Network


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

Hotspot


Dive into the research topics where Gary Tong is active.

Publication


Featured researches published by Gary Tong.


Clinical Therapeutics | 2012

Multicenter, Randomized, Double-Blind, Placebo-Controlled, Single-Ascending Dose Study of the Oral γ-Secretase Inhibitor BMS-708163 (Avagacestat): Tolerability Profile, Pharmacokinetic Parameters, and Pharmacodynamic Markers

Gary Tong; Jun-Sheng Wang; Oleksandr Sverdlov; Shu-Pang Huang; Randy Slemmon; Robert Croop; Lorna Castaneda; Huidong Gu; Oi Wong; Hewei Li; Robert M. Berman; Christina Smith; Charles F. Albright; Randy C. Dockens

BACKGROUND γ-Secretase inhibitors (GSIs) are being investigated for their potential to modify the progression of Alzheimer disease based on their ability to regulate amyloid-β (Aβ) accumulation. BMS-708163 (avagacestat) is an oral GSI designed for selective inhibition of Aβ synthesis currently in development for the treatment of mild to moderate and predementia AD. In addition to the desired effect on Aβ synthesis, GSIs affect Notch processing, which is thought to mediate some toxic adverse effects reported with this drug class. Avagacestat produced up to 190-fold greater selectivity for Aβ synthesis than Notch processing in preclinical studies and may therefore produce less toxic adverse events than other less selective compounds. Presented here are the results of the first in-human study for this new GSI compound. OBJECTIVE The goal of this study was to assess the tolerability profile, pharmacokinetic properties, and effects on pharmacodynamic markers (Aβ, trefoil factor family 3 protein, dual specificity phosphatase 6, and hairy and enhancer of split-1) of single, oral doses of avagacestat in healthy, young, male volunteers. METHODS This was a multicenter, randomized, double-blind, placebo-controlled, single-ascending dose study in 8 healthy young men (age, 18-45 years) per dosing panel. Each study participant was randomized to receive a single dose of placebo (n = 2) or avagacestat (n = 6 for each dose) as an oral solution in 1 of 9 sequential dose panels (0.3, 1.5, 5, 15, 50, 100, 200, 400, and 800 mg). For determination of avagacestat, blood samples were obtained before dosing and for up to 144 hours after dosing. For participants in the 800-mg avagacestat dose panel, additional samples were obtained at 216, 312, and 648 hours. For 40-amino acid isoform of Aβ (Aβ(1-40)) assessment, plasma samples were collected before avagacestat administration and up to 72 hours after dosing. RESULTS Avagacestat concentrations peaked quickly after oral administration and then had a biphasic decrease in concentrations with a prolonged terminal phase. Exposures were proportional with doses up to 200 mg. Avagacestat was well tolerated at single oral doses up to 800 mg, with a biphasic effect on plasma Aβ(1-40). Adverse events were predominately mild to moderate in severity with no evidence of dose dependence up to 200 mg. CONCLUSIONS Results from this single-ascending dose study suggest that avagacestat was tolerated at a single-dose range of 0.3 to 800 mg and suitable for further clinical development.


Journal of Pharmacology and Experimental Therapeutics | 2013

Pharmacodynamics of Selective Inhibition of γ -Secretase by Avagacestat

Charles F. Albright; Randy C. Dockens; Jere E. Meredith; Richard E. Olson; Randy Slemmon; Kimberley A. Lentz; Jun-Sheng Wang; Rex Denton; Gary Pilcher; Paul Rhyne; Joseph Raybon; Donna M. Barten; Catherine R. Burton; Jeremy H. Toyn; Sethu Sankaranarayanan; Craig Polson; Valerie Guss; Randy White; Frank Simutis; Thomas P. Sanderson; Kevin W. Gillman; John E. Starrett; Joanne J. Bronson; Oleksandr Sverdlov; Shu-Pang Huang; Lorna Castaneda; Howard Feldman; Vlad Coric; Robert Zaczek; John E. Macor

A hallmark of Alzheimer’s disease (AD) pathology is the accumulation of brain amyloid β-peptide (Aβ), generated by γ-secretase-mediated cleavage of the amyloid precursor protein (APP). Therefore, γ-secretase inhibitors (GSIs) may lower brain Aβ and offer a potential new approach to treat AD. As γ-secretase also cleaves Notch proteins, GSIs can have undesirable effects due to interference with Notch signaling. Avagacestat (BMS-708163) is a GSI developed for selective inhibition of APP over Notch cleavage. Avagacestat inhibition of APP and Notch cleavage was evaluated in cell culture by measuring levels of Aβ and human Notch proteins. In rats, dogs, and humans, selectivity was evaluated by measuring plasma blood concentrations in relation to effects on cerebrospinal fluid (CSF) Aβ levels and Notch-related toxicities. Measurements of Notch-related toxicity included goblet cell metaplasia in the gut, marginal-zone depletion in the spleen, reductions in B cells, and changes in expression of the Notch-regulated hairy and enhancer of split homolog-1 from blood cells. In rats and dogs, acute administration of avagacestat robustly reduced CSF Aβ40 and Aβ42 levels similarly. Chronic administration in rats and dogs, and 28-day, single- and multiple-ascending–dose administration in healthy human subjects caused similar exposure-dependent reductions in CSF Aβ40. Consistent with the 137-fold selectivity measured in cell culture, we identified doses of avagacestat that reduce CSF Aβ levels without causing Notch-related toxicities. Our results demonstrate the selectivity of avagacestat for APP over Notch cleavage, supporting further evaluation of avagacestat for AD therapy.


Alzheimers & Dementia | 2013

Frontotemporal degeneration, the next therapeutic frontier: Molecules and animal models for frontotemporal degeneration drug development

Adam L. Boxer; Michael Gold; Edward D. Huey; Fen-Biao Gao; Edward A. Burton; Tiffany W. Chow; Aimee W. Kao; Blair R. Leavitt; Bruce T. Lamb; Megan Grether; David S. Knopman; Nigel J. Cairns; Ian R. Mackenzie; Laura L. Mitic; Erik D. Roberson; Daniel Van Kammen; Marc Cantillon; Kathleen R. Zahs; Stephen Salloway; John C. Morris; Gary Tong; Howard Feldman; Howard Fillit; Susan Dickinson; Zaven S. Khachaturian; Margaret Sutherland; Robert V. Farese; Bruce L. Miller; Jeffrey L. Cummings

Frontotemporal degeneration (FTD) is a common cause of dementia for which there are currently no approved therapies. Over the past decade, there has been an explosion of knowledge about the biology and clinical features of FTD that has identified a number of promising therapeutic targets as well as animal models in which to develop drugs. The close association of some forms of FTD with neuropathological accumulation of tau protein or increased neuroinflammation due to progranulin protein deficiency suggests that a drugs success in treating FTD may predict efficacy in more common diseases such as Alzheimers disease. A variety of regulatory incentives, clinical features of FTD such as rapid disease progression, and relatively pure molecular pathology suggest that there are advantages to developing drugs for FTD as compared with other more common neurodegenerative diseases such as Alzheimers disease. In March 2011, the Frontotemporal Degeneration Treatment Study Group sponsored a conference entitled “FTD, the Next Therapeutic Frontier,” which focused on preclinical aspects of FTD drug development. The goal of the meeting was to promote collaborations between academic researchers and biotechnology and pharmaceutical researchers to accelerate the development of new treatments for FTD. Here we report the key findings from the conference, including the rationale for FTD drug development; epidemiological, genetic, and neuropathological features of FTD; FTD animal models and how best to use them; and examples of successful drug development collaborations in other neurodegenerative diseases.


Alzheimers & Dementia | 2013

The advantages of frontotemporal degeneration drug development (part 2 of frontotemporal degeneration: The next therapeutic frontier)

Adam L. Boxer; Michael Gold; Edward D. Huey; William T. Hu; Howard J. Rosen; Joel H. Kramer; Fen-Biao Gao; Edward A. Burton; Tiffany W. Chow; Aimee W. Kao; Blair R. Leavitt; Bruce T. Lamb; Megan Grether; David S. Knopman; Nigel J. Cairns; Ian R. Mackenzie; Laura L. Mitic; Erik D. Roberson; Daniel Van Kammen; Marc Cantillon; Kathleen R. Zahs; George R. Jackson; Stephen Salloway; John C. Morris; Gary Tong; Howard Feldman; Howard Fillit; Susan Dickinson; Zaven S. Khachaturian; Margaret Sutherland

Frontotemporal degeneration (FTD) encompasses a spectrum of related neurodegenerative disorders with behavioral, language, and motor phenotypes for which there are currently no effective therapies. This is the second of two articles that summarize the presentations and discussions that occurred at two symposia in 2011 sponsored by the Frontotemporal Degeneration Treatment Study Group, a collaborative group of academic and industry researchers that is devoted to developing treatments for FTD. This article discusses the current status of FTD clinical research that is relevant to the conduct of clinical trials, and why FTD research may be an attractive pathway for developing therapies for neurodegenerative disorders. The clinical and molecular features of FTD, including rapid disease progression and relatively pure molecular pathology, suggest that there are advantages to developing drugs for FTD as compared with other dementias. FTD qualifies as orphan indication, providing additional advantages for drug development. Two recent sets of consensus diagnostic criteria will facilitate the identification of patients with FTD, and a variety of neuropsychological, functional, and behavioral scales have been shown to be sensitive to disease progression. Moreover, quantitative neuroimaging measurements demonstrate progressive brain atrophy in FTD at rates that may surpass Alzheimers disease. Finally, the similarities between FTD and other neurodegenerative diseases with drug development efforts already underway suggest that FTD researchers will be able to draw on this experience to create a road map for FTD drug development. We conclude that FTD research has reached sufficient maturity to pursue clinical development of specific FTD therapies.


Journal of Medicinal Chemistry | 2012

Discovery of (5S,6S,9R)-5-Amino-6-(2,3-difluorophenyl)-6,7,8,9-tetrahydro-5H-cyclohepta[b]pyridin-9-yl 4-(2-oxo-2,3-dihydro-1H-imidazo[4,5-b]pyridin-1-yl)piperidine-1-carboxylate (BMS-927711): An Oral Calcitonin Gene-Related Peptide (CGRP) Antagonist in Clinical Trials for Treating Migraine

Guanglin Luo; Ling Chen; Charles M. Conway; Rex Denton; Deborah Keavy; Laura Signor; Walter Kostich; Kimberley A. Lentz; Kenneth S. Santone; Richard Schartman; Marc Browning; Gary Tong; John G. Houston; Gene M. Dubowchik; John E. Macor

Calcitonin gene-related peptide (CGRP) receptor antagonists have demonstrated clinical efficacy in the treatment of acute migraine. Herein, we describe the design, synthesis, and preclinical characterization of a highly potent, oral CGRP receptor antagonist BMS-927711 (8). Compound 8 has good oral bioavailability in rat and cynomolgus monkey, attractive overall preclinical properties, and shows dose-dependent activity in a primate model of CGRP-induced facial blood flow. Compound 8 is presently in phase II clinical trials.


Alzheimer's Research & Therapy | 2015

An integrated multi-study analysis of intra-subject variability in cerebrospinal fluid amyloid-β concentrations collected by lumbar puncture and indwelling lumbar catheter

Brendan P. Lucey; Celedon Gonzales; Ujjwas Das; Jinhe Li; Eric Siemers; J. Randall Slemmon; Randall J. Bateman; Yafei Huang; Gerard B. Fox; Jurgen A.H.R. Claassen; Diane Slats; Marcel M. Verbeek; Gary Tong; Holly Soares; Mary J. Savage; Matthew E. Kennedy; Magnus Sjögren; Richard Margolin; Xia Chen; Martin R. Farlow; Robert A. Dean; Jeffrey F. Waring

IntroductionAmyloid-β (Aβ) has been investigated as a diagnostic biomarker and therapeutic drug target. Recent studies found that cerebrospinal fluid (CSF) Aβ fluctuates over time, including as a diurnal pattern, and increases in absolute concentration with serial collection. It is currently unknown what effect differences in CSF collection methodology have on Aβ variability. In this study, we sought to determine the effect of different collection methodologies on the stability of CSF Aβ concentrations over time.MethodsGrouped analysis of CSF Aβ levels from multiple industry and academic groups collected by either lumbar puncture (n=83) or indwelling lumbar catheter (n=178). Participants were either placebo or untreated subjects from clinical drug trials or observational studies. Participants had CSF collected by lumbar puncture or lumbar catheter for quantitation of Aβ concentration by enzyme linked immunosorbent assay. Data from all sponsors was converted to percent of the mean for Aβ40 and Aβ42 for comparison. Repeated measures analysis of variance was performed to assess for factors affecting the linear rise of Aβ concentrations over time.ResultsAnalysis of studies collecting CSF via lumbar catheter revealed tremendous inter-subject variability of Aβ40 and Aβ42 as well as an Aβ diurnal pattern in all of the sponsors’ studies. In contrast, Aβ concentrations from CSF samples collected at two time points by lumbar puncture showed no significant differences. Repeated measures analysis of variance found that only time and draw frequency were significantly associated with the slope of linear rise in Aβ40 and Aβ42 concentrations during the first 6 hours of collection.ConclusionsBased on our findings, we recommend minimizing the frequency of CSF draws in studies measuring Aβ levels and keeping the frequency standardized between experimental groups. The Aβ diurnal pattern was noted in all sponsors’ studies and was not an artifact of study design. Averaging Aβ concentrations at each time point is recommended to minimize the effect of individual variability. Indwelling lumbar catheters are an invaluable research tool for following changes in CSF Aβ over 24-48 hours, but factors affecting Aβ concentration such as linear rise and diurnal variation need to be accounted for in planning study designs.


British Journal of Clinical Pharmacology | 2013

A contrast in safety, pharmacokinetics and pharmacodynamics across age groups after a single 50 mg oral dose of the γ-secretase inhibitor avagacestat

Gary Tong; Jun-Sheng Wang; Oleksandr Sverdlov; Shu-Pang Huang; Randy Slemmon; Robert Croop; Lorna Castaneda; Huidong Gu; Oi Wong; Hewei Li; Robert M. Berman; Christina Smith; Charles F. Albright; Randy C. Dockens

AIM To evaluate the single dose pharmacokinetics, pharmacodynamics, and preliminary tolerability of the γ-secretase inhibitor BMS-708163 (avagacestat) in young and elderly men and women. METHODS All subjects received double-blinded administration of a single 50 mg dose of avagacestat in capsule form or matching placebo. Main evaluations included pharmacokinetics, safety, plasma amyloid-β (Aβ)(1-40) concentratios and exploration of Notch biomarkers. RESULTS Avagacestat 50 mg capsule was well tolerated and rapidly absorbed among young and elderly subjects, with a median t(max) between 1 and 2 h post dose and an average half-life between 41 and 71 h. In general, subjects aged 75 years or more had higher AUC(0,∞) values than those aged less than 75 years. An exploratory analysis of Aβ(1-40) serum concentrations showed a pattern of decreasing concentrations over the first 4-6 h followed by a rise above baseline that was maintained until the end of the assessment period. Adverse events were generally mild, occurring more frequently in elderly subjects, with no observed difference between subjects receiving avagacestat and placebo. No dose limiting gastrointestinal effects of avagacestat were observed and exploratory biomarkers of Notch inhibition did not change significantly. CONCLUSIONS The favourable safety profile and pharmacokinetic effects of avagacestat in this study support its continued development, especially in the target population of elderly subjects with mild cognitive impairment or Alzheimers disease.


Alzheimers & Dementia | 2010

The safety, tolerability, pharmacokinetics and pharmacodynamics of single doses of BMS-708163 in young and elderly subjects

Gary Tong; Jun-Sheng Wang; Alex Sverdlov; Shu-Pang Huang; Randy Slemmon; Robert Croop; Lorna Castaneda; Huidong Gu; Oi Wong; Hewei Li; Robert M. Berman; Christina Smith; Charles F. Albright; Randy C. Dockens; Santiago Arroyo

improved cognitive function in a mouse model of AD (APP/PS1). Methods: To track BMDCs, bone marrow harvested from the mice carrying green fluorescent protein (GFP) was transplanted to the APP/PS1 mice. Thereafter, SCF+G-CSF were subcutaneous administered for 12 days, beginning at the age of 8 months when Ab plaques had appeared in the brain. To evaluate special learning and memory, water maze test was performed before and 9 months after treatment. Results: We observed that SCF+G-CSF-treated mice showed significantly shorter escape latency and that the number of senile plaques was significantly reduced by SCF+G-CSF treatment. In addition, a significant high number of GFP+/Iba1+ cells surrounding Ab plaques was seen in SCF+G-CSF-treated mice. Conclusions: These data suggest that SCF+G-CSF treatment may delay AD progress by enhancing clearance of Ab deposits via BM-derived microglial cells. These data would help in developing an effective treatment to improve health of AD.


Alzheimers & Dementia | 2018

The National Institute on Aging and the Alzheimer's Association Research Framework for Alzheimer's disease : Perspectives from the Research Roundtable

David S. Knopman; Samantha Budd Haeberlein; Maria C. Carrillo; James Hendrix; Geoff Kerchner; Richard Margolin; Paul Maruff; David S. Miller; Gary Tong; Maria B. Tome; Melissa E. Murray; Peter T. Nelson; Mary Sano; Niklas Mattsson; David L. Sultzer; Thomas J. Montine; Clifford R. Jack; Hartmuth C. Kolb; Ronald C. Petersen; Prashanthi Vemuri; Megan Zoschg Canniere; Julie A. Schneider; Susan M. Resnick; Gary Romano; Argonde C. van Harten; David A. Wolk; Lisa J. Bain; Eric Siemers

The Alzheimers Associations Research Roundtable met in November 2017 to explore the new National Institute on Aging and the Alzheimers Association Research Framework for Alzheimers disease. The meeting allowed experts in the field from academia, industry, and government to provide perspectives on the new National Institute on Aging and the Alzheimers Association Research Framework. This review will summarize the “A, T, N System” (Amyloid, Tau, and Neurodegeneration) using biomarkers and how this may be applied to clinical research and drug development. In addition, challenges and barriers to the potential adoption of this new framework will be discussed. Finally, future directions for research will be proposed.


Alzheimers & Dementia | 2011

An assessment of variability in CSF biomarkers in clinical experimental models: a meta-analysis

Jeffrey F. Waring; Diane Slats; Celedon Gonzales; Robert A. Dean; David Lee; Eric Siemers; Jurgen A.H.R. Claassen; Jinhe Li; David LeBlond; Randy Slemmon; Randall J. Bateman; Gary Tong; Gerard B. Fox

all biomarkers considered for this review. Six percent of studies included more than 500 patients. It was unclear whether scan/test interpretation was conducted blind to knowledge of conversion to dementia in 62% of reports. Similarly, it was unclear whether decisions about conversion to dementia were made without knowledge of scan/test results in 63% of reports. Uninterpretable or intermediate test results were either not reported, or reported unclearly in 79% of cases. It was rarely possible to make an assessment, on the basis of the report, that the spectrum of patients in the cohort was representative of the patients whowould receive the test in practice. 21 studies of CSF or plasma Abeta reported conversion in 1128 patients. 26 studies of CSF tau reported conversion in 1054 patients. 22 structural MRI studies reported conversion in 1343 patients. 11 studies of FDG-PET reported conversion in 270 patients. 5 studies of PET based amyloid imaging reported conversion in 27 patients.Conclusions: The quality of reporting of longitudinal studies of biomarkers raises the possibility that substantial biases may be present. Multiple publication is commonplace in studies of diagnostic accuracy in dementia. Whilst this reflects developing techniques and continuing monitoring of subjects for outcomes, greater transparency is required so as to allow easier compilation of these data. A more formal process with cohort registration, reporting of ’intention to diagnose’ data, and clear independence of clinical and test evaluations is required. PET-based tests have the smallest evidence base. The small sample size of most studies limits their overall generalizeabilty. Funders, including those seeking licences for new products, should support large-scale, representative evaluations in that part of the population who would be most likely to utilize the test. Critical evaluation of the evidence base for diagnostic biomarkers is of major importance to the field of dementia. Without it, there is a risk that future clinical care and research will be built on assumptions about diagnostic validity that are wrong.

Collaboration


Dive into the Gary Tong's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hewei Li

Bristol-Myers Squibb

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge