Network


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

Hotspot


Dive into the research topics where Marian Iwamoto is active.

Publication


Featured researches published by Marian Iwamoto.


Drug Metabolism and Disposition | 2007

Metabolism and Disposition in Humans of Raltegravir (MK-0518), an Anti-AIDS Drug Targeting the Human Immunodeficiency Virus 1 Integrase Enzyme

Kelem Kassahun; Ian McIntosh; Donghui Cui; David Hreniuk; Shelia Merschman; Kenneth C. Lasseter; Neal Azrolan; Marian Iwamoto; John A. Wagner; Larissa Wenning

Raltegravir is a potent human immunodeficiency virus 1 (HIV-1) integrase strand transfer inhibitor that is being developed as a novel anti-AIDS drug. The absorption, metabolism, and excretion of raltegravir were studied in healthy volunteers after a single oral dose of 200 mg (200 μCi) of [14C]raltegravir. Plasma, urine, and fecal samples were collected at specified intervals up to 240 h postdose, and the samples were analyzed for total radioactivity, parent compound, and metabolites. Radioactivity was eliminated in substantial amounts in both urine (32%) and feces (51%). The elimination of radioactivity was rapid, since the majority of the recovered dose was attributable to samples collected through 24 h. In extracts of urine, two components were detected and were identified as raltegravir and the glucuronide of raltegravir (M2), and each accounted for 9% and 23% of the dose recovered in urine, respectively. Only a single radioactive peak, which was identified as raltegravir, was detected in fecal extracts; raltegravir in feces is believed to be derived, at least in part, from the hydrolysis of M2 secreted in bile, as demonstrated in rats. The major entity in plasma was raltegravir, which represented 70% of the total radioactivity, with the remaining radioactivity accounted for by M2. Studies using cDNA-expressed UDP-glucuronosyltransferases (UGTs), form-selective chemical inhibitors, and correlation analysis indicated that UGT1A1 was the main UGT isoform responsible for the formation of M2. Collectively, the data indicate that the major mechanism of clearance of raltegravir in humans is UGT1A1-mediated glucuronidation.


Clinical Pharmacology & Therapeutics | 2008

Safety, Tolerability, and Pharmacokinetics of Raltegravir After Single and Multiple Doses in Healthy Subjects

Marian Iwamoto; La Wenning; As Petry; M Laethem; M De Smet; Jt Kost; Sa Merschman; Km Strohmaier; Steven Ramael; Kenneth C. Lasseter; Julie A. Stone; Keith M. Gottesdiener; John A. Wagner

Raltegravir is a novel human immunodeficiency virus‐1 integrase inhibitor with potent in vitro activity (95% inhibitory concentration (IC95)=33 nM in 50% human serum). Three double‐blind, randomized, placebo‐controlled, pharmacokinetic, safety, and tolerability studies were conducted: (1) single‐dose escalation study (10–1,600 mg), (2) multiple‐dose escalation study (100–800 mg q12 h×10 days), and (3) single‐dose female study (400 mg). Raltegravir was rapidly absorbed with a terminal half‐life (t½) ∼7–12 h. Approximately 7–14% of raltegravir was excreted unchanged in urine. Area under the curve (AUC)0–∞ was similar between male and female subjects. After multiple‐dose administration, steady state was achieved within 2 days; there was little to modest accumulation of raltegravir. Trough levels were >33 nM for dose levels of 100 mg and greater. Raltegravir is generally well tolerated at doses of up to 1,600 mg/day given for up to 10 days and exhibits a pharmacokinetic profile supportive of twice‐daily dosing with multiple doses of 100 mg and greater achieving trough levels >33 nM.


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.


Antimicrobial Agents and Chemotherapy | 2009

Effect of Rifampin, a Potent Inducer of Drug-Metabolizing Enzymes, on the Pharmacokinetics of Raltegravir

Larissa Wenning; William D. Hanley; Diana M. Brainard; Amelia S. Petry; Kalyan Ghosh; Bo Jin; Eric Mangin; Thomas Marbury; Jolene Kay Berg; Jeffrey A. Chodakewitz; Julie A. Stone; Keith M. Gottesdiener; John A. Wagner; Marian Iwamoto

ABSTRACT Raltegravir is a human immunodeficiency virus type 1 integrase strand transfer inhibitor that is metabolized by glucuronidation via UGT1A1 and may be affected by inducers of UGT1A1, such as rifampin (rifampicin). Two pharmacokinetic studies were performed in healthy subjects: study 1 examined the effect of administration of 600-mg rifampin once daily on the pharmacokinetics of a single dose of 400-mg raltegravir, and study 2 examined the effect of 600-mg rifampin once daily on the pharmacokinetics of 800-mg raltegravir twice daily compared to 400-mg raltegravir twice daily without rifampin. Raltegravir coadministered with rifampin resulted in lower plasma raltegravir concentrations: in study 1, the geometric mean ratios (GMRs) and 90% confidence intervals (90% CIs) for the plasma raltegravir concentration determined 12 h postdose (C12), area under the concentration-time curve from 0 h to ∞ (AUC0-∞), and maximum concentration of drug in plasma (Cmax) (400-mg raltegravir plus rifampin/400-mg raltegravir) were 0.39 (0.30, 0.51), 0.60 (0.39, 0.91), and 0.62 (0.37, 1.04), respectively. In study 2, the GMRs and 90% CIs for raltegravir C12, AUC0-12, and Cmax (800-mg raltegravir plus rifampin/400-mg raltegravir) were 0.47 (0.36, 0.61), 1.27 (0.94, 1.71), and 1.62 (1.12, 2.33), respectively. Doubling the raltegravir dose to 800 mg when coadministered with rifampin therefore compensates for the effect of rifampin on raltegravir exposure (AUC0-12) but does not overcome the effect of rifampin on raltegravir trough concentrations (C12). Coadministration of rifampin and raltegravir is not contraindicated; however, caution should be used, since raltegravir trough concentrations in the presence of rifampin are likely to be at the lower limit of clinical experience.


Clinical Cancer Research | 2006

A Study to Determine the Effects of Food and Multiple Dosing on the Pharmacokinetics of Vorinostat Given Orally to Patients with Advanced Cancer

Eric H. Rubin; Nancy G. B. Agrawal; Evan J. Friedman; Pamela Scott; Kathryn E. Mazina; Linda Sun; Lihong Du; Justin L. Ricker; Stanley R. Frankel; Keith Gottesdiener; John A. Wagner; Marian Iwamoto

Purpose: This phase I study, conducted in advanced-stage cancer patients, assessed the safety and tolerability of oral vorinostat (suberoylanilide hydroxamic acid), single-dose and multiple-dose pharmacokinetics of vorinostat, and the effect of a high-fat meal on vorinostat pharmacokinetics. Experimental Design: Patients (n = 23) received single doses of 400 mg vorinostat on day 1 (fasted) and day 5 (fed) with 48 hours of pharmacokinetic sampling on both days. Patients received 400 mg vorinostat once daily on days 7 to 28. On day 28, vorinostat was given (fed) with pharmacokinetic sampling for 24 hours after dose. Results: The apparent t1/2 of vorinostat was short (∼1.5 hours). A high-fat meal was associated with a small increase in the extent of absorption and a modest decrease in the rate of absorption. A short lag time was observed before detectable levels of vorinostat were observed in the fed state, and Tmax was delayed. Vorinostat concentrations were qualitatively similar following single-dose and multiple-dose administration; the accumulation ratio based on area under the curve was 1.21. The elimination of vorinostat occurred primarily through metabolism, with <1% of the given dose recovered intact in urine. The most common vorinostat-related adverse experiences were mild to moderate nausea, anorexia, fatigue, increased blood creatinine, and vomiting. Conclusions: Vorinostat concentrations were qualitatively similar after single and multiple doses. A high-fat meal increased the extent and modestly decreased the rate of absorption of vorinostat; this effect is not anticipated to be clinically meaningful. Continued investigation of 400 mg vorinostat given once daily in phase II and III efficacy studies is warranted.


The Journal of Clinical Pharmacology | 2008

Lack of a Pharmacokinetic Effect of Raltegravir on Midazolam: In Vitro/In Vivo Correlation

Marian Iwamoto; Kelem Kassahun; Matthew D. Troyer; William D. Hanley; Ping Lu; Alisha Rhoton; Amelia S. Petry; Kalyan Ghosh; Eric Mangin; Emanuel P. DeNoia; Larissa Wenning; Julie A. Stone; Keith M. Gottesdiener; John A. Wagner

Raltegravir is a novel HIV‐1 integrase inhibitor with potent in vitro activity (95% inhibitory concentration = 33 nM in 50% human serum). In vitro characterization of raltegravir inhibition potential was assessed against a panel of cytochrome P450 (CYP) enzymes. An open‐label, 2‐period study was conducted to assess the effect of raltegravir on the pharmacokinetics of midazolam, a sensitive CYP 3A4 probe substrate: period 1, 2.0 mg of midazolam; period 2, 400 mg of raltegravir every 12 hours for 14 days with 2.0 mg of midazolam on day 14. There was no meaningful in vitro effect of raltegravir on inhibition of a panel of CYP enzymes and induction of CYP 3A4. In the presence of raltegravir, midazolam area under the curve extrapolated to infinity (AUC0‐infin) and maximum plasma concentration (Cmax) geometric mean ratios were similar (geometric mean ratios and 90% confidence intervals: 0.92 [0.82, 1.03] (P = .208) and 1.03 [0.87, 1.22] (P = .751), respectively). No substantial differences were observed in Tmax (P = .750) or apparent half‐life (P = .533) of midazolam. Plasma levels of midazolam were not substantially affected by raltegravir, which implies that raltegravir is not a clinically important inducer or inhibitor of CYP 3A4 and that raltegravir would not be expected to affect the pharmacokinetics of other drugs metabolized by CYP 3A4 to a clinically meaningful extent.


Clinical Infectious Diseases | 2008

Atazanavir Modestly Increases Plasma Levels of Raltegravir in Healthy Subjects

Marian Iwamoto; Larissa Wenning; Goutam C. Mistry; Amelia S. Petry; Sarah Y. Liou; Kaylan Ghosh; Sheila Breidinger; Neal Azrolan; Maria J. Gutierrez; William E. Bridson; Julie A. Stone; Keith M. Gottesdiener; John A. Wagner

Raltegravir is an HIV integrase inhibitor that is metabolized through glucuronidation by uridine diphosphate glucuronosyltransferase 1A1, and its use is anticipated in combination with atazanavir (a uridine diphosphate glucuronosyltransferase 1A1 inhibitor). Two pharmacokinetic studies of healthy subjects assessed the effect of multiple-dose atazanavir or ritonavir-boosted atazanavir on raltegravir levels in plasma. Atazanavir and atazanavir plus ritonavir modestly increase plasma levels of raltegravir.


Antimicrobial Agents and Chemotherapy | 2008

Minimal Pharmacokinetic Interaction between the Human Immunodeficiency Virus Nonnucleoside Reverse Transcriptase Inhibitor Etravirine and the Integrase Inhibitor Raltegravir in Healthy Subjects

Matt S. Anderson; Thomas N. Kakuda; William D. Hanley; Jutta L. Miller; James Kost; Randall Stoltz; Larissa Wenning; Julie A. Stone; Richard M. W. Hoetelmans; John A. Wagner; Marian Iwamoto

ABSTRACT Etravirine, a next-generation nonnucleoside reverse transcriptase inhibitor, and raltegravir, an integrase strand transfer inhibitor, have separately demonstrated potent activity in treatment-experienced, human immunodeficiency virus (HIV)-infected patients. An open-label, sequential, three-period study with healthy, HIV-seronegative subjects was conducted to assess the two-way interaction between etravirine and raltegravir for potential coadministration to HIV-infected patients. In period 1, 19 subjects were administered 400 mg raltegravir every 12 h (q12 h) for 4 days, followed by a 4-day washout; in period 2, subjects were administered 200 mg etravirine q12 h for 8 days; and in period 3, subjects were coadministered 400 mg raltegravir and 200 mg etravirine q12 h for 4 days. There was no washout between periods 2 and 3. Doses were administered with a moderate-fat meal. Etravirine had only modest effects on the pharmacokinetics of raltegravir, while raltegravir had no clinically meaningful effect on the pharmacokinetics of etravirine. For raltegravir coadministered with etravirine relative to raltegravir alone, the geometric mean ratio (GMR) and 90% confidence interval (CI) were 0.90 and 0.68 to 1.18, respectively, for the area under the concentration curve from 0 to 12 h (AUC0-12), 0.89 and 0.68 to 1.15, respectively, for the maximum concentration of drug in serum (Cmax), and 0.66 and 0.34 to 1.26, respectively, for the trough drug concentration (C12); the GMR (90% CI) for etravirine coadministered with raltegravir relative to etravirine alone was 1.10 (1.03, 1.16) for AUC0-12, 1.04 (0.97, 1.12) for Cmax, and 1.17 (1.10, 1.26) for C12. All drug-related adverse clinical experiences were mild and generally transient in nature. No grade 3 or 4 adverse experiences or discontinuations due to adverse experiences occurred. Coadministration of etravirine and raltegravir was generally well tolerated; the data suggest that no dose adjustment for either drug is necessary.


Annual Review of Pharmacology and Toxicology | 2009

Emerging Pharmacology: Inhibitors of Human Immunodeficiency Virus Integration

Daria J. Hazuda; Marian Iwamoto; Larissa Wenning

The first integrase inhibitor licensed to treat HIV-1 infection was approved in late 2007, more than a decade after the introduction of the first inhibitors of the HIV-1 reverse transcriptase and protease. The unique biochemical and molecular mechanism of action of this novel class of antiretroviral drugs is the fundamental basis for their activity in treating multidrug-resistant HIV-1 infection and is important for understanding both the cellular and in vivo pharmacology and metabolism of these agents. In addition, available pharmacokinetic and drug interaction data for raltegravir and elvitegravir, the two integrase inhibitors that are the most advanced in clinical development to date, are reviewed.


Clinical Infectious Diseases | 2009

Effects of Omeprazole on Plasma Levels of Raltegravir

Marian Iwamoto; Larissa Wenning; Bach-Yen Nguyen; Hedy Teppler; Allison Moreau; Rand R. Rhodes; William D. Hanley; Bo Jin; Charlotte M. Harvey; Sheila Breidinger; Neal Azrolan; H. Frank Farmer; Robin Isaacs; Jeffery A. Chodakewitz; Julie A. Stone; John A. Wagner

Raltegravir, a human immunodeficiency virus type 1 (HIV-1) integrase inhibitor, has pH-dependent solubility. Raltegravir plasma concentration increases with omeprazole coadministration in healthy subjects; this is likely secondary to an increase in bioavailability attributable to increased gastric pH. Increased gastric pH has been reported in HIV-1-infected individuals, and the effects of omeprazole in this intended population may be diminished. Further investigation is necessary.

Collaboration


Dive into the Marian Iwamoto's collaboration.

Researchain Logo
Decentralizing Knowledge