with ceftazidime, a cephalosporin, and the majority of our patients were on tobramycin with piperacillin-tazobactam, an extended spectrum penicillin plus beta-lactam. This contributes new information not previously examined in a pediatric cystic fibrosis population.
Collapse
Affiliation(s)
- Ashley R Deschamp
- Pediatric Pulmonology, University of Nebraska Medical Center and Children's Hospital and Medical Center, Omaha, NE, USA
| | - Rebecca S Pettit
- Indiana University School of Medicine and Riley Hospital for Children at Indiana University Health, Indianapolis, IN, USA
| | - Jennifer A Donaldson
- Indiana University School of Medicine and Riley Hospital for Children at Indiana University Health, Indianapolis, IN, USA
| | - James E Slaven
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Stephanie D Davis
- Indiana University School of Medicine and Riley Hospital for Children at Indiana University Health, Indianapolis, IN, USA
| |
Collapse
|
29
|
Singh M, Yau YCW, Wang S, Waters V, Kumar A. MexXY efflux pump overexpression and aminoglycoside resistance in cystic fibrosis isolates of Pseudomonas aeruginosa from chronic infections. Can J Microbiol 2017; 63:929-938. [PMID: 28922614 DOI: 10.1139/cjm-2017-0380] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In this study, we analyzed 15 multidrug-resistant cystic fibrosis isolates of Pseudomonas aeruginosa from chronic lung infections for expression of 4 different multidrug efflux systems (MexAB-OprM, MexCD-OprJ, MexEF-OprN, and MexXY), using quantitative reverse transcriptase PCR. Overexpression of MexXY pump was observed in all of the isolates tested. Analysis of regulatory genes that control the expression of these 4 efflux pumps revealed a number of previously uncharacterized mutations. Our work shows that MexXY pump overexpression is common in cystic fibrosis isolates and could be contributing to their reduced aminoglycoside susceptibility. Further, we also identified novel mutations in the regulatory genes of the 4 abovementioned Resistance-Nodulation-Division superfamily pumps that may be involved in the overexpression of these pumps.
Collapse
Affiliation(s)
- Manu Singh
- a Department of Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Yvonne C W Yau
- b Division of Microbiology, Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Shirley Wang
- a Department of Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Valerie Waters
- c Division of Infectious Diseases, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Ayush Kumar
- a Department of Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada.,d Manitoba Chemosensory Biology Group, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
30
|
Virant-Young D, Thomas J, Woiderski S, Powers M, Carlier J, McCarty J, Kupchick T, Larder A. Cystic Fibrosis: A Novel Pharmacologic Approach to Cystic Fibrosis Transmembrane Regulator Modulation Therapy. J Osteopath Med 2016; 115:546-55. [PMID: 26322933 DOI: 10.7556/jaoa.2015.112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Therapy for cystic fibrosis (CF) has progressed during the past several decades. Much of this progress is because of advances in genetic testing to precisely identify the underlying cause of CF transmembrane regulator (CFTR) dysfunction. However, with more than 1900 mutations that can produce a faulty CFTR, the management of CF can remain a challenge. Several innovative drugs recently approved by the Food and Drug Administration, termed genetic modulators, target the underlying disease by modulating the CFTR defect. This review provides physicians with an established simple classification scheme to guide their use of these drugs. The treatment challenge of 1900 CFTR mutations has been simplified into 6 physiologic classes, each paired with an available therapy to offer patients the most functional improvement. Drug therapy monitoring, adverse effects, and indications for discontinuation must also be considered.
Collapse
|
31
|
Dassner AM, Sutherland C, Girotto J, Nicolau DP. In vitro Activity of Ceftolozane/Tazobactam Alone or with an Aminoglycoside Against Multi-Drug-Resistant Pseudomonas aeruginosa from Pediatric Cystic Fibrosis Patients. Infect Dis Ther 2016; 6:129-136. [PMID: 27943223 PMCID: PMC5336416 DOI: 10.1007/s40121-016-0141-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Indexed: 11/29/2022] Open
Abstract
INTRODUCTION Gram-negative multi-drug resistance is an emerging threat among pediatric patients with cystic fibrosis (CF). Ceftolozane/tazobactam (C/T) is an extended-spectrum cephalosporin/beta-lactamase inhibitor combination that has been shown to maintain activity against MDR P. aeruginosa isolates. The understanding of C/T effectiveness in pediatric patients is extremely limited. Minimum inhibitory concentration (MIC) testing and time-kill analyses were performed to better understand the antimicrobial susceptibility and potential role of C/T. METHODS Non-duplicate clinical respiratory MDR P. aeruginosa isolates (n = 5) from four pediatric CF patients were identified. MICs were determined for these isolates using CLSI broth microdilution methods. Time-kill analyses were performed using multiples of C/T alone, and combinations of C/T 2× and 8× the MIC with 30 mg/L tobramycin or 80 mg/L amikacin for all isolates. Cell counts were determined by serial dilution plating. RESULTS Isolates had variable susceptibilities to C/T (range 0.5-8 mg/L), tobramycin (range 2 to >64 mg/L) and amikacin (range 8 to >256 mg/L). Time-kill analyses revealed an average of 0.71 (range -0.6 to 4.4), 1.50 (range 0.8-2.0) and 2.1 (range 1.2-3.4) log-kill at 4×, 8× and 16× the C/T MIC, respectively. At a tobramycin MIC of 32 mg/L, combination therapy showed synergistic benefit when the isolate was C/T susceptible. C/T and amikacin combination therapy showed synergistic activity at an amikacin MIC >256 mg/L when C/T MIC was 2 mg/L (4.7 log-kill at 2× C/T MIC and 4.0 log-kill at 8× C/T MIC). CONCLUSION C/T appears to be a promising treatment option for treatment of MDR P. aeruginosa in pediatric CF patients, both alone and in combination with tobramycin or amikacin. Interestingly, the benefit of C/T combination therapy with amikacin may be more pronounced than with the addition of tobramycin. Further evaluation of such combination regimens in pediatric CF patients is warranted.
Collapse
Affiliation(s)
| | - Christina Sutherland
- Center for Anti-Infective Research and Development, Hartford Hospital, Hartford, CT, USA
| | - Jennifer Girotto
- Connecticut Children's Medical Center, Hartford, CT, USA.,University of Connecticut School of Pharmacy, Storrs, CT, USA
| | - David P Nicolau
- Center for Anti-Infective Research and Development, Hartford Hospital, Hartford, CT, USA. .,Division of Infectious Diseases, Hartford Hospital, Hartford, CT, USA.
| |
Collapse
|
32
|
Khanam S, Guragain M, Lenaburg DL, Kubat R, Patrauchan MA. Calcium induces tobramycin resistance in Pseudomonas aeruginosa by regulating RND efflux pumps. Cell Calcium 2016; 61:32-43. [PMID: 28034459 DOI: 10.1016/j.ceca.2016.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 10/27/2016] [Accepted: 11/17/2016] [Indexed: 11/24/2022]
Abstract
Pseudomonas aeruginosa is an opportunistic multidrug resistant pathogen causing severe chronic infections. Our previous studies showed that elevated calcium (Ca2+) enhances production of several virulence factors and plant infectivity of the pathogen. Here we show that Ca2+ increases resistance of P. aeruginosa PAO1 to tobramycin, antibiotic commonly used to treat Pseudomonas infections. LC-MS/MS-based comparative analysis of the membrane proteomes of P aeruginosa grown at elevated versus not added Ca2+, determined that the abundances of two RND (resistance-nodulation-cell division) efflux pumps, MexAB-OprM and MexVW-OprM, were increased in the presence of elevated Ca2+. Analysis of twelve transposon mutants with disrupted RND efflux pumps showed that six of them (mexB, muxC, mexY, mexJ, czcB, and mexE) contribute to Ca2+-induced tobramycin resistance. Transcriptional analyses by promoter activity and RT-qPCR showed that the expression of mexAB, muxABC, mexXY, mexJK, czcCBA, and mexVW is increased by elevated Ca2+. Disruption of mexJ, mexC, mexI, and triA significantly decreased Ca2+-induced plant infectivity of the pathogen. Earlier, our group showed that PAO1 maintains intracellular Ca2+ (Ca2+in) homeostasis, which mediates Ca2+ regulation of P. aeruginosa virulence, and identified four putative Ca2+ transporters involved in this process (Guragain et al., 2013). Here we show that three of these transporters (PA2435, PA2092, PA4614) play role in Ca2+-induced tobramycin resistance and one of them (PA2435) contributes to Ca2+ regulation of mexAB-oprM promoter activity. Furthermore, mexJ, czcB, and mexE contribute to the maintenance of Ca2+in homeostasis. This provides the first evidence that Ca2+in homeostasis mediates Ca2+ regulation of RND transport systems, which contribute to Ca2+-enhanced tobramycin resistance and plant infectivity in P. aeruginosa.
