WO2015184011A2 - Agonists of the apelin receptor and methods of use thereof - Google Patents

Agonists of the apelin receptor and methods of use thereof Download PDF

Info

Publication number
WO2015184011A2
WO2015184011A2 PCT/US2015/032748 US2015032748W WO2015184011A2 WO 2015184011 A2 WO2015184011 A2 WO 2015184011A2 US 2015032748 W US2015032748 W US 2015032748W WO 2015184011 A2 WO2015184011 A2 WO 2015184011A2
Authority
WO
WIPO (PCT)
Prior art keywords
optionally substituted
compound
apelin
group
phenyl
Prior art date
Application number
PCT/US2015/032748
Other languages
French (fr)
Other versions
WO2015184011A3 (en
Inventor
Anthony B. Pinkerton
Layton H. SMITH
Original Assignee
Sanford-Burnham Medical Research Institute
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 Sanford-Burnham Medical Research Institute filed Critical Sanford-Burnham Medical Research Institute
Priority to US15/313,938 priority Critical patent/US10570128B2/en
Publication of WO2015184011A2 publication Critical patent/WO2015184011A2/en
Publication of WO2015184011A3 publication Critical patent/WO2015184011A3/en
Priority to US16/742,818 priority patent/US10988475B2/en
Priority to US17/208,919 priority patent/US20210363142A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41961,2,4-Triazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/42Oxazoles
    • A61K31/422Oxazoles not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/427Thiazoles not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/04Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings

Definitions

  • This invention relates generally to small molecule agonists of the apelin receptor (APJ) and, more specifically, to compounds for the treatment of apelin receptor-mediated diseases and disorders.
  • APJ apelin receptor
  • G protein-coupled receptors are activated by a plethora of molecules including neuropeptides, polypeptide hormones and non-peptides such as biogenic amines, lipids, nucleotides and ions. They are classically composed of seven membrane-spanning domains and constitute one of the largest and most diverse gene families in the mammalian genome. Some novel GPCRs do not have obvious endogenous ligands and are termed orphan receptors, a number of which appear to be constitutively active.
  • Both APJ and apelin have been implicated as the key mediators of physiological responses to multiple homeostatic perturbations, including cardiovascular control, water balance, hypothalamic-pituitary-adrenal (HPA) axis regulation and metabolic homeostasis.
  • HPA hypothalamic-pituitary-adrenal
  • Homeostatic stability is critical in mammalian organisms, and knowledge as to how this vital function is regulated and how this mechanism can go wrong in pathological conditions is still limited.
  • APJ was first identified as an orphan GPCR, with closest identity to the angiotensin II (Ang II) receptor, type ATi a .
  • Ang II angiotensin II
  • APJ remained an orphan receptor until 1998 when a 36-amino acid peptide termed apelin, for APJ endogenous ligand was identified.
  • the receptor was deorphanised when its cognate ligand, apelin, was isolated from bovine stomach extracts. Recently, the apelinergic system has been shown to be critically involved in multiple homeostatic processes.
  • the protein structure of APJ is typical of a GPCR, containing seven hydrophobic transmembrane domains, with consensus sites for phosphorylation by protein kinase A (P A), palmitoylation and glycosylation.
  • P A protein kinase A
  • the N-terminal glycosylation of GPCRs has been implicated in receptor expression, stability, correct folding of the nascent protein and ligand binding.
  • the palmitoylation of the C-terminal tail has been reported to play a role in membrane association and, combined with receptor phosphorylation, these fatty acid modifications can influence the internalization, dimerization and ligand binding of a GPCR.
  • Structural studies on APJ have determined that amino acids in both the N-terminal (e.g., Asp 23 and Gkf °) and C-terminal portions of the receptor are required for internalization.
  • APLN human apelin
  • Apln chromosome Xq25- 26.1 and possesses one intron within its open reading frame of ⁇ 6kb.
  • Apln the genes are termed Apln and are located at chromosomal locations Xq35 and XA3.2 respectively.
  • the core promoter regions of these genes have been identified as -207/- 1 and -100/+ 74 bp in rats and humans respectively.
  • a CAAT box but no TATA box, sequence is present in the rat and human promoter regions.
  • rat and human preproapelin cDNAs do not have a classical Kozak consensus sequence surrounding the initiating methionine codon.
  • Preproapelin 77-amino acid preproprotein
  • Bovine, human, rat and mouse preproapelin precursors have 76-95% homology and appear to exist endogenously as a dimeric protein, as a consequence of disulfide bridges formed between cysteine residues.
  • apelin-36 would constitute a mature form of the peptide.
  • C-terminal portion of preproapelin also contained lysine (Lys, K) and arginine (Arg, R) residues, and given their potential as sites for proteolytic cleavage, the existence of apelin-17 and apelin-13 peptides was predicted, along with a pyroglutamylated form of apelin- 13 ((Pyr')apelin-13).
  • apelin-36 and apelin- 13 have been determined in aqueous solution, indicating that both possess an unordered structure.
  • the amino acid sequence homology of the mature apelin-36 peptide is more conserved between species than that of preproapelin, with 86-100% homology between bovine, human, rat and mouse amino acid sequences, while the 23 C-terminal amino acids have 100% homology between species, suggesting an important physiological role.
  • APJ does not bind Ang II (O'Dowd et al. 1993)
  • apelin- 13 shares a limited homology (four amino acids) with the vasoconstrictive peptide.
  • Ang I- converting enzyme 2 which catalyzes the C-terminal dipeptide cleavage of Ang I to Ang II, or Ang II to Ang 1-7, also acts on apelin-13 with a high catalytic efficiency, removing the C-terminal phenylalanine (Phe, F) residue.
  • this cleavage may not inactivate the peptide, as the apelin isoform K 16P, which lacks the terminal Phe, while ineffective at inducing receptor internalization or regulating blood pressure (BP) (effects associated with the full peptide), still binds to APJ and inhibits forskol in-stimulated cAMP production.
  • BP blood pressure
  • APJ and apelin mRNAs and proteins are widely distributed in the CNS and peripheral tissues, whether the levels of mRNAs present in most of the regions of the brain and tissues are functionally relevant is not yet known.
  • APJ binds numerous apelin isoforms and signals through various G proteins to a variety of signaling pathways to culminate in different patterns of activation and
  • APJ has also been reported to heterodimerize with other GPCRs and to signal in the absence of an endogenous ligand.
  • the C-terminal region of the apelin peptide may be responsible for its overall biological activity.
  • N-terminal deletions of apelin-17 reveal that the 12 C-terminal amino acids may be the core requirements for the internalization and biological potency of APJ.
  • Apelin- 17 induces the internalization of APJ, which decreases with every N-terminal deletion to apelin- 12, while the deletion of the terminal F amino acid results in a peptide that no longer internalizes APJ or affects arterial BP.
  • the N-terminal residues within the RPRL motif (residues 2-5) of apelin-13 are critical for functional potency, and the C-terminal sequence KGPM (residues 8-1 1 ) is important for binding activity and for internalization.
  • the five N-terminal and two C-terminal amino acids of apelin- 17 are not required for binding of the peptide to APJ or activation of receptor signaling (e.g., cAMP production). Although this may indicate a possible dissociation between the conformational states of the receptor responsible for receptor signaling and internalization, it is also possible that different ligand isoforms may induce differential receptor trafficking and signaling.
  • apelin/APJ Although progress has been made in recent years in clarifying the physiological significance of apelin/APJ, much remains to be discovered about the expression of the apelinergic system and precisely how it affects numerous physiological functions. Since the discovery of the apelin ligand, both apelin and APJ have been implicated as key regulators of central and peripheral responses to multiple homeostatic perturbations. These include playing pivotal roles in the regulation of cardiovascular function, angiogenesis, fluid homeostasis and energy metabolism and acting as neuroendocrine modulators of the HPA axis responses to stress. It is becoming apparent that the apelinergic system may play a pathophysiological role within many of these regulatory systems.
  • ISHH angiotensinogen
  • Ang II is part of the rennin-angiotensin system (RAS), which controls extracellular fluid volume and arterial vasoconstriction, thereby regulating mean arterial blood pressure (MABP).
  • RAS rennin-angiotensin system
  • MABP mean arterial blood pressure
  • the central actions of the RAS include the regulation of drinking behavior, salt appetite and VP secretion.
  • the RAS plays a critical role in the pathogenesis of heart failure.
  • apelin exerts many physiological effects that appear to oppose those exerted by Ang II. More recently, apelin has been shown to block many Ang II-initiated processes, perhaps partly by dimerization between APJ and AT,.
  • APJ plays a role in the regulation of BP comes from a study on mice with a global deletion of APJ, where a transient decrease in systolic BP observed in conscious wild-type (WT) mice following i.p. injection of (Pyr')apelin- 13 is abolished in APJ KO mice.
  • peripheral apelin is a vasodilator in the human saphenous vein, in vessels denuded of endothelium, apelin acts as a vasoconstrictor. Therefore, peripheral apelin may act as an antihypertensive factor, and sensitivity to the peripheral administration of apelin might be altered in hypertensive disease.
  • the apelinergic system has an important role in cardiac function.
  • infusion of apelin- 16 induces a potent dose-dependent positive inotropic effect, with an EC3 ⁇ 4, of 40-125 pM in humans and—33 pM in rats, an effect also observed in the failing heart.
  • administration of apelin increases myocardial contraction while reducing cardiac preload and afterload, without causing hypertrophy.
  • apelin increases the shortening of sarcomeres in cardiomyocytes, an effect that is impaired in isolated ventricular myocytes from apelin and APJ KO mice.
  • Apelin KO mice have an impaired response to cardiac pressure overload, thus suggesting a role for apelin/APJ in the sustainability and amplification of the cardiac response to stress.
  • EHT essential hypertension
  • the apelinergic system plays a role in the Cripto signaling pathway (which stimulates signaling by the transforming growth factor Nodal or growth/differentiation factors 1 and 3, via activin type IB and type IIB receptors) in mammalian cardiac myogenesis.
  • APJ KO mice cardiovascular development defects have been reported in APJ KO mice, where a loss of homozygous mutants has been described, but not in apelin KO mice, indicating possible ligand-independent effects of the receptor. This effect may perhaps be explained by the recent report that APJ signals independently of apelin in response to cardiac mechanical stretch. APJ KO embryos at El 0.5, when lethality begins, have poorly developed vasculature of the yolk sac, delayed formation of the atrioventricular cushion and unusually formed cardinal veins and dorsal aorta.
  • APJ KOs that survive do not reveal any apparent morphological differences; however, they have decreased vascular smooth muscle layer recruitment and myocardial defects including thinning of the myocardium, enlarged right ventricles and ventricular septal defects, suggesting an involvement of apelin/APJ signaling in cardiovascular development.
  • Apelin appears to have a role to play in the pathophysiology of the cardiovascular system - it has been implicated in vascular diseases, heart failure, and ischemia and subsequent reperfusion.
  • vascular diseases the expression of apelin is up-regulated in the atherosclerosis of human coronary artery.
  • its role is undetermined, as conflicting evidence has been found in KO studies, indicating both antagonistic and inducing roles for apelin in atherosclerotic formation.
  • plasma apelin levels rise in the early stages of disease and stabilize or lower as the condition develops.
  • APJ mRNA is decreased in the weakened and enlarged heart of humans with idiopathic dilated
  • Apelin may have a cardioprotective role in hypoxia and ischemia, where the cardiac levels of apelin and APJ respectively are increased. Apelin may also play a protective a role in ischemia /reperfusion injury, although the method of signaling appears to be independent of the characteristic myocardial kinase cascade, termed the reperfusion injury salvage kinase pathway. Post-infarct treatment with (Pyr')apelin- 13 reduces infarct size and increases HR, with a long-term antioxidant cardioprotective action. [0021 ] Apelin is an angiogenic factor and mitogen of endothelial cells.
  • apelin is required for the normal development of frog heart and formation of murine blood vessels. Additionally, the development of the retinal vasculature is stunted in apelin KO mice, and apelin is necessary for hypoxia-induced retinal angiogenesis, and is also involved in non- neovascular remodeling of the retina.
  • the apelinergic system has been implicated in tumor neoangiogenesis.
  • the expression of apelin and APJ is up-regulated in microvascular proliferations, while tumor cell lines overexpressing apelin show increased growth.
  • the pathophysiological effects of apelin in angiogenesis have also been reported for the liver, where the apelinergic system is a factor in portosystemic collaterization and splanchnic neovascularization in portal hypotensive rats as well as in neovascularization during liver cirrhosis.
  • apelin may have therapeutic effects in ischemia recovery due to vessel regeneration and endothelial proliferation and blood vessel diameter regulation.
  • VP along with OT, is synthesized primarily in the neurones of the mPVN and SON, which project to the posterior pituitary and release the peptides into the systemic circulation.
  • the predominant endocrine function of VP from this source is to increase water permeability in the renal collecting duct cells, thereby allowing the retention of water.
  • apelin and APJ mRNAs are increased by dehydration, while the labelling of VP-immunoreactive cells decreases, implying the differential regulation of these peptides in response to dehydration.
  • abnormal fluid homeostasis has been demonstrated in APJ KO mice, manifested by a decrease in drinking behavior and an inability to concentrate urine to levels observed in controls during water deprivation, suggesting an antidiuretic effect of apelin in vivo.
  • apelin induces diuresis and has direct effects on renal vasculature.
  • APJ is also necessary in dehydration-induced signaling in the subfornical organ, implicating the apelinergic pathway in responses to hyperosmotic stimuli.
  • a number of studies have pointed out an emerging involvement of apelin in energy metabolism and a role for adipocyte-derived apelin in the (patho)-physiology of obesity has been reported. Both apelin and APJ mRNAs are present in mouse, human and rat adipose tissue, and their levels increase in adipose tissue and plasma with obesity. This highlights APJ as an interesting therapeutic target for metabolic disorders.
  • the expression of plasma apelin is increased only in obese humans and in mouse models of obesity associated with hyperinsulinemia, indicating that obesity or high-fat feeding may not be the main cause for the rise in the expression of apelin, and implying a close relationship between apelin and insulin both in vivo and in vitro.
  • Insulin directly acts on adipocytes in vitro to stimulate the production of apelin, and the expression of apelin mRNA is down-regulated in the adipocytes of mice treated with the ⁇ -cell toxin streptozotocin, which leads to a fall in plasma insulin levels.
  • nutritional status influences apelin levels in vivo - fasting inhibits plasma levels, which are then restored by re-feeding - thus strengthening the implication that insulin regulates apelin gene expression and secretion.
  • apelin perhaps through APJ expressed in pancreatic islet ⁇ -cells, regulates the secretion of insulin - apelin inhibits glucose-stimulated insulin secretion in vivo in mice and in isolated islets of Langerhans in vitro.
  • apelin has been shown to alleviate diabetes-induced reduction of pancreatic islet mass and to improve the insulin content of pancreatic islets in type 1 diabetic mice.
  • Apelin may have a positive effect in the metabolic syndrome (a combination of risk factors that when occurring together increase the risk of coronary artery disease, stroke and type 2 diabetes (T2D)).
  • Apelin KO mice have reduced insulin sensitivity, are glucose intolerant and are hyperinsulinemic.
  • the peripheral administration of apelin reduces peak plasma glucose concentrations by increasing glucose uptake in skeletal muscle and adipose tissue and improves insulin sensitivity in both apelin KO and obese high-fat diet fed mice, with the insulin-sensitizing effects continuing for up to 4 weeks, with no tolerance to the actions of apelin.
  • Apelin increases glucose uptake, both in vitro and in vivo, through both insulin-dependent and -independent pathways.
  • Apelin may also decrease body adiposity, independently of altered food intake, by increasing energy expenditure through the activation of mitochondrial uncoupling proteins 1 and 3.
  • Clinical studies have shown a promising therapeutic value for apelin, as apelin displays beneficial glucose-lowering effects in human adipose tissue and plasma apelin levels correlate with glucose and HbA l c levels.
  • Apelin is linked to the pathogenesis of T2D - plasma apelin concentrations are increased in insulin- resistant patients, in type T2D patients and in morbidly obese T2D individuals, perhaps indicating a compensatory role of apelin in the reduction of insulin resistance.
  • plasma apelin levels are reduced in newly diagnosed T2D patients and increased in T2D patients and obese non-diabetic individuals.
  • APJ is localized in the hypothalamic pPVN and the anterior pituitary gland, key areas involved in the stress response. Apelin mRNA is also present in these areas, co-localizing with VP in the mPVN, SON and pituitary. Additionally, apelin immunostaining of cell bodies and fibers is highest in the hypothalamus, with large numbers of apelin-positive cell bodies present in the PVN and SON. The presence of APJ and apelin in VP- and CRH-containing hypothalamic nuclei, which are pivotal to the HPA axis responses to stress, suggests a role for apelin/APJ in neuroadenohypophysial hormone release.
  • a role for apelin in the regulation of the HPA axis responses to stress is supported by studies showing that central administration of (Pyr')apelin-13 increases the expression of c-fos, an indicator of neuronal activity, in the PVN. Furthermore, administration of apelin-13 stimulates the release of CRH and VP from hypothalamic extracts in vitro, effects consistent with stimulation of the stress axis. APJ mRNA levels increase in the PVN in response to acute and chronic stress and following adrenalectomy, implying negative regulation of the expression of APJ mRNA by glucocorticoids. Additionally, dexamethasone, a glucocorticoid agonist, decreases apelin mRNA levels in 3T3-L1 mouse adipocytes.
  • Apelin may potentially stimulate the secretion of ACTH either directly at the level of the pituitary corticotroph or via an indirect action on the hypothalamus involving the release of both VP and CRH. Consistent with the expression of apelin and APJ in anterior pituitary corticotrophs, administration of apelin- 17 directly increases the release of ACTH, while also augmenting K * -stimulated ACTH release, in an ex vivo perfusion system of anterior pituitary glands, suggesting possible autocrine or paracrine functions for apelin in this tissue. Central administration of (Pyr')apelin-13 in rats also increases plasma ACTH and CORT levels while decreasing prolactin, luteinizing hormone and follicle-stimulating hormone levels.
  • APJ has been shown to play a regulatory role in the modulation of the HPA axis responses to some acute stressors including LPS challenge (an immune stressor), insulin-induced hypoglycemia (a metabolic stressor) and forced swim (a physical/psychological stressor).
  • LPS challenge an immune stressor
  • insulin-induced hypoglycemia a metabolic stressor
  • forced swim a physical/psychological stressor
  • Apelin was first isolated from stomach extracts, and studies on the actions of apelin in the gastrointestinal system have found functional, and possible cell survival, roles.
  • apelin/APJ may be regulators of hormone and gastric acid secretion.
  • Apelin/APJ may also have a direct effect on vascular smooth muscle, including vasoconstriction, which may affect renal glomerular hemodynamic function in the rat kidney.
  • Some studies have also proposed an immunological role for apelin as it reduces the production of cytokines in mouse spleen cells, suggesting that apelin may modulate neonatal immune responses through rodent and bovine colostrum and milk.
  • APJ is also a co-receptor of HIV entry into target cells, an action that is blocked by apelin.
  • APJ may contribute to HIV- 1 infection and pathogenesis in CNS-based cells as viral envelope proteins can mediate fusion with APJ-positive, cluster of differentiation 4 (CD4)-negative cells, provided that CD4 is added in trans, and HIV can infect APJ-expressing cells despite their CD4 status.
  • Other possible roles for apelin and APJ in the rodent CNS include antinociception, enhancement of depressive behavior, and facilitation of passive avoidance learning.
  • Apelin may also have a role in neuroprotection, as apelin pre-treatment protects hippocampal neurones against N- methyl-D-aspartate (NMDA) receptor-mediated excitotoxic injury, possibly via the phosphorylation of Akt and ERK1 /2, and prevents apoptosis in cultured mouse cortical neurones.
  • NMDA N- methyl-D-aspartate
  • apelin and APJ are expressed in osteoblasts where they may induce cell proliferation and promote survival; however, an increase in bone mass can be observed in apeiin KO mice.
  • apeiin has been reported to have a potential role in the pathophysiology of osteoarthritis (OA), as apeiin is present in synovial fluid, and OA patients have elevated plasma apeiin concentrations. Blood plasma levels of apeiin are reduced in patients with polycystic ovary syndrome, consistent with the role played by apelin/APJ in metabolic disturbances such as insulin resistance.
  • apeiin Elevated levels of apeiin have been detected in many pathological states or disease processes, such as heart disease, atherosclerosis, tumor angiogenesis and diabetes. However, in many systems, apeiin has been shown to have positive effects, for example in the cardiovascular system, where it has a cardioprotective effect. This has led to speculation that apeiin and APJ could be beneficial targets for therapeutic strategies for a number of diseases and disorders.
  • the present invention is based on the seminal discovery of a series of potent small molecule agonists of the apeiin receptor, which are useful for the treatment of diseases including heart failure, chronic kidney disease, hypertension, and metabolic disorders such as insulin resistance/diabetes and obesity.
  • the compounds disclosed herein are highly specific for the apeiin receptor versus the angiotensin II receptor (ATI).
  • A is selected from the group consisting of optionally substituted aryl, optionally substituted heteroaryl, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted alkoxy, optionally substituted cycloalkyl, hydrogen, hydroxyl, -CN and halogen;
  • B is selected from the group consisting of optionally substituted aryl, optionally substituted heteroaryl, optionally substituted arylalkyl, optionally substituted heteroarylalkyl, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted cycloalkyl, hydrogen, hydroxyl, -CN and halogen;
  • C is selected from the group consisting of optionally substituted aryl, optionally substituted heteroaryl, optionally substituted arylalkyl, optionally substituted heteroarylalkyl, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted cycloalkyl, hydrogen, hydroxyl, -CN and halogen;
  • Y is selected from null, a bond, O, S, SO, S0 2 , NH, -C(0)-N(R' )-, -C(0)-0-, - N(R 2 )- and -C(R 3 )(R 4 )-;
  • X 1 and X 2 are each independently selected from CR S and N;
  • R 1 , R 2 , R 3 and R 4 are each independently selected from the group consisting of of optionally substituted aryl, optionally substituted 5- or 6-membered heteroaryl, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted alkoxy, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, hydrogen and hydroxyl;
  • R 5 is independently selected from the group consisting of hydrogen, halogen, -CN, -C(0)-N(R 6 )-R 7 , -C(0)-0-R 8 , optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted alkoxy, haloalkyl, haloalkoxy, optionally substituted aryl and optionally substituted 5- or 6- membered heteroaryl; and
  • R 6 , R 7 and R 8 are each independently selected from the group consisting of of optionally substituted aryl, optionally substituted 5- or 6-membered heteroaryl, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted alkoxy, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, hydrogen and hydroxyl.
  • R 6 , R 7 and R 8 are each independently selected from the group consisting of of optionally substituted aryl, optionally substituted 5- or 6-membered heteroaryl, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted alkoxy, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, hydrogen and hydroxyl.
  • R 6 , R 7 and R 8 are each independently selected from the group consisting of of optionally substituted aryl, optionally substituted 5- or 6-membered heteroaryl, optionally substituted alkyl, optionally substituted heteroalkyl, optional
  • A is optionally substituted aryl or optionally substituted heteroaryl.
  • A is optionally substituted phenyl or optionally substituted 5- or 6-membered heteroaryl, wherein the optional substituents are selected from the group consisting of hydrogen, halogen, -CN, optionally substituted carboxyl, amido, nitro, amino, optionally substituted alkyl, optionally substituted alkoxy and optionally substituted haloalkoxy and optionally substituted haloalkyl.
  • A is selected from the group consisting of:
  • Y is selected from the group consisting of S, SO, S0 2 , O and N. In certain embodiments, Y is S, O or N. In one embodiment, Y is an optionally substituted Ci-Cgalkyl group. In other embodiments, Y is S, O, N or NH. In one embodiment, Y is S. In another embodiment, Y is S and X 2 is CH. In yet another embodiment, Y is O and X 2 is N. In a further embodiment, Y is NH and X " is N. In other aspects, C is optionally substituted alkyl, optionally substituted arylalkyl or optionally substituted heteroarylalkyl.
  • C is an optionally substituted alkyl group.
  • C is substituted alkyl, where C is an alkyl substituted with a substituent selected from optionally substituted aryl or optionally substituted heteroaryl.
  • C is an alkyl group substituted with optionally substituted aryl, including but not limited to optionally substituted phenyl.
  • C is an alkyl substituted with optionally substituted monocyclic heteroaryl.
  • C is an alkyl substituted with optionally substituted bicyclic heteroaryl.
  • C is optionally substituted heteroarylalkyl.
  • C is substituted heteroarylalkyl, where C is a heteroarylalkyl substituted with a substituent selected from the group consisting of hydrogen, halogen, -CN, optionally substituted carboxyl, amido, nitro, amino, optionally substituted alkyl, optionally substituted alkoxy and optionally substituted haloalkoxy and optionally substituted haloalkyi.
  • a substituent selected from the group consisting of hydrogen, halogen, -CN, optionally substituted carboxyl, amido, nitro, amino, optionally substituted alkyl, optionally substituted alkoxy and optionally substituted haloalkoxy and optionally substituted haloalkyi.
  • C is optionally substituted arylalkyl, where the optional substituents are selected from the group consisting of hydrogen, halogen, -CN, optionally substituted carboxyl, amido, nitro, amino, optionally substituted alkyl, optionally substituted alkoxy and optionally substituted haloalkoxy and optionally substituted haloalkyi .
  • C is selected from the group consisting of:
  • C is:
  • R 9 is optionally substituted aryl, wherein the optional substituents are selected from the group consisting of hydrogen, halogen, -CN, optionally substituted carboxyl, amido, nitro, amino, optionally substituted alkyl, optionally substituted alkoxy and optionally substituted haloalkoxy and optionally substituted haloalkyl.
  • X 3 is S. In other embodiments, X 3 is O. 9 is selected from:
  • B is optionally substituted aryl, such as optionally substituted phenyl, optionally substituted heteroaryl, such as optionally substituted pyridinyl, or optionally substituted heteroarylalkyl.
  • B is substituted aryl, , substituted heteroaryl or substituted heteroarylalkyl, where substituted aryl, such as substituted phenyl,, substituted heteroaryl, such as substituted pyridinyl, or substituted heteroarylalkyl are substituted with a substituent selected from the group consisting of hydrogen, halogen, -CN, optionally substituted carboxyl, amido, nitro, amino, optionally substituted alkyl, optionally substituted alkoxy and optionally substituted haloalkoxy and optionally substituted haloalkyl.
  • B is substituted phenyl, where the phenyl is substituted with at least one alkoxy, such as methoxy.
  • B is .
  • B is selected from the group consisting of:
  • X 1 and X 2 are each independently N.
  • composition comprising a compound of structural Formula I, or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable excipient.
  • a method of preventing or treating in a subject an apelin-mediated disease or disorder comprising administering to the subject a therapeutically effective amount of a compound of structural Formula I thereby preventing or treating the disease or disorder is also provided herein.
  • the subject is a human.
  • the disease or disorder is a cardiovascular disease or disorder, coronary heart disease, stroke, heart failure, systolic heart failure, diastolic heart failure, diabetic heart failure, heart failure with preserved ejection fraction, cardiomyopathy, myocardial infarction, left ventricular dysfunction, left ventricular dysfunction after myocardial infarction, cardiac hypertrophy, myocardial remodeling, myocardial remodeling after infarction, myocardial remodeling after cardiac surgery or valvular heart disease.
  • the disease or disorder is a metabolic disease or disorder, metabolic syndrome, insulin resistance, diabetes mellitus, diabetic late complications, diabetic macro- and micro-vasculopathies, diabetic nephropathy, diabetic retinopathy, diabetic neuropathies or cardiac autonomic neuropathy.
  • the disease or disorder is caused by CNS-dependent or CNS- independent disturbed fluid homeostasis, acute or chronic renal failure, hypertension, pulmonary hypertension, portal hypertension or systolic hypertension.
  • the disease or disorder is a vascular disease or disorder, vascular permeability, nonfunctional blood vessels, vascular hypertrophy, vascular remodeling, vascular stiffness, atherosclerosis, peripheral arterial occlusive disease (PAOD), restenosis, thrombosis, vascular permeability disorders, ischemia, reperfusion damage, ischemia or reperfusion damage of the heart, kidney or retina, or a combination thereof.
  • the methods provided herein further comprise coadministering an additional compound in combination with a compound of structural Formula I for the treatment of a disease or disorder, such as, by way of example only, coadministering an additional apelin receptor agonist in combination with a compound of structural Formula I for the treatment of an apelin-mediated disease or disorder or coadministering a chemotherapeutic agent in combination with a compound of structural Formula I for the treatment of cancer.
  • an additional compound in combination with a compound of structural Formula I for the treatment of a disease or disorder such as, by way of example only, coadministering an additional apelin receptor agonist in combination with a compound of structural Formula I for the treatment of an apelin-mediated disease or disorder or coadministering a chemotherapeutic agent in combination with a compound of structural Formula I for the treatment of cancer.
  • acyl refers to a carbonyl attached to an alkenyl, alkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, or any other moiety were the atom attached to the carbonyl is carbon.
  • An "acetyl” group which is a type of acyl, refers to a ⁇ C(0)CH 3 group.
  • An "alkylcarbonyl” or “alkanoyl” group refers to an alkyl group attached to the parent molecular moiety through a carbonyl group. Examples of such groups include methylcarbonyl and ethylcarbonyl.
  • acyl groups include formyl, alkanoyl and aroyl.
  • alkenyl refers to a straight- chain or branched-chain hydrocarbon radical having one or more double bonds and containing from 2 to 20 carbon atoms. In certain embodiments, said alkenyl will comprise from 2 to 6 carbon atoms.
  • alkoxy refers to an alkyl ether radical, wherein the term alkyl is as defined below.
  • suitable alkyl ether radicals include methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, iso-butoxy, sec-butoxy, tert- butoxy, and the like.
  • alkyl refers to a straight-chain or branched-chain alkyl radical containing from 1 to 20 carbon atoms.
  • said alkyl will comprise from 1 to 10 carbon atoms. In further embodiments, said alkyl will comprise from 1 to 6 carbon atoms.
  • Alkyl groups may be optionally substituted as defined herein. Examples of alkyl radicals include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, pentyl, iso-amyl, hexyl, octyl, noyl and the like.
  • alkylene refers to a saturated aliphatic group derived from a straight or branched chain saturated hydrocarbon attached at two or more positions, such as methylene (— CH 2 ⁇ ). Unless otherwise specified, the term “alkyl” may include “alkylene” groups.
  • alkylamino refers to an alkyl group attached to the parent molecular moiety through an amino group. Suitable alkylamino groups may be mono- or dialkylated, forming groups such as, for example, N-methylamino, N-ethylamino, ⁇ , ⁇ -dimethylamino, ⁇ , ⁇ -ethylmethylamino and the like.
  • alkylidene refers to an alkenyl group in which one carbon atom of the carbon-carbon double bond belongs to the moiety to which the alkenyl group is attached.
  • alkylthio refers to an alkyl thioether (R— S— ) radical wherein the term alkyl is as defined above and wherein the sulfur may be singly or doubly oxidized.
  • suitable alkyl thioether radicals include methylthio, ethylthio, n-propylthio, isopropylthio, n-butylthio, iso-butylthio, sec-butylthio, tert-butylthio, methanesulfonyl, ethanesulfinyl, and the like.
  • alkynyl refers to a straight- chain or branched chain hydrocarbon radical having one or more triple bonds and containing from 2 to 20 carbon atoms. In certain embodiments, said alkynyl comprises from 2 to 6 carbon atoms. In further embodiments, said alkynyl comprises from 2 to 4 carbon atoms.
  • alkynylene refers to a carbon-carbon triple bond attached at two positions such as ethynylene (— C:::C— ,— C ⁇ C— ).
  • alkynyl radicals include ethynyl, propynyl, hydroxypropynyl, butyn- l -yl, butyn-2-yl, pentyn- l -yl, 3-methylbutyn-l -yl, hexyn-2-yl, and the like.
  • alkynyl may include "alkynylene” groups.
  • acylamino as used herein, alone or in combination, embraces an acyl group attached to the parent moiety through an amino group.
  • amino is acetylamino (CH 3 C(0)NH ⁇ ).
  • amino refers to ⁇ N(R)(R') or - -N + (R)(R')(R"), wherein R, R' and R" are independently selected from the group consisting of hydrogen, alkyl, acyl, heteroalkyl, aryl, cycloaikyl, heteroaryl, and heterocycloaikyl, any of which may themselves be optionally substituted.
  • amino acid means a substituent of the form ⁇ NRCH(R')C(0)OH, wherein R is typically hydrogen, but may be cyclized with N (for example, as in the case of the amino acid proline), and R' is selected from the group consisting of hydrogen, alkyl, heteroalkyl, cycloaikyl, heterocycloaikyl, aryl, heteroaryl, amino, amido, cycloalkylalkyl, heterocycloalkylalkyl, arylalkyl, heteroarylalkyl, aminoalkyl, amidoalkyl, hydroxyalkyl, thiol, thioalkyl, alkylthioalkyl, and alkylthio, any of which may be optionally substituted.
  • amino acid includes all naturally occurring amino acids as well as synthetic analogues.
  • aryl as used herein, alone or in combination, means a carbocyclic aromatic system containing one, two or three rings wherein such rings may be attached together in a pendent manner or may be fused.
  • aryl embraces aromatic radicals such as benzyl, phenyl, naphthyl, anthracenyl, phenanthryl, indanyl, indenyl, annulenyl, azulenyl, tetrahydronaphthyl, and biphenyl.
  • arylalkenyl or “aralkenyl,” as used herein, alone or in combination, refers to an aryl group attached to the parent molecular moiety through an alkenyl group.
  • arylalkoxy or “aralkoxy,” as used herein, alone or in combination, refers to an aryl group attached to the parent molecular moiety through an alkoxy group.
  • arylalkyl or “aralkyl,” as used herein, alone or in combination, refers to an aryl group attached to the parent molecular moiety through an alkyl group.
  • arylalkynyl or “aralkynyl,” as used herein, alone or in combination, refers to an aryl group attached to the parent molecular moiety through an alkynyl group.
  • arylalkanoyl or “aralkanoyl” or “aroyl,” as used herein, alone or in combination, refers to an acyl radical derived from an aryl-substituted alkanecarboxylic acid such as benzoyl, naphthoyl, phenylacetyl, 3-phenylpropionyl(hydrocinnamoyl), 4- phenylbutyryl, (2-naphthyl)acetyl, 4-chlorohydrocinnamoyl, and the like.
  • aryloxy as used herein, alone or in combination, refers to an aryl group attached to the parent molecular moiety through an oxy.
  • benzo and "benz,” as used herein, alone or in combination, refer to the divalent radical ⁇ ⁇ ⁇ 4 derived from benzene. Examples include benzothiophene and benzimidazole.
  • carbamate refers to an ester of carbamic acid (--NHCOO-- ) which may be attached to the parent molecular moiety from either the nitrogen or acid end, and which may be optionally substituted as defined herein.
  • O-carbamyl refers to a— OC(0)NRR', group— with R and R' as defined herein.
  • N-carbamyl as used herein, alone or in combination, refers to a ROC(0)NR' ⁇ group, with R and R' as defined herein.
  • carbonyl when alone includes formyl [— C(0)H] and in combination is a --C(O)— group.
  • carboxyl refers to— C(0)OH, O-carboxy, C-carboxy, or the corresponding "carboxylate” anion, such as is in a carboxylic acid salt.
  • An "O-carboxy” group refers to a RC(0)0 ⁇ group, where R is as defined herein.
  • a “C-carboxy” group refers to a ⁇ C(0)OR groups where R is as defined herein.
  • cyano as used herein, alone or in combination, refers to— CN.
  • cycloalkyl or, alternatively, “carbocycle,” as used herein, alone or in combination, refers to a saturated or partially saturated monocyclic, bicyciic or tricyclic alkyl radical wherein each cyclic moiety contains from 3 to 12 carbon atom ring members and which may optionally be a benzo fused ring system which is optionally substituted as defined herein.
  • said cycloalkyl will comprise from 5 to 7 carbon atoms.
  • cycloalkyl radicals examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, octahydronaphthyl, 2,3-dihydro-l H-indenyl, adamantyl and the like.
  • "Bicyciic” and "tricyclic” as used herein are intended to include both fused ring systems, such as decahydronaphthalene, octahydronaphthalene as well as the multicyclic (multicentered) saturated or partially unsaturated type.
  • esters refers to a carboxyl group bridging two moieties linked at carbon atoms.
  • ether typically refers to an oxy group bridging two moieties linked at carbon atoms.
  • "Ether” may also include polyethers, such as, for example, --RO(CH 2 ) 2 0(CH 2 ) 2 0(CH 2 ) 2 OR', --RO(CH 2 ) 2 0(CH 2 ) 2 OR * , - RO(CH 2 ) 2 OR', and --RO(CH 2 ) 2 OH.
  • halo or halogen
  • haloalkoxy refers to a haloalkyl group attached to the parent molecular moiety through an oxygen atom.
  • haloalkyl refers to an alkyl radical having the meaning as defined above wherein one or more hydrogens are replaced with a halogen. Specifically embraced are monohaloalkyl, dihaloalkyl and polyhaloalkyl radicals.
  • a monohaloalkyl radical for one example, may have an iodo, bromo, chloro or fluoro atom within the radical.
  • Dihalo and polyhaloalkyl radicals may have two or more of the same halo atoms or a combination of different halo radicals.
  • haloalkyl radicals include fluoromethyl, difluoromethyl, trifluoromethyl, chloromethyl, dichloromethyl, trichloromethyl, pentafluoroethyl, heptafluoropropyl, difluorochloromethyl,
  • dichlorofluoromethyl difluoroethyl, difluoropropyl, dichloroethyl and dichloropropyl.
  • Haloalkylene refers to a haloalkyl group attached at two or more positions. Examples include fluoromethylene ( ⁇ CFH--), difluoromethylene (--CF 2 --), chloromethylene (-CHC1--) and the like.
  • heteroalkyl refers to a stable straight or branched chain, or cyclic hydrocarbon radical, or combinations thereof, fully saturated or containing from 1 to 3 degrees of unsaturation, consisting of the stated number of carbon atoms and from one to three heteroatoms selected from the group consisting of O, N, and S, and wherein the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized.
  • the heteroatom(s) O, N and S may be placed at any interior position of the heteroalkyl group. Up to two heteroatoms may be consecutive, such as, for example, ⁇ CH 2 ⁇ NH ⁇ OCH 3 .
  • heteroalkyl may include ethers.
  • heteroaryl refers to 3 to 7 membered unsaturated heteromonocyclic rings, or fused polycyclic rings in which at least one of the fused rings is unsaturated, wherein at least one atom is selected from the group consisting of O, S, and N. In certain embodiments, said heteroaryl will comprise from 5 to 7 carbon atoms.
  • the term also embraces fused polycyclic groups wherein heterocyclic radicals are fused with aryl radicals, wherein heteroaryl radicals are fused with other heteroaryl radicals, or wherein heteroaryl radicals are fused with cycloalkyl radicals.
  • heteroaryl groups include pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, pyranyl, furyl, thienyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, thiadiazolyl, isothiazolyl, indolyl, isoindolyl, indolizinyl, benzimidazolyl, quinolyl, isoquinolyl, quinoxalinyl, quinazolinyl, indazolyl, benzotriazolyl, benzodioxolyl, benzopyranyl, benzoxazolyl, benzoxadiazolyl, benzothiazolyl, benzothiadiazolyl, benzofuryl, benzothienyl, chromonyl,
  • tetrazolopyridazinyl tetrahydroisoquinolmyl
  • thienopyridinyl furopyridinyl
  • pyrrolopyridinyl exemplary tricyclic heterocyclic groups include carbazolyl, benzidolyl, phenanthrolinyl, dibenzofuranyl, acridinyl, phenanthridinyl, xanthenyl and the like.
  • heterocycloalkyl and, interchangeably, “heterocycle,” as used herein, alone or in combination, each refer to a saturated, partially unsaturated, or fully unsaturated monocyclic, bicyclic, or tricyclic heterocyclic radical containing at least one heteroatom as ring members, wherein each said heteroatom may be independently selected from the group consisting of nitrogen, oxygen, and sulfur
  • said heterocycloalkyl will comprise from 1 to 4 heteroatoms as ring members.
  • said heterocycloalkyl will comprise from 1 to 4 heteroatoms as ring members.
  • heterocycloalkyl will comprise from 1 to 2 heteroatoms ring members.
  • said heterocycloalkyl will comprise from 3 to 8 ring members in each ring. In further embodiments, said heterocycloalkyl will comprise from 3 to 7 ring members in each ring. In yet further embodiments, said heterocycloalkyl will comprise from 5 to 6 ring members in each ring.
  • "Heterocycloalkyl” and “heterocycle” are intended to include sugars, sulfones, sulfoxides, N-oxides of tertiary nitrogen ring members, and carbocyclic fused and benzo fused ring systems; additionally, both terms also include systems where a heterocycle ring is fused to an aryl group, as defined herein, or an additional heterocycle group. Examples of heterocycloalkyl groups include aziridinyl, azetidinyl, 1 ,3-benzodioxolyl,
  • dihydroisoindolyl dihydroisoquinolinyl, dihydrocinnolinyl, dihydrobenzodioxinyl, dihydro[l ,3]oxazolo[4,5-b]pyridinyl, benzothiazolyl, dihydroindolyl, dihy-dropyridinyl, 1,3- dioxanyl, 1 ,4-dioxanyl, 1 ,3-dioxolanyl, isoindolinyl, morpholinyl, piperazinyl, pyrrolidinyl, tetrahydropyridinyl, piperidinyl, thiomorpholinyl, and the like.
  • the heterocycloalkyl groups may be optionally substituted unless specifically prohibited.
  • hydrazinyl as used herein, alone or in combination, refers to two amino groups joined by a single bond, i.e., --N--N— .
  • hydroxamic acid refers to ⁇ C(0)ON(R)0(R'), wherein R and R' are as defined herein, or the corresponding "hydroxamate” anion, including any corresponding hydroxamic acid salt.
  • hydroxyalkyl refers to a hydroxy group attached to the parent molecular moiety through an alkyl group.
  • isocyanato refers to a --NCO group.
  • isothiocyanato refers to a— NCS group.
  • linear chain of atoms refers to the longest straight chain of atoms independently selected from carbon, nitrogen, oxygen and sulfur.
  • lower means containing from 1 to and including 6 carbon atoms.
  • mercaptyl as used herein, alone or in combination, refers to an RS— group, where R is as defined herein.
  • nitro refers to — NOi.
  • perhaloalkoxy refers to an alkoxy group where all of the hydrogen atoms are replaced by halogen atoms.
  • perhaloalkyi refers to an alkyl group where all of the hydrogen atoms are replaced by halogen atoms.
  • phosphoamide refers to a phosphate group [(OH) 2 P(0)0-] in which one or more of the hydroxyl groups has been replaced by nitrogen, amino, or amido.
  • phosphonate refers to a group of the form ROP(OR')(OR)0— wherein R and R' are selected from the group consisting of hydrogen, alkyl, acyl, heteroalkyl, aryl, cycloalkyi, heteroaryl, and heterocycloalkyl, any of which may themselves be optionally substituted.
  • Phosphonate includes "phosphate [(OH) 2 P(0)0 ⁇ ] and related phosphoric acid anions which may form salts.
  • sulfonate refers to the --SO 3 H group and its anion as the sulfonic acid is used in salt formation.
  • thia and thio refer to a— S ⁇ group or an ether wherein the oxygen is replaced with sulfur.
  • the oxidized derivatives of the thio group namely sulfmyl and sulfonyl, are included in the definition of thia and thio.
  • thiol as used herein, alone or in combination, refers to an ⁇ SH group.
  • thiocarbonyl when alone includes thioformyl— C(S)H and in combination is a— C(S) ⁇ group.
  • N-thiocarbamyl refers to an ROC(S)NR' ⁇ group, with R and R' as defined herein.
  • O-thiocarbamyl refers to a ⁇ OC(S)NRR', group with R and R' as defined herein.
  • thiocyanato refers to a— CNS group.
  • trihalomethanesulfonamido refers to a X 3 CS(0) R ⁇ group with X is a halogen and R as defined herein.
  • trihalomethanesulfonyl refers to a X3CS(0) 2 ⁇ group where X is a halogen.
  • trimethoxy refers to a X 3 CO-- group where X is a halogen.
  • trimethysilyl as used herein, alone or in combination, refers to a silicone group substituted at its three free valences with groups as l isted herein under the definition of substituted amino. Examples include trimethysilyl, tert-butyldimethylsilyl, triphenylsilyl and the like.
  • any definition herein may be used in combination with any other definition to describe a composite structural group.
  • the trailing element of any such definition is that which attaches to the parent moiety.
  • the composite group alkylamido would represent an alkyl group attached to the parent molecule through an amido group
  • the term alkoxyalkyl would represent an alkoxy group attached to the parent molecule through an alkyl group.
  • null When a group is defined to be “null,” what is meant is that said group is absent.
  • a “null” group occurring between two other groups may also be understood to be a collapsing of flanking groups. For example, if in --(CHi)Gi G 2 G3, the element G 2 were null, said group would become— (CH 2 )GiG3.
  • optionally substituted means the anteceding group may be substituted or unsubstituted.
  • substituents of an "optionally substituted” group may include, without lim itation, one or more substituents independently selected from the following groups or a particular designated set of groups, alone or in combination: lower alkyl, lower alkenyl, lower alkynyl, lower alkanoyl, lower heteroalkyl, lower
  • heterocycloalkyl lower haloalkyl, lower haloalkenyl, lower haloalkynyl, lower perhaloalkyl, lower perhaloalkoxy, lower cycloalkyl, phenyl, aryl, aryloxy, lower alkoxy, lower haloalkoxy, oxo, lower acyloxy, carbonyl, carboxyl, lower aikylcarbonyl, lower carboxyester, lower carboxamido, cyano, hydrogen, halogen, hydroxy, amino, lower alkylamino, arylamino, amido, nitro, thiol, lower alkylthio, lower haloalkylthio, lower perhaloalkylthio, arylthio, sulfonate, sulfonic acid, trisubstituted silyl, N3, SH, SCH 3 , C(0)CH3, CO2CH3, CO2H, pyridinyl, thiophen
  • Two substituents may be joined together to form a fused five-, six-, or seven-membered carbocyclic or heterocyclic ring consisting of zero to three heteroatoms, for example forming methylenedioxy or ethylenedioxy.
  • An optionally substituted group may be unsubstituted (e.g.,— CH2CH3), fully substituted (e.g.,— CF2CF3), monosubstituted (e.g., ⁇ CF CHbF) or substituted at a level anywhere in-between fully substituted and monosubstituted (e.g.,— CH2CF3).
  • substituents are recited without qualification as to substitution, both substituted and unsubstituted forms are encompassed.
  • R or the term R' appearing by itself and without a number designation, unless otherwise defined, refers to a moiety selected from the group consisting of hydrogen, hydroxyl, halogen, alkyl, cycloalkyl, heteroalkyl, aryl, heteroaryl and heterocycloalkyl, any of which may be optionally substituted.
  • R and R' groups should be understood to be optionally substituted as defined herein.
  • Asymmetric centers exist in the compounds of the present invention. These centers are designated by the symbols “R” or “S,” depending on the configuration of substituents around the chiral carbon atom. It should be understood that the invention encompasses all stereochemical isomeric forms, including diastereomeric, enantiomeric, and epimeric forms, as well as d-isomers and 1 -isomers, and mixtures thereof.
  • Individual stereoisomers of compounds can be prepared synthetically from commercially available starting materials which contain chiral centers or by preparation of mixtures of enantiomeric products followed by separation such as conversion to a mixture of diastereomers followed by separation or recrystallization, chromatographic techniques, direct separation of enantiomers on chiral chromatographic columns, or any other appropriate method known in the art.
  • Starting compounds of particular stereochemistry are either commercially available or can be made and resolved by techniques known in the art.
  • the compounds of the present invention may exist as geometric isomers.
  • the present invention includes all cis, trans, syn, anti,
  • compounds may exist as tautomers, including keto-enol tautomers; all tautomeric isomers are provided by this invention.
  • the compounds of the present invention can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. In general, the solvated forms are considered equivalent to the unsolvated forms for the purposes of the present invention.
  • bond refers to a covalent linkage between two atoms, or two moieties when the atoms joined by the bond are considered to be part of larger substructure.
  • a bond may be single, double, or triple unless otherwise specified.
  • a dashed line between two atoms in a drawing of a molecule indicates that an additional bond may be present or absent at that position.
  • disease as used herein is intended to be generally synonymous, and is used interchangeably with, the terms “disorder” and “condition” (as in medical condition), in that all reflect an abnormal condition of the body or of one of its parts that impairs normal functioning and is typically manifested by distinguishing signs and symptoms.
  • combination therapy means the administration of two or more therapeutic agents to treat a therapeutic condition or disorder described in the present disclosure. Such administration encompasses co-administration of these therapeutic agents in a substantially simultaneous manner, such as in a single capsule having a fixed ratio of active ingredients or in multiple, separate capsules for each active ingredient. In addition, such administration also encompasses use of each type of therapeutic agent in a sequential manner. In either case, the treatment regimen will provide beneficial effects of the drug combination in treating the conditions or disorders described herein.
  • the phrase "therapeutically effective” is intended to qualify the amount of active ingredients used in the treatment of a disease or disorder. This amount will achieve the goal of reducing or eliminating the said disease or disorder.
  • treatment of a patient is intended to include prophylaxis.
  • patient means all mammals including humans. Examples of patients include humans, cows, dogs, cats, goats, sheep, pigs, and rabbits. Preferably, the patient is a human.
  • prodrug refers to a compound that is made more active in vivo. Certain of the present compounds can also exist as prodrugs, as described in Hydrolysis in Drug and Prodrug Metabolism: Chemistry, Biochemistry, and Enzymology (Testa, Bernard and Mayer, Joachim M. Wiley- VHCA, Zurich, Switzerland 2003). Prodrugs of the compounds described herein are structurally modified forms of the compound that readily undergo chemical changes under physiological conditions to provide the compound. Additionally, prodrugs can be converted to the compound by chemical or biochemical methods in an ex vivo
  • prodrugs can be slowly converted to a compound when placed in a transdermal patch reservoir with a suitable enzyme or chemical reagent.
  • Prodrugs are often useful because, in some situations, they may be easier to administer than the compound, or parent drug. They may, for instance, be bioavailable by oral administration whereas the parent drug is not.
  • the prodrug may also have improved solubility in pharmaceutical compositions over the parent drug.
  • a wide variety of prodrug derivatives are known in the art, such as those that rely on hydrolytic cleavage or oxidative activation of the prodrug.
  • prodrug a compound which is administered as an ester (the "prodrug"), but then is metabolically hydrolyzed to the carboxylic acid, the active entity.
  • prodrug a compound which is administered as an ester
  • Additional examples include peptidyl derivatives of a compound.
  • therapeutically acceptable prodrug refers to those prodrugs or zwitterions which are suitable for use in contact with the tissues of patients without undue toxicity, irritation, and allergic response, are commensurate with a reasonable benefit/risk ratio, and are effective for their intended use.
  • the compounds of the present invention can exist as therapeutically acceptable salts.
  • the present invention includes compounds listed above in the form of salts, including acid addition salts. Suitable salts include those formed with both organic and inorganic acids. Such acid addition salts will normally be pharmaceutically acceptable. However, salts of non- pharmaceutically acceptable salts may be of utility in the preparation and purification of the compound in question. Basic addition salts may also be formed and be pharmaceutically acceptable.
  • Pharmaceutical Salts Properties, Selection, and Use (Stahl, P. Heinrich. Wiley- VCHA, Zurich, Switzerland, 2002).
  • terapéuticaally acceptable salt represents salts or zwitterionic forms of the compounds of the present invention which are water or oil-soluble or dispersible and therapeutically acceptable as defined herein.
  • the salts can be prepared during the final isolation and purification of the compounds or separately by reacting the appropriate compound in the form of the free base with a suitable acid.
  • Representative acid addition salts include acetate, adipate, alginate, L-ascorbate, aspartate, benzoate,
  • benzenesulfonate (besylate), bisulfate, butyrate, camphorate, camphorsulfonate, citrate, digluconate, formate, fumarate, gentisate, glutarate, glycerophosphate, glycolate, hemisulfate, heptanoate, hexanoate, hippurate, hydrochloride, hydrobromide, hydroiodide, 2- hydroxyethansulfonate (isethionate), lactate, maleate, malonate, DL-mandelate,
  • basic groups in the compounds of the present invention can be quaternized with methyl, ethyl, propyl, and butyl chlorides, bromides, and iodides; dimethyl, diethyl, dibutyl, and diamyl sulfates; decyl, lauryl, myristyl, and steryl chlorides, bromides, and iodides; and benzyl and phenethyl bromides.
  • acids which can be employed to form therapeutically acceptable addition salts include inorganic acids such as hydrochloric, hydrobromic, sulfuric, and phosphoric, and organic acids such as oxalic, maleic, succinic, and citric. Salts can also be formed by coordination of the compounds with an alkali metal or alkaline earth ion.
  • the present invention contemplates sodium, potassium, magnesium, and calcium salts of the compounds disclosed herein, and the like.
  • Basic addition salts can be prepared during the final isolation and purification of the compounds by reacting a carboxyl group with a suitable base such as the hydroxide, carbonate, or bicarbonate of a metal cation or with ammonia or an organic primary, secondary, or tertiary amine.
  • a suitable base such as the hydroxide, carbonate, or bicarbonate of a metal cation or with ammonia or an organic primary, secondary, or tertiary amine.
  • the cations of therapeutically acceptable salts include lithium, sodium, potassium, calcium, magnesium, and aluminum, as well as nontoxic quaternary amine cations such as ammonium, tetramethylammonium, tetraethylammonium,
  • Other representative organic amines useful for the formation of base addition salts include ethylenediamine, ethanolamine, diethanolamine, piperidine, and piperazine.
  • compositions which comprise one or more of certain compounds of the present invention, or one or more pharmaceutically acceptable salts, esters, prodrugs, amides, or solvates thereof, together with one or more pharmaceutically acceptable carriers thereof and optionally one or more other therapeutic ingredients.
  • the carrier(s) must be "acceptable” in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. Proper formulation is dependent upon the route of administration chosen.
  • compositions disclosed herein may be manufactured in any manner known in the art, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or compression processes.
  • compositions include those suitable for oral, parenteral (including
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Typically, these methods include the step of bringing into association a compound of the subject invention or a pharmaceutically acceptable salt, ester, amide, prodrug or solvate thereof ("active ingredient") with the carrier which constitutes one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both and then, if necessary, shaping the product into the desired formulation.
  • Formulations of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a
  • the active ingredient may also be presented as a bolus, electuary or paste.
  • compositions which can be used orally include tablets, push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. Tablets may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with binders, inert diluents, or lubricating, surface active or dispersing agents. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredient therein. All formulations for oral administration should be in dosages suitable for such administration.
  • the push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added.
  • Dragee cores are provided with suitable coatings.
  • concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • the compounds may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in powder form or in a freeze- dried (lyophilized) condition requiring only the addition of the sterile liquid earner, for example, saline or sterile pyrogen-free water, immediately prior to use.
  • sterile liquid earner for example, saline or sterile pyrogen-free water
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
  • Formulations for parenteral administration include aqueous and non-aqueous (oily) sterile injection solutions of the active compounds which may contain antioxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • the compounds may be formulated with suitable polymeric or
  • hydrophobic materials for example as an emulsion in an acceptable oil
  • ion exchange resins for example as an emulsion in an acceptable oil
  • sparingly soluble derivatives for example, as a sparingly soluble salt
  • compositions may take the form of tablets, lozenges, pastilles, or gels formulated in conventional manner.
  • Such compositions may comprise the active ingredient in a flavored basis such as sucrose and acacia or tragacanth.
  • the compounds may also be formulated in rectal compositions such as
  • suppositories or retention enemas e.g., containing conventional suppository bases such as cocoa butter, polyethylene glycol, or other glycerides.
  • Certain compounds of the present invention may be administered topically, that is by non-systemic administration. This includes the application of a compound of the present invention externally to the epidermis or the buccal cavity and the instillation of such a compound into the ear, eye and nose, such that the compound does not significantly enter the blood stream.
  • systemic administration refers to oral, intravenous, intraperitoneal and intramuscular administration.
  • Formulations suitable for topical administration include liquid or semi-liquid preparations suitable for penetration through the skin to the site of inflammation such as gels, liniments, lotions, creams, ointments or pastes, and drops suitable for administration to the eye, ear or nose.
  • the active ingredient for topical administration may comprise, for example, from 0.001% to 10% w/w (by weight) of the formulation. In certain embodiments, the active ingredient may comprise as much as 10% w/w. In other embodiments, it may comprise less than 5% w/w. In certain embodiments, the active ingredient may comprise from 2% w/w to 5% w/w. In other embodiments, it may comprise from 0.1% to 1 % w/w of the formulation.
  • Gels for topical or transdermal administration may comprise, generally, a mixture of volatile solvents, nonvolatile solvents, and water.
  • the volatile solvent component of the buffered solvent system may include lower (C 1 -C6) alkyl alcohols, lower alkyl glycols and lower glycol polymers.
  • the volatile solvent is ethanol.
  • the volatile solvent component is thought to act as a penetration enhancer, while also producing a cooling effect on the skin as it evaporates.
  • the nonvolatile solvent portion of the buffered solvent system is selected from lower alkylene glycols and lower glycol polymers. In certain embodiments, propylene glycol is used.
  • the nonvolatile solvent slows the evaporation of the volatile solvent and reduces the vapor pressure of the buffered solvent system.
  • the amount of this nonvolatile solvent component, as with the volatile solvent, is determined by the pharmaceutical compound or drug being used. When too little of the nonvolatile solvent is in the system, the pharmaceutical compound may crystallize due to evaporation of volatile solvent, while an excess may result in a lack of bioavailability due to poor release of drug from solvent mixture.
  • the buffer component of the buffered solvent system may be selected from any buffer commonly used in the art; in certain embodiments, water is used. A common ratio of ingredients is about 20% of the nonvolatile solvent, about 40% of the volatile solvent, and about 40% water.
  • chelators and gelling agents Appropriate gelling agents can include, but are not limited to, semisynthetic cellulose derivatives (such as hydroxypropylmethylcellulose) and synthetic polymers, and cosmetic agents.
  • Lotions include those suitable for application to the skin or eye.
  • An eye lotion may comprise a sterile aqueous solution optionally containing a bactericide and may be prepared by methods similar to those for the preparation of drops.
  • Lotions or liniments for application to the skin may also include an agent to hasten drying and to cool the skin, such as an alcohol or acetone, and/or a moisturizer such as glycerol or an oil such as castor oil or arachis oil.
  • Creams, ointments or pastes are semi-solid formulations of the active ingredient for external application. They may be made by mixing the active ingredient in finely-divided or powdered form, alone or in solution or suspension in an aqueous or non-aqueous fluid, with the aid of suitable machinery, with a greasy or non-greasy base.
  • the base may comprise hydrocarbons such as hard, soft or liquid paraffin, glycerol, beeswax, a metallic soap; a mucilage; an oil of natural origin such as almond, corn, arachis, castor or olive oil; wool fat or its derivatives or a fatty acid such as stearic or oleic acid together with an alcohol such as propylene glycol or a macrogel.
  • the formulation may incorporate any suitable surface active agent such as an anionic, cationic or non-ionic surfactant such as a sorbitan ester or a polyoxyethylene derivative thereof.
  • Suspending agents such as natural gums, cellulose derivatives or inorganic materials such as silicaceous silicas, and other ingredients such as lanolin, may also be included.
  • Drops may comprise sterile aqueous or oily solutions or suspensions and may be prepared by dissolving the active ingredient in a suitable aqueous solution of a bactericidal and/or fungicidal agent and/or any other suitable preservative, and, in certain embodiments, including a surface active agent.
  • the resulting solution may then be clarified by filtration, transferred to a suitable container which is then sealed and sterilized by autoclaving or maintaining at 98- 100 °C for half an hour.
  • the solution may be sterilized by filtration and transferred to the container by an aseptic technique.
  • bactericidal and fungicidal agents suitable for inclusion in the drops are phenylmercuric nitrate or acetate (0.002%), benzalkonium chloride (0.01%) and chlorhexidine acetate (0.01%).
  • Suitable solvents for the preparation of an oily solution include glycerol, diluted alcohol and propylene glycol.
  • Formulations for topical administration in the mouth include lozenges comprising the active ingredient in a flavored basis such as sucrose and acacia or tragacanth, and pastilles comprising the active ingredient in a basis such as gelatin and glycerin or sucrose and acacia.
  • compounds may be conveniently delivered from an insufflator, nebulizer pressurized packs or other convenient means of delivering an aerosol spray.
  • Pressurized packs may comprise a suitable propellant such as
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • the compounds according to the invention may take the form of a dry powder composition, for example a powder mix of the compound and a suitable powder base such as lactose or starch.
  • the powder composition may be presented in unit dosage form, in for example, capsules, cartridges, gelatin or blister packs from which the powder may be administered with the aid of an inhalator or insufflator.
  • Preferred unit dosage formulations are those containing an effective dose, as herein below recited, or an appropriate fraction thereof, of the active ingredient.
  • formulations described above may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavoring agents.
  • Compounds may be administered orally or via injection at a dose of from 0.1 to 500 mg/kg per day.
  • the dose range for adult humans is generally from 5 mg to 2 g/day.
  • Tablets or other forms of presentation provided in discrete units may conveniently contain an amount of one or more compounds which is effective at such dosage or as a multiple of the same, for instance, units containing 5 mg to 500 mg, usually around 10 mg to 200 mg.
  • the amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
  • the compounds can be administered in various modes, e.g. orally, topically, or by injection.
  • the precise amount of compound administered to a patient will be the
  • the specific dose level for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diets, time of administration, route of administration, rate of excretion, drug combination, the precise disorder being treated, and the severity of the indication or condition being treated. Also, the route of administration may vary depending on the condition and its severity.
  • the compounds described herein may be administered in combination with another therapeutic agent.
  • another therapeutic agent such as a pharmaceutically acceptable salt, ester, or prodrug thereof.
  • the therapeutic effectiveness of one of the compounds described herein may be enhanced by administration of an adjuvant (i.e., by itself the adjuvant may only have minimal therapeutic benefit, but in combination with another therapeutic agent, the overall therapeutic benefit to the patient is enhanced).
  • the benefit of experienced by a patient may be increased by administering one of the compounds described herein with another therapeutic agent (which also includes a therapeutic regimen) that also has therapeutic benefit.
  • another therapeutic agent which also includes a therapeutic regimen
  • increased therapeutic benefit may result by also providing the patient with another therapeutic agent for diabetes.
  • the overall benefit experienced by the patient may simply be additive of the two therapeutic agents or the patient may experience a synergistic benefit.
  • the multiple therapeutic agents may be administered in any order or even simultaneously. If simultaneously, the multiple therapeutic agents may be provided in a single, unified form, or in multiple forms (by way of example only, either as a single pill or as two separate pills). One of the therapeutic agents may be given in multiple doses, or both may be given as multiple doses. If not simultaneous, the timing between the multiple doses may be any duration of time ranging from a few minutes to four weeks.
  • APJ apelin receptor
  • Apelin is the endogenous peptide ligand for the G-protein coupled receptor (GPCR) APJ (angiotensin II receptor-like 1 , AGTRL-1 and APLNR). Until the discovery of apelin, APJ was an orphan GPCR. APJ is coupled to Gai, and has been shown in cell culture to inhibit adenylate cyclase. The APJ gene encodes a receptor that most closely resembles the angiotensin receptor ATI ; however, t he APJ receptor does not bind angiotensin.
  • GPCR G-protein coupled receptor
  • apelin reduces the extent of atherosclerotic lesions in ApoE /- mice , and opposes the development of abdominal aortic aneurysms. Additional studies have also revealed that APJ forms a heterodimer with the Ang II receptor ATI , and that this complex facilitates antagonism of Ang II signaling by apelin. Despite these exciting results, there remains a multitude of unanswered questions regarding the role of apelin and APJ in the physiology and pathology.
  • the series of molecules disclosed herein are potent (EC50 ⁇ 0.01 u for most potent cases), and exhibit dose dependent agonist of the apelin receptor.
  • the compounds disclosed herein are selective for the apelin receptor, and have been shown to be highly specific for the apelin receptor versus the angiotensin II receptor (ATI), the most closely related receptor.
  • the compounds disclosed herein are low in molecular weight ( ⁇ 500), and are typically lipophilic.
  • apelin receptor agonists would be useful as therapeutic regimens for chronic kidney disease.
  • vasodilatation by apelins is reduced by two thirds during nitric oxide synthase inhibition but is unaffected by prostacyclin inhibition.
  • the apelin receptor has been linked to direct cardiac actions.
  • exogenous apelin increases contractility at subnanomolar concentrations in atrial strips (Maguire et al. Hypertension 2009;54:598-604) and whole rat hearts (Szokodi et al. Circ. Res. 2002;91 , 434-440).
  • acute apelin infusion increases myocardial contractility independently of its effects on loading conditions.
  • Apelin and the APJ receptor are both expressed also in the kidney and many areas of the brain. Synthesis in certain brain regions involved in fluid homeostasis are regulated by vasopressin. To the contrary, intracerebral injection of apelin directly inhibits vasopressin release leading to a 40% reduction in plasma vasopressin concentrations (Reaux-Le Goazigo et al. Endocrinology 2004; 145 :4392- 4400).
  • a link of apelins to metabolic syndrome is suggested by pre-clinical data.
  • Apelins are produced by adipose tissue and may influence glucose and lipid metabolism as adipocytokines (Boucher et al. Endocrinology 2005; 146: 1764-1771 ).
  • Acute intravenous administration of 'pyf-apelin- 13 stimulates glucose utilization in normal and obese insulin- resistant mice (Dray C et al. Cell Metab 2008;8:437-445). These acute effects were explained by a direct effect of 'pyr-apelin- 13 on glucose uptake into skeletal muscle.
  • Mice deficient for the apelins have reduced insulin sensitivity which can be corrected by sub-chronic supplementation with apelin via minipumps.
  • apelin receptor agonists demonstrate a unique hemodynamic profile in enhancing myocardial contractility without inducing left ventricular hypertrophy.
  • ventricular pre- and afterload is reduced by reduced peripheral resistance.
  • apelin increases contractility at least to the same extent in the failing compared to normal myocardium (Dai et al. Eur J Pharmacol
  • apelin receptor agonists may be administered with the reperfusion solution directly after acute myocardial infarction and thereby display potential benefits in both restoring cardiac survival and function.
  • Another application, especially of oral bioavailable small molecule apelin receptor agonists could be to start in a patient with an acute myocardial infarction with an intravenous formulation during reperfusion and continue later, e.g., outside the clinic, with an oral bioavailable formulation of the same drug component.
  • intravenous or oral administration of apelin receptor agonists could be envisaged in patients with acute heart failure.
  • Agonists of the apelin receptor may also represent a novel class of antihypertensive agents.
  • administration of apelin peptides lowers blood pressure, greatly enhanced in hypertensive animals compared with normotensive controls.
  • modest but significant effects on blood pressure lowering could be demonstrated in normotensive middle-aged subjects.
  • intravenously applied apelin peptides lower blood pressure stronger in hypertensive patient populations, similar to the situation in normotensive vs. hypertensive rats, needs to be evaluated.
  • Application of apelin peptides in hypertensive patients is strongly limited by the need of intravenous administration route.
  • small molecule apelin receptor agonists as claimed in this patent application may have a much wider application in these patients due to better oral bioavailability.
  • Apelin receptor agonists appear to have beneficial effects on additional vascular based diseases.
  • apelin infusion inhibits atherosclerosis progress and completely abrogates angiotensin II- accelerated detrimental effects independent of blood pressure (Chun et al. J Clin Invest 2008; 1 1 8:3343- 3354).
  • double knockout mice deficient in for the apelin receptor ligand and
  • Apolipoprotein-E accelerated atherosclerosis could be observed compared versus single Apolipoprotein-E-knockout. It needs to be mentioned, that also pro-atherosclerotic effects of the apelin receptor have been described in a combined mice knockout-model of the apelin receptor and apolipoprotein-E ApoE (Hashimoto et al. Am J Pathol 2007: 108: 1432-1438). Overall these results are difficult to reconcile: Most probably very different fat feeding regimens or different genetic backgrounds and so called off-target genetic effects best explain the observed differences.
  • apelin treatment resulted in reduced aneurysm by 50% in a mouse model of abdominal aortic aneurysms (Leeper et al. Am J Physiol Heart Circ Physiol 2009;296:H 1329- 1335), an effect explained by the authors by a direct anti-inflammatory effect within the vessel wall.
  • apelin receptor agonists may play an important role in maturation of newly formed blood vessels.
  • Bood 2010; 1 15;3 166-3174 recently described in a model of vascular remodeling after hind limb ischemia in mice, that apelins induce the maturation into enlarged and non-leaky blood vessels for functional recovery.
  • VEGF vascular endothelial growth factor
  • apelins cause nitric oxide-mediated vasodilatation in forearm resistance vessels of healthy subjects.
  • the role of apelin receptor agonists in preventing human vascular disease merits further investigations. These investigations will be strongly facilitated by small molecule apelin receptor agonists, as claimed in this patent application, because the oral bioavailability allows for much easier chronic administration routes.
  • apelin receptor agonists may provide additional benefits.
  • Apelins are produced also by adipose tissue and influence glucose and lipid metabolism as adipocytokines. Mice with no apelin receptor ligands have reduced insulin sensitivity which can be corrected by the administration of exogenous apelin.
  • Acute and sub-chronic positive effects of apelins on glucose utilizations following a glucose load have been described in insulin- resistant animal strains. Although the translation of these effects to man needs to be performed, apelin receptor agonists may offer additional therapeutic options especially in insulin-resistant patients, insufficiently dealing with increased plasma glucose load in metabolic syndrome and diabetes.
  • APJ modulators in particular in view of safety and selectivity.
  • the compounds disclosed herein are selective for the apelin receptor, and have been shown to be highly specific for the apelin receptor versus the angiotensin II receptor (ATI ), the most closely related receptor. Broad selectivity profiling experiments revealed no significant off target binding.
  • the compounds disclosed herein show favorable in vitro absorption, distribution, metabolism, and excretion (ADME) properties and excellent in vivo pharmacokinetics.
  • the compounds of the present disclosure are contemplated to be useful as novel therapies for the treatment for a range of cardiovascular, renal and metabolic conditions.
  • Step 2 The mixture of 2-(4-methoxybenzoyl)-N-(2- methoxyphenyl)hydrazinecarbothioamide ( 100 mg, 0.30 mmol) and K 2 CO 3 (170 mg, 1.23 mmol) in water (2.0 mL) was heated to reflux. The reaction was monitored by LC-MS. When it was completed, the solution was cooled to room temperature, washed with with ⁇ 2 ⁇ 3 (4 mL), and the aqueous layer was acidified with IN HC1.
  • Step 1 Step 1 :
  • Step 2 [0272] To a solution of 4-chloromethyl-2-phenyl-thiazole (50.0 mg, 0.240 mmol) in acetone (10 mL) was added 4-(2-methoxyphenyl)-5-(2-thienyl)-l,2,4-triazole-3-thiol (83.9 mg, 0.290 mmol) and K 2 C0 3 (100 mg, 0.720 mmol). Then the mixture was stirred at 60 °C for 2 hrs. After cooling to room temperature, the reactant was filtered. The filtrate was evaporated in vacuum to dryness. The residue was diluted with EA (10 mL).
  • Biological activity was determined in a cell-based assay. This assay detects activation of the receptor by reporting on intracellular cyclic adenosine monophosphate (cAMP) level.
  • Chinese hamster ovary cells engineered to express a stably integrated human apelin receptor (APJ, AGTRL-1 , APLNR) were plated into the wells of a microtiter plate. Cells were stimulated with forskolin to increase intracellular cAMP.
  • compounds were added at a range of final concentrations 0.0-100 ⁇ , in DMSO (not to exceed a final DMSO concentration of > 1 % v/v), and allowed to incubate with the cells for 0.5h.
  • DMSO not to exceed a final DMSO concentration of > 1 % v/v
  • Compound potency is reported as the effective concentration required to effect 50% (EC50) of the control (apelin- 13, 1 nM) response in micromolar ( ⁇ ) units.
  • Compound efficacy (% resp) is reported as a percentage of the maximal apelin- 13 response at I nM (control). Potencies were calculated by fitting a point logistic curve to a concentration response curve consisting of at least 10 data points spanning the effective range of the compounds in a cell-based assay that measures cAMP. [0439] Table 1.

Abstract

Provided herein are small molecule agonists of the apelin receptor for the treatment of disease. The compounds disclosed herein are useful for the treatment of a range of cardiovascular, renal and metabolic conditions.

