WO2003048108A2 - Compounds for treatment of inflammation, diabetes and related disorders - Google Patents

Compounds for treatment of inflammation, diabetes and related disorders Download PDF

Info

Publication number
WO2003048108A2
WO2003048108A2 PCT/US2002/038150 US0238150W WO03048108A2 WO 2003048108 A2 WO2003048108 A2 WO 2003048108A2 US 0238150 W US0238150 W US 0238150W WO 03048108 A2 WO03048108 A2 WO 03048108A2
Authority
WO
WIPO (PCT)
Prior art keywords
optionally substituted
aryl
alkyl
phenoxy
phenyl
Prior art date
Application number
PCT/US2002/038150
Other languages
French (fr)
Other versions
WO2003048108A3 (en
Inventor
Partha Neogi
Debendranath Dey
Ta-Kai Li
Joseph Fuller
Liang Chen
Original Assignee
Theracos, 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
Priority to KR1020047008107A priority Critical patent/KR100941197B1/en
Application filed by Theracos, Inc filed Critical Theracos, Inc
Priority to EP02804467A priority patent/EP1448515A2/en
Priority to MXPA04005168A priority patent/MXPA04005168A/en
Priority to NZ533645A priority patent/NZ533645A/en
Priority to CA2468302A priority patent/CA2468302C/en
Priority to CN028271009A priority patent/CN1615295B/en
Priority to AU2002357032A priority patent/AU2002357032B2/en
Priority to US10/430,677 priority patent/US7323496B2/en
Publication of WO2003048108A2 publication Critical patent/WO2003048108A2/en
Publication of WO2003048108A3 publication Critical patent/WO2003048108A3/en
Priority to US12/004,064 priority patent/US20080103302A1/en
Priority to US12/004,039 priority patent/US20080108825A1/en
Priority to US12/004,075 priority patent/US20080188654A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom 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
    • C07D213/62Oxygen or sulfur atoms
    • C07D213/63One oxygen atom
    • C07D213/64One oxygen atom attached in position 2 or 6
    • C07D213/6432-Phenoxypyridines; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • 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
    • A61P3/06Antihyperlipidemics
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C233/00Carboxylic acid amides
    • C07C233/01Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C233/16Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by singly-bound oxygen atoms
    • C07C233/24Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by singly-bound oxygen atoms with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by a carbon atom of a six-membered aromatic ring
    • C07C233/25Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by singly-bound oxygen atoms with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by a carbon atom of a six-membered aromatic ring having the carbon atom of the carboxamide group bound to a hydrogen atom or to a carbon atom of an acyclic saturated carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C235/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms
    • C07C235/02Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton
    • C07C235/28Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton the carbon skeleton being acyclic and unsaturated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C235/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms
    • C07C235/02Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton
    • C07C235/32Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton the carbon skeleton containing six-membered aromatic rings
    • C07C235/34Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton the carbon skeleton containing six-membered aromatic rings having the nitrogen atoms of the carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C235/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms
    • C07C235/42Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings and singly-bound oxygen atoms bound to the same carbon skeleton
    • C07C235/44Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings and singly-bound oxygen atoms bound to the same carbon skeleton with carbon atoms of carboxamide groups and singly-bound oxygen atoms bound to carbon atoms of the same non-condensed six-membered aromatic ring
    • C07C235/56Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to carbon atoms of six-membered aromatic rings and singly-bound oxygen atoms bound to the same carbon skeleton with carbon atoms of carboxamide groups and singly-bound oxygen atoms bound to carbon atoms of the same non-condensed six-membered aromatic ring having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C237/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups
    • C07C237/02Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C237/22Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton having nitrogen atoms of amino groups bound to the carbon skeleton of the acid part, further acylated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C271/00Derivatives of carbamic acids, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups
    • C07C271/06Esters of carbamic acids
    • C07C271/08Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms
    • C07C271/10Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atoms of the carbamate groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C271/16Esters of carbamic acids having oxygen atoms of carbamate groups bound to acyclic carbon atoms with the nitrogen atoms of the carbamate groups bound to hydrogen atoms or to acyclic carbon atoms to carbon atoms of hydrocarbon radicals substituted by singly-bound oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C271/00Derivatives of carbamic acids, i.e. compounds containing any of the groups, the nitrogen atom not being part of nitro or nitroso groups
    • C07C271/62Compounds containing any of the groups, X being a hetero atom, Y being any atom, e.g. N-acylcarbamates
    • C07C271/64Y being a hydrogen or a carbon atom, e.g. benzoylcarbamates
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C275/00Derivatives of urea, i.e. compounds containing any of the groups, the nitrogen atoms not being part of nitro or nitroso groups
    • C07C275/46Derivatives of urea, i.e. compounds containing any of the groups, the nitrogen atoms not being part of nitro or nitroso groups containing any of the groups, X being a hetero atom, Y being any atom, e.g. acylureas
    • C07C275/48Y being a hydrogen or a carbon atom
    • C07C275/50Y being a hydrogen or an acyclic carbon atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C281/00Derivatives of carbonic acid containing functional groups covered by groups C07C269/00 - C07C279/00 in which at least one nitrogen atom of these functional groups is further bound to another nitrogen atom not being part of a nitro or nitroso group
    • C07C281/06Compounds containing any of the groups, e.g. semicarbazides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C317/00Sulfones; Sulfoxides
    • C07C317/16Sulfones; Sulfoxides having sulfone or sulfoxide groups and singly-bound oxygen atoms bound to the same carbon skeleton
    • C07C317/22Sulfones; Sulfoxides having sulfone or sulfoxide groups and singly-bound oxygen atoms bound to the same carbon skeleton with sulfone or sulfoxide groups bound to carbon atoms of six-membered aromatic rings of the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/24Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D213/54Radicals substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/56Amides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom 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
    • C07D213/72Nitrogen atoms
    • C07D213/75Amino or imino radicals, acylated by carboxylic or carbonic acids, or by sulfur or nitrogen analogues thereof, e.g. carbamates
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/16Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms acylated on ring nitrogen atoms
    • C07D295/18Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms acylated on ring nitrogen atoms by radicals derived from carboxylic acids, or sulfur or nitrogen analogues thereof
    • C07D295/182Radicals derived from carboxylic acids
    • C07D295/185Radicals derived from carboxylic acids from aliphatic carboxylic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/12Systems containing only non-condensed rings with a six-membered ring
    • C07C2601/14The ring being saturated

Definitions

  • R ⁇ , R 2 , R 3 , R4, R5, R ⁇ and R are each independently selected from the group consisting of
  • R12, R13, Ri8, R19 and R 2 o are each independently selected from the group consisting of
  • R15, Ri6, and R ⁇ are each independently selected from the group consisting of
  • Alkoxycarbonyl alone or in combination, means a radical of the type
  • these compounds are useful for treatment of disorders associated with insulin resistance, such as polycystic ovary syndrome, and for the treatment of inflammation, inflammatory and immunological diseases, particularly those mediated by pro-inflammatory cytokines (such as TNF-alpha, IL-1 beta and IL- 6), type 4 phosphodiesterase (PDE4), type 3 phosphodiesterase (PDE3), p44/42 mitogen activated protein (MAP) kinase, cyclooxygenase-2 (COX-2) and/or inducible nitric oxide synthase (iNOS).
  • pro-inflammatory cytokines such as TNF-alpha, IL-1 beta and IL- 6
  • PDE4 type 4 phosphodiesterase
  • PDE3 type 3 phosphodiesterase
  • MAP mitogen activated protein
  • COX-2 cyclooxygenase-2
  • iNOS inducible nitric oxide synthase
  • the invention discloses compounds of the Formulas l-XIII
  • R 8 and Rg are each independently selected from the group consisting of
  • R"' may be H or optionally substituted C1-C20 alkyl, optionally substituted C 2 -C 2 o alkenyl, optionally substituted Cr C 20 acyl, optionally substituted C1-C20 acyloxy and optionally substituted C ⁇ - C 20 alkoxycarbonyl;
  • Z is CRdR e Rf where Rd, R e and R f are each independently selected from the group consisting of
  • Q is NR b R c where Rb and R c are independently selected from the group consisting of
  • the compounds of the invention are useful for the treatment of diabetes, characterized by the presence of elevated blood glucose levels, that is, hyperglycemic disorders such as diabetes mellitus, including both type 1 and 2 diabetes, as well as other hyperglycemic related disorders such as obesity, increased cholesterol, hyperiipidemia such as hypertriglyceridemia, kidney related disorders and the like.
  • hyperglycemic disorders such as diabetes mellitus, including both type 1 and 2 diabetes, as well as other hyperglycemic related disorders such as obesity, increased cholesterol, hyperiipidemia such as hypertriglyceridemia, kidney related disorders and the like.
  • the compounds are also useful for the treatment of disorders linked to insulin resistance and/or hyperinsulinemia, which include, in addition to diabetes, hyperandrogenic conditions such as polycystic ovary syndrome (Ibanez et al., J.
  • the compounds of this invention may be used in formulations using acceptable pharmaceutical vehicles for enteral, or parenteral, administration, such as, for example, water, alcohol, gelatin, gum arabic, lactose, amylase, magnesium stearate, talc, vegetable oils, polyalkylene glycol, and the like.
  • acceptable pharmaceutical vehicles for enteral, or parenteral, administration such as, for example, water, alcohol, gelatin, gum arabic, lactose, amylase, magnesium stearate, talc, vegetable oils, polyalkylene glycol, and the like.
  • the compounds can be formulated in solid form, e.g., as tablets, capsules, drages and suppositories, or in the liquid form, e.g., solutions, suspensions and emulsions.
  • the preparations may also be delivered transdermally or by topical application.
  • Scheme 1 details the synthesis of compounds 1-6.
  • Scheme 2 details the synthesis of 17. It is to be understood that the Schemes 1 and 2 are representative schemes and are not intended to be limited to the compounds disclosed. SCHEME II
  • Step 1 Synthesis of 3-(3, ⁇ -dimethoxyphenyl)-2-(4-hvdroxyphenyl)- acrylic acid (2).
  • 3, ⁇ -dimethoxybenzaldehyde 120 g, 0.72 mol
  • p-hydroxyphenyl acetic acid 110 g, 0.72 mol
  • acetic anhydride 240 mL
  • triethylamine 161 mL, 1.6 equiv.
  • Step 2 Synthesis of 3-(3, ⁇ -dimethoxyphenyl)-2-r4-(4- formylphenoxy)-phenyl1-acrylic acid (3). 2 (64.0 g, 0.21 mol) was dissolved in 320 mL anhydrous DMSO under nitrogen, and potassium tetf-butoxide (48.0 g, 0.43 mol) was added in lots. When the solution became homogenous, p- fluorobenzaldehyde (27 mL, 0.22 mol) was added and the mixture was heated at 100°C for ⁇ hr. After cooling to room temperature, the solution was poured into 1 L water and extracted with ether (2 x 600 mL).
  • CDI intermediate of 38 was converted to 40 by reacting it with morpholine in 94% yield.
  • the effect of treatment with 1 on glucose uptake was measured in 3T3- L1 differentiated adipocytes.
  • the assay was conducted essentially according to the method of Tafuri SR, Endocrinology, 137, 4706-4712 (1996).
  • the adipocytes were incubated with different concentrations of the test compound for 48 hours in Dulbecco's modified Eagle's medium (DMEM) containing 10% fetal bovine serum (FBS), then washed and incubated in glucose-free, serum-free medium for 60 minutes at 37°C. Then 14 C-deoxyglucose was added and the cells were incubated for 30 minutes at room temperature.
  • DMEM Dulbecco's modified Eagle's medium
  • FBS fetal bovine serum
  • Glucose uptake was calculated as a percentage of the basal level seen in cells not treated with drug. As shown in FIG. 1 , treatment with 1 resulted in a dose-dependent increase in glucose uptake.
  • mice When signs of arthritis appeared, mice were assigned into four treatment groups: vehicle control (0.6% carboxymethylcellulose (CMC)); compound 31 (40 mg/kg suspension in CMC); compound 31 (100 mg/kg in CMC); positive control (dexamethasone; ⁇ mg/kg).
  • CMC carboxymethylcellulose
  • the animals were dosed per oral by gavage, twice daily for 14 days, at a dose volume of 260 ⁇ l per mouse per dose.
  • the study was scored blindly to the different treatment groups. Mice were weighed and arthritis was scored three times a week. Arthritis was scored as a count of affected limbs and digits, evaluated as: erythema and swelling of tarsal, the ankle to the metatarsal joints, up to restriction of movement and deformity of the joints.

Abstract

Novel acyl urea, thiourea, carbamate, thiocarbamate and related compounds are provided which are effective in inhibiting the cytokine-mediated inflammatory response in cultured cells, in ameliorating bone destruction, in an animal model of arthritis and in lowering blood glucose levels in animal models of Type II diabetes mellitus. The compounds are disclosed as useful for a variety of treatments including the treatment of diabetes mellitus, insulin resistance, inflammation, inflammatory diseases, immunological diseases and cancer.

Description

COMPOUNDS FOR TREATMENT OF INFLAMMATION, DIABETES AND
RELATED DISORDERS
CROSS REFERENCE TO RELATED APPLICATION
[0001] This application claims priority from U.S. Provisional Application No.
60/334,818, filed November 29, 2001 , and is incorporated herein, in its entirety, by reference.
FIELD OF THE INVENTION [0002] The invention is directed to compounds, for example, heterocyclic derivatives of acyl urea, thiourea, carbamate and thiocarbamate compounds, that provide a variety of useful pharmacological effects. The compounds are useful, for example, in lowering blood glucose levels in hyperglycemic disorders, such as diabetes mellitus, and for treating related disorders, such as obesity and hyperlipidemia. Furthermore, these compounds are useful for treatment of disorders associated with insulin resistance, such as polycystic ovary syndrome, and for the treatment of inflammation, inflammatory and immunological diseases, particularly those mediated by pro-inflammatory cytokines (such as TNF-alpha, IL-1 beta and IL- 6), type 4 and type 3 phosphodiesterase (PDE4 and PDE3, respectively), p44/42 mitogen-activated protein (MAP) kinase, cyclooxygenase-2 (COX-2) and/or inducible nitric oxide synthase (iNOS).
BACKGROUND [0003] The causes of diabetes mellitus are not yet known, although both genetics and environment seem to be major factors. Type 1 diabetes, also known as insulin-dependent diabetes mellitus (IDDM), is an autoimmune disease in which the responsible autoantigen is still unidentified. Since their insulin-producing pancreatic cells are destroyed, Type 1 diabetics need to take insulin parenterally to survive. On the other hand, type 2 diabetes, also called non-insulin-dependent diabetes mellitus (NIDDM), the more common form, is a metabolic disorder resulting from the body's inability to make a sufficient amount of insulin or to properly use the insulin that is produced. Impaired insulin secretion and insulin resistance are considered the major defects; however, the precise genetic factors involved in the mechanism remain unknown.
[0004] Other than insulin administered parenterally and as shown in Table 1 , there are generally four major classes of oral hypoglycemic agents currently used in the treatment of diabetes mellitus:
Figure imgf000003_0001
TABLE 1
[0005] As is shown in the above table, each of the current agents available for use in treatment of diabetes mellitus has several disadvantages. Accordingly, there is a need for the identification and development of new agents, particularly, water soluble agents which can be orally administered, for use in the treatment of diabetes mellitus and other hyperglycemic disorders.
[0006] Moreover, while the thiazolidinedione class has gained more widespread use in recent years as insulin sensitizers to combat "insulin resistance", a condition in which the patient becomes less responsive to the effects of insulin, there is a need for frequent liver testing for patients using these compounds. In fact, the known thiazolidinediones are not effective for a significant portion of the patient population. In addition, the first drug in this class to be approved by the FDA, troglitazone, was withdrawn from the market due to problems of liver toxicity. Thus, there is a continuing need for nontoxic, more widely effective insulin sensitizers.
[0007] As indicated above, the invention is also directed to the treatment of immunological diseases or inflammation, in particular, such diseases as are mediated by cytokines, COX-2 and iNOS. The principal elements of the immune system are macrophages or antigen-presenting cells, T cells and B cells. Macrophages are important mediators of inflammation and also provide the necessary "help" for T cell stimulation and proliferation. For example, macrophages make the cytokines IL-1 , IL-12 and TNF-alpha, all of which are potent pro-inflammatory molecules. Cytokine production may lead to the secretion of other cytokines, altered cellular function, cell division or differentiation. In addition, activation of macrophages results in the induction of enzymes, such as COX-2 and iNOS, and in the production of free radicals capable of damaging normal cells. Many factors activate macrophages, including bacterial products, superantigens and interferon gamma. It is believed that phosphotyrosine kinases and other cellular kinases are involved in the activation process. Since macrophages are sentinel to the development of an immune response, agents that modify their function, specifically their cytokine secretion profile, are likely to determine the direction and potency of the immune response.
[0008] Inflammation is the body's normal response to injury or infection.
However, in inflammatory diseases such as rheumatoid arthritis, pathologic inflammatory processes can lead to morbidity and mortality. The cytokine tumor necrosis factor-alpha (TNF-alpha) plays a central role in the inflammatory response and has been targeted as a point of intervention in inflammatory disease. TNF-alpha is a polypeptide hormone released by activated macrophages and other cells. At low concentrations, TNF-alpha participates in the protective inflammatory response by activating leukocytes and promoting their migration to extravascular sites of inflammation (Moser et al., J Clin Invest, 83:444-56, 1989). At higher concentrations, TNF-alpha can act as a potent pyrogen and induce the production of other pro- inflammatory cytokines (Haworth et al., Eur J Immunol, 21 :2576-79, 1991 ; Brennan et al., Lancet, 2:244-7, 1989). TNF-alpha also stimulates the synthesis of acute- phase proteins. In rheumatoid arthritis, a chronic and progressive inflammatory disease affecting about 1% of the adult U.S. population, TNF-alpha mediates the cytokine cascade that leads to joint damage and destruction (Arend et al., Arthritis Rheum, 38:151-60, 1995). Inhibitors of TNF-alpha, including soluble TNF receptors (etanercept) (Goldenberg, Clin Ther, 21 :75-87, 1999) and anti-TNF-alpha antibody (infliximab) (Luong et al., Ann Pharmacother, 34:743-60, 2000), the contents of each of which are incorporated herein by reference, have recently been approved by the U.S. Food and Drug Administration (FDA) as agents for the treatment of rheumatoid arthritis.
[0009] Elevated levels of TNF-alpha have also been implicated in many other disorders and disease conditions, including cachexia (Fong et al., Am J Physiol, 256:R659-65, 1989), septic shock syndrome (Tracey et al., Proc Soc Exp Biol Med, 200:233-9, 1992), osteoarthritis (Venn et al., Arthritis Rheum, 36:819-26, 1993), inflammatory bowel disease such as Crohn's disease and ulcerative colitis (Murch et al., Gut, 32:913-7, 1991), Behcet's disease (Akoglu et al., J Rheumatol, 17:1107-8, 1990), Kawasaki disease (Matsubara et al., Clin Immunol Immunopathol, 56:29-36, 1990), cerebral malaria (Grau et al., N Engl J Med, 320:1586-91 , 1989), adult respiratory distress syndrome (Millar et al., Lancet 2:712-4, 1989), asbestosis and silicosis (Bissonnette et al., Inflammation, 13:329-39, 1989), pulmonary sarcoidosis (Baughman et al., J Lab Clin Med, 115:36-42, 1990), asthma (Shah et al., Clin Exp Allergy, 25:1038-44, 1995), AIDS (Dezube et al., J Acquir Immune Defic Syndr, 5:1099-104, 1992), meningitis (Waage et al., Lancet, 1 :356-7, 1987), psoriasis (Oh et al., J Am Acad Dermatol, 42:829-30, 2000), spondyloarthritides such as ankylosing spondylitis (Braun et al., Curr Opin Rheumatol 13:245-9, 2001 ; Marzo- Ortega et al., Arthritis Rheum 44:2112-7, 2001), graft versus host reaction (Nestel et al., J Exp Med, 175:405-13, 1992), multiple sclerosis (Sharief et al., N Engl J Med, 325:467-72, 1991), systemic lupus erythematosus (Maury et al., Int J Tissue React, 11:189-93, 1989), diabetes (Hotamisligil et al., Science, 259:87-91 , 1993) and atherosclerosis (Bruunsgaard et al., Clin Exp Immunol, 121:255-60, 2000), the contents of each of which are incorporated herein by reference. It can be seen from the references cited above that inhibitors of TNF-alpha are potentially useful in the treatment of a wide variety of diseases.
[00010] lnterleukin-6 (IL-6) is another pro-inflammatory cytokine that exhibits pleiotropy and redundancy of action. IL-6 participates in the immune response, inflammation and hematopoiesis. It is a potent inducer of the hepatic acute phase response and is a powerful stimulator of the hypothalamic-pituitary-adrenal axis that is under negative control by glucocorticoids. IL-6 promotes the secretion of growth hormone but inhibits release of thyroid stimulating hormone. Elevated levels of IL-6 are seen in several inflammatory diseases, and inhibition of the IL-6 cytokine subfamily has been suggested as a strategy to improve therapy for rheumatoid arthritis (Carroll et al., Inflamm Res, 47:1-7, 1998). In addition, IL-6 has been implicated in the progression of atherosclerosis and the pathogenesis of coronary heart disease (Yudkin et al., Atherosclerosis, 148:209-14, 1999). Overproduction of IL-6 is also seen in steroid withdrawal syndrome, conditions related to deregulated vasopressin secretion, and osteoporosis associated with increased bone resorption, such as in cases of hyperparathyroidism and sex-steroid deficiency (Papanicolaou et al., Ann Intern Med, 128:127-37, 1998). Since excessive production of IL-6 is implicated in several disease states, it is highly desirable to develop compounds that inhibit IL-6 secretion.
[00011] The cytokine IL-1 beta also participates in the inflammatory response.
It stimulates thymocyte proliferation, fibroblast growth factor activity, and the release of prostaglandin from synovial cells. Elevated or unregulated levels of the cytokine IL-1 beta have been associated with a number of inflammatory diseases and other disease states, including but not limited to adult respiratory distress syndrome (Meduri et al, Chest 107:1062-73, 1995), allergy (Hastie et al, Cytokine 8:730-8, 1996), Alzheimer's disease (O'Barr et al, J Neuroimmunol 109:87-94, 2000), anorexia (Laye et al, Am J Physiol Regul Integr Comp Physiol 279:R93-8, 2000), asthma (Sousa et al, Thorax 52:407-10, 1997), atherosclerosis (Dewberry et al, Arterioscler Thromb Vase Biol 20:2394-400, 2000), brain tumors (llyin et al, Mol Chem Neuropathol 33:125-37, 1998), cachexia (Nakatani et al, Res Commun Mol Pathol Pharmacol 102:241-9, 1998), carcinoma (Ikemoto et al, Anticancer Res 20:317-21 , 2000), chronic arthritis (van den Berg et al, Clin Exp Rheumatol 17:S105- 14, 1999), chronic fatigue syndrome (Cannon et al, J Clin Immunol 17:253-61 , 1997), CNS trauma (Herx et al, J Immunol 165:2232-9, 2000), epilepsy (De Simoni et al, Eur J Neurosci 12:2623-33, 2000), fibrotic lung diseases (Pan et al, Pathol Int 46:91- 9, 1996), fulminant hepatic failure (Sekiyama et al, Clin Exp Immunol 98:71-7, 1994), gingivitis (Biesbrock et al, Monogr Oral Sci 17:20-31 , 2000), glomerulonephritis (Kluth et al, J Nephrol 12:66-75, 1999), Guillain-Barre syndrome (Zhu et al, Clin Immunol Immunopathol 84:85-94, 1997), heat hyperalgesia (Opree et al, J Neurosci 20:6289-93, 2000), hemorrhage and endotoxemia (Parsey et al, J Immunol 160:1007-13, 1998), inflammatory bowel disease (Olson et al, J Pediatr Gastroenterol Nutr 16:241-6, 1993), leukemia (Estrov et al, Leuk Lymphoma 24:379- 91 , 1997), leukemic arthritis (Rudwaleit et al, Arthritis Rheum 41 :1695-700, 1998), systemic lupus erythematosus (Mao et al, Autoimmunity 24:71-9, 1996), multiple sclerosis (Martin et al, J Neuroimmunol 61 :241-5, 1995), osteoarthritis (Hernvann et al, Osteoarthritis Cartilage 4:139-42, 1996), osteoporosis (Zheng et al, Maturitas 26:63-71, 1997), Parkinson's disease (Bessler et al, Biomed Pharmacother 53:141-6, 1999), POEMS syndrome (Gherardi et al, Blood 83:2587-93, 1994), pre-term labor (Dudley, J Reprod Immunol 36:93-109, 1997), psoriasis (Bonifati et al, J Biol Regul Homeost Agents 11 :133-6, 1997), reperfusion injury (Clark et al, J Surg Res 58:675- 81 , 1995), rheumatoid arthritis (Seitz et al, J Rheumatol 23:1512-6, 1996), septic shock (van Deuren et al, Blood 90:1101-8, 1997), systemic vasculitis (Brooks et al, Clin Exp Immunol 106:273-9, 1996), temporal mandibular joint disease (Nordahl et al, Eur J Oral Sci 106:559-63, 1998), tuberculosis (Tsao et al, Tuber Lung Dis 79:279-85, 1999), viral rhinitis (Roseler et al, Eur Arch Otorhinolaryngol Suppl 1:S61- 3, 1995), the contents of each of which are incorporated herein by reference, and pain and/or inflammation resulting from strain, sprain, trauma, surgery, infection or other disease processes. Since overproduction of IL-1 beta is associated with numerous disease conditions, it is desirable to develop compounds that inhibit the production or activity of IL-1 beta. [00012] Cyclooxygenase is an enzyme that catalyzes a rate-determining step in the biosynthesis of prostaglandins, which are important mediators of inflammation and pain. The enzyme occurs as at least two distinct isomers, cyclooxygenase-1 (COX-1) and cyclooxygenase-2 (COX-2). The COX-1 isomer is constitutively expressed in the gastric mucosa, platelets and other cells and is involved in the maintenance of homeostasis in mammals, including protecting the integrity of the digestive tract. The COX-2 isomer, on the other hand, is not constitutively expressed but rather is induced by various agents, such as cytokines, mitogens, hormones and growth factors. In particular, COX-2 is induced during the inflammatory response (DeWitt DL, Biochim Biophys Acta, 1083:121-34, 1991 ; Seibert et al., Receptor, 4:17-23, 1994.). Aspirin and other conventional non-steroid anti-inflammatory drugs (NSAIDs) are non-selective inhibitors of both COX-1 and COX-2. They can be effective in reducing inflammatory pain and swelling, but since they hamper the protective action of COX-1 , they produce undesirable side effects of gastrointestinal pathology. Therefore, agents that selectively inhibit COX-2 but not COX-1 are preferable for treatment of inflammatory diseases. Recently, a diarylpyrazole sulfonamide (celecoxib) that selectively inhibits COX-2 has been approved by the FDA for use in the treatment of osteoarthritis and adult rheumatoid arthritis (Luong et al., Ann Pharmacother, 34:743-60, 2000; Penning et al., J Med Chem, 40:1347-65, 1997). Another selective COX-2 inhibitor, rofecoxib, has been approved by the FDA for treatment of osteoarthritis, acute pain and primary dysmenorrhea (Scott and Lamb, Drugs, 58:499-606, 1999; Morrison et al., Obstet Gynecol, 94:504-8, 1999; Saag et al, Arch Fam Med, 9:1124-34, 2000). COX-2 is also expressed in many cancers and precancerous lesions, and there is mounting evidence that selective COX-2 inhibitors may be useful for treating and preventing colorectal, breast and other cancers (Taketo MM, J Natl Cancer Inst, 90:1609-20, 1998; Foumier et al., J Cell Biochem Suppl, 34:97-102, 2000; Masferrer et al., Cancer Res, 60:1306-11 , 2000), the contents of each of which are incorporated herein by reference. In 1999 celecoxib was approved by the FDA as an adjunct to usual care for patients with familial adenomatous polyposis, a condition which, left untreated, generally leads to colorectal cancer.
[00013] Production of nitric oxide by iNOS has been associated with both beneficial and detrimental effects in inflammation, inflammatory diseases and related disorders. For example, deleterious effects have been implicated in the pathogenesis of abdominal aortic aneurysms (Johanning et al, J Vase Surg 33:579-86, 2001), acute endotoxemia (Henningsson et al, Am J Physiol Cell Physiol 280:C1242-54, 2001), amyotrophic lateral sclerosis (Sasaki et al, Neurosci Lett 291 :44-8, 2000), atherosclerosis (Behr-Roussel et al, Circulation 102:1033-8, 2000), bladder cancer (Wolf et al, Virchows Arch 437:662-6, 2000), colon cancer (Watanabe et al, Biofactors 12:129-33, 2000), cystitis (Alfieri et al, Naunyn Schmiedebergs Arch Pharmacol 363:353-7, 2001), HIV-1 encephalitis (Zhao et al, J Neuroimmunol 115:182-91 , 2001), inflammatory bowel disease (Singer et al, Gastroenterology 111 :871-85, 1996), multiple sclerosis (Pozza et al, Brain Res 855:39-46, 2000), osteoarthritis (Pelletier et al, Osteoarthritis Cartilage 7:416-8, 1999), osteoporosis (Armour et al, J Bone Miner Res 14:2137-42, 1999), portal hypertension (Schroeder et al, Dig Dis Sci Dec 45:2405-10, 2000), pulmonary edema in endotoxin shock (Lee et al, Clin Exp Pharmacol Physiol 28:315-20, 2001), rheumatoid arthritis (van't Hof et al, Rheumatology (Oxford) 39:1004-8, 2000), sepsis (Nishina et al, Anesth Analg 92:959-66, 2001), severe burn/smoke inhalation injury (Soejima et al, Am J Respir Crit Care Med 163:745-52, 2001), and ulcerative colitis (Ikeda et al, Am J Gastroenterol 92:1339-41 , 1997), the contents of each of which are incorporated herein by reference. Since the production of nitric oxide by iNOS has been implicated in the pathogenesis of inflammatory and related immunological diseases, it is desirable to develop compounds that inhibit iNOS activity or expression.
[00014] Phosphodiesterases (PDEs) are responsible for the hydrolysis of intracellular cyclic adenosine and guanosine monophosphate (cAMP and cGMP), which converts these second messengers into their inactive forms. There are 11 major families of PDEs, designated PDE1 to PDE11. Type 4 phosphodiesterase (PDE4) is found in airway smooth muscle cells and in immune and inflammatory cells. PDE4 activity has been associated with a wide variety of inflammatory and autoimmune diseases, and PDE4 inhibitors have been studied as potential therapeutic agents for such diseases as asthma, chronic obstructive pulmonary disease, rheumatoid arthritis, multiple sclerosis and type 2 diabetes (Burnouf and Pruniaux, Current Pharm Des, 8:1256-96, 2002; Dal Piaz and Giovannoni, Eur J Med Chem, 35:463-80, 2000). Type 3 phosphodiesterase (PDE3) is localized in platelets and cardiac and vascular smooth muscle cells. Inhibitors of PDE3 have been proposed as possible drugs for the treatment of acute respiratory distress syndrome (Schermuly et al, J Pharmacol Exp Ther, 292:512-20, 2000), cancer (Shimizu et al, Anticancer Drugs, 13:875-80, 2002; Murata et al, Anticancer Drugs, 12:79-83, 2001), cardiomyopathy (Alharethi and Movsesian, Expert Opin Investig Drugs, 11 :1529-36, 2002), congestive heart failure (Movsesian, J Am Coll Cardiol, 34:318-24, 1999), erectile dysfunction (Kuthe et al, Curr Opin Investig Drugs, 3:1489-95, 2002), and T-cell-mediated autoimmune disorders (Bielekova et al, J Immunol 164:1117-24, 2000), the contents of each of which are incorporated herein by reference.
[00015] Activation of lymphocyte and macrophage immune response to pathogens involve complex intracellular signaling pathways involving a cascade of various phosphorylating enzymes, kinases that ultimately regulate cytokine production and cell apoptosis. Key kinases include p44/42 MAP kinase (also known as ERK1/ERK2), P38 MAP kinase, MEK, and IRAK/NFkB. While different processes utilize different aspects of the pathway, the bacterial coat-derived protein LPS has been shown to activate multiple mitogen-activated protein kinases, including the extracellular signal-regulated receptor kinases ERK1 and ERK2. LPS-induced TNF- alpha production by human monocytes involves activation of ERK1/ERK2 (van der Bruggen et al, Infect Immun, 67:3824-9, 1999). As TNF-alpha is a key mediator of autoimmune disease, blocking the ERK pathway has potential for the treatment of inflammatory and immunological diseases such as lupus (Yi et al, J Immunol, 165:6627-34, 2000), rheumatoid arthritis (Neff et al, Cell Microbiol, 3:703-12, 2001 ; Schett et al, Arthritis Rheum, 43:2501-12, 2000), psoriasis (van der Bruggen et al, Infect Immun, 67:3824-9, 1999) and destruction of pancreatic islet beta cells in Type I diabetes (Pavlovic et al, Eur Cytokine Netw 11 :267-74, 2000), the contents of each of which are incorporated herein by reference.
[00016] It will be appreciated from the foregoing that, while there have been extensive prior efforts to provide compounds for inhibiting, for example, TNF-alpha, IL-1 beta, IL-6, COX-2, PDE4 or other agents considered responsible for inflammation or inflammatory diseases, e.g. arthritis, there still remains a need for new and improved compounds for effectively treating or inhibiting such diseases. A principal object of the invention is to provide compounds which are effective for such treatments as well as for the treatment of, for example, diabetes, coronary heart disease, insulin resistance and related disorders.
SUMMARY OF THE INVENTION [00017] The invention is directed to compounds, for example, heterocyclic derivatives of acyl urea, thiourea, carbamate and thiocarbamate compounds, for providing a variety of useful pharmacological effects. The compounds area seful, for example, in lowering blood glucose levels in hyperglycemic disorders, such as diabetes mellitus, and for treating related disorders, such as obesity and hyperiipidemia. Furthermore, these compounds are useful for treatment of disorders associated with insulin resistance, such as polycystic ovary syndrome, and for the treatment of inflammation and immunological diseases, particularly those mediated by pro-inflammatory cytokines (such as TNF-alpha, IL-1 beta and IL-6), type 4 phosphodiesterase (PDE4), type 3 phosphodiesterase (PDE3), p44/42 mitogen activated protein (MAP) kinase, cyclooxygenase-2 (COX-2) and/or inducible nitric oxide synthase (iNOS). In particular, the invention discloses compounds of the Formulas l-XIII as well as the pharmaceutically acceptable salts and solvates thereof:
Figure imgf000011_0001
Figure imgf000012_0001
Figure imgf000013_0001
[00018] wherein the stereocenters marked with an asterisk (*) may be R- or S-; the bond represented by a dashed line plus a solid line may be a double bond or a single bond, and when the bond is a double bond it may be in the E or Z configuration, and when the bond is a single bond the resulting stereocenters may have the R- or S- configuration; and
[00019] Rι, R2, R3, R4, R5, Rδ and R are each independently selected from the group consisting of
H; optionally substituted C1-C20 linear or branched alkyl including chloroalkyl or fluoroalkyl; optionally substituted C2-C2o linear or branched alkenyl; optionally substituted C6-C20 aryl, linear or branched alkylaryl, linear or branched alkenylaryl; COOR where R is H, optionally substituted Cι-C20 alkyl, optionally substituted C2-C20 alkenyl or optionally substituted C6-Ci0 aryl, sodium, potassium or other pharmaceutically acceptable counter-ion such as calcium, magnesium, ammonium, tromethamine and the like; CONR'R", where R' and R" are independently H, optionally substituted C1-C20 alkyl, optionally substituted C2-C2o alkenyl or optionally substituted C6-Cιo aryl or where NR'R" represents a cyclic moiety such as morpholine, piperidine, piperazine and the like; optionally substituted Cι-C6 amidoalkyl; NH2; Cι-C20 alkylamino, bis(alkylamino), cycloalkylamino or cyclic amino; OH; optionally substituted C1-C20 alkoxy including trifluoromethoxy and the like; optionally substituted C1-C20 alkanoyl; optionally substituted C1-C20 acyloxy; halo; optionally substituted C1-C20 alkylcarboxylamino; cyano; nitro; SO2NR'"R"" where R'" and R"" are independently H, C^o alkyl or aryl; SO2R'" where R"' is H, C1-C20 alkyl or aryl; SOsR'" where R"' is H, C1-C20 alkyl or aryl; and C4-C8 heterocycles such as tetrazolyl, imidazolyl, pyrrolyl, pyridyl, indolyl and the like; and wherein when individual aromatic rings possess adjacent substituents, these substituents may be joined to form a ring such as a methylenedioxy or ethylenedioxy group, and the like, including lactones and lactams;
