Richard Apodaca
Johnson & Johnson Pharmaceutical Research and Development
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Richard Apodaca.
British Journal of Pharmacology | 2004
Ann J. Barbier; C Berridge; Christine Dugovic; A D Laposky; Sandy J. Wilson; Jamin D. Boggs; Leah Aluisio; Brian Lord; Curt Mazur; Cindy Pudiak; X Langlois; Wei Xiao; Richard Apodaca; Nicholas I. Carruthers; Timothy W. Lovenberg
1‐[4‐(3‐piperidin‐1‐yl‐propoxy)‐benzyl]‐piperidine (JNJ‐5207852) is a novel, non‐imidazole histamine H3 receptor antagonist, with high affinity at the rat (pKi=8.9) and human (pKi=9.24) H3 receptor. JNJ‐5207852 is selective for the H3 receptor, with negligible binding to other receptors, transporters and ion channels at 1 μM. JNJ‐5207852 readily penetrates the brain tissue after subcutaneous (s.c.) administration, as determined by ex vivo autoradiography (ED50 of 0.13 mg kg−1 in mice). In vitro autoradiography with 3H‐JNJ‐5207852 in mouse brain slices shows a binding pattern identical to that of 3H‐R‐α‐methylhistamine, with high specific binding in the cortex, striatum and hypothalamus. No specific binding of 3H‐JNJ‐5207852 was observed in brains of H3 receptor knockout mice. In mice and rats, JNJ‐5207852 (1–10 mg kg−1 s.c.) increases time spent awake and decreases REM sleep and slow‐wave sleep, but fails to have an effect on wakefulness or sleep in H3 receptor knockout mice. No rebound hypersomnolence, as measured by slow‐wave delta power, is observed. The wake‐promoting effects of this H3 receptor antagonist are not associated with hypermotility. A 4‐week daily treatment of mice with JNJ‐5207852 (10 mg kg−1 i.p.) did not lead to a change in body weight, possibly due to the compound being a neutral antagonist at the H3 receptor. JNJ‐5207852 is extensively absorbed after oral administration and reaches high brain levels. The data indicate that JNJ‐5207852 is a novel, potent and selective H3 antagonist with good in vitro and in vivo efficacy, and confirm the wake‐promoting effects of H3 receptor antagonists.
Bioorganic & Medicinal Chemistry Letters | 2008
John M. Keith; Richard Apodaca; Wei Xiao; Mark Seierstad; Kanaka Pattabiraman; Jiejun Wu; Michael Webb; Mark J. Karbarz; Sean Brown; Sandy J. Wilson; Brian Scott; Chui-Se Tham; Lin Luo; James Palmer; Sandra R. Chaplan; J. Guy Breitenbucher
A series of thiadiazolopiperazinyl aryl urea fatty acid amide hydrolase (FAAH) inhibitors is described. The molecules were found to inhibit the enzyme by acting as mechanism-based substrates, forming a covalent bond with Ser241. SAR and PK properties are presented.
Anesthesia & Analgesia | 2009
Mark J. Karbarz; Lin Luo; Leon Chang; Chui-Se Tham; James Palmer; Sandy J. Wilson; Michelle Wennerholm; Sean Brown; Brian Scott; Richard Apodaca; John M. Keith; Jiejun Wu; James Guy Breitenbucher; Sandra R. Chaplan; Michael Webb
Fatty acid amide hydrolase (FAAH) is an integral membrane enzyme within the amidase-signature family. It catalyzes the hydrolysis of several endogenous biologically active lipids, including anandamide (arachidonoyl ethanolamide), oleoyl ethanolamide, and palmitoyl ethanolamide. These endogenous FAAH substrates have been shown to be involved in a variety of physiological and pathological processes, including synaptic regulation, regulation of sleep and feeding, locomotor activity, pain and inflammation. Here we describe the biochemical and biological properties of a potent and selective FAAH inhibitor, 4-(3-phenyl-[1,2,4]thiadiazol-5-yl)-piperazine-1-carboxylic acid phenylamide (JNJ-1661010). The time-dependence of apparent IC50 values at rat and human recombinant FAAH, dialysis and mass spectrometry data indicate that the acyl piperazinyl fragment of JNJ-1661010 forms a covalent bond with the enzyme. This bond is slowly hydrolyzed, with release of the piperazinyl fragment and recovery of enzyme activity. The lack of inhibition observed in a rat liver esterase assay suggests that JNJ-1661010 is not a general esterase inhibitor. JNJ-1661010 is >100-fold preferentially selective for FAAH-1 when compared to FAAH-2. JNJ-1661010 dose-dependently increases arachidonoyl ethanolamide, oleoyl ethanolamide, and palmitoyl ethanolamide in the rat brain. The compound attenuates tactile allodynia in the rat mild thermal injury model of acute tissue damage and in the rat spinal nerve ligation (Chung) model of neuropathic pain. JNJ-1661010 also diminishes thermal hyperalgesia in the inflammatory rat carrageenan paw model. These data suggest that FAAH inhibitors with modes of action similar to JNJ-1661010 may be useful clinically as broad-spectrum analgesics.
Bioorganic & Medicinal Chemistry | 2008
Scott D. Bembenek; John M. Keith; Michael A. Letavic; Richard Apodaca; Ann J. Barbier; Lisa Dvorak; Leah Aluisio; Kirsten L. Miller; Timothy W. Lovenberg; Nicholas I. Carruthers
Currently, the only clinically effective treatment for Alzheimers disease (AD) is the use of acetylcholinesterase (AChE) inhibitors. These inhibitors have limited efficacy in that they only treat the symptoms and not the disease itself. Additionally, they often have unpleasant side effects. Here we consider the viability of a single molecule having the actions of both an AChE inhibitor and histamine H(3) receptor antagonist. Both histamine H(3) receptor antagonists and AChE inhibitors improve and augment cholinergic neurotransmission in the cortex. However, whereas an AChE inhibitor will impart its effect everywhere, a histamine H(3) antagonist will raise acetylcholine levels mostly in the brain as its mode of action will primarily be on the central nervous system. Therefore, the combination of both activities in a single molecule could be advantageous. Indeed, studies suggest an appropriate dual-acting compound may offer the desired therapeutic effect with fewer unpleasant side effects [CNS Drugs2004, 18, 827]. Further, recent studies(2) indicate the peripheral anionic site (PAS) of AChE interacts with the beta-amyloid (betaA) peptide. Consequently, a molecule capable of disrupting this interaction may have a significant impact on the production of or the aggregation of betaA. This may result in slowing down the progression of the disease rather than only treating the symptoms as current therapies do. Here, we detail how the use of the available crystal structure information, pharmacophore modeling and docking (automated, manual, classical, and QM/MM) lead to the identification of an AChE inhibitor-histamine H(3) receptor antagonist. Further, based on our models we speculate that this dual-acting compound may interact with the PAS. Such a dual-acting compound may be able to affect the pathology of AD in addition to providing symptomatic relief.
European Journal of Medicinal Chemistry | 2009
Devin M. Swanson; Chandra R. Shah; Brian Lord; Kirsten L. Morton; Lisa Dvorak; Curt Mazur; Richard Apodaca; Wei Xiao; Jamin D. Boggs; Mark A. Feinstein; Sandy J. Wilson; Ann J. Barbier; Pascal Bonaventure; Timothy W. Lovenberg; Nicholas I. Carruthers
A series of small molecules consisting of a heterocyclic core flanked by two basic functionalities were synthesized and screened for in vitro affinity at the human histamine H(3) receptor (hH(3)R). Nine of the twenty-eight compounds tested were found to possess a hH(3)R K(i) of less than 5 nM and consisted of a diverse range of central hetero-aromatic linkers (pyridine, pyrazine, oxazole, isoxazole, thiazole, furan, thiophene, and pyrrole). One member of this series, (4-isopropyl-piperazin-1-yl)-(6-piperidin-1-ylmethyl-pyridin-3-yl)-methanone (37), was found to be a high affinity, selective antagonist that crosses the blood-brain barrier and occupies H(3) receptors after oral administration in the rat.
European Journal of Medicinal Chemistry | 2009
Curt A. Dvorak; Richard Apodaca; Wei Xiao; Jill A. Jablonowski; Pascal Bonaventure; Christine Dugovic; Jonathan Shelton; Brian Lord; Kirsten L. Miller; Lisa Dvorak; Timothy W. Lovenberg; Nicholas I. Carruthers
A series of (4-aminobutyn-1-yl)benzylamines were prepared and the SAR around three key areas: (1) the amine attached to the butynyl linker (R(3)R(4)N-); (2) the benzylamine moiety (R(1)R(2)N-); and (3) the point of attachment of the benzylamine group (R(1)R(2)N- in the ortho, meta, or para positions) was examined. One compound, 4-[3-(4-piperidin-1-yl-but-1-ynyl)-benzyl]-morpholine (9s) was chosen for further profiling and found to be a selective histamine H(3) antagonist with desirable drug-like properties. Ex vivo receptor occupancy studies established that 9s does occupy H(3) binding sites in the brain of rats after oral administration. Subcutaneous doses of 9s (10mg/kg) given during the natural sleep phase demonstrated robust wake-promoting effects.
ACS Medicinal Chemistry Letters | 2015
Michael A. Letavic; Leah Aluisio; Richard Apodaca; Manoj Bajpai; Ann J. Barbier; Anne Bonneville; Pascal Bonaventure; Nicholas I. Carruthers; Christine Dugovic; Ian C. Fraser; Michelle Kramer; Brian Lord; Timothy W. Lovenberg; Lilian Y. Li; Kiev S. Ly; Heather M. McAllister; Neelakandha S. Mani; Kirsten L. Morton; Anthony Ndifor; S. Diane Nepomuceno; Chennagiri R. Pandit; Steven Sands; Chandra R. Shah; Jonathan Shelton; Sandra S. Snook; Devin M. Swanson; Wei Xiao
The preclinical characterization of novel phenyl(piperazin-1-yl)methanones that are histamine H3 receptor antagonists is described. The compounds described are high affinity histamine H3 antagonists. Optimization of the physical properties of these histamine H3 antagonists led to the discovery of several promising lead compounds, and extensive preclinical profiling aided in the identification of compounds with optimal duration of action for wake promoting activity. This led to the discovery of two development candidates for Phase I and Phase II clinical trials.
British Journal of Pharmacology | 2009
Ann J. Barbier; C Berridge; Christine Dugovic; A D Laposky; Sandy J. Wilson; Jamin D. Boggs; Leah Aluisio; Brian Lord; Curt Mazur; Cindy Pudiak; X Langlois; Wei Xiao; Richard Apodaca; Nicholas I. Carruthers; Timothy W. Lovenberg
1 1-[4-(3-piperidin-1-yl-propoxy)-benzyl]-piperidine (JNJ-5207852) is a novel, non-imidazole histamine H3 receptor antagonist, with high affinity at the rat (pKi=8.9) and human (pKi=9.24) H3 receptor. JNJ-5207852 is selective for the H3 receptor, with negligible binding to other receptors, transporters and ion channels at 1 microm. 2 JNJ-5207852 readily penetrates the brain tissue after subcutaneous (s.c.) administration, as determined by ex vivo autoradiography (ED50 of 0.13 mg kg(-1) in mice). In vitro autoradiography with 3H-JNJ-5207852 in mouse brain slices shows a binding pattern identical to that of 3H-R-alpha-methylhistamine, with high specific binding in the cortex, striatum and hypothalamus. No specific binding of 3H-JNJ-5207852 was observed in brains of H3 receptor knockout mice. 3 In mice and rats, JNJ-5207852 (1-10 mg kg(-1) s.c.) increases time spent awake and decreases REM sleep and slow-wave sleep, but fails to have an effect on wakefulness or sleep in H3 receptor knockout mice. No rebound hypersomnolence, as measured by slow-wave delta power, is observed. The wake-promoting effects of this H3 receptor antagonist are not associated with hypermotility. 4 A 4-week daily treatment of mice with JNJ-5207852 (10 mg kg(-1) i.p.) did not lead to a change in body weight, possibly due to the compound being a neutral antagonist at the H3 receptor. 5 JNJ-5207852 is extensively absorbed after oral administration and reaches high brain levels. 6 The data indicate that JNJ-5207852 is a novel, potent and selective H3 antagonist with good in vitro and in vivo efficacy, and confirm the wake-promoting effects of H3 receptor antagonists.
Journal of Medicinal Chemistry | 2003
Richard Apodaca; Curt A. Dvorak; Wei Xiao; Ann J. Barbier; Jamin D. Boggs; Sandy J. Wilson; Timothy W. Lovenberg; Nicholas I. Carruthers
Journal of Medicinal Chemistry | 2007
F. Anthony Romero; Wu Du; Inkyu Hwang; Thomas J. Rayl; F. Scott Kimball; Donmienne Leung; Heather Hoover; Richard Apodaca; J. Guy Breitenbucher; Benjamin F. Cravatt; Dale L. Boger