WO2014074668A1 - Modulators of gpr119 and the treatment of disorders related thereto - Google Patents

Modulators of gpr119 and the treatment of disorders related thereto Download PDF

Info

Publication number
WO2014074668A1
WO2014074668A1 PCT/US2013/068864 US2013068864W WO2014074668A1 WO 2014074668 A1 WO2014074668 A1 WO 2014074668A1 US 2013068864 W US2013068864 W US 2013068864W WO 2014074668 A1 WO2014074668 A1 WO 2014074668A1
Authority
WO
WIPO (PCT)
Prior art keywords
yloxy
pharmaceutically acceptable
pharmaceutical
disorder
pharmaceutical agent
Prior art date
Application number
PCT/US2013/068864
Other languages
French (fr)
Inventor
Robert M. Jones
Daniel J. Buzard
Sangdon Han
Sun Hee Kim
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 WO2014074668A1 publication Critical patent/WO2014074668A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings

Definitions

  • the present invention relates to compounds of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, that are useful as single pharmaceutical agents or in combination with one or more additional pharmaceutical agents, such as, an inhibitor of DPP-IV, a biguanide, an alpha-glucosidase inhibitor, an insulin analogue, a sulfonylurea, an SGLT2 inhibitor, a meglitinide, a thiazolidinedione, an anti-diabetic peptide analogue, or a DGAT-1 inhibitor, in the treatment of, for example, a disorder selected from: a GPRl 19-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic-related disorder; type 2 diabetes; obesity; and complications related thereto.
  • additional pharmaceutical agents such as, an inhibitor of DPP-IV, a bigu
  • 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 is characterized by a relative disparity between endogenous insulin production and insulin requirements, leading to elevated blood glucose levels.
  • endogenous insulin production in NIDDM 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.
  • NIDDM is more common among Native Americans, African-Americans, Latinos, and Hispanics.
  • the onset can be insidious or even clinically unapparent, making diagnosis difficult.
  • NIDDM neurodegenerative disease
  • 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).
  • 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 25% of all post-menopausal white women, and the proportion rises to 70% in women older than 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.
  • 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.
  • Crohn' s disease 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 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.
  • GPR119 is a G protein-coupled receptor (GPR119; e.g., human GPR119, GenBank® Accession No. AAP72125 and alleles thereof; e.g., mouse GPR119, GenBank® Accession No. AY288423 and alleles thereof) and is selectively expressed on pancreatic beta cells. GPR119 activation leads to elevation of a level of intracellular cAMP, consistent with GPR119 being coupled to Gs. Agonists to GPR119 stimulate glucose-dependent insulin secretion in vitro and lower an elevated blood glucose level in vivo; see, e.g., International Applications WO
  • GPR119 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).
  • GPR119 agonists also stimulate the release of Glucose-dependent Insulinotropic Polypeptide (GIP), Glucagon-Like Peptide-1 (GLP-1), 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 GPR119 agonists and the release of:
  • GIP Glucose-dependent Insulinotropic Polypeptide
  • GLP-1 Glucagon-Like Peptide-1
  • PYY Peptide YY
  • GLP-1 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
  • GPR119 agonists enhance incretin release and therefore can be used in treatment of disorders related to the incretins, such as, GIP, GLP-1, and PYY.
  • GIP and GLP-1 are substrates for the enzyme DPP-IV. Jones and co-workers (Jones, et al., Ann. Rep. Med.
  • 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(l-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 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.
  • 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.
  • GLP-1 Glucagon-Like Peptide-1
  • Glucagon-like peptide-1 (GLP-1) is an incretin hormone derived from the
  • GLP-1 mediates its actions through a specific G protein-coupled receptor (GPCR), namely GLP-1R.
  • 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.
  • 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-1 has been shown to be cardioprotective in a rat model of myocardial infarction [Bose et al, Diabetes (2005) 54: 146- 151], and GLP-1R 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] .
  • 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- 1R 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.
  • 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 jejunum, high in ileum and colon, and highest in rectum (Lundberg et al, PNAS USA (1982) 79:4471-4475; Adrian et al, Gastroenterol.
  • PYYi_ 36 is generated from PYYi_ 36 by cleavage of the N-terminal Tyr and Pro residues by dipeptidyl peptidase IV.
  • PYY 3 . 36 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.
  • 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 . 36 was found to significantly decrease appetite and reduce food intake by 33% over 24 hours.
  • Peripheral administration of PYY 3 . 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 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 PYY 3 . 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- 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).
  • 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 PYY 3 . 36 inhibit bowel motility and work to prevent diarrhea (EP1902730; also see Cox, Peptides (2007) 28:345-351).
  • Y2R agonists such as PYYi_ 36 and PYY 3 . 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 PYY 3 _ 36 can confer protection in rodent models of pancreatitis (Vona-Davis et al, Peptides (2007) 28:334-338).
  • 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 PYY 3 . 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 PYY 3 . 36 promote wound healing. It has been reported that ischemic angiogenesis is impaired in Y2R-deficient mice (Lee et al, J. Clin. Invest.
  • Y2R such as PYYi_ 36 and PYY 3 .
  • 36 promotes revascularization and restoration of function of ischemic tissue. It has been reported that agonists of Y2R such as and PYY 3 . 36 mediate increases in collateral-dependent blood flow in a rat model of peripheral arterial disease (Cruze et al, Peptides (2007) 28:269-280).
  • PYY and Y2R agonists such as PYY 3 . 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
  • 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
  • Adiponectin is an adipokine with potent anti-inflammatory properties (Ouchi et al, Clin Chim Acta (2007) 380:24-30; Tilg 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.
  • adiponectin has been implicated in high density lipoprotein (HDL) assembly (Oku et al, FEBS Letters (2007) 581 :5029-5033).
  • HDL high density lipoprotein
  • 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 (2007) 115: 1408- 1416). Adiponectin has been reported to confer protection against myocardial ischemia- 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 protect against myocyte and capillary loss (Shibata et al, J. Mol. 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 Mol. 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)-a-induced expression of the adhesion molecules VCAM-1, E-selectin and ICAM- 1 in human aortic endothelial cells (HAECs) (Ouchi et al, Circulation (1999) 100:2473-2476) and to inhibit production of TNF-a 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-a- 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).
  • One aspect of the present invention is directed to compounds, as described herein, and pharmaceutically acceptable salts, solvates, and hydrates thereof, which bind to and modulate the activity of a GPCR, referred to herein as GPR119, and uses thereof.
  • GPR119 a GPCR
  • One aspect of the present invention encompasses, inter alia, certain cyclohexane derivatives selected from compounds of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • X is N or CH
  • R 1 is selected from: Ci-C 6 alkoxycarbonyl, C 3 -C 7 cycloalkoxycarbonyl,
  • heterocyclyloxycarbonyl, and heteroaryl each optionally substituted with one or more substituents selected from: C 1 -C6 alkyl, C 1 -C6 haloalkyl, and halogen; provided that R 1 is a group other than 4-(trifluoromethyl)pyrimidin-2-yl.
  • One aspect of the present invention encompasses, inter alia, certain cyclohexane derivatives selected from compounds of Formula (lb) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • One aspect of the present invention encompasses, inter alia, certain cyclohexane derivatives selected from compounds of Formula (Id) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • compositions comprising a compound of the present invention.
  • One aspect of the present invention pertains to methods for preparing a composition comprising the step of admixing a compound of the present invention, and a pharmaceutically acceptable carrier.
  • One aspect of the present invention pertains to a pharmaceutical product selected from: a pharmaceutical composition, a formulation, a unit dosage form, and a kit; each comprising a compound of the present invention.
  • One aspect of the present invention pertains to pharmaceutical compositions comprising a compound of the present invention, and a pharmaceutically acceptable carrier.
  • One aspect of the present invention pertains to methods for preparing a pharmaceutical composition
  • methods for preparing a pharmaceutical composition comprising the step of admixing a compound of the present invention, and a pharmaceutically acceptable carrier.
  • One aspect of the present invention pertains to pharmaceutical compositions obtained by any of the methods described herein.
  • compositions comprising a compound of the present invention, and a second pharmaceutical agent.
  • One aspect of the present invention pertains to methods for preparing a composition comprising the step of admixing a compound of the present invention, and a second pharmaceutical agent.
  • One aspect of the present invention pertains to compositions obtained by any of the methods described herein.
  • One aspect of the present invention pertains to a pharmaceutical product selected from: a pharmaceutical composition, a formulation, a unit dosage form, a combined preparation, a twin pack, and a kit; each comprising a compound of the present invention, and a second pharmaceutical agent.
  • One aspect of the present invention pertains to pharmaceutical compositions comprising a compound of the present invention, a second pharmaceutical agent, and a pharmaceutically acceptable carrier.
  • One aspect of the present invention pertains to methods for preparing a pharmaceutical composition
  • methods for preparing a pharmaceutical composition comprising the step of admixing a compound of the present invention, a second pharmaceutical agent, and a pharmaceutically acceptable carrier.
  • One aspect of the present invention pertains to methods for modulating the activity of a GPRl 19 receptor, comprising administering to an individual in need thereof, a therapeutically effective amount of: a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention.
  • One aspect of the present invention pertains to methods for modulating the activity of a GPRl 19 receptor, comprising prescribing to an individual in need thereof, a therapeutically effective amount of: a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention.
  • One aspect of the present invention pertains to the use of a compound of the present invention in the manufacture of a medicament for modulating the activity of a GPRl 19 receptor in an individual.
  • One aspect of the present invention pertains to a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention; for use in a method of treatment of the human or animal body by therapy.
  • One aspect of the present invention pertains to a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention; for use in a method of modulating the activity of a GPR119 receptor in an individual.
  • One aspect of the present invention pertains to methods for modulating the activity of a GPR119 receptor, comprising administering to an individual in need thereof, a therapeutically effective amount of a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention; each in combination with a therapeutically effective amount of a second pharmaceutical agent.
  • One aspect of the present invention pertains to methods for modulating the activity of a GPR119 receptor, comprising prescribing to an individual in need thereof, a therapeutically effective amount of a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention; each in combination with a therapeutically effective amount of a second pharmaceutical agent.
  • One aspect of the present invention pertains to the use of a compound of the present invention in combination with a second pharmaceutical agent in the manufacture of a medicament for modulating the activity of a GPR119 receptor in an individual.
  • One aspect of the present invention pertains to the use of a pharmaceutical agent in combination with a compound of the present invention, in the manufacture of a medicament for modulating the activity of a GPR119 receptor in an individual.
  • One aspect of the present invention pertains to a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention; each in combination with a second pharmaceutical agent for use in a method of treatment of the human or animal body by therapy.
  • One aspect of the present invention pertains to a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention; each in combination with a second pharmaceutical agent for use in a method of modulating the activity of a GPR119 receptor in an individual.
  • One aspect of the present invention pertains to a pharmaceutical agent in combination with a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention; for use in a method of treatment of the human or animal body by therapy.
  • One aspect of the present invention pertains to a pharmaceutical agent in combination with a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention; for use in modulating the activity of a GPRl 19 receptor in an individual.
  • the pharmaceutical agent or the second pharmaceutical agent is selected from: a DPP-IV inhibitor, a biguanide, an alpha-glucosidase inhibitor, an insulin analogue, a sulfonylurea, an SGLT2 inhibitor, a meglitinide, a thiazolidinedione, an antidiabetic peptide analogue, and a DGAT-1 inhibitor.
  • modulating the activity of a GPRl 19 receptor is agonizing a GPRl 19 receptor.
  • agonizing a GPRl 19 receptor is selected from:
  • Figure 1 shows the in vivo effects of Compound 7 on glucose homeostasis in male
  • Figure 2 shows the in vivo effects of Compound 10 on glucose homeostasis in male
  • Figure 3 shows the in vivo effects of Compound 7 and Compound 10 on glucose excursion in terms of percent glycemic inhibition (i.e., percent AUC reduction) in male 129SVE mice.
  • Figure 4 shows a general synthetic scheme for the preparation of intermediates useful in preparing compounds of Formula (I).
  • Figure 5 shows a general synthetic scheme for the preparation of compounds of Formula (I) and intermediates of the present invention.
  • Figure 6 shows a general synthetic scheme for the preparation of compounds of Formula (I) wherein R 1 is oxadiazole optionally substituted with one or more substituents selected from: Ci-C6 alkyl, Ci-C6 haloalkyl, and halogen ⁇ i.e., R is selected from H, Ci-C6 alkyl, C 1 -C6 haloalkyl, and halogen).
  • Figure 7 shows a general synthetic scheme for the preparation of compounds of Formula (I) wherein R 1 is Ci-C6 alkoxycarbonyl, C 3 -C 7 cycloalkoxycarbonyl, or heterocyclyloxycarbonyl each optionally substituted with one or more substituents selected from: C 1 -C6 alkyl, C 1 -C6 haloalkyl, and halogen (i.e., R b is C 1 -C6 alkyl, C 3 -C 7 cycloalkyl, or heterocyclyl each optionally substituted with one or more substituents selected from: C 1 -C6 alkyl, C 1 -C6 haloalkyl, and halogen); and wherein R 1 is heteroaryl optionally substituted with one or more substituents selected from: C 1 -C6 alkyl, C 1 -C6 haloalkyl, and halogen.
  • R 1 is Ci-C6 alkoxycarbonyl, C 3 -C
  • agonist refers to a moiety that interacts with and activates a G-protein-coupled receptor, for instance a GPR119-receptor, and can thereby initiate a physiological or pharmacological response characteristic of that receptor.
  • a G-protein-coupled receptor for instance a GPR119-receptor
  • an agonist may activate an intracellular response upon binding to a receptor, or enhance GTP binding to a membrane.
  • antagonist refers to a moiety that competitively binds to the receptor at the same site as an agonist (for example, the endogenous ligand), but which does not activate the intracellular response initiated by the active form of the receptor and can thereby inhibit the intracellular responses by an agonist or partial agonist.
  • An antagonist does not diminish the baseline intracellular response in the absence of an agonist or partial agonist.
  • GPR119 includes the human amino acid sequences found in GenBank® accession number AY288416, and naturally-occurring allelic variants thereof, and mammalian orthologs thereof.
  • a human GPR119 for use in screening and testing of the compounds of the invention is provided in the nucleotide sequence of Seq. ID.No: l and the corresponding amino acid sequence in Seq. ID.No:2 found in PCT Application No.
  • hydrate refers to 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.
  • solvate refers to 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.
  • in need of treatment and the term “in need thereof” when referring to treatment are used interchangeably and refer to 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
  • mice refers to any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates, and most preferably humans.
  • inverse agonist refers to a moiety that binds to the endogenous form of the receptor or to the constitutively activated form of the receptor and which inhibits the baseline intracellular response initiated by the active form of the receptor below the normal base level of activity which is observed in the absence of an agonist or partial agonist, or decreases GTP binding to a membrane.
  • the baseline intracellular response is inhibited in the presence of the inverse agonist by at least 30%, more preferably by at least 50% and most preferably by at least 75%, as compared with the baseline response in the absence of the inverse agonist.
  • modulate or modulating refers to an increase or decrease in the amount, quality, response or effect of a particular activity, function or molecule.
  • composition refers to 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
  • 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.
  • terapéuticaally effective amount refers to 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 by 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
  • 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).
  • unit dosage form refers to a single dose form which is capable of being administered to a subject, and which can be readily handled and packaged, remaining as a physically and chemically stable unit comprising a compound of the present invention (i.e., compound used neat, such as in a dry powder inhaler (DPI) consisting of neat drug substance or for use in sublingual or buccal administration) or a pharmaceutically acceptable composition comprising a compound of the present invention such as in a pill, capsule, tablet, and the like.
  • DPI dry powder inhaler
  • C 1 -C6 alkoxy refers to a radical comprising a C 1 -C6 alkyl group attached directly to an oxygen atom, wherein Ci-C 6 alkyl has the same definition as found herein. Some embodiments contain 1 to 5 carbons. Some embodiments contain 1 to 4 carbons. Some embodiments contain 1 to 3 carbons. Some embodiments contain one or two carbons. Examples of an alkoxy group include, but are not limited to methoxy, ethoxy, w-propoxy, isopropoxy, n- butoxy, i-butoxy, isobutoxy, s-butoxy, and the like.
  • Ci-C6 alkoxycarbonyl refers to a radical consisting of a Ci-C6 alkoxy group attached to a carbonyl group, wherein Ci-C6 alkoxy has the same definition as found herein. Examples include, but are not limited to, methoxycarbonyl, ethoxycarbonyl, propoxycarbonyl, isopropoxycarbonyl, butoxycarbonyl, butoxycarbonyl, isobutoxycarbonyl, and tert- butoxycarbonyl.
  • Ci-C6 alkyl refers to a straight or branched carbon radical containing 1 to 6 carbons. Some embodiments contain 1 to 5 carbons. Some embodiments contain 1 to 4 carbons. Some embodiments contain 1 to 3 carbons. Some embodiments contain one or two carbons.
  • alkyl group examples include, but are not limited to, methyl, ethyl, n -propyl, isopropyl, n- butyl, i-butyl, isobutyl, i-butyl, pentyl, isopentyl, i-pentyl, neopentyl, 1-methylbutyl [i.e., -CH(CH 3 )CH 2 CH 2 CH 3 ], 2-methylbutyl [i.e. , -CH 2 CH(CH 3 )CH 2 CH 3 ], n-hexyl, and the like.
  • cyano refers to the group -CN.
  • C 3 -C 7 cycloalkoxycarbonyl refers to a radical consisting of a C 3 -C 7 cycloalkoxy group attached to a carbonyl group, wherein C 3 -C 7 cycloalkoxy has the same definition as found herein. Examples include, but are not limited to, cyclopropoxycarbonyl, cyclobutoxycarbonyl, cyclopentyloxycarbonyl, cyclohexyloxycarbonyl, and
  • C 3 -C 7 cycloalkoxy refers to a radical comprising a C 3 -C 7 cycloalkyl group attached directly to an oxygen atom, wherein C 3 -C 7 cycloalkyl has the same definition as found herein. Some embodiments contain 3 to 6 carbons. Some embodiments contain 3 to 5 carbons. Some embodiments contain 3 to 4 carbons. Examples of a cycloalkoxy group include, but are not limited to cyclopropoxy, cyclobutoxy, cyclopentyloxy, cyclohexyloxy, and cycloheptyloxy.
  • C 3 -C 7 cycloalkyl refers to a saturated ring radical containing 3 to 7 ring carbons. Some embodiments contain 3 to 4 carbons. Some embodiments contain 3 to 5 carbons. Some embodiments contain 4 to 6 carbons. Some embodiments contain 5 to 6 carbons.
  • Examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cycloheptyl.
  • Ci-C6 haloalkyl refers to a radical comprising a Ci-C6 alkyl group substituted with one or more halogens, wherein Ci-C6 alkyl has the same definition as found herein.
  • the Ci-C6 haloalkyl may be fully substituted in which case it can be represented by the formula C q L 2q+ i, wherein L is a halogen and "q" is 1, 2, 3, 4, 5 or 6. When more than one halogen is present then they may be the same or different and selected from: fluorine, chlorine, bromine, and iodine.
  • haloalkyl contains 1 to 5 carbons. In some embodiments, haloalkyl contains 1 to 4 carbons.
  • haloalkyl contains 1 to 3 carbons. In some embodiments, haloalkyl contains one or two carbons. Examples of a haloalkyl group include, but are not limited to, fluoromethyl, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, pentafluoroethyl, 2-fluoropropan-2-yl, 1,1-difluoropropyl, 1,3- difluoropropan-2-yl, (S)- l-fluoropropan-2-yl, (R)-l-fluoropropan-2-yl, l, l, l-trifluoropropan-2- yl, l, l, l,3,3,3-hexafluoropropan-2-yl, and the like.
  • halogen refers to a fluoro, chloro, bromo or iodo group.
  • heteroaryl refers to a ring system containing 5 to 10 ring atoms, that may contain a single ring or two fused rings, and wherein at least one ring is aromatic and at least one ring atom of the aromatic ring is a heteroatom selected from, for example: O, S and N, wherein N is optionally substituted with H, C 1 -C4 acyl, C 1 -C4 alkyl, or O (i.e., forming an N- oxide) and S is optionally substituted with one or two oxygens.
  • the aromatic ring contains one heteroatom.
  • the aromatic ring contains two heteroatoms.
  • the aromatic ring contains three heteroatoms.
  • Examples include furanyl, thienyl, pyrrolyl, imidazolyl, oxazolyl, thiazolyl, isoxazolyl, pyrazolyl, isothiazolyl, oxadiazolyl, triazolyl, tetrazolyl, thiadiazolyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, quinolinyl, isoquinolinyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, indolyl, isoindolyl, indazolyl, indolizinyl, purinyl, naphthyridinyl, pteridinyl, carbazolyl, acridinyl.
  • Some embodiments are directed to 5-membered heteroaryl rings.
  • Examples of a 5-membered heteroaryl ring include furanyl, thienyl, pyrrolyl, imidazolyl, oxazolyl, thiazolyl, isoxazolyl, pyrazolyl, isothiazolyl, oxadiazolyl, triazolyl, tetrazolyl, thiadiazolyl, and the like.
  • Some embodiments are directed to 6-membered heteroaryl rings. Examples of a 6-membered heteroaryl ring include pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, and the like.
  • Some embodiments are directed to 8 to 10-membered heteroaryl rings.
  • Examples of a 8 to 10-membered heteroaryl ring include quinolinyl, isoquinolinyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, indolyl, isoindolyl, indazolyl, indolizinyl, purinyl, naphthyridinyl, pteridinyl, carbazolyl, acridinyl. phenazinyl,
  • heterocyclyl refers to a non-aromatic ring radical containing 3 to 10 ring atoms, wherein one, two or three ring atoms are heteroatoms selected independently from, for example: O, S, and N, wherein N is optionally substituted with H, C1-C4 acyl or C1-C4 alkyl; and S is optionally substituted with one or two oxygens.
  • heterocyclyl group examples include, but are not limited to, aziridinyl, azetidinyl, piperidinyl, morpholinyl, piperazinyl, pyrrolidinyl, [l,3]-dioxolanyl, thiomorpholinyl, [l,4]oxazepanyl, 1, 1-dioxothiomorpholinyl, azepanyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydrothiopyranyl, l-oxo-hexahydro- ⁇ 4 -thiopyranyl, l, l-dioxo-hexahydro- ⁇ 6 -thiopyranyl, oxetanyl, and azabicyclo[3.2.1]octanyl.
  • heterocyclyl refers to piperidin-4-yl, 3-azabicyclo[3.2.1]octan-8-yl, and 8- azabicyclo[3.2.1] octan- 3 - y 1.
  • heterocyclyloxy refers to a radical comprising a heterocyclyl group attached to an oxygen radical, wherein heterocyclyl has the same definition as described herein.
  • heterocyclyloxycarbonyl refers to a radical comprising a heterocyclyloxy group attached to a carbonyl group, wherein heterocyclyloxy has the same definition as described herein.
  • One aspect of the present invention provides, inter alia, compounds selected from compounds of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • X and R 1 have the same definitions as described herein, supra and infra.
  • One aspect of the present invention provides, inter alia, compounds selected from compounds of Formula (lb) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • One aspect of the present invention provides, inter alia, compounds selected from compounds of Formula (Id) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • One aspect of the present invention provides, inter alia, compounds selected from compounds of Formulae (I), (lb), and (Id), and pharmaceutically acceptable salts, solvates, and hydrates thereof; wherein X and R 1 have the same definitions as described herein, supra and infra.
  • some embodiments of the present invention include every combination of one or more embodiments pertaining to the chemical groups represented by the variables and generic chemical formulae as described herein or every combination of one or more compounds of Formula (I), and related Formulae, together/in combination with every combination of one or more pharmaceutical agents, such as an inhibitor of DPP-IV, a biguanide, an alpha-glucosidase inhibitor, a DGAT- 1 inhibitor, and the like, either specifically disclosed herein or specifically disclosed in any reference recited herein just as if each and every combination was individually and explicitly recited.
  • pharmaceutical agents such as an inhibitor of DPP-IV, a biguanide, an alpha-glucosidase inhibitor, a DGAT- 1 inhibitor, and the like, either specifically disclosed herein or specifically disclosed in any reference recited herein just as if each and every combination was individually and explicitly recited.
  • substituted indicates that at least one hydrogen atom of the chemical group is replaced by a non-hydrogen substituent or group, the non-hydrogen substituent or group can be monovalent or divalent. When the substituent or group is divalent, then it is understood that this group is further substituted with another substituent or group.
  • a chemical group herein when a chemical group herein is "substituted" it may have up to the full valance of substitution; for example, a methyl group can be substituted by 1, 2, or 3 substituents, a methylene group can be substituted by one or two substituents, a phenyl group can be substituted by 1, 2, 3, 4, or 5 substituents, a naphthyl group can be substituted by 1, 2, 3, 4, 5, 6, or 7 substituents, and the like.
  • substituted with one or more substituents refers to the substitution of a group with one substituent up to the total number of substituents physically allowed by the group. Further, when a group is substituted with more than one group they can be identical or they can be different.
  • Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution. It is understood that the various tautomeric forms are within the scope of the compounds of the present invention.
  • trans meso isomers of compounds of the present invention are named herein using the designation (lr,4r) as shown below:
  • X is N or CH.
  • X is N.
  • X is CH.
  • R 1 is selected from: Ci-C 6 alkoxycarbonyl, C 3 -C 7
  • cycloalkoxycarbonyl, heterocyclyloxycarbonyl, and heteroaryl each optionally substituted with one or more substituents selected from: C 1 -C6 alkyl, C 1 -C6 haloalkyl, and halogen; provided that R 1 is a group other than 4-(trifluoromethyl -2-yl:
  • R 1 is selected from: C 1 -C6 alkoxycarbonyl, C3-C7
  • cycloalkoxycarbonyl cycloalkoxycarbonyl, heterocyclyloxycarbonyl, and heteroaryl; each optionally substituted with one or more substituents selected from: chloro, fluoro, 2-fluoropropan-2-yl, isopropyl, methyl, 2,2,2-trifluoroethyl, and trifluoromethyl.
  • R 1 is selected from: ieri-butoxycarbonyl, isopropoxycarbonyl, cyclobutoxycarbonyl, cyclopentyloxycarbonyl, cyclopropoxycarbonyl, (pyrrolidin-3- yloxy)carbonyl, 1,2,4-oxadiazolyl, (oxetan-3-yloxy)carbonyl, and pyrimidin-2-yl; each optionally substituted with one or more substituents selected from: Ci-C 6 alkyl, Ci-C 6 haloalkyl, and halogen.
  • R 1 is selected from: ieri-butoxycarbonyl, isopropoxycarbonyl, cyclobutoxycarbonyl, cyclopentyloxycarbonyl, cyclopropoxycarbonyl, (pyrrolidin-3- yloxy)carbonyl, 1,2,4-oxadiazolyl, (oxetan-3-yloxy)carbonyl, and pyrimidin-2-yl; each optionally substituted with one or more substituents selected from: chloro, fluoro, 2- fluoropropan-2-yl, isopropyl, methyl, 2,2,2-trifluoroethyl, and trifluoromethyl.
  • R 1 is selected from: C 1 -C6 alkoxycarbonyl optionally substituted with one or more halogen substituents.
  • R 1 is selected from: C 1 -C6 alkoxycarbonyl optionally substituted with one or more fluoro substituents. In some embodiments, R 1 is selected from: ieri-butoxycarbonyl and
  • isopropoxycarbonyl each optionally substituted with one or more halogen substituents.
  • R 1 is selected from: ieri-butoxycarbonyl and
  • isopropoxycarbonyl each optionally substituted with one or more fluoro substituents.
  • R 1 is selected from: C 3 -C 7 cycloalkoxycarbonyl optionally substituted with one or more substituents selected from: C 1 -C6 alkyl and C 1 -C6 haloalkyl.
  • R 1 is selected from: C 3 -C 7 cycloalkoxycarbonyl optionally substituted with one or more substituents selected from: methyl and trifluoromethyl.
  • R 1 is selected from: cyclobutoxycarbonyl
  • cyclopentyloxycarbonyl, and cyclopropoxycarbonyl each optionally substituted with one or more substituents selected from: C 1 -C6 alkyl and C 1 -C6 haloalkyl.
  • R 1 is selected from cyclobutoxycarbonyl
  • cyclopentyloxycarbonyl and cyclopropoxycarbonyl; each optionally substituted with one or more substituents selected from: methyl and trifluoromethyl.
  • R 1 is selected from: heterocyclyloxycarbonyl optionally substituted with one or more C 1 -C6 haloalkyl substituents.
  • R 1 is selected from: heterocyclyloxycarbonyl optionally substituted with one or more substituents selected from: 2,2,2-trifluoroethyl and trifluoromethyl.
  • R 1 is selected from: (pyrrolidin-3-yloxy)carbonyl and (oxetan-3- yloxy)carbonyl; each optionally substituted with one or more C 1 -C6 haloalkyl substituents.
  • R 1 is selected from: (pyrrolidin-3-yloxy)carbonyl and (oxetan-3- yloxy)carbonyl; each optionally substituted with one or more substituents selected from: 2,2,2- trifluoroethyl and trifluoromethyl.
  • R 1 is selected from: heteroaryl optionally substituted with one or more substituents selected from: C 1 -C6 alkyl, C 1 -C6 haloalkyl, and halogen.
  • R 1 is selected from: heteroaryl optionally substituted with one or more substituents selected from: chloro, 2-fluoropropan-2-yl, isopropyl, and trifluoromethyl.
  • R 1 is selected from: 1,2,4-oxadiazolyl, and pyrimidin-2-yl; each optionally substituted with one or more substituents selected from: C 1 -C 6 alkyl, C 1 -C6 haloalkyl, and halogen.
  • R 1 is selected from: 1,2,4-oxadiazolyl, and pyrimidin-2-yl; each optionally substituted with one or more substituents selected from: chloro, 2-fluoropropan-2-yl, isopropyl, and trifluoromethyl.
  • R 1 is selected from: (l-methylcyclopropoxy)carbonyl, 5- chloropyrimidin-2-yl, (1,1,1 -trifluoropropan-2-yloxy)carbonyl, ( 1 - (2,2,2- trifluoroethyl)pyrrolidin-3-yloxy)carbonyl, 3-(trifluoromethyl)- 1 ,2,4-oxadiazol-5-yl, 5- (trifluoromethyl)- 1 ,2,4-oxadiazol-3-yl, ( 1 -(trifluoromethyl)cyclobutoxy)carbonyl, (3- (trifluoromethyl)oxetan-3-yloxy)carbonyl, (1,1,1 -trifluoro-2-methylpropan-2-yloxy)carbonyl, (l,l,l,3,3,3-hexafluoropropan-2-yloxy)carbonyl, (l-(trifluoromethyl)cyclopentyloxy)carbonyl,
  • R 1 is (l-methylcyclopropoxy)carbonyl. In some embodiments, R 1 is 5-chloropyrimidin-2-yl. In some embodiments, R 1 is ( 1,1,1 -trifluoropropan-2- yloxy)carbonyl. In some embodiments, R 1 is (l-(2,2,2-trifluoroethyl)pyrrolidin-3- yloxy)carbonyl. In some embodiments, R 1 is 3-(trifluoromethyl)-l,2,4-oxadiazol-5-yl. In some embodiments, R 1 is 5-(trifluoromethyl)-l,2,4-oxadiazol-3-yl.
  • R 1 is (1- (trifluoromethyl)cyclobutoxy)carbonyl. In some embodiments, R 1 is (3-(trifluoromethyl)oxetan- 3-yloxy)carbonyl. In some embodiments, R 1 is ( 1,1,1 -trifluoro-2-methylpropan-2- yloxy)carbonyl. In some embodiments, R 1 is (l,l,l,3,3,3-hexafluoropropan-2-yloxy)carbonyl. In some embodiments, R 1 is (l-(trifluoromethyl)cyclopentyloxy)carbonyl.
  • R 1 is 5-(2-fluoropropan-2-yl)-l,2,4-oxadiazol-3-yl. In some embodiments, R 1 is 3-(2- fluoropropan-2-yl)-l,2,4-oxadiazol-5-yl. In some embodiments, R 1 is 3-isopropyl- 1,2,4- oxadiazol-5-yl.
  • One aspect of the present invention encompasses certain cyclohexyl derivatives selected from compounds of Formula (lb) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • R 1 is selected from: Ci-C6 alkoxycarbonyl, C 3 -C 7 cycloalkoxycarbonyl,
  • heterocyclyloxycarbonyl, and heteroaryl each optionally substituted with one or more substituents selected from: chloro, fluoro, 2-fluoropropan-2-yl, methyl, 2,2,2-trifluoroethyl, and trifluoromethyl.
  • One aspect of the present invention encompasses certain cyclohexyl derivatives selected from compounds of Formula (lb) and pharmaceutically acceptable salts, solvates, and hydrates thereof, wherein:
  • R 1 is selected from: ieri-butoxycarbonyl, isopropoxycarbonyl, cyclobutoxycarbonyl, cyclopentyloxycarbonyl, cyclopropoxycarbonyl, (pyrrolidin-3-yloxy)carbonyl, 1,2,4- oxadiazolyl, (oxetan-3-yloxy)carbonyl, and pyrimidin-2-yl; each optionally substituted with one or more substituents selected from: Ci-C6 alkyl, Ci-C6 haloalkyl, and halogen.
  • One aspect of the present invention encompasses certain cyclohexyl derivatives selected from compounds of Formula (lb) and pharmaceutically acceptable salts, solvates, and hydrates thereof, wherein:
  • R 1 is selected from: ieri-butoxycarbonyl, isopropoxycarbonyl, cyclobutoxycarbonyl, cyclopentyloxycarbonyl, cyclopropoxycarbonyl, (pyrrolidin-3-yloxy)carbonyl, 1,2,4- oxadiazolyl, (oxetan-3-yloxy)carbonyl, and pyrimidin-2-yl; each optionally substituted with one or more substituents selected from: chloro, fluoro, 2-fluoropropan-2-yl, methyl, 2,2,2- trifluoroethyl, and trifluoromethyl.
  • One aspect of the present invention encompasses certain cyclohexyl derivatives selected from compounds of Formula (lb) and pharmaceutically acceptable salts, solvates, and hydrates thereof, wherein:
  • R 1 is selected from: (l-methylcyclopropoxy)carbonyl, 5-chloropyrimidin-2-yl, (1,1,1- trifluoropropan-2-yloxy)carbonyl, (l-(2,2,2-trifluoroethyl)pyrrolidin-3-yloxy)carbonyl, 3- (trifluoromethyl)-l,2,4-oxadiazol-5-yl, 5-(trifluoromethyl)-l,2,4-oxadiazol-3-yl, (1- (trifluoromethyl)cyclobutoxy)carbonyl, (3-(trifluoromethyl)oxetan-3-yloxy)carbonyl, (1,1,1- trifluoro-2-methylpropan-2-yloxy)carbonyl, (l,l,l,3,3,3-hexafluoropropan-2-yloxy)carbonyl, (l-(trifluoromethyl)cyclopentyloxy)carbonyl, 5-(2-fluoropropan-2-
  • One aspect of the present invention encompasses certain cyclohexyl derivatives selected from compounds of Formula (Id) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • R 1 is selected from: Ci-C6 alkoxycarbonyl, C 3 -C 7 cycloalkoxycarbonyl, and heteroaryl; each optionally substituted with one or more substituents selected from: chloro, fluoro, 2- fluoropropan-2-yl, isopropyl, and methyl.
  • One aspect of the present invention encompasses certain cyclohexyl derivatives selected from compounds of Formula (Id) and pharmaceutically acceptable salts, solvates, and hydrates thereof, wherein:
  • R 1 is selected from: isopropoxycarbonyl, cyclopropoxycarbonyl, 1,2,4-oxadiazolyl, and pyrimidin-2-yl; each optionally substituted with one or more substituents selected from: Ci-C6 alkyl, Ci-C6 haloalkyl, and halogen.
  • One aspect of the present invention encompasses certain cyclohexyl derivatives selected from compounds of Formula (Id) and pharmaceutically acceptable salts, solvates, and hydrates thereof, wherein:
  • R 1 is selected from: isopropoxycarbonyl, cyclopropoxycarbonyl, 1,2,4-oxadiazolyl, and pyrimidin-2-yl; each optionally substituted with one or more substituents selected from: chloro, fluoro, 2-fluoropropan-2-yl, isopropyl, and methyl.
  • One aspect of the present invention encompasses certain cyclohexyl derivatives selected from compounds of Formula (Id) and pharmaceutically acceptable salts, solvates, and hydrates thereof, wherein:
  • R 1 is selected from: (l-methylcyclopropoxy)carbonyl, 5-chloropyrimidin-2-yl, (1,1, 1- trifluoropropan-2-yloxy)carbonyl, 5-(2-fluoropropan-2-yl)-l,2,4-oxadiazol-3-yl, 3-(2- fluoropropan-2-yl)-l,2,4-oxadiazol-5-yl, and 3-isopropyl-l,2,4-oxadiazol-5-yl.
  • Some embodiments of the present invention include every combination of one or more compounds selected from the following group shown in Table A.
  • individual compounds and chemical genera of the present invention for example those compounds found in Table A including, isomers, diastereoisomers and enantiomers thereof, encompass all pharmaceutically acceptable salts, solvates, and hydrates, thereof.
  • mesoisomers of individual compounds and chemical genera of the present invention for example those compounds found in Table A, encompass all pharmaceutically acceptable salts, solvates and particularly hydrates, thereof.
  • the compounds of the Formula (I) and genera related thereto may be prepared according to relevant published literature procedures that are used by one skilled in the art. Exemplary reagents and procedures for these reactions appear hereinafter in the working Examples. Protection and deprotection may be carried out by procedures generally known in the art (see, for example, Greene, T. W. and Wuts, P. G. M., Protecting Groups in Organic Synthesis, 3 rd Edition, 1999 [Wiley]).
  • the present invention embraces, each isomer, each diastereoisomer, each enantiomer and mixtures thereof of each compound and generic formulae disclosed herein just as if they were each individually disclosed with the specific stereochemical designation for each chiral carbon. Separation of the individual isomers and enantiomers (such as, by chiral HPLC, recrystallization of diastereoisomeric mixtures and the like) or selective synthesis (such as, by enantiomeric selective syntheses and the like) of the individual isomers can be accomplished by the application of various methods which are well known to practitioners in the art.
  • compositions Compositions, Methods, Indications, Pharmaceutical Products, Combinations, and Uses of Compounds of the Present Invention.
  • compositions comprising a compound of the present invention.
  • One aspect of the present invention pertains to pharmaceutical products selected from: a pharmaceutical composition, a formulation, a unit dosage form, and a kit; each comprising a compound of the present invention.
  • One aspect of the present invention pertains to pharmaceutical compositions comprising a compound of the present invention, and a pharmaceutically acceptable carrier.
  • One aspect of the present invention pertains to methods for preparing a pharmaceutical composition comprising the step of admixing a compound of the present invention, and a pharmaceutically acceptable carrier; some embodiments pertain to pharmaceutical compositions obtained by any of the methods described herein.
  • One aspect of the present invention pertains to compositions comprising a compound of the present invention, and a second pharmaceutical agent.
  • a pharmaceutical agent and “a second pharmaceutical agent”
  • these terms in some aspects be further limited to a pharmaceutical agent that is not a compound of Formula (I) or a compounds related thereto.
  • a pharmaceutical agent and “a second pharmaceutical agent” may refer to a pharmaceutical agent that is not detectable or has an EC 50 that is greater than a value selected from: 50 ⁇ , 10 ⁇ , 1 ⁇ , and 0.1 ⁇ in a GPRl 19 receptor activity assay as described in Example 4.
  • One aspect of the present invention pertains to methods for preparing a composition comprising the step of admixing a compound of the present invention, and a second pharmaceutical agent; some embodiments pertain to compositions obtained by any of the methods described herein.
  • One aspect of the present invention pertains to pharmaceutical products selected from: a pharmaceutical composition, a formulation, a unit dosage form, a combined preparation, a twin pack, and a kit; each comprising a compound of the present invention, and a second pharmaceutical agent.
  • One aspect of the present invention pertains to pharmaceutical compositions comprising a compound of the present invention, a second pharmaceutical agent, and a pharmaceutically acceptable carrier.
  • One aspect of the present invention pertains to methods for preparing a pharmaceutical composition
  • methods for preparing a pharmaceutical composition comprising the step of admixing a compound of the present invention, a second pharmaceutical agent, and a pharmaceutically acceptable carrier; some embodiments pertain to pharmaceutical compositions obtained by any of the methods described herein.
  • One aspect of the present invention pertains to methods selected from one or more of the following: 1) for modulating the activity of a GPRl 19 receptor; 2) for agonizing a GPRl 19 receptor; 3) for increasing the secretion of an incretin in an individual; 4) increasing a blood incretin level in an individual; and 5) for treating/treatment of a disorder selected from: a GPR119-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic-related disorder; type 2 diabetes; and obesity; in an individual; comprising: A) administering to an individual in need thereof or B) prescribing to an individual in need thereof, a therapeutically effective amount of: a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention; each optionally in combination with a therapeutically effective amount of a second pharmaceutical agent.
  • Some embodiments pertain to methods comprising administering to an individual in need thereof a therapeutically effective amount of: a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention. Some embodiments pertain to methods for prescribing to an individual in need thereof, a therapeutically effective amount of: a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention.
  • One aspect of the present invention pertains to the use of a compound of the present invention, optionally in combination with a second pharmaceutical agent, in the manufacture of a medicament, selected from one or more of the following: 1) for modulating the activity of a GPR119 receptor in an individual; 2) for agonizing a GPR119 receptor; 3) for increasing the secretion of an incretin in an individual; 4) increasing a blood incretin level in an individual; and 5) for treating/treatment of a disorder selected from: a GPR119-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic- related disorder; type 2 diabetes; and obesity.
  • One aspect of the present invention pertains to a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention; optionally in combination with a second pharmaceutical agent, for use in a method of treatment of the human or animal body by therapy.
  • One aspect of the present invention pertains to one or more of the following: methods of the present invention, compounds of the present invention; compositions of the present invention; pharmaceutical products of the present invention; and pharmaceutical compositions of the present invention; optionally in combination with a second pharmaceutical agent, for use in a method selected from one or more of the following: 1) for modulating the activity of a GPR119 receptor; 2) for agonizing a GPR119 receptor; 3) for increasing the secretion of an incretin in an individual; 4) increasing a blood incretin level in an individual; and 5) for treating/ treatment of a disorder selected from: a GPR119-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic- related disorder; type 2 diabetes; and obesity.
  • a second pharmaceutical agent for use in a method selected from one or more of the following: 1) for modulating the activity of a GPR119 receptor; 2)
  • One aspect of the present invention pertains to a pharmaceutical agent in combination with a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention; for use in a method of treatment of the human or animal body by therapy.
  • One aspect of the present invention pertains to a pharmaceutical agent in combination with a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention; for use in a method selected from one or more of the following: 1) for modulating the activity of a GPR119 receptor; 2) for agonizing a GPR119 receptor; 3) for increasing the secretion of an incretin in an individual; 4) increasing a blood incretin level in an individual; and 5) for treating/treatment of a disorder selected from: a GPR119-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic- related disorder; type 2 diabetes; and obesity.
  • Some embodiments pertain to methods, uses, compounds, and pharmaceutical agents, each as described herein, for modulating the activity of a GPR119 receptor. Some embodiments pertain to methods, uses, compounds, and pharmaceutical agents, each as described herein, for agonizing a GPR119 receptor. Some embodiments pertain to methods, uses, compounds, and pharmaceutical agents, each as described herein, for increasing the secretion of an incretin in an individual. Some embodiments pertain to methods, uses, compounds, and pharmaceutical agents, each as described herein, for increasing a blood incretin level in an individual.
  • Some embodiments pertain to methods, uses, compounds, and pharmaceutical agents, each as described herein, for treatment of a disorder selected from: a GPR119-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic-related disorder; and obesity; in an individual.
  • a disorder selected from: a GPR119-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic-related disorder; and obesity; in an individual.
  • the disorder is a GPR119-related disorder. In some embodiments, the disorder is a GPR119-related disorder.
  • the disorder is a condition ameliorated by increasing secretion of an incretin. In some embodiments, the disorder is a condition ameliorated by increasing a blood incretin level. In some embodiments, the disorder is a condition characterized by low bone mass. In some embodiments, the disorder is a neurological disorder. In some embodiments, the disorder is a metabolic-related disorder. In some embodiments, the disorder is obesity. In some
  • the disorder is type 2 diabetes. In some embodiments, the disorder is hyperglycemia. In some embodiments, the disorder is hyperlipidemia. In some embodiments, the disorder is hypertriglyceridemia. In some embodiments, the disorder is type 1 diabetes. In some embodiments, the disorder is dyslipidemia. In some embodiments, the disorder is syndrome X.
  • the pharmaceutical product comprises a pharmaceutical composition. In some embodiments, the pharmaceutical product comprises a formulation. In some embodiments, the pharmaceutical product comprises a unit dosage form. In some embodiments, the pharmaceutical product comprises a kit. In some embodiments, the pharmaceutical product comprises a combined preparation. In some embodiments, the pharmaceutical product comprises a twin pack.
  • the compound and pharmaceutical agent or the second pharmaceutical agent are administered simultaneously, separately, or sequentially. In some embodiments, the compound and the pharmaceutical agent or the second pharmaceutical agent are administered simultaneously. In some embodiments, the compound and the pharmaceutical agent or the second pharmaceutical agent are administered separately. In some embodiments, the compound and the pharmaceutical agent or the second pharmaceutical agent are administered sequentially.
  • the incretin is GLP-1. In some embodiments, the incretin is GIP. In some embodiments, the incretin is PYY.
  • a DPP-IV inhibitor a biguanide
  • an alpha-glucosidase inhibitor an insulin analogue
  • a sulfonylurea an SGLT2 inhibitor
  • meglitinide a thiazolidinedione
  • an antidiabetic peptide analogue a DGAT-1 inhibitor
  • One aspect of the present invention pertains to compositions, methods, pharmaceutical products, pharmaceutical compositions, uses; compounds, and pharmaceutical agents, each as described herein, wherein: 1) the compound and the pharmaceutical agent or the second pharmaceutical agent are provided in amounts which give a synergistic effect in treating the disorder; 2) the amount of the compound alone is substantially therapeutically ineffective at treating the disorder; and/or 3) the amount of the pharmaceutical agent alone or the second pharmaceutical agent alone is substantially therapeutically ineffective at treating the disorder.
  • One aspect of the present invention relates to methods for preparing pharmaceutical products of the present invention comprising the steps: mixing said compound with a first pharmaceutically acceptable carrier to prepare a compound unit dosage form; mixing said second pharmaceutical agent with a second pharmaceutically acceptable carrier to prepare a second pharmaceutical agent unit dosage form; and combining said compound unit dosage form and said second pharmaceutical agent unit dosage form in a combined unit dosage form for simultaneous, separate, or sequential use.
  • the first pharmaceutically acceptable carrier is different from the second pharmaceutically acceptable carrier. In some embodiments, the different
  • pharmaceutically acceptable carriers are suitable for administration by the same route.
  • the different pharmaceutically acceptable carriers are suitable for administration by different routes.
  • the first pharmaceutically acceptable carrier is substantially the same as the second pharmaceutically acceptable carrier.
  • the substantially the same pharmaceutically acceptable carriers are suitable for oral administration.
  • a compound as described herein or a pharmaceutical composition thereof can be utilized for modulating the activity of GPR119- receptor and therefore related diseases, conditions and/or disorders related thereto such as those described herein.
  • modulating the activity includes the treatment of a GPRl 19- related disorder.
  • a GPRl 19-related disorder is a condition ameliorated by increasing a blood incretin level.
  • a GPRl 19-related disorder is a condition characterized by low bone mass.
  • a GPRl 19-related disorder is a neurological disorder.
  • a GPRl 19-related disorder is a metabolic-related disorder.
  • a GPRl 19-related disorder is type 2 diabetes.
  • a GPRl 19-related disorder is obesity.
  • Some embodiments of the present invention include every combination of one or more conditions characterized by low bone mass selected from: osteopenia, osteoporosis, rheumatoid arthritis, osteoarthritis, periodontal disease, alveolar bone loss, osteotomy bone loss, childhood idiopathic bone loss, Paget' s disease, bone loss due to metastatic cancer, osteolytic lesions, curvature of the spine, and loss of height.
  • the neurological disorder selected from: stroke and
  • Some embodiments of the present invention include every combination of one or more metabolic-related disorders selected from: type 1 diabetes, type 2 diabetes mellitus, and conditions associated therewith, such as, but not limited to, coronary heart disease, ischemic stroke, restenosis after angioplasty, peripheral vascular disease, intermittent claudication, 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, 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, vascular restenosis, hyperglycemia, hyperinsulinemia, hyperlipidemia, hypertriglyceridemia, insulin resistance, impaired glucose metabolism, erectile dysfunction, skin and connective tissue disorders, foot ulcerations and ulcerative colitis, endothelial dysfunction and impaired vascular compliance.
  • ITT impaired glucose tolerance
  • impaired fasting plasma glucose glucose
  • Some embodiments of the present invention include every combination of one or more metabolic-related disorders selected from: diabetes, 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, 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, motor-neuron disease, traumatic brain injury, spinal cord injury, and obesity.
  • the disorder is type 2 diabetes. In some embodiments, the disorder is hyperglycemia. In some embodiments, the disorder is hyperlipidemia. In some embodiments, the disorder is hypertriglyceridemia. In some embodiments, the disorder is type 1 diabetes. In some embodiments, the disorder is dyslipidemia. In some embodiments, the disorder is syndrome X. In some embodiments, the disorder is obesity.
  • One aspect of the present invention pertains to methods for weight management, comprising administering to an individual in need thereof, a therapeutically effective amount of a compound of the present invention in combination with a therapeutically effective amount of a pharmaceutical agent, such as any agent described herein; wherein the compound and the pharmaceutical agent.
  • the weight management comprises weight loss. In some embodiments, the weight management comprises maintenance of weight loss. In some embodiments, the weight management further comprises a reduced-calorie diet. In some embodiments, the weight management further comprises a program of regular exercise. In some embodiments, the weight management further comprises both a reduced-calorie diet and a program of regular exercise.
  • the individual in need of weight management is a patient with an initial body mass of index > 40 kg/m 2 ; > 39 kg/m 2 ; > 38 kg/m 2 ; > 37 kg/m 2 ; > 36 kg/m 2 ; > 35 kg/m 2 ; > 34 kg/m 2 ; > 33 kg/m 2 ; > 32 kg/m 2 ; > 31 kg/m 2 ; > 30 kg/m 2 ; > 29 kg/m 2 ; > 28 kg/m 2 ; > 27 kg/m 2 ; > 26 kg/m 2 ; > 25 kg/m 2 ; > 24 kg/m 2 ; > 23 kg/m 2 ; > 22 kg/m 2 ; > 21 kg/m 2 ; or > 20 kg/m 2 ; and the patient optionally has at least one or at least two weight related comorbid condition(s).
  • the comorbid condition(s) when present are selected from: hypertension, dyslipidemia, cardiovascular disease, glucose intolerance, and sleep apnea.
  • Formulations may be prepared by any suitable method, typically by uniformly mixing the active compound(s) 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
  • 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. 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 ampule. 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, 20 th Edition, 2000, Lippincott Williams & Wilkins, (Editors: Gennaro et al.)
  • a compound of the invention may, 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.
  • Transdermal patches dispense a drug at a controlled rate by presenting the drug for absorption in an efficient manner with minimal 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.
  • the compounds of the invention may thus be placed into the form of pharmaceutical formulations 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.
  • 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.
  • active ingredient defined in the context of a “pharmaceutical composition” refers to a component of a pharmaceutical composition that provides the primary pharmacological effect, as opposed to an "inactive ingredient” which would generally be recognized as providing no pharmaceutical benefit.
  • the dose when using the compounds of the present invention can vary within wide limits and as is customary and is 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 conducted or on whether further active compounds are administered in addition to the compounds of the present invention.
  • Representative doses of the present invention include, but not limited to, about 0.001 mg to about 5000 mg, about 0.001 mg to about 2500 mg, about 0.001 mg to about 1000 mg, 0.001 mg 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.
  • Multiple doses may be administered during the day, especially when relatively large amounts are deemed to be needed, for example 2, 3, or 4 doses. Depending on the individual and as deemed appropriate from the patient's physician or caregiver it may be necessary to deviate upward or downward from the doses described herein.
  • the amount of active ingredient, or an active salt or derivative 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.
  • a model system typically an animal model
  • these extrapolations may merely be based on the weight of the animal 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.
  • compositions of this invention are selected in accordance with a variety 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 dosage and dosage regimen outside these typical ranges can be tested and, where appropriate, may be used in the methods of this invention.
  • 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.
  • the daily dose can be divided, especially when relatively large amounts are administered as deemed appropriate, into several, for example 2, 3, or 4 part administrations. If appropriate, depending on individual behavior, it may be necessary to deviate upward or downward from the daily dose indicated.
  • the compounds of the present invention can be administrated 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, solvate, or hydrate 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 desire 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 active compound; 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” refers to 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.
  • 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.
  • Formulations 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.
  • parenteral administration e.g. by injection, for example bolus injection or continuous infusion
  • 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.
  • a suitable vehicle e.g. sterile, pyrogen-free water
  • Aqueous formulations 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.
  • 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.
  • Formulations suitable for topical administration in the mouth include lozenges comprising 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 formulations 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 formulation in which the active ingredient is provided in a pressurized pack with a suitable propellant.
  • aerosol formulation 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.
  • the compound In formulations intended for administration to the respiratory tract, including intranasal formulations, 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, formulations 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 is 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.
  • Representative acids include, but are not limited to, acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethenesulfonic, dichloroacetic, formic, fumaric, gluconic, glutamic, hippuric, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic,
  • Certain compounds of the present invention which contain a carboxylic acid functional group may optionally exist as pharmaceutically acceptable salts containing non-toxic, pharmaceutically acceptable metal cations and cations derived from organic bases.
  • Representative metals include, but are not limited to, aluminium, calcium, lithium, magnesium, potassium, sodium, zinc and the like. In some embodiments the pharmaceutically acceptable metal is sodium.
  • Organic bases include, but are not limited to, benzathine (A ⁇ A ⁇ -dibenzylefhane-l ⁇ -diamine), chloroprocaine (2- (diethylamino)ethyl 4-(chloroamino)benzoate), choline, diethanolamine, ethylenediamine, meglumine ((2R,3R,4R,5S)-6-(methylamino)hexane-l,2,3,4,5-pentaol), procaine (2- (diethylamino)ethyl 4-aminobenzoate), and the like.
  • Certain pharmaceutically acceptable salts are listed in Berge, et al., Journal of Pharmaceutical Sciences, 66: 1-19 (1977).
  • the acid addition salts may be obtained as the direct products 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.
  • Pro-drugs refers to compounds that have been modified with specific chemical groups known in the art and when administered into an individual these groups undergo biotransformation to give the parent compound. Pro-drugs can thus be viewed as compounds of the invention containing one or more specialized non-toxic protective groups used in a transient manner to alter or to eliminate a property of the compound. In one general aspect, the "pro-drug” approach is utilized to facilitate oral absorption.
  • T. Higuchi and V. Stella Prodrugs as Novel Delivery Systems Vol. 14 of the A.C.S. Symposium Series; and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987.
  • Some embodiments of the present invention include a method of producing a pharmaceutical composition for "combination-therapy" comprising admixing at least one compound according to any of the compound embodiments disclosed herein, together with at least one known pharmaceutical agent as described herein and a pharmaceutically acceptable carrier.
  • GPR119 receptor modulators are utilized as active ingredients in pharmaceutical compositions, these are not intended for use in humans only, but in non- human mammals as well.
  • active agents such as GPR119 receptor modulators
  • livestock animals e.g., horses, cows, etc.
  • the dosage forms described herein may comprise, as the active component, either a compound described herein or a pharmaceutically acceptable salt or as a pharmaceutically acceptable solvate or hydrate thereof.
  • various hydrates and solvates of the compounds described herein 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 K.J. Guillory, "Generation of Polymorphs, Hydrates, Solvates, and Amorphous Solids," in: Polymorphism in Pharmaceutical Solids, ed. Harry G. England, Vol.
  • one aspect of the present invention pertains to methods of administering hydrates and solvates of compounds described herein and/or their pharmaceutical acceptable salts, that can be isolated and characterized by 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-Infrared spectroscopy
  • powder X-ray diffraction (XRPD) powder X-ray diffraction
  • Karl Fisher titration high resolution X-ray diffraction
  • 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.
  • Crystalline forms can be identified by their unique solid state signature with respect to, for example, differential scanning calorimetry (DSC), powder X-ray diffraction (PXRD), and other solid state methods.
  • DSC differential scanning calorimetry
  • PXRD powder X-ray diffraction
  • thermogravimetric analysis TGA
  • Karl Fischer analysis Karl Fischer analysis
  • DSC DSC thermograms
  • the values relating to DSC thermograms can vary by plus or minus about 4 °C (+ 4 °C).
  • the values relating to DSC thermograms can also vary by plus or minus about 20 joules per gram (+ 20 joules per gram).
  • DSC can be used to observe desolvation events.
  • DSC thermogram values relate to desolvation events, the values are understood to be estimates.
  • Scan rate and pan closure can influence DSC values for desolvation events, which can vary by plus or minus about 25 °C.
  • DSC values for desolvation events are typically recorded using a sample in an aluminum pan with an uncrimped lid and a scan rate of 10 °C/min.
  • the relative intensities of the peaks can vary, depending upon the sample preparation technique, the sample mounting procedure and the particular instrument employed. Moreover, instrument variation and other factors can often affect the 2 lvalues. Therefore, the peak assignments of diffraction patterns can vary by plus or minus 0.2 °2 ⁇ (+ 0.2 °2 ⁇ ).
  • the features reported herein can vary by plus or minus about 5 °C (+ 5 °C).
  • the TGA features can also vary by plus or minus about 2% (+ 2%) weight change due to, for example, sample variation.
  • DMS dynamic moisture sorption
  • a compound of the invention can be administered as the sole active pharmaceutical agent (i.e., mono-therapy), or it can be used in combination with one or more pharmaceutical agents (i.e., combination-therapy), such as pharmaceutical agents, such as, known anti-diabetic agents, either administered together or separately for the treatment of the diseases, conditions, and 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 thereof a therapeutically effective amount of a compound of Formula (I) and pharmaceutically acceptable salts, solvates and hydrates thereof, in combination with one or more pharmaceutical agents, such as anti-diabetic agents, as described herein.
  • a metabolic related disorder including a weight-related disorder, such as obesity
  • the combination can be used by mixing the respective active components, a compound of Formula (I) and a pharmaceutical agent, either together or independently optionally with a physiologically acceptable carrier, excipient, binder, diluent, etc., as described herein, and administering the mixture or mixtures either orally or non- orally as a pharmaceutical composition(s).
  • a compound of Formula (I) is administered as a combination therapy with another active compound the compound of Formula (I) and the pharmaceutical agent can be formulated as separate pharmaceutical compositions given at the same time or at different times; or the compound of Formula (I) and the pharmaceutical agent can be formulated together as a single unit dosage.
  • 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: A r -(piperidin-l-yl)-5-(4-chlorophenyl)-l-(2,4- dichlorophenyl)-4-methyl-l/i-pyrazole-3-carboxamide]; melanin concentrating hormone antagonists; le
  • 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 calorie-controlled diet.
  • combination-therapy of the compounds of the present invention with 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 Formula (I) in combination with at least one pharmaceutical agent selected from the group consisting of: sulfonylureas (for example, tolbutamide (Orinase); acetohexamide (Dymelor); tolazamide (Tolinase); chlorpropamide (Diabinese); glipizide (Glucotrol); glyburide (Diabeta, Micronase, Glynase); glimepiride (Amaryl); gliclazide (Diamicron); and sulfonylureas known in the art); meglitinides (for example, repaglinide (Prandin), nateglinide (Starlix), mitiglinide, and other meglitinides known in the art); biguanides (for example,
  • 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 other angiotensin converting enzyme inhibitors known in the art); angiotensin II receptor antagonists [for example, losartan (and the potassium salt form), and other angiotensin II receptor antagonists known in the art; adip
  • Suitable pharmaceutical agents that can be used in conjunction with compounds of the present invention include, but are not limited to: amylin agonists (for example, pramlintide); insulin secretagogues (for example, GLP-1 agonists, exendin-4, and insulinotropin (Liraglutide (NN2211)); acyl Co A cholesterol acetyltransferase inhibitors (for example, ezetimibe, eflucimibe, and other acyl Co A cholesterol acetyltransferase inhibitors known in the art);
  • amylin agonists for example, pramlintide
  • insulin secretagogues for example, GLP-1 agonists, exendin-4, and insulinotropin (Liraglutide (NN2211)
  • acyl Co A cholesterol acetyltransferase inhibitors for example, ezetimibe, eflucimibe, and other acyl Co A cholesterol acetyltransferase inhibitors known in the art
  • cholesterol absorption inhibitors for example, ezetimibe, pamaqueside and other cholesterol absorption inhibitors known in the art
  • cholesterol ester transfer protein inhibitors for example, Torcetrapib (CP-529,414), Dalcetrapib (JTT-705), CETi-1, and other cholesterol ester transfer protein inhibitors known in the art
  • microsomal triglyceride transfer protein inhibitors for example, implitapide, and other microsomal triglyceride transfer protein inhibitors known in the art
  • cholesterol modulators for example, ibrolipim (NO-1886), and other cholesterol modulators known in the art
  • bile acid modulators for example, GT103-279 and other bile acid modulators known in the art
  • insulin signaling pathway modulators for example, inhibitors of protein tyrosine phosphatases (PTPases); non-small molecule mimetics and inhibitors of glutamine- fructose-6-phosphate amido transferase (GFAT); compounds influencing a dysregulated hepati
  • PDHK dehydrogenase kinase
  • insulin sensitivity enhancers insulin secretion enhancers
  • inhibitors of gastric emptying a 2 -adrenergic antagonists
  • RXR retinoid X receptor
  • DPP-IV dipeptidyl peptidase-4
  • Some aspects of the present invention include compounds of Formula (I) that can be employed in any of the methods, pharmaceutical products, uses, compounds, and
  • the two distinct pharmaceutical agents are selected from any of the pharmaceutical agents, or classes of pharmaceutical agents described herein.
  • the two distinct pharmaceutical agents are selected from: an inhibitor of DPP- IV, a biguanide, an alpha-glucosidase inhibitor, an insulin analogue, a sulfonylurea, a SGLT2 inhibitor, a meglitinide, a thiazolidinedione, an anti-diabetic peptide analogue, and a DGAT-1 inhibitor.
  • the two distinct pharmaceutical agents include every combination selected from pharmaceutical agents of the following group: an inhibitor of DPP- IV, a biguanide, an alpha-glucosidase inhibitor, a sulfonylurea, a SGLT2 inhibitor, and a DGAT-1 inhibitor.
  • Some embodiments of the present invention include every combination of one or more compounds selected from compounds of the following group and pharmaceutically acceptable salts, solvates, and hydrates thereof: an inhibitor of DPP-IV 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; (l l S',3 l S',5 l S')-2-[2( l S')-amino-2-(3-hydroxyadamantan-l-yl)acetyl]-2- azabicyclo[3.1.0]hexane-3-carbonitrile; 2-[6-[3(R)-aminopi
  • a-glucosidase inhibitor selected from: acarbose ((2R3R,4R,5R)-4-((2R3R,4R,55,6R)-5-((2R,3R,45,5.S , ,6R)-3,4-dihydroxy-6-methyl-5- ((15,4R,5 l S',6 l S r )-4,5,6-trihydroxy-3-(hydroxymethyl)cyclohex-2-enylamino)tetrahydro-2/i-pyran-
  • acarbose ((2R3R,4R,5R)-4-((2R3R,4R,55,6R)-5-((2R,3R,45,5.S , ,6R)-3,4-dihydroxy-6-methyl-5- ((15,4R,5 l S',6 l S r )-4,5,6-trihydroxy-3-(hydroxymethyl)cyclohex-2-en
  • glibenclamide also known as glyburide (Diabeta, Micronase, Glynase, 5-chloro-A r -(4-(A r - (cyclohexylcarbamoyl)sulfamoyl)phenethyl)-2-methoxybenzamide); glimepiride (Amaryl, 3- ethyl-4-methyl-A r -(4-(A r -((lr,4r)-4-methylcyclohexylcarbamoyl)sulfamoyl)phenethyl)-2-oxo- 2,5-dihydro-l/i-pyrrole-l-carboxamide); and gliclazide (Diamicron, ⁇ '- (hexahydrocyclopenta
  • DGAT-1 inhibitor selected from: (lr,4r)-4-(4-(5-(3,4-difluorophenylamino)-l,3,4-oxadiazole- 2-carboxamido)-3-fluorophenyl)cyclohexanecarboxylic acid; ira «5-(4- ⁇ 4-[5-(6-trifluoromethyl- pyridin-3ylamino)-pyridin-2-yl] -phenyl ⁇ -cyclohexyl)-acetic acid; and ⁇ iraws-4-(4-(4-amino-5- oxo-7, 8-dihydropyrimido[5.4-f] [f]oxazepin-6(5//)yl)phenyllcyclohexyl ⁇ acetic acid.
  • the two distinct pharmaceutical agents include every combination selected from pharmaceutical agents of the following group: sitagliptin, vildagliptin, saxagliptin, alogliptin, linagliptin, phenformin, metformin, buformin, acarbose, miglitol, voglibose, tolbutamide, acetohexamide, tolazamide, chlorpropamide, glipizide, glibenclamide, glimepiride, gliclazide, dapagliflozin, remigliflozin, and sergliflozin.
  • pharmaceutical agents of the following group sitagliptin, vildagliptin, saxagliptin, alogliptin, linagliptin, phenformin, metformin, buformin, acarbose, miglitol, voglibose, tolbutamide, acetohexamide,
  • Dipeptidyl peptidase IV exhibits catalytic activity against a broad range of peptide substrates that includes peptide hormones, neuropeptides, and chemokines.
  • the incretins glucagon-like peptide 1 (GLP-1), 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.
  • Peptide YY is a gut peptide that has been implicated in modulating satiety (Chaudhri et al, Annu Rev Physiol (2008) 70:239-255). PYY is released into the circulation as PYYi_ 36 and PYY 3 _ 36 (Eberlein et al, Peptides (1989) 10:797-803). PYY 3 . 36 is generated from PYYi_ 36 by cleavage of the N-terminal Tyr and Pro residues by DPP-IV. Both pharmacological and genetic attenuation of DPP-IV activity is associated with enhanced incretin action, increased insulin, and lower blood glucose in vivo.
  • 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 therapeutics, for example, oral administration of vildagliptin (l-[2-(3-hydroxyadamant-l- ylamino)acetyl]pyrrolidine-2(S)-carbonitrile) or sitagliptin (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) 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.
  • 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 DPP-IV inhibitors," Biochim. Biophys. Acta, 1751 : 33-44 (2005), and (2) 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, 15: 1387-1407 (2005).
  • 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-known 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) 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. Na.t Prod. (2004) 67:999- 1004.
  • 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, 0-benzoyl hydroxylamine, as described, for example, 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. Pat. Nos. 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 2002006
  • 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 Nos. 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 05/122
  • 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 Nos. 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 126
  • JP 2003238566 JP 2002531547, JP 2002527504, JP 2002517401, JP 2002516318, JP 2002363157, JP 2002356472, JP 2002356471, JP 2002265439, JP 2001510442, JP
  • the DPP-IV inhibitor has an IC 50 of less than about 10 ⁇ , less than about 1 ⁇ , 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.
  • the DPP-IV inhibitor has an IC 50 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 1 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 PPCE, DPP-II, DPP-8 and DPP-9 of at least about 10-fold. 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 PPCE, DPP-II, DPP-8 and DPP-9 of at least about 100-fold.
  • 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 PPCE, DPP-II, DPP-8 and DPP-9 of at least about 10-fold. 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 PPCE, DPP-II, DPP-8 and DPP-9 of at least about 1000-fold.
  • 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 compounds of the following group 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; l-[2-(3- hydroxyadamant-l-ylamino)acetyl]pyrrolidine-2(5)-carbonitrile; (15,35,55)-2-[2(5)-amino-2-(3- hydroxyadamantan- 1 -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-t
  • 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-l-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.
  • EMEA European Medicines Evaluation Agency
  • Some embodiments of the present invention include every combination of one or more compounds selected from compounds disclosed in WO2003/004498 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • the DPP-IV inhibitor 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-l-one, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the DPP-IV inhibitor is (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 phosphate:
  • the DPP-IV inhibitor is crystalline (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 phosphate monohydrate.
  • Vildagliptin (Galvus®, LAF-237, l-[2-(3-hydroxyadamant- l- ylaniino)acetyl]pyrrolidine-2(S)-carbonitrile) is another DPP-IV inhibitor and was first commercialized in Brazil and Mexico by Novartis for oral, once-daily treatment of type 2 diabetes.
  • 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. Vildagliptin was approved in Japan in 2010.
  • the compound, l-[2-(3-hydroxyadamant-l-ylamino)acetyl]pyrrolidine-2( l 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 compounds disclosed in WO2000/034241 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • the DPP-IV inhibitor is selected from l-[2-(3- hydroxyadamant- 1 -ylamino)acetyl]pyrrolidine-2(3 ⁇ 4-carbonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the DPP-IV inhibitor is l-[2-(3-hydroxyadamant-l-ylamino)acetyl]pyrrolidine- 2(S)-carbonitrile HCI:
  • Saxagliptin (OnglyzaTM, BMS-477118, (l£3£5S)-2-[2(S)-ainino-2-(3- hydroxyadamantan-l-yl)acetyl]-2-azabicyclo[3.1.0]hexane-3-carbonitrile) is another DPP-IV inhibitor, which 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 approved in the E.U. for the treatment of type 2 diabetes independently or in combination with metformin. Phase 3 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 compounds disclosed in WO2001/068603 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • the DPP-IV inhibitor is selected from (lS,3S,5S)-2-[2(S)-amino-2-(3- hydroxyadamantan- 1 -yl)acetyl] -2-azabicyclo [3.1.0]hexane-3-carbonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the compound, 2-[6-[3(R)-aminopiperidin-l-yl]-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 compounds disclosed in WO 2005/095381 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • the DPP-IV inhibitor is selected from 2-[6-[3(R)- aminopiperidin-l-yl]-3-methyl-2,4-dioxo- l,2,3,4-tetrahydropyrimidin-l-ylmethyl]benzonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the DPP-IV inhibitor is 2-[6-[3(R)-aminopiperidin-l- yl] -3-methyl-2,4-dioxo-l,2,3,4- tetrahydropyrimidin-l-ylmethyl]benzonitrile benzoate:
  • Linagliptin (BI-1356, Tradjenta®, 8-[3(R)-aminopiperidin-l-yl]-7-(2-butynyl)-3- methyl-l-(4-methylquinazolin-2-ylmethyl)xanthine) is an inhibitor of DPP-IV approved by the FDA in May 2011 as an adjunct to diet and excerise to improve glycemic control in adults with 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 compounds disclosed in WO2004/018468 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • the DPP-IV inhibitor is selected from 8-[3(R)-aminopiperidin-l-yl]-7-(2-butynyl)-3-methyl- l-(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-l-yl]-7-(2-butynyl)-3-methyl-l-(4-methylquinazolin-2-ylmethyl)xanthine.
  • Dutogliptin (PHX-1 149, l-[A r -[3(R)-pyrrolidinyl]glycyl]pyrrolidin-2(R)-yl boronic acid) is a DPP-IV inhibitor in phase 3 clinical trials by Phenomix and Forest for the oral, once-daily treatment of type 2 diabetes.
  • 7V-[3(R)-pyrrolidinyl]glycyl] pyrrolidin-2(R)-yl boronic acid, and pharmaceutically acceptable salts thereof are disclosed in international patent publication WO2005/047297.
  • Some embodiments of the present invention include every combination of one or more compounds selected from compounds disclosed in
  • the DPP-IV inhibitor is selected from l- ⁇ -fS ⁇ -pyrrolidinylJglycylJpyrrolidin- 2(R)-yl boronic acid, and ph lvates, and hydrates thereof:
  • the DPP-IV inhibitor is l- oronic acid tartrate:
  • Melogliptin (GRC-8200, 4(S)-fluoro- 1 - [2- [( lR,3S)-3-( IH- 1 ,2,4-triazol- 1 - ylmethyl)cyclopentylamino]acetyl]pyrrolidine-2( l S r )-carbonitrile) is a DPP-IV inhibitor currently undergoing phase 2 clinical trials by Glenmark Pharmaceuticals and Merck KGaA for the treatment of type 2 diabetes.
  • the DPP-IV inhibitor is selected from 4(S)-fluoro-l-[2-[(lR,3S)-3-(l#-l,2,4- triazol- 1 -ylmethyl)cyclopentylamino]acetyl]pyrrolidine-2( l S')-carbonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • Carmegliptin (R-1579, 1-[(2S,3S, 1 lbS)-2-arnino-9, 10-dimethoxy-2,3,4,6,7,l lb- hexahydro-l/i-pyrido[2,l-a]isoquinolin-3-yl]-4( l S r )-(fluoromethyl)pyrrolidin-2-one) is a DPP-IV inhibitor.
  • the compound, l-[(2 l S',3 l S', l lb l S')-2-amino-9, 10-dimethoxy-2,3,4,6,7,l lb-hexahydro- l/i-pyrido[2,l-a]isoquinolin-3-yl]-4( l S r )-(fluoromethyl)pyrrolidin-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 compounds disclosed in WO2005/000848 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • the DPP-IV inhibitor is selected from l-[(2 l S',3 l S', l lb l S')-2-amino-9, 10-dimethoxy- 2,3,4,6,7, l lb-hexahydro- l/i-pyrido[2, l -a]isoquinolin-3-yl]-4( l S')-(fluoromethyl)pyrrolidin-2- one, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • ethylamino]acetylpyrrolidine a DPP-IV inhibitor in US patent publication US 2007/0112059.
  • Some embodiments of the present invention include every combination of one or more compounds selected from compounds disclosed in US 2007/0112059 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • the DPP-IV inhibitor is selected from (2S,4S)-2-cyano-4-fluoro-l -[(2 -hydroxy- 1, 1- 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. Some embodiments of the present invention include every combination of one or more compounds selected from compounds disclosed in US publication US 2007/0167468 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • the DPP-IV inhibitor is selected from 8-(cw-hexahydro-pyrrolo[3,2-b]pyrrol-l-yl)-3-methyl-7-(3-methyl-but-2-enyl)-l -(2-0X0-2- 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 compounds disclosed in WO2007/148185 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • One such compound is l-((3 l S',4 l S r )-4-amino-l-(4-(3,3-difluoropyrrolidin-l-yl)- l,3,5-triazin-2-yl)pyrrolidin-3-yl)-5,5difluoropiperidin-2-one.
  • the DPP- IV inhibitor is selected from l-((3 l S',4 l S r )-4-amino-l-(4-(3,3-difluoropyrrolidin-l-yl)-l,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(l/i,3//)-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 compounds disclosed in WO2005/095381 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • One such compound is (R)-2-((6-(3-aminopiperidin- l-yl)-3-methyl-2,4-dioxo- 3,4-dihydropyrimidin-l(2//)-yl)methyl)-4-fluorobenzonitrile.
  • the DPP- IV inhibitor is selected from (R)-2-((6-(3-aminopiperidin-l-yl)-3-methyl-2,4-dioxo-3,4- dihydropyrimidin- l(2//)-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(2//)-yl)methyl)-4-fluorobenzonitrile succinic acid salt as described in international patent publication WO2008/067465.
  • the DPP- IV inhibitor is crystalline (R)-2-((6-(3-aminopiperidin-l-yl)-3-methyl-2,4-dioxo-3,4- dihydropyrimidin- l(2//)-yl)methyl)-4-fluorobenzonitrile succinic acid salt:
  • One such compound is 5- ⁇ (S)-2-[2-((S)-2-cyano-pyrrolidin-l-yl)-2-oxo- ethylamino] -propyl ⁇ -5-( l/i-tetrazol-5-yl) 10, 11 -dihydro-5/i-dibenzo[a,d]cycloheptene-2,8- dicarboxylic acid bis-dimethylamide.
  • the DPP-IV inhibitor is selected from 5- ⁇ (S)-2- [2-((S)-2-cyano-pyrrolidin- 1 -yl)-2-oxo-ethylamino] -propyl ⁇ -5-( l/i-tetrazol-5- yl) 10, l l-dihydro-5/i-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 compounds disclosed in WO2002/0014271 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • One such compound is ((2S,4S)-4-(4-(3-methyl-l -phenyl- l/i-pyrazol-5- yl)piperazin-l-yl)pyrrolidin-2-yl)(thiazolidin-3-yl)methanone.
  • the DPP- IV inhibitor is selected from ((2 l S',4 l S r )-4-(4-(3-methyl-l-phenyl-l/i-pyrazol-5-yl)piperazin-l- 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 crystalline forms of ((2S,4S)-4-(4-(3-methyl-l- phenyl- l/i-pyrazol-5-yl)piperazin- 1 -yl)pyrrolidin-2-yl)(thiazolidin-3-yl)methanone salt as described in international patent publication WO2006/088129 and US publication
  • the DPP-IV inhibitor is crystalline ((25,45)-4-(4-(3- methyl- 1 -phenyl- l/i-pyrazol-5-yl)piperazin- 1 -yl)pyrrolidin-2-yl)(thiazolidin-3-yl)methanone 2.5 hydrobromide salt:
  • the DPP- IV inhibitor is crystalline ((25,45)-4-(4-(3-mefhyl-l -phenyl- l/i-pyrazol-5-yl)piperazin-l-yl)pyrrolidin-2-yl)(thiazolidin-3- yl)methanone di-hydrobromide salt.
  • Some embodiments of the present invention include every combination of one or more compounds selected from compounds disclosed in WO2008/114857 and US publication US 2008/0146818, and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • One such compound is (2 l S',4 l S r )-l-[2-[(4-ethoxycarbonylbicyclo[2.2.2]oct-l-yl)amino]acetyl]-4-fluoropyrrolidine-2- carbonitrile.
  • the DPP-IV inhibitor is selected from (25,45)- 1- [2- [(4- ethoxycarbonylbicyclo[2.2.2]oct-l-yl)amino]acetyl]-4-fluoropyrrolidine-2-carbonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • Some embodiments of the present invention include every combination of one or more compounds selected from compounds disclosed in WO2006/068163 and US publication US 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.
  • the DPP-IV inhibitor is selected from (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, solvates, and hydrates thereof:
  • the DPP-IV inhibitor is selected from 2-( ⁇ 6-[(3R)-3-amino-3- methylpiperidin-l-yl]-l,3-dimethyl-2,4-dioxo- 1,2,3, 4-tetrahydro-5/i-pyrrolo[3,2-d]pyrimidin-5- yl ⁇ methyl)-4-fluorobenzonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • Hoffmann-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 compounds disclosed in WO 03/037327 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • One such compound is (23 ⁇ 4-l- ⁇ [2-(5-mefhyl-2-phenyl-oxazol-4-yl)- ethylamino] -acetyl ⁇ -pyrrolidine-2-carbonitrile.
  • the DPP-IV inhibitor is selected from (2S)-l- ⁇ [2-(5-methyl-2-phenyl-oxazol-4-yl)-ethylamino] -acetyl ⁇ -pyrrolidine-2- carbonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the DPP-IV inhibitor 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 is selected from (2S)-l- ⁇ [1, 1 -dimethyl-3-(4-pyridin-3-yl-imidazol- 1 -yl)-propylamino] -acetyl ⁇ -pyrrolidine-2- carbonitrile, and pharmaceu ates thereof:
  • the DPP-IV inhibitor is (2S)-l- ⁇ [l,l-dimethyl-3-(4-pyridin-3-yl- imidazol- 1 -yl)-propylamino] -acetyl ⁇ -pyrrolidine-2-carbonitrile methansulfonic acid:
  • the DPP-IV inhibitor is (25)- 1- ⁇ [1, 1 -dimethyl-3-(4-pyridin-3-yl-imidazol- 1 -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. Some embodiments of the present invention include every combination of one or more compounds selected from compounds disclosed in
  • WO2005/1 16014 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • One such compound is (3,3-difluoropyrrolidin-l-yl)-((2 l S , ,45)-4-(4-(pyrimidin-2-yl)piperazin-l- yl)pyrrolidin-2-yl)methanone.
  • the DPP-IV inhibitor is selected from (3,3-difluoropyrrolidin- 1 - ⁇ 1)-((25,45)-4-(4-( ⁇ (1 ⁇ -2- ⁇ 1) ⁇ - 1 -yl)pyrrolidin-2- yl)methanone, and pharmac ates thereof:
  • GlaxoSmithKline 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,43 ⁇ 4-l-[(2S)-2-amino-3,3-bis(4- fluorophenyl)propanoyl]-4-fluoropyrrolidine-2-carbonitrile (Denagliptin).
  • the DPP-IV inhibitor is selected from (2S,43 ⁇ 4-l-[(23 ⁇ 4-2-amino-3,3-bis(4- fluorophenyl)propanoyl]-4-fluoropyrrolidine-2-carbonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • One salt disclosed is (2S,4S)-l-[(2S)-2- amino-3,3-bis(4-fluorophenyl)propanoyl]-4-fluoropyrrolidine-2-carbonitrile j-toluenesulfonic acid salt (also referred to as (2 l S',4 l S r )-4-fluoro-l-[4-fluoro- -(4-fluorophenyl)-L-phenylalanyl]-2- pyrrolidinecarbonitrile j-toluenesulfonic acid salt, or Denagliptin tosylate).
  • j-toluenesulfonic acid salt also referred to as (2 l S',4 l S r )-4-fluoro-l-[4-fluoro- -(4-fluorophenyl)-L-phenylalanyl]-2- pyrrolidinecarbonitrile j-toluenesulfonic
  • the DPP-IV inhibitor is (2S,43 ⁇ 4-l-[(2S)-2-amino-3,3-bis(4- fluorophenyl)propanoyl]-4-fluoropyrrolidine-2-carbonitrile j-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 (2 l S',5R)-5-ethynyl-l- ⁇ A i -(4-methyl-l-(4-carboxy-pyridin-2- yl)piperidin-4-yl)glycyl ⁇ pyrrolidine-2-carbonitrile.
  • the DPP-IV inhibitor is selected from (2 l S',5R)-5-ethynyl-l- ⁇ A i -(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 (15,6 ⁇ )-3- ⁇ [3- (trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8//)-yl]carbonyl ⁇ -6-(2,4,5- trifluorophenyl)cyclohex-3-en-l -amine.
  • the DPP-IV inhibitor is selected from (l l S',6R)-3- ⁇ [3-(trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8//)- yl]carbonyl ⁇ -6-(2,4,5-trifluorophenyl)cyclohex-3-en-l-amine, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the biguanides are a class of drugs that stimulate anaerobic glycolysis, increase the sensitivity to insulin in the peripheral tissues, inhibit glucose absorption from the intestine, suppress of hepatic gluconeogenesis, and inhibit fatty acid oxidation.
  • biguanides include phenformin ((phenylethyl)biguanide), metformin (dimethylbiguanide), buformin
  • the pharmaceutical agent or said second pharmaceutical agent is a biguanide selected from the following biguanide: (phenylethyl)biguanide, dimethylbiguanide, butylbiguanide, l-(p-chlorophenyl)-5- isopropylbiguanide, and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • the pharmaceutical agent or the second pharmaceutical agent is a biguanide selected from (phenylethyl)biguanide (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the pharmaceutical agent or the second pharmaceutical agent is a biguanide selected from dimethylbiguanide (chemical structure shown below) and
  • the pharmaceutical agent or the second pharmaceutical agent is a biguanide selected from butylbiguanide (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof; the chemical structure is as follows:
  • the pharmaceutical agent or the second pharmaceutical agent is a biguanide selected from l-(p-chlorophenyl)-5-isopropylbiguanide (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof; the chemical structure is as follows:
  • the pharmaceutical agent or said second pharmaceutical agent is a biguanide selected from the following biguanides: metformin, phenformin, buformin, and proguanil.
  • the pharmaceutical agent or the second pharmaceutical agent is metformin.
  • the pharmaceutical agent or the second pharmaceutical agent is phenformin.
  • the pharmaceutical agent or the second pharmaceutical agent is buformin.
  • the pharmaceutical agent or the second pharmaceutical agent is proguanil.
  • Alpha-Gluocosidase Inhibitors ⁇ -Glucosidase inhibitors belong to the class of drugs which competitively inhibit digestive enzymes such as a-amylase, maltase, a-dextrinase, sucrase, etc. in the pancreas and or small intestine.
  • the reversible inhibition by a-glucosidase inhibitors retard, diminish or otherwise reduce blood glucose levels by delaying the digestion of starch and sugars.
  • a-glucosidase inhibitors include acarbose ((2R,3R,4R,5R)-4- ((2R,3R,4R,5 l S , ,6R)-5-((2R,3R,4 l S , ,5 l S , ,6R)-3,4-dihydroxy-6-methyl-5-((l l S , ,4R,5.S , ,6 l S , )-4,5,6- trihydroxy-3-(hydroxymethyl)cyclohex-2-enylamino)tetrahydro-2/i-pyran-2-yloxy)-3,4- dihydroxy-6-(hydroxymethyl)tetrahydro-2/i-pyran-2-yloxy)-2,3,5,6-tetrahydroxyhexanal), miglitol ((2R,3R,4R,5 l S r )-l-(2-hydroxyethyl)-2-(hydroxymethyl)
  • the pharmaceutical agent or said second pharmaceutical agent is a ⁇ -glucosidase inhibitor selected from the following ⁇ -glucosidase inhibitors:
  • the pharmaceutical agent or the second pharmaceutical agent is a ⁇ -glucosidase inhibitor selected from (2 ⁇ ,3 ⁇ ,4 ⁇ ,5 ⁇ )-4-((2 ⁇ ,3 ⁇ ,4 ⁇ ,55,6 ⁇ )-5-((2 ⁇ ,3 ⁇ ,45,55,6 ⁇ )- 3,4-dihydroxy-6-methyl-5-((l l S',4R,5 l S',6 l S')-4,5,6-trihydroxy-3-(hydroxymethyl)cyclohex-2- enylamino)tetrahydro-2/i-pyran-2-yloxy)-3,4-dihydroxy-6-(hydroxymethyl)tetrahydro-2/i- pyran-2-yloxy)-2,3,5,6-tetrahydroxyhexanal (chemical structure shown below) and
  • the pharmaceutical agent or the second pharmaceutical agent is a ⁇ -glucosidase inhibitor selected from (2R,3R,4R,5S)-l-(2-hydroxyefhyl)-2- (hydroxymethyl)piperidine-3,4,5-triol (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the pharmaceutical agent or the second pharmaceutical agent is a a-glucosidase inhibitor selected from (lS,2S,3R,4S,5S)-5-(l,3-dihydroxypropan-2-ylamino)-l- (hydroxymethyl)cyclohexane-l,2,3,4-tetraol (chemical structure shown below) and
  • the pharmaceutical agent or the second pharmaceutical agent is an alpha-glucosidase inhibitor selected from: acarbose, miglitol, and voglibose.
  • the pharmaceutical agent or the second pharmaceutical agent is acarbose.
  • the pharmaceutical agent or the second pharmaceutical agent is miglitol.
  • the pharmaceutical agent or the second pharmaceutical agent is voglibose.
  • insulin analogue refers to the naturally occurring human hormone and insulin receptor ligands (i.e., synthetic insulin analogues). Insulin receptor ligands are structurally different from the natural human hormone, but have substantially the same activity as human insulin in terms of glycemic control.
  • an insulin analogue examples include, NPH insulin (also known as Humulin N, Novolin N, NPH Lletin II, and insulin isophane), insulin lispro (28B-L-lysine-29B-L-proline-insulin, wherein insulin is human insulin), insulin aspart (28B-L-aspartic acid-insulin, wherein insulin is human insulin), insulin glulisine (3B-L-lysine- 29B-L-glutamic acid-insulin, wherein insulin is human insulin), and insulin analogues known in the art.
  • NPH insulin also known as Humulin N, Novolin N, NPH Lletin II, and insulin isophane
  • insulin lispro 28B-L-lysine-29B-L-proline-insulin, wherein insulin is human insulin
  • insulin aspart 28B-L-aspartic acid-insulin, wherein insulin is human insulin
  • insulin glulisine 3B-L-lysine- 29B-L-gluta
  • NPH insulin is marketed by Eli Lilly and Company under the name Humulin N, and is considered as an intermediate-acting insulin analogue given to help control the blood sugar level of those with diabetes.
  • Insulin lispro is marketed by Eli Lilly and Company under the name Humalog, and is considered a rapid acting insulin analogue.
  • Insulin aspart is marketed by Novo Nordisk and sold as NovoRapid. Insulin aspart is considered a fast acting insulin analogue.
  • Insulin glulisine was developed by Sanofi-Aventis and is sold under the trade name Apidra. Insulin glulisine is considered a rapid acting insulin analogue but shorter duration of action compared to human insulin.
  • the pharmaceutical agent or the second pharmaceutical agent is an insulin analogue selected from NPH insulin and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is an insulin analogue selected from insulin lispro and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is an insulin analogue selected from insulin aspart and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is an insulin analogue selected from insulin glulisine and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • the sulfonylureas are drugs which promote secretion of insulin from pancreatic beta cells by transmitting signals of insulin secretion via receptors in the cell membranes.
  • Examples of a sulfonylurea include tolbutamide (Orinase, A r -(butylcarbamoyl)-4- methylbenzenesulfonamide); acetohexamide (Dymelor, 4-acetyl-A r -
  • glibenclamide also known as glyburide (Diabeta, Micronase, Glynase, 5-chloro-A r -(4-(A r - (cyclohexylcarbamoyl)sulfamoyl)phenethyl)-2-methoxybenzamide); glimepiride (Amaryl, 3- ethyl-4-methyl-A r -(4-(A r -((lr,4r)-4-methylcyclohexylcarbamoyl)sulfamoyl)phenethyl)-2-oxo- 2,5-dihydro- l/i-pyrrole-l-carboxamide); gliclazide (Diamicron, ⁇ '- (hexahydrocyclopenta
  • the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from sulfonylureas:
  • the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from A r -(butylcarbamoyl)-4-methylbenzenesulfonamide (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from 4-acetyl-A L (cyclohexylcarbamoyl)benzenesulfonamide (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from A r -(azepan-l-ylcarbamoyl)-4-methylbenzenesulfonamide (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from 4-chloro-A L (propylcarbamoyl)benzenesulfonamide (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from A r -(4-(A r -(cyclohexylcarbamoyl)sulfamoyl)phenethyl)-5- methylpyrazine-2-carboxamide (chemical structure shown below) and pharmaceutically acceptable salts, sol
  • the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from 5-chloro-A r -(4-(A r -(cyclohexylcarbamoyl)sulfamoyl)phenethyl)-2- methoxybenzamide (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from 3-ethyl-4-methyl-A r -(4-(A r -((lr,4r)-4- methylcyclohexylcarbamoyl)sulfamoyl)phenethyl)-2-oxo-2,5-dihydro- 1/i-pyrrole- 1 - carboxamide (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from A r -(hexahydrocyclopenta[c]pyrrol-2(l//)-ylcarbamoyl)-4- methylbenzenesulfonamide (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from the following sulfonylureas and pharmaceutically acceptable salts, solvates, and hydrates thereof: glipizide, glimepiride, and glibenclamide.
  • the pharmaceutical agent or the second pharmaceutical agent is tolbutamide.
  • the pharmaceutical agent or the second pharmaceutical agent is acetohexamide.
  • the pharmaceutical agent or the second pharmaceutical agent is tolazamide.
  • the pharmaceutical agent or the second pharmaceutical agent is chlorpropamide.
  • the pharmaceutical agent or the second pharmaceutical agent is glipizide.
  • the pharmaceutical agent or the second pharmaceutical agent is glyburide.
  • the pharmaceutical agent is glimepiride.
  • the pharmaceutical agent or the second pharmaceutical agent is gliclazide.
  • Sodium-glucose transporter-2 (SGLT2) inhibitors belong to the class of drugs which inhibit the protein SGLT2 and the reabsorption of glucose in the kidney. The inhibition by SGLT2 inhibitors retard, diminish, or otherwise reduce the amount of glucose that is reabsorbed and therefore is eliminated in the urine.
  • SGLT2 inhibitors include dapagliflozin ((2 l S , ,3R,4R,5.S , ,6R)-2-(4-chloro-3-(4-ethoxybenzyl)phenyl)-6- (hydroxymethyl)tetrahydro-2/i-pyran-3,4,5-triol, Bristol-Myers Squibb and AstraZeneca), remogliflozin (ethyl ((2R,3 l S',4 l S',5R,6 l S r )-3,4,5-trihydroxy-6-(4-(4-isopropoxybenzyl)-l-isopropyl- 5-methyl- l/i-pyrazol-3-yloxy)tetrahydro-2/i-pyran-2-yl)methyl carbonate, Glaxo SmithKline), ASP1941 (Kotobuki/Astellas), canagliflozin ((25,3 ⁇ ,4
  • the pharmaceutical agent or the second pharmaceutical agent is a SGLT2 inhibitor selected from the following SGLT2 inhibitors:
  • the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from (2S,3R,4R,5S,6R)-2-(4-chloro-3-(4-efhoxybenzyl)phenyl)-6- (hydroxymethyl)tetrahydro-2/i-pyran-3,4,5-triol (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from ethyl ((2R,3 l S',4 l S',5R,6 l S r )-3,4,5-trihydroxy-6-(4-(4-isopropoxybenzyl)- l-isopropyl-5-methyl-l/i-pyrazol-3-yloxy)tetrahydro-2/i-pyran-2-yl)methyl carbonate (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from ethyl ((2R,3S,4S,5R,63 ⁇ 4-3,4,5-trihydroxy-6-(2-(4- methoxybenzyl)phenoxy)tetrahydro-2/i-pyran-2-yl)methyl carbonate (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the pharmaceutical agent or the second pharmaceutical agent is a SGLT2 inhibitor selected from: dapagliflozin, remigliflozin, and sergliflozin.
  • the pharmaceutical agent or the second pharmaceutical agent is dapagliflozin.
  • the pharmaceutical agent or the second pharmaceutical agent is remigliflozin.
  • the pharmaceutical agent or the second pharmaceutical agent is sergliflozin.
  • Astellas and Kotobuki disclosed a series of SGLT2 inhibitors in international patent publication WO2004/080990. Some embodiments of the present invention include every combination of one or more compounds selected from compounds disclosed in
  • Some embodiments of the present invention include every combination of one or more compounds selected from compounds disclosed in WO2004/007517 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • One such compound is (2R,3S, 4 l S , ,5R,6,S , )-2-(hydroxymethyl)-6-(2-(4-methoxybenzyl)thiophen-3-yloxy)tetrahydro-2H- pyran-3,4,5-triol.
  • the SGLT2 inhibitor is selected from (2R,3S,4S,5R,6S)- 2-(hydroxymethyl)-6-(2-(4-methoxybenzyl)thiophen-3-yloxy)tetrahydro-2H-pyran-3,4,5-triol, and pharmaceutically accepta hydrates thereof:
  • Some embodiments of the present invention include every combination of one or more compounds selected from compounds disclosed in WO2005/012326 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • One such compound is (25,3R,4R,55,6R)-2-(3-((5-(4-fluorophenyl)thiophen-2-yl)methyl)-4-methylphenyl)-6- (hydroxymethyl)tetrahydro-2/i-pyran-3,4,5-triol.
  • the SGLT2 inhibitor is selected from (25,3R,4R,55,6R)-2-(3-((5-(4-fluorophenyl)thiophen-2-yl)methyl)-4- methylphenyl)-6-(hydroxymethyl)tetrahydro-2/i-pyran-3,4,5-triol, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • Boehringer Ingelheim disclosed a series of SGLT2 inhibitors in international patent publication WO2005/092877. Some embodiments of the present invention include every combination of one or more compounds selected from compounds disclosed in
  • Chugai disclosed a series of SGLT2 inhibitors in international patent publication WO2006/080421. Some embodiments of the present invention include every combination of one or more compounds selected from compounds disclosed in WO2006/080421 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • Lexicon disclosed a series of SGLT2 inhibitors in international patent publication WO2008/109591. Some embodiments of the present invention include every combination of one or more compounds selected from compounds disclosed in WO2008/109591 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • Meglitinides The meglitinides promote secretion of insulin by binding to the pancreatic beta cells in a similar manner as sulfonylureas but at an alternative binding site.
  • meglitinides include Novo Nordisk's repaglinide (Prandin, (3 ⁇ 4-2-ethoxy-4-(2-(3-methyl-l-(2-(piperidin-l- yl)phenyl)butylamino)-2-oxoethyl)benzoic acid), nateglinide (Starlix, (R)-2-((lr,4R)-4- isopropylcyclohexanecarboxamido)-3-phenylpropanoic acid), mitiglinide (( l S r )-2-benzyl-4- ((3aR aS) H ⁇ so dol-2(3H,3aHAH,5H,6H H aH)-yl)-4-oxob tanoic acid
  • the pharmaceutical agent or the second pharmaceutical agent is a meglitinide selected from the following meglitinides: (3 ⁇ 4-2-ethoxy-4-(2-(3-mefhyl- l-(2- (piperidin- l-yl)phenyl)butylamino)-2-oxoethyl)benzoic acid; (R)-2-((lr,4R)-4- isopropylcyclohexanecarboxamido)-3-phenylpropanoic acid; (S)-2-benzyl-4-((3aR,7aS)- IH- isoindol-2(3/i,3a/i,4/i,5/i,6/i,7/i,7a//)-yl)-4-oxobutanoic acid; and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • the pharmaceutical agent or the second pharmaceutical agent is (S)-2-efhoxy-4-(2-(3-mefhyl- 1 -(2-(piperidin- 1 -yl)phenyl)butylamino)-2-oxoethyl)benzoic acid (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from (R)-2-((lr,4R)-4-isopropylcyclohexanecarboxamido)-3- phenylpropanoic acid (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from (S)-2-benzyl-4-((3aR,7aS)- l/i-isoindol-
  • the pharmaceutical agent or the second pharmaceutical agent is a meglitinide selected from the following meglitinides: repaglinide, nateglinide, mitiglinide, and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • the pharmaceutical agent or the second pharmaceutical agent is a meglitinide selected from repaglinide and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • the pharmaceutical agent or the second pharmaceutical agent is a meglitinide selected from nateglinide and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • the pharmaceutical agent or the second pharmaceutical agent is a meglitinide selected from mitiglinide and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • Thiazolidinediones belong to the class of drugs more commonly known as TZDs. These drugs act by binding to the nuclear receptor peroxisome proliferator-activated receptor gamma (PPARy) activate transcription of a number of specific genes leading to a decrease in insulin resistance.
  • TZDs nuclear receptor peroxisome proliferator-activated receptor gamma
  • thiazolidinediones examples include rosiglitazone (Avandia, 5-(4-(2- (methyl(pyridin-2-yl)amino)ethoxy)benzyl)thiazolidine-2,4-dione), pioglitazone (Actos, 5-(4-(2- (5-ethylpyridin-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione), troglitazone (Rezulin, 5-(4-((6- hydroxy-2,5,7,8-tetramethylchroman-2-yl)methoxy)benzyl)thiazolidine-2,4-dione), rivoglitazone (5-(4-((6-methoxy-l-methyl-l/i-benzo[d]imidazol-2- yl)methoxy)benzyl)thiazolidine-2,4-dione), ciglitazone(5-(4-((
  • the pharmaceutical agent or the second pharmaceutical agent is a meglitinide selected from: 5-(4-(2-(methyl(pyridin-2-yl)amino)ethoxy)benzyl)thiazolidine-2,4- dione; 5-(4-(2-(5-ethylpyridin-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione; 5-(4-((6-methoxy-l/i- benzo[d]imidazol-2-yl)methoxy)benzyl)thiazolidine-2,4-dione; 5-(4-((l- methylcyclohexyl)methoxy)benzyl)thiazolidine-2,4-dione; and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • the pharmaceutical agent or the second pharmaceutical agent is 5-(4-(2-(methyl(pyridin-2-yl)amino)ethoxy)benzyl)thiazolidine-2,4-dione (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the pharmaceutical agent or the second pharmaceutical agent is 5-(4-(2-(5-ethylpyridin-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the pharmaceutical agent or the second pharmaceutical agent is 5-(4-((6-hydroxy-2,5,7,8-tetramethylchroman-2-yl)methoxy)benzyl)thiazolidine-2,4-dione (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the pharmaceutical agent or the second pharmaceutical agent is 5-(4-((6-methoxy-l -methyl- l/i-benzo[d]imidazol-2-yl)methoxy)benzyl)thiazolidine-2,4-dione (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the pharmaceutical agent or the second pharmaceutical agent is 5-(4-((l-methylcyclohexyl)methoxy)benzyl)thiazolidine-2,4-dione (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • the pharmaceutical agent or the second pharmaceutical agent is a thiazolidinedione selected from rosiglitazone and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a thiazolidinedione selected from pioglitazone and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a thiazolidinedione selected from troglitazone and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • the pharmaceutical agent or the second pharmaceutical agent is a thiazolidinedione selected from rivoglitazone and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a
  • thiazolidinedione selected from ciglitazone and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • Anti-diabetic peptide analogues are peptides that promote secretion of insulin by acting as an incretin mimetic, such as, GLP-1 and GIP.
  • examples of an anti-diabetic peptide analog include, exenatide, liraglutide, taspoglutide, and anti-diabetic peptides analogues know in the art.
  • the pharmaceutical agent or the second pharmaceutical agent is an anti-diabetic peptide analogue selected from: exenatide; liraglutide; and taspoglutide.
  • the pharmaceutical agent or the second pharmaceutical agent is exenatide.
  • the pharmaceutical agent or the second pharmaceutical agent is liraglutide.
  • the pharmaceutical agent or the second pharmaceutical agent is taspoglutide.
  • the pharmaceutical agent or the second pharmaceutical agent is L-histidylglycyl-L-a-glutamylglycyl-L-threonyl-L-phenylalanyl-L-threonyl-L-seryl-L-a- aspartyl-L-leucyl-L-seryl-L-lysyl-L-glutaminyl-L-methionyl-L-a-glutamyl-L-a-glutamyl-L-a-glutamyl-L-a- glutamyl-L-alanyl-L-valyl-L-arginyl-L-leucyl-L-phenylalanyl-L-isoleucyl-L-a-glutamyl-L- tryptophyl-L-leucyl-L-lysyl-L-asparaginylglycyl-L-prolyl-L-seryl-L
  • the pharmaceutical agent or the second pharmaceutical agent is L-histidyl-L-alanyl-L-a-glutamylglycyl-L-threonyl-L-phenylalanyl-L-threonyl-L-seryl-L-a- aspartyl-L-valyl-L-seryl-L-seryl-L-tyrosyl-L-leucyl-L-a-glutamylglycyl-L-glutaminyl-L-alanyl- L-alanyl-N6-[A r -(l-oxohexadecyl)-L-a-glutamyl]-L-lysyl-L-a-glutamyl-L-phenylalanyl-L- isoleucyl-L-alanyl-L-tryptophyl-L-leucyl-L-valyl-L-arginylgly
  • the pharmaceutical agent or the second pharmaceutical agent is H 2 N-His-2-methyl-Ala-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Gly-Gln-Ala- Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Val-Lys-2-methyl-Ala-Arg-CONH 2 (taspoglutide) and pharmaceutically acceptable salts, solvates, and hydrates thereof.
  • Triglycerides represent the major form of energy stored in eukaryotes. Disorders and/or imbalances in triglyceride metabolism are implicated in the pathogenesis of and increased risk for metabolic related disorders, such as, obesity, insulin resistance syndrome and type 2 diabetes, nonalcoholic fatty liver disease, and coronary heart disease.
  • Triglyceride biosynthesis and the resulting TG burden on tissues are largely controlled by two major pathways in humans. Both of these pathways converge at an intermediate diacylglycerols (DAG) which are subsequently converted to triglycerides through acylation by a fatty acid acyl-CoA catalyzed by the acyl-CoA:diacylglycerol acyltransferases (DGAT).
  • DAG diacylglycerols
  • DGAT consists of DGAT-1 and DGAT-2.
  • Inhibition of acyl-CoA:diacylglycerol acyltransferase-1 (DGAT-1) is one of the most advanced targets for altering lipid biosyntheis as evidenced by on going human clinical trials.
  • the pharmaceutical agent or said second pharmaceutical agent is a DGAT-1 inhibitor known in the art.
  • the pharmaceutical agent or said second pharmaceutical agent is a DGAT-1 inhibitor selected from the DGAT-1 inhibitors described in the following patent applications and pharmaceutically acceptable salts, solvates, and hydrates thereof: WO 2004/047755, WO 2004/100881, WO 2005/0727401, WO 2006/044775, WO 2006/06019020, WO 2006/082010, WO2006/113919, WO 2006/134317, WO2007/126957, WO2012/051488, WO2009/016462.
  • the pharmaceutical agent or said second pharmaceutical agent is a DGAT-1 inhibitor selected from the following DGAT-1 inhibitors and pharmaceutically acceptable salts, solvates, and hydrates thereof:
  • Another object of the present invention relates to radio-labeled compounds of the present invention that would be useful not only in radio-imaging but also in assays, both in vitro and in vivo, for localizing and quantitating GPR119 receptors in tissue samples, including human and for identifying GPR119 receptor ligands by inhibition binding of a radio-labeled compound. It is a further object of this invention to develop novel GPR119 receptor assays of which comprise such radio-labeled compounds.
  • the present disclosure includes all isotopes of atoms occurring in the present compounds, intermediates, salts and crystalline forms thereof.
  • Isotopes include those atoms having the same atomic number but different mass numbers.
  • One aspect of the present invention includes every combination of one or more atoms in the present compounds, intermediates, salts, and crystalline forms thereof that is replaced with an atom having the same atomic number but a different mass number.
  • One such example is the replacement of an atom that is the most naturally abundant isotope, such as 3 ⁇ 4 or 12 C, found in one the present compounds, intermediates, salts, and crystalline forms thereof, with a different atom that is not the most
  • isotopes of hydrogen include 2 H (deuterium) and 3 H (tritium).
  • isotopes of carbon include n C, 13 C, and 14 C.
  • Isotopes of oxygen include O, O, and C.
  • An isotope of fluorine includes 18 F.
  • An isotope of sulfur includes 35 S.
  • An isotope of chlorine includes 36 C1.
  • Isotopes of bromine include 75 Br, 76 Br, 77 Br, and 82 Br.
  • Isotopes of iodine include 123 1, 124 I, 125 I, and 131 I.
  • compositions such as, those prepared during synthesis, preformulation, and the like, and pharmaceutical compositions, such as, those prepared with the intent of using in a mammal for the treatment of one or more of the disorders described herein, comprising one or more of the present compounds, intermediates, salts, and crystalline forms thereof, wherein the naturally occurring distribution of the isotopes in the composition is perturbed.
  • compositions and pharmaceutical compositions comprising compounds as described herein wherein the compound is enriched at one or more positions with an isotope other than the most naturally abundant isotope.
  • 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 (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 Drawings and Examples infra, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.
  • Synthetic methods for incorporating radio-isotopes into organic compounds are applicable to compounds of the invention and are well known in the art. These synthetic methods, for example, incorporating activity levels of tritium into target molecules, are as follows:
  • Tritium Gas Exposure Labeling This procedure involves exposing precursors containing exchangeable protons to tritium gas in the presence of a suitable catalyst.
  • N-Mefhylation using Methyl Iodide [ 3 H] This procedure is usually employed to prepare O-methyl or N-mefhyl (3H) products by treating appropriate precursors with high specific activity methyl iodide (3H). This method in general allows for higher specific activity, such as for example, about 70-90 Ci/mmol.
  • Synthetic methods for incorporating activity levels of 125 I into target molecules include:
  • Aryl and heteroaryl bromide exchange with 125 I This method is generally a two step process.
  • the first step is the conversion of the aryl or heteroaryl bromide to the corresponding tri-alkyltin intermediate using for example, a Pd catalyzed reaction [i.e. Pd(Ph 3 P) 4 ] or through an aryl or heteroaryl lithium, in the presence of a tri-alkyltinhalide or hexaalkylditin [e.g., (CH 3 ) 3 SnSn(CH 3 )3] .
  • Pd catalyzed reaction i.e. Pd(Ph 3 P) 4
  • a tri-alkyltinhalide or hexaalkylditin e.g., (CH 3 ) 3 SnSn(CH 3 )3
  • a radiolabeled GPR119 receptor compound of Formula (I) can be used in a screening assay to identify/evaluate compounds.
  • a newly synthesized or identified compound i.e., test compound
  • test compound can be evaluated for its ability to reduce binding of the "radiolabeled compound of Formula (I)" to a GPR119 receptor. Accordingly, the ability of a test compound to compete with the "radio-labeled compound of Formula (I)" for the binding to a GPR119 receptor directly correlates to its binding affinity.
  • Certain labeled compounds of the present invention bind to certain GPR119 receptors.
  • the labeled compound has an IC 50 less than about 500 ⁇ , in another embodiment the labeled compound has an IC 50 less than about 100 ⁇ , in yet another embodiment the labeled compound has an IC 50 less than about 10 ⁇ , in yet another embodiment the labeled compound has an IC 50 less than about 1 ⁇ and in still yet another embodiment the labeled inhibitor has an IC 50 less than about 0.1 ⁇ .
  • Example 1 Syntheses of Compounds of the Present Invention.
  • TLC Thin-layer chromatography
  • 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 under reduced pressure on a Biichi rotary evaporator.
  • LCMS spec HPLC -pumps: LC-10AD VP, Shimadzu Inc.; HPLC system controller: SCL-IOA VP, Shimadzu Inc; UV-Detector: SPD-10A 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.
  • Example 1.1 Preparation of 1-Methylcyclopropyl 4-((lr,4r)-4-(3-Cyanopyrazin-2- yloxy)cyclohexyloxy)piperidine-l-carboxylate (Compound 1).
  • Step 1 Preparation of 1-Methylcyclopropyl 4-((lr,4r)-4-Hydroxycyclohexyloxy) piperidine- 1 -carboxylate.
  • Step 2 Preparation of 1-Methylcyclopropyl 4-((lr,4r)-4-(3-Cyanopyrazin-2- yloxy)cyclohexyloxy)piperidine-l-carboxylate (Compound 1).
  • Example 1.2 Preparation of 3-((lr,4r)-4-(l-(5-Chloropyrimidin-2-yl)piperidin-4- yloxy)cyclohexyloxy)pyrazine-2-carbonitrile (Compound 2).
  • Step 1 Preparation of (lr,4r)-4-(Piperidin-4-yloxy)cyclohexanol.
  • Step 2 tert-Butyl 4-((lr,4r)-4-Hydroxycyclohexyloxy)piperidine-l-carboxylate.
  • Step 3 Preparation of teri-Butyl 4-((lr,4r)-4-(3-Cyanopyrazin-2- yloxy)cyclohexyloxy)piperidine-l-carboxylate.
  • Step 4 Preparation of 3-((lr,4r)-4-(Piperidin-4-yloxy)cyclohexyloxy)pyrazine-2- carbonitrile Hydrochloride.
  • Step 5 Preparation of 3-((lr,4r)-4-(l-(5-Chloropyrimidin-2-yl)piperidin-4- yloxy)cyclohexyloxy)pyrazine-2-carbonitrile (Compound 2).
  • Example 1.3 Preparation of (/f)-l,l,l-Trifluoropropan-2-yl 4-((lr,4r)-4-(3-Cyanopyrazin- 2-yloxy)cyclohexyloxy)piperidine-l-carboxylate (Compound 3).
  • Example 1.5 Preparation of (/f)-l-(2,2,2-Trifluoroethyl)pyrrolidin-3-yl 4-((lr,4r)-4-(3- Cyanopyrazin-2-yloxy)cyclohexyloxy)piperidine-l-carboxylate (Compound 5).
  • Step 1 Preparation of (/f)-l-(tert-Butoxycarbonyl)pyrrolidin-3-yl 4-((lr,4r)-4-(3- Cyanopyrazin-2-yloxy)cyclohexyloxy)piperidine-l-carboxylate.
  • Step 2 Preparation of (/f)-Pyrrolidin-3-yl 4-((lr,4r)-4-(3-Cyanopyrazin-2- yloxy)cyclohexyloxy)piperidine-l-carboxylate Hydrochloride.
  • Step 3 Preparation of (K)-l-(2,2,2-Trifluoroethyl)pyrrolidin-3-yl 4-((lr,4r)-4-(3- Cyanopyrazin-2-yloxy)cyclohexyloxy)piperidine-l-carboxylate (Compound 5).
  • Example 1.7 Preparation of 3-((lr,4r)-4-(l-(3-(Trifluoromethyl)-l,2,4-oxadiazol-5- yl)piperidin-4-yloxy)cyclohexyloxy)pyrazine-2-carbonitrile (Compound 7).
  • Step 1 Preparation of (lr,4r)-4-(l-(3-(Trifluoromethyl)-l,2,4-oxadiazol-5- yl)piperidin-4-yloxy)cyclohexanol.
  • Step 2 Preparation of -((lr,4r)-4-(l-(3-(Trifluoromethyl)-l,2,4-oxadiazol-5- yl)piperidin-4-yloxy)cyclohexyloxy)pyrazine-2-carbonitrile (Compound 7).
  • Examplel.8 Preparation of 3-((lr,4r)-4-(l-(5-(Trifluoromethyl)-l,2,4-oxadiazol-3- yl)piperidin-4-yloxy)cyclohexyloxy)pyrazine-2-carbonitrile (Compound 8).
  • Step 1 Preparation of (lr,4r)-4-(l-(5-(Trifluoromethyl)-l,2,4-oxadiazol-3- yl)piperidin-4-yloxy)cyclohexanol.
  • Step 2 Preparation of 3-((lr,4r)-4-(l-(5-(Trifluoromethyl)-l,2,4-oxadiazol-3- yl)piperidin-4-yloxy)cyclohexyloxy)pyrazine-2-carbonitrile (Compound 8).
  • Example 1.9 Preparation of l-(Trifluoromethyl)cyclobutyl 4-((lr,4r)-4-(3-Cyanopyrazin- 2-yloxy)cyclohexyloxy)piperidine-l-carboxylate (Compound 9).
  • Step 1 Preparation of l-(Trifluoromethyl)cyclobutyl l//-Imidazole-l-carboxylate.
  • Step 2 Preparation of l-(Trifluoromethyl)cyclobutyl 4-((lr,4r)-4-(3-Cyanopyrazin- 2-yloxy)cyclohexyloxy)piperidine-l-carboxylate (Compound 9).
  • Step 1 Preparation of Perfluorophenyl 3-(Trifluoromethyl)oxetan-3-yl carbonate.
  • Step 2 Preparation of 3-(Trifluoromethyl)oxetan-3-yl 4-((lr,4r)-4-(3- Cyanopyrazin-2-yloxy)cyclohexyloxy)piperidine- 1-carboxylate (Compound 10).
  • Example 1.11 Preparation of l,l,l-Trifluoro-2-methylpropan-2-yl 4-((lr,4r)-4-(3- Cyanopyrazin-2-yloxy)cyclohexyloxy)piperidine- 1-carboxylate (Compound 11).
  • Step 1 Preparation of l,l,l-Trifluoro-2-methylpropan-2-yl l//-Imidazole-l- carboxylate.
  • Step 2 Preparation of l,l,l-Trifluoro-2-methylpropan-2-yl 4-((lr,4r)-4-(3- Cyanopyrazin-2-yloxy)cyclohexyloxy)piperidine- 1-carboxylate (Compound 11).
  • Example 1.12 Preparation of l,l,l,3,3,3-Hexafluoropropan-2-yl 4-((lr,4r)-4-(3- Cyanopyrazin-2-yloxy)cyclohexyloxy)piperidine- 1-carboxylate (Compound 12).
  • Example 1.13 Preparation of l-(Trifluoromethyl)cyclopentyl 4-((lr,4r)-4-(3- Cyanopyrazin-2-yloxy)cyclohexyloxy)piperidine- 1-carboxylate (Compound 13).
  • Step 1 Preparation of l-(Trifluoromethyl)cyclopentyl l//-Imidazole-l- carboxylate.
  • Step 2 Preparation of l-(Trifluoromethyl)cyclopentyl 4-((lr,4r)-4-(3- Cyanopyrazin-2-yloxy)cyclohexyloxy)piperidine- 1-carboxylate (Compound 13).
  • Example 1.14 Preparation of 3-((lr,4r)-4-(l-(5-(2-Fluoropropan-2-yl)-l,2,4-oxadiazol-3- yl)piperidin-4-yloxy)cyclohexyloxy)pyrazine-2-carbonitrile (Compound 14).
  • Step 2 Preparation of (lr,4r)-4-(l-(5-(2-Fluoropropan-2-yl)-l,2,4-oxadiazol-3- yl)piperidin-4-yloxy)cyclohexanol.
  • Step 3 Preparation of 3-((lr,4r)-4-(l-(5-(2-Fluoropropan-2-yl)-l,2,4-oxadiazol-3- yl)piperidin-4-yloxy)cyclohexyloxy)pyrazine-2-carbonitrile (Compound 14).
  • Example 1.15 Preparation of 3-((lr,4r)-4-(l-(3-(2-Fluoropropan-2-yl)-l,2,4-oxadiazol-5- yl)piperidin-4-yloxy)cyclohexyloxy)pyrazine-2-carbonitrile (Compound 15) .
  • Step 1 Preparation of (lr,4r)-4-(l-(3-(2-Fluoropropan-2-yl)-l,2,4-oxadiazol-5- yl)piperidin-4-yloxy)cyclohexanol.
  • Step 2 Preparation of 3-((lr,4r)-4-(l-(3-(2-Fluoropropan-2-yl)-l,2,4-oxadiazol-5- yl)piperidin-4-yloxy)cyclohexyloxy)pyrazine-2-carbonitrile (Compound 15).
  • Residue was purified by Biotage® column chromatography (Si0 2 , hexane/AcOEt gradient). Fractions containing product were concentrated and re-purified by HPLC (CH 3 CN/H 2 0 gradient + 0.1% TFA). Fractions containing product were partly concentrated and residue was extracted with 1 M NaHC0 3 and CH 2 C1 2 . Organic phases were dried over MgS0 4 , filtered, and concentrated to afford the title compound (125 mg, 0.290 mmol, 61.7%) as a white solid.
  • Example 1.16 Preparation of 3-((lr,4r)-4-(l-(3-(2-Fluoropropan-2-yl)-l,2,4-oxadiazol-5- yl)piperidin-4-yloxy)cyclohexyloxy)picolinonitrile (Compound 16).
  • Example 1.17 Preparation of 3-((lr,4r)-4-(l-(5-(2-Fluoropropan-2-yl)-l,2,4-oxadiazol-3- yl)piperidin-4-yloxy)cyclohexyloxy)picolinonitrile (Compound 17).
  • Example 1.18 Preparation of 3-((lr,4r)-4-(l-(5-Chloropyrimidin-2-yl)piperidin-4- yloxy)cyclohexyloxy)picolinonitrile (Compound 18).
  • Example 1.20 Preparation of (5)-l,l,l-Trifluoropropan-2-yl 4-((lr,4r)-4-(2-Cyanopyridin- 3-yloxy)cyclohexyloxy)piperidine-l-carboxylate (Compound 20).
  • Example 1.21 Preparation of 3-((lr,4r)-4-(l-(3-Isopropyl-l,2,4-oxadiazol-5-yl)piperidin-4- yloxy)cyclohexyloxy)picolinonitrile (Compound 21).
  • Example 1.22 Preparation of 1-Methylcyclopropyl 4-((lr,4r)-4-(2-Cyanopyridin-3- yloxy)cyclohexyloxy)piperidine-l-carboxylate (Compound 22).
  • Example 2 In vivo Effects of GPR119 agonists on Incretin Hormone GIP release.
  • Compounds are delivered orally via a gavage needle (p.o., volume 4 mL kg), and after 45 min a blood sample is collected to determine plasma total GIP levels.
  • the control group receive vehicle (PET: 80%PEG: 10%Ethanol: 10%Tween80TM).
  • Plasma GIP levels are determined using a Total GIP ELISA kit from Millipore.
  • GPR119 agonists have been shown to increase GIP release compared to control in this assay; see for example, WO2011/127051, WO2012/040279, and WO2012/145361 ; the disclosure of each is herein incorporated by reference in its entirety.
  • Example 3 In vivo effects of a Representative Compound of the Present Invention on
  • Glucose Homeostasis oral glucose tolerance test (oGTT) in male 129SVE mice.
  • FIG 2 With a separate group receiving vehicle (20% hydroxypropyl-beta-cyclodextrin) as control.
  • vehicle 20% hydroxypropyl-beta-cyclodextrin
  • the glucose bolus was administered.
  • Levels of blood glucose were assessed using a glucometer (One-Touch UltraTM, LifeScan) at time -30 minute (prior to compound administration), at 0 min (at time when glucose bolus was given), and at 20, 40, 60, 120 minutes post glucose bolus.
  • Plasma glucose levels are shown in Table 2.
  • the reduction in the glucose excursion area under the curve (AUC) from -30 to 120 min in GPRl 19 agonist- treated animals relative to vehicle control is shown in Figure 3.
  • the glycemic suppression or percent inhibition of glucose is also shown in Table 3.
  • Example 4 Homogeneous Time-Resolved Fluorescence (HTRF®) Assay For Direct cAMP Measurement.
  • HTRF® Homogeneous Time-Resolved Fluorescence
  • GPRl 19 agonists were evaluated in an HTRF ® cAMP detection assay according to the manufacturer's instructions (Cisbio, cAMP Dynamic 2 Assay Kit; #62AM4PEJ) using CHO-Kl cells stably expressing GPRl 19. Briefly, CHO-Kl cells were transduced with a lentiviral vector encoding the nucleotide sequence of GPRl 19 (NCBI mRNA and protein reference sequences: NM_178471.2 & NP_848566 (human) and NM_181770.1 & NP_861435.1 (rat), (GPRl 19 has also been referred to as Glucose-Dependent Insulinotropic Receptor (GDIR)). The N-terminus of the GPRl 19 nucleotide sequence was modified to replace the first methionine-coding codon with a nucleotide sequence coding for a standard, nine amino acid, hemagglutinin tag.
  • GDIR Glucose
  • test compounds were solubilized in DMSO, serially diluted in DMSO and then diluted in assay buffer before being added to the cells. Test compounds were evaluated in triplicate, using 10-point, 5-fold serial dilutions starting at 10 ⁇ . The final DMSO concentration in the assay was 0.5%.
  • the compound A r -(2-fluoro-4-(methylsulfonyl) phenyl)-6-(4-(3-isopropyl-l,2,4-oxadiazol-5-yl)piperidin-l-yl)- 5-nitropyrimidin-4-amine (WO2004/065380) was used as a positive control in each runset while assay buffer containing 0.5% DMSO was used as the negative control.
  • the HTRF® assay was used to determine EC5 0 values for GPR119 agonists.
  • Each of the Compounds 1 to 22 as shown in Table A was observed to have an hGPRl 19 EC 50 value ranging from about 33.1 nM to about 0.3 nM.
  • Each of the Compounds 1 to 22 as shown in Table A was observed to have an rGPRl 19 EC5 0 value ranging from about 155.0 nM to about 3.7 nM.

Abstract

The present invention relates to compounds of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, Formula (I), that are useful as single pharmaceutical agents or in combination with one or more additional pharmaceutical agents, such as, an inhibitor of DPP-IV, a biguanide, an alpha-glucosidase inhibitor, an insulin analogue, a sulfonylurea, an SGLT2 inhibitor, a meglitinide, a thiazolidinedione, an anti-diabetic peptide analogue, or a DGAT-1 inhibitor, in the treatment of, for example, a disorder selected from: a GPR119-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic-related disorder; type 2 diabetes; obesity; and complications related thereto.

Description

MODULATORS OF GPRl 19 AND THE TREATMENT OF
DISORDERS RELATED THERETO
FIELD OF THE INVENTION
The present invention relates to compounds of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof, that are useful as single pharmaceutical agents or in combination with one or more additional pharmaceutical agents, such as, an inhibitor of DPP-IV, a biguanide, an alpha-glucosidase inhibitor, an insulin analogue, a sulfonylurea, an SGLT2 inhibitor, a meglitinide, a thiazolidinedione, an anti-diabetic peptide analogue, or a DGAT-1 inhibitor, in the treatment of, for example, a disorder selected from: a GPRl 19-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic-related disorder; type 2 diabetes; obesity; and complications related thereto.
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 unapparent, 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 3rd 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, BMJ 310, 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 imgf000004_0001
25.0 - 29.9 Overweight
30.0 - 34.9 Obesity (Class I)
35.0 - 39.9 Obesity (Class II)
> 40 Extreme Obesity (Class III)
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 25% of all post-menopausal white women, and the proportion rises to 70% in women older than 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 mortality, 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 Osteoporosis.)
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. GPR119 GPR119 is a G protein-coupled receptor (GPR119; e.g., human GPR119, GenBank® Accession No. AAP72125 and alleles thereof; e.g., mouse GPR119, GenBank® Accession No. AY288423 and alleles thereof) and is selectively expressed on pancreatic beta cells. GPR119 activation leads to elevation of a level of intracellular cAMP, consistent with GPR119 being coupled to Gs. Agonists to GPR119 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, GPR119 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).
GPR119 agonists also stimulate the release of Glucose-dependent Insulinotropic Polypeptide (GIP), Glucagon-Like Peptide-1 (GLP-1), 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 GPR119 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-1, 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, GPR119 agonists enhance incretin release and therefore can be used in treatment of disorders related to the incretins, such as, GIP, GLP-1, and PYY. However, a number of the incretins, such as, GIP and GLP-1, 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 GPR119 agonist, (2-Fluoro-4-methanesulfonyl-phenyl)-{6-[4- (3-isopropyl-[ 1 ,2,4]oxadiazol-5-yl)-piperidin- 1 -yl] -5-nitro-pyrimidin-4-yl } -amine (see, compound Bl 11 in WO 2004/065380), and a DPP-IV inhibitor acutely increased plasma GLP-1 levels and improved glucose tolerance to a significantly greater degree than either agent alone.
G. Glucose-dependent Insulinotropic Polypeptide (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(l-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; Mcintosh 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, Mol Cell Endocrinol (2001) 177:35-41; Xie et al, Bone (2005) 3 ':759-769; and Tsukiyama et al, Mol 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-1)
Glucagon-like peptide-1 (GLP-1) is an incretin hormone derived from the
posttranslaltional modification of proglucagon and secreted by gut endocrine cells. GLP- 1 mediates its actions through a specific G protein-coupled receptor (GPCR), namely GLP-1R. 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-1 has been shown to be cardioprotective in a rat model of myocardial infarction [Bose et al, Diabetes (2005) 54: 146- 151], and GLP-1R 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 Sci (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- 1R 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 jejunum, 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 et al, 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), 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
Figure imgf000011_0001
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; Tilg 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 (2007) 115: 1408- 1416). Adiponectin has been reported to confer protection against myocardial ischemia- 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 protect against myocyte and capillary loss (Shibata et al, J. Mol. 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 Mol. 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)-a-induced expression of the adhesion molecules VCAM-1, E-selectin and ICAM- 1 in human aortic endothelial cells (HAECs) (Ouchi et al, Circulation (1999) 100:2473-2476) and to inhibit production of TNF-a 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-a in inflammation has been demonstrated by the ability of agents that block the action of TNF-a to treat a range of inflammatory conditions. TNF- a- 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, Scand J 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.
SUMMARY OF THE INVENTION
One aspect of the present invention is directed to compounds, as described herein, and pharmaceutically acceptable salts, solvates, and hydrates thereof, which bind to and modulate the activity of a GPCR, referred to herein as GPR119, and uses thereof. One aspect of the present invention encompasses, inter alia, certain cyclohexane derivatives selected from compounds of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000013_0001
(I)
wherein:
X is N or CH; and
R1 is selected from: Ci-C6 alkoxycarbonyl, C3-C7 cycloalkoxycarbonyl,
heterocyclyloxycarbonyl, and heteroaryl; each optionally substituted with one or more substituents selected from: C1-C6 alkyl, C1-C6 haloalkyl, and halogen; provided that R1 is a group other than 4-(trifluoromethyl)pyrimidin-2-yl.
One aspect of the present invention encompasses, inter alia, certain cyclohexane derivatives selected from compounds of Formula (lb) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000013_0002
(lb)
One aspect of the present invention encompasses, inter alia, certain cyclohexane derivatives selected from compounds of Formula (Id) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000013_0003
(Id)
One aspect of the present invention pertains to compositions comprising a compound of the present invention.
One aspect of the present invention pertains to methods for preparing a composition comprising the step of admixing a compound of the present invention, and a pharmaceutically acceptable carrier.
One aspect of the present invention pertains to a pharmaceutical product selected from: a pharmaceutical composition, a formulation, a unit dosage form, and a kit; each comprising a compound of the present invention. One aspect of the present invention pertains to pharmaceutical compositions comprising a compound of the present invention, and a pharmaceutically acceptable carrier.
One aspect of the present invention pertains to methods for preparing a pharmaceutical composition comprising the step of admixing a compound of the present invention, and a pharmaceutically acceptable carrier.
One aspect of the present invention pertains to pharmaceutical compositions obtained by any of the methods described herein.
One aspect of the present invention pertains to compositions comprising a compound of the present invention, and a second pharmaceutical agent.
One aspect of the present invention pertains to methods for preparing a composition comprising the step of admixing a compound of the present invention, and a second pharmaceutical agent.
One aspect of the present invention pertains to compositions obtained by any of the methods described herein.
One aspect of the present invention pertains to a pharmaceutical product selected from: a pharmaceutical composition, a formulation, a unit dosage form, a combined preparation, a twin pack, and a kit; each comprising a compound of the present invention, and a second pharmaceutical agent.
One aspect of the present invention pertains to pharmaceutical compositions comprising a compound of the present invention, a second pharmaceutical agent, and a pharmaceutically acceptable carrier.
One aspect of the present invention pertains to methods for preparing a pharmaceutical composition comprising the step of admixing a compound of the present invention, a second pharmaceutical agent, and a pharmaceutically acceptable carrier.
One aspect of the present invention pertains to methods for modulating the activity of a GPRl 19 receptor, comprising administering to an individual in need thereof, a therapeutically effective amount of: a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention.
One aspect of the present invention pertains to methods for modulating the activity of a GPRl 19 receptor, comprising prescribing to an individual in need thereof, a therapeutically effective amount of: a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention.
One aspect of the present invention pertains to the use of a compound of the present invention in the manufacture of a medicament for modulating the activity of a GPRl 19 receptor in an individual. One aspect of the present invention pertains to a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention; for use in a method of treatment of the human or animal body by therapy.
One aspect of the present invention pertains to a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention; for use in a method of modulating the activity of a GPR119 receptor in an individual.
One aspect of the present invention pertains to methods for modulating the activity of a GPR119 receptor, comprising administering to an individual in need thereof, a therapeutically effective amount of a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention; each in combination with a therapeutically effective amount of a second pharmaceutical agent.
One aspect of the present invention pertains to methods for modulating the activity of a GPR119 receptor, comprising prescribing to an individual in need thereof, a therapeutically effective amount of a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention; each in combination with a therapeutically effective amount of a second pharmaceutical agent.
One aspect of the present invention pertains to the use of a compound of the present invention in combination with a second pharmaceutical agent in the manufacture of a medicament for modulating the activity of a GPR119 receptor in an individual.
One aspect of the present invention pertains to the use of a pharmaceutical agent in combination with a compound of the present invention, in the manufacture of a medicament for modulating the activity of a GPR119 receptor in an individual.
One aspect of the present invention pertains to a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention; each in combination with a second pharmaceutical agent for use in a method of treatment of the human or animal body by therapy.
One aspect of the present invention pertains to a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention; each in combination with a second pharmaceutical agent for use in a method of modulating the activity of a GPR119 receptor in an individual.
One aspect of the present invention pertains to a pharmaceutical agent in combination with a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention; for use in a method of treatment of the human or animal body by therapy.
One aspect of the present invention pertains to a pharmaceutical agent in combination with a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention; for use in modulating the activity of a GPRl 19 receptor in an individual.
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is selected from: a DPP-IV inhibitor, a biguanide, an alpha-glucosidase inhibitor, an insulin analogue, a sulfonylurea, an SGLT2 inhibitor, a meglitinide, a thiazolidinedione, an antidiabetic peptide analogue, and a DGAT-1 inhibitor.
In some embodiments, modulating the activity of a GPRl 19 receptor is agonizing a GPRl 19 receptor. In some embodiments, agonizing a GPRl 19 receptor is selected from:
increasing the secretion of an incretin in an individual; increasing a blood incretin level in an individual; a GPRl 19-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic-related disorder; and obesity.
These and other aspects of the invention disclosed herein will be set forth in greater detail as the patent disclosure proceeds.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 shows the in vivo effects of Compound 7 on glucose homeostasis in male
129SVE mice (oral glucose tolerance test (oGTT)).
Figure 2 shows the in vivo effects of Compound 10 on glucose homeostasis in male
129SVE mice (oral glucose tolerance test (oGTT)).
Figure 3 shows the in vivo effects of Compound 7 and Compound 10 on glucose excursion in terms of percent glycemic inhibition (i.e., percent AUC reduction) in male 129SVE mice.
Figure 4 shows a general synthetic scheme for the preparation of intermediates useful in preparing compounds of Formula (I).
Figure 5 shows a general synthetic scheme for the preparation of compounds of Formula (I) and intermediates of the present invention.
Figure 6 shows a general synthetic scheme for the preparation of compounds of Formula (I) wherein R1 is oxadiazole optionally substituted with one or more substituents selected from: Ci-C6 alkyl, Ci-C6 haloalkyl, and halogen {i.e., R is selected from H, Ci-C6 alkyl, C1-C6 haloalkyl, and halogen).
Figure 7 shows a general synthetic scheme for the preparation of compounds of Formula (I) wherein R1 is Ci-C6 alkoxycarbonyl, C3-C7 cycloalkoxycarbonyl, or heterocyclyloxycarbonyl each optionally substituted with one or more substituents selected from: C1-C6 alkyl, C1-C6 haloalkyl, and halogen (i.e., Rb is C1-C6 alkyl, C3-C7 cycloalkyl, or heterocyclyl each optionally substituted with one or more substituents selected from: C1-C6 alkyl, C1-C6 haloalkyl, and halogen); and wherein R1 is heteroaryl optionally substituted with one or more substituents selected from: C1-C6 alkyl, C1-C6 haloalkyl, and halogen.
DETAILED DESCRIPTION OF THE INVENTION
DEFINITIONS
For clarity and consistency, the following definitions will be used throughout this patent document.
The term "agonist" as used herein refers to a moiety that interacts with and activates a G-protein-coupled receptor, for instance a GPR119-receptor, and can thereby initiate a physiological or pharmacological response characteristic of that receptor. For example, an agonist may activate an intracellular response upon binding to a receptor, or enhance GTP binding to a membrane.
The term "antagonist" as used herein refers to a moiety that competitively binds to the receptor at the same site as an agonist (for example, the endogenous ligand), but which does not activate the intracellular response initiated by the active form of the receptor and can thereby inhibit the intracellular responses by an agonist or partial agonist. An antagonist does not diminish the baseline intracellular response in the absence of an agonist or partial agonist.
The term "GPR119" as used herein includes the human amino acid sequences found in GenBank® accession number AY288416, and naturally-occurring allelic variants thereof, and mammalian orthologs thereof. A human GPR119 for use in screening and testing of the compounds of the invention is provided in the nucleotide sequence of Seq. ID.No: l and the corresponding amino acid sequence in Seq. ID.No:2 found in PCT Application No.
WO2005/007647.
The term "hydrate" as used herein refers to 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 term "solvate" as used herein refers to 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 "in need of treatment" and the term "in need thereof" when referring to treatment are used interchangeably and refer to 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" refers to any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates, and most preferably humans.
The term "inverse agonist" refers to a moiety that binds to the endogenous form of the receptor or to the constitutively activated form of the receptor and which inhibits the baseline intracellular response initiated by the active form of the receptor below the normal base level of activity which is observed in the absence of an agonist or partial agonist, or decreases GTP binding to a membrane. Preferably, the baseline intracellular response is inhibited in the presence of the inverse agonist by at least 30%, more preferably by at least 50% and most preferably by at least 75%, as compared with the baseline response in the absence of the inverse agonist.
The term "modulate or modulating" refers to an increase or decrease in the amount, quality, response or effect of a particular activity, function or molecule.
The term "pharmaceutical composition" refers to 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 "therapeutically effective amount" refers to 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 by 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).
The term "unit dosage form" refers to a single dose form which is capable of being administered to a subject, and which can be readily handled and packaged, remaining as a physically and chemically stable unit comprising a compound of the present invention (i.e., compound used neat, such as in a dry powder inhaler (DPI) consisting of neat drug substance or for use in sublingual or buccal administration) or a pharmaceutically acceptable composition comprising a compound of the present invention such as in a pill, capsule, tablet, and the like.
CHEMICAL GROUP, MOIETY OR RADICAL
The term "C1-C6 alkoxy" refers to a radical comprising a C1-C6 alkyl group attached directly to an oxygen atom, wherein Ci-C6 alkyl has the same definition as found herein. Some embodiments contain 1 to 5 carbons. Some embodiments contain 1 to 4 carbons. Some embodiments contain 1 to 3 carbons. Some embodiments contain one or two carbons. Examples of an alkoxy group include, but are not limited to methoxy, ethoxy, w-propoxy, isopropoxy, n- butoxy, i-butoxy, isobutoxy, s-butoxy, and the like.
The term "Ci-C6 alkoxycarbonyl" refers to a radical consisting of a Ci-C6 alkoxy group attached to a carbonyl group, wherein Ci-C6 alkoxy has the same definition as found herein. Examples include, but are not limited to, methoxycarbonyl, ethoxycarbonyl, propoxycarbonyl, isopropoxycarbonyl, butoxycarbonyl, butoxycarbonyl, isobutoxycarbonyl, and tert- butoxycarbonyl.
The term "Ci-C6 alkyl" refers to a straight or branched carbon radical containing 1 to 6 carbons. Some embodiments contain 1 to 5 carbons. Some embodiments contain 1 to 4 carbons. Some embodiments contain 1 to 3 carbons. Some embodiments contain one or two carbons. Examples of an alkyl group include, but are not limited to, methyl, ethyl, n -propyl, isopropyl, n- butyl, i-butyl, isobutyl, i-butyl, pentyl, isopentyl, i-pentyl, neopentyl, 1-methylbutyl [i.e., -CH(CH3)CH2CH2CH3], 2-methylbutyl [i.e. , -CH2CH(CH3)CH2CH3], n-hexyl, and the like.
The term "carbonyl" refers to a C=0 group.
The term "cyano" refers to the group -CN.
The term "C3-C7 cycloalkoxycarbonyl" refers to a radical consisting of a C3-C7 cycloalkoxy group attached to a carbonyl group, wherein C3-C7 cycloalkoxy has the same definition as found herein. Examples include, but are not limited to, cyclopropoxycarbonyl, cyclobutoxycarbonyl, cyclopentyloxycarbonyl, cyclohexyloxycarbonyl, and
cycloheptyloxycarbonyl.
The term "C3-C7 cycloalkoxy" refers to a radical comprising a C3-C7 cycloalkyl group attached directly to an oxygen atom, wherein C3-C7 cycloalkyl has the same definition as found herein. Some embodiments contain 3 to 6 carbons. Some embodiments contain 3 to 5 carbons. Some embodiments contain 3 to 4 carbons. Examples of a cycloalkoxy group include, but are not limited to cyclopropoxy, cyclobutoxy, cyclopentyloxy, cyclohexyloxy, and cycloheptyloxy.
The term "C3-C7 cycloalkyl" refers to a saturated ring radical containing 3 to 7 ring carbons. Some embodiments contain 3 to 4 carbons. Some embodiments contain 3 to 5 carbons. Some embodiments contain 4 to 6 carbons. Some embodiments contain 5 to 6 carbons.
Examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cycloheptyl.
The term "Ci-C6 haloalkyl" refers to a radical comprising a Ci-C6 alkyl group substituted with one or more halogens, wherein Ci-C6 alkyl has the same definition as found herein. The Ci-C6 haloalkyl may be fully substituted in which case it can be represented by the formula CqL2q+i, wherein L is a halogen and "q" is 1, 2, 3, 4, 5 or 6. When more than one halogen is present then they may be the same or different and selected from: fluorine, chlorine, bromine, and iodine. In some embodiments, haloalkyl contains 1 to 5 carbons. In some embodiments, haloalkyl contains 1 to 4 carbons. In some embodiments, haloalkyl contains 1 to 3 carbons. In some embodiments, haloalkyl contains one or two carbons. Examples of a haloalkyl group include, but are not limited to, fluoromethyl, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, pentafluoroethyl, 2-fluoropropan-2-yl, 1,1-difluoropropyl, 1,3- difluoropropan-2-yl, (S)- l-fluoropropan-2-yl, (R)-l-fluoropropan-2-yl, l, l, l-trifluoropropan-2- yl, l, l, l,3,3,3-hexafluoropropan-2-yl, and the like.
The term "halogen" refers to a fluoro, chloro, bromo or iodo group.
The term "heteroaryl" refers to a ring system containing 5 to 10 ring atoms, that may contain a single ring or two fused rings, and wherein at least one ring is aromatic and at least one ring atom of the aromatic ring is a heteroatom selected from, for example: O, S and N, wherein N is optionally substituted with H, C1-C4 acyl, C1-C4 alkyl, or O (i.e., forming an N- oxide) and S is optionally substituted with one or two oxygens. In some embodiments, the aromatic ring contains one heteroatom. In some embodiments, the aromatic ring contains two heteroatoms. In some embodiments, the aromatic ring contains three heteroatoms. Examples include furanyl, thienyl, pyrrolyl, imidazolyl, oxazolyl, thiazolyl, isoxazolyl, pyrazolyl, isothiazolyl, oxadiazolyl, triazolyl, tetrazolyl, thiadiazolyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, quinolinyl, isoquinolinyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, indolyl, isoindolyl, indazolyl, indolizinyl, purinyl, naphthyridinyl, pteridinyl, carbazolyl, acridinyl. phenazinyl, phenothiazinyl, phenoxazinyl, benzoxazolyl, benzothiazolyl, 1/i-benzimidazolyl, imidazopyridinyl, benzothienyl, benzofuranyl, isobenzofuran, 2,3- dihydrobenzofuranyl, 4/i-benzo[l,3]dioxinyl, 3,4-dihydro-l/i-isoquinolinyl, 1,4,6,7-tetrahydro- imidazo[4,5-c]pyridinyl, 7, 8-dihydro-5/i-[ 1,6] naphthyridinyl, 5,6-dihydro-8/i- [l,2,4]triazolo[4,3-a]pyrazinyl, benzo[l,3]dioxolyl, pyrazolo[l,5-a]pyrimidinyl, 1,2,3,4- tetrahydroquinolinyl, and the like. Some embodiments are directed to 5-membered heteroaryl rings. Examples of a 5-membered heteroaryl ring include furanyl, thienyl, pyrrolyl, imidazolyl, oxazolyl, thiazolyl, isoxazolyl, pyrazolyl, isothiazolyl, oxadiazolyl, triazolyl, tetrazolyl, thiadiazolyl, and the like. Some embodiments are directed to 6-membered heteroaryl rings. Examples of a 6-membered heteroaryl ring include pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, and the like. Some embodiments are directed to 8 to 10-membered heteroaryl rings. Examples of a 8 to 10-membered heteroaryl ring include quinolinyl, isoquinolinyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, indolyl, isoindolyl, indazolyl, indolizinyl, purinyl, naphthyridinyl, pteridinyl, carbazolyl, acridinyl. phenazinyl,
phenothiazinyl, phenoxazinyl, benzoxazolyl, benzothiazolyl, 1/i-benzimidazolyl,
imidazopyridinyl, benzothienyl, benzofuranyl, isobenzofuran, 2,3-dihydrobenzofuranyl, 4H- benzo[l,3]dioxinyl, 3,4-dihydro-l/i-isoquinolinyl, l,4,6,7-tetrahydro-imidazo[4,5-c]pyridinyl, 7,8-dihydro-5/i-[l,6]naphthyridinyl, 5,6-dihydro-8/i-[l,2,4]triazolo[4,3-a]pyrazinyl, benzo[l,3]dioxolyl, pyrazolo[l,5-a]pyrimidinyl, 1,2,3,4-tetrahydroquinolinyl, and the like.
The term "heterocyclyl" refers to a non-aromatic ring radical containing 3 to 10 ring atoms, wherein one, two or three ring atoms are heteroatoms selected independently from, for example: O, S, and N, wherein N is optionally substituted with H, C1-C4 acyl or C1-C4 alkyl; and S is optionally substituted with one or two oxygens. Examples of a heterocyclyl group include, but are not limited to, aziridinyl, azetidinyl, piperidinyl, morpholinyl, piperazinyl, pyrrolidinyl, [l,3]-dioxolanyl, thiomorpholinyl, [l,4]oxazepanyl, 1, 1-dioxothiomorpholinyl, azepanyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydrothiopyranyl, l-oxo-hexahydro-^4-thiopyranyl, l, l-dioxo-hexahydro-^6-thiopyranyl, oxetanyl, and azabicyclo[3.2.1]octanyl. In some embodiments "heterocyclyl" refers to piperidin-4-yl, 3-azabicyclo[3.2.1]octan-8-yl, and 8- azabicyclo[3.2.1] octan- 3 - y 1.
The term "heterocyclyloxy" refers to a radical comprising a heterocyclyl group attached to an oxygen radical, wherein heterocyclyl has the same definition as described herein.
The term "heterocyclyloxycarbonyl" refers to a radical comprising a heterocyclyloxy group attached to a carbonyl group, wherein heterocyclyloxy has the same definition as described herein.
COMPOUNDS OF THE INVENTION
One aspect of the present invention provides, inter alia, compounds selected from compounds of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000021_0001
(I)
wherein X and R1 have the same definitions as described herein, supra and infra. One aspect of the present invention provides, inter alia, compounds selected from compounds of Formula (lb) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000022_0001
(lb)
wherein X and R1 have the same definitions as described herein, supra and infra.
One aspect of the present invention provides, inter alia, compounds selected from compounds of Formula (Id) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000022_0002
(Id)
wherein X and R1 have the same definitions as described herein, supra and infra.
One aspect of the present invention provides, inter alia, compounds selected from compounds of Formulae (I), (lb), and (Id), and pharmaceutically acceptable salts, solvates, and hydrates thereof; wherein X and R1 have the same definitions as described herein, supra and infra.
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. All combinations of the embodiments pertaining to the chemical groups represented by the variables {e.g., X and R1) contained within the generic chemical formulae described herein, for example, (I), (lb), and (Id) are specifically embraced by the present invention just as if each and every combination was individually and explicitly recited, to the extent that such combinations embrace compounds that result in stable compounds {i.e., compounds that can be isolated, characterized and tested for biological activity). In addition, all subcombinations of the chemical groups listed in the embodiments describing such variables, as well as all subcombinations of methods, uses, compounds, and medical indications as described herein, are also specifically embraced by the present invention just as if each and every subcombination of chemical groups and subcombination of methods, uses, compounds, and medical indications were individually and explicitly recited herein. In addition, some embodiments include every combination of one or more pharmaceutical agents, such as an inhibitor of DPP-IV, a biguanide, an alpha-glucosidase inhibitor, and the like, either specifically disclosed herein or specifically disclosed in any reference recited herein just as if each and every combination was individually and explicitly recited. Still further, some embodiments of the present invention include every combination of one or more embodiments pertaining to the chemical groups represented by the variables and generic chemical formulae as described herein or every combination of one or more compounds of Formula (I), and related Formulae, together/in combination with every combination of one or more pharmaceutical agents, such as an inhibitor of DPP-IV, a biguanide, an alpha-glucosidase inhibitor, a DGAT- 1 inhibitor, and the like, either specifically disclosed herein or specifically disclosed in any reference recited herein just as if each and every combination was individually and explicitly recited.
As used herein, "substituted" indicates that at least one hydrogen atom of the chemical group is replaced by a non-hydrogen substituent or group, the non-hydrogen substituent or group can be monovalent or divalent. When the substituent or group is divalent, then it is understood that this group is further substituted with another substituent or group. When a chemical group herein is "substituted" it may have up to the full valance of substitution; for example, a methyl group can be substituted by 1, 2, or 3 substituents, a methylene group can be substituted by one or two substituents, a phenyl group can be substituted by 1, 2, 3, 4, or 5 substituents, a naphthyl group can be substituted by 1, 2, 3, 4, 5, 6, or 7 substituents, and the like. Likewise, "substituted with one or more substituents" refers to the substitution of a group with one substituent up to the total number of substituents physically allowed by the group. Further, when a group is substituted with more than one group they can be identical or they can be different.
Compounds of the invention can also include tautomeric forms, such as keto-enol tautomers and the like. Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution. It is understood that the various tautomeric forms are within the scope of the compounds of the present invention.
It is understood and appreciated that compounds of Formula (I) and formulae related thereto may have one or more chiral centers and therefore can exist as enantiomers and/or diastereoisomers. The invention is understood to extend to and embrace all such enantiomers, diastereoisomers and mixtures thereof, including but not limited to racemates. It is understood that compounds of Formula (I) and formulae used throughout this disclosure represent all individual enantiomers and mixtures thereof, unless stated or shown otherwise.
It is understood and appreciated that compounds of Formula (I) and formulae related thereto exist as the trans meso isomers. The trans meso isomers of compounds of the present invention are named herein using the designation (lr,4r) as shown below:
Figure imgf000024_0001
(I)
The Group X:
In some embodiments, X is N or CH.
In some embodiments, X is N.
In some embodiments, X is CH.
The Group R1:
In some embodiments, R1 is selected from: Ci-C6 alkoxycarbonyl, C3-C7
cycloalkoxycarbonyl, heterocyclyloxycarbonyl, and heteroaryl; each optionally substituted with one or more substituents selected from: C1-C6 alkyl, C1-C6 haloalkyl, and halogen; provided that R1 is a group other than 4-(trifluoromethyl -2-yl:
Figure imgf000024_0002
In some embodiments, R1 is selected from: C1-C6 alkoxycarbonyl, C3-C7
cycloalkoxycarbonyl, heterocyclyloxycarbonyl, and heteroaryl; each optionally substituted with one or more substituents selected from: chloro, fluoro, 2-fluoropropan-2-yl, isopropyl, methyl, 2,2,2-trifluoroethyl, and trifluoromethyl.
In some embodiments, R1 is selected from: ieri-butoxycarbonyl, isopropoxycarbonyl, cyclobutoxycarbonyl, cyclopentyloxycarbonyl, cyclopropoxycarbonyl, (pyrrolidin-3- yloxy)carbonyl, 1,2,4-oxadiazolyl, (oxetan-3-yloxy)carbonyl, and pyrimidin-2-yl; each optionally substituted with one or more substituents selected from: Ci-C6 alkyl, Ci-C6 haloalkyl, and halogen.
In some embodiments, R1 is selected from: ieri-butoxycarbonyl, isopropoxycarbonyl, cyclobutoxycarbonyl, cyclopentyloxycarbonyl, cyclopropoxycarbonyl, (pyrrolidin-3- yloxy)carbonyl, 1,2,4-oxadiazolyl, (oxetan-3-yloxy)carbonyl, and pyrimidin-2-yl; each optionally substituted with one or more substituents selected from: chloro, fluoro, 2- fluoropropan-2-yl, isopropyl, methyl, 2,2,2-trifluoroethyl, and trifluoromethyl.
In some embodiments, R1 is selected from: C1-C6 alkoxycarbonyl optionally substituted with one or more halogen substituents.
In some embodiments, R1 is selected from: C1-C6 alkoxycarbonyl optionally substituted with one or more fluoro substituents. In some embodiments, R1 is selected from: ieri-butoxycarbonyl and
isopropoxycarbonyl; each optionally substituted with one or more halogen substituents.
In some embodiments, R1 is selected from: ieri-butoxycarbonyl and
isopropoxycarbonyl; each optionally substituted with one or more fluoro substituents.
In some embodiments, R1 is selected from: C3-C7 cycloalkoxycarbonyl optionally substituted with one or more substituents selected from: C1-C6 alkyl and C1-C6 haloalkyl.
In some embodiments, R1 is selected from: C3-C7 cycloalkoxycarbonyl optionally substituted with one or more substituents selected from: methyl and trifluoromethyl.
In some embodiments, R1 is selected from: cyclobutoxycarbonyl,
cyclopentyloxycarbonyl, and cyclopropoxycarbonyl; each optionally substituted with one or more substituents selected from: C1-C6 alkyl and C1-C6 haloalkyl.
In some embodiments, R1 is selected from cyclobutoxycarbonyl,
cyclopentyloxycarbonyl, and cyclopropoxycarbonyl; each optionally substituted with one or more substituents selected from: methyl and trifluoromethyl.
In some embodiments, R1 is selected from: heterocyclyloxycarbonyl optionally substituted with one or more C1-C6 haloalkyl substituents.
In some embodiments, R1 is selected from: heterocyclyloxycarbonyl optionally substituted with one or more substituents selected from: 2,2,2-trifluoroethyl and trifluoromethyl.
In some embodiments, R1 is selected from: (pyrrolidin-3-yloxy)carbonyl and (oxetan-3- yloxy)carbonyl; each optionally substituted with one or more C1-C6 haloalkyl substituents.
In some embodiments, R1 is selected from: (pyrrolidin-3-yloxy)carbonyl and (oxetan-3- yloxy)carbonyl; each optionally substituted with one or more substituents selected from: 2,2,2- trifluoroethyl and trifluoromethyl.
In some embodiments, R1 is selected from: heteroaryl optionally substituted with one or more substituents selected from: C1-C6 alkyl, C1-C6 haloalkyl, and halogen.
In some embodiments, R1 is selected from: heteroaryl optionally substituted with one or more substituents selected from: chloro, 2-fluoropropan-2-yl, isopropyl, and trifluoromethyl.
In some embodiments, R1 is selected from: 1,2,4-oxadiazolyl, and pyrimidin-2-yl; each optionally substituted with one or more substituents selected from: C1-C6 alkyl, C1-C6 haloalkyl, and halogen.
In some embodiments, R1 is selected from: 1,2,4-oxadiazolyl, and pyrimidin-2-yl; each optionally substituted with one or more substituents selected from: chloro, 2-fluoropropan-2-yl, isopropyl, and trifluoromethyl.
In some embodiments, R1 is selected from: (l-methylcyclopropoxy)carbonyl, 5- chloropyrimidin-2-yl, (1,1,1 -trifluoropropan-2-yloxy)carbonyl, ( 1 - (2,2,2- trifluoroethyl)pyrrolidin-3-yloxy)carbonyl, 3-(trifluoromethyl)- 1 ,2,4-oxadiazol-5-yl, 5- (trifluoromethyl)- 1 ,2,4-oxadiazol-3-yl, ( 1 -(trifluoromethyl)cyclobutoxy)carbonyl, (3- (trifluoromethyl)oxetan-3-yloxy)carbonyl, (1,1,1 -trifluoro-2-methylpropan-2-yloxy)carbonyl, (l,l,l,3,3,3-hexafluoropropan-2-yloxy)carbonyl, (l-(trifluoromethyl)cyclopentyloxy)carbonyl, 5-(2-fluoropropan-2-yl)-l,2,4-oxadiazol-3-yl, 3-(2-fluoropropan-2-yl)-l,2,4-oxadiazol-5-yl, and 3-isopropyl-l,2,4-oxadiazol-5-yl.
In some embodiments, R1 is (l-methylcyclopropoxy)carbonyl. In some embodiments, R1 is 5-chloropyrimidin-2-yl. In some embodiments, R1 is ( 1,1,1 -trifluoropropan-2- yloxy)carbonyl. In some embodiments, R1 is (l-(2,2,2-trifluoroethyl)pyrrolidin-3- yloxy)carbonyl. In some embodiments, R1 is 3-(trifluoromethyl)-l,2,4-oxadiazol-5-yl. In some embodiments, R1 is 5-(trifluoromethyl)-l,2,4-oxadiazol-3-yl. In some embodiments, R1 is (1- (trifluoromethyl)cyclobutoxy)carbonyl. In some embodiments, R1 is (3-(trifluoromethyl)oxetan- 3-yloxy)carbonyl. In some embodiments, R1 is ( 1,1,1 -trifluoro-2-methylpropan-2- yloxy)carbonyl. In some embodiments, R1 is (l,l,l,3,3,3-hexafluoropropan-2-yloxy)carbonyl. In some embodiments, R1 is (l-(trifluoromethyl)cyclopentyloxy)carbonyl. In some embodiments, R1 is 5-(2-fluoropropan-2-yl)-l,2,4-oxadiazol-3-yl. In some embodiments, R1 is 3-(2- fluoropropan-2-yl)-l,2,4-oxadiazol-5-yl. In some embodiments, R1 is 3-isopropyl- 1,2,4- oxadiazol-5-yl.
Certain Combinations:
One aspect of the present invention encompasses certain cyclohexyl derivatives selected from compounds of Formula (lb) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000026_0001
(lb)
wherein:
R1 is selected from: Ci-C6 alkoxycarbonyl, C3-C7 cycloalkoxycarbonyl,
heterocyclyloxycarbonyl, and heteroaryl; each optionally substituted with one or more substituents selected from: chloro, fluoro, 2-fluoropropan-2-yl, methyl, 2,2,2-trifluoroethyl, and trifluoromethyl.
One aspect of the present invention encompasses certain cyclohexyl derivatives selected from compounds of Formula (lb) and pharmaceutically acceptable salts, solvates, and hydrates thereof, wherein:
R1 is selected from: ieri-butoxycarbonyl, isopropoxycarbonyl, cyclobutoxycarbonyl, cyclopentyloxycarbonyl, cyclopropoxycarbonyl, (pyrrolidin-3-yloxy)carbonyl, 1,2,4- oxadiazolyl, (oxetan-3-yloxy)carbonyl, and pyrimidin-2-yl; each optionally substituted with one or more substituents selected from: Ci-C6 alkyl, Ci-C6 haloalkyl, and halogen. One aspect of the present invention encompasses certain cyclohexyl derivatives selected from compounds of Formula (lb) and pharmaceutically acceptable salts, solvates, and hydrates thereof, wherein:
R1 is selected from: ieri-butoxycarbonyl, isopropoxycarbonyl, cyclobutoxycarbonyl, cyclopentyloxycarbonyl, cyclopropoxycarbonyl, (pyrrolidin-3-yloxy)carbonyl, 1,2,4- oxadiazolyl, (oxetan-3-yloxy)carbonyl, and pyrimidin-2-yl; each optionally substituted with one or more substituents selected from: chloro, fluoro, 2-fluoropropan-2-yl, methyl, 2,2,2- trifluoroethyl, and trifluoromethyl.
One aspect of the present invention encompasses certain cyclohexyl derivatives selected from compounds of Formula (lb) and pharmaceutically acceptable salts, solvates, and hydrates thereof, wherein:
R1 is selected from: (l-methylcyclopropoxy)carbonyl, 5-chloropyrimidin-2-yl, (1,1,1- trifluoropropan-2-yloxy)carbonyl, (l-(2,2,2-trifluoroethyl)pyrrolidin-3-yloxy)carbonyl, 3- (trifluoromethyl)-l,2,4-oxadiazol-5-yl, 5-(trifluoromethyl)-l,2,4-oxadiazol-3-yl, (1- (trifluoromethyl)cyclobutoxy)carbonyl, (3-(trifluoromethyl)oxetan-3-yloxy)carbonyl, (1,1,1- trifluoro-2-methylpropan-2-yloxy)carbonyl, (l,l,l,3,3,3-hexafluoropropan-2-yloxy)carbonyl, (l-(trifluoromethyl)cyclopentyloxy)carbonyl, 5-(2-fluoropropan-2-yl)-l,2,4-oxadiazol-3-yl, and 3-(2-fluoropropan-2-yl)-l,2,4-oxadiazol-5-yl.
One aspect of the present invention encompasses certain cyclohexyl derivatives selected from compounds of Formula (Id) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000027_0001
(W)
wherein:
R1 is selected from: Ci-C6 alkoxycarbonyl, C3-C7 cycloalkoxycarbonyl, and heteroaryl; each optionally substituted with one or more substituents selected from: chloro, fluoro, 2- fluoropropan-2-yl, isopropyl, and methyl.
One aspect of the present invention encompasses certain cyclohexyl derivatives selected from compounds of Formula (Id) and pharmaceutically acceptable salts, solvates, and hydrates thereof, wherein:
R1 is selected from: isopropoxycarbonyl, cyclopropoxycarbonyl, 1,2,4-oxadiazolyl, and pyrimidin-2-yl; each optionally substituted with one or more substituents selected from: Ci-C6 alkyl, Ci-C6 haloalkyl, and halogen. One aspect of the present invention encompasses certain cyclohexyl derivatives selected from compounds of Formula (Id) and pharmaceutically acceptable salts, solvates, and hydrates thereof, wherein:
R1 is selected from: isopropoxycarbonyl, cyclopropoxycarbonyl, 1,2,4-oxadiazolyl, and pyrimidin-2-yl; each optionally substituted with one or more substituents selected from: chloro, fluoro, 2-fluoropropan-2-yl, isopropyl, and methyl.
One aspect of the present invention encompasses certain cyclohexyl derivatives selected from compounds of Formula (Id) and pharmaceutically acceptable salts, solvates, and hydrates thereof, wherein:
R1 is selected from: (l-methylcyclopropoxy)carbonyl, 5-chloropyrimidin-2-yl, (1,1, 1- trifluoropropan-2-yloxy)carbonyl, 5-(2-fluoropropan-2-yl)-l,2,4-oxadiazol-3-yl, 3-(2- fluoropropan-2-yl)-l,2,4-oxadiazol-5-yl, and 3-isopropyl-l,2,4-oxadiazol-5-yl.
Some embodiments of the present invention include every combination of one or more compounds selected from the following group shown in Table A.
Table A
Figure imgf000028_0001
Figure imgf000029_0001
Figure imgf000030_0001
Additionally, individual compounds and chemical genera of the present invention, for example those compounds found in Table A including, isomers, diastereoisomers and enantiomers thereof, encompass all pharmaceutically acceptable salts, solvates, and hydrates, thereof. Further, mesoisomers of individual compounds and chemical genera of the present invention, for example those compounds found in Table A, encompass all pharmaceutically acceptable salts, solvates and particularly hydrates, thereof.
The compounds of the Formula (I) and genera related thereto may be prepared according to relevant published literature procedures that are used by one skilled in the art. Exemplary reagents and procedures for these reactions appear hereinafter in the working Examples. Protection and deprotection may be carried out by procedures generally known in the art (see, for example, Greene, T. W. and Wuts, P. G. M., Protecting Groups in Organic Synthesis, 3rd Edition, 1999 [Wiley]).
It is understood that the present invention embraces, each isomer, each diastereoisomer, each enantiomer and mixtures thereof of each compound and generic formulae disclosed herein just as if they were each individually disclosed with the specific stereochemical designation for each chiral carbon. Separation of the individual isomers and enantiomers (such as, by chiral HPLC, recrystallization of diastereoisomeric mixtures and the like) or selective synthesis (such as, by enantiomeric selective syntheses and the like) of the individual isomers can be accomplished by the application of various methods which are well known to practitioners in the art.
Certain Embodiments: Compositions, Methods, Indications, Pharmaceutical Products, Combinations, and Uses of Compounds of the Present Invention.
In addition to the foregoing, without limitation, certain other embodiments are described and provided below.
Certain Compositions of the Present Invention:
One aspect of the present invention pertains to compositions comprising a compound of the present invention. One aspect of the present invention pertains to pharmaceutical products selected from: a pharmaceutical composition, a formulation, a unit dosage form, and a kit; each comprising a compound of the present invention. One aspect of the present invention pertains to pharmaceutical compositions comprising a compound of the present invention, and a pharmaceutically acceptable carrier. One aspect of the present invention pertains to methods for preparing a pharmaceutical composition comprising the step of admixing a compound of the present invention, and a pharmaceutically acceptable carrier; some embodiments pertain to pharmaceutical compositions obtained by any of the methods described herein. One aspect of the present invention pertains to compositions comprising a compound of the present invention, and a second pharmaceutical agent.
In any of the embodiments that recites the terms "a pharmaceutical agent" and "a second pharmaceutical agent", it is appreciated that these terms in some aspects be further limited to a pharmaceutical agent that is not a compound of Formula (I) or a compounds related thereto. It is understood that the terms "a pharmaceutical agent" and "a second pharmaceutical agent" may refer to a pharmaceutical agent that is not detectable or has an EC50 that is greater than a value selected from: 50 μΜ, 10 μΜ, 1 μΜ, and 0.1 μΜ in a GPRl 19 receptor activity assay as described in Example 4.
One aspect of the present invention pertains to methods for preparing a composition comprising the step of admixing a compound of the present invention, and a second pharmaceutical agent; some embodiments pertain to compositions obtained by any of the methods described herein. One aspect of the present invention pertains to pharmaceutical products selected from: a pharmaceutical composition, a formulation, a unit dosage form, a combined preparation, a twin pack, and a kit; each comprising a compound of the present invention, and a second pharmaceutical agent. One aspect of the present invention pertains to pharmaceutical compositions comprising a compound of the present invention, a second pharmaceutical agent, and a pharmaceutically acceptable carrier. One aspect of the present invention pertains to methods for preparing a pharmaceutical composition comprising the step of admixing a compound of the present invention, a second pharmaceutical agent, and a pharmaceutically acceptable carrier; some embodiments pertain to pharmaceutical compositions obtained by any of the methods described herein.
Certain Methods, Pharmaceutical Products, Combinations, and Uses of the Present Invention:
One aspect of the present invention pertains to methods selected from one or more of the following: 1) for modulating the activity of a GPRl 19 receptor; 2) for agonizing a GPRl 19 receptor; 3) for increasing the secretion of an incretin in an individual; 4) increasing a blood incretin level in an individual; and 5) for treating/treatment of a disorder selected from: a GPR119-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic-related disorder; type 2 diabetes; and obesity; in an individual; comprising: A) administering to an individual in need thereof or B) prescribing to an individual in need thereof, a therapeutically effective amount of: a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention; each optionally in combination with a therapeutically effective amount of a second pharmaceutical agent.
Some embodiments pertain to methods comprising administering to an individual in need thereof a therapeutically effective amount of: a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention. Some embodiments pertain to methods for prescribing to an individual in need thereof, a therapeutically effective amount of: a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention.
One aspect of the present invention pertains to the use of a compound of the present invention, optionally in combination with a second pharmaceutical agent, in the manufacture of a medicament, selected from one or more of the following: 1) for modulating the activity of a GPR119 receptor in an individual; 2) for agonizing a GPR119 receptor; 3) for increasing the secretion of an incretin in an individual; 4) increasing a blood incretin level in an individual; and 5) for treating/treatment of a disorder selected from: a GPR119-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic- related disorder; type 2 diabetes; and obesity.
One aspect of the present invention pertains to a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention; optionally in combination with a second pharmaceutical agent, for use in a method of treatment of the human or animal body by therapy.
One aspect of the present invention pertains to one or more of the following: methods of the present invention, compounds of the present invention; compositions of the present invention; pharmaceutical products of the present invention; and pharmaceutical compositions of the present invention; optionally in combination with a second pharmaceutical agent, for use in a method selected from one or more of the following: 1) for modulating the activity of a GPR119 receptor; 2) for agonizing a GPR119 receptor; 3) for increasing the secretion of an incretin in an individual; 4) increasing a blood incretin level in an individual; and 5) for treating/ treatment of a disorder selected from: a GPR119-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic- related disorder; type 2 diabetes; and obesity.
One aspect of the present invention pertains to a pharmaceutical agent in combination with a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention; for use in a method of treatment of the human or animal body by therapy.
One aspect of the present invention pertains to a pharmaceutical agent in combination with a compound of the present invention; a composition of the present invention; a pharmaceutical product of the present invention; or a pharmaceutical composition of the present invention; for use in a method selected from one or more of the following: 1) for modulating the activity of a GPR119 receptor; 2) for agonizing a GPR119 receptor; 3) for increasing the secretion of an incretin in an individual; 4) increasing a blood incretin level in an individual; and 5) for treating/treatment of a disorder selected from: a GPR119-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic- related disorder; type 2 diabetes; and obesity.
Some embodiments pertain to methods, uses, compounds, and pharmaceutical agents, each as described herein, for modulating the activity of a GPR119 receptor. Some embodiments pertain to methods, uses, compounds, and pharmaceutical agents, each as described herein, for agonizing a GPR119 receptor. Some embodiments pertain to methods, uses, compounds, and pharmaceutical agents, each as described herein, for increasing the secretion of an incretin in an individual. Some embodiments pertain to methods, uses, compounds, and pharmaceutical agents, each as described herein, for increasing a blood incretin level in an individual. Some embodiments pertain to methods, uses, compounds, and pharmaceutical agents, each as described herein, for treatment of a disorder selected from: a GPR119-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic-related disorder; and obesity; in an individual.
In some embodiments, the disorder is a GPR119-related disorder. In some
embodiments, the disorder is a condition ameliorated by increasing secretion of an incretin. In some embodiments, the disorder is a condition ameliorated by increasing a blood incretin level. In some embodiments, the disorder is a condition characterized by low bone mass. In some embodiments, the disorder is a neurological disorder. In some embodiments, the disorder is a metabolic-related disorder. In some embodiments, the disorder is obesity. In some
embodiments, the disorder is type 2 diabetes. In some embodiments, the disorder is hyperglycemia. In some embodiments, the disorder is hyperlipidemia. In some embodiments, the disorder is hypertriglyceridemia. In some embodiments, the disorder is type 1 diabetes. In some embodiments, the disorder is dyslipidemia. In some embodiments, the disorder is syndrome X.
In some embodiments, the pharmaceutical product comprises a pharmaceutical composition. In some embodiments, the pharmaceutical product comprises a formulation. In some embodiments, the pharmaceutical product comprises a unit dosage form. In some embodiments, the pharmaceutical product comprises a kit. In some embodiments, the pharmaceutical product comprises a combined preparation. In some embodiments, the pharmaceutical product comprises a twin pack.
In some embodiments, the compound and pharmaceutical agent or the second pharmaceutical agent are administered simultaneously, separately, or sequentially. In some embodiments, the compound and the pharmaceutical agent or the second pharmaceutical agent are administered simultaneously. In some embodiments, the compound and the pharmaceutical agent or the second pharmaceutical agent are administered separately. In some embodiments, the compound and the pharmaceutical agent or the second pharmaceutical agent are administered sequentially.
In some embodiments, the incretin is GLP-1. In some embodiments, the incretin is GIP. In some embodiments, the incretin is PYY.
One aspect of the present invention pertains to compositions, methods, pharmaceutical products, pharmaceutical compositions, uses; compounds, and pharmaceutical agents, each as described herein, wherein the pharmaceutical agent or the second pharmaceutical agent is selected from: a DPP-IV inhibitor, a biguanide, an alpha-glucosidase inhibitor, an insulin analogue, a sulfonylurea, an SGLT2 inhibitor, a meglitinide, a thiazolidinedione, an antidiabetic peptide analogue, and a DGAT-1 inhibitor. As provided herein are specific non-limiting examples of each of these agents.
One aspect of the present invention pertains to compositions, methods, pharmaceutical products, pharmaceutical compositions, uses; compounds, and pharmaceutical agents, each as described herein, wherein: 1) the compound and the pharmaceutical agent or the second pharmaceutical agent are provided in amounts which give a synergistic effect in treating the disorder; 2) the amount of the compound alone is substantially therapeutically ineffective at treating the disorder; and/or 3) the amount of the pharmaceutical agent alone or the second pharmaceutical agent alone is substantially therapeutically ineffective at treating the disorder.
One aspect of the present invention relates to methods for preparing pharmaceutical products of the present invention comprising the steps: mixing said compound with a first pharmaceutically acceptable carrier to prepare a compound unit dosage form; mixing said second pharmaceutical agent with a second pharmaceutically acceptable carrier to prepare a second pharmaceutical agent unit dosage form; and combining said compound unit dosage form and said second pharmaceutical agent unit dosage form in a combined unit dosage form for simultaneous, separate, or sequential use.
In some embodiments, the first pharmaceutically acceptable carrier is different from the second pharmaceutically acceptable carrier. In some embodiments, the different
pharmaceutically acceptable carriers are suitable for administration by the same route. In some embodiments, the different pharmaceutically acceptable carriers are suitable for administration by different routes. In some embodiments, the first pharmaceutically acceptable carrier is substantially the same as the second pharmaceutically acceptable carrier. In some embodiments, the substantially the same pharmaceutically acceptable carriers are suitable for oral administration.
Certain Indications of the Present Invention:
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 GPR119- receptor and therefore related diseases, conditions and/or disorders related thereto such as those described herein.
In some embodiments, modulating the activity includes the treatment of a GPRl 19- related disorder. In some embodiments, a GPRl 19-related disorder is a condition ameliorated by increasing a blood incretin level. In some embodiments, a GPRl 19-related disorder is a condition characterized by low bone mass. In some embodiments, a GPRl 19-related disorder is a neurological disorder. In some embodiments, a GPRl 19-related disorder is a metabolic-related disorder. In some embodiments, a GPRl 19-related disorder is type 2 diabetes. In some embodiments, a GPRl 19-related disorder is obesity.
Some embodiments of the present invention include every combination of one or more conditions characterized by low bone mass selected from: osteopenia, osteoporosis, rheumatoid arthritis, osteoarthritis, periodontal disease, alveolar bone loss, osteotomy bone loss, childhood idiopathic bone loss, Paget' s disease, bone loss due to metastatic cancer, osteolytic lesions, curvature of the spine, and loss of height.
In some embodiments, the neurological disorder selected from: stroke and
Parkinsonism.
Some embodiments of the present invention include every combination of one or more metabolic-related disorders selected from: type 1 diabetes, type 2 diabetes mellitus, and conditions associated therewith, such as, but not limited to, coronary heart disease, ischemic stroke, restenosis after angioplasty, peripheral vascular disease, intermittent claudication, 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, 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, vascular restenosis, hyperglycemia, hyperinsulinemia, hyperlipidemia, hypertriglyceridemia, insulin resistance, impaired glucose metabolism, erectile dysfunction, skin and connective tissue disorders, foot ulcerations and ulcerative colitis, endothelial dysfunction and impaired vascular compliance.
Some embodiments of the present invention include every combination of one or more metabolic-related disorders selected from: diabetes, 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, 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, motor-neuron disease, traumatic brain injury, spinal cord injury, and obesity.
In some embodiments, the disorder is type 2 diabetes. In some embodiments, the disorder is hyperglycemia. In some embodiments, the disorder is hyperlipidemia. In some embodiments, the disorder is hypertriglyceridemia. In some embodiments, the disorder is type 1 diabetes. In some embodiments, the disorder is dyslipidemia. In some embodiments, the disorder is syndrome X. In some embodiments, the disorder is obesity.
One aspect of the present invention pertains to methods for weight management, comprising administering to an individual in need thereof, a therapeutically effective amount of a compound of the present invention in combination with a therapeutically effective amount of a pharmaceutical agent, such as any agent described herein; wherein the compound and the pharmaceutical agent.
In some embodiments, the weight management comprises weight loss. In some embodiments, the weight management comprises maintenance of weight loss. In some embodiments, the weight management further comprises a reduced-calorie diet. In some embodiments, the weight management further comprises a program of regular exercise. In some embodiments, the weight management further comprises both a reduced-calorie diet and a program of regular exercise.
In some embodiments, the individual in need of weight management is a patient with an initial body mass of index > 40 kg/m2; > 39 kg/m2; > 38 kg/m2; > 37 kg/m2; > 36 kg/m2; > 35 kg/m2; > 34 kg/m2; > 33 kg/m2; > 32 kg/m2; > 31 kg/m2; > 30 kg/m2; > 29 kg/m2; > 28 kg/m2; > 27 kg/m2; > 26 kg/m2; > 25 kg/m2; > 24 kg/m2; > 23 kg/m2; > 22 kg/m2; > 21 kg/m2; or > 20 kg/m2; and the patient optionally has at least one or at least two weight related comorbid condition(s).
In some embodiments, the comorbid condition(s) when present are selected from: hypertension, dyslipidemia, cardiovascular disease, glucose intolerance, and sleep apnea.
Formulations and Compositions
Formulations may be prepared by any suitable method, typically by uniformly mixing the active compound(s) 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 ampule. 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 et al.)
While it is possible that, for use in the prophylaxis or treatment, a compound of the invention may, 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.
Pharmaceutical formulations include those suitable for oral, rectal, nasal, topical (including buccal and sub-lingual), vaginal or parenteral (including intramuscular, subcutaneous 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 minimal 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 formulations 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.
Compounds of the present invention or a solvate, hydrate or physiologically functional derivative thereof can be used as active ingredients in pharmaceutical compositions, specifically as GPR119 receptor modulators. The term "active ingredient", defined in the context of a "pharmaceutical composition", refers to a component of a pharmaceutical composition that provides the primary pharmacological effect, as opposed to an "inactive ingredient" which would generally be recognized as providing no pharmaceutical benefit.
The dose when using the compounds of the present invention can vary within wide limits and as is customary and is 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 conducted or on whether further active compounds are administered in addition to the compounds of the present invention.
Representative doses of the present invention include, but not limited to, about 0.001 mg to about 5000 mg, about 0.001 mg to about 2500 mg, about 0.001 mg to about 1000 mg, 0.001 mg 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. Multiple doses may be administered during the day, especially when relatively large amounts are deemed to be needed, for example 2, 3, or 4 doses. Depending on the individual and as deemed appropriate from the patient's physician or caregiver it may be necessary to deviate upward or downward from the doses described herein.
The amount of active ingredient, or an active salt or derivative 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. In some circumstances, these extrapolations may merely be based on the weight of the animal 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 the type, 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 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 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 dosage and dosage regimen outside these typical ranges can be tested and, where appropriate, may be used in the methods of this invention.
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. The daily dose can be divided, especially when relatively large amounts are administered as deemed appropriate, into several, for example 2, 3, or 4 part administrations. If appropriate, depending on individual behavior, it may be necessary to deviate upward or downward from the daily dose indicated.
The compounds of the present invention can be administrated 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, solvate, or hydrate 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 desire 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 active compound; 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" refers to 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.
Formulations 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 formulations 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.
Formulations suitable for topical administration in the mouth include lozenges comprising 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 formulations 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 formulation 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 formulations intended for administration to the respiratory tract, including intranasal formulations, 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, formulations 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 is 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.
Representative acids include, but are not limited to, acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethenesulfonic, dichloroacetic, formic, fumaric, gluconic, glutamic, hippuric, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic,
methanesulfonic, mucic, nitric, oxalic, pamoic, pantothenic, phosphoric, succinic, sulfiric, tartaric, oxalic, j-toluenesulfonic and the like. Certain compounds of the present invention which contain a carboxylic acid functional group may optionally exist as pharmaceutically acceptable salts containing non-toxic, pharmaceutically acceptable metal cations and cations derived from organic bases. Representative metals include, but are not limited to, aluminium, calcium, lithium, magnesium, potassium, sodium, zinc and the like. In some embodiments the pharmaceutically acceptable metal is sodium. Representative organic bases include, but are not limited to, benzathine (A^A^-dibenzylefhane-l^-diamine), chloroprocaine (2- (diethylamino)ethyl 4-(chloroamino)benzoate), choline, diethanolamine, ethylenediamine, meglumine ((2R,3R,4R,5S)-6-(methylamino)hexane-l,2,3,4,5-pentaol), procaine (2- (diethylamino)ethyl 4-aminobenzoate), and the like. Certain pharmaceutically acceptable salts are listed in Berge, et al., Journal of Pharmaceutical Sciences, 66: 1-19 (1977).
The acid addition salts may be obtained as the direct products 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.
Compounds of the present invention can be converted to "pro-drugs." The term "prodrugs" refers to compounds that have been modified with specific chemical groups known in the art and when administered into an individual these groups undergo biotransformation to give the parent compound. Pro-drugs can thus be viewed as compounds of the invention containing one or more specialized non-toxic protective groups used in a transient manner to alter or to eliminate a property of the compound. In one general aspect, the "pro-drug" approach is utilized to facilitate oral absorption. A thorough discussion is provided in T. Higuchi and V. Stella, Prodrugs as Novel Delivery Systems Vol. 14 of the A.C.S. Symposium Series; and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987.
Some embodiments of the present invention include a method of producing a pharmaceutical composition for "combination-therapy" comprising admixing at least one compound according to any of the compound embodiments disclosed herein, together with at least one known pharmaceutical agent as described herein and a pharmaceutically acceptable carrier.
It is noted that when the GPR119 receptor modulators are utilized as active ingredients in pharmaceutical compositions, these are not intended for use in humans only, but in non- human mammals as well. Recent advances in the area of animal health-care mandate that consideration be given for the use of active agents, such as GPR119 receptor modulators, for the treatment of a GPR119 receptor-associated disease or disorder in companionship animals (e.g., cats, dogs, etc.) and in livestock animals (e.g., horses, cows, etc.) Those of ordinary skill in the art are readily credited with understanding the utility of such compounds in such settings.
Hydrates and Solvates
It is understood that when the phrase "pharmaceutically acceptable salts, solvates, and hydrates" or the phrase "pharmaceutically acceptable salt, solvate, or hydrate" is used when referring to compounds described herein, it embraces pharmaceutically acceptable solvates and/or hydrates of the compounds, pharmaceutically acceptable salts of the compounds, as well as pharmaceutically acceptable solvates and/or hydrates of pharmaceutically acceptable salts of the compounds. It is also understood that when the phrase "pharmaceutically acceptable solvates and hydrates" or the phrase "pharmaceutically acceptable solvate or hydrate" is used when referring to salts described herein, it embraces pharmaceutically acceptable solvates and/or hydrates of such salts.
It will be apparent to those skilled in the art that the dosage forms described herein may comprise, as the active component, either a compound described herein or a pharmaceutically acceptable salt or as a pharmaceutically acceptable solvate or hydrate thereof. Moreover, various hydrates and solvates of the compounds described herein 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 K.J. Guillory, "Generation of Polymorphs, Hydrates, Solvates, and Amorphous Solids," in: Polymorphism in Pharmaceutical Solids, ed. Harry G. Britain, Vol. 95, Marcel Dekker, Inc., New York, 1999. Accordingly, one aspect of the present invention pertains to methods of administering hydrates and solvates of compounds described herein and/or their pharmaceutical acceptable salts, that can be isolated and characterized by 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).
Polymorphs 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 exhibited 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 other 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).
Crystalline forms can be identified by their unique solid state signature with respect to, for example, differential scanning calorimetry (DSC), powder X-ray diffraction (PXRD), and other solid state methods.
Further characterization with respect to water or solvent content of crystalline forms can be determined by any of the following methods, for example, thermogravimetric analysis (TGA), Karl Fischer analysis, and the like.
For DSC, it is known that the temperatures observed will depend upon sample purity, the rate of temperature change, as well as sample preparation technique and the particular instrument employed. Thus, the values relating to DSC thermograms can vary by plus or minus about 4 °C (+ 4 °C). The values relating to DSC thermograms can also vary by plus or minus about 20 joules per gram (+ 20 joules per gram).
DSC can be used to observe desolvation events. When DSC thermogram values relate to desolvation events, the values are understood to be estimates. Scan rate and pan closure can influence DSC values for desolvation events, which can vary by plus or minus about 25 °C. DSC values for desolvation events are typically recorded using a sample in an aluminum pan with an uncrimped lid and a scan rate of 10 °C/min.
For PXRD, the relative intensities of the peaks can vary, depending upon the sample preparation technique, the sample mounting procedure and the particular instrument employed. Moreover, instrument variation and other factors can often affect the 2 lvalues. Therefore, the peak assignments of diffraction patterns can vary by plus or minus 0.2 °2Θ (+ 0.2 °2Θ).
For TGA, the features reported herein can vary by plus or minus about 5 °C (+ 5 °C). The TGA features can also vary by plus or minus about 2% (+ 2%) weight change due to, for example, sample variation.
Further characterization with respect to hygroscopicity of a crystalline form can be gauged by, for example, dynamic moisture sorption (DMS). The DMS features can vary by plus or minus about 5% (+ 5%) relative humidity. The DMS features can also vary by plus or minus about 5% (+ 5%) weight change. COMBINATION THERAPY
A compound of the invention can be administered as the sole active pharmaceutical agent (i.e., mono-therapy), or it can be used in combination with one or more pharmaceutical agents (i.e., combination-therapy), such as pharmaceutical agents, such as, known anti-diabetic agents, either administered together or separately for the treatment of the diseases, conditions, and 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 thereof a therapeutically effective amount of a compound of Formula (I) and pharmaceutically acceptable salts, solvates and hydrates thereof, in combination with one or more pharmaceutical agents, such as anti-diabetic agents, as described herein.
In accordance with the present invention, the combination can be used by mixing the respective active components, a compound of Formula (I) and a pharmaceutical agent, either together or independently optionally with a physiologically acceptable carrier, excipient, binder, diluent, etc., as described herein, and administering the mixture or mixtures either orally or non- orally as a pharmaceutical composition(s). When a compound of Formula (I) is administered as a combination therapy with another active compound the compound of Formula (I) and the pharmaceutical agent can be formulated as separate pharmaceutical compositions given at the same time or at different times; or the compound of Formula (I) and the pharmaceutical agent can be formulated together as a single unit dosage.
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: Ar-(piperidin-l-yl)-5-(4-chlorophenyl)-l-(2,4- dichlorophenyl)-4-methyl-l/i-pyrazole-3-carboxamide]; melanin concentrating hormone antagonists; leptin (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 (GLP- 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 (H3R) antagonists or inverse 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 calorie-controlled diet.
It is understood that the scope of combination-therapy of the compounds of the present invention with 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 Formula (I) in combination with at least one pharmaceutical agent selected from the group consisting of: sulfonylureas (for example, tolbutamide (Orinase); acetohexamide (Dymelor); tolazamide (Tolinase); chlorpropamide (Diabinese); glipizide (Glucotrol); glyburide (Diabeta, Micronase, Glynase); glimepiride (Amaryl); gliclazide (Diamicron); and sulfonylureas known in the art); meglitinides (for example, repaglinide (Prandin), nateglinide (Starlix), mitiglinide, and other meglitinides known in the art); biguanides (for example, phenformin, metformin, buformin, and biguanides known in the art); a-glucosidase inhibitors (for example, acarbose, miglitol, and a-glucosidase inhibitors known in the art); thiazolidinediones - peroxisome proliferators-activated receptor-γ {i.e., PPAR-γ) agonists (for example, rosiglitazone (Avandia), pioglitazone (Actos), troglitazone (Rezulin), rivoglitazone, ciglitazone, and thiazolidinediones known in the art); insulin and insulin analogues; anti-diabetic peptide analogues (for example, exenatide, liraglutide, taspoglutide, and anti-diabetic peptides analogues know in the art); HMG- CoA reductase inhibitors (for example, rosuvastatin, pravastatin and its sodium salt, simvastatin, lovastatin, atorvastatin, fluvastatin, cerivastatin, rosuvastatin, pitavastatin, pravastatin, and other 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 other 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 other angiotensin converting enzyme inhibitors known in the art); angiotensin II receptor antagonists [for example, losartan (and the potassium salt form), and other angiotensin II receptor antagonists known in the art; adiponectin; squalene synthesis inhibitors {for example, (S)-a-[bis[2,2- dimethyl-l-oxopropoxy)methoxy] phosphinyl]-3-phenoxybenzenebutanesulfonic acid, mono potassium salt (BMS-188494) and other 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 simultaneously.
Suitable pharmaceutical agents that can be used in conjunction with compounds of the present invention include, but are not limited to: amylin agonists (for example, pramlintide); insulin secretagogues (for example, GLP-1 agonists, exendin-4, and insulinotropin (Liraglutide (NN2211)); acyl Co A cholesterol acetyltransferase inhibitors (for example, ezetimibe, eflucimibe, and other acyl Co A cholesterol acetyltransferase inhibitors known in the art);
cholesterol absorption inhibitors (for example, ezetimibe, pamaqueside and other cholesterol absorption inhibitors known in the art); cholesterol ester transfer protein inhibitors (for example, Torcetrapib (CP-529,414), Dalcetrapib (JTT-705), CETi-1, and other cholesterol ester transfer protein inhibitors known in the art); microsomal triglyceride transfer protein inhibitors (for example, implitapide, and other microsomal triglyceride transfer protein inhibitors known in the art); cholesterol modulators (for example, ibrolipim (NO-1886), and other cholesterol modulators known in the art); bile acid modulators (for example, GT103-279 and other bile acid modulators known in the art); insulin signaling pathway modulators; inhibitors of protein tyrosine phosphatases (PTPases); non-small molecule mimetics and inhibitors of glutamine- fructose-6-phosphate amido transferase (GFAT); compounds influencing a dysregulated hepatic glucose production; inhibitors of glucose-6-phosphatase (G6Pase); inhibitors of fructose- 1,6- bisphosphatase (F-l,6-BPase); inhibitors of glycogen phosphorylase (GP); glucagon receptor antagonists; inhibitors of phosphoenolpyruvate carboxykinase (PEPCK); pyruvate
dehydrogenase kinase (PDHK) inhibitors; insulin sensitivity enhancers; insulin secretion enhancers; inhibitors of gastric emptying; a2-adrenergic antagonists; retinoid X receptor (RXR) agonists; and dipeptidyl peptidase-4 (DPP-IV) inhibitors; DGAT-1 inhibitors; and the like. Tripartite Combinations
Some aspects of the present invention include compounds of Formula (I) that can be employed in any of the methods, pharmaceutical products, uses, compounds, and
pharmaceutical agents, as described herein, in combination with two distinct pharmaceutical agents.
In some embodiments, the two distinct pharmaceutical agents are selected from any of the pharmaceutical agents, or classes of pharmaceutical agents described herein. In some embodiments, the two distinct pharmaceutical agents are selected from: an inhibitor of DPP- IV, a biguanide, an alpha-glucosidase inhibitor, an insulin analogue, a sulfonylurea, a SGLT2 inhibitor, a meglitinide, a thiazolidinedione, an anti-diabetic peptide analogue, and a DGAT-1 inhibitor. In some embodiments, the two distinct pharmaceutical agents include every combination selected from pharmaceutical agents of the following group: an inhibitor of DPP- IV, a biguanide, an alpha-glucosidase inhibitor, a sulfonylurea, a SGLT2 inhibitor, and a DGAT-1 inhibitor.
Some embodiments of the present invention include every combination of one or more compounds selected from compounds of the following group and pharmaceutically acceptable salts, solvates, and hydrates thereof: an inhibitor of DPP-IV 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; (llS',3lS',5lS')-2-[2(lS')-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)-aminopiperidin-l-yl]-7-(2- butynyl)-3-methyl-l-(4-methylquinazolin-2-ylmethyl)xanthine; 1-[N-[3(R)- pyrrolidinyl]glycyl]pyrrolidin-2(R)-yl boronic acid; 4(¾-ί1υοΓθ-1-[2-[(ϋ?,35)-3-(1/ί-1,2,4- triazol-l-ylmethyl)cyclopentylamino]acetyl]pyrrolidine-2(lS')-carbonitrile; 1-[(25,35, 1 ^5)-2- amino-9, 10-dimethoxy-2,3,4,6,7,l lb-hexahydro-l/i-pyrido[2, l-a]isoquinolin-3-yl]-4(lSr)- (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-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-l-yl)-l,3,5-triazin-2-y^
(R)-2-((6-(3-aminopiperidin- l-yl)-3-methyl-2,4-dioxo-3,4-dihydropyrimidin-l(2//)-yl)methyl)- 4-fluorobenzonitrile; 5- { (S)-2- [2-((S)-2-cyano-pyrrolidin- 1 -yl)-2-oxo-ethylamino] -propyl } -5- (l/i-tetrazol-5-yl) 10, l l-dihydro-5/i-dibenzo[a,d]cycloheptene-2,8-dicarboxylic acid bis- dimethylamide; ((2S,4S)-4-(4-(3-methyl- 1 -phenyl- l/i-pyrazol-5-yl)piperazin- 1 -yl)pyrrolidin-2- yl)(thiazolidin-3-yl)methanone; (2S,4¾-l-[2-[(4-ethoxycarbonylbicyclo[2.2.2]oct-l- 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-pyrro 2-({6- [(3R)-3-amino-3-methylpiperidin-l-yl]-l,3-dimethyl-2,4-dioxo-l,2,3,4-tetrahydro-5/i- pyrrolo[3,2-d]pyrimidin-5-yl}methyl)-4-fluorobenzonitrile; (2S)-l-{ [2-(5-mefhyl-2-phenyl- oxazol-4-yl)-ethylamino] -acetyl } -pyrrolidine-2-carbonitrile; (2S)- 1 - { [ 1 , 1 -dimefhyl-3-(4-pyridin-
3- yl-imidazol- 1 -yl)-propylamino] -acetyl } -pyrrolidine-2-carbonitrile; (3,3-difluoropyrrolidin- 1 - yl)-((2,S,,4lS,)-4-(4-(pyrimidin-2-yl)piperazin-l-yl)pyrrolidin-2-yl)methanone; (25,45)-1-[(25)-2- amino-3,3-bis(4-fluorophenyl)propanoyl]-4-fluoropyrrolidine-2-carbonitrile; (2lS',5R)-5-ethynyl-
1- {Ar-(4-methyl-l-(4-carboxy-pyridin-2-yl)piperidin-4-yl)glycyl}pyrrolidine-2-carbonitrile; and (15,6R)-3-{ [3-(trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8//)-yl]carbonyl}-6- (2,4,5-trifluorophenyl)cyclohex-3-en-l -amine; a biguanide selected from: phenformin
((phenylethyl)biguanide); metformin (dimethylbiguanide); buformin (butylbiguanide); and proguanil (l-(p-chlorophenyl)-5-isopropylbiguanide); an a-glucosidase inhibitor selected from: acarbose ((2R3R,4R,5R)-4-((2R3R,4R,55,6R)-5-((2R,3R,45,5.S,,6R)-3,4-dihydroxy-6-methyl-5- ((15,4R,5lS',6lSr)-4,5,6-trihydroxy-3-(hydroxymethyl)cyclohex-2-enylamino)tetrahydro-2/i-pyran-
2- yloxy)-3,4-dihydroxy-6-(hydroxymethyl)tetrahydro-2/i-pyran-2-yloxy)-2,3,5,6- tetrahy droxyhexanal) ; miglitol ( (2R, 3R, 4R, 5 ¾ - 1 - (2 -hy droxy ethy 1) -2 - (hydroxymethyl)piperidine-3,4,5-triol); and voglibose ((lS,2S,3R,4S,5S)-5-(l,3- dihydroxypropan-2-ylamino)-l-(hydroxymethyl)cyclohexane-l,2,3,4-tetraol); an insulin analogue selected from: NPH insulin (also known as Humulin N, Novolin N, NPH Lletin II, and insulin isophane); insulin lispro (28B-L-lysine-29B-L-proline-insulin, wherein insulin is human insulin); insulin aspart (28B-L-aspartic acid-insulin, wherein insulin is human insulin); and insulin glulisine (3B-L-lysine-29B-L-glutamic acid-insulin, wherein insulin is human insulin); a sulfonylurea selected from: tolbutamide (Orinase, Ar-(butylcarbamoyl)-4- methylbenzenesulfonamide); acetohexamide (Dymelor, 4-acetyl-Ar-
(cyclohexylcarbamoyl)benzenesulfonamide); tolazamide (Tolinase, .V-(azepan-l-ylcarbamoyl)-
4- methylbenzenesulfonamide); chlorpropamide (Diabinese, 4-chloro-Ar- (propylcarbamoyl)benzenesulfonamide); glipizide (Glucotrol, N-(4-(N-
(cyclohexylcarbamoyl)sulfamoyl)phenethyl)-5-methylpyrazine-2-carboxamide); glibenclamide, also known as glyburide (Diabeta, Micronase, Glynase, 5-chloro-Ar-(4-(Ar- (cyclohexylcarbamoyl)sulfamoyl)phenethyl)-2-methoxybenzamide); glimepiride (Amaryl, 3- ethyl-4-methyl-Ar-(4-(Ar-((lr,4r)-4-methylcyclohexylcarbamoyl)sulfamoyl)phenethyl)-2-oxo- 2,5-dihydro-l/i-pyrrole-l-carboxamide); and gliclazide (Diamicron, Λ'- (hexahydrocyclopenta[c]pyrrol-2(l//)-ylcarbamoyl)-4-methylbenzenesulfonamide); a SGLT2 inhibitor selected from: dapagliflozin ((2S,3RAR,5S,6R)-2-(4-chloio-3-(4- ethoxybenzyl)phenyl)-6-(hydroxymethyl)tetrahydro-2/i-pyran-3,4,5-triol); remogliflozin (ethyl ((2R,3,S,,4lS,,5R,6lS,)-3,4,5-trihydroxy-6-(4-(4-isopropoxybenzyl)-l-isopropyl-5-methyl-l/i- pyrazol-3-yloxy)tetrahydro-2/i-pyran-2-yl)methyl carbonate); ASP1941, canagliflozin ((2lS,,3R,4R,5.S,,6R)-2-(3-((5-(4-fluorophenyl)thiophen-2-yl)methyl)-4-methylphenyl)-6- (hydroxymethyl)tetrahydro-2/i-pyran-3,4,5-triol); ISIS 388626; sergliflozin (ethyl
((2R,3^',4lS',5R,6lSr)-3,4,5-trihydroxy-6-(2-(4-methoxybenzyl)phenoxy)tetrahydro-2/i-pyran-2- yl)methyl carbonate), AVE2268 ((2R,3S,4S,5R,6¾-2-(hydroxymethyl)-6-(2-(4- methoxybenzyl)thiophen-3-yloxy)tetrahydro-2H-pyran-3,4,5-triol), BI10773, CSG453; and LX4211; a meglitinide selected from: repaglinide (Prandin, (S)-2-ethoxy-4-(2-(3-mefhyl-l-(2- (piperidin-l-yl)phenyl)butylamino)-2-oxoethyl)benzoic acid); nateglinide (Starlix, (R)-2- ((lr,4R)-4-isopropylcyclohexanecarboxamido)-3-phenylpropanoic acid); and mitiglinide ((5)-2- benzyl-4-((3aR,7a¾- m-isoindol-2(3/i,3a/i,4/i,5/i,6/i,7/i,7a//)-yl)-4-oxobutanoic acid); a thiazolidinedione selected from: rosiglitazone (Avandia, 5-(4-(2-(methyl(pyridin-2- yl)amino)ethoxy)benzyl)thiazolidine-2,4-dione); pioglitazone (Actos, 5-(4-(2-(5-ethylpyridin-2- yl)ethoxy)benzyl)thiazolidine-2,4-dione); troglitazone (Rezulin, 5-(4-((6-hydroxy-2,5,7,8- tetramethylchroman-2-yl)methoxy)benzyl)thiazolidine-2,4-dione); rivoglitazone (5-(4-((6- methoxy-1 -methyl- l/i-benzo[d]imidazol-2-yl)methoxy)benzyl)thiazolidine-2,4-dione); and ciglitazone (5-(4-((l-methylcyclohexyl)methoxy)benzyl)thiazolidine-2,4-dione); an antidiabetic peptide analogue selected from: exenatide; liraglutide; and taspoglutide; and a
DGAT-1 inhibitor selected from: (lr,4r)-4-(4-(5-(3,4-difluorophenylamino)-l,3,4-oxadiazole- 2-carboxamido)-3-fluorophenyl)cyclohexanecarboxylic acid; ira«5-(4-{4-[5-(6-trifluoromethyl- pyridin-3ylamino)-pyridin-2-yl] -phenyl }-cyclohexyl)-acetic acid; and {iraws-4-(4-(4-amino-5- oxo-7, 8-dihydropyrimido[5.4-f] [f]oxazepin-6(5//)yl)phenyllcyclohexyl} acetic acid.
In some embodiments, the two distinct pharmaceutical agents include every combination selected from pharmaceutical agents of the following group: sitagliptin, vildagliptin, saxagliptin, alogliptin, linagliptin, phenformin, metformin, buformin, acarbose, miglitol, voglibose, tolbutamide, acetohexamide, tolazamide, chlorpropamide, glipizide, glibenclamide, glimepiride, gliclazide, dapagliflozin, remigliflozin, and sergliflozin.
Dipeptidyl Peptidase 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-1), 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. Peptide YY (PYY) is a gut peptide that has been implicated in modulating satiety (Chaudhri et al, Annu Rev Physiol (2008) 70:239-255). 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 DPP-IV. 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 therapeutics, for example, oral administration of vildagliptin (l-[2-(3-hydroxyadamant-l- ylamino)acetyl]pyrrolidine-2(S)-carbonitrile) or sitagliptin (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) 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 DPP-IV inhibitors," Biochim. Biophys. Acta, 1751 : 33-44 (2005), and (2) 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, 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-known 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. Na.t 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, 0-benzoyl hydroxylamine, as described, for example, 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. Pat. Nos. 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 Nos. 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 Nos. 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 μΜ, less than about 1 μΜ, 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 IC50 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 1 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 PPCE, DPP-II, DPP-8 and DPP-9 of at least about 10-fold. 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 PPCE, DPP-II, DPP-8 and DPP-9 of at least about 100-fold. 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 PPCE, DPP-II, DPP-8 and DPP-9 of at least about 10-fold. 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 PPCE, DPP-II, DPP-8 and DPP-9 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 compounds of the following group 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; l-[2-(3- hydroxyadamant-l-ylamino)acetyl]pyrrolidine-2(5)-carbonitrile; (15,35,55)-2-[2(5)-amino-2-(3- hydroxyadamantan- 1 -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)-aminopiperidin-l-yl]-7-(2-butynyl)-3-methyl-l-(4-methylquinazolin-2- ylmethyl)xanthine; l-[Ar-[3(R)-pyrrolidinyl]glycyl]pyrrolidin-2(R)-yl boronic acid; 4(5)-fluoro- 1 - [2- [( lR,35)-3-( 1 H- 1 ,2,4-triazol- 1 -ylmethyl)cyclopentylamino]acetyl]pyrrolidine-2(5)- carbonitrile; l-[(2lS,,3.S,,l lblS,)-2-amino-9, 10-dimethoxy-2,3,4,6,7, l lb-hexahydro- m-pyrido[2, l- a]isoquinolin-3-yl]-4(5)-(fluoromefhyl)pyrrolidin-2-one; (25,45)-2-cyano-4-fluoro-l-[(2- hydroxy- 1, 1 -dimethyl) ethylamino]acetylpyrrolidine; 8-(cw-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- ((3lS',4lS')-4-amino-l-(4-(3,3-difluoropyrrolidin-l-yl)-l,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(2//)-yl)methyl)-4-fluorobenzonitrile; 5-{ (lS')-2-[2-((lS')-2-cyano-pyrrolidin-l- yl)-2-oxo-ethylamino]-propyl }-5-(l/i-tetrazol-5-yl)10,l l-dihydro-5/i- dibenzo[a,d]cycloheptene-2,8-dicarboxylic acid bis-dimethylamide; ((25,45)-4-(4-(3-mefhyl-l- phenyl- l/i-pyrazol-5-yl)piperazin- 1 -yl)pyrrolidin-2-yl)(thiazolidin-3-yl)methanone; (25,45)- 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-5/i-pyrrolo[3,2-d]pyrimidin-5-yl }methyl)-4-fluorobenzonitrile; (25)- 1 - { [2-(5-methyl-2-phenyl-oxazol-4-yl)-ethylamino] -acetyl } -pyrrolidine-2-carbonitrile; (25)- 1 - { [ 1 , 1 -dimethyl-3-(4-pyridin-3-yl-imidazol- 1 -yl)-propylamino] -acetyl } -pyrrolidine-2- carbonitrile; (33-difluoropyrrolidin-l-yl)-((25,45)-4-(4-(pyrimidin-2-yl)piperazin-l- yl)pyrrolidin-2-yl)methanone; (25,45)- 1 -[(25)-2-amino-3, 3-bis(4-fluorophenyl)propanoyl]-4- fluoropyrrolidine-2-carbonitrile; (25,5R)-5-ethynyl- 1 - {Ar-(4-methyl- l-(4-carboxy-pyridin-2- yl)piperidin-4-yl)glycyl}pyrrolidine-2-carbonitrile; and (15,6R)-3-{ [3-(trifluoromethyl)-5,6- dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8//)-yl]carbonyl}-6-(2,4,5-trifluorophenyl)cyclohex-3- en-1 -amine.
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-l-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-l-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 compounds disclosed in WO2003/004498 and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the DPP-IV inhibitor 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-l-one, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000057_0001
In some embodiments, the DPP-IV inhibitor is (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 phosphate:
Figure imgf000057_0002
The crystalline form of (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 phosphate salt monohydrate is disclosed in international patent publication WO2005/003135. In some embodiments, the DPP-IV inhibitor is crystalline (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 phosphate monohydrate.
Vildagliptin (Galvus®, LAF-237, l-[2-(3-hydroxyadamant- l- ylaniino)acetyl]pyrrolidine-2(S)-carbonitrile) is another 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. Vildagliptin was approved in Japan in 2010. The compound, l-[2-(3-hydroxyadamant-l-ylamino)acetyl]pyrrolidine-2(lS')-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 compounds disclosed in WO2000/034241 and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the DPP-IV inhibitor is selected from l-[2-(3- hydroxyadamant- 1 -ylamino)acetyl]pyrrolidine-2(¾-carbonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000058_0001
Certain salts of the compound, l-[2-(3-hydroxyadamant-l-ylamino)acetyl]pyrrolidine-2(S)- 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 HCI:
Figure imgf000058_0002
Saxagliptin (Onglyza™, BMS-477118, (l£3£5S)-2-[2(S)-ainino-2-(3- hydroxyadamantan-l-yl)acetyl]-2-azabicyclo[3.1.0]hexane-3-carbonitrile) is another DPP-IV inhibitor, which 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 approved in the E.U. for the treatment of type 2 diabetes independently or in combination with metformin. Phase 3 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 compounds disclosed in WO2001/068603 and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the DPP-IV inhibitor is selected from (lS,3S,5S)-2-[2(S)-amino-2-(3- hydroxyadamantan- 1 -yl)acetyl] -2-azabicyclo [3.1.0]hexane-3-carbonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000059_0001
Takeda has filed for regulatory approval of the DPP-IV inhibitor, alogliptin (SYR-322, 2-[6-[3(R)-aminopiperidin- 1 -yl] -3-methyl-2,4-dioxo- 1 ,2,3,4-tetrahydropyrimidin- 1- ylmethyl]benzonitrile) in Japan and the U.S for the once-daily, oral treatment of type 2 diabetes. The compound, 2-[6-[3(R)-aminopiperidin-l-yl]-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 compounds disclosed in WO 2005/095381 and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the DPP-IV inhibitor is selected from 2-[6-[3(R)- aminopiperidin-l-yl]-3-methyl-2,4-dioxo- l,2,3,4-tetrahydropyrimidin-l-ylmethyl]benzonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000059_0002
The crystalline form of 2-[6-[3(R)-aminopiperidin-l-yl]-3-methyl-2,4-dioxo-l,2,3,4- tetrahydropyrimidin-l-ylmethyl]benzonitrile is disclosed in international patent publication WO2007/035372. In some embodiments, the DPP-IV inhibitor is 2-[6-[3(R)-aminopiperidin-l- yl] -3-methyl-2,4-dioxo- -tetrahydropyrimidin- 1 -ylmethyl]benzonitrile benzoate:
Figure imgf000059_0003
Linagliptin (BI-1356, Tradjenta®, 8-[3(R)-aminopiperidin-l-yl]-7-(2-butynyl)-3- methyl-l-(4-methylquinazolin-2-ylmethyl)xanthine) is an inhibitor of DPP-IV approved by the FDA in May 2011 as an adjunct to diet and excerise to improve glycemic control in adults with 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 compounds disclosed in WO2004/018468 and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the DPP-IV inhibitor is selected from 8-[3(R)-aminopiperidin-l-yl]-7-(2-butynyl)-3-methyl- l-(4- methylquinazolin-2-ylmethyl)xanthine, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000060_0001
Certain polymorphs of the compound, 8-[3(R)-aminopiperidin- l-yl]-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-l-yl]-7-(2-butynyl)-3-methyl-l-(4-methylquinazolin-2-ylmethyl)xanthine.
Dutogliptin (PHX-1 149, l-[Ar-[3(R)-pyrrolidinyl]glycyl]pyrrolidin-2(R)-yl boronic acid) is a DPP-IV inhibitor in phase 3 clinical trials by Phenomix and Forest for the oral, once-daily treatment of type 2 diabetes. The compound, l-|7V-[3(R)-pyrrolidinyl]glycyl] pyrrolidin-2(R)-yl boronic acid, and pharmaceutically acceptable salts thereof are disclosed in international patent publication WO2005/047297. Some embodiments of the present invention include every combination of one or more compounds selected from compounds disclosed in
WO2005/047297 and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the DPP-IV inhibitor is selected from l-^-fS^-pyrrolidinylJglycylJpyrrolidin- 2(R)-yl boronic acid, and ph lvates, and hydrates thereof:
Figure imgf000060_0002
The crystalline form of l-[Ar-[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- oronic acid tartrate:
Figure imgf000060_0003
Melogliptin (GRC-8200, 4(S)-fluoro- 1 - [2- [( lR,3S)-3-( IH- 1 ,2,4-triazol- 1 - ylmethyl)cyclopentylamino]acetyl]pyrrolidine-2(lSr)-carbonitrile) is a DPP-IV inhibitor currently undergoing phase 2 clinical trials by Glenmark Pharmaceuticals and Merck KGaA for the treatment of type 2 diabetes. The compound, 4(¾-ί1υοΓθ-1-[2-[(ϋ?,35)-3-(1/ί-1,2,4-ΐΓΪ3ζο1-1- ylmethyl)cyclopentylamino]acetyl]pyrrolidine-2(lSr)-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 compounds disclosed in
WO2006/040625 and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the DPP-IV inhibitor is selected from 4(S)-fluoro-l-[2-[(lR,3S)-3-(l#-l,2,4- triazol- 1 -ylmethyl)cyclopentylamino]acetyl]pyrrolidine-2(lS')-carbonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000061_0001
Carmegliptin (R-1579, 1-[(2S,3S, 1 lbS)-2-arnino-9, 10-dimethoxy-2,3,4,6,7,l lb- hexahydro-l/i-pyrido[2,l-a]isoquinolin-3-yl]-4(lSr)-(fluoromethyl)pyrrolidin-2-one) is a DPP-IV inhibitor. The compound, l-[(2lS',3lS', l lblS')-2-amino-9, 10-dimethoxy-2,3,4,6,7,l lb-hexahydro- l/i-pyrido[2,l-a]isoquinolin-3-yl]-4(lSr)-(fluoromethyl)pyrrolidin-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 compounds disclosed in WO2005/000848 and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the DPP-IV inhibitor is selected from l-[(2lS',3lS', l lblS')-2-amino-9, 10-dimethoxy- 2,3,4,6,7, l lb-hexahydro- l/i-pyrido[2, l -a]isoquinolin-3-yl]-4(lS')-(fluoromethyl)pyrrolidin-2- one, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000061_0002
Taisho disclosed (2S,4S)-2-cyano-4-fluoro- 1 - [(2-hydroxy- 1 , 1 -dimethyl)
ethylamino]acetylpyrrolidine, a DPP-IV inhibitor in US patent publication US 2007/0112059. Some embodiments of the present invention include every combination of one or more compounds selected from compounds disclosed in US 2007/0112059 and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the DPP-IV inhibitor is selected from (2S,4S)-2-cyano-4-fluoro-l -[(2 -hydroxy- 1, 1- dimethyl)ethylamino]acetylpyrrolidine, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000062_0001
Sanofi-Aventis disclosed a series of substituted bicyclic 8-pyrrolidineoxanthine derivatives as DPP- IV inhibitors in US publication US 2007/0167468. Some embodiments of the present invention include every combination of one or more compounds selected from compounds disclosed in US publication US 2007/0167468 and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the DPP-IV inhibitor is selected from 8-(cw-hexahydro-pyrrolo[3,2-b]pyrrol-l-yl)-3-methyl-7-(3-methyl-but-2-enyl)-l -(2-0X0-2- phenylethyl)-3,7-dihydro-purine-2,6-dione, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000062_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 compounds disclosed in WO2007/148185 and pharmaceutically acceptable salts, solvates, and hydrates thereof. One such compound is l-((3lS',4lSr)-4-amino-l-(4-(3,3-difluoropyrrolidin-l-yl)- l,3,5-triazin-2-yl)pyrrolidin-3-yl)-5,5difluoropiperidin-2-one. In some embodiments, the DPP- IV inhibitor is selected from l-((3lS',4lSr)-4-amino-l-(4-(3,3-difluoropyrrolidin-l-yl)-l,3,5-triazin- 2-yl)pyrrolidin-3-yl)-5,5difluoropiperidin-2-one, and pharmaceutically acceptable salts, solvates, and hydrates thereof
Figure imgf000062_0003
Syrrx disclosed a series of substituted pyrimidine-2,4(l/i,3//)-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 compounds disclosed in WO2005/095381 and pharmaceutically acceptable salts, solvates, and hydrates thereof. One such compound is (R)-2-((6-(3-aminopiperidin- l-yl)-3-methyl-2,4-dioxo- 3,4-dihydropyrimidin-l(2//)-yl)methyl)-4-fluorobenzonitrile. In some embodiments, the DPP- IV inhibitor is selected from (R)-2-((6-(3-aminopiperidin-l-yl)-3-methyl-2,4-dioxo-3,4- dihydropyrimidin- l(2//)-yl)methyl)-4-fluorobenzonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000063_0001
Various crystalline forms of (R)-2-((6-(3-aminopiperidin-l-yl)-3-methyl-2,4-dioxo-3,4- dihydropyrimidin- l(2//)-yl)methyl)-4-fluorobenzonitrile succinic acid salt are disclosed in international patent publication WO2008/067465. 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(2//)-yl)methyl)-4-fluorobenzonitrile succinic acid salt as described in international patent publication WO2008/067465. In some embodiments, the DPP- IV inhibitor is crystalline (R)-2-((6-(3-aminopiperidin-l-yl)-3-methyl-2,4-dioxo-3,4- dihydropyrimidin- l(2//)-yl)methyl)-4-fluorobenzonitrile succinic acid salt:
Figure imgf000063_0002
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 compounds disclosed in WO2006/116157 and pharmaceutically acceptable salts, solvates, and hydrates thereof. One such compound is 5- {(S)-2-[2-((S)-2-cyano-pyrrolidin-l-yl)-2-oxo- ethylamino] -propyl } -5-( l/i-tetrazol-5-yl) 10, 11 -dihydro-5/i-dibenzo[a,d]cycloheptene-2,8- dicarboxylic acid bis-dimethylamide. In some embodiments, the DPP-IV inhibitor is selected from 5- { (S)-2- [2-((S)-2-cyano-pyrrolidin- 1 -yl)-2-oxo-ethylamino] -propyl } -5-( l/i-tetrazol-5- yl) 10, l l-dihydro-5/i-dibenzo[a,d]cycloheptene-2,8-dicarboxylic acid bis-dimethylamide, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000064_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 compounds disclosed in WO2002/0014271 and pharmaceutically acceptable salts, solvates, and hydrates thereof. One such compound is ((2S,4S)-4-(4-(3-methyl-l -phenyl- l/i-pyrazol-5- yl)piperazin-l-yl)pyrrolidin-2-yl)(thiazolidin-3-yl)methanone. In some embodiments, the DPP- IV inhibitor is selected from ((2lS',4lSr)-4-(4-(3-methyl-l-phenyl-l/i-pyrazol-5-yl)piperazin-l- yl)pyrrolidin-2-yl)(thiazolidin-3-yl)methanone, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000064_0002
Various crystalline forms of ((2lS',4lSr)-4-(4-(3-methyl-l-phenyl-l/i-pyrazol-5-yl)piperazin-l- yl)pyrrolidin-2-yl)(thiazolidin-3-yl)methanone salts are disclosed in international patent publication WO2006/088129 and US publication 2009/0216016. One embodiment of the present invention pertains to any one or more crystalline forms of ((2S,4S)-4-(4-(3-methyl-l- phenyl- l/i-pyrazol-5-yl)piperazin- 1 -yl)pyrrolidin-2-yl)(thiazolidin-3-yl)methanone salt as described in international patent publication WO2006/088129 and US publication
2009/0216016. In some embodiments, the DPP-IV inhibitor is crystalline ((25,45)-4-(4-(3- methyl- 1 -phenyl- l/i-pyrazol-5-yl)piperazin- 1 -yl)pyrrolidin-2-yl)(thiazolidin-3-yl)methanone 2.5 hydrobromide salt:
Figure imgf000065_0001
or a mono or a dihydrate thereof. In some embodiments, the DPP- IV inhibitor is crystalline ((25,45)-4-(4-(3-mefhyl-l -phenyl- l/i-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 US 2008/0146818. Some embodiments of the present invention include every combination of one or more compounds selected from compounds disclosed in WO2008/114857 and US publication US 2008/0146818, and pharmaceutically acceptable salts, solvates, and hydrates thereof. One such compound is (2lS',4lSr)-l-[2-[(4-ethoxycarbonylbicyclo[2.2.2]oct-l-yl)amino]acetyl]-4-fluoropyrrolidine-2- carbonitrile. In some embodiments, the DPP-IV inhibitor is selected from (25,45)- 1- [2- [(4- ethoxycarbonylbicyclo[2.2.2]oct-l-yl)amino]acetyl]-4-fluoropyrrolidine-2-carbonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000065_0002
Dainippon Sumitomo disclosed a series of bicyclic pyrrole derivatives as DPP-IV inhibitors in international patent publication WO2006/068163 and US publication US
2009/0192129. Some embodiments of the present invention include every combination of one or more compounds selected from compounds disclosed in WO2006/068163 and US publication US 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. In some embodiments, the DPP-IV inhibitor is selected from (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, solvates, and hydrates thereof:
Figure imgf000066_0001
Dainippon Sumitomo disclosed 2-({6-[(3R)-3-amino-3-methylpiperidin- l-yl]-l,3- dimethyl-2,4-dioxo- 1,2,3, 4-tetrahydro-5/i-pyrrolo[3,2-d]pyrimidin-5-yl}methyl)-4- fluorobenzonitrile as a DPP-IV inhibitor in international patent publication WO2009/084497. In some embodiments, the DPP-IV inhibitor is selected from 2-({6-[(3R)-3-amino-3- methylpiperidin-l-yl]-l,3-dimethyl-2,4-dioxo- 1,2,3, 4-tetrahydro-5/i-pyrrolo[3,2-d]pyrimidin-5- yl}methyl)-4-fluorobenzonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000066_0002
Hoffmann-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 compounds disclosed in WO 03/037327 and pharmaceutically acceptable salts, solvates, and hydrates thereof. One such compound is (2¾-l-{ [2-(5-mefhyl-2-phenyl-oxazol-4-yl)- ethylamino] -acetyl }-pyrrolidine-2-carbonitrile. In some embodiments, the DPP-IV inhibitor is selected from (2S)-l-{ [2-(5-methyl-2-phenyl-oxazol-4-yl)-ethylamino] -acetyl }-pyrrolidine-2- carbonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000066_0003
Various crystalline forms of (2lS')-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 is (2S)-l-{ [2-(5- methyl-2-phenyl-oxazol-4-yl)-ethylamino]-acetyl}-pyrrolidine-2-carbonitrile methansulfonic acid salt (i.e., mesylate):
Figure imgf000067_0001
Other compounds disclosed by Hoffmann-La Roche in international patent publication WO 03/037327 include (2S)- 1 - { [ 1 , 1 -dimethyl-3-(4-pyridin-3-yl-imidazol- 1 -yl)-propylamino] - acetyl }-pyrrolidine-2-carbonitrile, and pharmaceutically acceptable salts thereof, such as the methansulfonic acid salt. In some embodiments, the DPP-IV inhibitor is selected from (2S)-l- { [1, 1 -dimethyl-3-(4-pyridin-3-yl-imidazol- 1 -yl)-propylamino] -acetyl } -pyrrolidine-2- carbonitrile, and pharmaceu ates thereof:
Figure imgf000067_0002
In some embodiments, the DPP-IV inhibitor is (2S)-l-{ [l,l-dimethyl-3-(4-pyridin-3-yl- imidazol- 1 -yl)-propylamino] -acetyl } -pyrrolidine-2-carbonitrile methansulfonic acid:
Figure imgf000067_0003
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. In some embodiments, the DPP-IV inhibitor is (25)- 1- { [1, 1 -dimethyl-3-(4-pyridin-3-yl-imidazol- 1 -yl)-propylamino] -acetyl } -pyrrolidine-2- carbonitrile fumaric acid salt (i.e., fumarate):
Figure imgf000067_0004
Pfizer disclosed a series of proline derivatives as DPP-IV inhibitors in international patent publication WO2005/ 116014. Some embodiments of the present invention include every combination of one or more compounds selected from compounds disclosed in
WO2005/1 16014 and pharmaceutically acceptable salts, solvates, and hydrates thereof. One such compound is (3,3-difluoropyrrolidin-l-yl)-((2lS,,45)-4-(4-(pyrimidin-2-yl)piperazin-l- yl)pyrrolidin-2-yl)methanone. In some embodiments, the DPP-IV inhibitor is selected from (3,3-difluoropyrrolidin- 1 -γ1)-((25,45)-4-(4-(ργΓΪηιϊ(1ϊη-2-γ1)ρϊρεΓαζϊη- 1 -yl)pyrrolidin-2- yl)methanone, and pharmac ates thereof:
Figure imgf000068_0001
GlaxoSmithKline 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,4¾-l-[(2S)-2-amino-3,3-bis(4- fluorophenyl)propanoyl]-4-fluoropyrrolidine-2-carbonitrile (Denagliptin). In some embodiments, the DPP-IV inhibitor is selected from (2S,4¾-l-[(2¾-2-amino-3,3-bis(4- fluorophenyl)propanoyl]-4-fluoropyrrolidine-2-carbonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000068_0002
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 (2S,4S)-l-[(2S)-2- amino-3,3-bis(4-fluorophenyl)propanoyl]-4-fluoropyrrolidine-2-carbonitrile j-toluenesulfonic acid salt (also referred to as (2lS',4lSr)-4-fluoro-l-[4-fluoro- -(4-fluorophenyl)-L-phenylalanyl]-2- pyrrolidinecarbonitrile j-toluenesulfonic acid salt, or Denagliptin tosylate). In some embodiments, the DPP-IV inhibitor is (2S,4¾-l-[(2S)-2-amino-3,3-bis(4- fluorophenyl)propanoyl]-4-fluoropyrrolidine-2-carbonitrile j-toluenesulfonic acid salt:
Figure imgf000068_0003
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 (2lS',5R)-5-ethynyl-l-{Ai-(4-methyl-l-(4-carboxy-pyridin-2- yl)piperidin-4-yl)glycyl}pyrrolidine-2-carbonitrile. In some embodiments, the DPP-IV inhibitor is selected from (2lS',5R)-5-ethynyl-l-{Ai-(4-methyl-l-(4-carboxy-pyridin-2-yl)piperidin-4- yl)glycyl}pyrrolidine-2-carbonitrile, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000069_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 (15,6^)-3-{ [3- (trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8//)-yl]carbonyl}-6-(2,4,5- trifluorophenyl)cyclohex-3-en-l -amine. In some embodiments, the DPP-IV inhibitor is selected from (llS',6R)-3-{ [3-(trifluoromethyl)-5,6-dihydro[l,2,4]triazolo[4,3-a]pyrazin-7(8//)- yl]carbonyl}-6-(2,4,5-trifluorophenyl)cyclohex-3-en-l-amine, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000069_0002
Biguanides
The biguanides are a class of drugs that stimulate anaerobic glycolysis, increase the sensitivity to insulin in the peripheral tissues, inhibit glucose absorption from the intestine, suppress of hepatic gluconeogenesis, and inhibit fatty acid oxidation. Examples of biguanides include phenformin ((phenylethyl)biguanide), metformin (dimethylbiguanide), buformin
(butylbiguanide), proguanil (l-(p-chlorophenyl)-5-isopropylbiguanide), and biguanides known in the art.
In some embodiments, the pharmaceutical agent or said second pharmaceutical agent is a biguanide selected from the following biguanide: (phenylethyl)biguanide, dimethylbiguanide, butylbiguanide, l-(p-chlorophenyl)-5- isopropylbiguanide, and pharmaceutically acceptable salts, solvates, and hydrates thereof.
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a biguanide selected from (phenylethyl)biguanide (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000070_0001
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a biguanide selected from dimethylbiguanide (chemical structure shown below) and
pharmaceutically acceptable salts, solvates, and hydrates thereof; the chemical structure is as follows:
Figure imgf000070_0002
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a biguanide selected from butylbiguanide (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof; the chemical structure is as follows:
Figure imgf000070_0003
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a biguanide selected from l-(p-chlorophenyl)-5-isopropylbiguanide (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof; the chemical structure is as follows:
Figure imgf000070_0004
In some embodiments, the pharmaceutical agent or said second pharmaceutical agent is a biguanide selected from the following biguanides: metformin, phenformin, buformin, and proguanil. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is metformin. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is phenformin. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is buformin. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is proguanil.
Alpha-Gluocosidase Inhibitors α-Glucosidase inhibitors belong to the class of drugs which competitively inhibit digestive enzymes such as a-amylase, maltase, a-dextrinase, sucrase, etc. in the pancreas and or small intestine. The reversible inhibition by a-glucosidase inhibitors retard, diminish or otherwise reduce blood glucose levels by delaying the digestion of starch and sugars. Some representative examples of a-glucosidase inhibitors include acarbose ((2R,3R,4R,5R)-4- ((2R,3R,4R,5lS,,6R)-5-((2R,3R,4lS,,5lS,,6R)-3,4-dihydroxy-6-methyl-5-((llS,,4R,5.S,,6lS,)-4,5,6- trihydroxy-3-(hydroxymethyl)cyclohex-2-enylamino)tetrahydro-2/i-pyran-2-yloxy)-3,4- dihydroxy-6-(hydroxymethyl)tetrahydro-2/i-pyran-2-yloxy)-2,3,5,6-tetrahydroxyhexanal), miglitol ((2R,3R,4R,5lSr)-l-(2-hydroxyethyl)-2-(hydroxymethyl)piperidine-3,4,5-triol), voglibose ((llS',2lS',3R,4lS',5lSr)-5-(l,3-dihydroxypropan-2-ylamino)-l-(hydroxymethyl)cyclohexane-l,2,3,4- tetraol), and α-glucosidase inhibitors known in the art.
In some embodiments, the pharmaceutical agent or said second pharmaceutical agent is a α-glucosidase inhibitor selected from the following α-glucosidase inhibitors:
(2R,3R,4R,5R)-4-((2R,3R,4R,5.S,,6R)-5-((2R,3R,4lS,,5lS,,6R)-3,4-dihydroxy-6-methyl-5- ((llS',4R,5lS',6lSr)-4,5,6-trihydroxy-3-(hydroxymethyl)cyclohex-2-enylamino)tetrahydro-2/i-pyran- 2-yloxy)-3,4-dihydroxy-6-(hydroxymethyl)tetrahydro-2/i-pyran-2-yloxy)-2, 3,5,6- tetrahydroxyhexanal; (2R,3R,4R,5S)- 1 -(2-hydroxyethyl)-2-(hydroxymethyl)piperidine-3,4,5- triol; (llS',2lS',3R,4lS',5lSr)-5-(l,3-dihydroxypropan-2-ylamino)-l-(hydroxymethyl)cyclohexane- 1,2,3,4-tetraol; and pharmaceutically acceptable salts, solvates, and hydrates thereof.
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a α-glucosidase inhibitor selected from (2^,3^,4^,5^)-4-((2^,3^,4^,55,6^)-5-((2^,3^,45,55,6^)- 3,4-dihydroxy-6-methyl-5-((llS',4R,5lS',6lS')-4,5,6-trihydroxy-3-(hydroxymethyl)cyclohex-2- enylamino)tetrahydro-2/i-pyran-2-yloxy)-3,4-dihydroxy-6-(hydroxymethyl)tetrahydro-2/i- pyran-2-yloxy)-2,3,5,6-tetrahydroxyhexanal (chemical structure shown below) and
pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000071_0001
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a α-glucosidase inhibitor selected from (2R,3R,4R,5S)-l-(2-hydroxyefhyl)-2- (hydroxymethyl)piperidine-3,4,5-triol (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000072_0001
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a a-glucosidase inhibitor selected from (lS,2S,3R,4S,5S)-5-(l,3-dihydroxypropan-2-ylamino)-l- (hydroxymethyl)cyclohexane-l,2,3,4-tetraol (chemical structure shown below) and
pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000072_0002
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is an alpha-glucosidase inhibitor selected from: acarbose, miglitol, and voglibose. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is acarbose. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is miglitol. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is voglibose.
Insulin and Insulin Analogues
The term "insulin analogue" refers to the naturally occurring human hormone and insulin receptor ligands (i.e., synthetic insulin analogues). Insulin receptor ligands are structurally different from the natural human hormone, but have substantially the same activity as human insulin in terms of glycemic control. Examples of an insulin analogue include, NPH insulin (also known as Humulin N, Novolin N, NPH Lletin II, and insulin isophane), insulin lispro (28B-L-lysine-29B-L-proline-insulin, wherein insulin is human insulin), insulin aspart (28B-L-aspartic acid-insulin, wherein insulin is human insulin), insulin glulisine (3B-L-lysine- 29B-L-glutamic acid-insulin, wherein insulin is human insulin), and insulin analogues known in the art.
NPH insulin is marketed by Eli Lilly and Company under the name Humulin N, and is considered as an intermediate-acting insulin analogue given to help control the blood sugar level of those with diabetes. Insulin lispro is marketed by Eli Lilly and Company under the name Humalog, and is considered a rapid acting insulin analogue. Insulin aspart is marketed by Novo Nordisk and sold as NovoRapid. Insulin aspart is considered a fast acting insulin analogue. Insulin glulisine was developed by Sanofi-Aventis and is sold under the trade name Apidra. Insulin glulisine is considered a rapid acting insulin analogue but shorter duration of action compared to human insulin.
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is an insulin analogue selected from NPH insulin and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is an insulin analogue selected from insulin lispro and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is an insulin analogue selected from insulin aspart and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is an insulin analogue selected from insulin glulisine and pharmaceutically acceptable salts, solvates, and hydrates thereof.
Sulfonylureas
The sulfonylureas are drugs which promote secretion of insulin from pancreatic beta cells by transmitting signals of insulin secretion via receptors in the cell membranes. Examples of a sulfonylurea include tolbutamide (Orinase, Ar-(butylcarbamoyl)-4- methylbenzenesulfonamide); acetohexamide (Dymelor, 4-acetyl-Ar-
(cyclohexylcarbamoyl)benzenesulfonamide); tolazamide (Tolinase, .V-(azepan-l-ylcarbamoyl)- 4-methylbenzenesulfonamide); chlorpropamide (Diabinese, 4-chloro-Ar- (propylcarbamoyl)benzenesulfonamide); glipizide (Glucotrol, N-(4-(N-
(cyclohexylcarbamoyl)sulfamoyl)phenethyl)-5-methylpyrazine-2-carboxamide); glibenclamide, also known as glyburide (Diabeta, Micronase, Glynase, 5-chloro-Ar-(4-(Ar- (cyclohexylcarbamoyl)sulfamoyl)phenethyl)-2-methoxybenzamide); glimepiride (Amaryl, 3- ethyl-4-methyl-Ar-(4-(Ar-((lr,4r)-4-methylcyclohexylcarbamoyl)sulfamoyl)phenethyl)-2-oxo- 2,5-dihydro- l/i-pyrrole-l-carboxamide); gliclazide (Diamicron, Λ'- (hexahydrocyclopenta[c]pyrrol-2(l//)-ylcarbamoyl)-4-methylbenzenesulfonamide); and sulfonylureas known in the art.
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from sulfonylureas:
Ar-(4-(Ar-(cyclohexylcarbamoyl)sulfamoyl)phenethyl)-5-methylpyrazine-2- carboxamide); 5-chloro-Ar-(4-(Ar-(cyclohexylcarbamoyl)sulfamoyl)phenethyl)-2- methoxybenzamide; 3-ethyl-4-methyl-Ai-(4-(Ai-((lr,4r)-4- methylcyclohexylcarbamoyl)sulfamoyl)phenethyl)-2-oxo-2,5-dihydro- 1/i-pyrrole- 1 - carboxamide; and pharmaceutically acceptable salts, solvates, and hydrates thereof.
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from Ar-(butylcarbamoyl)-4-methylbenzenesulfonamide (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000074_0001
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from 4-acetyl-AL(cyclohexylcarbamoyl)benzenesulfonamide (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000074_0002
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from Ar-(azepan-l-ylcarbamoyl)-4-methylbenzenesulfonamide (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000074_0003
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from 4-chloro-AL(propylcarbamoyl)benzenesulfonamide (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000074_0004
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from Ar-(4-(Ar-(cyclohexylcarbamoyl)sulfamoyl)phenethyl)-5- methylpyrazine-2-carboxamide (chemical structure shown below) and pharmaceutically acceptable salts, sol
Figure imgf000074_0005
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from 5-chloro-Ar-(4-(Ar-(cyclohexylcarbamoyl)sulfamoyl)phenethyl)-2- methoxybenzamide (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000075_0001
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from 3-ethyl-4-methyl-Ar-(4-(Ar-((lr,4r)-4- methylcyclohexylcarbamoyl)sulfamoyl)phenethyl)-2-oxo-2,5-dihydro- 1/i-pyrrole- 1 - carboxamide (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000075_0002
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from Ar-(hexahydrocyclopenta[c]pyrrol-2(l//)-ylcarbamoyl)-4- methylbenzenesulfonamide (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000075_0003
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from the following sulfonylureas and pharmaceutically acceptable salts, solvates, and hydrates thereof: glipizide, glimepiride, and glibenclamide. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is tolbutamide. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is acetohexamide. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is tolazamide. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is chlorpropamide. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is glipizide. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is glyburide. In some embodiments, the pharmaceutical agent or the second
pharmaceutical agent is glimepiride. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is gliclazide. SGLT2 inhibitors
Sodium-glucose transporter-2 (SGLT2) inhibitors belong to the class of drugs which inhibit the protein SGLT2 and the reabsorption of glucose in the kidney. The inhibition by SGLT2 inhibitors retard, diminish, or otherwise reduce the amount of glucose that is reabsorbed and therefore is eliminated in the urine. Some representative examples of SGLT2 inhibitors include dapagliflozin ((2lS,,3R,4R,5.S,,6R)-2-(4-chloro-3-(4-ethoxybenzyl)phenyl)-6- (hydroxymethyl)tetrahydro-2/i-pyran-3,4,5-triol, Bristol-Myers Squibb and AstraZeneca), remogliflozin (ethyl ((2R,3lS',4lS',5R,6lSr)-3,4,5-trihydroxy-6-(4-(4-isopropoxybenzyl)-l-isopropyl- 5-methyl- l/i-pyrazol-3-yloxy)tetrahydro-2/i-pyran-2-yl)methyl carbonate, Glaxo SmithKline), ASP1941 (Kotobuki/Astellas), canagliflozin ((25,3^,4^,55,6^)-2-(3-((5-(4- fluorophenyl)thiophen-2-yl)methyl)-4-methylphenyl)-6-(hydroxymethyl)tetrahydro-2/i-pyran- 3,4,5-triol, Johnson & Johnson/Mitsubishi/Tanabe), ISIS 388626 (an antisense oligonucleotide, Isis Pharmaceuticals), sergliflozin (ethyl ((2R,3S,4S,5R,6S)-3,4,5-trihydroxy-6-(2-(4- methoxybenzyl)phenoxy)tetrahydro-2/i-pyran-2-yl)methyl carbonate, GlaxoSmithKline), AVE2268 ((2R,3.S,,4lS,,5R,6lS,)-2-(hydroxymethyl)-6-(2-(4-methoxybenzyl)thiophen-3- yloxy)tetrahydro-2H-pyran-3,4,5-triol, Sanofi-Aventis), BI10773 (Boehringer Ingelheim), CSG453 (Chugai/Roche), LX4211 (Lexicon), and SGLT2 inhibitors known in the art.
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a SGLT2 inhibitor selected from the following SGLT2 inhibitors:
(2lS,,3R,4R,5,S,,6R)-2-(4-chloro-3-(4-ethoxybenzyl)phenyl)-6-(hydroxymethyl)tetrahydro- 2#-pyran-3,4,5-triol; ethyl ((2R,3.S,,4lS,,5R,6lS,)-3,4,5-trihydroxy-6-(4-(4-isopropoxybenzyl)-l- isopropyl-5-methyl- l/i-pyrazol-3-yloxy)tetrahydro-2/i-pyran-2-yl)methyl carbonate; ethyl ((2R,3lS',4lS',5R,6lSr)-3,4,5-trihydroxy-6-(2-(4-methoxybenzyl)phenoxy)tetrahydro-2/i-pyran-2- yl)methyl carbonate; (2R,3lS',4lS',5R,6lSr)-2-(hydroxymethyl)-6-(2-(4-methoxybenzyl)thiophen-3- yloxy)tetrahydro-2H-pyran-3,4,5-triol; (2lS,,3R,4R,5.S,,6R)-2-(3-((5-(4-fluorophenyl)thiophen-2- yl)methyl)-4-methylphenyl)-6-(hydroxymethyl)tetrahydro-2/i-pyran-3,4,5-triol; and pharmaceutically acceptable salts, solvates, and hydrates thereof.
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from (2S,3R,4R,5S,6R)-2-(4-chloro-3-(4-efhoxybenzyl)phenyl)-6- (hydroxymethyl)tetrahydro-2/i-pyran-3,4,5-triol (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000076_0001
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from ethyl ((2R,3lS',4lS',5R,6lSr)-3,4,5-trihydroxy-6-(4-(4-isopropoxybenzyl)- l-isopropyl-5-methyl-l/i-pyrazol-3-yloxy)tetrahydro-2/i-pyran-2-yl)methyl carbonate (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000077_0001
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from ethyl ((2R,3S,4S,5R,6¾-3,4,5-trihydroxy-6-(2-(4- methoxybenzyl)phenoxy)tetrahydro-2/i-pyran-2-yl)methyl carbonate (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000077_0002
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a SGLT2 inhibitor selected from: dapagliflozin, remigliflozin, and sergliflozin. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is dapagliflozin. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is remigliflozin. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is sergliflozin.
Astellas and Kotobuki disclosed a series of SGLT2 inhibitors in international patent publication WO2004/080990. Some embodiments of the present invention include every combination of one or more compounds selected from compounds disclosed in
WO2004/080990 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
Aventis disclosed a series of SGLT2 inhibitors in international patent publication WO2004/007517. Some embodiments of the present invention include every combination of one or more compounds selected from compounds disclosed in WO2004/007517 and pharmaceutically acceptable salts, solvates, and hydrates thereof. One such compound is (2R,3S, 4lS,,5R,6,S,)-2-(hydroxymethyl)-6-(2-(4-methoxybenzyl)thiophen-3-yloxy)tetrahydro-2H- pyran-3,4,5-triol. In some embodiments, the SGLT2 inhibitor is selected from (2R,3S,4S,5R,6S)- 2-(hydroxymethyl)-6-(2-(4-methoxybenzyl)thiophen-3-yloxy)tetrahydro-2H-pyran-3,4,5-triol, and pharmaceutically accepta hydrates thereof:
Figure imgf000078_0001
Tanabe disclosed a series of SGLT2 inhibitors in international patent publication WO2005/012326. Some embodiments of the present invention include every combination of one or more compounds selected from compounds disclosed in WO2005/012326 and pharmaceutically acceptable salts, solvates, and hydrates thereof. One such compound is (25,3R,4R,55,6R)-2-(3-((5-(4-fluorophenyl)thiophen-2-yl)methyl)-4-methylphenyl)-6- (hydroxymethyl)tetrahydro-2/i-pyran-3,4,5-triol. In some embodiments, the SGLT2 inhibitor is selected from (25,3R,4R,55,6R)-2-(3-((5-(4-fluorophenyl)thiophen-2-yl)methyl)-4- methylphenyl)-6-(hydroxymethyl)tetrahydro-2/i-pyran-3,4,5-triol, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000078_0002
Boehringer Ingelheim disclosed a series of SGLT2 inhibitors in international patent publication WO2005/092877. Some embodiments of the present invention include every combination of one or more compounds selected from compounds disclosed in
WO2005/092877 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
Chugai disclosed a series of SGLT2 inhibitors in international patent publication WO2006/080421. Some embodiments of the present invention include every combination of one or more compounds selected from compounds disclosed in WO2006/080421 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
Lexicon disclosed a series of SGLT2 inhibitors in international patent publication WO2008/109591. Some embodiments of the present invention include every combination of one or more compounds selected from compounds disclosed in WO2008/109591 and pharmaceutically acceptable salts, solvates, and hydrates thereof.
Meglitinides The meglitinides promote secretion of insulin by binding to the pancreatic beta cells in a similar manner as sulfonylureas but at an alternative binding site. Examples of meglitinides include Novo Nordisk's repaglinide (Prandin, (¾-2-ethoxy-4-(2-(3-methyl-l-(2-(piperidin-l- yl)phenyl)butylamino)-2-oxoethyl)benzoic acid), nateglinide (Starlix, (R)-2-((lr,4R)-4- isopropylcyclohexanecarboxamido)-3-phenylpropanoic acid), mitiglinide ((lSr)-2-benzyl-4- ((3aR aS) H^so dol-2(3H,3aHAH,5H,6H H aH)-yl)-4-oxob tanoic acid), and the like.
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a meglitinide selected from the following meglitinides: (¾-2-ethoxy-4-(2-(3-mefhyl- l-(2- (piperidin- l-yl)phenyl)butylamino)-2-oxoethyl)benzoic acid; (R)-2-((lr,4R)-4- isopropylcyclohexanecarboxamido)-3-phenylpropanoic acid; (S)-2-benzyl-4-((3aR,7aS)- IH- isoindol-2(3/i,3a/i,4/i,5/i,6/i,7/i,7a//)-yl)-4-oxobutanoic acid; and pharmaceutically acceptable salts, solvates, and hydrates thereof.
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is (S)-2-efhoxy-4-(2-(3-mefhyl- 1 -(2-(piperidin- 1 -yl)phenyl)butylamino)-2-oxoethyl)benzoic acid (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000079_0001
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from (R)-2-((lr,4R)-4-isopropylcyclohexanecarboxamido)-3- phenylpropanoic acid (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000079_0002
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a sulfonylurea selected from (S)-2-benzyl-4-((3aR,7aS)- l/i-isoindol-
2(3/i,3a/i,4/i,5/i,6/i,7/i,7a//)-yl)-4-oxobutanoic acid (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000080_0001
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a meglitinide selected from the following meglitinides: repaglinide, nateglinide, mitiglinide, and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a meglitinide selected from repaglinide and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a meglitinide selected from nateglinide and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a meglitinide selected from mitiglinide and pharmaceutically acceptable salts, solvates, and hydrates thereof.
Thiazolidinediones
Thiazolidinediones belong to the class of drugs more commonly known as TZDs. These drugs act by binding to the nuclear receptor peroxisome proliferator-activated receptor gamma (PPARy) activate transcription of a number of specific genes leading to a decrease in insulin resistance. Examples of thiazolidinediones include rosiglitazone (Avandia, 5-(4-(2- (methyl(pyridin-2-yl)amino)ethoxy)benzyl)thiazolidine-2,4-dione), pioglitazone (Actos, 5-(4-(2- (5-ethylpyridin-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione), troglitazone (Rezulin, 5-(4-((6- hydroxy-2,5,7,8-tetramethylchroman-2-yl)methoxy)benzyl)thiazolidine-2,4-dione), rivoglitazone (5-(4-((6-methoxy-l-methyl-l/i-benzo[d]imidazol-2- yl)methoxy)benzyl)thiazolidine-2,4-dione), ciglitazone(5-(4-((l- methylcyclohexyl)methoxy)benzyl)thiazolidine-2,4-dione), and thiazolidinediones known in the art.
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a meglitinide selected from: 5-(4-(2-(methyl(pyridin-2-yl)amino)ethoxy)benzyl)thiazolidine-2,4- dione; 5-(4-(2-(5-ethylpyridin-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione; 5-(4-((6-methoxy-l/i- benzo[d]imidazol-2-yl)methoxy)benzyl)thiazolidine-2,4-dione; 5-(4-((l- methylcyclohexyl)methoxy)benzyl)thiazolidine-2,4-dione; and pharmaceutically acceptable salts, solvates, and hydrates thereof.
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is 5-(4-(2-(methyl(pyridin-2-yl)amino)ethoxy)benzyl)thiazolidine-2,4-dione (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000081_0001
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is 5-(4-(2-(5-ethylpyridin-2-yl)ethoxy)benzyl)thiazolidine-2,4-dione (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000081_0002
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is 5-(4-((6-hydroxy-2,5,7,8-tetramethylchroman-2-yl)methoxy)benzyl)thiazolidine-2,4-dione (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000081_0003
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is 5-(4-((6-methoxy-l -methyl- l/i-benzo[d]imidazol-2-yl)methoxy)benzyl)thiazolidine-2,4-dione (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000081_0004
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is 5-(4-((l-methylcyclohexyl)methoxy)benzyl)thiazolidine-2,4-dione (chemical structure shown below) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000081_0005
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a thiazolidinedione selected from rosiglitazone and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a thiazolidinedione selected from pioglitazone and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a thiazolidinedione selected from troglitazone and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a thiazolidinedione selected from rivoglitazone and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is a
thiazolidinedione selected from ciglitazone and pharmaceutically acceptable salts, solvates, and hydrates thereof.
Anti-Diabetic Peptide Analogues
Anti-diabetic peptide analogues are peptides that promote secretion of insulin by acting as an incretin mimetic, such as, GLP-1 and GIP. Examples of an anti-diabetic peptide analog include, exenatide, liraglutide, taspoglutide, and anti-diabetic peptides analogues know in the art.
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is an anti-diabetic peptide analogue selected from: exenatide; liraglutide; and taspoglutide. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is exenatide. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is liraglutide. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is taspoglutide.
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is L-histidylglycyl-L-a-glutamylglycyl-L-threonyl-L-phenylalanyl-L-threonyl-L-seryl-L-a- aspartyl-L-leucyl-L-seryl-L-lysyl-L-glutaminyl-L-methionyl-L-a-glutamyl-L-a-glutamyl-L-a- glutamyl-L-alanyl-L-valyl-L-arginyl-L-leucyl-L-phenylalanyl-L-isoleucyl-L-a-glutamyl-L- tryptophyl-L-leucyl-L-lysyl-L-asparaginylglycylglycyl-L-prolyl-L-seryl-L-serylglycyl-L-alanyl- L-prolyl-L-prolyl-L-prolyl- L-serinamide (i.e., exenatide) and pharmaceutically acceptable salts, solvates, and hydrates thereof.
In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is L-histidyl-L-alanyl-L-a-glutamylglycyl-L-threonyl-L-phenylalanyl-L-threonyl-L-seryl-L-a- aspartyl-L-valyl-L-seryl-L-seryl-L-tyrosyl-L-leucyl-L-a-glutamylglycyl-L-glutaminyl-L-alanyl- L-alanyl-N6-[Ar-(l-oxohexadecyl)-L-a-glutamyl]-L-lysyl-L-a-glutamyl-L-phenylalanyl-L- isoleucyl-L-alanyl-L-tryptophyl-L-leucyl-L-valyl-L-arginylglycyl-L-arginyl-glycine
(liraglutide) and pharmaceutically acceptable salts, solvates, and hydrates thereof. In some embodiments, the pharmaceutical agent or the second pharmaceutical agent is H2N-His-2-methyl-Ala-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Gly-Gln-Ala- Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Val-Lys-2-methyl-Ala-Arg-CONH2 (taspoglutide) and pharmaceutically acceptable salts, solvates, and hydrates thereof.
Acyl-CoA:Diacylglycerol Acyltransferases (DGAT-1) Inhibitors
Triglycerides (TG) represent the major form of energy stored in eukaryotes. Disorders and/or imbalances in triglyceride metabolism are implicated in the pathogenesis of and increased risk for metabolic related disorders, such as, obesity, insulin resistance syndrome and type 2 diabetes, nonalcoholic fatty liver disease, and coronary heart disease.
Triglyceride biosynthesis and the resulting TG burden on tissues are largely controlled by two major pathways in humans. Both of these pathways converge at an intermediate diacylglycerols (DAG) which are subsequently converted to triglycerides through acylation by a fatty acid acyl-CoA catalyzed by the acyl-CoA:diacylglycerol acyltransferases (DGAT). DGAT consists of DGAT-1 and DGAT-2. Inhibition of acyl-CoA:diacylglycerol acyltransferase-1 (DGAT-1) is one of the most advanced targets for altering lipid biosyntheis as evidenced by on going human clinical trials.
In some embodiments, the pharmaceutical agent or said second pharmaceutical agent is a DGAT-1 inhibitor known in the art.
In some embodiments, the pharmaceutical agent or said second pharmaceutical agent is a DGAT-1 inhibitor selected from the DGAT-1 inhibitors described in the following patent applications and pharmaceutically acceptable salts, solvates, and hydrates thereof: WO 2004/047755, WO 2004/100881, WO 2005/0727401, WO 2006/044775, WO 2006/06019020, WO 2006/082010, WO2006/113919, WO 2006/134317, WO2007/126957, WO2012/051488, WO2009/016462.
In some embodiments, the pharmaceutical agent or said second pharmaceutical agent is a DGAT-1 inhibitor selected from the following DGAT-1 inhibitors and pharmaceutically acceptable salts, solvates, and hydrates thereof:
(lr,4r)-4-(3-fluoro-4-(5-(4-(trifluoromethyl)phenylamino)-l,3,4-oxadiazole-2- carboxamido)phenyl)cyclohexanecarboxylic acid (see WO2010/070343, Example 1);
(lr,4r)-4-(4-(5-(4-(difluoromethoxy)phenylamino)-l,3,4-oxadiazole-2-carboxamido)-3- fluorophenyl)cyclohexanecarboxylic acid (see WO2010/070343, Example 2);
(lr,4r)-4-(3-fluoro-4-(5-(4-(trifluoromethoxy)phenylamino)-l,3,4-oxadiazole-2- carboxamido)phenyl)cyclohexanecarboxylic acid (see WO2010/070343, Example 3);
(lr,4r)-4-(4-(5-(3-chlorophenylamino)-l,3,4-oxadiazole-2-carboxamido)-3- fluorophenyl)cyclohexanecarboxylic acid (see WO2010/070343, Example 4); (l r,4r)-4-(4-(5-(3,4-difluorophenylamino)- l,3,4-oxadiazole-2-carboxamido)-3- fluorophenyl)cyclohexanecarboxylic acid (see WO2010/070343, Example 5);
(lr,4r)-4-(3-fluoro-4-(5-(2,4,5-trifluorophenylamino)-l,3,4-oxadiazole-2- carboxamido)phenyl)cyclohexanecarboxylic acid (see WO2010/070343, Example 6);
(l5,45)-4-(4-(5-(3,4-difluorophenylamino)- l,3,4-oxadiazole-2-carboxamido)-3- fluorophenyl)cyclohexanecarboxylic acid (see WO2010/070343, Example 7);
(l5,45)-4-(3-fluoro-4-(5-(2,4,5-trifluorophenylamino)-l,3,4-oxadiazole-2- carboxamido)phenyl)cyclohexanecarboxylic acid (see WO2010/070343, Example 8);
trans-(4- { 4- [5-(6-trifluoromethyl-pyridin-3ylamino)-pyridin-2-yl] -phenyl } -cyclohexyl)- acetic acid (see WO2007/126957, Example 5-1 and WO2012/051488, Examples 1-10 for formulations related thereto; alternative name: 2-((lr,4r)-4-(4-(5-(6-(trifluoromethyl)pyridin-3- ylamino)pyridin-2-yl)phenyl)cyclohexyl)acetic acid); and
{ ira«5-4-(4-(4-amino-5-oxo-7,8-dihydropyrimido[5.4-f] [fjoxazepin- 6(5//)yl)phenyllcyclohexyl } acetic acid (see WO2009/016462, Example 2; alternative name: 2- ((lr,4r)-4-(4-(4-amino-5-oxo-7,8-dihydropyrimido[5,4-f] [l,4]oxazepin-6(5H)- yl)phenyl)cyclohexyl)acetic acid).
Other Utilities
Another object of the present invention relates to radio-labeled compounds of the present invention that would be useful not only in radio-imaging but also in assays, both in vitro and in vivo, for localizing and quantitating GPR119 receptors in tissue samples, including human and for identifying GPR119 receptor ligands by inhibition binding of a radio-labeled compound. It is a further object of this invention to develop novel GPR119 receptor assays of which comprise such radio-labeled compounds.
The present disclosure includes all isotopes of atoms occurring in the present compounds, intermediates, salts and crystalline forms thereof. Isotopes include those atoms having the same atomic number but different mass numbers. One aspect of the present invention includes every combination of one or more atoms in the present compounds, intermediates, salts, and crystalline forms thereof that is replaced with an atom having the same atomic number but a different mass number. One such example is the replacement of an atom that is the most naturally abundant isotope, such as ¾ or 12C, found in one the present compounds, intermediates, salts, and crystalline forms thereof, with a different atom that is not the most
2 3 1 11 13 14 12 naturally abundant isotope, such as H or H (replacing H), or C, C, or C (replacing C). A compound wherein such a replacement has taken place is commonly referred to as being an isotopically-labeled compound. Isotopic-labeling of the present compounds, intermediates, salts, and crystalline forms thereof can be accomplished using any one of a variety of different synthetic methods know to those of ordinary skill in the art and they are readily credited with understanding the synthetic methods and available reagents needed to conduct such isotopic- labeling. By way of general example, and without limitation, isotopes of hydrogen include 2H (deuterium) and 3H (tritium). Isotopes of carbon include nC, 13C, and 14C. Isotopes of nitrogen
13 15 15 17 18
include N and N. Isotopes of oxygen include O, O, and C. An isotope of fluorine includes 18F. An isotope of sulfur includes 35S. An isotope of chlorine includes 36C1. Isotopes of bromine include 75Br, 76Br, 77Br, and 82Br. Isotopes of iodine include 1231, 124I, 125I, and 131I. Another aspect of the present invention includes compositions, such as, those prepared during synthesis, preformulation, and the like, and pharmaceutical compositions, such as, those prepared with the intent of using in a mammal for the treatment of one or more of the disorders described herein, comprising one or more of the present compounds, intermediates, salts, and crystalline forms thereof, wherein the naturally occurring distribution of the isotopes in the composition is perturbed. Another aspect of the present invention includes compositions and pharmaceutical compositions comprising compounds as described herein wherein the compound is enriched at one or more positions with an isotope other than the most naturally abundant isotope. Methods are readily available to measure such isotope perturbations or enrichments, such as, mass spectrometry, and for isotopes that are radio-isotopes additional methods are available, such as, radio-detectors used in connection with HPLC or GC.
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 Drawings 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 the scarcer 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. These synthetic methods, for example, incorporating activity levels of tritium into target molecules, are as follows:
A. Catalytic Reduction with Tritium Gas: This procedure normally yields high specific activity products and requires halogenated or unsaturated precursors. B. Reduction with Sodium Borohydride [¾] : This procedure is rather inexpensive and requires precursors containing reducible functional groups such as aldehydes, ketones, lactones, esters and the like.
C. Reduction with Lithium Aluminum Hydride [3H] : This procedure offers products at almost theoretical specific activities. It also requires precursors containing reducible functional groups such as aldehydes, ketones, lactones, esters and the like.
D. Tritium Gas Exposure Labeling: This procedure involves exposing precursors containing exchangeable protons to tritium gas in the presence of a suitable catalyst.
E. N-Mefhylation using Methyl Iodide [3H] : This procedure is usually employed to prepare O-methyl or N-mefhyl (3H) products by treating appropriate precursors with high specific activity methyl iodide (3H). This method in general allows for higher specific activity, such as for example, about 70-90 Ci/mmol.
Synthetic methods for incorporating activity levels of 125I into target molecules include:
A. Sandmeyer and like reactions: This procedure transforms an aryl amine or a heteroaryl amine into a diazonium salt, such as a diazonium tetrafluoroborate salt and subsequently to 125I labeled compound using Na125I. A represented procedure was reported by Zhu, G-D. and co-workers in /. Org. Chem. , 2002, 67, 943-948.
B. Ortho 125Iodination of phenols: This procedure allows for the incorporation of 125I at the ortho position of a phenol as reported by Collier, T. L. and co-workers in J. Labelled Compd. Radiopharm. , 1999, 42, S264-S266.
C. Aryl and heteroaryl bromide exchange with 125I: This method is generally a two step process. The first step is the conversion of the aryl or heteroaryl bromide to the corresponding tri-alkyltin intermediate using for example, a Pd catalyzed reaction [i.e. Pd(Ph3P)4] or through an aryl or heteroaryl lithium, in the presence of a tri-alkyltinhalide or hexaalkylditin [e.g., (CH3)3SnSn(CH3)3] . A representative procedure was reported by Le Bas, M.-D. and co-workers in /. Labelled Compd. Radiopharm. 2001, 44, S280-S282.
A radiolabeled GPR119 receptor compound of Formula (I) 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 "radiolabeled compound of Formula (I)" to a GPR119 receptor. Accordingly, the ability of a test compound to compete with the "radio-labeled compound of Formula (I)" for the binding to a GPR119 receptor directly correlates to its binding affinity.
Certain labeled compounds of the present invention bind to certain GPR119 receptors. In one embodiment the labeled compound has an IC50 less than about 500 μΜ, in another embodiment the labeled compound has an IC50 less than about 100 μΜ, in yet another embodiment the labeled compound has an IC50 less than about 10 μΜ, in yet another embodiment the labeled compound has an IC50 less than about 1 μΜ and in still yet another embodiment the labeled inhibitor has an IC50 less than about 0.1 μΜ.
Other uses of the disclosed receptors and methods will become apparent to those skilled in the art based upon, inter alia, a review of this disclosure.
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: Syntheses of Compounds of the Present Invention.
Illustrated syntheses for compounds of the present invention are shown in Figures 4 through 7 where the variables have the same definitions as used throughout this disclosure.
The compounds of the invention and their syntheses are further illustrated by 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, supra and infra, are named according to AutoNom version 2.2, AutoNom 2000, CS ChemDraw Ultra Version 7.0.1, or 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 (¾ NMR) spectra were recorded on 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, dd = doublet of doublets, ddd = doublet of doublet of doublets, dt = doublet of triplets, t = triplet, td = triplet of doublets, tt = triplet of triplets, q = quartet, quin = quintet, m = multiplet, bs = broad singlet, bt = broad triplet. Microwave irradiations were carried out using a Smith Synthesizer™ or an Emrys Optimizer™ (Biotage™). Thin-layer chromatography (TLC) was performed on silica gel 60 F254 (Merck), preparatory thin-layer chromatography (prep TLC) was preformed 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 under reduced pressure on a Biichi rotary evaporator.
LCMS spec: HPLC -pumps: LC-10AD VP, Shimadzu Inc.; HPLC system controller: SCL-IOA VP, Shimadzu Inc; UV-Detector: SPD-10A 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. Example 1.1: Preparation of 1-Methylcyclopropyl 4-((lr,4r)-4-(3-Cyanopyrazin-2- yloxy)cyclohexyloxy)piperidine-l-carboxylate (Compound 1).
Step 1: Preparation of 1-Methylcyclopropyl 4-((lr,4r)-4-Hydroxycyclohexyloxy) piperidine- 1 -carboxylate.
To a mixture of (lr,4r)-4-(piperidin-4-yloxy)cyclohexanol (2.45 g, 12.29 mmol) and triethylamine (2 mL, 14.35 mmol) in 100 mL CH2C12 was added 2,5-dioxopyrrolidin-l-yl 1- methylcyclopropyl carbonate (2.7 g, 12.66 mmol, containing approximately 5% isobutyl carbonate). After stirring at room temperature (rt) for 1 h the mixture was transferred into a separatory funnel and extracted with water. The organic phase was dried over MgS04, filtered, and concentrated. The residue was purified by Biotage® column chromatography (Si02, hexane/AcOEt gradient) to give 1- methylcyclopropyl 4-((lr,4r)-4-hydroxycyclohexyloxy)piperidine-l -carboxylate (3.62 g, 12.17 mmol, 99.0%). Exact mass calculated for C16H27N04: 297.19, found: LC/MS m/z = 298.2 (M+H+); ¾ NMR (400 MHz, CDC13) δ ppm 0.59-0.63 (m, 2H), 0.84-0.87 (m, 2H), 1.29-1.37 (m, 4H), 1.40- 1.53 (m, 2H), 1.54 (s, 3H), 1.73-1.78 (m, 2H), 1.92-1.98 (m, 4H), 3.06-3.1 1 (m, 2H), 3.35-3.39 (m, 1H), 3.51-3.57 (m, 1H), 3.66-3.74 (m, 3H).
Step 2: Preparation of 1-Methylcyclopropyl 4-((lr,4r)-4-(3-Cyanopyrazin-2- yloxy)cyclohexyloxy)piperidine-l-carboxylate (Compound 1).
To solution of 1 -methylcyclopropyl 4-((lr,4r)-4-hydroxycyclohexyloxy)piperidine-l- carboxylate (70 mg, 0.235 mmol) in THF at rt was added potassium 2-methylpropan-2-olate (0.353 mL, 0.353 mmol) under N2. After stirring for 15 min at room temperature, the reaction mixture was treated with 3-chloropyrazine-2-carbonitrile (49.27 mg, 0.353 mmol). The reaction mixture was stirred for an additional 20 min, quenched with 1 N HC1, diluted with H20, and extracted with EtOAc (twice). The combined organic layers were washed with 1 N HC1 and brine, dried, and concentrated. The residue was purified by column chromatography to afford the title compound (75 mg, 80%). Exact mass calculated for C2iH28N404: 400.2, found: LC/MS m/z = 401.6 (M+H+); ¾ NMR (400 MHz, CDC13) δ ppm 0.59-0.64 (m, 2H), 0.83-0.88 (m, 2H), 1.45-1.58 (m, 4H), 1.54 (s, 3H), 1.64-1.74 (m, 2H), 1.74-1.84 (m, 2H), 1.96-2.05 (m, 2H), 2.08-2.17 (m, 2H), 3.06-3.18 (m, 2H), 3.52-3.60 (m, 2H), 3.64-3.86 (bs, 2H). 5.17-5.25 (m, 1H), 8.23 (d, / = 2,5 Hz, 1H), 8.29 (d, / = 2.5 Hz, 1H).
Example 1.2: Preparation of 3-((lr,4r)-4-(l-(5-Chloropyrimidin-2-yl)piperidin-4- yloxy)cyclohexyloxy)pyrazine-2-carbonitrile (Compound 2).
Step 1: Preparation of (lr,4r)-4-(Piperidin-4-yloxy)cyclohexanol.
Method A
A mixture of (lr,4r)-4-(pyridin-4-yloxy)cyclohexanol (20.5 g, 106.1 mmol), 10% palladium on carbon (50% water, 22.5 g, 10.6 mmol), and sodium tetraborate decahydrate (borax, 4.05 g, 10.62 mmol) in 200 mL isopropanol (IPA) in a steel bomb was filled with hydogen to ca. 20 bar and stirred at 80 °C (oil bath). The steel bomb was refilled with hydrogen to ca. 20 bar three times within 8 h. After stirring over night, the steel bomb was refilled again to 20 bar (and then again after 4 and 8 hours). After stirring over the weekend, the reaction was complete. Solids were filtered off through celite, washed with additional IPA, and dried under high vacuum to afford the title compound (20.6 g, 103.4 mmol, 97.4%) as a white solid. Exact mass calculated for CnH21N02: 199.2, found: LC/MS m/z = 200.0 (M+H+); ¾ NMR (400 MHz, CDC13) δ ppm 1.25-1.51 (m, 8H), 1.81-1.90 (m, 2H), 1.91-2.00 (m, 4H), 2.55-2.64 (m, 2H), 3.04-3.12 (dt, / = 13.14, 3.79 Hz, 2H), 3.34-3.47 (m, 2H), 3.63-3.71 (m, 1H).
Method B
Using the following procedure (lr,4r)-4-(piperidin-4-yloxy)cyclohexanol was isoloated from a reaction utilizing (\r,Ar)l(\s, s) mixture of 4-(pyridin-4-yloxy)cyclohexanol. A mixture of 4-(pyridin-4-yloxy)cyclohexanol (-70:30 (trans/cis), 20.4 g, 105.6 mmol), 10% palladium on carbon (50% water, 22.4 g, 10.5 mmol) and sodium tetraborate decahydrate (4.0 g, 10.49 mmol) in 200 mL IPA in a steel bomb under ca. 22 bar hydrogen was stirred at 80 °C (oil bath) and refilled four times over the next 2 days. The pressure was released and the mixture was allowed to cool to rt while nitrogen was bubbled through it. The resulting solids were filtered off, washed with IPA, and the filtrate was partly concentrated whereupon a solid precipitated. The solid was filtered, washed with additional IPA, and dried under high vacuum to afford the title compound (10.8 g, 54.2 mmol) as a white solid. The filtrate was partly concentrated. The resulting solid was filtered and washed with additional IPA to give another 2.6 g (13.0 mmol) of the title compound.
Step 2: tert-Butyl 4-((lr,4r)-4-Hydroxycyclohexyloxy)piperidine-l-carboxylate.
To a suspension of (lr,4r)-4-(piperidin-4-yloxy)cyclohexanol (13.45 g, 67.49 mmol) and triethylamine (11 mL, 78.92 mmol) in 400 mL CH2C12, di-feri-butyl dicarbonate (16.2 g, 74.23 mmol) was added in small portions. After stirring at rt for 1 h, the solution was extracted with water. The organic phase was dried over MgS04, filtered, and concentrated. The residue was purified by Biotage® column chromatography (Si02, hexane/AcOEt gradient) to afford the title compound (19.61 g, 65.50 mmol, 97.0%) as a white solid. Exact mass calculated for Ci6H29N04: 299.21, found: LC/MS m/z = 300.4 (M+H+); ¾ NMR (400 MHz, CDC13) δ ppm 1.27-1.52 (m, 16H), 1.74-1.80 (m, 2H), 1.89-2.02 (m, 4H), 3.02-3.09 (m, 2H), 3.34-3.41 (m, 1H), 3.50-3.56 (m, 1H), 3.65-3.81 (m, 3H).
Step 3: Preparation of teri-Butyl 4-((lr,4r)-4-(3-Cyanopyrazin-2- yloxy)cyclohexyloxy)piperidine-l-carboxylate.
To a solution of ieri-butyl 4-((lr,4r)-4-hydroxycyclohexyloxy)piperidine-l-carboxylate (4 g, 13.36 mmol) in THF (10 mL) was added 1 M potassium i-butoxide in THF (16.03 mL, 16.03 mmol) at 0 °C under nitrogen. After stirring for 10 min, 3-chloropyrazine-2-carbonitrile (2.051 g, 14.70 mmol) in THF (6 mL) was added into the reaction. The reaction mixture was slowly warmed to rt and stirred for 1 h, then quenched with water and 1 N HC1, extracted with EtOAc, washed with brine, dried with anhydrous MgS04, and concentrated. The residue was purified by silica gel column chromatography with 40% ethyl acetate hexanes to afford the title compound (4.8 g, 11.93 mmol, 89.3% yield). Exact mass calculated for C21H30N4O4: 402.2, found LC/MS m/z = 403.2 (M+H+); :H NMR (400 MHz, CDC13) δ ppm 1.46 (s, 9H), 1.46-1.58 (m, 4H), 1.65-1.72 (m, 2H), 1.75-1.82 (m, 2H), 1.96-2.05 (m, 2H), 2.10-2.18 (m, 2H), 3.05-3.12 (m, 2H), 3.52-3.58 (m, 2H), 3.73-3.82 (m, 2H), 5.18-5.25 (m, 1H), 8.22 (d, / = 2.5 Hz, 1H), 8.29 (d, 7 = 2.5 Hz, 1H).
Step 4: Preparation of 3-((lr,4r)-4-(Piperidin-4-yloxy)cyclohexyloxy)pyrazine-2- carbonitrile Hydrochloride.
To a solution of ieri-butyl 4-((lr,4r)-4-(3-cyanopyrazin-2- yloxy)cyclohexyloxy)piperidine-l-carboxylate (4.8 g, 1 1.93 mmol) in anhydrous diethyl ether (20 mL) was added 4M hydrogen chloride in dioxane (29.81 mL, 1 19.3 mmol). The reaction mixture was stirred at rt overnight. The light yellow solid was filtered, washed with dioxane and dried to afford the title compound (3.95 g, 1 1.67 mmol, 97% yield). Exact mass calculated for C16H22N402: 302.2, found LC/MS m/z = 303.4 (M+H+).
Step 5: Preparation of 3-((lr,4r)-4-(l-(5-Chloropyrimidin-2-yl)piperidin-4- yloxy)cyclohexyloxy)pyrazine-2-carbonitrile (Compound 2).
The reaction mixture of 3-((l r,4r)-4-(piperidin-4-yloxy)cyclohexyloxy)pyrazine-2- carbonitrile hydrochloride (100 mg, 0.295 mmol), 2,5-dichloropyrimidine (87.94 mg, 0.590 mmol) and triethylamine (0.123 mL, 0.885 mmol) in IPA (2 mL) was heated at 120 °C for 3 h under microwave irradiation. The light yellow precipitate was collected, washed with IPA, dried to afford the title compound (114 mg, 0.275 mmol, 93.1% yield). Exact mass calculated for C20H23ClN6O2: 414.2, found LC/MS m/z = 415.4 (M+H+); ¾ NMR (400 MHz, CDC13) δ ppm 1.50-1.62 (m, 4H), 1.65-1.75 (m, 2H), 1.84- 1.92 (m, 2H), 1.99-2.07 (m, 2H), 2.10-2.18 (m, 2H), 3.39-3.46 (m, 2H), 3.58-3.71 (m, 2H), 4.19-4.26 (m, 2H), 5.18-5.25 (m, 1H), 8.21 (s, 2H), 8.22 (d, J = 2.5 Hz, 1H), 8.29 (d, 7 = 2.5 Hz, 1H).
Example 1.3: Preparation of (/f)-l,l,l-Trifluoropropan-2-yl 4-((lr,4r)-4-(3-Cyanopyrazin- 2-yloxy)cyclohexyloxy)piperidine-l-carboxylate (Compound 3).
(R)-l, l, l-Trifluoropropan-2-ol (0.297 g, 1.092 mmol) and Ι, Γ-carbonyldiimidazole (0.173 g, 1.066 mmol) in THF (3 mL) was stirred for 1 h at rt, then 3-((lr,4r)-4-(piperidin-4- yloxy)cyclohexyloxy)pyrazine-2-carbonitrile hydrochloride (0.1 g, 0.266 mmol) was added, followed by triethylamine (0.223 mL, 1.599 mmol). The reaction mixture was stirred at reflux overnight. Water was added, and the mixture was extracted with ethyl acetate. The combined organics were concentrated. The crude was purified by silica gel chromatography with 40% ethyl acetate/hexanes to afford the title compound (112 mg, 0.253 mmol, 95.0% yield) as an oil. Exact mass calculated for C^H^N^: 442.2, found LC/MS m/z = 443.4 (M+H+); ¾ NMR (400 MHz, CDC13) δ ppm 1.39 (d, J = 6.6 Hz, 3H), 1.48-1.58 (m, 4H), 1.64-1.73 (m, 2H), 1.75- 1.85 (m, 2H), 1.96-2.04 (m, 2H), 2.08-2.16 (m, 2H), 3.20-3.34 (m, 2H), 3.52-3.65 (m, 2H), 3.70-3.80 (m, 2H), 5.18-5.25 (m, 2H), 8.22 (d, / = 2.5 Hz, 1H), 8.28 (d, / = 2.5 Hz, 1H).
Example 1.4: Preparation of (S)-l,l,l-Trifluoropropan-2-yl 4-((lr,4r)-4-(3-Cyanopyrazin- 2-yloxy)cyclohexyloxy)piperidine-l-carboxylate (Compound 4).
(S)-l, l, l-trifluoropropan-2-ol (0.158 g, 1.092 mmol) and Ι, -carbonyldiimidazole (0.173 g, 1.066 mmol) in THF (3 mL) was stirred for 1 h at rt, then 3-((lr,4r)-4-(piperidin-4- yloxy)cyclohexyloxy)pyrazine-2-carbonitrile hydrochloride (0.1 g, 0.266 mmol) was added, followed by triethylamine (0.223 mL, 1.599 mmol). The reaction mixture was stirred at reflux overnight. Water was added, extracted with ethyl acetate. The combined organics were concentrated. The crude was purified by silica gel chromatography with 40% ethyl acetate/hexanes to afford the title compound (114 mg, 0.258 mmol, 96.7% yield) as oil. Exact mass calculated for C^H^N^: 442.2, found LC/MS m/z = 443.4 (M+H+); ¾ NMR (400 MHz, CDCI3) δ ppm 1.38 (d, J = 6.6 Hz, 3H), 1.48-1.58 (m, 4H), 1.64-1.73 (m, 2H), 1.75-1.85 (m, 2H), 1.96-2.04 (m, 2H), 2.08-2.16 (m, 2H), 3.20-3.34 (m, 2H), 3.52-3.65 (m, 2H), 3.70-3.80 (m, 2H), 5.18-5.25 (m, 2H), 8.22 (d, / = 2.5 Hz, 1H), 8.28 (d, / = 2.5 Hz, 1H).
Example 1.5: Preparation of (/f)-l-(2,2,2-Trifluoroethyl)pyrrolidin-3-yl 4-((lr,4r)-4-(3- Cyanopyrazin-2-yloxy)cyclohexyloxy)piperidine-l-carboxylate (Compound 5).
Step 1: Preparation of (/f)-l-(tert-Butoxycarbonyl)pyrrolidin-3-yl 4-((lr,4r)-4-(3- Cyanopyrazin-2-yloxy)cyclohexyloxy)piperidine-l-carboxylate.
(R)-ieri-Butyl 3-hydroxypyrrolidine-l-carboxylate (0.150 g, 0.799 mmol) and 1,1' - carbonyldiimidazole (0.130 g, 0.799 mmol) in THF (3 mL) was stirred for 1 h at rt, then 3- ((lr,4r)-4-(piperidin-4-yloxy)cyclohexyloxy)pyrazine-2-carbonitrile hydrochloride (0.1 g, 0.295 mmol) was added, followed by triethylamine (0.223 mL, 1.599 mmol). The reaction mixture was stirred at reflux overnight, diluted with water and extracted with ethyl acetate. The combined organics were concentrated. The residue was purified by silica gel column chromatography with 85% ethyl acetate/hexanes to give the title compound (102 mg, 0.198 mmol, 74.2% yield) as oil. Exact mass calculated for C26H37N506: 515.2, found LC/MS m/z = 516.4 (M+H+).
Step 2: Preparation of (/f)-Pyrrolidin-3-yl 4-((lr,4r)-4-(3-Cyanopyrazin-2- yloxy)cyclohexyloxy)piperidine-l-carboxylate Hydrochloride.
(R)-l-(ieri-Butoxycarbonyl)pyrrolidin-3-yl 4-((lr,4r)-4-(3-cyanopyrazin-2- yloxy)cyclohexyloxy)piperidine-l-carboxylate (102 mg, 0.198 mmol) in diethyl ether (1 mL) was added 4 N hydrogen chloride in dioxane (1.978 mL, 7.913 mmol). The reaction mixture was stirred at rt overnight. The white precipitate was collected and dried to give (R)-pyrrolidin-3-yl 4-((lr,4r)-4-(3-cyanopyrazin-2-yloxy)cyclohexyloxy)piperidine-l-carboxylate hydrochloride (89 mg, 0.197 mmol, 99.5% yield). Exact mass calculated for C2iH29N504: 414.2, found LC/MS m/z = 415.4 (M+H+).
Step 3: Preparation of (K)-l-(2,2,2-Trifluoroethyl)pyrrolidin-3-yl 4-((lr,4r)-4-(3- Cyanopyrazin-2-yloxy)cyclohexyloxy)piperidine-l-carboxylate (Compound 5).
To a stirred solution of (R)-pyrrolidin-3-yl 4-((lr,4r)-4-(3-cyanopyrazin-2- yloxy)cyclohexyloxy)piperidine-l-carboxylate hydrochloride (89 mg, 0.197 mmol) in DCM at rt was added triethylamine (82.34 μΐ, 0.591 mmol) and 2,2,2-trifluoroethyl
trifluoromethanesulfonate (91.41 mg, 0.394 mmol). The reaction mixture was stirred for 2 h at rt, then concentrated. The residue was purified by silica gel column chromatography with 70% ethyl acetate hexanes to afford the title compound (90 mg, 0.181 mmol, 91.9% yield). Exact mass calculated for C^HsoFsNsC : 497.2, found LC/MS m/z = 498.6 (M+H+); ¾ NMR (400 MHz, CDC13) δ ppm 1.48-1.58 (m, 4H), 1.64-1.73 (m, 2H), 1.75-1.85 (m, 2H), 1.87-1.96 (m, IH), 1.96-2.07 (m, 2H), 2.08-2.16 (m, 2H), 2.16-2.26 (m, IH), 2.68-2.74 (m, IH), 2.80-2.85 (dd, / = 3.5, 2.6 Hz, IH), 2.88-2.95 (m, IH), 3.05-3.15 (m, 3H), 3.15-3.24 (m, 2H), 3.53-3.62 (m, 2H), 3.72-3.82 (m, 2H), 5.12-5.17 (m, IH), 5.17-5.25 (m, IH), 8.21 (d, J = 2.5 Hz, IH), 8.28 (d, 7 = 2.5 Hz, IH).
Example 1.6: Preparation of (S)-l-(2,2,2-Trifluoroethyl)pyrrolidin-3-yl 4-((lr,4r)-4-(3- Cyanopyrazin-2-yloxy)cyclohexyloxy)piperidine-l-carboxylate (Compound 6).
To a stirred solution of (S)-pyrrolidin-3-yl 4-((lr,4r)-4-(3-cyanopyrazin-2- yloxy)cyclohexyloxy)piperidine-l-carboxylate hydrochloride (93 mg, 0.206 mmol), prepared in a manner similar to that described in Example 1.5 from Steps 1 and 2, starting with (S)-tert- butyl 3-hydroxypyrrolidine-l-carboxylate, in DCM (2 mL) at rt was added triethylamine (86.04 μΐ, 0.617 mmol) and 2,2,2-trifluoroethyl trifluoromethanesulfonate (95.52 mg, 0.412 mmol). The reaction mixture was stirred for 2 h at rt and concentrated. The residue was purified by silica gel column chromatography with 70% ethyl acetate/hexanes to afford the title compound (90 mg, 0.181 mmol, 87.9% yield). Exact mass calculated for CzsHsoFsNsC^: 497.2, found LC/MS m/z = 498.6 (M+H+); ¾ NMR (400 MHz, CDC13) δ ppm 1.48-1.58 (m, 4H), 1.64-1.73 (m, 2H), 1.75-1.85 (m, 2H), 1.87-1.96 (m, IH), 1.96-2.07 (m, 2H), 2.09-2.18 (m, 2H), 2.18-2.26 (m, IH), 2.68-2.74 (m, IH), 2.80-2.85 (dd, J = 3.5 and 2.6 Hz, IH), 2.88-2.95 (m, IH), 3.05- 3.15 (m, 3H), 3.15-3.24 (m, 2H), 3.53-3.62 (m, 2H), 3.72-3.82 (m, 2H), 5.12-5.17 (m, IH), 5.17-5.25 (m, IH), 8.21 (d, J = 2.5 Hz, IH), 8.28 (d, J = 2.5 Hz, IH). Example 1.7: Preparation of 3-((lr,4r)-4-(l-(3-(Trifluoromethyl)-l,2,4-oxadiazol-5- yl)piperidin-4-yloxy)cyclohexyloxy)pyrazine-2-carbonitrile (Compound 7).
Step 1: Preparation of (lr,4r)-4-(l-(3-(Trifluoromethyl)-l,2,4-oxadiazol-5- yl)piperidin-4-yloxy)cyclohexanol.
To a solution of 4-((lr,4r)-4-hydroxycyclohexyloxy)piperidine- l-carbonitrile (0.6 g, 2.675 mmol) and (Z)-2,2,2-trifluoro-Ar'-hydroxyacetimidamide (0.617 g, 4.815 mmol) in THF (5 mL) under nitrogen was added 0.5 M zinc(II) chloride in THF (16.05 mL, 8.025 mmol). The mixture was stirred overnight at 40 °C, cooled down, then 4 N hydrogen chloride in dioxane (4.012 mL, 16.05 mmol) was added and the mixture was heated at 120 °C for 5 h under microwave irradiation, quenched with aqueous saturated sodium bicarbonate and the organic layer was separated. The aqueous layer was extracted with DCM and the combined organic layers were concentrated. The residue was purified by silica gel column chromatography with 60% ethyl acetate hexanes to afford the title compound (256 mg, 0.763 mmol, 28.5% yield) as a light yellow oil. Exact mass calculated for C14H20F3N3O3: 335.1, found LC/MS m/z = 336.4 (M+H+).
Step 2: Preparation of -((lr,4r)-4-(l-(3-(Trifluoromethyl)-l,2,4-oxadiazol-5- yl)piperidin-4-yloxy)cyclohexyloxy)pyrazine-2-carbonitrile (Compound 7).
To a stirred solution of (lr,4r)-4-(l-(3-(trifluoromethyl)-l,2,4-oxadiazol-5-yl)piperidin- 4-yloxy)cyclohexanol (0.256 g, 0.763 mmol) and 3-chloropyrazine-2-carbonitrile (0.160 g, 1.145 mmol) in THF (3 mL) was added 1 M solution of potassium ieri-butoxide in THF (0.916 mL, 0.916 mmol) at ice- water bath under N2. The reaction mixture was stirred for 15 min, quenched with water, and extracted with EtOAc. The combined organics were dried with anhydrous MgS04, then concentrated. The residue was purified by silica gel column chromatography with 45% ethyl acetate/hexanes and semi preparative HPLC to afford the title compound (130 mg, 0.297 mmol, 38.8% yield) as oil, which became a white solid upon standing. Another ca. 60 mg was obtained in 95% purity. Exact mass calculated for
Ci9H21F3N603: 438.2, found LC/MS m/z = 439.4 (M+H+); ¾ NMR (400 MHz, CDC13) δ ppm 1.51-1.61 (m, 2H), 1.66-1.80 (m, 4H), 1.85- 1.95 (m, 2H), 1.98-2.08 (m, 2H), 2.10-2.18 (m, 2H), 3.56-3.65 (m, 3H), 3.70-3.78 (m, 1H), 3.80-3.88 (m, 2H), 5.20-5.26 (m, 1H), 8.23 (d, / = 2.5 Hz, 1H), 8.29 (d, / = 2.5 Hz, 1H).
Examplel.8: Preparation of 3-((lr,4r)-4-(l-(5-(Trifluoromethyl)-l,2,4-oxadiazol-3- yl)piperidin-4-yloxy)cyclohexyloxy)pyrazine-2-carbonitrile (Compound 8).
Step 1: Preparation of (lr,4r)-4-(l-(5-(Trifluoromethyl)-l,2,4-oxadiazol-3- yl)piperidin-4-yloxy)cyclohexanol.
To a solution of 50% hydroxylamine in water (0.277 mL, 4.521 mmol) was added a solution of 4-((lr,4r)-4-hydroxycyclohexyloxy)piperidine-l-carbonitrile (0.338 g, 1.507 mmol) in EtOH (4 mL). The reaction was stirred overnight at 60 °C. Then concentrated to dryness to give (Ζ)-Ν' -hydroxy-4-(( 1 r,4r)-4-hydroxycyclohexyloxy)piperidine- 1 -carboximidamide, which was dissolved in DCM (5 mL), triethylamine (0.525 mL, 3.767 mmol) was added at ice-water bath, followed by 2,2,2-trifluoroacetic anhydride (0.419 mL, 3.014 mmol). The reaction mixture was stirred for 30 min. Water was added, extracted with ethyl acetate. The combined organics were concentrated. The residue was purified by silica gel column chromatography with 60% ethyl acetate hexanes to afford the title compound (180 mg, 0.537 mmol, 35.6% yield) as oil. Exact mass calculated for Ci4H2oF3N303: 335.1, found LC/MS mlz = 336.4 (M+H+).
Step 2: Preparation of 3-((lr,4r)-4-(l-(5-(Trifluoromethyl)-l,2,4-oxadiazol-3- yl)piperidin-4-yloxy)cyclohexyloxy)pyrazine-2-carbonitrile (Compound 8).
To a solution of (lr,4r)-4-(l-(5-(trifluoromethyl)-l,2,4-oxadiazol-3-yl)piperidin-4- yloxy)cyclohexanol (0.18 g, 0.537 mmol) and 3-chloropyrazine-2-carbonitrile (89.89 mg, 0.644 mmol) in THF (3 mL) was added 1 M solution of potassium i-butoxide in THF (0.644 mL, 0.644 mmol) at ice-water bath temperature under N2. After stirring for 10 min, crude LC/MS showed disappearance of starting material and presence of some amount of product, and decomposed byproducts. Water was added into the reaction, extracted with ethyl acetate. The combined organics were concentrated. The residue was purified by semi preparative HPLC (30- 95% CH3CN/H20 with 0.1% TFA over 30 min). The combined fractions were neutralized with sat. NaHC03, extracted with DCM, then concentrated to afford the title compound (23 mg, 52.46 μηιοΐ, 9.8% yield). Exact mass calculated for Ci9H21F3N603: 438.2, found LC/MS mlz = 439.4 (M+H+); ¾ NMR (400 MHz, CDC13) δ ppm 1.50-1.60 (m, 2H), 1.62-1.75 (m, 4H), 1.85- 1.95 (m, 2H), 1.98-2.08 (m, 2H), 2.10-2.18 (m, 2H), 3.30-3.36 (m, 2H), 3.56-3.63 (m, 1H), 3.63-3.70 (m, 1H), 3.73-3.82 (m, 2H), 5.18-5.26 (m, 1H), 8.23 (d, / = 2.5 Hz, 1H), 8.29 (d, / = 2.5 Hz, 1H).
Example 1.9: Preparation of l-(Trifluoromethyl)cyclobutyl 4-((lr,4r)-4-(3-Cyanopyrazin- 2-yloxy)cyclohexyloxy)piperidine-l-carboxylate (Compound 9).
Step 1: Preparation of l-(Trifluoromethyl)cyclobutyl l//-Imidazole-l-carboxylate.
To a solution of cyclobutanone (1.27 mL, 17.0 mmol) and
trimethyl(trifluoromethyl)silane (3.18 mL, 20.4 mmol) in THF (5 mL) was slowly added 1 M tetrabutylammonium fluoride solution in THF (0.17 mL, 0.17 mmol). The mixture was stirred at rt for 2 h and then cooled to 0 °C. The mixture was quenched with 6 N HC1 (aq) (25 mL) and then stirred at rt for 2 h. The mixture was extracted with ether (2 x 50 mL) and the combined organic layers were washed with brine, dried over anhydrous Na2S04, filtered, and concentrated under reduced pressure (350 mmHg at rt water bath) to afford a crude brown solution of 1- (trifluoromethyl)cyclobutanol in THF/ether. To this solution was added 1, 1 '- carbonyldiimidazole (CDI; 2.75 g, 17.0 mmol) and the mixture was heated at 70 °C for 2 h. Additional CDI (1.10 g, 6.78 mmol, 0.4 eq) was added and the reaction was heated at 70 °C for an additional 2 h. The reaction was quenched with water and brine. The mixture was extracted with DCM and the organic layer was dried under reduced pressure. The residue was purified by Biotage® column chromatography to afford the title compound (2.49 g, 10.6 mmol, 62.6%) as an off-white solid. Exact mass calculated for C9H9F3N2O2: 234.1, found: LC/MS m/z = 235.0 (M+H+); ¾ NMR (400 MHz, CDC13) δ ppm 1.89-2.04 (m, 1H), 2.04-2.19 (m, 1H), 2.64-2.78 (m, 2H), 2.84-2.97 (m, 2H), 7.09 (s, 1H), 7.40 (t, 7 = 1.39 Hz, 1H), 8.1 1 (s, 1H).
Step 2: Preparation of l-(Trifluoromethyl)cyclobutyl 4-((lr,4r)-4-(3-Cyanopyrazin- 2-yloxy)cyclohexyloxy)piperidine-l-carboxylate (Compound 9).
To a suspension of 3-((lr,4r)-4-(piperidin-4-yloxy)cyclohexyloxy)pyrazine-2- carbonitrile hydrochloride (100.0 mg, 0.2950 mmol) and l-(trifluoromethyl)cyclobutyl IH- imidazole-l-carboxylate (76.0 mg, 0.325 mmol) in THF (8 mL) under nitrogen in a sealed tube was added triethylamine (0.21 mL, 1.5 mmol). The mixture was heated at 150 °C for 1 h under microwave irradiation. About 24% of starting material still remained and thus more 1- (trifluoromethyl)cyclobutyl 1/i-imidazole-l-carboxylate (27.0 mg, 0.115 mmol, 0.39 eq) was added and the mixture was heated at 150 °C for an addition 30 min under microwave irradiation. The reaction was complete and the mixture was concentrated under reduced pressure. The residue was purified by Biotage® column chromatography to afford the title compound (1 13.6 mg, 0.242 mmol, 82.2%) an off-white gum. Exact mass calculated for C22H27F3N4O4: 468.2, found: LC/MS m/z = 469.2 (M+H+); ¾ NMR (400 MHz, CDC13) δ ppm 1.47-1.62 (m, 4H), 1.63-1.73 (m, 2H), 1.74-1.91 (m, 3H), 1.92-2.06 (m, 3H), 2.07-2.20 (m, 2H), 2.44-2.62 (m, 2H), 2.75-2.90 (m, 2H), 3.17-3.28 (m, 2H), 3.50-3.66 (m, 2H), 3.74 (bs, 2H), 5.15-5.28 (m, 1H), 8.23 (d, 7 = 2.53 Hz, 1H), 8.29 (d, 7 = 2.53 Hz, 1H).
Example 1.10: Preparation of 3-(Trifluoromethyl)oxetan-3-yl 4-((lr,4r)-4-(3- Cyanopyrazin-2-yloxy)cyclohexyloxy)piperidine- 1-carboxylate (Compound 10).
Step 1: Preparation of Perfluorophenyl 3-(Trifluoromethyl)oxetan-3-yl carbonate.
To a solution of oxetan-3-one (0.980 g, 13.6 mmol) and trimethyl(trifluoromethyl)silane (3.50 mL, 22.4 mmol) in THF (10 mL) was slowly added 1 M tetrabutylammonium fluoride solution in THF (1.36 mL, 1.36 mmol). The mixture was stirred at rt for 2 h and then cooled to 0 °C. The mixture was quenched with 6 N HC1 (aq) (29 mL) and then stirred at rt for 2 h. The mixture was extracted with ether (2 x 50 mL) and the combined organic layers were washed with brine, dried over anhydrous Na2S04, filtered, and concentrated under reduced pressure (350 mmHg at rt water bath) to afford a crude brown solution of 3-(trifluoromethyl)oxetan-3-ol in THF/ether. ¾ NMR (400 MHz, CDC13) δ ppm 4.62 (d, 7 = 8.34 Hz, 2H), 4.80 (d, 7 = 7.33 Hz, 2H). To this solution was added bis(perfluorophenyl) carbonate (6.432 g, 16.32 mmol) and MeCN (6 mL). To this at 0 °C was added triethylamine (6.10 mL, 43.5 mmol) dropwise. The mixture was stirred at rt for 23 h. The mixture was concentrated under reduced pressure and the residue was purified by Biotage® column chromatography to afford the title compound (2.3514 g, 6.678 mmol, 49.1%) as yellow oil. ¾ NMR (400 MHz, CDC13) δ ppm 4.91 (d, J = 9.85 Hz, 2H), 5.04 (d, 7 = 8.59 Hz, 2H).
Step 2: Preparation of 3-(Trifluoromethyl)oxetan-3-yl 4-((lr,4r)-4-(3- Cyanopyrazin-2-yloxy)cyclohexyloxy)piperidine- 1-carboxylate (Compound 10).
To a suspension of 3-((lr,4r)-4-(piperidin-4-yloxy)cyclohexyloxy)pyrazine-2- carbonitrile hydrochloride (120 mg, 0.354 mmol) and perfluorophenyl 3- (trifluoromethyl)oxetan-3-yl carbonate (0.137 g, 0.390 mmol) in DCM (5 mL) under nitrogen was added triethylamine (0.25 mL, 1.8 mmol). The mixture was stirred at rt for 2 h. The mixture was quenched with saturated NaHC03 (aq) and the organic layer was separated. The aqueous layer was extracted with DCM (IX) and the combined organics were concentrated under reduced pressure. The residue was purified by Biotage® column chromatography to afford the title compound (157.8 mg, 0.335 mmol, 94.7%) as a white solid. Exact mass calculated for C21H25F3N405: 470.2, found: LC/MS m/z = 471.2 (M+H+); ¾ NMR (400 MHz, CDC13) δ ppm 1.47-1.64 (m, 4H), 1.65-1.76 (m, 2H), 1.82 (bs, 2H), 1.95-2.07 (m, 2H), 2.07-2.19 (m, 2H), 3.31 (bs, 2H), 3.52-3.61 (m, 1H), 3.61-3.68 (m, 1H), 3.70-3.78 (m, 2H), 4.84 (d, 7 = 9.35 Hz, 2H), 4.95-5.03 (m, 2H), 5.17-5.28 (m, 1H), 8.23 (d, J = 2.53 Hz, 1H), 8.29 (d, J = 2.53 Hz, 1H).
Example 1.11: Preparation of l,l,l-Trifluoro-2-methylpropan-2-yl 4-((lr,4r)-4-(3- Cyanopyrazin-2-yloxy)cyclohexyloxy)piperidine- 1-carboxylate (Compound 11).
Step 1: Preparation of l,l,l-Trifluoro-2-methylpropan-2-yl l//-Imidazole-l- carboxylate.
A solution of l, l, l-trifluoro-2-methylpropan-2-ol (500.0 mg, 3.903 mmol) and 1,1 '- carbonyldiimidazole (1.27 g, 7.81 mmol) in THF (20 mL) under nitrogen was stirred at 60 °C for 16 h (14% conversion to product by NMR). The reaction was heated at 60 °C for an additional 94 h but was not complete (83% conversion to product). The reaction was stopped. The reaction mixture was concentrated under reduced pressure and the residue was purified by Biotage® column chromatography to afford the title compound (703.7 mg, 3.167 mmol, 81.1%) as a white solid. Exact mass calculated for ΰ8Η9Ε3Ν202: 222.1, found: LC/MS m/z = 223.2 (M+H+); ¾ NMR (500 MHz, CDC13) δ ppm 1.84 (s, 6H), 7.26 (s, 1H), 7.37 (s, 1H), 8.09 (s, 1H).
Step 2: Preparation of l,l,l-Trifluoro-2-methylpropan-2-yl 4-((lr,4r)-4-(3- Cyanopyrazin-2-yloxy)cyclohexyloxy)piperidine- 1-carboxylate (Compound 11). To a suspension of 3-((lr,4r)-4-(piperidin-4-yloxy)cyclohexyloxy)pyrazine-2- carbonitrile hydrochloride (100.0 mg, 0.2950 mmol) and l, l, l-trifluoro-2-methylpropan-2-yl lH-imidazole-l-carboxylate (72.1 mg, 0.325 mmol) in THF (8 mL) was added triethylamine (0.21 mL, 1.5 mmol). The mixture was heated at 90 °C for 5.5 h. The reaction was not complete. To this mixture was added more l, l, l-trifluoro-2-methylpropan-2-yl 1/i-imidazole- l- carboxylate (27.0 mg, 0.122 mmol, 0.4 eq) and the mixture was heated at 150 °C for 1 h under microwave irradiation. The reaction was almost finished and thus stopped. The mixture was directly concentrated under reduced pressure and the residue was purified by Biotage® column chromatography and prep HPLC (2nd purification). Pure fractions were combined, neutralized with saturated NaHC03 (aq), and concentrated. The solid was collected by vacuum filtration to afford the title compound (67.4 mg, 0.148 mmol, 50.0%) as a white solid. Exact mass calculated for C2iH27F3N404: 456.2, found: LC/MS m/z = 457.4 (M+H+); ¾ NMR (400 MHz, CDC13) δ ppm 1.46-1.61 (m, 4H), 1.63-1.74 (m, 2H), 1.68 (s, 6H), 1.81 (bs, 2H), 1.95-2.07 (m, 2H), 2.07- 2.19 (m, 2H), 3.13-3.26 (m, 2H), 3.50-3.65 (m, 2H), 3.72 (bs, 2H), 5.13-5.29 (m, 1H), 8.23 (d, 7 = 2.53 Hz, 1H), 8.29 (d, 7 = 2.53 Hz, 1H).
Example 1.12: Preparation of l,l,l,3,3,3-Hexafluoropropan-2-yl 4-((lr,4r)-4-(3- Cyanopyrazin-2-yloxy)cyclohexyloxy)piperidine- 1-carboxylate (Compound 12).
A solution of l, l, l,3,3,3-hexafluoropropan-2-ol (92.0 μΐ, 0.885 mmol) in THF (5 mL) under nitrogen was added Ι, Γ -carbonyldiimidazole (71.8 mg, 0.443 mmol) and the mixture was stirred at rt for 2 h. To this was added 3-((lr,4r)-4-(piperidin-4-yloxy)cyclohexyloxy)pyrazine- 2-carbonitrile hydrochloride (100.0 mg, 0.295 mmol) and triethylamine (0.21 mL, 1.5 mmol). The mixture was heated at 150 °C for 1 h under microwave irradiation. The reaction was complete but formed a side product due to unreacted CDI. The mixture was directly concentrated under reduced pressure and the residue was purified by prep HPLC. Pure fractions were combined, neutralized with saturated NaHC03 (aq), and concentrated under reduced pressure. The aqueous residue was extracted with DCM (twice) and the combined organics were dried over anhydrous Na2S04, filtered, and concentrated under reduced pressure to afford the title compound (77.0 mg, 0.155 mmol, 52.6%) as a white solid. Exact mass calculated for C2oH22F6N404: 496.2, found: LC/MS m/z = 497.2 (M+H+); ¾ NMR (400 MHz, CDC13) δ ppm 1.48-1.76 (m, 6H), 1.77-1.89 (m, 2H), 1.96-2.06 (m, 2H), 2.08-2.18 (m, 2H), 3.35-3.45 (m, 2H), 3.54-3.61 (m, 1H), 3.63-3.70 (m, 1H), 3.71-3.81 (m, 2H), 5.14-5.31 (m, 1H), 5.75 (quin, 7 = 6.32 Hz, 1H), 8.23 (d, 7 = 2.53 Hz, 1H), 8.29 (d, 7 = 2.53 Hz, 1H).
Example 1.13: Preparation of l-(Trifluoromethyl)cyclopentyl 4-((lr,4r)-4-(3- Cyanopyrazin-2-yloxy)cyclohexyloxy)piperidine- 1-carboxylate (Compound 13). Step 1: Preparation of l-(Trifluoromethyl)cyclopentyl l//-Imidazole-l- carboxylate.
To a solution of cyclopentanone (1.0 mL, 11 mmol) and
trimethyl(trifluoromethyl)silane (2.12 mL, 13.6 mmol) in THF (10 mL) under nitrogen was slowly added 1 M tetrabutyl ammonium fluoride in THF (1.13 mL, 1.13 mmol) (exothermic). The mixture was stirred at rt for 2.5 h and then cooled to 0 °C. The mixture was quenched with 6 N HC1 (aq) (25 mL) and then stirred at rt for 1.5 h. The mixture was extracted with ether (2 x 50 mL) and the combined organic layers were washed with brine, dried over anhydrous Na2S04, filtered, and concentrated under reduced pressure (350 mmtig at rt water bath) to afford the crude brown solution of l-(trifluoromethyl)cyclopentanol in THF/ether. To this solution was added Ι, Γ -carbonyldiimidazole (2.75 g, 17.0 mmol) and THF (10 mL) under nitrogen and the mixture was heated at 70 °C overnight. The mixture was concentrated under reduced pressure and the residue was purified by Biotage® column chromatography to afford the title compound (2.2306 g, 8.987 mmol, 79.5%) as a light yellow oil. Exact mass calculated for C10H11F3N2O2: 248.1, found: LC/MS m/z = 249.2 (M+H+); ¾ NMR (400 MHz, CDC13) δ ppm 1.72-1.88 (m, 2H), 2.02-2.17 (m, 2H), 2.21-2.33 (m, 2H), 2.36-2.53 (m, 2H), 7.08 (s, 1H), 7.32-7.41 (m, 1H), 8.09 (s, 1H).
Step 2: Preparation of l-(Trifluoromethyl)cyclopentyl 4-((lr,4r)-4-(3- Cyanopyrazin-2-yloxy)cyclohexyloxy)piperidine- 1-carboxylate (Compound 13).
To a suspension of 3-((lr,4r)-4-(piperidin-4-yloxy)cyclohexyloxy)pyrazine-2- carbonitrile hydrochloride (100.0 mg, 0.2950 mmol) and l-(trifluoromethyl)cyclopentyl IH- imidazole- 1-carboxylate (87.9 mg, 0.354 mmol) in THF (8 mL) under nitrogen was added triefhylamine (0.21 mL, 1.5 mmol). The mixture was heated at 150 °C for 1 h under microwave irradiation. The mixture was directly concentrated under reduced pressure and the residue was purified by prep HPLC. Pure fractions were combined, neutralized with saturated NaHC03 (aq), and concentrated. The aqueous residue was extracted with DCM (twice) and the combined organics were dried over anhydrous Na2S04, filtered, and concentrated under reduced pressure to afford the title compound (125.1 mg, 0.259 mmol, 87.9%) as a white solid. Exact mass calculated for C23¥i29F3N404: 482.2, found: LC/MS m/z = 483.2 (M+H+); :H NMR (400 MHz, CDCI3) δ ppm 1.47-1.61 (m, 4H), 1.61-1.74 (m, 4H), 1.81 (bs, 2H), 1.94-2.18 (m, 8H), 2.25- 2.41 (m, 2H), 3.14-3.28 (m, 2H), 3.52-3.64 (m, 2H), 3.73 (bs, 2H), 5.15-5.29 (m, 1H), 8.23 (d, 7 = 2.53 Hz, 1H), 8.29 (d, 7 = 2.53 Hz, 1H).
Example 1.14: Preparation of 3-((lr,4r)-4-(l-(5-(2-Fluoropropan-2-yl)-l,2,4-oxadiazol-3- yl)piperidin-4-yloxy)cyclohexyloxy)pyrazine-2-carbonitrile (Compound 14).
Step 1: Preparation of N'-Hydroxy-4-((lr,4r)-4-hydroxycyclohexyloxy)piperidine- 1-carboximidamide. Hydroxylamine (0.52 mL, 7.82 mmol) was added to a solution of 4-((lr,4r)-4- hydroxycyclohexyloxy)piperidine- l-carbonitrile (1.25 g, 5.58 mmol) in EtOH (33.03 mL) and the reaction was warmed to 45 °C and stirred for 16 h. Toluene was added and the reaction was concentrated. LC/MS m/z = 258.2 (M+H+).
Step 2: Preparation of (lr,4r)-4-(l-(5-(2-Fluoropropan-2-yl)-l,2,4-oxadiazol-3- yl)piperidin-4-yloxy)cyclohexanol.
To a solution of 2-fluoro-2-methylpropanoic acid (0.71 g, 6.70 mmol) in DMA (6.6 mL) at rt was added Ι, Γ -carbonyldiimidazole (0.91 g, 5.58 mmol). The reaction was stirred for 1.5 h and and the resulting mixture was added dropwise to a solution of Ar-hydroxy-4-((lr,4r)-4- hydroxycyclohexyloxy)piperidine- l-carboximidamide (1.44 g, 5.58 mmol) in DMA (27.0 mL) at 0 °C. The reaction was warmed to 60 °C and stirred for 1 h. An additional 0.5 equivalents of 2-fluoro-2-methylpropanoic acid and Ι, Γ -carbonyldiimidazole were briefly stirred in DMA and added to the reaction mixture at rt. The mixture was warmed to 60 °C and stirred for 1 h. The reaction was cooled to room temperature and concentrated to dryness. The crude product was purified by column chromatography (silica gel, 0-5-10% MeOH in DCM) to afford 1.43 g of the title compound. LC/MS m/z = 328.2 (M+H+).
Step 3: Preparation of 3-((lr,4r)-4-(l-(5-(2-Fluoropropan-2-yl)-l,2,4-oxadiazol-3- yl)piperidin-4-yloxy)cyclohexyloxy)pyrazine-2-carbonitrile (Compound 14).
Potassium 2-methylpropan-2-olate (0.73 mL, 0.73 mmol) was added to a solution of ( 1 r,4r)-4-( 1 -(5-(2-fluoropropan-2-yl)- 1 ,2,4-oxadiazol-3-yl)piperidin-4-yloxy)cyclohexanol (0.20 g, 0.61 mmol) in THF (0.47 mL) at 0 °C. The reaction mixture was stirred for 10 min and a solution of 3-chloropyrazine-2-carbonitrile (93.8 mg, 0.67 mmol) in THF (0.5 mL) was added. The reaction was warmed to room temperature and stirred for 1 h. The reaction was quenched with water and EtOAc and 1.5 M aqueous HC1 were added. The EtOAc layer was removed and the aqueous mixture was extracted once with EtOAc. The combined EtOAc solution was dried (sodium sulfate), filtered, and concentrated. The resulting residue was purified by column chromatography to afford the title compound (191 mg). LC/MS m/z = 431.4 (M+H+). ¾ NMR (400 MHz, CDC13) δ ppm 1.50-1.74 (m, 6H), 1.78 (d, / = 21 Hz, 6H), 1.85- 1.94 (m, 2H), 1.98- 2.07 (m, 2H), 2.09-2.18 (m, 2H), 3.19-3.28 (m, 2H), 3.56-3.67 (m, 2H), 3.75-3.83 (m, 2H), 5.19-5.26 (m, 1H), 8.23 (d, / = 2.6 Hz, 1H), 8.29 (d, / = 2.6 Hz, 1H).
Example 1.15: Preparation of 3-((lr,4r)-4-(l-(3-(2-Fluoropropan-2-yl)-l,2,4-oxadiazol-5- yl)piperidin-4-yloxy)cyclohexyloxy)pyrazine-2-carbonitrile (Compound 15) .
Step 1: Preparation of (lr,4r)-4-(l-(3-(2-Fluoropropan-2-yl)-l,2,4-oxadiazol-5- yl)piperidin-4-yloxy)cyclohexanol.
To a solution of 4-((lr,4r)-4-hydroxycyclohexyloxy)piperidine- l-carbonitrile (0.2 g, 0.892 mmol) and (Z)-2-fluoro-Ar -hydroxy-2-methylpropanimidamide (0.193 g, 1.605 mmol) in THF (4 mL) was added 0.5 M zinc(II) chloride solution in THF (3.567 mL, 1.783 mmol). The mixture was stirred at rt for 3.5 h. To this solution was added 4 N hydrogen chloride solution in dioxane (1.337 mL, 5.350 mmol). The mixture was heated at 60 °C overnight. The reaction was not complete and thus the mixture was heated at 120 °C for 2 h under microwave irradiation. The mixture was quenched with saturated NaHC03 (aq) and the organic layer was separated. The aqueous layer was extracted with DCM (twice) and the combined organics were concentrated under reduced pressure and purified by Biotage® column chromatography to afford the title compound (0.1637 g, 0.500 mmol, 56.1%) as a light yellow oil. Exact mass calculated for Ci6H26FN303: 327.2, found: LC/MS m/z = 328.2 (M+H+); ¾ NMR (400 MHz, CDCl3) 5 ppm 1.29-1.41 (m, 4H), 1.62-1.72 (m, 2H), 1.72 (d, J = 24 Hz, 6H), 1.82- 1.91 (m, 2H), 1.92-2.02 (m, 4H), 3.35-3.52 (m, 3H), 3.62-3.75 (m, 2H), 3.79-3.89 (m, 2H).
Step 2: Preparation of 3-((lr,4r)-4-(l-(3-(2-Fluoropropan-2-yl)-l,2,4-oxadiazol-5- yl)piperidin-4-yloxy)cyclohexyloxy)pyrazine-2-carbonitrile (Compound 15).
To a solution of (lr,4r)-4-(l-(3-(2-fluoropropan-2-yl)- l,2,4-oxadiazol-5-yl)piperidin-4- yloxy)cyclohexanol (154 mg, 0.470 mmol) and 3-chloropyrazine-2-carbonitrile (83.8 mg, 0.601 mmol) in 2 mL THF, 1 M potassium ieri-butoxide in THF (0.6 mL, 0.600 mmol) was added slowly. After stirring at rt for 0.5 h, the dark brown solution was extracted with water and CH2C12. Organic phases were dried over MgS04, filtered, and concentrated. Residue was purified by Biotage® column chromatography (Si02, hexane/AcOEt gradient). Fractions containing product were concentrated and re-purified by HPLC (CH3CN/H20 gradient + 0.1% TFA). Fractions containing product were partly concentrated and residue was extracted with 1 M NaHC03 and CH2C12. Organic phases were dried over MgS04, filtered, and concentrated to afford the title compound (125 mg, 0.290 mmol, 61.7%) as a white solid. Exact mass calculated for C2iH27FN603: 430.21, found: LC/MS m/z = 431.2 (M+H+); ¾ NMR (400 MHz, CDC13) δ ppm 1.51-1.60 (m, 2H), 1.66-1.76 (m, 10H), 1.86- 1.93 (m, 2H), 1.99-2.06 (m, 2H), 2.10-2.17 (m, 2H), 3.47-3.53 (m, 2H), 3.56-3.62 (m, IH), 3.66-3.72 (m, IH), 3.82-3.89 (m, 2H), 5.20-5.27 (m, IH), 8.23 (d, J = 2.5 Hz, IH), 8.30 (d, J = 2.5 Hz, IH).
Example 1.16: Preparation of 3-((lr,4r)-4-(l-(3-(2-Fluoropropan-2-yl)-l,2,4-oxadiazol-5- yl)piperidin-4-yloxy)cyclohexyloxy)picolinonitrile (Compound 16).
To a solution of (lr,4r)-4-(l-(3-(2-fluoropropan-2-yl)- l,2,4-oxadiazol-5-yl)piperidin-4- yloxy)cyclohexanol (177 mg, 0.541 mmol) in 2 mL THF, 1 M potassium 2-methylpropan-2- olate in THF (0.6 mL, 0.600 mmol) was added. After stirring at rt for 10 min, 3- chloropicolinonitrile (150 mg, 1.083 mmol) was added. After stirring for 30 min, the mixture was extracted with brine and EtOAc. Organic phases were concentrated and purified by HPLC (H20/CH3CN gradient + 0.1% TFA). Fractions containing product were partly concentrated and residue was extracted with 1 M NaHC03 and CH2C12. Organic phases were dried over MgS04, filtered, and concentrated to afford the title compound (80.5 mg, 0.187 mmol, 34.7%) as a thick oil that solidified upon storage. Exact mass calculated for C22H28FN5O3: 429.22, found: LC/MS m/z = 430.2 (M+H+); ¾ NMR (400 MHz, CDC13) δ ppm 1.51- 1.58 (m, 2H), 1.65-1.78 (m, 10H), 1.85-1.92 (m, 2H), 1.99-2.13 (m, 4H), 3.48-3.54 (m, 2H), 3.61-3.71 (m, 2H), 3.82-3.88 (m, 2H), 4.50-4.55 (m, IH), 7.33-7.35 (m, IH), 7.42-7.45 (m, IH), 8.27-8.28 (m, IH).
Example 1.17: Preparation of 3-((lr,4r)-4-(l-(5-(2-Fluoropropan-2-yl)-l,2,4-oxadiazol-3- yl)piperidin-4-yloxy)cyclohexyloxy)picolinonitrile (Compound 17).
To a solution of (lr,4r)-4-(l-(5-(2-fluoropropan-2-yl)- l,2,4-oxadiazol-3-yl)piperidin-4- yloxy)cyclohexanol (109 mg, 0.333 mmol) in 1.5 mL THF, potassium 2-methylpropan-2-olate (0.4 mL, 0.400 mmol) was added. After stirring at rt for 10 min, 3-chloropicolinonitrile (94 mg, 0.678 mmol) was added. After stirring at rt for 15 min, the mixture was extracted with AcOEt and brine. Organic phases were concentrated and purified by HPLC (CH3CN/H20 gradient + 0.1% TFA). Fractions containing product were partly concentrated and residue was extracted with 1 M NaHC03 and CH2C12. Organic phases were dried over MgS04, filtered, and concentrated. Residue was re-purified by Biotage® column chromatography (hexane/ AcOEt gradient) to afford the title compound (66 mg, 0.154 mmol, 46.2%) a colorless thick oil. Exact mass calculated for C22H28FN503: 429.22, found: LC/MS m/z = 430.2 (M+H+); ¾ NMR (400 MHz, CDCI3) δ ppm 1.50-1.77 (m, 9H), 1.81 (s, 3H), 1.86-1.93 (m, 2H), 1.99-2.14 (m, 4H), 3.21-3.27 (m, 2H), 3.60-3.67 (m, 2H), 3.75-3.81 (m, 2H), 3.48-3.54 (m, IH), 7.33-7.35 (m, IH), 7.42-7.45 (m, IH), 8.26-8.27 (m, IH).
Example 1.18: Preparation of 3-((lr,4r)-4-(l-(5-Chloropyrimidin-2-yl)piperidin-4- yloxy)cyclohexyloxy)picolinonitrile (Compound 18).
A mixture of 3-((lr,4r)-4-(piperidin-4-yloxy)cyclohexyloxy)picolinonitrile dihydrochloride (100 mg, 0.267 mmol, prepared in a manner similar to that described in
Example 1.2, Steps 3 and 4), 2,5-dichloropyrimidine (79.60 mg, 0.534 mmol) and triethylamine (0.112 mL, 0.801 mmol) in IPA (2 mL) was heated at 120 °C for 3 h under microwave irradiation. The white precipitate was collected, washed with IPA, dried to afford the title compound (80 mg, 0.193 mmol, 72.3% yield). Exact mass calculated for C2iH24ClN502: 413.2, found LC/MS m/z = 414.4 (M+H+); :H NMR (400 MHz, CDC13) δ ppm 1.48-1.62 (m, 4H), 1.68- 1.77 (m, 2H), 1.84-1.92 (m, 2H), 2.00-2.15 (m, 4H), 3.39-3.46 (m, 2H), 3.63-3.70 (m, 2H), 4.18-4.25 (m, 2H), 4.47-4.53 (m, IH), 7.34 (dd, / = 8.6 and 1.1 Hz, IH), 7.44 (dd, / = 8.8 and 4.5 Hz, IH), 8.21 (s, 2H), 8.27 (dd, / = 4.5 and 1.2 Hz, IH).
Example 1.19: Preparation of (/f)-l,l,l-trifluoropropan-2-yl 4-((lr,4r)-4-(2-cyanopyridin- 3-yloxy)cyclohexyloxy)piperidine-l-carboxylate (Compound 19). (R)-l, l, l-Trifluoropropan-2-ol (99.96 mg, 0.876 mmol) and Ι, -carbonyldiimidazole (0.139 g, 0.855 mmol) in THF (3 mL) was stirred for 1 h at rt, then 3-((lr,4r)-4-(piperidin-4- yloxy)cyclohexyloxy)picolinonitrile dihydrochloride (80 mg, 0.214 mmol) was added, followed by triethylamine (0.179 mL, 1.282 mmol). The reaction mixture was stirred at reflux overnight. Water was added and extracted with ethyl acetate. The combined organics were concentrated. The crude was purified by silica gel chromatography with 40% ethyl acetate/hexanes to afford the title compound (85 mg, 0.193 mmol, 90.1 % yield) as oil. Exact mass calculated for C2iH26F3N304: 441.2, found LC/MS m/z = 442.4 (M+H+); ¾ NMR (400 MHz, CDC13) δ ppm 1.40 (d, 7 = 6.6 Hz, 3H), 1.48-1.58 (m, 4H), 1.66-1.86 (m, 4H), 1.98-2.12 (m, 4H), 3.20-3.32 (m, 2H), 3.58-3.65 (m, 2H), 3.70-3.80 (m, 2H), 4.48-4.54 (m, IH), 5.20-5.28 (m, IH), 7.35 (dd, 7 = 8.7 and 1.1 Hz, IH), 7.44 (dd, 7 = 8.7 and 4.5 Hz, IH), 8.27 (dd, 7 = 4.5 and 1.2 Hz, IH).
Example 1.20: Preparation of (5)-l,l,l-Trifluoropropan-2-yl 4-((lr,4r)-4-(2-Cyanopyridin- 3-yloxy)cyclohexyloxy)piperidine-l-carboxylate (Compound 20).
(S)-l, l, l-Trifluoropropan-2-ol (0.127 g, 0.876 mmol) and Ι,Γ -carbonyldiimidazole (0.139 g, 0.855 mmol) in THF (3 mL) was stirred for 1 h at rt, then 3-((lr,4r)-4-(piperidin-4- yloxy)cyclohexyloxy)picolinonitrile dihydrochloride (80 mg, 0.214 mmol) was added, followed by triethylamine (0.179 mL, 1.282 mmol). The reaction mixture was stirred at reflux overnight, water was added and extracted with ethyl acetate. The combined organics were concentrated. The crude was purified by silica gel chromatography with 40% ethyl acetate/hexanes to afford the title compound (82 mg, 0.186 mmol, 86.9% yield) as oil. Exact mass calculated for C2iH26F3N304: 441.2, found LC/MS m/z = 442.4 (M+H+); ¾ NMR (400 MHz, CDC13) δ ppm 1.40 (d, 7 = 6.6 Hz, 3H), 1.48-1.58 (m, 4H), 1.66-1.86 (m, 4H), 1.98-2.12 (m, 4H), 3.20-3.32 (m, 2H), 3.58-3.65 (m, 2H), 3.70-3.80 (m, 2H), 4.48-4.54 (m, IH), 5.20-5.28 (m, IH), 7.35 (dd, 7 = 8.7 and 1.1 Hz, IH), 7.44 (dd, 7 = 8.7 and 4.5 Hz, IH), 8.27 (dd, 7 = 4.5 and 1.2 Hz, IH).
Example 1.21: Preparation of 3-((lr,4r)-4-(l-(3-Isopropyl-l,2,4-oxadiazol-5-yl)piperidin-4- yloxy)cyclohexyloxy)picolinonitrile (Compound 21).
Potassium 2-methylpropan-2-olate (17.4 mg, 0.155 mmol) was added to a solution of (lr,4r)-4-(l-(5-isopropyl- l,2,4-oxadiazol-3-yl)piperidin-4-yloxy)cyclohexanol (0.02 g, 64.6 μηιοΐ) and 3-chloropicolinonitrile (8.9 mg, 64.6 μηιοΐ) in THF (1.0 mL) at room temperature. The reaction was stirred for 2h, then diluted with water and extracted with EtOAc. The extracts were dried (sodium sulfate), filtered, and concentrated. The crude product was purified by column chromatography (silica gel, 25-50-75% EtOAc in hexanes) and then thin layer chromatography (silica gel, 75% EtOAc in hexanes) to afford the title compound (18 mg). LC/MS m/z = 412.6 (M+H+). ¾ NMR (400 MHz, CDC13) δ ppm 1.28 (d, 7 = 7.1 Hz, 6H), 1.48- 1.59 (m, 2H), 1.62-1.78 (m, 4H), 1.84-1.93 (m, 2H), 1.98-2.14 (m, 4H), 2.88 (septet, 7 = 7.0 Hz, 1H), 3.41-3.49 (m, 2H), 3.60-3.70 (m, 2H), 3.79-3.87 (m, 2H), 4.48-4.55 (m, 1H), 7.34 (dd, / = 8.6, 1.3 Hz, 1H), 7.44 (dd, J = 8.6, 4.5 Hz, 1H), 8.27 (dd, J = 4.5, 1.2 Hz, 1H).
Example 1.22: Preparation of 1-Methylcyclopropyl 4-((lr,4r)-4-(2-Cyanopyridin-3- yloxy)cyclohexyloxy)piperidine-l-carboxylate (Compound 22).
A soultion of 1-methylcyclopropyl 4-((lr,4r)-4-hydroxycyclohexyloxy)piperidine-l- carboxylate (0.12 g, 0.404 mmol) in THF (3 mL) was treated with potassium 2-methylpropan-2- olate (0.484 mL, 0.484 mmol) under N2. After stirring for 10 min at room temperature, the reaction was treated with 3-chloropicolinonitrile (67.09 mg, 0.484 mmol), and stirred for an additional 1 h at 30 °C. The reaction was quenched with 1 N HC1, diluted with EtOAc, washed with 1 N HC1 and brine, dried, and concentrated. The residue was purified by column chromatography to afford the title compopund (80 mg, 50%). Exact mass calculated for C22H29N3O4: 399.2, found: LC/MS mlz = 400.4 (M+H+); ¾ NMR (400 MHz, CDC13) δ ppm 0.59-0.64 (m, 2H), 0.83-0.89 (m, 2H), 1.43-1.56 (m, 4H), 1.54 (s, 3H), 1.66-1.82 (m, 4H), 1.96-2.13 (m, 4H), 3.07-3.18 (m, 2H), 3.52-3.63 (m, 2H), 3.64-3.83 (bs, 2H). 4.46-4.53 (m, 1H), 7.33 (dd, J = 8.8 and 1.0 Hz, 1H), 7.43 (dd, J = 8.6 and 4.6 Hz, 1H), 8.23 (dd, 7 = 4.6 and 1.2 Hz, 1H).
Example 2: In vivo Effects of GPR119 agonists on Incretin Hormone GIP release.
Male 129SVE mice (approximately 8-week old) are fasted for 18 h and randomly grouped (n = 6) to receive vehicle or a GPR119 agonist (3 or 30 mg kg body weight).
Compounds are delivered orally via a gavage needle (p.o., volume 4 mL kg), and after 45 min a blood sample is collected to determine plasma total GIP levels. The control group receive vehicle (PET: 80%PEG: 10%Ethanol: 10%Tween80™). Plasma GIP levels are determined using a Total GIP ELISA kit from Millipore. GPR119 agonists have been shown to increase GIP release compared to control in this assay; see for example, WO2011/127051, WO2012/040279, and WO2012/145361 ; the disclosure of each is herein incorporated by reference in its entirety.
Example 3: In vivo effects of a Representative Compound of the Present Invention on
Glucose Homeostasis (oral glucose tolerance test (oGTT)) in male 129SVE mice.
Male 129SVE mice (approximately 8-week old) were fasted for 18 h and randomly grouped (n = 6) to receive a GPR119 agonist (Compound 7 and Compound 10) at 3, or 30 mg/kg (mg/kg body weight). Each compound was delivered orally via a gavage needle (p.o., volume 4 mL/kg) 30 minutes prior to glucose bolus (3g/kg) (time = -30 min in Figure 1 and
Figure 2), with a separate group receiving vehicle (20% hydroxypropyl-beta-cyclodextrin) as control. At time 0 min. the glucose bolus was administered. Levels of blood glucose were assessed using a glucometer (One-Touch Ultra™, LifeScan) at time -30 minute (prior to compound administration), at 0 min (at time when glucose bolus was given), and at 20, 40, 60, 120 minutes post glucose bolus. Plasma glucose levels are shown in Table 2. The reduction in the glucose excursion area under the curve (AUC) from -30 to 120 min in GPRl 19 agonist- treated animals relative to vehicle control is shown in Figure 3. The glycemic suppression or percent inhibition of glucose is also shown in Table 3.
Table 2 (Plasma lucose levels n =6)
Figure imgf000104_0001
Table 3 Gl cemic Su ression in oGTT in Mice
Figure imgf000104_0002
Example 4: Homogeneous Time-Resolved Fluorescence (HTRF®) Assay For Direct cAMP Measurement.
GPRl 19 agonists were evaluated in an HTRF® cAMP detection assay according to the manufacturer's instructions (Cisbio, cAMP Dynamic 2 Assay Kit; #62AM4PEJ) using CHO-Kl cells stably expressing GPRl 19. Briefly, CHO-Kl cells were transduced with a lentiviral vector encoding the nucleotide sequence of GPRl 19 (NCBI mRNA and protein reference sequences: NM_178471.2 & NP_848566 (human) and NM_181770.1 & NP_861435.1 (rat), (GPRl 19 has also been referred to as Glucose-Dependent Insulinotropic Receptor (GDIR)). The N-terminus of the GPRl 19 nucleotide sequence was modified to replace the first methionine-coding codon with a nucleotide sequence coding for a standard, nine amino acid, hemagglutinin tag.
Following transduction, cells expressing the GPRl 19 receptor were isolated and a single clone was isolated following standard dilution-cloning procedures. On the day of the assay, cultured CHO-GPR119 cells were harvested, suspended in assay buffer and plated into 384-well assay plates (PerkinElmer Proxiplate #6008280) at a density of 2,000 cells per well. A cAMP standard curve was added to each plate. Test compounds were solubilized in DMSO, serially diluted in DMSO and then diluted in assay buffer before being added to the cells. Test compounds were evaluated in triplicate, using 10-point, 5-fold serial dilutions starting at 10 μΜ. The final DMSO concentration in the assay was 0.5%. Compounds and cells were incubated for 1 h at room temperature and then detection reagents were added to each well (cAMP-D2 in cell lysis buffer, followed by europium cryptate-labeled anti-cAMP antibody). Plates were then incubated at room temperature for 1 h prior to reading. Time-resolved fluorescence measurements were collected on PerkinElmer Envision™ or BMG Pherastar™ microplate readers. The compound Ar-(2-fluoro-4-(methylsulfonyl) phenyl)-6-(4-(3-isopropyl-l,2,4-oxadiazol-5-yl)piperidin-l-yl)- 5-nitropyrimidin-4-amine (WO2004/065380) was used as a positive control in each runset while assay buffer containing 0.5% DMSO was used as the negative control. The HTRF® assay was used to determine EC50 values for GPR119 agonists.
Certain representative compounds of the present invention and their corresponding EC50 values (hGPRl 19) are shown in Table B.
Table B
Figure imgf000105_0001
Each of the Compounds 1 to 22 as shown in Table A was observed to have an hGPRl 19 EC50 value ranging from about 33.1 nM to about 0.3 nM.
Certain representative compounds of the present invention and their corresponding values (rGPRl 19) are shown in Table C.
Table C
Figure imgf000105_0002
Each of the Compounds 1 to 22 as shown in Table A was observed to have an rGPRl 19 EC50 value ranging from about 155.0 nM to about 3.7 nM.
Those skilled in the art will recognize that various modifications, additions, and substitutions 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.
Citation of any reference throughout this application is not to be construed as an admission that such reference is prior art to the present application.

Claims

We claim:
A compound selected from compounds of Formula (I) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000106_0001
(I)
wherein:
X is N or CH; and
R1 is selected from: Ci-C6 alkoxycarbonyl, C3-C7 cycloalkoxycarbonyl, heterocyclyloxycarbonyl, and heteroaryl; each optionally substituted with one or more substituents selected from: Ci-C6 alkyl, Ci-C6 haloalkyl, and halogen; provided that R1 is a group other than 4-(trifluoromethyl)pyrimidin-2-yl.
2. The compound according to claim 1, wherein X is N. 3. The compound according to claim 1, wherein X is CH. 4. The compound according to any one of claims 1 to 3, wherein R1 is selected from: tert- butoxycarbonyl, isopropoxycarbonyl, cyclobutoxycarbonyl, cyclopentyloxycarbonyl, cyclopropoxycarbonyl, (pyrrolidin-3-yloxy)carbonyl, 1,2,4-oxadiazolyl, (oxetan-3- yloxy)carbonyl, and pyrimidin-2-yl; each optionally substituted with one or more substituents selected from: chloro, fluoro, 2-fluoropropan-2-yl, isopropyl, methyl, 2,2,2- trifluoroefhyl, and trifluoromethyl.
5. The compound according to any one of claims 1 to 3, wherein R1 is selected from: tert- butoxycarbonyl and isopropoxycarbonyl; each optionally substituted with one or more fluoro substituents.
6. The compound according to any one of claims 1 to 3, wherein R1 is selected from
cyclobutoxycarbonyl, cyclopentyloxycarbonyl, and cyclopropoxycarbonyl; each optionally substituted with one or more substituents selected from: methyl and trifluoromethyl. The compound according to any one of claims 1 to 3, wherein R1 is selected from: (pyrrolidin-3-yloxy)carbonyl and (oxetan-3-yloxy)carbonyl; each optionally substituted with one or more substituents selected from: 2,2,2-trifluoroethyl and trifluoromethyl.
The compound according to any one of claims 1 to 3, wherein R1 is selected from: 1,2,4-oxadiazolyl, and pyrimidin-2-yl; each optionally substituted with one or more substituents selected from: chloro, 2-fluoropropan-2-yl, isopropyl, and trifluoromethyl.
The compound according to any one of claims 1 to 3, wherein R1 is selected from: (1- methylcyclopropoxy)carbonyl, 5-chloropyrimidin-2-yl, (1,1, 1 -trifluoropropan-2- yloxy)carbonyl, ( 1 -(2,2,2-trifluoroethyl)pyrrolidin-3-yloxy)carbonyl, 3- (trifluoromethyl)-l,2,4-oxadiazol-5-yl, 5-(trifluoromethyl)-l,2,4-oxadiazol-3-yl, (1- (trifluoromethyl)cyclobutoxy)carbonyl, (3-(trifluoromethyl)oxetan-3-yloxy)carbonyl, (1, 1, 1 -trifluoro-2-methylpropan-2-yloxy)carbonyl, (1,1, 1 ,3,3,3-hexafluoropropan-2- yloxy)carbonyl, (l-(trifluoromethyl)cyclopentyloxy)carbonyl, 5-(2-fluoropropan-2-yl)- l,2,4-oxadiazol-3-yl, 3-(2-fluoropropan-2-yl)- l,2,4-oxadiazol-5-yl, and 3-isopropyl- 1 ,2,4-oxadiazol-5-yl.
The compound according to claim 1, selected from compounds of Formula (lb) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000107_0001
(lb)
wherein:
R1 is selected from: Ci-C6 alkoxycarbonyl, C3-C7 cycloalkoxycarbonyl, heterocyclyloxycarbonyl, and heteroaryl; each optionally substituted with one or more substituents selected from: chloro, fluoro, 2-fluoropropan-2-yl, methyl, 2,2,2- trifluoroethyl, and trifluoromethyl.
The compound according to claim 1, selected from compounds of Formula (lb) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000107_0002
(lb)
wherein: R1 is selected from: ieri-butoxycarbonyl, isopropoxycarbonyl,
cyclobutoxycarbonyl, cyclopentyloxycarbonyl, cyclopropoxycarbonyl, (pyrrolidin-3- yloxy)carbonyl, 1,2,4-oxadiazolyl, (oxetan-3-yloxy)carbonyl, and pyrimidin-2-yl; each optionally substituted with one or more substituents selected from: C1-C6 alkyl, C1-C6 haloalkyl, and halogen.
The compound according to claim 1, selected from compounds of Formula (lb) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000108_0001
(lb)
wherein:
R1 is selected from: (l-methylcyclopropoxy)carbonyl, 5-chloropyrimidin-2-yl, (1,1,1 -trifluoropropan-2-yloxy)carbonyl, ( 1 -(2,2,2-trifluoroethyl)pyrrolidin-3- yloxy)carbonyl, 3-(trifluoromethyl)-l,2,4-oxadiazol-5-yl, 5-(trifluoromethyl)- 1,2,4- oxadiazol-3-yl, ( 1 -(trifluoromethyl)cyclobutoxy)carbonyl, (3-(trifluoromethyl)oxetan-3 yloxy)carbonyl, (l,l,l-trifluoro-2-methylpropan-2-yloxy)carbonyl, (1,1,1,3,3,3- hexafluoropropan-2-yloxy)carbonyl, ( 1 -(trifluoromethyl)cyclopentyloxy)carbonyl, 5-(2 fluoropropan-2-yl)-l,2,4-oxadiazol-3-yl, and 3-(2-fluoropropan-2-yl)-l,2,4-oxadiazol-5 yi-
The compound according to claim 1, selected from compounds of Formula (Id) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000108_0002
(Id)
wherein:
R1 is selected from: Ci-C6 alkoxycarbonyl, C3-C7 cycloalkoxycarbonyl, and heteroaryl; each optionally substituted with one or more substituents selected from: chloro, fluoro, 2-fluoropropan-2-yl, isopropyl, and methyl.
14. The compound according to claim 1, selected from compounds of Formula (Id) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000109_0001
(Id)
wherein:
R1 is selected from: isopropoxycarbonyl, cyclopropoxycarbonyl, 1,2,4- oxadiazolyl, and pyrimidin-2-yl; each optionally substituted with one or more substituents selected from: Ci-C6 alkyl, Ci-C6 haloalkyl, and halogen.
The compound according to claim 1, selected from compounds of Formula (Id) and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Figure imgf000109_0002
(Id)
wherein:
R1 is selected from: (l-methylcyclopropoxy)carbonyl, 5-chloropyrimidin-2-yl, (l, l, l-trifluoropropan-2-yloxy)carbonyl, 5-(2-fluoropropan-2-yl)-l,2,4-oxadiazol-3-yl, 3-(2-fluoropropan-2-yl)-l,2,4-oxadiazol-5-yl, and 3-isopropyl-l,2,4-oxadiazol-5-yl.
16. A compound according to claim 1 selected from the following compounds and
pharmaceutically acceptable salts, solvates, and hydrates thereof:
1 -methylcyclopropyl 4-(( 1 r,4r)-4-(3-cyanopyrazin-2- yloxy)cyclohexyloxy)piperidine- 1 -carboxylate;
3-((lr,4r)-4-(l-(5-chloropyrimidin-2-yl)piperidin-4- yloxy)cyclohexyloxy)pyrazine-2-carbonitrile;
(R)- 1, 1, 1 -trifluoropropan-2-yl 4-(( 1 r,4r)-4-(3-cyanopyrazin-2- yloxy)cyclohexyloxy)piperidine- 1 -carboxylate;
(S)-l, l, l-trifluoropropan-2-yl 4-((lr,4r)-4-(3-cyanopyrazin-2- yloxy)cyclohexyloxy)piperidine- 1 -carboxylate;
(R)-l-(2,2,2-trifluoroethyl)pyrrolidin-3-yl 4-((lr,4r)-4-(3-cyanopyrazin-2- yloxy)cyclohexyloxy)piperidine- 1 -carboxylate;
(S)-l-(2,2,2-trifluoroethyl)pyrrolidin-3-yl 4-((lr,4r)-4-(3-cyanopyrazin-2- yloxy)cyclohexyloxy)piperidine- 1 -carboxylate;
3-((lr,4r)-4-(l-(3-(trifluoromethyl)-l,2,4-oxadiazol-5-yl)piperidin-4- yloxy)cyclohexyloxy)pyrazine-2-carbonitrile; 3-((lr,4r)-4-(l-(5-(trifluoromethyl)-l,2,4-oxadiazol-3-yl)piperidin-4- yloxy)cyclohexyloxy)pyrazine-2-carbonitrile;
1 -(trifluoromethyl)cyclobutyl 4-(( 1 r,4r)-4-(3-cyanopyrazin-2- yloxy)cyclohexyloxy)piperidine- 1 -carboxylate;
3-(trifluoromethyl)oxetan-3-yl 4-((l r,4r)-4-(3-cyanopyrazin-2- yloxy)cyclohexyloxy)piperidine- 1 -carboxylate;
1, 1, 1 -trifluoro-2-methylpropan-2-yl 4-(( 1 r,4r)-4-(3-cyanopyrazin-2- yloxy)cyclohexyloxy)piperidine- 1 -carboxylate;
l, l, l,3,3,3-hexafluoropropan-2-yl 4-((lr,4r)-4-(3-cyanopyrazin-2- yloxy)cyclohexyloxy)piperidine- 1 -carboxylate;
1 -(trifluoromethyl)cyclopentyl 4-(( 1 r,4r)-4-(3-cyanopyrazin-2- yloxy)cyclohexyloxy)piperidine- 1 -carboxylate;
3-(( 1 r,4r)-4-( 1 -(5-(2-fluoropropan-2-yl)- 1 ,2,4-oxadiazol-3-yl)piperidin-4- yloxy)cyclohexyloxy)pyrazine-2-carbonitrile;
3-(( 1 r,4r)-4-( 1 -(3-(2-fluoropropan-2-yl)- 1 ,2,4-oxadiazol-5-yl)piperidin-4- yloxy)cyclohexyloxy)pyrazine-2-carbonitrile;
3-(( 1 r,4r)-4-( 1 -(3-(2-fluoropropan-2-yl)- 1 ,2,4-oxadiazol-5-yl)piperidin-4- yloxy)cyclohexyloxy)picolinonitrile;
3-((lr,4r)-4-(l-(5-(2-fluoropropan-2-yl)-l,2,4-oxadiazol-3-yl)piperidin-4- yloxy)cyclohexyloxy)picolinonitrile;
3-((lr,4r)-4-(l-(5-chloropyrimidin-2-yl)piperidin-4- yloxy)cyclohexyloxy)picolinonitrile;
(R)- 1, 1, 1 -trifluoropropan-2-yl 4-(( 1 r,4r)-4-(2-cyanopyridin-3- yloxy)cyclohexyloxy)piperidine- 1 -carboxylate;
(S)-l, l, l-trifluoropropan-2-yl 4-((lr,4r)-4-(2-cyanopyridin-3- yloxy)cyclohexyloxy)piperidine- 1 -carboxylate;
3-((lr,4r)-4-(l-(3-isopropyl-l,2,4-oxadiazol-5-yl)piperidin-4- yloxy)cyclohexyloxy)picolinonitrile; and
1-methylcyclopropyl 4-((lr,4r)-4-(2-cyanopyridin-3- yloxy)cyclohexyloxy)piperidine- 1 -carboxylate.
17. A composition comprising a compound according to any one of claims 1 to 16.
18. A pharmaceutical product selected from: a pharmaceutical composition, a formulation, a unit dosage form, and a kit; each comprising a compound according to any one of claims 1 to 16.
19. A pharmaceutical composition comprising a compound according to any one of claims 1 to 16, and a pharmaceutically acceptable carrier.
20. A method for preparing a pharmaceutical composition comprising the step of admixing a compound according to any one of claims 1 to 16, and a pharmaceutically acceptable carrier.
21. A pharmaceutical product selected from: a pharmaceutical composition, a formulation, a unit dosage form, a combined preparation, a twin pack, and a kit; each comprising a compound according to any one of claims 1 to 16, and a second pharmaceutical agent.
22. A method for preparing a pharmaceutical composition comprising the step of admixing a compound according to any one of claims 1 to 16, a second pharmaceutical agent, and a pharmaceutically acceptable carrier.
23. A method for the treatment of a disorder selected from: a GPR119-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic-related disorder; and obesity; in an individual; comprising administering to said individual in need thereof, a therapeutically effective amount of: a compound according to any one of claims 1 to 16; a composition according to claim 17; a pharmaceutical product according to claim 18 or 21; or a pharmaceutical composition according to claim 19.
24. Use of a compound according to any one of claims 1 to 16 or a composition according to claim 17; in the manufacture of a medicament for treating a disorder in an individual, wherein said disorder is selected from: a GPR119-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic-related disorder; and obesity.
25. A compound according to any one of claims 1 to 16; a composition according to claim 17; a pharmaceutical product according to claim 18 or 21; or a pharmaceutical composition according to claim 19; for use in a method of treatment of the human or animal body by therapy.
26. A compound according to any one of claims 1 to 16; a composition according to claim 17; a pharmaceutical product according to claim 18 or 21; or a pharmaceutical composition according to claim 19; for use in a method of treating a disorder in an individual, wherein said disorder is selected from: a GPR119-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic-related disorder; and obesity.
27. A method for the treatment of a disorder selected from: a GPR119-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic-related disorder; and obesity; in an individual; comprising administering to said individual in need thereof, a therapeutically effective amount of a compound according to any one of claims 1 to 16; a composition according to claim 17; a pharmaceutical product according to claim 18; or a pharmaceutical composition according to claim 19; each in combination with a therapeutically effective amount of a second pharmaceutical agent.
28. Use of a compound according to any one of claims 1 to 16 or a composition according to claim 17 in combination with a second pharmaceutical agent, in the manufacture of a medicament for the treatment of a disorder selected from: a GPR119-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic-related disorder; and obesity.
29. Use of a pharmaceutical agent in combination with a compound according to any one of claims 1 to 16 or a composition according to claim 17, in the manufacture of a medicament for the treatment of a disorder selected from: a GPR119-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic-related disorder; and obesity.
30. A compound according to any one of claims 1 to 16; a composition according to claim 17; a pharmaceutical product according to claim 18; or a pharmaceutical composition according to claim 19; in combination with a second pharmaceutical agent for use in a method of treatment of the human or animal body by therapy.
- I l l -
31. A compound according to any one of claims 1 to 16; a composition according to claim 17; a pharmaceutical product according to claim 18; or a pharmaceutical composition according to claim 19; in combination with a second pharmaceutical agent for use in a method of treating a disorder selected from: a GPRl 19-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic-related disorder; and obesity; in an individual.
32. A pharmaceutical agent in combination with a compound according to any one of claims 1 to 16; a composition according to claim 17; a pharmaceutical product according to claim 18; or a pharmaceutical composition according to claim 19; for use in a method of treatment of the human or animal body by therapy.
33. A pharmaceutical agent in combination with a compound according to any one of claims 1 to 16; a composition according to claim 17; a pharmaceutical product according to claim 18; or a pharmaceutical composition according to claim 19; for use in a method of treating a disorder selected from: a GPRl 19-related disorder; a condition ameliorated by increasing secretion of an incretin; a condition ameliorated by increasing a blood incretin level; a condition characterized by low bone mass; a neurological disorder; a metabolic-related disorder; and obesity; in an individual.
34. The method according to claim 23 or 27; the use according to any one of claims 24, 28, and 29; the compound according to claim 26 or 31; the composition according to claim 26 or 31; the pharmaceutical product according to claim 26 or 31; the pharmaceutical composition according to claim 26 or 31; or the pharmaceutical agent according to claim 33; wherein said incretin is GLP-1.
35. The method according to claim 23 or 27; the use according to any one of claims 24, 28, and 29; the compound according to claim 26 or 31; the composition according to claim 26 or 31; the pharmaceutical product according to claim 26 or 31; the pharmaceutical composition according to claim 26 or 31; or the pharmaceutical agent according to claim 33; wherein said incretin is GIP.
36. The method according to claim 23 or 27; the use according to any one of claims 24, 28, and 29; the compound according to claim 26 or 31; the composition according to claim 26 or 31; the pharmaceutical product according to claim 26 or 31; the pharmaceutical composition according to claim 26 or 31; or the pharmaceutical agent according to claim 33; wherein said incretin is PYY.
37. The method according to claim 23 or 27; the use according to any one of claims 24, 28, and 29; the compound according to claim 26 or 31; the composition according to claim 26 or 31; the pharmaceutical product according to claim 26 or 31; the pharmaceutical composition according to claim 26 or 31; or the pharmaceutical agent according to claim 33; wherein said disorder is a metabolic-related disorder selected from: diabetes, 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, glycosuria, metabolic acidosis, a cataract, 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, 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, motor-neuron disease, traumatic brain injury, spinal cord injury, and obesity.
38. The method according to claim 23 or 27; the use according to any one of claims 24, 28, and 29; the compound according to claim 26 or 31; the composition according to claim 26 or 31; the pharmaceutical product according to claim 26 or 31; the pharmaceutical composition according to claim 26 or 31; or the pharmaceutical agent according to claim 33; wherein said disorder is type 2 diabetes.
39. The composition according to any one of claims 30 to 33; the method according to claim 22 or 27; the pharmaceutical product according to any one of claims 21, and 30 to 33; the pharmaceutical composition according to any one of claims 30 to 33; the method according to claim 27; the use according to claim 28 or 29; the compound according to any one of claims 30 to 33; or the pharmaceutical agent according to claim 32 or 33; wherein said pharmaceutical agent or said second pharmaceutical agent is selected from: A. an inhibitor of DPP-IV selected from the following inhibitors of DPP-IV 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(5)-carbonitrile;
(llS,,3.S,,5lS,)-2-[2(lS,)-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-[Ar-[3(R)-pyrrolidinyl]glycyl]pyrrolidin-2(R)-yl boronic acid;
4(5)-fluoro- 1 - [2- [( lR,35)-3-( \H- 1 ,2,4-triazol- 1 - ylmethyl)cyclopentylamino]acetyl]pyrrolidine-2(lSr)-carbonitrile;
14(25,35, UbS)-2-ainino-9, 10-dimethoxy-2,3,4,6,7, 1 lb-hexahydro-l - pyrido[2, 1 -a]isoquinolin-3-yl] -4(5)-(fluoromethyl)pyrrolidin-2-one;
(25,45)-2-cyano-4-fluoro-l-[(2-hydroxy- 1, 1 -dimethyl)
ethylamino] acetylpyrrolidine;
8-(cw-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- ((35,45)-4-amino-l-(4-(3,3-difluoropyrrolidin-l-yl)-l,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(2//)-yl)methyl)-4-fluorobenzonitrile;
5- { (5)-2- [2-((5)-2-cyano-pyrrolidin- 1 -yl)-2-oxo-ethylamino] -propyl }-5-(l/f- tetrazol-5-yl)10,l l-dihydro-5/i-dibenzo[a,d]cycloheptene-2,8-dicarboxylic acid bis- dimethylamide;
((25,45)-4-(4-(3-methyl- l-phenyl-l/i-pyrazol-5-yl)piperazin-l-yl)pyrrolidin-2- yl)(thiazolidin-3-yl)methanone;
(25,45)- 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-5/i-pyrrolo[3,2-d]pyrimidin-5-yl}methyl)-4-fluorobenzonitrile;
(25)- 1- { [2-(5-methyl-2-phenyl-oxazol-4-yl)-ethylamino] -acetyl } -pyrrolidine-2- carbonitrile;
(25)- 1- { [1, 1 -dimethyl-3-(4-pyridin-3-yl-imidazol- 1 -yl)-propylamino] -acetyl } - pyrrolidine-2-carbonitrile; (33-difluoropyrrolidin-l-yl)-((25,45)-4-(4-(pyrimidin-2-yl)piperazin-l- yl)pyrrolidin-2-yl) methano ne ;
(2,S,,4lS,)-l-[(2lS,)-2-amino-3,3-bis(4-fluorophenyl)propanoyl]-4- fluoropyrrolidine-2-carbonitrile;
(2S,5R)-5-ethynyl- 1 - {Ar-(4-methyl- 1 -(4-carboxy-pyridin-2-yl)piperidin-4- yl)glycyl }pyrrolidine-2-carbonitrile; and
(15,6R)-3-{ [3 trifluorome l)-5,6-dmydro[l,2,4]triazolo[4,3-a]pyraziii-7(8/i)- yl]carbonyl}-6-(2,4,5-trifluorophenyl)cyclohex-3-en-l -amine;
B. a biguanide selected from the following biguanides, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
(phenylethyl)biguanide;
dimethylbiguanide;
butylbiguanide; and
l-(p-chlorophenyl)-5-isopropylbiguanide;
C. an alpha-glucosidase inhibitor selected from the following alpha-glucosidase inhibitors, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
(2R,3R,4R,5R)-4-((2R,3R,4R,5.S,,6R)-5-((2R,3R,4lS,,5lS,,6R)-3,4-dihydroxy-6- methyl-S-CCl.S'^R^.S'^^^^^-trihydroxy-S-Chydroxymethy^cyclohex^- enylamino)tetrahydro-2/i-pyran-2-yloxy)-3,4-dihydroxy-6-(hydroxymethyl)tetrahydro- 2/i-pyran-2-yloxy)-2,3,5,6-tetrahydroxyhexanal;
(2R,3R,4R,5lSr)-l-(2-hydroxyethyl)-2-(hydroxymethyl)piperidine-3,4,5-triol; and
(lS,2S,3R,4S,5S)-5-(l,3-dihydroxypropan-2-ylamino)-l- (hydroxymethyl)cyclohexane- 1 ,2,3,4-tetraol;
D. a sulfonylurea selected from the following sulfonylureas, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
Ar-(4-(Ar-(cyclohexylcarbamoyl)sulfamoyl)phenethyl)-5-methylpyrazine-2- carboxamide);
5-chloro-Ar-(4-(Ar-(cyclohexylcarbamoyl)sulfamoyl)phenethyl)-2- methoxybenzamide; and
3-ethyl-4-methyl-N-(4-(N-((lr,4r)-4- methylcyclohexylcarbamoyl)sulfamoyl)phenethyl)-2-oxo-2,5-dihydro- 1/i-pyrrole- 1 - carboxamide;
E. a SGLT2 inhibitor selected from the following SGLT2 inhibitors, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
(2lS,,3R,4R,5.S,,6R)-2-(4-chloro-3-(4-ethoxybenzyl)phenyl)-6- (hydroxymethyl)tetrahydro-2/i-pyran-3,4,5-triol; ethyl ((2R,3lS,,4lS,,5R,6.S,)-3,4,5-trihydroxy-6-(4-(4-isopropoxybenzyl)-l- isopropyl-5-methyl-l/i-pyrazol-3-yloxy)tetrahydro-2/i-pyran-2-yl)methyl carbonate; and
ethyl ((2R,3S,4S,5R,6¾-3,4,5-trihydroxy-6-(2-(4- methoxybenzyl)phenoxy)tetrahydro-2/i-pyran-2-yl)methyl carbonate; and
F. a meglitinide selected from the following meglitinides, and pharmaceutically acceptable salts, solvates, and hydrates thereof:
(¾-2-ethoxy-4-(2-(3-methyl- 1 -(2-(piperidin- 1 -yl)phenyl)butylamino)-2- oxoethyl)benzoic acid;
(R)-2-((lr,4R)-4-isopropylcyclohexanecarboxamido)-3-phenylpropanoic acid; and
(^^-benzyl^-CCSaRJa^-m-isoindol^CS^Sa/i^/i^/i^/iJ/iJa/^-yl)^- oxobutanoic acid.
PCT/US2013/068864 2012-11-08 2013-11-07 Modulators of gpr119 and the treatment of disorders related thereto WO2014074668A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201261723937P 2012-11-08 2012-11-08
US61/723,937 2012-11-08

Publications (1)

Publication Number Publication Date
WO2014074668A1 true WO2014074668A1 (en) 2014-05-15

Family

ID=49667569

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/068864 WO2014074668A1 (en) 2012-11-08 2013-11-07 Modulators of gpr119 and the treatment of disorders related thereto

Country Status (1)

Country Link
WO (1) WO2014074668A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110862395A (en) * 2019-11-13 2020-03-06 株洲千金药业股份有限公司 Preparation method of raw material compound for preparing tadalafil important impurities
US11279702B2 (en) 2020-05-19 2022-03-22 Kallyope, Inc. AMPK activators
US11407768B2 (en) 2020-06-26 2022-08-09 Kallyope, Inc. AMPK activators

Citations (255)

* 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
CA2123128A1 (en) 1991-11-22 1993-05-27 Roger Snow 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
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
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
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
WO1999061431A1 (en) 1998-05-28 1999-12-02 Probiodrug Gesellschaft für Arzneimittelforschung mbH New dipeptidyl peptidase iv effectors
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
WO2000010549A1 (en) 1998-08-21 2000-03-02 Point Therapeutics, Inc. 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
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
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
WO2002001427A1 (en) 2000-06-29 2002-01-03 Worldsmart Technology Pty Ltd Protecting against impulse expenditure
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
WO2002030891A1 (en) 2000-10-06 2002-04-18 Tanabe Seiyaku Co., Ltd. Aliphatic 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
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
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
WO2003000181A2 (en) 2001-06-20 2003-01-03 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
WO2003000250A1 (en) 2001-06-25 2003-01-03 Ferring Bv 3-fluoro-pyrrolidines as antidiabetic agents
WO2003002530A2 (en) 2001-06-27 2003-01-09 Smithkline Beecham Corporation Pyrrolidines 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
WO2003002531A2 (en) 2001-06-27 2003-01-09 Smithkline Beecham Corporation 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
WO2003002553A2 (en) 2001-06-27 2003-01-09 Smithkline Beecham Corporation Fluoropyrrolidines as dipeptidyl peptidase inhibitors
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
WO2003004496A1 (en) 2001-07-03 2003-01-16 Novo Nordisk A/S Dpp-iv-inhibiting purine derivatives for the treatment 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
WO2003024942A1 (en) 2001-09-14 2003-03-27 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
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
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
US20030105077A1 (en) 2001-07-03 2003-06-05 Kanstrup Anders Bendtz Heterocyclic compounds that are inhibitors of the enzyme DPP-IV
WO2003045228A2 (en) 2001-11-26 2003-06-05 Trustees Of Tufts College Methods for treating autoimmune disorders, and reagents related thereto
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
WO2003055881A1 (en) 2001-12-27 2003-07-10 F. Hoffmann-La Roche Ag Pyrido(2,1-a)isoquinoline derivatives as dpp-iv inhibitors
US20030130199A1 (en) 2001-06-27 2003-07-10 Von Hoersten Stephan Dipeptidyl peptidase IV inhibitors and their uses as anti-cancer agents
WO2003057666A2 (en) 2001-12-26 2003-07-17 Guilford Pharmaceuticals Inhibitors of dipeptidyl peptidase iv
WO2003068748A1 (en) 2002-02-13 2003-08-21 F. Hoffmann-La Roche Ag Novel pyridine- and quinoline-derivatives
WO2003068757A1 (en) 2002-02-13 2003-08-21 F. Hoffmann-La Roche Ag Novel pyridin- and pyrimidin-derivatives
JP2003238566A (en) 2001-02-02 2003-08-27 Takeda Chem Ind Ltd Condensed heterocyclic compound
EP1338592A1 (en) 2002-02-22 2003-08-27 Nippon Zoki Pharmaceutical Co., Ltd. Novel 2-phenylpiperazine derivatives
EP1338651A1 (en) 2000-12-01 2003-08-27 Yamanouchi Pharmaceutical Co. Ltd. Method of screening remedy for diabetes
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
WO2004007517A1 (en) 2002-07-11 2004-01-22 Aventis Pharma Deutschland Gmbh Novel thiophenylglycoside derivatives, methods for production thereof, medicaments comprising said compounds and use thereof
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
JP2004026820A (en) 2002-05-09 2004-01-29 Taisho Pharmaceut Co Ltd Dipeptidyl peptidase iv inhibitor
WO2004009544A1 (en) 2002-07-23 2004-01-29 Yamanouchi Pharmaceutical Co., Ltd. 2-cyano-4-fluoropyrrolidine derivative or its salt
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
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
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
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)
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
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
WO2004047755A2 (en) 2002-11-22 2004-06-10 Japan Tobacco Inc. Fused bicyclic nitrogen-containing heterocycles
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
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
WO2004052362A1 (en) 2002-12-10 2004-06-24 Novartis Ag Combination of an dpp-iv inhibitor and a ppar-alpha compound
WO2004052850A2 (en) 2002-12-09 2004-06-24 Bristol-Myers Squibb Company Methods and compounds producing dipeptidyl peptidase iv inhibitors and intermediates thereof
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
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
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
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
WO2004080990A1 (en) 2003-03-14 2004-09-23 Astellas Pharma Inc. C-glycoside derivatives and salts thereof
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
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
WO2004087053A2 (en) 2003-03-25 2004-10-14 Syrrx, Inc. Dipeptidyl peptidase 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
WO2004100881A2 (en) 2003-05-09 2004-11-25 Bayer Pharmaceuticals Corporation Preparation and use of aryl alkyl acid derivatives for the treatment of obesity
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)
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
WO2004103993A1 (en) 2003-05-14 2004-12-02 Syrrx, Inc. Dipeptidyl peptidase inhibitors
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
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
US20040259902A1 (en) 2003-06-20 2004-12-23 Markus Boehringer Pyrido [2,1-a] isoquinoline derivatives
WO2004110436A1 (en) 2003-06-06 2004-12-23 Merck & Co., Inc. Fused indoles as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
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)
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
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
WO2005011601A2 (en) 2003-08-01 2005-02-10 Pfizer Products, Inc. 6-menbered heteroaryl compounds for the treatment of neurodegenerative disorders
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
WO2005012326A1 (en) 2003-08-01 2005-02-10 Tanabe Seiyaku Co., Ltd. Novel compounds having inhibitory activity against sodium-dependant transporter
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
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
US20050059724A1 (en) 2003-07-25 2005-03-17 Aventis Pharma Deutschland Gmbh Novel cyanopyrrolidides, 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)
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
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
WO2005030751A2 (en) 2003-09-08 2005-04-07 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
WO2005033099A2 (en) 2003-10-03 2005-04-14 Glenmark Pharmaceuticals Ltd. Novel dipeptidyl peptidase iv inhibitors; processes for their preparation and compositions thereof
WO2005032590A1 (en) 2003-10-03 2005-04-14 Takeda Pharmaceutical Company Limited Remedy for diabetes
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
WO2005072740A2 (en) 2004-01-30 2005-08-11 Japan Tobacco Inc. Anorectic compounds
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
WO2005082348A2 (en) 2004-02-23 2005-09-09 Trustees Of Tufts College Inhibitors of dipeptidylpeptidase iv for regulating glucose metabolism
WO2005082849A1 (en) 2004-02-23 2005-09-09 Trustees Of Tufts College Lactams as conformationally constrained peptidomimetic inhibitors
WO2005087235A1 (en) 2004-03-09 2005-09-22 National Health Research Institutes Pyrrolidine compounds
WO2005092877A1 (en) 2004-03-16 2005-10-06 Boehringer Ingelheim International Gmbh Glucopyranosyl-substituted benzol derivatives, drugs containing said compounds, the use thereof and method for the production thereof
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
WO2006001161A1 (en) 2004-06-28 2006-01-05 Kabushiki Kaisha Toshiba Storage medium processing method, storage medium processing apparatus, and program
WO2006019020A1 (en) 2004-08-16 2006-02-23 Sankyo Company, Limited Substituted ureas
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
WO2006044775A2 (en) 2004-10-15 2006-04-27 Bayer Pharmaceuticals Corporation Preparation and use of biphenyl-4-yl-carbonylamino acid derivatives for the treatment of obesity
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
WO2006080421A1 (en) 2005-01-28 2006-08-03 Chugai Seiyaku Kabushiki Kaisha Spiroketal derivative and use thereof as diabetic medicine
WO2006082010A1 (en) 2005-02-07 2006-08-10 F.Hoffmann-La Roche Ag Inhibitors of diacylglycerol acyltransferase (dgat)
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
WO2006113919A2 (en) 2005-04-19 2006-10-26 Bayer Pharmaceuticals Corporation Aryl alkyl acid derivatives for and use thereof
WO2006116157A2 (en) 2005-04-22 2006-11-02 Alantos Pharmaceuticals Holding, Inc. Dipeptidyl peptidase-iv inhibitors
WO2006134317A1 (en) 2005-06-11 2006-12-21 Astrazeneca Ab Oxadiazole derivatives as dgat 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
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
WO2007126957A2 (en) 2006-03-31 2007-11-08 Novartis Ag New compounds
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
WO2008011485A2 (en) 2006-07-19 2008-01-24 Allergan, Inc. Methods for treating chronic pain
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
WO2008067465A1 (en) 2006-11-29 2008-06-05 Takeda Pharmaceutical Company Limited Polymorphs of succinate salt of 2-[6-(3-amino-piperidin-1-yl)-3-methyl-2,4-dioxo-3,4-dihydro-2h-pyrimidin-1-ylmethy]-4-fluor-benzonitrile and methods of use therefor
US20080146818A1 (en) 2004-02-05 2008-06-19 Yasumichi Fukuda Bicycloester Derivative
WO2008109591A1 (en) 2007-03-08 2008-09-12 Lexicon Pharmaceuticals, Inc. Phlorizin analogs as inhibitors of sodium glucose co-transporter 2
WO2009016462A2 (en) 2007-08-02 2009-02-05 Pfizer Products Inc. Substituted bicyclolactam compounds
WO2009084497A1 (en) 2007-12-28 2009-07-09 Dainippon Sumitomo Pharma Co., Ltd. Methyl-substituted piperidine derivative
WO2010070343A1 (en) 2008-12-19 2010-06-24 Astrazeneca Ab 1,3,4-oxadiazole derivatives and their uses to treat diabetes
WO2011127051A1 (en) 2010-04-06 2011-10-13 Arena Pharmaceuticals, Inc. Modulators of the gpr119 receptor and the treatment of disorders related thereto
WO2012040279A1 (en) 2010-09-22 2012-03-29 Arena Pharmaceuticals, Inc. Modulators of the gpr119 receptor and the treatment of disorders related thereto
WO2012051488A1 (en) 2010-10-14 2012-04-19 Novartis Ag Pharmaceutical compositions containing a dgat1 inhibitor
WO2012145361A1 (en) 2011-04-19 2012-10-26 Arena Pharmaceuticals, Inc. Modulators of the gpr119 receptor and the treatment of disorders related thereto

Patent Citations (424)

* 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
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
WO1991016339A1 (en) 1990-04-14 1991-10-31 New England Medical Center Hospitals, Inc. 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
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
CA2123128A1 (en) 1991-11-22 1993-05-27 Roger Snow Method for making a prolineboronate ester
WO1993010127A1 (en) 1991-11-22 1993-05-27 Boehringer Ingelheim Pharmaceuticals, Inc. 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
WO1995015309A1 (en) 1993-12-03 1995-06-08 Ferring B.V. Dp-iv-serine protease inhibitors
EP0731789A1 (en) 1993-12-03 1996-09-18 Ferring B.V. Dp-iv-serine protease inhibitors
JPH09509921A (en) 1993-12-03 1997-10-07 フェーリング ベスローテン フェンノートシャップ Enzyme 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
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
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
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
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
US6040145A (en) 1997-05-07 2000-03-21 Tufts University Potentiation of the immune response
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
US6100234A (en) 1997-05-07 2000-08-08 Tufts University 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
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
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
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
WO1999061431A1 (en) 1998-05-28 1999-12-02 Probiodrug Gesellschaft für Arzneimittelforschung mbH New dipeptidyl peptidase iv effectors
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
EP1215207A2 (en) 1998-05-28 2002-06-19 Probiodrug AG Salts of isoleucyl-thiazolidine and -pyrrolidine and their use as dipeptidylpeptidase inhibitors
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
DE19828113A1 (en) 1998-06-24 2000-01-05 Probiodrug Ges Fuer Arzneim Prodrugs of Dipeptidyl Peptidase IV Inhibitors
WO1999067278A1 (en) 1998-06-24 1999-12-29 Probiodrug Gesellschaft für Arzneimittelforschung mbH 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
EP1104293A1 (en) 1998-08-21 2001-06-06 Point Therapeutics, Inc. Regulation of substrate activity
CA2339537A1 (en) 1998-08-21 2000-03-02 Barbara Wallner Regulation of substrate activity
WO2000010549A1 (en) 1998-08-21 2000-03-02 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
WO2000023421A1 (en) 1998-10-22 2000-04-27 Idun Pharmaceuticals, Inc. (SUBSTITUTED)ACYL DIPEPTIDYL INHIBITORS OF THE ICE/ced-3 FAMILY OF CYSTEINE PROTEASES
US6242422B1 (en) 1998-10-22 2001-06-05 Idun Pharmacueticals, 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
EP1123272A1 (en) 1998-10-22 2001-08-16 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
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
WO2000034241A1 (en) 1998-12-10 2000-06-15 Novartis Ag N-substituted 2-cyanopyrrolidines
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
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
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
JP2000327689A (en) 1999-05-17 2000-11-28 Microbial Chem Res Found Sulfostin analogue, and production of sulfostin and its analogue
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
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
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
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
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
WO2001052825A2 (en) 2000-01-21 2001-07-26 Novartis Ag Combinations comprising dipeptidylpeptidase-iv inhibitors and antidiabetic agents
EP1248604A2 (en) 2000-01-21 2002-10-16 Novartis AG Combinations comprising dipeptidylpeptidase-iv inhibitors and antidiabetic agents
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
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
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
US20020103384A1 (en) 2000-01-24 2002-08-01 Anders Kanstrup Therapeutically active and selective heterocyclic compounds that 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
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
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
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
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
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
EP1280797A1 (en) 2000-04-26 2003-02-05 Ferring B.V. 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
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
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
WO2001096295A2 (en) 2000-06-13 2001-12-20 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
WO2002001427A1 (en) 2000-06-29 2002-01-03 Worldsmart Technology Pty Ltd Protecting against impulse expenditure
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
WO2002030890A1 (en) 2000-10-06 2002-04-18 Tanabe Seiyaku Co., Ltd. Nitrogenous five-membered ring compounds
JP2002356472A (en) 2000-10-06 2002-12-13 Tanabe Seiyaku Co Ltd 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
WO2002030891A1 (en) 2000-10-06 2002-04-18 Tanabe Seiyaku Co., Ltd. Aliphatic nitrogenous five-membered ring compounds
JP2002356471A (en) 2000-10-06 2002-12-13 Tanabe Seiyaku Co Ltd Aliphatic nitrogen-containing five-membered ring compound
US20040229926A1 (en) 2000-10-06 2004-11-18 Tanabe Seiyaku Co., Ltd. Aliphatic nitrogen - containing 5 - 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
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
US20070112059A1 (en) 2000-11-10 2007-05-17 Taisho Pharmaceutical Co., Ltd. Cyanopyrrolidine derivatives
EP1338651A1 (en) 2000-12-01 2003-08-27 Yamanouchi Pharmaceutical Co. Ltd. Method of screening remedy for diabetes
EP1354882A1 (en) 2000-12-27 2003-10-22 Kyowa Hakko Kogyo Co., Ltd. Dipeptidyl peptidase iv inhibitor
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
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
WO2002062764A1 (en) 2001-02-02 2002-08-15 Takeda Chemical Industries, Ltd. Fused heterocyclic compounds
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
WO2002068420A1 (en) 2001-02-24 2002-09-06 Boehringer Ingelheim Pharma Gmbh & Co. Kg Xanthine derivative, production and use thereof as a medicament
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
US20040077645A1 (en) 2001-02-24 2004-04-22 Frank Himmelsbach Xanthine derivatives,production and use thereof as medicament
US20020198205A1 (en) 2001-02-24 2002-12-26 Frank Himmelsbach 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
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
US20040106656A1 (en) 2001-03-27 2004-06-03 Ashton Wallace T 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
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)
US6716843B2 (en) 2001-03-28 2004-04-06 Les Laboratoires Servier 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
US20030087950A1 (en) 2001-03-28 2003-05-08 Denanteuil Guillaume New alpha-amino acid sulphonyl compounds
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
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
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
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
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
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
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)
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
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
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
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
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
EP1399154A1 (en) 2001-06-25 2004-03-24 Ferring BV 3-fluoro-pyrrolidines as antidiabetic agents
WO2003002530A2 (en) 2001-06-27 2003-01-09 Smithkline Beecham Corporation Pyrrolidines 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
EP1399469A2 (en) 2001-06-27 2004-03-24 Probiodrug AG Peptide structures useful for competitive modulation of dipeptidyl peptidase iv catalysis
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
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
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
EP1399470A2 (en) 2001-06-27 2004-03-24 Probiodrug AG Dipeptidyl peptidase iv inhibitors and their uses as anti-cancer agents
EP1399420A2 (en) 2001-06-27 2004-03-24 SmithKline Beecham Corporation Pyrrolidines as dipeptidyl peptidase inhibitors
EP1399433A2 (en) 2001-06-27 2004-03-24 Smithkline Beecham Corporation Fluoropyrrolidines as dipeptidyl peptidase inhibitors
EP1399471A2 (en) 2001-06-27 2004-03-24 Probiodrug AG Use of dipeptidyl peptidase iv inhibitors as therapeutics for neurological disorders
WO2003072556A1 (en) 2001-06-27 2003-09-04 Prosidion Ltd. Glutaminyl based dpiv inhibitors
US20030119750A1 (en) 2001-06-27 2003-06-26 Hans-Ulrich Demuth Use of dipeptidyl peptidase IV 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
WO2003002595A2 (en) 2001-06-27 2003-01-09 Probiodrug Ag Dipeptidyl peptidase iv inhibitors and their uses as anti-cancer agents
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
WO2003002593A2 (en) 2001-06-27 2003-01-09 Probiodrug Ag Peptide structures useful for competitive modulation of dipeptidyl peptidase iv catalysis
WO2003002553A2 (en) 2001-06-27 2003-01-09 Smithkline Beecham Corporation Fluoropyrrolidines as dipeptidyl peptidase inhibitors
EP1406873A2 (en) 2001-06-27 2004-04-14 Smithkline Beecham Corporation 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
WO2003002531A2 (en) 2001-06-27 2003-01-09 Smithkline Beecham Corporation Fluoropyrrolidines as dipeptidyl peptidase inhibitors
US20030105077A1 (en) 2001-07-03 2003-06-05 Kanstrup Anders Bendtz 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
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
US6869947B2 (en) 2001-07-03 2005-03-22 Novo Nordisk A/S Heterocyclic compounds that are inhibitors of the enzyme DPP-IV
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
US6699871B2 (en) 2001-07-06 2004-03-02 Merck & Co., Inc. Beta-amino heterocyclic 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
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
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
WO2003015775A1 (en) 2001-08-17 2003-02-27 Probiodrug Ag New dipeptidyl peptidase iv inhibitors and their uses for lowering blood pressure levels
US20030119738A1 (en) 2001-09-06 2003-06-26 Andre Niestroj Novel inhibitors of dipeptidyl peptidase I
WO2003022871A2 (en) 2001-09-06 2003-03-20 Probiodrug Ag Peptides having a c- terminal hydroxylamino group as 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
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
US20040259883A1 (en) 2001-09-14 2004-12-23 Hiroshi Sakashita Thiazolidine derivative and medicinal use thereof
EP1463727A2 (en) 2001-09-19 2004-10-06 Novo Nordisk A/S Heterocyclic compounds that are inhibitors of the enzyme dpp-iv
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
WO2003035057A1 (en) 2001-10-23 2003-05-01 Ferring B.V. Inhibitors of dipeptidyl peptidase iv
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
WO2003035067A1 (en) 2001-10-23 2003-05-01 Ferring B.V. Novel dipeptidyl peptidase iv (dp-iv) inhibitors as anti-diabetic agents
US20050004205A1 (en) 2001-10-23 2005-01-06 Evans David M 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
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
US20030130281A1 (en) 2001-10-26 2003-07-10 Markus Boehringer 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
EP1442049A2 (en) 2001-11-09 2004-08-04 Probiodrug AG Substituted amino ketone compounds
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
WO2003045228A2 (en) 2001-11-26 2003-06-05 Trustees Of Tufts College Methods for treating autoimmune disorders, and reagents related thereto
CA2466870A1 (en) 2001-11-26 2003-06-05 Trustees Of Tufts College Methods for treating autoimmune disorders, and reagents related thereto
EP1469873A2 (en) 2001-11-26 2004-10-27 Trustees of Tufts College Peptidomimetic inhibitors of post-proline cleaving enzymes
WO2003057666A2 (en) 2001-12-26 2003-07-17 Guilford Pharmaceuticals Inhibitors of dipeptidyl peptidase iv
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
US6727261B2 (en) 2001-12-27 2004-04-27 Hoffman-La Roche Inc. Pyrido[2,1-A]Isoquinoline derivatives
EP1461337A1 (en) 2001-12-27 2004-09-29 F. Hoffmann-La Roche Ag Pyrido[2,1-a]isoquinoline derivatives as dpp-iv inhibitors
WO2003055881A1 (en) 2001-12-27 2003-07-10 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
US20030232788A1 (en) 2002-02-08 2003-12-18 Idun Pharmaceuticals, Inc. (Substituted)acyl dipeptidyl inhibitors of the ICE/ced-3 family of cysteine proteases
WO2003072528A2 (en) 2002-02-08 2003-09-04 Idun Pharmaceuticals, Inc. (substituted)acyl dipeptidyl inhibitors of the ice/ced-3 family of cysteine proteases
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
WO2003068757A1 (en) 2002-02-13 2003-08-21 F. Hoffmann-La Roche Ag Novel pyridin- and pyrimidin-derivatives
US20030195188A1 (en) 2002-02-13 2003-10-16 Markus Boehringer Pyridine and quinoline derivatives
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
US6867205B2 (en) 2002-02-13 2005-03-15 Hoffman-La Roche Inc. Pyridine and pyrimidine derivatives
EP1338592A1 (en) 2002-02-22 2003-08-27 Nippon Zoki Pharmaceutical Co., Ltd. Novel 2-phenylpiperazine derivatives
JP2004043429A (en) 2002-02-25 2004-02-12 Eisai Co Ltd New xanthine derivative and dppiv inhibitor
US20040082570A1 (en) 2002-02-25 2004-04-29 Eisai Co., Ltd. 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
EP1490335A2 (en) 2002-03-25 2004-12-29 Merck & Co., Inc. Beta-amino heterocyclic dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
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
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
US20030225102A1 (en) 2002-04-08 2003-12-04 Torrent Pharmaceuticals Ltd. Novel compounds and therapeutic uses thereof
EP1492777A1 (en) 2002-04-08 2005-01-05 Torrent Pharmaceuticals Ltd Thiazolidine-4-carbonitriles and analogues and their use as dipeptidyl-peptidase inhibitors
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
WO2003101958A2 (en) 2002-06-04 2003-12-11 Pfizer Products Inc. Flourinated 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
US20040242898A1 (en) 2002-06-04 2004-12-02 Pfizer Inc Synthesis of 3,3,4,4-tetrafluoropyrrolidine and novel dipeptidyl peptidase-IV inhibitor compounds
EP1513808A2 (en) 2002-06-04 2005-03-16 Pfizer Products 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
WO2003104229A1 (en) 2002-06-06 2003-12-18 エーザイ株式会社 Novel fused imidazole derivative
US20040116328A1 (en) 2002-06-06 2004-06-17 Eisai Co., Ltd. Condensed imidazole derivatives
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
WO2004000327A1 (en) 2002-06-24 2003-12-31 Zaidan Hojin Biseibutsu Kagaku Kenkyu Kai Therapeutic agent for type 2 diabetes
JP2004026678A (en) 2002-06-24 2004-01-29 Microbial Chem Res Found 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
WO2004007517A1 (en) 2002-07-11 2004-01-22 Aventis Pharma Deutschland Gmbh Novel thiophenylglycoside derivatives, methods for production thereof, medicaments comprising said compounds and use 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
WO2004014860A2 (en) 2002-08-08 2004-02-19 Takeda Pharmaceutical Company Limited Fused heterocyclic compounds as peptidase inhibitors
JP2004315496A (en) 2002-08-08 2004-11-11 Takeda Chem Ind Ltd Fused heterocyclic ring compound
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
WO2004018469A1 (en) 2002-08-22 2004-03-04 Boehringer Ingelheim Pharma Gmbh & Co. Kg Novel purine derivatives, production and use thereof as medicaments
DE10238477A1 (en) 2002-08-22 2004-03-04 Boehringer Ingelheim Pharma Gmbh & Co. Kg New purine derivatives, their production and their use as medicines
DE10238470A1 (en) 2002-08-22 2004-03-04 Boehringer Ingelheim Pharma Gmbh & Co. Kg New xanthine derivatives, their production and their use as medicines
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
WO2004047755A2 (en) 2002-11-22 2004-06-10 Japan Tobacco Inc. Fused bicyclic nitrogen-containing heterocycles
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
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
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
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
WO2004067509A1 (en) 2003-01-31 2004-08-12 Sanwa Kagaku Kenkyusho Co., Ltd. Compound inhibiting dipeptidyl peptidase iv
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
WO2004076433A1 (en) 2003-02-28 2004-09-10 Aic Dipeptidyl peptidase inhibitors
WO2004076434A1 (en) 2003-02-28 2004-09-10 Aic Dipeptidyl peptidase inhibitors
WO2004080990A1 (en) 2003-03-14 2004-09-23 Astellas Pharma Inc. C-glycoside derivatives and salts thereof
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
WO2004100881A2 (en) 2003-05-09 2004-11-25 Bayer Pharmaceuticals Corporation Preparation and use of aryl alkyl acid derivatives for the treatment of obesity
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
WO2004103993A1 (en) 2003-05-14 2004-12-02 Syrrx, Inc. Dipeptidyl peptidase inhibitors
WO2004104216A2 (en) 2003-05-21 2004-12-02 Bayer Healthcare Ag Diagnostics and therapeutics for diseases associated with dipeptidylpeptidase iv (dpp4)
WO2004104215A2 (en) 2003-05-21 2004-12-02 Bayer Healthcare Ag Diagnostics and therapeutics for diseases associated with dipeptidylpeptidase 7 (dpp7)
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
WO2004110436A1 (en) 2003-06-06 2004-12-23 Merck & Co., Inc. Fused indoles as dipeptidyl peptidase inhibitors for the treatment or prevention of diabetes
WO2004110375A2 (en) 2003-06-06 2004-12-23 Merck & Co., Inc. Combination therapy for the treatment 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
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
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
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
WO2005000846A1 (en) 2003-06-20 2005-01-06 F.Hoffmann-La Roche Ag Hexahydropyridoisoqinolines as dpp-iv inhibitors
WO2005000848A1 (en) 2003-06-20 2005-01-06 F. Hoffmann-La Roche Ag Pyrido` 2, 1-a - isoquinoline derivatives as dpp-iv inhibitors
WO2005003135A1 (en) 2003-06-24 2005-01-13 Merck & Co., Inc. Phosphoric acid salt of a dipeptidyl peptidase-iv inhibitor
US20050032804A1 (en) 2003-06-24 2005-02-10 Cypes Stephen Howard 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
US20050059716A1 (en) 2003-07-25 2005-03-17 Aventis Pharma Deutschland Gmbh Novel bicyclic cyanoheterocycles, 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
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
US20050059724A1 (en) 2003-07-25 2005-03-17 Aventis Pharma Deutschland Gmbh Novel cyanopyrrolidides, process for their preparation and their use as medicaments
WO2005011581A2 (en) 2003-07-31 2005-02-10 Merck & Co., Inc. Hexahydrodiazepinones as dipeptidyl peptidase-iv inhibitors for the treatment or prevention of diabetes
WO2005012326A1 (en) 2003-08-01 2005-02-10 Tanabe Seiyaku Co., Ltd. Novel compounds having inhibitory activity against sodium-dependant transporter
WO2005012249A2 (en) 2003-08-01 2005-02-10 Bristol-Myers Squibb Company Adamantyglycine- based inhibitors of dipeptidyl peptidase iv for the treatment of diabetes
US20050038020A1 (en) 2003-08-01 2005-02-17 Hamann Lawrence G. Adamantylglycine-based inhibitors of dipeptidyl peptidase IV and methods
WO2005011601A2 (en) 2003-08-01 2005-02-10 Pfizer Products, Inc. 6-menbered heteroaryl compounds for the treatment of neurodegenerative disorders
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
WO2005072740A2 (en) 2004-01-30 2005-08-11 Japan Tobacco Inc. Anorectic compounds
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
WO2005092877A1 (en) 2004-03-16 2005-10-06 Boehringer Ingelheim International Gmbh Glucopyranosyl-substituted benzol derivatives, drugs containing said compounds, the use thereof and method for the production thereof
WO2005116014A1 (en) 2004-05-12 2005-12-08 Pfizer Products Inc. Proline derivatives and their use as dipeptidyl peptidase iv inhibitors
WO2006001161A1 (en) 2004-06-28 2006-01-05 Kabushiki Kaisha Toshiba Storage medium processing method, storage medium processing apparatus, and program
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
WO2006019020A1 (en) 2004-08-16 2006-02-23 Sankyo Company, Limited Substituted ureas
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
WO2006044775A2 (en) 2004-10-15 2006-04-27 Bayer Pharmaceuticals Corporation Preparation and use of biphenyl-4-yl-carbonylamino acid derivatives for the treatment of obesity
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
WO2006080421A1 (en) 2005-01-28 2006-08-03 Chugai Seiyaku Kabushiki Kaisha Spiroketal derivative and use thereof as diabetic medicine
WO2006082010A1 (en) 2005-02-07 2006-08-10 F.Hoffmann-La Roche Ag Inhibitors of diacylglycerol acyltransferase (dgat)
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
WO2006113919A2 (en) 2005-04-19 2006-10-26 Bayer Pharmaceuticals Corporation Aryl alkyl acid derivatives for and use thereof
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
WO2006134317A1 (en) 2005-06-11 2006-12-21 Astrazeneca Ab Oxadiazole derivatives as dgat 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
WO2007126957A2 (en) 2006-03-31 2007-11-08 Novartis Ag New compounds
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
WO2008011485A2 (en) 2006-07-19 2008-01-24 Allergan, Inc. Methods for treating chronic pain
WO2008027273A2 (en) 2006-08-30 2008-03-06 Phenomix Corporation Solid citrate and tartrate salts of dpp-iv inhibitors
WO2008067465A1 (en) 2006-11-29 2008-06-05 Takeda Pharmaceutical Company Limited Polymorphs of succinate salt of 2-[6-(3-amino-piperidin-1-yl)-3-methyl-2,4-dioxo-3,4-dihydro-2h-pyrimidin-1-ylmethy]-4-fluor-benzonitrile and methods of use therefor
WO2008109591A1 (en) 2007-03-08 2008-09-12 Lexicon Pharmaceuticals, Inc. Phlorizin analogs as inhibitors of sodium glucose co-transporter 2
WO2009016462A2 (en) 2007-08-02 2009-02-05 Pfizer Products Inc. Substituted bicyclolactam compounds
WO2009084497A1 (en) 2007-12-28 2009-07-09 Dainippon Sumitomo Pharma Co., Ltd. Methyl-substituted piperidine derivative
WO2010070343A1 (en) 2008-12-19 2010-06-24 Astrazeneca Ab 1,3,4-oxadiazole derivatives and their uses to treat diabetes
WO2011127051A1 (en) 2010-04-06 2011-10-13 Arena Pharmaceuticals, Inc. Modulators of the gpr119 receptor and the treatment of disorders related thereto
WO2012040279A1 (en) 2010-09-22 2012-03-29 Arena Pharmaceuticals, Inc. Modulators of the gpr119 receptor and the treatment of disorders related thereto
WO2012051488A1 (en) 2010-10-14 2012-04-19 Novartis Ag Pharmaceutical compositions containing a dgat1 inhibitor
WO2012145361A1 (en) 2011-04-19 2012-10-26 Arena Pharmaceuticals, Inc. Modulators of the gpr119 receptor and the treatment of disorders related thereto

Non-Patent Citations (123)

* Cited by examiner, † Cited by third party
Title
"Bioreversible Carriers in Drug Design", 1987, AMERICAN PHARMACEUTICAL ASSOCIATION AND PERGAMON PRESS
"World Health Organization Technical Report Series 921", PREVENTION AND MANAGEMENT OF OSTEOPOROSIS, 2003
ABBOTT ET AL., BRAIN RES, vol. 1043, 2005, pages 139 - 144
ABE ET AL., J. NA.T 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
AM. J. PHYSIOL., vol. 268, 1995, pages G71 - G81
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
BERGE ET AL., JOURNAL OF PHARMACEUTICAL SCIENCES, vol. 66, 1977, pages 1 - 19
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, J PATHOL, 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
CHAUDHRI ET AL., ANNU REV PHYSIOL, vol. 70, 2008, pages 239 - 255
COLLIER, T. L., J. LABELLED COMPD RADIOPHARM., vol. 42, 1999, pages S264 - S266
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
GREENE, T. W.; WUTS, P. G. M.: "Protecting Groups in Organic Synthesis, 3rd Edition,", 1999, WILEY
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 DPP-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. O. ET AL., SCIENCE, vol. 280, 1998, pages 1371 - 1374
JONES ET AL., ANN. REP. MED CHEM, vol. 44, 2009, pages 149 - 170
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, XP002453370, DOI: doi:10.1517/13543776.15.10.1387
K.J. GUILLORY: "Polymorphism in Pharmaceutical Solids", vol. 95, 1999, MARCEL DEKKER, INC., article "Generation of Polymorphs, Hydrates, Solvates, and Amorphous Solids", pages: 202 - 209
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 BAS, M.-D., J. LABELLED COMPD. RADIOPHARM., vol. 44, 2001, pages S280 - S282
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
MARSO ET AL., DIABETES CARE, 5 February 2008 (2008-02-05)
MATSUDA ET AL., J BIOL 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., LIFE SCIENCES, vol. 41, 1987, pages 2157 - 2165
NAUCK ET AL., DIABETES, vol. 53, no. 3, 2004, pages 8190 - 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 NOTES, 2005, pages 11
OHASHI ET AL., HYPERTENSION, vol. 47, 2006, pages 1108 - 1116
OKADA ET AL., ENDOCRINOLOGY, 1993, pages 180
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
REMINGTON: "The Science and Practice of Pharmacy, 20,h Edition,", 2000, LIPPINCOTT WILLIAMS & WILKINS
RENSHAW ET AL., CURRENT DRUG TARGETS, vol. 6, 2005, pages 171 - 179
RIMOIN, D. L.: "Emery and Rimoin's Principles and Practice of Medical Genetics", vol. 1, 1996, pages: 1401 - 1402
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., NATMED, 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)
T. HIGUCHI; V. STELLA: "Pro-drugs as Novel Delivery Systems", vol. 14, A.C.S. SYMPOSIUM SERIES
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 OF DISEASE, vol. 20, 2005, pages 760 - 772
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 J PHYSIOL ENDOCRINOL METAB, vol. 292, 2007, pages E543 - E548
ZHU, G-D., J. ORG. CHEM., vol. 67, 2002, pages 943 - 948

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110862395A (en) * 2019-11-13 2020-03-06 株洲千金药业股份有限公司 Preparation method of raw material compound for preparing tadalafil important impurities
US11279702B2 (en) 2020-05-19 2022-03-22 Kallyope, Inc. AMPK activators
US11851429B2 (en) 2020-05-19 2023-12-26 Kallyope, Inc. AMPK activators
US11407768B2 (en) 2020-06-26 2022-08-09 Kallyope, Inc. AMPK activators

Similar Documents

Publication Publication Date Title
US20210340133A1 (en) Modulators of the gpr119 receptor and the treatment of disorders related thereto
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
WO2011005929A1 (en) Piperidine derivative and its use for the treatment of diabets and obesity
WO2012170702A1 (en) Modulators of the gpr119 receptor and the treatment of disorders related thereto
WO2013055910A1 (en) Modulators of the gpr119 receptor and the treatment of disorders related thereto
JP2009542702A (en) Treatment of metabolic regulators and disorders related to metabolism
EA018703B1 (en) Compounds and compositions as modulators of gpr119 activity
JP7106623B2 (en) Compounds active against nuclear receptors
WO2022219495A1 (en) 2-((4-((s)-2-(4-chloro-2-fluorophenyl)-2-methylbenzo[d][1,3]dioxol-4-yl)piperidin-1-yl)methyl)-1-(((s)-oxetan-2-yl)methyl)-1h-imidazole derivatives as activators of the glp1 receptor for the treatment of obesity
WO2014074668A1 (en) Modulators of gpr119 and the treatment of disorders related thereto
EA040124B1 (en) GPR119 RECEPTOR MODULATORS AND THE TREATMENT OF RELATED DISORDERS
EP4323354A1 (en) 2-((4-((s)-2-(4-chloro-2-fluorophenyl)-2-methylbenzo[d][1,3]dioxol-4-yl)piperidin-1-yl)methyl)-1-(((s)-oxetan-2-yl)methyl)-1h-imidazole derivatives as activators of the glp1 receptor for the treatment of obesity
WO2023152698A1 (en) 2-((4-((s)-2-(4-chloro-2-fluorophenyl)-2-methylbenzo[d][1,3]dioxol-4-yl)piperidin-1-yl)methyl)-1-(((s)-oxetan-2-yl)methyl)-1h-imidazole derivatives as activators of the glp1 receptor for the treatment of obesity
JP2024514826A (en) 2-((4-((S)-2-(4-chloro-2-fluorophenyl)-2-methylbenzo[D][1,3]dioxol-4-yl)piperidin-1-yl)methyl)-1-(((S)-oxetan-2-yl)methyl)-1H-imidazole derivatives as activators of the GLP1 receptor for the treatment of obesity

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: 13795912

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: 13795912

Country of ref document: EP

Kind code of ref document: A1