1
|
Ishak A, Mazonakis N, Spernovasilis N, Akinosoglou K, Tsioutis C. Bactericidal versus bacteriostatic antibacterials: clinical significance, differences and synergistic potential in clinical practice. J Antimicrob Chemother 2024:dkae380. [PMID: 39471409 DOI: 10.1093/jac/dkae380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2024] Open
Abstract
Antibacterial activity can be classified as either bactericidal or bacteriostatic, using methods such as the MBC/MIC ratio and time-kill curves. However, such categorization has proven challenging in clinical practice, as these definitions only apply under specific laboratory conditions, which may differ from clinical settings. Several factors, such as the specific bacteria or infectious medium, can affect the action of antibiotics, with many antibacterials exerting both activities. These definitions have also led to the belief that bactericidal antibacterials are superior to bacteriostatic, especially in more severe cases, such as endocarditis, neutropenia and bacteraemia. Additionally, current dogma dictates against the combination of bactericidal and bacteriostatic antibacterials in clinical practice, due to potential antagonism. This review aimed to assess the differences in antibacterial activity of bactericidal and bacteriostatic antibacterials based on in vitro and in vivo studies and examine their antagonistic or synergistic effects. Our findings show that specific bacteriostatic agents, such as linezolid and tigecycline, are clinically non-inferior to bactericidals in multiple infections, including pneumonia, intra-abdominal infections, and skin and soft tissue infections. Studies also support using several bacteriostatic agents as salvage therapies in severe infections, such as neutropenic fever and endocarditis. Additionally, not all combinations of bacteriostatic and bactericidal agents appear to be antagonistic, with many combinations, such as linezolid and rifampicin, already being used. The findings should be interpreted with caution, as most evidence is from observational studies and there is a need for randomized controlled trials to assess their effectiveness and combinations, especially within the context of rising antimicrobial resistance.
Collapse
Affiliation(s)
- Angela Ishak
- Department of Internal Medicine, 48202 Henry Ford Hospital, Detroit, MI, USA
| | - Nikolaos Mazonakis
- Department of Internal Medicine, Thoracic Diseases General Hospital Sotiria, 11527 Athens, Greece
| | - Nikolaos Spernovasilis
- Department of Infectious Diseases, German Oncology Centre, 4108 Limassol, Cyprus
- School of Medicine, University of Crete, 71500 Heraklion, Greece
| | - Karolina Akinosoglou
- School of Medicine, University of Patras, 26504 Rio, Greece
- Department of Internal Medicine and Infectious Diseases, University General Hospital of Patras, 26504 Rio, Greece
| | - Constantinos Tsioutis
- School of Medicine, European University Cyprus, 6 Diogenes str, Nicosia 2404, Cyprus
| |
Collapse
|
2
|
Pracher L, Zeitlinger M. Preclinical and clinical studies in the drug development process of European Medicines Agency-approved non-HIV antiviral agents: a narrative review. Clin Microbiol Infect 2024:S1198-743X(24)00480-4. [PMID: 39389465 DOI: 10.1016/j.cmi.2024.10.001] [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: 06/14/2024] [Revised: 09/17/2024] [Accepted: 10/02/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Viral diseases represent a substantial global health challenge, necessitating the urgent development of effective antiviral medications. OBJECTIVES This review aims to present a thorough examination of systemic antiviral drugs approved by the European Medicines Agency (EMA) since its founding, excluding those targeting HIV, with a focus on preclinical and clinical studies in the drug development process. SOURCES Data was extracted from the European Public Assessment Reports and Summary of Product Characteristics issued by the EMA. CONTENT In total, 21 currently approved agents were analysed with a focus on preclinical and clinical studies. The majority of substances have been approved for hepatitis C (38%) and B (19%) followed by influenza and SARS-CoV-2 (14% and 10%, respectively). A smaller subset obtained approval for the indications of hepatitis D, cytomegalovirus, and pox viruses. As for preclinical studies, heterogeneity in the methods used for efficacy studies was observed, which is at least partly explained by the diverse nature of viruses and their hosts and the lack of general guidelines for antiviral pharmacokinetics and pharmacodynamics studies by the EMA. Clinical studies varied in sample sizes, ranging from a few hundred to several thousand patients. Many antiviral agents have a high potential for cytochrome P450 (CYP) and other enzyme interactions, resulting in the need for a high number of drug-drug interaction studies. Special market authorizations are available, including conditional approval for urgently required drugs such as nirmatrelvir/ritonavir for the treatment of COVID-19, and authorization under exceptional circumstances when comprehensive data cannot be provided, as seen with tecovirimat for pox viruses. IMPLICATIONS Streamlining the drug development process of antiviral substances and providing more guidelines would be crucial given the ongoing demand for effective treatment options for existing and new viral diseases.
Collapse
Affiliation(s)
- Lena Pracher
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
3
|
Dyer CJ, De Waele JJ, Roberts JA. Antibiotic dose optimisation in the critically ill: targets, evidence and future strategies. Curr Opin Crit Care 2024; 30:439-447. [PMID: 39150038 DOI: 10.1097/mcc.0000000000001187] [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: 08/17/2024]
Abstract
PURPOSE OF REVIEW To highlight the recent evidence for antibiotic pharmacokinetics and pharmacodynamics (PK/PD) in enhancing patient outcomes in sepsis and septic shock. We also summarise the limitations of available data and describe future directions for research to support translation of antibiotic dose optimisation to the clinical setting. RECENT FINDINGS Sepsis and septic shock are associated with poor outcomes and require antibiotic dose optimisation, mostly due to significantly altered pharmacokinetics. Many studies, including some randomised controlled trials have been conducted to measure the clinical outcome effects of antibiotic dose optimisation interventions including use of therapeutic drug monitoring. Current data support antibiotic dose optimisation for the critically ill. Further investigation is required to evolve more timely and robust precision antibiotic dose optimisation approaches, and to clearly quantify whether any clinical and health-economic benefits support expanded use of this treatment intervention. SUMMARY Antibiotic dose optimisation appears to improve outcomes in critically ill patients with sepsis and septic shock, however further research is required to quantify the level of benefit and develop a stronger knowledge of the role of new technologies to facilitate optimised dosing.
Collapse
Affiliation(s)
- Christopher J Dyer
- Herston Institute of Infectious Diseases (HeIDI), Metro North Health
- Pharmacy Department
- Departments of Pharmacy and Intensive Care Medicine, Royal Brisbane and Women's Hospital (RBWH), Herston, Australia
| | - Jan J De Waele
- Department of Critical Care Medicine, Ghent University Hospital
- Dept of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Jason A Roberts
- Herston Institute of Infectious Diseases (HeIDI), Metro North Health
- Pharmacy Department
- Departments of Pharmacy and Intensive Care Medicine, Royal Brisbane and Women's Hospital (RBWH), Herston, Australia
- UQ Centre for Clinical Research (UQCCR), Faculty of Medicine, University of Queensland, Herston, Australia
| |
Collapse
|
4
|
Romano JE, Lo CP, Villadoniga GB, Mays T. Pharmacokinetics of ceftiofur crystalline-free acid in plasma and seminal plasma in beef bulls. Theriogenology 2024; 226:328-334. [PMID: 38959843 DOI: 10.1016/j.theriogenology.2024.06.027] [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: 03/01/2024] [Revised: 05/31/2024] [Accepted: 06/26/2024] [Indexed: 07/05/2024]
Abstract
The objective of this study was to compare the plasma (PL) and seminal plasma (SP) pharmacokinetic profile of ceftiofur (CEFT) and desuroylceftiofur acetamide (DFCA) after administration of CEFT crystalline-free acid (CCFA) by SC route in two sites of the ear in beef bulls. Four clinically healthy Hereford bulls received a comprehensive physical exam and subsequently a breeding-soundness examination, CBC, and chemistry profile panel. All bulls were diagnosed healthy and satisfactory potential breeders. In one group (n = 2), a single dose of CCFA was administered SC route at the base of the ear (BOE) at a dose of 6.6 mg/kg of body weight. The second group (n = 2) was also administered by SC route in the middle third of the posterior aspect of the ear (MTE). The concentrations of CEFT and DFCA in PL and SP were determined by a high-performance liquid chromatography mass spectrometry (HPLC-MS). Blood and semen samples were collected before the administration of CCFA and at 12, 24, 36, 48, 72, 96, 120, 144, and 168 h after injection. No levels of CEFT were detected in PL and only in 20 of the 40 SP samples (P = 0.0001). The mean level of CEFT in SP was 0.11 % in comparison with the DFCA level. DFCA was found in all PL and SP samples. Therefore, DFCA was chosen to be utilized in the study of the pharmacokinetics parameters both in PL and SP. There were no differences in the mean PL levels of DFCA for the two sites of SC administration between the BOE (102.9 ± 78.9 ng/mL; X ± SD) and to MTE (116.1 ± 70.2 ng/mL; P = 0.58). The mean SP levels of DFCA after administration in the BOE was 857 ± 747 ng/mL, and for the MTE was 549 ± 488 ng/mL without differences between both sites (P = 0.15). The mean level of DFCA in PL was 109.5 ± 74.0 ng/mL, which was lower than the mean SP levels of 695 ± 103 ng/mL (P = 0.001). Moreover, the PL peak DFCA concentration (Cmax) was 229 ± 46 ng/mL at 36.0 ± 29.4 h (Tmax) post-administration. The SP Cmax was 1851 ± 533 ng/mL at 30.0 ± 28.6 h (Tmax) post-administration. The Cmax between PL and SP were distinctive (P = 0.004) without any differences in Tmax between PL and SP (P = 0.60). The terminal half-life for PL DFCA (47.4 ± 29.3 h) was not different than in SP (53.1 ± 23.6 h; P = 0.77). The PL area under the curve concentration time from the first to the last sample (AUC0-last) was 18,984 ± 4841 ng/mL/h, which was significatively smaller compared with 125,677 ± 59,445 ng/mL/h for SP AUC0-last (P = 0.04). The PL mean residence time from the first to the last sample (MRT0-last) was 69.7 ± 15.1 h, and it was similar than for SP of 66.5 ± 7.7 h (P = 0.69). From the present investigation, based in its pharmacokinetic features, it was concluded that CCFA should be an appropriate antibiotic that could be used for the treatment of bull genital infections when its indication is properly outlined. To study the pharmacokinetics of CCFA in SP, DFCA metabolite was appropriated.
Collapse
Affiliation(s)
- Juan E Romano
- 3 R Ranch Livestock Co, Somerville, TX, 778793, USA.
| | - Chih-Ping Lo
- Texas A&M Veterinary Medical Diagnostic Laboratory, College Station, TX, 77843, USA
| | | | - Travis Mays
- Texas A&M Veterinary Medical Diagnostic Laboratory, College Station, TX, 77843, USA
| |
Collapse
|
5
|
Bhavnani SM, Hammel JP, Lakota EA, Liolios K, Trang M, Rubino CM, Steenbergen JN, Friedrich L, Tzanis E, Ambrose PG. Assessment of pharmacokinetics-pharmacodynamics to support omadacycline dosing regimens for the treatment of patients with acute bacterial skin and skin structure infections. Antimicrob Agents Chemother 2024; 68:e0128123. [PMID: 39082883 PMCID: PMC11373226 DOI: 10.1128/aac.01281-23] [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: 10/19/2023] [Accepted: 02/08/2024] [Indexed: 09/05/2024] Open
Abstract
Pharmacokinetic-pharmacodynamic (PK-PD) relationships for efficacy were evaluated using data from omadacycline-treated patients with acute bacterial skin and skin structure infections (ABSSSI) enrolled in two phase 3 studies. Patients received omadacycline 100 mg intravenously (IV) every 12 hours for two doses, followed by 100 mg IV every 24 hours (q24h), with the option to switch to 300 mg oral (PO) q24h after 3 days or 450 mg PO q24h for two doses, followed by 300 mg PO q24h for a total duration of 7-14 days. Clinical response was evaluated at 48-72 hours [early clinical response (ECR)], end of treatment (EOT), and 7-14 days after EOT. Using a population pharmacokinetic (PK) model and PK data from patients with Staphylococcus aureus at baseline, omadacycline free-drug plasma area under the concentration-time curve (AUC) values were determined, and the relationships between free-drug plasma AUC:MIC ratio and dichotomous efficacy endpoints were evaluated. Using these relationships, the population PK model, simulation, and an omadacycline MIC distribution for S. aureus, mean percent probabilities of response were evaluated. Statistically significant PK--PD relationships were identified for ECR (P = 0.016 and 0.013 for optimized two- and three-group free-drug plasma AUC:MIC ratios, respectively). At an MIC value of 0.5 µg/mL, percent probabilities of model-predicted success for ECR based on the univariable PK-PD relationships using continuous and two-group free-drug plasma AUC:MIC ratio variables were 91.9 and 95.6%, respectively, for the IV-to-PO dosing regimen and 89.3 and 88.4%, respectively, for the PO-only dosing regimen. These data support for omadacycline IV-to-PO and PO-only dosing regimens for ABSSSI and an omadacycline susceptibility breakpoint of 0.5 µg/mL for S. aureus.
Collapse
Affiliation(s)
- Sujata M Bhavnani
- Institute for Clinical Pharmacodynamics, Inc., Schenectady, New York, USA
| | - Jeffrey P Hammel
- Institute for Clinical Pharmacodynamics, Inc., Schenectady, New York, USA
| | - Elizabeth A Lakota
- Institute for Clinical Pharmacodynamics, Inc., Schenectady, New York, USA
| | - Kathryn Liolios
- Institute for Clinical Pharmacodynamics, Inc., Schenectady, New York, USA
| | - Michael Trang
- Institute for Clinical Pharmacodynamics, Inc., Schenectady, New York, USA
| | | | | | | | - Evan Tzanis
- Paratek Pharmaceuticals, Inc., King of Prussia, Pennsylvania, USA
| | - Paul G Ambrose
- Institute for Clinical Pharmacodynamics, Inc., Schenectady, New York, USA
| |
Collapse
|
6
|
Gras-Martín L, Plaza-Diaz A, Zarate-Tamames B, Vera-Artazcoz P, Torres OH, Bastida C, Soy D, Ruiz-Ramos J. Risk Factors Associated with Antibiotic Exposure Variability in Critically Ill Patients: A Systematic Review. Antibiotics (Basel) 2024; 13:801. [PMID: 39334976 PMCID: PMC11428266 DOI: 10.3390/antibiotics13090801] [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: 07/19/2024] [Revised: 08/15/2024] [Accepted: 08/22/2024] [Indexed: 09/30/2024] Open
Abstract
(1) Background: Knowledge about the behavior of antibiotics in critically ill patients has been increasing in recent years. Some studies have concluded that a high percentage may be outside the therapeutic range. The most likely cause of this is the pharmacokinetic variability of critically ill patients, but it is not clear which factors have the greatest impact. The aim of this systematic review is to identify risk factors among critically ill patients that may exhibit significant pharmacokinetic alterations, compromising treatment efficacy and safety. (2) Methods: The search included the PubMed, Web of Science, and Embase databases. (3) Results: We identified 246 observational studies and ten clinical trials. The most studied risk factors in the literature were renal function, weight, age, sex, and renal replacement therapy. Risk factors with the greatest impact included renal function, weight, renal replacement therapy, age, protein or albumin levels, and APACHE or SAPS scores. (4) Conclusions: The review allows us to identify which critically ill patients are at a higher risk of not reaching therapeutic targets and helps us to recognize the extensive number of risk factors that have been studied, guiding their inclusion in future studies. It is essential to continue researching, especially in real clinical practice and with clinical outcomes.
Collapse
Affiliation(s)
- Laura Gras-Martín
- Pharmacy Department, Hospital de la Santa Creu i Sant Pau, Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Sat Quintí 77-79, 08041 Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Adrián Plaza-Diaz
- Pharmacy Department, Hospital de la Santa Creu i Sant Pau, Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Sat Quintí 77-79, 08041 Barcelona, Spain
| | - Borja Zarate-Tamames
- Pharmacy Department, Hospital de la Santa Creu i Sant Pau, Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Sat Quintí 77-79, 08041 Barcelona, Spain
| | - Paula Vera-Artazcoz
- Institut de Recerca Sant Pau (IR SANT PAU), Sat Quintí 77-79, 08041 Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Intensive Care Department, Hospital de la Santa Creu i Sant Pau, Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
| | - Olga H Torres
- Institut de Recerca Sant Pau (IR SANT PAU), Sat Quintí 77-79, 08041 Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Geriatric Unit, Internal Medicine Department, Hospital de la Santa Creu i Sant Pau, Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
| | - Carla Bastida
- Pharmacy Department, Division of Medicines, Hospital Clinic of Barcelona, Villarroel 170, 08036 Barcelona, Spain
- Department of Pharmacology, Toxicology and Therapeutical Chemistry, Faculty of Pharmacy, Universitat de Barcelona, Campus Diagonal, Av. de Joan XXIII, 27-31, 08028 Barcelona, Spain
| | - Dolors Soy
- Pharmacy Department, Division of Medicines, Hospital Clinic of Barcelona, Villarroel 170, 08036 Barcelona, Spain
- Department of Pharmacology, Toxicology and Therapeutical Chemistry, Faculty of Pharmacy, Universitat de Barcelona, Campus Diagonal, Av. de Joan XXIII, 27-31, 08028 Barcelona, Spain
| | - Jesús Ruiz-Ramos
- Pharmacy Department, Hospital de la Santa Creu i Sant Pau, Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Sat Quintí 77-79, 08041 Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| |
Collapse
|
7
|
Berryhill BA, Gil-Gil T, Witzany C, Goldberg DA, Vega NM, Regoes RR, Levin BR. The dynamics of Staphylococcal infection and their treatment with antibiotics and bacteriophage in the Galleria mellonella model system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.23.609294. [PMID: 39229007 PMCID: PMC11370618 DOI: 10.1101/2024.08.23.609294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Critical to our understanding of infections and their treatment is the role the innate immune system plays in controlling bacterial pathogens. Nevertheless, many in vivo systems are made or modified such that they do not have an innate immune response. Use of these systems denies the opportunity to examine the synergy between the immune system and antimicrobial agents. In this study we demonstrate that the larva of Galleria mellonella is an effective in vivo model for the study of the population and evolutionary biology of bacterial infections and their treatment. To do this we test three hypotheses concerning the role of the innate immune system during infection. We show: i) sufficiently high densities of bacteria are capable of saturating the innate immune system, ii) bacteriostatic drugs and bacteriophages are as effective as bactericidal antibiotics in preventing mortality and controlling bacterial densities, and iii) minority populations of bacteria resistant to a treating antibiotic will not ascend. Using a highly virulent strain of Staphylococcus aureus and a mathematical computer-simulation model, we further explore how the dynamics of the infection within the short term determine the ultimate infection outcome. We find that excess immune activation in response to high densities of bacteria leads to a strong but short-lived immune response which ultimately results in a high degree of mortality. Overall, our findings illustrate the utility of the G. mellonella model system in conjunction with established in vivo models in studying infectious disease progression and treatment.
Collapse
Affiliation(s)
- Brandon A. Berryhill
- Department of Biology, Emory University; Atlanta, Georgia, 30322, USA
- Program in Microbiology and Molecular Genetics, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University; Atlanta, GA, 30322, USA
| | - Teresa Gil-Gil
- Department of Biology, Emory University; Atlanta, Georgia, 30322, USA
| | | | - David A. Goldberg
- Department of Biology, Emory University; Atlanta, Georgia, 30322, USA
| | - Nic M. Vega
- Department of Biology, Emory University; Atlanta, Georgia, 30322, USA
| | - Roland R. Regoes
- Institute of Integrative Biology, ETH Zurich, Zurich, Switzerland
| | - Bruce R. Levin
- Department of Biology, Emory University; Atlanta, Georgia, 30322, USA
| |
Collapse
|
8
|
Minichmayr IK, Dreesen E, Centanni M, Wang Z, Hoffert Y, Friberg LE, Wicha SG. Model-informed precision dosing: State of the art and future perspectives. Adv Drug Deliv Rev 2024; 215:115421. [PMID: 39159868 DOI: 10.1016/j.addr.2024.115421] [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: 06/18/2024] [Revised: 07/19/2024] [Accepted: 08/01/2024] [Indexed: 08/21/2024]
Abstract
Model-informed precision dosing (MIPD) stands as a significant development in personalized medicine to tailor drug dosing to individual patient characteristics. MIPD moves beyond traditional therapeutic drug monitoring (TDM) by integrating mathematical predictions of dosing, and considering patient-specific factors (patient characteristics, drug measurements) as well as different sources of variability. For this purpose, rigorous model qualification is required for the application of MIPD in patients. This review delves into new methods in model selection and validation, also highlighting the role of machine learning in improving MIPD, the utilization of biosensors for real-time monitoring, as well as the potential of models integrating biomarkers for efficacy or toxicity for precision dosing. The clinical evidence of TDM and MIPD is discussed for various medical fields including infection medicine, oncology, transplant medicine, and inflammatory bowel diseases, thereby underscoring the role of pharmacokinetics/pharmacodynamics and specific biomarkers. Further research, particularly randomized clinical trials, is warranted to corroborate the value of MIPD in enhancing patient outcomes and advancing personalized medicine.
Collapse
Affiliation(s)
- I K Minichmayr
- Dept. of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - E Dreesen
- Clinical Pharmacology and Pharmacotherapy Unit, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - M Centanni
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Z Wang
- Clinical Pharmacology and Pharmacotherapy Unit, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Y Hoffert
- Clinical Pharmacology and Pharmacotherapy Unit, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - L E Friberg
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - S G Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Hamburg, Germany.
| |
Collapse
|
9
|
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
|
10
|
Deshpande D, Magombedze G, Boorgula GD, Chapagain M, Srivastava S, Gumbo T. Ceftriaxone Efficacy for Mycobacterium avium Complex Lung Disease in the Hollow Fiber and Translation to Sustained Sputum Culture Conversion in Patients. J Infect Dis 2024; 230:e230-e240. [PMID: 38036299 PMCID: PMC11326821 DOI: 10.1093/infdis/jiad545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/17/2023] [Accepted: 11/28/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND Only 35.6%-50.8% of patients with Mycobacterium avium complex (MAC) pulmonary disease achieve sustained sputum culture conversion (SSCC) on treatment with the azithromycin-ethambutol-rifabutin standard of care (SOC). We tested the efficacy of ceftriaxone, a β-lactam with a lung-to-serum penetration ratio of 12.18-fold. METHODS We mimicked lung concentration-time profiles of 7 ceftriaxone once-daily doses for 28 days in the hollow fiber system model of intracellular MAC (HFS-MAC). Monte Carlo experiments were used for dose selection. We also compared once-daily ceftriaxone monotherapy to 3-drug SOC against 5 MAC clinical isolates in HFS-MAC using γ (kill) slopes, and translated to SSCC rates. RESULTS Ceftriaxone killed 1.02-3.82 log10 colony-forming units (CFU)/mL, at optimal dose of 2 g once-daily. Ceftriaxone killed all 5 strains below day 0 versus 2 of 5 for SOC. The median γ (95% confidence interval [CI]) was 0.49 (.47-.52) log10 CFU/mL/day for ceftriaxone and 0.38 (.34-.43) log10 CFU/mL/day for SOC. In patients, the SOC was predicted to achieve SSCC rates (CI) of 39.3% (36%-42%) at 6 months. The SOC SSCC was 50% at 8.18 (3.64-27.66) months versus 3.58 (2.20-7.23) months for ceftriaxone, shortening time to SSCC 2.35-fold. CONCLUSIONS Ceftriaxone is a promising agent for creation of short-course chemotherapy.
Collapse
Affiliation(s)
| | | | | | - Moti Chapagain
- Department of Cellular and Molecular Biology, School of Medicine, University of Texas Health Science Center at Tyler
| | - Shashikant Srivastava
- Baylor University Medical Center, Dallas
- Department of Medicine, School of Medicine, University of Texas at Tyler
- Department of Cellular and Molecular Biology, School of Medicine, University of Texas Health Science Center at Tyler
| | - Tawanda Gumbo
- Mathematical Modeling and AI Department, Praedicare Inc, Dallas
- Hollow Fiber System and Experimental Therapeutics Laboratories, Praedicare Inc, Dallas, Texas
| |
Collapse
|
11
|
Stoddard M, Yuan L, Cooper J, Carcillo Neumar C, Hibner B, Gardner H, Chakravarty A. Can we do better with Mylotarg? Model-based assessment of opportunities to improve therapeutic index. Toxicol Appl Pharmacol 2024; 490:117034. [PMID: 39009139 DOI: 10.1016/j.taap.2024.117034] [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: 03/08/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/17/2024]
Abstract
Late-stage clinical trial failures increase the overall cost and risk of bringing new drugs to market. Determining the pharmacokinetic (PK) drivers of toxicity and efficacy in preclinical studies and early clinical trials supports quantitative optimization of drug schedule and dose through computational modeling. Additionally, this approach permits prioritization of lead candidates with better PK properties early in development. Mylotarg is an antibody-drug conjugate (ADC) that attained U.S. Food and Drug Administration (FDA) approval under a fractionated dosing schedule after 17 years of clinical trials, including a 10-year period on the market resulting in hundreds of fatal adverse events. Although ADCs are often considered lower risk for toxicity due to their targeted nature, off-target activity and liberated payload can still constrain dosing and drive clinical failure. Under its original schedule, Mylotarg was dosed infrequently at high levels, which is typical for ADCs because of their long half-lives. However, our PK modeling suggests that these regimens increase maximum plasma concentration (Cmax)-related toxicities while producing suboptimal exposures to the target receptor. Our analysis demonstrates that the benefits of dose fractionation for Mylotarg tolerability should have been obvious early in the drug's clinical development and could have curtailed the proliferation of ineffective Phase III studies. We also identify schedules likely to be even more efficacious without compromising on tolerability. Alternatively, a longer-circulating Mylotarg formulation could obviate the need for dose fractionation, allowing superior patient convenience. Early-stage PK optimization through quantitative modeling methods can accelerate clinical development and prevent late-stage failures.
Collapse
Affiliation(s)
| | - Lin Yuan
- Fractal Therapeutics, Lexington, MA, USA
| | | | | | | | | | | |
Collapse
|
12
|
Song X, Zeng M, Yang T, Han M, Yan S. Individualized, dynamic, and full-course vancomycin dosing prediction: a study on the customized dose model. Front Pharmacol 2024; 15:1414347. [PMID: 39021838 PMCID: PMC11252542 DOI: 10.3389/fphar.2024.1414347] [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: 04/11/2024] [Accepted: 06/06/2024] [Indexed: 07/20/2024] Open
Abstract
Purpose The single-point trough-based therapeutic drug monitoring (TDM) and Bayesian forecasting approaches are still limited in individualized and dynamic vancomycin delivery. Until recently, there has not yet been enough focus on the direct integration of pharmacokinetic/pharmacodynamic (PK/PD) and TDM to construct a customized dose model (CDM) for vancomycin to achieve individualized, dynamic, and full-course dose prediction from empirical to follow-up treatment. This study sought to establish CDM for vancomycin, test its performance and superiority in clinical efficacy prediction, formulate a CDM-driven full-course dosage prediction strategy to overcome the above challenge, and predict the empirical vancomycin dosages for six Staphylococci populations and four strains in patients with various creatinine clearance rates (CLcr). Methods The PK/PD and concentration models derived from our earlier research were used to establish CDM. The receiver operating characteristic (ROC) curve, with the area under ROC curve (AUCR) as the primary endpoint, for 21 retrospective cases was applied to test the performance and superiority of CDM in clinical efficacy prediction by comparison to the current frequently-used dose model (FDM). A model with an AUCR of at least 0.8 was considered acceptable. Based on the availability of TDM, the strategy of CDM-driven individualized, dynamic, and full-course dose prediction for vancomycin therapy was formulated. Based on the CDM, Monte Carlo simulation was used to predict the empirical vancomycin dosages for the target populations and bacteria. Results Four CDMs and the strategy of CDM-driven individualized, dynamic, and full-course dose prediction for vancomycin therapy from empirical to follow-up treatment were constructed. Compared with FDM, CDM showed a greater AUCR value (0.807 vs. 0.688) in clinical efficacy prediction. The empirical vancomycin dosages for six Staphylococci populations and four strains in patients with various CLcr were predicted. Conclusion CDM is a competitive individualized dose model. It compensates for the drawbacks of the existing TDM technology and Bayesian forecasting and offers a straightforward and useful supplemental approach for individualized and dynamic vancomycin delivery. Through mathematical modeling of the vancomycin dosage, this study achieved the goal of predicting doses individually, dynamically, and throughout, thus promoting "mathematical knowledge transfer and application" and also providing reference for quantitative and personalized research on similar drugs.
Collapse
Affiliation(s)
- Xiangqing Song
- Department of Pharmacy, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Meizi Zeng
- Department of Pharmacy, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Tao Yang
- Department of Pharmacy, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Mi Han
- Department of Pharmacy, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Shipeng Yan
- Office of Cancer Prevention Research, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
13
|
Xu N, Shi Y, Wang Y, Mak W, Yang W, Ng KW, Wu Y, Tang Z, He Q, Yan G, Xiang X, Zhu X. Development and Quality Control of a Population Pharmacokinetic Model Library for Caspofungin. Pharmaceutics 2024; 16:819. [PMID: 38931940 PMCID: PMC11207296 DOI: 10.3390/pharmaceutics16060819] [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: 05/06/2024] [Revised: 05/30/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Caspofungin is an echinocandin antifungal agent commonly used as the first-line therapy for invasive candidiasis, salvage therapy for invasive aspergillosis, and empirical therapy for presumed fungal infections. Pharmacokinetic (PK) variabilities and suboptimal exposure have been reported for caspofungin, increasing the risk of insufficient efficacy. OBJECTIVE This work aimed to develop a caspofungin population pharmacokinetic (popPK) library and demonstrate its utility by assessing the probability of target attainment across diverse settings. METHODS We established a caspofungin popPK model library following a rigorous literature review, re-implementing selected models in R with rxode2. Quality control procedures included a comparison of different studies and assessing covariate impacts. Model libraries were primarily used to perform Monte Carlo simulations to estimate target attainment and guide personalized dosing in Candida infections. RESULTS A total of 13 models, one- or two-compartment models, were included. The most significant covariates were body size (weight and body surface area), liver function, and albumin level. The results show that children and adults showed considerable differences in pharmacokinetics. For C. albicans and C. parapsilosis, none of the populations achieved a PTA of ≥90% at their respective susceptible MIC values. In contrast, for C. glabrata, 70% of the adult studies reached a PTA of ≥90%, while all pediatric studies achieved the same PTA level. CONCLUSION At the recommended dosage, adult patients showed notably lower exposure to caspofungin compared to pediatric patients. Considering body size, liver function, and serum albumin is crucial when determining caspofungin dosage regimens. Furthermore, further research is required to comprehensively understand the pharmacokinetics of caspofungin in pediatric patients.
Collapse
Affiliation(s)
- Nuo Xu
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai 201203, China; (N.X.); (Y.S.); (W.M.); (W.Y.); (Z.T.); (Q.H.)
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha 410000, China
| | - Yufei Shi
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai 201203, China; (N.X.); (Y.S.); (W.M.); (W.Y.); (Z.T.); (Q.H.)
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha 410000, China
| | - Yixue Wang
- Department of Critical Care Medicine, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai 200000, China; (Y.W.); (G.Y.)
| | - Wenyao Mak
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai 201203, China; (N.X.); (Y.S.); (W.M.); (W.Y.); (Z.T.); (Q.H.)
| | - Wenyu Yang
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai 201203, China; (N.X.); (Y.S.); (W.M.); (W.Y.); (Z.T.); (Q.H.)
| | - Kar Weng Ng
- Department of Pharmacotherapy Services, Hospital Kuala Lumpur, Ministry of Health, Kuala Lumpur 50586, Malaysia;
| | - Yue Wu
- Department of Clinical Pharmacy, Shenzhen Children’s Hospital, Medical College, Shantou University, Shenzhen 518000, China;
| | - Zhijia Tang
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai 201203, China; (N.X.); (Y.S.); (W.M.); (W.Y.); (Z.T.); (Q.H.)
| | - Qingfeng He
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai 201203, China; (N.X.); (Y.S.); (W.M.); (W.Y.); (Z.T.); (Q.H.)
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha 410000, China
| | - Gangfeng Yan
- Department of Critical Care Medicine, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai 200000, China; (Y.W.); (G.Y.)
| | - Xiaoqiang Xiang
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai 201203, China; (N.X.); (Y.S.); (W.M.); (W.Y.); (Z.T.); (Q.H.)
| | - Xiao Zhu
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai 201203, China; (N.X.); (Y.S.); (W.M.); (W.Y.); (Z.T.); (Q.H.)
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha 410000, China
| |
Collapse
|
14
|
Bradley JS, Bulitta JB, Cook R, Yu PA, Iwamoto C, Hesse EM, Chaney D, Yu Y, Kennedy JL, Sue D, Karchmer AW, Bower WA, Hendricks K. Central Nervous System Antimicrobial Exposure and Proposed Dosing for Anthrax Meningitis. Clin Infect Dis 2024; 78:1451-1457. [PMID: 38412060 PMCID: PMC11175673 DOI: 10.1093/cid/ciae093] [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] [Revised: 01/31/2024] [Accepted: 02/16/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND The high mortality of systemic anthrax is likely a consequence of the severe central nervous system inflammation that occurs in anthrax meningitis. Effective treatment of such infections requires, at a minimum, adequate cerebrospinal fluid (CSF) antimicrobial concentrations. METHODS We reviewed English medical literature and regulatory documents to extract information on serum and CSF exposures for antimicrobials with in vitro activity against Bacillus anthracis. Using CSF pharmacokinetic exposures and in vitro B. anthracis susceptibility data, we used population pharmacokinetic modeling and Monte Carlo simulations to determine whether a specific antimicrobial dosage would likely achieve effective CSF antimicrobial activity in patients with normal to inflamed meninges (ie, an intact to markedly disrupted blood-brain barrier). RESULTS The probability of microbiologic success at achievable antimicrobial dosages was high (≥95%) for ciprofloxacin, levofloxacin (500 mg every 12 hours), meropenem, imipenem/cilastatin, penicillin G, ampicillin, ampicillin/sulbactam, doxycycline, and minocycline; acceptable (90%-95%) for piperacillin/tazobactam and levofloxacin (750 mg every 24 hours); and low (<90%) for vancomycin, amikacin, clindamycin, and linezolid. CONCLUSIONS Prompt empiric antimicrobial therapy of patients with suspected or confirmed anthrax meningitis may reduce the high morbidity and mortality. Our data support using several β-lactam-, fluoroquinolone-, and tetracycline-class antimicrobials as first-line and alternative agents for treatment of patients with anthrax meningitis; all should achieve effective microbiologic exposures. Our data suggest antimicrobials that should not be relied on to treat suspected or documented anthrax meningitis. Furthermore, the protein synthesis inhibitors clindamycin and linezolid can decrease toxin production and may be useful components of combination therapy.
Collapse
Affiliation(s)
- John S Bradley
- Department of Pediatrics, University of California–San Diego School of Medicine and Rady Children's Hospital, San Diego, California, USA
| | - Jürgen B Bulitta
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Rachel Cook
- Oak Ridge Institute for Science and Education, CDC Fellowship Program, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Patricia A Yu
- Division of Preparedness and Emerging Infections, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Chelsea Iwamoto
- Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Elisabeth M Hesse
- Division of Preparedness and Emerging Infections, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Danielle Chaney
- Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Yon Yu
- Division of Preparedness and Emerging Infections, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Jordan L Kennedy
- Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - David Sue
- Division of Preparedness and Emerging Infections, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Adolf W Karchmer
- Division of Infectious Disease, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - William A Bower
- Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Katherine Hendricks
- Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
15
|
Ferran AA, Roques BB, Chapuis L, Kuroda T, Lacroix MZ, Toutain PL, Bousquet-Melou A, Lallemand EA. Predicted efficacy and tolerance of different dosage regimens of benzylpenicillin in horses based on a pharmacokinetic study with three IM formulations and one IV formulation. Front Vet Sci 2024; 11:1409266. [PMID: 38881781 PMCID: PMC11176610 DOI: 10.3389/fvets.2024.1409266] [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: 03/29/2024] [Accepted: 05/10/2024] [Indexed: 06/18/2024] Open
Abstract
Introduction Benzylpenicillin (BP) is a first-line antibiotic in horses but there are discrepancies between manufacturers and literature recommendations regarding dosing regimen. Objectives of this study were to evaluate pharmacokinetics and local tolerance of four different formulations of BP in adult horses, and to suggest optimized dosing regimen according to the formulation. Methods A cross-over design was used in 3 phases for the intramuscular injection of three different products: procaine BP alone, procaine BP/ benzathine BP combination or penethamate hydriodide were administered IM in the gluteal muscles of 6 horses for 3 days. Single IV administration of sodium BP was performed to the same horses with a dose of 22,000 IU BP/kg bwt 39 weeks after last IM injection. BP plasma concentrations were determined by UPLC assay coupled with mass spectrometry and a PK/PD analysis was conducted to predict the efficacy of various dosing regimens by estimating values of the fT>MIC index for different minimum inhibitory concentrations (MIC). Tolerance at the site of IM injection was monitored by creatine kinase activity quantified with a validated chemistry system and clinical scorings. Results and discussion Except one neurological reaction following one administration of penethamate hydriodide, the tolerance was good. Procaine BP alone, procaine BP/benzathine BP combination or penethamate hydriodide intramuscular administrations at a dosage of 22,000 IU BP/kg bwt q24h for 5 days would yield plasma concentrations that should be effective against bacteria with MIC of ≤0.256, 0.125 or 0.064 mg/L respectively. Of all the tested treatments, the use of a sodium BP by IV Constant Rate Infusion (CRI) for 10 hours a day was deemed to be the most efficient. All the formulations tested in this study are adequate to treat infections with susceptible Streptococcus equi.
Collapse
Affiliation(s)
- Aude A Ferran
- INTHERES, Université de Toulouse, INRAE, ENVT, Toulouse, France
| | | | - Laura Chapuis
- INTHERES, Université de Toulouse, INRAE, ENVT, Toulouse, France
| | - Taisuke Kuroda
- Clinical Veterinary Medicine Division, Equine Research Institute, Japan Racing Association, Shimotsuke, Japan
| | | | - Pierre-Louis Toutain
- INTHERES, Université de Toulouse, INRAE, ENVT, Toulouse, France
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, London, United Kingdom
| | | | | |
Collapse
|
16
|
KURODA T, MINAMIJIMA Y, NIWA H, MITA H, TAMURA N, FUKUDA K, TOUTAIN PL, OHTA M. Pharmacokinetics/pharmacodynamics cut-off determination for fosfomycin using Monte Carlo simulation in healthy horses. J Vet Med Sci 2024; 86:413-420. [PMID: 38346727 PMCID: PMC11061577 DOI: 10.1292/jvms.23-0476] [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: 11/24/2023] [Accepted: 01/26/2024] [Indexed: 04/12/2024] Open
Abstract
Fosfomycin (FOM) is an approved veterinary medicinal product for large animals in Japan, but Clinical breakpoint (CBP) for antimicrobial susceptibility test (AST) is not defined for animals. This study aimed at conducting a pharmacokinetics/pharmacodynamics (PK/PD) analysis to determine the PK/PD cutoff for the CBP in horses. Drug concentrations following single intravenous administration (IV) of 20 mg/kg body weight (BW) FOM in nine horses were measured using liquid chromatography/mass spectrometry. The data were modelled using a nonlinear mixed-effects model, followed by Monte Carlo simulations. A 90% probability of target attainment for a PK/PD target of the ratio of Area Under the free plasma concentration-time curve divided by the minimal inhibitory concentration (MIC) >24 hr was set as PK/PD cut-off. The PK/PD cutoff for FOM 20 mg/kg BW q12 hr IV was estimated with the MIC value of ≤16.0 mg/L, and this regimen was considered effective against E. coli (MIC90; 16.0 mg/L) in healthy horses based on the MIC90 values of the wild population. Owing to the relevance of FOM to human health, veterinarians should use q 12 hr FOM 20 mg /kg against E. coli infections with an MIC <16 µg/mL, as suggested by our PK/PD cutoff after AST.
Collapse
Affiliation(s)
- Taisuke KURODA
- Clinical Veterinary Medicine Division, Equine Research
Institute, Japan Racing Association, Tochigi, Japan
| | - Yohei MINAMIJIMA
- Drug Analysis Department, Laboratory of Racing Chemistry,
Tochigi, Japan
| | - Hidekazu NIWA
- Microbiology Division, Equine Research Institute, Japan
Racing Association, Tochigi, Japan
| | - Hiroshi MITA
- Clinical Veterinary Medicine Division, Equine Research
Institute, Japan Racing Association, Tochigi, Japan
| | - Norihisa TAMURA
- Clinical Veterinary Medicine Division, Equine Research
Institute, Japan Racing Association, Tochigi, Japan
| | - Kentaro FUKUDA
- Clinical Veterinary Medicine Division, Equine Research
Institute, Japan Racing Association, Tochigi, Japan
| | - Pierre-Louis TOUTAIN
- Comparative Biomedical Sciences, The Royal Veterinary
College, London, United Kingdom
- Intheres, Ecole Nationale Vétérinaire de Toulouse,
France
| | - Minoru OHTA
- Clinical Veterinary Medicine Division, Equine Research
Institute, Japan Racing Association, Tochigi, Japan
| |
Collapse
|
17
|
Jiao Y, Yan J, Sutaria DS, Lu P, Vicchiarelli M, Reyna Z, Ruiz-Delgado J, Burk E, Moon E, Shah NR, Spellberg B, Bonomo RA, Drusano GL, Louie A, Luna BM, Bulitta JB. Population pharmacokinetics and humanized dosage regimens matching the peak, area, trough, and range of amikacin plasma concentrations in immune-competent murine bloodstream and lung infection models. Antimicrob Agents Chemother 2024; 68:e0139423. [PMID: 38289076 PMCID: PMC10916399 DOI: 10.1128/aac.01394-23] [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: 10/26/2023] [Accepted: 12/22/2023] [Indexed: 03/07/2024] Open
Abstract
Amikacin is an FDA-approved aminoglycoside antibiotic that is commonly used. However, validated dosage regimens that achieve clinically relevant exposure profiles in mice are lacking. We aimed to design and validate humanized dosage regimens for amikacin in immune-competent murine bloodstream and lung infection models of Acinetobacter baumannii. Plasma and lung epithelial lining fluid (ELF) concentrations after single subcutaneous doses of 1.37, 13.7, and 137 mg/kg of body weight were simultaneously modeled via population pharmacokinetics. Then, humanized amikacin dosage regimens in mice were designed and prospectively validated to match the peak, area, trough, and range of plasma concentration profiles in critically ill patients (clinical dose: 25-30 mg/kg of body weight). The pharmacokinetics of amikacin were linear, with a clearance of 9.93 mL/h in both infection models after a single dose. However, the volume of distribution differed between models, resulting in an elimination half-life of 48 min for the bloodstream and 36 min for the lung model. The drug exposure in ELF was 72.7% compared to that in plasma. After multiple q6h dosing, clearance decreased by ~80% from the first (7.35 mL/h) to the last two dosing intervals (~1.50 mL/h) in the bloodstream model. Likewise, clearance decreased by 41% from 7.44 to 4.39 mL/h in the lung model. The humanized dosage regimens were 117 mg/kg of body weight/day in mice [administered in four fractions 6 h apart (q6h): 61.9%, 18.6%, 11.3%, and 8.21% of total dose] for the bloodstream and 96.7 mg/kg of body weight/day (given q6h as 65.1%, 16.9%, 10.5%, and 7.41%) for the lung model. These validated humanized dosage regimens and population pharmacokinetic models support translational studies with clinically relevant amikacin exposure profiles.
Collapse
Affiliation(s)
- Yuanyuan Jiao
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Jun Yan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Dhruvitkumar S. Sutaria
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Peggy Lu
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Michael Vicchiarelli
- Institute for Therapeutic Innovation, College of Medicine, University of Florida, Orlando, Florida, USA
| | - Zeferino Reyna
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Juan Ruiz-Delgado
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Elizabeth Burk
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Eugene Moon
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Nirav R. Shah
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Brad Spellberg
- Los Angeles County-USC (LAC+USC) Medical Center, Los Angeles, California, USA
| | - Robert A. Bonomo
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Proteomics and Bioinformatics, Case Western Reserve University, Cleveland, Ohio, USA
- Louis Stokes Cleveland Department of Veterans Affairs, Cleveland, Ohio, USA
- Case VA Center for Antimicrobial Resistance and Epidemiology (Case VA CARES), Cleveland, Ohio, USA
| | - George L. Drusano
- Institute for Therapeutic Innovation, College of Medicine, University of Florida, Orlando, Florida, USA
| | - Arnold Louie
- Institute for Therapeutic Innovation, College of Medicine, University of Florida, Orlando, Florida, USA
| | - Brian M. Luna
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Jürgen B. Bulitta
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Orlando, Florida, USA
| |
Collapse
|
18
|
Straub A, Stapf M, Brands RC, Kübler AC, Lâm TT, Vollmer A, Gubik S, Scherf-Clavel O, Hartmann S. Investigation of clindamycin concentrations in human plasma and jawbone tissue in patients with osteonecrosis of the jaw: A prospective trial. J Craniomaxillofac Surg 2024; 52:355-362. [PMID: 38368214 DOI: 10.1016/j.jcms.2024.01.014] [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: 08/10/2023] [Revised: 10/18/2023] [Accepted: 01/16/2024] [Indexed: 02/19/2024] Open
Abstract
The aim of this study was to investigate the jawbone concentration of clindamycin (CLI) in patients with an osteonecrosis of the jaw (ONJ). Patients with medication-related ONJ (MRONJ) and osteoradionecrosis (ORN) with an antibiotic treatment with CLI were included. Plasma, vital and necrotic bone samples were collected. Plasma and jawbone samples were analyzed by liquid chromatography-tandem mass spectrometry. Patients with MRONJ exhibited a mean plasma CLI concentration of 9.6 μg/mL (SD ± 3.6 μg/mL) and mean concentrations of 2.3 μg/g CLI (SD ± 1.4 μg/g) and 2.1 μg/g CLI (SD ± 2.4 μg/g) in vital and necrotic bone samples, without statistical significance (p = 0.79). In patients with ORN, mean concentration in plasma was 12.0 μg/mL (SD ± 2.6 μg/mL), in vital bone 2.1 μg/g (SD ± 1.5 μg/g), and in necrotic bone 1.7 μg/g (SD ± 1.2 μg/g). Vital and necrotic bone concentrations did not differ significantly (p = 0.88). The results demonstrate that CLI concentrations are considerably lower than in plasma, but sufficient for most bacteria present in ONJ. Within the limitations of the study, it seems that CLI is a relevant alternative to other antibiotics in the treatment of ONJ because it reaches adequate concentrations in jawbone.
Collapse
Affiliation(s)
- Anton Straub
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Würzburg, Pleicherwall 2, 97070, Würzburg, Germany.
| | - Maximilian Stapf
- Institute for Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Roman C Brands
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Würzburg, Pleicherwall 2, 97070, Würzburg, Germany
| | - Alexander C Kübler
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Würzburg, Pleicherwall 2, 97070, Würzburg, Germany
| | - Thiên-Trí Lâm
- Institute for Hygiene and Microbiology, University of Würzburg, Josef-Schneider-Str. 2/E1, 97080, Würzburg, Germany
| | - Andreas Vollmer
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Würzburg, Pleicherwall 2, 97070, Würzburg, Germany
| | - Sebastian Gubik
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Würzburg, Pleicherwall 2, 97070, Würzburg, Germany
| | - Oliver Scherf-Clavel
- Institute for Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074, Würzburg, Germany; Aalen University, Faculty of Chemistry, Beethovenstraße 1, 73430, Aalen, Germany
| | - Stefan Hartmann
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Würzburg, Pleicherwall 2, 97070, Würzburg, Germany
| |
Collapse
|
19
|
Nakashima H, Miyazaki M, Kuwamura T, Oda K, Haga Y, Imakyure O. Relationship between Target Time above Minimum Inhibitory Concentration Achievement Rate of Meropenem Using Monte Carlo Simulation and In-Hospital Survival in Patients with Pseudomonas aeruginosa Bacteremia. Antibiotics (Basel) 2024; 13:219. [PMID: 38534654 DOI: 10.3390/antibiotics13030219] [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: 01/24/2024] [Revised: 02/16/2024] [Accepted: 02/26/2024] [Indexed: 03/28/2024] Open
Abstract
Pseudomonas aeruginosa bacteremia is associated with a high mortality rate, and meropenem (MEPM) is commonly used to treat it. However, the relationship between the time above the minimum inhibitory concentration (fT>MIC) of MEPM and its therapeutic efficacy in P. aeruginosa bacteremia has not been explored. This study aimed to investigate this relationship by defining the target % fT>MIC of MEPM as 75%. The retrospective study spanned 14 years and included hospitalized patients treated with MEPM for P. aeruginosa bacteremia. Monte Carlo simulation was used to calculate the probability of target attainment (PTA) for each patient, and the threshold for a PTA of 75% fT>MIC associated with in-hospital survival was determined using receiver operating characteristic (ROC) curves. The ROC curve-derived PTA associated with improved in-hospital survival was 65.0%, a significant finding in multivariate logistic regression analysis adjusted for patient background factors (odds ratio: 20.49, 95% confidence interval: 3.02-245.23, p = 0.005). This result suggests a dosing regimen that achieves a PTA of at least 65% when the target fT>MIC of MEPM for treating P. aeruginosa bacteremia is defined as 75%.
Collapse
Affiliation(s)
- Hajime Nakashima
- Department of Pharmacy, Japan Community Health Care Organization Kyushu Hospital, Fukuoka 806-0034, Japan
| | - Motoyasu Miyazaki
- Department of Pharmacy, Fukuoka University Chikushi Hospital, Fukuoka 818-8502, Japan
| | - Tsuneo Kuwamura
- Department of Pharmacy, Japan Community Health Care Organization Kurume General Hospital, Fukuoka 830-0013, Japan
| | - Kazutaka Oda
- Department of Pharmacy, Kumamoto University Hospital, Kumamoto 860-8556, Japan
| | - Yumi Haga
- Department of Clinical Laboratory, Japan Community Health Care Organization Kyushu Hospital, Fukuoka 806-0034, Japan
| | - Osamu Imakyure
- Department of Pharmacy, Fukuoka University Chikushi Hospital, Fukuoka 818-8502, Japan
| |
Collapse
|
20
|
Rox K, Medina E. Aerosolized delivery of ESKAPE pathogens for murine pneumonia models. Sci Rep 2024; 14:2558. [PMID: 38297183 PMCID: PMC10830452 DOI: 10.1038/s41598-024-52958-9] [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: 06/25/2023] [Accepted: 01/25/2024] [Indexed: 02/02/2024] Open
Abstract
Murine pneumonia models for ESKAPE pathogens serve to evaluate novel antibacterials or to investigate immunological responses. The majority of published models uses intranasal or to a limited extent the intratracheal instillation to challenge animals. In this study, we propose the aerosol delivery of pathogens using a nebulizer. Aerosol delivery typically results in homogeneous distribution of the inoculum in the lungs because of lower particle size. This is of particular importance when compounds are assessed for their pharmacokinetic and pharmacodynamic (PK/PD) relationships as it allows to conduct several analysis with the same sample material. Moreover, aerosol delivery has the advantage that it mimics the 'natural route' of respiratory infection. In this short and concise study, we show that aerosol delivery of pathogens resulted in a sustained bacterial burden in the neutropenic lung infection model for five pathogens tested, whereas it gave a similar result in immunocompetent mice for three out of five pathogens. Moreover, a substantial bacterial burden in the lungs was already achieved 2 h post inhalation. Hence, this study constitutes a viable alternative for intranasal administration and a refinement of murine pneumonia models for PK/PD assessments of novel antibacterial compounds allowing to study multiple readouts with the same sample material.
Collapse
Affiliation(s)
- Katharina Rox
- Department of Chemical Biology, Helmholtz Centre for Infection Research (HZI), Inhoffenstraße 7, 38124, Braunschweig, Germany.
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 38124, Braunschweig, Germany.
| | - Eva Medina
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 38124, Braunschweig, Germany
- Infection Immunology Group, Helmholtz Centre for Infection Research (HZI), Inhoffenstraße 7, 38124, Braunschweig, Germany
| |
Collapse
|
21
|
Bissantz C, Zampaloni C, David-Pierson P, Dieppois G, Guenther A, Trauner A, Winther L, Stubbings W. Translational PK/PD for the Development of Novel Antibiotics-A Drug Developer's Perspective. Antibiotics (Basel) 2024; 13:72. [PMID: 38247631 PMCID: PMC10812724 DOI: 10.3390/antibiotics13010072] [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: 11/30/2023] [Revised: 12/23/2023] [Accepted: 12/28/2023] [Indexed: 01/23/2024] Open
Abstract
Antibiotic development traditionally involved large Phase 3 programs, preceded by Phase 2 studies. Recognizing the high unmet medical need for new antibiotics and, in some cases, challenges to conducting large clinical trials, regulators created a streamlined clinical development pathway in which a lean clinical efficacy dataset is complemented by nonclinical data as supportive evidence of efficacy. In this context, translational Pharmacokinetic/Pharmacodynamic (PK/PD) plays a key role and is a major contributor to a "robust" nonclinical package. The classical PK/PD index approach, proven successful for established classes of antibiotics, is at the core of recent antibiotic approvals and the current antibacterial PK/PD guidelines by regulators. Nevertheless, in the case of novel antibiotics with a novel Mechanism of Action (MoA), there is no prior experience with the PK/PD index approach as the basis for translating nonclinical efficacy to clinical outcome, and additional nonclinical studies and PK/PD analyses might be considered to increase confidence. In this review, we discuss the value and limitations of the classical PK/PD approach and present potential risk mitigation activities, including the introduction of a semi-mechanism-based PK/PD modeling approach. We propose a general nonclinical PK/PD package from which drug developers might choose the studies most relevant for each individual candidate in order to build up a "robust" nonclinical PK/PD understanding.
Collapse
Affiliation(s)
- Caterina Bissantz
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Claudia Zampaloni
- Roche Pharma Research and Early Development, Cardiovascular, Metabolism, Immunology, Infectious Diseases and Ophthalmology (CMI2O), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Pascale David-Pierson
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Guennaelle Dieppois
- Roche Pharma Research and Early Development, Cardiovascular, Metabolism, Immunology, Infectious Diseases and Ophthalmology (CMI2O), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Andreas Guenther
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Andrej Trauner
- Roche Pharma Research and Early Development, Cardiovascular, Metabolism, Immunology, Infectious Diseases and Ophthalmology (CMI2O), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Lotte Winther
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - William Stubbings
- Product Development, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| |
Collapse
|
22
|
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: 0] [Impact Index Per Article: 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
|
23
|
KURODA T, MINAMIJIMA Y, NIWA H, MITA H, TAMURA N, FUKUDA K, OHTA M. Pharmacokinetic/pharmacodynamic analysis of cephalothin after intramuscular administration in Thoroughbred horses. J Equine Sci 2023; 34:111-114. [PMID: 38274556 PMCID: PMC10806359 DOI: 10.1294/jes.34.111] [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: 06/21/2023] [Accepted: 08/02/2023] [Indexed: 01/27/2024] Open
Abstract
A pharmacokinetic/pharmacodynamic (PK/PD) approach was used to determine a dosage regimen of cephalothin (CET) after intramuscular (IM) administration in horses. CET plasma concentrations were measured in eight horses after a single IM administration of 11 mg/kg bwt of CET. The data were modeled using a nonlinear mixed-effect model, and the probability of target attainment (PTA) of the PK/PD target was calculated for 5,000 horses generated by Monte Carlo simulations. IM administrations of CET at 11 mg/kg bwt q 8 hr and q 6 hr achieved a PTA of 90% against the MIC90 of S. zooepidemicus and S. aureus, respectively, and were considered to be effective dosage regimens. The total dose for the IM administration recommended in this study was lower than that for intravenous (IV) administration in previous studies.
Collapse
Affiliation(s)
- Taisuke KURODA
- Equine Research Institute, Japan Racing
Association, Tochigi 329-0412, Japan
| | | | - Hidekazu NIWA
- Equine Research Institute, Japan Racing
Association, Tochigi 329-0412, Japan
| | - Hiroshi MITA
- Equine Research Institute, Japan Racing
Association, Tochigi 329-0412, Japan
| | - Norihisa TAMURA
- Equine Research Institute, Japan Racing
Association, Tochigi 329-0412, Japan
| | - Kentaro FUKUDA
- Equine Research Institute, Japan Racing
Association, Tochigi 329-0412, Japan
| | - Minoru OHTA
- Equine Research Institute, Japan Racing
Association, Tochigi 329-0412, Japan
| |
Collapse
|
24
|
Bradley JS, Makieieva N, Tøndel C, Roilides E, Kelly MS, Patel M, Vaddady P, Maniar A, Zhang Y, Paschke A, Chen LF. Pharmacokinetics, Safety, and Tolerability of Imipenem/Cilastatin/Relebactam in Children with Confirmed or Suspected Gram-Negative Bacterial Infections: A Phase 1b, Open-Label, Single-Dose Clinical Trial. J Clin Pharmacol 2023; 63:1387-1397. [PMID: 37562063 DOI: 10.1002/jcph.2334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 08/02/2023] [Indexed: 08/12/2023]
Abstract
Imipenem/cilastatin/relebactam is approved for the treatment of serious gram-negative bacterial infections in adults. This study assessed the pharmacokinetics (PK), safety, and tolerability of a single dose of imipenem/cilastatin/relebactam (with a fixed 2:1 ratio of imipenem/cilastatin to relebactam, and with a maximum dose of 15 mg/kg imipenem and 15 mg/kg cilastatin [≤500 mg imipenem and ≤500 mg cilastatin] and 7.5 mg/kg relebactam [≤250 mg relebactam]) in children with confirmed/suspected gram-negative bacterial infections receiving standard-of-care antibacterial therapy. In this phase 1, noncomparative study (ClinicalTrials.gov identifier, NCT03230916), PK parameters from 46 children were analyzed using both population modeling and noncompartmental analysis. The PK/pharmacodynamic (PD) target for imipenem was percent time of the dosing interval that unbound plasma concentration exceeded the minimum inhibitory concentration (%fT>MIC) of ≥30% (MIC = 2 mcg/mL). For relebactam, the PK/PD target was a free drug area under the plasma concentration-time curve (AUC) normalized to MIC (at 2 mcg/mL) of ≥8.0 (equivalent to an AUC from time zero extrapolated to infinity of ≥20.52 mcg·h/mL). Safety was assessed up to 14 days after drug infusion. For imipenem, the ranges for the geometric mean %fT>MIC and maximum concentration (Cmax ) across age cohorts were 56.5%-93.7% and 32.2-38.2 mcg/mL, respectively. For relebactam, the ranges of the geometric mean Cmax and AUC from 0 to 6 hours across age cohorts were 16.9-21.3 mcg/mL and 26.1-55.3 mcg·h/mL, respectively. In total, 8/46 (17%) children experienced ≥1 adverse events (AEs) and 2/46 (4%) children experienced nonserious AEs that were deemed drug related by the investigator. Imipenem and relebactam exceeded plasma PK/PD targets; single doses of imipenem/cilastatin/relebactam were well tolerated with no significant safety concerns identified. These results informed imipenem/cilastatin/relebactam dose selection for further pediatric clinical evaluation.
Collapse
Affiliation(s)
- John S Bradley
- Department of Pediatrics, University of California San Diego School of Medicine and Rady Children's Hospital of San Diego, San Diego, CA, USA
| | - Nataliia Makieieva
- Department of Pediatrics, Kharkiv National Medical University, Kharkiv, Ukraine
| | - Camilla Tøndel
- Department of Clinical Science, University of Bergen, and Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | - Emmanuel Roilides
- Third Department of Pediatrics, Infectious Diseases Unit, School of Medicine, Aristotle University and Hippokration General Hospital, Thessaloniki, Greece
| | - Matthew S Kelly
- Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | | | - Pavan Vaddady
- Merck & Co. Inc, Rahway, NJ, USA
- Daiichi Sankyo, Inc., Basking Ridge, NJ, USA
| | | | | | | | | |
Collapse
|
25
|
Tamma PD, Harris PNA, Mathers AJ, Wenzler E, Humphries RM. Breaking Down the Breakpoints: Rationale for the 2022 Clinical and Laboratory Standards Institute Revised Piperacillin-Tazobactam Breakpoints Against Enterobacterales. Clin Infect Dis 2023; 77:1585-1590. [PMID: 36001445 DOI: 10.1093/cid/ciac688] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/11/2022] [Accepted: 08/22/2022] [Indexed: 11/14/2022] Open
Abstract
Piperacillin-tazobactam (PTZ) is one of the most common antibiotics administered to hospitalized patients. Its broad activity against gram-negative, gram-positive, and anaerobic pathogens; efficacy in clinical trials across diverse infection types and patient populations; and generally favorable toxicity profile make it a particularly appealing antibiotic agent. PTZ susceptibility interpretive criteria (ie, breakpoints) for the Enterobacterales were initially established in 1992, as the drug was undergoing approval by the US Food and Drug Administration. In the ensuing 30 years, changes in the molecular epidemiology of the Enterobacterales and its impact on PTZ susceptibility testing, mounting pharmacokinetic/pharmacodynamic data generated from sophisticated techniques such as population pharmacokinetic modeling and Monte Carlo simulation, and disturbing safety signals in a large clinical trial prompted the Clinical Laboratory and Standards Institute (CLSI) to review available evidence to determine the need for revision of the PTZ breakpoints for Enterobacterales. After an extensive literature review and formal voting process, the susceptibility criteria were revised in the 2022 CLSI M100 document to the following: ≤8/4 µg/mL (susceptible), 16/4 µg/mL (susceptible dose-dependent), and ≥32/4 µg/mL (resistant). Herein, we provide a brief overview of the CLSI process of antibiotic breakpoint revisions and elaborate on the available data that ultimately led to the decision to revise the PTZ breakpoints.
Collapse
Affiliation(s)
- Pranita D Tamma
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Patrick N A Harris
- Faculty of Medicine, UQ Center for Clinical Research, Royal Brisbane and Women's Hospital Campus, University of Queensland, Brisbane, Australia
| | - Amy J Mathers
- Department of Medicine and Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - Eric Wenzler
- Department of Pharmacy Practice, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Romney M Humphries
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
26
|
Igarashi Y, Takemura W, Liu X, Kojima N, Morita T, Chuang VTG, Enoki Y, Taguchi K, Matsumoto K. In vivo Pharmacokinetic/Pharmacodynamic Analysis of the Efficacy of the Cefepime/Nacubactam Combination Against β-Lactamase-Producing Enterobacterales based on the Instantaneous MIC Concept. Pharm Res 2023; 40:2423-2431. [PMID: 37783926 DOI: 10.1007/s11095-023-03608-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 09/15/2023] [Indexed: 10/04/2023]
Abstract
PURPOSE Nacubactam (NAC) is a novel diazabicyclooctane β-lactamase inhibitor used in combination with cefepime (CFPM). In this study, we aimed to determine the target pharmacokinetics (PK) and pharmacodynamics (PD) values of CFPM/NAC in mice infected with β-lactamase-producing Enterobacterales, such as the carbapenemase-producing Enterobacterales. METHODS Three strains of β-lactamase-producing Enterobacterales, Klebsiella pneumoniae MSC 21444, Escherichia coli MSC 20662, and K. pneumoniae ATCC BAA-1898, were used for checkerboard assays and fractionation studies and dose-range studies. A PK study was performed in neutropenic mice. Additionally, PK/PD analysis was performed based on the instantaneous minimum inhibitory concentration (MICi) concept. RESULTS Checkerboard measurements revealed that higher NAC concentrations decreased the CFPM MIC in a concentration-dependent manner. In all tested strains, fT > MICi calculated from the PK experiments showed a high correlation with the mean change in the bacterial count of thigh-infected mice in the in vivo PD study, suggesting that fT > MICi is an optimal PK/PD parameter for monitoring the CFPM/NAC combination. The target fT > MICi values for CFPM/NAC to achieve a bacteriostatic effect, 1-log10-kill, and 2-log10-kill values were 30, 49, and 94%, respectively. CONCLUSIONS Our results indicate that fT > MICi is a PK/PD parameter is suitable for monitoring the CFPM/NAC combination. The minimum target value for achieving a static effect against β-lactamase-producing Enterobacterales is 30%.
Collapse
Affiliation(s)
- Yuki Igarashi
- Division of Pharmacodynamics, Keio University Faculty of Pharmacy, 1-5-30 Shibakoen, Minato-Ku, Tokyo, 105-8512, Japan
| | - Wataru Takemura
- Division of Pharmacodynamics, Keio University Faculty of Pharmacy, 1-5-30 Shibakoen, Minato-Ku, Tokyo, 105-8512, Japan
| | - Xiaoxi Liu
- Division of Pharmacodynamics, Keio University Faculty of Pharmacy, 1-5-30 Shibakoen, Minato-Ku, Tokyo, 105-8512, Japan
| | - Nana Kojima
- Division of Pharmacodynamics, Keio University Faculty of Pharmacy, 1-5-30 Shibakoen, Minato-Ku, Tokyo, 105-8512, Japan
| | - Takumi Morita
- Division of Pharmacodynamics, Keio University Faculty of Pharmacy, 1-5-30 Shibakoen, Minato-Ku, Tokyo, 105-8512, Japan
| | - Victor Tuan Giam Chuang
- Discipline of Pharmacy, Curtin Medical School, Faculty of Health Sciences, Curtin University, GPO Box U1987, Perth, Western Australia, 6845, Australia
| | - Yuki Enoki
- Division of Pharmacodynamics, Keio University Faculty of Pharmacy, 1-5-30 Shibakoen, Minato-Ku, Tokyo, 105-8512, Japan
| | - Kazuaki Taguchi
- Division of Pharmacodynamics, Keio University Faculty of Pharmacy, 1-5-30 Shibakoen, Minato-Ku, Tokyo, 105-8512, Japan.
| | - Kazuaki Matsumoto
- Division of Pharmacodynamics, Keio University Faculty of Pharmacy, 1-5-30 Shibakoen, Minato-Ku, Tokyo, 105-8512, Japan
| |
Collapse
|
27
|
Sitovs A, Skadins I, Purvina S, Bandere D. In vitro and ex vivo antibacterial activity of levofloxacin against Pasteurella multocida and Escherichia coli isolated from rabbits (Oryctolagus cuniculus) - A preliminary study. J Vet Pharmacol Ther 2023; 46:332-343. [PMID: 37060264 DOI: 10.1111/jvp.13383] [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: 01/27/2023] [Revised: 04/04/2023] [Accepted: 04/05/2023] [Indexed: 04/16/2023]
Abstract
Levofloxacin veterinary formulations are available in Argentina, China and India for the use in dogs, cattle, pig and sheep, but not currently in the rabbit. Only the extra-label use in rabbits is possible. Levofloxacin is not labelled for veterinary use in the EU or the USA. The activity of levofloxacin against rabbit pathogens Pasteurella multocida (P. multocida) and Escherichia coli (E. coli) was evaluated. Minimum inhibitory concentration (MIC) and minimal bactericidal concentration (MBC) were determined in broth and serum for 10 P. multocida isolates and 5 E. coli isolates from rabbits. One isolate of each bacterial species was used for the time-killing curve study in vitro and ex vivo. In vitro AUC24 /MIC ratios were used for building the inhibitory pharmacodynamic Imax model. The P. multocida MIC were 0.008-0.5 μg/mL, MBC - 0.015-0.5 μg/mL. Escherichia coli MIC was 0.008-0.03 μg/mL and MBC - 0.03-0.25 μg/mL. Bacterial counts were reduced to the limit of detection after 24 h with levofloxacin concentrations of 2 MIC and higher. All serum samples from rabbits treated with levofloxacin eliminated the bacteria within 24 h. AUC24 /MIC ratios for bacteriostatic, bactericidal and bacterial elimination effects for P. multocida and E. coli isolates were 21, 29 and 75 h and 27, 32 and 60 h, respectively. Proposed daily doses against P. multocida (MIC = 0.015 μg/mL) and E. coli (MIC = 0.03 μg/mL) isolates were calculated as ≤0.91 and ≤1.43 mg/kg, respectively. Fluoroquinolones are categorized by WHO as 'highest priority critically important antimicrobials'. Considering the increasing importance of antimicrobial stewardship, antimicrobials from a lower importance category that are active against the isolate of interest should be used in preference to fluoroquinolones. Fluoroquinolone use in veterinary medicine should be based on antimicrobial susceptibility testing in order to mitigate the risk to public health and prevent the spread of bacterial resistance.
Collapse
Affiliation(s)
- Andrejs Sitovs
- Department of Pharmacology, Riga Stradins University, Riga, Latvia
| | - Ingus Skadins
- Department of Biology and Microbiology, Riga Stradins University, Riga, Latvia
| | - Santa Purvina
- Department of Pharmacology, Riga Stradins University, Riga, Latvia
| | - Dace Bandere
- Department of Pharmaceutical Chemistry, Riga Stradins University, Riga, Latvia
| |
Collapse
|
28
|
Yedle R, Reniguntla MK, Puttaswamy R, Puttarangappa P, Hiremath S, Nanjundappa M, Jayaraman R. Neutropenic Rat Thigh Infection Model for Evaluation of the Pharmacokinetics/Pharmacodynamics of Anti-Infectives. Microbiol Spectr 2023; 11:e0013323. [PMID: 37260385 PMCID: PMC10433970 DOI: 10.1128/spectrum.00133-23] [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: 01/10/2023] [Accepted: 05/10/2023] [Indexed: 06/02/2023] Open
Abstract
The neutropenic mouse infection model is extensively used to characterize the pharmacokinetics/pharmacodynamics (PK/PD) of anti-infective agents. However, it is difficult to evaluate agents following intravenous (i.v.) infusions using this model. Furthermore, in many drug discovery programs, lead identification and optimization is performed in rats, and pharmacology is performed in mice. Alternative models of infection are needed for robust predictions of PK/PD in humans. The rat is an alternative model of infection which can overcome the shortcomings of the mouse model. However, the rat neutropenic thigh infection (NTI) model has not been adequately characterized for evaluation of the PK/PD of anti-infectives. The aim of this study was to characterize the PK/PD of ciprofloxacin against bacterial pathogens in a rat NTI model. We studied the PK/PD relationships of ciprofloxacin against wild-type Escherichia coli, Acinetobacter baumannii, Pseudomonas aeruginosa, and Klebsiella pneumoniae in neutropenic Wistar rats following administration of 10, 30, and 100 mg/kg as single intravenous boluses and 30- and 60-min infusions. The PK/PD of ciprofloxacin against all four pathogens was AUC/MIC dependent and independent of the duration of administration at 10, 30, and 100 mg/kg. At human-equivalent rat doses, the PK/PD targets of ciprofloxacin achieved in rats for microbiological cure were similar to those reported in human patients. The neutropenic rat thigh infection model can be used to evaluate anti-infective agents intended to be administered as infusions in the clinic, and it complements the mouse model, increasing the robustness of PK/PD predictions in humans. IMPORTANCE Many antibiotics are administered as intravenous infusions in the clinic, especially in intensive care units. Anti-infective drug discovery companies develop clinical candidates that are intended to be administered as i.v. infusions in the clinic. However, there are no well-characterized models with which they can evaluate the PK/PD of the candidates following i.v. infusions. The neutropenic rat thigh infection model reported in this study helps in evaluating anti-infective agents that are intended to be administered as i.v. infusions in the clinic. The rat model is useful for simulating the clinical conditions for i.v. infusions for treatment of infections, such as acute bacterial skin and skin structure, lung, and urinary tract infections. This model is predictive of efficacy in humans and can serve as an additional confirmatory model, along with the mouse model, for determining the proof of concept and for making robust predictions of efficacy in humans.
Collapse
Affiliation(s)
- Randhir Yedle
- TheraIndx Lifesciences Pvt. Ltd., Nelamangala, Bangalore, India
| | | | | | | | | | | | | |
Collapse
|
29
|
Stocker SL, Alffenaar JWC. Predicting early bactericidal activity using pre-clinical data for tuberculosis drugs: a platform for model-informed drug discovery and development. Eur Respir J 2023; 62:2301108. [PMID: 37652563 DOI: 10.1183/13993003.01108-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 07/14/2023] [Indexed: 09/02/2023]
Affiliation(s)
- Sophie L Stocker
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
- Department of Clinical Pharmacology and Toxicology, St. Vincent's Hospital, Sydney, Australia
- Sydney Institute for Infectious Diseases, The University of Sydney, Sydney, Australia
| | - Jan-Willem C Alffenaar
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Sydney Institute for Infectious Diseases, The University of Sydney, Sydney, Australia
- Westmead Hospital, Sydney, Australia
| |
Collapse
|
30
|
Bentley S, Cheong J, Gudka N, Makhecha S, Hadjisymeou-Andreou S, Standing JF. Therapeutic drug monitoring-guided dosing for pediatric cystic fibrosis patients: recent advances and future outlooks. Expert Rev Clin Pharmacol 2023; 16:715-726. [PMID: 37470695 DOI: 10.1080/17512433.2023.2238597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/10/2023] [Accepted: 07/17/2023] [Indexed: 07/21/2023]
Abstract
INTRODUCTION Medicine use in children with cystic fibrosis (CF) is complicated by inconsistent pharmacokinetics at variance with the general population, a lack of research into this and its effects on clinical outcomes. In the absence of established dose regimens, therapeutic drug monitoring (TDM) is a clinically relevant tool to optimize drug exposure and maximize therapeutic effect by the bedside. In clinical practice though, use of this is variable and limited by a lack of expert recommendations. AREAS COVERED We aimed to review the use of TDM in children with CF to summarize recent developments, current recommendations, and opportunities for future directions. We searched PubMed for relevant publications using the broad search terms "cystic fibrosis" in combination with the specific terms "therapeutic drug monitoring (TDM)" and "children." Further searches were undertaken using the name of identified drugs combined with the term "TDM." EXPERT OPINION Further research into the use of Bayesian forecasting and the relationship between exposure and response is required to personalize dosing, with the opportunity for the development of expert recommendations in children with CF. Use of noninvasive methods of TDM has the potential to improve accessibility to TDM in this cohort.
Collapse
Affiliation(s)
- Siân Bentley
- Pharmacy Department, Royal Brompton Hospital, London, UK
| | - Jamie Cheong
- Pharmacy Department, Royal Brompton Hospital, London, UK
| | - Nikesh Gudka
- Pharmacy Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | | | | | - Joseph F Standing
- Pharmacy Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
- Infection, Immunity and Inflammation,great Ormond Street Institute of Child Health, University College London, London, UK
| |
Collapse
|
31
|
Joynt GM, Ling L, Wong WT, Lipman J. Therapeutic drug monitoring of carbapenem antibiotics in critically ill patients: an overview of principles, recommended dosing regimens, and clinical outcomes. Expert Rev Clin Pharmacol 2023; 16:703-714. [PMID: 36942827 DOI: 10.1080/17512433.2023.2194629] [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: 11/28/2022] [Accepted: 03/20/2023] [Indexed: 03/23/2023]
Abstract
INTRODUCTION The importance of antibiotic treatment for sepsis in critically ill septic patients is well established. Consistently achieving the dose of antibiotics required to optimally kill bacteria, minimize the development of resistance, and avoid toxicity is challenging. The increasing understanding of the pharmacokinetic and pharmacodynamic (PK/PD) characteristics of antibiotics, and the effects of critical illness on key PK/PD parameters, is gradually re-shaping how antibiotics are dosed in critically ill patients. AREAS COVERED The PK/PD characteristics of commonly used carbapenem antibiotics, the principles of the application of therapeutic drug monitoring (TDM), and current as well as future methods of utilizing TDM to optimally devise dosing regimens will be reviewed. The limitations and evidence-base supporting the use of carbapenem TDM to improve outcomes in critically ill patients will be examined. EXPERT OPINION It is important to understand the principles of TDM in order to correctly inform dosing regimens. Although the concept of TDM is attractive, and the ability to utilize PK software to optimize dosing in the near future is expected to rapidly increase clinicians' ability to meet pre-defined PK/PD targets more accurately, current evidence provides only limited support for the use of TDM to guide carbapenem dosing in critically ill patients.
Collapse
Affiliation(s)
- Gavin Matthew Joynt
- Department of Anaesthesia and Intensive Care, the Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lowell Ling
- Department of Anaesthesia and Intensive Care, the Chinese University of Hong Kong, Hong Kong SAR, China
| | | | - Jeffrey Lipman
- Department of Intensive Care Services, Royal Brisbane and Women's Hospital, Brisbane, Australia
- Division of Anaesthesia Intensive Care, Pain and Emergency Medicine, Nîmes University Hospital, University of Montpellier, Nîmes, France
- University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Jamieson Trauma Institute, Royal Brisbane and Women's Hospital, Brisbane, Australia
| |
Collapse
|
32
|
Tashiro S, Taguchi K, Enoki Y, Matsumoto K. Antimicrobial Efficacy Evaluations of Metronidazole against Clostridioides difficile Infection using Fecal Pharmacokinetic and Pharmacodynamic Analyses. Pharm Res 2023; 40:1799-1806. [PMID: 37226025 DOI: 10.1007/s11095-023-03537-6] [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: 02/07/2023] [Accepted: 05/13/2023] [Indexed: 05/26/2023]
Abstract
OBJECTIVES The pharmacokinetics/pharmacodynamics (PK/PD) characteristics of metronidazole (MNZ) in Clostridioides difficile infection (CDI) remain unclear. We aimed to determine the PK/PD characteristics of MNZ using a fecal PK/PD analysis model. METHODS Susceptibility testing, time-kill studies, and post-antibiotic effect (PAE) measurements were performed to evaluate in vitro PD profiles. MNZ was subcutaneously administered to mice infected with C. difficile ATCC® 43255 to evaluate in vivo PK and PD profiles, followed by determining fecal PK/PD indices with target value. RESULTS MNZ exerted concentration-dependent bactericidal activities with minimum inhibitory concentration (MIC) and PAE being 0.79 µg/mL and 4.8 h, respectively, against C. difficile ATCC® 43255. The reduction in vegetative cells in feces and treatment outcomes were most closely correlated with the ratio of the area under the fecal drug concentration-time curve from 0 to 24 h to the MIC (fecal AUC24/MIC). The target value of fecal AUC24/MIC to achieve a 1 log10 reduction in vegetative cells was 188. Upon meeting the target value, high survival rates (94.5%) and low clinical sickness score grading (5.2) were achieved in the CDI mouse models. CONCLUSIONS The PK/PD index and its target value of MNZ for CDI treatment was fecal AUC24/MIC ≥ 188. These findings may contribute to the effective clinical use of MNZ.
Collapse
Affiliation(s)
- Sho Tashiro
- Division of Pharmacodynamics, Keio University Faculty of Pharmacy, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Kazuaki Taguchi
- Division of Pharmacodynamics, Keio University Faculty of Pharmacy, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan.
| | - Yuki Enoki
- Division of Pharmacodynamics, Keio University Faculty of Pharmacy, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Kazuaki Matsumoto
- Division of Pharmacodynamics, Keio University Faculty of Pharmacy, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| |
Collapse
|
33
|
Hilbert DW, DeRyke CA, Motyl M, Hackel M, Young K. Relebactam restores susceptibility of resistant Pseudomonas aeruginosa and Enterobacterales and enhances imipenem activity against chromosomal AmpC-producing species: analysis of global SMART 2018-2020. BMC Microbiol 2023; 23:165. [PMID: 37312049 PMCID: PMC10262423 DOI: 10.1186/s12866-023-02864-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 04/18/2023] [Indexed: 06/15/2023] Open
Abstract
BACKGROUND Carbapenem-resistant bacteria are an increasing problem in clinical practice; thus, it is important to identify β-lactamase inhibitors (e.g., relebactam) that can restore carbapenem susceptibility. We report analyses of relebactam enhancement of imipenem activity against both imipenem-nonsusceptible (NS) and imipenem-susceptible (S) Pseudomonas aeruginosa and Enterobacterales. Gram-negative bacterial isolates were collected for the ongoing Study for Monitoring Antimicrobial Resistance Trends global surveillance program. Clinical and Laboratory Standards Institute-defined broth microdilution minimum inhibitory concentrations (MIC) were used to determine the imipenem and imipenem/relebactam antibacterial susceptibilities of P. aeruginosa and Enterobacterales isolates. RESULTS Between 2018 and 2020, 36.2% of P. aeruginosa (N = 23,073) and 8.2% of Enterobacterales (N = 91,769) isolates were imipenem-NS. Relebactam restored imipenem susceptibility in 64.1% and 49.4% of imipenem-NS P. aeruginosa and Enterobacterales isolates, respectively. Restoration of susceptibility was largely observed among K. pneumoniae carbapenemase-producing Enterobacterales and carbapenemase-negative P. aeruginosa. Relebactam also caused a lowering of imipenem MIC among imipenem-S P. aeruginosa and Enterobacterales isolates from chromosomal Ambler class C β-lactamase (AmpC)-producing species. For both imipenem-NS and imipenem-S P. aeruginosa isolates, relebactam reduced the imipenem MIC mode from 16 μg/mL to 1 μg/mL and from 2 μg/mL to 0.5 μg/mL, respectively, compared with imipenem alone. CONCLUSIONS Relebactam restored imipenem susceptibility among nonsusceptible isolates of P. aeruginosa and Enterobacterales and enhanced imipenem susceptibility among susceptible isolates of P. aeruginosa and isolates from Enterobacterales species that can produce chromosomal AmpC. The reduced imipenem modal MIC values with relebactam may result in a higher probability of target attainment in patients.
Collapse
Affiliation(s)
| | | | | | - Meredith Hackel
- International Health Management Associates, Inc, Schaumburg, IL, USA
| | | |
Collapse
|
34
|
Gill CM, Nicolau DP. In Vivo Pharmacodynamic Profile of EVER206, a Novel Polymyxin Antimicrobial, against Gram-Negative Bacteria in the Murine Thigh Infection Model. Antimicrob Agents Chemother 2023; 67:e0173822. [PMID: 37022170 PMCID: PMC10190685 DOI: 10.1128/aac.01738-22] [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] [Received: 12/30/2022] [Accepted: 03/15/2023] [Indexed: 04/07/2023] Open
Abstract
The objective was to determine the magnitude of the EVER206 free-plasma area under the concentration time curve (fAUC)/MIC target associated with bacteriostasis and 1-log10 kill against clinically relevant Gram-negative bacteria in the murine thigh model. Twenty-seven clinical isolates (Pseudomonas aeruginosa, n = 10; Escherichia coli, n = 9; Klebsiella pneumoniae, n = 5; Enterobacter cloacae, n = 2; and Klebsiella aerogenes, n = 1) were tested. Mice were pretreated with cyclophosphamide (induce neutropenia) and uranyl nitrate (increase the exposure of test compound through predictable renal dysfunction). Two hours postinoculation, five doses of EVER206 were administered subcutaneously. EVER206 pharmacokinetics were determined in infected mice. Data were fit using maximum effect (Emax) models to elucidate the fAUC/MIC targets for stasis and 1-log10 bacterial kill (reported as mean [range] by species). EVER206 MICs (mg/L) ranged from 0.25 to 2 mg/L (P. aeruginosa), 0.06 to 2 mg/L (E. coli), 0.06 to 0.125 mg/L (E. cloacae), 0.06 mg/L (K. aerogenes), and 0.06 to 2 mg/L (K. pneumoniae). In vivo, the mean 0-h baseline bacterial burden was 5.57 ± 0.39 log10 CFU/thigh. Stasis was achieved in 9/10 P. aeruginosa (fAUC/MIC, 88.13 [50.33 to 129.74]), 9/9 E. coli (fAUC/MIC, 112.84 [19.19 to 279.38]), 2/2 E. cloacae (fAUC/MIC, 259.28 [124.08 to 394.47]), 0/1 K. aerogenes, and 4/5 K. pneumoniae (fAUC/MIC, 99.26 [62.3 to 144.43]) isolates tested. 1-log10 kill was achieved in 9/10 for P. aeruginosa (fAUC/MIC, 106.43 [55.22 to 152.08]), 3/9 for E. coli (fAUC/MIC, 258.96 [74.08 to 559.4]), and 1/2 for E. cloacae (fAUC/MIC, 255.33). Using the murine thigh model, the fAUC/MIC targets of EVER206 were assessed across a broad MIC distribution. Integrating these data with microbiologic and clinical exposure data will aid in determining the clinical dose of EVER206.
Collapse
Affiliation(s)
- Christian M. Gill
- Center for Anti-Infective Research and Development, Hartford Hospital, Hartford, Connecticut, USA
| | - David P. Nicolau
- Center for Anti-Infective Research and Development, Hartford Hospital, Hartford, Connecticut, USA
- Division of Infectious Diseases, Hartford Hospital, Hartford, Connecticut, USA
| |
Collapse
|
35
|
Legg A, Carmichael S, Chai MG, Roberts JA, Cotta MO. Beta-Lactam Dose Optimisation in the Intensive Care Unit: Targets, Therapeutic Drug Monitoring and Toxicity. Antibiotics (Basel) 2023; 12:antibiotics12050870. [PMID: 37237773 DOI: 10.3390/antibiotics12050870] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/31/2023] [Accepted: 05/01/2023] [Indexed: 05/28/2023] Open
Abstract
Beta-lactams are an important family of antibiotics used to treat infections and are commonly used in critically ill patients. Optimal use of these drugs in the intensive care unit (ICU) is important because of the serious complications from sepsis. Target beta-lactam antibiotic exposures may be chosen using fundamental principles of beta-lactam activity derived from pre-clinical and clinical studies, although the debate regarding optimal beta-lactam exposure targets is ongoing. Attainment of target exposures in the ICU requires overcoming significant pharmacokinetic (PK) and pharmacodynamic (PD) challenges. For beta-lactam drugs, the use of therapeutic drug monitoring (TDM) to confirm if the desired exposure targets are achieved has shown promise, but further data are required to determine if improvement in infection-related outcomes can be achieved. Additionally, beta-lactam TDM may be useful where a relationship exists between supratherapeutic antibiotic exposure and drug adverse effects. An ideal beta-lactam TDM service should endeavor to efficiently sample and report results in identified at-risk patients in a timely manner. Consensus beta-lactam PK/PD targets associated with optimal patient outcomes are lacking and should be a focus for future research.
Collapse
Affiliation(s)
- Amy Legg
- Menzies School of Health Research, Tiwi, Darwin, NT 0810, Australia
- Herston Infectious Diseases Institute, Herston, Brisbane, QLD 4029, Australia
| | - Sinead Carmichael
- Royal Brisbane and Women's Hospital, Departments of Intensive Care Medicine and Pharmacy, Brisbane, QLD 4029, Australia
| | - Ming G Chai
- Faculty of Medicine, University of Queensland Centre for Clinical Research (UQCCR), Brisbane, QLD 4029, Australia
| | - Jason A Roberts
- Herston Infectious Diseases Institute, Herston, Brisbane, QLD 4029, Australia
- Royal Brisbane and Women's Hospital, Departments of Intensive Care Medicine and Pharmacy, Brisbane, QLD 4029, Australia
- Faculty of Medicine, University of Queensland Centre for Clinical Research (UQCCR), Brisbane, QLD 4029, Australia
- Division of Anaesthesiology Critical Care Emergency and Pain Medicine, Nîmes University Hospital, University of Montpellier, 30029 Nîmes, France
| | - Menino O Cotta
- Faculty of Medicine, University of Queensland Centre for Clinical Research (UQCCR), Brisbane, QLD 4029, Australia
| |
Collapse
|
36
|
Arends SJR, Butler D, Scangarella-Oman N, Castanheira M, Mendes RE. Antimicrobial Activity of Gepotidacin Tested against Escherichia coli and Staphylococcus saprophyticus Isolates Causing Urinary Tract Infections in Medical Centers Worldwide (2019 to 2020). Antimicrob Agents Chemother 2023; 67:e0152522. [PMID: 36877017 PMCID: PMC10112209 DOI: 10.1128/aac.01525-22] [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] [Received: 11/16/2022] [Accepted: 02/02/2023] [Indexed: 03/07/2023] Open
Abstract
The in vitro activities of gepotidacin and comparator agents against 3,560 Escherichia coli and 344 Staphylococcus saprophyticus collected from female (81.1%) and male (18.9%) patients with urinary tract infections (UTIs) in a global prospective surveillance program in 2019 to 2020 were determined. Isolates collected from 92 medical centers in 25 countries, including the United States, Europe, Latin America, and Japan, were tested for susceptibility by reference methods in a central monitoring laboratory. Gepotidacin inhibited 98.0% (3,488/3,560 isolates) of E. coli and 100% (344/344 isolates) of S. saprophyticus at gepotidacin concentrations of ≤4 μg/mL and ≤0.25 μg/mL, respectively. This activity was largely unaffected with isolates that demonstrated resistance phenotypes to other oral standard-of-care antibiotics, including amoxicillin-clavulanic acid, cephalosporins, fluoroquinolones, fosfomycin, nitrofurantoin, and trimethoprim-sulfamethoxazole. Gepotidacin also inhibited 94.3% (581/616 isolates) of E. coli isolates with an extended-spectrum β-lactamase-producing phenotype, 97.2% (1,085/1,129 isolates) of E. coli isolates resistant to ciprofloxacin, 96.1% (874/899) of E. coli isolates resistant to trimethoprim-sulfamethoxazole, and 96.3% (235/244 isolates) of multidrug-resistant E. coli isolates at gepotidacin concentrations of ≤4 μg/mL. In summary, gepotidacin demonstrated potent activity against a large collection of contemporary UTI E. coli and S. saprophyticus strains collected from patients worldwide. These data support the further clinical development of gepotidacin as a potential treatment option for patients with uncomplicated UTIs.
Collapse
|
37
|
Chaira T, Subramani C, Barman TK. ADME, Pharmacokinetic Scaling, Pharmacodynamic and Prediction of Human Dose and Regimen of Novel Antiviral Drugs. Pharmaceutics 2023; 15:pharmaceutics15041212. [PMID: 37111697 PMCID: PMC10146820 DOI: 10.3390/pharmaceutics15041212] [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: 01/23/2023] [Revised: 03/29/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
The search for new drugs is an extremely time-consuming and expensive endeavour. Much of that time and money go into generating predictive human pharmacokinetic profiles from preclinical efficacy and safety animal data. These pharmacokinetic profiles are used to prioritize or minimize the attrition at later stages of the drug discovery process. In the area of antiviral drug research, these pharmacokinetic profiles are equally important for the optimization, estimation of half-life, determination of effective dose, and dosing regimen, in humans. In this article we have highlighted three important aspects of these profiles. First, the impact of plasma protein binding on two primary pharmacokinetic parameters-volume of distribution and clearance. Second, interdependence of primary parameters on unbound fraction of the drug. Third, the ability to extrapolate human pharmacokinetic parameters and concentration time profiles from animal profiles.
Collapse
Affiliation(s)
- Tridib Chaira
- Department of Pharmacology, SGT University, Gurugram 122505, Haryana, India
| | - Chandru Subramani
- Department of Pathology, Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Tarani Kanta Barman
- Department of Pathology, Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77550, USA
| |
Collapse
|
38
|
Khalid K, Rox K. All Roads Lead to Rome: Enhancing the Probability of Target Attainment with Different Pharmacokinetic/Pharmacodynamic Modelling Approaches. Antibiotics (Basel) 2023; 12:antibiotics12040690. [PMID: 37107052 PMCID: PMC10135278 DOI: 10.3390/antibiotics12040690] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
In light of rising antimicrobial resistance and a decreasing number of antibiotics with novel modes of action, it is of utmost importance to accelerate development of novel treatment options. One aspect of acceleration is to understand pharmacokinetics (PK) and pharmacodynamics (PD) of drugs and to assess the probability of target attainment (PTA). Several in vitro and in vivo methods are deployed to determine these parameters, such as time-kill-curves, hollow-fiber infection models or animal models. However, to date the use of in silico methods to predict PK/PD and PTA is increasing. Since there is not just one way to perform the in silico analysis, we embarked on reviewing for which indications and how PK and PK/PD models as well as PTA analysis has been used to contribute to the understanding of the PK and PD of a drug. Therefore, we examined four recent examples in more detail, namely ceftazidime-avibactam, omadacycline, gepotidacin and zoliflodacin as well as cefiderocol. Whereas the first two compound classes mainly relied on the ‘classical’ development path and PK/PD was only deployed after approval, cefiderocol highly profited from in silico techniques that led to its approval. Finally, this review shall highlight current developments and possibilities to accelerate drug development, especially for anti-infectives.
Collapse
Affiliation(s)
- Kashaf Khalid
- Department of Chemical Biology, Helmholtz Centre for Infection Research (HZI), Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Katharina Rox
- Department of Chemical Biology, Helmholtz Centre for Infection Research (HZI), Inhoffenstraße 7, 38124 Braunschweig, Germany
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany
| |
Collapse
|
39
|
Asempa TE, Kois AK, Gill CM, Nicolau DP. Phenotypes, genotypes and breakpoints: an assessment of β-lactam/β-lactamase inhibitor combinations against OXA-48. J Antimicrob Chemother 2023; 78:636-645. [PMID: 36626311 DOI: 10.1093/jac/dkac425] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 09/21/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Two of the three recently approved β-lactam agent (BL)/β-lactamase inhibitor (BLI) combinations have higher CLSI susceptibility breakpoints (ceftazidime/avibactam 8 mg/L; meropenem/vaborbactam 4 mg/L) compared with the BL alone (ceftazidime 4 mg/L; meropenem 1 mg/L). This can lead to a therapeutic grey area on susceptibility reports depending on resistance mechanism. For instance, a meropenem-resistant OXA-48 isolate (MIC 4 mg/L) may appear as meropenem/vaborbactam-susceptible (MIC 4 mg/L) despite vaborbactam's lack of OXA-48 inhibitory activity. METHODS OXA-48-positive (n = 51) and OXA-48-negative (KPC, n = 5; Klebsiella pneumoniae wild-type, n = 1) Enterobacterales were utilized. Susceptibility tests (broth microdilution) were conducted with ceftazidime/avibactam, imipenem/relebactam and meropenem/vaborbactam, as well as their respective BL partner. Antimicrobial activity of all six agents was evaluated in the murine neutropenic thigh model using clinically relevant exposures. Efficacy was assessed as the change in bacterial growth at 24 h, compared with 0 h controls. RESULTS On average, the three BL/BLI agents resulted in robust bacteria killing among OXA-48-negative isolates. Among OXA-48-positive isolates, poor in vivo activity with imipenem/relebactam was concordant with its resistant phenotypic profile. Variable meropenem/vaborbactam activity was observed among isolates with a 'susceptible' MIC of 4 mg/L. Only 30% (7/23) of isolates at meropenem/vaborbactam MICs of 2 and 4 mg/L met the ≥1-log bacterial reduction threshold predictive of clinical efficacy in serious infections. In contrast, ceftazidime/avibactam resulted in marked bacterial density reduction across the range of MICs, and 96% (49/51) of isolates exceeded the ≥1-log bacterial reduction threshold. CONCLUSIONS Data demonstrate that current imipenem/relebactam and ceftazidime/avibactam CLSI breakpoints are appropriate. Data also suggest that higher meropenem/vaborbactam breakpoints relative to meropenem can translate to potentially poor clinical outcomes in patients infected with OXA-48-harbouring isolates.
Collapse
Affiliation(s)
- Tomefa E Asempa
- Center for Anti-Infective Research and Development, Hartford Hospital, Hartford, CT, USA
| | - Abigail K Kois
- Center for Anti-Infective Research and Development, Hartford Hospital, Hartford, CT, USA
| | - Christian M Gill
- Center for Anti-Infective Research and Development, Hartford Hospital, Hartford, CT, USA
| | - David P Nicolau
- Center for Anti-Infective Research and Development, Hartford Hospital, Hartford, CT, USA
- Division of Infectious Diseases, Hartford Hospital, Hartford, CT, USA
| |
Collapse
|
40
|
Pharmacokinetic/Pharmacodynamic Index Linked to In Vivo Efficacy of the Ampicillin-Ceftriaxone Combination against Enterococcus faecalis. Antimicrob Agents Chemother 2023; 67:e0096622. [PMID: 36695584 PMCID: PMC9933695 DOI: 10.1128/aac.00966-22] [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: 01/26/2023] Open
Abstract
Combination therapy with ampicillin plus ceftriaxone (AMP+CRO) is the first-line therapy for treating severe infections due to Enterococcus faecalis. However, the pharmacokinetic/pharmacodynamic (PK/PD) index linked to the in vivo efficacy of the combination is not yet defined, hindering dose optimization in the clinic. Because classical PK/PD indices are not directly applicable to antimicrobial combinations, two novel indices were tested in the optimized murine model of infection by E. faecalis to delineate the potentiation of AMP by CRO: the time above the CRO threshold (T>threshold) and the time above the AMP instantaneous MIC (T>MICi). The potential clinical relevance was evaluated by simulating human doses of AMP and CRO. Hill's equation fitted well the exposure-response data in terms of T>threshold, with a CRO threshold of 1 mg/L. The required exposures were 46%, 49%, and 52% for stasis and 1- and 2-log10 killing, respectively. Human ceftriaxone doses of 2 g every 12 h (q12h) would reach the target in >90% of strains with thresholds ≤64 mg/L. The AMP T>MICi index also fitted well, and the required exposures were 37%, 41%, and 46% for stasis and 1- and 2-log10 killing, respectively. In humans, the addition of CRO would allow use of lower AMP doses to reach the same T>MICi and to treat strains with higher MICs. This is the first report of the PK/PD indices and required magnitudes linked to AMP+CRO against E. faecalis; these results can be used as the basis to guide the design of clinical trials to improve combined therapy against enterococci.
Collapse
|
41
|
Igarashi Y, Takemura W, Liu X, Kojima N, Morita T, Chuang VTG, Enoki Y, Taguchi K, Matsumoto K. Development of an optimized and practical pharmacokinetics/pharmacodynamics analysis method for aztreonam/nacubactam against carbapenemase-producing K. pneumoniae. J Antimicrob Chemother 2023; 78:991-999. [PMID: 36775998 PMCID: PMC10068424 DOI: 10.1093/jac/dkad033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 01/25/2023] [Indexed: 02/14/2023] Open
Abstract
BACKGROUND Nacubactam, a new β-lactamase inhibitor with antibacterial activity, is being developed as a single drug to be co-administered with cefepime or aztreonam. However, determining pharmacokinetics/pharmacodynamics (PK/PD) parameters in β-lactam/β-lactamase inhibitor combinations remains challenging. We aimed to establish a practical PK/PD analysis method for aztreonam/nacubactam that incorporates instantaneous MIC (MICi). METHODS Based on chequerboard MIC measurements, MICi of aztreonam against carbapenemase-producing Klebsiella pneumoniae in the presence of nacubactam was simulated. RESULTS The mean change in the bacterial count of thigh-infected mice in an in vivo PD study was plotted based on %fT>MICi and analysed using the inhibitory effect sigmoid Imax model. fT>MICi calculated from the PK experiments showed a high correlation with the in vivo bactericidal effect, suggesting that fT>MICi is the optimal PK/PD parameter for aztreonam/nacubactam. The target values of fT>MICi achieving growth inhibition, 1 log10 kill and 2 log10 kill, were 22, 38% and 75%, respectively. CONCLUSIONS The PK/PD analysis method proposed in this study is promising for determining practical PK/PD parameters in combination therapy. In addition, this is the first report of aztreonam/nacubactam showing a potent in vivo therapeutic effect against NDM-producing K. pneumoniae.
Collapse
Affiliation(s)
- Yuki Igarashi
- Division of Pharmacodynamics, Keio University Faculty of Pharmacy, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Wataru Takemura
- Division of Pharmacodynamics, Keio University Faculty of Pharmacy, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Xiaoxi Liu
- Division of Pharmacodynamics, Keio University Faculty of Pharmacy, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Nana Kojima
- Division of Pharmacodynamics, Keio University Faculty of Pharmacy, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Takumi Morita
- Division of Pharmacodynamics, Keio University Faculty of Pharmacy, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Victor Tuan Giam Chuang
- Discipline of Pharmacy, Curtin Medical School, Faculty of Health Sciences, Curtin University, GPO Box U1987, Perth, Western Australia 6845, Australia
| | - Yuki Enoki
- Division of Pharmacodynamics, Keio University Faculty of Pharmacy, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Kazuaki Taguchi
- Division of Pharmacodynamics, Keio University Faculty of Pharmacy, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Kazuaki Matsumoto
- Division of Pharmacodynamics, Keio University Faculty of Pharmacy, 1-5-30 Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| |
Collapse
|
42
|
A liquid chromatography-tandem mass spectrometry method for the quantification of ampicillin/sulbactam and clindamycin in jawbone, plasma, and platelet-rich fibrin: Application to patients with osteonecrosis of the jaw. J Pharm Biomed Anal 2023; 224:115167. [PMID: 36435082 DOI: 10.1016/j.jpba.2022.115167] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/13/2022] [Accepted: 11/14/2022] [Indexed: 11/22/2022]
Abstract
Ampicillin in combination with sulbactam is a widely used drug choice for infection prophylaxis, especially in oral and maxillofacial surgery. Clindamycin serves as an alternative in patients with known allergy to β-lactam antibiotics. To ensure effective prophylaxis, it is important to achieve sufficiently high concentrations of active antibiotic substances in the tissues affected by the surgery. To this end, a LC-MS/MS method was developed and validated that allows the quantification of ampicillin, sulbactam and clindamycin in jawbone, plasma, and so-called platelet-rich fibrin (PRF). Validation was performed in accordance with the European Medicines Agency guidelines for bioanalytical method validation. For all matrices, sample processing was carried out by protein precipitation with acetonitrile or methanol 80%, containing the isotope labelled internal standards (IS) of the three drugs. Analytes were separated on a pentaflourophenyl column at 20 °C using gradient elution. Furthermore, detection was accomplished by electrospray ionisation in positive-ion mode (ampicillin, clindamycin and corresponding IS) and negative-ion mode (sulbactam and corresponding IS) in combination with multiple reaction monitoring. Depending on the analyte and the matrix under investigation, calibration curves ranged from 0.14 to 59.8 µg/g (jawbone - ampicillin), 2.0-1000 µg/mL (plasma - ampicillin), and 1.0-495 µg/mL (PRF - ampicillin). All analytes fulfilled the requirements of the guideline regarding sensitivity, linearity, selectivity, carryover, within-run and between run accuracy and precision, matrix effect and extraction recovery in all matrices. The method was successfully applied to measure concentrations of ampicillin, sulbactam and clindamycin in real-life samples obtained in routine clinical practice.
Collapse
|
43
|
Scangarella-Oman NE, Hossain M, Perry CR, Tiffany C, Powell M, Swift B, Dumont EF. Dose selection for a phase III study evaluating gepotidacin (GSK2140944) in the treatment of uncomplicated urogenital gonorrhoea. Sex Transm Infect 2023; 99:64-69. [PMID: 36411033 PMCID: PMC9887395 DOI: 10.1136/sextrans-2022-055518] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 09/30/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Gepotidacin is a novel, first-in-class triazaacenaphthylene antibiotic that inhibits bacterial DNA replication by a distinct mechanism of action and is active against most strains of Neisseria gonorrhoeae (N. gonorrhoeae). Phase II data suggested higher exposures were needed for efficacy and to suppress resistance development. A translational approach using in vitro pharmacokinetic/pharmacodynamic (PK/PD) and clinical data was used to select a gepotidacin dose for a phase III study. In this narrative review of previously shown data, we summarise how a translational approach based on in vitro PK/PD and population PK modelling and simulation data was undertaken to select a dosing regimen for the ongoing phase III gepotidacin study in participants with uncomplicated urogenital gonorrhoea. METHODS For dose selection, prior in vitro minimum inhibitory concentrations (MICs) and PK/PD data were available. PK modelling was conducted to determine a dose that would limit plasma concentrations to less than 14 µg/mL (as concentrations above this are associated with QT prolongation and effects associated with acetylcholinesterase inhibition) while maintaining ≥90% probability of target attainment (PTA) for efficacy and resistance suppression against N. gonorrhoeae isolates with gepotidacin MICs ≤1 µg/mL. RESULTS Two 3000 mg gepotidacin doses, administered 10-12 hours apart, resulted in PTA of ≥97.5% and ≥91.7% for gepotidacin MICs ≤1 µg/mL for the ratio of the area under the free drug plasma concentration-time curve over 24 hours to the MIC (fAUC0-24/MIC) efficacy, and resistance suppression targets of 40 and 46, respectively, but limited the occurrence of maximum plasma concentrations ≥14 µg/mL. CONCLUSIONS Two gepotidacin 3000 mg oral doses 10-12 hours apart provide ~2-fold higher systemic exposures, increase efficacy for higher gepotidacin MIC N. gonorrhoeae isolates, reduce resistance potential and limit plasma concentrations of potential safety concern, compared with higher doses.
Collapse
Affiliation(s)
| | | | | | | | - Marcy Powell
- GSK, Research Triangle Park, North Carolina, USA
| | | | | |
Collapse
|
44
|
Hirano T, Ohge H, Ikawa K, Uegami S, Watadani Y, Shigemoto N, Yoshimura K, Kitagawa H, Kaiki Y, Morikawa N, Takahashi S. Pharmacokinetics of flomoxef in plasma, peritoneal fluid, peritoneum, and subcutaneous adipose tissue of patients undergoing lower gastrointestinal surgery: Dosing considerations based on site-specific pharmacodynamic target attainment. J Infect Chemother 2023; 29:186-192. [PMID: 36341996 DOI: 10.1016/j.jiac.2022.10.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/08/2022] [Accepted: 10/25/2022] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Flomoxef is generally used to treat abdominal infections and as antibiotic prophylaxis during lower gastrointestinal surgery. It is reportedly effective against extended-spectrum beta-lactamase (ESBL)-producing Enterobacteriaceae and an increasingly valuable alternative to carbapenems. However, its abdominal pharmacokinetics remain unclear. Herein, pharmacokinetic analysis of flomoxef in the abdominal tissue was conducted to simulate dosing regimens for pharmacodynamic target attainment in abdominal sites. METHODS Flomoxef (1 g) was administered intravenously to a patient 30 min before commencing elective lower gastrointestinal surgery. Samples of plasma, peritoneal fluid, peritoneum, and subcutaneous adipose tissue were collected during surgery. The flomoxef tissue concentrations were measured. Accordingly, non-compartmental and compartmental pharmacokinetic parameters were calculated, and simulations were conducted to evaluate site-specific pharmacodynamic target values. RESULTS Overall, 41 plasma samples, 34 peritoneal fluid samples, 38 peritoneum samples, and 41 subcutaneous adipose samples from 10 patients were collected. The mean peritoneal fluid-to-plasma ratio in the areas under the drug concentration-time curve was 0.68, the mean peritoneum-to-plasma ratio was 0.40, and the mean subcutaneous adipose tissue-to-plasma was 0.16. The simulation based on these results showed the dosing regimens (q8h [3 g/day] and q6h [4 g/day]) achieved the bactericidal effect (% T > minimum inhibitory concentration [MIC] = 40%) in all tissues at an MIC of 1 mg/L. CONCLUSIONS We elucidated the pharmacokinetics of flomoxef and simulated pharmacodynamics target attainment in the abdominal tissue. This study provides evidence concerning the use of optimal dosing regimens for treating abdominal infection caused by strains like ESBL-producing bacteria.
Collapse
Affiliation(s)
- Toshinori Hirano
- Department of Surgery, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima City, Hiroshima Prefecture, 734-8551, Japan.
| | - Hiroki Ohge
- Department of Infectious Diseases, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima City, Hiroshima Prefecture, 734-8551, Japan.
| | - Kazuro Ikawa
- Department of Clinical Pharmacotherapy, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima City, Hiroshima Prefecture, 734-8551, Japan.
| | - Shinnosuke Uegami
- Department of Surgery, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima City, Hiroshima Prefecture, 734-8551, Japan.
| | - Yusuke Watadani
- Department of Surgery, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima City, Hiroshima Prefecture, 734-8551, Japan.
| | - Norifumi Shigemoto
- Department of Infectious Diseases, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima City, Hiroshima Prefecture, 734-8551, Japan.
| | - Kosuke Yoshimura
- Department of Surgery, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima City, Hiroshima Prefecture, 734-8551, Japan.
| | - Hiroki Kitagawa
- Department of Infectious Diseases, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima City, Hiroshima Prefecture, 734-8551, Japan.
| | - Yuki Kaiki
- Department of Surgery, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima City, Hiroshima Prefecture, 734-8551, Japan.
| | - Norifumi Morikawa
- Department of Clinical Pharmacotherapy, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima City, Hiroshima Prefecture, 734-8551, Japan.
| | - Shinya Takahashi
- Department of Surgery, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima City, Hiroshima Prefecture, 734-8551, Japan.
| |
Collapse
|
45
|
Omolabi KF, Reddy N, Mdanda S, Ntshangase S, Singh SD, Kruger HG, Naicker T, Govender T, Bajinath S. The in vitro and in vivo potential of metal-chelating agents as metallo-beta-lactamase inhibitors against carbapenem-resistant Enterobacterales. FEMS Microbiol Lett 2023; 370:6912242. [PMID: 36521842 DOI: 10.1093/femsle/fnac122] [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: 05/11/2022] [Revised: 11/28/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
The recent surge in beta-lactamase resistance has created superbugs, which pose a current and significant threat to public healthcare. This has created an urgent need to keep pace with the discovery of inhibitors that can inactivate these beta-lactamase producers. In this study, the in vitro and in vivo activity of 1,4,7-triazacyclononane-1,4,7 triacetic acid (NOTA)-a potential metallo-beta-lactamase (MBL) inhibitor was evaluated in combination with meropenem against MBL producing bacteria. Time-kill studies showed that NOTA restored the efficacy of meropenem against all bacterial strains tested. A murine infection model was then used to study the in vivo pharmacokinetics and efficacy of this metal chelator. The coadministration of NOTA and meropenem (100 mg/kg.bw each) resulted in a significant decrease in the colony-forming units of Klebsiella pneumoniae NDM-1 over an 8-h treatment period (>3 log10 units). The findings suggest that chelators, such as NOTA, hold strong potential for use as a MBL inhibitor in treating carbapenem-resistant Enterobacterale infections.
Collapse
Affiliation(s)
- Kehinde F Omolabi
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Private Bag X54001,Durban 4000, South Africa
| | - Nakita Reddy
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Private Bag X54001,Durban 4000, South Africa
| | - Sipho Mdanda
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Private Bag X54001,Durban 4000, South Africa
| | - Sphamandla Ntshangase
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Private Bag X54001,Durban 4000, South Africa
| | - Sanil D Singh
- Department of Pharmaceutical Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4000, South Africa
| | - Hendrik G Kruger
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Private Bag X54001,Durban 4000, South Africa
| | - Tricia Naicker
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Private Bag X54001,Durban 4000, South Africa
| | - Thavendran Govender
- Department of Chemistry, University of Zululand, Private Bag X1001, KwaDlangezwa 3886, South Africa
| | - Sooraj Bajinath
- Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Private Bag X54001,Durban 4000, South Africa.,Department of Pharmaceutical Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4000, South Africa.,School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
| |
Collapse
|
46
|
Kuroda T, Minamijima Y, Mita H, Tamura N, Fukuda K, Kuwano A, Toutain PL, Sato F. Rational determination of cefazolin dosage regimen in horses based on pharmacokinetics/pharmacodynamics principles and Monte Carlo simulations. J Vet Pharmacol Ther 2023; 46:62-67. [PMID: 36245288 DOI: 10.1111/jvp.13099] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/30/2022] [Accepted: 10/03/2022] [Indexed: 01/11/2023]
Abstract
A pharmacokinetics/pharmacodynamics (PK/PD) approach was used to determine the best empirical dosage regimen of cefazolin (CEZ) after intramuscular (IM) administration of CEZ in horses. Seven horses received a single IM or intravenous (IV) administration of CEZ of 5 mg/kg bodyweight (BW) according to a crossover design. CEZ plasma concentrations were measured using LC-MS/MS. The plasma concentrations in these seven horses and those of six other horses obtained in a previous study with an IV CEZ dose of 10 mg/kg were modelled simultaneously using NonLinear Mixed-Effect modelling followed by Monte Carlo simulations to establish a rational dosage regimen. A 90% Probability of Target Attainment (PTA) for a PK/PD target of a free plasma concentration exceeding MIC90 (fT > MIC ) for 40% of the dosing interval was set for selecting an effective dosing regimen. The typical half-life of absorption and bioavailability after IM administration were 1.25 h and 96.8%, respectively. A CEZ dosage regimen of 5 mg/kg BW q12h IM administration achieved therapeutic concentrations to control both Streptococcus zooepidemicus and Staphylococcus aureus. For the same dose, the fT > MIC after IM administration was significantly longer than after IV administration, and the IM route should be favoured by clinicians for its efficiency and convenience.
Collapse
Affiliation(s)
- Taisuke Kuroda
- Clinical Veterinary Medicine Division, Equine Research Institute Japan Racing Association, Shimotsuke, Japan
| | - Yohei Minamijima
- Laboratory of Racing Chemistry, Drug Analysis Department, Utsunomiya, Japan
| | - Hiroshi Mita
- Clinical Veterinary Medicine Division, Equine Research Institute Japan Racing Association, Shimotsuke, Japan
| | - Norihisa Tamura
- Clinical Veterinary Medicine Division, Equine Research Institute Japan Racing Association, Shimotsuke, Japan
| | - Kentaro Fukuda
- Clinical Veterinary Medicine Division, Equine Research Institute Japan Racing Association, Shimotsuke, Japan
| | - Atsutoshi Kuwano
- Clinical Veterinary Medicine Division, Equine Research Institute Japan Racing Association, Shimotsuke, Japan
| | - Pierre-Louis Toutain
- Comparative Biomedical Sciences, The Royal Veterinary College, London, UK.,Intheres, Ecole Nationale Vétérinaire de Toulouse, Toulouse, France
| | - Fumio Sato
- Clinical Veterinary Medicine Division, Equine Research Institute Japan Racing Association, Shimotsuke, Japan
| |
Collapse
|
47
|
Hemmersbach-Miller M, Balevic SJ, Winokur PL, Landersdorfer CB, Gu K, Chan AW, Cohen-Wolkowiez M, Conrad T, An G, Kirkpatrick CMJ, Swamy GK, Walter EB, Schmader KE. Population Pharmacokinetics of Piperacillin/Tazobactam Across the Adult Lifespan. Clin Pharmacokinet 2023; 62:127-139. [PMID: 36633812 PMCID: PMC9969806 DOI: 10.1007/s40262-022-01198-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND AND OBJECTIVE Piperacillin/tazobactam is one of the most frequently used antimicrobials in older adults. Using an opportunistic study design, we evaluated the pharmacokinetics of piperacillin/tazobactam as a probe drug to evaluate changes in antibacterial drug exposure and dosing requirements, including in older adults. METHODS A total of 121 adult patients were included. The population pharmacokinetic models that best characterized the observed plasma concentrations of piperacillin and tazobactam were one-compartment structural models with zero-order input and linear elimination. RESULTS Among all potential covariates, estimated creatinine clearance had the most substantial impact on the elimination clearance for both piperacillin and tazobactam. After accounting for renal function and body size, there was no remaining impact of frailty on the pharmacokinetics of piperacillin and tazobactam. Monte Carlo simulations indicated that renal function had a greater impact on the therapeutic target attainment than age, although these covariates were highly correlated. Frailty, using the Canadian Study of Health and Aging Clinical Frailty Scale, was assessed in 60 patients who were ≥ 65 years of age. CONCLUSIONS The simulations suggested that adults ≤ 50 years of age infected with organisms with higher minimum inhibitory concentrations may benefit from continuous piperacillin/tazobactam infusions (12 g/day of piperacillin component) or extended infusions of 4 g every 8 hours. However, for a target of 50% fT + minimum inhibitory concentration, dosing based on renal function is generally preferable to dosing by age, and simulations suggested that patients with creatinine clearance ≥ 120 mL/min may benefit from infusions of 4 g every 8 hours for organisms with higher minimum inhibitory concentrations.
Collapse
Affiliation(s)
- Marion Hemmersbach-Miller
- Division of Infectious Diseases, Department of Internal Medicine, Duke University Medical Center, Durham, NC, USA
- Duke Clinical Research Institute, Durham, NC, USA
- ICON Plc, North Wales, PA, USA
| | - Stephen J Balevic
- Duke Clinical Research Institute, Durham, NC, USA
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
- Division of Rheumatology and Immunology, Department of Internal Medicine, Duke University Medical Center, Durham, NC, USA
| | - Patricia L Winokur
- Division of Infectious Diseases, Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | | | - Kenan Gu
- Division of Microbiology and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Austin W Chan
- Division of Infectious Diseases, Department of Internal Medicine, Duke University Medical Center, Durham, NC, USA
| | - Michael Cohen-Wolkowiez
- Duke Clinical Research Institute, Durham, NC, USA
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | | | - Guohua An
- College of Pharmacy, University of Iowa, Iowa City, IA, USA
| | - Carl M J Kirkpatrick
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Geeta K Swamy
- Department of Obstetrics and Gynecology, Obstetrics Clinical Research, Duke University Medical System, Durham, NC, USA
| | - Emmanuel B Walter
- Duke Clinical Research Institute, Durham, NC, USA
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Kenneth E Schmader
- Division of Geriatrics, Department of Medicine, Duke University Medical Center, Duke Box 3469, Durham, NC, 27710, USA.
- Geriatric Research Education and Clinical Center (GRECC), Durham Veterans Affairs Health Care System, Durham, NC, USA.
| |
Collapse
|
48
|
Liu X, Tashiro S, Igarashi Y, Takemura W, Kojima N, Morita T, Hayashi M, Enoki Y, Taguchi K, Matsumoto K. Differences in Pharmacokinetic/Pharmacodynamic Parameters of Tedizolid Against VRE and MRSA. Pharm Res 2023; 40:187-196. [PMID: 36329373 DOI: 10.1007/s11095-022-03425-5] [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: 07/24/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022]
Abstract
PURPOSE Vancomycin-resistant enterococci (VRE) have recently become a major cause of nosocomial infections and a global public health concern. Tedizolid exhibits powerful antibacterial activity against VRE in vitro, but its pharmacokinetic/pharmacodynamic (PK/PD) parameters remain unclear. Therefore, we aimed to determine the PK/PD indices of tedizolid action on VRE and the mechanisms underlying the PK/PD indices differences of tedizolid against VRE and methicillin-resistant Staphylococcus aureus (MRSA). METHODS Optimal PK/PD target values of tedizolid were determined in vitro, based on time-kill curves and post-antibiotic effects (PAEs), and in vivo, using mouse models of thigh infection with VRE and MRSA strains. RESULTS The tedizolid bactericidal activity on VRE and MRSA was time-dependent. Correlations were closest between fAUC24/MIC and the tedizolid PK/PD index against MRSA and VRE. To achieve 1 log10 kill tedizolid fAUC24/MIC in neutropenic mouse models of thigh infection with VRE and MRSA should be 14.2 and 138.5, respectively. The PAEs of tedizolid against VRE and MRSA were 2.39 and 0.99 h, respectively. CONCLUSION Tedizolid showed bactericidal effects against VRE even in neutropenic mice unlike MRSA, which could be attributed to its longer PAE against VRE. Hence, we hypothesize that tedizolid treatment against VRE infections is promising for achieving therapeutic success in clinical.
Collapse
Affiliation(s)
- Xiaoxi Liu
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, 1-5-30 Shiba Koen, Minato-Ku, Tokyo, 105-8512, Japan
| | - Sho Tashiro
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, 1-5-30 Shiba Koen, Minato-Ku, Tokyo, 105-8512, Japan
| | - Yuki Igarashi
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, 1-5-30 Shiba Koen, Minato-Ku, Tokyo, 105-8512, Japan
| | - Wataru Takemura
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, 1-5-30 Shiba Koen, Minato-Ku, Tokyo, 105-8512, Japan
| | - Nana Kojima
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, 1-5-30 Shiba Koen, Minato-Ku, Tokyo, 105-8512, Japan
| | - Takumi Morita
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, 1-5-30 Shiba Koen, Minato-Ku, Tokyo, 105-8512, Japan
| | - Marina Hayashi
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, 1-5-30 Shiba Koen, Minato-Ku, Tokyo, 105-8512, Japan
| | - Yuki Enoki
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, 1-5-30 Shiba Koen, Minato-Ku, Tokyo, 105-8512, Japan.
| | - Kazuaki Taguchi
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, 1-5-30 Shiba Koen, Minato-Ku, Tokyo, 105-8512, Japan
| | - Kazuaki Matsumoto
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, 1-5-30 Shiba Koen, Minato-Ku, Tokyo, 105-8512, Japan
| |
Collapse
|
49
|
Pharmacokinetics and Pharmacodynamics (PK/PD) of Corallopyronin A against Methicillin-Resistant Staphylococcus aureus. Pharmaceutics 2022; 15:pharmaceutics15010131. [PMID: 36678760 PMCID: PMC9860980 DOI: 10.3390/pharmaceutics15010131] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a World Health Organization’s high priority pathogen organism, with an estimated > 100,000 deaths worldwide in 2019. Thus, there is an unmet medical need for novel and resistance-breaking anti-infectives. The natural product Co-rallopyronin A (CorA), currently in preclinical development for filariasis, is efficacious against MRSA in vitro. In this study, we evaluated the pharmacokinetics of CorA after dosing in mice. Furthermore, we determined compound concentrations in target compartments, such as lung, kidney and thigh tissue, using LC-MS/MS. Based on the pharmacokinetic results, we evaluated the pharmacodynamic profile of CorA using the standard neutropenic thigh and lung infection models. We demonstrate that CorA is effective in both standard pharmacodynamic models. In addition to reaching effective levels in the lung and muscle, CorA was detected at high levels in the thigh bone. The data presented herein encourage the further exploration of the additional CorA indications treatment of MRSA- and methicillin-sensitive S. aureus- (MSSA) related infections.
Collapse
|
50
|
Yu D, Bio LL. Shedding Light on Amoxicillin, Amoxicillin-clavulanate, and Cephalexin Dosing in Children from a Pharmacist's Perspective. J Pediatric Infect Dis Soc 2022; 11:594-602. [PMID: 36112500 DOI: 10.1093/jpids/piac105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 09/15/2022] [Indexed: 12/30/2022]
Abstract
Selection of an antibiotic and dosing regimen requires consideration of multiple factors including microbiological data, site of infection, pharmacokinetics, and how it relates to the pharmacodynamic target. Given the multiple dosage regimens of amoxicillin with/without clavulanate and cephalexin, we review the principles of dose selection from a pharmacist's perspective.
Collapse
Affiliation(s)
- Diana Yu
- Department of Pharmacy, Doernbecher Children's Hospital, Oregon Health and Science University, Portland, Oregon, USA
| | - Laura L Bio
- Department of Pharmacy, Lucile Packard Children's Hospital Stanford, Palo Alto, California, USA
| |
Collapse
|