Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Med Chem ; 54(12): 4219-33, 2011 Jun 23.
Article in English | MEDLINE | ID: mdl-21557540

ABSTRACT

Mining of an in-house collection of angiotensin II type 1 receptor antagonists to identify compounds with activity at the peroxisome proliferator-activated receptor-γ (PPARγ) revealed a new series of imidazo[4,5-b]pyridines 2 possessing activity at these two receptors. Early availability of the crystal structure of the lead compound 2a bound to the ligand binding domain of human PPARγ confirmed the mode of interaction of this scaffold to the nuclear receptor and assisted in the optimization of PPARγ activity. Among the new compounds, (S)-3-(5-(2-(1H-tetrazol-5-yl)phenyl)-2,3-dihydro-1H-inden-1-yl)-2-ethyl-5-isobutyl-7-methyl-3H-imidazo[4,5-b]pyridine (2l) was identified as a potent angiotensin II type I receptor blocker (IC(50) = 1.6 nM) with partial PPARγ agonism (EC(50) = 212 nM, 31% max) and oral bioavailability in rat. The dual pharmacology of 2l was demonstrated in animal models of hypertension (SHR) and insulin resistance (ZDF rat). In the SHR, 2l was highly efficacious in lowering blood pressure, while robust lowering of glucose and triglycerides was observed in the male ZDF rat.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/chemical synthesis , Antihypertensive Agents/chemical synthesis , Hypoglycemic Agents/chemical synthesis , Imidazoles/chemical synthesis , PPAR gamma/agonists , Pyridines/chemical synthesis , Administration, Oral , Angiotensin II Type 1 Receptor Blockers/chemistry , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Antihypertensive Agents/chemistry , Antihypertensive Agents/pharmacology , Biological Availability , Blood Glucose/analysis , Crystallography, X-Ray , Drug Partial Agonism , Humans , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Imidazoles/chemistry , Imidazoles/pharmacology , Insulin Resistance , Male , Models, Molecular , Pyridines/chemistry , Pyridines/pharmacology , Radioligand Assay , Rats , Rats, Inbred SHR , Stereoisomerism , Structure-Activity Relationship , Transcriptional Activation , Triglycerides/blood
2.
Chem Res Toxicol ; 23(3): 664-76, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20151638

ABSTRACT

Throughout the past decade, the expectations from the regulatory agencies for safety, drug-drug interactions (DDIs), pharmacokinetic, and disposition characterization of new chemical entities (NCEs) by pharmaceutical companies seeking registration have increased. DDIs are frequently assessed using in silico, in vitro, and in vivo methodologies. However, a key gap in this screening paradigm is a full structural understanding of time-dependent inhibition (TDI) on the cytochrome P450 systems, particularly P450 3A4. To address this, a number of high-throughput in vitro assays have been developed. This work describes an automated assay for TDI using two concentrations at two time points (2 + 2 assay). Data generated with this assay for over 2000 compounds from multiple therapeutic programs were used to generate in silico Bayesian classification models of P450 3A4-mediated TDI. These in silico models were validated using several external test sets and multiple random group testing (receiver operator curve value >0.847). We identified a number of substructures that were likely to elicit TDI, the majority containing indazole rings. These in vitro and in silico approaches have been implemented as a part of the Pfizer screening paradigm. The Bayesian models are available on the intranet to guide synthetic strategy, predict whether a NCE is likely to cause a TDI via P450 3A4, filter for in vitro testing, and identify substructures important for TDI as well as those that do not cause TDI. This represents an integrated in silico-in vitro strategy for addressing P450 3A4 TDI and improving the efficiency of screening.


Subject(s)
Cytochrome P-450 CYP3A Inhibitors , Cytochrome P-450 CYP3A/metabolism , Enzyme Inhibitors/pharmacology , Microsomes, Liver/metabolism , Enzyme Inhibitors/chemistry , Humans , Models, Biological , Time Factors
3.
Eur J Pharmacol ; 627(1-3): 16-25, 2010 Feb 10.
Article in English | MEDLINE | ID: mdl-19850035

ABSTRACT

Protein kinase C (PKC) family members such as PKCbetaII may become activated in the hyperglycemic state associated with diabetes. Preclinical and clinical data implicate aberrant PKC activity in the development of diabetic microvasculature abnormalities. Based on this potential etiological role for PKC in diabetic complications, several therapeutic PKC inhibitors have been investigated in clinical trials for the treatment of diabetic patients. In this report, we present the discovery and preclinical evaluation of a novel class of 3-amino-pyrrolo[3,4-c]pyrazole derivatives as inhibitors of PKC that are structurally distinct from the prototypical indolocarbazole and bisindolylmaleimide PKC inhibitors. From this pyrrolo-pyrazole series, several compounds were identified from biochemical assays as potent, ATP-competitive inhibitors of PKC activity with high specificity for PKC over other protein kinases. These compounds were also found to block PKC signaling activity in multiple cellular functional assays. PF-04577806, a representative from this series, inhibited PKC activity in retinal lysates from diabetic rats stimulated with phorbol myristate acetate. When orally administered, PF-04577806 showed good exposure in the retina of diabetic Long-Evans rats and ameliorated retinal vascular leakage in a streptozotocin-induced diabetic rat model. These novel PKC inhibitors represent a promising new class of targeted protein kinase inhibitors with potential as therapeutic agents for the treatment of patients with diabetic microvascular complications.


Subject(s)
Diabetes Complications/metabolism , Drug Discovery , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Retinal Diseases/metabolism , Retinal Vessels/drug effects , Signal Transduction/drug effects , Administration, Oral , Animals , Cattle , Cell Line , Diabetes Complications/drug therapy , Diabetes Complications/enzymology , Disease Models, Animal , Humans , Male , Protein Kinase C/metabolism , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Pyrazoles/administration & dosage , Pyrazoles/chemistry , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Rats , Retinal Diseases/drug therapy , Retinal Diseases/enzymology , Retinal Vessels/metabolism , Retinal Vessels/pathology , Substrate Specificity
4.
Bioorg Med Chem Lett ; 19(9): 2409-12, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19346128

ABSTRACT

The synthesis and SAR of tolylamines with 5-HT(6) receptor antagonist activity is presented. The amine, core aromatic, peripheral aromatic, and ether linker moieties of HTS hit 1 were modulated and the effect on potency at 5-HT(6) examined. Tolylpiperidine ether 9h was found to possess desirable pharmacokinetic (PK) properties, and was also shown to enhance cognition in the rat novel object recognition paradigm.


Subject(s)
Amines/chemistry , Receptors, Serotonin/chemistry , Serotonin Receptor Agonists/chemical synthesis , Animals , Chemistry, Organic/methods , Chemistry, Pharmaceutical/methods , Drug Design , Ethers/chemistry , Inhibitory Concentration 50 , Kinetics , Models, Chemical , Rats , Serotonin Receptor Agonists/chemistry , Serotonin Receptor Agonists/pharmacology , Structure-Activity Relationship
5.
J Pharm Sci ; 98(8): 2857-67, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19116953

ABSTRACT

As the cost of discovering and developing new pharmaceutically relevant compounds continues to rise, it is increasingly important to select the right molecules to prosecute very early in drug discovery. The development of high throughput in vitro assays of hepatic metabolic clearance has allowed for vast quantities of data generation; however, these large screens are still costly and remain dependant on animal usage. To further expand the value of these screens and ultimately aid in animal usage reduction, we have developed an in silico model of rat liver microsomal (RLM) clearance. This model combines a large amount of rat clearance data (n = 27,697) generated at multiple Pfizer laboratories to represent the broadest possible chemistry space. The model predicts RLM stability (with 82% accuracy and a kappa value of 0.65 for test data set) based solely on chemical structural inputs, and provides a clear assessment of confidence in the prediction. The current in silico model should help accelerate the drug discovery process by using confidence-based stability-driven prioritization, and reduce cost by filtering out the most unstable/undesirable molecules. The model can also increase efficiency in the evaluation of chemical series by optimizing iterative testing and promoting rational drug design.


Subject(s)
Computational Biology/methods , Computational Biology/standards , Microsomes, Liver/metabolism , Models, Biological , Animals , Metabolic Clearance Rate/drug effects , Predictive Value of Tests , Rats
6.
Expert Opin Drug Metab Toxicol ; 2(4): 591-608, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16859407

ABSTRACT

Optimisation of oral bioavailability is a continuing challenge for the pharmaceutical and biotechnology industries. The number of potential drug candidates requiring in vivo evaluation has significantly increased with the advent of combinatorial chemistry. In addition, drug discovery programmes are increasingly forced into more lipophilic and lower solubility chemical space. To aid in the use of in vitro and in silico tools as well as reduce the number of in vivo studies required, a team-based discussion tool is proposed that provides a 'road map' to guide the selection of profiling assays that should be considered when optimising oral bioavailability. This road map divides the factors that contribute to poor oral bioavailability into two interrelated categories: absorption and metabolism. This road map provides an interface for cross discipline discussions and a systematic approach to the experimentation that drives the drug discovery process towards a common goal - acceptable oral bioavailability using minimal resources in an acceptable time frame.


Subject(s)
Biological Availability , Drug Industry , Pharmaceutical Preparations/metabolism , Chemistry, Pharmaceutical , Hepatocytes/metabolism , Humans , Intestinal Absorption , Permeability , Pharmaceutical Preparations/administration & dosage , Solubility
7.
Int J Pharm ; 308(1-2): 133-9, 2006 Feb 03.
Article in English | MEDLINE | ID: mdl-16352407

ABSTRACT

PK express module is a physiologically based model of first pass metabolism, which integrates in vitro data with an in silico physiologically based pharmacokinetic (PBPK) model to predict human bioavailability (F(H)). There are three required inputs: FDp (Fraction dose absorbed, final parameter from iDEA absorption module), protein binding (fu) and disappearance kinetics in human hepatocytes. Caco-2 permeability, aqueous solubility (at multiple pH's), estimated dose and chemical structure are inputs required for the estimation of FDp (Norris et al., 2000; Stoner et al., 2004) and were determined for all compounds in our laboratory or obtained from literature. Protein binding data was collected from literature references and/or Pfizer database. Human hepatocyte data was generated in-house using an automated human hepatocyte method (using Tecan Genesis Workstation) as described previously (). Sixteen compounds (commercial and Pfizer compounds) were chosen to evaluate the PK express model and the bioavailability predicted from the module was compared with known clinical endpoints. For majority of the 16 compounds (approximately 80%), the PK express model F(H) values were comparable to the known human bioavailability (F(H)) (within 23.7 units of the known human (true) F, except for PF 3, PF 4, PF 6). In conclusion, the PK express model integrates a number of key readily available discovery parameters and provides estimates of human performance by integrating in silico and experimental variables built on a physiological based pharmacokinetic model. Information from this model in conjunction with other ADME data (e.g., P450 inhibition) will enable progression of most promising compounds for further in vivo PK and/or efficacy studies.


Subject(s)
Drug Evaluation, Preclinical/methods , Models, Biological , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Biological Availability , Caco-2 Cells , Hepatocytes/metabolism , Humans , Hydrogen-Ion Concentration , In Vitro Techniques , Intestinal Absorption , Intestinal Mucosa/metabolism , Pharmaceutical Preparations/chemistry , Protein Binding , Reproducibility of Results , Retrospective Studies , Solubility
8.
J Pharm Sci ; 93(5): 1131-41, 2004 May.
Article in English | MEDLINE | ID: mdl-15067690

ABSTRACT

The pharmaceutical industry has large investments in compound library enrichment, high throughput biological screening, and biopharmaceutical (ADME) screening. As the number of compounds submitted for in vitro ADME screens increases, data analysis, interpretation, and reporting will become rate limiting in providing ADME-structure-activity relationship information to guide the synthetic strategy for chemical series. To meet these challenges, a software tool was developed and implemented that enables scientists to explore in vitro and in silico ADME and chemistry data in a multidimensional framework. The present work integrates physicochemical and ADME data, encompassing results for Caco-2 permeability, human liver microsomal half-life, rat liver microsomal half-life, kinetic solubility, measured log P, rule of 5 descriptors (molecular weight, hydrogen bond acceptors, hydrogen bond donors, calculated log P), polar surface area, chemical stability, and CYP450 3A4 inhibition. To facilitate interpretation of this data, a semicustomized software solution using Spotfire was designed that allows for multidimensional data analysis and visualization. The solution also enables simultaneous viewing and export of chemical structures with the corresponding ADME properties, enabling a more facile analysis of ADME-structure-activity relationship. In vitro and in silico ADME data were generated for 358 compounds from a series of human immunodeficiency virus protease inhibitors, resulting in a data set of 5370 experimental values which were subsequently analyzed and visualized using the customized Spotfire application. Implementation of this analysis and visualization tool has accelerated the selection of molecules for further development based on optimum ADME characteristics, and provided medicinal chemistry with specific, data driven structural recommendations for improvements in the ADME profile.


Subject(s)
Chemistry, Pharmaceutical/methods , Drug Design , Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/metabolism , Absorption , Animals , Caco-2 Cells , Humans , Microsomes, Liver/metabolism , Rats , Software , Solubility , Tissue Distribution
9.
Int J Pharm ; 269(1): 241-9, 2004 Jan 09.
Article in English | MEDLINE | ID: mdl-14698595

ABSTRACT

The objective of the analysis described herein is to examine the in vitro/in vivo relationship of estimated bioavailability values and also the applicability of the estimated in vitro bioavailability to lead candidate selection in drug discovery. To this end, in vitro ADME data from screening assays as well as in vivo rat pharmacokinetic (PK) data were compiled for 140 compounds across therapeutic areas from the Pfizer library in Ann Arbor. The compounds span a broad range of structural types, including neutral, basic, and acidic compounds. Solubility and Caco-2 permeability data from in vitro ADME screening were used to calculate the fraction dose absorbed (FDp) using the physiologically based IDEA model. In vitro metabolic stability (t(1/2)) from human and rat liver microsomal incubations was converted to an in vitro intrinsic clearance value (CL(int)'), which was then scaled up to reflect in vivo clearance (CL) and hepatic extraction as described by Obach et al. [J. Pharmcol. Exp. Ther. 283 (1997) 46]. Subsequently, the in vitro/in vivo relationship between the measured bioavailability (F(obs)) in rats and the estimated bioavailability (F(est)) from FDp and predicted CL values was examined. The observed data suggest that compounds with low estimated in vitro bioavailability (F(est)<15%) are more likely to have low in vivo bioavailability (F(obs)<30%). Therefore, the present study indicates that in vitro estimation of bioavailability is an efficient tool to eliminate compounds having low bioavailability prior to in vivo characterization and therefore can be used to reduce attrition due to poor ADME properties in drug development.


Subject(s)
Models, Biological , Pharmacokinetics , Administration, Oral , Animals , Biological Availability , Caco-2 Cells , Drug Design , Half-Life , Humans , In Vitro Techniques , Microsomes, Liver/metabolism , Pharmaceutical Preparations/metabolism , Rats , Solubility , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...