Network


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

Hotspot


Dive into the research topics where David M. Stresser is active.

Publication


Featured researches published by David M. Stresser.


Journal of Pharmacological and Toxicological Methods | 2000

Fluorometric screening for metabolism-based drug--drug interactions.

Charles L. Crespi; David M. Stresser

Inhibition of cytochromes P-450 (CYP) is a principal mechanism for metabolism-based drug interactions. In vitro methods for quantitatively measuring the extent of CYP inhibition are well-established. Classical methods use drug molecules as substrates and HPLC-based analysis. However, methodologies, which do not require HPLC separations for data acquisition generally offer higher throughputs and lower costs. Multiwell plate-based, direct, fluorometric assays for the activities of the five principal drug-metabolizing enzymes are available and parameters for the use of these substrates to measure CYP inhibition have been established. This methodology is quantitative, rapid, reproducible, and compatible with common high throughput screening instrumentation. This article describes approaches to establishing this methodology in a drug-discovery support program.


Annals of the New York Academy of Sciences | 2006

Fluorometric High-Throughput Screening for Inhibitors of Cytochrome P450

Vaughn P. Miller; David M. Stresser; Andrew P. Blanchard; Stephanie D. Turner; Charles L. Crespi

Abstract: Rapid screening for cytochrome P450 inhibitors is part of the current paradigm for avoiding development of drugs likely to give clinical pharmacokinetic drug‐drug interactions and associated toxicities. We have developed microtiter plate‐based, direct, fluorometric assays for the activities of the principal human drug‐metabolizing enzymes, CYP1A2, CYP2C8, CYP2C9, CYP2C19, CYP2D6, and CYP3A4, as well as for CYP2A6, which is an important enzyme in environmental toxicology. These assays are rapid and compatible with existing high‐throughput assay instrumentation. For CYP1A2, CYP2C8, CYP2C9, CYP2C19, and CYP2D6, the potency of enzyme inhibition (IC50) is consistent regardless of the probe substrate or assay method employed. In contrast, CYP3A4 inhibition for an individual inhibitor shows significant differences in potency (>300‐fold) depending on the probe substrate being used. We have investigated these differences through the use of several structurally distinct fluorescent substrates for CYP3A4 and several classical substrate probes (e.g., testosterone, nifedipine, and midazolam), with a panel of known, clinically significant, CYP3A4 inhibitors. The use of multiple probe substrates appears to be needed to characterize the inhibition potential of xenobiotics for CYP3A4.


Drug Metabolism and Disposition | 2009

Evaluation of multiple in vitro systems for assessment of CYP3A4 induction in drug discovery: human hepatocytes, pregnane X receptor reporter gene, and Fa2N-4 and HepaRG cells.

Dermot F. McGinnity; George Zhang; Jane R. Kenny; Geraldine A. Hamilton; Sara Otmani; Karen R. Stams; Suzzette Haney; Patrick Brassil; David M. Stresser; Robert J. Riley

Prototypic CYP3A4 inducers were tested in a pregnane X receptor (PXR) reporter gene assay, Fa2N-4 cells, HepaRG cells, and primary human hepatocytes, along with negative controls, using CYP3A4 mRNA and activity endpoints, where appropriate. Over half of the compounds tested (14 of 24) were identified as time-dependent inhibitors of CYP3A4 and high mRNA/activity ratios (>10) were consistent with CYP3A4 time-dependent inhibition for compounds such as troleandomycin, ritonavir, and verapamil. Induction response was compared between two human donors; there was an excellent correlation in the EC50 estimates (r2 = 0.89, p < 0.001), and a weak but statistically significant correlation was noted for maximum observed induction at an optimum concentration (Emax) (r2 = 0.38, p = 0.001). Emax and EC50 estimates determined from the PXR reporter gene assay and Fa2N-4 and HepaRG cells were compared with those from hepatocytes. Overall, EC50 values generated using hepatocytes agreed with those generated in the PXR reporter gene assay (r2 = 0.85, p < 0.001) and Fa2N-4 (r2 = 0.65, p < 0.001) and HepaRG (r2 = 0.99, p < 0.001) cells. However, Emax values generated in hepatocytes were only significantly correlated to those determined in Fa2N-4 (r2 = 0.33, p = 0.005) and HepaRG cells (r2 = 0.79, p < 0.001). “Gold standard” cytochrome P450 induction data can be generated using primary human hepatocytes, but a restricted, erratic supply and interdonor variability somewhat restrict routine application within a drug discovery setting. HepaRG cells are a valuable recent addition to the armory of in vitro tools for assessing CYP3A4 induction and seem to be an excellent surrogate of primary cells.


Drug Metabolism and Disposition | 2006

HUMAN CYTOCHROME P450 INDUCTION AND INHIBITION POTENTIAL OF CLEVIDIPINE AND ITS PRIMARY METABOLITE H152/81

J. George Zhang; Shangara S. Dehal; Thuy Ho; Jennifer Johnson; Catherine Chandler; Andrew P. Blanchard; Robert J. Clark; Charles L. Crespi; David M. Stresser; James Wong

Clevidipine is a short-acting dihydropyridine calcium channel antagonist under development for treatment of perioperative hypertension. Patients treated with clevidipine are likely to be comedicated. Therefore, the potential for clevidipine and its major metabolite H152/81 to elicit drug interactions by induction or inhibition of cytochrome P450 was investigated. Induction of CYP1A2, CYP2C9, and CYP3A4 was examined in primary human hepatocytes treated with clevidipine at 1, 10, and 100 μM. Clevidipine was found to be an inducer of CYP3A4, but not of CYP1A2 or CYP2C9, at the 10 μM and 100 μM concentrations of clevidipine tested. Induction response for CYP3A4 to 100 μM clevidipine was approximately 20% of that of the positive control inducer rifampicin. The response of H152/81 was similar. Using cDNA-expressed enzymes, clevidipine inhibited CYP2C9, CYP2C19, and CYP3A4 activities with IC50 values below 10 μM, whereas CYP1A2, CYP2D6, and CYP2E1 activities were not substantially inhibited (IC50 values >70 μM). The Ki values for CYP2C9 and CYP2C19 were 1.7 and 3.3 μM, respectively, and those for CYP3A4 were 8.3 and 2.9 μM, using two substrates, testosterone and midazolam, respectively. These values are at least 10 times higher than the highest clevidipine concentration typically seen in the clinic. Little or no inhibition by H152/81 was found for the enzyme activities mentioned above (IC50 values ≥ 69 μM). The present study demonstrates that it is highly unlikely for clevidipine or its major metabolite to cause cytochrome P450-related drug interactions when used in the dose range required to manage hypertension in humans.


Methods in Enzymology | 2002

Design and application of fluorometric assays for human cytochrome P450 inhibition.

Charles L. Crespi; Vaughn P. Miller; David M. Stresser

Publisher Summary One important aspect of drug safety is the extent to which a new drug entity (NDE) causes metabolism-based pharmacokinetic interactions with co-administered medications. Therefore, potential drug-drug interactions may be tested for each enzyme of interest with a suitable probe substrate. The validity of these assays depends on the use of human enzymes and the probe substrate/assay conditions measuring the activity of the subject enzyme with a high degree of specificity. However, if enzyme mixtures, such as human fiver microsomes, are to be used probe substrate choices are more limited, as substrates with multiple pathways of metabolism are usually inappropriate. The chapter discusses some general considerations for conducting fluorometric cytochrome P450 enzyme (CYP) inhibition assays and then provides a general method for conducting the assay for several of the drug-metabolizing CYPs. Assays should be conducted under initial rate conditions formation of the metabolite should be linear with respect to enzyme concentration and incubation time, and the total consumption of the substrate should be less than 20%.


Endocrine Research | 2002

ASSESSING PREGNANCY RISKS OF AZOLE ANTIFUNGALS USING A HIGH THROUGHPUT AROMATASE INHIBITION ASSAY

Laura Kragie; Stephanie D. Turner; Christopher J. Patten; Charles L. Crespi; David M. Stresser

Human aromatase (CYP19) converts C19 androgens to aromatic C18 estrogenic steroids. Its activity is critical for early and mid pregnancy maintenance and in regulating parturition in late pregnancy. Past studies have utilized placental microsome tritiated water release assay to assess drug-hormone interactions with estrogen synthesis. We compared data from human placental assays with BD Gentests high throughput recombinant CYP19 enzyme assay using the fluorometric substrate dibenzylfluorescein. We tested a panel of azole antifungal agents that are commonly administered to women of childbearing potential, for their potential to inhibit aromatase. Potency varied by several orders of magnitude. Plasma and tissue levels of some azole drugs following oral or topical administration are at or above these IC50 values. These include the oral agents fluconazole and ketoconazole, and the topical agents econazole, bifonazole, clotrimazole, miconazole, and sulconazole. Conclusions: 1. Recombinant enzyme assay data are comparable to the human placental assay data in both SAR rank order and potency. 2. Plasma and tissue levels of some azole drugs following oral or topical administration are at or above these IC50 values. Therefore, some azole drugs may disrupt estrogen production in pregnancy, affecting pregnancy outcome. 3. Recombinant CYP19 assay using the fluorometric substrate dibenzylfluorescein, demonstrates rapid screening potential for chemicals that may affect pregnancy outcome as a result of CYP19 inhibition.


Drug Metabolism and Disposition | 2012

Sequential Metabolism of AMG 487, a Novel CXCR3 Antagonist, Results in Formation of Quinone Reactive Metabolites That Covalently Modify CYP3A4 Cys239 and Cause Time-Dependent Inhibition of the Enzyme

Kirk R. Henne; Thuy Tran; Brooke M. VandenBrink; Dan A. Rock; Divesh Aidasani; Raju Subramanian; Andrew K. Mason; David M. Stresser; Yohannes Teffera; Simon Wong; Michael G. Johnson; Xiaoqi Chen; George Tonn; Bradley K. Wong

CYP3A4-mediated biotransformation of (R)-N-(1-(3-(4-ethoxyphenyl)-4-oxo-3,4-dihydropyrido[2,3-d]pyrimidin-2-yl)ethyl)-N-(pyridin-3-ylmethyl)-2-(4-(trifluoromethoxy)phenyl)acetamide (AMG 487) was previously shown to generate an inhibitory metabolite linked to dose- and time-dependent pharmacokinetics in humans. Although in vitro activity loss assays failed to demonstrate CYP3A4 time-dependent inhibition (TDI) with AMG 487, its M2 phenol metabolite readily produced TDI when remaining activity was assessed using either midazolam or testosterone (KI = 0.73–0.74 μM, kinact = 0.088–0.099 min−1). TDI investigations using an IC50 shift method successfully produced inhibition attributable to AMG 487, but only when preincubations were extended from 30 to 90 min. The shift magnitude was ∼3× for midazolam activity, but no shift was observed for testosterone activity. Subsequent partition ratio determinations conducted for M2 using recombinant CYP3A4 showed that inactivation was a relatively inefficient process (r = 36). CYP3A4-mediated biotransformation of [3H]M2 in the presence of GSH led to identification of two new metabolites, M4 and M5, which shifted focus away from M2 being directly responsible for TDI. M4 (hydroxylated M2) was further metabolized to form reactive intermediates that, upon reaction with GSH, produced isomeric adducts, collectively designated M5. Incubations conducted in the presence of [18O]H2O confirmed incorporation of oxygen from O2 for the majority of M4 and M5 formed (>75%). Further evidence of a primary role for M4 in CYP3A4 TDI was generated by protein labeling and proteolysis experiments, in which M4 was found to be covalently bound to Cys239 of CYP3A4. These investigations confirmed a primarily role for M4 in CYP3A4 inactivation, suggesting that a more complex metabolic pathway was responsible for generation of inhibitory metabolites affecting AMG 487 human pharmacokinetics.


Drug Metabolism Letters | 2010

A Multi-Endpoint Evaluation of Cytochrome P450 1A2, 2B6 and 3A4 Induction Response in Human Hepatocyte Cultures After Treatment with β - Naphthoflavone, Phenobarbital and Rifampicin

Jingxian Zhang; Thuy Ho; Alanna L. Callendrello; Charles L. Crespi; David M. Stresser

U.S. FDA and EMEA guidance recommend that the preferred in vitro model for cytochrome P450 induction testing is human hepatocytes coupled with acceptable inducers as controls. However, there are surprisingly few published studies characterizing this model system for dose and time-dependence response to model inducing compounds. The concentration-dependent response and time-course for the induction of CYP1A2, CYP2B6 and CYP3A4 by inducing agents β-naphthoflavone, phenobarbital and rifampicin, respectively were examined in two or more donors using multiple end-points (mRNA, enzyme activity and Western blot analysis). Depending on the endpoint, exposure time for maximal response of CYP induction potential for the three enzymes ranged from 24 to 72 hours. Of the concentrations of BNF, PB and RIF tested, those which gave the maximal response were found to be 33 µM, > 2 mM and 10 µM, respectively.


Drug Metabolism and Disposition | 2009

Differential Time-and NADPH-Dependent Inhibition of CYP2C19 by Enantiomers of Fluoxetine

David M. Stresser; Andrew K. Mason; Elke S. Perloff; Thuy Ho; Charles L. Crespi; Andre A. Dandeneau; Ling Morgan; Shangara S. Dehal

Fluoxetine [±-N-methyl-3-phenyl-3-[(α, α, (-trifluoro-p-tolyl)oxy]-propylamine)] a selective serotonin reuptake inhibitor, is widely used in treating depression and other serotonin-dependent disease conditions. Racemic, (R)- and (S)-fluoxetine are potent reversible inhibitors of CYP2D6, and the racemate has been shown to be a mechanism-based inhibitor of CYP3A4. Racemic fluoxetine also demonstrates time- and concentration-dependent inhibition of CYP2C19 catalytic activity in vitro. In this study, we compared fluoxetine, its (R)- and (S)-enantiomers, ticlopidine, and S-benzylnirvanol as potential time-dependent inhibitors of human liver microsomal CYP2C19. In a reversible inhibition protocol (30 min preincubation with liver microsomes without NADPH), we found (R)-, (S)- and racemic fluoxetine to be moderate inhibitors with IC50 values of 21, 93, and 27 μM, respectively. However, when the preincubation was supplemented with NADPH, IC50 values shifted to 4.0, 3.4, and 3.0 μM, respectively resulting in IC50 shifts of 5.2-, 28-, and 9.3-fold. Ticlopidine showed a 1.8-fold shift in IC50 value, and S-benzylnirvanol shifted right (0.41-fold shift). Follow-up KI and kinact determinations with fluoxetine confirmed time-dependent inhibition [KI values of 6.5, 47, and 14 μM; kinact values of 0.023, 0.085, 0.030 min–1 for (R)-, (S)-, and racemate, respectively]. Although the (S)-isomer exhibits a much lower affinity for CYP2C19 inactivation relative to the (R)-enantiomer, it exhibits a more rapid rate of inactivation. Racemic norfluoxetine exhibited an 11-fold shift (18–1.5 μM) in IC50 value, suggesting that conversion of fluoxetine to this metabolite represents a metabolic pathway leading to time-dependent inhibition. These data provide an improved understanding of the drug-interaction potential of fluoxetine.


Drug Metabolism and Disposition | 2014

Evaluation of calibration curve-based approaches to predict clinical inducers and noninducers of CYP3A4 with plated human hepatocytes.

Zhang Jg; Thuy Ho; Callendrello Al; Robert J. Clark; Santone Ea; Kinsman S; Xiao D; Fox Lg; Heidi J. Einolf; David M. Stresser

Cytochrome P450 (P450) induction is often considered a liability in drug development. Using calibration curve–based approaches, we assessed the induction parameters R3 (a term indicating the amount of P450 induction in the liver, expressed as a ratio between 0 and 1), relative induction score, Cmax/EC50, and area under the curve (AUC)/F2 (the concentration causing 2-fold increase from baseline of the dose-response curve), derived from concentration-response curves of CYP3A4 mRNA and enzyme activity data in vitro, as predictors of CYP3A4 induction potential in vivo. Plated cryopreserved human hepatocytes from three donors were treated with 20 test compounds, including several clinical inducers and noninducers of CYP3A4. After the 2-day treatment, CYP3A4 mRNA levels and testosterone 6β-hydroxylase activity were determined by real-time reverse transcription polymerase chain reaction and liquid chromatography–tandem mass spectrometry analysis, respectively. Our results demonstrated a strong and predictive relationship between the extent of midazolam AUC change in humans and the various parameters calculated from both CYP3A4 mRNA and enzyme activity. The relationships exhibited with non-midazolam in vivo probes, in aggregate, were unsatisfactory. In general, the models yielded better fits when unbound rather than total plasma Cmax was used to calculate the induction parameters, as evidenced by higher R2 and lower root mean square error (RMSE) and geometric mean fold error. With midazolam, the R3 cut-off value of 0.9, as suggested by US Food and Drug Administration guidance, effectively categorized strong inducers but was less effective in classifying midrange or weak inducers. This study supports the use of calibration curves generated from in vitro mRNA induction response curves to predict CYP3A4 induction potential in human. With the caveat that most compounds evaluated here were not strong inhibitors of enzyme activity, testosterone 6β-hydroxylase activity was also demonstrated to be a strong predictor of CYP3A4 induction potential in this assay model.

Collaboration


Dive into the David M. Stresser's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shangara S. Dehal

Worcester Foundation for Biomedical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anthony J. Lee

University of South Carolina

View shared research outputs
Researchain Logo
Decentralizing Knowledge