Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
BMC Infect Dis ; 16(1): 621, 2016 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-27809794

RESUMO

BACKGROUND: Bacillus anthracis, the causative agent of anthrax, is a spore forming and toxin producing rod-shaped bacterium that is classified as a category A bioterror agent. This pathogenic microbe can be transmitted to both animals and humans. Clinical presentation depends on the route of entry (direct contact, ingestion, injection or aerosolization) with symptoms ranging from isolated skin infections to more severe manifestations such as cardiac or pulmonary shock, meningitis, and death. To date, anthrax is treatable if antibiotics are administered promptly and continued for 60 days. However, if treatment is delayed or administered improperly, the patient's chances of survival are decreased drastically. In addition, antibiotics are ineffective against the harmful anthrax toxins and spores. Therefore, alternative therapeutics are essential. In this review article, we explore and discuss advances that have been made in anthrax therapy with a primary focus on alternative pre-approved and novel antibiotics as well as anti-toxin therapies. METHODS: A literature search was conducted using the University of Manitoba search engine. Using this search engine allowed access to a greater variety of journals/articles that would have otherwise been restricted for general use. In order to be considered for discussion for this review, all articles must have been published later than 2009. RESULTS: The alternative pre-approved antibiotics demonstrated high efficacy against B. anthracis both in vitro and in vivo. In addition, the safety profile and clinical pharmacology of these drugs were already known. Compounds that targeted underexploited bacterial processes (DNA replication, RNA synthesis, and cell division) were also very effective in combatting B. anthracis. In addition, these novel compounds prevented bacterial resistance. Targeting B. anthracis virulence, more specifically the anthrax toxins, increased the length of which treatment could be administered. CONCLUSIONS: Several novel and pre-existing antibiotics, as well as toxin inhibitors, have shown increasing promise. A combination treatment that targets both bacterial growth and toxin production would be ideal and probably necessary for effectively combatting this armed bacterium.


Assuntos
Antraz/tratamento farmacológico , Antibacterianos/uso terapêutico , Antitoxinas/uso terapêutico , alfa-Globulinas/uso terapêutico , Antibióticos Antineoplásicos/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Antígenos de Bactérias , Bacillus anthracis , Toxinas Bacterianas , DNA Helicases/antagonistas & inibidores , Daunorrubicina/análogos & derivados , Daunorrubicina/uso terapêutico , Doxorrubicina/uso terapêutico , Descoberta de Drogas , Fluoroquinolonas , Humanos , Indutores de Interferon/uso terapêutico , Levofloxacino , Linezolida , Moxifloxacina , Ofloxacino , Policetídeos/uso terapêutico , Inibidores de Serina Proteinase/uso terapêutico , Tilorona/uso terapêutico , Virulência
2.
Drugs ; 75(3): 253-70, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25673021

RESUMO

Tedizolid phosphate is a novel oxazolidinone prodrug (converted to the active form tedizolid by phosphatases in vivo) that has been developed and recently approved (June 2014) by the United States FDA for the treatment of acute bacterial skin and skin structure infections (ABSSSIs) caused by susceptible Gram-positive pathogens, including methicillin-resistant Staphylococcus aureus (MRSA). Tedizolid is an oxazolidinone, but differs from other oxazolidinones by possessing a modified side chain at the C-5 position of the oxazolidinone nucleus which confers activity against certain linezolid-resistant pathogens and has an optimized C- and D-ring system that improves potency through additional binding site interactions. The mechanism of action of tedizolid is similar to other oxazolidinones and occurs through inhibition of bacterial protein synthesis by binding to 23S ribosomal RNA (rRNA) of the 50S subunit of the ribosome. As with other oxazolidinones, the spontaneous frequency of resistance development to tedizolid is low. Tedizolid is four- to eightfold more potent in vivo than linezolid against all species of staphylococci, enterococci, and streptococci, including drug-resistant phenotypes such as MRSA and vancomycin-resistant enterococci (VRE) and linezolid-resistant phenotypes. Importantly, tedizolid demonstrates activity against linezolid-resistant bacterial strains harboring the horizontally transmissible cfr gene, in the absence of certain ribosomal mutations conferring reduced oxazolidinone susceptibility. With its half-life of approximately 12 h, tedizolid is dosed once daily. It demonstrates linear pharmacokinetics, has a high oral bioavailability of approximately 90 %, and is primarily excreted by the liver as an inactive, non-circulating sulphate conjugate. Tedizolid does not require dosage adjustment in patients with any degree of renal dysfunction or hepatic dysfunction. Studies in animals have demonstrated that the pharmacodynamic parameter most closely associated with the efficacy of tedizolid is fAUC(0-24h)/MIC. In non-neutropenic animals, a dose-response enhancement was observed with tedizolid and lower exposures were required compared to neutropenic cohorts. Two Phase III clinical trials have demonstrated non-inferiority of a once-daily tedizolid 200 mg dose for 6-10 days versus twice-daily 600 mg linezolid for the treatment of ABSSSIs. Both trials used the primary endpoint of early clinical response at 48-72 h; however, one trial compared oral formulations while the other initiated therapy with the parenteral formulation and allowed oral sequential therapy following initial clinical response. Throughout its development, tedizolid has demonstrated that it is well tolerated and animal studies have shown a lower propensity for neuropathies with long-term use than its predecessor linezolid. Data from the two completed Phase III clinical trials demonstrated that the studied tedizolid regimen (200 mg once daily for 6 days) had significantly less impact on hematologic parameters as well as significantly less gastrointestinal treatment-emergent adverse effects (TEAEs) than its comparator linezolid. As with linezolid, tedizolid is a weak, reversible MAO inhibitor; however, a murine head twitch model validated to assess serotonergic activity reported no increase in the number of head twitches with tedizolid even at doses that exceeded the C max in humans by up to 25-fold. Tyramine and pseudoephedrine challenge studies in humans have also reported no meaningful MAO-related interactions with tedizolid. With its enhanced in vitro activity against a broad-spectrum of Gram-positive aerobic bacteria, convenient once-daily dosing, a short 6-day course of therapy, availability of both oral and intravenous routes of administration, and an adverse effect profile that appears to be more favorable than linezolid, tedizolid is an attractive agent for use in both the hospital and community settings. Tedizolid is currently undergoing additional Phase III clinical trials for the treatment of hospital-acquired bacterial pneumonia (HABP) and ventilated nosocomial pneumonia (VNP).


Assuntos
Antibacterianos/uso terapêutico , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Infecções por Bactérias Gram-Positivas/tratamento farmacológico , Organofosfatos/uso terapêutico , Oxazóis/uso terapêutico , Dermatopatias Bacterianas/tratamento farmacológico , Animais , Antibacterianos/administração & dosagem , Antibacterianos/efeitos adversos , Antibacterianos/farmacocinética , Ensaios Clínicos como Assunto , Bactérias Gram-Positivas/efeitos dos fármacos , Bactérias Gram-Positivas/crescimento & desenvolvimento , Infecções por Bactérias Gram-Positivas/microbiologia , Humanos , Testes de Sensibilidade Microbiana , Estrutura Molecular , Organofosfatos/administração & dosagem , Organofosfatos/efeitos adversos , Organofosfatos/farmacocinética , Oxazóis/administração & dosagem , Oxazóis/efeitos adversos , Oxazóis/farmacocinética , Dermatopatias Bacterianas/microbiologia
3.
Crit Care Clin ; 29(4): 841-52, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24094380

RESUMO

Vancomycin-resistant enterococci (VRE) consist mainly of Enterococcus faecalis and E faecium, the latter mostly hospital-acquired. In addition, E gallinarum and E casseliflavus are intrinsically vancomycin-resistant and are community-acquired. VRE have become common in many hospitals throughout the world and, once established, are very difficult to eradicate. VRE are difficult to treat; therefore, infection control measures in hospitals are of prime importance in preventing the establishment of these pathogens. Most severe VRE infections will need combination therapy because many of the effective antimicrobial agents, when used alone, have only a bacteriostatic effect.


Assuntos
Aminoglicosídeos/uso terapêutico , Antibacterianos/uso terapêutico , Enterococcus/efeitos dos fármacos , Infecções por Bactérias Gram-Positivas/tratamento farmacológico , Resistência a Vancomicina , beta-Lactamas/uso terapêutico , Aminoglicosídeos/farmacologia , Antibacterianos/farmacologia , Infecção Hospitalar/tratamento farmacológico , Quimioterapia Combinada , Infecções por Bactérias Gram-Positivas/transmissão , Humanos , Controle de Infecções/métodos , Testes de Sensibilidade Microbiana , Vancomicina/farmacologia , Vancomicina/uso terapêutico , beta-Lactamas/farmacologia
4.
Expert Opin Drug Metab Toxicol ; 9(6): 789-99, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23590397

RESUMO

INTRODUCTION: Ceftobiprole is a cephalosporin with activity against methicillin-resistant Staphylococcus aureus, Enterobacteriaceae, and Pseudomonas aeruginosa with a promising role in the treatment of hospital-acquired pneumonia (HAP). Cure rates, however, with ceftobiprole at the doses studied may be inferior to conventional treatment in the ventilator-acquired subset of HAP. AREAS COVERED: Literature was sought using PubMed and through abstracts from the Interscience Conference on Antimicrobial Agents and Chemotherapy (2006 - 2012) and the European Congress of Clinical Microbiology and Infectious Diseases (2007 - 2012). The authors used the search terms "ceftobiprole," "BAL9141," "RO63-9141," "BAL5788," and 'RO5788." The article discusses the activity, mechanism of action, pharmacokinetics (PK), pharmacodynamics (PD), and clinical trials of ceftobiprole in HAP. The article also provides discussion of how PK/PD parameters play a role in the outcome of HAP treatment and how dosing in ventilator-associated pneumonia (VAP) should be reconsidered in light of altered PK/PD. EXPERT OPINION: In patients with normal PK and non-VAP, ceftobiprole is effective for the treatment of HAP in the recommended doses, ceftobiprole is unlikely to achieve the desired PD targets when PK parameters are altered in VAP (e.g., increased volume of distribution and clearance). In these settings, off-label use at higher doses may overcome these limitations; but in the presence of alternative therapies, it cannot be currently recommended.


Assuntos
Antibacterianos/uso terapêutico , Cefalosporinas/uso terapêutico , Pneumonia Bacteriana/tratamento farmacológico , Antibacterianos/farmacocinética , Antibacterianos/farmacologia , Cefalosporinas/farmacocinética , Cefalosporinas/farmacologia , Ensaios Clínicos como Assunto , Infecção Hospitalar/tratamento farmacológico , Infecção Hospitalar/microbiologia , Relação Dose-Resposta a Droga , Humanos , Pneumonia Bacteriana/microbiologia , Pneumonia Associada à Ventilação Mecânica/tratamento farmacológico , Pneumonia Associada à Ventilação Mecânica/microbiologia , Resultado do Tratamento
5.
Drugs ; 73(2): 159-77, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23371303

RESUMO

Avibactam (formerly NXL104, AVE1330A) is a synthetic non-ß-lactam, ß-lactamase inhibitor that inhibits the activities of Ambler class A and C ß-lactamases and some Ambler class D enzymes. This review summarizes the existing data published for ceftazidime-avibactam, including relevant chemistry, mechanisms of action and resistance, microbiology, pharmacokinetics, pharmacodynamics, and efficacy and safety data from animal and human trials. Although not a ß-lactam, the chemical structure of avibactam closely resembles portions of the cephem bicyclic ring system, and avibactam has been shown to bond covalently to ß-lactamases. Very little is known about the potential for avibactam to select for resistance. The addition of avibactam greatly (4-1024-fold minimum inhibitory concentration [MIC] reduction) improves the activity of ceftazidime versus most species of Enterobacteriaceae depending on the presence or absence of ß-lactamase enzyme(s). Against Pseudomonas aeruginosa, the addition of avibactam also improves the activity of ceftazidime (~fourfold MIC reduction). Limited data suggest that the addition of avibactam does not improve the activity of ceftazidime versus Acinetobacter species or most anaerobic bacteria (exceptions: Bacteroides fragilis, Clostridium perfringens, Prevotella spp. and Porphyromonas spp.). The pharmacokinetics of avibactam follow a two-compartment model and do not appear to be altered by the co-administration of ceftazidime. The maximum plasma drug concentration (C(max)) and area under the plasma concentration-time curve (AUC) of avibactam increase linearly with doses ranging from 50 mg to 2,000 mg. The mean volume of distribution and half-life of 22 L (~0.3 L/kg) and ~2 hours, respectively, are similar to ceftazidime. Like ceftazidime, avibactam is primarily renally excreted, and clearance correlates with creatinine clearance. Pharmacodynamic data suggest that ceftazidime-avibactam is rapidly bactericidal versus ß-lactamase-producing Gram-negative bacilli that are not inhibited by ceftazidime alone.Clinical trials to date have reported that ceftazidime-avibactam is as effective as standard carbapenem therapy in complicated intra-abdominal infection and complicated urinary tract infection, including infection caused by cephalosporin-resistant Gram-negative isolates. The safety and tolerability of ceftazidime-avibactam has been reported in three phase I pharmacokinetic studies and two phase II clinical studies. Ceftazidime-avibactam appears to be well tolerated in healthy subjects and hospitalized patients, with few serious drug-related treatment-emergent adverse events reported to date.In conclusion, avibactam serves to broaden the spectrum of ceftazidime versus ß-lactamase-producing Gram-negative bacilli. The exact roles for ceftazidime-avibactam will be defined by efficacy and safety data from further clinical trials. Potential future roles for ceftazidime-avibactam include the treatment of suspected or documented infections caused by resistant Gram-negative-bacilli producing extended-spectrum ß-lactamase (ESBL), Klebsiella pneumoniae carbapenemases (KPCs) and/or AmpC ß-lactamases. In addition, ceftazidime-avibactam may be used in combination (with metronidazole) for suspected polymicrobial infections. Finally, the increased activity of ceftazidime-avibactam versus P. aeruginosa may be of clinical benefit in patients with suspected or documented P. aeruginosa infections.


Assuntos
Antibacterianos/uso terapêutico , Compostos Azabicíclicos/uso terapêutico , Ceftazidima/uso terapêutico , Inibidores de beta-Lactamases , Antibacterianos/química , Antibacterianos/farmacologia , Compostos Azabicíclicos/química , Compostos Azabicíclicos/farmacologia , Infecções Bacterianas/tratamento farmacológico , Ceftazidima/química , Ceftazidima/farmacologia , Ensaios Clínicos como Assunto , Quimioterapia Combinada , Humanos , Testes de Sensibilidade Microbiana
6.
Drugs ; 70(7): 859-86, 2010 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-20426497

RESUMO

Dalbavancin, oritavancin and telavancin are semisynthetic lipoglycopeptides that demonstrate promise for the treatment of patients with infections caused by multi-drug-resistant Gram-positive pathogens. Each of these agents contains a heptapeptide core, common to all glycopeptides, which enables them to inhibit transglycosylation and transpeptidation (cell wall synthesis). Modifications to the heptapeptide core result in different in vitro activities for the three semisynthetic lipoglycopeptides. All three lipoglycopeptides contain lipophilic side chains, which prolong their half-life, help to anchor the agents to the cell membrane and increase their activity against Gram-positive cocci. In addition to inhibiting cell wall synthesis, telavancin and oritavancin are also able to disrupt bacterial membrane integrity and increase membrane permeability; oritavancin also inhibits RNA synthesis. Enterococci exhibiting the VanA phenotype (resistance to both vancomycin and teicoplanin) are resistant to both dalbavancin and telavancin, while oritavancin retains activity. Dalbavancin, oritavancin and telavancin exhibit activity against VanB vancomycin-resistant enterococci. All three lipoglycopeptides demonstrate potent in vitro activity against Staphylococcus aureus and Staphylococcus epidermidis regardless of their susceptibility to meticillin, as well as Streptococcus spp. Both dalbavancin and telavancin are active against vancomycin-intermediate S. aureus (VISA), but display poor activity versus vancomycin-resistant S. aureus (VRSA). Oritavancin is active against both VISA and VRSA. Telavancin displays greater activity against Clostridium spp. than dalbavancin, oritavancin or vancomycin. The half-life of dalbavancin ranges from 147 to 258 hours, which allows for once-weekly dosing, the half-life of oritavancin of 393 hours may allow for one dose per treatment course, while telavancin requires daily administration. Dalbavancin and telavancin exhibit concentration-dependent activity and AUC/MIC (area under the concentration-time curve to minimum inhibitory concentration ratio) is the pharmacodynamic parameter that best describes their activities. Oritavancin's activity is also considered concentration-dependent in vitro, while in vivo its activity has been described by both concentration and time-dependent models; however, AUC/MIC is the pharmacodynamic parameter that best describes its activity. Clinical trials involving patients with complicated skin and skin structure infections (cSSSIs) have demonstrated that all three agents are as efficacious as comparators. The most common adverse effects reported with dalbavancin use included nausea, diarrhoea and constipation, while injection site reactions, fever and diarrhoea were commonly observed with oritavancin therapy. Patients administered telavancin frequently reported nausea, taste disturbance and insomnia. To date, no drug-drug interactions have been identified for dalbavancin, oritavancin or telavancin. All three of these agents are promising alternatives for the treatment of cSSSIs in cases where more economical options such as vancomycin have been ineffective, in cases of reduced vancomycin susceptibility or resistance, or where vancomycin use has been associated with adverse events.


Assuntos
Aminoglicosídeos/uso terapêutico , Antibacterianos/uso terapêutico , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Glicopeptídeos/uso terapêutico , Infecções por Bactérias Gram-Positivas/tratamento farmacológico , Teicoplanina/análogos & derivados , Aminoglicosídeos/efeitos adversos , Aminoglicosídeos/química , Aminoglicosídeos/farmacocinética , Animais , Antibacterianos/efeitos adversos , Antibacterianos/química , Antibacterianos/farmacocinética , Ensaios Clínicos como Assunto , Interações Medicamentosas , Glicopeptídeos/efeitos adversos , Glicopeptídeos/química , Glicopeptídeos/farmacocinética , Infecções por Bactérias Gram-Positivas/microbiologia , Humanos , Lipoglicopeptídeos , Testes de Sensibilidade Microbiana , Estrutura Molecular , Teicoplanina/efeitos adversos , Teicoplanina/química , Teicoplanina/farmacocinética , Teicoplanina/uso terapêutico , Resultado do Tratamento
8.
Antimicrob Agents Chemother ; 49(4): 1323-5, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15793105

RESUMO

Antibiotic combinations are used to enhance antibacterial efficacy and to prevent the development of resistance. We have tested a possible synergistic effect of several antibacterial combinations on Bacillus anthracis. The in vitro activities of antibiotic combinations against two strains of B. anthracis, strain Sterne and the Russian anthrax vaccine strain STi, were tested by the fractional inhibitory concentration (FIC) method, derived from the MICs of the agents in combination, and by measuring the rate of bacterial killing over time by several antibiotic combinations. The FIC results showed that synergism against both B. anthracis strains was observed only with the combination of rifampin and clindamycin. The telithromycin-amoxicillin combination showed synergism against strain Sterne only. All other combinations were either indifferent or antagonistic. The results of the bacterial time-kill study demonstrated indifferent effects for all combinations. These in vitro results demonstrate the difficulties in obtaining synergistic combinations of antibiotics against B. anthracis.


Assuntos
Antibacterianos/farmacologia , Bacillus anthracis/efeitos dos fármacos , Bacillus anthracis/crescimento & desenvolvimento , Sinergismo Farmacológico , Bacillus anthracis/classificação , Clindamicina/farmacologia , Antagonismo de Drogas , Testes de Sensibilidade Microbiana/métodos , Rifampina/farmacologia
9.
J Antimicrob Chemother ; 51(3): 665-9, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12615869

RESUMO

OBJECTIVES: Pleural empyema is a serious complication of pneumonia, the optimal therapy of which is still unknown. The objective of this study was to evaluate the use of moxifloxacin in this condition. METHODS: Pleural empyema was induced in rabbits by intrapleural administration of Pasteurella multocida (10(5-6) cfu) or turpentine (0.3 mL) followed 3 h later by instillation of Streptococcus pneumoniae (ATCC 49619) (10(6) cfu) into the pleural cavity. The MICs of moxifloxacin for S. pneumoniae and P. multocida were 0.4 and 0.05 mg/L, respectively. Starting 30 h following S. pneumoniae challenge intramuscular moxifloxacin 12.5 and 25 mg/kg was administered x 4 (every 12 h). Pleural empyema fluid samples were obtained for bacterial count at 12 h intervals following the first three moxifloxacin administrations. Moxifloxacin levels in pleural empyema and serum samples were obtained at 0, 30, 60, 120, 240, 360 and 480 min and 12 h after the 4th dose and determined by bioassay. RESULTS: In control animals, S. pneumoniae (and P. multocida) persisted in the pleural empyema. S. pneumoniae also persisted in the pleural empyema fluid when moxifloxacin was administered at 12.5 mg/kg (x4 administrations). Mean serum and pleural empyema peak moxifloxacin levels (following the 25 mg/kg dose) were 7.6 (+/-3.2) and 4.8 (+/-2.5) mg/L, respectively. Pleural empyema peak moxifloxacin concentration lagged 1 h after serum moxifloxacin. Serum and pleural empyema half-lives were approximately 1.5 and approximately 6 h, respectively. Serum AUC(1-12) was 29.4 (+/-6.8) mg.h/L and serum area under the inhibitory concentration curve (AUIC) was 73.5 mg.h/L. Pleural empyema AUC(1-12) was 34.3 (+/-11.7) mg/L and pleural empyema AUIC was 85.8 mg.h/L. S. pneumoniae was eradicated from pleural empyema following a single dose of moxifloxacin 25 mg/kg in 52% of the animals and in 96% following four doses. Moxifloxacin was also effective in eradication of P. multocida. The rate of pleural empyema sterilization was related to moxifloxacin serum AUIC (r = 0.82) as well as serum peak moxifloxacin level (r = 0.84), but not to pleural empyema AUIC (r = 0.19) or pleural empyema peak levels. The results were similar for both methods of induction of pleural empyema. CONCLUSIONS: Moxifloxacin appears to penetrate well into experimental pleural empyema and effectively sterilize it from S. pneumoniae. Sterilization of S. pneumoniae is related to serum AUIC rather than to moxifloxacin pharmacokinetics in pleural empyema.


Assuntos
Anti-Infecciosos/uso terapêutico , Compostos Aza , Modelos Animais de Doenças , Empiema Pleural/tratamento farmacológico , Fluoroquinolonas , Infecções Pneumocócicas/tratamento farmacológico , Quinolinas , Animais , Anti-Infecciosos/metabolismo , Empiema Pleural/metabolismo , Masculino , Moxifloxacina , Pleura/efeitos dos fármacos , Infecções Pneumocócicas/metabolismo , Coelhos , Terebintina/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA