Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
PLoS Comput Biol ; 19(6): e1010823, 2023 06.
Article in English | MEDLINE | ID: mdl-37319311

ABSTRACT

Tuberculosis (TB) continues to be one of the deadliest infectious diseases in the world, causing ~1.5 million deaths every year. The World Health Organization initiated an End TB Strategy that aims to reduce TB-related deaths in 2035 by 95%. Recent research goals have focused on discovering more effective and more patient-friendly antibiotic drug regimens to increase patient compliance and decrease emergence of resistant TB. Moxifloxacin is one promising antibiotic that may improve the current standard regimen by shortening treatment time. Clinical trials and in vivo mouse studies suggest that regimens containing moxifloxacin have better bactericidal activity. However, testing every possible combination regimen with moxifloxacin either in vivo or clinically is not feasible due to experimental and clinical limitations. To identify better regimens more systematically, we simulated pharmacokinetics/pharmacodynamics of various regimens (with and without moxifloxacin) to evaluate efficacies, and then compared our predictions to both clinical trials and nonhuman primate studies performed herein. We used GranSim, our well-established hybrid agent-based model that simulates granuloma formation and antibiotic treatment, for this task. In addition, we established a multiple-objective optimization pipeline using GranSim to discover optimized regimens based on treatment objectives of interest, i.e., minimizing total drug dosage and lowering time needed to sterilize granulomas. Our approach can efficiently test many regimens and successfully identify optimal regimens to inform pre-clinical studies or clinical trials and ultimately accelerate the TB regimen discovery process.


Subject(s)
Tuberculosis, Multidrug-Resistant , Tuberculosis , Animals , Mice , Antitubercular Agents , Moxifloxacin/therapeutic use , Tuberculosis/drug therapy
2.
Antimicrob Agents Chemother ; 66(9): e0079022, 2022 09 20.
Article in English | MEDLINE | ID: mdl-36047786

ABSTRACT

Bioactive forms of oral ß-lactams were screened in vitro against Mycobacterium abscessus with and without the bioactive form of the oral ß-lactamase inhibitor avibactam ARX1796. Sulopenem was equally active without avibactam, while tebipenem, cefuroxime, and amoxicillin required avibactam for optimal activity. Systematic pairwise combination of the four ß-lactams revealed strong bactericidal synergy for each of sulopenem, tebipenem, and cefuroxime combined with amoxicillin in the presence of avibactam. These all-oral ß-lactam combinations warrant clinical evaluation.


Subject(s)
Lung Diseases , Mycobacterium abscessus , Amoxicillin/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Azabicyclo Compounds/pharmacology , Carbapenems , Cefuroxime , Humans , Lactams , Lung Diseases/drug therapy , Microbial Sensitivity Tests , beta-Lactamase Inhibitors/pharmacology , beta-Lactamase Inhibitors/therapeutic use , beta-Lactamases , beta-Lactams/pharmacology
3.
Sci Transl Med ; 14(633): eabj3860, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35196022

ABSTRACT

A search for alternative Mycobacterium abscessus treatments led to our interest in the two-component regulator DosRS, which, in Mycobacterium tuberculosis, is required for the bacterium to establish a state of nonreplicating, drug-tolerant persistence in response to a variety of host stresses. We show here that the genetic disruption of dosRS impairs the adaptation of M. abscessus to hypoxia, resulting in decreased bacterial survival after oxygen depletion, reduced tolerance to a number of antibiotics in vitro and in vivo, and the inhibition of biofilm formation. We determined that three antimalarial drugs or drug candidates, artemisinin, OZ277, and OZ439, can target DosS-mediated hypoxic signaling in M. abscessus and recapitulate the phenotypic effects of genetically disrupting dosS. OZ439 displayed bactericidal activity comparable to standard-of-care antibiotics in chronically infected mice, in addition to potentiating the activity of antibiotics used in combination. The identification of antimalarial drugs as potent inhibitors and adjunct inhibitors of M. abscessus in vivo offers repurposing opportunities that could have an immediate impact in the clinic.


Subject(s)
Antimalarials , Mycobacterium Infections, Nontuberculous , Mycobacterium abscessus , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Antimalarials/pharmacology , Antimalarials/therapeutic use , Mice , Microbial Sensitivity Tests , Mycobacterium Infections, Nontuberculous/drug therapy , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium abscessus/physiology
5.
Article in English | MEDLINE | ID: mdl-33468476

ABSTRACT

Intra-abdominal candidiasis (IAC) is one of the most common yet underappreciated forms of invasive candidiasis. IAC is difficult to treat, and therapeutic failure and drug-resistant breakthrough infections are common in some institutions despite the use of echinocandins as first-line agents. Fosmanogepix (FMGX, formerly APX001) is a first-in-class antifungal prodrug that can be administered both intravenously and orally. FMGX is currently in phase 2 clinical development for the treatment of life-threatening invasive fungal infections. To explore the pharmacological properties and therapeutic potential of FMGX for IAC, we evaluated both drug penetration and efficacy of the active moiety manogepix (MGX, formerly APX001A) in liver tissues in a clinically relevant IAC mouse model infected with Candida albicans Matrix-assisted laser desorption ionization-mass spectrometry imaging (MALDI-MSI) and laser capture microdissection (LCM)-directed absolute drug quantitation were employed to evaluate drug penetration into liver abscess lesions both spatially and quantitatively. The partitioning of MGX into lesions occurred slowly after a single dose; however, robust accumulation in the lesion was achieved after 3 days of repeated dosing. Associated with this drug penetration pattern, reduction in fungal burden and clearance in the liver were observed in mice receiving the multiday FMGX regimen. In comparison, administration of micafungin resulted in marginal reduction in fungal burden at the end of 4 days of treatment. These results suggest that FMGX is a promising candidate for the treatment of IAC.


Subject(s)
Antifungal Agents , Candidiasis, Invasive , Animals , Antifungal Agents/therapeutic use , Candidiasis, Invasive/drug therapy , Echinocandins , Micafungin , Mice , Microbial Sensitivity Tests
6.
Front Immunol ; 11: 891, 2020.
Article in English | MEDLINE | ID: mdl-32477361

ABSTRACT

In 2017 over 550,000 estimated new cases of multi-drug/rifampicin resistant tuberculosis (MDR/RR-TB) occurred, emphasizing a need for new treatment strategies. Linezolid (LZD) is a potent antibiotic for drug-resistant Gram-positive infections and is an effective treatment for TB. However, extended LZD use can lead to LZD-associated host toxicities, most commonly bone marrow suppression. LZD toxicities may be mediated by IL-1, an inflammatory pathway important for early immunity during M. tuberculosis infection. However, IL-1 can contribute to pathology and disease severity late in TB progression. Since IL-1 may contribute to LZD toxicity and does influence TB pathology, we targeted this pathway with a potential host-directed therapy (HDT). We hypothesized LZD efficacy could be enhanced by modulation of IL-1 pathway to reduce bone marrow toxicity and TB associated-inflammation. We used two animal models of TB to test our hypothesis, a TB-susceptible mouse model and clinically relevant cynomolgus macaques. Antagonizing IL-1 in mice with established infection reduced lung neutrophil numbers and partially restored the erythroid progenitor populations that are depleted by LZD. In macaques, we found no conclusive evidence of bone marrow suppression associated with LZD, indicating our treatment time may have been short enough to avoid the toxicities observed in humans. Though treatment was only 4 weeks (the FDA approved regimen at the time of study), we observed sterilization of the majority of granulomas regardless of co-administration of the FDA-approved IL-1 receptor antagonist (IL-1Rn), also known as Anakinra. However, total lung inflammation was significantly reduced in macaques treated with IL-1Rn and LZD compared to LZD alone. Importantly, IL-1Rn administration did not impair the host response against Mtb or LZD efficacy in either animal model. Together, our data support that inhibition of IL-1 in combination with LZD has potential to be an effective HDT for TB and the need for further research in this area.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Interleukin-1beta/antagonists & inhibitors , Linezolid/therapeutic use , Tuberculosis/drug therapy , Animals , Disease Models, Animal , Inflammation , Macaca , Male , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Mycobacterium tuberculosis/drug effects , Tuberculosis, Multidrug-Resistant/drug therapy
7.
Article in English | MEDLINE | ID: mdl-32041718

ABSTRACT

Doxycycline, an FDA-approved tetracycline, is used in tuberculosis in vivo models for the temporal control of mycobacterial gene expression. In these models, animals are infected with recombinant Mycobacterium tuberculosis carrying genes of interest under transcriptional control of the doxycycline-responsive TetR-tetO unit. To minimize fluctuations of plasma levels, doxycycline is usually administered in the diet. However, tissue penetration studies to identify the minimum doxycycline content in food achieving complete repression of TetR-controlled genes in tuberculosis (TB)-infected organs and lesions have not been conducted. Here, we first determined the tetracycline concentrations required to achieve silencing of M. tuberculosis target genes in vitro Next, we measured doxycycline concentrations in plasma, major organs, and lung lesions in TB-infected mice and rabbits and compared these values to silencing concentrations measured in vitro We found that 2,000 ppm doxycycline supplemented in mouse and rabbit feed is sufficient to reach target concentrations in TB lesions. In rabbit chow, the calcium content had to be reduced 5-fold to minimize chelation of doxycycline and deliver adequate oral bioavailability. Clearance kinetics from major organs and lung lesions revealed that doxycycline levels fall below concentrations that repress tet promoters within 7 to 14 days after doxycycline is removed from the diet. In summary, we have shown that 2,000 ppm doxycycline supplemented in standard mouse diet and in low-calcium rabbit diet delivers concentrations adequate to achieve full repression of tet promoters in infected tissues of mice and rabbits.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Doxycycline/pharmacokinetics , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , Tuberculosis/metabolism , Animal Feed , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/therapeutic use , Biological Availability , Calcium/pharmacology , Disease Models, Animal , Doxycycline/administration & dosage , Doxycycline/therapeutic use , Female , Gene Silencing , Lung/metabolism , Mice , Rabbits , Tetracycline Resistance , Tissue Distribution/genetics , Transgenes
8.
J Cyst Fibros ; 18(5): 714-720, 2019 09.
Article in English | MEDLINE | ID: mdl-31138497

ABSTRACT

BACKGROUND: Nontuberculous mycobacteria are recognized as a concern for cystic fibrosis (CF) patients due to increasing disease prevalence and the potential for detrimental effects on pulmonary function and mortality. Current standard of care involves prolonged systemic antibiotics, which often leads to severe side effects and poor treatment outcomes. In this study, we investigated the tolerability and efficacy of a novel inhaled therapeutic in various mouse models of NTM disease. METHODS: We developed clofazimine inhalation suspension (CIS), a novel formulation of clofazimine developed for inhaled administration. To determine the efficacy, minimum inhibitory concentrations were evaluated in vitro, and tolerability of CIS was determined in naïve mouse models over various durations. After establishing tolerability, CIS efficacy was tested in in vivo infection models of both Mycobacterium avium and M. abscessus. Lung and plasma clofazimine levels after chronic treatments were evaluated. RESULTS: Clofazimine inhalation suspension demonstrated antimycobacterial activity in vitro, with MIC values between 0.125 and 2 µg/ml for M. avium complex and M. abscessus. Administration into naïve mice showed that CIS was well tolerated at doses up to 28 mg/kg over 28 consecutive treatments. In vivo, CIS was shown to significantly improve bacterial elimination from the lungs of both acute and chronic NTM-infected mouse models compared to negative controls and oral clofazimine administration. Clofazimine concentrations in lung tissue were approximately four times higher than the concentrations achieved by oral dosing. CONCLUSION: Clofazimine inhalation suspension is a well tolerated and effective novel therapeutic candidate for the treatment of NTM infections in mouse models.


Subject(s)
Anti-Bacterial Agents , Clofazimine , Cystic Fibrosis , Mycobacterium Infections, Nontuberculous/drug therapy , Nontuberculous Mycobacteria/drug effects , Administration, Inhalation , Aerosols , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/pharmacokinetics , Biological Availability , Clofazimine/administration & dosage , Clofazimine/adverse effects , Clofazimine/pharmacokinetics , Cystic Fibrosis/drug therapy , Cystic Fibrosis/microbiology , Cystic Fibrosis/physiopathology , Mice , Microbial Sensitivity Tests/methods , Tissue Distribution , Treatment Outcome
9.
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
10.
Article in English | MEDLINE | ID: mdl-28739797

ABSTRACT

Intra-abdominal candidiasis (IAC) is a prominent invasive fungal infection associated with high mortality. Prompt antifungal therapy and source control are crucial for successful treatment. Echinocandin antifungal drugs are first-line agents; however, their clinical effectiveness is highly variable, with known potential for breakthrough resistance, and little is known about drug exposure at the site of infection. Using matrix-assisted desorption ionization mass spectrometry imaging technology, we investigated the spatial and quantitative distribution in tissue lesions for two echinocandin drugs, micafungin and CD101, in a clinically relevant IAC mouse model. Drug accumulation within lesions was observed with both drugs at their humanized therapeutic doses. CD101, but not micafungin, accumulated in lesions at levels above the mutant prevention concentration of the infecting strain. These findings indicate that current echinocandin drugs are limited by penetration at the site of infection and have implications for clinical outcomes and emergence of resistance in patients with IAC.


Subject(s)
Abdominal Abscess/drug therapy , Antifungal Agents/therapeutic use , Candida albicans/drug effects , Candidiasis/drug therapy , Echinocandins/pharmacokinetics , Lipopeptides/pharmacokinetics , Animals , Disease Models, Animal , Drug Resistance, Fungal/physiology , Echinocandins/therapeutic use , Female , Lipopeptides/therapeutic use , Micafungin , Mice , Microbial Sensitivity Tests , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
11.
Antimicrob Agents Chemother ; 60(7): 4197-205, 2016 07.
Article in English | MEDLINE | ID: mdl-27139472

ABSTRACT

Pyrazinamide (PZA) is a prodrug requiring conversion to pyrazinoic acid (POA) by an amidase encoded by pncA for in vitro activity. Mutation of pncA is the most common cause of PZA resistance in clinical isolates. To determine whether the systemic delivery of POA or host-mediated conversion of PZA to POA could circumvent such resistance, we evaluated the efficacy of orally administered and host-derived POA in vivo Dose-ranging plasma and intrapulmonary POA pharmacokinetics and the efficacy of oral POA or PZA treatment against PZA-susceptible tuberculosis were determined in BALB/c and C3HeB/FeJ mice. The activity of host-derived POA was assessed in rabbits infected with a pncA-null mutant and treated with PZA. Median plasma POA values for the area under the concentration-time curve from 0 h to infinity (AUC0-∞) were 139 to 222 µg·h/ml and 178 to 287 µg·h/ml after doses of PZA and POA of 150 mg/kg of body weight, respectively, in mice. Epithelial lining fluid POA concentrations in infected mice were comparable after POA and PZA administration. In chronically infected BALB/c mice, PZA at 150 mg/kg reduced lung CFU counts by >2 log10 after 4 weeks. POA was effective only at 450 mg/kg, which reduced lung CFU counts by ∼0.7 log10 POA had no demonstrable bactericidal activity in C3HeB/FeJ mice, nor did PZA administered to rabbits infected with a PZA-resistant mutant. Oral POA administration and host-mediated conversion of PZA to POA producing plasma POA exposures comparable to PZA administration was significantly less effective than PZA. These results suggest that the intrabacillary delivery of POA and that producing higher POA concentrations at the site of infection will be more effective strategies for maximizing POA efficacy.


Subject(s)
Antitubercular Agents/therapeutic use , Pyrazinamide/analogs & derivatives , Tuberculosis/drug therapy , Animals , Antitubercular Agents/pharmacokinetics , Female , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/pathogenicity , Pyrazinamide/pharmacokinetics , Pyrazinamide/therapeutic use , Rabbits
12.
J Chromatogr B Analyt Technol Biomed Life Sci ; 1009-1010: 138-43, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26730550

ABSTRACT

Moxifloxacin (MFX) and levofloxacin (LFX), class of fluoroquinolone antibiotics, are the two most prescribed drugs to multidrug resistant tuberculosis (MDR-TB) patients. A single, sensitive and reliable LC-ESI-MS/MS method was developed and validated to simultaneously quantitate the levels of these drugs in human serum where enrofloxacin (EFX) was used as internal standard (IS). Quantification was achieved by multiple reaction monitoring of selected mass transitions from precursor ions to product ions m/z 402.2→384.2 for MFX, 362.2→318.2 for LFX, and 362.1→318.3 for EFX. Calibration curves were plotted using concentrations ranging between 0.23-1000ng/mL for MFX and 0.13-1000ng/mL for LFX, and the correlation coefficients (r(2)) were in excess of 0.999. Intra- and inert-day accuracy was ranged between 92.1-104% with mean recoveries of 96% and 95.5% for MFX and LFX, respectively and precision was <9% at all quality control concentration levels. Matrix effect analysis showed extraction efficiency of 93.0-94.6% for MFX and 90.9-99.5% for LFX. Application of the developed method to real sample analysis resulted in efficient quantification of MFX and LFX in serum samples obtained from ten MDR-TB patients. The result indicated that the method could be applied as a potential drug monitoring tool to accurately analyze MFX and LFX within a short run time.


Subject(s)
Antitubercular Agents/blood , Chromatography, High Pressure Liquid/methods , Fluoroquinolones/blood , Levofloxacin/blood , Tandem Mass Spectrometry/methods , Tuberculosis, Multidrug-Resistant/drug therapy , Drug Monitoring/methods , Humans , Limit of Detection , Moxifloxacin , Tuberculosis, Multidrug-Resistant/blood
13.
Antimicrob Agents Chemother ; 60(2): 735-43, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26574016

ABSTRACT

Pyrazinamide (PZA) is one of only two sterilizing drugs in the first-line antituberculosis regimen. Its activity is strongly pH dependent; the MIC changes by several orders of magnitude over a range of pH values that may be encountered in various in vivo compartments. We recently reported selective inactivity of PZA in a subset of C3HeB/FeJ mice with large caseous lung lesions. In the present study, we evaluated whether such inactivity was explained by poor penetration of PZA into such lesions or selection of drug-resistant mutants. Despite demonstrating similar dose-proportional PZA exposures in plasma, epithelial lining fluid, and lung lesions, no dose response was observed in a subset of C3HeB/FeJ mice with the highest CFU burden. Although PZA-resistant mutants eventually replaced the susceptible bacilli in BALB/c mice and in C3HeB/FeJ mice with low total CFU burdens, they never exceeded 1% of the total population in nonresponding C3HeB/FeJ mice. The selective inactivity of PZA in large caseous lesions of C3HeB/FeJ mice is best explained by the neutral pH of liquefying caseum.


Subject(s)
Antitubercular Agents/therapeutic use , Drug Resistance, Bacterial/physiology , Mycobacterium tuberculosis/drug effects , Pyrazinamide/therapeutic use , Tuberculosis, Pulmonary/drug therapy , Animals , Disease Models, Animal , Genome, Bacterial/genetics , Hydrogen-Ion Concentration , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Microbial Sensitivity Tests , Mycobacterium tuberculosis/genetics
14.
Sci Transl Med ; 5(214): 214ra168, 2013 Dec 04.
Article in English | MEDLINE | ID: mdl-24307692

ABSTRACT

New chemotherapeutic compounds against multidrug-resistant Mycobacterium tuberculosis (Mtb) are urgently needed to combat drug resistance in tuberculosis (TB). We have identified and characterized the indolcarboxamides as a new class of antitubercular bactericidal agent. Genetic and lipid profiling studies identified the likely molecular target of indolcarboxamides as MmpL3, a transporter of trehalose monomycolate that is essential for mycobacterial cell wall biosynthesis. Two lead candidates, NITD-304 and NITD-349, showed potent activity against both drug-sensitive and multidrug-resistant clinical isolates of Mtb. Promising pharmacokinetic profiles of both compounds after oral dosing in several species enabled further evaluation for efficacy and safety. NITD-304 and NITD-349 were efficacious in treating both acute and chronic Mtb infections in mouse efficacy models. Furthermore, dosing of NITD-304 and NITD-349 for 2 weeks in exploratory rat toxicology studies revealed a promising safety margin. Finally, neither compound inhibited the activity of major cytochrome P-450 enzymes or the hERG (human ether-a-go-go related gene) channel. These results suggest that NITD-304 and NITD-349 should undergo further development as a potential treatment for multidrug-resistant TB.


Subject(s)
Antitubercular Agents/pharmacology , Indoles/pharmacology , Mycobacterium tuberculosis/drug effects , Tuberculosis, Multidrug-Resistant/drug therapy , Administration, Oral , Animals , Antitubercular Agents/administration & dosage , Antitubercular Agents/pharmacokinetics , Antitubercular Agents/toxicity , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Biological Availability , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Drug Resistance, Multiple, Bacterial/genetics , Humans , Indoles/administration & dosage , Indoles/pharmacokinetics , Indoles/toxicity , Injections, Intravenous , Membrane Transport Proteins/drug effects , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/metabolism , Rats , Rats, Wistar , Tuberculosis, Multidrug-Resistant/diagnosis , Tuberculosis, Multidrug-Resistant/microbiology
15.
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
16.
J Med Chem ; 54(16): 5639-59, 2011 Aug 25.
Article in English | MEDLINE | ID: mdl-21755942

ABSTRACT

The (S)-2-nitro-6-(4-(trifluoromethoxy)benzyloxy)-6,7-dihydro-5H-imidazo[2,1-b][1,3]oxazine named PA-824 (1) has demonstrated antitubercular activity in vitro and in animal models and is currently in clinical trials. We synthesized derivatives at three positions of the 4-(trifluoromethoxy)benzylamino tail, and these were tested for whole-cell activity against both replicating and nonreplicating Mycobacterium tuberculosis (Mtb). In addition, we determined their kinetic parameters as substrates of the deazaflavin-dependent nitroreductase (Ddn) from Mtb that reductively activates these pro-drugs. These studies yielded multiple compounds with 40 nM aerobic whole cell activity and 1.6 µM anaerobic whole cell activity: 10-fold improvements over both characteristics from the parent molecule. Some of these compounds exhibited enhanced solubility with acceptable stability to microsomal and in vivo metabolism. Analysis of the conformational preferences of these analogues using quantum chemistry suggests a preference for a pseudoequatorial orientation of the linker and lipophilic tail.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proteins/metabolism , Mycobacterium tuberculosis/drug effects , Nitroimidazoles/pharmacology , Nitroreductases/metabolism , Prodrugs/pharmacology , Animals , Antitubercular Agents/chemistry , Antitubercular Agents/pharmacokinetics , Drug Evaluation, Preclinical/methods , Humans , Kinetics , Metabolic Clearance Rate , Mice , Microbial Sensitivity Tests , Microsomes, Liver/metabolism , Molecular Structure , Mycobacterium tuberculosis/enzymology , Nitroimidazoles/chemistry , Nitroimidazoles/pharmacokinetics , Prodrugs/chemistry , Prodrugs/metabolism , Structure-Activity Relationship , Substrate Specificity , Tuberculosis/drug therapy , Tuberculosis/microbiology
17.
Antimicrob Agents Chemother ; 55(8): 3677-83, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21628535

ABSTRACT

Mycobacterium tuberculosis is the causative agent of a pulmonary epidemic that is estimated to infect one-third of the world's population and that has an increased incidence of multidrug resistance. The evaluation of new chemical entities against M. tuberculosis is hampered by the lack of biological tools to help predict efficacy, from early drug development to clinical trials. As the rat is the animal species of choice in the pharmaceutical industry, we have developed a rat model of acute and chronic phases of M. tuberculosis infection for drug efficacy testing. In this model, we have evaluated the impact of tuberculosis drugs on T cell response using the enzyme-linked immunospot assay methodology. Infected rats treated with isoniazid (INH) or rifampin (RIF) responded to therapy, the potency of which was comparable to that seen in the mouse. Peripheral blood mononuclear cells from infected rats produced gamma interferon (IFN-γ) in response to RD-1 antigens, such as the 6-kDa early secretory antigen target (ESAT-6) and the 10-kDa culture filtrate protein (CFP-10). A decrease in IFN-γ spot-forming cells (SFCs) was consistently observed in response to drug treatment. In both the acute- and chronic-phase models, the T cell response was more sensitive to ESAT-6 than to CFP-10. The SFC count in response to ESAT-6 appears to be an indicator of bacterial killing in the rat. Collectively, our data suggest that the ESAT-6 response could be used as a potential surrogate of drug efficacy in the rat and that such a readout could help shorten drug testing during preclinical development.


Subject(s)
Antigens, Bacterial/immunology , Antitubercular Agents/therapeutic use , Bacterial Proteins/immunology , Mycobacterium tuberculosis/drug effects , Rifampin/pharmacology , Tuberculosis/drug therapy , Tuberculosis/immunology , Animals , Antibiotics, Antitubercular/pharmacology , Antibiotics, Antitubercular/therapeutic use , Antitubercular Agents/pharmacology , Disease Models, Animal , Drug Discovery , Drug Evaluation, Preclinical , Drug Resistance, Multiple, Bacterial , Enzyme-Linked Immunospot Assay , Female , Interferon-gamma/biosynthesis , Isoniazid/pharmacology , Isoniazid/therapeutic use , Mycobacterium tuberculosis/immunology , Rats , Rats, Wistar , Rifampin/therapeutic use , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , T-Lymphocytes/microbiology , Tuberculosis/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL