Size | Price | |
---|---|---|
500mg | ||
1g | ||
Other Sizes |
Targets |
Glucagon Receptor
|
---|---|
ln Vitro |
Adomeglivant cannot block the cAMP-increasing effect of pancreatic islets [2]. Adomeglivant exhibits high resistance to group B GPCRs and resonates with the oscillatory binding motif potential in GluR, GLP-1R and GIP-R [2].
Glucagon and LY2409021 both target glucagon and GLP-1 receptors. LY2409021 blocks GLP-1 and Ex-4 agonist action at the GLP-1R. LY2409021 is a GIP-R antagonist but fails to block adenosine action at A2B receptors. LY2409021 blocks GGP817 agonist action at the GluR and GLP-1R. The GluR allosteric inhibitors LY2409021 and MK 0893 antagonized glucagon and GLP-1 action at the GLP-1R, whereas des-His1-[Glu9]glucagon antagonized glucagon action at the GluR, while having minimal inhibitory action versus glucagon or GLP-1 at the GLP-1R [2]. |
ln Vivo |
Adomeglivant (LY2409021) (5 mg/kg; ip) completely eliminates the hypertensive effects of CNO (clozapine-N-oxide) in Avpires-Cre+ electrodes. (CNO is a cardiovascular drug stimulant that can induce hM3Dq-induced membrane myocardial infarction and increase the firing rate of hM3Dq-expressing arginine vasopressin (AVP) neurons) [3] Animal model: Avpires-Cre+ small Rat [3] Dosage: 5 mg/kg Administration method: intraperitoneal injection, 30 minutes before CNO. Results: The hyperglycemic effect of CNO was completely eliminated.
To establish the contribution of glucagon to this hyperglycaemic response, we pre-treated mice with the glucagon receptor antagonist Adomeglivant (LY2409021) . This completely abolished the hyperglycaemic action of CNO (Fig. 4b). Similarly, to understand the contribution of vasopressin 1b receptor (V1bR) signalling, we pre-treated mice with the V1bR antagonist SSR149415. This also abolished the hyperglycaemic effect of CNO (Fig. 4b). Measurements of plasma glucagon during CNO treatment revealed it was elevated by ~40% [3]. |
Enzyme Assay |
FRET reporter assay in a 96-well format [2]
HEK293 cells stably expressing recombinant GPCRs were plated at 80% confluence on 96-well clear-bottom assay plates coated with rat tail collagen. Cells were then transduced for 16 h with H188 virus at a density of ∼60,000 cells/well under conditions in which the multiplicity of infection was equivalent to 25 viral particles per cell. The culture media were removed and replaced by 200 μl/well of a standard extracellular saline (SES) solution supplemented with 11 mm glucose and 0.1% BSA. The composition of the SES was (in mm): 138 NaCl, 5.6 KCl, 2.6 CaCl2, 1.2 MgCl2, 11.1 glucose, and 10 Hepes (295 mosmol, pH 7.4). Real-time kinetic assays of FRET were performed using a FlexStation 3 microplate reader equipped with excitation and emission light monochromators. Excitation light was delivered at 435/9 nm (455 nm cutoff), and emitted light was detected at 485/15 nm (cyan fluorescent protein) or 535/15 nm (yellow fluorescent protein). The emission intensities were the averages of 12 excitation flashes for each time point per well. Test solutions dissolved in SES were placed in V-bottom 96-well plates, and an automated pipetting procedure was used to transfer 50 μl of each test solution to each well of the assay plate containing monolayers of these cells. The 485/535 emission ratio was calculated for each well, and the mean ± S.D. values for 12 wells were averaged. These FRET ratio values were normalized using baseline subtraction so that a y axis value of 0 corresponds to the initial baseline FRET ratio, whereas a value of 100 corresponds to a 100% increase (i.e. doubling) of the FRET ratio. The time course of the ΔFRET ratio was plotted after exporting data to Origin 8.0. Origin 8.0 was also used for nonlinear regression analysis to quantify dose-response relationships. |
Cell Assay |
HEK293 cells stably expressing the human GLP-1R at a density of 150,000 receptors/cell were used. HEK293 cells stably expressing the rat GlucR at a density of 250,000 receptors/cell were obtained from T. P. Sakmar, A. M. Cypess, and C. G. Unson. HEK293 cells stably expressing the rat GIP-R at a receptor density that has yet to be determined were obtained from T. J. Kieffer. HEK293 cells stably expressing H188 were generated by O. G. Chepurny in the Holz laboratory. All cell cultures were maintained in Dulbecco's modified Eagle's medium containing 25 mm glucose and supplemented with 10% fetal bovine serum and 1% penicillin/streptomycin. Cell cultures equilibrated at 37 °C in a humidified incubator that was gassed with 5% CO2 were passaged once a week [2].
|
Animal Protocol |
AAV-DIO-hM3Dq-mCherry was injected bilaterally into the supraoptic nucleus (SON) of Avpires-Cre+ mice. Mice were fasted for 4 hours (beginning at 10:00 am), and then CNO (or saline vehicle) was injected (3 mg/kg i.p.). In the same cohort, during a different trial, the glucagon receptor antagonist Adomeglivant (LY2409021)Adomeglivant (LY2409021)
AAV-DIO-hM3Dq was injected into ThCre+ mice, targeting A1/C1 neurons. CNO (1 mg/kg) was then injected (i.p.). Antagonists (or vehicle) for the V1bR (SSR149415, 30 mg/kg) or glucagon receptor (GCGR; Adomeglivant (LY2409021)
|
ADME/Pharmacokinetics |
Prediction of pharmacokinetics of CAA and PIB in human body and comparison of the results with the already known antagonists [1]
Certain structural and molecular features of compounds govern their pharmacokinetic properties in our body. Qikprop module of Schrodinger was used to evaluated the drug likeliness of all the four inhibitors. The obtained values for molecular weight, number of hydrogen bond donors, number of hydrogen bond acceptors and logP were used to assess violation of Lipinski's rule of five if any. To further account for the potential of the compounds to act as efficient drug candidates, their absorption, distribution, metabolism and excretion (ADME) properties were also calculated in silico using Qikprop. Physico-chemical properties and pharmacokinetics of GCGR inhibitors- CAA, PIB, MK-0893, LY2409021 [1] To supplement the information gained from binding affinity prediction, Qikprop was used to calculate various other physically significant descriptors and pharmaceutically relevant properties of these small molecules. Qikprop predicts these molecular properties and provides significant ranges for comparing their values with those of 95% of already known pharmaceutical drugs. The descriptor, "#star" denotes the number of outlying properties of the molecule i.e., the properties which do not fall within the range of values for already known drugs. So, lesser the number better is the druglikeness of the small molecule. MK-0893, which showed the highest binding affinity, had a #star value of 4 whereas all the other three compounds had 0 #star. Hence, except for MK-0893, the computed properties for the other three compounds did not lie outside the required range and were very similar to that of the known drugs. Lipinski's rule of five is a thumb rule which determines the likeliness of a drug to be orally active based on four molecular properties. Table 1 lists the values of all four properties for these four compounds. MK-0893 with a molecular weight of 588.48 and logP value of 8.18 was not satisfying the lipinski's rule (molecular weight < 500, no. of hydrogen bond donors < 5, no. of hydrogen bond acceptors < 10, logP < 5). Solvent accessible surface area (SASA) and especially polar surface area (PSA) dictate the passive transport of molecules through membranes thereby giving an estimate about the transport properties of the drugs. The total SASA for MK-0893, CAA, PIB and LY2409021 was well within the range given by QikProp. Using some knowledge based set of rules Qikprop also calculates the percentage probability of the drug getting orally absorbed in the human body. This value has been shown to correlate well with the human oral absorption. PIB showed the highest oral absorption with a percentage value of 100%. Out of rest three, LY2409021 had the least value of 28.78 %. Central nervous system activity is another parameter that needs to be considered for assessing the safety issue. CAA was found to be highly CNS inactive whereas PIB was predicted to possess some minimal amount of CNS activity. Blood brain barrier (BBB) separates the human brain from the direct contact of circulatory system, thus protecting the brain for unwanted solute particles. Both the predicted compounds were shown to be BBB negative ensuring their administration safe for the brain. Even though MK-0893 had higher affinity for GCGR, CAA and PIB were found to be better than this known inhibitor in many aspects. The new compounds showed better druglikeness with acceptable values of ADME properties. This clearly delineates the distinctive potential of CAA and PIB as prospective lead inhibitors of GCGR for the treatment of type II diabetes mellitus. |
References |
|
Additional Infomation |
Objective: Type 2 diabetes (T2D) pathophysiology includes fasting and postprandial hyperglucagonemia, which has been linked to hyperglycemia via increased endogenous glucose production (EGP). We used a glucagon receptor antagonist (LY2409021) and stable isotope tracer infusions to investigate the consequences of hyperglucagonemia in T2D.
Design: A double-blinded, randomized, placebo-controlled crossover study was conducted.
Methods: Ten patients with T2D and ten matched non-diabetic controls underwent two liquid mixed meal tests preceded by single-dose administration of LY2409021 (100 mg) or placebo. Double-tracer technique was used to quantify EGP. Antagonist selectivity toward related incretin receptors was determined in vitro.
Results: Compared to placebo, LY2409021 lowered the fasting plasma glucose (FPG) from 9.1 to 7.1 mmol/L in patients and from 5.6 to 5.0 mmol/L in controls (both P < 0.001) by mechanisms involving reduction of EGP. Postprandial plasma glucose excursions (baseline-subtracted area under the curve) were unaffected by LY2409021 in patients and increased in controls compared to placebo. Glucagon concentrations more than doubled during glucagon receptor antagonism. The antagonist interfered with both glucagon-like peptide 1 and glucose-dependent insulinotropic polypeptide receptors, complicating the interpretation of the postprandial data.
Conclusions: LY2409021 lowered FPG concentrations but did not improve postprandial glucose tolerance after a meal in patients with T2D and controls. The metabolic consequences of postprandial hyperglucagonemia are difficult to evaluate using LY2409021 because of its antagonizing effects on the incretin receptors.[4]
Adomeglivant has been investigated for the basic science of Type 2 Diabetes. ADOMEGLIVANT is a small molecule drug with a maximum clinical trial phase of II (across all indications) and has 3 investigational indications. Background: Interaction of the small peptide hormone glucagon with glucagon receptor (GCGR) stimulates the release of glucose from the hepatic cells during fasting; hence GCGR performs a significant function in glucose homeostasis. Inhibiting the interaction between glucagon and its receptor has been reported to control hepatic glucose overproduction and thus GCGR has evolved as an attractive therapeutic target for the treatment of type II diabetes mellitus. Results: In the present study, a large library of natural compounds was screened against 7 transmembrane domain of GCGR to identify novel therapeutic molecules that can inhibit the binding of glucagon with GCGR. Molecular dynamics simulations were performed to study the dynamic behaviour of the docked complexes and the molecular interactions between the screened compounds and the ligand binding residues of GCGR were analysed in detail. The top scoring compounds were also compared with already documented GCGR inhibitors- MK-0893 and LY2409021 for their binding affinity and other ADME properties. Finally, we have reported two natural drug like compounds PIB and CAA which showed good binding affinity for GCGR and are potent inhibitor of its functional activity. Conclusion: This study contributes evidence for application of these compounds as prospective small ligand molecules against type II diabetes. Novel natural drug like inhibitors against the 7 transmembrane domain of GCGR have been identified which showed high binding affinity and potent inhibition of GCGR. [1] G protein-coupled receptors (GPCRs) for glucagon (GluR) and glucagon-like peptide-1 (GLP-1R) are normally considered to be highly selective for glucagon and GLP-1, respectively. However, glucagon secreted from pancreatic α-cells may accumulate at high concentrations to exert promiscuous effects at the β-cell GLP-1R, as may occur in the volume-restricted microenvironment of the islets of Langerhans. Furthermore, systemic administration of GluR or GLP-1R agonists and antagonists at high doses may lead to off-target effects at other receptors. Here, we used molecular modeling to evaluate data derived from FRET assays that detect cAMP as a read-out for GluR and GLP-1R activation. This analysis established that glucagon is a nonconventional GLP-1R agonist, an effect inhibited by the GLP-1R orthosteric antagonist exendin(9-39) (Ex(9-39)). The GluR allosteric inhibitors LY2409021 and MK 0893 antagonized glucagon and GLP-1 action at the GLP-1R, whereas des-His1-[Glu9]glucagon antagonized glucagon action at the GluR, while having minimal inhibitory action versus glucagon or GLP-1 at the GLP-1R. When testing Ex(9-39) in combination with des-His1-[Glu9]glucagon in INS-1 832/13 cells, we validated a dual agonist action of glucagon at the GluR and GLP-1R. Hybrid peptide GGP817 containing glucagon fused to a fragment of peptide YY (PYY) acted as a triagonist at the GluR, GLP-1R, and neuropeptide Y2 receptor (NPY2R). Collectively, these findings provide a new triagonist strategy with which to target the GluR, GLP-1R, and NPY2R. They also provide an impetus to reevaluate prior studies in which GluR and GLP-1R agonists and antagonists were assumed not to exert promiscuous actions at other GPCRs. [2] Hypoglycaemia is a major barrier to the treatment of diabetes. Accordingly, it is important that we understand the mechanisms regulating the circulating levels of glucagon – the body’s principle blood glucose-elevating hormone which is secreted from alpha-cells of the pancreatic islets. In isolated islets, varying glucose over the range of concentrations that occur physiologically between the fed and fuel-deprived states (from 8 to 4 mM) has no significant effect on glucagon secretion and yet associates with dramatic changes in plasma glucagon in vivo. The identity of the systemic factor that stimulates glucagon secretion in vivo remains unknown. Here, we show that arginine-vasopressin (AVP), secreted from the posterior pituitary, stimulates glucagon secretion. Glucagon-secreting alpha-cells express high levels of the vasopressin 1b receptor (V1bR). Activation of AVP neurons in vivo increased circulating AVP, stimulated glucagon release and evoked hyperglycaemia; effects blocked by pharmacological antagonism of either the glucagon receptor or vasopressin 1b receptor. AVP also mediates the stimulatory effects of dehydration and hypoglycaemia produced by exogenous insulin and 2-deoxy-D-glucose on glucagon secretion. We show that the A1/C1 neurons of the medulla oblongata, which are known to be activated by hypoglycaemia, drive AVP neuron activation in response to insulin-induced hypoglycaemia. Hypoglycaemia also increases circulating levels of copeptin (derived from the same pre-pro hormone as AVP) in humans and this hormone stimulates glucagon secretion from isolated human islets. In patients with type 1 diabetes, hypoglycaemia failed to increase both plasma copeptin and glucagon. These findings provide a new mechanism for the central regulation of glucagon secretion in both health and disease.[3] |
Molecular Formula |
C32H36F3NO4
|
---|---|
Molecular Weight |
555.63
|
Exact Mass |
555.259
|
Elemental Analysis |
C, 69.17; H, 6.53; F, 10.26; N, 2.52; O, 11.52
|
CAS # |
872260-19-0
|
Related CAS # |
872260-47-4 (racemic); 1488363-78-5; 488363-78-5 (S-isomer); 872260-19-0 (R-isomer)
|
PubChem CID |
91936837
|
Appearance |
Typically exists as solids at room temperature
|
LogP |
7.53
|
InChi Key |
FASLTMSUPQDLIB-HHHXNRCGSA-N
|
InChi Code |
InChI=1S/C32H36F3NO4/c1-20-18-26(19-21(2)29(20)23-10-12-25(13-11-23)31(3,4)5)40-27(14-16-32(33,34)35)22-6-8-24(9-7-22)30(39)36-17-15-28(37)38/h6-13,18-19,27H,14-17H2,1-5H3,(H,36,39)(H,37,38)/t27-/m1/s1
|
Chemical Name |
3-[[4-[(1R)-1-[4-(4-tert-butylphenyl)-3,5-dimethylphenoxy]-4,4,4-trifluorobutyl]benzoyl]amino]propanoic acid
|
Synonyms |
(+)-LY2409021; Adomeglivant, (+)-; RIM88PH2RA; UNII-RIM88PH2RA; 872260-19-0; (+)-adomeglivant;
|
HS Tariff Code |
2934.99.9001
|
Storage |
Powder -20°C 3 years 4°C 2 years In solvent -80°C 6 months -20°C 1 month |
Shipping Condition |
Room temperature (This product is stable at ambient temperature for a few days during ordinary shipping and time spent in Customs)
|
Solubility (In Vitro) |
May dissolve in DMSO (in most cases), if not, try other solvents such as H2O, Ethanol, or DMF with a minute amount of products to avoid loss of samples
|
---|---|
Solubility (In Vivo) |
Note: Listed below are some common formulations that may be used to formulate products with low water solubility (e.g. < 1 mg/mL), you may test these formulations using a minute amount of products to avoid loss of samples.
Injection Formulations
Injection Formulation 1: DMSO : Tween 80: Saline = 10 : 5 : 85 (i.e. 100 μL DMSO stock solution → 50 μL Tween 80 → 850 μL Saline)(e.g. IP/IV/IM/SC) *Preparation of saline: Dissolve 0.9 g of sodium chloride in 100 mL ddH ₂ O to obtain a clear solution. Injection Formulation 2: DMSO : PEG300 :Tween 80 : Saline = 10 : 40 : 5 : 45 (i.e. 100 μL DMSO → 400 μLPEG300 → 50 μL Tween 80 → 450 μL Saline) Injection Formulation 3: DMSO : Corn oil = 10 : 90 (i.e. 100 μL DMSO → 900 μL Corn oil) Example: Take the Injection Formulation 3 (DMSO : Corn oil = 10 : 90) as an example, if 1 mL of 2.5 mg/mL working solution is to be prepared, you can take 100 μL 25 mg/mL DMSO stock solution and add to 900 μL corn oil, mix well to obtain a clear or suspension solution (2.5 mg/mL, ready for use in animals). View More
Injection Formulation 4: DMSO : 20% SBE-β-CD in saline = 10 : 90 [i.e. 100 μL DMSO → 900 μL (20% SBE-β-CD in saline)] Oral Formulations
Oral Formulation 1: Suspend in 0.5% CMC Na (carboxymethylcellulose sodium) Oral Formulation 2: Suspend in 0.5% Carboxymethyl cellulose Example: Take the Oral Formulation 1 (Suspend in 0.5% CMC Na) as an example, if 100 mL of 2.5 mg/mL working solution is to be prepared, you can first prepare 0.5% CMC Na solution by measuring 0.5 g CMC Na and dissolve it in 100 mL ddH2O to obtain a clear solution; then add 250 mg of the product to 100 mL 0.5% CMC Na solution, to make the suspension solution (2.5 mg/mL, ready for use in animals). View More
Oral Formulation 3: Dissolved in PEG400  (Please use freshly prepared in vivo formulations for optimal results.) |
Preparing Stock Solutions | 1 mg | 5 mg | 10 mg | |
1 mM | 1.7998 mL | 8.9988 mL | 17.9976 mL | |
5 mM | 0.3600 mL | 1.7998 mL | 3.5995 mL | |
10 mM | 0.1800 mL | 0.8999 mL | 1.7998 mL |
*Note: Please select an appropriate solvent for the preparation of stock solution based on your experiment needs. For most products, DMSO can be used for preparing stock solutions (e.g. 5 mM, 10 mM, or 20 mM concentration); some products with high aqueous solubility may be dissolved in water directly. Solubility information is available at the above Solubility Data section. Once the stock solution is prepared, aliquot it to routine usage volumes and store at -20°C or -80°C. Avoid repeated freeze and thaw cycles.
Calculation results
Working concentration: mg/mL;
Method for preparing DMSO stock solution: mg drug pre-dissolved in μL DMSO (stock solution concentration mg/mL). Please contact us first if the concentration exceeds the DMSO solubility of the batch of drug.
Method for preparing in vivo formulation::Take μL DMSO stock solution, next add μL PEG300, mix and clarify, next addμL Tween 80, mix and clarify, next add μL ddH2O,mix and clarify.
(1) Please be sure that the solution is clear before the addition of next solvent. Dissolution methods like vortex, ultrasound or warming and heat may be used to aid dissolving.
(2) Be sure to add the solvent(s) in order.