Network


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

Hotspot


Dive into the research topics where Kevin J. Petty is active.

Publication


Featured researches published by Kevin J. Petty.


Clinical Pharmacology & Therapeutics | 2003

Effects of the neurokinin1 receptor antagonist aprepitant on the pharmacokinetics of dexamethasone and methylprednisolone

Jacqueline McCrea; Anup Majumdar; Michael R. Goldberg; Marian Iwamoto; Cynthia Gargano; Deborah Panebianco; Michael Hesney; Christopher Lines; Kevin J. Petty; Paul J. Deutsch; M. Gail Murphy; Keith M. Gottesdiener; D. Ronald Goldwater; Robert A. Blum

Aprepitant is a neurokinin1 receptor antagonist that, in combination with a corticosteroid and a 5‐hydroxytryptamine3 receptor antagonist, has been shown to be very effective in the prevention of chemotherapy‐induced nausea and vomiting. At doses used for the management of chemotherapy‐induced nausea and vomiting, aprepitant is a moderate inhibitor of cytochrome P4503A4 and may be used in conjunction with corticosteroids such as dexamethasone and methylprednisolone, which are substrates of cytochrome P4503A4. The effects of aprepitant on the these 2 corticosteroids were evaluated.


The Journal of Clinical Pharmacology | 2004

Evaluation of Potential Inductive Effects of Aprepitant on Cytochrome P450 3A4 and 2C9 Activity

Craig R. Shadle; Yih Lee; Anup Majumdar; Kevin J. Petty; Cynthia Gargano; Thomas E. Bradstreet; Judith K. Evans; Robert A. Blum

The NK1 receptor antagonist aprepitant (EMEND®), developed for use in combination with a 5HT3 receptor antagonist and a corticosteroid to prevent highly emetogenic chemotherapy‐induced nausea and vomiting (CINV), has been shown to have a moderate inhibitory effect as well as a possible inductive effect on cytochrome P450 (CYP) 3A4. Aprepitant has been noted to produce modest decreases in plasma S(−)‐warfarin concentrations, suggesting potential induction of CYP2C9. Because metabolism of some chemotherapeutic agents may involve CYP3A4, the potential inductive effect of the CINV dosing regimen of aprepitant on this metabolic pathway was evaluated using intravenous midazolam, a sensitive probe substrate of CYP3A4. The time course of induction of CYP2C9 by aprepitant was also evaluated using oral tolbutamide, a probe substrate of CYP2C9. In this double‐blind, randomized, placebo‐controlled, single‐center study, 24 healthy subjects were randomized (12 subjects per group) to receive either an aprepitant 3‐day regimen (aprepitant 125 mg p.o. on day 1 and aprepitant 80 mg p.o. on days 2 and 3) or matching placebo. All subjects also received probe drugs (midazolam 2 mg i.v. and tolbutamide 500 mg p.o.) once prior to aprepitant dosing (baseline) and again on days 4, 8, and 15. The ratio (aprepitant/placebo) of the geometric mean area under the plasma concentration curve (AUC) fold‐change from baseline for midazolam was 1.25 on day 4 (p < 0.01), 0.81 on day 8 (p < 0.01), and 0.96 on day 15 (p = 0.646). The ratio (aprepitant/placebo) of the geometric mean AUC fold‐change from baseline for tolbutamide was 0.77 on day 4 (p < 0.01), 0.72 on day 8 (p < 0.001), and 0.85 on day 15 (p = 0.05). Assessed using intravenous midazolam as a probe, aprepitant 125/80 mg p.o. administered over days 1 to 3 produced clinically insignificant weak inhibition (day 4) and induction (day 8) of CYP3A4 activity and no effect on CYP3A4 activity on day 15. Assessed using oral tolbutamide as a probe, the aprepitant regimen also produced modest induction of CYP2C9 activity on days 4 and 8, which resolved nearly to baseline by day 15. Thus, the aprepitant regimen for CINV results in modest, transient induction of CYPs 3A4 and 2C9 in the 2 weeks following administration.


Clinical Therapeutics | 2003

Effects of aprepitant on the pharmacokinetics of ondansetron and granisetron in healthy subjects

Robert A. Blum; Anup Majumdar; Jacqueline McCrea; John Busillo; Laura H. Orlowski; Deborah Panebianco; Michael Hesney; Kevin J. Petty; Michael R. Goldberg; M. Gail Murphy; Kevin M. Gottesdiener; Carolyn M. Hustad; Christian D. Lates; Walter K. Kraft; Sandi Van Buren; Scott A. Waldman; Howard E. Greenberg

BACKGROUND The neurokinin-1-receptor antagonist aprepitant, when given in combination with a corticosteroid and a 5-hydroxytryptamine type 3 (5-HT(3))-receptor antagonist, has been shown to be effective for the prevention of acute and delated chemotherapy-induced nausea and vomiting (CINV). OBJECTIVE Two studies were conducted to determine whether concomitant administration of aprepitant altered the pharmacokinetic profiles of ondansetron and granisetron, two 5-HT(3)-receptor antagonists commonly used as antiemetic therapy for CINV. METHODS The 2 studies were randomized, open-label, crossover trials conducted in healthy subjects aged between 18 and 46 years. Study 1 involved the following 2 treatment regimens: aprepitant 375 mg PO, dexamethasone 20 mg PO, and ondansetron 32 mg IV on day 1, followed by aprepitant 250 mg PO and dexamethasone 8 mg PO on days 2 through 5; and dexamethasone 20 mg PO and ondansetron 32 mg IV on day 1, followed by dexamethasone 8 mg PO on days 2 through 5. Study 2 involved the following 2 treatment regimens: aprepitant 125 mg PO with granisetron 2 mg PO on day 1, followed by aprepitant 80 mg PO on days 2 and 3; and granisetron 2 mg PO on day 1 only. Individual plasma samples were used to estimate area under the plasma concentration-time curve from time zero to infinity (AUC(0- infinity )), peak plasma concentration, and apparent terminal elimination half-life (t(12)) of both ondansetron and granisetron. RESULTS Study 1 included 19 subjects (10 women, 9 men), and study 2 included 18 subjects (11 men, 7 women). Coadministration of aprepitant 375 mg produced a small but statistically significant increase in the AUC(0- infinity ) for intravenous ondansetron (from 1268.3 to 1456.5 ng.h/mL; P = 0.019), with no significant effect on peak concentration at the end of the infusion (360.8 ng/mL with aprepitant vs 408.4 ng/mL without) or t(12) (5.0 vs 4.5 hours, respectively). Coadministration of aprepitant 125 mg/80 mg did not alter the mean pharmacokinetic characteristics of oral granisetron (AUC(0- infinity ), 101.4 ng.h/mL with aprepitant vs 92.2 ng.h/mL without; maximum plasma concentration, 9.0 ng/mL with and without aprepitant; time to maximum plasma concentration, both 3.0 hours; t(12), 6.5 vs 6.9 hours, respectively). CONCLUSION Concomitant administration of aprepitant had no clinically significant effect on the mean pharmacokinetic characteristics of either ondansetron or granisetron in these healthy subjects.


Clinical Pharmacology & Therapeutics | 2003

Effects of aprepitant on cytochrome P450 3A4 activity using midazolam as a probe.

Anup Majumdar; Jacqueline McCrea; Deborah Panebianco; Michael Hesney; James Dru; M.L Constanzer; Michael R. Goldberg; Gail Murphy; Keith M. Gottesdiener; Christopher R. Lines; Kevin J. Petty; R.A. Blum

Aprepitant is a neurokinin1 receptor antagonist that enhances prevention of chemotherapy‐induced nausea and vomiting when added to conventional therapy with a corticosteroid and a 5‐hydroxytryptamine3 (5‐HT3) antagonist. Because aprepitant may be used with a variety of chemotherapeutic agents and ancillary support drugs, which may be substrates of cytochrome P450 (CYP) 3A4, assessment of the potential of this drug to inhibit CYP3A4 activity in vivo is important. The effect of aprepitant on in vivo CYP3A4 activity in humans with oral midazolam used as a sensitive probe of CYP3A4 activity was evaluated in this study.


The Journal of Clinical Pharmacology | 2006

Pharmacokinetics of Aprepitant After Single and Multiple Oral Doses in Healthy Volunteers

Anup Majumdar; Laura Howard; Michael R. Goldberg; Lisa Hickey; M.L Constanzer; Paul Rothenberg; Tami M. Crumley; Deborah Panebianco; Thomas E. Bradstreet; Arthur J. Bergman; Scott A. Waldman; Howard E. Greenberg; Kathleen Butler; A. Knops; Inge De Lepeleire; Nicole Michiels; Kevin J. Petty

Aprepitant is the first NK1 receptor antagonist approved for use with corticosteroids and 5HT3 receptor antagonists to prevent chemotherapy‐induced nausea and vomiting (CINV). The effective dose to prevent CINV is a 125‐mg capsule on day 1 followed by an 80‐mg capsule on days 2 and 3. Study 1 evaluated the bioavailability of the capsules and estimated the effect of food. The mean (95% confidence interval [CI]) bioavailabilities of 125‐mg and 80‐mg final market composition (FMC) capsules, as assessed by simultaneous administration of stable isotope‐labeled intravenous (IV) aprepitant (2 mg) and FMC capsules, were 0.59 (0.53, 0.65) and 0.67 (0.62, 0.73), respectively. The geometric mean (90% CI) area under the plasma concentration time curve (AUC) ratios (fed/fasted) were 1.2 (1.10, 1.30) and 1.09 (1.00, 1.18) for the 125‐mg and 80‐mg capsule, respectively, demonstrating that aprepitant can be administered independently of food. Study 2 defined the pharmacokinetics of aprepitant administered following the 3‐day regimen recommended to prevent CINV (125 mg/80 mg/80 mg). Consistent daily plasma exposures of aprepitant were obtained following this regimen, which was generally well tolerated.


Journal of Pharmacology and Experimental Therapeutics | 2009

In vitro and in vivo properties of 3-tert-butyl-7-(5-methylisoxazol-3-yl)-2-(1-methyl-1H-1,2,4-triazol-5-ylmethoxy)-pyrazolo[1,5-d]-[1,2,4]triazine (MRK-016), a GABAA receptor alpha5 subtype-selective inverse agonist

John R. Atack; Karen A. Maubach; Keith A. Wafford; Desmond O'Connor; A. David Rodrigues; David C. Evans; F. David Tattersall; Mark Stuart Chambers; Angus Murray Macleod; Wai Si Eng; Christine Ryan; Eric Hostetler; Sandra Sanabria; Raymond E. Gibson; Stephen Krause; H. Donald Burns; Richard Hargreaves; Nancy G. B. Agrawal; Ruth M. McKernan; M. Gail Murphy; Kevin J. Gingrich; Gerard R. Dawson; Donald G. Musson; Kevin J. Petty

3-tert-Butyl-7-(5-methylisoxazol-3-yl)-2-(1-methyl-1H-1,2,4-triazol-5-ylmethoxy)-pyrazolo[1,5-d][1,2,4]triazine (MRK-016) is a pyrazolotriazine with an affinity of between 0.8 and 1.5 nM for the benzodiazepine binding site of native rat brain and recombinant human α1-, α2-, α3-, and α5-containing GABAA receptors. It has inverse agonist efficacy selective for the α5 subtype, and this α5 inverse agonism is greater than that of the prototypic α5-selective compound 3-(5-methylisoxazol-3-yl)-6-[(1-methyl-1,2,3-triazol-4-hdyl)methyloxy]-1,2,4-triazolo[3,4-a]phthalazine (α5IA). Consistent with its greater α5 inverse agonism, MRK-016 increased long-term potentiation in mouse hippocampal slices to a greater extent than α5IA. MRK-016 gave good receptor occupancy after oral dosing in rats, with the dose required to produce 50% occupancy being 0.39 mg/kg and a corresponding rat plasma EC50 value of 15 ng/ml that was similar to the rhesus monkey plasma EC50 value of 21 ng/ml obtained using [11C]flumazenil positron emission tomography. In normal rats, MRK-016 enhanced cognitive performance in the delayed matching-to-position version of the Morris water maze but was not anxiogenic, and in mice it was not proconvulsant and did not produce kindling. MRK-016 had a short half-life in rat, dog, and rhesus monkey (0.3–0.5 h) but had a much lower rate of turnover in human compared with rat, dog, or rhesus monkey hepatocytes. Accordingly, in human, MRK-016 had a longer half-life than in preclinical species (∼3.5 h). Although it was well tolerated in young males, with a maximal tolerated single dose of 5 mg corresponding to an estimated occupancy in the region of 75%, MRK-016 was poorly tolerated in elderly subjects, even at a dose of 0.5 mg, which, along with its variable human pharmacokinetics, precluded its further development.


The Journal of Clinical Pharmacology | 2003

Pharmacokinetics of Etoricoxib in Patients with Renal Impairment

Nancy G. B. Agrawal; Catherine Z. Matthews; Ralph S. Mazenko; Walter F. Kline; E.J. Woolf; Arturo G. Porras; Leslie A. Geer; Peggy Wong; Meehung Cho; Josee Cote; Thomas Marbury; Jack W. Moncrief; Harry Alcorn; Suzanne K. Swan; Marshall Sack; Richard Robson; Kevin J. Petty; Jules I. Schwartz; Keith M. Gottesdiener

The effect of renal insufficiency on the pharmacokinetics of etoricoxib, a selective inhibitor of cyclooxygenase‐2, was examined in 23 patients with varying degrees of renal impairment (12 moderate [creatinine clearance between 30 and 50 mL/min/1.73 m2], 5 severe [creatinine clearance below 30 mL/min/1.73 m2], and 6 with end‐stage renal disease requiring hemodialysis) following administration of single 120‐mg oral doses of etoricoxib. Even the most severe renal impairment was found to have little effect on etoricoxib pharmacokinetics. The low recovery of etoricoxib in dialysate (less than 6% of the dose) supports that hemodialysis also has little effect on etoricoxib pharmacokinetics, and binding of etoricoxib to plasma proteins was generally unaffected by renal disease. Single doses of etoricoxib were generally well tolerated by patients with renal impairment. Based on pharmacokinetic considerations, dosing adjustments are not necessary for patients with any degree of renal impairment. However, because patients with advanced renal disease (creatinine clearance below 30 mL/min/1.73 m2) are likely to be very sensitive to any further compromise of renal function, and there is no long‐term clinical experience in these patients, the use of etoricoxib is not recommended in patients with advanced renal disease.


The Journal of Clinical Pharmacology | 2003

Lack of Effect of Aprepitant on Digoxin Pharmacokinetics in Healthy Subjects

Martin Feuring; Yih Lee; Laura H. Orlowski; Nicole Michiels; Marina De Smet; Anup Majumdar; Kevin J. Petty; Michael R. Goldberg; M. Gail Murphy; Keith M. Gottesdiener; Michael Hesney; L. Ellen Brackett; Martin Wehling

Aprepitant is a highly selective neurokinin‐1 receptor antagonist that, in combination with a corticosteroid and a 5‐hydroxytryptamine3 (5HT3) receptor antagonist, has been shown to be efficacious in the prevention of highly emetogenic chemotherapy‐induced nausea and vomiting. In vitro data suggest that aprepitant is a substrate and a weak inhibitor of P‐glycoprotein. Thus, the effect of aprepitant on the pharmacokinetics of digoxin, a P‐glycoprotein substrate, was examined in a double‐blind, placebo‐controlled, randomized, two‐period crossover study in 12 healthy subjects. Each subject received daily oral doses of digoxin 0.25 mg on Days 1 through 13 during both treatment periods. Aprepitant 125 mg (or matching placebo) was coadministered orally with digoxin on Day 7, and aprepitant 80 mg (or matching placebo) was coadministered orally with digoxin on Days 8 to 11. Aprepitant did not affect the pharmacokinetics of digoxin. The geometric mean ratios (90% confidence interval [CI]) for plasma AUC0–24 h of digoxin (with/without aprepitant) were 0.99 (0.91, 1.09) and 0.93 (0.83, 1.05) on Days 7 and 11, respectively, and the geometric mean ratios (90% CI) for the 24‐hour urinary excretion of immunoreactive digoxin (with/without aprepitant) were 0.91 (0.80, 1.04) and 1.00 (0.91, 1.09) on Days 7 and 11, respectively. Thus, aprepitant, when dosed as a 5‐day regimen, did not interact with a known substrate of the P‐glycoprotein transporter.


The Journal of Clinical Pharmacology | 2007

Effect of Aprepitant on the Pharmacokinetics of Intravenous Midazolam

Anup Majumdar; Kerri X. Yan; Diana V. Selverian; Suna Barlas; M.L Constanzer; James Dru; Jacqueline McCrea; Tuli Ahmed; Glen S. Frick; Walter K. Kraft; Kevin J. Petty; Howard E. Greenberg

Oral aprepitant 125 mg, an antiemetic and a moderate inhibitor of the metabolism of oral midazolam, was assessed for interaction with intravenous midazolam in 12 subjects randomized to intravenous midazolam 2 mg ± oral aprepitant 125 mg. The hypothesis was that midazolam AUC would not change by more than 2‐fold (consistent with no more than weak inhibition) when midazolam + aprepitant was compared with midazolam alone. An AUC geometric mean ratio (midazolam + aprepitant/midazolam) with 90% confidence interval upper bound ≤2.0 (an increase in midazolam felt to be of modest clinical significance in the highly monitored perioperative period) was prespecified. Aprepitant increased intravenous midazolam AUC0–∞ 1.47‐fold (90% confidence interval, 1.36–1.59), which fell within the prespecified criterion.


Clinical Pharmacokinectics | 2005

Effect of Impaired Renal Function and Haemodialysis on the Pharmacokinetics of Aprepitant

Arthur J. Bergman; Thomas Marbury; Trisha Fosbinder; Suzanne K. Swan; Lisa Hickey; Thomas E. Bradstreet; John Busillo; Kevin J. Petty; Kala-Jyoti Viswanathan Aiyer; M.L. Constanzer; Su-Er W. Huskey; Anup Majumdar

AbstractBackground: The neurokinin NK1-receptor antagonist aprepitant has demonstrated efficacy in preventing highly emetogenic chemotherapy-induced nausea and vomiting. Objective: The objective of the present study was to investigate the effects of impaired renal function on the pharmacokinetics and safety of aprepitant. Subjects and methods: A total of 32 patients and healthy subjects were evaluated in this open-label, two-part study. Pharmacokinetic parameters after a single oral dose of aprepitant 240mg were measured in eight patients with end-stage renal disease (ESRD) requiring haemodialysis, eight patients with severe renal insufficiency (SRI [24-hour creatinine clearance <30 mL/min/1.73m2]) and 16 healthy and age-, sex- and weight-matched subjects (controls). Results: In ESRD patients, the area under the plasma concentration-time curve (AUC) from 0 to 48 hours (AUC48) for aprepitant was on average approximately 36% lower than that observed in control subjects (ratio [ESRD patients/healthy controls] of geometric means = 0.64), but the 90% confidence interval 0.52, 0.78 for the ratio of true mean AUC48 fell within the predefined target interval of 0.5, 2.0. Also in ESRD patients, there was no statistically or clinically significant difference in unbound aprepitant AUC (which may be more clinically relevant than total aprepitant AUC) when compared with healthy control subjects. Aprepitant pharmacokinetic parameters in ESRD patients were clinically similar when haemodialysis was initiated at 4 hours or 48 hours after aprepitant administration. In SRI patients, the ratio (SRI patients/healthy controls) of aprepitant AUC from zero to infinity (AUC∞) geometric mean value was 0.79 with a 90% confidence interval of 0.56, 1.10. On average, in SRI patients the principal aprepitant pharmacokinetic parameters (AUC∞, initial maximum plasma concentration [Cmax], time to initial Cmax, and apparent elimination half-life) were not statistically different from those obtained in healthy control subjects. Aprepitant was generally well tolerated in both ESRD and SRI patients. Conclusion: The results of this study demonstrate that ESRD, haemodialysis and SRI have no clinically meaningful effect on aprepitant pharmacokinetics. Therefore, no dosage adjustment of aprepitant is warranted in SRI or ESRD patients.

Collaboration


Dive into the Kevin J. Petty's collaboration.

Top Co-Authors

Avatar

Anup Majumdar

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Michael R. Goldberg

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Cynthia Gargano

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge