Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Nat Metab ; 6(2): 290-303, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38316982

ABSTRACT

Obesity is a major public health crisis. Multi-specific peptides have emerged as promising therapeutic strategies for clinical weight loss. Glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are endogenous incretins that regulate weight through their receptors (R). AMG 133 (maridebart cafraglutide) is a bispecific molecule engineered by conjugating a fully human monoclonal anti-human GIPR antagonist antibody to two GLP-1 analogue agonist peptides using amino acid linkers. Here, we confirm the GIPR antagonist and GLP-1R agonist activities in cell-based systems and report the ability of AMG 133 to reduce body weight and improve metabolic markers in male obese mice and cynomolgus monkeys. In a phase 1, randomized, double-blind, placebo-controlled clinical study in participants with obesity ( NCT04478708 ), AMG 133 had an acceptable safety and tolerability profile along with pronounced dose-dependent weight loss. In the multiple ascending dose cohorts, weight loss was maintained for up to 150 days after the last dose. These findings support continued clinical evaluation of AMG 133.


Subject(s)
Glucagon-Like Peptide 1 , Glucagon-Like Peptide-1 Receptor Agonists , Mice , Animals , Humans , Male , Glucagon-Like Peptide 1/therapeutic use , Weight Loss , Peptides/therapeutic use , Obesity/drug therapy , Obesity/metabolism
2.
Cell Rep Med ; 2(5): 100263, 2021 05 18.
Article in English | MEDLINE | ID: mdl-34095876

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) regulate glucose and energy homeostasis. Targeting both pathways with GIP receptor (GIPR) antagonist antibody (GIPR-Ab) and GLP-1 receptor (GLP-1R) agonist, by generating GIPR-Ab/GLP-1 bispecific molecules, is an approach for treating obesity and its comorbidities. In mice and monkeys, these molecules reduce body weight (BW) and improve many metabolic parameters. BW loss is greater with GIPR-Ab/GLP-1 than with GIPR-Ab or a control antibody conjugate, suggesting synergistic effects. GIPR-Ab/GLP-1 also reduces the respiratory exchange ratio in DIO mice. Simultaneous receptor binding and rapid receptor internalization by GIPR-Ab/GLP-1 amplify endosomal cAMP production in recombinant cells expressing both receptors. This may explain the efficacy of the bispecific molecules. Overall, our GIPR-Ab/GLP-1 molecules promote BW loss, and they may be used for treating obesity.


Subject(s)
Body Weight/physiology , Glucagon-Like Peptide 1/metabolism , Obesity/metabolism , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Animals , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide-1 Receptor/metabolism , Glucose Tolerance Test/methods , Haplorhini/metabolism , Mice, Obese
3.
Sci Transl Med ; 10(472)2018 12 19.
Article in English | MEDLINE | ID: mdl-30567927

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) receptor (GIPR) has been identified in multiple genome-wide association studies (GWAS) as a contributor to obesity, and GIPR knockout mice are protected against diet-induced obesity (DIO). On the basis of this genetic evidence, we developed anti-GIPR antagonistic antibodies as a potential therapeutic strategy for the treatment of obesity and observed that a mouse anti-murine GIPR antibody (muGIPR-Ab) protected against body weight gain, improved multiple metabolic parameters, and was associated with reduced food intake and resting respiratory exchange ratio (RER) in DIO mice. We replicated these results in obese nonhuman primates (NHPs) using an anti-human GIPR antibody (hGIPR-Ab) and found that weight loss was more pronounced than in mice. In addition, we observed enhanced weight loss in DIO mice and NHPs when anti-GIPR antibodies were codosed with glucagon-like peptide-1 receptor (GLP-1R) agonists. Mechanistic and crystallographic studies demonstrated that hGIPR-Ab displaced GIP and bound to GIPR using the same conserved hydrophobic residues as GIP. Further, using a conditional knockout mouse model, we excluded the role of GIPR in pancreatic ß-cells in the regulation of body weight and response to GIPR antagonism. In conclusion, these data provide preclinical validation of a therapeutic approach to treat obesity with anti-GIPR antibodies.


Subject(s)
Glucagon-Like Peptide-1 Receptor/agonists , Obesity/drug therapy , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Adipocytes/metabolism , Animals , Antibodies/pharmacology , Antibodies/therapeutic use , Diet , Drug Therapy, Combination , Feeding Behavior , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon-Like Peptides/analogs & derivatives , Glucagon-Like Peptides/pharmacology , Glucagon-Like Peptides/therapeutic use , Humans , Immunoglobulin Fc Fragments/pharmacology , Immunoglobulin Fc Fragments/therapeutic use , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Liraglutide/pharmacology , Liraglutide/therapeutic use , Mice, Obese , Obesity/pathology , Primates , Receptors, Gastrointestinal Hormone/metabolism , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Respiration , Weight Gain/drug effects , Weight Loss/drug effects
4.
Sci Transl Med ; 9(412)2017 Oct 18.
Article in English | MEDLINE | ID: mdl-29046435

ABSTRACT

In search of metabolically regulated secreted proteins, we conducted a microarray study comparing gene expression in major metabolic tissues of fed and fasted ob/ob mice and C57BL/6 mice. The array used in this study included probes for ~4000 genes annotated as potential secreted proteins. Circulating macrophage inhibitory cytokine 1 (MIC-1)/growth differentiation factor 15 (GDF15) concentrations were increased in obese mice, rats, and humans in comparison to age-matched lean controls. Adeno-associated virus-mediated overexpression of GDF15 and recombinant GDF15 treatments reduced food intake and body weight and improved metabolic profiles in various metabolic disease models in mice, rats, and obese cynomolgus monkeys. Analysis of the GDF15 crystal structure suggested that the protein is not suitable for conventional Fc fusion at the carboxyl terminus of the protein. Thus, we used a structure-guided approach to design and successfully generate several Fc fusion molecules with extended half-life and potent efficacy. Furthermore, we discovered that GDF15 delayed gastric emptying, changed food preference, and activated area postrema neurons, confirming a role for GDF15 in the gut-brain axis responsible for the regulation of body energy intake. Our work provides evidence that GDF15 Fc fusion proteins could be potential therapeutic agents for the treatment of obesity and related comorbidities.


Subject(s)
Growth Differentiation Factor 15/therapeutic use , Obesity/drug therapy , Animals , Crystallography, X-Ray , Dependovirus/metabolism , Diet , Food Preferences , Gastric Emptying , Growth Differentiation Factor 15/chemistry , Humans , Macaca fascicularis , Male , Mice, Inbred C57BL , Mice, Obese , Neurons/physiology , Obesity/pathology , Rats, Sprague-Dawley , Receptors, Fc/metabolism , Recombinant Fusion Proteins/metabolism , Up-Regulation
5.
ACS Med Chem Lett ; 7(7): 666-70, 2016 Jul 14.
Article in English | MEDLINE | ID: mdl-27437074

ABSTRACT

Two 1-(4-aryl-5-alkyl-pyridin-2-yl)-3-methylurea glucokinase activators were identified with robust in vivo efficacy. These two compounds possessed higher solubilities than the previously identified triaryl compounds (i.e., AM-2394). Structure-activity relationship studies are presented along with relevant pharmacokinetic and in vivo data.

6.
ACS Med Chem Lett ; 7(7): 714-8, 2016 Jul 14.
Article in English | MEDLINE | ID: mdl-27437083

ABSTRACT

Glucokinase (GK) catalyzes the phosphorylation of glucose to glucose-6-phosphate. We present the structure-activity relationships leading to the discovery of AM-2394, a structurally distinct GKA. AM-2394 activates GK with an EC50 of 60 nM, increases the affinity of GK for glucose by approximately 10-fold, exhibits moderate clearance and good oral bioavailability in multiple animal models, and lowers glucose excursion following an oral glucose tolerance test in an ob/ob mouse model of diabetes.

7.
Cell Metab ; 21(5): 731-8, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25955208

ABSTRACT

"Browning," the appearance and activation of brown-in-white (brite) adipose cells within inguinal white adipose tissue (iWAT), and induction of uncoupling protein 1 (UCP1) correlate with fibroblast growth factor-21 (FGF21)-induced weight loss and glucose homeostasis improvements. Therefore, antiobesity therapies targeting browning and brite adipocyte activation are currently being sought. To test the dependence of weight loss on browning, we examined whether this event was responsible for FGF21-Fc's beneficial effects. Lean and diet-induced obese mice housed at 21°C or 30°C that received FGF21-Fc exhibited similar degrees of body weight reduction and glucose homeostasis improvement. Substantial browning of iWAT occurred only in FGF21-Fc-treated lean mice housed at 21°C. Further, FGF21-Fc-treated Ucp1(-/-) mice showed robust improvements in body weight, glucose homeostasis, and plasma lipids, associated with increased energy expenditure and FGF21-Fc-induced Ppargc1 expression in iWAT. We conclude that FGF21 requires neither UCP1 nor brite adipocytes to elicit weight loss and improve glucose homeostasis.


Subject(s)
Adipose Tissue, White/drug effects , Anti-Obesity Agents/therapeutic use , Fibroblast Growth Factors/therapeutic use , Obesity/drug therapy , Adipocytes, Brown/drug effects , Adipocytes, Brown/pathology , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/physiopathology , Adipose Tissue, White/physiopathology , Animals , Diet/adverse effects , Energy Metabolism/drug effects , Gene Expression Regulation/drug effects , Glucose/metabolism , Hypoglycemic Agents/therapeutic use , Ion Channels/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Mitochondrial Proteins/genetics , Obesity/etiology , Obesity/genetics , Obesity/physiopathology , Thermogenesis/drug effects , Uncoupling Protein 1
8.
J Med Chem ; 56(24): 10132-41, 2013 Dec 27.
Article in English | MEDLINE | ID: mdl-24294923

ABSTRACT

Acetyl-CoA carboxylase (ACC) is a target of interest for the treatment of metabolic syndrome. Starting from a biphenyloxadiazole screening hit, a series of piperazine oxadiazole ACC inhibitors was developed. Initial pharmacokinetic liabilities of the piperazine oxadiazoles were overcome by blocking predicted sites of metabolism, resulting in compounds with suitable properties for further in vivo studies. Compound 26 was shown to inhibit malonyl-CoA production in an in vivo pharmacodynamic assay and was advanced to a long-term efficacy study. Prolonged dosing with compound 26 resulted in impaired glucose tolerance in diet-induced obese (DIO) C57BL6 mice, an unexpected finding.


Subject(s)
Acetyl-CoA Carboxylase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Oxadiazoles/pharmacology , Piperazines/pharmacology , Acetyl-CoA Carboxylase/metabolism , Animals , Diet/adverse effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Molecular Structure , Oxadiazoles/chemical synthesis , Oxadiazoles/chemistry , Piperazines/chemical synthesis , Piperazines/chemistry , Structure-Activity Relationship
9.
Nature ; 504(7480): 437-40, 2013 Dec 19.
Article in English | MEDLINE | ID: mdl-24226772

ABSTRACT

Glucose homeostasis is a vital and complex process, and its disruption can cause hyperglycaemia and type II diabetes mellitus. Glucokinase (GK), a key enzyme that regulates glucose homeostasis, converts glucose to glucose-6-phosphate in pancreatic ß-cells, liver hepatocytes, specific hypothalamic neurons, and gut enterocytes. In hepatocytes, GK regulates glucose uptake and glycogen synthesis, suppresses glucose production, and is subject to the endogenous inhibitor GK regulatory protein (GKRP). During fasting, GKRP binds, inactivates and sequesters GK in the nucleus, which removes GK from the gluconeogenic process and prevents a futile cycle of glucose phosphorylation. Compounds that directly hyperactivate GK (GK activators) lower blood glucose levels and are being evaluated clinically as potential therapeutics for the treatment of type II diabetes mellitus. However, initial reports indicate that an increased risk of hypoglycaemia is associated with some GK activators. To mitigate the risk of hypoglycaemia, we sought to increase GK activity by blocking GKRP. Here we describe the identification of two potent small-molecule GK-GKRP disruptors (AMG-1694 and AMG-3969) that normalized blood glucose levels in several rodent models of diabetes. These compounds potently reversed the inhibitory effect of GKRP on GK activity and promoted GK translocation both in vitro (isolated hepatocytes) and in vivo (liver). A co-crystal structure of full-length human GKRP in complex with AMG-1694 revealed a previously unknown binding pocket in GKRP distinct from that of the phosphofructose-binding site. Furthermore, with AMG-1694 and AMG-3969 (but not GK activators), blood glucose lowering was restricted to diabetic and not normoglycaemic animals. These findings exploit a new cellular mechanism for lowering blood glucose levels with reduced potential for hypoglycaemic risk in patients with type II diabetes mellitus.


Subject(s)
Carrier Proteins/antagonists & inhibitors , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Adaptor Proteins, Signal Transducing , Animals , Blood Glucose/metabolism , Carrier Proteins/metabolism , Cell Nucleus/enzymology , Crystallography, X-Ray , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/enzymology , Disease Models, Animal , Hepatocytes , Humans , Hyperglycemia/blood , Hyperglycemia/drug therapy , Hyperglycemia/enzymology , Hypoglycemic Agents/chemistry , Liver/cytology , Liver/enzymology , Liver/metabolism , Male , Models, Molecular , Organ Specificity , Phosphorylation/drug effects , Piperazines/chemistry , Piperazines/metabolism , Piperazines/pharmacology , Piperazines/therapeutic use , Protein Binding/drug effects , Protein Transport/drug effects , Rats , Rats, Wistar , Sulfonamides/chemistry , Sulfonamides/metabolism , Sulfonamides/pharmacology , Sulfonamides/therapeutic use
10.
PLoS One ; 8(4): e61432, 2013.
Article in English | MEDLINE | ID: mdl-23630589

ABSTRACT

The endocrine hormone FGF21 has attracted considerable interest as a potential therapeutic for treating diabetes and obesity. As an alternative to the native cytokine, we generated bispecific Avimer polypeptides that bind with high affinity and specificity to one of the receptor and coreceptor pairs used by FGF21, FGFR1c and ß-Klotho. These Avimers exhibit FGF21-like activity in in vitro assays with potency greater than FGF21. In a study conducted in obese male cynomolgus monkeys, animals treated with an FGFR1c/ß-Klotho bispecific Avimer showed improved metabolic parameters and reduced body weight comparable to the effects seen with FGF21. These results not only demonstrate the essential roles of FGFR1c and ß-Klotho in mediating the metabolic effects of FGF21, they also describe a first bispecific activator of this unique receptor complex and provide validation for a novel therapeutic approach to target this potentially important pathway for treating diabetes and obesity.


Subject(s)
Anti-Obesity Agents/pharmacology , Fibroblast Growth Factors/physiology , Membrane Proteins/antagonists & inhibitors , Obesity/drug therapy , Peptides/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Amino Acid Sequence , Animals , Anti-Obesity Agents/pharmacokinetics , Binding Sites , Binding, Competitive , Body Weight/drug effects , Cell Line , Drug Evaluation, Preclinical , Fibroblast Growth Factors/chemistry , Insulin/blood , Klotho Proteins , Macaca fascicularis , Male , Membrane Proteins/biosynthesis , Mice , Molecular Mimicry , Molecular Sequence Data , Obesity/blood , Peptides/pharmacokinetics , Protein Binding , Rats , Receptor, Fibroblast Growth Factor, Type 4/chemistry , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/biosynthesis , Serum Albumin/pharmacokinetics , Serum Albumin/pharmacology , Signal Transduction , Triglycerides/blood
11.
Sci Transl Med ; 4(162): 162ra153, 2012 Nov 28.
Article in English | MEDLINE | ID: mdl-23197570

ABSTRACT

Fibroblast growth factor 21 (FGF21) is a distinctive member of the FGF family with potent beneficial effects on lipid, body weight, and glucose metabolism and has attracted considerable interest as a potential therapeutic for treating diabetes and obesity. As an alternative to native FGF21, we have developed a monoclonal antibody, mimAb1, that binds to ßKlotho with high affinity and specifically activates signaling from the ßKlotho/FGFR1c (FGF receptor 1c) receptor complex. In obese cynomolgus monkeys, injection of mimAb1 led to FGF21-like metabolic effects, including decreases in body weight, plasma insulin, triglycerides, and glucose during tolerance testing. Mice with adipose-selective FGFR1 knockout were refractory to FGF21-induced improvements in glucose metabolism and body weight. These results in obese monkeys (with mimAb1) and in FGFR1 knockout mice (with FGF21) demonstrated the essential role of FGFR1c in FGF21 function and suggest fat as a critical target tissue for the cytokine and antibody. Because mimAb1 depends on ßKlotho to activate FGFR1c, it is not expected to induce side effects caused by activating FGFR1c alone. The unexpected finding of an antibody that can activate FGF21-like signaling through cell surface receptors provided preclinical validation for an innovative therapeutic approach to diabetes and obesity.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Diabetes Mellitus/drug therapy , Fibroblast Growth Factors/immunology , Glucuronidase/metabolism , Obesity/drug therapy , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Antibodies, Monoclonal/pharmacology , Body Weight/genetics , Diabetes Mellitus/blood , Epitopes/chemistry , Glucose/metabolism , Glucuronidase/immunology , Humans , Klotho Proteins , Macaca fascicularis , Mice , Obesity/blood , Obesity/complications , Phosphates/blood , Receptor, Fibroblast Growth Factor, Type 1/agonists , Signal Transduction/drug effects , Triglycerides/blood
12.
Endocrinology ; 153(9): 4192-203, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22798348

ABSTRACT

Fibroblast growth factor 21 (FGF21), a hormone with short half-life, has consistently shown strong pharmacological efficacy. We first assessed the efficacy of murine recombinant FGF21 in C57BL6 lean mice for 5 wk. We then generated a long-acting FGF21 molecule by fusing a Fc to a variant of human recombinant FGF21 (hrFGF21) that contained two engineered mutations [L98R, P171G; Fc-FGF21(RG)] and tested it in C57BL6 diet-induced obese mice and obese rhesus monkeys. We compared its metabolic properties with those of the hrFGF21. Groups of diet-induced obese mice were treated for 36 d with different doses of hrFGF21 (01, 0.3, and 1 mg/kg twice daily) and with Fc-FGF21(RG) (2.3 mg/kg, every 5 d). Body weight, glucose, insulin, cholesterol, and triglyceride levels were decreased after treatment with either compound. A glucose tolerance test (GTT) was also improved. Obese rhesus monkeys were treated with hrFGF21 (once a day) and Fc-FGF21(RG) (once a week) in a dose-escalation fashion. Doses started at 0.1 and 0.3 mg/kg and ended at 3 and 5 mg/kg for hrFGF21 and Fc-FGF21(RG), respectively. Doses were escalated every 2 wk, and animals were followed up for a washout period of 3 wk. Body weight, glucose, insulin, cholesterol, and triglyceride levels and the GTT profile were decreased to a greater extent with Fc-FGF21(RG) than with hrFGF21. The PK-PD relationship of Fc-FGF21(RG) exposure and triglyceride reduction was also conducted with a maximum response model. In conclusion, in more than one species, Fc-FGF21(RG) chronically administered once a week showed similar or greater efficacy than hrFGF21 administered daily.


Subject(s)
Fibroblast Growth Factors/administration & dosage , Fibroblast Growth Factors/therapeutic use , Obesity/drug therapy , Animals , Blood Glucose/drug effects , Body Weight , Drug Administration Schedule , Glucose Tolerance Test , Macaca mulatta , Mice , Mice, Inbred C57BL , Triglycerides/blood
13.
J Lipid Res ; 53(4): 643-52, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22287724

ABSTRACT

The aim of this study is to investigate the capability of an apoA-I mimetic with multiple amphipathic helices to form HDL-like particles in vitro and in vivo. To generate multivalent helices and to track the peptide mimetic, we have constructed a peptibody by fusing two tandem repeats of 4F peptide to the C terminus of a murine IgG Fc fragment. The resultant peptidbody, mFc-2X4F, dose-dependently promoted cholesterol efflux in vitro, and the efflux potency was superior to monomeric 4F peptide. Like apoA-I, mFc-2X4F stabilized ABCA1 in J774A.1 and THP1 cells. The peptibody formed larger HDL particles when incubated with cultured cells compared with those by apoA-I. Interestingly, when administered to mice, mFc-2X4F increased both pre-ß and α-1 HDL subfractions. The lipid-bound mFc-2X4F was mostly in the α-1 migrating subfraction. Most importantly, mFc-2X4F and apoA-I were found to coexist in the same HDL particles formed in vivo. These data suggest that the apoA-I mimetic peptibody is capable of mimicking apoA-I to generate HDL particles. The peptibody and apoA-I may work cooperatively to generate larger HDL particles in vivo, either at the cholesterol efflux stage and/or via fusion of HDL particles that were generated by the peptibody and apoA-I individually.


Subject(s)
Apolipoprotein A-I/pharmacology , Peptides/pharmacology , Recombinant Fusion Proteins/chemistry , 3T3 Cells , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/chemistry , Amino Acid Sequence , Animals , Apolipoprotein A-I/chemistry , Cholesterol/blood , Dose-Response Relationship, Drug , HEK293 Cells , Hep G2 Cells , High-Density Lipoproteins, Pre-beta/chemistry , Humans , Immunoglobulin Fc Fragments/chemistry , Lipoproteins, HDL/blood , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Peptides/administration & dosage , Peptides/chemistry , Protein Stability , Protein Structure, Secondary , Recombinant Fusion Proteins/pharmacology , Tandem Repeat Sequences
14.
J Pharmacol Exp Ther ; 338(1): 70-81, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21471191

ABSTRACT

Pharmacologic contributions of directly agonizing glucagon-like peptide 1 (GLP-1) receptor or antagonizing glucagon receptor (GCGR) on energy state and glucose homeostasis were assessed in diet-induced obese (DIO) mice. Metabolic rate and respiratory quotient (RQ), hyperglycemic clamp, stable isotope-based dynamic metabolic profiling (SiDMAP) studies of (13)C-labeled glucose during glucose tolerance test (GTT) and gene expression were assessed in cohorts of DIO mice after a single administration of GLP-1 analog [GLP-1-(23)] or anti-GCGR antibody (Ab). GLP-1-(23) and GCGR Ab similarly improved GTT. GLP-1-(23) decreased food intake and body weight trended lower. GCGR Ab modestly decreased food intake without significant effect on body weight. GLP-1-(23) and GCGR Ab decreased RQ with GLP-1, causing a greater effect. In a hyperglycemic clamp, GLP-1-(23) reduced hepatic glucose production (HGP), increased glucose infusion rate (GIR), increased glucose uptake in brown adipose tissue, and increased whole-body glucose turnover, glycolysis, and rate of glycogen synthesis. GCGR Ab slightly decreased HGP, increased GIR, and increased glucose uptake in the heart. SiDMAP showed that GLP-1-(23) and GCGR Ab increased (13)C lactate labeling from glucose, indicating that liver, muscle, and other organs were involved in the rapid disposal of glucose from plasma. GCGR Ab and GLP-1-(23) caused different changes in mRNA expression levels of glucose- and lipid metabolism-associated genes. The effect of GLP-1-(23) on energy state and glucose homeostasis was greater than GCGR Ab. Although GCGR antagonism is associated with increased circulating levels of GLP-1, most GLP-1-(23)-associated pharmacologic effects are more pronounced than GCGR Ab.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Blood Glucose/drug effects , Energy Metabolism/drug effects , Glucagon-Like Peptide 1/analogs & derivatives , Homeostasis/drug effects , Obesity/metabolism , Receptors, Glucagon/antagonists & inhibitors , Animals , Blood Glucose/physiology , Body Weight/drug effects , Body Weight/physiology , Dietary Fats/administration & dosage , Drug Delivery Systems/methods , Energy Metabolism/physiology , Glucagon-Like Peptide 1/administration & dosage , Glucagon-Like Peptide 1/physiology , Glucagon-Like Peptide-1 Receptor , Homeostasis/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Obesity/drug therapy , Receptors, Glucagon/physiology
15.
ACS Med Chem Lett ; 2(11): 824-7, 2011 Nov 10.
Article in English | MEDLINE | ID: mdl-24900270

ABSTRACT

All eight of the major active metabolites of (S)-2-((1S,2S,4R)-bicyclo[2.2.1]heptan-2-ylamino)-5-isopropyl-5-methylthiazol-4(5H)-one (AMG 221, compound 1), an inhibitor of 11ß-hydroxysteroid dehydrogenase type 1 that has entered the clinic for the treatment of type 2 diabetes, were synthetically prepared and confirmed by comparison with samples generated in liver microsomes. After further profiling, we determined that metabolite 2 was equipotent to 1 on human 11ß-HSD1 and had lower in vivo clearance and higher bioavailability in rat and mouse. Compound 2 was advanced into a pharmacodynamic model in mouse where it inhibited adipose 11ß-HSD1 activity.

16.
Am J Physiol Endocrinol Metab ; 299(4): E624-32, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20647556

ABSTRACT

Antagonism of the glucagon receptor (GCGR) is associated with increased circulating levels of glucagon-like peptide-1 (GLP-1). To investigate the contribution of GLP-1 to the antidiabetic actions of GCGR antagonism, we administered an anti-GCGR monoclonal antibody (mAb B) to wild-type mice and GLP-1 receptor knockout (GLP-1R KO) mice. Treatment of wild-type mice with mAb B lowered fasting blood glucose, improved glucose tolerance, and enhanced glucose-stimulated insulin secretion during an intraperitoneal glucose tolerance test (ipGTT). In contrast, treatment of GLP-1R KO mice with mAb B had little efficacy during an ipGTT. Furthermore, pretreatment with the GLP-1R antagonist exendin-(9-39) diminished the antihyperglycemic effects of mAb B in wild-type mice. To determine the mechanism whereby mAb B improves glucose tolerance, we generated a monoclonal antibody that specifically antagonizes the human GLP-1R. Using a human islet transplanted mouse model, we demonstrated that pancreatic islet GLP-1R signaling is required for the full efficacy of the GCGR antagonist. To identify the source of the elevated GLP-1 observed in GCGR mAb-treated mice, we measured active GLP-1 content in pancreas and intestine from db/db mice treated with anti-GCGR mAb for 8 wk. Elevated GLP-1 in GCGR mAb-treated mice was predominantly derived from increased pancreatic GLP-1 synthesis and processing. All together, these data show that pancreatic GLP-1 is a significant contributor to the glucose-lowering effects observed in response to GCGR antagonist treatment.


Subject(s)
Glucagon-Like Peptide 1/physiology , Glucagon/physiology , Islets of Langerhans/physiology , Receptors, Glucagon/antagonists & inhibitors , Animals , Antibodies, Monoclonal/pharmacology , Disease Models, Animal , Female , Glucagon/blood , Glucagon-Like Peptide-1 Receptor , Glucose Tolerance Test , Islets of Langerhans/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Peptide Fragments/pharmacology , Receptors, Glucagon/blood , Receptors, Glucagon/physiology , Signal Transduction/drug effects
17.
J Med Chem ; 53(11): 4481-7, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20465278

ABSTRACT

Thiazolones with an exo-norbornylamine at the 2-position and an isopropyl group on the 5-position are potent 11beta-HSD1 inhibitors. However, the C-5 center was prone to epimerization in vitro and in vivo, forming a less potent diastereomer. A methyl group was added to the C-5 position to eliminate epimerization, leading to the discovery of (S)-2-((1S,2S,4R)-bicyclo[2.2.1]heptan-2-ylamino)-5-isopropyl-5-methylthiazol-4(5H)-one (AMG 221). This compound decreased fed blood glucose and insulin levels and reduced body weight in diet-induced obesity mice.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , Drug Discovery/methods , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Thiazoles/administration & dosage , Thiazoles/pharmacology , 11-beta-Hydroxysteroid Dehydrogenase Type 1/chemistry , Administration, Oral , Animals , Dogs , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Humans , Male , Mice , Models, Molecular , Protein Conformation , Rats , Thiazoles/chemistry , Thiazoles/pharmacokinetics
18.
AAPS J ; 11(4): 700-9, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19851873

ABSTRACT

Elevated basal concentrations of glucagon and reduced postprandial glucagon suppression are partly responsible for the increased hepatic glucose production seen in type 2 diabetic patients. Recently, it was demonstrated that an antagonistic human monoclonal antibody (mAb) blocking glucagon receptor (GCGR) has profound glucose-lowering effects in various animal models. To further understand the effects on glucose homeostasis mediated by such an antibody, a pharmacokinetic-pharmacodynamic (PK-PD) study was conducted in a diabetic ob/ob mouse model. Four groups of ob/ob mice were randomized to receive single intraperitoneal administration of placebo, 0.6, 1, or 3 mg/kg of mAb GCGR, a fully human mAb against GCGR. The concentration-time data were used for noncompartmental and compartmental analysis. A semi-mechanistic PK-PD model incorporating the glucose-glucagon inter-regulation and the hypothesized inhibitory effect of mAb GCGR on GCGR signaling pathway via competitive inhibition was included to describe the disposition of glucose and glucagon over time. The pharmacokinetics of mAb GCGR was well characterized by a two-compartment model with parallel linear and nonlinear saturable eliminations. Single injection of mAb GCGR caused a rapid glucose-lowering effect with blood glucose concentrations returning to baseline by 4 to 18 days with increasing dose from 0.6 to 3 mg/kg. Elevation of glucagon concentrations was also observed in a dose-dependent manner. The results illustrated that the feedback relationship between glucose and glucagon in the presence of mAb GCGR could be quantitatively described by the developed model. The model may provide additional understanding in the underlying mechanism of GCGR antagonism by mAb.


Subject(s)
Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/pharmacokinetics , Receptors, Glucagon/antagonists & inhibitors , Algorithms , Animals , Blood Glucose/metabolism , CHO Cells , Cricetinae , Cricetulus , Diabetes Mellitus, Type 2/drug therapy , Glucagon/blood , Humans , Immunoglobulin G/immunology , Male , Mice , Mice, Obese , Models, Statistical , Postprandial Period
19.
J Pharmacol Exp Ther ; 331(3): 871-81, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19720878

ABSTRACT

Uncontrolled hepatic glucose output (HGO) contributes significantly to the pathological hyperglycemic state of patients with type 2 diabetes. Glucagon, through action on its receptor, stimulates HGO, thereby leading to increased glycemia. Antagonizing the glucagon signaling pathway represents an attractive therapeutic approach for the treatment of type 2 diabetes. We previously reported the generation and characterization of several high-affinity monoclonal antibodies (mAbs) targeting the glucagon receptor (GCGR). In the present study, we demonstrate that a 5-week treatment of diet-induced obese mice with mAb effectively normalized nonfasting blood glucose. Similar treatment also reduced fasting blood glucose without inducing hypoglycemia or other undesirable metabolic perturbations. In addition, no hypoglycemia was found in db/db mice that were treated with a combination of insulin and mAb. Long-term treatment with the mAb caused dose-dependent hyperglucagonemia and minimal to mild alpha-cell hyperplasia in lean mice. There was no evidence of pancreatic alpha-cell neoplastic transformation in mice treated with mAb for as long as 18 weeks. Treatment-induced hyperglucagonemia and alpha-cell hyperplasia were reversible after treatment withdrawal for periods of 4 and 10 weeks, respectively. It is noteworthy that pancreatic beta-cell function was preserved, as demonstrated by improved glucose tolerance throughout the 18-week treatment period. Our studies further support the concept that long-term inhibition of GCGR signaling by a mAb could be an effective approach for controlling diabetic hyperglycemia.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Glucagon-Secreting Cells/pathology , Glucagon/blood , Glucose/metabolism , Hyperglycemia/drug therapy , Hypoglycemic Agents/therapeutic use , Receptors, Glucagon/antagonists & inhibitors , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Blood Glucose , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Hyperglycemia/blood , Hyperglycemia/metabolism , Hyperplasia , Hypoglycemia/blood , Hypoglycemia/metabolism , Hypoglycemia/prevention & control , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/adverse effects , Insulin/administration & dosage , Insulin/therapeutic use , Male , Mice , Mice, Inbred C57BL , Obesity/blood , Obesity/metabolism , Tachyphylaxis
20.
J Pharmacol Exp Ther ; 329(1): 102-11, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19129372

ABSTRACT

Antagonizing the glucagon signaling pathway represents an attractive therapeutic approach for reducing excess hepatic glucose production in patients with type 2 diabetes. Despite extensive efforts, there is currently no human therapeutic that directly inhibits the glucagon/glucagon receptor pathway. We undertook a novel approach by generating high-affinity human monoclonal antibodies (mAbs) to the human glucagon receptor (GCGR) that display potent antagonistic activity in vitro and in vivo. A single injection of a lead antibody, mAb B, at 3 mg/kg, normalized blood glucose levels in ob/ob mice for 8 days. In addition, a single injection of mAb B dose-dependently lowered fasting blood glucose levels without inducing hypoglycemia and improved glucose tolerance in normal C57BL/6 mice. In normal cynomolgus monkeys, a single injection improved glucose tolerance while increasing glucagon and active glucagon-like peptide-1 levels. Thus, the anti-GCGR mAb could represent an effective new therapeutic for the treatment of type 2 diabetes.


Subject(s)
Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , Glucose/metabolism , Homeostasis/drug effects , Receptors, Glucagon/antagonists & inhibitors , Animals , Blood Glucose/metabolism , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Endocytosis/drug effects , Flow Cytometry , Glucose Tolerance Test , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Kinetics , Ligands , Macaca fascicularis , Male , Mice , Mice, Inbred C57BL , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...