MXPA06011123A - Tetraazabenzo[e]azulene derivatives and analogs thereof - Google Patents

Tetraazabenzo[e]azulene derivatives and analogs thereof

Info

Publication number
MXPA06011123A
MXPA06011123A MXPA/A/2006/011123A MXPA06011123A MXPA06011123A MX PA06011123 A MXPA06011123 A MX PA06011123A MX PA06011123 A MXPA06011123 A MX PA06011123A MX PA06011123 A MXPA06011123 A MX PA06011123A
Authority
MX
Mexico
Prior art keywords
alkyl
phenyl
group
alkoxy
halo
Prior art date
Application number
MXPA/A/2006/011123A
Other languages
Spanish (es)
Inventor
Hammond Marlys
O Keefe Cameron Kimberly
Louis Elliott Richard
Original Assignee
Pfizer Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Pfizer Inc filed Critical Pfizer Inc
Publication of MXPA06011123A publication Critical patent/MXPA06011123A/en

Links

Abstract

This invention relates to CCK-A agonists of Formula (I) wherein R1-R4, A, B, X, D, E and G are as defined in the specificiation, as well as, among other things, pharmaceutical compositions containing the compounds and methods of use of the compounds and compositions. The compounds are useful in treating obesity.

Description

DERIVATIVES OF TETRAAZABENZOI? 1AZULENE AND ITS ANALOGS FIELD OF THE INVENTION This invention relates to compounds of formula (I), pharmaceutical compositions comprising the compounds, either alone or in combination with other pharmaceutical agents, methods for using the compounds and combinations thereof. intermediates and methods useful in the preparation of the compounds. The compounds of formula (I) are cholesystokinin A receptor agonists (CCK-A) and, therefore, are useful, for example, for weight management and the treatment of obesity and associated diseases. BACKGROUND OF THE INVENTION Obesity is a major public health concern because of its increased prevalence and associated health risks. In addition, obesity can affect the quality of life of a person through limited mobility and decreased physical resistance as well as through social, academic and employment discrimination. Obesity and overweight are generally defined by the body mass index (BMI) that is correlated with total body fat and serves as a measure of risk for certain diseases. The BMI is calculated by the weight in kilograms divided by the height in square meters (kg / m2). Overweight is usually defined as a BMI of 25-29.9 kg / m2, and obesity is usually defined as a BMI of 30 kg / m2 or more. See, for example, National Heart, Lung, and Blood Institute, Clinical Guidelines on the Identification, Evaluation, and Treatment of Overweight and Obesity in Adults, The Evidence Report, Washington, DC: U.S. Department of Health and Human Services, NIH publication no. 98-4083 (1998). Recent studies have found that obesity and its associated health risks are not limited to adults, but also affect children and adolescents to a surprising degree. According to the Center for Disease Control, the percentage of children and adolescents who are defined as overweight has been more than double than since the beginning of the 70s, and approximately 15 percent of children and adolescents are currently overweight. Risk factors for heart disease, such as high cholesterol and high blood pressure, occur with an increased frequency in overweight children and adolescents compared to subjects with normal weight of similar age. Also, type 2 diabetes, previously considered an adult diabetes, has increased enormously in children and adolescents. The conditions of overweight and obesity are closely linked to type 2 diabetes. It has recently been estimated that overweight adolescents have a 70% chance of becoming overweight or obese adults. The probability increases to approximately 80% if at least one relative is overweight or obese. The most immediate consequence of being overweight, as perceived by the children themselves, is social discrimination. There are possible adverse health consequences from being overweight or being obese, as these individuals have a risk increased ailments (co-morbidities) such as hypertension, dyslipidemia, type 2 diabetes (non-insulin dependent), insulin resistance, glucose intolerance, hyperinsulinemia, coronary heart disease, angina pectoris, congestive heart failure, stroke , gallstones, cholescystitis, cholelithiasis, gout, osteoarthritis, obstructive sleep apnea and respiratory problems, gallbladder disease, certain forms of cancer (for example, endometrial, breast, prostate and colon) and psychological disorders (such as depression, eating disorders, distorted body image and low self-esteem). The negative health consequences of obesity make it the second leading cause of death that can be prevented in the United States and confer a significant economic and psychosocial effect on society. See McGinnís M, Foege WR, "Current Causes of Death in the United States," JAMA, 270, 2207-12 (1993). Obesity is currently recognized as a chronic disease that requires treatment to reduce its associated health risks. Although weight loss is an important consequential treatment, one of the main objectives of obesity management is to improve cardiovascular and metabolic indexes in order to reduce the morbidity and mortality related to obesity. It has been shown that 5-10% loss of body weight can substantially improve metabolic rates, such as blood glucose, blood pressure and lipid levels. Therefore, it is believed that a 5-10% intentional reduction in body weight can reduce morbidity and mortality.
Prescription drugs currently available to treat obesity generally reduce weight by inducing mainly satiety or decreasing the absorption of dietary fat. Satiety is achieved by increasing the synaptic levels of norepinephrine, serotonin, or both. For example, stimulation of subtypes 1 B, 1D and 2C of serotonin receptors and 1- and 2-adrenergic receptors decreases food intake by regulating satiety. See Bray GA, "The New Era of Drug Treatment, Phannacologic Treatment of Obesity: Symposium Overview," Obes Res., 3 (suppl 4), 415s-7s (1995). Adrenergic agents (eg, diethylpropion, benzophetamine, phendimetrazine, mazindol and phentermine) act by modulating the central norepinephrine and dopamine receptors through the promotion of catecholamine release. Previous adrenergic weight loss drugs (eg, amphetamines, methamphetamines, and phenmetrazine), which are strongly involved in dopamine trajectories, are no longer recommended due to the risk of their abusive use. Fenfluramine and dexfenfluramine, which are both serotonergic agents used to regulate appetite, are no longer available for use. Cholecystokinin (CCK) is a brain-gut peptide that acts as a gastrointestinal, neuratransmitting, and neuromodulating hormone in the central and peripheral nervous systems. Cholecystokinin is a peptide that exists in multiple active forms of variable lengths (for example, CCK-58, CCK-39, CCK-33, CCK-8 and CCK-4) with different predominant forms in different species. The cholecystokinin-58 is the main molecular form in the intestine of man, dog and cat but not in the pig, cattle or rat. See, for example, G.A. Eberlien, V.E. Eysselein and H. Goebell, 1988, Peptides 9, p. 993-998. The peripheral effects of CCK, of which the O-sulfated octapeptide CCK-8S is believed to be the predominant form, are focused on its function as a gastrointestinal satiety factor. It has been shown that CCK is released from mucosal cells I of the duodenum and jejunum in response to food, particularly in response to fats or proteins in food. Once released, the CCK initiates a certain number of coordinated responses to promote digestion and regulate food intake, which include mediating the emptying of bile from the gallbladder, regulating the release of digestive enzymes from the pancreas, controlling the emptying Gastric by regulation of the pyloric sphincter, as well as neuronal signaling for the CNS (Central Nervous System) through the vagal afferent neurons. In the CNS, the CCK has been found in numerous anatomical locations, including the cerebral cortex, hippocampus, septum, tonsils, olfactory bulbs, hypothalamus, thalamus, parabranchial nuclei, raphe nucleus, substantia nigra, ventral mesencephalon, solitary tract nucleus, ventral medulla and spinal cord. See, for example, T. Hokfeit et al., 1988, J. Chem. Neuroanat. 1, pag. 11-52; J.J. Vanderhaeghen, J.C. Signeu and W. Gepts, 1975, Nature 257, p. 604-605; and J-J. Vanderhaegen and S.N. Schiffmann (1992) p. 38-56, Eds. C.T. Dourish, S.J. Cooper, S.D. Iversen and L.L.
Iversen, Oxford University Press, Oxford. The neuronal CCk is believed to mediate a number of events in the CNS, including the dopaminergic modulation nuerotransmission and anxiolytic effects, and also affects cognition and nociception. See, for example, J.N. Crawley and R.L. Corwin, 1994, Peptides, 15: 731-755; N.S. Baber, C.T. Dourish, and D.R. Hill, Pain (1989), 39 (3), 307-28; and P. De Tullio, J. Delarge and B. Pirotte, Expert Opinion on Investigational Drugs (2000), 9 (1), 129-146. Cholecystokinin has also been shown to mediate its various hormonal and neuromodulatory functions through two subtypes of receptors: the CCK-A (CCKi) and CCK-B (CCK2) subtypes (see, for example, GN Woodruff and J. Hughes, Annu, Rev. Pharmacol, Toxicol. (1991), 31: 469-501) who have both been sequenced and cloned from rats (see, for example, SA Wank et al. (1992) Proc. Acad. Sci. USA, 89, 8691-8695) and humans (see, for example, JR Pisegna et al., 1992, Biochem Biophys., Res. Commun. 189, pp. 296-303). The two receptor subtypes CCK-K and CCK-B belong to the seven superfamilies of receptors coupled to the transmembrane G protein. The nucleotide sequences of the peripheral CCK-A receptor and central CCK-A receptor are the same in humans; similarly, the human CCK-B receptor and the gastrin receptor have been found to be the same. See, for example, S.A. Wank et al., (1994), NY Acad. Sci. 713, p.49-66.
The CCK-A receptor is located predominantly in the periphery, including pancreatic azine cells, pyloric sphincter, gallbladder and vagal afferents, where pancreatic exocrine secretion mediates gastric emptying and contraction of the gallbladder, and transmits signals of satiety post-prandial to the CNS. In addition, the CCK-A receptor is located in discrete zones in the CNS, including the solitary tract nucleus, postrema area and the mid dorsal hypothalamus. The CCK-B receptor is located predominantly in the CNS, and is less predominant in the periphery. A number of studies suggest that CCK mediates the satiety effect through the CCK-A receptor, which diffuses the postprandial satiety signal through the vagal afferents to the CNS. See, for example, G.P. Smith et al., Science 213 (1981) p. 1036-1037; and J.N. Crawley et al., J. Pharmacol. Exp. Ther., 257 (1991) p. 1076-1080. For example, it has been described that CCK and CCK agonists can decrease the ingestion of foods in animals, including rats (see, for example, J. Gibbs, RC Young and GP Smith, 1973, J. Comp.Physiol.Sychol. 84: 488-95), dogs and primates (including man) (see, for example, BA Himick and RE Peter, 1994, Am. J. Physiol. 267.R841-R851; Y. Hirosue et al., 1993, Am. J. Physiol. 265: R481-R486; and KE Asin et al., 1992, Pharmacol. Biochem. Behav. 42: 699-704), and that this anorexic effect is mediated through the CCK-A receptor located in vagal afferent fibers (see, for example, CT Dourish, 1992, In Multiple cholecystokinin receptors in the CNS, CT Dourish, SJ Cooper, SD Iversen and LL Iversen, editors, Oxford University Press, New York, NY, p. 234-253; G.P. Smith and J. Gibbs, 1992, In Multiple cholecystokinin receptors in the CNS, C. T. Dourish, S.J. Cooper, S.D. Iversen and L.L. Iversen, editors, Oxford University Press, New York, NY, p. 166-182; J.N. Crawley and R.L. Corwin, 1994, Peptides, 15: 731-755; and G.P. Smith et al., 1981, Science 213, p.1036-1037). Other lines of evidence supporting the intervention of the CCK-A receptor in the regulation of food intake include the discovery that OLTF rats (lacking the CCK-A receptor) are insensitive to the anorexigenic action of CCK. Also, it has been described that CCK-A selectable antagonisms, but not CCK-B antagonists, block the anorexic actions of CCK and CCK analogues and increase feeding in animals (see, eg, G. Hewson et al. al., 1988, Br. J. Pharmacol. 93: 79-84.; R.D. Reidelberger and M.F. O'Rourke, 1989, Am. J. Physiol. 257: R1512-R1518; T.H. Moran et al., 1993, Am. J. Physiol. 265: R620-R624; and M. Covasa and R.C. Ritter, Peptides (New York, NY, US) (2001), 22 (8), 1339-1348) including humans (see, for example, OM Wolkowitz et al., 1990, Biol. Psychiatry, 28: 169-173 ). Finally, it has been described that the infusion of CCK or selective agonists of CCK-A reduces the ingestion in quantity and calories of food in animals, including humans (see, for example, L. Degen et al., Peptides (New York). , NY) (2001), 22 (8), 1265-1269, HR Kissileff et al., Am. J. Clin. Nutr. 34 (1981), pp. 154-160; A. Ballinger et al., Clin Sci. 89 (1995), 375-381; and RJ Lieverse et al., Gastroenterology 106 (1994), 1451-1454).
The development of non-peptide CCK-A agonists has been described in the literature. For example, Sanofi has described in U.S. Pat. No. 5,798,353 that certain 3-acylamino-5- (polysulfuric phenyl) -1,4-benzodiazepin-2-ones act as agonists of CCK-A. Certain 1, 5-benzodiazepinones have been described as being CCK-A agonists having anorexic activity in rodents (see, for example, EE Sugg. Al., (1998) Pharmaceutical Biotechnology 11 (InÃgragration of Pharmaceutical Discovery and Development): 507 -524). R.G. Sherrill et al., N Bioorganic & Medicinal Chemist Letters (2001), 11 (9), 1145-1148 describe certain 1,4-benzodiazepines which are agonists of peripheral CCK-A receptors with anorexic activity in rat feeding models. A series of 3- (1H-indazol-3-ylmethyl) -1,5-benzodiazepines is described by B.R. Henke went to. in J. Med. Chem. (1997), 40 (17), 2706-2725 and J. Med. Chem. (1996), 39 (14), 2655-2658 which are orally active CCK-A agonists. Although ongoing research is still ongoing, there is still a need for a more effective and safe therapeutic approach to reduce or prevent weight gain. SUMMARY OF THE INVENTION The present invention relates to a compound of formula (I) ( wherein A, B, X, D, E and G are independently -C (R5) - or - N-; with the proviso that not more than two of A, B, X and D are N at the same time and at least one of E and G is N; R1 is selected from the group consisting of (C2-C6) alkyl, halo (C6-C6) halo-substituted, alkylamino (Ci-Cß), di (alkyl d-Cß) amino, alkylamino (Ci-CβJ-alkyl) (Ci-Cd), di-alkylamino (d-CeJ-alkyl (Ci-Cß), aryl, aryl-alkyl (C? -C6), heleroaryl-A, heeroaryl-A-alkyl (Ci-Cß), a heterocycle -A from 4 to 7 partial members or safned complememenie, a heierociclil-A-alkyl (Ci-Cß) of 4 to 7 members partially or fully saturated or cycloalkyl (C3-C) -alkyl (Ci-Cß) partially or completely saturated and, when none of R6 and R7 is phenylmethyl, R1 is selected from said group and partially or fully saturated cycloalkyl (C3-C7), wherein heeroaryl-A is selected from the group consisting of thienyl, thiazolyl, isoiazolyl, indolyl, 2-pyridyl, pyridazinyl, pyrimidyl, oxazolyl, furanyl, imidazolyl, isoxazolyl, quinolinyl, isoquinolinyl, benzoyl phenyl, benzofuranyl, pyrazinyl and pyrazolyl, and the partially or fully saturated helerocycle A Urea is selected from the group consisting of pyranyl, morpholinyl and teirahydrofuranyl, and wherein the aryl, heeroaryl-A, partially or fully saturated helerocyclo-A or the partial or complete cycloalkyl group saturated or from a group is optionally susiiuid with 1 to 3 subsitutes independently selected from the group consisting of halo, (C1-C3) alkoxy, (C1-C3) alkoxy halo-suslifuido, -OH, (C1-C3) alkyl. -CN and (C1-C3) alkyl halo-susiluido; R2 is -CH2C (O) N (R6) (R7); one of R3 and R4 is H, halo, alkyl (CI-CT), alkoxy (Ci-Ce) or a partially or fully saturated cyano (C3-C7) cycloalkyl and the other of R3 and R4 is-C (R8) (R9) ) (R10); or R3 and R4 are taken together to form = CHR11; each R5 is independently selected from the group consisting of H, alkoxy (d-Ce), -OH, halo, -CN, -NH2 and -NO2; one of R6 and R7 is (C3-C6) alkyl or a (C3-C7) cycloalkyl partially or fully saturated and the other of R6 and R7 is phenyl optionally substituted with 1 to 3 substitutes independently selected from the group consisting of -OH , -CN, halo, halo-substituted (Ci-Cβ) alkyl, halo-substituted (C 1 -C 3) alkoxy, (C C β) alkyl and (C 1 -C 3) alkoxy; phenylmethyl in which the phenyl moiety is optionally substituted with 1 to 3 substituents independently selected from the group consisting of -OH, -CN, halo-substituted alkyl (Ci-Cβ), halo-substituted alkoxy (C 1 -C 3) and alkoxy (C1-C3); or heteroaryl-B optionally substituted with 1 to 3 substituents independently selected from the group consisting of -OH, -CN, halo, halo-substituted (C -Cß) alkyl, halo-substituted (C1-C3) alkoxy, alkyl (d-) Cß) and (C 1 -C 3) alkoxy; and wherein heteroaryl-B is selected from the group consisting of thienyl, thiazolyl, isothiazolyl. isoquinolinyl, quinolinyl, 3- or 4-pyridyl, pyrimidyl, oxazolyl, furanyl, imidazolyl, isoxazolyl, pyrazinyl, pyridazinyl and pyrazolyl; two of R8, R9 and R10 are independently H or alkyl (Ci-Cß) and the other of R8, R9 and R10 is phenyl, a partially or fully saturated cycloalkyl (C3-C7), heteroaryl-C or a heterocycle-B of 4 to 7 members partially or completely saturated, wherein heteroaryl-C is selected from the group consisting of indol-2-yl, indol-3-yl, indazol-3-yl, 7-azaindol-2-yl and 7- azaindol-3-yl; and said phenyl, partially or fully saturated cycloalkyl, heteroaryl-C or partially or fully saturated heterocycle-B is optionally substituted on the carbon atom (s) with 1 to 3 substituents independently selected from the group consisting of alkoxy (C? -C6), F, Cl, -CN, -OH, -CO2H, tetrazole and halo-substituted (C? -C6) alkoxy; and R11 is phenyl, a partially or fully saturated cycloalkyl (C3-C7), heteroaryl-C or a 4- to 7-membered heteroaryl-B or fully saturated system, in which heeroaryl-C is selected from the group consisting of indole 2-yl, indol-3-yl, indazol-3-yl, 7-azaindol-2-yl and 7-azaindol-3-yl; said phenyl, partially or fully saturated cycloalkyl, heteroaryl-C or partially or completely saturated heterocycle-B is optionally susiiorated on the carbon atom (s) with 1 to 3 substitutes independently selected from the group consisting of alkoxy ( Ci-Cβ), F, Cl, -CN, -OH, -CO 2 H, εefrazol and alkoxy (Ci-Cβ) halo-susíiuuido; Y a pharmaceutically acceptable salt thereof or a prodrug of said compound or said salt. In one aspect of the invention, one of A, B, X and D in formula (I) is -N- and the others of A, B, X and D are -C (R5) -. In a further aspect, two of A, B, X and D are -N- and the others of A, B, X and D are -C (R5) -. In a preferred embodiment, each of A, B, X and D in formula (I) is -C (R5) -, as set forth in formula (II) below.
Each of the substituents R1, R2, R3, R4 and R5 and each of the ring atoms E and G for the compounds of formula (II) are as defined above and subsequently, generally and preferably, for the compounds of formula (I). In one embodiment of the invention, E is -N- and G is -C (R5) - in the formula (I). In another embodiment, E is -C (R5) - and G is -N-. Preferably, each of E and G is -N-. The susfiíuyeníes and other parameters for these modalities are as previously defined and subsequently, general and preferentially, for the compounds of formula (I). A preferred embodiment of the invention refers to the compounds of formula (I) wherein A and D are -CH-, X and B are -C (R5) - and E and G are -N- as set out in the formula (lll) later. Each of the substituents in the formula (III) is as defined above and subsequently, generally and preferably, for the compounds of formula (I). (ni) R1 in the formula (I) is preferably selected from the group consisting of (C2-C6) alkyl, -CF3, phenyl, phenyl-(C1-C3) alkyl, heteroaryl-A, heleroaryl-A-alkyl ( C1-C3), a 4 to 7 membered heteroaryl-A partially or completely saturated, a 4 to 7 membered heteroaryl-C1-C3 alkyl (partially or fully saturated) and a (C3-C7) cycloalkyl-alkyl ( C1-C3) partially or completely saturated, when none of R6 and R7 is phenylmeryl, R1 is also preferably a partially or fully salted (C3-C7) cycloalkyl. More preferably, R is phenyl or 2-pyridyl or, when none of R6 and R7 is phenylmeryl, R1 is also more preferably a (C5-C7) cycloalkyl partially or completely saturated. Still more preferably, R1 is phenyl or cyclohexyl when none of R6 and R7 is phenylmethyl. Most preferably, R1 is phenyl. R1 heteroaryl-A is preferably selected from the group consisting of nicienyl, 2-pyridyl, pyridazinyl, pyrimidyl, pyrazinyl and pyrazolyl.
More preferably, it will be a 2-pyridyl group.
The aryl R1, heteroaryl-A, partially or completely saturated heterocycle or the partially or fully saturated cycloalkyl group or a part of a group is optionally substituted, preferably with 1 to 3 substituents independently selected preferably from the group consisting of F, Cl, (C1-C3) alkoxy, -OH, (C1-C3) alkyl, -CN and -CF3; more preferably, the group consisting of F, Cl, (C1-C3) alkoxy, -OH, (C1-C3) alkyl, and -CF3; Especially enlre the group consisting of F, Cl, (C1-C3) alkoxy, -OH and (C1-C3) alkyl. When R1 is phenyl, it is most preferably unsubstituted, but if substituted, it is most preferably substituted with 1 to 3 F atoms or an -OH group. Each R 5 in the formula (I) is preferably selected from the group consisting of H, alkoxy (d-C), -OH, F, Cl and -CN; more preferably, include the group consisting of H, -OH and F; still more preferably between H and F. Most preferably, each R 5 is H. When each of A, B, X and D in the formula (I) is -C (R 5) -, preferably at least one of the R 5 is H, more preferably at least two or more of esters R5 are H. Most preferably, each of these R5 is H. When one of E and G in formula (I) is -C (R5) -, that is R5 is Also preferably, one of R6 and R7 in the formula (I) is a branched chain (C3-C6) alkyl, more preferably a (C3-C5) alkyl of branched chain; still more preferably a branched chain alkyl (C3 or C) and, most preferably, isopropyl. The other of R6 and R7 in the formula (I) is preferably phenyl, phenylmethyl or heteroaryl-B wherein the phenyl group, the phenyl part of the phenylmethyl group or the heteroaryl-B group is optionally substituted. Preferably, the phenyl group of R6 or R7 is optionally susiiluted with 1 to 3 independently selected from the group consisting of -OH, -CN, F, Cl, (C1-C3) alkyl substituted with F- or Cl-, alkoxy (C1-C3) substituted with F- or Cl-, (C1-C4) alkyl and (C1-C3) alkoxy. More preferably, the phenyl group is optionally substituted with 1 to 3 substituents independently selected from the group consisting of -OH, -CN, F, Cl, (C1-C3) alkyl substituted with F, (C1-C3) alkoxy substituted with F, (C 1 -C 4) alkyl and (C 1 -C 3) alkoxy. Still more preferably, the phenyl group is optionally substituted with 1 to 3 substituents independently selected from the group consisting of -OH, F, Cl, -CF3, -OCF3, (C1-C4) alkyl and (C1-C3) alkoxy. Still more preferably, the phenyl group is optionally substituted with 1 or 2 independently selected from the group consisting of -OH, F, Cl, -CF3, -OCF3, (C1-C4) alkyl and (C1-C3) alkoxy. Still more preferably, the phenyl group is optionally susiiluted with 1 or 2 substituents independently selected from the group consisting of F, Cl, -CF3, -OCF3, -CH3 and (C1-C3) alkoxy. Still more preferably, the phenyl group is optionally substituted with 1 or 2 atoms of F. When one of R6 and R7 is an optionally substituted phenyl group, is preferably phenyl unsubstituted or 4-F-phenyl. Preferably, the phenyl moiety of the phenylmeryl group of R 6 or R 7 is optionally substituted with 1 to 3 substituents independently selected from the group consisting of -OH, -CN and (C 1 -C 3) alkoxy. More preferably, the phenyl moiety is optionally substituted with 1 or 2 subsides independently selected from the group consisting of -OH and -OCH3. Most preferably, the phenyl moiety of the phenylmeryl group is unsubstituted. Preferably, the heteroaryl-B group of Rd or R7 is optionally substituted with 1 to 3 independently selected susfifuyenles in the group consisting of -OH, -CN, F, Cl, alkyl (C1-C3) substituted with F or Cl, alkoxy (C1-C3) suslifuido with F or Cl, (C1-C4) alkyl and (C1-C3) alkoxy. More preferably, the heleroaryl-B group is optionally susiiluted with 1 to 3 substituents independently selected from the group consisting of -OH, -CN, F, Cl, (C1-C3) alkyl substituted with F, (C1-C3) alkoxy substituted with F, (C 1 -C 4) alkyl and (C 1 -C 3) alkoxy. Still more preferably, the heteroaryl-B group is optionally substituted with 1 to 3 subsides independently selected from the group consisting of -OH, F, Cl, -CF3, -OCF3, (C1-C4) alkyl and (C1-C3) alkoxy ). Still more preferably, it is optionally substituted with 1 or 2 substituents independently selected from the group consisting of F, Cl, -CF3, -OCF3. -CH3 and (C1-C3) alkoxy. Even more preferably, the group heteroaryl-B is optionally substituted with 1 or 2 substituents independently selected from the group consisting of F, Cl and (C 1 -C 3) alkoxy. Still more preferably, heteroaryl-B is optionally substituted with 1 or 2 substituents independently selected from the group consisting of Cl, -OCH3 and -OCH2CH3. In a particularly preferred embodiment, the heleroaryl-B is substituted with a substituent selected from a group previously described, generally or preferably, in particular -OCH3. The heteroaryl-B of R6 or R7 is preferably selected from the group consisting of thienyl, 3- or 4-pyridyl, pyrimidyl and pyrazinyl. More preferably, it will be a 3- or 4-pyridyl group, particularly a 3-pyridyl group, which may be unsubstituted or, preferably, substituted as described above. When the 3-pyridyl group is monosubstituted, it is preferably substituted at the C-6. In a particularly preferred embodiment, the heleroaryl group is 6-meioxypyrid-3-yl. In a preferred embodiment, one of R3 and R4 in the formula (I) is H, (C1-C3) alkyl or (C1-C3) alkoxy; more preferably H or (C 1 -C 3) alkyl, for example, -CH 3; most preferably, H. The other of R3 and R4 in the formula (I) is -C (R8) (R9) (R10). For -C (R8) (R9) (R10) in the compounds of formula (I), two of R8, R9 and R10 are preferably H and the one of R8, R9 and R10 is a heeroaryl-C group. Preferably, heteroaryl-C is selected from the group consisting of indol-3-yl, indazol-3-yl and 7-azaindol-3-yl. More preferably, heteroaryl-C is an indol-3-yl or indazol-3-yl group; most preferably, an indol-3-yl group. The heteroaryl-C group is optionally substituted on the carbon atom (s), preferably placed on the phenyl or pyridyl ring of the heteroaryl-C group, with 1 to 3 substituents; preferably, 1 or 2 sustiluyeníes; most preferably, 1 susliluyenie. The substituents are independently selected and preferably from the group consisting of (C 1 -C 3) alkoxy, F, Cl, CN, -OH, -CO 2 H, feyrazol and (C 1 -C 3) alkoxy substituted with F (eg, -OCF 3); more preferably enriches F and Cl. Still more preferably, the heteroaryl-C group is optionally substituted with 1 or 2 or 3 (preferably 1) F atoms. Most preferably, the heteroaryl is unsubstituted. In a modalidadallernafiva R3 and R4 are joined together to form = CHR11. In this embodiment, when R3 and R4 in the formula (I) are combined together, R11 is the same as the "other" of R8, R9 and R10 as defined above and subsequently, generally and preferably. A preferred embodiment of the invention is shown in the formula wherein X1 is -CH- and X2 is -N- or -C (R12) -, or X1 is N- and X2 is -C (R12) -. Preferably, X1 is -CH- or -N- and X2 is -C (R12) -. Each R12, independently, is preferably selected from the group consisting of H, (C1-C3) alkoxy, F, Cl, CN, -OH, -CO2H, telrazole and (C1-C3) alkoxy susiiuid with F (e.g. -OCF3); more preferably enter H, F and Cl; still more preferably between H and F; with the proviso, however, that no more than three of R12 are different from H. Most preferably, each R12 is H. The other susfiíuyeníes and paramephros in formula (IV) are as defined earlier and subsequently, in a general and preferred way. Formula (V), in which substitutes and other parameters are as defined above, generally and preferably, represented a preferred subgenre of formula (IV).
(V) Preferred compounds of the present invention include: 2- [4- (6-fluoro-1 H -indol-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6, 10b -y-azezabenzo [e] azulen-6-yl] -N-isopropyl-N- (6-meioxy-pyridin-3-yl) -acetamide, 2- [4- (1 H-indol-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-yl-aza-benzo [e] azulen-6-yl] -N -isopropyl-N- (6-meloxy-pyridin-3-yl) -aceamide, 2- [4- (5-fluoro-1 H -indol-3-ylmethyl) -5-oxo-1-phenyM, 5-dihydro -2,3,6, 10b-yl-azabenzo [eJazulen-6-yl] -N-isopropyl-N- (6-methoxy-pyridin-3-yl) -acetamide, 2- [1 -cyclohexyl-4-] (1 H-indol-3-methylmethyl) -5-oxo-4,5-dihydro-2,3,6,10b-terazazabenzo [e] azulen-6-yl] -N-isopropyl-N- (6- meioxy-pyridin-3-yl) -acelamide, 2- [1- (3-hydroxy-phenyl) -4- (1 H -indol-3-ylmethyl) -5-oxo-4,5-dihydro-2,3, 6,10b-tetrazaza-benzo [e] azulen-6-yl] -N-isopropyl-N- (6-mephoxy-pyridin-3-yl) -aceamide, N-benzyl-2- [8,9-difluoro- 4- (5-fluoro-1 H -indol-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-lelraaza-benzo [e] azulen-6-yl ] -N-isopropyl-acefamide, 2- [1- (3-hydroxy-phenyl) -4- (1 H -indol-3-ylmeryl) -5-oxo-4,5-dihydro-2,3,6, 10b-leiraza-benzo [e] azulen -6-yl] -N-isopropyl-N-phenyl-acetylamide, N-benzyl-2- [8,9-difluoro-4- (1 H -indol-3-ylmelyyl) -5-oxo-1-phenyl-4 , 5-dihydro-2,3,6,10b-ioaraza-benzo [e] azulen-6-yl) -N-isopropyl-acetylamide, N-benzyl-2- [8,9-difluoro-4- (6- fluoro-1H-indol-3-ylmeryl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-leiraza-benzo [e] azulen-6-yl] -N-isopropyl- acelamide, N-isopropyl-2- [5-oxo-1-phenyl-4- (1H-pyrrolo [2,3-b] pyridin-3-ylmefyl) -4,5-dihydro-2,3,6,10b-ephase -benzo [e] azulen-6-yl] -N-phenyl-acetylamide, N-benzyl-2- [4- (1 H -indol-3-ylmethyl) -5-oxo-1-phenyl-4,5-dih Dro-2,3,6,10b-ee-arazabenzo [e] azulen-6-yl] -N-isopropyl-acetylamide, N- (6-chloro-pyridin-3-yl) -2- [4- (1 H -indol-3-ylmeryl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-tetraaza-benzo [e] azulen-6-yl] -N-isopropyl -acetamide, N- (6-eloxy-pyridin-3-yl) -2- [4- (1H-indo) -3-ylmethyl) -5-oxo-1-phenyl-4,5-dihi-dro-2, 3,6,10b-tetraaza-benzo [e] azulen-6-yl] -N-isopropyl-acetamide, 2- [4- (1 H -indazol-3-ylmethyl) -5-oxo-1-phenyl- 4,5-dihydro-2,3,6,10b-iora-azabenzo [e] azulen-6-yl] -N-isopropyl-N- (6-methoxy-pyridin-3-yl) -acetamide, 2- [4- (1 H-indazol-3-ylmef ii) -5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-yl-aza-benzo [e] azulen-6-yl] - N-isopropyl-N-phenyl-acetylamide, 2- [4- (1 H -indazol-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6, 10-tertiary- azabenzofe] azulen-6-yl] -N-isopropyl-N-phenyl-acetemide, N-benzyl-2- [8,9-difluoro-4- (1H-indazol-3-ylmethyl) -5-oxo-1- phenyl-4,5-dihi-dro-2,3,6, 10b-fetraaza-benzo [e] azulen-6-yl] -N-isopropyl-acetylamide, N-bendl-2- [4- (1H-indazole -3-ylmeiyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6, 10b-tetraaza-benzo [e] azulen-6-yl] -N-isopropyl-acetamide, 2- [ 4- (1 H-indol-3-ylmeryl) -4-methyl-5-oxo-1-phenyl-4,5-dihydro-2,3,6, 10b-ephzazabenzo [e] azulen-6-yl] - N-isopropyl-N-phenyl-acefamide, 2- [1 - (2-fluoro-phenyl) -4- (1 H-indazo l-3-ylmellyl) -5-oxo-4,5-dihydro-2,3,6, 10b-leiraza-benzo [e] azulen-6-yl) -N-isopropyl-N-phenyl-acetamide, 2- [1- (3-fluoro-phenyl) -4- (1H-indazol-3-ylmethyl) - 5-oxo-4,5-dihydro-2,3,6, 10b-tetraaza-benzo [e] azulen-6-yl] -N-isopropyl-N-phenyl-acetamide, 2- [1 -cyclohexyl-4] - (1 H-indazol-3-ylmeiyl) -5-oxo-4,5-dihydro-2,3,6, 10b-tetraazabenzo [e] azulen-6-yl] -N-isopropyl-N-phenyl-acetamide , 2- [1- (4-fluoro-phenyl) -4- (1 H -indazol-3-ylmethyl) -5-oxo-4,5-dihydro-2,3,6, 10b-tetraaza-benzo [e] azulen-6-yl] -N-isopropyl-N-phenyl-acetamide, N- (4-fluoro-phenyl) -2- [4- (1 H -indazol-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-fefraza -benzo [eJazulen-6-yl-N-isopropyl-acetamide, 2- [4- (1 H -indol-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6 10b-yl-azabenzo [e] azulen-6-yl] -N-isopropyl-N-phenyl-acelamide, 2- [4- (1 H -indol-3-ylmethyl) -5-oxo-1-phenyl- 4,5-dihydro-2,3,6, 10b-terazaza-benzo [e] azulen-6-yl] -N-isopropyl-phenyl-acetamide, 2- [1-cyclohexyl-4- (1H-indole -3-ylmethyl) -5-oxo-4,5-dihydro-2,3,6,10b-teirazaza-benzo [e] azulen-6-yl] -N-isopropyl-N-phenylaliamide, 2- [ 1- (2-fluoro-phenyl) -4- (1 H -indol-3-ylmethyl) -5-oxo-4,5-dihydro-2,3,6, 10b-lelraazabenzo [e] azulen-6-yl ] -N-isopropyl-N-phenyl-acetamide, and 2- [4- (1 H-indol-3-ylmef-ylene) -5-oxo-1-phenyl W, 5-dihydro-2,3,6, 10b -elzaza-benzo [e] azulen-6-yl] -N-isopropyl-N-phenyl-acetamide; or a pharmaceutically acceptable salt thereof. A subset of preferred compound groups includes: 2- [4- (1 H-indol-3-ylmeiyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6, 10b-tef-raaza-benzo [e] azulen-6-yl] -N-isopropyl-N-phenyl-acefamide; N-benzyl-2- [4- (1 H -indol-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-lelraazabenzo [e] azulen-6-yl ] -N-isopropyl-acetylamide; N-benzyl-2- [4- (1 H -indazol-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-letraza-benzo [e] azulen-6 -yl] -N-isopropyl-acetamide; 2- [4- (1 H -indazol-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6, 10b-yl-azabenzo [e] azulen-6-yl] -N-isopropyl-N-phenyl-acetamide; 2- [4- (1 H-lndol-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6,1 Ob-etraza-benzo [e] azulen-6-yl ) -N-isopropyl-N- (6-mephoxy-pyridin-3-yl) -aceamide; or a pharmaceutically acceptable salt thereof. An additional subset of preferred compound strands includes the enanlomers: (-) 2- [4- (1 H -indol-3-ylmeiyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6 , 10b-letterazabenzo [e] azulen-6-yl] -N-isopropyl-N-phenyl-acetamide; (-) N-benzyl-2- [4- (1 H -indol-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-lerazaza-benzo [e ] azulen-6-yl] -N-isopropyl-acelamide; (-) N-benzyl-2- [4- (1 H-indazol-3-ylmefyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6, 10b-ioraza-benzo [e] azulen-6-yl] -N-isopropyl-acetamide; (-) 2- [4- (1H-indazol-3-ylmeiyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-feirazaza nzo [e] azulen-6 -yl] -N-isopropyl-N-phenyl-acetylamide; (-) 2- [4- (1 H -Indol-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-yl-araza-benzo [e] azulen-6-yl] -N-isopropyl-N- (6-meioxy-pyridin-3-yl) -acefamide; or a pharmaceutically acceptable salt thereof. Another aspect of the invention relates to intermediates of formula (D) or formula (F-1) or salts thereof which are useful intermediates in the preparation of the compounds of formula (I).
(F-1) A, B, X, D, R1 and R2 are as defined above for the compounds of formula (I), generally and preferably. R is alkyl (CrC6) or cycloalkyl (C3-C6); preferably (C 1 -C 4) alkyl; more preferably straight-chain (C 1 -C 4) alkyl such as -CH 3 or -CH 2 CH 3. The formula (D-1), in which R is as defined above for the formula (D) and R7 is as defined above for the compounds of the formula (I), generally and preferably, represents a preferred subgenre of the The formula (F-1 a), in which R7 is as defined above for the compounds of formula (I), generally and preferably, represents a preferred subgenus of the formula (F-1).
(F-1a) A further aspect of the present invention relates to a process for the preparation of a compound of formula (C) (C) comprising contacting a compound of formula (A) with a compound of formula (B) in the presence of an acid catalyst, wherein A, B, X and D are. as previously defined for formula (I), generally and preferred. Each R is independently alkyl (Ci-Cß) or cycloalkyl (C3-Cß); preferably alkyl (C1-C4); more preferably straight-chain (C1-C4) alkyl such as -CH3 or -CH2CH3.
(A) (B) The acid catalyst may be an inorganic acid, for example, hydrochloric acid or sulfuric acid; an organic sulfonic acid, for example, benzene sulphonic acid or para-loluenesulfonic acid; or a carboxylic acid, for example, acetic acid. Acetic acid is a preferred catalyst. The process will usually be carried out in the presence of a solvent, preferably an aprolic solvent such as DMF, acetone, mephilyl-cephone, ethyl acephalous, methylene chloride, chloroform, dioxane, THF, toluene or xylenes. More preferably, the solvent is a hydrocarbon solvent such as toluene or xylene (s); especially xylene (s). The process may be carried out at ambient temperature of about 25 ° C or at an elevated temperature, generally in the range of about 50 ° C to reflux, with a preferred preservation of about 150 ° C to 200 ° C. The process can be carried out at atmospheric pressure or under positive pressure, for example up to 10, 20, 30, 40 or 50 atmospheres. In a preferred laboratory-scale embodiment, a solution of the compound of formula (B) in an aprotic solvent is added to a heated solution of the compound of formula (A) and the acid catalyst in an aprotic solvent in a reaction vessel and the whole it is heated to an elevated temperature, as described more particularly in the preparations (3A) and (3B) in the example section. The product (C), under preferred conditions, can be isolated from the reaction mixture simply by filtration.
Also provided is a process for the preparation of a compound of formula (D), a compound of formula (E), a compound of formula (F-1) or a compound of formula (1-1) which comprises converting the compound of formula (C) produced by the procedure described above into the compound of formula (D), the compound of formula (E), the compound of formula (F1) or the compound of formula (11), wherein A, B, X, D, R, R1, R2, R3, and R4 are as defined above, generally and preferably. This invention also relates to salts and solvates, including hydrates, of the compounds of the invention. The compounds of the invention and the intermediates are of basic nature and are capable of forming a wide variety of salts with various inorganic and organic acids. The acids that can be used to prepare the salts by addition of acids Pharmaceutically acceptable compounds are those which form salts by the addition of non-toxic acids, ie salts con taining pharmacologically acceptable anions. Some of the compounds of the invention and the intermediates are of acid nature and are capable of forming salts with various bases. Sodium and potassium salts are preferred. The present invention also relates to prodrugs of the present compounds. The compounds of formula (I) having free carboxy groups, amino or hydroxy can be converted, for example, into esters or amides which act as prodrugs. In another embodiment of the invention, a pharmaceutical composition comprising a compound of formula (I) is provided. In a preferred embodiment, the composition also comprises at least one additional pharmaceutical agent, which is preferably an amino-obesity organism. The additional pharmaceutical agent may also be a useful agent in the treatment of a co-morbidity of the primary indication for the composition. The composition preferably comprises a therapeutically effective amount of a compound of formula (I) or a therapeutically effective amount of a combination of a compound of formula (I) and an additional pharmaceutical agent. The composition preferably also comprises a pharmaceutically acceptable excipient, diluent or carrier. A method for bringing a disease, condition or disorder modulated by an agonist of CCK-A receptors is also provided. animals, which comprises administering to an animal in need of this treatment an therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a solvate or hydrate of said compound or said salt. The compound of formula (I) can be used alone or in combination with at least one additional pharmaceutic agent, preferably an ani-obesity agent or an agent useful in the treatment of a co-morbidity of the disease, condition or transorbium. Diseases, conditions or conditions modulated by an agonist of CCK-A receptors in animals include obesity, overweight and gallstones. The co-morbidities of these diseases, states or transferences with probability will be incidentally improved. Accordingly, a method for treating obesity in a mammal is provided, which comprises administering to a mammal in need of this treatment a therapeutically effective amount of a compound of formula (I), a pharmaceutically acceptable salt thereof or a solvate or hydrate thereof. compound or said salt. The compound of formula (I) can be used alone or in combination with at least one additional pharmaceutical agent, preferably an anti-obesity agent. A method for weight management in an animal is also provided which comprises administering to the animal a weight management amount of a compound of formula (I), a pharmaceutically acceptable salt thereof or a solvate or hydrate of said compound or said Salt. The compound of formula (I) can be used alone or in combination with less an additional pharmaceutical agent, preferably an anti-obesity agent. The present invention also provides a method for reducing the ingestion of food in an animal, which comprises administering to the animal a reducing amount of the food intake of a compound of formula (I), a pharmaceutically acceptable salt thereof or a solvate or hydrate. of said compound or said salt. The compound of formula (I) can be used alone or in combination with at least one additional pharmaceutical agent, preferably an ani-obesity agent. A method for preventing gallbladder stones in an animal is also provided, which comprises administering to the animal a canine to prevent the formation of vesicle stones of a compound of formula (I), a pharmaceutically acceptable salt thereof, or a solvate or hydrate of said compound or said salt. The compound of formula (I) can be used alone or in combination with at least one additional pharmaceutic agent, preferably a useful agent in the treatment or prevention of gall bladder stones. A further aspect of the present invention relates to a pharmaceutical test kit to be used by a consumer in the obesity trayemienio. The test kit comprises (a) a suitable dosage form comprising a compound of formula (I), and (b) instructions that describe a method for using the dosage form to treat or prevent obesity.
The invention also relates to the combination of separate pharmaceutical compositions in the form of a test kit. What is provided in this aspect of the invention is a pharmaceutical test kit comprising: (a) a first pharmaceutical composition comprising a compound of formula (I), (b) a second pharmaceutical composition comprising a second compound useful for the pathology of obesity, the prevention of the formation of vesicular stones or the treatment of a co-morbidity of obesity; and (c) a container containing the first and second compositions. Normally, the test kit will also comprise instructions for the administration of the separate components. The shape of the assay kit is especially advantageous when the separate components are preferably administered in different dosage forms or different dosage ranges. An example of a test kit of the present invention is a so-called blister pack. Blister packs are widely used in the pharmaceutical industry for the packaging of unit dosage forms (tablets, capsules and the like). Blister packs generally consist of a sheet of a relatively rigid material covered with a sheet of a preferably transparent plastic material. During the packaging process, hollow spaces are formed in the plastic sheet. The hollow spaces have the size and shape of the tablets or capsules that are going to be packaged.
Next, the tablets or capsules are placed in the hollow spaces and the relatively rigid sheet of material is sealed against the plastic sheet on the face of the sheet which is opposite to the direction in which the hollow spaces are formed. As a result, the tablets or capsules are sealed in the hollow spaces between the plastic sheet and the sheet. Preferably, the strength of the sheet is such that the tablets or capsules can be removed from the blister pack by applying a manual pressure on the hollow spaces whereby an opening is formed in the sheet at the hollow space location. The tablet or capsule can then be withdrawn through the opening. It may be desirable to provide a memory aid in the test kit, for example, in the form of numbers next to the tablets or capsules, so that the numbers correspond to the days of the regimen in which the tablets or capsules should be ingested. specified. Another example of this memory aid is a calendar printed on the essay kit, for example, as follows "first week, Monday, Tuesday, etc., second week, Monday, Tuesday ..." etc. Other variations of memory aids will be readily apparent. A "daily dose" can be a single pill or capsule or several pills or capsules to be taken on a given day. Also, a daily dose of compounds of the present invention may consist of a tablet or capsules while a daily dose of the second compound may consist of several tablets or capsules and vice versa. The memory aid should reflect this.
DEFINITIONS As used in the present specification, the following terms have the meanings assigned to them, unless otherwise specified. The term "alkyl" means a straight or branched chain hydrocarbon radical of the general formula CnH2n + ?. For example, the term "(C1-C6) alkyl" refers to a monovalent, saturated, straight-chain or branched aliphatic group containing 1 to 6 carbon atoms (eg, methyl, ethyl, n-propyl, i- propyl, n-butyl, i-butyl, s-butyl, t-buyl, n-pentyl, 1-methylbutyl, 2-methylbuyl, 3-methylbutyl, neopenlyl, 3,3-dimethylpropyl, hexyl, 2-methylpentyl and the like) . Analogously, the alkyl part of a group, for example, an alkoxy, acyl, alkylamino, dialkylamino or alkyl group, has the same meaning as before. "Alkyl substituted with halo" refers to an alkyl group substituted with 1 or more halogen atoms (e.g., -CH2CI, -CHF2, -CF3, -C2F5, and the like). Similarly, terms such as "alkyl substituted with F" or "alkyl substituted with Cl" means that the alkyl group is substituted with one or more fluorine or chlorine atoms, respectively. The term "acyl" refers to carbonyl groups substituted with alkyl, cycloalkyl, heteroaryl, aryl, and heeroaryl. For example, acyl includes groups such as alkanoyl (Ci-Cß) (for example, formyl, acetyl, propionyl and the like), cycloalkylcarbonyl (Cß-Cß) (for example, cyclopropylcarbonyl, cyclobuylcarbonyl, cyclopenylcarbonyl and the like), heterocyclylcarbonyl (eg example, pyrrolidinylcarbonyl, pyrrolid-2-one-5-carbonyl, piperidinylcarbonyl, piperazinylcarbonyl, lerahydrofuranylcarbonyl and the like), aroyl (for example, benzoyl) and heteroaroyl (for example, thiophenyl-2-carbonyl, thiophenyl-3-carbonyl, furanyl- 2-carbonyl, furanyl-3-carbonyl, 1H-pyrrolyl-2-carbonyl, 1H-pyrrolyl-3-carbonyl, banzo [b] -phophenyl-2-carbonyl, ele. The term "halo" means F, Cl, Br or I. Preferably, halo will be F, Cl or Br, more preferably F or Cl. "Ar" means aryl The term "aryl" refers to aromatic moieties having a single ring system (eg, phenyl) or condensed rings (eg, naphthalene, anthracene, phenanthrene, etc.) A typical aryl group has a 6- to 10-membered aromatic carbocyclic ring system, unless otherwise stated, the aryl group can be linked to the chemical substance or residue by any one of the carbon atoms in the aromatic ring system. The aryl part (ie, the aromatic resin) of a group (eg, arylalkyl) has the same meaning as above. The term "partially or completely saturated carbocyclic ring" (also referred to as "partially or fully saturated cycloalkyl") refers to non-aromatic rings that are partially or completely hydrogenated and can exist as a single ring, bicyclic ring or ring spiral. Unless otherwise specified, the carbocyclic ring is generally a 3- to 8-membered ring (preferably a 3 to 6 membered ring). For example, carbocyclic / cycloalkyl rings partially or fully saluted include groups such as cyclopropylo, cyclopropenyl, cyclobuyl, cyclobuhyl, cyclopentyl, cyclopentenyl, cyclopentadienyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, norbornyl (bicyclo [2.2.1] hepfyl), norbornenyl, bicyclo [2.2.2] oclyl and the like. Unless otherwise stated, the carbocyclic group may be attached to the chemical substance or residue by any one of the carbon atoms in the cycloalkyl ring system. The cycloalkyl group of a group (for example, cycloalkylalkyl, cycloalkylamino, etc.) has the same meaning as above. The term "partially or fully saturated heimeric cyclic ester" (also referred to as "partially saturated or fully saturated heterocycle" or "partially saturated or fully saturated heterocycle") refers to non-aromatic rings that are partially or completely halogenated, contain at least one hetero ring in the ring and they can exist in the form of a single ring, a bicyclic ring or a spiral ring. Unless otherwise specified, the heterocyclic ring is generally a 4- to 7-membered ring containing 1 to 4 heteroatoms (preferably 1 or 2 heteroaloms) independently selected from sulfur, oxygen or nitrogen. Heterocyclic rings include groups such as epoxy, aziridinyl, pyranyl, teirahydrofuranyl, pyrrolidinyl, N-meilypyrrolidinyl, imidazolidinyl, piperidinyl, piperazinyl, pyrazolidinyl, morpholino, epifhydrolienyl, teirahydrolienyl-1,1-dioxide, and the like. Unless otherwise stated, the heterocyclic group may be linked to the chemical moiety or entity by any one of the atoms of the ring in the heterocyclic ring system. The heterocyclic part of a group (e.g., heterocyclylalkyl) has the same meaning as above. The term "heteroaryl" refers to aromatic moieties containing at least one heteroalome (eg, oxygen, sulfur, nihologen or combinations thereof) with a 5- to 10-membered aromatic ring system (eg, pyridyl, pyrazolyl, indolyl, indazolyl, azaindolyl, ynyl, furanyl, benzofuranyl, oxazolyl, isoxazolyl, imidazolyl, lelrazolyl, pyrimidyl, pyrazinyl, pyridazinyl, thiazolyl, isothiazolyl, quinolinyl, isoquinolinyl, benzothiophenyl, etc.). The heteroaromatic moiety can consist of a single ring or fused ring system. A typical single heeroaryl system is a 5- to 6-membered ring that contains one ring independently selected from oxygen, sulfur, and nylrogen, and a typical fused heteroaryl ring system is a ring system of 9 to 10 members containing one to four heteroatoms independently selected from oxygen, sulfur and nitrogen. Unless otherwise stated, the heteroaryl group can be attached to the entity or chemical moiety by any one of the atoms in the aromatic ring system (e.g., imidazol-1-yl, imidazol-2-yl, imidazole-4) -yl, imidazol-5-yl The heteroaryl part of a group (e.g., heteroarylalkyl) has the same meaning as above The term "solvate" refers to a molecular complex of a compound with one or more solvent molecules For the solvaíos of the compounds of formula (I) (including prodrugs and pharmaceutically acceptable salts thereof), solvent molecules are those commonly used in pharmaceutical engineering, which are known to be harmless to the recipient, for example, water, ethanol and the like . The term "hydrate" refers to a solvate in which the solvent molecule is water. The term "protecting group" or "Pg" refers to a sustiluyenie that is commonly employed to block or protect a particular functionality while other functional groups in the compound react. For example, an "amino protecting group" is a susfiluyenle linked to an amino group that blocks or protects the amino functionality in the compound. Suitable amino protecting groups include acetyl, trifluoroacetyl, t-buxoxycarbonyl (BOC), benzyloxycarbonyl (CBz) and 9-fluorenylmethyleneoxycarbonyl (Fmoc). Analogously, a "hydroxy group" refers to a hydroxy group that blocks or proigates the hydroxy functionality. Suitable proxy-hydroxy groups include acelyl and silyl. A "carboxy protecting group" refers to a susíiuuyenfe that blocks or protects the carboxy functionality as an ester group. Common carboxy protecting groups include -CH2CH2S? 2Ph, cyanoethyl, 2- (dimethylsilyl) ethyIle, 2- (methylsilyl-oxoxymethyl, 2- (p-loluenesulfonyl) ethyIle, 2- (p-niirophenylsulphenyl) ethyl, 2- (diphenylphosphino) ethyl, nihroeroyl and the like For a general description of pro-void groups and their use, see TW Greene, Proieclive Groups in Organic Synfhesis, John Wiley &; Sons, New York, 1991.
The term "prodrugs" means compounds that are precursors of drugs that, following administration, release the drug in vivo through some chemical or physiological process (eg, a prodrug upon being brought to physiological pH or through an action enzyme is converted to the desired drug form). The term "pharmaceutically acceptable" means that the substance or composition is chemically and / or lexicologically compatible with the other ingredients comprising a formulation, and / or with the animal being treated therewith. The term "therapeutically effective" is intended to qualify an amount of an agent to be used in the therapy of a disease, condition or disorder that (i) would prevent or prevent the particular disease, condition or disorder, (ii) attenuate, improve or eliminates one or more of the symptoms of the particular disease, condition or disorder, or (i i i) prevents or delays the onset of one or more symptoms of the particular disease, condition or disorder described in the present specification. The terms "Iraíar" and "Irafamienfo" encompass a preventive approach, that is, prophylactic and paliafive. The term "animal" means human beings as well as other members of warm blood of the animal kingdom that possess a homeosphobic mechanism, including mammals (for example, companion animals, zoo animals and animals that are sources of food) and birds. Some examples of pets are dogs (for example, dogs), felines (for example, Gauls) and horses; Some examples of animals that are sources of food are pigs, cows, sheep, poultry and the like. Preferably, the animal is a mammal. Preferably, the mammal is a human being, a companion animal or an animal that is a source of food. Most preferably, the animal is a human being. The term "compounds of the present invention" and the like (unless otherwise specifically identified) means the compounds of formulas (I) as defined above in a general and preferred manner (including all embodiments), their prodrugs, pharmaceutically salts acceptable compounds and / or prodrugs and hydrates or solvates of the compounds, salts and / or prodrugs, as well as all stereoisomers, atropisomers, tauimers and labeled iso-epimene derivatives of the compounds of formula (I). DETAILED DESCRIPTION OF THE INVENTION In general, the components of the present invention can be prepared by methods described in the present specification or by other procedures within the knowledge of a person having ordinary knowledge in the medicinal chemistry art, which include procedures analogous to those described in the art to produce compounds that are similar or analogous to the present compounds and have substitutes that are similar to those of the present compounds. Certain intermediates and processes for the preparation of the present compounds are provided as Additional features of the present invention are illustrated by means of the following reaction schemes. These procedures can be carried out by sequential or convergent synthetic routes. Other procedures are described in the experimental section. Purification procedures include crystallization and normal phase or reverse phase chromatography. In the following discussion relative to the reaction schemes, certain common abbreviations and acronyms are used including: AcOH (acetic acid), DMF (dimethylformamide), DMSO (dimethylsulfoxide), NH 4 OAC (ammonium acetate), NMP (N-methylpyrrolidone) ) OTS (p-toluenesulfonyloxy), Pg (protecting group) and THF (tetrahydrofuran). Scheme I below illustrates a means for preparing compounds of formula (I) wherein E and G are both -N-. The structures of Scheme I below, A, B, X, D, R, R1, R2, R3 and R4 are as defined above, generally and preferably.
Scheme 1 According to Scheme I, the compound of formula (C) is prepared by coupling a diamine of formula A with the appropriate bisalkoxy-acrylate of formula (B), such as the 3,3-diephoxyacrylic acid eyl ester, for example, by heating the two compounds in a suitable solvent, such as toluene or xylene (s) in the presence of an acid catalyst such as acetic acid. The diamines of formula (A) are commercially available, and can be prepared by methods described in the literature, or can be prepared by methods analogous to those described in the literature for analogous compounds or by methods within the knowledge of a person with ordinary knowledge in the medicinal chemistry art from readily available starting materials. The iminoether of formula (C) is then N-alkylated with an alkylating agent R2L in which L is a leaving group such as Cl, Br, I or OTs (for example, Br as in 2-bromo-N-isopropyl-N) phenyl acetamide) using an inert solvent such as NMP or DMF and a base such as lithium hexamethyldisilazide or sodium hydroxide, at an approximate temperature of -20 ° C to approximately 70 ° C for approximately 2 hours to approximately 48 hours, to supply the N-alkylated imino ether of formula (D). Alternatively, the compound of formula (C) is alkylated, under the conditions described above, with an alkylating agent L-CH2C02Pg (wherein L is a leaving group and Pg is a protecting group) such as 2-haloacetic acid ester ( for example, benzyl 2-bromoacetate) and the ester protecting group is separated to provide the corresponding carboxylic acid, ie, the compound of formula (D) wherein R2 is -CH2COOH. The carboxylic acid compound is then coupled with an amine HNR6R7 using standard methods of amide couplings to provide the compound of formula (D) wherein R2 is -CH2C (O) N (R6) (R7). The compound of formula D in which R2 is -CH2C (O) N (R6) (R7) is then converted to the triazine derivative of formula (F-1) by condensation of the compound of formula (D) with a acyl hydrazide of general formula R1CONHNH2 in an organic solvent, such as glacial acetic acid or toluene, at a temperature in the range of about 0 ° C to that of reflux. Alternately, the compound of formula (C) is converted to the triazine of formula (E) under the conditions described above for the formation of triazine rings. The compound of formula (E) is then N-alkylated to form the compound of formula (F-1) in the manner previously described for the alkylation of the compound of formula (C). The compound of formula (F-1) is alkylated at the C-3 carbon atom using a suitable electrophile such as (R8) (R9) (R10) C-halide or -tosylate (e.g., R3L wherein R3 is -C (R8) (R9) (R10) and L is a labile group such as Cl, Br, I or OTs), a suitable base such as lithium hexamethyldisilazide or sodium hydride, in an inert solvent such as DMF, NMP or THF, at a lemperairy in the range of about -20 ° C to about 70 ° C, to provide the monoalkylated product of formula (1-1) in which R3 is -C (R8) (R9) (R10) and R4 is H. relication of this process with an appropriate base and an alkylating agent (for example, R 4 L wherein R 4 is alkyl or cycloalkyl and L is a leaving group such as Cl, Br, I or OTs) provides the compound of formula (1-1) wherein R3 is -C (R8) (R9) (R10) and R4 is alkyl or cycloalkyl. Compounds in which one of R3 and R4 is halo can be prepared by analogously forming an anion in C-3 with a strong base, such as lithium hexamethyldisilazide or sodium hydride, in an inert solvent, such as DMF, NMP or THF, and a temperature in the range of about .78 ° C at room temperature, and subsequently trapping the anion with a suitable halogenating agent (for example, Br2, C (diethylamino) sulfur (DAST) trifluoride (tetrafluoroborate) of 1- (chlorometii) - 4-fluoro-1, 4-diazoniabicyclo [2.2.2] octane (Selecfluor®, available from Air Products and Chemicals, Inc., 7201, Hamilton Boulevard, Allentown, PA)) to obtain the halogenated compound at C-3. Similarly, trapping the anion at C-3 with a suitable oxygenating reagent, such as 2- (phenylsulfonyl) -3-phenyloxaziridine or triamide oxodiperoximolibedene (pyridine) hexamethylphosphoric (MoOPh) provides the hydroxy compound at C-3, which may be subsequently converted to the C-3 alkoxy compound by an O-alkylation with an alkyl halide and a suitable base under standard conditions. Alternatively, the compound of formula (F-1) is condensed with an aldehyde R11CHO, such as ndol-3-carboxaldehyde, in an organic solvent such as toluene or xylene (s) preferentially in the presence of a base such as piperidine, at an the temperature range is at reflux temperature, to provide the corresponding alpha-bephana-unsalurified medium (ie, wherein R3 and R4 are combined together to form = CHR11) which can be reduced under standard conditions (e.g. Zn-AcOH; H2, Pd-C, to provide the compound of formula (1-1) in which one of R3 and R4 is H and the other of R3 and R4 is -CH2R10 (R11 is the same as R10).
Scheme II below illustrates a means for preparing compounds of formula (I) in which E -N- and G is -C (R5) -. In the formulas of Scheme II below, A, B, X, D, R, R1, R2, R3, R4 and R5 are as defined above, generally and preferably.
Scheme II According to Scheme II, the amidine of formula (G) is prepared by treating an iminoether of formula (C) with NH 3 or a source of ammonia (for example, NH 4 OAC). The amidine is then condensed, under standard conditions, with, for example, a bromoketone of formula R 1 CH (Br) C (O) R 5, to provide the imidazole of formula (H). Alternately, the methyl ether of formula (C) can be condensed with a alpha-amino-celone of formula H2NCH (R5) C (O) R1 to provide the imidazole of formula (H) (see, for example, M. Langlois ef al., J. Heferocycl. Chem: (1982), 19 ( 1), 193-200). The imidazole of formula (H) is then subjected to N1-alkylation (ie, the introduction of the R2 substitute), using similar procedures as described above for the N-alkylation of the compound of formula (C) or (E) in Scheme I, to provide the N-alkylated imidazole of formula (F-2). This compound is then alkylated at C3, using conditions similar to those indicated for conversion of the compound of formula (F-1) into the compound of formula (1-1) in Scheme I, to provide the compound of formula (1-) 2) of Scheme II. Scheme Ill below illustrates a means for preparing compounds of formula (I) wherein E is -C (R5) - and G is -N-. In the structures of Scheme III below, A, B, X, D, R, R 1, R 2, R 3, R 4 and R 5 are as defined above, generally and preferably.
Scheme III The compounds of formula (I-3) can be synthesized by methods analogous to those described in the chemical and patent literature for similar or analogous compounds (see, for example, "Diazepine derivatives," Net. Appl., NL 7803585 (1978), Armin Walser, "Imidazodiazepine derivalives," Ger. Offen., DE 2813549 (1978), and Armin Walser and Rodney lan Fryer, "lmidazo [1, 5-a] [1, 5] benzodiazepines", documen- 4080323 (1978). For example, the compound of formula (D) is condensed with a nitroalkane anion (which can be generated by stirring a microalkane with a strong base, such as lithium hexamethyldisilazide, sodium hydride, potassium t-buioxide or lithium diisopropylamide, in an aprotic organic solvent such as THF, DMSO or DMF, for example) at a temperature in the range of approximately 30 ° C to about 100 ° C to provide the compound of formula (J). This compound is subsequently reduced using a metal calyzer, such as palladium, platinum or nickel, in the presence of hydrogen, to provide the compound of formula (K). The compound of formula (K) is acylated with an acylation agent suitable for the inroduction of R 1 CO-, for example, an acyl halide / base or carboxylic acid / coupling agent (for example EDCI, N, N-carbonyldiimidazole) for provide the compound of formula (L), which is subsequently subjected to standard dewatering ring closure conditions to provide the compound of formula (M). Oxidation of the compound of formula (M) to provide the compound of formula (F-3) is carried out using an oxidant such as manganese dioxide or potassium permanganate. The compound of formula (F-3) can be converted to the compound of formula (1-3) in a manner analogous to that described for the conversion of the compound of formula (F-1) into the compound of formula (1-1 ) in Scheme I. Conventional methods and / or techniques of separation and purification known to one of ordinary skill in the art can be used to isolate the compounds of the present invention, as well as the various intermediates related thereto. These techniques will be fine known by an ordinary expert in the art and can include, for example, all types of chromatography (high pressure liquid chromatography (HPLC)), column chromatography using common adsorbents such as silica gel and thin layer chromatography), recrystallization and differential (i.e. liquid-liquid) extraction techniques. The compounds of the present invention can be isolated or used by themselves or in the form of their pharmaceutically acceptable salts, solvates and / or hydrates. The term "salts" includes inorganic as well as organic salts. These salts can be prepared in situ during the final isolation and purification of a compound, or by reacting the compound separately with a suitable organic or inorganic acid or base and isolating the salt thus formed. The salts of the intermediates need not be pharmaceutically acceptable. The pharmaceutically acceptable acid addition salts representative of the present compounds include hydrochloride, hydrobromide, hydroiodide, nitrate, sulfate, bisulfate, phosphate, acid phosphate, isonicolinalo, acetate, lactal, salicylate, citrate, acid citrate, tartrate, pantothenate salts, bitartraio, ascorbalo, succinalo, maleafo, geníisinalo, fumaraío, gluconaio, glucuronalo, sacrallo, formialo, benzoato, gluíamalo, methanesulfonato, ethanesulfonate, benzenesulfonato, p-toluenosulfonaio, pamoato, palmitato, malonato, estearafo, laurafo, malafo, borato, hexafluorofosfalo, naphthylate, glucoheptonate, lactobionate and lauryl sulphonate and the like. A preferred salt of the compounds is the hydrochloride salt.
The salts formed with bases include cations based on alkaline and alkaline earth metals such as sodium, lithium, potassium, calcium, magnesium and the like, as well as ammonium, quaternary ammonium and cations derived from mono-, bi- or tri-amines included, but without limitation, ammonium, tetramethylammonium and tetraethylammonium and calions derived from mellylamine, ethylamine, dimethylamine, trimethylamine, iaryrylamine and the like. See, for example, Berge, ef al., J. Farm. Sci., 66, 1-19 (1977). The present invention also includes prodrugs of the compounds of formula (I). As used herein, the term "prodrug" means a compound that is transformed in vivo to produce a compound of formula I or a pharmaceutically acceptable salt, hydrate or solvate of the compound. The transformation can be produced by various mechanisms, such as through hydrolysis in the blood. An exposure to the use of prodrugs is provided by T. Higuchi and W. Stella, "Pro-drugs as Novel Delivery Systems," Vol. 14 of the A.C.S. Symposium Series; in Bioreversible Carriers in Drug Design, ed. Edward B. Rocha, American Pharmaceutical Association and Pergamon Press, 1987; in Advanced Drug Delivery Reviews, 1996, 19, 115; and in J. Med. Chem. 1996, 39, 10. For example, if a compound of the present invention contains a carboxylic acid functional group, a prodrug may comprise an ester formed by the substitution of the hydrogen atom of the acid group. with a group such as alkyl (Ci-Cß), alkanoyl (C2-Ci2) -oximethyl, 1- (alkanoyloxy) ethylene having from 4 to 9 carbon atoms, 1-methyl-1- (alkanoyloxy) -ethyl having from 5 to 10 carbon atoms, alkoxycarbonyloxymethyl having from 3 to 6 carbon atoms, 1- (alkoxycarbonyloxy) ) elly having 4 to 7 carbon atoms, 1-methyl-1 - (alkoxycarbonyloxy) elly having from 5 to 8 carbon atoms, N- (alkoxycarbonyl) aminomethyl having from 3 to 9 carbon atoms, 1- (N- (alkoxycarbonyl) amino) ethyl having from 4 to 10 carbon atoms, 3-phthalidyl, 4-crolonolacf onyl, gamma-bulirolaclon-4-yl, di-N, N-alkylamino (C? -C2) - (C2-C3) alkyl (such as β-dimethylaminoelyl), carbamoyl-(C1-C2) alkyl, N, N-dialkyl (C1-C2) carbamoyl-(C1-C2) alkyl and piperidino-, pyrrolidino- or morpholino-alkyl (C2-C3). Analogously, if a compound of the present invention contains an alcohol functional group, a prodrug can be formed by substituting the hydrogen atom of the alcohol group with a group such as alkanoyloxy (Ci-CßHimelil, 1- (alkanoyloxy (C? -C6)) ellyl , 1-meityl-1 - (alkanoyloxy (CrC6)) efyl, alkoxy (Ci-Cey-carbonyloxymethyl, N-alkoxy (CrC6) -carbonylaminomethyl, succinoyl, alkanoyl (Ci-Cß), a-amino-alkanoyl (C1-C4) ), arylacyl and a-aminoacyl, or a-aminoacyl-a-aminoacyl, in each a-aminoacyl group is independently selected from the L-amino acids that are produced naurally, P (O) (OH) 2, P (O ) (O-alkyl (C? -Ce)) 2 or glycosyl (the radical resulting from the separation of a hydroxyl group from the hemiaceal form of a carbohydrate) If a compound of the present invention contains an amino functional group, a prodrug can be formed by replacing a hydrogen atom in the amino group with a group such as R-carbonyl; RO-carbonyl, (R ') (R) -N-carbonyl in which R and R' are each independently afflic (C1-C10), cycloalkyl (C3-C77 or benzyl; or R-carbonyl is a natural a-aminoacyl) or naive a-aminoacyl of naive a-aminoacyl; -C (OH) C (O) OY wherein Y 'is H, alkyl (C? -C6) or benzyl; -C (OY °) Y1 wherein Y ° is (C 1 -C 4) alkyl and Y 1 is alkyl (C.-Cß), carboxy-alkyl (C? -C6), amino-(C1-C4) alkyl or mono-N- or di-N, N-alkylamino (C) ? -C6) -alkyl; or - C (Y2) Y3 wherein Y2 is H or me yl and Y3 is mono-N- or di-N, N-alkylamino (C1-Ca), morpholino, piperidin-1-yl or pyrrolidin-1-yl Many of the compounds of the present invention and certain intermediates contain one or more asymmetric or chiral centers (for example, the carbon atom C-3 carrying R3 and R4 in the formula (I)), and these compounds therefore exist in different stereoisomeric forms (e.g., enantiomers and diastereoisomers). Many of the present compounds also exhibit atropism. All stereoisomeric forms of the intermediates and compounds of the present invention, as well as their mixtures, which include racemic and diastereomeric mixtures, possess useful properties in the treatment of the conditions disclosed herein or are useful intermediates for the preparation of The compounds having these properties form part of the present invention. Generally, one of the enantiomers will be more biologically acidic than the other enantiomer. However, the less active enantiomer can be converted to a racemic mixture by epimerization in the stereocenter C-3 using a strong base, such as sodium methoxide in methanol, for example. The racemic mixture can then be prepared in the form of each enanfomer using standard conditions, such as resolution or chiral chromatography. In addition, the present invention encompasses all geometric isomers and atropisomers. For example, if an intermediate or compound of the present invention contains a double bond or a fused ring, both cis and trans forms, as well as mixtures thereof, are encompassed within the scope of the invention. The diastereomeric mixtures can be separated in the form of individual diastereomers on the basis of their physical and chemical differences by methods well known to those skilled in the art, such as by chromatography and / or fractional crystallization. The enaniomers can be separated by the use of a chiral HPLC column. They can also be separated by converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (eg, a chiral auxiliary such as a chiral alcohol or a Mosher acid chloride), separating the diastereoisomers and converting (eg, hydrolyzing ) the individual diastereomers in the corresponding pure enantiomers. The compounds of the present invention and the intermediates can exist in unsolvated forms as well as solvated with solvents such as water, ethanol, isopropanol and the like, and both solvated and unsolvated forms are included within the scope of the invention. The solvates to be used in the characteristic methods of the invention should be with pharmaceutically acceptable solvents. A number of compounds of the present invention and intermediates thereof exhibit iauomerism and, therefore, may exist in different tautomeric forms under certain conditions. The term "tautomer" or "taulomeric form" refers to structural isomers of different energies that are interconvertible through a low energy barrier. For example, protonic tauimers (also known as pro-thyrotropic tautomers) include inlerconversions to proton migration, such as zeolite-enol and imino-enamine. A specific example of a proton lautomer is an imidazole moiety in which the hydrogen atom can migrate between the ring nitrogen atoms. The valence lautomers include interconversions by rearrangement of some of the electrons in the bonds. All these tautomeric forms (e.g., all keto-enol and imino-enamino forms) are within the scope of the invention. The disclosure of any particular tautomeric form in any of the structural formulas of the present descriptive mem- ory is not intended to be limiting with respect to that form, but is intended to be representative of the complete tautomeric set. The present invention also encompasses isotopically-labeled compounds which are the same as the compounds of formula (I) or their intermediates except that one or more atoms are substituted with an atom having an atomic weight or mass number. differ from the atomic weight or mass number usually found in nature. Examples of isotopes that can be incorporated in the intermediates or compounds of the invention include isotopes of hydrogen, carbon, nihorogen, oxygen, phosphorus, sulfur, fluorine, iodine and chlorine, such as H2, H3, C11, C13 'C14, N13, N15 , O15, O17, O18, P31, P32, S35, F18, I123, I125 and Cl36, respectively. The compounds of the present invention, their prodrugs and pharmaceutically acceptable salts, hydrates and solvates of said compounds and of said prodrugs which contain the aforementioned isotopes and / or other isotope isotopes of other atoms are within the scope of the present invention. Some isotopically-labeled compounds of the present invention (for example, those labeled with H3 and C14) are useful in fabric dissipation tests on compounds and / or substrates. The tritiated isotopes (ie, H3) and carbon-14 (ie, C14) are particularly preferred for their ease of preparation and detection capability. In addition, substitution with heavier isotopes such as deuterium (i.e., H2) may provide certain therapeutic advantages resulting from greater metabolic stability (eg, increased half-life in vivo or reduced dosage requirements) and, therefore, may be preferred. in some circumstances. Positron-emitting isotopes such as O15, N13, C11 and F18 are useful for studies by positron emission tomography (PET) to examine the occupation of substrate receptors. The isotopically-labeled compounds of the present invention can be prepared generally following procedures analogous to those described in the Schemes and / or Examples of the present specification by substituting a reagent that is not isotopically labeled with an isotopically labeled reagent. The compounds of the present invention are useful, for example, for bringing diseases, conditions and conditions modulated by colecisloquinin A receptors (for example, receptacle agonies CCK-A). Such diseases, conditions, and conditions include obesity and gallstones, as well as non-obese overweight conditions and normal weight states in which control or weight management is desired in order to avoid the development of an obese or overweight state. overweight, or just to maintain an optimal healthy weight. Also, the compounds of the present invention are useful for bringing or preventing diseases, conditions and transients that are clinical sequelae or co-morbidities of obesity such as hypertension, dyslipidemia, type 2 diabetes (non-insulin dependent), resistance to insulin, glucose tolerance, hyperinsulinemia, coronary heart disease, angina pectoris, congestive heart failure, stroke, gallstones, cholecystitis, cholelithiasis, obesity, osteoarthritis, obstructive sleep apnea and respiratory problems, gallbladder disease, certain forms of cancer (for example, endomelrial, breast, prostate and colon) and psychological stressors (such as depression, feeding frasins, distorted body image and low self-esteem). Additionally, the present compounds are useful in the irradiation of any combination in which weight loss is desirable or food intake is reduced.
Therefore, the present invention provides methods of treatment or prevention of these diseases, conditions and / or disorders modulated by agonists of CCK-A receptors in a mammal comprising administering to the animal in need of this treatment a compound of formula (I), preferably a therapeutically effective amount thereof. The present compounds will generally be administered in the form of a pharmaceutical composition. Accordingly, the present invention also provides pharmaceutical compositions comprising a therapeutically effective composition of a compound of formula (I) in admixture with a pharmaceutically acceptable excipient, diluent or carrier, as well as methods for using these compositions in the treatment of diseases, conditions and / or disorders that are modulated by agonists of CCK-A receptors in an animal, or clinical sequelae or co-morbidities thereof, which comprises administering this pharmaceutical composition to an animal in need of such treatment. The compounds of formula (I) and the compositions containing them are also useful in the manufacture of a medicament for the therapeutic applications mentioned herein. The compounds of the present invention can be administered to a patient at dosage levels in the range of about 0.1 mg to about 3,000 mg per day. The dosage for a human being will generally vary in the range of approximately 1 mg to approximately 1,000 mg per day; more frequently from about 1 mg to about 400 mg or 500 mg per day; preferably from about 1 mg to about 200 mg or 250 mg per day; more preferably, from about 1 mg to about 75 mg or 100 mg per day; usually about 1 mg to 50 mg or 60 mg per day. The specific dosage and dosage range that can be used depends on a certain number of facors, which include the age and weight of the patient, the mode of administration, the severity of the disease, the condition and / or the condition that is being eroded. and the pharmacological acíividad of the compound that was being administered. The administration of dosing intervals and optimal dosages for a particular patient is denoted by the ordinary knowledge of the technique. The compounds of this invention can be used in combination with other pharmaceutical agents (sometimes referred to herein as a "combination") for the treatment of diseases, states and / or disorders mentioned in the present specification or its co-mbrbilities. Therefore, methods of treatment that include the administration of compounds of the present invention in combination with other pharmaceutical agents are also provided by the present invention. Suitable pharmaceutical agents that can be used in the aspect of combinations of the present invention include agents anli-obesity as antagonists of cannabinoid-1 (CB-1) (as rimonabanf), inhibitors of 11β-hydroxy-sleroid-dehydrogenase-1 (11β-HSD type 1), agonists of peptides YY (PYY) and PYY (such as PYY3-36), MCR-4 agonists, monoamine reuptake inhibitors (such as sibulramine), sympathomimetic agents, ß3 adrenergic receptor agonists, dopamine receptor agonists (such as bromocriptine), analogs of stimulatory hormone receptors of melanocytes (as shown in US Pat. No. 6,716,810), 5HT2c receptor agonists, melanin-concentrating hormone antagonists, leptin (OB protein), leptin receptor agonists. galanin antagonists, lipase inhibitors (such as tephhydrolipstaphine, ie, orlisia), bombesin receptor agonists, Y-neuropeptide receptor anolagonies (NPY) (eg, NPY Y5 receptor antagonists), thyromimine agents, dehydroheprosyrrone, agonists or anonymiasis of glucocorticoid receptors, orexin receptor antagonists, glucagon-like peptide-1 receptor agonists, ciliary neurotropic factors (such as available at Regeneran Pharmaceulicals companies. Inc., Tarryfown, NY and Procter & Gamble Company, Cincinnati, OH), antagonists of aguei related human prolein (AGRP), ghrelin receptor antagonists, antagonists or inverse agonists of histamine 3 receptors, neuromene U receptor agonists, inhibitors of MTP / APoB secretion , T-type calcium channel blockers (such as zonisamide), opioid receptor antagonists (such as described in the PCT patent application publications No. WO 03/101963 and WO 2004/026305) and the like. Preferred NPY receptor antagonists include NPY Y5 receptor antagonists, such as the spiro-compounds described in US Pat. 6,566,367, 6,649,624, 6,638,942, 6,605,720, 6,495,559, 6,462,053, 6,388,077, 6,335,345 and 6,326,375, publications of US patent applications. No. 2002/0151456 and 2003/036652 and publications of PCT patent applications No. WO 03/010175; WO 03/082190 and WO 02/048152. All references cited above are incorporated by reference to the present specification. Preferred anti-obesity agents include orlistai (U.S. Patent Nos. 5,274,143, 5,420,305, 5,540,917, and 5,643,874), sibuiramine (U.S. Patent No. 4,929,629), bromocriptine (U.S. Patent Nos. 3,752,814 and 3,752,888), ephedrine, lepline, pseudoephedrine, zonisamide (U.S. Patent No. 4,172,896) and peptide YY3-36 or an analog or derivative thereof. same (publication of US Patent Application No. 2002/0141985 and publication of PCT application No. WO 03/026591). All references cited above are incorporated as reference to the present descriptive memory. The compounds of this invention can also be used in combination with other pharmaceutical agents (eg, LDL-cholesterol lowering agents, triglyceride lowering agents) for the treatment of the diseases / conditions mentioned herein. descriptive memory. For example, the present compounds may be used in combination with a HMG-CoA-reducyase inhibitor (such as alorvastatin, simvastana, fluvasatin, pravasilain, cerivasatin, rosuvastatin or pifavasilatin), an inhibitor of HMG-CoA-synase, an inhibitor of expression Gene of HMG-CoA-Reduclase, an inhibitor of squalene-synilleia / epoxidase / cyclase, an inhibitor of cholesterol synthesis, an inhibitor of cholesterol absorption (such as ezetimibe), a CETP inhibitor (such as torceyrapib), a modulator of PPAR or another cholesterol-lowering agent such as a fibrate, niacin, an ion exchange resin, an antioxidant, an ACAT inhibitor (such as avasimibe, CS-505 (Sankyo) and eflucimibe) or a bile acid sequestrant. Other pharmaceutical agents useful in the practice of the combination aspect of the invention include inhibitors of bile acid reabsorption.inhibitors of bile acid transporters loyal, inhibitors of ACC, antihypertensive agents (such as Norvasc®), antibiotics, anlidiabéficos (as meforformin, pfenformina or buformina), aclivadores of PPARy, secreíagogues of insulin (as sulfonilureas and glinidas), insulin , aldose-reductase inhibitors (ARI) (eg, zopolresil), sorbitol dehydrogenase inhibitors (SDI)) and aniinflammatory agents such as aspirin or, preferably, an amphiinflammatory agent that inhibits cyclooxygenase-2 (Cox-2) to a measure greater than what is inhibited by cyclooxygenase-1 (Cox-1) such as celecoxib (U.S. Patent No. 5,466,823), valdecoxib (US Pat. No. 5,633,272, parecoxib (Patenie de USA No. 5,932,598), deracoxib (CAS RN 169590-41-4), rofecoxib ((CAS RN 162011-90-7), elorlcoxib (CAS RN 202409-33-4) or lumiracoxib (CAS RN 220991-20-8). The compounds of the present invention can also be administered in combination with naturally occurring substances that act to lower plasma cholesterol levels. Natural substances that are produced in a natural way are commonly called null-arauic products and include, for example, garlic extract, exudates from the Hoodia plant and niacin. A slow release form of niacin is available commercially under the trademark Niaspan. Niacin can also be combined with other leprosy agents such as lobalaine, which is an inhibitor of HMG-CoA reductase. This combination therapy is known as Advicor® (Kos Pharmaceulicals Inc.). The compounds of formula (I) of the present invention can also be used in combination with anti-hyperlensing agents. Preferred antihypertensive agents include calcium channel blockers such as Cardizem®, Adalat®, Calan®, Cardene®, Covera®, Dilacor®, DynaCirc®, Procardia XL®, Sular®, Tiazac®, Vascor®, Verelan®, Isoptin® , Nimolope 'Norvasc®, and Plendil®; angiotensin-converting enzyme (ACE) inhibitors, such as Accupril®, Alface®, Caplopril®, Lotensin®, Mavik®, Monopril®, Prinivil®, Univasc®, Vasoiec® and Zestril®. Diabetes (especially type II diabetes), insulin resistance, impaired glucose tolerance, and the like can be addressed through the administration of an epistemically effective dose of a drug. compound of formula (I), preferably in combination with one or more other agents (eg, insulin) that are useful for treating diabetes. Any glycogen phosphorylase inhibitor can be used as the second agent in combination with a compound of formula (I) of the present invention. The term "glycogen phosphorylase inhibitor" refers to compounds that inhibit the bioconversion of glycogen to glucose-1-phosphate, which is catalyzed by the enzyme glycogen phosphorylase. The activity of inhibiting glycogen phosphorylase is easily determined by standard assays well known in the art (for example, J. Med. Chem. 41 (1998) 2934-2938)). The glycogen phosphorylase inhibitors of interest in the present invention include those described in PCT Patent Application Publication Nos. WO 96/39384 and WO 96/39385. The references cited above are incorporated by reference to the present specification. The aldose reductase inhibitors are also useful in the practice of the combination aspect of the present invention. Compound spheres inhibit the bioconversion of glucose in sorbiol, which is catalyzed by the enzyme aldose reductase. The inhibition of aldose reductase is easily determined by standard assays (for example, J. Malone, Diabetes, 29: 861-864 (1980) "Red Cell Sorbitol, an Indicator of Diabetic Control", incorporated by reference to the present specification. ). A variety of aldose reductase inhibitors are known to those skilled in the art. The aforementioned references are incorporated as reference to the present descriptive memory. Any sorbitol dehydrogenase inhibitor may be used in combination with a compound of formula (I) of the present invention. The term "sorbilol dehydrogenase inhibitor" refers to compounds that inhibit the bioconversion of sorbitol to fructose, which is catalyzed by the enzyme sorbiol dehydrogenase. The inhibitory activity of sorbitol dehydrogenase is easily determined by the use of standard assays well known in the art (eg, Analyt, Biochem (2000) 280: 329-331). Sorbitol dehydrogenase inhibitors of interest include those described in U.S. Pat. Nos. 5,728,704 and 5,866,578. The references cited above are incorporated by reference to the present specification. Any glucosidase inhibitor can be used in the combination aspect of the present invention. These compounds inhibit the enzymatic hydrolysis of complex carbohydrates by glycoside hydrolases such as arnilasa or maliasa in simple bioavailable sugars, for example, glucose. The rapid metabolic action of the glucosidases, particularly following the intake of high levels of carbohydrates, leads to a state of alimentary hyperglycemia which, in adipose or diabetic subjects, leads to an increased secretion of insulin, increased synthesis of fats and a reduction of fat degradation. Following these hyperglycidemia, hypoglemia frequently occurs due to the increased levels of insulin present. Additionally, it is known that the Chyme that remains in the stomach favors the production of gastric juice, which initiates or favors the development of gastritis or duodenal ulcers. Consequently, glucosidase inhibitors are known to be useful in accelerating the passage of carbohydrates through the stomach and inhibiting the absorption of glucose in the intestine. In addition, the conversion of carbohydrates into fatty tissue lipids and the subsequent incorporation of dietary fat in the form of fatty tissue deposits is consequently reduced or delayed, with the concomitant advantage of reducing or preventing the harmful abnormalities resulting therefrom. The inhibition activity of glusidase is easily determined by those skilled in the art according to standard tests (for example, Biochemisíry (1969) 8: 4214), incorporated as reference to the present descriptive specification. A generally preferred glucosidase inhibitor includes an amylase inhibitor. An amylase inhibitor is a glucosidase inhibitor that inhibits the enzymatic degradation of starch or glycogen in maltose. This amylase inhibition activity is easily determined by the use of standard assays (eg, Methods Enzymol. (1955) 1: 149, incorporated by reference herein). The inhibition of this enzymatic degradation is advantageous for reducing the amounts of bioavailable sugars, including glucose and maltose, and the concomitant detrimental states resulting therefrom. Preferred glucosidase inhibitors include acarbose, adiposine, boglibose, miglitol, emiglitate, camiglibose, lendamisate, trestatin, pradimicin-Q and salvosatin. The ascarbose glucosidase inhibitor and various amino-sugar derivatives related thereto are disclosed in U.S. Pat. Nos. 4,062,950 and 4,174,439, respectively. The glucosidase inhibitor adiposine is described in US Pat. No. 4,254,256. The glusidase inhibitor voglibose, 3,4-dideoxy-4 - [[2-hydroxy-1- (hydroxymethyl) efyl] amino] -2-C- (hydroxymethyl) -D-epi-inosylol, and various pseudo-amino sugars susliluted in N related thereto are described in U.S. Pat. No. 4,701,559. The glucosidase inhibitor miglitol, (2R, 3R, 4R, 5S) -1- (2-hydroxyethyl) -2- (hydroxymethyl) -3,4,5-piperidinoiriol, and various related 3,4,5-trihydroxypiperidines with it are described in U.S. Pat. No. 4,639,436. The glycosidase inhibitor hemiglite, p- [2 - [(2R, 3R, 4R, 5S) -3,4,5-ylhydroxy-2- (hydroxymethyl) piperidino] ethoxy] -benzoic acid ester, various derivatives related thereto and their salts by addition of pharmaceutically acceptable acids are described in U.S. Pat. No. 5,192,772. The glucosidase inhibitor MDL-25637, 2,6-dideoxy-7-O-β-D-glucopyranosyl-2,6-imino-D-glycero-1-gluco-heptif ol, various homodisaccharides related thereto and their salts by the addition of pharmaceutically acceptable acids are described in U.S. Pat. No. 4,634,765. The glucosidase inhibitor camíglibosa, 6-deoxy-6 - [(2R, 3R, 4R, 5S) -3,4,5-иhydroxy-2- (hydroxymethyl) piperidino] -D-glucopyranoside-sesfíclohydrate, its related derivatives of deoxy-nojirimycin, various pharmaceutically acceptable salts thereof and synthetic methods for their preparation are described in US Pat. n ° . 157,116 and 5,504,078. The glycosidase inhibitor salbostatin and various pseudosaccharides related thereto are described in U.S. Pat. No. 5,091,524. All references cited above are incorporated by reference to the present specification. The amylase inhibitors of interest in the present specification are described in U.S. Pat. No. 4,451,455, U.S. Pat. No. 4,623,714 (A1-3688 and the various cyclic polypeptides related thereto) and U.S. Pat. No. 4,273,765 (trestatin, which consists of a mixture of trestatin A, threeiaine B and fresilane C, and the various amino sugars containing trehalose related thereto). All references above diadas are incorporated by reference to the present specification. The dosage of the additional pharmaceutical agent generally depends on a number of factors including the health status of the subject being treated, the extent of the desired treatment, the nature and type of the concurrent therapy and, if present, the frequency of the treatment. Irrationality and the nature of the desired effect. In general, the dosage range of the additional pharmaceutical agent is in the range of about 0.001 mg to about 100 mg per kilogram of body weight of the individual per day, preferably from about 0.1 mg to about 10 mg per kilogram of body weight of the body. individual per day. However, some variability of the general dosage range may also be necessary depending on the age and weight of the subject that is being brought, the planned administration route, the particular antiobesity agent that is being administered and the like. The determination of dosing ranges and optimal dosages for a particular patient are also within the capabilities of one of ordinary skill in the art who can take advantage of the present disclosure. According to the methods of treatment of the invention, a compound of the present invention or a combination is administered to a subject in need of this treatment, preferably in the form of a pharmaceutical composition. In the combination aspect of the invention, the compound of the present invention and the other pharmaceutical agent (s) can be administered separately or in a pharmaceutical composition which understand both. It is generally preferred that this administration be oral. When a combination of a compound of the present invention and at least one pharmaceutical agent is administered jointly, this administration can be sequential in time or simultaneous. Generally, simultaneous administration of drug combinations is preferred. For sequential administration, a compound of the present invention and the additional pharmaceutical agent can be administered in any order. It is generally preferred that this administration be oral. It is especially preferred that this administration be oral and simultaneous. When a compound of the present invention and the pharmaceutical agent additional are administered sequentially, the administration of each can be by the same or different methods. Accordingly, a compound of the present invention or a combination can be administered to a patient separately or together in any conventional oral, rectal, transdermal, parenteral (e.g., intravenous, intramuscular or subcutaneous), intracisternal, intravaginal, intraperitoneal, dosage form. topical (for example, powder, ointment, cream, spray or lotion), buccal or nasal (for example, spraying, dropping or inhálate). The compounds of the invention or combinations will generally be administered in admixture with one or more suitable excipients, diluents or pharmaceutical vehicles known in the art and selected taking into account the intended route of administration and standard pharmaceutical practice. The compound of the invention or the combination can be formulated to provide dosage forms of immediate, delayed, modified, sustained, pulsed or conirrolated release, depending on the desired route of administration and the specificity of the release profile, considered with the therapeutic needs. The pharmaceutical compositions comprise a compound of the invention or a combination in an amount generally in the range of about 1% to about 75%, 80%, 85%, 90% or even 95% (by weight) of the composition, usually in the the range of about 1%, 2% or 3% up to about 50%, 60% or 70%, more frequently in the range of about 1%, 2% or 3% to less than 50%, such as about 25%, 30% or 35%. Methods for preparing the various pharmaceutical compositions with a specific amount of active compound are known to those skilled in the art. For example, see Remington: The Practice of Pharmacy, Lippincott Williams and Wiikins, Baltimore MD, 201 ed. 2000. Compositions suitable for parenteral injection generally include sterile and pharmaceutically acceptable solutions, dispersions, suspensions or emulsions, aqueous or non-aqueous, and sterile powders to be reconstituted in the form of sterile injectable solutions or dispersions. Examples of suitable aqueous and non-aqueous vehicles or diluents (including solvents and vehicles) include water, ethanol, polyols (propylene glycol, polyethylene glycol, glycerol and the like), its proper mixtures, triglycerides include vegetable oils such as olive oil and injectable organic esters such as ethyl oleate. A preferred vehicle is the Migliol® brand, caprylic / capric acid ester with glycerin or propylene glycol (eg, Migliol® 812, Migliol® 829 or Migliol® 840) available from Condea Vista Co., Cranford, NJ. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants.
These compositions for parenteral injection may also contain excipients such as preservatives, humectanis, emulsifiers and dispersants. The prevention of contamination by microorganisms of the compositions can be carried out with various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid and the like. It may also be desirable to include isotonic agents, for example, sugars, sodium chloride and the like. Prolonged absorption of injectable pharmaceutical compositions can be carried out by the use of agents capable of delaying absorption, for example, aluminum monostearate and gelatin. Solid dosage forms for oral administration include capsules, tablets, chewing gums, lozenges, pills, powders and preparations in the form of multi-particles (granules). In these solid dosage forms, a compound of the present invention or a combination is mixed with at least one excipient, diluent or inert carrier. Suitable excipients, diluents or vehicles include materials such as sodium citrate or dicalcium phosphate and / or (a) fillers or fillers (e.g., microcrystalline cellulose (available as Avicel® from FMC Corp.), starches, lactose , sucrose, mannitol, silicic acid, xylitol, sorbitol, dextrose, calcium hydrogen phosphate, dexyrin, alpha-cyclodextrin, beia-cyclodextrin, polyethylene glycol, medium chain fatty acids, titanium oxide, magnesium oxide, aluminum oxide and similar), (b) binders (eg, carboxymethylcellulose, methylcellulose, hydroxypropylcellulose, hydroxypropylmethylcellulose, gelfine, gum arabic, ethylcellulose, polyvinyl alcohol, pullulan, pregelatinized starch, agar, urea, alginates, gelatin, polyvinyl pyrrolidone, sucrose, gum arabic and the like); (c) humectanis (e.g., glycerol and the like); (d) disintegrating agents (eg, agar-agar, calcium carbonate, potato starch or tapioca, alginic acid, certain complex silicates, sodium carbonate, sodium lauryl-sulfate, starch-sodium glycolate (available as Exployab® from Edward Mendell Co.), reliculated polyvinylpyrrolidone, croscarmellose sodium lipo A (available as Ac-di-sol®), polyacrylline-polasium (an ion exchange resin) and the like); (e) solution retardants (e.g., paraffin and the like); (f) absorption accelerators (e.g., quaternary ammonium compounds and the like); (g) wetting agents (for example, cetyl alcohol, glycerol monostearate and the like); (h) adsorbents (e.g., kaolin, bentonif and the like); and / or (i) lubricants (eg, talc, calcium acetate, magnesium esarylation, stearic acid, polyoxyl esarylation, cetanol, talc, hydrogenated castor oil, sucrose esters and fatty acids, dimethyl polysiloxane, microcrystalline wax, wax of yellow bees, beeswax, white, solid polyethylene glycols, sodium lauryl sulfate and the like). In the case of capsules and tablets, the dosage forms may also comprise oxidizing agents. Solid compositions of a similar type may also be used as loading mates in hard or soft filled gelatin capsules using excipients, diluents or carriers such as lactose or milk sugar, as well as high molecular weight polyethylene glycols, and the like. Solid dosage forms such as tablets, dragees, capsules and granules with reveslimienlas and cortices can be prepared, as enteric and other well known techniques in the art. They may also contain opacifying agents, and may also be of a composition that release the compound of the present invention and / or the additional pharmaceutical agent in a remanent manner. Examples of included compositions that can be used are polymer substrates and waxes. The drug may also be in a micro-encapsulated form, if appropriate, with one or more of the aforementioned excipients, diluents or vehicles. For tablets, the active agent will normally comprise less than 50% (by weight) of the formulation, for example, less than about 10% as 5% or 2.5% by weight. The predominant part of the formulation comprises excipients, diluents or vehicles as fillers, disintegrants, lubricants and, optionally, flavors. The composition of these excipients is well known in the art. Frequently, the loading / diluting materials will comprise mixtures of two or more of the following components: microcrystalline cellulose, mannitol, lactose (of any / all types), starch and dicalcium phosphate. Mixtures of excipients, diluents or vehicles normally comprise less than about 98% of the formulation and preferably less Approximately 95%, for example, approximately 93.5%. Preferred disintegrants include Ac-di-sol®, Explotab®, starch and sodium lauryl sulfate. When present, a disintegrating agent will readily comprise less than about 10% of the formulation or less than about 5%, for example, about 3%. When present, a lubricant will usually comprise less than about 5% of the formulation or less than about 3%, for example, about 1%. A preferred lubricant is magnesium stearate. The tablets can be made by standard tabletting processes, for example, direct compression or wet, dry or melt granulation, a melt thickening and extrusion process. The tablet cores may be mono- or multi-layer (s) and may be coated with appropriate backings known in the art. Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups and elixirs. In addition to the compound of the present invention or the combination, the liquid dosage form may contain excipients, diluents or inert carriers commonly used in the art, such as water and other solvents, solubilizing agents and emulsifiers such as, for example, ethyl alcohol, alcohol isopropyl, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (for example, cottonseed oil, peanut oil, corn germ oil, olive oil, castor oil, sesame seed oil and the like), Mlglyol® (available from CONDEA Vista Co ., Cranford, NJ.), Glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters and sorbitan, or mixtures of these substances, and the like. Apart from these excipients, diluents or inert carriers, the composition may also include moisturizing, emulsifying and / or suspending agents and sweetening agents, flavors and / or perfumes. Oral liquid forms of the compounds of the invention or combinations include solutions, in which the active compound is dissolvingly compliant. Examples of solvents include all pharmaceutically suitable solvents suitable for oral administration, particularly those in which the compounds of the invention show good solubility, for example, poly-ethylene glycol, polypropylene glycol, edible oils and systems based on glyceryl and glycerides. Systems based on glyceryl and glycerides include, for example, the following products with trademarks (and the corresponding generic products): Captex® 355 EP (tricrapilafo / capillary glyceryl, from Abifec, Columbus, OH), Crodamol® GTC / C (medium-chain triglyceride, from Croda, Cowick Hall, United Kingdom) or Labrafac® CC (medium-chain triglycerides, from Gatlefosse), Capíex® 500P glyceryl-iriaceia, iriacetin, d ela company Abitec), Caprnul® MCM (mono- and di-glycerides of medium chain, from Abitec), Migyol® 812 (caprylic / capric triglyceride, from Condea, Cranford, NJ), Migyol® 829 (triglyceride caprylic / capric / succinic, from the company Condea), Migyol® 840 (dicaprylate / propylene glycol dicaprafo, from the company Condea), Labrafil® M1944CS (oleyl-macrogol-6 glycerides, from Galíefosse), Peceol® (monooleaf from glyceryl, from the company Gaííefosse) and Maisine® 35-1 (glyceryl monooleate, from the company Gatiefosse). Of particular interest are the medium chain iriglyceride oils (approximately Ce to Cio). These solvents frequently constitute the predominant part of the composition, ie more than about 50%, usually more than about 80%, for example about 95%, 97% or 99%. Other excipients can also be included, diluents or vehicles with the solvents mainly as agents for masking flavors, agents for the palate and aromatics, antioxidants, stabilizers, texture and viscosity modifiers, solubilizers and the like. The suspensions, in addition to the compounds of the present invention or combination, may further comprise excipients, diluents or carriers as suspending agents, for example, ethoxylated isostearyl alcohols, polyoxyethylene-sorbium esters and sorbitan, microcrystalline cellulose, aluminum melahydroxide, benioniía, agar-agar and íragacanfo, or mixtures of these substances, and the like.
The compositions for partial or vaginal administration preferably comprise suppositories, which can be prepared by mixing a compound of the present invention or a combination with suitable non-irritating excipients, diluents or vehicles such as cocoa butter, polyethylene glycol or suppository wax which are solid at temperaure ordinary ambienie, but liquid to the corporal temperalura and, therefore, are fused in the rectum or the vaginal cavity thus releasing the active component (s). Dosage forms for topical administration of the compounds of the present invention or combinations include ointments, creams, lotions, powders and sprays. The drugs are mixed with a pharmaceutically acceptable excipient, diluent or carrier, and any preservatives, buffers or propellants that may be necessary or desired. For any of the present compounds that are sparingly soluble in water, for example, less than about 1 μg / ml, liquid compositions in nonaqueous solubilizing solvents such as the above-mentioned medium chain triglyceride oils are a preferred dosage form. Solid amorphous dispersions, including dispersions formed by a spray drying process, are also a preferred dosage form for sparingly soluble compounds of the invention. By a "solid amorphous dispersion" is meant a solid material in which at least a part of the compound sparingly soluble in the amorphous form and dispersed in a water soluble polymer. By "amorphous" it is meant that the sparingly soluble compound is not crystalline. By "crystalline" it is meant that the compound exhibits a wide range order in three dimensions of at least 100 repeating units in each dimension. Therefore, the term "amorphous" is intended to include not only a material that has essentially no order, but also a material that may have some small degree of order, but the order is in less than three dimensions and / or is only over short distances. The amorphous material can be characterized by techniques known in the state of the art such as powder X-ray diffraction crystallography (PXRD), solid-state NMR or thermal techniques such as differential scanning calorimetry (DSC). Preferably, at least one main part (ie, at least about 60% by weight) of the sparingly soluble compound in the solid amorphous dispersion is amorphous. The compound may exist in the solid amorphous dispersion in relatively pure amorphous domains or zones, such as a solid solution of the compound homogeneously distributed by the polymer or any combination of these esides or the states which are intermediate therein. Preferably, the solid amorphous dispersion is substantially homogeneous so that the amorphous compound is dispersed as evenly as possible throughout the polymer. As used herein, "homogeneous homogeneous" means that the fraction of the compound that is present in relatively pure amorphous domains or zones in the solid amorphous dispersion is relatively small, on the order of less than about 20% by weight and preferably less than about 10% by weight of the total amount of drug. Water-soluble polymers suitable for use in solid amorphous dispersions must be inert, in the sense that they do not react chemically with the sparingly soluble compound in an adverse manner, are pharmaceutically acceptable and have at least some solubility in an aqueous solution. Physiologically relevant pHs (For example, 1-8). The polymer may be neutral or ionizable, and must have an aqueous solubility of at least 0.1 mg / ml over at least part of the pH range of 1-8. Water-soluble polymers suitable for use with the present invention may be cellulosic or non-cellulosic. The polymers can be neutral or ionizable in aqueous solution. Of these, ionizable and cellulosic polymers are preferred, and cellulosic polymers are more preferred. Examples of water-soluble polymers include hydroxypropyl methyl cellulose acetate succinate (HPMCAS), hydroxypropylmethyl cellulose (HPMC), hydroxypropylmethyl cellulose (HPMCP) flake, carboxy methyl ethyl cellulose (CMED) , cellulose acetate-phthalate (CAP), cellulose acetyl trimelliac (CAT), polyvinylpyrrolidone (PVP), hydroxypropyl cellulose (HPC), melil-cellulose (MC), block copolymers of ethylene oxide and propylene oxide (PEO / PPO, also known as poloxamers) and their mixtures. Especially preferred polymers include HPMCAS, HPMC, HPMCP, CMEC, CAP, CAT, PVP, poloxamers and mixtures thereof. The most preferred is HPMCAS. See European Patent Application Publication No. 0,901,786 A2, the description of which is given as reference to the present specification. Solid amorphous dispersions can be prepared by any process to form solid amorphous dispersions that result in at least a major part (at least 60%) of the sparingly soluble compound being in the amorphous state. These procedures include mechanical, thermal and solvent procedures. Examples of mechanical methods include trilling and extrusion; melt processes including high temperature melting, solvent modified melting and melt casting procedures; and solvent procedures that include solvent-free precipitation, spray-overs and spray-drying. See, for example, the following U.S. Patents, the pertinent disclosures of which are incorporated by reference herein: Nos. 5,456,923 and 5,939,099, which describe the formation of dispersions by extrusion processes; No. 5,340,591 and 4,673,564, which describe the formation of dispersions by means of trifuration processes; and Nos. 5,707,646 and 4,894,235, which describe the formation of dispersions by means of thickening procedures in molten state. In a procedure Preferred, the solid amorphous dispersion is formed by spray drying, as described in European Patent Application Publication No. 0,901,786 A2. In this process, the compound and the polymer are dissolved in a solvent, such as acetone or methanol, and the solvent is then rapidly removed from the solution by spray drying to form the solid amorphous dispersion. Solid amorphous dispersions can be prepared to contain up to about 99% by weight of the compound, for example, 1% by weight, 5% by weight, 10% by weight, 25% by weight, 50% by weight, 75% by weight , 95% by weight or 98% by weight as desired. The solid dispersion can be used as the dosage form by itself or can serve as a production use (MUP) in the preparation of other dosage forms such as capsules, tablets, solutions or suspensions. An example of an aqueous suspension is an aqueous suspension of a 1: 1 (w / w) compost / HPMCAS-HF spray-dried dispersion containing 2.5 mg / ml of 2% polysorbate-80 compound. The solid dispersions to be used in a tablet or capsule will generally be mixed with other excipients or adjuvants that are normally found in these dosage forms. For example, an example of filler material for capsules contains a spray-dried dispersion (60%) 2: 1 (w / w) of compound / HPMCAS-MF, lactose (rapid flow) (15%), microcrystalline cellulose (for example, Avicel®-102) (15.8%), sodium starch (7%), sodium lauryl sulfa (2%) and magnesium stearate (1%). HPMCAS polymers are available in low, medium and high grades such as Agoat (R) -LF, Agoat (R) -MF and Agoat (R) -HF respectively, from Shin-Eisu Chemical Co., LTD, Tokyo, Japan. Generally, the superior qualities of MF and HF are preferred. The pharmaceutical composition for the application can be packaged in a variety of ways depending on the method used to administer the drug. Generally, an article for dispensing includes a container having the pharmaceutical formulation deposited therein in an appropriate form. Suitable containers are well known to those skilled in the art and include materials such as bottles (of plas- mic and glass), sachets, ampoules, plastic bags, mei- alic cylinders and the like. The container may also include a childproof assembly to prevent indiscreet access to the container. further, the container is deposited on it a label that describes the contents of the container. The effique may also include appropriate warnings. USE IN VETERINARY MEDICINE The following paragraphs describe examples of formulations, dosages, etc. useful for non-human animals. The administration of the compounds of the present invention and the combinations of the compounds of the present invention with anti-obesity agents can be carried out orally or non-orally.
A quality of a compound of the present invention or a combination of a compound of the present invention with an anti-obesity agent is administered so that an effective dose is received. Generally, the daily dose that is administered orally to an animal is about 0.01 and about 1,000 mg / kg of body weight, for example, about 0.01 to about 300 mg / kg or about 0.01. and about 100 mg / kg or between about 0.01 and about 50 mg / kg or about 0.01 and about 25 mg / kg, or about 0.01 and about 100 mg / kg or about 0.01 and about 5 mg / kg. mg / kg of body weight. Conveniently, a compound of the present invention (or combination) can be carried in the drinking water so that a therapeutic dosage of the compound is ingested with the daily supply of water. The compound may be measured directly in the drinking water, preferably in the form of a water-soluble liquid concentrate (such as an aqueous solution of a water-soluble salt). Conveniently, a compound of the present invention (or combination) can also be added directly to foods, such as in the form of a food supplement for animals, also called premix or concentrate. A preliminary or concentrated mixture of the compound in an excipient, diluyenle or vehicle is more commonly used for the inclusion of the agenie in food. The excipients, Suitable diluents or vehicles are liquids or solids, as desired, such as water, various foods such as alfalfa meal, soy meal, cottonseed oil meal, flaxseed oil quote, corn cob meal and corn, molasses, urea, bone meal and mineral mixtures, as is commonly used in poultry feeds. A particularly effective excipient, diluent or vehicle is the animal's own feed; that is, a small part of these foods. The vehicle facilitates a uniform distribution of the compound in the finished foods with which the premix is combined. Preferably, the compound is mixed thoroughly in the premix and subsequently with the food. In this regard, the compound can be dispersed or dissolved in a suitable oily vehicle such as soybean oil, corn oil, cottonseed oil and the like, or in a volatile organic solvent and then combined with the carrier. It will be appreciated that the proportions of compound in the concentrate are susceptible to wide variation since the amount of the compound in the finished foods can be adjusted by combining the appropriate ratio of premix with the food to obtain a desired level of compound. Concentrates of high potency can be combined by the food manufacturer with a protein carrier such as soybean meal and you will hear meals, as described above, to produce concentrated supplements, which are suitable for feeding the animals directly. In these cases, animals are allowed to consume the diet habitual. Alternately, the concentrated supplements can be added directly to the food to produce a finished, unbalanced food containing an ineradically effective level of a compound of the present invention. The mixtures are thoroughly combined by standard procedures, as in a double-wrap mixer, to ensure homogeneity. If the supplement is used as a top coating for the food, this also helps to ensure a uniform distribution of the compound through the top of the coated food. Drinking water and foodstuffs effective to increase the lean meat deposit and to improve the ratio of lean meat to fat are generally prepared by mixing a compound of the present invention with a sufficient amount of animal feed., to provide from about 10"3 to about 500 ppm of the compound in food or water .. Preferred medicated feeds for pigs, cattle, sheep and goats generally contain from about 1 to about 400 grams of a compound of the present invention. (or combination) per tonne of food, and minimum calorie for animal animals is usually from about 50 to about 300 grams per tonne of food.
Preferred foods for domestic poultry and pet commonly contain from about 1 to about 400 grams and preferably from about 10 to about 400 grams of a compound of the present invention (or combination) per ton of food. The compounds of the present invention can be administered to an animal parenterally. Standard injectable granules or solutions or suspensions are useful for parenteral administration. In general, parenteral administration includes the injection of a sufficient amount of a compound of the present invention (or combination) to provide the animal with about 0.01 to about 20 mg / kg / day of body weight of the drug. The preferred dosage for domestic poultry, pigs, cattle, sheep, goats and pets is in the range of about 0.05 to about 10 mg / kg / day of body weight of drug. The compounds of the present invention (or combination) can be prepared in the form of a granulate and be administered in the form of an implant, usually under the skin of the head or ear of the animal in which an increase in the deposit of lean meat and an improvement in the ratio of lean meat to fat. Granules containing an effective amount of a compound of the present invention, pharmaceutical composition or combination can be prepared by mixing a compound of the present invention or combination with a diluyenle such as carbowax, carnauba wax and the like, and a lubricant, such as magnesium or calcium stearate, which can be added to improve the granulation process. Naturally, it is recognized that more than one pellet can be administered to an animal to achieve the desired dose level that will provide the increase in lean meat deposit and the improvement in the ratio of lean meat to fat desired. In addition, implants can be prepared periodically during the treatment period of the animal in order to maintain the appropriate drug level in the body of the animal. The compounds of the present invention can also be administered orally to non-human animals, for example companion animals such as dogs, cats and horses, and animal sources of food, in the same dosage forms as those used for humans, by example, tablets, capsules, solutions, suspensions, pastes, powders, etc. Paste formulations can be prepared by dispersing the drug in a pharmaceutically acceptable oil such as peanut oil, sesame oil, corn oil or the like. The present invention has various advantageous veterinary features. For the pet owner or veterinarian who wishes to increase lean meat and / or remove unwanted fat from domestic animals, the present invention provides a means by which to perform I am For breeders of poultry, calves and pigs, the use of the present invention method produces lean animals that result in higher selling prices in the meat industry. The embodiments of invention are illustrated by means of the following examples. However, it should be understood that the embodiments of the invention are not limited to the specific details of these examples, since variations of the same will be known, or will be evident taking into account the present description, for an ordinary expert in the art. . EXAMPLES Unless otherwise specified, starting materials are generally available from commercial sources such as Aldrich Chemicals Co. (Milwaukee, WI), Lancaster Synthesis, Inc. (Windham, NH), Acros Organics (Fairlawn, NJ), Maybridge Chemical Company, Lid. (Cornwail, England), Tyger Scientific (Princeton, NJ), and AstraZeneca Pharmaceuticals (London, England) or can be prepared using methods known to those skilled in the art from readily available materials. GENERAL EXPERIMENTAL PROCEDURES NMR spectra were recorded on a Varian Unity® device 400 (available from Varian Inc., Palo Alio, CA) at ambient temperature of 400 and 500 MHz H1, respectively. Chemical shifts are expressed in parts per million in relation to the residual solvent as an infernal reference. The shapes of the peaks are indicated as follows: s, singlefe; d, doublet; l, complete; q, quartet; m, multiple; br s, single width; v br s, singlele very wide, br m, mulliplete wide. In some cases only the representative peaks of H1 NMR are provided. The mass spectra were recorded by means of a direct flow analysis using positive and negative chemical ionization at atmospheric pressure (APcl) exploration modes. A ZMD mass spectrometer model APcI / MS Waíers equipped with a Gilson 215 liquid handling system was used to carry out the experiments. The analysis by mass spectrometry was also done by means of a gradient method of RP-HPLC for chromatographic separation. The identification of molecular weights was recorded by means of positive and negative electrospray ionization (ESI) scanning modes. A mass spectrometer model ZMD or LCZ Water / Micromass ESI / MS (Waters Corp., Milford, MA) equipped with a Gilson 215 liquid handling system (Gilson, Inc., Middelton, Wl) was used and a device was used. HP 1100 DAD (Hewlett Packard) to carry out the experiments. When the intensity of the ions containing chlorine or bromine is described, the expected intensity ratio (approximately 3: 1 for ions containing CI35 / CI37 and 1: 1 for ions containing Br79 / Br81) was observed and only the lower mass ion. The optical rotations were determined in a Perkin Elmer® 241 polarimeter (available from PerkinElmer Inc., Wellesley, MA) using the sodium D line (? = 589 nm) at the indicated temperature and are expressed as follows [a] Dtemp, concentration (c = g / 100 ml) and solvent . Column chromatography was performed with Baker® silica gel (40 μm, JT Baker, Phillipsburg, NJ) or Silica Gel 50 silica gel (EM Sciences®, Gibbstown, NJ) on glass columns, on Biotage® columns (Bioíage , Inc., Charloíiesville, USA) or using a Combinflash separation system subjected to low hydrogen pressure. Radial chromatography was performed using a Chromatotron® device (Harrison Research). The selected purifications were performed using liquid chromatography of Shimadzu preparation. Chiral separations were performed using a Chirlapak AD, (S, S) -Whelk-O 1 or Chiralcel OD column. References to "enantiomer 1" or "enantiomer 2" refer merely to the order in which the compounds are eluted from the column. In the following discussion, certain common abbreviations and acronyms were used that include: AcOH (acrylic acid), DMAP (4-dimethylaminopyridine), DMF (dimethylformamide), EIO2 (diethyl ether), EtOAc (ethyl acetate), EtOH ( ethanol), Et 3 N (triethylamine), KHMDS (potassium hexamethyldisilazane), MeOH (methanol), NaBH (OAc) 3 (sodium triaceotroxyborohydride), NaHMDS (sodium hexamethyldisilazane), TFA (trifluoroacetic acid) and THF (teirahydrofuran). Example KA) Preparation of 2-f4- (6-Fluoro-1 H -indol-3-ylmeryl) -5-oxo-1-phenyll-4,5-dihydro-2,3,6,10-b-eneaza-benzofe] azulen-6-yl- N-O-propyl-N- (6-meioxy-pyr-d-n-3-yl) -aceiamide Epaque A: 2-f4- (6-Fluoro-1 H-indol-3-ylmeiylene-5-oxo-1-phenyl-4,5-dihydro-2.3.6.10b-eiraza-benzofelazulen-6-yl) -N- isopropyl-N- (6-meioxy-pyridin-3-yl) -acetamide To a solution of N-isopropyl-N- (6-methoxy-pyridin-3-yl) -2- (5-oxo-1- phenyl-4,5-dihydro-2,3,6,10b-epzaza-benzo [e] azulen-6-yl) -aceamide (Preparation 11) (150 mg, 0.311 mmol) and 6-fluoro-1H- indole-3-carbaldehyde (61 mg, 0.373 mmol) in toluene (20 ml) was added piperidine (100 μl). The reaction vessel was equipped with a Dean-Slark separator containing íoluene and 4A molecular sieves and heated to reflux for 24 hours. The solution was cooled to room temperature and diluted with water. The aqueous solution was washed with ElOAc (3x) and the combined organic extracts were dried (MgSO 4), filtered and concentrated. The residue was purified by medium pressure chromatography eluting with a gradient of dissolve (10% ElOAc in hexanes to 100% EOAc) to provide 158 mg of 2- [4- (6-fluoro-1 H-indole-3- illmethylene) -5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-ioaraza- benzo [ejazulen-6-yl] -N-isopropyl-N- (6-meioxy-pyridin-3-yl) -acetamide. MS 628.3 (M + 1), 626.2 (M-1). Step B: (2-F4- (6-Fluoro-1H-indol-3-oxo-5-ilmeíin-1-fen¡l-4.5-dihydro-2.3.6.10b-ieiraaza-benzofe1azulen-6-¡pN- Isopropyl- (6-meioxy-pyridin-3-p-acetylamide) To a solution of 2- [4- (6-fluoro-1 H -indol-3-ylmelylene) -5-oxo- 1-phenyl-4,5-dihydro-2,3,6,10b-tetraaza-benzo [e] azulen-6-yl] -N-isopropyl-N- (6-methoxy-pyridin-3-yl) - acetamide (158 mg, 0.252 mmol) in MeOH (20 ml) was added ammonium formate (463 mg, 7.34 mmol) and 10% palladium on carbon (112 mg) .The reaction was refluxed for 24 hours. the suspension was filtered hot, rinsed with 10% MeOH in CH2CI2 (3x) and CH2CI2) (3x). the combined organic filfrados is conceníraron and the residue was purified by medium pressure chromatography eluting with a solvent gradient (2 % MeOH in CH 2 Cl 2 to 8% MeOH in CH 2 Cl 2) to yield 110 mg of 2- [4- (6-fluoro-1 H -indol-3-ylmethyl) -5-oxo-1-phenyl-4,5-d HYDR-2,3,6, 10b-yl-raaza-benzo [e] azulen-6-yl] -N-isopropyl-N- (6-meioxy-pyridin-3-yl) -acetami da H1 NMR (CD3OD) d 8.08 (d, 1H), 7.39-7.62 (m, 9H) 7.15 (m, 2H), 6.93 (m, 3H), 6.68 (m, 1H), 4.80 (m, 1H), 4.55 (m, 11-1), 4.20 (m, 1H), 3.95 (s, 3H), 3.88 (m, 1H), 3.77 (m, 1H), 3.67 (m, 1H), 1.01 (m, 6H); MS 630 (M + 1), 628.5 (M-1) Example KB) Preparation of (-) 2-14- (1 H-lndol-3-ylmeryl) -5-oxo-1-phenyl-4,5-dhydro-2,3,6,10-b-earaza-benzo [e1azulen-6-il ) -N-isopropyl- (6-meioxy-pyridyl-3-yl) -acetamide Step A: 2-r 4 - (1 H-lndol-3-ylmelylene) -5-oxo-1-phenyl-4,5-dihydro-2,3,6,10-tetraaza-benzo [e] azulen-6-yl] -N-isopropyl-N - (6-methoxy-pyridin-3-yl) -aceiamide Following the procedure described for Example 1 (A), Eíapa A, N-isopropyl-N- (6-meioxy-pyridin-3-yl) -2 was condensed - (5-Oxo-1-phenyl-4,5-dihydro-2,3,6,10b-tetraaza-benzo [e] azulen-6-yl) -acetamide (Preparation 11) (293 mg, 0.608 mmol ) with 1H-indole-3-carbaldehyde (106 mg, 0.729 mmol). Purification by medium pressure chromatography eluting with a solvent gradient (2% MeOH in CH 2 Cl 2 at 10% MeOH in CH 2 Cl 2) yielded 286 mg of 2- [4- (1 H -indole-3-ylmephylene) - 5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-teiraza-benzo [e] azulen-6-yl] -N-isopropyl-N- (6-mephoxy-pyridin-3-) il) -aceiamide. H1 NMR (CD3OD) d 8.25 (s) and 7.90 (s, total 1H), 8.16 (s, 1H), 7.58-7.67 (m, 4H), 7.37-7 , 53 (m, 6H), 7.07-7.17 (m, 3H), 6.94 (m, 2H), 4.90 (m, 1H), 4.55 (m, 1H), 4, 25 (m, 1H), 3.96 (m, 4?), 1.10 (m, 6H); MS 610.8 (M + 1), 608.5 (M-1).
Eíapa B: 2-r4- (1H-lndol-3-ilmetin-5-oxo-1-phenyl-4.5-dihydro-2.3.6.10b-teíraaza-benzo [e1azulen-6-ill-N-isoprop¡lN- ( 6-meioxy-pyridin-3-p-acetamide Following the procedure described for Example 1 (A), Step B, 2- [4- (1H-indole-3-ylmethyl) -5- was reduced. oxo-1-phenyl-4,5-dihydro-2,3,6,10b-tetraaza-benzo [e] azulen-6-yl] -N-isopropyl-N- (6-methoxy-pyridin-3-yl) acetamida (276 mg, 0.453 mmol) and purified by medium pressure chromatography eluting with a solvent gradient (CH2Cl2 at 5% MeOH in CH2CI2) to yield 86 mg of 2- [4- (1 H -indole) 3-ylmeryl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6, 10b-leiraza-benzo [e] azulen-6-yl] -N-isopropyl-N- (6- meioxy-pyridin-3-yl) -acetamide in the form of a racemic mixture H1 NMR (CD3OD) d 8.08 (d, 1H), 7.61 (m, 3H), 7.39-7.53 (m , 6H), 7.25 (d, 1H), 7.12 (m, 2H), 7.00 (m, 11-1), 6.90 (m, 3H), 4.78 (m, 1H) , 4.55 (m, 1H), 4.15 (m, 1H), 3.95 (s, 3H), 3.89 (m, 1H), 3.67-3.80 (m, 2H), 1.01 (m, 6H); MS 612.2 (M + 1), 610.5 (M-1) Step C: (-) 2-r4- (1H- indole-3-ilmet¡n-5-oxo-1-phenyl-4.5-dihydro- 2,3.6.10b-teíraazabenzofe1azulen-6-ilj-N-¡sopropil-N- (6-meioxi-p¡ridin-3- L) -aceiamide. enantiomer 1 The racemic product of Step B, 2- [4- (1H-indol-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6, 10b-fe-phaza- benzo [e] azulen-6-yl] -N-isopropyl-N- (6-meioxy-pyridin-3-yl) -acetamide (217 mg, 0.355 mmol), was separated into its enanfomers by high pressure chromatography using a Chiralpak AD column (5 cm x 50 cm), eluting with heptane in EtOH (75:12), using a flow rate of 85 ml / minute to provide the enantiomer 1 which was a Renewal time of 7.04 minutes. The enanfiomer was dissolved in EOAc and the organic layer was washed with water (1x) and brine (1x). The organic extract was dried (MgSO 4), filtered and concentrated to give 85 mg of 2- [4- (1 H -indol-3-ylmefyl) -5-oxo-1-phenyl-4,5-dihydroxy). 2,3,6, 10b-ioaraza-benzo [e] azulen-6-yl] -N-isopropyl-N- (6-meloxy-pyridin-3-yl) -acelamide (enantiomer 1). H1 NMR (CD3OD) d 8.08 (d, 1H), 7.61 (m, 3H), 7.39-7.53 (m, 6H), 7.25 (d, 1H), 7.14 ( m, 2H), 7.00 (t, 1H), 6.90 (m, 3H), 4.78 (m, 1H), 4.54 (m, 1H), 4.11 (m, 1H), 3.95 (s, 3H), 3.89 (t, 1H), 3.68-3.82 (m, 2H), 1.01 (m, 6H); MS 612.9 (M + 1), 610.5 (M-1); [a] D21 -79.6 (c 0.255, eilanol). Example KC) Preparation of 2- | '4- (5-Fluoro-1 H -indole-3-methyl-p-5-oxo-1-phenyl-4,5-dihydro-2.3.6.10-amino-acetylase- benzo [ejazulen-6-yl] -N-isopropyl-N- (6-meioxy-pyridin-3-iD-aceiamide Epaque A: 2- | "4- (5-Fluoro-1 H-indol-3-ylmeiylene) -5-oxo-1-phenyl-4,5-dihydro-2.3.6.10b-feiraza-benzore1azulen-6-in-N -isopropyl-N- (6-meioxy-pyridin-3-yl) -acetamide Following the procedure described for Example 1 (A), Step A, N-isopropyl-N- (6-methoxy-pyridine-3) was condensed. -il) -2- (5-oxo-1-phenyl-4,5- dihydro-2,3,6,10b-terazazabenzo [e] azulen-6-yl) -acelamide (Preparation 11) (120 mg, 0.207 mmol) with 5-fluoro-1H-indole-3-carbaldehyde (48 mg 0.248 mmol). Purification by medium pressure chromatography eluting with a gradient of solvents (1% MeOH in CH 2 Cl 2 to 7% MeOH in CH 2 Cl 2) yielded 85 mg of 2- [4- (5-fluoro-1 H -indol-3-ylmethylene) -5-oxo-1-phenyl-4,5-dihydro-2, 3,6,10b-teirazazabenzo [e] azulen-6-yl] -N-isopropyl-N- (6-meioxy-pyridin-3-) L) -acephamide. MS 628.8 (M + 1), 626.4 (M-1). Step B: 2-f4- (5-Fluoro-1 H -indole-3-ylmetin-5-oxo-1-phenyl-4,5-dihydro-2.3.6.10b-tetraaza-benzophe] azulen-6-ip-N -sopropyl-N- (6-meioxy-pyridin-3-yl) -aceylamide Following the procedure described for Example 1 (A), EIApa B, was reduced and purified 2- [4- (5 -fluoro-1H-indol-3-llmet-ene) -5-oxo-1-phenyl-4,5-dihydro-2, 3,6,10b-ephzazabenzo [e] azulen-6-yl] - N-isopropyl-N- (6-methoxy-pyridin-3-yl) -acetamide (85 mg, 0.135 mmol) by medium pressure chromatography eluting with a gradient of solvents (2% MeOH in CH 2 Cl 2 at 9% MeOH in CH 2 Cl 2) to yield 42 mg of 2- [4- (5-fluoro-1 H -indol-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6, 10b-leiraza-benzo [e] azulen-6-yl] -N-isopropyl-N- (6-mephoxy-pyridin-3-yl) -acetamide H1 NMR (CD3OD) d 8.08 (d, 1H ), 7.38-7.65 (m, 8H), 7.19 (m, 3H), 7.14 (f, 1H), 6.90 (d, 2H), 6.76 (f, 1H) , 4.80 (m, 1H), 4.50 (m, 1H), 4.12 (m, 1H), 3.94 (s, 3H), 3.87 (i, 1H), 3.74 ( m, 1H), 3.64 (m, 1H), 1.01 (m, 6H), MS 630.4 (M + 1), 628.5 (M-1) Example KD) Preparation of 2- [1-Cyclohexyl-4- (1 H -indol-3-ylmethyl) -5-oxo-4,5-dihydro-2,3,6,10-b-ezaza-benzofe] azulen-6-ylj-N-isopropyl -N- (6-meioxi-p¡r¡din-3 -l) -aceíamída Epaque A: 2-ri-Cyclohexyl-4- (1H-indol-3-ylmethylene) -5-oxo-4,5-dihydro-2.3.6.10b-feiraza-benzofe-1azulen-6-yl] -N-isopropyl -N- (6-meioxy-pyridin-3-iD-aceylamide Following the procedure described for Example 1 (A), Efapa A, 2- (1-cyclohexyl-5-oxo-4,5-dihydro-2,3,6,10b-tetraaza-benzo [e] azulen-6-yl) -N-isopropyl-N- was condensed (6-methoxypyridin-3-yl) -acelamide (Preparation 8 (A)) (90 mg, 0.184 mmol) with 1-indol-3-carbaldehyde (32 mg, 0.221 mmol). Purification by medium pressure chromatography eluting with a gradient of solvents (10% EtOAc in hexanes to 100% EtOAc) yielded 72.5 mg of 2- [1-cyclohexyl-4- (1H-indol-3-ylmethylene) -5-oxo-4,5-dihydro-2, 3,6,10b-l-ezaza-benzo [e] azulen-6-yl] -N-isopropyl-N- (6-meioxypyridin-3-yl) -acetamide. H1 NMR (CDCl3) d 8.63 (s, 1H), 8.16 (m, 1H), 7.88-7.96 (m, 1H), 7.45-7.76 (m, 2H), 7.12-7.44 (m, 6H), 6.83 (m, 1H), 5.10 (m, 1H), 4.11-4.35 (m, 1H), 3.94 (f, 2H), 3.84 (dd, 1H), 2.94 (m, 1H), 2.25 (m, 1H), 1.99 (m, 2H), 1.70 (m, 6H), 1, 35 (m, 3H), 1.15 (m, 6H); MS 616.4 (M + 1), 614.3 (M-1).
Stage B: 2-M-Cyclohexyl-4- (1 H -indol-3-ylmeryl-5-oxo-4,5-d -hydro-2.3.6.10b-eeryaraza-benzofe1azulen-6-l1-N -soprop) lN- (6-meioxy-pyridin-3-yl) -acetamide Following the procedure described for Example 1 (A), Step B, was reduced 2- [1-cyclohexyl-4- (1H-indol-3-ylmethylene) -5-oxo-4,5-dihydro-2,3,6,10b-tefraza-benzo [e] azulen-6-yl] -N-isopropyl-N- (6-meloxy-pyridin-3-yl) -acetamide (72.5 mg, 0.118 mmol). Purification by medium pressure chromatography eluting with a gradient of solvents (10% of EOAc in hexanes to 100% of EOAc) provided 19.7 mg of 2- [1-cyclohexyl-4- (1H-indo) -3-yl. mef il) -5-oxo-4,5-dihydro-2,3,6, 10b-ioraza-benzo [e] azulen-6-yl] -N-isopropyl-N- (6-meioxy-pyridin-3-) il) -aceiamide. H1 NMR (CDCl3) d 8.21 (1, 1 H), 7.02-8.09 (m, 10H), 6.84 (cid, 1 H), 5.02 (m, 1H), 4, 18 (1, 1H), 3.56-3.96 (m, 5H), 2.79 (m, 1H), 2.16 (m, 1 H), 1.92 (m, 1H), 1, 69 (m, 7H), 1.53 (m, 1H), 1.31 (m, 2H), 1.05 (m, 6H); MS 618.7 (M + 1). Example KE) Preparation of 2- | "1- (3-Hydroxy-phenyl-4- (1 H -indol-3-ylmethyl) -5-oxo-4,5-dihydro-2.3.6.10-teraza-benzore-1-azulene 6-il1-N-isopropyl-N- (6-meioxy-pyridin-3-ip-acetylamide) Step A: 2-M- (3-Benzyloxy-phenyl-4- (1 H -indol-3-ylmelylene) -5-oxo-4,5-dihydro-2,3,6.10b-tetraaza-benzo [e | azulen-6-] il] -N-isopropyl-N- (6-methoxy-pyridin-3-yl) -acetamide Following the procedure described for Example 1 (A), Eíapa A was condensed 2- [1- (3- benzyloxyphenyl) -5-oxo-4,5-dihydro-2, 3,6,1 Ob-bozaza-benzo [e] azulen-6-yl] -N-isopropyl-N- (6-methoxy-pyridine) -3-yl) -acelamide (Preparation 6) (129 mg, 0.219 mmol) with 1 H-indole-3-carbaldehyde (70 mg, 0.48 mmol) A purification by medium pressure chromatography eluting with a gradient of solvents (10% EtOAc in hexanes at 100% EtOAc) yielded 60 mg of 2- [1- (3-benzyloxy-phenyl) -4- (1H-indol-3-ylmethylene) -5-oxo-4,5- Hydro-2,3,6,10b-tetraaza-benzo [e] azulen-6-yl] -N-isopropyl-N- (6-methoxy-pyridin-3-yl) -aceiamide H1 NMR (CD3OD) d 8.13 (m, 1H), 7.64 (m, 2H), 7.23-7.48 (m, 13H), 7.13 (m, 3H), 6.93 (m, 2H), 4.94 (m, 2H), 4.85 (m, 1H), 4.63 (m, 1H), 4.20-4.40 (m, 1H), 3.96 (m, 1H) ), 3.85 (d, 1H), 0.90-1.05 (m, 6H), MS 716.5 (M + 1), 714.6 (M-1). Step B: 2-f 1- (3-Hydroxy-phenan-4- (1 H -indol-3-ylmeiyl) -5-oxo-4,5-dihydro-2,3,6.10b-tetraaza-benzofejazulen-6 -ilj-N-isopropyl-N- (6-meioxy-pyridin-3-yl) -acetamide Following the procedure described for Example 1 (A), Step B, 2- [1- (3-benzyloxyphen L) -4- (1H-indol-3-ylmethylene) -5-oxo-4,5-d-hydroxy-2,3,6,10b-yl-aza-benzo [e] azulen-6-yl] -N-isopropyl -N- (6-meioxy-pyridin-3-yl) -acetamide (60 mg, 0.084 mmol) and was deprotected for 48 hours.Purification by medium pressure chromatography eluting with a gradient of solvents (10% EtOAc in hexanes at 100% EtOAc) yielded 39.5 mg of 2- [1- (3-hydroxy-phenyl) -4- (1 H -indole-3-methyl) -5- oxo-4,5-dihydro-2,3,6, 10b-tetraaza-benzo [e] azulen-6-yl] -N-isopropyl-N- (6-methoxy-pyridin-3-ii) -acetamide. H NMR (CD3OD) d 8.15 (d, 2H), 7.63 (s, 1H), 6.91-7.44 (m, 14H), 4.80-5.05 (m, 2H), 4.10-4.60 (m, 3H), 3.95 (s, 3H), 3.40-3.90 (m, IH), 1.13 (m, 6H); MS 627 (M + 1), 625 (M-1). Example KF) Preparation of N-Benzyl-2- [8.9-difluoro-4- (5-fluoro-1 H -indol-3-ylmeryl) -5-oxo-1-phenyl-4,5-dihydro-2.3. 6.10b-tef raaza-benzofe1azulen-6-yl] -N-isopropyl-acefamide Step A: N-Benzyl-2-r8.9-difluoro-4- (5-fluoro-1H-indol-3-lmethylene) -5-oxo-1-phenyl-4,5-dihydro-2.3. 6.10b-1α-benzorejazulen-6-yl-1-N-isopropyl-acefamide Following the procedure described for Example 1 (A), Step A, N-benzyl-2- (8,9-difluoro-5-oxo-) was condensed. 1-phenyl-4,5-dihydro-2,3,6,10b-epzaza-benzo [e] azulen-6-yl) -N-isopropyl-acelamide (Preparation 9) (100 mg, 0.198 mmol) with 5- fluoro-1H-indole-3-carbaldehyde (38.8 mg, 0.238 mmol). Purification by means of medium pressure chromatography eluting with 50% of EOAc in CH2Cl2 yielded 91.5 mg of N-benzyl-2- [8,9-difluoro-4- (5-fluoro- 1 H-indol-3-ylmelenyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6, 10b-yl-raaza-benzo [e] a-zulen-6-yl] -N- isopropyl-acetamide. H1 NMR (CDCl3) d 7.92 (d, 1H), 7.10-7.67 (m, 13H), 6.84 (m, 1H), 6.64 (m, 1H), 4.37- 4.97 (m, 5H), 4.20 (m, 1H), 1.20 (m, 6H); MS 647.4 (M + 1), 645.3 (M-1). Step B: N-Benzyl-2- (8.9-difluoro-4- (5-fluoro-1 H-indol-3-methylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2.3.6.10-teraza -benzo [e1azulen-6-yl1-N-isopropyl-acetamide Following the procedure described for Example 1 (A), Step B, N-benzyl-2- [8,9-difluoro-4- (5-fluoro -1H-indo) -3-ylmethylene) -5-oxo-1-phenyl-4,5-dihydro-2,3,6, 10b-lelraza-benzo [e] azulen-6-yl] -N-isopropyl -acetylamide (91.5 mg, 0.140 mmol) to yield 77.2 mg of N-benzyl-2- [8,9-difluoro-4- (5-fluoro-1 H -indol-3-ylmethyl) -5- oxo-1-phenyl-4,5-dihydro-2,3,6, 10b-telraza-benzo [e] azulen-6-ii] -N-isopropyl-acelamide. H1 NMR (CDCl3) d 8.04 (d, 1H), 7.16-7.60 (m, 13H), 6.85 (m, 1H), 6.66 (m, 1H), 4.34- 4.90 (m, 5H), 3.83 (m, 2H), 3.64 (m, 1H), 1.23 (m, 3H), 1.13 (m, 3H); MS 649.5 (M + 1), 647.5 (M-1). Example KG) Preparation of 2- (1- (3-Hydroxy-phenyl) -4- (1 H -indol-3-ylmefiD-5-oxo-4,5-dihydro-2.3.6.10b-fefraaza-benzorejazulen-6-ilj -N-isopropyl-N-phenyl-acetylamide Step A: 2-M- (3-Hydroxy-phenyl-4- (1 H -indol-3-ietiene) -5-oxo-4,5-dihydro-2.3.6.1 Ob-teiraza-benzorelazole- 6-ylj-N-isopropyl-N-phenyl-acetamide Following the procedure described for Example 1 (A), EIAPA A, 2- [1- (3-hydroxyphenyl) -5-oxo-4,5-dihydro-2, 3,6,1 Ob-leiraza-benzo [e] azulen-6-yl) -N-isopropyl was condensed -N-phenyl-acelamide (Preparation 12) (244 mg, 0.522 mmol) for 48 hours with 1 H-indole-3-carbaldehyde (105 mg, 0.723 mmol). Purification by medium pressure chromatography eluting with a gradient of solvents (ElOAc to 5% MeOH in EOAc to 10% MeOh in ElOAc) yielded 177.9 mg of 2- [1- (3-hydroxy-phenii) -4 - (1H-indol-3-ylmethylene) -5-oxo-4,5-dihydro-2,3,6,10b-tetraaza-benzo [e] azulen-6-yl] -N- -propyl-N-phenyl -acetamide. H1 NMR (CD3OD) d 8.23 (d, 1H), 7.90 (s, 1H), 6.90-7.63 (m, 16H), 4.95 (m, 1H), 3.68- 4.50 (m, 3H), 1.11 (m, 6H); MS 595.3 (M + 1), 593.3 (M-1). Step B: 2-H- (3-Hydroxy-phenin-4- (1 H -indol-3-ylmethyl) -5-oxo-4,5-dihydro-2,3,6.10b-fefraaza-benzofe1azulen-6-il -N-isopropyl-N-phenyl-acefamide Following the procedure described for Example 1 (A), Step B, 2- [1- (3-hydroxyphenyl) -4- (1H-indol-3-ylmethylene) -5-oxo-4,5-dihydro-2,3,6, 10b-lefraazabenzo [e] azulen was reduced -6-yl] -N-isopropyl-N-phenyl-acetamide (177.9 mg, 0.299 mmol) lasted 20 hours and was purified by reverse phase medium pressure chromatography (C-18) eluting with a gradient of solvents ( 15% 0.1% formic acid / CH3CN in 0.1% formic acid / water at 100% formic acid 0.1% / CH3CN) to produce 77 mg of 2- [1 - (3-hydroxy phenyl) -4- (1 H-ndol-3-ylmethyl) -5-oxo-4,5-dihydro-2,3,6, 10b- tetraaza-benzo [e] azulen-6-yl] -N-isopropyl-N-feryl-acetamide. H1 NMR (CD3OD) d 7.53 (m, 7H), 7.32 (m, 3H), 7.22 (t, 3H), 6.90-7.03 (m, 5H), 4.82 ( m, 2H), 4.45 (d, 1H), 4.12 (d, 1H), 3.93 (d, 1H), 3.76 (d, 1H), 1. 04 (m, 6H); MS 597.4 (M + 1), 595.5 (M-1). Example KH) Preparation of N-Benzyl-2-f8.9-difluoro-4- (1 H -indol-3-ylmethyl-D-5-oxo-1-phenyl-4,5-dihydro-2.3.6.10b-tetraaza- benzo [e1azulen-6-il1-N-isopropyl-acetamide] Step A: N-Benzyl-2-f8.9-d-fluoro-4- (1 H -indol-3-ylmethylene) -5-oxo-1-phenyl-4,5-dihydro-2.3.6.10b-tetraaza -benzore1azulen-6-pN-isopropyl-acetamide Following the procedure described for Example 1 (A), Step A, N-benzyl-2- (8,9-difluoro-5-oxo-1-) was condensed. phenyl-4,5-dihydro-2,3,6,10b-epzaza-benzo [e] azulen-6-yl) -N-isopropylamide (Preparation 9) (200 mg, 0.399 mmol) for 24 hours with 1H -indol-3-carbaldehyde (70 mg, 0.479 mmol). Purification by medium pressure chromatography eluting with a gradient of solvents (CH2Cl2 at 20% acetone in CH2Cl2) afforded 190 mg of N-benzyl-2- [8,9-difluoro-4- (1H-indol-3-ylmethylene )-5- oxo-1-phenyl-4,5-dihydro-2,3,6,10b-lelaraza-benzo [e] azulen-6-yl] -N-isopropyl-acetamide. MS 629.3 (M + 1), 627.3 (M-1). Step B: N-Benzyl-2-f8.9-difluoro-4- (1 H -indole-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2.3.6.10b-tetraaza-benzo [ e] azulen-6-yl-1-N-isopropyl-acetamide Following the procedure described for Example 1 (A), Efapa B, N-benzyl-2- [8,9-difluoro-4 (1H-indo) -3-ylmethylene) -5-oxo-1-phenyl-4,5-dihydro-2,3 was reduced 6, 10b-pherazaazabenzo [e] azulen-6-yl] -N-isopropyl-acetamide (185 mg, 0.294 mmol) for 24 hours. The reaction was filtered through Celite® and the filtrate was concentrated in vacuo. The residue was dissolved in ElOAc and washed with aqueous NaHCO3 (1x). The organic solution was dried (Na2SO4), filtered and concentrated in vacuo. Purification by preparative chromatography eluting with 5% MeOH in CH2Cl2 yielded 35 mg of N-benzyl-2- [8,9-difiuoro-4- (1H-indol-3-ylmethyl) -5-oxo-1-phenyl- 4,5-dihydro-2,3,6,10b-tetraaza-benzo [e] azulen-6-yl] -N-isopropyl-acetamide. H1 NMR (CD2Cl2) d 8.30 (d, 1H), 6.98-7.64 (m, 15H), 6.68 (q, 1 H), 4.06-4.90 (m, 5H) , 3.73-3.89 (m, 3H), 1, 08-1.28 (m, 6H); MS 631.3 (M + 1), 629.3 (M-1) Example Kl) Preparation of N-Benzyl-2-r8.9-difluoro-4- (6-fluoro-1H-indol-3-ylmethyl- 5-oxo-1-phenyl-4,5-dihydro-2.3.6.10b-tetraaza-benzorejazulen-6-ylj-N-isopropyl-acetamide H, C Hfi;? 9 Stage A: N-Benzyl-2-F8.9-difluoro-4- (6-fluoro-1 H -indole-3-methylmethyl-5-oxo-1-pheny1,5-dihydro-2.3.6.10b -tetraaza-benzore azulen-6-pN-isopropyl-acetylamide Following the procedure described for Example 1 (A), Eíapa A, condensed N-benzyl-2- (8,9-difluoro-5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-eeiraza-benzo [e] azulen -6-yl) -N-isopropyl-acelamide (Preparation 9) (50 mg, 0.099 mmol) with 6-fluorindole-3-carboxaldehyde (19.4 mg, 0.119 mmol). Purification by medium pressure chromatography eluting with 50% of EOAc in CH2CI2 yielded 23.5 mg of N-benzyl-2- [8,9-d, fluoro-4- (6-fluoro-1 H-indole-3- illmethylene) -5-oxo-1-phenyl-4,5-dihydro-2, 3,6,10b-leiraza-benzo [e] azulen-6-yl] -N-isopropyl-aceiamide. H1 NMR (CDCl3) d 7.94 (d, 1H), 7.61 (m, 3H), 7.27-7.49 (m, 9H), 6.95 (m, 1H), 6.86 ( m, 1H), 6.63 (m, 1H), 4.35-5.0 (m, 5H), 4.20 (m, 1H), 1.23 (m, 6H); MS 647.3 (M + 1), 645.3 (M-1). Step B: N-Benzyl-2-r8.9-difluoro-4- (6-fluoro-1 H -indol-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2.3.6.10-aminoaza -benzofejazulen-6-yl] -N-isopropyl-acetylamide Following the procedure described for Example 1 (A), Step B, N-benzyl-2- [8,9-difluoro-4- (6- fluoro-1H-indol-3-ylmethylene) -5-oxo-1-phenyl-4,5-dihydro-2, 3,6,10b-tetraaza-benzo [e] azulen-6-yl] -N-ysopropyl- acetamide (23 mg, 0.035 mmol) at 80 ° C for 24 hours. The reaction was filtered through Celite® and concentrated in vacuo. The residue was dissolved in EtOAc and the organic solution was washed with aqueous NaHC 3, dried (MgSO 4), filtered and concentrated to provide 22.7 mg of N-benzyl-2- [8,9-difiuoro] -4- (6-fluoro- 1 H-indol-3-ylmef-yl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6, 10b-tetraaza-benzo [e] azulen-6-yl] -N -isopropyl-acetylamide. H1 NMR (CDCl3) d 8.40 (s) and 8.28 (s, total 1H), 7.10-7.60 (m, 12H), 6.94 (d, 1H), 6.77 (f , 1 H), 6.64 (m, IH), 4.31-4.95 (m, 5H), 3.61-3.93 (m, 3H), 1.07-1.30 (m, 6H); MS 649.5 (M + 1), 647.5 (M-1). Example KJ) Preparation of N-lsopropyl-2-r5-oxo-1-phenyl-4- (1H-pyrrolo [2.3-bjp¡r¡d¡n-3-ilmet¡p-4.5-dihidro-2.3.6.10b -leiraza-benzofe1azulen-6-nN-phenyl-acetylamide Stage A :. N-isopropyl-2-r5-oxo-1-phenyl-4- (1H-pyrrolof2.3-bjpyridin-3-methylmethylene) -4.5-dihydro-2.3.6.10b-ioaraza-benzo | e1azulen-6 -yl] -N-phenyl-acetylamide Following the procedure described for Example 1 (A), Step A, N-isopropyl-2- (5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-y-aza-benzo [e] azulen-6-yl) -N-phenyl- was condensed aceiamide (Preparation 7 (A)) (500 mg, 1.10 mmol) with 1H-pyrrolo [2,3-b] pyridino-3-carbaldehyde (160 mg, 1.16 mmol) in pyridine (5 mL). Purification by medium-pressure chromatography eluted with a gradient of solvents (5% acetone in CH 2 Cl 2 at 50% acetone in CH 2 Cl 2 at 10% MeOH in CH 2 Cl 2) yielded 300 mg of N-isopropyl-2- [5-oxo- 1-phenyl-4- (1H-pyrrolo [2,3-b] pyridin-3-ylmeiylene) -4,5-dihydro-2,3,6, 10b-eeryza-benzo [e] azulen-6- il] -N-phenyl-acetylamide. H1 NMR (CDCI3) d 8.31 (d, 1H), 8.05 (d) and 7.96 (d, iohal 1H), 7.71 (m, 1H), 7.59 (m, 2H), 7.33-7, 51 (m, 9H), 7.17 (m, 2H), 7.01 (m, 1H), 6.80 (1, 1H), 5.09 (m, 1H), 4.50 (d) and 4.30 (d, iohal 1H), 4.15 (m, 1H), 3.92 (d, 1H), 1.14 (m, 6H); MS 580.6 (M + 1). Step B: N-lsopropyl-2-r5-oxo-1-phenyl-4- (1 H-Pyrrolo [2,3-blPyridin-3-ylmeiyl) -4.5-d -hydro-2.3.6.10b-eyiraza-benzo [ejazulen-6-lN-phenyl-acetylamide Following the procedure described for Example 1 (A), Step B, N-iopropyl-2- [5-oxo-1-phenyl-4- (1H) was reduced. -pyrrolo [2,3-b] pyridin-3-ylmelylene) -4,5-dihydro-2,3,6,10b-tefraazabenzo [e] azulen-6-yl] -N-phenyl-acetamide (300 mg, 0.518 mmol) at 60 ° C for 6.5 hours. The reaction was filtered through Celite® and concentrated in vacuo. The residue was dissolved in EtOAc and the organic solution was washed with water (1x). The organic solution was dried (Na2SO4), filtered and concentrated in vacuo. Purification using reverse phase high pressure chromatography (C-18) eluting with a solvent gradient for 6 minutes (35% 0.1% NH 4 OH / CH 3 CN in 0.1% NH 4 OH / 100% H 2 O NH 4 OH 0.1% / CH3CN) gave 100 mg of N-isopropyl-2-. [5-oxo-l-phenyl-4- (1H-pyrrolo [2,3-b] pyridin-3-ylmethyl) -4,5-dihydro-2,3,6, 10b-telraza- benzo [e] azulene-6-yl-N-phenyl-acetylamide. H1 NMR (CDCl3) d 8.22 (d, 1 H), 8.00 (d, 1H), 7.55 (m, 3H), 7.32-7.48 (m, 9H), 7.18 (m, 1H), 7.05 (m, 2H), 6.85 (d, 1H), 4.97 (m, 11-0, 4.27 (d, 1H), 4.06 (m, 1H ), 3.87 (d, 2H), 3.70 (f, 1H), 1.09 (m, 6H), MS 582.6 (M + 1), 580.5 (M-1).
Example KK) Preparation of -N-Benzyl-2-y4-. { 1 H-3-methyl-3-oxo-1-phenyl-4,5-dihydro-2.3.6.10-ioaraza-benzofe1azulen-6-in-N-iopropyl-acetamide. enantiomer 1 Step A: N-Benzyl-2-f4- (1 H -indol-3-ylmephylene) -5-oxo-1-phenyl-4,5-dhydro-2,3,6,1-tetraaza-benzofe-1azulen-6-yl] -N- isopropyl-acetamide Following the procedure described for Example 1 (A), Efapa A, N-benzyl-N-isopropyl-2- (5-oxo-1-phenyl-4,5-dihydro-2,3,6 was condensed , 10b-tetraaza-benzo [e] azulen-6-yl) -acetamide (Preparation 10) (13.9 g, 29.8 mmol) with 1H-indole-3-carbaldehyde (5.2 g, 35.8 mmol ) for 48 hours. The volatiles were concentrated in vacuo to provide 18.5 g of N-benzyl-2- [4- (1 H -indol-3-ylmethylene) -5-oxo-1-phenyl-4,5-dihydro-2,3 , 6,10b-tetraaza-benzo [e] azulen-6-yl] -N-lsopropyl-acetamide. H1 NMR (CDCl3) d 8.01 (d, 1H), 7.67-7.50 (m, 4H), 7.43-7.11 (m, 12H), 6.99 (m, 1H), 6.99 (m, 1H), 6.80 (m, 1H), 4.96 (m, 1H), 4.71-4.49 (m, 4H), 4.23 (m, 1H), 1 20 (m, 6H); MS 593.3 (M + 1). Step B: N-Benzyl-2-f4-1H-indol-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2.3.6.10b-tetraaza-benzofe1azulen-6-ip-N-isopropyl- acetamide Following the procedure described for Example 1 (A), Step B, N-benzyl-2- [4- (1H-indol-3-ylmethylene) -5-oxo-1-phenyl-4 was reduced., 5-dihydro-2,3,6, 10b-tetraazabenzo [e] azulen-6-yl] -N-isopropyl-acetamide (18.5 g, 31.2 mmol) at 60 ° C for 24 hours. The reaction was filtered through Celite® and concentrated in vacuo. The residue was dissolved in CH2Cl2 and the organic solution was washed with water. The organic layer was dried (Na2SO4), filtered and concentrated in vacuo. The residue was dissolved in toluene and stirred at 60 ° C for 24 h. The solids were filtered to provide 12.4 g of N-benzyl-2- [4- (1 H -indol-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6. , 10b-tetraaza-benzo [e] azulen-6-yl] -N-isopropyl-acetamide in the form of a racemic mixture. H1 NMR (CDCl3) d 8.02 (d, 1H), 7.57 (d, 3H), 7.46-7.23 (m, 8H), 7.17-7.01 (m, 6H), 6.83 (dd, 1H), 4.96-4.40 (m, 5H), 4.17-3.89 (m, 2H), 3.80 (m) and 3.72 (m, total 1H ), 1.20 (dd, 3H), 1, 13 (dd, 3H); MS 595.3 (M + 1). Step C: -N-Benzyl-2-f4- (1 H -indole-3-methyl-5-oxo-1-phenyl-4,5-dihydro-2.3.6.1 Ob-tetraaza-benzofelazulen-6-illN-isopropyl- acetamide enaniomer 1 The racemate of the Epaque B, N-benzyl-2- [4- (1 H -indol-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3, 6,10b-telraaza-benzo [e] azulen-6-yl] -N-isopropyl-acelamide (13.56 g, 22.8 mmol) was separated in its enantiomers by means of high pressure chromatography using a column ( S, S-Whelk-O 1 (5 cm x 25 cm), eluting with heptane in EtOH (60:40), using a flow rate of 140 ml / minute to provide enaniomer 1 having a retention time of 17 minutes. The active enantiomer (enantiomer 1) was dissolved in CH2CI and the organic solution was washed with aqueous NaHC 3 (1x) and brine (1x). The organic solution was dried (Na2SO4), filtered and concentrated to provide 6.4 g of (-) - N-behcyl-2- [4- (1H-indol-3-methyl-5-oxo) -1-phenyl-4,5-dihydro-2,3,6,10b-tetraaza-benzo [eJazulen-6-yl] -N-isopropyl-acetamide (enantiomer 1). A mixture of the solid (6.4 g, 10.77 mmol) in EtOH (250 mL) was heated at 45 ° C for 96 hours, cooled slowly to room temperature for 4 hours, filtered and washed with a minimum amount of EtOH to provide 5 g of (-) - N-benzyl-2- [4- (1 H -indol-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6, 10b-terazazabenzo [e] azulen-6-yl] -N-isopropyl-acetylamide (enantiomer 1) (crystalline enantiomer 1), mp 244-245 ° C. H1 NMR (DMSO-de) d 10.79-10.77 (d, 1H), 7.76-7.74 (d, 1H), 7.56-7.24 (m, 7H), 7.20 -7.12 (m, 5H), 7.02-6.91 (m, 2H), 6.91-6.83 (m, 2H), 5.20 (m, 0.5H), 4.95 to 4.83 (m, 1H), 460-4.51 (m, 2H), 4.47 (m, 0.5H), 4.25-4.21 (m, 1H), 3.84-3, 78 (m, 2H), 3.64-3.58 (m, 2H), 1, 12-1.10 (d, 1.7H), 1.00-0.99 (d, 1.7H), 0.94-0.92 (d, 2.6H); [a] D20 -49.3 (c 1, ethanol). Example 2 (A) Preparation of N- (6-chloro-pyridin-3ip-2-f4- (1 H -indol-3-ylmei-p-5-oxo-1-phenyl-4,5-dihydro- 2.3.6.10b-teiraza-benzofe] azulen-6-nN-isopropyl-acetamide jn-sa To a solution of [4- (1H-indol-3-methylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-telraaza-benzo [e] azulen-6 acid ii) -acetic (Preparation 15) (30 mg, 0.06 mmol) and (6-chloro-pyridin-3-yl) -isopropyl-amine (Preparation 2 (A)) (10.1 g, 0.06 mmol ) in benzene (2 ml) was added PCI3 (2.0 M in CH2Cl2, 0.1 ml, 0.198 mmol) and the reaction was refluxed for 24 hours. The reaction was cooled to room temperature and diluted with CH2Cl2. The organic solution was washed sequentially with aqueous NaHC 3 (1x), water (1x), 1N HCl (1x) and water (1x). The organic solution was dried (MgSO4) and the volatile elements were concentrated in vacuo. The residue was purified by medium pressure chromatography eluting with 10% ElOH in CH 2 Cl 2 to provide 5 mg of N- (6-chloro-pyridin-3-yl) -2- [4- (1 H-indole-3 -ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-y-aza-benzo [e] azulen-6-yl] -N-isopropyl-acetamide. H1 NMR (CDCl3) d 8.26 (s, 1 H), 8.17 (s, 1H), 7.76-7.29 (m, 11H), 7.06 (m, 3H), 6.83 (d, 1H), 5.04-4.87 (m, 1H), 4.45-4.15 (m, 1H), 3.94-3.82 (m, 3H), 3.72 (m , 1H), 0.06 (s, 6H); MS 616.6 (M + 1), 614.5 (M-1). Example 2 B) Preparation of N- (6-Eioxy-pyridin-3-in-2-y4- (1 H -indole-3-ylmein-5-oxo-1-phenyl-4,5-dihydro-2.3.6.10b -irazaaza-benzo | 'e1azulen-6-in-N-sopropyl-acetylamide Following the procedure described for Example 2 (A), [4- (1 H -indol-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6, 10b was reacted -tetraaza-benzo [e] azulen-6-yl] -acetic (Preparation 15) with (6-eloxy-pyridin-3-yl) -isopropylamine (Preparation 2 (B)) (117 mg, 0.65 mmol ) and PCI3 in dichloroean at 100 ° C. Purification by medium-pressure chromatography eluted with a gradient of solvents (5% 0.5% NH 4 OH / MeOH in CH 2 Cl 2 at 10% 0.5% NH 4 OH / MeOH in CH 2 Cl 2) gave 85.1 mg of N- (6-eioxy-pyridin-3-yl) -2- [4- (1 H -indol-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6, 10b-tetraaza-benzo [e] azulen-6-yl] -N-isopropylacetamide. H1 NMR (CD3OD) d 8.05 (d, 1H), 7.62 (m, 3H), 7.38-7.52 (m, 6H), 7.24 (d, 1H), 7.11 ( m, 2H), 6.99 (t, 1H), 6.89 (ni, 3H), 4.78 (m, 1H), 4.53 (m, 1H), 4.35 (q, 2H), 4.12 (m, 1H), 3.88 (f, 1H), 3.78 (dd, 1H), 3.69 (dd, 1H), 1.38 (i, 3H), 0.99 (m , 6H); MS 626.7 (M + 1), 624.6 (M-1). Example 3 (A) Preparation of 2-f4- (1H-lndazol-3-ylmef-5-oxo-1-phenyl-4,5-dhydro-2.3.6.10b-feiraaza-benzo [e1azulen-6 -ilj-isopropyl-N- (6-meioxy-pyridin-3-n-acetylamide) Step A: 3- (6-. {Triopropyl- (6-methoxy-pyridin-3-yl) -carbamoyl] -methyl} -5-oxo-1-phenyl-5,6-dihydro-tert-butyl ester. -4H-2.3.6.1.0b-αetraaza-benzofe1azulen-4-ylmeryl) -indazole-1-carboxylic acid To a solution of N-isopropyl-N- (6-meioxy-pyridin-3-yl) -2- (5-oxo-1-phenyl-4,5-dihydro-2,3,6, 10b-eiraza- benzo [e] azulen-6-yl) -acetamide (Preparation 11) (520 mg, 1.079 mmol) in DMF (100 ml) at 0 ° C was added NaH (60% in acetyl)., 50 mg, 1.27 mmol). The reaction was stirred at 0 ° C for 45 min. And 3-bromomethyl-indazole-1-carboxylic acid tert-buíyl ester (423 mg, 1.36 mmol) in DMF (5 mL) was added. The reaction was stirred at room temperature for 24 hours and diluted with brine. The aqueous solution was washed with ElOAc (3x). The combined organic solutions were washed with brine (1x), dried (MgSO 4), filled and concentrated in vacuo to give 3- (6- { [Isopropyl- (6-methoxy) tert -butyl ester. -pyridin-3-yl) -carbamoyl] -methyl) -5-oxo-1-phenyl-5,6-dihydro-4H-2,3,6, 10b-tetraaza-benzo [e] azulen-4-ylmeity -indazole-1-carboxylic acid. H1 NMR (CD3OD) d 8.05 (d, 1H), 6.80-7.75 (m, 15H), 4.75 (m) and 4.60 (f, total 3H), 3.85-4 , 20 (m, 6H), 1.65 (m, 9H), 0.98 (m, 6H); MS 713.6 (M + 1), 711.5 (M-1). Step B: 2-r4- (1H-lndazol-3-ylmethyl-5-oxo-1-phenyl-4,5-dihydro-2.3.6.10b-tetraaza-benzo [e] azulen-6-yN-isopropyl-N- (6-methoxy-pyridin-3-ip-acetamide The product from Step A, 3- (6-. {[[Isopropyl- (6-mephoxy-pyridin-3-yl) tert-buffolester) -carbamoyl] -methyl.} - 5-oxo-1-phenyl-5,6-dihydro-4 H-2,3,6, 10b-ephaza-benzo [e] azulen-4-ylmefyl) -indazole-1 carboxylic acid was dissolved in CH2CI2 (8 ml) and TFA (2 ml) was added.The reaction was stirred at room temperature for 24 hours and concentrated in vacuo, purification by medium pressure chromatography eluting with a gradient of solvents ( 1% of MeOH in EtOAc to 5% MeOH in EtOAc) gave 172 mg of 2- [4- (1 H -indazol-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3, 6,10b-tetraazabenzo [e] azulen-6-yl] -N-isopropyl-N- (6-methoxy-pyridin-3-yl) -acetamide. H1 NMR (CD3OD) δ 8.04 (d, 1H), 7.88 (d, 1H), 7.64 (m, 2H), 7.56 (d, 2H), 7.49 (t, 2H) , 7.39 (q, 3H), 7.31 (t, 1H), 7.19 (t, 1H), 7.11 (t, 1H), 6.95 (d, 1H), 6.86 ( m, 1H), 4.72 (m, 1H), 4.59 (m, 1H), 4.41 (t, 1H), 3.99-4.12 (m, 2H), 3.90 (m , 4H), 0.95 (m, 6H); MS 613.8 (M + 1), 611.5 (m-1). Example 3 (B) Preparation of 2- [4- (1 H-ldazol-3-ylmethyl-D-5-oxo-1-phenyl-4,5-dihydro-2.3.6. IOb-. Tetraaza-benzofelazulen-6-in -N-isopropyl-N-phenyl-acetamide Stage A: 3-f6-ITIsopropanyl-phenyl-carbamoyl) -methyl-5-oxo-1-phenyl-5,6-dihydro-2,3,6,10,10-benzo [e] azulen-4-ferric acid ester -ilmefil} -indazole-1-carboxylic Following the procedure described for Example 3 (A), Step A, N-isopropyl-2- (5-oxo-1-phenyl-4,5-dihydro-2,3,6, 10b-αetraaza-benzo [e] a zulen-6-yl) -N-phenyl-acelamide (Preparation 7 (A)) (411.7 mg, 0.9118 mmol) with 3-bromomelyl acid-3-bromomethyl ester -indazol-1-carboxylic acid (309 mg, 0.993 mmol). Purification by means of medium pressure chromatography eluting with a gradient of solvents (EtOAc to 5% MeOH in EtOAc) provided 391.9 mg of 3- tert-buíyl ester. { 6 - [(isopropyl-phenyl-carbamoyl) -mef] -5-oxo-1-phenyl-5,6-dihydro-4H-2,3,6, 10b-leraza-benzo [e] azulen-4- ilmelil} -indazol-1-carboxylic acid. H1 NMR (CD3OD) d 7.99 (m, 2H), 7.22-7.66 (m, 15H), 7.01 (dd, 1H), 4.53-4.81 (m, 3H), 3.88-4.21 (m, 3H), 1.65 (m, 9H), 0.94 (m, 6H). Stage B: 2-r4- (1H-lndazol-3-llmetin-5-oxo-1-phenyl-4,5-dihydro-2.3.6.10b-teiraza-benzofe] azulen-6-ip-N-¡ sopropil-N-phenyl-acetylamide To a solution of 3- ({6 - [(isopropyl-phenyl-carbamoyl) -methyl] -5-oxo-1-phenyl-5,6-dihydro} -deric acid ester. 4H-2,3,6,10b-telraza-benzo [e] azulen-4-ylmethyl] -indazole-1-carboxylic acid (391.9 mg, 0.674 mmol) in dioxane (8 mL) was added 4M HCl in dioxane (6 ml) The reaction was stirred at room temperature for 24 hours and diluted with EtOAc The organic solution was washed with NaHCO 3 and brine, dried (MgSO 4), filtered and concentrated in vacuo. by mean pressure chromatography eluting with a gradient of solvents (10% MeOH in EOAc) gave 260.9 mg of 2- [4- (1H-indazol-3-ylmethyl) -5-oxo-1-phenyl- 4,5-dihydro-2,3,6,10b-tetraaza-benzo [e] azulen-6-yl] -N-isopropyl-N-phenylacetamide in the form of a racemic mixture H1 NMR (CD3OD) d 7.90 (d, 1H), 7.65 (d, 2H), 7.34-7.54 (m, 12H), 7.22 (t , 1H), 7.13 (1, 1H), 6.99 (d, 1H), 4.74 (m, 1 H), 4.65 (d, 1H), 4.42 (1, 1H), 3.92-4.12 (m, 3H), 0.99 (m, 6H); MS 582.7 (M + 1), 580.4 (M-1).
ALTERNATIVE PREPARATION: Stage A: N-lsopropyl-2- (5-oxo-1-phenyl-4,5-dihydro) -2.3.6.10b-teiraza-benzo [e1azulen-6-yl) -N-phenyl-acetamide To a solution of 1-phenyl-4H, 6H-2,3,6,10b-tetraaza-benzo [e] azulen-5-one (Preparation 4 (A)) (2.5 g, 9.05 mmol) in DMF (40 ml ) at 0 ° C was added sodium hydride (60% in oil, 0.36 g, 9.0 mmol). The reaction was warmed to room temperature and stirred for 40 minutes. The reaction was cooled to -6 ° C and a solution of 2-bromo-N-isopropyl-N-phenyl-acetamide (Preparation 1 (A)) (2.55 g, 9.95 mmol) in DMF (20%) was added. ml) was added dropwise to the reaction mixture for 0.5 hours, maintaining the internal temperature below -3 ° C. The reaction mixture was stirred for 105 minutes below -3 ° C and brine (200 ml) was added. The aqueous solution was washed with ethyl acetate (200 ml). The organic layer was washed with brine, dried over sodium sulfate and concentrated to 1/3 of its volume. Hexanes were slowly added until a solid separated. The white solid was collected by filtration and rinsed with ether and hexanes to provide 2.96 g of N-isopropyl-2- (5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b- leiraza-benzo [e] azulen-6-yl) -N-phenylaceiamide. This intermediate was further treated according to Example 3 (B), Steps A and B above to provide 2- [4- (1 H -indazol-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2 , 3,6,10b-ioaraza-benzo [e] azulen-6-yl] -N-isopropyl-N-phenylacetamide in the form of a racemic mixture.
Example 3 (C) Preparation of (-) 2- [4- (1H-lndazol-3-ylmeiyl) -5-oxo-1-phenyl-4,5-dihydro-2.3.6.10b-tetraaza-benzolre1azulen-6-nN -isopropyl-N-phenol-acetamida. enantiomer 2 2- [4- (1H-indazol-3-ylmethyl) -5-oxo-1-phenyl-4,5-d -hydro-2,3,6, 10b-tetraaza-benzo [e was separated ] azulen-6-yl] -N-isopropyl-N-phenyl-acetamide (Example 3 (B)) racemic in the form of its enaniomers using a Chiralcel OD column (10 cm x 50 cm eluting with heptane / efanol (3: 1) ) containing 0.05% diethylamine, using a flow rate of 250 ml / minute, to provide (-) 2- [4- (1 H -indazol-3-ylmethyl) -5-oxo-1-phenyl-4, 5-dihydro-2,3,6, 10b-terazaza-benzo [e] azulen-6-yl] -N-isopropyl-N-phenyl-acetamide (enantiomer 2) having a retention time of 9.21 minutes. H1 NMR (CD2Cl2) d 7.94 (d, 1H), 7.62 (d, 2H), 7.34-7.47 (m, 10H), 7.10-7.22 (m, 4H), 6.89 (d, 1H), 4.84 (m, 1H), 4.41 (br d, 1H), 4.35 (t, 1H), 3.93-4.11 (m, 3H), 1.00 (m, 6H); MS 582.5 (M + 1); [a] D20 -109.6 (c 1.1, eneol) Example 3 (D) Preparation of N-Benzyl-2-r8 .9-difluoro-4- (1 H-indazol-3-iimetiD-5-oxo-1-phenyl-4,5-dihydro-2.3.6.10b-teiraza-benzorelazulen-6-nN-isopropyl-acetamide Stage A: 3- (6-R (Benzyl-isopropyl-carbamoyl) -methin-8,9-difluoro-5-oxo-1-phenyl-5,6-dihydro-4H-2,3,6,10-tetraaza- benzofe1azulen-4-ylmethyl) -indazole-1-carboxylic acid Following the procedure described in Example 3 (A), Epaque A, N-benzyl-2- (8,9-difluoro-5-oxo-1-phenyl- 4,5-dihydro-2, 3,6,1 Ob-ephaza-benzo [e] azulen-6-yl) -N-isopropyl-acefamide with 3-bromomethyl-indazole-1-carboxylic acid alkyl ester ( 136 mg, 0.439 mmol). The reaction was diluted with a buffer solution to pH 6.8 and the aqueous solution was extracted with EtOAc. The organic solution was washed with brine (4x). dried (Na2SO4), filtered and concentrated in vacuo. Purification by means of medium pressure chromatography eluting with a gradient of solvents (CH2Cl2 at 30% of MeOH in CH 2 Cl 2) gave 100 mg of 3-acid eeryc-buyl ester. { 6- [(benzyl-isopropyl-carbamoyl) -methyl] -8,9-difluoro-5-oxo-1-phenyl-5,6-dihydro-4H-2,3,6, 10b-ioraza-benzo [e] azulen-4-ilmelil} indazol-1-carboxylic acid MS 732.5 (M + 1), 730.4 (M-1). Eiaoa B: N-Benzyl-2-b8.9-difluoro-4- (1 H-indazol-3-limelin-5-oxo-1-phenyl-4,5-dihydro-2.3.6.10b-eiciraza-benzofelazulen -6-yl] -N-isopropyl-acetylamide To a solution of 3-, {6 - [(benzyl-isopropyl-carbamoyl) methyl] -8,9-difluoro-5-oxo-1-tert-butyl ester. phenyl-5,6-dihydro-4H-2,3,6,10b-teiraza-benzo [e] azulen-4-ylmethyl] -indane-1-carboxylic acid (100 mg, 0.137 mmol) in dioxane (1 ml) was added HCl (4.0M in dioxane, 2 ml) The reaction was stirred at room temperature for 24 hours The volatile components were concentrated in vacuo and the residue was dissolved in CH2Cl2. it was washed with aqueous NaHC03 (1x), dried (Na2SO4), filtered and concentrated in vacuo. A purification by preparative chromatography eluting with 5% MeOH in CH 2 Cl 2 yielded 45 mg of N-benzyl-2- [8,9-difluoro-4- (1 H -indazol-3-methylmethyl) -5-oxo-1- phenyl-4,5-dihydro-2,3,6,10b-lerazaza-benzo [e] -azulen-6-yl] -N-isopropyl-acelamide. H1 NMR (CD2Cl2) d 7.92 (m, 1H), 7.04-7.66 (m, 14H), 6.67 (s, 1H), 4.22-4.96 (m, 6H), 3.91-4.13 (m, 21-1), 1.01-1.22 (m, 6H); MS 632.3 (M + 1), 630.2 (M-1). Example 3 (E) Preparation of (-) N-Benzyl-2-14- (1H-indazol-3-ylmeiyl) -5-oxo-1-phenyl-4,5-dihydro-2.3.6.10b-eeryza-benzore-1azulen-6 -in-N-isopropyl-acetylamide. enaniomer 2 Stage A: Etheric acidic acid 3-. { 6 - [(Benzyl-isopropyl-carbamo? O-mefyl] -5-oxo-1-phenyl-5,6-dihydro-4H-2,3,6,10-tefraza-benzo [e] azu-len-4-ylmeityl. Indazole-1-carboxylic Following the procedure described in Example 3 (A), Step A, N-benzyl-N-isopropyl-2- (5-oxo-1-phenyl-4,5-dihydro-2) was alkylated. , 3,6,1 Ob-ioraza-benzo [e] azulen-6-yl) -acefamide with 3-bromomethyl-indazole-1-carboxylic acid tert-buíyl ester (150 mg, 0.473 mmol) The reaction was diluted with a buffer solution at pH 6.8 and the aqueous solution was washed with EOAc. organic solution was washed with brine (3x), dried (Na2SO4), filtered and concentrated in vacuo. The residue was purified (2x) by preparative chromatography eluting with 5% acetone in CH2Cl2 followed by medium pressure chromatography eluting with a gradient of solvents (2% acetone in CH2Cl2a 14% acetone in CH2Cl2) to provide 19.1 mg of 3-tert-butyl ester. { 6 - [(benzyl-isopropyl-carbamoyl) -methyl] -5-oxo-1-phenyl-5,6-dihydro-4H-2,3,6,10b-teiraza-benzo [e] azulen-4-ylmelyl} -indazol-1-carboxylic acid. H1 NMR (CD2Cl2) d 8.05 (m, 1H), 7.12-7.71 (m, 15H), 7.03 (m, 1 H), 6.92 (d, 1H), 5.21 (d) and 4.90 (d, iohal 1H), 4.61-4.72 (m, 2H), 4.49 (s, 1H), 4.43 (t, 1H), 4.25-4 , 32 (m, 1H), 3.96-4.15 (m, 2H), 1.61 (d, 9H), 1, 16 (dd, 3H), 1.04 (dd, 3H); MS 696.4 (M + 1). Step B: N-Benzyl-2-r4- (1 H-indazol-3-ylmellyl) -5-oxo-1-phenyl-4,5-dhydro-2.3.6.10b-teiraza-benzo [e] azulen-6 -l] -N-isopropyl-acetylamide Following the procedure described in Example 3 (B), Step B, tert-buíyl ester of 3-acid was desprolected. { 6 - [(benzyl-isopropylcarbamoyl) -methyl] -5-oxo-1-phenyl-5,6-dihydro-4H-2,3,6, 10b-tetraazabenzo [e] azulene-4-ylmethyl} - indazol-1-carboxylic acid (19.1 mg, 0.274 mmol) with HCl (4.0M in dioxane, 0.8 ml) for 24 hours. The reaction was diluted with EfOAc and the organic solution was washed with aqueous NaHCO3 (1x), dried (Na2S4), filtered and concentrated. Purification by preparative chromaeography eluting with 5% MeOH in CH 2 Cl 2 yielded 8.5 mg of N-benzyl-2- [4- (1 H -indazol-3-ylmethyl) -5-oxo-1-phenyl-4,5- dihydro-2,3,6,10b-yl-aza-benzo [e] azulen-6-yl] -N-isopropyl-acelamide in the form of a racemic mixture. H1 NMR (CD2CI2) d 7.97 (t, 1H), 7.12-7.64 (m, 15H), 7.05 (m, 1H), 6.92 (m, 1H), 5.08 (d) and 4.81 (d, total 1H), 4, 60-4.74 (m, 2H), 4.49 (s, 1H), 4.45 (t, 1H), 4.29-4.39 (m, 1H), 3.96-4.13 ( m, 2H), 1, 16 (dd, 3H), 1, 05 (dd, 3H); MS 596.2 (M + 1), 594.2 (M-1). Step C: (-) N-Benzyl-2-r4- (1 H -indadazol-3-ylmethyl-5-oxo-1-phenyl-4,5-dihydro-2, 3.6.10b-tetraaza-benzofe] azulen-6-yl ] -N-isopropyl-acetamide enaniomer 2 The racemic production of EIAP B, N-benzyl-2- [4- (1 H-indazol-3-ylmethyl) -5-oxo-1-phenyl-4,5- dihydro-2,3,6,10b-telraza-benzo [e] azulen-6-yl] -N-isopropyl-acelamide (1.57 g, 2.64 mmol), was separated as its enaniomers using a column Chiralcel OD (10 cm x 25 cm, eluting with 50% EOH in heptane, using a flow rate of 250 ml / minute.) The enantiomer having a retention time of 17.396 minutes (enantiomer 2) was dissolved in CH2Cl2 and the organic solution it was washed with aqueous NaHC 3 (1x) and brine (1x), the organic solution was dried (Na 2 SO 4), filtered and concentrated to provide 660 mg of (-) N-benzyl-2- [4- (1 H). -indazol-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-tetraaza-benzo [ejazulen-6-yl] -N-isopropylacetamide (enaniomer 2) H1 NMR (CD2Cl2) d 7.97 (1, 1H), 7.12-7.65 (m, 15H), 7.05 (m, 1H), 6.92 (m, 1H), 5.10 (d) and 4.82 (d, total 1H), 4.60-4.73 (m, 2H), 4.49 (s, 1H) , 4.45 (t, 1H), 4.29-4.39 (m, 1H), 3.96-4.13 (m, 2H), 1.16 (dd, 3H), 1.05 (dd) , 3H); MS 596.1 (M + 1), 594.0 (M-1); [α] D20-145.3 (c 1.01, ethanol).
Example 4 Preparation of 2- [4- (1 H-lndol-3-ylmefyl-4-methyl-5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-tetraaza-benzore] azulen-6 -nN-isopropyl-N-phenol cetamyl Stage A: Etheric acid-buflic acid 3-. { 6 - [(lsopropyl-phenyl-carbamoyl) -methyl] -5-oxo-1-phenyl-5,6-dihydro-4H-2.3.6.1 Ob-fef-raaza-benciljazulen-4-ylmeityl} indole-1-carboxylic acid To a solution of 2- [4- (1H-indol-3-ylmeryl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-tetraaza-benzo [ e] azulen-6-yl] -N-isopropyl-N-phenyl-acetamide (Example 6 (A)) (250 mg, 0.431 mmol) in CH 2 Cl 2 (6 mL) at 0 ° C was added DMAP (5 mg, , 0431 mmol) and a dicarbonate solution of di-tert-buty (103 mg, 0.47 mmol) in CH2Cl2 (3 mL). The reaction was stirred at room temperature for 4.5 hours. The reaction was diluted with water and the organic layer was dried (Na2SO4), filtered and concentrated. Purification by means of medium pressure chromatography eluting with a gradient of solvents (10% acetone in CH 2 Cl 2 to 40% acetone in CH 2 Cl 2) gave 290 mg of 3-tert-buiyl ester. { 6 - [(isopropyl-phenyl-carbamoyl) -methyl] -, 5-oxo-1-phenyl-5,6-dihydro-4H-2,3,6,10b-l-aza-benzo [e] azulen-4-ylmeil } -indol-1 -carboxylic. H1 NMR (CDCl3) d 7.33-8.10 (m, 15H), 6.90-7.20 (m, 4H), 4.15-4.90 (m, 2H), 3.65-4 , 05 (m, 4H), 1.63 (m 9H), 0.97 (m, 6H); MS 681.2 (M + 1).
Step B: 3- (6-Ylsopropyl-phenyl-carbamoyl) -methyl-4-mephyl-5-oxo-1-phenyl-5,6-dihydro-4H-2,3,6.10b-tetraazaic acid etruric acid ester -benzoy1azulen-4-ylmeiyl} -indole-1-carboxylic acid To a solution of 3- tert-butyl ester. { 6 - [(isopropyl-phenyl-carbamoyl) -methyl] -5-oxo-1-phenyl-5,6-dihydro-4H-2,3,6, 10b-tetraaza-benzo [e] azulen-4-ylmethyl} -indole-1-carboxylic acid (100 mg, 0.147 mmol) in DMF (2 ml) at -5 ° C was added KHMDS (0.5 M in THF, 322 μl, 0.161 mmol). The solution was stirred at -5 ° C for 20 minutes and methyl iodide (10 μL, 0.161 mmol) was added. The reaction was stirred at room temperature for 24 hours, diluted with ElOAc and the organic solution was washed with brine (3x). The organic solution was dried (Na2SO), filtered and concentrated. The residue was purified by preparative chromatography (2x) eluting with 50% EtOAC in hexanes to provide 34 mg of 3-tert-butyl ester. { 6 - [(sopropii-phenyl-carbamoyl) -methyl] -4-methyl-5-oxo-1-phenyl-5,6-dihydro-4H-2, 3,6,1 Ob-tetraaza-benzo [e] azulen-4-ilmeil} -indol-1 -carboxylic. H1 NMR (CD2Cl2) d 8.05 (d, 1H), 7.67 (d, 1H), 7.36-7.54 (m, 10H), 7.08-7.30 (m, 6H), 6.90 (d, 1H), 5.02 (m, 1H), 4.29 (d, 1H), 4.11 (m, 1 H), 2.78 (s, 2H), 1.80 ( s, 3H), 1, 64 (s, 9H), 1, 10 (m, 6H); MS 695.3 (M + 1). Step C: 2- (4- (1H-lndol-3-ylmethyl) -4-methyl-5-oxo-1-phenyl-4,5-dihydro-2.3.6.10b-tetraaza-benzpe1azulen-6-ip-N -sopropyl-N-phenyl-acetamide To a solution of 3- ({6 - [(isopropyl-phenyl-carbamoyl) -methyl] -4-methyl-5-oxo-1-phenyl- tert-buíyl ester. 5,6-Dihydro-4H-2,3,6,10b-tetraaza-benzo [e] azulen-4-ylmethyl] -indol-1-carboxylic acid in dioxane (1 ml) was added HCl (4.0M in dioxane, 550 μl) and the reaction was stirred at ambient temperature for 50 hours. The volatile components were concentrated in vacuo and the residue was dissolved in CH2Cl2. The organic solution was washed with aqueous NaHC 3 (1x), dried (Na 2 SO 4), filtered and concentrated. Purification by preparative chromoiography eluting with 40% of acetyone in CH 2 Cl 2 yielded 17 mg of 2- [4- (1 H -indol-3-methylmethyl) -4-methyl-5-oxo-1-phenyl-4,5-dihydro -2,3,6, 10b-ioraza-benzo [e] azulen-6-yl] -N-isopropyl-N-phenylacelamide. H1 NMR (CD2Cl2) d 8.37 (s, 1H), 7.78 (m, 1H), 7.53-7.23 (m, 13H), 7.11 (m, 2H), 7.02 ( 1H), 6.90 (d, 1H), 5.03 (m, 1H), 4.33 (d, 1H), 3.76-3.58 (m, IH), 2.83 (m , 2H), 1.75 (s, 3H), 1.11 (m, 6H); MS 595.2 (M + 1). Example 5 (A) Preparation of 2- [1- (2-Fluorophenyl) -4- (1 H -indazol-3-ylmeiyl) -5-oxo-4,5-dihydro-2,3,6,10-teiraza-benzo [ elazulen-6-pN-isopropyl-N-phenyl-acetamide Following the procedure described for Preparation 4 (C), 2-fluoro-benzoic acid hydrazide was reacted with 2- (4-eioxy-2-oxo-2,3-dihydro-benzo [b] [1,4] diazepin-1-yl) -N-isopropyl-N-phenyl-acetylamide (Preparation (B)). This intermediate was continued to be sprayed according to Example 3 (B) Lower A and B Epaque to provide 2- [1- (2-fluoro-phenyl) -4- (1H-indazol-3-ylmethyl) -5-oxo- 4,5-dihydro-2,3,6,10b-letraazabenzo [e] azulen-6-yl] -N-isopropyl-N-phenyl-acetamide. H1 NMR (CD2Cl2) d 7.92 (d, 1H), 7.96 (dt, 1H), 7.59 (d, 1H), 7.33-7.50 (m, 7H), 7.15- 7.25 (m, 4H), 7.02-7.10 (m, 2H), 6.82 (dd, 1 H), 4.90-4.96 (m, 1H), 4.38 (t , 1H), 4.17 (d, 1H), 3.92-4.03 (m, 3H), 1. 04 (m, 6H); MS 600 (M + 1). Example 5 (B) Preparation of 2-1-1- (3-Fluoro-phenyD-4- (1 H -indazol-3-ylmeipy-5-oxo-4,5-dihydro-2,3,6.10b-teiraza-benzo [e ] azulen-6-pN-isopropyl-N-phenyl-acetamide Following the procedure described for Preparation 4 (C), Step A, 3-fluoro-benzoic acid hydrazide was reacted with 2- (4-ethoxy-2-oxo-2,3-dihydrobenzo [b] [1.4 ] diazepin-1-yl) -N-isopropyl-N-phenyI-acetamide (Preparation 5 (B)). This intermediate was further treated according to Example 3 (B) Steps A and B to provide 2- [1- (3-fluoro-phenyl) -4- (1 H -indazol-3-ylmethyl) -5-oxo-4,5 -dihydro-2,3,6,10b-tetraaza-benzo [e] azulen-6-yl] -N-isopropyl-N-phenyl-acetamide. H1 NMR (CD2Cl2) d 7.95 (d, 1H), 7.35-7.50 (m, 10H), 7.15-7.21 (m, 5H), 6.91 (d, 1H), 4.76-4.83 (m, 1H), 4.52 (d, 1H), 4.34 (t, 1H), 3.92-4.08 (m, 3H), 0.98 (dd, 6H); MS 600 (M + 1).
Example 5 (C) Preparation of 2- [1-Cyclohexyl-4- (1 H-indazol-3-ylmeill) -5-oxo-4,5-dihydro-2.3.6.1 O-tetraaza-benzoylazulen-6-ip-N- isopropyl-N-phenyl-acetylamide Following the procedure described for Preparation 4 (C), Step A, cyclohexanecarboxylic acid hydrazide (Preparation 14) was reacted with 2- (4-ethoxy-2-oxo-2,3-dihydro-benzo [b] [1,4] diazepin-1-yl) -N- isopropyl-N-phenylacetamide (Preparation 5 (B)). This intermediate was further treated according to Example 3 (B) Steps A and B to provide 2- [1-cyclohexyl-4- (1 H -indazol-3-ylmethyl) -5-oxo-4,5-dihydro-2, 3.6, 10b-lerazaza-benzo [e] azulen-6-yl] -N-isopropyl-phenylacetamide. H1 NMR (CD2Cl2) d 7.92 (d, 1H), 7.51-7.61 (m, 3H), 7.37-7.50 (m, 6H), 7.16-7.20 (m , 3H), 4.85 (m, 1H), 4.24 (m, 1H), 4.00 (d, 1H), 3.93 (d, 1H), 2.77-2.85 (m, 1H), 2.17 (d, 1H), 1.91 (d, 2H), 1.07-1.66. (br.m, 3H), 1.01 (dd, 6H); MS 588 (M + 1). Example 5 (D) Preparation of 2- [1- (4-Fuuoro-phenyl-4- (1H-indazol-3-ylmethy) -5-oxo-4,5-dihydro-2.3.6.10b-tefrazaz- benzore1azulen-6-yl] -N-isopropyl-N-phenyl-acetylamide Following the procedure described for Example 4 (C), 4-fluorobenzoic acid hydrazide was reacted with 2- (4-eloxy-2-oxo-2,3-dihydro-benzo [b] [1,4] diazepin- il) -N-isopropyl-N-phenyl-acefamide (Preparation 5 (B)). This medium was further treated according to Example 3 (B) Steps A and B to provide 2- [1- (4-fluorophenyl) -4- (1 H -indazol-3-ylmethyl) -5-oxo-4,5-dihydro -2,3,6, 10b-ephazaza-benzo [e] azulen-6-yl] -N-isopropyl-N-phenyl-acetamide. H1 NMR (CD2Cl2) d 7.92 (d, 1H), 7.64-7.67 (m, 2H), 7.32-7.44 (m, 7H), 7.07-7.20 (m , 6H), 6.87 (d, 1H), 4.77-4.84 (m, 1H), 4.48 (d, 1H), 4.32-4.36 (m, 1H), 3, 91-4.05 (m, 3H), 0.98 (dd, 6H); MS 600 (M + 1). Example 5 (E) Preparation of N- (4-Fluoro-phenyl) -2-r4- (1 H -indazole-3-ylmethyl-5-oxo-1-phenyl-4,5-dihydro-2.3.6.10b- teiraaza-benzore1azulen-6-yl-N-isopropyl-acetamide Following the procedure described for Preparation 4 (C), Step A of the Alternate Preparation, 1-phenyl-4H, 6H-2, 3,6,10b was alkylated -fetraaza-benzo [e] azulen-5-one (Preparation 4 (A)) (1.0 g, 3.62 mmol) with 2-bromo-N- (4-fluoro-phenyl) -N-isopropyl-acefamide (Preparation 1 (D)) (1.1 g, 3.98 mmol) This intermediate was followed according to Example 3 (B) Steps A and B to give N- (4-fluorophenyl) -2- [4- (1 H-indazol-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-feiraza-benzo [e] azulen-6-yl] -N-isopropyl- acetylamide, H1 NMR (CD2CI2) d 7.94 (d, 1H), 7.63 (d, 2H), 7.38-7.59 (br.m, 7H), 7.12-7.29 (br m, 6H), 6.90 (d, 1H), 4.79-4.86 (m, 1H), 4.36-4.45 (br.m, 2H), 3.95-4.05 (br.m, 3H), 0.98 (dd, 6H), MS 600 (M + 1).
Example 6 (A) Preparation of 2-f4- (1 H-lndol-3-ylmef-5-oxo-1-phenyl-4,5-dihydro-2.3.6.10b-tetra aza-benzoyl-azulen-6-in-N -isopropyl-N-phenyl-acetamide Stage A: 2-f4- (1 H-lndol-3-methylmethylene) -5-oxo-1-phenyl-4,5-dihydro-2.3.6.10b-teiraza-benzofe] azulen-6-ip-N-sopropil -N-phenyl-acetylamide To a solution of N-isopropyl-2- (5-oxo-1-phenyl-4,5-dihydro-2,3,6, 10b-teirazazazo [e] azulen-6-yl) - N-phenyl-acetylamide (Preparation 7 (A)) (1.2 g, 2.7 mmol) in uelen (15 ml) was added 1 H-indole-3-carboaldehyde (0.46 g, 3.2 mmol) and piperidine (0.4 ml). The reaction mixture was refluxed for 14 hours. Activated 4A® molecular sieves (2 g) were added and the reaction mixture was refluxed for 24 hours. The components were separated by filtration and the filtrate was concentrated. The residue was triturated with melylene chloride and the solid was collected through filtration and dried under vacuum to provide 1.5 g of 2- [4- (1H-indol-3-ylmethylene) -5-oxo-1- phenyl-4,5-dihydro-2,3,6,101b-tetraaza-benzo [e] azulen-6-yl] -N-isopropyl-N-phenyl-acetamide. The filtrate was concentrated and purified by chromatography (20% acetone / methylene chloride) to give an additional 320 mg of 2- [4- (1 H-indol-3-methylmethylene) -5-oxo-1-phenyl- 4,5-dihydro-2,3,6,10b-leiraza-benzo [e] azulen-6-yl] -N-isopropyl-N-phenyl-acetamide. MS 579.2 (M + 1).
Epaque B: 2- [4- (1H-lndol-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-ietraazabenzo [e] azulen-6-yl] - N-isopropyl-N-phenyl-acetamide To 10% Pd / C and EiOH was added 2- [4- (1H-indol-3-phenylmethylene) -5-oxo-1-phenyl-4,5-dihydro-2 , 3, 6,10b-ephaza-benzo [e] azulen-6-yl] -N-isopropyl-N-phenylacetylamide (0.71 g, 1.2 mmol). An additional 25 ml of EOAc followed by ammonium formate (0.77 g, 12.2 mmol) was added. The reaction mixture was heated at 80 ° C for 6 hours. The catalyst was removed by filtration through Celite® and the filtrate was concentrated. The residue was dissolved in ethyl acetate and the organic solution was washed with saturated NaHC 3. The organic phase was dried over sodium sulfate and concentrated. The crude product was purified by chromatography (0% to 18% acetone / methylene chloride) to provide 128 mg of 2- [4- (1H-indol-3-ylmeryl) -5-oxo-1-phenyl-4 , 5-dihydro-2,3,6, 10b-leirazazabenzo [e] azulen-6-yl] -N-isopropyl-N-phenyl-acetamide in the form of a racemic mixture. H1 NMR (CD2Cl2) d 8.18 (s, 1H), 7.64 (dd, 2H), 7.55 (dd, 1H), 7.510-7.41 (m, 8H), 7.36 (dd, 1H), 7.25 (m, 3H), 7.09 (m, 2H), 7.027 (m, 1H), 6.85 (dd, 1H), 4.86 (m, 1H), 4.40 (br d, 1H), 4.04 (dd, 1H), 3.79 (m, 3H), 1.03 (dd, 6H); MS 581.4 (M + 1). Example 6 (B) Preparation of (-) 2-r4- (1H-lndol-3-methylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2.3.6.10b-tetraaza-benzofe1azulen-6-in- N -sopropyl-N-phenyl-acetamide. enantiomer 1 Separated were 2- [4- (1H-indol-3-methylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6,1 Ob-oef raazabenzo [e] azulen- 6-yl] -N-isopropyl-N-phenol-acetylamide (Example 6 (A)) in its enaniomers using a Chiralpak AD column (10 cm x 50 cm), eluting with heptane / efanol (80:20) using a flow rate of 250 ml / minute to provide (-) 2- [4 - (1 H-indo) -3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6, 10b-tetraazabenzo [e] azulen-6-yl] - N-isopropyl-N-phenyl-acetamide (enanlimer 1) having a retention time of 8.64 hours. MS 581 (M + 1); [a] D20 -91.4 (c 1, 04, eianol). Example 6 (C) Preparation of 2- [1-Cyclohexyl-4- (1H-indol-3-ylmeiip-5-oxo-4,5-dihydro-2.3.6.10b-eiseraza-benzo [e] azulen-6 il1-N-isopropyl-N-phenyl-acetylamide Following the procedure described for Example 6 (A), Eíapa A; 2- (1-cyclohexyl-5-oxo-4,5-dihydro-2,3,6,10b-eeryaraza-benzo [e] azulen-6-yl) -N-isopropyl-N-phenylacelamide was condensed (Preparation 8) (B)) with 1 H -indole-3-carbaldehyde. This medium was further reduced according to Example 6 (A) Efapa B to provide 2- [1-cyclohexyl-4- (1H-indol-3-ylmeryl) -5-oxo-4,5-dihydro-2, 3,6 , 10b-telraazabenzo [e] azulen-6-yl] -N-isopropyl-N-phenyl-acefamide. H1 NMR (CD2Cl2) d 8.44 (s, 1H), 7.42-7.48 (m, 4H), 7.28-7.42 (m, 4H), 7.17-7.19 (m , 2H), 7.07-7.10 (m, 1H), 6.98-7.02 (m, 1H), 4.90-4.94 (m, 1H), 4.07-, 15 ( m, 1H), 3.91 (d, 1H), 3.67-3.73 (m, 2H), 2.83 (br.m, 1H), 2.19 (br.m, 1H), 1, 89-1.92 (ni, 2H), 1, 68 (br.s, 4H), 1, 50-1.59 (m, 1H), 1.32-1, 35 (m, 2H), 1, 23 (1, 1H), 1, 06 (m, 6H); MS 587 (M + 1). Example 6 (D) Preparation of 2 - ("1 - (2-Fluoro-phenyl) -4- (1 H -indol-2, -ylmefiD-5-oxo-4,5-dihydro-2.3.6.10b-eieraza-benzo [e1azulen-6-yl] -N-isopropyl-N-phenylaliamide Following the procedure described for Example 6 (A), Step A, 2- (1- (2-fluorophenyl) -5-oxo-4,5-dihydro-2, 3,6,1 Ob-eieiraza-benzo [e] azulen-6-yl) -N-isopropyl was condensed -N-phenyl-acetamide (Preparation 7 (B)) with 1H-indole-3-carbaldehyde. This intermediate was further treated according to Example 6 (A) Step B to provide 2- [1- (2-fluoro-phenyl) -4- (1 H -indol-3-ylmeryl) -5-oxo-4,5- dihydro-2,3,6, 10b-lefraazabenzo [e] azulen-6-yl] -N-isopropyl-N-phenyl-acetamide. H1 NMR (CD2Cl2) d 8.26 (s, 1H), 7.71-7.74 (m, 1H), 7.60-7.67 (m, 2H). 7.40-7.59 (m, 6H), 7.34-7.38 (m, 2H), 7.23-7.32 (br.m, 2H), 7.14-7.20 (m , 1H), 7.05-7.12 (m, 3H), 6.81-6.83 (m, 1H), 4.98-5.03 (m, 1H), 4.28 (d, 1 H), 3.93 (d, 1H), 3.83-3.84 (m, 3H), 1.12 (d, 6H); MS 599 (M + 1). Example 7 Preparation of 2-f4- (1H-lndol-3-ylmethylene) -5-oxo-1-phenyl-4,5-dihydro-2.3.6.10b-teiraza-benzo [e1azulen-6-ylj-N-isopropyl-N-phenyl] -aceylamide To a solution of N-isopropyl-2- (5-oxo-1-phenyl-4,5-dihydro-2,3,6, 10b-lef-raaza-benzo [e] azulen-6-yl) -N-phenyl -acetamide (Preparation 7 (A)) (1.2 g, 2.7 mmol) and piperidine (0.4 mL, 1.33 mmol) in toluene was added 1 H -indole-3-carbaldehyde (0.46 g, 3.2 mmoi). The reaction mixture was heated to 110 ° C for 31 hours. Added 4A® molecular sieves (2 g) were added. The reaction mixture was heated for 24 hours at 110 ° C. The reaction was cooled to room temperalure and the molecular variables were filtered with the aid of toluene. The filtrate was concentrated and the residue was purified by chromatography (12% -20% acetone / methylene chloride) to provide 1.5 g of 2- [4- (1 H -indol-3-ylmethylene) -5-oxo- 1-phenyl-4,5-dihydro-2,3,6,1 Ob-tetraaza-benzo [e] azulen-6-yl] -N-isopropyl-N-phenyl-acetamide in the form of a solid. MS 579.2 (M + 1). Example 8 Each of the compounds of the following Table can be prepared by procedures similar to those described in the previous Examples.
Preparation 1 (A) Preparation of 2-Bromo-N-isopropyl-N-phenyl-acetamide To a solution of N-isopropylaniline (21.58 g, 180 mmol) in CH2Cl2 (350 mL) was added triethylamine (26.7 mL, 190 mmol). The solution was cooled to 0 ° C and bromoacetyl bromide (14.0 ml, 160 mmol) was added slowly over 1 hour. The reaction was warmed to room temperature and stirred for 24 hours. The reaction was diluted with 5% aqueous HCl. The organic solution was washed with 5% aqueous HCl (2x), aqueous NaHC03 (1x) and brine (1x). The organic solution was filtered through a silica gel cartridge, eluting with CH2Cl2. A recrystallization from hexanes (60 ml) provided 22.47 g of 2-bromo-N-isopropyl-N-phenyl-acefamide. H1 NMR (CD2Cl2) d 7.45 (m, 3H), 7.21 (m, 2H), 4.89 (m, 1H), 3.52 (s, 2H), 1.05 (d, 6H); MS 256.2 (M + 1). Preparation 1 (B) Preparation of 2-Bromo-N-isopropyl-N- (6-mephoxy-pyridine-3-D-acetamide) Meto 1: Step A: lsopropylidene- (6-methoxy-pyridin-3-yl) -amine To a solution of 5-amino-2-meioxypyridine (18 g, 161 mmol) in methanol (80 ml) was added acetone (20 ml). ml, 177 mmol). The reaction was refluxed for 24 hours and the volatile components were concentrated in vacuo to provide 21.3 g of isopropylidene- (6-meioxy-pyridin-3-yl) -amine. H1 NMR (CDCl3) d 7.59 (d, 1H), 7.31 (m, 1H), 6.70 (d, 1H), 3.91 (s, 3H), 2.20 (s, 3H) 1.85 (s, 3H). Step B: lsopropyl- (6-methoxy-pyridin-3-yl-amine) To a solution of isopropylidene- (4-meloxy-phenyl) -amine (21.3 g, 129 mmol) in a mixture of EtOH (100 mL) and MeOH (50 mL) at 0 ° C was added NaBH 4 (14.7 g, 389 mmol) in 3 parts. The reaction was stirred at ambient temperature for 3 hours and diluted with water. The aqueous layer was washed with ElOAc (3x) and the combined organic extracts were dried (MgSO4), filtered and concentrated. The residue was purified by medium pressure chromatography eluting with a gradient of solvents (2% MeOH in CH 2 Cl 2 to 12% MeOH in CH 2 Cl 2) to provide 11.26 g of isopropyl- (6-methoxy-pyridin-3-yl) -amine. H1 NMR (CDCl3) d 7.55 (d, 1H), 6.96 (dd, 1H), 6.61 (d, 1H), 3.86 (s, 3H), 3.52 (m, 1H) , 1.19 (d, 6H). Method 2: Step A: lsopropyl- (6-methoxy-pyridin-3-iP-amine) To a solution of 5-amino-2-methoxypyridine (747 mg, 6.02 mmol) in CH2Cl2 (50 mL) was added acetone ( 500 μl) and sodium triacetoxyborohydride (1.95 g, 9.20 mmol). The reaction was stirred at ambient temperature for 20 hours and diluted with aqueous NaHCO3. The aqueous solution was washed with CH2Cl2 (3x) and the combined organic solutions were washed with brine (1x), dried (MgSO4), filtered and concentrated. The residue was purified by medium pressure chromatography eluting with a gradient of solvents (5% EtOAc in hexanes to 50% EtOAc in hexanes) to provide 810 mg of isopropyl- (6-methoxy-pyridin-3-yl) -am. Na H1 NMR (CDCl3) d 7.55 (s, 11-1), 6.96 (dd, 1H), 6.61 (d, 1H), 3.86 (s, 3H), 3.52 (m, 1H), 1.19 (d, 6H). Step B: 2-Bromo-N-isopropyl-N- (6-methoxy-pyridin-3-yl) -acetemide To a solution of isopropyl- (6-meioxy-pyridin-3-yl) -amine (6, 73 g, 41.0 mmol) in CH2Cl2 (130 ml) at 0 ° C was added diisopropylethylamine (7.15 ml, 41.0 mmol) followed by bromoacetyl bromide (8.28 g, 41.0 mmol) in CH2Cl2 (60 ml ) for 0.5 hours. The reaction was stirred at ambient temperature for 24 hours and diluted with water. The organic solution was washed with brine (1x), dried (MgSO 4), filtered and concentrated. The residue was purified by medium pressure chromatography eluting with a gradient of solvents (1% MeOH in CH 2 Cl 2 to 5% MeOH in CH 2 Cl 2) to provide 2.27 g of 2-bromo-N-isopril-N- (6- mexixy-pyridin-3-yl) -acetamide. H1 NMR (CD3OD) δ 8.08 (d, 1H), 7.62 (dd, 1H), 6.139 (d, 1H), 4.84 (m, 1H), 3.9 (s, 3H), 3 , 60 (s, 2H), 1, 06 (m, 6H). Preparation KC) Preparation of N-Benzyl-2-bromo-N-isopropyl-acetylamide. To a solution of N-isopropylbenzylamine (40 ml, 240 mmol) in CHCl 3 (300 ml) was added 3N (36.8 ml). The reaction was cooled to 0 ° C and bromoacetyl bromide (21.8 mL, 251 mmol) was added. The mixture was stirred at room temperature for 24 hours and diluted with CH2Cl2. The organic solution was washed consecutively with 5% aqueous HCl (1x) and aqueous NaHC 3 (1x). The organic solution was dried (Na 2 S 4), filtered and concentrated. The residue was dissolved in 40% EtOAc in hexanes and filtered through a silica gel cartridge to provide 52.2 g of N-benzyl-2-bromo-N-isopropyl acetamide. H1 NMR (CD2CI2) d 7.20-7.40 (m, 5H), 4.53 (d, 2H), 4.22 (m) and 4.74 (m, total 1H), 4.01 (s) , 1H), 3.72 (s, 1H), 1.20 (d, 3H), 1.12 (d, 3H). Preparation 1 (D) Preparation of 2-Bromo-N- (4-fluoro-phen-pN-isopropyl-acetamide) Following the procedure described for Preparation 1 (A), it was alkylated (4-fluoro-phenyl) -isopropyl-amine (13.70 g, 89.54 mmol) with bromoacetyl bromide (7.78 mL, 89.54 mmol) to provide 15.02 g of 2-bromo-N- (4-fluoro-phenyl) -N-isopropyl. -acetamide in the form of an oil H1 NMR (CD2CI2) d 7.14-7.23 (m, 4H), 4.90 (m, 1H), 3.53 (s, 2H), 1.05 (d , 6H).
Preparation 2 (A) Preparation of (6-Chloro-pyridin-3-P-isopropyl-amine To a solution of 2-chloro-5-aminopyridine (500 mg, 3.88 mmol) and acetone (250 μL, 4.27 mmol) in dichloroethane (13 mL) was added NaBH (OAc) 3 (989 mg, 4, 66 mmol) and AcOH (330 μl, 5.82 mmol). The reaction was stirred for 24 hours and diluted with 1N NaOH. The aqueous solution was washed with CH 2 Cl 2 (3x). The combined organic solutions were dried (Na 2 SO 4), filtered and concentrated. Purification by medium pressure chromatography eluting with a gradient of solvents (2% MeOH in CH 2 Cl 2 to 5% MeOH in CH 2 Cl 2) gave 505.2 mg of (6-chloro-pyridin-3-yl) -isopropyl- amine. H1 NMR (CD3OD) d 7.64 (d, 1H), 7.10 (d, 1H), 6.99 (dd, 1H), 3.55 (m, 1H), 1.17 (d, 6H) . Preparation 2 (B) Preparation of 6-Ethoxy-pyridin-3-yl) -isopropyl-amine * ~ Following the procedure described for Preparation 2 (A), 6-eioxy-pyridin-3-ylamine (10 g, 72.4 mmol) was reacted with aceilone (4.8 mL) to provide 11.18 g of (6 mL). -ethoxy-pyridin-3-yl) -isopropyl-amine. H1 NMR (CD3OD) d 7.48 (d, 1H), 7.11 (dd, 1H), 6.61 (d, 1H), 4.14 (q, 2H), 3.48 (m, 1H), 1 , 52 (1, 3H), 1, 14 (d, 6 H); MS 181.3 (M + 1). Preparation 3 (A) Preparation of 4-Eioxy-1,3-dihydro-benzo [b1H .41-diazepin-2-one To a solution of phenylenediamine (17.2 g, 159.2 mmol) and acetic acid (0.4 ml) in xylenes (225 ml) at 140 ° C was added 3,3-diephoxyacrylic acid erythyl ester (30.0 g, 159.2 mmol) in xylenes (80 ml) was to be enjoyed during 50 minutes The reaction was heated at 140 ° C for 2 hours, cooled to room temperature and stirred for 24 h. The resulting soft precipitate was filtered, washed with ether (100 ml) and dried under vacuum to provide 18.37 g of 4-ethoxy-1,3-dihydro-benzo [b] [1,4] diazepin-2-one as a white fluffy solid. MS 205 (M + 1). Preparation 3 (B) Preparation of 4-Eioxy-7,8-difluoro-1,3-dihydro-benzofbjf 1,4] diazepin-2-one To a solution of 1,2-diamino-4,5-difluorobenzene (4.0 g, 27.75 mmol) in xylenes (50 ml) at 140 ° C was added acetic acid (200 μl) followed by an ester solution. erylyl of 3,3-diethyxyacrylic acid (5.22 g, 27.75 mmol) in xylenes (25 ml) for 1 hour. The reaction was heated at 140 ° C for 2 hours, cooled to room temperature and concentrated. The residue was triturated in hexanes to give 5.38 g of 4-ethoxy-7,8-d-fluoro-1,3-dihydrobenzo [b] [1,4] -diazepin-2-one. H1 NMR (CDCl3) d 7.05 (q, 1H), 6.87 (q, 1H), 4.29 (q, 2H), 3.18 (s, 21-1), 1.35 (t, 3H). Preparation 4 (A) Preparation of 1-Phenyl-4H.6H-2.3.6.10b-feiraza-benzofejazulen-5- To a solution of 4-ethoxy-1,3-dihydro-benzo [b] [1,4] d-azepin-2-one (Preparation 3 (A) (5 g, 24 mmol) in glacial AcOH (75 ml) Benzoic acid hydrazide (3.33 g, 24.5 mmol) was added The reaction mixture was heated at 120 ° C for 2 hours and cooled to room temperature The solvent was removed in vacuo by azeopyric distillation with hepyan ( 2x) The residue was dissolved in a minimum amount of methylene chloride and 50 ml of ethyl acetate and the solution was poured slowly with stirring into a solution of 100 ml of saturated NaHCO3 / 100 ml of water. ml) and the mixture was stirred for 5 minutes.The precipitate was collected by filtration, washed with water and the minimum amount of ether to provide 5.89 g of 1-phenyl-4H, 6H-2,3,6, 10b -leazaza-benzo [e] azulen-5-one, H1 NMR (CDCl3) d 9.02 (br s, 1H), 7.48-7.36 (m, 6H), 7.10 (i, 1H) 6.95 (d, 1H), 4.23 (d, 1H), 3.60 (d, 1H), MS 277 (M + 1).
Preparation 4 (B) Preparation of 1-Cyclohexyl-4H.6H-2,3,6.10b-teiraza-benzofejazulen-5-one To a solution of 4-eioxy-1,3-dihydro-benzo [b] [1,4] diazepin-2-one (Preparation 3 (A)) (5 g, 24.5 mmol) in glacial acetic acid ( 75 ml) was added cyclohexanecarboxylic acid hydrazide (Preparation 14) (3.5 g, 24.5 mmol). The reaction was heated at 120 ° C for 2.5 hours, cooled to ambient temperature and EOAc (50 ml), water (50 ml) and aqueous NaHC 3 (50 ml) were added. The mixture was stirred for 5 minutes and ET2O (125 ml) was added. The suspension was filtered for 15 minutes and the solids were removed by filtration with the aid of EIO2 to provide 5.92 g of 1-cyclohexyl-4H, 6H-2,3,6, 10b-telraza-benzo [e] azulene. 5-one. H1 NMR (CDCl3) d 8.36 (s, NH), 7.50 (m, 1?), 7.41 (m, 2H), 7.30 (d, 1H), 4.13 (d, 1H), 3.47 (d, 1H), 2.87 (m, 1H) ), 2.24 (d, 1H), 2.00 (m, 21-i), 1.72-1.51 (m, 4H), 1.33 (m, 2H), 1.16 (m, 1H); MS 283.4 (M + 1), 281.3 (M-1). Preparation 4 (C) Preparation of 8.9-Pfluoro-1-phenyl-4H.6H-2.3.6.10b-telraza-benzo [e] azulen-5-one To a solution of 4-eioxy-7,8-difluoro-1,3-dihydro-benzo [b] [1,4] diazepin-2-one (Preparation 3 (B)) (5.38 g, 22.4 mmol) in acetic acid (100 ml) was added benzoic hydrazide (3.11 g, 22.85 mmoi). The reaction was heated at 120 ° C for 18 hours and the volatile components were concentrated in vacuo. The residue was dissolved in CH2Cl2 and the organic solution was washed with aqueous NaHCOs (1x), dried Na2S4), filtered and concentrated. The solid residue was frittered with a warm mixture of EtOAc and hexanes and filtered. The solids were trilured in 50% hexanes in EIO2 and filtered to provide 3.97 g of 8,9-difluoro-1-phenyl-4H, 6H-2,3,6,10b-eeryza-benzo [e] azulen. -5-ona. H1 NMR (DMSO-de) d 7.36 (m, 6H), 7.06 (m, 1H), 3.91 (d, 1H), 3.76 (d, 1H); MS 313.1 (M + 1), 311.1 (M-1). Preparation 5 (A) Preparation of 2- (4-Ethoxy-2-oxo-2,3-dihydro-benzofb] [1,41-diazepin-1-yl) -N-isopropyl-N- (6-meioxy-pyrid) n-3-yl) -acetamide To a solution of 4-eioxy-1,3-dihydro-benzo [b] [1,4] diazepin-2-one (Preparation 3 (A)) (2 g, 9.79 mmol) in DMF (40 mL) at 0 ° C NaH (60% in oil, 431 mg, 10.8 mmol) was added. The reaction was stirred at 0 ° C for 0.5 hour and a solution of 2-bromo-N-isopropyl-N- (6-methoxy-pyridin-3-yl) -acelamide (Preparation 1 (B)) ( 2.95 g, 10.28 mmol) in DMF (5 ml). The reaction was stirred at ambient temperature for 2 hours and diluted with water, brine and CH2Cl2. The aqueous solution was washed with CH2Cl2 (3x) and the combined organic components were washed with brine, dried (MgSO4), filtered and concentrated in vacuo. Purification by medium pressure chromatography eluting with a gradient of solvents (hexanes to 70% EtOAc in hexanes) yielded 2.87 g of 2- (4-ethoxy-2-oxo-2,3-dihydro-benzo [b] [1,4] d.azepin-1-yl) -N-isopropyl-N- (6-meioxy-pyridin-3-yl) -acetamide. H1 NMR (CDCl3) d 8.14 (s) and 7.93 (s, total 1H), 7.54 (m, 1H), 7.31 (m, 1H), 7.24-7.08 (m , 3H), 6.80 (m, 1H), 5.05 (m, 1H), 4.29 (m, 2H), 4.09 (m, 1H), 3.95 (m, 3H), 3 , 67 (d, 1H), 3.31 (d, 1H), 3.08 (d, 1H), 1.32 (m, 3H), 1.09 (m, 6H). Preparation 5 (B) Preparation of 2- (4-Eioxy-2-oxo-2,3-dihydro-benzofb] | '1,4] diazepin-1-iO-N-isopropyl-N-phenyl-acetamide To a solution of 4-ethoxy-1,3-dihydro-benzo [b] [1,4] diazepin-2-one (Preparation 3 (A) (5.0 g, 24.5 mmol) in DMF (100 ml) was added potassium bistrimethylsilyl amide (49 ml of 0.5 M solution in toluene, 24.5 mmol) 0 ° C. The reaction mixture was stirred for 20 minutes and cooled to -17 ° C. A solution of 2-bromo-N-isopropyl-N-phenyl-acetamide (Preparation 1 (A)) (6, 9 g, 27 mmol) in DMF (50 ml) dropwise so that the internal temperature remained below -15 ° C. The reaction mixture was stirred at -16 ° C for 1 hour, was boosted at ambient temperature and was diluted with ethyl acetate.The organic phase was washed with brine, dried over sodium sulfate and He concentrated. The resulting pale brown solid was rinsed with ether to obtain 3.2 g of 2- (4-ethoxy-2-oxo-2,3-dihydro-benzo [b] [1,4] diazepin-1-yl) -N -isopropyl-N-phenyl-acetamide in the form of a white powder. The filtrate was concentrated and the residue was triturated with ether / hexanes. The solid was collected by fill-up to provide a second crop of 2- (4-eioxy-2-oxo-2,3-dihydro-benzo [b] [1,4] -diazepin-1-yl) -N-isopropyl- N-phenyl-acetylamide (3.4 g). Preparation 6 Preparation of 2- [1- (3-Benzyloxy-phenyl) -5-oxo-4,5-dihydro-2.3.6.10b-yl-aza-benzo [e] azulen-6-yl] -N-isopropyl-N- ( 6-meioxy-pyridin-3-yl) -acetamide Following the procedure described for Preparation 4 (B), 2- (4-ethoxy-2-oxo-2,3-dihydro-benzo [b] [1,4] diazepin-1-yl) -N- was reacted isopropyl-N- (6-methoxy-pyridin-3-yl) -acetamide (Preparation 5 (A) (150 mg, 0.365 mmol) with 3-benzyloxybenzylhydrazide (88 mg, 0.365 mmol) to give 129 mg of 2- [1- (3-benzyloxy-phenyl) -5-oxo-4,5-dihydro-2,3,6,10b-tetraaza-benzo [e] azulen-6-yl] -N-isopropyl-N- (6 -methoxy-pyridin-3-yl) -acetamide, H1 NMR (CD3OD) d 8.06 (d, 1H), 7.59 (m, 2H), 7.51 (t, 1H), 7.40-7 , 28 (m, 8H), 7.19 (t, 1H), 7.13 (d, 1H), 6.97 (d, 1H), 6.91 (d, 1H), 5.01 (d, 1H), 4.91 (d, 1H), 4.76 (m, 1H), 4.60 (m, 1H), 4.12 (m, 1H), 3.95 (s, 4H), 3, 75 (d, 1H), 0.91 (m, 6H); MS 89.8 (M + 1), 587.5 (M-1).
Preparation 7 (A) Preparation of N-lsopropyl-2- (5-oxo-1-phenyl-4,5-dihydro-2,3,6, 10b-ieirazazabenzof31azulen-6-yl) -N-phenyl-acefamide To a solution of 2- (4-eioxy-2-oxo-2,3-dihydro-benzo [b] [1,4] diazepin-1-yl) -N-isopropyl-N-phenyl-acetamide (Preparation 5 ( B)) (71.14 g, 0.187 mol) in 1.04 I of AcOH was added benzoic acid hydrazide (27.28 g, 0.196 mol) in one portion. The reaction was heated to 80 ° C and stirred for 4 hours. The reaction was cooled to room temperature and the AcOH was removed in vacuo to give an off white solid. The solid was dissolved in 1 l of methylene chloride. The organic solution was washed with 1 l of saline NaHCO3 solution followed by brine and dried over Na2SO4. The methylene chloride solution was diluted with an equal volume of methyl-urea-builder-ether and the solution was concentrated under vacuum for a low volume causing a white solid to precipitate. The precipitate was collected in a sintered glass funnel and rinsed with methylene-tert-butyl ether. The solid was dried under vacuum to provide 81, 41 g of N-isopropyl-2- (5-oxo-1-phenyl-4,5-dihydro-2,3,6, 10b-tetraaza-benzo [e] azulen-6-yl) -N-phenyl- acefamide in the form of a white solid. H1 NMR (CD2CI2) d 7.57-7.35 (m, 10H), 7.27 (br s, 1H), 7.21 (br s, 1H), 7.10 (l, IH), 6.87 (d, 1H), 4.89 (m, 1H), 4.19-3.99 ( m, 3H), 3.53 (d, 1H), 1.03 (m, 6H); MS 452.3 (M + 1), 450.5 (M-1). Preparation 7 B) Preparation of 2- (1- (2-fluorophenin-5-oxo-4,5-dihydro-2.3.6.10b-ieiraazabenzo | e] azulen-6-iD-N-isopropyl-N-phenyl- aceiamide Following the procedure described in Preparation 7 (A), 2- (4-eloxi-2-oxo-2,3-dihydro-benzo [b] [1,4] diazepin-1-yl) -N is reacted - soproyl-N-phenyl-acelamide (Preparation 5 (B) (200 mg, 0.527 mmol) with 2-fluorobenzohydrazide (81 mg, 0.527 mmol) Purification by medium pressure chromatography eluting with a solvent gradient (CH2Cl2) 3% MeOH in CH 2 Cl 2) gave 180.2 mg of 2- (1- (2-fluorophenyl) -5-oxo-4,5-dihydro-2,3,6,10b-teiraza-benzo [e] azulen- 6-yl) -N-isopropyl-N-phenyl-acetamide H1 NMR (CD2CI2) d 7.73 (m, 1), 7.64 (d, 1), 7.91-7.18 (m, 8 ), 7.12-7.00 (m, 2), 6.83 (d, 1), 4.96 (m, 1), 4.23 (d, 1), 4.06 (d, 1) 3.80 (d, 1), 3.58 (d.1), 1.09 (d.6), MS 470.3 (M + 1) Preparation 8 A) Preparation of 2- (1-Cyclohexyl) -5-oxo-4,5-dihydro-2.3.6.10b-teiraza-benzofe1azulen-6-yl) -N-isopropyl-N- (6-mephoxy-pyridin-3) -iO-acetamide To a solution of 1-cyclohexyl-4H, 6H-2,3,6,10b-tetraaza-benzo [e] azulen-5-one (Preparation 4 (B)) (100 mg, 0.354 mmol) in DMF (10 ml ) at 0 ° C NaH (60% in oil, 15 mg, 0.372 mmol) was added. The reaction was stirred at 0 ° C for 30 minutes, cooled to -10 ° C and 2-bromo-N-isopropyl-N- (6-meioxy-pyridin-3-yl) -acetamide ( Preparation 1 (B)) (107 mg, 0.372 mmol) in DMF (1 ml). The reaction was stirred at ambient lemperauria for 24 h and diluted with water. The aqueous solution was washed with EtOAc (3x). The combined organic solutions were dried (MgSO 4), filtered and concentrated. The residue was purified by medium pressure chromatography eluting with a gradient of solvents (1% ElOAc in hexanes to 100% ElOAc) to provide 861.8 mg of 2- (1-cyclohexyl-5-oxo-4,5- dihydro-2,3,6, 10b-telraza-benzo [e] azulen-6-yl) -N-isopropyl-N- (6-methoxy-pyridin-3-yl) -acetamide. H1 NMR (CDCl3) d 8.11-7.48 (m, total 3H), 7.36 (m, 3H), 6.79 (dd, 1H), 4.99 (m, 1H), 4.12 (m, 2H), 3.92 (d, 3H), 3.65 (d) and 3.82 (d, total 1H), 3.46 (dd, 1H), 2.83 (m, 1H), 2.20 (d, 1H), 2.03-1.86 (m, 3H), 1.68 (m, 2H), 1.54 (m, 2H) 1.31-1.41 (m, 2H) ), 1.07 (m, 6H); MS 489.4 (M + 1), 487.4 (M-1). Preparation 8 (B) Preparation of 2- (1-cyclohexyl-5-oxo-4,5-dihydro-2.3.6.10b-tetraaza-benzofe] azulen-6-yl) -N-isopropyl-N-phenyl-acetamide Following the procedure described for Preparation 8 (A), 1-cyclohexyl-4H, 6H-2,3,6,10b-tetraaza-benzo [e] azulen-5-one was alkylated (Preparation 4 (B)) (640 mg, 0.266 mmol) with 2-bromo-N-isopropyl-N-phenyl-acetamide (Preparation 1 (A)) (580 mg, 0.226 mmol). Purification by means of medium pressure chromatography eluting with a gradient of solvents (CH 2 Cl 2 to 4% MeOH in CH 2 Cl 2) gave 668 mg of 2- (1-cyclohexyl-5- oxo-4,5-dihydro-2,3,6,10b-tetraaza-benzo [e] azulen-6-yl) -N-isopropyl-N-phenyl-acetamide. H1 NMR (CDCl3) d 7.62-7.31 (m, 9), 4.93 (m, 1), 4.10 (d, 1). 3.91 (d, 1), 3.76 (d, 1), 3.45 (d, 1), 2.85 (m, 1), 2.19 (br.d, 1), 1.91 (m, 2), 1.69-1.11 (m, 7), 1.06 (m, 6): MS 458.4 (M + 1). Preparation 9 Preparation of N-Benzyl-2- (8,9-difluoro-5-oxo-1-phenyl-4,5-dihydro-2,3,6, 10b-tetraaza-benzofejazulen-6-ip-N-isopropyl-acetamide To a solution of KHMDS (0.5 M in THF, 16.6 mL, 8.33 mmol) at 0 ° C was added 8,9-difluoro-1-phenyl-4H, 6H-2,3,6,10b -tetraaza-benzo [e] azulen-5-one (Preparation 4 (C)) (2.0 g, 6.4 mmol) in DMF (20 ml). The reaction was stirred at 0 ° C for 35 minutes, cooled to -10 ° C and a solution of N-benzyl-2-bromo-N-isopropyl-acetamide (Preparation 1 (C)) (1.9 g) was added. 7.0 mmol) in DMF (20 ml). The reaction was stirred at -10 ° C for 2 hours and at room temperature for 24 hours. The reaction was quenched with a pH 6.8 buffer and the aqueous solution was washed with EtOAc (3x). The combined organic solutions were washed with brine (4x), dried (Na2SO4), filtered and concentrated. Purification by means of medium pressure chromatography eluting with a gradient of solvents (CH2CL2 to 3% MeOH in CH2Cl2) gave 2.01 g of N-benzyl-2- (8,9-difluoro-5-oxo-1-phenyl- 4,5-dihydro- 2,3,6,10b-tetraazabenzo [e] azulen-6-yl) -N-isopropyl-acetamide. H1 NMR (CD2Cl2) d 7.60-7.17 (m, 11H), 6.75 (q, 1H), 4.85-4.38 (m, 4H), 4.10 (q, 1H), 3.60 (q, 1H), 3.41 (s, 1H), 1, 21 (q, 3H), 1.13 (q, 3H); MS 502.4 (M + 1), 500.3 (M-1). Preparation 10 Preparation of N-Benzyl-N-isopropyl-2- (5-oxo-1-phenyl-4,5-dihydro-2,3,6,10-tetraaza-benzo [e] azulen-6-yl) -acetamide To a solution of 1-phenyl-4H, 6H-2,3,6,10b-tetraaza-benzo [e] azulen-5-one (Preparation 4 (A)) (30.0 g, 108 mmol) in DMF ( 200 ml) at 0 ° C was added NaHMDS (1.0 M in THF, 13.6 g, 118 mmol) and N-benzyl-2-bromo-N-isopropyl-acetamide (Preparation 1 (A)) (35, 2 g, 130 mmol) in DMF (25 ml). The reaction was stirred at room temperature for 24 hours and diluted with a phosphate buffer at pH 6.8. The aqueous solution was washed with EtOAc (3x). The combined organic solutions were washed with brine (3x), dried (Na 2 S 4), filtered and concentrated. Purification by medium pressure chromatography eluting with a gradient of solvents (CH2Cl2 at 12% acetone in CH2Cl2) gave 13.9 g of N-benzyl-N-isopropyl-2- (5-oxo-1-phenyl-4, 5-dihydro-2,3,6, 10b-teiraza-benzo [e] azulen-6-yl) -acetemide. H1 NMR (CDCl3) d 7.64-7.55 (m, 2H), 7.44-7.08 (ni, 11H), 6.89 (m, 1H), 4.96-4.08 (m , 6H), 3.57 (m, 1H), 1.17 (m, 6H).
Preparation 11 Preparation of N-lsopropyl-N- (6-methoxy-pyridin-3-iD-2- (5-oxo-1-phenyl-4,5-dithia-2,3,6-di-1-aza-a) benzore1azulen-6-ip-aceiamida To a solution of 1-phenyl-4H, 6H-2,3,6,10b-ielraza-benzo [e] azulen-5-one (Preparation 4 (A)) (6.05 g, 21.9 mmol) in DMF (120 ml) at 0 ° C was added NaH (60% in oil, 920 mg, 23.0 mmol). The reaction was stirred at 0 ° C for 30 min. And isopropyl- (6-methoxy-pyridin-3-yl) -amine (Preparation 1 (B)) (6.21 g, 23.0 mmol) was added. The reaction was stirred at ambient temperature for 24 hours and diluted with water. The aqueous solution was washed with EOAc (3x). The combined organic solutions were washed with water, dried (MgSO4), filtered and concentrated. Purification by medium pressure chromatography eluting with a gradient of solvents (2% MeOH in CH 2 Cl 2 to 8% MeOH in CH 2 Cl 2) afforded 8.16 g of N-isopropyl-N- (6-methoxy-pyridin-3-yl) ) -2- (5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-leraza-benzo [e] azulen-6-yl) -acetamide. H1 NMR (CD3OD) d 8.10 (m, 1H), 7.62 (m, 4H), 7.52 (t, 2H), 7.43 (t, 2H), 7.19 (t, 1H) , 6.94 (t, 2H), 4.82 (m, 1H), 4.52 (d, 1H), 4.16 (m, 1H), 3.96 (m, 4H), 3.79 ( d, 1H), 1.02 (m, 6H); MS 483.3 (M + 1), 481.1 (M-1).
Preparation 12 Preparation of 2-f1- (3-Hydroxy-phenan-5-oxo-4,5-dihydro-2.3.6.10b-tetraazabenzo [e1azulen-6-yl] -N-isopropyl-N-phenyl) -acetemida To a solution of 2- (4-ethoxy-2-oxo-2,3-dihydro-benzo [b] [1,4] diazepin-1-yl) -N-isopropyl-N-phenyl-acefamide (Preparation 5 ( B)) (177 mg, 0.466 mmol) in glacial acetic acid (4 ml) was added 3-hydroxy-benzohydrazide (90 mg, 0.591 mmol). The reaction was heated to 120 ° C for 3 hours, cooled to room temperature and concentrated in vacuo. The residue was trifuged with 50% EIO20 in hexanes and the solids filtered. The solids were dissolved in CH2CI2 and the organic solution was washed with aqueous NaHCO3, dried (MgSO4) and concentrated to provide 244 mg of 2- [1- (3-hydroxy-phenyl) -5-oxo-4, 5-dihydro-2,3,6,10b-telraazabenzo [e] azulen-6-yl] -N-isopropyl-N-phenyl-acetamide. H1 NMR (CD3OD) d 7.74 (s, 2H), 7.56-7.18 (m, 10H), 6.91 (m, 1H), 4.86 (m, 1H), 4.30- 4.10 (m, 2H), 3.98 (d, 1H), 3.71 (d, 1H), 1.06 (m, 6H). Preparation 13 Preparation of 5-Oxo-1-phenyl-4,5-dihydro-2,3,6.10b-terazaza-benzo [e] azulen-6-yl-acetic acid eerc-buiyl ester Following the procedure described for Preparation 13 (A), 1-phenyl-4H, 6H-2,3,6,10b-tetraaza-benzo [e] azulen-5-one was alkylated (Preparation 4 (A)) (5). , 0 g, 18.1 mmol) with urea-buíilo bromoacety (2.94 mL, 19.9 mmol). A solution with Ef O (100 ml), hexanes (30 ml) and EOAc (10 ml) yielded 3.13 g of tert-buílic acid ester (5-oxo-1-phenyl-4,5-dihydro-2, 3,6, 10b-tetraaza-benzo [e] azulen-6-yl) -acetic. H1 NMR (CD3OD) d 7.68 (d, 1H), 7.60-7.51 (m, 4H), 7.43 (m, 2H), 7.23 (t, 1H), 6.99 ( d, 1H), 4.86 (d, 1H), 4.44 (d, 1H), 3.99 (d, 1H), 3.83 (d, 1H), 1.36 (s, 9H); MS 391.4 (M + 1), 389.3 (M-1). Preparation 14 Preparation of cyclohexanecarboxylic acid hydrazide To a solution of methylcyclohexane carboxylate (12 g, 83.9 mmol) in MeOH. (50 ml) was added hydrazine (5.3 ml, 1.67 moles). The reaction mixture was heated at 65 ° C overnight. The reaction mixture was cooled to ambient temperature and the resulting solid was collected by filtration and dried under vacuum to provide 4.0 g of cyclohexanecarboxylic acid hydrazide. Preparation 15 Preparation of acid [4- (1H-lndol-3-ylmethyl-5-oxo-1-phenyl-4,5-dihydro-2.3.6.10b-iefraaza-benzo | 'e1azulen-6-yl-acéíico Eíapa A : F4- (1H-lndol-3-ylmeiylene) -5-oxo-1-phenyl-4,5-dihydro-2,3,6, 10b-eeryaraza-benzofejazulen-6-yl-acetic Acidic acid ester To a solution of tert-butyl ester of (5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-teiraza-benzo [e] azulen-6-yl) -acetic acid (Preparation 13 (B)) (3.66 g, 9.37 mmol) in íoluene (94 mL) was added 1 H, ndol-3-carbaldehyde (1.63 g, 11.2 mmol) and piperidine (2.78 mL, 28.1 mmol). The reaction was heated to 110 ° C in a Soxhlei device for 10 hours and stirred at ambient temperature for 24 hours. The precipitate was filtered and washed with toluene to provide 6.47 g of [4- (1 H-indole-3-methylmethylene) -5-oxo-1-phenyl-4,5-dihydro-4-oxo-1-pyridine-3-hydroxy-3-hydroxyethyl ester. 2,3,6,10b-tetraaza-benzo [e] azulen-6-yl-acetic acid. MS 518.5 (M + 1). Step B: f4- (1 H-lndol-3-ylmef-5-oxo-1-phenyl-4,5-dihydro-2.3.6.10b-eierazazabenzore] azulen-6-α-acylic acid ions. Following the procedure described for Example 1 (A), Step B, 4- (1H-lndol-3-ylmeiylene) -5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-leiraza-benzo [e] acid tertiary-bufl ester was reduced azulen-6-yl] -acetic acid (4.84 g, 9.37 mmol) in EOH (150 ml) lasted 3 hours at 80 ° C. The residue was dissolved in EtOAc and washed with aqueous NH 4 Cl (1x) and brine (1x). The organic solution was dried (Na2SO4), filtered and concentrated to provide 4.26 g of [4- (1H-indol-3-ylmethyl) -5-oxo-1-phenyl-4-tert-butyl ester. , 5-dihydro-2,3,6,10b-ioraza-benzo [e] azulen-6-yl] -acelic. H1 NMR (DMSO-de) d 7.65 (d, 1H), 7.53-7.38 (m, 7H), 7.27 (m, 1H), 7.18 (m, 2H), 6, 98 (m, 1H), 6.89 (m, 2H), 4.75 (d, 1H), 4.43 (d, 1H), 3.82 (1, 1H), 3.59 (m, 2H) ), 1.22 (s, 9H). Step C: f4- (lH-lndol-3-ylmein-5-oxo-1-phenyl-4,5-dihydro- 2.3.6.1 Qb-ieirazazabenzofe'lazulen-6-yl-acetic acid To a solution of urea-buffyl ester of [4- (1H-Indsl-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-tetraaza-benzo [e] azulen-6-yl] -acetic acid (4.26 g, 8.19 mmol) in CH2Cl2 (27 mL) was added TFA (9.5 mL, 0.123 mmol). The reaction was stirred at ambient temperature for 8 hours and concentrated in vacuo. The residue was dissolved in CH2Cl2 (50 ml) and Et2? (15 ml) and stirred for 24 hours. The precipitate was filtered and washed with Et2Ü to provide 2.33 g of [4- (1H-indol-3-ylmethyl) -5-oxo-1-phenyl-4,5-d-hydro-2,3 acid, 6,10b-tetraaza-benzo [e] azulen-6-yl] -acetic. H1 NMR (DMSO-d6) d 10.79 (s, 1H), 7.70 (d, 1H), 7.54-7.38 (m, 7H), 7.26 (d, 1H), 7, 17 (m, 2H), 6.98 (t, 1H), 6.88 (m, 2H), 4.90 (d, 1H), 4.49 (d, 1 1-0, 3.82 (i , 1H), 3.59 (m, 2H) BIOLOGICAL TESTS The usefulness of the compounds of the present invention as pharmaceutically active agents in the treatment of metabolic diseases (such as those mentioned above) in animals, particularly mammals (e.g. human beings) is demonstrated by the activity of the compounds of the present invention in conventional assays and in the in vitro and in vivo assays described below.These tests also provide a means by which the activities of the compounds of the invention can be compared. present invention with the activities of known compounds The results of these comparisons are useful for determining the dosage levels The compounds of Examples 1 (A) -7 above were tested in the uni assay. n of CCK receptor described later, and the IC50 values for these compounds were found to be in the range of about 10 nM to about 125 nM under the conditions of that essay. The compounds of the above Examples 1 (A) -7 were also tested in the functional assay of the CCK receptor described below, and the EC50 values for the functional CCK-A agonism for these compounds were found to be in the range of about 50 nM to about 1,000 nM under the conditions of that assay. COLECISTOQUININE RECEIVER UNION ASSAY (CCK) To determine binding affinity, the compounds were tested using membranes prepared from CHO cells that were stably transfected and expressing human or rat CCK-A receptor. The cell membranes were prepared from a T-75 cell of cells by pelleting cells at 1,000 x g at 4 ° C for 5 minutes and resuspending them in 1 ml of homogenization buffer (1 mM EDTA)., 1 mM EGTA, 1 mM sodium bicarbonate pH 7.4, 100 μg / ml benzamidine ,. 100 μg / ml bacitracin, 5 μg / ml leupepin and 5 μg / ml aprotinin). After shaking on ice for 10 minutes, the cells were homogenized with. a Dounce homogenizer. Nuclei and non-lysar cells were separated by centrifugation at 1,000 x g at 4 ° C for 10 minutes. The supernatant was transferred to a new tube and then centrifuged at 25,000 g at 4 ° C for 20 minutes. The pellet was resuspended in 5 ml of binding buffer (20 mM HEPES, pH 7.4, 5 mM MgCl 2, 118 mM NaCl, 5 mM KCl, 1 mM EGTA, 100 μg / ml benzamidine, 100 μg / ml of bacitracin, 5 μg / ml of leupeptin and μg / ml aproininin). Protein concentration was determined using a BCA protein determination assay (Pierce). The binding assay was performed in a 96-well form using 5 μg (rat CCK-A receptors) or 10 μg (human CCK-A receptors) of membranes in 200 μl of binding buffer (described above) per well. Unlabeled CCK-8 (Sigma) or compounds were diluted in binding buffer and 25 μl of each was added to the assay plate so that its final concentration range was 10 μM to 0.01 nM. [125] -CCK8 (Amersham) was diluted to 0.75 nM in the binding buffer and 25 μl was added to each well (the final concentration was 75 pM). The assay plates were incubated at 30 ° C for 75 min. With gentle agitation. Nonspecific counts were suppressed using a Packard Fillermaf harvester (Packard 96-well Unfilter plate with GF / C membrane previously wetted in 0.3% PEI) and washed with cold wash buffer (20 mM HEPES, 5 mM MgCl 2). , 118 mM NaCl, 5 mM KCl, 1 mM EGTA, pH 7.4). After drying, the plates were counted by means of the Trilux 1450 Microbeta device from Wallac. The data was analyzed using a GraphPad Prism computer program to determine IC50 values. FUNCTIONAL TEST OF CCK RECEIVER To determine the functional agonist activity of the compounds, calcium mobilization was measured by FLIPR (Fluorometry Imaging Plate Reader, Molecular Devices Dorporaion, Sunnyvale, CA) in CHO cells stably expressing receptors.
CCK-A human or rat. In a 384-well white / clear poly-D-lysine culture plate, 15,000 cells were placed in 50 μl of medium per well and grown at 37 ° C, 5% CO2 for 24 hours. After removing the media, the cells were inoculated with 30 μl per well of Flú-4 cell loading dye (Molecular Probes, Eugene OR) in FLIPR phantom sterilized by filtration (50% cell-mediated medium, 50% solution of balanced Hank's salts, 20 mM HEPES, pH 7.4, 1 mM CaCb) with 0.64 mg / ml probenecid (Sigma), according to manufacturer's instructions. The cells were incubated for one hour at 37 ° C, 5% CO2. Drug plaques containing 50 μl of CCK-8 (Sigma) or compounds diluted in FLIPR buffer were assembled. Then, 15 μl of each compound was added to the assay plates so that the final concentration range was 100 μM to 0.01 nM after the FLIPR analysis. The EC50 values were determined using a GraphPad Prism computer program. FOOD INTAKE Sprague-Dawley male rats (274-325 grams) were acclimated to a self-ingested food intake and a system for evaluating locomotor activity during the night. Damages of food weight and locomotive activity were collected by computer acquisition at 10-minute intervals. Immediately before starting the dark cycle on the second day, the rats (n = 5-7 / group) were given a vehicle PO or IP dose (propylene glycol, 1 ml / kg + solution). saline, 8 ml / kg) or test compound (1-12 mg / kg in 1 ml / kg of propylene glycol + 8 ml / kg of saline) The food intake was checked until the next day. treatment were compared using a t-test in pairs to determine the statistical significance between groups.
BILIARY MOVEMENT TEST OF MOUSE BILIARY MOUSE Male mice C57BI / 6J, approximately 8 weeks old, were fasted for 18 hours and then administered vehicle orally (0.5% methylcellulose / 0.1% Tween 80) or compound. Was an ethylene / propylene glycol / H2 vehicle used? in a ratio of 2: 3: 5 for an inlraperiíoneal (i.p.) compound administration. A dose volume of 5 μl / g body weight in an oral administration and 1 ml / kg in an i.p. administration was used. After 30 minutes, the roots (n = 5 / lrafamienlo) were sacrificed by cervical dislocation and the biliary vesicles were removed and weighed. The ED50 values for the emptying of the gallbladder were determined by means of Graphpad Prism. all the groups of traiamaienío were arranged at random using the program www.randomizaiion.com.

Claims (15)

    CLAIMS 1.- A compound of formula (I) ) wherein A, B, X, D, E and G are independently -C (R5) - or -N-; with the proviso that not more than two of A, B, X and D are N at the same time and at least one of E and G is N; R1 is selected from the group consisting of (C2-C6) alkyl, halo-substituted alkyl (Ci-Cß), alkylamino (Ci-Cβ), di (C alqu-C6 alkyl) amino, alkylamino (C ?Ce) -alkyl (Ci-Cß), di-alkylamino (Ci-CßJ-alkyl (d-Cß), aryl, aryl-alkyl (Ci-Cß), heteroaryl-A, heteroaryl-A-alkyl (Ci-Cß), a partially or fully saturated heterocyclyl-A-4 to 7-membered heterocyclyl-A-alkyl (C-Cß) of 4 to 7 members, partially or completely saturated, and a (C3-C) cycloalkyl-alkyl (Ci-Cß) partially or completely saturated and, when none of R6 and R7 is phenylmethyl, R1 is selected from said group and partially (or completely) cycloalkyl (C3-C) in which heteroaryl-A is selected from the group consisting of thienyl, thiazolyl, isothiazolyl, indolyl , 2-pyridyl, pyridazinyl, pyrimidyl, oxazolyl, furanyl, imidazolyl, isoxazolyl, quinolinyl, isoquinollyl, benzothiophenyl, benzofuranyl, plrazinyl and pyrazolyl, and the partially or fully saturated heterocycle-A is selected from the group consisting of pyranyl, morpholinyl and teirahydrofuranyl, and wherein the aryl, heleroaryl-A , partial or fully saturated helerocycle-A or the partially or fully saturated cycloalkyl group or part of a group is optionally susliluted with 1 to 3 subsides independently selected from the group consisting of halo, (C1-C3) alkoxy, C1- (C1-) alkoxy C3) halo-substituted, -OH, (C1-C3) alkyl, -CN and halo-substituted alkyl (C1-C3); R2 is -CH2C (O) N (R6) (R7); one of R3 and R4 is H, halo, (C? -C6) alkyl, (C? -C6) alkoxy or a partially or fully saturated cycloalkyl (C3-C) and the other of R3 and R4 is -C (R8) (R9) (R10); or R3 and R4 are joined together to form = CHR11; each R5 is independently selected from the group consisting of H, alkoxy (Ci-Cß), -OH, halo, -CN, -NH2 and -NO2; one of Rd and R7 is (C3-C6) alkyl or a (C3-C7) cycloalkyl partially or fully saturated and the other of R6 and R7 is phenyl optionally substituted with 1 to 3 substituents independently selected from the group consisting of -OH , -CN, halo, halo-substituted alkyl (Ci-Cß), alkoxy (C1-C3) halo-substituted, alkyl (Ci-Cß) and (C 1 -C 3) alkoxy; phenylmethyl in which the
  1. The phenllo moiety is optionally substituted with 1 to 3 substituents independently selected from the group consisting of -OH, -CN, halo-substituted alkyl (d-Cß), halo-substituted (C1-C3) alkoxy, and (C1-C3) alkoxy; or heteroaryl-B optionally substituted with 1 to 3 substituents independently selected from the group consisting of -OH, -CN, halo, halo-substituted (Ci-Cß) alkyl, halo-substituted (C 1 -C 3) alkoxy, alkyl (d) -Cβ) and (C1-C3) alkoxy; and wherein said heteroaryl-B is selected from the group consisting of thienyl, thiazolyl, isothiazolyl, isoquinolinyl, quinolinyl, 3- or 4-pyridyl, pyrimidyl, oxazolyl, furanyl, imidazolyl, isoxazolyl, pyrazinyl, pyridazinyl and pyrazolyl; two of R8, R9 and R10 are independently H or alkyl (Ci-Cß) and the ring of R8, R9 and R10 is phenyl, a (C3-C7) cycloalkyl partially or completely saturated, heeroaryl-C or a heterocycle-B of 4 to 7 members partially or completely in the saftered state, in which heteroaryl-C is selected from the group consisting of indol-2-yl, indol-3-yl, indazol-3-yl, 7-azaindol-2-yl and 7- azaindoI-3-yl; and said phenyl, partially or fully saturated cycloalkyl, heteroaryl-C or partially or fully-functional heterocycle-B is optionally substituted in the carbon atom (s) with 1 to 3 independently selected from the group consisting of alkoxy. (C? -C6), F, Cl, -CN, -OH, -COH, idrazole and (C? -C6) alkoxy halo-susiiuuide; and R11 is phenyl, a (C3-C7) partial cycloalkyl or fully saturated, heeroaryl-C or a 4- to 7-membered heterocycle-B or saturated complex, in which heteroaryl-C is selected from the group consisting of indol-2-lyl, indol-3-yl, indazol-3-yl, 7-azaindol-2-yl and 7-azaindol-3-yl; said phenyl, partially or fully saturated cycloaicyl, heeroaryl-C or partially or fully saturated heterocycle-B is optionally substituted on the carbon atom (s) with 1 to 3 substituents independently selected from the group consisting of alkoxy ( Ci-Cβ), F, Cl, -CN, -OH, -CO 2 H, tetrazole and halo-substituted halo (C 1 -C 2) alkoxy; a pharmaceutically acceptable salt thereof, a prodrug of said compound or said salt or a solvate or hydrate of said compound, said salt or said prodrug.
  2. 2. The compound according to claim 1, of formula (III) (Hl) wherein R1, R2, R3, R4 and R5 are as defined in claim 1; or a pharmaceutically acceptable salt thereof.
  3. 3. The compound according to claim 1 or 2, wherein one of R3 and R4 is H, (C1-C3) alkyl or (C1-C3) alkoxy, and the r of R3 and R4 is -C (R8) (R9) (R10); or R3 and R4 are taken together to form = CHR11; R1 is selected from the group consisting of (C2-C6) alkyl, -FC3, phenyl, phenyl-(C1-C3) alkyl, heeroaryl-A, heteroaryl-A-C1-C3 alkyl, a heterocycle-A of 4 to 7 members partially or completely saturated, a partially or fully saturated heterocyclyl-C1-C3 alkyl of 4 to 7 members and a (C3-C7) cycloalkyl (C1-C3) alkyl partially or completely saturated, and when none of R6 and R7 is phenylmethyl, R1 is selected from said group and partially or fully saturated cycloalkyl (C3-C7), wherein heteroaryl-A is selected from the group consisting of thienyl, 2-pyridyl, pyrimidyl, pyrazinyl and pyrazolyl; and wherein the partially or fully saturated phenyl, heteroaryl-A, heterocyclic-A or partially or fully saturated cycloalkyl group or part of a group is optionally substituted with 1 to 3 substituents independently selected from the group consisting of F, Cl, alkoxy (C1-C3), -OH, (C1-C3) alkyl, -CN and -CF3; or a pharmaceutically acceptable salt thereof.
  4. 4. The compound according to any one of claims 1-3, wherein each R5 is H; one of R6 and R7 is branched chain (C3-C6) alkyl and the r of R6 and R7 is phenyl optionally substituted with 1 to 3 substitutes independently selected from the group consisting of -OH, -CN, F, Cl, (C 1 -C 3) alkyl substituted with F or Cl, (C 1 -C 3) alkoxy substituted with F or Cl, (C 1 -C 4) alkyl and (C 1 -C 3) alkoxy; phenylmethyl in which the phenyl moiety is optionally substituted with 1 to 3 members independently selected from the group consisting of -OH, -CN and (C1-C3) alkoxy; or heteroaryl-B optionally substituted with 1 to 3 substituents independently selected from the group consisting of -OH, -CN, F, Cl, (C1-C3) alkyl substituted with F or Cl, (C1-C3) alkoxy susiiluted with F or Cl, (C1-C4) alkyl and (C1-C3) alkoxy; and wherein said heleroaryl-B is selected from the group consisting of thienyl, 3- or 4-pyridyl, pyrimidyl and pyrazinyl; two of R8, R9 and R10 are H and the r of R8, R9 and R10 is heteroarii-C selected from the group consisting of indol-3-yl, indazol-3-yl and 7-azaindol-3-yl, and said heteroaryl-C is optionally substituted on the carbon atom (s) with 1 to 3 substituents independently selected from the group consisting of (C1-C3) alkoxy, F, Cl, -CN, -OH, - CO2H, tefrazol and (C1-C3) alkoxy susiiuuid with F; and R11 is a heleroaryl-C selected from the group consisting of indol-3-yl, indazol-3-yl and 7-azaindol-3-yl, and said heteroaryl-C is optionally substituted in the (or) anomers ( s) of carbon with 1 to 3 substituents independently selected from the group consisting of (C 1 -C 3) alkoxy, F, Cl, -CN, -OH, -CO 2 H, telrazole and (C 1 -C 3) alkoxy substituted with F; or a pharmaceutically acceptable salt thereof.
  5. 5. - The compound according to any one of claims 1-4, of formula (IV)
  6. (IV) wherein R3 is H or (C1-C3) alkyl; X1 is -CH- and X2 is -N- or -C (R12) -, or X1 is -N- and X2 is -C (R12) -; and each R12 is independently selected from the group consisting of H, F and Cl; with the condition, however, that no more than three of R12 are different from H; or a pharmaceutically acceptable salt thereof. 6. The compound according to any one of claims 1-5, wherein R1 is phenyl or 2-pyridyl or, when none of R6 and R7 is phenylmethyl, R1 may also be a partial or complete (C5-C7) cycloalkyl. salified, in which the phenyl, 2-pyridyl or (C5-C7) cycloalkyl partially or fully saturated is optionally substituted with 1 to 3 independently selected substituents in the group consisting of F, Cl, (d-C3) alkoxy, -OH , (C1-C3) alkyl and -CF3; R3 is H; one of R6 and R7 is a branched chain (C3-C5) alkyl and the other of R6 and R7 is phenyl optionally substituted with 1 to 3 substituents independently selected from the group consisting of -OH, -CN, F, Cl, (C 1 -C 3) alkyl substituted with F, (C 1 -C 3) alkoxy substituted with F, (C 1 -C 4) alkyl and (C 1 -C 3) alkoxy; phenylmethyl optionally substituted with 1 or 2 substituents independently selected from the group consisting of -OH and -OCH 3; or 3- or 4-pyridyl optionally substituted with 1 to 3 independently selected aminoides from the group consisting of -OH, -CN, F, Cl, (C1-C3) alkyl substituted with F, (C1-C3) alkoxy substituted with F, (C 1 -C 4) alkyl and (C 1 -C 3) alkoxy; or a pharmaceutically acceptable salt thereof.
  7. 7. The compound according to any one of claims 1-6, wherein R1 is phenyl, or cyclohexyl when none of R6 and R7 is phenylmethyl, wherein the phenyl is optionally substituted with 1 to 3 atoms of F or a group -OH; one of R6 and R7 is branched chain alkyl (C3 or C4) and the other of R6 and R7 is phenyl optionally substituted with 1 to 3 substituents independently selected from the group consisting of -OH, F, Cl, -CF3, - OCF3, (C1-C4) alkyl and (C1-C3) alkoxy; phenylmethyl; or 3- or 4-pyridyl optionally susiiuuid with 1 to 3 substituents independently selected from the group consisting of -OH, F, Cl, -CF3, -OCF3, (C1-C4) alkyl and (C1-C3) alkoxy; or a pharmaceutically acceptable salt thereof.
  8. 8. - A compound according to any one of claims 1-7, of Formula (V)
  9. (V) in which X1 is -CH- or -N-; and one of R6 and R7 is isopropyl and the other of R6 and R7 is phenyl optionally substituted with 1 or 2 F atoms; phenylmethyl; or 3- or 4-pyridyl optionally susliuid with 1 or 2 independently selected substituents in the group consisting of F, Cl, -OCH3 and -OCH2CH3; and each R12 is independently H or F; with the condition, however, that no more than three of R12 are F; or a pharmaceutically acceptable salt thereof. 9. The compound according to any one of claims 1-8, wherein X1 is -CH-; one of R6 and R7 is isopropyl and the other of R6 and R7 is phenyl or phenylmethyl; and each R12 is H; or a pharmaceutically acceptable salt thereof.
  10. 10. The compound according to any one of claims 1-8, wherein X1 is -CH-; one of R6 and R7 is sopropyl and the other of R6 and R7 is 3-pyridyl optionally substituted with 1 or 2 substituents independently selected from the group consisting of F, Cl, -OCH3 and -OCH2CH3; and each R12 is H; or a pharmaceutically acceptable salt thereof.
  11. 11.- The compote N-benzyl-2- [4- (1H-indol-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-tetraaza-benzo [ e] azulen-6-yl] -N-isopropyl-acetamide; or a pharmaceutically acceptable salt thereof.
  12. 12.- The compound (-) N-benzyl-2- [4- (1H-indol-3-ylmethyl) -5-oxo-1-phenyl-4,5-dihydro-2,3,6,10b-lefraza -benzo [e] azulen-6-yl] -N-isopropyl-acetamide.
  13. 13. A pharmaceutical composition, comprising a compound according to any one of claims 1-12 or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable excipient, diluent or vehicle.
  14. 14. A method for treating obesity in an animal, comprising administering to an animal in need of this treatment a therapeutically effective amount of a compound according to any one of claims 1-12 or a pharmaceutically acceptable salt thereof.
  15. 15. - A compound of Formula (D) or Formula (F-1) (F-1) wherein A, B, X and D are independently -C (R5) - or -N-, with the proviso that not more than two A, B, X and D are N at the same time; R is alkyl (CI-CT) or cyclo (C3-C6) alkyl; R1 is selected from the group consisting of (C2-C6) alkyl, halo-substituted alkyl (Ci-Cß), alkylamino (d-Cß), di (C 1 -Ce) amino, alkylamino (C 6 -C 6) -alkyl (C1-C6), di-alkylamino (C? -Ce) -alkyl (C? -C6), aryl, aryl-alkyl (Ci-Cß), heeroaryl-A, heteroaryl-A-alkyl (Ci-Cß) ), a 4 to 7-membered heteroaryl-A partially or completely saturated, a 4-to-7-membered heteroaryl-C-alkyl (Ci-Cß) or a saturated and a (C3-C7) -alkyl (Ci-Cß) cycloalkyl. ) partially or completely saturated and, when none of R6 and R7 is phenylmethyl, R1 is selected from said group and (C3-C7) cycloalkyl partially or completely saturated; wherein heteroaryl-A is selected from the group consisting of thienyl, isoazolyl, isopiazolyl, indolyl, 2-pyridyl, pyridazinyl, pyrimidyl, oxazolyl, furanyl, imidazolyl, isoxazolyl, quinolinyl, isoquinolinyl, benzothiophenyl, benzofuranyl, pyrazinyl and pyrazolyl, and the partial or fully saturated heterocycle is selected from the group consisting of pyranyl, morpholinyl and teirahydrofuranyl, and wherein the aryl, heteroaryl-A, partially or fully saturated heteroaryl-A or the cycloalkyl group partially or fully salted or from a group is optionally substituted with 1 to 3 selected independent subsitutes in the group consisting of halo, (C 1 -C 3) alkoxy, (C 1 -C 3) alkoxy-halo-substituted, -OH, (C 1 -C 3) alkyl. -CN and (C1-C3) alkyl halo-susíifuido; R2 is -CH2C (O) N (R6) (R7); each R5 is independently selected from the group consisting of H, alkoxy (C? -C6), -OH, halo, -CN, -NH2 and -NO2; one of R6 and R7 is (C3-C6) alkyl or a (C3-C7) cycloalkyl partially or completely saturated and the other of R6 and R7 is phenyl optionally susiiuuid with 1 to 3 susíifuyeníes independently selected from the group consisting of -OH , -CN, halo, halo-substituted alkyl (d-Cß), alkoxy (C1-C3) halo-suslifuido, alkyl (d-Cß) and (C1-C3) alkoxy; phenylmelyl in which the The phenyl ring is optionally susíifuido with 1 to 3 independently selected substances in the group consisting of -OH, -CN, halo-substituted (Ci-Cß) alkyl, halo-substituted (C 1 -C 3) alkoxy and (C 1 -C 3) alkoxy; or heteroaryl-B optionally substituted with 1 to 3 substituents independently selected from the group consisting of -OH, -CN, halo, halo-substituted alkyl (d-Cβ), halo-substituted (C1-C3) alkoxy, alkyl (C1) -Cβ) and (C1-C3) alkoxy; and in which heeroaryl B is selected from the group consisting of thienyl, thiazolyl, isothiazolyl, isoquinolinyl, quinolinyl, 3- or 4-pyridyl, pyrimidyl, oxazolyl, furanyl, imidazolyl, isoxazolyl, pyrazinyl, pyridazinyl and pyrazolyl; or a salt of it.
MXPA/A/2006/011123A 2004-05-25 2006-09-27 Tetraazabenzo[e]azulene derivatives and analogs thereof MXPA06011123A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US60/574,033 2004-05-25

Publications (1)

Publication Number Publication Date
MXPA06011123A true MXPA06011123A (en) 2007-04-20

Family

ID=

Similar Documents

Publication Publication Date Title
US7265104B2 (en) Tetraazabenzo[e]azulene derivatives and analogs thereof
KR100263625B1 (en) 1,5-benzodiazepine derivatives and their use in medicine
EP0167919B1 (en) Benzodiazepine derivatives and pharmaceutical compositions containing them
US5618812A (en) Benzodiazepine derivatives
KR100840852B1 (en) Tetraazabenzo[e]azulene derivatives and analogs thereof
US5569654A (en) Benzodiazepinones
US4663321A (en) Triazolobenzodiazepines and pharmaceutical use
EP0284256A1 (en) Benzodiazepine analogs
EP0434369A1 (en) New benzodiazepine analogs
EP0490590A1 (en) New benzodiazepine analogs
US4847248A (en) 1,4-Benzodiazepines with 5- and 6-membered heterocyclic rings and their use as cholecystokinins and gastrin antagonists
US5859007A (en) CCK or gastrin modulating benzo b! 1,4! diazepines derivatives
EP0272866A1 (en) 1,4-Benzodiazepines with 5-membered heterocyclic rings
RU2152939C1 (en) 5-heterocyclo-1,5-benzodiazepines and their pharmaceutically acceptable salts
IL101514A (en) Benzodiazepinone derivatives and cholecystokinin antagonist pharmaceutical compositions containing them
US4735941A (en) 1,4-benzodiazepines with 5- and 6-membered heterocyclic rings, useful as gastrointestinal and CNS agents
MXPA06011123A (en) Tetraazabenzo[e]azulene derivatives and analogs thereof
HUT74091A (en) 1,5-benzodiazepine derivatives having cck and/or gastrin antagonistic activity
US5486514A (en) Carbamate derivatives
WO2010067233A1 (en) 1,2,4 triazolo [4, 3 -a] [1,5] benzodiazepin-5 (6h) -ones as agonists of the cholecystokinin-1 receptor (cck-ir)
WO1995006041A1 (en) 3-phenylureido-1,4-benzodiazepines useful as selective cck-b antagonists
EP0272868A1 (en) 1,4-Benzodiazepines with 6-membered heterocyclic rings
SI9300328A (en) 1,5-Benzodiazepine Derivates