Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Clin Infect Dis ; 72(12): e1103-e1111, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-33393598

RESUMEN

In November 2019, the Food and Drug Administration (FDA) approved cefiderocol for the treatment of complicated urinary tract infections (cUTI) including pyelonephritis caused by susceptible gram-negative bacteria in adults with limited to no alternative treatment options based on a randomized, double-blind, noninferiority cUTI trial (APEKS-cUTI). In a randomized, open-label trial (CREDIBLE-CR) in patients with cUTI, nosocomial pneumonia, bloodstream infections, or sepsis due to carbapenem-resistant gram-negative bacteria, an increase in all-cause mortality was observed in patients treated with cefiderocol as compared to best available therapy. The cause of the increased mortality was not established, but some deaths were attributed to treatment failure. Preliminary data from a randomized, double-blind trial (APEKS-NP) in patients with nosocomial pneumonia due to carbapenem-susceptible gram-negative bacteria showed a similar rate of mortality as compared to meropenem. We describe the uncertainties and challenges in the interpretation of the CREDIBLE-CR trial and some benefit-risk considerations for the use of cefiderocol in clinical practice. Clinical Trials Registration: NCT02321800.


Asunto(s)
Antibacterianos , Cefalosporinas , Adulto , Antibacterianos/uso terapéutico , Bacterias Gramnegativas , Humanos , Estados Unidos , United States Food and Drug Administration , Cefiderocol
2.
Artículo en Inglés | MEDLINE | ID: mdl-32122895

RESUMEN

Animal models of bacterial infection have been widely used to explore the in vivo activity of antibacterial drugs. These data are often submitted to the U.S. Food and Drug Administration to support human use in an investigational new drug application (IND). To better understand the range and scientific use of animal models in regulatory submissions, a database was created surveying recent pneumonia models submitted as part of IND application packages. The IND studies were compared to animal models of bacterial pneumonia published in the scientific literature over the same period of time. In this review, we analyze the key experimental design elements, such as animal species, immune status, pathogens selected, and route of administration, and study endpoints.


Asunto(s)
Antituberculosos/farmacología , Modelos Animales de Enfermedad , Drogas en Investigación , Neumonía Bacteriana/tratamiento farmacológico , Neumonía Bacteriana/microbiología , Animales , Antituberculosos/uso terapéutico , Bases de Datos Factuales , Humanos , Aplicación de Nuevas Drogas en Investigación , Estados Unidos , United States Food and Drug Administration
3.
Artículo en Inglés | MEDLINE | ID: mdl-33106262

RESUMEN

The U.S. Food and Drug Administration (FDA) hosted a public workshop entitled "Advancing Animal Models for Antibacterial Drug Development" on 5 March 2020. The workshop mainly focused on models of pneumonia caused by Pseudomonas aeruginosa and Acinetobacter baumannii The program included discussions from academic investigators, industry, and U.S. government scientists. The potential use of mouse, rabbit, and pig models for antibacterial drug development was presented and discussed.


Asunto(s)
Acinetobacter baumannii , Antibacterianos , Animales , Antibacterianos/uso terapéutico , Desarrollo de Medicamentos , Ratones , Modelos Animales , Conejos , Porcinos , Estados Unidos , United States Food and Drug Administration
4.
Antimicrob Agents Chemother ; 59(9): 5768-74, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26169396

RESUMEN

Information about intralesional pharmacokinetics (PK) and spatial distribution of tuberculosis (TB) drugs is limited and has not been used to optimize dosing recommendations for new or existing drugs. While new techniques can detect drugs and their metabolites within TB granulomas, they are invasive, rely on accurate resection of tissues, and do not capture dynamic drug distribution in the tissues of interest. In this study, we assessed the in situ distribution of (11)C-labeled rifampin in live, Mycobacterium tuberculosis-infected mice that develop necrotic lesions akin to human disease. Dynamic positron emission tomography (PET) imaging was performed over 60 min after injection of [(11)C]rifampin as a microdose, standardized uptake values (SUV) were calculated, and noncompartmental analysis was used to estimate PK parameters in compartments of interest. [(11)C]rifampin was rapidly distributed to all parts of the body and quickly localized to the liver. Areas under the concentration-time curve for the first 60 min (AUC0-60) in infected and uninfected mice were similar for liver, blood, and brain compartments (P > 0.53) and were uniformly low in brain (10 to 20% of blood values). However, lower concentrations were noted in necrotic lung tissues of infected mice than in healthy lungs (P = 0.03). Ex vivo two-dimensional matrix-assisted laser desorption ionization (MALDI) imaging confirmed restricted penetration of rifampin into necrotic lung lesions. Noninvasive bioimaging can be used to assess the distribution of drugs into compartments of interest, with potential applications for TB drug regimen development.


Asunto(s)
Antituberculosos/farmacocinética , Mycobacterium tuberculosis/patogenicidad , Rifampin/farmacocinética , Animales , Femenino , Ratones , Tomografía de Emisión de Positrones , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Tuberculosis/metabolismo , Tuberculosis/microbiología
6.
Clin Pharmacol Ther ; 112(5): 1004-1007, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35792715

RESUMEN

In this report, we describe our scientific approach for including effluent flow rate (QE )-based dosing recommendations of cefiderocol for patients receiving continuous renal replacement therapy (CRRT) in the product labeling. The total clearance (CL) of cefiderocol in patients receiving CRRT was estimated as the sum of patients' nonrenal clearance (CLnonrenal ) and extracorporeal clearance by CRRT (CLCRRT ), based on the following rationale: (a) The renal clearance (CLrenal ) of cefiderocol is assumed to be negligible in patients receiving CRRT, (b) CLnonrenal represents the CRRT patients' own remaining systemic clearance and is estimated from the observed clearance in participants with creatinine clearance (CLcr) < 15 mL/minute without undergoing hemodialysis, and (c) CLCRRT was estimated by the product of unbound (free) fraction of plasma drug concentration (fu ) and QE because the free fraction of low-molecular-weight compounds like cefiderocol (752 Da) can be completely filtered by CRRT, regardless of CRRT modality. Hence, cefiderocol CL in CRRT patients was calculated by the equation of CL = CLnonrenal + fu × QE . Accordingly, the cefiderocol dosing regimens for patients receiving CRRT in clinically relevant ranges of QE were determined with the goal of achieving an average daily area under the concentration-time curve (AUC) observed in patients not receiving CRRT. Subsequently, pharmacokinetic (PK) simulations demonstrated that cefiderocol PK profiles following the QE -based dosing in patients receiving CRRT would be similar to those in patients not receiving CRRT.


Asunto(s)
Terapia de Reemplazo Renal Continuo , Humanos , Creatinina , Antibacterianos , Enfermedad Crítica/terapia , Terapia de Reemplazo Renal , Cefiderocol
7.
ACS Infect Dis ; 4(11): 1635-1644, 2018 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-30067329

RESUMEN

Staphylococcus aureus is the leading cause of life-threatening infections, frequently originating from unknown or deep-seated foci. Source control and institution of appropriate antibiotics remain challenges, especially with infections due to methicillin-resistant S. aureus (MRSA). In this study, we developed a radiofluorinated analog of para-aminobenzoic acid (2-[18F]F-PABA) and demonstrate that it is an efficient alternative substrate for the S. aureus dihydropteroate synthase (DHPS). 2-[18F]F-PABA rapidly accumulated in vitro within laboratory and clinical (including MRSA) strains of S. aureus but not in mammalian cells. Biodistribution in murine and rat models demonstrated localization at infection sites and rapid renal elimination. In a rat model, 2-[18F]F-PABA positron emission tomography (PET) rapidly differentiated S. aureus infection from sterile inflammation and could also detect therapeutic failures associated with MRSA. These data suggest that 2-[18F]F-PABA has the potential for translation to humans as a rapid, noninvasive diagnostic tool to identify, localize, and monitor S. aureus infections.


Asunto(s)
Ácido 4-Aminobenzoico/farmacología , Staphylococcus aureus Resistente a Meticilina/aislamiento & purificación , Tomografía Computarizada por Tomografía de Emisión de Positrones , Infecciones Estafilocócicas/diagnóstico por imagen , Infecciones Estafilocócicas/diagnóstico , Animales , Infección Hospitalaria/diagnóstico , Infección Hospitalaria/diagnóstico por imagen , Infección Hospitalaria/microbiología , Femenino , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Ratones , Ratones Endogámicos CBA , Ratas , Ratas Sprague-Dawley
8.
J Nucl Med ; 58(1): 144-150, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27635025

RESUMEN

The modern patient is increasingly susceptible to bacterial infections including those due to multidrug-resistant organisms (MDROs). Noninvasive whole-body analysis with pathogen-specific imaging technologies can significantly improve patient outcomes by rapidly identifying a source of infection and monitoring the response to treatment, but no such technology exists clinically. METHODS: We systematically screened 961 random radiolabeled molecules in silico as substrates for essential metabolic pathways in bacteria, followed by in vitro uptake in representative bacteria-Staphylococcus aureus, Escherichia coli, Pseudomonas aeruginosa, and mycobacteria. Fluorine-labeled analogs, that could be developed as PET-based imaging tracers, were evaluated in a murine myositis model. RESULTS: We identified 3 novel, nontoxic molecules demonstrating selective bacterial uptake: para-aminobenzoic acid (PABA), with uptake in all representative bacteria including Mycobacterium tuberculosis; mannitol, with selective uptake in S. aureus and E. coli; and sorbitol, accumulating only in E. coli None accumulated in mammalian cells or heat-killed bacteria, suggesting metabolism-derived specificity. In addition to an extended bacterial panel of laboratory strains, all 3 molecules rapidly accumulated in respective clinical isolates of interest including MDROs such as methicillin-resistant S. aureus, extended-spectrum ß-lactamase-producing, and carbapenem-resistant Enterobacteriaceae. In a murine myositis model, fluorine-labeled analogs of all 3 molecules could rapidly detect and differentiate infection sites from sterile inflammation in mice (P = 0.03). Finally, 2-deoxy-2-[F-18]fluoro-d-sorbitol (18F-FDS) can be easily synthesized from 18F-FDG. PET, with 18F-FDS synthesized using current good manufacturing practice, could rapidly differentiate true infection from sterile inflammation to selectively localize E. coli infection in mice. CONCLUSION: We have developed a systematic approach that exploits unique biochemical pathways in bacteria to develop novel pathogen-specific imaging tracers. These tracers have significant potential for clinical translation to specifically detect and localize a broad range of bacteria, including MDROs.


Asunto(s)
Ácido 4-Aminobenzoico/farmacocinética , Bacterias/metabolismo , Infecciones Bacterianas/diagnóstico por imagen , Infecciones Bacterianas/microbiología , Manitol/farmacocinética , Sorbitol/farmacocinética , Bacterias/clasificación , Bacterias/citología , Marcaje Isotópico/métodos , Tomografía de Emisión de Positrones/métodos , Radiofármacos/farmacocinética , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
9.
Nucl Med Biol ; 41(10): 777-84, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25195017

RESUMEN

Nearly 20 years after the World Health Organization declared tuberculosis (TB) a global public health emergency, TB still remains a major global threat with 8.6 million new cases and 1.3 million deaths annually. Mycobacterium tuberculosis adapts to a quiescent physiological state, and is notable for complex interaction with the host, producing poorly-understood disease states ranging from latent infection to fully active disease. Of the approximately 2.5 billion people latently infected with M. tuberculosis, many will develop reactivation disease (relapse), years after the initial infection. While progress has been made on some fronts, the alarming spread of multidrug-resistant, extensively drug-resistant, and more recently totally-drug resistant strains is of grave concern. New tools are urgently needed for rapidly diagnosing TB, monitoring TB treatments and to allow unique insights into disease pathogenesis. Nuclear bioimaging is a powerful, noninvasive tool that can rapidly provide three-dimensional views of disease processes deep within the body and conduct noninvasive longitudinal assessments of the same patient. In this review, we discuss the application of nuclear bioimaging to TB, including the current state of the field, considerations for radioprobe development, study of TB drug pharmacokinetics in infected tissues, and areas of research and clinical needs that could be addressed by nuclear bioimaging. These technologies are an emerging field of research, overcome several fundamental limitations of current tools, and will have a broad impact on both basic research and patient care. Beyond diagnosis and monitoring disease, these technologies will also allow unique insights into understanding disease pathogenesis; and expedite bench-to-bedside translation of new therapeutics. Finally, since molecular imaging is readily available for humans, validated tracers will become valuable tools for clinical applications.


Asunto(s)
Diagnóstico por Imagen/métodos , Mycobacterium tuberculosis/aislamiento & purificación , Medicina Nuclear , Tuberculosis/diagnóstico , Humanos , Tuberculosis/microbiología
10.
Sci Transl Med ; 6(259): 259ra146, 2014 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-25338757

RESUMEN

The Enterobacteriaceae are a family of rod-shaped Gram-negative bacteria that normally inhabit the gastrointestinal tract and are the most common cause of Gram-negative bacterial infections in humans. In addition to causing serious multidrug-resistant, hospital-acquired infections, a number of Enterobacteriaceae species are also recognized as biothreat pathogens. As a consequence, new tools are urgently needed to specifically identify and localize infections due to Enterobacteriaceae and to monitor antimicrobial efficacy. In this report, we used commercially available 2-[(18)F]-fluorodeoxyglucose ((18)F-FDG) to produce 2-[(18)F]-fluorodeoxysorbitol ((18)F-FDS), a radioactive probe for Enterobacteriaceae, in 30 min. (18)F-FDS selectively accumulated in Enterobacteriaceae, but not in Gram-positive bacteria or healthy mammalian or cancer cells in vitro. In a murine myositis model, (18)F-FDS positron emission tomography (PET) rapidly differentiated true infection from sterile inflammation with a limit of detection of 6.2 ± 0.2 log10 colony-forming units (CFU) for Escherichia coli. Our findings were extended to models of mixed Gram-positive and Gram-negative thigh co-infections, brain infection, Klebsiella pneumonia, and mice undergoing immunosuppressive chemotherapy. This technique rapidly and specifically localized infections due to Enterobacteriaceae, providing a three-dimensional holistic view within the animal. Last, (18)F-FDS PET monitored the efficacy of antimicrobial treatment, demonstrating a PET signal proportionate to the bacterial burden. Therapeutic failures associated with multidrug-resistant, extended-spectrum ß-lactamase (ESBL)-producing E. coli infections were detected in real time. Together, these data show that (18)F-FDS is a candidate imaging probe for translation to human clinical cases of known or suspected infections owing to Enterobacteriaceae.


Asunto(s)
Infecciones por Enterobacteriaceae/diagnóstico por imagen , Tomografía de Emisión de Positrones , Sorbitol/análogos & derivados , Animales , Antiinfecciosos/farmacología , Antiinfecciosos/uso terapéutico , Neoplasias Encefálicas/diagnóstico por imagen , Línea Celular , Infecciones por Enterobacteriaceae/tratamiento farmacológico , Escherichia coli/patogenicidad , Femenino , Humanos , Inmunocompetencia/efectos de los fármacos , Inflamación/patología , Infecciones por Klebsiella/diagnóstico por imagen , Infecciones por Klebsiella/tratamiento farmacológico , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/fisiología , Ratones , Pruebas de Sensibilidad Microbiana , Miositis/diagnóstico por imagen , Radiografía , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Proc Natl Acad Sci U S A ; 102(12): 4548-53, 2005 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-15767566

RESUMEN

Mycobacterium tuberculosis (Mtb) is an obligate aerobe that is capable of long-term persistence under conditions of low oxygen tension. Analysis of the Mtb genome predicts the existence of a branched aerobic respiratory chain terminating in a cytochrome bd system and a cytochrome aa(3) system. Both chains can be initiated with type II NADH:menaquinone oxidoreductase. We present a detailed biochemical characterization of the aerobic respiratory chains from Mtb and show that phenothiazine analogs specifically inhibit NADH:menaquinone oxidoreductase activity. The emergence of drug-resistant strains of Mtb has prompted a search for antimycobacterial agents. Several phenothiazines analogs are highly tuberculocidal in vitro, suppress Mtb growth in a mouse model of acute infection, and represent lead compounds that may give rise to a class of selective antibiotics.


Asunto(s)
Antituberculosos/farmacología , Inhibidores Enzimáticos/farmacología , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/enzimología , Quinona Reductasas/antagonistas & inhibidores , Animales , Antituberculosos/química , Transporte de Electrón/genética , Complejo III de Transporte de Electrones/metabolismo , Inhibidores Enzimáticos/química , Escherichia coli/genética , Femenino , Genes Bacterianos , Ratones , Ratones Endogámicos BALB C , Mycobacterium tuberculosis/genética , Consumo de Oxígeno/efectos de los fármacos , Fenotiazinas/química , Fenotiazinas/farmacología , Quinona Reductasas/genética , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/genética , Tuberculosis Pulmonar/tratamiento farmacológico , Tuberculosis Pulmonar/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA