Network


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

Hotspot


Dive into the research topics where Josee Cote is active.

Publication


Featured researches published by Josee Cote.


The Lancet | 2007

Effect of the cholesteryl ester transfer protein inhibitor, anacetrapib, on lipoproteins in patients with dyslipidaemia and on 24-h ambulatory blood pressure in healthy individuals: two double-blind, randomised placebo-controlled phase I studies

Rajesh Krishna; Matt S. Anderson; Arthur J. Bergman; Bo Jin; Marissa Fallon; Josee Cote; Kim Rosko; Cynthia Chavez-Eng; Ryan Lutz; Daniel M. Bloomfield; Maria del Carmen Gutierrez; James Doherty; Fredrick Bieberdorf; Jeffrey A. Chodakewitz; Keith M. Gottesdiener; John A. Wagner

BACKGROUND The inhibition of cholesteryl ester transfer protein (CETP) is considered a potential new mechanism for treatment of dyslipidaemia. Anacetrapib (MK-0859) is a CETP inhibitor currently under development. We aimed to assess anacetrapibs effects as monotherapy on low-density lipoprotein cholesterol (LDL-C) and high-density lipoprotein cholesterol (HDL-C) and on 24-h ambulatory blood pressure. METHODS We did two double-blind, randomised, placebo-controlled phase I studies. In the first study, 50 patients with dyslipidaemia (LDL-C 100-190 mg/dL; 40 active, 10 placebo) aged 18-75 years received anacetrapib doses of 0, 10, 40, 150, or 300 mg orally once a day with a meal for 28 days. Standard lipid and lipoprotein monitoring, safety monitoring, and anacetrapib concentrations for pharmacokinetics were done. In the second study, 22 healthy participants aged 45-75 years received either 150 mg of anacetrapib once a day or matching placebo with a meal for 10 days in each crossover period, in a randomised sequence, with at least a 14-day washout between the treatment periods. Continuous 24-h ambulatory blood pressure monitoring was done on day -1 and day 10 of each treatment period in this study. The primary or secondary endpoints of safety and tolerability were assessed in both studies by monitoring clinical adverse experiences, physical examinations, vital signs, 12-lead electrocardiogram, and laboratory safety. Analysis was per protocol. These trials are registered with ClinicalTrials.gov, number NCT00565292 and NCT00565006. FINDINGS In the dyslipidaemia study, one patient withdrew consent and one was excluded from the data analysis for HDL-C and LDL-C because complete pre-dose measurements were not available. Anacetrapib produced dose-dependent lipid-altering effects with peak lipid-altering effects of 129% (mean 51.1 [SD 3.8]-114.9 [7.9] mg/dL) increase in HDL-C and a 38% (138.2 [11.4]-77.6 [7.9] mg/dL) decrease in LDL-C in patients with dyslipidaemia. In the 24-h ambulatory blood pressure study in healthy individuals, least squares difference between anacetrapib and placebo groups on day 10 were 0.60 (90% CI -1.54 to 2.74; p=0.634) mm Hg for systolic blood pressure and 0.47 (90% CI -0.90 to 1.84; p=0.561) mm Hg for diastolic blood pressure. INTERPRETATION Anacetrapib seems to exhibit HDL-C increases greater than those seen with other investigational drugs in this class and LDL-C lowering effects similar to statins. Despite greater lipid-altering effects relative to other members of this class, anacetrapib seems not to increase blood pressure, suggesting that potent CETP inhibition by itself might not lead to increased blood pressure.


Proceedings of the National Academy of Sciences of the United States of America | 2007

[18F]MK-9470, a positron emission tomography (PET) tracer for in vivo human PET brain imaging of the cannabinoid-1 receptor

H. Donald Burns; Koen Van Laere; Sandra M. Sanabria-Bohórquez; Terence G. Hamill; Guy Bormans; Wai-si Eng; Ray E Gibson; Christine Ryan; Brett Connolly; Shil Patel; Stephen Krause; Amy Vanko; Anne Van Hecken; Patrick Dupont; Inge De Lepeleire; Paul Rothenberg; S. Aubrey Stoch; Josee Cote; William K. Hagmann; James P. Jewell; Linus S. Lin; Ping Liu; Mark T. Goulet; Keith M. Gottesdiener; John A. Wagner; Jan de Hoon; Luc Mortelmans; Tung M. Fong; Richard Hargreaves

[18F]MK-9470 is a selective, high-affinity, inverse agonist (human IC50, 0.7 nM) for the cannabinoid CB1 receptor (CB1R) that has been developed for use in human brain imaging. Autoradiographic studies in rhesus monkey brain showed that [18F]MK-9470 binding is aligned with the reported distribution of CB1 receptors with high specific binding in the cerebral cortex, cerebellum, caudate/putamen, globus pallidus, substantia nigra, and hippocampus. Positron emission tomography (PET) imaging studies in rhesus monkeys showed high brain uptake and a distribution pattern generally consistent with that seen in the autoradiographic studies. Uptake was blocked by pretreatment with a potent CB1 inverse agonist, MK-0364. The ratio of total to nonspecific binding in putamen was 4–5:1, indicative of a strong specific signal that was confirmed to be reversible via displacement studies with MK-0364. Baseline PET imaging studies in human research subject demonstrated behavior of [18F]MK-9470 very similar to that seen in monkeys, with very good test–retest variability (7%). Proof of concept studies in healthy young male human subjects showed that MK-0364, given orally, produced a dose-related reduction in [18F]MK-9470 binding reflecting CB1R receptor occupancy by the drug. Thus, [18F]MK-9470 has the potential to be a valuable, noninvasive research tool for the in vivo study of CB1R biology and pharmacology in a variety of neuropsychiatric disorders in humans. In addition, it allows demonstration of target engagement and noninvasive dose-occupancy studies to aid in dose selection for clinical trials of CB1R inverse agonists.


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.


Clinical Pharmacology & Therapeutics | 2008

Multiple-dose pharmacodynamics and pharmacokinetics of anacetrapib, a potent cholesteryl ester transfer protein (CETP) inhibitor, in healthy subjects.

Rajesh Krishna; Arthur J. Bergman; Bo Jin; M Fallon; Josee Cote; P Van Hoydonck; Tine Laethem; Isaias Noel Gendrano; K Van Dyck; Deborah Hilliard; O Laterza; Karen Snyder; Cynthia Chavez-Eng; Ryan Lutz; J Chen; Dm Bloomfield; M De Smet; L. Van Bortel; Maria J. Gutierrez; Nidal Al-Huniti; Kevin Dykstra; Keith M. Gottesdiener; John A. Wagner

Cholesteryl ester transfer protein (CETP) is a plasma protein that catalyzes the heteroexchange of cholesteryl esters from high‐density lipoprotein (HDL) and triglycerides to apolipoprotein B–containing lipoproteins, especially very low–density lipoproteins (LDL‐C). 1, 2


Science Translational Medicine | 2012

Niacin Lipid Efficacy Is Independent of Both the Niacin Receptor GPR109A and Free Fatty Acid Suppression

Brett Lauring; Andrew K.P. Taggart; James R. Tata; Richard L. Dunbar; Luzelena Caro; Kang Cheng; Jayne Chin; Steven L. Colletti; Josee Cote; Sauzanne Khalilieh; Jiajun Liu; Wen-Lin Luo; Alexandra MacLean; Laurence B. Peterson; Adam B. Polis; Waheeda Sirah; Tsuei-Ju Wu; Xuan Liu; Lan Jin; Kenneth K. Wu; P. Douglas Boatman; Graeme Semple; Dominic P. Behan; Daniel T. Connolly; Eseng Lai; John A. Wagner; Samuel D. Wright; Cynthia Cuffie; Yale B. Mitchel; Daniel J. Rader

GPR109A is not the target mediating niacin’s lipid efficacy and the free fatty acid hypothesis does not explain niacin’s mechanism of action. Breaking Free of the “FFA Hypothesis” Free fatty acids (FFAs) appear in the blood plasma after a meal. Niacin—a vitamin that helps to regulate lipid levels in the body—is given to patients to reduce the amount of FFAs. It also works to raise “good” cholesterol [high-density lipoprotein (HDL)] and lower both “bad” cholesterol [low-density lipoprotein (LDL)] and triglycerides. The “FFA hypothesis” suggests that niacin works to exert these beneficial lipid effects by limiting the amount of FFAs available to synthesize triglycerides. Lauring, Taggart, and colleagues now challenge this long-standing theory. In studies in mice and humans, the authors debunk the hypothesis, showing that the effect on HDL, LDL, and triglycerides is not directly linked to FFAs. To study the lipid-modifying effects of niacin (nicotinic acid), Lauring et al. used a genetic, humanized mouse model lacking the LDL receptor. In these animals, niacin increased HDL cholesterol levels, as expected. Lack of GPR109A in these animals blocked the anti-lipolytic effect of nicotinic acid on FFAs but had no effect on drug-related changes in plasma HDL and LDL cholesterol or triglyceride levels. Treatment of the mice with a GPR109A agonist, MK-1903, also caused an anti-lipolytic effect but did not affect levels of triglyceride or LDL and HDL cholesterol. Together, these in vivo preclinical studies suggest that niacin works to lower FFAs through GPR109A but has an independent mechanism of action on other lipids. The authors addressed the role of GPR109A in humans by testing the effects of MK-1903 and of another synthetic GPR109A agonist in clinical trials. Both agonists affected FFA lipolysis but had only minor effects on HDL cholesterol and triglyceride levels in patients, thus mirroring results seen in animals and showing that niacin works independently of GPR109A to modify dyslipidemia. The studies by Lauring et al. point to a new, yet-uncovered mechanism of action for niacin’s beneficial effects on lipids in the blood. Despite overturning the FFA hypothesis and potentially redirecting drug development away from GPR109A agonists, niacin could still be useful for treating other diseases in patients, including atherosclerosis and inflammation, where GPR109A plays a major role in cell signaling. Nicotinic acid (niacin) induces beneficial changes in serum lipoproteins and has been associated with beneficial cardiovascular effects. Niacin reduces low-density lipoprotein, increases high-density lipoprotein, and decreases triglycerides. It is well established that activation of the seven-transmembrane Gi-coupled receptor GPR109A on Langerhans cells results in release of prostaglandin D2, which mediates the well-known flushing side effect of niacin. Niacin activation of GPR109A on adipocytes also mediates the transient reduction of plasma free fatty acid (FFA) levels characteristic of niacin, which has been long hypothesized to be the mechanism underlying the changes in the serum lipid profile. We tested this “FFA hypothesis” and the hypothesis that niacin lipid efficacy is mediated via GPR109A by dosing mice lacking GPR109A with niacin and testing two novel, full GPR109A agonists, MK-1903 and SCH900271, in three human clinical trials. In mice, the absence of GPR109A had no effect on niacin’s lipid efficacy despite complete abrogation of the anti-lipolytic effect. Both MK-1903 and SCH900271 lowered FFAs acutely in humans; however, neither had the expected effects on serum lipids. Chronic FFA suppression was not sustainable via GPR109A agonism with niacin, MK-1903, or SCH900271. We conclude that the GPR109A receptor does not mediate niacin’s lipid efficacy, challenging the long-standing FFA hypothesis.


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.


British Journal of Clinical Pharmacology | 2009

Single‐dose pharmacokinetics and pharmacodynamics of anacetrapib, a potent cholesteryl ester transfer protein (CETP) inhibitor, in healthy subjects

Rajesh Krishna; Amit Garg; Deborah Panebianco; Josee Cote; Arthur J. Bergman; Pascale Van Hoydonck; Tine Laethem; Kristien Van Dyck; Jingjing Chen; Cynthia Chavez-Eng; Laura Archer; Ryan Lutz; Deborah Hilliard; Karen Snyder; Bo Jin; Luc Van Bortel; Kenneth C. Lasseter; Nidal Al-Huniti; Kevin Dykstra; Keith M. Gottesdiener; John A. Wagner

AIMS Anacetrapib is an orally active and potent inhibitor of CETP in development for the treatment of dyslipidaemia. These studies endeavoured to establish the safety, tolerability, pharmacokinetics and pharmacodynamics of rising single doses of anacetrapib, administered in fasted or fed conditions, and to preliminarily assess the effect of food, age, gender and obesity on the single-dose pharmacokinetics and pharmacodynamics of anacetrapib. METHODS Safety, tolerability, anacetrapib concentrations and CETP activity were evaluated. RESULTS Anacetrapib was rapidly absorbed, with peak concentrations occurring at approximately 4 h post-dose and an apparent terminal half-life ranging from approximately 9 to 62 h in the fasted state and from approximately 42 to approximately 83 h in the fed state. Plasma AUC and C(max) appeared to increase in a less than approximately dose-dependent manner in the fasted state, with an apparent plateau in absorption at higher doses. Single doses of anacetrapib markedly and dose-dependently inhibited serum CETP activity with peak effects of approximately 90% inhibition at t(max) and approximately 58% inhibition at 24 h post-dose. An E(max) model best described the plasma anacetrapib concentration vs CETP activity relationship with an EC(50) of approximately 22 nm. Food increased exposure to anacetrapib; up to approximately two-three-fold with a low-fat meal and by up to approximately six-eight fold with a high-fat meal. Anacetrapib pharmacokinetics and pharmacodynamics were similar in elderly vs young adults, women vs men, and obese vs non-obese young adults. Anacetrapib was well tolerated and was not associated with any meaningful increase in blood pressure. CONCLUSIONS Whereas food increased exposure to anacetrapib significantly, age, gender and obese status did not meaningfully influence anacetrapib pharmacokinetics and pharmacodynamics.


The Journal of Clinical Pharmacology | 2007

Effect of a Single Cyclosporine Dose on the Single‐Dose Pharmacokinetics of Sitagliptin (MK‐0431), a Dipeptidyl Peptidase‐4 Inhibitor, in Healthy Male Subjects

Rajesh Krishna; Arthur J. Bergman; Patrick Larson; Josee Cote; Kenneth C. Lasseter; Stacey Dilzer; Amy Wang; Wei Zeng; Li Chen; John A. Wagner; Gary A. Herman

Sitagliptin (MK‐0431) is an orally active, potent, and selective dipeptidyl peptidase‐4 inhibitor used for the treatment of patients with type 2 diabetes mellitus. Sitagliptin has been shown to be a substrate for P‐glycoprotein in preclinical studies. Cyclosporine was used as a probe P‐glycoprotein inhibitor at a high dose to evaluate the potential effect of potent P‐glycoprotein inhibition on single‐dose sitagliptin pharmacokinetics in healthy male subjects. Eight healthy young men received a single oral 600‐mg dose of cyclosporine with a single 100‐mg oral sitagliptin dose and a single oral 100‐mg sitagliptin dose alone in an open‐label, randomized, 2‐period, crossover study. Single doses of sitagliptin with or without single doses of cyclosporine were generally well tolerated. The sitagliptin AUC0‐∞ geometric mean ratio was 1.29 with a 90% confidence interval of (1.24, 1.34). The sitagliptin Cmax geometric mean ratio was 1.68 with a 90% confidence interval of (1.35, 2.08). Cyclosporine coadministration did not appear to affect apparent sitagliptin renal clearance, t1/2, or C24 h, suggesting that effects of these high doses of cyclosporine are more likely due to enhanced absorption of sitagliptin, potentially through inhibition of intestinal P‐glycoprotein. These results rationalize the use of a single high‐dose cyclosporine as a probe inhibitor of P‐glycoprotein for compound candidates whose elimination is less dependent on CYP3A4‐mediated metabolism.


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

Assessment of the CYP3A‐Mediated Drug Interaction Potential of Anacetrapib, a Potent Cholesteryl Ester Transfer Protein (CETP) Inhibitor, in Healthy Volunteers

Rajesh Krishna; Arthur J. Bergman; Bo Jin; Amit Garg; Brad Roadcap; Rita Chiou; James Dru; Josee Cote; Tine Laethem; Regina W. Wang; Varsha Didolkar; Eva Vets; Keith M. Gottesdiener; John A. Wagner

In this study, midazolam was used as a probe‐sensitive CYP3A substrate to investigate the effect of anacetrapib on CYP3A activity, and ketoconazole was used as a probe‐inhibitor to investigate the effect of potent CYP3A inhibition on the pharmacokinetics of anacetrapib, a novel cholesteryl ester transfer protein inhibitor in development for the treatment of dyslipidemia. Two partially blinded, randomized, 2‐period, fixed‐sequence studies were performed. Safety, tolerability, and midazolam and anacetrapib plasma concentrations were assessed. All treatments were generally well tolerated. The geometric mean ratios (90% confidence interval) of midazolam with anacetrapib/midazolam alone for AUC0‐∞ and Cmax were 1.04 (0.94, 1.14) and 1.15 (0.97, 1.37), respectively. Exposure to anacetrapib was increased by ketoconazole—specifically, the geometric mean ratios (90% confidence interval) of anacetrapib with ketoconazole/anacetrapib alone for AUC0‐∞ and Cmax were 4.58 (3.68, 5.71) and 2.37 (2.02, 2.78), respectively. The study showed that anacetrapib does not inhibit or induce CYP3A activity. Furthermore, anacetrapib appears to be a moderately sensitive substrate of CYP3A.

Collaboration


Dive into the Josee Cote'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

Richard Hargreaves

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Wai-si Eng

United States Military Academy

View shared research outputs
Researchain Logo
Decentralizing Knowledge