Network


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

Hotspot


Dive into the research topics where Laigao Chen is active.

Publication


Featured researches published by Laigao Chen.


Journal of Medicinal Chemistry | 2013

Design and selection parameters to accelerate the discovery of novel central nervous system positron emission tomography (PET) ligands and their application in the development of a novel phosphodiesterase 2A PET ligand.

Lei Zhang; Anabella Villalobos; Elizabeth Mary Beck; Thomas Bocan; Thomas A. Chappie; Laigao Chen; Sarah Grimwood; Steven D. Heck; Christopher John Helal; Xinjun Hou; John M. Humphrey; Jiemin Lu; Marc B. Skaddan; Timothy J. McCarthy; Patrick Robert Verhoest; Travis T. Wager; Kenneth Zasadny

To accelerate the discovery of novel small molecule central nervous system (CNS) positron emission tomography (PET) ligands, we aimed to define a property space that would facilitate ligand design and prioritization, thereby providing a higher probability of success for novel PET ligand development. Toward this end, we built a database consisting of 62 PET ligands that have successfully reached the clinic and 15 radioligands that failed in late-stage development as negative controls. A systematic analysis of these ligands identified a set of preferred parameters for physicochemical properties, brain permeability, and nonspecific binding (NSB). These preferred parameters have subsequently been applied to several programs and have led to the successful development of novel PET ligands with reduced resources and timelines. This strategy is illustrated here by the discovery of the novel phosphodiesterase 2A (PDE2A) PET ligand 4-(3-[(18)F]fluoroazetidin-1-yl)-7-methyl-5-{1-methyl-5-[4-(trifluoromethyl)phenyl]-1H-pyrazol-4-yl}imidazo[5,1-f][1,2,4]triazine, [(18)F]PF-05270430 (5).


Journal of Medicinal Chemistry | 2014

Discovery and preclinical characterization of 1-methyl-3-(4-methylpyridin-3-yl)-6-(pyridin-2-ylmethoxy)-1H-pyrazolo-[3,4-b]pyrazine (PF470): a highly potent, selective, and efficacious metabotropic glutamate receptor 5 (mGluR5) negative allosteric modulator.

Lei Zhang; Gayatri Balan; Gabriela Barreiro; Brian P. Boscoe; Lois K. Chenard; Julie Cianfrogna; Michelle Marie Claffey; Laigao Chen; Karen J. Coffman; Susan E. Drozda; Joshua R. Dunetz; Kari R. Fonseca; Paul Galatsis; Sarah Grimwood; John T. Lazzaro; Jessica Y. Mancuso; Emily L. Miller; Matthew R. Reese; Bruce N. Rogers; Isao Sakurada; Marc B. Skaddan; Deborah L. Smith; Antonia F. Stepan; Patrick Trapa; Jamison B. Tuttle; Patrick Robert Verhoest; Daniel P. Walker; Ann S. Wright; Margaret M. Zaleska; Kenneth Zasadny

A novel series of pyrazolopyrazines is herein disclosed as mGluR5 negative allosteric modulators (NAMs). Starting from a high-throughput screen (HTS) hit (1), a systematic structure-activity relationship (SAR) study was conducted with a specific focus on balancing pharmacological potency with physicochemical and pharmacokinetic (PK) properties. This effort led to the discovery of 1-methyl-3-(4-methylpyridin-3-yl)-6-(pyridin-2-ylmethoxy)-1H-pyrazolo[3,4-b]pyrazine (PF470, 14) as a highly potent, selective, and orally bioavailable mGluR5 NAM. Compound 14 demonstrated robust efficacy in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-rendered Parkinsonian nonhuman primate model of l-DOPA-induced dyskinesia (PD-LID). However, the progression of 14 to the clinic was terminated because of a potentially mechanism-mediated finding consistent with a delayed-type immune-mediated type IV hypersensitivity in a 90-day NHP regulatory toxicology study.


Nuclear Medicine and Biology | 2012

The synthesis and in vivo evaluation of [18F]PF-9811: a novel PET ligand for imaging brain fatty acid amide hydrolase (FAAH).

Marc B. Skaddan; Lei Zhang; Douglas S. Johnson; Aijun Zhu; Kenneth Zasadny; Richard V. Coelho; Kyle Kuszpit; Gwen Currier; Kuo-Hsien Fan; Elizabeth Mary Beck; Laigao Chen; Susan E. Drozda; Gayatri Balan; Micah J. Niphakis; Benjamin F. Cravatt; Kay Ahn; Thomas Bocan; Anabella Villalobos

INTRODUCTION Fatty acid amide hydrolase (FAAH) is responsible for the enzymatic degradation of the fatty acid amide family of signaling lipids, including the endogenous cannabinoid (endocannabinoid) anandamide. The involvement of the endocannabinoid system in pain and other nervous system disorders has made FAAH an attractive target for drug development. Companion molecular imaging probes are needed, however, to assess FAAH inhibition in the nervous system in vivo. We report here the synthesis and in vivo evaluation of [(18)F]PF-9811, a novel PET ligand for non-invasive imaging of FAAH in the brain. METHODS The potency and selectivity of unlabeled PF-9811 were determined by activity-based protein profiling (ABPP) both in vitro and in vivo. [(18)F]PF-9811 was synthesized in a 3-step, one-pot reaction sequence, followed by HPLC purification. Biological evaluation was performed by biodistribution and dynamic PET imaging studies in male rats. The specificity of [(18)F]PF-9811 uptake was evaluated by pre-administration of PF-04457845, a potent and selective FAAH inhibitor, 1h prior to radiotracer injection. RESULTS Biodistribution studies show good uptake (SUV~0.8 at 90 min) of [(18)F]PF-9811 in rat brain, with significant reduction of the radiotracer in all brain regions (37%-73% at 90 min) in blocking experiments. Dynamic PET imaging experiments in rat confirmed the heterogeneous uptake of [(18)F]PF-9811 in brain regions with high FAAH enzymatic activity, as well as statistically significant reductions in signal following pre-administration of the blocking compound PF-04457845. CONCLUSIONS [(18)F]PF-9811 is a promising PET imaging agent for FAAH. Biodistribution and PET imaging experiments show that the tracer has good uptake in brain, regional heterogeneity, and specific binding as determined by blocking experiments with the highly potent and selective FAAH inhibitor, PF-04457845.


Journal of Medicinal Chemistry | 2015

The Discovery and Characterization of the α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid (AMPA) Receptor Potentiator N-{(3S,4S)-4-[4-(5-Cyano-2-thienyl)phenoxy]tetrahydrofuran-3-yl}propane-2-sulfonamide (PF-04958242)

Christopher L. Shaffer; Nandini Chaturbhai Patel; Jacob Bradley Schwarz; Renato J. Scialis; Yunjing Wei; Xinjun J. Hou; Longfei Xie; Kapil Karki; Dianne K. Bryce; Sarah Osgood; William E. Hoffmann; John T. Lazzaro; Cheng Chang; Dina McGinnis; Susan M. Lotarski; JianHua Liu; R. Scott Obach; Mark L. Weber; Laigao Chen; Kenneth Zasadny; Patricia A. Seymour; Christopher J. Schmidt; Mihály Hajós; Raymond S. Hurst; Jayvardhan Pandit; Christopher J. O’Donnell

A unique tetrahydrofuran ether class of highly potent α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor potentiators has been identified using rational and structure-based drug design. An acyclic lead compound, containing an ether-linked isopropylsulfonamide and biphenyl group, was pharmacologically augmented by converting it to a conformationally constrained tetrahydrofuran to improve key interactions with the human GluA2 ligand-binding domain. Subsequent replacement of the distal phenyl motif with 2-cyanothiophene to enhance its potency, selectivity, and metabolic stability afforded N-{(3S,4S)-4-[4-(5-cyano-2-thienyl)phenoxy]tetrahydrofuran-3-yl}propane-2-sulfonamide (PF-04958242, 3), whose preclinical characterization suggests an adequate therapeutic index, aided by low projected human oral pharmacokinetic variability, for clinical studies exploring its ability to attenuate cognitive deficits in patients with schizophrenia.


Xenobiotica | 2017

Quantitative projection of human brain penetration of the H3 antagonist PF-03654746 by integrating rat-derived brain partitioning and PET receptor occupancy.

Aarti Sawant-Basak; Laigao Chen; Christopher L. Shaffer; Donna Palumbo; Anne W. Schmidt; Elaine Tseng; Douglas K. Spracklin; Jean-Dominique Gallezot; David Labaree; Nabeel Nabulsi; Yiyun Huang; Richard E. Carson; Timothy J. McCarthy

Abstract 1. Unbound brain drug concentration (Cb,u), a valid surrogate of interstitial fluid drug concentration (CISF), cannot be directly determined in humans, which limits accurately defining the human Cb,u:Cp,u of investigational molecules. 2. For the H3R antagonist (1R,3R)-N-ethyl-3-fluoro-3-[3-fluoro-4-(pyrrolidin-1-lmethyl)phenyl]cyclobutane-1-carboxamide (PF-03654746), we interrogated Cb,u:Cp,u in humans and nonhuman primate (NHP). 3. In rat, PF-03654746 achieved net blood–brain barrier (BBB) equilibrium (Cb,u:Cp,u of 2.11). 4. In NHP and humans, the PET receptor occupancy-based Cp,u IC50 of PF-03654746 was 0.99 nM and 0.31 nM, respectively, which were 2.1- and 7.4-fold lower than its in vitro human H3 Ki (2.3 nM). 5. In an attempt to understand this higher-than-expected potency in humans and NHP, rat-derived Cb,u:Cp,u of PF-03654746 was integrated with Cp,u IC50 to identify unbound (neuro) potency of PF-03654746, nIC50. 6. The nIC50 of PF-03654746 was 2.1 nM in NHP and 0.66 nM in human which better correlated (1.1- and 3.49-fold lower) with in vitro human H3 Ki (2.3 nM). 7. This correlation of the nIC50 and in vitro hH3 Ki suggested the translation of net BBB equilibrium of PF-03654746 from rat to NHP and humans, and confirmed the use of Cp,u as a reliable surrogate of Cb,u. 8. Thus, nIC50 quantitatively informed the human Cb,u:Cp,u of PF-03654746.


The Journal of Nuclear Medicine | 2016

Preclinical Evaluation of 18F-PF-05270430, a Novel PET Radioligand for the Phosphodiesterase 2A Enzyme

Laigao Chen; Nabeel Nabulsi; Mika Naganawa; Kenneth Zasadny; Marc B. Skaddan; Lei Zhang; Soheila Najafzadeh; Shu-fei Lin; Christopher John Helal; Tracey Boyden; Cheng Chang; Jim Ropchan; Richard E. Carson; Anabella Villalobos; Yiyun Huang

The enzyme phosphodiesterase 2A (PF-05270430) is a potential target for development of novel therapeutic agents for the treatment of cognitive impairments. The goal of the present study was to evaluate the PDE2A ligand 18F-PF-05270430, 4-(3-fluoroazetidin-1-yl)-7-methyl-5-(1-methyl-5-(4-(trifluoromethyl)phenyl)-1H-pyrazol-4-yl)imidazo[1,5-f][1,2,4]triazine, in nonhuman primates. Methods: 18F-PF-05270430 was radiolabeled by 2 methods via nucleophilic substitution of its tosylate precursor. Tissue metabolite analysis in rodents and PET imaging in nonhuman primates under baseline and blocking conditions were performed to determine the pharmacokinetic and binding characteristics of the new radioligand. Various kinetic modeling approaches were assessed to select the optimal method for analysis of imaging data. Results: 18F-PF-05270430 was synthesized in greater than 98% radiochemical purity and high specific activity. In the nonhuman primate brain, uptake of 18F-PF-05270430 was fast, with peak concentration (SUVs of 1.5–1.8 in rhesus monkeys) achieved within 7 min after injection. The rank order of uptake was striatum > neocortical regions > cerebellum. Regional time–activity curves were well fitted by the 2-tissue-compartment model and the multilinear analysis-1 (MA1) method to arrive at reliable estimates of regional distribution volume (VT) and binding potential (BPND) with 120 min of scan data. Regional VT values (MA1) ranged from 1.28 mL/cm3 in the cerebellum to 3.71 mL/cm3 in the putamen, with a BPND of 0.25 in the temporal cortex and 1.92 in the putamen. Regional BPND values estimated by the simplified reference tissue model (SRTM) were similar to those from MA1. Test–retest variability in high-binding regions (striatum) was 4% ± 6% for MA1 VT, 13% ± 6% for MA1 BPND, and 13% ± 7% SRTM BPND, respectively. Pretreatment of animals with the PDE2A inhibitor PF-05180999 resulted in a dose-dependent reduction of 18F-PF-05270430 specific binding, with a half maximal effective concentration of 69.4 ng/mL in plasma PF-05180999 concentration. Conclusion: 18F-PF-05270430 displayed fast and reversible kinetics in nonhuman primates, as well as specific binding blockable by a PDE2A inhibitor. This is the first PET tracer with desirable imaging properties and demonstrated ability to image and quantify PDE2A in vivo.


Journal of Medicinal Chemistry | 2017

Discovery and Characterization of (R)-6-Neopentyl-2-(pyridin-2-ylmethoxy)-6,7-dihydropyrimido[2,1-c][1,4]oxazin-4(9H)-one (PF-06462894), an Alkyne-Lacking Metabotropic Glutamate Receptor 5 Negative Allosteric Modulator Profiled in both Rat and Nonhuman Primates

Antonia F. Stepan; Michelle Marie Claffey; Matthew R. Reese; Gayatri Balan; Gabriela Barreiro; Jason Barricklow; Michael John Bohanon; Brian P. Boscoe; Gregg D. Cappon; Lois K. Chenard; Julie Cianfrogna; Laigao Chen; Karen J. Coffman; Susan E. Drozda; Joshua R. Dunetz; Somraj Ghosh; Xinjun Hou; Christopher Houle; Kapil Karki; John T. Lazzaro; Jessica Y. Mancuso; John M. Marcek; Emily L. Miller; Mark A. Moen; Steven V. O’Neil; Isao Sakurada; Marc B. Skaddan; Vinod D. Parikh; Deborah L. Smith; Patrick Trapa

We previously observed a cutaneous type IV immune response in nonhuman primates (NHP) with the mGlu5 negative allosteric modulator (NAM) 7. To determine if this adverse event was chemotype- or mechanism-based, we evaluated a distinct series of mGlu5 NAMs. Increasing the sp3 character of high-throughput screening hit 40 afforded a novel morpholinopyrimidone mGlu5 NAM series. Its prototype, (R)-6-neopentyl-2-(pyridin-2-ylmethoxy)-6,7-dihydropyrimido[2,1-c][1,4]oxazin-4(9H)-one (PF-06462894, 8), possessed favorable properties and a predicted low clinical dose (2 mg twice daily). Compound 8 did not show any evidence of immune activation in a mouse drug allergy model. Additionally, plasma samples from toxicology studies confirmed that 8 did not form any reactive metabolites. However, 8 caused the identical microscopic skin lesions in NHPs found with 7, albeit with lower severity. Holistically, this work supports the hypothesis that this unique toxicity may be mechanism-based although additional work is required to confirm this and determine clinical relevance.


Archive | 2015

Region-Based Data-Driven Intensity Normalization for Group Comparison of Functional Brain Images

Zhiyong Xie; Aijun Zhu; Laigao Chen; Timothy J. McCarthy

Intensity normalization is widely used to remove the confounding effect of global change exhibited in PET or SPECT brain images such that the local activity can be detected. Improper estimate of global change may induce a biased normalization. To separate the global change from local measurements, an iterative method is proposed to identify reference regions that are not affected by the local activity. From more than one hundred predefined anatomical regions, the reference regions are selected based on their intensity similarity between two groups. Weighted least squares regression is used to compute linear intensity transformations to align intensities of corresponding reference regions across all subjects. Studies with simulated data demonstrated that the proposed method performed better in recovering real intensity change comparing with global mean normalization and with Andersson’s data-driven method.


Journal of Veterinary Medical Science | 2008

MicroPET Detection of Regional Brain Activation Induced by Colonic Distention in a Rat Model of Visceral Hypersensitivity

Katsuyo Ohashi; Katsuomi Ichikawa; Laigao Chen; Michael J. Callahan; Kenneth Zasadny; Yoichi Kurebayashi


Diabetologia | 2018

Decreased VMAT2 in the pancreas of humans with type 2 diabetes mellitus measured in vivo by PET imaging

Gary W. Cline; Mika Naganawa; Laigao Chen; Kristin Chidsey; Santos Carvajal-Gonzalez; Sylvester Pawlak; Michelle Rossulek; Yanwei Zhang; Jason Bini; Timothy J. McCarthy; Richard E. Carson; Roberto A. Calle

Researchain Logo
Decentralizing Knowledge