Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Toxicol Pathol ; 44(8): 1072-1083, 2016 12.
Article in English | MEDLINE | ID: mdl-27638646

ABSTRACT

There is a great need for improved diagnostic and prognostic accuracy of potential cardiac toxicity in drug development. This study reports the evaluation of several commercially available biomarker kits by 3 institutions (SRI, Eli Lilly, and Pfizer) for the discrimination between myocardial degeneration/necrosis and cardiac hypertrophy as well as the assessment of the interlaboratory and interplatform variation in results. Serum concentrations of natriuretic peptides (N-terminal pro-atrial natriuretic peptide [NT-proANP] and N-terminal pro-brain natriuretic peptide [NT-proBNP]), cardiac and skeletal troponins (cTnI, cTnT, and sTnI), myosin light chain 3 (Myl3), and fatty acid binding protein 3 (FABP3) were assessed in rats treated with minoxidil (MNX) and isoproterenol (ISO). MNX caused increased heart-to-body weight ratios and prominent elevations in NT-proANP and NT-proBNP concentrations detected at 24-hr postdose without elevation in troponins, Myl3, or FABP3 and with no abnormal histopathological findings. ISO caused ventricular leukocyte infiltration, myocyte fibrosis, and necrosis with increased concentrations of the natriuretic peptides, cardiac troponins, and Myl3. These results reinforce the advantages of a multimarker strategy in elucidating the underlying cause of cardiac insult and detecting myocardial tissue damage at 24-hr posttreatment. The interlaboratory and interplatform comparison analyses also showed that the data obtained from different laboratories and platforms are highly correlated and reproducible, making these biomarkers widely applicable in preclinical studies.


Subject(s)
Biomarkers/blood , Drug Discovery/standards , Drug-Related Side Effects and Adverse Reactions/blood , Heart/drug effects , Laboratories/standards , Animals , Cardiotoxicity , Drug Evaluation, Preclinical , Myocardium/metabolism , Myocardium/pathology , Rats
2.
mBio ; 6(6): e01741-15, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26530386

ABSTRACT

UNLABELLED: Linezolid is an oxazolidinone with potent activity against Mycobacterium tuberculosis. Linezolid toxicity in patients correlates with the dose and duration of therapy. These toxicities are attributable to the inhibition of mitochondrial protein synthesis. Clinically relevant linezolid regimens were simulated in the in vitro hollow-fiber infection model (HFIM) system to identify the linezolid therapies that minimize toxicity, maximize antibacterial activity, and prevent drug resistance. Linezolid inhibited mitochondrial proteins in an exposure-dependent manner, with toxicity being driven by trough concentrations. Once-daily linezolid killed M. tuberculosis in an exposure-dependent manner. Further, 300 mg linezolid given every 12 hours generated more bacterial kill but more toxicity than 600 mg linezolid given once daily. None of the regimens prevented linezolid resistance. These findings show that with linezolid monotherapy, a clear tradeoff exists between antibacterial activity and toxicity. By identifying the pharmacokinetic parameters linked with toxicity and antibacterial activity, these data can provide guidance for clinical trials evaluating linezolid in multidrug antituberculosis regimens. IMPORTANCE: The emergence and spread of multidrug-resistant M. tuberculosis are a major threat to global public health. Linezolid is an oxazolidinone that is licensed for human use and has demonstrated potent activity against multidrug-resistant M. tuberculosis. However, long-term use of linezolid has shown to be toxic in patients, often resulting in thrombocytopenia. We examined therapeutic linezolid regimens in an in vitro model to characterize the exposure-toxicity relationship. The antibacterial activity against M. tuberculosis was also assessed for these regimens, including the amplification or suppression of resistant mutant subpopulations by the chosen regimen. Higher exposures of linezolid resulted in greater antibacterial activity, but with more toxicity and, for some regimens, increased resistant mutant subpopulation amplification, illustrating the trade-off between activity and toxicity. These findings can provide valuable insight for designing optimal dosage regimens for linezolid that are part of the long combination courses used to treat multidrug-resistant M. tuberculosis.


Subject(s)
Antitubercular Agents/administration & dosage , Antitubercular Agents/adverse effects , Linezolid/administration & dosage , Linezolid/adverse effects , Mycobacterium tuberculosis/drug effects , Tuberculosis/drug therapy , Cell Line , Cell Survival , Drug Evaluation, Preclinical , Humans , Microbial Viability , Models, Biological
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.
Int J Toxicol ; 33(4): 282-287, 2014 07.
Article in English | MEDLINE | ID: mdl-24819520

ABSTRACT

N1-Benzylated dihydroquinolin-6-ols and their corresponding esters display exceptional activity against African trypanosomes in vitro, and administration of members of this class of compounds to trypanosome-infected mice results in cures in a first-stage African trypanosomiasis model. Since a quinone imine intermediate has been implicated in the antiparasitic mechanism of action of these compounds, evaluation of the hepatotoxic, mutagenic, and methemoglobin-promoting effects of these agents was performed. 1-Benzyl-1,2-dihydro-2,2,4-trimethylquinolin-6-ol hydrochloride and 1-benzyl-1,2-dihydro-2,2,4-trimethylquinolin-6-yl acetate showed outstanding in vitro selectivity for Trypanosoma brucei compared to the HepG2, Hep3B, Huh7, and PLC5 hepatocyte cell lines. 1-Benzyl-1,2-dihydro-2,2,4-trimethylquinolin-6-ol hydrochloride and 1-(2-methoxybenzyl)-1,2-dihydro-2,2,4-trimethylquinolin-6-yl acetate were not mutagenic when screened in the Ames assay, with or without metabolic activation. The latter 2 compounds promoted time- and dose-dependent formation of methemoglobin when incubated in whole human blood, but such levels were below those typically required to produce symptoms of methemoglobinemia in humans. Although compounds capable of quinone imine formation require careful evaluation, these in vitro studies indicate that antitrypanosomal dihydroquinolines merit further study as drug candidates against the neglected tropical disease human African trypanosomiasis.


Subject(s)
Acetates/adverse effects , Drugs, Investigational/adverse effects , Hepatocytes/drug effects , Methemoglobin/metabolism , Quinolines/adverse effects , Quinolinium Compounds/adverse effects , Trypanocidal Agents/adverse effects , Acetates/metabolism , Acetates/pharmacology , Activation, Metabolic , Animals , Cell Line , Cell Survival/drug effects , Drug Design , Drug Evaluation, Preclinical , Drugs, Investigational/chemical synthesis , Drugs, Investigational/metabolism , Drugs, Investigational/pharmacology , Hemoglobins/chemistry , Hemoglobins/metabolism , Hepatocytes/enzymology , Hepatocytes/metabolism , Humans , Inhibitory Concentration 50 , Kinetics , Methemoglobin/chemistry , Mutagenicity Tests , Oxidation-Reduction , Quinolines/chemical synthesis , Quinolines/metabolism , Quinolines/pharmacology , Quinolinium Compounds/metabolism , Quinolinium Compounds/pharmacology , Rats , Trypanocidal Agents/chemical synthesis , Trypanocidal Agents/metabolism , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/drug effects , Trypanosoma brucei brucei/growth & development
5.
J Med Chem ; 55(13): 6087-93, 2012 Jul 12.
Article in English | MEDLINE | ID: mdl-22708838

ABSTRACT

Previously reported studies identified analogues of propafenone that had potent antimalarial activity, reduced cardiac ion channel activity, and properties that suggested the potential for clinical development for malaria. Careful examination of the bioavailability, pharmacokinetics, toxicology, and efficacy of this series of compounds using rodent models revealed orally bioavailable compounds that are nontoxic and suppress parasitemia in vivo. Although these compounds possess potential for further preclinical development, they also carry some significant challenges.


Subject(s)
Antimalarials/chemistry , Antimalarials/pharmacokinetics , Malaria/drug therapy , Plasmodium berghei/drug effects , Propafenone/analogs & derivatives , Administration, Oral , Animals , Antimalarials/administration & dosage , Chloroquine/pharmacology , Cytochrome P-450 CYP2D6/metabolism , Cytochrome P-450 CYP2D6 Inhibitors , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Interactions , Female , HEK293 Cells , Hep G2 Cells , Humans , Mice , Mice, Inbred ICR , Microsomes, Liver/metabolism , Parasitemia/drug therapy , Structure-Activity Relationship
6.
J Med Chem ; 53(9): 3685-95, 2010 May 13.
Article in English | MEDLINE | ID: mdl-20361799

ABSTRACT

Among the known antimalarial drugs, chloroquine (CQ) and other 4-aminoquinolines have shown high potency and good bioavailability. Yet complications associated with drug resistance necessitate the discovery of effective new antimalarial agents. ADMET prediction studies were employed to evaluate a library of new molecules based on the 4-aminoquinolone-related structure of CQ. Extensive in vitro screening and in vivo pharmacokinetic studies in mice helped to identify two lead molecules, 18 and 4, with promising in vitro therapeutic efficacy, improved ADMET properties, low risk for drug-drug interactions, and desirable pharmacokinetic profiles. Both 18 and 4 are highly potent antimalarial compounds, with IC(50) values of 5.6 and 17.3 nM, respectively, against the W2 (CQ-resistant) strain of Plasmodium falciparum (for CQ, IC(50) = 382 nM). When tested in mice, these compounds were found to have biological half-lives and plasma exposure values similar to or higher than those of CQ; they are therefore desirable candidates to pursue in future clinical trials.


Subject(s)
Aminoquinolines/pharmacology , Antimalarials/chemistry , Aminoquinolines/therapeutic use , Animals , Antimalarials/pharmacology , Drug Evaluation, Preclinical , Half-Life , Mice , Pharmacokinetics , Plasmodium falciparum/drug effects , Small Molecule Libraries , Toxicology
7.
Proc Natl Acad Sci U S A ; 106(15): 6099-104, 2009 Apr 14.
Article in English | MEDLINE | ID: mdl-19332801

ABSTRACT

To prevent sexually transmitted HIV, the most desirable active ingredients of microbicides are antiretrovirals (ARVs) that directly target viral entry and avert infection at mucosal surfaces. However, most promising ARV entry inhibitors are biologicals, which are costly to manufacture and deliver to resource-poor areas where effective microbicides are urgently needed. Here, we report a manufacturing breakthrough for griffithsin (GRFT), one of the most potent HIV entry inhibitors. This red algal protein was produced in multigram quantities after extraction from Nicotiana benthamiana plants transduced with a tobacco mosaic virus vector expressing GRFT. Plant-produced GRFT (GRFT-P) was shown as active against HIV at picomolar concentrations, directly virucidal via binding to HIV envelope glycoproteins, and capable of blocking cell-to-cell HIV transmission. GRFT-P has broad-spectrum activity against HIV clades A, B, and C, with utility as a microbicide component for HIV prevention in established epidemics in sub-Saharan Africa, South Asia, China, and the industrialized West. Cognizant of the imperative that microbicides not induce epithelial damage or inflammatory responses, we also show that GRFT-P is nonirritating and noninflammatory in human cervical explants and in vivo in the rabbit vaginal irritation model. Moreover, GRFT-P is potently active in preventing infection of cervical explants by HIV-1 and has no mitogenic activity on cultured human lymphocytes.


Subject(s)
Algal Proteins/pharmacology , HIV Fusion Inhibitors/adverse effects , HIV Fusion Inhibitors/pharmacology , HIV-1/drug effects , Lectins/pharmacology , Algal Proteins/genetics , Algal Proteins/isolation & purification , Algal Proteins/metabolism , Animals , Cell Proliferation/drug effects , Cells, Cultured , Cervix Uteri/surgery , Cervix Uteri/virology , Cytokines/biosynthesis , Drug Evaluation, Preclinical , Female , HIV Envelope Protein gp120/metabolism , HIV Infections/prevention & control , HIV Infections/transmission , HIV Infections/virology , HIV-1/metabolism , Humans , Lectins/genetics , Lectins/isolation & purification , Lectins/metabolism , Plant Lectins , Protein Binding , Rabbits , Tissue Culture Techniques , Tissue Transplantation , Nicotiana/genetics , Nicotiana/metabolism
8.
Mol Imaging Biol ; 8(5): 284-91, 2006.
Article in English | MEDLINE | ID: mdl-16924428

ABSTRACT

[(18)F]1-(2-Fluoroethyl)-4-[(4-cyanophenoxy)methyl]piperidine ([(18)F]SFE) is a novel, selective, high-affinity sigma-1 receptor radioligand that has been preclinically well characterized in rodents. To support an investigational new drug (IND) application for the first evaluation of [(18)F]SFE in humans, single-organ and whole-body radiation adsorbed doses associated with [(18)F]SFE injection were estimated from rat distribution data. In addition, single- and multiple-dose toxicity studies were conducted in rabbits and in dogs. Multiple-dose toxicity studies in rabbits and single-dose toxicity studies in beagles suggest at least a 100-fold safety margin for humans studies at a mass dose limit of 4.0 mug per intravenous injection, based on the combined no observable adverse effect levels (NOAEL, mg/m(2)) measured in these species. Radiation dosimetry estimates obtained from rat biodistribution analyses of [(18)F]SFE suggest that most tissues would receive about 0.010-0.020 mGy/MBq, while the adrenal glands, brain, bone, liver, lungs, and spleen would receive slightly higher doses (0.024-0.044 mGy/MBq). The adrenal glands were identified as the critical organ, because they received the highest adsorbed radiation dose. The total exposure resulting from a 5 mCi administration of [(18)F]SFE is well below the FDA-defined limits for yearly cumulative and per-study exposures to research participants. These combined results support the expectation that [(18)F]SFE will be safe for use in human positron emission tomography (PET) imaging studies with the administration of 5 mCi and a mass dose equal to or less than 4.0 mug SFE per injection.


Subject(s)
Piperidines/administration & dosage , Piperidines/toxicity , Receptors, sigma/metabolism , Animals , Cardiovascular System/diagnostic imaging , Cardiovascular System/drug effects , Dogs , Drug Evaluation, Preclinical , Female , Male , Maximum Tolerated Dose , Piperidines/adverse effects , Positron-Emission Tomography/methods , Rabbits , Radiation Dosage , Radioactive Tracers , Tissue Distribution , Sigma-1 Receptor
SELECTION OF CITATIONS
SEARCH DETAIL