Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 161
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Pharmacokinet Pharmacodyn ; 51(1): 5-31, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37573528

RESUMEN

The current demand for pharmacometricians outmatches the supply provided by academic institutions and considerable investments are made to develop the competencies of these scientists on-the-job. Even with the observed increase in academic programs related to pharmacometrics, this need is unlikely to change in the foreseeable future, as the demand and scope of pharmacometrics applications keep expanding. Further, the field of pharmacometrics is changing. The field largely started when Lewis Sheiner and Stuart Beal published their seminal papers on population pharmacokinetics in the late 1970's and early 1980's and has continued to grow in impact and use since its inception. Physiological-based pharmacokinetics and systems pharmacology have grown rapidly in scope and impact in the last decade and machine learning is just on the horizon. While all these methodologies are categorized as pharmacometrics, no one person can be an expert in everything. So how do you train future pharmacometricians? Leading experts in academia, industry, contract research organizations, clinical medicine, and regulatory gave their opinions on how to best train future pharmacometricians. Their opinions were collected and synthesized to create some general recommendations.


Asunto(s)
Farmacología , Humanos , Farmacocinética , Selección de Profesión
2.
Antimicrob Agents Chemother ; 67(12): e0072723, 2023 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-37975660

RESUMEN

It is unclear whether plasma is a reliable surrogate for target attainment in the epithelial lining fluid (ELF). The objective of this study was to characterize meropenem target attainment in plasma and ELF using prospective samples. The first 24-hour T>MIC was evaluated vs 1xMIC and 4xMIC targets at the patient (i.e., fixed MIC of 2 mg/L) and population [i.e., cumulative fraction of response (CFR) according to EUCAST MIC distributions] levels for both plasma and ELF. Among 65 patients receiving ≥24 hours of treatment, 40% of patients failed to achieve >50% T>4xMIC in plasma and ELF, and 30% of patients who achieved >50% T>4xMIC in plasma had <50% T>4xMIC in ELF. At 1xMIC and 4xMIC targets, 3% and 25% of patients with >95% T>MIC in plasma had <50% T>MIC in ELF, respectively. Those with a CRCL >115 mL/min were less likely to achieve >50%T>4xMIC in ELF (P < 0.025). In the population, CFR for Escherichia coli at 1xMIC and 4xMIC was >97%. For Pseudomonas aeruginosa, CFR was ≥90% in plasma and ranged 80%-85% in ELF at 1xMIC when a loading dose was applied. CFR was reduced in plasma (range: 75%-81%) and ELF (range: 44%-60%) in the absence of a loading dose at 1xMIC. At 4xMIC, CFR for P. aeruginosa was 60%-86% with a loading dose and 18%-62% without a loading dose. We found that plasma overestimated ELF target attainment inup to 30% of meropenem-treated patients, CRCL >115 mL/min decreased target attainment in ELF, and loading doses increased CFR in the population.


Asunto(s)
Antibacterianos , Infecciones por Pseudomonas , Humanos , Meropenem/uso terapéutico , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Estudios Prospectivos , Infecciones por Pseudomonas/tratamiento farmacológico , Plasma , Pruebas de Sensibilidad Microbiana
3.
J Pediatr ; 259: 113419, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37044372

RESUMEN

OBJECTIVES: To evaluate implementation of rifamycin-based regimens (RBR) for pediatric tuberculosis infection (TBI) treatment among 3 provider settings in a high-incidence county. STUDY DESIGN: A multicenter, retrospective observational study was performed across 3 sites in Los Angeles County: an academic center (AC), a general pediatrics federally qualified health center (FQHC), and department of public health (DPH) tuberculosis clinics. Patients initiated on TBI treatment age 1 months to 17 years between 2018 and 2020 were included. RBRs were defined as regimens: 3 months of weekly rifapentine and isoniazid, 4 months of daily rifampin, and 3 months of daily isoniazid and rifampin. RESULTS: We included 424 patients: 51 from AC, 327 from DPH, and 46 from FQHC. RBR use nearly doubled during the study period (from 43% in 2018 to 82% in 2020; P < .001). FQHC had the shortest time to chest radiograph and treatment initiation; however, AC and DPH were 4 times as likely to prescribe an RBR compared to FQHC (95% CI, 2.1-7.8). AC and DPH had similar completion rates (74%) and were 2.6 times as likely to complete treatment compared to FQHC (95% CI, 1.4-4.9). CONCLUSIONS: The use of RBRs for pediatric TBI varies significantly by clinical setting but is improving over time. Strategies are needed to improve RBR uptake, standardize care, and increase treatment completion, particularly among general pediatricians.


Asunto(s)
Tuberculosis Latente , Pediatría , Tuberculosis , Humanos , Niño , Lactante , Rifampin/uso terapéutico , Isoniazida/uso terapéutico , Antituberculosos/uso terapéutico , Tuberculosis/tratamiento farmacológico , Quimioterapia Combinada
4.
Antimicrob Agents Chemother ; 66(10): e0069522, 2022 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-36165631

RESUMEN

Mycobacterium tuberculosis (Mtb) exists in various metabolic states, including a nonreplicating persister (NRP) phenotype which may affect response to therapy. We have adopted a model-informed strategy to accelerate discovery of effective Mtb treatment regimens and previously found pretomanid (PMD), moxifloxacin (MXF), and bedaquiline (BDQ) to readily kill logarithmic- and acid-phase Mtb. Here, we studied multiple concentrations of each drug in flask-based, time-kill studies against NRP Mtb in single-, two- and three-drug combinations, including the active M2 metabolite of BDQ. We used nonparametric population algorithms in the Pmetrics package for R to model the data and to simulate the 95% confidence interval of bacterial population decline due to the two-drug combination regimen of PMD + MXF and compared this to observed declines with three-drug regimens. PMD + MXF at concentrations equivalent to average or peak human concentrations effectively eradicated Mtb. Unlike other states for Mtb, we observed no sustained emergence of less susceptible isolates for any regimen. The addition of BDQ as a third drug significantly (P < 0.05) shortened time to total bacterial suppression by 3 days compared to the two-drug regimen, similar to our findings for Mtb in logarithmic or acid growth phases.


Asunto(s)
Mycobacterium tuberculosis , Animales , Humanos , Antituberculosos/farmacología , Moxifloxacino/farmacología , Combinación de Medicamentos , Fenotipo
5.
Antimicrob Agents Chemother ; 66(2): e0204621, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-34902271

RESUMEN

Cefepime is the second most common cephalosporin used in U.S. hospitals. We aim to develop and validate a cefepime population pharmacokinetic (PK) model and integrate it into a precision dosing tool for implementation. Two data sets (680 patients) were used to build the cefepime PK model in Pmetrics, and three data sets (34 patients) were used for the validation. A separate application data set (115 patients) was used for the implementation and validation of a precision dosing tool. The model support points and covariates were used to generate the optimal initial dose (OID). Cefepime PK was described by a two-compartment model including weight and creatinine clearance (CrCl) as covariates. The median rate of elimination was 0.30 h-1 (adults) and 0.96 h-1 (children), the central volume of distribution was 13.85 L, and the rate of transfer from the central to the peripheral compartments was 1.22 h-1 and from the peripheral to the central compartments was 1.38 h-1. After integration in BestDose, the observed versus predicted cefepime concentration fit using the application data set was excellent (R2 > 0.98), and the median difference between what was observed and what BestDose predicted on a second occasion was 4%. For the OID, cefepime at a 0.5- to 1-g 4-h infusion every 8 to 24 h (q8 to 24 h) with a CrCl of <70 mL/min was needed to achieve a target range of free trough:MIC 1 to 4 at a MIC of 8 mg/L, while continuous infusion was needed for higher CrCl and weight values. In conclusion, we developed and validated a cefepime model for clinical application. The model was integrated in a precision dosing tool for implementation, and the median concentration prediction bias was 4%. The OID algorithm was provided.


Asunto(s)
Antibacterianos , Cefalosporinas , Adulto , Antibacterianos/farmacocinética , Cefepima/farmacocinética , Cefalosporinas/farmacocinética , Niño , Humanos , Pruebas de Sensibilidad Microbiana
6.
J Antimicrob Chemother ; 77(8): 2209-2216, 2022 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-35696555

RESUMEN

BACKGROUND: Echinocandins are commonly used in treatment and prophylaxis of invasive fungal diseases. Intravenous daily dosing for prophylaxis in the outpatient setting can however become a hurdle for adequate compliance in the paediatric population. OBJECTIVES: Simulations were performed to assess extended twice-weekly dosing for antifungal prophylaxis using caspofungin. METHODS: A population pharmacokinetic model was developed based on previously published data from children aged 3 months to 17 years. Using the final model, Monte Carlo simulations were performed to assess the dose needed for adequate exposure in a twice-weekly setting. Mean weekly AUC0-24 h/MIC together with reported AUC0-24 h from previously reported paediatric trials were used to guide adequate exposure. RESULTS AND CONCLUSIONS: A two-compartment model with linear elimination and allometric scaling using fixed exponents was found most adequate to describe the given paediatric populations. Simulations showed that a 200 mg/m2 twice-weekly regimen with maximal 200 mg total dose should result in exposures matching registered daily dosing as well as commonly used pharmacokinetic/pharmacodynamic targets.


Asunto(s)
Antifúngicos , Infecciones Fúngicas Invasoras , Antifúngicos/farmacología , Caspofungina , Niño , Equinocandinas , Humanos , Infecciones Fúngicas Invasoras/tratamiento farmacológico , Método de Montecarlo
7.
J Antimicrob Chemother ; 77(5): 1334-1343, 2022 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-35170719

RESUMEN

BACKGROUND: Neonatal sepsis is a serious bacterial infection of neonates, globally killing up to 680 000 babies annually. It is frequently complicated by antimicrobial resistance, particularly in low- and middle-income country (LMIC) settings with widespread resistance to the WHO's recommended empirical regimen of ampicillin and gentamicin. OBJECTIVES: We assessed the utility of flomoxef and fosfomycin as a potential alternative empirical regimen for neonatal sepsis in these settings. METHODS: We studied the combination in a 16-arm dose-ranged hollow-fibre infection model (HFIM) experiment and chequerboard assays. We further assessed the combination using clinically relevant regimens in the HFIM with six Enterobacterales strains with a range of flomoxef/fosfomycin MICs. RESULTS: Pharmacokinetic/pharmacodynamic modelling of the HFIM experimental output, along with data from chequerboard assays, indicated synergy of this regimen in terms of bacterial killing and prevention of emergence of fosfomycin resistance. Flomoxef monotherapy was sufficient to kill 3/3 strains with flomoxef MICs ≤0.5 mg/L to sterility. Three of three strains with flomoxef MICs ≥8 mg/L were not killed by fosfomycin or flomoxef monotherapy; 2/3 of these were killed with the combination of the two agents. CONCLUSIONS: These data suggest that flomoxef/fosfomycin could be an efficacious and synergistic regimen for the empirical treatment of neonatal sepsis in LMIC settings with prevalent antimicrobial resistance. Our HFIM results warrant further assessment of the flomoxef/fosfomycin combination in clinical trials.


Asunto(s)
Fosfomicina , Sepsis Neonatal , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Cefalosporinas , Farmacorresistencia Bacteriana , Fosfomicina/farmacología , Fosfomicina/uso terapéutico , Humanos , Recién Nacido , Pruebas de Sensibilidad Microbiana , Sepsis Neonatal/tratamiento farmacológico
8.
J Antimicrob Chemother ; 77(12): 3349-3357, 2022 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-36177766

RESUMEN

BACKGROUND: Annual mortality from neonatal sepsis is an estimated 430 000-680 000 infants globally, most of which occur in low- and middle-income countries (LMICs). The WHO currently recommends a narrow-spectrum ß-lactam (e.g. ampicillin) and gentamicin as first-line empirical therapy. However, available epidemiological data demonstrate high rates of resistance to both agents. Alternative empirical regimens are needed. Flomoxef and amikacin are two off-patent antibiotics with potential for use in this setting. OBJECTIVES: To assess the pharmacodynamics of flomoxef and amikacin in combination. METHODS: The pharmacodynamic interaction of flomoxef and amikacin was assessed in chequerboard assays and a 16-arm dose-ranged hollow-fibre infection model (HFIM) experiment. The combination was further assessed in HFIM experiments mimicking neonatal plasma exposures of clinically relevant doses of both drugs against five Enterobacterales isolates with a range of flomoxef/amikacin MICs. RESULTS: Flomoxef and amikacin in combination were synergistic in bacterial killing in both assays and prevention of emergence of amikacin resistance in the HFIM. In the HFIM assessing neonatal-like drug exposures, the combination killed 3/5 strains to sterility, (including 2/5 that monotherapy with either drug failed to kill) and failed to kill the 2/5 strains with flomoxef MICs of 32 mg/L. CONCLUSIONS: We conclude that the combination of flomoxef and amikacin is synergistic and is a potentially clinically effective regimen for the empirical treatment of neonatal sepsis in LMIC settings and is therefore suitable for further assessment in a clinical trial.


Asunto(s)
Amicacina , Sepsis Neonatal , Lactante , Recién Nacido , Humanos , Amicacina/farmacología , Amicacina/uso terapéutico , Sepsis Neonatal/tratamiento farmacológico , Cefalosporinas , Pruebas de Sensibilidad Microbiana , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Atención a la Salud
9.
J Antimicrob Chemother ; 77(11): 2956-2959, 2022 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-35869779

RESUMEN

OBJECTIVES: Critical illness reduces ß-lactam pharmacokinetic/pharmacodynamic (PK/PD) attainment. We sought to quantify PK/PD attainment in patients with hospital-acquired pneumonia. METHODS: Meropenem plasma PK data (n = 70 patients) were modelled, PK/PD attainment rates were calculated for empirical and definitive targets, and between-patient variability was quantified [as a coefficient of variation (CV%)]. RESULTS: Attainment of 100% T>4×MIC was variable for both empirical (CV% = 92) and directed (CV% = 33%) treatment. CONCLUSIONS: Individualization is required to achieve suggested PK/PD targets in critically ill patients.


Asunto(s)
Antibacterianos , Neumonía , Humanos , Meropenem/uso terapéutico , Meropenem/farmacología , Pruebas de Sensibilidad Microbiana , Antibacterianos/uso terapéutico , Antibacterianos/farmacología , Estudios Prospectivos , Enfermedad Crítica/terapia , Unidades de Cuidados Intensivos , Neumonía/tratamiento farmacológico , Hospitales
10.
Br J Clin Pharmacol ; 2022 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-36482842

RESUMEN

Patients are often switched between generic formulations of the same drug, but in some cases generic interchangeability is questioned. For generic drugs to be approved, bioequivalence with the innovator drug should be demonstrated, but evidence of bioequivalence is not required in the intended patient population or relative to other approved generics. AIM: We aim to identify pathophysiological pharmacokinetic subpopulations for whom there is a difference in comparative bioavailability compared to a healthy population. METHODS: We used simulated exposures from a nonparametric model of multiple generics and the originator gabapentin. Exposure was simulated for virtual populations with pharmacokinetic characteristics beyond those of healthy subjects with regard to rate of absorption, volume of distribution and reduced renal function. Virtual parallel design bioequivalence studies were performed using a random sample of 24 simulated subjects, with standard acceptance criteria. RESULTS: Results indicated increased pharmacokinetic variability for patient populations with a lower rate of absorption or a reduced renal function, but no change in the average comparable bioavailability ratio. This increased variability results in a reduced likelihood of demonstrating bioequivalence. Observations were similar for comparisons between all different formulations, as well as between subjects who received the identical formulation in a repeated fashion. No relevant effect was observed for simulations with increased volume of distribution. CONCLUSION: Our simulations indicate that the reduced likelihood of demonstrating bioequivalence for subjects with altered pharmacokinetics is not influenced by a formulation switch, nor does the average comparable bioavailability ratio change, therefore these results support generic interchangeability and current approval requirements for generics.

11.
Ther Drug Monit ; 44(1): 121-132, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34882107

RESUMEN

ABSTRACT: Individualization of vancomycin dosing based on therapeutic drug monitoring (TDM) data is known to improve patient outcomes compared with fixed or empirical dosing strategies. There is increasing evidence to support area-under-the-curve (AUC24)-guided TDM to inform vancomycin dosing decisions for patients receiving therapy for more than 48 hours. It is acknowledged that there may be institutional barriers to the implementation of AUC24-guided dosing, and additional effort is required to enable the transition from trough-based to AUC24-based strategies. Adequate documentation of sampling, correct storage and transport, accurate laboratory analysis, and pertinent data reporting are required to ensure appropriate interpretation of TDM data to guide vancomycin dosing recommendations. Ultimately, TDM data in the clinical context of the patient and their response to treatment should guide vancomycin therapy. Endorsed by the International Association of Therapeutic Drug Monitoring and Clinical Toxicology, the IATDMCT Anti-Infectives Committee, provides recommendations with respect to best clinical practice for vancomycin TDM.


Asunto(s)
Antiinfecciosos , Vancomicina , Antibacterianos , Área Bajo la Curva , Monitoreo de Drogas/métodos , Humanos
12.
J Community Health ; 47(1): 39-52, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34297272

RESUMEN

Childhood vaccine hesitancy has been studied extensively before the COVID-19 pandemic. The pandemic presented new barriers to pediatric vaccinations. Furthermore, the development of COVID-19 vaccines has complicated factors underlying vaccine hesitancy. We performed a cross-sectional mobile phone-based survey at Children's Hospital Los Angeles querying parents regarding perspectives on vaccines before and during the pandemic. Our primary aim was to understand the impact of the pandemic on routine childhood vaccine hesitancy. Secondarily, we examined intent to vaccinate, COVID-19 vaccine hesitancy, and key contributing demographic factors. Among 252 participants, we found overall increased childhood vaccine hesitancy (p = 0.006), increased risk perception (p = 0.006), and unchanged vaccine confidence during the COVID-19 pandemic. Increased hesitancy did not translate into decreased intent to vaccinate with routine childhood vaccines or influenza vaccines. During the pandemic, households with higher income (50-99 K, > 100 K) correlated with decreased routine childhood vaccine hesitancy, while Hispanic ethnicity and African American race had increased risk perception. For COVID-19 vaccine hesitancy, households with higher income (> 100 K) correlated with decreased hesitancy, while non-White ethnicity and race had increased risk perception. We found that routine childhood vaccine hesitancy increased during the COVID-19 pandemic, mainly due to increased risk perception. Key contributing demographic factors behind both childhood vaccine hesitancy and COVID-19 vaccine hesitancy included household income and race. Understanding factors behind routine childhood vaccine hesitancy is crucial to maintaining pediatric vaccination rates and promoting vaccine confidence during and after the COVID-19 pandemic.


Asunto(s)
COVID-19 , Vacunas contra la Influenza , Vacunas contra la COVID-19 , Niño , Estudios Transversales , Humanos , Pandemias , Padres , SARS-CoV-2 , Vacunación , Vacilación a la Vacunación
13.
Artículo en Inglés | MEDLINE | ID: mdl-33468465

RESUMEN

The repurposed agent moxifloxacin has become an important addition to the physician's armamentarium for the therapy of Mycobacterium tuberculosis When a drug is administered, we need to have metrics for success. As for most antimicrobial chemotherapy, we contend that for Mycobacterium tuberculosis therapy, these metrics should be a decline in the susceptible bacterial burden and the suppression of amplification of less-susceptible populations. To achieve optimal outcomes relative to these metrics, a dose and schedule of administration need to be chosen. For large populations of patients, there are true between-patient differences in important pharmacokinetic parameters. These distributions of parameter values may have an impact on these metrics, depending on what measure of drug exposure drives the metrics. To optimize dose and schedule choice of moxifloxacin, we performed a dose fractionation experiment in the hollow fiber infection model. We examined 12-, 24-, and 48-h dosing intervals with doses of 200, 400, and 800 mg for each interval, respectively. Within each interval, we had an arm where half-lives of 12, 8, and 4 h were simulated. We attempted to keep the average concentration (Cavg) or area under the concentration-time curve (AUC) constant across arms. We found that susceptible bacterial load decline was linked to Cavg, as we had indicated previously. Resistance suppression, a nonmonotonic function, had minimum concentration (Cmin) as the linked index. The 48-h interval with the 4-h half-life had the largest less-susceptible population. Balancing bacterial kill, resistance suppression, toxicity (linked to peak concentration [Cpeak]), and adherence, we conclude that the dose of 400 mg daily is optimal for moxifloxacin.


Asunto(s)
Antituberculosos , Tuberculosis , Antituberculosos/uso terapéutico , Área Bajo la Curva , Fluoroquinolonas , Semivida , Humanos , Pruebas de Sensibilidad Microbiana , Moxifloxacino , Tuberculosis/tratamiento farmacológico
14.
Artículo en Inglés | MEDLINE | ID: mdl-33318004

RESUMEN

Vancomycin induces exposure-related acute kidney injury. However, the pharmacokinetic-toxicodynamic (PK-TD) relationship remains unclear. Sprague-Dawley rats received intravenous (i.v.) vancomycin doses of 300 mg/kg/day and 400 mg/kg/day, divided into once-, twice-, three-times-, or four-times-daily doses (i.e., QD, BID, TID, or QID) over 24 h. Up to 8 samples plus a terminal sample were drawn during the 24-h dosing period. Twenty-four-hour urine was collected and assayed for kidney injury molecule-1 (KIM-1). Vancomycin was quantified via liquid chromatography-tandem mass spectrometry (LC-MS/MS). Following terminal sampling, nephrectomy and histopathologic analyses were conducted. PK analyses were conducted using Pmetrics. PK exposures (i.e., area under the concentration-time curve from 0 to 24 h [AUC0-24] and maximum concentration from 0 to 24 h [Cmax0-24]) were calculated for each rat, and PK-TD relationships were discerned. A total of 53-rats generated PK-TD data. A 2-compartment model fit the data well (Bayesian observed versus predicted concentrations; R2 = 0.96). KIM-1 values were greater in QD and BID groups (P for QD versus TID, <0.002; P for QD versus QID, <0.004; P for BID versus TID, <0.002; and P for BID versus QID, <0.004). Exposure-response relationships were observed between KIM-1 versus Cmax0-24 and AUC0-24 (R2 = 0.7 and 0.68). Corrected Akaike's information criterion showed Cmax0-24 as the most predictive PK-TD driver for vancomycin-induced kidney injury (VIKI) (-5.28 versus -1.95). While PK-TD indices are often intercorrelated, maximal concentrations and fewer doses (for the same total daily amount) resulted in increased VIKI in our rat model.


Asunto(s)
Espectrometría de Masas en Tándem , Vancomicina , Animales , Área Bajo la Curva , Teorema de Bayes , Cromatografía Liquida , Riñón , Ratas , Ratas Sprague-Dawley , Vancomicina/efectos adversos
15.
Antimicrob Agents Chemother ; 65(10): e0069321, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34339275

RESUMEN

Mycobacterium tuberculosis metabolic state affects the response to therapy. Quantifying the effect of antimicrobials in the acid and nonreplicating metabolic phases of M. tuberculosis growth will help to optimize therapy for tuberculosis. As a brute-force approach to all possible drug combinations against M. tuberculosis in all different metabolic states is impossible, we have adopted a model-informed strategy to accelerate the discovery. Using multiple concentrations of each drug in time-kill studies, we examined single drugs and two- and three-drug combinations of pretomanid, moxifloxacin, and bedaquiline plus its active metabolite against M. tuberculosis in its acid-phase metabolic state. We used a nonparametric modeling approach to generate full distributions of interaction terms between pretomanid and moxifloxacin for susceptible and less susceptible populations. From the model, we could predict the 95% confidence interval of the simulated total bacterial population decline due to the 2-drug combination regimen of pretomanid and moxifloxacin and compare this to observed declines with 3-drug regimens. We found that the combination of pretomanid and moxifloxacin at concentrations equivalent to average or peak human concentrations effectively eradicated M. tuberculosis in its acid growth phase and prevented emergence of less susceptible isolates. The addition of bedaquiline as a third drug shortened time to total and less susceptible bacterial suppression by 8 days compared to the 2-drug regimen, which was significantly faster than the 2-drug kill.


Asunto(s)
Mycobacterium tuberculosis , Animales , Antituberculosos/uso terapéutico , Combinación de Medicamentos , Quimioterapia Combinada , Humanos , Moxifloxacino
16.
Artículo en Inglés | MEDLINE | ID: mdl-33199386

RESUMEN

The Mycobacterium tuberculosis drug discovery effort has generated a substantial number of new/repurposed drugs for therapy for this pathogen. The arrival of these drugs is welcome, but another layer of difficulty has emerged. Single agent therapy is insufficient for patients with late-stage tuberculosis because of resistance emergence. To achieve our therapeutic ends, it is requisite to identify optimal combination regimens. These regimens go through a lengthy and expensive evaluative process. If we have a modest group of 6 to 8 new or repurposed agents, this translates into 15 to 28 possible 2-drug combinations. There is neither time nor resources to give an extensive evaluation for all combinations. We sought a screening procedure that would identify combinations that had a high likelihood of achieving good bacterial burden decline. We examined pretomanid, moxifloxacin, linezolid, and bedaquiline in log-phase growth, acid-phase growth, and nonreplicative persister (NRP) phase in the Greco interaction model. We employed the interaction term α and the calculated bacterial burden decline as metrics to rank different regimens in different metabolic states. No relationship was found between α and bacterial kill. We chose bacterial kill as the prime metric. The combination of pretomanid plus moxifloxacin emerged as the clear frontrunner, as the largest bacterial declines were seen in log phase and acid phase with this regimen and it was second best in NRP phase. Bedaquiline also produced good kill. This screening process may identify optimal combinations that can be further evaluated in both the hollow-fiber infection model and in animal models of Mycobacterium tuberculosis infection.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Animales , Antituberculosos/farmacología , Antituberculosos/uso terapéutico , Combinación de Medicamentos , Quimioterapia Combinada , Humanos , Tuberculosis/tratamiento farmacológico
17.
Antimicrob Agents Chemother ; 65(10): e0106021, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34339278

RESUMEN

Vancomycin area under the concentration curve (AUC) is known to predict vancomycin-induced acute kidney injury (AKI). Data were analyzed from a rat model (n = 48) and two prospective clinical studies (PROVIDE [n = 263] and CAMERA2 [n = 291]). A logit-link model was used to calculate the multiplicative factors between the probability of AKI from clinical studies and in the rat. The rat was 2.7 to 4.2 times more sensitive to AKI between AUCs of 199.5 and 794.3 mg·h/liter, respectively.


Asunto(s)
Lesión Renal Aguda , Vancomicina , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/tratamiento farmacológico , Animales , Antibacterianos/efectos adversos , Área Bajo la Curva , Estudios Prospectivos , Ratas , Vancomicina/efectos adversos
18.
Antimicrob Agents Chemother ; 65(7): e0029321, 2021 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-33972238

RESUMEN

Antimicrobial resistance (particularly through extended-spectrum ß-lactamase and aminoglycoside-modifying enzyme production) in neonatal sepsis is a global problem, particularly in low- and middle-income countries, with significant mortality rates. High rates of resistance are reported for the current WHO-recommended first-line antibiotic regimen for neonatal sepsis, i.e., ampicillin and gentamicin. We assessed the utility of fosfomycin and amikacin as a potential alternative regimen to be used in settings of increasingly prevalent antimicrobial resistance. The combination was studied in a 16-arm dose-ranged hollow-fiber infection model (HFIM) experiment. The combination of amikacin and fosfomycin enhanced bactericidal activity and prevented the emergence of resistance, compared to monotherapy with either antibiotic. Modeling of the experimental quantitative outputs and data from checkerboard assays indicated synergy. We further assessed the combination regimen at clinically relevant doses in the HFIM with nine Enterobacterales strains with high fosfomycin and amikacin MICs and demonstrated successful kill to sterilization for 6/9 strains. From these data, we propose a novel combination breakpoint threshold for microbiological success for this antimicrobial combination against Enterobacterales strains, i.e., MICF × MICA < 256 (where MICF and MICA are the fosfomycin and amikacin MICs, respectively). Monte Carlo simulations predict that a standard fosfomycin-amikacin neonatal regimen would achieve >99% probability of pharmacodynamic success for strains with MICs below this threshold. We conclude that the combination of fosfomycin with amikacin is a viable regimen for the empirical treatment of neonatal sepsis and is suitable for further clinical assessment in a randomized controlled trial.


Asunto(s)
Antibacterianos , Fosfomicina , Sepsis Neonatal , Amicacina , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Farmacorresistencia Bacteriana , Fosfomicina/farmacología , Fosfomicina/uso terapéutico , Humanos , Recién Nacido , Pruebas de Sensibilidad Microbiana , Sepsis Neonatal/tratamiento farmacológico
19.
Antimicrob Agents Chemother ; 65(8): e0029021, 2021 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-34031051

RESUMEN

Isavuconazole, administered as the water-soluble prodrug isavuconazonium sulfate, is a new triazole agent used to treat invasive fungal infections. This phase 1 study evaluated the pharmacokinetics (PK), safety, and tolerability of isavuconazole in 46 immunocompromised pediatric patients, stratified by age (1 to <6 [intravenous (i.v.) only], 6 to <12, and 12 to <18 years), receiving 10 mg/kg body weight (maximum, 372 mg) isavuconazonium sulfate either i.v. or orally. A population PK model using weight-based allometric scaling was constructed with the pediatric i.v. and oral data plus i.v. data from a phase 1 study in adults. The best model was a 3-compartment model with combined zero-order and first-order input, with linear elimination. Stepwise covariate modeling was performed in Perl-speaks-NONMEM version 4.7.0. None of the covariates examined, including age, sex, race, and body mass index, were statistically significant for any of the PK parameters. The area under the concentration-time curve at steady state (AUCSS) was predicted for pediatric patients using 1,000 Monte Carlo simulations per age cohort for each administration route. The probability of target attainment (AUCSS range, 60 to 233 µg · h/ml) was estimated; this target range was derived from plasma drug exposures in adults receiving the recommended clinical dose. Predicted plasma drug exposures were within the target range for >80% and >76% of simulated pediatric patients following i.v. or oral administration, respectively. Intravenous and oral administration of isavuconazonium sulfate at the studied dosage of 10 mg/kg was well tolerated and resulted in exposure in pediatric patients similar to that in adults. (This study has been registered at ClinicalTrials.gov under identifier NCT03241550).


Asunto(s)
Infecciones Fúngicas Invasoras , Triazoles , Administración Oral , Adolescente , Niño , Preescolar , Humanos , Lactante , Infecciones Fúngicas Invasoras/tratamiento farmacológico , Nitrilos/uso terapéutico , Piridinas/efectos adversos , Triazoles/uso terapéutico
20.
J Antimicrob Chemother ; 76(7): 1661-1665, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-33843994

RESUMEN

The need for precision dosing has been challenged on the basis of insufficient evidence. Herein, we argue that adequate evidence exists to conduct therapeutic drug monitoring (TDM) and precisely target antibiotic exposures. While achievement of any antibiotic concentration does not guarantee efficacy sans toxicity for any single patient, stochastic control optimizes the probability of achieving favourable responses across patients. We argue that variability in targets (such as the organism's MIC) can be considered with models. That is, complexity alone does not relegate the decision-making framework to 'clinician intuition'. We acknowledge the exposure-response relationships are modified by patient-specific factors (other drugs, baseline organ functional status etc.) and describe how precision dosing can inform clinical decision making rather than protocolize it. Finally, we call for randomized, controlled trials; however, we suggest that these trials are not necessary to make TDM standard of care for multiple classes of antibiotics.


Asunto(s)
Antibacterianos , Monitoreo de Drogas , Antibacterianos/uso terapéutico , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA