Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Neuropharmacology ; 148: 77-86, 2019 04.
Article in English | MEDLINE | ID: mdl-30553828

ABSTRACT

Headaches are highly disabling and are among the most common neurological disorders worldwide. Despite the high prevalence of headache, therapeutic options are limited. We recently identified the delta opioid receptor (DOR) as an emerging therapeutic target for migraine. In this study, we examined the effectiveness of a hallmark DOR agonist, SNC80, in disease models reflecting diverse headache disorders including: chronic migraine, post-traumatic headache (PTH), medication overuse headache by triptans (MOH), and opioid-induced hyperalgesia (OIH). To model chronic migraine C57BL/6J mice received chronic intermittent treatment with the known human migraine trigger, nitroglycerin. PTH was modeled by combining the closed head weight drop model with the nitroglycerin model of chronic migraine. For MOH and OIH, mice were chronically treated with sumatriptan or morphine, respectively. The development of periorbital and peripheral allodynia was observed in all four models; and SNC80 significantly inhibited allodynia in all cases. In addition, we also determined if chronic daily treatment with SNC80 would induce MOH/OIH, and we observed limited hyperalgesia relative to sumatriptan or morphine. Together, our results indicate that DOR agonists could be effective in multiple headache disorders, despite their distinct etiology, thus presenting a novel therapeutic target for headache.


Subject(s)
Benzamides/therapeutic use , Headache Disorders/drug therapy , Piperazines/therapeutic use , Receptors, Opioid, delta/agonists , Animals , Benzamides/adverse effects , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Headache Disorders/chemically induced , Hyperalgesia/drug therapy , Male , Mice , Morphine , Nitroglycerin , Piperazines/adverse effects , Receptors, Opioid, delta/therapeutic use , Sumatriptan
2.
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
3.
Curr Opin Pharmacol ; 32: 77-84, 2017 02.
Article in English | MEDLINE | ID: mdl-27936408

ABSTRACT

Opioid chemistry and biology occupy a pivotal place in the history of pharmacology and medicine. Morphine offers unmatched efficacy in alleviating acute pain, but is also associated with a host of adverse side effects. The advent of biased agonism at G protein-coupled receptors has expanded our understanding of intracellular signaling and highlighted the concept that certain ligands are able to differentially modulate downstream pathways. The ability to target one pathway over another has allowed for the development of biased ligands with robust clinical efficacy and fewer adverse events. In this review we summarize these concepts with an emphasis on biased mu opioid receptor pharmacology and highlight how far opioid pharmacology has evolved.


Subject(s)
Analgesics, Opioid/pharmacology , Pain/drug therapy , Receptors, Opioid, mu/agonists , Analgesics, Opioid/adverse effects , Animals , Drug Design , Humans , Ligands , Morphine/adverse effects , Morphine/pharmacology , Pain/physiopathology , Receptors, Opioid, mu/metabolism , Signal Transduction/drug effects
4.
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
5.
JACC Heart Fail ; 3(3): 193-201, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25650371

ABSTRACT

The BLAST-AHF (Biased Ligand of the Angiotensin Receptor Study in Acute Heart Failure) study is designed to test the efficacy and safety of TRV027, a novel biased ligand of the angiotensin-2 type 1 receptor, in patients with acute heart failure (AHF). AHF remains a major public health problem, and no currently-available therapies have been shown to favorably affect outcomes. TRV027 is a novel biased ligand of the angiotensin-2 type 1 receptor that antagonizes angiotensin-stimulated G-protein activation while stimulating ß-arrestin. In animal models, these effects reduce afterload while increasing cardiac performance and maintaining stroke volume. In initial human studies, TRV027 appears to be hemodynamically active primarily in patients with activation of the renin-angiotensin-aldosterone system, a potentially attractive profile for an AHF therapeutic. BLAST-AHF is an international prospective, randomized, phase IIb, dose-ranging study that will randomize up to 500 AHF patients with systolic blood pressure ≥120 mm Hg and ≤200 mm Hg within 24 h of initial presentation to 1 of 3 doses of intravenous TRV027 (1, 5, or 25 mg/h) or matching placebo (1:1:1:1) for at least 48 h and up to 96 h. The primary endpoint is a composite of 5 clinical endpoints (dyspnea, worsening heart failure, length of hospital stay, 30-day rehospitalization, and 30-day mortality) combined using an average z-score. Secondary endpoints will include the assessment of dyspnea and change in amino-terminal pro-B-type natriuretic peptide. The BLAST-AHF study will assess the efficacy and safety of a novel biased ligand of the angiotensin-2 type 1 receptor in AHF.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Clinical Trials as Topic/methods , Heart Failure/drug therapy , Receptor, Angiotensin, Type 1/metabolism , Acute Disease , Humans , Receptor, Angiotensin, Type 1/drug effects
6.
Trends Pharmacol Sci ; 35(7): 308-16, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24878326

ABSTRACT

Drug discovery targeting G protein-coupled receptors (GPCRs) is no longer limited to seeking agonists or antagonists to stimulate or block cellular responses associated with a particular receptor. GPCRs are now known to support a diversity of pharmacological profiles, a concept broadly referred to as functional selectivity. In particular, the concept of ligand bias, whereby a ligand stabilizes subsets of receptor conformations to engender novel pharmacological profiles, has recently gained increasing prominence. This review discusses how biased ligands may deliver safer, better tolerated, and more efficacious drugs, and highlights several biased ligands that are in clinical development. Biased ligands targeting the angiotensin II type 1 receptor and the µ opioid receptor illustrate the translation of the biased ligand concept from basic biology to clinical drug development.


Subject(s)
Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Drug Design , Humans , Ligands , Oligopeptides/pharmacology , Receptor, Angiotensin, Type 1/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Opioid, mu/metabolism , Spiro Compounds/pharmacology , Thiophenes/pharmacology
7.
Pain ; 155(9): 1829-1835, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24954166

ABSTRACT

Opioids provide powerful analgesia but also efficacy-limiting adverse effects, including severe nausea, vomiting, and respiratory depression, by activating µ-opioid receptors. Preclinical models suggest that differential activation of signaling pathways downstream of these receptors dissociates analgesia from adverse effects; however, this has not yet translated to a treatment with an improved therapeutic index. Thirty healthy men received single intravenous injections of the biased ligand TRV130 (1.5, 3, or 4.5mg), placebo, or morphine (10mg) in a randomized, double-blind, crossover study. Primary objectives were to measure safety and tolerability (adverse events, vital signs, electrocardiography, clinical laboratory values), and analgesia (cold pain test) versus placebo. Other measures included respiratory drive (minute volume after induced hypercapnia), subjective drug effects, and pharmacokinetics. Compared to morphine, TRV130 (3, 4.5mg) elicited higher peak analgesia (105, 116 seconds latency vs 75 seconds for morphine, P<.02), with faster onset and similar duration of action. More subjects doubled latency or achieved maximum latency (180 seconds) with TRV130 (3, 4.5mg). Respiratory drive reduction was greater after morphine than any TRV130 dose (-15.9 for morphine versus -7.3, -7.6, and -9.4 h*L/min, P<.05). More subjects experienced severe nausea after morphine (n=7) than TRV130 1.5 or 3mg (n=0, 1), but not 4.5mg (n=9). TRV130 was generally well tolerated, and exposure was dose proportional. Thus, in this study, TRV130 produced greater analgesia than morphine at doses with less reduction in respiratory drive and less severe nausea. This demonstrates early clinical translation of ligand bias as an important new concept in receptor-targeted pharmacotherapy.


Subject(s)
Analgesics, Opioid/adverse effects , Morphine/adverse effects , Pain/drug therapy , Spiro Compounds/adverse effects , Thiophenes/adverse effects , Adolescent , Adult , Analgesia , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/therapeutic use , Cross-Over Studies , Dizziness/chemically induced , Dose-Response Relationship, Drug , Double-Blind Method , Headache/chemically induced , Healthy Volunteers , Humans , Male , Middle Aged , Morphine/administration & dosage , Morphine/therapeutic use , Spiro Compounds/administration & dosage , Spiro Compounds/therapeutic use , Thiophenes/administration & dosage , Thiophenes/therapeutic use , Vomiting/chemically induced , Young Adult
8.
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
9.
J Biol Chem ; 289(20): 14211-24, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24668815

ABSTRACT

The concept of "biased agonism" arises from the recognition that the ability of an agonist to induce a receptor-mediated response (i.e. "efficacy") can differ across the multiple signal transduction pathways (e.g. G protein and ß-arrestin (ßarr)) emanating from a single GPCR. Despite the therapeutic promise of biased agonism, the molecular mechanism(s) whereby biased agonists selectively engage signaling pathways remain elusive. This is due in large part to the challenges associated with quantifying ligand efficacy in cells. To address this, we developed a cell-free approach to directly quantify the transducer-specific molecular efficacies of balanced and biased ligands for the angiotensin II type 1 receptor (AT1R), a prototypic GPCR. Specifically, we defined efficacy in allosteric terms, equating shifts in ligand affinity (i.e. KLo/KHi) at AT1R-Gq and AT1R-ßarr2 fusion proteins with their respective molecular efficacies for activating Gq and ßarr2. Consistent with ternary complex model predictions, transducer-specific molecular efficacies were strongly correlated with cellular efficacies for activating Gq and ßarr2. Subsequent comparisons across transducers revealed that biased AT1R agonists possess biased molecular efficacies that were in strong agreement with the signaling bias observed in cellular assays. These findings not only represent the first measurements of the thermodynamic driving forces underlying differences in ligand efficacy between transducers but also support a molecular mechanism whereby divergent transducer-specific molecular efficacies generate biased agonism at a GPCR.


Subject(s)
Receptor, Angiotensin, Type 1/agonists , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction , Allosteric Regulation , HEK293 Cells , Humans , Ligands , Recombinant Fusion Proteins/metabolism , Thermodynamics
10.
Chem Biol ; 21(4): 459-469, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24631122

ABSTRACT

Optimal tuning of enzyme signaling is critical for cellular homeostasis. We use fluorescence resonance energy transfer reporters in live cells to follow conformational transitions that tune the affinity of a multidomain signal transducer, protein kinase C (PKC), for optimal response to second messengers. This enzyme comprises two diacylglycerol sensors, the C1A and C1B domains, that have a sufficiently high intrinsic affinity for ligand so that the enzyme would be in a ligand-engaged, active state if not for mechanisms that mask its domains. We show that both diacylglycerol sensors are exposed in newly synthesized PKC and that conformational transitions following priming phosphorylations mask the domains so that the lower affinity sensor, the C1B domain, is the primary diacylglycerol binder. The conformational rearrangements of PKC serve as a paradigm for how multimodule transducers optimize their dynamic range of signaling.


Subject(s)
Protein Kinase C/chemistry , Protein Kinase C/metabolism , Signal Transduction , Animals , COS Cells , Cattle , Cells, Cultured , Chlorocebus aethiops , Diglycerides/chemistry , Diglycerides/metabolism , Fluorescence Resonance Energy Transfer , Mice , Models, Molecular , Phosphorylation , Protein Conformation , Rats
11.
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
12.
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
13.
Am J Physiol Heart Circ Physiol ; 305(6): H856-66, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23873795

ABSTRACT

In the present study, we compared the cardioprotective effects of TRV120023, a novel angiotensin II (ANG II) type 1 receptor (AT1R) ligand, which blocks G protein coupling but stimulates ß-arrestin signaling, against treatment with losartan, a conventional AT1R blocker in the treatment of cardiac hypertrophy and regulation of myofilament activity and phosphorylation. Rats were subjected to 3 wk of treatment with saline, ANG II, ANG II + losartan, ANG II + TRV120023, or TRV120023 alone. ANG II induced increased left ventricular mass compared with rats that received ANG II + losartan or ANG II + TRV120023. Compared with saline controls, ANG II induced a significant increase in pCa50 and maximum Ca(2+)-activated myofilament tension but reduced the Hill coefficient (nH). TRV120023 increased maximum tension and pCa50, although to lesser extent than ANG II. In contrast to ANG II, TRV120023 increased nH. Losartan blocked the effects of ANG II on pCa50 and nH and reduced maximum tension below that of saline controls. ANG II + TRV120023 showed responses similar to those of TRV120023 alone; compared with ANG II + losartan, ANG II + TRV120023 preserved maximum tension and increased both pCa50 and cooperativity. Tropomyosin phosphorylation was lower in myofilaments from saline-treated hearts compared with the other groups. Phosphorylation of cardiac troponin I was significantly reduced in ANG II + TRV120023 and TRV120023 groups versus saline controls, and myosin-binding protein C phosphorylation at Ser(282) was unaffected by ANG II or losartan but significantly reduced with TRV120023 treatment compared with all other groups. Our data indicate that TRV120023-related promotion of ß-arrestin signaling and enhanced contractility involves a mechanism promoting the myofilament response to Ca(2+) via altered protein phosphorylation. Selective activation of ß-arrestin-dependent pathways may provide advantages over conventional AT1R blockers.


Subject(s)
Arrestins/metabolism , Calcium/metabolism , Cardiomegaly/drug therapy , Cardiomegaly/physiopathology , Heart Ventricles/physiopathology , Myofibrils/drug effects , Oligopeptides/administration & dosage , Angiotensin II , Animals , Cardiomegaly/chemically induced , Cardiotonic Agents/administration & dosage , Heart Ventricles/drug effects , Heart Ventricles/pathology , Male , Myocardial Contraction/drug effects , Organ Size/drug effects , Rats , Rats, Sprague-Dawley , Treatment Outcome , Up-Regulation/drug effects , beta-Arrestins
14.
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
15.
Trends Cardiovasc Med ; 23(7): 242-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23499300

ABSTRACT

G protein-coupled receptors have been successfully targeted by numerous therapeutics including drugs that have transformed the management of cardiovascular disease. However, many GPCRs, when activated or blocked by drugs, elicit both beneficial and adverse pharmacology. Recent work has demonstrated that in some cases, the salutary and deleterious signals linked to a specific GPCR can be selectively targeted by "biased ligands" that entrain subsets of a receptor's normal pharmacology. This review briefly summarizes the advances and current state of the biased ligand field, focusing on an example: biased ligands targeting the angiotensin II type 1 receptor. These compounds exhibit unique pharmacology, distinct from classic agonists or antagonists, and one such molecule is now in clinical development for the treatment of acute heart failure.


Subject(s)
Angiotensin II Type 1 Receptor Blockers , Heart Failure , Receptors, G-Protein-Coupled , Angiotensin II Type 1 Receptor Blockers/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Drug Discovery , Heart Failure/drug therapy , Heart Failure/metabolism , Humans , Ligands , Molecular Targeted Therapy/adverse effects , Molecular Targeted Therapy/methods , Receptors, G-Protein-Coupled/classification , Receptors, G-Protein-Coupled/therapeutic use
16.
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
17.
Circ Heart Fail ; 5(5): 627-34, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22891045

ABSTRACT

BACKGROUND: TRV120027 is a novel ß-arrestin biased ligand of the angiotensin II type 1 receptor; it antagonizes canonical G-protein-mediated coupling while, in contrast to classical angiotensin II type 1 receptor antagonists, it engages ß-arrestin-mediated signaling. Consequently, TRV120027 inhibits angiotensin II-mediated vasoconstriction while, via ß-arrestin coupling, it increases cardiomyocyte contractility. We hypothesized that TRV120027 would elicit beneficial cardiorenal actions when added to furosemide in experimental heart failure. METHODS AND RESULTS: Two groups of anesthetized dogs (n=6 each) with tachypacing-induced heart failure were studied. After a baseline clearance, 1 group (F+V) received furosemide (1 mg/kg per hour) plus saline for 90 minutes, whereas the other (F+T) received the same dose of furosemide plus TRV120027 (0.3 and 1.5 µg/kg per minute for 45 minutes each); 2 clearances were done during drug infusion. After a washout, a postinfusion clearance was done; *P<0.05 between groups. F+V and F+T increased diuresis and natriuresis to a similar extent during drug administration, but urine flow* and urinary sodium excretion* were higher in the postinfusion clearance with F+T. Glomerular filtration rate was preserved in both groups. Renal blood flow increased with F+T but this was not significant versus F+V. Compared with F+V, F+T decreased mean arterial pressure*, systemic* and pulmonary* vascular resistances, and atrial natriuretic peptide*. Pulmonary capillary wedge pressure* decreased to a larger extent with F+T than with F+V. CONCLUSIONS: When added to furosemide, TRV120027, a novel ß-arrestin biased angiotensin II type 1 receptor ligand, preserved furosemide-mediated natriuresis and diuresis, while reducing cardiac preload and afterload. These results provide support for TRV120027 as a promising novel therapeutic for the treatment of heart failure.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Arrestins/metabolism , Diuretics/pharmacology , Furosemide/pharmacology , Heart Failure/drug therapy , Heart/drug effects , Kidney/drug effects , Oligopeptides/pharmacology , Receptor, Angiotensin, Type 1/drug effects , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Animals , Arterial Pressure/drug effects , Atrial Natriuretic Factor/blood , Cardiac Pacing, Artificial , Disease Models, Animal , Diuresis/drug effects , Diuretics/administration & dosage , Dogs , Drug Therapy, Combination , Furosemide/administration & dosage , Glomerular Filtration Rate/drug effects , Heart/physiopathology , Heart Failure/metabolism , Heart Failure/physiopathology , Infusions, Intravenous , Kidney/metabolism , Kidney/physiopathology , Ligands , Male , Natriuresis/drug effects , Oligopeptides/administration & dosage , Pulmonary Wedge Pressure/drug effects , Receptor, Angiotensin, Type 1/metabolism , Renal Circulation/drug effects , Signal Transduction/drug effects , Time Factors , Urodynamics/drug effects , Vascular Resistance/drug effects , beta-Arrestins
18.
Am J Physiol Heart Circ Physiol ; 303(8): H1001-10, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22886417

ABSTRACT

Pharmacological blockade of the ANG II type 1 receptor (AT1R) is a common therapy for treatment of congestive heart failure and hypertension. Increasing evidence suggests that selective engagement of ß-arrestin-mediated AT1R signaling, referred to as biased signaling, promotes cardioprotective signaling. Here, we tested the hypothesis that a ß-arrestin-biased AT1R ligand TRV120023 would confer cardioprotection in response to acute cardiac injury compared with the traditional AT1R blocker (ARB), losartan. TRV120023 promotes cardiac contractility, assessed by pressure-volume loop analyses, while blocking the effects of endogenous ANG II. Compared with losartan, TRV120023 significantly activates MAPK and Akt signaling pathways. These hemodynamic and biochemical effects were lost in ß-arrestin-2 knockout (KO) mice. In response to cardiac injury induced by ischemia reperfusion injury or mechanical stretch, pretreatment with TRV120023 significantly diminishes cell death compared with losartan, which did not appear to be cardioprotective. This cytoprotective effect was lost in ß-arrestin-2 KO mice. The ß-arrestin-biased AT1R ligand, TRV120023, has cardioprotective and functional properties in vivo, which are distinct from losartan. Our data suggest that this novel class of drugs may provide an advantage over conventional ARBs by supporting cardiac function and reducing cellular injury during acute cardiac injury.


Subject(s)
Acute Coronary Syndrome/drug therapy , Adaptor Proteins, Signal Transducing/metabolism , Arrestins/metabolism , Cardiotonic Agents/pharmacology , Losartan/pharmacology , Oligopeptides/pharmacology , Acute Coronary Syndrome/metabolism , Acute Coronary Syndrome/pathology , Acute Disease , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Arrestins/genetics , Cell Survival/drug effects , Cell Survival/physiology , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Myocardium/metabolism , Myocardium/pathology , Signal Transduction/drug effects , Signal Transduction/physiology , beta-Arrestin 2 , beta-Arrestins
19.
Mol Syst Biol ; 8: 590, 2012 Jun 26.
Article in English | MEDLINE | ID: mdl-22735336

ABSTRACT

Seven-transmembrane receptors (7TMRs) are involved in nearly all aspects of chemical communications and represent major drug targets. 7TMRs transmit their signals not only via heterotrimeric G proteins but also through ß-arrestins, whose recruitment to the activated receptor is regulated by G protein-coupled receptor kinases (GRKs). In this paper, we combined experimental approaches with computational modeling to decipher the molecular mechanisms as well as the hidden dynamics governing extracellular signal-regulated kinase (ERK) activation by the angiotensin II type 1A receptor (AT(1A)R) in human embryonic kidney (HEK)293 cells. We built an abstracted ordinary differential equations (ODE)-based model that captured the available knowledge and experimental data. We inferred the unknown parameters by simultaneously fitting experimental data generated in both control and perturbed conditions. We demonstrate that, in addition to its well-established function in the desensitization of G-protein activation, GRK2 exerts a strong negative effect on ß-arrestin-dependent signaling through its competition with GRK5 and 6 for receptor phosphorylation. Importantly, we experimentally confirmed the validity of this novel GRK2-dependent mechanism in both primary vascular smooth muscle cells naturally expressing the AT(1A)R, and HEK293 cells expressing other 7TMRs.


Subject(s)
Arrestins/metabolism , G-Protein-Coupled Receptor Kinase 2/metabolism , G-Protein-Coupled Receptor Kinases/metabolism , GTP-Binding Proteins/metabolism , Models, Biological , Signal Transduction , Cell Line , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , G-Protein-Coupled Receptor Kinase 3/metabolism , G-Protein-Coupled Receptor Kinase 5/metabolism , Humans , Kidney/cytology , Kidney/embryology , Kidney/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Receptor, Angiotensin, Type 1/metabolism , beta-Arrestins
20.
Circ Heart Fail ; 4(6): 770-8, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21835984

ABSTRACT

BACKGROUND: The angiotensin II type 1 receptor (AT1R) plays a key role in regulating cardiorenal function. Classic "unbiased" AT1R antagonists block receptor coupling to both G(αq) and ß-arrestin-mediated signals, which desensitize G-protein signaling as well as transduce G-protein-independent signals. TRV120027 is a novel ß-arrestin-biased AT1R ligand, which engages ß-arrestins while blocking G-protein signaling. At the AT1R, TRV120027 can inhibit angiotensin II-mediated vasoconstriction, whereas, through ß-arrestin coupling, increase cardiomyocyte contractility. We defined for the first time the acute cardiorenal actions of TRV120027 in healthy and heart failure (HF) canines. METHODS AND RESULTS: Healthy and HF canines (induced by tachypacing) were anesthetized. After instrumentation and equilibration, a 30-minute baseline clearance was performed, followed by further clearance with escalating doses of intravenous TRV120027 (0.01, 0.1, 1, 10, and 100 µg/kg per minute) and a 30-minute washout. In healthy canines, TRV120027 decreased pulmonary capillary wedge pressure and systemic and renal vascular resistances, while increasing cardiac output, renal blood flow, glomerular filtration rate, and urinary sodium excretion. In HF canines, TRV120027 decreased mean arterial pressure, right atrial pressure, and pulmonary capillary wedge pressure, systemic and renal vascular resistances and increased cardiac output and renal blood flow. Glomerular filtration rate and urinary sodium excretion were maintained. CONCLUSIONS: We report for the first time the cardiorenal actions of the novel ß-arrestin-biased AT1R ligand TRV120027. In both normal and HF canines, TRV120027 demonstrated cardiac unloading actions while preserving renal function. With this beneficial pharmacological profile, TRV120027 represents a novel strategy for the treatment of HF.


Subject(s)
Arrestins/metabolism , Heart Failure/drug therapy , Heart/drug effects , Kidney/drug effects , Oligopeptides/pharmacology , Oligopeptides/therapeutic use , Receptor, Angiotensin, Type 1/metabolism , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Cardiac Output/drug effects , Cardiac Output/physiology , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Glomerular Filtration Rate/drug effects , Glomerular Filtration Rate/physiology , Heart/physiopathology , Heart Failure/metabolism , Heart Failure/physiopathology , Kidney/physiopathology , Ligands , Male , Sodium/urine , Treatment Outcome , Vascular Resistance/drug effects , Vascular Resistance/physiology , beta-Arrestins
SELECTION OF CITATIONS
SEARCH DETAIL