Network


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

Hotspot


Dive into the research topics where Sandra M. Sanabria-Bohórquez is active.

Publication


Featured researches published by Sandra M. Sanabria-Bohórquez.


Proceedings of the National Academy of Sciences of the United States of America | 2007

[18F]MK-9470, a positron emission tomography (PET) tracer for in vivo human PET brain imaging of the cannabinoid-1 receptor

H. Donald Burns; Koen Van Laere; Sandra M. Sanabria-Bohórquez; Terence G. Hamill; Guy Bormans; Wai-si Eng; Ray E Gibson; Christine Ryan; Brett Connolly; Shil Patel; Stephen Krause; Amy Vanko; Anne Van Hecken; Patrick Dupont; Inge De Lepeleire; Paul Rothenberg; S. Aubrey Stoch; Josee Cote; William K. Hagmann; James P. Jewell; Linus S. Lin; Ping Liu; Mark T. Goulet; Keith M. Gottesdiener; John A. Wagner; Jan de Hoon; Luc Mortelmans; Tung M. Fong; Richard Hargreaves

[18F]MK-9470 is a selective, high-affinity, inverse agonist (human IC50, 0.7 nM) for the cannabinoid CB1 receptor (CB1R) that has been developed for use in human brain imaging. Autoradiographic studies in rhesus monkey brain showed that [18F]MK-9470 binding is aligned with the reported distribution of CB1 receptors with high specific binding in the cerebral cortex, cerebellum, caudate/putamen, globus pallidus, substantia nigra, and hippocampus. Positron emission tomography (PET) imaging studies in rhesus monkeys showed high brain uptake and a distribution pattern generally consistent with that seen in the autoradiographic studies. Uptake was blocked by pretreatment with a potent CB1 inverse agonist, MK-0364. The ratio of total to nonspecific binding in putamen was 4–5:1, indicative of a strong specific signal that was confirmed to be reversible via displacement studies with MK-0364. Baseline PET imaging studies in human research subject demonstrated behavior of [18F]MK-9470 very similar to that seen in monkeys, with very good test–retest variability (7%). Proof of concept studies in healthy young male human subjects showed that MK-0364, given orally, produced a dose-related reduction in [18F]MK-9470 binding reflecting CB1R receptor occupancy by the drug. Thus, [18F]MK-9470 has the potential to be a valuable, noninvasive research tool for the in vivo study of CB1R biology and pharmacology in a variety of neuropsychiatric disorders in humans. In addition, it allows demonstration of target engagement and noninvasive dose-occupancy studies to aid in dose selection for clinical trials of CB1R inverse agonists.


Cell Metabolism | 2008

The acyclic CB1R inverse agonist taranabant mediates weight loss by increasing energy expenditure and decreasing caloric intake

Carol Addy; Hamish Wright; Koen Van Laere; Ira Gantz; Ngozi Erondu; Bret Musser; Kaifeng Lu; Jinyu Yuan; Sandra M. Sanabria-Bohórquez; Aubrey Stoch; Cathy Stevens; Tung M. Fong; Inge De Lepeleire; Caroline Cilissen; Josee Cote; Kim Rosko; Isaias Noel Gendrano; Allison Martin Nguyen; Barry Gumbiner; Paul Rothenberg; Jan de Hoon; Guy Bormans; Marleen Depré; Wai-si Eng; Eric Ravussin; Samuel Klein; John E. Blundell; Gary A. Herman; H. Donald Burns; Richard Hargreaves

