Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 3947, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38729951

RESUMO

Gram-negative bacteria (GNB) are a major cause of neonatal sepsis in low- and middle-income countries (LMICs). Although the World Health Organization (WHO) reports that over 80% of these sepsis deaths could be prevented through improved treatment, the efficacy of the currently recommended first- and second-line treatment regimens for this condition is increasingly affected by high rates of drug resistance. Here we assess three well known antibiotics, fosfomycin, flomoxef and amikacin, in combination as potential antibiotic treatment regimens by investigating the drug resistance and genetic profiles of commonly isolated GNB causing neonatal sepsis in LMICs. The five most prevalent bacterial isolates in the NeoOBS study (NCT03721302) are Klebsiella pneumoniae, Acinetobacter baumannii, E. coli, Serratia marcescens and Enterobacter cloacae complex. Among these isolates, high levels of ESBL and carbapenemase encoding genes are detected along with resistance to ampicillin, gentamicin and cefotaxime, the current WHO recommended empiric regimens. The three new combinations show excellent in vitro activity against ESBL-producing K. pneumoniae and E. coli isolates. Our data should further inform and support the clinical evaluation of these three antibiotic combinations for the treatment of neonatal sepsis in areas with high rates of multidrug-resistant Gram-negative bacteria.


Assuntos
Acinetobacter baumannii , Antibacterianos , Bactérias Gram-Negativas , Infecções por Bactérias Gram-Negativas , Klebsiella pneumoniae , Testes de Sensibilidade Microbiana , Sepse Neonatal , Humanos , Antibacterianos/uso terapêutico , Antibacterianos/farmacologia , Sepse Neonatal/microbiologia , Sepse Neonatal/tratamento farmacológico , Recém-Nascido , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Negativas/genética , Bactérias Gram-Negativas/isolamento & purificação , Infecções por Bactérias Gram-Negativas/tratamento farmacológico , Infecções por Bactérias Gram-Negativas/microbiologia , Acinetobacter baumannii/efeitos dos fármacos , Acinetobacter baumannii/isolamento & purificação , Acinetobacter baumannii/genética , Klebsiella pneumoniae/efeitos dos fármacos , Klebsiella pneumoniae/isolamento & purificação , Klebsiella pneumoniae/genética , Amicacina/farmacologia , Amicacina/uso terapêutico , Fosfomicina/farmacologia , Fosfomicina/uso terapêutico , beta-Lactamases/genética , beta-Lactamases/metabolismo , Escherichia coli/efeitos dos fármacos , Escherichia coli/genética , Escherichia coli/isolamento & purificação , Países em Desenvolvimento , Farmacorresistência Bacteriana Múltipla/genética , Quimioterapia Combinada , Serratia marcescens/efeitos dos fármacos , Serratia marcescens/genética , Serratia marcescens/isolamento & purificação , Enterobacter cloacae/efeitos dos fármacos , Enterobacter cloacae/genética , Enterobacter cloacae/isolamento & purificação , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo
2.
Front Cell Infect Microbiol ; 13: 1282902, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38029240

RESUMO

Objectives: This study summarizes the epidemiological characteristics, species distribution, and drug sensitivity of clinical nontuberculous mycobacteria (NTM) isolates at the Public Health Clinical Center of Chengdu, China, from January 2017 to December 2022. Methods: We retrospectively analyzed data from patients with clinically isolated NTM strains. Chi-square analysis assessed the rate of Mycobacterium strain isolation over 6 years. Results: The number of samples tested for Mycobacterium tuberculosis (MTB) and/or NTM increased each year, while MTB detection decreased and NTM detection rose significantly each year (P=0.03). The average age of NTM patients was 51 ± 17.53 years, with a 14.1% HIV infection rate. The predominant isolates were Mycobacterium avium-intracellulare (MAC) and M. chelonae/M. abscessus, with 96.4% of cases being of Han ethnicity. Amikacin, moxifloxacin, and clarithromycin were effective against M. avium and M. intracellulare; linezolid, amikacin, and cefoxitin were effective against M. chelonae/M. abscessus. Over 90% of NTM cases originated from the respiratory tract. Conclusion: The NTM isolation rate in Southwest China has risen in recent years, primarily among elderly patients with a high HIV co-infection rate. The main NTM isolates were MAC and M. chelonae/M. abscessus. Amikacin, moxifloxacin, clarithromycin, and linezolid exhibited strong antibacterial activity against SGM, while amikacin and linezolid displayed relatively better antibacterial activity against RGM. The prevalence of NTM infection may be positively associated with regional economic development and health conditions.


Assuntos
Infecções por HIV , Infecções por Mycobacterium não Tuberculosas , Tuberculose , Humanos , Idoso , Adulto , Pessoa de Meia-Idade , Micobactérias não Tuberculosas , Claritromicina/farmacologia , Amicacina/farmacologia , Linezolida/farmacologia , Linezolida/uso terapêutico , Infecções por HIV/complicações , Infecções por HIV/epidemiologia , Moxifloxacina/farmacologia , Estudos Retrospectivos , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Infecções por Mycobacterium não Tuberculosas/epidemiologia , Infecções por Mycobacterium não Tuberculosas/microbiologia , Tuberculose/epidemiologia , China/epidemiologia , Testes de Sensibilidade Microbiana
3.
J Antimicrob Chemother ; 77(12): 3349-3357, 2022 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-36177766

RESUMO

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.


Assuntos
Amicacina , Sepse Neonatal , Lactente , Recém-Nascido , Humanos , Amicacina/farmacologia , Amicacina/uso terapêutico , Sepse Neonatal/tratamento farmacológico , Cefalosporinas , Testes de Sensibilidade Microbiana , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Atenção à Saúde
4.
BMC Microbiol ; 22(1): 175, 2022 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-35804298

RESUMO

BACKGROUND: Recently, Mycobacterium avium complex (MAC) infections have been increasing, especially in immunocompromised and older adults. The rapid increase has triggered a global health concern due to limited therapeutic strategies and adverse effects caused by long-term medication. To provide more evidence for the treatment of MAC, we studied the in vitro inhibitory activities of 17 antimicrobial agents against clinical MAC isolates. RESULTS: A total of 111 clinical MAC isolates were enrolled in the study and they were identified as M. intracellulare, M. avium, M. marseillense, M. colombiense, M. yongonense, and two isolates could not be identified at the species level. MAC strains had relatively low (0-21.6%) resistance to clarithromycin, amikacin, bedaquiline, rifabutin, streptomycin, and clofazimine, and the resistant rates to isoniazid, rifampin, linezolid, doxycycline, and ethionamide were very high (72.1-100%). In addition, M. avium had a significantly higher resistance rate than that of M. intracellulare for ethambutol (92.3% vs 40.7%, P < 0.001), amikacin (15.4% vs 1.2%, P = 0.049), and cycloserine (69.2% vs 25.9%, P = 0.004). CONCLUSIONS: Our results supported the current usage of macrolides, rifabutin, and aminoglycosides in the regimens for MAC infection, and also demonstrated the low resistance rate against new drugs, such as clofazimine, tedizolid, and bedaquiline, suggesting the possible implementation of these drugs in MAC treatment.


Assuntos
Anti-Infecciosos , Infecção por Mycobacterium avium-intracellulare , Idoso , Amicacina/farmacologia , Antibacterianos/farmacologia , Anti-Infecciosos/farmacologia , Claritromicina/farmacologia , Clofazimina/farmacologia , Humanos , Testes de Sensibilidade Microbiana , Complexo Mycobacterium avium , Infecção por Mycobacterium avium-intracellulare/tratamento farmacológico , Infecção por Mycobacterium avium-intracellulare/microbiologia , Rifabutina/farmacologia
5.
Microb Pathog ; 164: 105402, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35038548

RESUMO

Brucella spp. are facultative intracellular pathogens that can persistently colonize animal host cells and cause zoonotic brucellosis. Brucellosis affects public health and safety and even affects economic development. Our lab found that a Brucella strain isolated from Marmota himalayana exhibited amikacin resistance. To annotate and analyze the potential resistance genes in this strain, we utilized sequencing platforms in this study and cloned potential resistance genes. The findings showed that the isolated strain belonged to B. abortus biovar 1 and was similar to B. abortus 2308. The isolate had amikacin resistance genes encoding aminoglycoside 3'-phosphotransferase. Based on the results of genome analysis, the isolated strain may have obtained amikacin resistance genes from Salmonella spp. through Tn3 family transposons. Notably, this study establishes a foundation for further research on the resistance mechanism of Brucella spp. and provides data that may be useful for the prevention and control of drug-resistant Brucella strains.


Assuntos
Brucella abortus , Brucelose , Amicacina/farmacologia , Animais , Brucella abortus/genética , Marmota
6.
Curr Top Med Chem ; 20(11): 1042-1055, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32250224

RESUMO

BACKGROUND: Due to the appearance of resistant bacterial strains against the antimicrobial drugs and the reduced efficiency of these valuable resources, the health of a community and the economies of countries have been threatened. OBJECTIVE: In this study, the antibacterial assessment of zinc sulfide nanoparticles (ZnS NPs) against Streptococcus pyogenes and Acinetobacter baumannii has been performed. METHODS: ZnS NPs were synthesized through a co-precipitation method using polyvinylpyrrolidone (PVP), polyvinyl alcohol (PVA) and polyethylene glycol (PEG-4000). The size and morphology of the synthesized ZnS NPs were determined by a scanning electron microscope (SEM) and it was found that the average size of the applied NPs was about 70 nm. In order to evaluate the antibacterial effect of the synthesized ZnS NPs, various concentrations (50µg/mL, 100 µg/mL and 150 µg/mL) of ZnS NPs were prepared. Antibacterial assessments were performed through the disc diffusion method in Mueller Hinton Agar (MHA) culture medium and also the optical density (OD) method was performed by a UV-Vis spectrophotometer in Trypticase™ Soy Broth (TSB) medium. Then, in order to compare the antibacterial effects of the applied NPs, several commercial antibiotics including penicillin, amikacin, ceftazidime and primaxin were used. RESULTS: The achieved results indicated that the antibacterial effects of ZnS NPs had a direct relation along with the concentrations and the concentration of 150 µg/mL showed the highest antibacterial effect in comparison with others. In addition, the ZnS NPs were more effective on Acinetobacter baumannii. CONCLUSION: The findings of this research suggest a novel approach against antibiotic resistance.


Assuntos
Acinetobacter baumannii/efeitos dos fármacos , Antibacterianos/química , Nanopartículas Metálicas/química , Streptococcus pyogenes/efeitos dos fármacos , Sulfetos/química , Compostos de Zinco/química , Amicacina/farmacologia , Animais , Antibacterianos/farmacologia , Ceftazidima/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Combinação Imipenem e Cilastatina/farmacologia , Desenvolvimento de Medicamentos , Resistência Microbiana a Medicamentos , Humanos , Testes de Sensibilidade Microbiana , Penicilinas/farmacologia , Polietilenoglicóis/química , Álcool de Polivinil/química , Povidona/química , Ratos , Sulfetos/farmacologia , Compostos de Zinco/farmacologia
7.
Artigo em Inglês | MEDLINE | ID: mdl-31843996

RESUMO

Mycobacterium abscessus is intrinsically resistant to most antimicrobial agents. The emerging infections caused by M. abscessus and the lack of effective treatment call for rapid attention. Here, we intended to construct a selectable marker-free autoluminescent M. abscessus strain (designated UAlMab) as a real-time reporter strain to facilitate the discovery of effective drugs and regimens for treating M. abscessus The UAlMab strain was constructed using the dif/Xer recombinase system. In vitro and in vivo activities of several drugs, including clofazimine and TB47, a recently reported cytochrome bc1 inhibitor, were assessed using UAlMab. Furthermore, the efficacy of multiple drug combinations, including the clofazimine and TB47 combination, were tested against 20 clinical M. abscessus isolates. The UAlMab strain enabled us to evaluate drug efficacy both in vitro and in live BALB/c mice in a real-time, noninvasive fashion. Importantly, although TB47 showed marginal activity either alone or in combination with clarithromycin, amikacin, or roxithromycin, the drug markedly potentiated the activity of clofazimine, both in vitro and in vivo This study demonstrates that the use of the UAlMab strain can significantly facilitate rapid evaluation of new drugs and regimens. The clofazimine and TB47 combination is effective against M. abscessus, and dual/triple electron transport chain (ETC) targeting can be an effective therapeutic approach for treating mycobacterial infections.


Assuntos
Antibacterianos/farmacologia , Clofazimina/farmacologia , Complexo III da Cadeia de Transporte de Elétrons/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Infecções por Mycobacterium não Tuberculosas/tratamento farmacológico , Mycobacterium abscessus/efeitos dos fármacos , Amicacina/farmacologia , Animais , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Claritromicina/farmacologia , Combinação de Medicamentos , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Farmacorresistência Bacteriana Múltipla/genética , Sinergismo Farmacológico , Transporte de Elétrons/efeitos dos fármacos , Complexo III da Cadeia de Transporte de Elétrons/genética , Complexo III da Cadeia de Transporte de Elétrons/metabolismo , Feminino , Engenharia Genética/métodos , Luminescência , Camundongos , Camundongos Endogâmicos BALB C , Testes de Sensibilidade Microbiana , Infecções por Mycobacterium não Tuberculosas/enzimologia , Infecções por Mycobacterium não Tuberculosas/microbiologia , Infecções por Mycobacterium não Tuberculosas/patologia , Mycobacterium abscessus/genética , Mycobacterium abscessus/metabolismo , Imagem Óptica/métodos , Recombinases/genética , Recombinases/metabolismo , Roxitromicina/farmacologia
8.
Artigo em Inglês | MEDLINE | ID: mdl-29876099

RESUMO

Background: A high level of antibiotic consumption in France means antimicrobial resistance requires rigorous monitoring. The Tigecycline Evaluation and Surveillance Trial (T.E.S.T.) is a global surveillance study that monitors the in vitro activities of tigecycline and a panel of marketed antimicrobials against clinically important Gram-positive and Gram-negative isolates. Methods: Annually clinically relevant strains were prospectively included in the survey through a national network of hospital-based laboratories. MICs were determined locally by broth microdilution using CLSI guidelines. Antimicrobial susceptibility was assessed using European Committee on Antimicrobial Susceptibility Testing breakpoints. Results: Thirty-three centres in France collected 26,486 isolates between 2004 and 2016. Enterococcus species were highly susceptible (≥94.4%) to linezolid, tigecycline and vancomycin. Staphylococcus aureus, including methicillin-resistant S. aureus (MRSA), were susceptible (≥99.9%) to tigecycline, vancomycin and linezolid. Between 2004 and 2016, 27.7% of S. aureus isolates were MRSA, decreasing from 28.0% in 2013 to 23.5% in 2016. Susceptibility of Streptococcus pneumoniae isolates was 100% to vancomycin, and > 99.0% to levofloxacin, linezolid and meropenem; 3.0% were penicillin-resistant S. pneumoniae (100% susceptibility to vancomycin and linezolid). Escherichia coli isolates were highly susceptible (> 98.0%) to meropenem, tigecycline and amikacin. The rate of extended-spectrum ß-lactamase (ESBL) positive E. coli increased from 2004 (3.0%), but was stable from 2012 (23.1%) to 2016 (19.8%). Susceptibility of Klebsiella pneumoniae isolates was 99.4% to meropenem and 96.5% to amikacin. The proportion of ESBL-positive K. pneumoniae isolates increased from 2004 (7.5%) to 2012 (33.3%) and was highest in 2016 (43.6%). A. baumannii was susceptible to meropenem (81.0%) and amikacin (74.9%); none of the 6.2% of isolates identified as multidrug-resistant (MDR) was susceptible to any agents with breakpoints. P. aeruginosa isolates were most susceptible to amikacin (88.5%), and MDR rates were 13.6% in 2013 to 4.0% in 2016; susceptibility of MDR isolates was no higher than 31.4% to amikacin. Conclusions: Rates of MRSA decreased slowly, while rates of ESBL-positive E. coli and K. pneumoniae increased from 2004 to 2016. Susceptibility of Gram-positive isolates to vancomycin, tigecycline, meropenem and linezolid was well conserved, as was susceptibility of Gram-negative isolates to tigecycline and meropenem. The spread of MDR non-fermentative isolates must be carefully monitored.


Assuntos
Farmacorresistência Bacteriana Múltipla/fisiologia , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Acinetobacter baumannii/efeitos dos fármacos , Acinetobacter baumannii/isolamento & purificação , Amicacina/farmacologia , Enterococcus/efeitos dos fármacos , Enterococcus/isolamento & purificação , Escherichia coli/efeitos dos fármacos , Escherichia coli/isolamento & purificação , França , Humanos , Klebsiella pneumoniae/efeitos dos fármacos , Klebsiella pneumoniae/isolamento & purificação , Levofloxacino/farmacologia , Linezolida/farmacologia , Meropeném/farmacologia , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/isolamento & purificação , Testes de Sensibilidade Microbiana , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/isolamento & purificação , Streptococcus pneumoniae/efeitos dos fármacos , Streptococcus pneumoniae/isolamento & purificação , Tigeciclina/farmacologia , Vancomicina/farmacologia
9.
Artigo em Inglês | MEDLINE | ID: mdl-27821448

RESUMO

Optimizing antibiotic combinations is promising to combat multidrug-resistant Pseudomonas aeruginosa This study aimed to systematically evaluate synergistic bacterial killing and prevention of resistance by carbapenem and aminoglycoside combinations and to rationally optimize combination dosage regimens via a mechanism-based mathematical model (MBM). We studied monotherapies and combinations of imipenem with tobramycin or amikacin against three difficult-to-treat double-resistant clinical P. aeruginosa isolates. Viable-count profiles of total and resistant populations were quantified in 48-h static-concentration time-kill studies (inoculum, 107.5 CFU/ml). We rationally optimized combination dosage regimens via MBM and Monte Carlo simulations against isolate FADDI-PA088 (MIC of imipenem [MICimipenem] of 16 mg/liter and MICtobramycin of 32 mg/liter, i.e., both 98th percentiles according to the EUCAST database). Against this isolate, imipenem (1.5× MIC) combined with 1 to 2 mg/liter tobramycin (MIC, 32 mg/liter) or amikacin (MIC, 4 mg/liter) yielded ≥2-log10 more killing than the most active monotherapy at 48 h and prevented resistance. For all three strains, synergistic killing without resistance was achieved by ≥0.88× MICimipenem in combination with a median of 0.75× MICtobramycin (range, 0.032× to 2.0× MICtobramycin) or 0.50× MICamikacin (range, 0.25× to 0.50× MICamikacin). The MBM indicated that aminoglycosides significantly enhanced the imipenem target site concentration up to 3-fold; achieving 50% of this synergistic effect required aminoglycoside concentrations of 1.34 mg/liter (if the aminoglycoside MIC was 4 mg/liter) and 4.88 mg/liter (for MICs of 8 to 32 mg/liter). An optimized combination regimen (continuous infusion of imipenem at 5 g/day plus a 0.5-h infusion with 7 mg/kg of body weight tobramycin) was predicted to achieve >2.0-log10 killing and prevent regrowth at 48 h in 90.3% of patients (median bacterial killing, >4.0 log10 CFU/ml) against double-resistant isolate FADDI-PA088 and therefore was highly promising.


Assuntos
Aminoglicosídeos/farmacologia , Antibacterianos/farmacologia , Carbapenêmicos/farmacologia , Pseudomonas aeruginosa/efeitos dos fármacos , Amicacina/farmacologia , Sinergismo Farmacológico , Imipenem/farmacologia , Testes de Sensibilidade Microbiana , Modelos Teóricos , Método de Monte Carlo , Tobramicina/farmacologia
10.
Antimicrob Agents Chemother ; 60(2): 1097-105, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26643335

RESUMO

Multidrug therapy is a standard practice when treating infections by nontuberculous mycobacteria (NTM), but few treatment options exist. We conducted this study to define the drug-drug interaction between clofazimine and both amikacin and clarithromycin and its contribution to NTM treatment. Mycobacterium abscessus and Mycobacterium avium type strains were used. Time-kill assays for clofazimine alone and combined with amikacin or clarithromycin were performed at concentrations of 0.25× to 2× MIC. Pharmacodynamic interactions were assessed by response surface model of Bliss independence (RSBI) and isobolographic analysis of Loewe additivity (ISLA), calculating the percentage of statistically significant Bliss interactions and interaction indices (I), respectively. Monte Carlo simulations with predicted human lung concentrations were used to calculate target attainment rates for combination and monotherapy regimens. Clofazimine alone was bacteriostatic for both NTM. Clofazimine-amikacin was synergistic against M. abscessus (I = 0.41; 95% confidence interval [CI], 0.29 to 0.55) and M. avium (I = 0.027; 95% CI, 0.007 to 0.048). Based on RSBI analysis, synergistic interactions of 28.4 to 29.0% and 23.2 to 56.7% were observed at 1× to 2× MIC and 0.25× to 2× MIC for M. abscessus and M. avium, respectively. Clofazimine-clarithromycin was also synergistic against M. abscessus (I = 0.53; 95% CI, 0.35 to 0.72) and M. avium (I = 0.16; 95% CI, 0.04 to 0.35), RSBI analysis showed 23.5% and 23.3 to 53.3% at 2× MIC and 0.25× to 0.5× MIC for M. abscessus and M. avium, respectively. Clofazimine prevented the regrowth observed with amikacin or clarithromycin alone. Target attainment rates of combination regimens were >60% higher than those of monotherapy regimens for M. abscessus and M. avium. The combination of clofazimine with amikacin or clarithromycin was synergistic in vitro. This suggests a potential role for clofazimine in treatment regimens that warrants further evaluation.


Assuntos
Amicacina/farmacologia , Antibacterianos/farmacologia , Claritromicina/farmacologia , Clofazimina/farmacologia , Mycobacterium avium/efeitos dos fármacos , Micobactérias não Tuberculosas/efeitos dos fármacos , Interações Medicamentosas , Sinergismo Farmacológico , Quimioterapia Combinada , Testes de Sensibilidade Microbiana , Método de Monte Carlo , Mutação , Mycobacterium avium/genética , Mycobacterium avium/crescimento & desenvolvimento , Micobactérias não Tuberculosas/genética , Micobactérias não Tuberculosas/crescimento & desenvolvimento
11.
Antimicrob Agents Chemother ; 60(3): 1242-8, 2015 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-26643339

RESUMO

The treatment of pulmonary Mycobacterium abscessus disease is associated with very high failure rates and easily acquired drug resistance. Amikacin is the key drug in treatment regimens, but the optimal doses are unknown. No good preclinical model exists to perform formal pharmacokinetics/pharmacodynamics experiments to determine these optimal doses. We developed a hollow-fiber system model of M. abscessus disease and studied amikacin exposure effects and dose scheduling. We mimicked amikacin human pulmonary pharmacokinetics. Both amikacin microbial kill and acquired drug resistance were linked to the peak concentration-to-MIC ratios; the peak/MIC ratio associated with 80% of maximal kill (EC80) was 3.20. However, on the day of the most extensive microbial kill, the bacillary burden did not fall below the starting inoculum. We performed Monte Carlo simulations of 10,000 patients with pulmonary M. abscessus infection and examined the probability that patients treated with one of 6 doses from 750 mg to 4,000 mg would achieve or exceed the EC80. We also examined these doses for the ability to achieve a cumulative area under the concentration-time curve of 82,232 mg · h/liter × days, which is associated with ototoxicity. The standard amikacin doses of 750 to 1,500 mg a day achieved the EC80 in ≤ 21% of the patients, while a dose of 4 g/day achieved this in 70% of the patients but at the cost of high rates of ototoxicity within a month or two. The susceptibility breakpoint was an MIC of 8 to 16 mg/liter. Thus, amikacin, as currently dosed, has limited efficacy against M. abscessus. It is urgent that different antibiotics be tested using our preclinical model and new regimens developed.


Assuntos
Amicacina/farmacocinética , Antibacterianos/farmacologia , Antibacterianos/farmacocinética , Testes de Sensibilidade Microbiana/métodos , Micobactérias não Tuberculosas/efeitos dos fármacos , Amicacina/farmacologia , Relação Dose-Resposta a Droga , Humanos , Testes de Sensibilidade Microbiana/instrumentação , Modelos Biológicos , Método de Monte Carlo , Taxa de Mutação , Micobactérias não Tuberculosas/genética , Micobactérias não Tuberculosas/patogenicidade
12.
Infect Dis (Lond) ; 47(12): 902-7, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26365683

RESUMO

BACKGROUND: Acinetobacter baumannii is one of the most prevalent causes of severe hospital-acquired infections and is responsible for the dramatic increase in carbapenem resistance in Croatia in the last 5 years. Such data have encouraged multicenter research focused on the organism's ability to form biofilm, susceptibility to antibiotics, and particular genotype lineage. METHODS: Biofilm formation in 109 unrelated clinical isolates of A. baumannii recovered in six cities of Southern Croatia was investigated. Genotyping was performed by pulsed-field gel electrophoresis and antibiotic profile was tested by applying the disc diffusion method and confirmed by determining the minimum inhibitory concentrations. The ability to form biofilm in vitro was determined from overnight cultures of the collected isolates on microtiter plates, after staining with crystal violet, and quantified at 570 nm after solubilization with ethanol. The statistical relevance was calculated in an appropriate program with level of statistical confidence. RESULTS: There was no significant difference in biofilm formation due to the genotype lineage. Isolates collected from intensive care units (ICUs) and isolated from respiratory samples were more likely to create a biofilm compared with isolates from other departments and other samples. There was a significant difference in the ability to produce biofilm in relation to antibiotic resistance pattern. A large proportion of A. baumannii isolates that were resistant to ampicillin/sulbactam, carbapenems, and amikacin were found to be biofilm-negative. In contrast, isolates susceptible and intermediately susceptible to ampicillin/sulbactam, carbapenems, and amikacin were biofilm producers. CONCLUSION: Clinical isolates of A. baumannii from respiratory samples in ICUs with a particular susceptibility pattern are more prone to form biofilm.


Assuntos
Infecções por Acinetobacter/microbiologia , Acinetobacter baumannii/genética , Acinetobacter baumannii/fisiologia , Biofilmes/crescimento & desenvolvimento , Farmacorresistência Bacteriana Múltipla , Infecções por Acinetobacter/tratamento farmacológico , Acinetobacter baumannii/efeitos dos fármacos , Amicacina/farmacologia , Ampicilina/farmacologia , Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Carbapenêmicos/farmacologia , Croácia , Infecção Hospitalar , Farmacorresistência Bacteriana Múltipla/genética , Eletroforese em Gel de Campo Pulsado , Genótipo , Humanos , Unidades de Terapia Intensiva , Testes de Sensibilidade Microbiana , Método de Monte Carlo , Sistema Respiratório/microbiologia , Infecções Respiratórias/microbiologia , Sulbactam/farmacologia , Urina/microbiologia , Ferimentos e Lesões/microbiologia
13.
Antimicrob Agents Chemother ; 59(10): 6344-51, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26248375

RESUMO

Based on a previously derived population pharmacokinetic model, a novel neonatal amikacin dosing regimen was developed. The aim of the current study was to prospectively evaluate this dosing regimen. First, early (before and after second dose) therapeutic drug monitoring (TDM) observations were evaluated for achieving target trough (<3 mg/liter) and peak (>24 mg/liter) levels. Second, all observed TDM concentrations were compared with model-predicted concentrations, whereby the results of a normalized prediction distribution error (NPDE) were considered. Subsequently, Monte Carlo simulations were performed. Finally, remaining causes limiting amikacin predictability (i.e., prescription errors and disease characteristics of outliers) were explored. In 579 neonates (median birth body weight, 2,285 [range, 420 to 4,850] g; postnatal age 2 days [range, 1 to 30 days]; gestational age, 34 weeks [range, 24 to 41 weeks]), 90.5% of the observed early peak levels reached 24 mg/liter, and 60.2% of the trough levels were <3 mg/liter (93.4% ≤5 mg/liter). Observations were accurately predicted by the model without bias, which was confirmed by the NPDE. Monte Carlo simulations showed that peak concentrations of >24 mg/liter were reached at steady state in almost all patients. Trough values of <3 mg/liter at steady state were documented in 78% to 100% and 45% to 96% of simulated cases with and without ibuprofen coadministration, respectively; suboptimal trough levels were found in patients with postnatal age <14 days and current weight of >2,000 g. Prospective evaluation of a model-based neonatal amikacin dosing regimen resulted in optimized peak and trough concentrations in almost all patients. Slightly adapted dosing for patient subgroups with suboptimal trough levels was proposed. This model-based approach improves neonatal dosing individualization.


