Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
ACS Infect Dis ; 9(2): 342-364, 2023 02 10.
Article in English | MEDLINE | ID: mdl-36706233

ABSTRACT

SQ109 is a tuberculosis drug candidate that has high potency against Mycobacterium tuberculosis and is thought to function at least in part by blocking cell wall biosynthesis by inhibiting the MmpL3 transporter. It also has activity against bacteria and protozoan parasites that lack MmpL3, where it can act as an uncoupler, targeting lipid membranes and Ca2+ homeostasis. Here, we synthesized 18 analogs of SQ109 and tested them against M. smegmatis, M. tuberculosis, M. abscessus, Bacillus subtilis, and Escherichia coli, as well as against the protozoan parasites Trypanosoma brucei, T. cruzi, Leishmania donovani, L. mexicana, and Plasmodium falciparum. Activity against the mycobacteria was generally less than with SQ109 and was reduced by increasing the size of the alkyl adduct, but two analogs were ∼4-8-fold more active than SQ109 against M. abscessus, including a highly drug-resistant strain harboring an A309P mutation in MmpL3. There was also better activity than found with SQ109 with other bacteria and protozoa. Of particular interest, we found that the adamantyl C-2 ethyl, butyl, phenyl, and benzyl analogs had 4-10× increased activity against P. falciparum asexual blood stages, together with low toxicity to a human HepG2 cell line, making them of interest as new antimalarial drug leads. We also used surface plasmon resonance to investigate the binding of inhibitors to MmpL3 and differential scanning calorimetry to investigate binding to lipid membranes. There was no correlation between MmpL3 binding and M. tuberculosis or M. smegmatis cell activity, suggesting that MmpL3 is not a major target in mycobacteria. However, some of the more active species decreased lipid phase transition temperatures, indicating increased accumulation in membranes, which is expected to lead to enhanced uncoupler activity.


Subject(s)
Malaria , Mycobacterium abscessus , Mycobacterium tuberculosis , Parasites , Tuberculosis , Animals , Humans , Antitubercular Agents/pharmacology , Parasites/metabolism , Bacterial Proteins/metabolism , Tuberculosis/microbiology , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Lipids
2.
Microorganisms ; 10(2)2022 Feb 17.
Article in English | MEDLINE | ID: mdl-35208914

ABSTRACT

BACKGROUND: The turnaround times for phenotypic tests used to monitor the bacterial load of Mycobacterium tuberculosis, in both clinical and preclinical studies, are delayed by the organism's slow growth in culture media. The existence of differentially culturable populations of M.tuberculosis may result in an underestimate of the true number. Moreover, culture methods are susceptible to contamination resulting in loss of critical data points. OBJECTIVES: We report the adaptation of our robust, culture-free assay utilising 16S ribosomal RNA, developed for sputum, to enumerate the number of bacteria present in animal tissues as a tool to improve the read-outs in preclinical drug efficacy studies. METHODS: Initial assay adaptation was performed using naïve mouse lungs spiked with known quantities of M. tuberculosis and an internal RNA control. Tissues were homogenised, total RNA extracted, and enumeration performed using RT-qPCR. We then evaluated the utility of the assay, in comparison to bacterial counts estimated using growth assays on solid and liquid media, to accurately inform bacterial load in tissues from M. tuberculosis-infected mice before and during treatment with a panel of drug combinations. RESULTS: When tested on lung tissues derived from infected mice, the MBL assay produced comparable results to the bacterial counts in solid culture (colony forming units: CFU). Notably, under specific drug treatments, the MBL assay was able to detect a significantly higher number of M. tuberculosis compared to CFU, likely indicating the presence of bacteria that were unable to produce colonies in solid-based culture. Additionally, growth recovery in liquid media using the most probable number (MPN) assay was able to account for the discrepancy between the MBL assay and CFU number, suggesting that the MBL assay detects differentially culturable sub-populations of M. tuberculosis. CONCLUSIONS: The MBL assay can enumerate the bacterial load in animal tissues in real time without the need to wait for extended periods for cultures to grow. The readout correlates well with CFUs. Importantly, we have shown that the MBL is able to measure specific populations of bacteria not cultured on solid agar. The adaptation of this assay for preclinical studies has the potential to decrease the readout time of data acquisition from animal experiments and could represent a valuable tool for tuberculosis drug discovery and development.

4.
Front Microbiol ; 9: 2271, 2018.
Article in English | MEDLINE | ID: mdl-30319580

ABSTRACT

The current standard of care therapy for pulmonary Mycobacterium kansasii infection is isoniazid (300 mg/day), rifampin (600 mg/day), and ethambutol (15 mg/kg/day) for 12 months after achieving sputum culture negativity. Rifampin is the key drug in this regimen. The contribution of isoniazid is unclear since its in vitro MICs against M. kansasii are near the peak achievable serum levels and more than 100-fold greater than the MICs for Mycobacterium tuberculosis. Ethambutol likely decreases the emergence of rifampin resistant organisms. There are several new drug classes (e.g., quinolones, macrolides, nitroimidazoles, diarylquinolines, and clofazimine) that exhibit antimycobacterial activities against M. tuberculosis but have not yet been adequately studied against M. kansasii infections. The evaluation of in vitro activities of these agents as well as their study in new regimens in comparison to the standard of care regimen in mouse infection models should be undertaken. This knowledge will inform development of human clinical trials of new regimens in comparison to the current standard of care regimen. It is likely that shorter and more effective therapy is achievable with currently available drugs.

5.
Bioorg Med Chem Lett ; 27(17): 3987-3991, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28778468

ABSTRACT

To develop agents for the treatment of infections caused by Mycobacterium tuberculosis, a novel phenotypic screen was undertaken that identified a series of 2-N-aryl thiazole-based inhibitors of intracellular Mycobacterium tuberculosis. Analogs were optimized to improve potency against an attenuated BSL2 H37Ra laboratory strain cultivated in human macrophage cells in vitro. The insertion of a carboxylic acid functionality resulted in compounds that retained potency and greatly improved microsomal stability. However, the strong potency trends we observed in the attenuated H37Ra strain were inconsistent with the potency observed for virulent strains in vitro and in vivo.


Subject(s)
Anti-Bacterial Agents/pharmacology , Mycobacterium tuberculosis/drug effects , Thiazoles/pharmacology , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Dose-Response Relationship, Drug , Humans , Macrophages/drug effects , Macrophages/microbiology , Mice , Microbial Sensitivity Tests , Molecular Structure , Structure-Activity Relationship , Thiazoles/chemical synthesis , Thiazoles/chemistry
6.
Antimicrob Agents Chemother ; 59(3): 1455-65, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25534737

ABSTRACT

New drugs to treat drug-resistant tuberculosis are urgently needed. Extensively drug-resistant and probably the totally drug-resistant tuberculosis strains are resistant to fluoroquinolones like moxifloxacin, which target gyrase A, and most people infected with these strains die within a year. In this study, we found that a novel aminobenzimidazole, VXc-486, which targets gyrase B, potently inhibits multiple drug-sensitive isolates and drug-resistant isolates of Mycobacterium tuberculosis in vitro (MICs of 0.03 to 0.30 µg/ml and 0.08 to 5.48 µg/ml, respectively) and reduces mycobacterial burdens in lungs of infected mice in vivo. VXc-486 is active against drug-resistant isolates, has bactericidal activity, and kills intracellular and dormant M. tuberculosis bacteria in a low-oxygen environment. Furthermore, we found that VXc-486 inhibits the growth of multiple strains of Mycobacterium abscessus, Mycobacterium avium complex, and Mycobacterium kansasii (MICs of 0.1 to 2.0 µg/ml), as well as that of several strains of Nocardia spp. (MICs of 0.1 to 1.0 µg/ml). We made a direct comparison of the parent compound VXc-486 and a phosphate prodrug of VXc-486 and showed that the prodrug of VXc-486 had more potent killing of M. tuberculosis than did VXc-486 in vivo. In combination with other antimycobacterial drugs, the prodrug of VXc-486 sterilized M. tuberculosis infection when combined with rifapentine-pyrazinamide and bedaquiline-pyrazinamide in a relapse infection study in mice. Furthermore, the prodrug of VXc-486 appeared to perform at least as well as the gyrase A inhibitor moxifloxacin. These findings warrant further development of the prodrug of VXc-486 for the treatment of tuberculosis and nontuberculosis mycobacterial infections.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Benzimidazoles/therapeutic use , Mycobacterium Infections/drug therapy , Topoisomerase II Inhibitors/therapeutic use , Animals , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microbial Sensitivity Tests
7.
Antimicrob Agents Chemother ; 59(3): 1534-41, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25534740

ABSTRACT

Previous studies indicated that inhibition of efflux pumps augments tuberculosis therapy. In this study, we used timcodar (formerly VX-853) to determine if this efflux pump inhibitor could increase the potency of antituberculosis (anti-TB) drugs against Mycobacterium tuberculosis in in vitro and in vivo combination studies. When used alone, timcodar weakly inhibited M. tuberculosis growth in broth culture (MIC, 19 µg/ml); however, it demonstrated synergism in drug combination studies with rifampin, bedaquiline, and clofazimine but not with other anti-TB agents. When M. tuberculosis was cultured in host macrophage cells, timcodar had about a 10-fold increase (50% inhibitory concentration, 1.9 µg/ml) in the growth inhibition of M. tuberculosis and demonstrated synergy with rifampin, moxifloxacin, and bedaquiline. In a mouse model of tuberculosis lung infection, timcodar potentiated the efficacies of rifampin and isoniazid, conferring 1.0 and 0.4 log10 reductions in bacterial burden in lung, respectively, compared to the efficacy of each drug alone. Furthermore, timcodar reduced the likelihood of a relapse infection when evaluated in a mouse model of long-term, chronic infection with treatment with a combination of rifampin, isoniazid, and timcodar. Although timcodar had no effect on the pharmacokinetics of rifampin in plasma and lung, it did increase the plasma exposure of bedaquiline. These data suggest that the antimycobacterial drug-potentiating activity of timcodar is complex and drug dependent and involves both bacterial and host-targeted mechanisms. Further study of the improvement of the potency of antimycobacterial drugs and drug candidates when used in combination with timcodar is warranted.


Subject(s)
Antitubercular Agents/pharmacology , Pyridines/pharmacology , Animals , Antitubercular Agents/pharmacokinetics , Cell Line , Drug Synergism , Female , Humans , Macrophages/immunology , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Mycobacterium tuberculosis/drug effects
8.
Biochem Pharmacol ; 86(2): 222-30, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23680030

ABSTRACT

The genus Mycobacterium includes non-pathogenic species such as M. smegmatis, and pathogenic species such as M. tuberculosis, the causative agent of tuberculosis (TB). Treatment of TB requires a lengthy regimen of several antibiotics, whose effectiveness has been compromised by the emergence of resistant strains. New antibiotics that can shorten the treatment course and those that have not been compromised by bacterial resistance are needed. In this study, we report that thiadiazolidinones, a relatively little-studied heterocyclic class, inhibit the activity of mycobacterial alanine racemase, an essential enzyme that converts l-alanine to d-alanine for peptidoglycan synthesis. Twelve members of the thiadiazolidinone family were evaluated for inhibition of M. tuberculosis and M. smegmatis alanine racemase activity and bacterial growth. Thiadiazolidinones inhibited M. tuberculosis and M. smegmatis alanine racemases to different extents with 50% inhibitory concentrations (IC50) ranging from <0.03 to 28µM and 23 to >150µM, respectively. The compounds also inhibited the growth of these bacteria, including multidrug resistant strains of M. tuberculosis. The minimal inhibitory concentrations (MIC) for drug-susceptible M. tuberculosis and M. smegmatis ranged from 6.25µg/ml to 100µg/ml, and from 1.56 to 6.25µg/ml for drug-resistant M. tuberculosis. The in vitro activities of thiadiazolidinones suggest that this family of compounds might represent starting points for medicinal chemistry efforts aimed at developing novel antimycobacterial agents.


Subject(s)
Alanine Racemase/antagonists & inhibitors , Mycobacterium smegmatis/drug effects , Mycobacterium tuberculosis/drug effects , Thiadiazoles/pharmacology , Alanine Racemase/chemistry , Alanine Racemase/metabolism , Amino Acid Sequence , Catalysis , Molecular Sequence Data , Mycobacterium smegmatis/enzymology , Mycobacterium tuberculosis/enzymology , Sequence Homology, Amino Acid , Spectrometry, Mass, Electrospray Ionization , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
9.
Vaccines (Basel) ; 1(1): 34-57, 2013 Jan 11.
Article in English | MEDLINE | ID: mdl-26343849

ABSTRACT

Early attempts to improve BCG have focused on increasing the expression of prominent antigens and adding recombinant toxins or cytokines to influence antigen presentation. One such modified BCG vaccine candidate has been withdrawn from human clinical trials due to adverse effects. BCG was derived from virulent Mycobacterium bovis and retains much of its capacity for suppressing host immune responses. Accordingly, we have used a different strategy for improving BCG based on reducing its immune suppressive capacity. We made four modifications to BCG Tice to produce 4dBCG and compared it to the parent vaccine in C57Bl/6 mice. The modifications included elimination of the oxidative stress sigma factor SigH, elimination of the SecA2 secretion channel, and reductions in the activity of iron co-factored superoxide dismutase and glutamine synthetase. After IV inoculation of 4dBCG, 95% of vaccine bacilli were eradicated from the spleens of mice within 60 days whereas the titer of BCG Tice was not significantly reduced. Subcutaneous vaccination with 4dBCG produced greater protection than vaccination with BCG against dissemination of an aerosolized challenge of M. tuberculosis to the spleen at 8 weeks post-challenge. At this time, 4dBCG-vaccinated mice also exhibited altered lung histopathology compared to BCG-vaccinated mice and control mice with less well-developed lymphohistiocytic nodules in the lung parenchyma. At 26 weeks post-challenge, 4dBCG-vaccinated mice but not BCG-vaccinated mice had significantly fewer challenge bacilli in the lungs than control mice. In conclusion, despite reduced persistence in mice a modified BCG vaccine with diminished antioxidants and glutamine synthetase is superior to the parent vaccine in conferring protection against M. tuberculosis. The targeting of multiple immune suppressive factors produced by BCG is a promising strategy for simultaneously improving vaccine safety and effectiveness.

10.
Biochem Pharmacol ; 83(3): 368-77, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22146584

ABSTRACT

Methicillin-resistant Staphylococcus aureus (MRSA) is a human pathogen and a major cause of hospital-acquired infections. New antibacterial agents that have not been compromised by bacterial resistance are needed to treat MRSA-related infections. We chose the S. aureus cell wall synthesis enzyme, alanine racemase (Alr) as the target for a high-throughput screening effort to obtain novel enzyme inhibitors, which inhibit bacterial growth. Among the 'hits' identified was a thiadiazolidinone with chemical properties attractive for lead development. This study evaluated the mode of action, antimicrobial activities, and mammalian cell cytotoxicity of the thiadiazolidinone family in order to assess its potential for development as a therapeutic agent against MRSA. The thiadiazolidones inhibited Alr activity with 50% inhibitory concentrations (IC50) ranging from 0.36 to 6.4 µM, and they appear to inhibit the enzyme irreversibly. The series inhibited the growth of S. aureus, including MRSA strains, with minimal inhibitory concentrations (MICs) ranging from 6.25 to 100 µg/ml. The antimicrobial activity showed selectivity against Gram-positive bacteria and fungi, but not Gram-negative bacteria. The series inhibited human HeLa cell proliferation. Lead development centering on the thiadiazolidinone series would require additional medicinal chemistry efforts to enhance the antibacterial activity and minimize mammalian cell toxicity.


Subject(s)
Alanine Racemase/antagonists & inhibitors , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Methicillin-Resistant Staphylococcus aureus/enzymology , Thiadiazoles/chemistry , Thiadiazoles/pharmacology , Alanine Racemase/metabolism , Anti-Bacterial Agents/administration & dosage , Drug Delivery Systems/methods , HeLa Cells , Humans , Methicillin-Resistant Staphylococcus aureus/drug effects , Microbial Sensitivity Tests , Thiadiazoles/classification
11.
Am J Vet Res ; 72(11): 1465-75, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22023124

ABSTRACT

OBJECTIVE: To investigate the effect of opsonization of Rhodococcus equi with R. equi-specific antibodies in plasma on bacterial viability and phagocyte activation in a cell culture model of infection. SAMPLE: Neutrophils and monocyte-derived macrophages from 6 healthy 1-week-old foals and 1 adult horse. PROCEDURES: Foal and adult horse phagocytes were incubated with either opsonized or nonopsonized bacteria. Opsonization was achieved by use of plasma containing high or low concentrations of R. equi-specific antibodies. Phagocyte oxidative burst activity was measured by use of flow cytometry, and macrophage tumor necrosis factor (TNF)-α production was measured via an ELISA. Extracellular and intracellular bacterial viability was measured with a novel R. equi-luciferase construct that used a luminometer. RESULTS: Opsonized bacteria increased oxidative burst activity in adult horse phagocytes, and neutrophil activity was dependent on the concentration of specific antibody. Secretion of TNF-α was higher in macrophages infected with opsonized bacteria. Opsonization had no significant effect on bacterial viability in macrophages; however, extracellular bacterial viability was decreased in broth containing plasma with R. equi-specific antibodies, compared with viability in broth alone. CONCLUSIONS AND CLINICAL RELEVANCE: The use of plasma enriched with specific antibodies for the opsonization of R. equi increased the activation of phagocytes and decreased bacterial viability in the extracellular space. Although opsonized R. equi increased TNF-α secretion and oxidative burst in macrophages, additional factors may be necessary for effective intracellular bacterial killing. These data have suggested a possible role of plasma antibody in protection of foals from R. equi pneumonia.


Subject(s)
Actinomycetales Infections/veterinary , Bronchopneumonia/veterinary , Horse Diseases/immunology , Microbial Viability , Opsonin Proteins/metabolism , Phagocytosis , Rhodococcus equi/immunology , Actinomycetales Infections/immunology , Animals , Animals, Newborn , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Bacterial Proteins/blood , Bacterial Proteins/immunology , Bronchopneumonia/immunology , Enzyme-Linked Immunosorbent Assay/veterinary , Female , Horses , Macrophages/immunology , Macrophages/metabolism , Male , Neutrophils/immunology , Neutrophils/metabolism , Phagocytes/immunology , Phagocytes/metabolism , Respiratory Burst , Rhodococcus equi/physiology , Tumor Necrosis Factor-alpha/metabolism
12.
PLoS One ; 6(5): e20374, 2011.
Article in English | MEDLINE | ID: mdl-21637807

ABSTRACT

BACKGROUND: In an effort to discover new drugs to treat tuberculosis (TB) we chose alanine racemase as the target of our drug discovery efforts. In Mycobacterium tuberculosis, the causative agent of TB, alanine racemase plays an essential role in cell wall synthesis as it racemizes L-alanine into D-alanine, a key building block in the biosynthesis of peptidoglycan. Good antimicrobial effects have been achieved by inhibition of this enzyme with suicide substrates, but the clinical utility of this class of inhibitors is limited due to their lack of target specificity and toxicity. Therefore, inhibitors that are not substrate analogs and that act through different mechanisms of enzyme inhibition are necessary for therapeutic development for this drug target. METHODOLOGY/PRINCIPAL FINDINGS: To obtain non-substrate alanine racemase inhibitors, we developed a high-throughput screening platform and screened 53,000 small molecule compounds for enzyme-specific inhibitors. We examined the 'hits' for structural novelty, antimicrobial activity against M. tuberculosis, general cellular cytotoxicity, and mechanism of enzyme inhibition. We identified seventeen novel non-substrate alanine racemase inhibitors that are structurally different than any currently known enzyme inhibitors. Seven of these are active against M. tuberculosis and minimally cytotoxic against mammalian cells. CONCLUSIONS/SIGNIFICANCE: This study highlights the feasibility of obtaining novel alanine racemase inhibitor lead compounds by high-throughput screening for development of new anti-TB agents.


Subject(s)
Alanine Racemase/antagonists & inhibitors , Anti-Infective Agents/pharmacology , Enzyme Inhibitors/classification , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays/methods , Mycobacterium tuberculosis/drug effects , Alanine Dehydrogenase/metabolism , Alanine Racemase/chemistry , Alanine Racemase/metabolism , Alanine Racemase/pharmacology , Anti-Infective Agents/analysis , Anti-Infective Agents/chemistry , Anti-Infective Agents/classification , Cell Death/drug effects , Enzyme Inhibitors/analysis , Enzyme Inhibitors/chemistry , HeLa Cells , Humans , Inhibitory Concentration 50 , Kinetics , Mass Spectrometry , Microbial Sensitivity Tests , Substrate Specificity/drug effects
13.
Antimicrob Agents Chemother ; 54(8): 3478-80, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20516287

ABSTRACT

ACH-702 is a new isothiazoloquinolone with potent in vitro and in vivo activities against important bacterial pathogens, including Staphylococcus aureus. In this study, ACH-702 was found to have promising in vitro antibacterial activity against Mycobacterium tuberculosis, with MICs of

Subject(s)
Antitubercular Agents/pharmacology , Drug Resistance, Bacterial , Mycobacterium tuberculosis/drug effects , Quinolones/pharmacology , Thiazoles/pharmacology , Antitubercular Agents/chemistry , DNA Gyrase/genetics , DNA Gyrase/metabolism , Humans , Microbial Sensitivity Tests , Mycobacterium tuberculosis/isolation & purification , Quinolones/chemistry , Thiazoles/chemistry , Topoisomerase II Inhibitors , Tuberculosis/microbiology
14.
Antimicrob Agents Chemother ; 53(12): 5114-21, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19770283

ABSTRACT

Antileishmanial therapy is suboptimal due to toxicity, high cost, and development of resistance to available drugs. Pyrazinamide (PZA) is a constituent of short-course tuberculosis chemotherapy. We investigated the effect of PZA on Leishmania major promastigote and amastigote survival. Promastigotes were more sensitive to the drug than amastigotes, with concentrations at which 50% of parasites were inhibited (MIC(50)) of 16.1 and 8.2 microM, respectively (48 h posttreatment). Moreover, 90% of amastigotes were eliminated at 120 h posttreatment, indicating that longer treatments will result in parasite elimination. Most strikingly, PZA treatment of infected C57BL/6 mice resulted in protection against disease and in a 100-fold reduction in the parasite burden. PZA treatment of J774 cells and bone marrow-derived dendritic cells and macrophages increased interleukin 12, tumor necrosis factor alpha, and activation marker expression, as well as nitric oxide production, suggesting that PZA enhances effective immune responses against the parasite. PZA treatment also activates dendritic cells deficient in Toll-like receptor 2 and 4 expression to initiate a proinflammatory response, confirming that the immunostimulatory effect of PZA is directly caused by the drug and is independent of Toll-like receptor stimulation. These results not only are strongly indicative of the promise of PZA as an alternative antileishmanial chemotherapy but also suggest that PZA causes collateral immunostimulation, a phenomenon that has never been reported for this drug.


Subject(s)
Antiprotozoal Agents/therapeutic use , Antitubercular Agents/therapeutic use , Leishmaniasis, Cutaneous/drug therapy , Pyrazinamide/therapeutic use , Animals , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , Cell Line , Cell Survival/drug effects , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation/drug effects , Interleukin-10/metabolism , Interleukin-12/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Nitric Oxide/metabolism , Tumor Necrosis Factor-alpha/metabolism
15.
Eur J Med Chem ; 44(10): 4169-78, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19524330

ABSTRACT

Structural modification of the frontline antitubercular isonicotinic acid hydrazide (INH) provides lipophilic adaptations (3-46) of the drug in which the hydrazine moiety of the parent compound has been chemically blocked from the deactivating process of N(2)-acetylation by N-arylaminoacetyl transferases. As a class, these compounds show high levels of activity against Mycobacterium tuberculosis in vitro and in tuberculosis-infected macrophages. They provide strong protection in tuberculosis-infected mice and have low toxicity. With some representatives of this class achieving early peak plasma concentrations approximately three orders of magnitude above minimum inhibitory concentration, they may serve as tools for improving our understanding of INH-based treatment modalities, particularly for those patients chronically underdosed in conventional INH therapy.


Subject(s)
Antitubercular Agents/chemistry , Antitubercular Agents/therapeutic use , Isoniazid/chemistry , Isoniazid/therapeutic use , Mycobacterium tuberculosis/drug effects , Schiff Bases/chemistry , Tuberculosis/drug therapy , Animals , Antitubercular Agents/pharmacology , Female , Isoniazid/analogs & derivatives , Isoniazid/pharmacology , Macrophages/drug effects , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Structure-Activity Relationship
16.
Tuberculosis (Edinb) ; 88(5): 410-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18674969

ABSTRACT

Pyrazinamide is unusual among anti-tuberculous agents in its ability to promote a durable cure and shorten the duration of therapy. Yet the basis for this effect is not well understood. A particularly effective strategy for the development of new drugs can be to synthetically manipulate the well-established structures to improve either the spectrum of activity or some pharmacological properties. Similar to previously described aminomethylene amides such as morphazinamide, it was found that novel aminomethylene amides can have in vitro activity at higher than the very acidic pH conditions where pyrazinamide is inactive as well as retaining activity against pyrazinamide-resistant M. tuberculosis. These new compounds have shown an improved anti-tuberculous activity in infected human macrophages relative to pyrazinamide. Compound 1, in combination with rifamycin, was especially effective in both infected human macrophages and in a murine model of infection. The activity of these analogs against pyrazinamide-resistant strains suggests that the development of second generation pyrazinamide analogs may be especially fruitful.


Subject(s)
Antitubercular Agents/pharmacology , Monocytes/microbiology , Mycobacterium tuberculosis/drug effects , Pyrazinamide/analogs & derivatives , Animals , Drug Resistance, Multiple, Bacterial/drug effects , Humans , Macrophages/microbiology , Mice , Pyrazinamide/pharmacology , Treatment Outcome
17.
J Infect Chemother ; 10(3): 185-8, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15290460

ABSTRACT

The activity against Mycobacterium avium complex (MAC) of varying doses of grepafloxacin (GRE; 25 mg/kg, 50 mg/kg, 100 mg/kg, and 200 mg/kg) were compared to clarithromycin (CLA; 100 mg/kg and 200 mg/kg), ethambutol (EMB; 100 mg/kg), and rifabutin (RBT; 10 mg/kg) using an intranasal (IN) infection model compared to an intravenous (IV) infection model. Beige mice (C57BL6/J-Lyst bg J/+) were infected intranasally with about 10(6) organisms and for the IV model about 10(7) organisms. Treatment for both models was started 1 week postinfection and given by gavage 5 days/week for 4 weeks. At the initiation of therapy, an early control group was killed to determine the initial organism load. Three days following the completion of therapy, drug-treated groups of mice and the late control group were killed and the response to therapy measured. The most effective agents were CLA and RBT. GRE and EMB had modest activities in both the IN and the IV models. A matched comparison between IN and IV challenges for each of the agents used revealed greater suppression of MAC in the IN model compared to the IV model.


Subject(s)
Fluoroquinolones/pharmacology , Mycobacterium avium Complex/drug effects , Piperazines/pharmacology , Administration, Intranasal , Animals , Clarithromycin/administration & dosage , Clarithromycin/pharmacology , Disease Models, Animal , Ethambutol/administration & dosage , Ethambutol/pharmacology , Female , Fluoroquinolones/administration & dosage , Injections, Intravenous , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Piperazines/administration & dosage , Random Allocation , Rifabutin/administration & dosage , Rifabutin/pharmacology
18.
Scand J Infect Dis ; 36(2): 150-4, 2004.
Article in English | MEDLINE | ID: mdl-15061673

ABSTRACT

A case of recurrent Mycobacterium xenopi infection presenting as Pott's disease in a patient receiving etanercept for severe rheumatoid arthritis is described. A 49-y-old Caucasian male had received a total of 11 months of anti-mycobacterial therapy for hip infection acquired 15 months earlier; he presented with progressive back pain, which was diagnosed as Pott's disease. He had been treated with etanercept in addition to his prior immunosuppressive agents after the diagnosis of hip infection.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Immunoglobulin G/therapeutic use , Mycobacterium Infections, Nontuberculous/diagnosis , Mycobacterium xenopi/isolation & purification , Prosthesis-Related Infections/drug therapy , Receptors, Tumor Necrosis Factor/therapeutic use , Arthritis, Rheumatoid/complications , Arthritis, Rheumatoid/diagnosis , DNA, Bacterial/analysis , Etanercept , Follow-Up Studies , Hip Prosthesis/adverse effects , Humans , Male , Middle Aged , Mycobacterium Infections, Nontuberculous/complications , Mycobacterium Infections, Nontuberculous/drug therapy , Polymorphism, Restriction Fragment Length , Prosthesis-Related Infections/diagnosis , Recurrence , Risk Assessment , Severity of Illness Index , Treatment Outcome
19.
J Antimicrob Chemother ; 53(4): 641-5, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14973154

ABSTRACT

OBJECTIVE: Designing a more rapid method to test antimycobacterial agents in a murine model would significantly improve the drug development process. We describe a short-course in vivo treatment model that could be used to screen potential antituberculous drugs. METHODS: In this model, C57BL/6 mice were infected intranasally with approximately 10(6) viable Mycobacterium tuberculosis organisms. Treatment began 1 day post-infection and was administered for 2 days. Mice were euthanized 3 days post-infection and their right lungs were removed and cell counts determined. Several antimycobacterial agents with superior in vivo activity in a 4 week treatment model were tested to evaluate the short-course treatment model. RESULTS: Two days of isoniazid (25 mg/kg), rifampicin (20 mg/kg), PNU-100480 (100 mg/kg), gatifloxacin (100 mg/kg), levofloxacin (100 mg/kg) and sparfloxacin (100 mg/kg) were all able to significantly reduce the mycobacterial load in the lungs compared with the untreated control mice. CONCLUSIONS: Use of this model to screen potential chemotherapeutic agents will save time and resources.


Subject(s)
Antitubercular Agents/administration & dosage , Disease Models, Animal , Tuberculosis/drug therapy , Acetamides/administration & dosage , Animals , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Evaluation, Preclinical , Female , Fluoroquinolones/administration & dosage , Gatifloxacin , Isoniazid/administration & dosage , Linezolid , Mice , Mice, Inbred C57BL , Oxazolidinones/administration & dosage , Rifampin/administration & dosage , Time Factors , Tuberculosis/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...