Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
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.
Eur J Clin Microbiol Infect Dis ; 39(5): 799-826, 2020 May.
Article in English | MEDLINE | ID: mdl-31853742

ABSTRACT

Currently, there is a trend of increasing incidence in pulmonary non-tuberculous mycobacterial infections (PNTM) together with a decrease in tuberculosis (TB) incidence, particularly in developed countries. The prevalence of PNTM in underdeveloped and developing countries remains unclear as there is still a lack of detection methods that could clearly diagnose PNTM applicable in these low-resource settings. Since non-tuberculous mycobacteria (NTM) are environmental pathogens, the vicinity favouring host-pathogen interactions is known as important predisposing factor for PNTM. The ongoing changes in world population, as well as socio-political and economic factors, are linked to the rise in the incidence of PNTM. Development is an important factor for the improvement of population well-being, but it has also been linked, in general, to detrimental environmental consequences, including the rise of emergent (usually neglected) infectious diseases, such as PNTM. The rise of neglected PNTM infections requires the expansion of the current efforts on the development of diagnostics, therapies and vaccines for mycobacterial diseases, which at present, are mainly focused on TB. This review discuss the current situation of PNTM and its predisposing factors, as well as the efforts and challenges for their control.


Subject(s)
Disease Management , Mycobacterium Infections, Nontuberculous/microbiology , Respiratory Tract Infections/microbiology , Animals , Clinical Trials as Topic , Global Health , Host-Pathogen Interactions , Humans , Mycobacterium Infections, Nontuberculous/epidemiology , Mycobacterium Infections, Nontuberculous/prevention & control , Nontuberculous Mycobacteria , Prevalence , Respiratory Tract Infections/complications
3.
Article in English | MEDLINE | ID: mdl-31405862

ABSTRACT

The Mycobacterium tuberculosis mycolate flippase MmpL3 has been the proposed target for multiple inhibitors with diverse chemical scaffolds. This diversity in chemical scaffolds has made it difficult to predict compounds that inhibit MmpL3 without whole-genome sequencing of isolated resistant mutants. Here, we describe the identification of four new inhibitors that select for resistance mutations in mmpL3. Using these resistant mutants, we conducted a targeted whole-cell phenotypic screen of 163 novel M. tuberculosis growth inhibitors for differential growth inhibition of wild-type M. tuberculosis compared to the growth of a pool of 24 unique mmpL3 mutants. The screen successfully identified six additional putative MmpL3 inhibitors. The compounds were bactericidal both in vitro and against intracellular M. tuberculosisM. tuberculosis cells treated with these compounds were shown to accumulate trehalose monomycolates, have reduced levels of trehalose dimycolate, and displace an MmpL3-specific probe, supporting MmpL3 as the target. The inhibitors were mycobacterium specific, with several also showing activity against the nontuberculous mycobacterial species M. abscessus Cluster analysis of cross-resistance profiles generated by dose-response experiments for each combination of 13 MmpL3 inhibitors against each of the 24 mmpL3 mutants defined two clades of inhibitors and two clades of mmpL3 mutants. Pairwise combination studies of the inhibitors revealed interactions that were specific to the clades identified in the cross-resistance profiling. Additionally, modeling of resistance-conferring substitutions to the MmpL3 crystal structure revealed clade-specific localization of the residues to specific domains of MmpL3, with the clades showing differential resistance. Several compounds exhibited high solubility and stability in microsomes and low cytotoxicity in macrophages, supporting their further development. The combined study of multiple mutants and novel compounds provides new insights into structure-function interactions of MmpL3 and small-molecule inhibitors.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proteins/genetics , Benzamides/pharmacology , Benzothiazoles/pharmacology , Drug Resistance, Bacterial/drug effects , Membrane Transport Proteins/genetics , Mycobacterium tuberculosis/drug effects , Pyridines/pharmacology , Antitubercular Agents/chemical synthesis , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Benzamides/chemical synthesis , Benzothiazoles/chemical synthesis , Binding Sites , Biological Transport/drug effects , Cord Factors/antagonists & inhibitors , Cord Factors/biosynthesis , Cord Factors/metabolism , Drug Resistance, Bacterial/genetics , Galactans/metabolism , Gene Expression , High-Throughput Screening Assays , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Microbial Sensitivity Tests , Models, Molecular , Mutation , Mycobacterium abscessus/drug effects , Mycobacterium abscessus/genetics , Mycobacterium abscessus/growth & development , Mycobacterium abscessus/metabolism , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/metabolism , Mycolic Acids/metabolism , Protein Binding , Protein Structure, Secondary , Pyridines/chemical synthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Whole Genome Sequencing
4.
Article in English | MEDLINE | ID: mdl-30803965

ABSTRACT

Fluoroquinolones represent the pillar of multidrug-resistant tuberculosis (MDR-TB) treatment, with moxifloxacin, levofloxacin, or gatifloxacin being prescribed to MDR-TB patients. Recently, several clinical trials of "universal" drug regimens, aiming to treat drug-susceptible and drug-resistant TB, have included a fluoroquinolone. In the absence of clinical data comparing their side-by-side efficacies in controlled MDR-TB trials, a pharmacological rationale is needed to guide the selection of the most efficacious fluoroquinolone. The present studies were designed to test the hypothesis that fluoroquinolone concentrations (pharmacokinetics) and activity (pharmacodynamics) at the site of infection are better predictors of efficacy than the plasma concentrations and potency measured in standard growth inhibition assays and are better suited to determinations of whether one of the fluoroquinolones outperforms the others in rabbits with active TB. We first measured the penetration of these fluoroquinolones in lung lesion compartments, and their potency against bacterial populations that reside in each compartment, to compute lesion-centric pharmacokinetic-pharmacodynamic (PK/PD) parameters. PK modeling methods were used to quantify drug penetration from plasma to tissues at human-equivalent doses. On the basis of these metrics, moxifloxacin emerged with a clear advantage, whereas plasma-based PK/PD favored levofloxacin (the ranges of the plasma AUC/MIC ratio [i.e., the area under the concentration-time curve over 24 h in the steady state divided by the MIC] are 46 to 86 for moxifloxacin and 74 to 258 for levofloxacin). A comparative efficacy trial in the rabbit model of active TB demonstrated the superiority of moxifloxacin in reducing bacterial burden at the lesion level and in sterilizing cellular and necrotic lesions. Collectively, these results show that PK/PD data obtained at the site of infection represent an adequate predictor of drug efficacy against TB and constitute the baseline required to explore synergies, antagonism, and drug-drug interactions in fluoroquinolone-containing regimens.


Subject(s)
Antitubercular Agents/therapeutic use , Fluoroquinolones/therapeutic use , Animals , Levofloxacin/therapeutic use , Microbial Sensitivity Tests , Moxifloxacin/therapeutic use , Rabbits , Tandem Mass Spectrometry , Tuberculosis, Multidrug-Resistant/drug therapy
5.
ACS Infect Dis ; 5(4): 598-617, 2019 04 12.
Article in English | MEDLINE | ID: mdl-30652474

ABSTRACT

The synthesis, absolute stereochemical configuration, complete biological characterization, mechanism of action and resistance, and pharmacokinetic properties of ( S)-(-)-acidomycin are described. Acidomycin possesses promising antitubercular activity against a series of contemporary drug susceptible and drug-resistant M. tuberculosis strains (minimum inhibitory concentrations (MICs) = 0.096-6.2 µM) but is inactive against nontuberculosis mycobacteria and Gram-positive and Gram-negative pathogens (MICs > 1000 µM). Complementation studies with biotin biosynthetic pathway intermediates and subsequent biochemical studies confirmed acidomycin inhibits biotin synthesis with a Ki of approximately 1 µM through the competitive inhibition of biotin synthase (BioB) and also stimulates unproductive cleavage of S-adenosyl-l-methionine (SAM) to generate the toxic metabolite 5'-deoxyadenosine. Cell studies demonstrate acidomycin selectively accumulates in M. tuberculosis providing a mechanistic basis for the observed antibacterial activity. The development of spontaneous resistance by M. tuberculosis to acidomycin was difficult, and only low-level resistance to acidomycin was observed by overexpression of BioB. Collectively, the results provide a foundation to advance acidomycin and highlight BioB as a promising target.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Sulfurtransferases/antagonists & inhibitors , Thiazolidines/pharmacology , Tuberculosis/microbiology , Animals , Antitubercular Agents/chemical synthesis , Antitubercular Agents/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biological Products/chemical synthesis , Biological Products/chemistry , Biological Products/pharmacology , Biotin/biosynthesis , Caproates/chemical synthesis , Caproates/chemistry , Caproates/pharmacology , Drug Resistance, Bacterial , Humans , Kinetics , Mice , Microbial Sensitivity Tests , Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/genetics , Sulfurtransferases/chemistry , Sulfurtransferases/genetics , Sulfurtransferases/metabolism , Thiazolidines/chemical synthesis , Thiazolidines/chemistry , Tuberculosis/drug therapy
6.
J Exp Med ; 215(8): 1975-1986, 2018 08 06.
Article in English | MEDLINE | ID: mdl-30018074

ABSTRACT

In the 1970s, inclusion of pyrazinamide (PZA) in the drug regimen of tuberculosis (TB) patients for the first 2 mo achieved a drastic reduction of therapy duration. Until now, however, the mechanisms underlying PZA's unique contribution to efficacy have remained controversial, and animal efficacy data vary across species. To understand how PZA kills bacterial populations present in critical lung lesion compartments, we first characterized a rabbit model of active TB, showing striking similarities in lesion types and fates to nonhuman primate models deemed the most appropriate surrogates of human TB. We next employed this model with lesion-centric molecular and bacteriology readouts to demonstrate that PZA exhibits potent activity against Mycobacterium tuberculosis residing in difficult-to-sterilize necrotic lesions. Our data also indicate that PZA is slow acting, suggesting that PZA administration beyond the first 2 mo may accelerate the cure. In conclusion, we provide a pharmacodynamic explanation for PZA's treatment-shortening effect and deliver new tools to dissect the contribution of immune response versus drug at the lesion level.


Subject(s)
Antitubercular Agents/therapeutic use , Pyrazinamide/therapeutic use , Tuberculosis/drug therapy , Tuberculosis/immunology , Animals , Antitubercular Agents/pharmacology , Disease Models, Animal , Female , Humans , Microbial Viability/drug effects , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/physiology , Necrosis , Neutrophils/drug effects , Neutrophils/metabolism , Pyrazinamide/pharmacology , Rabbits , Tuberculosis/microbiology , Tuberculosis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL