Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
PLoS Negl Trop Dis ; 15(3): e0009013, 2021 03.
Article in English | MEDLINE | ID: mdl-33651812

ABSTRACT

BACKGROUND: There is a continued need to develop effective and safe treatments for visceral leishmaniasis (VL). Preclinical studies on pharmacokinetics and pharmacodynamics of anti-infective agents, such as anti-bacterials and anti-fungals, have provided valuable information in the development and dosing of these agents. The aim of this study was to characterise the pharmacokinetic and pharmacodynamic properties of the anti-leishmanial drugs AmBisome and miltefosine in a preclinical disease model of VL. METHODOLOGY / PRINCIPAL FINDINGS: BALB/c mice were infected with L. donovani (MHOM/ET/67/HU3) amastigotes. Groups of mice were treated with miltefosine (orally, multi-dose regimen) or AmBisome (intravenously, single dose regimen) or left untreated as control groups. At set time points groups of mice were killed and plasma, livers and spleens harvested. For pharmacodynamics the hepatic parasite burden was determined microscopically from tissue impression smears. For pharmacokinetics drug concentrations were measured in plasma and whole tissue homogenates by LC-MS. Unbound drug concentrations were determined by rapid equilibrium dialysis. Doses exerting maximum anti-leishmanial effects were 40 mg/kg for AmBisome and 150 mg/kg (cumulatively) for miltefosine. AmBisome displayed a wider therapeutic range than miltefosine. Dose fractionation at a total dose of 2.5 mg/kg pointed towards concentration-dependent anti-leishmanial activity of AmBisome, favouring the administration of large doses infrequently. Protein binding was >99% for miltefosine and amphotericin B in plasma and tissue homogenates. CONCLUSION / SIGNIFICANCE: Using a PK/PD approach we propose optimal dosing strategies for AmBisome. Additionally, we describe pharmacokinetic and pharmacodynamic properties of miltefosine and compare our findings in a preclinical disease model to available knowledge from studies in humans. This approach also presents a strategy for improved use of animal models in the drug development process for VL.


Subject(s)
Amphotericin B/pharmacokinetics , Antiprotozoal Agents/pharmacokinetics , Leishmaniasis, Visceral/drug therapy , Phosphorylcholine/analogs & derivatives , Amphotericin B/therapeutic use , Animals , Antiprotozoal Agents/therapeutic use , Drug Therapy, Combination , Homeodomain Proteins/genetics , Humans , Liver/parasitology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Parasite Load , Phosphorylcholine/pharmacokinetics , Phosphorylcholine/therapeutic use , Protein Binding/physiology
2.
Antimicrob Agents Chemother ; 60(10): 6271-80, 2016 10.
Article in English | MEDLINE | ID: mdl-27503647

ABSTRACT

The recent development and spread of extensively drug-resistant and totally drug-resistant resistant (TDR) strains of Mycobacterium tuberculosis highlight the need for new antitubercular drugs. Protein synthesis inhibitors have played an important role in the treatment of tuberculosis (TB) starting with the inclusion of streptomycin in the first combination therapies. Although parenteral aminoglycosides are a key component of therapy for multidrug-resistant TB, the oxazolidinone linezolid is the only orally available protein synthesis inhibitor that is effective against TB. Here, we show that small-molecule inhibitors of aminoacyl-tRNA synthetases (AARSs), which are known to be excellent antibacterial protein synthesis targets, are orally bioavailable and effective against M. tuberculosis in TB mouse infection models. We applied the oxaborole tRNA-trapping (OBORT) mechanism, which was first developed to target fungal cytoplasmic leucyl-tRNA synthetase (LeuRS), to M. tuberculosis LeuRS. X-ray crystallography was used to guide the design of LeuRS inhibitors that have good biochemical potency and excellent whole-cell activity against M. tuberculosis Importantly, their good oral bioavailability translates into in vivo efficacy in both the acute and chronic mouse models of TB with potency comparable to that of the frontline drug isoniazid.


Subject(s)
Antitubercular Agents/pharmacology , Leucine-tRNA Ligase/antagonists & inhibitors , Mycobacterium tuberculosis/drug effects , Protein Synthesis Inhibitors/pharmacology , Administration, Oral , Animals , Antitubercular Agents/administration & dosage , Antitubercular Agents/chemistry , Antitubercular Agents/pharmacokinetics , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Female , Humans , Leucine-tRNA Ligase/chemistry , Leucine-tRNA Ligase/genetics , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred Strains , Microbial Sensitivity Tests , Mycobacterium smegmatis/drug effects , Mycobacterium smegmatis/genetics , Mycobacterium tuberculosis/genetics , Protein Synthesis Inhibitors/administration & dosage , Protein Synthesis Inhibitors/chemistry , Protein Synthesis Inhibitors/pharmacokinetics , Structure-Activity Relationship , Tuberculosis/drug therapy , Vero Cells
3.
Sci Transl Med ; 7(296): 296ra111, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26180101

ABSTRACT

Malaria is one of the most significant causes of childhood mortality, but disease control efforts are threatened by resistance of the Plasmodium parasite to current therapies. Continued progress in combating malaria requires development of new, easy to administer drug combinations with broad-ranging activity against all manifestations of the disease. DSM265, a triazolopyrimidine-based inhibitor of the pyrimidine biosynthetic enzyme dihydroorotate dehydrogenase (DHODH), is the first DHODH inhibitor to reach clinical development for treatment of malaria. We describe studies profiling the biological activity, pharmacological and pharmacokinetic properties, and safety of DSM265, which supported its advancement to human trials. DSM265 is highly selective toward DHODH of the malaria parasite Plasmodium, efficacious against both blood and liver stages of P. falciparum, and active against drug-resistant parasite isolates. Favorable pharmacokinetic properties of DSM265 are predicted to provide therapeutic concentrations for more than 8 days after a single oral dose in the range of 200 to 400 mg. DSM265 was well tolerated in repeat-dose and cardiovascular safety studies in mice and dogs, was not mutagenic, and was inactive against panels of human enzymes/receptors. The excellent safety profile, blood- and liver-stage activity, and predicted long half-life in humans position DSM265 as a new potential drug combination partner for either single-dose treatment or once-weekly chemoprevention. DSM265 has advantages over current treatment options that are dosed daily or are inactive against the parasite liver stage.


Subject(s)
Antimalarials/chemistry , Enzyme Inhibitors/chemistry , Malaria, Falciparum/drug therapy , Malaria, Falciparum/prevention & control , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Pyrimidines/chemistry , Triazoles/chemistry , Administration, Oral , Animals , Antimalarials/pharmacokinetics , Area Under Curve , Caco-2 Cells , Crystallography, X-Ray , Dihydroorotate Dehydrogenase , Dogs , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacokinetics , Haplorhini , Humans , Inhibitory Concentration 50 , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Sequence Data , Oxidoreductases Acting on CH-CH Group Donors/chemistry , Plasmodium falciparum , Pyrimidines/pharmacokinetics , Rabbits , Substrate Specificity , Triazoles/pharmacokinetics
4.
J Infect Dis ; 211 Suppl 3: S96-S106, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-26009618

ABSTRACT

BACKGROUND: Antimicrobial pharmacokinetic-pharmacodynamic (PK/PD) science is vital to early antibiotic drug development to enable more efficient dose-effect study designs, identification of doses that may suppress drug resistance and choice of susceptibility breakpoints. Proper conduct of such studies is essential in the field of tuberculosis. METHODS: We conducted an exhaustive review of literature on the hollow fiber system (HFS) model, murine model, and guinea pig model of tuberculosis as well as clinical studies to identify PK/PD studies that have been applied to antituberculosis therapy. Lessons learned are presented as recommendations and standards for both industry and academia in the field of antituberculosis drug development. RESULTS: PK/PD studies have been performed for both first-line and experimental antituberculosis agents. When properly designed exposure-effect and dose-fractionation studies have been performed in preclinical models, optimal drug exposures, and PK/PD parameters identified in these models have been found to be similar to clinical studies. Susceptibility breakpoints identified using these methods differed from previous concentrations in the literature but were found to be similar to those in prospective clinical studies. CONCLUSIONS: Preclinical PK/PD studies are essential value added in the development of antituberculosis agents. We provide 8 recommendations and standards for the proper conduct of such studies.


Subject(s)
Antitubercular Agents/pharmacology , Antitubercular Agents/pharmacokinetics , Tuberculosis/drug therapy , Animals , Clinical Trials as Topic , Drug Evaluation, Preclinical , Humans
5.
PLoS One ; 8(6): e66967, 2013.
Article in English | MEDLINE | ID: mdl-23825598

ABSTRACT

The emergence of resistance to available antimalarials requires the urgent development of new medicines. The recent disclosure of several thousand compounds active in vitro against the erythrocyte stage of Plasmodium falciparum has been a major breakthrough, though converting these hits into new medicines challenges current strategies. A new in vivo screening concept was evaluated as a strategy to increase the speed and efficiency of drug discovery projects in malaria. The new in vivo screening concept was developed based on human disease parameters, i.e. parasitemia in the peripheral blood of patients on hospital admission and parasite reduction ratio (PRR), which were allometrically down-scaled into P. berghei-infected mice. Mice with an initial parasitemia (P0) of 1.5% were treated orally for two consecutive days and parasitemia measured 24 h after the second dose. The assay was optimized for detection of compounds able to stop parasite replication (PRR = 1) or induce parasite clearance (PRR >1) with statistical power >99% using only two mice per experimental group. In the P. berghei in vivo screening assay, the PRR of a set of eleven antimalarials with different mechanisms of action correlated with human-equivalent data. Subsequently, 590 compounds from the Tres Cantos Antimalarial Set with activity in vitro against P. falciparum were tested at 50 mg/kg (orally) in an assay format that allowed the evaluation of hundreds of compounds per month. The rate of compounds with detectable efficacy was 11.2% and about one third of active compounds showed in vivo efficacy comparable with the most potent antimalarials used clinically. High-throughput, high-content in vivo screening could rapidly select new compounds, dramatically speeding up the discovery of new antimalarial medicines. A global multilateral collaborative project aimed at screening the significant chemical diversity within the antimalarial in vitro hits described in the literature is a feasible task.


Subject(s)
Antimalarials/pharmacology , Drug Evaluation, Preclinical/methods , Plasmodium berghei/drug effects , Animals , Antimalarials/therapeutic use , Feasibility Studies , Female , Humans , Malaria/complications , Malaria/drug therapy , Mice , Parasitemia/complications , Plasmodium berghei/physiology , Time Factors
6.
Tuberculosis (Edinb) ; 92(6): 453-88, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22940006

ABSTRACT

In drug development, there are typically a series of preclinical studies that must be completed with new compounds or regimens before use in humans. A sequence of in vitro assays followed by in vivo testing in validated animal models to assess the activity against Mycobacterium tuberculosis, pharmacology and toxicity is generally used for advancing compounds against tuberculosis in a preclinical stage. A plethora of different assay systems and conditions are used to study the effect of drug candidates on the growth of M. tuberculosis, making it difficult to compare data from one laboratory to another. The Bill and Melinda Gates Foundation recognized the scientific gap to delineate the spectrum of variables in experimental protocols, identify which of these are biologically significant, and converge towards a rationally derived standard set of optimized assays for evaluating compounds. The goals of this document are to recommend protocols and hence accelerate the process of TB drug discovery and testing. Data gathered from preclinical in vitro and in vivo assays during personal visits to laboratories and an electronic survey of methodologies sent to investigators is reported. Comments, opinions, experiences as well as final recommendations from those currently engaged in such preclinical studies for TB drug testing are being presented. Certain in vitro assays and mouse efficacy models were re-evaluated in the laboratory as head-to-head experiments and a summary is provided on the results obtained. It is our hope that this information will be a valuable resource for investigators in the field to move forward in an efficient way and that key variables of assays are included to ensure accuracy of results which can then be used for designing human clinical trials. This document then concludes with remaining questions and critical gaps that are in need of further validation and experimentation.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proteins/drug effects , Drug Evaluation, Preclinical/methods , Mycobacterium tuberculosis/drug effects , Tuberculosis/drug therapy , Animals , Antitubercular Agents/pharmacokinetics , Disease Models, Animal , Drug Design , Drug Evaluation, Preclinical/trends , Humans , Mice , Mycobacterium tuberculosis/pathogenicity , Tuberculosis/prevention & control
7.
J Biol Chem ; 285(43): 33054-33064, 2010 Oct 22.
Article in English | MEDLINE | ID: mdl-20702404

ABSTRACT

Plasmodium falciparum, the causative agent of the most deadly form of human malaria, is unable to salvage pyrimidines and must rely on de novo biosynthesis for survival. Dihydroorotate dehydrogenase (DHODH) catalyzes the rate-limiting step in the pyrimidine biosynthetic pathway and represents a potential target for anti-malarial therapy. A high throughput screen and subsequent medicinal chemistry program identified a series of N-alkyl-5-(1H-benzimidazol-1-yl)thiophene-2-carboxamides with low nanomolar in vitro potency against DHODH from P. falciparum, P. vivax, and P. berghei. The compounds were selective for the parasite enzymes over human DHODH, and x-ray structural data on the analog Genz-667348, demonstrated that species selectivity could be attributed to amino acid differences in the inhibitor-binding site. Compounds from this series demonstrated in vitro potency against the 3D7 and Dd2 strains of P. falciparum, good tolerability and oral exposure in the mouse, and ED(50) values in the 4-day murine P. berghei efficacy model of 13-21 mg/kg/day with oral twice-daily dosing. In particular, treatment with Genz-667348 at 100 mg/kg/day resulted in sterile cure. Two recent analogs of Genz-667348 are currently undergoing pilot toxicity testing to determine suitability as clinical development candidates.


Subject(s)
Antimalarials/pharmacology , Enzyme Inhibitors/pharmacology , Malaria, Falciparum/drug therapy , Malaria, Falciparum/enzymology , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Plasmodium falciparum/enzymology , Protozoan Proteins/antagonists & inhibitors , Animals , Cell Line , Dihydroorotate Dehydrogenase , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Imidazoles/pharmacology , Mice , Mice, Inbred NOD , Mice, SCID , Plasmodium berghei/enzymology , Plasmodium vivax/enzymology , Rats
8.
Nature ; 465(7296): 311-5, 2010 May 20.
Article in English | MEDLINE | ID: mdl-20485428

ABSTRACT

Malaria caused by Plasmodium falciparum is a disease that is responsible for 880,000 deaths per year worldwide. Vaccine development has proved difficult and resistance has emerged for most antimalarial drugs. To discover new antimalarial chemotypes, we have used a phenotypic forward chemical genetic approach to assay 309,474 chemicals. Here we disclose structures and biological activity of the entire library-many of which showed potent in vitro activity against drug-resistant P. falciparum strains-and detailed profiling of 172 representative candidates. A reverse chemical genetic study identified 19 new inhibitors of 4 validated drug targets and 15 novel binders among 61 malarial proteins. Phylochemogenetic profiling in several organisms revealed similarities between Toxoplasma gondii and mammalian cell lines and dissimilarities between P. falciparum and related protozoans. One exemplar compound displayed efficacy in a murine model. Our findings provide the scientific community with new starting points for malaria drug discovery.


Subject(s)
Antimalarials/analysis , Antimalarials/pharmacology , Drug Discovery , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Animals , Antimalarials/isolation & purification , Cell Line , Drug Evaluation, Preclinical , Drug Resistance/drug effects , Drug Therapy, Combination , Erythrocytes/drug effects , Erythrocytes/parasitology , Humans , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Mice , Phenotype , Phylogeny , Plasmodium falciparum/metabolism , Reproducibility of Results , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
9.
Antimicrob Agents Chemother ; 54(5): 2262-4, 2010 May.
Article in English | MEDLINE | ID: mdl-20160054

ABSTRACT

Murine models of Mycobacterium tuberculosis infection are essential tools in drug discovery. Here we describe a fast standardized 9-day acute assay intended to measure the efficacy of drugs against M. tuberculosis growing in the lungs of immunocompetent mice. This assay is highly reproducible, allows good throughput, and was validated for drug lead optimization using isoniazid, rifampin, ethambutol, pyrazinamide, linezolid, and moxifloxacin.


Subject(s)
Antitubercular Agents/pharmacology , Disease Models, Animal , Drug Discovery , Mice, Inbred C57BL , Tuberculosis, Pulmonary/drug therapy , Acetamides/pharmacology , Animals , Aza Compounds/pharmacology , Ethambutol/pharmacology , Fluoroquinolones , Immunocompetence , Inhalation Exposure , Isoniazid/pharmacology , Linezolid , Mice , Moxifloxacin , Oxazolidinones/pharmacology , Pyrazinamide/pharmacology , Quinolines/pharmacology , Reproducibility of Results , Rifampin/pharmacology , Tuberculosis, Pulmonary/immunology
SELECTION OF CITATIONS
SEARCH DETAIL