Cannabinoid 1 receptor (CB1R) inverse agonists are emerging as a potential obesity therapy. However, the physiological mechanisms by which these agents modulate human energy balance are incompletely elucidated. Here, we describe a comprehensive clinical research study of taranabant, a structurally novel acyclic CB1R inverse agonist. Positron emission tomography imaging using the selective CB1R tracer [(18)F]MK-9470 confirmed central nervous system receptor occupancy levels ( approximately 10%-40%) associated with energy balance/weight-loss effects in animals. In a 12-week weight-loss study, taranabant induced statistically significant weight loss compared to placebo in obese subjects over the entire range of evaluated doses (0.5, 2, 4, and 6 mg once per day) (p < 0.001). Taranabant treatment was associated with dose-related increased incidence of clinical adverse events, including mild to moderate gastrointestinal and psychiatric effects. Mechanism-of-action studies suggest that engagement of the CB1R by taranabant leads to weight loss by reducing food intake and increasing energy expenditure and fat oxidation.


Journal of Pharmacology and Experimental Therapeutics | 2013

In Vivo Quantification of Calcitonin Gene-Related Peptide Receptor Occupancy by Telcagepant in Rhesus Monkey and Human Brain Using the Positron Emission Tomography Tracer [11C]MK-4232

Eric Hostetler; Aniket Joshi; Sandra M. Sanabria-Bohórquez; Hong Fan; Zhizhen Zeng; Mona Purcell; Liza Gantert; Kerry Riffel; Mangay Williams; Stacey O'Malley; Patricia Miller; Harold G. Selnick; Steven N. Gallicchio; Ian M. Bell; Christopher A. Salvatore; Stefanie A. Kane; Chi-Chung Li; Richard Hargreaves; Tjibbe de Groot; Guy Bormans; Anne Van Hecken; Inge Derdelinckx; Jan de Hoon; Tom Reynders; Ruben Declercq; Inge De Lepeleire; W Dexter Kennedy; Rebecca Blanchard; Eugene E. Marcantonio; Cyrille Sur

Calcitonin gene-related peptide (CGRP) is a potent neuropeptide whose agonist interaction with the CGRP receptor (CGRP-R) in the periphery promotes vasodilation, neurogenic inflammation and trigeminovascular sensory activation. This process is implicated in the cause of migraine headaches, and CGRP-R antagonists in clinical development have proven effective in treating migraine-related pain in humans. CGRP-R is expressed on blood vessel smooth muscle and sensory trigeminal neurons and fibers in the periphery as well as in the central nervous system. However, it is not clear what role the inhibition of central CGRP-R plays in migraine pain relief. To this end, the CGRP-R positron emission tomography (PET) tracer [11C]MK-4232 (2-[(8R)-8-(3,5-difluorophenyl)-6,8-[6-11C]dimethyl-10-oxo-6,9-diazaspiro[4.5]decan-9-yl]-N-[(2R)-2′-oxospiro[1,3-dihydroindene-2,3′-1H-pyrrolo[2,3-b]pyridine]-5-yl]acetamide) was discovered and developed for use in clinical PET studies. In rhesus monkeys and humans, [11C]MK-4232 displayed rapid brain uptake and a regional brain distribution consistent with the known distribution of CGRP-R. Monkey PET studies with [11C]MK-4232 after intravenous dosing with CGRP-R antagonists validated the ability of [11C]MK-4232 to detect changes in CGRP-R occupancy in proportion to drug plasma concentration. Application of [11C]MK-4232 in human PET studies revealed that telcagepant achieved only low receptor occupancy at an efficacious dose (140 mg PO). Therefore, it is unlikely that antagonism of central CGRP-R is required for migraine efficacy. However, it is not known whether high central CGRP-R antagonism may provide additional therapeutic benefit.


Journal of Psychopharmacology | 2011

MRK-409 (MK-0343), a GABAA receptor subtype-selective partial agonist, is a non-sedating anxiolytic in preclinical species but causes sedation in humans.

