Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Thomayant Prueksaritanont is active.

Publication


Featured researches published by Thomayant Prueksaritanont.


Clinical Pharmacology & Therapeutics | 1992

Bioavailability of cyclosporine with concomitant rifampin administration is markedly less than predicted by hepatic enzyme induction.

Mary F. Hebert; John P. Roberts; Thomayant Prueksaritanont; Leslie Z. Benet

The pharmacokinetics of cyclosporine was studied in six healthy volunteers after administration of the drug orally (10 mg/kg) and intravenously (3 mg/kg) with and without concomitant rifampin administration. Both blood and plasma (separated at 37° C) samples were analyzed for cyclosporine concentration. For blood and plasma, respectively, clearances of cyclosporine were calculated to be 0.30 and 0.55 L/hr/kg, values for volume of distribution at steady state were 1.31 and 1.68 L/kg, and bioavailabilities were 27% and 33% during the pre‐rifampin phase. Post‐rifampin phase clearances of cyclosporine were 0.42 and 0.79 L/hr/kg, values for volume of distribution at steady state were 1.36 and 1.35 L/kg, and bioavailabilities were 10% and 9% for blood and plasma, respectively. Rifampin not only induces the hepatic metabolism of cyclosporine but also decreases its bioavailability to a greater extent than would be predicted by the increased metabolism. The decreased bioavailability most probably can be explained by an induction of intestinal cytochrome P450 enzymes, which appears to be markedly greater than the induction of hepatic metabolism.


Journal of Medicinal Chemistry | 2010

Discovery of the dual orexin receptor antagonist [(7R)-4-(5-chloro-1,3-benzoxazol-2-yl)-7-methyl-1,4-diazepan-1-yl][5-methyl-2-(2H-1,2,3-triazol-2-yl)phenyl]methanone (MK-4305) for the treatment of insomnia.

Christopher D. Cox; Michael J. Breslin; David B. Whitman; John D. Schreier; Georgia B. McGaughey; Michael J. Bogusky; Anthony J. Roecker; Swati P. Mercer; Rodney A. Bednar; Wei Lemaire; Joseph G. Bruno; Duane R. Reiss; C. Meacham Harrell; Kathy L. Murphy; Susan L. Garson; Scott M. Doran; Thomayant Prueksaritanont; Wayne B. Anderson; Cuyue Tang; Shane Roller; Tamara D. Cabalu; Donghui Cui; George D. Hartman; Steven D. Young; Ken S. Koblan; Christopher J. Winrow; John J. Renger; Paul J. Coleman

Despite increased understanding of the biological basis for sleep control in the brain, few novel mechanisms for the treatment of insomnia have been identified in recent years. One notable exception is inhibition of the excitatory neuropeptides orexins A and B by design of orexin receptor antagonists. Herein, we describe how efforts to understand the origin of poor oral pharmacokinetics in a leading HTS-derived diazepane orexin receptor antagonist led to the identification of compound 10 with a 7-methyl substitution on the diazepane core. Though 10 displayed good potency, improved pharmacokinetics, and excellent in vivo efficacy, it formed reactive metabolites in microsomal incubations. A mechanistic hypothesis coupled with an in vitro assay to assess bioactivation led to replacement of the fluoroquinazoline ring of 10 with a chlorobenzoxazole to provide 3 (MK-4305), a potent dual orexin receptor antagonist that is currently being tested in phase III clinical trials for the treatment of primary insomnia.


Journal of Medicinal Chemistry | 2008

Kinesin spindle protein (KSP) inhibitors. 9. Discovery of (2S)-4-(2,5-difluorophenyl)-n-[(3R,4S)-3-fluoro-1-methylpiperidin-4-yl]-2-(hydroxymethyl)-N-methyl-2-phenyl-2,5-dihydro-1H-pyrrole-1-carboxamide (MK-0731) for the treatment of taxane-refractory cancer.

Christopher D. Cox; Paul J. Coleman; Michael J. Breslin; David B. Whitman; Robert M. Garbaccio; Mark E. Fraley; Carolyn A. Buser; Eileen S. Walsh; Kelly Hamilton; Michael D. Schaber; Robert B. Lobell; Weikang Tao; Joseph P. Davide; Ronald E. Diehl; Marc Abrams; Vicki J. South; Hans E. Huber; Maricel Torrent; Thomayant Prueksaritanont; Chunze Li; Donald E. Slaughter; Elizabeth Mahan; Carmen Fernandez-Metzler; Youwei Yan; Lawrence C. Kuo; Nancy E. Kohl; George D. Hartman

Inhibition of kinesin spindle protein (KSP) is a novel mechanism for treatment of cancer with the potential to overcome limitations associated with currently employed cytotoxic agents. Herein, we describe a C2-hydroxymethyl dihydropyrrole KSP inhibitor ( 11) that circumvents hERG channel binding and poor in vivo potency, issues that limited earlier compounds from our program. However, introduction of the C2-hydroxymethyl group caused 11 to be a substrate for cellular efflux by P-glycoprotein (Pgp). Utilizing knowledge garnered from previous KSP inhibitors, we found that beta-fluorination modulated the p K a of the piperidine nitrogen and reduced Pgp efflux, but the resulting compound ( 14) generated a toxic metabolite in vivo. Incorporation of fluorine in a strategic, metabolically benign position by synthesis of an N-methyl-3-fluoro-4-(aminomethyl)piperidine urea led to compound 30 that has an optimal in vitro and metabolic profile. Compound 30 (MK-0731) was recently studied in a phase I clinical trial in patients with taxane-refractory solid tumors.


British Journal of Clinical Pharmacology | 1999

Metabolic interactions between mibefradil and HMG-CoA reductase inhibitors: an in vitro investigation with human liver preparations

Thomayant Prueksaritanont; Bennett Ma; Cuyue Tang; Yuan Meng; Carol Assang; Ping Lu; Paul J. Reider; Jiunn H. Lin; Thomas A. Baillie

AIMS To determine the effects of mibefradil on the nletabolism in human liver microsomal preparations of the HMG-CoA reductase inhibitors simvastatin, lovastatin, atorvastatin, cerivastatin and fluvastatin. METHODS Metabolism of the above five statins (0.5, 5 or 10 microM), as well as of specific CYP3A4/5 and CYP2C8/9 marker substrates, was examined in human liver microsomal preparations in the presence and absence of mibefradil (0.1-50 microM). RESULTS Mibefradil inhibited, in a concentration-dependent fashion, the metabolism of the four statins (simvastatin, lovastatin, atorvastatin and cerivastatin) known to be substrates for CYP3A. The potency of inhibition was such that the IC50 values (<1 microM) for inhibition of all of the CYP3A substrates fell within the therapeutic plasma concentrations of mibefradil, and was comparable with that of ketoconazole. However, the inhibition by mibefradil, unlike that of ketoconazole, was at least in part mechanism-based. Based on the kinetics of its inhibition of hepatic testosterone 6beta-hydroxylase activity, mibefradil was judged to be a powerful mechanism-based inhibitor of CYP3A4/5, with values for Kinactivation, Ki and partition ratio (moles of mibefradil metabolized per moles of enzyme inactivated) of 0.4 min(-1), 2.3 microM and 1.7, respectively. In contrast to the results with substrates of CYP3A, metabolism of fluvastatin, a substrate of CYP2C8/9, and the hydroxylation of tolbutamide, a functional probe for CYP2C8/9, were not inhibited by mibefradil. CONCLUSION Mibefradil, at therapeutically relevant concentrations, strongly suppressed the metabolism in human liver microsomes of simvastatin, lovastatin, atorvastatin and cerivastatin through its inhibitory effects on CYP3A4/5, while the effects of mibefradil on fluvastatin, a substrate for CYP2C8/9, were minimal in this system. Since mibefradil is a potent mechanism-based inhibitor of CYP3A4/5, it is anticipated that clinically significant drug-drug interactions will likely ensue when mibefradil is coadministered with agents which are cleared primarily by CYP3A-mediated pathways.


Drug Metabolism and Disposition | 2012

Lymphatic Transport and Catabolism of Therapeutic Proteins after Subcutaneous Administration to Rats and Dogs

Weirong Wang; Nancy Chen; Xiaolan Shen; Paul K. Cunningham; Scott E. Fauty; Kimberly Michel; Bo Wang; Xuening Hong; Christine Adreani; Christian N. Nunes; Chris V. Johnson; Kuo‐Chang Yin; Michelle Groff; Yan Zou; Liming Liu; Lora Hamuro; Thomayant Prueksaritanont

The mechanism underlying subcutaneous absorption of macromolecules and factors that can influence this process were studied in rats using PEGylated erythropoietins (EPOs) as model compounds. Using a thoracic lymph duct cannulation (LDC) model, we showed that PEGylated EPO was absorbed from the subcutaneous injection site mainly via the lymphatic system in rats, which is similar to previous reports in sheep. After subcutaneous administration, the serum exposure was reduced by ∼70% in LDC animals compared with that in the control animals, and most of the systemically available dose was recovered in the lymph. In both LDC and intact rats, the total radioactivity recoveries in excreta after subcutaneous administration were high (70–80%), indicating that catabolism, not poor absorption, was the main cause for the observed low bioavailability (30–40%). Moreover, catabolism of PEGylated EPO was found with both rat subcutaneous tissue homogenate and lymph node cell suspensions, and a significant amount of dose-related breakdown fragments was found in the lymph of LDC rats. In addition, the bioavailability of PEGylated EPOs was shown to be 2- to 4-fold lower in “fat rats,” indicating that physiologic features pertinent to lymphatic transport can have a profound impact on subcutaneous absorption. Limited studies in dogs also suggested similar subcutaneous absorption mechanisms. Collectively, our results suggest that the lymphatic absorption mechanism for macromolecules is probably conserved among commonly used preclinical species, e.g., rats and dogs, and that mechanistic understanding of the subcutaneous absorption mechanism and associated determinants should be helpful in biologic drug discovery and development.


Drug Metabolism and Disposition | 2006

In Vitro and in Vivo CYP3A64 Induction and Inhibition Studies in Rhesus Monkeys: A Preclinical Approach for CYP3A-Mediated Drug Interaction Studies

Thomayant Prueksaritanont; Yuhsin Kuo; Cuyue Tang; Chunze Li; Yue Qiu; Bing Lu; Kristie Strong-Basalyga; Karen Richards; Brian A. Carr; Jiunn H. Lin

In this study, induction and inhibition of rhesus monkey CYP3A64 versus human CYP3A4 were characterized in vitro, and the corresponding pharmacokinetic consequences were evaluated in rhesus monkeys. In monkey hepatocytes, rifampin markedly induced CYP3A64 mRNA (EC50 = 0.5 μM; Emax = 6-fold) and midazolam (MDZ) 1′-hydroxylase activity (EC50 = 0.2 μM; Emax = 2-fold). Compound A (N-[2(R)-hydroxy-1(S)-indanyl-5-[2(S)-(1,1-dimethylethylaminocarbonyl)-4-[(furo[2,3-b]pyridin-5-yl)-methyl]piperazin-1-yl]-4(S)-hydroxy-2(R)-phenylmethylpentanamide), a known potent and mechanism-based inhibitor of CYP3A4, strongly inhibited the formation of 1′-hydroxy MDZ by recombinant CYP3A64 in a concentration- and time-dependent manner (KI = 0.25 μM; kinact = 0.4 min–1). Similar corresponding results also were obtained with human CYP3A4 in the presence of rifampin or compound A. In rhesus monkeys, MDZ exhibited a relatively high metabolic clearance (primarily via 1′-hydroxylation followed by glucuronidation) and a low hepatic availability (Fh = 16%). Consistent with the induction of hepatic metabolism of a high-clearance compound, pretreatment with rifampin (18 mg/kg p.o. for 5 days) did not significantly affect the i.v. kinetics of MDZ, but caused a pronounced reduction (∼10-fold) in the systemic exposure to MDZ and, consequently, its Fh following intrahepatic portal vein administration (i.pv.) of MDZ. A comparable extent of the pharmacokinetic interaction also was obtained after a 1.8 mg/kg rifampin dose. Also consistent with the in vitro CYP3A64 inhibition finding, compound A (6 mg/kg i.v.) markedly increased (10-fold) the i.pv. administered MDZ exposure. At the doses studied, plasma concentrations of rifampin or compound A reached or exceeded their respective in vitro EC50 or KI values. These findings suggest the potential applicability of the in vitro-in vivo relationship approach in rhesus monkeys for studying CYP3A-mediated interactions in humans.


Journal of Biological Chemistry | 2010

Discovery of the Selective Androgen Receptor Modulator MK-0773 Using a Rational Development Strategy Based on Differential Transcriptional Requirements for Androgenic Anabolism Versus Reproductive Physiology

Azriel Schmidt; Donald B. Kimmel; Chang Bai; Angela Scafonas; SuJane Rutledge; Robert L. Vogel; Sheila McElwee-Witmer; Fang Chen; Pascale V. Nantermet; Viera Kasparcova; Chih-Tai Leu; Hai-Zhuan Zhang; Mark E. Duggan; Michael A. Gentile; Paul Hodor; Brenda Pennypacker; Patricia Masarachia; Evan E. Opas; Sharon Adamski; Tara E. Cusick; Jiabing Wang; Helen J. Mitchell; Yuntae Kim; Thomayant Prueksaritanont; James J. Perkins; Robert S. Meissner; George D. Hartman; Leonard P. Freedman; Shun-ichi Harada; William J. Ray

Selective androgen receptor modulators (SARMs) are androgen receptor (AR) ligands that induce anabolism while having reduced effects in reproductive tissues. In various experimental contexts SARMs fully activate, partially activate, or even antagonize the AR, but how these complex activities translate into tissue selectivity is not known. Here, we probed receptor function using >1000 synthetic AR ligands. These compounds produced a spectrum of activities in each assay ranging from 0 to 100% of maximal response. By testing different classes of compounds in ovariectomized rats, we established that ligands that transactivated a model promoter 40–80% of an agonist, recruited the coactivator GRIP-1 <15%, and stabilized the N-/C-terminal interdomain interaction <7% induced bone formation with reduced effects in the uterus and in sebaceous glands. Using these criteria, multiple SARMs were synthesized including MK-0773, a 4-aza-steroid that exhibited tissue selectivity in humans. Thus, AR activated to moderate levels due to reduced cofactor recruitment, and N-/C-terminal interactions produce a fully anabolic response, whereas more complete receptor activation is required for reproductive effects. This bimodal activation provides a molecular basis for the development of SARMs.


Pharmaceutical Research | 2011

Theoretical Analysis of Interplay of Therapeutic Protein Drug and Circulating Soluble Target: Temporal Profiles of ‘Free’ and ‘Total’ Drug and Target

Cuyue Tang; Thomayant Prueksaritanont

ABSTRACTPurposeTo systemically investigate, for a therapeutic protein with a circulating soluble target, how the interplay of target dynamics and drug pharmacokinetics defines the ‘total’ and ‘free’ drug and target temporal profiles.MethodBy extending the established rapid-binding target-mediated drug disposition (TMDD) pharmacokinetic model to circulating soluble targets, the temporal profiles of ‘total’ and ‘free’ drug and target were simulated with varying binding affinity (KD), target baseline (Rss), target turnover, and drug dose level. Two sets of published experimental data were compared with the simulated results.ResultsBinding to a circulating soluble target could lead to a divergence of the ‘free’ drug from the ‘total’ drug. Simulations show this divergent magnitude determined by KD and Rss, with the temporal profile being defined by target turnover and drug dose level. As divergence proceeds, starting at the distribution phase, ‘free’ drug would decline faster but eventually parallel ‘total’ drug at the terminal phase, giving rise to a steeper distribution phase and comparable terminal half-life, relative to the ’total’ form. The model also allows for estimation of the dynamic change of ‘total’ and ‘free’ target in response to the treatment of a therapeutic protein drug, facilitating dose level and regimen design to achieve desired ‘free’ target suppression. Model predictions compared favorably with two sets of published experimental data.ConclusionsTheoretical analyses identified key variables governing the different temporal profiles of ‘total’ and ‘free’ drug and target. The rapid-binding TMDD model reasonably captured the features of the interplay of drug pharmacokinetics and target dynamics for two reported cases.


Biochemical Pharmacology | 1992

Transport and metabolism of cyclosporine in isolated rat hepatocytes: The effects of lipids

Thomayant Prueksaritanont; Masahiro Koike; Betty-Ann Hoener; Leslie Z. Benet

The effects of lipids on the uptake and metabolism of cyclosporine (CyA) were investigated in isolated rat hepatocytes. In the absence of lipids, CyA was rapidly taken up (reaching apparent steady state within 5 min) and highly associated with the cells (more than 80%). The CyA uptake was concentration independent over the concentration range studied (0.6 to 11.2 micrograms/mL). Metabolism, however, was relatively slow and saturable. Except for cholesterol (at concentrations up to 15.5 mM), all lipids tested [oleic acid; low density lipoproteins (LDL); and high density lipoproteins (HDL)] reduced CyA cell uptake as well as its metabolism in a concentration-dependent manner. The effects of LDL were much more pronounced when compared to those of HDL and oleic acid. At an LDL concentration of 1 microM, drug uptake, indicated by the cell-associated concentration at steady state, was about 49% of the control value, while CyA metabolism was inhibited completely. Drug uptake of about 82 and 91% and CyA disappearance of 75 and 68% of the relevant control values were observed with HDL and oleic acid at concentrations of 10 microM and 0.7 mM, respectively. Apparently, lipids decreased CyA metabolism by reducing the concentration of CyA available for transport into the cells. These findings further support the suggestion of an important role for plasma lipids in the disposition of CyA.


The Journal of Clinical Pharmacology | 1994

Pharmacokinetics of orally and intravenously administered cyclosporine in pre-kidney transplant patients.

Francesco T. Aweeka; Stephen J. Tomlanovich; Thomayant Prueksaritanont; Suneel K. Gupta; Leslie Z. Benet

The pharmacokinetics of cyclosporine (CSA) and four metabolites were evaluated in eight hemodialysis subjects awaiting renal transplantation to compare metabolic patterns with those observed in post‐transplant patients and normal volunteers. Each subject received a single 4‐mg/kg intravenous and a single 10‐mg/kg oral dose separated by a 1‐week washout period. Blood samples were collected before and at .5, 1, 1.5, 2, 2.5, 3, 4, 6, 8, 10, 12, 14, and 24 hours after CSA dosing. Cyclosporine blood, plasma, and metabolite (M17, M1, M18, M21) levels were determined by high‐pressure liquid chromatography. Mean (±standard deviation) CSA blood clearance was .47 ± .15 L/hour/kg, steady‐state volume of distribution (Vss) was 1.9 ± .5 L/kg, and mean residence time (MRT) was 4.4 ± 1.8 hours after intravenous dosing. With plasma, mean clearance was .70 ± .31 L/hour/kg, Vss was 2.4 ±1.2 L/kg, and MRT was 3.7 ± 2.2 hours. Cyclosporine bioavailability (F) averaged 24 ± 11 and 24 ± 15%, using blood and plasma, respectively. Values for clearance and Vss were approximately 30 to 100% greater than comparable estimates in healthy volunteers, but F and MRT were not altered to this extent. These changes might be explained on the basis of decreased protein binding in uremic patients. The area under the curve ratio for M17 and M1 to CSA increased an average of 1.7‐ and 3.9‐fold, respectively, after oral dosing compared with intravenous administration, indicating increased conversion during first‐pass metabolism.

Collaboration


Dive into the Thomayant Prueksaritanont's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jiunn H. Lin

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

George D. Hartman

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kathy L. Murphy

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Lynn M. Gorham

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Douglas J. Pettibone

United States Military Academy

View shared research outputs
Researchain Logo
Decentralizing Knowledge