Network


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

Hotspot


Dive into the research topics where Wai-si Eng is active.

Publication


Featured researches published by Wai-si Eng.


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.


Current Opinion in Chemical Biology | 1999

Positron emission tomography neuroreceptor imaging as a tool in drug discovery, research and development.

H. Donald Burns; Terence G. Hamill; Wai-si Eng; Barbara Francis; Christine Fioravanti; Raymond E. Gibson

Improved communication and cooperation between research-driven drug companies and academic positron emission tomography (PET) centers, coupled with improvements in PET camera resolution, the availability of small animal PET cameras and a growing list of neuroreceptor-specific PET tracers, have all contributed to a substantial increase in the use and value of PET as a tool in central nervous system drug discovery and development.


Current Pharmaceutical Design | 2000

Non-invasive radiotracer imaging as a tool for drug development.

Raymond E. Gibson; H. Donald Burns; Terence G. Hamill; Wai-si Eng; Barbara E. Francis; Christine Ryan

Non-Invasive Radiotracer Imaging (NIRI) uses either short-lived positron-emitting isotopes, such as 11C and 18F, for Positron Emis ion Tomography (PET) or single photon emitting nuclides, e.g., 123I, which provide images using planar imaging or Single-Photon Emission Computed Tomography (SPECT). These high-resolution imaging modalities provide anatomical distribution and localization of radiolabeled drugs, which can be used to generate real time receptor occupancy and off-rate studies in humans. This can be accomplished by either isotopically labeling a potential new drug (usually with 11C), or indirectly by studying how the unlabelled drug inhibits specific radioligand binding in vivo. Competitive blockade studies can be accomplished using a radiolabeled analogue which binds to the site of interest, rather than a radiolabeled version of the potential drug. Imaging, particularly PET imaging, can be used to demonstrate the effect of a drug through a biochemical marker of processes such as glucose metabolism or blood flow. NIRI as a development tool in the pharmaceutical industry is gaining increased acceptance as its unique ability to provide such critical information in human subjects is recognized. This section will review recent examples that illustrate the utility of NIRI, principally PET, in drug development, and the potential of imaging advances in the development of cancer drugs and gene therapy. Finally, we provide a brief overview of the design of new radiotracers for novel targets.


Journal of Psychopharmacology | 2011

Preclinical and clinical pharmacology of TPA023B, a GABAA receptor α2/α3 subtype-selective partial agonist.

John R. Atack; David James Hallett; S Tye; Keith A. Wafford; Christine Ryan; Sandra M. Sanabria-Bohórquez; Wai-si Eng; Raymond E. Gibson; Hd Burns; Gerard R. Dawson; Rw Carling; Lj Street; A Pike; I. De Lepeleire; K. Van Laere; Guy Bormans; Jn de Hoon; A. Van Hecken; Ruth M. McKernan; Mg Murphy; Richard Hargreaves

In the accompanying paper we describe how MRK-409 unexpectedly produced sedation in man at relatively low levels of GABAA receptor occupancy (∼10%). Since it was not clear whether this sedation was mediated via the α2/α3 or α1 GABAA subtype(s), we characterized the properties of TPA023B, a high-affinity imidazotriazine which, like MRK-409, has partial agonist efficacy at the α2 and α3 subtype but is an antagonist at the α1 subtype, at which MRK-409 has weak partial agonism. TPA023B gave dose- and time-dependent occupancy of rat brain GABAA receptors as measured using an in vivo [3H]flumazenil binding assay, with 50% occupancy corresponding to a respective dose and plasma drug concentration of 0.09 mg/kg and 19 ng/mL, the latter of which was similar to that observed in mice (25 ng/mL) and comparable to values obtained in baboon and man using [11C]flumazenil PET (10 and 5.8 ng/mL, respectively). TPA023B was anxiolytic in rodent and primate (squirrel monkey) models of anxiety (elevated plus maze, fear-potentiated startle, conditioned suppression of drinking, conditioned emotional response) yet had no significant effects in rodent or primate assays of ataxia and/or myorelaxation (rotarod, chain-pulling, lever pressing), up to doses (10 mg/kg) corresponding to occupancy of greater than 99%. In man, TPA023B was well tolerated at a dose (1.5 mg) that produced occupancy of >50%, suggesting that the sedation previously seen with MRK-409 is due to the partial agonist efficacy of that compound at the α1 subtype, and highlighting the importance of antagonist efficacy at this particular GABAA receptor population for avoiding sedation in man.


Synapse | 2011

Synthesis, characterization, and monkey PET studies of [18F]MK-1312, a PET tracer for quantification of mGluR1 receptor occupancy by MK-5435

Eric Hostetler; Wai-si Eng; Aniket D. Joshi; Sandra M. Sanabria-Bohórquez; Hiroshi Kawamoto; Satoru Ito; Stacey O'Malley; Stephen Krause; Christine Ryan; Shil Patel; Mangay Williams; Kerry Riffel; Gentaroh Suzuki; Satoshi Ozaki; Hisashi Ohta; Jacquelynn J. Cook; H. Donald Burns; Richard Hargreaves

Two moderately lipophilic, high affinity ligands for metabotropic glutamate receptor subtype 1 (mGluR1) were radiolabeled with a positron‐emitting radioisotope and evaluated in rhesus monkey as potential PET tracers. Both ligands were radiolabeled with fluorine‐18 via nucleophilic displacement of the corresponding 2‐chloropyridine precursor with [18F]potassium fluoride. [18F]MK‐1312 was found to have a suitable signal for quantification of mGluR1 receptors in nonhuman primates and was more thoroughly characterized. In vitro autoradiographic studies with [18F]MK‐1312 in rhesus monkey and human brain tissue slices revealed an uptake distribution consistent with the known distribution of mGluR1, with the highest uptake in the cerebellum, moderate uptake in the hippocampus, thalamus, and cortical regions, and lowest uptake in the caudate and putamen. In vitro saturation binding studies in rhesus monkey and human cerebellum homogenates confirmed that [18F]MK‐1312 binds to a single site with a Bmax/Kd ratio of 132 and 98, respectively. PET studies in rhesus monkey with [18F]MK‐1312 showed high brain uptake and a regional distribution consistent with in vitro autoradiography results. Blockade of [18F]MK‐1312 uptake with mGluR1 allosteric antagonist MK‐5435 dose‐dependently reduced tracer uptake in all regions of gray matter to a similarly low level of tracer uptake. This revealed a large specific signal useful for determination of mGluR1 receptor occupancy in rhesus monkey. Taken together, these results are promising for clinical PET studies with [18F]MK‐1312 to determine mGluR1 occupancy of MK‐5435. Synapse 2011.


Synapse | 2009

Inverse agonist histamine H3 receptor PET tracers labelled with carbon-11 or fluorine-18

Terence G. Hamill; Nagaaki Sato; Makoto Jitsuoka; Shigeru Tokita; Sandra Sanabria; Wai-si Eng; Christine Ryan; Stephen Krause; Norihiro Takenaga; Shil Patel; Zhizhen Zeng; David L. Williams; Cyrille Sur; Richard Hargreaves; H. Donald Burns

Two histamine H3 receptor (H3R) inverse agonist PET tracers have been synthesized and characterized in preclinical studies. Each tracer has high affinity for the histamine H3 receptor, has suitable lipophilicity, and neither is a substrate for the P‐glycoprotein efflux pump. A common phenolic precursor was used to synthesize each tracer with high specific activity and radiochemical purity by an alkylation reaction using either [11C]MeI or [18F]FCD2Br. Autoradiographic studies in rhesus monkey and human brain slices showed that each tracer had a widespread distribution with high binding densities in frontal cortex, globus pallidus and striatum, and lower uptake in cerebellum. The specificity of this expression pattern was demonstrated by the blockade of the autoradiographic signal by either the H3R agonist R‐α‐methylhistamine or a histamine H3R inverse agonist. In vivo PET imaging studies in rhesus monkey showed rapid uptake of each tracer into the brain with the same distribution seen in the autoradiographic studies. Each tracer could be blocked by pretreatment with a histamine H3R inverse agonist giving a good specific signal. Comparison of the in vitro metabolism of each compound showed slower metabolism in human liver microsomes than in rhesus monkey liver microsomes, with each compound having a similar clearance rate in humans. The in vivo metabolism of 1b in rhesus monkey showed that at 60 min, ∼35% of the circulating counts were due to the parent. These tracers are very promising candidates as clinical PET tracers to both study the histamine H3R system and measure receptor occupancy of H3R therapeutic compounds. Synapse 63:1122–1132, 2009.


Synapse | 2011

The synthesis and preclinical evaluation in rhesus monkey of [18F]MK-6577 and [11C]CMPyPB glycine transporter 1 positron emission tomography radiotracers

Terence G. Hamill; Wai-si Eng; Andrew Jennings; Richard Thomas Lewis; Steven R. Thomas; Suzanne Wood; Leslie J. Street; David D. Wisnoski; Scott E. Wolkenberg; Craig W. Lindsley; Sandra M. Sanabria-Bohórquez; Shil Patel; Kerry Riffel; Christine Ryan; Jacquelynn J. Cook; Cyrille Sur; H. Donald Burns; Richard Hargreaves

Two positron emission tomography radiotracers for the glycine transporter 1 (GlyT1) are reported here. Each radiotracer is a propylsulfonamide‐containing benzamide and was labeled with either carbon‐11 or fluorine‐18. [11C]CMPyPB was synthesized by the alkylation of a 3‐hydroxypyridine precursor using [11C]MeI, and [18F]MK‐6577 was synthesized by a nucleophilic aromatic substitution reaction using a 2‐chloropyridine precursor. Each tracer shows good uptake into rhesus monkey brain with the expected distribution of highest uptake in the pons, thalamus, and cerebellum and lower uptake in the striatum and gray matter of the frontal cortex. In vivo blockade and chase studies of [18F]MK‐6577 showed a large specific signal and reversible binding. In vitro autoradiographic studies with [18F]MK‐6577 showed a large specific signal in both rhesus monkey and human brain slices and a distribution consistent with the in vivo results and those reported in the literature. In vivo metabolism studies in rhesus monkeys demonstrated that only more‐polar metabolites are formed for each tracer. Of these two tracers, [18F]MK‐6577 was more extensively characterized and is a promising clinical positron emission tomography tracer for imaging GlyT1 and for measuring GlyT1 occupancy of therapeutic compounds. Synapse, 2011.


Molecular Imaging and Biology | 2003

Image-derived input function for [11C]flumazenil kinetic analysis in human brain

Sandra M. Sanabria-Bohórquez; Alex Maes; Patrick Dupont; Guy Bormans; Tjibbe de Groot; Alexandre Coimbra; Wai-si Eng; Tine Laethem; Inge De Lepeleire; Jay Gambale; Jose M. Vega; H. Donald Burns

PURPOSE We describe a method for analysis of [11C]flumazenil data using an input curve directly derived from the positron emission tomography (PET) images. PROCEDURE The shape of the tracer plasma curve was obtained from the product of the intact flumazenil fraction in plasma in six arterial samples and the internal carotid artery time-activity curve (TAC). The resulting curve was calibrated using the [11C]flumazenil concentration in three of the six samples. The curve peak was recovered by adding an exponential function to the scaled curve whose parameters were estimated from simultaneous fittings of several tissue TACs assuming that all regions share the same input. RESULTS Good agreement was found between the image-derived and the experimental plasma curves in six subjects. Distribution volumes were highly correlated with linear regression slope and intercept values between [0.94, 1.03] and [-0.10, 0.16], respectively. CONCLUSION The proposed method is suitable for benzodiazepine receptor quantification requiring only a few blood samples.


NeuroImage | 2013

Evaluation of [18F]MK-0911, a positron emission tomography (PET) tracer for opioid receptor-like 1 (ORL1), in rhesus monkey and human

Eric Hostetler; Sandra M. Sanabria-Bohórquez; Wai-si Eng; Aniket D. Joshi; Shailendra Patel; Raymond E. Gibson; Stacey O'Malley; Stephen Krause; Christine Ryan; Kerry Riffel; Sheng Bi; Osamu Okamoto; Hiroshi Kawamoto; Satoshi Ozaki; Hisashi Ohta; Tjibbe de Groot; Guy Bormans; Marleen Depré; Jan de Hoon; Inge De Lepeleire; Tom Reynders; Jacquelynn J. Cook; H. Donald Burns; Michael F. Egan; William Cho; Koen Van Laere; Richard Hargreaves

Antagonism of the central opioid receptor like-1 receptor (ORL1) has been implicated in cognition, and has been a focus of drug discovery efforts to ameliorate the cognitive deficits that remain during the stable treatment of schizophrenia with current antipsychotics. In order to facilitate dose selection for phase II clinical testing an ORL1-specific PET tracer was developed to determine drug plasma concentration versus occupancy relationships in order to ensure that the doses selected and the degree of target engagement were sufficient to ensure adequate proof of concept testing. MK-0911 is a selective, high affinity antagonist for the ORL1 receptor radiolabeled with high specific activity (18)F for positron emission tomography (PET) studies. Evaluation of [(18)F]MK-0911 in rhesus monkey PET studies showed a pattern of brain uptake which was consistent with the known distribution of ORL1. In vitro autoradiography with [(18)F]MK-0911 in rhesus monkey and human brain tissue slices showed a regional distribution that was consistent with in vivo imaging results in monkey. Pre-treatment of rhesus monkeys with high doses of structurally diverse ORL1 antagonists MK-0584, MK-0337, or MK-5757 achieved blockade of [(18)F]MK-0911 in all gray matter regions. Baseline PET studies with [(18)F]MK-0911 in healthy human subjects showed tracer distribution and kinetics similar to that observed in rhesus monkey. Quantification of [(18)F]MK-0911 uptake in repeat human baseline PET studies showed a test-retest variability in volume of distribution (V(T)) averaging 3% across brain regions. Humans dosed orally with MK-5757 showed reduced [(18)F]MK-0911 tracer concentration in brain proportional with MK-5757 dose and plasma level. [(18)F]MK-0911 was useful for determining MK-5757-induced receptor occupancy of ORL1 to guide MK-5757 dose-selection for clinical proof-of-concept studies. Additionally, [(18)F]MK-0911 may be a useful tool for studying the pharmacology of ORL1 in various human populations and disease states.

Collaboration


Dive into the Wai-si Eng's collaboration.

Top Co-Authors

Avatar

Richard Hargreaves

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Christine Ryan

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Terence G. Hamill

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Sandra Sanabria

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Raymond E. Gibson

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Stephen Krause

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Guy Bormans

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kerry Riffel

United States Military Academy

View shared research outputs
Researchain Logo
Decentralizing Knowledge