John R. Atack; Ka Wafford; Lj Street; Gr Dawson; S Tye; K. Van Laere; Guy Bormans; Sandra M. Sanabria-Bohórquez; I. De Lepeleire; Jn de Hoon; A. Van Hecken; Hd Burns; Rm McKernan; Mg Murphy; Richard Hargreaves

MRK-409 binds to α1-, α2-, α3- and α5-containing human recombinant GABAA receptors with comparable high affinity (0.21–0.40 nM). However, MRK-409 has greater agonist efficacy at the α3 compared with α1 subtypes (respective efficacies relative to the full agonist chlordiazepoxide of 0.45 and 0.18). This compound readily penetrates the brain in rats and occupies the benzodiazepine site of GABAA receptors, measured using an in vivo [3H]flumazenil binding assay, with an Occ50 of 2.2 mg/kg p.o. and a corresponding plasma EC50 of 115 ng/mL. Behaviourally, the α3-preferring agonist efficacy profile of MRK-409 produced anxiolytic-like activity in rodent and primate unconditioned and conditioned models of anxiety with minimum effective doses corresponding to occupancies, depending on the particular model, ranging from ∼35% to 65% yet there were minimal overt signs of sedation at occupancies greater than 90%. In humans, however, safety and tolerability studies showed that there was pronounced sedation at a dose of 2 mg, resulting in a maximal tolerated dose of 1 mg. This 2 mg dose corresponded to a Cmax plasma concentration of 28 ng/mL, which, based on the rodent plasma EC50 for occupancy of 115 ng/mL, suggested that sedation in humans occurs at low levels of occupancy. This was confirmed in human positron emission tomography studies, in which [11C]flumazenil uptake following a single dose of 1 mg MRK-409 was comparable to that of placebo, indicating that occupancy of GABAA receptor benzodiazepine binding sites by MRK-409 was below the limits of detection (i.e. <10%). Taken together, these data show that MRK-409 causes sedation in humans at a dose (2 mg) corresponding to levels of occupancy considerably less than those predicted from rodent models to be required for anxiolytic efficacy (∼35–65%). Thus, the preclinical non-sedating anxiolytic profile of MRK-409 did not translate into humans and further development of this compound was halted.


Journal of Psychopharmacology | 2011

Preclinical and clinical pharmacology of TPA023B, a GABAA receptor α2/α3 subtype-selective partial agonist.

John R. Atack; David James Hallett; S Tye; Keith A. Wafford; Christine Ryan; Sandra M. Sanabria-Bohórquez; Wai-si Eng; Raymond E. Gibson; Hd Burns; Gerard R. Dawson; Rw Carling; Lj Street; A Pike; I. De Lepeleire; K. Van Laere; Guy Bormans; Jn de Hoon; A. Van Hecken; Ruth M. McKernan; Mg Murphy; Richard Hargreaves

In the accompanying paper we describe how MRK-409 unexpectedly produced sedation in man at relatively low levels of GABAA receptor occupancy (∼10%). Since it was not clear whether this sedation was mediated via the α2/α3 or α1 GABAA subtype(s), we characterized the properties of TPA023B, a high-affinity imidazotriazine which, like MRK-409, has partial agonist efficacy at the α2 and α3 subtype but is an antagonist at the α1 subtype, at which MRK-409 has weak partial agonism. TPA023B gave dose- and time-dependent occupancy of rat brain GABAA receptors as measured using an in vivo [3H]flumazenil binding assay, with 50% occupancy corresponding to a respective dose and plasma drug concentration of 0.09 mg/kg and 19 ng/mL, the latter of which was similar to that observed in mice (25 ng/mL) and comparable to values obtained in baboon and man using [11C]flumazenil PET (10 and 5.8 ng/mL, respectively). TPA023B was anxiolytic in rodent and primate (squirrel monkey) models of anxiety (elevated plus maze, fear-potentiated startle, conditioned suppression of drinking, conditioned emotional response) yet had no significant effects in rodent or primate assays of ataxia and/or myorelaxation (rotarod, chain-pulling, lever pressing), up to doses (10 mg/kg) corresponding to occupancy of greater than 99%. In man, TPA023B was well tolerated at a dose (1.5 mg) that produced occupancy of >50%, suggesting that the sedation previously seen with MRK-409 is due to the partial agonist efficacy of that compound at the α1 subtype, and highlighting the importance of antagonist efficacy at this particular GABAA receptor population for avoiding sedation in man.


Synapse | 2011

Synthesis, characterization, and monkey PET studies of [18F]MK-1312, a PET tracer for quantification of mGluR1 receptor occupancy by MK-5435

Eric Hostetler; Wai-si Eng; Aniket D. Joshi; Sandra M. Sanabria-Bohórquez; Hiroshi Kawamoto; Satoru Ito; Stacey O'Malley; Stephen Krause; Christine Ryan; Shil Patel; Mangay Williams; Kerry Riffel; Gentaroh Suzuki; Satoshi Ozaki; Hisashi Ohta; Jacquelynn J. Cook; H. Donald Burns; Richard Hargreaves

Two moderately lipophilic, high affinity ligands for metabotropic glutamate receptor subtype 1 (mGluR1) were radiolabeled with a positron‐emitting radioisotope and evaluated in rhesus monkey as potential PET tracers. Both ligands were radiolabeled with fluorine‐18 via nucleophilic displacement of the corresponding 2‐chloropyridine precursor with [18F]potassium fluoride. [18F]MK‐1312 was found to have a suitable signal for quantification of mGluR1 receptors in nonhuman primates and was more thoroughly characterized. In vitro autoradiographic studies with [18F]MK‐1312 in rhesus monkey and human brain tissue slices revealed an uptake distribution consistent with the known distribution of mGluR1, with the highest uptake in the cerebellum, moderate uptake in the hippocampus, thalamus, and cortical regions, and lowest uptake in the caudate and putamen. In vitro saturation binding studies in rhesus monkey and human cerebellum homogenates confirmed that [18F]MK‐1312 binds to a single site with a Bmax/Kd ratio of 132 and 98, respectively. PET studies in rhesus monkey with [18F]MK‐1312 showed high brain uptake and a regional distribution consistent with in vitro autoradiography results. Blockade of [18F]MK‐1312 uptake with mGluR1 allosteric antagonist MK‐5435 dose‐dependently reduced tracer uptake in all regions of gray matter to a similarly low level of tracer uptake. This revealed a large specific signal useful for determination of mGluR1 receptor occupancy in rhesus monkey. Taken together, these results are promising for clinical PET studies with [18F]MK‐1312 to determine mGluR1 occupancy of MK‐5435. Synapse 2011.


Synapse | 2011

The synthesis and preclinical evaluation in rhesus monkey of [18F]MK-6577 and [11C]CMPyPB glycine transporter 1 positron emission tomography radiotracers

Terence G. Hamill; Wai-si Eng; Andrew Jennings; Richard Thomas Lewis; Steven R. Thomas; Suzanne Wood; Leslie J. Street; David D. Wisnoski; Scott E. Wolkenberg; Craig W. Lindsley; Sandra M. Sanabria-Bohórquez; Shil Patel; Kerry Riffel; Christine Ryan; Jacquelynn J. Cook; Cyrille Sur; H. Donald Burns; Richard Hargreaves

Two positron emission tomography radiotracers for the glycine transporter 1 (GlyT1) are reported here. Each radiotracer is a propylsulfonamide‐containing benzamide and was labeled with either carbon‐11 or fluorine‐18. [11C]CMPyPB was synthesized by the alkylation of a 3‐hydroxypyridine precursor using [11C]MeI, and [18F]MK‐6577 was synthesized by a nucleophilic aromatic substitution reaction using a 2‐chloropyridine precursor. Each tracer shows good uptake into rhesus monkey brain with the expected distribution of highest uptake in the pons, thalamus, and cerebellum and lower uptake in the striatum and gray matter of the frontal cortex. In vivo blockade and chase studies of [18F]MK‐6577 showed a large specific signal and reversible binding. In vitro autoradiographic studies with [18F]MK‐6577 showed a large specific signal in both rhesus monkey and human brain slices and a distribution consistent with the in vivo results and those reported in the literature. In vivo metabolism studies in rhesus monkeys demonstrated that only more‐polar metabolites are formed for each tracer. Of these two tracers, [18F]MK‐6577 was more extensively characterized and is a promising clinical positron emission tomography tracer for imaging GlyT1 and for measuring GlyT1 occupancy of therapeutic compounds. Synapse, 2011.


Molecular Imaging and Biology | 2003

Image-derived input function for [11C]flumazenil kinetic analysis in human brain

Sandra M. Sanabria-Bohórquez; Alex Maes; Patrick Dupont; Guy Bormans; Tjibbe de Groot; Alexandre Coimbra; Wai-si Eng; Tine Laethem; Inge De Lepeleire; Jay Gambale; Jose M. Vega; H. Donald Burns

PURPOSE We describe a method for analysis of [11C]flumazenil data using an input curve directly derived from the positron emission tomography (PET) images. PROCEDURE The shape of the tracer plasma curve was obtained from the product of the intact flumazenil fraction in plasma in six arterial samples and the internal carotid artery time-activity curve (TAC). The resulting curve was calibrated using the [11C]flumazenil concentration in three of the six samples. The curve peak was recovered by adding an exponential function to the scaled curve whose parameters were estimated from simultaneous fittings of several tissue TACs assuming that all regions share the same input. RESULTS Good agreement was found between the image-derived and the experimental plasma curves in six subjects. Distribution volumes were highly correlated with linear regression slope and intercept values between [0.94, 1.03] and [-0.10, 0.16], respectively. CONCLUSION The proposed method is suitable for benzodiazepine receptor quantification requiring only a few blood samples.


NeuroImage | 2011

Synthesis, characterization, and monkey positron emission tomography (PET) studies of [18F]Y1-973, a PET tracer for the neuropeptide Y Y1 receptor.

Eric Hostetler; Sandra M. Sanabria-Bohórquez; Hong Fan; Zhizhen Zeng; Liza Gantert; Mangay Williams; Patricia Miller; Stacey O'Malley; Minoru Kameda; Makoto Ando; Nagaaki Sato; Satoshi Ozaki; Shigeru Tokita; Hisashi Ohta; David L. Williams; Cyrille Sur; Jacquelynn J. Cook; H. Donald Burns; Richard Hargreaves

Neuropeptide Y receptor subtype 1 (NPY Y1) has been implicated in appetite regulation, and antagonists of NPY Y1 are being explored as potential therapeutics for obesity. An NPY Y1 PET tracer is useful for determining the level of target engagement by NPY Y1 antagonists in preclinical and clinical studies. Here we report the synthesis and evaluation of [(18)F]Y1-973, a novel PET tracer for NPY Y1. [(18)F]Y1-973 was radiolabeled by reaction of a primary chloride with [(18)F]KF/K2.2.2 followed by deprotection with HCl. [(18)F]Y1-973 was produced with high radiochemical purity (>98%) and high specific activity (>1000 Ci/mmol). PET studies in rhesus monkey brain showed that the distribution of [(18)F]Y1-973 was consistent with the known NPY Y1 distribution; uptake was highest in the striatum and cortical regions and lowest in the pons, cerebellum nuclei, and brain stem. Blockade of [(18)F]Y1-973 uptake with NPY Y1 antagonist Y1-718 revealed a specific signal that was dose-dependently reduced in all regions of grey matter to a similarly low level of tracer uptake, indicative of an NPY Y1 specific signal. In vitro autoradiographic studies with [(18)F]Y1-973 in rhesus monkey and human brain tissue slices revealed an uptake distribution consistent with the in vivo PET studies. Highest binding density was observed in the dentate gyrus, caudate-putamen, and cortical regions; moderate binding density in the hypothalamus and thalamus; and lowest binding density in the globus pallidus and cerebellum. In vitro saturation binding studies in rhesus monkey and human caudate-putamen homogenates confirmed a similarly high B(max)/K(d) ratio for [(18)F]Y1-973, suggesting the tracer may provide a specific signal in human brain of similar magnitude to that observed in rhesus monkey. [(18)F]Y1-973 is a suitable PET tracer for imaging NPY Y1 in rhesus monkey with potential for translation to human PET studies.


NeuroImage | 2013

Evaluation of [18F]MK-0911, a positron emission tomography (PET) tracer for opioid receptor-like 1 (ORL1), in rhesus monkey and human

Eric Hostetler; Sandra M. Sanabria-Bohórquez; Wai-si Eng; Aniket D. Joshi; Shailendra Patel; Raymond E. Gibson; Stacey O'Malley; Stephen Krause; Christine Ryan; Kerry Riffel; Sheng Bi; Osamu Okamoto; Hiroshi Kawamoto; Satoshi Ozaki; Hisashi Ohta; Tjibbe de Groot; Guy Bormans; Marleen Depré; Jan de Hoon; Inge De Lepeleire; Tom Reynders; Jacquelynn J. Cook; H. Donald Burns; Michael F. Egan; William Cho; Koen Van Laere; Richard Hargreaves

Antagonism of the central opioid receptor like-1 receptor (ORL1) has been implicated in cognition, and has been a focus of drug discovery efforts to ameliorate the cognitive deficits that remain during the stable treatment of schizophrenia with current antipsychotics. In order to facilitate dose selection for phase II clinical testing an ORL1-specific PET tracer was developed to determine drug plasma concentration versus occupancy relationships in order to ensure that the doses selected and the degree of target engagement were sufficient to ensure adequate proof of concept testing. MK-0911 is a selective, high affinity antagonist for the ORL1 receptor radiolabeled with high specific activity (18)F for positron emission tomography (PET) studies. Evaluation of [(18)F]MK-0911 in rhesus monkey PET studies showed a pattern of brain uptake which was consistent with the known distribution of ORL1. In vitro autoradiography with [(18)F]MK-0911 in rhesus monkey and human brain tissue slices showed a regional distribution that was consistent with in vivo imaging results in monkey. Pre-treatment of rhesus monkeys with high doses of structurally diverse ORL1 antagonists MK-0584, MK-0337, or MK-5757 achieved blockade of [(18)F]MK-0911 in all gray matter regions. Baseline PET studies with [(18)F]MK-0911 in healthy human subjects showed tracer distribution and kinetics similar to that observed in rhesus monkey. Quantification of [(18)F]MK-0911 uptake in repeat human baseline PET studies showed a test-retest variability in volume of distribution (V(T)) averaging 3% across brain regions. Humans dosed orally with MK-5757 showed reduced [(18)F]MK-0911 tracer concentration in brain proportional with MK-5757 dose and plasma level. [(18)F]MK-0911 was useful for determining MK-5757-induced receptor occupancy of ORL1 to guide MK-5757 dose-selection for clinical proof-of-concept studies. Additionally, [(18)F]MK-0911 may be a useful tool for studying the pharmacology of ORL1 in various human populations and disease states.

Collaboration


Dive into the Sandra M. Sanabria-Bohórquez's collaboration.

Top Co-Authors

Avatar

Richard Hargreaves

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Guy Bormans

Catholic University of Leuven

View shared research outputs
Top Co-Authors

Avatar

Koen Van Laere

The Catholic University of America

View shared research outputs
Top Co-Authors

Avatar

Terence G. Hamill

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Jacquelynn J. Cook

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Wai-si Eng

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Jan de Hoon

Catholic University of Leuven

View shared research outputs
Researchain Logo
Decentralizing Knowledge