Network


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

Hotspot


Dive into the research topics where Thomas R. MacGregor is active.

Publication


Featured researches published by Thomas R. MacGregor.


Hiv Clinical Trials | 2004

Pharmacokinetic Characterization of Different Dose Combinations of Coadministered Tipranavir and Ritonavir in Healthy Volunteers

Thomas R. MacGregor; John P. Sabo; Stephen Norris; Philip Johnson; Lawrence Galitz; Scott McCallister

Abstract Purpose: To characterize the steady-state pharmacokinetic combination of the nonpeptidic protease inhibitor tipranavir (TPV) with ritonavir (RTV) in 95 healthy adult volunteers, a phase 1, single-center, open-label, randomized, parallel-group trial was conducted. Method: Participants received 250-mg self-emulsifying drug delivery system (SEDDS) capsules of TPV at doses between 250 mg and 1250 mg twice daily for 11 days, then received one or two RTV 100-mg SEDDS capsules, in addition to the TPV capsules, for the next 21 days. Results: Coadministration of TPV and RTV (TPV/r) resulted in a greater than 20-fold increase in steady-state TPV trough concentrations (Cssmin) as compared with TPV at steady state alone. Mean TPV Cssmin was above a preliminary target threshold of 20 μM with all but one of the RTV-boosted doses; without boosting, none of the TPV-alone doses exceeded the threshold. The average steady-state Cssmin for TPV 500 mg and 750 mg with RTV 100 mg or 200 mg were 20 to 57 times the protein-adjusted TPV IC90 for protease inhibitor-resistant HIV-1. An erythromycin breath test, a surrogate marker for cytochrome P450 isoenzyme 3A4 activity, indicated that all TPV/r combinations given provided net inhibition of this isoenzyme. The most frequent treatment-related adverse events were mild gastrointestinal symptoms. Conclusion: This phase 1 study demonstrated that RTV-boosted TPV achieves concentrations that are expected to be effective in treating drug-experienced patients.


Biopharmaceutics & Drug Disposition | 1999

Single dose pharmacokinetics and bioavailability of nevirapine in healthy volunteers

Michael Lamson; John P. Sabo; Thomas R. MacGregor; Joseph W. Pav; Lois Rowland; Amale Hawi; Michael L. Cappola; Patrick A. Robinson

The results of two randomized, single‐dose, crossover bioavailability studies are presented which describe the pharmacokinetics and oral bioavailability of nevirapine, a novel nonnucleoside antiretroviral drug. In the first study 12 healthy male volunteers received nevirapine 15 mg via short‐term i.v. infusion or orally as a 50 mg tablet or reference solution (50 mg/200 mL). Following the i.v. dose, nevirapine had a low systemic clearance (Mean±S.D., Cl=1.4±0.3 L/h) and a prolonged elimination phase (t1/2β=52.8±14.8 h; MRT=81.4±22.4 h). Nevirapine absolute bioavailability was 93±9% and 91±8% for the tablet and oral solution, respectively. In the second study, 24 healthy male volunteers were administered nevirapine as a 200 mg production‐line tablet or oral reference solution (200 mg/200 mL). There was no significant difference in bioavailability between the tablet and reference solution. Overall, comparison of the pharmacokinetic parameters between the 50 and 200 mg doses indicates that nevirapine is well absorbed at clinically relevant doses. The absorption profiles using deconvolution revealed no evidence of differential enzyme induction between the two doses or routes of administration following a single dose. Copyright


Aaps Pharmsci | 2000

Pharmacokinetics of nevirapine and lamivudine in patients with HIV-1 infection.

John P. Sabo; Michael Lamson; Gerhard Leitz; Chan-Loi Yong; Thomas R. MacGregor

The purpose of this parallel treatment group, double-blind. multicenter study was to lharacterize the pharmacokinetics of nevirapine and lamivudine when coadministered to patients with the HIV-1 infection. This pharmacokinetic interaction study was nested within a larger Phase III clinical trial conducted to characterize the safety and efficacy of coadministered nevirapine and lamivudine. One hundred HIV-1 infected patients with CD4+ lymphocyte counts=200 cells/mm3 and who were on a background of nucleoside (zidovudine [ZDV], didanosine [ddl], zalcitabine [ddC], stavudine [d4T]) therapy were randomly assigned to be treated with either nucleoside+ lamivudine+nevirapine or nucleoside+lamivudine+ placebo. Each patient underwent blood sampling at defined times for the purpose of determining the concentration of nevirapine in plasma and lamivudine in serum under steady-state conditions. Each patient was also monitored closely for concomitant administration of other drugs, including ZDV, ddl, ddC, d4T and cotrimoxazole. The pharmacokinetics of nevirapine and lamivudine were characterized using nonlinear mixed-effects modeling. There were no reported serious adverse events during the 40-day pharmacokinetic study. The results of the modeling analysis revealed that nevirapine had no effect on the pharmacokinetics of lamivudine. Estimates of the apparent clearance for nevirapine (CL/F=3.3 L/hour; 95% confidence interval [Cl] 2.9 to 3.7 L/hour) and lamivudine (CL/F 27.6 L/hour; 95% Cl 22 to 33.2 L/hour) were consistent with the values reported in earlier trials. However, the results also showed that concomitant administration of lamivudine with cotrimoxazole resulted in a 31% reduction in the apparent clearance of lamivudine, resulting in a 43% increase in the average steady-state lamivudine serum concentrations. These results indicate that chronic concurrent administration of cotrimoxazole with lamivudine may significantly affect the steady-state pharmacokinetics of lamivudine.


Clinical Pharmacology & Therapeutics | 1985

Pharmacokinetics of Transdermally Delivered Clonidine

Thomas R. MacGregor; Kandace M. Matzek; James J. Keirns; Rudolf G A van Wayjen; Abraham van den Ende; Rudolf G L van Tol

We detail a series of pharmacokinetic investigations to determine the dose linearity, the effect of site of application, the duration of steady‐state plasma concentrations, and the effect of chronic application when clonidine is administered transdermally. Dose linearity was assessed in six subjects with normotension after application of increasing sizes of transdermal clonidine systems (3.5, 7.0, and 10.5 cm2 size) to the upper outer arm. Of the six subjects studied, five had linear relationships between clonidine plasma concentrations at steady state and system size of >0.975; in the sixth subject the correlation was >0.90. The mean steady‐state plasma concentrations with 3.5, 7.0, and 10.5 cm2 systems were 0.39, 0.84, and i.12 ng/ml, respectively. The influence of site and duration of application on the absorption of transdermal clonidine was studied in eight subjects with normotension by use of the 3.5 cm2 system. The mean steady‐state plasma concentration over the time interval from 3 to 7 days after application to the arm or to the chest did not significantly differ. When a system was left on the chest or arm for a total of 11 days (4 days beyond the recommended time to change systems), the plasma concentrations of seven of eight subjects with application to the arm and of six of eight subjects with application to the chest remained constant through day 11. The influence of consecutive applications of 3.5 cm2 transdermal clonidine systems on steady‐state plasma clonidine concentrations was also studied in eight subjects with normotension over an 11‐day period. Steady‐state plasma concentrations were achieved in all eight subjects within 3 days after application of the initial transdermal clonidine system. On days 4 and 7 the previous transdermal system was removed and a new system was applied to the alternate arm. The mean steady‐state plasma concentration (3 to 11 days) for the eight subjects was 0.32 ng/ml. There was no apparent difference between the mean plasma concentration at the time the clonidine system was removed and 24 hours after application of a new system.


Antimicrobial Agents and Chemotherapy | 2005

Interaction of Ritonavir-Boosted Tipranavir with Loperamide Does Not Result in Loperamide-Associated Neurologic Side Effects in Healthy Volunteers

Geoffrey Mukwaya; Thomas R. MacGregor; David Hoelscher; Thomas Heming; Daniel Legg; Kelli Kavanaugh; Phillip Johnson; John P. Sabo; Scott McCallister

ABSTRACT Loperamide (LOP) is a peripherally acting opioid receptor agonist used for the management of chronic diarrhea through the reduction of gut motility. The lack of central opioid effects is partly due to the efflux activity of the multidrug resistance transporter P-glycoprotein (P-gp) at the blood-brain barrier. The protease inhibitors are substrates for P-gp and have the potential to cause increased LOP levels in the brain. Because protease inhibitors, including tipranavir (TPV), are often associated with diarrhea, they are commonly used in combination with LOP. The level of respiratory depression, the level of pupil constriction, the pharmacokinetics, and the safety of LOP alone compared with those of LOP-ritonavir (RTV), LOP-TPV, and LOP-TPV-RTV were evaluated in a randomized, open-label, parallel-group study with 24 healthy human immunodeficiency virus type 1-negative adults. Respiratory depression was assessed by determination of the ventilatory response to carbon dioxide. Tipranavir-containing regimens (LOP-TPV and LOP-TPV-RTV) caused decreases in the area under the concentration-time curve from time zero to infinity for LOP (51% and 63% decreases, respectively) and its metabolite (72% and 77% decreases, respectively), whereas RTV caused increases in the levels of exposure of LOP (121% increase) and its metabolite (44% increase). In vitro and in vivo data suggest that TPV is a substrate for and an inducer of P-gp activity. The respiratory response to LOP in combination with TPV and/or RTV was not different from that to LOP alone. There was no evidence that LOP had opioid effects in the central nervous system, as measured indirectly by CO2 response curves and pupillary response in the presence of TPV and/or RTV.


Journal of Acquired Immune Deficiency Syndromes | 2004

A 14-day dose-response study of the efficacy, safety, and pharmacokinetics of the nonpeptidic protease inhibitor Tipranavir in treatment-naive HIV-1-infected patients

Scott McCallister; Hernan Valdez; Kevin Curry; Thomas R. MacGregor; Marie Borin; F. William Freimuth; Yenyung Wang; Douglas L. Mayers

Abstract:Tipranavir (TPV), a novel nonpeptidic protease inhibitor (NPPI), was administered to treatment-naive HIV-1–infected patients over 14 days in a randomized, multicenter, open-label, parallel-group trial to evaluate the efficacy and tolerability of a self-emulsifying drug delivery system (SEDDS) formulation, in combination with ritonavir (RTV). Of the 31 patients enrolled, 10 were randomized to receive TPV 1200 mg twice daily (TPV 1200), 10 patients received TPV 300 mg + RTV 200 mg twice daily (TPV/r 300/200), and 11 patients received TPV 1200 mg + RTV 200 mg twice daily (TPV/r 1200/200). The median baseline viral load and CD4 cell count were 4.96 log10 copies/mL and 244 cells/mm3, respectively. After 14 days, the median decrease in viral load was −0.77 log10 in the TPV 1200 group, −1.43 log10 in the TPV/r 300/200 group, and −1.64 log10 in the TPV/r 1200/200 group. TPV exposure was increased by 24- and 70-fold in the TPV/r 300/200 and 1200/200 groups, respectively, compared with TPV 1200 alone. There were no significant differences across treatment arms with regard to drug-related adverse events. TPV/r appeared to be safe, effective, and well tolerated during 14 days of treatment.


Clinical Pharmacology & Therapeutics | 1999

Nevirapine Induces Both CYP3A4 and CYP2B6 Metabolic Pathways

Michael Lamson; Thomas R. MacGregor; P. Riska; D. Erickson; P. Maxfield; L. Rowland; M. Gigliotti; P. Robinson; S. Azzam; James J. Keirns

Clinical Pharmacology & Therapeutics (1999) 65, 137–137; doi:


Clinical Pharmacology & Therapeutics | 2010

A Phenotype–Genotype Approach to Predicting CYP450 and P‐Glycoprotein Drug Interactions With the Mixed Inhibitor/Inducer Tipranavir/Ritonavir

Julie B. Dumond; M. Vourvahis; Naser L. Rezk; K. B. Patterson; H. C. Tien; Nicholas J. White; S. H. Jennings; S. O. Choi; J. Li; M. J. Wagner; N. M. La-Beck; M. Drulak; J. P. Sabo; M. A. Castles; Thomas R. MacGregor; Angela D. M. Kashuba

The effects of tipranavir/ritonavir (TPV/r) on hepatic and intestinal P‐glycoprotein (P‐gp) and cytochrome P450 (CYP) enzyme activity were evaluated in 23 volunteers. The subjects received oral (p.o.) caffeine, warfarin + vitamin K, omeprazole, dextromethorphan, and midazolam and digoxin (p.o. and intravenous (i.v.)) at baseline, during the first three doses of TPV/r (500 mg/200 mg b.i.d.), and at steady state. Plasma area under the curve (AUC)0–∞ and urinary metabolite ratios were used for quantification of protein activities. A single dose of TPV/r had no effect on the activity of CYP1A2 and CYP2C9; it weakly inhibited CYP2C19 and P‐gp; and it potently inhibited CYP2D6 and CYP3A. Multiple dosing produced weak induction of CYP1A2, moderate induction of CYP2C19, potent induction of intestinal P‐gp, and potent inhibition of CYP2D6 and CYP3A, with no significant effects on CYP2C9 and hepatic P‐gp. Several P450/transporter single‐nucleotide polymorphisms correlated with the baseline phenotype but not with the extent of inhibition or induction. Although mixed induction and inhibition are present, this approach offers an understanding of drug interaction mechanisms and ultimately assists in optimizing the clinical use of TPV/r.


Journal of Acquired Immune Deficiency Syndromes | 2008

Pharmacokinetics, safety, and efficacy of tipranavir boosted with ritonavir alone or in combination with other boosted protease inhibitors as part of optimized combination antiretroviral therapy in highly treatment-experienced patients (BI Study 1182.51).

Sharon Walmsley; Christine Katlama; Adriano Lazzarin; Keikawus Arestéh; Gerald Pierone; Gary Blick; Margaret Johnson; Ulrich Meier; Thomas R. MacGregor; Johnathan G Leith

Background:Given the limited treatment options for patients with high-level resistance, antiretroviral (ARV) regimens based on concomitant use of 2 ritonavir (RTV)-boosted protease inhibitors (PIs) were considered a therapeutic option. Methods:Boehringer Ingelheim (BI) study 1182.51 examined the pharmacokinetic profile, safety, and efficacy of RTV-boosted tipranavir (TPV/r), alone and in combination with comparator PIs (CPIs) in 315 triple-class-experienced, HIV-infected patients. Results:Two weeks after single PI therapy, the addition of TPV/r reduced plasma trough levels 52%, 80%, and 56% for lopinavir (LPV), saquinavir (SQV), and amprenavir (APV) recipients, respectively. After 2 weeks, a TPV/r-only regimen reduced HIV viral load (VL) by a median of 1.06 log10 copies/mL. VL reductions at 2 weeks between single-boosted CPIs were difficult to compare, because the numbers of patients maintaining their previous failing PI after randomization were different. At week 4, patients initiating treatment with TPV-containing regimens sustained VL reduction (median decrease of 1.27 log10 copies/mL). Patients adding TPV to regimens at week 2 achieved median reductions from a baseline of 1.19 log10, 0.96 log10, and 1.12 log10 copies/mL at week 4 in dual-boosted LPV, SQV, and APV groups, respectively. At 24 weeks, VL reductions (median: −0.24 to −0.47 log10 copies/mL) were comparable between treatment groups. Conclusions:The efficacy of a dual PI regimen depended on the presence of TPV, with additional recycled CPIs having limited activity, even in drug-resistant patient populations with plasma trough concentrations regarded as likely to be adequate in this study. No clear guidelines exist about ARV plasma trough concentrations in treatment-experienced patients, however.


Antimicrobial Agents and Chemotherapy | 2007

Steady-State Disposition of the Nonpeptidic Protease Inhibitor Tipranavir when Coadministered with Ritonavir

Linzhi Chen; John P. Sabo; Elsy Philip; Yanping Mao; Stephen Norris; Thomas R. MacGregor; Jan M. Wruck; Sandra Garfinkel; Mark Castles; Amy Brinkman; Hernan Valdez

ABSTRACT The pharmacokinetic and metabolite profiles of the antiretroviral agent tipranavir (TPV), administered with ritonavir (RTV), in nine healthy male volunteers were characterized. Subjects received 500-mg TPV capsules with 200-mg RTV capsules twice daily for 6 days. They then received a single oral dose of 551 mg of TPV containing 90 μCi of [14C]TPV with 200 mg of RTV on day 7, followed by twice-daily doses of unlabeled 500-mg TPV with 200 mg of RTV for up to 20 days. Blood, urine, and feces were collected for mass balance and metabolite profiling. Metabolite profiling and identification was performed using a flow scintillation analyzer in conjunction with liquid chromatography-tandem mass spectrometry. The median recovery of radioactivity was 87.1%, with 82.3% of the total recovered radioactivity excreted in the feces and less than 5% recovered from urine. Most radioactivity was excreted within 24 to 96 h after the dose of [14C]TPV. Radioactivity in blood was associated primarily with plasma rather than red blood cells. Unchanged TPV accounted for 98.4 to 99.7% of plasma radioactivity. Similarly, the most common form of radioactivity excreted in feces was unchanged TPV, accounting for a mean of 79.9% of fecal radioactivity. The most abundant metabolite in feces was a hydroxyl metabolite, H-1, which accounted for 4.9% of fecal radioactivity. TPV glucuronide metabolite H-3 was the most abundant of the drug-related components in urine, corresponding to 11% of urine radioactivity. In conclusion, after the coadministration of TPV and RTV, unchanged TPV represented the primary form of circulating and excreted TPV and the primary extraction route was via the feces.

Collaboration


Dive into the Thomas R. MacGregor's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge