Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters











Database
Language
Publication year range
1.
Exp Biol Med (Maywood) ; 242(16): 1579-1585, 2017 10.
Article in English | MEDLINE | ID: mdl-28622731

ABSTRACT

Tissue chips are poised to deliver a paradigm shift in drug discovery. By emulating human physiology, these chips have the potential to increase the predictive power of preclinical modeling, which in turn will move the pharmaceutical industry closer to its aspiration of clinically relevant and ultimately animal-free drug discovery. Despite the tremendous science and innovation invested in these tissue chips, significant challenges remain to be addressed to enable their routine adoption into the industrial laboratory. This article describes the main steps that need to be taken and highlights key considerations in order to transform tissue chip technology from the hands of the innovators into those of the industrial scientists. Written by scientists from 13 pharmaceutical companies and partners at the National Institutes of Health, this article uniquely captures a consensus view on the progression strategy to facilitate and accelerate the adoption of this valuable technology. It concludes that success will be delivered by a partnership approach as well as a deep understanding of the context within which these chips will actually be used. Impact statement The rapid pace of scientific innovation in the tissue chip (TC) field requires a cohesive partnership between innovators and end users. Near term uptake of these human-relevant platforms will fill gaps in current capabilities for assessing important properties of disposition, efficacy and safety liabilities. Similarly, these platforms could support mechanistic studies which aim to resolve challenges later in development (e.g. assessing the human relevance of a liability identified in animal studies). Building confidence that novel capabilities of TCs can address real world challenges while they themselves are being developed will accelerate their application in the discovery and development of innovative medicines. This article outlines a strategic roadmap to unite innovators and end users thus making implementation smooth and rapid. With the collective contributions from multiple international pharmaceutical companies and partners at National Institutes of Health, this article should serve as an invaluable resource to the multi-disciplinary field of TC development.


Subject(s)
Drug Evaluation, Preclinical/methods , Microchip Analytical Procedures/methods , Microfluidics/methods , Drug Industry , Humans , Lab-On-A-Chip Devices
2.
Drug Metab Dispos ; 44(8): 1286-95, 2016 08.
Article in English | MEDLINE | ID: mdl-27298338

ABSTRACT

(R)-2-((2-(1H-pyrrolo[2,3-b]pyridin-3-yl)pyrimidin-4-yl)amino)-2-methyl-N-(2,2,2-trifluoroethyl)butanamide (VX-509, decernotinib) is an oral Janus kinase 3 inhibitor that has been studied in patients with rheumatoid arthritis. Patients with rheumatoid arthritis often receive multiple medications, such as statins and steroids, to manage the signs and symptoms of comorbidities, which increases the chances of drug-drug interactions (DDIs). Mechanism-based inhibition is a subset of time-dependent inhibition (TDI) and occurs when a molecule forms a reactive metabolite which irreversibly binds and inactivates drug-metabolizing enzymes, potentially increasing the systemic load to toxic concentrations. Traditionally, perpetrating compounds are screened using human liver microsomes (HLMs); however, this system may be inadequate when the precipitant is activated by a non-cytochrome P450 (P450)-mediated pathway. Even though studies assessing competitive inhibition and TDI using HLM suggested a low risk for CYP3A4-mediated DDI in the clinic, VX-509 increased the area under the curve of midazolam, atorvastatin, and methyl-prednisolone by approximately 12.0-, 2.7-, and 4.3-fold, respectively. Metabolite identification studies using human liver cytosol indicated that VX-509 is converted to an oxidative metabolite, which is the perpetrator of the DDIs observed in the clinic. As opposed to HLM, hepatocytes contain the full complement of drug-metabolizing enzymes and transporters and can be used to assess TDI arising from non-P450-mediated metabolic pathways. In the current study, we highlight the role of aldehyde oxidase in the formation of the hydroxyl-metabolite of VX-509, which is involved in clinically significant TDI-based DDIs and represents an additional example in which a system-dependent prediction of TDI would be evident.


Subject(s)
Aldehyde Oxidase/pharmacology , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Cytochrome P-450 CYP3A/metabolism , Heterocyclic Compounds, 2-Ring/pharmacology , Janus Kinase Inhibitors/pharmacology , Liver/enzymology , Microsomes, Liver/enzymology , Valine/analogs & derivatives , Adult , Aged , Aldehyde Oxidase/metabolism , Biotransformation , Cells, Cultured , Cytochrome P-450 CYP3A Inhibitors/metabolism , Cytochrome P-450 CYP3A Inhibitors/toxicity , Dose-Response Relationship, Drug , Drug Interactions , Female , Hepatocytes/enzymology , Heterocyclic Compounds, 2-Ring/metabolism , Heterocyclic Compounds, 2-Ring/toxicity , Humans , Hydroxylation , Janus Kinase Inhibitors/metabolism , Janus Kinase Inhibitors/toxicity , Kinetics , Male , Middle Aged , Risk Assessment , Valine/metabolism , Valine/pharmacology , Valine/toxicity , Young Adult
3.
J Med Chem ; 58(18): 7195-216, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26230873

ABSTRACT

While several therapeutic options exist, the need for more effective, safe, and convenient treatment for a variety of autoimmune diseases persists. Targeting the Janus tyrosine kinases (JAKs), which play essential roles in cell signaling responses and can contribute to aberrant immune function associated with disease, has emerged as a novel and attractive approach for the development of new autoimmune disease therapies. We screened our compound library against JAK3, a key signaling kinase in immune cells, and identified multiple scaffolds showing good inhibitory activity for this kinase. A particular scaffold of interest, the 1H-pyrrolo[2,3-b]pyridine series (7-azaindoles), was selected for further optimization in part on the basis of binding affinity (Ki) as well as on the basis of cellular potency. Optimization of this chemical series led to the identification of VX-509 (decernotinib), a novel, potent, and selective JAK3 inhibitor, which demonstrates good efficacy in vivo in the rat host versus graft model (HvG). On the basis of these findings, it appears that VX-509 offers potential for the treatment of a variety of autoimmune diseases.


Subject(s)
Autoimmune Diseases/drug therapy , Heterocyclic Compounds, 2-Ring/chemistry , Janus Kinase 3/antagonists & inhibitors , Valine/analogs & derivatives , Animals , Cell Line , Databases, Chemical , Dogs , Female , Graft vs Host Disease/drug therapy , Graft vs Host Disease/immunology , Haplorhini , Heterocyclic Compounds, 2-Ring/pharmacokinetics , Heterocyclic Compounds, 2-Ring/pharmacology , Humans , Janus Kinase 2/chemistry , Janus Kinase 3/chemistry , Male , Mice , Mice, Inbred BALB C , Mice, Inbred CBA , Microsomes, Liver/metabolism , Models, Molecular , Rats , Rats, Inbred Lew , Rats, Sprague-Dawley , Stereoisomerism , Structure-Activity Relationship , Valine/chemistry , Valine/pharmacokinetics , Valine/pharmacology
4.
Hepatology ; 60(6): 1826-37, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24811404

ABSTRACT

UNLABELLED: Understanding hepatitis C virus (HCV) replication has been limited by access to serial samples of liver, the primary site of viral replication. Our understanding of how HCV replicates and develops drug-resistant variants in the liver is limited. We studied 15 patients chronically infected with genotype 1 HCV treated with telaprevir (TVR)/pegylated-interferon alpha/ribavirin. Hepatic fine needle aspiration was performed before treatment and at hour 10, days 4 and 15, and week 8 after initiation of antiviral therapy. We measured viral kinetics, resistance patterns, TVR concentrations, and host transcription profiles. All patients completed all protocol-defined procedures that were generally well tolerated. First-phase HCV decline (baseline/treatment day 4) was significantly slower in liver than in plasma (slope plasma: -0.29; liver, -0.009; P < 0.001), whereas second-phase decline (posttreatment days 4-15) did not differ between the two body compartments (-0.11 and -0.15, respectively; P = 0.1). TVR-resistant variants were detected in plasma, but not in liver (where only wild-type virus was detected). Based upon nonstructural protein 3 sequence analysis, no compartmentalization of viral populations was observed between plasma and liver compartments. Gene expression profiling revealed strong tissue-specific expression signatures. Human intrahepatic TVR concentration, measured for the first time, was lower, compared to plasma, on a gram per milliliter basis. We found moderate heterogeneity between HCV RNA levels from different intrahepatic sites, indicating differences in hepatic microenvironments. CONCLUSION: These data support an integrated model for HCV replication wherein the host hepatic milieu and innate immunity control the level of viral replication, and the early antiviral response observed in the plasma is predominantly driven by inhibition of hepatic high-level HCV replication sites.


Subject(s)
Hepacivirus/drug effects , Hepatitis C, Chronic/drug therapy , Liver/virology , Oligopeptides/pharmacokinetics , RNA, Viral/blood , Adolescent , Adult , Aged , Biopsy, Fine-Needle , Drug Resistance, Viral , Female , Gene Expression , Hepacivirus/genetics , Hepatitis C, Chronic/metabolism , Humans , Liver/drug effects , Liver/metabolism , Male , Middle Aged , Models, Statistical , Oligopeptides/therapeutic use , Phylogeny , Treatment Outcome , Young Adult
5.
Drug Dev Ind Pharm ; 36(3): 315-22, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20170280

ABSTRACT

CONTEXT: VX-702 is a novel p38 mitogen-activated protein kinase inhibitor being developed to treat rheumatoid arthritis. OBJECTIVE: To characterize the renal excretion profile of VX-702 using the isolated perfused rat kidney (IPRK) model. METHODS: Studies were performed to assess the dose linearity of VX-702 excretion and to evaluate the effect of inhibitors of organic anion (probenecid) and organic cation (cimetidine) transport systems on VX-702 disposition. VX-702 excretion was studied over a range of doses targeting concentrations between 100 and 600 ng/mL. VX-702 (600 ng/mL) was also co-perfused with probenecid (1 mM) and cimetidine (2 mM). The results were compared to parallel experiments performed with methotrexate (MTX). RESULTS: VX-702 excretion was linear over the range of doses studied, and clearance data were consistent with net reabsorption by the kidney. Transport inhibition studies indicate that VX-702 is not a substrate for renal organic anion and organic cation transport systems. MTX (500 ng/mL) also displayed net reabsorption in the IPRK, but secretory transport was inhibited upon co-administration with probenecid. This finding is consistent with previous IPRK studies that demonstrated inhibitory effects of NSAIDS on MTX excretion. CONCLUSION: Overall, this study suggests that a renal drug-drug interaction between VX-702 and MTX would be unlikely if these medications were co-administered.


Subject(s)
Antirheumatic Agents/metabolism , Enzyme Inhibitors/metabolism , Kidney/metabolism , Methotrexate/metabolism , Phenylurea Compounds/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Algorithms , Animals , Cimetidine/pharmacology , Drug Interactions , Enzyme Inhibitors/analysis , Enzyme Inhibitors/chemistry , Glomerular Filtration Rate/drug effects , Immunologic Factors/metabolism , Kidney/drug effects , Kinetics , Male , Methotrexate/analysis , Methotrexate/chemistry , Organic Anion Transporters/antagonists & inhibitors , Organic Anion Transporters/physiology , Organic Cation Transport Proteins/antagonists & inhibitors , Organic Cation Transport Proteins/physiology , Osmolar Concentration , Perfusion , Phenylurea Compounds/analysis , Phenylurea Compounds/chemistry , Probenecid/pharmacology , Rats , Rats, Sprague-Dawley , Ultrafiltration
6.
J Pharm Sci ; 96(10): 2547-65, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17688284

ABSTRACT

A number of new molecular entities (NMEs) selected for full-scale development based on their safety and pharmacological data suffer from undesirable physicochemical and biopharmaceutical properties, which lead to poor pharmacokinetics and distribution after in vivo administration. An optimization of the preformulation studies to develop a dosage form with proper drug delivery system to achieve desirable pharmacokinetic and toxicological properties can aid in the accelerated development of these NMEs into therapies. Nanoparticulate drug delivery systems show a promising approach to obtain desirable druglike properties by altering the biopharmaceutics and pharmacokinetics properties of the molecule. Apart from the advantages of enhancing potential for systemic administration, nanoparticulate drug delivery systems can also be used for site-specific delivery, thus alleviating unwanted toxicity due to nonspecific distribution, improve patient compliance, and provide favorable clinical outcomes. This review summarizes some of the parameters and approaches that can be used to evaluate nanoparticulate drug delivery systems in early stages of formulation development.


Subject(s)
Drug Carriers , Drug Design , Nanotechnology , Pharmaceutical Preparations/chemistry , Polymers/chemistry , Technology, Pharmaceutical/methods , Animals , Chemistry, Pharmaceutical , Dendrimers , Drug Compounding , Gastrointestinal Tract/enzymology , Gastrointestinal Tract/metabolism , Humans , Intestinal Absorption , Lipids/chemistry , Liposomes , Micelles , Nanoparticles , Osmolar Concentration , Permeability , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Protein Binding , Solubility
7.
J Control Release ; 100(1): 135-44, 2004 Nov 05.
Article in English | MEDLINE | ID: mdl-15491817

ABSTRACT

Commercially available doxorubicin-loaded long-circulating liposomes (Doxil, Alza Pharmaceuticals) were modified with the monoclonal nucleosome (NS)-specific 2C5 antibody (mAb 2C5) that recognizes a broad variety of tumors via the tumor cell surface-bound NSs. For incorporation into liposomes, mAb 2C5 was modified with poly(ethylene glycol)-phosphatidyl ethanolamine conjugate (PEG-PE) with the free PEG terminus activated with the p-nitrophenylcarbonyl group (pNP-PEG-PE). Derivatives of mAb 2C5 containing a variable number of PEG-PE residues (10-32) per protein molecule were prepared with a reasonably good preservation of the antibody specific activity even at the highest degree of modification. PEG-PE-modified antibody quantitatively incorporated into the liposomal membrane of doxorubicin-loaded liposomes with a loss of not more than 20% of the encapsulated doxorubicin. 2C5-targeted Doxil liposomes acquired the ability to recognize NSs and specifically bind to various tumor cells. Doxorubicin-loaded long-circulating liposomes modified with the mAb 2C5 kill various tumor cells in vitro with the efficiency higher than non-targeted doxorubicin-loaded liposomes.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Neoplasm/administration & dosage , Doxorubicin/administration & dosage , Liposomes , Neoplasms/therapy , Nucleosomes/immunology , Animals , Cell Line, Tumor , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL