Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters










Publication year range
1.
SLAS Discov ; 29(4): 100159, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38723666

ABSTRACT

To confirm target engagement of hits from our high-throughput screening efforts, we ran biophysical assays on several hundreds of hits from 15 different high-throughput screening campaigns. Analyzing the biophysical assay results from these screening campaigns led us to conclude that we could be more strategic in our biophysical analysis of hits by first confirming activity in a thermal shift assay (TSA) and then confirming activity in either a surface plasmon resonance (SPR) assay or a temperature-related intensity change (TRIC) assay. To understand how this new workflow shapes the quality of the final hits, we compared TSA/SPR or TSA/TRIC confirmed and unconfirmed hits to one another using four measures of compound quality: quantitative estimate of drug-likeness (QED), Pan-Assay Interference Compounds (PAINS), promiscuity, and aqueous solubility. In general, we found that the biophysically confirmed hits performed better in the compound quality metrics than the unconfirmed hits, demonstrating that our workflow not only confirmed target engagement of the hits but also enriched for higher quality hits.

2.
J Med Chem ; 63(20): 11602-11614, 2020 10 22.
Article in English | MEDLINE | ID: mdl-32965113

ABSTRACT

A comprehensive understanding of structure-reactivity relationships is critical to the design and optimization of cysteine-targeted covalent inhibitors. Herein, we report glutathione (GSH) reaction rates for N-phenyl acrylamides with varied substitutions at the α- and ß-positions of the acrylamide moiety. We find that the GSH reaction rates can generally be understood in terms of the electron donating or withdrawing ability of the substituent. When installed at the ß-position, aminomethyl substituents with amine pKa's > 7 accelerate, while those with pKa's < 7 slow the rate of GSH addition at pH 7.4, relative to a hydrogen substituent. Although a computational model was able to only approximately capture experimental reactivity trends, our calculations do not support a frequently invoked mechanism of concerted amine/thiol proton transfer and C-S bond formation and instead suggest that protonated aminomethyl functions as an electron-withdrawing group to reduce the barrier for thiolate addition to the acrylamide.


Subject(s)
Acrylamides/chemical synthesis , Glutathione/chemistry , Acrylamides/chemistry , Amines/chemistry , Cysteine/chemistry , Molecular Structure , Structure-Activity Relationship
3.
ACS Chem Biol ; 11(10): 2734-2743, 2016 10 21.
Article in English | MEDLINE | ID: mdl-27434622

ABSTRACT

The efficacy of therapeutic antibodies that induce antibody-dependent cellular cytotoxicity can be improved by reduced fucosylation. Consequently, fucosylation is a critical product attribute of monoclonal antibodies produced as protein therapeutics. Small molecule fucosylation inhibitors have also shown promise as potential therapeutics in animal models of tumors, arthritis, and sickle cell disease. Potent small molecule metabolic inhibitors of cellular protein fucosylation, 6,6,6-trifluorofucose per-O-acetate and 6,6,6-trifluorofucose (fucostatin I), were identified that reduces the fucosylation of recombinantly expressed antibodies in cell culture in a concentration-dependent fashion enabling the controlled modulation of protein fucosylation levels. 6,6,6-Trifluorofucose binds at an allosteric site of GDP-mannose 4,6-dehydratase (GMD) as revealed for the first time by the X-ray cocrystal structure of a bound allosteric GMD inhibitor. 6,6,6-Trifluorofucose was found to be incorporated in place of fucose at low levels (<1%) in the glycans of recombinantly expressed antibodies. A fucose-1-phosphonate analog, fucostatin II, was designed that inhibits fucosylation with no incorporation into antibody glycans, allowing the production of afucosylated antibodies in which the incorporation of non-native sugar is completely absent-a key advantage in the production of therapeutic antibodies, especially biosimilar antibodies. Inhibitor structure-activity relationships, identification of cellular and inhibitor metabolites in inhibitor-treated cells, fucose competition studies, and the production of recombinant antibodies with varying levels of fucosylation are described.


Subject(s)
Fucose/metabolism , Hydro-Lyases/metabolism , Small Molecule Libraries , Animals , CHO Cells , Cricetinae , Cricetulus , Crystallography, X-Ray , Fucose/antagonists & inhibitors , Guanosine Diphosphate Mannose/metabolism , Mass Spectrometry , Molecular Structure , Surface Plasmon Resonance
4.
J Med Chem ; 58(24): 9663-79, 2015 Dec 24.
Article in English | MEDLINE | ID: mdl-26551034

ABSTRACT

The HTS-based discovery and structure-guided optimization of a novel series of GKRP-selective GK-GKRP disrupters are revealed. Diarylmethanesulfonamide hit 6 (hGK-hGKRP IC50 = 1.2 µM) was optimized to lead compound 32 (AMG-0696; hGK-hGKRP IC50 = 0.0038 µM). A stabilizing interaction between a nitrogen atom lone pair and an aromatic sulfur system (nN → σ*S-X) in 32 was exploited to conformationally constrain a biaryl linkage and allow contact with key residues in GKRP. Lead compound 32 was shown to induce GK translocation from the nucleus to the cytoplasm in rats (IHC score = 0; 10 mg/kg po, 6 h) and blood glucose reduction in mice (POC = -45%; 100 mg/kg po, 3 h). X-ray analyses of 32 and several precursors bound to GKRP were also obtained. This novel disrupter of GK-GKRP binding enables further exploration of GKRP as a potential therapeutic target for type II diabetes and highlights the value of exploiting unconventional nonbonded interactions in drug design.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Glucokinase/metabolism , Hypoglycemic Agents/chemistry , Sulfonamides/chemistry , Thiophenes/chemistry , Active Transport, Cell Nucleus , Animals , Blood Glucose/metabolism , Cell Nucleus/metabolism , Crystallography, X-Ray , Cytoplasm/metabolism , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , Male , Mice , Microsomes, Liver/metabolism , Models, Molecular , Molecular Conformation , Protein Binding , Rats, Sprague-Dawley , Stereoisomerism , Structure-Activity Relationship , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology , Thiophenes/pharmacokinetics , Thiophenes/pharmacology
5.
J Med Chem ; 58(23): 9171-8, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26580091

ABSTRACT

Success in the design of targeted covalent inhibitors depends in part on a knowledge of the factors influencing electrophile reactivity. In an effort to further develop an understanding of structure-reactivity relationships among N-arylacrylamides, we determined glutathione (GSH) reaction rates for a family of N-arylacrylamides independently substituted at ortho-, meta-, and para-positions with 11 different groups common to inhibitor design. We find that substituent effects on reaction rates show a linear Hammett correlation for ortho-, meta-, and para-substitution. In addition, we note a correlation between (1)H and (13)C NMR chemical shifts of the acrylamide with GSH reaction rates, suggesting that NMR chemical shifts may be a convenient surrogate measure of relative acrylamide reactivity. Density functional theory calculations reveal a correlation between computed activation parameters and experimentally determined reaction rates, validating the use of such methodology for the screening of synthetic candidates in a prospective fashion.


Subject(s)
Acrylamides/chemistry , Acrylamides/pharmacology , Glutathione/metabolism , Drug Discovery , Kinetics , Magnetic Resonance Spectroscopy , Models, Molecular , Thermodynamics
6.
J Med Chem ; 58(11): 4462-82, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-25914941

ABSTRACT

The glucokinase-glucokinase regulatory protein (GK-GKRP) complex plays an important role in controlling glucose homeostasis in the liver. We have recently disclosed a series of arylpiperazines as in vitro and in vivo disruptors of the GK-GKRP complex with efficacy in rodent models of type 2 diabetes mellitus (T2DM). Herein, we describe a new class of aryl sulfones as disruptors of the GK-GKRP complex, where the central piperazine scaffold has been replaced by an aromatic group. Conformational analysis and exploration of the structure-activity relationships of this new class of compounds led to the identification of potent GK-GKRP disruptors. Further optimization of this novel series delivered thiazole sulfone 93, which was able to disrupt the GK-GKRP interaction in vitro and in vivo and, by doing so, increases cytoplasmic levels of unbound GK.


Subject(s)
Aminopyridines/pharmacology , Carrier Proteins/antagonists & inhibitors , Glucokinase/antagonists & inhibitors , Hypoglycemic Agents/pharmacology , Liver/drug effects , Small Molecule Libraries/pharmacology , Sulfones/chemistry , Aminopyridines/chemistry , Animals , Carrier Proteins/metabolism , Crystallography, X-Ray , Glucokinase/metabolism , Glucose/metabolism , Hypoglycemic Agents/chemistry , Liver/cytology , Liver/metabolism , Models, Molecular , Molecular Conformation , Molecular Structure , Rats , Rats, Sprague-Dawley , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Sulfones/pharmacology
7.
J Med Chem ; 57(14): 5949-64, 2014 Jul 24.
Article in English | MEDLINE | ID: mdl-25001129

ABSTRACT

Structure-activity relationship investigations conducted at the 5-position of the N-pyridine ring of a series of N-arylsulfonyl-N'-2-pyridinyl-piperazines led to the identification of a novel bis-pyridinyl piperazine sulfonamide (51) that was a potent disruptor of the glucokinase-glucokinase regulatory protein (GK-GKRP) interaction. Analysis of the X-ray cocrystal of compound 51 bound to hGKRP revealed that the 3-pyridine ring moiety occupied a previously unexplored binding pocket within the protein. Key features of this new binding mode included forming favorable contacts with the top face of the Ala27-Val28-Pro29 ("shelf region") as well as an edge-to-face interaction with the Tyr24 side chain. Compound 51 was potent in both biochemical and cellular assays (IC50=0.005 µM and EC50=0.205 µM, respectively) and exhibited acceptable pharmacokinetic properties for in vivo evaluation. When administered to db/db mice (100 mg/kg, po), compound 51 demonstrated a robust pharmacodynamic effect and significantly reduced blood glucose levels up to 6 h postdose.


Subject(s)
Carrier Proteins/antagonists & inhibitors , Glucokinase/antagonists & inhibitors , Glucokinase/metabolism , Piperazines/pharmacology , Small Molecule Libraries/pharmacology , Binding Sites/drug effects , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Crystallography, X-Ray , Dose-Response Relationship, Drug , Glucokinase/chemistry , Humans , Models, Molecular , Molecular Conformation , Piperazines/chemical synthesis , Piperazines/chemistry , Protein Binding/drug effects , Pyridines/chemistry , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship
8.
J Med Chem ; 57(7): 3094-116, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24611879

ABSTRACT

We have recently reported a novel approach to increase cytosolic glucokinase (GK) levels through the binding of a small molecule to its endogenous inhibitor, glucokinase regulatory protein (GKRP). These initial investigations culminated in the identification of 2-(4-((2S)-4-((6-amino-3-pyridinyl)sulfonyl)-2-(1-propyn-1-yl)-1-piperazinyl)phenyl)-1,1,1,3,3,3-hexafluoro-2-propanol (1, AMG-3969), a compound that effectively enhanced GK translocation and reduced blood glucose levels in diabetic animals. Herein we report the results of our expanded SAR investigations that focused on modifications to the aryl carbinol group of this series. Guided by the X-ray cocrystal structure of compound 1 bound to hGKRP, we identified several potent GK-GKRP disruptors bearing a diverse set of functionalities in the aryl carbinol region. Among them, sulfoximine and pyridinyl derivatives 24 and 29 possessed excellent potency as well as favorable PK properties. When dosed orally in db/db mice, both compounds significantly lowered fed blood glucose levels (up to 58%).


Subject(s)
Carrier Proteins/antagonists & inhibitors , Diabetes Mellitus/drug therapy , Glucokinase/antagonists & inhibitors , Hepatocytes/drug effects , Microsomes, Liver/drug effects , Piperazines/chemistry , Sulfonamides/pharmacology , Animals , Biological Availability , Blood Glucose/metabolism , Carrier Proteins/metabolism , Crystallography, X-Ray , Diabetes Mellitus/metabolism , Disease Models, Animal , Glucokinase/metabolism , Hepatocytes/metabolism , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Mice , Microsomes, Liver/metabolism , Models, Molecular , Piperazines/pharmacology , Rats , Stereoisomerism , Structure-Activity Relationship , Sulfonamides/chemistry
9.
J Med Chem ; 57(2): 309-24, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-24405172

ABSTRACT

Small molecule activators of glucokinase have shown robust efficacy in both preclinical models and humans. However, overactivation of glucokinase (GK) can cause excessive glucose turnover, leading to hypoglycemia. To circumvent this adverse side effect, we chose to modulate GK activity by targeting the endogenous inhibitor of GK, glucokinase regulatory protein (GKRP). Disrupting the GK-GKRP complex results in an increase in the amount of unbound cytosolic GK without altering the inherent kinetics of the enzyme. Herein we report the identification of compounds that efficiently disrupt the GK-GKRP interaction via a previously unknown binding pocket. Using a structure-based approach, the potency of the initial hit was improved to provide 25 (AMG-1694). When dosed in ZDF rats, 25 showed both a robust pharmacodynamic effect as well as a statistically significant reduction in glucose. Additionally, hypoglycemia was not observed in either the hyperglycemic or normal rats.


Subject(s)
Carrier Proteins/metabolism , Glucokinase/metabolism , Hypoglycemic Agents/chemistry , Piperazines/chemistry , Animals , Binding Sites , Carrier Proteins/chemistry , Crystallography, X-Ray , Glucokinase/chemistry , Hepatocytes/drug effects , Hepatocytes/metabolism , High-Throughput Screening Assays , Humans , Hypoglycemia/chemically induced , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/pharmacology , Piperazines/adverse effects , Piperazines/pharmacology , Protein Conformation , Protein Transport , Rats , Rats, Zucker , Stereoisomerism , Structure-Activity Relationship , Sulfonamides/adverse effects , Sulfonamides/chemistry , Sulfonamides/pharmacology
10.
J Med Chem ; 57(2): 325-38, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-24405213

ABSTRACT

In the previous report , we described the discovery and optimization of novel small molecule disruptors of the GK-GKRP interaction culminating in the identification of 1 (AMG-1694). Although this analogue possessed excellent in vitro potency and was a useful tool compound in initial proof-of-concept experiments, high metabolic turnover limited its advancement. Guided by a combination of metabolite identification and structure-based design, we have successfully discovered a potent and metabolically stable GK-GKRP disruptor (27, AMG-3969). When administered to db/db mice, this compound demonstrated a robust pharmacodynamic response (GK translocation) as well as statistically significant dose-dependent reductions in fed blood glucose levels.


Subject(s)
Carrier Proteins/metabolism , Glucokinase/metabolism , Hypoglycemic Agents/chemistry , Piperazines/chemical synthesis , Sulfonamides/chemical synthesis , Alkynes/chemical synthesis , Alkynes/pharmacokinetics , Alkynes/pharmacology , Animals , Blood Glucose/metabolism , Carrier Proteins/chemistry , Glucokinase/chemistry , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , Mice , Microsomes, Liver/metabolism , Models, Molecular , Morpholines/chemical synthesis , Morpholines/pharmacokinetics , Morpholines/pharmacology , Piperazines/pharmacokinetics , Piperazines/pharmacology , Protein Binding , Protein Transport , Rats , Stereoisomerism , Structure-Activity Relationship , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology
11.
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
12.
Bioorg Med Chem Lett ; 23(24): 6625-8, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24215889

ABSTRACT

A series of urea based calcimimetics was optimized for potency and oral bioavailability. Crucial to this process was overcoming the poor pharmacokinetic properties of lead thiazole 1. Metabolism-guided modifications, characterized by the use of metabolite identification (ID) and measurement of time dependent inhibition (TDI) of CYP3A4, were essential to finding a compound suitable for oral dosing. Calcimimetic 18 exhibited excellent in vivo potency in a 5/6 nephrectomized rat model and cross-species pharmacokinetics.


Subject(s)
Hyperparathyroidism, Secondary/drug therapy , Thiazoles/chemistry , Thiazoles/therapeutic use , Urea/analogs & derivatives , Administration, Oral , Animals , Biological Availability , Half-Life , Hyperparathyroidism, Secondary/metabolism , Hyperparathyroidism, Secondary/pathology , Male , Parathyroid Hormone/metabolism , Protein Binding , Rats , Rats, Sprague-Dawley , Receptors, Calcium-Sensing/chemistry , Receptors, Calcium-Sensing/metabolism , Thiazoles/pharmacokinetics
13.
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
14.
Bioorg Med Chem Lett ; 23(23): 6396-400, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24120542

ABSTRACT

A series of aminooxadiazoles was optimized for inhibition of Cdc7. Early lead isoquinoline 1 suffered from modest cell potency (cellular IC50=0.71 µM measuring pMCM2), low selectivity against structurally related kinases, and high IV clearance in rats (CL=18 L/h/kg). Extensive optimization resulted in azaindole 26 (Cdc7 IC50=1.1 nM, pMCM2 IC50=32 nM) that demonstrated robust lowering of pMCM2 in a mouse pharmacodynamic (PD) model when dosed orally. Modifications to improve the pharmacokinetic profile of this series were guided by trapping experiments with glutathione in rat hepatocytes.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Oxadiazoles/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Disease Models, Animal , Female , Mice , Mice, Nude , Molecular Structure , Oxadiazoles/chemical synthesis , Oxadiazoles/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Rats , Structure-Activity Relationship , Xenograft Model Antitumor Assays
16.
Bioorg Med Chem Lett ; 22(1): 527-31, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22104144

ABSTRACT

We reveal how a N-scan SAR strategy (systematic substitution of each CH group with a N atom) was employed for quinolinone-based S1P(1) agonist 5 to modulate physicochemical properties and optimize in vitro and in vivo activity. The diaza-analog 17 displays improved potency (hS1P(1) RI; 17: EC(50)=0.020 µM, 120% efficacy; 5: EC(50)=0.070 µM, 110% efficacy) and selectivity (hS1P(3) Ca(2+) flux; 17: EC(50) >25 µM; 5: EC(50)=1.5 µM, 92% efficacy), as well as enhanced pharmacokinetics (17: CL=0.15 L/h/kg, V(dss)=5.1L/kg, T(1/2)=24h, %F=110; 5: CL=0.93L/h/kg, V(dss)=11L/kg, T(1/2)=15 h, %F=60) and pharmacodynamics (17: 1.0mg/kg po, 24h PLC POC=-67%; 5: 3mg/kg po, 24h PLC POC=-51%) in rat.


Subject(s)
Chemistry, Physical/methods , Quinolones/pharmacology , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/chemistry , Animals , Area Under Curve , Cardiovascular Diseases/metabolism , Drug Design , Female , Humans , Immunosuppressive Agents/pharmacology , In Vitro Techniques , Kinetics , Lymphocytes/cytology , Lymphocytes/metabolism , Models, Chemical , Multiple Sclerosis/drug therapy , Quinolones/chemistry , Rats , Rats, Inbred Lew , Structure-Activity Relationship
17.
ACS Med Chem Lett ; 3(1): 74-8, 2012 Jan 12.
Article in English | MEDLINE | ID: mdl-24900374

ABSTRACT

The optimization of a series of S1P1 agonists with limited activity against S1P3 is reported. A polar headgroup was used to improve the physicochemical and pharmacokinetic parameters of lead quinolinone 6. When dosed orally at 1 and 3 mg/kg, the azahydroxymethyl analogue 22 achieved statistically significant lowering of circulating blood lymphocytes 24 h postdose. In rats, a dose-proportional increase in exposure was measured when 22 was dosed orally at 2 and 100 mg/kg.

19.
Bioorg Med Chem Lett ; 21(11): 3384-9, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21514825

ABSTRACT

The discovery of novel and highly potent oxopiperazine based B1 receptor antagonists is described. Compared to the previously described arylsulfonylated (R)-3-amino-3-phenylpropionic acid series, the current compounds showed improved in vitro potency and metabolic stability. Compound 17, 2-((2R)-1-((4-methylphenyl)sulfonyl)-3-oxo-2-piperazinyl)-N-((1R)-6-(1-piperidinylmethyl)-1,2,3,4-tetrahydro-1-naphthalenyl)acetamide, showed EC(50) of 10.3 nM in a rabbit biochemical challenge model. The practical syntheses of chiral arylsulfonylated oxopiperazine acetic acids are also described.


Subject(s)
Acetamides/therapeutic use , Bradykinin B1 Receptor Antagonists , Inflammation/drug therapy , Pain/drug therapy , Piperazines/therapeutic use , Acetamides/chemical synthesis , Acetamides/chemistry , Animals , Dogs , Inhibitory Concentration 50 , Mice , Models, Animal , Molecular Structure , Piperazines/chemical synthesis , Piperazines/chemistry , Rabbits , Rats , Receptor, Bradykinin B1/chemistry , Stereoisomerism , Structure-Activity Relationship
20.
J Pharmacol Exp Ther ; 337(3): 681-91, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21422163

ABSTRACT

Calcimimetics are positive allosteric modulators to the calcium-sensing receptor (CaSR). Activation of the CaSR inhibits the secretion of parathyroid hormone (PTH), stimulates the secretion of calcitonin, and decreases serum calcium (Ca(2+)). Cinacalcet, a second-generation calcimimetic, is used therapeutically to control PTH in patients with chronic kidney disease who are on dialysis with secondary hyperparathyroidism. A calcimimetic that displays increased separation of PTH versus Ca(2+) lowering in patients would potentially allow the use of calcimimetics to treat patients in earlier stages of renal disease because hypocalcemia can develop in this population. Toward this end, we developed a third-generation calcimimetic, determined the molecular pharmacological properties of it using an operation model of allosteric modulation/agonism, and measured the compound effects on PTH, serum ionized Ca(2+), and calcitonin levels in 5/6 nephrectomized rats. We found the new molecule effectively reduced PTH levels without promoting calcitonin secretion or hypocalcemia. Furthermore, our third-generation molecule was less efficacious at promoting calcitonin secretion from human thyroid carcinoma cells compared with 3-(2-chlorophenyl)-N-((1R)-1-(3-methoxyphenyl)ethyl)-1-propanamine (R-568), a first-generation calcimimetic. These data provide evidence that calcimimetics with increased potency can be used to lower PTH without production of significant hypocalcemia because the threshold for inhibition of PTH secretion is much lower than the threshold for calcitonin secretion.


Subject(s)
Aniline Compounds/pharmacology , Biphenyl Compounds/pharmacology , Calcitonin/metabolism , Calcium/agonists , Calcium/metabolism , Diethylamines/pharmacology , Hyperparathyroidism, Secondary/drug therapy , Parathyroid Hormone/metabolism , Receptors, Calcium-Sensing/metabolism , Animals , Biphenyl Compounds/administration & dosage , CHO Cells , Calcitonin/blood , Calcium/blood , Cricetinae , Cricetulus , Diethylamines/administration & dosage , HEK293 Cells , Humans , Hyperparathyroidism, Secondary/etiology , Hypocalcemia/complications , Inositol Phosphates/metabolism , Kidney Failure, Chronic/complications , Male , Parathyroid Glands/drug effects , Parathyroid Hormone/blood , Phenethylamines , Phosphorylation/drug effects , Propylamines , Rats , Rats, Sprague-Dawley , Renal Dialysis/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...