Network


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

Hotspot


Dive into the research topics where Maurice Emery is active.

Publication


Featured researches published by Maurice Emery.


Proceedings of the National Academy of Sciences of the United States of America | 2009

A proprotein convertase subtilisin/kexin type 9 neutralizing antibody reduces serum cholesterol in mice and nonhuman primates

Joyce Chi Yee Chan; Derek E. Piper; Qiong Cao; Dongming Liu; Chadwick Terence King; Wei Wang; Jie Tang; Qiang Liu; Jared Higbee; Zhen Xia; Yongmei Di; Susan Shetterly; Ziva Arimura; Heather Salomonis; William G. Romanow; Stephen T. Thibault; Richard Zhang; Ping Cao; Xiaoping Yang; Timothy Yu; Mei Lu; Marc W. Retter; Gayle Kwon; Kirk Henne; Oscar Pan; Mei-Mei Tsai; Bryna Fuchslocher; Evelyn Yang; Lei Zhou; Ki Jeong Lee

Proprotein convertase subtilisin/kexin type 9 (PCSK9) regulates serum LDL cholesterol (LDL-C) by interacting with the LDL receptor (LDLR) and is an attractive therapeutic target for LDL-C lowering. We have generated a neutralizing anti-PCSK9 antibody, mAb1, that binds to an epitope on PCSK9 adjacent to the region required for LDLR interaction. In vitro, mAb1 inhibits PCSK9 binding to the LDLR and attenuates PCSK9-mediated reduction in LDLR protein levels, thereby increasing LDL uptake. A combination of mAb1 with a statin increases LDLR levels in HepG2 cells more than either treatment alone. In wild-type mice, mAb1 increases hepatic LDLR protein levels ≈2-fold and lowers total serum cholesterol by up to 36%: this effect is not observed in LDLR−/− mice. In cynomolgus monkeys, a single injection of mAb1 reduces serum LDL-C by 80%, and a significant decrease is maintained for 10 days. We conclude that anti-PCSK9 antibodies may be effective therapeutics for treating hypercholesterolemia.


Journal of the American College of Cardiology | 2012

Effects of AMG 145 on low-density lipoprotein cholesterol levels: results from 2 randomized, double-blind, placebo-controlled, ascending-dose phase 1 studies in healthy volunteers and hypercholesterolemic subjects on statins.

Clapton Dias; Adam Shaywitz; Scott M. Wasserman; Brian Smith; Bing Gao; Dina Stolman; Caroline Crispino; Karen Smirnakis; Maurice Emery; Alexander Colbert; John P. Gibbs; Marc W. Retter; Blaire Cooke; Stephen Uy; Mark Matson; Evan A. Stein

OBJECTIVES The aim of this study was to evaluate the safety, tolerability, and effects of AMG 145 on low-density lipoprotein cholesterol (LDL-C) in healthy and hypercholesterolemic subjects on statin therapy. BACKGROUND Proprotein convertase subtilisin/kexin type 9 (PCSK9) down-regulates surface expression of the low-density lipoprotein receptor (LDL-R), increasing serum LDL-C. AMG 145, a fully human monoclonal antibody to PCSK9, prevents PCSK9/LDL-R interaction, restoring LDL-R recycling. METHODS Healthy adults (phase 1a) were randomized to 1 dose of AMG 145: 7, 21, 70, 210, or 420 mg SC; 21 or 420 mg IV; or matching placebo. Hypercholesterolemic adults (phase 1b) receiving low- to moderate-dose statins were randomized to multiple SC doses of AMG 145: 14 or 35 mg once weekly (QW) ×6, 140 or 280 mg every 2 weeks (Q2W) ×3, 420 mg every 4 weeks ×2, or matching placebo. Eleven subjects receiving high-dose statins and 6 subjects with heterozygous familial hypercholesterolemia were randomized to SC AMG 145 140 mg or placebo Q2W ×3. RESULTS In the trials (AMG 145 n = 85, placebo n = 28), AMG 145 reduced LDL-C up to 64% (p < 0.0001) versus placebo after 1 dose ≥21 mg and up to 81% (p < 0.001) with repeated doses ≥35 mg QW. No serious adverse events (AEs) occurred. Overall incidence of treatment-emergent AEs was similar in AMG 145 versus placebo groups: 69% versus 71% (phase 1a); 65% versus 64% (phase 1b). CONCLUSIONS In phase 1 studies, AMG 145 significantly reduced serum LDL-C in healthy and hypercholesterolemic statin-treated subjects, including those with heterozygous familial hypercholesterolemia or taking the highest doses of atorvastatin or rosuvastatin, with an overall AE profile similar to placebo.


Journal of Medicinal Chemistry | 2010

Discovery of a Potent, Orally Active 11β-Hydroxysteroid Dehydrogenase Type 1 Inhibitor for Clinical Study: Identification of (S)-2-((1S,2S,4R)-Bicyclo[2.2.1]heptan-2-ylamino)-5-isopropyl-5-methylthiazol-4(5H)-one (AMG 221)

Murielle M. Véniant; Clarence Hale; Randall W. Hungate; Kyung Gahm; Maurice Emery; Janan Jona; Smriti Joseph; Jeffrey Adams; Andrew Hague; George A. Moniz; Jiandong Zhang; Michael D. Bartberger; Vivian Li; Rashid Syed; Steven R. Jordan; Renee Komorowski; Michelle Chen; Rod Cupples; Ki Won Kim; David J. St. Jean; Lars Johansson; Martin Henriksson; Meredith Williams; Jerk Vallgarda; Christopher Fotsch; Minghan Wang

Thiazolones with an exo-norbornylamine at the 2-position and an isopropyl group on the 5-position are potent 11beta-HSD1 inhibitors. However, the C-5 center was prone to epimerization in vitro and in vivo, forming a less potent diastereomer. A methyl group was added to the C-5 position to eliminate epimerization, leading to the discovery of (S)-2-((1S,2S,4R)-bicyclo[2.2.1]heptan-2-ylamino)-5-isopropyl-5-methylthiazol-4(5H)-one (AMG 221). This compound decreased fed blood glucose and insulin levels and reduced body weight in diet-induced obesity mice.


Journal of Medicinal Chemistry | 2008

2-amino-1,3-thiazol-4(5H)-ones as potent and selective 11beta-hydroxysteroid dehydrogenase type 1 inhibitors: enzyme-ligand co-crystal structure and demonstration of pharmacodynamic effects in C57Bl/6 mice.

Lars Johansson; Christopher Fotsch; Michael D. Bartberger; Victor M. Castro; Michelle Chen; Maurice Emery; Sonja Gustafsson; Clarence Hale; Dean Hickman; Evert Homan; Steven R. Jordan; Renee Komorowski; Aiwen Li; Kenneth McRae; George A. Moniz; Guy Matsumoto; Carlos Orihuela; Gunnar Palm; Murielle M. Véniant; Minghan Wang; Meredith Williams; Jiandong Zhang

11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) has attracted considerable attention during the past few years as a potential target for the treatment of diseases associated with metabolic syndrome. In our ongoing work on 11beta-HSD1 inhibitors, a series of new 2-amino-1,3-thiazol-4(5 H)-ones were explored. By inserting various cycloalkylamines at the 2-position and alkyl groups or spirocycloalkyl groups at the 5-position of the thiazolone, several potent 11beta-HSD1 inhibitors were identified. An X-ray cocrystal structure of human 11beta-HSD1 with compound 6d (Ki=28 nM) revealed a large lipophilic pocket accessible by substitution off the 2-position of the thiazolone. To increase potency, analogues were prepared with larger lipophilic groups at this position. One of these compounds, the 3-noradamantyl analogue 8b, was a potent inhibitor of human 11beta-HSD1 (Ki=3 nM) and also inhibited 11beta-HSD1 activity in lean C57Bl/6 mice when evaluated in an ex vivo adipose and liver cortisone to cortisol conversion assay.


The Journal of Clinical Pharmacology | 2011

Population Pharmacokinetic/Pharmacodynamic Model of Subcutaneous Adipose 11β‐Hydroxysteroid Dehydrogenase Type 1 (11β‐HSD1) Activity After Oral Administration of AMG 221, a Selective 11β‐HSD1 Inhibitor

John P. Gibbs; Maurice Emery; Ian McCaffery; Brian G. Smith; Megan A. Gibbs; Anna Akrami; John M. Rossi; Katherine Paweletz; Marc R. Gastonguay; Edgar Bautista; Minghan Wang; Riccardo Perfetti; Oranee Daniels

Inhibition of 11β‐HSD1 is hypothesized to improve measures of insulin sensitivity and hepatic glucose output in patients with type II diabetes. AMG 221 is a potent, small molecule inhibitor of 11β‐HSD1. The objective of this analysis is to describe the pharmacokinetic/pharmacodynamic (PK/PD) relationship between AMG 221 and 11β‐HSD1 inhibition in ex vivo adipose tissue samples. Healthy, obese subjects were administered a single dose of 3, 30, or 100 mg of oral AMG 221 (n = 44) or placebo (n = 11). Serial blood samples were collected over 24 hours. Subcutaneous adipose tissue samples were collected by open biopsy. Population PK/PD analysis was conducted using NONMEM. The inhibitory effects (mean ± standard error of the estimate) of AMG 221 on 11β‐HSD1 activity were directly related to adipose concentrations with Imax (the maximal inhibition of 11β‐HSD1 activity) and IC50 (the plasma AMG 221 concentration associated with 50% inhibition of enzyme activity) of 0.975 ± 0.003 and 1.19 ± 0.12 ng/mL, respectively. The estimated baseline 11β‐HSD1 enzyme activity was 755 ± 61 pmol/mg. An equilibration rate constant (keo) of 0.220 ± 0.021 h–1 described the delay between plasma and adipose tissue AMG 221 concentrations. AMG 221 potently blocked 11β‐HSD1 activity producing sustained inhibition for the 24‐hour study duration as measured in ex vivo adipose samples. Early characterization of concentration‐response relationships can support rational selection of dose and regimen for future studies.


Expert Review of Cardiovascular Therapy | 2015

Evolocumab (AMG 145) for primary hypercholesterolemia

Gisle Langslet; Maurice Emery; Scott M. Wasserman

Evolocumab is a fully human monoclonal IgG2 antibody that inhibits proprotein convertase subtilisin/kexin type 9, a protein that targets LDL receptors for degradation and thereby reduces the liver’s ability to remove LDL-C from the blood. In Phase II and III trials in more than 6000 subjects with primary hypercholesterolemia, evolocumab reduced LDL-C by 50–75% compared with placebo and by 35–45% compared with ezetimibe. Evolocumab reduced the proatherogenic lipid profile, including Lp(a), and modestly increased HDL-C and ApoA1. In subjects with homozygous familial hypercholesterolemia, evolocumab reduced LDL-C by 30%. Safety and tolerability of evolocumab was similar to that of placebo and ezetimibe. The ongoing FOURIER trial, anticipated to report in 2017, will provide definitive evidence on cardiovascular endpoints and additional long-term safety.


Journal of the American College of Cardiology | 2012

EFFECTS OF AMG 145, A FULLY HUMAN MONOCLONAL ANTIBODY AGAINST PCSK9, ON LOW-DENSITY LIPOPROTEIN CHOLESTEROL IN SUBJECTS TAKING STATINS: A PHASE 1, RANDOMIZED, DOUBLE-BLIND, PLACEBO-CONTROLLED, ASCENDING MULTIPLE-DOSE STUDY

Clapton Dias; Adam Shaywitz; Blaire Cooke; Stephen Uy; Maurice Emery; John P. Gibbs; Caroline Crispino; Karen Smirnakis; Brian Smith; Bing Gao; Scott M. Wasserman; Evan Stein

Proprotein convertase subtilisin/kexin type 9 (PCSK9) downregulates surface expression of the low density lipoprotein (LDL) receptor (LDL-R), increasing circulating LDL cholesterol (LDL-C). Statins increase LDL-R and PCSK9 levels. AMG 145, a fully human monoclonal antibody against PCSK9, has been


The Journal of Clinical Pharmacology | 2017

Impact of Target-Mediated Elimination on the Dose and Regimen of Evolocumab, a Human Monoclonal Antibody Against Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9).

John P. Gibbs; Sameer Doshi; Mita Kuchimanchi; Anita Grover; Maurice Emery; Michael Dodds; Megan A. Gibbs; Ransi Somaratne; Scott M. Wasserman; Dirk Blom

Understanding the pharmacokinetic (PK) and pharmacodynamic (PD) relationship of a therapeutic monoclonal antibody against proprotein convertase subtilisin/kexin type 9 (PCSK9) exhibiting target‐mediated drug disposition (TMDD) is critical for selecting optimal dosing regimens. We describe the PK/PD relationship of evolocumab using a mathematical model that captures evolocumab binding and removal of unbound PCSK9 as well as reduction in circulating low‐density lipoprotein cholesterol (LDL‐C). Data were pooled from 2 clinical studies: a single‐dose escalation study in healthy subjects (7‐420 mg SC; n = 44) and a multiple‐dose escalation study in statin‐treated hypercholesterolemic patients (14 mg weekly to 420 mg monthly [QM] SC; n = 57). A TMDD model described the time course of unbound evolocumab concentrations and removal of unbound PCSK9. The estimated linear clearance and volume of evolocumab were 0.256 L/day and 2.66 L, respectively, consistent with other monoclonal antibodies. The time course of LDL‐C reduction was described by an indirect response model with the elimination rate of LDL‐C being modulated by unbound PCSK9. The concentration of unbound PCSK9 associated with half‐maximal inhibition (IC50) of LDL‐C elimination was 1.46 nM. Based on simulations, 140 mg every 2 weeks (Q2W) and 420 mg QM were predicted to achieve a similar time‐averaged effect of 69% reduction in LDL‐C in patients on statin therapy, suggesting that an approximate 3‐fold dose increase is required for a 2‐fold extension in the dosing interval. Evolocumab dosing regimens of 140 mg Q2W or 420 mg QM were predicted to result in comparable reductions in LDL‐C over a monthly period, consistent with results from recently completed phase 3 studies.


ACS Medicinal Chemistry Letters | 2011

Synthesis and Evaluation of the Metabolites of AMG 221, a Clinical Candidate for the Treatment of Type 2 Diabetes.

Aiwen Li; Chester Chenguang Yuan; David Chow; Michelle Chen; Maurice Emery; Clarence Hale; Xiping Zhang; Raju Subramanian; David J. St. Jean; Renee Komorowski; Murielle M. Véniant; Minghan Wang; Christopher Fotsch

All eight of the major active metabolites of (S)-2-((1S,2S,4R)-bicyclo[2.2.1]heptan-2-ylamino)-5-isopropyl-5-methylthiazol-4(5H)-one (AMG 221, compound 1), an inhibitor of 11β-hydroxysteroid dehydrogenase type 1 that has entered the clinic for the treatment of type 2 diabetes, were synthetically prepared and confirmed by comparison with samples generated in liver microsomes. After further profiling, we determined that metabolite 2 was equipotent to 1 on human 11β-HSD1 and had lower in vivo clearance and higher bioavailability in rat and mouse. Compound 2 was advanced into a pharmacodynamic model in mouse where it inhibited adipose 11β-HSD1 activity.


Combinatorial Chemistry & High Throughput Screening | 2010

Application of In Vivo Animal Models to Characterize the Pharmacokinetic and Pharmacodynamic Properties of Drug Candidates in Discovery Settings

Benny Amore; John P. Gibbs; Maurice Emery

A goal of preclinical discovery is the identification of drug candidates suitable for clinical testing. Successful integration of in vitro and in vivo experimental data sets can afford projections of human dose regimens anticipated to be safe and therapeutically beneficial. While in vitro experiments guide new chemical syntheses and are essential to understanding drug action and disposition, in vivo characterizations provide unique insight into complex biological systems that control concentrations at the site of action and pharmacologic response. Pharmacokinetic and pharmacodynamic (PK/PD) concepts underlying drug disposition and response provide a quantitative framework with which to identify potential clinical candidates. To improve throughput in earlier stages of drug discovery, in vivo pharmacokinetic study designs such as cassette dosing and sparse sampling schemes have been utilized. In later stages of discovery, pharmacokinetic studies using chemical inhibitors or surgical and genetic animal models are used to characterize the underlying determinants of drug disposition. In a complimentary fashion, modeling of in vivo pharmacodynamic effects may quantitatively link biomarkers to pharmacological response, validate in vitro to in vivo correlations and underwrite predictions of efficacious exposure targets. When applied to in vivo discovery data, PK/PD models have aided in understanding mechanisms of pharmacological response such as receptor theory in the central nervous system and cell turnover concepts in infectious disease and oncology. This review considers the role of in vivo testing toward understanding the pharmacokinetic and pharmacodynamic attributes of lead candidates in drug discovery.

Collaboration


Dive into the Maurice Emery's collaboration.

Researchain Logo
Decentralizing Knowledge