Network


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

Hotspot


Dive into the research topics where Pamela L. Tannenbaum is active.

Publication


Featured researches published by Pamela L. Tannenbaum.


Journal of Neurogenetics | 2011

Promotion of Sleep by Suvorexant—A Novel Dual Orexin Receptor Antagonist

Christopher J. Winrow; Anthony L. Gotter; Christopher D. Cox; Scott M. Doran; Pamela L. Tannenbaum; Michael J. Breslin; Susan L. Garson; Steven V. Fox; Charles M. Harrell; Joanne Stevens; Duane R. Reiss; Donghui Cui; Paul J. Coleman; John J. Renger

Abstract: Orexins/hypocretins are key neuropeptides responsible for regulating central arousal and reward circuits. Two receptors respond to orexin signaling, orexin 1 receptor (OX1R) and orexin 2 receptor (OX2R) with partially overlapping nervous system distributions. Genetic studies suggest orexin receptor antagonists could be therapeutic for insomnia and other disorders with disruptions of sleep and wake. Suvorexant (MK-4305) is a potent, selective, and orally bioavailable antagonist of OX1R and OX2R currently under clinical investigation as a novel therapy for insomnia. Examination of Suvorexant in radioligand binding assays using tissue from transgenic rats expressing the human OX2R found nearly full receptor occupancy (>90%) at plasma exposures of 1.1 μM. Dosed orally Suvorexant significantly and dose-dependently reduced locomotor activity and promoted sleep in rats (10, 30, and 100 mg/kg), dogs (1 and 3 mg/kg), and rhesus monkeys (10 mg/kg). Consistent cross-species sleep/wake architecture changes produced by Suvorexant highlight a unique opportunity to develop dual orexin antagonists as a novel therapy for insomnia.


ChemMedChem | 2012

Discovery of [(2R,5R)-5-{[(5-fluoropyridin-2-yl)oxy]methyl}-2-methylpiperidin-1-yl][5-methyl-2-(pyrimidin-2-yl)phenyl]methanone (MK-6096): a dual orexin receptor antagonist with potent sleep-promoting properties.

Paul J. Coleman; John D. Schreier; Christopher D. Cox; Michael J. Breslin; David B. Whitman; Michael J. Bogusky; Georgia B. McGaughey; Rodney A. Bednar; Wei Lemaire; Scott M. Doran; Steven V. Fox; Susan L. Garson; Anthony L. Gotter; C. Meacham Harrell; Duane R. Reiss; Tamara D. Cabalu; Donghui Cui; Thomayant Prueksaritanont; Joanne Stevens; Pamela L. Tannenbaum; Richard G. Ball; Joyce Stellabott; Steven D. Young; George D. Hartman; Christopher J. Winrow; John J. Renger

Insomnia is a common disorder that can be comorbid with other physical and psychological illnesses. Traditional management of insomnia relies on general central nervous system (CNS) suppression using GABA modulators. Many of these agents fail to meet patient needs with respect to sleep onset, maintenance, and next‐day residual effects and have issues related to tolerance, memory disturbances, and balance. Orexin neuropeptides are central regulators of wakefulness, and orexin antagonism has been identified as a novel mechanism for treating insomnia with clinical proof of concept. Herein we describe the discovery of a series of α‐methylpiperidine carboxamide dual orexin 1 and orexin 2 receptor (OX1R/OX2R) antagonists (DORAs). The design of these molecules was inspired by earlier work from this laboratory in understanding preferred conformational properties for potent orexin receptor binding. Minimization of 1,3‐allylic strain interactions was used as a design principle to synthesize 2,5‐disubstituted piperidine carboxamides with axially oriented substituents including DORA 28. DORA 28 (MK‐6096) has exceptional in vivo activity in preclinical sleep models, and has advanced into phase II clinical trials for the treatment of insomnia.


Science Translational Medicine | 2013

Orexin Receptor Antagonists Differ from Standard Sleep Drugs by Promoting Sleep at Doses That Do Not Disrupt Cognition

Jason M. Uslaner; Spencer J. Tye; Donnie M. Eddins; Xiaohai Wang; Steven V. Fox; Alan T. Savitz; Jacquelyn Binns; Christopher E. Cannon; Susan L. Garson; Lihang Yao; Robert Hodgson; Joanne Stevens; Mark R. Bowlby; Pamela L. Tannenbaum; Joseph Brunner; Terrence P. McDonald; Anthony L. Gotter; Scott D. Kuduk; Paul J. Coleman; Christopher J. Winrow; John J. Renger

Selective orexin receptor antagonism promotes sleep at doses lower than those that impair attention and memory in contrast to the GABA receptor–positive allosteric modulators currently in use. Sleep Without the After Effects Currently available treatments for insomnia can produce a number of central nervous system–based cognitive side effects, including the potential to impair memory and attention. Recently, selective dual orexin receptor antagonists, such as suvorexant and almorexant, have been shown to promote sleep onset and maintenance in clinical trials for patients with insomnia. In new work, Uslaner and colleagues compared sleep-promoting doses to the cognitive-impairing doses for an orexin receptor antagonist, DORA-22, versus sleep drugs currently in use: zolpidem, diazepam, or eszopiclone. At doses that produced equivalent amounts of sleep in rat and rhesus monkey, zolpidem, diazepam, and eszopiclone significantly disrupted attention and memory, whereas DORA-22 promoted sleep at doses that did not exert measurable effects on cognition. Furthermore, when compared to the other insomnia treatments that modulate γ-aminobutyric acid (GABA) receptor function, the authors saw greater separation for orexin receptor antagonism between doses that promoted sleep and doses that reduced expression of a hippocampal gene involved in synaptic plasticity called Arc. These findings suggest that dual orexin receptor antagonists might provide an effective treatment for insomnia with a greater therapeutic margin for sleep versus cognitive disturbances compared to the GABAA-positive allosteric modulators currently available. Current treatments for insomnia, such as zolpidem (Ambien) and eszopiclone (Lunesta), are γ-aminobutyric acid type A (GABAA)–positive allosteric modulators that carry a number of side effects including the potential to disrupt cognition. In an effort to develop better tolerated medicines, we have identified dual orexin 1 and 2 receptor antagonists (DORAs), which promote sleep in preclinical animal models and humans. We compare the effects of orally administered eszopiclone, zolpidem, and diazepam to the dual orexin receptor antagonist DORA-22 on sleep and the novel object recognition test in rat, and on sleep and two cognition tests (delayed match to sample and serial choice reaction time) in the rhesus monkey. Each compound’s minimal dose that promoted sleep versus the minimal dose that exerted deficits in these cognitive tests was determined, and a therapeutic margin was established. We found that DORA-22 has a wider therapeutic margin for sleep versus cognitive impairment in rat and rhesus monkey compared to the other compounds tested. These data were further supported with the demonstration of a wider therapeutic margin for DORA-22 compared to the other compounds on sleep versus the expression of hippocampal activity–regulated cytoskeletal-associated protein (Arc), an immediate-early gene product involved in synaptic plasticity. These findings suggest that DORAs might provide an effective treatment for insomnia with a greater therapeutic margin for sleep versus cognitive disturbances compared to the GABAA-positive allosteric modulators currently in use.


ChemMedChem | 2014

Discovery of 5′′-Chloro-N-[(5,6-dimethoxypyridin-2-yl)methyl]-2,2′:5′,3′′-terpyridine-3′-carboxamide (MK-1064): A Selective Orexin 2 Receptor Antagonist (2-SORA) for the Treatment of Insomnia

Anthony J. Roecker; Swati P. Mercer; John D. Schreier; Christopher D. Cox; Mark E. Fraley; Justin T. Steen; Wei Lemaire; Joseph G. Bruno; C. Meacham Harrell; Susan L. Garson; Anthony L. Gotter; Steven V. Fox; Joanne Stevens; Pamela L. Tannenbaum; Thomayant Prueksaritanont; Tamara D. Cabalu; Donghui Cui; Joyce Stellabott; George D. Hartman; Steven D. Young; Christopher J. Winrow; John J. Renger; Paul J. Coleman

The field of small‐molecule orexin antagonist research has evolved rapidly in the last 15 years from the discovery of the orexin peptides to clinical proof‐of‐concept for the treatment of insomnia. Clinical programs have focused on the development of antagonists that reversibly block the action of endogenous peptides at both the orexin 1 and orexin 2 receptors (OX1R and OX2R), termed dual orexin receptor antagonists (DORAs), affording late‐stage development candidates including Merck’s suvorexant (new drug application filed 2012). Full characterization of the pharmacology associated with antagonism of either OX1R or OX2R alone has been hampered by the dearth of suitable subtype‐selective, orally bioavailable ligands. Herein, we report the development of a selective orexin 2 antagonist (2‐SORA) series to afford a potent, orally bioavailable 2‐SORA ligand. Several challenging medicinal chemistry issues were identified and overcome during the development of these 2,5‐disubstituted nicotinamides, including reversible CYP inhibition, physiochemical properties, P‐glycoprotein efflux and bioactivation. This article highlights structural modifications the team utilized to drive compound design, as well as in vivo characterization of our 2‐SORA clinical candidate, 5′′‐chloro‐N‐[(5,6‐dimethoxypyridin‐2‐yl)methyl]‐2,2′:5′,3′′‐terpyridine‐3′‐carboxamide (MK‐1064), in mouse, rat, dog, and rhesus sleep models.


ChemMedChem | 2012

Discovery of a Pharmacologically Active Antagonist of the Two-Pore-Domain Potassium Channel K2P9.1 (TASK-3)

Craig A. Coburn; Yunfu Luo; Mingxiang Cui; Jiabing Wang; Richard Soll; Jingchao Dong; Bin Hu; Michael A. Lyon; Vincent P. Santarelli; Richard L. Kraus; Yun Gregan; Yi. Wang; Steven V. Fox; Jacquelyn Binns; Scott M. Doran; Duane R. Reiss; Pamela L. Tannenbaum; Anthony L. Gotter; Peter T. Meinke; John J. Renger

TWIK‐related acid‐sensitive K+ (K2P9.1, TASK‐3) ion channels have the capacity to regulate the activity of neuronal pathways by influencing the resting membrane potential of neurons on which they are expressed. The central nervous system (CNS) expression of these channels suggests potential roles in neurologic disorders, and it is believed that the development of TASK‐3 antagonists could lead to the therapeutic treatment of a number of neurological conditions. While a therapeutic potential for TASK‐3 channel modulation exists, there are only a few documented examples of potent and selective small‐molecule channel blockers. Herein, we describe the discovery and lead optimization efforts for a novel series of TASK‐3 channel antagonists based on a 5,6,7,8‐tetrahydropyrido[4,3‐d]pyrimidine high‐throughput screening lead from which a subseries of potent and selective inhibitors were identified. One compound was profiled in detail with respect to its physical properties and demonstrated pharmacological target engagement as indicated by its ability to modulate sleep architecture in rodent electroencephalogram (EEG) telemetry models.


Neuropsychopharmacology | 2013

Quantitative electroencephalography within sleep/wake states differentiates GABAA modulators eszopiclone and zolpidem from dual orexin receptor antagonists in rats.

Steven V. Fox; Anthony L. Gotter; Spencer J. Tye; Susan L. Garson; Alan T. Savitz; Jason M. Uslaner; Joseph Brunner; Pamela L. Tannenbaum; Terrence P. McDonald; Robert Hodgson; Lihang Yao; Mark R Bowlby; Scott D. Kuduk; Paul J. Coleman; Richard Hargreaves; Christopher J. Winrow; John J. Renger

Dual orexin receptor antagonists (DORAs) induce sleep by blocking orexin 1 and orexin 2 receptor-mediated activities responsible for regulating wakefulness. DORAs represent a potential alternative mechanism to the current standard of care that includes the γ-aminobutyric acid (GABA)A receptor-positive allosteric modulators, eszopiclone and zolpidem. This work uses an innovative method to analyze electroencephalogram (EEG) spectral frequencies within sleep/wake states to differentiate the effects of GABAA modulators from DORA-22, an analog of the DORA MK-6096, in Sprague–Dawley rats. The effects of low, intermediate, and high doses of eszopiclone, zolpidem, and DORA-22 were examined after first defining each compound’s ability to promote sleep during active-phase dosing. The EEG spectral frequency power within specific sleep stages was calculated in 1-Hz intervals from 1 to 100 Hz within each sleep/wake state for the first 4 h after the dose. Eszopiclone and zolpidem produced marked, dose-responsive disruptions in sleep stage-specific EEG spectral profiles compared with vehicle treatment. In marked contrast, DORA-22 exhibited marginal changes in the spectral profile, observed only during rapid eye movement sleep, and only at the highest dose tested. Moreover, while eszopiclone- and zolpidem-induced changes were evident in the inactive period, the EEG spectral responses to DORA-22 were absent during this phase. These results suggest that DORA-22 differs from eszopiclone and zolpidem whereby DORA-22 promotes somnolence without altering the neuronal network EEG activity observed during normal sleep.


Brain Research | 2011

TASK-3 as a potential antidepressant target

Anthony L. Gotter; Vincent P. Santarelli; Scott M. Doran; Pamela L. Tannenbaum; Richard L. Kraus; Thomas W. Rosahl; Hamid Meziane; Marina Montial; Duane R. Reiss; Keith Wessner; Alexander McCampbell; Joanne Stevens; Joseph Brunner; Steven V. Fox; Victor N. Uebele; Douglas A. Bayliss; Christopher J. Winrow; John J. Renger

Modulation of TASK-3 (Kcnk9) potassium channels affect neurotransmitter release in thalamocortical centers and other sleep-related nuclei having the capacity to regulate arousal cycles and REM sleep changes associated with mood disorders and antidepressant action. Circumstantial evidence from this and previous studies suggest the potential for TASK-3 to be a novel antidepressant therapeutic target; TASK-3 knock-out mice display augmented circadian amplitude and exhibit sleep architecture characterized by suppressed REM activity. Detailed analysis of locomotor activity indicates that the amplitudes of activity bout duration and bout number are augmented in TASK-3 mutants well beyond that seen in wildtypes, findings substantiated by amplitude increases in body temperature and EEG recordings of sleep stage bouts. Polysomnographic analysis of TASK-3 mutants reveals increases in nocturnal active wake and suppressed REM sleep time while increased slow wave sleep typifies the inactive phase, findings that have implications for the cognitive impact of reduced TASK-3 activity. In direct measures of their resistance to despair behavior, TASK-3 knock-outs displayed significant decreases in immobility relative to wildtype controls in both tail suspension and forced swim tests. Treatment of wildtype animals with the antidepressant Fluoxetine markedly reduced REM sleep, while leaving active wake and slow wave sleep relatively intact. Remarkably, these effects were absent in TASK-3 mutants indicating that TASK-3 is either directly involved in the mechanism of this drugs action, or participates in parallel pathways that achieve the same effect. Together, these results support the TASK-3 channel to act as a therapeutic target for antidepressant action.


Scientific Reports | 2016

Orexin 2 Receptor Antagonism is Sufficient to Promote NREM and REM Sleep from Mouse to Man

Anthony L. Gotter; Charles M. Harrell; Joanne Stevens; Vladimir Svetnik; Ka Lai Yee; Xiaodong Li; Anthony J. Roecker; Steven V. Fox; Pamela L. Tannenbaum; Susan L. Garson; Inge De Lepeleire; Nicole Calder; Laura Rosen; Arie Struyk; Paul J. Coleman; W. Joseph Herring; John J. Renger; Christopher J. Winrow

Orexin neuropeptides regulate sleep/wake through orexin receptors (OX1R, OX2R); OX2R is the predominant mediator of arousal promotion. The potential for single OX2R antagonism to effectively promote sleep has yet to be demonstrated in humans. MK-1064 is an OX2R-single antagonist. Preclinically, MK-1064 promotes sleep and increases both rapid eye movement (REM) and non-REM (NREM) sleep in rats at OX2R occupancies higher than the range observed for dual orexin receptor antagonists. Similar to dual antagonists, MK-1064 increases NREM and REM sleep in dogs without inducing cataplexy. Two Phase I studies in healthy human subjects evaluated safety, tolerability, pharmacokinetics and sleep-promoting effects of MK-1064, and demonstrated dose-dependent increases in subjective somnolence (via Karolinska Sleepiness Scale and Visual Analogue Scale measures) and sleep (via polysomnography), including increased REM and NREM sleep. Thus, selective OX2R antagonism is sufficient to promote REM and NREM sleep across species, similarly to that seen with dual orexin receptor antagonism.


Frontiers in Neuroscience | 2013

Dual orexin receptor antagonists show distinct effects on locomotor performance, ethanol interaction and sleep architecture relative to gamma-aminobutyric acid-A receptor modulators

Andres D. Ramirez; Anthony L. Gotter; Steven V. Fox; Pamela L. Tannenbaum; Lihang Yao; Spencer J. Tye; Terrence P. McDonald; Joseph Brunner; Susan L. Garson; Duane R. Reiss; Scott D. Kuduk; Paul J. Coleman; Jason M. Uslaner; Robert Hodgson; Susan E. Browne; John J. Renger; Christopher J. Winrow

Dual orexin receptor antagonists (DORAs) are a potential treatment for insomnia that function by blocking both the orexin 1 and orexin 2 receptors. The objective of the current study was to further confirm the impact of therapeutic mechanisms targeting insomnia on locomotor coordination and ethanol interaction using DORAs and gamma-aminobutyric acid (GABA)-A receptor modulators of distinct chemical structure and pharmacological properties in the context of sleep-promoting potential. The current study compared rat motor co-ordination after administration of DORAs, DORA-12 and almorexant, and GABA-A receptor modulators, zolpidem, eszopiclone, and diazepam, alone or each in combination with ethanol. Motor performance was assessed by measuring time spent walking on a rotarod apparatus. Zolpidem, eszopiclone and diazepam [0.3–30 mg/kg administered orally (PO)] impaired rotarod performance in a dose-dependent manner. Furthermore, all three GABA-A receptor modulators potentiated ethanol- (0.25–1.5 g/kg) induced impairment on the rotarod. By contrast, neither DORA-12 (10–100 mg/kg, PO) nor almorexant (30–300 mg/kg, PO) impaired motor performance alone or in combination with ethanol. In addition, distinct differences in sleep architecture were observed between ethanol, GABA-A receptor modulators (zolpidem, eszopiclone, and diazepam) and DORA-12 in electroencephalogram studies in rats. These findings provide further evidence that orexin receptor antagonists have an improved motor side-effect profile compared with currently available sleep-promoting agents based on preclinical data and strengthen the rationale for further evaluation of these agents in clinical development.


BMC Neuroscience | 2014

Differential sleep-promoting effects of dual orexin receptor antagonists and GABAA receptor modulators

Anthony L. Gotter; Susan L. Garson; Joanne Stevens; Regina L Munden; Steven V. Fox; Pamela L. Tannenbaum; Lihang Yao; Scott D. Kuduk; Terrence P. McDonald; Jason M. Uslaner; Spencer J. Tye; Paul J. Coleman; Christopher J. Winrow; John J. Renger

BackgroundThe current standard of care for insomnia includes gamma-aminobutyric acid receptor A (GABAA) activators, which promote sleep as well as general central nervous system depression. Dual orexin receptor antagonists (DORAs) represent an alternative mechanism for insomnia treatment that induces somnolence by blocking the wake-promoting effects of orexin neuropeptides. The current study compares the role and interdependence of these two mechanisms on their ability to influence sleep architecture and quantitative electroencephalography (qEEG) spectral profiles across preclinical species.ResultsActive-phase dosing of DORA-22 induced consistent effects on sleep architecture in mice, rats, dogs, and rhesus monkeys; attenuation of active wake was accompanied by increases in both non─rapid eye movement (NREM) and rapid eye movement (REM) sleep. Eszopiclone, a representative GABAA receptor modulator, promoted sleep in rats and rhesus monkeys that was marked by REM sleep suppression, but had inconsistent effects in mice and paradoxically promoted wakefulness in dogs. Active-phase treatment of rats with DORA-12 similarly promoted NREM and REM sleep to magnitudes nearly identical to those seen during normal resting-phase sleep following vehicle treatment, whereas eszopiclone suppressed REM even to levels below those seen during the active phase. The qEEG changes induced by DORA-12 in rats also resembled normal resting-phase patterns, whereas eszopiclone induced changes distinct from normal active- or inactive-phase spectra. Co-dosing experiments, as well as studies in transgenic rats lacking orexin neurons, indicated partial overlap in the mechanism of sleep promotion by orexin and GABA modulation with the exception of the REM suppression exclusive to GABAA receptor modulation. Following REM deprivation in mice, eszopiclone further suppressed REM sleep while DORA-22 facilitated recovery including increased REM sleep.ConclusionDORAs promote NREM and importantly REM sleep that is similar in proportion and magnitude to that seen during the normal resting phase across mammalian animal models. While limited overlap exists between therapeutic mechanisms, orexin signaling does not appear involved in the REM suppression exhibited by GABAA receptor modulators. The ability of DORAs to promote proportional NREM and REM sleep following sleep deprivation suggests that this mechanism may be effective in alleviating recovery from sleep disturbance.

Collaboration


Dive into the Pamela L. Tannenbaum's collaboration.

Top Co-Authors

Avatar

Anthony L. Gotter

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Steven V. Fox

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Joanne Stevens

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Susan L. Garson

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Scott D. Kuduk

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Donghui Cui

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Tamara D. Cabalu

United States Military Academy

View shared research outputs
Researchain Logo
Decentralizing Knowledge