Network


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

Hotspot


Dive into the research topics where Carol Addy is active.

Publication


Featured researches published by Carol Addy.


Cell Metabolism | 2008

The acyclic CB1R inverse agonist taranabant mediates weight loss by increasing energy expenditure and decreasing caloric intake

Carol Addy; Hamish Wright; Koen Van Laere; Ira Gantz; Ngozi Erondu; Bret Musser; Kaifeng Lu; Jinyu Yuan; Sandra M. Sanabria-Bohórquez; Aubrey Stoch; Cathy Stevens; Tung M. Fong; Inge De Lepeleire; Caroline Cilissen; Josee Cote; Kim Rosko; Isaias Noel Gendrano; Allison Martin Nguyen; Barry Gumbiner; Paul Rothenberg; Jan de Hoon; Guy Bormans; Marleen Depré; Wai-si Eng; Eric Ravussin; Samuel Klein; John E. Blundell; Gary A. Herman; H. Donald Burns; Richard Hargreaves

Cannabinoid 1 receptor (CB1R) inverse agonists are emerging as a potential obesity therapy. However, the physiological mechanisms by which these agents modulate human energy balance are incompletely elucidated. Here, we describe a comprehensive clinical research study of taranabant, a structurally novel acyclic CB1R inverse agonist. Positron emission tomography imaging using the selective CB1R tracer [(18)F]MK-9470 confirmed central nervous system receptor occupancy levels ( approximately 10%-40%) associated with energy balance/weight-loss effects in animals. In a 12-week weight-loss study, taranabant induced statistically significant weight loss compared to placebo in obese subjects over the entire range of evaluated doses (0.5, 2, 4, and 6 mg once per day) (p < 0.001). Taranabant treatment was associated with dose-related increased incidence of clinical adverse events, including mild to moderate gastrointestinal and psychiatric effects. Mechanism-of-action studies suggest that engagement of the CB1R by taranabant leads to weight loss by reducing food intake and increasing energy expenditure and fat oxidation.


The Journal of Clinical Pharmacology | 2008

Multiple-Dose Pharmacokinetics, Pharmacodynamics, and Safety of Taranabant, a Novel Selective Cannabinoid-1 Receptor Inverse Agonist, in Healthy Male Volunteers

Carol Addy; Paul Rothenberg; Susie Li; Anup Majumdar; Nancy G. B. Agrawal; Hankun Li; Ling Zhong; Jinyu Yuan; Andrea Maes; Stephanie Dunbar; Josee Cote; Kim Rosko; Kristien Van Dyck; Inge De Lepeleire; Jan de Hoon; Anne Van Hecken; Marleen Depré; Annemie Knops; Keith M. Gottesdiener; Aubrey Stoch; John A. Wagner

Taranabant is a cannabinoid‐1 receptor inverse agonist for the treatment of obesity. This study evaluated the safety, pharmacokinetics, and pharmacodynamics of taranabant (5, 7.5, 10, or 25 mg once daily for 14 days) in 60 healthy male subjects. Taranabant was rapidly absorbed, with a median tmax of 1.0 to 2.0 hours and a t1/2 of approximately 74 to 104 hours. Moderate accumulation was observed in Cmax (1.18‐ to 1.40‐fold) and AUC0–24 h (1.5‐ to 1.8‐fold) over 14 days for the 5‐, 7.5‐, and 10‐mg doses, with an accumulation half‐life ranging from 15 to 21 hours. Steady state was reached after 13 days. After multiple‐dose administration, plasma AUC0–24 h and Cmax of taranabant increased dose proportionally (5–10 mg) and increased somewhat less than dose proportionally for 25 mg. Taranabant was generally well tolerated up to doses of 10 mg and exhibited multiple‐dose pharmacokinetics consistent with once‐daily dosing.


The Journal of Clinical Pharmacology | 2008

Safety, Tolerability, Pharmacokinetics, and Pharmacodynamic Properties of Taranabant, a Novel Selective Cannabinoid‐1 Receptor Inverse Agonist, for the Treatment of Obesity: Results From a Double‐Blind, Placebo‐Controlled, Single Oral Dose Study in Healthy Volunteers

Carol Addy; Susie Li; Nancy G. B. Agrawal; Julie A. Stone; Anup Majumdar; Ling Zhong; Hankun Li; Jinyu Yuan; Andrea Maes; Paul Rothenberg; Josee Cote; Kim Rosko; Corinne Cummings; Steven J. Warrington; Malcolm Boyce; Keith M. Gottesdiener; Aubrey Stoch; John A. Wagner

Taranabant is a novel cannabinoid CB‐1 receptor (CB1R) inverse agonist in clinical development for the treatment of obesity. This double‐blind, randomized, placebo‐controlled, single oral dose study evaluated the safety, tolerability, pharmacokinetics, and pharmacodynamics of taranabant (0.5–600 mg) in 24 healthy male volunteers. Single‐dose AUC0‐∞ and Cmax values for taranabant increased approximately linearly ith dose up to 200 mg, with slightly less than dose‐proportional increases in AUC0‐∞ and Cmax values for doses >200 mg. Plasma taranabant had a biphasic disposition, with a median tmax of 1 to 2.5 hours and a terminal elimination tl/2 of 38 to 69 hours. Coadministration of taranabant with a high‐fat meal led to a 14% increase in Cmax and a 74% increase in AUC0‐∞, Clinical adverse experiences ssociated with single doses of taranabant were generally mild and transient. Of the 198 clinical adverse experiences reported, the most common drug‐related ones were nausea (36), headache (22), drowsiness (14), abdominal discomfort/abdominal pain/stomachache (14), hiccups (9), dizziness (8), decreased appetite (7), increased bowel movement (7), mood change (6), tiredness (4), vomiting (4), and sweating increased (4). Taranabant has pharmacokinetic characteristics suitable for a once‐daily dosing regimen.


The Journal of Clinical Pharmacology | 2009

Single-Dose Administration of MK-0657, an NR2B-Selective NMDA Antagonist, Does Not Result in Clinically Meaningful Improvement in Motor Function in Patients With Moderate Parkinson's Disease

Carol Addy; Chris Assaid; David Hreniuk; Mark Stroh; Yang Xu; W. Joseph Herring; Aaron Ellenbogen; H. A. Jinnah; Louis Kirby; Mark Leibowitz; R. Malcolm Stewart; Daniel Tarsy; James Tetrud; S. Aubrey Stoch; Keith M. Gottesdiener; John A. Wagner

The glutamatergic system is thought to contribute to the motor disturbances observed in Parkinsons disease. Blockade of glutamatergic activity by a selective antagonist of the NR2B subunit of the N‐methyl‐D‐aspartate (NMDA) receptor is associated with improvement in motor symptoms in a preclinical model of Parkinsons disease. A randomized, double‐blind, double‐dummy, placebo‐controlled, 3‐period crossover study was conducted in patients with moderate Parkinsons disease to evaluate the pharmacologic activity of MK‐0657, an NR2B‐selctive NMDA receptor antagonist. Patients (n = 16) received single oral doses of MK‐0657 7 mg, carbidopa/levodopa 25/250 mg (LD) as a positive control, and placebo, after which motor function was serially evaluated by means of the Unified Parkinsons Disease Rating Scale—Motor Examination (UPDRS‐ME). LD administration resulted in significant improvement in the UPDRS‐ME relative to placebo (P = .025), confirming the sensitivity of the test paradigm; however, the UPDRS‐ME change following MK‐0657 administration showed no improvement compared with placebo (P = .110) despite exceeding the target MK‐0657 plasma concentration of 400 nM. Although the administration of MK‐0657 was generally well tolerated, it was associated with increases in systolic and diastolic blood pressure relative to placebo. The results of this study do not support ongoing clinical development of MK‐0657 as a novel monotherapy for Parkinsons disease.


Obesity | 2007

NPY5R antagonism does not augment the weight loss efficacy of orlistat or sibutramine.

Ngozi Erondu; Carol Addy; Kaifeng Lu; Madhuja Mallick; Bret Musser; Ira Gantz; Joseph Proietto; Arne Astrup; Søren Toubro; Aila M. Rissannen; Serena Tonstad; William G. Haynes; Keith M. Gottesdiener; Keith D. Kaufman; John M. Amatruda; Steven B. Heymsfield

Objective: Central counter‐regulatory mechanisms, including those related to the orexigenic hormone neuropeptide Y (NPY), may limit the weight loss observed with conventional pharmacological monotherapy. This study evaluated whether blockade of the NPY Y5 receptor (NPY5R) with the selective antagonist MK‐0557 potentiates sibutramine and orlistat weight loss effects.


The Journal of Clinical Pharmacology | 2016

Pharmacokinetics and Pharmacodynamics of Omarigliptin, a Once-weekly Dipeptidyl Peptidase-4 (DPP-4) Inhibitor, after Single and Multiple Doses in Healthy Subjects.

Rajesh Krishna; Carol Addy; Daniel Tatosian; Xiaoli S. Glasgow; Isaias Noel Gendrano; Martine Robberechts; Wouter Haazen; Jn de Hoon; Marleen Depré; Ashley Martucci; Joanna Z. Peng; Amy O. Johnson-Levonas; John A. Wagner; S. Aubrey Stoch

The pharmacokinetics (PK) and pharmacodynamics (PD) of omarigliptin, a novel once‐weekly DPP‐4 inhibitor, were assessed following single and multiple doses in healthy subjects. Absorption was rapid, and food did not influence single‐dose PK. Accumulation was minimal, and steady state was reached after 2 to 3 weeks. Weekly (area under the curve) AUC and Cmax displayed dose proportionality within the dose range studied at steady state. The average renal clearance of omarigliptin was ∼2 L/h. DPP‐4 inhibition ranged from ∼77% to 89% at 168 hours following the last of 3 once‐weekly doses over the dose range studied. Omarigliptin resulted in ∼2‐fold increases in weighted average postprandial active GLP‐1. Omarigliptin acts by stabilizing active GLP‐1, which is consistent with its mechanism of action as a DPP‐4 inhibitor. Administration of omarigliptin was generally well tolerated in healthy subjects, and both the PK and PD profiles support once‐weekly dosing. A model‐based assessment of QTc interval risk from the single ascending dose study predicted a low risk of QTc prolongation within the likely clinical dose range, a finding later confirmed in a thorough QT trial.


Journal of Clinical Psychopharmacology | 2012

Human Abuse Potential and Cognitive Effects of Taranabant, a Cannabinoid 1 Receptor Inverse Agonist: A Randomized, Double-Blind, Placebo- and Active-Controlled, Crossover Study in Recreational Polydrug Users

Kerri A. Schoedel; Carol Addy; Bijan Chakraborty; Kim Rosko; Stephanie Dunbar; Andrea Maes; Nancy Chen; Selwyn Aubrey Stoch; John G. Wagner; Jeff Chodakewitz; Edward M. Sellers

Introduction Taranabant is a cannabinoid 1 receptor inverse agonist that was in development for treatment of obesity. Because of central nervous system effects, the study was performed to assess the abuse potential and cognitive effects of taranabant in recreational polydrug users compared with phentermine, dronabinol, and placebo. Methods Stimulant- and cannabis-experienced polydrug users (N = 30) were randomized in a double-blind crossover study to receive taranabant 2, 4, 10, and 20 mg; phentermine 45 and 90 mg; dronabinol 20 mg; and placebo. Subjective and neurocognitive measures were administered for 24 hours, and peak/peak change from baseline effects were analyzed using a linear mixed-effects model. Results Phentermine 45 and 90 mg showed abuse-related subjective effects versus placebo, including drug liking, overall drug liking, and other positive/stimulant effects, whereas dronabinol 20 mg showed abuse-related positive, cannabis-like, and sedative effects. Taranabant was not significantly different from placebo on most of the subjective measures other than negative/dysphoric effects at the highest dose, and its effects were significantly less pronounced relative to phentermine and dronabinol on most measures. Phentermine improved cognitive/motor performance and dronabinol impaired motor/cognitive performance on some measures, whereas taranabant 4 and 20 mg had minor impairment effects on manual tracking. Conclusions The phentermine and dronabinol results demonstrate the validity and sensitivity of the study. Taranabant did not consistently show stimulant/cannabis-like effects or abuse potential in recreational polydrug users, indicating that cannabinoid 1 receptor inverse agonists/antagonists are unlikely to be recreationally abused.


The Journal of Clinical Pharmacology | 2009

Pharmacokinetics, Safety, and Tolerability of Phentermine in Healthy Participants Receiving Taranabant, a Novel Cannabinoid‐1 Receptor (CB1R) Inverse Agonist

Carol Addy; Patricia Jumes; Kimberly Rosko; Susie Li; Hankun Li; Andrea Maes; Amy O. Johnson-Levonas; Jeffrey A. Chodakewitz; S. Aubrey Stoch; John A. Wagner

This study assessed the potential pharmacokinetic interaction and safety/tolerability of taranabant and phentermine coadministration. This was a randomized, double‐blind, 3‐panel, fixed‐sequence study in healthy participants. Panels A, B, and C evaluated the safety/tolerability of phentermine 15 mg coadministered with taranabant 0.5, 1, and 2 mg for 7 days (panel A) and 28 days (panels B and C). In panels A and C, phentermine 15 mg was administered both with (7 days, panel A; 28 days, panel C) and without (7 days) taranabant 0.5 mg or 2 mg to evaluate pharmacokinetics. The primary endpoint was phentermine AUC0–24 h in panels A and C. Secondary endpoints were changes from baseline in blood pressure and heart rate for all panels. The geometric mean ratios and 90% confidence intervals for phentermine AUC0–24 h in the presence/absence of taranabant 0.5 mg and 2 mg were 1.08 (0.99, 1.17) and 1.04 (0.98, 1.10), respectively. No significant differences in blood pressure and heart rate were observed with any treatment versus placebo. Coadministration of taranabant 0.5 mg, 1 mg, and 2 mg with phentermine was well tolerated with no pharmacokinetic interaction and did not result in meaningful changes in blood pressure or heart rate versus placebo.


Journal of Diabetes Investigation | 2017

Single and multiple dose pharmacokinetics and pharmacodynamics of omarigliptin, a novel, once‐weekly dipeptidyl peptidase‐4 inhibitor, in healthy Japanese men

Saori Tsuchiya; Evan J. Friedman; Carol Addy; Akira Wakana; Daniel Tatosian; Yuki Matsumoto; Hideyo Suzuki; Eunkyung Kauh

Omarigliptin is a novel, potent, long‐acting oral dipeptidyl peptidase‐4 inhibitor being developed as a once‐weekly (q.w.) treatment for type 2 diabetes mellitus patients, with 25 mg and 12.5 mg tablets recently being approved as market formulations in Japan.


Clinical Therapeutics | 2016

Pharmacokinetic and Pharmacodynamic Effects of Multiple-dose Administration of Omarigliptin, a Once-weekly Dipeptidyl Peptidase-4 Inhibitor, in Obese Participants With and Without Type 2 Diabetes Mellitus.

Carol Addy; Daniel Tatosian; Xiaoli S. Glasgow; Isaias Noel Gendrano; Eunkyung Kauh; Ashley Martucci; Amy O. Johnson-Levonas; Diana Selverian; Catherine Z. Matthews; Marie Gutierrez; John A. Wagner; S. Aubrey Stoch

PURPOSE Omarigliptin (MK-3102) is a potent, oral, long-acting dipeptidyl peptidase (DPP)-4 inhibitor approved in Japan and in global development as a once-weekly treatment for type 2 diabetes mellitus (T2DM). The aim of this study was to investigate the pharmacokinetic (PK) and pharmacodynamic (PD) effects of omarigliptin in obese participants with and without T2DM. METHODS This was a Phase I, randomized, double-blind, placebo-controlled, multiple-dose study of 50-mg omarigliptin administered once weekly for 4 weeks. Participants included 24 obese but otherwise healthy subjects (panel A; omarigliptin, n = 18; placebo, n = 6) and 8 obese patients with T2DM (treatment naive, hemoglobin A1c ≥ 6.5% and ≤ 10.0% [panel B]; omarigliptin, n = 6; placebo, n = 2). Participants were 45 to 65 years of age with a body mass index of ≥ 30 and ≤ 40 kg/m(2). Blood sampling occurred at select time points, depending on the study panel, to evaluate the PK properties of omarigliptin, DPP-4 activity, active glucagon-like peptide 1 levels, and plasma glucose concentrations. Body weight was an exploratory end point. Due to sparse sampling in panel A, a thorough PK analysis was performed in obese patients with T2DM (panel B) only. PD analyses were performed in the overall study population (pooled panels A and B). FINDINGS PK profiles in obese participants with and without T2DM were similar to those observed in nonobese reference subjects (historical data). Steady state was achieved after 1 or 2 weekly doses in obese participants with and without T2DM. In obese patients with T2DM, omarigliptin was rapidly absorbed, with a median Tmax of 1 to 2.5 hours (days 1 and 22). Compared with those in reference subjects, the geometric mean ratios (95% CI) (Obese T2DM/reference) for steady-state plasma AUC0-168h, Cmax, and C168h were 0.80 (0.65-0.98), 0.86 (0.53-1.41), and 1.08 (0.88-1.33), respectively. Trough DPP-4 activity was inhibited by ~90%; postprandial (PP) 4-hour weighted mean active GLP-1 concentrations were increased ~2-fold; and PP glucose was significantly reduced with omarigliptin versus placebo in the pooled population. Omarigliptin was generally well-tolerated in the pooled population, and there were no hypoglycemic events. Consistent with other DPP-4 inhibitors, omarigliptin had no effect on body weight in this short-duration study. IMPLICATIONS The administration of omarigliptin was generally well-tolerated in obese participants with and without T2DM, and the favorable PK and PD profiles support once-weekly dosing. Omarigliptin may provide an important once-weekly treatment option for patients with T2DM. ClinicalTrials.gov identifier: NCT01088711.

Collaboration


Dive into the Carol Addy's collaboration.

Top Co-Authors

Avatar

John A. Wagner

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Paul Rothenberg

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Wai-si Eng

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Marleen Depré

Katholieke Universiteit Leuven

View shared research outputs
Researchain Logo
Decentralizing Knowledge