Collapse
Affiliation(s)
- Sharmily Khanam
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK, United States
| | - Manita Guragain
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK, United States
| | - Dirk L Lenaburg
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK, United States
| | - Ryan Kubat
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK, United States
| | - Marianna A Patrauchan
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK, United States.
| |
Collapse
|
33
|
Abstract
There is a high prevalence of Pseudomonas aeruginosa in patients with cystic fibrosis and clear epidemiologic links between chronic infection and morbidity and mortality exist. Prevention and early identification of infection are critical, and stand to improve with the advent of new vaccines and laboratory methods. Once the organism is identified, a variety of treatment options are available. Aggressive use of antipseudomonal antibiotics is the standard of care for acute pulmonary exacerbations in cystic fibrosis, and providers must take into account specific patient characteristics when making treatment decisions related to antibiotic selection, route and duration of administration, and site of care.
Collapse
Affiliation(s)
- Jaideep S Talwalkar
- Department of Internal Medicine, Yale School of Medicine, 333 Cedar Street, PO Box 208086, New Haven, CT 06520-8086, USA; Department of Pediatrics, Yale School of Medicine, 333 Cedar Street, PO Box 208084, New Haven, CT 06520-8084, USA.
| | - Thomas S Murray
- Department of Medical Sciences, Frank H Netter MD School of Medicine, Quinnipiac University, 275 Mount Carmel Avenue, Hamden, CT 06518, USA; Division of Infectious Diseases and Immunology, Connecticut Children's Medical Center, 282 Washington Street, Suite 2L, Hartford, CT 06106, USA
| |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW The current guidelines and recent clinical research in the management of Pseudomonas aeruginosa respiratory infections in cystic fibrosis (CF) are reviewed. Areas where further research is required will also be highlighted. RECENT FINDINGS P. aeruginosa is a key respiratory pathogen in CF. Inhaled tobramycin or colistin is recommended for early eradication to prevent establishment of chronic infection. Other antibiotic options are currently being investigated. The long-term success of eradication strategies is also now being assessed. The use of inhaled antibiotics in the management of chronic P. aeruginosa infection is an area of active investigation. Acute pulmonary exacerbations are still a major cause of morbidity and mortality. Guidelines continue to recommend combination intravenous therapy but further research is required to clarify the advantage of this approach. Multidrug resistance is common and potentially more effective antipseudomonal antibiotics may soon become available. SUMMARY The management of P. aeruginosa respiratory infection in CF remains a challenging area, especially in the setting of multidrug resistance. The role of inhaled antibiotics continues to be expanded. Further research is required in the key areas of eradication and management of chronic infection and acute pulmonary exacerbations to identify those treatments that optimize long-term, clinical benefits.
Collapse
|
35
|
Zobell JT, Epps K, Kittell F, Sema C, McDade EJ, Peters SJ, Duval MA, Pettit RS. Tobramycin and Beta-Lactam Antibiotic Use in Cystic Fibrosis Exacerbations: A Pharmacist Approach. J Pediatr Pharmacol Ther 2016; 21:239-46. [PMID: 27453702 DOI: 10.5863/1551-6776-21.3.239] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVES Survey suggests that recommended doses and dosage regimens for antipseudomonal antibiotics for the treatment of acute pulmonary exacerbations in cystic fibrosis (CF) patients are not used, and one way to address these disparities is the involvement of pharmacists who are dedicated to CF. This is the first survey specifically designed for pharmacists at Cystic Fibrosis Foundation (CFF)-accredited centers to identify how tobramycin and antipseudomonal beta-lactams are being used. The purpose of this survey is to quantify this information and to promote future study to allow for implementation of tobramycin and beta-lactam dosage and monitoring standardization. METHODS An anonymous national cross-sectional survey of pharmacists that are affliated with CFF-accredited programs was performed using Qualtrics.com. RESULTS The survey had a 48.5% response rate. Most pediatric pharmacists (78.6%) report using extended-interval tobramycin dosage. The most common reported starting dosage was 10 mg/kg every 24 hours; most centers aim for a maximum serum concentration (Cmax) between 20 and 40 mg/L (78.6%). A total of 26 adult pharmacists reported using extended-interval dosage (96%), using an initial dosage of 10 mg/kg/day. The most common parameters used to adjust dosage were Cmax and area under the curve (AUC; 31%); the Cmax goal was 20 to 40 mg/L (84.2%). Most respondents (79%) report using beta-lactams in combination with tobramycin. Extended-infusion and continuous-infusion beta-lactams were used more in adults than pediatric patients. CONCLUSIONS Most CF pharmacists report using extended-interval tobramycin. With the information from this survey, the establishment of future consensus recommendations by pharmacists for optimal and consistent tobramycin and antipseudomonal beta-lactam dosage and monitoring strategies needs to be considered.
Collapse
Affiliation(s)
| | - Kevin Epps
- St. Vincent's Hospital Riverside, Jacksonville, Florida
| | | | | | | | | | | | - Rebecca S Pettit
- Riley Hospital for Children at Indiana University Health, Indianapolis, Indiana
| |
Collapse
|
36
|
Brigg Turner R, Elbarbry F, Biondo L. Pharmacokinetics of once and twice daily dosing of intravenous tobramycin in paediatric patients with cystic fibrosis. J Chemother 2016; 28:304-7. [DOI: 10.1179/1973947815y.0000000077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
37
|
Assessing Predictive Performance of Published Population Pharmacokinetic Models of Intravenous Tobramycin in Pediatric Patients. Antimicrob Agents Chemother 2016; 60:3407-14. [PMID: 27001806 DOI: 10.1128/aac.02654-15] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 03/11/2016] [Indexed: 11/20/2022] Open
Abstract
Several population pharmacokinetic models describe the dose-exposure relationship of tobramycin in pediatric patients. Before the implementation of these models in clinical practice for dosage adjustment, their predictive performance should be externally evaluated. This study tested the predictive performance of all published population pharmacokinetic models of tobramycin developed for pediatric patients with an independent patient cohort. A literature search was conducted to identify suitable models for testing. Demographic and pharmacokinetic data were collected retrospectively from the medical records of pediatric patients who had received intravenous tobramycin. Tobramycin exposure was predicted from each model. Predictive performance was assessed by visual comparison of predictions to observations, by calculation of bias and imprecision, and through the use of simulation-based diagnostics. Eight population pharmacokinetic models were identified. A total of 269 concentration-time points from 41 pediatric patients with cystic fibrosis were collected for external evaluation. Three models consistently performed best in all evaluations and had mean errors ranging from -0.4 to 1.8 mg/liter, relative mean errors ranging from 4.9 to 29.4%, and root mean square errors ranging from 47.8 to 66.9%. Simulation-based diagnostics supported these findings. Models that allowed a two-compartment disposition generally had better predictive performance than those that used a one-compartment disposition model. Several published models of the pharmacokinetics of tobramycin showed reasonable low levels of bias, although all models seemed to have some problems with imprecision. This suggests that knowledge of typical pharmacokinetic behavior and patient covariate values alone without feedback concentration measurements from individual patients is not sufficient to make precise predictions.
Collapse
|
38
|
Potentiation of Aminoglycoside Activity in Pseudomonas aeruginosa by Targeting the AmgRS Envelope Stress-Responsive Two-Component System. Antimicrob Agents Chemother 2016; 60:3509-18. [PMID: 27021319 DOI: 10.1128/aac.03069-15] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/17/2016] [Indexed: 02/06/2023] Open
Abstract
A screen for agents that potentiated the activity of paromomycin (PAR), a 4,5-linked aminoglycoside (AG), against wild-type Pseudomonas aeruginosa identified the RNA polymerase inhibitor rifampin (RIF). RIF potentiated additional 4,5-linked AGs, such as neomycin and ribostamycin, but not the clinically important 4,6-linked AGs amikacin and gentamicin. Potentiation was absent in a mutant lacking the AmgRS envelope stress response two-component system (TCS), which protects the organism from AG-generated membrane-damaging aberrant polypeptides and, thus, promotes AG resistance, an indication that RIF was acting via this TCS in potentiating 4,5-linked AG activity. Potentiation was also absent in a RIF-resistant RNA polymerase mutant, consistent with its potentiation of AG activity being dependent on RNA polymerase perturbation. PAR-inducible expression of the AmgRS-dependent genes htpX and yccA was reduced by RIF, suggesting that AG activation of this TCS was compromised by this agent. Still, RIF did not compromise the membrane-protective activity of AmgRS, an indication that it impacted some other function of this TCS. RIF potentiated the activities of 4,5-linked AGs against several AG-resistant clinical isolates, in two cases also potentiating the activity of the 4,6-linked AGs. These cases were, in one instance, explained by an observed AmgRS-dependent expression of the MexXY multidrug efflux system, which accommodates a range of AGs, with RIF targeting of AmgRS undermining mexXY expression and its promotion of resistance to 4,5- and 4,6-linked AGs. Given this link between AmgRS, MexXY expression, and pan-AG resistance in P. aeruginosa, RIF might be a useful adjuvant in the AG treatment of P. aeruginosa infections.
Collapse
|
39
|
Paviour S, Hennig S, Staatz CE. Usage and monitoring of intravenous tobramycin in cystic fibrosis in Australia and the UK. JOURNAL OF PHARMACY PRACTICE AND RESEARCH 2016. [DOI: 10.1002/jppr.1145] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Sophie Paviour
- Pharmacy Department; Redcliffe Hospital; Brisbane Australia
| | - Stefanie Hennig
- School of Pharmacy; University of Queensland; Brisbane Australia
| | | |
Collapse
|
40
|
Ehsan Z, Clancy JP. Management of Pseudomonas aeruginosa infection in cystic fibrosis patients using inhaled antibiotics with a focus on nebulized liposomal amikacin. Future Microbiol 2015; 10:1901-12. [PMID: 26573178 DOI: 10.2217/fmb.15.117] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Pseudomonas aeruginosa (PsA) is a highly prevalent bacterial organism recovered from the lungs of cystic fibrosis (CF) patients and chronic PsA infection is linked to progressive pulmonary function decline. The eradication and treatment of this organism from CF airways is particularly challenging to CF care providers. Aerosolized antibiotics that target PsA help to slow down growth, maintain lung function and reduce the frequency of pulmonary exacerbations. In this review, we discuss the currently available inhaled antibiotics for management of PsA lung infections in CF patients, with a focus on liposomal amikacin for inhalation (LAI). LAI is a unique formulation of amikacin under development that enhances drug delivery and retention in CF airways via drug incorporation into neutral liposomes. Factors such as once-daily dosing, mucus and biofilm penetration and potentially prolonged off-drug periods make LAI a potentially attractive option to manage chronic PsA lung infections in CF patients.
