Network


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

Hotspot


Dive into the research topics where Adam Ogden is active.

Publication


Featured researches published by Adam Ogden.


Proceedings of the National Academy of Sciences of the United States of America | 2009

A class of selective antibacterials derived from a protein kinase inhibitor pharmacophore

J. Richard Miller; Steve Dunham; Igor Mochalkin; Craig Banotai; Matthew Bowman; Susan Buist; Bill Dunkle; Debra Hanna; H. James Harwood; Michael D. Huband; Alla Karnovsky; Michael Kuhn; Chris Limberakis; Jia Y. Liu; Shawn Mehrens; W. Thomas Mueller; Lakshmi Narasimhan; Adam Ogden; Jeff Ohren; J. V. N. Vara Prasad; John A. Shelly; Laura Skerlos; Mark C. Sulavik; V. Hayden Thomas; Steve VanderRoest; Li Ann Wang; Zhigang Wang; Amy Whitton; Tong Zhu; C. Kendall Stover

As the need for novel antibiotic classes to combat bacterial drug resistance increases, the paucity of leads resulting from target-based antibacterial screening of pharmaceutical compound libraries is of major concern. One explanation for this lack of success is that antibacterial screening efforts have not leveraged the eukaryotic bias resulting from more extensive chemistry efforts targeting eukaryotic gene families such as G protein-coupled receptors and protein kinases. Consistent with a focus on antibacterial target space resembling these eukaryotic targets, we used whole-cell screening to identify a series of antibacterial pyridopyrimidines derived from a protein kinase inhibitor pharmacophore. In bacteria, the pyridopyrimidines target the ATP-binding site of biotin carboxylase (BC), which catalyzes the first enzymatic step of fatty acid biosynthesis. These inhibitors are effective in vitro and in vivo against fastidious Gram-negative pathogens including Haemophilus influenzae. Although the BC active site has architectural similarity to those of eukaryotic protein kinases, inhibitor binding to the BC ATP-binding site is distinct from the protein kinase-binding mode, such that the inhibitors are selective for bacterial BC. In summary, we have discovered a promising class of potent antibacterials with a previously undescribed mechanism of action. In consideration of the eukaryotic bias of pharmaceutical libraries, our findings also suggest that pursuit of a novel inhibitor leads for antibacterial targets with active-site structural similarity to known human targets will likely be more fruitful than the traditional focus on unique bacterial target space, particularly when structure-based and computational methodologies are applied to ensure bacterial selectivity.


American Journal of Respiratory and Critical Care Medicine | 2009

Addition of PNU-100480 to First-Line Drugs Shortens the Time Needed to Cure Murine Tuberculosis

Kathy N. Williams; Steven Joseph Brickner; Charles K. Stover; Tong Zhu; Adam Ogden; Rokeya Tasneen; Sandeep Tyagi; Jacques Grosset; Eric L. Nuermberger

RATIONALE We recently reported strong bactericidal activity of the oxazolidinone PNU-100480 and its ability to increase the initial bactericidal effect of various combinations of first-line tuberculosis drugs and moxifloxacin in a murine model. OBJECTIVES To investigate whether the addition of PNU-100480 to the standard first-line regimen of rifampin, isoniazid, and pyrazinamide could shorten the duration of treatment necessary to prevent relapse after treatment discontinuation. METHODS Following aerosol infection with Mycobacterium tuberculosis H37Rv and a 13-day incubation period, control mice were treated with the first-line regimen while test mice received the same regimen with PNU-100480 or linezolid added for the first 2 or 4 months. Efficacy was assessed on the basis of quantitative cultures of lung homogenates performed monthly during treatment and 3 months after completion of 3, 4, 5, or 6 months of treatment to determine the relapse rate. MEASUREMENTS AND MAIN RESULTS After 2 months of treatment, mice receiving PNU-100480 in addition to the first-line regimen had lung CFU counts two orders of magnitude lower than control mice receiving the first-line regimen alone. Relapse rates after 4 months of treatment were 90, 35, and 5% when PNU-100480 was added to the first-line regimen for 0, 2, and 4 months, respectively. When the total treatment duration was 3 months, relapse rates were 85 and 35 to 45% when mice received PNU-100480 for 2 and 3 months, respectively; all control mice remained culture positive at the time of treatment completion with 17 to 72 CFU per lung. Addition of linezolid to the first-line regimen had an antagonistic effect resulting in higher CFU counts and failure to render mice culture-negative in 4 months of treatment. CONCLUSIONS Together with previous findings, these results confirm that PNU-100480, which is now in Phase I clinical testing, has sterilizing activity in the murine model and suggest that it may be capable of shortening treatment duration for drug-susceptible as well as drug-resistant tuberculosis in humans.


Bioorganic & Medicinal Chemistry Letters | 2013

Discovery of potent, selective, bioavailable phosphodiesterase 2 (PDE2) inhibitors active in an osteoarthritis pain model, Part I: Transformation of selective pyrazolodiazepinone phosphodiesterase 4 (PDE4) inhibitors into selective PDE2 inhibitors.

Mark Stephen Plummer; Joseph A. Cornicelli; Howard Roark; Donald James Skalitzky; Charles Stankovic; Susan Bove; Jayvardhan Pandit; Annise Paige Goodman; James Lester Hicks; Aurash Shahripour; David Beidler; Xiao Kang Lu; Brian Sanchez; Christopher Whitehead; Ron Sarver; Timothy Braden; Richard Gowan; Xi Qiang Shen; Katherine Welch; Adam Ogden; Nalini Sadagopan; Heidi Baum; Howard Miller; Craig Banotai; Cindy Spessard; Sandra Lightle

We identified potent, selective PDE2 inhibitors by optimizing residual PDE2 activity in a series of PDE4 inhibitors, while simultaneously minimizing PDE4 activity. These newly designed PDE2 inhibitors bind to the PDE2 enzyme in a cGMP-like mode in contrast to the cAMP-like binding mode found in PDE4. Structure activity relationship studies coupled with an inhibitor bound crystal structure in the active site of the catalytic domain of PDE2 identified structural features required to minimize PDE4 inhibition while simultaneously maximizing PDE2 inhibition.


Bioorganic & Medicinal Chemistry Letters | 2013

Discovery of potent selective bioavailable phosphodiesterase 2 (PDE2) inhibitors active in an osteoarthritis pain model. Part II: optimization studies and demonstration of in vivo efficacy.

Mark Stephen Plummer; Joseph A. Cornicelli; Howard Roark; Donald James Skalitzky; Charles Stankovic; Susan Bove; Jayvardhan Pandit; Annise Paige Goodman; James Lester Hicks; Aurash Shahripour; David Beidler; Xiao Kang Lu; Brian Sanchez; Christopher Whitehead; Ron Sarver; Timothy Braden; Richard Gowan; Xi Qiang Shen; Katherine Welch; Adam Ogden; Nalini Sadagopan; Heidi Baum; Howard Miller; Craig Banotai; Cindy Spessard; Sandra Lightle

Selective phosphodiesterase 2 (PDE2) inhibitors are shown to have efficacy in a rat model of osteoarthritis (OA) pain. We identified potent, selective PDE2 inhibitors by optimizing residual PDE2 activity in a series of phosphodiesterase 4 (PDE4) inhibitors, while minimizing PDE4 inhibitory activity. These newly designed PDE2 inhibitors bind to the PDE2 enzyme in a cGMP-like binding mode orthogonal to the cAMP-like binding mode found in PDE4. Extensive structure activity relationship studies ultimately led to identification of pyrazolodiazepinone, 22, which was >1000-fold selective for PDE2 over recombinant, full length PDEs 1B, 3A, 3B, 4A, 4B, 4C, 7A, 7B, 8A, 8B, 9, 10 and 11. Compound 22 also retained excellent PDE2 selectivity (241-fold to 419-fold) over the remaining recombinant, full length PDEs, 1A, 4D, 5, and 6. Compound 22 exhibited good pharmacokinetic properties and excellent oral bioavailability (F=78%, rat). In an in vivo rat model of OA pain, compound 22 had significant analgesic activity 1 and 3h after a single, 10 mg/kg, subcutaneous dose.


The Journal of Nuclear Medicine | 2016

First-in-Human Assessment of the Novel PDE2A PET Radiotracer 18F-PF-05270430

Mika Naganawa; Rikki Waterhouse; Nabeel Nabulsi; Shu-fei Lin; David Labaree; Jim Ropchan; Sanela Tarabar; Nicholas DeMartinis; Adam Ogden; Anindita Banerjee; Yiyun Huang; Richard E. Carson

This was a first-in-human study of the novel phosphodiesterase-2A (PDE2A) PET ligand 18F-PF-05270430. The primary goals were to determine the appropriate tracer kinetic model to quantify brain uptake and to examine the within-subject test–retest variability. Methods: In advance of human studies, radiation dosimetry was determined in nonhuman primates. Six healthy male subjects participated in a test–retest protocol with dynamic scans and metabolite-corrected input functions. Nine brain regions of interest were studied, including the striatum, white matter, neocortical regions, and cerebellum. Multiple modeling methods were applied to calculate volume of distribution (VT) and binding potentials relative to the nondisplaceable tracer in tissue (BPND), concentration of tracer in plasma (BPP), and free tracer in tissue (BPF). The cerebellum was selected as a reference region to calculate binding potentials. Results: The dosimetry study provided an effective dose of less than 0.30 mSv/MBq, with the gallbladder as the critical organ; the human target dose was 185 MBq. There were no adverse events or clinically detectable pharmacologic effects reported. Tracer uptake was highest in the striatum, followed by neocortical regions and white matter, and lowest in the cerebellum. Regional time–activity curves were well fit by multilinear analysis-1, and a 70-min scan duration was sufficient to quantify VT and the binding potentials. BPND, with mean values ranging from 0.3 to 0.8, showed the best intrasubject and intersubject variability and reliability. Test–retest variability in the whole brain (excluding the cerebellum) of VT, BPND, and BPP were 8%, 16%, and 17%, respectively. Conclusion: 18F-PF-05270430 shows promise as a PDE2A PET ligand, albeit with low binding potential values.


Neuropharmacology | 2017

In vivo measurement of PDE10A enzyme occupancy by positron emission tomography (PET) following single oral dose administration of PF-02545920 in healthy male subjects.

Marielle Delnomdedieu; Anton Forsberg; Adam Ogden; Patrik Fazio; Ching-Ray Yu; Per Stenkrona; Sridhar Duvvuri; William S. David; Nabil Al-Tawil; Ottavio V. Vitolo; Nahid Amini; Sangram Nag; Christer Halldin; Andrea Varrone

&NA; Phosphodiesterase 10A (PDE10A) is an enzyme highly enriched in the striatal medium spiny neurons. It is involved in the regulation of cytoplasmic levels of cAMP and cGMP and signaling within the basal ganglia. This study with PDE10A radioligand [18F]MNI‐659 was designed to measure the enzyme occupancy of PF‐02545920 in 8 healthy male volunteers (48 ± 4 years) after a single oral dose (10 mg or 20 mg) and to evaluate safety and tolerability. Arterial blood sampling was performed to obtain a metabolite‐corrected plasma input function for the quantification of [18F]MNI‐659 binding to PDE10A. The occupancy of PF‐02545920 was calculated with two different methods: In Method 1, [18F]MNI‐659 enzyme occupancy was calculated from the estimates of binding potential, using the cerebellum as a reference region; in Method 2, occupancy was estimated from the slope of the revised Lassens plot. Serum concentrations of PF‐02545920 were measured to determine the relationship between concentration and occupancy. Based on Method 1, striatal PDE10A occupancy increased with increasing PF‐02545920 dose: 14–27% at 10 mg dose (N = 4) and 45–63% at 20 mg dose (N = 3). Comparable occupancies were observed using Lassens plot Method 2: 10 mg: 14–37%; 20 mg: 46–55%. The relationship between exposure and occupancy was best described using an Emax model. The serum concentration associated with 50% occupancy was estimated to be 93.2 ng/mL. Single oral doses of 10 mg or 20 mg of PF‐02545920 were safe and well tolerated in healthy male volunteers [NCT# 01918202]. HighlightsSingle oral doses (10 mg, 20 mg) of PF‐02545920 were safe and well tolerated in healthy male volunteers.Enzyme occupancy of PF‐02545920 obtained using [18F]MNI‐659 PET was in the expected range.Striatal PDE10A occupancy increased with increasing PF‐02545920 dose and exposure, reaching ˜50% following the 20 mg single dose.A 20 mg oral dose of PF‐02545920 provides sufficient target brain occupancy for evaluation in future PDE10 clinical trials.


Antimicrobial Agents and Chemotherapy | 2012

Evaluation of pharmacokinetic/pharmacodynamic relationships of PD-0162819, a biotin carboxylase inhibitor representing a new class of antibacterial compounds, using in vitro infection models.

Adam Ogden; Michael Kuhn; Michael Dority; Susan Buist; Shawn Mehrens; Tong Zhu; Deqing Xiao; J. Richard Miller; Debra Hanna

ABSTRACT The present study investigated the pharmacokinetic/pharmacodynamic (PK/PD) relationships of a prototype biotin carboxylase (BC) inhibitor, PD-0162819, against Haemophilus influenzae 3113 in static concentration time-kill (SCTK) and one-compartment chemostat in vitro infection models. H. influenzae 3113 was exposed to PD-0162819 concentrations of 0.5 to 16× the MIC (MIC = 0.125 μg/ml) and area-under-the-curve (AUC)/MIC ratios of 1 to 1,100 in SCTK and chemostat experiments, respectively. Serial samples were collected over 24 h. For efficacy driver analysis, a sigmoid maximum-effect (Emax) model was fitted to the relationship between bacterial density changes over 24 h and corresponding PK/PD indices. A semimechanistic PK/PD model describing the time course of bacterial growth and death was developed. The AUC/MIC ratio best explained efficacy (r2 = 0.95) compared to the peak drug concentration (Cmax)/MIC ratio (r2 = 0.76) and time above the MIC (T>MIC) (r2 = 0.88). Static effects and 99.9% killing were achieved at AUC/MIC values of 500 and 600, respectively. For time course analysis, the net bacterial growth rate constant, maximum bacterial density, and maximum kill rate constant were similar in SCTK and chemostat studies, but PD-0162819 was more potent in SCTK than in the chemostat (50% effective concentration [EC50] = 0.046 versus 0.34 μg/ml). In conclusion, basic PK/PD relationships for PD-0162819 were established using in vitro dynamic systems. Although the bacterial growth parameters and maximum drug effects were similar in SCTK and the chemostat system, PD-0162819 appeared to be more potent in SCTK, illustrating the importance of understanding the differences in preclinical models. Additional studies are needed to determine the in vivo relevance of these results.


Journal of Neurology, Neurosurgery, and Psychiatry | 2018

J06 A randomized, double-blind, placebo-controlled phase ii efficacy and safety study of the PDE10A inhibitor PF-02545920 in huntington disease (amaryllis)

Marielle Delnomdedieu; Ye Tan; Adam Ogden; Zdenek Berger; Ralf Reilmann

Background Degeneration of striatal medium-spiny neurons underlies corticostriatal dysfunction in Huntington disease (HD). Inhibition of Phosphodiesterase-10A, an intracellular signaling regulator preferentially expressed in medium-spiny neurons, increases corticostriatal function in preclinical models providing a promising therapeutic target for HD. Aims To assess efficacy on motor function, safety and tolerability of the phosphodiesterase-10A inhibitor PF-02545920 in HD. Methods AMARYLLIS was a double-blind, placebo-controlled, randomized trial (ClinicalTrials.gov, NCT02197130). The primary outcome was the Unified-Huntington’s-Disease-Rating-Scale Total-Motor-Score (UHDRS-TMS). Patients with genetically confirmed symptomatic HD in stages I-II, UHDRS-TMS≥10 and no neuroleptic treatment, were randomized to 5 mg, 20 mg PF-02545920 or placebo twice daily for 26 weeks. Results 216 (79%) of 272 randomized subjects completed AMARYLLIS (5 mg=79, 20 mg=56, placebo=81). UHDRS-TMS and the secondary efficacy assessments UHDRS-Total-Maximum-Chorea score and Clinical-Global-Impression of Improvement showed no benefits compared to placebo. Pre-specified exploratory quantitative motor (Q-Motor) measures showed consistent and dose-dependent improvements. Adverse events were mild or moderate and occurred more frequently at the 20 mg (90%) compared to 5 mg (86%) dose and placebo (72%), most common were somnolence, fatigue, and weight-decrease. Adverse-event-related discontinuations were higher at 20 mg (26%) compared to 5 mg (14%) and placebo (6%). Serious adverse event were not dose-related and <10% across groups. Conclusions PF-02545920 was generally safe and sufficiently tolerated. This study did not provide evidence of efficacy in primary or secondary clinical endpoints. But rater-independent improvements in pre-specified Q-Motor measures suggested proof-of-concept for a dose-dependent central effect on motor coordination of unknown clinical significance. Exploration of higher doses possibly in earlier stages of HD may be considered.


Journal of Medicinal Chemistry | 2018

Identification of a Potent, Highly Selective, and Brain Penetrant Phosphodiesterase 2A Inhibitor Clinical Candidate

Christopher John Helal; Eric P. Arnold; Tracey Boyden; Cheng Chang; Thomas A. Chappie; Ethan L. Fisher; Mihály Hajós; John F. Harms; William E. Hoffman; John M. Humphrey; Jayvardhan Pandit; Zhijun Kang; Robin J. Kleiman; Bethany L. Kormos; Che-Wah Lee; Jiemin Lu; Noha Maklad; Laura McDowell; Dina McGinnis; Rebecca E. O’Connor; Christopher J. O’Donnell; Adam Ogden; Mary Piotrowski; Christopher J. Schmidt; Patricia A. Seymour; Hirokazu Ueno; Nichole Vansell; Patrick Robert Verhoest; Edward X. Yang

Computational modeling was used to direct the synthesis of analogs of previously reported phosphodiesterase 2A (PDE2A) inhibitor 1 with an imidazotriazine core to yield compounds of significantly enhanced potency. The analog PF-05180999 (30) was subsequently identified as a preclinical candidate targeting cognitive impairment associated with schizophrenia. Compound 30 demonstrated potent binding to PDE2A in brain tissue, dose responsive mouse brain cGMP increases, and reversal of N-methyl-d-aspartate (NMDA) antagonist-induced (MK-801, ketamine) effects in electrophysiology and working memory models in rats. Preclinical pharmacokinetics revealed unbound brain/unbound plasma levels approaching unity and good oral bioavailability resulting in an average concentration at steady state (Cav,ss) predicted human dose of 30 mg once daily (q.d.). Modeling of a modified release formulation suggested that 25 mg twice daily (b.i.d.) could maintain plasma levels of 30 at or above targeted efficacious plasma levels for 24 h, which became part of the human clinical plan.


Alzheimers & Dementia | 2011

Translational receptor occupancy for the 5-HT4 partial agonist PF-04995274 in rats, non-human primates and healthy volunteers

Sarah Grimwood; Elena M. Drummond; Ken Zasadny; Marc B. Skaddan; Michael Aaron Brodney; Karen Coffman; Timothy Nicholas; Sridhar Duvvuri; David Raunig; Claire Leurent; Carolyn Rowinski; Yo-sub Park; Adam Ogden; Aarti Sawant; Emily Miller; Tracey L. Rapp; Yi Wang; Beata Planeta-Wilson; Jim Ropchan; Richard E. Carson; Evan D. Morris; Yu-Shin Ding; Elyse Katz

Collaboration


Dive into the Adam Ogden's collaboration.

Researchain Logo
Decentralizing Knowledge