Assuntos
Amicacina/farmacocinética , Antibacterianos/farmacocinética , Monitoramento de Medicamentos , Modelos Estatísticos , Amicacina/sangue , Amicacina/farmacologia , Antibacterianos/sangue , Antibacterianos/farmacologia , Peso ao Nascer , Creatinina/sangue , Relação Dose-Resposta a Droga , Esquema de Medicação , Feminino , Idade Gestacional , Humanos , Lactente , Recém-Nascido , Recém-Nascido Prematuro , Masculino , Método de Monte Carlo , Medicina de Precisão , Estudos Prospectivos , Sepse/tratamento farmacológico , Sepse/microbiologia , Sepse/patologia
14.
PLoS One ; 8(10): e76687, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24204655

RESUMO

The rules that govern adaptive protein evolution remain incompletely understood. Aminoglycoside aminotransferase (3') type IIIa (hereafter abbreviated APH(3')-IIIa) is a good model enzyme because it inactivates kanamycin efficiently; it recognizes other aminoglycoside antibiotics, including amikacin, but not nearly as well. Here we direct the evolution of APH(3')-IIIa variants with increased activity against amikacin. After four rounds of random mutation and selection in Escherichia coli, the minimum inhibitory concentration of amikacin rose from 18 micrograms/mL (wild-type enzyme) to over 1200 micrograms/mL (clone 4.1). The artificially evolved 4.1 APH(3')-IIIa variant exhibited 19-fold greater catalytic efficiency (k cat/K M) than did the wild-type enzyme in reactions with amikacin. E. coli expressing the evolved 4.1 APH(3')-IIIa also exhibited a four-fold decrease in fitness (as measured by counting colony forming units in liquid cultures with the same optical density) compared with isogenic cells expressing the wild-type protein under non-selective conditions. We speculate that these fitness costs, in combination with the prevalence of other amikacin-modifying enzymes, hinder the evolution of APH(3')-IIIa in clinical settings.


Assuntos
Amicacina/farmacologia , Proteínas de Escherichia coli/genética , Canamicina Quinase/genética , Mutação/efeitos dos fármacos , Antibacterianos/farmacologia , Biocatálise , Cristalografia por Raios X , Evolução Molecular Direcionada/métodos , Relação Dose-Resposta a Droga , Estabilidade Enzimática , Escherichia coli/efeitos dos fármacos , Escherichia coli/genética , Escherichia coli/crescimento & desenvolvimento , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Aptidão Genética , Canamicina Quinase/química , Canamicina Quinase/metabolismo , Cinética , Testes de Sensibilidade Microbiana , Modelos Moleculares , Estrutura Terciária de Proteína
15.
Int J Antimicrob Agents ; 42(2): 155-60, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23756322

RESUMO

The objectives of this study were to conduct a comparative pharmacokinetic/pharmacodynamic (PK/PD) evaluation using Monte Carlo simulation of conventional versus high-dose extended-interval dosage (HDED) regimens of amikacin (AMK) in intensive care unit (ICU) patients for an Acinetobacter baumannii infection model. The simulation was performed in five populations (a control population and four subpopulations of ICU patients). Using a specific AMK PK/PD model and Monte Carlo simulation, the following were generated: simulated AMK steady-state plasma level curves; PK/PD efficacy indexes [time during which the serum drug concentration remains above the minimum inhibitory concentration (MIC) for a dosing period (%T>MIC) and ratio of peak serum concentration to MIC (Cmax/MIC)]; evolution of bacterial growth curves; and adaptive resistance to treatment. A higher probability of bacterial resistance was observed with the HDED regimen compared with the conventional dosage regimen. A statistically significant increase in Cmax/MIC and a statistically significant reduction in %T>MIC with the HDED regimen were obtained. A multiple linear relationship between CFU values at 24h with Cmax/MIC and %T>MIC was obtained. In conclusion, with the infection model tested, the likelihood of resistance to treatment may be higher against pathogens with a high MIC with the HDED regimen, considering that in many ICU patients the %T>MIC may be limited. If a sufficient value of %T>MIC (≥60%) is not reached, even though the Cmax/MIC is high, the therapeutic efficacy of the treatment may not be guaranteed. This study indicates that different AMK dosing strategies could directly influence the efficacy results in ICU patients.


Assuntos
Infecções por Acinetobacter/tratamento farmacológico , Acinetobacter baumannii/efeitos dos fármacos , Amicacina/administração & dosagem , Amicacina/farmacocinética , Antibacterianos/administração & dosagem , Antibacterianos/farmacocinética , Infecções por Acinetobacter/microbiologia , Amicacina/farmacologia , Antibacterianos/farmacologia , Estado Terminal , Humanos , Unidades de Terapia Intensiva , Testes de Sensibilidade Microbiana , Modelos Estatísticos , Plasma/química , Fatores de Tempo
16.
J Egypt Natl Canc Inst ; 25(1): 31-5, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23499204

RESUMO

BACKGROUND AND AIM: Appropriate antibiotic selection and timing of administration for prophylaxis are crucial to reduce the likelihood of surgical site infection (SSI) after a clean contaminated cancer surgery. Our aim is to compare the use of two prophylactic antibiotic (PA) regimens as regards efficacy, timing, and cost. PATIENTS AND METHODS: Two hundred patients with gastric, bladder, or colorectal cancer were randomized to receive preoperative PA, group A received penicillin G sodium and gentamicin and group B received clindamycin and amikacin intravenously. The demographic data of patients were collected, and they were observed for wound infections. RESULTS: Infected wounds occurred in 19 patients with a rate of 9.5%. Highest incidence of SSI was among bladder cancer patients (14.2%); p=0.044. The rate of SSI was 11% in group A, and 8% in group B, p=0.469. The cost of PA administered in group A was significantly less than that of group B (21.96±3.22LE versus 117.05±12.74LE, respectively; p<0.001). SSI tended to be higher among those who had longer time for antibiotic and incision (≥30min) than those who had shorter time interval (<30min), (13% vs. 6.5%, respectively). CONCLUSION: Both penicillin+gentamicin and clindamycin+amikacin are safe and effective for the prevention of SSI in clean contaminated operative procedures. In a resource limited hospital, a regimen including penicillin+gentamicin is a cost-effective alternative for the more expensive and broader coverage of clindamycin+amikacin. Timing of PA is effective in preventing SSIs when administered 30min before the start of surgery.


Assuntos
Amicacina/uso terapêutico , Antibacterianos/uso terapêutico , Antibioticoprofilaxia/economia , Clindamicina/uso terapêutico , Gentamicinas/uso terapêutico , Penicilina G/uso terapêutico , Infecção da Ferida Cirúrgica/prevenção & controle , Adulto , Amicacina/economia , Amicacina/farmacologia , Antibacterianos/economia , Antibacterianos/farmacologia , Clindamicina/economia , Clindamicina/farmacologia , Neoplasias Colorretais/cirurgia , Quimioterapia Combinada/economia , Feminino , Gentamicinas/economia , Gentamicinas/farmacologia , Humanos , Masculino , Pessoa de Meia-Idade , Penicilina G/economia , Penicilina G/farmacologia , Fatores de Risco , Infecções Estafilocócicas/prevenção & controle , Neoplasias Gástricas/cirurgia , Neoplasias da Bexiga Urinária/cirurgia
17.
J Infect Dis ; 201(6): 889-97, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20156065

RESUMO

BACKGROUND: Combination antimicrobial therapy is clinically used as a last-resort strategy to control multidrug-resistant bacterial infections. However, selection of antibiotics is often empirical, and conventional assessment of combined drug effect has not been correlated to clinical outcomes. Here, we report a quantitative method to assess combined killing of antimicrobial agents against 2 multidrug-resistant bacteria. METHODS: Combined time-kill studies were performed using clinically achievable concentrations for each 2-agent combination against clinical isolates of Acinetobacter baumannii and Pseudomonas aeruginosa. Bacterial burden observed at 24 h was mathematically modeled using a 3-dimensional response surface. Subsequently, a neutropenic murine pneumonia model with simulated clinical dosing exposures was used to validate our quantitative assessment of combined killing. RESULTS: Different antimicrobial combinations were found to have varying efficacy against the multidrug-resistant bacteria. As predicted by our quantitative method, cefepime plus amikacin was found to be the most superior combination, which was evidenced by a reduction in tissue bacterial burden and prolonged survival of infected animals. CONCLUSIONS: The consistency between the predictions of the mathematical model and in vivo observations substantiated the robustness of our quantitative method. These data highlighted a novel and promising method to guide rational selection of antimicrobial combination in the clinical setting.


Assuntos
Acinetobacter baumannii/efeitos dos fármacos , Antibacterianos/farmacologia , Pneumonia Bacteriana/tratamento farmacológico , Pseudomonas aeruginosa/efeitos dos fármacos , Amicacina/farmacocinética , Amicacina/farmacologia , Amicacina/uso terapêutico , Animais , Antibacterianos/farmacocinética , Antibacterianos/uso terapêutico , Cefepima , Cefalosporinas/farmacocinética , Cefalosporinas/farmacologia , Cefalosporinas/uso terapêutico , Modelos Animais de Doenças , Farmacorresistência Bacteriana Múltipla , Quimioterapia Combinada , Feminino , Humanos , Pulmão/efeitos dos fármacos , Pulmão/microbiologia , Camundongos , Ofloxacino/farmacocinética , Ofloxacino/farmacologia , Ofloxacino/uso terapêutico , Pneumonia Bacteriana/microbiologia , Pneumonia Bacteriana/mortalidade , Reação em Cadeia da Polimerase , Distribuição Aleatória , Análise de Sobrevida
18.
J Clin Pharm Ther ; 33(4): 409-17, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18613859

RESUMO

BACKGROUND AND OBJECTIVE: Monitoring of renal function in cystic fibrosis (CF) patients is essential. The dosage regimen of amikacin is regularly modified according to the patient's glomerular filtration rate (GFR). The aim of the study was to evaluate the use of cystatin C (CyC) for monitoring amikacin therapy along with other markers of renal tubular and glomerular function, and damage [N-acetyl-beta-d glucosaminidase (NAG), creatinine level and creatinine clearance]. METHODS: We compared the GFR, estimated from the serum concentrations of creatinine (Cockcroft-Gault formula) and CyC (Grubb's formula). Seventy-one patients (mean age 12 years; range 4-28 years) with CF were treated by intermittent intravenous infusion of amikacin. Tubular nephrotoxicity was investigated by measurement of urine NAG/urine creatinine ratio (U-NAG/U-creatinine). Concentrations of all markers were measured before starting amikacin therapy and at days 3, 5, 7, 10 and 12. Fluorescence polarization analysis, turbidimetry, enzymatic phototometric creatinine deaminase method and fluorimetry were used for determination of serum amikacin, serum CyC, creatinine and urine NAG activity. Receiver operating characteristic (ROC) analysis was performed to assess the influence of GFR estimated from serum creatinine and serum CyC for the prediction of amikacin clearance during aminoglycoside therapy. RESULTS: Significant differences in the rate of U-NAG/U-creatinine were noted before and after treatment with amikacin (P < 0.001). Serum creatinine levels and creatinine clearance at the end of amikacin therapy (12th day) did not show any significant differences in comparison with the levels measured before the start of therapy (0th day). At days 5, 7, 10 and 12, serum CyC levels showed a significant elevation (P < 0.001), and CyC clearance showed a significant decrease (P < 0.001) in comparison with the levels measured at day 0. The ratio of amikacin clearance/creatinine clearance decreased with therapy whereas the amikacin clearance/CyC and amikacin clearance/CyC clearance increased. CONCLUSION: We showed that the rate of U-NAG/U-creatinine is a suitable marker for monitoring tubular nephrotoxicity in CF patients. Serum creatinine and estimated creatinine clearance are modest predictors of GFR in CF patients. CyC appears to be a better marker of GFR than serum creatinine concentration or creatinine clearance in our study. Serum CyC levels and CyC clearance showed greater ability to predict amikacin clearance during therapy than creatinine clearance.


Assuntos
Amicacina/farmacologia , Antibacterianos/farmacologia , Cistatinas/sangue , Fibrose Cística/tratamento farmacológico , Taxa de Filtração Glomerular , Acetilglucosaminidase/urina , Adolescente , Adulto , Amicacina/administração & dosagem , Amicacina/farmacocinética , Antibacterianos/administração & dosagem , Antibacterianos/farmacocinética , Biomarcadores/sangue , Criança , Pré-Escolar , Creatinina/sangue , Creatinina/urina , Cistatina C , Fibrose Cística/sangue , Monitoramento de Medicamentos , Feminino , Humanos , Infusões Intravenosas , Rim/efeitos dos fármacos , Rim/patologia , Masculino , Curva ROC , Fatores de Tempo
19.
Antimicrob Agents Chemother ; 52(8): 2898-904, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18505848

RESUMO

Treatment of multidrug-resistant bacterial infections poses a therapeutic challenge to clinicians; combination therapy is often the only viable option for multidrug-resistant infections. A quantitative method was developed to assess the combined killing abilities of antimicrobial agents. Time-kill studies (TKS) were performed using a multidrug-resistant clinical isolate of Acinetobacter baumannii with escalating concentrations of cefepime (0 to 512 mg/liter), amikacin (0 to 256 mg/liter), and levofloxacin (0 to 64 mg/liter). The bacterial burden data in single and combined (two of the three agents with clinically achievable concentrations in serum) TKS at 24 h were mathematically modeled to provide an objective basis for comparing various antimicrobial agent combinations. Synergy and antagonism were defined as interaction indices of <1 and >1, respectively. A hollow-fiber infection model (HFIM) simulating various clinical (fluctuating concentrations over time) dosing exposures was used to selectively validate our quantitative assessment of the combined killing effect. Model fits in all single-agent TKS were satisfactory (r(2) > 0.97). An enhanced combined overall killing effect was seen in the cefepime-amikacin combination (interactive index, 0.698; 95% confidence interval [CI], 0.675 to 0.722) and the cefepime-levofloxacin combination (interactive index, 0.929; 95% CI, 0.903 to 0.956), but no significant difference in the combined overall killing effect for the levofloxacin-amikacin combination was observed (interactive index, 0.994; 95% CI, 0.982 to 1.005). These assessments were consistent with observations in HFIM validation studies. Our method could be used to objectively rank the combined killing activities of two antimicrobial agents when used together against a multidrug-resistant A. baumannii isolate. It may offer better insights into the effectiveness of various antimicrobial combinations and warrants further investigations.


Assuntos
Acinetobacter baumannii/efeitos dos fármacos , Anti-Infecciosos/farmacologia , Farmacorresistência Bacteriana/efeitos dos fármacos , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Infecções por Acinetobacter/microbiologia , Amicacina/farmacologia , Cefepima , Cefalosporinas/farmacologia , Antagonismo de Drogas , Sinergismo Farmacológico , Humanos , Levofloxacino , Testes de Sensibilidade Microbiana , Ofloxacino/farmacologia
20.
Clin Microbiol Infect ; 11(4): 319-25, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15760430

RESUMO

A guinea-pig pneumonia model involving imipenem-susceptible and imipenem-resistant strains of Acinetobacter baumannii was developed to assess the in-vitro and in-vivo activities of imipenem, alone or in combination with amikacin, and the pharmacokinetic and pharmacodynamic parameters. Serum levels were measured by bioassay (imipenem) or immunoassay (amikacin), followed by calculation of pharmacokinetic and pharmacodynamic parameters (Cmax, AUC, t1/2, Cmax/MIC, AUC/MIC, and Deltat/MIC). In-vivo efficacy was evaluated by comparing bacterial counts in the lungs of treatment groups with end-of-therapy controls by anova and post-hoc tests. Decreases in the Cmax (13.4%), AUC (13%), t1/2 (25%) and Deltat/MIC (11.8-32.2%) of imipenem were observed when it was administered with amikacin, compared with administration of imipenem alone. Similarly, decreases in the Cmax (34.5%), AUC (11.6%), Cmax/MIC (34.5%) and AUC/MIC (11.7%) of amikacin were observed when it was administered with imipenem. Bacterial counts in lungs were reduced by imipenem (p 0.004) with the imipenem-susceptible strain, and by amikacin (p 0.001) with the imipenem-resistant strain. The combination of imipenem plus amikacin was inferior to imipenem alone with the imipenem-susceptible strain (p 0.01), despite their in-vitro synergy, and was inferior to amikacin alone with the imipenem-resistant strain (p < 0.0001). In summary, combined use of imipenem with amikacin was less efficacious than monotherapy, probably because of a drug-drug interaction that resulted in decreased pharmacokinetic and pharmacodynamic parameters for both antimicrobial agents.


Assuntos
Infecções por Acinetobacter/microbiologia , Acinetobacter baumannii/efeitos dos fármacos , Amicacina/farmacologia , Amicacina/uso terapêutico , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Imipenem/farmacologia , Imipenem/uso terapêutico , Pneumonia Bacteriana/microbiologia , Infecções por Acinetobacter/tratamento farmacológico , Infecções por Acinetobacter/metabolismo , Animais , Modelos Animais de Doenças , Farmacorresistência Bacteriana Múltipla , Sinergismo Farmacológico , Quimioterapia Combinada , Feminino , Cobaias , Pulmão/efeitos dos fármacos , Pulmão/microbiologia , Testes de Sensibilidade Microbiana , Pneumonia Bacteriana/tratamento farmacológico , Pneumonia Bacteriana/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA