WO2011005929A1 - Piperidine derivative and its use for the treatment of diabets and obesity - Google Patents

Piperidine derivative and its use for the treatment of diabets and obesity Download PDF

Info

Publication number
WO2011005929A1
WO2011005929A1 PCT/US2010/041325 US2010041325W WO2011005929A1 WO 2011005929 A1 WO2011005929 A1 WO 2011005929A1 US 2010041325 W US2010041325 W US 2010041325W WO 2011005929 A1 WO2011005929 A1 WO 2011005929A1
Authority
WO
WIPO (PCT)
Prior art keywords
methyl
carbonitrile
amino
disease
acetyl
Prior art date
Application number
PCT/US2010/041325
Other languages
French (fr)
Inventor
Robert M. Jones
Juerg Lehmann
Original Assignee
Arena Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Arena Pharmaceuticals, Inc. filed Critical Arena Pharmaceuticals, Inc.
Publication of WO2011005929A1 publication Critical patent/WO2011005929A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system

Definitions

  • the present invention relates to the GPRl 19 agonist, N-(2-fluoro-4-(methylsulfonyl) phenyl)-5-methyl-6-( 1 -(5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine (Compound 1 , Formula (I)), and pharmaceutically acceptable salts, solvates, and hydrates thereof, that are useful in the treatment of GPRl 19-related disorders, such as, metabolic-related disorders and complications thereof, such as, diabetes and obesity.
  • Diabetes mellitus is a serious disease afflicting over 100 million people worldwide. In the United States, there are more than 12 million diabetics, with 600,000 new cases diagnosed each year.
  • Diabetes mellitus is a diagnostic term for a group of disorders characterized by abnormal glucose homeostasis resulting in elevated blood sugar.
  • IDDM insulin-dependent diabetes mellitus
  • NIDDM non-insulin-dependent diabetes mellitus
  • the etiology of the different types of diabetes is not the same; however, everyone with diabetes has two things in common: overproduction of glucose by the liver and little or no ability to move glucose out of the blood into the cells where it becomes the body's primary fuel.
  • Diabetes is a syndrome with interrelated metabolic, vascular, and neuropathic components.
  • the metabolic syndrome generally characterized by hyperglycemia, comprises alterations in carbohydrate, fat and protein metabolism caused by absent or markedly reduced insulin secretion and/or ineffective insulin action.
  • the vascular syndrome consists of abnormalities in the blood vessels leading to cardiovascular, retinal and renal complications. Abnormalities in the peripheral and autonomic nervous systems are also part of the diabetic syndrome.
  • IDDM is characterized by low or undetectable levels of endogenous insulin production caused by destruction of the insulin-producing ⁇ cells of the pancreas, the characteristic that most readily distinguishes IDDM from NIDDM. IDDM, once termed juvenile-onset diabetes, strikes young and older adults alike.
  • NIDDM type 2
  • NIDDM Approximately 90 to 95% of people with diabetes have type 2 (or NIDDM). NIDDM subjects produce insulin, but the cells in their bodies are insulin resistant: the cells don't respond properly to the hormone, so glucose accumulates in their blood. NIDDM is characterized by a relative disparity between endogenous insulin production and insulin requirements, leading to elevated blood glucose levels. In contrast to IDDM, there is always some endogenous insulin production in NIDDM; many NIDDM patients have normal or even elevated blood insulin levels, while other NIDDM patients have inadequate insulin production (Rotwein, R. et al. N. Engl. J. Med. 308, 65-71 (1983)). Most people diagnosed with NIDDM are age 30 or older, and half of all new cases are age 55 and older. Compared with whites and Asians, NIDDM is more common among Native Americans, African- Americans, Latinos, and Hispanics. In addition, the onset can be insidious or even clinically inapparent, making diagnosis difficult.
  • obesity is characterized by hyperinsulinemia and insulin resistance, a feature shared with NIDDM, hypertension and atherosclerosis.
  • Kidney disease also called nephropathy
  • Diabetes occurs when the kidney's "filter mechanism” is damaged and protein leaks into urine in excessive amounts and eventually the kidney fails. Diabetes is also a leading cause of damage to the retina at the back of the eye and increases risk of cataracts and glaucoma.
  • diabetes is associated with nerve damage, especially in the legs and feet, which interferes with the ability to sense pain and contributes to serious infections. Taken together, diabetes complications are one of the nation's leading causes of death.
  • Obesity and diabetes are among the most common human health problems in industrialized societies. In industrialized countries a third of the population is at least 20% overweight. In the United States, the percentage of obese people has increased from 25% at the end of the 1970's, to 33% at the beginning the 1990's. Obesity is one of the most important risk factors for NIDDM. Definitions of obesity differ, but in general, a subject weighing at least 20% more than the recommended weight for his/her height and build is considered obese. The risk of developing NIDDM is tripled in subjects 30% overweight, and three-quarters with NIDDM are overweight.
  • Obesity which is the result of an imbalance between caloric intake and energy expenditure, is highly correlated with insulin resistance and diabetes in experimental animals and human.
  • Whether someone is classified as overweight or obese can be determined by a number of different methods, such as, on the basis of their body mass index (BMI) which is calculated by dividing body weight (kg) by height squared (m 2 ).
  • BMI body mass index
  • m 2 height squared
  • Overweight is defined as a BMI in the range 25-30 kg/m 2
  • obesity as a BMI greater than 30 kg/m 2 (see TABLE below).
  • problems with this definition in that it does not take into account the proportion of body mass that is muscle in relation to fat (adipose tissue).
  • obesity can be defined on the basis of body fat content: greater than 25% and 30% in males and females, respectively.
  • Coronary insufficiency, atheromatous disease, and cardiac insufficiency are at the forefront of the cardiovascular complication induced by obesity. It is estimated that if the entire population had an ideal weight, the risk of coronary insufficiency would decrease by 25% and the risk of cardiac insufficiency and of cerebral vascular accidents by 35%. The incidence of coronary diseases is doubled in subjects less than 50 years of age who are 30% overweight.
  • Atherosclerosis is a complex disease characterized by inflammation, lipid accumulation, cell death and fibrosis. Atherosclerosis is characterized by cholesterol deposition and monocyte infiltration into the subendothelial space, resulting in foam cell formation. Thrombosis subsequent to atherosclerosis leads to myocardial infarction and stroke. Atherosclerosis is the leading cause of mortality in many countries, including the United States. (See, e.g., Ruggeri, Nat Med (2002) 8:1227-1234; Arehart et al, Circ Res, Circ. Res. (2008) 102:986-993.)
  • Osteoporosis is a disabling disease characterized by the loss of bone mass and microarchitectural deterioration of skeletal structure leading to compromised bone strength, which predisposes a patient to increased risk of fragility fractures. Osteoporosis affects more than 75 million people in Europe, Japan and the United States, and causes more than 2.3 million fractures in Europe and the United States alone. In the United States, osteoporosis affects at least 80 years. One in three women older than 50 years will have an osteoporotic fracture that causes a considerable social and financial burden on society. The disease is not limited to women; older men also can be affected. By 2050, the worldwide incidence of hip fracture in men is projected to increase by 310% and 240% in women.
  • IBD Inflammatory bowel disease
  • Crohn's disease ulcerative colitis
  • ulcerative proctitis U.S. medical costs of inflammatory bowel disease for 1990 have been estimated to be $1.4 to $1.8 billion. Lost productivity has been estimated to have added an additional $0.4 to $0.8 billion, making the estimated cost of inflammatory bowel disease $1.8 to $2.6 billion.
  • Enteritis refers to inflammation of the intestine, especially the small intestine, a general condition that can have any of numerous different causes. Enterocolitis refers to inflammation of the small intestine and colon.
  • CD Crohn's disease
  • Granulomatous colitis is another name for Crohn's disease that affects the colon.
  • Ileitis is CD of the ileum which is the third part of the small intestine.
  • Crohn's colitis is CD affecting part or all of the colon.
  • Ulcerative colitis (UC) is an inflammatory disease of the large intestine, commonly called the colon. UC causes inflammation and ulceration of the inner lining of the colon and rectum. The inflammation of UC is usually most severe in the rectal area with severity diminishing (at a rate that varies from patient to patient) toward the cecum, where the large and small intestine join. Inflammation of the rectum is called proctitis.
  • Inflammation of the sigmoid colon (located just above the rectum) is called sigmoiditis. Inflammation involving the entire colon is termed pancolitis. The inflammation causes the colon to empty frequently resulting in diarrhea. As the lining of the colon is destroyed ulcers form releasing mucus, pus and blood. Ulcerative proctitis is a form of UC that affects only the rectum.
  • GPRl 19 is a G protein-coupled receptor (GPRl 19; e.g., human GPRl 19, GenBank ® Accession No. AAP72125 and alleles thereof; e.g., mouse GPRl 19, GenBank ® Accession No. AY288423 and alleles thereof) and is selectively expressed on pancreatic beta cells. GPRl 19 activation leads to elevation of a level of intracellular cAMP, consistent with GPRl 19 being coupled to Gs.
  • GPRl 19 G protein-coupled receptor
  • GPRl 19 Agonists to GPRl 19 stimulate glucose-dependent insulin secretion in vitro and lower an elevated blood glucose level in vivo; see, e.g., International Applications WO 04/065380 and WO 04/076413, and EP 1338651, the disclosure of each of which is herein incorporated by reference in its entirety.
  • GPRl 19 has also been referred to as RUP3 (see, International Application WO 00/31258) and as Glucose-Dependent Insulinotropic Receptor
  • GPRl 19 agonists also stimulate the release of Glucose-dependent Insulinotropic
  • GIP Polypeptide
  • GLP-I Glucagon-Like Peptide- 1
  • PYY Peptide YY
  • GPRl 19 agonists enhance incretin release and therefore can be used in treatment of disorders related to the incretins, such as, GIP, GLP-I, and PYY.
  • GIP and GLP-I are substrates for the enzyme DPP-IV. Jones and co-workers (Jones, et al., Ann. Rep. Med.
  • G Glucose-dependent Insulinotropic Poloypeptide
  • GIP Glucose-dependent insulinotropic polypeptide
  • gastric inhibitory polypeptide is a peptide incretin hormone of 42 amino acids that is released from duodenal endocrine K cells after meal ingestion. The amount of GIP released is largely dependent on the amount of glucose consumed. GIP has been shown to stimulate glucose-dependent insulin secretion in pancreatic beta cells. GIP mediates its actions through a specific G protein-coupled receptor, namely GIPR.
  • GIP contains an alanine at position 2, it is an excellent substrate for dipeptidyl peptidase-4 (DPP-IV), an enzyme regulating the degradation of GIP.
  • DPP-IV dipeptidyl peptidase-4
  • Full-length GIP( 1 -42) is rapidly converted to bioinactive GIP(3-42) within minutes of secretion from the gut K cell. Inhibition of DPP-IV has been shown to augment GIP bioactivity.
  • GIP has been shown to promote bone formation.
  • GIP has been shown to activate osteoblastic receptors, resulting in increases in collagen type I synthesis and alkaline phosphatase activity, both associated with bone formation.
  • GIP has been shown to inhibit osteoclast activity and differentiation in vitro.
  • GIP administration has been shown to prevent the bone loss due to ovariectomy.
  • GIP receptor (GIPR) knockout mice evidence a decreased bone size, lower bone mass, altered bone microarchitecture and biochemical properties, and altered parameters for bone turnover, especially in bone formation.
  • GIP Glucagon-Like Peptide-1
  • Glucagon- like peptide-1 (GLP-I) is an incretin hormone derived from the
  • GLP-I mediates its actions through a specific G protein-coupled receptor (GPCR), namely GLP-IR.
  • GPCR G protein-coupled receptor
  • GLP- 1 is best characterized as a hormone that regulates glucose homeostasis. GLP- 1 has been shown to stimulate glucose-dependent insulin secretion and to increase pancreatic beta cell mass.
  • GLP- 1 has also been shown to reduce the rate of gastric emptying and to promote satiety.
  • the efficacy of GLP- 1 peptide agonists in controlling blood glucose in Type 2 diabetics has been demonstrated in several clinical studies [see, e.g., Nauck et al., Drug News Perspect (2003) 16:413-422], as has its efficacy in reducing body mass [Zander et al., Lancet (2002) 359:824- 830].
  • GLP- 1 receptor agonists are additionally useful in protecting against myocardial infarction and against cognitive and neurodegenerative disorders.
  • GLP-I has been shown to be cardioprotective in a rat model of myocardial infarction [Bose et al., Diabetes (2005) 54: 146- 151], and GLP-IR has been shown in rodent models to be involved in learning and
  • Type 2 diabetes are characterized by a deficiency in GLP- 1 [see, e.g., Nauck et al., Diabetes (2004) 53 Suppl 3 :S 190- 196].
  • Peptide YY is a 36 amino acid peptide originally isolated in 1980 from porcine intestine (Tatemoto et al, Nature (1980) 285:417-418). PYY is secreted from enteroendocrine L- cells within both the large and small intestine. It has been shown that in rat and human gut concentrations of immunoreactive PYY are low in duodenum and jenunum, high in ileum and colon, and highest in rectum (Lundberg et al, PNAS USA (1982) 79:4471-4475; Adrian et al, Gastroenterol.
  • PYY 3 _ 36 is generated from PYYi_ 36 by cleavage of the N-terminal Tyr and Pro residues by dipeptidyl peptidase IV.
  • PYY 3 _ 3 6 is the predominant form of PYY in human postprandial plasma (Grandt et al, Regul. Pept. (1994) 51 : 151-159).
  • PYYi_ 36 and PYY 3 _ 36 have been reported to have comparable agonist activity at NPY Y2 receptor (Y2R), a G protein- coupled receptor (Parker et al, Br. J. Pharmacol. (2008) 153:420-431); however, PYY 3 _ 36 has been reported to be a high-affinity Y2R selective agonist (Keire et al, Am. J. Physiol.
  • mice were treated with weight loss in mice after peripheral administration (Morley Qt ⁇ , Life Sciences (1987) 41 :2157-2165).
  • Peripheral administration of PYY 3 _ 36 has been reported to markedly reduce food intake and weight gain in rats, to decrease appetite and food intake in humans, and to decrease food intake in mice, but not in Y2R-null mice, which was said to suggest that the food intake effect requires the Y2R.
  • infusion of PYY 3 _ 3 6 was found to significantly decrease appetite and reduce food intake by 33% over 24 hours.
  • Peripheral administration of PYY 3 _ 3 6 has been reported to reduce food intake, body weight gain and glycemic indices in diverse rodent models of metabolic diseases of both sexes (Pittner et al, Int. J. Obes. Relat. Metab. Disord. (2004) 28:963-971). It has been reported that blockade of Y2R with the specific antagonist BIIE-246 attenuates the effect of peripherally administered endogenous and exogenous PYY 3 _ 36 for reducing food intake (Abbott et al, Brain Res (2005) 1043:139-144).
  • peripheral administration of a novel long- acting selective Y2R polyethylene glycol-conjugated peptide agonist reduces food intake and improves glucose metabolism (glucose disposal, plasma insulin and plasma glucose) in rodents (Ortiz et al, JPET (2007) 323:692-700; Lamb et al, J. Med. Chem. (2007) 50:2264-2268). It has been reported that PYY ablation in mice leads to the development of hyperinsulinemia and obesity (Boey et al, Diabetologia (2006) 49: 1360-1370). It has been reported that peripheral administration of a long-acting, potent and highly selective Y2R agonist inhibits food intake and promotes fat metabolism in mice (Balasubramaniam et al, Peptides (2007) 28:235-240).
  • Y2R agonists such as PYYi_36 and PYY3_36 can confer protection against epileptic seizures, such as against kainate seizures (El Bahh et al, Eur. J. Neurosci. (2005) 22: 1417-1430; Woldbye et al, Neurobiology of Disease (2005) 20:760-772).
  • Y2R agonists such as PYYi_ 36 and PYY 3 - 36 act as proabsorbtive (or anti-secretory) hormones, increasing upon intravenous administration the absorption of both water and sodium in various parts of the bowel (Bilchik et al, Gastroenterol. (1993) 105:1441-
  • Y2R agonists such as PYYi_36 and PYY3_36 can confer protection against inflammatory bowel disease such as ulcerative colitis and Crohn's disease (WO
  • PYY-deficient mice exhibit an osteopenic phenotype, i.e. that PYY can increase bone mass and/or can confer protection against loss of bone mass (e.g., decreases loss of bone mass) (Wortley et al, Gastroenterol. (2007) 133: 1534-1543). It has been reported that PYY 3 _ 36 can confer protection in rodent models of pancreatitis (Vona-Davis et al, Peptides (2007) 28:334-338).
  • PYY and Y2R agonists such as PYY3_36 can suppress tumor growth in the cases of, e.g., pancreatic cancer such as pancreatic ductal adenocarcinoma, breast cancer such as breast infiltrative ductal adenocarcinoma, colon cancer such as colon
  • Adiponectin is an adipokine with potent anti-inflammatory properties (Ouchi et al, Clin Chim Acta (2007) 380:24-30; TiIg et al,
  • Adiponectin exerts anti-atherogenic effects by targeting vascular endothelial cells and macrophages and insulin-sensitizing effects, predominantly in muscle and liver (Kubota et al, J. Biol. Chem. (2002) 277:25863-25866; Maeda et al, Nat. Med. (2002) 8:731-737).
  • Low adiponectin levels have been reported to be associated with atherogenic lipoproteins in dyslipidemia (elevated triglycerides, small dense LDL cholesterol, low HDL cholesterol) (Marso et al, Diabetes Care (2008) Feb 5 Epub ahead of print).
  • Adiponectin has been implicated in high density lipoprotein (HDL) assembly (Oku et al, FEBS Letters (2007)
  • Adiponectin has been found to ameliorate the abnormalities of metabolic syndrome, including insulin resistance, hyperglycemia, and dyslipidemia, in a mouse model of obesity-linked metabolic syndrome associated with decreased adiponectin levels (Hara et al,
  • Adiponectin has been reported to stimulate angiogenesis in response to tissue ischemia (Shibata et al, J. Biol. Chem. (2004) 279:28670-28674). Adiponectin has been reported to prevent cerebral ischemic injury through endothelial nitric oxide synthase- dependent mechanisms (Nishimura et al, Circulation (2008) 117:216-223). Adiponectin has been reported to confer protection against myocardial ischemia-reperfusion injury (Shibata et al, Nat
  • Adiponectin has been reported to confer protection against myocardial ischaemia-reperfusion injury via AMP- activated protein kinase, Akt, and nitric oxide (Gonon et al, Cardiovasc Res. (2008) 78:116-122).
  • Adiponectin has been reported to confer protection against the development of systolic dysfunction following myocardial infarction, through its abilities to suppress cardiac hypertrophy and interstitial fibrosis, and proctect against myocyte and capillary loss (Shibata et al, J. MoI. Cell Cardiol. (2007) 42: 1065-1074). Adiponectin has been reported to confer protection against inflammatory lung disease; adiponectin-deficient mice exhibit an emphysema-like phenotype (Summer et al, Am J. Physiol. Lung Cell MoI. Physiol (March 7, 2008)). Adiponectin has been reported to confer protection against allergic airway inflammation and airway
  • hyperresponsiveness such as may be associated with asthma (Shore et al, J. Allergy Clin.
  • Adiponectin has been suggested to confer protection against pulmonary arterial hypertension by virtue of its insulin-sensitizing effects (Hansmann et al, Circulation (2007) 115: 1275-1284). Adiponectin has been reported to ameliorate obesity-related hypertension, with said amelioration of hypertension being associated in part with upregulated prostacyclin expression (Ohashi et al, Hypertension (2006) 47:1108-1116).
  • Adiponectin has been reported to decrease tumor necrosis factor (TNF)- ⁇ - induced expression of the adhesion molecules VCAM-I, E-selectin and ICAM-I in human aortic endothelial cells (HAECs) (Ouchi et al, Circulation (1999) 100:2473-2476) and to inhibit production of TNF- ⁇ in macrophages (Yokota et al, Blood (2000) 96:1723-1732). Adiponectin has been reported to confer protection against restenosis after vascular intervention (Matsuda et al, J Biol Chem (2002) 277:37487-37491).
  • TNF- ⁇ -mediated inflammatory conditions encompass rheumatoid arthritis, inflammatory bowel disease such as Crohn's disease, ankylosing spondylitis, psoriasis, ischemic brain injury, cardiac allograft rejection, asthma, and the like (Bradley, J Pathol (2008) 214:149-160).
  • Figure 1 shows a powder X-ray diffraction (PXRD) pattern for Compound 1 as prepared according to the procedure described in Example 6.
  • FIG. 2 shows a thermogravimetric analysis (TGA) thermogram for Compound 1 as prepared according to the procedure described in Example 6.
  • FIG. 3 shows a differential scanning calorimetry (DSC) thermogram for Compound 1 as prepared according to the procedure described in Example 6.
  • the present invention is drawn to N-(2-fluoro-4-(methylsulfonyl) phenyl)-5-methyl-6-(l- (5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine (Compound 1, Formula (I)) and pharmaceutically acceptable salts, solvates, and hydrates thereof, which bind to and modulate the activity of a GPCR, referred to herein as GPRl 19, and uses thereof.
  • GPRl 19, as used herein includes the human sequences found in GeneBank accession number AY288416, naturally-occurring allelic variants, mammalian orthologs, and recombinant mutants thereof.
  • a preferred human GPRl 19 for use in screening and testing of the compounds of the invention is provided in the nucleotide sequence of Seq. ID.No: 1 and the corresponding amino acid sequence in Seq. ID.No:2 found in PCT Application No. WO2005/007647, the disclosure which is herein incorporated by reference in its entirety.
  • One aspect of the present invention encompasses compounds selected from the following compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • compositions comprising a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, and a pharmaceutically acceptable carrier.
  • compositions comprising a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, and an inhibitor of dipeptidyl peptidase IV (DPP-IV).
  • DPP-IV dipeptidyl peptidase IV
  • compositions wherein the inhibitor of DPP-IV is selected from the following compounds and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • compositions wherein the inhibitor of DPP-IV is selected from the following compounds and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • compositions wherein the inhibitor of DPP-IV is selected from the following compounds and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • Some embodiments of the present invention in regard to the inhibitor of DPP-IV include every combination of one or more inhibitors of DPP-IV and pharmaceutically acceptable salts, solvates, and hydrates thereof, selected from the following group:
  • the inhibitor of dipeptidyl peptidase IV is selected from the following compound and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the inhibitor of dipeptidyl peptidase IV is 3(R)-amino-l-[3- (trifluoromethyl)-5,6,7,8-tetrahydro[l,2,4]triazolo[4,3-a]pyrazin-7-yl]-4-(2,4,5- trifluorophenyl)butan- 1 -one phosphate (1 : 1) monohydrate.
  • compositions further comprising a pharmaceutically acceptable carrier.
  • compositions wherein:
  • the compound is selected from the following compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the inhibitor of dipeptidyl peptidase IV is selected from the following compound and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • compositions further comprising a pharmaceutically acceptable carrier.
  • One aspect of the present invention pertains to methods of modulating the activity of a GPRl 19 receptor by contacting the receptor with a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof.
  • One aspect of the present invention pertains to methods of agonizing a GPRl 19 receptor by contacting the receptor with a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof.
  • One aspect of the present invention pertains to methods for the treatment of a GPRl 19 receptor related disorder in an individual, comprising administering to the individual in need thereof, a therapeutically effective amount of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof.
  • the GPRl 19 receptor related disorder is a metabolic-related disorder.
  • One aspect of the present invention pertains to methods for the treatment of a metabolic- related disorder in an individual, comprising administering to the individual in need thereof, a therapeutically effective amount of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof.
  • One aspect of the present invention pertains to methods for the treatment of a metabolic-related disorder selected from the group consisting of:
  • diabetes mellitus type 1 diabetes, type 2 diabetes, inadequate glucose tolerance, impaired glucose tolerance, insulin resistance, hyperglycemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia, atherosclerosis, stroke, syndrome X, hypertension, pancreatic beta-cell insufficiency, enteroendocrine cell insufficiency, glucosuria, metabolic acidosis, cataracts, diabetic nephropathy, diabetic neuropathy, peripheral neuropathy, diabetic coronary artery disease, diabetic cerebrovascular disease, diabetic peripheral vascular disease, diabetic retinopathy, metabolic syndrome, a condition related to diabetes mellitus, myocardial infarction, learning impairment, memory impairment, a neurodegenerative disorder, a condition ameliorated by increasing a blood GLP- 1 level in an individual with a neurodegenerative disorder, excitotoxic brain damage caused by severe epileptic seizures, Alzheimer's disease, Parkinson's disease, Huntington's disease, prion-associated disease, stroke, motor-
  • One aspect of the present invention pertains to methods for the treatment of type 2 diabetes in an individual, comprising administering to the individual in need thereof, a therapeutically effective amount of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof.
  • One aspect of the present invention pertains to methods for the treatment of
  • hyperglycemia in an individual comprising administering to the individual in need thereof, a therapeutically effective amount of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof.
  • One aspect of the present invention pertains to methods for the treatment of
  • hyperlipidemia in an individual comprising administering to the individual in need thereof, a therapeutically effective amount of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof.
  • a compound selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof.
  • One aspect of the present invention pertains to methods for the treatment of
  • hypertriglyceridemia in an individual comprising administering to the individual in need thereof, a therapeutically effective amount of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof.
  • One aspect of the present invention pertains to methods for the treatment of type 1 diabetes in an individual, comprising administering to the individual in need thereof, a therapeutically effective amount of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof.
  • One aspect of the present invention pertains to methods for the treatment of dyslipidemia in an individual, comprising administering to the individual in need thereof, a therapeutically effective amount of a compound, selected from the compound of Formula (I) and
  • One aspect of the present invention pertains to methods for the treatment of syndrome X in an individual, comprising administering to the individual in need thereof, a therapeutically effective amount of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof.
  • the individual is a mammal.
  • the mammal is a human.
  • One aspect of the present invention pertains to methods of treating obesity in an individual comprising administering to the individual in need of treatment a therapeutically effective amount of a compound, selected from the compound of Formula (I) and
  • One aspect of the present invention pertains to methods of decreasing food intake of an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof.
  • One aspect of the present invention pertains to methods of inducing satiety in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof.
  • One aspect of the present invention pertains to methods of controlling or decreasing weight gain of an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof.
  • One aspect of the present invention pertains to methods for the treatment of a GPRl 19 receptor related disorder in an individual, comprising administering to the individual in need thereof, a therapeutically effective amount of a compound according to claim 1 in combination with an inhibitor of DPP-IV, wherein the compound and the inhibitor of DPP-IV are administered simultaneously, separately, or sequentially.
  • the compound and the inhibitor of DPP-IV are administered simultaneously.
  • the compound and the inhibitor of DPP-IV are administered separately.
  • the compound and the inhibitor of DPP-IV are administered sequentially.
  • the GPRl 19 receptor related disorder is a metabolic-related disorder.
  • the metabolic-related disorder is selected from the group consisting of:
  • diabetes mellitus type 1 diabetes, type 2 diabetes, inadequate glucose tolerance, impaired glucose tolerance, insulin resistance, hyperglycemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia, atherosclerosis, stroke, syndrome X, hypertension, pancreatic beta-cell insufficiency, enteroendocrine cell insufficiency, glucosuria, metabolic acidosis, cataracts, diabetic nephropathy, diabetic neuropathy, peripheral neuropathy, diabetic coronary artery disease, diabetic cerebrovascular disease, diabetic peripheral vascular disease, diabetic retinopathy, metabolic syndrome, a condition related to diabetes mellitus, myocardial infarction, learning impairment, memory impairment, a neurodegenerative disorder, a condition ameliorated by increasing a blood GLP- 1 level in an individual with a neurodegenerative disorder, excitotoxic brain damage caused by severe epileptic seizures, Alzheimer's disease, Parkinson's disease, Huntington's disease, prion-associated disease, stroke, motor-
  • the metabolic-related disorder is type 2 diabetes.
  • the metabolic-related disorder is hyperglycemia.
  • the metabolic-related disorder is hyperlipidemia.
  • the metabolic-related disorder is hypertriglyceridemia.
  • the metabolic-related disorder is type 1 diabetes.
  • the metabolic-related disorder is dyslipidemia.
  • the metabolic-related disorder is syndrome X.
  • One aspect of the present invention pertains to methods of treating obesity in an individual comprising administering to the individual in need of treatment a therapeutically effective amount of a compound according to claim 1 in combination with an inhibitor of DPP- IV, wherein the compound and the inhibitor of DPP-IV are administered simultaneously, separately, or sequentially.
  • One aspect of the present invention pertains to methods of decreasing food intake of an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound according to claim 1 in combination with an inhibitor of DPP-IV, wherein the compound and the inhibitor of DPP-IV are administered simultaneously, separately, or sequentially.
  • One aspect of the present invention pertains to methods of inducing satiety in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound according to claim 1 in combination with an inhibitor of DPP-IV, wherein the compound and the inhibitor of DPP-IV are administered simultaneously, separately, or sequentially.
  • One aspect of the present invention pertains to methods of controlling or decreasing weight gain of an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound according to claim 1 in combination with an inhibitor of DPP-IV, wherein the compound and the inhibitor of DPP-IV are administered simultaneously, separately, or sequentially.
  • the compound and the inhibitor of DPP-IV are administered simultaneously.
  • the compound and the inhibitor of DPP-IV are administered separately.
  • the compound and the inhibitor of DPP-IV are administered sequentially.
  • One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for modulating the activity of a GPRl 19 receptor.
  • One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for agonizing a GPRl 19 receptor.
  • One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for the treatment of a GPRl 19 receptor related disorder.
  • One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for the treatment of a metabolic-related disorder.
  • One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for the treatment of a metabolic- related disorder selected from the group consisting of:
  • diabetes mellitus type 1 diabetes, type 2 diabetes, inadequate glucose tolerance, impaired glucose tolerance, insulin resistance, hyperglycemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia, atherosclerosis, stroke, syndrome X, hypertension, pancreatic beta-cell insufficiency, enteroendocrine cell insufficiency, glucosuria, metabolic acidosis, cataracts, diabetic nephropathy, diabetic neuropathy, peripheral neuropathy, diabetic coronary artery disease, diabetic cerebrovascular disease, diabetic peripheral vascular disease, diabetic retinopathy, metabolic syndrome, a condition related to diabetes mellitus, myocardial infarction, learning impairment, memory impairment, a neurodegenerative disorder, a condition ameliorated by increasing a blood GLP- 1 level in an individual with a neurodegenerative disorder, excitotoxic brain damage caused by severe epileptic seizures, Alzheimer's disease, Parkinson's disease, Huntington's disease, prion-associated disease, stroke, motor-
  • One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for the treatment of type 2 diabetes.
  • One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for the treatment of hyperglycemia.
  • One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for the treatment of hyperlipidemia.
  • One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for the treatment of
  • One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for the treatment of type 1 diabetes.
  • One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for the treatment of dyslipidemia.
  • One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for the treatment of syndrome X.
  • One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for the treatment of obesity.
  • One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for decreasing food intake in an individual.
  • One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for inducing satiety in an individual.
  • One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for controlling or decreasing weight gain in an individual.
  • One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in a method of treatment of the human or animal body by therapy.
  • One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in a method of modulating the activity of a GPRl 19 receptor.
  • One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in a method of agonizing a GPRl 19 receptor.
  • One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in a method of treatment of a GPRl 19 receptor related disorder.
  • One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in a method of treatment of a metabolic-related disorder.
  • One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in a method of treatment of a metabolic-related disorder selected from the group consisting of:
  • diabetes mellitus type 1 diabetes, type 2 diabetes, inadequate glucose tolerance, impaired glucose tolerance, insulin resistance, hyperglycemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia, atherosclerosis, stroke, syndrome X, hypertension, pancreatic beta-cell insufficiency, enteroendocrine cell insufficiency, glucosuria, metabolic acidosis, cataracts, diabetic nephropathy, diabetic neuropathy, peripheral neuropathy, diabetic coronary artery disease, diabetic cerebrovascular disease, diabetic peripheral vascular disease, diabetic retinopathy, metabolic syndrome, a condition related to diabetes mellitus, myocardial infarction, learning impairment, memory impairment, a neurodegenerative disorder, a condition ameliorated by increasing a blood GLP- 1 level in an individual with a neurodegenerative disorder, excitotoxic brain damage caused by severe epileptic seizures, Alzheimer's disease, Parkinson's disease, Huntington's disease, prion-associated disease, stroke, motor-
  • One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in a method of treatment of type 2 diabetes.
  • One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in a method of treatment of hyperglycemia.
  • One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in a method of treatment of hyperlipidemia.
  • One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in a method of treatment of hypertriglyceridemia.
  • One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in a method of treatment of type 1 diabetes.
  • One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in a method of treatment of dyslipidemia.
  • One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in a method of treatment of syndrome X.
  • One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in a method of treatment of obesity.
  • One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in decreasing food intake in an individual.
  • One aspect of the present invention pertains to compounds of the present invention for use in inducing satiety in an individual.
  • One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in controlling or decreasing weight gain in an individual.
  • the individual is a mammal. In some embodiments, the mammal is a human. In some embodiments, the human has a body mass index of about 18.5 to about 45 or greater. In some embodiments, the human has a body mass index of about 18.5 to about 45. In some embodiments, the human has a body mass index of about 25 to about 45. In some embodiments, the human has a body mass index of about 30 to about 45. In some embodiments, the human has a body mass index of about 35 to about 45.
  • the present invention includes every combination of one or more metabolic-related disorders selected from the group consisting of diabetes mellitus, type 1 diabetes, type 2 diabetes, inadequate glucose tolerance, impaired glucose tolerance, insulin resistance, hyperglycemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia, atherosclerosis, stroke, syndrome X, hypertension, pancreatic beta-cell insufficiency, enteroendocrine cell insufficiency, glucosuria, metabolic acidosis, cataracts, diabetic nephropathy, diabetic neuropathy, peripheral neuropathy, diabetic coronary artery disease, diabetic cerebrovascular disease, diabetic peripheral vascular disease, diabetic retinopathy, metabolic syndrome, a condition related to diabetes mellitus, myocardial infarction, learning impairment, memory impairment, a neurodegenerative disorder, a condition ameliorated by increasing a blood GLP- 1 level in an individual with a neurodegenerative disorder, excitotoxic brain damage caused by severe epileptic seizures, Alzheimer's disease,
  • Some embodiments of the present invention include every combination of one or more metabolic-related disorder selected from the group consisting of type 2 diabetes, hyperglycemia, hyperinsulinemia, hyperlipidemia, hypertriglyceridemia, insulin resistance, type 1 diabetes, idiopathic type 1 diabetes (type Ib), latent autoimmune diabetes in adults (LADA), early-onset type 2 diabetes (EOD), youth-onset atypical diabetes (YOAD), maturity onset diabetes of the young (MODY), malnutrition-related diabetes, gestational diabetes, coronary heart disease, vascular restenosis, restenosis, restenosis after angioplasty, peripheral vascular disease, claudication, intermittent claudication, cell death associated with myocardial infarction (e.g.
  • necrosis and apoptosis dyslipidemia, post-prandial lipemia, conditions of impaired glucose tolerance (IGT), impaired glucose metabolism, conditions of impaired glucose metabolism, conditions of impaired fasting plasma glucose, metabolic acidosis, ketosis, arthritis, obesity, osteoporosis, hypertension, congestive heart failure, left ventricular hypertrophy, peripheral arterial disease, diabetic retinopathy, macular degeneration, cataract, diabetic nephropathy, glomerulosclerosis, chronic renal failure, diabetic neuropathy, metabolic syndrome, syndrome X, premenstrual syndrome, angina pectoris, thrombosis, atherosclerosis, ischemic stroke, transient ischemic attacks, stroke, erectile dysfunction, skin and connective tissue disorders, foot ulcerations, ulcerative colitis, endothelial dysfunction, and impaired vascular compliance.
  • ITT impaired glucose tolerance
  • impaired glucose metabolism conditions of impaired glucose metabolism, conditions of impaired fasting plasma glucose
  • metabolic acidosis ketosis
  • ketosis arthritis
  • obesity osteoporos
  • One aspect of the present invention pertains to methods of preparing compositions comprising the step of admixing a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, and a pharmaceutically acceptable carrier.
  • One aspect of the present invention pertains to methods of preparing pharmaceutically compositions comprising the step of admixing a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, and a pharmaceutically acceptable carrier.
  • One aspect of the present invention pertains to methods of preparing compositions comprising the step of admixing a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, and an inhibitor of DPP-IV.
  • One aspect of the present invention pertains to methods of preparing compositions comprising the step of admixing a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, with an inhibitor of DPP-IV, and a pharmaceutically acceptable carrier.
  • One aspect of the present invention pertains to methods of preparing compositions further comprising the step of preparing a unit dosage form from the composition.
  • One aspect of the present invention pertains to methods of preparing a dosage form of a pharmaceutical composition for the treatment of a GPRl 19 receptor related disorder in an individual, the pharmaceutical composition comprising a compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, and an inhibitor of DPP-IV; the method comprising:
  • One aspect of the present invention pertains to methods of preparing a dosage form of a pharmaceutical composition for the treatment of a GPRl 19 receptor related disorder selected from the group consisting of:
  • diabetes mellitus type 1 diabetes, type 2 diabetes, inadequate glucose tolerance, impaired glucose tolerance, insulin resistance, hyperglycemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia, atherosclerosis, stroke, syndrome X, hypertension, pancreatic beta-cell insufficiency, enteroendocrine cell insufficiency, glucosuria, metabolic acidosis, cataracts, diabetic nephropathy, diabetic neuropathy, peripheral neuropathy, diabetic coronary artery disease, diabetic cerebrovascular disease, diabetic peripheral vascular disease, diabetic retinopathy, metabolic syndrome, a condition related to diabetes mellitus, myocardial infarction, learning impairment, memory impairment, a neurodegenerative disorder, a condition ameliorated by increasing a blood GLP- 1 level in an individual with a neurodegenerative disorder, excitotoxic brain damage caused by severe epileptic seizures, Alzheimer's disease,
  • Parkinson's disease Huntington's disease, prion-associated disease, stroke, motor-neuron disease, traumatic brain injury, spinal cord injury, and obesity.
  • the combined preparation is in the form of a twin pack.
  • the first pharmaceutically acceptable carrier and the second pharmaceutically acceptable carrier in steps (a) and (b) are different.
  • the first pharmaceutically acceptable carrier and the second pharmaceutically acceptable carrier in steps (a) and (b) are substantially the same.
  • the combined preparation is for simultaneous use.
  • the combined preparation is for separate use.
  • the combined preparation is for sequential use.
  • One aspect of the present invention pertains to methods of preparing a dosage form of a pharmaceutical composition for increasing blood GLP- 1 level in an individual, the
  • composition comprising a compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, and an inhibitor of DPP-IV; the method comprising:
  • the combined preparation is in the form of a twin pack.
  • the first pharmaceutically acceptable carrier and the second pharmaceutically acceptable carrier in steps (a) and (b) are different.
  • the first pharmaceutically acceptable carrier and the second pharmaceutically acceptable carrier in steps (a) and (b) are substantially the same.
  • the combined preparation is for simultaneous use.
  • the combined preparation is for separate use.
  • the combined preparation is for sequential use.
  • the amount of the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, alone and the amount of the inhibitor of DPP-IV alone are therapeutically ineffective in increasing a blood GLP- 1 level in the individual.
  • One aspect of the present invention pertains to compounds of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, for use in combination with an inhibitor of DPP-IV for the treatment of a condition ameliorated by increasing a blood GLP- 1 level in a mammal.
  • the amount of compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, alone is therapeutically ineffective for treatment of a condition ameliorated by increasing a blood GLP- 1 level in a mammal.
  • One aspect of the present invention pertains to an inhibitor of DPP-IV for use in combination with compounds of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, for the treatment of a condition ameliorated by increasing a blood GLP- 1 level in a mammal.
  • the amount of the inhibitor of DPP-IV alone is therapeutically ineffective for treatment of a condition ameliorated by increasing a blood GLP- 1 level in a mammal.
  • condition ameliorated by increasing a blood GLP- 1 level is selected from the group consisting of:
  • diabetes mellitus type 1 diabetes, type 2 diabetes, inadequate glucose tolerance, impaired glucose tolerance, insulin resistance, hyperglycemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia, atherosclerosis, stroke, syndrome X, hypertension, pancreatic beta-cell insufficiency, enteroendocrine cell insufficiency, glucosuria, metabolic acidosis, cataracts, diabetic nephropathy, diabetic neuropathy, peripheral neuropathy, diabetic coronary artery disease, diabetic cerebrovascular disease, diabetic peripheral vascular disease, diabetic retinopathy, metabolic syndrome, a condition related to diabetes mellitus, myocardial infarction, learning impairment, memory impairment, a neurodegenerative disorder, a condition ameliorated by increasing a blood GLP- 1 level in an individual with a neurodegenerative disorder, excitotoxic brain damage caused by severe epileptic seizures, Alzheimer's disease, Parkinson's disease, Huntington's disease, prion-associated disease, stroke, motor-
  • compositions comprising compounds of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, for use in combination with an inhibitor of DPP-IV for the treatment of a condition ameliorated by increasing a blood GLP- 1 level in a mammal.
  • the amount of the compound alone is therapeutically ineffective for treatment of a condition ameliorated by increasing a blood GLP- 1 level in a mammal.
  • One aspect of the present invention pertains to pharmaceutical compositions comprising an inhibitor of DPP-IV for use in combination with compounds of Formula (I) and
  • condition ameliorated by increasing a blood GLP- 1 level is selected from the group consisting of:
  • diabetes mellitus type 1 diabetes, type 2 diabetes, inadequate glucose tolerance, impaired glucose tolerance, insulin resistance, hyperglycemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia, atherosclerosis, stroke, syndrome X, hypertension, pancreatic beta-cell insufficiency, enteroendocrine cell insufficiency, glucosuria, metabolic acidosis, cataracts, diabetic nephropathy, diabetic neuropathy, peripheral neuropathy, diabetic coronary artery disease, diabetic cerebrovascular disease, diabetic peripheral vascular disease, diabetic retinopathy, metabolic syndrome, a condition related to diabetes mellitus, myocardial infarction, learning impairment, memory impairment, a neurodegenerative disorder, a neurodegenerative disorder, excitotoxic brain damage caused by severe epileptic seizures, Alzheimer's disease, Parkinson's disease, Huntington's disease, prion-associated disease, stroke, motor-neuron disease, traumatic brain injury, spinal cord injury, and obesity.
  • One aspect of the present invention pertains to dosage forms of a pharmaceutical composition as described herein.
  • One aspect of the present invention pertains to combined preparations comprising a compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, and an inhibitor of DPP-IV for simultaneous, separate or sequential use for the treatment of diabetes mellitus or a condition related thereto.
  • the amount of the compound selected from compounds of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, alone and the amount of an inhibitor of DPP-IV are therapeutically ineffective in lowering blood glucose levels in a subject.
  • the compounds and inhibitors of DPP-IV are admixed with pharmaceutically acceptable carriers. In some embodiments, the compound and the inhibitor of DPP-IV are admixed with different pharmaceutically acceptable carriers.
  • the condition related to diabetes mellitus is selected from the group consisting of hyperglycemia, impaired glucose tolerance, insulin resistance, pancreatic beta-cell insufficiency, enteroendocrine cell insufficiency, glucosuria, metabolic acidosis, cataracts, diabetic nephropathy, diabetic neuropathy, diabetic retinopathy, diabetic coronary artery disease, diabetic cerebrovascular disease, diabetic peripheral vascular disease, metabolic syndrome, hyperlipidemia, atherosclerosis, stroke, hypertension, and obesity.
  • twin packs comprising compounds of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, and inhibitors of DPP-IV as a combined preparation for the treatment of diabetes mellitus or a condition related thereto.
  • the compound and the inhibitor of DPP-IV are admixed with different pharmaceutically acceptable carriers.
  • the different pharmaceutically acceptable carriers are suitable for administration by different routes.
  • the condition related to diabetes mellitus is selected from the group consisting of hyperglycemia, impaired glucose tolerance, insulin resistance, pancreatic beta-cell insufficiency, enteroendocrine cell insufficiency, glucosuria, metabolic acidosis, cataracts, diabetic nephropathy, diabetic neuropathy, diabetic retinopathy, diabetic coronary artery disease, diabetic cerebrovascular disease, diabetic peripheral vascular disease, metabolic syndrome, hyperlipidemia, atherosclerosis, stroke, hypertension, and obesity.
  • kits comprising (a) a first package comprising a compound according to claim 1 or a pharmaceutical composition thereof, and (b) a second package comprising an inhibitor of DPP-IV.
  • the present disclosure includes all isotopes of atoms occurring in the present compounds, salts and crystalline forms thereof.
  • Isotopes include those atoms having the same atomic number but different mass numbers.
  • isotopes of hydrogen include 2 H (deuterium) and 3 H (tritium).
  • isotopes of carbon include 13 C and 14 C.
  • agonist is intended to mean a moiety that interacts and activates the receptor, such as, the GPRl 19 receptor and initiates a physiological or pharmacological response characteristic of that receptor. For example, when moieties activate the intracellular response upon binding to the receptor, or enhance GTP binding to membranes.
  • composition is intended to mean a material comprising at least two components; for example, and without limitation, a composition comprising a compound of the present invention and a pharmaceutically acceptable carrier.
  • contacting is intended to mean bringing the indicated moieties together, whether in an in vitro system or an in vivo system.
  • "contacting" a GPRl 19 receptor with a compound of the invention includes the administration of a compound of the present invention to an individual, preferably a human, having a GPRl 19 receptor, as well as, for example, introducing a compound of the invention into a sample containing a cellular or more purified preparation containing a GPRl 19 receptor.
  • hydrate as used herein means a compound of the invention or a salt thereof, that further includes a stoichiometric or non- stoichiometric amount of water bound by non- covalent intermolecular forces.
  • in need of treatment and "in need thereof,” when referring to treatment are used interchangeably to mean a judgment made by a caregiver (e.g. physician, nurse, nurse practitioner, etc. in the case of humans; veterinarian in the case of animals, including non-human mammals) that an individual or animal requires or will benefit from treatment. This judgment is made based on a variety of factors that are in the realm of a caregiver's expertise, but that includes the knowledge that the individual or animal is ill, or will become ill, as the result of a disease, condition or disorder that is treatable by the compounds of the invention. Accordingly, the compounds of the invention can be used in a protective or preventive manner; or compounds of the invention can be used to alleviate, inhibit or ameliorate the disease, condition or disorder.
  • a caregiver e.g. physician, nurse, nurse practitioner, etc. in the case of humans; veterinarian in the case of animals, including non-human mammals
  • the term "individual” or “subject” is intended to mean any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates and most preferably humans.
  • the individual is a human and in certain embodiments, the human is an infant, child, adolescent or adult.
  • the individual is at risk for developing a GPRl 19-related disorder.
  • the individual is at risk for developing a metabolic-related disease or disorder.
  • Individuals who are at risk include, but are not limited to, those with hereditary history of a metabolic-related disease or disorder, or those in a state of physical health which puts them at risk for a metabolic-related disease or disorder.
  • the individual has been determined, by the care-giver or someone acting under the guidance of the care-giver, to have a metabolic-related disease or disorder.
  • modulate or modulating is intended to mean an increase or decrease in the amount, quality, response or effect of a particular activity, function or molecule.
  • composition is intended to mean a composition comprising at least one active ingredient; including but not limited to, salts, solvates, and hydrates of compounds of the present invention; whereby the composition is amenable to investigation for a specified, efficacious outcome in a mammal (for example, without limitation, a human).
  • a mammal for example, without limitation, a human.
  • solvate means a compound of the invention or a salt, thereof, that further includes a stoichiometric or non- stoichiometric amount of a solvent bound by non- covalent intermolecular forces.
  • Preferred solvents are volatile, non-toxic, and/or acceptable for administration to humans in trace amounts.
  • terapéuticaally effective amount is intended to mean the amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue, system, animal, individual or human that is being sought by a researcher, veterinarian, medical doctor or other clinician or caregiver; or in an individual, which includes one or more of the following:
  • Preventing the disease for example, preventing a disease, condition or disorder in an individual that may be predisposed to the disease, condition or disorder but does not yet experience or display the pathology or symptomatology of the disease;
  • Inhibiting the disease for example, inhibiting a disease, condition or disorder in an individual that is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., arresting further development of the pathology and/or symptomatology); and
  • Ameliorating the disease for example, ameliorating a disease, condition or disorder in an individual that is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., reversing the pathology and/or symptomatology).
  • 6-(l-(5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine is a "full agonist”.
  • the compound N-(2-fluoro-4-(methylsulfonyl)phenyl)-5-methyl-6-(l-(5-methylpyrazin-2- yl)piperidin-4-yloxy)pyrimidin-4-amine increased cAMP levels in CHO cells expressing human GPRl 19 (GDIR) with an observed EC 50 of 6 nM (see Example IA).
  • N-(2-fluoro-4- (methylsulfonyl)phenyl)-5-methyl-6-(l -(5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4- amine stimulated GLP-I release from mouse GLUTag cells with an observed EC 50 of 54.1 nM (see Example IB) and is a potent and efficacious GPRl 19 agonist in normal Sprague-Dawley rats in vivo.
  • N-(2-fluoro-4-(methylsulfonyl)phenyl)-5-methyl-6-(l-(5- methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine potently increased glucose clearance during the oral glucose tolerance test (oGTT) (see Example 2). Further, N-(2-fluoro-4-(methylsulfonyl)phenyl)-5-methyl-6-(l-(5- methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine potently increased glucose clearance during the oral glucose tolerance test (oGTT) (see Example 2). Further, N-(2-fluoro-4-
  • N-(2-fluoro-4-(methylsulfonyl)phenyl)-5- methyl-6-(l-(5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine inhibited 3A4, 2C9, and
  • 2C19 with approximate IC 50 values for inhibition of 9.05 ⁇ M, 8.82 ⁇ M, and >10 ⁇ M, respectively and showed no inhibition of 1 A2 and 2D6.
  • the approximate IC 50 value of inhibition for 2C9 was 6 ⁇ M and for the other P450s tested (i.e., 2Cl 9 and 3A4) the IC 5 O values were >30 ⁇ M (see Examples 4 and 5).
  • One aspect of the present invention pertains to compounds selected from the following compound N-(2-fluoro-4-(methylsulfonyl)phenyl)-5-methyl-6-(l-(5-methylpyrazin-2- yl)piperidin-4-yloxy)pyrimidin-4-amine (Compound 1), as shown in Formula (I):
  • compounds of the invention are useful in the treatment of additional diseases. Without limitation, these include the following.
  • insulin resistance The most significant pathologies in type 2 diabetes are impaired insulin signaling at its target tissues ("insulin resistance") and failure of the insulin-producing cells of the pancreas to secrete an appropriate degree of insulin in response to a hyperglycemic signal.
  • Current therapies to treat the latter include inhibitors of the ⁇ -cell ATP-sensitive potassium channel to trigger the release of endogenous insulin stores, or administration of exogenous insulin. Neither of these achieves accurate normalization of blood glucose levels and both carry the risk of inducing hypoglycemia. For these reasons, there has been intense interest in the development of pharmaceuticals that function in a glucose-dependent action, i.e. potentiators of glucose signaling.
  • Physiological signaling systems which function in this manner are well-characterized and include the gut peptides GLPl, GIP and PACAP. These hormones act via their cognate G-protein coupled receptor to stimulate the production of cAMP in pancreatic ⁇ -cells. The increased cAMP does not appear to result in stimulation of insulin release during the fasting or preprandial state.
  • a series of biochemical targets of cAMP signaling including the ATP-sensitive potassium channel, voltage-sensitive potassium channels and the exocytotic machinery, are modified in such a way that the insulin secretory response to a postprandial glucose stimulus is markedly enhanced. Accordingly, agonists of novel, similarly functioning, ⁇ -cell GPCRs, including GPRl 19, would also stimulate the release of endogenous insulin and consequently promote normoglycemia in type 2 diabetes.
  • cAMP for example as a result of GLP- 1 stimulation, promotes ⁇ -cell proliferation, inhibits ⁇ -cell death and thus improves islet mass.
  • This positive effect on ⁇ -cell mass is expected to be beneficial in both type 2 diabetes, where insufficient insulin is produced, and type 1 diabetes, where ⁇ -cells are destroyed by an inappropriate autoimmune response.
  • Some ⁇ -cell GPCRs, including GPRl 19, are also present in the hypothalamus where they modulate hunger, satiety, decrease food intake, controlling or decreasing weight and energy expenditure. Hence, given their function within the hypothalamic circuitry, agonists or inverse agonists of these receptors mitigate hunger, promote satiety and therefore modulate weight.
  • the metabolic-related disorder is selected from the group consisting of type 2 diabetes, hyperglycemia, hyperinsulinemia, hyperlipidemia, hypertriglyceridemia, insulin resistance, type 1 diabetes, idiopathic type 1 diabetes (type Ib), latent autoimmune diabetes in adults (LADA), early-onset type 2 diabetes (EOD), youth-onset atypical diabetes (YOAD), maturity onset diabetes of the young (MODY), malnutrition-related diabetes, gestational diabetes, cardiovascular disease, coronary heart disease, vascular restenosis, restenosis, restenosis after angioplasty, peripheral vascular disease, claudication, intermittent claudication, cell death associated with myocardial infarction (e.g.
  • ITT impaired glucose tolerance
  • impaired glucose metabolism conditions of impaired glucose metabolism, conditions of impaired fasting plasma glucose, metabolic acidosis, ketosis, arthritis, obesity, osteoporosis, hypertension, congestive heart failure, left ventricular hypertrophy, peripheral arterial disease, diabetic retinopathy, macular degeneration, cataract, diabetic nephropathy, glomerulosclerosis, chronic renal failure, diabetic neuropathy, metabolic syndrome, syndrome X, premenstrual syndrome, angina pectoris, thrombosis, atherosclerosis, ischemic stroke, transient ischemic attacks, stroke, erectile dysfunction, skin and connective tissue disorders, foot ulcerations, ulcerative colitis, endothelial dysfunction, and impaired vascular compliance.
  • COMPOSITIONS AND SALTS impaired glucose tolerance
  • compositions comprising a compound selected from N-(2-fluoro-4-(methylsulfonyl)phenyl)-5-methyl-6-(l -(5-methylpyrazin-2- yl)piperidin-4-yloxy)pyrimidin-4-amine (Compound 1), and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • a further aspect of the present invention pertains to compositions comprising a compound selected from N-(2-fluoro-4-(methylsulfonyl)phenyl)-5-methyl-6-(l-(5-methylpyrazin- 2-yl)piperidin-4-yloxy)pyrimidin-4-amine (Compound 1), and pharmaceutically acceptable salts, solvates, and hydrates thereof, and one or more pharmaceutically acceptable carriers.
  • Compound 1 N-(2-fluoro-4-(methylsulfonyl)phenyl)-5-methyl-6-(l-(5-methylpyrazin- 2-yl)piperidin-4-yloxy)pyrimidin-4-amine
  • a further aspect of the present invention pertains to pharmaceutical compositions comprising a compound selected from N-(2-fluoro-4-(methylsulfonyl)phenyl)-5-methyl-6-(l-(5- methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine (Compound 1), and pharmaceutically acceptable salts, solvates, and hydrates thereof, and one or more pharmaceutically acceptable carriers.
  • Compound 1 N-(2-fluoro-4-(methylsulfonyl)phenyl)-5-methyl-6-(l-(5- methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine
  • compositions comprising N-(2- fluoro-4-(methylsulfonyl)phenyl)-5-methyl-6-(l-(5-methylpyrazin-2-yl)piperidin-4- yloxy)pyrimidin-4-amine and a pharmaceutically acceptable carrier.
  • composition comprising admixing N-(2-fluoro-4-(methylsulfonyl)phenyl)-5- methyl-6-(l-(5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier.
  • Compositions may be prepared by any suitable method, typically by uniformly mixing the active compound with liquids or finely divided solid carriers, or both, in the required proportions, and then, if necessary, forming the resulting mixture into a desired shape.
  • Liquid preparations for oral administration may be in the form of solutions, emulsions, aqueous or oily suspensions, and syrups.
  • the oral preparations may be in the form of dry powder that can be reconstituted with water or another suitable liquid vehicle before use.
  • Parenteral dosage forms may be prepared by dissolving the compound of the invention in a suitable liquid vehicle and filter sterilizing the solution before filling and sealing an appropriate vial or ampoule. These are just a few examples of the many appropriate methods well known in the art for preparing dosage forms.
  • a compound of the present invention can be formulated into pharmaceutical
  • compositions using techniques well known to those in the art. Suitable pharmaceutically- acceptable carriers, outside those mentioned herein, are known in the art; for example, see Remington, The Science and Practice of Pharmacy, 20th Edition, 2000, Lippincott Williams & Wilkins, (Editors: Gennaro, A. R., et al.).
  • a compound of the invention can, in an alternative use, be administered as a raw or pure chemical, it is preferable however to present the compound or active ingredient as a pharmaceutical formulation or composition further comprising a pharmaceutically acceptable carrier.
  • compositions comprising a compound of the invention or a pharmaceutically acceptable salt or derivative thereof together with one or more pharmaceutically acceptable carriers thereof and/or prophylactic ingredients.
  • the carrier(s) must be "acceptable” in the sense of being compatible with the other ingredients of the composition and not overly deleterious to the recipient thereof.
  • compositions include those suitable for oral, rectal, nasal, topical (including buccal and sub-lingual), vaginal or parenteral (including intramuscular, sub-cutaneous and intravenous) administration or in a form suitable for administration by inhalation, insufflation or by a transdermal patch.
  • Transdermal patches dispense a drug at a controlled rate by presenting the drug for absorption in an efficient manner with a minimum of degradation of the drug.
  • transdermal patches comprise an impermeable backing layer, a single pressure sensitive adhesive and a removable protective layer with a release liner.
  • a desired efficacious transdermal patch based upon the needs of the artisan.
  • compositions and unit dosages thereof may thus be placed into the form of pharmaceutical compositions and unit dosages thereof, and in such form may be employed as solids, such as tablets or filled capsules, or liquids such as solutions, suspensions, emulsions, elixirs, gels or capsules filled with the same, all for oral use, in the form of suppositories for rectal administration; or in the form of sterile injectable solutions for parenteral (including subcutaneous) use.
  • Such pharmaceutical compositions and unit dosage forms thereof may comprise conventional ingredients in conventional proportions, with or without additional active compounds or principles, and such unit dosage forms may contain any suitable effective amount of the active ingredient commensurate with the intended daily dosage range to be employed.
  • the pharmaceutical composition may be in the form of, for example, a tablet, capsule, suspension or liquid.
  • the pharmaceutical composition is preferably made in the form of a dosage unit containing a particular amount of the active ingredient.
  • Examples of such dosage units are capsules, tablets, powders, granules or a suspension, with conventional additives such as lactose, mannitol, corn starch or potato starch; with binders such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins; with disintegrators such as corn starch, potato starch or sodium carboxymethyl-cellulose; and with lubricants such as talc or magnesium stearate.
  • the active ingredient may also be administered by injection as a composition wherein, for example, saline, dextrose or water may be used as a suitable pharmaceutically acceptable carrier.
  • the dose when using the compounds of the present invention can vary within wide limits, and as is customary and known to the physician, it is to be tailored to the individual conditions in each individual case. It depends, for example, on the nature and severity of the illness to be treated, on the condition of the patient, on the compound employed or on whether an acute or chronic disease state is treated or prophylaxis is conducted or on whether further active compounds are administered in addition to the compounds of the present invention.
  • doses of the present invention include, but are not limited to, about 0.001 mg to about 5000 mg, about 0.001 to about 2500 mg, about 0.001 to about 1000 mg, 0.001 to about 500 mg, 0.001 mg to about 250 mg, about 0.001 mg to 100 mg, about 0.001 mg to about 50 mg, and about 0.001 mg to about 25 mg.
  • the desired dose may conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example, as two, three, four or more sub-doses per day.
  • the sub-dose itself may be further divided, e.g., into a number of discrete loosely spaced administrations. Depending on the individual and as deemed appropriate from the patient's physician or care-giver it may be necessary to deviate upward or downward from the doses described herein.
  • a compound of the invention, or pharmaceutically acceptable salt thereof, required for use in treatment will vary not only with the particular salt selected but also with the route of administration, the nature of the condition being treated and the age and condition of the patient and will ultimately be at the discretion of the attendant physician or clinician.
  • animal models include, but are not limited to, the rodent diabetes model as described in Example 1 , infra (as well as other animal models known in the art, such as those reported by Reed and Scribner in Diabetes, Obesity and Metabolism, 1, 1999, 75-86).
  • these extrapolations may merely be based on the weight of the animal in the respective model in comparison to another, such as a mammal, preferably a human, however, more often, these extrapolations are not simply based on weights, but rather incorporate a variety of factors.
  • Representative factors include, but are not limited to, the age, weight, sex, diet and medical condition of the patient, the severity of the disease, the route of administration, pharmacological considerations such as the activity, efficacy, pharmacokinetic and toxicology profiles of the particular compound employed, whether a drug delivery system is utilized, on whether an acute or chronic disease state is being treated or prophylaxis is conducted or on whether further active compounds are administered in addition to the compounds of the present invention and as part of a drug combination.
  • the dosage regimen for treating a disease condition with the compounds and/or compositions of this invention is selected in accordance with a variety of factors as cited above.
  • the actual dosage regimen employed may vary widely and therefore may deviate from a preferred dosage regimen and one skilled in the art will recognize that a dosage and a dosage regimen outside these typical ranges can be tested and, where appropriate, may be used in the methods of this invention.
  • the compounds of the present invention can be administered in a wide variety of oral and parenteral dosage forms. It will be obvious to those skilled in the art that the following dosage forms may comprise, as the active component, either a compound of the invention or a pharmaceutically acceptable salt of a compound of the invention.
  • a suitable pharmaceutically acceptable carrier can be either solid, liquid or a mixture of both.
  • Solid form preparations include powders, tablets, pills, capsules, cachets, suppositories, and dispersible granules.
  • a solid carrier can be one or more substances which may also act as diluents, flavoring agents, solubilizers, lubricants, suspending agents, binders, preservatives, tablet disintegrating agents, or an encapsulating material.
  • the carrier is a finely divided solid which is in a mixture with the finely divided active component.
  • the active component is mixed with the carrier having the necessary binding capacity in suitable proportions and compacted to the desired shape and size.
  • the powders and tablets may contain varying percentage amounts of the active compound.
  • a representative amount in a powder or tablet may contain from 0.5 to about 90 percent of the compound of the invention; however, an artisan would know when amounts outside of this range are necessary.
  • Suitable carriers for powders and tablets are magnesium carbonate, magnesium stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, a low melting wax, cocoa butter, and the like.
  • preparation is intended to include the formulation of the active compound with encapsulating material as carrier providing a capsule in which the active component, with or without carriers, is surrounded by a carrier, which is thus in association with it.
  • carrier providing a capsule in which the active component, with or without carriers, is surrounded by a carrier, which is thus in association with it.
  • cachets and lozenges are included. Tablets, powders, capsules, pills, cachets, and lozenges can be used as solid forms suitable for oral administration.
  • a low melting wax such as an admixture of fatty acid glycerides or cocoa butter
  • the active component is dispersed homogeneously therein, as by stirring.
  • the molten homogenous mixture is then poured into convenient sized molds, allowed to cool, and thereby to solidify.
  • compositions suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or sprays containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
  • Liquid form preparations include solutions, suspensions, and emulsions, for example, water or water-propylene glycol solutions.
  • parenteral injection liquid preparations can be formulated as solutions in aqueous polyethylene glycol solution.
  • injectable preparations for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • Suitable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • the compounds according to the present invention may thus be formulated for parenteral administration (e.g. by injection, for example bolus injection or continuous infusion) and may be presented in unit dose form in ampoules, pre-filled syringes, small volume infusion or in multi- dose containers with an added preservative.
  • the pharmaceutical 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 active ingredient may be in powder form, obtained by aseptic isolation of sterile solid or by lyophilization from solution, for constitution with a suitable vehicle, e.g. sterile, pyrogen- free water, before use.
  • Aqueous compositions suitable for oral use can be prepared by dissolving or suspending the active component in water and adding suitable colorants, flavors, stabilizing and thickening agents, as desired.
  • Aqueous suspensions suitable for oral use can be made by dispersing the finely divided active component in water with viscous material, such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, or other well known suspending agents.
  • solid form preparations which are intended to be converted, shortly before use, to liquid form preparations for oral administration.
  • liquid forms include solutions, suspensions, and emulsions.
  • These preparations may contain, in addition to the active component, colorants, flavors, stabilizers, buffers, artificial and natural sweeteners, dispersants, thickeners, solubilizing agents, and the like.
  • the compounds according to the invention may be formulated as ointments, creams or lotions, or as a transdermal patch.
  • Ointments and creams may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agents.
  • Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilizing agents, dispersing agents, suspending agents, thickening agents, or coloring agents.
  • compositions suitable for topical administration in the mouth include lozenges comprising the active agent in a flavored base, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert base such as gelatin and glycerin or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
  • Solutions or suspensions are applied directly to the nasal cavity by conventional means, for example with a dropper, pipette or spray.
  • the compositions may be provided in single or multi-dose form. In the latter case of a dropper or pipette, this may be achieved by the patient administering an appropriate, predetermined volume of the solution or suspension. In the case of a spray, this may be achieved for example by means of a metering atomizing spray pump.
  • Administration to the respiratory tract may also be achieved by means of an aerosol composition in which the active ingredient is provided in a pressurized pack with a suitable propellant.
  • an aerosol composition in which the active ingredient is provided in a pressurized pack with a suitable propellant.
  • the compounds of the present invention or pharmaceutical compositions comprising them are administered as aerosols, for example as nasal aerosols or by inhalation, this can be carried out, for example, using a spray, a nebulizer, a pump nebulizer, an inhalation apparatus, a metered inhaler or a dry powder inhaler.
  • Pharmaceutical forms for administration of the compounds of the present invention as an aerosol can be prepared by processes well-known to the person skilled in the art.
  • solutions or dispersions of the compounds of the present invention in water, water/alcohol mixtures or suitable saline solutions can be employed using customary additives, for example benzyl alcohol or other suitable preservatives, absorption enhancers for increasing the bioavailability, solubilizers, dispersants and others, and, if appropriate, customary propellants, for example include carbon dioxide, CFCs, such as, dichlorodifluoromethane, trichlorofluoromethane, or dichlorotetrafluoroethane; and the like.
  • the aerosol may conveniently also contain a surfactant such as lecithin.
  • the dose of drug may be controlled by provision of a metered valve.
  • compositions intended for administration to the respiratory tract including intranasal compositions
  • the compound will generally have a small particle size for example of the order of 10 microns or less. Such a particle size may be obtained by means known in the art, for example by micronization.
  • compositions adapted to give sustained release of the active ingredient may be employed.
  • the active ingredients may be provided in the form of a dry powder, for example, a powder mix of the compound in a suitable powder base such as lactose, starch, starch derivatives such as hydroxypropylmethyl cellulose and polyvinylpyrrolidone (PVP).
  • a powder mix of the compound in a suitable powder base such as lactose, starch, starch derivatives such as hydroxypropylmethyl cellulose and polyvinylpyrrolidone (PVP).
  • the powder carrier will form a gel in the nasal cavity.
  • the powder composition may be presented in unit dose form for example in capsules or cartridges of, e.g., gelatin, or blister packs from which the powder may be administered by means of an inhaler.
  • the pharmaceutical preparations are preferably in unit dosage forms.
  • the preparation can be subdivided into unit doses containing appropriate quantities of the active component.
  • the unit dosage form can be a packaged preparation, the package containing discrete quantities of preparation, such as packeted tablets, capsules, and powders in vials or ampoules.
  • the unit dosage form can be a capsule, tablet, cachet, or lozenge itself, or it can be the appropriate number of any of these in packaged form.
  • Tablets or capsules for oral administration and liquids for intravenous administration are preferred compositions.
  • the compounds according to the invention may optionally exist as pharmaceutically acceptable salts including pharmaceutically acceptable acid addition salts prepared from pharmaceutically acceptable non-toxic acids including inorganic and organic acids.
  • the acid addition salts may be obtained as the direct product of compound synthesis.
  • the free base may be dissolved in a suitable solvent containing the appropriate acid, and the salt isolated by evaporating the solvent or otherwise separating the salt and solvent.
  • the compounds of this invention may form solvates with standard low molecular weight solvents using methods known to the skilled artisan.
  • GPRl 19 agonist and pharmaceutically acceptable salts, solvates and hydrates thereof are utilized as active ingredients in a composition, such as a pharmaceutical composition, that they are not necessarily intended for use in only humans, but in other non- human mammals as well.
  • active agents such as GPRl 19 receptor modulators
  • GPRl 19 receptor modulators for the treatment of obesity in domestic animals (e.g., cats and dogs), and GPRl 19 receptor modulators in other animals, such as livestock, where no disease or disorder is evident (e.g., food-oriented animals such as cows, chickens, fish, etc.).
  • the compounds of the present invention can be administrated in a wide variety of oral and parenteral dosage forms. It will be apparent to those skilled in the art that the following dosage forms may comprise, as the active component, a compound selected from the compound
  • one aspect of the present invention pertains to hydrates and solvates of N-(2-fluoro-4-(methylsulfonyl) phenyl)-5- methyl-6-(l -(5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine and pharmaceutical acceptable salts, that can be isolated and characterized by one or more of the methods known in the art, such as, thermogravimetric analysis (TGA), TGA-mass spectroscopy, TGA-Infrared spectroscopy, powder X-ray diffraction (XRPD), Karl Fisher titration, high resolution X-ray diffraction, and the like.
  • TGA thermogravimetric analysis
  • TGA-mass spectroscopy TGA-mass spectroscopy
  • TGA-Infrared spectroscopy powder X-ray diffraction (XRPD)
  • Karl Fisher titration high resolution X-ray diffraction, and the like.
  • Polymorphism is the ability of a substance to exist as two or more crystalline phases that have different arrangements and/or conformations of the molecules in the crystal lattice.
  • Polymorphs show the same properties in the liquid or gaseous state but they behave differently in the solid state.
  • drugs can also exist as salts and other multicomponent crystalline phases.
  • solvates and hydrates may contain an API host and either solvent or water molecules, respectively, as guests.
  • the guest compound is a solid at room temperature, the resulting form is often called a cocrystal.
  • Salts, solvates, hydrates, and cocrystals may show polymorphism as well. Crystalline phases that share the same API host, but differ with respect to their guests, may be referred to as
  • Solvates contain molecules of the solvent of crystallization in a definite crystal lattice. Solvates, in which the solvent of crystallization is water, are termed hydrates. Because water is a constituent of the atmosphere, hydrates of drugs may be formed rather easily.
  • GPRl 19 receptor related diseases can be utilized for modulating the activity of GPRl 19 receptor related diseases, conditions and/or disorders as described herein.
  • modulating the activity of GPRl 19 receptor related diseases include the treatment of metabolic related disorders.
  • Metabolic related disorders include, but are not limited to, hyperlipidemia, type 1 diabetes, type 2 diabetes, and conditions associated therewith, such as, but not limited to coronary heart disease, ischemic stroke, restenosis after angioplasty, peripheral vascular disease, claudication, intermittent claudication, cell death associated with myocardial infarction (e.g.
  • necrosis and apoptosis necrosis and apoptosis
  • dyslipidemia post-prandial lipemia
  • ITT impaired glucose tolerance
  • conditions of impaired fasting plasma glucose metabolic acidosis, ketosis, arthritis, obesity, osteoporosis, hypertension, congestive heart failure, left ventricular pressure
  • hypertrophy peripheral arterial disease, diabetic retinopathy, macular degeneration, cataract, diabetic nephropathy, glomerulosclerosis, chronic renal failure, diabetic neuropathy, metabolic syndrome, syndrome X, premenstrual syndrome, coronary heart disease, angina pectoris, thrombosis, atherosclerosis, myocardial infarction, transient ischemic attacks, stroke, vascular restenosis, hyperglycemia, hyperinsulinemia, hyperlipidemia, hypertriglyceridemia, insulin resistance, impaired glucose metabolism, conditions of impaired glucose tolerance, conditions of impaired fasting plasma glucose, obesity, erectile dysfunction, skin and connective tissue disorders, foot ulcerations, ulcerative colitis, endothelial dysfunction and impaired vascular compliance.
  • metabolic related disorders include type 1 diabetes, type 2 diabetes, inadequate glucose tolerance, insulin resistance, hyperglycemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia and syndrome X.
  • Other examples of modulating the activity of GPRl 19 receptor related diseases include the treatment of obesity and/or overweight by decreasing food intake, inducing satiation (i.e., the feeling of fullness), controlling weight gain, decreasing body weight and/or affecting metabolism such that the recipient loses weight and/or maintains weight.
  • a compound of the invention can be administered as the sole active pharmaceutical agent (i.e., mono-therapy), the compound can also be used in combination with one or more pharmaceutical agents (i.e., combination-therapy) either administered together or separately for the treatment of the diseases / conditions / disorders described herein. Therefore, another aspect of the present invention includes methods of treatment of a metabolic related disorder, including a weight related disorder, such as obesity, comprising administering to an individual in need of prophylaxis and/or treatment a therapeutically effective amount of a compound of the present invention in combination with one or more additional pharmaceutical agent as described herein.
  • a metabolic related disorder including a weight related disorder, such as obesity
  • Suitable pharmaceutical agents that can be used in combination with the compounds of the present invention include anti-obesity agents such as apolipoprotein-B secretion/microsomal triglyceride transfer protein (apo-B/MTP) inhibitors, MCR-4 agonists, cholescystokinin-A (CCK- A) agonists, serotonin and norepinephrine reuptake inhibitors (for example, sibutramine), sympathomimetic agents, ⁇ 3 adrenergic receptor agonists, dopamine agonists (for example, bromocriptine), melanocyte-stimulating hormone receptor analogues, cannabinoid 1 receptor antagonists [for example, SR141716: N-(piperidin-l-yi)-5-(4-chlorophenyl)-l-(2,4- dichlorophenyl)-4-methyl-lH-pyrazole-3-carboxamide], melanin concentrating hormone antagonists, leptons (the
  • GPP ghrelin receptor antagonists
  • histamine 3 receptor antagonists or reverse agonists neuromedin U receptor agonists
  • neuromedin U receptor agonists for example, noradrenergic anorectic agents (for example, phentermine, mazindol and the like) and appetite suppressants (for example, bupropion).
  • noradrenergic anorectic agents for example, phentermine, mazindol and the like
  • appetite suppressants for example, bupropion
  • anti-obesity agents including the agents set forth infra, are well known, or will be readily apparent in light of the instant disclosure, to one of ordinary skill in the art.
  • the anti-obesity agents are selected from the group consisting of orlistat, sibutramine, bromocriptine, ephedrine, leptin, and pseudoephedrine.
  • compounds of the present invention and combination therapies are administered in conjunction with exercise and/or a sensible diet.
  • combination-therapy of the compounds of the present invention with other anti-obesity agents, anorectic agents, appetite suppressant and related agents is not limited to those listed above, but includes in principle any combination with any pharmaceutical agent or pharmaceutical composition useful for the treatment of overweight and obese individuals.
  • combination-therapy of the compounds of the present invention with other pharmaceutical agents is not limited to those listed herein, supra or infra, but includes in principle any combination with any pharmaceutical agent or pharmaceutical composition useful for the treatment of diseases, conditions or disorders that are linked to metabolic related disorders.
  • Some embodiments of the present invention include methods of treatment of a disease, disorder, condition or complication thereof as described herein, comprising administering to an individual in need of such treatment a therapeutically effective amount or dose of a compound of the present invention in combination with at least one pharmaceutical agent selected from the group consisting of: sulfonylureas (for example, glyburide, glipizide, glimepiride and other sulfonylureas known in the art), meglitinides (for example, repaglinide, nateglinide and other meglitinides known in the art), biguanides (for example, biguanides include phenformin, metformin, buformin, and biguanides known in the art), ⁇ -glucosidase inhibitors [for example, acarbose, N-(l,3-dihydroxy-2-propyl)valiolamine (generic name; voglibose), miglitol, and ⁇
  • PPAR- ⁇ ) agonists for example, rosiglitazone, pioglitazone, tesaglitazar, netoglitazone, GW- 409544, GW-501516 and PPAR- ⁇ agonists known in the art
  • insulin insulin analogues
  • HMG- CoA reductase inhibitors for example, rosuvastatin, pravastatin and its sodium salt, simvastatin, lovastatin, atorvastatin, fluvastatin, cerivastatin, rosuvastatin, pitavastatin, BMS's "superstatin", and HMG-CoA reductase inhibitors known in the art
  • cholesterol-lowering drugs for example, fibrates that include: bezafibrate, beclobrate, binifibrate, ciplofibrate, clinofibrate, clofibrate, clofibric acid, etofibrate, fenofibrate,
  • Suitable pharmaceutical agents that can be used in conjunction with compounds of the present invention include, but not limited to, amylin agonists (for example, pramlintide), insulin secretagogues (for example, GLP-I agonists; exendin-4; insulinotropin (NN2211); acyl CoA cholesterol acetyltransferase inhibitors (for example, ezetimibe, eflucimibe, and like compounds), cholesterol absorption inhibitors (for example, ezetimibe, pamaqueside and like compounds), cholesterol ester transfer protein inhibitors (for example, CP-529414, JTT-705, CETi-I, and like compounds), microsomal triglyceride transfer protein inhibitors (for example, implitapide, and like compounds), cholesterol modulators (for example, NO- 1886, and like compounds), bile acid modulators (for example, GT103-279 and like compounds), insulin signaling pathway modulators, like inhibitors of protein tyrosine phosphatases (PTPases
  • PPCK phosphoenolpyruvate carboxykinase
  • PDHK pyruvate dehydrogenase kinase
  • insulin sensitivity enhancers insulin secretion enhancers
  • inhibitors of gastric emptying ⁇ 2 -adrenergic antagonists
  • PvXR retinoid X receptor
  • DPP-IV dipeptidyl peptidase-4
  • Dipeptidyl peptidase IV (DPP-IV, EC 3.4.14.5) exhibits catalytic activity against a broad range of peptide substrates that includes peptide hormones, neuropeptides, and chemokines.
  • GIP which stimulate glucose-dependent insulin secretion and otherwise promote blood glucose homeostasis, are rapidly cleaved by DPP-IV at the position 2 alanine leading to inactivation of their biological activity.
  • DPP-IV activity is associated with enhanced incretin action, increased insulin, and lower blood glucose in vivo. Genetic attenuation of DPP-IV activity has been shown to provide resistance to obesity and to improve insulin sensitivity.
  • Inhibitors of DPP-IV have shown to be useful as theraputics, for example, oral administration of vildagliptin (l-[2-(3-hydroxyadamant-l- ylamino)acetyl]pyrrolidine-2(S)-carbonitrile) or sitagliptin (3(R)-ammo-l-[3-(trifluoromethyl)- 5,6,7,8-tetrahydro[l,2,4]triazolo[4,3-a]pyrazin-7-yl]-4-(2,4,5-trifluorophenyl) butan-1-one) to human patients suffering with type 2 diabetes has been found to reduce fasting glucose and postprandial glucose excursion in association with significantly reduced HbAi c levels.
  • suitable pharmaceutical agents include inhibitors of DPP-IV that can be used in conjunction with compounds of the present invention either dosed separately or together.
  • Inhibitors of DPP-IV are well in the art or can be readily identified and their in vitro biological activity determined using any number of methods available, for example, O'Brien, M., Daily, B., Schurria, M., "Assay for DPPIV activity using a homogeneous, luminescent method," Cell Notes, Issue 11, 2005; see also the DPPIV-GloTM Protease Assay Technical Bulletin #TB339.
  • DPP-IV inhibitors are described in Villhauer et al., J Med Chem (2003)
  • DPP-IV inhibitors include, but are not limited to, dipeptide derivatives or dipeptide mimetics such as alanine-pyrrolidide, isoleucine-thiazolidide, and the pseudosubstrate N-valyl prolyl, O-benzoyl hydroxylamine, as described e.g. in U.S. Pat. No. 6,303,661.
  • DPP-IV inhibitors selected from the DPP-IV inhibitors found in U.S. Pat. Nos. 6,869,947, 6,867,205, 6,861,440, 6,849,622, 6,812,350, 6,803,357, 6,800,650, 6,727,261, 6,716,843, 6,710,040, 6,706,742, 6,645,995, 6,617,340, 6,699,871, 6,573,287, 6,432,969, 6,395,767, 6,380,398, 6,303,661, 6,242,422, 6,166,063, 6,100,234, and 6,040,145.
  • DPP-IV inhibitors selected from the DPP-IV inhibitors found in U.S. Patent Application
  • DPP-IV inhibitors selected from the DPP-IV inhibitors found in International Patent Application Publication Numbers: WO 2005/087235, WO 2005/082348, WO 2005/082849, WO
  • Some embodiments of the present invention include every combination of one or more DPP-IV inhibitors selected from the DPP-IV inhibitors found in Patent Publication Numbers: EP 1517907, EP 1513808, EP 1492777, EP 1490335, EP 1489088, EP 1480961, EP 1476435, EP 1476429, EP 1469873, EP 1465891, EP 1463727, EP 1461337, EP 1450794, EP 1446116, EP
  • the DPP-IV inhibitor has an IC 50 of less than about 10 ⁇ M, less than about 1 ⁇ M, less than about 100 nM, less than about 75 nM, less than about 50 nM, less than about 25 nM, less than about 20 nM, less than about 15 nM, less than about 10 nM, less than about 5 nM, less than about 4 nM, less than about 3 nM, less than about 2 nM, or less than about
  • the DPP-IV inhibitor has an IC 5 O of less than about 50 nM, less than about 25 nM, less than about 20 nM, less than about 15 nM, less than about 10 nM, less than about 5 nM, less than about 4 nM, less than about 3 nM, less than about 2 nM, or less than about I nM.
  • the DPP-IV inhibitor is a selective DPP-IV inhibitor, wherein the selective DPP-IV inhibitor has a selectivity for human plasma DPP-IV over one or more of the following enzymes, post proline cleaving enzyme (PPCE), dipeptidyl peptidase II (DPPII), dipeptidyl peptidase-8 (DPP-8), and dipeptidyl peptidase-9 (DPP-9) of at least about 10-fold, more preferably of at least about 100-fold, and most preferably of at least about 1000-fold.
  • PPCE post proline cleaving enzyme
  • DPPII dipeptidyl peptidase II
  • DPP-8 dipeptidyl peptidase-8
  • DPP-9 dipeptidyl peptidase-9
  • the DPP-IV inhibitor is orally active.
  • the DPP-IV inhibitor is an inhibitor of human DPP-IV.
  • Some embodiments of the present invention include every combination of one or more compounds selected from: 3(R)-amino-l-[3-(trifluoromethyl)-5,6,7,8-tetrahydro[l,2,4]triazolo[4,3-a]pyrazin-7-yl]- 4-(2,4,5-trifluorophenyl) butan- 1 -one; 1 -[2-(3-hydroxyadamant- 1 -ylamino)acetyl]pyrrolidine- 2(S)-carbonitrile; (lS,3S,5S)-2-[2(S)-amino-2-(3-hydroxyadamantan-l-yl)acetyl]-2- azabicyclo[3.1.0]hexane-3-carbonitrile; 2-[6-[3(R)-aminopiperidin-l-yl]-3-methyl-2,4-dioxo- l,2,3,4-tetrahydropyrimidin-l-ylmethyl
  • Some embodiments of the present invention include every combination of one or more compounds selected from: (2S,4S)-l-[(2S)-2-amino-3,3-bis(4-fluorophenyl)propanoyl]-4- fluoropyrrolidine-2-carbonitrile; (2S,5R)-5-ethynyl- 1 - ⁇ N-(4-methyl- 1 -(4-carboxy-pyridin-2- yl)piperidin-4-yl)glycyl ⁇ pyrrolidine-2-carbonitrile; and ( ⁇ S,6R)-?>- ⁇ [3-(trifluoromethyl)-5,6- dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl]carbonyl ⁇ -6-(2,4,5-trifluorophenyl)cyclohex-3-en- 1 -amine; and pharmaceutically acceptable salts, solvates and hydrates thereof.
  • Sitagliptin phosphate (Januvia, MK-0431, dihydrogenphosphate salt of 3(R)-amino-l-[3- (trifluoromethyl)-5,6,7,8-tetrahydro[l,2,4]triazolo[4,3-a]pyrazin-7-yl]-4-(2,4,5-trifluorophenyl) butan-1-one) is marketed by Merck & Co. for once-daily oral treatment of type 2 diabetes.
  • the DPP-IV inhibitor of the present invention is selected from 3(R)-amino-l-[3-(trifluoromethyl)-5,6,7,8-tetrahydro[l,2,4]triazolo[4,3-a]pyrazin- 7-yl]-4-(2,4,5-trifluorophenyl) butan-1-one, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the dipeptidyl peptidase IV (DPP4) inhibitor is 3(R)-amino-l-[3- (trifluoromethyl)-5,6,7,8-tetrahydro[l,2,4]triazolo[4,3-a]pyrazin-7-yl]-4-(2,4,5-trifluorophenyl) butan-1-one phosphate:
  • the dipeptidyl peptidase IV (DPP4) inhibitor is crystalline 3(R)-amino-l-[3-(trifluoromethyl)-5,6,7,8-tetrahydro[l,2,4]triazolo[4,3-a]pyrazin-7- yl]-4-(2,4,5-trifluorophenyl) butan-1-one phosphate monohydrate.
  • Vildagliptin (Galvus, LAF-237, l-[2-(3-hydroxyadamant-l-ylamino)acetyl]pyrrolidine- 2( 1 S)-carbonitrile) is another dipeptidyl-peptidase IV (DPP-IV) inhibitor and was first commercialized in Brazil and Mexico by Novartis for oral, once-daily treatment of type 2 diabetes.
  • DPP-IV dipeptidyl-peptidase IV
  • MAA marketing authorization application
  • the compound, l-[2-(3-hydroxyadamant-l-ylamino)acetyl]pyrrolidine-2(S)-carbonitrile, is disclosed in international patent publication WO2000/034241.
  • Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in WO2000/034241 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • the DPP-IV inhibitor of the present invention is selected from 1- [2-(3-hydroxyadamant- 1 -ylamino)acetyl]pyrrolidine-2(S)-carbonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the DPP-IV inhibitor is l-[2-(3-hydroxyadamant-l-ylamino)acetyl]pyrrolidine-
  • Saxagliptin (Onglyza, BMS-477118, (l£,3£,5S)-2-[2(S)-amino-2-(3-hydroxyadamantan- l-yl)acetyl]-2-azabicyclo[3.1.0]hexane-3-carbonitrile) is another DPP-IV inhibitor and was launched in 2009 by AstraZeneca and Bristol-Myers Squibb in the U.S. for the treatment of type
  • the DPP-IV inhibitor of the present invention is selected from (lS,3S,5S)-2-[2(S)-amino-2-(3-hydroxyadamantan-l-yl)acetyl]-2-azabicyclo[3.1.0]hexane-3- carbonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • SYR-322 (Alogliptin, 2-[6-[3(R)-aminopiperidin-l-yl]-3-methyl-2,4-dioxo-l,2,3,4- tetrahydropyrimidin- 1 -ylmethyl]benzonitrile) is a DPP-IV inhibitor which regulatory approval has been filed in Japan and the U.S. by Takeda for the once-daily, oral treatment of type 2 diabetes.
  • the compound, 2-[6-[3(R)-ammopiperidin-l-yi]-3-methyl-2,4-dioxo-l,2,3,4- tetrahydropyrimidin-l-ylmethyl]benzonitrile, and pharmaceutically acceptable salts thereof are disclosed in international patent publication WO 2005/095381.
  • Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in WO 2005/095381 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • the DPP-IV inhibitor of the present invention is selected from 2-[6-[3(R)-aminopiperidin- 1 -yl]-3-methyl-2,4-dioxo- 1 ,2,3,4-tetrahydropyrimidin- 1 - ylmethyljbenzonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the DPP-IV inhibitor is 2-[6-[3(R)-aminopiperidin- 1 -yl]-3-methyl-2,4-dioxo- 1,2,3,4-tetrahydropyrimidin- 1 - ylmethyljbenzonitrile benzoate:
  • BI- 1356 (Ondero, 8-[3(R)-aminopiperidin- 1 -yl]-7-(2-butynyl)-3-methyl- 1 -(4- methylquinazolin-2-ylmethyl)xanthine) is a DPP-IV inhibitor and is in phase III clinical development at Boehringer Ingelheim to evaluate its potential as an add-on therapy to metformin for the treatment of type 2 diabetes.
  • the compound, 8-[3(R)-aminopiperidin-l-yl]-7-(2-butynyl)- 3 -methyl- l-(4-methylquinazolin-2-ylmethyl)xanthine is disclosed in international patent publication WO2004/018468.
  • Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in WO2004/018468 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • the DPP-IV inhibitor of the present invention is selected from 8-[3(R)- aminopiperidin- 1 -yl]-7-(2-butynyl)-3-methyl- 1 -(4-methylquinazolin-2-ylmethyl)xanthine, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the DPP-IV inhibitor is a crystalline form of 8-[3(R)- aminopiperidin- 1 -yl]-7-(2-butynyl)-3-methyl- 1 -(4-methylquinazolin-2-ylmethyl)xanthine.
  • PHX-1149 (Dutogliptin, l-[N-[3(R)-pyrrolidinyl]glycyl]pyrrolidin-2(R)-yl boronic acid) is a DPP-IV inhibitor in phase III clinical trials by Phenomix and Forest for the oral, once-daily treatment of type 2 diabetes.
  • the compound, l-[N-[3(R)-pyrrolidinyl]glycyl] pyrrolidin-2(R)-yl boronic acid, and pharmaceutically acceptable salts are disclosed in international patent publication WO2005/047297.
  • Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in WO2005/047297 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • the DPP-IV inhibitor of the present invention is selected from 1-[N- [3 (R)- pyrrolidinyl]glycyl]pyrrolidin-2(R)-yl boronic acid, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the DPP-IV inhibitor is l-[N-[3(R)-pyrrolidinyl]glycyl]pyrrolidin-2(R)-yl boronic acid tartrate: GRC-8200 (Melogliptin, 4(5)-fluoro-l-[2-[(l ⁇ ,35)-3-(lH-l,2,4-triazol-l- ylmethyl)cyclopentylammo]acetyl]pyrrolidine-2(S)-carbonitrile) is a DPP-IV inhibitor and is currently undergoing phase II clinical trials by Glenmark Pharmaceuticals and Merck KGaA for the treatment of type 2 diabetes.
  • the compound, 4(5)-fluoro-l-[2-[(lR,35)-3-(lH-l,2,4-triazol-l- ylmethyl)cyclopentylammo]acetyl]pyrrolidine-2(S)-carbonitrile, is disclosed in international patent publication WO2006/040625.
  • Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in WO2006/040625 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • the DPP-IV inhibitor of the present invention is selected from 4(S)-fluoro-l-[2- [(lR,3S)-3-(lH-l,2,4-triazol-l-ylmethyl)cyclopentylamino]acetyl]pyrrolidine-2(S)-carbonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • R- 1579 (Carmegliptin, l-[(2S,3S,l lbS)-2-amino-9,10-dimethoxy-2,3,4,6,7,l Ib- hexahydro-lH-pyrido[2,l-a]isoquinolin-3-yl]-4(S)-(fluoromethyl)pyrrolidin-2-one) is a DPP-IV inhibitor.
  • the compound, l-[(2S,3S,l lb£)-2-amino-9,10-dimethoxy-2,3,4,6,7,l lb-hexahydro-lH- pyrido[2,l-a]isoqumoh ' n-3-yl]-4(S)-(fluoromethyl)pyrroh ' din-2-one, is disclosed in international patent publication WO2005/000848.
  • Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in WO2005/000848 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • the DPP-IV inhibitor of the present invention is selected from l-[(2S,3S,l lbS)-2- amino-9,10-dimethoxy-2,3,4,6,7,l lb-hexahydro-lH-pyrido[2,l-a]isoquinolin-3-yl]-4(S)- (fluoromethyl)pyrrolidin-2-one, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the DPP- IV inhibitor of the present invention is selected from (2S,4S)-2-cyano-4-fluoro-l-[(2-hydroxy- l,l-dimethyl)ethylamino]acetylpyrrolidine, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • Sanofi-Aventis disclosed a series of substituted bicyclic 8-pyrrolidineoxanthine derivatives as DPP-IV inhibitors in US publication US 2007/0167468 (incorporated by reference in its entirety). Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in US publication 2007/0167468 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • the DPP-IV inhibitor of the present invention is selected from 8-(cw-hexahydro-pyrrolo[3,2-b]pyrrol- 1 -yl)-3-methyl-7-(3-methyl-but-2-enyl)- 1 -(2-oxo-2-phenylethyl)-3,7-dihydro-purine-2,6-dione, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • Pfizer disclosed a series of 3-amino-pyrrolidine-4-lactam derivatives as DPP-IV inhibitors in international patent publication WO2007/148185.
  • Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in WO2007/148185 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • One such compound is l-((3S,4S)-4-ammo-l-(4-(3,3-difluoropyrroridin-l-yl)- l,3,5-triazin-2-yl)pyrrolidin-3-yl)-5,5difluoropiperidin-2-one, and pharmaceutically acceptable salts thereof.
  • the DPP-IV inhibitor of the present invention is selected from 1 -((3S,4S)-4-amino- 1 -(4-(3,3-difluoropyrrolidin- 1 -yl)- 1 ,3,5-triazin-2-yl)pyrrolidin-3-yl)- 5,5difluoropiperidin-2-one, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • Syrrx disclosed a series of substituted pyrimidine-2,4(lH,3H)-dione derivatives as DPP- IV inhibitors in international patent publication WO2005/095381.
  • Some embodiments of the present invention include every combination of one or more compounds selected from the DPP- IV inhibitors disclosed in WO2005/095381 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • One such compound is (R)-2-((6-(3-aminopiperidm-l-yl)-3-methyl-2,4-dioxo- 3,4-dihydropyrimidin-l(2H)-yl)methyl)-4-fluorobenzonitrile, and pharmaceutically acceptable salts thereof.
  • the DPP-IV inhibitor of the present invention is selected from (R)-2-((6-(3-aminopiperidin- 1 -yl)-3-methyl-2,4-dioxo-3,4-dihydropyrimidin- 1 (2H)- yl)methyl)-4-fluorobenzonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • One embodiment of the present invention pertains to any one or more crystalline forms of (R)-2-((6-(3-aminopiperidin-l-yl)-3-methyl-2,4- dioxo-3,4-dihydropyrimidin-l(2H)-yl)methyl)-4-fluorobenzonitrile succinic acid salt as described in international patent publication WO2005/0031335.
  • the dipeptidyl peptidase IV (DPP4) inhibitor is crystalline (R)-2-((6-(3-ammopiperidin-l-yl)-3-methyl-2,4- dioxo-3,4-dihydropyrimidin-l(2H)-yl)methyl)-4-fluorobenzonitrile succinic acid salt:
  • One such compound is 5- ⁇ (S)-2-[2-((S)-2-cyano-pyrrolid in-l-yl)-2-oxo- ethylamino] -propyl ⁇ -5-(lH-tetrazol-5-yl) 10, 11 -dihydro-5H-dibenzo[a,d]cycloheptene-2,8- dicarboxylic acid bis-dimethylamide, and pharmaceutically acceptable salts thereof.
  • the DPP-IV inhibitor of the present invention is selected from 5- ⁇ (S)-2-[2-((S)-2- cyano-pyrrolid in- 1 -yl)-2-oxo-ethylamino]-propyl ⁇ -5-(lH-tetrazol-5-yl) 10, 11 -dihydro-5H- dibenzo[a,d]cycloheptene-2,8-dicarboxylic acid bis-dimethylamide, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • Some embodiments of the present invention include every combination of one or more compounds selected from the DPP- IV inhibitors disclosed in WO2002/0014271 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • One such compound is ((2S,4S)-4-(4-(3-methyl- 1 -phenyl- lH-pyrazol-5- yl)piperazin-l-yl)pyrrolidin-2-yl)(thiazolidin-3-yl)methanone, and pharmaceutically acceptable salts thereof.
  • the DPP-IV inhibitor of the present invention is selected from ((2S,4S)-4-(4-(3-methyl- 1 -phenyl- lH-pyrazol-5-yl)piperazin- 1 -yl)pyrrolidin-2- yl)(thiazolidin-3-yl)methanone, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • One embodiment of the present invention pertains to any one or more of the crystalline forms of ((2S, 4S)-4-(4-(3 -methyl- 1 -phenyl- lH-pyrazol-5-yl)piperazin- 1 -yl)pyrrolidin-2-yl)(thiazolidin-3- yl)methanone salt described in international patent publication WO2006/088129 and US publication 2009/0216016.
  • the DPP-IV inhibitor is crystalline ((2S,4S)-4- (4-(3 -methyl- 1 -phenyl- lH-pyrazol-5-yl)piperazin-l-yl)pyrrolidin-2-yl)(thiazolidin-3- yl)methanone 2.5 hydrobromide salt:
  • the dipeptidyl peptidase IV (DPP4) inhibitor is crystalline ((2S,45)-4-(4-(3-methyl-l-phenyl-lH-pyrazol-5-yl)piperazin-l- yl)pyrrolidin-2-yl)(thiazolidin-3-yl)methanone di-hydrobromide salt.
  • Kyorin disclosed a series of pyrrolidinecarbonitrile derivatives as DPP-IV inhibitors in international patent publication WO2008/114857 and US publication 2008/0146818. Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in WO2008/114857 and US publication
  • the DPP-IV inhibitor of the present invention is selected from (2S,4S)-l-[2-[(4- ethoxycarbonylbicyclo[2.2.2]oct- 1 -yl)amino]acetyl]-4-fluoropyrrolidine-2-carbonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • Dainippon Sumitomo disclosed a series of bicyclic pyrrole derivatives as DPP-IV inhibitors in international patent publication WO2006/068163 and US publication 2009/0192129.
  • Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in WO2006/068163 and US publication 2009/0192129 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • One such compound is 6-[(3R)-3-amino-piperidin-l-yl]-5-(2-chloro-5-fluoro-benzyl)-l,3- dimethyl-l,5dihydro-pyrrolo[3,2-d]pyrimidine-2,4-dione, and pharmaceutically acceptable salts thereof.
  • the DPP-IV inhibitor of the present invention is selected from (6- [(3R)-3-amino-piperidin- l-yl]-5-(2-chloro-5-fluoro-benzyl)- 1 ,3-dimethyl- 1 ,5dihydro-pyrrolo[3,2- d]pyrimidine-2,4-dione, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the DPP-IV inhibitor of the present invention is selected from 2-( ⁇ 6-[(3R)-3-amino-3-methylpiperidin- 1 -yl]- 1 ,3-dimethyl-2,4-dioxo- 1 ,2,3,4-tetrahydro- 5H-pyrrolo[3,2-d]pyrimidin-5-yl ⁇ methyl)-4-fluorobenzonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • Some embodiments of the present invention include every combination of one or more compounds selected from the DPP- IV inhibitors disclosed in WO 03/037327 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • One such compound is (2S)-l- ⁇ [2-(5-methyl-2-phenyl-oxazol-4-yl)- ethylamino]-acetyl ⁇ -pyrrolidine-2-carbonitrile, and pharmaceutically acceptable salts thereof.
  • the DPP-IV inhibitor of the present invention is selected from (2S)-I- ⁇ [2-(5- methyl-2-phenyl-oxazol-4-yl)-ethylamino]-acetyl ⁇ -pyrrolidine-2-carbonitrile, and
  • the DPP-IV inhibitor of the present invention is (2S)-l- ⁇ [2-(5-methyl-2-phenyl-oxazol-4-yl)-ethylamino]-acetyl ⁇ -pyrrolidine-2- carbonitrile methansulfonic acid salt (i.e., mesylate):
  • the DPP-IV inhibitor of the present invention is selected from (2S)- 1 - ⁇ [ 1 , 1 -dimethyl-3-(4-pyridin-3-yl-imidazol- 1 -yl)-propylamino]-acetyl ⁇ - pyrrolidine-2-carbonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the DPP-IV inhibitor of the present invention is (2S)- 1- ⁇ [1,1- dimethyl-3-(4-pyridin-3-yl-imidazol- 1 -yl)-propylamino]-acetyl ⁇ -pyrrolidine-2-carbonitrile methansulfonic acid:
  • the DPP-IV inhibitor of the present invention is (2S)-l- ⁇ [l,l-dimethyl-3-(4- pyridin-3-yl-imidazol-l-yl)-propylamino]-acetyl ⁇ -pyrrolidine-2-carbonitrile fumaric acid salt (i.e., fumarate):
  • Pfizer disclosed a series of proline derivatives as DPP-IV inhibitors in international patent publication WO2005/116014 (incorporated by reference in its entirety).
  • embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in WO2005/116014 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • One such compound is (3,3-difluoropyrrolidin-l- yl)-((2S,4S)-4-(4-(pyrimidin-2yl)piperazin- 1 -yl)pyrrolidin-2-yl)methanone, and pharmaceutically acceptable salts thereof.
  • the DPP-IV inhibitor of the present invention is selected from (3,3-difluoropyrrolidin- 1 -yl)-((2S,4S)-4-(4-(pyrimidin-2yl)piperazin- 1 - yl)pyrrolidin-2-yl)methanone, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • Glaxo SmithKline disclosed a series of fluoropyrrolidine derivatives as DPP-IV inhibitors in international patent publication WO 03/002531.
  • Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in WO 03/037327 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • One such compound is (2S,4S)-l-[(2S)-2-amino-3,3-bis(4-fluorophenyl)propanoyi]-4- fluoropyrrolidine-2-carbonitrile (Denagliptin).
  • the DPP-IV inhibitor of the present invention is selected from (2S,4S)-l-[(2S)-2-amino-3,3-bis(4-fluorophenyl)propanoyl]-4- fluoropyrrolidine-2-carbonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • One salt disclosed is (25 1 ,45)-l-[(25)-2-amino-3,3-bis(4- fluorophenyl)propanoyl]-4-fluoropyrrolidine-2-carbonitrile/>-toluenesulfonic acid salt (also referred to as (2S,45)-4-fluoro- 1 -[4-fluoro- ⁇ -(4-fluorophenyl)-L-phenylalanyl]-2- pyrrolidinecarbonitrile/>-toluenesulfonic acid salt, or Denagliptin tosylate).
  • (2S,45)-4-fluoro- 1 -[4-fluoro- ⁇ -(4-fluorophenyl)-L-phenylalanyl]-2- pyrrolidinecarbonitrile/>-toluenesulfonic acid salt or Denagliptin tosylate.
  • the DPP-IV inhibitor of the present invention is (2S,4S)-l-[(2S)-2-amino-3,3-bis(4- fluorophenyl)propanoyl]-4-fluoropyrrolidine-2-carbonitrile/>-toluenesulfonic acid salt:
  • Abbott disclosed a series of substituted pyrrolidinyl derivatives as DPP-IV inhibitors in international patent publication WO 2004/026822.
  • Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in WO 2004/026822 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • One such compound is (2S,5R)-5-ethynyl-l- ⁇ N-(4-methyl-l-(4-carboxy-pyridin-2- yl)piperidin-4-yl)glycyl ⁇ pyrrolidine-2-carbonitrile.
  • the DPP-IV inhibitor of the present invention is selected from (2S,5R)-5-ethynyl-l- ⁇ N-(4-methyl-l-(4-carboxy-pyridin- 2-yl)piperidin-4-yl)glycyl ⁇ pyrrolidine-2-carbonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • Abbott has further disclosed a series of substituted cyclohexanyl/cyclohexenyl derivatives as DPP-IV inhibitors in international patent publication WO 2007/027651.
  • Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in WO 2007/027651 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • One such compound is (15,6R)-3- ⁇ [3- (trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl]carbonyl ⁇ -6-(2,4,5- trifluorophenyl)cyclohex-3-en-l -amine.
  • the DPP-IV inhibitor of the present invention is selected from (lS,6R)-3- ⁇ [3-(trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3- a]pyrazin-7(8H)-yl]carbonyl ⁇ -6-(2,4,5-trifluorophenyl)cyclohex-3-en- 1 -amine, and
  • the combination can be used by mixing the respective active components, a compound of the present invention and pharmaceutical agent, either all together or independently with a physiologically acceptable carrier, excipient, binder, diluent, etc., as described herein above, and administering the mixture or mixtures either orally or non-orally as a pharmaceutical composition.
  • a compound or a mixture of compounds of the present invention are administered as a combination therapy with another active compound the therapeutic agents can be formulated as separate pharmaceutical compositions given at the same time or at different times; or the compound or a mixture of compounds of the present invention and the therapeutic agent(s) can be formulated together as a single unit dosage.
  • Another object of the present invention relates to radio-labeled compounds that are useful not only in radio-imaging but also in assays, both in vitro and in vivo, for localizing and quantitating the GPRl 19 receptor in tissue samples, including human, and for identifying GPRl 19 receptor ligands by inhibition binding of a radio-labeled compound. It is a further object of this invention to develop novel GPRl 19 receptor assays of which comprise such radio-labeled compounds.
  • Compounds of the invention can also include all isotopes of atoms occurring in the intermediates and/or the final compound and pharmaceutically acceptable salts, solvates and hydrates thereof.
  • Isotopes include those atoms having the same atomic number but different mass numbers.
  • An isotopically-labeled or radio-labeled compound is one which is identical to compounds disclosed herein, but for the fact that one or more atoms is replaced or substituted by an atom having an atomic mass or mass number different from the atomic mass or mass number typically found in nature (i.e., naturally occurring).
  • Suitable radionuclides that may be incorporated in compounds of the present invention include but are not limited to H (also written as D for deuterium), 3 H (also written as T for tritium), 11 C, 13 C, 14 C, 13 N, 15 N, 15 0, 17 0, 18 0, 18 F, 35 S, 36 Cl, 82 Br, 75 Br, 76 Br, 77 Br, 123 1, 124 1, 125 I and 131 I.
  • the radionuclide that is incorporated in the instant radio-labeled compounds will depend on the specific application for which the radio- labeled compound will be used.
  • a radio-labeled or labeled compound is a compound of the present invention that has incorporated at least one radionuclide; in some embodiments the radionuclide is selected from the group consisting of 3 H, 14 C, 125 1, 35 S, and 82 Br.
  • isotopically-labeled compounds of the present invention are useful in compound and/or substrate tissue distribution assays.
  • the radionuclide 3 H and/or 14 C isotopes are useful in these studies.
  • substitution with heavier isotopes such as deuterium (i.e., 2 H) may afford certain therapeutic advantages resulting from greater metabolic stability
  • Isotopically labeled compounds of the present invention can generally be prepared by following procedures analogous to those disclosed in the Schemes supra and Examples infra, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent. Other synthetic methods that are useful are discussed infra. Moreover, it should be understood that all of the atoms represented in the compounds of the invention can be either the most commonly occurring isotope of such atoms or a more scarce radio-isotope or nonradioactive isotope.
  • Synthetic methods for incorporating radio-isotopes into organic compounds are applicable to compounds of the invention and are well known in the art, for example, tritium gas exposure labeling. This procedure involves exposing precursors containing exchangeable protons to tritium gas in the presence of a suitable catalyst.
  • a radio-labeled GPRl 19 receptor compound of present invention can be used in a screening assay to identify/evaluate compounds.
  • a newly synthesized or identified compound i.e., test compound
  • the ability of a test compound to compete with the radio-labeled compound of the present invention for the binding to the GPRl 19 receptor directly correlates to its binding affinity.
  • the labeled compounds of the present invention bind to the GPRl 19 receptor.
  • the labeled compound has an IC 5 O less than about 500 ⁇ M, in another embodiment the labeled compound has an IC 50 less than about 100 ⁇ M, in yet another embodiment the labeled compound has an IC 50 less than about 10 ⁇ M, in yet another embodiment the labeled compound has an IC 50 less than about 1 ⁇ M, in still yet another embodiment the labeled inhibitor has an IC 50 less than about 0.1 ⁇ M, in still yet another embodiment the labeled inhibitor has an IC 50 less than about 0.01 ⁇ M, and in still yet another embodiment the labeled inhibitor has an IC 50 less than about 0.005 ⁇ M.
  • Example 1 Primary Pharmacology for ⁇ l-fluoro- ⁇ methylsulfonylJphenylJ-S-methyl- ⁇ - (l-(5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine (Compound 1) - In Vitro Assays.
  • GDIR Glucose-Dependent Insulino tropic Receptor
  • CHO cells expressing human GDIR (AGDIR) and rat GDIR (rGDIR) were used. CHO cells were cultured in DMEM/F12K media containing pen/strep, 10% FBS and the appropriate antibiotic for selection (500 ⁇ g/mL G418 for rat and 3 ⁇ g/mL puromycin human clone cells). To establish assay conditions, 100 ⁇ L of cells were seeded at 5 x 105 cells/mL (2 x 105 cells/mL) into a lysine-coated 96 well culture plates and allowed to recover for 24 hours. After a 24-hour attachment and growth period, media was replaced with DMEM/F12 containing pen/strep, and 250 ⁇ M isobutylmethylxanthine (IBMX, phosphdiesterase inhibitor).
  • IBMX isobutylmethylxanthine
  • GluTag cells in growth medium (DMEM GIBCO Cat# 11995-065, 10% FBS GIBCO Cat# 16000-044) were seeded in a 96-well plate at 90,000 cells/well and incubated in a CO 2 incubator at 37 0 C for 24 hours.
  • the growth medium was removed by aspiration and 200 ⁇ L of assay medium (DMEM with 5 mM glucose and 0.1% BSA) was added to each well. After incubating at 37 0 C for 30 min, the assay medium was removed and each well was replenished with 175 ⁇ L of fresh assay medium with a DPP-IV inhibitor. Next, 25 ⁇ L of challenge solution was added to each well and the reaction was incubated for 2 hours at 37 0 C.
  • assay medium DMEM with 5 mM glucose and 0.1% BSA
  • Example 2 In vivo Effects of a Single Dose of ⁇ '-(2-fluoro-4-(methylsulfonyl)phenyl)-5- methyl-6-(l-(5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine on Oral Glucose Tolerance Test (oGTT) in Male SD Rats (PK/PD study) at 0.3 and 3 mg/kg.
  • the oral glucose tolerance test measures the ability of an animal to metabolize a bolus of exogenous glucose. A decrease in glucose AUC excursion from baseline indicates better glucose control.
  • Male SD rats (6 - 7 weeks of age) were housed one rat per cage in a temperature- controlled room with 12-hour light/dark cycle. They were allowed ad libitum access to water and food. Rats were fasted overnight before the study. These rats were first dosed with vehicle (0.5%
  • HPMC HPMC
  • Compound 1 at 0.3, or 3 mg/kg doses via oral gavage at 8:30 am.
  • rats were administered glucose (2 g/kg, using 50% G) by oral gavage and the tail blood samples were collected to measure blood glucose and plasma insulin at 0, 30, 60, and 120 min.
  • a separate group of rats were dosed in similar fashion and blood samples collected at 1, 2, 4, 6, 8, and 24 h after dosing for PK analysis.
  • Compound 1 significantly reduced the blood glucose AUC during OGTT at both the 0.3 and 3 mg/kg doses.
  • the compound levels in the plasma showed a dose-related increase.
  • Example 3 Effects of Single Dose of ⁇ '-(2-fluoro-4-(methylsulfonyl)phenyl)-5-methyl-6-(l- (5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine (Compound 1) on oGTT Conducted in Normal Male Cynomolgus Monkeys. The effect of Compound 1 on blood glucose control was tested in healthy male
  • Cynomolgus monkeys Na ⁇ ve, healthy, male Cynomolgus monkeys were used in this study. Monkeys were divided into 3 groups based on basal fasting blood glucose levels. Blood samples were collected before compound dosing, glucose administration, and at 30, 60, and 120 min after glucose nasal gavage (6 g/kg using 50% glucose solution). The results of this study indicate that Compound 1 at the 1.5 mg/kg dose significantly reduced blood glucose levels during oral glucose test.
  • Human cytochrome P450 proteins are a family of enzymes involved in the metabolism of endogenous substrates and detoxication of drugs and other foreign chemicals. P450 enzymes convert various compounds into more polar molecules, and converted molecules are easier to remove from the body. Drug-drug interactions occur when two drugs are co-administered and compete for the same P450 enzyme possibly resulting in incomplete detoxication. Conditions of the assay are summarized in the following table.
  • N-(2-Fluoro-4-(methylsulfonyl)phenyl)-5-methyl-6-(l-(5-methylpyrazin-2-yl)piperidin-4- yloxy)pyrimidin-4-amine was observed to inhibit isoenzymes 3A4, 2C9 and 2Cl 9 by roughly 50 % at 10 ⁇ M with IC 50 values of 9.05, 8.82 and > 10 ⁇ M, respectively. There was no observed inhibition of 1 A2 or 2D6.
  • Example 5 Inhibition of P450 2C9 and 2C19 in HLM by ⁇ -(2-Fluoro-4- (methylsulfonyl)phenyl)-5-methyl-6-(l-(5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin- 4-amine (Compound 1).
  • the metabolic incubations with standard probe substrates were conducted using a 96-well plate format.
  • the general incubation mixture contained 0.5 mg/mL microsomal protein, 0 ⁇ M to 50 ⁇ M N-(2-fluoro-4- (methylsulfonyl)phenyl)-5-methyl-6-(l-(5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4- amine (as inhibitor), 1 mM NADPH and 100 mM potassium phosphate buffer containing 3 mM MgCl 2 and 1 mM EDTA (pH 7.5) in a final volume of 100 ⁇ L.
  • CYP2C9 150 ⁇ M
  • tolbutamide was added to the reaction mixture and incubated for 10 min at 37 0 C.
  • 50 ⁇ M (S)-mephenytoin was added to the reaction mixture and incubated for 40 min at 37 0 C.
  • Parallel control incubations conducted with tolbutamide and (S)-mephenytoin, without the addition of Compound 1, were used as positive controls of the reactions. All of the assays were conducted in triplicate.
  • the incubations were terminated by the addition of 100 ⁇ L acetonitrile containing one of the following compounds as an internal standard: 4'-hydroxytolbutamide-d9 for CYP2C9 and 4'-hydroxymephenytoin-d 3 for CYP2C19 (final concentration 100 ng/mL).
  • the 96-well plates were centrifuged at 3000 rpm for 10 min and 100 ⁇ L supernatant was transferred to a clean 96-well plate for analysis by LC/MS/MS.
  • TLC Thin-layer chromatography
  • PK6F silica gel 60 A 1 mm plates (Whatman)
  • column chromatography was carried out on a silica gel column using Kieselgel 60, 0.063-0.200 mm (Merck). Evaporation was done in vacuo on a Buchi rotary evaporator. Celite 545 ® was used during palladium filtrations.
  • LCMS specs 1) PC: HPLC-pumps: LC-IOAD VP, Shimadzu Inc.; HPLC system controller: SCL-IOA VP, Shimadzu Inc; UV-Detector: SPD-IOA VP, Shimadzu Inc;
  • Example 6.2 Preparation of 4-Chloro-5-methyl-6-(l-(5-methylpyrazin-2-yl)piperidin-4- yloxy)pyrimidine.
  • This material was further characterized by powder X-ray diffraction (XRPD), thermogravimetric analysis (TGA), and differential scanning calorimetry (DSC).
  • XRPD powder X-ray diffraction
  • TGA thermogravimetric analysis
  • DSC differential scanning calorimetry
  • the powder X- ray diffraction (XRPD) pattern shows that the material is crystalline (see Figure 1).
  • the thermogravimetric analysis (TGA) thermogram shows the material has an initial weight loss of about -1.0% in the range of the melting temperature (from about 130 0 C to about 180 0 C, see Figure 2).
  • the differential scanning calorimetry (DSC) thermogram shows the material has a narrow endothermic event at a melting temperature with an onset and peak temperature of about 180.7 ° and about 182.8 0 C respectively, with an enthalpy of 74.3 J/g due to melting of the sample and subsequent exothermic decomposition (see Figure 3).

Abstract

The present invention relates to the GPR119 agonist, N-(2-fluoro-4-(methylsulfonyl)phenyl)-5-methyl-6-(1-(5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine (Compound 1, Formula (I)), and pharmaceutically acceptable salts, solvates, and hydrates thereof, that are useful in the treatment of GPR119-related disorders, such as, metabolic-related disorders and complications thereof, such as, diabetes and obesity.

Description

PIPERIDINE DERIVATIVE AND ITS USE FOR THE TREATMENT OF DIABETS AND OBESITY
FIELD OF THE INVENTION
The present invention relates to the GPRl 19 agonist, N-(2-fluoro-4-(methylsulfonyl) phenyl)-5-methyl-6-( 1 -(5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine (Compound 1 , Formula (I)), and pharmaceutically acceptable salts, solvates, and hydrates thereof, that are useful in the treatment of GPRl 19-related disorders, such as, metabolic-related disorders and complications thereof, such as, diabetes and obesity.
BACKGROUND OF THE INVENTION
A. Diabetes Mellitus
Diabetes mellitus is a serious disease afflicting over 100 million people worldwide. In the United States, there are more than 12 million diabetics, with 600,000 new cases diagnosed each year.
Diabetes mellitus is a diagnostic term for a group of disorders characterized by abnormal glucose homeostasis resulting in elevated blood sugar. There are many types of diabetes, but the two most common are type 1 (also referred to as insulin-dependent diabetes mellitus or IDDM) and type 2 (also referred to as non-insulin-dependent diabetes mellitus or NIDDM).
The etiology of the different types of diabetes is not the same; however, everyone with diabetes has two things in common: overproduction of glucose by the liver and little or no ability to move glucose out of the blood into the cells where it becomes the body's primary fuel.
People who do not have diabetes rely on insulin, a hormone made in the pancreas, to move glucose from the blood into the cells of the body. However, people who have diabetes either don't produce insulin or can't efficiently use the insulin they produce; therefore, they can't move glucose into their cells. Glucose accumulates in the blood creating a condition called hyperglycemia, and over time, can cause serious health problems.
Diabetes is a syndrome with interrelated metabolic, vascular, and neuropathic components. The metabolic syndrome, generally characterized by hyperglycemia, comprises alterations in carbohydrate, fat and protein metabolism caused by absent or markedly reduced insulin secretion and/or ineffective insulin action. The vascular syndrome consists of abnormalities in the blood vessels leading to cardiovascular, retinal and renal complications. Abnormalities in the peripheral and autonomic nervous systems are also part of the diabetic syndrome.
About 5% to 10% of the people who have diabetes have IDDM. These individuals don't produce insulin and therefore must inject insulin to keep their blood glucose levels normal. IDDM is characterized by low or undetectable levels of endogenous insulin production caused by destruction of the insulin-producing β cells of the pancreas, the characteristic that most readily distinguishes IDDM from NIDDM. IDDM, once termed juvenile-onset diabetes, strikes young and older adults alike.
Approximately 90 to 95% of people with diabetes have type 2 (or NIDDM). NIDDM subjects produce insulin, but the cells in their bodies are insulin resistant: the cells don't respond properly to the hormone, so glucose accumulates in their blood. NIDDM is characterized by a relative disparity between endogenous insulin production and insulin requirements, leading to elevated blood glucose levels. In contrast to IDDM, there is always some endogenous insulin production in NIDDM; many NIDDM patients have normal or even elevated blood insulin levels, while other NIDDM patients have inadequate insulin production (Rotwein, R. et al. N. Engl. J. Med. 308, 65-71 (1983)). Most people diagnosed with NIDDM are age 30 or older, and half of all new cases are age 55 and older. Compared with whites and Asians, NIDDM is more common among Native Americans, African- Americans, Latinos, and Hispanics. In addition, the onset can be insidious or even clinically inapparent, making diagnosis difficult.
The primary pathogenic lesion on NIDDM has remained elusive. Many have suggested that primary insulin resistance of the peripheral tissues is the initial event. Genetic epidemiological studies have supported this view. Similarly, insulin secretion abnormalities have been argued as the primary defect in NIDDM. It is likely that both phenomena are important contributors to the disease process (Rimoin, D. L., et. al. Emery and Rimoin's Principles and Practice of Medical Genetics 3r
Ed. 1 :1401-1402 (1996)).
Many people with NIDDM have sedentary lifestyles and are obese: they weigh
approximately 20% more than the recommended weight for their height and build. Furthermore, obesity is characterized by hyperinsulinemia and insulin resistance, a feature shared with NIDDM, hypertension and atherosclerosis.
The patient with diabetes faces a 30% reduced lifespan. After age 45, people with diabetes are about three times more likely than people without diabetes to have significant heart disease and up to five times more likely to have a stroke. These findings emphasize the inter-relations between risks factors for NIDDM and coronary heart disease and the potential value of an integrated approach to the prevention of these conditions (Perry, I. J., et al., BMJ310, 560-564 (1995)).
Diabetes has also been implicated in the development of kidney disease, eye diseases and nervous-system problems. Kidney disease, also called nephropathy, occurs when the kidney's "filter mechanism" is damaged and protein leaks into urine in excessive amounts and eventually the kidney fails. Diabetes is also a leading cause of damage to the retina at the back of the eye and increases risk of cataracts and glaucoma. Finally, diabetes is associated with nerve damage, especially in the legs and feet, which interferes with the ability to sense pain and contributes to serious infections. Taken together, diabetes complications are one of the nation's leading causes of death. B. Obesity
Obesity and diabetes are among the most common human health problems in industrialized societies. In industrialized countries a third of the population is at least 20% overweight. In the United States, the percentage of obese people has increased from 25% at the end of the 1970's, to 33% at the beginning the 1990's. Obesity is one of the most important risk factors for NIDDM. Definitions of obesity differ, but in general, a subject weighing at least 20% more than the recommended weight for his/her height and build is considered obese. The risk of developing NIDDM is tripled in subjects 30% overweight, and three-quarters with NIDDM are overweight.
Obesity, which is the result of an imbalance between caloric intake and energy expenditure, is highly correlated with insulin resistance and diabetes in experimental animals and human.
However, the molecular mechanisms that are involved in obesity-diabetes syndromes are not clear.
During early development of obesity, increased insulin secretion balances insulin resistance and protects patients from hyperglycemia (Le Stunff, et al. Diabetes 43, 696-702 (1989)). However, after several decades, β cell function deteriorates and non-insulin-dependent diabetes develops in about 20% of the obese population (Pederson, P. Diab. Metab. Rev. 5, 505-509 (1989)) and (Brancati, F. L., et al., Arch. Intern. Med. 159, 957-963 (1999)). Given its high prevalence in modern societies, obesity has thus become the leading risk factor for NIDDM (Hill, J. O., et al., Science 280, 1371- 1374 (1998)). However, the factors which predispose a fraction of patients to alteration of insulin secretion in response to fat accumulation remain unknown.
Whether someone is classified as overweight or obese can be determined by a number of different methods, such as, on the basis of their body mass index (BMI) which is calculated by dividing body weight (kg) by height squared (m2). Thus, the units of BMI are kg/m2 and it is possible to calculate the BMI range associated with minimum mortality in each decade of life. Overweight is defined as a BMI in the range 25-30 kg/m2, and obesity as a BMI greater than 30 kg/m2 (see TABLE below). There are problems with this definition in that it does not take into account the proportion of body mass that is muscle in relation to fat (adipose tissue). To account for this, alternately, obesity can be defined on the basis of body fat content: greater than 25% and 30% in males and females, respectively. CLASSIFICATION OF WEIGHT BY BODY MASS INDEX (BMI)
Figure imgf000005_0001
As the BMI increases there is an increased risk of death from a variety of causes that is independent of other risk factors. The most common diseases with obesity are cardiovascular disease (particularly hypertension), diabetes (obesity aggravates the development of diabetes), gall bladder disease (particularly cancer) and diseases of reproduction. Research has shown that even a modest reduction in body weight can correspond to a significant reduction in the risk of developing coronary heart disease.
Obesity considerably increases the risk of developing cardiovascular diseases as well. Coronary insufficiency, atheromatous disease, and cardiac insufficiency are at the forefront of the cardiovascular complication induced by obesity. It is estimated that if the entire population had an ideal weight, the risk of coronary insufficiency would decrease by 25% and the risk of cardiac insufficiency and of cerebral vascular accidents by 35%. The incidence of coronary diseases is doubled in subjects less than 50 years of age who are 30% overweight.
C. Atherosclerosis
Atherosclerosis is a complex disease characterized by inflammation, lipid accumulation, cell death and fibrosis. Atherosclerosis is characterized by cholesterol deposition and monocyte infiltration into the subendothelial space, resulting in foam cell formation. Thrombosis subsequent to atherosclerosis leads to myocardial infarction and stroke. Atherosclerosis is the leading cause of mortality in many countries, including the United States. (See, e.g., Ruggeri, Nat Med (2002) 8:1227-1234; Arehart et al, Circ Res, Circ. Res. (2008) 102:986-993.)
D. Osteoporosis
Osteoporosis is a disabling disease characterized by the loss of bone mass and microarchitectural deterioration of skeletal structure leading to compromised bone strength, which predisposes a patient to increased risk of fragility fractures. Osteoporosis affects more than 75 million people in Europe, Japan and the United States, and causes more than 2.3 million fractures in Europe and the United States alone. In the United States, osteoporosis affects at least 80 years. One in three women older than 50 years will have an osteoporotic fracture that causes a considerable social and financial burden on society. The disease is not limited to women; older men also can be affected. By 2050, the worldwide incidence of hip fracture in men is projected to increase by 310% and 240% in women. The combined lifetime risk for hip, forearm, and vertebral fractures presenting clinically is around 40%, equivalent to the risk for cardiovascular disease. Osteoporotic fractures therefore cause substantial mortaility, morbidity, and economic cost. With an ageing population, the number of osteoporotic fractures and their costs will at least double in the next 50 years unless effective preventive strategies are developed. (See, e.g., Atik et al., Clin Orthop Relat Res (2006) 443: 19-24; Raisz, J Clin Invest (2005) 115:3318-3325; and
World Health Organization Technical Report Series 921 (2003), Prevention and Management of Osteopoosis.)
E. Inflammatory Bowel Disease (IBD)
Inflammatory bowel disease (IBD) is the general name for diseases that cause inflammation in the intestines and includes, e.g. Crohn's disease, ulcerative colitis, ulcerative proctitis. U.S. medical costs of inflammatory bowel disease for 1990 have been estimated to be $1.4 to $1.8 billion. Lost productivity has been estimated to have added an additional $0.4 to $0.8 billion, making the estimated cost of inflammatory bowel disease $1.8 to $2.6 billion. (See, e.g., Pearson, Nursing Times (2004) 100:86-90; Hay et al, J Clin Gastroenterol (1992) 14:309-317;
Keighley et al, Ailment Pharmacol Ther (2003) 18:66-70.)
Enteritis refers to inflammation of the intestine, especially the small intestine, a general condition that can have any of numerous different causes. Enterocolitis refers to inflammation of the small intestine and colon.
Crohn's disease (CD) is an inflammatory process that can affect any portion of the digestive tract, but is most commonly seen in the last part of the small intestine otherwise called the (terminal) ileum and cecum. Altogether this area is also known as the ileocecal region. Other cases may affect one or more of: the colon only, the small bowel only (duodenum, jejunum and/or ileum), the anus, stomach or esophagus. In contrast with ulcerative colitis, CD usually does not affect the rectum, but frequently affects the anus instead. The inflammation extends deep into the lining of the affected organ. The inflammation can cause pain and can make the intestines empty frequently, resulting in diarrhea. Crohn's disease may also be called enteritis. Granulomatous colitis is another name for Crohn's disease that affects the colon. Ileitis is CD of the ileum which is the third part of the small intestine. Crohn's colitis is CD affecting part or all of the colon. Ulcerative colitis (UC) is an inflammatory disease of the large intestine, commonly called the colon. UC causes inflammation and ulceration of the inner lining of the colon and rectum. The inflammation of UC is usually most severe in the rectal area with severity diminishing (at a rate that varies from patient to patient) toward the cecum, where the large and small intestine join. Inflammation of the rectum is called proctitis. Inflammation of the sigmoid colon (located just above the rectum) is called sigmoiditis. Inflammation involving the entire colon is termed pancolitis. The inflammation causes the colon to empty frequently resulting in diarrhea. As the lining of the colon is destroyed ulcers form releasing mucus, pus and blood. Ulcerative proctitis is a form of UC that affects only the rectum.
F. GPRl 19
GPRl 19 is a G protein-coupled receptor (GPRl 19; e.g., human GPRl 19, GenBank® Accession No. AAP72125 and alleles thereof; e.g., mouse GPRl 19, GenBank® Accession No. AY288423 and alleles thereof) and is selectively expressed on pancreatic beta cells. GPRl 19 activation leads to elevation of a level of intracellular cAMP, consistent with GPRl 19 being coupled to Gs. Agonists to GPRl 19 stimulate glucose-dependent insulin secretion in vitro and lower an elevated blood glucose level in vivo; see, e.g., International Applications WO 04/065380 and WO 04/076413, and EP 1338651, the disclosure of each of which is herein incorporated by reference in its entirety. In the literature, GPRl 19 has also been referred to as RUP3 (see, International Application WO 00/31258) and as Glucose-Dependent Insulinotropic Receptor
GDIR (see, Jones, et. al. Expert Opin. Ther. Patents (2009), 19(10): 1339-1359).
GPRl 19 agonists also stimulate the release of Glucose-dependent Insulinotropic
Polypeptide (GIP), Glucagon-Like Peptide- 1 (GLP-I), and at least one other L-cell peptide, Peptide YY (PYY) (Jones, et. al. Expert Opin. Ther. Patents (2009), 19(10): 1339-1359); for specific references related to GPRl 19 agonists and the release of:
GIP, see Shah, Current Opinion in Drug Discovery & Development, (2009) 12:519-532; Jones, et al., Ann. Rep. Med. Chem., (2009) 44:149-170; WO 2007/120689; and WO 2007/120702;
GLP-I, see Shah, Current Opinion in Drug Discovery & Development, (2009) 12:519-532; Jones, et al., Ann. Rep. Med. Chem., (2009) 44:149-170; Schwartz et. al., Cell Metabolism, 2010, 11 :445-447; and WO 2006/076231 ; and
PYY, see Schwartz et. al., Cell Metabolism, 2010, 11 :445-447; and WO 2009/126245.
As mentioned above, GPRl 19 agonists enhance incretin release and therefore can be used in treatment of disorders related to the incretins, such as, GIP, GLP-I, and PYY. However, a number of the incretins, such as, GIP and GLP-I, are substrates for the enzyme DPP-IV. Jones and co-workers (Jones, et al., Ann. Rep. Med. Chem., (2009) 44:149-170) have demonstrated that a combined administration of a GPRl 19 agonist, (2-Fluoro-4-methanesulfonyl-phenyl)- {6-[4-(3- isopropyl-[l,2,4]oxadiazol-5-yl)-piperidin-l-yl]-5-nitro-pyrimidin-4-yl} -amine (see, compound Bl 11 in WO 2004/065380), and a DPP-IV inhibitor acutely increased plasma GLP-I levels and improved glucose tolerance to a significantly greater degree than either agent alone.
G. Glucose-dependent Insulinotropic Poloypeptide (GIP)
Glucose-dependent insulinotropic polypeptide (GIP, also known as gastric inhibitory polypeptide) is a peptide incretin hormone of 42 amino acids that is released from duodenal endocrine K cells after meal ingestion. The amount of GIP released is largely dependent on the amount of glucose consumed. GIP has been shown to stimulate glucose-dependent insulin secretion in pancreatic beta cells. GIP mediates its actions through a specific G protein-coupled receptor, namely GIPR.
As GIP contains an alanine at position 2, it is an excellent substrate for dipeptidyl peptidase-4 (DPP-IV), an enzyme regulating the degradation of GIP. Full-length GIP( 1 -42) is rapidly converted to bioinactive GIP(3-42) within minutes of secretion from the gut K cell. Inhibition of DPP-IV has been shown to augment GIP bioactivity. (See, e.g., Drucker, Cell Metab (2006) 3:153-165; Mclntosh et al., Regul Pept (2005) 128: 159-165; Deacon, Regul Pept (2005) 128: 117-124; and Ahren et al., Endocrinology (2005) 146:2055-2059.). Analysis of full length bioactive GIP, for example in blood, can be carried out using N-terminal-specific assays (see, e.g., Deacon et al, J Clin Endocrinol Metab (2000) 85:3575-3581).
Recently, GIP has been shown to promote bone formation. GIP has been shown to activate osteoblastic receptors, resulting in increases in collagen type I synthesis and alkaline phosphatase activity, both associated with bone formation. GIP has been shown to inhibit osteoclast activity and differentiation in vitro. GIP administration has been shown to prevent the bone loss due to ovariectomy. GIP receptor (GIPR) knockout mice evidence a decreased bone size, lower bone mass, altered bone microarchitecture and biochemical properties, and altered parameters for bone turnover, especially in bone formation. (See, e.g., Zhong et al, Am J Physiol Endocrinol Metab (2007) 292:E543-E548; Bollag et al., Endocrinology (2000) 141 : 1228-1235; Bollag et al., MoI Cell Endocrinol (2001) 177:35-41; Xie et al., Bone (2005) 37:759-769; and
Tsukiyama et al., MoI Endocrinol (2006) 20:1644-1651.)
The usefulness of GIP for maintaining or increasing bone density or formation has been acknowledged by the United State Trademark and Patent Office by issuance of United States Patent No. 6,410,508 for the treatment of reduced bone mineralization by administration of GIP peptide. However, current GIP peptide agonists suffer from a lack of oral bioavailability, negatively impacting patient compliance. An attractive alternative approach is to develop an orally active composition for increasing an endogenous level of GIP activity. H. Glucagon-Like Peptide-1 (GLP-I)
Glucagon- like peptide-1 (GLP-I) is an incretin hormone derived from the
posttranslaltional modification of proglucagon and secreted by gut endocrine cells. GLP-I mediates its actions through a specific G protein-coupled receptor (GPCR), namely GLP-IR. GLP- 1 is best characterized as a hormone that regulates glucose homeostasis. GLP- 1 has been shown to stimulate glucose-dependent insulin secretion and to increase pancreatic beta cell mass.
GLP- 1 has also been shown to reduce the rate of gastric emptying and to promote satiety. The efficacy of GLP- 1 peptide agonists in controlling blood glucose in Type 2 diabetics has been demonstrated in several clinical studies [see, e.g., Nauck et al., Drug News Perspect (2003) 16:413-422], as has its efficacy in reducing body mass [Zander et al., Lancet (2002) 359:824- 830].
GLP- 1 receptor agonists are additionally useful in protecting against myocardial infarction and against cognitive and neurodegenerative disorders. GLP-I has been shown to be cardioprotective in a rat model of myocardial infarction [Bose et al., Diabetes (2005) 54: 146- 151], and GLP-IR has been shown in rodent models to be involved in learning and
neuroprotection [During et al., Nat. Med. (2003) 9: 1173-1179; and Greig et al., Ann N Y Acad Sd
(2004) 1035:290-315].
Certain disorders such as Type 2 diabetes are characterized by a deficiency in GLP- 1 [see, e.g., Nauck et al., Diabetes (2004) 53 Suppl 3 :S 190- 196].
Current GLP- 1 peptide agonists suffer from a lack of oral bioavailability, negatively impacting patient compliance. Efforts to develop orally bioavailable non-peptidergic, small- molecule agonists of GLP-IR have so far been unsuccessful [Mentlein, Expert Opin Investig Drugs (2005) 14:57-64]. An attractive alternative approach is to develop an orally active composition for increasing an endogenous level of GLP- 1 in the blood. I. Peptide YY (PYY)
Peptide YY (PYY) is a 36 amino acid peptide originally isolated in 1980 from porcine intestine (Tatemoto et al, Nature (1980) 285:417-418). PYY is secreted from enteroendocrine L- cells within both the large and small intestine. It has been shown that in rat and human gut concentrations of immunoreactive PYY are low in duodenum and jenunum, high in ileum and colon, and highest in rectum (Lundberg et al, PNAS USA (1982) 79:4471-4475; Adrian et al, Gastroenterol. (1985) 89:1070-1077; Ekblad et al, Peptides (2002) 23:251-261; Ueno et al, Regul Pept (2008) 145: 12-16). (PYY expression in rat been reported to also extend to alpha cells of the islets of Langerhans and to cells in the medulla oblongata (Ekblad et al, Peptides (2002) 23:251- 261 ; PYY is released into the circulation as PYYi_36 and PYY3_36 (Eberlein et al, Peptides (1989)
10:797-803). PYY3_36 is generated from PYYi_36 by cleavage of the N-terminal Tyr and Pro residues by dipeptidyl peptidase IV. PYY3_36 is the predominant form of PYY in human postprandial plasma (Grandt et al, Regul. Pept. (1994) 51 : 151-159). PYYi_36 and PYY3_36 have been reported to have comparable agonist activity at NPY Y2 receptor (Y2R), a G protein- coupled receptor (Parker et al, Br. J. Pharmacol. (2008) 153:420-431); however, PYY3_36 has been reported to be a high-affinity Y2R selective agonist (Keire et al, Am. J. Physiol.
Gastrointest. Liver Physiol. (2000) 279:G126-G131). PYY was subsequently reported to reduce high- fat food intake in rats after peripheral administration (Okada et al, Endocrinology
Supplement (1993) 180) and to cause weight loss in mice after peripheral administration (Morley Qt Ά\, Life Sciences (1987) 41 :2157-2165).
Peripheral administration of PYY3_36 has been reported to markedly reduce food intake and weight gain in rats, to decrease appetite and food intake in humans, and to decrease food intake in mice, but not in Y2R-null mice, which was said to suggest that the food intake effect requires the Y2R. In human studies, infusion of PYY3_36 was found to significantly decrease appetite and reduce food intake by 33% over 24 hours. Infusion of PYY3_36 to reach the normal postprandial circulatory concentrations of the peptide led to peak serum levels of PYY3_36 within 15 minutes, followed by a rapid decline to basal levels within 30 minutes. It was reported that there was significant inhibition of food intake in the 12-hour period following the PYY3_36 infusion, but was essentially no effect on food intake in the 12-hour to 24-hour period. In a rat study, repeated administration of PYY3_36 intraperitoneally (injections twice daily for 7 days) reduced cumulative food intake (Batterham et al, Nature (2002) 418:650-654; Renshaw et al, Current Drug Targets (2005) 6: 171-179).
Peripheral administration of PYY3_36 has been reported to reduce food intake, body weight gain and glycemic indices in diverse rodent models of metabolic diseases of both sexes (Pittner et al, Int. J. Obes. Relat. Metab. Disord. (2004) 28:963-971). It has been reported that blockade of Y2R with the specific antagonist BIIE-246 attenuates the effect of peripherally administered endogenous and exogenous PYY3_36 for reducing food intake (Abbott et al, Brain Res (2005) 1043:139-144). It has been reported that peripheral administration of a novel long- acting selective Y2R polyethylene glycol-conjugated peptide agonist reduces food intake and improves glucose metabolism (glucose disposal, plasma insulin and plasma glucose) in rodents (Ortiz et al, JPET (2007) 323:692-700; Lamb et al, J. Med. Chem. (2007) 50:2264-2268). It has been reported that PYY ablation in mice leads to the development of hyperinsulinemia and obesity (Boey et al, Diabetologia (2006) 49: 1360-1370). It has been reported that peripheral administration of a long-acting, potent and highly selective Y2R agonist inhibits food intake and promotes fat metabolism in mice (Balasubramaniam et al, Peptides (2007) 28:235-240).
There is evidence that agents which stimulate PYY synthesis in vivo can confer protection against diet- induced and genetic obesity and can improve glucose tolerance (Boey et al, Neuropeptides (2008) 42: 19-30).
It has been reported that Y2R agonists such as PYYi_36 and PYY3_36 can confer protection against epileptic seizures, such as against kainate seizures (El Bahh et al, Eur. J. Neurosci. (2005) 22: 1417-1430; Woldbye et al, Neurobiology of Disease (2005) 20:760-772).
It has been reported that Y2R agonists such as PYYi_36 and PYY3-36 act as proabsorbtive (or anti-secretory) hormones, increasing upon intravenous administration the absorption of both water and sodium in various parts of the bowel (Bilchik et al, Gastroenterol. (1993) 105:1441-
1448; Liu et al, J. Surg. Res. (1995) 58:6-11; Nightingale et al, Gut (1996) 39:267-272; Liu et al, Am Surg (1996) 62:232-236; Balasubramaniam et al, J. Med. Chem. (2000) 43:3420-3427). It has been reported that Y2R agonists such as PYY analogues inhibit secretion and promote absorption and growth in the intestinal epithelium (Balasubramaniam et al, J. Med. Chem. (2000) 43:3420- 3427). It has been reported that PYY promotes intestinal growth in normal rats (Gomez et al, Am.
J. Physiol. (1995) 268:G71-G81). It has been reported that Y2R agonists such as PYYi_36 and PYY3_36 inhibit bowel motility and work to prevent diarrhea (EP1902730; also see Cox, Peptides (2007) 28:345-351).
It has been reported that Y2R agonists such as PYYi_36 and PYY3_36 can confer protection against inflammatory bowel disease such as ulcerative colitis and Crohn's disease (WO
03/105763). It has been reported that PYY-deficient mice exhibit an osteopenic phenotype, i.e. that PYY can increase bone mass and/or can confer protection against loss of bone mass (e.g., decreases loss of bone mass) (Wortley et al, Gastroenterol. (2007) 133: 1534-1543). It has been reported that PYY3_36 can confer protection in rodent models of pancreatitis (Vona-Davis et al, Peptides (2007) 28:334-338).
It has been reported that angiogenesis is impaired in Y2R-deficient mice (Lee et al, Peptides (2003) 24:99-106), i.e. that agonists of Y2R such as PYYi_36 and PYY3_36 promote angiogenesis. It has been reported that would healing is impaired in Y2R-deficient mice
(Ekstrand et al, PNAS USA (2003) 100:6033-6038), i.e. that agonists of Y2R such as PYYi_36 and PYY3-36 promote wound healing. It has been reported that ischemic angiogenesis is impaired in Y2R-deficient mice (Lee et al, J. Clin. Invest. (2003) 111 : 1853-1862), i.e. that agonists of Y2R such as PYYi_36 and PYY3_36 promotes revascularization and restoration of function of ischemic tissue. It has been reported that agonists of Y2R such as PYYi_36 and PYY3_36 mediate increases in collateral-dependent blood flow in a rat model of peripheral arterial disease (Cruze et al, Peptides
(2007) 28:269-280).
It has been reported that PYY and Y2R agonists such as PYY3_36 can suppress tumor growth in the cases of, e.g., pancreatic cancer such as pancreatic ductal adenocarcinoma, breast cancer such as breast infiltrative ductal adenocarcinoma, colon cancer such as colon
adenocarcinoma and Barrett's adenocarcinoma (Liu et al, Surgery (1995) 118:229-236; Liu et al,
J. Surg. Res. (1995) 58:707-712; Grise et al, J. Surg. Res. (1999) 82: 151-155; Tseng et al, Peptides (2002) 23:389-395; McFadden et al, Am. J. Surg. (2004) 188:516-519).
It has been reported that stimulation of Y2R such as by PYY3_36 leads to an increase in plasma adiponectin (Ortiz et al, JPET (2007) 323:692-700). Adiponectin is an adipokine with potent anti-inflammatory properties (Ouchi et al, Clin Chim Acta (2007) 380:24-30; TiIg et al,
Nat. Rev. Immunol. (2006) 6:772-783). Adiponectin exerts anti-atherogenic effects by targeting vascular endothelial cells and macrophages and insulin-sensitizing effects, predominantly in muscle and liver (Kubota et al, J. Biol. Chem. (2002) 277:25863-25866; Maeda et al, Nat. Med. (2002) 8:731-737). Low adiponectin levels have been reported to be associated with atherogenic lipoproteins in dyslipidemia (elevated triglycerides, small dense LDL cholesterol, low HDL cholesterol) (Marso et al, Diabetes Care (2008) Feb 5 Epub ahead of print). Adiponectin has been implicated in high density lipoprotein (HDL) assembly (Oku et al, FEBS Letters (2007)
581 :5029-5033). Adiponectin has been found to ameliorate the abnormalities of metabolic syndrome, including insulin resistance, hyperglycemia, and dyslipidemia, in a mouse model of obesity-linked metabolic syndrome associated with decreased adiponectin levels (Hara et al,
Diabetes Care (2006) 29: 1357- 1362). Adiponectin has been reported to stimulate angiogenesis in response to tissue ischemia (Shibata et al, J. Biol. Chem. (2004) 279:28670-28674). Adiponectin has been reported to prevent cerebral ischemic injury through endothelial nitric oxide synthase- dependent mechanisms (Nishimura et al, Circulation (2008) 117:216-223). Adiponectin has been reported to confer protection against myocardial ischemia-reperfusion injury (Shibata et al, Nat
Med (2005) 11 : 1096-1103; Tao et al, Circulation (2001) 115: 1408-1416). Adiponectin has been reported to confer protection against myocardial ischaemia-reperfusion injury via AMP- activated protein kinase, Akt, and nitric oxide (Gonon et al, Cardiovasc Res. (2008) 78:116-122).
Adiponectin has been reported to confer protection against the development of systolic dysfunction following myocardial infarction, through its abilities to suppress cardiac hypertrophy and interstitial fibrosis, and proctect against myocyte and capillary loss (Shibata et al, J. MoI. Cell Cardiol. (2007) 42: 1065-1074). Adiponectin has been reported to confer protection against inflammatory lung disease; adiponectin-deficient mice exhibit an emphysema-like phenotype (Summer et al, Am J. Physiol. Lung Cell MoI. Physiol (March 7, 2008)). Adiponectin has been reported to confer protection against allergic airway inflammation and airway
hyperresponsiveness such as may be associated with asthma (Shore et al, J. Allergy Clin.
Immunol (2006) 118:389-395). Adiponectin has been suggested to confer protection against pulmonary arterial hypertension by virtue of its insulin-sensitizing effects (Hansmann et al, Circulation (2007) 115: 1275-1284). Adiponectin has been reported to ameliorate obesity-related hypertension, with said amelioration of hypertension being associated in part with upregulated prostacyclin expression (Ohashi et al, Hypertension (2006) 47:1108-1116). Adiponectin has been reported to decrease tumor necrosis factor (TNF)-α- induced expression of the adhesion molecules VCAM-I, E-selectin and ICAM-I in human aortic endothelial cells (HAECs) (Ouchi et al, Circulation (1999) 100:2473-2476) and to inhibit production of TNF-α in macrophages (Yokota et al, Blood (2000) 96:1723-1732). Adiponectin has been reported to confer protection against restenosis after vascular intervention (Matsuda et al, J Biol Chem (2002) 277:37487-37491). The central role of TNF-α in inflammation has been demonstrated by the ability of agents that block the action of TNF-α to treat a range of inflammatory conditions. TNF-α-mediated inflammatory conditions encompass rheumatoid arthritis, inflammatory bowel disease such as Crohn's disease, ankylosing spondylitis, psoriasis, ischemic brain injury, cardiac allograft rejection, asthma, and the like (Bradley, J Pathol (2008) 214:149-160). See, e.g., Yamamoto et al, Clinical Science (2002) 103:137-142; Behre, ScandJ Clin Lab Invest (2007) 67:449-458; Guerre-Millo, Diabetes & Metabolism (2008) 34: 12-18; Parker et al, Br. J. Pharmacol. (2008) 153:420-431.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 shows a powder X-ray diffraction (PXRD) pattern for Compound 1 as prepared according to the procedure described in Example 6.
Figure 2 shows a thermogravimetric analysis (TGA) thermogram for Compound 1 as prepared according to the procedure described in Example 6.
Figure 3 shows a differential scanning calorimetry (DSC) thermogram for Compound 1 as prepared according to the procedure described in Example 6. SUMMARY OF THE INVENTION
The present invention is drawn to N-(2-fluoro-4-(methylsulfonyl) phenyl)-5-methyl-6-(l- (5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine (Compound 1, Formula (I)) and pharmaceutically acceptable salts, solvates, and hydrates thereof, which bind to and modulate the activity of a GPCR, referred to herein as GPRl 19, and uses thereof. The term GPRl 19, as used herein, includes the human sequences found in GeneBank accession number AY288416, naturally-occurring allelic variants, mammalian orthologs, and recombinant mutants thereof. A preferred human GPRl 19 for use in screening and testing of the compounds of the invention is provided in the nucleotide sequence of Seq. ID.No: 1 and the corresponding amino acid sequence in Seq. ID.No:2 found in PCT Application No. WO2005/007647, the disclosure which is herein incorporated by reference in its entirety.
One aspect of the present invention encompasses compounds selected from the following compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000014_0001
One aspect of the present invention pertains to pharmaceutical compositions comprising a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, and a pharmaceutically acceptable carrier.
One aspect of the present invention pertains to compositions comprising a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, and an inhibitor of dipeptidyl peptidase IV (DPP-IV).
One aspect of the present invention pertains to compositions wherein the inhibitor of DPP-IV is selected from the following compounds and pharmaceutically acceptable salts, solvates, and hydrates thereof:
3(R)-amino-l-[3-(trifluoromethyl)-5,6,7,8-tetrahydro[l,2,4]triazolo[4,3-a]pyrazin-7-yl]- 4-(2,4,5-trifluorophenyl) butan-1-one;
1 -[2-(3-hydroxyadamant- 1 -ylamino)acetyl]pyrrolidine-2(S)-carbonitrile;
(lS,3S,5S>2-[2(S>amino-2-(3-hydroxyadamantan-l-yl)acetyl]-2- azabicyclo[3.1.0]hexane-3-carbonitrile;
2-[6-[3(R)-aminopiperidin- 1 -yl]-3-methyl-2,4-dioxo- 1 ,2,3,4-tetrahydropyrimidin- 1 - ylmethyljbenzonitrile; 8-[3(R)-aminopiperidin- 1 -yl]-7-(2-butynyl)-3-methyl- 1 -(4-methylquinazolin-2- ylmethyl)xanthine;
l-[N-[3(R)-pyrrolidinyl]glycyl]pyrrolidin-2(R)-yl boronic acid;
4(5)-fluoro-l-[2-[(lΛ,35)-3-(lH-l,2,4-triazol-l- ylmethyl)cyclopentylamino]acetyl]pyrrolidme-2(£)-carbonitrile;
1 -[(2S,3S,l lb£)-2-amino-9,l 0-dimethoxy-2,3,4,6,7,l lb-hexahydro-lH-pyrido[2,l- a]isoquinolin-3-yl]-4(S)-(fluoromethyl)pyrrolidin-2-one;
(2S,4S)-2-cyano-4-fluoro- 1 -[(2-hydroxy- 1 , 1 -dimethyl) ethylamino]acetylpyrrolidine;
8-(c^-hexahydro-pyrrolo[3,2-b]pyrrol-l-yl)-3-methyl-7-(3-methyl-but-2-enyl)-l-(2-oxo- 2-phenylethyl)-3,7-dihydro-purine-2,6-dione;
1 -((3S,45)-4-amino- 1 -(4-(3,3-difluoropyrrolidin- 1 -yl)- 1 ,3,5-triazin-2-yl)pyrrolidin-3-yl)- 5,5difluoropiperidin-2-one;
(R)-2-((6-(3-aminopiperidin- 1 -yl)-3-methyl-2,4-dioxo-3,4-dihydropyrimidin- 1 (2H)- yl)methyl)-4-fluorobenzonitrile;
5-{(S)-2-[2-((S)-2-cyano-pyrrolid in-l-yl)-2-oxo-ethylamino]-propyl}-5-(lH-tetrazol-5- yl)10,l l-dihydro-5H-dibenzo[a,d]cycloheptene-2,8-dicarboxylic acid bis-dimethylamide;
((2S, 4S)-4-(4-(3 -methyl- 1 -phenyl- lH-pyrazol-5-yl)piperazin- 1 -yl)pyrrolidin-2- yl)(thiazolidin-3-yl)methanone;
(2S,AS)- 1 -[2-[(4-ethoxycarbonylbicyclo[2.2.2]oct- 1 -yl)amino] acetyl] -4- fluoropyrrolidine-2-carbonitrile;
6-[(3R)-3-amino-piperidin- 1 -yl]-5-(2-chloro-5-fluoro-benzyl)- 1 ,3-dimethyl- 1 ,5dihydro- pyrrolo[3,2-d]pyrimidine-2,4-dione;
2-({6-[(3R)-3-amino-3-methylpiperidin- 1 -yl]- 1 ,3-dimethyl-2,4-dioxo- 1 ,2,3,4-tetrahydro- 5H-pyrrolo[3,2-d]pyrimidin-5-yl}methyl)-4-fluorobenzonitrile;
(2S)-l-{[2-(5-methyl-2-phenyl-oxazol-4-yl)-ethylamino]-acetyl}-pyrrolidine-2- carbonitrile;
(2S)- 1 - { [ 1 , 1 -dimethyl-3-(4-pyridin-3-yl-imidazol- 1 -yl)-propylamino]-acetyl} - pyrrolidine-2-carbonitrile;
(3,3-difluoropyrrolidin- 1 -yl)-((2S,4S)-4-(4-(pyrimidin-2yl)piperazin- 1 -yl)pyrrolidin-2- yl)methanone;
(2S,4S)-l-[(2S)-2-amino-3,3-bis(4-fluorophenyl)propanoyl]-4-fluoropyrrolidine-2- carbonitrile;
(2S,5R)-5-ethynyl- 1 - {N-(4-methyl- 1 -(4-carboxy-pyridin-2-yl)piperidin-4- yl)glycyl}pyrrolidine-2-carbonitrile; and (lS,6R)-3- {[3-(trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)- yl]carbonyl} -6-(2,4,5-trifluorophenyl)cyclohex-3-en- 1 -amine.
One aspect of the present invention pertains to compositions wherein the inhibitor of DPP-IV is selected from the following compounds and pharmaceutically acceptable salts, solvates, and hydrates thereof:
3(R)-amino-l-[3-(trifluoromethyl)-5,6,7,8-tetrahydro[l,2,4]triazolo[4,3-a]pyrazin-7-yl]- 4-(2,4,5-trifluorophenyl)butan- 1 -one;
1 -[2-(3-hydroxyadamant- 1 -ylamino)acetyl]pyrrolidine-2(S)-carbonitrile;
(lS,3S,5S>2-[2(S>amino-2-(3-hydroxyadamantan-l-yl)acetyl]-2- azabicyclo[3.1.0]hexane-3-carbonitrile;
2-[6-[3(R)-aminopiperidin- 1 -yl]-3-methyl-2,4-dioxo- 1 ,2,3,4-tetrahydropyrimidin- 1 - ylmethyljbenzonitrile;
8-[3(R)-aminopiperidin- 1 -yl]-7-(2-butynyl)-3-methyl- 1 -(4-methylquinazolin-2- ylmethyl)xanthine;
1 -[N-[3(R)-pyrrolidinyl]glycyl]pyrrolidin-2(R)-yl boronic acid;
4(5)-fluoro-l-[2-[(lR,3S)-3-(lH-l,2,4-triazol-l-ylmethyl)cyclopen1ylamino] acetyl] pyrrolidine-2(S)-carbonitrile;
1 -[(2S,3S,l lb£)-2-amino-9,l 0-dimethoxy-2,3,4,6,7,l lb-hexahydro-lH-pyrido[2,l- a]isoquinolm-3-yl]-4(S)-(fluoromethyl)pyrrolidin-2-one; and
(2S,4S)- 1 -[(2S)-2-amino-3,3-bis(4-fluorophenyl)propanoyl]-4-fluoropyrrolidine-2- carbonitrile.
One aspect of the present invention pertains to compositions wherein the inhibitor of DPP-IV is selected from the following compounds and pharmaceutically acceptable salts, solvates, and hydrates thereof:
3(R)-amino-l-[3-(trifluoromethyl)-5,6,7,8-tetrahydro[l,2,4]triazolo[4,3-a]pyrazin-7-yl]-
4-(2,4,5-trifluorophenyl)butan- 1 -one;
1 -[2-(3-hydroxyadamant- 1 -ylamino)acetyl]pyrrolidine-2(S)-carbonitrile;
(lS,3S,5S)-2-[2(S)-amino-2-(3-hydroxyadamantan-l-yl)acetyl]-2- azabicyclo[3.1.0]hexane-3-carbonitrile;
2-[6-[3(R)-aminopiperidin- 1 -yl]-3-methyl-2,4-dioxo- 1 ,2,3,4-tetrahydropyrimidin- 1 - ylmethyljbenzonitrile;
8-[3(R)-aminopiperidin- 1 -yl]-7-(2-butynyl)-3-methyl- 1 -(4-methylquinazolin-2- ylmethyl)xanthine;
l-[N-[3(R)-pyrrolidinyl]glycyl]pyrrolidin-2(R)-yl boronic acid; 4(5)-fluoro-l-[2-[(lR,35)-3-(lH-l,2,4-triazol-l-ylmethyl)cyclopen1ylamino] acetyl] pyrrolidine-2(S)-carbonitrile; and
1 -[(2S,3S,l lb£)-2-amino-9,l 0-dimethoxy-2,3,4,6,7,l lb-hexahydro-lH-pyrido[2,l- a]isoquinohn-3-yl]-4(S)-(fluoromethyl)pyrrolidin-2-one.
Some embodiments of the present invention in regard to the inhibitor of DPP-IV include every combination of one or more inhibitors of DPP-IV and pharmaceutically acceptable salts, solvates, and hydrates thereof, selected from the following group:
3(R)-amino-l-[3-(trifluoromethyl)-5,6,7,8-tetrahydro[l,2,4]triazolo[4,3-a]pyrazin-7-yl]- 4-(2,4,5-trifluorophenyl)butan- 1 -one;
1 -[2-(3-hydroxyadamant- 1 -ylamino)acetyl]pyrrolidine-2(S)-carbonitrile;
(lS,3S,5S>2-[2(S>amino-2-(3-hydroxyadamantan-l-yl)acetyl]-2- azabicyclo[3.1.0]hexane-3-carbonitrile;
2-[6-[3(R)-aminopiperidin- 1 -yl]-3-methyl-2,4-dioxo- 1 ,2,3,4-tetrahydropyrimidin- 1 - ylmethyljbenzonitrile;
8-[3(R)-aminopiperidin- 1 -yl]-7-(2-butynyl)-3-methyl- 1 -(4-methylquinazolin-2- ylmethyl)xanthine;
l-[N-[3(R)-pyrrolidinyl]glycyl]pyrrolidin-2(R)-yl boronic acid;
4(5)-fluoro-l-[2-[(lR,3S)-3-(lH-l,2,4-triazol-l-ylmethyl)cyclopen1ylamino] acetyl] pyrrolidine-2(S)-carbonitrile; and
\-[(2S,3S,\ lbS)-2-amino-9,10-dimethoxy-2,3,4,6,7,l lb-hexahydro-lH-pyrido[2,l- a]isoquinolm-3-yl]-4(S)-(fluoromethyl)pyrrolidin-2-one.
In some embodiments, the inhibitor of dipeptidyl peptidase IV is selected from the following compound and pharmaceutically acceptable salts, solvates, and hydrates thereof:
3(R)-amino-l-[3-(trifluoromethyl)-5,6,7,8-tetrahydro[l,2,4]triazolo[4,3-a]pyrazin-7-yl]- 4-(2,4,5-trifluorophenyl)butan- 1 -one.
In some embodiments, the inhibitor of dipeptidyl peptidase IV is 3(R)-amino-l-[3- (trifluoromethyl)-5,6,7,8-tetrahydro[l,2,4]triazolo[4,3-a]pyrazin-7-yl]-4-(2,4,5- trifluorophenyl)butan- 1 -one phosphate (1 : 1) monohydrate.
One aspect of the present invention pertains to compositions, further comprising a pharmaceutically acceptable carrier.
One aspect of the present invention pertains to compositions, wherein:
the compound is selected from the following compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000018_0001
00 ; and
the inhibitor of dipeptidyl peptidase IV is selected from the following compound and pharmaceutically acceptable salts, solvates, and hydrates thereof:
3(R)-amino-l-[3-(trifluoromethyl)-5,6,7,8-tetrahydro[l,2,4]triazolo[4,3-a]pyrazin-7-yl]- 4-(2,4,5-trifluorophenyl)butan-l-one. Some embodiments of the present invention pertains to compositions, further comprising a pharmaceutically acceptable carrier.
One aspect of the present invention pertains to methods of modulating the activity of a GPRl 19 receptor by contacting the receptor with a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof.
One aspect of the present invention pertains to methods of agonizing a GPRl 19 receptor by contacting the receptor with a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof.
One aspect of the present invention pertains to methods for the treatment of a GPRl 19 receptor related disorder in an individual, comprising administering to the individual in need thereof, a therapeutically effective amount of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof.
In some embodiments, the GPRl 19 receptor related disorder is a metabolic-related disorder.
One aspect of the present invention pertains to methods for the treatment of a metabolic- related disorder in an individual, comprising administering to the individual in need thereof, a therapeutically effective amount of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof.
One aspect of the present invention pertains to methods for the treatment of a metabolic- related disorder selected from the group consisting of:
diabetes mellitus, type 1 diabetes, type 2 diabetes, inadequate glucose tolerance, impaired glucose tolerance, insulin resistance, hyperglycemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia, atherosclerosis, stroke, syndrome X, hypertension, pancreatic beta-cell insufficiency, enteroendocrine cell insufficiency, glucosuria, metabolic acidosis, cataracts, diabetic nephropathy, diabetic neuropathy, peripheral neuropathy, diabetic coronary artery disease, diabetic cerebrovascular disease, diabetic peripheral vascular disease, diabetic retinopathy, metabolic syndrome, a condition related to diabetes mellitus, myocardial infarction, learning impairment, memory impairment, a neurodegenerative disorder, a condition ameliorated by increasing a blood GLP- 1 level in an individual with a neurodegenerative disorder, excitotoxic brain damage caused by severe epileptic seizures, Alzheimer's disease, Parkinson's disease, Huntington's disease, prion-associated disease, stroke, motor-neuron disease, traumatic brain injury, spinal cord injury, and obesity.
One aspect of the present invention pertains to methods for the treatment of type 2 diabetes in an individual, comprising administering to the individual in need thereof, a therapeutically effective amount of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof.
One aspect of the present invention pertains to methods for the treatment of
hyperglycemia in an individual, comprising administering to the individual in need thereof, a therapeutically effective amount of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof.
One aspect of the present invention pertains to methods for the treatment of
hyperlipidemia in an individual, comprising administering to the individual in need thereof, a therapeutically effective amount of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof.
One aspect of the present invention pertains to methods for the treatment of
hypertriglyceridemia in an individual, comprising administering to the individual in need thereof, a therapeutically effective amount of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof.
One aspect of the present invention pertains to methods for the treatment of type 1 diabetes in an individual, comprising administering to the individual in need thereof, a therapeutically effective amount of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof.
One aspect of the present invention pertains to methods for the treatment of dyslipidemia in an individual, comprising administering to the individual in need thereof, a therapeutically effective amount of a compound, selected from the compound of Formula (I) and
pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof.
One aspect of the present invention pertains to methods for the treatment of syndrome X in an individual, comprising administering to the individual in need thereof, a therapeutically effective amount of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof.
In some embodiments, the individual is a mammal.
In some embodiments, the mammal is a human.
One aspect of the present invention pertains to methods of treating obesity in an individual comprising administering to the individual in need of treatment a therapeutically effective amount of a compound, selected from the compound of Formula (I) and
pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof.
One aspect of the present invention pertains to methods of decreasing food intake of an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof.
One aspect of the present invention pertains to methods of inducing satiety in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof.
One aspect of the present invention pertains to methods of controlling or decreasing weight gain of an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof.
One aspect of the present invention pertains to methods for the treatment of a GPRl 19 receptor related disorder in an individual, comprising administering to the individual in need thereof, a therapeutically effective amount of a compound according to claim 1 in combination with an inhibitor of DPP-IV, wherein the compound and the inhibitor of DPP-IV are administered simultaneously, separately, or sequentially.
In some embodiments, the compound and the inhibitor of DPP-IV are administered simultaneously.
In some embodiments, the compound and the inhibitor of DPP-IV are administered separately.
In some embodiments, the compound and the inhibitor of DPP-IV are administered sequentially.
In some embodiments, the GPRl 19 receptor related disorder is a metabolic-related disorder. In some embodiments, the metabolic-related disorder is selected from the group consisting of:
diabetes mellitus, type 1 diabetes, type 2 diabetes, inadequate glucose tolerance, impaired glucose tolerance, insulin resistance, hyperglycemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia, atherosclerosis, stroke, syndrome X, hypertension, pancreatic beta-cell insufficiency, enteroendocrine cell insufficiency, glucosuria, metabolic acidosis, cataracts, diabetic nephropathy, diabetic neuropathy, peripheral neuropathy, diabetic coronary artery disease, diabetic cerebrovascular disease, diabetic peripheral vascular disease, diabetic retinopathy, metabolic syndrome, a condition related to diabetes mellitus, myocardial infarction, learning impairment, memory impairment, a neurodegenerative disorder, a condition ameliorated by increasing a blood GLP- 1 level in an individual with a neurodegenerative disorder, excitotoxic brain damage caused by severe epileptic seizures, Alzheimer's disease, Parkinson's disease, Huntington's disease, prion-associated disease, stroke, motor-neuron disease, traumatic brain injury, spinal cord injury, and obesity.
In some embodiments, the metabolic-related disorder is type 2 diabetes.
In some embodiments, the metabolic-related disorder is hyperglycemia.
In some embodiments, the metabolic-related disorder is hyperlipidemia.
In some embodiments, the metabolic-related disorder is hypertriglyceridemia.
In some embodiments, the metabolic-related disorder is type 1 diabetes.
In some embodiments, the metabolic-related disorder is dyslipidemia.
In some embodiments, the metabolic-related disorder is syndrome X.
One aspect of the present invention pertains to methods of treating obesity in an individual comprising administering to the individual in need of treatment a therapeutically effective amount of a compound according to claim 1 in combination with an inhibitor of DPP- IV, wherein the compound and the inhibitor of DPP-IV are administered simultaneously, separately, or sequentially.
One aspect of the present invention pertains to methods of decreasing food intake of an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound according to claim 1 in combination with an inhibitor of DPP-IV, wherein the compound and the inhibitor of DPP-IV are administered simultaneously, separately, or sequentially.
One aspect of the present invention pertains to methods of inducing satiety in an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound according to claim 1 in combination with an inhibitor of DPP-IV, wherein the compound and the inhibitor of DPP-IV are administered simultaneously, separately, or sequentially.
One aspect of the present invention pertains to methods of controlling or decreasing weight gain of an individual comprising administering to the individual in need thereof a therapeutically effective amount of a compound according to claim 1 in combination with an inhibitor of DPP-IV, wherein the compound and the inhibitor of DPP-IV are administered simultaneously, separately, or sequentially.
In some embodiments, the compound and the inhibitor of DPP-IV are administered simultaneously.
In some embodiments, the compound and the inhibitor of DPP-IV are administered separately.
In some embodiments, the compound and the inhibitor of DPP-IV are administered sequentially.
One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for modulating the activity of a GPRl 19 receptor.
One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for agonizing a GPRl 19 receptor.
One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for the treatment of a GPRl 19 receptor related disorder.
One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for the treatment of a metabolic- related disorder.
One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for the treatment of a metabolic- related disorder selected from the group consisting of:
diabetes mellitus, type 1 diabetes, type 2 diabetes, inadequate glucose tolerance, impaired glucose tolerance, insulin resistance, hyperglycemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia, atherosclerosis, stroke, syndrome X, hypertension, pancreatic beta-cell insufficiency, enteroendocrine cell insufficiency, glucosuria, metabolic acidosis, cataracts, diabetic nephropathy, diabetic neuropathy, peripheral neuropathy, diabetic coronary artery disease, diabetic cerebrovascular disease, diabetic peripheral vascular disease, diabetic retinopathy, metabolic syndrome, a condition related to diabetes mellitus, myocardial infarction, learning impairment, memory impairment, a neurodegenerative disorder, a condition ameliorated by increasing a blood GLP- 1 level in an individual with a neurodegenerative disorder, excitotoxic brain damage caused by severe epileptic seizures, Alzheimer's disease, Parkinson's disease, Huntington's disease, prion-associated disease, stroke, motor-neuron disease, traumatic brain injury, spinal cord injury, and obesity.
One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for the treatment of type 2 diabetes.
One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for the treatment of hyperglycemia.
One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for the treatment of hyperlipidemia.
One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for the treatment of
hypertriglyceridemia.
One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for the treatment of type 1 diabetes.
One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for the treatment of dyslipidemia.
One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for the treatment of syndrome X. One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for the treatment of obesity.
One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for decreasing food intake in an individual.
One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for inducing satiety in an individual.
One aspect of the present invention pertains to the use of a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, or a composition thereof in the manufacture of a medicament for controlling or decreasing weight gain in an individual.
One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in a method of treatment of the human or animal body by therapy.
One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in a method of modulating the activity of a GPRl 19 receptor.
One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in a method of agonizing a GPRl 19 receptor.
One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in a method of treatment of a GPRl 19 receptor related disorder.
One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in a method of treatment of a metabolic-related disorder.
One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in a method of treatment of a metabolic-related disorder selected from the group consisting of:
diabetes mellitus, type 1 diabetes, type 2 diabetes, inadequate glucose tolerance, impaired glucose tolerance, insulin resistance, hyperglycemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia, atherosclerosis, stroke, syndrome X, hypertension, pancreatic beta-cell insufficiency, enteroendocrine cell insufficiency, glucosuria, metabolic acidosis, cataracts, diabetic nephropathy, diabetic neuropathy, peripheral neuropathy, diabetic coronary artery disease, diabetic cerebrovascular disease, diabetic peripheral vascular disease, diabetic retinopathy, metabolic syndrome, a condition related to diabetes mellitus, myocardial infarction, learning impairment, memory impairment, a neurodegenerative disorder, a condition ameliorated by increasing a blood GLP- 1 level in an individual with a neurodegenerative disorder, excitotoxic brain damage caused by severe epileptic seizures, Alzheimer's disease, Parkinson's disease, Huntington's disease, prion-associated disease, stroke, motor-neuron disease, traumatic brain injury, spinal cord injury, and obesity.
One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in a method of treatment of type 2 diabetes.
One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in a method of treatment of hyperglycemia.
One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in a method of treatment of hyperlipidemia.
One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in a method of treatment of hypertriglyceridemia.
One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in a method of treatment of type 1 diabetes.
One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in a method of treatment of dyslipidemia.
One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in a method of treatment of syndrome X.
One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in a method of treatment of obesity.
One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in decreasing food intake in an individual.
One aspect of the present invention pertains to compounds of the present invention for use in inducing satiety in an individual.
One aspect of the present invention pertains to compounds of the present invention or compositions thereof for use in controlling or decreasing weight gain in an individual.
In some embodiments, the individual is a mammal. In some embodiments, the mammal is a human. In some embodiments, the human has a body mass index of about 18.5 to about 45 or greater. In some embodiments, the human has a body mass index of about 18.5 to about 45. In some embodiments, the human has a body mass index of about 25 to about 45. In some embodiments, the human has a body mass index of about 30 to about 45. In some embodiments, the human has a body mass index of about 35 to about 45. The present invention includes every combination of one or more metabolic-related disorders selected from the group consisting of diabetes mellitus, type 1 diabetes, type 2 diabetes, inadequate glucose tolerance, impaired glucose tolerance, insulin resistance, hyperglycemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia, atherosclerosis, stroke, syndrome X, hypertension, pancreatic beta-cell insufficiency, enteroendocrine cell insufficiency, glucosuria, metabolic acidosis, cataracts, diabetic nephropathy, diabetic neuropathy, peripheral neuropathy, diabetic coronary artery disease, diabetic cerebrovascular disease, diabetic peripheral vascular disease, diabetic retinopathy, metabolic syndrome, a condition related to diabetes mellitus, myocardial infarction, learning impairment, memory impairment, a neurodegenerative disorder, a condition ameliorated by increasing a blood GLP- 1 level in an individual with a neurodegenerative disorder, excitotoxic brain damage caused by severe epileptic seizures, Alzheimer's disease, Parkinson's disease, Huntington's disease, prion- associated disease, stroke, motor-neuron disease, traumatic brain injury, spinal cord injury, and obesity.
Some embodiments of the present invention include every combination of one or more metabolic-related disorder selected from the group consisting of type 2 diabetes, hyperglycemia, hyperinsulinemia, hyperlipidemia, hypertriglyceridemia, insulin resistance, type 1 diabetes, idiopathic type 1 diabetes (type Ib), latent autoimmune diabetes in adults (LADA), early-onset type 2 diabetes (EOD), youth-onset atypical diabetes (YOAD), maturity onset diabetes of the young (MODY), malnutrition-related diabetes, gestational diabetes, coronary heart disease, vascular restenosis, restenosis, restenosis after angioplasty, peripheral vascular disease, claudication, intermittent claudication, cell death associated with myocardial infarction (e.g. necrosis and apoptosis), dyslipidemia, post-prandial lipemia, conditions of impaired glucose tolerance (IGT), impaired glucose metabolism, conditions of impaired glucose metabolism, conditions of impaired fasting plasma glucose, metabolic acidosis, ketosis, arthritis, obesity, osteoporosis, hypertension, congestive heart failure, left ventricular hypertrophy, peripheral arterial disease, diabetic retinopathy, macular degeneration, cataract, diabetic nephropathy, glomerulosclerosis, chronic renal failure, diabetic neuropathy, metabolic syndrome, syndrome X, premenstrual syndrome, angina pectoris, thrombosis, atherosclerosis, ischemic stroke, transient ischemic attacks, stroke, erectile dysfunction, skin and connective tissue disorders, foot ulcerations, ulcerative colitis, endothelial dysfunction, and impaired vascular compliance.
One aspect of the present invention pertains to methods of preparing compositions comprising the step of admixing a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, and a pharmaceutically acceptable carrier.
One aspect of the present invention pertains to methods of preparing pharmaceutically compositions comprising the step of admixing a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, and a pharmaceutically acceptable carrier.
One aspect of the present invention pertains to methods of preparing compositions comprising the step of admixing a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, and an inhibitor of DPP-IV.
One aspect of the present invention pertains to methods of preparing compositions comprising the step of admixing a compound, selected from the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, with an inhibitor of DPP-IV, and a pharmaceutically acceptable carrier.
One aspect of the present invention pertains to methods of preparing compositions further comprising the step of preparing a unit dosage form from the composition.
One aspect of the present invention pertains to methods of preparing a dosage form of a pharmaceutical composition for the treatment of a GPRl 19 receptor related disorder in an individual, the pharmaceutical composition comprising a compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, and an inhibitor of DPP-IV; the method comprising:
(a) mixing the compound with a first pharmaceutically acceptable carrier to prepare a dosage form of the compound;
(b) mixing the inhibitor of DPP-IV with a second pharmaceutically acceptable carrier to prepare a dosage form of the inhibitor of DPP-IV; and
(c) providing the dosage forms as a combined preparation for simultaneous, separate, or sequential use.
One aspect of the present invention pertains to methods of preparing a dosage form of a pharmaceutical composition for the treatment of a GPRl 19 receptor related disorder selected from the group consisting of:
diabetes mellitus, type 1 diabetes, type 2 diabetes, inadequate glucose tolerance, impaired glucose tolerance, insulin resistance, hyperglycemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia, atherosclerosis, stroke, syndrome X, hypertension, pancreatic beta-cell insufficiency, enteroendocrine cell insufficiency, glucosuria, metabolic acidosis, cataracts, diabetic nephropathy, diabetic neuropathy, peripheral neuropathy, diabetic coronary artery disease, diabetic cerebrovascular disease, diabetic peripheral vascular disease, diabetic retinopathy, metabolic syndrome, a condition related to diabetes mellitus, myocardial infarction, learning impairment, memory impairment, a neurodegenerative disorder, a condition ameliorated by increasing a blood GLP- 1 level in an individual with a neurodegenerative disorder, excitotoxic brain damage caused by severe epileptic seizures, Alzheimer's disease,
Parkinson's disease, Huntington's disease, prion-associated disease, stroke, motor-neuron disease, traumatic brain injury, spinal cord injury, and obesity.
In some embodiments, the combined preparation is in the form of a twin pack.
In some embodiments, the first pharmaceutically acceptable carrier and the second pharmaceutically acceptable carrier in steps (a) and (b) are different.
In some embodiments, the first pharmaceutically acceptable carrier and the second pharmaceutically acceptable carrier in steps (a) and (b) are substantially the same.
In some embodiments, the combined preparation is for simultaneous use.
In some embodiments, the combined preparation is for separate use.
In some embodiments, the combined preparation is for sequential use.
One aspect of the present invention pertains to methods of preparing a dosage form of a pharmaceutical composition for increasing blood GLP- 1 level in an individual, the
pharmaceutical composition comprising a compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, and an inhibitor of DPP-IV; the method comprising:
(a) mixing the compound with a first pharmaceutically acceptable carrier to prepare a dosage form of the compound;
(b) mixing the inhibitor of DPP-IV with a second pharmaceutically acceptable carrier to prepare a dosage form of the inhibitor of DPP-IV; and
(c) providing the dosage forms as a combined preparation for simultaneous, separate, or sequential use.
In some embodiments, the combined preparation is in the form of a twin pack.
In some embodiments, the first pharmaceutically acceptable carrier and the second pharmaceutically acceptable carrier in steps (a) and (b) are different.
In some embodiments, the first pharmaceutically acceptable carrier and the second pharmaceutically acceptable carrier in steps (a) and (b) are substantially the same.
In some embodiments, the combined preparation is for simultaneous use.
In some embodiments, the combined preparation is for separate use.
In some embodiments, the combined preparation is for sequential use. In some embodiments, the amount of the compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, alone and the amount of the inhibitor of DPP-IV alone are therapeutically ineffective in increasing a blood GLP- 1 level in the individual.
One aspect of the present invention pertains to compounds of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, for use in combination with an inhibitor of DPP-IV for the treatment of a condition ameliorated by increasing a blood GLP- 1 level in a mammal.
In some embodiments, the amount of compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, alone is therapeutically ineffective for treatment of a condition ameliorated by increasing a blood GLP- 1 level in a mammal.
One aspect of the present invention pertains to an inhibitor of DPP-IV for use in combination with compounds of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, for the treatment of a condition ameliorated by increasing a blood GLP- 1 level in a mammal.
In some embodiments, the amount of the inhibitor of DPP-IV alone is therapeutically ineffective for treatment of a condition ameliorated by increasing a blood GLP- 1 level in a mammal.
In some embodiments, the condition ameliorated by increasing a blood GLP- 1 level is selected from the group consisting of:
diabetes mellitus, type 1 diabetes, type 2 diabetes, inadequate glucose tolerance, impaired glucose tolerance, insulin resistance, hyperglycemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia, atherosclerosis, stroke, syndrome X, hypertension, pancreatic beta-cell insufficiency, enteroendocrine cell insufficiency, glucosuria, metabolic acidosis, cataracts, diabetic nephropathy, diabetic neuropathy, peripheral neuropathy, diabetic coronary artery disease, diabetic cerebrovascular disease, diabetic peripheral vascular disease, diabetic retinopathy, metabolic syndrome, a condition related to diabetes mellitus, myocardial infarction, learning impairment, memory impairment, a neurodegenerative disorder, a condition ameliorated by increasing a blood GLP- 1 level in an individual with a neurodegenerative disorder, excitotoxic brain damage caused by severe epileptic seizures, Alzheimer's disease, Parkinson's disease, Huntington's disease, prion-associated disease, stroke, motor-neuron disease, traumatic brain injury, spinal cord injury, and obesity.
One aspect of the present invention pertains to pharmaceutical compositions comprising compounds of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, for use in combination with an inhibitor of DPP-IV for the treatment of a condition ameliorated by increasing a blood GLP- 1 level in a mammal.
In some embodiments, the amount of the compound alone is therapeutically ineffective for treatment of a condition ameliorated by increasing a blood GLP- 1 level in a mammal.
One aspect of the present invention pertains to pharmaceutical compositions comprising an inhibitor of DPP-IV for use in combination with compounds of Formula (I) and
pharmaceutically acceptable salts, solvates, and hydrates thereof for the treatment of a condition ameliorated by increasing a blood GLP- 1 level in a mammal.
In some embodiments, the condition ameliorated by increasing a blood GLP- 1 level is selected from the group consisting of:
diabetes mellitus, type 1 diabetes, type 2 diabetes, inadequate glucose tolerance, impaired glucose tolerance, insulin resistance, hyperglycemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia, atherosclerosis, stroke, syndrome X, hypertension, pancreatic beta-cell insufficiency, enteroendocrine cell insufficiency, glucosuria, metabolic acidosis, cataracts, diabetic nephropathy, diabetic neuropathy, peripheral neuropathy, diabetic coronary artery disease, diabetic cerebrovascular disease, diabetic peripheral vascular disease, diabetic retinopathy, metabolic syndrome, a condition related to diabetes mellitus, myocardial infarction, learning impairment, memory impairment, a neurodegenerative disorder, a neurodegenerative disorder, excitotoxic brain damage caused by severe epileptic seizures, Alzheimer's disease, Parkinson's disease, Huntington's disease, prion-associated disease, stroke, motor-neuron disease, traumatic brain injury, spinal cord injury, and obesity.
One aspect of the present invention pertains to dosage forms of a pharmaceutical composition as described herein.
One aspect of the present invention pertains to combined preparations comprising a compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, and an inhibitor of DPP-IV for simultaneous, separate or sequential use for the treatment of diabetes mellitus or a condition related thereto.
In some embodiments, the amount of the compound selected from compounds of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, alone and the amount of an inhibitor of DPP-IV are therapeutically ineffective in lowering blood glucose levels in a subject.
In some embodiments, the compounds and inhibitors of DPP-IV are admixed with pharmaceutically acceptable carriers. In some embodiments, the compound and the inhibitor of DPP-IV are admixed with different pharmaceutically acceptable carriers.
In some embodiments, the condition related to diabetes mellitus is selected from the group consisting of hyperglycemia, impaired glucose tolerance, insulin resistance, pancreatic beta-cell insufficiency, enteroendocrine cell insufficiency, glucosuria, metabolic acidosis, cataracts, diabetic nephropathy, diabetic neuropathy, diabetic retinopathy, diabetic coronary artery disease, diabetic cerebrovascular disease, diabetic peripheral vascular disease, metabolic syndrome, hyperlipidemia, atherosclerosis, stroke, hypertension, and obesity.
One aspect of the present invention pertains to twin packs comprising compounds of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, and inhibitors of DPP-IV as a combined preparation for the treatment of diabetes mellitus or a condition related thereto.
In some embodiments, the compound and the inhibitor of DPP-IV are admixed with different pharmaceutically acceptable carriers.
In some embodiments, the different pharmaceutically acceptable carriers are suitable for administration by different routes.
In some embodiments, the condition related to diabetes mellitus is selected from the group consisting of hyperglycemia, impaired glucose tolerance, insulin resistance, pancreatic beta-cell insufficiency, enteroendocrine cell insufficiency, glucosuria, metabolic acidosis, cataracts, diabetic nephropathy, diabetic neuropathy, diabetic retinopathy, diabetic coronary artery disease, diabetic cerebrovascular disease, diabetic peripheral vascular disease, metabolic syndrome, hyperlipidemia, atherosclerosis, stroke, hypertension, and obesity.
One aspect of the present invention pertains to kits comprising (a) a first package comprising a compound according to claim 1 or a pharmaceutical composition thereof, and (b) a second package comprising an inhibitor of DPP-IV.
These and other aspects of the invention disclosed herein will be set forth in greater detail as the patent disclosure proceeds. DETAILED DESCRIPTION OF THE INVENTION
It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination. Accordly, all combinations of uses and medical indications described herein specifically embraced by the present invention just as if each and every subcombination of uses and medical indications was individually and explicitly recited herein.
The present disclosure includes all isotopes of atoms occurring in the present compounds, salts and crystalline forms thereof. Isotopes include those atoms having the same atomic number but different mass numbers. By way of general example, and without limitation, isotopes of hydrogen include 2H (deuterium) and 3H (tritium). Isotopes of carbon include 13C and 14C.
The invention is described herein in detail using the terms defined below unless otherwise specified.
The term "agonist" is intended to mean a moiety that interacts and activates the receptor, such as, the GPRl 19 receptor and initiates a physiological or pharmacological response characteristic of that receptor. For example, when moieties activate the intracellular response upon binding to the receptor, or enhance GTP binding to membranes.
The term "composition" is intended to mean a material comprising at least two components; for example, and without limitation, a composition comprising a compound of the present invention and a pharmaceutically acceptable carrier.
The term "contact or contacting" is intended to mean bringing the indicated moieties together, whether in an in vitro system or an in vivo system. Thus, "contacting" a GPRl 19 receptor with a compound of the invention includes the administration of a compound of the present invention to an individual, preferably a human, having a GPRl 19 receptor, as well as, for example, introducing a compound of the invention into a sample containing a cellular or more purified preparation containing a GPRl 19 receptor.
The term "hydrate" as used herein means a compound of the invention or a salt thereof, that further includes a stoichiometric or non- stoichiometric amount of water bound by non- covalent intermolecular forces.
The terms "in need of treatment" and "in need thereof," when referring to treatment are used interchangeably to mean a judgment made by a caregiver (e.g. physician, nurse, nurse practitioner, etc. in the case of humans; veterinarian in the case of animals, including non-human mammals) that an individual or animal requires or will benefit from treatment. This judgment is made based on a variety of factors that are in the realm of a caregiver's expertise, but that includes the knowledge that the individual or animal is ill, or will become ill, as the result of a disease, condition or disorder that is treatable by the compounds of the invention. Accordingly, the compounds of the invention can be used in a protective or preventive manner; or compounds of the invention can be used to alleviate, inhibit or ameliorate the disease, condition or disorder. The term "individual" or "subject" is intended to mean any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates and most preferably humans. In another embodiment, the individual is a human and in certain embodiments, the human is an infant, child, adolescent or adult. In one embodiment, the individual is at risk for developing a GPRl 19-related disorder. In one embodiment, the individual is at risk for developing a metabolic-related disease or disorder. Individuals who are at risk include, but are not limited to, those with hereditary history of a metabolic-related disease or disorder, or those in a state of physical health which puts them at risk for a metabolic-related disease or disorder. In another embodiment, the individual has been determined, by the care-giver or someone acting under the guidance of the care-giver, to have a metabolic-related disease or disorder.
The term "modulate or modulating" is intended to mean an increase or decrease in the amount, quality, response or effect of a particular activity, function or molecule.
The term "pharmaceutical composition" is intended to mean a composition comprising at least one active ingredient; including but not limited to, salts, solvates, and hydrates of compounds of the present invention; whereby the composition is amenable to investigation for a specified, efficacious outcome in a mammal (for example, without limitation, a human). Those of ordinary skill in the art will understand and appreciate the techniques appropriate for determining whether an active ingredient has a desired efficacious outcome based upon the needs of the artisan.
The term "solvate" as used herein means a compound of the invention or a salt, thereof, that further includes a stoichiometric or non- stoichiometric amount of a solvent bound by non- covalent intermolecular forces. Preferred solvents are volatile, non-toxic, and/or acceptable for administration to humans in trace amounts.
The term "therapeutically effective amount" is intended to mean the amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue, system, animal, individual or human that is being sought by a researcher, veterinarian, medical doctor or other clinician or caregiver; or in an individual, which includes one or more of the following:
(1) Preventing the disease; for example, preventing a disease, condition or disorder in an individual that may be predisposed to the disease, condition or disorder but does not yet experience or display the pathology or symptomatology of the disease;
(2) Inhibiting the disease; for example, inhibiting a disease, condition or disorder in an individual that is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., arresting further development of the pathology and/or symptomatology); and
(3) Ameliorating the disease; for example, ameliorating a disease, condition or disorder in an individual that is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., reversing the pathology and/or symptomatology).
COMPOUNDS OF THE PRESENT INVENTION
The novel compound N-(2-fluoro-4-(methylsulfonyl)phenyl)-5-methyl-6-(l-(5- methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine (Compound 1) is a potent and selective agonist of GPRl 19. In the HTRF cAMP assay, N-(2-fluoro-4-(methylsulfonyl)phenyl)-5-methyl-
6-(l-(5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine is a "full agonist". The compound N-(2-fluoro-4-(methylsulfonyl)phenyl)-5-methyl-6-(l-(5-methylpyrazin-2- yl)piperidin-4-yloxy)pyrimidin-4-amine increased cAMP levels in CHO cells expressing human GPRl 19 (GDIR) with an observed EC50 of 6 nM (see Example IA). N-(2-fluoro-4- (methylsulfonyl)phenyl)-5-methyl-6-(l -(5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4- amine stimulated GLP-I release from mouse GLUTag cells with an observed EC50 of 54.1 nM (see Example IB) and is a potent and efficacious GPRl 19 agonist in normal Sprague-Dawley rats in vivo. In each of these models, N-(2-fluoro-4-(methylsulfonyl)phenyl)-5-methyl-6-(l-(5- methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine potently increased glucose clearance during the oral glucose tolerance test (oGTT) (see Example 2). Further, N-(2-fluoro-4-
(methylsulfonyl)phenyl)-5-methyl-6-(l-(5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4- amine significantly reduced blood glucose levels during the oral glucose tolerance test (see Example 3) at a dose of 1.5 mg/kg.
In recombinant CYP enzyme preparations, N-(2-fluoro-4-(methylsulfonyl)phenyl)-5- methyl-6-(l-(5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine inhibited 3A4, 2C9, and
2C19 with approximate IC50 values for inhibition of 9.05 μM, 8.82 μM, and >10 μM, respectively and showed no inhibition of 1 A2 and 2D6. In human liver microsome preparations, the approximate IC50 value of inhibition for 2C9 was 6 μM and for the other P450s tested (i.e., 2Cl 9 and 3A4) the IC5O values were >30μM (see Examples 4 and 5).
One aspect of the present invention pertains to compounds selected from the following compound N-(2-fluoro-4-(methylsulfonyl)phenyl)-5-methyl-6-(l-(5-methylpyrazin-2- yl)piperidin-4-yloxy)pyrimidin-4-amine (Compound 1), as shown in Formula (I):
Figure imgf000035_0001
(I)
and pharmaceutically acceptable salts, solvates, and hydrates thereof.
INDICATIONS AND METHODS OF TREATMENT
In addition to the foregoing beneficial uses for compounds of the present invention as disclosed herein, compounds of the invention are useful in the treatment of additional diseases. Without limitation, these include the following.
The most significant pathologies in type 2 diabetes are impaired insulin signaling at its target tissues ("insulin resistance") and failure of the insulin-producing cells of the pancreas to secrete an appropriate degree of insulin in response to a hyperglycemic signal. Current therapies to treat the latter include inhibitors of the β-cell ATP-sensitive potassium channel to trigger the release of endogenous insulin stores, or administration of exogenous insulin. Neither of these achieves accurate normalization of blood glucose levels and both carry the risk of inducing hypoglycemia. For these reasons, there has been intense interest in the development of pharmaceuticals that function in a glucose-dependent action, i.e. potentiators of glucose signaling.
Physiological signaling systems which function in this manner are well-characterized and include the gut peptides GLPl, GIP and PACAP. These hormones act via their cognate G-protein coupled receptor to stimulate the production of cAMP in pancreatic β-cells. The increased cAMP does not appear to result in stimulation of insulin release during the fasting or preprandial state. However, a series of biochemical targets of cAMP signaling, including the ATP-sensitive potassium channel, voltage-sensitive potassium channels and the exocytotic machinery, are modified in such a way that the insulin secretory response to a postprandial glucose stimulus is markedly enhanced. Accordingly, agonists of novel, similarly functioning, β-cell GPCRs, including GPRl 19, would also stimulate the release of endogenous insulin and consequently promote normoglycemia in type 2 diabetes.
It is also established that increased cAMP, for example as a result of GLP- 1 stimulation, promotes β-cell proliferation, inhibits β-cell death and thus improves islet mass. This positive effect on β-cell mass is expected to be beneficial in both type 2 diabetes, where insufficient insulin is produced, and type 1 diabetes, where β-cells are destroyed by an inappropriate autoimmune response. Some β-cell GPCRs, including GPRl 19, are also present in the hypothalamus where they modulate hunger, satiety, decrease food intake, controlling or decreasing weight and energy expenditure. Hence, given their function within the hypothalamic circuitry, agonists or inverse agonists of these receptors mitigate hunger, promote satiety and therefore modulate weight.
It is also well-established that metabolic diseases exert a negative influence on other physiological systems. Thus, there is often the co-development of multiple disease states (e.g. type 1 diabetes, type 2 diabetes, inadequate glucose tolerance, insulin resistance, hyperglycemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia, obesity or cardiovascular disease in "syndrome X") or diseases which clearly occur secondary to diabetes mellitus (e.g. kidney disease, peripheral neuropathy). Thus, it is expected that effective treatment of the diabetic condition will in turn be of benefit to such interconnected disease states.
In some embodiments of the present invention the metabolic-related disorder is selected from the group consisting of type 2 diabetes, hyperglycemia, hyperinsulinemia, hyperlipidemia, hypertriglyceridemia, insulin resistance, type 1 diabetes, idiopathic type 1 diabetes (type Ib), latent autoimmune diabetes in adults (LADA), early-onset type 2 diabetes (EOD), youth-onset atypical diabetes (YOAD), maturity onset diabetes of the young (MODY), malnutrition-related diabetes, gestational diabetes, cardiovascular disease, coronary heart disease, vascular restenosis, restenosis, restenosis after angioplasty, peripheral vascular disease, claudication, intermittent claudication, cell death associated with myocardial infarction (e.g. necrosis and apoptosis), dyslipidemia, post-prandial lipemia, conditions of impaired glucose tolerance (IGT), impaired glucose metabolism, conditions of impaired glucose metabolism, conditions of impaired fasting plasma glucose, metabolic acidosis, ketosis, arthritis, obesity, osteoporosis, hypertension, congestive heart failure, left ventricular hypertrophy, peripheral arterial disease, diabetic retinopathy, macular degeneration, cataract, diabetic nephropathy, glomerulosclerosis, chronic renal failure, diabetic neuropathy, metabolic syndrome, syndrome X, premenstrual syndrome, angina pectoris, thrombosis, atherosclerosis, ischemic stroke, transient ischemic attacks, stroke, erectile dysfunction, skin and connective tissue disorders, foot ulcerations, ulcerative colitis, endothelial dysfunction, and impaired vascular compliance. COMPOSITIONS AND SALTS
Another aspect of the present invention pertains to compositions comprising a compound selected from N-(2-fluoro-4-(methylsulfonyl)phenyl)-5-methyl-6-(l -(5-methylpyrazin-2- yl)piperidin-4-yloxy)pyrimidin-4-amine (Compound 1), and pharmaceutically acceptable salts, solvates, and hydrates thereof. A further aspect of the present invention pertains to compositions comprising a compound selected from N-(2-fluoro-4-(methylsulfonyl)phenyl)-5-methyl-6-(l-(5-methylpyrazin- 2-yl)piperidin-4-yloxy)pyrimidin-4-amine (Compound 1), and pharmaceutically acceptable salts, solvates, and hydrates thereof, and one or more pharmaceutically acceptable carriers.
A further aspect of the present invention pertains to pharmaceutical compositions comprising a compound selected from N-(2-fluoro-4-(methylsulfonyl)phenyl)-5-methyl-6-(l-(5- methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine (Compound 1), and pharmaceutically acceptable salts, solvates, and hydrates thereof, and one or more pharmaceutically acceptable carriers.
Some embodiments of the present invention pertain to compositions comprising N-(2- fluoro-4-(methylsulfonyl)phenyl)-5-methyl-6-(l-(5-methylpyrazin-2-yl)piperidin-4- yloxy)pyrimidin-4-amine and a pharmaceutically acceptable carrier.
Some embodiments of the present invention include a method of producing a
pharmaceutical composition comprising admixing N-(2-fluoro-4-(methylsulfonyl)phenyl)-5- methyl-6-(l-(5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier.
Compositions may be prepared by any suitable method, typically by uniformly mixing the active compound with liquids or finely divided solid carriers, or both, in the required proportions, and then, if necessary, forming the resulting mixture into a desired shape.
Conventional excipients, such as binding agents, fillers, acceptable wetting agents, tabletting lubricants, and disintegrants may be used in tablets and capsules for oral administration. Liquid preparations for oral administration may be in the form of solutions, emulsions, aqueous or oily suspensions, and syrups. Alternatively, the oral preparations may be in the form of dry powder that can be reconstituted with water or another suitable liquid vehicle before use.
Additional additives such as suspending or emulsifying agents, non-aqueous vehicles (including edible oils), preservatives, and flavorings and colorants may be added to the liquid preparations. Parenteral dosage forms may be prepared by dissolving the compound of the invention in a suitable liquid vehicle and filter sterilizing the solution before filling and sealing an appropriate vial or ampoule. These are just a few examples of the many appropriate methods well known in the art for preparing dosage forms.
A compound of the present invention can be formulated into pharmaceutical
compositions using techniques well known to those in the art. Suitable pharmaceutically- acceptable carriers, outside those mentioned herein, are known in the art; for example, see Remington, The Science and Practice of Pharmacy, 20th Edition, 2000, Lippincott Williams & Wilkins, (Editors: Gennaro, A. R., et al.).
While it is possible that, for use in the treatment, a compound of the invention can, in an alternative use, be administered as a raw or pure chemical, it is preferable however to present the compound or active ingredient as a pharmaceutical formulation or composition further comprising a pharmaceutically acceptable carrier.
The invention thus further provides pharmaceutical compositions comprising a compound of the invention or a pharmaceutically acceptable salt or derivative thereof together with one or more pharmaceutically acceptable carriers thereof and/or prophylactic ingredients. The carrier(s) must be "acceptable" in the sense of being compatible with the other ingredients of the composition and not overly deleterious to the recipient thereof.
Pharmaceutical compositions include those suitable for oral, rectal, nasal, topical (including buccal and sub-lingual), vaginal or parenteral (including intramuscular, sub-cutaneous and intravenous) administration or in a form suitable for administration by inhalation, insufflation or by a transdermal patch. Transdermal patches dispense a drug at a controlled rate by presenting the drug for absorption in an efficient manner with a minimum of degradation of the drug.
Typically, transdermal patches comprise an impermeable backing layer, a single pressure sensitive adhesive and a removable protective layer with a release liner. One of ordinary skill in the art will understand and appreciate the techniques appropriate for manufacturing a desired efficacious transdermal patch based upon the needs of the artisan.
The compounds of the invention, together with a conventional adjuvant, carrier, or diluent, may thus be placed into the form of pharmaceutical compositions and unit dosages thereof, and in such form may be employed as solids, such as tablets or filled capsules, or liquids such as solutions, suspensions, emulsions, elixirs, gels or capsules filled with the same, all for oral use, in the form of suppositories for rectal administration; or in the form of sterile injectable solutions for parenteral (including subcutaneous) use. Such pharmaceutical compositions and unit dosage forms thereof may comprise conventional ingredients in conventional proportions, with or without additional active compounds or principles, and such unit dosage forms may contain any suitable effective amount of the active ingredient commensurate with the intended daily dosage range to be employed.
For oral administration, the pharmaceutical composition may be in the form of, for example, a tablet, capsule, suspension or liquid. The pharmaceutical composition is preferably made in the form of a dosage unit containing a particular amount of the active ingredient.
Examples of such dosage units are capsules, tablets, powders, granules or a suspension, with conventional additives such as lactose, mannitol, corn starch or potato starch; with binders such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins; with disintegrators such as corn starch, potato starch or sodium carboxymethyl-cellulose; and with lubricants such as talc or magnesium stearate. The active ingredient may also be administered by injection as a composition wherein, for example, saline, dextrose or water may be used as a suitable pharmaceutically acceptable carrier.
The dose when using the compounds of the present invention can vary within wide limits, and as is customary and known to the physician, it is to be tailored to the individual conditions in each individual case. It depends, for example, on the nature and severity of the illness to be treated, on the condition of the patient, on the compound employed or on whether an acute or chronic disease state is treated or prophylaxis is conducted or on whether further active compounds are administered in addition to the compounds of the present invention. For example, doses of the present invention include, but are not limited to, about 0.001 mg to about 5000 mg, about 0.001 to about 2500 mg, about 0.001 to about 1000 mg, 0.001 to about 500 mg, 0.001 mg to about 250 mg, about 0.001 mg to 100 mg, about 0.001 mg to about 50 mg, and about 0.001 mg to about 25 mg. The desired dose may conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example, as two, three, four or more sub-doses per day. The sub-dose itself may be further divided, e.g., into a number of discrete loosely spaced administrations. Depending on the individual and as deemed appropriate from the patient's physician or care-giver it may be necessary to deviate upward or downward from the doses described herein.
The amount of a compound of the invention, or pharmaceutically acceptable salt thereof, required for use in treatment will vary not only with the particular salt selected but also with the route of administration, the nature of the condition being treated and the age and condition of the patient and will ultimately be at the discretion of the attendant physician or clinician. In general, one skilled in the art understands how to extrapolate in vivo data obtained in a model system, typically an animal model, to another, such as a human. Typically, animal models include, but are not limited to, the rodent diabetes model as described in Example 1 , infra (as well as other animal models known in the art, such as those reported by Reed and Scribner in Diabetes, Obesity and Metabolism, 1, 1999, 75-86). In some circumstances, these extrapolations may merely be based on the weight of the animal in the respective model in comparison to another, such as a mammal, preferably a human, however, more often, these extrapolations are not simply based on weights, but rather incorporate a variety of factors. Representative factors include, but are not limited to, the age, weight, sex, diet and medical condition of the patient, the severity of the disease, the route of administration, pharmacological considerations such as the activity, efficacy, pharmacokinetic and toxicology profiles of the particular compound employed, whether a drug delivery system is utilized, on whether an acute or chronic disease state is being treated or prophylaxis is conducted or on whether further active compounds are administered in addition to the compounds of the present invention and as part of a drug combination. The dosage regimen for treating a disease condition with the compounds and/or compositions of this invention is selected in accordance with a variety of factors as cited above. Thus, the actual dosage regimen employed may vary widely and therefore may deviate from a preferred dosage regimen and one skilled in the art will recognize that a dosage and a dosage regimen outside these typical ranges can be tested and, where appropriate, may be used in the methods of this invention.
The compounds of the present invention can be administered in a wide variety of oral and parenteral dosage forms. It will be obvious to those skilled in the art that the following dosage forms may comprise, as the active component, either a compound of the invention or a pharmaceutically acceptable salt of a compound of the invention.
For preparing pharmaceutical compositions from the compounds of the present invention, the selection of a suitable pharmaceutically acceptable carrier can be either solid, liquid or a mixture of both. Solid form preparations include powders, tablets, pills, capsules, cachets, suppositories, and dispersible granules. A solid carrier can be one or more substances which may also act as diluents, flavoring agents, solubilizers, lubricants, suspending agents, binders, preservatives, tablet disintegrating agents, or an encapsulating material.
In powders, the carrier is a finely divided solid which is in a mixture with the finely divided active component.
In tablets, the active component is mixed with the carrier having the necessary binding capacity in suitable proportions and compacted to the desired shape and size.
The powders and tablets may contain varying percentage amounts of the active compound. A representative amount in a powder or tablet may contain from 0.5 to about 90 percent of the compound of the invention; however, an artisan would know when amounts outside of this range are necessary. Suitable carriers for powders and tablets are magnesium carbonate, magnesium stearate, talc, sugar, lactose, pectin, dextrin, starch, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose, a low melting wax, cocoa butter, and the like.
The term "preparation" is intended to include the formulation of the active compound with encapsulating material as carrier providing a capsule in which the active component, with or without carriers, is surrounded by a carrier, which is thus in association with it. Similarly, cachets and lozenges are included. Tablets, powders, capsules, pills, cachets, and lozenges can be used as solid forms suitable for oral administration.
For preparing suppositories, a low melting wax, such as an admixture of fatty acid glycerides or cocoa butter, is first melted and the active component is dispersed homogeneously therein, as by stirring. The molten homogenous mixture is then poured into convenient sized molds, allowed to cool, and thereby to solidify.
Compositions suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or sprays containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
Liquid form preparations include solutions, suspensions, and emulsions, for example, water or water-propylene glycol solutions. For example, parenteral injection liquid preparations can be formulated as solutions in aqueous polyethylene glycol solution. Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables.
The compounds according to the present invention may thus be formulated for parenteral administration (e.g. by injection, for example bolus injection or continuous infusion) and may be presented in unit dose form in ampoules, pre-filled syringes, small volume infusion or in multi- dose containers with an added preservative. The pharmaceutical 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. Alternatively, the active ingredient may be in powder form, obtained by aseptic isolation of sterile solid or by lyophilization from solution, for constitution with a suitable vehicle, e.g. sterile, pyrogen- free water, before use.
Aqueous compositions suitable for oral use can be prepared by dissolving or suspending the active component in water and adding suitable colorants, flavors, stabilizing and thickening agents, as desired. Aqueous suspensions suitable for oral use can be made by dispersing the finely divided active component in water with viscous material, such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, or other well known suspending agents.
Also included are solid form preparations which are intended to be converted, shortly before use, to liquid form preparations for oral administration. Such liquid forms include solutions, suspensions, and emulsions. These preparations may contain, in addition to the active component, colorants, flavors, stabilizers, buffers, artificial and natural sweeteners, dispersants, thickeners, solubilizing agents, and the like.
For topical administration to the epidermis the compounds according to the invention may be formulated as ointments, creams or lotions, or as a transdermal patch.
Ointments and creams may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agents. Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilizing agents, dispersing agents, suspending agents, thickening agents, or coloring agents.
Compositions suitable for topical administration in the mouth include lozenges comprising the active agent in a flavored base, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert base such as gelatin and glycerin or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
Solutions or suspensions are applied directly to the nasal cavity by conventional means, for example with a dropper, pipette or spray. The compositions may be provided in single or multi-dose form. In the latter case of a dropper or pipette, this may be achieved by the patient administering an appropriate, predetermined volume of the solution or suspension. In the case of a spray, this may be achieved for example by means of a metering atomizing spray pump.
Administration to the respiratory tract may also be achieved by means of an aerosol composition in which the active ingredient is provided in a pressurized pack with a suitable propellant. If the compounds of the present invention or pharmaceutical compositions comprising them are administered as aerosols, for example as nasal aerosols or by inhalation, this can be carried out, for example, using a spray, a nebulizer, a pump nebulizer, an inhalation apparatus, a metered inhaler or a dry powder inhaler. Pharmaceutical forms for administration of the compounds of the present invention as an aerosol can be prepared by processes well-known to the person skilled in the art. For their preparation, for example, solutions or dispersions of the compounds of the present invention in water, water/alcohol mixtures or suitable saline solutions can be employed using customary additives, for example benzyl alcohol or other suitable preservatives, absorption enhancers for increasing the bioavailability, solubilizers, dispersants and others, and, if appropriate, customary propellants, for example include carbon dioxide, CFCs, such as, dichlorodifluoromethane, trichlorofluoromethane, or dichlorotetrafluoroethane; and the like. The aerosol may conveniently also contain a surfactant such as lecithin. The dose of drug may be controlled by provision of a metered valve.
In compositions intended for administration to the respiratory tract, including intranasal compositions, the compound will generally have a small particle size for example of the order of 10 microns or less. Such a particle size may be obtained by means known in the art, for example by micronization. When desired, compositions adapted to give sustained release of the active ingredient may be employed.
Alternatively the active ingredients may be provided in the form of a dry powder, for example, a powder mix of the compound in a suitable powder base such as lactose, starch, starch derivatives such as hydroxypropylmethyl cellulose and polyvinylpyrrolidone (PVP).
Conveniently the powder carrier will form a gel in the nasal cavity. The powder composition may be presented in unit dose form for example in capsules or cartridges of, e.g., gelatin, or blister packs from which the powder may be administered by means of an inhaler.
The pharmaceutical preparations are preferably in unit dosage forms. In such form, the preparation can be subdivided into unit doses containing appropriate quantities of the active component. The unit dosage form can be a packaged preparation, the package containing discrete quantities of preparation, such as packeted tablets, capsules, and powders in vials or ampoules. Also, the unit dosage form can be a capsule, tablet, cachet, or lozenge itself, or it can be the appropriate number of any of these in packaged form.
Tablets or capsules for oral administration and liquids for intravenous administration are preferred compositions.
The compounds according to the invention may optionally exist as pharmaceutically acceptable salts including pharmaceutically acceptable acid addition salts prepared from pharmaceutically acceptable non-toxic acids including inorganic and organic acids.
The acid addition salts may be obtained as the direct product of compound synthesis. In the alternative, the free base may be dissolved in a suitable solvent containing the appropriate acid, and the salt isolated by evaporating the solvent or otherwise separating the salt and solvent. The compounds of this invention may form solvates with standard low molecular weight solvents using methods known to the skilled artisan.
It is noted that when the GPRl 19 agonist and pharmaceutically acceptable salts, solvates and hydrates thereof are utilized as active ingredients in a composition, such as a pharmaceutical composition, that they are not necessarily intended for use in only humans, but in other non- human mammals as well. Indeed, recent advances in the area of animal health-care indicate that consideration be given for the use of active agents, such as GPRl 19 receptor modulators, for the treatment of obesity in domestic animals (e.g., cats and dogs), and GPRl 19 receptor modulators in other animals, such as livestock, where no disease or disorder is evident (e.g., food-oriented animals such as cows, chickens, fish, etc.). Those of ordinary skill in the art are readily credited with understanding the utility of such compounds in such settings.
PHARMACEUTICALLY ACCEPTABLE SALTS, SOLVATES, AND HYDRATES
It is understood that when the phrase "pharmaceutically acceptable salts, solvates and hydrates" is used when referring to N-(2-fluoro-4-(methylsulfonyl) phenyl)-5-methyl-6-(l -(5- methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine, it is intended to embrace:
1) Pharmaceutically acceptable salts of N-(2-fluoro-4-(methylsulfonyl) phenyl)-5-methyl- 6-(l-(5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine;
2) Solvates of N-(2-fluoro-4-(methylsulfonyl) phenyl)-5-methyl-6-(l-(5-methylpyrazin-2- yl)piperidin-4-yloxy)pyrimidin-4-amine and pharmaceutically acceptable salts thereof; and
3) Hydrates of N-(2-fluoro-4-(methylsulfonyl) phenyl)-5-methyl-6-(l-(5-methylpyrazin- 2-yl)piperidin-4-yloxy)pyrimidin-4-amine, and pharmaceutically acceptable salts thereof.
The compounds of the present invention can be administrated in a wide variety of oral and parenteral dosage forms. It will be apparent to those skilled in the art that the following dosage forms may comprise, as the active component, a compound selected from the compound
N-(2-fluoro-4-(methylsulfonyl) phenyl)-5-methyl-6-(l-(5-methylpyrazin-2-yl)piperidin-4- yloxy)pyrimidin-4-amine (Compound 1) and pharmaceutically acceptable salts, solvates, and hydrates thereof. Moreover, various hydrates and solvates of the compounds of the invention and their salts will find use as intermediates in the manufacture of pharmaceutical compositions. Typical procedures for making and identifying suitable hydrates and solvates, outside those mentioned herein, are well known to those in the art; see for example, pages 202-209 of KJ. Guillory, "Generation of Polymorphs, Hydrates, Solvates, and Amorphous Solids," in:
Polymorphism in Pharmaceutical Solids , ed. Harry G. Brittan, Vol. 95, Marcel Dekker, Inc., New York, 1999, incorporated herein by reference in its entirety. Accordingly, one aspect of the present invention pertains to hydrates and solvates of N-(2-fluoro-4-(methylsulfonyl) phenyl)-5- methyl-6-(l -(5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine and pharmaceutical acceptable salts, that can be isolated and characterized by one or more of the methods known in the art, such as, thermogravimetric analysis (TGA), TGA-mass spectroscopy, TGA-Infrared spectroscopy, powder X-ray diffraction (XRPD), Karl Fisher titration, high resolution X-ray diffraction, and the like. There are several commercial entities that provide quick and efficient services for identifying solvates and hydrates on a routine basis. Example companies offering these services include Wilmington PharmaTech (Wilmington, DE), Avantium Technologies (Amsterdam), and Aptuit (Greenwich, CT).
POLYMOPHS AND PSEUDOPOLYMORPHS
Polymorphism is the ability of a substance to exist as two or more crystalline phases that have different arrangements and/or conformations of the molecules in the crystal lattice.
Polymorphs show the same properties in the liquid or gaseous state but they behave differently in the solid state.
Besides single-component polymorphs, drugs can also exist as salts and other multicomponent crystalline phases. For example, solvates and hydrates may contain an API host and either solvent or water molecules, respectively, as guests. Analogously, when the guest compound is a solid at room temperature, the resulting form is often called a cocrystal. Salts, solvates, hydrates, and cocrystals may show polymorphism as well. Crystalline phases that share the same API host, but differ with respect to their guests, may be referred to as
pseudopolymorphs of one another.
Solvates contain molecules of the solvent of crystallization in a definite crystal lattice. Solvates, in which the solvent of crystallization is water, are termed hydrates. Because water is a constituent of the atmosphere, hydrates of drugs may be formed rather easily.
By way of example, Stahly recently published a polymorph screen of 245 compounds consisting of a "wide variety of structural types" that revealed about 90% of the compounds exhibiting multiple solid forms. Overall, approximately half the compounds were polymorphic, often having one to three forms. About one-third of the compounds formed hydrates, and about one-third formed solvates. Data from cocrystal screens of 64 compounds showed that 60% formed cocrystals other than hydrates or solvates. (G. P. Stahly, Crystal Growth & Design (2007), 7(6), 1007-1026.)
COMBINATION THERAPY
In the context of the present invention, a compound as described herein or a
pharmaceutical composition thereof can be utilized for modulating the activity of GPRl 19 receptor related diseases, conditions and/or disorders as described herein. Examples of modulating the activity of GPRl 19 receptor related diseases include the treatment of metabolic related disorders. Metabolic related disorders include, but are not limited to, hyperlipidemia, type 1 diabetes, type 2 diabetes, and conditions associated therewith, such as, but not limited to coronary heart disease, ischemic stroke, restenosis after angioplasty, peripheral vascular disease, claudication, intermittent claudication, cell death associated with myocardial infarction (e.g. necrosis and apoptosis), dyslipidemia, post-prandial lipemia, conditions of impaired glucose tolerance (IGT), conditions of impaired fasting plasma glucose, metabolic acidosis, ketosis, arthritis, obesity, osteoporosis, hypertension, congestive heart failure, left ventricular
hypertrophy, peripheral arterial disease, diabetic retinopathy, macular degeneration, cataract, diabetic nephropathy, glomerulosclerosis, chronic renal failure, diabetic neuropathy, metabolic syndrome, syndrome X, premenstrual syndrome, coronary heart disease, angina pectoris, thrombosis, atherosclerosis, myocardial infarction, transient ischemic attacks, stroke, vascular restenosis, hyperglycemia, hyperinsulinemia, hyperlipidemia, hypertriglyceridemia, insulin resistance, impaired glucose metabolism, conditions of impaired glucose tolerance, conditions of impaired fasting plasma glucose, obesity, erectile dysfunction, skin and connective tissue disorders, foot ulcerations, ulcerative colitis, endothelial dysfunction and impaired vascular compliance. In some embodiments, metabolic related disorders include type 1 diabetes, type 2 diabetes, inadequate glucose tolerance, insulin resistance, hyperglycemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia and syndrome X. Other examples of modulating the activity of GPRl 19 receptor related diseases include the treatment of obesity and/or overweight by decreasing food intake, inducing satiation (i.e., the feeling of fullness), controlling weight gain, decreasing body weight and/or affecting metabolism such that the recipient loses weight and/or maintains weight.
While a compound of the invention can be administered as the sole active pharmaceutical agent (i.e., mono-therapy), the compound can also be used in combination with one or more pharmaceutical agents (i.e., combination-therapy) either administered together or separately for the treatment of the diseases / conditions / disorders described herein. Therefore, another aspect of the present invention includes methods of treatment of a metabolic related disorder, including a weight related disorder, such as obesity, comprising administering to an individual in need of prophylaxis and/or treatment a therapeutically effective amount of a compound of the present invention in combination with one or more additional pharmaceutical agent as described herein.
Suitable pharmaceutical agents that can be used in combination with the compounds of the present invention include anti-obesity agents such as apolipoprotein-B secretion/microsomal triglyceride transfer protein (apo-B/MTP) inhibitors, MCR-4 agonists, cholescystokinin-A (CCK- A) agonists, serotonin and norepinephrine reuptake inhibitors (for example, sibutramine), sympathomimetic agents, β3 adrenergic receptor agonists, dopamine agonists (for example, bromocriptine), melanocyte-stimulating hormone receptor analogues, cannabinoid 1 receptor antagonists [for example, SR141716: N-(piperidin-l-yi)-5-(4-chlorophenyl)-l-(2,4- dichlorophenyl)-4-methyl-lH-pyrazole-3-carboxamide], melanin concentrating hormone antagonists, leptons (the OB protein), leptin analogues, leptin receptor agonists, galanin antagonists, lipase inhibitors (such as tetrahydrolipstatin, i.e., Orlistat), anorectic agents (such as a bombesin agonist), Neuropeptide-Y antagonists, thyromimetic agents, dehydroepiandrosterone or an analogue thereof, glucocorticoid receptor agonists or antagonists, orexin receptor antagonists, urocortin binding protein antagonists, glucagon- like peptide- 1 receptor agonists, ciliary neutrotrophic factors (such as Axokine™ available from Regeneron Pharmaceuticals, Inc., Tarrytown, NY and Procter & Gamble Company, Cincinnati, OH), human agouti-related proteins
(AGRP), ghrelin receptor antagonists, histamine 3 receptor antagonists or reverse agonists, neuromedin U receptor agonists, noradrenergic anorectic agents (for example, phentermine, mazindol and the like) and appetite suppressants (for example, bupropion).
Other anti-obesity agents, including the agents set forth infra, are well known, or will be readily apparent in light of the instant disclosure, to one of ordinary skill in the art.
In some embodiments, the anti-obesity agents are selected from the group consisting of orlistat, sibutramine, bromocriptine, ephedrine, leptin, and pseudoephedrine. In a further embodiment, compounds of the present invention and combination therapies are administered in conjunction with exercise and/or a sensible diet.
It is understood that the scope of combination-therapy of the compounds of the present invention with other anti-obesity agents, anorectic agents, appetite suppressant and related agents is not limited to those listed above, but includes in principle any combination with any pharmaceutical agent or pharmaceutical composition useful for the treatment of overweight and obese individuals.
It is understood that the scope of combination-therapy of the compounds of the present invention with other pharmaceutical agents is not limited to those listed herein, supra or infra, but includes in principle any combination with any pharmaceutical agent or pharmaceutical composition useful for the treatment of diseases, conditions or disorders that are linked to metabolic related disorders.
Some embodiments of the present invention include methods of treatment of a disease, disorder, condition or complication thereof as described herein, comprising administering to an individual in need of such treatment a therapeutically effective amount or dose of a compound of the present invention in combination with at least one pharmaceutical agent selected from the group consisting of: sulfonylureas (for example, glyburide, glipizide, glimepiride and other sulfonylureas known in the art), meglitinides (for example, repaglinide, nateglinide and other meglitinides known in the art), biguanides (for example, biguanides include phenformin, metformin, buformin, and biguanides known in the art), α-glucosidase inhibitors [for example, acarbose, N-(l,3-dihydroxy-2-propyl)valiolamine (generic name; voglibose), miglitol, and α- glucosidase inhibitors known in the art], peroxisome proliferators-activated receptor-γ (i.e.,
PPAR-γ) agonists (for example, rosiglitazone, pioglitazone, tesaglitazar, netoglitazone, GW- 409544, GW-501516 and PPAR-γ agonists known in the art), insulin, insulin analogues, HMG- CoA reductase inhibitors (for example, rosuvastatin, pravastatin and its sodium salt, simvastatin, lovastatin, atorvastatin, fluvastatin, cerivastatin, rosuvastatin, pitavastatin, BMS's "superstatin", and HMG-CoA reductase inhibitors known in the art), cholesterol-lowering drugs (for example, fibrates that include: bezafibrate, beclobrate, binifibrate, ciplofibrate, clinofibrate, clofibrate, clofibric acid, etofibrate, fenofibrate, gemfibrozil, nicofibrate, pirifibrate, ronifibrate, simfibrate, theofibrate, and fibrates known in the art; bile acid sequestrants which include: cholestyramine, colestipol and the like; and niacin), antiplatelet agents (for example, aspirin and adenosine diphosphate receptor antagonists that include: clopidogrel, ticlopidine and the like), angiotensin- converting enzyme inhibitors (for example, captopril, enalapril, alacepril, delapril; ramipril, lisinopril, imidapril, benazepril, ceronapril, cilazapril, enalaprilat, fosinopril, moveltopril, perindopril, quinapril, spirapril, temocapril, trandolapril, and angiotensin converting enzyme inhibitors known in the art), angiotensin II receptor antagonists [for example, losartan (and the potassium salt form)], angiotensin II receptor antagonists known in the art, adiponectin, squalene synthesis inhibitors (for example, (S)-α-[bis[2,2-dimethyl-l-oxopropoxy)methoxy] phosphinyl]- 3-phenoxybenzenebutanesulfonic acid, mono potassium salt (BMS- 188494) and squalene synthesis inhibitors known in the art), and the like. In some embodiments, compounds of the present invention and the pharmaceutical agents are administered separately. In further embodiments, compounds of the present invention and the pharmaceutical agents are
administered together.
Suitable pharmaceutical agents that can be used in conjunction with compounds of the present invention include, but not limited to, amylin agonists (for example, pramlintide), insulin secretagogues (for example, GLP-I agonists; exendin-4; insulinotropin (NN2211); acyl CoA cholesterol acetyltransferase inhibitors (for example, ezetimibe, eflucimibe, and like compounds), cholesterol absorption inhibitors (for example, ezetimibe, pamaqueside and like compounds), cholesterol ester transfer protein inhibitors (for example, CP-529414, JTT-705, CETi-I, and like compounds), microsomal triglyceride transfer protein inhibitors (for example, implitapide, and like compounds), cholesterol modulators (for example, NO- 1886, and like compounds), bile acid modulators (for example, GT103-279 and like compounds), insulin signaling pathway modulators, like inhibitors of protein tyrosine phosphatases (PTPases), non-small molecule mimetic compounds and inhibitors of glutamine-fructose-6-phosphate amidotransferase (GFAT), compounds influencing a dysregulated hepatic glucose production, like inhibitors of glucose-6- phosphatase (GόPase), inhibitors of fructose- 1,6-bisphosphatase (F-l,6-BPase), inhibitors of glycogen phosphorylase (GP), glucagon receptor antagonists and inhibitors of
phosphoenolpyruvate carboxykinase (PEPCK), pyruvate dehydrogenase kinase (PDHK) inhibitors, insulin sensitivity enhancers, insulin secretion enhancers, inhibitors of gastric emptying, α2-adrenergic antagonists, retinoid X receptor (PvXR) agonists, and dipeptidyl peptidase-4 (DPP-IV) inhibitors.
DIPEPTIDYL PEPTIDASE IV (DPP-IV) INHIBITORS
Dipeptidyl peptidase IV (DPP-IV, EC 3.4.14.5) exhibits catalytic activity against a broad range of peptide substrates that includes peptide hormones, neuropeptides, and chemokines. The incretins glucagon- like peptide 1 (GLP-I) and glucose-dependent insulinotropic polypeptide
(GIP), which stimulate glucose-dependent insulin secretion and otherwise promote blood glucose homeostasis, are rapidly cleaved by DPP-IV at the position 2 alanine leading to inactivation of their biological activity. Both pharmacological and genetic attenuation of DPP-IV activity is associated with enhanced incretin action, increased insulin, and lower blood glucose in vivo. Genetic attenuation of DPP-IV activity has been shown to provide resistance to obesity and to improve insulin sensitivity. Inhibitors of DPP-IV have shown to be useful as theraputics, for example, oral administration of vildagliptin (l-[2-(3-hydroxyadamant-l- ylamino)acetyl]pyrrolidine-2(S)-carbonitrile) or sitagliptin (3(R)-ammo-l-[3-(trifluoromethyl)- 5,6,7,8-tetrahydro[l,2,4]triazolo[4,3-a]pyrazin-7-yl]-4-(2,4,5-trifluorophenyl) butan-1-one) to human patients suffering with type 2 diabetes has been found to reduce fasting glucose and postprandial glucose excursion in association with significantly reduced HbAic levels. For reviews on the application of DPP-IV inhibitors for the treatment of Type 2 diabetes, reference is made to the following publications: (1) H. -U. Demuth, et al., "Type 2 diabetes-Therapy with dipeptidyl peptidase IV inhibitors," Biochim. Biophys. Acta, 1751: 33-44 (2005), and (2) K. Augustyns, et al., "Inhibitors of pro line-specific dipeptidyl peptidases: DPP-IV inhibitors as a novel approach for the treatment of Type 2 diabetes," Expert Opin. Ther. Patents, 15: 1387-1407 (2005).
Accordingly, suitable pharmaceutical agents include inhibitors of DPP-IV that can be used in conjunction with compounds of the present invention either dosed separately or together. Inhibitors of DPP-IV are well in the art or can be readily identified and their in vitro biological activity determined using any number of methods available, for example, O'Brien, M., Daily, B., Schurria, M., "Assay for DPPIV activity using a homogeneous, luminescent method," Cell Notes, Issue 11, 2005; see also the DPPIV-Glo™ Protease Assay Technical Bulletin #TB339.
Examples of DPP-IV inhibitors are described in Villhauer et al., J Med Chem (2003)
46:2774-2789, for LAF237; Ahren et al, J Clin Endocrinol Metab (2004) 89:2078-2084;
Villhauer et al., J Med Chem (2002) 45:2362-2365 for NVP-DPP728; Ahren et al, Diabetes Care (2002) 25:869-875 for NVP-DPP728; Peters et al., Bioorg Med Chem Lett (2004) 14: 1491-1493; Caldwell et al., Bioorg Med Chem Lett (2004) 14:1265-1268; Edmondson et al., Bioorg Med Chem Lett (2004) 14:5151-5155; and Abe et al., J Nat Prod (2004) 67:999-1004.
Specific examples of DPP-IV inhibitors include, but are not limited to, dipeptide derivatives or dipeptide mimetics such as alanine-pyrrolidide, isoleucine-thiazolidide, and the pseudosubstrate N-valyl prolyl, O-benzoyl hydroxylamine, as described e.g. in U.S. Pat. No. 6,303,661.
Some embodiments of the present invention include every combination of one or more
DPP-IV inhibitors selected from the DPP-IV inhibitors found in U.S. Pat. Nos. 6,869,947, 6,867,205, 6,861,440, 6,849,622, 6,812,350, 6,803,357, 6,800,650, 6,727,261, 6,716,843, 6,710,040, 6,706,742, 6,645,995, 6,617,340, 6,699,871, 6,573,287, 6,432,969, 6,395,767, 6,380,398, 6,303,661, 6,242,422, 6,166,063, 6,100,234, and 6,040,145.
Some embodiments of the present invention include every combination of one or more
DPP-IV inhibitors selected from the DPP-IV inhibitors found in U.S. Patent Application
Numbers: 2005059724, 2005059716, 2005043292, 2005038020, 2005032804, 2005004205, 2004259903, 2004259902, 2004259883, 2004254226, 2004242898, 2004229926, 2004180925, 2004176406, 2004138214, 2004116328, 2004110817, 2004106656, 2004097510, 2004087587, 2004082570, 2004077645, 2004072892, 2004063935, 2004034014, 2003232788, 2003225102,
2003216450, 2003216382, 2003199528, 2003195188, 2003162820, 2003149071, 2003134802, 2003130281, 2003130199, 2003125304, 2003119750, 2003119738, 2003105077, 2003100563, 2003087950, 2003078247, 2002198205, 2002183367, 2002103384, 2002049164, and
2002006899.
Some embodiments of the present invention include every combination of one or more
DPP-IV inhibitors selected from the DPP-IV inhibitors found in International Patent Application Publication Numbers: WO 2005/087235, WO 2005/082348, WO 2005/082849, WO
2005/079795, WO 2005/075426, WO 2005/072530, WO 2005/063750, WO 2005/058849, WO 2005/049022, WO 2005/047297, WO 2005/044195, WO 2005/042488, WO 2005/040095, WO 2005/037828, WO 2005/037779, WO 2005/034940, WO 2005/033099, WO 2005/032590, WO 2005/030751, WO 2005/030127, WO 2005/026148, WO 2005/025554, WO 2005/023762, WO 2005/020920, WO 05/19168, WO 05/12312, WO 05/12308, WO 05/12249, WO 05/11581, WO 05/09956, WO 05/03135, WO 05/00848, WO 05/00846, WO 04/112701, WO 04/111051, WO 04/111041, WO 04/110436, WO 04/110375, WO 04/108730, WO 04/104216, WO 04/104215,
WO 04/103993, WO 04/103276, WO 04/99134, WO 04/96806, WO 04/92128, WO 04/87650, WO 04/87053, WO 04/85661, WO 04/85378, WO 04/76434, WO 04/76433, WO 04/71454, WO 04/69162, WO 04/67509, WO 04/64778, WO 04/58266, WO 04/52362, WO 04/52850, WO 04/50022, WO 04/50658, WO 04/48379, WO 04/46106, WO 04/43940, WO 04/41820, WO 04/41795, WO 04/37169, WO 04/37181, WO 04/33455, WO 04/32836, WO 04/20407, WO
04/18469, WO 04/18468, WO 04/18467, WO 04/14860, WO 04/09544, WO 04/07468, WO 04/07446, WO 04/04661, WO 04/00327, WO 03/106456, WO 03/104229, WO 03/101958, WO 03/101448, WO 03/99279, WO 03/95425, WO 03/84940, WO 03/82817, WO 03/80633, WO 03/74500, WO 03/72556, WO 03/72528, WO 03/68757, WO 03/68748, WO 03/57666, WO 03/57144, WO 03/55881, WO 03/45228, WO 03/40174, WO 03/38123, WO 03/37327, WO
03/35067, WO 03/35057, WO 03/24965, WO 03/24942, WO 03/22871, WO 03/15775, WO 03/04498, WO 03/04496, WO 03/02530, WO 03/02596, WO 03/02595, WO 03/02593, WO 03/02553, WO 03/02531, WO 03/00181, WO 03/00180, WO 03/00250, WO 02/83109, WO 02/83128, WO 02/76450, WO 02/68420, WO 02/62764, WO 02/55088, WO 02/51836, WO 02/38541, WO 02/34900, WO 02/30891, WO 02/30890, WO 02/14271, WO 02/02560, WO
01/97808, WO 01/96295, WO 01/81337, WO 01/81304, WO 01/68603, WO 01/55105, WO 01/52825, WO 01/34594, WO 00/71135, WO 00/69868, WO 00/56297, WO 00/56296, WO 00/34241, WO 00/23421, WO 00/10549, WO 99/67278, WO 99/62914, WO 99/61431, WO 99/56753, WO 99/25719, WO 99/16864, WO 98/50066, WO 98/50046, WO 98/19998, WO 98/18763, WO 97/40832, WO 95/29691, WO 95/15309, WO 93/10127, WO 93/08259, and WO
91/16339.
Some embodiments of the present invention include every combination of one or more DPP-IV inhibitors selected from the DPP-IV inhibitors found in Patent Publication Numbers: EP 1517907, EP 1513808, EP 1492777, EP 1490335, EP 1489088, EP 1480961, EP 1476435, EP 1476429, EP 1469873, EP 1465891, EP 1463727, EP 1461337, EP 1450794, EP 1446116, EP
1442049, EP 1441719, EP 1426366, EP 1412357, EP1406873, EP 1406872, EP 1406622, EP 1404675, EP 1399420, EP 1399471, EP 1399470, EP 1399469, EP 1399433, EP 1399154, EP 1385508, EP 1377288, EP 1355886, EP 1354882, EP 1338592, EP 1333025, EP 1304327, EP 1301187, EP 1296974, EP 1280797, EP 1282600, EP 1261586, EP 1258476, EP 1254113, EP 1248604, EP 1245568, EP 1215207, EP 1228061, EP 1137635, EP 1123272, EP 1104293, EP 1082314, EP 1050540, EP 1043328, EP 0995440, EP 0980249, EP 0975359, EP 0731789, EP 0641347, EP 0610317, EP 0528858, CA 2466870, CA 2433090, CA 2339537, CA 2289125, CA 2289124, CA 2123128, DD 296075, DE 19834591, DE 19828113, DE 19823831, DE 19616486, DE 10333935, DE 10327439, DE 10256264, DE 10251927, DE 10238477, DE 10238470, DE
10238243, DE 10143840, FR 2824825, FR 2822826, JP2005507261, JP 2005505531, JP 2005502624, JP 2005500321, JP 2005500308, JP2005023038, JP 2004536115, JP 2004535445, JP 2004535433, JP 2004534836, JP 2004534815, JP 2004532220, JP 2004530729, JP
2004525929, JP 2004525179, JP 2004522786, JP 2004521149, JP 2004503531, JP 2004315496, JP 2004244412, JP 2004043429, JP 2004035574, JP 2004026820, JP 2004026678, JP
2004002368, JP 2004002367, JP 2003535898, JP 2003535034, JP 2003531204, JP 2003531191, JP 2003531118, JP 2003524591, JP 2003520849, JP 2003327532, JP 2003300977, JP
2003238566, JP 2002531547, JP 2002527504, JP 2002517401, JP 2002516318, JP 2002363157, JP 2002356472, JP 2002356471, JP 2002265439, JP 2001510442, JP 2000511559, JP
2000327689, JP 2000191616, JP 1998182613, JP 1998081666, JP 1997509921, JP 1995501078, and JP 1993508624.
In some embodiments, the DPP-IV inhibitor has an IC50 of less than about 10 μM, less than about 1 μM, less than about 100 nM, less than about 75 nM, less than about 50 nM, less than about 25 nM, less than about 20 nM, less than about 15 nM, less than about 10 nM, less than about 5 nM, less than about 4 nM, less than about 3 nM, less than about 2 nM, or less than about
1 nM. In some embodiments, the DPP-IV inhibitor has an IC5O of less than about 50 nM, less than about 25 nM, less than about 20 nM, less than about 15 nM, less than about 10 nM, less than about 5 nM, less than about 4 nM, less than about 3 nM, less than about 2 nM, or less than about I nM.
In some embodiments, the DPP-IV inhibitor is a selective DPP-IV inhibitor, wherein the selective DPP-IV inhibitor has a selectivity for human plasma DPP-IV over one or more of the following enzymes, post proline cleaving enzyme (PPCE), dipeptidyl peptidase II (DPPII), dipeptidyl peptidase-8 (DPP-8), and dipeptidyl peptidase-9 (DPP-9) of at least about 10-fold, more preferably of at least about 100-fold, and most preferably of at least about 1000-fold.
In some embodiments, the DPP-IV inhibitor is orally active.
In some embodiments, the DPP-IV inhibitor is an inhibitor of human DPP-IV.
Some embodiments of the present invention include every combination of one or more compounds selected from: 3(R)-amino-l-[3-(trifluoromethyl)-5,6,7,8-tetrahydro[l,2,4]triazolo[4,3-a]pyrazin-7-yl]- 4-(2,4,5-trifluorophenyl) butan- 1 -one; 1 -[2-(3-hydroxyadamant- 1 -ylamino)acetyl]pyrrolidine- 2(S)-carbonitrile; (lS,3S,5S)-2-[2(S)-amino-2-(3-hydroxyadamantan-l-yl)acetyl]-2- azabicyclo[3.1.0]hexane-3-carbonitrile; 2-[6-[3(R)-aminopiperidin-l-yl]-3-methyl-2,4-dioxo- l,2,3,4-tetrahydropyrimidin-l-ylmethyl]benzonitrile; 8-[3(R)-ammopiperidin-l-yi]-7-(2-butynyl)-
3-methyl- 1 -(4-methylquinazolin-2-ylmethyl)xanthine; 1 -[N-[3(R)-pyrrolidinyl]glycyl]pyrrolidin- 2(R)-yl boronic acid; 4(S>fluoro-l-[2-[(lR,3S)-3-(lH-l,2,4-triazol-l- ylmethyl)cyclopentylammo]acetyl]pyrrolidine-2(S)-carbonitrile; l-[(2S,3S,l lbS)-2-amino-9,10- dimethoxy-2,3,4,6,7, 1 lb-hexahydro- lH-pyrido[2, 1 -a]isoquinolin-3-yl]-4(S)- (fluoromethyl)pyrrolidin-2-one; (2S,4S)-2-cyano-4-fluoro- 1 -[(2-hydroxy- 1 , 1 -dimethyl) ethylamino]acetylpyrrolidine; 8-(czs-hexahydro-pyrrolo[3,2-b]pyrrol-l-yl)-3-methyl-7-(3-methyl- but-2-enyl)-l-(2-oxo-2-phenylethyl)-3,7-dihydro-purine-2,6-dione; l-((3£,45)-4-amino-l-(4-(3,3- difluoropyrrolidin- 1 -yl)- 1 ,3,5-triazin-2-yl)pyrrolidin-3-yl)-5,5difluoropiperidin-2-one; (R)-2-((6- (3-aminopiperidin- 1 -yl)-3-methyl-2,4-dioxo-3,4-dihydropyrimidin- 1 (2H)-yl)methyl)-4- fluorobenzonitrile; 5- {(S)-2-[2-((S)-2-cyano-pyrrolid in- 1 -yl)-2-oxo-ethylamino]-propyl} -5-(1H- tetrazol-5-yl)10,l l-dihydro-5H-dibenzo[a,d]cycloheptene-2,8-dicarboxylic acid bis- dimethylamide; ((2S, 4S)-4-(4-(3 -methyl- 1 -phenyl- lH-pyrazol-5-yl)piperazin- 1 -yl)pyrrolidin-2- yl)(thiazolidin-3-yl)methanone; (2S,AS)- 1 -[2-[(4-ethoxycarbonylbicyclo[2.2.2]oct- 1 - yl)amino]acetyl]-4-fluoropyrrolidine-2-carbonitrile; 6-[(3R)-3-amino-piperidin-l-yl]-5-(2-chloro- 5-fluoro-benzyl)-l,3-dimethyl-l,5dihydro-pyrrolo[3,2-d]pyrimidine-2,4-dione; 2-({6-[(3R)-3- amino-3-methylpiperidin- 1 -yl]- 1 ,3-dimethyl-2,4-dioxo- l,2,3,4-tetrahydro-5H-pyrrolo[3,2- d]pyrimidin-5-yl}methyl)-4-fluorobenzonitrile; (2S)-l-{[2-(5-methyl-2-phenyl-oxazol-4-yl)- ethylamino]-acetyl} -pyrrolidine-2-carbonitrile; (2S)- 1 - {[1 , 1 -dimethyl-3-(4-pyridin-3-yl-imidazol- 1 -yl)-propylamino]-acetyl} -pyrrolidine-2-carbonitrile; (3,3-difluoropyrrolidin- 1 -y\)-((2S,AS)-A- (4-(pyrimidin-2yl)piperazin-l-yl)pyrrolidin-2-yl)methanone; (2S,4S)-l-[(2S)-2-amino-3,3-bis(4- fluorophenyl)propanoyl]-4-fluoropyrrolidine-2-carbonitrile; (2S,5R)-5-ethynyl- 1 - {N-(4-methyl- 1 - (4-carboxy-pyridin-2-yl)piperidin-4-yl)glycyl}pyrrolidine-2-carbonitrile; and (\S,6R)-2>- {[3- (trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl]carbonyl}-6-(2,4,5- trifluorophenyl)cyclohex-3-en-l -amine; and pharmaceutically acceptable salts, solvates and hydrates thereof.
Some embodiments of the present invention include every combination of one or more compounds selected from:
3(R)-amino-l-[3-(trifluoromethyl)-5,6,7,8-tetrahydro[l,2,4]triazolo[4,3-a]pyrazin-7-yl]- 4-(2,4,5-trifluorophenyl) butan- 1-one; l-[2-(3-hydroxyadamant-l-ylamino)acetyl]pyrrolidine- 2(S)-carbonitrile; (lS,3S,5S)-2-[2(S)-amino-2-(3-hydroxyadamantan-l-yl)acetyl]-2- azabicyclo[3.1.0]hexane-3-carbonitrile; 2-[6-[3(R)-aminopiperidin-l-yl]-3-methyl-2,4-dioxo- l,2,3,4-tetrahydropyrimidin-l-ylmethyl]benzonitrile; 8-[3(R)-ammopiperidin-l-yi]-7-(2-butynyl)- 3-methyl- 1 -(4-methylquinazolin-2-ylmethyl)xanthine; 1 -[N-[3(R)-pyrrolidinyl]glycyl]pyrrolidin- 2(R)-yl boronic acid; 4(S>fluoro-l-[2-[(lR,3S)-3-(lH-l,2,4-triazol-l- ylmethyl)cyclopentylammo]acetyl]pyrrolidine-2(S)-carbonitrile; l-[(2S,3S,l lbS)-2-amino-9,10- dimethoxy-2,3,4,6,7, 1 lb-hexahydro- lH-pyrido[2, 1 -a]isoquinolin-3-yl]-4(S)- (fluoromethyl)pyrrolidin-2-one; (2S,4S)-2-cyano-4-fluoro- 1 -[(2-hydroxy- 1 , 1 -dimethyl) ethylamino]acetylpyrrolidine; 8-(czs-hexahydro-pyrrolo[3,2-b]pyrrol-l-yl)-3-methyl-7-(3-methyl- but-2-enyl)-l-(2-oxo-2-phenylethyl)-3,7-dihydro-purine-2,6-dione; l-((3S,4S)-4-amino-l-(4-(3,3- difluoropyrrolidin- 1 -yl)- 1 ,3,5-triazin-2-yl)pyrrolidin-3-yl)-5,5difluoropiperidin-2-one; (R)-2-((6- (3-aminopiperidin- 1 -yl)-3-methyl-2,4-dioxo-3,4-dihydropyrimidin- 1 (2H)-yl)methyl)-4- fluorobenzonitrile; 5-{(S)-2-[2-((S)-2-cyano-pyrrolid in-l-yl)-2-oxo-ethylamino]-propyl}-5-(lΗ- tetrazol-5-yl)10,l l-dihydro-5H-dibenzo[a,d]cycloheptene-2,8-dicarboxylic acid bis- dimethylamide; ((2S, 4S)-4-(4-(3 -methyl- 1 -phenyl- lH-pyrazol-5-yl)piperazin- 1 -yl)pyrrolidin-2- yl)(thiazolidin-3-yl)methanone; (2S,AS)- 1 -[2-[(4-ethoxycarbonylbicyclo[2.2.2]oct- 1 - yl)amino]acetyl]-4-fluoropyrrolidine-2-carbonitrile; 6-[(3R)-3-amino-piperidin-l-yl]-5-(2-chloro- 5-fluoro-benzyl)-l,3-dimethyl-l,5dihydro-pyrrolo[3,2-d]pyrimidine-2,4-dione; 2-({6-[(3R)-3- amino-3-methylpiperidin- 1 -yl]- 1 ,3-dimethyl-2,4-dioxo- 1 ,2,3,4-tetrahydro-5H-pyrrolo[3,2- d]pyrimidin-5-yl}methyl)-4-fluorobenzonitrile; (2S)-l-{[2-(5-methyl-2-phenyl-oxazol-4-yl)- ethylamino] -acetyl} -pyrrolidine-2-carbonitrile; (2S)- 1 - {[1 , 1 -dimethyl-3-(4-pyridin-3-yl-imidazol- 1 -yl)-propylamino] -acetyl} -pyrrolidine-2-carbonitrile; and (3,3-difluoropyrrolidin- 1 -yl)-((2S,4S)- 4-(4-(pyrimidin-2yl)piperazin-l-yl)pyrrolidin-2-yl)methanone; and pharmaceutically acceptable salts, solvates and hydrates thereof.
Some embodiments of the present invention include every combination of one or more compounds selected from: (2S,4S)-l-[(2S)-2-amino-3,3-bis(4-fluorophenyl)propanoyl]-4- fluoropyrrolidine-2-carbonitrile; (2S,5R)-5-ethynyl- 1 - {N-(4-methyl- 1 -(4-carboxy-pyridin-2- yl)piperidin-4-yl)glycyl}pyrrolidine-2-carbonitrile; and (\S,6R)-?>- {[3-(trifluoromethyl)-5,6- dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl]carbonyl}-6-(2,4,5-trifluorophenyl)cyclohex-3-en- 1 -amine; and pharmaceutically acceptable salts, solvates and hydrates thereof.
Sitagliptin phosphate (Januvia, MK-0431, dihydrogenphosphate salt of 3(R)-amino-l-[3- (trifluoromethyl)-5,6,7,8-tetrahydro[l,2,4]triazolo[4,3-a]pyrazin-7-yl]-4-(2,4,5-trifluorophenyl) butan-1-one) is marketed by Merck & Co. for once-daily oral treatment of type 2 diabetes.
Januvia was first launched in Mexico followed by commercialization in the U.S. In 2007, the product was approved by the European Medicines Evaluation Agency (EMEA) and is currently available in the U.K., Germany and Spain. In 2009, Januvia was approved and launched in Japan. In addition, Merck has also filed for approval of Januvia in the U.S. as an adjunct to diet and exercise and in combination with other therapies to improve glycemic control in the treatment of diabetes. The compound, 3(R)-amino-l-[3-(trifluoromethyl)-5,6,7,8-tetrahydro[l,2,4]triazolo[4,3- a]pyrazin-7-yl]-4-(2,4,5-trifluorophenyl) butan-1-one, and pharmaceutically acceptable salts thereof are disclosed in international patent publication WO2003/004498. Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in WO2003/004498 and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the DPP-IV inhibitor of the present invention is selected from 3(R)-amino-l-[3-(trifluoromethyl)-5,6,7,8-tetrahydro[l,2,4]triazolo[4,3-a]pyrazin- 7-yl]-4-(2,4,5-trifluorophenyl) butan-1-one, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000055_0001
In some embodiments, the dipeptidyl peptidase IV (DPP4) inhibitor is 3(R)-amino-l-[3- (trifluoromethyl)-5,6,7,8-tetrahydro[l,2,4]triazolo[4,3-a]pyrazin-7-yl]-4-(2,4,5-trifluorophenyl) butan-1-one phosphate:
Figure imgf000055_0002
A crystalline form of 3(R)-amino-l -[3-(trifluoromethyl)-5, 6,7,8- tetrahydro[l,2,4]triazolo[4,3-a]pyrazin-7-yl]-4-(2,4,5-trifluorophenyl) butan- 1 -one phosphate salt monohydrate is disclosed in international patent publication WO2005/0031335 (incorporated by reference in its entirety). In some embodiments, the dipeptidyl peptidase IV (DPP4) inhibitor is crystalline 3(R)-amino-l-[3-(trifluoromethyl)-5,6,7,8-tetrahydro[l,2,4]triazolo[4,3-a]pyrazin-7- yl]-4-(2,4,5-trifluorophenyl) butan-1-one phosphate monohydrate.
Vildagliptin (Galvus, LAF-237, l-[2-(3-hydroxyadamant-l-ylamino)acetyl]pyrrolidine- 2(1S)-carbonitrile) is another dipeptidyl-peptidase IV (DPP-IV) inhibitor and was first commercialized in Brazil and Mexico by Novartis for oral, once-daily treatment of type 2 diabetes. In 2008, a marketing authorization application (MAA) was approved in the E.U. for this indication and launch took place in the U.K. in March, 2008. An approvable letter has been received for the regulatory application filed in the U.S. Approval in Japan took place in 2010. The compound, l-[2-(3-hydroxyadamant-l-ylamino)acetyl]pyrrolidine-2(S)-carbonitrile, is disclosed in international patent publication WO2000/034241. Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in WO2000/034241 and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the DPP-IV inhibitor of the present invention is selected from 1- [2-(3-hydroxyadamant- 1 -ylamino)acetyl]pyrrolidine-2(S)-carbonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000056_0001
Certain salts of the compound, l-[2-(3-hydroxyadamant-l-ylamino)acetyl]pyrrolidine- 2(1S)-carbonitrile, are disclosed in international patent publication WO2007/019255. In some embodiments, the DPP-IV inhibitor is l-[2-(3-hydroxyadamant-l-ylamino)acetyl]pyrrolidine-
2(S)-carbonitrile HCl:
Figure imgf000056_0002
Saxagliptin (Onglyza, BMS-477118, (l£,3£,5S)-2-[2(S)-amino-2-(3-hydroxyadamantan- l-yl)acetyl]-2-azabicyclo[3.1.0]hexane-3-carbonitrile) is another DPP-IV inhibitor and was launched in 2009 by AstraZeneca and Bristol-Myers Squibb in the U.S. for the treatment of type
2 diabetes. In 2009, the product was also approved in the E.U. for the treatment of type 2 diabetes independently and in combination with metformin. Phase III clinical studies are ongoing in Japan for the treatment of type 2 diabetes. The compound, (lS,3S,5S)-2-[2(S)-amino-2-(3- hydroxyadamantan-l-yl)acetyl]-2-azabicyclo[3.1.0]hexane-3-carbonitrile, is disclosed in international patent publication WO2001/068603. Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in WO2001/068603 and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the DPP-IV inhibitor of the present invention is selected from (lS,3S,5S)-2-[2(S)-amino-2-(3-hydroxyadamantan-l-yl)acetyl]-2-azabicyclo[3.1.0]hexane-3- carbonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000057_0001
SYR-322 (Alogliptin, 2-[6-[3(R)-aminopiperidin-l-yl]-3-methyl-2,4-dioxo-l,2,3,4- tetrahydropyrimidin- 1 -ylmethyl]benzonitrile) is a DPP-IV inhibitor which regulatory approval has been filed in Japan and the U.S. by Takeda for the once-daily, oral treatment of type 2 diabetes. The compound, 2-[6-[3(R)-ammopiperidin-l-yi]-3-methyl-2,4-dioxo-l,2,3,4- tetrahydropyrimidin-l-ylmethyl]benzonitrile, and pharmaceutically acceptable salts thereof are disclosed in international patent publication WO 2005/095381. Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in WO 2005/095381 and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the DPP-IV inhibitor of the present invention is selected from 2-[6-[3(R)-aminopiperidin- 1 -yl]-3-methyl-2,4-dioxo- 1 ,2,3,4-tetrahydropyrimidin- 1 - ylmethyljbenzonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000057_0002
The crystalline form of 2-[6-[3(R)-aminopiperidin-l-yi]-3-methyl-2,4-dioxo-l,2,3,4- tetrahydropyrimidin- 1 -ylmethyljbenzonitrile is disclosed in international patent publication
WO2007/035372 (incorporated by reference in its entirety). In some embodiments, the DPP-IV inhibitor is 2-[6-[3(R)-aminopiperidin- 1 -yl]-3-methyl-2,4-dioxo- 1,2,3,4-tetrahydropyrimidin- 1 - ylmethyljbenzonitrile benzoate:
Figure imgf000057_0003
BI- 1356 (Ondero, 8-[3(R)-aminopiperidin- 1 -yl]-7-(2-butynyl)-3-methyl- 1 -(4- methylquinazolin-2-ylmethyl)xanthine) is a DPP-IV inhibitor and is in phase III clinical development at Boehringer Ingelheim to evaluate its potential as an add-on therapy to metformin for the treatment of type 2 diabetes. The compound, 8-[3(R)-aminopiperidin-l-yl]-7-(2-butynyl)- 3 -methyl- l-(4-methylquinazolin-2-ylmethyl)xanthine, is disclosed in international patent publication WO2004/018468. Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in WO2004/018468 and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the DPP-IV inhibitor of the present invention is selected from 8-[3(R)- aminopiperidin- 1 -yl]-7-(2-butynyl)-3-methyl- 1 -(4-methylquinazolin-2-ylmethyl)xanthine, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000058_0001
Certain polymorphs of the compound, 8-[3(R)-aminopiperidm-l-yi]-7-(2-butynyl)-3-methyl-l-(4- methylquinazolin-2-ylmethyl)xanthine, are disclosed in international patent publication WO 2007/128721. In some embodiments, the DPP-IV inhibitor is a crystalline form of 8-[3(R)- aminopiperidin- 1 -yl]-7-(2-butynyl)-3-methyl- 1 -(4-methylquinazolin-2-ylmethyl)xanthine.
PHX-1149 (Dutogliptin, l-[N-[3(R)-pyrrolidinyl]glycyl]pyrrolidin-2(R)-yl boronic acid) is a DPP-IV inhibitor in phase III clinical trials by Phenomix and Forest for the oral, once-daily treatment of type 2 diabetes. The compound, l-[N-[3(R)-pyrrolidinyl]glycyl] pyrrolidin-2(R)-yl boronic acid, and pharmaceutically acceptable salts are disclosed in international patent publication WO2005/047297. Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in WO2005/047297 and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the DPP-IV inhibitor of the present invention is selected from 1-[N- [3 (R)- pyrrolidinyl]glycyl]pyrrolidin-2(R)-yl boronic acid, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000058_0002
The crystalline form of l-[N-[3(R)-pyrrolidinyl]glycyl]pyrrolidin-2(R)-yl boronic acid tartrate is disclosed in international patent publication WO2008/027273. In some embodiments, the DPP-IV inhibitor is l-[N-[3(R)-pyrrolidinyl]glycyl]pyrrolidin-2(R)-yl boronic acid tartrate:
Figure imgf000058_0003
GRC-8200 (Melogliptin, 4(5)-fluoro-l-[2-[(lΛ,35)-3-(lH-l,2,4-triazol-l- ylmethyl)cyclopentylammo]acetyl]pyrrolidine-2(S)-carbonitrile) is a DPP-IV inhibitor and is currently undergoing phase II clinical trials by Glenmark Pharmaceuticals and Merck KGaA for the treatment of type 2 diabetes. The compound, 4(5)-fluoro-l-[2-[(lR,35)-3-(lH-l,2,4-triazol-l- ylmethyl)cyclopentylammo]acetyl]pyrrolidine-2(S)-carbonitrile, is disclosed in international patent publication WO2006/040625. Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in WO2006/040625 and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the DPP-IV inhibitor of the present invention is selected from 4(S)-fluoro-l-[2- [(lR,3S)-3-(lH-l,2,4-triazol-l-ylmethyl)cyclopentylamino]acetyl]pyrrolidine-2(S)-carbonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000059_0001
R- 1579 (Carmegliptin, l-[(2S,3S,l lbS)-2-amino-9,10-dimethoxy-2,3,4,6,7,l Ib- hexahydro-lH-pyrido[2,l-a]isoquinolin-3-yl]-4(S)-(fluoromethyl)pyrrolidin-2-one) is a DPP-IV inhibitor. The compound, l-[(2S,3S,l lb£)-2-amino-9,10-dimethoxy-2,3,4,6,7,l lb-hexahydro-lH- pyrido[2,l-a]isoqumoh'n-3-yl]-4(S)-(fluoromethyl)pyrroh'din-2-one, is disclosed in international patent publication WO2005/000848. Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in WO2005/000848 and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the DPP-IV inhibitor of the present invention is selected from l-[(2S,3S,l lbS)-2- amino-9,10-dimethoxy-2,3,4,6,7,l lb-hexahydro-lH-pyrido[2,l-a]isoquinolin-3-yl]-4(S)- (fluoromethyl)pyrrolidin-2-one, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000059_0002
Taisho disclosed (2S,4S)-2-cyano-4-fluoro- 1 -[(2-hydroxy- 1 , 1 -dimethyl)
ethylamino]acetylpyrrolidine as a DPP-IV inhibitor. The compound, (2S,4S)-2-cyano-4-fluoro-l- [(2-hydroxy-l,l-dimethyl)ethylamino]acetylpyrrolidine, is disclosed in US publication US 2007/0112059. Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in US 2007/0112059 and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the DPP- IV inhibitor of the present invention is selected from (2S,4S)-2-cyano-4-fluoro-l-[(2-hydroxy- l,l-dimethyl)ethylamino]acetylpyrrolidine, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000060_0001
Sanofi-Aventis disclosed a series of substituted bicyclic 8-pyrrolidineoxanthine derivatives as DPP-IV inhibitors in US publication US 2007/0167468 (incorporated by reference in its entirety). Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in US publication 2007/0167468 and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the DPP-IV inhibitor of the present invention is selected from 8-(cw-hexahydro-pyrrolo[3,2-b]pyrrol- 1 -yl)-3-methyl-7-(3-methyl-but-2-enyl)- 1 -(2-oxo-2-phenylethyl)-3,7-dihydro-purine-2,6-dione, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000060_0002
Pfizer disclosed a series of 3-amino-pyrrolidine-4-lactam derivatives as DPP-IV inhibitors in international patent publication WO2007/148185. Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in WO2007/148185 and pharmaceutically acceptable salts, solvates, and hydrates thereof. One such compound is l-((3S,4S)-4-ammo-l-(4-(3,3-difluoropyrroridin-l-yl)- l,3,5-triazin-2-yl)pyrrolidin-3-yl)-5,5difluoropiperidin-2-one, and pharmaceutically acceptable salts thereof. In some embodiments, the DPP-IV inhibitor of the present invention is selected from 1 -((3S,4S)-4-amino- 1 -(4-(3,3-difluoropyrrolidin- 1 -yl)- 1 ,3,5-triazin-2-yl)pyrrolidin-3-yl)- 5,5difluoropiperidin-2-one, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000061_0001
Syrrx disclosed a series of substituted pyrimidine-2,4(lH,3H)-dione derivatives as DPP- IV inhibitors in international patent publication WO2005/095381. Some embodiments of the present invention include every combination of one or more compounds selected from the DPP- IV inhibitors disclosed in WO2005/095381 and pharmaceutically acceptable salts, solvates, and hydrates thereof. One such compound is (R)-2-((6-(3-aminopiperidm-l-yl)-3-methyl-2,4-dioxo- 3,4-dihydropyrimidin-l(2H)-yl)methyl)-4-fluorobenzonitrile, and pharmaceutically acceptable salts thereof. In some embodiments, the DPP-IV inhibitor of the present invention is selected from (R)-2-((6-(3-aminopiperidin- 1 -yl)-3-methyl-2,4-dioxo-3,4-dihydropyrimidin- 1 (2H)- yl)methyl)-4-fluorobenzonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000061_0002
Various crystalline forms of (R)-2-((6-(3-ammopiperidin-l-yl)-3-methyl-2,4-dioxo-3,4- dihydropyrimidin- 1 (2H)-yl)methyl)-4-fluorobenzonitrile succinic acid salt have been disclosed in international patent publication WO2005/0031335. One embodiment of the present invention pertains to any one or more crystalline forms of (R)-2-((6-(3-aminopiperidin-l-yl)-3-methyl-2,4- dioxo-3,4-dihydropyrimidin-l(2H)-yl)methyl)-4-fluorobenzonitrile succinic acid salt as described in international patent publication WO2005/0031335. In some embodiments, the dipeptidyl peptidase IV (DPP4) inhibitor is crystalline (R)-2-((6-(3-ammopiperidin-l-yl)-3-methyl-2,4- dioxo-3,4-dihydropyrimidin-l(2H)-yl)methyl)-4-fluorobenzonitrile succinic acid salt:
Figure imgf000061_0003
Alantos disclosed a series of substituted 2-cyano-pyrrolidine derivatives as DPP-IV inhibitors in international patent publication WO2006/116157. Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in WO2006/116157 and pharmaceutically acceptable salts, solvates, and hydrates thereof. One such compound is 5-{(S)-2-[2-((S)-2-cyano-pyrrolid in-l-yl)-2-oxo- ethylamino] -propyl} -5-(lH-tetrazol-5-yl) 10, 11 -dihydro-5H-dibenzo[a,d]cycloheptene-2,8- dicarboxylic acid bis-dimethylamide, and pharmaceutically acceptable salts thereof. In some embodiments, the DPP-IV inhibitor of the present invention is selected from 5- {(S)-2-[2-((S)-2- cyano-pyrrolid in- 1 -yl)-2-oxo-ethylamino]-propyl} -5-(lH-tetrazol-5-yl) 10, 11 -dihydro-5H- dibenzo[a,d]cycloheptene-2,8-dicarboxylic acid bis-dimethylamide, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000062_0001
Mitsubishi disclosed a series of 2,4-disubstituted pyrrolidine derivatives as DPP-IV inhibitors in international patent publication WO2002/0014271. Some embodiments of the present invention include every combination of one or more compounds selected from the DPP- IV inhibitors disclosed in WO2002/0014271 and pharmaceutically acceptable salts, solvates, and hydrates thereof. One such compound is ((2S,4S)-4-(4-(3-methyl- 1 -phenyl- lH-pyrazol-5- yl)piperazin-l-yl)pyrrolidin-2-yl)(thiazolidin-3-yl)methanone, and pharmaceutically acceptable salts thereof. In some embodiments, the DPP-IV inhibitor of the present invention is selected from ((2S,4S)-4-(4-(3-methyl- 1 -phenyl- lH-pyrazol-5-yl)piperazin- 1 -yl)pyrrolidin-2- yl)(thiazolidin-3-yl)methanone, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000062_0002
Various crystalline forms of ((2S,4S)-4-(4-(3-methyl-l -phenyl- lH-pyrazol-5- yl)piperazin-l-yl)pyrrolidin-2-yl)(thiazolidin-3-yl)methanone salts have been disclosed in international patent publication WO2006/088129 and US publication 2009/0216016. One embodiment of the present invention pertains to any one or more of the crystalline forms of ((2S, 4S)-4-(4-(3 -methyl- 1 -phenyl- lH-pyrazol-5-yl)piperazin- 1 -yl)pyrrolidin-2-yl)(thiazolidin-3- yl)methanone salt described in international patent publication WO2006/088129 and US publication 2009/0216016. In some embodiments, the DPP-IV inhibitor is crystalline ((2S,4S)-4- (4-(3 -methyl- 1 -phenyl- lH-pyrazol-5-yl)piperazin-l-yl)pyrrolidin-2-yl)(thiazolidin-3- yl)methanone 2.5 hydrobromide salt:
Figure imgf000063_0001
or a mono or a dihydrate thereof. In some embodiments, the dipeptidyl peptidase IV (DPP4) inhibitor is crystalline ((2S,45)-4-(4-(3-methyl-l-phenyl-lH-pyrazol-5-yl)piperazin-l- yl)pyrrolidin-2-yl)(thiazolidin-3-yl)methanone di-hydrobromide salt.
Kyorin disclosed a series of pyrrolidinecarbonitrile derivatives as DPP-IV inhibitors in international patent publication WO2008/114857 and US publication 2008/0146818. Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in WO2008/114857 and US publication
2008/0146818, and pharmaceutically acceptable salts, solvates, and hydrates thereof. One such compound is (2S,AS)- 1 -[2-[(4-ethoxycarbonylbicyclo[2.2.2]oct- 1 -yl)amino] acetyl] -4- fluoropyrrolidine-2-carbonitrile, and pharmaceutically acceptable salts thereof. In some embodiments, the DPP-IV inhibitor of the present invention is selected from (2S,4S)-l-[2-[(4- ethoxycarbonylbicyclo[2.2.2]oct- 1 -yl)amino]acetyl]-4-fluoropyrrolidine-2-carbonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000063_0002
Dainippon Sumitomo disclosed a series of bicyclic pyrrole derivatives as DPP-IV inhibitors in international patent publication WO2006/068163 and US publication 2009/0192129. Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in WO2006/068163 and US publication 2009/0192129 and pharmaceutically acceptable salts, solvates, and hydrates thereof. One such compound is 6-[(3R)-3-amino-piperidin-l-yl]-5-(2-chloro-5-fluoro-benzyl)-l,3- dimethyl-l,5dihydro-pyrrolo[3,2-d]pyrimidine-2,4-dione, and pharmaceutically acceptable salts thereof. In some embodiments, the DPP-IV inhibitor of the present invention is selected from (6- [(3R)-3-amino-piperidin- l-yl]-5-(2-chloro-5-fluoro-benzyl)- 1 ,3-dimethyl- 1 ,5dihydro-pyrrolo[3,2- d]pyrimidine-2,4-dione, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000064_0001
Dainippon Sumitomo disclosed 2-( {6-[(3R)-3-amino-3-methylpiperidin- 1 -yl]- 1 ,3- dimethyl-2,4-dioxo-l,2,3,4-tetrahydro-5H-pyrrolo[3,2-d]pyrimidin-5-yl}methyl)-4- fluorobenzonitrile as a DPP-IV inhibitor in international patent publication WO2009/084497. Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in international patent publication WO2009/084497. In some embodiments, the DPP-IV inhibitor of the present invention is selected from 2-({6-[(3R)-3-amino-3-methylpiperidin- 1 -yl]- 1 ,3-dimethyl-2,4-dioxo- 1 ,2,3,4-tetrahydro- 5H-pyrrolo[3,2-d]pyrimidin-5-yl}methyl)-4-fluorobenzonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000064_0002
Ηoffmann-La Roche disclosed a series of N-substituted pyrrolidine derivatives as DPP-
IV inhibitors in international patent publication WO 03/037327. Some embodiments of the present invention include every combination of one or more compounds selected from the DPP- IV inhibitors disclosed in WO 03/037327 and pharmaceutically acceptable salts, solvates, and hydrates thereof. One such compound is (2S)-l- {[2-(5-methyl-2-phenyl-oxazol-4-yl)- ethylamino]-acetyl}-pyrrolidine-2-carbonitrile, and pharmaceutically acceptable salts thereof. In some embodiments, the DPP-IV inhibitor of the present invention is selected from (2S)-I- {[2-(5- methyl-2-phenyl-oxazol-4-yl)-ethylamino]-acetyl}-pyrrolidine-2-carbonitrile, and
pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000065_0001
Various crystalline forms of (2S)-l-{[2-(5-methyl-2-phenyl-oxazol-4-yl)-ethylamino]- acetyl}-pyrrolidine-2-carbonitrile methansulfonic acid salt are disclosed in international patent publication WO2006/100181. In some embodiments, the DPP-IV inhibitor of the present invention is (2S)-l-{[2-(5-methyl-2-phenyl-oxazol-4-yl)-ethylamino]-acetyl}-pyrrolidine-2- carbonitrile methansulfonic acid salt (i.e., mesylate):
Figure imgf000065_0002
Another compound disclosed by Hoffmann-La Roche in international patent publication WO 03/037327 is (2S)-l-{[l,l-dimethyl-3-(4-pyridin-3-yl-imidazol-l-yl)-propylamino]-acetyl}- pyrrolidine-2-carbonitrile, and pharmaceutically acceptable salts thereof, such as the methansulfonic acid salt. In some embodiments, the DPP-IV inhibitor of the present invention is selected from (2S)- 1 - { [ 1 , 1 -dimethyl-3-(4-pyridin-3-yl-imidazol- 1 -yl)-propylamino]-acetyl} - pyrrolidine-2-carbonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000065_0003
In some embodiments, the DPP-IV inhibitor of the present invention is (2S)- 1- {[1,1- dimethyl-3-(4-pyridin-3-yl-imidazol- 1 -yl)-propylamino]-acetyl} -pyrrolidine-2-carbonitrile methansulfonic acid:
Figure imgf000066_0001
Various crystalline forms of (2S)-l-{[l,l-dimethyl-3-(4-pyridin-3-yl-imidazol-l-yl)- propylamino]-acetyl}-pyrrolidine-2-carbonitrile fumaric acid salt are disclosed in international patent publication WO2007/071576 (incorporated by reference in its entirety). In some embodiments, the DPP-IV inhibitor of the present invention is (2S)-l-{[l,l-dimethyl-3-(4- pyridin-3-yl-imidazol-l-yl)-propylamino]-acetyl}-pyrrolidine-2-carbonitrile fumaric acid salt (i.e., fumarate):
Figure imgf000066_0002
Pfizer disclosed a series of proline derivatives as DPP-IV inhibitors in international patent publication WO2005/116014 (incorporated by reference in its entirety). Some
embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in WO2005/116014 and pharmaceutically acceptable salts, solvates, and hydrates thereof. One such compound is (3,3-difluoropyrrolidin-l- yl)-((2S,4S)-4-(4-(pyrimidin-2yl)piperazin- 1 -yl)pyrrolidin-2-yl)methanone, and pharmaceutically acceptable salts thereof. In some embodiments, the DPP-IV inhibitor of the present invention is selected from (3,3-difluoropyrrolidin- 1 -yl)-((2S,4S)-4-(4-(pyrimidin-2yl)piperazin- 1 - yl)pyrrolidin-2-yl)methanone, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000066_0003
Glaxo SmithKline disclosed a series of fluoropyrrolidine derivatives as DPP-IV inhibitors in international patent publication WO 03/002531. Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in WO 03/037327 and pharmaceutically acceptable salts, solvates, and hydrates thereof. One such compound is (2S,4S)-l-[(2S)-2-amino-3,3-bis(4-fluorophenyl)propanoyi]-4- fluoropyrrolidine-2-carbonitrile (Denagliptin). In some embodiments, the DPP-IV inhibitor of the present invention is selected from (2S,4S)-l-[(2S)-2-amino-3,3-bis(4-fluorophenyl)propanoyl]-4- fluoropyrrolidine-2-carbonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000067_0001
Various crystalline forms of (2S,4S)-l-[(2S)-2-amino-3,3-bis(4-fluorophenyl)propanoyl]- 4-fluoropyrrolidine-2-carbonitrile and salts have been disclosed in international patent publication WO 2005/009956. One salt disclosed is (251,45)-l-[(25)-2-amino-3,3-bis(4- fluorophenyl)propanoyl]-4-fluoropyrrolidine-2-carbonitrile/>-toluenesulfonic acid salt (also referred to as (2S,45)-4-fluoro- 1 -[4-fluoro-β-(4-fluorophenyl)-L-phenylalanyl]-2- pyrrolidinecarbonitrile/>-toluenesulfonic acid salt, or Denagliptin tosylate). In some
embodiments, the DPP-IV inhibitor of the present invention is (2S,4S)-l-[(2S)-2-amino-3,3-bis(4- fluorophenyl)propanoyl]-4-fluoropyrrolidine-2-carbonitrile/>-toluenesulfonic acid salt:
Figure imgf000067_0002
Abbott disclosed a series of substituted pyrrolidinyl derivatives as DPP-IV inhibitors in international patent publication WO 2004/026822. Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in WO 2004/026822 and pharmaceutically acceptable salts, solvates, and hydrates thereof. One such compound is (2S,5R)-5-ethynyl-l-{N-(4-methyl-l-(4-carboxy-pyridin-2- yl)piperidin-4-yl)glycyl}pyrrolidine-2-carbonitrile. In some embodiments, the DPP-IV inhibitor of the present invention is selected from (2S,5R)-5-ethynyl-l-{N-(4-methyl-l-(4-carboxy-pyridin- 2-yl)piperidin-4-yl)glycyl}pyrrolidine-2-carbonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000068_0001
Abbott has further disclosed a series of substituted cyclohexanyl/cyclohexenyl derivatives as DPP-IV inhibitors in international patent publication WO 2007/027651. Some embodiments of the present invention include every combination of one or more compounds selected from the DPP-IV inhibitors disclosed in WO 2007/027651 and pharmaceutically acceptable salts, solvates, and hydrates thereof. One such compound is (15,6R)-3-{[3- (trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl]carbonyl}-6-(2,4,5- trifluorophenyl)cyclohex-3-en-l -amine. In some embodiments, the DPP-IV inhibitor of the present invention is selected from (lS,6R)-3- {[3-(trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3- a]pyrazin-7(8H)-yl]carbonyl} -6-(2,4,5-trifluorophenyl)cyclohex-3-en- 1 -amine, and
pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000068_0002
In accordance with the present invention, the combination can be used by mixing the respective active components, a compound of the present invention and pharmaceutical agent, either all together or independently with a physiologically acceptable carrier, excipient, binder, diluent, etc., as described herein above, and administering the mixture or mixtures either orally or non-orally as a pharmaceutical composition. When a compound or a mixture of compounds of the present invention are administered as a combination therapy with another active compound the therapeutic agents can be formulated as separate pharmaceutical compositions given at the same time or at different times; or the compound or a mixture of compounds of the present invention and the therapeutic agent(s) can be formulated together as a single unit dosage.
OTHER UTILITIES
Another object of the present invention relates to radio-labeled compounds that are useful not only in radio-imaging but also in assays, both in vitro and in vivo, for localizing and quantitating the GPRl 19 receptor in tissue samples, including human, and for identifying GPRl 19 receptor ligands by inhibition binding of a radio-labeled compound. It is a further object of this invention to develop novel GPRl 19 receptor assays of which comprise such radio-labeled compounds.
Compounds of the invention can also include all isotopes of atoms occurring in the intermediates and/or the final compound and pharmaceutically acceptable salts, solvates and hydrates thereof. Isotopes include those atoms having the same atomic number but different mass numbers. An isotopically-labeled or radio-labeled compound is one which is identical to compounds disclosed herein, but for the fact that one or more atoms is replaced or substituted by an atom having an atomic mass or mass number different from the atomic mass or mass number typically found in nature (i.e., naturally occurring). Suitable radionuclides that may be incorporated in compounds of the present invention include but are not limited to H (also written as D for deuterium), 3H (also written as T for tritium), 11C, 13C, 14C, 13N, 15N, 150, 170, 180, 18F, 35S, 36Cl, 82Br, 75Br, 76Br, 77Br, 1231, 1241, 125I and 131I. The radionuclide that is incorporated in the instant radio-labeled compounds will depend on the specific application for which the radio- labeled compound will be used. For example, for in vitro GPRl 19 receptor labeling and competition assays, compounds that incorporate 3H, 14C, 82Br, 1251, 131I, or 35S will generally be most useful. For radio-imaging applications 11C, 18F, 1251, 1231, 1241, 1311, 75Br, 76Br or 77Br will generally be most useful.
It is understood that a radio-labeled or labeled compound is a compound of the present invention that has incorporated at least one radionuclide; in some embodiments the radionuclide is selected from the group consisting of 3H, 14C, 1251, 35S, and 82Br.
Certain isotopically-labeled compounds of the present invention are useful in compound and/or substrate tissue distribution assays. In some embodiments the radionuclide 3H and/or 14C isotopes are useful in these studies. Further, substitution with heavier isotopes such as deuterium (i.e., 2H) may afford certain therapeutic advantages resulting from greater metabolic stability
(e.g., increased in vivo half-life or reduced dosage requirements) and hence may be preferred in some circumstances. Isotopically labeled compounds of the present invention can generally be prepared by following procedures analogous to those disclosed in the Schemes supra and Examples infra, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent. Other synthetic methods that are useful are discussed infra. Moreover, it should be understood that all of the atoms represented in the compounds of the invention can be either the most commonly occurring isotope of such atoms or a more scarce radio-isotope or nonradioactive isotope. Synthetic methods for incorporating radio-isotopes into organic compounds are applicable to compounds of the invention and are well known in the art, for example, tritium gas exposure labeling. This procedure involves exposing precursors containing exchangeable protons to tritium gas in the presence of a suitable catalyst.
A radio-labeled GPRl 19 receptor compound of present invention can be used in a screening assay to identify/evaluate compounds. In general terms, a newly synthesized or identified compound (i.e., test compound) can be evaluated for its ability to reduce binding of the radio-labeled compound of the present invention to the GPRl 19 receptor. Accordingly, the ability of a test compound to compete with the radio-labeled compound of the present invention for the binding to the GPRl 19 receptor directly correlates to its binding affinity.
The labeled compounds of the present invention bind to the GPRl 19 receptor. In one embodiment the labeled compound has an IC5O less than about 500 μM, in another embodiment the labeled compound has an IC50 less than about 100 μM, in yet another embodiment the labeled compound has an IC50 less than about 10 μM, in yet another embodiment the labeled compound has an IC50 less than about 1 μM, in still yet another embodiment the labeled inhibitor has an IC50 less than about 0.1 μM, in still yet another embodiment the labeled inhibitor has an IC50 less than about 0.01 μM, and in still yet another embodiment the labeled inhibitor has an IC50 less than about 0.005 μM.
As will be recognized, the steps of the methods of the present invention need not be performed any particular number of times or in any particular sequence. Additional objects, advantages, and novel features of this invention will become apparent to those skilled in the art upon examination of the following examples thereof, which are intended to be illustrative and not intended to be limiting. EXAMPLES
Example 1: Primary Pharmacology for Λ^l-fluoro-^methylsulfonylJphenylJ-S-methyl-ό- (l-(5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine (Compound 1) - In Vitro Assays.
A. cAMP Generation in CHO or RIN-5F Cells Expressing Human or Rat
Glucose-Dependent Insulino tropic Receptor (GDIR).
CHO cells expressing human GDIR (AGDIR) and rat GDIR (rGDIR) were used. CHO cells were cultured in DMEM/F12K media containing pen/strep, 10% FBS and the appropriate antibiotic for selection (500 μg/mL G418 for rat and 3μg/mL puromycin human clone cells). To establish assay conditions, 100 μL of cells were seeded at 5 x 105 cells/mL (2 x 105 cells/mL) into a lysine-coated 96 well culture plates and allowed to recover for 24 hours. After a 24-hour attachment and growth period, media was replaced with DMEM/F12 containing pen/strep, and 250 μM isobutylmethylxanthine (IBMX, phosphdiesterase inhibitor).
Compound or DMSO (vehicle) were added to the cells and incubated for 45 minutes at 37 0C in the presence of 0%, 4% or 20% serum. After incubation, the cells were lysed and processed according to the manufacturer's instructions (Tropix cAMP-Screen™ System Chemi- luminescent Immunoassay System for the Quantitation of cAMP from Cultured Mammalian cells). Cell media was removed and 100 μL of Assay/Lysis Buffer was added and mixed for cAMP assays using an ELISA kit (Tropix cAMP-Screen™ System Chemiluminescent
Immunoassay System, Applied Biosystems, Foster City, CA). The EC50 calculations were calculated with GraphPadPrism data analysis software.
Potency of Compound 1 in cAMP generation in CHO- h GDIR and
CHO- r GDIR cells.
Figure imgf000071_0001
B. GLP-I release from GLUTag cells
GluTag cells in growth medium (DMEM GIBCO Cat# 11995-065, 10% FBS GIBCO Cat# 16000-044) were seeded in a 96-well plate at 90,000 cells/well and incubated in a CO2 incubator at 37 0C for 24 hours. The growth medium was removed by aspiration and 200 μL of assay medium (DMEM with 5 mM glucose and 0.1% BSA) was added to each well. After incubating at 37 0C for 30 min, the assay medium was removed and each well was replenished with 175 μL of fresh assay medium with a DPP-IV inhibitor. Next, 25 μL of challenge solution was added to each well and the reaction was incubated for 2 hours at 37 0C. At completion of the reaction, 180 μL of supernatant was transferred to a fresh 96-well plate that was then placed in an -80 0C deep freezer for storage until assay. GLP-I was measured using an ELISA KIT from Lincoln Research (Cat. # EGLP-35K) according to manufactures instructions. Potency of Compound 1 in GLP-I release from GLUTag cells
Figure imgf000072_0001
Example 2: In vivo Effects of a Single Dose of Λ'-(2-fluoro-4-(methylsulfonyl)phenyl)-5- methyl-6-(l-(5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine on Oral Glucose Tolerance Test (oGTT) in Male SD Rats (PK/PD study) at 0.3 and 3 mg/kg.
The oral glucose tolerance test (oGTT) measures the ability of an animal to metabolize a bolus of exogenous glucose. A decrease in glucose AUC excursion from baseline indicates better glucose control. Male SD rats (6 - 7 weeks of age) were housed one rat per cage in a temperature- controlled room with 12-hour light/dark cycle. They were allowed ad libitum access to water and food. Rats were fasted overnight before the study. These rats were first dosed with vehicle (0.5%
HPMC) or Compound 1 at 0.3, or 3 mg/kg doses via oral gavage at 8:30 am. One hour after compound dosing, rats were administered glucose (2 g/kg, using 50% G) by oral gavage and the tail blood samples were collected to measure blood glucose and plasma insulin at 0, 30, 60, and 120 min. A separate group of rats were dosed in similar fashion and blood samples collected at 1, 2, 4, 6, 8, and 24 h after dosing for PK analysis.
Compound 1 significantly reduced the blood glucose AUC during OGTT at both the 0.3 and 3 mg/kg doses. The compound levels in the plasma showed a dose-related increase.
Efficacy of Λ'-(2-fluoro-4-(methylsulfonyl)phenyl)-5-methyl-6-(l-(5-methylpyrazin-2- yl)piperidin-4-yloxy)pyrimidin-4-amine (Compound 1) on glucose excursion in
SD rats during oGTT (PD arm).
Figure imgf000073_0001
* Statistically different compared to the vehicle- treated group (One way
repeated-measures analysis of variance (ANOVA) with Dunnett's post test.
Plasma concentrations of jY-^-fluoro^^methylsulfonylJphenylJ-S-methyl-ό-
(l-(5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-
4-amine (Compound 1) during glucose excursion
in SD rats during OGTT (PK arm).
Compound 1
Treatment
0. 3 mg/kg 3 mg/kg
Compound 1 I h, (0 min of OGTT) 56 .6 ± 35.5 933. 0 ± 417 .6
Levels 2 h, (60 min of OGTT) 54 .9 ± 28.6 894 .3 ± 98. 2
(ng/mL) 4 h, (180 min of OGTT) 35 .5 ± 21.5 397. 0 ± 207 .7
Example 3: Effects of Single Dose of Λ'-(2-fluoro-4-(methylsulfonyl)phenyl)-5-methyl-6-(l- (5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine (Compound 1) on oGTT Conducted in Normal Male Cynomolgus Monkeys. The effect of Compound 1 on blood glucose control was tested in healthy male
Cynomolgus monkeys. Naϊve, healthy, male Cynomolgus monkeys were used in this study. Monkeys were divided into 3 groups based on basal fasting blood glucose levels. Blood samples were collected before compound dosing, glucose administration, and at 30, 60, and 120 min after glucose nasal gavage (6 g/kg using 50% glucose solution). The results of this study indicate that Compound 1 at the 1.5 mg/kg dose significantly reduced blood glucose levels during oral glucose test.
N-(2-Fluoro-4-(methylsulfonyl)phenyl)-5-methyl-6-(l-(5-methylpyrazin-2-yl)piperidin-4- yloxy)pyrimidin-4-amine (Compound 1) on Blood Glucose Levels During oGTT
in Cynomolgus Monkeys
Figure imgf000074_0001
Example 4:
Inhibition of Recombinant P450 Enzymes
Human cytochrome P450 proteins are a family of enzymes involved in the metabolism of endogenous substrates and detoxication of drugs and other foreign chemicals. P450 enzymes convert various compounds into more polar molecules, and converted molecules are easier to remove from the body. Drug-drug interactions occur when two drugs are co-administered and compete for the same P450 enzyme possibly resulting in incomplete detoxication. Conditions of the assay are summarized in the following table.
Specific Enzyme Stocks and Substrates for P450 Assay
Figure imgf000075_0001
N-(2-Fluoro-4-(methylsulfonyl)phenyl)-5-methyl-6-(l-(5-methylpyrazin-2-yl)piperidin-4- yloxy)pyrimidin-4-amine was observed to inhibit isoenzymes 3A4, 2C9 and 2Cl 9 by roughly 50 % at 10 μM with IC50 values of 9.05, 8.82 and > 10 μM, respectively. There was no observed inhibition of 1 A2 or 2D6.
Inhibition Profile of Λ'-(2-Fluoro-4-(methylsulfonyl)phenyl)-5-methyl-6-
(l-(5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin- 4-amine (Compound 1) in Recombinant CYP-450 Enzymes
Figure imgf000075_0002
Example 5: Inhibition of P450 2C9 and 2C19 in HLM by Λ^-(2-Fluoro-4- (methylsulfonyl)phenyl)-5-methyl-6-(l-(5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin- 4-amine (Compound 1).
The metabolic incubations with standard probe substrates (tolbutamide for CYP2C9 and (S)-mephenytoin for CYP2C19) were conducted using a 96-well plate format. The general incubation mixture contained 0.5 mg/mL microsomal protein, 0 μM to 50 μM N-(2-fluoro-4- (methylsulfonyl)phenyl)-5-methyl-6-(l-(5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4- amine (as inhibitor), 1 mM NADPH and 100 mM potassium phosphate buffer containing 3 mM MgCl2 and 1 mM EDTA (pH 7.5) in a final volume of 100 μL. For CYP2C9, 150 μM
tolbutamide was added to the reaction mixture and incubated for 10 min at 37 0C. For CYP2C19, 50 μM (S)-mephenytoin was added to the reaction mixture and incubated for 40 min at 37 0C. Parallel control incubations conducted with tolbutamide and (S)-mephenytoin, without the addition of Compound 1, were used as positive controls of the reactions. All of the assays were conducted in triplicate. The incubations were terminated by the addition of 100 μL acetonitrile containing one of the following compounds as an internal standard: 4'-hydroxytolbutamide-d9 for CYP2C9 and 4'-hydroxymephenytoin-d3 for CYP2C19 (final concentration 100 ng/mL). After the addition of the internal standard, the 96-well plates were centrifuged at 3000 rpm for 10 min and 100 μL supernatant was transferred to a clean 96-well plate for analysis by LC/MS/MS.
Inhibition Profile ofΛ'-(2-Fluoro-4-(methylsulfonyl)phenyl)-5-methyl-6-(l-(5- methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine in
Recombinant CYP-450 Enzymes
Figure imgf000076_0001
Example 6:
The synthesis of N-(2-fluoro-4-(methylsulfonyl)phenyl)-5-methyl-6-(l -(5-methylpyrazin- 2-yl)piperidin-4-yloxy)pyrimidin-4-amine is shown in the following examples. The following examples are provided to further define the invention without, however, limiting the invention to the particulars of these examples. The compounds described herein are named according to the CS ChemDraw Ultra Version 9.0.7. In certain instances common names are used and it is understood that these common names would be recognized by those skilled in the art. Chemistry: Proton nuclear magnetic resonance (IH NMR) spectra were recorded on a Varian Mercury Vx-400 equipped with a 4 nucleus auto switchable probe and z-gradient or a Bruker Avance-400 equipped with a QNP (Quad Nucleus Probe) or a BBI (Broad Band Inverse) and z-gradient. Chemical shifts are given in parts per million (ppm) with the residual solvent signal used as reference. NMR abbreviations are used as follows: s = singlet, d = doublet, t = triplet, q = quartet, m = multiplet, sept = septet, br = broad. Microwave irradiations were carried out using the Emrys Synthesizer (Personal Chemistry). Thin-layer chromatography (TLC) was performed on silica gel 60 F254 (Merck), preparatory thin-layer chromatography (prep TLC) was performed on PK6F silica gel 60 A 1 mm plates (Whatman), and column chromatography was carried out on a silica gel column using Kieselgel 60, 0.063-0.200 mm (Merck). Evaporation was done in vacuo on a Buchi rotary evaporator. Celite 545 ® was used during palladium filtrations.
LCMS specs: 1) PC: HPLC-pumps: LC-IOAD VP, Shimadzu Inc.; HPLC system controller: SCL-IOA VP, Shimadzu Inc; UV-Detector: SPD-IOA VP, Shimadzu Inc;
Autosampler: CTC HTS, PAL, Leap Scientific; Mass spectrometer: API 150EX with Turbo Ion Spray source, AB/MDS Sciex; Software: Analyst 1.2. 2) Mac: HPLC-pumps: LC-8A VP,
Shimadzu Inc; HPLC system controller: SCL-IOA VP, Shimadzu Inc. UV-Detector: SPD-IOA VP, Shimadzu Inc; Autosampler: 215 Liquid Handler, Gilson Inc; Mass spectrometer: API 150EX with Turbo Ion Spray source, AB/MDS Sciex; Software: Masschrom 1.5.2. Example 6.1: Preparation of l-(5-Methylpyrazin-2-yl)piperidin-4-ol.
Figure imgf000077_0001
A mixture of 2-bromo-5-methylpyrazine (73 g, 422 mmol) and piperidin-4-ol (107 g, 1055 mmol) in 470 mL isopropanol was loaded into two high-pressure vessels respectively. Reaction was stirred at 150 0C for 17 h. Solvent was removed under reduced pressure to half volume and the residue was diluted with DCM and water. The organic layer was separated and the aqueous layer was extracted with DCM. The organic layers were dried over anhydrous MgSθ4 and concentrated in vacuo. The residue was filtered through a plug of silica gel eluting with 100% EtOAc to afford l-(5-methylpyrazin-2-yl)piperidin-4-ol as solid (69.75 g, 361 mmol, 86 % yield). Exact mass calculated for Ci0H15N3O 193.12, found 194.3 [M+H]+.
Example 6.2: Preparation of 4-Chloro-5-methyl-6-(l-(5-methylpyrazin-2-yl)piperidin-4- yloxy)pyrimidine.
Figure imgf000078_0001
To a solution of isopropyl l-(5-methylpyrazin-2-yl)piperidin-4-ol (69.75 g, 361 mmol) and 4,6-dichloro-5-methylpyrimidine (61.8 g, 379 mmol) in 125 mL of THF, 1 M potassium-?- butoxide in THF (375 mL, 375 mmol) was added dropwise in ice-acetone bath at 0 0C. The reaction mixture was allowed to warm to room temperature and continued to stir for 17 h. An additional 5 mL of 1 M potassium-ϊ-butoxide in THF was added. After two additional hours of stirring no change was observed. The reaction mixture was filtered through Celite® and washed with EtOAc (500 mL). The solvent was concentrated under reduced pressure to approximately one third volume. The resulting suspension was filtered, the solid was washed with cold CH3CN and the filtrate was concentrated under reduced pressure. The residue was purified by column chromatography (Biotage®, silica gel, AcOEt:Hex, 1 : 1, 340 g size column) to provide 4-chloro- 5-methyl-6-(l-(5-methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidine as a solid (80.35g, 70%). Exact mass calculated for C15H18ClN5O 319.12, found 320.3 [M+H]+. Example 6.3: Preparation of Λ'-(2-Fluoro-4-(methylsulfonyl)phenyl)-5-methyl-6-(l-(5- methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidin-4-amine (Compound 1).
Figure imgf000078_0002
(I)
A 2 L three neck round bottom flask was charged with 4-chloro-5-methyl-6-(l-(5- methylpyrazin-2-yl)piperidin-4-yloxy)pyrimidine (63.8 g, 200 mmol), 2-fluoro-4- (methylsulfonyl)aniline (37.7 g, 199 mmol), diacetoxypalladium (2.24 g, 9.98 mmol), l,V-bis(άi-
?-butylphosphino)ferrocene (9.47 g, 19.71 mmol), and dioxane (1 L). N2 was bubbled through the solution for 5 min. Sodium 2-methylpropan-2-olate (46.02 g, 479 mmol) was added at room temperature and then the mixture was heated to 100 0C. A yellow solid formed and the color changed from dark brown to yellow. This mixture was maintained at 100 0C for 1 h. The resuling reaction mixture was poured into 2 L of ice-water. The solid was filtered (80.3 g) and suspended in acetonitrile (1 L) at 80 0C. Mixture was cooled down and the solid was filtered to give N-(I-
11 fluoro-4-(methylsulfonyl) phenyl)-5-methyl-6-( 1 -(5-methylpyrazin-2-yl)piperidin-4- yloxy)pyrimidin-4-amine as light yellow-brown solid (74.26 g, 157 mmol, 79 % yield). 1H NMR (CDCl3, 400 MHz) δ 1.90-1.95 (m, 2H), 2.12-2.17 (m, 2H), 2.12 (s, 3H), 2.44 (s, 3H), 3.08 (s, 3H), 3.50-3.56 (m, 2H), 3.89-3.95 (m, 2H), 5.44-5.46 (sept, J= 3.79 Hz, IH), 6.84-6.85 (d, J = 4.17 Hz, IH), 7.71-7.77 (m, 2H), 7.99 (s, IH), 8.13 (s, IH), 8.44 (s, IH), 8.84-8.88 (t, J= 8.34
Hz, IH). Exact mass calculated for C22H25FN6O3S 472.17, found 473.5 [M+H]+.
This material was further characterized by powder X-ray diffraction (XRPD), thermogravimetric analysis (TGA), and differential scanning calorimetry (DSC). The powder X- ray diffraction (XRPD) pattern shows that the material is crystalline (see Figure 1). The thermogravimetric analysis (TGA) thermogram shows the material has an initial weight loss of about -1.0% in the range of the melting temperature (from about 130 0C to about 180 0C, see Figure 2). The differential scanning calorimetry (DSC) thermogram shows the material has a narrow endothermic event at a melting temperature with an onset and peak temperature of about 180.7 ° and about 182.8 0C respectively, with an enthalpy of 74.3 J/g due to melting of the sample and subsequent exothermic decomposition (see Figure 3).
Those skilled in the art will recognize that various modifications, additions, substitutions, and variations to the illustrative examples set forth herein can be made without departing from the spirit of the invention and are, therefore, considered within the scope of the invention. All documents referenced above, including, but not limited to, printed publications, and provisional and regular patent applications, are incorporated herein by reference in their entirety.

Claims

CLAIMS We claim:
1. A compound selected from the following compound of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000080_0001
(I)
2. A pharmaceutical composition comprising a compound according to claim 1 and a
pharmaceutically acceptable carrier.
3. A composition comprising a compound according to claim 1 and an inhibitor of DPP-IV.
4. A composition according to claim 3, wherein said inhibitor of DPP-IV is selected from the following compounds and pharmaceutically acceptable salts, solvates, and hydrates thereof:
3(R)-amino-l-[3-(trifluoromethyl)-5,6,7,8-tetrahydro[l,2,4]triazolo[4,3- a]pyrazin-7-yl]-4-(2,4,5-trifluorophenyl) butan- 1 -one;
1 -[2-(3-hydroxyadamant- 1 -ylamino)acetyl]pyrrolidine-2(S)-carbonitrile;
(lS,3S,5S>2-[2(S)-amino-2-(3-hydroxyadamantan- 1 -yl)acetyl]-2- azabicyclo[3.1.0]hexane-3-carbonitrile;
2-[6-[3(R)-ammopiperidin-l-yl]-3-methyl-2,4-dioxo-l,2,3,4- tetrahydropyrimidin- 1 -ylmethyljbenzonitrile;
8-[3(R)-aminopiperidin- 1 -yl]-7-(2-butynyl)-3-methyl- 1 -(4-methylquinazolin-2- ylmethyl)xanthine;
l-[N-[3(R)-pyrrolidinyl]glycyl]pyrrolidin-2(R)-yl boronic acid;
4(S)-fluoro-l-[2-[(lR,3S)-3-(lH-l,2,4-triazol-l- ylmethyl)cyclopentylamino]acetyl]pyrrolidine-2(S)-carbonitrile;
1 -[(2S,3S,\ lbS)-2-amino-9,10-dimethoxy-2,3,4,6,7,l lb-hexahydro- IH- pyrido[2,l-a]isoquinolin-3-yl]-4(S)-(fluoromethyl)pyrrolidin-2-one;
(2S,4S)-2-cyano-4-fluoro- 1 -[(2-hydroxy- 1 , 1 -dimethyl)
ethylamino]acetylpyrrolidine; 8-(cw-hexahydro-pyrrolo[3,2-b]pyrrol- 1 -yl)-3-methyl-7-(3-methyl-but-2-enyl)- 1 - (2-oxo-2-phenylethyl)-3,7-dihydro-purine-2,6-dione;
1 -((3S,4S)-4-amino- 1 -(4-(3,3-difluoropyrrolidin- 1 -yl)- 1 ,3,5-triazin-2- yl)pyrrolidin-3-yl)-5,5difluoropiperidin-2-one;
(R)-2-((6-(3-aminopiperidin- 1 -yl)-3-methyl-2,4-dioxo-3,4-dihydropyrimidin- l(2H)-yl)methyl)-4-fluorobenzonitrile;
5- {(S)-2-[2-((S)-2-cyano-pyrrorid in- 1 -yl)-2-oxo-ethylamino]-propyl} -5-(1H- tetrazol-5-yl)10,l l-dihydro-5H-dibenzo[a,d]cycloheptene-2,8-dicarboxylic acid bis- dimethylamide;
((2S,4S)-4-(4-(3 -methyl- 1 -phenyl- lH-pyrazol-5-yl)piperazin- 1 -yl)pyrrolidin-2- yl)(thiazolidin- 3 -yl)methanone ;
(2S,4S)- 1 -[2-[(4-ethoxycarbonylbicyclo[2.2.2]oct- 1 -yl)amino] acetyl] -4- fluoropyrrolidine-2-carbonitrile;
6-[(3R)-3-amino-piperidin- 1 -yl]-5-(2-chloro-5-fluoro-benzyl)- 1 ,3-dimethyl- 1 ,5dihydro-pyrrolo[3,2-d]pyrimidine-2,4-dione;
2-({6-[(3R)-3-amino-3-methylpiperidin-l-yl]-l,3-dimethyl-2,4-dioxo-l,2,3,4- tetrahydro-5H-pyrrolo[3,2-d]pyrimidin-5-yl}methyl)-4-fluorobenzonitrile;
(2S)-l-{[2-(5-methyl-2-phenyl-oxazol-4-yl)-ethylamino]-acetyl}-pyrrolidine-2- carbonitrile;
(2S)- 1 - {[1 , 1 -dimethyl-3-(4-pyridin-3-yl-imidazol- 1 -yl)-propylamino]-acetyl} - pyrrolidine-2-carbonitrile;
(3,3-difluoropyrrolidin- 1 -yl)-((2S,4S)-4-(4-(pyrimidin-2yl)piperazin- 1 - yl)pyrrolidin-2-yl)methanone;
(2S,4S)-l-[(2S)-2-amino-3,3-bis(4-fluorophenyl)propanoyl]-4-fluoropyrrolidine- 2-carbonitrile;
(2S,5R)-5-ethynyl- 1 - {N-(4-methyl- 1 -(4-carboxy-pyridin-2-yl)piperidin-4- yl)glycyl}pyrrolidine-2-carbonitrile; and
(lS,6R)-3-{[3-(trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)- yljcarbonyl} -6-(2,4,5-trifluorophenyl)cyclohex-3-en- 1 -amine.
5. A pharmaceutical composition according to claim 3 or 4, further comprising a
pharmaceutically acceptable carrier.
6. A method for the treatment of a GPRl 19 receptor related disorder in an individual, comprising administering to said individual in need thereof, a therapeutically effective amount of a compound according to claim 1 , or a composition according to any one of claims 2 to 5.
7. A method according to claim 6, wherein said GPRl 19 receptor related disorder is a metabolic-related disorder.
8. A method according to claim 7, wherein said metabolic-related disorder is selected from the group consisting of:
diabetes mellitus, type 1 diabetes, type 2 diabetes, inadequate glucose tolerance, impaired glucose tolerance, insulin resistance, hyperglycemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia, atherosclerosis, stroke, syndrome X, hypertension, pancreatic beta-cell insufficiency, enteroendocrine cell insufficiency, glucosuria, metabolic acidosis, cataracts, diabetic nephropathy, diabetic neuropathy, peripheral neuropathy, diabetic coronary artery disease, diabetic
cerebrovascular disease, diabetic peripheral vascular disease, diabetic retinopathy, metabolic syndrome, a condition related to diabetes mellitus, myocardial infarction, learning impairment, memory impairment, a neurodegenerative disorder, a condition ameliorated by increasing a blood GLP- 1 level in an individual with a neurodegenerative disorder, excitotoxic brain damage caused by severe epileptic seizures, Alzheimer's disease, Parkinson's disease, Huntington's disease, prion-associated disease, stroke, motor-neuron disease, traumatic brain injury, spinal cord injury, and obesity.
9. A method according to claim 7, wherein said metabolic-related disorder is type 2
diabetes.
10. A method for the treatment of a GPRl 19 receptor related disorder in an individual, comprising administering to said individual in need thereof, a therapeutically effective amount of a compound according to claim 1 in combination with an inhibitor of DPP-IV, wherein said compound and said inhibitor of DPP-IV are administered simultaneously, separately, or sequentially.
11. A method according to claim 10, wherein said compound and said inhibitor of DPP-IV are administered simultaneously.
12. A method according to claim 10, wherein said compound and said inhibitor of DPP-IV are administered separately.
13. A method according to claim 10, wherein said compound and said inhibitor of DPP-IV are administered sequentially.
14. A method according to any one of claims 10 to 13, wherein said GPRl 19 receptor related disorder is a metabolic-related disorder.
15. A method according to claim 14, wherein said metabolic-related disorder is selected from the group consisting of:
diabetes mellitus, type 1 diabetes, type 2 diabetes, inadequate glucose tolerance, impaired glucose tolerance, insulin resistance, hyperglycemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia, atherosclerosis, stroke, syndrome X, hypertension, pancreatic beta-cell insufficiency, enteroendocrine cell insufficiency, glucosuria, metabolic acidosis, cataracts, diabetic nephropathy, diabetic neuropathy, peripheral neuropathy, diabetic coronary artery disease, diabetic cerebrovascular disease, diabetic peripheral vascular disease, diabetic retinopathy, metabolic syndrome, a condition related to diabetes mellitus, myocardial infarction, learning impairment, memory impairment, a neurodegenerative disorder, a condition ameliorated by increasing a blood GLP- 1 level in an individual with a neurodegenerative disorder, excitotoxic brain damage caused by severe epileptic seizures, Alzheimer's disease, Parkinson's disease, Huntington's disease, prion-associated disease, stroke, motor-neuron disease, traumatic brain injury, spinal cord injury, and obesity.
16. A method according to claim 14, wherein said metabolic-related disorder is type 2
diabetes.
17. A method according to any one of claims 10 to 16, wherein said inhibitor of DPP-IV is selected from the following compounds and pharmaceutically acceptable salts, solvates, and hydrates thereof: 3(R)-amino-l-[3-(trifluoromethyl)-5,6,7,8-tetrahydro[l,2,4]triazolo[4,3- a]pyrazin-7-yl]-4-(2,4,5-trifluorophenyl) butan- 1 -one;
1 -[2-(3-hydroxyadamant- 1 -ylamino)acetyl]pyrrolidine-2(S)-carbonitrile;
(lS,3S,5S>2-[2(S>amino-2-(3-hydroxyadamantan-l-yl)acetyl]-2- azabicyclo[3.1.0]hexane-3-carbonitrile;
2-[6-[3(R)-aminopiperidin-l-yl]-3-methyl-2,4-dioxo-l,2,3,4- tetrahydropyrimidin- 1 -ylmethyl]benzonitrile;
8-[3(R)-aminopiperidin- 1 -yl]-7-(2-butynyl)-3-methyl- 1 -(4-methylquinazolin-2- ylmethyl)xanthine;
1 -[N-[3(R)-pyrrolidinyl]glycyl]pyrrolidin-2(R)-yl boronic acid;
4(5)-fluoro-l-[2-[(lΛ,35)-3-(lH-l,2,4-triazol-l- ylmethyl)cyclopentylamino]acetyl]pyrrolidme-2(S)-carbonitrile;
1 -[(2S,3S,\ lbS>2-amino-9,10-dimethoxy-2,3,4,6,7,l lb-hexahydro- IH- pyrido[2,l-a]isoquinolin-3-yl]-4(S)-(fluoromethyl)pyrrolidin-2-one;
(2S,4S)-2-cyano-4-fluoro- 1 -[(2-hydroxy- 1 , 1 -dimethyl)
ethylamino]acetylpyrrolidine;
8-(c^-hexahydro-pyrrolo[3,2-b]pyrrol- 1 -yl)-3-methyl-7-(3-methyl-but-2-enyl)- 1 - (2-oxo-2-phenylethyl)-3,7-dihydro-purine-2,6-dione;
1 -((3S,45)-4-amino- 1 -(4-(3,3-difluoropyrrolidin- 1 -yl)- 1 ,3,5-triazin-2- yl)pyrrolidin-3-yl)-5,5difluoropiperidin-2-one;
(R)-2-((6-(3-aminopiperidin-l-yl)-3-methyl-2,4-dioxo-3,4-dihydropyrimidin- l(2H)-yl)methyl)-4-fluorobenzonitrile;
5- {(S)-2-[2-((S)-2-cyano-pyrrolid in- 1 -yl)-2-oxo-ethylamino]-propyl} -5-(1H- tetrazol-5-yl)10,l l-dihydro-5H-dibenzo[a,d]cycloheptene-2,8-dicarboxylic acid bis- dimethylamide;
((2S, 4S)-4-(4-(3 -methyl- 1 -phenyl- lH-pyrazol-5-yl)piperazin- 1 -yl)pyrrolidin-2- yl)(thiazolidin- 3 -yl)methanone ;
(2S,AS)- 1 -[2-[(4-ethoxycarbonylbicyclo[2.2.2]oct- 1 -yl)amino] acetyl] -4- fluoropyrrolidine-2-carbonitrile;
6-[(3R)-3-amino-piperidin-l-yl]-5-(2-chloro-5-fluoro-benzyl)-l,3-dimethyl- l,5dihydro-pyrrolo[3,2-d]pyrimidine-2,4-dione;
2-({6-[(3R)-3-amino-3-methylpiperidin-l-yl]-l,3-dimethyl-2,4-dioxo-l,2,3,4- tetrahydro-5H-pyrrolo[3,2-d]pyrimidin-5-yl}methyl)-4-fluorobenzonitrile; (2S)-l-{[2-(5-methyl-2-phenyl-oxazol-4-yl)-ethylamino]-acetyl}-pyrrolidine-2- carbonitrile;
(2S)- 1 - { [ 1 , 1 -dimethyl-3-(4-pyridin-3-yl-imidazol- 1 -yl)-propylamino]-acetyl} - pyrrolidine-2-carbonitrile;
(3,3-difluoropyrrolidin- 1 -yl)-((2S,4S)-4-(4-(pyrimidin-2yl)piperazin- 1 - yl)pyrrolidin-2-yl)methanone;
(2S,4S)-l-[(2S)-2-amino-3,3-bis(4-fluorophenyl)propanoyl]-4-fluoropyrrolidine- 2-carbonitrile;
(2£,5R)-5-ethynyl- 1 - {N-(4-methyl- 1 -(4-carboxy-pyridin-2-yl)piperidin-4- yl)glycyl}pyrrolidine-2-carbonitrile; and
(lS,6R)-3- {[3-(trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)- yl]carbonyl} -6-(2,4,5-trifluorophenyl)cyclohex-3-en- 1 -amine.
18. Use of a compound according to claim 1, or a composition according to any one of
claims 2 to 5, in the manufacture of a medicament for the treatment of a GPRl 19 receptor related disorder.
19. Use of a compound according to claim 1, or a composition according to any one of
claims 2 to 5, in the manufacture of a medicament for the treatment of a metabolic-related disorder.
20. Use of a compound according to claim 1, or a composition according to any one of
claims 2 to 5, in the manufacture of a medicament for the treatment of a metabolic-related disorder selected from the group consisting of:
diabetes mellitus, type 1 diabetes, type 2 diabetes, inadequate glucose tolerance, impaired glucose tolerance, insulin resistance, hyperglycemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia, atherosclerosis, stroke, syndrome X, hypertension, pancreatic beta-cell insufficiency, enteroendocrine cell insufficiency, glucosuria, metabolic acidosis, cataracts, diabetic nephropathy, diabetic neuropathy, peripheral neuropathy, diabetic coronary artery disease, diabetic
cerebrovascular disease, diabetic peripheral vascular disease, diabetic retinopathy, metabolic syndrome, a condition related to diabetes mellitus, myocardial infarction, learning impairment, memory impairment, a neurodegenerative disorder, a condition ameliorated by increasing a blood GLP- 1 level in an individual with a neurodegenerative disorder, excitotoxic brain damage caused by severe epileptic seizures, Alzheimer's disease, Parkinson's disease, Huntington's disease, prion-associated disease, stroke, motor-neuron disease, traumatic brain injury, spinal cord injury, and obesity.
21. Use of a compound according to claim 1 , or a composition according to any one of claims 2 to 5, in the manufacture of a medicament for the treatment of type 2 diabetes.
22. A compound according to claim 1, or a composition according to any one of claims 2 to 5, for use in a method of treatment of the human or animal body by therapy.
23. A compound according to claim 1, or a composition according to any one of claims 2 to 5, for use in a method of treatment of a GPRl 19 receptor related disorder.
24. A compound according to claim 1, or a composition according to any one of claims 2 to 5, for use in a method of treatment of a metabolic-related disorder.
25. A compound according to claim 1, or a composition according to any one of claims 2 to 5, for use in a method of treatment of a metabolic-related disorder selected from the group consisting of:
diabetes mellitus, type 1 diabetes, type 2 diabetes, inadequate glucose tolerance, impaired glucose tolerance, insulin resistance, hyperglycemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia, atherosclerosis, stroke, syndrome X, hypertension, pancreatic beta-cell insufficiency, enteroendocrine cell insufficiency, glucosuria, metabolic acidosis, cataracts, diabetic nephropathy, diabetic neuropathy, peripheral neuropathy, diabetic coronary artery disease, diabetic cerebrovascular disease, diabetic peripheral vascular disease, diabetic retinopathy, metabolic syndrome, a condition related to diabetes mellitus, myocardial infarction, learning impairment, memory impairment, a neurodegenerative disorder, a condition ameliorated by increasing a blood GLP- 1 level in an individual with a neurodegenerative disorder, excitotoxic brain damage caused by severe epileptic seizures, Alzheimer's disease, Parkinson's disease, Huntington's disease, prion-associated disease, stroke, motor-neuron disease, traumatic brain injury, spinal cord injury, and obesity.
26. A compound according to claim 1, or a composition according to any one of claims 2 to 5, for use in a method of treatment of type 2 diabetes.
27. A method of preparing a composition comprising the step of admixing a compound
according to claim 1 , and a pharmaceutically acceptable carrier.
28. A method of preparing a composition comprising the step of admixing a compound
according to claim 1 and an inhibitor of DPP-IV.
29. A method of preparing a composition comprising the step of admixing a compound
according to claim 1 , with an inhibitor of DPP-IV, and a pharmaceutically acceptable carrier.
30. A method of preparing a composition according to claim 28 or 29, wherein said inhibitor of DPP-IV is selected from the following compounds and pharmaceutically acceptable salts, solvates, and hydrates thereof:
3(R)-amino-l-[3-(trifluoromethyl)-5,6,7,8-tetrahydro[l,2,4]triazolo[4,3- a]pyrazin-7-yl]-4-(2,4,5-trifluorophenyl) butan- 1 -one;
1 -[2-(3-hydroxyadamant- 1 -ylamino)acetyl]pyrrolidine-2(S)-carbonitrile;
(lS,3S,5S>2-[2(S>amino-2-(3-hydroxyadamantan-l-yl)acetyl]-2- azabicyclo[3.1.0]hexane-3-carbonitrile;
2-[6-[3(R)-aminopiperidin-l-yl]-3-methyl-2,4-dioxo- 1,2,3,4- tetrahydropyrimidin- 1 -ylmethyljbenzonitrile;
8-[3(R)-aminopiperidin- 1 -yl]-7-(2-butynyl)-3-methyl- 1 -(4-methylquinazolin-2- ylmethyl)xanthine;
l-[N-[3(R)-pyrrolidinyl]glycyl]pyrrolidin-2(R)-yl boronic acid;
4(5)-fluoro-l-[2-[(lΛ,35)-3-(lH-l,2,4-triazol-l- ylmethyl)cyclopentylammo]acetyl]pyrrolidine-2(S)-carbonitrile;
l-[(2S,3S,l lbS>2-amino-9,10-dimethoxy-2,3,4,6,7,l lb-hexahydro-lH- pyrido[2, 1 -a]isoqumoh'n-3-yl]-4(S)-(fluoromethyl)pyrrolidin-2-one;
(2S,4S)-2-cyano-4-fluoro- 1 -[(2-hydroxy- 1 , 1 -dimethyl)
ethylamino]acetylpyrrolidine;
8-(c^-hexahydro-pyrrolo[3,2-b]pyrrol- 1 -yl)-3-methyl-7-(3-methyl-but-2-enyl)- 1 - (2-oxo-2-phenylethyl)-3,7-dihydro-purine-2,6-dione; 1 -((3S,45)-4-amino- 1 -(4-(3,3-difluoropyrrolidin- 1 -yl)- 1 ,3,5-triazin-2- yl)pyrrolidin-3-yl)-5,5difluoropiperidin-2-one;
(R)-2-((6-(3-ammopiperidin-l-yi)-3-methyl-2,4-dioxo-3,4-dihydropyrimidin- l(2H)-yl)methyl)-4-fluorobenzonitrile;
S-K^^-P-^^-cyano-pyrrolid in-l-y^^-oxo-ethylaminol-propylj-S-ClH- tetrazol-5-yl)10,l l-dihydro-5H-dibenzo[a,d]cycloheptene-2,8-dicarboxylic acid bis- dimethylamide;
((2S, 4ιS)-4-(4-(3 -methyl- 1 -phenyl- lH-pyrazol-5-yl)piperazin- 1 -yl)pyrrolidin-2- yl)(thiazolidin- 3 -yl)methanone ;
(2S,4S)- 1 -[2-[(4-ethoxycarbonylbicyclo[2.2.2]oct- 1 -yl)amino] acetyl] -4- fluoropyrrolidine-2-carbonitrile;
6-[(3R)-3-amino-piperidin- 1 -yl]-5-(2-chloro-5-fluoro-benzyl)- 1 ,3-dimethyl- l,5dihydro-pyrrolo[3,2-d]pyrimidine-2,4-dione;
2-({6-[(3R)-3-amino-3-methylpiperidin-l-yl]-l,3-dimethyl-2,4-dioxo-l,2,3,4- tetrahydro-5H-pyrrolo[3,2-d]pyrimidin-5-yl}methyl)-4-fluorobenzonitrile;
(2S)-l-{[2-(5-methyl-2-phenyl-oxazol-4-yl)-ethylamino]-acetyl}-pyrrolidine-2- carbonitrile;
(2S)- 1 - {[1 , 1 -dimethyl-3-(4-pyridin-3-yl-imidazol- 1 -yl)-propylamino] -acetyl} - pyrrolidine-2-carbonitrile;
(3,3-difluoropyrrolidin- 1 -yl)-((2S,4S)-4-(4-(pyrimidin-2yl)piperazin- 1 - yl)pyrrolidin-2-yl)methanone;
(2S,4S)-l-[(2S)-2-amino-3,3-bis(4-fluorophenyl)propanoyl]-4-fluoropyrrolidine- 2-carbonitrile;
(2£,5R)-5-ethynyl- 1 - {N-(4-methyl- 1 -(4-carboxy-pyridin-2-yl)piperidin-4- yl)glycyl}pyrrolidine-2-carbonitrile; and
(lS,6R)-3-{[3-(trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)- yljcarbonyl} -6-(2,4,5-trifluorophenyl)cyclohex-3-en- 1 -amine.
31. A compound according to claim 1 for use in combination with an inhibitor of DPP-IV for the treatment of a condition ameliorated by increasing a blood GLP- 1 level in a mammal.
32. A compound according to claim 31 , wherein the amount of said compound alone is therapeutically ineffective for treatment of a condition ameliorated by increasing a blood GLP- 1 level in a mammal.
33. A compound according to claim 31 or 32, wherein said condition ameliorated by increasing a blood GLP- 1 level is selected from the group consisting of:
diabetes mellitus, type 1 diabetes, type 2 diabetes, inadequate glucose tolerance, impaired glucose tolerance, insulin resistance, hyperglycemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia, atherosclerosis, stroke, syndrome X, hypertension, pancreatic beta-cell insufficiency, enteroendocrine cell insufficiency, glucosuria, metabolic acidosis, cataracts, diabetic nephropathy, diabetic neuropathy, peripheral neuropathy, diabetic coronary artery disease, diabetic cerebrovascular disease, diabetic peripheral vascular disease, diabetic retinopathy, metabolic syndrome, a condition related to diabetes mellitus, myocardial infarction, learning impairment, memory impairment, a neurodegenerative disorder, a condition ameliorated by increasing a blood GLP- 1 level in an individual with a neurodegenerative disorder, excitotoxic brain damage caused by severe epileptic seizures, Alzheimer's disease, Parkinson's disease, Huntington's disease, prion-associated disease, stroke, motor-neuron disease, traumatic brain injury, spinal cord injury, and obesity.
34. A compound according to claim 31 or 32, wherein said condition is type 2 diabetes. 35. A compound according to any one of claims 31 to 34, wherein said inhibitor of DPP-IV is selected from the following compounds and pharmaceutically acceptable salts, solvates, and hydrates thereof:
3(R)-amino-l-[3-(trifluoromethyl)-5,6,7,8-tetrahydro[l,2,4]triazolo[4,3- a]pyrazin-7-yl]-4-(2,4,5-trifluorophenyl) butan- 1 -one;
1 -[2-(3-hydroxyadamant- 1 -ylamino)acetyl]pyrrolidine-2(S)-carbonitrile;
(lS,3S,5S>2-[2(S>amino-2-(3-hydroxyadamantan-l-yl)acetyl]-2- azabicyclo[3.1.0]hexane-3-carbonitrile;
2-[6-[3(R)-aminopiperidin-l-yl]-3-methyl-2,4-dioxo-l,2,3,4- tetrahydropyrimidin- 1 -ylmethyljbenzonitrile;
8-[3(R)-aminopiperidin- 1 -yl]-7-(2-butynyl)-3-methyl- 1 -(4-methylquinazolin-2- ylmethyl)xanthine;
l-[N-[3(R)-pyrrolidinyl]glycyl]pyrrolidin-2(R)-yl boronic acid;
4(5)-fluoro-l-[2-[(lΛ,
35)-3-(lH-l,2,4-triazol-l- ylmethyl)cyclopentylamino]acetyl]pyrrolidine-2(£)-carbonitrile; 1 -[(2S,3S,l lbS>2-amino-9,10-dimethoxy-2,3,4,6,7,l lb-hexahydro- IH- pyrido[2,l-a]isoquinolin-3-yl]-4(S)-(fluoromethyl)pyrrolidin-2-one;
(2S,4S)-2-cyano-4-fluoro- 1 -[(2-hydroxy- 1 , 1 -dimethyl)
ethylamino]acetylpyrrolidine;
8-(cw-hexahydro-pyrrolo[3,2-b]pyrrol- 1 -yl)-3-methyl-7-(3-methyl-but-2-enyl)- 1 -
(2-oxo-2-phenylethyl)-3,7-dihydro-purine-2,6-dione;
1 -((3S,45)-4-amino- 1 -(4-(3,3-difluoropyrrolidin- 1 -yl)- 1 ,3,5-triazin-2- yl)pyrrolidin-3-yl)-5,5difluoropiperidin-2-one;
(R)-2-((6-(3-aminopiperidin-l-yl)-3-methyl-2,4-dioxo-3,4-dihydropyrimidin- 1 (2H)-yl)methyl)-4-fluorobenzonitrile;
5- {(S)-2-[2-((S)-2-cyano-pyrrolid in- 1 -yl)-2-oxo-ethylamino]-propyl} -5-(1H- tetrazol-5-yl)10,l l-dihydro-5H-dibenzo[a,d]cycloheptene-2,8-dicarboxylic acid bis- dimethylamide;
((2ιS',4ιS)-4-(4-(3-methyl- 1 -phenyl- lH-pyrazol-5-yl)piperazin- 1 -yl)pyrrolidin-2- yl)(thiazolidin-3-yl)methanone;
(2S,4S)- 1 -[2-[(4-ethoxycarbonylbicyclo[2.2.2]oct- 1 -yl)amino] acetyl] -4- fluoropyrrolidine-2-carbonitrile;
6-[(3R)-3-amino-piperidin- 1 -yl]-5-(2-chloro-5-fluoro-benzyl)- 1 ,3-dimethyl- l,5dihydro-pyrrolo[3,2-d]pyrimidine-2,4-dione;
2-({6-[(3R)-3-amino-3-methylpiperidin-l-yl]-l,3-dimethyl-2,4-dioxo-l,2,3,4- tetrahydro-5H-pyrrolo[3,2-d]pyrimidin-5-yl}methyl)-4-fluorobenzonitrile;
(2S)-l-{[2-(5-methyl-2-phenyl-oxazol-4-yl)-ethylamino]-acetyl}-pyrrolidine-2- carbonitrile;
(2S)- 1 - {[1 , 1 -dimethyl-3-(4-pyridin-3-yl-imidazol- 1 -yl)-propylamino]-acetyl} - pyrrolidine-2-carbonitrile;
(3,3-difluoropyrrolidin- 1 -yl)-((2S,4S)-4-(4-(pyrimidin-2yl)piperazin- 1 - yl)pyrrolidin-2-yl)methanone;
(2S,4S)-l-[(2S)-2-amino-3,3-bis(4-fluorophenyl)propanoyl]-4-fluoropyrrolidine- 2-carbonitrile;
(2S,5R)-5-ethynyl- 1 - {N-(4-methyl- 1 -(4-carboxy-pyridin-2-yl)piperidin-4- yl)glycyl}pyrrolidine-2-carbonitrile; and
(lS,6R)-3- {[3-(trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)- yljcarbonyl} -6-(2,4,5-trifluorophenyl)cyclohex-3-en- 1 -amine.
36. An inhibitor of DPP-IV for use in combination with a compound according to claim 1 for the treatment of a condition ameliorated by increasing a blood GLP- 1 level in a mammal.
37. An inhibitor of DPP-IV according to claim 36, wherein the amount of said inhibitor of DPP-IV alone is therapeutically ineffective for treatment of a condition ameliorated by increasing a blood GLP- 1 level in a mammal.
38. An inhibitor of DPP-IV according to claim 36 or 37, wherein said condition ameliorated by increasing a blood GLP- 1 level is selected from the group consisting of:
diabetes mellitus, type 1 diabetes, type 2 diabetes, inadequate glucose tolerance, impaired glucose tolerance, insulin resistance, hyperglycemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, dyslipidemia, atherosclerosis, stroke, syndrome X, hypertension, pancreatic beta-cell insufficiency, enteroendocrine cell insufficiency, glucosuria, metabolic acidosis, cataracts, diabetic nephropathy, diabetic neuropathy, peripheral neuropathy, diabetic coronary artery disease, diabetic
cerebrovascular disease, diabetic peripheral vascular disease, diabetic retinopathy, metabolic syndrome, a condition related to diabetes mellitus, myocardial infarction, learning impairment, memory impairment, a neurodegenerative disorder, a
neurodegenerative disorder, excitotoxic brain damage caused by severe epileptic seizures, Alzheimer's disease, Parkinson's disease, Huntington's disease, prion-associated disease, stroke, motor-neuron disease, traumatic brain injury, spinal cord injury, and obesity.
39. An inhibitor of DPP-IV according to claim 36 or 37, wherein said condition is type 2 diabetes.
40. An inhibitor of DPP-IV according to any one of claims 36 to 39, wherein said inhibitor of DPP-IV is selected from the following compounds and pharmaceutically acceptable salts, solvates, and hydrates thereof:
3(R)-amino-l-[3-(trifluoromethyl)-5,6,7,8-tetrahydro[l,2,4]triazolo[4,3- a]pyrazin-7-yl]-4-(2,4,5-trifluorophenyl) butan-1-one;
1 -[2-(3-hydroxyadamant- 1 -ylamino)acetyl]pyrrolidine-2(S)-carbonitrile;
(lS,3S,5S>2-[2(S>amino-2-(3-hydroxyadamantan-l-yi)acetyl]-2- azabicyclo[3.1.0]hexane-3-carbonitrile; 2-[6-[3(R)-aminopiperidin-l-yl]-3-methyl-2,4-dioxo-l,2,3,4- tetrahydropyrimidin- 1 -ylmethyl]benzonitrile;
8-[3(R)-aminopiperidin- 1 -yl]-7-(2-butynyl)-3-methyl- 1 -(4-methylquinazolin-2- ylmethyl)xanthine;
l-[N-[3(R)-pyrrolidinyl]glycyl]pyrrolidin-2(R)-yl boronic acid;
4(5)-fluoro-l-[2-[(lΛ,35)-3-(lH-l,2,4-triazol-l- ylmethyl)cyclopentylamino]acetyl]pyrrolidme-2(S)-carbonitrile;
1 -[(2S,3S,l lb5)-2-amino-9,10-dimethoxy-2,3,4,6,7,l lb-hexahydro- IH- pyrido[2,l-a]isoquinoh'n-3-yl]-4(S)-(fluoromethyl)pyrroh'din-2-one;
(2S,4S)-2-cyano-4-fluoro- 1 -[(2-hydroxy- 1 , 1 -dimethyl)
ethylamino]acetylpyrrolidine;
8-(c^-hexahydro-pyrrolo[3,2-b]pyrrol- 1 -yl)-3-methyl-7-(3-methyl-but-2-enyl)- 1 - (2-oxo-2-phenylethyl)-3,7-dihydro-purine-2,6-dione;
1 -((3S,45)-4-amino- 1 -(4-(3,3-difluoropyrrolidin- 1 -yl)- 1 ,3,5-triazin-2- yl)pyrrolidin-3-yl)-5,5difluoropiperidin-2-one;
(R)-2-((6-(3-aminopiperidin-l-yl)-3-methyl-2,4-dioxo-3,4-dihydropyrimidin- l(2H)-yl)methyl)-4-fluorobenzonitrile;
5- {(S)-2-[2-((S)-2-cyano-pyrrolid in- 1 -yl)-2-oxo-ethylamino]-propyl} -5-(1H- tetrazol-5-yl)10,l l-dihydro-5H-dibenzo[a,d]cycloheptene-2,8-dicarboxylic acid bis- dimethylamide;
((2ιS',4ιS)-4-(4-(3-methyl- 1 -phenyl- lH-pyrazol-5-yl)piperazin- 1 -yl)pyrrolidin-2- yl)(thiazolidin- 3 -yl)methanone ;
(2S,4S)- 1 -[2-[(4-ethoxycarbonylbicyclo[2.2.2]oct- 1 -yl)amino] acetyl] -4- fluoropyrrolidine-2-carbonitrile;
6-[(3R)-3-amino-piperidin-l-yl]-5-(2-chloro-5-fluoro-benzyl)-l,3-dimethyl- l,5dihydro-pyrrolo[3,2-d]pyrimidine-2,4-dione;
2-({6-[(3R)-3-amino-3-methylpiperidin-l-yl]-l,3-dimethyl-2,4-dioxo-l,2,3,4- tetrahydro-5H-pyrrolo[3,2-d]pyrimidin-5-yl}methyl)-4-fluorobenzonitrile;
(2S)-l-{[2-(5-methyl-2-phenyl-oxazol-4-yl)-ethylamino]-acetyl}-pyrrolidine-2- carbonitrile;
(2S)- 1 - { [ 1 , 1 -dimethyl-3-(4-pyridin-3-yl-imidazol- 1 -yl)-propylamino]-acetyl} - pyrrolidine-2-carbonitrile;
(3,3-difluoropyrrolidin- 1 -yl)-((2S,4S)-4-(4-(pyrimidin-2yl)piperazin- 1 - yl)pyrrolidin-2-yl)methanone; (2S,4S)-l-[(25)-2-amino-3,3-bis(4-fluorophenyl)propanoyl]-4-fluoropyrrolidine- 2-carbonitrile;
(2£,5R)-5-ethynyl- 1 - {N-(4-methyl- 1 -(4-carboxy-pyridin-2-yl)piperidin-4- yl)glycyl}pyrrolidine-2-carbonitrile; and
(lS,6R)-3- {[3-(trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)- yl]carbonyl} -6-(2,4,5-trifluorophenyl)cyclohex-3-en- 1 -amine.
41. A twin pack comprising a compound according to claim 1 and an inhibitor of DPP-IV as a combined preparation for the treatment of diabetes mellitus or a condition related thereto.
42. A twin pack according to claim 41, wherein said compound and said inhibitor of DPP-IV are admixed with the same or different pharmaceutically acceptable carriers.
43. A twin pack according to claim 41, wherein said compound and said inhibitor of DPP-IV are admixed with different pharmaceutically acceptable carriers.
44. A twin pack according to claim 43, wherein said different pharmaceutically acceptable carriers are suitable for administration by different routes.
45. A twin pack according to any one of claims 41 to 44, wherein said condition related to diabetes mellitus is selected from the group consisting of hyperglycemia, impaired glucose tolerance, insulin resistance, pancreatic beta-cell insufficiency, enteroendocrine cell insufficiency, glucosuria, metabolic acidosis, cataracts, diabetic nephropathy, diabetic neuropathy, diabetic retinopathy, diabetic coronary artery disease, diabetic cerebrovascular disease, diabetic peripheral vascular disease, metabolic syndrome, hyperlipidemia, atherosclerosis, stroke, hypertension, and obesity.
46. A twin pack according to any one of claims 41 to 45, wherein said inhibitor of DPP-IV is selected from the following compounds and pharmaceutically acceptable salts, solvates, and hydrates thereof:
3(R)-amino-l-[3-(trifluoromethyl)-5,6,7,8-tetrahydro[l,2,4]triazolo[4,3- a]pyrazin-7-yl]-4-(2,4,5-trifluorophenyl) butan- 1 -one;
1 -[2-(3-hydroxyadamant- 1 -ylamino)acetyl]pyrrolidine-2(S)-carbonitrile; (lS,3S,5S>2-[2(S>amino-2-(3-hydroxyadamantan-l-yl)acetyl]-2- azabicyclo[3.1.0]hexane-3-carbonitrile;
2-[6-[3(R)-aminopiperidin-l-yl]-3-methyl-2,4-dioxo-l,2,3,4- tetrahydropyrimidin- 1 -ylmethyl]benzonitrile;
8-[3(R)-aminopiperidin- 1 -yl]-7-(2-butynyl)-3-methyl- 1 -(4-methylquinazolin-2- ylmethyl)xanthine;
l-[N-[3(R)-pyrrolidinyl]glycyl]pyrrolidin-2(R)-yl boronic acid;
4(5)-fluoro-l-[2-[(lΛ,35)-3-(lH-l,2,4-triazol-l- ylmethyl)cyclopentylamino]acetyl]pyrrolidme-2(S)-carbonitrile;
l-[(2S,3S,l lbS)-2-amino-9,10-dimethoxy-2,3,4,6,7,l lb-hexahydro-lH- pyrido[2,l-a]isoquinoh'n-3-yl]-4(S)-(fluoromethyl)pyrroh'din-2-one;
(2S,4S)-2-cyano-4-fluoro- 1 -[(2-hydroxy- 1 , 1 -dimethyl)
ethylamino]acetylpyrrolidine;
8-(cw-hexahydro-pyrrolo[3,2-b]pyrrol- 1 -yl)-3-methyl-7-(3-methyl-but-2-enyl)- 1 - (2-oxo-2-phenylethyl)-3,7-dihydro-purine-2,6-dione;
1 -((3S,45)-4-amino- 1 -(4-(3,3-difluoropyrrolidin- 1 -yl)- 1 ,3,5-triazin-2- yl)pyrrolidin-3-yl)-5,5difluoropiperidin-2-one;
(R)-2-((6-(3-aminopiperidin-l-yl)-3-methyl-2,4-dioxo-3,4-dihydropyrimidin- l(2H)-yl)methyl)-4-fluorobenzonitrile;
S-K^^-P-^^-cyano-pyrrolid in-l-y^^-oxo-ethylaminol-propylj-S-ClH- tetrazol-5-yl)10,l l-dihydro-5H-dibenzo[a,d]cycloheptene-2,8-dicarboxylic acid bis- dimethylamide;
((2ιS',4ιS)-4-(4-(3-methyl- 1 -phenyl- lH-pyrazol-5-yl)piperazin- 1 -yl)pyrrolidin-2- yl)(thiazolidin- 3 -yl)methanone ;
(2S,4S)- 1 -[2-[(4-ethoxycarbonylbicyclo[2.2.2]oct- 1 -yl)amino] acetyl] -4- fluoropyrrolidine-2-carbonitrile;
6-[(3R)-3-amino-piperidin- 1 -yl]-5-(2-chloro-5-fluoro-benzyl)- 1 ,3-dimethyl- l,5dihydro-pyrrolo[3,2-d]pyrimidine-2,4-dione;
2-({6-[(3R)-3-amino-3-methylpiperidin-l-yl]-l,3-dimethyl-2,4-dioxo-l,2,3,4- tetrahydro-5H-pyrrolo[3,2-d]pyrimidin-5-yl}methyl)-4-fluorobenzonitrile;
(2S)-l-{[2-(5-methyl-2-phenyl-oxazol-4-yl)-ethylamino]-acetyl}-pyrrolidine-2- carbonitrile;
(2S)- 1 - {[1 , 1 -dimethyl-3-(4-pyridin-3-yl-imidazol- 1 -yl)-propylamino]-acetyl} - pyrrolidine-2-carbonitrile; (3,3-difluoropyrrolidin- 1 -yl)-((2£,4,S)-4-(4-(pyrimidin-2yl)piperazin- 1 - yl)pyrrolidin-2-yl)methanone;
(2S,4S)-l-[(2S)-2-amino-3,3-bis(4-fluorophenyl)propanoyl]-4-fluoropyrrolidine- 2-carbonitrile;
(2£,5R)-5-ethynyl- 1 - {N-(4-methyl- 1 -(4-carboxy-pyridin-2-yl)piperidin-4- yl)glycyl}pyrrolidine-2-carbonitrile; and
(lS,6R)-3- {[3-(trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)- yl]carbonyl} -6-(2,4,5-trifluorophenyl)cyclohex-3-en- 1 -amine.
47. A kit comprising (a) a first package comprising a compound according to claim 1 or a pharmaceutical composition thereof, and (b) a second package comprising an inhibitor of DPP-IV or a pharmaceutical composition thereof.
48. A kit according to claim 47, wherein said inhibitor of DPP-IV is selected from the
following compounds and pharmaceutically acceptable salts, solvates, and hydrates thereof:
3(R)-amino-l-[3-(trifluoromethyl)-5,6,7,8-tetrahydro[l,2,4]triazolo[4,3- a]pyrazin-7-yl]-4-(2,4,5-trifluorophenyl) butan- 1 -one;
1 -[2-(3-hydroxyadamant- 1 -ylamino)acetyl]pyrrolidine-2(S)-carbonitrile;
(lS,3S,5S>2-[2(S>amino-2-(3-hydroxyadamantan-l-yl)acetyl]-2- azabicyclo[3.1.0]hexane-3-carbonitrile;
2-[6-[3(R)-aminopiperidin-l-yl]-3-methyl-2,4-dioxo- 1,2,3,4- tetrahydropyrimidin- 1 -ylmethyljbenzonitrile;
8-[3(R)-aminopiperidin- 1 -yl]-7-(2-butynyl)-3-methyl- 1 -(4-methylquinazolin-2- ylmethyl)xanthine;
l-[N-[3(R)-pyrrolidinyl]glycyl]pyrrolidin-2(R)-yl boronic acid;
4(5)-fluoro-l-[2-[(lΛ,35)-3-(lH-l,2,4-triazol-l- ylmethyl)cyclopentylammo]acetyl]pyrrolidine-2(S)-carbonitrile;
l-[(2S,3S,l lbS)-2-amino-9,10-dimethoxy-2,3,4,6,7,l lb-hexahydro-lH- pyrido[2, 1 -a]isoquinolin-3-yl]-4(S)-(fluoromethyl)pyrrolidin-2-one;
(2S,4S)-2-cyano-4-fluoro- 1 -[(2-hydroxy- 1 , 1 -dimethyl)
ethylamino]acetylpyrrolidine;
8-(cw-hexahydro-pyrrolo[3,2-b]pyrrol- 1 -yl)-3-methyl-7-(3-methyl-but-2-enyl)- 1 - (2-oxo-2-phenylethyl)-3,7-dihydro-purine-2,6-dione; 1 -((3S,45)-4-amino- 1 -(4-(3,3-difluoropyrrolidin- 1 -yl)- 1 ,3,5-triazin-2- yl)pyrrolidin-3-yl)-5,5difluoropiperidin-2-one;
(R)-2-((6-(3-ammopiperidin-l-yl)-3-methyl-2,4-dioxo-3,4-dihydropyrimidin- l(2H)-yl)methyl)-4-fluorobenzonitrile;
S-K^^-P-^^-cyano-pyrrolid in-l-y^^-oxo-ethylaminol-propylj-S-ClH- tetrazol-5-yl)10,l l-dihydro-5H-dibenzo[a,d]cycloheptene-2,8-dicarboxylic acid bis- dimethylamide;
((2S, 4ιS)-4-(4-(3 -methyl- 1 -phenyl- lH-pyrazol-5-yl)piperazin- 1 -yl)pyrrolidin-2- yl)(thiazolidin- 3 -yl)methanone ;
(2S,4S)- 1 -[2-[(4-ethoxycarbonylbicyclo[2.2.2]oct- 1 -yl)amino] acetyl] -4- fluoropyrrolidine-2-carbonitrile;
6-[(3R)-3-amino-piperidin- 1 -yl]-5-(2-chloro-5-fluoro-benzyl)- 1 ,3-dimethyl- l,5dihydro-pyrrolo[3,2-d]pyrimidine-2,4-dione;
2-({6-[(3R)-3-amino-3-methylpiperidin-l-yl]-l,3-dimethyl-2,4-dioxo-l,2,3,4- tetrahydro-5H-pyrrolo[3,2-d]pyrimidin-5-yl}methyl)-4-fluorobenzonitrile;
(2S)-l-{[2-(5-methyl-2-phenyl-oxazol-4-yl)-ethylamino]-acetyl}-pyrrolidine-2- carbonitrile;
(2S)- 1 - {[1 , 1 -dimethyl-3-(4-pyridin-3-yl-imidazol- 1 -yl)-propylamino] -acetyl} - pyrrolidine-2-carbonitrile;
(3,3-difluoropyrrolidin- 1 -yl)-((2S,4S)-4-(4-(pyrimidin-2yl)piperazin- 1 - yl)pyrrolidin-2-yl)methanone;
(2S,4S)-l-[(2S)-2-amino-3,3-bis(4-fluorophenyl)propanoyl]-4-fluoropyrrolidine- 2-carbonitrile;
(2S,5R)-5-ethynyl- 1 - {N-(4-methyl- 1 -(4-carboxy-pyridin-2-yl)piperidin-4- yl)glycyl}pyrrolidine-2-carbonitrile; and
(lS,6R)-3-{[3-(trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8H)- yljcarbonyl} -6-(2,4,5-trifluorophenyl)cyclohex-3-en- 1 -amine.
PCT/US2010/041325 2009-07-09 2010-07-08 Piperidine derivative and its use for the treatment of diabets and obesity WO2011005929A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US27050209P 2009-07-09 2009-07-09
US61/270,502 2009-07-09

Publications (1)

Publication Number Publication Date
WO2011005929A1 true WO2011005929A1 (en) 2011-01-13

Family

ID=42751753

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/041325 WO2011005929A1 (en) 2009-07-09 2010-07-08 Piperidine derivative and its use for the treatment of diabets and obesity

Country Status (3)

Country Link
AR (1) AR077642A1 (en)
TW (1) TW201105327A (en)
WO (1) WO2011005929A1 (en)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012099915A1 (en) * 2011-01-19 2012-07-26 Hongwen Zhu Thiazolidine derivatives and their therapeutic use
WO2012170867A1 (en) 2011-06-09 2012-12-13 Rhizen Pharmaceuticals Sa Novel compounds as modulators of gpr-119
WO2014011926A1 (en) 2012-07-11 2014-01-16 Elcelyx Therapeutics, Inc. Compositions comprising statins, biguanides and further agents for reducing cardiometabolic risk
US8933083B2 (en) 2003-01-14 2015-01-13 Arena Pharmaceuticals, Inc. 1,2,3-trisubstituted aryl and heteroaryl derivatives as modulators of metabolism and the prophylaxis and treatment of disorders related thereto such as diabetes and hyperglycemia
US9096641B2 (en) 2013-06-05 2015-08-04 Evestra, Inc. Imidazolyl progesterone antagonists
US9776987B2 (en) 2013-11-26 2017-10-03 Chong Kun Dang Pharmaceutical Corp Amide derivatives for GPR119 agonist
US9944600B2 (en) 2012-06-12 2018-04-17 Chong Kun Dang Pharmaceutical Corp. Piperidine derivatives for GPR119 agonist
US10894787B2 (en) 2010-09-22 2021-01-19 Arena Pharmaceuticals, Inc. Modulators of the GPR119 receptor and the treatment of disorders related thereto
US11007175B2 (en) 2015-01-06 2021-05-18 Arena Pharmaceuticals, Inc. Methods of treating conditions related to the S1P1 receptor
US11534424B2 (en) 2017-02-16 2022-12-27 Arena Pharmaceuticals, Inc. Compounds and methods for treatment of primary biliary cholangitis
US11884626B2 (en) 2015-06-22 2024-01-30 Arena Pharmaceuticals, Inc. Crystalline L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclo-penta [b]indol-3-yl)acetic acid(Compound1) for use in S1P1 receptor-associated disorders

Citations (233)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991016339A1 (en) 1990-04-14 1991-10-31 New England Medical Center Hospitals, Inc. Inhibitors of dipeptidyl-aminopeptidase type iv
DD296075A5 (en) 1989-08-07 1991-11-21 Martin-Luther-Universitaet Halle-Wittenberg,De PROCESS FOR THE PREPARATION OF NEW INHIBITORS OF DIPEPTIDYL PEPTIDASE IV
WO1993008259A2 (en) 1991-10-22 1993-04-29 New England Medical Center Hospitals, Inc. Inhibitors of dipeptidyl-aminopeptidase type iv
WO1993010127A1 (en) 1991-11-22 1993-05-27 Boehringer Ingelheim Pharmaceuticals, Inc. Method for making a prolineboronate ester
WO1995015309A1 (en) 1993-12-03 1995-06-08 Ferring B.V. Dp-iv-serine protease inhibitors
WO1995029691A1 (en) 1994-04-28 1995-11-09 Georgia Tech Research Corporation Proline phosphonate derivatives
US5571815A (en) * 1992-03-14 1996-11-05 Hoechst Aktiengesellschaft Substituted pyrimidines, process for their preparation, and their use as pesticides and fungicides
DE19616486A1 (en) 1996-04-25 1997-10-30 Knoell Hans Forschung Ev Method of lowering blood glucose levels in mammals
JPH1081666A (en) 1996-06-12 1998-03-31 Ishihara Sangyo Kaisha Ltd Phthalimide derivative or salt thereof, its production and medicinal composition containing the same
WO1998018763A1 (en) 1996-10-25 1998-05-07 Tanabe Seiyaku Co., Ltd. Tetrahydroisoquinoline derivatives
WO1998019998A2 (en) 1996-11-07 1998-05-14 Novartis Ag N-substituted 2-cyanopyrrolidines
JPH10182613A (en) 1996-10-25 1998-07-07 Tanabe Seiyaku Co Ltd Tetraphydroisoquinoline derivative
WO1998050066A1 (en) 1997-05-07 1998-11-12 Trustees Of Tufts College Potentiation of the immune response through delivery of compounds binding a cytoplasmic dipeptidase
WO1998050046A1 (en) 1997-05-07 1998-11-12 Trustees Of Tufts College Use of cd26 inhibitor for the manufacture of a medicament for the treatment of hiv
WO1999016864A1 (en) 1997-09-29 1999-04-08 Point Therapeutics, Inc. Stimulation of hematopoietic cells in vitro
WO1999025719A1 (en) 1997-11-18 1999-05-27 Zaidan Hojin Biseibutsu Kagaku Kenkyu Kai Novel physiologically active substance sulphostin, process for producing the same, and use thereof
WO1999056753A1 (en) 1998-05-04 1999-11-11 Point Therapeutics, Inc. Hematopoietic stimulation
DE19823831A1 (en) 1998-05-28 1999-12-02 Probiodrug Ges Fuer Arzneim New pharmaceutical use of isoleucyl thiazolidide and its salts
WO1999062914A1 (en) 1998-06-05 1999-12-09 Point Therapeutics, Inc. Cyclic boroproline compounds
WO1999067278A1 (en) 1998-06-24 1999-12-29 Probiodrug Gesellschaft für Arzneimittelforschung mbH Prodrugs of dipeptidyl peptidase iv inhibitors
DE19834591A1 (en) 1998-07-31 2000-02-03 Probiodrug Ges Fuer Arzneim Use of substances that decrease the activity of dipeptidyl peptidase IV to increase blood sugar levels, e.g. for treating hypoglycemia
CA2339537A1 (en) 1998-08-21 2000-03-02 Barbara Wallner Regulation of substrate activity
WO2000023421A1 (en) 1998-10-22 2000-04-27 Idun Pharmaceuticals, Inc. (SUBSTITUTED)ACYL DIPEPTIDYL INHIBITORS OF THE ICE/ced-3 FAMILY OF CYSTEINE PROTEASES
WO2000031258A2 (en) 1998-11-20 2000-06-02 Arena Pharmaceuticals, Inc. Human orphan g protein-coupled receptors
WO2000034241A1 (en) 1998-12-10 2000-06-15 Novartis Ag N-substituted 2-cyanopyrrolidines
JP2000191616A (en) 1998-12-24 2000-07-11 Senju Pharmaceut Co Ltd New peptidylaldehyde derivative and medicine containing the same
WO2000056297A2 (en) 1999-03-23 2000-09-28 Ferring B.V. Pharmaceutical compositions comprising dipeptidy peptidase iv inhibitors for the promotion of growth
WO2000056296A2 (en) 1999-03-23 2000-09-28 Ferring Bv Compositions for improving fertility
WO2000069868A1 (en) 1999-05-17 2000-11-23 Zaidan Hojin Biseibutsu Kagaku Kenkyu Kai Sulphostin analogues and processes for the preparation of sulphostin and analogues thereof
WO2000071135A1 (en) 1999-05-25 2000-11-30 Point Therapeutics, Inc. Anti-tumor comprising boroproline compounds
WO2001034594A1 (en) 1999-11-12 2001-05-17 Guilford Pharmaceuticals, Inc. Dipeptidyl peptidase iv inhibitors and methods of making and using dipeptidyl peptidase iv inhibitors
WO2001052825A2 (en) 2000-01-21 2001-07-26 Novartis Ag Combinations comprising dipeptidylpeptidase-iv inhibitors and antidiabetic agents
WO2001055105A1 (en) 2000-01-24 2001-08-02 Novo Nordisk A/S N-substituted 2-cyanopyroles and -pyrrolines which are inhibitors of the enzyme dpp-iv
WO2001068603A2 (en) 2000-03-10 2001-09-20 Bristol-Myers Squibb Co. Cyclopropyl-fused pyrrolidine-based inhibitors of dipeptidyl iv, processes for their preparation, and their use
WO2001081304A1 (en) 2000-04-26 2001-11-01 Ferring Bv Inhibitors of dipeptidyl peptidase iv
WO2001081337A1 (en) 2000-04-26 2001-11-01 Ferring B.V. Inhibitors of dipeptidyl peptidase iv
WO2001096295A2 (en) 2000-06-13 2001-12-20 Novartis Ag 2-cyanopyrrolidine derivatives and their use as medicaments
WO2001097808A1 (en) 2000-06-19 2001-12-27 Smithkline Beecham Plc Combinations of depeptidyl peptidase iv inhibitors and other antidiabetic agents for the treatment of diabete mellitus
WO2002002560A2 (en) 2000-07-04 2002-01-10 Novo Nordisk A/S Purine-2,6-diones which are inhibitors of the enzyme dipeptidyl peptidase iv (dpp-iv)
US20020006899A1 (en) 1998-10-06 2002-01-17 Pospisilik Andrew J. Use of dipeptidyl peptidase IV effectors for lowering blood pressure in mammals
WO2002014271A1 (en) 2000-08-10 2002-02-21 Mitsubishi Pharma Corporation Proline derivatives and use thereof as drugs
WO2002030890A1 (en) 2000-10-06 2002-04-18 Tanabe Seiyaku Co., Ltd. Nitrogenous five-membered ring compounds
US6380398B2 (en) 2000-01-04 2002-04-30 Novo Nordisk A/S Therapeutically active and selective heterocyclic compounds that are inhibitors of the enzyme DPP-IV
WO2002034900A1 (en) 2000-10-27 2002-05-02 The University Of Sydney Dipeptidyl peptidases
WO2002038541A1 (en) 2000-11-10 2002-05-16 Taisho Pharmaceutical Co., Ltd. Cyanopyrrolidine derivatives
US6410508B1 (en) 1998-10-07 2002-06-25 Med College Georgia Res Inst Glucose-dependent insulinotropic peptide for use as an osteotropic hormone
WO2002051836A1 (en) 2000-12-27 2002-07-04 Kyowa Hakko Kogyo Co., Ltd. Dipeptidyl peptidase iv inhibitor
WO2002055088A1 (en) 2001-01-16 2002-07-18 Nippon Kayaku Kabushiki Kaisha Remedies for bone marrow suppression and infectious diseases and white blood cell count elevating agents
US6432969B1 (en) 2000-06-13 2002-08-13 Novartis Ag N-(substituted glycyl)-2 cyanopyrrolidines, pharmaceutical compositions containing them and their use in inhibiting dipeptidyl peptidase-IV
WO2002062764A1 (en) 2001-02-02 2002-08-15 Takeda Chemical Industries, Ltd. Fused heterocyclic compounds
WO2002068420A1 (en) 2001-02-24 2002-09-06 Boehringer Ingelheim Pharma Gmbh & Co. Kg Xanthine derivative, production and use thereof as a medicament
JP2002265439A (en) 2001-03-08 2002-09-18 Mitsubishi Pharma Corp Cyanopyrrolidine derivative and its use for medicine
EP1245568A1 (en) 2001-03-28 2002-10-02 Les Laboratoires Servier Sulfonyl derivatives of amino acids and their use as inhibitors of dipeptidyl-peptidase IV (DPP IV)
WO2002076450A1 (en) 2001-03-27 2002-10-03 Merck & Co., Inc. Dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2002083109A1 (en) 2001-04-11 2002-10-24 Ferring Bv Treatment of type 2 diabetes with inhibitors of dipeptidyl peptidase iv
WO2002083128A1 (en) 2001-04-12 2002-10-24 Bristol-Myers Squibb Company 2,1-oxazoline and 1,2-pyrazoline-based inhibitors of dipeptidyl peptidase iv and method
EP1258476A1 (en) 2001-05-15 2002-11-20 Les Laboratoires Servier Alpha-amino acid derivatives, method for their preparation and their use as dipeptidyl-peptidase IV inhibitors (DPP IV)
JP2002356472A (en) 2000-10-06 2002-12-13 Tanabe Seiyaku Co Ltd Nitrogen-containing five-membered ring compound
JP2002356471A (en) 2000-10-06 2002-12-13 Tanabe Seiyaku Co Ltd Aliphatic nitrogen-containing five-membered ring compound
WO2003000250A1 (en) 2001-06-25 2003-01-03 Ferring Bv 3-fluoro-pyrrolidines as antidiabetic agents
WO2003000180A2 (en) 2001-06-20 2003-01-03 Merck & Co., Inc. Dipeptidyl peptidase inhibitors for the treatment of diabetes
WO2003000181A2 (en) 2001-06-20 2003-01-03 Merck & Co., Inc. Dipeptidyl peptidase inhibitors for the treatment of diabetes
WO2003002593A2 (en) 2001-06-27 2003-01-09 Probiodrug Ag Peptide structures useful for competitive modulation of dipeptidyl peptidase iv catalysis
WO2003002595A2 (en) 2001-06-27 2003-01-09 Probiodrug Ag Dipeptidyl peptidase iv inhibitors and their uses as anti-cancer agents
WO2003002531A2 (en) 2001-06-27 2003-01-09 Smithkline Beecham Corporation Fluoropyrrolidines as dipeptidyl peptidase inhibitors
WO2003002553A2 (en) 2001-06-27 2003-01-09 Smithkline Beecham Corporation Fluoropyrrolidines as dipeptidyl peptidase inhibitors
WO2003002530A2 (en) 2001-06-27 2003-01-09 Smithkline Beecham Corporation Pyrrolidines as dipeptidyl peptidase inhibitors
WO2003004496A1 (en) 2001-07-03 2003-01-16 Novo Nordisk A/S Dpp-iv-inhibiting purine derivatives for the treatment of diabetes
WO2003004498A1 (en) 2001-07-06 2003-01-16 Merck & Co., Inc. Beta-amino tetrahydroimidazo (1, 2-a) pyrazines and tetrahydrotrioazolo (4, 3-a) pyrazines as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2003022871A2 (en) 2001-09-06 2003-03-20 Probiodrug Ag Peptides having a c- terminal hydroxylamino group as inhibitors of dipeptidyl peptidase i
WO2003024965A2 (en) 2001-09-19 2003-03-27 Novo Nordisk A/S Heterocyclic compounds that are inhibitors of the enzyme dpp-iv
WO2003024942A1 (en) 2001-09-14 2003-03-27 Mitsubishi Pharma Corporation Thiazolidine derivative and medicinal use thereof
WO2003035057A1 (en) 2001-10-23 2003-05-01 Ferring B.V. Inhibitors of dipeptidyl peptidase iv
WO2003035067A1 (en) 2001-10-23 2003-05-01 Ferring B.V. Novel dipeptidyl peptidase iv (dp-iv) inhibitors as anti-diabetic agents
WO2003037327A1 (en) 2001-10-26 2003-05-08 F. Hoffmann-La-Roche Ag N-substituted pyrrolidin derivatives as dipeptidyl peptidase iv inhibitors
WO2003038123A2 (en) 2001-10-31 2003-05-08 Novartis Ag Methods to treat diabetes and related conditions based on polymorphisms in the tcf1 gene
WO2003040174A2 (en) 2001-11-09 2003-05-15 Probiodrug Ag Substituted amino ketone compounds
CA2466870A1 (en) 2001-11-26 2003-06-05 Trustees Of Tufts College Methods for treating autoimmune disorders, and reagents related thereto
US20030105077A1 (en) 2001-07-03 2003-06-05 Kanstrup Anders Bendtz Heterocyclic compounds that are inhibitors of the enzyme DPP-IV
US20030119750A1 (en) 2001-06-27 2003-06-26 Hans-Ulrich Demuth Use of dipeptidyl peptidase IV inhibitors
US20030119738A1 (en) 2001-09-06 2003-06-26 Andre Niestroj Novel inhibitors of dipeptidyl peptidase I
US20030125304A1 (en) 2001-11-09 2003-07-03 Hans-Ulrich Demuth Substituted amino ketone compounds
US20030130199A1 (en) 2001-06-27 2003-07-10 Von Hoersten Stephan Dipeptidyl peptidase IV inhibitors and their uses as anti-cancer agents
WO2003055881A1 (en) 2001-12-27 2003-07-10 F. Hoffmann-La Roche Ag Pyrido(2,1-a)isoquinoline derivatives as dpp-iv inhibitors
WO2003057666A2 (en) 2001-12-26 2003-07-17 Guilford Pharmaceuticals Inhibitors of dipeptidyl peptidase iv
WO2003068757A1 (en) 2002-02-13 2003-08-21 F. Hoffmann-La Roche Ag Novel pyridin- and pyrimidin-derivatives
WO2003068748A1 (en) 2002-02-13 2003-08-21 F. Hoffmann-La Roche Ag Novel pyridine- and quinoline-derivatives
JP2003238566A (en) 2001-02-02 2003-08-27 Takeda Chem Ind Ltd Condensed heterocyclic compound
EP1338651A1 (en) 2000-12-01 2003-08-27 Yamanouchi Pharmaceutical Co. Ltd. Method of screening remedy for diabetes
EP1338592A1 (en) 2002-02-22 2003-08-27 Nippon Zoki Pharmaceutical Co., Ltd. Novel 2-phenylpiperazine derivatives
US20030162820A1 (en) 2002-02-28 2003-08-28 Hans-Ulrich Demuth Glutaminyl based DPIV inhibitors
WO2003072528A2 (en) 2002-02-08 2003-09-04 Idun Pharmaceuticals, Inc. (substituted)acyl dipeptidyl inhibitors of the ice/ced-3 family of cysteine proteases
US6617340B1 (en) 1999-07-29 2003-09-09 Novartis Ag N-(substituted glycyl)-pyrrolidines, pharmaceutical compositions containing them and their use in inhibiting dipeptidyl peptidase-IV
WO2003074500A2 (en) 2002-03-06 2003-09-12 Sanofi-Aventis N-aminoacetyl-pyrrolidine-2-carbonitriles and their use as ddp-iv inhibitors
WO2003080633A1 (en) 2002-03-25 2003-10-02 Nippon Kayaku Kabushiki Kaisha Novel $g(a)-amino-n-(diaminophosphinyl)lactam derivative
WO2003082817A2 (en) 2002-03-25 2003-10-09 Merck & Co., Inc. Beta-amino heterocyclic dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2003084940A1 (en) 2002-04-08 2003-10-16 Alangudi Sankaranarayanan Thiazolidine-4-carbonitriles and analogues and their use as dipeptidyl-peptidas inhibitors
JP2003300977A (en) 2002-04-10 2003-10-21 Sumitomo Pharmaceut Co Ltd Xanthine derivative
JP2003327532A (en) 2002-05-10 2003-11-19 Takeda Chem Ind Ltd Peptidase inhibitor
WO2003095425A1 (en) 2002-05-09 2003-11-20 Taisho Pharmaceutical Co.,Ltd. Cyanopyrrolidine derivatives
WO2003099279A1 (en) 2002-05-29 2003-12-04 Novartis Ag Combination of a dpp iv inhibitor and a cardiovascular compound
WO2003101448A1 (en) 2002-06-03 2003-12-11 Novartis Ag The use of substituted cyanopyrrolidines and combination preparations containing them for treating hyperlipidemia and associated diseases
WO2003101958A2 (en) 2002-06-04 2003-12-11 Pfizer Products Inc. Flourinated cyclic amides as dipeptidyl peptidase iv inhibitors
WO2003104229A1 (en) 2002-06-06 2003-12-18 エーザイ株式会社 Novel fused imidazole derivative
WO2003106456A2 (en) 2002-06-14 2003-12-24 Sanofi-Synthelabo New compounds
WO2003105763A2 (en) 2002-06-14 2003-12-24 Amylin Pharmaceuticals, Inc. Prevention and/or treatment of inflammatory bowel disease using pyy or agonists thereof
WO2004000327A1 (en) 2002-06-24 2003-12-31 Zaidan Hojin Biseibutsu Kagaku Kenkyu Kai Therapeutic agent for type 2 diabetes
JP2004002368A (en) 2002-04-04 2004-01-08 Tanabe Seiyaku Co Ltd Pharmaceutical composition
JP2004002367A (en) 2002-04-04 2004-01-08 Tanabe Seiyaku Co Ltd Pharmaceutical composition
WO2004004661A2 (en) 2002-07-09 2004-01-15 Point Therapeutics, Inc. Boroproline compound combination therapy
WO2004007446A1 (en) 2002-07-10 2004-01-22 Yamanouchi Pharmaceutical Co., Ltd. Novel azetidine derivative or salt thereof
WO2004007468A1 (en) 2002-07-15 2004-01-22 Merck & Co., Inc. Piperidino pyrimidine dipeptidyl peptidase inhibitors for the treatment of diabetes
WO2004009544A1 (en) 2002-07-23 2004-01-29 Yamanouchi Pharmaceutical Co., Ltd. 2-cyano-4-fluoropyrrolidine derivative or its salt
JP2004026820A (en) 2002-05-09 2004-01-29 Taisho Pharmaceut Co Ltd Dipeptidyl peptidase iv inhibitor
JP2004035574A (en) 2000-10-06 2004-02-05 Tanabe Seiyaku Co Ltd Aliphatic nitrogenous five-membered ring compound
JP2004043429A (en) 2002-02-25 2004-02-12 Eisai Co Ltd New xanthine derivative and dppiv inhibitor
WO2004014860A2 (en) 2002-08-08 2004-02-19 Takeda Pharmaceutical Company Limited Fused heterocyclic compounds as peptidase inhibitors
DE10238243A1 (en) 2002-08-21 2004-03-04 Boehringer Ingelheim Pharma Gmbh & Co. Kg New 8-(3-amino-piperidin-1-yl)-xanthine derivatives are dipeptidylpeptidase-IV inhibitors useful for, e.g. treating diabetes mellitus, arthritis or obesity
DE10238470A1 (en) 2002-08-22 2004-03-04 Boehringer Ingelheim Pharma Gmbh & Co. Kg New xanthine derivatives, their production and their use as medicines
WO2004018469A1 (en) 2002-08-22 2004-03-04 Boehringer Ingelheim Pharma Gmbh & Co. Kg Novel purine derivatives, production and use thereof as medicaments
WO2004018468A2 (en) 2002-08-21 2004-03-04 Boehringer Ingelheim Pharma Gmbh & Co. Kg 8-[3-amino-piperidin-1-yl]-xanthines, the production thereof and the use of the same as medicaments
WO2004020407A1 (en) 2002-08-29 2004-03-11 Taisho Pharmaceutical Co.,Ltd. Benzenesulfonate of 4-fluoro-2-cyanopyrrolidine derivative
US6710040B1 (en) 2002-06-04 2004-03-23 Pfizer Inc. Fluorinated cyclic amides as dipeptidyl peptidase IV inhibitors
WO2004026822A2 (en) 2002-09-19 2004-04-01 Abbott Laboratories Pharmaceutical compositions as inhibitors of dipeptidyl peptidase-iv (dpp-iv)
WO2004033455A2 (en) 2002-10-08 2004-04-22 Novo Nordisk A/S Hemisuccinate salts of heterocyclic dpp-iv inhibitors
WO2004032836A2 (en) 2002-10-07 2004-04-22 Merck & Co., Inc. Antidiabetic beta-amino heterocylcic dipeptidyl peptidase inhibitors
US20040082570A1 (en) 2002-02-25 2004-04-29 Eisai Co., Ltd. Xanthine derivative and DPPIV inhibitor
WO2004037169A2 (en) 2002-10-18 2004-05-06 Merck & Co., Inc. Beta-amino heterocyclic dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2004037181A2 (en) 2002-10-23 2004-05-06 Bristol-Myers Squibb Company Glycinenitrile-based inhibitors of dipeptidyl peptidase iv and methods
DE10251927A1 (en) 2002-11-08 2004-05-19 Boehringer Ingelheim Pharma Gmbh & Co. Kg New 1,7,8-trisubstituted xanthine derivatives, are dipeptidylpeptidase-IV inhibitors useful e.g. for treating diabetes mellitus type I or II, arthritis or obesity
US20040097510A1 (en) 2002-08-21 2004-05-20 Boehringer Ingelheim Pharma Gmbh & Co. Kg 8-[3-amino-piperidin-1-yl]-xanthines, the preparation thereof and their use as pharmaceutical compositions
WO2004041795A1 (en) 2002-10-30 2004-05-21 Guilford Pharmaceuticals Inc. Novel inhibitors of dipeptidyl peptidase iv
WO2004043940A1 (en) 2002-11-07 2004-05-27 Merck & Co., Inc. Phenylalanine derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2004046106A1 (en) 2002-11-18 2004-06-03 Pfizer Products Inc. Dipeptidyl peptidase iv inhibiting fluorinated cyclic amides
WO2004048379A1 (en) 2002-11-01 2004-06-10 Sumitomo Pharmaceuticals Co., Ltd. Xanthine compound
WO2004050022A2 (en) 2002-12-04 2004-06-17 Merck & Co., Inc. Phenylalanine derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2004050658A1 (en) 2002-12-03 2004-06-17 Boehringer Ingelheim Pharma Gmbh & Co. Kg Novel substituted imidazo-pyridinones and imidazo-pyridazeiones, the production and use thereof as medicaments
WO2004052850A2 (en) 2002-12-09 2004-06-24 Bristol-Myers Squibb Company Methods and compounds producing dipeptidyl peptidase iv inhibitors and intermediates thereof
WO2004052362A1 (en) 2002-12-10 2004-06-24 Novartis Ag Combination of an dpp-iv inhibitor and a ppar-alpha compound
DE10256264A1 (en) 2002-12-03 2004-06-24 Boehringer Ingelheim Pharma Gmbh & Co. Kg New trisubstituted dihydro-imidazo-pyridazinone or imidazo-pyridinone derivatives, useful as dipeptidylpeptidase-IV inhibitors for e.g. treating diabetes mellitus type I or II, arthritis or obesity
US20040138214A1 (en) 2002-11-08 2004-07-15 Boehringer Ingelheim Pharma Gmbh & Co. Kg Xanthine derivatives, the preparation thereof and their use as pharmaceutical compositions
WO2004058266A1 (en) 2002-12-20 2004-07-15 Merck & Co., Inc. 3-amino-4-phenylbutanoic acid derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2004064778A2 (en) 2003-01-17 2004-08-05 Merck & Co. Inc. 3-amino-4-phenylbutanoic acid derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2004065380A1 (en) 2003-01-14 2004-08-05 Arena Pharmaceuticals Inc. 1,2,3-trisubstituted aryl and heteroaryl derivatives as modulators of metabolism and the prpphylaxis and treatment of disorders related thereto such as diabetes and hyperglycemia
WO2004067509A1 (en) 2003-01-31 2004-08-12 Sanwa Kagaku Kenkyusho Co., Ltd. Compound inhibiting dipeptidyl peptidase iv
WO2004069162A2 (en) 2003-01-31 2004-08-19 Merck & Co., Inc. 3-amino-4-phenylbutanoic acid derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2004071454A2 (en) 2003-02-13 2004-08-26 Guilford Pharmaceuticals Inc. Substituted azetidine compounds as inhibitors of dipeptidyl peptidase iv
JP2004244412A (en) 2003-01-20 2004-09-02 Kotobuki Seiyaku Kk 2-cyanopyrrolidine derivative having substituent at 4-position, method for producing the same, and medicament containing the same
WO2004076433A1 (en) 2003-02-28 2004-09-10 Aic Dipeptidyl peptidase inhibitors
WO2004076434A1 (en) 2003-02-28 2004-09-10 Aic Dipeptidyl peptidase inhibitors
WO2004076413A2 (en) 2003-02-24 2004-09-10 Arena Pharmaceuticals, Inc. Phenyl- and pyridylpiperidine-derivatives as modulators of glucose metabolism
WO2004085661A2 (en) 2003-03-24 2004-10-07 Merck & Co., Inc Process to chiral beta-amino acid derivatives
WO2004085378A1 (en) 2003-03-19 2004-10-07 Merck & Co. Inc. Process for the preparation of chiral beta amino acid derivatives by asymmetric hydrogenation
US6803357B1 (en) 1998-02-02 2004-10-12 New England Medical Center Hospitals, Inc. Method of regulating glucose metabolism, and reagents related thereto
WO2004087053A2 (en) 2003-03-25 2004-10-14 Syrrx, Inc. Dipeptidyl peptidase inhibitors
WO2004087650A2 (en) 2003-03-27 2004-10-14 Merck & Co. Inc. Process and intermediates for the preparation of beta-amino acid amide dipeptidyl peptidase-iv inhibitors
EP1469873A2 (en) 2001-11-26 2004-10-27 Trustees of Tufts College Peptidomimetic inhibitors of post-proline cleaving enzymes
WO2004092128A1 (en) 2003-04-10 2004-10-28 Smithkline Beecham Corporation Anhydrous crystalline forms of (2s, 4s)-1-{(2r)-2-amino-3-‘4-methoxybenzyl)sulfonyl!-3-methylbutanoyl}-4-fluoropyrrolindine-2-carbonitrile
US6812350B2 (en) 2002-06-04 2004-11-02 Pfizer Inc. Synthesis of 3,3,4,4-tetrafluoropyrrolidine and novel dipeptidyl peptidase-IV inhibitor compounds
WO2004096806A1 (en) 2003-04-30 2004-11-11 Sumitomo Pharmaceuticals Co. Ltd. Fused imidazole derivative
JP2004315496A (en) 2002-08-08 2004-11-11 Takeda Chem Ind Ltd Fused heterocyclic ring compound
WO2004099134A2 (en) 2003-05-05 2004-11-18 Prosidion Ltd. Glutaminyl based dp iv-inhibitors
WO2004104215A2 (en) 2003-05-21 2004-12-02 Bayer Healthcare Ag Diagnostics and therapeutics for diseases associated with dipeptidylpeptidase 7 (dpp7)
WO2004103993A1 (en) 2003-05-14 2004-12-02 Syrrx, Inc. Dipeptidyl peptidase inhibitors
WO2004103276A2 (en) 2003-05-14 2004-12-02 Merck & Co., Inc. 3-amino-4-phenylbutanoic acid derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2004104216A2 (en) 2003-05-21 2004-12-02 Bayer Healthcare Ag Diagnostics and therapeutics for diseases associated with dipeptidylpeptidase iv (dpp4)
WO2004108730A1 (en) 2003-06-05 2004-12-16 Fujisawa Pharmaceutical Co., Ltd. 1h-imidazo`4,5-d!pyridazines as dpp-iv inhibitors for the treatment of niddm
US20040259903A1 (en) 2003-06-20 2004-12-23 Markus Boehringer Pyrido [2,1-a] isoquinoline derivatives
WO2004111041A1 (en) 2003-06-12 2004-12-23 Fujisawa Pharmaceutical Co., Ltd. Pyrrolidine, thiazolidine and oxazolidine compounds which inhibit dipeptidyl peptidase-iv (dpp)
WO2004111051A1 (en) 2003-06-18 2004-12-23 Boehringer Ingelheim International Gmbh Imidazo-pyridazinone derivatives and imidazo-pyridone derivatives, production thereof, and use thereof as medicaments
WO2004110436A1 (en) 2003-06-06 2004-12-23 Merck & Co., Inc. Fused indoles as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
US20040259902A1 (en) 2003-06-20 2004-12-23 Markus Boehringer Pyrido [2,1-a] isoquinoline derivatives
WO2004110375A2 (en) 2003-06-06 2004-12-23 Merck & Co., Inc. Combination therapy for the treatment of diabetes
WO2004112701A2 (en) 2003-06-17 2004-12-29 Merck & Co., Inc. Cyclohexylglycine derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2005003135A1 (en) 2003-06-24 2005-01-13 Merck & Co., Inc. Phosphoric acid salt of a dipeptidyl peptidase-iv inhibitor
JP2005023038A (en) 2003-07-04 2005-01-27 Taisho Pharmaceut Co Ltd Therapeutic agent for chronic renal disease
WO2005007647A1 (en) 2003-07-11 2005-01-27 Arena Pharmaceuticals, Inc. Trisubstituted aryl and heteroaryl derivatives as modulators of metabolism and the prophylaxis and treatment of disorders related thereto
WO2005009956A1 (en) 2003-07-21 2005-02-03 Smithkline Beecham Corporation (2s,4s)-4-fluoro-1-[4-fluoro-beta-(4-fluorophenyl)-l-phenylalanyl]-2-pyrrolidinecarbonitrile p-toluenesulfonic acid salt and anhydrous crystalline forms thereof
WO2005012312A1 (en) 2003-07-25 2005-02-10 Sanofi-Aventis Deutschland Gmbh Novel cyano thiazolides, methods for the production thereof, and use thereof as a medicament
WO2005012308A1 (en) 2003-07-25 2005-02-10 Sanofi-Aventis Deutschland Gmbh Novel cyanopyrrolidides, methods for the production thereof, and use of the same as medicaments
WO2005012249A2 (en) 2003-08-01 2005-02-10 Bristol-Myers Squibb Company Adamantyglycine- based inhibitors of dipeptidyl peptidase iv for the treatment of diabetes
WO2005011581A2 (en) 2003-07-31 2005-02-10 Merck & Co., Inc. Hexahydrodiazepinones as dipeptidyl peptidase-iv inhibitors for the treatment or prevention of diabetes
US20050043292A1 (en) 2003-08-20 2005-02-24 Pfizer Inc Fluorinated lysine derivatives as dipeptidyl peptidase IV inhibitors
WO2005020920A2 (en) 2003-09-02 2005-03-10 Merck & Co., Inc. Novel crystalline forms of a phosphoric acid salt of a dipeptidyl peptidase-iv inhibitor
US20050059716A1 (en) 2003-07-25 2005-03-17 Aventis Pharma Deutschland Gmbh Novel bicyclic cyanoheterocycles, process for their preparation and their use as medicaments
WO2005023762A1 (en) 2003-09-04 2005-03-17 Abbott Laboratories Pyrrolidine-2-carbonitrile derivatives and their use as inhibitors of dipeptidyl peptidase-iv (dpp-iv)
US20050059724A1 (en) 2003-07-25 2005-03-17 Aventis Pharma Deutschland Gmbh Novel cyanopyrrolidides, process for their preparation and their use as medicaments
WO2005025554A2 (en) 2003-09-09 2005-03-24 Japan Tobacco Inc. Dipeptidyl peptidase iv inhibitor
WO2005026148A1 (en) 2003-09-08 2005-03-24 Takeda San Diego, Inc. Dipeptidyl peptidase inhibitors
WO2005030751A2 (en) 2003-09-08 2005-04-07 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
WO2005030127A2 (en) 2003-09-23 2005-04-07 Merck & Co., Inc. Novel crystalline form of a phosphoric acid salt of a dipeptidyl peptidase-iv inhibitor
WO2005032590A1 (en) 2003-10-03 2005-04-14 Takeda Pharmaceutical Company Limited Remedy for diabetes
WO2005033099A2 (en) 2003-10-03 2005-04-14 Glenmark Pharmaceuticals Ltd. Novel dipeptidyl peptidase iv inhibitors; processes for their preparation and compositions thereof
WO2005034940A2 (en) 2003-10-15 2005-04-21 Imtm Gmbh Dual alanyl aminopeptidase and dipeptidyl peptidase iv inhibitors for functionally influencing different cells and for treating immunological, inflammatory, neuronal and other diseases
WO2005037779A2 (en) 2003-10-15 2005-04-28 Imtm Gmbh Novel dipeptidyl peptidase iv inhibitors used for functionally influencing different cells and treating immunological, inflammatory, neuronal, and other diseases
WO2005037828A1 (en) 2003-10-20 2005-04-28 Lg Life Sciences Ltd. Novel inhibitors of dpp-iv, methods of preparing the same, and pharmaceutical compositions containing the same as an active agent
WO2005040095A1 (en) 2003-10-16 2005-05-06 Astrazeneca Ab Inhibitors of dipeptidyl peptidase iv
WO2005042488A1 (en) 2003-10-31 2005-05-12 Takeda Pharmaceutical Company Limited Pyridine compounds as inhibitors of dipeptidyl peptidase iv
WO2005044195A2 (en) 2003-11-04 2005-05-19 Merck & Co., Inc. Fused phenylalanine derivatives as dipeptidyl peptidase-iv inhibitors for the treatment or prevention of diabetes
WO2005047297A1 (en) 2003-11-12 2005-05-26 Phenomix Corporation Heterocyclic boronic acid compounds
WO2005049022A2 (en) 2003-11-17 2005-06-02 Novartis Ag Use of dipeptidyl peptidase iv inhibitors
WO2005058849A1 (en) 2003-12-15 2005-06-30 Glenmark Pharmaceuticals Ltd. New dipeptidyl peptidase in inhibitors; process for their preparation and compositions containing them
WO2005063750A1 (en) 2003-12-23 2005-07-14 Boehringer Ingelheim International Gmbh Bicyclic imidazole compounds, the production thereof and their use as medicaments
WO2005072530A1 (en) 2004-01-16 2005-08-11 Merck & Co., Inc. Novel crystalline salts of a dipeptidyl peptidase-iv inhibitor
WO2005075426A1 (en) 2004-02-03 2005-08-18 Glenmark Pharmaceuticals Ltd. Novel dipeptidyl peptidase iv inhibitors; processes for their preparation and compositions thereof
WO2005079795A2 (en) 2004-02-20 2005-09-01 Novartis Ag Dpp-iv inhibitors for treating neurodegeneration and cognitive disorders
WO2005082849A1 (en) 2004-02-23 2005-09-09 Trustees Of Tufts College Lactams as conformationally constrained peptidomimetic inhibitors
WO2005082348A2 (en) 2004-02-23 2005-09-09 Trustees Of Tufts College Inhibitors of dipeptidylpeptidase iv for regulating glucose metabolism
WO2005087235A1 (en) 2004-03-09 2005-09-22 National Health Research Institutes Pyrrolidine compounds
WO2005095381A1 (en) 2004-03-15 2005-10-13 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
WO2005116014A1 (en) 2004-05-12 2005-12-08 Pfizer Products Inc. Proline derivatives and their use as dipeptidyl peptidase iv inhibitors
WO2006040625A1 (en) 2004-10-12 2006-04-20 Glenmark Pharmaceuticals S.A. Novel dipeptidyl peptidase iv inhibitors, pharmaceutical compositions containing them, and process for their preparation
WO2006068163A1 (en) 2004-12-24 2006-06-29 Dainippon Sumitomo Pharma Co., Ltd. Bicyclic pyrrole derivatives
WO2006076231A2 (en) 2005-01-10 2006-07-20 Arena Pharmaceuticals, Inc. Combination therapy for the treatment of diabetes and conditions related thereto and for the treatment of conditions ameliorated by increasing a blood glp-1 level
WO2006088129A1 (en) 2005-02-18 2006-08-24 Mitsubishi Pharma Corporation Salt of proline derivative, solvate thereof, and production method thereof
WO2006100181A2 (en) 2005-03-22 2006-09-28 F. Hoffmann-La Roche Ag New salt and polymorphs of a dpp-iv inhibitor
WO2006116157A2 (en) 2005-04-22 2006-11-02 Alantos Pharmaceuticals Holding, Inc. Dipeptidyl peptidase-iv inhibitors
WO2007019255A2 (en) 2005-08-04 2007-02-15 Novartis Ag Salts of vildagliptin
WO2007027651A2 (en) 2005-08-30 2007-03-08 Abbott Laboratories Pharmaceutical compositions as inhibitors of dipeptidyl peptidase-iv (dpp-iv)
WO2007035372A2 (en) 2005-09-16 2007-03-29 Takeda Pharmaceutical Company Limited Polymorphs of benzoate salt of 2-[[6-[(3r)-3-amino-1-piperidinyl]-3,4-dihydro-3-methyl-2,4-dioxo-1(2h)-pyrimidinyl]methyl]-benzonitrile and methods of use therefor
WO2007071576A1 (en) 2005-12-21 2007-06-28 F. Hoffmann-La Roche Ag New salt and polymorph of dpp-iv inhibitor
US20070167468A1 (en) 2004-08-06 2007-07-19 Sanofi-Aventis Deutschland Gmbh Substituted bicyclic 8-pyrr0lidinoxanthines, methods for their production, pharmaceutical formulations and their use
WO2007120689A2 (en) 2006-04-11 2007-10-25 Arena Pharmaceuticals, Inc. Methods of using gpr119 receptor to identify compounds useful for increasing bone mass in an individual
WO2007120702A2 (en) 2006-04-11 2007-10-25 Arena Pharmaceuticals, Inc. Use of gpr119 receptor agonists for increasing bone mass and for treating osteoporosis, and combination therapy relating thereto
WO2007128721A1 (en) 2006-05-04 2007-11-15 Boehringer Ingelheim Internationalgmbh Polymorphs
WO2007148185A2 (en) 2006-06-21 2007-12-27 Pfizer Products Inc. Substituted 3 -amino- pyrrolidino-4 -lactams as dpp inhibitors
WO2008027273A2 (en) 2006-08-30 2008-03-06 Phenomix Corporation Solid citrate and tartrate salts of dpp-iv inhibitors
EP1902730A1 (en) 2005-06-09 2008-03-26 Banyu Pharmaceutical Co., Ltd. Npy y2 agonist for use as therapeutic agent for disease accompanied by diarrhea
WO2008070692A2 (en) * 2006-12-06 2008-06-12 Smithkline Beecham Corporation Bicyclic compounds and use as antidiabetics
US20080146818A1 (en) 2004-02-05 2008-06-19 Yasumichi Fukuda Bicycloester Derivative
WO2008114857A1 (en) 2007-03-22 2008-09-25 Kyorin Pharmaceutical Co., Ltd. Method for producing aminoacetylpyrrolidinecarbonitrile derivative
WO2009084497A1 (en) 2007-12-28 2009-07-09 Dainippon Sumitomo Pharma Co., Ltd. Methyl-substituted piperidine derivative
WO2009126245A1 (en) 2008-04-07 2009-10-15 Arena Pharmaceuticals, Inc. Methods of using a g protein-coupled receptor to identify peptide yy (pyy) secretagogues and compounds useful in the treatment of conditions modulated by pyy

Patent Citations (401)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DD296075A5 (en) 1989-08-07 1991-11-21 Martin-Luther-Universitaet Halle-Wittenberg,De PROCESS FOR THE PREPARATION OF NEW INHIBITORS OF DIPEPTIDYL PEPTIDASE IV
WO1991016339A1 (en) 1990-04-14 1991-10-31 New England Medical Center Hospitals, Inc. Inhibitors of dipeptidyl-aminopeptidase type iv
EP0528858A1 (en) 1990-04-14 1993-03-03 New England Medical Center Inc Inhibitors of dipeptidyl-aminopeptidase type iv.
JPH05508624A (en) 1990-04-14 1993-12-02 ニュー イングランド メデカル センター ホスピタルズ インク Dipeptidyl-aminopeptidase type 4 inhibitor
WO1993008259A2 (en) 1991-10-22 1993-04-29 New England Medical Center Hospitals, Inc. Inhibitors of dipeptidyl-aminopeptidase type iv
EP0610317A1 (en) 1991-10-22 1994-08-17 New England Medical Center Inhibitors of dipeptidyl-aminopeptidase type iv
EP1050540A2 (en) 1991-10-22 2000-11-08 New England Medical Center Hospitals, Inc. Inhibitors of dipeptidyl-aminopeptidase type IV
WO1993010127A1 (en) 1991-11-22 1993-05-27 Boehringer Ingelheim Pharmaceuticals, Inc. Method for making a prolineboronate ester
CA2123128A1 (en) 1991-11-22 1993-05-27 Roger Snow Method for making a prolineboronate ester
JPH07501078A (en) 1991-11-22 1995-02-02 ベーリンガー インゲルハイム ファーマシューティカルズ インコーポレイテッド Production method of proline boronate ester
EP0641347A1 (en) 1991-11-22 1995-03-08 Boehringer Ingelheim Pharmaceuticals Inc. Method for making a prolineboronate ester
US5571815A (en) * 1992-03-14 1996-11-05 Hoechst Aktiengesellschaft Substituted pyrimidines, process for their preparation, and their use as pesticides and fungicides
EP0731789A1 (en) 1993-12-03 1996-09-18 Ferring B.V. Dp-iv-serine protease inhibitors
WO1995015309A1 (en) 1993-12-03 1995-06-08 Ferring B.V. Dp-iv-serine protease inhibitors
WO1995029691A1 (en) 1994-04-28 1995-11-09 Georgia Tech Research Corporation Proline phosphonate derivatives
DE19616486A1 (en) 1996-04-25 1997-10-30 Knoell Hans Forschung Ev Method of lowering blood glucose levels in mammals
WO1997040832A1 (en) 1996-04-25 1997-11-06 Probiodrug Gesellschaft für Arzneimittelforschung mbH Use of dipeptidyl peptidase iv effectors for lowering the blood glucose level in mammals
JP2001510442A (en) 1996-04-25 2001-07-31 プロビオドラッグ ゲゼルシャフト フュル アルツナイミッテルフォルシュング エムベーハー Use of dipeptidyl peptidase (IV) effector for lowering blood glucose in mammals
US6303661B1 (en) 1996-04-25 2001-10-16 Probiodrug Use of dipeptidyl peptidase IV effectors for lowering the blood glucose level in mammals
JPH1081666A (en) 1996-06-12 1998-03-31 Ishihara Sangyo Kaisha Ltd Phthalimide derivative or salt thereof, its production and medicinal composition containing the same
WO1998018763A1 (en) 1996-10-25 1998-05-07 Tanabe Seiyaku Co., Ltd. Tetrahydroisoquinoline derivatives
JPH10182613A (en) 1996-10-25 1998-07-07 Tanabe Seiyaku Co Ltd Tetraphydroisoquinoline derivative
WO1998019998A2 (en) 1996-11-07 1998-05-14 Novartis Ag N-substituted 2-cyanopyrrolidines
JP2000511559A (en) 1996-11-07 2000-09-05 ノバルティス アクチエンゲゼルシャフト N-substituted 2-cyanopyrrolidine
WO1998050046A1 (en) 1997-05-07 1998-11-12 Trustees Of Tufts College Use of cd26 inhibitor for the manufacture of a medicament for the treatment of hiv
EP0980249A1 (en) 1997-05-07 2000-02-23 Trustees Of Tufts College Use of cd26 inhibitor for the manufacture of a medicament for the treatment of hiv
WO1998050066A1 (en) 1997-05-07 1998-11-12 Trustees Of Tufts College Potentiation of the immune response through delivery of compounds binding a cytoplasmic dipeptidase
CA2289125A1 (en) 1997-05-07 1998-11-12 Trustees Of Tufts College Potentiation of the immune response through delivery of compounds binding a cytoplasmic dipeptidase
US6100234A (en) 1997-05-07 2000-08-08 Tufts University Treatment of HIV
CA2289124A1 (en) 1997-05-07 1998-11-12 Trustees Of Tufts College Use of cd26 inhibitor for the manufacture of a medicament for the treatment of hiv
US6040145A (en) 1997-05-07 2000-03-21 Tufts University Potentiation of the immune response
EP0975359A1 (en) 1997-05-07 2000-02-02 Trustees Of Tufts College Potentiation of the immune response through delivery of compounds binding a cytoplasmic dipeptidase
WO1999016864A1 (en) 1997-09-29 1999-04-08 Point Therapeutics, Inc. Stimulation of hematopoietic cells in vitro
EP1043328A1 (en) 1997-11-18 2000-10-11 Zaidan Hojin Biseibutsu Kagaku Kenkyu Kai Novel physiologically active substance sulphostin, process for producing the same, and use thereof
WO1999025719A1 (en) 1997-11-18 1999-05-27 Zaidan Hojin Biseibutsu Kagaku Kenkyu Kai Novel physiologically active substance sulphostin, process for producing the same, and use thereof
US6803357B1 (en) 1998-02-02 2004-10-12 New England Medical Center Hospitals, Inc. Method of regulating glucose metabolism, and reagents related thereto
WO1999056753A1 (en) 1998-05-04 1999-11-11 Point Therapeutics, Inc. Hematopoietic stimulation
EP1215207A2 (en) 1998-05-28 2002-06-19 Probiodrug AG Salts of isoleucyl-thiazolidine and -pyrrolidine and their use as dipeptidylpeptidase inhibitors
US20030134802A1 (en) 1998-05-28 2003-07-17 Hans-Ulrich Demuth Novel effectors of dipepetidyl peptidase IV
EP1304327A2 (en) 1998-05-28 2003-04-23 Probiodrug AG Glutamine-thiazolidide and -pyrrolidide and their use as dipeptidylpeptidase IV inhibitors
JP2002516318A (en) 1998-05-28 2002-06-04 プロバイオドラッグ ゲゼルシャフト フュア アルツナイミッテルフォルシュンク エムベーハー Novel effector of dipeptidyl peptidase IV
EP1082314A1 (en) 1998-05-28 2001-03-14 Probiodrug Gesellschaft für Arzneimittelforschung mbH New dipeptidyl peptidase iv effectors
WO1999061431A1 (en) 1998-05-28 1999-12-02 Probiodrug Gesellschaft für Arzneimittelforschung mbH New dipeptidyl peptidase iv effectors
DE19823831A1 (en) 1998-05-28 1999-12-02 Probiodrug Ges Fuer Arzneim New pharmaceutical use of isoleucyl thiazolidide and its salts
JP2002517401A (en) 1998-06-05 2002-06-18 ポイント セラピューティクス, インコーポレイテッド Cyclic boroproline compound
WO1999062914A1 (en) 1998-06-05 1999-12-09 Point Therapeutics, Inc. Cyclic boroproline compounds
JP2003524591A (en) 1998-06-24 2003-08-19 プロバイオドラッグ ゲゼルシャフト フュア アルツナイミッテルフォルシュンク エムベーハー Prodrugs of DPIV inhibitors
WO1999067278A1 (en) 1998-06-24 1999-12-29 Probiodrug Gesellschaft für Arzneimittelforschung mbH Prodrugs of dipeptidyl peptidase iv inhibitors
DE19828113A1 (en) 1998-06-24 2000-01-05 Probiodrug Ges Fuer Arzneim Prodrugs of Dipeptidyl Peptidase IV Inhibitors
US20020049164A1 (en) 1998-06-24 2002-04-25 Hans-Ulrich Demuth Prodrugs of DP IV-inhibitors
DE19834591A1 (en) 1998-07-31 2000-02-03 Probiodrug Ges Fuer Arzneim Use of substances that decrease the activity of dipeptidyl peptidase IV to increase blood sugar levels, e.g. for treating hypoglycemia
EP0995440A1 (en) 1998-07-31 2000-04-26 Probiodrug Gesellschaft für Arzneimittelforschung mbH Method to increase the blood, glucose level in mammals
WO2000010549A1 (en) 1998-08-21 2000-03-02 Point Therapeutics, Inc. Regulation of substrate activity
CA2339537A1 (en) 1998-08-21 2000-03-02 Barbara Wallner Regulation of substrate activity
EP1104293A1 (en) 1998-08-21 2001-06-06 Point Therapeutics, Inc. Regulation of substrate activity
US20020006899A1 (en) 1998-10-06 2002-01-17 Pospisilik Andrew J. Use of dipeptidyl peptidase IV effectors for lowering blood pressure in mammals
US6410508B1 (en) 1998-10-07 2002-06-25 Med College Georgia Res Inst Glucose-dependent insulinotropic peptide for use as an osteotropic hormone
US6242422B1 (en) 1998-10-22 2001-06-05 Idun Pharmacueticals, Inc. (Substituted)Acyl dipeptidyl inhibitors of the ice/ced-3 family of cysteine proteases
EP1123272A1 (en) 1998-10-22 2001-08-16 Idun Pharmaceuticals, Inc. (substituted)acyl dipeptidyl inhibitors of the ice/ced-3 family of cysteine proteases
WO2000023421A1 (en) 1998-10-22 2000-04-27 Idun Pharmaceuticals, Inc. (SUBSTITUTED)ACYL DIPEPTIDYL INHIBITORS OF THE ICE/ced-3 FAMILY OF CYSTEINE PROTEASES
JP2002527504A (en) 1998-10-22 2002-08-27 アイドゥン ファーマシューティカルズ, インコーポレイテッド ICE / ced-3 substituted acyldipeptidyl inhibitors of cysteine proteases
WO2000031258A2 (en) 1998-11-20 2000-06-02 Arena Pharmaceuticals, Inc. Human orphan g protein-coupled receptors
JP2002531547A (en) 1998-12-10 2002-09-24 ノバルティス アクチエンゲゼルシャフト N-substituted 2-cyanopyrrolidine
WO2000034241A1 (en) 1998-12-10 2000-06-15 Novartis Ag N-substituted 2-cyanopyrrolidines
EP1137635A1 (en) 1998-12-10 2001-10-04 Novartis AG N-substituted 2-cyanopyrrolidines
US6166063A (en) 1998-12-10 2000-12-26 Novartis Ag N-(substituted glycyl)-2-cyanopyrrolidines, pharmaceutical compositions containing them and their use in inhibiting dipeptidyl peptidase-IV
JP2000191616A (en) 1998-12-24 2000-07-11 Senju Pharmaceut Co Ltd New peptidylaldehyde derivative and medicine containing the same
WO2000056296A2 (en) 1999-03-23 2000-09-28 Ferring Bv Compositions for improving fertility
WO2000056297A2 (en) 1999-03-23 2000-09-28 Ferring B.V. Pharmaceutical compositions comprising dipeptidy peptidase iv inhibitors for the promotion of growth
JP2000327689A (en) 1999-05-17 2000-11-28 Microbial Chem Res Found Sulfostin analogue, and production of sulfostin and its analogue
WO2000069868A1 (en) 1999-05-17 2000-11-23 Zaidan Hojin Biseibutsu Kagaku Kenkyu Kai Sulphostin analogues and processes for the preparation of sulphostin and analogues thereof
WO2000071135A1 (en) 1999-05-25 2000-11-30 Point Therapeutics, Inc. Anti-tumor comprising boroproline compounds
US6617340B1 (en) 1999-07-29 2003-09-09 Novartis Ag N-(substituted glycyl)-pyrrolidines, pharmaceutical compositions containing them and their use in inhibiting dipeptidyl peptidase-IV
WO2001034594A1 (en) 1999-11-12 2001-05-17 Guilford Pharmaceuticals, Inc. Dipeptidyl peptidase iv inhibitors and methods of making and using dipeptidyl peptidase iv inhibitors
EP1228061A1 (en) 1999-11-12 2002-08-07 Guilford Pharmaceuticals Inc. Dipeptidyl peptidase iv inhibitors and methods of making and using dipeptidyl peptidase iv inhibitors
JP2003535034A (en) 1999-11-12 2003-11-25 ギルフォード ファーマシューティカルズ インコーポレイテッド Dipeptidyl peptidase IV inhibitors and methods for producing and using dipeptidyl peptidase IV inhibitors
US6380398B2 (en) 2000-01-04 2002-04-30 Novo Nordisk A/S Therapeutically active and selective heterocyclic compounds that are inhibitors of the enzyme DPP-IV
EP1248604A2 (en) 2000-01-21 2002-10-16 Novartis AG Combinations comprising dipeptidylpeptidase-iv inhibitors and antidiabetic agents
WO2001052825A2 (en) 2000-01-21 2001-07-26 Novartis Ag Combinations comprising dipeptidylpeptidase-iv inhibitors and antidiabetic agents
WO2001055105A1 (en) 2000-01-24 2001-08-02 Novo Nordisk A/S N-substituted 2-cyanopyroles and -pyrrolines which are inhibitors of the enzyme dpp-iv
EP1254113A1 (en) 2000-01-24 2002-11-06 Novo Nordisk A/S N-substituted 2-cyanopyroles and -pyrrolines which are inhibitors of the enzyme dpp-iv
JP2003520849A (en) 2000-01-24 2003-07-08 ノボ ノルディスク アクティーゼルスカブ N-substituted 2-cyanopyrroles and -pyrrolines which are inhibitors of the enzyme DPP-IV
US20020103384A1 (en) 2000-01-24 2002-08-01 Anders Kanstrup Therapeutically active and selective heterocyclic compounds that are inhibitors of the enzyme DPP-IV
US6645995B2 (en) 2000-01-24 2003-11-11 Novo Nordisk A/S Therapeutically active and selective heterocyclic compounds that are inhibitors of the enzyme DPP-IV
US6395767B2 (en) 2000-03-10 2002-05-28 Bristol-Myers Squibb Company Cyclopropyl-fused pyrrolidine-based inhibitors of dipeptidyl peptidase IV and method
EP1261586A2 (en) 2000-03-10 2002-12-04 Bristol-Myers Squibb Company Cyclopropyl-fused pyrrolidine-based inhibitors of dipeptidyl peptidase iv, processes for their preparation, and their use
WO2001068603A2 (en) 2000-03-10 2001-09-20 Bristol-Myers Squibb Co. Cyclopropyl-fused pyrrolidine-based inhibitors of dipeptidyl iv, processes for their preparation, and their use
JP2003531118A (en) 2000-03-10 2003-10-21 ブリストル−マイヤーズ スクイブ カンパニー Inhibitors and methods for dipeptidyl peptidase IV having a cyclopropyl-fused pyrrolidine skeleton
WO2001081304A1 (en) 2000-04-26 2001-11-01 Ferring Bv Inhibitors of dipeptidyl peptidase iv
EP1282600A1 (en) 2000-04-26 2003-02-12 Ferring BV Inhibitors of dipeptidyl peptidase iv
JP2003531191A (en) 2000-04-26 2003-10-21 フェリング ベスローテン フェンノートシャップ Inhibitors of dipeptidyl peptidase IV
JP2003531204A (en) 2000-04-26 2003-10-21 フェリング ベスローテン フェンノートシャップ Inhibitors of dipeptidyl peptidase IV
WO2001081337A1 (en) 2000-04-26 2001-11-01 Ferring B.V. Inhibitors of dipeptidyl peptidase iv
US20030216450A1 (en) 2000-04-26 2003-11-20 Evans David Michael Inhibitors of dipeptidyl peptidase IV
EP1280797A1 (en) 2000-04-26 2003-02-05 Ferring B.V. Inhibitors of dipeptidyl peptidase iv
WO2001096295A2 (en) 2000-06-13 2001-12-20 Novartis Ag 2-cyanopyrrolidine derivatives and their use as medicaments
JP2004503531A (en) 2000-06-13 2004-02-05 ノバルティス アクチエンゲゼルシャフト 2-Cyanopyrrolidine derivatives and their use as drugs
US6432969B1 (en) 2000-06-13 2002-08-13 Novartis Ag N-(substituted glycyl)-2 cyanopyrrolidines, pharmaceutical compositions containing them and their use in inhibiting dipeptidyl peptidase-IV
EP1296974A2 (en) 2000-06-13 2003-04-02 Novartis AG 2-cyanopyrrolidine derivatives and their use as medicaments
JP2003535898A (en) 2000-06-19 2003-12-02 スミスクライン ビーチャム パブリック リミテッド カンパニー Combination of dipeptidyl peptidase IV inhibitors and other antidiabetic agents for the treatment of diabetes mellitus
WO2001097808A1 (en) 2000-06-19 2001-12-27 Smithkline Beecham Plc Combinations of depeptidyl peptidase iv inhibitors and other antidiabetic agents for the treatment of diabete mellitus
EP1301187A2 (en) 2000-07-04 2003-04-16 Novo Nordisk A/S Heterocyclic compounds, which are inhibitors of the enzyme dpp-iv
US20040034014A1 (en) 2000-07-04 2004-02-19 Kanstrup Anders Bendtz Heterocyclic compounds, which are inhibitors of the enzyme DPP-IV
WO2002002560A2 (en) 2000-07-04 2002-01-10 Novo Nordisk A/S Purine-2,6-diones which are inhibitors of the enzyme dipeptidyl peptidase iv (dpp-iv)
WO2002014271A1 (en) 2000-08-10 2002-02-21 Mitsubishi Pharma Corporation Proline derivatives and use thereof as drugs
JP2002356472A (en) 2000-10-06 2002-12-13 Tanabe Seiyaku Co Ltd Nitrogen-containing five-membered ring compound
WO2002030891A1 (en) 2000-10-06 2002-04-18 Tanabe Seiyaku Co., Ltd. Aliphatic nitrogenous five-membered ring compounds
WO2002030890A1 (en) 2000-10-06 2002-04-18 Tanabe Seiyaku Co., Ltd. Nitrogenous five-membered ring compounds
US20040229926A1 (en) 2000-10-06 2004-11-18 Tanabe Seiyaku Co., Ltd. Aliphatic nitrogen - containing 5 - membered ring compound
JP2002356471A (en) 2000-10-06 2002-12-13 Tanabe Seiyaku Co Ltd Aliphatic nitrogen-containing five-membered ring compound
US20040063935A1 (en) 2000-10-06 2004-04-01 Kosuke Yasuda Aliphatic nitrogenous five-membered ring compounds
JP2004035574A (en) 2000-10-06 2004-02-05 Tanabe Seiyaku Co Ltd Aliphatic nitrogenous five-membered ring compound
US6849622B2 (en) 2000-10-06 2005-02-01 Tanabe Seiyaku Co., Ltd. Aliphatic nitrogenous five-membered ring compounds
WO2002034900A1 (en) 2000-10-27 2002-05-02 The University Of Sydney Dipeptidyl peptidases
WO2002038541A1 (en) 2000-11-10 2002-05-16 Taisho Pharmaceutical Co., Ltd. Cyanopyrrolidine derivatives
US20070112059A1 (en) 2000-11-10 2007-05-17 Taisho Pharmaceutical Co., Ltd. Cyanopyrrolidine derivatives
EP1333025A1 (en) 2000-11-10 2003-08-06 Taisho Pharmaceutical Co., Ltd Cyanopyrrolidine derivatives
US20040072892A1 (en) 2000-11-10 2004-04-15 Hiroshi Fukushima Cyanopyrrolidine derivatives
EP1338651A1 (en) 2000-12-01 2003-08-27 Yamanouchi Pharmaceutical Co. Ltd. Method of screening remedy for diabetes
US20040180925A1 (en) 2000-12-27 2004-09-16 Kenji Matsuno Dipeptidylpeptidase-IV inhibitor
WO2002051836A1 (en) 2000-12-27 2002-07-04 Kyowa Hakko Kogyo Co., Ltd. Dipeptidyl peptidase iv inhibitor
EP1354882A1 (en) 2000-12-27 2003-10-22 Kyowa Hakko Kogyo Co., Ltd. Dipeptidyl peptidase iv inhibitor
CA2433090A1 (en) 2000-12-27 2002-07-04 Kyowa Hakko Kogyo Co., Ltd. Dipeptidyl peptidase iv inhibitor
WO2002055088A1 (en) 2001-01-16 2002-07-18 Nippon Kayaku Kabushiki Kaisha Remedies for bone marrow suppression and infectious diseases and white blood cell count elevating agents
JP2003238566A (en) 2001-02-02 2003-08-27 Takeda Chem Ind Ltd Condensed heterocyclic compound
EP1355886A1 (en) 2001-02-02 2003-10-29 Takeda Chemical Industries, Ltd. Fused heterocyclic compounds
WO2002062764A1 (en) 2001-02-02 2002-08-15 Takeda Chemical Industries, Ltd. Fused heterocyclic compounds
US20040077645A1 (en) 2001-02-24 2004-04-22 Frank Himmelsbach Xanthine derivatives,production and use thereof as medicament
WO2002068420A1 (en) 2001-02-24 2002-09-06 Boehringer Ingelheim Pharma Gmbh & Co. Kg Xanthine derivative, production and use thereof as a medicament
US20020198205A1 (en) 2001-02-24 2002-12-26 Frank Himmelsbach Xanthine derivatives, the preparation thereof and their use as pharmaceutical compositions
JP2004522786A (en) 2001-02-24 2004-07-29 ベーリンガー インゲルハイム ファルマ ゲゼルシャフト ミット ベシュレンクテル ハフツング ウント コンパニー コマンディトゲゼルシャフト Xanthine derivatives, their preparation and their use as pharmaceutical compositions
US20040087587A1 (en) 2001-02-24 2004-05-06 Boehringer Ingelheim Pharma Gmbh & Co. Kg Xanthine derivatives, the preparation thereof and their use as pharmaceutical compositions
JP2002265439A (en) 2001-03-08 2002-09-18 Mitsubishi Pharma Corp Cyanopyrrolidine derivative and its use for medicine
EP1385508A1 (en) 2001-03-27 2004-02-04 Merck & Co., Inc. Dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
US20040106656A1 (en) 2001-03-27 2004-06-03 Ashton Wallace T Dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
JP2004525929A (en) 2001-03-27 2004-08-26 メルク エンド カムパニー インコーポレーテッド Dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2002076450A1 (en) 2001-03-27 2002-10-03 Merck & Co., Inc. Dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
US20030087950A1 (en) 2001-03-28 2003-05-08 Denanteuil Guillaume New alpha-amino acid sulphonyl compounds
FR2822826A1 (en) 2001-03-28 2002-10-04 Servier Lab NOVEL ALPHA-AMINO ACID SULPHONYL DERIVATIVES, PROCESS FOR PREPARING THEM AND PHARMACEUTICAL COMPOSITIONS CONTAINING SAME
US6716843B2 (en) 2001-03-28 2004-04-06 Les Laboratoires Servier Alpha-amino acid sulphonyl compounds
EP1245568A1 (en) 2001-03-28 2002-10-02 Les Laboratoires Servier Sulfonyl derivatives of amino acids and their use as inhibitors of dipeptidyl-peptidase IV (DPP IV)
WO2002083109A1 (en) 2001-04-11 2002-10-24 Ferring Bv Treatment of type 2 diabetes with inhibitors of dipeptidyl peptidase iv
JP2004525179A (en) 2001-04-11 2004-08-19 フェリング ベスローテン フェンノートシャップ Treatment of type II diabetes with dipeptidyl peptidase IV inhibitors
US6573287B2 (en) 2001-04-12 2003-06-03 Bristo-Myers Squibb Company 2,1-oxazoline and 1,2-pyrazoline-based inhibitors of dipeptidyl peptidase IV and method
WO2002083128A1 (en) 2001-04-12 2002-10-24 Bristol-Myers Squibb Company 2,1-oxazoline and 1,2-pyrazoline-based inhibitors of dipeptidyl peptidase iv and method
US20020183367A1 (en) 2001-04-12 2002-12-05 Sulsky Richard B. 2,1-Oxazoline and 1,2-pyrazoline-based inhibitors of dipeptidyl peptidase IV and method
EP1377288A1 (en) 2001-04-12 2004-01-07 Bristol-Myers Squibb Company 2,1-oxazoline and 1,2-pyrazoline-based inhibitors of dipeptidyl peptidase iv and method
JP2004532220A (en) 2001-04-12 2004-10-21 ブリストル−マイヤーズ スクイブ カンパニー Dipeptidyl peptidase IV 2,1-oxazoline and 1,2-pyrazolin based inhibitors and methods
EP1258476A1 (en) 2001-05-15 2002-11-20 Les Laboratoires Servier Alpha-amino acid derivatives, method for their preparation and their use as dipeptidyl-peptidase IV inhibitors (DPP IV)
JP2002363157A (en) 2001-05-15 2002-12-18 Lab Servier NEW alpha-AMINO ACID COMPOUND, METHOD FOR PREPARING THE SAME AND PHARMACEUTICAL COMPOSITION CONTAINING THE SAME
FR2824825A1 (en) 2001-05-15 2002-11-22 Servier Lab NOVEL ALPHA-AMINO-ACID DERIVATIVES, THEIR PREPARATION PROCESS AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
US6706742B2 (en) 2001-05-15 2004-03-16 Les Laboratories Servier Alpha-amino-acid compounds
US20030078247A1 (en) 2001-05-15 2003-04-24 De Nanteuil Guillaume Alpha-amino-acid compounds
EP1406872A2 (en) 2001-06-20 2004-04-14 Merck & Co., Inc. Dipeptidyl peptidase inhibitors for the treatment of diabetes
EP1406622A2 (en) 2001-06-20 2004-04-14 Merck & Co., Inc. Dipeptidyl peptidase inhibitors for the treatment of diabetes
WO2003000180A2 (en) 2001-06-20 2003-01-03 Merck & Co., Inc. Dipeptidyl peptidase inhibitors for the treatment of diabetes
JP2005500308A (en) 2001-06-20 2005-01-06 メルク エンド カムパニー インコーポレーテッド Dipeptidyl peptidase inhibitors for the treatment of diabetes
WO2003000181A2 (en) 2001-06-20 2003-01-03 Merck & Co., Inc. Dipeptidyl peptidase inhibitors for the treatment of diabetes
JP2004535433A (en) 2001-06-20 2004-11-25 メルク エンド カムパニー インコーポレーテッド Dipeptidyl peptidase inhibitors for the treatment of diabetes
EP1399154A1 (en) 2001-06-25 2004-03-24 Ferring BV 3-fluoro-pyrrolidines as antidiabetic agents
WO2003000250A1 (en) 2001-06-25 2003-01-03 Ferring Bv 3-fluoro-pyrrolidines as antidiabetic agents
JP2004534815A (en) 2001-06-25 2004-11-18 フェリング ベスローテン フェンノートシャップ 3-Fluoro-pyrrolidine as antidiabetic drug
JP2005500321A (en) 2001-06-27 2005-01-06 スミスクライン ビーチャム コーポレーション Fluoropyrrolidines as dipeptidyl peptidase inhibitors
WO2003002596A2 (en) 2001-06-27 2003-01-09 Probiodrug Ag Use of dipeptidyl peptidase iv inhibitors as therapeutics for neurological disorders
JP2004521149A (en) 2001-06-27 2004-07-15 プロバイオドラッグ アーゲー Novel dipeptidyl peptidase IV inhibitors and their use as anticancer agents
WO2003002595A2 (en) 2001-06-27 2003-01-09 Probiodrug Ag Dipeptidyl peptidase iv inhibitors and their uses as anti-cancer agents
WO2003072556A1 (en) 2001-06-27 2003-09-04 Prosidion Ltd. Glutaminyl based dpiv inhibitors
JP2004535445A (en) 2001-06-27 2004-11-25 スミスクライン ビーチャム コーポレーション Fluoropyrrolidines as dipeptidyl peptidase inhibitors
US20030119750A1 (en) 2001-06-27 2003-06-26 Hans-Ulrich Demuth Use of dipeptidyl peptidase IV inhibitors
EP1399470A2 (en) 2001-06-27 2004-03-24 Probiodrug AG Dipeptidyl peptidase iv inhibitors and their uses as anti-cancer agents
EP1406873A2 (en) 2001-06-27 2004-04-14 Smithkline Beecham Corporation Fluoropyrrolidines as dipeptidyl peptidase inhibitors
WO2003002593A2 (en) 2001-06-27 2003-01-09 Probiodrug Ag Peptide structures useful for competitive modulation of dipeptidyl peptidase iv catalysis
EP1399433A2 (en) 2001-06-27 2004-03-24 Smithkline Beecham Corporation Fluoropyrrolidines as dipeptidyl peptidase inhibitors
EP1399420A2 (en) 2001-06-27 2004-03-24 SmithKline Beecham Corporation Pyrrolidines as dipeptidyl peptidase inhibitors
WO2003002553A2 (en) 2001-06-27 2003-01-09 Smithkline Beecham Corporation Fluoropyrrolidines as dipeptidyl peptidase inhibitors
US20030130199A1 (en) 2001-06-27 2003-07-10 Von Hoersten Stephan Dipeptidyl peptidase IV inhibitors and their uses as anti-cancer agents
JP2004534836A (en) 2001-06-27 2004-11-18 プロバイオドラッグ アーゲー Novel use of dipeptidyl peptidase IV inhibitors
EP1399469A2 (en) 2001-06-27 2004-03-24 Probiodrug AG Peptide structures useful for competitive modulation of dipeptidyl peptidase iv catalysis
EP1399471A2 (en) 2001-06-27 2004-03-24 Probiodrug AG Use of dipeptidyl peptidase iv inhibitors as therapeutics for neurological disorders
WO2003002530A2 (en) 2001-06-27 2003-01-09 Smithkline Beecham Corporation Pyrrolidines as dipeptidyl peptidase inhibitors
JP2004530729A (en) 2001-06-27 2004-10-07 プロバイオドラッグ アーゲー Peptide structures useful for antagonizing dipeptidyl peptidase IV catalysis
WO2003002531A2 (en) 2001-06-27 2003-01-09 Smithkline Beecham Corporation Fluoropyrrolidines as dipeptidyl peptidase inhibitors
EP1404675A1 (en) 2001-07-03 2004-04-07 Novo Nordisk A/S Dpp-iv-inhibiting purine derivatives for the treatment of diabetes
JP2005502624A (en) 2001-07-03 2005-01-27 ノボ ノルディスク アクティーゼルスカブ Purine derivatives inhibiting DPP-IV for the treatment of diabetes
US20030105077A1 (en) 2001-07-03 2003-06-05 Kanstrup Anders Bendtz Heterocyclic compounds that are inhibitors of the enzyme DPP-IV
US6869947B2 (en) 2001-07-03 2005-03-22 Novo Nordisk A/S Heterocyclic compounds that are inhibitors of the enzyme DPP-IV
WO2003004496A1 (en) 2001-07-03 2003-01-16 Novo Nordisk A/S Dpp-iv-inhibiting purine derivatives for the treatment of diabetes
US20030100563A1 (en) 2001-07-06 2003-05-29 Edmondson Scott D. Beta-amino heterocyclic dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2003004498A1 (en) 2001-07-06 2003-01-16 Merck & Co., Inc. Beta-amino tetrahydroimidazo (1, 2-a) pyrazines and tetrahydrotrioazolo (4, 3-a) pyrazines as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
EP1412357A1 (en) 2001-07-06 2004-04-28 Merck & Co., Inc. Beta-amino tetrahydroimidazo(1,2-a)pyrazines and tetrahydrotriazolo(4,3-a)pyrazines as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
US6699871B2 (en) 2001-07-06 2004-03-02 Merck & Co., Inc. Beta-amino heterocyclic dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
JP2004536115A (en) 2001-07-06 2004-12-02 メルク エンド カムパニー インコーポレーテッド Β-aminotetrahydroimidazo (1,2-A) pyrazines and tetrahydrotriazolo (4,3-A) pyrazines as dipeptidyl peptidase inhibitors for treating or preventing diabetes
WO2003015775A1 (en) 2001-08-17 2003-02-27 Probiodrug Ag New dipeptidyl peptidase iv inhibitors and their uses for lowering blood pressure levels
JP2005505531A (en) 2001-08-17 2005-02-24 プロビオドルグ アーゲー Novel dipeptidyl peptidase IV inhibitors and their use to reduce blood pressure levels
WO2003022871A2 (en) 2001-09-06 2003-03-20 Probiodrug Ag Peptides having a c- terminal hydroxylamino group as inhibitors of dipeptidyl peptidase i
US20030119738A1 (en) 2001-09-06 2003-06-26 Andre Niestroj Novel inhibitors of dipeptidyl peptidase I
DE10143840A1 (en) 2001-09-06 2003-03-27 Probiodrug Ag New acylated hydroxamates useful for the treatment of e.g. wound healing
US20040259883A1 (en) 2001-09-14 2004-12-23 Hiroshi Sakashita Thiazolidine derivative and medicinal use thereof
WO2003024942A1 (en) 2001-09-14 2003-03-27 Mitsubishi Pharma Corporation Thiazolidine derivative and medicinal use thereof
EP1426366A1 (en) 2001-09-14 2004-06-09 Mitsubishi Pharma Corporation Thiazolidine derivative and medicinal use thereof
WO2003024965A2 (en) 2001-09-19 2003-03-27 Novo Nordisk A/S Heterocyclic compounds that are inhibitors of the enzyme dpp-iv
US20030199528A1 (en) 2001-09-19 2003-10-23 Kanstrup Anders B. Hetrocyclic compounds that are inhibitors of the enzyme DPP-IV
EP1463727A2 (en) 2001-09-19 2004-10-06 Novo Nordisk A/S Heterocyclic compounds that are inhibitors of the enzyme dpp-iv
WO2003035067A1 (en) 2001-10-23 2003-05-01 Ferring B.V. Novel dipeptidyl peptidase iv (dp-iv) inhibitors as anti-diabetic agents
WO2003035057A1 (en) 2001-10-23 2003-05-01 Ferring B.V. Inhibitors of dipeptidyl peptidase iv
US20050004205A1 (en) 2001-10-23 2005-01-06 Evans David M Novel dipeptidyl peptidase iv (dp-iv) inhibitors as anti-diabetic agents
EP1446116A1 (en) 2001-10-23 2004-08-18 Ferring B.V. Inhibitors of dipeptidyl peptidase iv
EP1450794A1 (en) 2001-10-23 2004-09-01 Ferring B.V. Novel dipeptidyl peptidase iv (dp-iv) inhibitors as anti-diabetic agents
WO2003037327A1 (en) 2001-10-26 2003-05-08 F. Hoffmann-La-Roche Ag N-substituted pyrrolidin derivatives as dipeptidyl peptidase iv inhibitors
US20030130281A1 (en) 2001-10-26 2003-07-10 Markus Boehringer DPP IV inhibitors
EP1441719A1 (en) 2001-10-26 2004-08-04 F. Hoffmann-La Roche Ag N-substituted pyrrolidin derivatives as dipeptidyl peptidase iv inhibitors
US6861440B2 (en) 2001-10-26 2005-03-01 Hoffmann-La Roche Inc. DPP IV inhibitors
WO2003038123A2 (en) 2001-10-31 2003-05-08 Novartis Ag Methods to treat diabetes and related conditions based on polymorphisms in the tcf1 gene
JP2005507261A (en) 2001-10-31 2005-03-17 ノバルティス アクチエンゲゼルシャフト Methods for treating diabetes and related conditions based on polymorphisms in the TCF1 gene
US20030125304A1 (en) 2001-11-09 2003-07-03 Hans-Ulrich Demuth Substituted amino ketone compounds
WO2003040174A2 (en) 2001-11-09 2003-05-15 Probiodrug Ag Substituted amino ketone compounds
EP1442049A2 (en) 2001-11-09 2004-08-04 Probiodrug AG Substituted amino ketone compounds
EP1469873A2 (en) 2001-11-26 2004-10-27 Trustees of Tufts College Peptidomimetic inhibitors of post-proline cleaving enzymes
CA2466870A1 (en) 2001-11-26 2003-06-05 Trustees Of Tufts College Methods for treating autoimmune disorders, and reagents related thereto
WO2003045228A2 (en) 2001-11-26 2003-06-05 Trustees Of Tufts College Methods for treating autoimmune disorders, and reagents related thereto
EP1465891A2 (en) 2001-12-26 2004-10-13 Guilford Pharmaceuticals Inc. Inhibitors of dipeptidyl peptidase iv
WO2003057144A2 (en) 2001-12-26 2003-07-17 Guilford Pharmaceuticals Change inhibitors of dipeptidyl peptidase iv
WO2003057666A2 (en) 2001-12-26 2003-07-17 Guilford Pharmaceuticals Inhibitors of dipeptidyl peptidase iv
EP1461337A1 (en) 2001-12-27 2004-09-29 F. Hoffmann-La Roche Ag Pyrido[2,1-a]isoquinoline derivatives as dpp-iv inhibitors
US20040176406A1 (en) 2001-12-27 2004-09-09 Gobbi Luca Claudio Pyrido [2,1-a] isoquinoline derivatives
US20030149071A1 (en) 2001-12-27 2003-08-07 Gobbi Luca Claudio Pyrido [2,1-a] isoquinoline derivatives
WO2003055881A1 (en) 2001-12-27 2003-07-10 F. Hoffmann-La Roche Ag Pyrido(2,1-a)isoquinoline derivatives as dpp-iv inhibitors
US6727261B2 (en) 2001-12-27 2004-04-27 Hoffman-La Roche Inc. Pyrido[2,1-A]Isoquinoline derivatives
WO2003072528A2 (en) 2002-02-08 2003-09-04 Idun Pharmaceuticals, Inc. (substituted)acyl dipeptidyl inhibitors of the ice/ced-3 family of cysteine proteases
US20030232788A1 (en) 2002-02-08 2003-12-18 Idun Pharmaceuticals, Inc. (Substituted)acyl dipeptidyl inhibitors of the ICE/ced-3 family of cysteine proteases
US20030216382A1 (en) 2002-02-13 2003-11-20 Markus Boehringer Pyridine and pyrimidine derivatives
EP1476429A1 (en) 2002-02-13 2004-11-17 F. Hoffmann-La Roche Ag Novel pyridine- and quinoline-derivatives
US6800650B2 (en) 2002-02-13 2004-10-05 Hoffmann-La Roche Inc. Pyridine and quinoline derivatives
US20030195188A1 (en) 2002-02-13 2003-10-16 Markus Boehringer Pyridine and quinoline derivatives
WO2003068757A1 (en) 2002-02-13 2003-08-21 F. Hoffmann-La Roche Ag Novel pyridin- and pyrimidin-derivatives
US6867205B2 (en) 2002-02-13 2005-03-15 Hoffman-La Roche Inc. Pyridine and pyrimidine derivatives
WO2003068748A1 (en) 2002-02-13 2003-08-21 F. Hoffmann-La Roche Ag Novel pyridine- and quinoline-derivatives
EP1476435A1 (en) 2002-02-13 2004-11-17 F. Hoffmann-La Roche Ag Novel pyridin- and pyrimidin-derivatives
EP1338592A1 (en) 2002-02-22 2003-08-27 Nippon Zoki Pharmaceutical Co., Ltd. Novel 2-phenylpiperazine derivatives
US20040082570A1 (en) 2002-02-25 2004-04-29 Eisai Co., Ltd. Xanthine derivative and DPPIV inhibitor
JP2004043429A (en) 2002-02-25 2004-02-12 Eisai Co Ltd New xanthine derivative and dppiv inhibitor
US20030162820A1 (en) 2002-02-28 2003-08-28 Hans-Ulrich Demuth Glutaminyl based DPIV inhibitors
EP1480961A1 (en) 2002-02-28 2004-12-01 Prosidion Limited Glutaminyl based dpiv inhibitors
WO2003074500A2 (en) 2002-03-06 2003-09-12 Sanofi-Aventis N-aminoacetyl-pyrrolidine-2-carbonitriles and their use as ddp-iv inhibitors
WO2003080633A1 (en) 2002-03-25 2003-10-02 Nippon Kayaku Kabushiki Kaisha Novel $g(a)-amino-n-(diaminophosphinyl)lactam derivative
WO2003082817A2 (en) 2002-03-25 2003-10-09 Merck & Co., Inc. Beta-amino heterocyclic dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
EP1490335A2 (en) 2002-03-25 2004-12-29 Merck & Co., Inc. Beta-amino heterocyclic dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
EP1489088A1 (en) 2002-03-25 2004-12-22 Nippon Kayaku Kabushiki Kaisha Novel alpha-amino-n-(diaminophosphinyl)lactam derivative
JP2004002368A (en) 2002-04-04 2004-01-08 Tanabe Seiyaku Co Ltd Pharmaceutical composition
JP2004002367A (en) 2002-04-04 2004-01-08 Tanabe Seiyaku Co Ltd Pharmaceutical composition
WO2003084940A1 (en) 2002-04-08 2003-10-16 Alangudi Sankaranarayanan Thiazolidine-4-carbonitriles and analogues and their use as dipeptidyl-peptidas inhibitors
EP1492777A1 (en) 2002-04-08 2005-01-05 Torrent Pharmaceuticals Ltd Thiazolidine-4-carbonitriles and analogues and their use as dipeptidyl-peptidase inhibitors
US20030225102A1 (en) 2002-04-08 2003-12-04 Torrent Pharmaceuticals Ltd. Novel compounds and therapeutic uses thereof
JP2003300977A (en) 2002-04-10 2003-10-21 Sumitomo Pharmaceut Co Ltd Xanthine derivative
WO2003095425A1 (en) 2002-05-09 2003-11-20 Taisho Pharmaceutical Co.,Ltd. Cyanopyrrolidine derivatives
JP2004026820A (en) 2002-05-09 2004-01-29 Taisho Pharmaceut Co Ltd Dipeptidyl peptidase iv inhibitor
JP2003327532A (en) 2002-05-10 2003-11-19 Takeda Chem Ind Ltd Peptidase inhibitor
WO2003099279A1 (en) 2002-05-29 2003-12-04 Novartis Ag Combination of a dpp iv inhibitor and a cardiovascular compound
WO2003101448A1 (en) 2002-06-03 2003-12-11 Novartis Ag The use of substituted cyanopyrrolidines and combination preparations containing them for treating hyperlipidemia and associated diseases
EP1513808A2 (en) 2002-06-04 2005-03-16 Pfizer Products Inc. Fluorinated cyclic amides as dipeptidyl peptidase iv inhibitors
US6710040B1 (en) 2002-06-04 2004-03-23 Pfizer Inc. Fluorinated cyclic amides as dipeptidyl peptidase IV inhibitors
US6812350B2 (en) 2002-06-04 2004-11-02 Pfizer Inc. Synthesis of 3,3,4,4-tetrafluoropyrrolidine and novel dipeptidyl peptidase-IV inhibitor compounds
WO2003101958A2 (en) 2002-06-04 2003-12-11 Pfizer Products Inc. Flourinated cyclic amides as dipeptidyl peptidase iv inhibitors
US20040242898A1 (en) 2002-06-04 2004-12-02 Pfizer Inc Synthesis of 3,3,4,4-tetrafluoropyrrolidine and novel dipeptidyl peptidase-IV inhibitor compounds
US20040116328A1 (en) 2002-06-06 2004-06-17 Eisai Co., Ltd. Condensed imidazole derivatives
WO2003104229A1 (en) 2002-06-06 2003-12-18 エーザイ株式会社 Novel fused imidazole derivative
WO2003105763A2 (en) 2002-06-14 2003-12-24 Amylin Pharmaceuticals, Inc. Prevention and/or treatment of inflammatory bowel disease using pyy or agonists thereof
WO2003106456A2 (en) 2002-06-14 2003-12-24 Sanofi-Synthelabo New compounds
EP1517907A2 (en) 2002-06-14 2005-03-30 Sanofi-Aventis Azabicyclo-octane and nonane derivatives with ddp-iv inhibiting activity
JP2004026678A (en) 2002-06-24 2004-01-29 Microbial Chem Res Found Therapeutic agent for type 2 diabetes
WO2004000327A1 (en) 2002-06-24 2003-12-31 Zaidan Hojin Biseibutsu Kagaku Kenkyu Kai Therapeutic agent for type 2 diabetes
WO2004004661A2 (en) 2002-07-09 2004-01-15 Point Therapeutics, Inc. Boroproline compound combination therapy
WO2004007446A1 (en) 2002-07-10 2004-01-22 Yamanouchi Pharmaceutical Co., Ltd. Novel azetidine derivative or salt thereof
WO2004007468A1 (en) 2002-07-15 2004-01-22 Merck & Co., Inc. Piperidino pyrimidine dipeptidyl peptidase inhibitors for the treatment of diabetes
WO2004009544A1 (en) 2002-07-23 2004-01-29 Yamanouchi Pharmaceutical Co., Ltd. 2-cyano-4-fluoropyrrolidine derivative or its salt
JP2004315496A (en) 2002-08-08 2004-11-11 Takeda Chem Ind Ltd Fused heterocyclic ring compound
WO2004014860A2 (en) 2002-08-08 2004-02-19 Takeda Pharmaceutical Company Limited Fused heterocyclic compounds as peptidase inhibitors
US20040097510A1 (en) 2002-08-21 2004-05-20 Boehringer Ingelheim Pharma Gmbh & Co. Kg 8-[3-amino-piperidin-1-yl]-xanthines, the preparation thereof and their use as pharmaceutical compositions
WO2004018468A2 (en) 2002-08-21 2004-03-04 Boehringer Ingelheim Pharma Gmbh & Co. Kg 8-[3-amino-piperidin-1-yl]-xanthines, the production thereof and the use of the same as medicaments
DE10238243A1 (en) 2002-08-21 2004-03-04 Boehringer Ingelheim Pharma Gmbh & Co. Kg New 8-(3-amino-piperidin-1-yl)-xanthine derivatives are dipeptidylpeptidase-IV inhibitors useful for, e.g. treating diabetes mellitus, arthritis or obesity
WO2004018467A2 (en) 2002-08-22 2004-03-04 Boehringer Ingelheim Pharma Gmbh & Co. Kg Phenacyl xanthine derivatives as dpp-iv inhibitor
DE10238470A1 (en) 2002-08-22 2004-03-04 Boehringer Ingelheim Pharma Gmbh & Co. Kg New xanthine derivatives, their production and their use as medicines
DE10238477A1 (en) 2002-08-22 2004-03-04 Boehringer Ingelheim Pharma Gmbh & Co. Kg New purine derivatives, their production and their use as medicines
WO2004018469A1 (en) 2002-08-22 2004-03-04 Boehringer Ingelheim Pharma Gmbh & Co. Kg Novel purine derivatives, production and use thereof as medicaments
WO2004020407A1 (en) 2002-08-29 2004-03-11 Taisho Pharmaceutical Co.,Ltd. Benzenesulfonate of 4-fluoro-2-cyanopyrrolidine derivative
WO2004026822A2 (en) 2002-09-19 2004-04-01 Abbott Laboratories Pharmaceutical compositions as inhibitors of dipeptidyl peptidase-iv (dpp-iv)
WO2004032836A2 (en) 2002-10-07 2004-04-22 Merck & Co., Inc. Antidiabetic beta-amino heterocylcic dipeptidyl peptidase inhibitors
WO2004033455A2 (en) 2002-10-08 2004-04-22 Novo Nordisk A/S Hemisuccinate salts of heterocyclic dpp-iv inhibitors
WO2004037169A2 (en) 2002-10-18 2004-05-06 Merck & Co., Inc. Beta-amino heterocyclic dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2004037181A2 (en) 2002-10-23 2004-05-06 Bristol-Myers Squibb Company Glycinenitrile-based inhibitors of dipeptidyl peptidase iv and methods
WO2004041795A1 (en) 2002-10-30 2004-05-21 Guilford Pharmaceuticals Inc. Novel inhibitors of dipeptidyl peptidase iv
WO2004048379A1 (en) 2002-11-01 2004-06-10 Sumitomo Pharmaceuticals Co., Ltd. Xanthine compound
WO2004043940A1 (en) 2002-11-07 2004-05-27 Merck & Co., Inc. Phenylalanine derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
US20040138214A1 (en) 2002-11-08 2004-07-15 Boehringer Ingelheim Pharma Gmbh & Co. Kg Xanthine derivatives, the preparation thereof and their use as pharmaceutical compositions
WO2004041820A1 (en) 2002-11-08 2004-05-21 Boehringer Ingelheim Pharma Gmbh & Co. Kg Novel xanthine derivatives, the production and the use thereof in the form of drugs
DE10251927A1 (en) 2002-11-08 2004-05-19 Boehringer Ingelheim Pharma Gmbh & Co. Kg New 1,7,8-trisubstituted xanthine derivatives, are dipeptidylpeptidase-IV inhibitors useful e.g. for treating diabetes mellitus type I or II, arthritis or obesity
US20040110817A1 (en) 2002-11-18 2004-06-10 Pfizer Inc Dipeptidyl peptidase IV inhibiting fluorinated cyclic amides
WO2004046106A1 (en) 2002-11-18 2004-06-03 Pfizer Products Inc. Dipeptidyl peptidase iv inhibiting fluorinated cyclic amides
WO2004050658A1 (en) 2002-12-03 2004-06-17 Boehringer Ingelheim Pharma Gmbh & Co. Kg Novel substituted imidazo-pyridinones and imidazo-pyridazeiones, the production and use thereof as medicaments
DE10256264A1 (en) 2002-12-03 2004-06-24 Boehringer Ingelheim Pharma Gmbh & Co. Kg New trisubstituted dihydro-imidazo-pyridazinone or imidazo-pyridinone derivatives, useful as dipeptidylpeptidase-IV inhibitors for e.g. treating diabetes mellitus type I or II, arthritis or obesity
WO2004050022A2 (en) 2002-12-04 2004-06-17 Merck & Co., Inc. Phenylalanine derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2004052850A2 (en) 2002-12-09 2004-06-24 Bristol-Myers Squibb Company Methods and compounds producing dipeptidyl peptidase iv inhibitors and intermediates thereof
WO2004052362A1 (en) 2002-12-10 2004-06-24 Novartis Ag Combination of an dpp-iv inhibitor and a ppar-alpha compound
WO2004058266A1 (en) 2002-12-20 2004-07-15 Merck & Co., Inc. 3-amino-4-phenylbutanoic acid derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2004065380A1 (en) 2003-01-14 2004-08-05 Arena Pharmaceuticals Inc. 1,2,3-trisubstituted aryl and heteroaryl derivatives as modulators of metabolism and the prpphylaxis and treatment of disorders related thereto such as diabetes and hyperglycemia
WO2004064778A2 (en) 2003-01-17 2004-08-05 Merck & Co. Inc. 3-amino-4-phenylbutanoic acid derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
JP2004244412A (en) 2003-01-20 2004-09-02 Kotobuki Seiyaku Kk 2-cyanopyrrolidine derivative having substituent at 4-position, method for producing the same, and medicament containing the same
WO2004067509A1 (en) 2003-01-31 2004-08-12 Sanwa Kagaku Kenkyusho Co., Ltd. Compound inhibiting dipeptidyl peptidase iv
WO2004069162A2 (en) 2003-01-31 2004-08-19 Merck & Co., Inc. 3-amino-4-phenylbutanoic acid derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2004071454A2 (en) 2003-02-13 2004-08-26 Guilford Pharmaceuticals Inc. Substituted azetidine compounds as inhibitors of dipeptidyl peptidase iv
WO2004076413A2 (en) 2003-02-24 2004-09-10 Arena Pharmaceuticals, Inc. Phenyl- and pyridylpiperidine-derivatives as modulators of glucose metabolism
WO2004076434A1 (en) 2003-02-28 2004-09-10 Aic Dipeptidyl peptidase inhibitors
WO2004076433A1 (en) 2003-02-28 2004-09-10 Aic Dipeptidyl peptidase inhibitors
WO2004085378A1 (en) 2003-03-19 2004-10-07 Merck & Co. Inc. Process for the preparation of chiral beta amino acid derivatives by asymmetric hydrogenation
WO2004085661A2 (en) 2003-03-24 2004-10-07 Merck & Co., Inc Process to chiral beta-amino acid derivatives
WO2004087053A2 (en) 2003-03-25 2004-10-14 Syrrx, Inc. Dipeptidyl peptidase inhibitors
WO2004087650A2 (en) 2003-03-27 2004-10-14 Merck & Co. Inc. Process and intermediates for the preparation of beta-amino acid amide dipeptidyl peptidase-iv inhibitors
WO2004092128A1 (en) 2003-04-10 2004-10-28 Smithkline Beecham Corporation Anhydrous crystalline forms of (2s, 4s)-1-{(2r)-2-amino-3-‘4-methoxybenzyl)sulfonyl!-3-methylbutanoyl}-4-fluoropyrrolindine-2-carbonitrile
WO2004096806A1 (en) 2003-04-30 2004-11-11 Sumitomo Pharmaceuticals Co. Ltd. Fused imidazole derivative
WO2004099134A2 (en) 2003-05-05 2004-11-18 Prosidion Ltd. Glutaminyl based dp iv-inhibitors
WO2004103993A1 (en) 2003-05-14 2004-12-02 Syrrx, Inc. Dipeptidyl peptidase inhibitors
WO2004103276A2 (en) 2003-05-14 2004-12-02 Merck & Co., Inc. 3-amino-4-phenylbutanoic acid derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
US20040254226A1 (en) 2003-05-14 2004-12-16 Syrrx, Inc. Dipeptidyl peptidase inhibitors
WO2004104215A2 (en) 2003-05-21 2004-12-02 Bayer Healthcare Ag Diagnostics and therapeutics for diseases associated with dipeptidylpeptidase 7 (dpp7)
WO2004104216A2 (en) 2003-05-21 2004-12-02 Bayer Healthcare Ag Diagnostics and therapeutics for diseases associated with dipeptidylpeptidase iv (dpp4)
WO2004108730A1 (en) 2003-06-05 2004-12-16 Fujisawa Pharmaceutical Co., Ltd. 1h-imidazo`4,5-d!pyridazines as dpp-iv inhibitors for the treatment of niddm
WO2004110375A2 (en) 2003-06-06 2004-12-23 Merck & Co., Inc. Combination therapy for the treatment of diabetes
WO2004110436A1 (en) 2003-06-06 2004-12-23 Merck & Co., Inc. Fused indoles as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2004111041A1 (en) 2003-06-12 2004-12-23 Fujisawa Pharmaceutical Co., Ltd. Pyrrolidine, thiazolidine and oxazolidine compounds which inhibit dipeptidyl peptidase-iv (dpp)
WO2004112701A2 (en) 2003-06-17 2004-12-29 Merck & Co., Inc. Cyclohexylglycine derivatives as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
DE10327439A1 (en) 2003-06-18 2005-01-05 Boehringer Ingelheim Pharma Gmbh & Co. Kg Novel imidazopyridazinone and imidazopyridone derivatives, their production and their use as pharmaceuticals
WO2004111051A1 (en) 2003-06-18 2004-12-23 Boehringer Ingelheim International Gmbh Imidazo-pyridazinone derivatives and imidazo-pyridone derivatives, production thereof, and use thereof as medicaments
WO2005000846A1 (en) 2003-06-20 2005-01-06 F.Hoffmann-La Roche Ag Hexahydropyridoisoqinolines as dpp-iv inhibitors
US20040259903A1 (en) 2003-06-20 2004-12-23 Markus Boehringer Pyrido [2,1-a] isoquinoline derivatives
US20040259902A1 (en) 2003-06-20 2004-12-23 Markus Boehringer Pyrido [2,1-a] isoquinoline derivatives
WO2005000848A1 (en) 2003-06-20 2005-01-06 F. Hoffmann-La Roche Ag Pyrido` 2, 1-a - isoquinoline derivatives as dpp-iv inhibitors
US20050032804A1 (en) 2003-06-24 2005-02-10 Cypes Stephen Howard Phosphoric acid salt of a dipeptidyl peptidase-IV inhibitor
WO2005003135A1 (en) 2003-06-24 2005-01-13 Merck & Co., Inc. Phosphoric acid salt of a dipeptidyl peptidase-iv inhibitor
JP2005023038A (en) 2003-07-04 2005-01-27 Taisho Pharmaceut Co Ltd Therapeutic agent for chronic renal disease
WO2005007647A1 (en) 2003-07-11 2005-01-27 Arena Pharmaceuticals, Inc. Trisubstituted aryl and heteroaryl derivatives as modulators of metabolism and the prophylaxis and treatment of disorders related thereto
WO2005009956A1 (en) 2003-07-21 2005-02-03 Smithkline Beecham Corporation (2s,4s)-4-fluoro-1-[4-fluoro-beta-(4-fluorophenyl)-l-phenylalanyl]-2-pyrrolidinecarbonitrile p-toluenesulfonic acid salt and anhydrous crystalline forms thereof
US20050059724A1 (en) 2003-07-25 2005-03-17 Aventis Pharma Deutschland Gmbh Novel cyanopyrrolidides, process for their preparation and their use as medicaments
DE10333935A1 (en) 2003-07-25 2005-02-24 Aventis Pharma Deutschland Gmbh New bicyclic cyano-heterocycles, process for their preparation and their use as pharmaceuticals
US20050059716A1 (en) 2003-07-25 2005-03-17 Aventis Pharma Deutschland Gmbh Novel bicyclic cyanoheterocycles, process for their preparation and their use as medicaments
WO2005012308A1 (en) 2003-07-25 2005-02-10 Sanofi-Aventis Deutschland Gmbh Novel cyanopyrrolidides, methods for the production thereof, and use of the same as medicaments
WO2005012312A1 (en) 2003-07-25 2005-02-10 Sanofi-Aventis Deutschland Gmbh Novel cyano thiazolides, methods for the production thereof, and use thereof as a medicament
WO2005011581A2 (en) 2003-07-31 2005-02-10 Merck & Co., Inc. Hexahydrodiazepinones as dipeptidyl peptidase-iv inhibitors for the treatment or prevention of diabetes
US20050038020A1 (en) 2003-08-01 2005-02-17 Hamann Lawrence G. Adamantylglycine-based inhibitors of dipeptidyl peptidase IV and methods
WO2005012249A2 (en) 2003-08-01 2005-02-10 Bristol-Myers Squibb Company Adamantyglycine- based inhibitors of dipeptidyl peptidase iv for the treatment of diabetes
US20050043292A1 (en) 2003-08-20 2005-02-24 Pfizer Inc Fluorinated lysine derivatives as dipeptidyl peptidase IV inhibitors
WO2005019168A2 (en) 2003-08-20 2005-03-03 Pfizer Products Inc. Fluorinated lysine derivatives as dipeptidyl peptidase iv inhibitors
WO2005020920A2 (en) 2003-09-02 2005-03-10 Merck & Co., Inc. Novel crystalline forms of a phosphoric acid salt of a dipeptidyl peptidase-iv inhibitor
WO2005023762A1 (en) 2003-09-04 2005-03-17 Abbott Laboratories Pyrrolidine-2-carbonitrile derivatives and their use as inhibitors of dipeptidyl peptidase-iv (dpp-iv)
WO2005026148A1 (en) 2003-09-08 2005-03-24 Takeda San Diego, Inc. Dipeptidyl peptidase inhibitors
WO2005030751A2 (en) 2003-09-08 2005-04-07 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
WO2005025554A2 (en) 2003-09-09 2005-03-24 Japan Tobacco Inc. Dipeptidyl peptidase iv inhibitor
WO2005030127A2 (en) 2003-09-23 2005-04-07 Merck & Co., Inc. Novel crystalline form of a phosphoric acid salt of a dipeptidyl peptidase-iv inhibitor
WO2005032590A1 (en) 2003-10-03 2005-04-14 Takeda Pharmaceutical Company Limited Remedy for diabetes
WO2005033099A2 (en) 2003-10-03 2005-04-14 Glenmark Pharmaceuticals Ltd. Novel dipeptidyl peptidase iv inhibitors; processes for their preparation and compositions thereof
WO2005034940A2 (en) 2003-10-15 2005-04-21 Imtm Gmbh Dual alanyl aminopeptidase and dipeptidyl peptidase iv inhibitors for functionally influencing different cells and for treating immunological, inflammatory, neuronal and other diseases
WO2005037779A2 (en) 2003-10-15 2005-04-28 Imtm Gmbh Novel dipeptidyl peptidase iv inhibitors used for functionally influencing different cells and treating immunological, inflammatory, neuronal, and other diseases
WO2005040095A1 (en) 2003-10-16 2005-05-06 Astrazeneca Ab Inhibitors of dipeptidyl peptidase iv
WO2005037828A1 (en) 2003-10-20 2005-04-28 Lg Life Sciences Ltd. Novel inhibitors of dpp-iv, methods of preparing the same, and pharmaceutical compositions containing the same as an active agent
WO2005042488A1 (en) 2003-10-31 2005-05-12 Takeda Pharmaceutical Company Limited Pyridine compounds as inhibitors of dipeptidyl peptidase iv
WO2005044195A2 (en) 2003-11-04 2005-05-19 Merck & Co., Inc. Fused phenylalanine derivatives as dipeptidyl peptidase-iv inhibitors for the treatment or prevention of diabetes
WO2005047297A1 (en) 2003-11-12 2005-05-26 Phenomix Corporation Heterocyclic boronic acid compounds
WO2005049022A2 (en) 2003-11-17 2005-06-02 Novartis Ag Use of dipeptidyl peptidase iv inhibitors
WO2005058849A1 (en) 2003-12-15 2005-06-30 Glenmark Pharmaceuticals Ltd. New dipeptidyl peptidase in inhibitors; process for their preparation and compositions containing them
WO2005063750A1 (en) 2003-12-23 2005-07-14 Boehringer Ingelheim International Gmbh Bicyclic imidazole compounds, the production thereof and their use as medicaments
WO2005072530A1 (en) 2004-01-16 2005-08-11 Merck & Co., Inc. Novel crystalline salts of a dipeptidyl peptidase-iv inhibitor
WO2005075426A1 (en) 2004-02-03 2005-08-18 Glenmark Pharmaceuticals Ltd. Novel dipeptidyl peptidase iv inhibitors; processes for their preparation and compositions thereof
US20080146818A1 (en) 2004-02-05 2008-06-19 Yasumichi Fukuda Bicycloester Derivative
WO2005079795A2 (en) 2004-02-20 2005-09-01 Novartis Ag Dpp-iv inhibitors for treating neurodegeneration and cognitive disorders
WO2005082849A1 (en) 2004-02-23 2005-09-09 Trustees Of Tufts College Lactams as conformationally constrained peptidomimetic inhibitors
WO2005082348A2 (en) 2004-02-23 2005-09-09 Trustees Of Tufts College Inhibitors of dipeptidylpeptidase iv for regulating glucose metabolism
WO2005087235A1 (en) 2004-03-09 2005-09-22 National Health Research Institutes Pyrrolidine compounds
WO2005095381A1 (en) 2004-03-15 2005-10-13 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
WO2005116014A1 (en) 2004-05-12 2005-12-08 Pfizer Products Inc. Proline derivatives and their use as dipeptidyl peptidase iv inhibitors
US20070167468A1 (en) 2004-08-06 2007-07-19 Sanofi-Aventis Deutschland Gmbh Substituted bicyclic 8-pyrr0lidinoxanthines, methods for their production, pharmaceutical formulations and their use
WO2006040625A1 (en) 2004-10-12 2006-04-20 Glenmark Pharmaceuticals S.A. Novel dipeptidyl peptidase iv inhibitors, pharmaceutical compositions containing them, and process for their preparation
US20090192129A1 (en) 2004-12-12 2009-07-30 Dainippon Sumitomo Pharma Co., Ltd. Bicyclic pyrrole derivatives
WO2006068163A1 (en) 2004-12-24 2006-06-29 Dainippon Sumitomo Pharma Co., Ltd. Bicyclic pyrrole derivatives
WO2006076231A2 (en) 2005-01-10 2006-07-20 Arena Pharmaceuticals, Inc. Combination therapy for the treatment of diabetes and conditions related thereto and for the treatment of conditions ameliorated by increasing a blood glp-1 level
WO2006088129A1 (en) 2005-02-18 2006-08-24 Mitsubishi Pharma Corporation Salt of proline derivative, solvate thereof, and production method thereof
US20090216016A1 (en) 2005-02-18 2009-08-27 Mitsubishi Pharma Corporation Salt of proline derivative, solvate thereof, and production method thereof
WO2006100181A2 (en) 2005-03-22 2006-09-28 F. Hoffmann-La Roche Ag New salt and polymorphs of a dpp-iv inhibitor
WO2006116157A2 (en) 2005-04-22 2006-11-02 Alantos Pharmaceuticals Holding, Inc. Dipeptidyl peptidase-iv inhibitors
EP1902730A1 (en) 2005-06-09 2008-03-26 Banyu Pharmaceutical Co., Ltd. Npy y2 agonist for use as therapeutic agent for disease accompanied by diarrhea
WO2007019255A2 (en) 2005-08-04 2007-02-15 Novartis Ag Salts of vildagliptin
WO2007027651A2 (en) 2005-08-30 2007-03-08 Abbott Laboratories Pharmaceutical compositions as inhibitors of dipeptidyl peptidase-iv (dpp-iv)
WO2007035372A2 (en) 2005-09-16 2007-03-29 Takeda Pharmaceutical Company Limited Polymorphs of benzoate salt of 2-[[6-[(3r)-3-amino-1-piperidinyl]-3,4-dihydro-3-methyl-2,4-dioxo-1(2h)-pyrimidinyl]methyl]-benzonitrile and methods of use therefor
WO2007071576A1 (en) 2005-12-21 2007-06-28 F. Hoffmann-La Roche Ag New salt and polymorph of dpp-iv inhibitor
WO2007120702A2 (en) 2006-04-11 2007-10-25 Arena Pharmaceuticals, Inc. Use of gpr119 receptor agonists for increasing bone mass and for treating osteoporosis, and combination therapy relating thereto
WO2007120689A2 (en) 2006-04-11 2007-10-25 Arena Pharmaceuticals, Inc. Methods of using gpr119 receptor to identify compounds useful for increasing bone mass in an individual
WO2007128721A1 (en) 2006-05-04 2007-11-15 Boehringer Ingelheim Internationalgmbh Polymorphs
WO2007148185A2 (en) 2006-06-21 2007-12-27 Pfizer Products Inc. Substituted 3 -amino- pyrrolidino-4 -lactams as dpp inhibitors
WO2008027273A2 (en) 2006-08-30 2008-03-06 Phenomix Corporation Solid citrate and tartrate salts of dpp-iv inhibitors
WO2008070692A2 (en) * 2006-12-06 2008-06-12 Smithkline Beecham Corporation Bicyclic compounds and use as antidiabetics
WO2008114857A1 (en) 2007-03-22 2008-09-25 Kyorin Pharmaceutical Co., Ltd. Method for producing aminoacetylpyrrolidinecarbonitrile derivative
WO2009084497A1 (en) 2007-12-28 2009-07-09 Dainippon Sumitomo Pharma Co., Ltd. Methyl-substituted piperidine derivative
WO2009126245A1 (en) 2008-04-07 2009-10-15 Arena Pharmaceuticals, Inc. Methods of using a g protein-coupled receptor to identify peptide yy (pyy) secretagogues and compounds useful in the treatment of conditions modulated by pyy

Non-Patent Citations (109)

* Cited by examiner, † Cited by third party
Title
ABBOTT ET AL., BRAIN RES, vol. 1043, 2005, pages 139 - 144
ABE ET AL., J NAT PROD, vol. 67, 2004, pages 999 - 1004
ADRIAN ET AL., GASTROENTEROL, vol. 89, 1985, pages 1070 - 1077
AHREN ET AL., DIABETES CARE, vol. 25, 2002, pages 869 - 875
AHREN ET AL., ENDOCRINOLOGY, vol. 146, 2005, pages 2055 - 2059
AHREN ET AL., J CLIN ENDOCRINOL METAB, vol. 89, 2004, pages 2078 - 2084
AREHART ET AL., CIRC RES, CIRC. RES., vol. 102, 2008, pages 986 - 993
ATIK ET AL., CLIN ORTHOP RELAT RES, vol. 443, 2006, pages 19 - 24
BALASUBRAMANIAM ET AL., J. MED. CHEM., vol. 43, 2000, pages 3420 - 3427
BALASUBRAMANIAM ET AL., PEPTIDES, vol. 28, 2007, pages 235 - 240
BATTERHAM ET AL., NATURE, vol. 418, 2002, pages 650 - 654
BEHRE, SCAND J CLIN LAB INVEST, vol. 67, 2007, pages 449 - 458
BILCHIK ET AL., GASTROENTEROL, vol. 105, 1993, pages 1441 - 1448
BOEY ET AL., DIABETOLOGIA, vol. 49, 2006, pages 1360 - 1370
BOEY ET AL., NEUROPEPTIDES, vol. 42, 2008, pages 19 - 30
BOLLAG ET AL., ENDOCRINOLOGY, vol. 141, 2000, pages 1228 - 1235
BOLLAG ET AL., MOL CELL ENDOCRINOL, vol. 177, 2001, pages 35 - 41
BOSE ET AL., DIABETES, vol. 54, 2005, pages 146 - 151
BRADLEY, JPATHOL, vol. 214, 2008, pages 149 - 160
BRANCATI, F. L. ET AL., ARCH. INTERN. MED., vol. 159, 1999, pages 957 - 963
CALDWELL ET AL., BIOORG MED CHEM LETT, vol. 14, 2004, pages 1265 - 1268
COX, PEPTIDES, vol. 28, 2007, pages 345 - 351
CRUZE ET AL., PEPTIDES, vol. 28, 2007, pages 269 - 280
DEACON ET AL., J CLIN ENDOCRINOL METAB, vol. 85, 2000, pages 3575 - 3581
DEACON, REGUL PEPT, vol. 128, 2005, pages 117 - 124
DRUCKER, CELL METAB, vol. 3, 2006, pages 153 - 165
DURING ET AL., NAT. MED., vol. 9, 2003, pages 1173 - 1179
EBERLEIN ET AL., PEPTIDES, vol. 10, 1989, pages 797 - 803
EDMONDSON ET AL., BIOORG MED CHEM LETT, vol. 14, 2004, pages 5151 - 5155
EKBLAD ET AL., PEPTIDES, vol. 23, 2002, pages 251 - 261
EKSTRAND ET AL., PNAS USA, vol. 100, 2003, pages 6033 - 6038
EL BAHH ET AL., EUR. J. NEUROSCI., vol. 22, 2005, pages 1417 - 1430
G. P. STAHLY, CRYSTAL GROWTH & DESIGN, vol. 7, no. 6, 2007, pages 1007 - 1026
GONON ET AL., CARDIOVASC RES., vol. 78, 2008, pages 116 - 122
GRANDT ET AL., REGUL. PEPT., vol. 51, 1994, pages 151 - 159
GREIG ET AL., ANN N YACAD SCI, vol. 1035, 2004, pages 290 - 315
GRISE ET AL., J. SURG. RES., vol. 82, 1999, pages 151 - 155
GUERRE-MILLO, DIABETES & METABOLISM, vol. 34, 2008, pages 12 - 18
H.-U. DEMUTH ET AL.: "Type 2 diabetes-Therapy with dipeptidyl peptidase IV inhibitors", BIOCHIM. BIOPHYS. ACTA, vol. 1751, 2005, pages 33 - 44
HANSMANN ET AL., CIRCULATION, vol. 115, 2007, pages 1275 - 1284
HARA ET AL., DIABETES CARE, vol. 29, 2006, pages 1357 - 1362
HAY ET AL., J CLIN GASTROENTEROL, vol. 14, 1992, pages 309 - 317
HILL, J. 0. ET AL., SCIENCE, vol. 280, 1998, pages 1371 - 1374
JONES ET AL., ANN. REP. MED. CHEM., vol. 44, 2009, pages 149 - 170
JONES, EXPERT OPIN. THER. PATENTS, vol. 19, no. 10, 2009, pages 1339 - 1359
K. AUGUSTYNS ET AL.: "Inhibitors of proline-specific dipeptidyl peptidases: DPP-IV inhibitors as a novel approach for the treatment of Type 2 diabetes", EXPERT OPIN. THER. PATENTS, vol. 15, 2005, pages 1387 - 1407
KEIGHLEY ET AL., AILMENT PHARMACOL THER, vol. 18, 2003, pages 66 - 70
KEIRE ET AL., AM. J. PHYSIOL. GASTROINTEST. LIVER PHYSIOL., vol. 279, 2000, pages G126 - G131
KUBOTA ET AL., J. BIOL. CHEM., vol. 277, 2002, pages 25863 - 25866
LAMB ET AL., J. MED. CHEM., vol. 50, 2007, pages 2264 - 2268
LE STUNFF ET AL., DIABETES, vol. 43, 1989, pages 696 - 702
LEE ET AL., J. CLIN. INVEST., vol. 111, 2003, pages 1853 - 1862
LEE ET AL., PEPTIDES, vol. 24, 2003, pages 99 - 106
LIU ET AL., AM SURG, vol. 62, 1996, pages 232 - 236
LIU ET AL., J. SURG. RES., vol. 58, 1995, pages 6 - 11
LIU ET AL., J. SURG. RES., vol. 58, 1995, pages 707 - 712
LIU ET AL., SURGERY, vol. 118, 1995, pages 229 - 236
LUNDBERG ET AL., PNAS USA, vol. 79, 1982, pages 4471 - 4475
MAEDA ET AL., NAT. MED., vol. 8, 2002, pages 731 - 737
MATSUDA ET AL., JBIOL CHEM, vol. 277, 2002, pages 37487 - 37491
MCFADDEN ET AL., AM. J. SURG., vol. 188, 2004, pages 516 - 519
MCINTOSH ET AL., REGUL PEPT, vol. 128, 2005, pages 159 - 165
MENTLEIN, EXPERT OPIN INVESTIG DRUGS, vol. 14, 2005, pages 57 - 64
MORLEY ET AL., LIJE SCIENCES, vol. 41, 1987, pages 2157 - 2165
NAUCK ET AL., DIABETES, vol. 53, no. 3, 2004, pages 190 - 196
NAUCK ET AL., DRUG NEWS PERSPECT, vol. 16, 2003, pages 413 - 422
NIGHTINGALE ET AL., GUT, vol. 39, 1996, pages 267 - 272
NISHIMURA ET AL., CIRCULATION, vol. 117, 2008, pages 216 - 223
O'BRIEN, M.; DAILY, B.; SCHURRIA, M.: "Assay for DPPIV activity using a homogeneous, luminescent method", CELL NOTE, 2005
OHASHI ET AL., HYPERTENSION, vol. 47, 2006, pages 1108 - 1116
OKU ET AL., FEBS LETTERS, vol. 581, 2007, pages 5029 - 5033
ORTIZ ET AL., JPET, vol. 323, 2007, pages 692 - 700
OUCHI ET AL., CIRCULATION, vol. 100, 1999, pages 2473 - 2476
OUCHI ET AL., CLIN CHIM ACTA, vol. 380, 2007, pages 24 - 30
PARKER ET AL., BR. J. PHARMACOL., vol. 153, 2008, pages 420 - 431
PEARSON, NURSING TIMES, vol. 100, 2004, pages 86 - 90
PEDERSON, P., DIAB. METAB. REV., vol. 5, 1989, pages 505 - 509
PERRY, I. J. ET AL., BMJ, vol. 310, 1995, pages 560 - 564
PETERS ET AL., BIOORG MED CHEM LETT, vol. 14, 2004, pages 1491 - 1493
PITTNER ET AL., INT. J. OBES. RELAT. METAB. DISORD., vol. 28, 2004, pages 963 - 971
RAISZ, J CLIN INVEST, vol. 115, 2005, pages 3318 - 3325
REED; SCRIBNER, DIABETES, OBESITY AND METABOLISM, vol. 1, 1999, pages 75 - 86
RENSHAW ET AL., CURRENT DRUG TARGETS, vol. 6, 2005, pages 171 - 179
ROTWEIN, R. ET AL., N. ENGL. J. MED., vol. 308, 1983, pages 65 - 71
RUGGERI, NAT MED, vol. 8, 2002, pages 1227 - 1234
SCHWARTZ, CELL METABOLISM, vol. 11, 2010, pages 445 - 447
SHAH, CURRENT OPINION IN DRUG DISCOVERY & DEVELOPMENT, vol. 12, 2009, pages 519 - 532
SHIBATA ET AL., J. BIOL. CHEM., vol. 279, 2004, pages 28670 - 28674
SHIBATA ET AL., J. MOL. CELL CARDIOL., vol. 42, 2007, pages 1065 - 1074
SHIBATA ET AL., NAT MED, vol. 11, 2005, pages 1096 - 1103
SHORE ET AL., J. ALLERGY CLIN. IMMUNOL, vol. 118, 2006, pages 389 - 395
SUMMER ET AL., AM J. PHYSIOL. LUNG CELL MOL. PHYSIOL, 7 March 2008 (2008-03-07)
TAO ET AL., CIRCULATION, vol. 115, 2007, pages 1408 - 1416
TATEMOTO ET AL., NATURE, vol. 285, 1980, pages 417 - 418
TILG ET AL., NAT. REV. IMMUNOL., vol. 6, 2006, pages 772 - 783
TSENG ET AL., PEPTIDES, vol. 23, 2002, pages 389 - 395
TSUKIYAMA ET AL., MOL ENDOCRINOL, vol. 20, 2006, pages 1644 - 1651
UENO ET AL., REGUL PEPT, vol. 145, 2008, pages 12 - 16
VILLHAUER ET AL., J MED CHEM, vol. 45, 2002, pages 2362 - 2365
VILLHAUER ET AL., J MED CHEM, vol. 46, 2003, pages 2774 - 2789
VONA-DAVIS ET AL., PEPTIDES, vol. 28, 2007, pages 334 - 338
WOLDBYE ET AL., NEUROBIOLOGY OFDISEASE, vol. 20, 2005, pages 760 - 772
WORLD HEALTH ORGANIZATION TECHNICAL REPORT SERIES 921, 2003
WORTLEY ET AL., GASTROENTEROL, vol. 133, 2007, pages 1534 - 1543
XIE ET AL., BONE, vol. 37, 2005, pages 759 - 769
YAMAMOTO ET AL., CLINICAL SCIENCE, vol. 103, 2002, pages 137 - 142
YOKOTA ET AL., BLOOD, vol. 96, 2000, pages 1723 - 1732
ZANDER ET AL., LANCET, vol. 359, 2002, pages 824 - 830
ZHONG ET AL., AM JPHYSIOL ENDOCRINOL METAB, vol. 292, 2007, pages E543 - E548

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8933083B2 (en) 2003-01-14 2015-01-13 Arena Pharmaceuticals, Inc. 1,2,3-trisubstituted aryl and heteroaryl derivatives as modulators of metabolism and the prophylaxis and treatment of disorders related thereto such as diabetes and hyperglycemia
US10894787B2 (en) 2010-09-22 2021-01-19 Arena Pharmaceuticals, Inc. Modulators of the GPR119 receptor and the treatment of disorders related thereto
WO2012099915A1 (en) * 2011-01-19 2012-07-26 Hongwen Zhu Thiazolidine derivatives and their therapeutic use
WO2012170867A1 (en) 2011-06-09 2012-12-13 Rhizen Pharmaceuticals Sa Novel compounds as modulators of gpr-119
US9944600B2 (en) 2012-06-12 2018-04-17 Chong Kun Dang Pharmaceutical Corp. Piperidine derivatives for GPR119 agonist
WO2014011926A1 (en) 2012-07-11 2014-01-16 Elcelyx Therapeutics, Inc. Compositions comprising statins, biguanides and further agents for reducing cardiometabolic risk
US9096641B2 (en) 2013-06-05 2015-08-04 Evestra, Inc. Imidazolyl progesterone antagonists
US9776987B2 (en) 2013-11-26 2017-10-03 Chong Kun Dang Pharmaceutical Corp Amide derivatives for GPR119 agonist
US11007175B2 (en) 2015-01-06 2021-05-18 Arena Pharmaceuticals, Inc. Methods of treating conditions related to the S1P1 receptor
US11884626B2 (en) 2015-06-22 2024-01-30 Arena Pharmaceuticals, Inc. Crystalline L-arginine salt of (R)-2-(7-(4-cyclopentyl-3-(trifluoromethyl)benzyloxy)-1,2,3,4-tetrahydrocyclo-penta [b]indol-3-yl)acetic acid(Compound1) for use in S1P1 receptor-associated disorders
US11534424B2 (en) 2017-02-16 2022-12-27 Arena Pharmaceuticals, Inc. Compounds and methods for treatment of primary biliary cholangitis

Also Published As

Publication number Publication date
TW201105327A (en) 2011-02-16
AR077642A1 (en) 2011-09-14

Similar Documents

Publication Publication Date Title
US20210340133A1 (en) Modulators of the gpr119 receptor and the treatment of disorders related thereto
WO2011005929A1 (en) Piperidine derivative and its use for the treatment of diabets and obesity
US20130023494A1 (en) Modulators of the gpr119 receptor and the treatment of disorders related thereto
US20140038889A1 (en) Modulators Of The GPR119 Receptor And The Treatment Of Disorders Related Thereto
US20140051714A1 (en) Modulators Of The GPR119 Receptor And The Treatment Of Disorders Related Thereto
US20140066369A1 (en) Modulators Of The GPR119 Receptor And The Treatment Of Disorders Related Thereto
US20140018371A1 (en) Modulators Of The GPR119 Receptor And The Treatment Of Disorders Related Thereto
WO2012170702A1 (en) Modulators of the gpr119 receptor and the treatment of disorders related thereto
JP2009542702A (en) Treatment of metabolic regulators and disorders related to metabolism
WO2013055910A1 (en) Modulators of the gpr119 receptor and the treatment of disorders related thereto
JP2009542699A (en) Treatment of metabolic regulators and disorders related to metabolism
JP2009508861A (en) Treatment of metabolic regulators and disorders related to metabolism
JP2008540426A (en) Combination of dipeptidyl peptidase IV inhibitor and cannabinoid CB1 receptor antagonist for the treatment of diabetes and obesity
US20060270722A1 (en) Combination of a dipeptidyl peptidase-IV inhibitor and a dual PPAR agonist for the treatment of diabetes and obesity
KR20130138768A (en) Fast-dissolve dosage forms of 5-ht2c agonists
EP2611433A2 (en) Non-hygroscopic salts of 5-ht2c agonists
WO2014074668A1 (en) Modulators of gpr119 and the treatment of disorders related thereto
EA040124B1 (en) GPR119 RECEPTOR MODULATORS AND THE TREATMENT OF RELATED DISORDERS

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: 10730966

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 10730966

Country of ref document: EP

Kind code of ref document: A1