1
|
Esmaeeli A, Ravan H, Hassanshahian M, Khaleghi M. Rapid LAMP-based detection of A. baumannii and aminoglycoside resistance genes in ESKAPE pathogens. Microb Pathog 2025; 202:107436. [PMID: 40021031 DOI: 10.1016/j.micpath.2025.107436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 01/20/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
The opportunistic pathogen Acinetobacter baumannii has emerged as a formidable challenge in the realm of healthcare-associated infections, primarily due to its remarkable capacity to develop resistance against a diverse array of antibiotics. In response to this pressing concern, this comprehensive study aims to design and validate a rapid and highly precise diagnostic tool, known as the Loop-Mediated Isothermal Amplification (LAMP) assay, for the detection of A. baumannii and its associated aminoglycoside resistance genes. Through meticulous comparative genomic analyses, the researchers have identified a unique 299-base pair segment within the glutathione S-transferase gene that exhibits exceptional specificity to A. baumannii. This discovery has enabled the development of a LAMP-based protocol that demonstrates complete specificity, with no cross-reactivity observed against other bacterial species. Furthermore, the assay has demonstrated a remarkable sensitivity threshold, capable of detecting as few as 10 colony-forming units per reaction for both the glutathione S-transferase target and the armA aminoglycoside resistance gene. Expanding the scope of the investigation, the researchers have also explored the distribution of five key aminoglycoside resistance genes (aadB, aacC1, aadA1, aphA6, and armA) across a diverse collection of bacterial isolates. Interestingly, the study has revealed that the aphA6 gene is the most widespread, being present in all the A. baumannii samples analyzed. The successful implementation of this LAMP-based diagnostic approach not only provides a rapid and precise means of identifying A. baumannii but also offers the ability to evaluate the presence of critical aminoglycoside resistance determinants. The adaptability of this methodology to other pathogens further underscores its potential as an invaluable tool in clinical diagnostics and epidemiological investigations. By enhancing the speed and accuracy of pathogen detection, this research holds promise for improving patient management and strengthening infection control strategies, ultimately contributing to better healthcare outcomes.
Collapse
Affiliation(s)
- Azam Esmaeeli
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Hadi Ravan
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran.
| | - Mehdi Hassanshahian
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Moj Khaleghi
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| |
Collapse
|
2
|
Perrier Q, Dumas AF, Pluchart H, Bartoli M, Gautier E, Bedouch P, Epaulard O. Recommended gentamicin peak plasma levels rarely reached, even with recommended dosages. Infect Dis Now 2025; 55:105076. [PMID: 40294703 DOI: 10.1016/j.idnow.2025.105076] [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: 02/14/2025] [Revised: 04/21/2025] [Accepted: 04/23/2025] [Indexed: 04/30/2025]
Abstract
INTRODUCTION The recommended gentamicin peak plasma concentration range is 32-40 mg/L; we aimed to determine how frequently it was reached, and for which gentamicin doses. PATIENTS AND METHODS We retrospectively reviewed 601 gentamicin peak plasma concentrations in 501 patients aged ≥15 in our institution between 2013 and 2023. RESULTS Median gentamicin dose was 5.9 mg/kg [IQR 4.1-7.9]. Median peak plasma concentration was 16.5 mg/L [IQR 10.8-22.8] and was strongly correlated with dose (p < 0.0001). Only 5.7 % of values were ≥32 mg/L, including 22.8 % for dose ≥10 mg/kg. CONCLUSION This suggests that existing recommendations regarding either dose or target concentration for gentamicin should be modified.
Collapse
Affiliation(s)
- Quentin Perrier
- Université Grenoble Alpes, INSERM, LBFA, U1055, Pharmacy Department, Grenoble Alpes University Hospital, Grenoble, France
| | - Anne-Flore Dumas
- Université Grenoble Alpes, Pharmacy Department, Grenoble Alpes University Hospital, Grenoble, France
| | - Hélène Pluchart
- Université Grenoble Alpes, Pharmacy Department, Grenoble Alpes University Hospital, Grenoble, France
| | - Mireille Bartoli
- Université Grenoble Alpes, Pharmacology, Toxicology and Pharmacogenetics Laboratory, Grenoble Alpes University Hospital, Grenoble, France
| | - Elodie Gautier
- Université Grenoble Alpes, Inserm, CHU Grenoble Alpes, HP2, 38000 Grenoble, France
| | - Pierrick Bedouch
- Université Grenoble Alpes, Pharmacy Department, Grenoble Alpes University Hospital, Grenoble, France
| | - Olivier Epaulard
- Université Grenoble Alpes, CIC-1406, INSERM, Groupe de Recherche en Infectiologie Clinique, Infectious Diseases Department, Grenoble Alpes University Hospital, Grenoble, France.
| |
Collapse
|
3
|
Yamamoto PA, Rodriguez-Vera L, Telles JP, Moreira FL, Roy S, Korn O, Coelho D, Migotto KC, de Carvalho FRT, Caruso P, França E Silva IL, Vozmediano V, de Moraes NV. A Bayesian Framework for Optimizing Amikacin Therapy in Critically Ill Patients With Cancer. Ther Drug Monit 2025:00007691-990000000-00338. [PMID: 40209112 DOI: 10.1097/ftd.0000000000001324] [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: 09/27/2024] [Accepted: 01/08/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND Amikacin (AMK) is used to treat gram-negative bacterial infections in intensive care unit (ICU) patients. However, its narrow therapeutic range and high interindividual variability can lead to toxicity and ineffectiveness. This study aimed to establish a roadmap for AMK therapeutic drug monitoring in critically ill patients with cancer to provide a Bayesian estimator of bedside applicability. METHODS An observational retrospective study was conducted on oncological patients admitted to the ICU, treated with AMK as a 30-min intravenous infusion at 5.8-39.2 mg/kg. The plasma concentrations were analyzed using a nonlinear mixed-effects modeling approach. Covariate analyses were performed using anthropometric and laboratory data, concomitant drugs, and comorbidities. The model predictive performance was compared with previous AMK dosing approaches using the Bland-Altman method. RESULTS The concentration-time profiles were best described using a one-compartment model with linear elimination. The estimated glomerular filtration rate was a significant covariate of clearance (CL), explaining 16% of the interpatient variability. Body weight was positively correlated with the volume of distribution, accounting for 4% of the variability. Our model reduced the bias in the estimates of individual CL values compared with that of other available methods and was further implemented in DoseMeRx for real-time application at the bedside. CONCLUSIONS This study provides an effective example of a Bayesian estimation method for individualizing AMK doses in critically ill patients with cancer. Collecting more comprehensive patient information, including additional biomarkers for renal function, could further refine the model and improve its predictive performance in this special population.
Collapse
Affiliation(s)
- Priscila Akemi Yamamoto
- Center for Pharmacometrics and System Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida
| | - Leyanis Rodriguez-Vera
- Center for Pharmacometrics and System Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida
- Model Informed Development, Regulatory Development and Consulting, CTI Laboratories, Covington, Kentucky
| | | | - Fernanda Lima Moreira
- Laboratory of Pharmacometrics, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | | | | | | | | | - Valvanera Vozmediano
- Center for Pharmacometrics and System Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida
- Model Informed Development, Regulatory Development and Consulting, CTI Laboratories, Covington, Kentucky
| | - Natalia Valadares de Moraes
- Center for Pharmacometrics and System Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida
| |
Collapse
|
4
|
van der Veer MAA, Smits A, de Haan TR, Franken LGW, van Kaam AH, Hodiamont CJ, Bijleveld YA, Allegaert K, Mathôt RAA. Amikacin dosing in neonates: evaluation of target attainment using a simplified and complex pharmacokinetic model-derived dosing regimen in clinical practice. Antimicrob Agents Chemother 2025; 69:e0111824. [PMID: 40067044 PMCID: PMC11963607 DOI: 10.1128/aac.01118-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 01/13/2025] [Indexed: 04/03/2025] Open
Abstract
Amikacin is frequently used for the treatment of neonatal sepsis. The Dutch Pediatric Formulary recommends a complex pharmacokinetic (PK) model-derived dosing regimen, which consists of dosing categories based on postnatal age and weight that results in adequate PK/pharmacodynamic (PK/PD) target attainment. However, a simplified dosing regimen may be easier to apply in clinical practice. We evaluated PK/PD target attainment of amikacin in neonates using this simplified or complex dosing regimen. This retrospective cohort study included neonates with routinely measured amikacin concentrations at the neonatal intensive care units of the Amsterdam University Medical Center (simplified dosing regimen) or University Hospitals Leuven (complex dosing regimen). Peak (Cmax) and trough (Cmin) concentrations and the area under the concentration-time curve (AUC) for the first dosing interval were calculated by Bayesian estimation for both populations. Targets of Cmax (≥15, ≥25, and ≥35 mg/L), Cmin (≤3 and ≤5 mg/L), and AUC/minimal inhibitory concentration (MIC: 2, 4, and 8 mg/L for Enterobacterales species) for bacteriostasis and 1-log reduction were evaluated. A target attainment of ≥90% was considered adequate. In total, 366 neonates (768 concentrations) and 579 neonates (1,195 concentrations) received the simplified and complex dosing regimen, respectively. Both regimens achieved target attainment of 100% for Cmax ≥ 15 mg/L, Cmin ≤ 5 mg/L, AUC/MIC for bacteriostasis, and AUC/MIC for 1-log reduction up to a MIC of 2 mg/L. Target attainment was achieved for less stringent targets (Cmax ≥ 15 mg/L, Cmin ≤ 5 mg/L, and AUC/MIC for bacteriostasis) with the simplified and complex amikacin dosing regimen. Clinicians can choose one of both dosing regimens, depending on their local circumstances, and the availability of integrated (electronic) prescription tools.
Collapse
Affiliation(s)
- Marlotte A. A. van der Veer
- Department of Pharmacy & Clinical Pharmacology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Anne Smits
- Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Timo R. de Haan
- Department of Neonatology, Emma Children’s Hospital, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Linda G. W. Franken
- Department of Pharmacy & Clinical Pharmacology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Anton H. van Kaam
- Department of Neonatology, Emma Children’s Hospital, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Caspar J. Hodiamont
- Medical Microbiology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Yuma A. Bijleveld
- Department of Pharmacy & Clinical Pharmacology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Karel Allegaert
- Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Ron A. A. Mathôt
- Department of Pharmacy & Clinical Pharmacology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
5
|
Peng ZH, Dickerson VM, Fajt VR, Gould EN, Droog M, Thieman Mankin KM. Serum amikacin concentrations in dogs with naturally occurring open wounds treated with topical amikacin in carboxymethylcellulose hydrogel. Vet Surg 2025; 54:453-460. [PMID: 39651702 PMCID: PMC11947291 DOI: 10.1111/vsu.14195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/28/2024] [Accepted: 11/11/2024] [Indexed: 12/11/2024]
Abstract
OBJECTIVE To quantify serum amikacin concentrations in dogs undergoing wound management with topical amikacin (45 mg/mL) 3% carboxymethylcellulose hydrogel. STUDY DESIGN Prospective clinical study. SAMPLE POPULATION Eleven client-owned dogs. METHODS Dogs with naturally occurring wounds, undergoing treatment with topical amikacin gel, were enrolled. A whole blood sample was collected prior to initial application of the gel. Up to a maximum dose of 30 mg/kg of gel, was applied directly on the wound and the wound was bandaged. Serial blood sampling was performed at approximately 2, 4, 8, 12, 18, 24, 32, 40, 48, 56, 64, and 72 h after application of amikacin gel. The sampling schedule was reset following each bandage change and new application of the gel. Up to 20 samples per dog were collected. The Siemens Syva EMIT Amikacin Assay was used to quantify the concentration of amikacin in each blood serum sample. The lower limit of quantification (LLOQ) of the test was 2.5 μg/mL. RESULTS Amikacin gel was applied a total of 31 times (dose range, 0.1-24.9 mg/kg). A total of 153 samples were analyzed. Five samples in three different dogs were above the LLOQ at approximately 2 h after gel application (range 2.75-3.82 μg/mL). All other samples were below the LLOQ. CONCLUSION Routine use of amikacin gel for open wound management did not result in serum amikacin levels above 5 μg/mL. CLINICAL SIGNIFICANCE Topical amikacin gel may be a safe treatment option for wounds in dogs with resistant infections or biofilms.
Collapse
Affiliation(s)
- Zong H. Peng
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical SciencesTexas A&M UniversityCollege StationTexasUSA
| | - Vanna M. Dickerson
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical SciencesTexas A&M UniversityCollege StationTexasUSA
| | - Virginia R. Fajt
- Department of Veterinary Physiology and Pharmacology, College of Veterinary MedicineTexas A&M UniversityCollege StationTexasUSA
| | - Emily N. Gould
- Gastrointestinal Lab, Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical SciencesTexas A&M UniversityCollege StationTexasUSA
| | - Madeline Droog
- Veterinary Medical Teaching Hospital, College of Veterinary Medicine and Biomedical SciencesTexas A&M UniversityCollege StationTexasUSA
| | - Kelley M. Thieman Mankin
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical SciencesTexas A&M UniversityCollege StationTexasUSA
| |
Collapse
|
6
|
Dupont V, Mourvillier B, Barbe C, Legros V, Jozwiak M, Merdji H, Dupuis C, Winiszewski H, Marchalot A, Lacave G, Neuville M, Sagnier A, Barbier F, Thivilier C, Ruiz S, Smonig R, Rosman J, Argaud L, Grangé S, Sarton B, Chillet P, Voiriot G, Kanagaratnam L, Djerada Z. Amikacin use in critically ill patients requiring renal replacement therapy: the AMIDIAL-ICU study. Ann Intensive Care 2025; 15:42. [PMID: 40133728 PMCID: PMC11937451 DOI: 10.1186/s13613-025-01461-z] [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: 09/03/2024] [Accepted: 03/13/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Acute kidney injury (AKI) requiring renal replacement therapy (RRT) is common in intensive care units (ICUs), yet optimal amikacin dosing in this context remains poorly understood. METHODS We conducted a prospective observational study across 18 French hospitals from April 2020 to January 2022. Adult ICU patients (aged > 18 years) receiving their first amikacin dose while on RRT were included. Data on demographics, RRT modalities, amikacin dosing, and therapeutic drug monitoring were collected. Using a pharmacokinetic modeling approach, we evaluated various amikacin regimens and simulated target attainment probabilities across different minimum inhibitory concentrations (MICs). RESULTS A total of 111 patients were included, with approximately two-thirds receiving continuous RRT. The median amikacin dose was 27 (25-30) mg/kg. Amikacin peak (Cmax) and trough concentrations were monitored in 53 (47.8%) and 76 (68.5%) patients, respectively. Continuous RRT and a history of chronic kidney disease reduced dialytic clearance. For a MIC ≤ 4 mg/L, a 15 mg/kg amikacin dose achieved Cmax/MIC and AUC/MIC targets in ≥ 90% of patients on intermittent dialysis, while 20 mg/kg was required for those on continuous dialysis. For a MIC = 8 mg/L, a 30 mg/kg dose was necessary to achieve Cmax/MIC ≥ 8. CONCLUSIONS Our findings highlight suboptimal adherence to amikacin monitoring guidelines in ICU patients on RRT. Using pharmacokinetic modeling, we identified amikacin dosing recommendations ranging from 15 to 35 mg/kg to optimize efficacy and minimize risks, depending on MIC and dialysis modality.
Collapse
Affiliation(s)
- Vincent Dupont
- Centre Hospitalier Universitaire de Reims, 45 rue Cognacq Jay, Reims, 51092, +33326787641, France.
- Université de Reims Champagne Ardenne, Reims, France.
| | - Bruno Mourvillier
- Centre Hospitalier Universitaire de Reims, 45 rue Cognacq Jay, Reims, 51092, +33326787641, France
- Université de Reims Champagne Ardenne, Reims, France
| | - Coralie Barbe
- Université de Reims Champagne Ardenne, Reims, France
| | - Vincent Legros
- Centre Hospitalier Universitaire de Reims, 45 rue Cognacq Jay, Reims, 51092, +33326787641, France
| | - Mathieu Jozwiak
- Unité de Recherche Clinique Côte d'Azur, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Hamid Merdji
- Centre Hospitalier Universitaire de Strasbourg, Strasbourg, France
| | - Claire Dupuis
- Centre Hospitalier Universitaire de Clermont Ferrand, Clermont Ferrand, France
| | | | | | | | | | - Anne Sagnier
- Centre Hospitalier de Beauvais, Beauvais, France
| | | | | | - Stéphanie Ruiz
- Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | | | - Jeremy Rosman
- Centre Hospitalier Intercommunal Nord Ardennes, Charleville-Mézières, France
| | | | - Steven Grangé
- Centre Hospitalier Universitaire de Rouen, Rouen, France
| | | | - Patrick Chillet
- Centre Hospitalier de Chalons en Champagne, Chalons en Champagne, France
| | - Guillaume Voiriot
- Service de Médecine Intensive Réanimation, Sorbonne Université, Centre de Recherche Saint-Antoine UMRS_938 INSERM, Assistance Publique- Hôpitaux de Paris, Hôpital Tenon, Paris, France
| | - Lukshe Kanagaratnam
- Centre Hospitalier Universitaire de Reims, 45 rue Cognacq Jay, Reims, 51092, +33326787641, France
- Université de Reims Champagne Ardenne, Reims, France
| | - Zoubir Djerada
- Centre Hospitalier Universitaire de Reims, 45 rue Cognacq Jay, Reims, 51092, +33326787641, France
- Université de Reims Champagne Ardenne, Reims, France
| |
Collapse
|
7
|
Trah J, Deindl P, Luister A, Langebrake C, Singer D, Ebenebe CU. Factors associated with elevated gentamicin trough levels in neonates: a retrospective analysis of dosing and clinical parameters. Front Pediatr 2025; 13:1510838. [PMID: 40166658 PMCID: PMC11956741 DOI: 10.3389/fped.2025.1510838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 02/03/2025] [Indexed: 04/02/2025] Open
Abstract
Objective Investigate determinants of elevated gentamicin trough levels in neonates. Methods This single-center retrospective analysis used a multivariate linear regression model to explore the relationship between gentamicin trough concentrations and factors such as creatinine levels, dosage, day of life, sex, CRP levels, and dosing interval in neonates. Results In 215 neonates, including 68 (31.6%) premature neonates with a postmenstrual age of ≤35 weeks, shorter dosing intervals, higher creatinine levels, and increased dosage were linked to higher gentamicin trough levels. Elevated CRP levels corresponded with lower trough levels. Conclusion This study highlights the critical role of dosing frequency, kidney function, and inflammatory status in influencing gentamicin trough levels in neonates. However, all gentamicin trough levels were within the 2 µg/ml threshold.
Collapse
Affiliation(s)
- Julian Trah
- Department of Neonatology and Pediatric Intensive Care Medicine, University Children’s Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department for Pediatric Rehabilitation, VAMED Rehabilitation Hospital, Geesthacht, Germany
| | - Philipp Deindl
- Department of Neonatology and Pediatric Intensive Care Medicine, University Children’s Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alexandra Luister
- Department of Neonatology and Pediatric Intensive Care Medicine, University Children’s Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Claudia Langebrake
- Hospital Pharmacy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dominique Singer
- Department of Neonatology and Pediatric Intensive Care Medicine, University Children’s Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Chinedu Ulrich Ebenebe
- Department of Neonatology and Pediatric Intensive Care Medicine, University Children’s Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
8
|
Kilcoyne I, Nieto J, Nottle BF, Flynn H, Knych HK. Morphine synovial fluid concentrations after intravenous regional limb perfusion in horses during standing sedation. Equine Vet J 2025; 57:502-512. [PMID: 38887833 DOI: 10.1111/evj.14114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 05/16/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND Addition of morphine to the perfusate while performing intravenous regional limb perfusion (IVRLP) may be helpful in treating painful infectious orthopaedic conditions of the distal limb. OBJECTIVES The main objective of this study was to determine synovial morphine concentrations following IVRLP with morphine alone or in combination with amikacin. STUDY DESIGN Randomised cross-over in vivo experiment. METHODS Six horses underwent IVRLP with 0.1 mg/kg morphine sulphate diluted to 60 mL using 0.9% NaCl (M group) or combined with 2 g amikacin and 0.9% NaCl (MA group) with a 2-week washout period between treatments. Synovial fluid was collected from the radiocarpal joint (RCJ) at 10, 20, 30, 120, 240, 480, 720 and 1440 min after IVRLP. The tourniquet was removed after the 30-min sample was collected. Synovial concentrations of morphine and major metabolites were measured using liquid chromatography-tandem mass spectrometry. Amikacin concentrations were quantified by a fluorescence polarisation immunoassay. RESULTS Measurable concentrations of morphine were apparent in the RCJ of all horses. Median CMAX of morphine in the M group was 4753.1 (2115.7-14 934.5) ng/mL and 4477 (3434.3-7363) ng/mL in the MA group (p = 0.5). Median CMAX of synovial amikacin was 322.6 (157.5-1371.6 μg/mL). MAIN LIMITATIONS Limitations include small sample size. Investigators were not blinded to the treatments and a third treatment group where amikacin alone was administered via IVRLP to the study population was not included. CONCLUSIONS IVRLP using morphine is a feasible technique and synovial morphine concentrations were measurable following IVRLP and were not affected when used concurrently with amikacin. Administration of morphine via IVRLP may be beneficial as an analgesic technique for orthopaedic conditions of the distal limb while limiting potential serious systemic side-effects.
Collapse
Affiliation(s)
- Isabelle Kilcoyne
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Jorge Nieto
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Bridget F Nottle
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Harriet Flynn
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Heather K Knych
- K.L. Maddy Equine Analytical Chemistry Laboratory, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| |
Collapse
|
9
|
Coggins SA, Greenberg RG. Pharmacokinetic and Pharmacodynamic Approaches to Optimize Antibiotic Use in Neonates. Clin Perinatol 2025; 52:67-86. [PMID: 39892955 DOI: 10.1016/j.clp.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Newborn infants (particularly those born preterm) are frequently exposed to empiric antibiotics at birth, and antibiotics are among the most commonly prescribed medications in neonatal intensive care units. Challenges in optimizing neonatal antibiotic dosing include: technical and ethical barriers to neonatal pharmacoanalytic study design and sampling, difficulty in extrapolating adult and pediatric data due to unique neonatal physiology, and a lack of validated pharmacodynamic targets specific to neonatal populations. In this review, we summarize basic concepts in pharmacokinetics (PK) and pharmacodynamics (PD), describe pharmacometric strategies utilized in contemporary PK/PD analyses, and review the evolution of PK/PD data guiding neonatal dosing among 3 commonly used antibiotics.
Collapse
Affiliation(s)
- Sarah A Coggins
- Department of Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Division of Neonatology (2 Main NW), Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104, USA.
| | - Rachel G Greenberg
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA; Duke Clinical Research Institute, 300 West Morgan Street Suite 800, Durham, NC 27701, USA
| |
Collapse
|
10
|
Tong DMH, Hughes MSA, Hu J, Pearson JC, Kubiak DW, Dionne BW, Hughes JH. Comparing two-sample log-linear exposure estimation with Bayesian model-informed precision dosing of tobramycin in adult patients with cystic fibrosis. Antimicrob Agents Chemother 2025; 69:e0104024. [PMID: 39791873 PMCID: PMC11823644 DOI: 10.1128/aac.01040-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 12/10/2024] [Indexed: 01/12/2025] Open
Abstract
Tobramycin dosing in patients with cystic fibrosis (CF) is challenged by its high pharmacokinetic (PK) variability and narrow therapeutic window. Doses are typically individualized using two-sample log-linear regression (LLR) to quantify the area under the concentration-time curve (AUC). Bayesian model-informed precision dosing (MIPD) may allow dose individualization with fewer samples; however, the relative performance of these methods is unknown. This single-center retrospective analysis included adult patients with CF receiving tobramycin from 2015 to 2022. Tobramycin concentrations were predicted using LLR or Bayesian estimation with two population PK models (Hennig and Alghanem). Then, both methods were used to estimate the AUC for simulated patients. For Bayesian estimation, AUC estimation with flattened priors and limited sampling strategies were also assessed. Predictions were evaluated using normalized root mean square error (nRMSE), mean percent error (MPE), and accuracy. The data set included 70 treatment courses, with 32 not evaluable by LLR due to detection limits or timing issues. Bayesian estimation demonstrated worse accuracy (47.1%-50.7% vs 75.7%), higher MPE (24.2%-32.4% vs -2.4%), and higher nRMSE (35.0%-39.4% vs 24.8%) than LLR for peak concentrations but performed better on troughs (accuracy: 92.0%-92.9% vs 84.6%). Bayesian estimation with flattened priors and a single sample at 4 h was comparable to LLR performance, with better accuracy (42.9%-68.0% vs 41.1% LLR), comparable MPE (-2.3% to -3.7% vs -0.5%) and nRMSE (11.3%-21.6% vs 17.3%). Bayesian estimation with one concentration and flattened priors can match LLR prediction accuracy. However, popPK models must be improved to better estimate peak samples.
Collapse
Affiliation(s)
| | | | - Jasmine Hu
- InsightRX, San Francisco, California, USA
| | - Jeffrey C. Pearson
- Department of Pharmacy, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - David W. Kubiak
- Department of Pharmacy, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Brandon W. Dionne
- Department of Pharmacy, Brigham and Women's Hospital, Boston, Massachusetts, USA
- School of Pharmacy and Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts, USA
| | | |
Collapse
|
11
|
Schellong P, Joean O, Pletz MW, Hagel S, Weis S. Treatment of Complicated Gram-Positive Bacteremia and Infective Endocarditis. Drugs 2025; 85:193-214. [PMID: 39720961 PMCID: PMC11802659 DOI: 10.1007/s40265-024-02135-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2024] [Indexed: 12/26/2024]
Abstract
The Gram-positive cocci Staphylococcus aureus, Streptococcus spp., and Enterococcus spp. are the most frequent causative organisms of bloodstream infections and infective endocarditis. "Complicated bacteremia" is a term used in S. aureus bloodstream infections and originally implied the presence of metastatic infectious foci (i.e. complications of S. aureus bacteremia). These complications demand longer antimicrobial treatment durations and, frequently, interventional source control. Several risk factors for the incidence of bacteremia complications have been identified and are often used for the definition of complicated bacteremia. Here, we discuss management and diagnostic approaches and treatment options for patients with complicated bacteremia, with particular focus on infective endocarditis. We also summarize the available evidence regarding imaging modalities and the choice of antimicrobial mono- or combination therapy according to resistance patterns for these pathogens as well as treatment durations and optimized application routes. Finally, we synopsize current and future areas of research in complicated bacteremia and infective endocarditis.
Collapse
Affiliation(s)
- Paul Schellong
- Institute for Infectious Disease and Infection Control, Jena University Hospital, Friedrich-Schiller-University, Am Klinikum 1, 07749, Jena, Germany.
- Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute, Jena, Germany.
| | - Oana Joean
- Institute for Infectious Disease and Infection Control, Jena University Hospital, Friedrich-Schiller-University, Am Klinikum 1, 07749, Jena, Germany
| | - Mathias W Pletz
- Institute for Infectious Disease and Infection Control, Jena University Hospital, Friedrich-Schiller-University, Am Klinikum 1, 07749, Jena, Germany
| | - Stefan Hagel
- Institute for Infectious Disease and Infection Control, Jena University Hospital, Friedrich-Schiller-University, Am Klinikum 1, 07749, Jena, Germany
| | - Sebastian Weis
- Institute for Infectious Disease and Infection Control, Jena University Hospital, Friedrich-Schiller-University, Am Klinikum 1, 07749, Jena, Germany
- Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute, Jena, Germany
- Department of Anaesthesiology and Intensive Care Medicine, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany
| |
Collapse
|
12
|
Cerenzio J, Andrade J, DeAngelis J, Truong J. The Effect of a Single-Dose Aminoglycoside With a Beta-Lactam for the Treatment of Gram-Negative Bacteremia. J Pharm Pract 2025; 38:141-148. [PMID: 39185751 DOI: 10.1177/08971900241273212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Introduction: Aminoglycosides possess activity against aerobic gram-negative organisms and are often used in combination with beta-lactam antibiotics. Previous studies evaluating combination therapy in gram-negative bacteremia have not shown clear benefits, however antimicrobial resistance was not prevalent in these studies. Our objective is to elucidate potential benefits of adding a single dose of an aminoglycoside to a beta-lactam in patients with gram-negative bacteremia. Methods: This study was a single-center, retrospective, cohort study including patients 18 years old or older and treated for at least 24 hours for a confirmed gram-negative bacteremia. Patients were divided into two groups: receipt of beta-lactam monotherapy (n = 164) and receipt of a beta-lactam in addition to a single dose of an aminoglycoside (n = 79) within 24 hours of bacteremia onset. The primary endpoint was infection-related 30-day mortality per provider documentation. Key secondary outcomes include incidence of acute kidney injury (AKI) and time to improvement of AKI. Data were analyzed using Chi-square or Fisher's exact tests, student's T test, and descriptive statistics as appropriate. Results: The primary outcome occurred in 13/164 vs 2/79 patients in the monotherapy and combination groups (P = 0.10). Incidence of AKI (14% vs. 12%) and time to recovery from AKI (90 hours; IQR [50 - 133] vs 78 hours; IQR [42 - 128]) were comparable between groups (P = 1.00 and P = 0.73, respectively). Conclusions: The addition of a single-dose aminoglycoside was not significantly associated with reduced mortality or increased time to recovery from AKI in our patient population. Larger studies, particularly in more severely ill patient populations, are needed.
Collapse
Affiliation(s)
| | - Justin Andrade
- The Brooklyn Hospital Center, Brooklyn, NY, USA
- Touro College of Pharmacy, New York, NY, USA
| | - Joanna DeAngelis
- Staten Island University Hospital - North Campus, Staten Island, NY, USA
| | - James Truong
- NewYork-Presbyterian Queens Hospital, Queens, NY, USA
| |
Collapse
|
13
|
Onita T, Ishihara N, Yano T. PK/PD-Guided Strategies for Appropriate Antibiotic Use in the Era of Antimicrobial Resistance. Antibiotics (Basel) 2025; 14:92. [PMID: 39858377 PMCID: PMC11759776 DOI: 10.3390/antibiotics14010092] [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: 12/19/2024] [Revised: 01/11/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Antimicrobial resistance (AMR) poses a critical global health threat, necessitating the optimal use of existing antibiotics. Pharmacokinetic/pharmacodynamic (PK/PD) principles provide a scientific framework for optimizing antimicrobial therapy, particularly to respond to evolving resistance patterns. This review examines PK/PD strategies for antimicrobial dosing optimization, focusing on three key aspects. First, we discuss the importance of drug concentration management for enhancing efficacy while preventing toxicity, considering various patient populations, including pediatric and elderly patients with their unique physiological characteristics. Second, we analyze different PK modeling approaches: the classic top-down approach exemplified by population PK analysis, the bottom-up approach represented by physiologically based PK modeling, and hybrid models combining both approaches for enhanced predictive performance. Third, we explore clinical applications, including nomogram-based dosing strategies, Bayesian estimation, and emerging artificial intelligence applications, for real-time dose optimization. Critical challenges in implementing PK/PD simulation are addressed, particularly the selection of appropriate PK models, the optimization of PK/PD indices, and considerations concerning antimicrobial concentrations at infection sites. Understanding these principles and challenges is crucial for optimizing antimicrobial therapy and combating AMR through improved dosing strategies.
Collapse
Affiliation(s)
| | | | - Takahisa Yano
- Department of Pharmacy, Shimane University Hospital, 89-1 Enya, Izumo 693-8501, Shimane, Japan
| |
Collapse
|
14
|
Grzeskowiak LE, Wynne S, Stark MJ. A Quantitative Examination and Comparison of the Ability of Australian Gentamicin Dosing Guidelines to Achieve Target Therapeutic Concentrations in Neonates. Antibiotics (Basel) 2025; 14:48. [PMID: 39858334 PMCID: PMC11759765 DOI: 10.3390/antibiotics14010048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 12/29/2024] [Accepted: 01/01/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Effective gentamicin dosing is crucial to the survival of neonates with suspected sepsis but requires a careful balance between attaining both effective peak and safe trough concentrations. We aimed to systematically compare existing gentamicin dosing guidelines for neonates in Australia to determine the extent to which they reach therapeutic targets. Methods: Simulations of a single gentamicin dose to a virtual representative neonatal population according to each Australian guideline were performed using population pharmacokinetic modelling. We determined the proportion of neonates who would achieve peak gentamicin concentrations of ≥5 or ≥10 mg/L and trough concentrations of ≤1 or ≤2 mg/L. We calculated the probability of target attainment (PTA) according to gestation at birth (22 to 40 weeks) and postnatal age (1-7, 8-14, 15-21, 22-28 days). Results: Five unique dosing guidelines were identified. Guidelines varied considerably with respect to dose (4.5 to 7 mg/kg), dosing interval (24 to 48 h), and characteristics used to individualise dosing regimens (e.g., gestation at birth and postnatal age). Guidelines were satisfactory in routinely achieving effective peak concentrations ≥ 5 mg/L, but PTAs for effective peak concentrations ≥ 10 mg/L varied considerably from 5% to 100% based on dose, gestation, and postnatal age. PTAs for trough concentrations ≤ 1 mg/L ranged from 0% to 100%, being lowest among extremely preterm infants. Conclusions: Current Australian gentamicin guidelines demonstrate significant variability in their ability to achieve defined therapeutic targets, necessitating efforts to improve standardisation of dosing recommendations. Further research to define optimal pharmacodynamic targets in neonates with respect to clinical outcomes are also urgently warranted.
Collapse
Affiliation(s)
- Luke E. Grzeskowiak
- College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia
- Adelaide Medical School, Robinson Research Institute, The University of Adelaide, Adelaide 5005, Australia;
- Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide 5000, Australia
- SA Pharmacy, Flinders Medical Centre, SA Health, Adelaide 5042, Australia;
| | - Sheree Wynne
- SA Pharmacy, Flinders Medical Centre, SA Health, Adelaide 5042, Australia;
| | - Michael J. Stark
- Adelaide Medical School, Robinson Research Institute, The University of Adelaide, Adelaide 5005, Australia;
- Department of Neonatal Medicine, Women’s and Children’s Hospital, Adelaide 5006, Australia
| |
Collapse
|
15
|
Hidalgo NJ, Juvany M, Guillaumes S, Hoyuela C, Vidal Ó, Pera M. Effect of topical gentamicin in preventing surgical site infection in elective incisional hernia repair in a randomized controlled trial. Sci Rep 2024; 14:28755. [PMID: 39567596 PMCID: PMC11579354 DOI: 10.1038/s41598-024-80112-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 11/14/2024] [Indexed: 11/22/2024] Open
Abstract
Surgical site infection (SSI) continues to be a common complication of surgery. The real benefit of using topical antibiotics for the prevention of SSI in abdominal hernia repair surgery is still unknown. This study aimed to evaluate the usefulness of topical gentamicin in SSI prophylaxis in incisional hernia repair with mesh. A randomized controlled trial was conducted in patients undergoing open incisional hernia repair. Patients were randomly assigned to one of two groups: in the gentamicin group, each layer of the abdominal wall was irrigated with gentamicin solution before wound closure, and in the saline solution group (placebo), each layer of the abdominal wall was irrigated with normal saline solution. The incidence of SSI and other surgical site complications was compared between both groups, and the presence of adverse effects with the use of topical gentamicin. Data from 146 patients were included for analysis: 74 in the gentamicin group and 72 in the saline solution group. SSI was observed in six patients (8.1%) in the gentamicin group and eight patients (11.1%) in the saline solution group, with no significant differences (p = 0.538) between both groups. No statistically significant differences were observed in the presentation of seroma, hematoma, and surgical wound dehiscence between both groups. No adverse effects were reported from topical application of gentamicin. In this clinical trial, the use of topical gentamicin in incisional hernia repair with mesh did not significantly reduce the incidence of SSI. EU Clinical Trials Register: EudraCT 2018-001860-45 (04/07/2019).
Collapse
Affiliation(s)
- Nils Jimmy Hidalgo
- Department of General and Digestive Surgery, Hospital Clínic Barcelona, Barcelona, 08036, Spain.
| | | | - Salvador Guillaumes
- Department of General and Digestive Surgery, Hospital Clínic Barcelona, Barcelona, 08036, Spain
| | - Carlos Hoyuela
- Department of Surgery, Hospital Universitario Mollet, Mollet, 08100, Spain
| | - Óscar Vidal
- Department of General and Digestive Surgery, Hospital Clínic Barcelona, Barcelona, 08036, Spain
| | - Miguel Pera
- Department of General and Digestive Surgery, Hospital Clínic Barcelona, Barcelona, 08036, Spain
| |
Collapse
|
16
|
Ariano RE, Zelenitsky SA, Davis C, Sathianathan C, Wolowich WR. Comparative simulation of intraperitoneal aminoglycoside regimens for patients with peritonitis on automated peritoneal dialysis. Perit Dial Int 2024; 44:438-444. [PMID: 38288583 DOI: 10.1177/08968608231221062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Intraperitoneal (IP) aminoglycosides (AGs) continue to be the cornerstone of empiric management of peritonitis. AG dosing during automated peritoneal dialysis (APD), however, has not been well studied in patients with peritonitis. We sought to identify differences in AG exposure in the peritoneum and plasma for two different dosing regimens with little supporting evidence in patients on APD with peritonitis. METHODS A retrospective design that utilised the peritoneal and plasma concentration-time data from a prior study of 18 continuous ambulatory peritoneal dialysis (CAPD) patients with peritonitis to generate an in silico peritoneal and plasma PK model. This model was then used to compare via simulation using Phoenix© WinNonlin Software with IP AG dosing for a loading-dose regimen (1.5 mg/kg first dose) versus a fixed-dose regimen (0.6 mg/kg/d) in patients on APD with peritonitis. RESULTS Outcome measures were (1) percentage of time where peritoneal peak concentrations/minimal inhibitory concentration (MIC) ratio >10, (2) AUC/MIC > 74 and (3) plasma Cmin concentrations. Both regimens resulted in > 90% optimal peak/MIC ratio and AUC/MIC ratios on days 1 and 5 of the dose protocol. The loading-dose regimen resulted in IP exposures that were 2.5 times greater in the peritoneal compartment on day 1. By day 5, both protocols resulted in similar accumulation of AG plasma Cmin concentrations of 2.5-3.4 mg/L versus 2.4-3.3 mg/L, respectively, for the loading-dose regimen versus fixed-dose regimen. CONCLUSIONS The current international guidelines for the treatment of peritoneal dialysis-associated peritonitis can continue to recommend the fixed-dose regimen for those on APD with the addition of plasma Cmin monitoring after 3 days to assess for drug accumulation.
Collapse
Affiliation(s)
- Robert E Ariano
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
- St. Boniface Hospital, Winnipeg, Canada
| | - Sheryl A Zelenitsky
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
- St. Boniface Hospital, Winnipeg, Canada
| | - Christine Davis
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
- St. Boniface Hospital, Winnipeg, Canada
- Manitoba Renal Program, Winnipeg, Canada
| | | | - William R Wolowich
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA
| |
Collapse
|
17
|
Khatibzadeh SM, Dahlgren LA, Caswell CC, Ducker WA, Werre SR, Bogers SH. Equine bone marrow-derived mesenchymal stromal cells reduce established S. aureus and E. coli biofilm matrix in vitro. PLoS One 2024; 19:e0312917. [PMID: 39480794 PMCID: PMC11527187 DOI: 10.1371/journal.pone.0312917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 10/16/2024] [Indexed: 11/02/2024] Open
Abstract
Biofilms reduce antibiotic efficacy and lead to complications and mortality in human and equine patients with orthopedic infections. Equine bone marrow-derived mesenchymal stromal cells (MSC) kill planktonic bacteria and prevent biofilm formation, but their ability to disrupt established orthopedic biofilms is unknown. Our objective was to evaluate the ability of MSC to reduce established S. aureus or E. coli biofilms in vitro. We hypothesized that MSC would reduce biofilm matrix and colony-forming units (CFU) compared to no treatment and that MSC combined with the antibiotic, amikacin sulfate, would reduce these components more than MSC or amikacin alone. MSC were isolated from 5 adult Thoroughbred horses in antibiotic-free medium. 24-hour S. aureus or E. coli biofilms were co-cultured in triplicate for 24 or 48 hours in a transwell plate system: untreated (negative) control, 30 μg/mL amikacin, 1 x 106 passage 3 MSC, and MSC with 30 μg/mL amikacin. Treated biofilms were photographed and biofilm area quantified digitally. Biomass was quantified via crystal violet staining, and CFU quantified following enzymatic digestion. Data were analyzed using mixed model ANOVA with Tukey post-hoc comparisons (p < 0.05). MSC significantly reduced S. aureus biofilms at both timepoints and E. coli biofilm area at 48 hours compared to untreated controls. MSC with amikacin significantly reduced S. aureus biofilms versus amikacin and E. coli biofilms versus MSC at 48 hours. MSC significantly reduced S. aureus biomass at both timepoints and reduced S. aureus CFU at 48 hours versus untreated controls. MSC with amikacin significantly reduced S. aureus biomass versus amikacin at 24 hours and S. aureus and E. coli CFU versus MSC at both timepoints. MSC primarily disrupted the biofilm matrix but performed differently on S. aureus versus E. coli. Evaluation of biofilm-MSC interactions, MSC dose, and treatment time are warranted prior to testing in vivo.
Collapse
Affiliation(s)
- Sarah M. Khatibzadeh
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States of America
| | - Linda A. Dahlgren
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States of America
| | - Clayton C. Caswell
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, Virginia, United States of America
| | - William A. Ducker
- Department of Chemical Engineering, College of Engineering, Virginia Tech, Blacksburg, VA, United States of America
| | - Stephen R. Werre
- Laboratory for Study Design and Statistical Analysis, Virginia-Maryland College of Veterinary Medicine, Blacksburg, Virginia, United States of America
| | - Sophie H. Bogers
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States of America
| |
Collapse
|
18
|
Wu M, Feng K, Wu X, Liu C, Zhu S, Martins FS, Yu M, Lv Z, Yan M, Sy SKB. Prediction of tissue exposures of polymyxin-B, amikacin and sulbactam using physiologically-based pharmacokinetic modeling. Front Microbiol 2024; 15:1435906. [PMID: 39435440 PMCID: PMC11491386 DOI: 10.3389/fmicb.2024.1435906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/19/2024] [Indexed: 10/23/2024] Open
Abstract
Background The combination antimicrobial therapy consisting of amikacin, polymyxin-B, and sulbactam demonstrated in vitro synergy against multi-drug resistant Acinetobacter baumannii. Objectives The objectives were to predict drug disposition and extrapolate their efficacy in the blood, lung, heart, muscle and skin tissues using a physiologically-based pharmacokinetic (PBPK) modeling approach and to evaluate achievement of target pharmacodynamic (PD) indices against A. baumannii. Methods A PBPK model was initially developed for amikacin, polymyxin-B, and sulbactam in adult subjects, and then scaled to pediatrics, accounting for both renal and non-renal clearances. The simulated plasma and tissue drug exposures were compared to the observed data from humans and rats. Efficacy was inferred using joint probability of target attainment of target PD indices. Results The simulated plasma drug exposures in adults and pediatrics were within the 0.5 to 2 boundary of the mean fold error for the ratio between simulated and observed means. Simulated drug exposures in blood, skin, lung, and heart were consistent with reported penetration ratio between tissue and plasma drug exposure. In a virtual pediatric population from 2 to <18 years of age using pediatric dosing regimens, the interpretive breakpoints were achieved in 85-90% of the population. Conclusion The utility of PBPK to predict and simulate the amount of antibacterial drug exposure in tissue is a practical approach to overcome the difficulty of obtaining tissue drug concentrations in pediatric population. As combination therapy, amikacin/polymyxin-B/sulbactam drug concentrations in the tissues exhibited sufficient penetration to combat extremely drug resistant A. baumannii clinical isolates.
Collapse
Affiliation(s)
- Mengyuan Wu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Kun Feng
- Women and Children Hospital, Qingdao University, Qingdao, China
| | - Xiao Wu
- Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Chang Liu
- Women and Children Hospital, Qingdao University, Qingdao, China
| | - Shixing Zhu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Frederico S. Martins
- Department of Clinical and Toxicological Analysis, Faculty of Pharmaceutical Sciences, Universidade de São Paulo, São Paulo, Brazil
| | - Mingming Yu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Zhihua Lv
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Meixing Yan
- Women and Children Hospital, Qingdao University, Qingdao, China
| | - Sherwin K. B. Sy
- Department of Statistics, Universidade Estadual de Maringá, Maringá, Paraná, Brazil
| |
Collapse
|
19
|
Zhao X, Ren J, Zheng X, Wang N, Wu Z, Liu H, Tian W, Zhao D, Xu Y, Han X. A phase I clinical study: Evaluation of safety, tolerability, and population pharmacokinetic-pharmacodynamic target attainment analysis of etimicin sulfate among healthy chinese participants. Int J Antimicrob Agents 2024; 64:107287. [PMID: 39084574 DOI: 10.1016/j.ijantimicag.2024.107287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 07/11/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND This phase I clinical study aimed to assess the safety, tolerability, and population pharmacokinetic-pharmacodynamics (PK-PD) target attainment analysis of etimicin sulfate in healthy participants, and to provide scientific reference for further development of clinical breakpoints. METHODS A total of 24 healthy Chinese subjects were enrolled in this study and received an etimicin sulfate infusion. A population PK model was constructed for the estimation of the PK profiles of etimicin sulfate. The area under the concentration-time curve divided by the minimum inhibitory concentration (AUC0-24h/MIC) and the peak concentration divided by the MIC (Cmax/MIC) were selected as the PK/PD indices. The probability of target attainment (PTA) was calculated for each designed dosing regimen using Monte Carlo simulations. The minimum MIC value with a PTA ≥ 90% for each regimen was considered as the PK/PD cutoff values. RESULTS Etimicin sulfate demonstrated safety, tolerability, and predictable PK characteristics. No deaths or serious adverse events were reported. Seven treatment-emergent adverse events (TEAEs) were reported by five participants; all TEAEs were minor and were rapidly relieved. A two-compartment model was developed and validated for describing the PK features of etimicin sulfate among Chinese healthy participants. The diagnostic goodness-of-fit plots and visual predictive check plots showed that this developed model could describe these data well. CONCLUSIONS The PTA results showed that etimicin sulfate provided clinical improvement against strains with an MIC of 0.5-1 mg/L and below, and antibacterial effect against strains with an MIC of 0.25 mg/L and below. However, etimicin sulfate had limited clinical efficacy for clinical isolates with MIC values > 1 mg/L.
Collapse
Affiliation(s)
- Xue Zhao
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jianwei Ren
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xin Zheng
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Na Wang
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhen Wu
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Hongzhong Liu
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wei Tian
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - DanDan Zhao
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yingchun Xu
- Department of Laboratory Medicine and Beijing Key Laboratory for Mechanisms Research and Precision Diagnosis of Invasive Fungal Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaohong Han
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
20
|
Paegelow JL, Schoonover MJ, Young JM, Maxwell LK, Taylor JD, Gilliam LL, Holbrook TC. Pharmacokinetics of amikacin after intravenous, intra-articular, and combined intravenous and intra-articular administration in healthy neonatal foals. J Vet Intern Med 2024; 38:1825-1834. [PMID: 38647141 PMCID: PMC11099780 DOI: 10.1111/jvim.17087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 04/09/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Pharmacokinetics of amikacin administered IV to neonatal foals are described, but little data are available regarding the plasma concentrations contributed by concurrent intra-articular (IA) administration. HYPOTHESIS/OBJECTIVES Compare the pharmacokinetics of amikacin when the total dose is administered IV compared to being divided between IV and IA routes of administration in neonatal foals and predict the plasma concentrations from various combined IV and IA dosing regimens. ANIMALS Eight healthy neonatal foals. METHODS Foals received 3 amikacin treatment protocols: (1) IV-only (25 mg/kg q24h IV), (2) concurrent IV and IA (16.7 mg/kg q24h IV and 8.3 mg/kg q24h into 1 tarsocrural joint), and (3) IA-only (8.3 mg/kg q24h into 1 tarsocrural joint). Protocols were administered for 3 days beginning at 7, 14, and 21 days of age. Plasma concentrations ≥53 μg/mL at 30 minutes were considered therapeutic for isolates with intermediate susceptibility. RESULTS Foal age was a significant variable. The IV-only protocol met or exceeded the 30-minute plasma concentrations considered therapeutic (mean μg/mL [95% confidence interval, CI]) in 7- to 9-day-old (54.0 [52.2-56.9]), 14- to 16-day-old (58.1 [55.2-61.0]), and 21- to 23-day-old (66.6 [63.7-69.6]) foals. Concurrent IV and IA protocol did not reach the 30-minute concentration considered therapeutic in 7- to 9-day-old foals (46.5 [43.6-49.4]) but did in 14- to 16-day-old (62.9 [60.0-65.8]) and 21-to 23-day-old (62.6 [59.7-65.6]) foals. CONCLUSIONS AND CLINICAL IMPORTANCE Concurrent IV and IA administration of amikacin produces 30-minute plasma concentrations considered therapeutic in foals 14 to 23 days old, but concentrations observed in younger foals might be below those considered therapeutic for isolates with intermediate susceptibility to amikacin.
Collapse
Affiliation(s)
- Jillian L. Paegelow
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineOklahoma State UniversityStillwaterOklahomaUSA
- Present address:
Innovative Equine ServicesRogersvilleMissouriUSA
| | - Mike J. Schoonover
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineOklahoma State UniversityStillwaterOklahomaUSA
| | - Jenna M. Young
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineOklahoma State UniversityStillwaterOklahomaUSA
- Present address:
Department of Veterinary Population Medicine, College of Veterinary MedicineUniversity of MinnesotaSt. PaulMinnesotaUSA
| | - Lara K. Maxwell
- Department of Physiological Sciences, College of Veterinary MedicineOklahoma State UniversityStillwaterOklahomaUSA
| | - Jared D. Taylor
- Department of Veterinary Pathobiology, College of Veterinary MedicineOklahoma State UniversityStillwaterOklahomaUSA
| | - Lyndi L. Gilliam
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineOklahoma State UniversityStillwaterOklahomaUSA
| | - Todd C. Holbrook
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineOklahoma State UniversityStillwaterOklahomaUSA
- Present address:
Department of Large Animal Clinical Sciences, College of Veterinary MedicineUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
21
|
Poulain C, Launey Y, Bouras M, Lakhal K, Dargelos L, Crémet L, Gibaud SA, Corvec S, Seguin P, Rozec B, Asehnoune K, Feuillet F, Roquilly A. Clinical evaluation of the BioFire Respiratory Pathogen Panel for the guidance of empirical antimicrobial therapy in critically ill patients with hospital-acquired pneumonia: A multicenter, quality improvement project. Anaesth Crit Care Pain Med 2024; 43:101353. [PMID: 38355044 DOI: 10.1016/j.accpm.2024.101353] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 01/13/2024] [Accepted: 01/14/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND We aimed to determine whether implementing antimicrobial stewardship based on multiplex bacterial PCR examination of respiratory fluid can enhance outcomes of critically ill patients with hospital-acquired pneumonia (HAP). METHODS We conducted a quality improvement study in two hospitals in France. Adult patients requiring invasive mechanical ventilation with a diagnosis of HAP were included. In the pre-intervention period (August 2019 to April 2020), antimicrobial therapy followed European guidelines. In the «intervention» phase (June 2020 to October 2021), treatment followed a multiplex PCR-guided protocol. The primary endpoint was a composite endpoint made of mortality on day 28, clinical cure between days 7 and 10, and duration of invasive mechanical ventilation on day 28. The primary outcome was analyzed with a DOOR strategy. RESULTS A total of 443 patients were included in 3 ICUs from 2 hospitals (220 pre-intervention; 223 intervention). No difference in the ranking of the primary composite outcome was found (DOOR: 50.3%; 95%CI, 49.9%-50.8%). The number of invasive mechanical ventilation-free days at day 28 was 10.0 [0.0; 19.0] in the baseline period and 9.0 [0.0; 20.0] days during the intervention period (p = 0.95). The time-to-efficient antimicrobial treatment was 0.43 ± 1.29 days before versus 0.55 ± 1.13 days after the intervention (p = 0.56). CONCLUSION Implementation of Rapid Multiplex PCR to guide empirical antimicrobial therapy for critically ill patients with HAP was not associated with better outcomes. However, adherence to stewardship was low, and the study may have had limited power to detect a clinically important difference.
Collapse
Affiliation(s)
- Cécile Poulain
- Nantes Université, CHU Nantes, INSERM, Anesthesie Réanimation, CIC 0004, F-44000 Nantes, France; Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000, Nantes, France.
| | - Yoann Launey
- Univ Rennes, CHU Rennes, Department of Anaesthesia, Critical Care and Perioperative Medicine, F-35000 Rennes, France
| | - Marwan Bouras
- Nantes Université, CHU Nantes, INSERM, Anesthesie Réanimation, CIC 0004, F-44000 Nantes, France; Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000, Nantes, France
| | - Karim Lakhal
- Nantes Université, CHU Nantes, INSERM, Anesthesie Réanimation, CIC 0004, F-44000 Nantes, France
| | - Laura Dargelos
- Nantes Université, CHU Nantes, INSERM, Anesthesie Réanimation, CIC 0004, F-44000 Nantes, France
| | - Lise Crémet
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000, Nantes, France; Nantes Université, CHU Nantes, Service de bactériologie-hygiène, pôle de biologie, Nantes, France
| | - Sophie-Anne Gibaud
- Nantes Université, CHU Nantes, Service de bactériologie-hygiène, pôle de biologie, Nantes, France
| | - Stéphane Corvec
- Nantes Université, CHU Nantes, Service de bactériologie-hygiène, pôle de biologie, Nantes, France
| | - Philippe Seguin
- Univ Rennes, CHU Rennes, Department of Anaesthesia, Critical Care and Perioperative Medicine, F-35000 Rennes, France
| | - Bertrand Rozec
- Nantes Université, CHU Nantes, INSERM, Anesthesie Réanimation, CIC 0004, F-44000 Nantes, France
| | - Karim Asehnoune
- Nantes Université, CHU Nantes, INSERM, Anesthesie Réanimation, CIC 0004, F-44000 Nantes, France; Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000, Nantes, France
| | - Fanny Feuillet
- Nantes Université, CHU de Nantes, DRI, Département promotion, cellule vigilances, Nantes, France; Nantes Université, CHU de Nantes, DRI, Plateforme de Méthodologie et de Biostatistique, Nantes, France
| | - Antoine Roquilly
- Nantes Université, CHU Nantes, INSERM, Anesthesie Réanimation, CIC 0004, F-44000 Nantes, France; Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000, Nantes, France
| |
Collapse
|
22
|
Darlow CA, Parrott N, Peck RW, Hope W. Development and application of neonatal physiology-based pharmacokinetic models of amikacin and fosfomycin to assess pharmacodynamic target attainment. CPT Pharmacometrics Syst Pharmacol 2024; 13:464-475. [PMID: 38108548 PMCID: PMC10941605 DOI: 10.1002/psp4.13097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/14/2023] [Accepted: 11/22/2023] [Indexed: 12/19/2023] Open
Abstract
Antimicrobial resistance increasingly complicates neonatal sepsis in a global context. Fosfomycin and amikacin are two agents being tested in an ongoing multicenter neonatal sepsis trial. Although neonatal pharmacokinetics (PKs) have been described for these drugs, the physiological variability within neonatal populations makes population PKs in this group uncertain. Physiologically-based pharmacokinetic (PBPK) models were developed in Simcyp for fosfomycin and amikacin sequentially for adult, pediatric, and neonatal populations, with visual and quantitative validation compared to observed data at each stage. Simulations were performed using the final validated neonatal models to determine drug exposures for each drug across a demographic range, with probability of target attainment (PTA) assessments. Successfully validated neonatal PBPK models were developed for both fosfomycin and amikacin. PTA analysis demonstrated high probability of target attainment for amikacin 15 mg/kg i.v. q24h and fosfomycin 100 mg/kg (in neonates aged 0-7 days) or 150 mg/kg (in neonates aged 7-28 days) i.v. q12h for Enterobacterales with fosfomycin and amikacin minimum inhibitory concentrations at the adult breakpoints. Repeat analysis in premature populations demonstrated the same result. PTA analysis for a proposed combination fosfomycin-amikacin target was also performed. The simulated regimens, tested in a neonatal sepsis trial, are likely to be adequate for neonates across different postnatal ages and gestational age. This work demonstrates a template for determining target attainment for antimicrobials (alone or in combination) in special populations without sufficient available PK data to otherwise assess with traditional pharmacometric methods.
Collapse
Affiliation(s)
- Christopher A. Darlow
- Antimicrobial Pharmacodynamics and Therapeutics, Department of PharmacologyUniversity of LiverpoolLiverpoolUK
| | - Neil Parrott
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre BaselBaselSwitzerland
| | - Richard W. Peck
- Antimicrobial Pharmacodynamics and Therapeutics, Department of PharmacologyUniversity of LiverpoolLiverpoolUK
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre BaselBaselSwitzerland
| | - William Hope
- Antimicrobial Pharmacodynamics and Therapeutics, Department of PharmacologyUniversity of LiverpoolLiverpoolUK
| |
Collapse
|
23
|
Gibson JE, Nandanwar N, Neely MN. Time-dependent pharmacodynamics of amikacin on Mycobacterium abscessus growth and resistance emergence. Microbiol Spectr 2024; 12:e0322223. [PMID: 38236037 PMCID: PMC10846206 DOI: 10.1128/spectrum.03222-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 12/07/2023] [Indexed: 01/19/2024] Open
Abstract
Mycobacterium abscessus pulmonary disease is increasing in prevalence globally, particularly for individuals with cystic fibrosis. These infections are challenging to treat due to a high rate of resistance. Amikacin is critical to treatment, but the development of toxicity, amikacin resistance, and treatment failure are significant challenges. Amikacin has been characterized previously as peak-dependent and extended-interval dosing is commonly used. In our hollow fiber infection model of M. abscessus, amikacin exhibited time-dependent rather than the expected peak-dependent pharmacodynamics. Humanized amikacin exposures with more frequent, short-interval dosing (continuous infusion or every 12 hours) yielded improved microbiological response compared to extended-interval dosing (every 24 hours or 1-3 times per week). Short-interval dosing inhibited growth with a mean (SD) maximum Δlog10 colony forming units of -4.06 (0.52), significantly more than extended-interval dosing (P = 0.0013) every 24 hours, -2.40 (0.58), or 1-3 times per week, -2.39 (0.38). Growth recovery, an indicator of resistance emergence, occurred at 6.56 (0.70) days with short-interval dosing but was significantly earlier with extended-interval dosing (P = 0.0032) every 24 hours, 3.88 (0.85) days, and 1-3 times per week, 3.27 (1.72) days. Microbiological response correlated best with the pharmacodynamic index of %T > minimum inhibitory concentration (MIC), with an EC80 for growth inhibition of ~40%T > MIC. We used a previously published population model of amikacin to determine the probability of achieving 40%T > MIC and show that current dosing strategies are far below this target, which may partially explain why treatment failure remains so high for these infections. These data support a cautious approach to infrequent amikacin dosing for the treatment of M. abscessus.IMPORTANCEPulmonary disease caused by Mycobacterium abscessus complex (MABSC) is increasing worldwide, particularly in patients with cystic fibrosis. MABSC is challenging to treat due to high levels of antibiotic resistance. Treatment requires 2-4 antibiotics over more than 12 months and has a significant risk of toxicity but still fails to eradicate infection in over 50% of patients with cystic fibrosis. Antibiotic dosing strategies have been largely informed by common bacteria such as Pseudomonas aeruginosa. The "pharmacodynamic" effects of amikacin, a backbone of MABSC treatment, were thought to be related to maximum "peak" drug concentration, leading to daily or three times weekly dosing. However, we found that amikacin MABSC kill and growth recovery, an indicator of antibiotic resistance, are dependent on how long amikacin concentrations are above the minimum inhibitory concentration, not how high the peak concentration is. Therefore, we recommend a re-evaluation of amikacin dosing to determine if increased frequency can improve efficacy.
Collapse
Affiliation(s)
- Joy E. Gibson
- Division of Infectious Diseases and the Laboratory of Applied Pharmacokinetics and Bioinformatics, The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, California, USA
- Department of Pediatrics, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Nishant Nandanwar
- Division of Infectious Diseases and the Laboratory of Applied Pharmacokinetics and Bioinformatics, The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, California, USA
| | - Michael N. Neely
- Division of Infectious Diseases and the Laboratory of Applied Pharmacokinetics and Bioinformatics, The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, California, USA
- Department of Pediatrics, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| |
Collapse
|
24
|
Zikou X, Vaia D, Vasiliki P, Panagiotis C, Stavros A. Use of Therapeutic Apheresis methods in ICU. Transfus Apher Sci 2024; 63:103853. [PMID: 38049358 DOI: 10.1016/j.transci.2023.103853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2023]
Abstract
Apheresis is a modern medical approach in which plasma or cellular components are separated from the whole blood. Apheresis can be either diagnostic or therapeutic. Diagnostic apheresis is typically applied in hematology and cancer research. Therapeutic Apheresis (TA) includes a broad spectrum of extracorporeal treatments applied in various medical specialties, including Intensive Care Unit (ICU). Considering the complexity of the pathophysiologic characteristics of various clinical entities and in particular sepsis, apheresis methods are becoming increasingly applicable. Therapeutic Plasma Exchange (TPE) is the most common used method in ICU. It is considered as first line therapy for Thrombotic Thrombocytopenic Purpura (TTP) and Guillain Barre Syndrome, while the current data for sepsis are scarce. Over the last decades, technologic evolution has led to increasing application of new and more selective methods based on adsorptive techniques. In this review we will describe the current data of characteristics of different techniques, safety and clinical impact of apheresis methods used in ICUs.
Collapse
|
25
|
Schlegtendal A, Rettberg S, Maier C, Brinkmann F, Koerner-Rettberg C. Necessity of Tobramycin trough Levels in Once Daily Iv-Treatment in Patients with Cystic Fibrosis. KLINISCHE PADIATRIE 2024; 236:116-122. [PMID: 38286409 DOI: 10.1055/a-2244-6903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2024]
Abstract
BACKGROUND Once daily intravenous (iv) treatment with tobramycin for Pseudomonas aeruginosa infection in patients with cystic fibrosis (pwCF) is frequently monitored by measuring tobramycin trough levels (TLs). Although the necessity of these TLs is recently questioned in pwCF without renal impairment, no study has evaluated this so far. The aim of this observational study was to evaluate the frequency of increased tobramycin TLs in pwCF treated with a once daily tobramycin dosing protocol. METHODS Patient records of all consecutive once daily iv tobramycin courses in 35 pwCF between 07/2009 and 07/2019 were analyzed for tobramycin level, renal function, co-medication and comorbidity. RESULTS Eight elevated TLs (2.9% of 278 courses) were recorded in four patients, two with normal renal function. One of these resolved without adjustment of tobramycin dosages suggesting a test timing or laboratory error. In the other patient the elevated tobramycin level decreased after tobramycin dosage adjustment. Six of the elevated levels occurred in two patients with chronic renal failure. In 15 other patients with reduced glomerular filtration rate (GFR) (36 courses) but normal range creatinine no case of elevated tobramycin trough levels was detected. Neither cumulative tobramycin dosages nor concomitant diabetes or nutritional status were risk factors for elevated TLs. CONCLUSION Our data show that elevated tobramycin TLs are rare but cannot be excluded, so determination of tobramycin TLs is still recommended for safety.
Collapse
Affiliation(s)
- Anne Schlegtendal
- Department of pediatrics, Ruhr-Universität Bochum Medizinische Fakultät, Bochum, Germany
| | - Sophia Rettberg
- Department of pediatrics, Ruhr-Universität Bochum Medizinische Fakultät, Bochum, Germany
| | - Christoph Maier
- Department of pediatrics, Ruhr-Universität Bochum Medizinische Fakultät, Bochum, Germany
| | - Folke Brinkmann
- Department of pediatrics, Ruhr-Universität Bochum Medizinische Fakultät, Bochum, Germany
| | | |
Collapse
|
26
|
Karimzadeh I, Abdollahpour-Alitappeh M, Ghaffari S, Mahi-Birjand M, Barkhordari A, Alemzadeh E. Aminoglycosides: Single- or Multiple-daily Dosing? An Updated Qualitative Systematic Review of Randomized Trials on Toxicity and Efficacy. Curr Mol Med 2024; 24:1358-1373. [PMID: 37533241 DOI: 10.2174/1566524023666230801160452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 06/17/2023] [Accepted: 06/22/2023] [Indexed: 08/04/2023]
Abstract
INTRODUCTION Aminoglycosides are among the first-choice antibiotics for routine clinical use. However, dose-limiting factors such as ototoxicity and nephrotoxicity are considered as serious complications of aminoglycosides. OBJECTIVE In this systematic review, the main goal was to investigate the efficacy and incidence of nephrotoxicity and ototoxicity of once-daily dosing (ODD) and multiple daily dosing (MDD) regimens of aminoglycosides through available randomized controlled trials (RCTs). METHODS We performed a literature-based research in relevant databases, including EMBASE, MEDLINE, and SCOPUS published between 1987 and 2023 using the keywords "aminoglycosides", "pharmacokinetics", "ODD", "MDD", "once daily", "multiple daily", "dosing regimen", "nephrotoxicity", "ototoxicity", "efficacy", "safety", and "toxicity". As so told, the results of this article were limited to papers available in English. Our initial search yielded 1124 results. After a review of the titles and abstracts of the articles, 803 articles were excluded from this study because they did not address the toxicity and effectiveness of ODD versus MDD of aminoglycosides. A total number of 20 studies on gentamicin, tobramycin, netilmicin, and amikacin met the inclusion criteria for the efficacy of aminoglycosides and their role in ototoxicity and nephrotoxicity were included in this review. Studies recruited different age classes, and the age of relevant cohorts varied from only a few days to more than 70 years. RESULTS The most common clinical condition in the included studies was cystic fibrosis. CONCLUSION In most studies, there were no significant differences between the two regimens regarding ototoxicity. In addition, the ODD regimens were safer than MDD concerning nephrotoxicity.
Collapse
Affiliation(s)
- Iman Karimzadeh
- Department of Clinical Pharmacy, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Shokouh Ghaffari
- Infectious Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Motahareh Mahi-Birjand
- Department of Clinical Pharmacy, School of Pharmacy, Infectious Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Amin Barkhordari
- Cellular and Molecular Research Center, Gerash University of Medical Sciences, Gerash, Iran
| | - Effat Alemzadeh
- Infectious Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
27
|
Lang M, Carvalho A, Baharoglu Z, Mazel D. Aminoglycoside uptake, stress, and potentiation in Gram-negative bacteria: new therapies with old molecules. Microbiol Mol Biol Rev 2023; 87:e0003622. [PMID: 38047635 PMCID: PMC10732077 DOI: 10.1128/mmbr.00036-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2023] Open
Abstract
SUMMARYAminoglycosides (AGs) are long-known molecules successfully used against Gram-negative pathogens. While their use declined with the discovery of new antibiotics, they are now classified as critically important molecules because of their effectiveness against multidrug-resistant bacteria. While they can efficiently cross the Gram-negative envelope, the mechanism of AG entry is still incompletely understood, although this comprehension is essential for the development of new therapies in the face of the alarming increase in antibiotic resistance. Increasing antibiotic uptake in bacteria is one strategy to enhance effective treatments. This review aims, first, to consolidate old and recent knowledge about AG uptake; second, to explore the connection between AG-dependent bacterial stress and drug uptake; and finally, to present new strategies of potentiation of AG uptake for more efficient antibiotic therapies. In particular, we emphasize on the connection between sugar transport and AG potentiation.
Collapse
Affiliation(s)
- Manon Lang
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, Paris, France
| | - André Carvalho
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, Paris, France
| | - Zeynep Baharoglu
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, Paris, France
| | - Didier Mazel
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Unité Plasticité du Génome Bactérien, Paris, France
| |
Collapse
|
28
|
Gras E, Vu TTT, Nguyen NTQ, Tran VG, Mao Y, Tran ND, Mai NH, Dong OX, Jung DH, Iorio NLPP, Povoa HCC, Pinheiro MG, Aguiar-Alves F, Weiss WJ, Zheng B, Cheng LI, Stover CK, Sellman BR, DiGiandomenico A, Gibault L, Valour F, Diep BA. Development and validation of a rabbit model of Pseudomonas aeruginosa non-ventilated pneumonia for preclinical drug development. Front Cell Infect Microbiol 2023; 13:1297281. [PMID: 38149013 PMCID: PMC10750358 DOI: 10.3389/fcimb.2023.1297281] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/22/2023] [Indexed: 12/28/2023] Open
Abstract
Background New drugs targeting antimicrobial resistant pathogens, including Pseudomonas aeruginosa, have been challenging to evaluate in clinical trials, particularly for the non-ventilated hospital-acquired pneumonia and ventilator-associated pneumonia indications. Development of new antibacterial drugs is facilitated by preclinical animal models that could predict clinical efficacy in patients with these infections. Methods We report here an FDA-funded study to develop a rabbit model of non-ventilated pneumonia with Pseudomonas aeruginosa by determining the extent to which the natural history of animal disease reproduced human pathophysiology and conducting validation studies to evaluate whether humanized dosing regimens of two antibiotics, meropenem and tobramycin, can halt or reverse disease progression. Results In a rabbit model of non-ventilated pneumonia, endobronchial challenge with live P. aeruginosa strain 6206, but not with UV-killed Pa6206, caused acute respiratory distress syndrome, as evidenced by acute lung inflammation, pulmonary edema, hemorrhage, severe hypoxemia, hyperlactatemia, neutropenia, thrombocytopenia, and hypoglycemia, which preceded respiratory failure and death. Pa6206 increased >100-fold in the lungs and then disseminated from there to infect distal organs, including spleen and kidneys. At 5 h post-infection, 67% of Pa6206-challenged rabbits had PaO2 <60 mmHg, corresponding to a clinical cut-off when oxygen therapy would be required. When administered at 5 h post-infection, humanized dosing regimens of tobramycin and meropenem reduced mortality to 17-33%, compared to 100% for saline-treated rabbits (P<0.001 by log-rank tests). For meropenem which exhibits time-dependent bactericidal activity, rabbits treated with a humanized meropenem dosing regimen of 80 mg/kg q2h for 24 h achieved 100% T>MIC, resulting in 75% microbiological clearance rate of Pa6206 from the lungs. For tobramycin which exhibits concentration-dependent killing, rabbits treated with a humanized tobramycin dosing regimen of 8 mg/kg q8h for 24 h achieved Cmax/MIC of 9.8 ± 1.4 at 60 min post-dose, resulting in 50% lung microbiological clearance rate. In contrast, rabbits treated with a single tobramycin dose of 2.5 mg/kg had Cmax/MIC of 7.8 ± 0.8 and 8% (1/12) microbiological clearance rate, indicating that this rabbit model can detect dose-response effects. Conclusion The rabbit model may be used to help predict clinical efficacy of new antibacterial drugs for the treatment of non-ventilated P. aeruginosa pneumonia.
Collapse
Affiliation(s)
- Emmanuelle Gras
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA, United States
- Université François Rabelais, Tours, France
| | - Trang T. T. Vu
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Nhu T. Q. Nguyen
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Vuvi G. Tran
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Yanjie Mao
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA, United States
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Nguyen D. Tran
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA, United States
- University of Medicine and Pharmacy, Ho Chi Minh City, Vietnam
| | - Nam H. Mai
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Oliver X. Dong
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA, United States
| | - David H. Jung
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Natalia L. P. P. Iorio
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA, United States
- Department of Basic Science, Fluminense Federal University, Nova Friburgo, Rio de Janeiro, Brazil
| | - Helvecio C. C. Povoa
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA, United States
- Department of Basic Science, Fluminense Federal University, Nova Friburgo, Rio de Janeiro, Brazil
| | - Marcos Gabriel Pinheiro
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Fabio Aguiar-Alves
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA, United States
- Pathology Program, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - William J. Weiss
- Pre-Clinical Services at UNT Health Science Center, Fort Worth, TX, United States
| | - Bo Zheng
- Clinical Pharmacology & DMPK, AstraZeneca, Gaithersburg, MD, United States
| | - Lily I. Cheng
- Early Vaccines and Immune Therapies, AstraZeneca, Gaithersburg, MD, United States
| | - Charles K. Stover
- Early Vaccines and Immune Therapies, AstraZeneca, Gaithersburg, MD, United States
| | - Bret R. Sellman
- Early Vaccines and Immune Therapies, AstraZeneca, Gaithersburg, MD, United States
| | | | - Laure Gibault
- Pathology Department, George Pompidou European Hospital, Paris, France
| | - Florent Valour
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA, United States
- Department of Infectious Diseases, Hospices Civils de Lyon, Lyon, France
- CIRI – Centre International de Recherche en Infectiologie, Inserm, U1111, University of Lyon, Lyon, France
- Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, University of Lyon, Lyon, France
| | - Binh An Diep
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
29
|
Jaiswal S, Agarwal A, Singh S, Mohan P. Therapeutic drug monitoring of Amikacin in hospitalized patients: A pilot study. Med J Armed Forces India 2023; 79:S119-S124. [PMID: 38144659 PMCID: PMC10746789 DOI: 10.1016/j.mjafi.2022.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 02/20/2022] [Indexed: 10/18/2022] Open
Abstract
Background Amikacin, an aminoglycoside, is a widely used parenteral antibiotic. Therapeutic drug monitoring (TDM) is recommended for aminoglycosides to avoid toxicity. However, the lack of infrastructure at most places precludes it. This pilot and novel study attempt to estimate the real-world serum levels of Amikacin in hospitalised patients. Methods Thirty admitted patients, given Amikacin injections, were included in the study. In addition, 15 clinical specimens isolated with gram-negative bacteria were tested for minimum inhibitory concentration (MIC) value of Amikacin. Trough and peak serum levels of Amikacin were estimated by high-pressure liquid chromatography (HPLC). Results The average MIC value of Amikacin estimated in our laboratory was 3.92 mcg/mL. Peak and trough serum levels of Amikacin ranged from 12.1 to 66.4 mcg/ml and 1.1 to 20.7 mcg/ml, respectively. More than 83% of our patients achieved peak Amikacin levels of 15 mcg/mL, and 37% had trough levels above 5 mcg/mL. These levels are desirable watersheds as per available literature. Conclusion Trough levels of Amikacin in all cases and a review of dosing according to MIC values are recommended to achieve drug safety and therapeutic efficacy.
Collapse
Affiliation(s)
- Sanjay Jaiswal
- Professor, Department of Pharmacology, Armed Forces Medical College, Pune, India
| | - Amit Agarwal
- Senior Advisor (Surgery) & Urologist, Command Hospital (Western Command), Chandimandir Cantt, Panchkula, India
| | - S.P. Singh
- Professor & Head, Department of Microbiology, Armed Forces Medical College, Pune, India
| | - Prafull Mohan
- Professor, Department of Pharmacology, Armed Forces Medical College, Pune, India
| |
Collapse
|
30
|
de Hoop-Sommen MA, van der Heijden JEM, Freriksen JJM, Greupink R, de Wildt SN. Pragmatic physiologically-based pharmacokinetic modeling to support clinical implementation of optimized gentamicin dosing in term neonates and infants: proof-of-concept. Front Pediatr 2023; 11:1288376. [PMID: 38078320 PMCID: PMC10702772 DOI: 10.3389/fped.2023.1288376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/02/2023] [Indexed: 02/12/2024] Open
Abstract
Introduction Modeling and simulation can support dosing recommendations for clinical practice, but a simple framework is missing. In this proof-of-concept study, we aimed to develop neonatal and infant gentamicin dosing guidelines, supported by a pragmatic physiologically-based pharmacokinetic (PBPK) modeling approach and a decision framework for implementation. Methods An already existing PBPK model was verified with data of 87 adults, 485 children and 912 neonates, based on visual predictive checks and predicted-to-observed pharmacokinetic (PK) parameter ratios. After acceptance of the model, dosages now recommended by the Dutch Pediatric Formulary (DPF) were simulated, along with several alternative dosing scenarios, aiming for recommended peak (i.e., 8-12 mg/L for neonates and 15-20 mg/L for infants) and trough (i.e., <1 mg/L) levels. We then used a decision framework to weigh benefits and risks for implementation. Results The PBPK model adequately described gentamicin PK. Simulations of current DPF dosages showed that the dosing interval for term neonates up to 6 weeks of age should be extended to 36-48 h to reach trough levels <1 mg/L. For infants, a 7.5 mg/kg/24 h dose will reach adequate peak levels. The benefits of these dose adaptations outweigh remaining uncertainties which can be minimized by routine drug monitoring. Conclusion We used a PBPK model to show that current DPF dosages for gentamicin in term neonates and infants needed to be optimized. In the context of potential uncertainties, the risk-benefit analysis proved positive; the model-informed dose is ready for clinical implementation.
Collapse
Affiliation(s)
- Marika A. de Hoop-Sommen
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, Netherlands
| | - Joyce E. M. van der Heijden
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jolien J. M. Freriksen
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, Netherlands
| | - Rick Greupink
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, Netherlands
| | - Saskia N. de Wildt
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, Nijmegen, Netherlands
- Department for Intensive Care, Radboud University Medical Center, Nijmegen, Netherlands
- Intensive Care and Pediatric Surgery, Erasmus MC, Rotterdam, Netherlands
| |
Collapse
|
31
|
Kilcoyne I, Nieto J, Magdesian KG, Nottle BF. Effects of a 10% dimethyl sulfoxide solution on radiocarpal joint amikacin pharmacokinetics during intravenous regional limb perfusion in standing sedated horses. Vet Surg 2023; 52:1171-1179. [PMID: 37280731 DOI: 10.1111/vsu.13973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 04/28/2023] [Accepted: 05/08/2023] [Indexed: 06/08/2023]
Abstract
OBJECTIVE To determine the effect of a 10% dimethyl sulfoxide (DMSO) solution on the peak concentration (CMAX ) of amikacin in the radiocarpal joint (RCJ) during intravenous regional limb perfusion (IVRLP) compared with 0.9% NaCl. STUDY DESIGN Randomized crossover study. ANIMALS Seven healthy adult horses. METHODS The horses underwent IVRLP with 2 g of amikacin sulfate diluted to 60 mL using a 10% DMSO or 0.9% NaCl solution. Synovial fluid was collected from the RCJ at 5, 10, 15, 20, 25, and 30 minutes after IVRLP. The wide rubber tourniquet placed on the antebrachium was removed after the 30 min sample. Amikacin concentrations were quantified by a fluorescence polarization immunoassay. The mean CMAX and time to peak concentration (TMAX ) of amikacin within the RCJ were determined. A one-sided paired t-test was used to determine the differences between treatments. The significance level was p < .05. RESULTS The mean ± SD CMAX in the DMSO group was 1361.8 ± 593 μg/mL and in the 0.9% NaCl group it was 860 ± 481.6 μg/mL (p = .058). Mean TMAX using the 10% DMSO solution was 23 and 18 min using the 0.9% NaCl perfusate (p = .161). No adverse effects were associated with use of the 10% DMSO solution. CONCLUSION Although there were higher mean peak synovial concentrations using the 10% DMSO solution no difference in synovial amikacin CMAX between perfusate type was detected (p = .058). CLINICAL SIGNIFICANCE Use of a 10% DMSO solution in conjunction with amikacin during IVRLP is a feasible technique and does not negatively affect the synovial amikacin levels achieved. Further research is warranted to determine other effects of using DMSO during IVRLP.
Collapse
Affiliation(s)
- Isabelle Kilcoyne
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Los Angeles, California, USA
| | - Jorge Nieto
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Los Angeles, California, USA
| | - K Gary Magdesian
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Los Angeles, California, USA
| | - Bridget F Nottle
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
32
|
Tilanus A, Drusano G. Inoculum-Based Dosing: A Novel Concept for Combining Time with Concentration-Dependent Antibiotics to Optimize Clinical and Microbiological Outcomes in Severe Gram Negative Sepsis. Antibiotics (Basel) 2023; 12:1581. [PMID: 37998783 PMCID: PMC10668771 DOI: 10.3390/antibiotics12111581] [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: 09/19/2023] [Revised: 10/22/2023] [Accepted: 10/23/2023] [Indexed: 11/25/2023] Open
Abstract
Certain classes of antibiotics show "concentration dependent" antimicrobial activity; higher concentrations result in increased bacterial killing rates, in contrast to "time dependent antibiotics", which show antimicrobial activity that depends on the time that antibiotic concentrations remain above the MIC. Aminoglycosides and fluoroquinolones are still widely used concentration-dependent antibiotics. These antibiotics are not hydrolyzed by beta-lactamases and are less sensitive to the inoculum effect, which can be defined as an increased MIC for the antibiotic in the presence of a relatively higher bacterial load (inoculum). In addition, they possess a relatively long Post-Antibiotic Effect (PAE), which can be defined as the absence of bacterial growth when antibiotic concentrations fall below the MIC. These characteristics make them interesting complementary antibiotics in the management of Multi-Drug Resistant (MDR) bacteria and/or (neutropenic) patients with severe sepsis. Global surveillance studies have shown that up to 90% of MDR Gram-negative bacteria still remain susceptible to aminoglycosides, depending on the susceptibility breakpoint (e.g., CLSI or EUCAST) being applied. This percentage is notably lower for fluoroquinolones but depends on the region, type of organism, and mechanism of resistance involved. Daily (high-dose) dosing of aminoglycosides for less than one week has been associated with significantly less nephro/oto toxicity and improved target attainment. Furthermore, higher-than-conventional dosing of fluoroquinolones has been linked to improved clinical outcomes. Beta-lactam antibiotics are the recommended backbone of therapy for severe sepsis. Since these antibiotics are time-dependent, the addition of a second concentration-dependent antibiotic could serve to quickly lower the bacterial inoculum, create PAE, and reduce Penicillin-Binding Protein (PBP) expression. Inadequate antibiotic levels at the site of infection, especially in the presence of high inoculum infections, have been shown to be important risk factors for inadequate resistance suppression and therapeutic failure. Therefore, in the early phase of severe sepsis, effort should be made to optimize the dose and quickly lower the inoculum. In this article, the authors propose a novel concept of "Inoculum Based Dosing" in which the decision for antibiotic dosing regimens and/or combination therapy is not only based on the PK parameters of the patient, but also on the presumed inoculum size. Once the inoculum has been lowered, indirectly reflected by clinical improvement, treatment simplification should be considered to further treat the infection.
Collapse
Affiliation(s)
- Alwin Tilanus
- Department of Infectious Diseases, Clinica Los Nogales, Calle 95 # 23-61, Bogota 110221, Colombia
| | - George Drusano
- Institute for Therapeutic Innovation, University of Florida, 6550 Sanger Road, Orlando, FL 32827, USA;
| |
Collapse
|
33
|
Grit GF, Toren-Wielema ML, Colin PJ, Touw DJ. Gentamicin Administration in Dialysis Patients: Before or After Hemodialysis? Ther Drug Monit 2023; 45:697-701. [PMID: 36730889 DOI: 10.1097/ftd.0000000000001058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 09/21/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND Gentamicin is used to treat severe infections and has a small therapeutic window. This study aimed to optimize the dosing strategy of gentamicin in intermittently hemodialyzed patients by simulating concentration-time profiles during pre- and postdialysis dosing, based on a published pharmacokinetic model. METHODS Pharmacokinetic simulations were performed with virtual patients, including septic patients, who were treated with gentamicin and received weekly hemodialysis with an interval of 48 h-48 h-72 h. The following dosing regimens were simulated: for nonseptic patients, 5 mg/kg gentamicin was given 1 h or 2 h before dialysis or a starting dose of 2.5 mg/kg and a maintenance dose of 1.5 mg/kg immediately after dialysis were given; for septic patients, 6 mg/kg gentamicin was given 1 h or 2 h before dialysis or a starting dose of 3 mg/kg and a maintenance dose of 1.8 mg/kg immediately were given after dialysis. The mean maximum concentration (C max ), area under the curve (AUC) 24 h , and target attainment (TA) of pharmacodynamic targets were calculated and compared. The following targets were adopted from the literature: C max >8 mg/L and <20 mg/L and AUC 24 h >70 mg·h/L and <120 mg·h/L. RESULTS In nonseptic patients, postdialysis dosing resulted in a TA of 35% for C max of >8 mg/L, 100% for <20 mg/L and AUC 24 h >70 mg·h/L, and 45% for <120 mg·h/L. Dosing 2 h before dialysis resulted in a TA of 100% for C max of >8 mg/L, 40% for <20 mg/L, 65% for AUC 24 h >70 mg·h/L, and 77% for <120 mg·h/L. Simulations of septic patients resulted in comparable outcomes with higher TAs for C max <20 mg/L (96%), AUC 24 h >70 mg·h/L (90%), and AUC 24 h <120 mg·h/L (53%) for dosing 1 h before dialysis. CONCLUSIONS Postdialysis dosing resulted in a low TA of C max >8 mg/L; however, predialysis dosing ensured a high TA of C max >8 mg/L and acceptable TA of C max <20 mg/L, AUC 24 h >70 mg·h/L, and AUC 24 h <120 mg·h/L, which could increase the efficacy of gentamicin. Therefore, clinicians should consider predialysis dosing of gentamicin in patients undergoing intermittent hemodialysis.
Collapse
Affiliation(s)
- Geeske F Grit
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; and
| | - Martha L Toren-Wielema
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; and
| | - Pieter J Colin
- Department of Anesthesiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Daan J Touw
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; and
| |
Collapse
|
34
|
Biénès T, Lyssens A, Machiels H, Hercot ME, Fastres A, Alexandru-Cosmin T, Deville M, Charlier C, Billen F, Clercx C. Intranasal and Serum Gentamicin Concentration: Comparison of Three Topical Administration Protocols in Dogs. Vet Sci 2023; 10:490. [PMID: 37624277 PMCID: PMC10457901 DOI: 10.3390/vetsci10080490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/14/2023] [Accepted: 07/21/2023] [Indexed: 08/26/2023] Open
Abstract
Antimicrobials' topical administration efficacy has not been assessed in dogs with upper respiratory tract disease. The aim was to compare the concentration of gentamicin in nasal lavage fluid (NALF) and in serum after three topical protocols. This was a prospective crossover study of ten healthy dogs. Gentamicin was nebulized for a duration of 1 week, twice a day, for 10 min in the first protocol (10-min protocol) and for 3 min in the second protocol (3-min protocol), while the third protocol consisted of the administration of 0.25 mL of gentamicin in each nostril (drop protocol). Median concentrations of gentamicin in NALF were 9.39 µg/mL (8.12-19.97 interquartile range), 4.96 µg/mL (4.60-6.43) and 137.00 µg/mL (110.5-162.00) in the 10-min protocol, 3-min protocol and drop protocol, respectively. The result for the drop protocol was significantly higher than those of both nebulization protocols in NALF (p = 0.039). In serum, the gentamicin concentration was 0.98 µg/mL (0.65-1.53) and 0.25 µg/mL (0.25-0.44) in the 10-min and 3-min protocols, respectively. Gentamicin was not detected in the serum of seven out of ten dogs in the drop protocol, and gentamicin was significantly higher in the 10-min protocol compared to the drop protocol (p = 0.001). This study found that the 10-min, 3-min and drop protocols achieved superior concentrations in NALF compared to the minimum inhibitory concentration for gentamicin-sensitive bacteria, while remaining below the toxic values in blood.
Collapse
Affiliation(s)
- Tom Biénès
- Department of Clinical Sciences, Faculty of Veterinary Medicine, B67 Sart Tilman, University of Liege, 4000 Liege, Belgium; (A.L.); (H.M.); (M.E.H.); (T.A.-C.); (F.B.)
| | - Aurélie Lyssens
- Department of Clinical Sciences, Faculty of Veterinary Medicine, B67 Sart Tilman, University of Liege, 4000 Liege, Belgium; (A.L.); (H.M.); (M.E.H.); (T.A.-C.); (F.B.)
| | - Hélène Machiels
- Department of Clinical Sciences, Faculty of Veterinary Medicine, B67 Sart Tilman, University of Liege, 4000 Liege, Belgium; (A.L.); (H.M.); (M.E.H.); (T.A.-C.); (F.B.)
| | - Marie Eve Hercot
- Department of Clinical Sciences, Faculty of Veterinary Medicine, B67 Sart Tilman, University of Liege, 4000 Liege, Belgium; (A.L.); (H.M.); (M.E.H.); (T.A.-C.); (F.B.)
| | - Aline Fastres
- Department of Clinical Sciences, Faculty of Veterinary Medicine, B67 Sart Tilman, University of Liege, 4000 Liege, Belgium; (A.L.); (H.M.); (M.E.H.); (T.A.-C.); (F.B.)
| | - Tutunaru Alexandru-Cosmin
- Department of Clinical Sciences, Faculty of Veterinary Medicine, B67 Sart Tilman, University of Liege, 4000 Liege, Belgium; (A.L.); (H.M.); (M.E.H.); (T.A.-C.); (F.B.)
| | - Marine Deville
- Laboratory of Clinical, Forensic, Industrial and Environmental Toxicology, Center for Interdisciplinary Research on Medicines (CIRM), University Hospital of Liege, 4130 Liege, Belgium; (M.D.); (C.C.)
| | - Corinne Charlier
- Laboratory of Clinical, Forensic, Industrial and Environmental Toxicology, Center for Interdisciplinary Research on Medicines (CIRM), University Hospital of Liege, 4130 Liege, Belgium; (M.D.); (C.C.)
| | - Frédéric Billen
- Department of Clinical Sciences, Faculty of Veterinary Medicine, B67 Sart Tilman, University of Liege, 4000 Liege, Belgium; (A.L.); (H.M.); (M.E.H.); (T.A.-C.); (F.B.)
| | - Cécile Clercx
- Department of Clinical Sciences, Faculty of Veterinary Medicine, B67 Sart Tilman, University of Liege, 4000 Liege, Belgium; (A.L.); (H.M.); (M.E.H.); (T.A.-C.); (F.B.)
| |
Collapse
|
35
|
Frost KJ, Hamilton RA, Hughes S, Jamieson C, Rafferty P, Troise O, Jenkins A. Systematic review of high-dose amikacin regimens for the treatment of Gram-negative infections based on EUCAST dosing recommendations. Eur J Hosp Pharm 2023; 30:189-195. [PMID: 36344247 PMCID: PMC10359793 DOI: 10.1136/ejhpharm-2022-003421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 10/17/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Updated European Committee on Antimicrobial Susceptibility Testing (EUCAST) amikacin breakpoints for Enterobacterales and Pseudomonas aeruginosa included revised dosing recommendations of 25-30 mg/kg to achieve key pharmacokinetic/pharmacodynamic parameters, higher than recommended in the British National Formulary. The objectives of this review were to identify clinical evidence for high-dose amikacin regimens and to determine drug exposures that are related to adverse events and toxicity. METHODS The literature search was conducted in October 2021 and updated in May 2022 using electronic databases for any study reporting adult participants treated with amikacin at doses ≥20 mg/kg/day. Reference lists of included papers were also screened for potential papers. Data were extracted for pharmacokinetic parameters and clinical outcomes, presented in a summary table and consolidated narratively. Meta-analysis was not possible. Each study was assessed for bias before, during and after the intervention using the ROBINS-I tool. RESULTS Nine studies (total 501 participants in 10 reports) were identified and included, eight of which were observational studies. Assessment of bias showed substantial flaws. Dosing regimens ranged from 25 to 30 mg/kg/day. Six studies adjusted the dose in obesity when participants had a body mass index of ≥30 kg/m2. Target peak serum concentrations ranged from 60 mg/L to 80 mg/L and 59.6-81.8% of patients achieved these targets, but there was no information on clinical outcomes. Two studies reported the impact of high-dose amikacin on renal function. No studies reporting auditory or vestibular toxicity were identified. CONCLUSION All included papers were limited by a significant risk of bias, while methodological and reporting heterogeneity made drawing conclusions challenging. Lack of information on the impact on renal function or ototoxicity means high-dose regimens should be used cautiously in older people. There is a need for a consensus guideline for high-dose amikacin to be written. TRIAL REGISTRATION NUMBER PROSPERO (CRD42021250022).
Collapse
Affiliation(s)
| | - Ryan A Hamilton
- Pharmacy, De Montfort University, Leicester, UK
- Pharmacy, Kettering General Hospital NHS Foundation Trust, Kettering, UK
| | - Stephen Hughes
- Pharmacy, Chelsea and Westminster Hospital NHS Foundation Trust, London, UK
| | - Conor Jamieson
- NHS England and NHS Improvement Midlands, Birmingham, UK
| | - Paul Rafferty
- Pharmacy, Southern Health and Social Care Trust, Portadown, UK
| | - Oliver Troise
- Pharmacy, Chelsea and Westminster Hospital NHS Foundation Trust, London, UK
| | - Abi Jenkins
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| |
Collapse
|
36
|
Thy M, Timsit JF, de Montmollin E. Aminoglycosides for the Treatment of Severe Infection Due to Resistant Gram-Negative Pathogens. Antibiotics (Basel) 2023; 12:antibiotics12050860. [PMID: 37237763 DOI: 10.3390/antibiotics12050860] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/01/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023] Open
Abstract
Aminoglycosides are a family of rapidly bactericidal antibiotics that often remain active against resistant Gram-negative bacterial infections. Over the past decade, their use in critically ill patients has been refined; however, due to their renal and cochleovestibular toxicity, their indications in the treatment of sepsis and septic shock have been gradually reduced. This article reviews the spectrum of activity, mode of action, and methods for optimizing the efficacy of aminoglycosides. We discuss the current indications for aminoglycosides, with an emphasis on multidrug-resistant Gram-negative bacteria, such as extended-spectrum β-lactamase-producing Enterobacterales, carbapenemase-producing Enterobacterales, multidrug-resistant Pseudomonas aeruginosa, and carbapenem-resistant Acinetobacter baumannii. Additionally, we review the evidence for the use of nebulized aminoglycosides.
Collapse
Affiliation(s)
- Michaël Thy
- Assistance Publique Hôpitaux de Paris (AP-HP), Service de Médecine Intensive et Réanimation Infectieuse, Hôpital Bichat Claude-Bernard, Université Paris Cité, 46 Rue Henri Huchard, 75018 Paris, France
- Equipe d'accueil (EA) 7323, Department of Pharmacology and Therapeutic Evaluation in Children and Pregnant Women, Université Paris Cité, 75018 Paris, France
| | - Jean-François Timsit
- Assistance Publique Hôpitaux de Paris (AP-HP), Service de Médecine Intensive et Réanimation Infectieuse, Hôpital Bichat Claude-Bernard, Université Paris Cité, 46 Rue Henri Huchard, 75018 Paris, France
- Unité mixte de Recherche (UMR) 1137, Infection, Antimicrobials, Modelization, Epidemiology (IAME), Institut National de la Recherche Médicale (INSERM), Université Paris Cité, 75018 Paris, France
| | - Etienne de Montmollin
- Assistance Publique Hôpitaux de Paris (AP-HP), Service de Médecine Intensive et Réanimation Infectieuse, Hôpital Bichat Claude-Bernard, Université Paris Cité, 46 Rue Henri Huchard, 75018 Paris, France
- Unité mixte de Recherche (UMR) 1137, Infection, Antimicrobials, Modelization, Epidemiology (IAME), Institut National de la Recherche Médicale (INSERM), Université Paris Cité, 75018 Paris, France
| |
Collapse
|
37
|
Khaled JM, Alharbi NS, Kadaikunnan S, Alobaidi AS, Nauman K, M.Ghilan AK, Almazyed AO, Aldosary MS, Al Rashedi S, Alfalih AM, Farrag MA, Alharbi MS. Distribution of Candida infections in patients and evaluation of the synergic interactions of some drugs against emerging fluconazole- and caspofungin-resistant C. auris. JOURNAL OF KING SAUD UNIVERSITY - SCIENCE 2023; 35:102617. [DOI: 10.1016/j.jksus.2023.102617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
38
|
Hollander EM, van Tuinen EL, Schölvinck EH, Bergman KA, Bourgonje AR, Gracchi V, Kneyber MCJ, Touw DJ, Mian P. Evaluation of Dosing Guidelines for Gentamicin in Neonates and Children. Antibiotics (Basel) 2023; 12:antibiotics12050810. [PMID: 37237713 DOI: 10.3390/antibiotics12050810] [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: 03/24/2023] [Revised: 04/17/2023] [Accepted: 04/23/2023] [Indexed: 05/28/2023] Open
Abstract
Although aminoglycosides are frequently prescribed to neonates and children, the ability to reach effective and safe target concentrations with the currently used dosing regimens remains unclear. This study aims to evaluate the target attainment of the currently used dosing regimens for gentamicin in neonates and children. We conducted a retrospective single-center cohort study in neonates and children receiving gentamicin between January 2019 and July 2022, in the Beatrix Children's Hospital. The first gentamicin concentration used for therapeutic drug monitoring was collected for each patient, in conjunction with information on dosing and clinical status. Target trough concentrations were ≤1 mg/L for neonates and ≤0.5 mg/L for children. Target peak concentrations were 8-12 mg/L for neonates and 15-20 mg/L for children. In total, 658 patients were included (335 neonates and 323 children). Trough concentrations were outside the target range in 46.2% and 9.9% of neonates and children, respectively. Peak concentrations were outside the target range in 46.0% and 68.7% of neonates and children, respectively. In children, higher creatinine concentrations were associated with higher gentamicin trough concentrations. This study corroborates earlier observational studies showing that, with a standard dose, drug concentration targets were met in only approximately 50% of the cases. Our findings show that additional parameters are needed to improve target attainment.
Collapse
Affiliation(s)
- Esther M Hollander
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Eline L van Tuinen
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Elisabeth H Schölvinck
- Department of Pediatric Infectious Diseases, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Klasien A Bergman
- Division of Neonatology, Department of Pediatrics, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Arno R Bourgonje
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Valentina Gracchi
- Division of Pediatric Nephrology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Martin C J Kneyber
- Division of Peadiatric Critical Care Medicine, Department of Paediatrics, Beatrix Children's Hospital Groningen, University Medical Center Groningen, University of Groningen Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Daan J Touw
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
- Department of Pharmaceutical Analysis, Groningen Research Institute for Pharmacy, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Paola Mian
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
39
|
El Hassani M, Thirion DJG, Koloskoff K, Matouk E, Simard C, Cloutier I, Pilote S, Marsot A. Development and Evaluation of a Height-Based Tobramycin Initial Dosing Nomogram for the Treatment of Adult Cystic Fibrosis Pulmonary Exacerbations. Ther Drug Monit 2023; 45:259-264. [PMID: 36730982 DOI: 10.1097/ftd.0000000000001053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 10/03/2022] [Indexed: 02/04/2023]
Abstract
ABSTRACT Tobramycin is widely used to treat pulmonary exacerbations of cystic fibrosis. Height has been previously found to be significantly more predictive of tobramycin pharmacokinetics than body weight. This study aimed to develop a height-based initial dosing nomogram and evaluate its performance in peak concentration (Cmax) precision relative to standard and fixed dosing. Monte Carlo simulations were performed to develop a nomogram representing the doses required to reach Cmax targets at different heights. Cmax data observed at 2 clinical centers [McGill University Health Centre (MUHC) and Institut universitaire de cardiologie et pneumologie de Québec (IUCPQ-UL)] were compared with population-predicted Cmax using the doses derived from the nomogram alongside a fixed dose. Height-based dosing resulted in significantly less variable-predicted Cmax values [coefficient of variation (CV) MUHC = 15.7% and IUCPQ-UL = 10.8%] than the Cmax values observed in clinical practice (CV MUHC = 30.0% and CV IUCPQ-UL = 26.9%) and predicted Cmax values obtained from a fixed dose (CV MUHC = 21.2% and CV IUCPQ-UL = 16.3%). An initial dosing nomogram was developed to help reduce pharmacokinetic variability in the observed Cmax. More precise dosing would allow for better clinical outcomes in adult patients with cystic fibrosis.
Collapse
Affiliation(s)
- Mehdi El Hassani
- Faculté de pharmacie, Université de Montréal, Montreal, Canada
- Laboratoire de suivi thérapeutique pharmacologique et pharmacocinétique, Faculté de pharmacie, Université de Montréal, Montreal, Canada
| | - Daniel J G Thirion
- Faculté de pharmacie, Université de Montréal, Montreal, Canada
- Department of Pharmacy, McGill University Health Centre, Montreal, Canada
| | - Kevin Koloskoff
- Faculté de pharmacie, Université de Montréal, Montreal, Canada
- Laboratoire de suivi thérapeutique pharmacologique et pharmacocinétique, Faculté de pharmacie, Université de Montréal, Montreal, Canada
| | - Elias Matouk
- Adult Cystic Fibrosis Clinic, Montreal Chest Institute, McGill University, Montreal, Canada
- Department of Medicine, McGill University, Montreal, Canada
- McGill University Health Center Research Institute, Montreal, Canada
| | - Chantale Simard
- Faculté de pharmacie, Université Laval, Quebec, Canada
- Centre de recherche, Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec, Canada; and
| | - Isabelle Cloutier
- Faculté de pharmacie, Université Laval, Quebec, Canada
- Département de pharmacie, Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec, Canada
| | - Sylvie Pilote
- Faculté de pharmacie, Université Laval, Quebec, Canada
| | - Amélie Marsot
- Faculté de pharmacie, Université de Montréal, Montreal, Canada
- Laboratoire de suivi thérapeutique pharmacologique et pharmacocinétique, Faculté de pharmacie, Université de Montréal, Montreal, Canada
| |
Collapse
|
40
|
Telles JP, Morales R, Yamada CH, Marins TA, D'Amaro Juodinis V, Sztajnbok J, Silva M, Bassetti BR, Albiero J, Tuon FF. Optimization of Antimicrobial Stewardship Programs Using Therapeutic Drug Monitoring and Pharmacokinetics-Pharmacodynamics Protocols: A Cost-Benefit Review. Ther Drug Monit 2023; 45:200-208. [PMID: 36622029 DOI: 10.1097/ftd.0000000000001067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/08/2022] [Indexed: 01/10/2023]
Abstract
PURPOSE Antimicrobial stewardship programs are important for reducing antimicrobial resistance because they can readjust antibiotic prescriptions to local guidelines, switch intravenous to oral administration, and reduce hospitalization times. Pharmacokinetics-pharmacodynamics (PK-PD) empirically based prescriptions and therapeutic drug monitoring (TDM) programs are essential for antimicrobial stewardship, but there is a need to fit protocols according to cost benefits. The cost benefits can be demonstrated by reducing toxicity and hospital stay, decreasing the amount of drug used per day, and preventing relapses in infection. Our aim was to review the data available on whether PK-PD empirically based prescriptions and TDM could improve the cost benefits of an antimicrobial stewardship program to decrease global hospital expenditures. METHODS A narrative review based on PubMed search with the relevant studies of vancomycin, aminoglycosides, beta-lactams, and voriconazole. RESULTS TDM protocols demonstrated important cost benefit for patients treated with vancomycin, aminoglycosides, and voriconazole mainly due to reduce toxicities and decreasing the hospital length of stay. In addition, PK-PD strategies that used infusion modifications to meropenem, piperacillin-tazobactam, ceftazidime, and cefepime, such as extended or continuous infusion, demonstrated important cost benefits, mainly due to reducing daily drug needs and lengths of hospital stays. CONCLUSIONS TDM protocols and PK-PD empirically based prescriptions improve the cost-benefits and decrease the global hospital expenditures.
Collapse
Affiliation(s)
- João Paulo Telles
- - AC Camargo Cancer Center, Infectious Diseases Department, São Paulo
- - Laboratory of Emerging Infectious Diseases, Pontifical Catholic University of Paraná, Curitiba
| | - Ronaldo Morales
- - Clinical Pharmacokinetics Center, School of Pharmaceutical Sciences, University of São Paulo
- - Pediatric Intensive Care Unit, Department of Pediatrics, Hospital Sírio-Libanês. São Paulo
| | - Carolina Hikari Yamada
- - Laboratory of Emerging Infectious Diseases, Pontifical Catholic University of Paraná, Curitiba
- - Hospital Universitário Evangélico Mackenzie, Department of Infectious Diseases, Curitiba
| | - Tatiana A Marins
- - Hospital Israelita Albert Einstein, Department of Clinical Pharmacy, São Paulo
| | | | - Jaques Sztajnbok
- - Instituto de Infectologia Emílio Ribas, São Paulo
- - Instituto da Criança do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (ICr/HC-FMUSP)
| | - Moacyr Silva
- - Hospital Israelita Albert Einstein, Department of Infection Prevention and Control, São Paulo
| | - Bil Randerson Bassetti
- - Hospital Santa Rita de Cássia, Department of Infectious Disease and Infection Control, Vitória ; and
| | - James Albiero
- - Universidade Estadual de Maringá, Pharmacy Department, Programa de Pós-Graduação em Assistência Farmacêutica, Maringá, Brazil
| | - Felipe Francisco Tuon
- - Laboratory of Emerging Infectious Diseases, Pontifical Catholic University of Paraná, Curitiba
| |
Collapse
|
41
|
Hamed MM, Abdelsamie AS, Rox K, Schütz C, Kany AM, Röhrig T, Schmelz S, Blankenfeldt W, Arce‐Rodriguez A, Borrero‐de Acuña JM, Jahn D, Rademacher J, Ringshausen FC, Cramer N, Tümmler B, Hirsch AKH, Hartmann RW, Empting M. Towards Translation of PqsR Inverse Agonists: From In Vitro Efficacy Optimization to In Vivo Proof-of-Principle. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204443. [PMID: 36596691 PMCID: PMC9929129 DOI: 10.1002/advs.202204443] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/28/2022] [Indexed: 06/17/2023]
Abstract
Pseudomonas aeruginosa (PA) is an opportunistic human pathogen, which is involved in a wide range of dangerous infections. It develops alarming resistances toward antibiotic treatment. Therefore, alternative strategies, which suppress pathogenicity or synergize with antibiotic treatments are in great need to combat these infections more effectively. One promising approach is to disarm the bacteria by interfering with their quorum sensing (QS) system, which regulates the release of various virulence factors as well as biofilm formation. Herein, this work reports the rational design, optimization, and in-depth profiling of a new class of Pseudomonas quinolone signaling receptor (PqsR) inverse agonists. The resulting frontrunner compound features a pyrimidine-based scaffold, high in vitro and in vivo efficacy, favorable pharmacokinetics as well as clean safety pharmacology characteristics, which provide the basis for potential pulmonary as well as systemic routes of administration. An X-ray crystal structure in complex with PqsR facilitated further structure-guided lead optimization. The compound demonstrates potent pyocyanin suppression, synergizes with aminoglycoside antibiotic tobramycin against PA biofilms, and is active against a panel of clinical isolates from bronchiectasis patients. Importantly, this in vitro effect translated into in vivo efficacy in a neutropenic thigh infection model in mice providing a proof-of-principle for adjunctive treatment scenarios.
Collapse
Affiliation(s)
- Mostafa M. Hamed
- Helmholtz‐Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) Campus E8.166123SaarbrückenGermany
- German Centre for Infection Research (DZIF)Partner Site Hannover‐Braunschweig Saarbrücken66123SaarbrückenGermany
| | - Ahmed S. Abdelsamie
- Helmholtz‐Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) Campus E8.166123SaarbrückenGermany
- German Centre for Infection Research (DZIF)Partner Site Hannover‐Braunschweig Saarbrücken66123SaarbrückenGermany
- Department of Chemistry of Natural and Microbial ProductsInstitute of Pharmaceutical and Drug Industries ResearchNational Research CentreEl‐Buhouth St.DokkiCairo12622Egypt
| | - Katharina Rox
- German Centre for Infection Research (DZIF)Partner Site Hannover‐Braunschweig Saarbrücken66123SaarbrückenGermany
- Department of Chemical Biology (CBIO)Helmholtz Centre for Infection Research (HZI)Inhoffenstr. 7 Braunschweig38124SaarbrückenGermany
| | - Christian Schütz
- Helmholtz‐Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) Campus E8.166123SaarbrückenGermany
- German Centre for Infection Research (DZIF)Partner Site Hannover‐Braunschweig Saarbrücken66123SaarbrückenGermany
| | - Andreas M. Kany
- Helmholtz‐Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) Campus E8.166123SaarbrückenGermany
- German Centre for Infection Research (DZIF)Partner Site Hannover‐Braunschweig Saarbrücken66123SaarbrückenGermany
| | - Teresa Röhrig
- Helmholtz‐Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) Campus E8.166123SaarbrückenGermany
- German Centre for Infection Research (DZIF)Partner Site Hannover‐Braunschweig Saarbrücken66123SaarbrückenGermany
| | - Stefan Schmelz
- Department of Structure and Function of Proteins (SFPR)Helmholtz Centre for Infection Research (HZI)Inhoffenstr. 7 Braunschweig38124SaarbrückenGermany
| | - Wulf Blankenfeldt
- Department of Structure and Function of Proteins (SFPR)Helmholtz Centre for Infection Research (HZI)Inhoffenstr. 7 Braunschweig38124SaarbrückenGermany
- Institute for BiochemistryBiotechnology and BioinformaticsTechnische Universität BraunschweigBraunschweigGermany
| | | | - José Manuel Borrero‐de Acuña
- Institute of MicrobiologyTechnische Universität Braunschweig38106BraunschweigGermany
- Braunschweig Integrated Centre of Systems Biology (BRICS)Technische Universität Braunschweig38106BraunschweigGermany
- Departamento de MicrobiologíaFacultad de BiologíaUniversidad de SevillaAv. de la Reina Mercedesno. 6SevillaCP 41012Spain
| | - Dieter Jahn
- Institute of MicrobiologyTechnische Universität Braunschweig38106BraunschweigGermany
- Braunschweig Integrated Centre of Systems Biology (BRICS)Technische Universität Braunschweig38106BraunschweigGermany
| | - Jessica Rademacher
- Department for Respiratory MedicineMedizinische Hochschule HannoverCarl‐Neuberg‐Str. 130625HannoverGermany
- Biomedical Research in Endstage and Obstructive Lung Disease (BREATH)German Center for Lung Research (DZL)30625HannoverGermany
| | - Felix C. Ringshausen
- Department for Respiratory MedicineMedizinische Hochschule HannoverCarl‐Neuberg‐Str. 130625HannoverGermany
- Biomedical Research in Endstage and Obstructive Lung Disease (BREATH)German Center for Lung Research (DZL)30625HannoverGermany
- European Reference Network on Rare and Complex Respiratory Diseases (ERN‐ LUNG)FrankfurtGermany
| | - Nina Cramer
- Department for Pediatric PneumologyAllergology and NeonatologyMedizinische Hochschule HannoverCarl‐Neuberg‐Str. 130625HannoverGermany
| | - Burkhard Tümmler
- Biomedical Research in Endstage and Obstructive Lung Disease (BREATH)German Center for Lung Research (DZL)30625HannoverGermany
- Department for Pediatric PneumologyAllergology and NeonatologyMedizinische Hochschule HannoverCarl‐Neuberg‐Str. 130625HannoverGermany
| | - Anna K. H. Hirsch
- Helmholtz‐Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) Campus E8.166123SaarbrückenGermany
- German Centre for Infection Research (DZIF)Partner Site Hannover‐Braunschweig Saarbrücken66123SaarbrückenGermany
- Department of PharmacySaarland University Campus E8.166123SaarbrückenGermany
| | - Rolf W. Hartmann
- Helmholtz‐Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) Campus E8.166123SaarbrückenGermany
- German Centre for Infection Research (DZIF)Partner Site Hannover‐Braunschweig Saarbrücken66123SaarbrückenGermany
- Department of PharmacySaarland University Campus E8.166123SaarbrückenGermany
| | - Martin Empting
- Helmholtz‐Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) Campus E8.166123SaarbrückenGermany
- German Centre for Infection Research (DZIF)Partner Site Hannover‐Braunschweig Saarbrücken66123SaarbrückenGermany
- Department of PharmacySaarland University Campus E8.166123SaarbrückenGermany
| |
Collapse
|
42
|
Coste A, Bellouard R, Deslandes G, Jalin L, Roger C, Ansart S, Dailly E, Bretonnière C, Grégoire M. Development of a Predictive Dosing Nomogram to Achieve PK/PD Targets of Amikacin Initial Dose in Critically Ill Patients: A Non-Parametric Approach. Antibiotics (Basel) 2023; 12:antibiotics12010123. [PMID: 36671324 PMCID: PMC9854650 DOI: 10.3390/antibiotics12010123] [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: 12/01/2022] [Revised: 12/30/2022] [Accepted: 01/04/2023] [Indexed: 01/10/2023] Open
Abstract
French guidelines recommend reaching an amikacin concentration of ≥8 × MIC 1 h after beginning infusion (C1h), with MIC = 8 mg/L for probabilistic therapy. We aimed to elaborate a nomogram guiding clinicians in choosing the right first amikacin dose for ICU patients in septic shock. A total of 138 patients with 407 observations were prospectively recruited. A population pharmacokinetic model was built using a non-parametric, non-linear mixed-effects approach. The total body weight (TBW) influenced the central compartment volume, and the glomerular filtration rate (according to the CKD-EPI formula) influenced its clearance. A dosing nomogram was produced using Monte Carlo simulations of the amikacin amount needed to achieve a C1h ≥ 8 × MIC. The dosing nomogram recommended amikacin doses from 1700 mg to 4200 mg and from 28 mg/kg to 49 mg/kg depending on the patient's TBW and renal clearance. However, a Cthrough ≤ 2.5 mg/L 24 h and 48 h after an optimal dose of amikacin was obtained with probabilities of 0.20 and 0.81, respectively. Doses ≥ 30 mg/kg are required to achieve a C1h ≥ 8 × MIC with MIC = 8 mg/L. Targeting a MIC = 8 mg/L should depend on local ecology.
Collapse
Affiliation(s)
- Anne Coste
- Service de Maladies Infectieuses et Tropicales, CHU de Brest, 29200 Brest, France
- Cibles et Médicaments des Infections et de l’Immunité, 9 IICiMed, UR1155, Nantes Université, 44000 Nantes, France
- Laboratoire de Traitement de l’Information Médicale, INSERM, UMR1101, Brest Université, 29200 Brest, France
- Correspondence:
| | - Ronan Bellouard
- Cibles et Médicaments des Infections et de l’Immunité, 9 IICiMed, UR1155, Nantes Université, 44000 Nantes, France
- Service de Pharmacologie Clinique, CHU Nantes, 44000 Nantes, France
| | | | - Laurence Jalin
- Unité de Neuro-Anesthésie-Réanimation, Groupe Hospitalier Pitié-Salpétrière, AP-HP, 75013 Paris, France
| | - Claire Roger
- Département d’anesthésie et réanimation, douleur et médecine d’urgence, CHU Carémeau, 30029 Nîmes, France
- UR UM 103 IMAGINE, Faculté de Médecine, Montpellier Université, 30029 Nîmes, France
| | - Séverine Ansart
- Service de Maladies Infectieuses et Tropicales, CHU de Brest, 29200 Brest, France
- Laboratoire de Traitement de l’Information Médicale, INSERM, UMR1101, Brest Université, 29200 Brest, France
| | - Eric Dailly
- Cibles et Médicaments des Infections et de l’Immunité, 9 IICiMed, UR1155, Nantes Université, 44000 Nantes, France
- Service de Pharmacologie Clinique, CHU Nantes, 44000 Nantes, France
| | - Cédric Bretonnière
- Service des Soins Intensifs de Pneumologie, CHU Nantes, 44000 Nantes, France
| | - Matthieu Grégoire
- Cibles et Médicaments des Infections et de l’Immunité, 9 IICiMed, UR1155, Nantes Université, 44000 Nantes, France
- Service de Pharmacologie Clinique, CHU Nantes, 44000 Nantes, France
| |
Collapse
|
43
|
Koishi N, Sasano H, Yoshizawa T, Shikuri M, Matsumoto H, Suzuki M, Fukui Y, Chonan M, Kimura T, Ichida H, Saiura A, Naito T. Successful Treatment of a Case of Metallo-Beta-Lactamase-Producing Raoultella ornithinolytica Bacteremia by Antimicrobial Stewardship Team Intervention and Therapeutic Drug Monitoring-Based Amikacin Treatment. Case Rep Infect Dis 2023; 2023:5574769. [PMID: 37065978 PMCID: PMC10104729 DOI: 10.1155/2023/5574769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/17/2023] [Accepted: 03/17/2023] [Indexed: 04/18/2023] Open
Abstract
An 80-year-old woman underwent pancreatoduodenectomy. Post-operation, she experienced a fever, and a culture of blood revealed metallo-beta-lactamase-producing Raoultella ornithinolytica. For treatments with aminoglycoside antimicrobial agents, a therapeutic drug monitoring-based dosing design can lower the risk of adverse events and enable appropriate treatment. Key Clinical Message. When aminoglycoside antimicrobial agents are administered for MBL-producing bacteremia, prescription suggestions based on TDM by antimicrobial stewardship team can reduce the occurrence of adverse events and enable appropriate treatment.
Collapse
Affiliation(s)
- Noriko Koishi
- Department of Pharmacy, Juntendo University Hospital, Tokyo, Japan
| | - Hiroshi Sasano
- Department of Pharmacy, Juntendo University Hospital, Tokyo, Japan
- Department of General Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
| | | | - Mika Shikuri
- Department of Pharmacy, Juntendo University Hospital, Tokyo, Japan
| | | | - Mai Suzuki
- Department of General Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Yukiko Fukui
- Department of General Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Masayoshi Chonan
- Department of Clinical Laboratory, Juntendo University Hospital, Tokyo, Japan
| | - Toshimi Kimura
- Department of Pharmacy, Juntendo University Hospital, Tokyo, Japan
| | - Hirofumi Ichida
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Akio Saiura
- Department of Hepatobiliary-Pancreatic Surgery, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Toshio Naito
- Department of General Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
44
|
Kayser N, Cunningham K, Alabsi S, Smith H. Targeting Lower Serum Trough Concentrations: A New Gentamicin Dosing Strategy for Suspected Neonatal Early-Onset Sepsis. J Pediatr Pharmacol Ther 2023; 28:71-77. [PMID: 36777986 PMCID: PMC9901316 DOI: 10.5863/1551-6776-28.1.71] [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: 11/10/2020] [Accepted: 06/09/2021] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Neonatal gentamicin dosing algorithms are not designed to achieve serum trough concentrations ≤1 mcg/mL. The purpose of our study was to evaluate a new gentamicin algorithm based on serum creatinine (SCr) and gestational age (GA) designed to achieve serum gentamicin trough concentrations ≤1 mcg/mL. METHODS A retrospective cohort study was conducted in a level IIIB neonatal intensive care unit. The incidence of elevated serum gentamicin troughs for this study was compared with the center's previously published results to evaluate the proposed dosing algorithm. Patients were included if gentamicin was administered within the first 7 days of life and a serum gentamicin trough concentration and a baseline SCr concentration were obtained. Patients were further subdivided into groups based on GA for data analysis: ≤30 weeks (group 1), 30-34 weeks (group 2), and ≥35 weeks (group 3). The SCr was considered mildly elevated (0.81-0.99 mg/dL) or elevated (≥1 mg/dL). The respective outcomes between the post-algorithm and control groups were examined using intention-to-treat analysis and Bayesian modeling to calculate rate differences. RESULTS Of the 2377 patients evaluated, 366 met the inclusion criteria. Significantly lower percentages of elevated serum gentamicin troughs were noted in groups 2 and 3 subsequent to the implementation of the dosing algorithm with 16% and 15% lower rate differences, respectively. Regardless of GA, there were significantly fewer elevated serum troughs in the post-implementation groups than in the control with mildly elevated and elevated SCr p < 0.001. CONCLUSIONS Using a dosing algorithm based on SCr significantly reduced the number of elevated serum trough rates in neonates with a GA greater than 30 weeks.
Collapse
Affiliation(s)
- Nicholas Kayser
- Department of Pharmacy (NK, KC), UnityPoint Des Moines–Blank Children's Hospital, Des Moines, IA
| | - Kelli Cunningham
- Department of Pharmacy (NK, KC), UnityPoint Des Moines–Blank Children's Hospital, Des Moines, IA
| | - Samir Alabsi
- Neonatal Intensive Care Unit (SA), UnityPoint Des Moines–Blank Children's Hospital, Des Moines, IA
| | - Hayden Smith
- Education and Research Department (HS), UnityPoint Des Moines–Iowa Methodist Medical Center, Des Moines, IA
| |
Collapse
|
45
|
Davido B, Crémieux AC, Vaugier I, Gatin L, Noussair L, Massias L, Laurent F, Saleh-Mghir A. Efficacy of ceftazidime-avibactam in various combinations for the treatment of experimental osteomyelitis due to Klebsiella pneumoniae carbapenemase (KPC)-producing Klebsiella pneumoniae. Int J Antimicrob Agents 2023; 61:106702. [PMID: 36476965 DOI: 10.1016/j.ijantimicag.2022.106702] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/24/2022] [Accepted: 11/26/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Optimal treatment of carbapenemase-producing Enterobacterales (CPE) bone infections is poorly defined. This study evaluated the efficacy of the novel beta-lactam-beta-lactamase inhibitor-ceftazidime-avibactam (CAZ-AVI)-with different antibiotic combinations in an experimental model of CPE osteomyelitis. METHODS KPC-99YC is a clinical strain of Klebsiella pneumoniae carbapenemase (KPC)-producing Klebsiella pneumoniae with intermediate susceptibility to meropenem (MIC 4 mg/L), gentamicin (MIC 0.25 mg/L), colistin (MIC 0.25 mg/L), fosfomycin (MIC 4 mg/L) and ceftazidime-avibactam (MIC 1 mg/L). Time-kill curves were performed at 4x MIC. Osteomyelitis was induced in rabbits by tibial injection of 2×108 CFU of KPC-99YC. Six groups started treatment 14 days later for 7 days: control, colistin, CAZ-AVI, CAZ-AVI plus gentamicin, CAZ-AVI plus colistin and CAZ-AVI plus fosfomycin. Antibiotic dosages were selected to simulate plasma concentrations obtained in humans. Treatment was evaluated according to bone cultures quantified in log10 CFU. RESULTS In vitro, CAZ-AVI plus colistin or gentamicin were rapidly bactericidal in contrast with CAZ-AVI plus fosfomycin. In vivo, compared with controls, colistin alone (P = 0.045) and CAZ-AVI alone or in combination significantly lowered bone bacterial counts (P < 0.001). Bone sterilisation was achieved in 67% and 100% of animals with combinations of CAZ-AVI plus colistin or gentamicin (P = 0.001 and P < 0.001, respectively) whereas other treatments were no different from controls. CAZ-AVI plus gentamicin provided greater bone bacterial reduction than CAZ-AVI plus colistin (P = 0.033). No CAZ-AVI-resistant strains emerged in treated rabbits, regardless of combination. CONCLUSIONS CAZ-AVI plus gentamicin was the best effective combination therapy. Combinations with CAZ-AVI appear to be a promising treatment of KPC-producing Klebsiella pneumoniae osteomyelitis.
Collapse
Affiliation(s)
- Benjamin Davido
- UMR 1173, Versailles Saint-Quentin University, Versailles, France; Raymond Poincaré Paris Saclay University Hospital, Garches, France.
| | - Anne-Claude Crémieux
- UMR 1173, Versailles Saint-Quentin University, Versailles, France; FHU PROTHEE, St Louis Hospital, Paris-Cité University, Paris, France
| | - Isabelle Vaugier
- CIC, Raymond Poincaré Paris Saclay University Hospital, Garches, France
| | - Laure Gatin
- UMR 1173, Versailles Saint-Quentin University, Versailles, France
| | - Latifa Noussair
- Microbiology Unit, Raymond Poincaré Paris Saclay University Hospital, Garches, France
| | - Laurent Massias
- Toxicology Unit, Bichat Paris Nord University Hospital, Paris, France
| | - Frederic Laurent
- Institut for Infectious Agents, Department of Bacteriology - CNR des staphylocoques, Croix-Rousse Hospital, North Biology Centre, Hospices Civils de Lyon, Lyon, France; Team "Staphylococcal pathogenesis", International Centre for Infectiology Research, INSERM U1111 - CNRS UMR5308 - ENS Lyon - Lyon 1 University, Lyon, France
| | - Azzam Saleh-Mghir
- UMR 1173, Versailles Saint-Quentin University, Versailles, France; Raymond Poincaré Paris Saclay University Hospital, Garches, France
| |
Collapse
|
46
|
When and How to Use MIC in Clinical Practice? Antibiotics (Basel) 2022; 11:antibiotics11121748. [PMID: 36551405 PMCID: PMC9774413 DOI: 10.3390/antibiotics11121748] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/22/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Bacterial resistance to antibiotics continues to be a global public health problem. The choice of the most effective antibiotic and the use of an adapted dose in the initial phase of the infection are essential to limit the emergence of resistance. This will depend on (i) the isolated bacteria and its resistance profile, (ii) the pharmacodynamic (PD) profile of the antibiotic used and its level of toxicity, (iii) the site of infection, and (iv) the pharmacokinetic (PK) profile of the patient. In order to take account of both parameters to optimize the administered treatment, a minimal inhibitory concentration (MIC) determination associated with therapeutic drug monitoring (TDM) and their combined interpretation are required. The objective of this narrative review is thus to suggest microbiological, pharmacological, and/or clinical situations for which this approach could be useful. Regarding the microbiological aspect, such as the detection of antibiotic resistance and its level, the preservation of broad-spectrum β-lactams is particularly discussed. PK-PD profiles are relevant for difficult-to-reach infections and specific populations such as intensive care patients, cystic fibrosis patients, obese, or elderly patients. Finally, MIC and TDM are tools available to clinicians, who should not hesitate to use them to manage their patients.
Collapse
|
47
|
Pajot O, Lakhal K, Lambert J, Gros A, Bruel C, Boulain T, Garot D, Das V, Timsit JF, Cerf C, Souweine B, Chaffaut C, Mentec H, Zahar JR, Mira JP, Jullien V. Empirical Antibiotic Therapy for Gram-Negative Bacilli Ventilator-Associated Pneumonia: Observational Study and Pharmacodynamic Assessment. Antibiotics (Basel) 2022; 11:antibiotics11111664. [PMID: 36421308 PMCID: PMC9686941 DOI: 10.3390/antibiotics11111664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/10/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Background: Strong evidence suggests a correlation between pharmacodynamics (PD) index and antibiotic efficacy while dose adjustment should be considered in critically ill patients due to modified pharmacokinetic (PK) parameters and/or higher minimum inhibitory concentrations (MICs). This study aimed to assess pharmacodynamic (PD) target attainment considering both antibiotics serum concentrations and measured MICs in these patients. Method: A multicentric prospective open-label trial conducted in 11 French ICUs involved patients with Gram-negative bacilli (GNB) ventilator-associated pneumonia (VAP) confirmed by quantitative cultures. Results: We included 117 patients. Causative GNBs were P. aeruginosa (40%), Enterobacter spp. (23%), E. coli (20%), and Klebsiella spp. (16%). Hence, 117 (100%) patients received β-lactams, 65 (58%) aminoglycosides, and two (1.5%) fluoroquinolones. For β-lactams, 83% of the patients achieved a Cmin/MIC > 1 and 70% had a Cmin/MIC > 4. In the case of high creatinine clearance (CrCL > 100 mL/min/1.73 m2), 70.4% of the patients achieved a Cmin/MIC ratio > 1 versus 91% otherwise (p = 0.041), and 52% achieved a Cmin/MIC ratio > 4 versus 81% (p = 0.018). For aminoglycosides, 94% of the patients had a Cmax/MIC ratio > 8. Neither β-lactams nor aminoglycosides PK/PD parameters were associated clinical outcomes, but our data suggest a correlation between β-lactams Cmin/MIC and microbiological success. Conclusion: In our ICU patients treated for GNB VAP, using recommended antibiotic dosage led in most cases to PK/PD targets attainment for aminoglycosides and β-lactams. High creatinine clearance should encourage clinicians to focus on PK/PD issues.
Collapse
Affiliation(s)
- Olivier Pajot
- Victor Dupouy Hospital, Intensive Care Unit, F-95100 Argenteuil, France
- Correspondence: ; Tel.: +33-134232455
| | - Karim Lakhal
- Service d’Anesthésie-Réanimation, Hôpital Laënnec, Centre Hospitalier Universitaire, F-44093 Nantes, France
| | - Jérome Lambert
- Department of Biostatistics and Medical Information, APHP, Saint-Louis Hospital, F-75010 Paris, France
| | - Antoine Gros
- Medical-Surgical Intensive Care Unit, André Mignot Hospital, F-78150 Le Chesnay, France
| | - Cédric Bruel
- Medical and Surgical Intensive Care Unit, Paris Saint-Joseph Hospital Network, F-75014 Paris, France
| | - Thierry Boulain
- Intensive Care Unit, Orleans Regional Hospital, 14 Avenue de L’Hôpital CS 86709, CEDEX 02, F-45067 Orléans, France
| | - Denis Garot
- Service de Médecine Intensive Réanimation, Hôpital Bretonneau, CHU Tours, F-37000 Tours, France
| | - Vincent Das
- Service de Médecine Intensive Réanimation, Centre Hospitalier Intercommunal André Grégoire, F-93100 Montreuil, France
| | - Jean François Timsit
- AP-HP, Bichat Hospital, Medical and Infectious Diseases Intensive Care Unit (MI2), F-75018 Paris, France
| | - Charles Cerf
- Intensive Care Unit, Foch Hospital, F-92150 Suresnes, France
| | - Bertrand Souweine
- CHU Clermont-Ferrand, Service de Réanimation Médicale, F-63000 Clermont-Ferrand, France
| | - Cendrine Chaffaut
- Department of Biostatistics and Medical Information, APHP, Saint-Louis Hospital, F-75010 Paris, France
| | - Hervé Mentec
- Victor Dupouy Hospital, Intensive Care Unit, F-95100 Argenteuil, France
| | - Jean Ralph Zahar
- AP-HP, Hôpital Avicenne, Prévention du Risque Infectieux, GH Paris Seine Saint-Denis, F-93000 Bobigny, France
| | - Jean Paul Mira
- Department of Medical Intensive Care, Cochin University Hospital, F-75014 Paris, France
| | - Vincent Jullien
- Pharmacology Unit, University Sorbonne Paris Nord, Groupe Hospitalier Paris Seine-Saint-Denis, Assistance Publique-Hôpitaux de Paris, Hôpital Jean Verdier, F-93140 Bondy, France
| |
Collapse
|
48
|
Sherwin E, King C, Hasen H, May S. Single-dose intravesical amikacin instillation for pyocystis in a patient with autonomic dysreflexia: A case report. J Spinal Cord Med 2022; 45:965-968. [PMID: 33983103 PMCID: PMC9661977 DOI: 10.1080/10790268.2021.1922832] [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] [Indexed: 10/21/2022] Open
Abstract
CONTEXT Pyocystis is an infection of the epithelium of the bladder and a frequent complication of anuria. Patients with spinal cord injury (SCI) at the sixth thoracic vertebra (T6) or higher are at a greater risk for autonomic dysreflexia (AD), which can be induced by infections such as pyocystis. Cases of pyocystis treatment with aminoglycoside instillations have been reported in the literature. FINDINGS We describe the case of a 59-year-old male with T1 American Spinal Injury Association (ASIA) Impairment Scale (AIS) grade A complete paraplegia, status post bilateral nephrectomy with recurrent episodes of AD suspected to be caused by pyocystis related to anuria. A bladder specimen culture grew Escherichia coli susceptible to amikacin with a minimum inhibitory concentration (MIC) of ≤ 8 mg/L. In the setting of anuria and with concern that intravenous antibiotics would not adequately reach the site of infection, we chose to treat the patient with a single-dose intravesical instillation of amikacin 25 mg/100 mL left to dwell for approximately 2 h. A repeat bladder culture showed no colonies. The patient remained stable with no episodes of AD and no signs or symptoms of infection one month following treatment. CONCLUSION The purpose of this case report is to add to the current literature on intravesical aminoglycoside instillations for pyocystis to aid clinicians in treating future cases, as well as to highlight pyocystis as a potential cause of AD in SCI patients with anuria.
Collapse
Affiliation(s)
- Erin Sherwin
- College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA,Correspondence to: Erin Sherwin, Science Center College of Pharmacy, University of Tennessee Health, 700 South 19th Street, Birmingham, AL35233, USA; Ph: (205) 933-8101 ext. 6401.
| | - Cynthia King
- Veteran’s Affairs Medical Center, Memphis, Tennessee,USA
| | - Howard Hasen
- Veteran’s Affairs Medical Center, Memphis, Tennessee,USA
| | - Shari May
- College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
49
|
Luxton TN, King N, Wälti C, Jeuken LJC, Sandoe JAT. A Systematic Review of the Effect of Therapeutic Drug Monitoring on Patient Health Outcomes during Treatment with Carbapenems. Antibiotics (Basel) 2022; 11:antibiotics11101311. [PMID: 36289971 PMCID: PMC9598625 DOI: 10.3390/antibiotics11101311] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/23/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
Adjusting dosing regimens based on measurements of carbapenem levels may improve carbapenem exposure in patients. This systematic review aims to describe the effect carbapenem therapeutic drug monitoring (TDM) has on health outcomes, including the emergence of antimicrobial resistance (AMR). Four databases were searched for studies that reported health outcomes following adjustment to dosing regimens, according to measurements of carbapenem concentration. Bias in the studies was assessed with risk of bias analysis tools. Study characteristics and outcomes were tabulated and a narrative synthesis was performed. In total, 2 randomised controlled trials (RCTs), 17 non-randomised studies, and 19 clinical case studies were included. Significant variation in TDM practice was seen; consequently, a meta-analysis was unsuitable. Few studies assessed impacts on AMR. No significant improvement on health outcomes and no detrimental effects of carbapenem TDM were observed. Five cohort studies showed significant associations between achieving target concentrations and clinical success, including suppression of resistance. Studies in this review showed no obvious improvement in clinical outcomes when TDM is implemented. Optimisation and standardisation of carbapenem TDM practice are needed to improve intervention success and enable study synthesis. Further suitably powered studies of standardised TDM are required to assess the impact of TMD on clinical outcomes and AMR.
Collapse
Affiliation(s)
- Timothy N. Luxton
- School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, UK
- Correspondence:
| | - Natalie King
- Leeds Institute of Health Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Christoph Wälti
- School of Electronic and Electrical Engineering, University of Leeds, Leeds LS2 9JT, UK
| | - Lars J. C. Jeuken
- Leiden Institute of Chemistry, Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | | |
Collapse
|
50
|
Krueger CK, Bruno JJ, Tverdek FP, Hernandez M, Abudayyeh A. Aminoglycoside Pharmacokinetics in Critically Ill Patients Undergoing Continuous Renal Replacement Therapy. Ann Pharmacother 2022; 57:629-636. [PMID: 36062536 DOI: 10.1177/10600280221120600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND There are few studies describing aminoglycoside pharmacokinetics during continuous renal replacement therapy (CRRT). OBJECTIVE To characterize the effect of CRRT on aminoglycoside clearance and volume of distribution (Vd). METHODS Retrospective observational pharmacokinetic study of adult critically ill oncologic patients who received a first dose of amikacin or tobramycin during CRRT between February 2012 and May 2017. Study outcomes included aminoglycoside clearance, Vd, and attainment of the target peak: MIC (minimum inhibitory concentration) ratio as a surrogate for dosing appropriateness. RESULTS In total, 80 patients were included, sustained low-efficiency dialysis (SLED), n = 49; continuous venovenous hemodialysis (CVVHD), n = 19; continuous venovenous hemofiltration (CVVH), n = 12. Fifty-one patients received amikacin at a median dose of 14.5 mg/kg per actual body weight and achieved a median peak level of 26.7 mg/L. Twenty-nine patients received tobramycin at a median dose of 6.5 mg/kg actual body weight and achieved a median peak level of 10.3 mg/L. The median aminoglycoside clearance was 63.1 mL/min and was similar between CRRT modality groups (P = 0.97). The median Vd was 0.47 L/kg and was different between the SLED and CVVH groups (P = 0.007). Attainment of target peak: MIC occurred in 29% in the total study population and 44% in the subgroup of 23 patients with isolates tested for aminoglycoside susceptibility. CONCLUSION AND RELEVANCE Critically ill oncology patients undergoing CRRT exhibited reduced clearance and expanded Vd that was not significantly different between CRRT modalities. Current dosing regimens led to low peak concentrations and poor attainment of pharmacokinetic targets.
Collapse
Affiliation(s)
- Chelsea K Krueger
- Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Dallas, TX, USA
| | - Jeffrey J Bruno
- Division of Pharmacy, MD Anderson Cancer Center, The University of Texas, Houston, TX, USA
| | - Frank P Tverdek
- Seattle Cancer Care Alliance, University of Washington, Seattle, WA, USA
| | - Mike Hernandez
- Department of Biostatistics, MD Anderson Cancer Center, The University of Texas, Houston, TX, USA
| | - Ala Abudayyeh
- Section of Nephrology, MD Anderson Cancer Center, The University of Texas, Houston, TX, USA
| |
Collapse
|