1
|
Schouwenburg S, Keij FM, Tramper-Stranders GA, Kornelisse RF, Reiss IKM, de Cock PAJG, Dhont E, Watt KM, Muller AE, Flint RB, Koch BCP, Allegaert K, Preijers T. A Pooled Population Pharmacokinetic Study of Oral and Intravenous Administration of Clavulanic Acid in Neonates and Infants: Targeting Effective Beta-Lactamase Inhibition. Clin Pharmacol Ther 2024. [PMID: 39205386 DOI: 10.1002/cpt.3423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024]
Abstract
Data published on the oral clavulanic acid pharmacokinetics in the pediatric population is lacking. This research aimed to describe clavulanic acid disposition following oral and intravenous administration and to provide insights into clavulanic acid exposure based on threshold concentrations for (pre-)term neonates and infants. This pooled population pharmacokinetic study combined four datasets for analysis in NONMEM v7.4.3. Clavulanic acid exposure was simulated using the percentage of time above the threshold concentrations (%fT > CT). Multiple dosage regimens and amoxicillin/clavulanic acid dosage ratios were evaluated. The cohort consisted of 89 (42 oral, 47 intravenous) subjects (403 samples) with a median (range) postnatal age 54.5 days (0-365), gestational age 37.4 weeks (23.0-41.7), and current bodyweight 3.9 kg (0.6-9.0). A one-compartment model with first-order absorption best described clavulanic acid pharmacokinetics with postnatal age as a covariate on the inter-individual variability of clearance. Oral bioavailability was 24.4% in neonates up to 10 days of age. An oral dosing regimen 90 mg/kg/day amoxicillin/clavulanic acid (4:1 ratio) resulted in 40.2% of simulated patients achieving 100% fT > CT,2mg/L. An amoxicillin/clavulanic acid ratio of 4:1 is preferred for neonatal oral regimens due to the higher exposure along the entire %fT > CT range (0-100%) as ratios higher than 4:1 might result in inadequate exposure. Our results highlight substantial exposure differences (%fT > CT) when using threshold concentrations of 1 mg/L vs. 2 mg/L. This first population pharmacokinetic model for clavulanic acid in neonates may serve as a foundational step for future research, once more precise clavulanic acid targets become available.
Collapse
Affiliation(s)
- Stef Schouwenburg
- Department of Hospital Pharmacy, Erasmus University Medical Centre, Rotterdam, The Netherlands
- Rotterdam Clinical Pharmacometrics Group, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Fleur M Keij
- Division of Neonatology, Department of Neonatal and Pediatric Intensive Care, Erasmus University Medical Centre-Sophia Children's Hospital, Rotterdam, The Netherlands
- Department of Paediatrics, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | - Gerdien A Tramper-Stranders
- Division of Neonatology, Department of Neonatal and Pediatric Intensive Care, Erasmus University Medical Centre-Sophia Children's Hospital, Rotterdam, The Netherlands
- Department of Paediatrics, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | - René F Kornelisse
- Division of Neonatology, Department of Neonatal and Pediatric Intensive Care, Erasmus University Medical Centre-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Irwin K M Reiss
- Division of Neonatology, Department of Neonatal and Pediatric Intensive Care, Erasmus University Medical Centre-Sophia Children's Hospital, Rotterdam, The Netherlands
- Department of Paediatrics, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | - Pieter A J G de Cock
- Department of Pharmacy, Ghent University Hospital, Ghent, Belgium
- Department of Pediatric Intensive Care, Ghent University Hospital, Ghent, Belgium
| | - Evelyn Dhont
- Department of Pediatric Intensive Care, Ghent University Hospital, Ghent, Belgium
- Department of Basic and Applied Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Kevin M Watt
- Duke University Medical Center, Durham, North Carolina, USA
- Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Anouk E Muller
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Centre, Rotterdam, The Netherlands
- Department of Medical Microbiology, Haaglanden Medisch Centrum, The Hague, The Netherlands
| | - Robert B Flint
- Department of Hospital Pharmacy, Erasmus University Medical Centre, Rotterdam, The Netherlands
- Rotterdam Clinical Pharmacometrics Group, Erasmus University Medical Centre, Rotterdam, The Netherlands
- Division of Neonatology, Department of Neonatal and Pediatric Intensive Care, Erasmus University Medical Centre-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Birgit C P Koch
- Department of Hospital Pharmacy, Erasmus University Medical Centre, Rotterdam, The Netherlands
- Rotterdam Clinical Pharmacometrics Group, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Karel Allegaert
- Department of Hospital Pharmacy, Erasmus University Medical Centre, Rotterdam, The Netherlands
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Tim Preijers
- Department of Hospital Pharmacy, Erasmus University Medical Centre, Rotterdam, The Netherlands
- Rotterdam Clinical Pharmacometrics Group, Erasmus University Medical Centre, Rotterdam, The Netherlands
| |
Collapse
|
2
|
Narayanan N, Mathers AJ, Wenzler E, Moore NM, Giske CG, Mendes RE, Edelstein PH. Amoxicillin-Clavulanate Breakpoints Against Enterobacterales: Rationale for Revision by the Clinical and Laboratory Standards Institute. Clin Infect Dis 2024; 79:516-523. [PMID: 38626241 PMCID: PMC11327796 DOI: 10.1093/cid/ciae201] [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: 01/24/2024] [Revised: 03/18/2024] [Accepted: 04/01/2024] [Indexed: 04/18/2024] Open
Abstract
Amoxicillin-clavulanate (AMC) is among the most frequently prescribed antibiotics globally. It has broad antibacterial activity against gram-positive, gram-negative, and anaerobic bacteria and has been used to treat infections caused by a broad range of pathogens. AMC breakpoints against Enterobacterales were initially set in the 1980s. However, since that time, increases in antibiotic resistance, advances in pharmacokinetic/pharmacodynamic analyses, and publication of additional clinical data prompted a reassessment by the Clinical and Laboratory Standards Institute (CLSI) Subcommittee on Antimicrobial Susceptibility Testing. Based on this contemporary reappraisal, the CLSI retained the Enterobacterales breakpoints but revised comments regarding dosing associated with use of the AMC breakpoints in the 2022 supplement of M100. This viewpoint provides insight into the CLSI breakpoint reevaluation process and summarizes the data and rationale used to support these revisions to the AMC Enterobacterales breakpoint.
Collapse
Affiliation(s)
- Navaneeth Narayanan
- Department of Pharmacy Practice and Administration, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
| | - Amy J Mathers
- Department of Medicine and Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - Eric Wenzler
- Roche Diagnostics Corporation, Indianapolis, Indiana, USA
| | - Nicholas M Moore
- Departments of Internal Medicine and Pathology, Rush University Medical Center, Chicago, Illinois, USA
| | - Christian G Giske
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska University Hospital and Karolinska Institute, Stockholm, Sweden
| | | | - Paul H Edelstein
- Department Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Department of Medicine, Molecular Immunity Unit, Cambridge Institute of Therapeutic Immunology and Infectious Diseases, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
3
|
Dinh A, Duran C, Ropers J, Bouchand F, Deconinck L, Matt M, Senard O, Lagrange A, Mellon G, Calin R, Makhloufi S, de Lastours V, Mathieu E, Kahn JE, Rouveix E, Grenet J, Dumoulin J, Chinet T, Pépin M, Delcey V, Diamantis S, Benhamou D, Vitrat V, Dombret MC, Renaud B, Claessens YE, Labarère J, Bedos JP, Aegerter P, Crémieux AC. Exclusive oral antibiotic treatment for hospitalized community-acquired pneumonia: a post-hoc analysis of a randomized clinical trial. Clin Microbiol Infect 2024; 30:1020-1028. [PMID: 38734138 DOI: 10.1016/j.cmi.2024.05.003] [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: 10/09/2023] [Revised: 04/09/2024] [Accepted: 05/05/2024] [Indexed: 05/13/2024]
Abstract
OBJECTIVES In this study, we aimed to assess the efficacy of different ways of administration and types of beta-lactams for hospitalized community-acquired pneumonia (CAP). METHODS In this post-hoc analysis of randomized controlled trials (RCT) on patients hospitalized for CAP (pneumonia short treatment trial) comparing 3-day vs. 8-day durations of beta-lactams, which concluded to non-inferiority, we included patients who received either amoxicillin-clavulanate (AMC) or third-generation cephalosporin (3GC) regimens, and exclusively either intravenous or oral treatment for the first 3 days (followed by either 5 days of oral placebo or AMC according to randomization). The choice of route and molecule was left to the physician in charge. The main outcome was a failure at 15 days after the first antibiotic intake, defined as temperature >37.9°C, and/or absence of resolution/improvement of respiratory symptoms, and/or additional antibiotic treatment for any cause. The primary outcome according to the route of administration was evaluated through logistic regression. Inverse probability treatment weighting with a propensity score model was used to adjust for non-randomization of treatment routes and potential confounders. The difference in failure rates was also evaluated among several sub-populations (AMC vs. 3GC treatments, intravenous vs. oral AMC, patients with multi-lobar infection, patients aged ≥65 years old, and patients with CURB65 scores of 3-4). RESULTS We included 200 patients from the original trial, with 93/200 (46.5%) patients only treated with intravenous treatment and 107/200 (53.5%) patients only treated with oral therapy. The failure rate at Day 15 was not significantly different among patients treated with initial intravenous vs. oral treatment [25/93 (26.9%) vs. 28/107 (26.2%), adjusted odds ratios (aOR) 0.973 (95% CI 0.519-1.823), p 0.932)]. Failure rates at Day 15 were not significantly different among the subgroup populations. DISCUSSION Among hospitalized patients with CAP, there was no significant difference in efficacy between initial intravenous and exclusive oral treatment. TRIAL REGISTRATION This trial is registered with ClinicalTrials.gov, NCT01963442.
Collapse
Affiliation(s)
- Aurélien Dinh
- Infectious Diseases Unit, Raymond-Poincaré University Hospital, AP-HP Paris Saclay University, Garches, France; Epidemiology and Modeling of Bacterial Evasion to Antibacterials Unit (EMEA), Institut Pasteur, Paris, France.
| | - Clara Duran
- Infectious Diseases Unit, Raymond-Poincaré University Hospital, AP-HP Paris Saclay University, Garches, France
| | - Jacques Ropers
- Clinical Research Unit, Pitié-Salpétrière University Hospital, AP-HP, Paris, France
| | - Frédérique Bouchand
- Department of Pharmacy, Raymond-Poincaré University Hospital, AP-HP Paris Saclay, Garches, France
| | - Laurène Deconinck
- Department of Infectious Disease, Bichat University Hospital, AP-HP, University of Paris, Paris, France
| | - Morgan Matt
- Infectious Diseases Unit, Raymond-Poincaré University Hospital, AP-HP Paris Saclay University, Garches, France
| | - Olivia Senard
- Department of Infectious Disease, Marne La Vallée Hospital, GHEF, Marne La Vallée, France
| | - Aurore Lagrange
- Department of Pneumology, Pontoise Hospital, Pontoise, France
| | - Guillaume Mellon
- Infectious Diseases Unit, Raymond-Poincaré University Hospital, AP-HP Paris Saclay University, Garches, France
| | - Ruxandra Calin
- Infectious Diseases Unit, Raymond-Poincaré University Hospital, AP-HP Paris Saclay University, Garches, France
| | - Sabrina Makhloufi
- Infectious Diseases Unit, Raymond-Poincaré University Hospital, AP-HP Paris Saclay University, Garches, France
| | | | | | - Jean-Emmanuel Kahn
- Internal Medicine, Ambroise-Paré University Hospital, AP-HP Paris Saclay, Boulogne-Billancourt, France
| | - Elisabeth Rouveix
- Internal Medicine, Ambroise-Paré University Hospital, AP-HP Paris Saclay, Boulogne-Billancourt, France
| | - Julie Grenet
- Emergency Medicine, Ambroise-Paré University Hospital, AP-HP Paris Saclay, Boulogne-Billancourt, France
| | - Jennifer Dumoulin
- Department of Pneumology, Ambroise-Paré University Hospital, AP-HP Paris Saclay, Boulogne-Billancourt, France
| | - Thierry Chinet
- Department of Pneumology, Ambroise-Paré University Hospital, AP-HP Paris Saclay, Boulogne-Billancourt, France
| | - Marion Pépin
- Department of Geriatric, Ambroise-Paré University Hospital, AP-HP Paris Saclay, Boulogne-Billancourt, France
| | - Véronique Delcey
- Internal Medicine, Lariboisière University Hospital, AP-HP, Paris, France
| | | | - Daniel Benhamou
- Department of Pneumology, Rouen University Hospital, Rouen, France
| | | | | | - Bertrand Renaud
- Department of Emergency, Cochin University Hospital, AP-HP, Paris, France
| | | | - José Labarère
- Quality of Care Unit, Grenoble University Hospital, Grenoble Alpes University, Grenoble, France
| | | | - Philippe Aegerter
- UMRS 1168 VIMA, INSERM, Versailles Saint-Quentin University, Versailles, France
| | | |
Collapse
|
4
|
Reza N, Gerada A, Stott KE, Howard A, Sharland M, Hope W. Challenges for global antibiotic regimen planning and establishing antimicrobial resistance targets: implications for the WHO Essential Medicines List and AWaRe antibiotic book dosing. Clin Microbiol Rev 2024; 37:e0013923. [PMID: 38436564 PMCID: PMC11324030 DOI: 10.1128/cmr.00139-23] [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] [Indexed: 03/05/2024] Open
Abstract
SUMMARYThe World Health Organisation's 2022 AWaRe Book provides guidance for the use of 39 antibiotics to treat 35 infections in primary healthcare and hospital facilities. We review the evidence underpinning suggested dosing regimens. Few (n = 18) population pharmacokinetic studies exist for key oral AWaRe antibiotics, largely conducted in homogenous and unrepresentative populations hindering robust estimates of drug exposures. Databases of minimum inhibitory concentration distributions are limited, especially for community pathogen-antibiotic combinations. Minimum inhibitory concentration data sources are not routinely reported and lack regional diversity and community representation. Of studies defining a pharmacodynamic target for ß-lactams (n = 80), 42 (52.5%) differed from traditionally accepted 30%-50% time above minimum inhibitory concentration targets. Heterogeneity in model systems and pharmacodynamic endpoints is common, and models generally use intravenous ß-lactams. One-size-fits-all pharmacodynamic targets are used for regimen planning despite complexity in drug-pathogen-disease combinations. We present solutions to enable the development of global evidence-based antibiotic dosing guidance that provides adequate treatment in the context of the increasing prevalence of antimicrobial resistance and, moreover, minimizes the emergence of resistance.
Collapse
Affiliation(s)
- Nada Reza
- Department of
Antimicrobial Pharmacodynamics and Therapeutics, Institute of Systems,
Molecular and Integrative Biology, University of
Liverpool, Liverpool,
United Kingdom
- Liverpool University
Hospitals NHS Foundation Trust,
Liverpool, United Kingdom
| | - Alessandro Gerada
- Department of
Antimicrobial Pharmacodynamics and Therapeutics, Institute of Systems,
Molecular and Integrative Biology, University of
Liverpool, Liverpool,
United Kingdom
- Liverpool University
Hospitals NHS Foundation Trust,
Liverpool, United Kingdom
| | - Katharine E. Stott
- Department of
Antimicrobial Pharmacodynamics and Therapeutics, Institute of Systems,
Molecular and Integrative Biology, University of
Liverpool, Liverpool,
United Kingdom
- Liverpool University
Hospitals NHS Foundation Trust,
Liverpool, United Kingdom
| | - Alex Howard
- Department of
Antimicrobial Pharmacodynamics and Therapeutics, Institute of Systems,
Molecular and Integrative Biology, University of
Liverpool, Liverpool,
United Kingdom
- Liverpool University
Hospitals NHS Foundation Trust,
Liverpool, United Kingdom
| | - Mike Sharland
- Centre for Neonatal
and Paediatric Infection, Institute for Infection and Immunity, St
George’s, University of London,
London, United Kingdom
| | - William Hope
- Department of
Antimicrobial Pharmacodynamics and Therapeutics, Institute of Systems,
Molecular and Integrative Biology, University of
Liverpool, Liverpool,
United Kingdom
- Liverpool University
Hospitals NHS Foundation Trust,
Liverpool, United Kingdom
| |
Collapse
|
5
|
Keij FM, Tramper-Stranders GA, Koch BCP, Reiss IKM, Muller AE, Kornelisse RF, Allegaert K. Pharmacokinetics of Clavulanic Acid in the Pediatric Population: A Systematic Literature Review. Clin Pharmacokinet 2022; 61:637-653. [PMID: 35355215 PMCID: PMC9095526 DOI: 10.1007/s40262-022-01116-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2022] [Indexed: 11/24/2022]
Abstract
Background and Objective Clavulanic acid is a commonly used β-lactam inhibitor in pediatrics for a variety of infections. Clear insight into its mode of action is lacking, however, and a target has not been identified. The dosing of clavulanic acid is currently based on that of the partner drug (amoxicillin or ticarcillin). Still, proper dosing of the compound is needed because clavulanic acid has been associated with adverse effects. In this systematic review, we aim to describe the current literature on the pharmacokinetics of clavulanic acid in the pediatric population Methods We performed a systematic search in MEDLINE, Embase.com, Cochrane Central, Google Scholar, and Web of Science. We included all published studies reporting pharmacokinetic data on clavulanic acid in neonates and children 0–18 years of age. Results The search resulted in 18 original studies that met the inclusion criteria. In general, the variation in drug exposure was large, which can be partly explained by differences in disease state, route of administration, or age. Unfortunately, the studies’ limited background information hampered in-depth assessment of the observed variability. Conclusion The pharmacokinetics of clavulanic acid in pediatric patients is highly variable, similar to reports in adults, but more pronounced. Significant knowledge gaps remain with regard to the population-specific explanation for this variability. Model-based pharmacokinetic studies that address both maturational and disease-specific changes in the pediatric population are therefore needed. Furthermore, additional pharmacodynamic studies are needed to define a clear target. The combined outcomes will eventually lead to pharmacokinetic-pharmacodynamic modeling of clavulanic acid and targeted exposure. Clinical Trial Registration PROSPERO CRD42020137253. Supplementary Information The online version contains supplementary material available at 10.1007/s40262-022-01116-3.
Collapse
Affiliation(s)
- Fleur M Keij
- Department of Pediatrics, Division of Neonatology, Erasmus MC Sophia Children's Hospital, Erasmus University Medical Center Rotterdam, Doctor Molenwaterplein 40, 3015 CN, Rotterdam, The Netherlands. .,Department of Pediatrics, Franciscus Gasthuis and Vlietland, Rotterdam, The Netherlands.
| | - Gerdien A Tramper-Stranders
- Department of Pediatrics, Division of Neonatology, Erasmus MC Sophia Children's Hospital, Erasmus University Medical Center Rotterdam, Doctor Molenwaterplein 40, 3015 CN, Rotterdam, The Netherlands.,Department of Pediatrics, Franciscus Gasthuis and Vlietland, Rotterdam, The Netherlands
| | - Birgit C P Koch
- Department of Hospital Pharmacy, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Irwin K M Reiss
- Department of Pediatrics, Division of Neonatology, Erasmus MC Sophia Children's Hospital, Erasmus University Medical Center Rotterdam, Doctor Molenwaterplein 40, 3015 CN, Rotterdam, The Netherlands
| | - Anouk E Muller
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Medical Microbiology, Haaglanden Medical Center, The Hague, The Netherlands
| | - René F Kornelisse
- Department of Pediatrics, Division of Neonatology, Erasmus MC Sophia Children's Hospital, Erasmus University Medical Center Rotterdam, Doctor Molenwaterplein 40, 3015 CN, Rotterdam, The Netherlands
| | - Karel Allegaert
- Department of Hospital Pharmacy, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Development and Regeneration, KU Leuven, Leuven, Belgium.,Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
6
|
An Assessment of Occasional Bio-Inequivalence for BCS1 and BCS3 Drugs: What are the Underlying Reasons? J Pharm Sci 2021; 111:124-134. [PMID: 34363838 DOI: 10.1016/j.xphs.2021.08.001] [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: 06/04/2021] [Revised: 08/02/2021] [Accepted: 08/02/2021] [Indexed: 11/20/2022]
Abstract
Despite having adequate solubility properties, bioequivalence (BE) studies performed on immediate release formulations containing BCS1/3 drugs occasionally fail. By systematically evaluating a set of 17 soluble drugs where unexpected BE failures have been reported and comparing to a set of 29 drugs where no such reports have been documented, a broad assessment of the risk factors leading to BE failure was performed. BE failures for BCS1/3 drugs were predominantly related to changes in Cmax rather than AUC. Cmax changes were typically modest, with minimal clinical significance for most drugs. Overall, drugs with a sharp plasma peak were identified as a key factor in BE failure risk. A new pharmacokinetic term (t½Cmax) is proposed to identify drugs at higher risk due to their peak plasma profile shape. In addition, the analysis revealed that weak acids, and drugs with particularly high gastric solubility are potentially more vulnerable to BE failure, particularly when these features are combined with a sharp Cmax peak. BCS3 drugs, which are often characterised as being more vulnerable to BE failure due to their potential for permeation and transit to be altered, particularly by excipient change, were not in general at greater risk of BE failures. These findings will help to inform how biowaivers may be optimally applied in the future.
Collapse
|
7
|
Wu YJ, Meanwell NA. Geminal Diheteroatomic Motifs: Some Applications of Acetals, Ketals, and Their Sulfur and Nitrogen Homologues in Medicinal Chemistry and Drug Design. J Med Chem 2021; 64:9786-9874. [PMID: 34213340 DOI: 10.1021/acs.jmedchem.1c00790] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Acetals and ketals and their nitrogen and sulfur homologues are often considered to be unconventional and potentially problematic scaffolding elements or pharmacophores for the design of orally bioavailable drugs. This opinion is largely a function of the perception that such motifs might be chemically unstable under the acidic conditions of the stomach and upper gastrointestinal tract. However, even simple acetals and ketals, including acyclic molecules, can be sufficiently robust under acidic conditions to be fashioned into orally bioavailable drugs, and these structural elements are embedded in many effective therapeutic agents. The chemical stability of molecules incorporating geminal diheteroatomic motifs can be modulated by physicochemical design principles that include the judicious deployment of proximal electron-withdrawing substituents and conformational restriction. In this Perspective, we exemplify geminal diheteroatomic motifs that have been utilized in the discovery of orally bioavailable drugs or drug candidates against the backdrop of understanding their potential for chemical lability.
Collapse
Affiliation(s)
- Yong-Jin Wu
- Small Molecule Drug Discovery, Bristol Myers Squibb Research and Early Development, 100 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Nicholas A Meanwell
- Department of Discovery and Chemistry and Molecular Technologies, Bristol-Myers Squibb PRI, PO Box 4000, Princeton, New Jersey 08543-4000, United States
| |
Collapse
|
8
|
Abbott IJ, Roberts JA, Meletiadis J, Peleg AY. Antimicrobial pharmacokinetics and preclinical in vitro models to support optimized treatment approaches for uncomplicated lower urinary tract infections. Expert Rev Anti Infect Ther 2020; 19:271-295. [PMID: 32820686 DOI: 10.1080/14787210.2020.1813567] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Urinary tract infections (UTIs) are extremely common. Millions of people, particularly healthy women, are affected worldwide every year. One-in-two women will have a recurrence within 12-months of an initial UTI. Inadequate treatment risks worsening infection leading to acute pyelonephritis, bacteremia and sepsis. In an era of increasing antimicrobial resistance, it is critical to provide optimized antimicrobial treatment. AREAS COVERED Literature was searched using PubMed and Google Scholar (up to 06/2020), examining the etiology, diagnosis and oral antimicrobial therapy for uncomplicated UTIs, with emphasis on urinary antimicrobial pharmacokinetics (PK) and the application of dynamic in vitro models for the pharmacodynamic (PD) profiling of pathogen response. EXPERT OPINION The majority of antimicrobial agents included in international guidelines were developed decades ago without well-described dose-response relationships. Microbiology laboratories still apply standard diagnostic methodology that has essentially remained unchanged for decades. Furthermore, it is uncertain how relevant standard in vitro susceptibility is for predicting antimicrobial efficacy in urine. In order to optimize UTI treatments, clinicians must exploit the urine-specific PK of antimicrobial agents. Dynamic in vitro models are valuable tools to examine the PK/PD and urodynamic variables associated with UTIs, while informing uropathogen susceptibility reporting, optimized dosing schedules, clinical trials and treatment guidelines.
Collapse
Affiliation(s)
- Iain J Abbott
- Department of Infectious Diseases, the Alfred Hospital and Central Clinical School, Monash University, Melbourne, Australia
| | - Jason A Roberts
- University of Queensland Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Australia.,School of Pharmacy, Centre for Translational Anti-infective Pharmacodynamics, The University of Queensland, Brisbane, Australia.,Department of Intensive Care Medicine, Royal Brisbane and Women's Hospital, Brisbane, Australia.,Division of Anaesthesiology Critical Care Emergency and Pain Medicine, Nîmes University Hospital, University of Montpellier, Nîmes, France
| | - Joseph Meletiadis
- Clinical Microbiology Laboratory, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, Haidari, Greece
| | - Anton Y Peleg
- Department of Infectious Diseases, the Alfred Hospital and Central Clinical School, Monash University, Melbourne, Australia.,Infection and Immunity Program, Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, Australia
| |
Collapse
|
9
|
Wu F, Zhou Y, Li L, Shen X, Chen G, Wang X, Liang X, Tan M, Huang Z. Computational Approaches in Preclinical Studies on Drug Discovery and Development. Front Chem 2020; 8:726. [PMID: 33062633 PMCID: PMC7517894 DOI: 10.3389/fchem.2020.00726] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 07/14/2020] [Indexed: 12/11/2022] Open
Abstract
Because undesirable pharmacokinetics and toxicity are significant reasons for the failure of drug development in the costly late stage, it has been widely recognized that drug ADMET properties should be considered as early as possible to reduce failure rates in the clinical phase of drug discovery. Concurrently, drug recalls have become increasingly common in recent years, prompting pharmaceutical companies to increase attention toward the safety evaluation of preclinical drugs. In vitro and in vivo drug evaluation techniques are currently more mature in preclinical applications, but these technologies are costly. In recent years, with the rapid development of computer science, in silico technology has been widely used to evaluate the relevant properties of drugs in the preclinical stage and has produced many software programs and in silico models, further promoting the study of ADMET in vitro. In this review, we first introduce the two ADMET prediction categories (molecular modeling and data modeling). Then, we perform a systematic classification and description of the databases and software commonly used for ADMET prediction. We focus on some widely studied ADMT properties as well as PBPK simulation, and we list some applications that are related to the prediction categories and web tools. Finally, we discuss challenges and limitations in the preclinical area and propose some suggestions and prospects for the future.
Collapse
Affiliation(s)
- Fengxu Wu
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Research Platform Service Management Center, Dongguan, China
- Key Laboratory of Pesticide & Chemical Biology, Ministry of Education, College of Chemistry, Central China Normal University, Wuhan, China
| | - Yuquan Zhou
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Research Platform Service Management Center, Dongguan, China
- The Second School of Clinical Medicine, Guangdong Medical University, Dongguan, China
| | - Langhui Li
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Research Platform Service Management Center, Dongguan, China
- Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Xianhuan Shen
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Research Platform Service Management Center, Dongguan, China
- Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Ganying Chen
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Research Platform Service Management Center, Dongguan, China
- The Second School of Clinical Medicine, Guangdong Medical University, Dongguan, China
| | - Xiaoqing Wang
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Research Platform Service Management Center, Dongguan, China
- Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Xianyang Liang
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Research Platform Service Management Center, Dongguan, China
- The Second School of Clinical Medicine, Guangdong Medical University, Dongguan, China
| | - Mengyuan Tan
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Research Platform Service Management Center, Dongguan, China
- Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Zunnan Huang
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Research Platform Service Management Center, Dongguan, China
- Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, China
- Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
| |
Collapse
|
10
|
Huttner A, Bielicki J, Clements MN, Frimodt-Møller N, Muller AE, Paccaud JP, Mouton JW. Oral amoxicillin and amoxicillin-clavulanic acid: properties, indications and usage. Clin Microbiol Infect 2019; 26:871-879. [PMID: 31811919 DOI: 10.1016/j.cmi.2019.11.028] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 11/22/2019] [Accepted: 11/25/2019] [Indexed: 10/25/2022]
Abstract
BACKGROUND Amoxicillin has been in use since the 1970s; it is the most widely used penicillin both alone and in combination with the β-lactamase clavulanic acid. OBJECTIVES In this narrative review, we re-examine the properties of oral amoxicillin and clavulanic acid and provide guidance on their use, with emphasis on the preferred use of amoxicillin alone. SOURCES Published medical literature (MEDLINE database via Pubmed). CONTENT While amoxicillin and clavulanic acid have similar half-lives, clavulanic acid is more protein bound and even less heat stable than amoxicillin, with primarily hepatic metabolism. It is also more strongly associated with gastrointestinal side effects, including Clostridium difficile infection, and, thus, in oral combination formulations, limits the maximum daily dose of amoxicillin that can be given. The first ratio for an amoxicillin-clavulanic acid combination was set at 4:1 due to clavulanic acid's high affinity for β-lactamases; ratios of 2:1, 7:1, 14:1 and 16:1 are currently available in various regions. Comparative effectiveness data for the different ratios are scarce. Amoxicillin-clavulanic acid is often used as empiric therapy for many of the World Health Organization's Priority Infectious Syndromes in adults and children, leading to extensive consumption, when some of these syndromes could be handled with a delayed antibiotic prescription approach or amoxicillin alone. IMPLICATIONS Using available epidemiological and pharmacokinetic data, we provide guidance on indications for amoxicillin versus amoxicillin-clavulanic acid and on optimal oral administration, including choice of combination ratio. More data are needed, particularly on heat stability, pharmacodynamic effects and emergence of resistance in 'real-world' clinical settings.
Collapse
Affiliation(s)
- A Huttner
- Division of Infectious Diseases, Geneva University Hospitals, Geneva, Switzerland.
| | - J Bielicki
- University of Basel Children's Hospital, Paediatric Infectious Diseases, Basel, Switzerland; Paediatric Infectious Diseases Research Group, St. George's University of London, London, UK
| | - M N Clements
- MRC Clinical Trials Unit at UCL, UCL, London, UK
| | - N Frimodt-Møller
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| | - A E Muller
- Department of Medical Microbiology, Haaglanden Medical Centre, The Hague, the Netherlands; Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - J-P Paccaud
- Global Antibiotic Research and Development Partnership, Geneva, Switzerland
| | - J W Mouton
- Department of Medical Microbiology, Haaglanden Medical Centre, The Hague, the Netherlands
| |
Collapse
|
11
|
de Kraker MEA, Sommer H, de Velde F, Gravestock I, Weiss E, McAleenan A, Nikolakopoulos S, Amit O, Ashton T, Beyersmann J, Held L, Lovering AM, MacGowan AP, Mouton JW, Timsit JF, Wilson D, Wolkewitz M, Bettiol E, Dane A, Harbarth S. Optimizing the Design and Analysis of Clinical Trials for Antibacterials Against Multidrug-resistant Organisms: A White Paper From COMBACTE's STAT-Net. Clin Infect Dis 2019; 67:1922-1931. [PMID: 30107400 PMCID: PMC6260160 DOI: 10.1093/cid/ciy516] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 06/15/2018] [Indexed: 01/08/2023] Open
Abstract
Innovations are urgently required for clinical development of antibacterials against multidrug-resistant organisms. Therefore, a European, public-private working group (STAT-Net; part of Combatting Bacterial Resistance in Europe [COMBACTE]), has reviewed and tested several innovative trials designs and analytical methods for randomized clinical trials, which has resulted in 8 recommendations. The first 3 focus on pharmacokinetic and pharmacodynamic modeling, emphasizing the pertinence of population-based pharmacokinetic models, regulatory procedures for the reassessment of old antibiotics, and rigorous quality improvement. Recommendations 4 and 5 address the need for more sensitive primary end points through the use of rank-based or time-dependent composite end points. Recommendation 6 relates to the applicability of hierarchical nested-trial designs, and the last 2 recommendations propose the incorporation of historical or concomitant trial data through Bayesian methods and/or platform trials. Although not all of these recommendations are directly applicable, they provide a solid, evidence-based approach to develop new, and established, antibacterials and address this public health challenge.
Collapse
Affiliation(s)
- Marlieke E A de Kraker
- Infection Control Program, Geneva University Hospitals and Faculty of Medicine, Switzerland
| | - Harriet Sommer
- Institute for Medical Biometry and Statistics, Faculty of Medicine and Medical Center, University of Freiburg, Germany
| | - Femke de Velde
- Department of Medical Microbiology and Infectious Diseases, Rotterdam, The Netherlands.,Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Isaac Gravestock
- Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Switzerland
| | - Emmanuel Weiss
- Université Paris Diderot, Paris, France.,APHP Anesthesiology and Critical Care Department, Beaujon Hospital, Paris, France
| | - Alexandra McAleenan
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, United Kingdom
| | - Stavros Nikolakopoulos
- Department of Biostatistics and Research Support, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, The Netherlands
| | - Ohad Amit
- GlaxoSmithKline, Collegeville, Pennsylvania
| | | | | | - Leonhard Held
- Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Switzerland
| | - Andrew M Lovering
- Bristol Centre for Antibiotic Research and Evaluation, Infection Sciences, North Bristol NHS Trust, Southmead Hospital, United Kingdom
| | - Alasdair P MacGowan
- Bristol Centre for Antibiotic Research and Evaluation, Infection Sciences, North Bristol NHS Trust, Southmead Hospital, United Kingdom
| | - Johan W Mouton
- Department of Medical Microbiology and Infectious Diseases, Rotterdam, The Netherlands
| | - Jean-François Timsit
- UMR 1137 IAME Inserm/Université Paris Diderot.,APHP Medical and Infectious Diseases ICU, Bichat Hospital, Paris, France
| | | | - Martin Wolkewitz
- Institute for Medical Biometry and Statistics, Faculty of Medicine and Medical Center, University of Freiburg, Germany
| | - Esther Bettiol
- Infection Control Program, Geneva University Hospitals and Faculty of Medicine, Switzerland
| | - Aaron Dane
- DaneStat Consulting Limited, Macclesfield, United Kingdom
| | - Stephan Harbarth
- Infection Control Program, Geneva University Hospitals and Faculty of Medicine, Switzerland
| | | |
Collapse
|
12
|
Monogue ML, Nicolau DP. Pharmacokinetics-pharmacodynamics of β-lactamase inhibitors: are we missing the target? Expert Rev Anti Infect Ther 2019; 17:571-582. [PMID: 31340665 DOI: 10.1080/14787210.2019.1647781] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Introduction: β-lactamase production in Gram-negative bacteria is a leading cause of antimicrobial resistance. β-lactamase inhibitors are therapeutic agents used in combination with a partner antimicrobial to overcome the production of these enzymes and restore antimicrobial activity. To address the ongoing threat of multi-drug resistant bacteria, a recent wave of β-lactamase inhibitor development has occurred. Emphasis on the pharmacokinetics and pharmacodynamics of these agents is needed to optimize their clinical impact. Areas covered: This review will describe methods currently used to define the pharmacokinetics/pharmacodynamics of β-lactamase inhibitors. Minimal focus will be on the structure and mechanism of β-lactamase inhibitors. Emphasis will be placed on the use of specific thresholds to normalize β-lactamase inhibitor exposure. In vitro and in vivo pharmacokinetic/pharmacodynamic data specific to FDA approved and pipeline β-lactamase inhibitors will be explored. Expert opinion: Describing the exposure-response relationship of β-lactamase inhibitors is an ongoing challenge due to the dynamic relationship of the β-lactamase inhibitor with the active partner compound. Pharmacokinetic/pharmacodynamic indices and target exposures lack generalizability, as they are often specific to the infecting organism and/or β-lactamase, rather than β-lactamase inhibitor class. Selected dosage regimens of new agents should be validated via the use of population target attainment analyses.
Collapse
Affiliation(s)
- Marguerite L Monogue
- a Center for Anti-infective Research and Development, Hartford Hospital , Hartford , CT , USA.,b Department of Pharmacy, University of Texas Southwestern , Dallas , TX , USA
| | - David P Nicolau
- a Center for Anti-infective Research and Development, Hartford Hospital , Hartford , CT , USA.,c Division of Infectious Diseases, Hartford Hospital , Hartford , CT , USA
| |
Collapse
|
13
|
Yang F, Yang F, Wang G, Xi W, Zhang C, Wang H. Pharmacokinetics of the amoxicillin-clavulanic acid combination after intravenous and oral administration in cats. J Vet Pharmacol Ther 2019; 42:511-517. [PMID: 31162674 DOI: 10.1111/jvp.12765] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/12/2019] [Accepted: 03/12/2019] [Indexed: 11/30/2022]
Abstract
The pharmacokinetic properties of amoxicillin (AMX) and clavulanic acid (CLV) were studied in healthy cats following single intravenous and oral dosage of 10 mg/kg of AMX and 2.5 mg/kg of CLV. The drug concentrations in plasma were determined by a high-performance liquid chromatographic-tandem mass spectrometry (LC-MS-MS) method validated for canine plasma and further subjected to noncompartmental analysis. After intravenous injection, no significant difference (p > 0.05) was found in the volume of distribution of these two compounds. In addition, AMX and CLV were both rapidly eliminated from plasma with a clearance of 0.453 and 0.921 L hr-1 kg-1 , respectively; however, a quicker elimination was observed for CLV (p < 0.01). After oral administration, both drugs were characterized by rapid absorption with an absorption half-life of 1.10 and 0.70 hr for AMX and CLV, respectively. Significant differences were observed between their absorption rates (p < 0.05). However, the oral bioavailabilities of AMX and CLV (75.57% and 98.15%, respectively) were not statistically different (p > 0.05). A total intravenous or oral dose at 12.5 mg/kg of AMX and CLV (4:1) is predicted to be effective for treating those bacterial species isolated from cats with a minimum inhibitory concentration (MIC) of ≤0.25 μg/ml for 12 hr, based on a time above the MIC (T > MIC) of 40%.
Collapse
Affiliation(s)
- Fan Yang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China.,Environmental and Animal Products Safety Laboratory of Key Discipline in University of Henan Province, Henan University of Science and Technology, Luoyang, China
| | - Fang Yang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Guoyong Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Wenyuan Xi
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Chaoshuo Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Han Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
14
|
Quantitative analysis, pharmacokinetics and metabolomics study for the comprehensive characterization of the salt-processing mechanism of Psoraleae Fructus. Sci Rep 2019; 9:661. [PMID: 30679561 PMCID: PMC6345873 DOI: 10.1038/s41598-018-36908-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 11/27/2018] [Indexed: 01/11/2023] Open
Abstract
Research based on quantitative analysis, pharmacokinetics and metabolomics was conducted to explore the effects of salt-processing on Psoraleae Fructus (PF). Quantitative analysis showed that the contents of bioactive components were higher in salt-processed Psoraleae Fructus (SPF) extract than in PF extract. Pharmacokinetics indicated that the overall AUC and tmax levels was higher, while Cmax was lower in the SPF group. In the metabolomics study, the differential influences of PF and SPF on 22 common biomarkers and associated metabolic pathways showed that salt-processing could enhance the effect of PF and reduce toxicity in the cardiovascular and renal systems. The internal correlations among these results, together with the influence of salt-processing, suggested that the effects of heating and newly generated surfactants during the salt-processing procedure were the primary causes of the changes in chemical composition and absorption characteristics, as well as the subsequent enhanced efficacy and minor toxicity.
Collapse
|