Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Antimicrob Agents Chemother ; 54(1): 411-7, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19884374

ABSTRACT

Nemonoxacin (TG-873870) is a novel nonfluorinated quinolone with broad-spectrum activities against Gram-positive and Gram-negative aerobic, anaerobic, and atypical pathogens, as well as against methicillin-resistant Staphylococcus aureus, vancomycin-resistant S. aureus, and multiple-resistant bacterial pathogens. We conducted a randomized, double-blind, placebo-controlled, dose-escalating study to ascertain the safety, tolerability, and pharmacokinetics of nemonoxacin. We enrolled 46 healthy volunteers and used a once-daily oral-dosing range of 75 to 1,000 mg for 10 days. Additionally, the food effect was evaluated in subjects in the 500-mg cohort. Nemonoxacin was generally safe and well tolerated, with no significant changes in the clinical laboratory tests or electrocardiograms. Adverse effects, including headache, contact dermatitis, and rash, were mild and resolved spontaneously. Nemonoxacin was rapidly absorbed within 2 h postdosing, and generally, a steady state was reached after 3 days. The maximum plasma concentration and the area under the plasma concentration-time curve were dose proportional over the dosing range. The elimination half-life was approximately 7.5 h and 19.7 h on days 1 and 10, respectively. Approximately 37 to 58% of the drug was excreted in the urine. Food affected the pharmacokinetics, with decreases in the maximum plasma concentration and area under the plasma concentration-time curve of 46% and 27%, respectively. However, the free AUC/MIC(90) of nemonoxacin was more than 100 under both the fasting and fed conditions, predicting the efficacy of nemonoxacin against most of the tested pathogens. In conclusion, the results support further clinical investigation of once-daily nemonoxacin administration for antibiotic-sensitive and antibiotic-resistant bacterial infections.


Subject(s)
Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/pharmacokinetics , Quinolones/adverse effects , Quinolones/pharmacokinetics , Adolescent , Adult , Anti-Bacterial Agents/administration & dosage , Dermatitis, Contact/etiology , Dose-Response Relationship, Drug , Double-Blind Method , Electrocardiography/drug effects , Enzymes/blood , Female , Headache/chemically induced , Humans , Male , Quinolones/administration & dosage , Young Adult
2.
Antimicrob Agents Chemother ; 54(10): 4098-106, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20660689

ABSTRACT

Nemonoxacin, a novel nonfluorinated quinolone, exhibits potent in vitro and in vivo activities against community-acquired pneumonia (CAP) pathogens, including multidrug-resistant Streptococcus pneumoniae. Patients with mild to moderate CAP (n = 265) were randomized to receive oral nemonoxacin (750 mg or 500 mg) or levofloxacin (500 mg) once daily for 7 days. Clinical responses were determined at the test-of-cure visit in intent-to-treat (ITT), clinical per protocol (PPc), evaluable-ITT, and evaluable-PPc populations. The clinical cure rates for 750 mg nemonoxacin, 500 mg nemonoxacin, and levofloxacin were 89.9%, 87.0%, and 91.1%, respectively, in the evaluable-ITT population; 91.7%, 87.7%, and 90.3%, respectively, in the evaluable-PPc population; 82.6%, 75.3%, and 80.0%, respectively, in the ITT population; and 83.5%, 78.0%, and 82.3%, respectively, in the PPc population. Noninferiority to levofloxacin was demonstrated in both the 750-mg and 500-mg nemonoxacin groups for the evaluable-ITT and evaluable-PPc populations, and also in the 750 mg nemonoxacin group for the ITT and PPc populations. Overall bacteriological success rates were high for all treatment groups in the evaluable-bacteriological ITT population (90.2% in the 750 mg nemonoxacin group, 84.8% in the 500 mg nemonoxacin group, and 92.0% in the levofloxacin group). All three treatments were well tolerated, and no drug-related serious adverse events were observed. Overall, oral nemonoxacin (both 750 mg and 500 mg) administered for 7 days resulted in high clinical and bacteriological success rates in CAP patients. Further, good tolerability and excellent activity against common causative pathogens were demonstrated. Nemonoxacin (750 mg and 500 mg) once daily is as effective and safe as levofloxacin (500 mg) once daily for the treatment of CAP.


Subject(s)
Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/therapeutic use , Levofloxacin , Ofloxacin/adverse effects , Ofloxacin/therapeutic use , Pneumonia/drug therapy , Quinolones/adverse effects , Quinolones/therapeutic use , Adult , Double-Blind Method , Female , Humans , Male , Middle Aged , Treatment Outcome
3.
Microb Drug Resist ; 26(2): 110-117, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31478786

ABSTRACT

Nemonoxacin is a nonfluorinated quinolone with good bactericidal effects against quinolone-resistant Gram-positive microorganisms. The in vitro inducible resistance of nemonoxacin against clinically relevant Gram-positive pathogens was compared with ciprofloxacin, levofloxacin, and moxifloxacin. Three strains of each bacterial species, including Streptococcus pneumoniae, Staphylococcus aureus, Enterococcus faecium, and Enterococcus faecalis, were cultured. All clinical isolates had wild-type gyrA, gyrB and parC, parE before further in vitro test. DNA sequencing for the quinolone resistance determination region (QRDR) of gyrase and topoisomerase genes was performed. Nemonoxacin had the lowest minimum inhibitory concentrations (MICs) among all quinolones. During exposure to nemonoxacin, the MIC values did not increase for S. aureus, E. faecium, and E. faecalis, and revealed fourfold increase of S. pneumoniae over three cycles of a stepwise resistance selection. DNA sequencing did not show inducible QRDR resistance of nemonoxacin group. Compared to other fluoroquinolones, nemonoxacin has a low potential for inducing resistant pathogens.


Subject(s)
Anti-Bacterial Agents/pharmacology , Fluoroquinolones/pharmacology , Gram-Positive Bacteria/drug effects , Quinolones/pharmacology , DNA Gyrase/genetics , DNA Topoisomerases/genetics , Dose-Response Relationship, Drug , Drug Resistance, Multiple, Bacterial , Gram-Positive Bacteria/genetics , Humans , Microbial Sensitivity Tests , Sequence Analysis, DNA , Taiwan
4.
J Microbiol Immunol Infect ; 52(5): 743-751, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30616912

ABSTRACT

BACKGROUND: Nemonoxacin, a novel nonfluorinated quinolone, has broad-spectrum antibacterial activity, including activity against antibiotic-resistant strains, and was developed for treating community-acquired pneumonia (CAP). This report provides an integrated safety summary of oral nemonoxacin from two phase II and one phase III clinical studies. METHODS: Patients with mild CAP were randomized for treatment with nemonoxacin 500 mg (NEMO-500MG), nemonoxacin 750 mg (NEMO-750MG), or levofloxacin 500 mg (LEVO), orally, once daily, for 7-10 days. Hematological, gastrointestinal, and hepatic disorders; electrocardiography abnormalities; and reported quinolone-associated clinical concerns were included in this analysis. RESULTS: A total of 520, 155, and 320 subjects were assigned to receive NEMO-500MG, NEMO-750MG, and LEVO, respectively. The incidence of adverse events (AEs) was the highest (54.8%) in the NEMO-750MG group (NEMO-500MG, 36.9%; NEMO-750MG, 54.8%; LEVO, 39.7%) and that of drug-related AEs was comparable between the three groups (NEMO-500MG, 22.9%; NEMO-750MG, 31.0%; LEVO, 22.5%). The majority (>80%) of the patients showed mild drug-related AEs and the distribution based on severity was similar between the groups. The most commonly reported drug-related AEs included neutropenia (NEMO-500MG, 2.5%; NEMO-750MG, 8.4%; LEVO, 4.4%), nausea (NEMO-500MG, 2.5%; NEMO-750MG, 7.1%; LEVO, 2.5%), leukopenia (NEMO-500MG, 2.3%; NEMO-750MG, 4.5%; LEVO, 3.1%), and increased alanine aminotransferase level (NEMO-500MG, 4.4%; NEMO-750MG, 0%; LEVO, 2.5%). CONCLUSION: Nemonoxacin was well tolerated and no clinically significant safety concerns were identified, suggesting that it possesses a desirable safety and tolerability profile similar to that of levofloxacin, and may be a suitable alternative to fluoroquinolones for treating patients with CAP.


Subject(s)
Community-Acquired Infections/drug therapy , Levofloxacin/therapeutic use , Pneumonia/drug therapy , Quinolones/therapeutic use , Safety , Administration, Oral , Adolescent , Adult , Aged , Aged, 80 and over , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/therapeutic use , China , Double-Blind Method , Drug Combinations , Female , Fluoroquinolones/therapeutic use , Humans , Levofloxacin/administration & dosage , Lung Diseases/drug therapy , Male , Middle Aged , Quinolones/administration & dosage , South Africa , Taiwan , Treatment Outcome , Young Adult
5.
Transplantation ; 102(12): 2002-2011, 2018 12.
Article in English | MEDLINE | ID: mdl-30095739

ABSTRACT

BACKGROUND: CXC motif chemokine receptor 4 (CXCR4) blockade is pursued as an alternative to mesenchymal stem cell treatment in transplantation based on our previous report that burixafor, through CXCR4 antagonism, mobilizes immunomodulatory mesenchymal stem cells. Here, we explored the efficacy of combining mycophenolate mofetil (MMF)-based immunosuppressants with repetitive burixafor administration. METHODS: Swine heterotopic cardiac allograft recipients received MMF and corticosteroids (control, n = 10) combined with burixafor as a 2-dose (burixafor2D, n = 7) or 2-dose plus booster injections (burixafor2D + B, n = 5) regimen. The efficacy endpoints were graft survival, freedom from first acute rejection, and the severity of intimal hyperplasia. Each specimen was sacrificed either at its first graft arrest or after 150 days. RESULTS: After 150 days, all specimens in the control group had died, but 28.5% of the burixafor2D group survived, and 60% of the burixafor2D + B group survived (P = 0.0088). Although the control group demonstrated acute rejection at a median of 33.5 days, the burixafor2D + B group survived without acute rejection for a median of 136 days (P = 0.0209). Burixafor administration significantly attenuated the incidence rate of acute rejection (P = 0.002) and the severity of intimal hyperplasia (P = 0.0097) at end point relative to the controls. These findings were associated with reduced cell infiltrates in the allografts, and modulation of C-reactive protein profiles in the circulation. CONCLUSIONS: The augmentation of conventional MMF plus corticosteroids with a CXCR4 antagonist is potentially effective in improving outcomes after heart transplantation in minipigs. Future studies are warranted into optimizing the therapeutic regimens for humans.


Subject(s)
Coronary Artery Disease/prevention & control , Graft Rejection/prevention & control , Graft Survival/drug effects , Heart Transplantation/adverse effects , Immunosuppressive Agents/pharmacology , Mycophenolic Acid/pharmacology , Receptors, CXCR4/antagonists & inhibitors , Acute Disease , Allografts , Animals , Coronary Artery Disease/immunology , Coronary Artery Disease/metabolism , Disease Models, Animal , Drug Therapy, Combination , Female , Graft Rejection/immunology , Graft Rejection/metabolism , Male , Receptors, CXCR4/immunology , Receptors, CXCR4/metabolism , Swine , Swine, Miniature , Time Factors
6.
J Med Chem ; 49(16): 4971-80, 2006 Aug 10.
Article in English | MEDLINE | ID: mdl-16884309

ABSTRACT

A potent SARS coronavirus (CoV) 3CL protease inhibitor (TG-0205221, Ki = 53 nM) has been developed. TG-0205221 showed remarkable activity against SARS CoV and human coronavirus (HCoV) 229E replications by reducing the viral titer by 4.7 log (at 5 microM) for SARS CoV and 5.2 log (at 1.25 microM) for HCoV 229E. The crystal structure of TG-0205221 (resolution = 1.93 A) has revealed a unique binding mode comprising a covalent bond, hydrogen bonds, and numerous hydrophobic interactions. Structural comparisons between TG-0205221 and a natural peptide substrate were also discussed. This information may be applied toward the design of other 3CL protease inhibitors.


Subject(s)
Antiviral Agents/chemical synthesis , Carbamates/chemical synthesis , Cysteine Endopeptidases/chemistry , Dipeptides/chemical synthesis , Viral Proteins/antagonists & inhibitors , Viral Proteins/chemistry , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Carbamates/chemistry , Carbamates/pharmacology , Cell Line , Chlorocebus aethiops , Coronavirus 229E, Human/drug effects , Coronavirus 3C Proteases , Crystallography, X-Ray , Dipeptides/chemistry , Dipeptides/pharmacology , Drug Stability , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Mice , Models, Molecular , Molecular Structure , Rats , Severe acute respiratory syndrome-related coronavirus/drug effects , Structure-Activity Relationship , Virus Replication/drug effects
7.
Microb Drug Resist ; 22(7): 578-584, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27267788

ABSTRACT

Selection of resistant strains in Streptococcus pneumoniae was studied in vitro with nemonoxacin, a novel nonfluorinated quinolone (NFQ), in comparison with quinolone benchmarks, ciprofloxacin, garenoxacin, and gatifloxacin. In stepwise resistance selection studies, a 256-fold loss of potency was observed after three to four steps of exposure to ciprofloxacin or garenoxacin. In contrast, the loss of potency was limited to eightfold after three steps of exposure to nemonoxacin and repeated attempts to isolate highly resistant organisms after four steps of exposure yielded isolates that could not be subcultured in liquid medium. The quinolone resistance-determining regions of the target genes, parC, parE, gyrA, and gyrB, were analyzed through DNA sequencing. Known mutations, especially in the hotspots of parC and gyrA, were selected with exposure to garenoxacin, ciprofloxacin, and gatifloxacin. In contrast, mutations selected with nemonoxacin were limited to GyrA, GyrB, and ParE, sparing ParC, which is known as a key driver of resistance in clinical isolates of S. pneumoniae. This observation is consistent with previous data using other NFQs, which showed no loss of potency due to ParC mutations in clinical isolates. This apparently unique feature of nemonoxacin is potentially attributable to the structural uniqueness of the NFQs, distinguishing them from the fluoroquinolones that are commonly prescribed for infections by S. pneumoniae.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/drug effects , Mutation , Quinolones/pharmacology , Streptococcus pneumoniae/drug effects , Ciprofloxacin/pharmacology , Culture Media/chemistry , DNA Gyrase/genetics , DNA Topoisomerase IV/genetics , Drug Resistance, Bacterial/genetics , Fluoroquinolones/pharmacology , Gatifloxacin , Gene Expression , Microbial Sensitivity Tests , Sequence Analysis, DNA , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/growth & development , Structure-Activity Relationship
8.
Cell Transplant ; 24(7): 1313-28, 2015.
Article in English | MEDLINE | ID: mdl-24823505

ABSTRACT

Interaction between chemokine stromal cell-derived factor 1 and the CXC chemokine receptor 4 (CXCR4) governs the sequestration and mobilization of bone marrow stem cells. We investigated the therapeutic potential of TG-0054, a novel CXCR4 antagonist, in attenuating cardiac dysfunction after myocardial infarction (MI). In miniature pigs (minipigs), TG-0054 mobilized CD34(+)CXCR4(+), CD133(+)CXCR4(+), and CD271(+)CXCR4(+) cells into peripheral circulation. After isolation and expansion, TG-0054-mobilized CD271(+) cells were proved to be mesenchymal stem cells (designated CD271-MSCs) since they had trilineage differentiation potential, surface markers of MSCs, and immunosuppressive effects on allogeneic lymphocyte proliferation. MI was induced in 22 minipigs using balloon occlusion of the left anterior descending coronary artery, followed by intravenous injections of 2.85 mg/kg of TG-0054 or saline at 3 days and 7 days post-MI. Serial MRI analyses revealed that TG-0054 treatment prevented left ventricular (LV) dysfunction at 12 weeks after MI (change of LV ejection fraction from baseline, -1.0 ± 6.2% in the TG-0054 group versus -7.9 ± 5.8% in the controls). The preserved cardiac function was accompanied by a significant decrease in the myocardial expression of TNF-α, IL-1ß, and IL-6 at 7 days post-MI. Moreover, the plasma levels of TNF-α, IL-1ß, and IL-6 were persistently suppressed by the TG-0054 treatment. Infusion of TG-0054-mobilized CD271-MSCs reduced both myocardial and plasma cytokine levels in a pattern, which is temporally correlated with TG-0054 treatment. This study demonstrated that TG-0054 improves the impaired LV contractility following MI, at least in part, by mobilizing MSCs to attenuate the postinfarction inflammation. This insight may facilitate exploring novel stem cell-based therapy for treating post-MI heart failure.


Subject(s)
Mesenchymal Stem Cells/metabolism , Myocardial Infarction/therapy , Receptors, CXCR4/antagonists & inhibitors , Ventricular Function, Left/physiology , Animals , Disease Models, Animal , Immunomodulation , Inflammation/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Porcine Reproductive and Respiratory Syndrome , Receptors, CXCR4/metabolism , Swine
9.
J Med Chem ; 45(8): 1644-55, 2002 Apr 11.
Article in English | MEDLINE | ID: mdl-11931618

ABSTRACT

When skeletons of Win compounds were used as templates, computer-assisted drug design led to the identification of a novel series of imidazolidinone derivatives with significant antiviral activity against enterovirus 71 (EV 71), the infection of which had resulted in about 80 fatalities during the 1998 epidemic outbreak in Taiwan. In addition to inhibiting all the genotypes (A, B, and C) of EV 71 in the submicromolar to low micromolar range, compounds 1 and 8 were extensively evaluated against a variety of viruses, showing potent activity against coxsackievirus A9 (IC(50) = 0.47-0.55 microM) and coxsackievirus A24 (IC(50) = 0.47-0.55 microM) as well as moderate activity against enterovirus 68 (IC(50) = 2.13 microM) and echovirus 9 (IC(50) = 2.6 microM). Our SAR studies revealed that imidazolidinone analogues with an aryl substituent at the para position of the phenoxyl ring, such as compounds 20, 21, 27, 57, 58, and 61, in general exhibited the highest activity against EV 71. Among them, compound 20 and its corresponding hydrochloride salt 57, in terms of potency and selectivity index, appear to be the most promising candidates in this series for further development of anti-EV-71 agents. Preliminary results of the study on the mode of action by a time-course experiment suggest that test compounds 1 and 8 can effectively inhibit the virus replication at the early stages, referring to virus attachment or uncoating. This indicates that the surface protein may be the target for this type of compounds.


Subject(s)
Antiviral Agents/chemical synthesis , Enterovirus/drug effects , Imidazoles/chemical synthesis , Pyridines/chemical synthesis , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cell Line , Chlorocebus aethiops , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Neutralization Tests , Oxadiazoles/chemistry , Oxazoles , Pyridines/chemistry , Pyridines/pharmacology , Structure-Activity Relationship , Viral Plaque Assay
10.
J Biol Chem ; 284(12): 7646-55, 2009 Mar 20.
Article in English | MEDLINE | ID: mdl-19144641

ABSTRACT

Human coxsackievirus (CV) belongs to the picornavirus family, which consists of over 200 medically relevant viruses. In picornavirus, a chymotrypsin-like protease (3C(pro)) is required for viral replication by processing the polyproteins, and thus it is regarded as an antiviral drug target. A 3C-like protease (3CL(pro)) also exists in human coronaviruses (CoV) such as 229E and the one causing severe acute respiratory syndrome (SARS). To combat SARS, we previously had developed peptidomimetic and zinc-coordinating inhibitors of 3CL(pro). As shown in the present study, some of these compounds were also found to be active against 3C(pro) of CV strain B3 (CVB3). Several crystal structures of 3C(pro) from CVB3 and 3CL(pro) from CoV-229E and SARS-CoV in complex with the inhibitors were solved. The zinc-coordinating inhibitor is tetrahedrally coordinated to the His(40)-Cys(147) catalytic dyad of CVB3 3C(pro). The presence of specific binding pockets for the residues of peptidomimetic inhibitors explains the binding specificity. Our results provide a structural basis for inhibitor optimization and development of potential drugs for antiviral therapies.


Subject(s)
Biomimetic Materials/chemistry , Cysteine Endopeptidases/chemistry , Cysteine Proteinase Inhibitors/chemistry , Severe acute respiratory syndrome-related coronavirus/enzymology , Viral Proteins/chemistry , Zinc/chemistry , Binding Sites , Biomimetic Materials/therapeutic use , Coronavirus 3C Proteases , Cysteine Proteinase Inhibitors/therapeutic use , Humans , Protein Structure, Tertiary , Severe Acute Respiratory Syndrome/drug therapy , Severe Acute Respiratory Syndrome/enzymology , Structure-Activity Relationship , Viral Proteins/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL