HK1126489B - Melanocortin type 4 receptor agonist piperidinoylpyrrolidines - Google Patents
Melanocortin type 4 receptor agonist piperidinoylpyrrolidines Download PDFInfo
- Publication number
- HK1126489B HK1126489B HK09105357.7A HK09105357A HK1126489B HK 1126489 B HK1126489 B HK 1126489B HK 09105357 A HK09105357 A HK 09105357A HK 1126489 B HK1126489 B HK 1126489B
- Authority
- HK
- Hong Kong
- Prior art keywords
- carbonyl
- pyrrolidin
- dimethylpiperidin
- hydroxy
- chloropyridin
- Prior art date
Links
Description
The present invention relates to certain classes of compounds, and pharmaceutically acceptable salts, solvates and prodrugs thereof, which are melanocortin 4(MC4) receptor agonists, in particular to selective MC4 agonist compounds, to their use in medicine, to compositions of said compounds, to processes for preparing them and to intermediates used in these processes. In particular, the present invention relates to MCR4 agonist piperidinecarbonylpyrrolidines (piperidinylpyrrolidines) compounds useful for the treatment of sexual dysfunction, obesity, diabetes and other diseases. [ the terms "MC 4", "MC 4 receptor", "MCR 4", "MC 4-R", and the like, are used interchangeably herein. ]
Melanocortins are pro-opiod melanocortin (POMC) -derived peptides that bind to and activate G-protein coupled receptors (GPCR's) of the melanocortin receptor family. Melanocortins regulate many physiological processes including sexual function and sexual behavior, food intake and metabolism. 5 melanocortin receptors, MCR1, MCR2, MCR3, MCR4, MCR5, have been cloned and expressed in a variety of tissues. MCR1 is specifically expressed in melanocytes and melanoma cells, MCR2 is the ACTH receptor and is expressed in adrenal tissue, MCR3 is mainly expressed in the brain and limbic system, MCR4 is widely expressed in the brain and spinal cord, and MCR5 is expressed in the brain and many peripheral tissues including skin, adipose tissue, skeletal muscle and lymphoid tissue. MCR3 may be involved in controlling sexual function, food intake and thermogenesis.
MC4-R is a G protein-coupled seven transmembrane receptor expressed primarily in the hypothalamus, hippocampus, and thalamus (Gantz et al 1993J Biol Chem 268: 15174-15179). This receptor is involved in central regulation of body weight: MC4-R is activated by alpha-melanocyte-stimulating hormone (MSH) derived from the precursor of opiomelanocortin, and agouti gene-related protein (AGRP) inactivates this receptor. alpha-MSH induces weight loss, however ectopic expression of the agouti protein leads to obesity in agouti mice (Fan et al 1993Nature 385: 165-168; Lu et al 1994Nature 371: 799-802). Additional evidence for the role of MC4-R in weight regulation comes from the gene knockout mouse model (Huszar et al 1997 Cell 88: 131-141) and the human haplotype deficiency mutation (Vaisse et al 1998NatGenet 20: 113-114; Yeo et al 1998NatGenet 20: 111-112; Hinney et al 1999J Clin Endocrinol Metab 84: 1483-1486). In MC 4-R-knockout mice, weight gain was observed in mice at 5 weeks of age. By 15 weeks, homozygous mutant female mice weighed twice as much as wild-type littermates, while homozygous mutant male mice weighed 50% more than wild-type control mice. MC4-R knockout heterozygous mice show an increase in body weight that is intermediate between that of wild type and homozygous mutant mice, thus demonstrating the gene dose effect of MC4-R ablation on body weight regulation. Homozygote mutant food intake was increased by-50% compared to the wild type relative (Huszar et al 1997 Cell 88: 131-141). [ from am.j.hum.genet., 65: 1501-1507, 1999]. Activation of MCR4 has been shown to induce penile erection in rodents, and inactivation of MCR4 can lead to obesity (reviewed in Hadley, 1999, Ann N Y Acad Sci, 885: 1-21, Wikberg et al 2000, Pharmacol Res., 42 (393), 420).
In Drugs Of The Future, 2004, 29 (10): 1065-1074, Chaki and Nakazato indicated potential therapeutic applications for ligands acting at the MC4 receptor. International patent application publication Nos. WO2005/077935, WO02/068387 and WO02/068388, and International patent application PCT/IB2006/002151 relate to specific piperidinyl carbonyl pyrrolidine MC4 agonists that are useful for treating sexual dysfunction, obesity, diabetes and other diseases. The above disclosure is incorporated herein by reference in its entirety for the therapeutic aspects of the MC4 agonists of the present invention.
The compounds of the invention are useful for treating diseases, disorders, or conditions responsive to activation of the MC4 receptor, including:
male and female sexual dysfunction, including hypoactive sexual desire disorder, sexual arousal disorder, female orgasmic disorder and/or dyspareunia disorder, male erectile dysfunction;
obesity (by reducing appetite, increasing metabolic rate, reducing fat intake, or reducing carbohydrate craving); and
diabetes (by increasing glucose tolerance and/or decreasing insulin resistance).
The compounds of the invention are useful in the treatment of other diseases, disorders or conditions including, but not limited to, hypertension, hyperlipidemia, osteoarthritis, cancer, gallbladder disease, sleep apnea, depression, anxiety, obsessive-compulsive disorders, neuropathy, insomnia/sleep disorders, drug abuse, pain, fever, inflammation, immunomodulation, rheumatoid arthritis, tanning of the skin, acne and other skin disorders, neuroprotection and enhanced cognitive memory including treatment of alzheimer's disease, treatment of lower urinary tract dysfunction (including urinary incontinence-overactive bladder, increased daytime frequency, nocturia, urgency, urinary incontinence (with involuntary leak conditions) including stress urinary incontinence, urge urinary incontinence and mixed urinary incontinence, overactive bladder with urinary incontinence, enuresis, nocturnal enuresis, persistent urinary incontinence, contextual urinary incontinence e.g. during sexual intercourse, And Lower Urinary Tract Symptoms (LUTS) associated with Benign Prostatic Hyperplasia (BPH), and other indications mentioned in the above-referenced patent applications.
The compounds of the invention are particularly suitable for the treatment of female sexual dysfunction, male erectile dysfunction, obesity, diabetes and conditions of lower urinary tract dysfunction.
The term "treatment" as used herein includes both prevention and control, i.e., the prevention and palliative treatment of a given condition.
Desirable properties of the compound MCR4 agonists of the invention include: desirable MCR4 agonist potency, as detailed below; selectivity for MCR4 agonism as compared to MCR1, and/or MCR5, and/or MCR3, as detailed below; desirable MC4R agonist potency and selectivity for MCR4 as compared to MCR1, and/or MCR5, and/or MCR 3; good biopharmaceutical properties, such as physical stability; solubility; oral bioavailability; suitable metabolic stability; ability to substitute AGRP from MC4 receptor.
The present invention provides compounds of formula (I), and pharmaceutically acceptable salts, solvates (including hydrates), and prodrugs thereof,
wherein
One of X and Y is N and the other is CH,
r is F, Cl, CN, CF3Or methoxy, with the proviso that when Y is N, R is not F or Cl,
R1is phenyl, 2-pyridyl, C3-C6Cycloalkyl or CH2(C3-C6Cycloalkyl) wherein the cyclic moiety is optionally substituted with one or more substituents independently selected from the group consisting of F, Cl, CN, methyl and methoxy,
R2h, F or Cl, with the proviso that when Y is N, R2Instead of F or Cl, the catalyst is,
het is a 6-membered ring containing 1 or 2N atoms, wherein the ring is aromatic, or contains 2 double bonds and an ═ O substituent in the ring, the ring being optionally substituted with one or more substituents independently selected from F, Cl, OH, CN, methyl, ethyl, NH2、NHCH3、N(CH3)2And a methoxy group,
alternatively, Het is a 6-membered ring containing 1 or 2N atoms, which 6-membered ring is fused in the 3, 4-position relative to the attachment to the pyrrolidine ring to a 5-membered aromatic ring containing one or two other N atoms, which 5-membered ring is optionally substituted with OH.
Non-limiting examples of suitable "Het" groups are shown below:
preferably X is N and Y is CH.
Preferably R is chloro.
Preferably R1Is phenyl optionally substituted with one or more substituents independently selected from F, Cl, CN, methyl and methoxy.
More preferably R1Is phenyl, 4-chlorophenyl or 4-fluorophenyl.
In another embodiment, R1Preferably C3-C6Cycloalkyl, more preferably cyclopropyl or cyclohexyl.
Preferably R2Is H or F.
More preferably R2Is H.
Preferably Het is pyridin-2-yl, pyridin-3-yl, pyridazin-3-yl, 6-oxo-1, 6-dihydropyridazin-3-yl, 6-oxo-1, 6-dihydropyridin-3-yl, 2-oxo-1, 2-dihydropyrimidin-4-yl, 6-oxo-1, 6-dihydropyrimidin-4-yl, 2-oxo-1, 2-dihydropyridin-4-yl, imidazo [1, 2-b ] pyridazin-6-yl, [1, 2, 4] triazolo [4, 3-b ] pyridazin-6-yl or 6-oxo-1, 6-dihydropyridin-2-yl, optionally substituted with one or more substituents independently selected from F, Cl, OH, CN, methyl, ethyl and methoxy.
More preferably Het is pyridin-2-yl, pyridin-3-yl, pyridazin-3-yl or 6-oxo-1, 6-dihydropyridazin-3-yl, optionally substituted with one or more substituents independently selected from OH, CN, F, methyl and methoxy.
More preferably Het is pyridin-2-yl or pyridazin-3-yl each substituted at the para position relative to the bond to the pyrrolidine moiety by OH, CN or methoxy.
Most preferably Het is pyridazin-3-yl substituted in the para position relative to the bond to the pyrrolidine moiety with OH, CN or methoxy.
Preferred compounds, salts, solvates and prodrugs include those wherein:
R1has a value (value) related to the specific compound described below.
Preferred compounds, salts, solvates and prodrugs include those wherein:
r has a value related to the specific compound described below.
Preferred compounds, salts, solvates and prodrugs include those wherein:
R2have values associated with the specific compounds described below.
Preferred compounds, salts, solvates and prodrugs include those wherein:
het has values related to the specific compounds described below.
Other preferred compounds, salts, solvates and prodrugs include those wherein:
R、R1、R2and Het has values associated with the specific compounds described below.
Preferably the compound is selected from:
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4-hydroxy-3, 5-dimethyl-4-phenylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- (5-Chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4-hydroxy-3, 5-dimethyl-4-pyridin-2-ylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4-hydroxy-3, 5-dimethyl-4-phenylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4-hydroxy-3, 5-dimethyl-4-phenylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- { [ (3R, 4R, 5S) -4- (4-chlorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } -4- (5-chloropyridin-2-yl) pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-Chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (5-Chloropyridin-2-yl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (3, 4-difluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4-hydroxy-4- (4-methoxyphenyl) -3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4-cyclohexyl-4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
(3R, 4R, 5S) -1- { [ (3S, 4S) -1- (6-chloropyridazin-3-yl) -4- (5-chloropyridin-2-yl) pyrrolidin-3-yl ] carbonyl } -4- (4-fluorophenyl) -3, 5-dimethylpiperidin-4-ol;
(3R, 4R, 5S) -1- { [ (3S, 4S) -4- (5-chloropyridin-2-yl) -1- (6-methoxypyridazin-3-yl) pyrrolidin-3-yl ] carbonyl } -4-cyclopropyl-3, 5-dimethylpiperidin-4-ol;
(3R, 4R, 5S) -1- { [ (3S, 4S) -4- (5-chloropyridin-2-yl) -1- (5-fluoropyridin-3-yl) pyrrolidin-3-yl ] carbonyl } -4- (4-fluorophenyl) -3, 5-dimethylpiperidin-4-ol;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -3-cyanopyridine;
6- [ (3S, 4S) -3- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } -4- (5-fluoropyridin-2-yl) pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- (5-fluoropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4-hydroxy-3, 5-dimethyl-4-phenylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- { [ (3R, 4R, 5S) -4- (4-chlorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } -4- (5-fluoropyridin-2-yl) pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- { [ (3R, 4R, 5S) -4- (4-chlorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } -4- (5-fluoropyridin-2-yl) pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-cyanopyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4R) -3- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } -4- (6-methoxypyridin-3-yl) pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } -4- (5-methoxypyridin-2-yl) pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
(3R, 4R, 5S) -4- (4-fluorophenyl) -1- { [ (3S, 4R) -1- (5-fluoropyridin-3-yl) -4- (6-methoxypyridin-3-yl) pyrrolidin-3-yl ] carbonyl } -3, 5-dimethylpiperidin-4-ol;
(3R, 4R, 5S) -1- { [ (3S, 4S) -4- (5-chloropyridin-2-yl) -1-pyridazin-3-ylpyrrolidin-3-yl ] carbonyl } -4- (4-fluorophenyl) -3, 5-dimethylpiperidin-4-ol;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
(3R, 4R, 5S) -1- { [ (3S, 4S) -4- (5-chloropyridin-2-yl) -1- [1, 2, 4] triazolo [4, 3-b ] pyridazin-6-ylpyrrolidin-3-yl ] carbonyl } -4- (4-fluorophenyl) -3, 5-dimethylpiperidin-4-ol;
(3R, 4R, 5S) -1- { [ (3S, 4S) -4- (5-chloropyridin-2-yl) -1-imidazo [1, 2-b ] pyridazin-6-ylpyrrolidin-3-yl ] carbonyl } -4- (4-fluorophenyl) -3, 5-dimethylpiperidin-4-ol;
4- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyrimidin-2 (1H) -one;
4- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -1-methylpyrimidin-2 (1H) -one;
(3R, 4R, 5S) -1- { [ (3S, 4S) -4- (5-chloropyridin-2-yl) -1- (6-methoxypyridazin-3-yl) pyrrolidin-3-yl ] carbonyl } -4- (4-fluorophenyl) -3, 5-dimethylpiperidin-4-ol;
6- [ (3S, 4S) -3- (5-chloro-3-fluoropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- (5-chloro-3-fluoropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidine, 1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-chloro-3-fluoropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (3, 5-difluoropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- (3, 5-difluoropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (3, 5-difluoropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- { [ (3R, 4R, 5S) -4- (4-chlorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } -4- (5-chloropyridin-2-yl) pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- (5-Chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (3, 4-difluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- { [ (3R, 4R, 5S) -4- (4-chlorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } -4- (5-chloropyridin-2-yl) pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-Chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (3, 4-difluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
and pharmaceutically acceptable salts, solvates (including hydrates), and prodrugs thereof.
More preferably the compound is selected from:
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4-hydroxy-3, 5-dimethyl-4-phenylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4-hydroxy-3, 5-dimethyl-4-phenylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4-hydroxy-3, 5-dimethyl-4-phenylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- { [ (3R, 4R, 5S) -4- (4-chlorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } -4- (5-chloropyridin-2-yl) pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-Chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (5-Chloropyridin-2-yl) -4-hydroxy-3, 5-dimethylpiperidine, 1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (3, 4-difluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
(3R, 4R, 5S) -1- { [ (3S, 4S) -4- (5-chloropyridin-2-yl) -1-pyridazin-3-ylpyrrolidin-3-yl ] carbonyl } -4- (4-fluorophenyl) -3, 5-dimethylpiperidin-4-ol;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
(3R, 4R, 5S) -1- { [ (3S, 4S) -4- (5-chloropyridin-2-yl) -1- [1, 2, 4] triazolo [4, 3-b ] pyridazin-6-ylpyrrolidin-3-yl ] carbonyl } -4- (4-fluorophenyl) -3, 5-dimethylpiperidin-4-ol;
(3R, 4R, 5S) -1- { [ (3S, 4S) -4- (5-chloropyridin-2-yl) -1-imidazo [1, 2-b ] pyridazin-6-ylpyrrolidin-3-yl ] carbonyl } -4- (4-fluorophenyl) -3, 5-dimethylpiperidin-4-ol;
4- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyrimidin-2 (1H) -one;
4- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -1-methylpyrimidin-2 (1H) -one;
6- [ (3S, 4S) -3- (5-chloro-3-fluoropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- (5-chloro-3-fluoropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-chloro-3-fluoropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (3, 5-difluoropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- (3, 5-difluoropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (3, 5-difluoropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- { [ (3R, 4R, 5S) -4- (4-chlorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } -4- (5-chloropyridin-2-yl) pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- (5-Chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (3, 4-difluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- { [ (3R, 4R, 5S) -4- (4-chlorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } -4- (5-chloropyridin-2-yl) pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-Chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (3, 4-difluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
and pharmaceutically acceptable salts, solvates (including hydrates), and prodrugs thereof.
Most preferably the compound is selected from:
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- { [ (3R, 4R, 5S) -4- (4-chlorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } -4- (5-chloropyridin-2-yl) pyrrolidine, 1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-Chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (5-Chloropyridin-2-yl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (3, 4-difluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine, 3(2H) -one;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- { [ (3R, 4R, 5S) -4- (4-chlorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } -4- (5-chloropyridin-2-yl) pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- (5-Chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (3, 4-difluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- { [ (3R, 4R, 5S) -4- (4-chlorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } -4- (5-chloropyridin-2-yl) pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-Chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (3, 4-difluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
and pharmaceutically acceptable salts, solvates (including hydrates), and prodrugs thereof.
Pharmaceutically acceptable salts of the compounds of formula (I) include acid addition salts thereof. Suitable acid addition salts are formed from acids which form non-toxic salts. Examples include acetate, adipate, aspartate, benzoate, benzenesulfonate, bicarbonate/carbonate, bisulfate/sulfate, borate, camphorsulfonate, citrate, cyclamate, edisylate, ethanesulfonate, formate, fumarate, glucoheptonate, gluconate, glucuronate, hexafluorophosphate, oxybenzoate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide, isethionate, lactate, malate, maleate, malonate, methanesulfonate, methylsulfate, naphthenate (naphylate), 2-naphthalenesulfonate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate (pamoate), phosphate/biphosphate/dihydrogen phosphate, dihydrogenphosphate, citrate, cyclamate, dihydrogensulfonate, Pyroglutamate, saccharinate, stearate, succinate, tannate, tartrate, tosylate, trifluoroacetate and xinafoate (xinofoate). Hemisalts of acids (hemisalts), such as hemisulfate, may also be formed. For an overview of suitable Salts, see Handbook of Pharmaceutical Salts: properties, Selection, and Usby Stahl and Wermuth (Wiley-VCH, 2002).
The compounds, salts, solvates and prodrugs of the present invention may exist in the form of tautomers (tautomeric), zwitterions, polymorphs, crystals, liquid crystals (liquid crystals), and the like. All such forms are included within the scope of the present invention. Examples of tautomeric relationships (compounds exemplified by the "Het" group) the "keto" and "enol" tautomers are included within the scope of "Het" for compounds of formula (I):
also included within the scope of the invention are multi-component complexes (in addition to salts and solvates) in which the drug and at least one other component are present in stoichiometric or non-stoichiometric amounts. Such complexes include clathrates (drug-host containing complexes) and co-crystals (co-crystals). The latter is generally defined as a crystalline complex of neutral molecular (neutral molecular) components linked together by non-covalent interactions, but may also be a complex of a neutral molecule and a salt.
Also included within the scope of the present invention are isotopically-labeled compounds of formula (I), for example, wherein2H、3H、13C、15N、18O or other isotopes, which can be prepared by appropriate variations of the synthetic methods described herein using methods and reagents known in the art or variations thereof.
As shown herein, so-called 'prodrugs' of the compounds of formula (I) are within the scope of the present invention. Thus, when some derivatives of the compounds of formula (I), which may themselves have little or no pharmacological activity, are administered into or onto the body, they can be converted into compounds of formula (I) having the desired activity, for example by hydrolytic cleavage. These derivatives are referred to as 'prodrugs'. For further information on the use of prodrugs seePro-drugs as Novel Delivery SystemsVol.14, ACS symposium series (T Higuchi and W Stella) andBioreversible Carriers in Drug Design,PergamonPress,1987(Ed.E B Roche,American Pharmaceutical Association)。
some of the compounds of formula (I) may themselves also act as prodrugs of other compounds of formula (I).
Conventional techniques for the preparation/separation of individual enantiomers include chiral synthesis from precursors suitable for optical purity or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral High Performance Liquid Chromatography (HPLC).
Alternatively, the racemate (or a racemic precursor) may be reacted with a suitable optically active compound, for example an alcohol, or, in the case of compounds of formula (I) containing an acidic or basic moiety, an acid or base such as tartaric acid or 1-phenylethylamine. The resulting diastereomeric mixtures can be separated by chromatography and/or fractional crystallization, or the two diastereomers can be converted into the corresponding pure enantiomer or enantiomers by methods known to the skilled artisan.
The chiral compounds of the invention (and chiral precursors thereof) can be obtained in enantiomerically enriched form using chromatography (typically HPLC) on an asymmetric resin with a mobile phase consisting of a hydrocarbon (typically heptane or hexane) containing 0 to 50% by volume isopropanol (typically 2 to 20%) and possibly 0 to 5% by volume alkylamine. The concentration of the eluent provides an enriched mixture. The absolute composition of the mobile phase will depend on the chiral stationary phase (asymmetric resin) selected.
The following route, which is included in the examples and preparations, represents a method of synthesizing the compounds of formula (I). The skilled person will appreciate that the compounds of the invention, and their intermediates, may be prepared by methods other than those specifically described herein, for example by variations of the methods described herein, for example methods known in the art. For suitable teachings of synthesis, functional group transformations, use of protecting groups, and the like, see, for example:
"Comprehensive Organic Transformations", RC Larock, VCH publishers Inc. (1989); advanced Organic Chemistry "by J.March, Wiley Interscience (1985); "design Organic Synthesis" by S Warren, Wiley Interscience (1978); "Organic Synthesis-The Disconnection apparatus", S Warren, WileyInetrscience (1982); "Guidebook to Organic Synthesis", RK Mackie and DMSmith, Longman (1982); "Protective Groups in Organic Synthesis", TW Greene and PGM Wuts, John Wiley and Sons, Inc. (1999); and "protecting Groups", PJ, Kocienski, Georg Thieme Verlag (1994); and any improved version of the standard method.
In the following general synthetic methods, the substituent R, R is shown unless otherwise indicated1、R2X, Y and Het are as above described for compounds of formula (I)And (4) defining.
The following pathway represents a method for synthesizing the compound of formula (I). The skilled person will appreciate that other equivalent methods may also be used.
Scheme 1 shows the preparation of compounds of formula (I) by peptide coupling of intermediates (II) and (III), if desired with the addition of a suitable base and/or additive (e.g. 1-hydroxybenzotriazole hydrate or 4-dimethylaminopyridine).
Scheme 1
Alternatively, the conditions used include stirring a solution of the piperidine of formula (II) and the acid of formula (III) together with 1- (3-dimethylaminopropyl) -3-ethyl-carbodiimide hydrochloride (EDCI), triethylamine or N-methylmorpholine, and 1-hydroxybenzotriazole hydrate (HOBt) in Dimethylformamide (DMF), Tetrahydrofuran (THF), Dichloromethane (DCM) or ethyl acetate (EtOAc) at room temperature. Other alternative suitable methods are stirring of the intermediate compound of formula (II) and the reaction of formula (III) with O-benzotriazol-1-yl-N, N, N ', N' -tetramethyluronium Hexafluorophosphate (HBTU) or 1-propylphosphonic acid cyclic anhydride in CH2Cl2Or a solution in EtOAc. Any suitable inert solvent can be used for the above conditions, wherein inert solvent refers to a solvent that does not contain a carboxylic acid or a primary or secondary amine. At least 1 equivalent of each coupling reagent should be used, and an excess of one or both coupling reagents may be used if desired.
According to another embodiment, the present invention provides novel intermediate compounds of formula (III).
Scheme 2 illustrates another route to compounds of general formula (I) having a series of Het groups by methods employing protecting groups.
Scheme 2
PG is a suitable nitrogen-protecting group.
The novel compounds of formulae (IV), (V) and (VI) are another embodiment of the present invention.
In scheme 2, the amine intermediate of formula (II) and the protected pyrrolidinoic acid intermediate of formula (IV) are coupled using standard peptide coupling procedures described in scheme 1 above to prepare a coupled and protected intermediate of formula (V), and the nitrogen protecting group is removed from formula (V) using standard deprotection protocols to afford the compound of formula (VI). Any suitable nitrogen Protecting group may be used (e.g., "Protecting Groups in organic Synthesis" 3)rdEdition t.w.greene and p.g.wuts, Wiley-Interscience, 1999). Commonly used nitrogen Protecting Groups (PG) suitable for use herein include tert-butoxycarbonyl (t-Boc), which is conveniently removed by treatment with an acid (e.g. trifluoroacetic acid or hydrochloric acid) in an organic solvent (e.g. dichloromethane or 1, 4-dioxane); and benzyl groups, which are removed by hydrogenation in the presence of a suitable catalyst or by treatment with 1-chloroethyl chloroformate.
A "Het" group (where "Het" is heteroaryl) may be introduced by replacement of a suitable leaving group, for example by replacement of a suitable leaving group from a heteroaromatic precursor of the formula "Het-L" where L is a suitable leaving group. Suitable leaving groups include halogen. In some cases, a transition metal catalyst (e.g., palladium, copper) may be desired, optionally in combination with a phosphine ligand (e.g., 1 '-dinaphthalene-2, 2' diylbiphenylphosphine) to give the desired coupled product.
Alternatively, a compound of formula (I) having a particular Het group may be converted to other compounds of formula (I) having a different Het group. For example:
i) compounds of formula (Ia) as depicted in scheme 3 wherein Het contains a suitable leaving group L, such as methoxy or chloro, may be converted to compounds of formula (Ib) by hydrolysis under acidic or basic conditions as depicted in scheme 3. Superior foodAcidic conditions are selected and it is especially preferred to treat the compound of formula (Ia) with acetic acid at reflux temperature. Alternatively, compounds of formula (Ia) wherein L is chloro may be reacted with a compound of formula Z-O-Wherein Z is a suitable oxygen protecting group, to give an intermediate of formula (Ia) wherein L is OZ. Deprotection then provides compounds of formula (Ib). For example, when Z ═ benzyl, it can be removed by hydrogenation in the presence of a suitable catalyst.
Scheme 3
ii) Compounds of formula Ic as described in scheme 4 (wherein Het is as described in scheme 4 and R3Is H) can be converted into compounds of the formula Id, in which R is3Is methyl or ethyl. Suitable bases include sodium hydride, lithium diisopropylamide (lithium diisopropylamide) and sodium hexamethyldisilazide, suitable alkylating agents include methyl iodide, methyl tosylate, dimethyl sulfate and ethyl iodide, and suitable solvents include tetrahydrofuran, dimethylformamide and N-methyl-2-pyrrolidone. Optional additives, such as lithium salts, e.g., lithium bromide, may also be present in the reaction mixture.
Scheme 4
Scheme 5 illustrates a route for the preparation of a pyrrolidinic acid intermediate of formula (IV) from an unsaturated intermediate of formula (VII).
Scheme 5
PG is a suitable nitrogen protecting group. PG (Picture experts group)2For suitable carboxylic acid protectionAnd (4) a base. The compounds of formulae (VIII), (IX), (X), (XI) and (XII) are either commercially available or known to those skilled in the literature and/or in the art relating to the preparation and/or the literature described herein.
The compounds of formula (VII) may be prepared by olefination of the aldehyde intermediate of formula (XI) to predominantly the desired trans isomer by Wittig or similar methods using a suitable ylide such as the methyl (triphenylphosphoranylidene) acetate or phosphonate anion, e.g. deprotection from trimethylphosphonoacetate.
There are many alternatives in the literature for the preparation of unsaturated intermediates of general formula (VII) including the protection of the precursor cinnamic acid derivative (VIII) using standard methods or Heck reaction of an aromatic halide (IX) with a suitable acrylate derivative (X), e.g. tert-butyl acrylate, in the presence of a palladium catalyst and a suitable base, e.g. triethylamine.
The resulting E-olefin intermediate of formula (VII) will undergo a [3+2] -azomethine ylide cycloaddition by reaction with a ylide precursor of formula (XII) to give pyrrolidine, almost exclusively in trans stereochemistry. The reaction requires an inert solvent, such as dichloromethane or toluene or tetrahydrofuran, and activation by one or more of the following conditions: (1) acid catalysts, such as TFA; (2) desilication alkylating agents, such as silver fluoride; (3) and (4) heating.
The compounds of the general formula (XIII) obtained from the cycloaddition reaction are racemates and can be resolved to give their constituent enantiomers (constitutive enantiomers) which can be achieved by preparative HPLC using a chiral stationary phase. Alternatively, the acid intermediate of formula (IV) may be resolved by standard methods, for example by reaction with an enantiomerically pure reagent to form a diastereomeric derivative, and the resulting diastereomers separated by physical means, followed by a leaving to give acid (IV).
The intermediate compounds of formula (XIII) may be protected by the protecting group PG2To a compound of general formula (IV). A number of methods can be used to effect this transformation (see Advanced organic chemistry: reactions, Mechanisms, and Structure, Fourth edition, March, Jerry, 1992, pp378-383, Wiley, New York, N.Y. USA). In particular, for base labile protecting groups, treatment of a compound of formula (XIII) with an aqueous solution of an alkali metal hydroxide, such as lithium, sodium or potassium hydroxide, in a suitable organic solvent affords the corresponding compound of formula (IV). Preferably, water-miscible organic co-solvents (e.g. 1, 4-dioxane or tetrahydrofuran) may also be used in such reactions. The reaction may be heated to assist hydrolysis if desired. Under acidic conditions, some protecting groups are more easily hydrolyzed, such as tert-butyl or benzhydryl esters (benzylesters). These esters can be treated with anhydrous acids such as trifluoroacetic acid or hydrochloric acid in inert organic solvents such as dichloromethane.
Novel compounds of formula (XIII) are another embodiment of the invention.
Scheme 6 illustrates another route to prepare the individual enantiomers of the pyrrolidinic acid intermediate of formula (IV) from the unsaturated intermediate of formula (VII) using oxazolidinone as a chiral auxiliary. The acid of formula (VIII) may be obtained by deprotection of (VII), then conversion to a mixed anhydride, then coupling with an oxazolidinone (wherein preferably R4Phenyl, tert-butyl or isopropyl) to give an intermediate of formula (XIV). Alternatively, a compound of formula (VII) (e.g. when PG is2═ OCOt-Bu) with a lithium salt of oxazolidinone in a suitable solvent such as THF to give compounds of formula (XIV).
The compound of formula (XIV) will undergo [3+2] by reaction with a compound of formula (XII)]-ylide cycloaddition of azomethine to give the diastereomers (XV) and (XVI), which can be separated by chromatography or crystallization and then hydrolyzed to give the pyrrolidine of formula (IV). Alternatively, the compound of formula (XVI) may be converted to a compound of formula (XIII), for example by treatment with sodium methoxide in dimethyl carbonate (when PG is present)2OMe).
Scheme 6
PG is selected from suitable nitrogen protecting groups. PG (Picture experts group)2Selected from suitable carboxylic acid protecting groups, and different groups are used for compounds (VII) and (XIII).
The novel compounds of formula (XVI) are another embodiment of the present invention.
Scheme 7 illustrates a route to the pyrrolidine acid intermediate of general formula (III) from the intermediate of general formula (XIII). Once the protecting group PG is removed using any suitable conventional technique, the Het group can be introduced by a suitable method as described in scheme 2. The acid protecting group PG is then removed as described in scheme 42To obtain the acid of the general formula (III).
Scheme 7
PG is selected from suitable nitrogen protecting groups. PG (Picture experts group)2Selected from suitable carboxylic acid protecting groups.
The novel compounds of formula (XVII) and (XVIII) are another embodiment of the present invention.
As depicted in scheme 8, piperidine intermediates of general formula (II) may be prepared by addition of an organometallic nucleophile to a ketone of general formula (XIX) containing a suitable nitrogen Protecting Group (PG) to give intermediates of general formula (XX). The stereochemistry of the addition is preferably such that the hydroxyl groups in the product are cis to the two methyl groups. Controlled addition to carbonyl systems, such as the one described, has been described in the literature (e.g.journal of medicinal chemistry (1964), 7(6), pp 726-8). These nucleophilic additions are typically carried out at low temperatures in anhydrous ethers or other non-polar solvents using grignard reagents, organolithium reagents or other suitable organometallic reagents. These organometallic reagents may be prepared by using suitable halide precursors (R)1-Br or R1-I) and n-butyllithium or tert-butyllithiumHalogen-metal exchange preparation of (a). Suitable protecting groups include benzyl, which is removed by hydrogenation; or Boc, which is removed by treatment with an acid such as TFA; or p-methoxybenzyl (PMB) which is removed by treatment with DDQ, CAN or 1-chloroethyl chloroformate to give the desired piperidine intermediate of formula (II). For some protecting groups, and under certain conditions, the protecting group is not stable to treatment with organometallic reagents, and both transformations can be achieved in one step, for example when PG ═ Boc, this protecting group can sometimes be left off when the intermediate of formula (XIX) is treated with organometallic reagents.
Scheme 8
PG is selected from suitable nitrogen protecting groups.
The skilled person will appreciate that in addition to protecting nitrogen or acid groups, as described above, in many cases during the synthesis of compounds of formula I, it is desirable to protect other groups, for example, a hydroxy group with a suitable protecting group, and then remove the protecting group. The method of deprotection of any particular group will depend on the protecting group. For examples of protection/deprotection methodologies see "Protective groups in Organic synthesis", TW Greene and PGM Wutz. For example, where the hydroxy group is protected as a methyl ether, the deprotection conditions may for example comprise refluxing in 48% aqueous HBr, or by stirring with boron tribromide in dichloromethane. Alternatively, where the hydroxyl group is protected as a benzyl ether, the deprotection conditions may, for example, comprise hydrogenation with a palladium catalyst in a hydrogen atmosphere.
The preparation of all of the above reactions and novel starting materials used in the above processes is conventional and suitable reaction conditions for their performance or preparation and the method of isolating the desired product will be well known to those skilled in the art relating to literature methods and examples and preparations herein.
Pharmaceutically acceptable salts of the compounds of formula (I) can be prepared rapidly by mixing together a solution of the compound of formula (I) and optionally an acid. The salt may precipitate out of solution and be collected by filtration, or may be recovered by evaporation of the solvent.
Pharmaceutically acceptable salts of the compounds of formula (I) may be prepared by one or more of the following three methods:
(i) reacting a compound of formula (I) with a desired acid;
(ii) removal of acid-or base-labile protecting groups from suitable precursors of compounds of formula (I), or by ring-opening of suitable ring precursors, such as lactones or lactams, using a desired acid; or
(iii) One salt of the compound of formula (I) is converted to another by reaction with a suitable acid or by a suitable ion exchange column.
All three reactions are usually carried out in solution. The resulting salt may precipitate out and be collected by filtration, or may be recovered by evaporation of the solvent. The degree of ionization in the resulting salt may vary from completely ionized to almost non-ionized.
The compounds of formula (I) of the present invention are useful as MCR4 agonists for the treatment of various disease states. Preferably, the MCR4 agonist exhibits functional potency (functional potency), expressed as EC, at the MC4 receptor50Less than about 1000nM, more preferably less than 500nM, yet more preferably less than about 100nM, and even more preferably less than about 50nM, where the EC of the functional potency of MCR450The measurement can be carried out using method E in International patent application publication No. WO 2005/077935. Using this approach, the compounds according to the invention exhibit functional potency at the MC4 receptor, expressed as EC50And less than about 1000 nM.
Preferred compounds herein exhibit functional potency at the MC4 receptor as defined herein before, and selectivity for MCR4 over MCR 1. Preferably, the MCR4 agonist is more selective for MCR4 than MCR1, wherein the MCR4 receptor agonist is at least about 10-fold, at least about 20-fold, more preferably at least about 30-fold, even more preferably at least about 100-fold, particularly more preferably at least about 300-fold, even more preferably at least about 500-fold, particularly at least about 1000-fold more functionally selective for MCR4 receptor than for MCR1 receptor, wherein the relative selectivity assessment is based on an assay for functional potency of MCR1 and MCR4 using the methods described herein.
Preferably, the MCR4 agonist is more selective for MCR4 than MCR3, wherein the MCR4 receptor agonist is at least about 10-fold, preferably at least about 30-fold, more preferably at least about 100-fold, more preferably to at least about 300-fold, even more preferably at least about 500-fold, especially at least about 1000-fold more functionally selective for MCR4 receptor than for MCR3 receptor, wherein the relative selectivity assessment is based on an assay of functional potency of MCR3 and MCR4 using the methods described herein.
Preferred compounds herein exhibit functional potency at the MCR4 receptor as defined herein before, and selectivity for MCR4 over MCR 5. Preferably, the MCR4 agonist is more selective for MCR4 than MCR5, wherein the MCR4 receptor agonist is at least about 10-fold, preferably at least about 30-fold, more preferably at least about 100-fold, more preferably at least about 300-fold, even more preferably at least about 500-fold, particularly at least about 1000-fold more functionally selective for MCR4 receptor than for MCR5 receptor, wherein the relative selectivity assessment is based on an assay of functional potency of MCR5 and MCR4 using the methods described herein.
Preferably, the MCR4 agonist is more selective for MCR4 than MCR1 and MCR3, wherein the MCR4 receptor agonist is at least about 10-fold, preferably at least about 30-fold, more preferably at least about 100-fold, more preferably at least about 300-fold, even more preferably at least about 1000-fold, selective for the MCR4 receptor as compared to the MCR1 and MCR3 receptors.
Preferred compounds herein exhibit functional potency at the MCR4 receptor as defined herein before and selectivity for MCR4 over MCR1 and MCR 5. Preferably, the MCR4 agonist is more selective for MCR4 than MCR1 and MCR5, wherein the MCR4 receptor agonist is at least about 10-fold, preferably at least about 30-fold, more preferably at least about 100-fold, more preferably at least about 300-fold, even more preferably at least about 500-fold, particularly at least about 1000-fold, selective for MCR1 and MCR5 receptors functionally for MCR4 receptors.
Preferably, the MCR4 agonist MCR4 is more selective than MCR3 and MCR5, wherein the MCR4 receptor agonist is at least about 10-fold, preferably at least about 30-fold, more preferably at least about 100-fold, more preferably at least about 300-fold, most preferably at least about 1000-fold selective for the MCR4 receptor as compared to the MCR3 and MCR5 receptors.
Combination therapy
The compounds of formula (I) or their salts, solvates or prodrugs of the invention may be used for the delivery of a combination of active agents that is therapeutically adjunctive to conditions of interest, such as sexual dysfunction, lower urinary tract disorders, obesity and/or diabetes. Moreover, in some cases, the compounds of formula (I) of the present invention or their salts, solvates, or prodrugs can be used for delivery in combination with effective active agents that help alleviate emesis. Some suitable co-effective active agents for use in the compositions of the present invention include:
1) compounds which modulate the action of natriuretic factors, especially atrial natriuretic factor (also known as atrial natriuretic peptide), natriuretic factors of type B and C, such as inhibitors or neutral endopeptidases and especially the compounds described and claimed in WO02/02513, WO02/03995, WO02/079143 and EP- cA-1258474, and especially the compound (2S) -2{ [1- {3-4 (-chlorophenyl) propyl ] amino } carbonyl) -cyclopentyl ] methyl } -4-methoxybutanoic acid of example 22 of WO 02/079143;
2) angiotensin converting enzyme inhibiting compounds such as enalapril, and combined inhibitors of angiotensin converting enzyme and neutral endopeptidase such as omatrazole;
3) substrates for NO-synthase, such as L-arginine;
4) cholesterol lowering agents, e.g. statins (e.g. atorvastatin/Lipitor)TM) And fibrates (fibrates);
5) estrogen receptor modulators and/or estrogen agonists and/or estrogen antagonists, preferably raloxifene or lasofoxifene ((-) -cis-6-phenyl-5- [4- (2-pyrrolidin-1-yl-ethoxy) -phenyl ] -5, 6,7, 8-tetrahydronaphthalen-2-ol, and pharmaceutically acceptable salts thereof, formulations thereof as described in WO 96/21656);
6) a PDE inhibitor, more preferably a PDE2, 3, 4, 5,7 or 8 inhibitor, preferably a PDE2 or PDE5 inhibitor and most preferably a PDE5 inhibitor (see below), preferably with an IC50 of less than 100nM against the respective enzyme (provided that the PDE3 and 4 inhibitors are administered only locally or by injection into the penis for the treatment of male erectile dysfunction);
7) vasoactive Intestinal Protein (VIP), VIP mimetics, VIP analogues, more preferably mediated by one or more VIP receptor subtypes VPAC1, VPAC or PACAP (pituitary adenylate cyclase activating peptide), one or more VIP receptor agonists or VIP analogues (such as Ro-125-1553) or VIP fragments, one or more alpha-adrenoceptor antagonists in combination with VIP (such as invigorp (vasoactive intestinal peptide and phentolamine preparation), Aviptadil (Aviptadil));
8) 5-hydroxytryptamine receptor agonists, antagonists or modulators, more preferably agonists, antagonists or modulators for the 5HT1A (including VML670[ WO02/074288] and flibanserin [ US2003/0104980]), 5HT2A, 5HT2C, 5HT3 and/or 5HT6 receptors, including those described in WO-09902159, WO-00002550 and/or WO-00028993;
9) testosterone replacement agents (including dehydroandrosterone), testosterone (such as Tostrelle)TM,LibiGelTM) Dihydrotestosterone or testosterone implant (Implant);
10) selective androgen receptor modulators, such as LGD-2226;
11) estrogen, estrogen and medroxyprogesterone or medroxyprogesterone acetate (MPA) (i.e., as a combination), or estrogen and methyltestosterone hormone replacement therapy (e.g., HRT, especially bemeili, Cenestin, Oestrofeminal, Equin, Estrace, noonkinide, eleste Solo, Estring, eastaderm TTS, eastaderm Matrix, dememectin, premelase, Preempro, Prempak, premix, este, estotex, Evitrest HS, tibolone);
12) modulators of norepinephrine, dopamine, and/or 5-hydroxytryptamine transport, such as bupropion, GW-320659;
13) a receptor agonist or modulator for oxytocin/vasopressin, preferably a selective oxytocin agonist or modulator;
14) agonists or modulators for dopamine receptors, preferably D3 or D4 selective agonists or modulators, such as apomorphine; and
15 antiemetics, e.g. 5-HT3An antagonist or a neurokinin-1 (NK-1) antagonist.
Suitable 5-HT3Antagonists include, but are not limited to, granisetron, ondansetron, tropisetron, ramosetron, palonosetron, indisetron, dolasetron, alosetron, and azasetron.
Suitable NK-1 antagonists include, but are not limited to, Aprepitant, casopiotant, epitant, cilapitant, netupitant, vestipitant, wofpitant, and 2- (R) - (1- (R) -3, 5-bis (trifluoromethyl) phenyl) ethoxy-4- (5- (dimethylamino) methyl-1, 2, 3-triazol-4-yl) methyl-3- (S) - (4-fluorophenyl) morpholine. See, for example, International patent application publication No. WO 2006/049933.
For use of the compounds of the present invention in the treatment of lower urinary tract dysfunction in particular, combinations with other agents may include, but are not limited to
● muscarinic acetylcholine receptor antagonists such as tolterodine;
● an alpha-adrenoceptor antagonist, in particular an alpha 1 adrenoceptor antagonist or an alpha 2 adrenoceptor antagonist;
● an alpha adrenergic receptor agonist or partial agonist, in particular an alpha 1 adrenergic receptor agonist or partial agonist, or an alpha 2 adrenergic receptor agonist or partial agonist;
● 5HT2C agonists (see WO 2004/096196);
● 5-hydroxytryptamine and norepinephrine reuptake inhibitor (SNRI);
norepinephrine Reuptake Inhibitors (NRIs), such as reboxetine, racemic or (S, S) -enantiomeric forms;
● capsaicin receptor (VR) antagonists, such as capsaicin;
● α 2 δ ligands, such as gabapentin or pregabalin;
● beta 3-adrenoceptor agonists;
● 5 a 5HT1a receptor antagonist or a 5HT1a receptor inverse agonist;
● prostanoid receptor antagonists, such as EP1 receptor antagonists.
With respect to the use of compounds of formula (I) in the treatment of obesity and related diseases, the compounds may also be used in combination with other anti-obesity agents. Suitable anti-obesity agents include cannabinoid 1(CB-1) receptor antagonists (e.g., rimonabant), apolipoprotein-B secretion/microsomal triglyceride transfer protein (apo-B/MTP) inhibitors (particularly gut (gut) selective MTP inhibitors such as edipatamide or dirlotamide), 11 beta-hydroxysteroid dehydrogenase-1 (11 beta-HSD type 1) inhibitors, peptide YY3-36And analogs thereof, cholecystokinin-A (CCK-A) agonists, monoamine reuptake inhibitors (e.g., sibutramine), sympathomimetics, beta 3 adrenoreceptor agonists, dopamine receptor agonists (e.g., bromocriptine), melanocyte-stimulating hormone receptor analogs, 5HT2c receptor agonists, melanin concentrating hormone antagonists, leptin (OB protein), leptin analogs, leptin receptor agonists, somatotropin neuropeptide antagonists, lipase inhibitors (e.g., tetrahydrolipin)Pravastatin, i.e. orlistat (orlistat), anorectic agents (e.g. bombesin agonists), neuropeptide-Y receptor antagonists (especially NPY-5 receptor antagonists), thyromimetics, dehydroepiandrosterone or analogs thereof, glucocorticoid receptor agonists or antagonists, orexin receptor antagonists, glucagon-like peptide-1 receptor agonists, ciliary neurotrophic factors (e.g. Axokine)TMCommercially available from Regeneron Pharmaceuticals, Inc., Tarrytown, NY and Procter&Gamble Company, Cincinnati, OH), human agomelatine-related protein (AGRP) inhibitors, grelin receptor antagonists, histamine 3 receptor antagonists or inverse agonists, neuregulin U receptor agonists, and the like. Other anti-obesity agents, including the preferred agents below, are well known to those skilled in the art or will be apparent from the disclosure herein. The compounds of the present invention may also be administered in combination with naturally occurring compounds that have the ability to lower plasma cholesterol levels. Such naturally occurring compounds are commonly referred to as nutraceuticals and include, for example, garlic extract, Hoodia plant extract, and niacin. Particularly preferred antiobesity agents are selected from CB-1 antagonists, gut (gut) selective MTP inhibitors, orlistat, sibutramine, bromocriptine, ephedrine, leptin, peptide YY3-36And analogs thereof, and pseudoephedrine. Preferably, the compounds and combination therapies of the present invention for the treatment of obesity and related conditions are administered in conjunction with exercise and a scientific diet. Preferred CB-1 antagonists include rimonabant (SR141716A, trade name Acomplia) described in U.S. Pat. No. 5,624,941TMAvailable from Sanofi-Synthelabo); and compounds described in us patent 5,747,524, 6,432,984 and 6,518,264; U.S. patent publications US2004/0092520, US2004/0157839, US2004/0214855, and US 2004/0214838; U.S. patent application serial No. 10/971599 filed on day 22 of month 10, 2004; and PCT patent publications WO02/076949, WO03/075660, WO04/048317, WO04/013120, and WO 04/012671. Preferred gut-selective MTP inhibitors include dirlotapede described in us patent 6,720,351; 4- (4- (4- (4- ((2- ((4-methyl-4H-1, 2, 4-triazol-3-ylthio) methyl) -2- (4-chlorophenyl) -1, 3-dioxolane (dioxolan) -4-yl) methoxy) phenyl) piperazin-1-yl) benzene described in U.S. Pat. Nos. 5,521,186 and 5,929,075-2-sec-butyl-2H-1, 2, 4-triazol-3 (4H) -one (R103757); and the inputapi described in us patent 6,265,431 (BAY 13-9952). Other representative anti-obesity agents for use in the combinations, pharmaceutical compositions and methods of the invention may be prepared using methods known to those of ordinary skill in the art, for example; sibutramine can be prepared according to the method described in U.S. patent No. 4,929,629; bromocriptine can be prepared as described in us patents 3,752,814 and 3,752,888; orlistat can be prepared according to the methods described in us patents 5,274,143, 5,420,305, 5,540,917 and 5,643,874; PYY can be prepared according to the procedures described in U.S. publication 2002/0141985 and WO03/0276373-36(including the like).
One of the preferences herein is the combination of a compound of the invention and one or more other therapeutic agents selected from the group consisting of: a PDE5 inhibitor; an NEP inhibitor; a D3 or D4 selective agonist or modulator; an estrogen receptor modulator and/or an estrogen agonist and/or an estrogen antagonist; testosterone replacement, testosterone or testosterone implant (imbplant); estrogen, estrogen and medroxyprogesterone or medroxyprogesterone acetate (MPA), or estrogen and methyltestosterone hormone replacement therapy.
A preferred combination for the treatment of MED is a combination of a compound of the invention and one or more PDE5 inhibitors and/or NEP inhibitors.
Preferred combinations for treating FSD are combinations of a compound of the invention and a PDE5 inhibitor, and/or a NEP inhibitor, and/or a D3 or D4 selective agonist or modulator, and/or an estrogen receptor modulator, an estrogen agonist, an estrogen antagonist, and/or a testosterone replacement drug, testosterone implant, and/or an estrogen, estrogen and medroxyprogesterone or medroxyprogesterone acetate (MPA), estrogen and methyltestosterone hormone replacement therapy drug.
A particularly preferred PDE5 inhibitor for use in the treatment of a combination product of MED or FSD is 5- [ 2-ethoxy-5- (4-methyl-1-piperazinylsulfonyl) phenyl ] -1-methyl-3-n-propyl-1, 6-dihydro-7H-pyrazolo [4, 3-d ] pyrimidin-7-one (sildenafil, especially as the citrate salt);
(6R, 12aR) -2, 3,6, 7, 12, 12 a-hexahydro-2-methyl-6- (3, 4-methylenedioxyphenyl) -pyrazino [2 ', 1': 6, 1] pyrido [3, 4-b ] indole-1, 4-dione (IC-351 or tadalafil);
2- [ 2-ethoxy-5- (4-ethyl-piperazin-1-yl-1-sulfonyl) -phenyl ] -5-methyl-7-propyl-3H-imidazo [5, 1-f ] [1, 2, 4] triazin-4-one (vardenafil); 5- (5-acetyl-2-butoxy-3-pyridinyl) -3-ethyl-2- (1-ethyl-3-azetidinyl) -2, 6-dihydro-7H-pyrazolo [4, 3-d ] pyrimidin-7-one; 5- (5-acetyl-2-propoxy-3-pyridyl) -3-ethyl-2- (1-isopropyl-3-azetidinyl) (azetidinyl)) -2, 6-dihydro-7H-pyrazolo [4, 3-d ] pyrimidin-7-one; 5- [ 2-ethoxy-5- (4-ethylpiperazin-1-ylsulfonyl) pyridin-3-yl ] -3-ethyl-2- [ 2-methoxyethyl ] -2, 6-dihydro-7H-pyrazolo [4, 3-d ] pyrimidin-7-one; 4- [ (3-chloro-4-methoxybenzyl) amino ] -2- [ (2S) -2- (hydroxymethyl) pyrrolidin-1-yl ] -N- (pyrimidin-2-ylmethyl) pyrimidine-5-carboxamide (carboxamide) (TA-1790); 3- (1-methyl-7-oxo-3-propyl-6, 7-dihydro-1H-pyrazolo [4, 3-d ] pyrimidin-5-yl) -N- [2- (1-methylpyrrolidin-2-yl) ethyl ] -4-propoxybenzenesulfonamide (DA8159) and pharmaceutically acceptable salts thereof.
Particularly preferred NEP inhibitors for the treatment of MED or FSD combination products are the compounds described in WO 02/079143.
Cross-reference herein to compounds contained in patents and patent applications that may be used in accordance with the present invention is made to therapeutically active compounds and specific examples as defined in the claims, particularly claim 1 (all incorporated by reference in their entirety).
If a combination of active agents is administered, they may be administered simultaneously, separately or sequentially in the same or different formulations.
Biological experiments
Melanocortin receptor agonist activity; selectivity is
Agonistic potency (EC) of compounds against melanocortin receptors type 1 and 3(MC 1 and MC3)
50
) Body of
And (4) external measurement.
Activation of Melanocortin (MC) receptors by agonists results in activation of intracellular adenylate cyclase, which synthesizes the second messenger signal molecule, 3 ', 5' -cyclic adenosine monophosphate (cAMP). Measurement of changes in cAMP levels and evaluation of MC1 and MC3 potencies after treatment of recombinant MC1 and MC3 cell lines with test Compounds (EC)50) The calculation is as follows:
stably transfected cell lines were established by stably transfecting Human Embryonic Kidney (HEK) or chinese hamster ovary cell lines with full-length cdnas encoding human MC1 or MC3 receptors, respectively, using standard molecular biology methods. The test compound was dissolved in dimethyl sulfoxide (DMSO) at 4 mM. An 11-point semilog unit incremental dilution series of test compounds, typically starting at 50uM, was diluted in a buffer containing Phosphate Buffered Saline (PBS), 2.5% DMSO, and 0.05% Pluronic F-127 surfactant. Between 80% and 90% of the confluent freshly cultured cells were collected and resuspended in Dulbecco's Modified Eagle's Medium (DMEM). Cells (10,000 cells for MC3, 20,000 cells for MC 1) were added to the test compound dilution series in 384-well measurement plates and incubated for 1 hour at 37 ℃. The relative cAMP concentration in each well was then determined using the β -galactosidase fragment complementation method using the discover cAMP II kit from GE Healthcare/Amersham Biosciences, england. In the case of MC1, DMEM contained 750. mu.M of 3-isobutyl-1-methylxanthine (IBMX) when resuspended cells were used in the experiment. The fluorescence readings for each experimental well were converted to percent effect relative to the maximum value for the control well corresponding to the maximum effect alpha melanocyte stimulating hormone concentration. Log using the conventional software SIGHT10Inhibition concentration vs. percent effect sigmoidal curve fit was performed and the EC was determined by the software as the concentration of test compound half the effect between the experimental points where the sigmoidal dose-response curve goes from low to high50. Each experiment included EC for alpha melanocyte stimulating hormone50It was determined that alpha melanocyte stimulating hormone was used as a standard to follow the consistency of the experiment, allowing EC to be obtained in different experiments50A fair comparison between the values.
EC for MC5 and MC4 were determined by experimental methods D and E in US2005/0176772 (pages 28-30), respectively50And (4) activity.
Nle4, D-Phe 7-alpha-MSH inhibition at MC4 receptor
Nle4, D-Phe7- α -MSH is a stable analog of melanocyte-stimulating hormone (MSH) and is an agonist of the MC4 receptor (MC 4R). Use of the alpha-cyclodextrin125I]Binding competition experiments with Nle4, D-Phe7- α -MSH compared to one can evaluate the ability of a compound to inhibit the binding of Nle4, D-Phe7- α -MSH to the membrane of MC4R expressing cells.
Cells expressing MC4R were homogenized and membrane fragments were separated by differential centrifugation. CHO-CRE MC4R cell membranes were coupled to PVT-PEI-WGA SPA beads type A for 2 hours, centrifuged at 1000RPM for 5 minutes and suspended to 300ug beads/ml (0.15ug membrane, 15ug beads/well). 50 μ l buffer (25mM HEPES, 1mM MgCl) per well in total volume2、2.5mM CaCl21% Pluronic F68, 1 tablet complete EDTA protease inhibitor tablet/50 ml pH7), bead/membrane mixture with 0.06nM [ 2], [ 2]125I]Nle4, D-Phe 7-alpha-MSH and 11 semilog concentrations of competitor ligand were incubated in duplicate. Nonspecific binding was determined by adding 100nM SHU 9119. The reaction was started by adding beads/membrane and the well plate was incubated at room temperature for 12 hours (first hour on the well plate shaker), after which the amount of radioactivity present was determined using a Wallac plate counter. Ki values were determined by data analysis using appropriate software.
Preferably, the compounds of the invention exhibit a binding constant at the MC4 receptor expressed as a Ki value for Nle4, D-Phe7- α -MSH of less than about 1000nM, or more preferably less than 500nM, but more preferably less than about 100nM, and more preferably less than about 50nM, as determined using the above-described experiments.
Nle4, D-Phe 7-alpha-MSH inhibition at MC3 receptor
Nle4, D-Phe7- α -MSH is a stable analog of melanocyte-stimulating hormone (MSH) and is an agonist of the MC3(MC3R) receptor. Use of the alpha-cyclodextrin125I]Binding competition experiments with Nle4, D-Phe7- α -MSH compared to compounds that inhibited the ability of Nle4, D-Phe7- α -MSH to bind to the membrane of MC3R expressing cells were evaluated.
Cells expressing MC3R were homogenized and membrane fragments were separated by differential centrifugation. CHO-CRE MC3R cell membranes were coupled to PVT-PEI-WGA SPA beads type A for 2 hours, centrifuged at 1000RPM for 5 minutes, and suspended to a final experimental concentration of 800ug beads/ml (1.2ug membrane, 40ug beads/well). 50 μ l buffer (25mM HEPES, 1mM MgCl) per well in total volume2、2.5mM CaCl21% Pluronic F68, 1 tablet of complete EDTA protease inhibitor/50 ml pH7), the bead/membrane mixture was mixed with 0.06nM [ 2], [125]Nle4, D-Phe 7-alpha-MSH and 11 semilog concentrations of competitor ligand were incubated in duplicate. Non-specific binding was determined by addition of 100nM SHU 9119. The reaction was started by adding beads/membrane and the well plate was incubated at room temperature for 12 hours (first hour on the well plate shaker), after which the amount of radioactivity present was determined using a Wallac plate counter. Ki values were determined by data analysis using appropriate software.
High Density drug-drug interaction (DDI) 3. mu.M cocktail Screen
Drug interactions are situations where a substance affects the activity of another drug, i.e., an effect is increased or decreased, or two drugs together produce a new effect that neither drug produces. Various processes can lead to drug interactions, but it is relatively common for one drug to affect the pharmacokinetics of another by inhibiting the cytochrome P450 that metabolizes the other drug. Because of the importance of this phenomenon, it is believed that the measurement of the DDI potential of New Chemical Entities (NCEs) early in the drug discovery process is important.
DDI cocktail screening in a fully automated fashion in Human Liver Microsomes (HLM) and the purpose of the screening was to provide a single point assay of the DDI potential of a new chemical entity (NCE; detected at 3. mu.M) for the 4 important cytochrome P450 enzymes 1A2, 2D6, 2C9 and 3A 4.
The substrate cocktail method for P450DDI uses human liver microsome and subtype-specific clinical drug probes and allows simultaneous assay of inhibition of P4501a2, 2D6, 2C9 and 3a4 activity in a single incubation. It was performed at high throughput and simultaneously detected metabolites by LC-MS/MS. This method has been well tested and evaluated using standard compounds. The probe substrates used are shown in the following table.
| Source of microsomes | The collected human liver microsomes are collected, |
| concentration of microsomes | 0.1mg/mL |
| P450 concentration | 0.03μM |
| Regeneration system | NADPH(1.3mM) |
| Experiment time | 8 minutes |
| Probe substrate (Probe-labeled enzyme) | Concentration of |
| Tacrine (1A2) | 2μM |
| Diclofenac (2C9) | 5μM |
| Dextromethorphan (2D6) | 5μM |
| Midazolam (3A4) | 2μM |
| Inhibitors | Concentration of |
| NCE (test Compound) | 3μM |
| Miconazole (general purpose control) | 3μM |
The presence of metabolites for each substrate was determined over time in the presence and absence of NCE (test compound/inhibitor) at a concentration of 3. mu.M. The inhibitory potency of the compounds was determined as a percentage value and is illustrated using the following figure. These data are then combined with other assays to evaluate the suitability of the NCEs and to aid in the design and development of compounds.
| % inhibition | IC50 |
| >75% | <1μM |
| 25-75% | 1-10μM |
| <25% | >10μM |
AGRP inhibition
The Agouti-related protein (AGRP) is a high affinity endogenous antagonist/inverse agonist of the MC4 receptor (Lu et al, 1994, Nature 371: 799-138; Ollman et al, 1997, Science 278: 135-138). Upregulation of AGRP levels by fasting (Mizuno)&Mobbs1999, Endocrinology.140: 4551-4557), it is therefore important to determine the ability of anti-obesity drugs to inhibit AGRP binding by acting on the MC4 receptor. It has now been determined that the C-terminal fragment of AGRP comprises the MC4R binding determinant (Yang et al, 1999, Mol Endocrinol 13: 148-125I]Competitive binding assays for AGRP (87-132) compounds can be evaluated for their ability to inhibit the binding of AGRP to the membrane of MC 4R-expressing cells. To this end, cells expressing MC4R were homogenized and membrane fragments were separated by differential centrifugation. In a total volume of 100. mu.l buffer (25mM HEPES, 1mM MgCl) per well2、2.5mMCaCl20.5% BSA pH7.0), CHO-CRE MC4R cell membrane (12. mu.g protein) and 0.3nM [ 2], [125I]AGRP (87-132) was incubated in duplicate with 11 semilog concentrations of competitor ligand. Non-specific binding was determined by adding 1 μ M SHU 9119. The reaction was started by adding the membrane and the well plates were incubated for 2 hours at room temperature. The reaction was stopped by rapid filtration on GF/C filter paper (pre-soaked in 1% PEI) using a vacuum collector, followed by 5 washes with 200. mu.l ice-cold wash buffer (binding buffer containing 500mM NaCl). The filter paper was immersed in 50. mu.l of scintillation fluid and the amount of radioactivity present was determined by liquid scintillation counting. Ki values were determined by data analysis using appropriate software.
Preferably, the compounds of the invention exhibit a binding constant at the MC4 receptor with a Ki value for AGRP of less than about 1000nM, or more preferably less than 500nM, yet more preferably less than about 100nM, and more preferably less than about 50nM, where the Ki value is determined using the above-described assay. Using this experiment, compounds according to the invention showed binding constants at the MC4 receptor, expressed as Ki values for AGRP, with Ki lower than about 1000 nM.
Food intake study:to evaluate the effect of MC4 agonist on food intake and body weight in male rats over 24 hours.
Approximately two weeks before the study began, rats were housed in a single cage and light-conditioned (9.30am-9.30pm) was switched. Rat adaptation technique experimental instrument 24 hours before study*(Technical scientific Equipment) (TSE) cage. On the morning of the study, rats were randomized by body weight to treatment groups (n-5/treatment). Each rat received an MC4 agonist or solvent orally before lights-off. Immediately after dosing, rats were returned to TSE cages and food intake and water consumption of rats was monitored throughout the study (24 hours). The motion behavior is monitored in the form of a beam break.
At the end of the study, the rats were sacrificed by exsanguination under isoflurane anesthesia. Blood was taken by cardiac puncture and used for analysis of drug concentration levels and biomarkers.
Data are presented as mean ± SEM, and comparisons between control and treatment groups were analyzed using ANOVA. p <0.05 was considered statistically significant.
In vitro metabolic rate determination (human liver microsome (HLM); Rat Liver Microsome (RLM) experiments)
Many drugs are metabolized by the cytochrome P450 monooxygenase system. This enzyme was found to be present in high concentrations in the liver and to bind to the endoplasmic reticulum of hepatocytes. The enzyme system can be obtained in a semi-purified state by preparation of liver microsome fragments. Determining the in vitro half-life of a compound in such a system provides a useful indication of metabolic stability.
Materials and reagents
All reagents were of the anaral grade.
1.200mM phosphate buffer (Sigma) -100 ml of 1M phosphate buffer pH7.4 dissolved in 400ml of MilliQ water. If necessary, the pH is adjusted to 7.4 with concentrated orthophosphoric acid, formulated monthly and stored in the refrigerator (2-8 ℃).
2.0.1M MgCl2.6H2O (BDH) -2.032g was dissolved in 100ml MilliQ water and stored in a refrigerator (2-8 ℃ C.).
3.0.02M NADP (Sigma) -15.3mg were dissolved in 1000. mu.l MilliQ water-and then stored in the refrigerator (2-8 ℃ C.) until use.
4.0.1M D-L Isocitric acid (Sigma) -129mg was dissolved in 5ml MilliQ water-and then stored in the refrigerator (2-8 ℃ C.) until use.
5. Isocitrate dehydrogenase, form IV (Sigma) -stored in the refrigerator (2-8 ℃).
6. Stock solutions of the substrate in miscible organic solvents such as methanol, ethanol or water (about 1mg/ml) are stored in a refrigerator (2-8 ℃).
7.50mM p-nitroanisole (PNA) (Aldrich) -7.65mg were dissolved in 1ml methanol and stored in the refrigerator (2-8 ℃ C.) until use.
8.50. mu.M p-nitrophenol (PNP) (Sigma) -0.69mg dissolved in 100ml water and stored in the refrigerator (2-8 ℃ C.).
9.20% trichloroacetic acid (TCA) (BDH) -20g was dissolved in 100ml MilliQ water, prepared in amber glassware, and stored at room temperature.
10.10M sodium hydroxide (BDH) -40g was dissolved in 100ml MilliQ water (note that this solution should be practiced when preparing this solution due to the exothermic nature of this reaction), prepared in "non-frangible" (safebreak) glassware, and stored at room temperature.
11. Liver or Supermix microsomes, stored at-80 ℃, dissolved immediately prior to use, kept on ice and dispensed (dispense).
MilliQ water.
13. A thermostatically controlled shaking water bath was set for incubation at about 37 ℃.
14. An agent (usually an organic solvent, acid or base) to stop the incubation.
Methodology for in vitro metabolic rate determination using liver and Supermix microsomes
The following procedure was used for a total incubation volume of 1.5 ml.
1. The following mixtures were prepared in test tubes:
| reagent | Stock solution concentration | Incubation concentration | Add volume (for 1.5ml incubation) |
| Phosphate buffer pH7.4 | 200mM | 50mM | 375μl |
| MgCl2 | 0.1M | 5mM | 75μl |
| Isocitric acid | 0.1M | 5mM | 75μl |
| Isocitrate dehydrogenase | On the bottle | 1 unit/ml* | See below* |
*This volume was calculated for each new batch of isocitrate dehydrogenase
For example, the protein concentration is 18mg/ml
Enzyme activity 3.3 units/mg
Thus, specific activity 3.3 × 18 units/ml 59 units/ml
For 1.5ml of incubation, one needs to do
2. The microparticles are thawed at room temperature and a sufficient amount of microparticles is added to give a final concentration of 0.5nmol cytochrome P450/ml of incubation, e.g. for 1.5ml of incubation the volume of microparticles added is:
3. sufficient MilliQ water was added to give a total incubation volume of 1.425 ml.
4. 237.5. mu.l of the incubation mixture was removed and placed in a test tube as a PNA positive control. Add 2.5. mu.l of PNA solution, mix (whirlimix), and place the tube in a holder in a thermostatically controlled shaking water bath.
5. 100 μ l was removed as a no substrate control and dispensed into test tubes. The detection tube is placed in a support of a constant temperature control oscillation water bath.
6. Substrate was added to the incubation. The initial concentration of substrate should be 1. mu.M. The volume of substrate required in the remaining 1.162.5ml of incubation solution) was calculated as follows:
note that the volume of organic solvent added should not exceed 0.1% of the total volume of the incubation fluid.
7. 100 μ l of the incubation mixture was removed and placed in a detection tube as a no cofactor control (no cofactontrol). Mix (Whirlimix) and place in a thermostatically controlled shaking water bath rack.
8. Tubes containing the incubation mixture, positive control and no-cofactor control tubes were preincubated in a thermostatically controlled shaking water bath set at 37 ℃ for 5 minutes.
9. The reaction was initiated by addition of NADP (75. mu.l per 1.162.5ml of incubation mixture, 12.5. mu.l to positive control tube and 5. mu.l to no substrate tube) and the first time point was taken immediately. PNA positive controls, no cofactor controls and no substrate tubes were incubated for total incubation time.
10. From 0 to 60 minutes 9 different sampling points (usually 0,3, 5, 10, 15, 20,30, 45 and 60min) 100. mu.l aliquots were taken and the reaction was stopped. Longer incubation times can be used, but after 120 minutes the microsomes denature. The reaction can be terminated by the addition of an organic solvent, acid or base. After the incubation period was complete, the no cofactor and no substrate controls were terminated in a similar manner, i.e., with the same reagents.
11.PNA Positive control protocol:
After the final sampling, the positive control was removed and 1ml 20% TCA was added to the tube. Tubes containing 250. mu.l of 50. mu.M PNP standard were also prepared and 1ml 20% TCA was added. The two tubes were mixed (Whirlimix) and held for about 5 minutes to allow the protein to precipitate.
Both tubes were centrifuged in the instrument set at 3500rpm for about 5 minutes. 1ml of the supernatant was removed and placed in a clean test tube and the residue was removed.
To this supernatant was added 1ml of 10M NaOH, mixed (Whirlimix), and kept mixed for about 5 minutes. The blank spectrophotometer had distilled water, after which the absorbance of the PNP standard versus distilled water was measured at 400 nm. Microsomal 4-nitrobenzyl ether O-demethylase activity was calculated as follows:
calculation of results
The activity value of the incubation must be equal to or greater than 85% of the average value of the batch of effective incubation. If this criterion is not met, the incubation must be repeated.
11. Samples were analyzed by substrate-specific experiments (including no cofactors and no substrate controls) to determine elimination kinetics.
Data analysis
The data obtained using the above procedure can be quantified in terms of the intrinsic clearance in vitro (Clint) of the substrate. Assuming substrate concentrations below Km, metabolism should be first order elimination kinetics, giving a log-linear curve of substrate elimination over time.
The in vitro half-life of a substrate can be determined by plotting the natural logarithm (1n) of the relative substrate concentration (e.g., drug/internal reference ratio) assay versus time and fitting an optimal line to this data. The slope of this line is the first order rate constant (k) for substrate elimination, determined by regression analysis. This rate constant can be converted to a half-life according to the following equation:
alternatively, the rate constant can be converted to intrinsic clearance (Clint) according to the following equation:
clint (μ l/min/mg) ═ k/protein concentration in incubation (mg/ml)*1000
Preferably, the compounds of the invention exhibit clearance, as determined by the above assay, as expressed by a clearance value, of less than about 200. mu.L/min/mg, more preferably less than about 100. mu.L/min/mg, yet more preferably less than about 50. mu.L/min/mg, and more preferably less than about 20. mu.L/min/mg. Using this experiment, the clearance of compounds according to the invention was tested to be below 200. mu.L/min/mg.
Method of administration
The compounds of the invention contemplated for use as medicaments may be administered as crystalline or amorphous products. Solid suppositories, powders or films can be obtained, for example, by precipitation, crystallization, freeze-drying, spray-drying or evaporation-drying. Microwave or radio frequency drying may be used for this purpose.
One or more compounds of the invention may be administered alone, or in combination with one or more other drugs (or, e.g., any combination thereof). In general, administration may be in the form of a formulation in combination with one or more pharmaceutically acceptable excipients (excipients). The term "excipient" is used herein to describe any ingredient other than a compound of the invention. The choice of excipients is widely varied and depends on factors such as the particular mode of administration, the effect of the excipients on solubility and stability, and the nature of the dosage form.
Pharmaceutical compositions suitable for delivery of the compounds of the invention and methods for their preparation will be apparent to those skilled in the art. These compositions and processes for their preparation can be found, for exampleRemington’s Pharmaceutical Sciences19 th edition (Mack publishing company, 1995).
The present invention therefore provides a pharmaceutical composition comprising a compound of formula (I) and a pharmaceutically acceptable diluent or carrier.
Any suitable route of administration may be employed for providing an effective dose of a compound of the invention to a mammal, particularly a human. For example, oral (including buccal and sublingual), rectal, topical, parenteral, ophthalmic, pulmonary, nasal, and the like may be used. Dosage forms include tablets, troches, dispersions, suspensions, solutions, capsules, emulsions, ointments, aerosols, and the like. Preferably, the compound of formula (I) is administered orally or intranasally.
The effective dosage of the active ingredient employed will vary with the particular compound employed, the mode of administration, the nature of the mammal being treated (e.g., body weight), the condition being treated, and the severity of the condition being treated. These dosages can be readily determined by one skilled in the art.
For the treatment of sexual dysfunction, a dose ranging from about 0.001 milligram (mg) to about 1000mg of a compound of the invention is administered, preferably, from about 0.001mg to about 500mg, more preferably from about 0.001mg to about 100mg, even more preferably, from about 0.001mg to about 50mg, and particularly, from about 0.002mg to about 25mg per kilogram of body weight, preferably in a single dose orally or nasally by nebulization. For example, oral administration requires a total daily dose of from about 0.1mg to about 1000mg, whereas an intravenous dose requires only from about 0.001mg to about 100 mg. The total daily dose may be administered in single or divided doses, and may be administered as prescribed, beyond the general dosage ranges given herein.
When treating obesity in combination with diabetes and/or hyperglycemia, or obesity alone, satisfactory results are obtained when the compounds of the invention are administered in a daily dose of from about 0.0001mg to about 1000mg, preferably from about 0.001mg to about 500mg, more preferably from about 0.005mg to about 100mg, and especially from about 0.005mg to about 50mg per kilogram of animal body weight, preferably in single or divided doses 2 to 6 times a day, or in sustained release dosage forms. In the case of a 70kg adult, the total daily dose is usually from about 0.7mg to about 3500 mg. Such dosing regimens may be adjusted to provide the optimal therapeutic response.
When treating diabetes and/or hyperglycemia, and other diseases or conditions in which compounds of formula I may be used, satisfactory results are obtained when the compounds of the invention are administered in daily doses of from about 0.001mg to about 100mg per kilogram of animal body weight, preferably in single or divided doses 2 to 6 times a day, or in sustained release dosage forms. In the case of a 70kg adult, the total daily dose will generally be from about 0.07mg to about 350 mg. Such dosing regimens may be adjusted to provide the optimal therapeutic response.
These doses are based on average body weight of about 65kg to 70kg of human subjects. For subjects outside this range, such as infants, elderly and obese persons, the physician can readily determine their dosage.
The compounds of the invention may be administered orally. Oral administration may include swallowing, so that the compound enters the gastrointestinal tract, and/or buccal, lingual or sublingual administration, so that the compound enters the blood stream from the mouth.
Dosage forms suitable for oral administration include solid, semi-solid and liquid systems, such as tablets; soft or hard capsules, liquids or powders comprising poly-or nano-particles; lozenges (including liquid-filled); a masticatory; gelling agent; a rapidly dispersing dosage form; a film; a vaginal gel; spraying; and buccal/mucoadhesive patches.
Liquid dosage forms include suspensions, solutions, syrups and elixirs. These dosage forms may be employed as a fill for soft or hard capsules (e.g., prepared from gelatin or hydroxypropylmethylcellulose) and typically include a carrier, for example, water, ethanol, polyethylene glycol, propylene glycol, methylcellulose, or a suitable oil, and one or more emulsifying agents and/or suspending agents. Liquid dosage forms may also be prepared by reconstitution of a solid, for example from a sachet.
The compounds of the present invention may also be used in fast dissolving, fast disintegrating dosage forms, such as Liang and Chen (2001) Expert Opinion in Therapeutic Patents,11(6) 981-.
For tablet dosage forms, depending on the dosage, the drug may constitute from 1 wt% to 80 wt% of the dosage form, more typically from 5 wt% to 60 wt% of the dosage form. In addition to the drug, tablets typically contain a disintegrant. Examples of disintegrants include sodium starch glycolate, sodium carboxymethylcellulose, calcium carboxymethylcellulose, croscarmellose sodium, crospovidone, polyvinylpyrrolidone, methylcellulose, microcrystalline cellulose, lower alkyl-substituted hydroxypropyl cellulose, starch, pregelatinized starch, and sodium alginate. Typically the disintegrant is from 1 to 25 wt% of the dosage form, preferably from 5 to 20 wt% of the dosage form.
Binders are commonly used to affect the binding properties of the tablet dosage form. Suitable binders include microcrystalline cellulose, gelatin, sugars, polyethylene glycol, natural and synthetic gums, polyvinylpyrrolidone, pregelatinized starch, hydroxypropyl cellulose, and hydroxypropyl methyl cellulose. Tablets may also include diluents such as lactose (monohydrate, spray dried hydrate, anhydrous form, etc.), mannitol, xylitol, dextrose, sucrose, sorbitol, microcrystalline cellulose, starch, and dibasic calcium phosphate dihydrate.
The tablets may also optionally include surfactants such as sodium lauryl sulfate and polysorbate 80, and glidants such as silicon dioxide and talc. When present, the surfactant may be from 0.2 wt% to 5 wt% of the tablet, and the glidant may be from 0.2 wt% to 1 wt% of the tablet.
Tablets also typically include lubricants such as magnesium stearate, calcium stearate, zinc stearate, sodium stearyl fumarate, and mixtures of magnesium stearate and sodium lauryl sulfate. The lubricant is typically 0.25 to 10 wt% of the tablet, preferably from 0.5 to 3 wt% of the tablet.
Other possible ingredients include antioxidants, coloring agents, flavoring agents, preservatives, and taste masking agents.
Exemplary tablets contain up to about 80% drug, from about 10 wt% to about 90 wt% binder, from about 0 wt% to about 85 wt% diluent, from about 2 wt% to about 10 wt% disintegrant, and from about 0.25 wt% to about 10 wt% lubricant.
The tablet mixture can be directly compressed or compressed by a roll tablet machine (roller) to form tablets. The tablet mixture or a portion of the mixture may be wet, dry or melt granulated, melt congealed, or extruded prior to tableting. The final formulation may comprise one or more layers, and may or may not be coated; and may even be encapsulated.
The preparation of the tablet is described inPharmaceutical Dosage Forms:TabletsVol.1, H.Lieberman and L Lachman (Marcel Dekker, New York, 1980).
Consumable oral films for human or veterinary use are typically flexible, water-soluble or water-swellable thin film dosage forms which dissolve rapidly or are mucoadhesive, and the films typically comprise a compound of formula I, a film-forming polymer, a binder, a solvent, a wetting agent, a plasticizer, a stabilizer or emulsifier, a viscosity-modifying agent, and a solvent. Some components of the formulation may serve multiple functions.
The compounds of formula I may be water soluble or water insoluble. The water soluble compound typically comprises from 1 wt% to 80 wt%, more typically from 20 wt% to 50 wt% of the solute. Less soluble compounds may constitute a greater proportion of the composition, typically up to 88% by weight of the solute. Alternatively, the compound of formula I may be in the form of multiparticulate beads.
The film forming polymer may be selected from natural polysaccharides, proteins, or synthetic hydrocolloids, and typically the proportion ranges from 0.01 to 99 wt%, more typically 30 to 80 wt%.
Other possible ingredients include antioxidants, colorants, flavors and odor modifiers, preservatives, saliva stimulating agents, cooling agents, co-solvents (including oils), lubricants, fillers, anti-foaming agents, surfactants, and taste masking agents.
The membranes according to the invention are generally obtained by evaporating a thin aqueous membrane coated on a peeled support or paper. This can be done in a drying oven or tube, typically a combination coater dryer, or by lyophilization or vacuum drying.
Solid formulations for oral administration may be formulated as immediate release and/or sustained release formulations. Sustained release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
Suitable sustained release formulations for the purposes of the present invention are described in U.S. Pat. No. 6,106,864. Suitable delivery techniques, e.g. high energy dispersion and penetration and coating of the particles are described in detail in the descriptionPharmaceutical Technology On-line25(2), 1-14, Verma et al (2001). The use of chewing gum to achieve controlled release is described in WO 00/35298.
The compound liquid of the present invention can be directly administered into blood, muscle or organs in vivo. Suitable modes of parenteral administration include intravenous, arterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular, intrasynovial and subcutaneous administration. Suitable devices for parenteral administration include needle (including microneedle) injectors, needleless injectors and infusion techniques.
Parenteral formulations are typically aqueous solutions which may contain excipients such as salts, carbohydrates and buffers (preferably at a pH of from 3 to 9), but, for some applications, they may more suitably be formulated as sterile aqueous solutions or in dry form for use in conjunction with a suitable vehicle such as sterile, pyrogen-free water.
Preparation of parenteral formulations under sterile conditions (e.g., by lyophilization) can be rapidly achieved using standard pharmaceutical techniques well known to those skilled in the art.
The solubility of the compound of formula (I) used in the parenteral solution formulation may be increased by using suitable formulation techniques, such as by adding agents that increase solubility.
Formulations for parenteral administration may be formulated as immediate release and/or sustained release formulations. Sustained release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release. Thus, the compounds of the present invention may be formulated for administration as a suspension or as a solid, semi-solid, or thixotropic liquid, as an implanted depot that provides sustained release of the active compound. Examples of such formulations include drug-coated stents (stents) and semi-solids and suspensions comprising drug-loaded poly (dl-lactic-co-glycolic acid) acid (PGLA) microspheres.
The compounds of the invention may be administered topically, dermally (internally), or transdermally to the skin or mucosa. Typical formulations for this purpose include gels, hydrogels, lotions, solutions, emulsions, ointments, dusting powders, dressings, foams, films, skin patches, sheets, implants, sponges, fibers, bandages and microemulsions. Liposomes may also be used. Typical carriers include alcohols, water, mineral oil, liquid paraffin, white paraffin, glycerol, polyethylene glycol and propylene glycol. Penetration enhancers-see, e.g., J pharm sci,88(10) 955 Across 958, Finnin and Morgan (10 months 1999).
Other means of topical administration include by electroporation, iontophoresis, sonophoresis and microneedle or needle-free (e.g. Powderject)TM,BiojectTMEtc.) for injection.
Formulations for topical administration may be formulated as immediate release and/or sustained release formulations. Sustained release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
The compounds of the invention may also be administered nasally or by inhalation, typically in the form of a dry powder from a dry powder inhaler (alone, as a mixture, for example with lactose, or as a particulate blend of ingredients, for example with a phospholipid such as phosphatidylcholine), or as a spray from a pressurised container, pump, spray, nebuliser (preferably a nebuliser using electrohydrodynamics to produce a fine mist), or with or without a suitable propellant such as 1,1, 1, 2-tetrafluoroethane or 1,1, 1, 2, 3, 3, 3-heptafluoropropane, or as nasal drops. For intranasal use, the powder may comprise a bioadhesive agent, for example, chitosan or cyclodextrin.
A pressurized container, pump, sprayer, nebulizer or atomizer containing a solution or suspension of a compound of the invention comprising, for example, ethanol, aqueous ethanol, or other suitable agent for dispersing, dissolving, or prolonging the release of the active agent, a propellant as a solvent, and optionally a surfactant such as sorbitan trioleate, oleic acid, or oligomeric lactic acid.
Prior to use in dry powder or suspension formulations, the drug product is micronized to a size suitable for delivery by inhalation (typically less than 5 microns). This may be accomplished by any suitable comminution method, such as spiral jet milling, fluidized bed jet milling, supercritical fluid methods to form nanoparticles, high pressure homogenization, or spray drying.
Capsules (e.g. made of gelatin or hydroxypropylmethylcellulose), blisters (blisters) and cartridges for use in an inhaler or insufflator may be formulated containing a powder mix of a compound of the invention, a suitable powder base such as lactose or starch, and a performance modifier such as l-leucine, mannitol, or magnesium stearate. Lactose may be in the anhydrous or monohydrate form, the latter being preferred. Other suitable excipients include dextran, glucose, maltose, sorbitol, xylitol, fructose, sucrose and trehalose.
Suitable solution formulations for use in nebulisers using electrohydrodynamics to produce a fine particle mist may contain from 1 μ g to 20mg of a compound of the invention per actuation and the volume per actuation may be from 1 μ l to 100 μ l. A typical formulation may comprise a compound of formula (I), propylene glycol, sterile water, ethanol and sodium chloride. Other solvents that may be used in place of propylene glycol include glycerol and polyethylene glycol.
Suitable flavoring agents, such as menthol and levomenthol, or sweetening agents, such as saccharin or saccharin sodium, may be added to those formulations of the invention for inhalation/intranasal administration.
Formulations for inhalation/intranasal administration using, for example, PGLA, may be formulated as immediate release and/or sustained release formulations. Sustained release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
In the case of dry powder inhalers and aerosols, the dosage unit (unit) is determined by a valve delivering a metered amount (metered dose). The unit site according to the invention is generally set to administer a metered dose or "spray (puff)" containing from 0.001mg to 10mg of the compound of formula (I). The total daily dose will generally range from 0.001mg to 40mg, which may be administered in a single dose, or more usually in divided doses throughout the day.
The compounds of the invention may be administered rectally or vaginally, for example in the form of suppositories, pessaries or enemas. Cocoa butter is a conventional suppository base, but a variety of other bases may be used as desired.
Formulations for rectal/vaginal administration may be formulated as immediate release and/or sustained release formulations. Sustained release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
The compounds of the invention may also be administered directly to the eye or ear, usually in the form of drops of micronized suspension or solution in isotonic, pH-adjusted sterile saline. Other formulations suitable for ophthalmic and otic administration include ointments, gels, biodegradable (e.g. absorbable gel sponge, collagen) and non-biodegradable (e.g. silicone) implants, wafers, lenses and particles or vesicular systems such as lipid vesicles (niosomes) or liposomes. Polymers such as crosslinked polyacrylic acid, polyvinyl alcohol, hyaluronic acid, cellulosic polymers (e.g., hydroxypropyl methylcellulose, hydroxyethyl cellulose, or methyl cellulose), or heteropolysaccharide polymers (e.g., agar gum) may be incorporated with preservatives such as benzalkonium chloride. These formulations can also be delivered iontophoretically.
Formulations for ophthalmic/otic administration may be formulated as immediate release and/or sustained release formulations. Sustained release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
The compounds of the present invention may be combined with water-soluble macromolecular entities such as cyclodextrins, and suitable derivatives thereof, or polyethylene glycol-containing polymers, to enhance their solubility, dissolution rate, taste masking, bioavailability and/or stability for use in any of the above modes of administration.
Drug-cyclodextrin complexes are found, for example, in most dosage forms and routes of administration. Both inclusion and non-inclusion complexes may be used. In addition to being complexed directly with the drug, cyclodextrins may also be used as an auxiliary additive, i.e. as a carrier, diluent or cosolvent. The most commonly used for these purposes are alpha-, beta-and gamma-cyclodextrins, examples of which are found in international patent applications WO91/11172, WO94/02518 and WO98/55148.
Since, for example, a combination of active compounds is administered for the purpose of treating a particular disease or condition, it is within the scope of the present invention to include two or more pharmaceutical compositions (at least one containing a compound according to the invention) which may be conveniently combined in a kit suitable for co-administration of the compositions.
Thus, the kit of the invention comprises two or more separate pharmaceutical compositions (at least one containing a compound of formula (I) according to the invention), and means for preserving said compositions separately, such as containers, separate bottles or separate foil pouches. Examples of such kits are blister packs (blister packs) similar to those used for wrapping tablets, capsules, etc.
The kits of the invention are particularly suitable for administration of different dosage forms (e.g. oral and parenteral), administration of separate compositions at different dosage intervals, or titration of separate compositions from one another. To aid compliance, the kit typically contains administration instructions and may provide so-called memory assistance.
Other aspects of the invention are set out in the claims.
Biological data
| Example No. 2 | MC4EC50(nM) | MC4MSHKi(nM) | MC3EC50(nM) | MC3MSH Ki(nM) | MC4AgRPKi(nM) | HLM(μL/min/mg) | RLM(μL/min/mg) | % inhibition of 3A4 metabolism at 3. mu.M |
| 1 | <0.52 | 43.9 | 39 | <7.0 | <8.5 | 54 | ||
| 2 | 4.53 | 27 | 4.26 | 124 | 59 | <12.0 | 31 | 64 |
| 3 | 11.9 | 16.4 | <7.0 | <19.8 | 60 | |||
| 4 | 55.9 | 51 | 60 | 74 | 38 | |||
| 5 | 137 | 203 | 552 | <7.0 | 27 | 34 | ||
| 6 | 18.9 | 1460 | 456 | <8.5 | 32 | |||
| 7 | 40 | 1600 | 714 | 21 | 11 | 19 | ||
| 8 | 27.5 | 1090 | 643 | >440 | >510 | 94 | ||
| 9 | 39.3 | 520 | 523 | 240 | 411 | 68 | ||
| 10 | 11.9 | 276 | 493 | 977 | 223 | 20.5 | 42 | 19 |
| 11 | 111 | 417 | 14 | |||||
| 12 | 91.6 | >352 | 88.5 | |||||
| 13 | 37.6 | 1020 | 106 | 6 | ||||
| 14 | 115 | 129 | <9.0 | <9.75 | ||||
| 15 | 53.9 | >33400 | ||||||
| 16 | 34.3 | 323 | 251 | <45.5 | <8.5 | 83 | ||
| 17 | 1.59 | 112 | 54.9 | 332 | 20 | >440 | >510 | 35 |
| 18 | 55.5 | >8520 | 965 | 67 | 68 | 34 | ||
| 19 | 6.5 | 85.4 | 425 | 568 | 79 | 21 | 21 | 50 |
| 20 | 16.4 | 251 | 1120 | 1020 | 253 | 31 | 51.5 | 35 |
| 21 | 2.81 | 283 | 66 | 144 | 58.5 | 25 | ||
| 22 | 19.8 | 1050 | 5560 | 5960 | 179 | 96 | 54 | |
| 23 | 4.75 | 65.7 | 185 | 264 | 79 | <15.5 | <8.5 | 35 |
| 24 | 59.3 | 2510 | >955 | 355 | >510 | 87 | ||
| 25 | 12.4 | 10.9 | 189 | <0.105 | 278 | 308 | 172 | 16 |
| 26 | 22.5 | 4210 | 363 | 70 | >510 | 61 | ||
| 27 | 4.54 | 390 | 100 | 51 | 31 | 64 | ||
| 28 | 24.4 | 938 | 1670 | 516 | 33 | 51 | 77 | |
| 29 | 7.54 | 40 | 105 | 36 | 59 | |||
| 30 | 5.77 | 61.5 | 112 | 141 | 69.5 | 38 | ||
| 31 | 22.9 | 410 | 526 | 188 | 292 | 60 | ||
| 32 | 8.78 | 150 | 76.6 | 456 | 160 | 8 | <8.5 | 31 |
| 33 | 13.4 | 193 | 220 | 186 | 72 | 19 | ||
| 34 | 39.2 | 375 | <7.0 | <8.5 | 50 | |||
| 35 | 62.2 | 878 | 386 | 100 | 92 | 46 | ||
| 36 | 168 | 1020 | <7.0 | 17 | 59 | |||
| 37 | 325 | 422 | 19.5 | 23 | 45 | |||
| 38 | 102 | 336 | <7.0 | <8.5 | 58 | |||
| 39 | 2100 | 751 | 95 | 58 | 30 | |||
| 40 | 69.8 | 580 | 430 | <7.0 | <8.5 | 64 |
| 41 | 655 | >33300 | 3750 | 21 | 30 | 52 | ||
| 42 | >20300 | 888 | 25 | 25 | 59 | |||
| 43 | 49.3 | 1590 | 248 | 37 | 44 | |||
| 44 | 284 | >9630 | 2130 | 27 | ||||
| 45 | 44.4 | 66.1 | 251 | <7.0 | <8.5 | 69 | ||
| 46 | 15 | 50.8 | 551 | 134 | <7.0 | <8.5 | 74 |
The effect of 3mg/kg and 10mg/kg of the compound of example 2 on accumulated food intake over 24 hours and on body weight change over 24 hours compared to vehicle is shown in figures 1 and 2.
The invention is illustrated by the following non-limiting examples, in which the following abbreviations and definitions are used:
APCI atmospheric pressure chemical ionization mass spectrum
[α]DSpecific rotation at 589nm
br width
Filtering agent
Chemical shift of delta
d doublet peak
dd two doublets
EI electrospray ionization
Ex examples
LRMS low resolution mass spectrum
m multiplet
Peak of mass m/z
NMR nuclear magnetic resonance
Prec precursor
Prep preparation
psi pounds per square inch
q quartet peak
s single peak
t triplet peak
tlc thin layer chromatography
For ease of synthesis, while in many instances the compounds are initially isolated in their free base form, they are typically converted to the corresponding hydrochloride salt for analytical identification purposes. To avoid ambiguity, both the free base and the HCl salt form are provided herein.
For avoidance of doubt, the compounds used herein were prepared using ACD Labs Name software v7.11TMAnd (4) naming.
Examples
Example 1
6- [ (3S, 4S) -3- (5-Chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-di
Methylpiperidin-1-yl]Carbonyl } pyrrolidin-1-yl]-3-cyanopyridine (nicotinonitrile)
2-chloro-5-cyanopyridine (96mg, 0.69mmol) was added to a solution of pyrrolidine (200mg, 0.46mmol) and N-ethyldiisopropylamine (0.32mL, 1.8mmol) from preparation 10 in acetonitrile (10mL), and the mixture was heated under nitrogen at 70 ℃ overnight. The solvent was removed in vacuo and the residue was purified by column chromatography (silica gel) eluting with dichloromethane, increasing polarity to 5% methanol in dichloromethane to give the title compound (191mg, 77%) as a white solid.1H NMR(CD3OD, 400MHz) delta 0.4-0.6(6H, 4x d), 0.78-0.82, 1.60-1.68 and 1.97-2.05(2H, 3x m), 2.73-2.80(1H,m), 3.00-3.20(1H, m), 3.68-4.37(8H, m), 6.62(1H, m), 7.01-7.08 and 7.37-7.48(5H, 2x m), 7.74(1H, m), 7.80 and 7.95(1H, 2 xdd), 8.40(1H, m), 8.57 and 8.60(1H, 2x d); LRMS (EI)+)534[MH+];[α]D 25=-47.0(c=0.215,MeOH).
Example 2
6- [ (3S, 4S) -3- (5-Chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-di
Methylpiperidin-1-yl]Carbonyl } pyrrolidin-1-yl]Pyridazine-3-carbonitriles
3-chloro-6-cyanopyridazine (prepared according to US3,637,691) (20mg, 0.14mmol) was added to a solution of pyrrolidine (40mg, 0.09mmol) and N-ethyldiisopropylamine (0.06mL, 0.37mmol) in acetonitrile (10mL) from preparation 10, and the mixture was heated under nitrogen at 70 ℃ overnight. The solvent was removed in vacuo and the residue was purified by column chromatography (silica gel) eluting with dichloromethane, increasing polarity to 5% methanol in dichloromethane to give the title compound (41mg, 83%) as a white solid.1HNMR(CD3OD, 400MHz) δ 0.4-0.6(6H, 4x d), 0.78-0.85, 1.60-1.69 and 1.97-2.10(2H, 3x m), 2.72-2.80(1H, m), 3.00-3.23(1H, m), 3.68-4.38(8H, m), 6.99-7.08 and 7.38-7.50(6H, 2x m), 7.70(1H, d), 7.80 and 7.94(1H, 2 xdd), 8.58 and 8.61(1H, 2x d); LRMS (EI)+)535[MH+].
Example 3
(3R, 4R, 5S) -1- { [ (3S, 4S) -1- (6-Chloropyridazin-3-yl) -4- (5-Chloropyridin-2-yl) pyrrolidin-3-yl]
Carbonyl } -4- (4-fluorophenyl) -3, 5-dimethylpiperidin-4-ol
1, 6-dichloropyridazine (173mg, 1.2mmol) was added to a solution of pyrrolidine (100mg, 0.23mmol) and N-ethyldiisopropylamine (0.16mL, 0.93mmol) from preparation 10 in acetonitrile (5mL), and the mixture was heated at 70 ℃ under nitrogen overnight. The solvent was removed in vacuo and the residue was purified by column chromatography (silica gel) eluting with dichloromethane, increasing polarity to 5% methanol in dichloromethane to give the title compound (52mg, 42%) as a white solid.1HNMR(CD3OD, 400MHz) δ 0.4-0.6(6H, 4x d), 0.78-0.85, 1.60-1.69 and 1.97-2.10(2H, 3x m), 2.72-2.80(1H, m), 3.00-3.23(1H, m), 3.68-4.38(8H, m), 6.99-7.08 and 7.38-7.50(6H, 2xm), 7.70(1H, d), 7.80 and 7.94(1H, 2 xdd), 8.58 and 8.61(1H, 2x d); LRMS (EI)+)544[MH+].
Example 4
(3R, 4R, 5S) -1- { [ (3S, 4S) -4- (5-Chloropyridin-2-yl) -1- (6-methoxypyridazin-3-yl) pyrrolidine
-3-yl]Carbonyl } -4- (4-fluorophenyl) -3, 5-dimethylpiperidin-4-ol
Sodium tert-butoxide (31mg, 0.32mmol), 3-chloro-6-methoxypyridazine (34mg, 0.23mmol), tris (dibenzylideneacetone) dipalladium (0) (8.5mg, 0.009mmol) and 2, 2 '-bis (diphenylphosphino) -1, 1' -dinaphthalene (11.5mg, 0.0185mmol) were added to a solution of pyrrolidine (100mg, 0.23mmol) from preparation 10 in toluene (10mL), and the mixture was heated under nitrogen at 80 ℃ overnight. The solvent was removed in vacuo and the residue was dissolved in ethyl acetate (25mL), washed with water, dried (MgSO4) and evaporated. Purification by chromatography (silica gel) eluting with dichloromethane, increasing polarity to 5% methanol in dichloromethane afforded the title compound (48mg, 40%),in the form of yellow foam.1H NMR(CD3OD,400MHz)δ0.4-0.62(d+t+t,6H),0.86(m,1H),1.65(m,1H),1.93-2.08(br,1H),2.75(m,1H),3.17(t,1H),3.74(m,3H),3.95(s,3H),3.96-4.21(m,3H),4.32(d,1H),6.98-7.1(m,5H),7.38-7.48(m,2H),7.79+7.48(2x dd,1H),8.56(d,1H);LRMS(EI+)540[MH+].
Example 5
(3R, 4R, 5S) -4- (4-chlorophenyl) -1- { [ (3S, 4S) -1- (6-chloropyridazin-3-yl) -4- (5-fluoropyridin-2-yl)
Pyrrolidin-3-yl radical]Carbonyl } -3, 5-dimethylpiperidin-4-ol
To (3R, 4R, 5S) -4- (4-chlorophenyl) -1- { [ (3S, 4S) -4- (5-fluoropyridin-2-yl) pyrrolidin-3-yl]Carbonyl } -3, 5-dimethylpiperidin-4-ol (prepared by the same method as used for the preparation of 10, starting from the aldehyde of preparation 17 and (3R, 4S, 5S) -4- (4-chlorophenyl) -3, 5-dimethylpiperidin-4-ol (according to International patent application publication No. WO 2005/077935)) (120mg, 0.28mmol) in dimethyl sulfoxide (2mL), 3, 6-dichloropyridazine (98mg, 0.56mmol), triethylamine (0.12mL, 0.84mmol) and cesium fluoride (13mg, 0.084mmol) were added. The mixture was stirred at 110 ℃ under nitrogen for 3 hours. The reaction mixture was diluted with methanol (5mL) and loaded on a SCX column, eluted with methanol to remove non-basic materials and dimethyl sulfoxide, then 2M NH3The solution in methanol elutes the basic product. The solvent was removed in vacuo to give the title compound (74mg, 49%) as a colourless gum.1H NMR(CD3OD, 400MHz) δ 0.44-0.60(6H, 4x d), 0.95-1.02, 1.64-1.73 and 1.94-2.09(2H, 3x m), 2.69-2.79(1H, m), 2.99-3.06 and 3.16-3.22(1H, 2x m), 3.74-3.79(3H, m), 4.01-4.24(4H, m), 4.32-4.36(1H, m), 7.02-7.07(1H, m), 7.29-7.70(7H, m), 8.45-8.49(1H, m); LRMS (EI +)544[ MH +].
Example 6
6- [ (3S, 4S) -3- { [ (3R, 4R, 5S) -4- (4-chlorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl]Carbonyls
-4- (5-fluoropyridin-2-yl) pyrrolidin-1-yl]Pyridazin-3 (2H) -ones
A solution of chloropyridazine (70mg, 0.13mmol) from example 5 was dissolved in degassed acetic acid and stirred under nitrogen at reflux overnight. The solvent was removed in vacuo and the residue was purified by column chromatography (silica gel) eluting with dichloromethane/methanol/aqueous ammonia 95/5/0.5. The title compound was obtained as an off-white solid (37mg, 55%).1H NMR(CD3OD, 400MHz) δ 0.43-0.59(6H, 4x d), 0.94-1.03, 1.64-1.73 and 1.93-2.03(2H, 3x m), 2.65-2.78(1H, m), 2.97-3.03 and 3.14-3.20(1H, 2x m), 3.62-3.79(3H, m), 3.86-4.17(4H, m), 4.31-4.35(1H, m), 6.88-6.91(1H, m), 7.30-7.42(5H, m), 7.47-7.57(1H, m), 7.63-7.68(1H, m), 8.44-8.48(1H, m); LRMS (EI +)526 MH].
Example 7
6- [ (3S, 4S) -3- { [ (3R, 4R, 5S) -4- (4-chlorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl]Carbonyls
-4- (5-fluoropyridin-2-yl) pyrrolidin-1-yl]-2-methylpyridazin-3 (2H) -one
To a solution of pyridazinone (30mg, 0.057mmol) from example 6 in dimethylformamide (2mL) was added a solution of 1M sodium hexamethyldisilazide (tetrahydrofuran) (0.07mL, 0.07mmol) in tetrahydrofuran (0.07 mL), and lithium bromide (6mg, 0.07 mmol). Stirring under nitrogen at room temperatureThe mixture was stirred for 30 minutes, then methyl iodide (0.004mL, 0.07mmol) was added and the mixture was stirred at room temperature under nitrogen for 2 hours. The solvent was removed in vacuo and the residue was purified by column chromatography (silica gel) eluting with dichloromethane/methanol/aqueous ammonia 95/5/0.5. The title compound was obtained as a yellow solid.1H NMR(CD3OD, 400MHz) δ 0.44-0.59(6H, 4x d), 0.94-1.03, 1.64-1.73 and 1.95-2.04(2H, 3x m), 2.68-2.78(1H, m), 2.95-3.01 and 3.14-3.21(1H, 2x m), 3.62-3.81(6H, m), 3.87-4.17(4H, m), 4.31-4.35(1H, m), 6.87-6.90(1H, m), 7.25-7.34(4H, m), 7.38-7.42(1H, m), 7.48-7.58(1H, m), 7.64-7.69(1H, m), 8.44-8.48(1H, m); LRMS (EI +)540[ MH +].
Example 8
(3R, 4R, 5S) -1- { [ (3S, 4S) -4- (5-Chloropyridin-2-yl) -1- (5-Fluoropyridin-3-yl) pyrrolidin-3-yl]
Carbonyl } -3, 5-dimethyl-4-pyridin-2-ylpiperidin-4-ol
To (3R, 4R, 5S) -1- { [ (3S, 4S) -4- (5-chloropyridin-2-yl) pyrrolidin-3-yl]Carbonyl } -3, 5-dimethyl-4-pyridin-2-ylpiperidin-4-ol (prepared by the same method as used for the preparation of the amine of 10, starting from a solution of (3R, 4S, 5S) -3, 5-dimethyl-4-pyridin-2-ylpiperidin-4-ol (starting according to International patent application publication No. WO2005/077935) (50mg, 0.12mmol) in toluene (5mL) was added 3-bromo, 5-fluoropyridine (25mg, 0.14mmol), tris (dibenzylideneacetone) dipalladium) (4.4mg, 0.0048mmol), BINAP (6mg, 0.0096mmol) and sodium tert-butoxide (26mg, 0.27 mmol). The mixture was stirred overnight at 80 ℃ under nitrogen. The solvent was removed in vacuo and the residue was purified by column chromatography (silica gel) eluting with dichloromethane/methanol/aqueous ammonia 99/1/0.1, increasing polarity to 95/5/0.5. The title compound (11mg, 18%) was obtained as a yellow gum.1H NMR(CD3OD,400MHz)δ0.33-0.53(6H, 4x d), 0.81-0.93, 1.81-1.92 and 2.08-2.28(2H, 3x m), 2.69-2.78(1H, m), 2.99-3.20(1H, m), 3.61-3.94(5H, m), 4.01-4.23(2H, m), 4.34-4.42(1H, m), 6.81-6.90(1H, m), 7.26-7.50(3H, m), 7.70-7.94(4H, m), 8.51-8.62(2H, m); LRMS (EI +)510[ MH +].
Example 9
(3R, 4R, 5S) -1- { [ (3S, 4S) -4- (5-Chloropyridin-2-yl) -1-pyridazin-3-ylpyrrolidin-3-yl]Carbonyls
3, 5-dimethyl-4-pyridin-2-ylpiperidin-4-ol
To (3R, 4R, 5S) -1- { [ (3S, 4S) -4- (5-chloropyridin-2-yl) pyrrolidine, 3-yl]Carbonyl } -3, 5-dimethyl-4-pyridin-2-ylpiperidin-4-ol (prepared by the same method as used for the preparation of the amine of 10, starting from a solution of (3R, 4S, 5S) -3, 5-dimethyl-4-pyridin-2-ylpiperidin-4-ol (according to International patent application publication No. WO 2005/077935)) (50mg, 0.12mmol) in DMSO (2mL) was added 3-chloropyridazine (28mg, 0.24mmol), cesium fluoride (18mg, 0.12mmol) and triethylamine (0.05mL, 0.36 mmol). The mixture was stirred overnight at 100 ℃ under nitrogen. The reaction mixture was diluted with 10mL ethyl acetate and washed with 3 × 20mL water. The combined aqueous extracts were extracted with 10mL ethyl acetate and the combined organic extracts were washed with 10mL brine and dried (MgSO)4) And then the solvent is removed in vacuo. The residue was purified by column chromatography (silica gel) eluting with dichloromethane/methanol/aqueous ammonia 99/1/0.1 increasing polarity to 95/5/0.5. The title compound (16mg, 27%) was obtained as a yellow gum.1HNMR(CD3OD, 400MHz) δ.0.20-0.37(6H, 4x d), 0.91-1.13, 1.67-1.76 and 1.94-2.09(2H, 3x m), 2.54-2.62(1H, m), 2.87-2.93 and 2.99-3.05(1H, 2x m), 3.61-3.74(3H, m), 3.86-4.08(4H, m), 4.20-4.26(1H, m), 6.83-6.88(1H, m), 7.11-7.44(4H, m), 7.63-7.78(2H, m), 8.30-8.46(3H, m); LRMS (EI +)493[ MH +].
Example 10
6-1(3S, 4S) -3- (5-Chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (5-Chloropyridin-2-yl) -4-hydroxy
-3, 5-dimethylpiperidin-1-yl]Carbonyl } pyrrolidin-1-yl]-2-methylpyridazin-3 (2H) -one
To a solution of the carboxylic acid from preparation 15 (42mg, 0.08mmol) in dichloromethane (3mL) was added N-ethyldiisopropylamine (0.04mL, 0.25mmol), 1-hydroxybenzotriazole (15mg, 0.095mmol) and 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide, and the mixture was stirred under nitrogen for 30 minutes. The amine from preparation 16 was added and the mixture was stirred under nitrogen overnight. The reaction mixture was diluted with dichloromethane (20mL) and washed with saturated NaHCO3(20mL) and brine (20 mL). The organic layer was MgSO4Dried, filtered and evaporated to give a yellow oily solid. Purification by column chromatography (silica gel), eluting with dichloromethane and dichloromethane/methanol to increase polarity to 95/5, gave the title compound as a yellow solid (31mg, 67%).1H NMR(CD3OD,400MHz)δ0.36-0.6(m,6H),1.18(br,1H),1.89(br,1H),2.16(br,1H),2.71(m,1H),3.13(m,1H),3.6-4.2(s+m,9H),4.33(d,1H),6.89(d,1H),7.2-7.5(m,3H),7.78-7.92(m,2H),8.56(d,2H);LRMS(EI+)558[MH+].
Examples 11 to 46
Examples 11-46 were prepared according to the methods described above, e.g., 1-10, from the appropriate pyridine aldehydes1And suitably 4-substituted 3, 5-dimethylpiperidin-4-ol2And (4) starting.
1.2-Methoxypyridine-5-carbaldehyde is commercially available. Other aldehydes were as described in preparations 2, 17, 18 and 22.
(3R, 4S, 5S) -4- (5-Chloropyridin-2-yl) -3, 5-dimethylpiperidin-4-ol as described in preparation 16. The synthesis of other desired piperidinols is described in International patent application publication No. WO 2005/077935.
Preparation of
Preparation 1
5-chloro-2-iodopyridine
Acetyl chloride (11.05mL, 0.155mol) was added to a solution of 2-bromo-5-chloropyridine (20.0g, 0.103mol) in acetonitrile (120mL), followed by sodium iodide (23.3g, 0.155mol), and the mixture was heated under reflux (equipped with a drying tube) for 3 hours. The reaction was cooled in an ice bath, cautiously basified with saturated aqueous potassium carbonate solution and then extracted with ethyl acetate (2 × 100 mL). The combined organic layers were washed with saturated aqueous sodium sulfite (200mL) and dried (MgSO)4) And areAnd (4) evaporating. The residue was then subjected to the same reaction and treatment conditions to ensure complete reaction and the title compound (18.71g, 75%) was obtained as a brown solid.1H NMR(CDCl3,400MHz)δ7.30(1H,dd),7.65(1H,d),8.35(1H,d);LRMS(APCI+)240[MH+]。
Preparation 2
5-chloropyridine-2-carbaldehyde
Iodide from preparation 2 (18.71g, 78.1mmol) was dissolved in tetrahydrofuran (100mL) and cooled to-15 ℃ under nitrogen. A solution of isopropyl magnesium chloride in tetrahydrofuran (2M, 42.2mL, 84.4mmol) was then added dropwise, ensuring that the temperature remained below 0 ℃. The reaction mixture was cooled to-15 deg.C, stirred for 1 hour, and dimethylformamide (9.0mL, 116mmol) was added dropwise, maintaining the temperature below 0 deg.C. The reaction mixture was warmed to room temperature and stirred for 1 hour, then cooled to 0 ℃ again, then carefully quenched by the dropwise addition of 2M HCl (100 mL). After the addition was complete, the mixture was stirred at room temperature for 30 minutes, and then the pH was adjusted to 6-7 by adding saturated aqueous sodium bicarbonate solution. The organic layer was separated and the aqueous layer was extracted with dichloromethane (2x200 mL). The combined organic layers were washed with water (200mL) and dried (MgSO)4) Then concentrated in a rotary evaporator, maintaining the temperature below 30 ℃ to give the crude product (13.7g) as a brown oil, which was used without further purification.1H NMR(CDCl3,400MHz)δ7.35(1H,d),7.95(1H,d),8.73(1H,s),10.02(1H,s);LRMS(APCI+)142[MH+].
Preparation 3
(2E) -3- (5-Chloropyridin-2-yl) acrylic acid tert-butyl ester
N-butyllithium (2.5M in hexane, 34mL, 85mmol) was added dropwise to a solution of tert-butyl diethylphosphonoacetate (19.1mL, 81mmol) in diethyl ether (80mL) at-78 ℃ under nitrogen and stirring was continued for 30 min. A solution of the crude aldehyde from preparation 2 (78.1 mmol of iodide from preparation 1) in diethyl ether (20mL) was then added dropwise, maintaining the temperature below-65 ℃. When the addition was complete, the mixture was warmed to room temperature over 2 hours and then carefully quenched by addition of saturated aqueous ammonium chloride (200 mL). The mixture was extracted with ether (2 × 150mL) and the combined organic extracts were washed with brine (200mL) and dried (MgSO)4) And evaporated. The residue was purified by column chromatography (silica gel) using pentane to increase the polarity to pentane/ethyl acetate 8:2 to give the title compound (13.34g, 74% over 2 steps) as an oil.1H NMR(CDCl3,400MHz)δ1.51,(9H,s),6.79(1H,d),7.35(1H,d),7.52,(1H,d),7.66(1H,dd),8.55(1H,d);LRMS(APCI+)240[MH+].
Preparation 4
(2E) Trifluoroacetic acid salt of (E) -3- (5-chloropyridin-2-yl) acrylic acid
A solution of trifluoroacetic acid (10mL) in dichloromethane (10mL) was added dropwise to an ice-cooled solution of the ester from preparation 3 (2.09g, 8.7mmol) in dichloromethane (10mL), and the resulting mixture was stirred at room temperature overnight. The solvent was removed in vacuo, toluene (10mL) was added and removed in vacuo, then dichloromethane (10mL) was added and removed in vacuo to give the title compound (2.44g, 94%) as a red solid.1H NMR(CD3OD,400MHz)δ6.86(1H,d),7.64(2H,m),7.87(1H,dd),8.59(1H,d);LRMS(APCI+)184[MH+].
Preparation 5
(4S) -4-benzyl-3- [ (2E) -3- (5-chloropyridin-2-yl) prop-2-enoyl]-1, 3-oxazolidin-2-one
A solution of the acid from preparation 4 (2.44g, 8.2mmol) in tetrahydrofuran (15mL) was cooled to-78 deg.C under nitrogen. Triethylamine (2.85mL, 20mmol) was added dropwise followed by trimethylacetyl chloride (1.11mL, 9.0mmol) at a rate controlled to maintain the temperature below-65 ℃. The mixture was then stirred at-78 ℃ for 2 hours. At-78 ℃ under nitrogennBuLi (2.5M in hexanes, 4.26mL, 10.7mmol) was added dropwise to a solution of (4S) -4-benzyl-1, 3-oxazolidin-2-one (1.74g, 9.8mmol) in tetrahydrofuran (15mL) at a rate controlled to maintain the temperature below-65 ℃. After stirring for 20 minutes at-78 ℃, the oxazolidinone anion solution was added to the mixed anhydride solution through a cannula at-78 ℃. The reaction mixture was stirred at-78 ℃ for 20 minutes and then slowly warmed to room temperature overnight. The reaction was quenched by the addition of saturated aqueous ammonium chloride (30mL) and then concentrated in vacuo to remove tetrahydrofuran. The solid precipitate was filtered and washed with diethyl ether to give the title compound (1.52g, 54%) as a pale yellow solid. The ether washings were evaporated to dryness, slurried in ether and then filtered to give further product (0.42g, 15%).1H NMR(CDCl3,400MHz)δ2.84(1H,t),3.37(1H,d),4.22(2H,m),4.78(1H,m),7.2-7.4(5H,m),7.51(1H,d),7.69(1H,d),7.86(1H,d),8.23(1H,d),8.62(1H,s);LRMS(APCI+)343[MH+].
Preparation 6
(4S) -4-benzyl-3- { [ (3S, 4S) -1-benzyl-4- (5-chloropyridin-2-yl) pyrrolidin-3-yl]Carbonyls
Base } -1, 3-oxazolidin-2-ones
Trifluoroacetic acid (90 μ L, 1.2mmol) was added to a suspension of oxazolidinone from preparation 5 (1.93g, 5.6mmol) in dichloromethane (20mL) and then N-benzyl-N- (methoxymethyl) trimethylsilylamine (2.3mL, 9.0mmol) was added dropwise over 10 minutes. After the addition was complete the reaction was stirred at room temperature overnight. The reaction mixture was treated with saturated aqueous sodium bicarbonate (20mL), and the layers were separated. The aqueous layer was extracted with dichloromethane (2 × 20mL) and the combined organic layers were dried (MgSO)4) And evaporated. The residue was purified by column chromatography (silica gel) eluting with ethyl acetate/pentane 2:8, increasing the polarity to 2:3 to give the undesired (4S) -4-benzyl-3- { [ (3R, 4R) -1-benzyl-4- (5-chloropyridine, 2-yl) pyrrolidin-3-yl]Carbonyl } -1, 3-oxazolidin-2-one (1.16g, 44%) as the first eluting component, and the desired (4S) -4-benzyl-3- { [ (3S, 4S) -1-benzyl-4- (5-chloropyridin-2-yl) pyrrolidin-3-yl]Carbonyl } -1, 3-oxazolidin-2-one (1.18g, 45%) was the second eluting component.1H NMR(CDCl3,400MHz)δ2.75(2H,m),2.92(1H,m),3.20(3H,m),3.27(1H,br),3.68(2H,br),4.14(2H,m),4.23(1H,m),4.50(1H,m),4.67(1H,m),7.10-7.40(11H,m),7.58(1H,dd),8.50(1H,d);LRMS(APCI+)476[MH+].
Preparation 7
(3S, 4S) -1-benzyl-4- (5-chloropyridin-2-yl) pyrrolidine-3-carboxylic acid methyl ester
Sodium methoxide (664mg, 12mmol) was added to a solution of oxazolidinone (1.17g, 2.5mmol) and dimethyl carbonate (1.03mL, 12mmol) from preparation 6 in dichloromethane (15mL),the reaction was then stirred at room temperature overnight. The reaction mixture was concentrated in vacuo, and the residue was partitioned between ethyl acetate (50mL) and water (30 mL). The aqueous layer was neutralized by addition of 2M HCl (. about.6 mL) and then concentrated in vacuo. The residue was triturated with acetonitrile (25mL) and then filtered. The filtrate was concentrated to give (3S, 4S) -1-benzyl-4- (5-chloropyridin-2-yl) pyrrolidine-3-carboxylic acid (123mg, 16%) as a yellow solid (see preparation 8 for spectroscopic data). The ethyl acetate layer was dried (MgSO)4) And evaporated. The residue was purified by column chromatography (silica gel) eluting with ethyl acetate/pentane 2:8, increasing polarity to 2:3 to give the title compound (371mg, 45%) as a colorless oil.1H NMR(CDCl3,400MHz)δ2.71(1H,t),2.97(1H,t),3.05(2H,m),3.23(1H,m),3.63(5H,m),3.82(1H,q),7.15-7.35(6H,m),7.55(1H,d),8.46(1H,s);LRMS(APCI+)331[MH+].
Preparation 8
(3S, 4S) -1-benzyl-4- (5-chloropyridin-2-yl) pyrrolidine-3-carboxylic acid dihydrochloride
A solution of NaOH (135mg, 3.3mmol) in water (5mL) was added to a solution of the ester from preparation 7 (371mg, 1.1mmol) in dioxane (10mL) and the mixture was stirred at room temperature overnight. The reaction mixture was concentrated in vacuo, dissolved in water (10mL) and neutralized with 2M HCl (. about.1.7 mL). The mixture was then concentrated in vacuo, triturated with acetonitrile (20mL) and filtered. The filtrate was acidified with 2M HCl ether solution and then concentrated in vacuo to give the title compound (290mg, 68%) as a solid.1H NMR(CD3OD,400MHz)δ3.40-4.20(6H,m),4.53(2H,m),7.40-7.60(6H,m),7.81(1H,d),8.60(1H,br);LRMS(APCI+)317[MH+].
Preparation 9
(3R, 4R, 5S) -1- { [ (3S, 4S) -1-benzyl-4- (5-Chloropyridin-2-yl) pyrrolidin-3-yl]Carbonyls
-4- (4-fluorophenyl) -3, 5-dimethylpiperidin-4-ol
1-hydroxybenzotriazole (230mg, 1.7mmol) and 1- (3-dimethylaminopropyl) -3-ethyl-carbodiimide hydrochloride (354mg, 1.8mmol) were added to a solution of the acid from preparation 8 (522mg, 1.5mmol) in dichloromethane (10mL) and triethylamine (1.03mL, 7.4mmol), then the mixture was stirred at room temperature for 30 minutes, then (3R, 4S, 5S) -4- (4-fluorophenyl) -3, 5-dimethylpiperidin-4-ol hydrochloride (prepared according to US 2005/176772) (384mg, 1.5mmol) was added. The mixture was stirred at room temperature overnight, the solvent was removed in vacuo, and the residue was partitioned between ethyl acetate (50mL) and saturated aqueous sodium bicarbonate (50 mL). The organic layer was washed with brine (50mL), dried (MgSO4) and evaporated. The residue was purified by column chromatography (silica gel) eluting with dichloromethane, increasing polarity to 5% methanol in dichloromethane to give the title compound (546mg, 71%) as an oil.1H NMR(CD3OD,400MHz)δ0.3-0.6(6H,4x d),1.23(1H,m),1.75-1.95(2H,m),2.72(1H,t),2.85(1H,m),2.90-3.20(3H,m),3.45-4.05(5H,m),4.32(1H,d),7.02(3H,m),7.20-7.50(7H,m),7.80,(1H,dd),8.50(1H,d);LRMS(APCI+)522[MH+].
Preparation 10
(3R, 4R, 5S) -1- { [ (3S, 4S) -4- (5-Chloropyridin-2-yl) pyrrolidin-3-yl]Carbonyl } -4- (4-fluorobenzene)
Yl) -3, 5-dimethylpiperidin-4-ol
1-chloroethyl chlorideThe formate ester (0.3mL, 2.8mmol) was added to a solution of the amide from preparation 9 (370mg, 0.7mmol) and N-ethyldiisopropylamine (0.27mL, 1.6mmol) in dichloromethane (10mL), and the mixture was heated at reflux for 3 h. After cooling to room temperature, the solvent was removed in vacuo and the residue was partitioned between 10% aqueous citric acid (30mL) and dichloromethane (30 mL). The organic layer was washed with water (30mL) and dried (MgSO)4) And evaporated. The resulting dark oil was dissolved in methanol (10mL) and heated at reflux for 3 hours. The solvent was removed in vacuo, and the residue was purified by column chromatography (silica gel) eluting with 5% methanol in dichloromethane, increasing polarity to 10% methanol in dichloromethane to give the title compound (305mg, 100%) as an oil.1H NMR(CD3OD, 400MHz) δ 0.4-0.6(6H, 4x d), 1.00-1.06, 1.77-1.82 and 1.98-2.05(2H, 3x m), 2.76-2.82(1H, m), 3.00-3.20(2H, m), 3.40-4.10(6H, m), 4.36(1H, m)7.00-7.50(5H, m), 7.85 and 7.95(1H, 2x dd), 8.61 and 8.63(1H, 2x d); LRMS (APCI)+)432[MH+].
Preparation 11
(3S, 4S) -4- (5-Chloropyridin-2-yl) pyrrolidine-3-carboxylic acid methyl ester
1-chloroethyl chloroformate (2.33mL, 21.4mmol) was added to a solution of the ester from preparation 7 (1.77g, 5.35mmol) and N-ethyldiisopropylamine (2.1mL, 12mmol) in dichloromethane (10mL), and the mixture was heated at reflux for 3 hours. After cooling to room temperature, the solvent was removed in vacuo and the residue was dissolved in methanol (10mL) and heated at reflux for 16 h. The solvent was removed in vacuo and the residue was purified by column chromatography (silica gel) eluting with dichloromethane, increasing polarity to a solution of 10% methanol in dichloromethane to afford the desired product and N-ethyldiisopropylamine as an oil. The oil was dissolved in ethyl acetate (30mL)And the resulting precipitate was filtered. The filtrate was concentrated in vacuo, and the residue was dissolved in acetonitrile (25 mL). The resulting precipitate was filtered to give the title compound (619mg, 48%) as a white solid.1H NMR(CD3OD,400MHz)δ3.46(m,1H),3.61-3.77(m,7H),3.74(s,3H),3.98(m,1H),7.42(d.1H),7.82(dd,1H),8.57(d,1H);LRMS(APCI+)241[MH+].
Preparation 12
(3S, 4S) -1- (6-Chloropyridazin-3-yl) -4- (5-Chloropyridin-2-yl) pyrrolidine-3-carboxylic acid methyl ester
To a solution of pyrrolidine (350mg, 1.50mmol) from preparation 11 in dimethylsulfoxide (10mL) was added 3, 6-dichloropyridazine (330mg, 2.20mmol), triethylamine (0.61mL, 4.40mmol), and cesium fluoride (220mg, 1.45 mmol). The mixture was stirred overnight at 80 ℃ under nitrogen. The reaction mixture was dissolved in 25mL ethyl acetate and washed with 20mL water. The organic layer was separated and the aqueous layer re-extracted with 25mL of ethyl acetate. The combined ethyl acetate extracts were over MgSO4Drying, filtration and evaporation gave a light orange oily solid which was purified by column chromatography eluting with dichloromethane increasing polarity to 95/5 dichloromethane/methanol. The title compound was obtained as a yellow solid.1H NMR(CD3OD,400MHz)δ3.66(s,1H),3.72(m,1H),3.78(t,1H),3.96-4.12(m,4H),7.04(d,1H),7.42(m,2H),7.79(d,1H),8.52(s,1H);LRMS(El+)353[MH+].
Preparation 13
(3S, 4S) -4- (5-Chloropyridin-2-yl) -1- (6-oxo-1, 6-dihydropyridazin-3-yl) pyrrolidine-3-carboxylic acid
Methyl ester
A solution of chloropyridazine (803mg, 2.27mmol) from preparation 12 was dissolved in deoxygenated acetic acid and heated at reflux under nitrogen for 44 hours. The solvent was removed in vacuo and 15mL of methanol was added. HCl gas was then bubbled into the reaction mixture until saturated, then the mixture was stirred under a dry tube overnight. Methanol was removed in vacuo, and the residue was partitioned between DCM and 10% K2CO3In the meantime. The organic layer was separated over MgSO4Drying, filtration and evaporation gave the title compound as a pale brown solid (577mg, 76%).1H NMR(CD3OD, 400MHz) δ 3.58-3.76 and 3.83-3.97(6H, 2x m), 3.65(3H, s), 6.89(1H, d), 7.31(1H, d), 7.39(1H, d), 7.79(1H, dd), 8.52(1H, d); LRMS (EI)+)335[MH+].
Preparation 14
(3S, 4S) -4- (5-Chloropyridin-2-yl) -1- (1-methyl-6-oxo-1, 6-dihydropyridazin-3-yl) pyrrolidine
-3-Carboxylic acid methyl ester
To a solution of pyridazinone (557mg, 1.66mmol) from preparation 13 in dimethylformamide (10mL) were added sodium hexamethyldisilazide (1M in tetrahydrofuran, 2.00mL, 2.00mmol) and lithium bromide (173mg, 2.00 mmol). The reaction was stirred under nitrogen for 10 minutes, then methyl iodide (0.083mL, 1.30mmol) was added. The reaction was stirred under nitrogen for 4 hours and then partitioned between ethyl acetate (30mL) and water (30 mL). The organic layer was separated, dried over MgSO4, filtered and evaporated to give a brown oil which was purified using column chromatography (silica gel) eluting with 100% dichloromethane increasing polarity to 95/5 dichloromethane/methanol. The title compound was obtained as a yellow oil (500mg, 90%).1H NMR(CD3OD,400MHz)δ3.64(m,9H),3.92(m,3H),6.87(d,1H),7.26(d,1H),7.37(d,1H),7.79(dd,1H),8.51(s,1H);LRMS(EI+)349[MH+].
Preparation 15
(3S, 4S) -4- (5-Chloropyridin-2-yl) -1- (1-methyl-6-oxo-1, 6-dihydropyridazin-3-yl) pyrrolidine
-3-carboxylic acid
To a solution of ester 14 from preparation 14(500mg, 1.43mmol) in dioxane (10mL) was added 5mL of aqueous solution of sodium hydroxide (172mg, 4.30 mmol). The reaction was stirred under a dry tube overnight. The solvent was removed in vacuo and the residue was dissolved in water, neutralized with 4 equivalents of 2M HCl, and evaporated. The residue was stirred with 20mL acetonitrile and then filtered to give a pale yellow solid (512mg, containing 3 equivalents NaCl-338mg product +174mg NaCl).1H NMR(CD3OD,400MHz)δ3.6(m,2H),3.64(s,3H),3.7(t,1H),3.9(m,3H),6.88(d,1H),7.27(d,1H),7.41(d,1H),7.79(dd,1H),8.52(s,1H);LRMS(EI+)335[MH+].
Preparation 16
(3R, 4S, 5S) -4- (5-Chloropyridin-2-yl) -3, 5-dimethylpiperidin-4-ol
Step A: (3R, 4S, 5S) -4- (5-Chloropyridin-2-yl) -1- (4-methoxybenzyl) -3, 5-dimethylpiperidine
-4-alcohols
A solution of 2-bromo-5-chloropyridine (6.0g, 31.2mmol) in toluene (90mL) was cooled to-78 ℃ under nitrogen. N-butyllithium (2.5M in hexanes) (15mL, 37.5mmol) was added dropwise over 12 min, and the mixture was stirred at-78 deg.C for 1 h. A solution of (3R, 5S) -1- (4-methoxybenzyl) -3, 5-dimethylpiperidin-4-one (prepared according to International patent application publication No. WO2005/077935) (6.93g, 28.1mmol) in toluene (15mL) is then added dropwise over 10 minutes, and the mixture is stirred at-78 ℃ for a further 3 hours and then warmed to room temperature. The mixture was quenched by pouring saturated ammonium chloride (100mL), then stirred for 5 minutes and the mixture was partitioned between water (50mL) and ethyl acetate (300 mL). The organic phase was separated and the aqueous phase was extracted with ethyl acetate (2x300 mL). The combined organic extracts were dried over magnesium sulfate, filtered and evaporated to dryness to give the crude intermediate. Purification by chromatography (silica gel) eluting with 2% methanol in dichloromethane with increasing polarity to 10% (10:1 methanol: 880 ammonia) in dichloromethane afforded (3R, 4S, 5S) -4- (5-chloropyridin-2-yl) -1- (4-methoxybenzyl) -3, 5-dimethylpiperidin-4-ol as an orange oil (8.48g, 83%).
And B: (3R, 4S, 5S) -4- (5-Chloropyridin-2-yl) -3, 5-dimethylpiperidin-4-ol
The product from step A (6.56g, 18.2mmol) was dissolved in anhydrous dichloromethane (100mL), triethylamine (2.02g, 20.0mmol) was added, and the solution was cooled to 5 ℃ under nitrogen. 1-Chloroethyl chloroformate (3.1g, 21.9mmol) was added dropwise to the stirred solution, and after the addition was complete the mixture was stirred at room temperature for a further 2.5 hours. The mixture was then washed with 10% aqueous potassium carbonate (3 × 50mL), dried over magnesium sulfate and evaporated to dryness. The crude oil was heated in methanol (100mL) at reflux for 2.5 hours, then the solvent was removed in vacuo. The residue was dissolved in dichloromethane (100mL) and methanol (10mL), solid potassium carbonate (10g) was added, and the heterogeneous mixture was stirred for 30 minutes. The solid potassium carbonate was filtered off and the filtrate was evaporated to dryness. The crude product was then purified by column chromatography (silica gel) eluting with 10% methanol in dichloromethane, with increasing polarity to 20% (10:1 methanol: 880 ammonia) in dichloromethane to give the title compound (3.37g, 77%) as a yellowish solid.1H NMR(400MHz,CDCl3)δ0.53(3H, s), 0.57(s, 3H), 2.60-2.71(m, 2H), 3.13(q, 2H), 3.32(d, 2H), 7.43(d, 1H), 7.78(dd, 1H), 8.50(1H, d), 9.58(br, 1H), 9.84(br, 1H); LRMS (APCI +)241 and 243 MH+]
Preparation 17
5-fluoropyridine-2-carbaldehyde
The title compound was prepared according to the procedures for preparation 1 and 2, starting from 2-bromo-5-fluoropyridine. A crude product containing tetrahydrofuran and diethyl ether was obtained, which was used without further purification.1H NMR(CDCl3,400MHz)δ7.57(1H,dt),8.03(1H,dd),8.62(1H,d),10.04(1H,s);LRMS(APCI+)126[MH+].
Preparation 18
6-formyl-3-cyanopyridines
A mixture of 6-methyl-3-cyanopyridine (10.0g, 84.6mmol) and iodine (20.0g, 78.8mmol) in dimethylsulfoxide (150mL) was heated at 150 ℃ under nitrogen for 20 minutes (reaction exhaust gas (reaction exhaust) was purified with a bleaching agent to remove dimethylsulfide). After cooling to room temperature, saturated aqueous sodium bicarbonate (200mL) was carefully added and the resulting mixture was extracted with toluene (3 × 100 mL). The combined organic extracts were washed with brine and dried (MgSO)4) And evaporated to give the desired product as an orange oil (5.65g, 50%), which was used without further purification.1H NMR(CDCl3,400MHz)δ8.06(1H,d),8.17(1H,dd),9.05(1H,d),10.12(1H,s).
Preparation 19
5-methoxy-2-methylpyridine
6-methylpyridin-3-ol (50.0g, 0.458mol) was added to a suspension of powdered KOH (103g, 1.83mol) in dimethylsulfoxide (750mL), and the mixture was stirred at room temperature under nitrogen for 1.5 hours. Methyl iodide (30mL, 68.3g, 0.481mol) was then added dropwise to the dark brown mixture over 1 hour (exothermic). After stirring at room temperature for 1.5 h, water (1.0L) was added and the mixture was extracted with ethyl acetate (2 × 300 mL). The combined organic extracts were washed with brine and dried (MgSO)4) And evaporated in a rotary evaporator at 40 ℃. The residue was purified by column chromatography (silica gel) eluting with pentane, increasing the polarity to ethyl acetate to give the volatile product as a 1:1 mixture with ethyl acetate (40g,. about.23.3 g product, 41%)1H NMR(CDCl3,400MHz)δ2.45(3H,s),3.79(3H,s),7.02(1H,d),7.08(1H,dd),8.16(1H,d).
Preparation 20
5-methoxy-2-methylpyridine 1-oxide
M-chloroperbenzoic acid (51.3g, 0.297mol) was added in portions to a solution of preparation 19 compound (40g of a 1:1 mixture with ethyl acetate, 23.3g, 189mmol) in dichloromethane (1500mL) and the mixture was stirred at room temperature for 2 hours. A solution of sodium sulfite (45g) in water (250mL) was then added to the reaction and the mixture was stirred for 15 minutes at which time a starch/KI test paper performed for the presence of oxidant showed a negative test result. The organic layer was separated over MgSO4Drying and evaporating to obtain a lightA yellow solid (54g) which is a 1:1 mixture of the desired product and m-chlorobenzoic acid (mCBA). It was subjected to the following step without further purification.1H NMR(CDCl3,400MHz)δ3.84(3H,s),6.97(1H,dd),7.19(1H,d),7.34(1H,t,mCBA),7.48(1H,d,mCBA),7.94(1H,d,mCBA),8.04(1H,s,mCBA),8.36(1H,d).
Preparation 21
(5-methoxypyridin-2-yl) methanol
Trifluoroacetic anhydride (28.2mL, 203mmol) was added dropwise to an ice-cooled solution of the product from preparation 20 (-135 mmol) in dichloromethane (500mL), the mixture was warmed to room temperature and stirred overnight. Tlc analysis showed that there was still a significant amount of starting material, so an additional portion of trifluoroacetic anhydride (15mL, 108mmol) was added dropwise and the mixture was stirred for an additional 27 hours. The reaction was quenched by careful addition of methanol (250mL) and stirred for 30 min, then concentrated in vacuo. 2.5M sodium hydroxide (100mL) was then carefully added and the mixture extracted with dichloromethane (4X100 mL). The combined organic extracts were dried (MgSO)4) And evaporation gave the desired product (12g, 64%) which was contaminated with recovered starting material (. about.15 mol%).1H NMR(CDCl3,400MHz)δ3.80(3H,s),4.40(1H,br),4.66(2H,s),7.16(1H,dd),7.20(1H,d),8.17(1H,d).
Preparation 22
5-methoxypyridine-2-carbaldehyde (carbaldehyde)
MnO of2(97.0g, 360mmol) in portionsPortions were added to a solution of the alcohol from preparation 21 (12g, 86mmol) in dichloromethane (500mL) and the resulting suspension was stirred at room temperature for 64 hours. By passingThe reaction mixture was filtered and the solvent was removed in vacuo to give the desired product (8.6g, 73%) as an orange oil, which was contaminated with 5-methoxy-2-methylpyridine 1-oxide (15 mol%). 1HNMR (CDCl3, 400MHz) delta 3.93(3H, s), 7.26(1H, dd), 7.91(1H, d), 8.37(1H, d), 9.94(1H, s).
Claims (20)
1. A compound of formula (I) or a pharmaceutically acceptable salt thereof:
wherein
One of X and Y is N and the other is CH,
r is F, Cl, CN, CF3Or methoxy, with the proviso that when Y is N, R is not F or Cl,
R1is phenyl, 2-pyridyl, C3-C6Cycloalkyl or CH2(C3-C6Cycloalkyl), wherein the cyclic moiety is optionally substituted with one or more substituents independently selected from the group consisting of F, Cl, CN, methyl, and methoxy,
R2h, F or Cl, with the proviso that when Y is N, R2Instead of F or Cl, the catalyst is,
het is a 6-membered ring containing 1 or 2N atoms, wherein the ring is aromatic, or contains 2 double bonds and an ═ O substituent in the ring, the ring being optionally substituted with one or more substituents independently selected from F, Cl, OH, CN, methyl, ethyl, NH2、NHCH3、N(CH3)2And a methoxy group.
2. A compound according to claim 1, or a pharmaceutically acceptable salt thereof, wherein X is N and Y is CH.
3. A compound according to claim 1, or a pharmaceutically acceptable salt thereof, wherein R is chloro.
4. A compound according to any one of claims 1, 2 or 3, or a pharmaceutically acceptable salt thereof, wherein
R1Is phenyl optionally substituted with one or more substituents independently selected from F, Cl, CN, methyl and methoxy.
5. A compound according to claim 4, or a pharmaceutically acceptable salt thereof, wherein R1Is phenyl, 4-chlorophenyl or 4-fluorophenyl.
6. A compound according to any one of claims 1, 2 or 3, or a pharmaceutically acceptable salt thereof, wherein
R1Is C3-C6A cycloalkyl group.
7. A compound according to claim 6 or a pharmaceutically acceptable salt thereof, wherein
R1Is cyclopropyl or cyclohexyl.
8. A compound according to any one of claims 1, 2 and 3, or a pharmaceutically acceptable salt thereof, wherein R2Is H or F.
9. A compound according to claim 8, or a pharmaceutically acceptable salt thereof, wherein R2Is H.
10. A compound according to any one of claims 1, 2 and 3, or a pharmaceutically acceptable salt thereof, wherein
Het is pyridin-2-yl, pyridin-3-yl, pyridazin-3-yl, 6-oxo-1, 6-dihydropyridazin-3-yl, 6-oxo-1, 6-dihydropyridin-3-yl, 2-oxo-1, 2-dihydropyrimidin-4-yl, 6-oxo-1, 6-dihydropyrimidin-4-yl, 2-oxo-1, 2-dihydropyridin-4-yl or 6-oxo-1, 6-dihydropyridin-2-yl, optionally substituted with one or more substituents independently selected from F, Cl, OH, CN, methyl, ethyl and methoxy.
11. A compound according to claim 10, or a pharmaceutically acceptable salt thereof, wherein Het is pyridin-2-yl, pyridin-3-yl, pyridazin-3-yl or 6-oxo-1, 6-dihydropyridazin-3-yl, optionally substituted with one or more substituents independently selected from OH, CN, F, methyl and methoxy.
12. A compound according to claim 11, or a pharmaceutically acceptable salt thereof, wherein Het is pyridin-2-yl or pyridazin-3-yl, each substituted at the para position relative to the bond to the pyrrolidine moiety with OH, CN or methoxy.
13. A compound according to claim 12, or a pharmaceutically acceptable salt thereof, wherein Het is pyridazin-3-yl substituted at the para position relative to the bond attached to the pyrrolidine moiety with OH, CN or methoxy.
14. A compound according to claim 1, selected from:
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4-hydroxy-3, 5-dimethyl-4-phenylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- (5-Chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4-hydroxy-3, 5-dimethyl-4-pyridin-2-ylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4-hydroxy-3, 5-dimethyl-4-phenylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4-hydroxy-3, 5-dimethyl-4-phenylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- { [ (3R, 4R, 5S) -4- (4-chlorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } -4- (5-chloropyridin-2-yl) pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-Chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (5-Chloropyridin-2-yl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (3, 4-difluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4-hydroxy-4- (4-methoxyphenyl) -3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4-cyclohexyl-4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
(3R, 4R, 5S) -1- { [ (3S, 4S) -1- (6-chloropyridazin-3-yl) -4- (5-chloropyridin-2-yl) pyrrolidin-3-yl ] carbonyl } -4- (4-fluorophenyl) -3, 5-dimethylpiperidin-4-ol;
(3R, 4R, 5S) -1- { [ (3S, 4S) -4- (5-chloropyridin-2-yl) -1- (6-methoxypyridazin-3-yl) pyrrolidin-3-yl ] carbonyl } -4-cyclopropyl-3, 5-dimethylpiperidin-4-ol;
(3R, 4R, 5S) -1- { [ (3S, 4S) -4- (5-chloropyridin-2-yl) -1- (5-fluoropyridin-3-yl) pyrrolidin-3-yl ] carbonyl } -4- (4-fluorophenyl) -3, 5-dimethylpiperidin-4-ol;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -3-cyanopyridine;
6- [ (3S, 4S) -3- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } -4- (5-fluoropyridin-2-yl) pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- (5-fluoropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4-hydroxy-3, 5-dimethyl-4-phenylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- { [ (3R, 4R, 5S) -4- (4-chlorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } -4- (5-fluoropyridin-2-yl) pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- { [ (3R, 4R, 5S) -4- (4-chlorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } -4- (5-fluoropyridin-2-yl) pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-cyanopyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4R) -3- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } -4- (6-methoxypyridin-3-yl) pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } -4- (5-methoxypyridin-2-yl) pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
(3R, 4R, 5S) -4- (4-fluorophenyl) -1- { [ (3S, 4R) -1- (5-fluoropyridin-3-yl) -4- (6-methoxypyridin-3-yl) pyrrolidin-3-yl ] carbonyl } -3, 5-dimethylpiperidin-4-ol;
(3R, 4R, 5S) -1- { [ (3S, 4S) -4- (5-chloropyridin-2-yl) -1-pyridazin-3-ylpyrrolidin-3-yl ] carbonyl } -4- (4-fluorophenyl) -3, 5-dimethylpiperidin-4-ol;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
(3R, 4R, 5S) -1- { [ (3S, 4S) -4- (5-chloropyridin-2-yl) -1- [1, 2, 4] triazolo [4, 3-b ] pyridazin-6-ylpyrrolidin-3-yl ] carbonyl } -4- (4-fluorophenyl) -3, 5-dimethylpiperidin-4-ol;
(3R, 4R, 5S) -1- { [ (3S, 4S) -4- (5-chloropyridin-2-yl) -1-imidazo [1, 2-b ] pyridazin-6-ylpyrrolidin-3-yl ] carbonyl } -4- (4-fluorophenyl) -3, 5-dimethylpiperidin-4-ol;
4- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyrimidin-2 (1H) -one;
4- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -1-methylpyrimidin-2 (1H) -one;
(3R, 4R, 5S) -1- { [ (3S, 4S) -4- (5-chloropyridin-2-yl) -1- (6-methoxypyridazin-3-yl) pyrrolidin-3-yl ] carbonyl } -4- (4-fluorophenyl) -3, 5-dimethylpiperidin-4-ol;
6- [ (3S, 4S) -3- (5-chloro-3-fluoropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- (5-chloro-3-fluoropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-chloro-3-fluoropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (3, 5-difluoropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- (3, 5-difluoropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (3, 5-difluoropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- { [ (3R, 4R, 5S) -4- (4-chlorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } -4- (5-chloropyridin-2-yl) pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- (5-Chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (3, 4-difluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- { [ (3R, 4R, 5S) -4- (4-chlorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } -4- (5-chloropyridin-2-yl) pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-Chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (3, 4-difluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
or a pharmaceutically acceptable salt thereof.
15. A compound according to claim 14, selected from:
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4-hydroxy-3, 5-dimethyl-4-phenylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4-hydroxy-3, 5-dimethyl-4-phenylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4-hydroxy-3, 5-dimethyl-4-phenylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- { [ (3R, 4R, 5S) -4- (4-chlorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } -4- (5-chloropyridin-2-yl) pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-Chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (5-Chloropyridin-2-yl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (3, 4-difluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
(3R, 4R, 5S) -1- { [ (3S, 4S) -4- (5-chloropyridin-2-yl) -1-pyridazin-3-ylpyrrolidin-3-yl ] carbonyl } -4- (4-fluorophenyl) -3, 5-dimethylpiperidin-4-ol;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
(3R, 4R, 5S) -1- { [ (3S, 4S) -4- (5-chloropyridin-2-yl) -1- [1, 2, 4] triazolo [4, 3-b ] pyridazin-6-ylpyrrolidin-3-yl ] carbonyl } -4- (4-fluorophenyl) -3, 5-dimethylpiperidin-4-ol;
(3R, 4R, 5S) -1- { [ (3S, 4S) -4- (5-chloropyridin-2-yl) -1-imidazo [1, 2-b ] pyridazin-6-ylpyrrolidin-3-yl ] carbonyl } -4- (4-fluorophenyl) -3, 5-dimethylpiperidin-4-ol;
4- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyrimidin-2 (1H) -one;
4- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -1-methylpyrimidin-2 (1H) -one;
6- [ (3S, 4S) -3- (5-chloro-3-fluoropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- (5-chloro-3-fluoropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-chloro-3-fluoropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (3, 5-difluoropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- (3, 5-difluoropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (3, 5-difluoropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- { [ (3R, 4R, 5S) -4- (4-chlorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } -4- (5-chloropyridin-2-yl) pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- (5-Chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (3, 4-difluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- { [ (3R, 4R, 5S) -4- (4-chlorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } -4- (5-chloropyridin-2-yl) pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-Chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (3, 4-difluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
or a pharmaceutically acceptable salt thereof.
16. A compound according to claim 15, selected from:
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- { [ (3R, 4R, 5S) -4- (4-chlorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } -4- (5-chloropyridin-2-yl) pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-Chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (5-Chloropyridin-2-yl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (3, 4-difluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (4-fluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] -2-methylpyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- { [ (3R, 4R, 5S) -4- (4-chlorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } -4- (5-chloropyridin-2-yl) pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- (5-Chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (3, 4-difluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazine-3-carbonitrile;
6- [ (3S, 4S) -3- { [ (3R, 4R, 5S) -4- (4-chlorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } -4- (5-chloropyridin-2-yl) pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
6- [ (3S, 4S) -3- (5-Chloropyridin-2-yl) -4- { [ (3R, 4R, 5S) -4- (3, 4-difluorophenyl) -4-hydroxy-3, 5-dimethylpiperidin-1-yl ] carbonyl } pyrrolidin-1-yl ] pyridazin-3 (2H) -one;
or a pharmaceutically acceptable salt thereof.
17. A pharmaceutical composition comprising a compound of any one of claims 1, 2, 3, 4, 5,6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or 16, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier or adjuvant.
18. A pharmaceutical composition comprising a compound of claim 1 and a diluent.
19. Use of a compound of any one of claims 1, 2, 3, 4, 5,6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or 16, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 17 or 18, in the manufacture of a medicament for the treatment of a disease, disorder or condition responsive to activation of the MC4 receptor.
20. Use according to claim 19, wherein the disease is sexual dysfunction, obesity, diabetes or lower urinary tract dysfunction.
Applications Claiming Priority (5)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US77629506P | 2006-02-23 | 2006-02-23 | |
| US60/776,295 | 2006-02-23 | ||
| US88784007P | 2007-02-02 | 2007-02-02 | |
| US60/887,840 | 2007-02-02 | ||
| PCT/IB2007/000456 WO2007096763A2 (en) | 2006-02-23 | 2007-02-19 | Melanocortin type 4 receptor agonist piperidinoylpyrrolidines |
Publications (2)
| Publication Number | Publication Date |
|---|---|
| HK1126489A1 HK1126489A1 (en) | 2009-09-04 |
| HK1126489B true HK1126489B (en) | 2013-08-02 |
Family
ID=
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| KR101036981B1 (en) | Melanocortin Type 4 Receptor Agonist Piperidinoylpyrrolidine | |
| KR102032007B1 (en) | Nitrogen-containing aromatic heterocyclic compound | |
| CN102171206B (en) | Diazepine and diazocane compounds as MC4 agonists | |
| CN112673001B (en) | Indazole carboxamides as kinase inhibitors | |
| JP2009503050A (en) | Piperidinoyl-pyrrolidine and piperidinoyl-piperidine compounds | |
| CN103140489A (en) | Urea derivatives and their therapeutic use in the treatment of, inter alia, diseases of the respiratory tract | |
| JP5243274B2 (en) | Piperidinoylpyrrolidine, a type 4 melanocortin receptor agonist | |
| HK1126489B (en) | Melanocortin type 4 receptor agonist piperidinoylpyrrolidines | |
| HK1158619B (en) | Diazepine and diazocane compounds as mc4 agonists | |
| HK1176062A1 (en) | Pyrazole compounds as crth2 antagonists | |
| HK1176062B (en) | Pyrazole compounds as crth2 antagonists |