Description

AGONISTS OF THE APELIN RECEPTOR AND METHODS OF USE THEREOF
BACKGROUND OF THE INVENTION
FIELD OF THE INVENTION
[0001] This invention relates generally to small molecule agonists of the apelin receptor (APJ) and, more specifically, to compounds for the treatment of apelin receptor-mediated diseases and disorders.
BACKGROUND INFORMATION
[0002] G protein-coupled receptors (GPCRs) are activated by a plethora of molecules including neuropeptides, polypeptide hormones and non-peptides such as biogenic amines, lipids, nucleotides and ions. They are classically composed of seven membrane-spanning domains and constitute one of the largest and most diverse gene families in the mammalian genome. Some novel GPCRs do not have obvious endogenous ligands and are termed orphan receptors, a number of which appear to be constitutively active. The cognate ligands for some of these orphan GPCRs have been identified, often based on the cellular and tissue distributions of the orphan GPCRs and occasionally using 'reverse pharmacology' where orphan GPCRs have been used to isolate novel endogenous substances. The human apelin receptor (APJ, gene symbol APLNR) first identified in 1993 (O'Dowd et al. Gene
1993; 136:355-60) is one such GPCR whose endogenous ligand, apelin, has been described.
[0003] Both APJ and apelin have been implicated as the key mediators of physiological responses to multiple homeostatic perturbations, including cardiovascular control, water balance, hypothalamic-pituitary-adrenal (HPA) axis regulation and metabolic homeostasis. Homeostatic stability is critical in mammalian organisms, and knowledge as to how this vital function is regulated and how this mechanism can go wrong in pathological conditions is still limited.
[0004] APJ was first identified as an orphan GPCR, with closest identity to the angiotensin II (Ang II) receptor, type ATia. APJ remained an orphan receptor until 1998 when a 36-amino acid peptide termed apelin, for APJ endogenous ligand was identified. In the ensuing years, the receptor was deorphanised when its cognate ligand, apelin, was isolated from bovine stomach extracts. Recently, the apelinergic system has been shown to be critically involved in multiple homeostatic processes.
[0005] The protein structure of APJ is typical of a GPCR, containing seven hydrophobic transmembrane domains, with consensus sites for phosphorylation by protein kinase A (P A), palmitoylation and glycosylation. The N-terminal glycosylation of GPCRs has been implicated in receptor expression, stability, correct folding of the nascent protein and ligand binding. Furthermore, the palmitoylation of the C-terminal tail has been reported to play a role in membrane association and, combined with receptor phosphorylation, these fatty acid modifications can influence the internalization, dimerization and ligand binding of a GPCR. Structural studies on APJ have determined that amino acids in both the N-terminal (e.g., Asp23 and Gkf °) and C-terminal portions of the receptor are required for internalization.
[0006] The gene encoding human apelin, termed APLN, is located on chromosome Xq25- 26.1 and possesses one intron within its open reading frame of ~6kb. In rat and mouse, the genes are termed Apln and are located at chromosomal locations Xq35 and XA3.2 respectively. The core promoter regions of these genes have been identified as -207/- 1 and -100/+ 74 bp in rats and humans respectively. Similar to APJ, a CAAT box, but no TATA box, sequence is present in the rat and human promoter regions. Furthermore, rat and human preproapelin cDNAs do not have a classical Kozak consensus sequence surrounding the initiating methionine codon.
[0007] Human and bovine APLN cDNA sequences encode a 77-amino acid preproprotein (preproapelin) containing a hydrophobic rich N-terminal region, likely to be a secretory signal sequence. Bovine, human, rat and mouse preproapelin precursors have 76-95% homology and appear to exist endogenously as a dimeric protein, as a consequence of disulfide bridges formed between cysteine residues.
[0008] There are several mature forms of the apelin peptide. As the sequence of the purified peptide corresponded to the 36 C-terminal amino acids of the preproapelin protein, it was predicted that apelin-36 would constitute a mature form of the peptide. Additionally, as the C-terminal portion of preproapelin also contained lysine (Lys, K) and arginine (Arg, R) residues, and given their potential as sites for proteolytic cleavage, the existence of apelin-17 and apelin-13 peptides was predicted, along with a pyroglutamylated form of apelin- 13 ((Pyr')apelin-13). These mature forms of apelin lack cysteine residues and are probably only present in monomeric form. The likely secondary structures of apelin-36 and apelin- 13 have been determined in aqueous solution, indicating that both possess an unordered structure. The amino acid sequence homology of the mature apelin-36 peptide is more conserved between species than that of preproapelin, with 86-100% homology between bovine, human, rat and mouse amino acid sequences, while the 23 C-terminal amino acids have 100% homology between species, suggesting an important physiological role. [0009] Although APJ does not bind Ang II (O'Dowd et al. 1993), apelin- 13 shares a limited homology (four amino acids) with the vasoconstrictive peptide. Moreover, Ang I- converting enzyme 2 (ACE2), which catalyzes the C-terminal dipeptide cleavage of Ang I to Ang II, or Ang II to Ang 1-7, also acts on apelin-13 with a high catalytic efficiency, removing the C-terminal phenylalanine (Phe, F) residue. However, this cleavage may not inactivate the peptide, as the apelin isoform K 16P, which lacks the terminal Phe, while ineffective at inducing receptor internalization or regulating blood pressure (BP) (effects associated with the full peptide), still binds to APJ and inhibits forskol in-stimulated cAMP production.
[0010] Although it is clear that APJ and apelin mRNAs and proteins are widely distributed in the CNS and peripheral tissues, whether the levels of mRNAs present in most of the regions of the brain and tissues are functionally relevant is not yet known.
(0011] Early studies of the expression of APJ mRNA by northern blot and quantitative PCR (qPCR) analyses have revealed strongest signals in the human caudate nucleus, corpus callosum, hippocampus, substantia nigra, subthalamic nucleus, medulla and spinal cord. Recently, the expression of APJ mRNA has also been demonstrated in the human cortex and hippocampus using a sensitive GPCR gene array profiling method - interestingly, APJ transcripts have also been detected in human bone marrow stromal cell lines. Transcriptomic analysis of multiple brain regions of human donors has revealed a widespread central expression of APJ mRNA with high levels in samples including the hippocampus (e.g., CA4 region), habenular nuclei, paraventricular nucleus (PVN) of the thalamus, supraoptic nucleus (SON) of the hypothalamus and various hindbrain structures. The salient feature of these studies is that APJ has been reported to have a widespread central distribution; although the function of APJ in the majority of brain regions is unknown, foremost among those regions probably important from a functional perspective include the PVN and SON of the hypothalamus.
[0012] In the periphery, the expression of human APJ mRNA was originally reported to be strongest in the spleen, with less expression being reported for the small intestine, colonic mucosa and ovary. A broader qPCR study has also reported strongest expression in the spleen, with high levels also being reported to be present in the placenta and weaker levels in the lung, stomach and intestine. (Pyr')apelin- 13-binding sites can be found within the media and intimal layers of muscular arteries and large elastic arteries and veins, while in the lung, apelin-binding sites have a predominantly vascular localization. Furthermore, APJ distribution in cardiovascular tissues, as demonstrated by immunohistochemistry'(IHC), indicates APJ to be present in ventricular cardiomyocytes, vascular smooth muscle cells (VS Cs) and intramyocardial endothelial cells.
[0013] APJ binds numerous apelin isoforms and signals through various G proteins to a variety of signaling pathways to culminate in different patterns of activation and
desensitization that may be tissue- and cell type-specific. Recently, APJ has also been reported to heterodimerize with other GPCRs and to signal in the absence of an endogenous ligand.
[0014] The C-terminal region of the apelin peptide may be responsible for its overall biological activity. N-terminal deletions of apelin-17 reveal that the 12 C-terminal amino acids may be the core requirements for the internalization and biological potency of APJ. Apelin- 17 induces the internalization of APJ, which decreases with every N-terminal deletion to apelin- 12, while the deletion of the terminal F amino acid results in a peptide that no longer internalizes APJ or affects arterial BP. The N-terminal residues within the RPRL motif (residues 2-5) of apelin-13 are critical for functional potency, and the C-terminal sequence KGPM (residues 8-1 1 ) is important for binding activity and for internalization. In contrast, the five N-terminal and two C-terminal amino acids of apelin- 17 are not required for binding of the peptide to APJ or activation of receptor signaling (e.g., cAMP production). Although this may indicate a possible dissociation between the conformational states of the receptor responsible for receptor signaling and internalization, it is also possible that different ligand isoforms may induce differential receptor trafficking and signaling. These studies provide information on the structural importance of key apelin residues critical for efficient binding, activity and internalization, which have proved significant in the design and synthesis of apelin analogs.
[0015] Although progress has been made in recent years in clarifying the physiological significance of apelin/APJ, much remains to be discovered about the expression of the apelinergic system and precisely how it affects numerous physiological functions. Since the discovery of the apelin ligand, both apelin and APJ have been implicated as key regulators of central and peripheral responses to multiple homeostatic perturbations. These include playing pivotal roles in the regulation of cardiovascular function, angiogenesis, fluid homeostasis and energy metabolism and acting as neuroendocrine modulators of the HPA axis responses to stress. It is becoming apparent that the apelinergic system may play a pathophysiological role within many of these regulatory systems. [0016] The central rriRNA expression of preproapelin in regions of the hippocampus, hypothalamus, thalamus and midbrain shares a distribution pattern, as shown by ISHH, similar to that of angiotensinogen (Ang II precursor). Ang II is part of the rennin-angiotensin system (RAS), which controls extracellular fluid volume and arterial vasoconstriction, thereby regulating mean arterial blood pressure (MABP). The central actions of the RAS include the regulation of drinking behavior, salt appetite and VP secretion. Importantly, the RAS plays a critical role in the pathogenesis of heart failure. Interestingly, apelin exerts many physiological effects that appear to oppose those exerted by Ang II. More recently, apelin has been shown to block many Ang II-initiated processes, perhaps partly by dimerization between APJ and AT,.
[0017] It is clear that apelin has both peripheral and central cardiovascular effects.
However, experiments carried out in animal models have yielded conflicting results about the role of peripheral apelin in the regulation of vascular tone, with both pressor and depressor responses being described. In anaesthetized intact rats, the overall effect of peripherally administered apelin is the reduction of MABP. This hypotensive action is blocked by the NOS inhibitor L-NAME, indicating a nitric oxide-mediated pathway. In conscious rats, the effect is even less clear, with both increases and decreases in MABP being reported.
Discrepancies among these reports may reflect the conscious state of the animal or the different apelin isoforms used in these studies; it is unknown which specific apelin peptide may be responsible for the (patho)-physiological roles of apelin. Further evidence that APJ plays a role in the regulation of BP comes from a study on mice with a global deletion of APJ, where a transient decrease in systolic BP observed in conscious wild-type (WT) mice following i.p. injection of (Pyr')apelin- 13 is abolished in APJ KO mice. However, while peripheral apelin is a vasodilator in the human saphenous vein, in vessels denuded of endothelium, apelin acts as a vasoconstrictor. Therefore, peripheral apelin may act as an antihypertensive factor, and sensitivity to the peripheral administration of apelin might be altered in hypertensive disease.
[0018] Additionally, the apelinergic system has an important role in cardiac function. In the isolated rat heart, infusion of apelin- 16 induces a potent dose-dependent positive inotropic effect, with an EC¾, of 40-125 pM in humans and—33 pM in rats, an effect also observed in the failing heart. In mice, administration of apelin increases myocardial contraction while reducing cardiac preload and afterload, without causing hypertrophy. Furthermore, apelin increases the shortening of sarcomeres in cardiomyocytes, an effect that is impaired in isolated ventricular myocytes from apelin and APJ KO mice. Apelin KO mice have an impaired response to cardiac pressure overload, thus suggesting a role for apelin/APJ in the sustainability and amplification of the cardiac response to stress. There is also evidence for a role in essential hypertension (EHT) as circulating levels of apelin- 12 are decreased in patients with EHT. Functionally, the apelinergic system plays a role in the Cripto signaling pathway (which stimulates signaling by the transforming growth factor Nodal or growth/differentiation factors 1 and 3, via activin type IB and type IIB receptors) in mammalian cardiac myogenesis.
[0019] Cardiovascular development defects have been reported in APJ KO mice, where a loss of homozygous mutants has been described, but not in apelin KO mice, indicating possible ligand-independent effects of the receptor. This effect may perhaps be explained by the recent report that APJ signals independently of apelin in response to cardiac mechanical stretch. APJ KO embryos at El 0.5, when lethality begins, have poorly developed vasculature of the yolk sac, delayed formation of the atrioventricular cushion and unusually formed cardinal veins and dorsal aorta. APJ KOs that survive do not reveal any apparent morphological differences; however, they have decreased vascular smooth muscle layer recruitment and myocardial defects including thinning of the myocardium, enlarged right ventricles and ventricular septal defects, suggesting an involvement of apelin/APJ signaling in cardiovascular development.
[0020] Apelin appears to have a role to play in the pathophysiology of the cardiovascular system - it has been implicated in vascular diseases, heart failure, and ischemia and subsequent reperfusion. In vascular diseases, the expression of apelin is up-regulated in the atherosclerosis of human coronary artery. Yet its role is undetermined, as conflicting evidence has been found in KO studies, indicating both antagonistic and inducing roles for apelin in atherosclerotic formation. During heart failure, plasma apelin levels rise in the early stages of disease and stabilize or lower as the condition develops. However, APJ mRNA is decreased in the weakened and enlarged heart of humans with idiopathic dilated
cardiomyopath. Apelin may have a cardioprotective role in hypoxia and ischemia, where the cardiac levels of apelin and APJ respectively are increased. Apelin may also play a protective a role in ischemia /reperfusion injury, although the method of signaling appears to be independent of the characteristic myocardial kinase cascade, termed the reperfusion injury salvage kinase pathway. Post-infarct treatment with (Pyr')apelin- 13 reduces infarct size and increases HR, with a long-term antioxidant cardioprotective action. [0021 ] Apelin is an angiogenic factor and mitogen of endothelial cells. Significantly, apelin is required for the normal development of frog heart and formation of murine blood vessels. Additionally, the development of the retinal vasculature is stunted in apelin KO mice, and apelin is necessary for hypoxia-induced retinal angiogenesis, and is also involved in non- neovascular remodeling of the retina.
[0022] The apelinergic system has been implicated in tumor neoangiogenesis. In brain tumors, the expression of apelin and APJ is up-regulated in microvascular proliferations, while tumor cell lines overexpressing apelin show increased growth. The pathophysiological effects of apelin in angiogenesis have also been reported for the liver, where the apelinergic system is a factor in portosystemic collaterization and splanchnic neovascularization in portal hypotensive rats as well as in neovascularization during liver cirrhosis. However, apelin may have therapeutic effects in ischemia recovery due to vessel regeneration and endothelial proliferation and blood vessel diameter regulation. These findings indicate that apelin is a crucial factor for angiogenesis and that there may be therapeutic potential in both the disruption of its signaling (e.g., tumors) and the stimulation of APJ expression (e.g., ischemia recovery).
[0023] The detection of APJ mRNA expression in areas of the brain critical for the control of fluid homeostasis led to the hypothesis that apelin may play a role in the regulation of body fluid balance. VP, along with OT, is synthesized primarily in the neurones of the mPVN and SON, which project to the posterior pituitary and release the peptides into the systemic circulation. The predominant endocrine function of VP from this source is to increase water permeability in the renal collecting duct cells, thereby allowing the retention of water.
[0024] The regulatory actions of apelin on thirst and drinking behavior have been reported. In water-replete animals, a significant increase in water intake is observed following i.p. or i.c.v. injection of apelin, whereas in other studies apelin has been reported to reduce water intake post i.c.v. injection or to have no effect. Additionally, in water-deprived rats, an inhibitory effect or lack of any effect of apelin on drinking behavior is observed, while in apelin KO mice, the dehydration-induced drinking response is comparable to that observed in WT mice. The expression of apelin and APJ mRNAs, and labelling of apelin-immunoreactive magnocellular cells, are increased by dehydration, while the labelling of VP-immunoreactive cells decreases, implying the differential regulation of these peptides in response to dehydration. Recently, however, abnormal fluid homeostasis has been demonstrated in APJ KO mice, manifested by a decrease in drinking behavior and an inability to concentrate urine to levels observed in controls during water deprivation, suggesting an antidiuretic effect of apelin in vivo. However, in lactating rats, apelin induces diuresis and has direct effects on renal vasculature. APJ is also necessary in dehydration-induced signaling in the subfornical organ, implicating the apelinergic pathway in responses to hyperosmotic stimuli.
[0025] A number of studies have pointed out an emerging involvement of apelin in energy metabolism and a role for adipocyte-derived apelin in the (patho)-physiology of obesity has been reported. Both apelin and APJ mRNAs are present in mouse, human and rat adipose tissue, and their levels increase in adipose tissue and plasma with obesity. This highlights APJ as an intriguing therapeutic target for metabolic disorders. However, the expression of plasma apelin is increased only in obese humans and in mouse models of obesity associated with hyperinsulinemia, indicating that obesity or high-fat feeding may not be the main cause for the rise in the expression of apelin, and implying a close relationship between apelin and insulin both in vivo and in vitro. Insulin directly acts on adipocytes in vitro to stimulate the production of apelin, and the expression of apelin mRNA is down-regulated in the adipocytes of mice treated with the β-cell toxin streptozotocin, which leads to a fall in plasma insulin levels. In mice, nutritional status influences apelin levels in vivo - fasting inhibits plasma levels, which are then restored by re-feeding - thus strengthening the implication that insulin regulates apelin gene expression and secretion. Additionally, apelin, perhaps through APJ expressed in pancreatic islet β-cells, regulates the secretion of insulin - apelin inhibits glucose-stimulated insulin secretion in vivo in mice and in isolated islets of Langerhans in vitro. Interestingly, in a recent study, apelin has been shown to alleviate diabetes-induced reduction of pancreatic islet mass and to improve the insulin content of pancreatic islets in type 1 diabetic mice.
[0026] Apelin may have a positive effect in the metabolic syndrome (a combination of risk factors that when occurring together increase the risk of coronary artery disease, stroke and type 2 diabetes (T2D)). Apelin KO mice have reduced insulin sensitivity, are glucose intolerant and are hyperinsulinemic. The peripheral administration of apelin reduces peak plasma glucose concentrations by increasing glucose uptake in skeletal muscle and adipose tissue and improves insulin sensitivity in both apelin KO and obese high-fat diet fed mice, with the insulin-sensitizing effects continuing for up to 4 weeks, with no tolerance to the actions of apelin. Apelin increases glucose uptake, both in vitro and in vivo, through both insulin-dependent and -independent pathways. Apelin may also decrease body adiposity, independently of altered food intake, by increasing energy expenditure through the activation of mitochondrial uncoupling proteins 1 and 3. Clinical studies have shown a promising therapeutic value for apelin, as apelin displays beneficial glucose-lowering effects in human adipose tissue and plasma apelin levels correlate with glucose and HbA l c levels. Apelin is linked to the pathogenesis of T2D - plasma apelin concentrations are increased in insulin- resistant patients, in type T2D patients and in morbidly obese T2D individuals, perhaps indicating a compensatory role of apelin in the reduction of insulin resistance. However, conversely, plasma apelin levels are reduced in newly diagnosed T2D patients and increased in T2D patients and obese non-diabetic individuals. The increased expression of apelin in plasma and adipose tissue of obese individuals can, however, be reversed by a hypocaloric diet. As a result of such studies, similarities between the function of apelin and that of insulin, and a link between this adipokine and glucose homeostasis, have been hypothesized.
[0027] As has been noted previously, APJ is localized in the hypothalamic pPVN and the anterior pituitary gland, key areas involved in the stress response. Apelin mRNA is also present in these areas, co-localizing with VP in the mPVN, SON and pituitary. Additionally, apelin immunostaining of cell bodies and fibers is highest in the hypothalamus, with large numbers of apelin-positive cell bodies present in the PVN and SON. The presence of APJ and apelin in VP- and CRH-containing hypothalamic nuclei, which are pivotal to the HPA axis responses to stress, suggests a role for apelin/APJ in neuroadenohypophysial hormone release.
[0028] A role for apelin in the regulation of the HPA axis responses to stress is supported by studies showing that central administration of (Pyr')apelin-13 increases the expression of c-fos, an indicator of neuronal activity, in the PVN. Furthermore, administration of apelin-13 stimulates the release of CRH and VP from hypothalamic extracts in vitro, effects consistent with stimulation of the stress axis. APJ mRNA levels increase in the PVN in response to acute and chronic stress and following adrenalectomy, implying negative regulation of the expression of APJ mRNA by glucocorticoids. Additionally, dexamethasone, a glucocorticoid agonist, decreases apelin mRNA levels in 3T3-L1 mouse adipocytes.
[0029] Apelin may potentially stimulate the secretion of ACTH either directly at the level of the pituitary corticotroph or via an indirect action on the hypothalamus involving the release of both VP and CRH. Consistent with the expression of apelin and APJ in anterior pituitary corticotrophs, administration of apelin- 17 directly increases the release of ACTH, while also augmenting K*-stimulated ACTH release, in an ex vivo perfusion system of anterior pituitary glands, suggesting possible autocrine or paracrine functions for apelin in this tissue. Central administration of (Pyr')apelin-13 in rats also increases plasma ACTH and CORT levels while decreasing prolactin, luteinizing hormone and follicle-stimulating hormone levels. However, increases in plasma ACTH and CORT levels observed after i.e. v. administration of (Pyr')apelin- 13 in mice are reduced to control levels by pre-treatment with the CRH receptor antagonist a-helical CRH.,_„, while (Pyr')apelin- 13-mediated increases in plasma ACTH levels are abolished in VP VI b receptor KO mice, indicating that apelin also modulates the release of ACTH via an indirect action on the hypothalamus involving both CRH- and VP-dependent mechanisms. Recently, using APJ KO mice, APJ has been shown to play a regulatory role in the modulation of the HPA axis responses to some acute stressors including LPS challenge (an immune stressor), insulin-induced hypoglycemia (a metabolic stressor) and forced swim (a physical/psychological stressor). These studies suggest that other peptides cannot compensate for the loss of APJ to directly, or indirectly, induce the release of ACTH in response to stress. Thus, the integration of neurobehavioral responses to stress may be more complicated than previously envisioned, with apelin/APJ exerting a pivotal neuroregulatory role.
[0030] Apelin was first isolated from stomach extracts, and studies on the actions of apelin in the gastrointestinal system have found functional, and possible cell survival, roles. In the gastrointestinal system, apelin/APJ may be regulators of hormone and gastric acid secretion. Apelin/APJ may also have a direct effect on vascular smooth muscle, including vasoconstriction, which may affect renal glomerular hemodynamic function in the rat kidney. Some studies have also proposed an immunological role for apelin as it reduces the production of cytokines in mouse spleen cells, suggesting that apelin may modulate neonatal immune responses through rodent and bovine colostrum and milk. APJ is also a co-receptor of HIV entry into target cells, an action that is blocked by apelin. APJ may contribute to HIV- 1 infection and pathogenesis in CNS-based cells as viral envelope proteins can mediate fusion with APJ-positive, cluster of differentiation 4 (CD4)-negative cells, provided that CD4 is added in trans, and HIV can infect APJ-expressing cells despite their CD4 status. Other possible roles for apelin and APJ in the rodent CNS include antinociception, enhancement of depressive behavior, and facilitation of passive avoidance learning. Apelin may also have a role in neuroprotection, as apelin pre-treatment protects hippocampal neurones against N- methyl-D-aspartate (NMDA) receptor-mediated excitotoxic injury, possibly via the phosphorylation of Akt and ERK1 /2, and prevents apoptosis in cultured mouse cortical neurones.
[0031 ] Furthermore, apelin and APJ are expressed in osteoblasts where they may induce cell proliferation and promote survival; however, an increase in bone mass can be observed in apeiin KO mice. Recently, apeiin has been reported to have a potential role in the pathophysiology of osteoarthritis (OA), as apeiin is present in synovial fluid, and OA patients have elevated plasma apeiin concentrations. Blood plasma levels of apeiin are reduced in patients with polycystic ovary syndrome, consistent with the role played by apelin/APJ in metabolic disturbances such as insulin resistance.
[0032] Elevated levels of apeiin have been detected in many pathological states or disease processes, such as heart disease, atherosclerosis, tumor angiogenesis and diabetes. However, in many systems, apeiin has been shown to have positive effects, for example in the cardiovascular system, where it has a cardioprotective effect. This has led to speculation that apeiin and APJ could be beneficial targets for therapeutic strategies for a number of diseases and disorders.
[0033] To date, there are few reports of selective small molecule apeiin receptor agonists, and thus far none of the reported agonists have favorable pharmacokinetic profiles. There is a need, therefore, for potent compounds that target and exhibit dose dependent agonism of the apeiin receptor.
SUMMARY OF THE INVENTION
[0034] The present invention is based on the seminal discovery of a series of potent small molecule agonists of the apeiin receptor, which are useful for the treatment of diseases including heart failure, chronic kidney disease, hypertension, and metabolic disorders such as insulin resistance/diabetes and obesity. The compounds disclosed herein are highly specific for the apeiin receptor versus the angiotensin II receptor (ATI).
[0035] Provided herein are compounds of structural Formula I, or a pharmaceutically acceptable salt, polymorph, solvate, tautomer, or N-oxide thereof:
Figure imgf000012_0001
(I) [0036] wherein:
[0037] A is selected from the group consisting of optionally substituted aryl, optionally substituted heteroaryl, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted alkoxy, optionally substituted cycloalkyl, hydrogen, hydroxyl, -CN and halogen;
[0038] B is selected from the group consisting of optionally substituted aryl, optionally substituted heteroaryl, optionally substituted arylalkyl, optionally substituted heteroarylalkyl, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted cycloalkyl, hydrogen, hydroxyl, -CN and halogen;
[0039] C is selected from the group consisting of optionally substituted aryl, optionally substituted heteroaryl, optionally substituted arylalkyl, optionally substituted heteroarylalkyl, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted cycloalkyl, hydrogen, hydroxyl, -CN and halogen;
[0040] Y is selected from null, a bond, O, S, SO, S02, NH, -C(0)-N(R' )-, -C(0)-0-, - N(R2)- and -C(R3)(R4)-;
[0041] X1 and X2 are each independently selected from CRS and N;
[0042] R1 , R2, R3 and R4 are each independently selected from the group consisting of of optionally substituted aryl, optionally substituted 5- or 6-membered heteroaryl, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted alkoxy, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, hydrogen and hydroxyl;
[0043] R5 is independently selected from the group consisting of hydrogen, halogen, -CN, -C(0)-N(R6)-R7, -C(0)-0-R8, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted alkoxy, haloalkyl, haloalkoxy, optionally substituted aryl and optionally substituted 5- or 6- membered heteroaryl; and
[0044] R6, R7 and R8 are each independently selected from the group consisting of of optionally substituted aryl, optionally substituted 5- or 6-membered heteroaryl, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted alkoxy, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, hydrogen and hydroxyl. [0045] Also provided herein are compounds of structural Formula II, or a
pharmaceutically acceptable salt, polymorph, solvate, tautomer, or N oxide thereof:
Figure imgf000014_0001
[0046] wherein C and B are as defined above for structural Formula I.
[0047] Provided herein are compounds of structural Formula III, or a pharmaceutically acceptable salt, polymorph, solvate, tautomer, or N oxide thereof:
Figure imgf000014_0002
(III)
[0048] wherein C is as defined above for structural Formula I.
[0049] Provided herein are compounds of structural Formula IV, or a pharmaceutically acceptable salt, polymorph, solvate, tautomer, or N oxide thereof:
Figure imgf000015_0001
(IV)
[0050] wherein X2 and Y are as defined above for structural Formula I.
[0051] In certain aspects, A is optionally substituted aryl or optionally substituted heteroaryl. In some embodiments, A is optionally substituted phenyl or optionally substituted 5- or 6-membered heteroaryl, wherein the optional substituents are selected from the group consisting of hydrogen, halogen, -CN, optionally substituted carboxyl, amido, nitro, amino, optionally substituted alkyl, optionally substituted alkoxy and optionally substituted haloalkoxy and optionally substituted haloalkyl.
[0052] In certain aspects, A is selected from the group consisting of:
Figure imgf000016_0001
[0053] In some aspects, Y is selected from the group consisting of S, SO, S02, O and N. In certain embodiments, Y is S, O or N. In one embodiment, Y is an optionally substituted Ci-Cgalkyl group. In other embodiments, Y is S, O, N or NH. In one embodiment, Y is S. In another embodiment, Y is S and X2 is CH. In yet another embodiment, Y is O and X2 is N. In a further embodiment, Y is NH and X" is N.In other aspects, C is optionally substituted alkyl, optionally substituted arylalkyl or optionally substituted heteroarylalkyl. In certain embodiments, C is an optionally substituted alkyl group. In certain aspects, C is substituted alkyl, where C is an alkyl substituted with a substituent selected from optionally substituted aryl or optionally substituted heteroaryl. In some embodiments, C is an alkyl group substituted with optionally substituted aryl, including but not limited to optionally substituted phenyl. In other embodiments, C is an alkyl substituted with optionally substituted monocyclic heteroaryl. In further embodiments, C is an alkyl substituted with optionally substituted bicyclic heteroaryl. In certain aspects, C is optionally substituted heteroarylalkyl. In one embodiment, C is substituted heteroarylalkyl, where C is a heteroarylalkyl substituted with a substituent selected from the group consisting of hydrogen, halogen, -CN, optionally substituted carboxyl, amido, nitro, amino, optionally substituted alkyl, optionally substituted alkoxy and optionally substituted haloalkoxy and optionally substituted haloalkyi. In another embodiment, C is optionally substituted arylalkyl, where the optional substituents are selected from the group consisting of hydrogen, halogen, -CN, optionally substituted carboxyl, amido, nitro, amino, optionally substituted alkyl, optionally substituted alkoxy and optionally substituted haloalkoxy and optionally substituted haloalkyi .
[0054] In certain aspects, C is selected from the group consisting of:
Figure imgf000017_0001
Figure imgf000017_0002
Figure imgf000017_0003
Figure imgf000018_0001
Figure imgf000018_0002
Figure imgf000019_0001
Figure imgf000020_0001
[0055] In certain aspects, C is:
Figure imgf000020_0002
[0057] wherein X3 is S or O; and
[0058] R9 is optionally substituted aryl, wherein the optional substituents are selected from the group consisting of hydrogen, halogen, -CN, optionally substituted carboxyl, amido, nitro, amino, optionally substituted alkyl, optionally substituted alkoxy and optionally substituted haloalkoxy and optionally substituted haloalkyl.
[0059] In some embodiments, X3 is S. In other embodiments, X3 is O. 9 is selected from:
Figure imgf000021_0001
[0061] In some embodiments, B is optionally substituted aryl, such as optionally substituted phenyl, optionally substituted heteroaryl, such as optionally substituted pyridinyl, or optionally substituted heteroarylalkyl. In certain aspects, B is substituted aryl, , substituted heteroaryl or substituted heteroarylalkyl, where substituted aryl, such as substituted phenyl,, substituted heteroaryl, such as substituted pyridinyl, or substituted heteroarylalkyl are substituted with a substituent selected from the group consisting of hydrogen, halogen, -CN, optionally substituted carboxyl, amido, nitro, amino, optionally substituted alkyl, optionally substituted alkoxy and optionally substituted haloalkoxy and optionally substituted haloalkyl. In one embodiment, B is substituted phenyl, where the phenyl is substituted with at least one alkoxy, such as methoxy.
[0062] In one embodiment, B is .
Figure imgf000021_0002
[0063] In certain aspects, B is selected from the group consisting of:
Figure imgf000022_0001
[0064] In certain aspects, X1 and X2 are each independently N.
[0065] Also provided herein is a pharmaceutical composition, comprising a compound of structural Formula I, or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable excipient.
[0066] A method of preventing or treating in a subject an apelin-mediated disease or disorder, comprising administering to the subject a therapeutically effective amount of a compound of structural Formula I thereby preventing or treating the disease or disorder is also provided herein. In one embodiment, the subject is a human.
[0067] In certain aspects the disease or disorder is a cardiovascular disease or disorder, coronary heart disease, stroke, heart failure, systolic heart failure, diastolic heart failure, diabetic heart failure, heart failure with preserved ejection fraction, cardiomyopathy, myocardial infarction, left ventricular dysfunction, left ventricular dysfunction after myocardial infarction, cardiac hypertrophy, myocardial remodeling, myocardial remodeling after infarction, myocardial remodeling after cardiac surgery or valvular heart disease. [0068] In other aspects the disease or disorder is a metabolic disease or disorder, metabolic syndrome, insulin resistance, diabetes mellitus, diabetic late complications, diabetic macro- and micro-vasculopathies, diabetic nephropathy, diabetic retinopathy, diabetic neuropathies or cardiac autonomic neuropathy.
[0069] In further aspects the disease or disorder is caused by CNS- dependent or CNS- independent disturbed fluid homeostasis, acute or chronic renal failure, hypertension, pulmonary hypertension, portal hypertension or systolic hypertension.
[0070] In other aspects, the disease or disorder is a vascular disease or disorder, vascular permeability, nonfunctional blood vessels, vascular hypertrophy, vascular remodeling, vascular stiffness, atherosclerosis, peripheral arterial occlusive disease (PAOD), restenosis, thrombosis, vascular permeability disorders, ischemia, reperfusion damage, ischemia or reperfusion damage of the heart, kidney or retina, or a combination thereof.
[0071] In some embodiments, the methods provided herein further comprise coadministering an additional compound in combination with a compound of structural Formula I for the treatment of a disease or disorder, such as, by way of example only, coadministering an additional apelin receptor agonist in combination with a compound of structural Formula I for the treatment of an apelin-mediated disease or disorder or coadministering a chemotherapeutic agent in combination with a compound of structural Formula I for the treatment of cancer.
[0072] As used herein, the terms below have the meanings indicated.
[0073] When ranges of values are disclosed, and the notation "from ni . . . to n2" is used, where ni and n2 are the numbers, then unless otherwise specified, this notation is intended to include the numbers themselves and the range between them. This range may be integral or continuous between and including the end values. By way of example, the range "from 2 to 6 carbons" is intended to include two, three, four, five, and six carbons, since carbons come in integer units. Compare, by way of example, the range "from 1 to 3 μΜ (micromoiar)," which is intended to include 1 μΜ, 3 μΜ, and everything in between to any number of significant figures (e.g., 1 .255 μΜ, 2.1 μΜ, 2.9999 μΜ, etc.).
[0074] The term "about," as used herein, is intended to qualify the numerical values which it modifies, denoting such a value as variable within a margin of error. When no particular margin of error, such as a standard deviation to a mean value given in a chart or table of data, is recited, the term "about" should be understood to mean that range which would encompass the recited value and the range which would be included by rounding up or down to that figure as well, taking into account significant figures.
[0075] The term "acyl," as used herein, alone or in combination, refers to a carbonyl attached to an alkenyl, alkyl, aryl, cycloalkyl, heteroaryl, heterocycloalkyl, or any other moiety were the atom attached to the carbonyl is carbon. An "acetyl" group, which is a type of acyl, refers to a ~C(0)CH3 group. An "alkylcarbonyl" or "alkanoyl" group refers to an alkyl group attached to the parent molecular moiety through a carbonyl group. Examples of such groups include methylcarbonyl and ethylcarbonyl. Examples of acyl groups include formyl, alkanoyl and aroyl.
[0076] The term "alkenyl," as used herein, alone or in combination, refers to a straight- chain or branched-chain hydrocarbon radical having one or more double bonds and containing from 2 to 20 carbon atoms. In certain embodiments, said alkenyl will comprise from 2 to 6 carbon atoms. The term "alkenylene" refers to a carbon-carbon double bond system attached at two or more positions such as ethenylene [(--CH=CH— ),(~C::C— )]. Examples of suitable alkenyl radicals include ethenyl, propenyl, 2-methylpropenyl, 1 ,4- butadienyl and the like. Unless otherwise specified, the term "alkenyl" may include
"alkenylene" groups.
[0077] The term "alkoxy," as used herein, alone or in combination, refers to an alkyl ether radical, wherein the term alkyl is as defined below. Examples of suitable alkyl ether radicals include methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, iso-butoxy, sec-butoxy, tert- butoxy, and the like.
[0078] The term "alkyl," as used herein, alone or in combination, refers to a straight-chain or branched-chain alkyl radical containing from 1 to 20 carbon atoms. In certain
embodiments, said alkyl will comprise from 1 to 10 carbon atoms. In further embodiments, said alkyl will comprise from 1 to 6 carbon atoms. Alkyl groups may be optionally substituted as defined herein. Examples of alkyl radicals include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, pentyl, iso-amyl, hexyl, octyl, noyl and the like.
[0079] The term "alkylene," as used herein, alone or in combination, refers to a saturated aliphatic group derived from a straight or branched chain saturated hydrocarbon attached at two or more positions, such as methylene (— CH2~). Unless otherwise specified, the term "alkyl" may include "alkylene" groups.
[0080] The term "alkylamino," as used herein, alone or in combination, refers to an alkyl group attached to the parent molecular moiety through an amino group. Suitable alkylamino groups may be mono- or dialkylated, forming groups such as, for example, N-methylamino, N-ethylamino, Ν,Ν-dimethylamino, Ν,Ν-ethylmethylamino and the like.
[0081] The term "alkylidene," as used herein, alone or in combination, refers to an alkenyl group in which one carbon atom of the carbon-carbon double bond belongs to the moiety to which the alkenyl group is attached.
[0082] The term "alkylthio," as used herein, alone or in combination, refers to an alkyl thioether (R— S— ) radical wherein the term alkyl is as defined above and wherein the sulfur may be singly or doubly oxidized. Examples of suitable alkyl thioether radicals include methylthio, ethylthio, n-propylthio, isopropylthio, n-butylthio, iso-butylthio, sec-butylthio, tert-butylthio, methanesulfonyl, ethanesulfinyl, and the like.
[0083] The term "alkynyl," as used herein, alone or in combination, refers to a straight- chain or branched chain hydrocarbon radical having one or more triple bonds and containing from 2 to 20 carbon atoms. In certain embodiments, said alkynyl comprises from 2 to 6 carbon atoms. In further embodiments, said alkynyl comprises from 2 to 4 carbon atoms. The term "alkynylene" refers to a carbon-carbon triple bond attached at two positions such as ethynylene (— C:::C— ,— C≡C— ). Examples of alkynyl radicals include ethynyl, propynyl, hydroxypropynyl, butyn- l -yl, butyn-2-yl, pentyn- l -yl, 3-methylbutyn-l -yl, hexyn-2-yl, and the like. Unless otherwise specified, the term "alkynyl" may include "alkynylene" groups.
[0084] The terms "amido" and "carbamoyl," as used herein, alone or in combination, refer to an amino group as described below attached to the parent molecular moiety through a carbonyl group, or vice versa. The term "C-amido" as used herein, alone or in combination, refers to a— C(=0)~N(R)2 group with R as defined herein. The term "N-amido" as used herein, alone or in combination, refers to a RC(=0)N(R')~ group, with R and R' as defined herein. The term "acylamino" as used herein, alone or in combination, embraces an acyl group attached to the parent moiety through an amino group. An example of an "acylamino" group is acetylamino (CH3C(0)NH~). [0085] The term "amino," as used herein, alone or in combination, refers to ~N(R)(R') or - -N+(R)(R')(R"), wherein R, R' and R" are independently selected from the group consisting of hydrogen, alkyl, acyl, heteroalkyl, aryl, cycloaikyl, heteroaryl, and heterocycloaikyl, any of which may themselves be optionally substituted.
[0086] The term "amino acid," as used herein, alone or in combination, means a substituent of the form ~NRCH(R')C(0)OH, wherein R is typically hydrogen, but may be cyclized with N (for example, as in the case of the amino acid proline), and R' is selected from the group consisting of hydrogen, alkyl, heteroalkyl, cycloaikyl, heterocycloaikyl, aryl, heteroaryl, amino, amido, cycloalkylalkyl, heterocycloalkylalkyl, arylalkyl, heteroarylalkyl, aminoalkyl, amidoalkyl, hydroxyalkyl, thiol, thioalkyl, alkylthioalkyl, and alkylthio, any of which may be optionally substituted. The term "amino acid" includes all naturally occurring amino acids as well as synthetic analogues.
[0087] The term "aryl," as used herein, alone or in combination, means a carbocyclic aromatic system containing one, two or three rings wherein such rings may be attached together in a pendent manner or may be fused. The term "aryl" embraces aromatic radicals such as benzyl, phenyl, naphthyl, anthracenyl, phenanthryl, indanyl, indenyl, annulenyl, azulenyl, tetrahydronaphthyl, and biphenyl.
[0088] The term "arylalkenyl" or "aralkenyl," as used herein, alone or in combination, refers to an aryl group attached to the parent molecular moiety through an alkenyl group.
[0089] The term "arylalkoxy" or "aralkoxy," as used herein, alone or in combination, refers to an aryl group attached to the parent molecular moiety through an alkoxy group.
[0090] The term "arylalkyl" or "aralkyl," as used herein, alone or in combination, refers to an aryl group attached to the parent molecular moiety through an alkyl group.
[0091] The term "arylalkynyl" or "aralkynyl," as used herein, alone or in combination, refers to an aryl group attached to the parent molecular moiety through an alkynyl group.
[0092] The term "arylalkanoyl" or "aralkanoyl" or "aroyl," as used herein, alone or in combination, refers to an acyl radical derived from an aryl-substituted alkanecarboxylic acid such as benzoyl, naphthoyl, phenylacetyl, 3-phenylpropionyl(hydrocinnamoyl), 4- phenylbutyryl, (2-naphthyl)acetyl, 4-chlorohydrocinnamoyl, and the like. [0093] The term aryloxy as used herein, alone or in combination, refers to an aryl group attached to the parent molecular moiety through an oxy.
[0094] The terms "benzo" and "benz," as used herein, alone or in combination, refer to the divalent radical ΟόΗ4 derived from benzene. Examples include benzothiophene and benzimidazole.
[0095] The term "carbamate," as used herein, alone or in combination, refers to an ester of carbamic acid (--NHCOO-- ) which may be attached to the parent molecular moiety from either the nitrogen or acid end, and which may be optionally substituted as defined herein.
[0096] The term "O-carbamyl" as used herein, alone or in combination, refers to a— OC(0)NRR', group— with R and R' as defined herein.
[0097] The term "N-carbamyl" as used herein, alone or in combination, refers to a ROC(0)NR'~ group, with R and R' as defined herein.
[0098] The term "carbonyl," as used herein, when alone includes formyl [— C(0)H] and in combination is a --C(O)— group.
[0099] The term "carboxyl" or "carboxy," as used herein, refers to— C(0)OH, O-carboxy, C-carboxy, or the corresponding "carboxylate" anion, such as is in a carboxylic acid salt. An "O-carboxy" group refers to a RC(0)0~ group, where R is as defined herein. A "C-carboxy" group refers to a ~C(0)OR groups where R is as defined herein.
[0100] The term "cyano," as used herein, alone or in combination, refers to— CN.
[0101] The term "cycloalkyl," or, alternatively, "carbocycle," as used herein, alone or in combination, refers to a saturated or partially saturated monocyclic, bicyciic or tricyclic alkyl radical wherein each cyclic moiety contains from 3 to 12 carbon atom ring members and which may optionally be a benzo fused ring system which is optionally substituted as defined herein. In certain embodiments, said cycloalkyl will comprise from 5 to 7 carbon atoms. Examples of such cycloalkyl radicals include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, octahydronaphthyl, 2,3-dihydro-l H-indenyl, adamantyl and the like. "Bicyciic" and "tricyclic" as used herein are intended to include both fused ring systems, such as decahydronaphthalene, octahydronaphthalene as well as the multicyclic (multicentered) saturated or partially unsaturated type. The latter type of isomer is exemplified in general by, bicyclo[l , l , l]pentane, camphor, adamantane, and bicyclo[3,2,l ]octane. [0102] The term "ester," as used herein, alone or in combination, refers to a carboxyl group bridging two moieties linked at carbon atoms.
[0103] The term "ether," as used herein, alone or in combination, typically refers to an oxy group bridging two moieties linked at carbon atoms. "Ether" may also include polyethers, such as, for example, --RO(CH2)20(CH2)20(CH2)2OR', --RO(CH2)20(CH2)2OR*, - RO(CH2)2OR', and --RO(CH2)2OH.
[0104] The term "halo," or "halogen," as used herein, alone or in combination, refers to fluorine, chlorine, bromine, or iodine.
[0105] The term "haloalkoxy," as used herein, alone or in combination, refers to a haloalkyl group attached to the parent molecular moiety through an oxygen atom.
[0106] The term "haloalkyl," as used herein, alone or in combination, refers to an alkyl radical having the meaning as defined above wherein one or more hydrogens are replaced with a halogen. Specifically embraced are monohaloalkyl, dihaloalkyl and polyhaloalkyl radicals. A monohaloalkyl radical, for one example, may have an iodo, bromo, chloro or fluoro atom within the radical. Dihalo and polyhaloalkyl radicals may have two or more of the same halo atoms or a combination of different halo radicals. Examples of haloalkyl radicals include fluoromethyl, difluoromethyl, trifluoromethyl, chloromethyl, dichloromethyl, trichloromethyl, pentafluoroethyl, heptafluoropropyl, difluorochloromethyl,
dichlorofluoromethyl, difluoroethyl, difluoropropyl, dichloroethyl and dichloropropyl.
"Haloalkylene" refers to a haloalkyl group attached at two or more positions. Examples include fluoromethylene (~CFH--), difluoromethylene (--CF2--), chloromethylene (-CHC1--) and the like.
[0107] The term "heteroalkyl," as used herein, alone or in combination, refers to a stable straight or branched chain, or cyclic hydrocarbon radical, or combinations thereof, fully saturated or containing from 1 to 3 degrees of unsaturation, consisting of the stated number of carbon atoms and from one to three heteroatoms selected from the group consisting of O, N, and S, and wherein the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized. The heteroatom(s) O, N and S may be placed at any interior position of the heteroalkyl group. Up to two heteroatoms may be consecutive, such as, for example, ~CH2~NH~OCH3. The term heteroalkyl may include ethers. [0108] The term "heteroaryl," as used herein, alone or in combination, refers to 3 to 7 membered unsaturated heteromonocyclic rings, or fused polycyclic rings in which at least one of the fused rings is unsaturated, wherein at least one atom is selected from the group consisting of O, S, and N. In certain embodiments, said heteroaryl will comprise from 5 to 7 carbon atoms. The term also embraces fused polycyclic groups wherein heterocyclic radicals are fused with aryl radicals, wherein heteroaryl radicals are fused with other heteroaryl radicals, or wherein heteroaryl radicals are fused with cycloalkyl radicals. Examples of heteroaryl groups include pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, pyranyl, furyl, thienyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, thiadiazolyl, isothiazolyl, indolyl, isoindolyl, indolizinyl, benzimidazolyl, quinolyl, isoquinolyl, quinoxalinyl, quinazolinyl, indazolyl, benzotriazolyl, benzodioxolyl, benzopyranyl, benzoxazolyl, benzoxadiazolyl, benzothiazolyl, benzothiadiazolyl, benzofuryl, benzothienyl, chromonyl, coumarinyl, benzopyranyl, tetrahydroquinolinyl,
tetrazolopyridazinyl, tetrahydroisoquinolmyl, thienopyridinyl, furopyridinyl, pyrrolopyridinyl and the like. Exemplary tricyclic heterocyclic groups include carbazolyl, benzidolyl, phenanthrolinyl, dibenzofuranyl, acridinyl, phenanthridinyl, xanthenyl and the like.
[0109] The terms "heterocycloalkyl" and, interchangeably, "heterocycle," as used herein, alone or in combination, each refer to a saturated, partially unsaturated, or fully unsaturated monocyclic, bicyclic, or tricyclic heterocyclic radical containing at least one heteroatom as ring members, wherein each said heteroatom may be independently selected from the group consisting of nitrogen, oxygen, and sulfur In certain embodiments, said heterocycloalkyl will comprise from 1 to 4 heteroatoms as ring members. In further embodiments, said
heterocycloalkyl will comprise from 1 to 2 heteroatoms ring members. In certain
embodiments, said heterocycloalkyl will comprise from 3 to 8 ring members in each ring. In further embodiments, said heterocycloalkyl will comprise from 3 to 7 ring members in each ring. In yet further embodiments, said heterocycloalkyl will comprise from 5 to 6 ring members in each ring. "Heterocycloalkyl" and "heterocycle" are intended to include sugars, sulfones, sulfoxides, N-oxides of tertiary nitrogen ring members, and carbocyclic fused and benzo fused ring systems; additionally, both terms also include systems where a heterocycle ring is fused to an aryl group, as defined herein, or an additional heterocycle group. Examples of heterocycloalkyl groups include aziridinyl, azetidinyl, 1 ,3-benzodioxolyl,
dihydroisoindolyl, dihydroisoquinolinyl, dihydrocinnolinyl, dihydrobenzodioxinyl, dihydro[l ,3]oxazolo[4,5-b]pyridinyl, benzothiazolyl, dihydroindolyl, dihy-dropyridinyl, 1,3- dioxanyl, 1 ,4-dioxanyl, 1 ,3-dioxolanyl, isoindolinyl, morpholinyl, piperazinyl, pyrrolidinyl, tetrahydropyridinyl, piperidinyl, thiomorpholinyl, and the like. The heterocycloalkyl groups may be optionally substituted unless specifically prohibited.
[0110] The term "hydrazinyl" as used herein, alone or in combination, refers to two amino groups joined by a single bond, i.e., --N--N— .
[0111 J The term "hydroxamic acid" as used herein, refers to ~C(0)ON(R)0(R'), wherein R and R' are as defined herein, or the corresponding "hydroxamate" anion, including any corresponding hydroxamic acid salt.
[0112] The term "hydroxy," as used herein, alone or in combination, refers to—OH.
[0113] The term "hydroxyalkyl," as used herein, alone or in combination, refers to a hydroxy group attached to the parent molecular moiety through an alkyl group.
[01 14] The term "imino," as used herein, alone or in combination, refers to =N~.
[0115] The term "iminohydroxy," as used herein, alone or in combination, refers to =N(OH) and =N--0~.
[0116] The term "isocyanato" refers to a --NCO group. [0117] The term "isothiocyanato" refers to a— NCS group.
[0118] The phrase "linear chain of atoms" refers to the longest straight chain of atoms independently selected from carbon, nitrogen, oxygen and sulfur.
[0119] The term "lower," as used herein, alone or in combination, means containing from 1 to and including 6 carbon atoms.
[0120] The term "mercaptyl" as used herein, alone or in combination, refers to an RS— group, where R is as defined herein.
[0121] The term "nitro," as used herein, alone or in combination, refers to — NOi.
[0122] The terms "oxy" or "oxa" as used herein, alone or in combination, refer to— O— .
[0123] The term "oxo," as used herein, alone or in combination, refers to =0.
[0124] The term "perhaloalkoxy" refers to an alkoxy group where all of the hydrogen atoms are replaced by halogen atoms. [0125] The term "perhaloalkyi" as used herein, alone or in combination, refers to an alkyl group where all of the hydrogen atoms are replaced by halogen atoms.
[0126] The term "phosphoamide" as used herein, alone or in combination, refers to a phosphate group [(OH)2P(0)0-] in which one or more of the hydroxyl groups has been replaced by nitrogen, amino, or amido.
[0127] The term "phosphonate" as used herein, alone or in combination, refers to a group of the form ROP(OR')(OR)0— wherein R and R' are selected from the group consisting of hydrogen, alkyl, acyl, heteroalkyl, aryl, cycloalkyi, heteroaryl, and heterocycloalkyl, any of which may themselves be optionally substituted. "Phosphonate" includes "phosphate [(OH) 2P(0)0~] and related phosphoric acid anions which may form salts.
[0128] The terms "sulfonate," "sulfonic acid," and "sulfonic," as used herein, alone or in combination, refers to the --SO3H group and its anion as the sulfonic acid is used in salt formation.
[0129] The term "sulfanyl," as used herein, alone or in combination, refers to— S— .
[0130] The term "sulfmyl," as used herein, alone or in combination, refers to --S(O)--.
[0131] The term "sulfonyl," as used herein, alone or in combination, refers to— S(0)2— .
[0132] The term "N-sulfonamido" refers to a RS(=0)2NR'~ group with R and R' as defined herein.
[0133] The term "S-sulfonamido" refers to a ~S(=0)2NRR', group, with R and R' as defined herein.
[0134] The terms "thia" and "thio," as used herein, alone or in combination, refer to a— S~ group or an ether wherein the oxygen is replaced with sulfur. The oxidized derivatives of the thio group, namely sulfmyl and sulfonyl, are included in the definition of thia and thio.
[0135] The term "thiol," as used herein, alone or in combination, refers to an ~SH group.
[0136] The term "thiocarbonyl," as used herein, when alone includes thioformyl— C(S)H and in combination is a— C(S)~ group.
[0137] The term "N-thiocarbamyl" refers to an ROC(S)NR'~ group, with R and R' as defined herein. [0138] The term "O-thiocarbamyl" refers to a ~OC(S)NRR', group with R and R' as defined herein.
[0139] The term "thiocyanato" refers to a— CNS group.
[0140] The term "trihalomethanesulfonamido" refers to a X3CS(0) R~ group with X is a halogen and R as defined herein.
[0141] The term "trihalomethanesulfonyl" refers to a X3CS(0)2~ group where X is a halogen.
[0142] The term "trihalomethoxy" refers to a X3CO-- group where X is a halogen.
[0143] The term "trisubstituted silyl," as used herein, alone or in combination, refers to a silicone group substituted at its three free valences with groups as l isted herein under the definition of substituted amino. Examples include trimethysilyl, tert-butyldimethylsilyl, triphenylsilyl and the like.
[0144] Any definition herein may be used in combination with any other definition to describe a composite structural group. By convention, the trailing element of any such definition is that which attaches to the parent moiety. For example, the composite group alkylamido would represent an alkyl group attached to the parent molecule through an amido group, and the term alkoxyalkyl would represent an alkoxy group attached to the parent molecule through an alkyl group.
[0145] When a group is defined to be "null," what is meant is that said group is absent. A "null" group occurring between two other groups may also be understood to be a collapsing of flanking groups. For example, if in --(CHi)Gi G2G3, the element G2 were null, said group would become— (CH2)GiG3.
[0146] The term "optionally substituted" means the anteceding group may be substituted or unsubstituted. When substituted, the substituents of an "optionally substituted" group may include, without lim itation, one or more substituents independently selected from the following groups or a particular designated set of groups, alone or in combination: lower alkyl, lower alkenyl, lower alkynyl, lower alkanoyl, lower heteroalkyl, lower
heterocycloalkyl, lower haloalkyl, lower haloalkenyl, lower haloalkynyl, lower perhaloalkyl, lower perhaloalkoxy, lower cycloalkyl, phenyl, aryl, aryloxy, lower alkoxy, lower haloalkoxy, oxo, lower acyloxy, carbonyl, carboxyl, lower aikylcarbonyl, lower carboxyester, lower carboxamido, cyano, hydrogen, halogen, hydroxy, amino, lower alkylamino, arylamino, amido, nitro, thiol, lower alkylthio, lower haloalkylthio, lower perhaloalkylthio, arylthio, sulfonate, sulfonic acid, trisubstituted silyl, N3, SH, SCH3, C(0)CH3, CO2CH3, CO2H, pyridinyl, thiophene, furanyl, lower carbamate, and lower urea. Two substituents may be joined together to form a fused five-, six-, or seven-membered carbocyclic or heterocyclic ring consisting of zero to three heteroatoms, for example forming methylenedioxy or ethylenedioxy. An optionally substituted group may be unsubstituted (e.g.,— CH2CH3), fully substituted (e.g.,— CF2CF3), monosubstituted (e.g., ~ CF CHbF) or substituted at a level anywhere in-between fully substituted and monosubstituted (e.g.,— CH2CF3). Where substituents are recited without qualification as to substitution, both substituted and unsubstituted forms are encompassed. Where a substituent is qualified as "substituted," the substituted form is specifically intended. Additionally, different sets of optional substituents to a particular moiety may be defined as needed; in these cases, the optional substitution will be as defined, often immediately following the phrase, "optionally substituted with."
[0147] The term R or the term R', appearing by itself and without a number designation, unless otherwise defined, refers to a moiety selected from the group consisting of hydrogen, hydroxyl, halogen, alkyl, cycloalkyl, heteroalkyl, aryl, heteroaryl and heterocycloalkyl, any of which may be optionally substituted. Such R and R' groups should be understood to be optionally substituted as defined herein. Whether an R group has a number designation or not, every R group, including R, R' and Rn where n = ( 1 , 2, 3, . . . n), every substituent, and every term should be understood to be independent of every other in terms of selection from a group. Should any variable, substituent, or term (e.g. aryl, heterocycle, R, etc.) occur more than one time in a formula or generic structure, its definition at each occurrence is independent of the definition at every other occurrence. Those of skill in the art will further recognize that certain groups may be attached to a parent molecule or may occupy a position in a chain of elements from either end as written. Thus, by way of example only, an unsymmetrical group such as ~C(0)N(R)— may be attached to the parent moiety at either the carbon or the nitrogen.
[0148] Asymmetric centers exist in the compounds of the present invention. These centers are designated by the symbols "R" or "S," depending on the configuration of substituents around the chiral carbon atom. It should be understood that the invention encompasses all stereochemical isomeric forms, including diastereomeric, enantiomeric, and epimeric forms, as well as d-isomers and 1 -isomers, and mixtures thereof. Individual stereoisomers of compounds can be prepared synthetically from commercially available starting materials which contain chiral centers or by preparation of mixtures of enantiomeric products followed by separation such as conversion to a mixture of diastereomers followed by separation or recrystallization, chromatographic techniques, direct separation of enantiomers on chiral chromatographic columns, or any other appropriate method known in the art. Starting compounds of particular stereochemistry are either commercially available or can be made and resolved by techniques known in the art. Additionally, the compounds of the present invention may exist as geometric isomers. The present invention includes all cis, trans, syn, anti, entgegen (E), and zusammen (Z) isomers as well as the appropriate mixtures thereof. Additionally, compounds may exist as tautomers, including keto-enol tautomers; all tautomeric isomers are provided by this invention. Additionally, the compounds of the present invention can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. In general, the solvated forms are considered equivalent to the unsolvated forms for the purposes of the present invention.
[0149] The term "bond" refers to a covalent linkage between two atoms, or two moieties when the atoms joined by the bond are considered to be part of larger substructure. A bond may be single, double, or triple unless otherwise specified. A dashed line between two atoms in a drawing of a molecule indicates that an additional bond may be present or absent at that position.
[0150] The term "disease" as used herein is intended to be generally synonymous, and is used interchangeably with, the terms "disorder" and "condition" (as in medical condition), in that all reflect an abnormal condition of the body or of one of its parts that impairs normal functioning and is typically manifested by distinguishing signs and symptoms.
[0151] The term "combination therapy" means the administration of two or more therapeutic agents to treat a therapeutic condition or disorder described in the present disclosure. Such administration encompasses co-administration of these therapeutic agents in a substantially simultaneous manner, such as in a single capsule having a fixed ratio of active ingredients or in multiple, separate capsules for each active ingredient. In addition, such administration also encompasses use of each type of therapeutic agent in a sequential manner. In either case, the treatment regimen will provide beneficial effects of the drug combination in treating the conditions or disorders described herein.
[0152] The phrase "therapeutically effective" is intended to qualify the amount of active ingredients used in the treatment of a disease or disorder. This amount will achieve the goal of reducing or eliminating the said disease or disorder. [0153] As used herein, reference to "treatment" of a patient is intended to include prophylaxis. The term "patient" means all mammals including humans. Examples of patients include humans, cows, dogs, cats, goats, sheep, pigs, and rabbits. Preferably, the patient is a human.
[0154] The term "prodrug" refers to a compound that is made more active in vivo. Certain of the present compounds can also exist as prodrugs, as described in Hydrolysis in Drug and Prodrug Metabolism: Chemistry, Biochemistry, and Enzymology (Testa, Bernard and Mayer, Joachim M. Wiley- VHCA, Zurich, Switzerland 2003). Prodrugs of the compounds described herein are structurally modified forms of the compound that readily undergo chemical changes under physiological conditions to provide the compound. Additionally, prodrugs can be converted to the compound by chemical or biochemical methods in an ex vivo
environment. For example, prodrugs can be slowly converted to a compound when placed in a transdermal patch reservoir with a suitable enzyme or chemical reagent. Prodrugs are often useful because, in some situations, they may be easier to administer than the compound, or parent drug. They may, for instance, be bioavailable by oral administration whereas the parent drug is not. The prodrug may also have improved solubility in pharmaceutical compositions over the parent drug. A wide variety of prodrug derivatives are known in the art, such as those that rely on hydrolytic cleavage or oxidative activation of the prodrug. An example, without limitation, of a prodrug would be a compound which is administered as an ester (the "prodrug"), but then is metabolically hydrolyzed to the carboxylic acid, the active entity. Additional examples include peptidyl derivatives of a compound. The term
"therapeutically acceptable prodrug," refers to those prodrugs or zwitterions which are suitable for use in contact with the tissues of patients without undue toxicity, irritation, and allergic response, are commensurate with a reasonable benefit/risk ratio, and are effective for their intended use.
[0155] The compounds of the present invention can exist as therapeutically acceptable salts. The present invention includes compounds listed above in the form of salts, including acid addition salts. Suitable salts include those formed with both organic and inorganic acids. Such acid addition salts will normally be pharmaceutically acceptable. However, salts of non- pharmaceutically acceptable salts may be of utility in the preparation and purification of the compound in question. Basic addition salts may also be formed and be pharmaceutically acceptable. For a more complete discussion of the preparation and selection of salts, refer to Pharmaceutical Salts: Properties, Selection, and Use (Stahl, P. Heinrich. Wiley- VCHA, Zurich, Switzerland, 2002). [0156] The term "therapeutically acceptable salt," as used herein, represents salts or zwitterionic forms of the compounds of the present invention which are water or oil-soluble or dispersible and therapeutically acceptable as defined herein. The salts can be prepared during the final isolation and purification of the compounds or separately by reacting the appropriate compound in the form of the free base with a suitable acid. Representative acid addition salts include acetate, adipate, alginate, L-ascorbate, aspartate, benzoate,
benzenesulfonate (besylate), bisulfate, butyrate, camphorate, camphorsulfonate, citrate, digluconate, formate, fumarate, gentisate, glutarate, glycerophosphate, glycolate, hemisulfate, heptanoate, hexanoate, hippurate, hydrochloride, hydrobromide, hydroiodide, 2- hydroxyethansulfonate (isethionate), lactate, maleate, malonate, DL-mandelate,
mesitylenesulfonate, methanesulfonate, naphthylenesulfonate, nicotinate, 2- naphthalenesulfonate, oxalate, pamoate, pectinate, persulfate, 3-phenylproprionate, phosphonate, picrate, pivalate, propionate, pyroglutamate, succinate, sulfonate, tartrate, L- tartrate, trichloroacetate, trifluoroacetate, phosphate, glutamate, bicarbonate, para- toluenesulfonate (p-tosylate), and undecanoate. Also, basic groups in the compounds of the present invention can be quaternized with methyl, ethyl, propyl, and butyl chlorides, bromides, and iodides; dimethyl, diethyl, dibutyl, and diamyl sulfates; decyl, lauryl, myristyl, and steryl chlorides, bromides, and iodides; and benzyl and phenethyl bromides. Examples of acids which can be employed to form therapeutically acceptable addition salts include inorganic acids such as hydrochloric, hydrobromic, sulfuric, and phosphoric, and organic acids such as oxalic, maleic, succinic, and citric. Salts can also be formed by coordination of the compounds with an alkali metal or alkaline earth ion. Hence, the present invention contemplates sodium, potassium, magnesium, and calcium salts of the compounds disclosed herein, and the like.
[0157] Basic addition salts can be prepared during the final isolation and purification of the compounds by reacting a carboxyl group with a suitable base such as the hydroxide, carbonate, or bicarbonate of a metal cation or with ammonia or an organic primary, secondary, or tertiary amine. The cations of therapeutically acceptable salts include lithium, sodium, potassium, calcium, magnesium, and aluminum, as well as nontoxic quaternary amine cations such as ammonium, tetramethylammonium, tetraethylammonium,
methylamine, dimethylamine, trimethylamine, triethylamine, diethylamine, ethylamine, tributylamine, pyridine, Ν,Ν-dimethylaniline, N-methylpiperidine, N-methylmorpholine, dicyclohexylamine, procaine, dibenzylamine, Ν,Ν-dibenzylphenethylamine, 1 -ephenamine, and Ν,Ν'-dibenzylethylenediamine. Other representative organic amines useful for the formation of base addition salts include ethylenediamine, ethanolamine, diethanolamine, piperidine, and piperazine.
[0158] While it may be possible for the compounds of the subject invention to be administered as the raw chemical, it is also possible to present them as a pharmaceutical formulation. Accordingly, provided herein are pharmaceutical formulations which comprise one or more of certain compounds of the present invention, or one or more pharmaceutically acceptable salts, esters, prodrugs, amides, or solvates thereof, together with one or more pharmaceutically acceptable carriers thereof and optionally one or more other therapeutic ingredients. The carrier(s) must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. Proper formulation is dependent upon the route of administration chosen. Any of the well-known techniques, carriers, and excipients may be used as suitable and as understood in the art; e.g., in Remington's Pharmaceutical Sciences. The pharmaceutical compositions disclosed herein may be manufactured in any manner known in the art, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or compression processes.
[0159] The formulations include those suitable for oral, parenteral (including
subcutaneous, intradermal, intramuscular, intravenous, intraarticular, and intramedullary), intraperitoneal, transmucosal, transdermal, rectal and topical (including dermal, buccal, sublingual and intraocular) administration although the most suitable route may depend upon for example the condition and disorder of the recipient. The formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Typically, these methods include the step of bringing into association a compound of the subject invention or a pharmaceutically acceptable salt, ester, amide, prodrug or solvate thereof ("active ingredient") with the carrier which constitutes one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both and then, if necessary, shaping the product into the desired formulation.
[0160] Formulations of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a
predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous liquid or a non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion. The active ingredient may also be presented as a bolus, electuary or paste.
[0161] Pharmaceutical preparations which can be used orally include tablets, push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. Tablets may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with binders, inert diluents, or lubricating, surface active or dispersing agents. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredient therein. All formulations for oral administration should be in dosages suitable for such administration. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
[0162] The compounds may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. The formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in powder form or in a freeze- dried (lyophilized) condition requiring only the addition of the sterile liquid earner, for example, saline or sterile pyrogen-free water, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described. [0163] Formulations for parenteral administration include aqueous and non-aqueous (oily) sterile injection solutions of the active compounds which may contain antioxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
[0164] In addition to the formulations described previously, the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the compounds may be formulated with suitable polymeric or
hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
[0165] For buccal or sublingual administration, the compositions may take the form of tablets, lozenges, pastilles, or gels formulated in conventional manner. Such compositions may comprise the active ingredient in a flavored basis such as sucrose and acacia or tragacanth.
[0166] The compounds may also be formulated in rectal compositions such as
suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter, polyethylene glycol, or other glycerides.
[0167] Certain compounds of the present invention may be administered topically, that is by non-systemic administration. This includes the application of a compound of the present invention externally to the epidermis or the buccal cavity and the instillation of such a compound into the ear, eye and nose, such that the compound does not significantly enter the blood stream. In contrast, systemic administration refers to oral, intravenous, intraperitoneal and intramuscular administration.
[0168] Formulations suitable for topical administration include liquid or semi-liquid preparations suitable for penetration through the skin to the site of inflammation such as gels, liniments, lotions, creams, ointments or pastes, and drops suitable for administration to the eye, ear or nose. The active ingredient for topical administration may comprise, for example, from 0.001% to 10% w/w (by weight) of the formulation. In certain embodiments, the active ingredient may comprise as much as 10% w/w. In other embodiments, it may comprise less than 5% w/w. In certain embodiments, the active ingredient may comprise from 2% w/w to 5% w/w. In other embodiments, it may comprise from 0.1% to 1 % w/w of the formulation.
[0169] Gels for topical or transdermal administration may comprise, generally, a mixture of volatile solvents, nonvolatile solvents, and water. In certain embodiments, the volatile solvent component of the buffered solvent system may include lower (C 1 -C6) alkyl alcohols, lower alkyl glycols and lower glycol polymers. In further embodiments, the volatile solvent is ethanol. The volatile solvent component is thought to act as a penetration enhancer, while also producing a cooling effect on the skin as it evaporates. The nonvolatile solvent portion of the buffered solvent system is selected from lower alkylene glycols and lower glycol polymers. In certain embodiments, propylene glycol is used. The nonvolatile solvent slows the evaporation of the volatile solvent and reduces the vapor pressure of the buffered solvent system. The amount of this nonvolatile solvent component, as with the volatile solvent, is determined by the pharmaceutical compound or drug being used. When too little of the nonvolatile solvent is in the system, the pharmaceutical compound may crystallize due to evaporation of volatile solvent, while an excess may result in a lack of bioavailability due to poor release of drug from solvent mixture. The buffer component of the buffered solvent system may be selected from any buffer commonly used in the art; in certain embodiments, water is used. A common ratio of ingredients is about 20% of the nonvolatile solvent, about 40% of the volatile solvent, and about 40% water. There are several optional ingredients which can be added to the topical composition. These include, but are not limited to, chelators and gelling agents. Appropriate gelling agents can include, but are not limited to, semisynthetic cellulose derivatives (such as hydroxypropylmethylcellulose) and synthetic polymers, and cosmetic agents.
[0170] Lotions include those suitable for application to the skin or eye. An eye lotion may comprise a sterile aqueous solution optionally containing a bactericide and may be prepared by methods similar to those for the preparation of drops. Lotions or liniments for application to the skin may also include an agent to hasten drying and to cool the skin, such as an alcohol or acetone, and/or a moisturizer such as glycerol or an oil such as castor oil or arachis oil.
[0171 ] Creams, ointments or pastes are semi-solid formulations of the active ingredient for external application. They may be made by mixing the active ingredient in finely-divided or powdered form, alone or in solution or suspension in an aqueous or non-aqueous fluid, with the aid of suitable machinery, with a greasy or non-greasy base. The base may comprise hydrocarbons such as hard, soft or liquid paraffin, glycerol, beeswax, a metallic soap; a mucilage; an oil of natural origin such as almond, corn, arachis, castor or olive oil; wool fat or its derivatives or a fatty acid such as stearic or oleic acid together with an alcohol such as propylene glycol or a macrogel. The formulation may incorporate any suitable surface active agent such as an anionic, cationic or non-ionic surfactant such as a sorbitan ester or a polyoxyethylene derivative thereof. Suspending agents such as natural gums, cellulose derivatives or inorganic materials such as silicaceous silicas, and other ingredients such as lanolin, may also be included.
[0172] Drops may comprise sterile aqueous or oily solutions or suspensions and may be prepared by dissolving the active ingredient in a suitable aqueous solution of a bactericidal and/or fungicidal agent and/or any other suitable preservative, and, in certain embodiments, including a surface active agent. The resulting solution may then be clarified by filtration, transferred to a suitable container which is then sealed and sterilized by autoclaving or maintaining at 98- 100 °C for half an hour. Alternatively, the solution may be sterilized by filtration and transferred to the container by an aseptic technique. Examples of bactericidal and fungicidal agents suitable for inclusion in the drops are phenylmercuric nitrate or acetate (0.002%), benzalkonium chloride (0.01%) and chlorhexidine acetate (0.01%). Suitable solvents for the preparation of an oily solution include glycerol, diluted alcohol and propylene glycol.
[0173] Formulations for topical administration in the mouth, for example buccally or sublingually, include lozenges comprising the active ingredient in a flavored basis such as sucrose and acacia or tragacanth, and pastilles comprising the active ingredient in a basis such as gelatin and glycerin or sucrose and acacia.
[0174] For administration by inhalation, compounds may be conveniently delivered from an insufflator, nebulizer pressurized packs or other convenient means of delivering an aerosol spray. Pressurized packs may comprise a suitable propellant such as
dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. Alternatively, for administration by inhalation or insufflation, the compounds according to the invention may take the form of a dry powder composition, for example a powder mix of the compound and a suitable powder base such as lactose or starch. The powder composition may be presented in unit dosage form, in for example, capsules, cartridges, gelatin or blister packs from which the powder may be administered with the aid of an inhalator or insufflator.
[0175] Preferred unit dosage formulations are those containing an effective dose, as herein below recited, or an appropriate fraction thereof, of the active ingredient.
[0176] It should be understood that in addition to the ingredients particularly mentioned above, the formulations described above may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavoring agents.
[0177] Compounds may be administered orally or via injection at a dose of from 0.1 to 500 mg/kg per day. The dose range for adult humans is generally from 5 mg to 2 g/day. Tablets or other forms of presentation provided in discrete units may conveniently contain an amount of one or more compounds which is effective at such dosage or as a multiple of the same, for instance, units containing 5 mg to 500 mg, usually around 10 mg to 200 mg.
[0178] The amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
[0179] The compounds can be administered in various modes, e.g. orally, topically, or by injection. The precise amount of compound administered to a patient will be the
responsibility of the attendant physician. The specific dose level for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diets, time of administration, route of administration, rate of excretion, drug combination, the precise disorder being treated, and the severity of the indication or condition being treated. Also, the route of administration may vary depending on the condition and its severity.
[0180] In certain instances, it may be appropriate to administer at least one of the compounds described herein (or a pharmaceutically acceptable salt, ester, or prodrug thereof) in combination with another therapeutic agent. By way of example only, if one of the side effects experienced by a patient upon receiving one of the compounds herein is hypertension, then it may be appropriate to administer an anti-hypertensive agent in combination with the initial therapeutic agent. Or, by way of example only, the therapeutic effectiveness of one of the compounds described herein may be enhanced by administration of an adjuvant (i.e., by itself the adjuvant may only have minimal therapeutic benefit, but in combination with another therapeutic agent, the overall therapeutic benefit to the patient is enhanced). Or, by way of example only, the benefit of experienced by a patient may be increased by administering one of the compounds described herein with another therapeutic agent (which also includes a therapeutic regimen) that also has therapeutic benefit. By way of example only, in a treatment for diabetes involving administration of one of the compounds described herein, increased therapeutic benefit may result by also providing the patient with another therapeutic agent for diabetes. In any case, regardless of the disease, disorder or condition being treated, the overall benefit experienced by the patient may simply be additive of the two therapeutic agents or the patient may experience a synergistic benefit.
[0181] In any case, the multiple therapeutic agents (at least one of which is a compound of the present invention) may be administered in any order or even simultaneously. If simultaneously, the multiple therapeutic agents may be provided in a single, unified form, or in multiple forms (by way of example only, either as a single pill or as two separate pills). One of the therapeutic agents may be given in multiple doses, or both may be given as multiple doses. If not simultaneous, the timing between the multiple doses may be any duration of time ranging from a few minutes to four weeks.
DETAILED DESCRIPTION OF THE INVENTION
[0182] The recently discovered apelin receptor (APJ, AGTRL- 1 , APLNR) system has emerged as a critical mediator of cardiovascular homeostasis and is involved in the pathogenesis of hypertension, heart failure, atherosclerosis and other cardiovascular diseases and is the subject of intense investigation. Disclosed herein is the first discovery and characterization of potent small molecule APJ functional agonists.
[0183] Apelin is the endogenous peptide ligand for the G-protein coupled receptor (GPCR) APJ (angiotensin II receptor-like 1 , AGTRL-1 and APLNR). Until the discovery of apelin, APJ was an orphan GPCR. APJ is coupled to Gai, and has been shown in cell culture to inhibit adenylate cyclase. The APJ gene encodes a receptor that most closely resembles the angiotensin receptor ATI ; however, t he APJ receptor does not bind angiotensin.
[0184] Underscoring the emerging importance of the apelin/APJ system, recent studies have shown that apelin reduces the extent of atherosclerotic lesions in ApoE /- mice , and opposes the development of abdominal aortic aneurysms. Additional studies have also revealed that APJ forms a heterodimer with the Ang II receptor ATI , and that this complex facilitates antagonism of Ang II signaling by apelin. Despite these exciting results, there remains a multitude of unanswered questions regarding the role of apelin and APJ in the physiology and pathology.
[0185] Disclosed herein is a series of small molecule agonists of the apelin receptor that have beend developed for the treatment of diseases such as heart failure, arrhythmia, atherosclerosis, and disorders where pathological angiogenesis is involved.
[0186] The series of molecules disclosed herein are potent (EC50≤ 0.01 u for most potent cases), and exhibit dose dependent agonist of the apelin receptor. The compounds disclosed herein are selective for the apelin receptor, and have been shown to be highly specific for the apelin receptor versus the angiotensin II receptor (ATI), the most closely related receptor. The compounds disclosed herein are low in molecular weight (<500), and are typically lipophilic.
[0187] Currently, it is conceived that these compounds would be useful as novel therapies for the treatment for a range of cardiovascular and metabolic conditions. In another aspect of the invention, apelin receptor agonists would be useful as therapeutic regimens for chronic kidney disease.
[0188] Administration of apelins cause vasodilatation in different pre-clinical models and accordingly, intravenous administration in rodents reduces mean arterial blood pressure, systemic venous tone and cardiac pre- and afterload (for a review see Barnes et al. Heart 2010, 96: 101 1 -1016). Vasodilatation to apelin in rodents is dependent on endothelium and mediated through nitric oxide and prostacyclin dependent pathways. Ishida and colleagues demonstrated in 2004, that a functional knockdown of the apelin receptor abolished blood pressure lowering effects of apelins, confirming that vascular effects of apelins are mediated by the apelin receptor specifically (Ishida et al. 2004, J Biol Chem;279:25, 25274-25279).
[0189] The vascular effects of apelin in preclinical studies translate into similar effects in humans (Japp et al., 2008 J Am Coll Cardiol 2008;52:908-913; Japp et al. Circulation 2010; 121 : 1818- 1827). Infusions of apelins increased forearm and coronary blood flow and lowered mean arterial pressure and peripheral vascular resistance in heart failure patients and healthy control subjects in heart failure patients without raising heart rates. An increased cardiax index could be noted, which may be explained by either direct effects on the cardiac muscle (see below) and/or reduction of pre- and afterload in the peripheral circulation. In man, vasodilatation by apelins is reduced by two thirds during nitric oxide synthase inhibition but is unaffected by prostacyclin inhibition. The apelin receptor has been linked to direct cardiac actions. In vitro, exogenous apelin increases contractility at subnanomolar concentrations in atrial strips (Maguire et al. Hypertension 2009;54:598-604) and whole rat hearts (Szokodi et al. Circ. Res. 2002;91 , 434-440). In healthy rodents, acute apelin infusion increases myocardial contractility independently of its effects on loading conditions.
Uniquely among current inotropic agents, chronic dosing causes a sustained increase in cardiac output without inducing left ventricular hypertrophy (Ashley et al. Cardiovasc Res 2005;65:73-82). While apelin-deficient mice display normal or only slightly impaired basal cardiac function at early life cycles, they demonstrate progressive cardiac dysfunction from 6 months of age and develop severe heart failure when subjected to chronic pressure overload (Kuba et al. Circ Res 2007; 101 ,e§2-42).
[0190] Controversial results have been published regarding the involvement of intracellular calcium on the contractility effects of apelin in cardiomyocytes. Two groups described that intracellular calcium is not a signaling mechanism. However, others reported at least a modest increase in the amplitude of the intracellular calcium ion transients in failing rat trabeculae and isolated cardiomyocytes (Dai ef a/.Eur J Pharmacol 2006;553;222-228; Wang et al. Am J Physiol heart circ Physiol 2008;294;H2540-46. Additionally, effects of apelins in pre-clinical models have been described. Apelins may have an important counter- regulatory role to vasopressin and hence fluid homoeostasis. Apelin and the APJ receptor are both expressed also in the kidney and many areas of the brain. Synthesis in certain brain regions involved in fluid homeostasis are regulated by vasopressin. To the contrary, intracerebral injection of apelin directly inhibits vasopressin release leading to a 40% reduction in plasma vasopressin concentrations (Reaux-Le Goazigo et al. Endocrinology 2004; 145 :4392- 4400).
[0191] A link of apelins to metabolic syndrome is suggested by pre-clinical data. Apelins are produced by adipose tissue and may influence glucose and lipid metabolism as adipocytokines (Boucher et al. Endocrinology 2005; 146: 1764-1771 ). Acute intravenous administration of 'pyf-apelin- 13 stimulates glucose utilization in normal and obese insulin- resistant mice (Dray C et al. Cell Metab 2008;8:437-445). These acute effects were explained by a direct effect of 'pyr-apelin- 13 on glucose uptake into skeletal muscle. Mice deficient for the apelins have reduced insulin sensitivity which can be corrected by sub-chronic supplementation with apelin via minipumps. Furthermore in insulin resistant homozygous leptin receptor mutant mice (db/db mice) a similar sub-chronic adminstration results in improved glucose utilization (Yue et al. Am J Physiol Endocrinol Metab 2009; 298:E59-67). Results with glucose utilization in apelin receptor knockout mice have not been published. Furthermore it is not reported yet, whether apelins significantly affect glucose handling in man.
[0192] The clinical and pre-clinical profile suggests applications of apelin receptor agonists in different patient populations and indications. In heart failure, apelins demonstrate a unique hemodynamic profile in enhancing myocardial contractility without inducing left ventricular hypertrophy. In parallel, ventricular pre- and afterload is reduced by reduced peripheral resistance. In pre-clincal models, apelin increases contractility at least to the same extent in the failing compared to normal myocardium (Dai et al. Eur J Pharmacol
2006;553:222-228). Irrespective of changes in receptor and ligand expression, these studies indicate agonism of the receptor is not diminished in situations of established heart failure. First data from clinical studies with acute apelin infusions are promising. In contrast to acetylcholine, another vaso-active principle, vascular and cardiac hemodynamic effects of apelins are preserved in chronic heart failure patients (Japp et al. Circulation 2010; 121 : 1818- 1827). These patients received optimal pharmacological treatment, suggesting that the effects of apelin were additive to established heart failure therapies like ACE-lnhibitors and/or β- blockers.
[0193] Regarding therapies targeting the diseased heart, acute beneficial effects of apelins after acute myocardial infarction may be envisaged. Two groups reported that in preclinical models of acute myocardial ischemia and reperfusion administration of apelins at reperfusion strongly reduces myocardial injury ( leinz et al. Regul Pept 2008; 146:271 -277; Simpkin et al. Basic Res Cardiol 2007; 102:518-28). Both groups published opposing results regarding the underlying signaling of this cardioprotective mechanism. Simpkin et al favor a mechanism based on activation of phosphatide-3- kinase, AKT kinase and P70S6 kinase, whereas Kleinz et al could not confirm activation of this pathway However, signaling pathways independent of PI-3-kinase, AKT-kinase and p70S6 kinase may also explain the benefical effects of apelin receptor agonists in ischemia-reperfusion injury. Apelin increases both phosphorylation and activity of key components within reperfusion injury salvage kinase pathway (Smith et al. Cardiovasc Drugs Ther 2007;21 :409-414). This pro- survival pathway is known to be associated with reduced ischemia-reperfusion-injury by preserving mitochondrial function. Despite the fact, preconditioning agents are difficult to implement in clinical practice, apelin receptor agonists may be administered with the reperfusion solution directly after acute myocardial infarction and thereby display potential benefits in both restoring cardiac survival and function. Another application, especially of oral bioavailable small molecule apelin receptor agonists, could be to start in a patient with an acute myocardial infarction with an intravenous formulation during reperfusion and continue later, e.g., outside the clinic, with an oral bioavailable formulation of the same drug component. Furthermore, intravenous or oral administration of apelin receptor agonists could be envisaged in patients with acute heart failure. Very often acute heart failure develops in the progression of chronic heart failure spontaneously as acute episodes of disease worsening but without signs of myocardial infarction. Patients are then hospitalized and stabilized during hospitalization by agents increasing the contractility of the disease heart muscle. Apelin receptor agonists display a unique hemodynamic profile suggesting a safe and efficient use in such patients.
[0194] Agonists of the apelin receptor may also represent a novel class of antihypertensive agents. In preclinical models, administration of apelin peptides lowers blood pressure, greatly enhanced in hypertensive animals compared with normotensive controls. In first clinical studies modest but significant effects on blood pressure lowering could be demonstrated in normotensive middle-aged subjects. Whether intravenously applied apelin peptides lower blood pressure stronger in hypertensive patient populations, similar to the situation in normotensive vs. hypertensive rats, needs to be evaluated. Application of apelin peptides in hypertensive patients is strongly limited by the need of intravenous administration route. However, small molecule apelin receptor agonists as claimed in this patent application may have a much wider application in these patients due to better oral bioavailability.
[0195] Apelin receptor agonists appear to have beneficial effects on additional vascular based diseases. In atherosclerotic mice deficient for the Apolipoprotein-E, apelin infusion inhibits atherosclerosis progress and completely abrogates angiotensin II- accelerated detrimental effects independent of blood pressure (Chun et al. J Clin Invest 2008; 1 1 8:3343- 3354). And in double knockout mice, deficient in for the apelin receptor ligand and
Apolipoprotein-E, accelerated atherosclerosis could be observed compared versus single Apolipoprotein-E-knockout. It needs to be mentioned, that also pro-atherosclerotic effects of the apelin receptor have been described in a combined mice knockout-model of the apelin receptor and apolipoprotein-E ApoE (Hashimoto et al. Am J Pathol 2007: 108: 1432-1438). Overall these results are difficult to reconcile: Most probably very different fat feeding regimens or different genetic backgrounds and so called off-target genetic effects best explain the observed differences. Independent of effects on atherosclerosis progression, apelin treatment resulted in reduced aneurysm by 50% in a mouse model of abdominal aortic aneurysms (Leeper et al. Am J Physiol Heart Circ Physiol 2009;296:H 1329- 1335), an effect explained by the authors by a direct anti-inflammatory effect within the vessel wall.
[0196] Furthermore, apelin receptor agonists may play an important role in maturation of newly formed blood vessels. idoya et al. (Blood 2010; 1 15;3 166-3174) recently described in a model of vascular remodeling after hind limb ischemia in mice, that apelins induce the maturation into enlarged and non-leaky blood vessels for functional recovery. Especially pathologically increased vascular permeability induced by VEGF under hypoxic conditions seems to be corrected by apelins.
[0197] In humans, apelins cause nitric oxide-mediated vasodilatation in forearm resistance vessels of healthy subjects. Based on promising preclinical data, the role of apelin receptor agonists in preventing human vascular disease merits further investigations. These investigations will be strongly facilitated by small molecule apelin receptor agonists, as claimed in this patent application, because the oral bioavailability allows for much easier chronic administration routes.
[0198] In patients with metabolic syndrome and diabetes, apelin receptor agonists may provide additional benefits. Apelins are produced also by adipose tissue and influence glucose and lipid metabolism as adipocytokines. Mice with no apelin receptor ligands have reduced insulin sensitivity which can be corrected by the administration of exogenous apelin. Acute and sub-chronic positive effects of apelins on glucose utilizations following a glucose load have been described in insulin- resistant animal strains. Although the translation of these effects to man needs to be performed, apelin receptor agonists may offer additional therapeutic options especially in insulin-resistant patients, insufficiently dealing with increased plasma glucose load in metabolic syndrome and diabetes. The simultaneous beneficial effects on blood glucose lowering and vascular and cardiac homeostasis are a unique advantage to therapeutic principles affecting blood glucose alone and open an avenue to macro- and microvascular diabetic late complications, like diabetic cardiomyopathies, diabetic retinophathy, diabetic macular edema, diabetic nephrophathy and diabetic neuropathy. Oral bioavailable small molecule apelin receptor agonist would strongly boost these areas of applications because they would be not restricted to intravenous or
subcutaneous administration routes.
[0199] There continues to be a need for further effective low molecular weight APJ modulators, in particular in view of safety and selectivity. The compounds disclosed herein are selective for the apelin receptor, and have been shown to be highly specific for the apelin receptor versus the angiotensin II receptor (ATI ), the most closely related receptor. Broad selectivity profiling experiments revealed no significant off target binding. The compounds disclosed herein show favorable in vitro absorption, distribution, metabolism, and excretion (ADME) properties and excellent in vivo pharmacokinetics. The compounds of the present disclosure are contemplated to be useful as novel therapies for the treatment for a range of cardiovascular, renal and metabolic conditions.
[0200] The following examples are intended to illustrate but not limit the invention.
EXAMPLE 1
2-[5-(4-Methoxy-phenyl)-4-(2-methoxy-phenyl)-4H-[ l ,2,4]triazol-3-ylsulfanylmethyl]-7- methyl-pyrido[l ,2-a]pyrimidin-4-one
Figure imgf000049_0001
Scheme 1 [0201] Step l :
[0202] The mixture of 4-methoxy-benzoic acid hydrazide (100 mg, 0.60 mmol) and 1 - isothiocyanato-2-methoxy-benzene (99.0 mg, 0.60 mmol) in acetonitrile (2.5 mL) was stirred at room temperature overnight. The reaction was monitored by LC-MS. When it was completed, the mixture was concentrated in vacuum to give 5-(4-methoxy-phenyl)-4-(2- methoxy-phenyl)-4H-[ l ,2,4]triazole-3-thiol ( 189 mg, yield: 95%), which was used directly without further purification.
[0203] 1 Η NMR (400 MHz, DMSO-i/6): δ = 10.47 (s, 1 H), 9.74 (s, 1 H), 9.17 (s, 1 H), 8.03 (s, 1 H), 7.91 (d, J = 8.0 Hz, 2H), 7.14 (t, J = 7.6 Hz, 1 H), 7.04-7.02 (m, 3H), 6.93 (m, 1 H), 3.83 (s, 3H), 3.74 (s, 3H).
[0204] Step 2: [0205] The mixture of 2-(4-methoxybenzoyl)-N-(2- methoxyphenyl)hydrazinecarbothioamide ( 100 mg, 0.30 mmol) and K2CO3 (170 mg, 1.23 mmol) in water (2.0 mL) was heated to reflux. The reaction was monitored by LC-MS. When it was completed, the solution was cooled to room temperature, washed with with Εί2<3 (4 mL), and the aqueous layer was acidified with IN HC1. The precipitate was collected by filtration and sequentially washed with water (4mL) and Et20 (4mL), dried in vacuum to give 5-(4-methoxy-phenyl)-4- (2-methoxy-phenyl)-4H-[ l ,2,4]triazole-3-thiol (70.4 mg, yield: 75%) of as yellow solid.
[0206] Ή NMR (400 MHz, DMSO-i¾): δ = 13.94 (s, 1 H), 7.49 (t, J = 8.4 Hz, 1 H), 7.38 (d, J = 8.0 Hz, 1 H), 7.24 (d, J = 7.6 Hz, 2H), 7.17 (d, J = 8.0 Hz, 1 H), 7.09 (t, J = 8.4 Hz, 1 H), 6.88 (d, J = 7.6 Hz, 2H), 3.72 (s, 3H), 3.59 (s, 3H).
[0207] Step 3:
[0208] To a solution of 5-(4-methoxy-phenyl)-4-(2-methoxy-phenyl) -4H-[ 1 ,2,4]triazole- 3-thiol (40.0 mg, 0.130 mmol) in acetone (2 mL) was added 2-chloromethyl -7-methyl- pyrido[l ,2-a]pyrimidin-4-one (27.0 mg, 0.13 mmol) and K2C03 (22 mg, 0.16 mmol). Then the mixture was warmed to 60°C and stirred until the reaction was completed. Aftet cooling to room temperature, the reactant was concentrated to dryness in vacuum. The residue was diluted with DCM (5 mL) and the suspension was washed with aqueous NaHC03 (5 mL). The organic layer was dried over anhydrous Na2SC>4, filtered and concentrated. The residue was purified by Pre-TLC (EA) to give 2-[5-(4-methoxy-phenyl)-4-(2-methoxy-phenyl)-4H- [l ,2,4]triazol-3-ylsulfanylmethyl]-7-methyl-pyrido[ l ,2-a]pyrimidin-4-one (63.1 g , yield: 76%) as a yellow solid.
[0209] Ή NMR(400 MHz, CDC13): 5 = 8.82 (s, 1 H), 7.58 (dd, J = 8.8, 2.0 Hz, 1 H), 7.51 (d, J= 9.2 Hz, 1 H), 7.43 (t, J= 8.8 Hz, 1 H), 7.36 (d, J= 8.8 Hz, 2H), 7.15 (dd, J= 8.0, 2.0 Hz, 2H), 7.04-6.90 (m, 2H), 6.77 (d, J= 8.8 Hz, 2H), 6.55 (s, 1 H), 4.48-4.40 (m, 2H), 3.76 (s, 3H), 3.66 (s, 3H), 2.42 (s, 3H). MS: m/z 486.2 (M+H+).
EXAMPLE 2
2-[5-Furan-2-yl-4-(2-methoxy-phenyl)-4H-[ l ,2,4]triazol-3-ylsulfanylmethyl]-7-methyl- pyrido[ 1 ,2-a]pyrimidin-4-one
Figure imgf000051_0001
[0210] Ή NMR (400 MHz, CDC13): 5 = 8.82 (s, IH), 7.58 (dd, J= 9.2, 2.0 Hz, IH), 7.56- 7.47 (m, 2H), 7.38 (d, J= 0.8 Hz, IH), 7.23 (dd, J= 8.0, 1.6 Hz, IH), 7.09-7.03 (m, 2H), 6.54 (s, IH), 6.31 (dd,J=3.2, 1.6 Hz, IH), 6.19 (d, J= 3.2 Hz, IH), 4.45 (s, 2H), 3.71 (s, 3H), 2.41 (s, 3H). MS: m/z 446.2 (M+H+).
EXAMPLE 3
2-[4-(2-Methoxy-phenyl)-5-phenyl-4H-[l,2,4]triazol-3-ylsulfanylmethyl]-7-methyl- pyrido[l,2-a]pyrimidin-4-one
Figure imgf000051_0002
[0211] This compound was prepared as described in EXAMPLE 1.
[0212] Ή NMR (300 MHz, DMSO-i0): 5 = 8.78 (s, IH), 7.86 (dd, J= 9.0, 1.8 Hz, IH), 7.58 (d,J=9.0 Hz, IH), 7.52 (td,J= 8.1, 1.5 Hz, IH), 7.43-7.29 (m, 6H), 7.22 (d,J= 8.1 Hz, IH), 7.04 (t, J= 7.5 Hz, IH), 6.43 (s, IH), 4.35 (s, 2H), 3.62 (s, 3H), 2.40 (s, 3H). MS: m/z 456.2 (M+H+).
EXAMPLE 4
[0213] 2-[4-(2-Methoxy-phenyl)-5-pyridin-4-yl-4H-[l,2,4]triazol-3-ylsulfanylmethyl]-7- methyI-pyrido[l,2-a]pyrimidin-4-one
Figure imgf000051_0003
[0214] This compound was prepared as described in EXAMPLE 1. [0215] Ή NMR (300 MHz, CDC13): δ = 8.82 (s, IH), 8.53 (d, J= 5.1 Hz, 2H), 7.61 (dd, J = 9.0, 2.1 Hz, IH), 7.53-7.46 (m, 2H), 7.33 (d, J= 5.7 Hz, 2H), 7.22 (d, J= 1.5 Hz, IH), 7.07 (d,J= 7.5 Hz, IH), 7.02 (d,J= 9.0 Hz, IH), 6.58 (s, IH), 4.50 (s, 2H), 3.64 (s, 3H), 2.42 (s, 3H). MS: m/z 457.2 (M+H+).
EXAMPLE 5
2-[4-(2-Methoxy-phenyl)-5-p-tolyI-4H-[l,2,4]triazol-3-ylsulfanylmethyl]-7-methyl- pyridof 1 ,2-a]pyrimidin-4-one
Figure imgf000052_0001
[0216] This compound was prepared as described in EXAMPLE 1.
[0217] Ή NMR (400 MHz, CDC13): δ = 8.82 (s, IH), 7.58 (dd,J= 9.2, 2.0 Hz, IH), 7.51 (d, J= 8.8 Hz, IH), 7.43 (td, J= 8.0, 2.0 Hz, IH), 7.31 (d, J= 8.0 Hz, 2H), 7.15 (dd, J= 8.0, 1.6 Hz, IH), 7.05 (d, J= 8.0 Hz, 2H), 7.05-6.93 (m, 2H), 6.56 (s, IH), 4.45 (d, J= 4.0 Hz, 2H), 3.65 (s, 3H), 2.42 (s, 3H), 2.30 (s, 3H). MS: m/z 470.2 (M+H+).
EXAMPLE 6
2-[5-(4-Dimethylamino-phenyl)-4-(2-methoxy-phenyl)-4H-[l,2,4]triazol-3- ylsulfanylmethyl]-7-methyl-pyrido[ 1 ,2-a]pyrimidin-4-one
Figure imgf000052_0002
[0218] This compound was prepared as described in EXAMPLE 1.
[0219] Ή NMR (400 MHz, CDCI3): δ = 8.82 (s, 1 H), 7.57 (dd, J = 9.2, 2.0 Hz, IH), 7.51 (d, J=5.2 Hz, IH), 7.43 (td,J=8.0, 1.6 Hz, IH), 7.29 (d,J=8.8Hz, 2H), 7.14 (d, J= 7.6, 1.6 Hz, IH), 7.02-6.94 (m, 2H), 6.56-6.52 (m, 2H), 6.52 (s, IH), 4.46-4.35 (m, 2H), 3.69 (s, 3H), 2.92 (s, 6H), 2.41 (s, 3H). MS: m/z 499.2 (M+H+). EXAMPLE 7
2-[4-(2-Methoxy-phenyl)-5-(4-nitro-phenyl)-4H-[l,2,4]triazol-3-ylsulfanylmethyl]-7-methyl- pyrido[ 1 ,2-a]pyrimidin-4-one
Figure imgf000053_0001
[0220] This compound was prepared as described in EXAMPLE 1.
[0221] Ή NMR (400 MHz, CDC13): δ = 8.83 (s, 1H), 8.13 (d, J = 8.4 Hz, 2H), 7.63 (d, J = 8.4 Hz, 2H), 7.60 (dd, J= 9.2, 2.0 Hz, 1H), 7.56-7.48 (m, 2H), 7.23 (dd, J= 7.6, 1.6 Hz, 1H), 7.09-7.04 (m, 1H), 7.04-6.99 (m, 1H), 6.59 (s, 1H),4.52 (d,J=1.6 Hz, 2H), 3.64 (s, 3H), 2.42 (s, 3H). MS: m/z 501.2 (M+H+).
EXAMPLE 8
2-[5-(5-Bromo-thiophen-2-yl)-4-(2-methoxy-phenyl)-4H-[l,2,4]triazol-3-ylsulfanylmethyl]- 7-methyI-pyrido[ 1 ,2-a]pyrimidin-4-one
Figure imgf000053_0002
[0222] This compound was prepared as described in EXAMPLE 1.
[0223] Ή NMR (300 MHz, CDC13): δ = 8.82 (s, 1H), 7.62-7.49 (m, 3H), 7.26-7.21 (m, 1H), 7.12-7.03 (m, 2H), 6.85 (dd, 7=3.9, 1.5 Hz, 1H), 6.73 (d,J=3.9, 1.5 Hz, 1H), 6.57 (s, 1H), 4.45 (s, 2H), 3.74 (s, 3H), 2.42 (s, 3H). MS: m/z 524.1 (M+H+).
EXAMPLE 9 -[5-(5-Chloro-thiophen-2-yl)-4-(2-methoxy-phenyI)-4H-[l,2,4]triazol-3-ylsulfanylmethyl]- -methyl-pyrido[l,2-a]pyrimidin-4-one
Figure imgf000054_0001
[0224] This compound was prepared as described in EXAMPLE 1.
[0225] Ή NMR (300 MHz, CDC13): δ = 8.82 (s, IH), 7.62-7.48 (m, 3H), 7.23 (dd, J = 7.8, 1.5 Hz, IH), 7.11-7.03 (m, 2H), 6.75 (d,J=6.9 Hz, IH), 6.71 (d,J=6.9 Hz, IH), 6.57 (s, IH), 4.45 (s, 2H), 3.74 (s, 3H), 2.42 (s, 3H). MS: m/z 497.1 (M+H+).
EXAMPLE 10
2-[5-(5-Bromo-furan-2-yl)-4-(2-methoxy-phenyl)-4H-[l,2,4]triazol-3-ylsulfanylmethyl]-7- methyl-pyrido[ 1 ,2-a]pyrimidin-4-one
Figure imgf000054_0002
[0226] This compound was prepared as described in EXAMPLE 1.
[0227] Ή NMR (300 MHz, CDC13): δ = 8.82 (s, IH), 7.59 (dd, J= 8.1, 2.1 Hz, IH), 7.55- 7.47 (m, 2H), 7.22 (dd,J=8.1, 1.5 Hz, IH), 7.09-7.01 (m, 2H), 6.54 (s, IH), 6.25 (d, J= 3.3 Hz, IH), 6.21 (d, J= 3.3 Hz, IH), 4.46 (s, 2H), 3.73 (s, 3H), 2.42 (s, 3H). MS: m/z 524.1 (M+H+).
EXAMPLE 11
2-[4-(2-Methoxy-phenyl)-5-(5-methyl-thiophen-2-yl)-4H-[l,2,4]triazol-3-ylsulfanylmethyl]- 7-methyl-pyrido[ 1 ,2-a]pyrimidin-4-one
Figure imgf000054_0003
[0228] This compound was prepared as described in EXAMPLE 1.
[0229] Ή NMR (400 MHz, CDC13): δ = 8.82 (s, IH), 7.58 (dd, J= 8.8, 1.6 Hz, IH), 7.55- 7.48 (m, 2H), 7.22 (d, J= 6.8 Hz, IH), 7.08-7.01 (m, 2H), 6.74 (d, J= 4.0 Hz, IH), 6.56 (s, I H), 6.54 (d, J= 3.2 Hz, I H), 4.47-4.38 (m, 2H), 3.72 (s, 3H), 2.41 (s, 3H), 2.40 (s, 3H). MS: m/z 476.2 (M+H+).
EXAMPLE 12
2-[4-(2-Methoxy-phenyI)-5-(3-methyl-thiophen-2-yl)-4H-[ l ,2,4]triazol-3-ylsulfanylmethyl]- 7-methyl-pyrido[ 1 ,2-a]pyrimidin-4-one
Figure imgf000055_0001
[0230] This compound was prepared as described in EXAMPLE 1 .
[0231] Ή NMR (400 MHz, CDCI3): 5 = 8.82 (s, I H), 7.59 (dd, J = 8.8, 1.6 Hz, I H), 7.52 (d, J = 8.8 Hz, I H), 7.45 (t, J = 8.4 Hz, I H), 7.18-7.10 (m, 2H), 7.06-6.95 (m, 2H), 6.82 (d, J = 5.2 Hz, I H), 6.59 (s, I H), 4.53-4.47 (m, 2H), 3.71 (s, 3H), 2.42 (s, 3H), 2.39 (s, 3H). MS: m/z 476.2 (M+H+).
EXAMPLE 13
4-[4-(2-Methoxy-phenyl)-5-(7-methyl-4-oxo-4H-pyrido[l ,2-a]pyrimidin-2- y lmethylsulfanyl)-4H-[ 1 ,2,4]triazol-3-yl]-benzonitrile
Figure imgf000055_0002
[0232] This compound was prepared as described in EXAMPLE 1 .
[0233] Ή NMR (400 MHz, CDCI3): δ = 8.82 (s, I H), 7.63-7.46 (m, 7H), 7.21 (dd, j = 1.6 Hz, I H), 7.08-6.96 (m, 2H), 6.58 (s, I H), 4.53-4.47 (m, 2H), 3.64 (s, 3H), 2.42 (s, 3H) MS: m/z 481 .2 (M+H+).
EXAMPLE 14
2-[4-(2-Methoxy-phenyl)-5-thiazol-2-yl-4H-[ l ,2,4]triazol-3-ylsulfanylmethyl]-7-methyl- pyridof 1 ,2-a]pyrimidin-4-one
Figure imgf000056_0001
[0234] This compound was prepared as described in EXAMPLE 1.
[0235] Ή NMR (400 MHz, CDC13): 5 = 8.82 (s, 1H), 7.65 (d, J= 3.2 Hz, 1H), 7.59 (dd, J = 8.8,2.0 Hz, 1H), 7.52 (d, J =9.2 Hz, 1H), 7.48 (t, J=8.4 Hz, 1H), 7.32 (d,J= 3.2 Hz, 1H), 7.29 (dd,J=7.6, 1.6 Hz, IH), 7.05 (t,J=8.4 Hz, 1H), 7.00 (d,J=8.0 Hz, 1H), 6.58 (s, 1H), 4.54-4.48 (m, 2H), 3.64 (s, 3H), 3.42 (s, 3H). MS: m/z 463.2 (M+H+).
EXAMPLE 15
2-[4-(2-Methoxy-phenyl)-5-(5-methyl-furan-2-yl)-4H-[l,2,4]triazol-3-ylsulfanylmethyl]-7- methyl-pyrido[ 1 ,2-a]pyrimidin-4-one
Figure imgf000056_0002
[0236] This compound was prepared as described in EXAMPLE 1.
[0237] Ή NMR (400 MHz, CDCL-): δ = 8.82 (s, 1H), 7.58 (dd, J= 8.8, 1.6 Hz, 1H), 7.54- 7.47 (m, 2H), 7.21 (d, J=7.6 Hz, 1H), 7.07-7.01 (m, 2H), 6.53 (s, 1H), 5.91-5.87 (m, 2H), 4.44 (s, 2H), 3.72 (s, 3H), 2.42 (s, 3H), 2.25 (s, 3H). MS: m/z 460.2 (M+H+).
EXAMPLE 16
2-[5-Furan-3-yl-4-(2-methoxy-phenyl)-4H-[l,2,4]triazol-3-ylsulfanylmethyl]-7-methyl- pyrido[ 1 ,2-a]pyrimidin-4-one
Figure imgf000056_0003
[0238] This compound was prepared as described in EXAMPLE 1. [0239] Ή NMR (400 MHz, DMSO-d6): δ = 8.78 (s, IH), 7.86 (dd, J= 8.8, 2.0 Hz, IH), 7.71 (s, IH), 7.64-7.56 (m, 2H), 7.41 (d,J=8.8Hz, IH), 7.33-7.27 (m, 2H), 7.11 (t,J=7.6 Hz, IH), 6.46 (s, IH), 6.41 (s, IH), 4.37-4.27 (m, 2H), 3.69 (s, 3H), 2.40 (s, 3H). MS: m/z 446.2 (M+H+).
EXAMPLE 17
2-[4-(2-Methoxy-phenyl)-5-thiophen-3-yl-4H-[l,2,4]triazol-3-ylsulfanylmethyl]-7-methyl- pyrido[ 1 ,2-a]pyrimidin-4-one
Figure imgf000057_0001
[0240] This compound was prepared as described in EXAMPLE 1.
[0241] Ή NMR (400 MHz, CDC13): δ = 8.82 (s, IH), 7.59 (dd, J= 8.4, 1.6 Hz, IH), 7.54- 7.46 (m, 2H), 7.32 (dd, J= 4.8, 1.6 Hz, IH), 7.23-7.20 (m, 2H), 7.08-7.04 (m, 3H), 6.57 (s, IH), 4.47-4.43 (m, 2H), 3.69 (s, 3H), 2.42 (s, 3H). MS: m/z 462.2 (M+H+).
EXAMPLE 18
2-[4-(2-Methoxy-phenyl)-5-(l -methyl- lH-pyrrol-2-yl)-4H-[l,2,4]triazol-3-ylsulfanylmethyI]- 7-methyl-pyrido[l,2-a]pyrimidin-4-one
Figure imgf000057_0002
[0242] This compound was prepared as described in EXAMPLE 1.
[0243] Ή NMR (400 MHz, DMSO-fl¾): δ = 8.78 (s, IH), 7.86 (dd, J= 8.4, 1.6 Hz, IH), 7.59 (d, J= 9.6 Hz, IH), 7.53 (t, J= 7.6 Hz, IH), 7.33 (d, J= 7.6 Hz, IH), 7.24 (d, J= 8.0 Hz, IH), 7.05 (t,J=7.6 Hz, IH), 6.90 (s, IH), 6.43 (s, IH), 5.94-5.85 (m, IH), 5.57-5.45 (m, IH), 4.33 (s, 2H), 3.83 (s, 3H), 3.66 (s, 3H), 2.40 (s, 3H). MS: m/z 459.2 (M+H+). EXAMPLE 19
2-[4-(2-Methoxy-phenyl)-5-( 1 -methyl- 1 H-imidazol-2-yl)-4H-[ 1 ,2,4]triazol-3- ylsulfanylmethyl]-7-methyl-pyrido[l,2-a]pyrimidin-4-one
Figure imgf000058_0001
[0244] This compound was prepared as described in EXAMPLE 1.
[0245] Ή NMR (400 MHz, OMSO-d6): δ = 8.78 (s, IH), 7.86 (dd, J= 8.8, 1.6 Hz, IH), 7.59 (d, J= 8.8 Hz, IH), 7.44 (t, J= 8.8 Hz, IH), 7.34-7.28 (m, 2H), 7.14 (d, J= 8.0 Hz, IH), 6.99 (t, J=8.8Hz, IH), 6.81 (s, IH), 6.43 (s, lH),4.38(s, 2H), 3.91 (s, 3H), 3.59 (s, 3H), 2.40 (s, 3H). MS: m/z 460.2 (M+H+).
EXAMPLE 20
2-[4-(4-Methoxy-phenyl)-5-thiophen-2-yl-4H-[l,2,4]triazol-3-ylsulfanylmethyl]-7-methyl- pyridof 1 ,2-a]pyrimidin-4-one
Figure imgf000058_0002
Scheme 2
[0246] This compound was prepared as described in EXAMPLE 1.
[0247] 'HNMR (400 MHz, CDC13): 5 = 8.83 (s, IH), 7.59 (dd, J= 9.2, 2.0 Hz, IH), 7.52 (d,J=8.8Hz, IH), 7.30 (dd, J =4.8, 1.6 Hz, IH), 7.22 (d,J=8.8Hz, 2H), 7.01 (d,J=8.8 Hz, 2H), 6.95-6.88 (m, 2H), 6.60 (s, IH), 4.47 (s, 2H), 3.88 (s, 3H), 2.42 (s, 3H). MS: m/z 462.1 (M+H+).
EXAMPLE 21
7-Methyl-2-(5-thiophen-2-yl-4-m-tolyl-4H-[l,2,4]triazol-3-ylsulfanylmethyl)-pyrido[l,2- a]pyrimidin-4-one
Figure imgf000059_0001
[0248] This compound was prepared as described in EXAMPLE 1.
[0249] 'HNMR (300 MHz, CDC13): δ = 8.85 (s, IH), 7.63-7.52 (m, 2H), 7.47-7.35 (m, 2H), 7.32-7.30 (m, IH), 7.13 (s, 2H), 6.95-6.89 (m, IH), 6.87-6.86 (m, IH), 6.62 (s, IH), 4.50 (s, 2H), 2.43 (d, J= 5.4 Hz, 6H). MS: m/z 446.1 (M+H+).
EXAMPLE 22
2-[4-(3-Methoxy-phenyl)-5-thiophen-2-yl-4H-[l,2,4]triazol-3-ylsulfanylmethyl]-7-methyl- pyrido[l,2-a]pyrimidin-4-one
Figure imgf000059_0002
[0250] This compound was prepared as described in EXAMPLE 1.
[0251] 'HNMR (400 MHz, CDC13) δ = 8.83 (s, IH), 7.59 (dd, J= 8.8, 2.0 Hz, IH), 7.52 (d, J =8.8 Hz, IH), 7.42 (t, J =8.0 Hz, IH), 7.31 (dd, J= 9.2, 2.0 Hz, IH), 7.09 (dd, J =4.0, 2.8 Hz, IH), 6.97-6.85 (m, 3H), 6.82 (t, J= 2.0 Hz, IH), 6.60 (s, IH), 4.48 (s, 2H), 3.81 (s, 3H), 2.42 (s, 3H). MS: m/z 462.1 (M+H+).
EXAMPLE 23
7-Methyl-2-(5-thiophen-2-yl-4-o-tolyl-4H-[l,2,4]triazol-3-ylsulfanylmethyl)-pyrido[l,2- a]pyrimidin-4-one
Figure imgf000060_0001
[0252] This compound was prepared as described in EXAMPLE 1 .
[0253] 'HNMR (400 MHz, CDC13) δ = 8.84 (s, I H), 7.59 (dd, J= 8.8, 2.4 Hz, I H), 7.53- 7.48 (m, 2H), 7.40-7.35 (m, 2H), 7.26 (overlap, 2H), 6.91 -6.87 (m, 1 H), 6.85 (dd, J = 3.6, 1.2 Hz, I H), 6.62 (s, I H), 4.52 (s, 2H), 2.42 (s, 3H), 2.02 (s, 3H). MS: m/z 446. 1 (M+H+).
EXAMPLE 24
2-[4-(2-Fluoro-phenyl)-5-thiophen-2-yl-4H-[l,2,4]triazol-3-ylsulfanylmethyl]-7-methyl- pyrido[ 1 ,2-a]pyrimidin-4-one
Figure imgf000060_0002
[0254] This compound was prepared as described in EXAMPLE 1 .
[0255] 'HNMR (400 MHz, CDCI3) δ = 84 (s, 1 H), 7.65-7.55 (m, 2H), 7.52 (d, J= 10.0 Hz, I H), 7.38-7.29 (m, 4H), 7.04-6.87 (m, 2H), 6.58 (s, I H), 4.57-4.37 (m, 2H), 2.42 (s, 3H). MS: m/z 450.1 (M+H+).
EXAMPLE 25
7-Methyl-2-[5-thiophen-2-yl-4-(4-trifluoromethoxy-phenyl)-4H-[ l ,2,4]triazol-3- ylsulfanylmethyl]-pyrido[ l ,2-a]pyrimidin-4-one
Figure imgf000060_0003
[0256] This compound was prepared as described in EXAMPLE 1 . [0257] 'HNMR (400 MHz, CDCI3) δ = 8.84 (s, IH), 7.61 (dd, J= 9.2, 1.6 Hz, IH), 7.52 (d,J=8.8Hz, IH), 7.37 (s, 4H), 7.34 (dd,J=5.0, 1.2 Hz, 1H), 6.97-6.92 (m, IH), 6.90 (dd, J= 3.6, 1.2 Hz, 1 H), 6.59 (s, 1 H), 4.49 (s, 2H), 2.43 (s, 3H). MS: m/z 516.1 (M+H+).
EXAMPLE 26
2-[4-(4-Chloro-phenyl)-5 hiophen-2-yl-4H-[l,2,4]triazol-3-ylsulfanylmethyl]-7-methyl- pyridof 1 ,2-a]pyrimidin-4-one
Figure imgf000061_0001
[0258] This compound was prepared as described in EXAMPLE 1.
[0259] 'HNMR (400 MHz, CDC13) 5 = 8.83 (s, IH), 7.61 (dd,J=9.0, 1.8 Hz, IH), 7.53 (s, IH), 7.49 (d, J= 8.8 Hz, 2H), 7.33 (d, J = 4.4 Hz, IH), 7.24 (s, 2H), 6.96-6.92 (m, IH), 6.90 (d,J= 3.2 Hz, IH), 6.57 (s, IH), 4.47 (s, 2H), 2.43 (s, 3H). MS: m/z 466.1 (M+H+).
EXAMPLE 27
7-Methyl-2-[5-thiophen-2-yl-4-(2-trifluoromethoxy-phenyl)-4H-[l,2,4]triazol-3- ylsulfanylmethyl]-pyrido[l,2-a]pyrimidin-4-one
Figure imgf000061_0002
[0260] This compound was prepared as described in EXAMPLE 1.
[0261] 'HNMR (400 MHz, CDC13) δ = 8.84 (s, IH), 7.70-7.56 (m, 2H), 7.53-7.40 (m, 4H), 7.32 (dd, J= 4.6, 1.0 Hz, 1 H), 6.89-6.88 (m, 2H), 6.59 (s, 1 H), 4.56-4.43 (m, 2H), 2.42 (s,3H). MS: m/z 516.1 (M+H+). EXAMPLE 28
2-[4-(3-Chloro-phenyl)-5-thiophen-2-yl-4H-[l ,2,4]triazol-3-ylsulfanylmethyl]-7-methyl- pyrido[ l ,2-a]pyrimidin-4-one
Figure imgf000062_0001
[0262] This compound was prepared as described in EXAMPLE 1.
[0263] ' HNMR (CDCb, 300 MHz) δ = 8.85 (s, I H), 7.62 (d, J = 9.0 Hz, I H), 7.52 (m, 3H), 7.35 (d, J = 5.4 Hz, 2H), 7.24 (d, J= 7.8 Hz, 1 H), 6.99 - 6.88 (m, 2H), 6.60 (s, I H), 4.50 (s, 2H), 2.44 (s, 3H). MS: m/z 466.1 (M+H+).
EXAMPLE 29
2-[4-(2-Chloro-phenyl)-5-thiophen-2-yl-4H-[ l ,2,4]triazoI-3-ylsulfanylmethyI]-7-methyl- pyrido[ 1 ,2-a]pyrimidin-4-one
Figure imgf000062_0002
[0264] This compound was prepared as described in EXAMPLE 1.
[0265] 'HNMR (CDC13, 300 MHz) δ = 8.78 (s, 1 H), 7.88-7.57 (m, 7H), 7.02 (t, J = 4.2 Hz, 1 H), 6.76 (d, J= 2.8 Hz, 1 H), 6.46 (s, 1 H), 4.50 (s, 2H), 2.44 (s, 3H). MS: m/z 466.1 (M+H+).
EXAMPLE 30
2-[4-(2-Chloro-phenyl)-5-thiophen-2-yl-4H-[ l,2,4]triazol-3-ylsulfanylmethyl]-7-methyl- pyrido[ l ,2-a]pyrimidin-4-one
Figure imgf000063_0001
[0266] This compound was prepared as described in EXAMPLE 1.
[0267] 'HNMR (CDC , 400 MHz) δ = 8.78 (s, 1 H), 8.34-8.32 (m, 1 H), 7.92 (dd, J= 8.8 Hz, 2.4 Hz, 1 H), 7.86 (d, J = 9.2 Hz, 1 H), 7.07-7.00 (m, 2H), 6.91 (d, J = 3.6 Hz, 1 H), 6.44 (s, 1H), 4.31 (s, 2H), 3.92 (s, 3H), 2.40 (s, 3H). MS: m/z 463.2 (M+H+).
EXAMPLE 31
4-(2-Methoxy-phenyl)-3-(2-phenyl-thiazol-4-ylmethylsulfanyl)-5-thiophen-2-yl-4H- [ l ,2,4]triazole
Figure imgf000063_0002
Scheme 3
[0268] Step 1 :
[0269] To a stirred solution of thiobenzamide (274 mg, 2.0 mmol), in ethanol (10 mL) and THF (4 mL) was added 1 ,3-dichloroacetone (280 mg, 2.2 mmol) at 65 °C, then the result solution was stirred overnight at this temperature. The mixture was cooled, evaporated in vacuum to dryness. The residue was purified by Pre-TLC to afford 4-chloromethyl-2-phenyl- thiazole (270 mg, yield: 64%) as yellow oil.
[0270] Ή NMR (400 MHz, CDC13): S = 8.01 -7.89 (m, 2H), 7.49 -7.41 (m, 3H), 7.31 (s, 1 H), 4.75 (s, 2H).
[0271] Step 2: [0272] To a solution of 4-chloromethyl-2-phenyl-thiazole (50.0 mg, 0.240 mmol) in acetone (10 mL) was added 4-(2-methoxyphenyl)-5-(2-thienyl)-l,2,4-triazole-3-thiol (83.9 mg, 0.290 mmol) and K2C03 (100 mg, 0.720 mmol). Then the mixture was stirred at 60 °C for 2 hrs. After cooling to room temperature, the reactant was filtered. The filtrate was evaporated in vacuum to dryness. The residue was diluted with EA (10 mL). The new suspension was washed with 5% aqueous solution of NaOH (10 mL), then brine (10 mL), dried over anhydrous Na2S04 and evaporated in vacuum to dryness. The residue was purified by Pre-TLC (EA/PE = 1/1) to give 4-(2-methoxy-phenyl)-3-(2-phenyl-thiazol-4- ylmethylsulfanyl)-5-thiophen-2-yl-4H-[l,2,4]triazole (107 mg, yield: 97%) as a yellow solid.
[0273] Ή NMR (400 MHz, DMSO-i¾): S = 7.90-7.86 (m, 2H), 7.65-7.58 (m, 3H), 7.49- 7.46 (m, 3H), 7.38 (dd,J= 7.6, 1.6 Hz, 1H), 7.31 (d,J=4.0 Hz, 1H), 7.10 (t, J = 2.8 Hz, 1H), 7.01 (dd,J= 1.6 Hz, 1H), 6.81-6.80 (m, 1H), 4.49 (s, 2H), 3.68 (s, 3H). MS: m/z 463.1
(M+H+).
EXAMPLE 32
4-(2-Methoxy-phenyl)-3-thiophen-2-yl-5-[2-(4-trifluoromethyl-phenyl)-thiazol-4- ylmethylsulfanyl]-4H-[ 1 ,2,4]triazole
Figure imgf000064_0001
[0274] This compound was prepared as described in EXAMPLE 31.
[0275] Ή NMR (400 MHz, D SO-d6): S = 8.10 (d, J= 3.6 Hz, 2H), 7.84 (d, J= 4.8 Hz, 1H), 7.71 (s, 1H), 7.65-7.61 (m, 2H), 7.39 (dd, J= 7.6, 1.2 Hz, 1H), 7.31 (d,J=7.6 Hz, 1H), 7.11 (t,J=4.8Hz, 1H), 7.01 (dd, J= 4.8, 3.6 Hz, 1H),6.80 (dd,J=3.6, 1.2 Hz, 1H), 4.52 (s, 2H), 3.68 (s, 3H). MS: m/z 531.1 (M+H+).
EXAMPLE 33
3-[2-(4-Fluoro-phenyl)-thiazol-4-ylmethylsulfanyl]-4-(2-methoxy-phenyl)-5-thiophen-2-yl- H-[l,2,4]triazole
Figure imgf000065_0001
[0276] This compound was prepared as described in EXAMPLE 31.
[0277] Ή NMR ( 400 MHz, DMSO-i¾): δ = 7.95-7.92 (m, 2H), 7.65-7.59 (m, 2H), 7.58 (s, IH), 7.40-7.29 (m, 4H), 7.11 (t,J= 1.5 Hz, IH), 7.01 (dd,J=4.0, 1.2 Hz, 2H), 6.82-6.80 (m, IH), 4.48 (s, 2H), 3.68 (s, 3H). MS: m/z 481.1 (M+H+).
EXAMPLE 34
4-(2-Methoxy-phenyl)-3-[2-(4-nitro-phenyl)-thiazol-4-ylmethylsulfanyl]-5-thiophen-2-yl-4H- [l,2,4]triazole
Figure imgf000065_0002
[0278] This compound was prepared as described in EXAMPLE 31.
[0279] Ή NMR (400 MHz,
Figure imgf000065_0003
8.37-8.29 (m, 2H), 8.17-8.13 (m, 2H), 7.81- 7.73 (m, 1 H), 7.68-7.59 (m, 2H), 7.43-7.35 (m, 1 H), 7.31 (d, J = 7.6 Hz, IH), 7.11 -6.99 (m, IH), 7.04 - 6.97 (m, IH), 6.84-6.76 (m, IH), 4.53 (s, 2H), 3.68 (s, 3H). MS: m/z 508.1( M+H+).
EXAMPLE 35
3-[2-(4-Chloro-phenyl)-thiazol-4-ylmethylsulfanyl]-4-(2-methoxy-phenyl)-5-thiophen-2-yl- 4H-[l,2,4]triazole
Figure imgf000065_0004
[0280] This compound was prepared as described in EXAMPLE 31. [0281] Ή NMR (400MHz, DMSO-<¾: S = 7.95-7.87 (m, 2Η), 7.69-7.58 (m, 3H), 7.54 (d, J= 8.4 Hz, 2H), 7.40-7.34 (m, IH), 7.34-7.28 (m, IH), 7.11 (dd, J= 7.6, 4.0 Hz, IH), 7.05- 6.98 (m, IH), 6.83-6.76 (m, IH), 4.48 (s, 2H), 3.68 (s, 3H). MS: m/z 497.1( M+H+).
EXAMPLE 36
4-(2-Methoxy-phenyl)-3-thiophen-2-yl-5-(2-p-tolyl-thiazol-4-ylmethylsulfanyl)-4H- [l,2,4]triazole
Figure imgf000066_0001
(0282] This compound was prepared as described in EXAMPLE 31.
[0283] Ή NMR (300 MHz, DMSO^): <5 = 7.77-7.75 (m, 2H), 7.66-7.58 (m, 2H), 7.52 (s, IH), 7.38 (d, J= 7.5 Hz, IH), 7.32-7.27 (m, 3H), 7.10 (t, J= 4.5 Hz, IH), 7.02-6.98 (m, IH), 6.81-6.80 (m, IH), 4.47 (s, 2H), 3.67 (s, 3H), 2.34 (s, 3H). MS: m/z 477.1 (M+H+).
EXAMPLE 37
4-(2-Methoxy-phenyl)-3-[2-(4-methoxy-phenyl)-oxazol-4-ylmethylsulfanyl]-5-thiophen-2- yl-4H-[l,2,4]triazole
Figure imgf000066_0002
[0284] This compound was prepared as described in EXAMPLE 31.
[0285] Ή NMR (400 MHz, DMSO-t¾): 0 = 7.81 (d, J= 8.8 Hz, 2H), 7.65-7.59 (m, 2H), 7.47 (s, IH), 7.39-7.36 (m, IH), 7.31 (d,J=8.4 Hz, IH), 7.10 (t, J = 7.6 Hz, IH), 7.03-7.00 (m, 3H), 6.80 (d, J= 3.6 Hz, IH), 4.46 (s, 2H), 3.81 (s, 3H), 3.68 (s, 3H). MS: m/z 493.1( M+H+).
EXAMPLE 38
4-(2-Methoxy-phenyl)-3-thiophen-2-yl-5-[2-(4-trifluoromethyl-phenyl)-oxazol-4- ylmethylsulfanyl]-4H-[l,2,4]triazole
Figure imgf000067_0001
[0286] This compound was prepared as described in EXAMPLE 31.
[0287] Ή NMR (400 MHz, DMSO-¾: δ = 8.14 (d, J= 7.6 Hz, 3H), 7.90 (d, 7 = 4.8 Hz, 2H), 7.65-7.61 (m, 2H), 7.45-7.43 (m, IH), 7.30(d,J=8.0 Hz, IH), 7.12 (t, 7= 3.6 Hz, IH), 7.01-6.99 (m, I H), 6.81 (dd,7=3.6, 1.2 Hz, IH), 4.33 (s, 2H), 3.67 (s, 3H). MS: m/z 515.1 (M+H+).
EXAMPLE 39
3-[2-(4-Fluoro-phenyl)-oxazol-4-ylmethylsulfanyl]-4-(2-methoxy-phenyl)-5-thiophen-2-yI- 4H-[l,2,4]triazole
Figure imgf000067_0002
[0288] This compound was prepared as described in EXAMPLE 31
[0289] Ή NMR (400 MHz, DMSO-flfe): δ = 8.05 (s, IH), 8.01-7.96 (m, 2H), 7.65-7.61 (m, 2H), 7.43(d, IH), 7.37 (t, 2H), 7.30 (d, IH), 7.13 (d, IH), 7.01 (d, IHd), 6.81 (d, IHd), 4.29 (s, 2H), 3.67 (s, 3H). MS: m/z 465.1 (M+H+).
EXAMPLE 40
3-[2-(4-Chloro-phenyl)-oxazol-4-ylmethyIsulfanyl]-4-(2-methoxy-phenyl)-5-thiophen-2- l- 4H-[l,2,4]triazole
Figure imgf000067_0003
[0290] This compound was prepared as described in EXAMPLE 31. [0291] Ή NMR (400 MHz, DMSO-<¾): δ = 8.08 (s, IH), 7.94 (d, J= 8.4 Hz, 2H), 7.64- 7.58 (m, 4H), 7.42 (d,J= 1.6 Hz, IH), 7.30 (d, J= 8.0 Hz, IH), 7.11 (t, IH), 7.01 (t, .7=2.0 Hz, lHd), 6.81-6.80 (m, IH), 4.30 (s, 2H), 3.67 (s, 3H). MS: m/z 481.1 (M+H+).
EXAMPLE 41
4-(2-Methoxy-phenyl)-3-thiophen-2-yl-5-(2-p-tolyl-oxazol-4-ylmethylsuIfanyl)-4H- [l,2,4]triazole
Figure imgf000068_0001
[0292] This compound was prepared as described in EXAMPLE 37.
[0293] Ή NMR (400 MHz, OMSO- 6): δ = 8.01 (d, J= 1.2 Hz, IH), 7.82 (d, J= 8.4 Hz, 2H), 7.64-7.59 (m, 2H), 7.43 (dd,J=3.6, 1.2 Hz, IH), 7.35-7.29 (m, 3H), 7.12(t, lH),7.01(t, J= 7.2 Hz, IH), 6.81 (s, IH), 4.29 (s, 2H), 3.67 (s, 3H), 2.37 (s, 3H). MS: m/z 461.1 (M+H+).
EXAMPLE 42
3-[2-(4-Bromo-phenyl)-oxazol-4-ylmethylsulfanyl]-4-(2-methoxy-phenyl)-5-thiophen-2-yl- 4H-[l,2,4]triazole
Figure imgf000068_0002
[0294] This compound was prepared as described in EXAMPLE 31.
[0295] Ή NMR (400 MHz, DMSO-i¾: δ = 8.08 (s, IH), 7.87-7.85 (m, 2H), 7.75-7.72 (m, 2H), 7.65-7.61 (m, 2H), 7.43 (dd,J=3.6, 1.2 Hz, IH), 7.31-7.28 (m, IH), 7.1 l(t, J= 6.8 Hz, 1 H), 7.03-6.99 (m, IH), 6.80 (s, IH), 4.30 (s, 2H), 3.66 (s, 3H). MS: m/z 525.0 (M+H+).
EXAMPLE 43
4-(2-Methoxy-phenyl)-3-thiophen-2-yl-5-[2-(4-trifluoromethoxy-phenyl)-oxazol-4- ylmethylsulfanyl]-4H-[ 1 ,2,4]triazoie
Figure imgf000069_0001
[0296] This compound was prepared as described in EXAMPLE 31.
[0297] Ή NMR (400MHz, DMSO-i¾: δ = 8.10 (s, 1H), 8.06 (d, J = 8.8 Hz, 2H), 7.64- 7.59 (m, 2H), 7.53 (d, J= 8.8 Hz, 1H), 7.44 (d, J= 6.8 Hz, 1H), 7.31(d, J= 4.8 Hz, 1H), 7.12 (t,J=6.0 Hz, 1H), 7.02 (t,J=3.0 Hz, 1H), 6.81 (s, 1H), 4.31 (s, 2H), 3.67 (s, 3H). MS: m/z 531.1 (M+H+).
EXAMPLE 44
2-[4-(2-Methoxy-phenyl)-5-thiophen-2-yl-4H-[l,2,4]triazol-3-ylsulfanylmethyl]-6-methyl- pyrido[ 1 ,2-a]pyrimidin-4-one
Figure imgf000069_0002
Scheme 4
[0298] Step 1 :
[0299] A mixture of 2-amino-6-picoline (324 mg, 3.0 mmol) and ethyl chloroacetate (594 mg, 3.60 mmol) in polyphosphoric acid (10 mL) was heated at 110 °C for 5 hrs. After cooling to room temperature, the mixture was quenched with water (50 mL). The pH value was adjusted to 6-7 with aqueous K2CO3. Then the new mixture was extracted with EA (50 mL *3).the organic layers were washed with brine (50 mL), dried over NaSC^ and filtered. The filtrate was evaporated in vacuum to give 2-chloromethyl-6-methyl-pyrido[l,2-a]pyrimidin- 4-one (360 mg, yield: 58%) as yellow solid.
[0300] Ή NMR (400 MHz, DMSO-<e): δ = 7.68 (dd, J= 4.0, 1.2 Hz, 1H), 7.40 (d, J= 7.6 Hz, 1H), 6.93 (d, J= 6.8 Hz, 1H), 6.36 (s, 1H), 4.58 (s, 2H), 2.93 (s, 3H). [0301] Step 2:
[0302] The procedure is similar to EXAMPLE 31.
[0303] Ή NMR (400MHz, DMSO-</6): δ = 7.71-7.56 (m, 3H), 7.54-7.40 (m, 1H), 7.33 (t, J=7.2 Hz, 2H), 7.19-7.10 (m, 1H), 7.03-6.98 (m, 1H), 6.94 (dd,J=3.6, 1.2 Hz, 1H), 6.84- 6.79 (m, 1 H), 6.30-6.22 (m, 1 H), 4.25 (s, 2H), 3.70 (s, 3H), 2.92 (s, 3H). MS: m/z 462.2
(M+H+).
EXAMPLE 45
7-Methoxy-2-[4-(2-methoxy-phenyl)-5-thiophen-2-yl-4H-[l,2,4]triazol-3- ylsulfanylmethyl]- pyridof 1 ,2-a]pyrim idin-4-one
Figure imgf000070_0001
[0304] This compound was prepared as described in EXAMPLE 44.
[0305] Ή NMR (300 MHz, DMSO-i¾: δ = 8.43 (d, J= 7.6 Hz, 1H), 7.80 (dd, J= 9.6, 3.0 Hz, 1H), 7.68-7.55 (m, 3H), 7.45 (dd,J=7.8, 1.5 Hz, 1H), 7.31 (d,J=3.6 Hz, 1H), 7.14- 7.10 (m, 1H), 7.00 (dd, J= 8.1, 0.9 Hz, 1H), 6.80 (dd, J=3.6, 1.2 Hz, 1H), 6.44 (s, 1H), 4.33 (s, 2H), 3.92 (s, 3H), 3.68 (s, 3H). MS: m/z 478.1 (M+H+).
EXAMPLE 46
7-Bromo-2-[4-(2-methoxy-phenyl)-5-thiophen-2-yI-4H-[l,2,4]triazol-3-ylsulfanylmethyl]- pyridof 1 ,2-a]pyrimidin-4-one
Figure imgf000070_0002
[0306] This compound was prepared as described in EXAMPLE 44.
[0307] Ή NMR (400MHz, OMSO-d6): δ = 8.98 (d, J= 2.8 Hz, 1H), 8.08 (dd, J= 9.2, 2.0 Hz, 1H), 7.63-7.60 (m, 3H), 7.46 (dd,J=7.6, 1.6 Hz, 1H), 7.31 (d,J=4.8Hz, 1H), 7.15- 7.12 (m, IH), 7.00 (dd, J= 5.2, 4.0 Hz, IH), 6.81 (dd, J
(s, 2H), 3.68 (s, 3H). MS: m/z 526.1 (M+H+).
EXAMPLE 47
2-[4-(2-Methoxy-phenyl)-5-thiophen-2-yl-4H-[l,2,4]triazol-3-ylsulfanylmethyl]-4- pyrido[ 1 ,2-a]pyrimidine-7-carbonitrile
Figure imgf000071_0001
[0308] This compound was prepared as described in EXAMPLE 44.
[0309] Ή NMR (400MHz, OMSO-d6): <5 = 9.42(d, J =3.0 Hz, IH), 8.11 (dd, J= 9.2, 2.0 Hz, 1 H), 7.68 (d, J = 1.2 Hz, 1 H), 7.64-7.52 (m, 2H), 7.50-7.44 (m, 1 H), 7.31 (d, J= 8.0 Hz, IH), 7.13 (t, J =8.0 Hz, 1 H), 7.00 (dd, J= 5.2, 4.0 Hz, IH), 6.80-6.77 (m, IH), 6.57 (s, IH), 4.35 (s, 2H), 3.68 (s, 3H). MS: m/z 473.1 (M+H+).
EXAMPLE 48
2-[4-(2-Methoxy-phenyl)-5-thiophen-2-yl-4H-[l,2,4]triazol-3-ylsulfanylmethyl]-7-methyl- pyrido[ 1 ,2-a]pyrimidin-4-one
Figure imgf000071_0002
[0310] This compound was prepared as described in EXAMPLE 44.
[0311] Ή NMR (400MHz, DMSO-i/6): δ = 8.78 (s, 1 H), 7.86 (dd, J= 9.0, 1.8 Hz, IH), 7.59-7.48 (m, 2H), 7.35 (dd,J= 5.1, 1.2 Hz, IH), 7.23-7.20 (m, 2H), 7.00 (td, J= 7.5, 0.9 Hz, IH), 6.94-6.90 (m, IH), 6.82-6.78 (m, IH), 4.21 (s, 2H), 3.66 (s, 3H), 2.50 (s, 3H). MS: m/z 62.2 (M+H+). EXAMPLE 49
7-Fluoro-2-[4-(2-methoxy-phenyl)-5-thiophen-2-yl-4H-[l,2,4]triazol-3-ylsulfanylmethyl]- pyrido[l,2-a]pyrimidin-4-one
Figure imgf000072_0001
[0312] This compound was prepared as described in EXAMPLE 44.
[0313] Ή NMR (400MHz, DMSO-ifc): δ = 8.92 (dd, J= 6.8, 3.2 Hz, IH), 8.13-8.09 (m, IH), 7.77-7.74 (m, IH), 7.66-7.60 (m, 2H), 7.46 (dd,J=7.6, 4.0 Hz, IH), 7.31 (d,J=4.0 Hz, IH), 7.13 (t, J= 3.6 Hz, IH), 7.04-6.99 (m, IH), 6.85-6.80 (m, IH), 6.50 (s, IH), 4.36 (s, 2H), 3.68 (s, 3H). MS: m/z: 466.1 (M+H+).
EXAMPLE 50
7-Chloro-2-[4-(2-methoxy-phenyl)-5-thiophen-2-yl-4H-[l,2,4]triazol-3-yIsulfanylmethyl]- pyrido[ 1 ,2-a]pyrimidin-4-one
Figure imgf000072_0002
[0314] This compound was prepared as described in EXAMPLE 44.
[0315] Ή NMR (400MHz, DMSO-¼): δ = 12.60 (IH, s), 8.02 (dd, J= 4.8, 4.0 Hz, IH), 7.72-7.59 (m, 3H), 7.47 (dd, J = 2.4, 2.4 Hz, IH), 7.31 (s, IH), 7.13 (t, J= 5.2 Hz, IH), 7.00- 6.98 (m, IH), 6.86-6.78 (m, IH), 6.52 (s, IH), 4.35 (s, 2H), 3.68 (s, 3H). MS: m/z: 482.1 (M+H+).
EXAMPLE 51
2-[4-(2-Methoxy-phenyl)-5-thiophen-2-yl-4H-[l,2,4]triazol-3-ylsulfanylmethyl]-8-methyl- pyrido[l,2-a]pyrimidin-4-one
Figure imgf000073_0001
[0316] This compound was prepared as described in EXAMPLE 44.
[0317] Ή NMR (400MHz, DMSO-i¾: 3 = 12.32 (s, 1 H), 7.98 (d, J = 8.0 Hz, 1 H), 7.48- 7.45 (m, 2H), 7.3 1 (d, 1 H), 7.23 (d, 1 Hd), 7. 17-7.09 (m, 1 H), 7.00 (d, 1 Hd), 6.81 (d, 1 Hd,), 6.38 (s, 1 H), 4.38-4.27 (m, 2H), 3.68 (s, 3H), 2.45 (s, 3H). MS: m/z 462.1 (M+H+)
EXAMPLE 52
2-[4-(2-Methoxy-phenyl)-5-thiophen-2-yl-4H-[ l ,2,4]triazol-3-ylsulfanylmethyl]- l H- quinazoiin-4-one
Figure imgf000073_0002
1 2 3
Scheme 5 [0318] Step l :
[0319] A mixture of 2-amino-benzoic acid ethyl ester (248 mg, 1.50 mmol) and 2- chloroethanenitrile (171 mg, 2.25 mmol) in HCl/dioxane (4 M, 10 mL) was heated at 70 °C for t, 3HLC showed the reaction was completed. The mixture was cooled, quenched with aqueous Na2CC>3 to PH= 6-7. The resulting solid was filtered and dried to afford 280 mg (yield: 95% ) of 2-chloromethyl- l H-quinazolin-4-one precipitated as a white solid.
[0320] Ή NMR (400MHz, DMSO-i/6): δ = 12.74- 12.37 (m, 1 H), 8.26-8.03 (m, 1 H), 7.93- 7.78 (m, 1 H), 7.68 (d, 1 H), 7.55 (t, 1 H), 4.56 (s, 2H).
[0321 ] Step 2:
[0322] The procedure is similar to EXAMPLE 31 . [0323] Ή NMR (400MHz, DMSO-i¾): δ = 8.31 (s, 1H), 7.88 (dd, J= 1.6, 1.2 Hz, 1H), 7.65-7.59 (d,J= 1.2 Hz, 2H), 7.49 (dd,J= 1.6, 1.2 Hz, 1H), 7.38 (dd, J= 7.6, 2.0 Hz, 1H), 7.32 (d, J= 8.4 Hz, 1H), 7.26 (d, J= 3.0 Hz, 1H), 7.15 (t, J= 6.0 Hz, 1H), 7.08 (t, J= 8.0 Hz, 1 H), 7.00 (t, J= 6.8 Hz, 1 H), 6.80 (d, J= 2.4 Hz, 1 H), 4.26 (s, 2H), 3.73 (s, 3H). MS: m/z 448.1 (M+H+).
EXAMPLE 53
2-(((4-(2-methoxyphenyl)-5-(thiophen-2-yl)-4H-l,2,4-triazol-3-yl)thio)methyl)-7- methylquinazolin-4(3H)-one
Figure imgf000074_0001
[0324] 1 H NMR (400MHz, DMSO-<¾): 0 = 12.32 (s, 1 H), 7.98 (d, , J = 8.0 Hz, 1 H), 7.66- 7.62 (m, 2H), 7.44 (d, , J = 1.2 Hz, 1 H), 7.39 (s, 1 H), 7.32 (d, , J- 4.0 Hz, 2H), 7.11 (t, , J = 5.6 Hz, 1H), 7.01 (t, ,J=4.0 Hz, 1H), 6.81 (d, ,J=2.4 Hz, 1H), 4.32 (s, 2H), 3.68 (s, 3H), 2.42 (s, 3H). MS: m/z 462.1 (M+H+).
EXAMPLE 54
2-[4-(2-Methoxy-phenyl)-5-thiophen-2-yl-4H-[l,2,4]triazol-3-ylsulfanylmethyl]-4- dihydro-quinazoline-6-carbonitrile
Figure imgf000074_0002
[0325] This compound was prepared as described in EXAMPLE 52.
[0326] 'H NMR(400 MHZ, DMSO-^): = 12.82 (s, 1H), 8.49 (d, J= 1.6 Hz, 1H), 8.14 (m, 1H), 7.72 (d, J=8.4 Hz, 1H), 7.65-7.61 (m, 2H), 7.46-7.44 (m, 1H), 7.31 (d, J=8.8Hz, 1H), 7.12 (d, J=7.6Hz, 1H), 7.03 (m, 1H), 6.81 (d,J=3.2Hz, 1H), 4.35 (s, 2H), 3.68 (s, 3H). MS: m/z 473.1 (M+H+). EXAMPLE 55
6-Fluoro-2-[4-(2-methoxy-phenyl)-5-thiophen-2-yl-4H-[l,2,4]triazol-3-ylsulfanylmethyl]- 1 H-quinazolin-4-one
Figure imgf000075_0001
[0327] This compound was prepared as described in EXAMPLE 52.
[0328] Ή NMR (300 MHz, DMSO-i/6): δ = 12.57 (s, IH), 7.78 (d, J= 7.8 Hz, IH), 7.70- 7.60 (m, 4H), 7.44 (m, IH), 7.32 (d, J= 8.1 Hz, IH), 7.11 (m, IH), 7.01 (m, IH), 6.81 (d, J = 2.7 Hz, IH), 4.33 (s, 2H), 3.68 (s, 3H). MS: m/z 466.1 (M+H+).
EXAMPLE 56
5-Fluoro-2-[4-(2-methoxy-phenyl)-5-thiophen-2-yl-4H-[l,2,4]triazol-3-ylsuIfanylmethyl]- 1 H-quinazolin-4-one
Figure imgf000075_0002
[0329] This compound was prepared as described in EXAMPLE 52.
[0330] Ή NMR (400 MHz, DMSO-</6): δ = 12.46 (s, IH), 7.79-7.72 (m, IH), 7.66-7.61 (m, 2H),7.45 (m, IH), 7.39(d,J=7.6 Hz, 1H),7.32 (d,J=7.6 Hz, lH),7.25(m, 1H),7.12 (t,J=7.6Hz, IH), 7.01 (m, IH), 6.81 (m, IH), 4.30 (s, 2H), 3.68 (s, 3H). MS: m/z 466.2 (M+H+).
EXAMPLE 57
8-Fluoro-2-[4-(2-methoxy-phenyl)-5-thiophen-2-yl-4H-[l,2,4]triazol-3-ylsulfanylmethyl]- 1 H-quinazolin-4-one
Figure imgf000076_0001
Scheme 6
[0331] Step l :
[0332] A solution of 3-fluoro-2-nitro-benzoic acid (370 mg, 2.0 mmol) in MeOH (10 mL) and C.H2SO4 ( 16 mL) was heated at reflux and stirred for overnight. TLC showed the reaction was completed. The mixture was cooled and evaporated in vacuum to give 3-fluoro-2-nitro- benzoic acid methyl ester (360 mg, yield: 90%) as a yellow oil, which was used for next step without further purificaiton.
[0333] Step 2:
[0334] To a stirred solution of 3-fluoro-2-nitro-benzoic acid methyl ester (350 mg, 1.75 mmol) in MeOH ( 10 mL) was amed Pd/C (35.0 mg, 10%, wet). The suspension was stirred under H2 at room temperature overnight. The reactant was filtered. The filtrate was evaporated in vacuum to afford 2-amino-3-fluoro-benzoic acid methyl ester (300 mg, yield: 00%) as a yellow oil, which was used for next step without further purificaiton.
[0335] Ή NMR (CDCI3, 300 MHz): δ = 7.66 (d, 1 H), 7.12 (m, 1 H), 6.58 (t, 1 H), 5.80 (s, 2H), 3.90 (s, 3H,).
[0336] Steps 3-4:
[0337] The procedure is similar to EXAMPLE 52. [0338] 'H MR(300 MHz, DMSO-i/6): 8= 12.64 (s, IH), 7.91 (d,J=8.1 Hz, IH), 7.72- 7.59 (m, 3H), 7.53-7.46(m, 2H), 7.32 (d, J= 8.1 Hz, IH), 7.11 (m, IH), 7.04-6.98 (m, IH), 6.81 (d,J=0.9 Hz, IH), 4.32 (s,2H), 3.68 (s,3H). MS: m/z 466.1 (M+H+).
EXAMPLE 58
2-[4-(2-Methoxy-phenyl)-5-thiophen-2-yl-4H-[l,2,4]triazol-3-ylsulfanylmethyl]-8-methyl- 1 H-quinazolin-4-one
Figure imgf000077_0001
[0339] This compound was prepared as described in EXAMPLE 52.
[0340] Ή NMR (400 MHz, OMSO-d6): δ = 12.38 (s, IH), 7.93 (d, J= 7.6 Hz, IH), 7.65- 7.60 (m, 3H), 7.40-7.36 (m, 2H), 7.32 (d, J= 7.6 Hz, IH), 7.09 (t, J= 7.6 Hz, IH), 7.00 (m, IH), 6.81-6.79 (m, IH), 4.25 (s, 2H), 3.67 (s, 3H), 2.38 (s, 3H). MS: m/z 462.1 (M+H+).
EXAMPLE 59
2-[4-(2-Methoxy-phenyl)-5-thiophen-2-yl-4H-[l,2,4]triazol-3-ylsulfanylmethyl]-6-methyl- 1 H-quinazolin-4-one
Figure imgf000077_0002
[0341] This compound was prepared as described in EXAMPLE 52.
[0342] Ή NMR (400 MHz, DMSO-i/6): δ = 7.90 (s, IH), 7.65-7.58 (m, 3H), 7.48 (d, J = 8.0 Hz, IH), 7.43 (m, IH), 7.32 (d,J=7.6Hz, IH), 7.13-7.10 (m, IH), 7.03-7.01 (m, IH), 6.83-6.79 (m, IH), 4.32 (s, 2H),3.68 (s, 3H), 2.43 (s, 3H). MS: m/z 462.1 (M+H+).
EXAMPLE 60
2-[4-(2-Methoxy-phenyl)-5-thiophen-2-yl-4H-[l,2,4]triazol-3-ylsulfanylmethyl]-7- trifluoromethyl-1 H-quinazolin-4-one
Figure imgf000078_0001
[0343] This compound was prepared as described in EXAMPLE 52.
[0344] Ή NMR (400 MHz, DMSO-i6): δ = 12.75 (s, IH), 8.32 (d, J= 8.0 Hz, IH), 7.90 (s, IH), 7.81 (d,J=8.0 Hz, IH), 7.66-7.61 (m, 2H), 7.48-7.44 (m, IH), 7.32 (d, J= 8.0 Hz, IH), 7.14-7.10 (m, IH), 7.01 (dd, J= 4.8, 3.6 Hz, IH), 6.80 (dd,J=3.6, 1.2 Hz, IH), 4.36 (s, 2H), 3.68 (s, 3H). MS: m/z 516.1 (M+H+).
EXAMPLE 61
6-Bromo-2-[4-(2-methoxy-phenyl)-5-thiophen-2-yl-4H-[l,2,4]triazol-3-ylsulfanylmethyl]- 1 H-quinazolin-4-one
Figure imgf000078_0002
[0345] This compound was prepared as described in EXAMPLE 52.
[0346] Ή NMR (400 MHz, DMSO-< ): δ = 12.62 (s, IH), 8.18 (d, J = 2.4 Hz, IH), 7.93 (m, IH), 7.66-7.63 (m, 2H), 7.54 (d,J=8.8 Hz, IH), 7.44 (m, IH), 7.32 (d, J= 7.6 Hz, IH), 7.11 (m, IH), 7.01 (m, IH), 6.81 (m, IH), 4.32 (s, 2H), 3.68 (s, 3H). MS: m/z 526.1 (M+H+).
EXAMPLE 62
6-Chloro-2-[4-(2-methoxy-phenyI)-5-thiophen-2-yl-4H-[l,2,4]triazol-3-ylsulfanylmethyI]- 1 H-quinazolin-4-one
Figure imgf000078_0003
[0347] This compound was prepared as described in EXAMPLE 52. [0348] Ή NMR (400 MHz, DMSO-c/e): δ = 12.61 (s, IH), 8.04 (d, J= 2.4 Hz, IH), 7.82 (m, IH), 7.66-7.60 (m, 3H), 7.44 (m, IH), 7.32 (d,J= 8.0 Hz, IH), 7.11 (m, IH), 7.01 (m, IH), 6.81 (m, IH), 4.33 (s, 2H), 3.68 (s, 3H). MS: m/z 482.1 (M+H+).
EXAMPLE 63
7-Fluoro-2-[4-(2-methoxy-phenyl)-5-thiophen-2-yl-4H-[l,2,4]triazol-3-ylsulfanylmethyl]- 1 H-quinazolin-4-one
Figure imgf000079_0001
[0349] This compound was prepared as described in EXAMPLE 56.
[0350] Ή NMR (300 MHz, DMSO- 6): δ = 12.53 (s, IH), 8.18-8.13 (m, IH), 7.66-7.60 (m, 2H), 7.47-7.30 (m, 4H), 7.11 (m, IH), 7.04-6.96 (m, IH), 6.81 (d,J=3.3 Hz, 1H),4.33 (s, 2H), 3.68 (s, 3H). MS: m/z 466.1 (M+H+).
EXAMPLE 64
2-[4-(2-Methoxy-phenyl)-5-thiophen-2-yl-4H-[l,2,4]triazol-3-ylsulfanylmethyl]-6- trifluoromethyl-lH-quinazoIin-4-one
Figure imgf000079_0002
[0351] This compound was prepared as described in EXAMPLE 52.
[0352] Ή NMR (400 MHz, DMSO-i6): δ = 12.80 (s, IH), 8.34 (s, IH), 8.10-8.08 (m, IH), 7.78 (d,J= 8.4 Hz, IH), 7.66-7.61 (m, 2H), 7.46 (dd, d,J=8.0, 1.6 Hz, IH), 7.33 (d, J = 8.4 Hz, IH), 7.15-7.11 (m, IH), 7.01 (dd, J= 6.4, 2.4, IH), 6.83-6.81 (m, IH), 4.37 (s, 2H), 3.68 (s, 3H). MS: m/z 516.1 (M+H+). EXAMPLE 65
6-Methoxy-2-[4-(2-methoxy-phenyl)-5-thiophen-2-yl-4H-[l,2,4]triazol-3-ylsulfanylmethyl]- 1 H-quinazol in-4-one
Figure imgf000080_0001
[0353] This compound was prepared as described in EXAMPLE 52.
[0354] Ή NMR (300 MHz, OMSO-d6): δ = 12.38 (s, 1H), 7.65-7.60 (m, 2H), 7.56-7.30 (m, 5H), 7.13-7.07 (m, 1H), 7.03-6.99 (m, 1H),6.80 (d, d,J=3.3 Hz, 1H), 4.31 (s, 2H), 3.86 (s, 3H), 3.68 (s, 3H). MS: m/z 478.1 (M+H+).
EXAMPLE 66
7-Chloro-2-[4-(2-methoxy-phenyl)-5-thiophen-2-yl-4H-[l,2,4]triazol-3-ylsulfanylmethyl]- 1 H-quinazolin-4-one
Figure imgf000080_0002
[0355] This compound was prepared as described in EXAMPLE 52.
[0356] Ή NMR (400 MHz, DMSO- ): δ = 12.59 (s, 1H), 8.09 (d, J= 8.4 Hz, 1H), 7.66- 7.57 (m,3H), 7.55 (m, 1H), 7.46 (d,J=7.6 Hz, 1H), 7.33 (d,J= 8.8 Hz, 1H), 7.11 (m, 1H), 7.03-6.96 (m, 1H), 6.81 (d, J= 3.6 Hz, 1H), 4.32 (s, 2H), 3.68 (s, 3H). MS: m/z 482.1 (M+H+).
EXAMPLE 67
7-Methoxy-2-[4-(2-methoxy-phenyl)-5-thiophen-2-yl-4H-[l,2,4]triazol-3-ylsuIfanylmethyl]- 1 H-quinazolin-4-one U 2015/032748
80
Figure imgf000081_0001
[0357] This compound was prepared as described in EXAMPLE 52.
[0358] Ή NMR (400 MHz, DMSO-i/6): δ = 12.43 (s, IH), 7.69-7.60 (m, 3H), 7.55 (d, J = 7.2 Hz, IH), 7.43 (t, J = 8.0 Hz, IH), 7.34-7.31 (m, 2H), 7.14-7.11 (m, IH), 7.04-6.98 (m, IH), 6.80 (d, J= 3.6 Hz, IH), 4.35-4.19 (m, 2H), 3.81 (s, 3H), 3.69 (s, 3H). MS: m/z
478.2(M+H+).
EXAMPLE 68
2-[4-(2-Methoxy-phenyl)-5-thiophen-2-yl-4H-[l,2,4]triazol-3-ylsulfanylmethyl]-quinoline
Figure imgf000081_0002
[0359] This compound was prepared as described in EXAMPLE 31.
[0360] Ή NMR (400 MHz, DMSO-c/6): δ = 8.35 (d, J= 8.4 Hz, IH), 7.64 (m, 2H), 7.75- 7.74 (m, IH), 7.64-7.59 (m, 4H), 7.43 (d, d,J=7.6, 1.6 Hz, 1H),7.32 (d,J=7.6 Hz, IH) 7.12 (t, d, J= 7.6 Hz, 1 H), 7.00 (dd, d, J = 4.4, 3.6 Hz, 1 H), 6.80 (dd, J = 3.6, 0.8 Hz, 1 H), 4.68 (s, 2H), 3.67 (s, 3H). MS: m/z 431 ,1(M+H+).
EXAMPLE 69
2-[4-(2-Methoxy-phenyl)-5-thiophen-2-yl-4H-[l,2,4]triazoI-3-ylsulfanylmethyl]-6- trifluoromethoxy-lH-quinazolin-4-one
Figure imgf000082_0001
Scheme 7
[0361 ] Step l :
[0362] To a stirred solution of (4-trifluoromethoxy-phenyl)-carbamic acid tert-butyl ester (410 mg, 1.48 mmol) in anhydrous THF ( 10 mL) was amed droppwise t-BuLi ( 1.9 mL, 2.96 mmol, 2 M in THF) at -78 °C. The mixture was stirred at this temperature for 1 hr. Then COT gas was bubbled into the reaction mixture at -75 °C, the result solution was stirred ovemight before quenched with aqueous NH4CI. The new suspension was extracted with EA (20 mL x2). The extracts were washed brine (30 mL), dried over anhydrous a2S04, filtered. The filtrate was evaporated in vacuum to afford 2-tert-butoxycarbonylamino-5-trifluoromethoxy- benzoic acid (360 mg, yield: 75%) as a yellow solid.
[0363] Ή NMR (400 MHz, DMSO-de): δ = 13.09 (s, 1 H), 8.22 (d, J = 8.0 Hz, 1 H), 7.82 (d, J = 1.6 Hz, 1 H ), 7.26 (dd, J = 8.0, 1.6 Hz, 1 H), 1 .46 (9H, s).
[0364] Step 2:
[0365] The procedure is similar to EXAMPLE 57. [0366] Step 3:
[0367] A solution of 2-tert-butoxycarbonyIamino-5-trifluoromethoxy-benzoic acid methyl ester (350 mg, 1.04 mmol) in HCl/dioxane (10 mL), was stirred at room temperature for 2 hrs. TLC showed the reaction was completed. The reaction mixture was evaporated in vacuum to give 2-amino-5-trifluoromethoxy-benzoic acid methyl ester (220 mg, yield: 75%) as a white solid.
[0368] Ή NMR (400 MHz, DMSO-flfe): 5 = 7.71 (d,J=1.6Hz, 1H), 7.20 (dd, J= 8.0, 1.6 Ηζ,ΙΗ), 6.60 (d, J= 8.0 Hz, 1H), 5.80 (brs, 2H), 3.89 (s, 3H).
[0369] Steps 4-5:
[0370] The procedures are similar to EXAMPLE 52.
[0371] 'HNMR (400 MHz, DMSO-c/6): δ = 12.69 (s, 1H), 7.94 (s, 1H), 7.83-7.69 (m, 2H), 7.65-7.60 (m, 2H), 7.44 (d, J= 6.0 Hz, 1H), 7.32 (d, J= 8.4 Hz, 1H), 7.11 (t, J= 7.6 Hz, 1H), 7.03-6.98 (m, 1H), 6.81 (d, J= 2.8 Hz, 1H), 4.34 (s, 2H), 3.67 (s, 3H). MS: m/z 532.1 (M+H+).
EXAMPLE 70
3-(4-Chloro-benzylsulfanyl)-4-(2-methoxy-phenyl)-5-thiophen-2-yl-4H-[l,2,4]triazole
Figure imgf000083_0001
Scheme 8
[0372] To a stirred solution of l-bromomethyl-4-chloro-benzene (50.0 mg, 0.240 mmol) and 5-(2-methoxy-phenyl)-4-thiophen-2-yl-cyclopenta-l,3-dienethiol (85.0 mg, 0.290 mmol) in acetone (10 mL) was added K2CO3 (100 mg, 0.720 mmol). The the mixture was stirred at 60 °C for 2 hrs. TLC indicated no SM. The mixture was cooled and filtered. The filtrate was evaporated in vacuum to dryness, the residue was diluted with EA (10 mL) and the new suspension was washed with 5% aqueos NaOH (10 mL, 5%) and brine(10 mL). The solution was dried over anhydrous Na2SC and filtered. The filtrate was evaporated in vacuo to dryness. The residue was purified by Pre-TLC (EA/PE = 1/1) to give 3-(4-chloro- benzylsulfanyl)-4-(2-methoxy-phenyl)-5-thiophen-2-yl-4H-[l,2,4]triazole (35.0 mg, yield: 35%) as a yellow solid.
10373] Ή NMR (400 MHz, DMSO-i/6): δ = 7.58-7.51 (m, 1H), 7.46-7.17 (m, 5H), 7.06 (m, 3H), 7.02-6.76 (m, 2H), 4.25 (s, 2H), 3.67 (s, 3H). MS: m/z 414.1 (M+H+). EXAMPLE 71
-Dichloro-benzylsulfanyl)-4-(2-methoxy-phenyl)-5-thiophen-2-yl-4H-[l ,2,4]triazole
Figure imgf000084_0001
[0374] This compound was prepared as described in EXAMPLE 70.
[0375] Ή NMR (400 MHz, DMSO-<¾): δ = 7.64-7.56 (m, 4H), 7.39-7.30 (m, 3H), 7.14 (m, 1 H), 7.00 (m, 1 H), 6.85-6.75 (m, 1 H), 4.34 (s, 2H), 3.67 (s, 3H). MS: m/z 448.0 (M+H+).
EXAMPLE 72
4-(2-Methoxy-phenyl)-3-(4-nitro-benzylsulfanyl)-5-thiophen-2-yl-4H-[ l,2,4]triazoIe
Figure imgf000084_0002
[0376] This compound was prepared as described in EXAMPLE 70.
[0377] Ή NMR (400 MHz, OMSO-d6): δ = 8.18 (d, J = 8.4Hz, 2H), 7.65-7.61 (m, 4H), 7.39 (m, J = 7.7, 1.5 Hz, I H), 7.31 (d, J= 8.4 Hz, I H), 7.17 (dt, J = 15.2, 5.3 Hz, I H), 7.07- 6.94 (m, I H), 6.88-6.73 (m, I H), 4.47 (s, 2H), 3.66 (s, 3H). MS: m/z 425.1 (M+H+).
EXAMPLE 73 uoro-benzylsuIfanyl)-4-( -methoxy-phenyl)-5-thiophen-2-yl-4H-[ l ,2,4]triazole
Figure imgf000084_0003
578] This compound was prepared as described in EXAMPLE 70.
$79] Ή NMR (400 MHz, DMSO-i 6): δ = 7.62 (m, 2H), 7.42-7.25 (m, 3H), 7.25-7.03 , 4H), 7.02-6.98 (m, I H), 6.80 (m, I H), 4.34 (s, 2H), 3.67 (s, 3H). MS: m/z 398.1 (M+H+). EXAMPLE 74
3-(2-Fluoro-benzylsulfanyl)-4-(2-methoxy-phenyl)-5-thiophen-2-yl-4H-[l ,2,4]triazole
Figure imgf000085_0001
[0380] This compound was prepared as described in EXAMPLE 70.
[0381] Ή NMR (400 MHz, DMSO-ifc): δ = 7.66-7.59 (m, 2H), 7.45-7.26 (m, 4H)), 7.21 - 7.08 (m, 3H), 7.04-6.96 (m, I H), 6.80 (dd, J = 3.6, 0.9 Hz, IH), 4.35 (s, 2H), 3.67 (s, 3H). MS: m/z 398.1 (M+H+).
EXAMPLE 75
4-(2-Methoxy-phenyl)-3-(4-met n-2-yl-4H-[l,2,4]triazole
Figure imgf000085_0002
[0382] This compound was prepared as described in EXAMPLE 70.
[0383] Ή NMR (400 MHz, DMSC s): δ = 7.63-7.61 (m, 2H), 7.31 (d, J = 7.6 Hz, 2H), 7.20 (d, J = 8.4 Hz, 2H), 7.12-7.09 (ra, 3H), 7.00 (m, I H), 6.80 (m, I H), 4.30 (s, 2H), 3.67 (s, 3H) , 2.63 (s, 3H). MS: m/z 394.1 (M+H+).
EXAMPLE 76
4-(2-Methoxy-phenyl)-3-(3-methyl-benzylsulfanyl)-5-thiophen-2-yl-4H-[ l ,2,4]triazole
Figure imgf000085_0003
[0384] This compound was prepared as described in EXAMPLE 70. [0385] 1 H NMR (400 MHz, DMSO-d6): δ = 7.64-7.61 (m, 2H), 7.32-7.25 (m, 2H), 7.19- 7.08 (m, 2H), 7.02-6.97 (m, 1 H), 6.79 (dd, J = 3.6, 0.9 Hz, 1 H), 4.30 (s, 2H), 3.68 (s, 3H), 2.25 (s, 3H). MS: m/z 394.1 (M+H+).
EXAMPLE 77
3-(3,5-Dif uoro-benzylsulfanyl)-4-(2-methoxy-phenyl)-5-thiophen-2-yl-4H-[ l ,2,4]triazole
Figure imgf000086_0001
[0386] This compound was prepared as described in EXAMPLE 69.
[0387] ]H NMR (400 MHz, DMSO-c/6): δ = 7.64-7.62 (m, 2H), 7.42-7.38 (m, 1 H), 7.32 (d, J = 8.0 Hz, 1 H), 7.17-7.07 (m, 4H), 7.02-6.98 (m, 1 H), 6.81 (dd, J = 4.0, 1 .2 Hz, 1 H), 4.36(d, J = 5.2 Hz, 2H), 3.67 (s, 3H). MS: m/z 416.1 (M+H+).
EXAMPLE 78
4-[4-(2-Methoxy-phenyl)-5-thiophen-2-yl-4H-[l ,2,4]triazol-3-ylsulfanylmethyl]-pyrimidine
Figure imgf000086_0002
Scheme 9
[0388] Step l :
[0389] The mixture of 4-methyl-pyrimidine (564 mg, 6.0 mmol) and 1 ,3,5-trichloro- [ l ,3,5]triazinane-2,4,6-trione (557 mg, 24.0 mmol) in CH3C1 (10 mL) was heated at reflux and stirred overnight. The mixture was filtered and the filtrate was evaporated in cacuitm. the residue purified by Pre-TLC (EA/PE = 1 : 1 ) to afford 4-chloromethyl-pyrimidine (400 mg, yield: 52%) as yellow oil. [0390] 1 H N R (DMSO-i 6, 400 MHz): δ = 9.28-9.1 1 (m, 1 H), 8.92-8.80 (m, 1 H), 7.74- 7.62 (m, 1 H), 4.80 (s, 2H).
[0391 J Step2:
[0392] To a mixture of 4-(2-methoxy-phenyl)-5-thiophen-2-yl-4H-[ 1 ,2,4]triazoIe-3-thiol (85.0 mg, 0.290 mmol) and 4-chloromethyI-pyrimidine (3 1 .0 mg, 0.240 mmo!) in acetone ( 10 mL) was added K2CO3 ( 100 mg, 0.720 mmol). The mixture was heated at 60 °C and stirred for 2 hrs. The mixture was filtered and the filtrate was evaporated in vacuum. The residue was dissolved in EA ( 10 mL). The new suspension was washed with 5% NaOH solution and brine, and then dried over Na2S04. The solution was evaporated in vacuum. The residue was purified by Pre-TLC (EA/PE = 1 /1 ) to afford 4-[4-(2-Methoxy-phenyl)-5-thiophen-2-yl-4H- [ l ,2,4]triazol-3-ylsulfanylmethyl]-pyrimidine (35 mg, yield: 38%) as yellow solid.
[0393] Ή NMR (400 MHz, DMSO-cfc): 6 = 9.10 (s, 1 H), 8.88-8.69 (m, 1 H), 7.67-7.57 (m, 3 H), 7.45 (d, J = 7.6 Hz, 1 H), 7.33 (d, J = 8.4 Hz, 1 H), 7.16 (t, J= 7.6 Hz, 1 H), 7.01 (t, J = 3.6 Hz, 1 H), 6.81 (d, J = 2.4 Hz, 1 H), 4.47 (s, 2H), 3.69 (s, 3H). MS: m/z 382.1 (M+H+).
EXAMPLE 79
2-[ l -(2-Methoxy-phenyl)-5-thiophen-2-yl-l H-imidazol-2-ylsulfanylmethyl]-7-methyl- pyrido[ l ,2-a]pyrimidin-4-one
Figure imgf000087_0001
Scheme 10 [0394] Step 1 :
[0395] A mixture of 2-bromo- l -(thiophen-2-yl)ethanone (410 mg, 2.0 mmol) and sodium diformylamide (210 mg, 2.2 mmol) in MeCN (10 mL) was heated at reflux for 4 hrs. After cooling to room temperature, the reactant was filtered and the filtrate was concentrated to dryness in vacuum. The residue was dissolved in the mixture solvent of EtOH (10 mL) and c. HCI ( 1 mL) and the mixture was stirred at room temperature overnight. The reactant was concentrated in vaccum to give 2-amino-l-(thiophen-2-yI)ethanone (HC1 salt, 140 mg, yield: 49%) as tan oil, which was used for next step without further purification.
[0396] 'HNMR (400 MHz, DMSO-i/6): δ = 8.50 (brs, 3H), 8.16 (d, J= 4.8 Hz, 1H), 8.13 (d, J=4.0 Hz, 1H), 7.33 (t,J=4.4 Hz, 1H), 4.51 (q,J=5.6Hz, 2H).
[0397] Step 2:
[0398] To a solution of 2-amino-l-(thiophen-2-yl)ethanone HC1 salt (210 mg, 1.2 mmol) and l-isothiocyanato-2-methoxybenzene (245 mg, 1.2 mmol) in 5 mL of water was added 2CO3 (500 mg, 3.6 mmol) and the mixture was heated at 50 °C for hours. After the reaction was completed showed by LCMS, the reaction solution was neutralized with IN HC1 to pH = 6-7. The aqueous phase was extracted with EA (10 mL x3) and the extracts were washed with brine (10 mL) and dried over Na2SC>4. The solution was filtered and concentrated to dryness in vacuum. The residue was purified by prep-TLC (EA/PE = 1/1) to give l-(2- methoxyphenyl)-5-(thiophen-2-yl)-lH-imidazole-2-thiol (140 mg, yield: 41%) as a yellow solid.
[0399] Step 4:
[0400] To a solution of l-(2-methoxyphenyl)-5-(thiophen-2-yl)-lH-imidazole-2-thiol (50.0 mg, 0.17 mmol) in acetone (2 mL) was added 2-(chloromethyI)-8-methyl-4H- pyrido[l,2-a]pyrimidin-4-one (40 mg, 0.19 mmol) and 2CO3 (71 mg, 0.51 mmol). Then the mixture was warmed to 60 °C and stirred until the reaction was completed (~2hrs). Aftet cooling to room temperature, the reactant was filtered and the filtrate was concentrated to dryness in vacuum. The residue was diluted with EA (5 mL) and the suspension was washed with aqueous 5% NaOH (5 mL), brine (5 mL). The organic layer was dried over anhydrous Na2S04, filtered and concentrated. The residue was purified by Prep-TLC (PE/EA = 1/1) to give 2-[l-(2-methoxy-phenyl)-5-thiophen-2-yl-lH-imidazol-2-ylsulfanylmethyl]-7-methyl- pyrido[l,2-a]pyrimidin-4-one (20 mg, yield: 25%) as a yellow solid.
[0401] 'HNMR (300 MHz, DMSO-i¾: 5 = 8.77 (s, 1H), 7.85 (dd, J= 9.0 Hz, 1.8 Hz, 1H), 7.61-7.48 (m, 2H), 7.39 (s, 1H), 7.36 (dd, J= 8.0 Hz, 1.1 Hz, 1H), 7.27-7.16 (m, 2H), 7.00 (t,J=7.5 Hz, 1H), 6.92 (dd,J=5.0 Hz, 3.8 Hz, 1H), 6.80 (dd,J= 5.0 Hz, 1.1 Hz, 1H), 6.34 (s, 1H), 4.21 (s, 2H), 3.66 (s, 3H), 2.39 (s, 3H). MS: m/z 461.1 ( M+H+). EXAMPLE 80
4-(2-Methoxy-phenyl)-3-(2-phenyl-oxazol-4-ylmethoxy)-5-thiophen-2-yl-4H-[l ,2,4]triazole
Figure imgf000089_0001
Scheme 1 1
[0402] Step l :
[0403] A suspension of l -(2-methoxyphenyl)-5-(thiophen-2-yl)- l H-imidazole-2 -thiol (0.7 g, 2.43 mmol), Mel (0.52 g, 3.64 mmol) and 2C03 ( 1 .0 g, 7.29 mmol) in THF (20 mL) was stirred at room temperature overnight. The reaction solution was evaporated in vacuum. The residue was dissolved in DCM ( 10 mL) and filtered. The filtrate was concentrated to give 1 - (2-methoxyphenyl)-2-(methylthio)-5-(thiophen-2-yl)- l H-imidazole (0.72 g, yield: 92%) as yellow oil.
[0404] Step 2:
[0405] A solution of l -(2-methoxyphenyl)-2-(methylthio)-5-(thiophen-2-yl)-l H-imidazole (0.72 g, 2.38 mmol) and m-CPBA ( 1.03 mg, 5.95 mmol) in DCM (20 mL) was stirred at room temperature overnight. The reaction solution was concentrated to diyness and the residue was purified by silica gel column (EA/PE = 1/5) to give l -(2-methoxyphenyl)-2- (methylsulfonyl)-5-(thiophen-2-yl)- l H-imidazole (0.39 g, yield: 48%) as a tan solid.
[0406] Step 3 :
[0407] A mixture of l -(2-methoxyphenyl)-2-(methylsulfonyl)-5-(thiophen-2-yl)- l H- imidazole (34 mg, 0.10 mmol), (2-phenylthiazol-4-yl)methanol (20 mg, 0.10 mmol) and KOBu' (35 mg, 0.3 1 mmol) in DMF (3 mL) was heated at 120 °C under MW irridation for 0.5 h. The suspension was concentrated to dryness and purified by prep-TLC (EA/PE = 1 /5) to give 4-(2-Methoxy-phenyl)-3-(2-phenyl-oxazol-4-ylmethoxy)-5-thiophen-2-yl-4H- [ l ,2,4]triazole (5.5 mg, yield: 12%) as yellow oil.
[0408] 1 HNMR (300 MHz, CDC13): δ = 7.95-7.91 (m, 2H), 7.47-7.41 (m, 4H), 7.27-7.22 (m, 2H), 7.09-6.99 (m, 4H), 6.86-6.82 (m, 1 H), 6.62 (dd, J= 3.6 Hz, 1 .2 Hz, 1 H), 5.61 (dd, J = 8.1 Hz, 0.6 Hz, 1 H), 3.68 (s, 3H). MS: m/z 446.2 (M+H+).
EXAMPLE 81
4-(2-Methoxy-phenyl)-3-(2-phenyl-oxazol-4-ylmethoxy)-5-thiophen-2-yl-4H-[ l ,2,4]triazole
Figure imgf000090_0001
[0409] This compound was prepared as described in EXAMPLE 69.
[0410] ' HNMR (400 MHz, CDCI3): δ = 8.02-7.99 (m, 2H), 7.88 (s, 1 H), 7.49-7.44 (m, 4H), 7.23 (d, J = 1.6 Hz, 2H), 7.05-7.01 (m, 2H), 6.90-6.86 (m, 2H), 5.50 (s, 2H), 3.63 (s, 3H). MS: m/z 43 1 .1 ( M+H+).
EXAMPLE 82
3-(benzyloxy)-4-(2-methoxyphenyl)-5-(thiophen-2-yl)-4H- l ,2,4-triazole
Figure imgf000090_0002
Scheme 12
[0411] A suspension of benzyl alcohol (44 mg, 0.4 mmol) and NaH (60% in mineral oil, 16 mg, 0.4 mmol) in anhydrous THF (2 mL) was stirred at room temperature for 20 mins. Then l -(2-methoxyphenyl)-2-(methylsulfonyI)-5-(thiophen-2-yl)-l ,2,4-triazole (67 mg, 0.2 mmol) was added and the mixture was stirred at room temperature for 12 hrs. The reaction was quenched with water ( 10 mL) and the mixture was extracted with EA ( 10 mL x3). The extracts were dried over Na^SC^ and the solution was concentrated to dryness. The residue was purified by prep-TLC (PE/EA = 1/1) to give 3-(benzyloxy)-4-(2-methoxyphenyl)-5- (thiophen-2-yl)-4H-l,2,4-triazole (43 mg, yield: 59%) as a white solid.
[0412] 'HNMR (400 MHz, DMSO-c/6): δ = 7.59-7.55 (m, 2H), 7.47 (dd, J= 7.8 Hz, 1.4 Hz, IH), 7.38-7.32 (m, 5H), 7.27 (d, J= 8.8 Hz, 2H), 7.11 (d, J= 7.6 Hz, IH), 6.98-6.96 (m, IH), 6.72 (d,J= 3.2 Hz, IH), 5.49-5.41 (m, 2H), 3.68 (s, 3H). MS: m/z 364.1 ( M+H+).
EXAMPLE 83
2-(((4-(2-methoxyphenyl)-5-(thiophen-2-yl)-4H-l,2,4-triazol-3-yl)oxy)methyl)-7-methyl-4H- pyrido[l,2-a]pyrimidin-4-one
Figure imgf000091_0001
[0413] This compound was prepared as described in EXAMPLE 82.
[0414] 'HNMR (400 MHz, CDC13): δ = 8.82 (s, IH), 7.62-7.52 (m, 3H), 7.35 (d, J= 7.6 Hz, IH), 7.27-7.25 (m, IH), 7.15-7.10 (m, 2H), 6.94-6.88 (m, 2H), 6.40 (s, IH), 5.61 (d,J = 14.8 Hz, IH), 5.44 (d, J= 15.6 Hz, IH), 3.81 (s, 3H), 2.43 (s, 3H). MS: m/z 446.2 ( M+H+).
EXAMPLE 84
2-[l-(2-Methoxy-phenyl)-5-thiophen-2-yl-lH-imidazol-2-yisulfanylmethyl]-7-methyl- pyrido[ 1 ,2-a]pyrimidin-4-one
Figure imgf000091_0002
[0415] This compound was prepared as described in EXAMPLE 82.
[0416] 'HNMR (400 MHz, CDC13): δ = 7.93-7.90 (m, 2H), 7.52-7.40 (m, 5H), 7.27-7.23 (m, 2H), 7.08-7.00 (m, 2H), 6.90-6.86 (m, 2H), 5.71-5.63 (m, 2H), 3.64 (s, 3H). MS: m/z 447.1 (M+H+). EXAMPLE 85
4-(2-Methoxy-phenyl)-3-(2-phenyl-oxazol-4-yImethoxy)-5-thiophen-2-yl-4H-[ l ,2,4]triazoIe
Figure imgf000092_0001
Scheme 13
[0417] Step l :
[0418] A mixture of thiophene-2-carboxylic acid (5.12 g, 40 mmol) and BOP (21.2 g, 48 mmol) and TEA (12 g, 0.12 mol) was stirred for 10 mins and 2-methoxyaniline (5.41 g, 44 mmol) was added. The reaction was stirred at room temperature overnight. The reactant was evaported in vacuum and the residue was dissolved in EA ( 100 mL). The mixture was washed with aqueous 1 0% NaOH and brine, and then dried over Na2S04. The solution was filtered and the filtrate was evaporated in vacuum to give N-(2-methoxyphenyl)thiophene-2- carboxamide (9.0 g, yield: 96%) as tan oil.
[0419] Step 2:
[0420] A mixture of N-(2-methoxyphenyl)thiophene-2-carboxamide (9.0 g, 38.6 mmol) and Lawesson's reagent ( 18.7 g, 46.3 mmol) in toluene (50 mL) was heated to reflux overnight. The reactant was evaporated in vacuum and the residue was purified by silica gel column (PE/EA = 8/1 ) to give N-(2-methoxyphenyl)thiophene-2-carbothioamide (8.6 g, yield: 89.4%) as a tan oil.
[0421 ] Step 3:
[0422] To a solution of N-(2-methoxyphenyl)thiophene-2-carbothioamide ( 1 .3 g, 5.2 mmol) in dioxane (60 mL) was added hydrazine monohydrate (2.6 g, 52 mmol), and the mixture was stirred at 50 °C ovemight. The reaction was quenced with water (20 mL) and the aqueous phase was extracted with EA (50 mL x2). The extracts were washed with brine (20 mL) and dried over Na2S04. The solution was filtered and the filtrate was evaporated in vaccum to give (E)-N-(2-methoxyphenyl)thiophene-2-carbohydrazonamide (1 .3 g, yield: 92%) as yellow oil.
[0423] 'HNMR (300 MHz, DMSO-afe): 8 = 7.34 (d, J = 5.1 Hz, 1 H), 6.94 (dd, J = 7.5, 1 .5 Hz, 1 H), 6.91 -6.87 (m, 1 H), 6.80-6.70 (m, 3H), 6.26 (dd, J = 7.2, 1 .8 Hz, 1 H), 6.18 (brs, 2H), 3.84 (s, 3H).
[0424] Step 4:
[0425] To a solution of (E)-N-(2-methoxyphenyl)thiophene-2-carbohydrazonamide ( 1.1 g, 4.5 mmol), cyano bromide (561 mg, 5.3mmol) in dioxane (80 mL) was added K2CO3 (1.86 g, 1.35 mmol) in water (8 mL), and the mixture was heated at 55 °C and stired at this temperature overnight. The reaction solution was concentrated in vacuum and the residue was purified by silica gel column (PE/EA = 4/1 ) to give 4-(2-methoxyphenyl)-5-(thiophen-2-yl)- 4H- l ,2,4-triazoi-3-amine (500 mg, not pure) as a yellow solid.
[0426] A solution of 4-(2-methoxyphenyl)-5-(thiophen-2-yl)-4H-l ,2,4-triazol-3-amine ( 1 .5 g, not pure), Boc 0 ( 1 .73 g, 8.3 mmol), DMAP (67 mg, 0.55 mmol) and TEA (0.5 mL) in THF (20 mL) was stirred at room temperature overnight. The reaction was quenched with water ( 10 mL) and the mixture was extracted with EA (25 mL x3). The extracts were washed with brine (10 mL) and dried over a2S04. The solution was filtered and the filtrate was concentrated to dryness in vacuum. The residue was purified by prep-TLC (PE/EA = 3/1 ) to give 4-(2-methoxyphenyl)-5-(thiophen-2-yl)-4H- l ,2,4-triazol-3-amine with Boc protected ( 1.0 g, yield: 53%) as a yellow oil.
[0427] A solution of the above Boc-protected amine in HCl/dioxane ( 10 mL ) was heated at 50 °C amd stirred for 2 hrs. The reactant was concentrated to dryness in vacuum. The residue was washed with EA and filtered to give 4-(2-methoxyphenyl)-5-(thiophen-2-yl)-4H- l ,2,4-triazol-3-amine HC1 salt (0.5 g, yield: 68%) as a yellow solid.
[0428] Step 5:
[0429] A solution of 2-(chloi methyl)-7-methyl-4H-pyrido[ l ,2-a]pyrimidin-4-one (35 mg, 0.16 mmol), 4-(2-methoxyphenyl)-5-(thiophen-2-yI)-4H- l ,2,4-triazol-3-amine (30 mg, 0.1 1 mmol) and K2C03 (46 mg, 0.33 mmol) in DMF (2 mL) was stiired at 170 °C under MW irradiation for 90 mins. The reaction solution was evaporated in vacuum and the residue was purified by prep-HPLC to give 4-(2-Methoxy-phenyl)-3-(2-phenyl-oxazol-4-ylmethoxy)-5- thiophen-2-yl-4H-[l,2,4]triazole (7.5 mg, yield: 15%) as a yellow solid.
[0430] 'HNMR (400 MHz, DMSO-d6): δ = 8.85-8.78 (m, 2H), 7.90-7.78 (m, 4H), 7.65 (d, J=8.8Hz, 1H), 7.44 (d, J=8.8Hz, 1H), 7.28 (t,J=7.6Hz, 1H), 7.10-7.07 (m, 1H), 6.96 (dd, j=4.0 Hz, 1.2 Hz, 1H), 6.38 (s, 1H), 5.49 (s, 2H), 3.84 (s, 3H), 2.43 (s, 3H). MS: m/z 445.2 (M+H+).
EXAMPLE 86
[4-(2-Methoxy-phenyI)-5-thiophen-2-yl-4H-[l,2,4]triazol-3-yl]-(2-p-tolyl-thiazol-4- ylmethyl)-amine
Figure imgf000094_0001
[0431] This compound was prepared as described in EXAMPLE 85.
[0432] 'HNMR (400 MHz, CD3OD): δ = 7.76 (d, J= 8.0 Hz, 2H), 7.69-7.62 (m, 1H), 7.54 (s, 1H), 7.51-7.48 (m, 2H), 7.26 (d, J= 8.0 Hz, 1H), 7.21-7.12 (m, 3H), 6.93-6.87 (m, 2H), 5.42 (s, 2H), 3.71 (s, 3H), 2.29 (s, 3H). MS: m/z 460.2 (M+H+).
EXAMPLE 87
[4-(2-Methoxy-phenyl)-5-thiophen-2-yl-4H-[l,2,4]triazol-3-yI]-(2-p-tolyl-oxazol-4- ylmethyl)-amine
Figure imgf000094_0002
[0433] This compound was prepared as described in EXAMPLE 85.
[0434] 'HNMR (400 MHz, DMSO-i/6): δ = 8.11 (s, 1 H), 7.88 (d, J= 8.8 Hz, 2H), 7.59- 7.56 (m, 2H), 7.46 (dd, J= 7.8 Hz, 1.4 Hz, 1H), 7.35 (d, J= 8.0 Hz, 2H), 7.27 (d, J= 8.0 Hz, 1 H),7.17-7.14 (m, 1 H), 6.97-6.93 (m, 1 H), 6.68 (dd, J = 3.6, 1 .2 Hz, 1 H), 5.00-4.99 (m, 2H), 3.69 (s, 3H), 2.37 (s, 3H). MS: m/z 444.2 (M+H+).
EXAMPLE 88
BIOLOGICAL DATA
[0435] This example illustrates the biological activity of various compounds tested against the APJ receptor.
[0436] Commercially available cell lines were used to screen chemical libraries to identify compounds that potently and selectively activated the human apelin receptor. The assays disclosed herein were performed using cell lines from DiscoveRx, Inc.
[0437] Biological activity was determined in a cell-based assay. This assay detects activation of the receptor by reporting on intracellular cyclic adenosine monophosphate (cAMP) level. Chinese hamster ovary cells engineered to express a stably integrated human apelin receptor (APJ, AGTRL-1 , APLNR), were plated into the wells of a microtiter plate. Cells were stimulated with forskolin to increase intracellular cAMP. Immediately following, compounds were added at a range of final concentrations 0.0-100 μΜ, in DMSO (not to exceed a final DMSO concentration of > 1 % v/v), and allowed to incubate with the cells for 0.5h. At the end of the incubation, cells were lysed to stop APJ signaling, and cAMP was quantified using a commercially available assay kit that applies enzyme fragment
complementation technology and bioluminescence, in a competitive ELISA format. The potency and efficacy of compounds at APJ are reported in reference to the canonical APJ receptor agonist apelin- 13 tested at 1 nM.
[0438] Compound potency is reported as the effective concentration required to effect 50% (EC50) of the control (apelin- 13, 1 nM) response in micromolar (μΜ) units. Compound efficacy (% resp) is reported as a percentage of the maximal apelin- 13 response at I nM (control). Potencies were calculated by fitting a point logistic curve to a concentration response curve consisting of at least 10 data points spanning the effective range of the compounds in a cell-based assay that measures cAMP. [0439] Table 1.
APJ cAMP
Example Number
ECso, μΜ / % resp
1 1.45/95
2 0.31 / 154
3 6.16/136
4 3.09 / 80
5 1.51 / 126
6 13.6/176
7 6.63/51
8 0.551 / 137
9 0.455 / 136
10 0.0182/ 150
11 0.0704/118
12 1.49 / 89
13 1.42/94
14 0.772/105
15 0.00444 / 52
16 0.0968 / 84
17 0.09/109
18 1.7/58
19 >10/57
20 5.0/10
21 2.37/113
22 1.24/134
23 0.179/109
24 >10
25 >10 / 52
26 >10/86
27 0.0174/82
28 >10/91
29 0.252/62
30 >10/31
31 0.635 / 133
32 1.46/33
33 1.17/80
34 >10/ 120
35 >10 / 90
36 >10/83
37 3.09/138
38 >I0 / 18
39 0.105/93 APJ cAMP
Example Number
EC50, μΜ / % resp
40 3.34/58
41 2.49/58
42 >10/33
43 3.89/102
44 0.0098/ 119
45 0.0705 / 130
46 0.0078/117
47 0.0252/54
48 0.028/103
49 0.0234/107
50 0.00674/ 110
51 0.451 / 125
52 0.24/113
53 1.03/59
54 0.0313/118
55 0.009/ 109
56 8.62/107
57 0.140/106
58 0.547/ 134
59 0.042/ 121
60 0.569/ 160
61 0.0315/80
62 0.0536/ 116
63 0.125/108
64 0.365 /92
65 0.304/ 101
66 0.482/87
67 0.59/59
68 0.211/79
69 1.37/59
70 2.03/134
71 1.22/99
72 0.14/ 106
73 10.2/166
74 15.0/80
75 2.74/70
76 11.3/ 159
77 8.44/111
78 >10/50
79 1.92/54
80 >10/31 APJ cAMP
Example Number
ECso, μΜ / % resp
81 0.238 / 101
82 > 10/ 52
83 0.045 / 52
84 0.680 / 109
85 > 10
86 >10
87 >10
[0440] Although the invention has been described with reference to the above examples, it will be understood that modifications and variations are encompassed within the spirit and scope of the invention. Accordingly, the invention is limited only by the following claims.

Claims

What is claimed is:
1 . A compound of structural Formula I, or a pharmaceutically acceptable salt, polymorph, solvate, tautomer, or N-o
Figure imgf000099_0001
wherein:
A is selected from the group consisting of optionally substituted aryl, optionally substituted heteroaryl, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted alkoxy, optionally substituted cycloalkyl, hydrogen, hydroxyl, -CN and halogen;
B is selected from the group consisting of optionally substituted aryl, optionally substituted heteroaryl, optionally substituted arylalkyl, optionally substituted heteroarylalkyl, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted cycloalkyl, hydrogen, hydroxyl, -CN and halogen;
C is selected from the group consisting of optionally substituted aryl, optionally substituted heteroaryl, optionally substituted arylalkyl, optionally substituted heteroarylalkyl, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted cycloalkyl, hydrogen, hydroxyl, -CN and halogen;
Y is selected from null, a bond, O, S, SO, S02, NH, -C(0)-N(R' )-, -C(0)-0-, -N(R2)- and - C(R3)(R4)-;
X and X" are each independently selected from CR and N;
R1 , R2, R3 and R4 are each independently selected from the group consisting of of optionally substituted aryl, optionally substituted 5- or 6-membered heteroaryl, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted alkoxy, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, hydrogen and hydroxyl; R5 is independently selected from the group consisting of hydrogen, halogen, -CN, -C(O)- N(R6)-R7, -C(0)-0-R8, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted alkoxy, haloalkyl, haloalkoxy, optionally substituted aryl and optionally substituted 5- or 6- membered heteroaryl; and
R6, R7 and Rs are each independently selected from the group consisting of of optionally substituted aryl, optionally substituted 5- or 6-membered heteroaryl, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted alkoxy, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, hydrogen and hydroxy 1.
2. The compound of claim 1, wherein A is optionally substituted aryl or optionally substituted heteroaryl.
3. The compound of claim 1 , wherein A is optionally substituted phenyl or optionally substituted 5- or 6-membered heteroaryl.
4. The compound of claim 3, wherein the optional substituents are selected from the group consisting of hydrogen, halogen, -CN, optionally substituted carboxyl, amido, nitro, amino, optionally substituted alkyl, optionally substituted alkoxy and optionally substituted haloalkoxy and optionally substituted haloalkyl.
5. The compound of claim 1 , wherein A is selected from the group consisting of
Figure imgf000101_0001
6. The compound of claim 1 , wherein Y is selected from the group consisting of S, SO, S02, O and N.
7. The compound of claim 5, wherein Y is selected from the group consisting of S, O and N.
8. The compound of claim 1 , wherein Y is S.
9. The compound of claim 1 , wherein C is optionally substituted alkyl, optionally substituted arylalkyl or optionally substituted heteroarylalkyl.
10. The compound of claim 1 , wherein C is substituted alkyl.
1 1 . The compound of claim 10, wherein the substituent on C is selected from optionally substituted aryl or optionally substituted heteroaryl.
12. The compound of claim 10, wherein the substituent on C is bicyclic heteroaryl.
13. The compound of claim 1 , wherein B is optionally substituted aryl.
14. The compound of claim 13, wherein B is optionally substituted phenyl.
15. The compound of claim 1 , wherein X1 and X2 are each independently N.
16. The compound of claim 1 , wherein the compound has structural Formula II, or a pharmaceutically acceptable salt, polymorph, solvate, tautomer, or N-oxide thereof:
Figure imgf000102_0001
17. The compound of claim 16, wherein C is optionally substituted heteroarylalkyl.
1 8. The compound of claim 16, wherein C is optionally substituted bicyclic
heteroarylalkyl.
19. The compound of claim 17, wherein the optional substituents are selected from the group consisting of hydrogen, halogen, -CN, optionally substituted carboxyl, amido, nitro, amino, optionally substituted alkyl, optionally substituted alkoxy and optionally substituted haloalkoxy and optionally substituted haloalkyl.
20. The compound of claim 16, wherein B is optionally substituted aryl or optionally substituted heteroaryl.
21. The compound of claim 16, wherein B is optionally substituted phenyl or optionally substituted pyridinyl.
22. The compound of claim 21 , wherein the optional substituents are selected from the group consisting of hydrogen, halogen, -CN, optionally substituted carboxyl, amido, nitro, amino, optionally substituted alkyl, optionally substituted alkoxy and optionally substituted haloalkoxy and optionally substituted haloalkyl.
23. The compound of claim 16, wherein C is selected from the group consisting of:
Figure imgf000103_0001
Figure imgf000103_0002
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000105_0002
Figure imgf000105_0003
24. The compound of claim 16, wherein B is selected from the group consisting of:
Figure imgf000106_0001
25. The compound of claim 16, wherein B is substituted phenyl.
26. The compoundof claim 25, wherein the phenyl is substituted with at least one alkoxy.
27. The compound of claim 26, wherein the at least one alkoxy is methoxy.
28. The compound of claim 27, wherein B is
Figure imgf000106_0002
Figure imgf000107_0001
Figure imgf000107_0002
107
Figure imgf000108_0001
Figure imgf000108_0002
Figure imgf000109_0001
Figure imgf000109_0002
Figure imgf000109_0003
30. The compound of claim 1 , or a pharmaceutically acceptable salt, polymorph, solvate, tautomer, or N-oxide thereof, wherein the compound has structural Formula III:
Figure imgf000109_0004
(III).
31. The compound of claim 30, wherein C is optionally substituted alkyl, optionally substituted, heteroarylalkyl or optionally substituted arylalkyl.
32. The compound of claim 31 , wherein C is substituted alkyl and the substituent is selected from optionally substituted aryl or optionally substituted heteroaryl.
33. The compound of claim 32, wherein C is substituted alkyl and the substituent is optionally substituted monocyclic heteroaryl or optionally substituted bicyclic heteroaryl.
34. The compound of claim 32, wherein C is substituted alkyl and the substituent is optionally substituted phenyl.
35. The compound of claim 30, wherein C is selected from:
Figure imgf000110_0001
36. The compound of claim 30, wherein C is
Figure imgf000110_0002
wherein X3 is S or O; and R9 is optionally substituted aryl, wherein the optional substituents are selected from the group consisting of hydrogen, halogen, -CN, optionally substituted carboxyl, amido, nitro, amino, optionally substituted alkyl, optionally substituted alkoxy and optionally substituted haloalkoxy and optionally substituted haloalkyl.
37. The com ound of claim 36, wherein X3 is S.
The compound of claim 37, wherein R9 is selected from
Figure imgf000111_0001
The com ound of claim 36, wherein XJ is O.
The compound of claim 39, wherein R9 is selected from
Figure imgf000111_0002
41 . The compound of claim 1 , wherein the compound has structural Formula IV, or a pharmaceutically acceptable salt, polymorph, solvate, tautomer, or N-oxide thereof:
Figure imgf000112_0001
(IV).
42 The compound of claim 41 , wherein Y is S, O or NH.
43 The compound of claim 41 , wherein Y is S and X2 is CH.
44 The compound of claim 41 , wherein Y is O and X2 is N.
45 The compound of claim 41 , wherein Y is NH and X2 is N.
46. A pharmaceutical composition comprising a compound of structural Formula I, or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable excipient.
47. A method of preventing or treating in a subject an apelin-mediated disease or disorder, comprising administering to the subject a therapeutically effective amount of a compound of structural Formula I, thereby preventing or treating the disease or disorder.
48. The method of claim 47, wherein the disease or disorder is a cardiovascular disease or disorder, coronary heart disease, stroke, heart failure, systolic heart failure, diastolic heart failure, diabetic heart failure, heart failure with preserved ejection fraction, cardiomyopathy, myocardial infarction, left ventricular dysfunction, left ventricular dysfunction after myocardial infarction, cardiac hypertrophy, myocardial remodeling, myocardial remodeling after infarction, myocardial remodeling after cardiac surgery or valvular heart disease.
49. The method of claim 48, wherein the disease or disorder is a metabolic disease or disorder, metabolic syndrome, insulin resistance, diabetes mellitus, diabetic late
complications, diabetic macro- and micro- vasculopathies, diabetic nephropathy, diabetic retinopathy, diabetic neuropathies or cardiac autonomic neuropathy.
50. The method of claim 49, wherein the disease or disorder is caused by CNS- dependent or CNS-independent disturbed fluid homeostasis, acute or chronic renal failure, hypertension, pulmonary hypertension, portal hypertension or systolic hypertension.
5 1 . The method of claim 50, wherein the disease or disorder is a vascular disease or disorder, vascular permeability, nonfunctional blood vessels, vascular hypertrophy, vascular remodeling, vascular stiffness, atherosclerosis, peripheral arterial occlusive disease (PAOD), restenosis, thrombosis, vascular permeability disorders, ischemia, reperfusion damage, ischemia or reperfusion damage of the heart, kidney or retina, or a combination thereof.
52. The method of claim 47, further comprising co-administering an additional apelin receptor agonist in combination with a compound of structural Formula I.
PCT/US2015/032748 2014-05-28 2015-05-27 Agonists of the apelin receptor and methods of use thereof WO2015184011A2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US15/313,938 US10570128B2 (en) 2014-05-28 2015-05-27 Agonists of the apelin receptor and methods of use thereof
US16/742,818 US10988475B2 (en) 2014-05-28 2020-01-14 Agonists of the apelin receptor and methods of use thereof
US17/208,919 US20210363142A1 (en) 2014-05-28 2021-03-22 Agonists of the apelin receptor and methods of use thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201462004195P 2014-05-28 2014-05-28
US62/004,195 2014-05-28

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US15/313,938 A-371-Of-International US10570128B2 (en) 2014-05-28 2015-05-27 Agonists of the apelin receptor and methods of use thereof
US16/742,818 Division US10988475B2 (en) 2014-05-28 2020-01-14 Agonists of the apelin receptor and methods of use thereof

Publications (2)

Publication Number Publication Date
WO2015184011A2 true WO2015184011A2 (en) 2015-12-03
WO2015184011A3 WO2015184011A3 (en) 2016-02-25

Family

ID=54700048

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/032748 WO2015184011A2 (en) 2014-05-28 2015-05-27 Agonists of the apelin receptor and methods of use thereof

Country Status (2)

Country Link
US (3) US10570128B2 (en)
WO (1) WO2015184011A2 (en)

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH1143120A (en) * 1997-07-28 1999-02-16 Ishikawajima Harima Heavy Ind Co Ltd After treatment device of container
WO2016187308A1 (en) * 2015-05-20 2016-11-24 Amgen Inc. Triazole agonists of the apj receptor
WO2017106396A1 (en) * 2015-12-16 2017-06-22 Bristol-Myers Squibb Company Heteroarylhydroxypyrimidinones as agonists of the apj receptor
WO2017165640A1 (en) * 2016-03-24 2017-09-28 Bristol-Myers Squibb Company 6-hydroxy-4-oxo-1,4-dihydropyrimidine-5-carboxamides as apj agonists
WO2017192485A1 (en) 2016-05-03 2017-11-09 Amgen Inc. Heterocyclic triazole compounds as agonists of the apj receptor
WO2017218633A1 (en) * 2016-06-14 2017-12-21 Bristol-Myers Squibb Company 6-hydroxy-5-(phenyl/heteroarylsulfonyl)pyrimidin-4(1h)-one as apj agonists
WO2018071526A1 (en) 2016-10-12 2018-04-19 Research Triangle Institute Heterocyclic apelin receptor (apj) agonists and uses thereof
WO2018093576A1 (en) 2016-11-16 2018-05-24 Amgen Inc. Alkyl substituted triazole compounds as agonists of the apj receptor
WO2018093580A1 (en) 2016-11-16 2018-05-24 Amgen Inc. Triazole pyridyl compounds as agonists of the apj receptor
WO2018093577A1 (en) 2016-11-16 2018-05-24 Amgen Inc. Cycloalkyl substituted triazole compounds as agonists of the apj receptor
WO2018093579A1 (en) 2016-11-16 2018-05-24 Amgen Inc. Triazole phenyl compounds as agonists of the apj receptor
WO2018097945A1 (en) 2016-11-16 2018-05-31 Amgen Inc. Heteroaryl-substituted triazoles as apj receptor agonists
WO2018097944A1 (en) 2016-11-16 2018-05-31 Amgen Inc. Triazole furan compounds as agonists of the apj receptor
WO2019089335A1 (en) 2017-11-03 2019-05-09 Amgen Inc. Fused triazole agonists of the apj receptor
US10392347B2 (en) 2015-10-14 2019-08-27 Bristol-Myers Squibb Company 2,4-dihydroxy-nicotinamides as APJ agonists
WO2019213006A1 (en) 2018-05-01 2019-11-07 Amgen Inc. Substituted pyrimidinones as agonists of the apj receptor
EP3438282A4 (en) * 2016-03-29 2020-05-06 Advanced Telecommunications Research Institute International Screening method for candidate substances for active component to prevent or treat at least one disease selected from the group consisting of renal hypofunction, chronic kidney disease and kidney failure
US10889565B2 (en) 2016-06-14 2021-01-12 Bristol-Myers Squibb Company 4-hydroxy-3-sulfonylpyridin-2(1H)-ones as APJ receptor agonists
US10988475B2 (en) 2014-05-28 2021-04-27 Sanford Burnham Prebys Medical Discovery Institute Agonists of the apelin receptor and methods of use thereof
US11180539B2 (en) 2016-03-29 2021-11-23 Karydo Therapeutix, Inc. Pharmaceutical composition or food composition, and method for assessing effect of active ingredient in vivo
US11244760B2 (en) 2015-06-25 2022-02-08 Karydo Therapeutix, Inc. Prediction device based on inter-organ cross talk system
US11345705B2 (en) 2017-08-09 2022-05-31 Sanford Burnham Prebys Medical Discovery Institute Substituted pyrazolo[1,5-a]pyrimidines as apelin receptor agonists
US11401244B2 (en) 2014-06-06 2022-08-02 Research Triangle Institute Apelin receptor (APJ) agonists and uses thereof
US11535630B2 (en) 2015-12-09 2022-12-27 Research Triangle Institute Apelin receptor (APJ) agonists and uses thereof

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020046753A1 (en) 2018-08-27 2020-03-05 Oregon Health & Science University Parp inhibitors for treating cancer and asthma

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2008518964A (en) 2004-11-01 2008-06-05 ユニバーシティ オブ サザン カリフォルニア Novel compounds for the treatment of cancer and diseases associated with angiogenic function
US20130158063A1 (en) * 2010-08-24 2013-06-20 Georgetown University Compounds, Compositions and Methods Related to PPAR Antagonists
WO2015184011A2 (en) 2014-05-28 2015-12-03 Sanford-Burnham Medical Research Institute Agonists of the apelin receptor and methods of use thereof

Cited By (71)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH1143120A (en) * 1997-07-28 1999-02-16 Ishikawajima Harima Heavy Ind Co Ltd After treatment device of container
US10988475B2 (en) 2014-05-28 2021-04-27 Sanford Burnham Prebys Medical Discovery Institute Agonists of the apelin receptor and methods of use thereof
US11401244B2 (en) 2014-06-06 2022-08-02 Research Triangle Institute Apelin receptor (APJ) agonists and uses thereof
KR20180011164A (en) * 2015-05-20 2018-01-31 암젠 인크 Triazole compounds of the APJ receptor
US9656997B2 (en) 2015-05-20 2017-05-23 Amgen Inc. Triazole agonists of the APJ receptor
US9656998B2 (en) 2015-05-20 2017-05-23 Amgen Inc. Intermediates for preparing triazole agonists of the APJ receptor
US10344016B2 (en) 2015-05-20 2019-07-09 Amgen Inc. Bromotriazole intermediates
US9745286B2 (en) 2015-05-20 2017-08-29 Amgen Inc. Triazole agonists of the APJ receptor
US9751864B2 (en) 2015-05-20 2017-09-05 Amgen Inc. Methods for preparing triazole agonists of the APJ receptor
KR102653667B1 (en) 2015-05-20 2024-04-03 암젠 인크 Triazole agonists of APJ receptors
WO2016187308A1 (en) * 2015-05-20 2016-11-24 Amgen Inc. Triazole agonists of the apj receptor
US9845310B2 (en) 2015-05-20 2017-12-19 Amgen Inc. Intermediates for preparing triazole agonists of the APJ receptor
EA034776B1 (en) * 2015-05-20 2020-03-19 Эмджен Инк. Triazole agonists of the apj receptor
US9868721B2 (en) 2015-05-20 2018-01-16 Amgen Inc. Triazole agonists of the APJ receptor
US10221162B2 (en) 2015-05-20 2019-03-05 Amgen Inc. Triazole agonists of the APJ receptor
JP2017061546A (en) * 2015-05-20 2017-03-30 アムジエン・インコーポレーテツド Triazole agonists of apj receptor
US10058550B2 (en) 2015-05-20 2018-08-28 Amgen Inc. Methods of treating heart failure
AU2016263564B2 (en) * 2015-05-20 2019-12-05 Amgen Inc. Triazole agonists of the APJ receptor
US9573936B2 (en) 2015-05-20 2017-02-21 Amgen Inc. Triazole agonists of the APJ receptor
US11244760B2 (en) 2015-06-25 2022-02-08 Karydo Therapeutix, Inc. Prediction device based on inter-organ cross talk system
US10392347B2 (en) 2015-10-14 2019-08-27 Bristol-Myers Squibb Company 2,4-dihydroxy-nicotinamides as APJ agonists
USRE49594E1 (en) 2015-12-09 2023-08-01 Research Triangle Institute Apelin receptor (APJ) agonists and uses thereof
US11535630B2 (en) 2015-12-09 2022-12-27 Research Triangle Institute Apelin receptor (APJ) agonists and uses thereof
US10669261B2 (en) 2015-12-16 2020-06-02 Bristl-Myers Squibb Company Heteroarylhydroxypyrimidinones as agonists of the APJ receptor
EA036736B1 (en) * 2015-12-16 2020-12-14 Бристол-Майерс Сквибб Компани Heteroarylhydroxypyrimidinones as agonists of the apj receptor
CN108602813A (en) * 2015-12-16 2018-09-28 百时美施贵宝公司 Heteroaryl hydroxypyrimidinone as apj receptor agonist
AU2016372048B2 (en) * 2015-12-16 2021-02-04 Bristol-Myers Squibb Company Heteroarylhydroxypyrimidinones as agonists of the APJ receptor
CN108602813B (en) * 2015-12-16 2021-10-29 百时美施贵宝公司 Heteroaryl hydroxypyrimidinones as APJ receptor agonists
JP2018538304A (en) * 2015-12-16 2018-12-27 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company Heteroarylhydroxypyrimidinone as an APJ agonist of the APJ receptor
WO2017106396A1 (en) * 2015-12-16 2017-06-22 Bristol-Myers Squibb Company Heteroarylhydroxypyrimidinones as agonists of the apj receptor
AU2017238504B2 (en) * 2016-03-24 2021-05-27 Bristol-Myers Squibb Company 6-hydroxy-4-oxo-1,4-dihydropyrimidine-5-carboxamides as APJ agonists
US10106528B2 (en) 2016-03-24 2018-10-23 Bristol-Myers Squibb Company 6-hydroxy-4-oxo-1,4-dihydropyrimidine-5-carboxamides as APJ agonists
WO2017165640A1 (en) * 2016-03-24 2017-09-28 Bristol-Myers Squibb Company 6-hydroxy-4-oxo-1,4-dihydropyrimidine-5-carboxamides as apj agonists
TWI744301B (en) * 2016-03-24 2021-11-01 美商必治妥美雅史谷比公司 6-hydroxy-4-oxo-1,4-dihydropyrimidine-5-carboxamides as apj agonists
US10590113B2 (en) 2016-03-24 2020-03-17 Bristol-Myers Squibb Company 6-hydroxy-4-oxo-1,4-dihydropyrimidine-5-carboxamides as APJ agonists
JP2019509319A (en) * 2016-03-24 2019-04-04 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company 6-Hydroxy-4-oxo-1,4-dihydropyrimidine-5-carboxamide as an APJ agonist
EA037162B1 (en) * 2016-03-24 2021-02-12 Бристол-Маерс Сквибб Компани 6-hydroxy-4-oxo-1,4-dihydropyrimidine-5-carboxamides as apj agonists
KR20180129836A (en) * 2016-03-24 2018-12-05 브리스톨-마이어스 스큅 컴퍼니 6-hydroxy-4-oxo-1,4-dihydropyrimidine-5-carboxamide as an APJ agonist
KR102433280B1 (en) 2016-03-24 2022-08-17 브리스톨-마이어스 스큅 컴퍼니 6-Hydroxy-4-oxo-1,4-dihydropyrimidine-5-carboxamide as an APJ agonist
US11180539B2 (en) 2016-03-29 2021-11-23 Karydo Therapeutix, Inc. Pharmaceutical composition or food composition, and method for assessing effect of active ingredient in vivo
EP3438282A4 (en) * 2016-03-29 2020-05-06 Advanced Telecommunications Research Institute International Screening method for candidate substances for active component to prevent or treat at least one disease selected from the group consisting of renal hypofunction, chronic kidney disease and kidney failure
US10150760B2 (en) 2016-05-03 2018-12-11 Amgen Inc. Compounds for use in preparing heterocyclic triazole agonists of the APJ receptor
US9988369B2 (en) 2016-05-03 2018-06-05 Amgen Inc. Heterocyclic triazole compounds as agonists of the APJ receptor
WO2017192485A1 (en) 2016-05-03 2017-11-09 Amgen Inc. Heterocyclic triazole compounds as agonists of the apj receptor
US10889565B2 (en) 2016-06-14 2021-01-12 Bristol-Myers Squibb Company 4-hydroxy-3-sulfonylpyridin-2(1H)-ones as APJ receptor agonists
CN109311821B (en) * 2016-06-14 2022-10-14 百时美施贵宝公司 6-hydroxy-5- (phenyl/heteroarylsulfonyl) pyrimidin-4 (1H) -ones as APJ agonists
WO2017218633A1 (en) * 2016-06-14 2017-12-21 Bristol-Myers Squibb Company 6-hydroxy-5-(phenyl/heteroarylsulfonyl)pyrimidin-4(1h)-one as apj agonists
US10508104B2 (en) 2016-06-14 2019-12-17 Bristol-Myers Squibb Company 6-hydroxy-5-(phenyl/heteroarylsulfonyl)pyrimidin-4(1H)-one as APJ agonists
CN109311821A (en) * 2016-06-14 2019-02-05 百时美施贵宝公司 6- hydroxyl -5- (phenyl/heteroarylsulfonyl) pyrimidine -4 (1H) -one as APJ agonist
WO2018071526A1 (en) 2016-10-12 2018-04-19 Research Triangle Institute Heterocyclic apelin receptor (apj) agonists and uses thereof
JP2019534848A (en) * 2016-10-12 2019-12-05 アールティーアイ インターナショナル Heterocyclic apelin receptor (APJ) agonists and their use
JP7104690B2 (en) 2016-10-12 2022-07-21 リサーチ トライアングル インスティテュート Heterocyclic apelin receptor (APJ) agonists and their use
US11926612B2 (en) 2016-10-12 2024-03-12 Research Triangle Institute Heterocyclic apelin receptor (APJ) agonists and uses thereof
RU2764039C2 (en) * 2016-10-12 2022-01-14 Рисерч Трайэнгл Инститьют Heterocyclic agonists of apelin receptor (apj) and their application
WO2018093577A1 (en) 2016-11-16 2018-05-24 Amgen Inc. Cycloalkyl substituted triazole compounds as agonists of the apj receptor
WO2018097945A1 (en) 2016-11-16 2018-05-31 Amgen Inc. Heteroaryl-substituted triazoles as apj receptor agonists
US10689367B2 (en) 2016-11-16 2020-06-23 Amgen Inc. Triazole pyridyl compounds as agonists of the APJ receptor
US11046680B1 (en) 2016-11-16 2021-06-29 Amgen Inc. Heteroaryl-substituted triazoles as APJ receptor agonists
US11191762B2 (en) 2016-11-16 2021-12-07 Amgen Inc. Alkyl substituted triazole compounds as agonists of the APJ Receptor
US11020395B2 (en) 2016-11-16 2021-06-01 Amgen Inc. Cycloalkyl substituted triazole compounds as agonists of the APJ receptor
WO2018093576A1 (en) 2016-11-16 2018-05-24 Amgen Inc. Alkyl substituted triazole compounds as agonists of the apj receptor
US10736883B2 (en) 2016-11-16 2020-08-11 Amgen Inc. Triazole furan compounds as agonists of the APJ receptor
WO2018093579A1 (en) 2016-11-16 2018-05-24 Amgen Inc. Triazole phenyl compounds as agonists of the apj receptor
WO2018093580A1 (en) 2016-11-16 2018-05-24 Amgen Inc. Triazole pyridyl compounds as agonists of the apj receptor
US10906890B2 (en) 2016-11-16 2021-02-02 Amgen Inc. Triazole phenyl compounds as agonists of the APJ receptor
WO2018097944A1 (en) 2016-11-16 2018-05-31 Amgen Inc. Triazole furan compounds as agonists of the apj receptor
US11345705B2 (en) 2017-08-09 2022-05-31 Sanford Burnham Prebys Medical Discovery Institute Substituted pyrazolo[1,5-a]pyrimidines as apelin receptor agonists
US11149040B2 (en) 2017-11-03 2021-10-19 Amgen Inc. Fused triazole agonists of the APJ receptor
WO2019089335A1 (en) 2017-11-03 2019-05-09 Amgen Inc. Fused triazole agonists of the apj receptor
US11807624B2 (en) 2018-05-01 2023-11-07 Amgen Inc. Substituted pyrimidinones as agonists of the APJ receptor
WO2019213006A1 (en) 2018-05-01 2019-11-07 Amgen Inc. Substituted pyrimidinones as agonists of the apj receptor

Also Published As

Publication number Publication date
US20200354355A1 (en) 2020-11-12
US10988475B2 (en) 2021-04-27
US10570128B2 (en) 2020-02-25
US20170197958A1 (en) 2017-07-13
WO2015184011A3 (en) 2016-02-25
US20210363142A1 (en) 2021-11-25

Similar Documents

Publication Publication Date Title
US10988475B2 (en) Agonists of the apelin receptor and methods of use thereof
US11673873B2 (en) Apelin receptor agonists and methods of use thereof
US20110312992A1 (en) Inhibitors of C-Kit and Uses Thereof
EP3551610B1 (en) Bicyclo[1.1.1]pentane inhibitors of dual leucine zipper (dlk) kinase for the treatment of disease
US20230037225A1 (en) SUBSTITUTED PYRAZOLO[1,5-a]PYRIMIDINES AS APELIN RECEPTOR AGONISTS
US11058688B2 (en) Imidazopiperazine inhibitors of transcription activating proteins
US20080021217A1 (en) Heterocyclic inhibitors of rho kinase
WO2007008529A2 (en) Celullar cholesterol absorption modifiers
US10392389B2 (en) Heterocyclic compounds for the inhibition of PASK
US20090318485A1 (en) Novel inhibitors of rho kinase
WO2007015866A2 (en) Inhibitors of p38 kinase and methods of treating inflammatory disorders
EP2681207B1 (en) Heterocyclic compounds for the inhibition of pask
US10899769B2 (en) Imidazopiperazinone inhibitors of transcription activating proteins
WO2010016846A1 (en) Heterocyclic modulators of tgr5 for treatment of disease
CN114302881A (en) Compounds useful in the treatment and prevention of muscle atrophy and other conditions
US20230312588A1 (en) Imidazopiperazine inhibitors of transcription activating proteins
US20230295173A1 (en) Imidazopiperazine inhibitors of transcription activating proteins
US20210361773A1 (en) Compounds and methods for selective proteolysis of glucocorticoid receptors

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 15800366

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 15313938

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 15800366

Country of ref document: EP

Kind code of ref document: A2