EP1899304A1 - Derives d'acide 5-amino-1h-pyrazole-3-carboxylique en tant qu'agonistes du recepteur couple aux proteines g (rcpg) rup38 pour traiter des troubles lies au metabolisme - Google Patents

Derives d'acide 5-amino-1h-pyrazole-3-carboxylique en tant qu'agonistes du recepteur couple aux proteines g (rcpg) rup38 pour traiter des troubles lies au metabolisme

Info

Publication number
EP1899304A1
EP1899304A1 EP06770837A EP06770837A EP1899304A1 EP 1899304 A1 EP1899304 A1 EP 1899304A1 EP 06770837 A EP06770837 A EP 06770837A EP 06770837 A EP06770837 A EP 06770837A EP 1899304 A1 EP1899304 A1 EP 1899304A1
Authority
EP
European Patent Office
Prior art keywords
alkoxy
compound according
alkyl
group
optionally substituted
Prior art date
Legal status (The legal status 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 status listed.)
Withdrawn
Application number
EP06770837A
Other languages
German (de)
English (en)
Inventor
Philip J. Skinner
Graeme Semple
Peter Webb
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Arena Pharmaceuticals Inc
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 EP1899304A1 publication Critical patent/EP1899304A1/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/14Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D231/38Nitrogen atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/12Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings

Definitions

  • Formula (I) and pharmaceutically acceptable salts thereof which exhibit useful pharmacological properties, for example, as agonists for the G protein-coupled receptor (GPCR) referred to herein as RUP38.
  • GPCR G protein-coupled receptor
  • compositions containing compounds of the invention are also provided by the present invention, and methods of using the compounds and compositions of the invention in the treatment of metabolic-related disorders, including dyslipidemia, atherosclerosis, coronary heart disease, insulin resistance, obesity, impaired glucose tolerance, atheromatous disease, hypertension, stroke, Syndrome X, heart disease, type 2 diabetes, hypo-HDL related atherosclerotic risk, ischemic cerebrovascular disease, peripheral vascular disease, stroke, myocardial infarction and the like.
  • metabolic-related disorders including dyslipidemia, atherosclerosis, coronary heart disease, insulin resistance, obesity, impaired glucose tolerance, atheromatous disease, hypertension, stroke, Syndrome X, heart disease, type 2 diabetes, hypo-HDL related atherosclerotic risk, ischemic cerebrovascular disease, peripheral vascular disease, stroke, myocardial infarction and the like.
  • Atherosclerosis and stroke are the numbers one and number three leading causes of death of both men and women in the United States.
  • Type 2 diabetes is a public health problem that is serious, widespread and increasing. Elevated levels of low density lipoprotein (LDL) cholesterol or low levels of high density lipoprotein (HDL) cholesterol are, independently, risk factors for atherosclerosis and associated cardiovascular pathologies.
  • high levels of plasma free fatty acids are associated with insulin resistance and type 2 diabetes.
  • One strategy for decreasing 0 LDL-cholesterol, increasing HDL-cholesterol, and decreasing plasma free fatty acids is to inhibit lipolysis in adipose tissue. This approach involves regulation of hormone sensitive lipase, which is the rate-limiting enzyme in lipolysis.
  • Lipolytic agents increase cellular levels of cAMP, which leads to activation of hormone sensitive lipase within adipocytes. Agents that lower intracellular cAMP levels, by contrast, would be antilipolytic. 5 It is also worth noting in passing that an increase in cellular levels of cAMP down- regulates the secretion of adiponectin from adipocytes [Delporte, ML et al. Biochem J (2002) July; the disclosure of which is incorporated by reference in its entirety]. Reduced levels of plasma adiponectin have been associated with metabolic-related disorders, including atherosclerosis, coronary heart disease, insulin resistance, type 2 diabetes, hypertension and obseity [Matsuda, M et al.
  • Compounds of the invention inhibit the production and release of free fatty acids from adipose tissue, likely via an inhibition of adenylyl cyclase, a decrease in intracellular cAMP levels, and a concomitant decrease in hormone sensitive lipase activity.
  • Agonists that down- regulate hormone sensitive lipase activity leading to a decrease in plasma free fatty acid levels are likely to have therapeutic value.
  • the consequence of decreasing plasma free fatty acids is twofold. First, it will ultimately lower LDL-cholesterol and raise HDL-cholesterol levels, independent risk factors, thereby reducing the risk of mortality due to cardiovascular incidence subsequent to atheroma formation. Second, it will provide an increase in insulin sensitivity in individuals with insulin resistance or type 2 diabetes.
  • Agonists of antilipolytic GPCRs having limited tissue distribution beyond adipose may be especially valuable in view of the diminished opportunity for potentially undesirable side-effects.
  • HDL is a "protective" lipoprotein [Gloria Lena Vega and Scott Grundy, Current Opinion in Lipidology, 7, 209-216 (1996)] and that increasing plasma levels of HDL may offer a direct protection against the development of atherosclerosis.
  • CHD coronary heart disease
  • HDL-C serum HDL-cholesterol
  • Atherosclerosis is the process of the accumulation of cholesterol within the arterial wall which results in the occlusion, or stenosis, of coronary and cerebral arterial vessels and subsequent myocardial infarction and stroke.
  • HDL Angiographic studies have shown that elevated levels of some HDL particles in humans appear to be correlated to a decreased number of sites of stenosis in the coronary arteries of humans (Miller et al., Br. Med. J., 282, 1741-1744 (1981)). There are several mechanisms by which HDL may protect against the progression of atherosclerosis. Studies in vitro have shown that HDL is capable of removing cholesterol from cells (Picardo et al., Arteriosclerosis, 6, 434-441 (1986)). Data of this nature suggest that one antiatherogenic property of HDL may lie in its ability to deplete tissue of excess free cholesterol and eventually lead to the delivery of this cholesterol to the liver (Glomset, J.
  • HDL Lipid Res., 9, 155- 167 (1968)). This has been supported by experiments showing efficient transfer of cholesterol from HDL to the liver (Glass et al., J. Biol. Chem., 258 7161-7167 (1983); McKinnon et al., J. Biol. Chem., 26, 2548-2552 (1986)).
  • HDL may serve as a reservoir in the circulation for apoproteins necessary for the rapid metabolism of triglyceride-rich lipoproteins (Grow and Fried, J. Biol. Chem., 253, 1834-1841 (1978); LagocM and Scanu, J. Biol. Chem., 255, 3701-3706 (1980); Schaefer et al., J. Lipid Res., 23, 1259-1273 (1982)).
  • Total Cholesterol/HDL-Cholesterol (i.e., TC/HDL) ratio represents a useful predictor as to the risk of an individual in developing a more serious condition, such as a HDL-related condition.
  • the classification of plasma lipid levels is shown in Table A:
  • the recommended Total Cholesterol/HDL-Cholesterol (i.e., TC/HDL) ratio indicates that a ratio of about 3.5 or less is ideal and a ratio of about 4.5 or greater is considered an increased "at risk.”
  • the value of determining the TC/HDL ratio is clearly evident in the circumstance where an individual presents with "normal" LDL and total cholesterol but possesses low HDL-cholesterol. Based on LDL and total cholesterol the individual may not qualify for treatment, however, factor in the HDL-cholesterol level then a more accurate risk assessment may be obtained. Thus, if the individual's level of HDL-cholesterol is such that the ratio is greater than about 4.5 then therapeutic intervention may be warranted.
  • a physician or care provider may determine the need of treatment based on a TC/HDL ratio; for example, a TC/HDL ratio of about 2.5 or greater, about 3.0 or greater, about 3.5 or greater, about 4.0 or greater, about 4.5 or greater, about 5.0 or greater, or a TC/HDL ratio of about 5.5 or greater.
  • agents that increase HDL levels or reduce total cholesterol/HDL ratios would be of great importance as antiatherosclerotic agents, and particularly useful in the treatment of coronary heart disease, ischemic cerebrovascular disease, peripheral vascular disease, dyslipoproteinimias and other HDL-related diseases.
  • the present invention is drawn to compounds which bind to and interact with the GPCR defined as RUP38 and uses thereof.
  • the term RTJP38 includes the human sequences found in GeneBank accession number D 10923.1, naturally-occurring allelic variants, mammalian orthologs, biologically active fragments and recombinant mutants thereof.
  • One aspect of the present invention encompasses certain carboxylic acid or ester derivatives as shown in Formula (I):
  • A is a Ci -3 alkylene optionally substituted with 1, 2, 3, or 4 substituents selected independently from the group consisting Of C 1-6 alkoxy, Q -6 alkyl, C 1-6 alkylsulfinyl, Ci -6 alkylsulfonyl, amino, Ci -6 alkylamino, C 2-6 dialkylamino, carbo-Ci -6 -alkoxy, carboxy, cyano, C 3-6 cycloalkyl, halogen, Ci -6 haloalkoxy, Ci -6 haloalkyl, C 1-6 haloalkylsulfinyl, C 1-6 haloalkylsulfonyl, Ci -6 haloalkylthio and hydroxyl;
  • B is a Ci -3 alkylene optionally substituted with 1, 2, 3, or 4 substituents selected independently from the group consisting of Ci -6 alkoxy, Ci -6 alkyl, Ci -6 alkylsulfinyl, Q -6 alkylsulfonyl, amino, Ci -6 alkylamino, C 2-6 dialkylamino, carbo-Ci -6 -alkoxy, carboxy, cyano, C 3-6 cycloalkyl, halogen, Ci -6 haloalkoxy, C 1-6 haloalkyl, Ci -6 haloalkylsulfinyl, C 1-6 haloalkylsulfonyl, C 1-6 haloalkylthio and hydroxyl, or B is a bond;
  • R 1 is H, aryl, heteroaryl, C 1-6 alkyl, C 3-7 cycloalkyl, C 2-6 alkenyl, or C 1-6 haloalkyl, wherein the Ri is optionally substituted with 1, 2, 3, 4, or 5 substituents selected independently from the group consisting of C 1-6 acyl, acyloxy, C 2-6 alkenyl, Ci -6 alkoxy, Ci -6 alkyl, C x-6 alkylcarboxamido, C 2-6 alkynyl, Ci -6 alkylsulfinyl, Q -6 alkylsulfonyl, Ci -6 alkylthio, Ci -6 alkylureyl, amino, Ci -6 alkylamino, C 2-6 dialkylamino, carbo-Ci -6 -alkoxy, carboxy, cyano, C 3-6 cycloalkyl, C 2-6 dialkylcarboxamido, halogen, Ci -6 haloalkoxy, Ci
  • R 2 is H, aryl, heteroaryl, C 1-6 alkyl, C 3-7 cycloalkyl, C 2-6 alkenyl, or Ci -6 haloalkyl, wherein the R 2 is optionally substituted with 1, 2, 3, 4, or 5 substituents selected independently from the group consisting of Q -6 acyl, acyloxy, C 2-6 alkenyl, C 1-6 alkoxy, Ci -6 alkyl, C ]-6 alkylcarboxamido, C 2-6 alkynyl, Ci -6 alkylsulf ⁇ nyl, Ci -6 alkylsulfonyl, Cj -6 alkylthio, Ci -6 alkylureyl, amino, Ci -6 alkylamino, C 2-6 dialkylamino, carbo-Ci -6 -alkoxy, carboxy, cyano, C3 -6 cycloalkyl, C 2-6 dialkylcarboxamido, halogen, Ci -6 haloal
  • R 3 is selected from the group consisting of H, Ci -6 acyl, acyloxy, C 2-6 alkenyl, Ci -6 alkoxy, Ci -6 alkyl, Q -6 alkylcarboxamido, C 2-6 alkynyl, Ci -6 alkylsulfinyl, Ci -6 alkylsulfonyl, Ci -6 alkylthio, Ci -6 alkylureyl, amino, Ci -6 alkylamino, C 2-6 dialkylamino, carbo-Q -6 -alkoxy, carboxy, cyano, C 3-6 cycloalkyl, C 2-6 dialkylcarboxamido, halogen, C !-6 haloalkoxy, Ci -6 haloalkyl, Q -6 haloalkylsulfinyl, Ci -6 haloalkylsulfonyl, Q -6 haloalkylthio, hydroxyl, nitro and thiol;
  • R 3 is selected from the group consisting of H, Q -6 acyl, acyloxy, C 2-6 alkenyl, Ci -6 alkoxy, Ci -6 alkyl, Ci -6 alkylcarboxamido, C 2-6 alkynyl, Q -6 alkylsulfinyl, Ci -6 alkylsulfonyl, Q -6 alkylthio, Ci -6 alkylureyl, amino, Ci -6 alkylamino, C 3-6 dialkylamino, carbo-Q.
  • Some embodiments of the present invention include a pharmaceutical composition comprising at least one compound of the present invention and a pharmaceutically acceptable carrier.
  • Some embodiments of the present invention include methods of modulating a RUP38 receptor comprising contacting the receptor with an effective amount of a compound of the present invention, wherein "compound(s) of the present invention” is intended to include Formula (I) and subgenera thereof.
  • the compound is an agonist of the RUP38 receptor.
  • Some embodiments of the present invention include methods of modulating a RUP38 receptor in an individual comprising contacting the receptor with a compound of the present invention.
  • the compound is an agonist of the RUP38 receptor.
  • the modulation of the RTJP38 receptor treats a metabolic-related disorder.
  • Some embodiments of the present invention include methods of modulating RTJP38 receptor function in a cell, tissue or individual comprising contacting the cell, tissue or individual with an effective amount of a compound of the present invention or a pharmaceutical composition thereof as described herein.
  • the RUP38 receptor function is associated with a metabolic-related disorder.
  • Some embodiments of the present invention include methods of raising HDL cholesterol levels in an individual comprising administering to the individual a therapeutically effective amount of a compound of of the present invention.
  • Some embodiments of the present invention include the use of compounds of the present invention for production of medicaments for use in the treatment of a metabolic disorder.
  • One aspect of the present invention pertains to compounds of the present invention as described herein, for use in methods of treatment of the human or animal body by therapy.
  • the metabolic-related disorder is selected from the group consisting of dyslipidemia, atherosclerosis, coronary heart disease, insulin resistance, obesity, impaired glucose tolerance, atheromatous disease, hypertension, stroke, Syndrome X, heart disease, type 2 diabetes, hypo-HDL related atherosclerotic risk, ischemic cerebrovascular disease, peripheral vascular disease, stroke, and myocardial infarction.
  • the metabolic-related disorder is selected from the group consisting of dyslipidemia, atherosclerosis, coronary heart disease, insulin resistance and type 2 diabetes.
  • the metabolic-related disorder is dyslipidemia.
  • One aspect of the present invention pertains to compounds of the present invention as described herein, for use in a method of raising HDL cholesterol levels of the human or animal body by therapy.
  • the individual in certain methods is a mammal. In some embodiments, the mammal is a human.
  • Some embodiments of the present invention include methods of producing pharmaceutical compositions comprising admixing at least one compound according to any of the compound embodiments disclosed herein and a pharmaceutically acceptable carrier.
  • the invention pertains to methods for alleviation of a symptom of any of the diseases, conditions or disorders mentioned herein.
  • Applicant reserves the right to exclude any one or more of the compounds from any of the embodiments of the invention.
  • Applicant additionally reserves the right to exclude any disease, condition or disorder from any of the embodiments of the invention.
  • Figure 1 shows histograms representing relative expression levels of RUP38 detected in different human tissues via DNA microarray.
  • the horizontal axis displays the different tissues, which are identified in Example 2(B) herein.
  • the vertical axis indicates level of expression of RUP38.
  • RUP38 (the signal toward the left of each of the histograms corresponding to primary adipocytes is identified by a vertical arrow above the bar, for ease of reference).
  • Figure 2 is a photograph of an ethidium bromide stained gel illustrating the relative expression of RUP25 (a GPCR related to RUP38, GeneBank accession number AB065876.1) and RUP38 as detected by RT-PCR using cDNA derived from a number of human tissues as template. Note the tissue expression for RUP38 in adipocycte, spleen and lung; also notice by comparison with RUP25, RUP38 has limited tissue distribution beyond adipose. The controls are shown in the far right three lanes.
  • Figure 3 depicts melanophores transfected with DNA plasmids expressing RUP38 without treatment. These cells are pigment-aggregated because RUP38 is a Gi-coupled receptor having a high basal level of activity, and therefore driving the aggregation to a measurable level in the absence of a ligand.
  • Isopropyl-lH-benzotriazole-5-carboxylic acid has no effect on C ⁇ O cells expressing RUP25 or
  • RUP19 an orphan GPCR also related to RUP38, GeneBank accession number AF345568.1.
  • NT indicates not tested.
  • AGONISTS shall mean moieties that interact and activate the receptor, such as the RUP38 receptor and initiates a physiological or pharmacological response characteristic of that receptor. For example, when moieties activate the intracellular response upon binding to the receptor, or enhance GTP binding to membranes.
  • ANGINA PECTORIS is intended herein to encompass the paroxysmal thoracic pain caused generally and most often by myocardial anoxia as a result of atherosclerosis.
  • ANTAGONISTS is intended to mean moieties that competitively bind to the receptor at the same site as agonists (for example, the endogenous ligand), but which do not activate the intracellular response initiated by the active form of the receptor, and can thereby inhibit the intracellular responses by agonists or partial agonists. Antagonists do not diminish the baseline intracellular response in the absence of an agonist or partial agonist.
  • ATHEROSCLEROSIS is intended herein to encompass disorders of the arteries that result in the progressive accumulation of smooth muscle cells and lipids within the intima.
  • Atherosclerosis is a disorder that is generally characterized by yellowish plaques of cholesterol, others lipids and cellular debris in the inner layers of the walls of the arteries. Over time the atherosclerotic plaque increases thus narrowing the lumen and reducing the blood flow to organs supplied by the artery. The plaque eventually creates a risk for thrombosis and is one of the major causes of coronary heart disease, angina pectoris, myocardial infarction and other disorders, such as, congestive heart failure, ischemic heart disease, peripheral vascular disease, stroke and the like.
  • Ci -6 acyl denotes an alkyl radical attached to a carbonyl wherein the definition of alkyl has the same definition as described herein; some examples include formyl, acetyl, propionyl, butanoyl, iso-butanoyl, and the like.
  • Ci -6 acyloxy denotes an acyl radical attached to an oxygen atom wherein acyl has the same definition has described herein; some examples include acetyloxy, propionyloxy, butanoyloxy, iso-butanoyloxy and the like.
  • C 2-6 alkenyl denotes a radical containing 2 to 4 carbons wherein at least one carbon-carbon double bond is present, some embodiments have 3 carbons, and some embodiments have 2 carbons.
  • E and Z isomers and mixtures of E and Z isomers are embraced by the term "alkenyl.” Examples of an alkenyl include vinyl, allyl, 2-butenyl, 3- butenyl, and the like.
  • Ci -6 alkoxy denotes a radical alkyl, as defined herein, attached directly to an oxygen atom. Examples include methoxy, ethoxy, n-propoxy, iso-propoxy, n- butoxy, t-butoxy, iso-butoxy and the like.
  • C 1-8 alkyl denotes a straight or branched carbon radical containing 1 to 8 carbons, 1 to 6 carbons, or 1 to 4 carbons respectively, some embodiments are 1 to 3 carbons, and some embodiments are 1 or 2 carbons.
  • alkyl examples include, but not limited to, methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, t-butyl, sec- butyl, n-pentyl, iso-pentyl, sec-pentyl, neo-pentyl, pent-3-yl, 2-methyl-but-l-yl, 1,2-dimethyl- prop-1-yl, n-hexyl, iso-hexyl, sec-hexyl, neo-hexyl, l-ethyl-2-methyl-prop-l-yl, 1,2,2-trimethyl- prop-1-yl, 1,1,2-trimethyl-prop-l-yl, 1-ethyl-l-methyl-prop-l-yl, 1,1-dimethyl-but-l-yl, 1,2- dimethyl-but-1-yl, 2,3-dimethyl-but-l-yl
  • Ci -6 alkylcarboxamido denotes a single alkyl group attached to an amide, wherein alkyl has the same definition as found herein.
  • the Ci -6 alkylcarboxamido may be represented by the following:
  • C 1-6 alkylsulf ⁇ nyl denotes an alkyl radical attached to a sulfoxide radical of the formula: -S(O)- wherein the alkyl radical has the same definition as described herein. Examples include methylsulfinyl, ethylsulfinyl and the like.
  • Cn alkylene refers to a divalent straight carbon group, such as, -CH 2 -, -CH 2 CH 2 -, -CH 2 CH 2 CH 2 -.
  • C x-6 alkylsulfonamide refers to the groups
  • Ci -6 alkylsulfonyl denotes an alkyl radical attached to a sulfone radical of the formula: -S(O) 2 - wherein the alkyl radical has the same definition as described herein. Examples include methylsulfonyl, ethylsulfonyl and the like.
  • Ci -6 alkylthio denotes an alkyl radical attached to a sulfide of the formula: -S- wherein the alkyl radical has the same definition as described herein. Examples include methylsulfanyl (i.e., CH 3 S-), ethylsulfanyl, isopropylsulfanyl and the like.
  • C 1-6 alkylamino denotes one alkyl radical attached to an amino radical wherein the alkyl radical has the same meaning as described herein. Some examples include methylamino, ethylamino, propylamino and the like.
  • C 1-6 alkylureyl denotes the group of the formula: -NC(O)N- wherein one are both of the nitrogens are substituted with the same or different Ci -6 alkyl group wherein alkyl has the same definition as described herein.
  • alkylureyl examples include, CH 3 NHC(O)NH-, NH 2 C(O)NCH 3 -, (CHa) 2 N(O)NH-, (CH 3 ) 2 N(O)NH-, (CH 3 ) 2 N(O)NCH 3 -, CH 3 CH 2 NHC(O)NH-, CH 3 CH 2 NHC(O)NCH 3 -, and the like.
  • C 2-6 alkynyl denotes a radical containing 2 to 6 carbons and at least one carbon-carbon triple bond, some embodiments are 2 to 4 carbons, some embodiments are 2 to 3 carbons, and some embodiments have 2 carbons.
  • alkynyl examples include ethynyl, ethynyl, 1-propynyl, 2-propynyl, 1-butynyl, 2-butynyl, 3-butynyl, 1-pentynyl, 2-pentynyl, 3- pentynyl, 4-pentynyl, 1-hexynyl, 2-hexynyl, 3-hexynyl, 4-hexynyl, 5-hexynyl and the like.
  • alkynyl includes di- and tri-ynes.
  • amino refers to -NH 2 .
  • aryl denotes an aromatic ring radical containing 6 to 10 ring carbons.
  • Examples include phenyl and naphthyl.
  • carbo-Ci -6 -alkoxy refers to an alkyl ester of a carboxylic acid, wherein the alkyl group is Ci -4 .
  • alkyl group is Ci -4 .
  • Examples include carbomethoxy, carboethoxy, carboisopropoxy and the like.
  • Carboxamide refers to the group -CONH 2 .
  • Carboxy refers to the group -CO 2 H.
  • C 3-7 cycloalkenyl denotes a non-aromatic ring radical containing 3 to 7 ring carbons and at least one double bond; some embodiments contain 5 to 7 carbons; some embodiments contain 3 to 6 carbons; some embodiments contain 3 to 5 carbons; some embodiments contain 3 to 4 carbons. Examples include cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclopentenyl, cyclohexenyl, and the like.
  • C 3-7 cycloalkyl or "C 3-6 cycloalkyl” denotes a non aromatic ring radical containing 3 to 7 carbons or 3 to 6 carbons respectively; some embodiments contain 5 to 7 carbons; some embodiments contain 3 to 5 carbons; some embodiments contain 3 to 4 carbons. Examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclopenyl, cyclohexyl, cycloheptyl and the like.
  • cyano denotes the group -CN.
  • C 2-6 dialkylamino denotes an amino substituted with two of the same or different alkyl radicals wherein alkyl radical has the same definition as described herein and the sum of the number of carbons between the two alkyl groups is 2 to 6. Some embodiments contain 3 to 6 carbons, wherein the sum of the number of carbons between the two alkyl groups is 3 to 6 (i.e., in some embodiments, C 3-6 dialkylamino include dialkylamino groups other than dimethylamino). In general, examples include dimethylamino, methylethylamino, diethylamino, diethylamino, and the like.
  • C 2-6 dialkylcarboxamido denotes two alkyl radicals, that are the same or different, attached to an amide group, wherein alkyl has the same definition as described herein.
  • a C 2-6 dialkylcarboxamido maybe represented by the following groups:
  • dialkylcarboxamide examples include N,N-dimethylcarboxamide, N-methyl-N-ethylcarboxamide, N- methyl-N-pentylcarboxamide and the like.
  • Ci -6 haloalkoxy denotes a haloalkyl, as defined herein, that is directly attached to an oxygen to form a difluoromethoxy, trifluoromethoxy, 2,2,2-trifluoroethoxy, pentafluoroethoxy and the like.
  • C 1-6 haloalkyl denotes an alkyl group, defined herein, wherein the alkyl is substituted with at least one halogen up to fully substituted represented by the formula C n L 2n+I , wherein L is a halogen; when more than one halogen is present then they may be the same or different and selected from F, Cl, Br or I. Examples include fluoromethyl, difluoromethyl, trifluoromethyl, chlorodifluoromethyl, 2,2,2-trifluoroethyl, pentafluoroethyl and the like.
  • Cj -6 haloalkylsulfinyl denotes a haloalkyl radical attached to a sulfoxide of the formula: -S(O)- wherein the alkyl radical has the same definition as described herein.
  • Examples include trifluoromethylsulfinyl, 2,2,2-trifluoroethylsulfinyl, 2,2-difluoroethylsulfinyl and the like.
  • Ci -6 haloalkylsulfonyl denotes a haloalkyl attached to a sulfone of the formula: -S(O) 2 - wherein haloalkyl has the same definition as described herein. Examples include trifluoromethylsulfonyl, 2,2,2-trifluoroethylsulfonyl, 2,2-difluoroethylsulfonyl and the like.
  • Ci -6 haloalkylthio denotes an alkylthio radical substituted with one or more halogens. Examples include trifluoromethylthio, 1,1-difluoroethylthio, 2,2,2-trifluoroethylthio and the like.
  • heteroaryl denotes an aromatic ring system that may be a single ring, two fused rings or three fused rings containing carbons and at least one ring heteroatom selected from O, S and N.
  • heteroaryl groups include, but not limited to, pyridyl, pyrazinyl, pyridazinyl, pyrimidinyl, triazinyl, quinoline, benzoxazole, benzothiazole, lH-benzimidazole, isoquinoline, quinazoline, quinoxaline, pyridinone and the like.
  • C 5-7 heterocycloalkyl denotes a non-aromatic carbon ring (i.e., cycloalkyl or cycloalkenyl as defined herein) wherein one, two or three ring carbons are replaced by one, two or three heteroatoms, such as, O, S, S(O), S(O) 2 , NH, and N-Ci. 4 -alkyl.
  • heterocycloalkyl include piperidinyl, morpholinyl, piperzinyl, pyrrolidinyl, and the like.
  • hydroxyl refers to the group -OH.
  • nitro denotes the group -NO 2 .
  • thiol denotes the group -SH.
  • COMPOSITION shall mean a material comprising at least two compounds or two components; for example, and not limitation, a Pharmaceutical Composition is a Composition.
  • CONGESTIVE HEART FAILURE shall refer to a disorder in which the heart loses its ability to pump blood efficiently. Congestive heart failure becomes more prevalent with advancing age. Ischemic heart disease is the most common cause of congestive heart failure, accounting for 60-70% of all cases. An increased venous pressure greater than 12 mtnHg is one of the major Framingham criteria for congestive heart failure, as is a reduction in cardiac output equivalent to a circulation time greater than 25 seconds.
  • CORONARY HEART DISEASE is intended herein to encompass disorders comprising a narrowing of the small blood vessels that supply blood and oxygen to the heart.
  • CORONARY HEART DISEASE usually results from the build up of fatty material and plaque. As the coronary arteries narrow, the flow of blood to the heart can slow or stop.
  • CORONARY HEART DISEASE can cause chest pain (stable angina), shortness of breath, heart attack, or other symptoms.
  • CONTACT or CONTACTING shall mean bringing the indicated moieties together, whether in an in vitro system or an in vivo system.
  • "contacting" a RUP38 receptor with a compound of the invention includes the administration of a compound of the present invention to an individual, preferably a human, having a RUP38 receptor, as well as, for example, introducing a compound of the invention into a sample containing a cellular or more purified preparation containing a RUP38 receptor.
  • HYPO-HDL RELATED ATHEROSCLEROTIC RISK refers to a HDL-related disorder that leads to or predisposes an individual to the risk of developing atherosclerosis (supra) by the presence of low levels of HDL cholesterol. Although low levels of HDL may depend on a variety of health factors, Table B can be used as a general guideline by physicians or caregivers in evaluating the individual.
  • the levels in Table B are useful as a general guideline for evaluational purposes, levels outside these values may depend on a variety of health factors, all of which would be recognized by a physician or caregiver.
  • IN NEED OF SUCH TREATMENT refers 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 is ill, or will be ill, as the result of a disease, condition or disorder that is treatable by the compounds of the invention.
  • treatment also refers in the alternative to "prophylaxis.” Therefore, in general, “in need of treatment” refers to the judgment of the caregiver that the individual is already ill, accordingly, the compounds of the present invention are used to alleviate, inhibit or ameliorate the disease, condition or disorder. Furthermore, the phrase also refers, in the alternative, to the judgment made by the caregiver that the individual will become ill. In this context, the compounds of the invention are used in a protective or preventive manner.
  • INDIVIDUAL refers to any animal, vertebrate, mammal and human; for example, mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates, and humans. In some embodiments, individual refers to human.
  • ISCHEMIC HEART DISEASE shall refer to a disorder caused by lack of oxygen to the tissues of the heart, in which muscles of the heart are affected and the heart cannot pump properly. Ischemic heart disease is the most common cardiomyopathy in the United States.
  • MYOCARDIAL INFARCTION shall refer to the damage or death of an area of heart muscle because of an inadequate supply of oxygen to that area.
  • Myocardial infarctions are often caused by a clot that blocks one of the coronary arteries (the blood vessels that bring blood and oxygen to heart muscle) or by a narrowing of an artery.
  • the clot or narrowing of an artery prevents blood and oxygen from reaching that area of the heart, leading to the death of heart cells in that area.
  • PERIPHERAL VASCULAR DISEASE refers to disorders affecting the blood vessels outside the heart and brain, such as, arteries, veins, and lymphatics of the extremities.
  • Peripheral vascular disease includes, but not limited to, peripheral artery disease (PAD), and the like.
  • PHARMACEUTICAL COMPOSITION shall mean a composition comprising at least one active ingredient, whereby the composition is amenable to investigation for a specified, efficacious outcome in a mammal (for example, and not limitation, a human).
  • a mammal for example, and not limitation, a human.
  • STROKE occurs when the arterial blood flow leading to or in the brain becomes blocked or ruptures and a result of this sudden diminution or loss of blood, the individual's neurological function is decreased. Blood carries oxygen and nutrients to the neurons (nerve cells) in the brain, so when the blood flow stops, the cells begin to die. As a result, the functions of the body controlled by the nerve cells can lose their ability to function.
  • Secondary brain damage resulting from the stroke can include cerebral cell destruction, or lesions, in the area surrounding the ischemic injury, in the case of focal ischemia, and also in areas of selective vulnerability in lesions, such as the hippocampus or basal ganglia, in the case of global ischemia. The secondary damage resulting from a stroke can often be manifested by functional impairment, such as but not limited to, loss of physical movement, loss of speech, loss of short-term and/or long-term memory.
  • 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, which includes one or more of the following:
  • Preventing the disease for example, preventing a disease, condition or disorder in an individual that may be predisposed to the disease, condition or disorder but does not yet experience or display the pathology or symptomatology of the disease,
  • Inhibiting the disease for example, inhibiting a disease, condition or disorder in an individual that is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., arresting further development of the pathology and/or symptomatology), and
  • Ameliorating the disease for example, ameliorating a disease, condition or disorder in an individual that is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., reversing the pathology and/or symptomatology).
  • One aspect of the present invention encompasses certain pyrazole carboxylic acid or ester derivatives as shown in Formula QL):
  • Compounds of the invention also include tautomeric forms, such as keto-enol tautomers, pyrazole tautomeric forms, and the like.
  • tautomeric forms such as keto-enol tautomers, pyrazole tautomeric forms, 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.
  • substituted indicates that at least one hydrogen atom of the chemical group is replaced by a non-hydrogen substituent or group.
  • a chemical group herein 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 1 or 2 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. It is understood that when a group is optionally substituted with more than one group then those groups can be same or different.
  • Compounds of the invention can also include all isotopes of atoms occurring in the intermediates or final compounds.
  • Isotopes include those atoms having the same atomic number but different mass numbers.
  • isotopes of hydrogen include deuterium and tritium.
  • Ri is H.
  • Ri is H and A is C] -3 alkylene.
  • R 1 and A together are selected from the group consisting of CH 3 , CH 2 CH 3 , and CH 2 CH 2 CH 3 .
  • Ri is aryl, heteroaryl, Ci -6 alkyl, C 3-7 cycloalkyl, C 2-6 alkenyl, or Ci- 6 haloalkyl, wherein the Ri is optionally substituted with 1, 2, 3, 4, or 5 substituents. In some embodiments, Ri is optionally substituted with 1, 2, 3, or 4 substituents. In some embodiments, Ri is optionally substituted with 1, 2, or 3 substituents. In some embodiments, Ri is optionally substituted with 1, or 2 substituents.
  • Ri is aryl optionally substituted with 1, 2, 3, 4, or 5 substituents selected independently from the group consisting of Ci -6 acyl, acyloxy, C 2-6 alkenyl, Ci -6 alkoxy, Ci -6 alkyl, Ci -6 alkylcarboxamido, C 2-6 alkynyl, Ci -6 alkylsulfmyl, Ci -6 alkylsulfonyl, Ci -6 alkylthio, Q -6 alkylureyl, amino, Ci -6 alkylamino, C 2-6 dialkylamino, carbo-Ci -6 -alkoxy, carboxy, cyano, C 3-6 cycloalkyl, C 2-6 dialkylcarboxamido, halogen, Ci -6 haloalkoxy, Ci -6 haloalkyl, Ci -6 haloalkylsulfmyl, C 1-6 haloalkylsulfonyl, Ci -6 haloal
  • Ri is aryl optionally substituted with 1, 2, 3, 4, or 5 substituents selected independently from the group consisting OfCi -6 acyl, C 2-6 alkenyl, Ci -6 alkoxy, Ci -6 alkyl, Ci -6 alkylcarboxamido, C 2-6 alkynyl, Ci -6 alkylsulfmyl, Ci -6 alkylsulfonyl, C 1-6 alkylthio, Ci -6 alkylureyl, Ci -6 alkylamino, C 2-6 dialkylamino, carbo-Ci -6 -alkoxy, carboxy, cyano, C 3-6 cycloalkyl, C 2-6 dialkylcarboxamido, halogen, Ci -6 haloalkoxy, Ci -6 haloalkyl, Ci -6 haloalkylsulfmyl, Ci -6 haloalkylsulfonyl, hydroxyl, and nitro.
  • substituents selected independently from the
  • R 1 is aryl optionally substituted with 1, 2, 3, 4 or 5 substituents selected independently from the group consisting of Q -6 acyl, C 1-6 alkoxy, C 1-6 alkyl, C 1-6 alkylamino, C 2-6 dialkylamino, carbo-Q -6 -alkoxy, carboxy, cyano, halogen, C 1-6 haloalkoxy, C 1-6 haloalkyl, hydroxyl and nitro.
  • R 1 is aryl optionally substituted with 1, 2, 3, 4 or 5 substituents selected independently from the group consisting of C 1-6 alkoxy, C 1-6 alkyl, carboxy, cyano, halogen, C 1-6 haloalkoxy, and Q -6 haloalkyl.
  • R 1 is aryl optionally substituted with halogen.
  • Ri is aryl optionally substituted with F.
  • Ri is phenyl
  • Ri is phenyl and compounds can be represented by Formula (Ib) as shown below:
  • R 5 -R 9 are each independently selected from the group consisting of H, Ci -6 acyl, acyloxy, C 2-6 alkenyl, Ci -6 alkoxy, Ci -6 alkyl, Ci -6 alkylcarboxamido, C 2 .
  • R 1 is aryl optionally substituted with two adjacent substituents together with the aryl form a C 5-7 cycloalkyl optionally substituted with halogen.
  • Ri is phenyl optionally substituted with two adjacent substituents together with the aryl to form a C 5-7 cycloalkyl optionally substituted with halogen.
  • compounds of the present invention can be represented by Formula (Id) as shown below:
  • R 1 is aryl optionally substituted with two adjacent substituents together with the aryl to form a C 5-7 heterocycloalkyl optionally substituted with halogen.
  • compounds of the present invention can be represented by Formula (Id) wherein 1, or 2 non-aromatic ring carbons are independently replaced with a heteroatom selected from the group consisting of O, S, S(O), S(O) 2 , NH, and N-C 1-4 -alkyl.
  • R 1 is a benzo[l,3]dioxolyl.
  • R 1 is a benzo[l,3]dioxol-4-yl.
  • R 1 is heteroaryl optionally substituted with 1, 2, 3, or 4 substituents selected independently from the group consisting of C 1-6 acyl, acyloxy, C 2-6 alkenyl, C 1-6 alkoxy, Ci -6 alkyl, Ci -6 alkylcarboxamido, C 2-6 alkynyl, C 1-6 alkylsulfmyl, C 1-6 alkylsulfonyl, Cj -6 alkylthio, C 1-6 alkylureyl, amino, Ci -6 alkylamino, C 2-6 dialkylamino, carbo-Ci.
  • substituents selected independently from the group consisting of C 1-6 acyl, acyloxy, C 2-6 alkenyl, C 1-6 alkoxy, Ci -6 alkyl, Ci -6 alkylcarboxamido, C 2-6 alkynyl, C 1-6 alkylsulfmyl, C 1-6 alkylsulfonyl, Cj -6 alkylthi
  • R is heteroaryl optionally substituted with 1, 2, 3, or 4 substituents selected independently from the group consisting of Ci -6 acyl, C 2-6 alkenyl, Q -6 alkoxy, Ci -6 alkyl, Ci -6 alkylcarboxamido, C 2-6 alkynyl, Ci -6 alkylsulfmyl, Ci -6 alkylsulfonyl, Ci -6 alkylthio, Ci -6 alkylureyl, Ci -6 alkylamino, C 2-6 dialkylamino, carbo-Ci.
  • substituents selected independently from the group consisting of Ci -6 acyl, C 2-6 alkenyl, Q -6 alkoxy, Ci -6 alkyl, Ci -6 alkylcarboxamido, C 2-6 alkynyl, Ci -6 alkylsulfmyl, Ci -6 alkylsulfonyl, Ci -6 alkylthio, Ci -6 alkylureyl, Ci -6
  • Ri is heteroaryl optionally substituted with 1, 2, 3, or 4 substituents selected independently from the group consisting of Ci -6 acyl, Ci -6 alkoxy, Ci -6 alkyl, Ci -6 alkylamino, C 2-6 dialkylamino, carbo-Q. 6 -alkoxy, carboxy, cyano, halogen, Ci -6 haloalkoxy, Ci -6 haloalkyl, hydroxyl and nitro.
  • Ri is heteroaryl optionally substituted with 1, 2, 3, or 4 substituents selected independently from the group consisting of Ci -6 alkoxy, Ci -6 alkyl, carboxy, cyano, halogen, Ci -6 haloalkoxy, and Ci -6 haloalkyl.
  • R] is heteroaryl optionally substituted with 1, 2, 3, or 4 substituents selected independently from the group consisting Of Q -6 alkyl, and halogen.
  • Ri is heteroaryl optionally substituted with 1, 2, 3, or 4 substituents selected independently from the group consisting of CH 3 , F, Cl, and Br.
  • Ri is a 5-member or 6-member heteroaryl. In some embodiments, Ri is a 5-member heteroaryl.
  • heteroaryl is selected from the group in Table 2 as shown below:
  • a imidazolyl ring can be bonded at one of the ring nitrogens (i.e., imidazol-1-yl group) or at one of the ring carbons (i.e., imidazol-2-yl, imidazol-4-yl or imiadazol-5-yl group) as illustrated below:
  • Ri is a 5-member heteroaryl selected from the group consisting of a furanyl, an isoxazolyl, an oxazolyl, an [l,2,4]-oxadiazolyl, an [l,3,4]-oxadiazolyl, a thienyl, an isothiazolyl, a thiazolyl, a [l,2,4]-thiadiazolyl, a [1,3,4-thiadiazolyl, a lH-pyrrolyl, a IH- pyrazolyl, an lH-imidazolyl, a IH-[1, 2,4]-triazolyl, a lH-[l,2,4]-triazolyl, and a lH-tetrazolyl.
  • Rj is a 5-member heteroaryl selected from the group consisting of thien-2-yl, thien-3-yl, furan-2-yl and furan-3-yl. It is understood that due to variations in chemical nomenclature that a group may have more than one chemical name to represent the group, for example, thien-2-yl is equivalent to thiophen-2-yl. Other examples exist in the art and those of ordinary skill are credited with the ability to recognize that they are present.
  • the heteroaryl is a 6-member heteroaryl.
  • heteroaryl is selected from the group in Table 3 as shown below:
  • heteroaryl group is bonded at any ring carbon.
  • Rj is a 6-member heteroaryl selected from the group consisting of a pyridyl, a pyrazine, and a pyrimidinyl.
  • Ri is a 6-member selected from the group consisting of pyridin-2- yl, pyridm-3-yl, and pyridin-4-yl.
  • Ri is alkyl optionally substituted with 1, 2, 3, 4 or 5 substituents selected independently from the group consisting Of Ci -6 acyl, acyloxy, C 2-6 alkenyl, Ci -6 alkoxy, Ci -6 alkyl, Ci -6 alkylcarboxamido, C 2-6 alkynyl, Ci -6 alkylsulfinyl, Ci -6 alkylsulfonyl, Ci -6 alkylthio, Ci -6 alkylureyl, amino, Ci -6 alkylamino, C 2-6 dialkylamino, carbo-Ci -6 -alkoxy, carboxy, cyano, C 3-6 cycloalkyl, C 2-6 dialkylcarboxamido, halogen, Ci -6 haloalkoxy, Ci -6 haloalkyl, Ci -6 haloalkylsulfinyl, Ci -6 haloalkylsulfonyl, Ci -6 hal
  • Ri is Ci -6 alkyl optionally substituted with 1, 2, 3, 4 or 5 substituents selected independently from the group consisting OfCi -6 acyl, C 2-6 alkenyl, C ]-6 alkoxy, Q -6 alkyl, Cj -6 alkylcarboxamido, C 2-6 alkynyl, Ci -6 alkylsulf ⁇ nyl, Ci -6 alkylsulfonyl, C ]-6 alkylthio, Ci -6 alkylureyl, C] -6 alkylamino, C 2-6 dialkylamino, carbo-Ci. 6 -alkoxy, carboxy, cyano,
  • Ri is Ci -6 alkyl optionally substituted with 1, 2, 3, 4 or 5 substituents selected independently from the group consisting of Ci -6 acyl, Ci -6 alkoxy, Ci -6 alkyl,
  • Ri is Ci -6 alkyl optionally substituted with 1, 2, 3, 4 or 5 substituents selected independently from the group consisting of Q -6 alkoxy, Ci -6 alkyl, carboxy, cyano, halogen, Ci -6 haloalkoxy, and Ci -6 haloalkyl.
  • R] is C 3-7 cycloalkyl optionally substituted with 1, 2, 3, 4 or 5 substituents selected independently from the group consisting Of Ci -6 alkoxy, Ci -6 alkyl, carboxy, cyano, halogen, Ci -6 haloalkoxy, and Ci -6 haloalkyl.
  • Ri is C 2-6 alkenyl optionally substituted with 1, 2, 3, 4 or 5 substituents selected independently from the group consisting of Ci -6 acyl, acyloxy, C 2-6 alkenyl,
  • Ri is C 2-6 alkenyl optionally substituted with 1, 2, 3, 4 or 5 substituents selected independently from the group consisting of C 1-6 alkoxy, Q -6 alkyl, carboxy, cyano, halogen, Ci -6 haloalkoxy, and C x-6 haloalkyl.
  • Ri is Ci -6 haloalkyl optionally substituted with 1, 2, 3, 4 or 5 substituents selected independently from the group consisting of Ci -6 acyl, acyloxy, C 2-6 alkenyl,
  • Ri is Ci -6 haloalkyl optionally substituted with 1, 2, 3, 4 or 5 substituents selected independently from the group consisting Of Ci -6 alkoxy, Q -6 alkyl, carboxy, cyano, halogen, and Ci -6 haloalkoxy.
  • Ra is H.
  • R 2 is H and A is Ci -3 alkylene.
  • R 2 and A together are selected from the group consisting of CH 3 , CH 2 CH 3 , CH 2 CH 2 CH 3 .
  • R 2 is H and A is a bond.
  • compounds of the present invention can be presented by Formula (II):
  • R 2 is aryl, heteroaryl, Ci -6 alkyl, C 3-7 cycloalkyl, C 2-6 alkenyl, or
  • Ci -6 haloalkyl wherein the R 2 is optionally substituted with 1, 2, 3, 4, or 5 substituents. In some embodiments, R 2 is optionally substituted with 1, 2, 3, or 4 substituents. In some embodiments,
  • R 2 is optionally substituted with 1, 2, or 3 substituents. In some embodiments, R 2 is optionally substituted with 1, or 2 substituents.
  • R 2 is aryl optionally substituted with 1, 2, 3, 4, or 5 substituents selected independently from the group consisting Of Ci -6 acyl, acyloxy, C 2-6 alkenyl, Ci -6 alkoxy, Ci -6 alkyl, Ci -6 alkylcarboxamido, C 2-6 alkynyl, Ci -6 alkylsulfinyl, Ci -6 alkylsulfonyl, Ci -6 alkylthio, Ci -6 alkylureyl, amino, Ci -6 alkylamino, C 2-6 dialkylamino, carbo-Ci -6 -alkoxy, carboxy, cyano, C 3-6 cycloalkyl, C 2-6 dialkylcarboxamido, halogen, Ci -6 haloalkoxy, Ci -6 haloalkyl, Ci -6 haloalkylsulfinyl, Ci -6 haloalkylsulfonyl, Q -6
  • R 2 is aryl optionally substituted with 1, 2, 3, 4, or 5 substituents selected independently from the group consisting of Ci -6 acyl, C 2-6 alkenyl, Ci -6 alkoxy, Q -6 alkyl,
  • R 2 is aryl optionally substituted with 1, 2, 3, 4 or 5 substituents selected independently from the group consisting of Q -6 acyl, Ci -6 alkoxy, Q -6 alkyl, Ci -6 alkylamino, C 2-6 dialkylamino, carbo-Q -6 -alkoxy, carboxy, cyano, halogen, Ci -6 haloalkoxy, Q. ⁇ haloalkyl, hydroxyl and nitro.
  • R 2 is aryl optionally substituted with 1, 2, 3, 4 or 5 substituents selected independently from the group consisting of Q -6 alkoxy, Ci -6 alkyl, carboxy, cyano, halogen, Q -6 haloalkoxy, and Ci -6 haloalkyl.
  • R 2 is aryl optionally substituted with halogen. In some embodiments, R 2 is aryl optionally substituted with F.
  • R 2 is phenyl. In some embodiments, R 2 is phenyl and compounds can be represented by Formula (Ih) as shown below:
  • R 1 0-R 14 are each independently selected from the group consisting of H, Ci -6 acyl, acyloxy, C 2 -6 alkenyl, Ci -6 alkoxy, Ci -6 alkyl, Ci -6 alkylcarboxamido, C 2-6 alkynyl, Ci -6 alkylsulfmyl, Ci -6 alkylsulfonyl, Ci -6 alkylthio, Ci -6 alkylureyl, amino, C 1-6 alkylamino, C 2-6 dialkylamino, carbo-Ci -6 -alkoxy, carboxy, cyano, C 3-6 cycloalkyl, C 2-6 dialkylcarboxamido, halogen, Ci -6 haloalkoxy, Ci -6 haloalkyl, Ci -6 haloalkylsulfmyl, C 1-6 haloalkylsulf
  • R 2 is phenyl optionally substituted with two adjacent substituents together with the aryl to form a C 5-7 cycloalkyl optionally substituted with halogen.
  • compounds of the present invention can be represented by Formula (Ij) as shown below:
  • R 2 is aryl optionally substituted with two adjacent substituents together with the aryl to form a C 5-7 heterocycloalkyl optionally substituted with halogen, m
  • compounds of the present invention can be represented by Formula (Ij) wherein 1, or 2 non-aromatic ring carbons are independently replaced with a heteroatom selected from the group consisting of O, S, S(O), S(O) 2 , NH, and N-Ci- 4 -alkyl.
  • R 2 is a benzo[l,3]dioxolyl. In some embodiments, R 2 is a benzo[l,3]dioxol-4-yl.
  • R 2 is heteroaryl optionally substituted with 1, 2, 3, or 4 substituents selected independently from the group consisting of Ci -6 acyl, acyloxy, C 2-6 alkenyl, Ci- 6 alkoxy, C 1-6 alkyl, Ci -6 alkylcarboxamido, C 2-6 alkynyl, Ci -6 alkylsulfinyl, Ci -6 alkylsulfonyl, Ci -6 alkylthio, Ci -6 alkylureyl, amino, Ci -6 alkylamino, C 2-6 dialkylamino, carbo-Ci -6 -alkoxy, carboxy, cyano, C 3-6 cycloalkyl, C 2-6 dialkylcarboxamido, halogen, Ci -6 haloalkoxy, C 1-6 haloalkyl, Ci -6 haloalkylsulfinyl, Ci -6 haloalkylsulfonyl, Q -6 hal
  • R 2 is heteroaryl optionally substituted with 1, 2, 3, or 4 substituents selected independently from the group consisting of Ci -6 acyl, C 2-6 alkenyl, Q -6 alkoxy, Ci -6 alkyl, Ci -6 alkylcarboxamido, C 2-6 alkynyl, Ci -6 alkylsulfinyl, Ci -6 alkylsulfonyl, Ci -6 alkylthio, Ci -6 alkylureyl, C J-6 alkylamino, C 2-6 dialkylamino, carbo-Ci -6 -alkoxy, carboxy, cyano, C 3-6 cycloalkyl, C 2-6 dialkylcarboxamido, halogen, Ci -6 haloalkoxy, Ci -6 haloalkyl, Ci -6 haloalkylsulfinyl, Ci -6 haloalkylsulfonyl, hydroxyl, nitro and thiol.
  • R 2 is heteroaryl optionally substituted with 1, 2, 3, or 4 substituents selected independently from the group consisting of Ci -6 acyl, Ci -6 alkoxy, Ci -6 alkyl, Ci -6 alkylamino, C 2-6 dialkylamino, carbo-Ci -6 -alkoxy, carboxy, cyano, halogen, Ci -6 haloalkoxy, Ci -6 haloalkyl, hydroxyl and nitro.
  • R 2 is heteroaryl optionally substituted with 1, 2, 3, or 4 substituents selected independently from the group consisting of Ci -6 alkyl, and halogen. In some embodiments, R 2 is heteroaryl optionally substituted with 1, 2, 3, or 4 substituents selected independently from the group consisting of CH 3 , F, Cl, and Br.
  • the heteroaryl is a 5-member or 6-member heteroaryl, each as described herein, for example as shown in Tables 2 and 3.
  • R 2 is a 5-member heteroaryl selected from the group consisting of a furanyl, an isoxazolyl, an oxazolyl, an [l,2,4]-oxadiazolyl, an [l,3,4]-oxadiazolyl, a thienyl, an isothiazolyl, a thiazolyl, a [l,2,4]-thiadiazolyl, a [1,3,4-thiadiazolyl, a lH-pyrrolyl, a IH- pyrazolyl, an lH-imidazolyl, a lH-[l,2,4]-triazolyl, a IH-[1, 2,4]-triazolyl, and a lH-tetrazolyl.
  • R 2 is a 5-member heteroaryl selected from the group consisting of thien-2-yl, thien-3-yl, furan-2-yl and furan-3-yl.
  • R 2 is a 6-member heteroaryl selected from the group consisting of a pyridyl, a pyrazine, and a pyrimidinyl.
  • R 2 is pyridin-2-yl, pyridin-3-yl, or pyridin-4-yl.
  • R 2 is Ci -6 alkyl optionally substituted with 1, 2, 3, 4 or 5 substituents selected independently from the group consisting of Q -6 acyl, acyloxy, C 2-6 alkenyl, Ci -6 alkoxy, Ci -6 alkyl, Ci -6 alkylcarboxamido, C 2-6 alkynyl, Ci -6 alkylsulfinyl, Q -6 alkylsulfonyl, Ci -6 alkylthio, Ci -6 alkylureyl, amino, Q -6 alkylamino, C 2-6 dialkylamino, carbo-Q -6 -alkoxy, carboxy, cyano, C 3-6 cycloalkyl, C 2-6 dialkylcarboxamido, halogen, Ci -6 haloalkoxy, Ci -6 haloalky
  • R 2 is Cj -6 alkyl optionally substituted with 1, 2, 3, 4 or 5 substituents selected independently from the group consisting of Ci -6 acyl, C 2-6 alkenyl, Ci -6 alkoxy, Ci -6 alkyl, Ci -6 alkylcarboxamido, C 2-6 alkynyl, Ci -6 alkylsulfinyl, Ci -6 alkylsulfonyl, Ci -6 alkylthio, Ci -6 alkylureyl, Ci -6 alkylamino, C 2-6 dialkylamino, carbo-Ci -6 -alkoxy, carboxy, cyano, C 3 .
  • R 2 is Cj -6 alkyl optionally substituted with 1, 2, 3, 4 or 5 substituents selected independently from the group consisting of Ci -6 acyl, Ci -6 alkoxy, Ci -6 alkyl, Ci -6 alkylamino, C 2-6 dialkylamino, carbo-Ci -6 -alkoxy, carboxy, cyano, halogen, Ci -6 haloalkoxy, Ci -6 haloalkyl, hydroxyl and nitro.
  • R 2 is Ci -6 alkyl optionally substituted with 1, 2, 3, 4 or 5 substituents selected independently from the group consisting OfCi -6 alkoxy, Q -6 alkyl, carboxy, cyano, halogen, Ci -6 haloalkoxy, and C ⁇ 6 haloalkyl.
  • R 2 is C 3-7 cycloalkyl optionally substituted with 1, 2, 3, 4 or 5 substituents selected independently from the group consisting OfCi -6 alkoxy, C x-6 alkyl, carboxy, cyano, halogen, Ci -6 haloalkoxy, and Ci -6 haloalkyl.
  • R 2 is C 2-6 alkenyl optionally substituted with 1, 2, 3, 4 or 5 substituents selected independently from the group consisting of Ci -6 acyl, acyloxy, C 2-6 alkenyl, Ci -6 alkoxy, Ci -6 alkyl, Ci -6 alkylcarboxamido, C 2-6 alkynyl, C J-6 alkylsulfinyl, Ci -6 alkylsulfonyl, Ci -6 alkylthio, Ci -6 alkylureyl, amino, Ci -6 alkylamino, C 2-6 dialkylamino, carbo-Ci -6 -alkoxy, carboxy, cyano, C 3-6 cycloalkyl, C 2-6 dialkylcarboxamido, halogen, Q -6 haloalkoxy, Ci -6 haloalkyl, Q -6 haloalkylsulfinyl, Ci -6 haloalkylsulfonyl,
  • R 2 is C 2-6 alkenyl optionally substituted with 1, 2, 3, 4 or 5 substituents selected independently from the group consisting Of Ci -6 alkoxy, Ci -6 alkyl, carboxy, cyano, halogen, Ci -6 haloalkoxy, and C 1-6 haloalkyl.
  • R 2 is Ci -6 haloalkyl optionally substituted with 1, 2, 3, 4 or 5 substituents selected independently from the group consisting of Ci -6 acyl, acyloxy, C 2-6 alkenyl, Ci -6 alkoxy, Ci -6 alkyl, Ci -6 alkylcarboxamido, C 2-6 alkynyl, Ci -6 alkylsulfinyl, Ci -6 alkylsulfonyl, Ci -6 alkylthio, Ci -6 alkylureyl, amino, Ci -6 alkylamino, C 2-6 dialkylamino, carbo-Q -6 -alkoxy, carboxy, cyano, C 3-6 cycloalkyl, C 2-6 dialkylcarboxamido, halogen, C 1-6 haloalkoxy, C] -6 haloalkylsulfinyl, Ci -6 haloalkylsulfonyl, Q -6 haloalkyl
  • R 2 is Ci -6 haloalkyl optionally substituted with 1, 2, 3, 4 or 5 substituents selected independently from the group consisting OfCi -6 alkoxy, Ci -6 alkyl, carboxy, cyano, halogen, and Ci -6 haloalkoxy.
  • A is Ci -3 alkylene optionally substituted with 1, 2, 3 or 4 substituents selected independently from the group consisting Of C 1-6 alkoxy, C 1-6 alkyl, Ci -6 alkylsulfmyl, Ci -6 alkylsulfonyl, amino, Ci -6 alkylamino, C 2-6 dialkylamino, carbo-Ci -6 -alkoxy, carboxy, cyano, C 3-6 cycloalkyl, halogen, Ci -6 haloalkoxy, Ci -6 haloalkyl, Ci -6 haloalkylsulf ⁇ nyl, Ci -6 haloalkylsulfonyl, Ci -6 haloalkylthio and hydroxyl.
  • A is C 1-3 alkylene optionally substituted with 1, 2, 3 or 4 substituents selected independently from the group consisting of Ci -6 alkoxy, Q -6 alkyl, carbo-Ci. 6-alkoxy, carboxy, cyano, halogen, Ci -6 haloalkoxy, C 1-6 haloalkyl and hydroxyl.
  • A is Ci -3 alkylene optionally substituted with 1, 2, 3 or 4 substituents selected independently from the group consisting of Ci -6 alkoxy, Ci -6 alkyl, carboxy, cyano, halogen and Ci -6 haloalkyl.
  • A is -CH 2 -, -CH(CH 3 )-, -C(CH 3 ) 2 - or -CH 2 CH 2 -.
  • B is Ci -3 alkylene optionally substituted with 1, 2, 3 or 4 substituents selected independently from the group consisting of Ci -6 alkoxy, Ci -6 alkyl, Ci -6 alkylsulfmyl, Ci -6 alkylsulfonyl, amino, Ci -6 alkylamino, C 2-6 dialkylamino, carbo-Ci -6 -alkoxy, carboxy, cyano, C 3-6 cycloalkyl, halogen, C 1-6 haloalkoxy, Ci -6 haloalkyl, Ci -6 haloalkylsulfinyl, Cj -6 haloalkylsulfonyl, Ci -6 haloalkylthio and hydroxyl.
  • B is Ci -3 alkylene optionally substituted with 1, 2, 3 or 4 substituents selected independently from the group consisting Of Ci -6 alkoxy, C x-6 alkyl, carbo-Q. 6 -alkoxy, carboxy, cyano, halogen, C 1-6 haloalkoxy, C 1-6 haloalkyl and hydroxyl.
  • B is C 1-3 alkylene optionally substituted with 1, 2, 3 or 4 substituents selected independently from the group consisting of Ci -6 alkoxy, Ci -6 alkyl, carboxy, cyano, halogen and Ci -6 haloalkyl.
  • B is -CH 2 -, -CH(CH 3 )-, -C(CH 3 ) 2 - or -CH 2 CH 2 -.
  • B is a bond.
  • B can also be referred to as being absent and it is understood that when B is a bond that R 2 is directly bonded to the nitrogen as illustrated in Formula (Im) below:
  • R 3 is selected from the group consisting of H, Ci -6 acyl, acyloxy, C 2-6 alkenyl, Ci -6 alkoxy, Ci -6 alkyl, C 1-6 alkylcarboxamido, C 2-6 alkynyl, C 1-6 alkylsulfinyl, C 1-6 alkylsulfonyl, C 1-6 alkylthio, C 1-6 alkylureyl, amino, C 1-6 alkylamino, C 2-6 dialkylamino, carbo-Q.
  • R 3 is selected from the group consisting of H, Ci -6 acyl, acyloxy, C 2-6 alkenyl, Ci -6 alkoxy, Ci -6 alkyl, Ci -6 alkylcarboxamido, C 2-6 alkynyl, Ci -6 alkylsulfinyl, Ci -6 alkylsulfonyl, Ci -6 alkylthio, Ci -6 alkylureyl, amino, Ci -6 alkylamino, C 3-6 dialkylamino, carbo-Q.
  • R 3 is selected from the group consisting of H, Ci -6 alkoxy, Q -6 alkyl, cyano, C 3-6 cycloalkyl, halogen, Ci -6 haloalkoxy, Q -6 haloalkyl, hydroxyl, nitro and thiol.
  • R 3 is selected from the group consisting of H, -CH 3 , -CH 2 CH 3 , cyano, F, Cl, Br, CF 3 and hydroxyl. In some embodiments, R 3 is H.
  • R 4 is Ci -6 alkyl. In some embodiments, R 4 is selected from the group consisting of -CH 3 , -CH 2 CH 3 , -(CH 2 ) 2 CH 3 , -(CH 2 ) 3 CH 3 , -(CH 2 ) 4 CH 3 , -(CH 2 ) 5 CH 3 , and -(CH 2 ) 6 CH 3 . In some embodiments, R 4 is H.
  • Ri-A together is benzyl, sec-butyl, 1 -methyl-butyl, thiophen-3-ylmethyl, 5-bromo- thiophen-2-ylmethyl, propyl, cyclopropylmethyl, 3 -methyl-butyl, 3-methyl-but-2-enyl, cyclohexylmethyl, phenethyl, hexyl, benzo[l,3]dioxol-4-ylmethyl, thiophen-2-ylniethyl, 5- methyl-thiophen-2-ylmethyl, furan-3-ylmethyl, 4-fluoro-benzyl, 3-fluoro-benzyl, or 2-fluoro- benzyl;
  • Compounds of the present invention can modulate the activity of the RUP38 receptor.
  • modulate is meant to refer to the ability to increase or decrease activity of the receptor.
  • compounds of the invention can be used in methods of modulating a RUP38 receptor comprising contacting the receptor with an effective amount of any one or more of the compounds as described herein.
  • compounds of the invention can be used in methods of modulating a RUP38 receptor for the treatment of a metabolic-related disorder in an individual in need of such modulation comprising contacting the receptor with a therapeutically-effective amount of a compound of Formula (I).
  • compounds of the invention increase activity of the RUP38 receptor.
  • compounds of the invention are agonists of the RUP38 receptor.
  • agonist refers to an agent that can stimulate the activity of the receptor (i.e., activate).
  • compounds of the invention are partial agonists of the RUP38 receptor.
  • Some embodiments of the present invention include methods of modulating the RUP 38 receptor function in a cell, tissue or individual comprising contacting the cell, tissue or individual with an effective amount of a compound of the present invention or a pharmaceutical composition thereof as described herein.
  • the RUP 38 receptor function is associated with a metabolic-related disorder.
  • Another aspect of the present invention pertains to methods of treatment of a metabolic- related disorder comprising administering to an individual in need of such treatment a therapeutically-effective amount of a compound of Formula (I).
  • Another aspect of the present invention pertains to methods of raising HDL cholesterol levels in an individual comprising administering to the individual a therapeutically-effective amount of a compound of Formula (I).
  • Another aspect of the present invention pertains to compounds of Formula (I), as described herein, for use in a method of treatment of the human or animal body by therapy.
  • Another aspect of the present invention pertains to compounds of Formula (I), as described herein, for use in a method of treatment of a metabolic-related disorder of the human or animal body by therapy.
  • Another aspect of the present invention pertains to compounds of Formula (I), as described herein, for use in a method of treatment of a metabolic-related disorder of the human or animal body by therapy wherein the metabolic-related disorder is selected from the group consisting of dyslipidemia, atherosclerosis, coronary heart disease, insulin resistance, obesity, impaired glucose tolerance, atheromatous disease, hypertension, stroke, Syndrome X, heart disease, type 2 diabetes, hypo-HDL related atherosclerotic risk, ischemic cerebrovascular disease, peripheral vascular disease, stroke, and myocardial infarction.
  • the metabolic-related disorder is selected from the group consisting of dyslipidemia, atherosclerosis, coronary heart disease, insulin resistance, obesity, impaired glucose tolerance, atheromatous disease, hypertension, stroke, Syndrome X, heart disease, type 2 diabetes, hypo-HDL related atherosclerotic risk, ischemic cerebrovascular disease, peripheral vascular disease, stroke, and myocardial infarction.
  • Another aspect of the present invention pertains to compounds of Formula (I), as described herein, for use in a method of treatment of a metabolic-related disorder of the human or animal body by therapy wherein the metabolic-related disorder is selected from the group consisting of dyslipidemia, atherosclerosis, coronary heart disease, insulin resistance and type 2 diabetes.
  • Another aspect of the present invention pertains to compounds of Formula (I), as described herein, for use in a method of treatment of atherosclerosis of the human or animal body by therapy.
  • Another aspect of the present invention pertains to compounds of Formula (I), as described herein, for use in a method of raising HDL cholesterol levels of the human or animal body by therapy.
  • Another aspect of the present invention pertains to uses of the compounds of Formula (I), as described herein, for the manufacture of a medicament for use in the treatment of a metabolic- related disorder.
  • Another aspect of the present invention pertains to uses of the compounds of Formula (I), as described herein, for the manufacture of a medicament for use in the treatment of a metabolic- related disorder selected from the group consisting of dyslipidemia, atherosclerosis, coronary heart disease, insulin resistance, obesity, impaired glucose tolerance, atheromatous disease, hypertension, stroke, Syndrome X, heart disease, type 2 diabetes, hypo-HDL related atherosclerotic risk, ischemic cerebrovascular disease, peripheral vascular disease, stroke, and myocardial infarction.
  • Another aspect of the present invention pertains to uses of the compounds of Formula (I), as described herein, for the manufacture of a medicament for use in the treatment of atherosclerosis. Another aspect of the present invention pertains to uses of the compounds of Formula (I), as described herein, for the manufacture of a medicament for use in raising HDL cholesterol levels in an individual.
  • the metabolic-related disorder is of the group consisting of dyslipidemia, atherosclerosis, coronary heart disease, insulin resistance, obesity, impaired glucose tolerance, atheromatous disease, hypertension, stroke, Syndrome X, heart disease, type 2 diabetes, hypo-HDL related atherosclerotic risk, ischemic condition such as ischemic cerebrovascular disease, stoke, peripheral vascular disease, stroke, myocardial infarction, and the like.
  • the metabolic-related disorder is dyslipidemia, atherosclerosis, coronary heart disease, insulin resistance and type 2 diabetes.
  • the metabolic-related disorder is dyslipidemia.
  • the metabolic-related disorder is atherosclerosis.
  • the metabolic-related disorder is coronary heart disease.
  • the metabolic-related disorder is insulin resistance.
  • the metabolic-related disorder is type 2 diabetes.
  • the individual is a mammal.
  • the mammal is a human.
  • Another aspect of the present invention pertains to methods of producing a pharmaceutical composition
  • a pharmaceutical composition comprising admixing or combining a compound of Formula (I), as described herein, and a pharmaceutically acceptable carrier.
  • phrases "pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgement, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • the present invention also includes pharmaceutically acceptable salts of the compounds described herein.
  • pharmaceutically acceptable salts refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form.
  • pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • the pharmaceutically acceptable salts of the present invention include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods.
  • such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred.
  • Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418 and Journal of Pharmaceutical Science, 66, 2 (1977), each of which is incorporated herein by reference in its entirety.
  • prodrugs refer to any covalently bonded carriers which release the active parent drug when administered to a mammalian subject.
  • Prodrugs can be prepared by modifying functional groups present in the compounds in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compounds.
  • Prodrugs include compounds wherein hydroxyl, amino, sulfhydryl, or carboxyl groups are bonded to any group that, when administered to a mammalian subject, cleaves to form a free hydroxyl, amino, sulfhydryl, or carboxyl group respectively.
  • prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol and amine functional groups in the compounds of the invention. Preparation and use of prodrugs is discussed in T. Higuchi and V. Stella, "Pro-drugs as Novel Delivery Systems,” Vol. 14 of the A.C.S. Symposium Series, and in Bior ⁇ versible Carriers in Drug
  • Combination Therapy While the compounds of the invention can be administered as the sole active pharmaceutical agent (i.e., mono-therapy), compounds of the invention can also be used in combination with other pharmaceutical agents (i.e., combination-therapy) for the treatment of the diseases/conditions/disorders described herein. Therefore, another aspect of the present invention includes methods of treatment comprising administering to an individual in need of treatment a therapeutically effective amount of a compound of the present invention, for example Formula (I), in combination with one or more additional pharmaceutical agent as described herein.
  • a therapeutically effective amount of a compound of the present invention for example Formula (I), in combination with one or more additional pharmaceutical agent as described herein.
  • Suitable pharmaceutical agents that can be used in combination with the compounds of the present invention include anti-obesity agents such as apolipoprotein-B secretion/microsomal triglyceride transfer protein (apo-B/MTP) inhibitors, MCR-4 agonists, cholescystokim ' n-A (CCK- A) agonists, serotonin and norepinephrine reuptake inhibitors (for example, sibutramine), sympathomimetic agensts, ⁇ 3 adrenergic receptor agonists, dopamine agonists (for example, bromocriptine), melanocyte-stimulating hormone receptor analogs, cannabinoid 1 receptor antagonists [for example, SR141716: N-(piperidin-l-yl) ⁇ 5-(4-chlorophenyl)-l-(2,4- dichlorophenyl)-4-methyl-lH-pyrazole-3-carboxamide], melanin concentrating hormone antagonists
  • the anti-obesity agents are selected from the group consisting of orlistat, sibutramine, bromocriptine, ephedrine, leptin, and pseudoephedrine.
  • compounds of the present invention and combination therapies are administered in conjunction with exercise and/or a sensible diet.
  • combination-therapy of the compounds of the present invention with other anti-obesity agents, anorectic agents, appetite suppressant and related agents is not limited to those listed above, but includes in principle any combination with any pharmaceutical agent or pharmaceutical composition useful for the treatment of overweight and obese individuals.
  • Suitable pharmaceutical agents in addition to anti-obesity agents, that can be used in combination with the compounds of the present invention include agents useful in the treatment of concomitant diseases.
  • concomitant diseases such as, metabolic-related disorders including congestive heart failure, type 2 diabetes, atherosclerosis, dyslipidemia, hyperinsulinemia, hypertension, insulin resistance, hyperglycemia, retinopathy, nephropathy, neuropathy, and the like.
  • Treatment for one or more of the diseases cited herein include the use of one or more pharmaceutical agents known in the art belonging to the classes of drugs referred to, but not limited to, the following: sulfonylureas, meglitinides, biguanides, ⁇ -glucosidase inhibitors, peroxisome proliferators-activated receptor- ⁇ (i.e., PPAR- ⁇ ) agonists, insulin, insulin analogues, HMG-CoA reductase inhibitors, cholesterol-lowering drugs (for example, f ⁇ brates that include: fenof ⁇ brate, bezafibrate, gemfibrozil, clofibrate and the like; 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
  • 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 diseases, conditions or disorders that are linked to overweight and obese individuals.
  • Some embodiments of the present invention include methods of treatment of a disease, disorder or condition as described herein comprising administering to an individual in need of such treatment a therapeutically effect amount or dose of a compound of the present invention in combination with at least one pharmaceutical agent selected from the group consisting of: sulfonylureas, meglitinides, biguanides, ⁇ -glucosidase inhibitors, peroxisome proliferators- activated receptor- ⁇ (i.e., PPAR- ⁇ ) agonists, insulin, insulin analogues, HMG-CoA reductase inhibitors, cholesterol-lowering drugs (for example, f ⁇ brates that include: fenof ⁇ brate, bezafibrate, gemfibrozil, clofibrate and the like; bile acid sequestrants which include: cholestyramine, colestipol and the like; and niacin), antiplatelet agents (for example, aspirin and adenosine diphosphate receptor antagonists that
  • Suitable pharmaceutical agents that can be used in conjunction with compounds of the present invention include ⁇ -glucosidase inhibitors.
  • ⁇ -Glucosidase inhibitors belong to the class of drugs which competitively inhibit digestive enzymes such as ⁇ -amylase, maltase, ⁇ -dextrinase, sucrase, etc. in the pancreas and or small intesting.
  • the reversible inhibition by ⁇ -glucosidase inhibitors retard, diminish or otherwise reduce blood glucose levels by delaying the digestion of starch and sugars.
  • ⁇ -glucosidase inhibitors include acarbose, N- (l,3-dihydroxy-2-propyl)valiolamine (generic name; voglibose), miglitol, and ⁇ -glucosidase inhibitors known in the art.
  • Suitable pharmaceutical agents that can be used in conjunction with compounds of the present invention include sulfonylureas.
  • the sulfonylureas (SU) are drugs which promote secretion of insulin from pancreatic ⁇ cells by transmitting signals of insulin secretion via SU receptors in the cell membranes.
  • Examples of the sulfonylureas include glyburide, glipizide, glimepiride and other sulfonylureas known in the art.
  • Suitable pharmaceutical agents that can be used in conjunction with compounds of the present invention include the meglitinides.
  • the meglitinides are benzoic acid derivatives represent a novel class of insulin secretagogues. These agents target postprandial hyperglycemia and show comparable efficacy to sulfonylureas in reducing HbAi 0 .
  • Examples of meglitinides include repaglinide, nateglinide and other meglitinides known in the art.
  • Suitable pharmaceutical agents that can be used in conjunction with compounds of the present invention include the biguanides.
  • the biguanides represent 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, metformin, buformin, and biguanides known in the art.
  • Suitable pharmaceutical agents that can be used in conjunction with compounds of the present invention include the ⁇ -glucosidase inhibitors.
  • Suitable pharmaceutical agents that can be used in conjunction with compounds of the present invention include the peroxisome proliferators-activated receptor- ⁇ (i.e., PPAR- ⁇ ) agonists.
  • the peroxisome proliferators-activated receptor- ⁇ agonists represent a class of compounds that activates the nuclear receptor PPAR- ⁇ and therefore regulate the transcription of insulin-responsive genes involved in the control of glucose production, transport and utilization. Agents in the class also facilitate the regulation of fatty acid metabolism.
  • Examples of PPAR- ⁇ agonists include rosiglitazone, pioglitazone, tesaglitazar, netoglitazone, GW-409544, GW-501516 and PPAR- ⁇ agonists known in the art.
  • Suitable pharmaceutical agents that can be used in conjunction with compounds of the present invention include the HMG-CoA reductase inhibitors.
  • the HMG-CoA reductase inhibitors are agents also referred to as Statin compounds that belong to a class of drugs that lower blood cholesterol levels by inhibiting hydroxymethylglutalyl CoA (HMG-CoA) reductase.
  • HMG-CoA reductase is the rate-limiting enzyme in cholesterol biosynthesis.
  • the statins lower serum LDL concentrations by upregulating the activity of LDL receptors and are responsible for clearing LDL from the blood.
  • statin compounds include rosuvastatin, pravastatin and its sodium salt, simvastatin, lovastatin, atorvastatin, fluvastatin, cerivastatin, rosuvastatin, pitavastatin, BMS 's "superstatin”, and HMG-CoA reductase inhibitors known in the art.
  • Suitable pharmaceutical agents that can be used in conjunction with compounds of the present invention include the angiotensin converting enzyme (ACE) inhibitors.
  • ACE angiotensin converting enzyme
  • the angiotensin converting enzyme inhibitors belong to the class of drugs that partially lower blood glucose levels as well as lowering blood pressure by inhibiting angiotensin converting enzymes.
  • angiotensin converting enzyme inhibitors examples include captopril, enalapril, alacepril, delapril; ramipril, lisinopril, imidapril, benazepril, ceronapril, cilazapril, enalaprilat, fosinopril, moveltopril, perindopril, quinapril, spirapril, temocapril, trandolapril, and angiotensin converting enzyme inhibitors known in the art.
  • Suitable pharmaceutical agents that can be used in conjunction with compounds of the present invention include the angiotensin II receptor antagonists.
  • Angiotensin II receptor antagonists target the angiotensin II receptor subtype 1 (i.e., ATI) and demonstrate a beneficial effect on hypertension.
  • Examples of angiotensin II receptor antagonists include losartan (and the potassium salt form), and angiotensin II receptor antagonists known in the art.
  • Suitable pharmaceutical agents that can be used in conjunction with compounds of the present invention include DP receptor antagonists.
  • DP receptor antagonists include those described in WO01/79169, EP03/062200, WO01/66520, WO03/022814, WO03/078409, WO2004/103370, and like antagonists.
  • amylin agonists for example, pramlintide
  • insulin secretagogues for example, GLP-I agonists; exendin-4; insulinotropin (NN2211); dipeptyl peptidase inhibitors (for example, NVP-DPP-728), acyl CoA cholesterol acetyltransferase inhibitors (for example, Ezetimibe, eflucimibe, and like compounds), cholesterol absorption inhibitors (for example, ezetimibe, pamaqueside and like compounds), cholesterol ester transfer protein inhibitors (for example, CP-529414, JTT-705, CETi-I, and like compounds), microsomal triglyceride transfer protein inhibitors (for example, implitapide, and like compounds), cholesterol modulators (for example, NO-1886, and like compounds), bile acid modul
  • Squalene synthesis inhibitors belong to a class of drugs that lower blood cholesterol levels by inhibiting synthesis of squalene.
  • examples of the squalene synthesis inhibitors include (S)- ⁇ -[Bis[2,2-dimethyl-l-oxopropoxy)methoxy] phosphinyl]-3-phenoxybenzenebutanesulfonic acid, mono potassium salt (BMS-188494) and squalene synthesis inhibitors known in the art.
  • compositions of the Present Invention also pertains to pharmaceutical compositions comprising one or more compounds of Formula (I) or any formulae disclosed herein, and one or more pharmaceutically acceptable carriers.
  • Some embodiments of the present invention include a method of producing a pharmaceutical composition comprising admixing at least one compound according to any of the compound embodiments disclosed herein and a pharmaceutically acceptable carrier.
  • 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.
  • 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 ampoule. These are just a few examples of the many appropriate methods well known in the art for preparing dosage forms.
  • a compound of the present invention can be formulated into pharmaceutical compositions using techniques well known to those in the art. Suitable pharmaceutically-acceptable carriers, outside those mentioned herein, are known in the art; for example, see Remington, The Science and Practice of Pharmacy, 20 th Edition, 2000, Lippincott Williams & Wilkins, (Editors: Gennaro, A. R., 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.
  • the invention thus further provides pharmaceutical formulations comprising a compound of the invention or a pharmaceutically acceptable salt thereof together with one or more pharmaceutically acceptable carriers thereof.
  • the carrier(s) must be "acceptable” in the sense of being compatible with the other ingredients of the formulation and not overly deleterious to the recipient thereof.
  • Transdermal patches dispense a drug at a controlled rate by presenting the drug for absorption in an efficient manner with a minimum of degradation of the drug.
  • transdermal patches comprise an impermeable backing layer, a single pressure sensitive adhesive and a removable protective layer with a release liner.
  • 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 is defined in the context of a "pharmaceutical composition” and shall mean 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.
  • doses of the present invention include, but not limited to, about 0.001 mg to about 5000 mg, about 0.001 to about 2500 mg, about 0.001 to about 1000 mg, 0.001 to about 500 mg, 0.001 mg to about 250 mg, about 0.001 mg to 100 mg, about 0.001 mg to about 50 mg, and about 0.001 mg to about 25 mg.
  • 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 care-giver 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
  • animal models include, but are not limited to, the rodents.
  • these extrapolations may merely be based on the weight of the animal in the respective model in comparison to another, such as a mammal, preferably a human, however, more often, these extrapolations are not simply based on weights, but rather incorporate a variety of factors.
  • Representative factors include, but not limited to, 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 on whether further active compounds are administered in addition to the compounds of the Formula (I) 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.
  • 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 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, flavouring 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" is intended to include the formulation of the active compound with encapsulating material as carrier providing a capsule in which the active component, with or without carriers, is surrounded by a carrier, which is thus in association with it.
  • 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.
  • the pharmaceutical compositions may take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the active ingredient may be in powder form, obtained by aseptic isolation of sterile solid or by lyophilization from solution, for constitution with a suitable vehicle, e.g. sterile, pyrogen-free water, before use.
  • Aqueous formulations suitable for oral use can be prepared by dissolving or suspending the active component in water and adding suitable colorants, flavours, stabilizing and thickening agents, as desired.
  • Aqueous suspensions suitable for oral use can be made by dispersing the finely divided active component in water with viscous material, such as natural or synthetic gums, resins, methylcellulose, sodium carboxymethylcellulose, or other well known suspending agents.
  • solid form preparations which are intended to be converted, shortly before use, to liquid form preparations for oral administration.
  • liquid forms include solutions, suspensions, and emulsions.
  • These preparations may contain, in addition to the active component, colorants, flavors, stabilizers, buffers, artificial and natural sweeteners, dispersants, thickeners, solubilizing agents, and the like.
  • the compounds according to the invention may be formulated as ointments, creams or lotions, or as a transdermal patch.
  • Ointments and creams may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agents.
  • Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilizing agents, dispersing agents, suspending agents, thickening agents, or coloring agents.
  • 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.
  • the compounds of the Formula (I) 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 Formula (I) 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 Formula (I) 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).
  • PVP polyvinylpyrrolidone
  • 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.
  • 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.
  • Compounds of the invention can be prepared using known organic synthesis techniques and can be synthesized according to any of the numerous possible synthetic routes.
  • the compounds described herein can be asymmetric (e.g., having one or more stereocenters). All stereoisomers, such as enantiomers and diastereomers, are intended unless otherwise indicated.
  • Compounds of the present invention that contain asymmetrically substituted carbon atoms can be isolated in optically active or racemic forms. Methods on how to prepare optically active forms from optically active starting materials are known in the art, such as by resolution of racemic mixtures or by stereoselective synthesis.
  • Suitable resolving agents for fractional recrystallization methods are, for example, optically active acids, such as the D and L forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or the various optically active camphorsulfonic acids such as ⁇ -camphorsulfonic acid.
  • optically active acids such as the D and L forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or the various optically active camphorsulfonic acids such as ⁇ -camphorsulfonic acid.
  • resolving agents suitable for fractional crystallization methods include stereoisomerically pure forms of ⁇ -methylbenzylamine (e.g., S and R forms, or diastereomerically pure forms), 2-phenylglycinol, norephedrine, ephedrine, N-methylephedrine, cyclohexylethylamine, 1,2-diaminocyclohexane, and the like.
  • Resolution of racemic mixtures can also be carried out by elution on a column packed with an optically active resolving agent (e.g., dinitrobenzoylphenylglycine).
  • Suitable elution solvent composition can be determined by one skilled in the art.
  • the reactions for preparing compounds of the invention can be carried out in suitable solvents which can be readily selected by one of skill in the art of organic synthesis.
  • suitable solvents can be substantially nonreactive with the starting materials (reactants), the intermediates, or products at the temperatures at which the reactions are carried out, i.e., temperatures which can range from the solvent's freezing temperature to the solvent's boiling temperature.
  • a given reaction can be carried out in one solvent or a mixture of more than one solvent.
  • suitable solvents for a particular reaction step can be selected.
  • amino-pyrazoles of Formula (B) can be prepared via reduction of the nitro group.
  • "PG” is a C 1-6 alkyl group.
  • amino-pyrazoles of Formula (B) can be converted to mono-N-substituted aminopyrazoles of Formula (C) via a reductive amination. This step involves two aspects, 1) the formation of an intermediate imine and 2) reduction of the imine to mono-N-substituted aminopyrazoles of Formula (C).
  • the formation of the imine and reduction can be conducted as one step without isolation of the imine or the reduction can be conducted with an isolated imine as a separate step.
  • a variety of aldehydes and ketones can be used in this step providing a wide variety of Ri-A- groups.
  • Mono-N-substituted aminopyrazoles of Formula (C) can be converted to compounds of the invention, wherein R 2 -B- is hydrogen, by removal of the protecting group to give compounds of Formula (D) wherein R 4 is hydrogen or, if "PG" was a protecting group other than a C 1-6 alkyl group, the carboxylic acid (R 4 is hydrogen) can be converted to an ester wherein R 4 is Ci_ 6 alkyl.
  • Mono-N-substituted aminopyrazoles of Formula (C) or (D) can be converted to di-N-substituted aminopyrazoles of Formula (E) or (I) via a similar second reductive step and, if needed, a conversion of group "PG" to R 4 as described supra.
  • amino-pyrazoles of Formula (B) can be converted to compounds of Formula (I) via a two part reductive amination utilizing only one procedural step; generally this method is useful for the preparation of symmetrical di-N- subsituted aminopyrazoles.
  • the Formulae described in Scheme I represent compounds wherein R 3 is hydrogen.
  • Another representative synthesis is set forth below in Scheme II for wherein R 3 is fluorine: Scheme II
  • R 3 is fluorine
  • the fluorination step can be implemented at any point during the syntheses.
  • aminopyrazoles of Formula (F) can be fluorinated to give fluoropyrazoles of Formula (G), wherein R 3 is F.
  • fluoropyrazoles of Formula (G) can be converted to compounds of Formulae (H, wherein R 3 is F), (J), (K) and (L).
  • fluorine can be introduced in a later step.
  • mono-N-substituted aminopyrazoles of Formula (H), wherein R 3 is hydrogen can undergo a fluorination step to give Compounds (J).
  • mono-N-substituted aminopyrazoles of Formula (H) can be converted to Compounds (K) and (L).
  • Suitable fluorinating agents include, l-chloromethyl-4-fluoro-l,4-diazoniabicyclo[2.2.2]octane bis- tetrafluoroborate (Selectfluor®), bis-(2-methoxyethyl)aminosulfur trifluoride (Deoxo-FluorTM), diethylaminosulfur trifluoride (DAST), bis(2-methoxyethyl)aminosulfur trifluoride (BAST), and the like.
  • One representative fluorinating procedure is as follows.
  • the resulting solution is heated to above RT, such as about 65 0 C, in a suitable reaction vessel, such as a sealed polypropylene flask, and the like, for a suitable time (monitored by an analytical method, such as, TLC, HPLC, and the like).
  • the solvent is removed under reduced pressure, and the resulting crude product is taken up in a suitable solvent, such as dichloromethane, ethyl acetate and the like, and is washed with aqueous hydrochloric acid (for example, about IM to 3M).
  • a suitable solvent such as dichloromethane, ethyl acetate and the like
  • aqueous hydrochloric acid for example, about IM to 3M.
  • the solvent is removed under reduced pressure and the residual purified, for example, by column chromatography (alumina, silica, and the like) to give the fluorinated pyrazole.
  • Preparation of compounds of the invention can involve the protection and deprotection of various chemical groups.
  • the need for protection and deprotection, and the selection of appropriate protecting groups can be readily determined by one skilled in the art.
  • the chemistry of protecting groups can be found, for example, in T.W. Green and P.G.M. Wuts, Protective Groups in Organic Synthesis, 3rd. Ed., Wiley & Sons, Inc., New York (1999), which is incorporated herein by reference in its entirety.
  • Reactions can be monitored according to any suitable method known in the art.
  • product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., 1 H or 13 C) infrared spectroscopy, spectrophotometry (e.g., UV- visible), or mass spectrometry, or by chromatography such as high performance liquid chromatography (HPLC) or thin layer chromatography.
  • spectroscopic means such as nuclear magnetic resonance spectroscopy (e.g., 1 H or 13 C) infrared spectroscopy, spectrophotometry (e.g., UV- visible), or mass spectrometry
  • chromatography such as high performance liquid chromatography (HPLC) or thin layer chromatography.
  • this compound can also have another tautomeric form as shown below and based on this chemical representation is appropriately named 5-[(thio ⁇ hen-3- ylmethyl)-amino]-lH-pyrazole-3-carboxylic acid:
  • a G protein-coupled receptor When a G protein-coupled receptor is in its active state, either as a result of ligand binding or constitutive activation, the receptor couples to a G protein and stimulates the release of GDP and subsequent binding of GTP to the G protein.
  • the alpha subunit of the G protein- receptor complex acts as a GTPase and slowly hydrolyzes the GTP to GDP, at which point the receptor normally is deactivated. Activated receptors continue to exchange GDP for GTP.
  • the non-hydrolyzable GTP analog, [ 35 S]GTPyS can be utilized to demonstrate enhanced binding of [ 35 S]GTPyS to membranes expressing activated receptors.
  • the assay utilizes the ability of G protein coupled receptors to stimulate [ 35 S]GTPyS binding to membranes expressing the relevant receptors.
  • the assay can, therefore, be used in the direct identification method to screen candidate compounds to endogenous GPCRs and non- endogenous, constitutively activated GPCRs.
  • the assay is generic and has application to drug discovery at all G protein-coupled receptors.
  • the [ 35 S]GTPyS assay is incubated in 20 mM HEPES and between 1 and about 2OmM MgCl 2 (this amount can be adjusted for optimization of results, although 2OmM is preferred) pH 7.4, binding buffer with between about 0.3 and about 1.2 nM [ 35 S]GTPyS (this amount can be adjusted for optimization of results, although 1.2 is preferred ) and 12.5 to 75 ⁇ g membrane protein (e.g, 293 cells expressing the recombinant GPCR; this amount can be adjusted for optimization) and 10 ⁇ M GDP (this amount can be changed for optimization) for 1 hour.
  • Wheatgerm agglutinin beads (25 ⁇ l; Amersham) are then added and the mixture incubated for another 30 minutes at room temperature. The tubes are then centrifuged at 1500 x g for 5 minutes at room temperature and then counted in a scintillation counter. 2.
  • a Flash PlateTM Adenylyl Cyclase kit (New England Nuclear; Cat. No. SMP004A) designed for cell-based assays can be modified for use with crude plasma membranes.
  • the Flash Plate wells can contain a scintillant coating which also contains a specific antibody recognizing cAMP.
  • the cAMP generated in the wells can be quantitated by a direct competition for binding of radioactive cAMP tracer to the cAMP antibody. The following serves as a brief protocol for the measurement of changes in cAMP levels in whole cells that express the receptors.
  • Transfected cells are harvested approximately twenty four hours after transient transfection. Media is carefully aspirated off and discarded. 10ml of PBS is gently added to each dish of cells followed by careful aspiration. ImI of Sigma cell dissociation buffer and 3ml of PBS are added to each plate. Cells are pipetted off the plate and the cell suspension is collected into a 50ml conical centrifuge tube. Cells are then centrifuged at room temperature at 1,100 rpm for 5 min. The cell pellet is carefully re-suspended into an appropriate volume of PBS (about 3ml/plate). The cells are then counted using a hemocytometer and additional PBS is added to give the appropriate number of cells (with a final volume of about 50 ⁇ l/well).
  • cAMP standards and Detection Buffer comprising 1 ⁇ Ci of tracer [ 125 I] cAMP (50 ⁇ l) to 11 ml Detection Buffer
  • Assay Buffer is prepared fresh for screening and contains 50 ⁇ l of Stimulation Buffer, 3ul of test compound (12 ⁇ M final assay concentration) and 50 ⁇ l cells.
  • Assay Buffer is stored on ice until utilized.
  • the assay preferably carried out e.g. in a 96-well plate, is initiated by addition of 50 ⁇ l of cAMP standards to appropriate wells followed by addition of 50ul of PBSA to wells H-11 and H12. 50 ⁇ l of Stimulation Buffer is added to all wells.
  • DMSO (or selected candidate compounds) is added to appropriate wells using a pin tool capable of dispensing 3 ⁇ l of compound solution, with a final assay concentration of 12uM test compound and lOO ⁇ l total assay volume.
  • the cells are then added to the wells and incubated for 60 min at room temperature.
  • lOO ⁇ l of Detection Mix containing tracer cAMP is then added to the wells. Plates are then incubated additional 2 hours followed by counting in a Wallac MicroBeta scintillation counter. Values of cAMP/well are then extrapolated from a standard cAMP curve which is contained within each assay plate.
  • TSHR is a Gs coupled GPCR that causes the accumulation of cAMP upon activation.
  • TSHR will be constitutively activated by mutating amino acid residue 623 (i.e., changing an alanine residue to an isoleucine residue).
  • a Gi coupled receptor is expected to inhibit adenylyl cyclase, and, therefore, decrease the level of cAMP production, which can make assessment of cAMP levels challenging.
  • An effective technique for measuring the decrease in production of cAMP as an indication of activation of a Gi coupled receptor can be accomplished by co- transfecting, most preferably, non-endogenous, constitutively activated TSHR (TSHR-A623I) (or an endogenous, constitutively active Gs coupled receptor) as a "signal enhancer" with a Gi linked target GPCR to establish a baseline level of cAMP.
  • TSHR-A623I non-endogenous, constitutively activated TSHR
  • Gs coupled receptor an endogenous, constitutively active Gs coupled receptor
  • this approach is preferably used in the direct identification of candidate compounds against Gi coupled receptors. It is noted that for a Gi coupled GPCR, when this approach is used, an inverse agonist of the target GPCR will increase the cAMP signal and an agonist will decrease the cAMP signal.
  • tube A will be prepared by mixing 2 ⁇ g DNA of each receptor transfected into the mammalian cells, for a total of 4 ⁇ g DNA ⁇ e.g., pCMV vector; pCMV vector with mutated THSR (TSHR-A623I); TSHR-A623I and GPCR, etc.) in 1.2ml serum free DMEM (Irvine Scientific, Irvine, CA); tube B will be prepared by mixing 120 ⁇ l lipofectamine (Gibco BRL) in 1.2ml serum free DMEM.
  • Tubes A and B will then be admixed by inversions (several times), followed by incubation at room temperature for 30- 45min. The admixture is referred to as the "transfection mixture”.
  • Plated 293 cells will be washed with IXPBS, followed by addition of 10ml serum free DMEM. 2.4ml of the transfection mixture will then be added to the cells, followed by incubation for 4hrs at 37°C/5% CO 2 . The transfection mixture will then be removed by aspiration, followed by the addition of 25ml of DMEM/10% Fetal Bovine Serum. Cells will then be incubated at 37°C/5% CO 2 . After 24hr incubation, cells will then be harvested and utilized for analysis.
  • a Flash PlateTM Adenylyl Cyclase kit (New England Nuclear; Cat. No. SMP004A) is designed for cell-based assays, but can be modified for use with crude plasma membranes depending on the need of the skilled artisan.
  • the Flash Plate wells will contain a scintillant coating which also contains a specific antibody recognizing cAMP.
  • the cAMP generated in the wells can be quantitated by a direct competition for binding of radioactive cAMP tracer to the c AMP antibody. The following serves as a brief protocol for the measurement of changes in cAMP levels in whole cells that express the receptors.
  • Transfected cells will be harvested approximately twenty four hours after transient transfection. Media will be carefully aspirated off and discarded. 10ml of PBS will be gently added to each dish of cells followed by careful aspiration. ImI of Sigma cell dissociation buffer and 3ml of PBS will be added to each plate. Cells will be pipetted off the plate and the cell suspension will be collected into a 50ml conical centrifuge tube. Cells will then be centrifuged at room temperature at 1,100 rpm for 5 min. The cell pellet will be carefully re-suspended into an appropriate volume of PBS (about 3ml/plate).
  • cAMP standards and Detection Buffer comprising 1 ⁇ Ci of tracer [ 125 I] cAMP (50 ⁇ l) to 11 ml Detection Buffer
  • Assay Buffer should be prepared fresh for screening and contained 50 ⁇ l of Stimulation Buffer, 3 ⁇ l of test compound (12 ⁇ M final assay concentration) and 50 ⁇ l cells, Assay Buffer can be stored on ice until utilized.
  • the assay can be initiated by addition of 50 ⁇ l of cAMP standards to appropriate wells followed by addition of 50 ⁇ l of PBSA to wells H-11 and H12. Fifty ⁇ l of Stimulation Buffer will be added to all wells. Selected compounds ⁇ e.g., TSH) will be added to appropriate wells using a pin tool capable of dispensing 3 ⁇ l of compound solution, with a final assay concentration of 12 ⁇ M test compound and lOO ⁇ l total assay volume. The cells will then be added to the wells and incubated for 60 min at room temperature. lOO ⁇ l of Detection Mix containing tracer cAMP will then be added to the wells. Plates were then incubated additional 2 hours followed by counting in a Wallac MicroBeta scintillation counter. Values of cAMP/well will then be extrapolated from a standard cAMP curve which is contained within each assay plate.
  • Stimulation Buffer will be added to all wells. Selected compounds ⁇ e.g.,
  • RT-PCR was applied to confirm the expression and to determine the tissue distribution of several novel human GPCRs. Oligonucleotides utilized were GPCR-specific and the human multiple tissue cDNA panels (MTC, Clontech) as templates. Taq DNA polymerase (Stratagene) were utilized for the amplification in a 40 ⁇ l reaction according to the manufacturer's instructions. 20 ⁇ l of the reaction will be loaded on a 1.5% agarose gel to analyze the RT-PCR products. Table E below lists RUP38, the cycle conditions and the primers utilized.
  • RT-PCR results for RUP38 are shown in in Figure 2.
  • RUP25 (a GPCR related to RUP38, GeneBank accession number AB065876.1) is included in Figure 2 to indicate that the amplification is specific to RUP38.
  • RNA samples were amplified, labeled, hybridized to the microarray, and data analyzed according to manufacturer's instructions.
  • the tissues in Figure 1 are: monocytes (CD 14+), monocytes
  • Adipose tissues were monitored for the level of gene expression of each of the GPCRs represented on the microarray. GPCRs were determined to be expressed if the expression index was greater than 100 (based upon and according to manufacturer's instructions). The data was analyzed and had indicated that classification of GPCRs with an expression index greater than 100 was reasonable because a number of known GPCRs had previously been reported to be expressed in neuronal tissues with an expression index greater than 100.
  • RUP38 Using the GeneChip, we discovered RUP38 to have high levels of expression in adipocytes indicating that, for example, that RUP38 may play a role in lipolysis ⁇ see, Goodman & Gilman's, The Pharmacological Basis of Therapeutics. 9 th Edition, page 235 (1996). See Figure 1.
  • Figure 1 is a plot representing the expression level of RUP38 in various tissues. Based upon this data, RUP38 is highly expressed by primary adipocytes.
  • membranes comprising the Target GPCR of interest and for use in the identification of candidate compounds as, e.g.,. inverse agonists, agonists, or antagonists are preferably prepared as follows: a. Materials "Membrane Scrape Buffer” is comprised of 2OmM HEPES and 1OmM EDTA, pH 7.4;
  • “Membrane Wash Buffer” is comprised of 20 mM HEPES and 0.1 mM EDTA, pH 7.4; "Binding Buffer” is comprised of 2OmM HEPES, 100 mM NaCl, and 10 mM MgCl 2 , pH 7.4. b. Procedure
  • the media will be aspirated from a confluent monolayer of cells, followed by rinse with 10ml cold PBS, followed by aspiration. Thereafter, 5ml of Membrane Scrape Buffer will be added to scrape cells; this will be followed by transfer of cellular extract into 50ml centrifuge tubes (centrifuged at 20,000 rpm for 17 minutes at 4°C). Thereafter, the supernatant will be aspirated and the pellet will be resuspended in 30ml Membrane Wash Buffer followed by centrifuge at 20,000 rpm for 17 minutes at 4°C. The supernatant will then be aspirated and the pellet resuspended in Binding
  • Duplicate tubes will be prepared, one including the membrane, and one as a control "blank". Each contained 800 ⁇ l Binding Buffer. Thereafter, lO ⁇ l of Bradford Protein Standard (lmg/ml) will be added to each tube, and lO ⁇ l of membrane Protein will then be added to just one tube (not the blank). Thereafter, 200 ⁇ l of Bradford Dye Reagent will be added to each tube, followed by vortex of each. After five (5) minutes, the tubes will be re-vortexed and the material therein will be transferred to cuvettes. The cuvettes will then be read using a CECIL 3041 spectrophotometer, at wavelength 595.
  • GDP Buffer consisted of 37.5 ml Binding Buffer and 2mg GDP (Sigma, cat. no. G-7127), followed by a series of dilutions in Binding Buffer to obtain 0.2 ⁇ M GDP (final concentration of GDP in each well was 0.1 ⁇ M GDP); each well comprising a candidate compound, has a final volume of 200 ⁇ l consisting of lOO ⁇ l GDP Buffer (final concentration, 0.1 ⁇ M GDP), 50 ⁇ l Membrane Protein in Binding Buffer, and 50 ⁇ l [ 35 S]GTPyS (0.6 nM) in Binding Buffer (2.5 ⁇ l [ 35 S]GTPyS per 10ml Binding Buffer).
  • Candidate compounds will be preferably screened using a 96-well plate format (these can be frozen at -80 0 C).
  • Membrane Protein or membranes with expression vector excluding the Target GPCR, as control), will be homogenized briefly until in suspension. Protein concentration will then be determined using the Bradford Protein Assay set forth above. Membrane Protein (and control) will then be diluted to 0.25mg/ml in Binding Buffer (final assay concentration,
  • Melanophores are skin cells found in lower vertebrates. They contain pigmented organelles termed melanosomes. Melanophores are able to redistribute these melanosomes along a microtubule network upon G-protein coupled receptor (GPCR) activation. The result of this pigment movement is an apparent lightening or darkening of the cells. fti melanophores, the decreased levels of intracellular cAMP that result from activation of a Gi-coupled receptor cause melanosomes to migrate to the center of the cell, resulting in a dramatic lightening in color. If cAMP levels are then raised, following activation of a Gs-coupled receptor, the melanosomes are re-dispersed and the cells appear dark again.
  • GPCR G-protein coupled receptor
  • the increased levels of diacylglycerol that result from activation of Gq-coupled receptors can also induce this re-dispersion.
  • the technology is also suited to the study of certain receptor tyrosine kinases.
  • the response of the melanophores takes place within minutes of receptor activation and results in a simple, robust color change. The response can be easily detected using a conventional absorbance microplate reader or a modest video imaging system. Unlike other skin cells, the melanophores derive from the neural crest and appear to express a full complement of signaling proteins. In particular, the cells express an extremely wide range of G-proteins and so are able to functionally express almost all GPCRs.
  • Melanophores can be utilized to identify compounds, including natural ligands, against GPCRs. This method can be conducted by introducing test cells of a pigment cell line capable of dispersing or aggregating their pigment in response to a specific stimulus and expressing an exogenous clone coding for the GCPR.
  • a stimulant e.g., melatonin
  • stimulating the cell with a stimulant to set an initial state of pigment disposition wherein the pigment is dispersed if activation of the GPCR induces pigment aggregation.
  • test cells are then contacted with chemical compounds, and it is determined whether the pigment disposition in the cells changed from the initial state of pigment disposition. Dispersion of pigments cells due to the candidate compound, including but not limited to a ligand, coupling to the GPCR will appear dark on a petri dish, while aggregation of pigments cells will appear light.
  • the cells were plated in 96-well plates (one receptor per plate). 48 hours post- transfection, half of the cells on each plate were treated with 1OnM melatonin. Melatonin activates an endogenous Gi-coupled receptor in the melanophores and causes them to aggregate their pigment. The remaining half of the cells were transferred to serum-free medium 0.7X L-15 (Gibco). After one hour, the cells in serum-free media remained in a pigment-dispersed state while the melatonin-treated cells were in a pigment-aggregated state. At this point, the cells were treated with a dose response of nicotinic acid (Sigma).
  • the melanophores would be expected to undergo a color change in response to the compound. If the receptor were either a Gs or Gq coupled receptor, then the melatonin- aggregated melanophores would undergo pigment dispersion. In contrast, if the receptor was a Gi-coupled receptor, then the pigment-dispersed cells would be expected to undergo a dose- dependent pigment aggregation.
  • RUP38 is Gi-coupled. Additionally, RTJP38 may be used in methods described herein to identify antagonists, agonists, inverse agonists, partial agonists, allosteric enhancers, and negative allosteric modulators. Preferably, the modulator is an agonist.
  • Figure 4 presents screening data via adenylyl cyclase assay for RUP38. Note that nicotinic acid does not activate inhibition of forskolin stimulated cAMP RUP38-expressing CHO cells whereas l-Isopropyl-lH-benzotriazole-S-carboxylic acid does.
  • SMP004A was utilized for direct identification of candidate compounds as agonists to RUP38 in accordance with the following protocol:
  • CHO cells stably transfected with RUP38 were harvested from flasks via non-enzymatic means. The cells were washed in PBS and resuspended in the manufacturer's Assay Buffer. Live cells were counted using a hemacytometer and Trypan blue exclusion, and the cell concentration was adjusted to 2x10 6 cells/mL.
  • cAMP standards and Detection Buffer comprising 2 ⁇ Ci of tracer [ 125 I]-cAMP (100 ⁇ l) to 11 mL Detection Buffer) were prepared and maintained in accordance with the manufacturer's instructions.
  • Candidate compounds identified as per above were added to their respective wells (preferably wells of a 96-well plate) at increasing concentrations (3 ⁇ l/well; 12 ⁇ M final assay concentration).
  • Assay Buffer a concentration of Assay Buffer
  • 100,000 cells in 50 ⁇ l of Assay Buffer were added and the mixture was then incubated for 30 minutes at room temperature, with gentle shaking.
  • lOO ⁇ l of Detection Buffer was added to each well, followed by incubation for 2-24 hours. Plates were counted in a Wallac MicroBetaTM plate reader using "Prot. #31" (as per manufacturer instructions).
  • the present invention also provides methods and compositions for the generation of mice and rats that express RUP38 recombinant human antilipolytic GPCR polyeptide of the invention.
  • transgenic mammals can be produced, e.g., by transfecting a pluripotential stem cell such as an ES cell with a polynucleotide encoding RUP38 polypeptide of the invention.
  • a pluripotential stem cell such as an ES cell
  • Successfully transformed ES cells can then be introduced into an early stage embryo that is then implanted into the uterus of a mammal of the same species.
  • the transformed (“transgenic”) cells will comprise part of the germ line of the resulting animal and adult animals comprising the transgenic cells in the germ line can then be mated to other animals, thereby eventually producing a population of transgenic animals that have the transgene in each of their cells and that can stably transmit the transgene to each of their offspring.
  • Other methods of introducing the polynucleotide can be used, for example introducing the polynucleotide encoding RUP38 polypeptide of the invention into a fertilized egg or early stage embryo via microinjection.
  • the transgene may be introduced into an animal by infection of zygotes with a retrovirus containing the transgene [Jaenisch, R, Proc Natl Acad Sci USA (1976) 73:1260-4].
  • Methods of making transgenic mammals are described, e.g., in Wall et al., J Cell Biochem (1992) 49:113-20; Hogan et al., in Manipulating the Mouse Embryo. A Laboratory Manual. (1986) Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N. Y.; in WO 91/08216; or in U.S. Patent No. 4,736, 866; all of which disclosures are hereby incorporated by reference in their entirety.
  • another means for evaluating a test compound is by determining binding affinities to the RUP38 receptor.
  • This type of assay generally requires a radiolabeled ligand to the RTJP38 receptor. Absent the use of known ligands for the RUP38 receptor and radiolabels thereof, compounds of Formula (I) can be labelled with a radioisotope and used in an assay for evaluating the affinity of a test compound to the RUP38 receptor.
  • a radiolabeled RUP38 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
  • the ability to compete with the "radio- labelled compound of Formula (I)" or Radiolabeled RUP38 Ligand for the binding to the RUP38 receptor directly correlates to its binding affinity of the test compound to the RUP38 receptor.
  • RUP38 RECEPTOR PREPARATION 293 cells human kidney, ATCC
  • transiently transfected with 10 ug human RUP38 receptor and 60 ul Lipofectamine per 15-cm dish
  • the cells are then centrifuged in a Beckman Coulter centrifuge for 20 minutes, 17,000 rpm (JA-25.50 rotor).
  • the pellet is resuspended in 20 mM Hepes + 1 mM EDTA, pH 7.4 and homogenized with a 50- mL Dounce homogenizer and again centrifuged. After removing the supernatant, the pellets are stored at -8O 0 C, until used in binding assay.
  • membranes are thawed on ice for 20 minutes and then 10 mL of incubation buffer (20 mM Hepes, 1 mM MgCl 2 , 100 mM NaCl, pH 7.4) added. The membranes are then vortexed to resuspend the crude membrane pellet and homogenized with a Brinkmann PT-3100 Polytron homogenizer for 15 seconds at setting 6. The concentration of membrane protein is determined using the BRL Bradford protein assay.
  • a total volume of 50ul of appropriately diluted membranes (diluted in assay buffer containing 5OmM Tris HCl (pH 7.4), 1OmM MgCl 2 , and ImM EDTA; 5-50ug protein) is added to 96-well polyproylene microtiter plates followed by addition of lOOul of assay buffer and 50ul of Radiolabeled RUP38 Ligand.
  • 50 ul of assay buffer is added instead of lOOul and an additional 50ul of lOuM cold RUP38 is added before 50ul of Radiolabeled RUP38 Ligand is added. Plates are then incubated at room temperature for 60- 120 minutes.
  • the binding reaction is terminated by filtering assay plates through a Microplate Devices GF/C Unifilter filtration plate with a Brandell 96-well plate harvester followed by washing with cold 50 mM Tris HCl, pH 7.4 containing 0.9% NaCl. Then, the bottom of the filtration plate are sealed, 50ul of Optiphase Supermix is added to each well, the top of the plates are sealed, and plates are counted in a Trilux MicroBeta scintillation counter. For compound competition studies, instead of adding lOOul of assay buffer, lOOul of appropriately diluted test compound is added to appropriate wells followed by addition of 50ul of Radiolabeled RUP38 Ligand.
  • test compounds are initially assayed at 1 and 0.1 ⁇ M and then at a range of concentrations chosen such that the middle dose would cause about 50% inhibition of a Radio- RUP38 Ligand binding (i.e., IC 50 ).
  • IC 50 Specific binding in the absence of test compound (B 0 ) is the difference of total binding (B ⁇ ) minus non-specific binding (NSB) and similarly specific binding (in the presence of test compound) (B) is the difference of displacement binding (B D ) minus nonspecific binding (NSB).
  • IC 50 is determined from an inhibition response curve, logit-log plot of % B/Bo vs concentration of test compound. Ki is calculated by the Cheng and Prustoff transformation:
  • Ki IC 50 / (l + [L]/K D ) where [L] is the concentration of a Radio-RUP38 Ligand used in the assay and K D is the dissociation constant of a Radio-RUP38 Ligand determined independently under the same binding conditions.
  • TLC Thin-layer chromatography
  • PK6F silica gel 60 A 1 mm plates (Whatman)
  • column chromatography was carried out on a silica gel column using Kieselgel 60, 0.063-0.200 mm (Merck). Evaporation was done in vacuo on a Buchi rotary evaporator. Celite 545 ® was used during palladium filtrations.
  • LCMS specs 1) PC: HPLC-pumps: LC-IOAD VP, Shimadzu Inc.; HPLC system controller: SCL-IOA VP, Shimadzu Inc; UV-Detector: SPD-IOA VP, Shimadzu Inc; Autosampler: CTC HTS, PAL, Leap Scientific; Mass spectrometer: API 150EX with Turbo Ion Spray source, AB/MDS Sciex; Software: Analyst 1.2. 2) Mac: HPLC-pumps: LC-8A VP, Shimadzu Inc; HPLC system controller: SCL-IOA VP, Shimadzu Inc.
  • UV-Detector SPD-IOA VP, Shimadzu Inc; Autosampler: 215 Liquid Handler, Gilson Inc; Mass spectrometer: API 150EX with Turbo Ion Spray source, AB/MDS Sciex Software: Masschrom 1.5.2.
  • 5-Dibenzylamino-lH-pyrazole-3-carboxylic acid ethyl ester was taken up in IM lithium hydroxide: tetrahydrofuran : methanol (1 : 5 : 1) (10 cm 3 ) and heated at 65 0 C for 18 hours. Solvent was removed under reduced pressure and the crude product was taken to a pH of 1 with 0.1M aqueous HCl and extracted with ethyl acetate (10 cm 3 ). The organic phase was dried over anhydrous Na 2 SO 4 , filtered, and solvent removed under reduced pressure to give 5- (dibenzylammo)-lH-pyrazole-3-carboxylic acid as a brown, glassy solid.
  • the other compounds in the Examples showed EC 50 activities in the membrane cyclase assay less than about 500 ⁇ M.
  • pCMV A variety of expression vectors are available to those in the art for purposes of producing a polypeptide of interest in a cell.
  • One suitable vector is pCMV, which is used in certain embodiments. This vector was deposited with the American Type Culture Collection (ATCC) on October 13, 1998 (10801 University Boulevard., Manassas, VA 20110-2209 USA) under the provisions of the Budapest Treaty for the International Recognition of the Deposit of Microorganisms for the Purpose of Patent Procedure. The DNA was tested by the ATCC and determined to be viable. The ATCC has assigned the following deposit number to pCMV: ATCC #203351.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Diabetes (AREA)
  • General Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Emergency Medicine (AREA)
  • Endocrinology (AREA)
  • Child & Adolescent Psychology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

L'invention concerne des dérivés d'acide ou d'ester carboxylique pyrazole de formule (I) ainsi que des sels pharmaceutiquement acceptables de ceux-ci présentant des propriétés pharmacologiques utiles, par exemple, comme agonistes du récepteur RCPG désignés ci-après RUP38. La présente invention porte également sur des compositions pharmaceutiques contenant des composés de l'invention, et sur des méthodes pour utiliser ces composés et ces compositions dans le traitement de troubles liés au métabolisme tels que la dyslipidémie, l'athérosclérose, l'insuffisance coronaire, la résistance à l'insuline, le diabète de type 2, le syndrome X et d'autres troubles similaires. L'invention concerne aussi l'utilisation desdits composés en combinaison avec d'autres agents actifs tels que ceux appartenant à la catégorie des inhibiteurs de l'α-glucosidase, des inhibiteurs de l'aldose réductase, des biguanides, des inhibiteurs de HMG-CoA réductase, des inhibiteurs de synthèse de squalène, des fibrates, des promoteurs du catabolisme LDL, des inhibiteurs de l'enzyme de conversion de l'angiotensine (ACE), des promoteurs de sécrétion de l'insuline et d'autres agents similaires.
EP06770837A 2005-05-23 2006-05-23 Derives d'acide 5-amino-1h-pyrazole-3-carboxylique en tant qu'agonistes du recepteur couple aux proteines g (rcpg) rup38 pour traiter des troubles lies au metabolisme Withdrawn EP1899304A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US68391205P 2005-05-23 2005-05-23
PCT/US2006/019730 WO2006127595A1 (fr) 2005-05-23 2006-05-23 Derives d'acide 5-amino-1h-pyrazole-3-carboxylique en tant qu'agonistes du recepteur couple aux proteines g (rcpg) rup38 pour traiter des troubles lies au metabolisme

Publications (1)

Publication Number Publication Date
EP1899304A1 true EP1899304A1 (fr) 2008-03-19

Family

ID=37024757

Family Applications (1)

Application Number Title Priority Date Filing Date
EP06770837A Withdrawn EP1899304A1 (fr) 2005-05-23 2006-05-23 Derives d'acide 5-amino-1h-pyrazole-3-carboxylique en tant qu'agonistes du recepteur couple aux proteines g (rcpg) rup38 pour traiter des troubles lies au metabolisme

Country Status (2)

Country Link
EP (1) EP1899304A1 (fr)
WO (1) WO2006127595A1 (fr)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2545823A1 (fr) 2003-11-21 2005-06-09 Arena Pharmaceuticals, Inc. Derives d'acide carboxylique 4-oxo-4,5-dihydrofurane-2 et methodes pour traiter des troubles du metabolisme
PT2713722T (pt) 2011-05-31 2017-06-27 Receptos Llc Novos estabilizadores e moduladores do receptor glp-1
EA031618B1 (ru) 2011-06-09 2019-01-31 Ризен Фармасьютикалз Са Соединения-модуляторы gpr-119
ES2689110T3 (es) 2011-10-06 2018-11-08 Bayer Cropscience Ag Heterociclilpiri(mi)dinilpirazoles como fungicidas
AU2013290100A1 (en) 2012-07-11 2015-01-29 Elcelyx Therapeutics, Inc. Compositions comprising statins, biguanides and further agents for reducing cardiometabolic risk
CN105593225B (zh) 2013-06-11 2019-04-16 赛尔基因第二国际有限公司 新型glp-1受体调节剂
JP2017538711A (ja) 2014-12-10 2017-12-28 セルジーン インターナショナル ツー エスエーアールエル Glp−1レセプターモジュレーター
IL276195B1 (en) * 2018-01-30 2024-02-01 Kainos Medicine Inc Salt forms of an organic compound
KR20240046893A (ko) * 2021-08-10 2024-04-11 텍사스 하트 인스티튜트 아미노아릴-인테그린 작용제 화합물

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001057034A1 (fr) * 2000-02-07 2001-08-09 Bristol-Myers Squibb Co. Inhibiteurs 3-aminopyrazole des kinases dependantes des cyclines
US6902902B2 (en) * 2001-11-27 2005-06-07 Arena Pharmaceuticals, Inc. Human G protein-coupled receptors and modulators thereof for the treatment of metabolic-related disorders
WO2004032928A1 (fr) * 2002-10-10 2004-04-22 Arena Pharmaceuticals, Inc. Derives d'acide 2h-pyrazole-3-carboxylique substitue en 5 utilises en tant qu'agents antilipotyques pour le traitement de troubles metaboliques, tels que la dyslipidemie

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2006127595A1 *

Also Published As

Publication number Publication date
WO2006127595A1 (fr) 2006-11-30

Similar Documents

Publication Publication Date Title
EP1899304A1 (fr) Derives d'acide 5-amino-1h-pyrazole-3-carboxylique en tant qu'agonistes du recepteur couple aux proteines g (rcpg) rup38 pour traiter des troubles lies au metabolisme
JP5221952B2 (ja) 3−アリール−5,6−ジ置換ピリダジン類
CA2748289C (fr) Procede de preparation de composes d'amide de dihydroindene, compositions pharmaceutiques contenant les composes, et leur utilisation comme inhibiteur de proteines kinases
US7241792B2 (en) Fused pyrazole derivatives and methods of treatment of metabolic-related disorders thereof
CA2664398C (fr) Derives de pyrazole en tant que modulateurs du recepteur 5ht2a de la serotonine utiles dans le traitement de troubles lies a ce recepteur
EP1606282B1 (fr) Derives d'aryle et heteroaryle susbtitues tenant lieu de modulateurs du metabolisme du glucose et prophylaxie et traitement de troubles associes
US11560369B2 (en) Crystalline forms and processes for the preparation of cannabinoid receptor modulators
CN102417508A (zh) 作为新陈代谢调节剂的稠合芳基和杂芳基衍生物以及预防和治疗与其相关的病症
US20070037836A1 (en) Mch receptor antagonists
US20060122240A1 (en) Benzotriazoles and methods of prophylaxis or treatment of metabolic-related disorders thereof
KR20020062277A (ko) Nos 억제제로서의 n-헤테로시클릭 유도체
JP2008523075A (ja) 5ht2cレセプター関連障害の処置に有用な5ht2cレセプターのモジュレーターとしてのn−ビアリールピペラジン誘導体およびn−アリールヘテロアリールピペラジン誘導体
JP2012180365A (ja) 1−アリール−4−置換イソキノリン類
DE602004001134T2 (de) Tetrazolderivate und verfahren zur behandlung von stoffwechselbedingten erkrankungen damit
JP2022527925A (ja) 芳香族アミン系化合物と、該化合物のarおよびbrd4二重阻害剤ならびに調節剤の製造における使用
DE60218099T2 (de) 2-Cyclohexyl-4-phenyl-1H-imidazolderivate als Liganden für den Neuropeptid Y5 Rezeptor
TW200845963A (en) Compounds with a combination of cannabinoid-CB1 antagonism and serotonin reuptake inhibition
EP1551403B1 (fr) Derives d'acide 2h-pyrazole-3-carboxylique substitue en 5 utilises en tant qu'agents antilipotyques pour le traitement de troubles metaboliques, tels que la dyslipidemie
CN105228609A (zh) 新的转录因子调节剂
US20070032537A1 (en) 5-Substituted 2h-pyrazole-3-carboxylic acid derivatives as agonists for the acid receptor rup25 for the treatment of dyslipidemia and related diseases
EP1701947B1 (fr) Derives d'acide carboxylique 4-oxo-4,5-dihydrofurane-2 et methodes pour traiter des troubles du metabolisme
CN117143019A (zh) 用作β2-肾上腺素受体别构拮抗剂的吡唑酰胺类衍生物
WO2014074700A1 (fr) Antagonistes du récepteur gpr119 et traitement de troubles associés à ceux-ci
TW200916442A (en) 4, 5-dihydro-(1H)-pyrazole derivatives as cannabinoid CB1receptor modulators

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20071212

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA HR MK YU

RIN1 Information on inventor provided before grant (corrected)

Inventor name: WEBB, PETER

Inventor name: SEMPLE, GRAEME

Inventor name: SKINNER, PHILIP, J.

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1109894

Country of ref document: HK

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20090114

REG Reference to a national code

Ref country code: HK

Ref legal event code: WD

Ref document number: 1109894

Country of ref document: HK