[00020] Rs and Rg are each independently selected from the group consisting of
H; optionally substituted C-ι-C2o linear or branched alkyl; optionally substituted C2-C20 linear or branched alkenyl; optionally substituted Cβ-Cio aryl or heteroaryl; COOR where R is H, optionally substituted C1-C20 alkyl, optionally substituted C2-C2o alkenyl or optionally substituted Cβ-Cio aryl, sodium, potassium or other pharmaceutically acceptable counter-ion such as calcium, magnesium, ammonium, tromethamine and the like; CONR'R", where R' and R" are independently H, alkoxy, optionally substituted Cι-C20 alkyl, optionally substituted C2-C20 alkenyl, optionally substituted C3-C10 cycloalkyl or cycloalkenyl or optionally substituted Cβ-C-to aryl or heteroaryl, preferably 2-, 3- or 4-pyridyl; or where NR'R" represents a cyclic moiety such as morpholine, piperidine, hydroxypiperidine, imidazole, piperazine, methylpiperazine and the like; NH2; Cι-C20 alkylamino, bis(alkylamino), cycloalkylamino or cyclic amino; OH; C1-C20 alkoxy; C-1-C20 alkanoyl; C1-C20 acyloxy; halo; C1-C20 alkylcarboxylamino; cyano; nitro; SO2NR'"R"" where R'" and R"" are independently H, C1-C20 alkyl or aryl; SO2R'" where R'" is H, Cι-C20 alkyl or aryl; SO3R'" where R'" is H, C1-C20 alkyl or aryl; and tetrazolyl; and wherein R8 and Rg together may be joined to form a C4-Cs heterocyclic ring, including lactone or lactam; [00021] Rio and Rn are each independently selected from the group consisting of
H; optionally substituted C1-C20 linear or branched alkyl; optionally substituted C2-C20 linear or branched alkenyl; optionally substituted C6-Cι0 aryl or heteroaryl; COOR where R is H, optionally substituted C1-C20 alkyl, optionally substituted C2-C2o alkenyl or optionally substituted C6-Cι0 aryl, sodium, potassium or other pharmaceutically acceptable counter-ion such as calcium, magnesium, ammonium, tromethamine and the like; CONR'R", where R' and R" are independently H, optionally substituted C1-C20 alkyl, optionally substituted C2-C20 alkenyl or optionally substituted C6-Cι0 aryl or where NR'R" represents a cyclic moiety such as morpholine, piperidine, piperazine and the like; NH2; C1-C20 alkylamino, bis(alkylamino), cycloalkylamino or cyclic amino; OH; C-1-C20 alkoxy; C1-C20 alkanoyl; C1-C20 acyloxy; halo; C1-C20 alkylcarboxylamino; cyano; nitro; SO2NR'"R"" where R'" and R"" are independently H, C1-C20 alkyl or aryl; SO2R'" where R"' is H, Cι-C20 alkyl or aryl; SO3R'" where R'" is H, C1-C20 alkyl or aryl; and tetrazolyl; and wherein R10 and Rn together may be joined to form a C4-C8 heterocyclic ring, including lactone or lactam;
[00022] R12, R13, Ri8, R19 and R2o are each independently selected from the group consisting of
H; optionally substituted C1-C20 linear or branched alkyl; optionally substituted C2-C20 linear or branched alkenyl; optionally substituted C6-C-ι0 aryl or heteroaryl; COOR where R is optionally substituted C^C^o alkyl, optionally substituted C2-C20 alkenyl or optionally substituted C6-Cι0 aryl, sodium, potassium or other pharmaceutically acceptable counter-ion such as calcium, magnesium, ammonium, tromethamine and the like; CONR'R", where R' and R" are independently H, optionally substituted C1-C20 alkyl, optionally substituted C2-C o alkenyl or optionally substituted C6-Cι0 aryl or where NR'R" represents a cyclic moiety such as morpholine, piperidine, piperazine and the like; Cι-C20 alkanoyl; C C20 alkylamido; C6-C2o aroyl or heteroaroyl; S02R"' where R1" is H, C1-C20 alkyl or aryl; morpholinocarbonylmethyl; piperazinocabonylmethyl; and piperadinocabonylmethyl; [00023] Rι2 and R13 may be absent, or R12 and R13 together may be an optionally substituted heterocyclic ring, preferably morpholine, piperidine, piperazine, and N-methyl piperidine.
[00024] Ri4 is selected from the group consisting of
H; optionally substituted C1-C20 linear or branched alkyl including chloroalkyl and fluoroalkyl; optionally substituted C2-C2o linear or branched alkenyl; optionally substituted Cβ-Cio aryl or heteroaryl; COOR where R is H, optionally substituted C1-C20 alkyl, optionally substituted C -C2o alkenyl or optionally substituted C6-Cι0 aryl, sodium, potassium or other pharmaceutically acceptable counter-ion such as calcium, magnesium, ammonium, tromethamine and the like; CONR'R", where R' and R" are independently H, optionally substituted C-i-C2o alkyl, optionally substituted C2- C2o alkenyl or optionally substituted C6-Cι0 aryl or where NR'R" represents a cyclic moiety such as morpholine, piperidine, piperazine and the like; cyano; and tetrazolyl;
[00025] R15, Ri6, and Rι are each independently selected from the group consisting of
H; optionally substituted Cι-C2o linear or branched alkyl including chloroalkyl and fluoroalkyl; optionally substituted C2-C2o linear or branched alkenyl; optionally substituted Cβ-Cio aryl or heteroaryl; COOR where R is H, optionally substituted C1-C20 alkyl, optionally substituted C2-C2o alkenyl or optionally substituted Cβ-Cio aryl, sodium, potassium or other pharmaceutically acceptable counter-ion such as calcium, magnesium, ammonium, tromethamine and the like; CONR'R", where R' and R" are independently H, optionally substituted C1-C20 alkyl, optionally substituted C2- C20 alkenyl or optionally substituted C6-Cιo aryl or where NR'R" represents a cyclic moiety such as morpholine, piperidine, piperazine and the like; NH2; C-i- C2o alkylamino, bis(alkylamino), cycloalkylamino or cyclic amino; OH; C1-C20 alkoxy; C1-C20 alkanoyl; C1-C20 acyloxy; halo; C1-C20 alkylcarboxylamino; cyano; nitro; SO2NR'"R"" where R'" and R"" are independently H, C C^ alkyl or aryl; SO2R'" where R"' is H, CrC^o alkyl or aryl; SO3R"' where R'" is H, C C2o alkyl or aryl; and tetrazolyl; [00026] X is independently selected from the group consisting of
O; N; S; S=O; SO2; or NR"1", where R""' may be H or optionally substituted C1-C20 alkyl, optionally substituted C2-C20 alkenyl, optionally substituted Cι- C20 acyl, optionally substituted C1-C20 acyloxy and optionally substituted C1- . C2o alkoxycarbonyl;
[00027] Y is independently O, S or NH;
[00028] Z is ORa where Ra is selected from the group consisting of
H; optionally substituted C1-C20 linear or branched alkyl including chloroalkyl or fluoroalkyl and the like; optionally substituted C2-C2o linear or branched alkenyl; optionally substituted C6-C10 aryl or heteroaryl; optionally substituted C6-C2o aroyl or heteroaroyl; optionally substituted C1-C20 alkanoyl; and SO2R'" where R'" is H, C C2o alkyl or aryl; or
[00029] Z is NRbRc where Rb and Rc are independently selected from the group consisting of
H; optionally substituted Cι-C2o linear or branched alkyl including chloroalkyl or fluoroalkyl and the like; optionally substituted C2-C2o linear or branched alkenyl; optionally substituted C6-C10 aryl or heteroaryl; optionally substituted C3-C10 cycloalkyl or cycloalkenyl; COOZ1 where Z-i is optionally substituted C-1-C20 alkyl, optionally substituted C2-C2o alkenyl or optionally substituted C6- C10 aryl; optionally substituted C6-C20 aroyl or heteroaroyl; optionally substituted C1-C20 alkanoyl; and SO2R'" where R"' is H, C1-C20 alkyl or aryl; and wherein Rb and Rc together may be joined to form a 3-6 membered ring such as aziridine, morpholine, piperidine, piperazine and the like; or
[00030] Z is CRdReRf where Rd, Re and Rf are each independently selected from the group consisting of
H; optionally substituted Cι-C20 linear or branched alkyl including chloroalkyl or fluoroalkyl and the like; optionally substituted C2-C2o linear or branched alkenyl; optionally substituted Cβ-Cio aryl or heteroaryl; optionally substituted C3-C10 cycloalkyl or cycloalkenyl; COOR where R is H, optionally substituted Cι-C20 alkyl, optionally substituted C2-C20 alkenyl or optionally substituted C6- C10 aryl, sodium, potassium or other pharmaceutically acceptable counter-ion such as calcium, magnesium, ammonium, tromethamine and the like; NH2; C1-C20 alkylamino, bis(alkylamino); cycloalkylamino or cyclic amino; OH; optionally substituted C -C2ø alkoxy including trifluoromethoxy and the like; optionally substituted C1-C20 alkanoyl; optionally substituted C1-C20 acyloxy; optionally substituted C6-C20 aroyl or heteroaroyl; halo; cyano; nitro; optionally substituted C1-C20 alkylcarboxylamino; SO2NR'"R"" where R'" and R"" are independently H, Cι-C20 alkyl or aryl; SO2R'" where R'" is H, C1-C20 alkyl or aryl; and SOsR'" where R'" is H, C1-C20 alkyl or aryl; and wherein Rd and Re together may be joined to form a 3-6 membered ring such as aziridine, morpholine, piperidine, piperazine and the like; and the resulting stereocenter may have the R- or S- configuration; or the grouping C(=Y)Z may represent hydrogen or R12 or may be absent.
[00031] Q is ORa where Ra is selected from the group consisting of
H; optionally substituted C C2o linear or branched alkyl including chloroalkyl or fluoroalkyl and the like; optionally substituted C2-C20 linear or branched alkenyl; optionally substituted C6-C10 aryl or heteroaryl; optionally substituted C6-C20 aroyl or heteroaroyl; optionally substituted CrC20 alkanoyl; and SO2R'" where R'" is H, C1-C20 alkyl or aryl; or
[00032] Q is NRbRc where Rb and Rc are independently selected from the group consisting of
H; optionally substituted C-t-C^o linear or branched alkyl including chloroalkyl or fluoroalkyl and the like; optionally substituted C2-C20 linear or branched alkenyl; optionally substituted C6-Cι0 aryl or heteroaryl; optionally substituted C3-C10 cycloalkyl or cycloalkenyl; COOZ1 where Z1 is optionally substituted C1-C20 alkyl, optionally substituted C2-C2o alkenyl or optionally substituted C6- C10 aryl; optionally substituted C6-C2o aroyl or heteroaroyl; optionally substituted C1-C20 alkanoyl; and SO2R'" where R"' is H, C C2o alkyl or aryl; and wherein Rb and Rc together may be joined to form a 3-6 membered ring such as aziridine, morpholine, piperidine, piperazine and the like; or
[00033] Q is SRg, SORg or SO2Rg where Rg is selected from the group consisting of
H; optionally substituted C1-C20 linear or branched alkyl including chloroalkyl or floroalkyl and the like; optionally substituted C2-C20 linear or branched alkenyl; optionally substituted C C2o acyl; optionally substituted C1-C20 alkoxycarbonyl; C2-C20 alkoxy; optionally substituted C6-Ci0 aryl or heteroaryl; and optionally substituted Cβ-Cio aroyl or heteroaroyl.
[00034] Group A is optionally substituted C2-C20 linear or branched alkenyl; optionally substituted C6-C20 aryl, linear or branched alkylaryl, linear or branched alkenylaryl; optionally substituted heteroaryls like pyridine, indole, morpholine, piperidine, piperazine, tetrazolyl and the like; COR where R is optionally substituted C1-C20 linear or branched alkyl; optionally substituted C2-C2o linear or branched alkenyl; optionally substituted C6-C2o aryl, linear or branched alkylaryl, linear or branched alkenylaryl; optionally substituted heteroaryls like pyridine, indole, morpholine, piperidine, piperazine, tetrazolyl and the like;
[00035] Group B is OH, C-ι-C2o alkoxy; SO2R where R may be H or linear or branched C1-C20 alkyl.
[00036] Group Het represents a heterocyclic ring which is pyridyl, indolyl, tetrazolyl, imidazolyl, morphonyl, piperidinyl, piperazinyl, thiophenyl or the like.
[00037] These compounds are useful for treating diabetes and other diseases linked to insulin resistance, such as coronary artery disease and peripheral vascular disease, and also for treating or inhibiting inflammation or inflammatory diseases such as inflammatory arthritides and collagen vascular diseases, which are caused by, for example, cytokines or inducible enzymes such as TNF-alpha, IL-1 , IL-6, iNOS and/or COX-2. The compounds are also useful for treating or preventing other diseases mediated by cytokines, iNOS and/or COX-2, such as cancer.
[00038] Another aspect of the invention is a method of treating diabetes and related diseases comprising the step of administering to a subject suffering from a diabetic or related condition a therapeutically effective amount of a compound of Formulas I - XIII. Additionally, the invention provides a method of treating inflammation or inflammatory diseases or diseases mediated by cytokines, iNOS, PDE4, PDE3, p44/42 MAP kinase and/or COX-2 by administering to a subject in need of such treatment an effective amount of a compound according to Formulas I - XIII. Further, pharmaceutical compositions containing a therapeutically effective amount of one or more compounds according to Formulas I - XIII together with a pharmaceutically or physiologically acceptable carrier, for use in the treatments contemplated herein, are also provided.
BRIEF DESCRIPTION OF THE DRAWINGS [00039] FIG. 1 shows a graph of the dose-dependent increase in glucose uptake in 3T3-L1 adipocytes treated with varying concentrations of a compound according to the invention.
[00040] FIG. 2 shows a graph of the enhancement of glucose uptake in 3T3-L1 adipocytes treated with a compound according to the invention in addition to varying concentrations of insulin.
[00041] FIG. 3 shows a graph of the lowering of blood glucose levels in ob/ob mice treated with a compound according to the invention.
[00042] FIGS. 4A and 4B show graphs of the lowering of serum triglycerides and free fatty acid levels, respectively, in ob/ob mice treated with a compound according to the invention. [00043] FIG. 5 shows a graph of the inhibition of LPS-induced TNF-alpha production in mouse RAW264.7 cells treated with varying concentrations of a compound according to the invention.
[00044] FIG. 6 shows a graph of the inhibition of LPS-induced IL-1 beta production in mouse RAW264.7 cells treated with varying concentrations of a compound according to the invention.
[00045] FIG. 7 shows a graph of the inhibition of LPS-induced IL-6 production in mouse RAW264.7 cells treated with varying concentrations of a compound according to the invention.
[00046] FIG. 8 shows photos of Western blots demonstrating the inhibition of
LPS-induced iNOS and COX-2 production in mouse RAW264.7 cells treated with varying concentrations of a compound according to the invention.
[00047] FIG. 9 shows a graph of median clinical scores over time demonstrating improvement of collagen induced arthritis in mice using varying concentrations of a compound according to the invention.
DETAILED DESCRIPTION OF THE INVENTION [00048] The invention is based on the discovery that the compounds described herein are useful in the treatment of diabetes and other diseases linked to insulin resistance, such as coronary artery disease and peripheral vascular disease, and also for the treatment or inhibition of inflammation or inflammatory diseases such as inflammatory arthritides and collagen vascular diseases, which are caused by, for example, cytokines or inducible enzymes such as TNF-alpha, IL-1 , IL-6, PDE4, PDE3, p44/42 MAP kinase, iNOS and/or COX-2.
Definitions
[00049] As utilized herein, the following terms, unless otherwise indicated, shall be understood to have the following meanings: [00050] "Alkyl", alone or in combination, means a straight-chain or branched- chain alkyl radical containing preferably 1-20 carbon atoms, more preferably 1-10 carbon atoms, and most preferably 1-6 carbon atoms. Exemplary alkyl radicals include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentyl, neopentyl, iso-amyl, hexyl and the like.
[00051] "Alkenyl", alone or in combination, means a straight-chain or branched- chain hydrocarbon radical having one or more double bonds, preferably 1-2 double bonds and more preferably one double bond, and containing preferably 2-20 carbon atoms, more preferably 2-10 carbon atoms, and still more preferably 2-6 carbon atoms. Exemplary alkenyl radicals include ethenyl, propenyl, 2-methylpropenyl, n- butenyl, isobutenyl, and include groups containing multiple sites of unsaturation such as 1 ,3 -butadiene and1 ,4-butadienyl and the like.
[00052] "Alkoxy", alone or in combination, means a radical of the type "R--O-" wherein R can be hydrogen, linear or branched alkyl, or linear or branched alkenyl as previously defined and "O" is an oxygen atom. Exemplary alkoxy radicals include methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, iso-butoxy, sec-butoxy, tert- butoxy and the like.
[00053] "Alkoxycarbonyl", alone or in combination, means a radical of the type
"R-O-C(O)-" wherein "R--O-" is an alkoxy radical as previously defined and "C(O)~" is a carbonyl radical. Exemplary alkoxycarbonyl groups include methoxycarbonyl and ethoxycarbonyl.
[00054] "Alkylcarboxylamino" means a group RCON(R)- where R can be independently hydrogen, linear or branched alkyl, or linear or branched alkenyl as previously defined.
[00055] "Alkanoyl", alone or in combination, means a radical of the type "R-
C(O)~" wherein "R" is an alkyl radical as previously defined and "~C(O)~" is a carbonyl radical. Exemplary alkanoyl radicals include acetyl, trifluoroacetyl, hydroxyacetyl, propionyl, butyryl, valeryl, 4-methyIvaleryl and the like. [00056] "Halo" or "halogen", alone or in combination, means chloro, bromo, fluoro or iodo radicals.
[00057] "Aryl", alone or in combination, means an aromatic carbocyclic radical containing about 6 to about 10 carbon atoms, which is optionally substituted with one or more substituents selected from alkyl, alkoxy, halogen, hydroxy, amino, azido, nitro, cyano, haloalkyl, carboxy, alkoxycarbonyl, cycloalkyl, alkanoylamino, amido, amidino, alkoxycarbonylamino, N-alkylamidino, alkylamino, dialkylamino, aminoalkyl, alkylaminoalkyl, dialkylaminoalkyl, N-alkylamido, N,N-dialkylamido, aralkoxycarbonylamino, alkylthio, alkylsulfinyl, alkylsulfonyl, oxo and the like. Exemplary aryl radicals include phenyl, o-tolyl, 4-methoxyphenyl, 2-(tert- butoxy)phenyl, 3-methyl-4-methoxyphenyl, 2-fluorophenyl, 2-chlorophenyl, 3- nitrophenyl, 3-aminophenyl, 3-acetamidophenyl, 2-amino-3-(aminomethyl)phenyl, 6- methyl-2-aminophenyl, 2-amino-3-methylphenyl, 4,6-dimethyl-2-aminophenyl, 4- hydroxyphenyl, 3-methyl-4-hydroxyphenyl, 4-(2-methoxyphenyl)phenyl, 2-amino-1- naphthyl, 2-naphthyl, 1-methyl-3-amino-2-naphthyl, 2,3-diamino-1-naphthyl, 4,8- dimethoxy-2-naphthyl and the like.
[00058] "Acyloxy" or "Acylamino" group means an oxygen or amino group, respectively, bonded to an acyl group (RCO) where R can be hydrogen, linear or branched alkyl, or linear or branched alkenyl.
[00059] "Alkylamido" means the group RN(H)CO- where R can be hydrogen, linear or branched alkyl, or linear or branched alkenyl, as previously defined.
[00060] The reference to "optionally substituted" in the definition of the compounds throughout this disclosure is intended to include any substituent which does not negatively affect the activity of the compounds. Typical substitution includes, for example, lower (C-ι-C6) alkyl; halogen such as fluoro, chloro and bromo; nitro; amino; lower alkylamino; carboxylate, lower alkyl carboxylate, hydroxy, lower alkoxy, sulfonamide, cyano, or the like.
[00061] The invention is directed to compounds, for example, heterocyclic derivatives of acyl urea, thiourea, carbamate and thiocarbamate compounds, that provide a variety of useful pharmacological effects. The compounds are useful, for example, in lowering blood glucose levels in hyperglycemic disorders, such as diabetes mellitus, and for treating related disorders, such as obesity and hyperiipidemia. Furthermore, these compounds are useful for treatment of disorders associated with insulin resistance, such as polycystic ovary syndrome, and for the treatment of inflammation, inflammatory and immunological diseases, particularly those mediated by pro-inflammatory cytokines (such as TNF-alpha, IL-1 beta and IL- 6), type 4 phosphodiesterase (PDE4), type 3 phosphodiesterase (PDE3), p44/42 mitogen activated protein (MAP) kinase, cyclooxygenase-2 (COX-2) and/or inducible nitric oxide synthase (iNOS). In particular, the invention discloses compounds of the Formulas l-XIII as well as the pharmaceutically acceptable salts and solvates thereof:
Figure imgf000024_0001
Figure imgf000025_0001
[00062] wherein the stereocenters marked with an asterisk (*) may be R- or S-; the bond represented by a dashed line plus a solid line may be a double bond or a single bond, and when the bond is a double bond it may be in the E or Z configuration, and when the bond is a single bond the resulting stereocenters may have the R- or S- configuration; and [00063] R-i, R2, R3, R4, R5, Rδ and R7 are each independently selected from the group consisting of
H; optionally substituted C1-C20 linear or branched alkyl including chloroalkyl or fluoroalkyl; optionally substituted C2-C2o linear or branched alkenyl; optionally substituted C6-C2o aryl, linear or branched alkylaryl, linear or branched alkenylaryl; COOR where R is H, optionally substituted Cι-C20 alkyl, optionally substituted C2-C2o alkenyl or optionally substituted C6-C10 aryl, sodium, potassium or other pharmaceutically acceptable counter-ion such as calcium, magnesium, ammonium, tromethamine and the like; CONR'R", where R' and R" are independently H, optionally substituted C1-C20 alkyl, optionally substituted C2-C20 alkenyl or optionally substituted C6-C10 aryl or where NR'R" represents a cyclic moiety such as morpholine, piperidine, piperazine and the like; optionally substituted Ci-Cβ amidoalkyl; NH2; Cι-C20 alkylamino, bis(alkylamino), cycloalkylamino or cyclic amino; OH; optionally substituted C1-C20 alkoxy including trifluoromethoxy and the like; optionally substituted C C2o alkanoyl; optionally substituted C1-C20 acyloxy; halo; optionally substituted C1-C20 alkylcarboxylamino; cyano; nitro; SO2NR'"R"" where R"' and R"" are independently H, C1-C20 alkyl or aryl; SO2R'" where R'" is H, C C2o alkyl or aryl; SO3R'" where R'" is H, C1-C20 alkyl or aryl; and C4-C8 heterocycles such as tetrazolyl, imidazolyl, pyrrolyl, pyridyl, indolyl and the like; or when individual aromatic rings possess adjacent substituents, these substituents may be joined to form a ring such as a methylenedioxy or ethylenedioxy group, and the like, including lactones and lactams;
[00064] R8 and Rg are each independently selected from the group consisting of
H; optionally substituted C-I-C20 linear or branched alkyl; optionally substituted C2-C20 linear or branched alkenyl; optionally substituted C6-C10 aryl or heteroaryl; COOR where R is H, optionally substituted C1-C20 alkyl, optionally substituted C2-C20 alkenyl or optionally substituted C6-Cι0 aryl, sodium, potassium or other pharmaceutically acceptable counter-ion such as calcium, magnesium, ammonium, tromethamine and the like; CONR'R", where R' and R" are independently H, alkoxy, optionally substituted C1-C20 alkyl, optionally substituted C2-C2o alkenyl, optionally substituted C3-C10 cycloalkyl or cycloalkenyl or optionally substituted Cβ-Cio aryl or heteroaryl, preferably 2-, 3- or 4-pyridyl or where NR'R" represents a cyclic moiety such as morpholine, piperidine, hydroxypiperidine, imidazole, piperazine, methylpiperazine and the like; NH2; C1-C20 alkylamino, bis(alkylamino), cycloalkylamino or cyclic amino; OH; C1-C20 alkoxy; C1-C20 alkanoyl; C1-C20 acyloxy; halo; C1-C20 alkylcarboxylamino; cyano; nitro; S02NR"'R"" where R'" and R"" are independently H, C^o alkyl or aryl; S02R"' where R'" is H, C C2o alkyl or aryl; S03R"' where R"' is H, C1-C20 alkyl or aryl; and tetrazolyl; wherein Rs and Rg together may be joined to form a C -Cs heterocyclic ring, including lactone or lactam;
[00065] R10 and Rn are each independently selected from the group consisting of
H; optionally substituted C1-C20 linear or branched alkyl; optionally substituted C2-C20 linear or branched alkenyl; optionally substituted C6-Cιo aryl or heteroaryl; COOR where R is H, optionally substituted C1-C20 alkyl, optionally substituted C2-C2o alkenyl or optionally substituted C6-Cιo aryl, sodium, potassium or other pharmaceutically acceptable counter-ion such as calcium, magnesium, ammonium, tromethamine and the like; CONR'R", where R' and R" are independently H, optionally substituted C1-C20 alkyl, optionally substituted C2-C2o alkenyl or optionally substituted Cβ-Cio aryl or where NR'R" represents a cyclic moiety such as morpholine, piperidine, piperazine and the like; NH2; C-ι-C2o alkylamino, bis(alkylamino), cycloalkylamino or cyclic amino; OH; C1-C20 alkoxy; C1-C20 alkanoyl; C1-C20 acyloxy; halo; C1-C20 alkylcarboxylamino; cyano; nitro; SO2NR'"R"" where R"' and R"" are independently H, C1-C20 alkyl or aryl; SO2R'" where R'" is H, C^o alkyl or aryl; SOsR'" where R'" is H, C1-C20 alkyl or aryl; and tetrazolyl; wherein R10 and R11 together may be joined to form a C4-Cs heterocyclic ring, including lactone or lactam;
[00066] R12, R13, Ris, R19 and R2o are each independently selected from the group consisting of H; optionally substituted C C2o linear or branched alkyl; optionally substituted C2-C20 linear or branched alkenyl; optionally substituted Cβ-Cι0 aryl or heteroaryl; COOR where R is optionally substituted C1-C20 alkyl, optionally substituted C2-C2o alkenyl or optionally substituted C6-C10 aryl, sodium, potassium or other pharmaceutically acceptable counter-ion such as calcium, magnesium, ammonium, tromethamine and the like; CONR'R", where R' and R" are independently H, optionally substituted C1-C20 alkyl, optionally substituted C2-C2o alkenyl or optionally substituted C6-C10 aryl or where NR'R" represents a cyclic moiety such as morpholine, piperidine, piperazine and the like; C1-C20 alkanoyl; C1-C20 alkylamido; C6-C20 aroyl or heteroaroyl; S02R"' where R"1 is H, C-1-C20 alkyl or aryl; morpholinocarbonylmethyl; piperazinocabonylmethyl; and piperadinocabonylmethyl;
[00067] R-12 and R13 may be absent, or R12 and R^ together may be an optionally substituted heterocyclic ring, preferably morpholine, piperidine, piperazine, and N-methyl piperidine.
[00068] R14 is selected from the group consisting of
H; optionally substituted Cι-C2o linear or branched alkyl including chloroalkyl and fluoroalkyl; optionally substituted C2-C20 linear or branched alkenyl; optionally substituted C6-Cι0 aryl or heteroaryl; COOR where R is H, optionally substituted C1-C2o alkyl, optionally substituted C2-C20 alkenyl or optionally substituted Cβ-Cio aryl, sodium, potassium or other pharmaceutically acceptable counter-ion such as calcium, magnesium, ammonium, tromethamine and the like; CONR'R", where R' and R" are independently H, optionally substituted C C2o alkyl, optionally substituted C - C20 alkenyl or optionally substituted C6-Cι0 aryl or where NR'R" represents a cyclic moiety such as morpholine, piperidine, piperazine and the like; cyano; and tetrazolyl;
[00069] Ri5> R16, and R17are each independently selected from the group consisting of
H; optionally substituted Cι-C20 linear or branched alkyl including chloroalkyl and fluoroalkyl; optionally substituted C2-C2o linear or branched alkenyl; optionally substituted C6-C10 aryl or heteroaryl; COOR where R is H, optionally substituted C1-C20 alkyl, optionally substituted C2-C20 alkenyl or optionally substituted C6-C10 aryl, sodium, potassium or other pharmaceutically acceptable counter-ion such as calcium, magnesium, ammonium, tromethamine and the like; CONR'R", where R' and R" are independently H, optionally substituted Cι-C2o alkyl, optionally substituted C2- C2o alkenyl or optionally substituted C6-Cιo aryl or where NR'R" represents a cyclic moiety such as morpholine, piperidine, piperazine and the like; NH2; Cι- C20 alkylamino, bis(alkylamino), cycloalkylamino or cyclic amino; OH; C1-C20 alkoxy; Cι-C2o alkanoyl; C1-C20 acyloxy; halo; C1-C20 alkylcarboxylamino; cyano; nitro; S02NR",RM" where R'" and R"" are independently H, C1-C20 alkyl or aryl; SO2R"' where R'" is H, Cι-C20 alkyl or aryl; SO3R'" where R"' is H, d- C2o alkyl or aryl; and tetrazolyl;
[00070] X is independently selected from the group consisting of
O; N; S; S=O; SO2; or NR"'", where R"'" may be H or optionally substituted C1-C20 alkyl, optionally substituted C2-C2o alkenyl, optionally substituted Cr C20 acyl, optionally substituted C1-C20 acyloxy and optionally substituted Cι- C20 alkoxycarbonyl;
[00071] Y is independently O, S or NH;
[00072] Z is ORa where Ra is selected from the group consisting of
H; optionally substituted C1-C20 linear or branched alkyl including chloroalkyl or fluoroalkyl and the like; optionally substituted C2-C2o linear or branched alkenyl; optionally substituted Cβ-Cio aryl or heteroaryl; optionally substituted C6-C2o aroyl or heteroaroyl; optionally substituted C1-C20 alkanoyl; and SO2R'" where R"' is H, C1-C20 alkyl or aryl; or
[00073] Z is NRbRc where Rb and Rc are independently selected from the group consisting of
H; optionally substituted C1-C20 linear or branched alkyl including chloroalkyl or fluoroalkyl and the like; optionally substituted C2-C2o linear or branched alkenyl; optionally substituted Cβ-C-io aryl or heteroaryl; COOZi where Z1 is optionally substituted C1-C20 alkyl, optionally substituted C2-C20 alkenyl or optionally substituted Cβ-Cio aryl; optionally substituted C6-C20 aroyl or heteroaroyl; optionally substituted C-i-C2o alkanoyl; and SO2R'" where R"' is H, Cι-C20 alkyl or aryl; and wherein Rb and Rc together may be joined to form a 3-6 membered ring such as aziridine, morpholine, piperidine, piperazine and the like; or
[00074] Z is CRdReRf where Rd, Re and Rf are each independently selected from the group consisting of
H; optionally substituted Cι-C20 linear or branched alkyl including chloroalkyl or fluoroalkyl and the like; optionally substituted C2-C20 linear or branched alkenyl; optionally substituted C6-Cι0 aryl or heteroaryl; COOR where R is H, optionally substituted Cι-C2o alkyl, optionally substituted C2-C20 alkenyl or optionally substituted C6-Cι0 aryl, sodium, potassium or other pharmaceutically acceptable counter-ion such as calcium, magnesium, ammonium, tromethamine and the like; NH2; C1-C20 alkylamino, bis(alkylamino); cycloalkylamino or cyclic amino; OH; optionally substituted C-1-C20 alkoxy including trifluoromethoxy and the like; optionally substituted C-i- C o alkanoyl; optionally substituted C1-C20 acyloxy; optionally substituted Cβ- C2o aroyl or heteroaroyl; halo; cyano; nitro; optionally substituted C1-C20 alkylcarboxylamino; SO2NR'"R"" where R'" and R"" are independently H, C-i- C2o alkyl or aryl; SO2R'" where R'" is H, C1-C20 alkyl or aryl; and SO3R'" where R'" is H, C1-C20 alkyl or aryl; and wherein Rd and Re together may be joined to form a 3-6 membered ring such as aziridine, morpholine, piperidine, piperazine and the like; and the resulting stereocenter may have the R- or S- configuration; or the grouping -C (=Y)Z may represent hydrogen or R12 or may be absent.
[00075] Q is ORa where Ra is selected from the group consisting of
H; optionally substituted C-ι-C2o linear or branched alkyl including chloroalkyl or fluoroalkyl and the like; optionally substituted C2-C2o linear or branched alkenyl; optionally substituted C6-Cι0 aryl or heteroaryl; optionally substituted C6-C2o aroyl or heteroaroyl; optionally substituted C1-C20 alkanoyl; and SO2R'" where R'" is H, C1-C20 alkyl or aryl; or
[00076] Q is NRbRc where Rb and Rc are independently selected from the group consisting of
H; optionally substituted C1-C20 linear or branched alkyl including chloroalkyl or fluoroalkyl and the like; optionally substituted C2-C20 linear or branched alkenyl; optionally substituted Cβ-C-io aryl or heteroaryl; optionally substituted C3-C10 cycloalkyl or cycloalkenyl; COOZ1 where Z1 is optionally substituted C1-C20 alkyl, optionally substituted C2-C2o alkenyl or optionally substituted Cβ- C10 aryl; optionally substituted C6-C20 aroyl or heteroaroyl; optionally substituted C1-C20 alkanoyl; and SO R'" where R"' is H, C1-C20 alkyl or aryl; and wherein Rb and Rc together may be joined to form a 3-6 membered ring such as aziridine, morpholine, piperidine, piperazine and the like; or
[00077] Q is SRg, SORg or SO2Rg where Rg is selected from the group consisting of
H; optionally substituted C1-C20 linear or branched alkyl including chloroalkyl or floroalkyl and the like; optionally substituted C2-C20 linear or branched alkenyl; optionally substituted C1-C20 acyl; optionally substituted C1-C20 alkoxycarbonyl; C2-C2o alkoxy; optionally substituted Cβ-Cio aryl or heteroaryl; and optionally substituted Cβ-Cio aroyl or heteroaroyl.
[00078] Group A is optionally substituted C2-C2o linear or branched alkenyl; optionally substituted C6-C2oaryl, linear or branched alkylaryl, linear or branched alkenylaryl; optionally substituted heteroaryls like pyridine, indole, morpholine, piperidine, piperazine, tetrazoly and the like; COR where R is optionally substituted C1-C20 linear or branched alkyl; optionally substituted C -C2o linear or branched alkenyl; optionally substituted C6-C2o aryl, linear or branched alkylaryl, linear or branched alkenylaryl; optionally substituted heteroaryls like pyridine, indole, morpholine, piperidine, piperazine, tetrazolyl and the like; [00079] Group B is OH, C1-C2o alkoxy; SO2R where R may be H or linear or branched C1-C20 alkyl.
[00080] Group Het represents a heterocyclic ring which is pyridyl, indolyl, tetrazolyl, imidazolyl, morphonyl, piperidinyl, piperazinyl, thiophenyl or the like.
[00081] These compounds are useful for treating diabetes and other diseases linked to insulin resistance, such as coronary artery disease and peripheral vascular disease, and also for treating or inhibiting inflammation or inflammatory diseases such as inflammatory arthritides and collagen vascular diseases, which are caused by, for example, cytokines or inducible enzymes such as TNF-alpha, IL-1 , IL-6, PDE4, PDE3, p44/42 MAP kinase, iNOS and/or COX-2. The compounds are also useful for treating or preventing other diseases mediated by cytokines, PDE4, PDE3, p44/42 MAP kinase, iNOS and/or COX-2, such as cancer.
[00082] As indicated above, the compounds of the invention include bonds, designated in Formulas I - XIII with a dashed line plus a solid line, that may be either a double bond or a single bond. When such a bond is a double bond, it may have either the E or Z configuration. On the other hand, when such a bond is a single bond, the resulting stereocenters may be in the R- and/or S- configurations. Likewise, compounds of the invention with other stereocenters, designated in Formulas I - XIII with an asterisk, may be R- and/or S- stereoisomers. The invention contemplates racemic mixtures of such stereoisomers as well as the individual, separated stereoisomers. The individual stereoisomers may be obtained by the use of an optically active resolving agent. Alternatively, a desired enantiomer may be obtained by stereospecific synthesis using an optically pure starting material of known configuration.
[00083] Generally, R- or S- refers to the configuration of the stereoisomers. The determination of whether the configuration is R- (rectus) or S- (sinister) is based on the priority of the atoms in a compound. Similarly, E- or Z- configuration is used when describing compounds with double bonds and wherein the determination is based on the priority of the atom on each carbon of a double bond.
[00084] The following compounds are representative of the preferred compounds according to Formula I:
[00085] 3-(3,5-Dimethoxyphenyl)-2-{4-[4-(3-oxo-3-ureidopropyl)- phenoxy]-phenyl}-acrylic acid methyl ester (1); [00086] 3-(3,δ-Dimethoxyphenyl)-2-{4-[4-(3-oxo-3-ureidopropyl)- phenoxy]-phenyl}-acrylic acid (6);
[00087] 3-(3,δ-Dimethoxyphenyl)-2-{4-[4-(3-ethoxycarbonylamino-3-oxo- propyl)-phenoxy]-phenyl}-acrylic acid methyl ester (8); [00088] 2-{4-[4-(3-Benzoyloxycarbonylamino-3-oxo-propyl)-phenoxy]- phenyl}-3-(3,δ-dimethoxyphenyl)-acrylic acid methyl ester (9); [00089] 3-(3,δ-Dimethoxyphenyl)-2-{4-[4-(3-oxo-3-ureidopropyl)- phenoxy]-phenyl}-propionic acid (10);
[00090] 3-(3,δ-Dimethoxyphenyl)-2-{4-[4-(3-oxo-3-ureidopropenyl)- phenoxy]-phenyl}-acrylic acid (11);
[00091] 3-(3,δ-Dimethoxyphenyl)-2-{4-[4-(3-oxo-3-ureidopropyl)- phenoxy]-phenyl}-acrylic acid ethyl ester (12); [00092] 3-(3,δ-Dimethoxyphenyl)-Λ/,Λ -dimethyl-2-{4-[4-(3-oxo-3- ureidopropyl)-phenoxy]-phenyl}-acrylamide (13);
[00093] 2-(4-{4-[3-(3-Cyclohexylureido)-3-oxopropyl]-phenoxy}-phenyl)-
3-(3,δ-dimethoxyphenyl)-acrylic acid (14).
[00094] The following are preferred compounds according to Formula II:
[3-(4-Phenoxyphenyl)-propionyl]-urea (15);
[00095] {3-[4-(4-Methoxyphenoxy)-phenyl]-acryIoyl}-urea (16).
[00096] The following are preferred compounds according to Formula III:
2-{4-[4-(3-AcetylureidomethyI)-phenoxy]-phenyl}-3-(3,δ-dimethoxyphenyl)-acrylic acid methyl ester (17);
[00097] 2-{4-[4-(3-Acetylthioureidomethyl)-phenoxy]-phenyl}-3-(3,δ- dimethoxyphenyl)-acrylic acid (18). [00098] The following are preferred compounds according to Formula IV:
1 -Acetyl-3-[4-(4-methoxyphenoxy)-benzyl]-urea (24);
[00099] Acetyl-3-[4-(3,4-dimethoxyphenoxy)-benzyl]-urea (25).
[000100] The following are more preferred compounds for their anti-inflammatory properties:
[000101] 3-(3,δ-Dimethoxyphenyl)-Λ/,Λ/-dimethyl-2-{4-[4-(3-oxo-3-ureidopropyl)- phenoxy]-phenyl}-acrylamide (13);
[000102] 2-{4-[4-(2-Carbamoylethyl)-phenoxy]-phenyl}-3-(3,δ-dimethoxyphenyl)-
N,N-dimethylacrylamide (31);
[000103] 3-(4-{4-[2-(3,δ-Dimethoxyphenyl)-1 -dimethylcarbamoylvinylj-phenoxy}- phenyl)-propionic acid ethyl ester (37);
[000104] N-{4-[2-(3,5-Dimethoxyphenyl)-1 -dimethylcarbamoylvinyl]-phenyl}-3- hydroxybenzamide (44);
[000105] 3-(3,δ-Dimethoxyphenyl)-2-(4-hydroxyphenyl)-N,N-dimethylacrylamide
(49);
[000106] [3-(4-{4-[2-(3,5-Dimethoxyphenyl)-1-(piperidine-1-carbonyl)-vinyl]- phenoxy}-phenyl)-propionyl]-urea (51 );
[000107] 2-{4-[4-(3-Acetylamino-3-oxopropyl)-phenoxy]-phenyl}-3-(4- fluorophenyl)-N,N-dimethylacrylamide (56);
[000108] 2-(4-{4-[2-(3,5-Dimethoxyphenyl)-1 -dimethylcarbamoylvinylj-phenoxy}- benzyl)-malonic acid (58); [000109] 2-(4-{4-[2-(3,δ-Dimethoxyphenyl)-1 -dimethylcarbamoylvinylj-phenoxy}- benzyl)-malonamide (59); [000110] 3-(3,δ-Dimethoxyphenyl)-N,N-dimethyl-2-[4-(pyridin-2-yloxy)-phenyl]- acrylamide (66);
[000111] N-{4-[2-(3,5-Dimethoxyphenyl)-1-dimethylcarbamoyl-vinyl]-phenyl}- benzamide (67) ;
[000112] 2-{4-[4-(1 -Dimethylcarbamoyl-2-pyridin-3-yl-vinyl)-phenoxy]-benzyl}- malonamide (71);
[000113] 3-{4-[4-(2-Benzo[1 ,3]dioxol-δ-yl-1 -dimethylcarbamoyl-vinyl)-phenoxy]- phenylj-propionic acid ethyl ester (69);
[000114] 3-Benzo[1 ,3]dioxol-5-yl-2-{4-[4-(2-carbamoylethyl)-phenoxy]-phenyl}- N,N-dimethyl-acrylamide (72); [000115] N,N-Dimethyl-2-{4-[4-(3-oxo-3-ureidopropyl)-phenoxy]-phenyl}-3- pyridin-3-yl-acrylamide (73);
[000116] The following are more preferred compounds for their antidiabetic properties:
[000117] 3-(3,5-Dimethoxyphenyl)-2-{4-[4-(3-ethoxycarbonylamino-3-oxo- propyl)-phenoxy]-phenyl}-acrylic acid methyl ester (8);
[000118] (4-{4-[2-(3,5-Dimethoxyphenyl)-1 -dimethylcarbamoyl-vinylj-phenoxy}- benzyl)-carbamic acid methyl ester (29);
[000119] 2-{4-[4-(2-Carbamoylethyl)-phenoxy]-phenyl}-3-(3,δ-dimethoxyphenyl)- N.N-dimethylacrylamide (31);
[000120] 3-(3,5-Dimethoxyphenyl)-N,N-dimethyl-2-{4-[4-(3-morpholin-4-yl-3- oxopropyl)-phenoxy]-phenyl}-acrylamide (40);
[000121] [3-(4-{4-[2-(3,δ-Dimethoxyphenyl)-1-(piperidine-1-carbonyl)-vinyl]- phenoxy}-phenyl)-propionyl]-urea (51 );
[000122] 2-{4-[4-(3-Acetylamino-3-oxopropyl)-phenoxy]-phenyl}-3-(4- fluorophenyl)-N,N-dimethylacrylamide (56);
[000123] 3-(3,5-Dimethoxyphenyl)-2-{4-[4-(3-oxo-3-ureidopropyl)- phenoxy]- phenyl}-N-pyridin-4-ylacrylamide (60);
[000124] N-(4-Chlorophenyl)-3-(3,5-dimethoxyphenyl)-2-{4-[4-(3-oxo-3- ureidopropyl)-phenoxy]-phenyl}-acrylamide (61);
[000125] 3-(3,δ-Dimethoxyphenyl)-N,N-dimethyl-2-(4-{4-[2-(2-morpholin-4-yl-2- oxoethylcarbamoyl)-ethyl]-phenoxy}-phenyl)-acrylamide (63); [000126] 3-(3,δ-Dimethoxyphenyl)-N,N-dimethyl-2-(4-{4-[3-(4-methylpiperazin-1- yl)-3-oxopropyl]-phenoxy}-phenyl)-acrylamide (64).
[000127] However, it will be appreciated that the invention also contemplates the provision and use of other compounds according to Formulas I - XIII.
[000128] The compounds according to the present invention may be combined with a physiologically acceptable carrier or vehicle to provide a pharmaceutical composition, such as, lyophilized powder in the form of tablet or capsule with various fillers and binders. Similarly, the compounds may be coadministered with other agents. Co-administration shall mean the administration of at least two agents to a subject so as to provide the beneficial effects of the combination of both agents. For example, the agents may be administered simultaneously or sequentially over a period of time. The effective dosage of a compound in the composition can be widely varied as selected by those of ordinary skill in the art and may be empirically determined. Moreover, the compounds of the present invention can be used alone or in combination with one or more additional agents depending on the indication and the desired therapeutic effect. For example, in the case of diabetes, insulin resistance and associated conditions or complications, including obesity and hyperiipidemia, such additional agent(s) may be selected from the group consisting of: insulin or an insulin mimetic, a sulfonylurea (such as acetohexamide, chlorpropamide, glimepiride, glipizide, glyburide, tolbutamide and the like) or other insulin secretagogue (such as nateglinide, repaglinide and the like), a thiazolidinedione (such as pioglitazone, rosiglitazone and the like) or other peroxisome proliferator-activated receptor (PPAR)-gamma agonist, a fibrate (such as bezafibrate, clofibrate, fenofibrate, gemfibrozol and the like) or other PPAR-alpha agonist, a PPAR-delta agonist, a biguanide (such as metformin), a statin (such as fluvastatin, lovastatin, pravastatin, simvastatin and the like) or other hydroxymethylglutaryl (HMG) CoA reductase inhibitor, an alpha-glucosidase inhibitor (such as acarbose, miglitol, voglibose and the like), a bile acid-binding resin (such as cholestyramine, celestipol and the like), a high density lipoprotein (HDL)-lowering agent such as apolipoprotein A-l (apoA1), niacin and the like, probucol and nicotinic acid. In the case of inflammation, inflammatory diseases, autoimmune disease and other such cytokine mediated disorders, the additional agent(s) may be selected from the group consisting of: a nonsteroidal anti-inflammatory drug (NSAID) (such as diclofenac, diflunisal, ibuprofen, naproxen and the like), a cyclooxygenase-2 inhibitor (such as celecoxib, rofecoxib and the like), a corticosteroid (such as prednisone, methylprednisone and the like) or other immunosuppressive agent (such as methotrexate, leflunomide, cyclophosphamide, azathioprine and the like), a disease- modifying antirheumatic drug (DMARD) (such as injectable gold, penicilliamine, hydroxychloroquine, sulfasalazine and the like), a TNF-alpha inhibitor (such as etanercept, infliximab and the like), other cytokine inhibitor (such as soluble cytokine receptor, anti-cytokine antibody and the like), other immune modulating agent (such as cyclosporin, tacrolimus, rapamycin and the like) and a narcotic agent (such as hydrocodone, morphine, codeine, tramadol and the like). The combination therapy contemplated by the invention includes, for example, administration of the inventive compound and additional agent(s) in a single pharmaceutical formulation as well as administration of the inventive compound and additional agent(s) in separate pharmaceutical formulations.
[000129] Another aspect of the invention is a method of treating diabetes and related diseases comprising the step of administering to a subject -suffering from a diabetic or related condition a therapeutically effective amount of a compound of Formulas I -XIII. Additionally, the invention provides a method of treating inflammation or inflammatory diseases or diseases mediated by cytokines, PDE4, PDE3, p44/42 MAP kinase, iNOS and/or COX-2 by administering to a subject in need of such treatment an effective amount of a compound according to Formulas I - XIII. Further, pharmaceutical compositions containing a therapeutically effective amount of one or more compounds according to Formulas I - XIII together with a pharmaceutically or physiologically acceptable carrier, for use in the treatments contemplated herein, are also provided.
[000130] The compounds of the invention are useful for the treatment of diabetes, characterized by the presence of elevated blood glucose levels, that is, hyperglycemic disorders such as diabetes mellitus, including both type 1 and 2 diabetes, as well as other hyperglycemic related disorders such as obesity, increased cholesterol, hyperiipidemia such as hypertriglyceridemia, kidney related disorders and the like. The compounds are also useful for the treatment of disorders linked to insulin resistance and/or hyperinsulinemia, which include, in addition to diabetes, hyperandrogenic conditions such as polycystic ovary syndrome (Ibanez et al., J. Clin Endocrinol Metab, 86:3626-30, 2000; Taylor A.E., Obstet Gynecol Clin North Am, 27:683-96, 2000), coronary artery disease such as atherosclerosis and vascular restenosis, and peripheral vascular disease. Additionally, the compounds of the present invention are also useful for the treatment of inflammation and immunological diseases that include those mediated by signaling pathways linked to pro-inflammatory cytokines, such as rheumatoid arthritis, ankylosing spondylitis, multiple sclerosis, inflammatory bowel disease, psoriasis, and contact and atopic dermatitis. [000131] By "treatment", it is meant that the compounds of the invention are administered in an amount which is at least sufficient to, for example, reduce the blood glucose level in a patient suffering from a hyperglycemic disorder or to inhibit or prevent the development of pro-inflammatory cytokine or like responses in a patient suffering from inflammatory or immunological disease. In the case of diabetes, the compound is usually administered in the amount sufficient to reduce the blood glucose level, free fatty acid level, triglyceride level and/or the like level sufficient to improve or alleviate the symptoms and/or reduce the risk of complications associated with elevated levels of these parameters. A variety of subjects may be treated with the present compounds to reduce blood glucose levels such as livestock, wild or rare animals, pets, as well as humans. The compounds may be administered to a subject suffering from hyperglycemic disorder using any convenient administration technique, including intravenous, intradermal, intramuscular, subcutaneous, oral and the like. However, oral daily dosage is preferred. The dosage delivered to the host will necessarily depend upon the route by which the compound is delivered, but generally ranges from about 0.1 to about 600 mg/kg human body weight or typically from about 0.1 to about 60 mg/kg human body weight. Generally similar types of administration and dosages are also contemplated when the compounds of the invention are used to treat inflammatory or immunological disease.
[000132] The compounds of this invention may be used in formulations using acceptable pharmaceutical vehicles for enteral, or parenteral, administration, such as, for example, water, alcohol, gelatin, gum arabic, lactose, amylase, magnesium stearate, talc, vegetable oils, polyalkylene glycol, and the like. The compounds can be formulated in solid form, e.g., as tablets, capsules, drages and suppositories, or in the liquid form, e.g., solutions, suspensions and emulsions. The preparations may also be delivered transdermally or by topical application.
[000133] The syntheses of representative compounds according to the present invention are illustrated in Schemes I and II. Further examples illustrating the syntheses of additional compounds according to the present invention are also given below. SCHEME
Figure imgf000039_0001
Figure imgf000039_0002
[000134] Scheme 1 details the synthesis of compounds 1-6. Scheme 2 details the synthesis of 17. It is to be understood that the Schemes 1 and 2 are representative schemes and are not intended to be limited to the compounds disclosed. SCHEME II
Figure imgf000040_0001
21 22
Figure imgf000040_0002
23 17
[000135] The following examples are provided to further illustrate the present invention and are not intended to limit the invention in any way.
EXAMPLE 1
Synthesis of 3-(3,δ-dimethoxyphenyl)-2-{4-[4-(3-oxo-3-ureidopropyl)-phenoxy]- phenylj-acrylic acid methyl ester (1) [see Scheme I]
[000136] Step 1 : Synthesis of 3-(3,δ-dimethoxyphenyl)-2-(4-hvdroxyphenyl)- acrylic acid (2). To a mixture of 3,δ-dimethoxybenzaldehyde (120 g, 0.72 mol) and p-hydroxyphenyl acetic acid (110 g, 0.72 mol) was added acetic anhydride (240 mL) and triethylamine (161 mL, 1.6 equiv.). This non-homogeneous mixture on heating becomes homogeneous at ~70°C. After being stirred at 130°C for 4 hr, the mixture was cooled to room temperature. HCI (16%, 600 mL) was added to the reaction mixture slowly in 30min keeping temperature below δ-10°C. The solid was dissolved in 3N aqueous NaOH (1.2 L) and stirred for 0.5 hr. The filtrate was acidified, maintaining a temperature at 25-30°C, with cone. HCI (-700 mL) to pH 1. The precipitated product was filtered and washed with water to give crude product (~300 g, wet cake). The crude product was dissolved by heating in ethanol and recrystallized by adding equal volume of water. The product was dried overnight in a vacuum oven at 40°C. Yield: 161 g, 74%. Analysis: 1HNMR (DMSO-d6): 512.48 (br, 1 H), 9.42 (s, 1 H), 7.69 (s, 1H), 6.9δ (d, J=8.0 Hz, 2H), 6.76 (d, J=8.0 Hz, 2H), 6.36 (t, J=2.2 Hz, 1 H), 6.27 (d, J=2.2 Hz, 2H), 3.56 (s, 6H).
[000137] (b) Step 2: Synthesis of 3-(3,δ-dimethoxyphenyl)-2-r4-(4- formylphenoxy)-phenyl1-acrylic acid (3). 2 (64.0 g, 0.21 mol) was dissolved in 320 mL anhydrous DMSO under nitrogen, and potassium tetf-butoxide (48.0 g, 0.43 mol) was added in lots. When the solution became homogenous, p- fluorobenzaldehyde (27 mL, 0.22 mol) was added and the mixture was heated at 100°C for δ hr. After cooling to room temperature, the solution was poured into 1 L water and extracted with ether (2 x 600 mL). The aqueous phase was acidified with δ% HCI to ~ pH 4 and the precipitated product was collected by suction filtration. The wet filter cake was dissolved in a minimum of boiling acetone and recrystallized with addition of water. After chilling to 4°C for 3 hr, the solid was collected by vacuum filtration. The product was dried overnight at 40°C in a vacuum oven. Yield: 62 g, 73%. Analysis: 1HNMR (DMSO-d6): 612.87 (s, 1 H), 9.94 (s, 1 H), 7.95 (d, J=8.2 Hz, 2H), 7.72 (s, 1 H), 7.27 (d, J=8.0 Hz, 2H), 7.19 (d, J=8.0 Hz, 2H), 7.15 (d, J=8.2 Hz, 2H), 6.42 (t, J=1.6 Hz, 1 H), 6.29 (d, J=2.0 Hz, 2H), 3.60 (s, 6H).
[000138] (c) Step 3: Synthesis of 3-(3.δ-dimethoxyphenyl)-2-(4-r4-(2- ethoxycarbonyl-vinvD-phenoxyl-phenvD-acrylic acid (4). Triethylphosphonoacetate (7.14 mL, 36 mmol) was added to a suspension of NaH (60% in mineral oil, 2.64 g, 66 mmol) in anhydrous THF (100 mL) at 0°C under argon, and the mixture was stirred for 16 min. A solution of aldehyde 3, (12.12 g, 30 mmol) in THF (100 mL) was added and the mixture was stirred for 1 h. The mixture was quenched with saturated aqueous ammonium chloride solution (δ mL), diluted with ethyl acetate (300 mL) and acidified with 5% aqueous HCI to pH 1. The ethyl acetate layer was separated, and the aqueous layer was extracted with ethyl acetate (100 mL). The combined organic layers were washed with brine, dried over anhydrous MgS04, filtered and concentrated. The crude product was purified by recrystallization from a mixture of chloroform/methanol. The compound was suspended in hot methanol (200 mL) and a minimum volume (-30-40 mL) of chloroform was added to yield 4. Yield: 12.39 g, 87.1%. Analysis: 1HNMR (DMSO-d6): 57.77 (d, J=8.4 Hz, 2H), 7.69 (s.1 H), 7.6δ (d, J=16 Hz, 2H), 7.23 (d, 8.8 Hz, 2H), 7.11 (d, J=8.8 Hz, 2H), 7.01 (d,.J=8.4 Hz, 2H), 6.67 (d, J=16 Hz, 2H), 6.41 (t, J=2 Hz, 1 H), 6.28 (d, J=1.6 Hz, 2H), 4.18 (q, J=7.2 Hz, 2H), 3.69 (s, 6H), 1.26 (t, J= 7.2 Hz, 3H).
[000139] (d) Step 4: Synthesis of 3-(3,5-dimethoχyphenyl)-2-(4-r4-(2- ethoxycarbonyl-ethvD-phenoxyl-phenvD-acrylic acid (5). To a suspension of Raney Ni (10.0 g, Raney 2800 nickel in water active catalyst) in ethanol-dioxane (2:1, 60 mL) was added a solution of 4 (13.0 g, 27.4 mmol) in a mixture of ethanol-dioxane (2:1 , 400 mL), and the resulting mixture was stirred vigorously for 16 hr under hydrogen at atmospheric pressure. Completion of the reaction was monitored by HPLC (time varies with the speed of stirring). Catalyst was filtered through a bed of Celite® diatomaceous earth, the bed was washed with ethanol-dioxane (2:1 , 200 mL), and solvent was evaporated. The solid obtained was dissolved in hot toluene (160 mL) and cooled at 4°C overnight. Solid separated was filtered and washed with ice-cold toluene (60 mL) and dried at δδ°C for 6 hr. Yield: 11.61 g, 90.5%. Analysis: 1HNMR (DMSO-de): 512.75 (s, 1 H), 7.68 (s, 1 H), 7.26 (d, J=8.4 Hz, 2H), 7.17 (d, J=8.4 Hz, 2H), 6.99 (d, J=8.4 Hz, 2H), 6.94 (d, J=8.4 Hz, 2H), 6.39 (t, J=2.0 Hz, 1 H), 6.27 (d, J=1.6 Hz, 2H), 4.06 (q, J=7.2 Hz, 2H), 3.57 (s, 6H), 2.84 (t, J=8 Hz, 2H), 2.60 (t, J=8 Hz, 2H), 1.15 (t, J=8 Hz, 3H).
[000140] (e) Step 5: Synthesis of 3-(3.δ-dimethoxyphenyl)-2-f4-r4-(3-oxo-3- ureido-propyD-phenoxyl-phenvD-acrylic acid (6). To a solution of sodium ethoxide in ethanol (21% w/w, 66 mL) under argon was added ethyl acetate (3.12 mL), then refluxed for 20 min. Urea (18 g, 0.3 mol) was dissolved in the above-mentioned sodium ethoxide in ethanol solution at 7δ°C. To this solution was added 5 (13 g, 0.027 mol) in one lot. After all dissolved, the resulting mixture was stirred at 7δ°C for another δ min, cooled quickly in 15 min to 15-20°C, TFA (13 mL) added, and then adjusted to pH 4-δ with 5% HCI. After stirring at room temperature for 1 hr, the mixture was slowly added to water (520 mL). The solid separated was filtered and refluxed in 10% isopropanol in ethyl acetate (150 mL) for 20 min. The mixture was allowed to cool to room temperature, then incubated overnight at 4°C. The mixture was filtered and solid was dried. Yield: 8.5 g. Analysis: 1HNMR (DMSO-d6): 512.35 (br, 1 H), 10.20 (s, 1 H), 7.75 (br, 1 H), 7.68 (s, 1 H), 7.26 (d, J=8.4 Hz, 2H), 7.17 (d, J=8.4 Hz, 2H), 6.99 (d, J=8.4 Hz, 2H), 6.94 (d, J=8.4 Hz, 2H), 6.39 (t, .J=2.4 Hz, 1 H), 6.27 (d, J=2.4 Hz, 2H), 3.57 (s, 6H), 2.81 (t, J=7.2 Hz, 2H), 2.54 (t, J=7.2 Hz, 2H).
[000141] (f) Step 6: Synthesis of S-fS.δ-dimethoxyphenvn^- -K-fS-oxo-S- ureido-propyD-phenoxyl-phenvD-acrylic acid methyl ester (1). To a stirred solution of 6 (δ g, 0.01 mol) in dry DMF (36 mL) under argon was added K2C03 (1.38 g, 0.01 mol). To this, dimethyl sulfate (3.8 g, 0.03 mol) was added and stirred at room temperature for 30 min. The reaction mixture was acidified with 5% aqueous HCI and extracted with ethyl acetate. The organic layer was dried over anhydrous magnesium sulfate and evaporated. The oily residue was dissolved in hexane/ethyl acetate (2:3, 30 mL) with stirring, and incubated overnight at 4°C for crystallization. The solid was collected by vacuum filtration and dried. Yield: 3.3 g, 66%. Analysis: 1HNMR (DMSO-d6): 510.17 (br, 1H), 7.72 (br, 2H), 7.72 (s, 1H), 7.2δ (d, J=8.4 Hz, 2H), 7.18 (d, J=6.8 Hz, 2H), 7.21 (s overlapped, 1 H), 7.01 (d, J=6.8 Hz, 2H), 6.96 (d, J=8.4 Hz, 2H), 6.41 (t, J=2.2 Hz, 1H), 6.28 (d, J=2.2 Hz, 2H), 3.73 (s, 3H), 3.57 (s, 6H), 2.84 (t, J=7.2 Hz, 2H), 2.61 (t, J=7.2 Hz, 2H).
EXAMPLE 2
Synthesis of 3-(3,5-dimethoxyphenyl)-2-{4-[4-(3-ethoxycarbonylamino-3-oxo-propyl)- phenoxy]-phenyl}-acrylic acid methyl ester (8)
[000142] 2-{4-[4-(2-Carbamoyl-ethyl)-phenoxy]-phenyl}-3-(3,δ-dimethoxyphenyl)- acrylic acid methyl ester (7) was obtained as a byproduct in the synthesis of 3-(3,5- dimethoxy-phenyl)-2-{4-[4-(2,4-dioxothiazolidin-δ-ylmethyl)-phenoxy]-phenyl}-acrylic acid methyl ester, performed essentially as shown in PCT/US99/09982 (WO 99/58127). 7 (460 mg, 1.0 mmol) was taken up in dry THF (6 mL) and cooled to - 78°C. To this solution, lithium diisopropyl amide (LDA) (2M, 0.56 mL, 1.1 mmol) was added and stirred for 10 min. Ethyl chloroformate (0.11 mL, 1.2 mmol) was added and stirred overnight at room temperature. The reaction was quenched with saturated aqueous ammonium chloride solution and ethyl acetate (60 mL) was added. The organic layer was washed with brine (2 x 20 mL), dried on anhydrous magnesium sulfate and evaporated under reduced pressure. The crude product was purified by silica gel chromatography and eluted with hexane-ethyl acetate (7:3). Yield: 264 mg, 49.8%.
Figure imgf000044_0001
8
[000143] Analysis: 1HNMR (DMSO-d6): 510.62 (s, 1 H), 7.70 (s, 1 H), 7.24 (d, J=8.4 Hz, 2H), 7.17 (d, J=8.4 Hz, 2H), 6.99 (d, J=8.4 Hz, 2H), 6.94 (d, J=8.4 Hz, 2H), 6.40 (t, J=2.1 Hz, 1H), 6.27 (d, J=2.1 Hz, 2H), 4.07 (q, J=7.2 Hz, 2H), 3.70 (s, 3H), 3.56 (s, 6H), 2.76 (m, 4H), 1.19 (t, J=7.2 Hz, 3H).
EXAMPLE 3
Synthesis of 2-{4-[4-(3-benzoyloxycarbonylamino-3-oxo-propyl)-phenoxy]-phenyI}-3-
(3,5-dimethoxyphenyl)-acrylic acid methyl ester (9)
[000144] 7 (1.38, 3.0 mmol) prepared as in Example 2 was taken up in dry THF (20 mL) and cooled to -78°C. To this solution, LDA (2M, 1.8 mL, 3.6 mmol) was added and stirred for 10 min. Benzyl chloroformate (0.67 g, 39 mmol) was added and stirred overnight at room temperature. The reaction was quenched with saturated aqueous ammonium chloride solution, and ethyl acetate (150 mL) was added. The organic layer was washed with brine (2 x 25 mL), dried on anhydrous magnesium sulfate and evaporated under reduced pressure. The crude product was purified by silica gel chromatography and eluted with hexane-ethyl acetate (7:3). Yield: 0.68g, 37.3%.
Figure imgf000045_0001
[000145] Analysis: 1HNMR (DMSO-d6): 510.66 (s, 1 H), 7.72 (s, 1 H), 7.38-7.39 (m, 5H), 7.26 (d, J=8.4 Hz, 2H), 7.18 (d, J=8.4 Hz, 2H), 7.00 (d, J=8.4 Hz, 2H), 6.94 (d, J=8.4 Hz, 2H), 6.41 (t, J=2.0 Hz, 1H), 6.28 (d, J=2.0 Hz, 2H), 5.12 (s, 2H), 3.72 (s, 3H), 3.57 (s, 6H), 2.79 (m, 4H).
EXAMPLE 4 Synthesis of 3-(3,5-dimethoxyphenyl)-2-{4-[4-(3-oxo-3-ureidopropyl)-phenoxy]- phenylj-propionic acid (10)
[000146] 3-(3,5-Dimethoxyphenyl)-2-{4-[4-(2-ethoxycarbonylvinyl)-phenoxy]- phenylj-acrylic acid (4, 2.37 g, 5.0 mmol) was dissolved in a mixture of ethanol- dioxane (2:1 , 150 mL), and palladium charcoal (10%, 500 mg) was added. The mixture was stirred under hydrogen for 15 hr. Catalyst was then removed by filtration, and solvent was evaporated under reduced pressure to yield 3-(3,5- dimethoxy-phenyl)-2-{4-[4-(2-ethoxycarbony!ethyl)-phenoxy]-phenyl}-propionic acid (18) quantitatively. Urea (0.21 g, 3.68 mmol) was dissolved in sodium ethoxide (2.7 M, 2.2 mL, δ.92 mmol) at 80°C under argon, and to this a solution of 18 (1.13 g, 2.37 mmol) in anhydrous ethanol (1 δ mL) was added and heated at this temperature for 13 hr. Ethanol was evaporated under reduced pressure, water (20 mL) was added, acidified to pH 1 by δ% aqueous HCI and extracted with ethyl acetate (60 mL). The organic layer was washed with water (2 x 2δ mL), brine (2 x 20 mL), dried over anhydrous magnesium sulfate and evaporated. The crude product was purified by silica gel chromatography and eluted with hexane-ethyl acetate (3:7) containing acetic acid (1%), followed by recrystallization from ethanol. Yield: 266 mg, 22.8%.
Figure imgf000046_0001
[000147] Analysis: 1HNMR (DMSO-d6): 512.37 (s, 1 H), 10.17 (s, 1 H), 7.74 (br, 1 H), 7.31 (d, J=9.2 Hz, 2H), 7.21 (d, J=9.2 Hz, 2H), 6.91 (d, J=8.4 Hz, 2H), 6.90 (d, J=8.4 Hz, 2H), 6.33 (d, J=2.0 Hz, 2H), 6.29 (t, J=2.0 Hz, 1 H), 3.83 (t, J=8.0 Hz, 1 H), 3.68 (s, 6H), 3.19 (dd, J= 14.4 & 8.4 Hz, 1 H), 2.88-2.80 (m, 3H), 2.69 (t, J=8.0 Hz, 2H).
EXAMPLE δ Synthesis of 3-(3,5-dimethoxyphenyl)-2-{4-[4-(3-oxo-3-ureidopropenyl)-phenoxy]- phenylj-acrylic acid (11)
[000148] Urea (0.21 g, 3.68 mmol) was dissolved in sodium ethoxide (2.7 M, 2.2 mL, δ.92 mmol) at 80°C under argon, and to this a solution of 4 (1.14 g, 2.37 mmol) in anhydrous ethanol (16 mL) was added and heated at this temperature for 13 hr. Ethanol was evaporated under reduced pressure, water (20 mL) was added, acidified to pH 1 by 6% aqueous HCI and extracted with ethyl acetate (60 mL). The organic layer was washed with water (2 x 26 mL), brine (2 x 20 mL), dried over anhydrous magnesium sulfate and evaporated. The crude product was purified by silica gel chromatography and eluted with hexane-ethyl acetate (3:7) containing acetic acid (1 %), followed by recrystallization from ethanol. Yield: 167 mg, 14.4%.
Figure imgf000047_0001
[000149] Analysis: 1HNMR (DMSO-d6): 512.51 (br, 1 H), 10.30 (s, 1 H), 7.92 (br, 1H), 7.77 (d, J=9.2 Hz, 2H), 7.68 (s, 1 H), 7.65 (d, J=16.0 Hz, 1 H), 7.30 (br, 1 H), 7.22 (d, J=8.8 Hz, 2H), 7.10 (d, J=8.8 Hz, 2H), 7.03 (d, J=9.2 Hz, 2H), 6.73 (d, J=16.0 Hz, 1H), 6.40 (t, J=2.0 Hz, 1 H), 6.28 (d, J=2 Hz, 2H), 3.59 (s, 6H).
EXAMPLE 6 Synthesis of 3-(3,5-dimethoxyphenyl)-2-{4-[4-(3-oxo-3-ureidopropyl)-phenoxy]- phenylj-acrylic acid ethyl ester (12)
[000150] To a stirred solution of 6 (0.40 g, 0.81 mmol) in dry DMSO (3 mL) was added K2C03 (0.14 g, 0.98 mmol). To this, diethyl sulfate (0.115 g, 0.91 mmol) was added and stirred at room temperature for 30 min. The reaction mixture was poured into water (30 mL) and extracted with ethyl acetate. The organic layer was dried over anhydrous magnesium sulfate and evaporated. The crude product was purified by column chromatography over silica gel and eluted with hexanes-ethyl acetate (3:1). Yield: 0.39 g, 92.2%.
Figure imgf000047_0002
[000151] Analysis: 1HNMR (DMSO-d6): 510.17 (s, 1 H), 7.74 (br, 1 H), 7.70 (s, 1 H), 7.25 (d, J=8.4 Hz, 2H), 7.24 (overlapped, 1 H), 7.18 (d, J=8.4 Hz, 2H), 7.00 (d, J=8.4 Hz, 2H), 6.95 (d, J=8.4 Hz, 2H), 6.41 (t, J=1.6 Hz, 1 H), 6.28 (d, J=1.6 Hz, 2H), 4.19 (q, J=8.0 Hz, 2H), 3.67 (s, 6H), 2.83 (t, J=7.2 Hz, 2H), 2.60 (t, J=7.2 Hz, 2H), 1.26 (t, J=8.0 Hz, 3H).
EXAMPLE 7 Synthesis of 3-(3,δ-dimethoxyphenyl)-Λ/,Λy-dimethyl-2-{4-[4-(3-oxo-3-ureido-propyl)- phenoxy]-phenyl}-acrylamide (13)
[000152] To a stirred solution of 6 (1.68 g, 3.43 mmol) in dry DMF (30 mL) was added carbonyldiimidazole (1.1 g, 6.86 mmol), and the reaction mixture was heated to 60°C for 1 hr. The reaction mixture was cooled to 0°C and a solution of dimethylamine in THF (2 M, 8.6 mL, 17.2 mmol) was added and stirred for 18 hr. The reaction mixture was diluted with water (100 mL) and extracted with ethyl acetate (100 mL). The organic phase was then rinsed sequentially with 10% citric acid (2 X δOmL), water (2 X δOmL), and brine (20 mL), then dried over anhydrous magnesium sulfate and evaporated. The crude product was purified by silica gel chromatography using hexane-ethyl acetate (3:7) containing 1 % acetic acid. Yield: 1.77 g, 100%.
Figure imgf000048_0001
[000153] Analysis: 1HNMR (DMSO-d6): 510.17 (br, 1 H), 7.74 (br, 1H), 7.27 (d, J=9.2 Hz, 2H), 7.23 (d, J=8.8 Hz, 2H), 7.23 (br, 1 H), 6.79 (d, J=9.2 Hz, 2H), 6.93 (d, J=8.8 Hz, 2H), 6.56 (s, 1 H), 6.34 (t, J=2 Hz, 1 H), 6.29 (s, 1 H), 6.28 (s, 1 H), 3.58 (s, 6H), 3.05 (br, 3H), 2.90 (br, 3H), 2.82 (t, J=7.2 Hz, J=8.0 Hz, 2H), 2.59 (t, J=8.0 Hz, J=7.2 Hz, 2H). EXAMPLE 8 Synthesis of 2-(4-{4-[3-(3-cyclohexylureido)-3-oxopropyl]-phenoxy}-phenyl)-3-(3,5- dimethoxyphenyl)-acrylic acid (14)
[000154] Cyclohexylurea (1.3 g, 9 mmol) was dissolved in sodium ethoxide in ethanol (21 % w/w, 3 mL) at 75°C. To this solution 5 was added (O.δ g, 1.1 mmol) in one lot. The resulting mixture was stirred at 7δ°C for δ min, then cooled quickly to 40-50°C. TFA (0.5 mL) was added and then 5% aqueous HCI (1 N, 0.6 mL). After stirring at room temperature for 1 hr, the mixture was left overnight at 4°C. The solid separated was filtered and refluxed in ethyl acetate (4 mL) for 20 min. The mixture was allowed to cool to room temperature, filtered and the crude product was purified by silica gel chromatography using hexane-ethyl acetate (1 :1). Yield: 0.27 g, 45%.
Figure imgf000049_0001
[000155] Analysis: 1HNMR (DMSO-d6): 512.74 (s, 1 H), 10.30 (s, 1 H), 8.32 (br, 1 H), 7.67 (s, 1 H), 7.24 (d, J=8.8 Hz, 2H), 7.16 (d, J=8.8 Hz, 2H), 6.90 (d, J=8.4 Hz, 2H), 6.94 (d, J=8.4 Hz, 2H), 6.34 (t, J=2.4 Hz, 1 H), 6.27 (d, J=2.4 Hz, 2H), 3.58 (s, 6H), 2.83 (t, J=7.6 Hz, 2H), 2.69 (t, J=7.6 Hz, 2H), 1.78 (m, 2H), 1.61 (m, 2H), 1.51 (m, 1 H), 1.32-1.16 (m, 5H).
EXAMPLE 9 Synthesis of [3-(4-phenόxyphenyl)-propionyl]-urea (15)
[000156] 4-Phenoxy-benzaldehyde was reacted with triethyl phosphonoacetate to yield 3-(4-phenoxyphenyl)-acrylic acid ethyl ester, which was then reduced with H2 using palladium-on-carbon catalyst to yield 3-(4-phenoxyphenyl)-propionic acid methyl ester (19). Urea (1.20 g, 19.99 mmol) was dissolved in sodium ethoxide (2 M, 6.7 mL, 13.4 mmol) at 80°C under argon, and to this a solution of 19 (1.71 g, 6.67 mmol) in anhydrous ethanol (8 mL) was added and heated at this temperature for 1 hr. Ethanol was evaporated under reduced pressure, water (20 mL) was added, acidified to pH 1 by δ% aqueous HCI and extracted with ethyl acetate (50 mL). The organic layer was washed with water (2 x 25 mL), brine (2 x 20 mL), dried over anhydrous magnesium sulfate and evaporated. The crude product was purified by silica gel chromatography and eluted with hexane-ethyl acetate (1 :1) containing acetic acid (1%) followed by recrystallization from ethanol. Yield: 113 mg, δ.6%.
Figure imgf000050_0001
19 15
[000157] Analysis: 1HNMR (DMSO-d6): 510.18 (s, 1 H), 7.74 (br, 1 H), 7.38 (d, J=7.6 Hz, 1 H), 7.36 (d, J=7.6 Hz, 1H), 7.22 (d, J=8.8 Hz, 2H), 7.17 (t, J=7.2 Hz, 1 H), 6.97 (d, J=7.2 Hz, 2H), 6.93 (d, J=8.8 Hz, 2H), 2.82 (t, J=7.2 Hz, 2H), 2.59 (t, J=7.2 Hz, 2H).
EXAMPLE 10
Synthesis of 2-{4-[4-(3-acetylureidomethyl)-phenoxy]-phenyl}-3-(3,5- dimethoxyphenyl)-acrylic acid methyl ester (17) [see Scheme II]
[000158] Step 1 : Synthesis of 3-(3,δ-dimethoxyphenyl)-2-r4-(4-hvdroxymethyl- phenoxy)-phenyll-acrylic acid methyl ester (22). 3-(3,δ-Dimethoxy-phenyl)-2-[4-(4- formylphenoxy)-phenyl]-acrylic acid methyl ester (21) was first prepared by converting the corresponding free acid (3) to the methyl ester by addition of DMF, K2CO3 and dimethyl sulfate in a manner analogous to Example 1 (f) above. Sodium borohydride (0.126 g, 3.3 mmol) was added to a suspension of 21 (1.26 g, 3 mmol) in ethanol (20 mL) and stirred at room temperature for 1 hr. The reaction was quenched with 5% aqueous HCI, and ethanol was evaporated under reduced pressure. Residue was taken up in ethyl acetate (60 mL) and washed with brine (2 x 20 mL), dried over anhydrous magnesium sulfate and evaporated. The crude product was purified by silica gel chromatography and eluted with hexanes-ethyl acetate (1 :1). Yield: 1.14 g, 95.0%. Analysis: 1HNMR (DMSO-d6): 57.72 (s, 1 H), 7.36 (d, J=8.8 Hz, 2H), 7.19 (d, J=8.8 Hz, 2H), 7.01 (d, J=8.4 Hz, 2H), 6.99 (d, J=8.4 Hz, 2H), 6.41 (t, J=2.4 Hz, 1 H), 6.28 (d, J=2.4 Hz, 2H), 5.18 (t, J=6.4 Hz, 1 H), 4.49 (d, J=4.8 Hz, 2H), 3.72 (s, 3H), 3.57 (s, 6H).
[000159] (b) Step 2: Synthesis of 2-f4-(4-bromomethylphenoxy)-phenyll-3-(3.δ- dimethoxyphenvD-acrylic acid methyl ester (23). To a stirred solution of 22 (1.06 g, 2.5 mmol) in dichloromethane (10 mL) at 10°C, PBr3 (1 M, 3.75 mL) was added and stirred for 1 hr. The reaction was quenched with saturated aqueous sodium bicarbonate solution. The organic layer was washed with water (20 mL), brine (2 x 30 mL), dried over anhydrous magnesium sulfate and evaporated. The crude product was purified by silica gel chromatography and eluted with hexanes-ethyl acetate (4:1). Yield: 0.85 g, 70.4%. Analysis: 1HNMR (DMSO-d6): 57.73 (s, 1H), 7.49 (d, J=8.4 Hz, 2H), 7.22 (d, J=8.4 Hz, 2H), 7.07 (d, J=8.4 Hz, 2H), 7.00 (d, J=8.4 Hz, 2H), 6.42 (t, J=2.4 Hz, 1 H), 6.28 (d, J=2.4 Hz, 2H), 4.74 (s, 2H), 3.73 (s, 3H), 3.58 (s, 6H).
[000160] (c) Synthesis of 2-{4-r4-(3-acetylureidomethyl)-phenoxy1-phenyl>-3- (3,5-dimethoxyphenyl)-acrylic acid methyl ester (17). To a stirred suspension of sodium hydride (60% in oil, 0.11 g, 2.8 mmol) in dimethylformamide (2 mL), N- acylurea (0.11 g, 1.12 mmol) was added and stirred at room temperature for 30 min. A solution of 23 (0.54 g, 1.12 mmol) in dimethylformamide (3 mL) was added and heated overnight at 80°C. The reaction was quenched with water and extracted with ethyl acetate (3 x 30 mL). The combined organic layer was washed with brine (2 x 25 mL), dried over anhydrous magnesium sulfate and evaporated. The crude product was purified by silica gel column chromatography and eluted with hexanes- ethyl acetate (3:7) containing 1% acetic acid. Yield: 0.16 g, 28.4%. Analysis: 1HNMR (DMSO-de): 58.34 (t, J=5.6 Hz, 1 H), 7.72 (s, 1 H), 7.29 (d, J=8.4 Hz, 1 H), 7.19 (d, J=8.4 Hz, 2H), 7.02 (d, J=8.4 Hz, 2H), 6.99 (d, J=8.4 Hz, 2H), 6.42 (t, J=8.4 Hz, 1 H), 6.28 (d, J=2.4 Hz, 2H), 4.24 (d, J=5.2 Hz), 3.73 (s, 3H), 3.57 (s, 6H), 1.87 (s, 3H). General Procedure for Conversion of Carboxylic Acids to Amides
[000161] A mixture of carboxylic acid (1.1 mmol) and carbonyldiimidazole (1.3 mmol) in DMF (20 mL) was heated at 60°C for 30 min. After the reaction mixture was cooled to room temperature, a solution of amine (2M, 1 mL, 2.0 mmol) was added and stirred for 18 hr. To the reaction mixture water (100 mL) was added and extracted with ethyl acetate (3 x 60 mL). The organic phase was washed with 10% citric acid (20 mL), water (2 x 60 mL), and brine (60 mL), then dried over anhydrous magnesium sulfate and removed the solvent. The crude product was purified by silica gel chromatography.
EXAMPLE 11 Synthesis of N,N-dimethyl-2-{4-[4-(3-oxo-3-ureidopropyl)-phenoxy]-phenyl}- acetamide (26)
[000162] Urea (0.78 g, 13 mmol) and 3-[4-(4-carboxymethylphenoxy)-phenyl]- propionic acid ethyl ester, 24 (O.δ g, 1.5 mmol) were dissolved in sodium ethoxide in ethanol (2M, 6.5 mL, 13 mmol) at 80°C under argon, and the reaction mixture was heated at this temperature for 1h. The reaction was then quenched by TFA (0.5 mL) after cooling to δ°C. Water (40 mL) was added to the reaction mixture. The crude product was filtered and purified by silica gel chromatography and eluted with hexane-ethyl acetate (1 :1) containing acetic acid (1%) followed by recrystallization from toluene yielded 25 (0.28 g, 64%).
[000163] Analysis: 1HNMR (DMSO-d6): 5 12.28 (br, 1 H), 7.73 (br, 1 H), 7.24 (d, J=8.8 Hz, 2H), 7.23, (br, 1 H), 7.21 (d, J=8.8 Hz, 2H), 6.93, (d, J=8.8 Hz, 2H), 6.92 (d, J=8.8 Hz, 2H), 3.54 (s, 2H), 2.81 (t, J=7.2 Hz, 2H), 2.58 (t, J=7.2 Hz, 2H).
[000164] Following the general procedure for conversion of carboxylic acids to amides mentioned above and using dimethyl amine as amine, 25 was converted to 26 in 97% yield. [000165] Analysis: 1HNMR (DMSO-d6): 510.17(s, 1H), 7.73 (s, 1 H), 7.22 (s, 1H), 7.21 (d, J=8.0 Hz, 2H), 7.19 (d, J=8.0 Hz, 2H), 6.92 (d, J=8.0 Hz, 2H), 6.90 (d, J=8.0 Hz, 2H), 3.66 (s, 2H), 3.00 (s, 3H), 2.81 (t, J=8.0 Hz, 2H), 2.68 (t, J=8.0 Hz, 2H).
Figure imgf000053_0001
24 2δ 26
EXAMPLE 12 Synthesis of (4-{4-[2-(3,5-dimethoxyphenyl)-1-dimethylcarbamoyl-vinyl]-phenoxy}- benzyl)-carbamic acid methyl ester (29)
[000166] Reaction of 3-(3,5-dimethoxyphenyl)-2-{4-[4-(2,4-dioxothiazolidin-3- ylmethyl)-phenoxy]-phenyl}-acrylic acid, 27, (0.4 g, 0.77 mmol) with δ% LiOH (2mL) in methanol (19 mL) was carried out at room temperature for 18 h. The reaction mixture was acidified to pH 3 by δ% aqueous HCI and extracted with ethyl acetate (2 x 60 mL). The organic layer was washed with water (2 x 60 mL), brine (2 x 20 mL), dried over anhydrous magnesium sulfate and evaporated. The crude product was purified by silica gel chromatography and eluted with hexane- ethyl acetate (1 :1) containing acetic acid (1 %). Yield (28): 0.31 g, 83%. Analysis: 1HNMR (DMSO-d6): δ 12.76 (br, 1H), 7.68 (t, J=4.6 Hz, 1H), 7.67 (s, 1H), 7.28 (d, J=8.8 Hz, 2H), 7.17 (d, J=8.8 Hz, 2H), 7.01 (d, J=8.8 Hz, 2H), 6.97 (d, J=8.8 Hz, 2H), 6.39 (t, J=2.8 Hz, 1 H), 6.27 (d, J=2.4 Hz, 2H), 4.17 (d, J=6.4 Hz, 2H), 3.58 (S, 6H), 3.56 (s, 3H).
[000167] Following the general procedure for conversion of carboxylic acids to amides mentioned above and using dimethyl amine as amine, 28 was converted to 29 in 96% yield. [000168] Analysis: 1HNMR (DMSO-d6): 5 7.68 (t, J=4.6 Hz, 1 H), 7.28 (d, J=8.8 Hz, 2H), 7.27 (d, J=8.8Hz, 2H), 6.98 (d, J=8.8 Hz, 2H), 6.96 (d, J=8.8 Hz, 2H), 6.67 (s, 1 H), 6.35 (t, J=2.8 Hz, 1 H), 6.28 (d, J=2.4 Hz, 2H), 4.16 (d, J=6.4 Hz, 2H), 3.59 (S, 6H), 3.δδ (s, 3H), 3.06 (br, 3H), 2.91 (br, 3H).
Figure imgf000054_0001
27 28 29
EXAMPLE 13 Synthesis of 2-{4-[4-(2-carbamoylethyl)-phenoxy]-phenyl}-3-(3,5-dimethoxyphenyl)-
N,N-dimethylacrylamide (31)
[000169] Urea (0.78 g, 13 mmol) and 3-(3,5-dimethoxyphenyl)-2-{4-[-4-(2- ethoxycarbonylethyl)-phenoxy]-phenyl}-acrylic acid 5 (0.45 g, 1.5 mmol) were dissolved in sodium ethoxide in ethanol (2M, 6.5 mL, 13 mmol) at 80°C under argon, and the reaction mixture was heated at this temperature for 5 h. The reaction was then quenched by TFA (O.δ mL) after cooling to 5°C. Water (40 mL) was added to the reaction mixture. The crude product was filtered and purified by silica gel chromatography and eluted with hexane-ethyl acetate (1 :1) containing acetic acid (1 %). Yield (30): 0.39 g, 93%.
[000170] Analysis: 1HNMR (DMSO-d6): 5 12.73 (br, 1H), 7.68 (s, 1H), 7.29 (br, 1 H), 7.24 (d, J=8.8 Hz, 2H), 7.65 (d, J=8.8 Hz, 2H), 6.99 (d, J=8.8 Hz, 2H), 6.92 (d, J=8.8 Hz, 2H), 6.78 (br, 1 H), 6.39 (t, J=2.4 Hz, 1H), 6.27 (d, J=2 Hz, 2H), 3.57 (s, 6H), 2.79 (t, J=8.0 Hz, 2H), 2.35 (t, J=8.0 Hz, 2H). [000171] Following the general procedure for conversion of carboxylic acids to amides mentioned above and using dimethyl amine as amine, 30 was converted to 31 in 98% yield.
[000172] Analysis: 1HNMR (DMSO-d6): δ 7.30 (br, 1H), 7.28 (d, J=8.8 Hz, 2H), 7.23 (d, J=8.8 Hz, 2H), 6.96 (d, J=8.8 Hz, 2H), 6.92 (d, J=8.8 Hz, 2H), 6.79 (br, 1 H), 6.66 (s, 1 H), 6.34 (t, J=2.4 Hz, 1 H), 6.28 (d, J=2 Hz, 2H), 3.68 (s, 6H), 3.05 (br, 3H), 2.90 (br, 3H), 2.77 (t, J=8.0 Hz, 2H), 2.34 (t, J=8.0 Hz, 2H).
Figure imgf000055_0001
30 31
EXAMPLE 14 Synthesis of 2-[4-(4-acetylaminophenoxy)-phenyl]-3-(3,5-dimethoxyphenyl)-N,N- dimethylacrylamide (34)
[000173] Compound 2 was reacted with 1-fluoro-4-nitrobenzene in the presence of NaH in DMF to give 3-(3,δ-dimethoxyphenyl)-2-[4-(4-nitrophenoxy)-phenyl]-acrylic acid (32). Reduction of 32 (10 g, 24 mmol) with zinc dust (16 g, 230 mmol) in acetic acid (100 mL) was accomplished at 120°C for 16 h, the mixture was cooled to room temperature. Water (260 mL) was slowly added to the reaction mixture. The precipitated product was filtered and washed with water (70 mL) to give crude product. The product was recrystallized from toluene. Yield (33): 9.7 g, 94%.
[000174] Analysis: 1HNMR (DMSO-d6): δ 12.36 (br, 1H), 9.96 (s,1H), 7.67 (s, 1 H), 7.60 (d, J=8.8 Hz, 2H), 7.16 (d, J=8.8Hz, 2H), 6.97 (d, J=8.8 Hz, 2H), 6.96 (d, J=8.8 Hz, 2H), 6.34 (t, J=2.8 Hz, 1 H), 6.28 (d, J=2.4 Hz, 2H), 3.68 (S, 6H), 2.03 (s, 3H). [000175] Following the general procedure for conversion of carboxylic acids to amides mentioned above and using dimethylamine as amine, 33 was converted to 34 in 98% yield.
[000176] Analysis: 1HNMR (DMSO-d6): δ 9.96 (s,.1 H), 7.60 (d, J=8.8 Hz, 2H), 7.25 (d, J=8.8Hz, 2H), 6.97 (d, J=8.8 Hz, 2H), 6.93 (d, J=8.8 Hz, 2H), 6.55 (s, 1 H), 6.34 (t, J=2.8 Hz, 1 H), 6.28 (d, J=2.4 Hz, 2H), 3.58 (S, 6H), 3.04 (br, 3H), 2.90 (br, 3H), 2.03 (s, 3H).
Figure imgf000056_0001
32 33 34
EXAMPLE 15 Synthesis of 3-(3,δ-dimethoxyphenyl)-2-[4-(4-methanesulfonylphenoxy)-phenyl]-N,N- dimethylacrylamide (36)
[000177] Compound 2 (3 g, 10 mmol) was dissolved in anhydrous DMF (70 mL) under nitrogen, and potassium carbonate (1.4 g, 10 mol) was added in lots. When the solution became homogeneous, 4-fluorophenyl methyl sulfone (1.74 g, 10 mmol) was added and the mixture was heated at 160°C for 2 h. After cooling to room temperature, the solution was poured into water (160 mL). The mixture was acidified with δ% HCI to - pH 4 and the solidified product was collected by suction filtration. The crude product was recrystallized with toluene. Yield(35): 4.3 g, 96%.
[000178] Analysis: 1HNMR (DMSO-d6): δ 12.72 (br, 1 H), 7.94 (d, J=8.8 Hz, 2H), 7.72 (s, 1 H), 7.80 (d, J=8.4Hz, 2H), 7.18 (d, J=8.8 Hz, 2H), 7.17 (d, J=8.4 Hz, 2H), 6.42 (t, J=2.8 Hz, 1 H), 6.28 (d, J=2.4 Hz, 2H), 3.59 (S, 6H), 3.21 (s, 3H). [000179] Following the general procedure for conversion of carboxylic acids to amides mentioned above and using dimethylamine as amine, 35 was converted to 36 in 96% yield.
[000180] Analysis: 1HNMR (DMSO-d6): δ 7.93 (d, J=8.8 Hz, 2H), 7.38 (d, J=8.4Hz, 2H), 7.17 (d, J=8.8 Hz, 2H), 7.16 (d, J=8.4 Hz, 2H), 6.62 (s, 1 H), 6.36 (t, J=2.8 Hz, 1 H), 6.29 (d, J=2.4 Hz, 2H), 3.69 (S, 6H), 3.20 (s, 3H), 3.08 (br, 3H), 2.92 (br, 3H).
Figure imgf000057_0001
36 36
EXAMPLE 16 Synthesis of 3-(4-{4-[2-(3,δ-dimethoxyphenyl)-1-dimethylcarbamoylvinyl]-phenoxy}- phenyl)-propionic acid ethyl ester (37)
[000181] Following the general procedure for conversion of carboxylic acids to amides mentioned above and using dimethyl amine as amine, 5 was converted to 37 in 97% yield.
[000182] Analysis: 1HNMR (DMSO-d6): δ 7.28 (d, J=8.8 Hz, 2H), 7.23 (d, J=8.8 Hz, 2H), 6.95 (d, J=8.8 Hz, 2H), 6.92 (d, J=8.8 Hz, 2H), 6.56 (s, 1 H), 6.34 (t, J=2.4 Hz, 1 H), 6.28 (d, J=2 Hz, 2H), 4.04 (q, J=6.8 Hz, 2H), 3.68 (s, 6H), 3.06 (br, 3H), 2.90 (br, 3H), 2.84 (t, J=8.4 Hz, 2H), 2.61 (t, J=8.4 Hz, 2H), 1.15 (t, J=6.4 Hz, 3H).
Figure imgf000058_0001
37
EXAMPLE 17
Synthesis of 2-{4-[4-(N-ureido-2-carbamoylethyl)-phenoxy]-phenyl}-3-(3,5- dimethoxyphenyl)-N,N- dimethylacrylamide (39)
[000183] Hydrolysis of 13 with 1 N NaOH yielded 38. The 1 ,1-carbonyl- diimidazole (CDI) derivative was made by the general procedure for conversion of carboxylic acids to amides mentioned above. The CDI intermediate of 38 was converted to 39 by reacting this with semicarbazide in 73% yield.
[000184] Analysis: 1HNMR (DMSO-d6): δ 9.48 (br, 1 H), 7.72 (br, 1 H), 7.28 (d, J=8.8 Hz, 2H), 7.25 (d, J=8.8 Hz, 2H), 6.95 (d, J=8.8 Hz, 2H), 6.92 (d, J=8.8 Hz, 2H), 6.56 (s, 1 H), 6.34 (t, J=2.4 Hz, 1 H), 6.28 (d, J=2 Hz, 2H), 5.86 (s, 2H), 3.58 (s, 6H), 3.05 (br, 3H), 2.90 (br, 3H), 2.77 (t, J=8.0 Hz, 2H), 2.39 (t, J=8.0 Hz, 2H).
Figure imgf000058_0002
38 39 EXAMPLE 18
Synthesis of 3-(3,δ-dimethoxyphenyl)-N,N-dimethyl-2-{4-[4-(3-morpholin-4-yl-3- oxopropyl)-phenoxy]-phenyl}-acrylamide (40)
[000185] The CDI intermediate of 38 was converted to 40 by reacting it with morpholine in 94% yield.
[000186] Analysis: 1HNMR (DMSO-d6): 5 7.27 (d, J=8.8 Hz, 2H), 7.26 (d| J=8.8 Hz, 2H), 6.96 (d, J=8.8 Hz, 2H), 6.92 (d, J=8.8 Hz, 2H), 6.56 (s, 1H), 6.34 (t, J=2.4 Hz, 1H), 6.28 (d, J=2 Hz, 2H), 3.58 (s, 6H), 3.49 (m, 4H), 3.41 (m, 4H), 3.05 (br, 3H), 2.90 (br, 3H), 2.77 (t, J=8.0 Hz, 2H), 2.39 (t, J=8.0 Hz, 2H).
Figure imgf000059_0001
38 40
EXAMPLE 19 Synthesis of 2-(4-{4-[2-(3,5-dimethoxyphenyI)-1-dimethylcarbamoylvinyl]-phenoxy}- benzyl)-malonic acid dimethyl ester (43)
[000187] Condensation of 3 with malonic acid dimethyl ester in the presence of sodium hydride as base resulted in 41, which on reduction with zinc/acetic acid yielded 42. Conversion of 42 to 43 was accomplished by the general procedure for conversion of carboxylic acids to amides mentioned above in 94% yield. Analysis: 1HNMR (DMSO-d6): δ 7.29 (d, J=8.8 Hz, 2H), 7.23 (d, J=8.8 Hz, 2H), 6.96 (d, J=8.8 Hz, 2H), 6.92 (d, J=8.8 Hz, 2H), 6.57 (s, 1 H), 6.34 (t, J=2.4 Hz, 1 H), 6.28 (d, J=2 Hz, 2H), 3.87 (t, J=8 Hz, 1 H), 3.61 (s, 6H), 3.68 (s, 6H), 3.08 (d, J=7.6 Hz, 2H), 3.05 (br, 3H), 2.91 (br, 3H).
Figure imgf000060_0001
41 42 43
EXAMPLE 20 Synthesis of N-{4-[2-(3,5-dimethoxyphenyl)-1 -dimethylcarbamoylvinyl]-phenyl}-3- hydroxybenzamide (44)
[000188] A mixture of 2-(4-aminophenyl)-3-(3,δ-dimethoxyphenyl)-N,N- dimethylacrylamide, 43, (0.59 g, 1.5 mmol), benzotriazol-l-yloxytris-(dimethylamino)- phosphonium hexafluorophosphate (BOP,0.88g, 2.0 mmol), 3-hydroxybenzoic acid (0.28g, 2.0 mmol), triethylamine (0.2 g, 2.0 mmol) in DMF (8.0 mL) was stirred for 3h at room temperature. The reaction mixture was poured in water (δOmL) and solid separated was filtered, dried and purity was checked by HPLC (97.6%).
[000189] Analysis: 1HNMR (DMSO-d6): δ 10.29 (s, 1 H), 9.81 (s, 1 H), 7.79 (d, J=6.8Hz, 2H), 7.43 (d, J=8.0Hz, 1 H), 7.37 (t, J=7.6Hz, 2H), 7.29 (d, J=8.4Hz, 2H), 7.02 (m, 1 H), 6.60 (s, 1 H), 6.40 (t, J=2.0Hz, 1H), 6.36 (d, J=2.0Hz, 2H), 3.63 (s, 6H), 3.08 (brs, 3H), 2.96 (brs, 3H).
Figure imgf000060_0002
EXAMPLE 21 Synthesis of N,N-dimethyl-2-{4-[4-(3-oxo-3-ureidopropenyl)-phenoxy]-phenyl}-3- pyridin-3-ylacrylamide (47)
[000190] Synthesis of 45 from 3-pyridinecarboxaldehyde was performed following Scheme I. Urea (0.78 g, 13 mmol) and 2-{4-[4-(2-ethoxycarbonyl-vinyI)- phenoxy]-phenyl}-3-pridin-3-ylacrylic acid, 45 (0.5 g, 1.2 mmol) was dissolved in sodium ethoxide in ethanol (2M, 6.5 mL, 13 mmol) at 80°C under argon, and the reaction mixture was heated at this temperature for 1 h. The reaction was then quenched by TFA (0.5 mL) after cooling to 5°C. Water (40 mL) was added to the reaction mixture. The crude product was filtered and purified by silica gel chromatography and eluted with hexanes-ethyl acetate (1 :1) containing acetic acid (1%) followed by recrystallization from toluene. Yield (46): 0.33 g, 63%.
[000191] Analysis: 1HNMR (DMSO-d6): δ 12.78 (br, 1 H), 10.29 (s, 1H), 8.42 (dd, J=4.8, 1.6 Hz, 1 H), 8.35 (d, J=2.4 Hz, 1 H), 7.92 (br, 1 H), 7.66 (d, J=16 Hz, 1 H), 7.64 (d, J=8.8 Hz, 2H), 7.36 (tt, J=8.4, 1.6 Hz, 1 H), 7.30 (br, 1 H), 7.28 (m, 1 H), 7.23 (d, J=8.8 Hz, 2H), 7.11 (d, J=8.8 Hz, 2H), 7.09 (d, J=8.8 Hz, 2H), 6.73 (d, J=16 Hz, 1H).
[000192] Following the general procedure for conversion of carboxylic acids to amides mentioned above, 46 was converted to 47.
[000193] Analysis: 1HNMR (DMSO-d6): 5 10.30 (s, 1H), 8.39 (dd, J=4.8, 1.6 Hz, 1H), 8.34 (d, J=2.4 Hz, 1 H), 7.92 (br, 1 H), 7.66 (d, J=16 Hz, 1 H), 7.64 (d, J=8.8 Hz, 2H), 7.45 (tt, J=8.4, 1.6 Hz, 1 H), 7.32 (br, 1 H), 7.29 (d, J=8.8 Hz, 2H), 7.26 (m, 1H), 7.11 (d, J=8.8 Hz, 2H), 7.05 (d, J=8.8 Hz, 2H), 6.73 (d, J=16 Hz, 1 H), 6.70 (s, 1 H), 3.07 (br, 3H), 2.93 (br, 3H).
Figure imgf000061_0001
45 46 47 EXAMPLE 22 Synthesis of 3-(3,δ-dimethoxyphenyl)-2-(4-hydroxyphenyl)-N,N-dimethylacrylamide
(49)
[000194] Following the general procedure for conversion of carboxylic acids to amides mentioned above and using dimethyl amine as amine, 2 was converted to 49.
[000195] Analysis: 1HNMR (DMSO-d6): 5 9.69 (s, 1 H), 7.07 (d, J=8.8, 2H), 6.73 (d, J=8.8Hz, 2H), 6.43 (s, 1 H), 6.23 (t, J=2.4Hz, 1 H), 6.29 (d, J=2.4Hz, 2H), 3.57 (s, 6H), 2.99 (brs, 3H), 2.89 (brs, 3H).
Figure imgf000062_0001
49
EXAMPLE 23
Synthesis of [3-(4-{4-[2-(3,5-dimethoxyphenyl)-1-(piperidine-1-carbonyl)-vinyl]- phenoxy}-phenyl)-propionyl]-urea (51 )
[000196] Following the general procedure for conversion of carboxylic acids to amides mentioned above and using piperidine as amine, 6 was converted to 51.
[000197] Analysis: 1HNMR (DMSO-d6): δ 10.16 (s, 1 H), 7.73 (brs, 1 H), 7.26 (d, J=8.8 Hz, 2H), 7.23 (d, J=8.8Hz, 2H), 6.98 (d, J=8.8Hz, 2H), 6.93 (d, J=8.8Hz, 2H), 6.56 (s, 1H), 6.34 (t, J=2.4Hz, 1 H), 6.29 (d, J=2.4Hz, 2H), 3.58 (s, 6H), 3.50 (br, 4H), 2.82 (t, J=7.6Hz, 2H), 2.59 (t, J=7.6Hz, 2H), 1.68 (br, 2H) 1.40-1.46 (br, 4H).
Figure imgf000063_0001
6 51
EXAMPLE 24 Synthesis of 3-(3,5-dimethoxyphenyl)-N,N-diethyl-2-{4-[4-(3-oxo-3-ureidopropyl)- phenoxy]-phenyl}-acrylamide (53)
[000198] Following the general procedure for conversion of carboxylic acids to amides mentioned above and using diethylamine as amine, 6 was converted to 53.
[000199] Analysis: 1HNMR (DMSO-d6): δ 10.17 (s, 1 H), 7.70 (brs, 1 H), 7.26 (overlapped d, J=8.8Hz, 2H), 7.23 (overlapped d, J=8.8Hz, 2H), 6.97 (d, J=8.8Hz, 2H), 6.92 (d, J=8.8Hz, 2H), 6.54 (s, 1 H), 6.34 (t, J=2.0Hz, 1 H), 6.29 (d, J=2.0Hz, 2H), 3.32-3.37 (br, 4H), 3.59 (s, 6H), 2.82 (t, J=7.6Hz, 2H), 2.59 (t, J=7.6Hz, 2H), 1.03 (br, 3H), 0.92 (br, 3H).
Figure imgf000063_0002
53 EXAMPLE 25
Synthesis of 2-{4-[4-(3-acetylamino-3-oxopropyl)-phenoxy]-phenyl}-3-(4- fluorophenyl)-N,N-dimethylacrylamide (56)
[000200] To a solution of {4-[4-(2-carbamoylethyl)-phenoxy]-phenyl}-acetic acid, 54, (0.45g, l .δmmol) in acetic anhydride (15 mL) was added 4-fluorobenzaldehyde (0.17 mL, 1.6 mmol) and potassium acetate (0.17g, 1.8 mmol) and refluxed overnight. Reaction mixture was poured in water (50 mL) and extracted with ethyl acetate (2 x 50mL). The crude product was purified by silica gel chromatography to yield 55.
[000201] Analysis: 1HNMR (DMSO-d6): δ 12.50 (br, 1 H), 10.64 (s, 1H), 7.74 (s,1H), 7.27 (d, J=8.4Hz, 2H), 7.10-7.15 (m, 6H), 6.99 (d, J=8.4Hz, 2H), 6.97 (d, J=8.4Hz, 2H), 2.81 (d, J=6.8Hz, 2H), 2.76 (d, J=6.8Hz, 2H), 2.15 (s, 3H).
[000202] Following the general procedure for conversion of carboxylic acids to amides mentioned above and using dimethylamine as amine, 55 was converted to 56.
[000203] Analysis: 1HNMR (DMSO-d6): δ 10.62 (s, 1 H), 7.26 (d, J=8.4Hz, 2H), 7.22 (d, J=8.4Hz, 2H), 7.15 (d, J=8.4Hz, 2H), 7.06 (d, J=8.4Hz, 2H), 6.97 (d, J=8.0Hz, 2H), 6.94 (d, J=8.0Hz, 2H), 6.63 (s,1 H), 2.81 (d, J=6.8Hz, 2H), 2.76 (d, J=6.8Hz, 2H), 2.16 (s, 3H).
Figure imgf000064_0001
EXAMPLE 26
Synthesis of 2-(4-{4-[2-(3,5-dimethoxyphenyl)-1 -dimethylcarbamoylvinylj-phenoxy}- benzyl)-malonic acid (58) and 2-(4-{4-[2-(3,5-dimethoxyphenyl)-1- dimethylcarbamoylvinyl]-phenoxy}-benzyl)-malonamide (59)
[000204] To a solution of 2-(4-{4-[2-(3,δ-dimethoxyphenyl)-1- dimethylcarbamoylvinyl]-phenoxy}-benzyl)-malonic acid dimethyl ester, 43 (0.40g, 0.73mmol) in DMF (6 mL) and ethanol (10 mL), ammonium hydroxide (20 mL, 28%) and IN NaOH (0.36 mL, 0.36 mmol) was added and stirred overnight at room temperature. Solvent was evaporated and the crude product was purified by silica gel chromatography to yield 58 and 59.
Analysis: 1HNMR (DMSO-d6 + D20) of 58: δ7.20 (d, J=8.4Hz, 2H), 7.17 (d, J=8.4Hz, 2H), 6.90 (d, J=8.4Hz, 2H), 6.81 (d, J=8.4Hz, 2H), 6.51 (s, 1 H), 6.29 (t, J=2.0Hz, 1 H), 6.21 (d, J=2.0Hz, 2H), 3.53 (s, 6H), 3.13 (br, 1H), 3.01 (brs, 3H), 2.92 (br, 2H), 2.86 (brs, 3H).
[000205] Analysis: 1HNMR (DMSO-d6) of 59: 5 5 7.28 (d, J=8.8 Hz, 2H), 7.26 (br, 2H), 7.22 (d, J=8.8 Hz, 2H), 7.03 (br, 2H), 6.97 (d, J=8.8 Hz, 2H), 6.90 (d, J=8.8 Hz, 2H), 6.56 (s, 1 H), 6.34 (t, J=2.4 Hz, 1 H), 6.28 (d, J=2 Hz, 2H), 3.58 (s, 6H), 3.29 (t, J=8 Hz, 1 H), 3.05 (br, 3H), 2.95 (d, J=7.6 Hz, 2H), 2.91 (br, 3H).
Figure imgf000065_0001
Figure imgf000065_0002
Figure imgf000065_0003
EXAMPLE 27 Synthesis of 3-(3,5-dimethoxyphenyl)-2-{4-[4-(3-oxo-3-ureidopropyl)- phenoxy]- phenyl}-N-pyridin-4-ylacrylamide (60)
[000206] Following the general procedure for conversion of carboxylic acids to amides mentioned above and using 4-aminopyridine as amine, 6 was converted to 60.
[000207] Analysis: 1HNMR (DMSO-d6): δ10.17 (s, 1H), 8.24 (brs, 1H), 7.71 (br, 2H), 7.63 (d, J=8.8Hz, 2H), 7.44 (s, 1 H), 7.2δ (d, J=8.4Hz, 2H), 7.22 (br, 1 H), 7.03 (d, J=9.2Hz, 2H), 7.99 (d, J=8.4Hz, 2H), 6.47 (d, J=2.4Hz, 2H), 6.43 (t, J=2.4Hz, 2H), 3.65 (s, 6H), 2.83 (t, J=7.6Hz, 2H), 2.60 (t, J=7.6Hz, 2H).
Figure imgf000066_0001
60
EXAMPLE 28
Synthesis of N-(4-chlorophenyl)-3-(3,5-dimethoxyphenyl)-2-{4-[4-(3-oxo-3- ureidopropyl)-phenoxy]-phenyl}-acrylamide (61 )
[000208] Following the general procedure for conversion of carboxylic acids to amides mentioned above and using 4-chloroaniline as amine, 6 was converted to 61.
[000209] Analysis: 1HNMR (DMSO-d6): δ 10.16 (s, 1 H), 8.24 (brs, 1 H), 7.65 (brs, 1 H), 7.53 (d, J=8.8Hz, 2H), 7.44 (s, 1 H), 7.25 (d, J=8.8Hz, 2H), 7.22 (br, 1 H), 7.03 (d, J=8.8Hz, 2H), 7.00 (d, J=8.8Hz, 2H), 6.47 (d, J=2.4Hz, 2H), 6.43 (d, J=2.4Hz, 1 H), 3.66 (s, 6H), 2.83 (t, J=8.0Hz, 2H), 2.60 (t, J=8.0Hz, 2H).
Figure imgf000067_0001
61
EXAMPLE 29
Synthesis of 3-(3,5-dimethoxyphenyl)-N,N-dimethyl-2-(4-{4-[2-(2-morpholin-4-yl-2- oxoethylcarbamoyl)-ethyl]-phenoxy}-phenyl)-acrylamide (63)
[000210] Following the general procedure for conversion of carboxylic acids to amides mentioned above and using 2-amino-1-morpholin-4-yl-ethanone as amine, 3- (4-{4-[2-(3,5-dimethoxyphenyl)-1-dimethylcarbamoylvinyl]-phenoxy}-phenyl)- propionic acid, 38, was converted to 63.
[000211] Analysis: 1 HNMR (DMSO-d6): δ 7.99 (t, J=δ.6Hz, 1 H), 7.27 (d, J=8.8Hz, 2H), 7.24 (d, J=8.8Hz, 2H), 6.97 (d, J=8.8Hz, 2H), 6.92 (d, J=8.8Hz, 2H), 6.66 (s, 1 H), 6.34 (t, J=2.0Hz, 1 H), 6.28 (d, J=2.0Hz, 2H), 3.93 (d, J=5.6Hz, 2H) 3.56 (s, 6H), 3.52-3.56 (m, 4H), 3.40-3.42 (m, 4H), 3.05 (brs, 3H), 2.91 (brs, 3H), 2.80 (t, J=7.6Hz, 2H), 2.46 (t, J=7.6Hz, 2H).
Figure imgf000067_0002
38 63 EXAMPLE 30
Synthesis of 3-(3,5-dimethoxyphenyl)-N,N-dimethyl-2-(4-{4-[3-(4-methylpiperazin-1- yl)-3-oxopropyl]-phenoxy}-phenyl)-acrylamide (64)
[000212] Following the general procedure for conversion of carboxylic acids to amides mentioned above and using 4-methylpiperazine as amine, 3-(4-{4-[2-(3,5- dimethoxyphenyl)-1 -dimethylcarbamoylvinyI]-phenoxy}-phenyl)-propionic acid, 38, was converted to 64.
[000213] Analysis: 1HNMR (DMSO-d6): δ 7.28 (d, J=2.8Hz, 2H), 7.26 (d, J=2.8Hz, 2H), 6.96 (d, J=8.8Hz, 2H), 6.92 (d, J=8.6Hz, 2H), 6.56 (s, 1 H), 6.34 (t, J=2.0Hz, 1H), 6.28 (d, J=2.0Hz, 2H), 6.19 (s, 6H), 3.40(dt, 2=18.0 and 4.8Hz), 3.04 (brs, 3H), 2.90 (brs, 3H), 2.79 (t, J=8.0, 2H), 2.60 (t, J=8.0Hz, 2H), 2.20 (t, J=5.2Hz, 2H), 2.14 (s, 3H).
Figure imgf000068_0001
38 64
EXAMPLE 31 Synthesis of 3-(3,5-dimethoxyphenyl)-N,N-dimethyl-2-[4-(pyridin-2-yIoxy)-phenyl]- acrylamide (66)
[000214] A solution of 3-(3,5-dimethoxyphenyl)-2-(4-hydroxyphenyl)-acrylic acid, 2, (0.6 g, 2.0 mmol), 2-fluoropyridine (0.19 g, 2.0 mmol) in dimethyl acetamide (4.0 mL) was heated in presence of potassium carbonate (0.28 g, 2.0 mmol) at 175°C for 2 h, and then quenched with water (2δ mL), neutralized with dilute HCI and extracted with ethyl acetate (2 x 50mL). Organic layer was dried and evaporated. The crude product was purified by silica gel chromatography to yield 65 (0.15 g, 19.9%). [000215] A mixture of 3-(3,δ-dimethoxyphenyl)- 2-[4-(pyridin-2-yloxy)-phenyl]- acrylic acid, 65, (0.11g, 0.3 mmol), benzotriazol-l-yloxytris-(dimethylamino)- phosphonium hexafluorophosphate (BOP, 0.16 g, 0.35 mmol), dimethylamine in THF (2M, 0.5 mL, 1.0 mmol), triethylamine ( 0.035 g, 035 mmol) in DMF (6.0 mL) was stirred for 3 h at room temperature. The reaction mixture was poured in water (50.0 mL) and extracted with ethyl acetate (2 x 50 mL). Solvent was evaporated under reduced pressure and residue was purified by silica gel chromatography to yield 66. [000216] Analysis: 1HNMR (DMSO-d6): δ 8.14 (m, 1 H), 7.88 (m, 1 H), 7.33 (d, J=8.8 Hz, 2H), 7.14 (m, 3H), 7.05 (d, J=8.4Hz, 2H), 6.59 (s, 1H), 6.34 (t, J=2.0Hz, 1 H), 6.31 (d, J=2.0Hz, 2H), 3.58(s, 6H), 3.10 (brs, 3H), 2.92 (brs, 3H).
Figure imgf000069_0001
65 66
EXAMPLE 32 Measurement of Increased Glucose Uptake in 3T3-L1 Adipocytes Treated With a
Compound of the Present Invention
[000217] The effect of treatment with 1 on glucose uptake was measured in 3T3- L1 differentiated adipocytes. The assay was conducted essentially according to the method of Tafuri SR, Endocrinology, 137, 4706-4712 (1996). The adipocytes were incubated with different concentrations of the test compound for 48 hours in Dulbecco's modified Eagle's medium (DMEM) containing 10% fetal bovine serum (FBS), then washed and incubated in glucose-free, serum-free medium for 60 minutes at 37°C. Then 14C-deoxyglucose was added and the cells were incubated for 30 minutes at room temperature. After washing, the cells were lysed (0.1% SDS) and the radioactivity was measured to determine the amount of glucose uptake. Glucose uptake was calculated as a percentage of the basal level seen in cells not treated with drug. As shown in FIG. 1 , treatment with 1 resulted in a dose-dependent increase in glucose uptake.
EXAMPLE 33
Measurement of Enhanced Glucose Uptake in 3T3-L1 Adipocytes Treated With
Insulin in Combination with a Compound of the Present Invention
[000218] The ability of 1 to enhance insulin-stimulated glucose uptake was assessed in 3T3-L1 adipocytes essentially as described above in Example 32. Adipocytes were incubated with either vehicle (0.1% DMSO) or test compound (δ μM 1) for 48 hours in DMEM plus 10% FBS. The cells were then serum-starved, incubated for 30 minutes with different concentrations of insulin, and then glucose uptake was carried out for 10 minutes at room temperature. When compared to treatment with vehicle, treatment with 1 enhanced the stimulation of glucose uptake by insulin (see Figure 2).
EXAMPLE 34
Measurement of the Glucose-Lowering Effect in ob/ob Mice Treated With a
Compound of the Present Invention
[000219] The glucose-lowering effect of 1 was measured in ob/ob mice, an animal model for type 2 diabetes. At the onset of diabetes, seven-week-old male ob/ob mice received daily oral doses of either vehicle (0.6% CMC) or 1 (10 mg/kg) by gavage for seven days. Blood glucose levels were measured on day 0 (24 hours prior to administration of the first dose), day 1 (immediately prior to the first dose), and on days 2, 4, 6 and 8 (24 hours following administration of the prior dose). Significant decreases in blood glucose levels were recorded on day 6 (36% decrease, p < 0.06) and day 8 (23% decrease, p < O.Oδ) in the drug-treated versus the vehicle-treated animals (see Figure 3). EXAMPLE 35 Measurement of the Lipid-Lowering Effects in ob/ob Mice Treated With a Compound of the Present Invention
[000220] The lipid-lowering effects of 1 also were measured in ob/ob mice following one week of treatment. In the experiment described above in Example 34, the concentrations of serum triglycerides and free fatty acids were determined on day 8. Significant decreases were observed in the levels of serum triglycerides (49% lower, p < 0.05) and free fatty acids (19% lower, p < O.Oδ) in the drug-treated versus the vehicle treated mice (see Figure 4).
EXAMPLE 36
Measurement of the Inhibition of LPS-induced TNF-alpha Production in RAW264.7
Cells Treated With a Compound of the Present Invention
[000221] The ability of 1 to inhibit LPS-induced TNF-alpha production was assessed in the mouse macrophage cell line RAW264.7. The RAW cells were preincubated with either 1 μM dexamethasone (Dex) or 10, 30 or 100 μM 1 for 1 hour at 37°C in RPMI-1640 containing 10% FBS. After 1 hour LPS (0.1 μg/ml) was added and cells were incubated an additional 6 hours. Cell supernatant was then collected, aliquoted and frozen at -70°C, and an aliquot used to determine the concentration of TNF-alpha by ELISA. As shown in Figure 5, treatment with 1 significantly inhibited the LPS-induced production of TNF-alpha. The inhibitory effect approached that seen with dexamethasone.
EXAMPLE 37
Measurement of the Inhibition of LPS-induced IL-1 Beta Production in RAW264.7
Cells Treated With a Compound of the Present Invention
[000222] The ability of 1 to inhibit LPS-induced IL-1 beta production was also examined in RAW264.7 cells. The RAW cells were preincubated with either 1 μM dexamethasone (Dex) or 10, 30 or 100 μM 1 for 1 hour at 37°C in RPMI-1640 containing 10% FBS. After 1 hour LPS (0.1 μg/ml) was added and cells were incubated an additional 6 hours. Cell supernatant was then collected, aliquoted and frozen at -70°C, and an aliquot used to determine the concentration of IL-1 beta by ELISA. As shown in Figure 6, treatment with 1 significantly inhibited the LPS-induced production of IL-1 beta. The inhibition seen with 1 was of the same approximate magnitude as that seen with dexamethasone.
EXAMPLE 38
Measurement of the Inhibition of LPS-induced IL-6 Production in RAW264.7 Cells
Treated With a Compound of the Present Invention
[000223] The ability of 1 to inhibit LPS-induced IL-6 production was also measured in RAW264.7 cells. The RAW cells were preincubated with either 1 μM dexamethasone (Dex) or 10, 30 or 100 μM 1 for 1 hour at 37°C in RPMI-1640 containing 10% FBS. After 1 hour LPS (0.1 μg/ml) was added and cells were incubated an additional 6 hours. Cell supernatant was then collected, aliquoted and frozen at -70°C, and an aliquot used to determine the concentration of IL-6 by ELISA. As shown in Figure 7, treatment with 1 significantly inhibited the LPS-induced production of IL-6. The inhibitory effect was greater than that observed with dexamethasone.
EXAMPLE 39
Measurement of the Inhibition of LPS-induced iNOS and COX-2 Production in
RAW264.7 Cells Treated With a Compound of the Present Invention
[000224] The ability of 1 to inhibit LPS-induced production of iNOS and COX-2 was also measured in RAW264.7 cells. The RAW cells were preincubated with either 1 μM dexamethasone (Dex) or 10, 30 or 100 μM 1 (Test Cpd) or other reference compound (Ref Cpd A or Ref Cpd B) for 1 hour at 37°C in RPMI-1640 containing 10% FBS. After 1 hour LPS (0.1 μg/ml) was added and cells were incubated an additional 6 hours. Cells receiving no drug treatment, incubated with or without LPS, served as controls. Cells were lysed and 25 μg/well of total protein was electrophoresed on 4-20% Tris-glycine SDS gels. Proteins were transferred to nitrocellulose membrane, and the resulting blot was probed with antibody to iNOS, then stripped and reprobed with antibody to COX-2, and then stripped and reprobed with antibody to COX-1. As shown in Figure 8, treatment with 1 exhibited dose- dependent inhibition of LPS-induced iNOS production. In addition, treatment with 1 selectively inhibited production of COX-2 but not COX-1 in LPS-stimulated cells.
EXAMPLE 40
Inhibition of LPS-induced TNF-alpha Release by Human Monocytes With
Compounds of the Present Invention
[000225] Frozen human elutriated monocytes (Advanced Biotechnologies Incorporated) were thawed and each 1-ιml vial mixed with -12 ml of 10% FBS complete medium (10% heat-inactivated fetal bovine serum in RPMI 1640 medium supplemented with 100 U/ml penicillin, 100 μg/ml streptomycin and 50 μM 2- mercaptoethanol). Cells were centrifuged at 800 rpm for 10 min at room temperature using a Beckman GS-6 centrifuge with GH-3.8 rotor, and the cell pellets were resuspended in 20% FBS complete medium (20% heat-inactivated FBS in RPMI 1640 medium supplemented with 100 U/ml penicillin, 100 μg/ml streptomycin and 50 μM 2-mercaptoethanol) and centrifuged again at 800 rpm for 10 min at room temperature. Cell pellets were resuspended in 20% FBS complete medium, and the cell suspensions were pooled and passed through a 70-micron cell strainer to remove any aggregates or clumps. The cell suspension was adjusted to 2.5 x 106 cells/ml in 20% FBS complete medium. Cell suspension (160 μl, 4 x 105 cells) was added into each well of a 96-well tissue-culture treated polystyrene plate and incubated at 37°C for 1 - 2.5 h. Cells were pretreated with vehicle (DMSO) or test compound (0.3, 1.0, 3.0, 10 or 30 μM) in 20% FBS complete medium for 1 h at 37°C. After pretreatment, lipopolysaccharides (LPS) from Salmonella typhimurium in 20% FBS complete medium were added to the cells. The final concentrations were 0.1% DMSO and 10 ng/ml LPS in a final volume of 200 μl/well. The cells were incubated for 20 h at 37°C, and then the supernatants were harvested and aliquots of the supernatants frozen at -80°C for subsequent analysis. Cells on the plates were assayed for cell viability using the MTS/PMS assay (Cory AH et al., Cancer Commun 3:207-212, 1991). The concentration of TNF-alpha in the cell supernatants was determined using quantitative sandwich enzyme immunoassay for human TNF- alpha (R&D Systems). The mean percent inhibition of TNF-alpha release relative to vehicle was calculated for each concentration of test compound from multiple determinations. As shown in Table 2, the compounds of the invention caused significant inhibition of LPS-induced TNF-alpha release by human monocytes.
TABLE 2
Figure imgf000075_0001
Cell viability < 70%
EXAMPLE 41 Stimulation of Glucose Uptake in 3T3-L1 Adipocytes With Compounds of the Present
Invention
[000226] Differentiation of mouse 3T3-L1 adipocytes was carried out using the method of Greenberg AS, et al., J Biol Chem 276:45456-61, 2001. Briefly, 3T3-L1 fibroblasts were differentiated to adipocytes by incubation in DMEM containing 10% FBS, 72 μg/ml porcine insulin, 0.5 mM 3-isobutylmethylxanthine (IBMX) and 400 ng/ml dexamethasone for 2 x 48 h at 37°C. Differentiated cells were maintained in media containing 10% FBS without insulin, IBMX or dexamethasone until they were used for experiments. The effect of treatment with compounds of the invention on glucose uptake by differentiated adipocytes was assessed essentially according to the method of Tafuri SR, Endocrinology 137:4706-12, 1996. Adipocytes were incubated with vehicle (0.1 % DMSO) or test compound (0.1 , 1.0 or 10 μM) for 48 h in DMEM containing 10% FBS, then washed and incubated in high-glucose, serum- free medium for 3 h at 37°C. Cells were then washed, incubated for 20 min in glucose-free, serum-free medium containing 100 nM insulin, then supplemented with 2.5 μCi/ml 14C-deoxyglucose in 0.1 mM cold deoxyglucose and further incubated for 10 min at room temperature. After washing, cells were lysed with 0.5% SDS and the radioactivity was measured in a scintillation counter to determine the amount of glucose uptake. The mean percent stimulation of glucose uptake relative to vehicle (set at 100%) was calculated for each concentration of test compound from triplicate determinations. As shown in Table 3, the compounds of the invention caused significant stimulation of glucose uptake in differentiated adipocytes.
TABLE 3
Test Percent Stimulation of Glucose Uptake
Compound 0.1 μM 1.0 μM 10 μM
31 107% 119% 161%
8 115% 132% 171%
60 93% 120% 229%
61 93% 120% 229%
51 93% 107% 136%
29 106% 124% 120%
40 126% 117% 126%
63 107% 112% 139%
64 108% 113% 127%
56 83% 100% 126%
EXAMPLE 42 Inhibition of PDE4 and PDE3 Activity With a Compound of the Present Invention
[000227] Compound 13 was examined for its ability to inhibit the activity of PDE4 and PDE3 enzymes. PDE4 partially purified from human U-937 promonocytic cells and PDE3 partially purified from human platelets were used. Test compound (1, 10 or 30 μM) or vehicle (0.1% DMSO) was incubated with 0.2 μg PDE4 enzyme or 1 μg PDE3 enzyme and 1 μM cAMP containing 0.01 μg [3H]cAMP in Tris buffer pH 7.5 for 20 min at 30°C. The reaction was terminated by boiling for 2 min and the resulting AMP was converted to adenosine by addition of 10 mg/ml snake venom nucleotidase and further incubation at 30°C for 10 min. Unhydrolyzed cAMP was bound to AGI-X2 resin, and remaining [3H]adenosine in the aqueous phase was quantitated by scintillation counting. The mean percent inhibition of PDE4 or PDE3 activity was calculated from duplicate determinations (Table 4). Compound 13 exhibited significant inhibition of both PDE4 (IC5o < μM) and PDE3 (IC50 = 13.6 μM) enzyme activities. TABLE 4
Enzyme Percent Inhibition of Enzyme Activity
Assay 1 μM 10 μM 30 μM
PDE4 86% 98% 102% PDE3 20% 62% 66%
EXAMPLE 43 Inhibition of LPS-induced Phosphorylation of p44/42 MAP Kinase With a Compound of the Present Invention
[000228] Compound 13 was examined for its ability to inhibit LPS-induced and LPS/IFN-gamma induced phosphorylation of p44/42 MAP kinase. RAW 264.7 gamma NO(-) cells were seeded at 1 x 106/well (2 ml per well) in 6-well tissue culture plates one day prior to the experiment. To start the experiment, cells were washed 2X with RPMI 1640 medium, 0.5% FBS, 100 U/ml penicillin, 100 μg/ml streptomycin, 1mM sodium pyruvate, and then pretreated with vehicle (0.1 % DMSO) or test compound (10 or 30 μM) at 37°C for 1 h. After pretreatment, cells were incubated in RPMI 1640 medium, 10% FBS, 100 U/ml penicillin, 100 μg/ml streptomycin, 1mM sodium pyruvate, containing 10 ng/ml LPS or LPS (10 ng/ml)/IFN-gamma (10 U/ml) at 37°C for 15 min. Cells were then washed 2X with cold PBS (pH 7.4) and lysed in 20 mM Tris-HCI (pH 7.5), 150 mM NaCI, 1 mM Na2EDTA, 1 mM EGTA, 1% Triton, 2.6 mM sodium pyrophosphate, 1 mM beta-glycerophosphate, 1 mM Na3VO4, 1 μg/ml leupeptin, 1 mM PMSF on ice for 10 min. Lysed cells were collected and centrifuged at -20,800 x g for 10 min at 4°C. Supernatants (lysates) were collected, aliquoted, and stored frozen at -80°C until use. Lysates (29 μg of total proteins per sample) were subjected to SDS-polyacrylamide (4-20%) gel electrophoresis, and the separated proteins were transferred to nitrocellulose membranes. Membranes were blocked with 5% non-fat dry milk, 10 mM Tris-HCI (pH 8.0), 160 mM NaCI, 0.1% Tween®-20 at room temperature for 1 h and then were blotted with mAb against phospho-p44/42 MAP kinase (Thr 202/Tyr 204) at room temperature for 1 h. The membranes were then washed and incubated with a horseradish peroxidase-linked anti-mouse secondary antibody at room temperature for 1 h. The signals were detected using ECL Western blotting detection reagents. The results showed that compound 13 inhibited LPS-induced phosphorylation of p44/42 MAP kinase at 30 μM but not 10 μM, while the compound inhibited LPS/IFN-gamma induced phosphorylation of p44/42 in a dose-dependent manner at 30 μM and 10 μM (data not shown).
EXAMPLE 44 Inhibition of Anti-CD3/Anti-CD28 Stimulated Lymphocyte Proliferation With a
Compound of the Present Invention
[000229] Compound 13 was examined for its ability to inhibit anti-CD3/anti-CD28 stimulated lymphocyte proliferation. Binding of antigen, or antibodies, to CD3/CD28 triggers activation and proliferation of T-lymphocytes, which are key steps involved in mounting an immune response (Abbas, Lichtman and Pober, Cellular and Molecular Immunology, 3rd edition, W.B. Saunders Company, Philadelphia, 1997). Mesenteric lymph nodes were collected from BALB/c mice (female, -8 weeks old), and the cells were isolated in PBS (pH 7.4) and mixed with culture medium (RPMI 1640 medium, 10% FBS, 100 U/ml penicillin, 100 μg/ml streptomycin, 50 μM 2-mercaptoethanol). The cell suspension was centrifuged at 900 rpm for 10 min at room temperature using a Beckman GS-6 centrifuge with GH-3.8 rotor. After centrifugation, cell pellets were resuspended in culture medium and centrifuged again at 900 rpm for 10 min at room temperature. Cell pellets were resuspended in culture medium and cells were counted. 2 x 105 lymph node cells per well were added into a 96-well cell culture plate. For the treatment (n=4), vehicle (DMSO) or test compound was added into cells. Cells were treated with purified hamster anti-mouse CD3ε (2 μg/ml) and anti- mouse CD28 (0.2 μg/ml) monoclonal antibodies or with culture medium. The final concentrations were 0.1 % DMSO and 10 μM test compound in a final volume of 200 μl per well. Cells were incubated at 37°C for 67 h, and then cells on plates were centrifuged at 900 rpm for 10 min at room temperature using a Beckman GS-6 Centrifuge with GH-3.8 rotor. 150 μl of supernatant from each well was subsequently harvested and frozen at -80°C for further analysis (ELISA). For the cells on the plate, 150 μl of culture medium was added into each well to replace the harvested supernatants and 40 μl of MTS/PMS solution was added into each well. After further incubation at 37°C for 140 min, the plate was read at 505 nm in a microplate spectrophotometer. The O.D. values (after subtracting the mean O.D. of blank wells) were used to compare the proliferation of treated cells. As shown in Table 5, 10 μM of compound 13 caused about 60% inhibition of the proliferation of mouse mesenteric lymph node cells stimulated by anti-CD3/anti-CD28 monoclonal antibodies. Inhibition of CD3/CD28 mediated T-cell proliferation demonstrates compound 13 is able to block an immunologically-relevant cellular response, probably via interactions with a step in the signal transduction cascade. This indicates that compound 13 has immunomodulatory activity, which may be useful for the treatment of immunoproliferative disorders.
TABLE 5
Treatment P.P. (Mean ± SD)
DMSO 0.020 ± 0.006 DMSO + anti-CD3/anti-CD28 mAbs 1.372 ± 0.125
Test compound + anti-CD3/anti-CD28 mAbs 0.578 ± 0.012
EXAMPLE 45 Improvement of Collagen Induced Arthritis in Mice Using a Compound of the Present
Invention
[000230] Collagen-induced arthritis (CIA) was induced in 45 DBA/1 J mice using immunization with chicken collagen Type II. The induction schedule was as follows: on Day 0, 100 μg/100 μl in Complete Freund's Adjuvant (CFA) intradermally; on Day 21 , 100 μg/100 μl in Incomplete Freund's Adjuvant subcutaneously; on Day 31 , 100 μg/100 μl in CFA subcutaneously; all given at the base of the tail. On Day 35 animals received lipopolysaccharides (detoxified from E. coli serotype O111 :B4; 40 μg/mL) intraperitoneally. When signs of arthritis appeared, mice were assigned into four treatment groups: vehicle control (0.6% carboxymethylcellulose (CMC)); compound 31 (40 mg/kg suspension in CMC); compound 31 (100 mg/kg in CMC); positive control (dexamethasone; δ mg/kg). The animals were dosed per oral by gavage, twice daily for 14 days, at a dose volume of 260 μl per mouse per dose. The study was scored blindly to the different treatment groups. Mice were weighed and arthritis was scored three times a week. Arthritis was scored as a count of affected limbs and digits, evaluated as: erythema and swelling of tarsal, the ankle to the metatarsal joints, up to restriction of movement and deformity of the joints. Plasma was collected from the animals 4 hours following the final dose, for measurement of circulating drug levels. At termination, animals were euthanized and hind limbs removed for histopathologic examination, hind limbs were collected in formalin. Decalcified tissue was sectioned longitudinally, parallel to the bones, and hematoxylin and eosin stained sections were scored using a standard rheumatoid arthritis scoring system by a veterinary histopathologist who was blinded to the treatment groups. Animals in all groups had moderate arthritis prior to the start of dosing (Day 0) as shown in Figure 9. The vehicle group exhibited an increase in severity over the course of the study with a tendency to plateau from about Day 10. The low dose of compound 31 had no apparent effect on the animals compared with vehicle controls. The high dose prevented the increase in severity, to about the same extent as dexamethasone. Histology showed that the vehicle group and the low-dose compound 31 group had marked chronic inflammation of synovium with pannus formation and destruction of bone and cartilage, while in the dexamethasone group the joints were within normal limits. At high dose of compound 31 there was a reduction in incidence and severity of pannus formation, inflammation cell infiltration and bone/cartilage damage. Thus a dose-dependent effect of compound 31 was observed on both the soft tissue and bone and cartilage, consistent with a disease- modifying activity of the compound in this model.
[000231] It will be evident from the above that the compounds according to the present invention not only lower blood glucose level, triglyceride level and free fatty acid level in diabetic conditions, but also inhibit TNF-alpha, IL-6, IL-1 beta, COX-2 and iNOS production in inflammation, as well as inhibit PDE4 and PDE3 activity, phosphorylation of p44/42 MAP kinase and lymphocyte proliferation. The properties demonstrated above indicate that the compounds of the invention should be useful in the treatment of disorders associated with insulin resistance, hyperglycemia, hyperiipidemia, coronary artery disease and peripheral vascular disease and for the treatment of inflammation, inflammatory diseases, immunological diseases, proliferative diseases and cancer, especially those mediated by cytokines, cyclooxygenase, phosphodiesterase and/or MAP kinase. EXAMPLE 46 Synthesis of N-{4-[2-(3,5-dimethoxyphenyl)-1 -dimethylcarbamoylvinylj-phenyl}- benzamide (67)
[000232] A mixture of 2-(4-aminophenyl)-3-(3,5-dimethoxyphenyl)-N,N- dimethylacrylamide, 43, (0.49g, 1.2 mmol) and benzoyl chloride (0.26 g, 1.8 mmol) in anhydrous benzene (18.0 mL) was heated at 90°C for 2 h. Solvent was evaporated and crude product was purified by silica gel chromatography. [000233] Analysis: 1HNMR (DMSO-d6): δ 10.33 (s, 1 H), 7.96 (d, J=8.8Hz, 2H), 7.76 (d, J=8.8Hz, 1 H), 7.51-7.62 (m, 3H), 7.26 (d, J=9.2Hz, 2H), 6.55 (s, 1H), 6.35 (t, J=2.0Hz, 1 H), 6.31 (d, J=2.0Hz, 2H), 3.58 (s, 6H), 3.03 (brs, 3H), 2.91 (brs, 3H).
Figure imgf000081_0001
EXAMPLE 47 Synthesis of 3-{4-[4-(2-benzo[1 ,3]dioxol-5-yl-1 -dimethylcarbamoylvinyl)-phenoxy]- phenylj-propionic acid ethyl ester (69)
[000234] A mixture of 3-{4-[4-(2-ethoxycarbonylethyl)-phenoxy]-phenyl}-2- oxopropionic acid, 24 (1.0 g, 3.0 mmol), piperonal (0.67 g, 0.4δ mmol), triethylamine (6.12 mL) and acetic anhydride (δ mL) was heated at 80°C for 3 h. Reaction mixture was poured in water (60 mL). Solid separated was filtered and boiled in toluene, cooled and filtered. Crude solid was purified by silica gel chromatography to yield 68, 0.3δg (yield, 26.0%).
[000235] A mixture of 4-benzo[1 ,3]dioxol-δ-yl-3-{4-[4-(2-ethoxycarbonylethyl)- phenoxy]-phenyl}-2-oxobut-3-enoic acid, 68, (0.08 g, 0.17 mmol), benzotriazol-1- yloxytris-(dimethylamino)-phosphonium hexafluorophosphate (BOP, 0.09 g, 0.21 mmol), triethylamine (36 μL, 0.2δ mmol), dimethylamine in THF (2M, 0.26 mL, 0.5 mmol) in DMF (2.0 mL) was stirred for 10 min at room temperature. Reaction mixture was poured in water (20 mL). Solid separated was filtered and boiled in toluene, cooled and filtered. Crude solid was purified by silica gel chromatography to yield 69. Analysis: 1HNMR (DMSO-d6): δ 7.24 (d, J=8.8Hz, 4H), 6.95 (overlapped d, J=8.8Hz, 4H), 6.80 (d, J=8.0Hz, 1H), 6.68 (d, J=8.0Hz, 1H), 6.54 (s, 1H), 6.61 (s, 1H), 5.96 (s, 2H), 4.05 (q, J=4.0Hz, 2H), 3.05 (brs, 3H), 2.85 (brs, 3H), 2.80 (t, J=6.0Hz, 2H), 2.60 (t, J=6.0Hz, 2H) and 1.15 (t, J=4.0Hz, 3H).
Figure imgf000082_0001
EXAMPLE 48 Synthesis of 2-{4-[4-(1 -dimethylcarbamoyl-2-pyridin-3-ylvinyl)-phenoxy]-benzyl}- malonamide (71)
[000236] To a solution of 2-{4-[4-(1-dimethylcarbamoyl-2-pyridin-3-ylvinyl)- phenoxy]-benzyl}-malonic acid dimethyl ester, 70 (0.49 g, 1.0 mmol), in DMF (δ mL), ammonium hydroxide (28% in water, 12 mL) was added and stirred overnight at room temperature. Reaction mixture was poured in water (30 mL) and extracted with chloroform (δ x 26 mL). The organic layer was dried on anhydrous magnesium sulfate and evaporated. The crude product was purified by silica gel chromatography to yield 71, 0.23g (yield, 24.6%).
[000237] Analysis: 1HNMR (CDCI3 + CD3OD): δ 8.32 (m, 2H), 7.40 (m, 1 H), 7.18 (overlapped d, J= 8.0 Hz, 2H), 7.20 (overlapped d, J= 8.0 Hz, 2H), 7.12 (m,1 H), 6.92 (d, J=8.0Hz, 2H), 6.84 (d, J=8.0Hz, 2H), 6.60 (s, 1 H), 3.22 (d, J=12.0 Hz), 3.12 (brd, J=12.0 Hz), 2.98 (brs, 3H), 2.96 (brs, 3H).
Figure imgf000083_0001
70 71
[000238] It will be appreciated that various modifications may be made in the invention as described above and as defined in the following claims wherein:

Claims

What is claimed is:
1. A compound having a structure selected from Formulas l-XIII:
Figure imgf000084_0001
Figure imgf000085_0001
wherein the stereocenters marked with an asterisk (*) are R- or S-; the bond represented by a dashed line plus a solid line is a double bond or a single bond; and wherein Riι R2, 3, R4, R5, Rε and R7 are each independently selected from the group consisting of
H; optionally substituted C1-C20 linear or branched alkyl, chloroalkyl or fluoroalkyl; optionally substituted C2-C20 linear or branched alkenyl; optionally substituted C6-C2o aryl, linear or branched alkylaryl, linear or branched alkenylaryl; COOR where R is H, optionally substituted O1-C20 alkyl, optionally substituted C2-C2o alkenyl or optionally substituted C6-C10 aryl, sodium, potassium, calcium, magnesium, ammonium, tromethamine; CONR'R", where R' and R" are independently H, optionally substituted C-r C2o alkyl, optionally substituted C2-C2o alkenyl or optionally substituted Cβ- C 0 aryl or where NR'R" represents a cyclic moiety selected from morpholine, piperidine, piperazine; optionally substituted Ci-Cβ amidoalkyl; NH2; Cι-C2o alkylamino, bis(alkylamino), cycloalkylamino or cyclic amino; OH; optionally substituted Cι-C2o alkoxy, optionally substituted C C2o alkanoyl; optionally substituted C1-C20 acyloxy; halo; optionally substituted C1-C20 alkylcarboxylamino; cyano; nitro; S02NR"'R"" where R'" and R"" are independently H, Cι-C20 alkyl or aryl; SO2R"' where R'" is H, C1-C20 alkyl or aryl; SO3R'" where R'" is H, C1-C20 alkyl or aryl; and C4-C8 heterocycles selected from tetrazolyl, imidazolyl, pyrrolyl, pyridyl and indolyl;
Rs and Rg are each independently selected from the group consisting of
H; optionally substituted C1-C20 linear or branched alkyl; optionally substituted C2-C2o linear or branched alkenyl; optionally substituted C6-C10 aryl or heteroaryl; COOR where R is H, optionally substituted C1-C20 alkyl, optionally substituted C2-C2o alkenyl or optionally substituted C6-Cι0 aryl, sodium, potassium, calcium, magnesium, ammonium, or tromethamine; CONR'R", where R' and R" are independently H, alkoxy, optionally substituted C1-C20 alkyl, optionally substituted C2-C2o alkenyl, optionally substituted C3-C10 cycloalkyl or cycloalkenyl or optionally substituted C6-Cι0 aryl or heteroaryl, or where NR'R" represents a cyclic moiety selected from morpholine, piperidine, hydroxypiperidine, imidazole, piperazine, or methylpiperazine; NH2; C1-C20 alkylamino, bis(alkylamino), cycloalkylamino or cyclic amino; OH; C C^o alkoxy; C1-C20 alkanoyl; C-ι-C20 acyloxy; halo; C1-C20 alkylcarboxylamino; cyano; nitro; SO2NR"'R"" where R'" and R"" are independently H, C1-C20 alkyl or aryl; S02R"' where R'" is H, Cι-C20 alkyl or aryl; S03R,M where R'" is H, C1- C20 alkyl or aryl; and tetrazolyl;
R10 and R11 are each independently selected from the group consisting of
H; optionally substituted Cι-C2o linear or branched alkyl; optionally substituted C2-C20 linear or branched alkenyl; optionally substituted C6-Cιo aryl or heteroaryl; COOR where R is H, optionally substituted C1-C20 alkyl, optionally substituted C2-C20 alkenyl or optionally substituted Cβ-Cio aryl, sodium, potassium, calcium, magnesium, ammonium, or tromethamine; CONR'R", where R' and R" are independently H, optionally substituted C -C2o alkyl, optionally substituted C2-C2o alkenyl or optionally substituted Cβ-Cio aryl or where NR'R" represents a cyclic moiety selected from morpholine, piperidine, or piperazine; NH2; C1-C20 alkylamino, bis(alkylamino), cycloalkylamino or cyclic amino; OH; C1-C20 alkoxy; C1-C20 alkanoyl; C1-C20 acyloxy; halo; C1-C20 alkylcarboxylamino; cyano; nitro; SO2NR'"R"" where R'" and R"" are independently H, C1-C20 alkyl or aryl; SO2R'" where R'" is H, C C2o alkyl or aryl; SO3R'" where R'" is H, C1-C20 alkyl or aryl; and tetrazolyl;
R12, Ri3> Ri8> R19 and R20 are each independently selected from the group consisting of
H; optionally substituted Cι-C20 linear or branched alkyl; optionally substituted C2-C20 linear or branched alkenyl; optionally substituted C6-Cιo aryl or heteroaryl; COOR where R is optionally substituted C1-C20 alkyl, optionally substituted C2-C2o alkenyl or optionally substituted C6-C10 aryl, sodium, potassium, calcium, magnesium, ammonium, or tromethamine; CONR'R", where R' and R" are independently H, optionally substituted C1-C20 alkyl, optionally substituted C2-C2o alkenyl or optionally substituted Cβ-Cio aryl or where NR'R" represents a cyclic moiety selected from morpholine, piperidine and piperazine ; C1-C20 alkanoyl; C1-C20 alkylamido; C6-C2o aroyl or heteroaroyl; SO2R'" where R'" is H, C^o alkyl or aryl; morpholinocarbonylmethyl; piperazinocabonylmethyl; and piperadinocabonylmethyl; R12 and R 3 may be absent, or Rι2 and Rι3 together may be an optionally substituted heterocyclic ring selected from morpholine, piperidine, piperazine, and N- methylpiperidine;
R14 is selected from the group consisting of
H; optionally substituted Cι-C2o linear or branched alkyl including chloroalkyl and fluoroalkyl; optionally substituted C2-C2o linear or branched alkenyl; optionally substituted Cβ-Cio aryl or heteroaryl; COOR where R is H, optionally substituted C1-C20 alkyl, optionally substituted C2-C20 alkenyl or optionally substituted Cβ-Cio aryl, sodium, potassium, calcium, magnesium, ammonium, or tromethamine ; CONR'R", where R' and R" are independently H, optionally substituted C1-C20 alkyl, optionally substituted C2-C20 alkenyl or optionally substituted Cβ-Cio aryl or where NR'R" represents a cyclic moiety selected from morpholine, piperidine and piperazine; cyano; and tetrazolyl;
R15, R16 and Rι7 are each independently selected from the group consisting of
H; optionally substituted Cι-C2o linear or branched alkyl including chloroalkyl and fluoroalkyl; optionally substituted C2-C2o linear or branched alkenyl; optionally substituted C6-Cιo aryl or heteroaryl; COOR where R is H, optionally substituted C1-C20 alkyl, optionally substituted C2-C20 alkenyl or optionally substituted Cβ-Cio aryl, sodium, potassium, calcium, magnesium, ammonium, and tromethamine; CONR'R", where R' and R" are independently H, optionally substituted C C2o alkyl, optionally substituted C2-C2o alkenyl or optionally substituted C6-Cιo aryl or where NR'R" represents a cyclic moiety selected from morpholine, piperidine, and piperazine; NH2; C1-C20 alkylamino, bis(alkylamino), cycloalkylamino or cyclic amino; OH; C1-C20 alkoxy; C1-C20 alkanoyl; C -C2o acyloxy; halo; C1-C20 alkylcarboxylamino; cyano; nitro; SO2NR,"R"" where R'" and R"" are independently H, C1-C20 alkyl or aryl; SO2R,M where R'" is H, C C2o alkyl or aryl; SO3R'" where R'" is H, C1-C20 alkyl or aryl; and tetrazolyl;
X is independently selected from
O; N; S; S=O; SO2; or NR"'", where R""' may be H or optionally substituted C1-C20 alkyl, optionally substituted C2-C2o alkenyl, optionally substituted d- C2o acyl, optionally substituted Cι-C2o acyloxy and optionally substituted Cr C2o alkoxycarbonyl;
Y is independently selected from O, S or NH;
Z is selected from the group consisting of
ORa, wherein Ra is selected from the group consisting of H; optionally substituted C1-C20 linear or branched alkyl, chloroalkyl or fluoroalkyl, optionally substituted C2-C20 linear or branched alkenyl; optionally substituted C6-Cι0 aryl or heteroaryl; optionally substituted C6-C2o aroyl or heteroaroyl; optionally substituted C1-C20 alkanoyl; and SO2R'" where R"' is H, C1-C-20 alkyl or aryl;
NRbRc, wherein Rb and Rc are independently selected from the group consisting of H; optionally substituted C1-C20 linear or branched alkyl, chloroalkyl or fluoroalkyl ; optionally substituted C2-C2o linear or branched alkenyl; optionally substituted C6-C 0 aryl or heteroaryl; optionally substituted C3-C10 cycloalkyl or cycloalkenyl; COOZ1 where Z-i is optionally substituted C1-C20 alkyl, optionally substituted C2-C20 alkenyl or optionally substituted C-6-C10 aryl; optionally substituted C6-C2o aroyl or heteroaroyl; optionally substituted Cι-C20 alkanoyl; and SO2R'" where R'" is H, C1-C20 alkyl or aryl; and wherein Rb and Rc together may be joined to form a 3-6 membered ring selected from aziridine, morpholine, piperidine and piperazine; and
CRdReRf, wherein R , Re and Rf are each independently selected from the group consisting of H; optionally substituted C1-C20 linear or branched alkyl, chloroalkyl or fluoroalkyl, optionally substituted C2-C20 linear or branched alkenyl; optionally substituted C6-C10 aryl or heteroaryl; optionally substituted C3-C10 cycloalkyl or cycloalkenyl; COOR where R is H, optionally substituted C1-C20 alkyl, optionally substituted C2-C20 alkenyl or optionally substituted C6-C 0 aryl, sodium, potassium, calcium, magnesium, ammonium, or tromethamine; NH2; C1-C20 alkylamino, bis(alkylamino); cycloalkylamino or cyclic amino; OH; optionally substituted C1-C20 alkoxy, trifluoromethoxy; optionally substituted C1-C20 alkanoyl; optionally substituted C1-C20 acyloxy; optionally substituted C6-C2o aroyl or heteroaroyl; halo; cyano; nitro; optionally substituted C1-C20 alkylcarboxylamino; S02NR"'R"" where R'" and R"" are independently H, C1-C20 alkyl or aryl; SO2R'" where R'" is H, C1-C20 alkyl or aryl; and SO3R'" where R'" is H, C-1-C-20 alkyl or aryl; or the grouping -C (=Y)Z may represent hydrogen or R12 or may be absent;
Q is selected from the group consisting of
ORa where Ra is selected from the group consisting of H; optionally substituted Cι-C2o linear or branched alkyl, chloroalkyl or fluoroalkyl; optionally substituted C2-C2o linear or branched alkenyl; optionally substituted Cδ-Cio aryl or heteroaryl; optionally substituted C6-C2o aroyl or heteroaroyl; optionally substituted C1-C20 alkanoyl; and SO2R"1 where R"' is H, C1-C20 alkyl or aryl; and
NRbRc where Rb and Rc are independently selected from the group consisting of H; optionally substituted C1-C20 linear or branched alkyl, chloroalkyl or fluoroalkyl; optionally substituted C2-C20 linear or branched alkenyl; optionally substituted C6-Cι0 aryl or heteroaryl; optionally substituted C3-C10 cycloalkyl or cycloalkenyl; COOZ1 where Z1 is optionally substituted C1-C20 alkyl, optionally substituted C2-C2o alkenyl or optionally substituted C6-Cιo aryl; optionally substituted C6-C2o aroyl or heteroaroyl; optionally substituted C C2o alkanoyl; and SO2R'" where R'" is H, d- C2o alkyl or aryl; and wherein Rb and Rc together may be joined to form a 3-6 membered ring such as aziridine, morpholine, piperidine, piperazine and the like; and
SRg, SORg or SO2Rg where Rg is selected from the group consisting of H; optionally substituted C1-C20 linear or branched alkyl, chloroalkyl or fluoroalkyl; optionally substituted C2-C2o linear or branched alkenyl; optionally substituted C1-C20 acyl; optionally substituted C1-C20 alkoxycarbonyl; C2-C20 alkoxy; optionally substituted C6-Cι0 aryl or heteroaryl; and optionally substituted C6-Cι0 aroyl or heteroaroyl.
Group A is optionally substituted C2-C2o linear or branched alkenyl; optionally substituted C6-C2o aryl, linear or branched alkylaryl, linear or branched alkenylaryl; optionally substituted heteroaryl selected from pyridine, indole, morpholine, piperidine, tetrazolyl and piperazine; COR where R is optionally substituted C-ι-C2o linear or branched alkyl; optionally substituted C2-C o linear or branched alkenyl; optionally substituted C6- C2o aryl, linear or branched alkylaryl, linear or branched alkenylaryl; optionally substituted heteroaryl selected from pyridine, indole, morpholine, piperidine, piperazine, and tetrazolyl;
Group B is OH, C1-C20 alkoxy; SO2R where R may be H or linear or branched C1-C20 alkyl;
Group Het represents a heterocyclic ring selected from pyridyl, indolyl, tetrazolyl, imidazolyl, morphonyl, piperidinyl, piperazinyl and thiophenyl.
2. The compound of Claim 1 wherein the bond represented by a dashed line plus a solid line is a double bond and is in the E or Z configuration.
3. The compound of Claim 1 wherein the bond represented by a dashed line plus a solid line is a single bond wherein the resulting stereocenters have the R- or S- configuration.
4. The compound of Claim 1 wherein at least one aromatic ring possesses more than one adjacent substituent, and wherein the substituents are joined to form a ring.
5. The compound of Claim 1 wherein Rs and Rg together are joined to form a C4-C8 heterocyclic ring.
6. The compound of Claim 1 wherein R10 and Rn together are joined to form a C4-Cs heterocyclic ring.
7. The compound of Claim 1 wherein Rd and Re together are joined to form a 3-6 membered ring and wherein the resulting stereocenter has an R- or S- configu ration.
8. The compound of Claim 1 which is of Formula I.
9. The compound of Claim 8 selected from the group consisting of:
3-(3,5-Dimethoxyphenyl)-2-{4-[4-(3-oxo-3-ureidopropyl)-phenoxy]- phenyl}-acrylic acid methyl ester;
3-(3,5-Dimethoxyphenyl)-2-{4-[4-(3-oxo-3-ureido-propyl)-phenoxy]- phenylj-acrylic acid;
3-(3,δ-Dimethoxyphenyl)-2-{4-[4-(3-ethoxycarbonylamino-3-oxo- propyl)-phenoxy]-phenyl}-acrylic acid methyl ester;
2-{4-[4-(3-Benzoyloxycarbonylamino-3-oxopropyl)-phenoxy]-phenyl}-3- (3,δ-dimethoxyphenyl)-acrylic acid methyl ester;
3-(3,δ-Dimethoxyphenyl)-2-{4-[4-(3-oxo-3-ureidopropyl)-phenoxy]- phenyl}-propionic acid;
3-(3,δ-Dimethoxyphenyl)-2-{4-[4-(3-oxo-3-ureidopropenyl)-phenoxy]- phenylj-acrylic acid;
3-(3,5-Dimethoxyphenyl)-2-{4-[4-(3-oxo-3-ureidopropyI)-phenoxy]- phenyl}-acrylic acid ethyl ester;
3-(3,5-Dimethoxyphenyl)-Λ/,Λ/-dimethyl-2-{4-[4-(3-oxo-3-ureido-propyl)- phenoxy]-phenyl}-acrylamide; and
2-(4-{4-[3-(3-Cyclohexylureido)-3-oxopropyl]-phenoxy}-phenyl)-3-(3,5- dimethoxyphenyl)-acrylic acid.
10. The compound of Claim 1 which is of Formula II.
11. The compound of Claim 10 selected from the group consisting of
[3-(4-Phenoxyphenyl)-propionyl]-urea; and {3-[4-(4-Methoxyphenoxy)-phenyl]-acryloyl}-urea.
12. The compound of Claim 1 which is of Formula III..
13. The compound of Claim 12 selected from the group consisting of 2-{4-[4-(3-Acetyiureidomethyl)-phenoxy]-phenyl}-3-(3,δ-dimethoxy-phenyl)- acrylic acid methyl ester; and
2-{4-[4-(3-Acetylthioureidomethyl)-phenoxy]-phenyl}-3-(3,δ-dimethoxy- phenyl)-acrylic acid.
14. The compound of Claim 1 which is of formula IV.
16. The compound of Claim 14 selected from the group consisting of he
1 -Acetyl-3-[4-(4-methoxyphenoxy)-benzyl]-urea; and 1-Acetyl-3-[4-(3,4-dimethoxyphenoxy)-benzyl]-urea.
16. A composition comprising the compound of Claim 1 and a pharmaceutically acceptable carrier.
17. A method of treatment of diabetes comprising the co-administration of the compound of Claim 1 and an agent selected from the group consisting of: insulin or an insulin mimetic, a sulfonylurea or other insulin secretagogue, a thiazolidinedione, a fibrate or other PPAR-alpha agonist, a PPAR-delta agonist, a biguanide, a statin or other hydroxymethylglutaryl (HMG) CoA reductase inhibitor, an alpha-glucosidase inhibitor, a bile acid-binding resin, apoA1 , niacin, probucol, and nicotinic acid.
18. A method of treatment of inflammatory or immunlogical disease comprising the co-administration of the compound of Claim 1 and an agent selected from the group consisting of: a nonsteroidal anti-inflammatory drug (NSAID), a cyclooxygenase-2 inhibitor, a corticosteroid or other immunosuppressive agent, a disease-modifying antirheumatic drug (DMARD), a TNF-alpha inhibitor, other cytokine inhibitor, other immune modulating agent, and a narcotic agent.
19. A method of treatment of inflammatory or immunological disease comprising the step of administering to a subject in need of such treatment a therapeutically effective amount of the compound of Claim 1.
20. The method of claim 19 wherein the compound is administered in combination with an agent selected from the group consisting of NSAID, cyclooxygenase-2 inhibitor, immunosuppressive agent, DMARD, TNF- alpha inhibitor, cytokine inhibitor, immune modulating agent and a narcotic agent.
21. The method of Claim 20 wherein the agent is corticosteroid or methotrexate.
22. A method of treatment for diabetes comprising the step of administering to a subject suffering from a diabetic condition a therapeutically effective amount of the compound of Claim 1.
23. The method of claim 20 wherein the compound is administered in combination with an agent selected from the group consisting of: insulin, insulin mimetic, insulin secretagogue, PPAR-gamma agonist, PPAR-alpha agonist, a PPAR-delta agonist, biguanide, HMG CoA reductase inhibitor, alpha-glucosidase inhibitor, bile acid-binding resin, apoAI , niacin, probucol and nicotinic acid.
24. The method of Claim 21 wherein the agent is selected from the group consisting of sulfonylurea, thiazolidinedione, fibrate, and statin.
26. The method of claim 22, wherein the agent is sulfonylurea.
26. A method of inhibiting the activity of TNF-alpha, IL-1 , IL-6, PDE4, PDE3, p44/42 MAP kinase, iNOS or COX-2 comprising administering to a host in need of such inhibition a therapeutically effective amount of the compound of Claim 1.
27. A method of inhibiting the undesired action of cytokine, phosphodiesterase, MAP kinase or cyclooxygenase comprising administering to a host in need of such treatment a therapeutically effective amount of the compound of Claim 1.
28. A method of treating hyperiipidemia comprising administering to a host in need of such treatment a therapeutically effective amount of the compound of Claim 1.
29. A method of treating coronary heart disease comprising administering to a host in need of such treatment a therapeutically effective amount of the compound of Claim 1.
30. A method of treating cancer or proliferative disease comprising administering to a host in need of such treatment a therapeutically effective amount of the compound of Claim 1.
31. The method of Claim 19 wherein the inflammatory or immunological disease is selected from the group consisting of: rheumatoid arthritis, osteoarthritis, ankylosing spondylitis, psoriasis, psoriatic arthritis, asthma, acute respiratory distress syndrome, chronic obstructive pulmonary disease, and multiple sclerosis.
32. A compound of the formula
Figure imgf000096_0001
wherein the compound is 3-(3,5-Dimethoxyphenyl)-2-{4-[4-(3-oxo-3- ureidopropyl)-phenoxy]-phenyl}-acrylic acid methyl ester.
33. A compound of the formula
Figure imgf000096_0002
wherein the compound is 3-(3,5-Dimethoxyphenyl)-/V,/V-dimethyl-2-{4-[4- (3-oxo-3-ureidopropyl)-phenoxy]-phenyl}-acrylamide.
34. An anti-inflammatory composition comprising an effective amount of a compound selected from the group consisting of:
3-(3,δ-Dimethoxyphenyl)-Λ/,/\/-dimethyl-2-{4-[4-(3-oxo-3-ureidopropyl)- phenoxy]-phenyl}-acrylamide (13); 2-{4-[4-(2-Carbamoylethyl)-phenoxy]-phenyl}-3-(3,δ-dimethoxyphenyl)-N,N- dimethylacrylamide (31);
3-(4-{4-[2-(3,δ-Dimethoxyphenyl)-1-dimethylcarbamoylvinyl]-phenoxy}- phenyl)-propionic acid ethyl ester (37);
N-{4-[2-(3,δ-Dimethoxyphenyl)-1-dimethylcarbamoylvinyl]-phenyl}-3- hydroxybenzamide (44);
3-(3,δ-Dimethoxyphenyl)-2-(4-hydroxyphenyl)-N,N-dimethylacrylamide (49); [3-(4-{4-[2-(3,δ-Dimethoxyphenyl)-1-(piperidine-1-carbonyl)-vinyl]-phenoxy}- phenyl)-propionyl]-urea (51);
2-{4-[4-(3-Acetylamino-3-oxopropyl)-phenoxy]-phenyl}-3-(4-fluorophenyl)-N,N- dimethylacrylamide (56);
2-(4-{4-[2-(3,δ-Dimethoxyphenyl)-1-dimethylcarbamoylvinyl]-phenoxy}- benzyl)-malonic acid (58);
2-(4-{4-[2-(3,δ-Dimethoxyphenyl)-1-dimethylcarbamoylvinyl]-phenoxy}- benzyl)-malonamide (59);
3-(3,δ-Dimethoxyphenyl)-N,N-dimethyl-2-[4-(pyridin-2-yloxy)-phenyl]- acrylamide (66);
N,N-dimethyl-2-{4-[4-(3-oxo-3-ureidopropenyl)-phenoxy]-phenyl}-3-pyridin-3- ylacrylamide (47);
N-{4-[2-(3,δ-Dimethoxyphenyl)-1-dimethylcarbamoyl-vinyl]-phenyl}-benzamide (67) ;
2-{4-[4-(1-Dimethylcarbamoyl-2-pyridin-3-yl-vinyl)-phenoxy]-benzyl}- malonamide (71);
3-{4-[4-(2-Benzo[1 ,3]dioxol-δ-yl-1-dimethylcarbamoyl-vinyl)-phenoxy]-phenyl}- propionic acid ethyl ester (69);
3-Benzo[1 ,3]dioxol-δ-yl-2-{4-[4-(2-carbamoylethyl)-phenoxy]-phenyl}-N,N- dimethyl-acrylamide (72);
N,N-Dimethyl-2-{4-[4-(3-oxo-3-ureidopropyl)-phenoxy]-phenyl}-3-pyridin-3-yl- acrylamide (73)
36. An antidiabetic composition comprising an effective amount of a compound selected from the group consisting of:
3-(3,5-Dimethoxyphenyl)-2-{4-[4-(3-ethoxycarbonylamino-3-oxo-propyl)- phenoxy]-phenyl}-acrylic acid methyl ester (8); (4-{4-[2-(3,5-Dimethoxyphenyl)-1-dimethylcarbamoyl-vinyl]-phenoxy}-benzyl)- carbamic acid methyl ester (29);
2-{4-[4-(2-CarbamoylethyI)-phenoxy]-phenyl}-3-(3,δ-dimethoxyphenyl)-N,N- dimethylacrylamide (31);
3-(3,δ-Dimethoxyphenyl)-N,N-dimethyl-2-{4-[4-(3-morpholin-4-yl-3-oxopropyl)- phenoxy]-phenyl}-acrylamide (40);
[3-(4-{4-[2-(3,5-Dimethoxyphenyl)-1-(piperidine-1-carbonyl)-vinyl]-phenoxy}- phenyl)-propionyl]-urea (51);
2-{4-[4-(3-Acetylamino-3-oxopropyl)-phenoxy]-phenyl}-3-(4-fluorophenyl)-N,N- dimethylacrylamide (56);
3-(3,δ-Dimethoxyphenyl)-2-{4-[4-(3-oxo-3-ureidopropyl)- phenoxy]-phenyl}-N- pyridin-4-ylacrylamide (60);
N-(4-Chlorophenyl)-3-(3,5-dimethoxyphenyl)-2-{4-[4-(3-oxo-3-ureidopropyl)- phenoxy]-phenyl}-acrylamide (61);
3-(3,δ-Dimethoxyphenyl)-N,N-dimethyl-2-(4-{4-[2-(2-morpholin-4-yl-2- oxoethylcarbamoyl)-ethyl]-phenoxy}-phenyl)-acrylamide (63); 3-(3,5-Dimethoxyphenyl)-N,N-dimethyl-2-(4-{4-[3-(4-methylpiperazin-1-yl)-3- oxopropyl]-phenoxy}-phenyl)-acrylamide (64).
36. 2-{4-[4-(2-Carbamoylethyl)-phenoxy]-phenyl}-3-(3,δ-dimethoxyphenyl)-N,N- dimethylacrylamide (31).
PCT/US2002/038150 1999-11-08 2002-11-27 Compounds for treatment of inflammation, diabetes and related disorders WO2003048108A2 (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
CN028271009A CN1615295B (en) 2001-11-29 2002-11-27 Compounds for treatment of inflammation, diabetes and related disorders
EP02804467A EP1448515A2 (en) 2001-11-29 2002-11-27 Compounds for treatment of inflammation, diabetes and related disorders
MXPA04005168A MXPA04005168A (en) 2001-11-29 2002-11-27 Compounds for treatment of inflammation, diabetes and related disorders.
NZ533645A NZ533645A (en) 2001-11-29 2002-11-27 Compounds for treatment of inflammation, diabetes and related disorders
CA2468302A CA2468302C (en) 2001-11-29 2002-11-27 Compounds for treatment of inflammation, diabetes and related disorders
KR1020047008107A KR100941197B1 (en) 2001-11-29 2002-11-27 Compounds for treatment of inflammation, diabetes and related disorders
AU2002357032A AU2002357032B2 (en) 2001-11-29 2002-11-27 Compounds for treatment of inflammation, diabetes and related disorders
US10/430,677 US7323496B2 (en) 1999-11-08 2003-05-07 Compounds for treatment of inflammation, diabetes and related disorders
US12/004,039 US20080108825A1 (en) 1999-11-08 2007-12-20 Compounds for treatment of inflammation, diabetes and related disorders
US12/004,064 US20080103302A1 (en) 2000-02-04 2007-12-20 Compounds for treatment of inflammation, diabetes and related disorders
US12/004,075 US20080188654A1 (en) 2001-11-29 2007-12-20 Compounds for treatment of inflammation, diabetes and related disorders

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US33481801P 2001-11-29 2001-11-29
US60/334,818 2001-11-29

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/777,551 Continuation-In-Part US20020002200A1 (en) 1999-11-08 2001-02-05 Novel diphenylethylene compounds

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US10430667 Continuation-In-Part 2003-05-05
US10/430,677 Continuation-In-Part US7323496B2 (en) 1999-11-08 2003-05-07 Compounds for treatment of inflammation, diabetes and related disorders

Publications (2)

Publication Number Publication Date
WO2003048108A2 true WO2003048108A2 (en) 2003-06-12
WO2003048108A3 WO2003048108A3 (en) 2003-10-16

Family

ID=23308958

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2002/038150 WO2003048108A2 (en) 1999-11-08 2002-11-27 Compounds for treatment of inflammation, diabetes and related disorders

Country Status (9)

Country Link
EP (1) EP1448515A2 (en)
KR (1) KR100941197B1 (en)
CN (1) CN1615295B (en)
AU (2) AU2002357032B2 (en)
CA (1) CA2468302C (en)
MX (1) MXPA04005168A (en)
NZ (2) NZ563604A (en)
SG (1) SG165988A1 (en)
WO (1) WO2003048108A2 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004019986A1 (en) * 2002-08-29 2004-03-11 Schering Aktiengesellschaft Methods of treating acute respiratory distress syndrome
EP1625207A2 (en) * 2003-05-07 2006-02-15 Theracos, Inc. Compounds for treatment of inflammation, diabetes and related disorders
WO2009045397A1 (en) * 2007-10-02 2009-04-09 Stowers Institute For Medical Research Methods for treating polycystic kidney desease (pkd) or other cyst forming diseases
WO2012030165A2 (en) 2010-08-31 2012-03-08 서울대학교산학협력단 Use of the fetal reprogramming of a ppar δ agonist
WO2012060594A2 (en) * 2010-11-05 2012-05-10 숙명여자대학교산학협력단 Anti-inflammatory composition containing a thiourea compound and a pharmaceutically acceptable salt thereof as an active ingredient

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8034921B2 (en) 2006-11-21 2011-10-11 Alnylam Pharmaceuticals, Inc. IRNA agents targeting CCR5 expressing cells and uses thereof
JP2013538862A (en) * 2010-10-07 2013-10-17 サズセ アーペーエス Anti-diabetic enol-type glucoside of phenylpyruvic acid

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2254549A1 (en) * 1973-12-12 1975-07-11 Takeda Chemical Industries Ltd 3-(Substd. phenyl)-2-chloropropionic acid derivs - hypolipidaemics and hypoglycaemics
WO2001034094A2 (en) * 1999-11-08 2001-05-17 Calyx Therapeutics, Inc. Novel compounds to treat diabetes and associated conditions

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR970006472B1 (en) * 1987-05-07 1997-04-28 오노 화아마슈티칼 캄파니 리미팃드 Novel cinnamoylamide derivatives
US5783593A (en) * 1993-11-04 1998-07-21 Abbott Laboratories Inhibitors of squalene synthetase and protein farnesyltransferase
DE19527305A1 (en) * 1995-07-26 1997-01-30 Hoechst Ag Substituted cinnamic acid guanidides, process for their preparation, their use as a medicament or diagnostic agent and medicament containing them
UA74781C2 (en) * 1999-04-02 2006-02-15 Abbott Lab Antiinflammatory and immumosuppressive compounds inhibiting cell adhesion
DE19941764A1 (en) * 1999-09-02 2001-03-15 Aventis Pharma Gmbh Substituted acylguanidines, processes for their preparation, their use as medicines or diagnostics and medicines containing them
WO2001017614A2 (en) * 1999-09-07 2001-03-15 Conjuchem, Inc. Methods and compositions containing succinimide or maleimide derivatives of antineoplastic agents
EP1218336A2 (en) * 1999-09-20 2002-07-03 Takeda Chemical Industries, Ltd. Melanin concentrating hormone antagonist
DE10024319A1 (en) * 2000-05-17 2001-11-22 Merck Patent Gmbh New bis-acylguanidine compounds are subtype-1 cellular sodium ion-proton antiporter inhibitors useful e.g. for treating arrhythmia, angina pectoris, infarction and insulin-independent diabetes mellitus

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2254549A1 (en) * 1973-12-12 1975-07-11 Takeda Chemical Industries Ltd 3-(Substd. phenyl)-2-chloropropionic acid derivs - hypolipidaemics and hypoglycaemics
WO2001034094A2 (en) * 1999-11-08 2001-05-17 Calyx Therapeutics, Inc. Novel compounds to treat diabetes and associated conditions

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004019986A1 (en) * 2002-08-29 2004-03-11 Schering Aktiengesellschaft Methods of treating acute respiratory distress syndrome
EP1625207A2 (en) * 2003-05-07 2006-02-15 Theracos, Inc. Compounds for treatment of inflammation, diabetes and related disorders
EP1625207A4 (en) * 2003-05-07 2008-11-12 Theracos Inc Compounds for treatment of inflammation, diabetes and related disorders
US8007790B2 (en) 2006-04-03 2011-08-30 Stowers Institute For Medical Research Methods for treating polycystic kidney disease (PKD) or other cyst forming diseases
WO2009045397A1 (en) * 2007-10-02 2009-04-09 Stowers Institute For Medical Research Methods for treating polycystic kidney desease (pkd) or other cyst forming diseases
WO2012030165A2 (en) 2010-08-31 2012-03-08 서울대학교산학협력단 Use of the fetal reprogramming of a ppar δ agonist
WO2012060594A2 (en) * 2010-11-05 2012-05-10 숙명여자대학교산학협력단 Anti-inflammatory composition containing a thiourea compound and a pharmaceutically acceptable salt thereof as an active ingredient
WO2012060594A3 (en) * 2010-11-05 2012-06-28 숙명여자대학교산학협력단 Anti-inflammatory composition containing a thiourea compound and a pharmaceutically acceptable salt thereof as an active ingredient

Also Published As

Publication number Publication date
AU2002357032A1 (en) 2003-06-17
KR100941197B1 (en) 2010-02-10
CN1615295B (en) 2010-11-03
CA2468302C (en) 2012-08-14
MXPA04005168A (en) 2005-02-17
AU2002357032B2 (en) 2009-01-08
SG165988A1 (en) 2010-11-29
NZ563604A (en) 2009-04-30
CA2468302A1 (en) 2003-06-12
CN1615295A (en) 2005-05-11
EP1448515A2 (en) 2004-08-25
KR20040091609A (en) 2004-10-28
NZ533645A (en) 2008-04-30
AU2009201342A1 (en) 2009-04-30
WO2003048108A3 (en) 2003-10-16

Similar Documents

Publication Publication Date Title
US20080188654A1 (en) Compounds for treatment of inflammation, diabetes and related disorders
JP4609691B2 (en) Heterocyclic compounds and pharmaceutical uses thereof
US7718682B2 (en) Heterocyclic analogs of diphenylethylene compounds
AU2009201342A1 (en) Compounds for treatment of inflammation, diabetes and related disorders
US20150018331A1 (en) Antibacterial agents
JP2001294586A (en) Piperazine as material for inhibiting fructose-1,6- bisphosphatase (fbp-ase)
US7105552B2 (en) Heterocyclic analogs of diphenylethylene compounds
KR20020019971A (en) Antibacterial agents
KR20080023758A (en) Novel derivatives of amino acids for treatment of obesity and related disorders
US20080103302A1 (en) Compounds for treatment of inflammation, diabetes and related disorders
US20080108825A1 (en) Compounds for treatment of inflammation, diabetes and related disorders
EP0808309B1 (en) Imidazole derivatives, their preparation and their use as s-adenosylmethionine decarboxylase (=samdc) inhibitors
RU2295520C2 (en) DERIVATIVES OF OXAZOLE AND THEIR USING AS INSULIN SENSITIZING AGENTS, PHARMACEUTICAL COMPOSITION POSSESSING ACTIVATING EFFECT ON PPARα AND/OR PPARγ
MXPA05012029A (en) Compounds for treatment of inflammation, diabetes and related disorders
JP2004531580A (en) Peptide deformylase inhibitor

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 10430677

Country of ref document: US

AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SC SD SE SG SI SK SL TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR IE IT LU MC NL PT SE SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2468302

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2002804467

Country of ref document: EP

Ref document number: 1162/CHENP/2004

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: PA/a/2004/005168

Country of ref document: MX

Ref document number: 1020047008107

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 2002357032

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 533645

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 20028271009

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2002804467

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: JP

WWW Wipo information: withdrawn in national office

Ref document number: JP

WWP Wipo information: published in national office

Ref document number: 10430677

Country of ref document: US