Network


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

Hotspot


Dive into the research topics where Anabella Villalobos is active.

Publication


Featured researches published by Anabella Villalobos.


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.


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).


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.


ACS Chemical Neuroscience | 2016

Central Nervous System Multiparameter Optimization Desirability: Application in Drug Discovery

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

Significant progress has been made in prospectively designing molecules using the central nervous system multiparameter optimization (CNS MPO) desirability tool, as evidenced by the analysis reported herein of a second wave of drug candidates that originated after the development and implementation of this tool. This simple-to-use design algorithm has expanded design space for CNS candidates and has further demonstrated the advantages of utilizing a flexible, multiparameter approach in drug discovery rather than individual parameters and hard cutoffs of physicochemical properties. The CNS MPO tool has helped to increase the percentage of compounds nominated for clinical development that exhibit alignment of ADME attributes, cross the blood-brain barrier, and reside in lower-risk safety space (low ClogP and high TPSA). The use of this tool has played a role in reducing the number of compounds submitted to exploratory toxicity studies and increasing the survival of our drug candidates through regulatory toxicology into First in Human studies. Overall, the CNS MPO algorithm has helped to improve the prioritization of design ideas and the quality of the compounds nominated for clinical development.


Nuclear Medicine and Biology | 2012

The synthesis and in vivo evaluation of [18F]PF-9811: a novel PET ligand for imaging brain fatty acid amide hydrolase (FAAH).

Marc B. Skaddan; Lei Zhang; Douglas S. Johnson; Aijun Zhu; Kenneth Zasadny; Richard V. Coelho; Kyle Kuszpit; Gwen Currier; Kuo-Hsien Fan; Elizabeth Mary Beck; Laigao Chen; Susan E. Drozda; Gayatri Balan; Micah J. Niphakis; Benjamin F. Cravatt; Kay Ahn; Thomas Bocan; Anabella Villalobos

INTRODUCTION Fatty acid amide hydrolase (FAAH) is responsible for the enzymatic degradation of the fatty acid amide family of signaling lipids, including the endogenous cannabinoid (endocannabinoid) anandamide. The involvement of the endocannabinoid system in pain and other nervous system disorders has made FAAH an attractive target for drug development. Companion molecular imaging probes are needed, however, to assess FAAH inhibition in the nervous system in vivo. We report here the synthesis and in vivo evaluation of [(18)F]PF-9811, a novel PET ligand for non-invasive imaging of FAAH in the brain. METHODS The potency and selectivity of unlabeled PF-9811 were determined by activity-based protein profiling (ABPP) both in vitro and in vivo. [(18)F]PF-9811 was synthesized in a 3-step, one-pot reaction sequence, followed by HPLC purification. Biological evaluation was performed by biodistribution and dynamic PET imaging studies in male rats. The specificity of [(18)F]PF-9811 uptake was evaluated by pre-administration of PF-04457845, a potent and selective FAAH inhibitor, 1h prior to radiotracer injection. RESULTS Biodistribution studies show good uptake (SUV~0.8 at 90 min) of [(18)F]PF-9811 in rat brain, with significant reduction of the radiotracer in all brain regions (37%-73% at 90 min) in blocking experiments. Dynamic PET imaging experiments in rat confirmed the heterogeneous uptake of [(18)F]PF-9811 in brain regions with high FAAH enzymatic activity, as well as statistically significant reductions in signal following pre-administration of the blocking compound PF-04457845. CONCLUSIONS [(18)F]PF-9811 is a promising PET imaging agent for FAAH. Biodistribution and PET imaging experiments show that the tracer has good uptake in brain, regional heterogeneity, and specific binding as determined by blocking experiments with the highly potent and selective FAAH inhibitor, PF-04457845.


Annual Reports in Medicinal Chemistry | 2012

Recent Advances in the Development of PET and SPECT Tracers for Brain Imaging

Lei Zhang; Anabella Villalobos

Abstract Positron emission tomography (PET) and single photon emission computed tomography (SPECT) are radiotracer-based noninvasive imaging techniques that provide quantitative binding information on specific target areas of interest. PET and SPECT have been proven to be particularly valuable for imaging targets in the central nervous system, enabling receptor occupancy measurements and dose selection for clinical candidates. In this review, we compare the advantages and limitations of these two imaging modalities; highlight recent advances in the PET field including new tools and design principles to facilitate tracer development and novel PDE10a, ORL1, and FAAH PET tracers; and review recent data in the field of SPECT in brain imaging.


EJNMMI Radiopharmacy and Chemistry | 2017

Strategies to facilitate the discovery of novel CNS PET ligands

Lei Zhang; Anabella Villalobos

Positron Emission Tomography (PET), as a non-invasive translatable imaging technology, can be incorporated into various stages of the CNS drug discovery process to provide valuable information for key preclinical and clinical decision-making. Novel CNS PET ligand discovery efforts in the industry setting, however, are facing unique challenges associated with lead design and prioritization, and budget constraints. In this review, three strategies aiming toward improving the central nervous system (CNS) PET ligand discovery process are described: first, early determination of receptor density (Bmax) and bio-distribution to inform PET viability and resource allocation; second, rational design and design prioritization guided by CNS PET design parameters; finally, a cost-effective in vivo specific binding assessment using a liquid chromatography-mass spectrometry (LC-MS/MS) “cold tracer” method. Implementation of these strategies allowed a more focused and rational CNS PET ligand discovery effort to identify high quality PET ligands for neuroimaging.


Scientific Reports | 2018

Dopamine D3 receptor antagonist reveals a cryptic pocket in aminergic GPCRs

Noelia Ferruz; Stefan Doerr; Michelle Vanase-Frawley; Yaozhong Zou; Xiaomin Chen; Eric S. Marr; Robin T. Nelson; Bethany L. Kormos; Travis T. Wager; Xinjun Hou; Anabella Villalobos; Simone Sciabola; Gianni De Fabritiis

The recent increase in the number of X-ray crystal structures of G-protein coupled receptors (GPCRs) has been enabling for structure-based drug design (SBDD) efforts. These structures have revealed that GPCRs are highly dynamic macromolecules whose function is dependent on their intrinsic flexibility. Unfortunately, the use of static structures to understand ligand binding can potentially be misleading, especially in systems with an inherently high degree of conformational flexibility. Here, we show that docking a set of dopamine D3 receptor compounds into the existing eticlopride-bound dopamine D3 receptor (D3R) X-ray crystal structure resulted in poses that were not consistent with results obtained from site-directed mutagenesis experiments. We overcame the limitations of static docking by using large-scale high-throughput molecular dynamics (MD) simulations and Markov state models (MSMs) to determine an alternative pose consistent with the mutation data. The new pose maintains critical interactions observed in the D3R/eticlopride X-ray crystal structure and suggests that a cryptic pocket forms due to the shift of a highly conserved residue, F6.52. Our study highlights the importance of GPCR dynamics to understand ligand binding and provides new opportunities for drug discovery.


Journal of Medicinal Chemistry | 2018

Design and Synthesis of Clinical Candidate PF-06751979: A Potent, Brain Penetrant, β-Site Amyloid Precursor Protein Cleaving Enzyme 1 (BACE1) Inhibitor Lacking Hypopigmentation

Brian T. O’Neill; Elizabeth Mary Beck; Christopher Ryan Butler; Charles E. Nolan; Cathleen Gonzales; Lei Zhang; Shawn D. Doran; Kimberly Lapham; Leanne M. Buzon; Jason K. Dutra; Gabriela Barreiro; Xinjun Hou; Luis Martinez-Alsina; Bruce N. Rogers; Anabella Villalobos; John C. Murray; Kevin Ogilvie; Erik LaChapelle; Cheng Chang; Lorraine Lanyon; Claire M. Steppan; Ashley Robshaw; Katherine Hales; Germaine Boucher; Karamjeet Pandher; Christopher Houle; Claude Ambroise; David Karanian; David Riddell; Kelly R. Bales

A major challenge in the development of β-site amyloid precursor protein cleaving enzyme 1 (BACE1) inhibitors for the treatment of Alzheimers disease is the alignment of potency, drug-like properties, and selectivity over related aspartyl proteases such as Cathepsin D (CatD) and BACE2. The potential liabilities of inhibiting BACE2 chronically have only recently begun to emerge as BACE2 impacts the processing of the premelanosome protein (PMEL17) and disrupts melanosome morphology resulting in a depigmentation phenotype. Herein, we describe the identification of clinical candidate PF-06751979 (64), which displays excellent brain penetration, potent in vivo efficacy, and broad selectivity over related aspartyl proteases including BACE2. Chronic dosing of 64 for up to 9 months in dog did not reveal any observation of hair coat color (pigmentation) changes and suggests a key differentiator over current BACE1 inhibitors that are nonselective against BACE2 in later stage clinical development.

Collaboration


Dive into the Anabella Villalobos's collaboration.

Researchain Logo
Decentralizing Knowledge