Collapse
Affiliation(s)
- Zarmina Ehsan
- Division of Pulmonary Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center & the University of Cincinnati, MLC 2021 3333 Burnet Avenue, Cincinnati, OH 45220, USA
| | - John P Clancy
- Division of Pulmonary Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center & the University of Cincinnati, MLC 2021 3333 Burnet Avenue, Cincinnati, OH 45220, USA
| |
Collapse
|
41
|
Molloy L, Nichols K. Infectious Diseases Pharmacotherapy for Children With Cystic Fibrosis. J Pediatr Health Care 2015; 29:565-78; quiz 579-80. [PMID: 26498903 DOI: 10.1016/j.pedhc.2015.07.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 07/29/2015] [Accepted: 07/30/2015] [Indexed: 12/15/2022]
Abstract
Cystic fibrosis (CF) affects several organs, most notably the lungs, which become predisposed to infections with potentially severe consequences. Because of physiologic changes and infection characteristics, unique approaches to antimicrobial agent selection, dosing, and administration are needed. To provide optimal acute and long-term care, pediatric health care providers must be aware of these patient features and common approaches to antimicrobial therapy in CF, which can differ significantly from those of other infectious diseases. The purpose of this article is to review common respiratory pathogens, pharmacology of commonly used antimicrobial agents, and unique pharmacokinetics and dosing strategies often used when treating children with CF.
Collapse
|
42
|
Ehsan Z, Clancy J. T100: nebulized-concentrated tobramycin formulation for treatment of Pseudomonas aeruginosainfection in cystic fibrosis patients. Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.1064308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
43
|
Cantón R, Máiz L, Escribano A, Olveira C, Oliver A, Asensio O, Gartner S, Roma E, Quintana-Gallego E, Salcedo A, Girón R, Barrio MI, Pastor MD, Prados C, Martínez-Martínez MT, Barberán J, Castón JJ, Martínez-Martínez L, Poveda JL, Vázquez C, de Gracia J, Solé A. Spanish Consensus on the Prevention and Treatment of Pseudomonas aeruginosa Bronchial Infections in Cystic Fibrosis Patients. ACTA ACUST UNITED AC 2015. [DOI: 10.1016/j.arbr.2014.09.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
44
|
Cantón R, Máiz L, Escribano A, Olveira C, Oliver A, Asensio O, Gartner S, Roma E, Quintana-Gallego E, Salcedo A, Girón R, Barrio MI, Pastor MD, Prados C, Martínez-Martínez MT, Barberán J, Castón JJ, Martínez-Martínez L, Poveda JL, Vázquez C, de Gracia J, Solé A. Spanish consensus on the prevention and treatment of Pseudomonas aeruginosa bronchial infections in cystic fibrosis patients. Arch Bronconeumol 2015; 51:140-50. [PMID: 25614377 DOI: 10.1016/j.arbres.2014.09.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Accepted: 09/22/2014] [Indexed: 01/22/2023]
Abstract
Pseudomonas aeruginosa is the main pathogen in bronchopulmonary infections in cystic fibrosis (CF) patients. It can only be eradicated at early infection stages while reduction of its bacterial load is the therapeutic goal during chronic infection or exacerbations. Neonatal screening and pharmacokinetic/pharmacodynamic knowledge has modified the management of CF-patients. A culture based microbiological follow-up should be performed in patients with no infection with P.aeruginosa. At initial infection, inhaled colistin (0,5-2MU/tid), tobramycin (300mg/bid) or aztreonam (75mg/tid) with or without oral ciprofloxacin (15-20mg/kg/bid, 2-3weeks) are recommended. In chronic infections, treatment is based on continuous administration of colistin or with a 28-day on-off regimen with tobramycin or aztreonam. During mild-moderate exacerbations oral ciprofloxacin (2-3weeks) can be administered while serious exacerbations must be treated with intravenous combination therapy (beta-lactam with an aminoglycoside or a fluoroquinolone). Future studies will support antibiotic rotation and/or new combination therapies. Epidemiological measures are also recommended to avoid new P.aeruginosa infections and "patient-to-patient transmission" of this pathogen.
Collapse
Affiliation(s)
- Rafael Cantón
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal e Instituto Ramón y Cajal de Investigaciones Sanitarias (IRYCIS), Madrid, España; Red Española de Investigación en Patología Infecciosa (REIPI), Instituto de Salud Carlos III, Madrid, España.
| | - Luis Máiz
- Unidad de Bronquiectasias y Fibrosis Quística, Servicio de Neumología, Hospital Universitario Ramón y Cajal e Instituto Ramón y Cajal de Investigaciones Sanitarias (IRYCIS), Madrid, España
| | - Amparo Escribano
- Unidad de Neumología Pediátrica y Fibrosis Quística, Servicio de Pediatría, Hospital Clínico Universitario, Universidad de Valencia, Valencia, España
| | - Casilda Olveira
- Unidad de Gestión Clínica de Enfermedades Respiratorias, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, España
| | - Antonio Oliver
- Red Española de Investigación en Patología Infecciosa (REIPI), Instituto de Salud Carlos III, Madrid, España; Servicio de Microbiología y Unidad de Investigación, Hospital Universitario Son Espases, Instituto de Investigación Sanitaria de Palma (IdISPa), Palma de Mallorca, España
| | - Oscar Asensio
- Unidad de Neumología y Alergia Pediátrica, Hospital Universitario de Sabadell. Corporació Sanitària Parc Taulí, Sabadell, Barcelona, España
| | - Silvia Gartner
- Unidad de Neumología Pediátrica y Fibrosis Quística, Hospital Universitario Vall d'Hebron, Barcelona, España
| | - Eva Roma
- Servicio de Farmacia, Hospital Universitario y Politécnico La Fe, Valencia, España
| | - Esther Quintana-Gallego
- Unidad de Fibrosis Quística, Servicio de Neumología, Hospital Universitario Virgen del Rocío, Sevilla, España
| | - Antonio Salcedo
- Unidad de Fibrosis Quística Interhospitalaria Niño Jesús-Gregorio Marañón, Madrid, España
| | - Rosa Girón
- Unidad de Bronquiectasias y Fibrosis Quística, Hospital Universitario La Princesa, Instituto La Princesa de Investigación Sanitaria, Madrid, España
| | - María Isabel Barrio
- Sección de Neumología Pediátrica y Unidad de Fibrosis Quística, Hospital Universitario La Paz, Madrid, España
| | - María Dolores Pastor
- Unidad de Neumología Pediátrica y Fibrosis Quística, Hospital Universitario Virgen de la Arrixaca, Murcia, España
| | - Concepción Prados
- Unidad de Fibrosis Quística y Bronquiectasias, Servicio de Neumología, Hospital Universitario La Paz, Madrid, España
| | | | - José Barberán
- Departamento de Medicina Interna, Hospital Montepríncipe, Universidad CEU San Pablo, Madrid, España
| | - Juan José Castón
- Unidad de Enfermedades Infecciosas, Hospital General Universitario de Ciudad Real, Ciudad Real, España
| | - Luis Martínez-Martínez
- Red Española de Investigación en Patología Infecciosa (REIPI), Instituto de Salud Carlos III, Madrid, España; Servicio de Microbiología, Hospital Universitario Marqués de Valdecilla-IDIVAL y Departamento de Biología Molecular, Universidad de Cantabria, Santander, España
| | - José Luis Poveda
- Servicio de Farmacia, Hospital Universitario y Politécnico La Fe, Valencia, España
| | - Carlos Vázquez
- Unidad de Neumología Pediátrica y Fibrosis Quística, Hospital Universitario de Cruces, Baracaldo, Vizcaya, España
| | - Javier de Gracia
- Servicio de Neumología y CIBER en Enfermedades Respiratorias (CibeRES), Hospital Universitari Vall d'Hebron, Universidad Autónoma, Barcelona, España
| | - Amparo Solé
- Unidad de Trasplante Pulmonar y Fibrosis Quística, Hospital Universitario y Politécnico la Fe, Valencia, España.
| | | |
Collapse
|
45
|
Chalhoub H, Tunney M, Elborn JS, Vergison A, Denis O, Plésiat P, Kahl BC, Van Bambeke F, Tulkens PM. Avibactam confers susceptibility to a large proportion of ceftazidime-resistant Pseudomonas aeruginosa isolates recovered from cystic fibrosis patients. J Antimicrob Chemother 2015; 70:1596-8. [PMID: 25587996 DOI: 10.1093/jac/dku551] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Hussein Chalhoub
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Michael Tunney
- CF & Airways Microbiology Research Group, Queen's University Belfast, Belfast, UK
| | - J Stuart Elborn
- CF & Airways Microbiology Research Group, Queen's University Belfast, Belfast, UK
| | - Anne Vergison
- Unité des maladies infectieuses, Hôpital Universitaire des Enfants Reine Fabiola, Brussels, Belgium
| | - Olivier Denis
- Laboratoire de microbiologie, Hôpital Erasme, Brussels, Belgium
| | - Patrick Plésiat
- Laboratoire de bactériologie, Hôpital Jean Minjoz, Besançon, France
| | - Barbara C Kahl
- Medical Microbiology, University Hospital Münster, Münster, Germany
| | - Françoise Van Bambeke
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Paul M Tulkens
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
46
|
Lutsar I, Telling K, Metsvaht T. Treatment option for sepsis in children in the era of antibiotic resistance. Expert Rev Anti Infect Ther 2014; 12:1237-52. [PMID: 25189378 DOI: 10.1586/14787210.2014.956093] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Sepsis caused by multidrug-resistant microorganisms is one of the most serious infectious diseases of childhood and poses significant challenges for pediatricians involved in management of critically ill children. This review discusses the use of pharmacokinetic/dynamic principles (i.e., prolonged infusion of β-lactams and vancomycin, once-daily administration of aminoglycosides and rationale of therapeutic drug monitoring) when prescribing antibiotics to critically ill patients. The potential of 'old' agents (i.e., colistin, fosfomycin) and newly approved antibiotics is critically reviewed. The pros and cons of combination antibacterial therapy are discussed and finally suggestions for the treatment of sepsis caused by multidrug-resistant organisms are provided.
Collapse
Affiliation(s)
- Irja Lutsar
- Institute of Medical Microbiology, University of Tartu, Ravila 19, 50411 Tartu, Estonia
| | | | | |
Collapse
|
47
|
Ehsan Z, Wetzel JD, Clancy JP. Nebulized liposomal amikacin for the treatment of Pseudomonas aeruginosa infection in cystic fibrosis patients. Expert Opin Investig Drugs 2014; 23:743-9. [PMID: 24597573 DOI: 10.1517/13543784.2014.895322] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Chronic lung infection with Pseudomonas aeruginosa (PsA) is associated with more-rapid pulmonary decline and acute pulmonary exacerbations in patients with cystic fibrosis (CF). The treatment and eradication of this organism from CF airways is particularly difficult, making it the focus of many therapeutic endeavors. Inhaled antibiotics used for PsA treatment help to suppress growth of the organism, maintain lung function and reduce the frequency of pulmonary exacerbations. AREAS COVERED Herein, the authors discuss the currently available inhaled antibiotics for management of lung infections in CF patients. They also describe the recent results from clinical trials of agents under development, with a focus on liposomal amikacin for inhalation (LAI, Arikace™). The article also provides a summary of the results from relevant articles published in the English. EXPERT OPINION LAI is a unique formulation of amikacin that enhances drug delivery and retention in CF airways via incorporation into neutral liposomes. Results of a recent Phase II trial suggest that LAI, with the capacity for once-daily dosing and prolonged off-drug periods, may be an attractive choice of inhaled antibiotic to manage PsA lung infections in CF patients. Further data from Phase III studies assessing the efficacy and safety of LAI should better elucidate its potential.
Collapse
Affiliation(s)
- Zarmina Ehsan
- University of Cincinnati, Cincinnati Children's Hospital Medical Center, Division of Pulmonary Medicine, Department of Pediatrics , MLC 2021 3333 Burnet Avenue, Cincinnati, OH 45220 , USA
| | | | | |
Collapse
|
48
|
Sherwin CMT, Zobell JT, Stockmann C, McCrory BE, Wisdom M, Young DC, Olson J, Ampofo K, Spigarelli MG. Pharmacokinetic and pharmacodynamic optimisation of intravenous tobramycin dosing among children with cystic fibrosis. J Pharmacokinet Pharmacodyn 2014; 41:71-9. [DOI: 10.1007/s10928-013-9348-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Accepted: 12/26/2013] [Indexed: 10/25/2022]
|