Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 12 de 12
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Biopharm Drug Dispos ; 42(4): 137-149, 2021 Apr.
Article de Anglais | MEDLINE | ID: mdl-33354831

RÉSUMÉ

Transforming growth factor beta (TGF-ß) is a pleiotropic cytokine that has a wide array of biological effects. For decades, tumor biology implicated TGF-ß as an attractive therapeutic target due to its immunosuppressive effects. Toward this end, multiple pharmaceutical companies developed a number of drug modalities that specifically target the TGF-ß pathway. BMS-986260 is a small molecule, selective TGF-ßR1 kinase inhibitor that was under preclinical development for oncology. In vivo studies across mouse, rat, dog, and monkey and cryopreserved hepatocytes predicted human pharmacokinetics (PK) and distribution of BMS-986260. Efficacy studies of BMS-986260 were undertaken in the MC38 murine colon cancer model, and target engagement, as measured by phosphorylation of SMAD2/3, was assessed in whole blood to predict the clinical efficacious dose. The human clearance is predicted to be low, 4.25 ml/min/kg. BMS-986260 provided a durable and robust antitumor response at 3.75 mg/kg daily and 1.88 mg/kg twice-daily dosing regimens. Phosphorylation of SMAD2/3 was 3.5-fold less potent in human monocytes than other preclinical species. Taken together, the projected clinical efficacious dose was 600 mg QD or 210 mg BID for 3 days followed by a 4-day drug holiday. Mechanism-based cardiovascular findings in the rat ultimately led to the termination of BMS-986260. This study describes the preclinical PK characterization and pharmacodynamics-based efficacious dose projection of a novel small molecule TGF-ßR1 inhibitor.


Sujet(s)
Adénocarcinome/traitement médicamenteux , Tumeurs du côlon/traitement médicamenteux , Inhibiteurs de protéines kinases/administration et posologie , Récepteur de type I du facteur de croissance transformant bêta/antagonistes et inhibiteurs , Adénocarcinome/anatomopathologie , Animaux , Lignée cellulaire tumorale , Tumeurs du côlon/anatomopathologie , Chiens , Relation dose-effet des médicaments , Femelle , Hépatocytes/métabolisme , Humains , Macaca fascicularis , Mâle , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Tumeurs expérimentales/traitement médicamenteux , Tumeurs expérimentales/anatomopathologie , Inhibiteurs de protéines kinases/pharmacocinétique , Inhibiteurs de protéines kinases/pharmacologie , Rats , Rat Sprague-Dawley , Spécificité d'espèce , Distribution tissulaire
2.
J Med Chem ; 60(23): 9703-9723, 2017 12 14.
Article de Anglais | MEDLINE | ID: mdl-29077405

RÉSUMÉ

Factor XIa (FXIa) is a blood coagulation enzyme that is involved in the amplification of thrombin generation. Mounting evidence suggests that direct inhibition of FXIa can block pathologic thrombus formation while preserving normal hemostasis. Preclinical studies using a variety of approaches to reduce FXIa activity, including direct inhibitors of FXIa, have demonstrated good antithrombotic efficacy without increasing bleeding. On the basis of this potential, we targeted our efforts at identifying potent inhibitors of FXIa with a focus on discovering an acute antithrombotic agent for use in a hospital setting. Herein we describe the discovery of a potent FXIa clinical candidate, 55 (FXIa Ki = 0.7 nM), with excellent preclinical efficacy in thrombosis models and aqueous solubility suitable for intravenous administration. BMS-962212 is a reversible, direct, and highly selective small molecule inhibitor of FXIa.


Sujet(s)
Anticoagulants/composition chimique , Anticoagulants/usage thérapeutique , Facteur XIa/antagonistes et inhibiteurs , Isoquinoléines/composition chimique , Isoquinoléines/usage thérapeutique , Thrombose/traitement médicamenteux , para-Aminobenzoates/composition chimique , para-Aminobenzoates/usage thérapeutique , Animaux , Anticoagulants/pharmacocinétique , Anticoagulants/pharmacologie , Coagulation sanguine/effets des médicaments et des substances chimiques , Cristallographie aux rayons X , Chiens , Découverte de médicament , Facteur XIa/composition chimique , Facteur XIa/métabolisme , Humains , Isoquinoléines/pharmacocinétique , Isoquinoléines/pharmacologie , Mâle , Simulation de docking moléculaire , Lapins , Rats , Bibliothèques de petites molécules/composition chimique , Bibliothèques de petites molécules/pharmacocinétique , Bibliothèques de petites molécules/pharmacologie , Bibliothèques de petites molécules/usage thérapeutique , Thrombose/sang , para-Aminobenzoates/pharmacocinétique , para-Aminobenzoates/pharmacologie
3.
Bioorg Med Chem Lett ; 25(20): 4412-8, 2015 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-26386604

RÉSUMÉ

Non-basic azolotriazinones were explored using an empirical free brain exposures-driven approach to identify potent MCHR1 antagonists for evaluation in in vivo efficacy studies. An optimized lead from this series, 1j (rMCHR1 Ki=1.8 nM), demonstrated a 6.9% reduction in weight gain relative to vehicle in a rat model at 30 mg/kg after 4 days of once-daily oral treatment as a glycine prodrug. Despite a promising efficacy profile, an assessment of the biliary toxicity risk of this compound rendered this compound non-progressible.


