Antimicrobial activity of moxifloxacin, gatifloxacin and six fluoroquinolones against Streptococcus pneumoniae. (73/758)

The in vitro and pharmacodynamic effects of moxifloxacin and gatifloxacin against Streptococcus pneumoniae were compared with six other fluoroquinolones. Organisms included penicillin-susceptible (54) and penicillin-non-susceptible (145) isolates from 1998-1999. Moxifloxacin and clinafloxacin demonstrated the greatest in vitro activity, with MIC90s of 0.13 mg/L, followed by trovafloxacin, grepafloxacin > gatifloxacin, sparfloxacin > levofloxacin > ciprofloxacin. There was no difference in fluoroquinolone activity between penicillin-susceptible and -non-susceptible strains. Pharmacodynamic analysis using published pharmacokinetic information indicates that all the agents tested except ciprofloxacin had an AUC/MIC90 > 30, with moxifloxacin having the greatest free-drug availability.  (+info)

A comparison of the bactericidal activity of quinolone antibiotics in a Mycobacterium fortuitum model. (74/758)

New agents are urgently needed to meet the threat of multiple drug-resistant tuberculosis and to manage infection with the naturally resistant non-tuberculosis mycobacteria. Earlier fluoroquinolones have been shown to have promising in-vitro activity, although mouse infection and clinical studies suggested that they lack sufficient bactericidal activity. Methods were evaluated to measure the bactericidal activity of fluoroquinolones and to compare the new agent moxifloxacin with other fluoroquinolones with M. fortuitum as a model system. The optimum bactericidal concentrations (OBC) for the fluoroquinolones were: moxifloxacin, 0.5 mg/L; ciprofloxacin and sparfloxacin, 2 mg/L and ofloxacin, 8 mg/L. The bactericidal indices (BI) for moxifloxacin, ciprofloxacin, sparfloxacin and ofloxacin were 1.8, 0.5, 0.2 and 0.2, respectively. Similar ranking was obtained when the time taken to produce one log10 reduction in viable count was calculated. These data indicate that moxifloxacin was the most bactericidal of the fluoroquinolones tested. Such methods provide a simple in-vitro measure that correlates with in-vivo models.  (+info)

Microbial drug resistance and the roles of the new antibiotics. (75/758)

Physicians should be cautious in prescribing broad-spectrum antibiotics, particularly vancomycin and the fluoroquinolones, because widespread use of these drugs is promoting antibiotic resistance. Resistance is now found in many organisms, including staphylococci, enterococci, streptococci, pneumococci, and Pseudomonas aeruginosa. Some resistant strains can be treated with alternative narrower-spectrum antibiotics. In addition, five newly licensed antibiotics are available, but they should be used judiciously because of their side effects, high cost, and ability to promote additional resistance.  (+info)

gyrA mutations associated with quinolone resistance in Bacteroides fragilis group strains. (76/758)

Mutations in the gyrA gene contribute considerably to quinolone resistance in Escherichia coli. Mechanisms for quinolone resistance in anaerobic bacteria are less well studied. The Bacteroides fragilis group are the anaerobic organisms most frequently isolated from patients with bacteremia and intraabdominal infections. Forty-four clinafloxacin-resistant and-susceptible fecal and clinical isolates of the B. fragilis group (eight Bacteroides fragilis, three Bacteroides ovatus, five Bacteroides thetaiotaomicron, six Bacteroides uniformis, and 22 Bacteroides vulgatus) and six ATCC strains of the B. fragilis group were analyzed as follows: (i) determination of susceptibility to ciprofloxacin, levofloxacin, moxifloxacin, and clinafloxacin by the agar dilution method and (ii) sequencing of the gyrA quinolone resistance-determining region (QRDR) located between amino acid residues equivalent to Ala-67 through Gln-106 in E. coli. Amino acid substitutions were found at hotspots at positions 82 (n = 15) and 86 (n = 8). Strains with Ser82Leu substitutions (n = 13) were highly resistant to all quinolones tested. Mutations in other positions of gyrA were also frequently found in quinolone-resistant and -susceptible isolates. Eight clinical strains that lacked mutations in their QRDR were susceptible to at least two of the quinolones tested. Although newer quinolones have good antimicrobial activity against the B. fragilis group, quinolone resistance in B. fragilis strains can be readily selected in vivo. Mutational events in the QRDR of gyrA seem to contribute to quinolone resistance in Bacteroides species.  (+info)

Resistance to moxifloxacin in toxigenic Clostridium difficile isolates is associated with mutations in gyrA. (77/758)

Clostridium difficile is the etiological agent of antibiotic-associated colitis and the most common cause of hospital-acquired infectious diarrhea. Fluoroquinolones such as ciprofloxacin are associated with lower risks of C. difficile-associated diarrhea. In this study, we have analyzed 72 C. difficile isolates obtained from patients with different clinical courses of disease, such as toxic megacolon and relapses; the hospital environment; public places; and horses. They were investigated for their susceptibilities to moxifloxacin (MXF), metronidazole (MEO), and vancomycin (VAN). Mutants highly resistant to fluoroquinolones were selected in vitro by stepwise exposure to increasing concentrations of MXF. The resulting mutants were analyzed for the presence of mutations in the quinolone resistance-determining regions of DNA gyrase (gyrA), the production of toxins A and B, and the epidemiological relationship of these isolates. These factors were also investigated using PCR-based methods. All strains tested were susceptible to MEO and VAN. Twenty-six percent of the clinical isolates (19 of 72) were highly resistant to MXF (MIC > or = 16 microg/ml). Fourteen of these 19 strains contained nucleotide changes resulting in amino acid substitutions at position 83 in the gyrA protein. Resistant strains selected in vitro did not contain mutations at that position. These findings indicate that resistance to MXF in a majority of cases may be due to amino acid substitution in the gyrA gene.  (+info)

Induction of Tmolt4 leukemia cell death by 3,3-disubstituted-6,6-pentamethylene-1,5-diazabicyclo[3.1.0]hexane-2-4-diones: specificity for type II inosine 5'-monophasphate dehydrogenase. (78/758)

Inosine 5'-monophosphate dehydrogenase (IMPDH), the rate-limiting enzyme in the de novo pathway for synthesis of guanine nucleotides, is essential for normal cell proliferation and function. New derivatives of the 1,5-diazabicyclo[3.1.0]hexane-2,4-diones were synthesized and examined for antiproliferative effects and selective inhibition of human IMPDH type II activity. The 3,3-disubstituted-6,6-pentamethylene-1,5-diazabicyclo[3.1.0]hexane-2,4-diones proved to be effective antiproliferative agents in tumor cell lines derived from murine and human leukemias, lymphomas, uterine carcinoma, glioma, and breast effusion with ED50 values (concentration of compound that inhibits 50% of cell growth) ranging from 3.3 to 16 microM. The agents acted as antimetabolites suppressing de novo purine biosynthesis at the key regulatory enzyme IMPDH, resulting in the specific suppression of dGTP pool levels by 19 to 64% and DNA synthesis by 39 to 68%. The derivatives were specific inhibitors of IMPDH type II activity as opposed to type I, acting in a competitive manner with respect to inosine 5'-monophosphate, K(i) values of 44.2 to 62 microM. In addition, effects of agents on Tmolt4 cell growth and DNA synthesis could be reversed by coincubation with guanosine. Unlike mycophenolic acid and tiazofurin, the 6,6-pentamethylene-1,5-diazabicyclo[3.1.0]hexane-2,4-diones specifically targeted type II IMPDH, where activity is increased in replicating or neoplastic cells, and did not suppress type I activity, where expression is relatively unaffected by cell proliferation or transformation. Agents were not inhibitors of normal human lung fibroblast cell growth, WI-38, most likely due to the observed isoform selectivity.  (+info)

Susceptibilities of Mycoplasma hominis, M. pneumoniae, and Ureaplasma urealyticum to GAR-936, dalfopristin, dirithromycin, evernimicin, gatifloxacin, linezolid, moxifloxacin, quinupristin-dalfopristin, and telithromycin compared to their susceptibilities to reference macrolides, tetracyclines, and quinolones. (79/758)

The susceptibilities of Mycoplasma hominis, Mycoplasma pneumoniae, and Ureaplasma urealyticum to eight new antimicrobial agents were determined by agar dilution. M. pneumoniae was susceptible to the new glycylcycline GAR-936 at 0.12 microg/ml and evernimicin at 4 microg/ml, but it was resistant to linezolid. It was most susceptible to dirithromycin, quinupristin-dalfopristin, telithromycin, reference macrolides, and josamycin. M. hominis was susceptible to linezolid, evernimicin, and GAR-936. It was resistant to macrolides and the ketolide telithromycin but susceptible to quinupristin-dalfopristin and josamycin. U. urealyticum was susceptible to evernimicin (8 to 16 microg/ml) and resistant to linezolid. It was less susceptible to GAR-936 (4.0 microg/ml) than to tetracycline (0.5 microg/ml). Telithromycin and quinupristin-dalfopristin were the most active agents against ureaplasmas (0.06 microg/ml). The new quinolone gatifloxacin was active against M. pneumoniae and M. hominis at 0.12 to 0.25 microg/ml and active against ureaplasmas at 1.0 microg/ml. The MICs of macrolides were markedly affected by pH, with an 8- to 32-fold increase in the susceptibility of M. pneumoniae as the pH increased from 6.9 to 7.8. A similar increase in susceptibility with increasing pH was also observed with ureaplasmas. Tetracyclines showed a fourfold increase of activity as the pH decreased 1 U, whereas GAR-936 showed a fourfold decrease in activity with a decrease in pH.  (+info)

Single- and multi-step resistance selection study of gemifloxacin compared with trovafloxacin, ciprofloxacin, gatifloxacin and moxifloxacin in Streptococcus pneumoniae. (80/758)

The ability of sequential subcultures in subinhibitory concentrations of gemifloxacin, trovafloxacin, ciprofloxacin, gatifloxacin and moxifloxacin to select resistant mutants was studied in 16 pneumococci [eight with ciprofloxacin MICs (mg/L) 0.25-1; four with 8-16; four with 16-32]. Subculturing was done 50 times, or until mutants with elevated MICs (> or = 4 x) to the selecting drug emerged. Subculturing in gemifloxacin selected six resistant mutants (gemifloxacin MICs 2 mg/L); trovafloxacin selected nine (trovafloxacin MICs 2-4 mg/L); ciprofloxacin selected 11 (ciprofloxacin MICs 8-128 mg/L); gatifloxacin selected 13; and moxifloxacin selected 12 (gatifloxacin or moxifloxacin MICs 2-16 mg/L). DNA sequencing showed that most mutants had mutations in ParC at Ser-79 or Asp-83 and in GyrA at Ser-81 or Glu-85; some mutants also had mutations in ParE or GyrB. Some new mutations were found in ParE or GyrB that have not yet been reported; GyrB mutation might be associated with moxifloxacin resistance. Both DNA gyrase and topoisomerase IV were thought to be the target of gemifloxacin; gemifloxacin also selected mutants with single modifications in gyrA, parC or parE alone among derived mutants by repeated exposure to subinhibitory concentrations of fluoroquinolones. In the presence of reserpine, most mutants had lower MICs of ciprofloxacin and gemifloxacin (4-32 x), and gatifloxacin (4-8 x), suggesting an efflux mechanism; none had lower trovafloxacin and moxifloxacin MICs. All quinolones tested selected for resistance; judicious use and proper dosing will be necessary to avoid resistance selection of newer broad-spectrum fluoroquinolones.  (+info)