Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Pharmacol Exp Ther ; 362(2): 254-262, 2017 08.
Article in English | MEDLINE | ID: mdl-28533287

ABSTRACT

Prescription opioids are a mainstay in the treatment of acute moderate to severe pain. However, chronic use leads to a host of adverse consequences including tolerance and opioid-induced hyperalgesia (OIH), leading to more complex treatment regimens and diminished patient compliance. Patients with OIH paradoxically experience exaggerated nociceptive responses instead of pain reduction after chronic opioid usage. The development of OIH and tolerance tend to occur simultaneously and, thus, present a challenge when studying the molecular mechanisms driving each phenomenon. We tested the hypothesis that a G protein-biased µ-opioid peptide receptor (MOPR) agonist would not induce symptoms of OIH, such as mechanical allodynia, following chronic administration. We observed that the development of opioid-induced mechanical allodynia (OIMA), a model of OIH, was absent in ß-arrestin1-/- and ß-arrestin2-/- mice in response to chronic administration of conventional opioids such as morphine, oxycodone and fentanyl, whereas tolerance developed independent of OIMA. In agreement with the ß-arrestin knockout mouse studies, chronic administration of TRV0109101, a G protein-biased MOPR ligand and structural analog of oliceridine, did not promote the development of OIMA but did result in drug tolerance. Interestingly, following induction of OIMA by morphine or fentanyl, TRV0109101 was able to rapidly reverse allodynia. These observations establish a role for ß-arrestins in the development of OIH, independent of tolerance, and suggest that the use of G protein-biased MOPR ligands, such as oliceridine and TRV0109101, may be an effective therapeutic avenue for managing chronic pain with reduced propensity for opioid-induced hyperalgesia.


Subject(s)
Analgesics, Opioid/administration & dosage , GTP-Binding Proteins/agonists , Hyperalgesia/drug therapy , Pain Measurement/drug effects , Receptors, Opioid, mu/agonists , Animals , Dose-Response Relationship, Drug , Drug Administration Schedule , GTP-Binding Proteins/physiology , HEK293 Cells , Humans , Hyperalgesia/pathology , Male , Mice , Mice, Inbred C57BL , Pain Measurement/methods , Receptors, Opioid, mu/physiology
2.
Circulation ; 135(11): 1056-1070, 2017 Mar 14.
Article in English | MEDLINE | ID: mdl-28104714

ABSTRACT

BACKGROUND: Biased agonism of the angiotensin II receptor is known to promote cardiac contractility. Our laboratory indicated that these effects may be attributable to changes at the level of the myofilaments. However, these signaling mechanisms remain unknown. Because a common finding in dilated cardiomyopathy is a reduction in the myofilament-Ca2+ response, we hypothesized that ß-arrestin signaling would increase myofilament-Ca2+ responsiveness in a model of familial dilated cardiomyopathy and improve cardiac function and morphology. METHODS: We treated a dilated cardiomyopathy-linked mouse model expressing a mutant tropomyosin (Tm-E54K) for 3 months with either TRV120067, a ß-arrestin 2-biased ligand of the angiotensin II receptor, or losartan, an angiotensin II receptor blocker. At the end of the treatment protocol, we assessed cardiac function using echocardiography, the myofilament-Ca2+ response of detergent-extracted fiber bundles, and used proteomic approaches to understand changes in posttranslational modifications of proteins that may explain functional changes. We also assessed signaling pathways altered in vivo and by using isolated myocytes. RESULTS: TRV120067- treated Tm-E54K mice showed improved cardiac structure and function, whereas losartan-treated mice had no improvement. Myofilaments of TRV120067-treated Tm-E54K mice had significantly improved myofilament-Ca2+ responsiveness, which was depressed in untreated Tm-E54K mice. We attributed these changes to increased MLC2v and MYPT1/2 phosphorylation seen only in TRV120067-treated mice. We found that the functional changes were attributable to an activation of ERK1/2-RSK3 signaling, mediated through ß-arrestin, which may have a novel role in increasing MLC2v phosphorylation through a previously unrecognized interaction of ß-arrestin localized to the sarcomere. CONCLUSIONS: Long-term ß-arrestin 2-biased agonism of the angiotensin II receptor may be a viable approach to the treatment of dilated cardiomyopathy by not only preventing maladaptive signaling, but also improving cardiac function by altering the myofilament-Ca2+ response via ß-arrestin signaling pathways.


Subject(s)
Cardiomyopathy, Dilated/physiopathology , beta-Arrestins/agonists , Angiotensin Receptor Antagonists/pharmacology , Angiotensin Receptor Antagonists/therapeutic use , Animals , Calcium/metabolism , Cardiomyopathy, Dilated/drug therapy , Cardiomyopathy, Dilated/metabolism , Disease Models, Animal , Female , Heart/diagnostic imaging , Heart/physiopathology , Losartan/pharmacology , Losartan/therapeutic use , Male , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Myofibrils/drug effects , Myofibrils/metabolism , Myosin Light Chains/genetics , Myosin Light Chains/metabolism , Myosin-Light-Chain Phosphatase/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction/drug effects , Tropomyosin/genetics , Tropomyosin/metabolism , beta-Arrestins/metabolism
3.
Cardiovasc Res ; 107(2): 226-34, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26045475

ABSTRACT

AIMS: Therapeutic approaches to treat familial dilated cardiomyopathy (DCM), which is characterized by depressed sarcomeric tension and susceptibility to Ca(2+)-related arrhythmias, have been generally unsuccessful. Our objective in the present work was to determine the effect of the angiotensin II type 1 receptor (AT1R) biased ligand, TRV120023, on contractility of hearts of a transgenic mouse model of familial DCM with mutation in tropomyosin at position 54 (TG-E54K). Our rationale is based on previous studies, which have supported the hypothesis that biased G-protein-coupled receptor ligands, signalling via ß-arrestin, increase cardiac contractility with no effect on Ca(2+) transients. Our previous work demonstrated that the biased ligand TRV120023 is able to block angiotensin-induced hypertrophy, while promoting an increase in sarcomere Ca(2+) response. METHODS AND RESULTS: We tested the hypothesis that the depression in cardiac function associated with DCM can be offset by infusion of the AT1R biased ligand, TRV120023. We intravenously infused saline, TRV120023, or the unbiased ligand, losartan, for 15 min in TG-E54K and non-transgenic mice to obtain left ventricular pressure-volume relations. Hearts were analysed for sarcomeric protein phosphorylation. Results showed that the AT1R biased ligand increases cardiac performance in TG-E54K mice in association with increased myosin light chain-2 phosphorylation. CONCLUSION: Treatment of mice with an AT1R biased ligand, acting via ß-arrestin signalling, is able to induce an increase in cardiac contractility associated with an increase in ventricular myosin light chain-2 phosphorylation. AT1R biased ligands may prove to be a novel inotropic approach in familial DCM.


Subject(s)
Cardiac Myosins/metabolism , Cardiomyopathy, Dilated/metabolism , Myocardial Contraction/physiology , Myosin Light Chains/metabolism , Oligopeptides/metabolism , Animals , Arrestins/metabolism , Disease Models, Animal , Female , Heart/physiopathology , Ligands , Male , Mice, Transgenic , Phosphorylation , beta-Arrestins
4.
Curr Opin Pharmacol ; 16: 108-15, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24834870

ABSTRACT

G protein-coupled receptors (GPCRs), in recent years, have been shown to signal via multiple distinct pathways. Furthermore, biased ligands for some receptors can differentially stimulate or inhibit these pathways versus unbiased endogenous ligands or drugs. Biased ligands can be used to gain a deeper understanding of the molecular targets and cellular responses associated with a GPCR, and may be developed into therapeutics with improved efficacy, safety and/or tolerability. Here we review examples and approaches to pathway validation that establish the relevance and therapeutic potential of distinct pathways that can be selectively activated or blocked by biased ligands.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Animals , Drug Discovery , Humans , Ligands
5.
J Clin Pharmacol ; 54(3): 351-7, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24122908

ABSTRACT

TRV130 is a G protein-biased ligand at the µ-opioid receptor. In preclinical studies it was potently analgesic while causing less respiratory depression and gastrointestinal dysfunction than morphine, suggesting unique benefits in acute pain management. A first-in-human study was conducted with ascending doses of TRV130 to explore its tolerability, pharmacokinetics, and pharmacodynamics in healthy volunteers. TRV130 was well-tolerated over the dose range 0.15 to 7 mg administered intravenously over 1 hour. TRV130 geometric mean exposure and Cmax were dose-linear, with AUC0-inf of 2.52 to 205.97 ng h/mL and Cmax of 1.04 to 102.36 ng/mL across the dose range tested, with half-life of 1.6-2.7 hours. A 1.5 mg dose of TRV130 was also well-tolerated when administered as 30, 15, 5, and 1 minute infusions. TRV130 pharmacokinetics were modestly affected by CYP2D6 phenotype: clearance was reduced by 53% in CYP2D6 poor metabolizers.TRV130 caused dose- and exposure-related pupil constriction, confirming central compartment µ-opioid receptor engagement. Marked pupil constriction was noted at 2.2, 4, and 7 mg doses. Nausea and vomiting observed at the 7 mg dose limited further dose escalation. These findings suggest that TRV130 may have a broad margin between doses causing µ-opioid receptor-mediated pharmacology and doses causing µ-opioid receptor-mediated intolerance.


Subject(s)
Analgesics , Spiro Compounds , Thiophenes , Administration, Intravenous , Adult , Analgesics/administration & dosage , Analgesics/adverse effects , Analgesics/pharmacokinetics , Analgesics/pharmacology , Cross-Over Studies , Double-Blind Method , Healthy Volunteers , Humans , Ligands , Male , Middle Aged , Nausea/chemically induced , Receptors, Opioid, mu/metabolism , Spiro Compounds/administration & dosage , Spiro Compounds/adverse effects , Spiro Compounds/pharmacokinetics , Spiro Compounds/pharmacology , Thiophenes/administration & dosage , Thiophenes/adverse effects , Thiophenes/pharmacokinetics , Thiophenes/pharmacology , Vomiting/chemically induced , Young Adult
6.
J Med Chem ; 56(20): 8019-31, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24063433

ABSTRACT

The concept of "ligand bias" at G protein coupled receptors has been introduced to describe ligands which preferentially stimulate one intracellular signaling pathway over another. There is growing interest in developing biased G protein coupled receptor ligands to yield safer, better tolerated, and more efficacious drugs. The classical µ opioid morphine elicited increased efficacy and duration of analgesic response with reduced side effects in ß-arrestin-2 knockout mice compared to wild-type mice, suggesting that G protein biased µ opioid receptor agonists would be more efficacious with reduced adverse events. Here we describe our efforts to identify a potent, selective, and G protein biased µ opioid receptor agonist, TRV130 ((R)-30). This novel molecule demonstrated an improved therapeutic index (analgesia vs adverse effects) in rodent models and characteristics appropriate for clinical development. It is currently being evaluated in human clinical trials for the treatment of acute severe pain.


Subject(s)
Acute Pain/drug therapy , Analgesics/pharmacology , Drug Discovery/methods , Receptors, Opioid, mu/agonists , Spiro Compounds/pharmacology , Thiophenes/pharmacology , Acute Pain/pathology , Analgesics/chemical synthesis , Analgesics/chemistry , Animals , Disease Models, Animal , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Mice , Models, Chemical , Molecular Structure , Rats , Receptors, Opioid, mu/metabolism , Severity of Illness Index , Spiro Compounds/chemical synthesis , Spiro Compounds/chemistry , Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/chemistry
7.
J Clin Pharmacol ; 53(9): 892-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23813302

ABSTRACT

TRV027 is a novel ß-arrestin biased peptide ligand of the angiotensin II type 1 receptor (AT1R). The compound antagonizes G protein coupling while simultaneously stimulating ß-arrestin-mediated signaling. In preclinical studies, TRV027 reversibly reduced blood pressure while preserving renal function in a dog tachypaced heart failure model and stimulating cardiomyocyte contractility in vitro. This profile suggests that TRV027 may have unique benefits in acute heart failure, a condition associated with renin-angiotensin system activation. A first-time-in-human study was conducted with ascending doses of TRV027 to explore its tolerability, pharmacokinetics and pharmacodynamics in healthy volunteers. Subjects' salt intake was restricted to stimulate RAS activation. In this study TRV027 was safe and well tolerated with a short-half-life (ranging between 2.4 and 13.2 minutes) and dose-proportional increases in systemic exposure. Consistent with the pre-clinical findings, TRV027 reduced blood pressure to a greater degree in subjects with RAS activation, measured as elevated plasma renin activity, than in those with normal PRA levels. This study in sodium-restricted healthy subjects suggests that TRV027 will successfully target a core mechanism of acute heart failure pathophysiology. Further clinical studies with TRV027 in patients with heart failure are underway.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Antihypertensive Agents/pharmacology , Oligopeptides/pharmacology , Adult , Angiotensin II Type 1 Receptor Blockers/blood , Antihypertensive Agents/blood , Blood Pressure/drug effects , Cross-Over Studies , Diet, Sodium-Restricted , Double-Blind Method , Female , Humans , Male , Middle Aged , Oligopeptides/blood , Renin/blood
8.
J Pharmacol Exp Ther ; 344(3): 708-17, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23300227

ABSTRACT

The concept of ligand bias at G protein-coupled receptors broadens the possibilities for agonist activities and provides the opportunity to develop safer, more selective therapeutics. Morphine pharmacology in ß-arrestin-2 knockout mice suggested that a ligand that promotes coupling of the µ-opioid receptor (MOR) to G proteins, but not ß-arrestins, would result in higher analgesic efficacy, less gastrointestinal dysfunction, and less respiratory suppression than morphine. Here we report the discovery of TRV130 ([(3-methoxythiophen-2-yl)methyl]({2-[(9R)-9-(pyridin-2-yl)-6-oxaspiro[4.5]decan-9-yl]ethyl})amine), a novel MOR G protein-biased ligand. In cell-based assays, TRV130 elicits robust G protein signaling, with potency and efficacy similar to morphine, but with far less ß-arrestin recruitment and receptor internalization. In mice and rats, TRV130 is potently analgesic while causing less gastrointestinal dysfunction and respiratory suppression than morphine at equianalgesic doses. TRV130 successfully translates evidence that analgesic and adverse MOR signaling pathways are distinct into a biased ligand with differentiated pharmacology. These preclinical data suggest that TRV130 may be a safer and more tolerable therapeutic for treating severe pain.


Subject(s)
Analgesics/pharmacology , GTP-Binding Proteins/metabolism , Gastrointestinal Tract/drug effects , Morphine/pharmacology , Receptors, Opioid, mu/metabolism , Respiratory System/drug effects , Animals , Arrestins/metabolism , Cell Line , Gastrointestinal Diseases/chemically induced , Gastrointestinal Diseases/drug therapy , Gastrointestinal Diseases/metabolism , HEK293 Cells , Humans , Ligands , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/metabolism , Respiratory Tract Diseases/chemically induced , Respiratory Tract Diseases/drug therapy , Respiratory Tract Diseases/metabolism , Signal Transduction/drug effects , beta-Arrestin 2 , beta-Arrestins
9.
Bioorg Med Chem Lett ; 17(16): 4670-7, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17560786

ABSTRACT

The design, synthesis, and SAR of a novel series of heterobiaryl phenethanolamine beta3 adrenergic receptor agonists are described. The furan analogue 49 was shown to elicit a significant dose-dependent lowering of plasma glucose in a rodent model of type 2 diabetes.


Subject(s)
Adrenergic beta-3 Receptor Agonists , Carboxylic Acids/chemistry , Carboxylic Acids/pharmacology , Binding Sites , Models, Molecular , Molecular Structure , Structure-Activity Relationship
10.
J Med Chem ; 49(9): 2758-71, 2006 May 04.
Article in English | MEDLINE | ID: mdl-16640337

ABSTRACT

The synthesis of a series of phenethanolamine aniline agonists that contain an aniline ring on the right-hand side of the molecule substituted at the meta position with a benzoic acid or a pyridyl carboxylate is described. Several of the analogues (e.g., 34, 36-38, 40, and 44) have high beta(3) adrenergic receptor (AR) potency and selectivity against beta(1) and beta(2) ARs in Chinese hamster ovary (CHO) cells expressing beta ARs. The dog pharmacokinetic profile of some of these analogues showed >25% oral bioavailability and po half-lives of at least 1.5 h. Among the compounds described herein, the 3,3'-biarylaniline carboxylate derivatives 36, 38 and the phenylpyridyl derivative 44 demonstrated outstanding in vitro properties and reasonable dog pharmacokinetic profiles. These three analogues also showed dose dependent beta(3) AR mediated responses in mice. The ease of synthesis and superior dog pharmacokinetics of compound 38 relative to that of 44 in combination with its in vitro profile led us to choose this compound as a development candidate for the treatment of type 2 diabetes.


Subject(s)
Adrenergic beta-3 Receptor Agonists , Aniline Compounds/chemistry , Ethanolamine/chemistry , Ethanolamine/pharmacology , Receptors, Adrenergic, beta-3/metabolism , Animals , Blood Glucose/metabolism , Cell Line , Cricetinae , Cyclic AMP/metabolism , Dogs , Ethanolamine/chemical synthesis , Glycosylation/drug effects , Hemoglobins/metabolism , Humans , Male , Mice , Molecular Structure , Structure-Activity Relationship
11.
J Biol Chem ; 281(16): 10856-64, 2006 Apr 21.
Article in English | MEDLINE | ID: mdl-16492667

ABSTRACT

Parathyroid hormone (PTH) regulates calcium homeostasis via the type I PTH/PTH-related peptide (PTH/PTHrP) receptor (PTH1R). The purpose of the present study was to identify the contributions of distinct signaling mechanisms to PTH-stimulated activation of the mitogen-activated protein kinases (MAPK) ERK1/2. In Human embryonic kidney 293 (HEK293) cells transiently transfected with hPTH1R, PTH stimulated a robust increase in ERK activity. The time course of ERK1/2 activation was biphasic with an early peak at 10 min and a later sustained ERK1/2 activation persisting for greater than 60 min. Pretreatment of HEK293 cells with the PKA inhibitor H89 or the PKC inhibitor GF109203X, individually or in combination reduced the early component of PTH-stimulated ERK activity. However, these inhibitors of second messenger dependent kinases had little effect on the later phase of PTH-stimulated ERK1/2 phosphorylation. This later phase of ERK1/2 activation at 30-60 min was blocked by depletion of cellular beta-arrestin 2 and beta-arrestin 1 by small interfering RNA. Furthermore, stimulation of hPTH1R with PTH analogues, [Trp1]PTHrp-(1-36) and [d-Trp12,Tyr34]PTH-(7-34), selectively activated G(s)/PKA-mediated ERK1/2 activation or G protein-independent/beta-arrestin-dependent ERK1/2 activation, respectively. It is concluded that PTH stimulates ERK1/2 through several distinct signal transduction pathways: an early G protein-dependent pathway meditated by PKA and PKC and a late pathway independent of G proteins mediated through beta-arrestins. These findings imply the existence of distinct active conformations of the hPTH1R responsible for the two pathways, which can be stimulated by unique ligands. Such ligands may have distinct and valuable therapeutic properties.


Subject(s)
Arrestins/physiology , GTP-Binding Proteins/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Receptor, Parathyroid Hormone, Type 1/metabolism , Arrestins/metabolism , Cell Line , Cells, Cultured , Culture Media, Serum-Free/pharmacology , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA/metabolism , DNA, Complementary/metabolism , GTP-Binding Proteins/metabolism , Humans , Immunoblotting , Immunoprecipitation , Indoles/pharmacology , Isoquinolines/pharmacology , Ligands , MAP Kinase Signaling System , Maleimides/pharmacology , Mutation , Phosphorylation , Protein Kinases/metabolism , RNA, Small Interfering/metabolism , Receptor, Parathyroid Hormone, Type 1/genetics , Signal Transduction , Sulfonamides/pharmacology , Time Factors , Transfection , beta-Arrestin 1 , beta-Arrestin 2 , beta-Arrestins
12.
J Biomol Screen ; 10(5): 476-84, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16093557

ABSTRACT

The authors demonstrate the use of a simple, universal G-protein-coupled receptor (GPCR) assay to screen for agonists for a specific GPCR. Cells stably expressing a green fluorescent protein (GFP)-labeled beta-arrestin fusion protein and the vasopressin V2 receptor (V2R) were used in a high-content screening (HCS) assay to screen a small peptide library for V2R agonists. Cells were treated with the peptides at a final concentration of 500 nM for 30 min. Agonist stimulation causes V2R internalization into endosomes. GFP-beta-arrestin remains associated with the V2R in endosomes, resulting in a fluorescent pattern of intracellular spots. Assay plates were automatically imaged and quantitatively analyzed using an HCS imaging platform and a fast turnkey image analysis application optimized for detection of receptor activation and intracellular spots. Hits were further evaluated to determine their potency. The combination of unique biology, automated high-content analysis, and a powerful means of validating hits results in better leads.


Subject(s)
Drug Evaluation, Preclinical/methods , Receptors, Vasopressin/agonists , Spectrometry, Fluorescence/methods , Animals , Arginine/chemistry , Arrestins/metabolism , Benzimidazoles/pharmacology , Cell Line, Tumor , Cloning, Molecular , Dose-Response Relationship, Drug , Endosomes/metabolism , Fluorescent Dyes/pharmacology , Gene Library , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/metabolism , Humans , Image Processing, Computer-Assisted , Luminescent Proteins/chemistry , Peptides/chemistry , Swine , Vasopressins/chemistry , beta-Arrestins
13.
J Med Chem ; 45(3): 567-83, 2002 Jan 31.
Article in English | MEDLINE | ID: mdl-11806709

ABSTRACT

Starting from phenethanolamine aniline leads 3a and 3b, we have identified a series of functionally potent and selective beta(3) adrenergic receptor (AR) agonists containing acylsulfonamide, sulfonylsulfonamide, or sulfonylurea groups within the aniline phenethanolamine series. In beta(3), beta(2), and beta(1) AR cAMP functional assays, 3a and other right-hand side (RHS) carboxylate analogues were found to be full agonists that were modestly selective against beta(1) or beta(2) ARs, while analogues lacking RHS acid functionality were active at beta(3) AR but not selective. Replacement of the carboxylate with acylthiazole and acylmethylsulfone gave potent, but only modestly selective, compounds. Increasing the size of the RHS sulfonamide substituent with phenyl or p-toluene afforded compounds with good potency and functional selectivity (beta(3) AR pEC(50) greater than 8; beta(1) and beta(2) AR selectivity greater than 40- and 500-fold, respectively). Our SAR studies suggest that the potency and selectivity profile of the best analogues reported here is a result of both the steric bulk and acidity of the RHS sulfonamide NH group. Although all of the analogues had a pharmacokinetic half-life of less than 2 h, acylsulfonamides 43 and 44 did show moderately low clearance in dogs. These two compounds were further evaluated by thermographic imaging in mice and were found to produce a robust thermogenic response via oral administration.


Subject(s)
Adrenergic beta-Agonists/chemical synthesis , Aniline Compounds/chemical synthesis , Receptors, Adrenergic, beta-3/drug effects , Sulfonamides/chemical synthesis , Sulfonylurea Compounds/chemical synthesis , Adrenergic beta-Agonists/chemistry , Adrenergic beta-Agonists/pharmacology , Aniline Compounds/chemistry , Aniline Compounds/pharmacology , Animals , Biological Availability , Body Temperature/drug effects , CHO Cells , Chromatography, High Pressure Liquid , Cricetinae , Cyclic AMP/biosynthesis , Dogs , Humans , Magnetic Resonance Spectroscopy , Male , Mass Spectrometry , Mice , Radioligand Assay , Stereoisomerism , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/pharmacology , Sulfonylurea Compounds/chemistry , Sulfonylurea Compounds/pharmacology , Thermography
SELECTION OF CITATIONS
SEARCH DETAIL
...