Sujet(s)
Encéphale/effets des médicaments et des substances chimiques , Obésité/traitement médicamenteux , Récepteur somatostatine/antagonistes et inhibiteurs , Triazines/pharmacologie , Animaux , Encéphale/métabolisme , Relation dose-effet des médicaments , Humains , Structure moléculaire , Obésité/métabolisme , Rats , Relation structure-activité , Triazines/administration et posologie , Triazines/composition chimique
4.
Bioorg Med Chem Lett ; 25(14): 2793-9, 2015 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-26022839

RÉSUMÉ

Our investigation of the structure-activity and structure-liability relationships for dihydropyrrolopyrazol-6-one MCHR1 antagonists revealed that off-rate characteristics, inferred from potencies in a FLIPR assay following a 2 h incubation, can impact in vivo efficacy. The in vitro and exposure profiles of dihydropyrrolopyrazol-6-ones 1b and 1e were comparable to that of the thienopyrimidinone counterparts 41 and 43 except for a much faster MCHR1 apparent off-rate. The greatly diminished dihydropyrrolopyrazol-6-one anti-obesity response may be the consequence of this rapid off-rate.


Sujet(s)
Agents antiobésité/composition chimique , Pyrazoles/composition chimique , Récepteur somatostatine/antagonistes et inhibiteurs , Animaux , Agents antiobésité/pharmacocinétique , Agents antiobésité/pharmacologie , Agents antiobésité/usage thérapeutique , Période , Humains , Obésité/traitement médicamenteux , Liaison aux protéines , Pyrazoles/pharmacocinétique , Pyrazoles/pharmacologie , Pyrazoles/usage thérapeutique , Rats , Rat Sprague-Dawley , Récepteur somatostatine/métabolisme , Relation structure-activité , Perte de poids/effets des médicaments et des substances chimiques
5.
Pharm Res ; 32(8): 2625-35, 2015 Aug.
Article de Anglais | MEDLINE | ID: mdl-25670525

RÉSUMÉ

PURPOSE: An unknown UV 280 nm absorbing peak was observed by SEC for protein stability samples formulated in L-histidine during a stress stability study. Understanding the source would enhance the confidence in the SEC results. We identified the unknown peak, studied the cause, and evaluated ways to eliminate it. METHODS: The unknown peak was fractionated by preparative size exclusion chromatography separations, and subsequently analyzed by Hydrophilic Interaction Chromatography (HILIC) coupled with Time-of-Flight (TOF) high resolution mass spectrometry. The possible degradation was also studied with the presence of different excipients, including metal cations, chelating agents, and amino acids. RESULTS: The unknown peak was identified to be trans-urocanic acid, a degradant of histidine, based on evidences from HILIC retention time, UV profile, accurate mass measurement, trans-cis isomerization, and pI measurement. The degradation from histidine to urocanic acids was not affected by the presence of Fe(2+), but slightly activated by Mn(2+). The chelating agents, EDTA and DTPA, counteracted the Mn(2+) effects. This degradation was evidenced to be caused by contamination. Adding alanine or cysteine as an excipient was found to reduce this degradation by 97 and 98%, respectively. CONCLUSIONS: L-histidine formulation buffer can be contaminated to induce histidine degradation to trans-urocanic acid, which shows a large UV 280 nm absorbing peak at the total permeation volume under SEC conditions. Amino acids alanine and cysteine effectively inhibit this histidine degradation.


Sujet(s)
Produits biologiques/administration et posologie , Produits biologiques/composition chimique , Histidine/composition chimique , Acide urocanique/composition chimique , Substances tampon , Chélateurs/composition chimique , Chimie pharmaceutique , Chromatographie en phase liquide à haute performance , Contamination de médicament , Stabilité de médicament , Excipients/composition chimique , Spectrométrie de masse , Protéines/composition chimique , Spectrophotométrie UV
6.
J Med Chem ; 57(18): 7509-22, 2014 Sep 25.
Article de Anglais | MEDLINE | ID: mdl-25165888

RÉSUMÉ

Identification of MCHR1 antagonists with a preclinical safety profile to support clinical evaluation as antiobesity agents has been a challenge. Our finding that a basic moiety is not required for MCHR1 antagonists to achieve high affinity allowed us to explore structures less prone to off-target activities such as hERG inhibition. We report the SAR evolution of hydroxylated thienopyrimidinone ethers culminating in the identification of 27 (BMS-819881), which entered obesity clinical trials as the phosphate ester prodrug 35 (BMS-830216).


Sujet(s)
Agents antiobésité/pharmacologie , Découverte de médicament , Obésité/traitement médicamenteux , Récepteur somatostatine/antagonistes et inhibiteurs , Animaux , Agents antiobésité/pharmacocinétique , Agents antiobésité/usage thérapeutique , Chiens , Canal potassique ERG1 , Canaux potassiques éther-à-go-go/antagonistes et inhibiteurs , Humains , Mâle , Rats
7.
J Med Chem ; 56(23): 9586-600, 2013 Dec 12.
Article de Anglais | MEDLINE | ID: mdl-24182233

RÉSUMÉ

Several strategies have been employed to reduce the long in vivo half-life of our lead CB1 antagonist, triazolopyridazinone 3, to differentiate the pharmacokinetic profile versus the lead clinical compounds. An in vitro and in vivo clearance data set revealed a lack of correlation; however, when compounds with <5% free fraction were excluded, a more predictable correlation was observed. Compounds with log P between 3 and 4 were likely to have significant free fraction, so we designed compounds in this range to give more predictable clearance values. This strategy produced compounds with desirable in vivo half-lives, ultimately leading to the discovery of compound 46. The progression of compound 46 was halted due to the contemporaneous marketing and clinical withdrawal of other centrally acting CB1 antagonists; however, the design strategy successfully delivered a potent CB1 antagonist with the desired pharmacokinetic properties and a clean off-target profile.


Sujet(s)
Pyridazines/pharmacocinétique , Récepteur cannabinoïde de type CB1/antagonistes et inhibiteurs , Triazoles/pharmacocinétique , Animaux , Cytochrome P-450 enzyme system/métabolisme , Découverte de médicament , Période , Liaison aux protéines , Pyridazines/composition chimique , Rats , Relation structure-activité , Triazoles/composition chimique
8.
J Pharm Sci ; 102(8): 2424-39, 2013 Aug.
Article de Anglais | MEDLINE | ID: mdl-23712759

RÉSUMÉ

Aggregation propensity is a critical attribute of protein therapeutics that can influence production, manufacturing, delivery, and potential activity and safety (immunogenicity). It is therefore imperative to select molecules with low aggregation propensity in the early stages of drug discovery to mitigate the risk of delays or failure in clinical development. Although many biophysical methods have been developed to characterize protein aggregation, most established methods are low-throughput, requiring large quantities of protein, lengthy assay times, and/or significant upstream sample preparation, which can limit application in early candidate screening. To avoid these limitations, we developed a reliable method to characterize aggregation propensity, by measuring the relative solubility of protein therapeutic candidates in the presence of the kosmotropic salt ammonium sulfate. Manual bench-scale and automated plate-based methods were applied to different protein therapeutic formats including Adnectins, domain antibodies, PEGylated Adnectins, Fc fusion proteins, and monoclonal antibodies. The kosmotrope solubility data agreed well with the aggregation propensity observed by established methods, while being amenable to high-throughput screening because of speed, simplicity, versatility and low protein material requirements. The results suggest that kosmotrope-based solubility assessment has broad applicability to selecting protein therapeutic candidates with low aggregation propensity and high "developability" to progress into development.


Sujet(s)
Sulfate d'ammonium/composition chimique , Anticorps/composition chimique , Animaux , Lignée cellulaire , Découverte de médicament , Humains , Stabilité protéique , Protéines de fusion recombinantes/composition chimique , Solubilité
9.
Chem Res Toxicol ; 24(6): 905-12, 2011 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-21574629

RÉSUMÉ

The development of compounds with the potential for genotoxicity poses significant safety risks as well as risks of attrition. Although genotoxicity evaluation of the parent molecule is routine and reasonably predictive, assessing the risk of commercialization when release of a genotoxic degradant and/or metabolite from a nongenotoxic parent molecule is suspected is much more challenging and resource intensive. Much of the risk of the formation of a genotoxic degradant/metabolite can be discharged with the conduct of carcinogenicity studies in models where the compound is formed, but this approach requires a great deal of time and resources. In this manuscript, we investigated the contribution of various factors (pH, serum instability, and hepatic metabolism) to the formation of a mutagenic aromatic amine from a potent and highly selective thyromimetic compound ([3-(3,5-dibromo-4-(4-hydroxy-3-isopropyl-5-methylphenoxy)-2-methylphenylamino)-3-oxopropanoic acid], compound 1), under in vitro conditions. The kinetic parameters obtained from in vitro experiments combined with the pharmacokinetics of 1in vivo (e.g., plasma concentration-time profile and clearance) were used to estimate the extent of in vivo formation of [4-(4-amino-2,6-dibromo-3-methylphenoxy)-2-isopropyl-6-methylphenol] (compound 2), in rats upon administration of a single oral dose of 1. The agreement between the predicted values (1.9% conversion of total administered dose) with the observed levels of 2 in rats (0.2%-2.2% of the 10 mg/kg dose, 10 mg/kg) further prompted the utilization of this approach to predict the extent of release of this mutagen in humans upon administration of 1. The projection of 0.13% conversion to 2 from an efficacious daily dose of 15 mg of 1 translated to the generation of 20 µg of 2 and provided the basis for the decision to terminate the development of 1.


Sujet(s)
Amines/toxicité , Anilides/toxicité , Hydrocarbures aromatiques/toxicité , Malonates/toxicité , Mutagènes/toxicité , Hormones thyroïdiennes/toxicité , Amines/métabolisme , Anilides/sang , Anilides/métabolisme , Animaux , Chiens , Haplorhini , Humains , Hydrocarbures aromatiques/métabolisme , Concentration en ions d'hydrogène , Foie/métabolisme , Mâle , Malonates/sang , Malonates/métabolisme , Souris , Modèles biologiques , Tests de mutagénicité , Mutagènes/métabolisme , Rats , Rat Sprague-Dawley , Sérum/métabolisme , Hormones thyroïdiennes/sang , Hormones thyroïdiennes/métabolisme
10.
J Pharm Sci ; 95(1): 37-44, 2006 Jan.
Article de Anglais | MEDLINE | ID: mdl-16307454

RÉSUMÉ

A rabbit model for investigating sublingual drug absorption was established yielding results consistent with clinical data reported in the literature. Using propranolol as a model compound the effect of formulation and dosing variables was explored as a means to characterize the limiting parameters of this model. In addition, verapamil and captopril were selected as reference compounds to compare this model to sublingual absorption in humans. Rabbits were dosed sublingually and systemic absorption was measured over time. Sublingual absorption of propranolol was dependent on dosing solution pH and volume. Intra-oral spray device did not affect the overall exposure compared to instillation using a syringe. Despite species and dosing regimen differences the relative bioavailabilities of propranolol and verapamil were very similar in rabbits and humans. In contrast, captopril absorption from the sublingual cavity of rabbits was low and did not agree with that observed in man. Here we report a sublingual rabbit model of drug delivery and its potential utility in preclinical development of intra-oral dosage forms.


Sujet(s)
Captopril/pharmacocinétique , Propranolol/pharmacocinétique , Vérapamil/pharmacocinétique , Administration par voie sublinguale , Animaux , Captopril/administration et posologie , Captopril/sang , Chromatographie en phase liquide à haute performance , Humains , Concentration en ions d'hydrogène , Modèles animaux , Propranolol/administration et posologie , Propranolol/sang , Lapins , Vérapamil/administration et posologie , Vérapamil/sang
12.
J Drug Target ; 10(1): 31-40, 2002 Feb.
Article de Anglais | MEDLINE | ID: mdl-11996084

RÉSUMÉ

The goal of this study was to evaluate the permeability characteristics of Calu-3, human bronchial epithelial cells to passive and actively transported drugs and to correlate the data with other in vitro models and rat lung absorption in vivo. Air-interface cultured Calu-3 cells grown on collagen-coated permeable filter supports formed "tight" polarized and well differentiated cell monolayers with apical microvilli and tight-junctional complexes. Within 8-10 days, cell monolayers developed trans-epithelial electrical resistance (TEER) > 1000 ohm cm2 and potential difference about 11-16 mV. Solute permeability was dependent on lipophilicity, and inversely related to molecular size. Calu-3 cells actively transported amino acids, nucleosides and dipeptide analogs, but not organic anions, organic cations or efflux pump substrates. The permeability characteristics of Calu-3 cells correlated well with primary cultured rabbit tracheal epithelial cells in vitro (r2 = 0.91), and the rate of drug absorption from the rat lung in vivo (r2 = 0.94). The absorption predicted from the regression equation correlated well with observed values. In conclusion, in vitro-in vivo correlation studies indicate that the Calu-3 cell culture model is a potentially useful model to predict absorption of inhalation delivery drug candidates.


Sujet(s)
Bronches/cytologie , Perméabilité des membranes cellulaires/physiologie , Cellules épithéliales/physiologie , Poumon/métabolisme , Absorption , Administration par inhalation , Algorithmes , Animaux , Transport biologique , Bronches/physiologie , Bronches/ultrastructure , Lignée cellulaire , Cellules cultivées , Phénomènes chimiques , Chimie physique , Cellules épithéliales/ultrastructure , Période , Humains , Microscopie électronique , Techniques de patch-clamp , Préparations pharmaceutiques/composition chimique , Préparations pharmaceutiques/métabolisme , Rats
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE