Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Nature ; 553(7686): 106-110, 2018 01 03.
Article in English | MEDLINE | ID: mdl-29300013

ABSTRACT

Class B G-protein-coupled receptors (GPCRs), which consist of an extracellular domain (ECD) and a transmembrane domain (TMD), respond to secretin peptides to play a key part in hormonal homeostasis, and are important therapeutic targets for a variety of diseases. Previous work has suggested that peptide ligands bind to class B GPCRs according to a two-domain binding model, in which the C-terminal region of the peptide targets the ECD and the N-terminal region of the peptide binds to the TMD binding pocket. Recently, three structures of class B GPCRs in complex with peptide ligands have been solved. These structures provide essential insights into peptide ligand recognition by class B GPCRs. However, owing to resolution limitations, the specific molecular interactions for peptide binding to class B GPCRs remain ambiguous. Moreover, these previously solved structures have different ECD conformations relative to the TMD, which introduces questions regarding inter-domain conformational flexibility and the changes required for receptor activation. Here we report the 3.0 Å-resolution crystal structure of the full-length human glucagon receptor (GCGR) in complex with a glucagon analogue and partial agonist, NNC1702. This structure provides molecular details of the interactions between GCGR and the peptide ligand. It reveals a marked change in the relative orientation between the ECD and TMD of GCGR compared to the previously solved structure of the inactive GCGR-NNC0640-mAb1 complex. Notably, the stalk region and the first extracellular loop undergo major conformational changes in secondary structure during peptide binding, forming key interactions with the peptide. We further propose a dual-binding-site trigger model for GCGR activation-which requires conformational changes of the stalk, first extracellular loop and TMD-that extends our understanding of the previously established two-domain peptide-binding model of class B GPCRs.


Subject(s)
Glucagon/analogs & derivatives , Receptors, Glucagon/chemistry , Receptors, Glucagon/metabolism , Crystallography, X-Ray , Drug Partial Agonism , Humans , Ligands , Models, Molecular , Protein Binding , Protein Conformation
2.
Nature ; 546(7657): 312-315, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28514449

ABSTRACT

The glucagon-like peptide-1 receptor (GLP-1R) and the glucagon receptor (GCGR) are members of the secretin-like class B family of G-protein-coupled receptors (GPCRs) and have opposing physiological roles in insulin release and glucose homeostasis. The treatment of type 2 diabetes requires positive modulation of GLP-1R to inhibit glucagon secretion and stimulate insulin secretion in a glucose-dependent manner. Here we report crystal structures of the human GLP-1R transmembrane domain in complex with two different negative allosteric modulators, PF-06372222 and NNC0640, at 2.7 and 3.0 Å resolution, respectively. The structures reveal a common binding pocket for negative allosteric modulators, present in both GLP-1R and GCGR and located outside helices V-VII near the intracellular half of the receptor. The receptor is in an inactive conformation with compounds that restrict movement of the intracellular tip of helix VI, a movement that is generally associated with activation mechanisms in class A GPCRs. Molecular modelling and mutagenesis studies indicate that agonist positive allosteric modulators target the same general region, but in a distinct sub-pocket at the interface between helices V and VI, which may facilitate the formation of an intracellular binding site that enhances G-protein coupling.


Subject(s)
Glucagon-Like Peptide-1 Receptor/chemistry , Glucagon-Like Peptide-1 Receptor/metabolism , Allosteric Regulation/drug effects , Allosteric Site/drug effects , Amino Acid Sequence , Aminopyridines/chemistry , Aminopyridines/metabolism , Aminopyridines/pharmacology , Benzamides/chemistry , Benzamides/metabolism , Benzamides/pharmacology , Crystallography, X-Ray , Glucagon-Like Peptide-1 Receptor/agonists , Humans , Models, Molecular , Phenylurea Compounds/chemistry , Phenylurea Compounds/metabolism , Phenylurea Compounds/pharmacology , Protein Domains
3.
Nature ; 546(7657): 259-264, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28514451

ABSTRACT

The human glucagon receptor, GCGR, belongs to the class B G-protein-coupled receptor family and plays a key role in glucose homeostasis and the pathophysiology of type 2 diabetes. Here we report the 3.0 Å crystal structure of full-length GCGR containing both the extracellular domain and transmembrane domain in an inactive conformation. The two domains are connected by a 12-residue segment termed the stalk, which adopts a ß-strand conformation, instead of forming an α-helix as observed in the previously solved structure of the GCGR transmembrane domain. The first extracellular loop exhibits a ß-hairpin conformation and interacts with the stalk to form a compact ß-sheet structure. Hydrogen-deuterium exchange, disulfide crosslinking and molecular dynamics studies suggest that the stalk and the first extracellular loop have critical roles in modulating peptide ligand binding and receptor activation. These insights into the full-length GCGR structure deepen our understanding of the signalling mechanisms of class B G-protein-coupled receptors.


Subject(s)
Receptors, Glucagon/chemistry , Receptors, Glucagon/classification , Allosteric Site/drug effects , Benzamides/chemistry , Benzamides/metabolism , Benzamides/pharmacology , Cell Membrane/metabolism , Cross-Linking Reagents/chemistry , Crystallography, X-Ray , Deuterium Exchange Measurement , Disulfides/chemistry , Humans , Ligands , Models, Molecular , Molecular Dynamics Simulation , Phenylurea Compounds/chemistry , Phenylurea Compounds/metabolism , Phenylurea Compounds/pharmacology , Protein Domains , Protein Stability , Receptors, Glucagon/agonists , Receptors, Glucagon/metabolism
4.
Nature ; 499(7459): 444-9, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23863937

ABSTRACT

Binding of the glucagon peptide to the glucagon receptor (GCGR) triggers the release of glucose from the liver during fasting; thus GCGR plays an important role in glucose homeostasis. Here we report the crystal structure of the seven transmembrane helical domain of human GCGR at 3.4 Å resolution, complemented by extensive site-specific mutagenesis, and a hybrid model of glucagon bound to GCGR to understand the molecular recognition of the receptor for its native ligand. Beyond the shared seven transmembrane fold, the GCGR transmembrane domain deviates from class A G-protein-coupled receptors with a large ligand-binding pocket and the first transmembrane helix having a 'stalk' region that extends three alpha-helical turns above the plane of the membrane. The stalk positions the extracellular domain (~12 kilodaltons) relative to the membrane to form the glucagon-binding site that captures the peptide and facilitates the insertion of glucagon's amino terminus into the seven transmembrane domain.


Subject(s)
Receptors, Glucagon/chemistry , Receptors, Glucagon/classification , Amino Acid Sequence , Binding Sites , Cell Membrane/metabolism , Crystallography, X-Ray , Glucagon/chemistry , Glucagon/metabolism , Humans , Ligands , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding , Protein Structure, Tertiary , Receptors, CXCR4/chemistry , Receptors, CXCR4/classification , Receptors, Glucagon/genetics , Receptors, Glucagon/metabolism
5.
Pharmacol Rev ; 68(4): 954-1013, 2016 10.
Article in English | MEDLINE | ID: mdl-27630114

ABSTRACT

The glucagon-like peptide (GLP)-1 receptor (GLP-1R) is a class B G protein-coupled receptor (GPCR) that mediates the action of GLP-1, a peptide hormone secreted from three major tissues in humans, enteroendocrine L cells in the distal intestine, α cells in the pancreas, and the central nervous system, which exerts important actions useful in the management of type 2 diabetes mellitus and obesity, including glucose homeostasis and regulation of gastric motility and food intake. Peptidic analogs of GLP-1 have been successfully developed with enhanced bioavailability and pharmacological activity. Physiologic and biochemical studies with truncated, chimeric, and mutated peptides and GLP-1R variants, together with ligand-bound crystal structures of the extracellular domain and the first three-dimensional structures of the 7-helical transmembrane domain of class B GPCRs, have provided the basis for a two-domain-binding mechanism of GLP-1 with its cognate receptor. Although efforts in discovering therapeutically viable nonpeptidic GLP-1R agonists have been hampered, small-molecule modulators offer complementary chemical tools to peptide analogs to investigate ligand-directed biased cellular signaling of GLP-1R. The integrated pharmacological and structural information of different GLP-1 analogs and homologous receptors give new insights into the molecular determinants of GLP-1R ligand selectivity and functional activity, thereby providing novel opportunities in the design and development of more efficacious agents to treat metabolic disorders.


Subject(s)
Glucagon-Like Peptide 1 , Receptors, G-Protein-Coupled , Animals , Glucagon-Like Peptide 1/chemistry , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/metabolism , Humans , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism
6.
PLoS One ; 17(3): e0264974, 2022.
Article in English | MEDLINE | ID: mdl-35245328

ABSTRACT

During recent years combining GLP-1 and glucagon receptor agonism with the purpose of achieving superior weight loss and metabolic control compared to GLP-1 alone has received much attention. The superior efficacy has been shown by several in preclinical models but has been difficult to reproduce in humans. In this paper, we present the pre-clinical evaluation of NN1177, a long-acting GLP-1/glucagon receptor co-agonist previously tested in clinical trials. To further investigate the contribution from the respective receptors, two other co-agonists (NN1151, NN1359) with different GLP-1-to-glucagon receptor ratios were evaluated in parallel. In the process of characterizing NN1177, species differences and pitfalls in traditional pre-clinical evaluation methods were identified, highlighting the translational challenges in predicting the optimal receptor balance in humans. In diet-induced obese (DIO) mice, NN1177 induced a dose-dependent body weight loss, primarily due to loss of fat mass, and improvement in glucose tolerance. In DIO rats, NN1177 induced a comparable total body weight reduction, which was in contrast mainly caused by loss of lean mass, and glucose tolerance was impaired. Furthermore, despite long half-lives of the three co-agonists, glucose control during steady state was seen to depend on compound exposure at time of evaluation. When evaluated at higher compound exposure, glucose tolerance was similarly improved for all three co-agonists, independent of receptor balance. However, at lower compound exposure, glucose tolerance was gradually impaired with higher glucagon receptor preference. In addition, glucose tolerance was found to depend on study duration where the effect of glucagon on glucose control became more evident with time. To conclude, the pharmacodynamic effects at a given GLP-1-to-glucagon ratio differs between species, depends on compound exposure and study length, complicating the identification of an optimally balanced clinical candidate. The present findings could partly explain the low number of clinical successes for this dual agonism.


Subject(s)
Glucagon , Receptors, Glucagon , Animals , Blood Glucose/metabolism , Glucagon/therapeutic use , Glucagon-Like Peptide 1/therapeutic use , Glucagon-Like Peptide-1 Receptor/metabolism , Mice , Mice, Obese , Obesity/metabolism , Rats , Receptors, Glucagon/metabolism , Weight Loss
7.
iScience ; 25(11): 105296, 2022 Nov 18.
Article in English | MEDLINE | ID: mdl-36325048

ABSTRACT

The pancreatic hormone, glucagon, is known to regulate hepatic glucose production, but recent studies suggest that its regulation of hepatic amino metabolism is equally important. Here, we show that chronic glucagon receptor activation with a long-acting glucagon analog increases amino acid catabolism and ureagenesis and causes alpha cell hypoplasia in female mice. Conversely, chronic glucagon receptor inhibition with a glucagon receptor antibody decreases amino acid catabolism and ureagenesis and causes alpha cell hyperplasia and beta cell loss. These effects were associated with the transcriptional regulation of hepatic genes related to amino acid uptake and catabolism and by the non-transcriptional modulation of the rate-limiting ureagenesis enzyme, carbamoyl phosphate synthetase-1. Our results support the importance of glucagon receptor signaling for amino acid homeostasis and pancreatic islet integrity in mice and provide knowledge regarding the long-term consequences of chronic glucagon receptor agonism and antagonism.

8.
Mol Metab ; 66: 101639, 2022 12.
Article in English | MEDLINE | ID: mdl-36400402

ABSTRACT

OBJECTIVE: Treatment with glucagon receptor antagonists (GRAs) reduces blood glucose but causes dyslipidemia and accumulation of fat in the liver. We investigated the acute and chronic effects of glucagon on lipid metabolism in mice. METHODS: Chronic effects of glucagon receptor signaling on lipid metabolism were studied using oral lipid tolerance tests (OLTTs) in overnight fasted glucagon receptor knockout (Gcgr-/-) mice, and in C57Bl/6JRj mice treated with a glucagon receptor antibody (GCGR Ab) or a long-acting glucagon analogue (GCGA) for eight weeks. Following treatment, liver tissue was harvested for RNA-sequencing and triglyceride measurements. Acute effects were studied in C57Bl/6JRj mice treated with a GRA or GCGA 1 h or immediately before OLTTs, respectively. Direct effects of glucagon on hepatic lipolysis were studied using isolated perfused mouse liver preparations. To investigate potential effects of GCGA and GRA on gastric emptying, paracetamol was, in separate experiments, administered immediately before OLTTs. RESULTS: Plasma triglyceride concentrations increased 2-fold in Gcgr-/- mice compared to their wild-type littermates during the OLTT (P = 0.001). Chronic treatment with GCGR Ab increased, whereas GCGA treatment decreased, plasma triglyceride concentrations during OLTTs (P < 0.05). Genes involved in lipid metabolism were upregulated upon GCGR Ab treatment while GCGA treatment had opposite effects. Acute GRA and GCGA treatment, respectively, increased (P = 0.02) and decreased (P = 0.003) plasma triglyceride concentrations during OLTTs. Glucagon stimulated hepatic lipolysis, evident by an increase in free fatty acid concentrations in the effluent from perfused mouse livers. In line with this, GCGR Ab treatment increased, while GCGA treatment decreased, liver triglyceride concentrations. The effects of glucagon appeared independent of changes in gastric emptying of paracetamol. CONCLUSIONS: Glucagon receptor signaling regulates triglyceride metabolism, both chronically and acutely, in mice. These data expand glucagon´s biological role and implicate that intact glucagon signaling is important for lipid metabolism. Glucagon agonism may have beneficial effects on hepatic and peripheral triglyceride metabolism.


Subject(s)
Glucagon , Receptors, Glucagon , Triglycerides , Animals , Mice , Acetaminophen/pharmacology , Glucagon/metabolism , Lipid Metabolism/physiology , Mice, Inbred C57BL , Receptors, Glucagon/metabolism , Triglycerides/blood , Triglycerides/metabolism
9.
Pharmacology ; 88(5-6): 340-8, 2011.
Article in English | MEDLINE | ID: mdl-22134089

ABSTRACT

Glucagon-like peptide-1 (GLP-1) activates the GLP-1 receptor (GLP-1R), which belongs to family B of the G-protein-coupled receptors. We previously identified a selective small molecule ligand, compound 2, that acted as a full agonist and allosteric modulator of GLP-1R. In this study, the structurally related small molecule, compound 3, stimulated cAMP production from GLP-1R, but not from the homologous glucagon receptor (GluR). The receptor selectivity encouraged a chimeric receptor approach to identify domains important for compound 3-mediated activation of GLP-1R. A subsegment of the GLP-1R transmembrane domain containing TM2 to TM5 was sufficient to transfer compound 3 responsiveness to GluR. Therefore, divergent residues in this subsegment of GLP-1R and GluR are responsible for the receptor selectivity of compound 3. Functional analyses of other chimeric receptors suggested that the existence of a helix-helix interface between TM1 and TM7 is important for the compound 3 response. Furthermore, site-directed mutagenesis revealed that a Phe195-Leu substitution in TM2 and a Thr391-Ala substitution in TM7 increased and decreased the efficacy of compound 3 without disturbing the potency or efficacy of GLP-1. Collectively, differential effects of receptor mutations suggest that TM2 and/or TM7 are important for compound 3-mediated activation of GLP-1R.


Subject(s)
Receptors, Glucagon/agonists , Receptors, Glucagon/chemistry , Cyclic AMP/metabolism , Glucagon/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor , HEK293 Cells , Humans , Ligands , Models, Molecular , Peptide Fragments/metabolism , Protein Structure, Secondary , Receptors, Glucagon/metabolism
10.
Article in English | MEDLINE | ID: mdl-31031702

ABSTRACT

The discovery of glucagon-like peptide-1 (GLP-1), an incretin hormone with important effects on glycemic control and body weight regulation, led to efforts to extend its half-life and make it therapeutically effective in people with type 2 diabetes (T2D). The development of short- and then long-acting GLP-1 receptor agonists (GLP-1RAs) followed. Our article charts the discovery and development of the long-acting GLP-1 analogs liraglutide and, subsequently, semaglutide. We examine the chemistry employed in designing liraglutide and semaglutide, the human and non-human studies used to investigate their cellular targets and pharmacological effects, and ongoing investigations into new applications and formulations of these drugs. Reversible binding to albumin was used for the systemic protraction of liraglutide and semaglutide, with optimal fatty acid and linker combinations identified to maximize albumin binding while maintaining GLP-1 receptor (GLP-1R) potency. GLP-1RAs mediate their effects via this receptor, which is expressed in the pancreas, gastrointestinal tract, heart, lungs, kidneys, and brain. GLP-1Rs in the pancreas and brain have been shown to account for the respective improvements in glycemic control and body weight that are evident with liraglutide and semaglutide. Both liraglutide and semaglutide also positively affect cardiovascular (CV) outcomes in individuals with T2D, although the precise mechanism is still being explored. Significant weight loss, through an effect to reduce energy intake, led to the approval of liraglutide (3.0 mg) for the treatment of obesity, an indication currently under investigation with semaglutide. Other ongoing investigations with semaglutide include the treatment of non-alcoholic fatty liver disease (NASH) and its use in an oral formulation for the treatment of T2D. In summary, rational design has led to the development of two long-acting GLP-1 analogs, liraglutide and semaglutide, that have made a vast contribution to the management of T2D in terms of improvements in glycemic control, body weight, blood pressure, lipids, beta-cell function, and CV outcomes. Furthermore, the development of an oral formulation for semaglutide may provide individuals with additional benefits in relation to treatment adherence. In addition to T2D, liraglutide is used in the treatment of obesity, while semaglutide is currently under investigation for use in obesity and NASH.

11.
J Chromatogr A ; 1603: 113-129, 2019 Oct 11.
Article in English | MEDLINE | ID: mdl-31262515

ABSTRACT

A protocol was defined which utilised peptides as probes for the characterisation of reversed phase chromatography peptide separation systems. These peptide probes successfully distinguished between differing stationary phases through the probe's hydrophobic, electrostatic, hydrogen bonding and aromatic interactions with the stationary phase, in addition, to more subtle interactions such as the phase's ability to separate racemic or isomeric probes. The dominating forces responsible for the chromatographic selectivity of peptides appear to be hydrophobic as well as electrostatic and polar in nature. This highlights the need for other types of stationary phase ligands with possibly mixed mode functionalities / electrostatic / polar interactions for peptide separations rather than the hydrophobic ligands which dominate small molecule separations. Selectivity differences are observed between phases, but it appears that it is the accessibility differences between these phases which play a crucial role in peptide separations i.e. accessibility to silanols, the hydrophobic acetonitrile / ligand layer or a thin adsorbed water layer on the silica surface.


Subject(s)
Chromatography, Reverse-Phase/methods , Peptides/isolation & purification , Amino Acid Sequence , Buffers , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Peptides/chemistry , Principal Component Analysis , Static Electricity
12.
Elife ; 72018 05 31.
Article in English | MEDLINE | ID: mdl-29851380

ABSTRACT

Transport of biologically active molecules across tight epithelial barriers is a major challenge preventing therapeutic peptides from oral drug delivery. Here, we identify a set of synthetic glycosphingolipids that harness the endogenous process of intracellular lipid-sorting to enable mucosal absorption of the incretin hormone GLP-1. Peptide cargoes covalently fused to glycosphingolipids with ceramide domains containing C6:0 or smaller fatty acids were transported with 20-100-fold greater efficiency across epithelial barriers in vitro and in vivo. This was explained by structure-function of the ceramide domain in intracellular sorting and by the affinity of the glycosphingolipid species for insertion into and retention in cell membranes. In mice, GLP-1 fused to short-chain glycosphingolipids was rapidly and systemically absorbed after gastric gavage to affect glucose tolerance with serum bioavailability comparable to intraperitoneal injection of GLP-1 alone. This is unprecedented for mucosal absorption of therapeutic peptides, and defines a technology with many other clinical applications.


Subject(s)
Absorption, Physiological , Glycosphingolipids/metabolism , Mucous Membrane/metabolism , Peptides/therapeutic use , Animals , Biological Transport, Active , Blood Glucose/metabolism , Cell Nucleus/metabolism , Ceramides/chemistry , Dogs , Epithelial Cells/metabolism , G(M1) Ganglioside/chemistry , G(M1) Ganglioside/metabolism , Glucagon-Like Peptide 1/metabolism , Madin Darby Canine Kidney Cells , Male , Mice, Inbred C57BL , Oligosaccharides/chemistry , Oligosaccharides/metabolism , Reproducibility of Results , Solutions , Structure-Activity Relationship , Transcytosis
13.
J Med Chem ; 50(1): 113-28, 2007 Jan 11.
Article in English | MEDLINE | ID: mdl-17201415

ABSTRACT

A weak human glucagon receptor antagonist with an IC50 of 7 microM was initially found by screening of libraries originally targeted to mimic the binding of the glucagon-like peptide (GLP-1) hormone to its receptor. Optimization of this hit for binding affinity for the glucagon receptor led to ligands with affinity in the nanomolar range. In addition to receptor binding, optimization efforts were made to stabilize the molecules against fast metabolic turnover. A potent antagonist of the human human glucagon receptor was obtained that had 17% oral availability in rats with a plasma half-life of 90 min. The major metabolites of this lead were identified and used to further optimize this series with respect to pharmacokinetic properties. This final optimization led to a potent glucagon antagonist that was orally available in rats and dogs and was efficacious in lowering blood glucose levels in a diabetic animal model.


Subject(s)
Hypoglycemic Agents/chemical synthesis , Receptors, Glucagon/antagonists & inhibitors , beta-Alanine/analogs & derivatives , beta-Alanine/chemical synthesis , Administration, Oral , Animals , Biological Availability , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Dogs , Humans , Hypoglycemic Agents/pharmacology , Mice , Mice, Obese , Rats , Structure-Activity Relationship , beta-Alanine/pharmacology
14.
Eur J Pharmacol ; 773: 24-31, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26808305

ABSTRACT

The pharmacological potential of Calcitonin gene-related peptide (CGRP) beyond vasodilation is not completely understood and studies are limited by the potent vasodilatory effect and the short half-life of CGRP. In particular, the effects of CGRP on metabolic diseases are not clarified. A peptide analogue of the α form of CGRP (αAnalogue) with prolonged half-life (10.2 ± 0.9h) in rodents was synthesised and used to determine specific metabolic effects in 3 rodent models; normal rats, diet-induced obese rats and the Leptin deficient mouse model (ob/ob mice). The αAnalogue (100 nmol/kg) induced elevated energy expenditure and reduced food intake after single dosing in normal rats. In addition, the αAnalogue increased levels of circulating Glucagon-Like Peptide-1 (GLP-1) by >60% and a specific concentration dependent CGRP-induced GLP-1 secretion was verified in a murine L-cell line. Two weeks treatment of the type 2 diabetic ob/ob mice with the αAnalogue caused reduction in fasting insulin levels (199 ± 36 pM vs 332 ± 68 pM) and a tendency to reduce fasting blood glucose (11.2 ± 1.1mM vs 9.5 ± 0.5mM) and % glycosylated haemoglobin (HbA1c) (5.88 ± 0.17 vs 5.12 ± 0.24), demonstrating a potential anti-diabetic effect. Furthermore, two weeks treatment of diet-induced obese rats with the αAnalogue caused reduction in food intake and a significant decline in body weight (3.6 ± 1.9 gvs. -36 ± 1.1g). We have demonstrated that long-acting CGRP analogues may have a therapeutic potential for the treatment of type 2 diabetes through positive metabolic effects and effect on GLP-1 secretion.


Subject(s)
Calcitonin Gene-Related Peptide/analogs & derivatives , Calcitonin Gene-Related Peptide/pharmacology , Glucagon-Like Peptide 1/metabolism , Animals , Body Weight/drug effects , CHO Cells , Calcitonin Gene-Related Peptide/administration & dosage , Cricetinae , Cricetulus , Eating/drug effects , Energy Metabolism/drug effects , Glucose/metabolism , Homeostasis/drug effects , Mice , Rats
15.
J Med Chem ; 58(18): 7370-80, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26308095

ABSTRACT

Liraglutide is an acylated glucagon-like peptide-1 (GLP-1) analogue that binds to serum albumin in vivo and is approved for once-daily treatment of diabetes as well as obesity. The aim of the present studies was to design a once weekly GLP-1 analogue by increasing albumin affinity and secure full stability against metabolic degradation. The fatty acid moiety and the linking chemistry to GLP-1 were the key features to secure high albumin affinity and GLP-1 receptor (GLP-1R) potency and in obtaining a prolonged exposure and action of the GLP-1 analogue. Semaglutide was selected as the optimal once weekly candidate. Semaglutide has two amino acid substitutions compared to human GLP-1 (Aib(8), Arg(34)) and is derivatized at lysine 26. The GLP-1R affinity of semaglutide (0.38 ± 0.06 nM) was three-fold decreased compared to liraglutide, whereas the albumin affinity was increased. The plasma half-life was 46.1 h in mini-pigs following i.v. administration, and semaglutide has an MRT of 63.6 h after s.c. dosing to mini-pigs. Semaglutide is currently in phase 3 clinical testing.


Subject(s)
Glucagon-Like Peptide 1/analogs & derivatives , Glucagon-Like Peptide 1/chemistry , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptides/chemistry , Administration, Intravenous , Animals , Cell Line , Cricetinae , Crystallography, X-Ray , Glucagon-Like Peptide 1/administration & dosage , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon-Like Peptides/administration & dosage , Glucagon-Like Peptides/pharmacology , Half-Life , Humans , Injections, Subcutaneous , Liraglutide/pharmacology , Male , Mice, Obese , Models, Molecular , Rats, Sprague-Dawley , Structure-Activity Relationship , Swine , Swine, Miniature
16.
J Med Chem ; 45(20): 4443-59, 2002 Sep 26.
Article in English | MEDLINE | ID: mdl-12238924

ABSTRACT

Reversible phosphorylation and dephosphorylation of key proteins on tyrosine residues are important parts of intracellular signaling triggered by hormones and other agents. Recent knock-out studies in mice have identified PTP1B as a potential target for the treatment of diabetes and obesity. As a consequence, a number of academic and industrial groups are aggressively pursuing the development of selective PTP1B inhibitors. In addition, other protein-tyrosine phosphatases (PTPs) appear to be critically involved in major diseases such as cancer and autoimmunity. Given the diversity of PTPs and their potential as drug targets in different diseases, we have taken a broad approach to develop active site-directed selective inhibitors of specific members of this family of enzymes. Using a high throughput screening, we have previously identified 2-(oxalylamino)benzoic acid 3a as a relatively weak but classical competitive inhibitor of several PTPs.(4) On the basis of our early studies, indicating that 3a might be used as a starting point for the synthesis of selective PTP inhibitors, we now present our efforts in expansion of this concept and provide here a number of new chemical scaffolds for the development of inhibitors of different members of the PTP family. Although the core structure of these inhibitors is charged, good oral bioavailability has been observed in rat for some compounds. Furthermore, we have observed enhancement of 2-deoxy-glucose accumulation in C2C12 cells with prodrug analogues.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Protein Tyrosine Phosphatases/antagonists & inhibitors , Pyridines/chemical synthesis , Thiophenes/chemical synthesis , Administration, Oral , Animals , Biological Availability , Cell Line , Crystallography, X-Ray , Deoxyglucose/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Mice , Models, Molecular , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Pyridines/chemistry , Pyridines/pharmacology , Rats , Structure-Activity Relationship , Thiophenes/chemistry , Thiophenes/pharmacology
17.
J Med Chem ; 45(26): 5755-75, 2002 Dec 19.
Article in English | MEDLINE | ID: mdl-12477359

ABSTRACT

Highly potent human glucagon receptor (hGluR) antagonists have been prepared employing both medicinal chemistry and targeted libraries based on modification of the core (proximal) dimethoxyphenyl group, the benzyl ether linkage, as well as the (distal) benzylic aryl group of the lead 2, 3-cyano-4-hydroxybenzoic acid (3,5-dimethoxy-4-isopropylbenzyloxybenzylidene)hydrazide. Electron-rich proximal aryl moieties such as mono- and dimethoxy benzenes, naphthalenes, and indoles were found to be active. The SAR was found to be quite insensitive regarding the linkage to the distal aryl group, since long and short as well as polar and apolar linkers gave highly potent compounds. The presence of a distal aryl group was not crucial for obtaining high binding affinity to the hGluR. In many cases, however, the affinity could be further optimized with substituted distal aryl groups. Representative compounds have been tested for in vitro metabolism, and structure-metabolism relationships are described. These efforts lead to the discovery of 74, NNC 25-2504, 3-cyano-4-hydroxybenzoic acid [1-(2,3,5,6-tetramethylbenzyl)-1H-indol-4-ylmethylene]hydrazide, with low in vitro metabolic turnover. 74 was a highly potent noncompetitive antagonist of the human glucagon receptor (IC(50) = 2.3 nM, K(B) = 760 pM) and of the isolated rat receptor (IC(50) = 430 pM, K(B) = 380 pM). Glucagon-stimulated glucose production from isolated primary rat hepatocytes was inhibited competitively by 74 (K(i) = 14 nM). This compound was orally available in dogs (F(po) = 15%) and was active in a glucagon-challenged rat model of hyperglucagonemia and hyperglycemia.


Subject(s)
Hydrazines/chemical synthesis , Indoles/chemical synthesis , Receptors, Glucagon/antagonists & inhibitors , Administration, Oral , Animals , Biological Availability , Cells, Cultured , Dogs , Glucagon/blood , Glucose/biosynthesis , Hepatocytes/metabolism , Humans , Hydrazines/pharmacokinetics , Hydrazines/pharmacology , Hyperglycemia/metabolism , Indoles/pharmacokinetics , Indoles/pharmacology , Male , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
18.
Angew Chem Int Ed Engl ; 38(10): 1417-1420, 1999 May 17.
Article in English | MEDLINE | ID: mdl-29711594

ABSTRACT

The latest member of the cyclophane family, the macrotricyclic tetrathiafulvalene (TTF) "belt", is now available. A general synthetic strategy for the construction of tetraconnected belt-type TTFs (shown schematically) has been developed, made possible by the use of a TTF with two different protecting groups. In the solid-state structure of one of the three TTF-belts prepared two chloroform molecules reside inside the spacious cavity.

19.
J Med Chem ; 55(5): 1969-77, 2012 Mar 08.
Article in English | MEDLINE | ID: mdl-22335602

ABSTRACT

We report in vitro and in vivo data of new α-melanocyte-stimulating hormone (α-MSH) analogues which are N-terminal modified with a long chain fatty acid derivative. While keeping the pharmacophoric motif (d-Phe-Arg-Trp) fixed, we tried to improve selectivity and physicochemical parameters like solubility and stability of these analogues by replacing amino acids further away from the motif. Receptor specific changes in binding affinity to the melanocortin receptors were observed between the acetyl derivatives and the fatty acid analogues. Furthermore, amino acids at the N-terminal of α-MSH (Ser-Tyr-Ser) not considered to be part of the pharmacophore were found to have an influence on the MC4/MC1 receptor selectivity. While the acetyl analogues have an in vivo effect for around 7 h, the long chain fatty acid analogues have an effect up to 48 h in an acute feeding study in male Sprague-Dawley rats after a single subcutaneous administration.


Subject(s)
Anti-Obesity Agents/chemical synthesis , Receptor, Melanocortin, Type 4/agonists , alpha-MSH/analogs & derivatives , alpha-MSH/chemical synthesis , Animals , Anti-Obesity Agents/pharmacokinetics , Anti-Obesity Agents/pharmacology , Eating/drug effects , Fatty Acids/chemical synthesis , Fatty Acids/pharmacokinetics , Fatty Acids/pharmacology , Injections, Subcutaneous , Male , Rats , Rats, Sprague-Dawley , Solubility , Structure-Activity Relationship , alpha-MSH/pharmacokinetics , alpha-MSH/pharmacology
20.
J Med Chem ; 52(9): 2989-3000, 2009 May 14.
Article in English | MEDLINE | ID: mdl-19385613

ABSTRACT

The aim of the work presented here was to design and synthesize potent human glucagon receptor antagonists with improved pharmacokinetic (PK) properties for development of pharmaceuticals for the treatment of type 2 diabetes. We describe the preparation of compounds with cyclic cores (5-aminothiazoles), their binding affinities for the human glucagon and GIP receptors, as well as affinities for rat, mouse, pig, dog, and monkey glucagon receptors. Generally, the compounds had slightly less glucagon receptor affinity compared to compounds of the previous series, but this was compensated for by much improved PK profiles in both rats and dogs with high oral bioavailabilities and sustained high plasma exposures. The compounds generally showed species selectivity for glucagon receptor binding with poor affinities for the rat, mouse, rabbit, and pig receptors. However, dog and monkey glucagon receptor affinities seem to reflect the human situation. One compound of this series, 18, was tested intravenously in an anesthetized glucagon-challenged monkey model of hyperglucagonaemia and hyperglycaemia and was shown dose-dependently to decrease glycaemia. Further, high plasma exposures and a long plasma half-life (5.2 h) were obtained.


Subject(s)
Receptors, Glucagon/antagonists & inhibitors , Thiazoles/pharmacology , Thiazoles/pharmacokinetics , Administration, Oral , Animals , Cell Line , Diabetes Mellitus, Type 2/drug therapy , Drug Design , Half-Life , Humans , Receptors, Glucagon/metabolism , Species Specificity , Thiazoles/chemistry , Thiazoles/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL