Network


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

Hotspot


Dive into the research topics where Jiunn H. Lin is active.

Publication


Featured researches published by Jiunn H. Lin.


Clinical Pharmacokinectics | 2003

Role of P-glycoprotein in pharmacokinetics: clinical implications.

Jiunn H. Lin; Masayo Yamazaki

P-glycoprotein, the most extensively studied ATP-binding cassette (ABC) transporter, functions as a biological barrier by extruding toxins and xenobiotics out of cells. In vitro and in vivo studies have demonstrated that P-glycoprotein plays a significant role in drug absorption and disposition. Because of its localisation, P-glycoprotein appears to have a greater impact on limiting cellular uptake of drugs from blood circulation into brain and from intestinal lumen into epithelial cells than on enhancing the excretion of drugs out of hepatocytes and renal tubules into the adjacent luminal space. However, the relative contribution of intestinal P-glycoprotein to overall drug absorption is unlikely to be quantitatively important unless a very small oral dose is given, or the dissolution and diffusion rates of the drug are very slow. This is because P-glycoprotein transport activity becomes saturated by high concentrations of drug in the intestinal lumen.Because of its importance in pharmacokinetics, P-glycoprotein transport screening has been incorporated into the drug discovery process, aided by the availability of transgenic mdr knockout mice and in vitro cell systems. When applying in vitro and in vivo screening models to study P-glycoprotein function, there are two fundamental questions: (i) can in vitro data be accurately extrapolated to the in vivo situation; and (ii) can animal data be directly scaled up to humans? Current information from our laboratory suggests that in vivo P-glycoprotein activity for a given drug can be extrapolated reasonably well from in vitro data. On the other hand, there are significant species differences in P-glycoprotein transport activity between humans and animals, and the species differences appear to be substrate-dependent.Inhibition and induction of P-glycoprotein have been reported as the causes of drug-drug interactions. The potential risk of P-glycoprotein-mediated drug interactions may be greatly underestimated if only plasma concentration is monitored. From animal studies, it is clear that P-glycoprotein inhibition always has a much greater impact on tissue distribution, particularly with regard to the brain, than on plasma concentrations. Therefore, the potential risk of P-glycoprotein-mediated drug interactions should be assessed carefully. Because of overlapping substrate specificity between cytochrome P450 (CYP) 3A4 and P-glycoprotein, and because of similarities in P-glycoprotein and CYP3A4 inhibitors and inducers, many drug interactions involve both P-glycoprotein and CYP3A4. Unless the relative contribution of P-glycoprotein and CYP3A4 to drug interactions can be quantitatively estimated, care should be taken when exploring the underlying mechanism of such interactions.


Pharmaceutical Research | 2006

CYP induction-mediated drug interactions : in vitro assessment and clinical implications

Jiunn H. Lin

Cytochrome P450 (CYP) induction-mediated interaction is one of the major concerns in clinical practice and for the pharmaceutical industry. There are two major issues associated with CYP induction: a reduction in therapeutic efficacy of comedications and an induction in reactive metabolite-induced toxicity. Because CYP induction is a metabolic liability in drug therapy, it is highly desirable to develop new drug candidates that are not potent CYP inducer to avoid the potential of CYP induction-mediated drug interactions. For this reason, today, many drug companies routinely include the assessment of CYP induction at the stage of drug discovery as part of the selection processes of new drug candidates for further clinical development. The purpose of this article is to review the molecular mechanisms of CYP induction and the clinical implications, including pharmacokinetic and pharmacodynamic consequences. In addition, factors that affect the degree of CYP induction and extrapolation of in vitro CYP induction data to in vivo situations will also be discussed. Finally, assessment of the potential of CYP induction at the drug discovery and development stage will be discussed.


Nature Medicine | 2000

A peptide–doxorubicin 'prodrug' activated by prostate-specific antigen selectively kills prostate tumor cells positive for prostate-specific antigen in vivo

Deborah Defeo-Jones; Victor M. Garsky; Bradley K. Wong; Dong-Mei Feng; Trina Bolyar; Kathleen M. Haskell; David M. Kiefer; Karen R. Leander; Elizabeth McAvoy; Patricia K. Lumma; Jenny Miu-Chun Wai; Edith T. Senderak; Sherri L. Motzel; Kevin P. Keenan; Matthew J. van Zwieten; Jiunn H. Lin; Roger M. Freidinger; Joel R. Huff; Allen Oliff; Raymond E. Jones

We covalently linked doxorubicin with a peptide that is hydrolyzable by prostate-specific antigen. In the presence of prostate tumor cells secreting prostate-specific antigen, the peptide moiety of this conjugate, L-377,202, was hydrolyzed, resulting in the release of leucine-doxorubicin and doxorubicin, which are both very cytotoxic to cancer cells. However, L-377,202 was much less cytotoxic than conventional doxorubicin to cells in culture that do not secrete prostate-specific antigen. L-377,202 was approximately 15 times more effective than was conventional doxorubicin at inhibiting the growth of human prostate cancer tumors in nude mice when both drugs were used at their maximally tolerated doses. Nude mice inoculated with human prostate tumor cells secreting prostate-specific antigen showed considerable reductions in tumor burden with minimal total body weight loss when treated with L-377,202. This improvement in therapeutic index correlated with the selective localization of leucine–doxorubicin and free doxorubicin in tissues secreting prostate-specific antigen after exposure to L-377,202.


Current Drug Metabolism | 2012

Enzyme kinetics of cytochrome P450-mediated reactions.

Magang Shou; Yuh Lin; Ping Lu; Cuyue Tang; Qin Mei; Dan Cui; Wei Tang; Jason S. Ngui; C. Charles Lin; Rominder Singh; Bradley K. Wong; James A. Yergey; Jiunn H. Lin; Paul G. Pearson; Thomas A. Baillie; A. David Rodrigues; Thomas H. Rushmore

The most common drug-drug interactions may be understood in terms of alterations of metabolism, associated primarily with changes in the activity of cytochrome P450 (CYP) enzymes. Kinetic parameters such as Km, Vmax, Ki and Ka, which describe metabolism-based drug interactions, are usually determined by appropriate kinetic models and may be used to predict the pharmacokinetic consequences of exposure to one or multiple drugs. According to classic Michaelis-Menten (M-M) kinetics, one binding site models can be employed to simply interpret inhibition (pure competitive, non-competitive and uncompetitive) or activation of the enzyme. However, some cytochromes P450, in particular CYP3A4, exhibit unusual kinetic characteristics. In this instance, the changes in apparent kinetic constants in the presence of inhibitor or activator or second substrate do not obey the rules of M-M kinetics, and the resulting kinetics are not straightforward and hamper mechanistic interpretation of the interaction in question. These unusual kinetics include substrate activation (autoactivation), substrate inhibition, partial inhibition, activation, differential kinetics and others. To address this problem, several kinetic models can be proposed, based upon the assumption that multiple substrate binding sites exist at the active site of a particular P450, and the resulting kinetic constants are, therefore, solved to adequately describe the observed interaction between multiple drugs. The following is an overview of some cytochrome P450-mediated classic and atypical enzyme kinetics, and the associated kinetic models. Applications of these kinetic models can provide some new insights into the mechanism of P450-mediated drug-drug interactions.


Current Drug Metabolism | 2002

Evaluation of Drug Interactions with P-Glycoprotein in Drug Discovery: In Vitro Assessment of the Potential for Drug-Drug Interactions with P-Glycoprotein

Jerome H. Hochman; Masayo Yamazaki; Tomoyuki Ohe; Jiunn H. Lin

The pharmacological effects of a drug are highly dependent on the absorption, metabolism, elimination, and distribution of the drug. In the past few years it has become apparent that transport proteins play a major role in regulating the distribution, elimination and metabolism of some drugs. As a consequence of our new understanding of the influence of transport proteins on the pharmacokinetic and pharmacodynamic behavior of drugs, increasing attention has been focused on the potential for drug-drug interactions arising from interactions with drug transport proteins. The efflux transporter P-glycoprotein (P-gp) has received the most attention with regard to its role in restricting drug absorption and distribution and as a potential source for variability in drug pharmacokinetics and pharmacodynamics. This review will focus on the evaluation of drug candidates to assess the potential for drug interactions at the level of P-gp. We will discuss the role of P-gp in drug disposition, the biochemistry of P-gp efflux as it relates to model systems to study drug interactions with P-gp, and the implementation of P-gp assay models within the drug discovery process.


Pharmaceutical Research | 2005

Interconversion Pharmacokinetics of Simvastatin and its Hydroxy Acid in Dogs: Effects of Gemfibrozil

Thomayant Prueksaritanont; Yue Qiu; Lillian Mu; Kimberly Michel; Janice Brunner; Karen Richards; Jiunn H. Lin

PurposeTo characterize the pharmacokinetics of simvastatin (SV) and simvastatin acid (SVA), a lactone–acid pair known to undergo reversible metabolism, and to better understand mechanisms underlying pharmacokinetic interactions observed between SV and gemfibrozil.MethodsPharmacokinetic studies were conducted after intravenous administration of SV and SVA to dogs pretreated with a vehicle or gemfibrozil. In vitro metabolism of SVA in dog hepatocytes as well as in vitro hepatic and plasma conversion of SV/SVA were investigated in the absence and presence of gemfibrozil.ResultsIn control animals, the irreversible elimination clearances of SV (CL10) and SVA (CL20) were 10.5 and 18.6xa0ml min−1 kg−1, respectively. The formation clearance of SVA from SV (CL12 = 4.8xa0ml min−1 kg−1) was 8-fold greater than that of SV from SVA (CL21 = 0.6xa0ml min−1 kg−1), and the recycled fraction was relatively minor (0.009). In gemfibrozil-treated animals, CL10 was essentially unchanged, whereas CL12, CL20, CL21, and recycled fraction were significantly decreased to 2.9, 9, 0.14xa0ml min−1 kg−1, and 0.003, respectively. In control dogs, values for real volume of distribution at steady state (Vss,real) of SV (2.3xa0L kg−1) were much larger than the corresponding values of SVA (0.3xa0L kg−1). Gemfibrozil treatment did not affect Vss,real of either SV or SVA. In dog hepatocytes, gemfibrozil modestly affected the formation of CYP3A-mediated oxidative metabolites (IC50 > 200xa0μM) and β-oxidative products (IC50 ∼100xa0μM), but markedly inhibited the glucuronidation-mediated lactonization of SVA and the glucuronidation of an SVA β-oxidation product (IC50 = 18xa0μM). In in vitro dog and human liver S9 and plasma, hydrolysis of SV to SVA was much faster than that of SVA to SV. Gemfibrozil (250xa0μM) had a minimal inhibitory effect on the hydrolysis of either SV to SVA or SVA to SV in dog and human liver S9, but had a significant (∼60%) inhibitory effect on the SV to SVA hydrolysis in both dog and human plasma.ConclusionsIn dogs, the interconversion process favored the formation of SVA and was less efficient than the irreversible elimination processes of SV and SVA. Treatment with gemfibrozil did not affect the distribution of SV/SVA, but rather affected the elimination of SVA and the SV/SVA interconversion processes. Gemfibrozil decreased CL20 and CL21 likely via its inhibitory effect on the glucuronidation of SVA, and not on the CYP3A-mediated oxidative metabolism of SV or SVA, the β-oxidation of SVA, nor the SVA to SV hydrolysis. The decrease in CL12 might be due in part to the inhibitory effect of gemfibrozil on SV to SVA hydrolysis in plasma. Similar rationales may also be applicable to studies in humans and/or other statin lactone–acid pairs.


British Journal of Clinical Pharmacology | 1999

Metabolic interactions between mibefradil and HMG-CoA reductase inhibitors

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

AIMSnTo 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.nnnMETHODSnMetabolism 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).nnnRESULTSnMibefradil 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.nnnCONCLUSIONnMibefradil, 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 | 2002

Glucuronidation of Statins in Animals and Humans: A Novel Mechanism of Statin Lactonization

Thomayant Prueksaritanont; Raju Subramanian; Xiaojun Fang; Bennett Ma; Yue Qiu; Jiunn H. Lin; Paul G. Pearson; Thomas A. Baillie


Journal of Pharmacology and Experimental Therapeutics | 2002

Mechanistic studies on metabolic interactions between gemfibrozil and statins

Thomayant Prueksaritanont; Jamie J. Zhao; Bennett Ma; Brad Roadcap; Cuyue Tang; Yue Qiu; Lida Liu; Jiunn H. Lin; Paul G. Pearson; Thomas A. Baillie


Journal of Pharmacology and Experimental Therapeutics | 2001

In Vitro Substrate Identification Studies for P-glycoprotein-Mediated Transport: Species Difference and Predictability of in Vivo Results

Masayo Yamazaki; William E. Neway; Tomoyuki Ohe; I-Wu Chen; Janice Rowe; Jerome H. Hochman; Masato Chiba; Jiunn H. Lin

Collaboration


Dive into the Jiunn H. Lin's collaboration.

Researchain Logo
Decentralizing Knowledge