Network


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

Hotspot


Dive into the research topics where Patrick Robert Verhoest is active.

Publication


Featured researches published by Patrick Robert Verhoest.


ACS Chemical Neuroscience | 2010

Moving beyond Rules: The Development of a Central Nervous System Multiparameter Optimization (CNS MPO) Approach To Enable Alignment of Druglike Properties

Travis T. Wager; Xinjun Hou; Patrick Robert Verhoest; Anabella Villalobos

The interplay among commonly used physicochemical properties in drug design was examined and utilized to create a prospective design tool focused on the alignment of key druglike attributes. Using a set of six physicochemical parameters ((a) lipophilicity, calculated partition coefficient (ClogP); (b) calculated distribution coefficient at pH = 7.4 (ClogD); (c) molecular weight (MW); (d) topological polar surface area (TPSA); (e) number of hydrogen bond donors (HBD); (f) most basic center (pK(a))), a druglikeness central nervous system multiparameter optimization (CNS MPO) algorithm was built and applied to a set of marketed CNS drugs (N = 119) and Pfizer CNS candidates (N = 108), as well as to a large diversity set of Pfizer proprietary compounds (N = 11 303). The novel CNS MPO algorithm showed that 74% of marketed CNS drugs displayed a high CNS MPO score (MPO desirability score ≥ 4, using a scale of 0-6), in comparison to 60% of the Pfizer CNS candidates. This analysis suggests that this algorithm could potentially be used to identify compounds with a higher probability of successfully testing hypotheses in the clinic. In addition, a relationship between an increasing CNS MPO score and alignment of key in vitro attributes of drug discovery (favorable permeability, P-glycoprotein (P-gp) efflux, metabolic stability, and safety) was seen in the marketed CNS drug set, the Pfizer candidate set, and the Pfizer proprietary diversity set. The CNS MPO scoring function offers advantages over hard cutoffs or utilization of single parameters to optimize structure-activity relationships (SAR) by expanding medicinal chemistry design space through a holistic assessment approach. Based on six physicochemical properties commonly used by medicinal chemists, the CNS MPO function may be used prospectively at the design stage to accelerate the identification of compounds with increased probability of success.


Journal of Pharmacology and Experimental Therapeutics | 2008

Preclinical Characterization of Selective Phosphodiesterase 10A Inhibitors: A New Therapeutic Approach to the Treatment of Schizophrenia

Christopher J. Schmidt; Douglas S. Chapin; J. Cianfrogna; M. L. Corman; Mihály Hajós; John F. Harms; W. E. Hoffman; L. A. Lebel; S. A. McCarthy; Frederick R. Nelson; C. Proulx-LaFrance; Mark J. Majchrzak; A. D. Ramirez; K. Schmidt; Patricia A. Seymour; J. A. Siuciak; F. D. Tingley; R. D. Williams; Patrick Robert Verhoest; Frank S. Menniti

We have recently proposed the hypothesis that inhibition of the cyclic nucleotide phosphodiesterase (PDE) 10A may represent a new pharmacological approach to the treatment of schizophrenia (Curr Opin Invest Drug 8:54–59, 2007). PDE10A is highly expressed in the medium spiny neurons of the mammalian striatum (Brain Res 985:113–126, 2003; J Histochem Cytochem 54:1205–1213, 2006; Neuroscience 139:597–607, 2006), where the enzyme is hypothesized to regulate both cAMP and cGMP signaling cascades to impact early signal processing in the corticostriatothalamic circuit (Neuropharmacology 51:374–385, 2006; Neuropharmacology 51:386–396, 2006). Our current understanding of the physiological role of PDE10A and the therapeutic utility of PDE10A inhibitors derives in part from studies with papaverine, the only pharmacological tool for this target extensively profiled to date. However, this agent has significant limitations in this regard, namely, relatively poor potency and selectivity and a very short exposure half-life after systemic administration. In the present report, we describe the discovery of a new class of PDE10A inhibitors exemplified by TP-10 (2-{4-[-pyridin-4-yl-1-(2,2,2-trifluoro-ethyl)-1H-pyrazol-3-yl]-phenoxymethyl}-quinoline succinic acid), an agent with greatly improved potency, selectivity, and pharmaceutical properties. These new pharmacological tools enabled studies that provide further evidence that inhibition of PDE10A represents an important new target for the treatment of schizophrenia and related disorders of basal ganglia function.


ACS Chemical Neuroscience | 2010

Defining Desirable Central Nervous System Drug Space through the Alignment of Molecular Properties, in Vitro ADME, and Safety Attributes

Travis T. Wager; Ramalakshmi Y. Chandrasekaran; Xinjun Hou; Matthew D. Troutman; Patrick Robert Verhoest; Anabella Villalobos; Yvonne Will

As part of our effort to increase survival of drug candidates and to move our medicinal chemistry design to higher probability space for success in the Neuroscience therapeutic area, we embarked on a detailed study of the property space for a collection of central nervous system (CNS) molecules. We carried out a thorough analysis of properties for 119 marketed CNS drugs and a set of 108 Pfizer CNS candidates. In particular, we focused on understanding the relationships between physicochemical properties, in vitro ADME (absorption, distribution, metabolism, and elimination) attributes, primary pharmacology binding efficiencies, and in vitro safety data for these two sets of compounds. This scholarship provides guidance for the design of CNS molecules in a property space with increased probability of success and may lead to the identification of druglike candidates with favorable safety profiles that can successfully test hypotheses in the clinic.


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 | 2015

Discovery and preclinical profiling of 3-[4-(morpholin-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-5-yl]benzonitrile (PF-06447475), a highly potent, selective, brain penetrant, and in vivo active LRRK2 kinase inhibitor.

Jaclyn Louise Henderson; Bethany L. Kormos; Matthew Merrill Hayward; Karen J. Coffman; Jayasankar Jasti; Ravi G. Kurumbail; Travis T. Wager; Patrick Robert Verhoest; G. Stephen Noell; Yi Chen; Elie Needle; Zdenek Berger; Stefanus J. Steyn; Christopher Houle; Warren D. Hirst; Paul Galatsis

Leucine rich repeat kinase 2 (LRRK2) has been genetically linked to Parkinsons disease (PD) by genome-wide association studies (GWAS). The most common LRRK2 mutation, G2019S, which is relatively rare in the total population, gives rise to increased kinase activity. As such, LRRK2 kinase inhibitors are potentially useful in the treatment of PD. We herein disclose the discovery and optimization of a novel series of potent LRRK2 inhibitors, focusing on improving kinome selectivity using a surrogate crystallography approach. This resulted in the identification of 14 (PF-06447475), a highly potent, brain penetrant and selective LRRK2 inhibitor which has been further profiled in in vivo safety and pharmacodynamic studies.


Journal of Medicinal Chemistry | 2009

Identification of a Brain Penetrant PDE9A Inhibitor Utilizing Prospective Design and Chemical Enablement as a Rapid Lead Optimization Strategy

Patrick Robert Verhoest; Caroline Proulx-Lafrance; Michael Corman; Lois K. Chenard; Christopher John Helal; Xinjun Hou; Robin J. Kleiman; Shenping Liu; Eric S. Marr; Frank S. Menniti; Christopher J. Schmidt; Michelle Vanase-Frawley; Anne W. Schmidt; Robert Williams; Frederick R. Nelson; Kari R. Fonseca; Spiros Liras

By use of chemical enablement and prospective design, a novel series of selective, brain penetrant PDE9A inhibitors have been identified that are capable of producing in vivo elevations of brain cGMP.


Journal of Pharmacology and Experimental Therapeutics | 2012

Phosphodiesterase 9A regulates central cGMP and modulates responses to cholinergic and monoaminergic perturbation in vivo.

Robin J. Kleiman; Douglas S. Chapin; Curt Christoffersen; Jody Freeman; Kari R. Fonseca; Kieran F. Geoghegan; Sarah Grimwood; Victor Guanowsky; Mihály Hajós; John F. Harms; Christopher John Helal; William E. Hoffmann; Geralyn P. Kocan; Mark J. Majchrzak; Dina McGinnis; Stafford McLean; Frank S. Menniti; Fredrick R. Nelson; Robin Roof; Anne W. Schmidt; Patricia A. Seymour; Diane Stephenson; Francis David Tingley; Michelle Vanase-Frawley; Patrick Robert Verhoest; Christopher J. Schmidt

Cyclic nucleotides are critical regulators of synaptic plasticity and participate in requisite signaling cascades implicated across multiple neurotransmitter systems. Phosphodiesterase 9A (PDE9A) is a high-affinity, cGMP-specific enzyme widely expressed in the rodent central nervous system. In the current study, we observed neuronal staining with antibodies raised against PDE9A protein in human cortex, cerebellum, and subiculum. We have also developed several potent, selective, and brain-penetrant PDE9A inhibitors and used them to probe the function of PDE9A in vivo. Administration of these compounds to animals led to dose-dependent accumulation of cGMP in brain tissue and cerebrospinal fluid, producing a range of biological effects that implied functional significance for PDE9A-regulated cGMP in dopaminergic, cholinergic, and serotonergic neurotransmission and were consistent with the widespread distribution of PDE9A. In vivo effects of PDE9A inhibition included reversal of the respective disruptions of working memory by ketamine, episodic and spatial memory by scopolamine, and auditory gating by amphetamine, as well as potentiation of risperidone-induced improvements in sensorimotor gating and reversal of the stereotypic scratching response to the hallucinogenic 5-hydroxytryptamine 2A agonist mescaline. The results suggested a role for PDE9A in the regulation of monoaminergic circuitry associated with sensory processing and memory. Thus, PDE9A activity regulates neuronal cGMP signaling downstream of multiple neurotransmitter systems, and inhibition of PDE9A may provide therapeutic benefits in psychiatric and neurodegenerative diseases promoted by the dysfunction of these diverse neurotransmitter systems.


ACS Medicinal Chemistry Letters | 2012

Discovery of Brain-Penetrant, Irreversible Kynurenine Aminotransferase II Inhibitors for Schizophrenia.

Amy B. Dounay; Marie Anderson; Bruce M. Bechle; Brian M. Campbell; Michelle Marie Claffey; Artem G. Evdokimov; Edelweiss Evrard; Kari R. Fonseca; Xinmin Gan; Somraj Ghosh; Matthew Merrill Hayward; Weldon Horner; Ji-Young Kim; Laura A. McAllister; Jayvardhan Pandit; Vanessa Paradis; Vinod D. Parikh; Matthew R. Reese; Suobao Rong; Michelle A. Salafia; Katherine Schuyten; Christine A. Strick; Jamison B. Tuttle; James Valentine; Hong Wang; Laura E. Zawadzke; Patrick Robert Verhoest

Kynurenine aminotransferase (KAT) II has been identified as a potential new target for the treatment of cognitive impairment associated with schizophrenia and other psychiatric disorders. Following a high-throughput screen, cyclic hydroxamic acid PF-04859989 was identified as a potent and selective inhibitor of human and rat KAT II. An X-ray crystal structure and (13)C NMR studies of PF-04859989 bound to KAT II have demonstrated that this compound forms a covalent adduct with the enzyme cofactor, pyridoxal phosphate (PLP), in the active site. In vivo pharmacokinetic and efficacy studies in rat show that PF-04859989 is a brain-penetrant, irreversible inhibitor and is capable of reducing brain kynurenic acid by 50% at a dose of 10 mg/kg (sc). Preliminary structure-activity relationship investigations have been completed and have identified the positions on this scaffold best suited to modification for further optimization of this novel series of KAT II inhibitors.


Journal of Medicinal Chemistry | 2012

Design and Discovery of 6-[(3S,4S)-4-Methyl-1-(pyrimidin-2-ylmethyl)pyrrolidin-3-yl]-1-(tetrahydro-2H-pyran-4-yl)-1,5-dihydro-4H-pyrazolo[3,4-d]pyrimidin-4-one (PF-04447943), a Selective Brain Penetrant PDE9A Inhibitor for the Treatment of Cognitive Disorders

Patrick Robert Verhoest; Kari R. Fonseca; Xinjun Hou; Caroline Proulx-Lafrance; Michael Corman; Christopher John Helal; Michelle Marie Claffey; Jamison B. Tuttle; Karen J. Coffman; Shenpinq Liu; Frederick R. Nelson; Robin J. Kleiman; Frank S. Menniti; Christopher J. Schmidt; Michelle Vanase-Frawley; Spiros Liras

6-[(3S,4S)-4-Methyl-1-(pyrimidin-2-ylmethyl)pyrrolidin-3-yl]-1-(tetrahydro-2H-pyran-4-yl)-1,5-dihydro-4H-pyrazolo[3,4-d]pyrimidin-4-one (PF-04447943) is a novel PDE9A inhibitor identified using parallel synthetic chemistry and structure-based drug design (SBDD) and has advanced into clinical trials. Selectivity for PDE9A over other PDE family members was achieved by targeting key residue differences between the PDE9A and PDE1C catalytic site. The physicochemical properties of the series were optimized to provide excellent in vitro and in vivo pharmacokinetics properties in multiple species including humans. It has been reported to elevate central cGMP levels in the brain and CSF of rodents. In addition, it exhibits procognitive activity in several rodent models and synaptic stabilization in an amyloid precursor protein (APP) transgenic mouse model. Recent disclosures from clinical trials confirm that it is well tolerated in humans and elevates cGMP in cerebral spinal fluid of healthy volunteers, confirming that it is a quality pharmacological tool for testing clinical hypotheses in disease states associated with impairment of cGMP signaling or cognition.


Expert Opinion on Drug Discovery | 2011

Strategies to optimize the brain availability of central nervous system drug candidates

Travis T. Wager; Anabella Villalobos; Patrick Robert Verhoest; Xinjun Hou; Christopher L. Shaffer

Introduction: Access to the CNS is essential for most neurotherapeutics to elicit their effects. Leveraging design strategies incorporating physicochemical properties, in vitro and in vivo assays to predict and measure brain penetration, and brain delivery approaches may enable the drug discovery community to improve access of drug candidates into the CNS compartment. Areas covered: This article reviews aspects of the most recent molecular design, in vitro and in vivo strategies, and delivery technologies to optimize the unbound brain concentrations (C b,u) of CNS molecules. Through this, the article provides insight into recent ideas and concepts in CNS drug molecule design, methods for evaluating CNS drug exposures and alternative approaches to maximize drug access to neurocompartments. Expert opinion: The most pharmacologically relevant measure in assessing a compounds pharmacodynamic response in the CNS is its C b,u. The utilization of emerging design strategies, together with in vitro and in vivo assays, may enable the design of molecules with optimal C b,u:C p,u (C p,u, unbound plasma concentration) and appropriate C b,u, to elicit a biological response from the neurotherapeutic target. Where drug properties intrinsically render a compound CNS impaired, using novel CNS delivery approaches may result in sufficient C b,u to furnish a biological response.

Researchain Logo
Decentralizing Knowledge