Dennis C. Dean
Merck & Co.
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Dennis C. Dean.
Science | 1996
Andrew D. Howard; Scott D. Feighner; Doris F. Cully; Joseph P. Arena; Paul A. Liberator; Charles Rosenblum; Michel Hamelin; Donna L. Hreniuk; Oksana C. Palyha; Jennifer Anderson; Philip S. Paress; Carmen Diaz; Michael Chou; Ken K. Liu; Karen Kulju McKee; Sheng-Shung Pong; Lee-Yuh Chaung; Alex Elbrecht; Mike Dashkevicz; Robert Heavens; M. Rigby; D.J.S. Sirinathsinghji; Dennis C. Dean; David G. Melillo; Arthur A. Patchett; Ravi P. Nargund; Patrick R. Griffin; Julie A. DeMartino; Sunil K. Gupta; James M. Schaeffer
Small synthetic molecules termed growth hormone secretagogues (GHSs) act on the pituitary gland and the hypothalamus to stimulate and amplify pulsatile growth hormone (GH) release. A heterotrimeric GTP-binding protein (G protein)-coupled receptor (GPC-R) of the pituitary and arcuate ventro-medial and infundibular hypothalamus of swine and humans was cloned and was shown to be the target of the GHSs. On the basis of its pharmacological and molecular characterization, this GPC-R defines a neuroendocrine pathway for the control of pulsatile GH release and supports the notion that the GHSs mimic an undiscovered hormone.
Journal of Pharmacology and Experimental Therapeutics | 2007
Xiaoyan Chu; Kelly Bleasby; Jocelyn Yabut; Xiaoxin Cai; Grace Chan; Michael J. Hafey; Shiyao Xu; Arthur J. Bergman; Matthew P. Braun; Dennis C. Dean; Raymond Evers
Sitagliptin, a selective dipeptidyl peptidase 4 inhibitor recently approved for the treatment of type 2 diabetes, is excreted into the urine via active tubular secretion and glomerular filtration in humans. In this report, we demonstrate that sitagliptin is transported by human organic anion transporter hOAT3 (Km = 162 μM), organic anion transporting polypeptide OATP4C1, and multidrug resistance (MDR) P-glycoprotein (Pgp), but not by human organic cation transporter 2 hOCT2, hOAT1, oligopeptide transporter hPEPT1, OATP2B1, and the multidrug resistance proteins MRP2 and MRP4. Our studies suggested that hOAT3, OATP4C1, and MDR1 Pgp might play a role in transporting sitagliptin into and out of renal proximal tubule cells, respectively. Sitagliptin did not inhibit hOAT1-mediated cidofovir uptake, but it showed weak inhibition of hOAT3-mediated cimetidine uptake (IC50 = 160 μM). hOAT3-mediated sitagliptin uptake was inhibited by probenecid, ibuprofen, furosemide, fenofibric acid, quinapril, indapamide, and cimetidine with IC50 values of 5.6, 3.7, 1.7, 2.2, 6.2, 11, and 79 μM, respectively. Sitagliptin did not inhibit Pgp-mediated transport of digoxin, verapamil, ritonavir, quinidine, and vinblastine. Cyclosporine A significantly inhibited Pgp-mediated transport of sitagliptin (IC50 = 1 μM). Our data indicate that sitagliptin is unlikely to be a perpetrator of drug-drug interactions with Pgp, hOAT1, or hOAT3 substrates at clinically relevant concentrations. Renal secretion of sitagliptin could be inhibited if coadministered with OAT3 inhibitors such as probenecid. However, the magnitude of interactions should be low, and the effects may not be clinically meaningful, due to the high safety margin of sitagliptin.
Bioorganic & Medicinal Chemistry Letters | 2008
Scott D. Edmondson; Lan Wei; Jinyou Xu; Jackie Shang; Shiyao Xu; Jianmei Pang; Ashok Chaudhary; Dennis C. Dean; Huaibing He; Barbara Leiting; Kathryn A. Lyons; Reshma A. Patel; Sangita B. Patel; Giovanna Scapin; Joseph K. Wu; Maria Beconi; Nancy A. Thornberry; Ann E. Weber
The synthesis, selectivity, rat pharmacokinetic profile, and drug metabolism profiles of a series of potent fluoroolefin-derived DPP-4 inhibitors (4) are reported. A radiolabeled fluoroolefin 33 was shown to possess a high propensity to form reactive metabolites, thus revealing a potential liability for this class of DPP-4 inhibitors.
Drug Metabolism and Disposition | 2010
Sanjeev Kumar; Eugene Y. Tan; Georgy Hartmann; Zachary Biddle; Arthur J. Bergman; James Dru; Jonathan Z. Ho; Allen N. Jones; Steve J. Staskiewicz; Matthew P. Braun; Bindhu V. Karanam; Dennis C. Dean; Isaias Noel Gendrano; Mark W. Graves; John A. Wagner; Rajesh Krishna
Anacetrapib is a novel cholesteryl ester transfer protein inhibitor being developed for the treatment of primary hypercholesterolemia and mixed dyslipidemia. The absorption, distribution, metabolism, and excretion of anacetrapib were investigated in an open-label study in which six healthy male subjects received a single oral dose of 150 mg and 165 μCi of [14C]anacetrapib. Plasma, urine, and fecal samples were collected at predetermined times for up to 14 days postdose and were analyzed for total radioactivity, the parent compound, and metabolites. The majority of the administered radioactivity (87%) was eliminated by fecal excretion, with negligible amounts present in urine (0.1%). The peak level of radioactivity in plasma (∼2 μM equivalents of [14C]anacetrapib) was achieved ∼4 h postdose. The parent compound was the major radioactive component (79–94% of total radioactivity) in both plasma and feces. Three oxidative metabolites, M1, M2, and M3, were detected in plasma and feces and were identified as the O-demethylated species (M1) and two secondary hydroxylated derivatives of M1 (M2 and M3). Each metabolite was detected at low levels, representing ≤14% of the radioactivity in plasma or fecal samples. In vitro data indicated that anacetrapib is metabolized mainly by CYP3A4 to form M1, M2, and M3. Overall, these data, along with those from other preclinical and clinical studies, indicate that anacetrapib probably exhibits a low-to-moderate degree of oral absorption in humans and the absorbed fraction of the dose is eliminated largely via CYP3A4-catalyzed oxidative metabolism, followed by excretion of metabolites by the biliary-fecal route.
Drug Metabolism and Disposition | 2010
Eugene Y. Tan; Georgy Hartmann; Qing Chen; Antonio Pereira; Scott M. Bradley; George A. Doss; Andy Shiqiang Zhang; Jonathan Z. Ho; Matthew P. Braun; Dennis C. Dean; Wei Tang; Sanjeev Kumar
The pharmacokinetics and metabolism of anacetrapib (MK-0859), a novel cholesteryl ester transfer protein inhibitor, were examined in rats and rhesus monkeys. Anacetrapib exhibited a low clearance in both species and a moderate oral bioavailability of ∼38% in rats and ∼13% in monkeys. The area under the plasma concentration-time curve in both species increased in a less than dose-proportional manner over an oral dose range of 1 to 500 mg/kg. After oral administration of [14C]anacetrapib at 10 mg/kg, ∼80 and 90% of the radioactive dose was recovered over 48 h postdose from rats and monkeys, respectively. The majority of the administered radioactive dose was excreted unchanged in feces in both species. Biliary excretion of radioactivity accounted for ∼15% and urinary excretion for less than 2% of the dose. Thirteen metabolites, resulting from oxidative and secondary glucuronic acid conjugation, were identified in rat and monkey bile. The main metabolic pathways consisted of O-demethylation (M1) and hydroxylation on the biphenyl moiety (M2) and hydroxylation on the isopropyl side chain (M3); these hydroxylations were followed by O-glucuronidation of these metabolites. A glutathione adduct (M9), an olefin metabolite (M10), and a propionic acid metabolite (M11) also were identified. In addition to parent anacetrapib, M1, M2, and M3 metabolites were detected in rat but not in monkey plasma. Overall, it appears that anacetrapib exhibits a low-to-moderate degree of absorption after oral dosing and majority of the absorbed dose is eliminated via oxidation to a series of hydroxylated metabolites that undergo conjugation with glucuronic acid before excretion into bile.
Drug Metabolism and Disposition | 2008
Ying Li; J. Greg Slatter; Zhoupeng Zhang; Yan Li; George A. Doss; Matthew P. Braun; Ralph A. Stearns; Dennis C. Dean; Thomas A. Baillie; Wei Tang
Recent clinical reports have suggested that the cyclooxygenase-2 inhibitor, lumiracoxib (Prexige), may cause a rare but serious hepatotoxicity in patients. In view of the close structural resemblance between lumiracoxib and diclofenac, a widely used nonsteroidal anti-inflammatory drug whose use also has been associated with rare cases of liver injury, it is possible that the toxicity of the two agents may share a common mechanism. Because it is believed that chemically reactive metabolites may play a role as mediators of diclofenac-mediated hepatotoxicity, the present in vitro study was carried out to test the hypothesis that lumiracoxib also undergoes metabolic activation when incubated with liver microsomal preparations and hepatocytes from rats and humans. By means of liquid chromatography tandem mass spectrometry and nuclear magnetic resonance spectrometry techniques, two previously unknown N-acetylcysteine (NAC) conjugates were identified, namely, 3′-NAC-4′-hydroxy lumiracoxib (M1) and 4′-hydroxy-6′-NAC-desfluoro lumiracoxib (M2), the structures of which reveal the intermediacy of an electrophilic quinone imine species. Based on the results of studies with immunoinhibitory antibodies, it was demonstrated that the formation of M1 and M2 in human liver microsomes was catalyzed by cytochrome P450 (P450) 2C9. These findings demonstrate that lumiracoxib is subject to P450-mediated bioactivation in both rat and human liver preparations, leading to the formation of a reactive intermediate analogous to species generated during the metabolism of diclofenac.
Journal of Labelled Compounds and Radiopharmaceuticals | 2000
Charles S. Elmore; Dennis C. Dean; David G. Melillo
A simple, efficient method for generation of 14CO from Ba14CO3 has been developed. Reduction of 14CO2 using LiBEt3H gave [14C] formate in good yield which was treated with conc. H2SO4 to effect dehydration to 14CO. Through direct attachment of a reaction vessel containing aryl substrate and Pd(0) catalyst,[14C]carbonylation reactions were performed without the use of a mercury transfer pump. [14C]Carbonylation reactions using 14CO generated in this manner have been shown to proceed in good yield with a variety of substrates. Copyright
Xenobiotica | 2008
Wei Tang; Ralph A. Stearns; Regina W. Wang; Randall R. Miller; Qing Chen; Jason S. Ngui; R. K. Bakshi; Ravi P. Nargund; Dennis C. Dean; Thomas A. Baillie
1-[(2R)-2-({[(1S,2S)-1-amino-1,2,3,4-tetrahydronaphthalen-2-yl]carbonyl}amino)-3-(4-chlorophenyl)propanoyl]-N-(tert-butyl)-4-cyclohexylpiperidine-4-carboxamide (1) is a potent melanocortin-4 receptor agonist that exhibited time-dependent inhibition of cytochrome P450 (P450) 3A in incubations with human liver microsomes. In incubations fortified with potassium cyanide, a cyano adduct was identified by liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis as a cyanonitrosotetrahydronaphthalenyl derivative. The detection of this adduct suggested that a nitroso species was involved in the formation of a metabolite intermediate (MI) complex that led to the observed P450 inactivation. Further evidence supporting this hypothesis derived from incubations of 1 with recombinant P450 3A4, which exhibited a λmax at approximately 450 nm. The species responsible for this absorbance required the presence of β-nicotinamide adenine dinucleotide phosphate reduced form (NADPH), increased with increasing incubation time and decreased following the addition of potassium ferricyanide to the incubation mixture, suggestive of an MI complex. Similar results were obtained with rat liver microsomes and with recombinant P450 3A1. When rats were dosed with indinavir as a P450 3A probe substrate, plasma exposure to indinavir increased three-fold following pretreatment with 1, consistent with drug–drug interaction projections based on the kinact and KI parameters for 1 in rat liver microsomes. A similar approach was used to predict the magnitude of the corresponding drug–drug interaction potential in humans dosed with a drug metabolized predominantly by P450 3A, and the forecast area under the curve (AUC) increase ranged from four- to ten-fold. These data prompted a decision to terminate further evaluation of 1 as a development candidate, and led to the synthesis of the methyl analogue 2. Methyl substitution α to the amino group in 2 was designed to reduce the propensity for formation of a nitroso intermediate and, indeed, 2 failed to exhibit time-dependent inhibition of P450 3A in human liver microsomal incubations. This case study highlights the importance of mechanistic studies in support of drug-discovery and decision-making processes.
Tetrahedron Letters | 1997
Dennis C. Dean; Michael A. Wallace; Tina M. Marks; David G. Melillo
Abstract Addition of one equivalent [14C]carbon dioxide to primary or secondary amines in the presence of ternary base, followed by reaction of the resulting [14C]carbamate salt with phosphorus oxychloride or thionyl chloride, represents a cost effective alternative to labeled phosgene.
Xenobiotica | 2008
Wei Tang; Ralph A. Stearns; Qing Chen; K. Bleasby; Y. Teffera; A. Colletti; M. Hafey; R. Evers; Dennis C. Dean; P. A. Magriotis; Thomas J. Lanza; Linus S. Lin; William K. Hagmann; T. A. Baillie
N-(1-(3,5-dichlorobenzenesulfonyl)-2S-methyl-azetidine-2-carbonyl)-L-4-(2′,6′-dimethoxyphenyl)phenylalanine (1) is a potent antagonist of the very late activating (VLA) antigen-4. During initial screening, 1 exhibited an apparent plasma clearance (CL) of 227 ml min−1 kg−1 in Sprague–Dawley rats following an intravenous bolus dose formulated in an aqueous solution containing 40% polyethylene glycol. Such a high CL value led to speculation that the elimination of compound 1 involved extra-hepatic tissues. However, the apparent plasma CL was reduced to 97 ml in−1 kg−1 when a 2-min time point was added to sample collections, and further decreased to 48 ml min−1 kg−1 after the dose was formulated in rat plasma. The lung extraction of 1 in rats was negligible whereas the hepatic extraction was ≥90%, based on comparison of area under the curve (AUC) values derived from intra-artery, intravenous, and portal vein administration. In rats dosed intravenously with [14C]-1, approximately 91% of the radioactivity was recovered in bile over 48 h, with 85% accounted for in the first 4-h samples. The biliary radioactivity profile consisted of approximately 30% intact parent compound, 20% 1-glucuronide, and 50% oxidation products resulting from O-demethylation or hydroxylation reactions. When incubated with rat liver microsomes, oxidative metabolism of 1 was inhibited completely by 1-aminobenzotriazole (ABT), whereas the oxidation and glucuronidation reactions were little affected in the presence of cyclosporin A (CsA). In contrast, the hepatic extraction of 1 in rats was unperturbed in animals pre-dosed with ABT, but was reduced approximately 60% following treatment with CsA. In vitro, 1 was a substrate of the rat organic anion transporter Oatp1b2, and its cellular uptake was inhibited by CsA. In addition, the hepatic extraction of 1 was approximately 30% lower in Eisai hyperbilirubinaemic rats which lack functional multidrug resistant protein-2 (MRP2). Collectively, these data suggest that the clearance of 1 in rats likely is a result of the combined processes of hepatic oxidation, glucuronidation and biliary excretion, all of which are facilitated by active hepatic uptake of parent compound and subsequent active efflux of both unchanged parent and its metabolites into bile. It was concluded, therefore, that multiple mechanisms contribute to the clearance of 1 in rats, and suggest that appropriate pharmacokinetic properties might be difficult to achieve for this class of compounds. This case study demonstrates that an integrated strategy, incorporating both rapid screening and mechanistic investigations, is of particular value in supporting drug discovery efforts and decision-making processes.