Network


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

Hotspot


Dive into the research topics where Lynn Hyde is active.

Publication


Featured researches published by Lynn Hyde.


Science Translational Medicine | 2016

The BACE1 inhibitor verubecestat (MK-8931) reduces CNS β-amyloid in animal models and in Alzheimer’s disease patients

Matthew E. Kennedy; Andrew W. Stamford; Xia Chen; Kathleen Cox; Jared N. Cumming; Marissa Dockendorf; Michael F. Egan; Larry Ereshefsky; Robert Hodgson; Lynn Hyde; Stanford Jhee; Huub Jan Kleijn; Reshma Kuvelkar; Wei Li; Britta A. Mattson; Hong Mei; John Palcza; Jack D. Scott; Michael Tanen; Matthew D. Troyer; Jack Tseng; Julie A. Stone; Eric M. Parker

The BACE1 inhibitor verubecestat safely reduces β-amyloid deposition in rats, monkeys, healthy human subjects, and patients with Alzheimer’s disease. Getting to first BACE The discovery of BACE1 inhibitors that reduce β-amyloid peptides in Alzheimer’s disease (AD) patients has been an encouraging development in the quest for a disease-modifying therapy. Kennedy and colleagues now report the discovery of verubecestat, a structurally unique, orally bioavailable small molecule that potently inhibits brain BACE1 activity resulting in a reduction in Aβ peptides in the cerebrospinal fluid of animals, healthy volunteers, and AD patients. No dose-limiting toxicities were observed in chronic animal toxicology studies or in phase 1 human studies, thus reducing safety concerns raised by previous reports of BACE inhibitors and BACE1 knockout mice. β-Amyloid (Aβ) peptides are thought to be critically involved in the etiology of Alzheimer’s disease (AD). The aspartyl protease β-site amyloid precursor protein cleaving enzyme 1 (BACE1) is required for the production of Aβ, and BACE1 inhibition is thus an attractive target for the treatment of AD. We show that verubecestat (MK-8931) is a potent, selective, structurally unique BACE1 inhibitor that reduced plasma, cerebrospinal fluid (CSF), and brain concentrations of Aβ40, Aβ42, and sAPPβ (a direct product of BACE1 enzymatic activity) after acute and chronic administration to rats and monkeys. Chronic treatment of rats and monkeys with verubecestat achieved exposures >40-fold higher than those being tested in clinical trials in AD patients yet did not elicit many of the adverse effects previously attributed to BACE inhibition, such as reduced nerve myelination, neurodegeneration, altered glucose homeostasis, or hepatotoxicity. Fur hypopigmentation was observed in rabbits and mice but not in monkeys. Single and multiple doses were generally well tolerated and produced reductions in Aβ40, Aβ42, and sAPPβ in the CSF of both healthy human subjects and AD patients. The human data were fit to an amyloid pathway model that provided insight into the Aβ pools affected by BACE1 inhibition and guided the choice of doses for subsequent clinical trials.


Bioorganic & Medicinal Chemistry Letters | 2012

Structure based design of iminohydantoin BACE1 inhibitors: Identification of an orally available, centrally active BACE1 inhibitor

Jared N. Cumming; Elizabeth M. Smith; Lingyan Wang; Jeffrey Misiaszek; James Durkin; Jianping Pan; Ulrich Iserloh; Yusheng Wu; Zhaoning Zhu; Corey Strickland; Johannes Voigt; Xia Chen; Matthew E. Kennedy; Reshma Kuvelkar; Lynn Hyde; Kathleen Cox; Leonard Favreau; Michael Czarniecki; William J. Greenlee; Brian A. McKittrick; Eric M. Parker; Andrew W. Stamford

From an initial lead 1, a structure-based design approach led to identification of a novel, high-affinity iminohydantoin BACE1 inhibitor that lowers CNS-derived Aβ following oral administration to rats. Herein we report SAR development in the S3 and F subsites of BACE1 for this series, the synthetic approaches employed in this effort, and in vivo data for the optimized compound.


Journal of Pharmacology and Experimental Therapeutics | 2015

MLi-2, a potent, selective and centrally active compound for exploring the therapeutic potential and safety of LRRK2 kinase inhibition

Matthew J. Fell; Christian Mirescu; Kallol Basu; Boonlert Cheewatrakoolpong; Duane Demong; J. Michael Ellis; Lynn Hyde; Yinghui Lin; Carrie G. Markgraf; Hong Mei; Michael D. Miller; Frederique M. Poulet; Jack D. Scott; Michelle Smith; Zhizhang Yin; Xiaoping Zhou; Eric M. Parker; Matthew E. Kennedy; John A. Morrow

Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are the most common genetic cause of familial and sporadic Parkinson’s disease (PD). That the most prevalent mutation, G2019S, leads to increased kinase activity has led to a concerted effort to identify LRRK2 kinase inhibitors as a potential disease-modifying therapy for PD. An internal medicinal chemistry effort identified several potent and highly selective compounds with favorable drug-like properties. Here, we characterize the pharmacological properties of cis-2,6-dimethyl-4-(6-(5-(1-methylcyclopropoxy)-1H-indazol-3-yl)pyrimidin-4-yl)morpholine (MLi-2), a structurally novel, highly potent, and selective LRRK2 kinase inhibitor with central nervous system activity. MLi-2 exhibits exceptional potency in a purified LRRK2 kinase assay in vitro (IC50 = 0.76 nM), a cellular assay monitoring dephosphorylation of LRRK2 pSer935 LRRK2 (IC50 = 1.4 nM), and a radioligand competition binding assay (IC50 = 3.4 nM). MLi-2 has greater than 295-fold selectivity for over 300 kinases in addition to a diverse panel of receptors and ion channels. Acute oral and subchronic dosing in MLi-2 mice resulted in dose-dependent central and peripheral target inhibition over a 24-hour period as measured by dephosphorylation of pSer935 LRRK2. Treatment of MitoPark mice with MLi-2 was well tolerated over a 15-week period at brain and plasma exposures >100× the in vivo plasma IC50 for LRRK2 kinase inhibition as measured by pSer935 dephosphorylation. Morphologic changes in the lung, consistent with enlarged type II pneumocytes, were observed in MLi-2-treated MitoPark mice. These data demonstrate the suitability of MLi-2 as a compound to explore LRRK2 biology in cellular and animal models.


Bioorganic & Medicinal Chemistry Letters | 2011

Discovery of fused 5,6-bicyclic heterocycles as γ-secretase modulators

Jun Qin; Pawan Dhondi; Xianhai Huang; Mihirbaran Mandal; Zhiqiang Zhao; Dmitri Pissarnitski; Wei Zhou; Robert Aslanian; Zhaoning Zhu; William J. Greenlee; John W. Clader; Lili Zhang; Mary Cohen-Williams; Nicholas Jones; Lynn Hyde; Anandan Palani

We herein report the discovery of four series of fused 5,6-bicyclic heterocycles as γ-secretase modulators. Synthesis and SAR of these series are discussed. These compounds represent a new class of γ-secretase modulators that demonstrate moderate to good in vitro potency in inhibiting Aβ(42) production.


Bioorganic & Medicinal Chemistry Letters | 2010

Iminoheterocycles as γ-secretase modulators

John P. Caldwell; Chad E. Bennett; Troy Mccracken; Robert Mazzola; Thomas Bara; Alexei V. Buevich; Duane A. Burnett; Inhou Chu; Mary Cohen-Williams; Hubert Josein; Lynn Hyde; Julie Lee; Brian A. McKittrick; Lixin Song; Giuseppe Terracina; Johannes Voigt; Lili Zhang; Zhaoning Zhu

The synthesis of a novel series of iminoheterocycles and their structure-activity relationship (SAR) as modulators of gamma-secretase activity will be detailed. Encouraging SAR generated from a monocyclic core led to a structurally unique bicyclic core. Selected compounds exhibit good potency as gamma-secretase modulators, excellent rat pharmacokinetics, and lowering of Abeta42 levels in various in vivo models.


Molecular Neurodegeneration | 2015

Analysis of tau post-translational modifications in rTg4510 mice, a model of tau pathology.

Lixin Song; Sherry X. Lu; Xuesong Ouyang; Jerry Melchor; Julie Lee; Giuseppe Terracina; Xiaohai Wang; Lynn Hyde; J. Fred Hess; Eric M. Parker; Lili Zhang

BackgroundMicrotubule associated protein tau is the major component of the neurofibrillary tangles (NFTs) found in the brains of patients with Alzheimer’s disease and several other neurodegenerative diseases. Tau mutations are associated with frontotemperal dementia with parkinsonism on chromosome 17 (FTDP-17). rTg4510 mice overexpress human tau carrying the P301L FTDP-17 mutation and develop robust NFT-like pathology at 4–5 months of age. The current study is aimed at characterizing the rTg4510 mice to better understand the genesis of tau pathology and to better enable the use of this model in drug discovery efforts targeting tau pathology.ResultsUsing a panel of immunoassays, we analyzed the age-dependent formation of pathological tau in rTg4510 mice and our data revealed a steady age-dependent accumulation of pathological tau in the insoluble fraction of brain homogenates. The pathological tau was associated with multiple post-translational modifications including aggregation, phosphorylation at a wide variety of sites, acetylation, ubiquitination and nitration. The change of most tau species reached statistical significance at the age of 16 weeks. There was a strong correlation between the different post-translationally modified tau species in this heterogeneous pool of pathological tau. Total tau in the cerebrospinal fluid (CSF) displayed a multiphasic temporal profile distinct from the steady accumulation of pathological tau in the brain. Female rTg4510 mice displayed significantly more aggressive accumulation of pathological tau in the brain and elevation of total tau in CSF than their male littermates.ConclusionThe immunoassays described here were used to generate the most comprehensive description of the changes in various tau species across the lifespan of the rTg4510 mouse model. The data indicate that development of tauopathy in rTg4510 mice involves the accumulation of a pool of pathological tau that carries multiple post-translational modifications, a process that can be detected well before the histological detection of NFTs. Therapeutic treatment targeting tau should therefore aim to reduce all tau species associated with the pathological tau pool rather than reduce specific post-translational modifications. There is still much to learn about CSF tau in physiological and pathological processes in order to use it as a translational biomarker in drug discovery.


Bioorganic & Medicinal Chemistry Letters | 2010

Tricyclic sulfones as orally active γ-secretase inhibitors: Synthesis and structure–activity relationship studies

T.K. Sasikumar; Li Qiang; Duane A. Burnett; David Cole; Ruo Xu; Hongmei Li; William J. Greenlee; John W. Clader; Lili Zhang; Lynn Hyde

Tricyclic sulfones were designed as gamma-secretase inhibitors and found to have excellent potency. Extensive SAR shows that a large number of sulfonamides at position 7 of the tricycle are very well tolerated. Compounds such as 15a and 15c showed remarkable in vivo potency.


Bioorganic & Medicinal Chemistry Letters | 2013

The discovery of fused oxadiazepines as gamma secretase modulators for treatment of Alzheimer’s disease

Hongmei Li; Jun Qin; Pawan Dhondi; Wei Zhou; Monica Vicarel; Thomas Bara; David Cole; Hubert Josien; Dmitri Pissarnitski; Zhaoning Zhu; Anandan Palani; Robert Aslanian; John W. Clader; Michael Czarniecki; William J. Greenlee; Mary Cohen-Williams; Lynn Hyde; Lixin Song; Lili Zhang; Inhou Chu; Xianhai Huang

In an attempt to further improve overall profiles of the oxadiazine series of GSMs, in particular the hERG activity, conformational modifications of the core structure resulted in the identification of fused oxadiazepines such as 7i which had an improved hERG inhibition profile and was a highly efficacious GSM in vitro and in vivo in rats. These SAR explorations offer opportunities to identify potential drugs to treat Alzheimers disease.


Journal of Medicinal Chemistry | 2016

Structure-Based Design of an Iminoheterocyclic β-Site Amyloid Precursor Protein Cleaving Enzyme (BACE) Inhibitor that Lowers Central Aβ in Nonhuman Primates

Mihirbaran Mandal; Yusheng Wu; Jeffrey Misiaszek; Guoqing Li; Alexei V. Buevich; John P. Caldwell; Xiaoxiang Liu; Robert Mazzola; Peter Orth; Corey Strickland; Johannes Voigt; Hongwu Wang; Zhaoning Zhu; Xia Chen; Michael Grzelak; Lynn Hyde; Reshma Kuvelkar; Presscott T. Leach; Giuseppe Terracina; Lili Zhang; Qi Zhang; Maria S. Michener; Brad Smith; Kathleen Cox; Diane Grotz; Leonard Favreau; Kaushik Mitra; Irina Kazakevich; Brian A. McKittrick; William J. Greenlee

We describe successful efforts to optimize the in vivo profile and address off-target liabilities of a series of BACE1 inhibitors represented by 6 that embodies the recently validated fused pyrrolidine iminopyrimidinone scaffold. Employing structure-based design, truncation of the cyanophenyl group of 6 that binds in the S3 pocket of BACE1 followed by modification of the thienyl group in S1 was pursued. Optimization of the pyrimidine substituent that binds in the S2-S2″ pocket of BACE1 remediated time-dependent CYP3A4 inhibition of earlier analogues in this series and imparted high BACE1 affinity. These efforts resulted in the discovery of difluorophenyl analogue 9 (MBi-4), which robustly lowered CSF and cortex Aβ40 in both rats and cynomolgus monkeys following a single oral dose. Compound 9 represents a unique molecular shape among BACE inhibitors reported to potently lower central Aβ in nonrodent preclinical species.


European Journal of Pharmacology | 2011

The anxiolytic-like profile of the nociceptin receptor agonist, endo-8-[bis(2-chlorophenyl)methyl]-3-phenyl-8-azabicyclo[3.2.1] octane-3-carboxamide (SCH 655842): Comparison of efficacy and side effects across rodent species

Sherry X. Lu; Guy A. Higgins; Robert Hodgson; Lynn Hyde; Robert A. Del Vecchio; Donald H. Guthrie; Tatiana M. Kazdoba; Martha F. McCool; Cynthia Morgan; Ana Bercovici; Ginny D. Ho; Deen Tulshian; Eric M. Parker; John C. Hunter; Geoffrey B. Varty

The endogenous opioid-like peptide, nociceptin, produces anxiolytic-like effects that are mediated via the nociceptin (NOP) receptor. Similarly, synthetic, non-peptide NOP agonists produce robust anxiolytic-like effects although these effects are limited by marked side effects. In the present studies, the effects of a novel NOP receptor agonist, SCH 655842, were examined in rodent models sensitive to anxiolytic drugs and tests measuring potential adverse affects. Oral administration of SCH 655842 produced robust, anxiolytic-like effects in three species, i.e., rat, guinea pig, and mouse. Specifically, SCH 655842 was effective in rat conditioned lick suppression (3-10 mg/kg) and fear-potentiated startle (3-10 mg/kg) tests, a guinea pig pup vocalization test (1-3 mg/kg), as well as in mouse Geller-Seifter (30 mg/kg) and marble burying (30 mg/kg) tests. The anxiolytic-like effect of SCH 655842 in the conditioned lick suppression test was attenuated by the NOP antagonist, J-113397. In mice, SCH 655842 reduced locomotor activity and body temperature at doses similar to the anxiolytic-like dose and these effects were absent in NOP receptor knockout mice. In rats, SCH 655842 did not produce adverse behavioral effects up to doses of 70-100 mg/kg. Pharmacokinetic studies in the rat confirmed dose-related increases in plasma and brain levels of SCH 655842 across a wide oral dose range. Taken together, SCH 655842 may represent a NOP receptor agonist with improved tolerability compared to other members of this class although further studies are necessary to establish whether this extends to higher species.

Researchain Logo
Decentralizing Knowledge