Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Antimicrob Agents Chemother ; : e0059524, 2024 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-39133023

RESUMO

Bacillus anthracis, the causative agent of anthrax, is among the most likely bacterial pathogens to be used in a biological attack. Inhalation anthrax is a serious, life-threatening form of infection, and the mortality from acute inhaled anthrax can approach 100% if not treated early and aggressively. Food and Drug Administration-approved antibiotics indicated for post-exposure prophylaxis (PEP) or treatment of anthrax are limited. This study assessed the in vitro activity and in vivo efficacy of omadacycline and comparators against clinical isolates of B. anthracis, including a ciprofloxacin-resistant isolate. Minimum inhibitory concentrations (MICs) of omadacycline, ciprofloxacin, and doxycycline were determined against animal and human clinical isolates of B. anthracis, including the ciprofloxacin-resistant Ames strain BACr4-2. Mice were challenged with aerosolized BACr4-2 spores, and survival was monitored for 28 days post-challenge. Treatment was initiated 24 h after aerosol challenge and administered for 14 days. Omadacycline demonstrated in vitro activity against 53 B. anthracis isolates with an MIC range of ≤0.008-0.25 µg/mL, and an MIC50/MIC90 of 0.015/0.03 µg/mL. Consistent with this, omadacycline demonstrated in vivo efficacy in a PEP mouse model of inhalation anthrax caused by the Ames BACr4-2 ciprofloxacin-resistant B. anthracis isolate. Omadacycline treatment significantly increased survival compared with the vehicle control group and the ciprofloxacin treatment group. As antibiotic resistance rates continue to rise worldwide, omadacycline may offer an alternative PEP or treatment option against inhalation anthrax, including anthrax caused by antibiotic-resistant B. anthracis.

2.
Ann Pharmacother ; 57(2): 184-192, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-35656828

RESUMO

OBJECTIVE: The objective of this systematic review is to summarize in vitro, preclinical, and human data related to omadacycline and Clostridioides difficile infection (CDI). DATA SOURCES: PubMed and Google Scholar were searched for "omadacycline" AND ("Clostridium difficile" OR "C difficile" OR "Clostridioides difficile") for any studies published before February 15, 2022. The US Food and Drug Administration (FDA) Adverse Events Reporting System (AERS) was searched for omadacycline (for reports including "C. difficile" or "CDI" or "gastrointestinal infection"). The publications list publicly available at Paratek Pharmaceuticals, Inc. Web site was reviewed. STUDY SELECTION AND DATA EXTRACTION: Publications presenting primary data on omadacycline and C. difficile published in English were included. DATA SYNTHESIS: Preclinical and clinical evidence was extracted from 14 studies. No case reports in indexed literature and no reports on FDA AERS were found. Omadacycline has potent in vitro activity against many C. difficile clinical strains and diverse ribotypes. In phase 3 studies, there were no reports of CDI in patients who received omadacycline for either community-acquired bacterial pneumonia or acute bacterial skin and skin structure infection. RELEVANCE TO PATIENT CARE AND CLINICAL PRACTICE: Omadacycline should be considered a low-risk antibiotic regarding its propensity to cause CDI. CONCLUSIONS: Reducing the burden of CDI on patients and the health care system should be a priority. Patients with appropriate indications who are at heightened risk of CDI may be suitable candidates for omadacycline therapy. In these patients, omadacycline may be preferable to antibiotics with a high CDI risk.


Assuntos
Clostridioides difficile , Infecções por Clostridium , Infecções Comunitárias Adquiridas , Humanos , Clostridioides , Antibacterianos/efeitos adversos , Infecções por Clostridium/tratamento farmacológico , Infecções por Clostridium/epidemiologia , Bactérias , Infecções Comunitárias Adquiridas/tratamento farmacológico
3.
Antimicrob Agents Chemother ; 66(1): e0170421, 2022 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-34662184

RESUMO

The incidence of nontuberculous mycobacterial diseases in the United States is rising and has surpassed that of tuberculosis. Most notable among the nontuberculous mycobacteria is Mycobacteroides abscessus, an emerging environmental opportunistic pathogen capable of causing chronic infections. M. abscessus disease is difficult to treat, and the current treatment recommendations include repurposed antibiotics, several of which are associated with undesirable side effects. In this study, we have evaluated the activity of omadacycline, a new tetracycline derivative, against M. abscessus using in vitro and in vivo approaches. Omadacycline exhibited an MIC90 of 0.5 µg/mL against a panel of 32 contemporary M. abscessus clinical isolates, several of which were resistant to antibiotics that are commonly used for treatment of M. abscessus disease. Omadacycline combined with clarithromycin, azithromycin, cefdinir, rifabutin, or linezolid also exhibited synergism against several M. abscessus strains and did not exhibit antagonism when combined with an additional nine antibiotics also commonly considered to treat M. abscessus disease. Concentration-dependent activity of omadacycline was observed in time-kill assessments. Efficacy of omadacycline was evaluated in a mouse model of lung infection against four M. abscessus strains. A dose equivalent to the 300-mg standard oral human dose was used. Compared to the untreated control group, within 4 weeks of treatment, 1 to 3 log10 fewer M. abscessus CFU were observed in the lungs of mice treated with omadacycline. Treatment outcome was biphasic, with bactericidal activity observed after the first 2 weeks of treatment against all four M. abscessus strains.


Assuntos
Infecções por Mycobacterium não Tuberculosas , Mycobacterium abscessus , Animais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Claritromicina/farmacologia , Camundongos , Testes de Sensibilidade Microbiana , Infecções por Mycobacterium não Tuberculosas/tratamento farmacológico , Infecções por Mycobacterium não Tuberculosas/microbiologia , Tetraciclinas/farmacologia , Tetraciclinas/uso terapêutico
4.
Antimicrob Agents Chemother ; 66(1): e0170321, 2022 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-34723626

RESUMO

Omadacycline, vancomycin, and rifampin, as well as rifampin combination therapies, were evaluated in an experimental rat model of methicillin-resistant Staphylococcus aureus (MRSA) osteomyelitis. All treatment groups had less MRSA recovered than saline-treated animals. The emergence of rifampin resistance was observed in 3 of 16 animals with rifampin monotherapy and none with rifampin combination therapy. After treatment, the median tibial bacterial loads were 6.04, 0.1, 4.81, and 5.24 log10 CFU/g for saline-, rifampin-, vancomycin-, and omadacycline-treated animals, respectively. Omadacycline or vancomycin administered with rifampin yielded no detectable MRSA. Omadacycline administered with rifampin deserves evaluation in humans as a potential treatment for osteomyelitis.


Assuntos
Staphylococcus aureus Resistente à Meticilina , Osteomielite , Infecções Estafilocócicas , Animais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Quimioterapia Combinada , Testes de Sensibilidade Microbiana , Osteomielite/tratamento farmacológico , Osteomielite/microbiologia , Ratos , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/microbiologia , Tetraciclinas
6.
Artigo em Inglês | MEDLINE | ID: mdl-31451507

RESUMO

New drugs with novel mechanisms of resistance are desperately needed to address both community and nosocomial infections due to Gram-negative bacteria. One such potential target is LpxC, an essential enzyme that catalyzes the first committed step of lipid A biosynthesis. Achaogen conducted an extensive research campaign to discover novel LpxC inhibitors with activity against Pseudomonas aeruginosa We report here the in vitro antibacterial activity and pharmacodynamics of ACHN-975, the only molecule from these efforts and the first ever LpxC inhibitor to be evaluated in phase 1 clinical trials. In addition, we describe the profiles of three additional LpxC inhibitors that were identified as potential lead molecules. These efforts did not produce an additional development candidate with a sufficiently large therapeutic window and the program was subsequently terminated.


Assuntos
Antibacterianos/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Pseudomonas aeruginosa/efeitos dos fármacos , Catálise/efeitos dos fármacos , Humanos , Pseudomonas aeruginosa/metabolismo
7.
Artigo em Inglês | MEDLINE | ID: mdl-29866862

RESUMO

Plazomicin and comparator agents were tested by using the CLSI reference broth microdilution method against 4,825 clinical isolates collected during 2014 and 2015 in 70 U.S. hospitals as part of the ALERT (Antimicrobial Longitudinal Evaluation and Resistance Trends) program. Plazomicin (MIC50/MIC90, 0.5/2 µg/ml) inhibited 99.2% of 4,362 Enterobacteriaceae at ≤4 µg/ml. Amikacin, gentamicin, and tobramycin inhibited 98.9%, 90.3%, and 90.3% of these isolates, respectively, by applying CLSI breakpoints. The activities of plazomicin were similar among Enterobacteriaceae species, with MIC50 values ranging from 0.25 to 1 µg/ml, with the exception of Proteus mirabilis and indole-positive Proteeae that displayed MIC50 values of 2 µg/ml. For 97 carbapenem-resistant Enterobacteriaceae (CRE), which included 87 isolates carrying blaKPC, plazomicin inhibited all but 1 isolate at ≤2 µg/ml (99.0% and 98.9%, respectively). Amikacin and gentamicin inhibited 64.9% and 56.7% of the CRE isolates at the respective CLSI breakpoints. Plazomicin inhibited 96.5 and 95.5% of the gentamicin-resistant isolates, 96.9 and 96.5% of the tobramycin-resistant isolates, and 64.3 and 90.0% of the amikacin-resistant isolates according to CLSI and EUCAST breakpoints, respectively. The activities of plazomicin against Pseudomonas aeruginosa (MIC50/MIC90, 4/16 µg/ml) and Acinetobacter species (MIC50/MIC90, 2/16 µg/ml) isolates were similar. Plazomicin was active against coagulase-negative staphylococci (MIC50/MIC90, 0.12/0.5 µg/ml) and Staphylococcus aureus (MIC50/MIC90, 0.5/0.5 µg/ml) but had limited activity against Enterococcus spp. (MIC50/MIC90, 16/64 µg/ml) and Streptococcus pneumoniae (MIC50/MIC90, 32/64 µg/ml). Plazomicin activity against the Enterobacteriaceae tested, including CRE and isolates carrying blaKPC from U.S. hospitals, supports the development plan for plazomicin to treat serious infections caused by resistant Enterobacteriaceae in patients with limited treatment options.


Assuntos
Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Enterobacteriáceas Resistentes a Carbapenêmicos/efeitos dos fármacos , Pseudomonas aeruginosa/efeitos dos fármacos , Sisomicina/análogos & derivados , Staphylococcus aureus/efeitos dos fármacos , beta-Lactamases/genética , Acinetobacter/efeitos dos fármacos , Acinetobacter/genética , Acinetobacter/crescimento & desenvolvimento , Acinetobacter/isolamento & purificação , Amicacina/farmacologia , Proteínas de Bactérias/metabolismo , Enterobacteriáceas Resistentes a Carbapenêmicos/genética , Enterobacteriáceas Resistentes a Carbapenêmicos/crescimento & desenvolvimento , Enterobacteriáceas Resistentes a Carbapenêmicos/isolamento & purificação , Expressão Gênica , Gentamicinas/farmacologia , Infecções por Bactérias Gram-Negativas/tratamento farmacológico , Infecções por Bactérias Gram-Negativas/epidemiologia , Infecções por Bactérias Gram-Negativas/microbiologia , Infecções por Bactérias Gram-Positivas/tratamento farmacológico , Infecções por Bactérias Gram-Positivas/epidemiologia , Infecções por Bactérias Gram-Positivas/microbiologia , Hospitais , Humanos , Testes de Sensibilidade Microbiana , Plasmídeos/química , Plasmídeos/metabolismo , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/crescimento & desenvolvimento , Pseudomonas aeruginosa/isolamento & purificação , Sisomicina/farmacologia , Staphylococcus aureus/genética , Staphylococcus aureus/crescimento & desenvolvimento , Staphylococcus aureus/isolamento & purificação , Streptococcus pneumoniae/efeitos dos fármacos , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/crescimento & desenvolvimento , Streptococcus pneumoniae/isolamento & purificação , Tobramicina/farmacologia , Estados Unidos/epidemiologia , beta-Lactamases/metabolismo
8.
J Antimicrob Chemother ; 73(12): 3346-3354, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30219857

RESUMO

Background: Plazomicin is a next-generation aminoglycoside that was developed to overcome common aminoglycoside-resistance mechanisms. Objectives: We evaluated the activity of plazomicin and comparators against clinical isolates collected from 26 European and adjacent countries during 2014 and 2015 as part of the Antimicrobial Longitudinal Evaluation and Resistance Trends (ALERT) global surveillance programme. Methods: All 4680 isolates collected from 45 hospitals were tested for susceptibility to antimicrobials using the reference broth microdilution method. Selected isolates were screened for genes encoding carbapenemases, aminoglycoside-modifying enzymes (AMEs) and 16S rRNA methyltransferases. Results: Plazomicin (MIC50/90 0.5/2 mg/L) inhibited 95.8% of Enterobacteriaceae at ≤2 mg/L, including carbapenem-resistant Enterobacteriaceae (MIC50/90 0.25/128 mg/L). Plazomicin was more active compared with other aminoglycosides against isolates carrying blaKPC (MIC50/90 0.25/2 mg/L), isolates carrying blaOXA-48-like (MIC50/90 0.25/16 mg/L) and carbapenemase-negative isolates (MIC50/90 0.25/1 mg/L). Approximately 60% of the isolates harbouring blaVIM and blaNDM-1 carried 16S rRNA methyltransferases (mainly rmtB and armA). AME genes were detected among 728 isolates and 99.0% of these were inhibited by plazomicin at ≤2 mg/L. Plazomicin activity against Pseudomonas aeruginosa (MIC50/90 4/8 mg/L) was similar to amikacin activity (MIC50/90 2/16 mg/L). Plazomicin demonstrated activity against CoNS (MIC50/90 0.12/0.25 mg/L) and Staphylococcus aureus (MIC50/90 0.5/1 mg/L). Plazomicin activity was limited against Acinetobacter spp. (MIC50/90 8/>128 mg/L), Enterococcus spp. (MIC50/90 32/128 mg/L) and Streptococcus pneumoniae (MIC50/90 32/64 mg/L). Conclusions: Plazomicin demonstrated activity against Enterobacteriaceae isolates tested in this study, including isolates carrying AMEs and a high percentage of the carbapenem-non-susceptible isolates. Plazomicin displayed activity against staphylococci.


Assuntos
Aminoglicosídeos/farmacologia , Antibacterianos/farmacologia , Farmacorresistência Bacteriana Múltipla , Enterobacteriaceae/efeitos dos fármacos , Enterobacteriaceae/genética , Sisomicina/análogos & derivados , Enterobacteriáceas Resistentes a Carbapenêmicos/efeitos dos fármacos , Enterobacteriáceas Resistentes a Carbapenêmicos/genética , Enterobacteriáceas Resistentes a Carbapenêmicos/isolamento & purificação , Enterobacteriaceae/enzimologia , Infecções por Enterobacteriaceae/microbiologia , Monitoramento Epidemiológico , Europa (Continente) , Humanos , Testes de Sensibilidade Microbiana , RNA Ribossômico 16S/genética , Sisomicina/farmacologia
10.
mSphere ; 9(7): e0038124, 2024 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-38980071

RESUMO

Treatment outcomes for Mycobacteroides abscessus (Mab, also known as Mycobacterium abscessus) disease are still unsatisfactory, mainly due to issues with drug toxicity, tolerability, and efficacy. Treating Mab disease is challenging due to its high baseline antibiotic resistance, initial requirement for intravenous therapy, and poor medication tolerance. Omadacycline, a new tetracycline, is active against Mab. Since any new antibiotic effective against Mab is expected to be used in combination with other antibiotics, we evaluated the efficacy of two triple-drug combinations comprising omadacycline, omadacycline + amikacin + imipenem, and omadacycline + clofazimine + linezolid against two contemporary Mab clinical isolates in a mouse model of Mab lung disease. Antibiotic administration was initiated 1-week post-infection and was given daily, with Mab burden in the lungs at treatment completion serving as the endpoint. Omadacycline alone moderately reduced Mab levels and maintained better health in mice compared to untreated ones, which typically suffered from the infection. The omadacycline + clofazimine + linezolid combination showed immediate bactericidal activity and enhanced efficacy over 6 weeks, particularly against the more resistant strain (M9507). However, the clofazimine + linezolid combination lacked early bactericidal activity. When combined with amikacin and imipenem, omadacycline did not improve the regimen's effectiveness over 4 weeks of treatment. Our study showed that omadacycline + clofazimine + linezolid exhibited significant bactericidal activity over an extended treatment duration. However, adding omadacycline to amikacin and imipenem did not improve regimen effectiveness against the evaluated clinical isolates within 4 weeks. Further research in Mab disease patients is needed to determine the most effective omadacycline-containing regimen.IMPORTANCEMycobacteroides abscessus is a common environmental bacterium that causes infections in people with compromised lung function, including those with bronchiectasis, cystic fibrosis, chronic obstructive pulmonary disease, and weakened immune systems, especially among older individuals. Treating M. abscessus disease is challenging due to the limited effectiveness and toxicity of current antibiotics, which often require prolonged use. Omadacycline, a new antibiotic, shows promise against M. abscessus. Using a mouse model that mimics M. abscessus disease in humans, we studied the effectiveness of including omadacycline with recommended antibiotics. Adding omadacycline to clofazimine and linezolid significantly improved treatment outcomes, rapidly clearing the bacteria from the lungs and maintaining effectiveness throughout. This oral combination is convenient for patients. However, adding omadacycline to amikacin and imipenem did not improve treatment effectiveness within 4 weeks. Further study with M. abscessus patients is necessary to optimize omadacycline-based treatment strategies for this disease.


Assuntos
Amicacina , Antibacterianos , Clofazimina , Modelos Animais de Doenças , Quimioterapia Combinada , Imipenem , Linezolida , Infecções por Mycobacterium não Tuberculosas , Mycobacterium abscessus , Tetraciclinas , Animais , Clofazimina/administração & dosagem , Clofazimina/uso terapêutico , Linezolida/administração & dosagem , Linezolida/uso terapêutico , Camundongos , Antibacterianos/administração & dosagem , Antibacterianos/uso terapêutico , Amicacina/administração & dosagem , Amicacina/uso terapêutico , Tetraciclinas/administração & dosagem , Tetraciclinas/uso terapêutico , Tetraciclinas/farmacologia , Mycobacterium abscessus/efeitos dos fármacos , Imipenem/administração & dosagem , Imipenem/uso terapêutico , Imipenem/farmacologia , Infecções por Mycobacterium não Tuberculosas/tratamento farmacológico , Infecções por Mycobacterium não Tuberculosas/microbiologia , Feminino , Resultado do Tratamento , Testes de Sensibilidade Microbiana , Pneumopatias/tratamento farmacológico , Pneumopatias/microbiologia , Administração Oral , Pulmão/microbiologia
11.
Tuberculosis (Edinb) ; 146: 102482, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38364332

RESUMO

Mycobacteroides abscessus (Mab, also known as Mycobacterium abscessus) causes opportunistic pulmonary and soft tissue infections that are difficult to cure with existing treatments. Omadacycline, a new tetracycline antibiotic, exhibits potent in vitro and in vivo activity against Mab. As regimens containing multiple antibiotics are required to produce a durable cure for Mab disease, we assessed efficacies of three three-drug combinations in a pre-clinical mouse model of pulmonary Mab disease to identify companion drugs with which omadacycline exhibits the highest efficacy. Additionally, we assessed the susceptibility of Mab recovered from mouse lungs after four weeks of exposure to the three triple-drug regimens. Among the three-drug regimens, omadacycline + imipenem + amikacin produced the largest reduction in Mab burden, whereas omadacycline + imipenem + linezolid exhibited the most effective early bactericidal activity. Omadacycline + linezolid + clofazimine, a regimen that can be administered orally, lacked early bactericidal activity but produced a gradual reduction in the lung Mab burden over time. The robust efficacy exhibited by these three regimens in the mouse model supports their further evaluation in patients with Mab lung disease. As we were unable to isolate drug-resistant Mab mutants at the completion of four weeks of treatment, these triple-drug combinations show promise of producing durable cure and minimizing selection of resistant mutants.


Assuntos
Infecções por Mycobacterium não Tuberculosas , Mycobacterium abscessus , Mycobacterium tuberculosis , Humanos , Animais , Camundongos , Linezolida/farmacologia , Infecções por Mycobacterium não Tuberculosas/tratamento farmacológico , Infecções por Mycobacterium não Tuberculosas/microbiologia , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Tetraciclinas/farmacologia , Tetraciclinas/uso terapêutico , Imipenem/farmacologia , Combinação de Medicamentos , Testes de Sensibilidade Microbiana
12.
J Bacteriol ; 195(7): 1525-37, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23354745

RESUMO

Previously, it was shown that an aconitase (citB) null mutation results in a vast overaccumulation of citrate in the culture fluid of growing Bacillus subtilis cells, a phenotype that causes secondary effects, including the hyperexpression of the citB promoter. B. subtilis aconitase is a bifunctional protein; to determine if either or both activities of aconitase were responsible for this phenotype, two strains producing different mutant forms of aconitase were constructed, one designed to be enzymatically inactive (C450S [citB2]) and the other designed to be defective in RNA binding (R741E [citB7]). The citB2 mutant was a glutamate auxotroph and accumulated citrate, while the citB7 mutant was a glutamate prototroph. Unexpectedly, the citB7 strain also accumulated citrate. Both mutant strains exhibited overexpression of the citB promoter and accumulated high levels of aconitase protein. These strains and the citB null mutant also exhibited increased levels of citrate synthase protein and enzyme activity in cell extracts, and the major citrate synthase (citZ) transcript was present at higher-than-normal levels in the citB null mutant, due at least in part to a >3-fold increase in the stability of the citZ transcript compared to the wild type. Purified B. subtilis aconitase bound to the citZ 5' leader RNA in vitro, but the mutant proteins did not. Together, these data suggest that wild-type aconitase binds to and destabilizes the citZ transcript in order to maintain proper cell homeostasis by preventing the overaccumulation of citrate.


Assuntos
Aconitato Hidratase/metabolismo , Bacillus subtilis/genética , Bacillus subtilis/metabolismo , Ciclo do Ácido Cítrico/genética , Regulação Bacteriana da Expressão Gênica , Aconitato Hidratase/genética , Sítios de Ligação , Ácido Cítrico/metabolismo , DNA Bacteriano/metabolismo , Perfilação da Expressão Gênica , Ácido Glutâmico/metabolismo , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo
13.
Cell Microbiol ; 14(4): 529-45, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22188208

RESUMO

Rickettsiae are obligate intracellular pathogens that are transmitted to humans by arthropod vectors and cause diseases such as spotted fever and typhus. Although rickettsiae require the host cell actin cytoskeleton for invasion, the cytoskeletal proteins that mediate this process have not been completely described. To identify the host factors important during cell invasion by Rickettsia parkeri, a member of the spotted fever group (SFG), we performed an RNAi screen targeting 105 proteins in Drosophila melanogaster S2R+ cells. The screen identified 21 core proteins important for invasion, including the GTPases Rac1 and Rac2, the WAVE nucleation-promoting factor complex and the Arp2/3 complex. In mammalian cells, including endothelial cells, the natural targets of R. parkeri, the Arp2/3 complex was also crucial for invasion, while requirements for WAVE2 as well as Rho GTPases depended on the particular cell type. We propose that R. parkeri invades S2R+ arthropod cells through a primary pathway leading to actin nucleation, whereas invasion of mammalian endothelial cells occurs via redundant pathways that converge on the host Arp2/3 complex. Our results reveal a key role for the WAVE and Arp2/3 complexes, as well as a higher degree of variation than previously appreciated in actin nucleation pathways activated during Rickettsia invasion.


Assuntos
Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/microbiologia , Rickettsia/patogenicidade , Família de Proteínas da Síndrome de Wiskott-Aldrich/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Complexo 2-3 de Proteínas Relacionadas à Actina/genética , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Células COS , Linhagem Celular , Chlorocebus aethiops , Proteínas de Drosophila/genética , Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Células Endoteliais/microbiologia , Imunofluorescência , Interações Hospedeiro-Patógeno , Humanos , Camundongos , Viabilidade Microbiana , Plasmídeos/genética , Plasmídeos/metabolismo , Interferência de RNA , Rickettsia/genética , Rickettsia/metabolismo , Infecções por Rickettsia/microbiologia , Transdução de Sinais , Transfecção , Células Vero , Família de Proteínas da Síndrome de Wiskott-Aldrich/genética , Proteínas rac de Ligação ao GTP/genética
14.
mSphere ; 8(2): e0066522, 2023 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-36912629

RESUMO

Mycobacteroides abscessus is an opportunistic pathogen in people with structural lung conditions such as bronchiectasis, chronic obstructive pulmonary disease, and cystic fibrosis. Pulmonary M. abscessus infection causes progressive symptomatic and functional decline as well as diminished lung function and is often incurable with existing antibiotics. We investigated the efficacy of a new tetracycline, omadacycline, in combination with existing antibiotics recommended to treat this indication, in a mouse model of M. abscessus lung disease. Amikacin, azithromycin, bedaquiline, biapenem, cefoxitin, clofazimine, imipenem, linezolid, and rifabutin were selected as companions to omadacycline. M. abscessus burden in the lungs of mice over a 4-week treatment duration was considered the endpoint. Omadacycline in combination with linezolid, imipenem, cefoxitin, biapenem, or rifabutin exhibited early bactericidal activity compared to any single drug. Using three M. abscessus isolates, we also determined the in vitro frequency of spontaneous resistance against omadacycline to be between 1.9 × 10-10 and 6.2 × 10-10 and the frequency of persistence against omadacycline to be between 5.3 × 10-6 and 1.3 × 10-5. Based on these findings, the combination of omadacycline and select drugs that are included in the recent treatment guidelines may exhibit improved potency to treat M. abscessus lung disease. IMPORTANCE M. abscessus disease incidence is increasing in the United States. This disease is difficult to cure with existing antibiotics. In this study, we describe the efficacy of a new tetracycline antibiotic, omadacycline, in combination with an existing antibiotic to treat this disease. A mouse model of M. abscessus lung disease was used to assess the efficacies of these experimental treatment regimens. Omadacycline in combination with select existing antibiotics exhibited bactericidal activity during the early phase of treatment.


Assuntos
Fibrose Cística , Mycobacterium abscessus , Animais , Camundongos , Linezolida , Cefoxitina , Testes de Sensibilidade Microbiana , Antibacterianos/uso terapêutico , Tetraciclinas/uso terapêutico , Imipenem , Rifabutina
15.
Mil Med ; 2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-37963013

RESUMO

INTRODUCTION: Combat-related wound infections complicate the recovery of wounded military personnel, contributing to overall morbidity and mortality. Wound infections in combat settings present unique challenges because of the size and depth of the wounds, the need to administer emergency care in the field, and the need for subsequent treatment in military facilities. Given the increase in multidrug-resistant pathogens, a novel, broad-spectrum antibiotic is desired across this continuum of care when the standard of care fails. Omadacycline was FDA-approved in 2018 for treatment of adults with acute bacterial skin and skin structure infections (ABSSSI), as well as community-acquired bacterial pneumonia (CABP). It is a broad-spectrum antibiotic with activity against gram-positive, gram-negative, and atypical bacterial pathogens, including multidrug-resistant species. Omadacycline can overcome commonly reported tetracycline resistance mechanisms, ribosomal protection proteins, and efflux pumps, and is available in once-daily intravenous or oral formulations. In this review, we discuss the potential role of omadacycline, which is included in the Department of Defense Formulary, in the context of combat wound infections. MATERIALS AND METHODS: A literature review was undertaken for manuscripts published before July 21, 2023. This included a series of publications found via PubMed and a bibliography made publicly available on the Paratek Pharmaceuticals, Inc. website. Publications presenting primary data published in English on omadacycline in relation to ESKAPEE (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, Escherichia coli, and Enterobacter species) pathogens and Clostridioides difficile, including in vitro, in vivo, and clinical data were included. RESULTS: Of 260 identified records, 66 were included for evidence review. Omadacycline has in vitro activity against almost all the ESKAPEE pathogens, apart from P. aeruginosa. Importantly, it has activity against the four most prevalent bacterial pathogens that cause wound infections in the military healthcare system: S. aureus, including methicillin-resistant S. aureus, A. baumannii, K. pneumoniae, and E. coli. In vivo studies in rats have shown that omadacycline is rapidly distributed in most tissues, with the highest tissue-to-blood concentration ratios in bone mineral. The clinical efficacy of omadacycline has been assessed in three separate Phase 3 studies in patients with ABSSSI (OASIS-1 and OASIS-2) and with CABP (OPTIC). Overall, omadacycline has an established safety profile in the treatment of both ABSSSI and CABP. CONCLUSIONS: Omadacycline has broad-spectrum activity, the option to be orally administered and an established safety profile, making it a potentially attractive replacement for moxifloxacin in the military individual first aid kit, especially when accounting for the increasing resistance to fluoroquinolones. Further studies and clinical evaluation are warranted to support broader use of omadacycline to treat combat wound infections in the military healthcare system.

16.
JAC Antimicrob Resist ; 4(1): dlab190, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34988444

RESUMO

OBJECTIVES: To evaluate the effect of sub-growth-inhibitory concentrations of omadacycline on Staphylococcus aureus ATCC 10832 haemolytic activity in vitro. METHODS: Following determination of the MICs of omadacycline and comparator antibiotics, the strain was grown in the presence of individual antibiotics and the percentage of haemolysis assayed; 'washout' experiments were performed with omadacycline only. RESULTS: Omadacycline inhibited S. aureus haemolytic activity in vitro at sub-growth-inhibitory concentrations. Inhibition was maintained at least 4 h after removal of extracellular drug. CONCLUSIONS: Omadacycline's in vitro potency and suppression of virulence factors might contribute to its efficacy in the treatment of acute bacterial skin and skin structure infections and community-acquired bacterial pneumonia caused by virulent strains of S. aureus. This finding could be relevant for other organisms and virulence factors that depend on new protein synthesis.

17.
Diagn Microbiol Infect Dis ; 104(3): 115785, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35988350

RESUMO

This study used surveillance data from a global program of clinical bacterial isolates to determine whether a tetracycline susceptible result can be used to predict an omadacycline susceptible result. Categorical agreement, very major error rates, and minor error rates were calculated for Staphylococcus aureus (MSSA and MRSA; n=38,364), S. lugdunensis (n=335), Streptococcus pneumoniae (n=11,725), S. pyogenes (n=3,390), S. anginosus group (n=622), Haemophilus spp. (n=6,419), Enterococcus faecalis (n=7,065), Klebsiella pneumoniae (n=10,313), and Enterobacter cloacae (n=4,418). Across the organisms, for which omadacycline has an FDA breakpoint established, a tetracycline susceptible result showed ≥96.3% categorical agreement in predicting an omadacycline susceptible result. The rates of very major errors were below the guideline-suggested level (<1.5%). Omadacycline retained activity against most (88.7-100% Gram-positive and 54-98.6% Gram-negative) tetracycline-resistant isolates. For laboratories that do not have the capability to perform susceptibility testing for omadacycline, one-sided surrogate testing for tetracycline can be a practical alternative.


Assuntos
Antibacterianos , Tetraciclinas , Antibacterianos/farmacologia , Humanos , Testes de Sensibilidade Microbiana , Tetraciclina/farmacologia , Tetraciclinas/farmacologia
18.
J Glob Antimicrob Resist ; 30: 96-99, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35500838

RESUMO

OBJECTIVES: Antibiotics are associated with increased risk of Clostridioides difficile infection, which has limited treatment options. We assessed in vitro activity of omadacycline (an aminomethylcycline antibiotic) against the C. difficile infection strain and efficacy in a hamster model of C. difficile-associated diarrhoea. METHODS: Omadacycline, clindamycin, tigecycline, vancomycin, and metronidazole minimum inhibitory concentrations (MICs) for the infection-model strain (C. difficile ATCC 43596) were determined. Hamsters were pretreated with subcutaneous clindamycin (10 mg/kg) and infected 24 h later with C. difficile ATCC 43596; 24 h post infection, they received oral omadacycline (50 mg/kg/day), vancomycin (50 mg/kg/day), or vehicle for 5 days. Efficacy was reported as survival. RESULTS: Omadacycline was as active as tigecycline, vancomycin, and metronidazole (MIC 0.06 mg/L); clindamycin showed no activity. Median survival in hamsters was: 12 days, omadacycline; 2 days, vancomycin; 4 days, clindamycin pretreatment only. CONCLUSION: Omadacycline exhibited potent in vitro activity against C. difficile and showed efficacy in a model of C. difficile-associated diarrhoea.


Assuntos
Clostridioides difficile , Animais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Clindamicina/farmacologia , Clindamicina/uso terapêutico , Clostridioides , Cricetinae , Diarreia/tratamento farmacológico , Metronidazol/farmacologia , Metronidazol/uso terapêutico , Tetraciclinas , Tigeciclina , Vancomicina/farmacologia , Vancomicina/uso terapêutico
20.
Open Forum Infect Dis ; 6(4): ofz123, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30968059

RESUMO

Plazomicin is an aminoglycoside that was approved in June 2018 by the US Food and Drug Administration for the treatment of complicated urinary tract infections, including pyelonephritis, due to Escherichia coli, Klebsiella pneumoniae, Enterobacter cloacae, and Proteus mirabilis. Plazomicin was engineered to overcome the most common aminoglycoside resistance mechanism, inactivation by aminoglycoside-modifying enzymes, but is not active against the less common 16S ribosomal RNA methyltransferases (16S-RMTase), which confer target site modification. As an aminoglycoside, plazomicin maintains activity against Enterobacteriaceae that express resistance mechanisms to other antibiotic classes, including metallo-ß-lactamases. Therefore, in the absence of a 16S-RMTase, plazomicin is active against metallo-ß-lactamase-producing Enterobacteriaceae.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA