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.
Drug Metab Lett ; 7(1): 15-22, 2013 Mar.
Article in English | MEDLINE | ID: mdl-24138031

ABSTRACT

PF-022 (1) is a novel polycyclic benzothiophene kinase inhibitor selective for mitogen-activated protein kinase-activated protein kinase 2 (MK2). Compound 1 emerged as an inhibitor bearing submicromolar potency against MK2 (IC50 5 nM) and demonstrated projected human pharmacokinetics sufficient for oral dosing. However, following a single, oral administration of 1 to beagle dogs, animals experienced an acute liver injury characterized by increases in biomarkers associated with hepatotoxicity; particularly noteworthy was the reversible elevation in bile salts and total bilirubin. Accompanying this observation was an ADME appraisal which included hepatic bioactivation of 1 in multiple species and the in vitro inhibition of P-glycoprotein (P-gp; IC50 21 µM). Simply attenuating the bioactivation via structural modification proved ineffective in improving the in vivo tolerability of this polycyclic scaffold. Hence, disruption of hepatobiliary transporters by the compound series was hypothesized as the likely mechanism contributing to the acute hepatotoxicity. Indeed, closer in vitro examination employing transporter gene overexpressing MDCK cell lines and membrane vesicles revealed potent compound-dependent inhibition of human multi-drug resistance-associated protein 2 (MRP2/ABCC2; IC50 38 µM) and bile salt export pump (BSEP/ABCB11; IC50 10 µM), two crucial hepatobiliary transport proteins accountable for bilirubin and bile salt homeostasis, respectively. Subsequent introduction of pKa-altering modifications to a second generation compound PF029 proved successful in reducing its affinity for these key efflux transporters (MRP2 IC50 >>80 µM; BSEP IC50 > 70 µM; P-gp > 90 µM), consequently mitigating this overt organ toxicity in dogs.


Subject(s)
Bile/metabolism , Carrier Proteins/antagonists & inhibitors , Chemical and Drug Induced Liver Injury/pathology , Liver/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/toxicity , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Area Under Curve , Cell Line , Dogs , Drug Resistance, Multiple , Fluoresceins/pharmacokinetics , Fluorescent Dyes , Male , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/metabolism , Taurocholic Acid/metabolism
2.
Toxicol Sci ; 108(2): 492-500, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19223659

ABSTRACT

CP-724,714, a potent and selective orally active HER2 tyrosine kinase inhibitor, was discontinued from clinical development due to unexpected hepatotoxicity in cancer patients. Based on the clinical manifestation of the toxicity, CP-724,714 likely exerted its hepatotoxicity via both hepatocellular injury and hepatobiliary cholestatic mechanisms. The direct cytotoxic effect, hepatobiliary disposition of CP-724,714, and its inhibition of active canalicular transport of bile constituents were evaluated in established human hepatocyte models and in vitro transporter systems. CP-724,714 exhibited direct cytotoxicity using human hepatocyte imaging assay technology with mitochondria identified as a candidate organelle for its off-target toxicity. Additionally, CP-724,714 was rapidly taken up into human hepatocytes, partially via an active transport process, with an uptake clearance approximately fourfold higher than efflux clearance. The major human hepatic uptake transporter, OATP1B1, and efflux transporters, multidrug resistance protein 1 (MDR1) and breast cancer resistance protein, were involved in hepatobiliary clearance of CP-724,714. Furthermore, CP-724,714 displayed a concentration-dependent inhibition of cholyl-lysyl fluorescein and taurocholate (TC) efflux into canaliculi in cryopreserved and fresh cultured human hepatocytes, respectively. Likewise, CP-724,714 inhibited TC transport in membrane vesicles expressing human bile salt export pump with an IC(50) of 16 microM. Finally, CP-724,714 inhibited the major efflux transporter in bile canaliculi, MDR1, with an IC(50) of approximately 28 microM. These results suggest that inhibition of hepatic efflux transporters contributed to hepatic accumulation of drug and bile constituents leading to hepatocellular injury and hepatobiliary cholestasis. This study provides likely explanations for clinically observed adverse liver effects of CP-724,714.


Subject(s)
Carrier Proteins/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/toxicity , Liver/metabolism , Quinazolines/pharmacokinetics , Quinazolines/toxicity , Receptor, ErbB-2/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/metabolism , Bile/metabolism , Bile Acids and Salts/metabolism , Calcium/metabolism , Cells, Cultured , Fluoresceins/metabolism , Hepatocytes/drug effects , Hepatocytes/enzymology , Humans , Liver-Specific Organic Anion Transporter 1/antagonists & inhibitors , Liver-Specific Organic Anion Transporter 1/metabolism , Membrane Potentials/drug effects , Mitochondrial Membranes/drug effects , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Multidrug Resistance-Associated Proteins/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism
3.
Drug Metab Dispos ; 36(2): 268-75, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17962372

ABSTRACT

Thirty-one structurally diverse marketed central nervous system (CNS)-active drugs, one active metabolite, and seven non-CNS-active compounds were tested in three P-glycoprotein (P-gp) in vitro assays: transwell assays using MDCK, human MDR1-MDCK, and mouse Mdr1a-MDCK cells, ATPase, and calcein AM inhibition. Additionally, the permeability for these compounds was measured in two in vitro models: parallel artificial membrane permeation assay and apical-to-basolateral apparent permeability in MDCK. The exposure of the same set of compounds in brain and plasma was measured in P-gp knockout (KO) and wild-type (WT) mice after subcutaneous administration. One drug and its metabolite, risperidone and 9-hydroxyrisperidone, of the 32 CNS compounds, and 6 of the 7 non-CNS drugs were determined to have positive efflux using ratio of ratios in MDR1-MDCK versus MDCK transwell assays. Data from transwell studies correlated well with the brain-to-plasma area under the curve ratios between P-gp KO and WT mice for the 32 CNS compounds. In addition, 3300 Pfizer compounds were tested in MDR1-MDCK and Mdr1a-MDCK transwell assays, with a good correlation (R(2) = 0.92) between the efflux ratios in human MDR1-MDCK and mouse Mdr1a-MDCK cells. Permeability data showed that the majority of the 32 CNS compounds have moderate to high passive permeability. This work has demonstrated that in vitro transporter assays help in understanding the role of P-gp-mediated efflux activity in determining the disposition of CNS drugs in vivo, and the transwell assay is a valuable in vitro assay to evaluate human P-gp interaction with compounds for assessing brain penetration of new chemical entities to treat CNS disorders.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Central Nervous System Agents/metabolism , Central Nervous System/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Adenosine Triphosphatases/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Fluoresceins/metabolism , Fluorescent Dyes/metabolism , Humans , Mice , Permeability , Pharmaceutical Preparations/metabolism , Phosphatidylcholines/metabolism
4.
Drug Metab Dispos ; 35(11): 2111-8, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17686907

ABSTRACT

The role of transporters in the disposition of (+)-2-[4-({[2-(benzo[1,3]dioxol-5-yloxy)-pyridine-3-carbonyl]-amino}-methyl)-3-fluoro-phenoxy]-propionic acid (CP-671,305), an orally active inhibitor of phosphodiesterase-4, was examined. In bile duct-exteriorized rats, a 7.4-fold decrease in the half-life of CP-671,305 was observed, implicating enterohepatic recirculation. Statistically significant differences in CP-671,305 pharmacokinetics (clearance and area under the curve) were discernible in cyclosporin A- or rifampicin-pretreated rats. Considering that cyclosporin A and rifampicin inhibit multiple uptake/efflux transporters, the interactions of CP-671,305 with major human hepatic drug transporters, multidrug resistance protein 1 (MDR1), multidrug resistance-associated protein 2 (MRP2), breast cancer resistant protein (BCRP), and organic anion-transporting polypeptide (OATPs) were evaluated in vitro. CP-671,305 was identified as a substrate of MRP2 and BCRP, but not MDR1. CP-671,305 was a substrate of human OATP2B1 with a high affinity (Km = 4 microM) but not a substrate for human OATP1B1 or OATP1B3. Consistent with these results, examination of hepatobiliary transport of CP-671,305 in hepatocytes indicated active uptake followed by efflux into bile canaliculi. Upon examination as a substrate for major rat hepatic Oatps, CP-671,305 displayed high affinity (Km = 12 microM) for Oatp1a4. The role of rat Mrp2 in the biliary excretion was also examined in Mrp2-deficient rats. The observations that CP-671,305 pharmacokinetics were largely unaltered suggested that compromised biliary clearance of CP-671,305 was compensated by increased urinary clearance. Overall, these studies suggest that hepatic transporters play an important role in the disposition and clearance of CP-671,305 in rat and human, and as such, these studies should aid in the design of clinical drug-drug interaction studies.


Subject(s)
Membrane Transport Proteins/metabolism , Phosphodiesterase 4 Inhibitors , Phosphodiesterase Inhibitors/pharmacokinetics , Propionates/pharmacokinetics , Pyridines/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Bile/metabolism , CHO Cells , Cell Line , Cricetinae , Cricetulus , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Male , Membrane Transport Proteins/genetics , Molecular Structure , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , Phosphodiesterase Inhibitors/chemistry , Phosphodiesterase Inhibitors/metabolism , Propionates/chemistry , Propionates/metabolism , Pyridines/chemistry , Pyridines/metabolism , Rats , Rats, Mutant Strains , Rats, Sprague-Dawley , Rats, Wistar , Transfection
5.
Toxicol Sci ; 90(2): 451-9, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16410371

ABSTRACT

Treatment with the antidepressant nefazodone has been associated with clinical idiosyncratic hepatotoxicty. Using membranes expressing human bile salt export pump (BSEP), human sandwich hepatocytes, and intact rats, we compared nefazodone and its marketed analogs, buspirone and trazodone. We found that nefazodone caused a strong inhibition of BSEP (IC(50) = 9 microM), inhibition of taurocholate efflux in human hepatocytes (IC(50) = 14 microM), and a transient increase in rat serum bile acids 1 h after oral drug administration. Buspirone or trazodone had no effect on biliary transport system. Nefazodone produced time- and concentration-dependent toxicity in human hepatocytes with IC(50) = 18 microM and 30 microM measured by inhibition of protein synthesis after 6 h and 24 h incubation, respectively. Toxicity was correlated with the amount of unmetabolized nefazodone. Partial recovery in toxicity by 24 h has been associated with metabolism of nefazodone to sulfate and glucuronide conjugates. The saturation of nefazodone metabolism resulted in sustained decrease in protein synthesis and cell death at 50 microM. The toxicity was not observed with buspirone or trazodone. Addition of 1-aminobenzotriazole (ABT), an inhibitor of CYP450, resulted in enhancement of nefazodone toxicity at 10 microM and was associated with accumulation of unmetabolized nefazodone. In human liver microsomes, ABT also prevented metabolism of nefazodone and formation of glutathione conjugates. We suggest that inhibition of bile acid transport by nefazodone is an indicator of potential hepatotoxicity. Our findings are consistent with the clinical experience and suggest that described methodology can be applied in the selection of nonhepatotoxic drug candidates.


Subject(s)
Antidepressive Agents, Second-Generation/toxicity , Bile Acids and Salts/metabolism , Biological Transport/drug effects , Triazoles/toxicity , Animals , Bile Acids and Salts/blood , Bile Canaliculi/drug effects , Bile Canaliculi/metabolism , Buspirone/toxicity , Cells, Cultured , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Piperazines , Rats , Rats, Sprague-Dawley , Trazodone/toxicity
6.
Chem Biol Interact ; 159(2): 156-68, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16356485

ABSTRACT

CI-1034, an endothelin-A receptor antagonist was being developed for pulmonary hypertension. Drug-drug interaction studies using human hepatic microsomes were conducted to assess CYP1A2, CYP2C9, CYP2C19, CYP3A4 and CYP2D6 inhibition potential; CYP3A4 induction potential was evaluated using primary human hepatocytes. CI-1034 moderately inhibited CYP2C9 (IC(50) 39.6 microM) and CYP3A4 activity (IC(50) 21.6 microM); CYP3A4 inhibition was metabolism-dependent. In human hepatocytes, no increase in CYP3A4 activity was observed in vitro, while mRNA was induced 15-fold, similar to rifampin, indicating that CI-1034 is both an inhibitor and inducer of CYP3A4. A 2-week clinical study was conducted to assess pharmacokinetics, pharmacodynamics and safety. No significant changes were observed in [formula: see text] between days 1 and 14. However, reversible elevations of serum liver enzymes were observed with a 50mg BID dose and the program was terminated. To further understand the interactions of CI-1034 in the liver and possible mechanisms of the observed hepatotoxicity, we evaluated the effect of CI-1034 on bile acid transport and previously reported that CI-1034 inhibited biliary efflux of taurocholate by 60%, in vitro. This indicated that inhibition of major hepatic transporters could be involved in the observed hepatotoxicity. We next evaluated the in vitro inhibition potential of CI-1034 with the major hepatic transporters OATP1B1, OATP1B3, OATP2B1, MDR1, MRP2 and OCT. CI-1034 inhibited OATP1B1 (K(i) 2 microM), OATP1B3 (K(i) 1.8 microM) and OATP2B1 activity (K(i) 3.3 microM) but not OCT, MDR1 or MRP2 mediated transport. Our data indicates that CI-1034 is an inhibitor of major hepatic transporters and inhibition of bile efflux may have contributed to the observed clinical hepatotoxicity. We recommend that in vitro drug-drug interaction panels include inhibition and induction studies with transporters and drug metabolizing enzymes, to more completely assess potential in vivo interactions or toxicity.


Subject(s)
Endothelin A Receptor Antagonists , Thiazines/pharmacology , Blotting, Western , Cells, Cultured , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/genetics , Double-Blind Method , Drug Interactions , Enzyme Inhibitors/pharmacology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Microsomes, Liver/metabolism , Organic Anion Transporters/metabolism , Placebos , RNA, Messenger/genetics , Thiazines/metabolism , Thiazines/pharmacokinetics
7.
Drug Metab Dispos ; 33(4): 537-46, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15616150

ABSTRACT

The present study examined the interaction of four 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (atorvastatin, lovastatin, and simvastatin in acid and lactone forms, and pravastatin in acid form only) with multidrug resistance gene 1 (MDR1, ABCB1) P-glycoprotein, multidrug resistance-associated protein 2 (MRP2, ABCC2), and organic anion-transporting polypeptide 1B1 (OATP1B1, SLCO21A6). P-glycoprotein substrate assays were performed using Madin-Darby canine kidney (MDCK) cells expressing MDR1, and the efflux ratios [the ratio of the ratio of basolateral-to-apical apparent permeability and apical-to-basolateral permeability between MDR1 and MDCK] were 1.87, 2.32/4.46, 2.17/3.17, and 0.93/2.00 for pravastatin, atorvastatin (lactone/acid), lovastatin (lactone/acid), and simvastatin (lactone/acid), respectively, indicating that these compounds are weak or moderate substrates of P-glycoprotein. In the inhibition assays (MDR1, MRP2, Mrp2, and OATP1B1), the IC50 values for efflux transporters (MDR1, MRP2, and Mrp2) were >100 microM for all statins in acid form except lovastatin acid (>33 microM), and the IC50 values were up to 10-fold lower for the corresponding lactone forms. In contrast, the IC50 values for the uptake transporter OATP1B1 were 3- to 7-fold lower for statins in the acid form compared with the corresponding lactone form. These data demonstrate that lactone and acid forms of statins exhibit differential substrate and inhibitor activities toward efflux and uptake transporters. The interconversion between the lactone and acid forms of most statins exists in the body and will potentially influence drug-transporter interactions, and may ultimately contribute to the differences in pharmacokinetic profiles observed between statins.


Subject(s)
Estradiol/analogs & derivatives , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Membrane Transport Proteins/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Organic Anion Transporters/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adenosine Triphosphate/metabolism , Animals , Atorvastatin , Cell Line , Dogs , Estradiol/metabolism , Fluoresceins/metabolism , Heptanoic Acids/chemistry , Heptanoic Acids/pharmacokinetics , Hydrolysis , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemistry , Lovastatin/chemistry , Lovastatin/pharmacokinetics , Multidrug Resistance-Associated Protein 2 , Permeability , Pyrroles/chemistry , Pyrroles/pharmacokinetics , Simvastatin/chemistry , Simvastatin/pharmacokinetics , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...