AU2002239371B2 - Use of(-) (3-halomethylphenoxy) (4-halophenyl) acetic acid derivatives for treatment of insulin resistance, type 2 diabetes, hyperlipidemia and hyperuricemia - Google Patents

Use of(-) (3-halomethylphenoxy) (4-halophenyl) acetic acid derivatives for treatment of insulin resistance, type 2 diabetes, hyperlipidemia and hyperuricemia Download PDF

Info

Publication number
AU2002239371B2
AU2002239371B2 AU2002239371A AU2002239371A AU2002239371B2 AU 2002239371 B2 AU2002239371 B2 AU 2002239371B2 AU 2002239371 A AU2002239371 A AU 2002239371A AU 2002239371 A AU2002239371 A AU 2002239371A AU 2002239371 B2 AU2002239371 B2 AU 2002239371B2
Authority
AU
Australia
Prior art keywords
alkyl
alkoxy
conr
compound
group
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
AU2002239371A
Other versions
AU2002239371A1 (en
Inventor
Jian Luo
Kenneth L. Lusky
Zuchun Zhao
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
CymaBay Therapeutics Inc
Original Assignee
Metabolex 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 claimed from AU53162/00A external-priority patent/AU775909C/en
Priority claimed from US09/724,788 external-priority patent/US6624194B1/en
Application filed by Metabolex Inc filed Critical Metabolex Inc
Priority to AU2002239371A priority Critical patent/AU2002239371B2/en
Publication of AU2002239371A1 publication Critical patent/AU2002239371A1/en
Application granted granted Critical
Publication of AU2002239371B2 publication Critical patent/AU2002239371B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Description

USE OF (3-HALOMETHYLPHENOXY) (4-HALOPHENYL) ACETIC ACID DERIVATIVES FOR TREATMENT OF INSULIN RESISTANCE, TYPE 2 DIABETES, HYPERLIPIDEMIA AND
HYPERURICEMIA
FIELD OF THE INVENTION The present invention relates to the use of (3-halomethylphenoxy) (4halophenyl) acetic acid derivatives and compositions in the treatment of insulin resistance, Type 2 diabetes, hyperlipidemia and hyperuricemia. It further relates to (3halomethylphenoxy) (4-halophenyl) acetic acid derivatives that are useful for the treatment of insulin resistance, Type 2 diabetes, hyperlipidemia and hyperuricemia.
BACKGROUND OF THE INVENTION Diabetes mellitus, commonly called diabetes, refers to a disease process derived from multiple causative factors and characterized by elevated levels of plasma glucose, referred to as hyperglycemia. See, LeRoith, D. et al., DIABETES MELLITUS (Lippincott-Raven Publishers, Philadelphia, PA U.S.A. 1996), and all references cited therein. According to the American Diabetes Association, diabetes millituis estimated to affect approximately 6% of the world population. Uncontrolled hyperglycemia is associated with increased and premature mortality due to an increased risk for microvascular and macrovascular diseases, including nephropathy, neuropathy, retinopathy, hypertension, cerebrovascular disease and coronary heart disease. Therefore, control of glucose homeostasis is a critically important approach for the treatment of diabetes.
There are two major forms of diabetes: Type 1 diabetes (formerly referred to as insulin-dependent diabetes or IDDM); and Type 2 diabetes (formerly referred to as non-insulin dependent diabetes or NIDDM).
Type 1 diabetes is the result of an absolute deficiency of insulin, the hormone which regulates glucose utilization. This insulin deficiency is usually characterized by p-cell destruction within the Islets of Langerhans in the pancreas, which WO 02/044113 PCT/US01/44603 usually leads to absolute insulin deficiency. Type 1 diabetes has two forms: Immune- Mediated Diabetes Mellitus, which results from a cellular mediated autoimmune destruction of the 3 cells of the pancreas; and Idiopathic Diabetes Mellitus, which refers to forms of the disease that have no known etiologies.
Type 2 diabetes is a disease characterized by insulin resistance accompanied by relative, rather than absolute, insulin deficiency. Type 2 diabetes can range from predominant insulin resistance with relative insulin deficiency to predominant insulin deficiency with some insulin resistance. Insulin resistance is the diminished ability of insulin to exert its biological action across a broad range of concentrations. In insulin resistant individuals, the body secretes abnormally high amounts of insulin to compensate for this defect. When inadequate amounts of insulin are present to compensate for insulin resistance and adequately control glucose, a state of impaired glucose tolerance develops. In a significant number of individuals, insulin secretion declines further and the plasma glucose level rises, resulting in the clinical state of diabetes. Type 2 diabetes can be due to a profournd resistance to insulin stimulating regulatory effects on glucose and lipid metabolism in the main insulin-sensitive tissues: muscle, liver and adipose tissue. This resistance to insulin responsiveness results in insufficient insulin activation of glucose uptake, oxidation and storage in muscle and inadequate insulin repression of lipolysis in adipose tissue and of glucose production and secretion in liver. In Type 2 diabetes, free fatty acid levels are often elevated in obese and some non-obese patients and lipid oxidation is increased.
Premature development of atherosclerosis and increased rate of cardiovascular and peripheral vascular diseases arc characteristic features of patients with diabetes. Hyperlipidemia is an important precipitating factor for these diseases.
Hyperlipidemia is a condition generally characterized by an abnormal increase in serum lipids in the bloodstream and is an important risk factor in developing atherosclerosis and heart disease. For a review of disorders of lipid metabolism, see, Wilson, J. et al., Disorders of Lipid Metabolism, Chapter 23,Textbook of Endocrinology, 9 t i Edition, Sanders Company, Philadelphia, PA U.S.A. 1998; this reference and all references cited therein are herein incorated by reference). Serum lipoproteins are the carriers for lipids in the circulation. They are classified according to their density: chylomicrons; very low-density lipoproteins (VLDL); intermediate density lipoproteins (IDL); low density lipoproteins (LDL); and high density lipoproteins (HDL).
Hyperlipidemia is usually classified as primary or secondary hyperlipidemia. Primary WO 02/044113 PCT/US01/44603 hyperlipidemia is generally caused by genetic defects, while secondary hyperlipidemia is generally caused by other factors, such as various disease states, drugs, and dietary factors. Alternatively, hyperlipidemia can result from both a combination of primary and secondary causes of hyperlipidemia. Elevated cholesterol levels are associated with a number of disease states, including coronary artery disease, angina pectoris, carotid artery disease, strokes, cerebral arteriosclerosis, and xanthoma.
Dyslipidemia, or abnormal levels of lipoproteins in blood plasma, is a frequent occurrence among diabetics, and has been shown to be one of the main contributors to the increased incidence of coronary events and deaths among diabetic subjects (see, Joslin, E. Ann. Chim. Med. (1927) 5: 1061-1079). Epidemiological studies since then have confirmed the association and have shown a several-fold increase in coronary deaths among diabetic subjects when compared with nondiabetic subjects (see, Garcia, M. J. et al., Diabetes (1974) 23: 105-11 (1974); and Laakso, M. and Lehto, Diabetes Reviews (1997) 294-315). Several lipoprotein abnormalities have been described among diabetic subjects (Howard et al., Artherosclerosis (1978) 153-162).
Previous studies from the 1970's have demonstrated the effectiveness of racemic 2-acetamidoethyl (4-chlorophenyl) (3-trifluoromethylphenoxy) acetate (also known as "halofenate") as a potential therapeutic agent to treat Type 2 diabetes, hyperlipidemia and hyperuricemia (see, Bollofer, U.S. 3,517,050; Jain, A. et al., N. Eng. J. Med. (1975) 293: 1283-1286; Kudzma, D. et al., Diabetes (1977) 25: 291-95; Kohl, E. et al., Diabetes Care (1984) 7: 19-24; McMahon, F.G. et al., Univ. Mich. Med.
Center J. (1970) 36: 247-248; Simori, C. et al., Lipids (1972) 7: 96-99; Morgan, J.P. et al., Clin. Pharmacol. Therap. (1971) 12: 517-524, Aronow, W.S. et al., Clin. Pharmacol Ther (1973) 14: 358-365 and Fanelli, G.M. et al., J. Pharm. Experimental Therapeutics (1972) 180:377-396). In these previous studies, the effect of racemic halofenate on diabetes was observed when combined with sulfonylureas. A minimal effect on glucose was observed in patients with diabetes treated with racemic halofenate alone. However, significant side effects were noted including gastrointestinal bleeding from stomach and peptic ulcers (see, Friedberg, S.J. et al., Clin. Res. (1986) Vol. 34, No. 2: 682A).
In addition, there were some indications of drug-drug interactions of racemic halofenate with agents such as warfarin sulfate (also referred to as 3-(alphaacetonylbenzyl)-4-hydroxycoumarin or Coumadin T M (Dupont Pharmaceuticals, E. I.
Dupont de Nemours and Co., Inc., Wilmington, DE (see, Vesell, E. S. and WO 02/044113 PCT/US01/44603 Passantanti, Fed. Proc. (1972) 31(2): 538). Coumadin T M is an anticoagulant that acts by inhibiting the synthesis of vitamin K dependent clotting factors (which include Factors II, VII, IX, and X, and the anticoagulant proteins C and Coumadin TM is believed to be stereospecifically metabolized by hepatic microsomal enzymes (the cytochrome P450 enzymes). The cytochrome P450 isozymes involved in the metabolism of Coumadin include 2C9, 2C19, 2C8, 2C18, 1A2, and 3A4. 2C9 is likely to be the principal form of human liver P450 which modulates in vivo drug metabolism of several drugs including the anticoagulant activity of CoumadinTM (see, Miners, J. O. et al., Bri. J. Clin. Pharmacol. (1998) 45: 525-538).
Drugs that inhibit the metabolism of CoumadinTM result in a further decrease in vitamin K dependent clotting factors that prevents coagulation more than desired in patients receiving such therapy patients at risk for pulmonary or cerebral embolism from blood clots in their lower extremities, heart or other sites). Simple reduction of the dose of anticoagulant is often difficult as one needs to maintain adequate anticoagulation to prevent blood clots from forming. The increased anticoagulation from drug-drug interaction results in a significant risk to such patients with the possibility of severe bleeding from soft tissue injuries, gastrointestinal sites gastric or duodenal ulcers) or other lesions aortic aneurysm). Bleeding in the face of too much anticoagulation constitutes a medical emergency and can result in death if it is not treated immediately with appropriate therapy.
Cytochrome P450 2C9 is also known to be involved in the metabolism of several other commonly used drugs, including dilantin, sulfonylureas, such as tolbutamide and several nonstcroidal anti-inflammatory agents, such as ibuprofen.
Inhibition of this enzyme has the potential to cause other adverse effects related to drugdrug interactions, in addition to those described above for Coumadin T M (see, e.g., Pelkonen, O. et al., Xenobiotica (1998) 28: 1203-1253; Linn, J.H. and Lu, Clin.
Pharmacokinet. (1998) 35(5): 361-390).
Solutions to the above difficulties and deficiencies are needed before halofenate becomes effective for routine treatment of insulin resistance, Type 2 diabetes, hyperlipidemia and hyperuricemia. The present invention fulfills this and other needs by providing compounds, compositions and methods for alleviating insulin resistance, Type 2 diabetes, hyperlipidemia and hyperuricemia, while presenting a better adverse effect profile.
SUMMARY OF THE INVENTION This present invention provides a method of treating Type 2 diabetes in a mammal. The method comprises administering-to the manmmal a therapeutically effective amount of the stereoisomer of a compound of Formula I, X 0
R
X(2 x 4
X
3
(I)
wherein R is a member selected from the group consisting of a alkoxy OCH 2
CH
3 heteroalkoxy OCH 2 CH20CH2CH 3 aryloxy OCsH 5 heteroaryloxy OCsH 4 lower aralkoxy, di-lower alkylamino-lower alkoxy, benzamido-lower alkoxy, ureido-lower alkoxy, N'-lower alkyl-ureido-lower alkoxy, carbamoyl-lower alkoxy, halophenoxy substituted lower alkoxy, carbamoyl substituted phenoxy, carbonyl-lower alkylamino, N,N-di-lower alkylamino-lower alkylamino, halo substituted lower alkylamino, hydroxy substituted lower alkylamino, lower alkanoyloxy substituted lower alkylamino, ureido, arylsulfonamido NHTs), alkylsulfonamido NHMs) and lower alkoxycarbonylamino; X' is a halogen; and X' 2
X
3 and X 4 are each independently selected from hydrogen and halogen with the proviso that no more than two of X 2
X
3 and X4are hydrogen; or a pharmaceutically acceptable salt thereof, wherein the compound is substantially free of its stereoisomer.
Some such methods further comprise a compound of Formula II: R 2 0 1 0 X2 X1 x 3
(II)
wherein R 2 is a member selected from the group consisting of alyl, heteroalkyl, aryl, heteroaryl, phenyl-lower alkyl, benzamido-lower alkyl, di-lower alkylamino-lower alkyl, WO 02/044113 WO 02/44113PCT[USOI/44603 ureido-lower alkyl, N'-lower alkyl-ureido-lower alkyl, carbarnoyl-lower alkyl, halophenoxy substituted lower alkyl and carbamnoyl substituted phenyl.
Examples of such R 2 groups include, without limitation, the following:
C
1
-C
5 alkyl, C 1 -C8-cyclic alkyl, C 2
-C
5 alkenyl, and C 2
-C
5 alkynyl, wherein the groups are optionally substituted with one or more halogen atoms;, phenyl, naphthyl and pyridyl, wherein the groups are optionally substituted with one or more substituents selected from the group consisting of halo, C 1
-C
4 alkyl, C 1
-C
4 alkoxy, -NO 2 -S(O)m(Ci-Csalkyl), -OH, -NR 3
R
4 -C0 2
-CONR
3
R
4
-NR
3
COR
4 -NR'CONRWR and -CF,;
-(CHR
3
)R
4
-R
7 0R 3
-R
7
O
2
CR
8
NR
3 R 4
-R
7
COR
6
-R
7
NR
3 COR'; -R>{R 6
*R
-(CH
2 )oCH(R 3
)(CH
2 )qO 2
CR
9
-(CH
2
).CH(R
3
)(CH
2 )qNR 4
COR
9
-(CH
2 )oCH(R 3
)(CH
2 )qNR 4
CONR
3
R
4
-(CH
2 0
,CH(R
2
)(CH
2 )qN7R 4
COOR'
0
-(CH
2 )oCH(R)(CH 2 )qNR 4
SO
2
R
1
-(CHR'),CO
2
R
12
-(CHR
3
)PNR
3
R
4
-(CHR
3
),CONR
13
R'
0 OA-0 CH 2 )t CHA) and R3 0 R1 i R 15 R16 ,N o ,I-0 0 Subscripts m, o, q, s, t, u, v and w are integers as follows: mn is 0 to 2; o and q are 0 to pis ito 5;sis I to 3;tis 1 to5;uis~to 1;vis ito 3;andwis I to(2v+ R 3 and R 4 are independently H, C I-C 5 alkyl, phenyl or benzyl. R 5 is H, C I-C 5 alkyl or NR 3
R
4 R 6 is phenyl, naphthyl, pyridyl, imidazolyl, indoxyl, indolizinyl, oxazolyl, thiazolyl, thienyl, pyrimidyl, or I -pyrazolyl optionally substituted with one or more sub stituents selected from the group consisting of halo, CI-C 4 alkyl, CI-C 4 alkoxy, -NO 2 -S(O)m(CI-Csalkyl), -OH, -NRWR, -C0 2 -CONRWR, -NR'COR 4
-NR
3
CONR
3
R
4 and R' is a
C
1 -Cs saturated or unsaturated, straight-chain, branched or cyclic alkylene or alkylidene group optionally substituted with one or more groups selected from halo, hydroxyl, thiol, amino, monoalkyl amino, dialkyl amino, acylamino, carboxyl, alkylcarboxyl, acyl, aryl, aroyl, aralkyl, cyano, nitro, alkoxy, alkenyloxy, alkylcarbonyloxy and arylcarbonyloxy.
R8 is a C 1
-C
8 straight-chain or branched alkylene or alkylidene optionally substituted with one or more groups selected from amino, monoalkyl amino, dialkyl amino, acylamino, hydroxyl, thiol, methylthiol, carboxyl and phenyl. R9 and R 10 are independently H, CI-C alkyl, optionally substituted with one or more groups consisting of C -C 5 alkoxy aryl and heteroaryl, wherein the aryl is phenyl or naphthyl optionally substituted with one or more WO 02/044113 WO 02/44113PCTIIJS01I44603 substituents selected from the group consisting of halo, CI-C 4 alkyl, CI-C 4 alkoxy, -NO 2 -S(O)m(Ci-C 5 alkyl), -OH, -NR 3
R
4 -C0 2
-CONR
3
R
4 -NR'COR 4
-NR'CONR
3 R 4 and and wherein the heteroaryl is pyridyl optionally substituted with one or more substituents selected from the group consisting of halo, CI-C 4 alkyl, CI-C 4 alkoxy, -NO 2
S(O)M(CI-C
5 alkyl), -OH, -NR 3
R
4 -C0 2 CORR, -NR 3
COR
4
-KR'CONR
and R 1 1 is methyl or phenyl, wherein the phenyl is optionally substituted with methyl and/or -NO 2 R 1 2 is H, CI-Cs alkyl, phenyl, benzyl, naphthyl or pyridyl, wherein the C,-C 5 alkyl, phenyl, naphthyl, benzyl and pyridyl are optionally substituted with one or more substituents selected from the group consisting of halo, C 1
-C
4 alkyl, C 1
-C
4 alkoxy, -NO 2 5 alkyl), -OH, -NR 3
R
4 -C0Rs, -CONR 3 R 4
-NR'COR
4
-NR'CONR
3
R
4 and R1 3 and R 14 are independently the following: alkyl, alkenyl, aryl, aralkyl or cycloalkyl, wherein the groups are optionally substituted with one or more substituents selected from the group consisting of halo, CI-C 4 alkyl, C 1
-C
4 alkoxy, -NO 2
(CI-C
5 alkyl), -OH, -NR'R 4 -C0 2
R
5
-CONR
3
R
4
-NR'COR
4
-NR
3
CONR
3
R
4
-CHNR
3
R
4 00CR' 8 and and wherein R 1 3 and R 1 4 are included as
-(CHR,)CCONR
3 NR3 'R1 4 i -N \0 -N -OR 3 -N NR 3 -N /\NR 3 OOR3
\OH
R
3 000 R R'NOC -N or -0Nt COOR 3
R
1 5 is CF, or C ,-C 5 alkyl, wherein CI-C 5 alkyl. is optionally substituted with the following substituents: C 1
-C
5 alkoxy; phenyl, optionally substituted with one or more substituents selected from the group consisting of halo, C,-C 4 alkyl, CI-C 4 alkoxy, -NO 2
-C
5 alkyl), -OH, -NR 3
R
4 -C0 2 R 5
-CONR
3
R
4
-NR
3
COR
4
-NR
3
CONR
3
R
4 and benzyl, optionally substituted with one or more substituents selected from the group consisting of halo, CI-C 4 alkyl, CI-C 4 alkoxy, -N0 2 -S(O)m,(CI-Csalkyl), -OH,
-NR
3
R
4 -C0 2
R
5
-CONR
3
R
4
-NR'COR
4
-NR'CONR
3
R
4 and R" 6 is H, 0,-C alkyl or benzyl. R 17 is CI-C 5 alkyl, C 3
-C
8 cyclic alkyl, phenyl or benzyl. R1 is H, alkyl, aryl, aralkyl or cycloalkyl, where the group is optionally substituted with one or more substituents selected from the group consisting of halo, CI-C 4 alkyl, Ci-C 4 alkoxy, -NO 2 -S(O)m(Ci-Csalkyl), -OH, -NR 3
R
4 -C0 2
R
5
-CONR
3
R
4
-NR
3
COR
4
-NR
3
CONR
3
R
4 and -CvF,. The groups X 2
X
3 and X 4 have the meanings provided with reference to s Formula I.
In other aspects, the general methods will use a compound of Formula III: CH3
HN
0 0 CO O
CF
3
(III)
The preferred compound of Formula III is known as 2-acetamidoethyl 4chlorophenyl-(3-trifluoromethylphenoxy)-acetate" or halofenate".
The present invention further provides a method for treating insulin resistance in a mammal. This method comprises administering to the mammal a therapeutically effective amount of the stereoisomer of a compound of Formula I as defined herein, or a pharmaceutically acceptable salt thereof, wherein the compound is substantially free of is its stereoisomer. In certain preferred embodiments, the methods use a compound of Formula II.
The present invention further provides a method of treating hyperuricemia in a mammal. This method comprises administering to the mammal a therapeutically effective amount of a compound of Formula I as defined herein, or a pharmaceutically acceptable salt thereof, wherein the compound is substantially free of its stereoisomer.
In certain preferred embodiment, the methods use a compound of Formula II.
In another aspect the present invention also provides a method for treating insulin resistance in a mammal, comprising administering to said mammal a therapeutically effective amount of the stereoisomer of a compound of Formula I, 0
R
X
2
X
4
X
3
(I)
wherein: [R:\L3H]637720spcci.doc:aak R is hydroxy or comprises a hydroxy group in an ester linkage which can be hydrolyzed in vivo to provide a pharmacologically effective amount of a compound of Formula I wherein R is hydroxy; X' is a halogen; and
X
2
X
3 and X 4 are each independently selected from hydrogen and halogen with the proviso that no more than two of X 2
X
3 and X 4 are hydrogen; or a pharmaceutically acceptable salt thereof, wherein the compound is substantially free of its stereoisomer.
In another aspect the present invention provides a method for treating type 2 diabetes in a mammal, comprising administering to said mammal a therapeutically effective amount of the stereoisomer of a compound of Formula I, O
R
X
2
X
4
X
3
(I)
wherein: R is hydroxy or comprises a hydroxy group in an ester linkage which can be hydrolyzed in vivo to provide a pharmacologically effective amount of a compound of formula I wherein R is hydroxy; X' is a halogen; and
X
2
X
3 and X 4 are each independently selected from hydrogen and halogen with the proviso that no more than two of X 2
X
3 and X 4 are hydrogen; or a pharmaceutically acceptable salt thereof, and wherein the compound is substantially free of its stereoisomer.
In another aspect the present invention provides a method for treating hyperuricemia in a mammal, comprising administering to said mammal a therapeutically effective amount of the stereoisomer of a compound of Formula I,
X
1
R
X2
X
4
X
3
(I)
wherein: [R:\LIBH]637720spci.doc:aak R is hydroxy or comprises a hydroxy group in an ester linkage which can be hydrolyzed in vivo to provide a pharmacologically effective amount of a compound of formula I wherein R is hydroxy; X' is a halogen; and CN 5 X 2
X
3 and X 4 are each independently selected from hydrogen and halogen with the proviso that no more than two of X 2
X
3 and X 4 are hydrogen; or a pharmaceutically acceptable salt thereof, and 0 wherein the compound is substantially free of its stereoisomer.
SThe present invention further provides the use of the stereoisomer of a compound of Formula I as defined herein, or a pharmaceutically acceptable salt thereof, wherein the compound is substantially free of its stereoisomer, in the manufacture of a medicament for treating Type 2 diabetes.
The present invention also provides the use of the stereoisomer of a compound of Formula I as defined herein, or a pharmaceutically acceptable salt thereof, wherein the compound is substantially free of its stereoisomer, in the manufacture of a medicament for treating insulin resistance.
The present invention also provides the use of the stereoisomer of a compound of Formula I as defined herein, or a pharmaceutically acceptable salt thereof, wherein the compound is substantially free of its stereoisomer, in the manufacture of a medicament for treating hyperuricemia.
The present invention also relates to compounds and pharmaceutical compositions.
The compounds are of Formula I, Formula II or Formula III. The pharmaceutical compositions comprise a pharmaceutically acceptable carrier and a therapeutically effective amount of a compound of Formula I, Formula II or Formula III.
Thus, the present invention also provides a pharmaceutical composition when used for treating insulin resistance, type 2 diabetes, or hyperuricemia, said composition comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of the stereoisomer of a compound of Formula I as defined herein, or a pharmaceutically acceptable salt thereof, wherein the compound is substantially free of its stereoisomer.
In a further aspect the present invention provides a pharmaceutical composition when used to treat insulin resistance in a mammal, said composition comprising a therapeutically effective amount of the stereoisomer of a compound of Formula I, [R:\LIBHI637720specidocaak >(X2 x 4
X
3 wherein: R is hydroxy or comprises a hydroxy group in an ester linkage which can be hydrolyzed in vivo to provide a pharmacologically effective amount of a compound of formula I wherein R is hydroxy; X' is a halogen; and
X
2
X
3 and X 4 are each independently selected from hydrogen and halogen with the 0o proviso that no more than two of X 2
X
3 and X 4 are hydrogen; or a pharmaceutically acceptable salt thereof, and wherein the compound is substantially free of its stereoisomer.
In a further aspect the present invention provides a pharmaceutical composition when used to treat type 2 diabetes in a mammal, said composition comprising a therapeutically effective amount of the stereoisomer of a compound of Formula I,
SR
X
4
X
3
(I)
wherein: R is hydroxy or comprises a hydroxy group in an ester linkage which can be hydrolyzed in vivo to provide a pharmacologically effective amount of a compound of formula I wherein R is hydroxy; X' is a halogen; and
X
2
X
3 and X 4 are each independently selected from hydrogen and halogen with the proviso that no more than two of X 2
X
3 and X 4 are hydrogen; or a pharmaceutically acceptable salt thereof, and wherein the compound is substantially free of its stereoisomer.
In a further aspect the present invention provides a pharmaceutical composition when used to treat hyperuricemia in a mammal, said composition comprising a therapeutically effective amount of the stereoisomer of a compound of Formula I, [R:\LIBH637720speci.doc:aak
X
2
X
4
X
3
(I)
wherein: R is hydroxy or comprises a hydroxy group in an ester linkage which can be hydrolyzed in vivo to provide a pharmacologically effective amount of a compound of formula I wherein R is hydroxy; X' is a halogen; and
X
2
X
3 and X 4 are each independently selected from hydrogen and halogen with the to proviso that no more than two of X 2
X
3 and X 4 are hydrogen; or a pharmaceutically acceptable salt thereof, and wherein the compound is substantially free of its stereoisomer.
[R:\LIBH]637720specidoc:aak WO 02/044113 PCT/US01/44603 BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 shows the inhibition of cytochrome P450 2C9 (CYP2C9) activity by racemic halofenic acid, halofenic acid and halofenic acid. The hydroxylation of tolbutamide was measured in the presence of increasing concentrations of these compounds. Racemic halofenic acid inhibited CYP 2C9 activity with an IC50 of 0.45 tM and halofenic acid inhibited CYP 2C9 with an IC50 of 0.22 ItM. In contrast, the halofenic acid was 20-fold less potent with an apparent IC50 of 3.5 pM.
Figure 2 shows the time course of glucose-lowering following a single oral dose of racemic halofenate, enantiomer of halofenate or enantiomer of halofenate at 250 mg/kg in diabetic ob/ob mice. The enantiomer showed the most rapid onset of action and the longest duration of action. The decrease in glucose was significant (p<0.05) for the enantiomer compared to control for all points from 3 to 24 hours. Racemic halofenate and the enantiomer were also significant (p<0.05) for all points from 4.5 to 24 hours. The plasma glucose at 24 hours was 217 16.4 mg/dl in animals treated with the enantiomer, compared to 306 28.5 mg/dl and 259.3 20.8 mg/dl for animals treated with the enantiomer and the racemate, respectively. The plasma glucose in the vehicle treated controls was 408 16.2 mg/dl at 24 hours. The enantiomer was more effective and significantly different (p<0.05) from the enantiomer at both the 3 hour and 24 hour time points.
Figure 3 shows the ability of racemic halofenate and both the and enantiomers ofhalofenate to lower plasma glucose in diabetic ob/ob mice following daily oral administration. The racemate was given at a dose of 250 mg/kg/day and the enantiomers were given at doses of 125 mg/kg/day and 250 mg/kg/day. Significant decreases in glucose levels relative to control animals were observed in animals treated with racemic halofenate and both the and enantiomers. At the low dose (125 mg/kg) of treatment with the and enantiomers, the enantiomer was significant at 6, 27 and 30 hours whereas the enantiomer was significant at only 6 and 27 hours.
Figure 4 shows the plasma insulin levels in the ob/ob mice treated with racemic halofenate and both the and enantiomers ofhalofenate in diabetic ob/ob mice following daily oral administration. The racemate was given at a dose of 250 mg/kg/day and the enantiomers were given at doses of 125 mg/kg/day and 250 mg/kg/day. Relative to the vehicle control, insulins were lower in the animals treated with either the racemate or either of the enantiomers of halofenate. At the high dose, the WO 02/044113 PCT/US01/44603 greatest extent of reduced plasma insulin was noted at 27 and 30 hours in animals treated with both the and enantiomers of halofenate following two days of treatment.
Figure 5 shows plasma glucose levels following an overnight fast in ob/ob mice after 5 days treatment with vehicle, racemic halofenate at 250 mg/kg/day, enantiomer of halofenate at 125 mg/kg/day and 250 mg/kg/day or enantiomer of halofenate at 125 mg/kg/day or 250 mg/kg/day. The control animals were hyperglycemic with plasma glucose levels of 185.4 12.3 mg/dl. All of the animals treated with halofenate showed significant (p 0.01) reductions in glucose. The high doses of both enantiomers lowered the glucose to near normal levels at 127.3 8.0 mg/dl and 127.2 9.7 mg/dl for the enantiomer and enantiomer treated animals, respectively.
Figure 6 shows the overnight fasting plasma insulin levels in the ob/ob mice treated with vehicle, racemic halofenate at 250 mg/kg/day, enantiomer at 125 mg/kg/day and 250 mg/kg/day or enantiomer of halofenate at 125 mg/kg/day or 250 mg/kg/day for 5 days. Significantly lower plasma insulins were observed in animals receiving both doses of enantiomer. The low dose of enantiomer of halofenate did not lower plasma insulin, although the high dose of the enantiomer resulted in a decrease in plasma insulin.
Figure 7A shows plasma glucose levels following an oral glucose challenge in Zucker fatty rats, a model of insulin resistance and Impaired Glucose Tolerance. These animals were treated with either a vehicle control, racemic halofenate, halofenate or halofenate 5.5 hours prior to the glucose challenge. The racemate was given at 100 mg/kg and both of the enantiomers were given at 50 and 100 mg/kg. In the control animals the glucose rose to >250 mg/dl 30 minutes after the challenge, a clear indication of impaired glucose tolerance. The plasma glucose was reduced in rats that had received racemic halofenate, especially between 30 60 minutes after the challenge.
Animals that received'the halofenate at 100 mg/kg had the greatest degree of glucoselowering of all the treated animals. Animals treated with the halofenate had lower glucose levels that persisted at 90-120 minutes, compared to those rats treated with the racemate or halofenate. Figure 7B compares the incremental area under the curve (AUC) for the animals in each group. Significant changes (p<0.05) were noted in the groups treated with both doses of the halofenate. Although the AUC was lower in the other groups relative to the control, the changes were not significant.
WO 02/044113 PCT/US01/44603 Figure 8 shows the results of a short insulin tolerance test in Zucker fatty rats that were treated with either a vehicle control, halofenate (50 mg/kg/day) or halofenate (50 mg/kg/day) for 5 days. This test is a measure of the insulin sensitivity of the test animals, the slope of the decline in glucose representing a direct measure of insulin responsiveness. The halofenate-treated animals were significantly more insulin sensitive than the vehicle-treated (p 0.01) or the halofenate-treated (p 0.05) animals.
Figure 9A shows plasma cholesterol levels in Zucker Diabetic Fatty rats treated for 13 days with racemic halofenate, enantiomer or enantiomer at mg/kg/day, 25 mg/kg/day or 25 mg/kg/day, respectively, relative to a vehicle treated control group. In both the enantiomer and racemate treated animals, the plasma cholesterol declined with treatment. The cholesterol in the enantiomer treated animals remained relatively constant, whereas cholesterol rose in the control animals. Figure 9B compares the differences in plasma cholesterol between the control group and the treated groups. The enantiomer was the most active of the species tested.
Figure 10A shows plasma cholesterol levels in Zucker Diabetic Fatty rats treated for 14 days with either enantiomer or enantiomer of halofenate at either 12.5 mg/kg/day (Low dose) or 37.5 mg/kg/day (High dose) relative to a vehicle treated control group. In the animals treated with the high dose, the enantiomer resulted in the greatest extent of cholesterol lowering. Figure 10B compares the differences in plasma cholesterol between the control and treated groups. There were significant differences in the animals treated with the enantiomer after 7 days at the low dose and after both 7 and 14 days at the high dose. The enantiomer showed significance only after 7 days of treatment at the high dose.
Figure 11A shows plasma triglyceride levels in Zucker Diabetic Fatty rats treated with either enantiomer or enantiomer at either 12.5 mg/kg/day (Low dose) or 37.5 mg/kg/day (High dose) relative to a vehicle treated control group. Animals treated with the high dose of the enantiomer had the lowest triglyceride levels of all the treatment groups. Figure 11B compares the differences in plasma triglyceride between the control and treated groups. At 7 days, the high dose of both the and enantiomers showed significant lowering of plasma triglyceride.
Figure 12 shows plasma glucose levels in Zucker Diabetic Fatty rats treated with vehicle, halofenate or halofenate at day 0, day 2 and day 3. Treatment WO 02/044113 PCT/US01/44603 with halofenate significantly reduced plasma glucose concentrations as compared to vehicle-treated animals.
Figure 13 shows plasma glucose concentrations in a control group of C57BL/6J db/db mice versus in a group treated with halofenate. Plasma glucose levels in the control group increased progressively as animals aged, while the increase of plasma glucose levels in the halofenate treated group was prevented or significantly delayed.
Figure 14 shows plasma insulin levels in a control group of C57BL/6J db/db mice versus in a group treated with halofenate. Treatment with halofenate maintained the plasma insulin concentration, while plasma insulin in the control group decreased progressively.
Figure 15 shows the percentage of non-diabetic mice in a control group of C57BL/6J db/db mice versus in a group treated with halofenate. About 30% of mice in the halofenate treated group did not develop diabetes (plasma glucose levels <250 mg/dl), while all of the control group did by the age of 10 weeks.
Figure 16 shows plasma triglyceride levels in a control group of C57BL/6J db/db mice versus in a group treated with halofenate. Treatment with halofenate alleviated hyperlipidemia, while there was no alleviation in the control group.
Figure 17 shows the effect halofenate and halofenate on plasma uric acid levels in oxonic acid induced hyperuricemic rats. Oral administration of(-) halofenate significantly reduced plasma uric acid levels. Halofenate also lowered plasma uric acid levels, but it was not statistically significant.
Figure 18 is a chromatogram illustrating the reversed phase chiral separation of compound 19.6, to which 2% of the undesired enantiomer has been added.
Figure 19 is a chromatogram illustrating the normal phase separation of a partly racemized product, compound 19.25, to which 2% of the undesired enantiomer has been added.
Figures 20A-20G illustrate the hydrolysis in plasma of various prodrug esters of the present invention. In each graph, the loss of prodrug compound is seen with a concomitant rise in the plasma concentration of halofenic acid.
WO 02/044113 PCT/US01/44603
DEFINITIONS
The term "mammal" includes, without limitation, humans, domestic animals dogs or cats), farm animals (cows, horses, or pigs), monkeys, rabbits, mice, and laboratory animals.
The term "insulin resistance" can be defined generally as a disorder of glucose metabolism. More specifically, insulin resistance can be defined as the diminished ability of insulin to exert its biological action across a broad range of concentrations producing less than the expected biologic effect. (see, Reaven, G. M., .1 Basic Clin. Phys. Pharm. (1998) 9: 387-406 and Flier, J. Ann Rev. Med. (1983) 34: 145-60). Insulin resistant persons have a diminished ability to properly metabolize glucose and respond poorly, if at all, to insulin therapy. Manifestations of insulin resistance include insufficient insulin activation of glucose uptake, oxidation and storage in muscle and inadequate insulin repression of lipolysis in adipose tissue and of glucose production and secretion in liver. Insulin resistance can cause or contribute to polycystic ovarian syndrome, Impaired Glucose Tolerance (IGT), gestational diabetes, hypertension, obesity, atherosclerosis and a variety of other disorders. Eventually, the insulin resistant individuals can progress to a point where a diabetic state is reached. The association of insulin resistance with glucose intolerance, an increase in plasma triglyceride and a decrease in high-density lipoprotein cholesterol concentrations, high blood pressure, hyperuricemia, smaller denser low-density lipoprotein particles, and higher circulating levels ofplaminogen activator inhibitor-i), has been referred to as "Syndrome X" (see, Reaven, G. Physiol. Rev. (1995) 75: 473-486).
The term "diabetes mellitus" or "diabetes" means a disease or condition that is generally characterized by metabolic defects in production and utilization of glucose which result in the failure to maintain appropriate blood sugar levels in the body.
The result of these defects is elevated blood glucose, referred to as "hyperglycemia." Two major forms of diabetes are Type 1 diabetes and Type 2 diabetes. As described above, Type 1 diabetes is generally the result of an absolute deficiency of insulin, the hormone which regulates glucose utilization. Type 2 diabetes often occurs in the face of normal, or even elevated levels of insulin and can result from the inability of tissues to respond appropriately to insulin. Most Type 2 diabetic patients are insulin resistant and have a relative deficiency of insulin, in that insulin secretion can not compensate for the resistance of peripheral tissues to respond to insulin. In addition, many Type 2 diabetics WO 02/044113 PCT/US01/44603 are obese. Other types of disorders of glucose homeostasis include Impaired Glucose Tolerance, which is a metabolic stage intermediate between normal glucose homeostasis and diabetes, and Gestational Diabetes Mellitus, which is glucose intolerance in pregnancy in women with no previous history of Type 1 or Type 2 diabetes.
The term "secondary diabetes" is diabetes resulting from other identifiable etiologies which include: genetic defects of P cell function maturity onset-type diabetes of youth, referred to as "MODY," which is an early-onset form of Type 2 diabetes with autosomal inheritance; see, Fajans S. et al., Diabet. Med. (1996) (9 Suppl S90-5 and Bell, G. et al.,Annu. Rev. Physiol. (1996) 58: 171-86; genetic defects in insulin action; diseases of the exocrine pancreas hemochromatosis, pancreatitis, and cystic fibrosis); certain endocrine diseases in which excess hormones interfere with insulin action growth hormone in acromegaly and cortisol in Cushing's syndrome); certain drugs that suppress insulin secretion phenytoin) or inhibit insulin action estrogens and glucocorticoids); and diabetes caused by infection rubella, Coxsackie, and CMV); as well as other genetic syndromes.
The guidelines for diagnosis for Type 2 diabetes, impaired glucose tolerance, and gestational diabetes have been outlined by the American Diabetes Association (see, The Expert Committee on the Diagnosis and Classification of Diabetes Mellitus, Diabetes Care, (1999) Vol 2 (Suppl S5-19).
The term "halofenic acid" refers to the acid form of 4-Chlorophenyl-(3trifluoromethylphenoxy)-acetic acid.
The term "hyperinsulinemia" refers to the presence of an abnormally elevated level of insulin in the blood.
The term "hyperuricemia" refers to the presence of an abnormally elevated level of uric acid in the blood.
The term "secretagogue" means a substance or compound that stimulates secretion. For example, an insulin secretagogue is a substance or compound that stimulates secretion of insulin.
The term "hemoglobin" or "Hb" refers to a respiratory pigment present in erythrocytes, which is largely responsible for oxygen transport. A hemoglobin molecule comprises four polypeptide subunits (two a chain systems and two p chain systems, respectively). Each subunit is formed by association of one globin protein and one heme molecule which is an iron-protoporphyrin complex. The major class of hemoglobin found in normal adult hemolysate is adult hemoglobin (referred to as "HbA"; also WO 02/044113 PCT/US01/44603 referred to HbAo for distinguishing it from glycated hemoglobin, which is referred to as "HbAI," described infra) having a 2 3 2 subunits. Trace components such as HbA 2 (ct 2 2 can also be found in normal adult hemolysate.
Among classes of adult hemoglobin HbAs, there is a glycated hemoglobin (referred to as "HbAi," or "glycosylated hemoglobin"), which may be further fractionated into HbA,, HbAia2, HbAlb, and HbAIc with an ion exchange resin fractionation. All of these subclasses have the same primary structure, which is stabilized by formation of an aldimine (Schiff base) by the amino group of N-terminal valine in the p subunit chain of normal hemoglobin HbA and glucose (or, glucose-6-phosphate or fructose) followed by formation of ketoamine by Amadori rearrangement.
The term "glycosylated hemoglobin" (also referred to as "HbAlc,", "GHb", "hemoglobin glycosylated", "diabetic control index" and "glycohemoglobin"; hereinafter referred to as "hemoglobin Alc") refers to a stable product of the nonenzymatic glycosylation of the p-chain of hemoglobin by plasma glucose. Hemoglobin Aic comprises the main portion of glycated hemoglobins in the blood. The ratio of glycosylated hemoglobin is proportional to blood glucose level. Therefore, hemoglobin Ale rate of formation directly increases with increasing plasma glucose levels. Since glycosylation occurs at a constant rate during the 120-day lifespan of an erythrocyte, measurement of glycosylated hemoglobin levels reflect the average blood glucose level for an individual during the preceding two to three months. Therefore determination of the amount of glycosylated hemoglobin HbAc can be a good index for carbohydrate metabolism control. Accordingly, blood glucose levels of the last two months can be estimated on the basis of the ratio of HbAic to total hemoglobin Hb. The analysis of the hemoglobin Alc in blood is used as a measurement enabling long-term control of blood glucose level (see, Jain, et al., Diabetes (1989) 38: 1539-1543; Peters et al., JAMA (1996) 276: 1246-1252).
The term "symptom" of diabetes, includes, but is not limited to, polyuria, polydipsia, and polyphagia, as used herein, incorporating their common usage. For example, "polyuria" means the passage of a large volume of urine during a given period; "polydipsia" means chronic, excessive thirst; and "polyphagia" means excessive eating.
Other symptoms of diabetes include, increased susceptibility to certain infections (especially fungal and staphylococcal infections), nausea, and ketoacidosis (enhanced production of ketone bodies in the blood).
WO 02/044113 PCT/US01/44603 The term "complication" of diabetes includes, but is not limited to, microvascular complications and macrovascular complications. Microvascular complications are those complications which generally result in small blood vessel damage. These complications include, retinopathy (the impairment or loss of vision due to blood vessel damage in the eyes); neuropathy (nerve damage and foot problems due to blood vessel damage to the nervous system); and nephropathy (kidney disease due to blood vessel damage in the kidneys). Macrovascular complications are those complications which generally result from large blood vessel damage. These complications include, cardiovascular disease and peripheral vascular disease.
Cardiovascular disease refers to diseases of blood vessels of the heart. See. Kaplan, R. et al., "Cardiovascular diseases" in HEALTH AND HUMAN BEHAVIOR, pp. 206-242 (McGraw-Hill, New York 1993). Cardiovascular disease is generally one of several forms, including, hypertension (also referred to as high blood pressure), coronary heart disease, stroke, and rheumatic heart disease. Peripheral vascular disease refers to diseases of any of the blood vessels outside of the heart. It is often a narrowing of the blood vessels that carry blood to leg and arm muscles.
The term "atherosclerosis" encompasses vascular diseases and conditions that are recognized and understood by physicians practicing in the relevant fields of medicine. Atherosclerotic cardiovascular disease, coronary heart disease (also known as coronary artery disease or ischemic heart disease); cerebrovascular disease and peripheral vessel disease are all clinical manifestations of atherosclerosis and are therefore encompassed by the terms "atherosclerosis" and "atherosclerotic disease".
The term "antihyperlipidemic" refers to the lowering of excessive lipid concentrations in blood to desired levels.
The term "antiuricemic" refers to the lowering of excessive uric acid concentrations in blood to desired levels.
The term "hyperlipidemia" refers to the presence of an abnormally elevated level of lipids in the blood. Hyperlipidemia can appear in at least three forms: hypercholesterolemia, an elevated cholesterol level; hypertriglyceridemia, i.e., an elevated triglyceride level; and combined hyperlipidemia, a combination of hypercholesterolemia and hypertriglyceridemia.
The term "modulate" refers to the treating, prevention, suppression, enhancement or induction of a function or condition. For example, the compounds of the WO 02/044113 PCT/US01/44603 present invention can modulate hyperlipidemia by lowering cholesterol in a human, thereby suppressing hyperlipidemia.
The term "treating" means the management and care of a human subject for the purpose of combating the disease, condition, or disorder and includes the administration of a compound of the present invention to prevent the onset of the symptoms or complications, alleviating the symptoms or complications, or eliminating the disease, condition, or disorder.
The term "preventing" means the management and care of a human subject such that the onset of symptoms of a disease, condition or disorder does not occur.
The term "cholesterol" refers to a steroid alcohol that is an essential component of cell membranes and myelin sheaths and, as used herein, incorporates its common usage. Cholesterol also serves as a precursor for steroid hormones and bile acids.
The term "triglyceride(s)" as used herein, incorporates its common usage. TGs consist of three fatty acid molecules esterified to a glycerol molecule and serve to store fatty acids which are used by muscle cells for energy production or are taken up and stored in adipose tissue.
Because cholesterol and TGs are water insoluble, they must be packaged in special molecular complexes known as "lipoproteins" in order to be transported in the plasma. Lipoproteins can accumulate in the plasma due to overproduction and/or deficient removal. There are at least five distinct lipoproteins differing in size, composition, density, and function. In the cells of the small of the intestine, dietary lipids are packaged into large lipoprotein complexes called "chylomicrons", which have a high TG and low-cholesterol content. In the liver, TG and cholesterol esters are packaged and released into plasma as TG-rich lipoprotein called very low density lipoprotein whose primary function is the endogenous transport of TGs made in the liver or released by adipose tissue. Through enzymatic action, VLDL can be either reduced and taken up by the liver, or transformed into intermediate density lipoprotein IDL, is in turn, either taken up by the liver, or is further modified to form the low density lipoprotein LDL is either taken up and broken down by the liver, or is taken up by extrahepatic tissue. High density lipoprotein helps remove cholesterol from peripheral tissues in a process called reverse cholesterol transport.
WO 02/044113 PCT/US01/44603 The term "dyslipidemia" refers to abnormal levels oflipoproteins in blood plasma including both depressed and/or elevated levels of lipoproteins elevated levels ofLDL, VLDL and depressed levels of HDL).
Exemplary Primary Hyperlipidemia include, but are not limited to, the following: Familial Hyperchylomicronemia, a rare genetic disorder which causes a deficiency in an enzyme, LP lipase, that breaks down fat molecules. The LP lipase deficiency can cause the accumulation of large quantities of fat or lipoproteins in the blood; Familial Hypercholesterolemia, a relatively common genetic disorder caused where the underlying defect is a series of mutations in the LDL receptor gene that result in malfunctioning LDL receptors and/or absence of the LDL receptors. This brings about ineffective clearance of LDL by the LDL receptors resulting in elevated LDL and total cholesterol levels in the plasma; Familial Combined Hyperlipidemia, also known as multiple lipoprotein-type hyperlipidemia; an inherited disorder where patients and their affected first-degree relatives can at various times manifest high cholesterol and high triglycerides.
Levels ofHDL cholesterol are often moderately decreased; Familial Defective Apolipoprotein B-100 is a relatively common autosomal dominant genetic abnormality. The defect is caused by a single nucleotide mutation that produces a substitution of glutamine for arginine which can cause reduced affinity of LDL particles for the LDL receptor. Consequently, this can cause high plasma LDL and total cholesterol levels; Familial Dysbetaliproteinemia, also referred to as Type III Hyperlipoproteinemia, is an uncommon inherited disorder resulting in moderate to severe elevations of serum TG and cholesterol levels with abnormal apolipoprotein E function.
HDL levels are usually normal; and Familial Hypertriglyceridemia, is a common inherited disorder in which the concentration of plasma VLDL is elevated. This can cause mild to moderately elevated triglyceride levels (and usually not cholesterol levels) and can often be associated with low plasma HDL levels.
Risk factors in exemplary Secondary Hyperlipidemia include, but are not limited to, the following: disease risk factors, such as a history of Type 1 diabetes, Type 2 diabetes, Cushing's syndrome, hypothroidism and certain types of renal failure; WO 02/044113 PCT/US01/44603 drug risk factors, which include, birth control pills; hormones, such as estrogen, and corticosteroids; certain diuretics; and various 13 blockers; dietary risk factors include dietary fat intake per total calories greater than 40%; saturated fat intake per total calories greater than 10%; cholesterol intake greater than 300 mg per day; habitual and excessive alcohol use; and obesity.
The terms "obese" and "obesity" refers to, according to the World Health Organization, a Body Mass Index (BMI) greater than 27.8 kg/m 2 for men and 27.3 kg/m 2 for women (BMI equals weight (kg)/height Obesity is linked to a variety of medical conditions including diabetes and hyperlipidemia. Obesity is also a known risk factor for the development of Type 2 diabetes (See, Barrett-Conner, Epidemol. Rev. (1989) 11: 172-181; and Knowler, et al., Am. J. Clin. Nutr. (1991) 53:1543-1551).
"Pharmaceutically acceptable salts" refer to the non-toxic alkali metal, alkaline earth metal, and ammonium salts commonly used in the pharmaceutical industry including the sodium, potassium, lithium, calcium, magnesium, barium, ammonium, and protamine zinc salts, which are prepared by methods well known in the art. The term also includes non-toxic acid addition salts, which are generally prepared by reacting the compounds of the present invention with a suitable organic or inorganic acid.
Representative salts include, but are not limited to, the hydrochloride, hydrobromide, sulfate, bisulfate, acetate, oxalate, valerate, oleate, laurate, borate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, napsylate, and the like.
"Pharmaceutically acceptable acid addition salt" refers to those salts which retain the biological effectiveness and properties of the free bases and which are not biologically or otherwise undesirable, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, malic acid, malonic acid, succinic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, menthanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like. For a description of pharmaceutically acceptable acid addition salts as prodrugs. See, Bundgaard, ed., Design of Prodrugs (Elsevier Science Publishers, Amsterdam 1985).
"Pharmaceutically acceptable ester" refers to those esters which retain, upon hydrolysis of the ester bond, the biological effectiveness and properties of the carboxylic acid or alcohol and are not biologically or otherwise undesirable. For a description of pharmaceutically acceptable esters as prodrugs, see Bundgaard, supra.
WO 02/044113 PCT/US01/44603 These esters are typically formed from the corresponding carboxylic acid and an alcohol.
Generally, ester formation can be accomplished via conventional synthetic techniques.
(See, March Advanced Organic Chemistry, 3rd Ed., p. 1157 (John Wiley Sons, New York 1985) and references cited therein, and Mark et al., Encyclopedia of Chemical Technology, (1980) John Wiley Sons, New York). The alcohol component of the ester will generally comprise: a C 2
C-
12 aliphatic alcohol that can or can not contain one or more double bonds and can or can not contain branched carbons; or (ii) a C7C12 aromatic or heteroaromatic alcohols. The present invention also contemplates the use of those compositions which are both esters as described herein and at the same time are the pharmaceutically acceptable acid addition salts thereof "Pharmaceutically acceptable amide" refers to those amides which retain, upon hydrolysis of the amide bond, the biological effectiveness and properties of the carboxylic acid or amine and are not biologically or otherwise undesirable. For a description of pharmaceutically acceptable amides as prodrugs, see, Bundgaard, ed., supra. These amides are typically formed from the corresponding carboxylic acid and an amine. Generally, amide formation can be accomplished via conventional synthetic techniques. See, March et al., Advanced Organic Chemistry, 3rd Ed., p. 1152 (John Wiley Sons, New York 1985), and Mark et al., Encyclopedia of Chemical Technology, (John Wiley Sons, New York 1980). The present invention also contemplates the use of those compositions which are both amides as described herein and at the same time are the pharmaceutically acceptable acid addition salts thereof.
DETAILED DESCRIPTION General The present invention is directed to use of a preferred (3halomethylphenoxy) (4-halophenyl) acetic acid derivatives having the following general formula: 0
R
XF a I X Formula I WO 02/044113 PCT/US01/44603 In Formula I, R is a functional group including, but not limited to, the following: alkoxy, heteroalkoxy, aryloxy, heteroaryloxy, lower aralkoxy, phenyllower alkoxy such as benzyloxy, phenethyloxy; di-lower alkylamino-lower alkoxy and the nontoxic, pharmacologically acceptable acid addition salts thereof, e.g., dimethylaminoethoxy, diethylaminoethoxy hydrochloride, diethylaminoethoxy citrate, diethylaminopropoxy; benzamido-lower alkoxy, benzamidoethoxy or benzamidopropoxy; ureido-lower alkoxy, ureidoethoxy or 1 -methyl-2-ureidoethoxy; N'-lower alkyl-ureido-lower alkoxy, R'NH-CONH-CH 2 n-O- wherein R' represents lower alkyl and n is an integer having a value of from 1 to about 5, N'-ethylureidoethoxy or N'-ethyl-ureidopropoxy; carbamoyl-lower alkoxy, e.g., carbamoylmethoxy or carbamoylethoxy; halophenoxy substituted lower alkoxy, 2- (4-chlorophenoxy) ethoxy or 2 (4 chlorophenoxy)-2-methylpropoxy; carbamoyl substituted phenoxy, 2-carbamoylphenoxy; carboxy-lower alkylamino and the nontoxic, pharmacologically acceptable amine addition salts thereof, e.g., carboxymethylamino cyclohexylamine salt or carboxyethylamine; N,N-di-lower alkylamino-lower alkylamino and the nontoxic, pharmacologically acceptable acid solution salts thereof, N,N-dimethylaminoethylamino hydrochloride, N,Ndiethylaminoethylamino, N,N-diethylaminoethylamino citrate, or N,Ndimethylaminopropylamino citrate; halo substituted lower alkylamino, 2chloroethylamino or 4-chlorobutylamino; hydroxy substituted lower alkylamino, 2hydroxyethylamino, or 3-hydroxypropylamino; lower alkanoyloxy substituted lower alkylamino, acetoxyethylamino or acetoxypropylamino; ureido; arylsulfonamido,
-NHSO
2 Ar; alkylsulfonamido, -NHSO 2
CH
3 lower alkoxycarbonylamino, e.g., methoxycarbonylamino -NHCOOCH 3 or ethyoxycarbonylamino
CHCOOC
2
H
5 In a preferred embodiment, R is selected such that it is a hydrolyzable moiety, such as an ester or amide, and upon hydrolysis of the ester or amide bond, the compound is biologically active such as pharmaceutically acceptable esters or amides as prodrugs. XI, in Formula I, is a halogen; and X 2
X
3 and X 4 are each independently selected from hydrogen and halogen with the proviso that no more than two of X2, X 3 and
X
4 are hydrogen. For each ofX' through X 4 the halogen can be chloro, bromo, fluoro or iodo, preferably chloro or fluoro.
WO 02/044113 PCT/US01/44603 In a preferred embodiment, the present invention relates to use of the (3 -halomethylphenoxy)(4-halophenyl) acetic acid derivatives having the following general formula: R 2 0 0.
x 1 X4 X Formula II In Formula II, R2 is a functional group including, but not limited to, the following: alkyl, heteroalkyl, aryl, heteroaryl, phenyl-lower alkyl, benzamido-lower alkyl, di-lower alkylamino-lower alkyl, ureido-lower alkyl, N'-lower alkyl-ureido-lower alkyl, carbamoyl-lower alkyl, halophenoxy substituted lower alkyl and carbamoyl substituted phenyl. Certain groups are preferred, including phenyl-lower alkyl, benzyl; benzamido-lower alkyl, benzamidoethyl; di-lower alkylamino-lower alkyl, e.g., dimethylaminoethyl or diethylaminopropyl; ureido-lower alkyl, ureidoethyl or 1methyl-2-ureidoethyl; N'-lower alkyl-ureido-lower alkyl, N'-ethyl-ureidoethyl or N'-ethyl-ureidopropyl; carbamoyl-lower alkyl, carbamoylmethyl or carbamoylethyl; halophenoxy substituted lower alkyl, 2-(4-chlorophenoxy)ethyl or 2-(4chlorophenoxy)-2methylpropyl; and carbamoyl substituted phenyl, 2carbamoylphenyl. X 1 in Formula II, is a halogen; and X 2
X
3 and X 4 are each independently selected from hydrogen and halogen with the proviso that no more than two of X 2
X
3 and X 4 are hydrogen. For each of X' through X 4 the halogen can be chloro, bromo, fluoro or iodo, preferably chloro or fluoro. Particularly preferred embodiments are those in which X' is Cl or F, more preferably Cl, and X 2
X
3 and X 4 are combined with the carbon atom to which each is attached to form -CF 3
-CF
2 C1, -CF 2 H and -CH 2
F.
Thus, the term "3-halomethylphenoxy" refers to those phenoxy groups having a monohalomethyl, dihalomethyl or trihalomethyl substituent, preferably a dihalomethyl or trihalomethyl substituent, more preferably, difluoromethyl or trifluoromethyl.
Returning to a discussion of R 2 such groups include, without limitation, the following: CI-C 5 alkyl, C 1 -Cs-cyclic alkyl, C 2
-C
5 alkenyl, and C 2
-C
5 alkynyl, wherein the groups are optionally substituted with one or more halogen atoms; phenyl, naphthyl WO 02/044113 WO 02144113PCTIUS01/44603 and pyridyl, wherein the groups are optionally substituted with one or more substituents selected from the group consisting of halo, C I-C 4 alkyl, C I-C 4 alkoxy, -NO 2 -S(0)m(C I Csalkyl), -OH, -NR 3
R
4 -C0 2
-CONR
3 R, -NR'COR 4
-NR
3
CONR
3
R
4 and -CF,; -(CHR )R -R7OR'; -RO7 2 CR'NR R -RO7OR -R NRICORI; -R 7 NF RI;
-(CH
2 ),oCH(R 3
)(CH
2 )qO 2
CR
9
-(CH
2 )oCH(R 3
)(CH
2 )qKR 4
COR
9
-(CH
2
),CH(R
3
)(CH
2 )qNR 4
CONRR
4
-(CH
2 ),oCH(R 3
)(CH
2 )qNR 4 COOR 0;
-(CH
2 )cCH(R 3
)(CH
2 )qNR 4
SO
2
R'
1 -(CHR),COR 1 2; -(CHI)PNR R;
-(CHR
3 ),CONR'R 1R 4 0 OA (111 t J_ (CHA) R 3 0 1 16R 16- N_ and _1O Subscripts mn, o, q, s, t, u, v and w are integers as follows: mn is 0 to 2; o and q are 0 to pis I to 5;sis I to 3;tis 1 to5;uis0to l;vis 1 to3;andwis 1 to(2v+ R 3 and R 4 are independently H, C 1
-C
5 alkyl, phenyl or benzyl. Ri is H, C 1
-C
5 alkyl or NR 3
R
4 R 6 is phenyl, naphthyl, pyridyl, imidazolyl, indoxyl, indolizinyl, oxazolyl, thiazolyl, thienyl, pyrimidyl, or 1-pyrazolyl optionally substituted with one or more substituents selected from the group consisting of halo, Cl-C 4 alkyl, C 1 -C4 alkoxy, -NO 2 -S(O)m(CI-Csalkyl), -OH, -NR 3
R
4 -C0 2 -CONRWR, -NR'COR 4
-NR
3
CONR
3
R
4 and R 7 is a
C
1
I-C
8 saturated or unsaturated, straight-chain, branched or cyclic alkylene or alkylidene group optionally substituted with one or more groups selected from halo, hydroxyl, thiol, amino, monoalkyl. amino, dialkyl amino, acylamino, carboxyl, alkylcarboxyl, acyl, aryl, aroyl, aralkyl, cyano, nitro, alkoxy, alkenyloxy, alkylcarbonyloxy and arylcarbonyloxy.
R
8 is a C 1 -Cs straight-chain or branched alkylene or alkylildene optionally substituted with one or more groups selected from amino, monoalkyl amino, dialkyl amino, acylamino, hydroxyl, thiol, methyithiol, carboxyl and phenyl. R 9 and R 1 0 are independently H, C 1
-C
alkyl, optionally substituted with one or more groups consisting of C -C 5 alkoxy aryl and heteroaryl, wherein the aryl is phenyl or naphthyl optionally substituted with one or more substituents selected from the group consisting of halo, CI-C 4 alkyl, CI-C 4 alkoxy, -NO 2 -S(O),(Ci-Csalkyl), -OH, -NR 3
R
4 -C0 2 R 5
-CONR
3
R
4
-NR
3
COR
4
-NR
3 CONR 'R 4 and -CFw, and wherein the heteroaryl is pyridyl optionally substituted with one or more substituents selected from the group consisting of halo, CI-C 4 alkyl, C 1
-C
4 alkoxy, -NO 2 -S(O)n(Cj-Csalkyl), -OH, -NR 3
R
4 -C0 2
-CONR
3
R
4
-NR
3
COR
4
-NR
3
CONR
3
R
4 WO 02/044113 WO 02144113PCT/US01I44603 and R1 1 is methyl or phenyl, wherein the phenyl is optionally substituted with methyl and/or -NO 2 R 12 is H, C I-C 5 alkyl, phenyl, benzyl, naphthyl or pyridyl, wherein the C 1
-C
5 alkyl, phenyl, naphthyl, benzyl and pyridyl are optionally substituted with one or more substituents selected from the group consisting of halo, CI-C 4 alkyl, C 1
-C
4 alkoxy, -NO 2 -S(O)m(CI-Csalkyl), -OH, -NR 3
R
4 -C0 2
R
5
-CONR
3
R
4
-NR
3
COR
4
-NR'CONR
3 R and R 13 and R 14 are independently the following: alkyl, alkenyl, aryl, aralkyl or cycloalkyl, wherein the groups are optionally substituted with one or more substituents selected from the group consisting of halo, CI-C 4 alkyl, CI-C 4 alkoxy, -NO 2 -S(O)m(CI-C 5 alkyl), -OH, -NR 3
R
4 -C0 2 R 5
-CONR
3 R1, -NRCOR 4
-NR
3
CONR
3
R
4 -CHNRR, -00CR 1 8 and and wherein R 1 3 and R 14 are included as
-(CHR,)CONR
3 R, NR 13
R
1 is -N -ND OR -N \/NR -N \-\NR 3
COOR
3
\OH
R 00C R RNOC -N or -0 COOR 3 R 1 5 is CF, or CI -C 5 alkyl, wherein C I-C5 alkyl is optionally substituted with the following substituents: C 1
-C
5 alkoxy; phenyl, optionally substituted with one or more substituents selected from the group consisting of halo, C 1
-C
4 alkyl, CI-C 4 alkoxy, -NO 2 -S(O)m(C,-Csalkyl), -OH, -NRR, -C0 2 -CONRWR, -NR 3
COR
4
-NR'CONR
3
R
4 and benzyl, optionally substituted with one or more substituents selected from the group consisting of halo, C 1
-C
4 alkyl, C 1
-C
4 alkoxy, -NO 2 -S(O)nm(CI-Csalkyl), -OH,
-NR
3
R
4 -C0 2
R
5
-CONR
3
-NR
3 COR 4
-NR'CONR
3
R
4 and R" 6 is H, C 1
-C,
alkyl or benzyl. R 17 is C 1
-C
5 alkyl, C 3
-C
8 cyclic alkyl, phenyl or benzyl. R' 8 is H, alkyl, aryl, aralkyl or cycloalkyl, where the group is optionally substituted with one or more substituents selected from the group consisting of halo, CI-C 4 alkyl, CI-C 4 alkoxy, -NO 2 -S(O)m,(C 1
-C
5 alkyI), -OH, -NR 3
R
4 -C0 2
-CONR
3
R
4
_NR
3
COR
4
-NR
3
CONR
3
R
4 and WO 02/044113 PCT/US01/44603 In particularly preferred embodiments, R 2 is selected from -R 7 0R 3
-R'O
2
CRNR
3
R
4
-RCOR
6
-R
7
NR
3
COR
6
-R
7 NR3R6;
-(CH
2 )oCH(R 3
)(CH
2 )qNR 4 COR; -(CH 2 )oCH(R 3
)(CH
2 )qNR 4
CONR
3
R
4
-(CH
2 )oCH(R 3
)(CH
2 )qNR 4 COOR'O; -(CH 2 )oCH(R 3
)(CH
2 )qNR 4
SO
2
R
1
-(CHR
3 )pCO 2 R1 2
-(CHR
3
)CONR
13
R
14 0 A O '(CH2t CH 2
R
3 o R16/N and O O -R17 in which the alkyl portions of any R groups have from one to four carbon atoms, and in further preferred embodiments are unsubstituted alkyl portions of from 1 to 4 carbon atoms.
In the most preferred embodiments, the compound is one of compounds 19.1 through 19.29 in the examples below.
In a further preferred embodiment, the present invention relates to the use of a compound having the formula:
CH
3
HN-
0 0 CI0 0
CF
3 Formula III The compound of Formula III is referred to as 2-acetamidoethyl 4chlorophenyl-(3-trifluoromethylphenoxy) acetate" (also referred to as halofenate").
With reference to each of the formulae above, the substituent groups have art recognized meanings. More particularly, terms such as alkyl are meant to be groups having from 1 to 8 carbon atoms, unless otherwise noted. Similarly, "alkylene" and the like alkoxy, heteroalkyl) refer to groups having 8 or fewer carbon atoms, unless otherwise stated. When the term "lower" is used to refer to an alkyl group (or variant thereof, such as alkoxy, alkanamido, etc.), the group is intended to have from 1 to 4 carbon atoms. The terms "aryl" and "heteroaryl" refer to monocyclic and fused bicyclic groups such as phenyl, naphthyl, pyridyl, benzimidazolyl, quinolinyl, and the like.
Preferred groups for aryl are phenyl and naphthyl, while a preferred heteroaryl group is pyridyl.
Changes in drug metabolism mediated by inhibition of cytochrome P450 enzymes has a very high potential to precipitate significant adverse effects in patients.
Such effects were previously noted in patients treated with racemic halofenate. In the present studies, racemic halofenic acid was found to inhibit cytochrome P450 2C9, an enzyme known to play a significant role in the metabolism of specific drugs. This can lead to significant problems with drug interactions with anticoagulants, anti-inflammatory agents and other drugs metabolized by this enzyme. However, quite surprisingly, a substantial difference was observed between the enantiomers of halofenic acid in their inability to inhibit cytochrome P450 2C9, the enantiomer being about twenty-fold less active whereas the enantiomer was quite potent (see Example Thus, use of the enantiomer of compounds in Formula I, Formula II or Formula II will avoid the inhibition of this enzyme and the adverse effects on drug metabolism previously observed with racemic halofenate.
The present invention encompasses a method of treating insulin resistance in a mammal, the method comprising: administering to the mammal a therapeutically effective amount of a compound having the general structure of Formula I or a pharmaceutically acceptable salt thereof. In a presently preferred embodiment, the compound has the general structure of Formula II. In a further preferred embodiment, the compound has the structure of Formula IR. Quite surprisingly, the method avoids the adverse effects associated with the administration of a racemic mixture of halofenate by providing an amount of the stereoisomer of the compounds in Formula I, Formula II or Formula I which is insufficient to cause the adverse effects associated with the inhibition of cytochrome P450 2C9.
The present invention also encompasses a method of treating Type 2 diabetes in a mammal, the method comprising: administering to the mammal a therapeutically effective amount of a compound having the general structure of Formula I or a pharmaceutically acceptable salt thereof. In a presently preferred embodiment, the compound has the general structure of Formula II In a further preferred embodiment, the compound has the structure of Formula II. Quite surprisingly, the method avoids the adverse effects associated with the administration of a racemic mixture of halofenate by providing an amount of the stereoisomer of the compounds in Formula I, Formula II 27 or Formula II which is insufficient to cause the adverse effects associated with the inhibition of cytochrome P450 2C9.
Also disclosed herein is a method of modulating hyperlipidemia in a mammal, the method comprising: administering to the mammal a therapeutically effective amount of a compound having the general structure of Formula I or a pharmaceutically acceptable salt thereof. In a presently preferred embodiment, the compound has the general structure of Formula II. In a further preferred embodiment, the compound has the structure of Formula Im. Quite surprisingly, the method avoids the adverse effects associated with the administration of a racemic mixture of halofenate by providing an amount of the stereoisomer of the compounds in Formula I, Formula II or Formula III which is insufficient to cause the adverse effects associated with the inhibition ofcytochrome P450 2C9.
The racemic mixture of the halofenate a 1:1 racemic mixture of the two enantiomers) possesses antihyperlipidemic activity and provides therapy and a reduction of hyperglycemia related to diabetes when combined with certain other drugs commonly used to treat this disease. However, this racemic mixture, while offering the expectation of efficacy, causes adverse effects. The term "adverse effects" includes, but is not limited to, nausea, gastrointestinal ulcers, and gastrointestinal bleeding. Other side effects that have been reported with racemic halofenate include potential problems with drug-drug interactions, especially including difficulties controlling anticoagulation with CoumadinTM. Utilizing the substantially pure compounds of the present invention results in clearer dose related definitions of efficacy, diminished adverse effects, and accordingly, an improved therapeutic index. As such, it has now been discovered that it is more desirable and advantageous to administer the enantiomer of halofenate instead of racemic halofenate.
The present invention further encompasses a method of treating hyperuricemia in a mammal, the method comprising: administering to the mammal a therapeutically effective amount of a compound having the general structure of Formula I or a pharmaceutically acceptable salt thereof. In a presently preferred embodiment, the compound has the general structure of Formula II. In a further preferred embodiment, the compound has the structure of Formula III. Quite surprisingly, the method avoids the adverse effects associated with the administration of a racemic mixture of halofenate by providing an amount of the stereoisomer of the compounds in Formula I, Formula II WO 02/044113 PCT/US01/44603 or Formula III which is insufficient to cause the adverse effects associated with the inhibition ofcytochrome P450 2C9.
Enantiomers of Formula I. Formula II and Formula III Many organic compounds exist in optically active forms, they have the ability to rotate the plane of plane-polarized light. In describing an optically active compound, the prefixes R and S are used to denote the absolute configuration of the molecule about its chiral center(s). The prefixes and or and are employed to designate the sign of rotation of plane-polarized light by the compound, with or 1 meaning that the compound is "levorotatory" and with or d is meaning that the compound is "dextrorotatory". There is no correlation between nomenclature for the absolute stereochemistry and for the rotation of an enantiomer. For a given chemical structure, these compounds, called "stereoisomers," are identical except that they are mirror images of one another. A specific stereoisomer can also be referred to as an "enantiomer," and a mixture of such isomers is often called an "enantiomeric" or "racemic" mixture. See, Streitwiesser, A. Heathcock, INTRODUCTION TO ORGANIC CHEMISTRY, 2 nd Edition, Chapter 7 (MacMillan Publishing Co., U.S.A. 1981).
The chemical synthesis of the racemic mixture ofhalofenates (3trihalomethylphenoxy) (4-halophenyl) acetic acid derivatives can be performed by the methods described in U.S. Patent No. 3,517,050, the teaching of which are incorporated herein by reference. The synthesis of the compounds of the present invention is further described in the Examples, supra. The individual enantiomers can be obtained by resolution of the racemic mixture of enantiomers using conventional means known to and used by those of skill in the art. See, Jaques, et al., in ENANTIOMERS, RACEMATES, AND RESOLUTIONS, John Wiley and Sons, New York (1981). Other standard methods of resolution known to those skilled in the art, including but not limited to, simple crystallization and chromatographic resolution, can also be used (see, e.g., STEREOCHEMISTRY OF CARBON COMPOUNDS (1962) E. L. Eliel, McGraw Hill; Lochmuller, J. Chromatography (1975) 113, 283-302). Additionally, the compounds of the present invention, the optically pure isomers, can be prepared from the racemic mixture by enzymatic biocatalytic resolution. Enzymatic biocatalytic resolution has been described previously (see, U.S. Patent Nos. 5,057,427 and 5,077,217, the disclosures of which are incorporated herein by reference). Other methods of obtaining enantiomers WO 02/044113 PCT/US01/44603 include stereospecific synthesis (see, Li, A. J. et al., Pharm. Sci. (1997) 86: 1073- 1077).
The term "substantially free of its stereoisomer," as used herein, means that the compositions contain a substantially greater proportion of the isomer of halofenate in relation to the isomer. In a preferred embodiment, the term "substantially free of its stereoisomer," as used herein, means that the composition is at least 90% by weight of the isomer and 10% by weight or less of the isomer. In a more preferred embodiment, the term "substantially free of its stereoisomer," as used herein, means that the composition contains at least 99% by weight of the isomer and 1% by weight or less of the isomer. In the most preferred embodiment, the term "substantially free of its stereoisomer," means that the composition contains greater than 99% by weight of the isomer. These percentages are based upon the total amount of halofenate in the composition. The terms "substantially optically pure isomer of halofenate," "substantially optically pure halofenate," "optically pure isomer of halofenate" and "optically pure halofenate" all refer to the isomer and are encompassed by the above-described amounts. In addition, the terms "substantially optically pure isomer of halofenate," "substantially optically pure halofenate," "optically pure isomer of halofenate" and "optically pure halofenate" all refer to the isomer and are encompassed by the above-described amounts.
The term "enantiomeric excess" or "ee" is related to the term "optical purity" in that both are measures of the same phenomenon. The value of ee will be a number from 0 to 100, 0 being racemic and 100 being pure, single enantiomer. A compound that is referred to as 98% optically pure can be described as 96% ee.
Combination Therapy With Additional Active Agents The compositions can be formulated and administered in the same manner as detailed below. "Formulation" is defined as a pharmaceutical preparation that contains a mixture of various excipients and key ingredients that provide a relatively stable, desirable and useful form of a compound or drug. For the present invention, "formulation" is included within the meaning of the term "composition." The compounds of the present invention can be used effectively alone or in combination with one or more additional active agents depending on the desired target therapy (see, Turner, N. et al. Prog. Drug Res. (1998) 51: 33-94; Haffner, S. Diabetes Care (1998) 21: 160-178; and DeFronzo, R. et al. Diabetes Reviews (1997) Vol. 5 No. A number of studies WO 02/044113 PCT/US01/44603 have investigated the benefits of combination therapies with oral agents (see, e.g., Mahler, J. Clin. Endocrinol. Metab. (1999) 84: 1165-71; United Kingdom Prospective Diabetes Study Group: UKPDS 28, Diabetes Care (1998) 21: 87-92; Bardin, C. CURRENT THERAPY IN ENDOCRINOLOGY AND METABOLISM, 6 th Edition (Mosby Year Book, Inc., St. Louis, MO 1997); Chiasson, J. et al., Ann. Intern. Med. (1994) 121: 928- 935; Coniff, R. et al., Clin. Ther. (1997) 19: 16-26; Coniff, R. et al., Am. J. Med. (1995) 98: 443-451; and Iwamoto, Y. et al., Diabet. Med. (1996) 13 365-370; Kwiterovich, P.
Am. J. Cardiol (1998) 82(12A): 3U-17U). These studies indicate that diabetes and hyperlipidemia modulation can be further improved by the addition of a second agent to the therapeutic regimen. Combination therapy includes administration of a single pharmaceutical dosage formulation which contains a compound having the general structure of Formula I (or Formula II or Formula III) and one or more additional active agents, as well as administration of a compound of Formula I (or Formula II or Formula III) and each active agent in its own separate pharmaceutical dosage formulation. For example, a compound of Formula I and an HMG-CoA reductase inhibitor can be administered to the human subject together in a single oral dosage composition, such as a tablet or capsule, or each agent can be administered in separate oral dosage formulations.
Where separate dosage formulations are used, a compound of Formula I and one or more additional active agents can be administered at essentially the same time concurrently), or at separately staggered times sequentially). Combination therapy is understood to include all these regimens.
An example of combination therapy that modulates (prevents the onset of the symptoms or complications associated) atherosclerosis, wherein a compound of Formula I is administered in combination with one or more of the following active agents: an antihyperlipidemic agent; a plasma HDL-raising agent; an antihypercholesterolemic agent, such as a cholesterol biosynthesis inhibitor, an hydroxymethylglutaryl (HMG) CoA reductase inhibitor (also referred to as statins, such as lovastatin, simvastatin, pravastatin, fluvastatin, and atorvastatin), an HMG-CoA synthase inhibitor, a squalene epoxidase inhibitor, or a squalene synthetase inhibitor (also known as squalene synthase inhibitor); an acyl-coenzyme A cholesterol acyltransferase (ACAT) inhibitor, such as melinamide; probucol; nicotinic acid and the salts thereof and niacinamide; a cholesterol absorption inhibitor, such as p-sitosterol; a bile acid sequestrant anion exchange resin, such as cholestyramine, colestipol or dialkylaminoalkyl derivatives of a cross-linked dextran; an LDL (low density lipoprotein) receptor inducer; fibrates, such as clofibrate, WO 02/044113 PCT/US01/44603 bezafibrate, fenofibrate, and gemfibrizol; vitamin B 6 (also known as pyridoxine) and the pharmaceutically acceptable salts thereof, such as the HCI salt; vitamin B12 (also known as cyanocobalamin); vitamin B 3 (also known as nicotinic acid and niacinamide, supra); anti-oxidant vitamins, such as vitamin C and E and beta carotene; a beta-blocker; an angiotensin II antagonist; an angiotensin converting enzyme inhibitor; and a platelet aggregation inhibitor, such as fibrinogen receptor antagonists glycoprotein IIb/IIIa fibrinogen receptor antagonists) and aspirin. As noted above, the compounds of Formula I can be administered in combination with more than one additional active agent, for example, a combination of a compound of Formula I with an HMG-CoA reductase inhibitor lovastatin, simvastatin and pravastatin) and aspirin, or a compound of Formula I with an HMG-CoA reductase inhibitor and a P blocker.
Another example of combination therapy can be seen in treating obesity or obesity-related disorders, wherein the compounds of Formula I can be effectively used in combination with, for example, phenylpropanolamine, phentermine, diethylpropion, mazindol; fenfluramine, dexfenfluramine, phentiramine, 03 adrenoceptor agonist agents; sibutramine, gastrointestinal lipase inhibitors (such as orlistat), and leptins. Other agents used in treating obesity or obesity-related disorders wherein the compounds of Formula I can be effectively used in combination with, for example, neuropeptide Y, enterostatin, cholecytokinin, bombesin, amylin, histamine H 3 receptors, dopamine D 2 receptors, melanocyte stimulating hormone, corticotrophin releasing factor, galanin and gamma amino butyric acid (GABA).
Still another example of combination therapy can be seen in modulating diabetes (or treating diabetes and its related symptoms, complications, and disorders), wherein the compounds of Formula I can be effectively used in combination with, for example, sulfonylureas (such as chlorpropamide, tolbutamide, acetohexamide, tolazamide, glyburide, gliclazide, glynase, glimepiride, and glipizide), biguanides (such as metformin), thiazolidinediones (such as ciglitazone, pioglitazone, troglitazone, and rosiglitazone); dehydroepiandrosterone (also referred to as DHEA or its conjugated sulphate ester, DHEA-SO 4 antiglucocorticoids; TNFa inhibitors; a-glucosidase inhibitors (such as acarbose, miglitol, and voglibose), pramlintide (a synthetic analog of the human hormone amylin), other insulin secretogogues (such as repaglinide, gliquidone, and nateglinide), insulin, as well as the active agents discussed above for treating atherosclerosis.
WO 02/044113 PCT/US01/44603 A further example of combination therapy can be seen in modulating hyperlipidemia (treating hyperlipidemia and its related complications), wherein the compounds of Formula I can be effectively used in combination with, for example, statins (such as fluvastatin, lovastatin, pravastatin or simvastatin), bile acid-binding resins (such as colestipol or cholestyramine), nicotinic acid, probucol, betacarotene, vitamin E, or vitamin C.
In accordance with the present invention, a therapeutically effective amount of a compound of Formula I (or Formula II or Formula III) can be used for the preparation of a pharmaceutical composition useful for treating diabetes, treating hyperlipidemia, treating hyperuricemia, treating obesity, lowering triglyceride levels, lowering cholesterol levels, raising the plasma level of high density lipoprotein, and for treating, preventing or reducing the risk of developing atherosclerosis.
Additionally, an effective amount of a compound of Formula I (or Formula II or Formula III) and a therapeutically effective amount of one or more active agents selected from the group consisting of: an antihyperlipidemic agent; a plasma HDL-raising agent; an antihypercholesterolemic agent, such as a cholesterol biosynthesis inhibitor, for example, an HMG-CoA reductase inhibitor, an HMG-CoA synthase inhibitor, a squalene epoxidase inhibitor, or a squalene synthetase inhibitor (also known as squalene synthase inhibitor); an acyl-coenzyme A cholesterol acyltransferase inhibitor; probucol; nicotinic acid and the salts thereof; niacinamide; a cholesterol absorption inhibitor; a bile acid sequestrant anion exchange resin; a low density lipoprotein receptor inducer; clofibrate, fenofibrate, and gemfibrozil; vitamin B 6 and the pharmaceutically acceptable salts thereof; vitamin B 1 2 an anti-oxidant vitamin; a 1 -blocker; an angiotensin II antagonist; an angiotensin converting enzyme inhibitor; a platelet aggregation inhibitor; a fibrinogen receptor antagonist; aspirin; phentiramines, 03 adrenergic receptor agonists; sulfonylureas, biguanides, a-glucosidase inhibitors, other insulin secretogogues, and insulin can be used together for the preparation of a pharmaceutical composition useful for the abovedescribed treatments.
Pharmaceutical Formulations and Methods of Administration In the methods of the present invention, the compounds of Formula I, Formula II, and Formula III can be delivered or administered to a mammal, a human patient or subject, alone, in the form of a pharmaceutically acceptable salt or hydrolyzable precursor thereof, or in the form of a pharmaceutical composition where the compound is WO 02/044113 PCT/US01/44603 mixed with suitable carriers or excipient(s) in a therapeutically effective amount. By a "therapeutically effective dose", "therapeutically effective amount", or, interchangeably, "pharmacologically acceptable dose" or "pharmacologically acceptable amount", it is meant that a sufficient amount of the compound of the present invention, alternatively, a combination, for example, a compound of the present invention, which is substantially free of its stereoisomer, and a pharmaceutically acceptable carrier, will be present in order to achieve a desired result, alleviating a symptom or complication of Type 2 diabetes.
The compounds of Formula I, Formula II, and Formula III that are used in the methods of the present invention can be incorporated into a variety of formulations for therapeutic administration. More particularly, the compounds of Formula I (or Formula II or Formula III) can be formulated into pharmaceutical compositions by combination with appropriate, pharmaceutically acceptable carriers or diluents, and can be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, pills, powders, granules, dragees, gels, slurries, ointments, solutions, suppositories, injections, inhalants and aerosols. As such, administration of the compounds can be achieved in various ways, including oral, buccal, rectal, parenteral, intraperitoneal, intradermal, transdermal, intratracheal administration. Moreover, the compound can be administered in a local rather than systemic manner, in a depot or sustained release formulation. In addition, the compounds can be administered in a liposome.
In addition, the compounds of Fommla I, Formula II or Formula III can be formulated with common excipients, diluents or carriers, and compressed into tablets, or formulated as elixirs or solutions for convenient oral administration, or administered by the intramuscular or intravenous routes. The compounds can be administered transdermally, and can be formulated as sustained release dosage forms and the like.
Compounds of Formula I, Formula II, or Formula III can be administered alone, in combination with each other, or they can be used in combination with other known compounds (discussed supra). In pharmaceutical dosage forms, the compounds can be administered in the form of their pharmaceutically acceptable salts thereof. They can contain hydrolyzable moieties. They can also be used alone or in appropriate association, as well as in combination with, other pharmaceutically active compounds.
Suitable formulations for use in the present invention are found in Remington's Pharmaceutical Sciences (Mack Publishing Company (1985) Philadelphia, PA, 17th which is incorporated herein by reference. Moreover, for a brief review of WO 02/044113 PCT/US01/44603 methods for drug delivery, see, Langer, Science (1990) 249:1527-1533, which is incorporated herein by reference. The pharmaceutical compositions described herein can be manufactured in a manner that is known to those of skill in the art, by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes. The following methods and excipients are merely exemplary and are in no way limiting.
For injection, the compounds can be formulated into preparations by dissolving, suspending or emulsifying them in an aqueous or nonaqueous solvent, such as vegetable or other similar oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids or propylene glycol; and if desired, with conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifying agents, stabilizers and preservatives. Preferably, the compounds of the present invention can be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer. For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
For oral administration, the compounds of Formula I, Formula II, or Formula III can be formulated readily by combining with pharmaceutically acceptable carriers that are well known in the art. Such carriers enable the compounds to be formulated as tablets, pills, dragees, capsules, emulsions, lipophilic and hydrophilic suspensions, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated. Pharmaceutical preparations for oral use can be obtained by mixing the compounds with a solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP). If desired, disintegrating agents can be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions can be used, which can optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer WO 02/044113 PCT/US01/44603 solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments can be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
Pharmaceutical preparations which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds can be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers can be added. All formulations for oral administration should be in dosages suitable for such administration.
For buccal administration, the compositions can take the form of tablets or lozenges formulated in conventional manner.
For administration by inhalation, the compounds for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas, or from propellant-free, dry-powder inhalers. In the case of a pressurized aerosol the dosage unit can be determined by providing a valve to deliver a metered amount. Capsules and cartridges of, gelatin for use in an inhaler or insufflator can be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
The compounds can be formulated for parenteral administration by injection, by bolus injection or continuous infusion. Formulations for injection can be presented in unit dosage form, in ampules or in multidose containers, with an added preservative. The compositions can take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and can contain formulator agents such as suspending, stabilizing and/or dispersing agents.
Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds can be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions can contain substances which increase the viscosity of the suspension, such as sodium WO 02/044113 PCT/US01/44603 carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension can also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions. Alternatively, the active ingredient can be in powder form for constitution with a suitable vehicle, sterile pyrogen-free water, before use.
The compounds can also be formulated in rectal compositions such as suppositories or retention enemas, containing conventional suppository bases such as cocoa butter, carbowaxes, polyethylene glycols or other glycerides, all of which melt at body temperature, yet are solidified at room temperature.
In addition to the formulations described previously, the compounds can also be formulated as a depot preparation. Such long acting formulations can be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the compounds can be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
Alternatively, other delivery systems for hydrophobic pharmaceutical compounds can be employed. Liposomes and emulsions are well known examples of delivery vehicles or carriers for hydrophobic drugs. In a presently preferred embodiment, long-circulating, stealth, liposomes can be employed. Such liposomes are generally described in Woodle, et al., U.S. Patent No. 5,013,556, the teaching of which is hereby incorporated by reference. The compounds of the present invention can also be administered by controlled release means and/or delivery devices such as those described in U.S. Pat. Nos. 3,845,770; 3,916,899; 3,536,809; 3,598,123; and 4,008,719; the disclosures of which are hereby incorporated by reference.
Certain organic solvents such as dimethylsulfoxide (DMSO) also can be employed, although usually at the cost of greater toxicity. Additionally, the compounds can be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent. Various types of sustainedrelease materials have been established and are well known by those skilled in the art.
Sustained-release capsules can, depending on their chemical nature, release the compounds for a few hours up to over 100 days.
The pharmaceutical compositions also can comprise suitable solid or gel phase carriers or excipients. Examples of such carriers or excipients include but are not WO 02/044113 PCT/US01/44603 limited to calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
Pharmaceutical compositions suitable for use in the present invention include compositions wherein the active ingredients are contained in a therapeutically effective amount. The amount of composition administered will, of course, be dependent on the subject being treated, on the subject's weight, the severity of the affliction, the manner of administration and the judgment of the prescribing physician. Determination of an effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.
For any compound used in the method of the present invention, a therapeutically effective dose can be estimated initially from cell culture assays or animal models.
Moreover, toxicity and therapeutic efficacy of the compounds described herein can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, by determining the LD 5 0 (the dose lethal to 50% of the population) and the ED 5 0 (the dose therapeutically effective in 50% of the population).
The dose ratio between toxic and therapeutic effect is the therapeutic index and can be expressed as the ratio between LD 5 0 and EDs 5 0 Compounds Which exhibit high therapeutic indices are preferred. The data obtained from these cell culture assays and animal studies can be used in formulating a dosage range that is not toxic for use in human. The dosage of such compounds lies preferably within a range of circulating concentrations that include the EDs 50 with little or no toxicity. The dosage can vary within this range depending upon the dosage form employed and the route of administration utilized. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. (See, Fingl et al. 1975 In: The Pharmacological Basis of Therapeutics, Ch. 1).
The amount of active compound that can be combined with a carrier material to produce a single dosage form will vary depending upon the disease treated, the mammalian species, and the particular mode of administration. However, as a general guide, suitable unit doses for the compounds of the present invention can, for example, preferably contain between 100 mg to about 3000 mg of the active compound. A preferred unit dose is between 500 mg to about 1500 mg. A more preferred unit dose is between 500 to about 1000 mg. Such unit doses can be administered more than once a day, for example 2, 3, 4, 5 or 6 times a day, but preferably I or 2 times per day, so that the WO 02/044113 PCT/US01/44603 total daily dosage for a 70 kg adult is in the range of 0.1 to about 250 mg per kg weight of subject per administration. A preferred dosage is 5 to about 250 mg per kg weight of subject per administration, and such therapy can extend for a number of weeks or months, and in some cases, years. It will be understood, however, that the specific dose level for any particular patient will depend on a variety of factors including the activity of the specific compound employed; the age, body weight, general health, sex and diet of the individual being treated; the time and route of administration; the rate of excretion; other drugs which have previously been administered; and the severity of the particular disease undergoing therapy, as is well understood by those of skill in the area.
A typical dosage can be one 10 to about 1500 mg tablet taken once a day, or, multiple times per day, or one time-release capsule or tablet taken once a day and containing a proportionally higher content of active ingredient. The time-release effect can be obtained by capsule materials that dissolve at different pH values, by capsules that release slowly by osmotic pressure, or by any other known means of controlled release.
It can be necessary to use dosages outside these ranges in some cases as will be apparent to those skilled in the art. Further, it is noted that the clinician or treating physician will know how and when to interrupt, adjust, or terminate therapy in conjunction with individual patient response.
Protecting Groups Certain compounds having the general structure of Formula I and II may require the use of protecting groups to enable their successful elaboration into the desired structure. Protecting groups can be chosen with reference to Greene, T. et al., Protective Groups in Organic Synthesis, John Wiley Sons, Inc., 1991. The blocking groups are readily removable, they can be removed, if desired, by procedures which will not cause cleavage or other disruption of the remaining portions of the molecule.
Such procedures include chemical and enzymatic hydrolysis, treatment with chemical reducing or oxidizing agents under mild conditions, treatment with fluoride ion, treatment with a transition metal catalyst and a nucleophile, and catalytic hydrogenation.
Examples of suitable hydroxyl protecting groups are: trimethylsilyl, triethylsilyl, o-nitrobenzyloxycarbonyl, p-nitrobenzyloxycarbonyl, t-butyldiphenylsilyl, t- WO 02/044113 WO 02/44113PCT/USOI/44603 butyldimethylsilyl, benzyloxycarbonyl, t-butyloxycarbonyl, 2,2,2trichioroethyloxycarbonyl, and allyloxycarbonyl. Examples of suitable carboxyl protecting groups are benzhydryl, o-nitrobenzyl, p-nitrobenzyl, 2-naphthylmethyl, allyl, 2-chioroallyl, benzyl, 2,2,2-trichioroethyl, trimethylsilyl, t-butyldimethylsilyl, tbutyldiphenylsilyl, 2-(trimethylsilyl)ethyl, phenacyl, p-methoxybcnzyl, acetonyl, pmethoxyphenyl, 4-pyridylmethyl and t-butyl.
Process Processes for making the compounds of the present invention are generally depicted in Schemes 1 and 2 (and further described in the Examples): Scheme 1: 0 ci CN 20% NaOH OH
C
8 H6CIN 170.60O 151.60 106 0
OH
0-0-t
C
15
H
10 C1F 3 0 3 330.69
CF,
0 ciC cat. py 0 1 §HC1 2
F
3 0 2
CF
3 0HN 349.14 CHN
C
9 HBrC]O 2 1. SOC1 2 118.97 263.52 2. Br 2 159.81 0 3. MeOH 32.04 OMe
CI
-Br
C
7
H
5
F
3 0 HO 162.11 NaOH CF 3 KOH H 2 0
C
16
H
12
CIF
3 0 3 344.72 CH,
HNCH
3 Miine 1 0 ,HCF34 415.80 2 103.12 CF 3 Iti.lu According to Scheme 1, a substituted phenyl acetonitrile is converted to a substituted phenyl acetic acid. The substituted phenyl acetic acid is converted to an activated acid derivative acid chloride), followed by halogenation at the alphacarbon and esterification with an alcohol. The halogenated ester is treated with a substituted phenol 3-trifluoromethyiphenol), yielding an aryl ether, which is hydrolyzed to form a carboxylic acid derivative. The acid derivated is converted to an WO 02/044113 PCT/US01/44603 activated acid derivative and subsequently treated with a nucleophile Nacetylethanolamine) to afford the desired product.
Scheme 2:
CH
3
HN-
0 1) Thionyl Chloride 0 0 OH 2. Bromine O 3. N-Acetylethanolamine Br 1. KOH/IPA HO CF 3
CH
3
HN
CF
3 According to Scheme 2, a substituted phenyl acetic acid is converted to an activated acid derivative acid chloride) followed by halogenation at the alphacarbon. The activated acid portion of the molecule is reacted with a nucleophile Nacetylethanolamine) to provide a protected acid. The halogenated, protected acid is treated with a substituted phenol 3-trifluoromethylphenol), yielding the desired product.
The stereoisomers of the compounds of the present invention can be prepared by using reactants or reagents or catalysts in their single enantiomeric form in the process wherever possible or by resolving the mixture of stereoisomers by conventional methods, discussed supra and in the Examples. Some of the preferred methods include use of microbial resolution, resolving the diastereomeric salts formed with chiral acids or chiral bases and chromatography using chiral supports.
Kits WO 02/044113 PCT/US01/44603 In addition, the present invention provides for kits with unit doses of the compounds of Formula I, Formula II, or Formula III either in oral or injectable doses.
In addition to the containers containing the unit doses will be an informational package insert describing the use and attendant benefits of the drugs in alleviating symptoms and/or complications associated with Type 2 diabetes as well as in alleviating hyperlipidemia and hyperuricemia. Preferred compounds and unit doses are those described herein above.
EXAMPLES
The compounds of Formula I, Formula II, or Formula III of the present invention can be readily prepared using the process set forth in Scheme 1, supra, and from the following examples.
EXAMPLE 1 This example relates to the preparation of Methyl Bromo-(4chlorophenyl)-acetate.
0 1. SOC 2
O
OH 2. Br 2 OMe CI 3. MeOH CI Br C8H 7
CO
2 CgHBrCIO 2 170.60 263.52 The initial compound listed in Scheme 1, 4-chlorophenylacetic acid, is readily available from several commercial sources Aldrich and Fluka).
A 5-L Morton reactor equipped with a magnetic stirrer, a pot temperature control, and addition funnel was vented through a gas scrubber and charged with pchlorophenylacetic acid (720 gm, 4.2 moles) and SOC12 (390 ml, 630 gm, 5.3 moles).
The reaction was stirred, heated and held at 550 +50 C for 1 hour. Bromine (220 ml., 670 gm, 5.3 moles) was then added over 20 min. and stirred at 550+50 C for 16 hours. The temperature was raised to 800 C for 7 hours and then cooled to 90 C in an ice-water bath.
Methanol (2.0 L, 1.6 kg, 49.4 moles) was then carefully added. The solvent was stripped to obtain 2 liquids weighing 1.28kg. These were dissolved in a mixture of 0.84 L water and 2.1 L ether and separated. The organic phase was washed once with 0.78 L aqueous NaCl and dried over 0.13 kg MgSO 4 This was filtered through Whatman #1 filter paper and stripped of solvent to obtain 0.985 kg of orange liquid. The proton WO 02/044113 PCT/US01/44603 NMR showed this to be 80% product and 19% non-brominated ester. The HPLC showed 82% product and 18% non-brominated ester. HPLC was run on a Zorbax SB-C8 column at 30° C measuring 250 X 4.6 mm and 5 I particle size. The mobile phase was 60:40 acetonitrile: 0.1% H 3 P0 4 at 1.5 ml/min. Detection was at 210 nm. The injected sample of 1 jl was dissolved in acetonitrile at a concentration of 10 mg/ml. The product had a retention time of 5.0 min. and that of the non-brominated ester was 3.8 min. This crude product was purified by vacuum distillation to obtain 96% pure product with an 84% yield. The product proton NMR (CDC1 3 300 MHz) showed shifts at 3.79 3H), 5.32 1H) and 7.20-7.55 4H) ppm.
EXAMPLE 2 This example relates to the preparation of Methyl 4-Chlorophenyl-(3trifluoromethylphenoxy)-acetate.
0 OMe K-tBuO OMe Cl Ot HO- CQ C1 BrTHF
C
g HaBrCIO 2
C
7 H5F 3 0 C 1 6
H
12
CIF
3 0 3
CF
3 26352 162.11 344.72 This step was similar to the same step in U.S. 3,517,050 with one exception, potassium t-butoxide was used in place of sodium methoxide to prevent generation of the corresponding methyl ether. A 5-L Morton reactor equipped with an overhead stirrer, a pot temperature detector, and addition funnel and under a nitrogen atmosphere was charged with methyl bromo-(4-chlorophenyl)-acetate (830 gm, moles) and THF (600 ml). The reactor was cooled to 140 +30 C in an ice-water bath and then a similarly cooled solution of trifluoromethyl-m-cresol (530 gm, 3.3 moles) in 1.0 M potassium t-butoxide in THF (3.1 L, 3.1 moles) was added. The reaction proceeded exothermically with a typical temperature rise exceeding 250 C and the addition was controlled to maintain a temperature of 150+2° C and stirred at ambient temperature for 2 hours. HPLC was run on a Zorbax SB-C8 column at 300 C measuring 250 x 4.6 mm and 5 p. particle size. The mobile phase was 60:40 acetonitrile: 0.1% H 3 P0 4 at ml/min. Detection was at 210 nm. The injected sample of 1 pl was dissolved in acetonitrile at a concentration of 10 mg/ml. The product had a retention time of 9.6 min., the starting ester eluted at 5.0 min., the phenol at 3.0 and the non-brominated ester at 3.8 min. The solvent was stripped using a rotary evaporator to obtain a yellow slush that was dissolved in a mixture of 4.0 L water and 12.0 L ether. The mixture was separated and WO 02/044113 PCT/US01/44603 the organic phase was washed once with 1.6 L 5% aqueous NaOH followed by 1.6 L water and finally 1.6 L 25% aqueous NaCI. The organic phase was dried over 0.32 kg MgSO 4 and filtered through Whatman #1 filter paper. The solvent was stripped to obtain 1.0 kg of damp, off-white crystals. This was recrystallized on the rotary evaporator by dissolving in 1.0 L methylcyclohexane at 750 C and then cooling to 200 C.
The crystals were filtered through Whatman #1 filter paper and washed with three 0.25 L portions of cool (150 C) methylcyclohexane. The wet product (0.97 kg) was dried overnight to obtain 0.81 kg of 98% pure product that corresponds to a 79% yield. The product proton NMR (CDC13, 300 MHz) shows shifts at 3.75 3H), 5.63 1H) and 7.05-7.55 8H).
EXAMPLE 3 This example relates to the preparation of 4-Chlorophenyl-(3trifluoromethylphenoxy)-acetic Acid 0 o S-OMe OH ci l' KOH H 2 0 CI O
S
E 1
OH
CeH2,CIF30 3 a C 15 Hs CIF 3 0 3 344.72
C
F3 330.69 CF 3 A 12-L Morton reactor with magnetic stirrer, pot temperature controller, a reflux condenser and under a nitrogen atmosphere was charged with methyl 4chlorophenyl-(3-trifluoromethylphenoxy)-acetate (810 gm, 2.3 moles) and absolute ethanol (5.8 L) and heated with stirring to 570 C to dissolve the solid. A solution of KOH (520 gm, 9.3 moles) in 0.98 L water was added. The solution was refluxed for 30 min.
and solvent was stripped by a rotary evaporator to obtain 2.03 kg of a mixture of two nearly colorless liquids. These were dissolved in water (16 L) and treated with 16 gm neutral Norit, then filtered through a pad of infusorial earth retained on Whatman #1 filter paper. The pH of the filtrate was lowered from an initial range of 13 to a range of 1 to 2 by adding a total of 2.75 L of 3 M HCI (8.25 moles). A very sticky solid formed after the addition of the first 2.30 L of acid and ether (7 L) was added at this point. The two layers were separated and the organic layer was dried over MgSO 4 (230 gm) and filtered through Whatman #1 filter paper. The solvent was then stripped to obtain 0.85 kg water-white syrup. The material was then recrystallized on the rotary evaporator by adding methylcyclohexane (800 ml) and cooling to 18° C with slow rotation. The WO 02/044113 PCT/US01/44603 temperature was then dropped to 5° C, the crystals were filtered, and washed 5 times with 0.10 L portions of cold C) methylcyclohexane to obtain 0.59 kg wet crystals. The wet crystals were dried to obtain 0.48 kg (62% yield) product with no p-chlorophenylacetic acid detectable in the proton NMR. The product proton NMR (CDC13, 300 MHz) shows shifts at 5.65 1H), 7.02-7.58 8H) and 10.6 1H).
EXAMPLE 4 This example relates to the preparation of resolved enantiomers of 4- Chlorophenyl-(3-trifluoromethylphenoxy)-acetic Acid.
Ci OH followed OH CHO N by Ho C1 5 HioCIF 3 0 3
CF
3 1 9
H
2
N
2 0 Resolved CF 3 330.69 294.40 A 12-L open-top Morton reactor with an overhead stirrer was charged with 4-chlorophenyl-(3-trifluoromethyl-phenoxy)-acetic acid (350 gm, 1.06 moles) and isopropanol (4.0 L) and heated to 650+30 C. A slurry cinchonidine (300 gm, 1.02 moles) in isopropanol (2.0 L) was added, rinsing all solid into the reactor with an additional 0.8 L of isopropanol. The temperature dropped from 650 to 560 C and a transparent, orange solution ultimately formed and the mixture was held at 550+50 C for 2 hours. Fine crystals were collected by filtration through Whatman #1 filter paper, washing once with 0.7 L hot (55' C) isopropanol. The crystals were dried for 16 hours at ambient temperature in a 12.6-L vacuum oven under a 5 LPM nitrogen flow. The dry solid weighed 0.37 kg and had an 80% enantiomeric excess (ee) of the enantiomer.
The enantiomeric excess was determined by HPLC using a 250 x 4.6 mm R,R-WhelkO-l column at ambient temperature. Injected samples were 20 gl of 2 mg/ml solutions of the samples in ethanol. The column was eluted with 95:5:0.4, hexane:isopropanol:acetic acid at a flow of 1 ml/min. Detection was at 210 nm. The enantiomer eluted at 7 to 8 min.
and the enantiomer at 11 to 13 min. The mother liquor dropped a second crop almost immediately that was filtered, washed, and dried to afford 0.06 kg salt that has a 90% ee of the (-)-enantiomer. Similarly third, fourth and fifth crops weighing 0.03 kg, 0.03 kg and 0.7 kg, respectively, were obtained; with enantiomer excesses of 88%, 89% and 92%, respectively.
WO 02/044113 PCT/US01/44603 The crude salt (320 gm) was recrystallized from a mixture of ethanol (5.9 L) methanol (1.2 The mixture was heated with overhead stirring to dissolve, cooled at ambient temperature for 16 hours, filtered and washed twice with 0.20 L of 5:1 ethanol:methanol. The crystals were dried to obtain 0.24 kg of the enantiomer that had an ee of 97%. This corresponded to an 80% recovery of this isomer. The resolved salt was suspended in a mixture of ether (6.5 L) and water (4.0 L) with overhead stirring. The pH was lowered to 0-1 as measured by pH indicating strips with a solution of concentrated H 2
SO
4 (0.13 L) in water (2.5 The phases were separated and the organic phase and washed twice with 6.5 L portions of water. Ether (1.9 L) was added and the organic layer washed once more with 6.5 L water. After the final separation, 0.1 L of 25% aqueous NaCl was added clean up any slight emulsion. The product was dried over 0.19 kg MgSO 4 filtered and solvent removed solvent to obtain 0.13 kg of water-white syrup that solidifies on cooling. This corresponded to a 97% recovery of product that had a 95% ee of the enantiomer. [ac]D +5.814c (c.=0.069 in methyl alcohol).
The combined, crude salt (200 gm) was recrystallized from isopropanol (3.1 The mixture was heated to dissolve almost all of the solid and fast-filtered to remove insoluble solids. The mixture was then cooled with stirring at ambient temperature for 16 hours, filtered, washed, and dried to obtain 0.16 kg of the enantiomer that has an ee of 97%. This corresponds to a 49% recovery of this isomer.
The enantiomer of the acid was isolated in the same manner as described above for the acid. The resolved salt was suspended in ether and water, the pH lowered with concentrated H 2
SO
4 and the product extracted in the organic phase.
EXAMPLE A. Preparation of 4-Chlorophenyl-(3-trifluoromethylphenoxy)acetyl Chloride ci °OH SOCI, C HCI 3 0 C15H1,CIF303 l C1i5HCI2F302 330.69 CF 3 349.14 CF 3 A 2-L evaporation flask with magnetic stirrer, Claissen adapter, pot thermometer and a reflux condenser routed to a gas scrubber was charged with 4- WO 02/044113 PCT/US01/44603 chlorophenyl-(3-trifluoromethylphenoxy)-acetic acid (143 g, 0.42 mole based on 97% purity) and CHCl 3 (170 ml) and heated to boiling in order to dissolve. SOC12 (38 ml, 62.1 gm, 0.52 mole) was added. The mixture was heated to reflux (680 C final) for 4.5 hours and then stripped of volatiles to obtain 151 g yellow, turbid liquid (103% apparent yield).
The material was used in the next step without further purification.
B. Preparation of 4-Chlorophenyl-(3-trifluoromethylphenoxy)acetyl Chloride 0 SCHCI 1 oCIF30 3 C1HgCI 2F302 33069 CF 3 349.14 CF 3 A 3-L evaporation flask with magnetic stirrer, Claissen adapter, pot thermometer and a reflux condenser routed to a gas scrubber was charged with 4chlorophenyl-(3-trifluoromethylphenoxy)-acetic acid (131 g, 0.37 mole) and CHC1 3 (152 ml) and heated to boiling in order to dissolve. SOC1 2 (35 ml, 56.5 g, 0.48 mole) was added. The mixture was heated to reflux (700 C final) for 4 hours and then stripped of volatiles to obtain 139 g liquid. The material was used in the next step without further purification.
EXAMPLE 6 A. Preparation of 2-Acetamidoethyl 4-Chlorophenyl-(3trifluoromethylphenoxy)-acetate C15HgC2F301 CH ether CI- 349.14 C 4
H
9 N02 10 C 349.14 CF 3 103.12 C 1 9
H
1 7
CIF
3 N0 4 415.80
CF
3 A 3-L round-bottom flask with magnetic stirrer, pot thermometer, under a nitrogen atmosphere and in an ice-water bath was charged with DMF (420 ml), pyridine (37 ml, 36 g, 0.46 mole) and N-acetoethanolamine (39 ml, 43 g, 0.42 mole). The mixture was cooled to 0° to 50 C and a solution of crude 4-chlorophenyl-(3trifluoromethylphenoxy)-acetyl chloride (151 gm, 0.42 mole based on 100% yield of previous step) in ether (170 ml) was added over a 40 min. period so as to maintain the pot WO 02/044113 PCT/US01/44603 temperature below 130 C. The mixture was stirred at ambient temperature for 16 hours and dissolved by adding water (960 ml) followed by ethyl acetate (630 ml). The water addition proceeded exothermically raising the temperature from 240 to 340 C. Ethyl acetate addition caused a temperature drop to 300 C. The layers were separated and the aqueous phase extracted once with ethyl acetate (125 ml). The combined organic layers were extracted once with 7% aqueous NaHCO 3 (125 ml) and five times with 60 ml portions of water and then twice with 60 ml portions of 25% aqueous NaC1. The product was dried over MgSO 4 (42 g) and filtered through Whatman #1 filter paper.
Solvent was stripped using a rotary evaporator to obtain 160 g of a yellow syrup corresponding to an 80% yield based on the proton nmr that shows 87% product, 8% EtOAc, 4% non-brominated amide, and 1% DMF. This syrup was dissolved in MTBE (225 ml) at ambient temperature and chilled (-150 C) 85% hexanes (400 ml) was added with stirring. Two liquids formed, then crystals, then the mixture formed a solid. The solid mass was scraped onto a Buchner funnel fitted with Whatman packed down and washed three times with 100 ml portions of 1:1 MTBE:hexanes to obtain 312 g wet product which dries to 127 gm, corresponding to a 73% yield.
B. Preparation of 2-Acetamidoethyl 4-Chlorophenyl-(3trifluoromethylphenoxy)-acetate 0
CH
3
-C
l H DMF HN-" ec HO CHoat:eCo. C15H 9
CI
2
F
3 o 2 0 g2 ether ci/ V" 349.14 F C4HgNo 2 100 c 349.14 CF 3 103.12 C,9H 17
CIF
3 N0 4 415.80
CF
3 A 3-L round-bottom flask with magnetic stirrer, pot thermometer, under a nitrogen atmosphere and in an ice-water bath was charged with DMF (365 ml), pyridine (33 ml, 32.3 g, 0.41 mole) and N-acetoethanolamine (34 ml, 38.1 g, 0.37 mole). The mixture was cooled to 00 to 50 C and a solution of crude 4-chlorophenyl-(3trifluoromethylphenoxy)-acetyl chloride (139 gm, 0.37 mole based on 100% yield of previous step) in ether (155 ml) was added over a 25 min. period so as to maintain the pot temperature below 130 C. The mixture was stirred at ambient temperature 40 hours and dissolved by adding water (850 ml) followed by ethyl acetate (550 ml). The water addition proceeded exothermically raising the temperature from 240 to 340 C. Ethyl acetate addition caused a temperature drop to 300 C. The layers were separated and the WO 02/044113 PCT/US01/44603 aqueous phase extracted once with ethyl acetate (110 ml). The combined organic layers were washed twice with 55 ml portions of water and then five times with 55 ml portions of 25% aqueous NaCI and dried over 30 g MgSO 4 and filtered through Whatman #1 filter paper. Solvent was stripped using a rotary evaporator to obtain 168 g yellow liquid corresponding to an 86% yield based on the proton nmr that shows 79% product, 9% EtOAc, 8% non-brominated amide, and 4% DMF. The product was crystallized in an 800-ml beaker by dissolving in MTBE (200 ml) at ambient temperature, cooling at -150 for 1.4 hours, adding 200 ml 85% hexanes and then chilling 1 hour. The solid mass was scraped out onto a Buchner funnel fitted with Whatman packed down and washed once with 1:1 MTBE:hexanes (100 ml) to obtain 201 gm wet product. The product was dried under nitrogen flow and triturated with 85% hexanes (700 ml) using an overhead stirrer. The material was filtered and dried to obtain 87 gm product. [a]D +2.7690 (c.=0.048 in methyl alcohol). [a]D-2.7160 (c.=0.049 in methyl alcohol). The and enantiomers were also analyzed by HPLC using a 250 x 4.6 mm R,R- WhelkO-1 column at ambient temperature. Injected samples were 20 pl of 2 mg/ml solutions of the samples in ethanol. The column was eluted with 60:40, isopropanol:hexane at a flow of 1 ml/min. Detection was at 220 nm. The enantiomer eluted at 5.0 to 5.2 min. and the enantiomer at 5.7 to 5.9 min.
EXAMPLE 7 This example relates to the inhibition of cytochrome P450 2C9 (CYP2C9) by the compounds of the present invention.
14 Tolbutamide hydroxylation activity (100 utM C-tolbutamide; 1 mM NADPH) was assayed in pooled human liver microsomes (0.6 mg protein/ml)) for minutes at 37 0 C both with and without test compounds. Racemic halofenic acid, halofenic acid and halofenic acid were tested (0.25 gM to 40 IM). As shown in Figure 1, racemic halofenic acid inhibited CYP2C9-mediated tolbutamide hydroxylation activity in human liver microsomes with an apparent IC5o of 0.45 utM. A substantial difference was noted in the ability of the enantiomers of halofenic acid to inhibit CYP2C9. The halofenic acid had an apparent IC 5 0 of 0.22 j[M whereas the halofenic acid was almost 20-fold less potent with an apparent IC5o of 3.6 uM.
WO 02/044113 PCT/US01/44603 EXAMPLE 8 This example relates to the time course of glucose-lowering for the compounds of the present invention.
A. Material and Methods Male, 9-10 weeks old, C57BL/6J ob/ob mice were purchased from The Jackson Laboratory (Bar Harbor, ME, USA). Animals were housed (4-5 mice/cage) under standard laboratory conditions at 22 0 C and 50% relative humidity, and were maintained on a diet of Purina rodent chow and water ad libitum. Prior to treatment, blood was collected from the tail vein of each animal. Mice that had non-fasting plasma glucose levels between 300 and 500 mg/dl were used. Each treatment group consisted of 10 mice that were distributed so that the mean glucose levels were equivalent in each group at the start of the study. Mice were dosed orally once by gavage with either vehicle, racemic halofenate (250 mg/kg), halofenate (250 mg/kg) or halofenate (250 mg/kg). All compounds were delivered in a liquid formulation contained 5% dimethyl sulfoxide (DMSO), 1% tween 80 and 2.7% methylcellulose. The gavage volume was ml/kg. Blood samples were taken at 1.5, 3, 4.5, 6, 7.5, 9 and 24 hour after the dose and analyzed for plasma glucose. Plasma glucose concentrations were determined colorimetrically using glucose oxidase method (Sigma Chemical Co, St. Louis, MO, USA). Significance difference between groups (comparing drug-treated to vehicletreated or between drug-treated groups) was evaluated using Student unpaired t-test.
B. Results As illustrated in Figure 2, racemic halofenate significantly reduced plasma glucose concentrations at most of the timepoints with the peak activity at 9 hours. Halofenate showed a plasma glucose reduction as-early as 1.5 hours and reached its peak activity at 3 hours. The plasma glucose concentrations remained low up to 24 hours. Halofenate did not show significant activity until 4.5 hours and the peak activity was at hours. Plasma glucose started to rebound afterward. There were significant differences between and enantiomers of halofenate at the 3 and 24-hour timepoints. The activity of the halofenate was more rapid onset and sustained longer.
EXAMPLE 9 This example relates to the Glucose lowering activity of the compounds of the present invention.
WO 02/044113 PCT/US01/44603 A. Materials and Methods Male, 8-9 weeks old, C57BL/6J ob/ob mice were purchased from The Jackson Laboratory (Bar Harbor, ME, USA). Animals were housed (4-5 mice/cage) under standard laboratory conditions at 22 0 C and 50% relative humidity, and were maintained on a diet of Purina rodent chow and water ad libitum. Prior to treatment, blood was collected from the tail vein of each animal. Mice that had non-fasting plasma glucose levels between 300 and 520 mg/dL were used. Each treatment group consisted of mice that were distributed so that the mean glucose levels were equivalent in each group as the start of the study. Mice were dosed orally by gavage once a day for 5 days with either vehicle, racemic halofenate (250 mg/kg), halofenate (125 and 250 mg/kg) or halofenate (125 and 250 mg/kg). Racemic halofenate was delivered in 2.7% (w/v) methylcellulose and both the enantiomer and enantiomer were delivered in a liquid formulation contained 5% dimethyl sulfoxide (DMSO), 1% tween 80 and 2.7% methylcellulose. The gavage volume was 10 ml/kg. Blood samples were taken at 3, 6, 27, 30 and 120 hour after the first dose and analyzed for plasma glucose and insulin. The animals were fasted overnight (14 hours) before the 120 hours sampling.
Plasma glucose concentrations were determined colorimetrically using glucose oxidase method (Sigma Chemical Co, St. Louis, MO, USA). Plasma insulin concentrations were determined by using the Rat Insulin RIA Kit from Linco Research Inc. (St. Charles, MO, USA). Significance difference between groups (comparing drug-treated to vehicletreated) was evaluated using Student unpaired t-test.
B. Results As illustrated in Figure 3, halofenate significantly reduced plasma glucose concentrations at 6, 27 and 30 hours. Halofenate at both dosage levels significantly lowered plasma glucose concentrations at 6, 27 and 30 hours. The high-dose (250 mg/kg) was also active at 3 hours. Halofenate at 125 mg/kg showed plasma glucose reduction at 6 and 27 hours, where at 250 mg/kg, lowered plasma glucose concentrations were observed at 3, 6, 27 and 30 hours. Plasma insulin levels are shown in Figure 4. Racemic halofenate significantly reduced insulin at 6 and 27 hours. Plasma insulins were significantly reduced in the halofenate group at 27 hours at both doses and was significantly reduced at 30 hours in the animals treated with 250 mg/kg/day. halofenate significantly reduced insulin at 27 and 30 hours at both doses. At 125 mg/kg/day a significant reduction was also observed after 6 hours. After fasting overnight (at 120 hours), all treatments reduced plasma glucose concentrations WO 02/044113 PCT/US01/44603 significantly (Figure Plasma insulins were significantly reduced in all halofenate treated groups except the halofenate at 125 mg/kg/day (Figure 6).
EXAMPLE This example relates to the improvement in Insulin Resistance and Impaired Glucose Tolerance for the compounds of the present invention.
A. Materials and Methods Male, 8-9 weeks old Zucker fa/fa rats (Charles River, were housed (2-3 rats/cage) under standard laboratory conditions at-22 0 C and 50% relative humidity, and were maintained on a diet of Purina rodent chow and water ad libitum. Prior to treatment, rats were assigned to 6 groups based on body weight. Each treatment group consisted of 8 rats. Rats were dosed orally once by gavage with either vehicle, racemic halofenate (100 mg/kg), halofenate (50 or 100 mg/kg) or halofenate (50 or 100 mg/kg). All compounds were delivered in a liquid formulation contained 5% dimethyl sulfoxide (DMSO), 1% tween 80 and 2.7% methylcellulose. The gavage volume was 10 ml/kg. All rats received an oral glucose challenge (1.9 g/kg) 5.5 hours after the treatment and 4 hours after withdrawal of the food. Blood samples were taken at 0, 60, 90, 120, and 180 minutes following the glucose challenge for plasma glucose measurement. The vehicle, halofenate (50 mg/kg) and halofenate (50 mg/kg) groups were subjected to an insulin challenge following daily gavage of the respective treatments for 5 days. On day 5, rats received the intravenous insulin (0.75 U/kg) hours after the last dose and 4 hours after withdrawal of the food. Blood samples were taken at 3, 6, 9, 12, 15 and 18 minutes following the insulin injection for plasma glucose measurement. Plasma glucose concentrations were determined colorimetrically using glucose oxidase method (Sigma Chemical Co, St. Louis, MO, Significance difference between groups (comparing drug-treated to vehicle-treated or between drugtreated groups) was evaluated using Student unpaired t-test.
B. Results As illustrated in Figure 7A, Zucker fatty rats with Impaired Glucose Tolerance had lower plasma glucose levels after a glucose challenge following treatment with halofenate. The halofenate was the most effective in lowering the glucose and had an effect that persisted longer than the racemate or enantiomer. Figure 7B shows the incremental area under the curve (AUC) for all the treatment groups. The animals treated with the halofenate showed significant reductions in the glucose area relative WO 02/044113 PCT/US01/44603 to vehicle-treated controls. Although the AUC was decreased in the groups treated with the racemate or halofenate, the effects were not as great as in the halofenatetreated rats and the differences were not statistically significant.
Changes in insulin sensitivity were assessed by monitoring the fall in glucose after an intravenous injection of insulin. The slope of the line is a direct indication of the insulin sensitivity of the test animal. As shown in Figure 8, the insulin sensitivity was improved significantly after 5 days of treatment with halofenate compared to the vehicle-treated controls (p 0.01) and animals treated with halofenate (p 0.05). Treatment with halofenate had a small effect on insulin sensitivity that was not significantly different from the vehicle-treated control (p 0.083).
Treatment with halofenate substantially reduced the insulin resistance in the Zucker fatty rat, a well-established model of Impaired Glucose Tolerance and insulin resistance.
EXAMPLE 11 This example relates to the lipid lowering activity of the compounds of the present invention.
A. Materials and methods Male Zucker diabetic fatty (ZDF) rats were obtained from GMI Laboratories (Indianapolis, IN) at 9 weeks of age. Vehicle or enantiomers of halofenate administered by oral gavage on a daily basis starting at 74 days of age. Initial blood samples were obtained for analysis one day before treatment and at the indicated times in the treatment protocol. Blood was analyzed for plasma triglyceride and cholesterol by standard techniques.
B. Results In experiment I animals received a dose of 25 mg/kg/day. As shown in Figure 9A and Figure 9B, a significant decrease in plasma cholesterol was noted only in animals treated with the (-)halofenate after 7 and 13 days of treatment. In Experiment II, animals at 107 days of age received daily doses of either 12.5 mg/kg/day or 37.5 mg/kg/day of the and enantiomers of halofenate. As shown in Figure 10A and Figure 10B, the plasma cholesterol was significantly lower on the high dose after 7 days but not after 14 days of treatment with the halofenate. In contrast, for the halofenate at the low dose, a significant decrease in cholesterol was observed after 7 days.
At the high dose a much greater decline in plasma cholesterol was noted that was apparent both after 7 and 14 days of treatment. As shown in Figure 11A and Figure 11B, WO 02/044113 PCT/US01/44603 a significant decrease in plasma triglyceride was also noted 7 days after treatment at the high dose which was of greater magnitude in animals treated with the enantiomer of halofenate.
EXAMPLE 12 This example relates to the glucose lowering activity of halofenate analogs and halofenate analogs.
A. Materials and methods Male, 8-9 weeks old, C57BL/6J ob/ob mice were purchased from The Jackson Laboratory (Bar Harbor, ME, USA). Animals were housed (4-5 mice/cage) under standard laboratory conditions at 22 3 0 C temperature and 50 20% relative humidity, and were maintained on a diet of Purina rodent chow and water ad libitum.
Prior to treatment, blood was collected from the tail vein of each animal. Mice that had non-fasting plasma glucose levels between 250 and 500 mg/dl were used. Each treatment group consisted of 8-10 mice that were distributed so that the mean glucose levels were equivalent in each group at the start of the study. Mice were dosed orally by gavage once a day for 1-3 days with either vehicle, halofenic acid, analog 14, 29, 33, 34, 36, 37, or 38 at 125 mg/kg or analog 29, 36, 37 or 38 at 150 mg/kg. Compounds were delivered in a liquid formulation containing 5% dimethyl sulfoxide (DMSO), 1% tween 80 and 0.9% methylcellulose. The gavage volume was 10 ml/kg.
Blood samples were taken at 6 hours after the each dose and analyzed for plasma glucose.
Food intake and body weight were measured daily. Plasma glucose concentrations were determined colorimetrically using glucose oxidase method (Sigma Chemical Co, St.
Louis, MO, USA). Significant difference between groups (comparing drug-treated to vehicle-treated) was evaluated using the Student unpaired t-test.
B. Results As illustrated in Table 2, compounds were evaluated in 5 different experiments. Single dose halofenic acid significantly reduced plasma glucose concentrations at 6 hours. Analog 14 significantly lowered plasma glucose concentrations at 6, 30 and 54 hours. Analog 33 significantly lowered plasma glucose concentrations at 6 and 54 hours. Analog 29 and 38 significantly lowered plasma glucose concentrations at 6, 30 and 54 hours. Analog 35 and 36 significantly lowered plasma glucose concentrations at 30 and 54 hours. Analog 37 significantly lowered plasma WO 02/044113 WO 02/44113PCT/US01/44603 glucose concentrations at 54 hours. Single dose analogs 29, 36, 37 and 38 significantly reduced plasma glucose concentrations at 6 hours. Compound treatments did not affect the animal's food intake and body weight.
0 R 2 x 0 cX 3 Formula HI Table 1: and Halofenate analogs. Compounds described in reference to Formula 11.
Cmpd No. X CX 3
R
halofenic Cl CF 3
H
acid 14 F CF 3
(CH
2 2
NHAC
29 Br CF 3
(CH
2 2 NIJAc 33 Cl CF 3
(CH
2 3
CH
3 Cl CF 3
(CH
2 2
N(CH
3 2 36 Cl CF 3
(CH
2 2 NHCOPh 37 Cl CF 3
CH
2 CONH4 2 38 Cl CF 3
CH
2
CON(CH
3 2 WO 02/044113 WO 02/44113PCTIIJS01I44603 Table 2 Glucose-lowerine Activities of (±)Halofenate and (-)Halofenate Analogs Predose 6 hours 30 hours 54 hours Glucose Glucose P Glucose P Glucose P (mgldl) (mg/dI) VALUE (mg/dl) VALUE (mgf dl) VALUE vs. veh vs. veb vs. veb Vehicle 313±18 303±19.8 NA NA (-)halofenic 312.9±17.7 163.8±11.8 0.0011 NA NA acid Vehicle 360.2±27.8 405.8±25.8 356.0±27.6 386.1±20.6 (±)Analog 14 361.0±17.1 328.9+34.1 0.0444 267.0±21.3 0.0099 293.0±29.4 0.0092 Vehicle 291.6±18.5 363.0±25.1 340.8±30.0 351.5±23.8 (±L)Analog 33 292.0±19.1 227.5±13.2 0.0001 298.0±15.3 0.1119 286.6±9.9 0.0125 Vehicle 387.1±14.3 371.5±24.2 326.2±L22.5 374.0+37.9 (±L)Analog 29 387.1±16.0 299.7±24.5 0.0259 237.4±14.9 0.0020 293.3±9.7 0.0268 (±:)Analog 35 387.0±18.0 319.6±26.7 0.0834 276.8±17.6 0.0504 286.2±31.5 0.0458 (±)Analog 37 3 87.4±L18.8 345.4±=19.7 NS 312.5121.7 N S 285.1±114.7 0.0210 Vehicle 329.6±16.1 361.8±23.2 346.5±24.6 379.2±24.4 (±L)Analog 36 329.7±17.6 300.5±27.3 0.0522 249.7±k8.6 0.0008 2 72.2+L18.4 0.0013 (±)Analog 38 329.4±18.9 303.2±18.2 0.0312 245.6±15.6 0.0014 243.1±10.6 0.0000 Vehicle 373.0±13.6 405.8±33.7 NA NA (-)Analog 36 373.2±15.5 281.1±18.2 0.0019 NA jNA (-)Analog 37 373.4±16.1 271.7+22.5 0.0018 NA NA (-)Analog 38 373.4±16.1 251.2+23.6 0.0007 NA NA (-)Analog 29 372.2±17.1 333.5±16.1 0.0353 NA NA WO 02/044113 PCT/US01/44603 EXAMPLE 13 This example relates to a comparison between the activities of(-) halofenate and halofenate.
A. Materials and methods Male 8-9 week old ZDF rats were purchased from Genetic Models, Inc.
(Indianapolis, IN). Animals were housed (3 rats/cage) under standard laboratory conditions at 22 3 °C temperature and 50 20% relative humidity, and were maintained on a diet of Purina rodent chow and water ad libitum. Prior to treatment, blood was collected from the tail vein of each animal. Rats that had 4-hour fasting plasma glucose levels between 200 and 500 mg/dL were used. Each treatment group consisted of 8-10 rats that were distributed so that the mean glucose levels were equivalent in each group at the start of the study. Rats were dosed orally by gavage once a day for 3 days with either vehicle, halofenate or halofenate at 50 mg/kg. Compounds were delivered in a liquid fomulation containing 5% dimethyl sulfoxide (DMSO), 1% tween and 0.9% (wiv) methylcellulose. The gavage volume was 5 ml/kg. Blood samples were taken at 5 hours post dose on day 2 and 3. Plasma glucose concentrations were determined colorimetrically using glucose oxidase method (Sigma Chemical Co, St.
Louis, MO, USA). Significant difference between groups (comparing drug-treated to vehicle-treated) was evaluated using the Student unpaired t-test.
B. Results Oral administration halofenate at 50 mg/kg significantly reduced plasma glucose concentrations while halofenate at the same dosage levels failed to reduce plasma glucose concentrations as compared to vehicle-treated animals (Figure 12).
EXAMPLE 14 This example relates to a pharmacokinetic study of halofenate and halofenate.
A. Materials and methods Male 225-250 g SD rats were purchased from Charles River. Animals were housed (3 rats/cage) under standard laboratory conditions at 22 3 C temperature and 50 20% relative humidity, and were maintained on a diet of Purina rodent chow and water ad libitum. A catheter was placed in the left carotid artery under sodium pentobarbital (50 mg/kg and animals were allowed to recover for 2 days before WO 02/044113 PCT/US01/44603 treatment. Single dose halofenate or halofenate at 50 mg/kg were administered by oral gavage. Compounds were delivered in a liquid formulation containing 5% (v/v) dimethyl sulfoxide (DMSO), 1% tween 80 and 0.9% methylcellulose. The gavage volume was 5 ml/kg. Blood samples were collected at 1, 2, 4, 6, 8, 12, 24, 48, 72, 96 and 120 hours post dose. The plasma samples were analyzed for each enantiomeric acid halofenic acid and halofenic acid) by a chiral specific HPLC assay, since the esters are prodrugs, which are designed to convert to their respective enantiomeric acids in vivo.
B. Results After oral administration of halofenate, both halofenic acid and halofenic acid were detected in the plasma samples. As shown in table 3, it appeared that the two enantiomeric acids had different dispositional profiles. The elimination of(-) halofenic acid was much slower than halofenic acid. As a result, the AUC of(-) halofenic acid was significantly higher than the AUC for halofenic acid, 4708.0 vs.
758.0 tg-h/mL and the terminal half-life was 46.8 vs. 14.3 hours.
After oral administration of halofenate, the dispositional profile of halofenic acid was basically identical to the administration of halofenate as the terminal half-life is the same (Table The Cmax and AUC halofenic acid were proportionally higher simply due to higher amount of halofenate administered (Table Halofenic acid was also detected in the plasma but the concentration was much lower than halofenic acid. It is speculated that halofenic acid was formed in vivo since the terminal half-life (T1/ 2 of both acids was similar.
These results suggest the use halofenate is more desirable since the AUC of halofenic acid was significantly higher than the AUC for halofenic acid.
WO 02/044113 WO 02/44113PCT[USOI/44603 Table 3: Pharmacokinetic Analysis of Halofenate (-Enantiomer) and() Halofenate Enantiomer) Drug administered ()Halofenate (n 3) ()Halofenate (n =1 Enantiomner Dose administered* 50 mg/kg 0 (metabolite) 25 mg/kg 25 mg/kg ([tg/mL) 114.6+29.7 2.4 ±0.5 65.2 30.5 Tmax (hours) 8-12 6-12 12 6 AUC(g-h/mL) 7159+ 1103 164.3 +79.3 4708 758 T 1/ 2 (hours) 46.4 4.7 F 41.7 11.8 46.8 1 14.3 The dose of each enantiomer in halofenate is 50% of the total dose of the racemic mixture.
Table 4: Plasma Concentrations of halofenic acid and halofenic acid following a single dose of H- halofenate.
Compoun I Analyzed (Vg/mL) Time (hour) halofenic acid I halofenic acid 8 Rat 9Rat 11 IRat 8 O Ra 9 Ra1 4122.3 36.9 94.5 1.67 BOL 1.95 6128.3 56.5 116.3 2.96 BOL 1.73 8 128.2 79.0 127.8 2.58 BOL 2.06 12 135.3 80.6 104.8 2.85 2.23 2.08 24 82.5 73.1 66.5 2.22 1.29 1.86 48 56.2 44.5 47.1 1.64 1.03 1.14 72 39.7 37.4 30.8 1.25 BQL BOL 96 131.1 N/ 24. BQL N/A BQL 120 1120.3 1N/A N/A IBQL N/A N/A *BQL Below Quantifiable Limit 1.00 j tg/rnL N/A Sample not available WO 02/044113 PCT/US01/44603 EXAMPLE This example relates to the prevention of the development of diabetes and the alleviation of hypertriglyceridemia by halofenate.
A. Materials and methods Male, 4 weeks old, C57BL/6J db/db mice were purchased from The Jackson Laboratory (Bar Harbor, ME, USA). Animals were housed (5 mice/cage) under standard laboratory conditions at 22 3°C temperature and 50 20% relative humidity, and were maintained on a powder diet of Purina rodent chow (#8640) and water ad libitum. Prior to treatment, blood was collected from the tail vein of each animal for plasma glucose, insulin and triglyceride concentrations. Mice were distributed so that the mean glucose levels and body weight were equivalent in each group at the start of the study. The control group (20 mice) was put on powder chow mixed with 5% sucrose and the treatment group (20 mice) was put on powder chow mixed with 5% sucrose and halofenate. The amount of halofenate in the chow was adjusted continuously according the animal's body weight and food intake to meet the target dosage of 150 mg/kg/day. Blood samples were taken at 8-10 AM once a week for 9 weeks under nonfasting condition. Food intake and body weight were measured every 1-3 days. Plasma glucose and triglyceride concentrations were determined colorimetrically using kits from Sigma Chemical Co (No. 315 and No. 339, St. Louis, MO, USA). Plasma insulin levels were measured using RIA assay kit purchased from Linco Research (St. Charles, MO).
Significant differences between groups (comparing drug-treated to vehicle-treated) was evaluated using Student unpaired t-test.
B. Results C57BL/6J db/db mice at 4 weeks of age are in a pre-diabetic state. Their plasma glucose concentrations are normal, but the plasma insulin concentrations are significantly elevated. As illustrated in Figure 13, the plasma glucose concentrations in both groups were normal at the start of the experiment. Following the natural course of diabetes development, plasma glucose levels in the control group increased progressively as the animals aged, while the increase of plasma glucose levels in the halofenate treated group was prevented or significantly delayed. As depicted in Figure 15, about of mice did not develop diabetes in the halofenate treated group when diabetes is defined as plasma glucose levels >250 mg/dl. On the other hand, none of the mice in the control group was free of diabetes by the age of 10 weeks. Consistent with the plasma WO 02/044113 PCT/US01/44603 glucose finding, plasma insulin in the control group decreased progressively, indicating deterioration of the ability of the pancreas to secret insulin. Halofenate treatment maintained the plasma insulin concentration, indicating prevention of the deterioration of pancreatic function (Figure 14).
Figure 16 shows progression of the plasma triglyceride concentrations versus age in C57BL/6J db/db mice. Halofenate administration alleviated the increase of plasma triglyceride concentration over the course of the experiment.
EXAMPLE 16 This example describes the preparation of 2-Acetamidoethyl 4- Chlorophenyl-(3-trifluoro methylphenoxy)-acetate halofenate).
CH
3
HN
0 1)Thionyl Chloride 0 0 cl )OH 2. Bromine CI 0 3. N-Acotylethanolamine Br 4-Chlorophenylacetic acid was combined with 1,2-dichloroethane and the resulting solution was heated to 45 Thionyl chloride was added to the reaction mixture, which was heated at 60 oC for 18 hours. The reaction was allowed to cool to room temperature and was then added slowly to a solution of N-acetylethanolamine in dichloromethane. After stirring 30 min., the reaction was quenched with aqueous potassium carbonate and sodium thiosulfate. The organic layer was washed with water, dried over magnesium sulfate and filtered. Removal of the solvent by rotary evaporation provided N-acetylaminoethyl 2-bromo-2-(4-chlorophenyl)acetate as an oil.
CH3 CH 3
HN
C
HN-
H
SFC OH 1i. KOH/IPA C Br l J 2. Resolulion 3-Hydroxybenzotrifluoride was added to a solution of potassium hydroxide in isopropanol. N-acetylaminoethyl 2-bromo-2-(4-chlorophenyl)acetate in isopropanol was added to the isopropanol/phenoxide solution and stirred at room temperature for 4 hours. The isopropanol was removed by vacuum distillation, and the resulting slush was dissolved in ethyl acetate and washed twice with water and once with brine. After drying over magnesium sulfate and filtration, the solvent was removed to WO 02/044113 PCT/US01/44603 give crude product as an oil. The crude product was dissolved in hot toluene/hexanes (1:1 v/v) and cooled to between 0 and 10 °C to crystallize the product. The filter cake was washed with hexanes/toluene (1:1 v/v) and then dried under vacuum at 50 OC. The isolated solid was dissolved in hot 1:6 isopropanol in hexanes. After cooling, the pure racemic 2-Acetamidoethyl 4-Chlorophenyl-(3-trifluoro methylphenoxy)-acetate formed as a crystalline solid. The solid was collected by filtration, the filter cake washed with 1:6 isopropanol in hexanes and dried under vacuum at 50 oC.
The racemic compound was dissolved in a solution of 20% isopropanol (IPA) and 80% hexane at 2.5% (wt/wt). The resulting solution was passed over a Whelk- O R,R Chiral Stationary Phase (CSP) in continuous fashion until >98% ee extract could be removed. The solvent was evaporated from the extract under reduced pressure to provide 2-Acetamidoethyl 4-Chlorophenyl-(3-trifluoro methylphenoxy)-acetate. (The Simulated Moving Bed resolution was conducted by Universal Pharm Technologies LLC of 70 Flagship Drive, North Andover, MA 01845.) EXAMPLE 17 This example relates to the lowering of plasma uric acid levels through the administration of halofenate.
A. Materials and methods Male SD rats, weight 275-300 g were purchased from Charles River.
Animals were housed (3 rats/cage) under standard laboratory conditions at 22 3 0
C
temperature and 50 20% relative humidity, and were maintained on a powder diet of Purina rodent chow (#8640) and water ad libitum. To establish a hyperuricemic state, animals were put on a diet containing 2.5% of oxonic acid (Sigma Chemical Co, St. Louis, MO, USA) throughout the experiment. Oxonic acid elevates plasma uric acid by inhibiting uricase. Rats were screened for plasma uric acid levels 3 days after they were placed on the diet, and those that had extreme plasma uric acid levels were excluded. Rats were assigned to one of three groups and the mean uric acid levels were equivalent in each group. Rats were dosed orally by gavage once a day for 3 days with either vehicle, halofcnate or halofenate at 50 mg/kg. On the 4 t h day, respective rats received halofenate or halofenate at 100 mg/kg and all rats received an i.p.
WO 02/044113 PCT/US01/44603 injection ofoxonic acid (250 mg/kg) 4 hours after the oral gavage. Halofenate and halofenate were delivered in a liquid formulation containing 5% dimethyl sulfoxide (DMSO), 1% tween 80 and 0.9% methylcellulose. Oxonic acid was delivered in a liquid formulation containing 0.9% methylcellulose. The gavage and injection volumes were 5 ml/kg. Blood samples were taken at 6 hours post oral gavage on day 4.
Plasma uric acid levels were determined colorimetrically using the Infinity Uric Acid Reagent (Sigma Chemical Co, St. Louis, MO, USA). Significant difference between the groups (comparing drug-treated to vehicle-treated) was evaluated using the Student unpaired t-test.
B. Results As shown in Figure 17, oral administration of halofenate significantly reduced plasma uric acid levels. Halofenate also lowered plasma uric acid levels, but it was not statistically significant.
EXAMPLE 18 This example relates to the inhibition of cytochrome P450 isoforms by the compounds of the present invention.
A. Materials and methods The following probe substrates were used to investigate the inhibitory potential of the test article on the cytochrome P450 isoforms 1A2, 2A6, 2C9, 2C19, 2D6, 2E1 and 3A4: 100 pM phenacetin (CYP1A2), 1 pM coumarin (CY)2A6), 150 jM tolbutamide (CYP2C9), 50 pM S-mephenytoin (CYP2C19), 16 jIM dextromethorphan (CYP2D6), 50 jM chlorzoxazone (CYP2E1), and 80 pM testosterone (CYP3A4). The activity of each isoform was determined in human hepatic microsomes in the presence and absence of the test article.
Unless otherwise noted, all incubations were conducted at 37 OC. The sample size was N 3 for all test and positive control conditions and N 6 for all vehicle control conditions. Halofenic acid (MW 330) was prepared at room temperature as 1000X stocks in methanol, then diluted with Tris buffer to achieve final concentrations of 0.33, 1.0, 3.3, 10 and 33.3 pM, each containing 0.1% methanol. A vehicle control (VC) consisting of microsomes and substrate in Tris buffer containing 0.1% methanol without WO 02/044113 PCT/US01/44603 the test article was included for all experimental groups. Positive control (PC) mixtures were prepared using the following known CYP450 inhibitors: 5 uM furafylline (CYP1A2), 250 |JM tranylcypromine (CYP2A6), 50 ptM sulfaphenazole (CYP2C9), itM omeprazole (CYP2C19), 1 [tM quinidine (CYP2D6), 100 4tM 4-methylpyrazole (CYP2E1), and 5 tM ketoconazole (CYP3A4). A chromatographic interference control (CIC) was included to investigate the possibility of chromatographic interference by the test article and its metabolites. The test article at 33.3 Ig/mL) was incubated with 1X microsomal protein, 1X NRS, and 10 jiL of an appropriate organic for an appropriate time period as described below.
Stable, frozen lots of pooled adult male and female hepatic microsomes prepared by differential centrifugation of liver homogenates were used in this study (see, Guengerich, F.P. (1989). Analysis and characterization of enzymes. In Principles and Methods of Toxicology Hayes, 777-813. Raven Press, New York.).
Incubation mixtures were prepared in Tris buffer to contain microsomal protein (1 mg/mL), each concentration of the probe substrates (as 100X stocks), and the test article (at each concentration) or PC as appropriate for each isoform. After a preincubation at 37 NADPH regenerating system (NRS) was added to initiate the reactions, and the samples were incubated at 37 °C for the following time periods: minutes for phenacetin (CYP1A6), 20 minutes for coumarin (CYP2A6), 40 minutes for tolbutamide (CYP2C9), 30 minutes for S-mephenytoin (CYP2C19), 15 minutes for dextromethorphan (CYP2D6), 20 minutes for chlorozoxazone (CYP2E1), and 10 minutes for testosterone (CYP3A4). Incubation reactions were terminated at the appropriate time with the addition of an equal volume of methanol, except for the incubations with Smephenytoin, which were terminated with the addition of 100 JL of perchloric acid. All substrates were evaluated near their respective Km concentrations, as previously indicated.
After each incubation, the activities of the P450 isoforms were determined by measuring the rates of metabolism for the respective probe substrates. The metabolites monitored for each probe substrate were as follows: acetaminophen for CYPIA2; 7hydroxycoumarin for CYP2A6; 4-hydroxytolbutramide for CYP2C9; 4hydroxymephenytoin for CYP2C19; dextrorphan for CYP2D6; 6-hydroxychlorzoxazone for CYP2E1; and 6p-hydroxytestosterone for CYP3A4. Activities were analyzed using HPLC (In Vitro Technologies, Inc., Baltimore, MD).
Inhibition was calculated using the following equation: WO 02/044113 PCT/US01/44603 Percent Inhibition [(vehicle control treatment)/vehicle control] x 100 Percent inhibition data for the test article was presented in a tabular format. Descriptive statistics (mean and standard deviation) of each test article concentration were calculated, then presented to show inhibitory potency. IC 50 values were also calculated for the test article using a 4-parameter curve fitting equation in Softmax 2.6.1.
Measures of time, temperature, and concentration in this example are approximate.
B. Results The results for each of the 7 isoforms of cytochrome P450, expressed as metabolic activity and percentage of inhibition, are presented in Tables 5-8. Halofenic acid inibited 4-hydroxytolbutamide production (CYP2C9, IC50 11 tiM) and also inhibited 4-hydroxymephenytoin production (CYP2C19) at the 10 and 33 jtM dose levels. Inhibition of other CYP450 isoforms was not observed. It should be noted that the IC50 for CYP2C9 in this experiment was approximately three times that reported in Example 7 (11 itM as compared to 3.6 jM). This result is most likely due, at least in part, to the use of a lower purity halofenic acid (lower ee) in Example 7.
WO 02/044113 WO 02144113PCTIUS01/446O3 Table 5: Hepatic microsomal activities of phenacetin (CYP1A2) and coumnarin (CYP2A6) in male and female human microsomes incubated with halofenic acid at doses of 0.33, 1.0, 3.3, 10, and 33.3 jaM Control/ Phenacetin Coumnarin Test Cone AC Production %7-HC% Article (4aM) (pmolmg Inhibitio Production Inhibition protein/min) n (pmollmg protein/rnin) CIC 33.3 0.00 0.00 NA 0.00 ±0.00 N VC 0.1% 118±2 0 32.0±1.4 0 FUR 5 54.5±1.3 54 NA NA TRAN 250 NA NA 0.00±0.00 100 H- 0.33 116 ±2 1 33.3±0.7 -4 halofenic 1.0 118 ±2 0 32.6±0.7 -2 acid 3.3 119 ±2 -1 32.1±0.7 0 119±2 -1 33.1±0.7 -3 33.3 119 ±2 -1 32.3±0.7 -1
IC
50 NA
NA
Values are the mean ±standard deviation of N =3 samples (VC: N Abbreviations: Conc, concentration; AC, acetaminophen; 7-HC, 7-hydroxycoumnarin; CIC, chromatographic interference control; VC, vehicle control 1% methanol); NA, not applicable; FUR, furafylline; TRAN, tranylcypromine.
WO 02/044113 WO 02/44113PCT/US01/44603 Table 6: Hepatic microsomal activities of tolbutamide (CYP2C9) and Smephenytoin (CYP2CI9) in male and female human microsomes incubated with(halofenic acid at doses of 0.33, 1.0, 3.3, 10, and 33.3 p.M Control/ Tolbutamide S-Mephenytoin Test Conc 4-OH TB %4-OH ME% Article (piM) Production Inhibitio Production Inhibition (pmollmg n (pmollmg protein/min) protein/mmn) CIC 33.3 0.00+±0.00 NA 0.00 0.00 NA VC 0.1% 43.0 1.4 0 3.17 0.29 0 OMP 10 NA NA 1.58 ±0.05 SFZ 50 BQL -400 NA NA H- 0.33 41.0 0.9 5 3.03 0.03 4 halofenic 1.0 38.6 ±0.5 10 3.01 ±0.07 acid 3.3 34.2 ±0.2 21 2.69 ±0.12 22.7 0.6 47 2.43 0.09 23 33.3 12.7 0.2 71 1.80 0.07 43
IC
50 -11.335 pM >33.3 PM Values are the mean ±standard deviation of N 3 samples (VC: N Abbreviations: Conc, concentration; 4-OH1 TB, 4-hydroxytolbutamide; 4-OH ME, 4hydroxymephenytoin; GIG, chromatographic interference control; VC, vehicle control 1% methanol); NA, not applicable; OMP, omeprazole; SFZ, sulfaphenazole; BQL, below quantifiable limit.
WO 02/044113 WO 02/44113PCT/US01/44603 Table 7: Hepatic microsomal activities of dextromethorphan (CYP2D6) and chlorzoxazone (CYP2E1) in male and female human microsomes incubated with Hhalofenic aid at doses of 0.33 11.3.3. 10, and 33.3 u.M Control/ Dextromethorphan Chlorzoxazone Test Cone DEX Production %6-OH1 CZX% Article 4 1 M) (Pmollmg Inhibitio Production Inhibition protein/min) n (pMollmg protein/min) dCi 33.3 0.00 0.00 NA 0.00 0.00 NA VC 0.1% Ill1±6 0 246 ±5 0 4-MP 100 NA NA BQL -100 QUIN I BQL -100 NA NA H- 0.33 107 ±4 3 238 ±4 3 halofenic 1.0 110 ±2 1 244 ±1 1 acid 3.3 104 ±3 6 239 ±4 3 107 ±1 4 244 ±6 1 33.3 106 ±4 5. 239 ±4 3
IC
50 NA NA Values are the mean standard deviation of N 3 samples (VC: N Abbreviations: Cone, concentration; DEX, dextrorphan; 6-OH4 CZX, 6-hydroxychlorzoxazone; dId, chromatographic interference control; VC, vehicle control 1% methanol); NA, not applicable; 4-MP, 4-methylpyrazole; QUIN, quinidine; BQL, below quantifiable limit.
WO 02/044113 PCT/US01/44603 Table 8 Hepatic microsomal activities of testosterone (CYP3A4) in male and female human microsomes incubated with halofenic acid at doses of 0.33, 1.0, 3.3, 10, and 33.3 itM Control/ Testosterone Test Cone 6P-OHT Production Inhibition Article (pM) (pmol/mg protein/min) CIC 33.3 0.00 0.00 NA VC 0.1% 1843 ±9 0 KTZ 5 32.4 0.2 98.2 0.33 1816 12 halofenic 1.0 1851 14 0 acid 3.3 1810 ±3 1.8 1819+4 1.3 33.3 1816±6 ICso NA Values are the mean standard deviation ofN 3 samples (VC: N Abbreviations: Conc, concentration; 6P-OHT, 6p-hydroxytestosterone; CIC, chromatographic interference control; VC, vehicle control methanol); NA, not applicable; KTZ, ketoconazole; BQL, below quantifiable limit.
EXAMPLE 19 This example illustrates the preparation of various prodrug esters of the enantiomer of CPTA. Representative coupling methods are provided (Methods Method Preparation of the cesium salt of(-)-CPTA by sub-stoichiometric addition of base and reaction with benzyl bromide. Preparation of (-)-benzyl 4-chloroalpha-(3-trifluoromethylphenoxy)benzeneacetate To 100.2 mg (0.303 mmol) of(-)-CPTA was added 0.82 ml MeOH and 0.2 ml H 2 0. The solution was cooled (0-5 0 C) and 0.148 mmol of Cs 2
CO
3 (0.296 mmol available base) from a cooled 20% stock solution was added. The resulting pH was between 5.5 and 6. The cooling was removed and the mixture stirred for 15 min. The WO 02/044113 PCT/US01/44603 MeOH/H 2 0 was stripped and 2ml dry benzene was added and thoroughly stripped at 0 C. Another 2 ml benzene was added and again evaporated at 45 oC. Anhydrous benzene (0.5 ml) and anhydrous DMF (0.5 ml) were added to the cesium salt CPTA, the solution was cooled to 0 °C and benzyl bromide (dissolved in 2001l of anhydrous 50/50 benzene/DMF and cooled) was added dropwise. After 5-10 min the ice cooling was removed and the flask allowed to warm to room temperature. After 6 h the reaction was complete (LC/MS). Methyl chloroform (8 ml) was added and washed twice with ice cold 0.2 N NaHCO 3 The organic layer was then washed with cold 0.1 N HC1, dried (Na 2
SO
4 and stripped to yield (-)-benzyl 4-chloro-alpha-(3-trifluoromethylphenoxy)benzeneacetate as a clear viscous oil (108 mg, 85% yield). Chiral analysis (Ultron ES-OVM reversed phase albumin protein column): 98% ee. LC/MS purity: 100%.
Method 2: HATU-mediated coupling. Preparation of (-)-2-ethoxycarbonylaminoethyl 4-chloro-alpha-(3trifluoromethylphenoxy)benzeneacetate To a cooled (0-5 solution of (-)-4-chloro-alpha-(3trifluoromethylphenoxy)benzeneacetic acid (0.100 g, 0.302 mmol) and ethyl N-(2hydroxyethyl)carbamate (44 mg, 0.333 mmol) in methylene chloride was added pyridine (36 mg, 0.454 mmol) from a 25% stock solution in methylene chloride. HATU (115 mg, 0.302 mmol) was added in one portion and after 20 min the suspension was allowed to warm to room temperature. The reaction was monitored by LC/MS and was complete after 3.5 h. The product mix was partitioned between methylene chloride 5 ml) and 0.2 M NaHCO 3 (5 ml). The aqueous was extracted a total of three times with methylene chloride. The organic extracts were combined, washed with 2 M HCI (3X5 ml), dried (Na 2
SO
4 and evaporated to yield (-)-2-ethoxycarbonylaminoethyl 4-chloro-alpha-(3trifluoromethylphenoxy)benzeneacetate as a clear viscous oil (104 mg, 77% yield).
Chiral analysis (Ultron ES-OVM reversed phase albumin protein column): 97% ee, LC/MS purity: 97%.
Method 3: p-Toluenesulfonic acid catalyzed ester formation. Preparation methoxyethyl 4-chloro-alpha-(3-trifluoromethylphenoxy)benzeneacetate WO 02/044113 PCT/US01/44603 To 100 mg (0.302 mmol) of(-)-CPTA was added 150 mg (1.97 mmol) of methoxyethanol, 4 ml benzene and 16.1mg of p-toluenesulfonic acid. The solution was refluxed for 3 h with use of a Dean-Stark trap for removal of the benzene-water azeotrope. The reaction was cooled, evaporated under reduced pressure and partitioned between methylene chloride (5 ml) and 0.2M Na2CO 3 (5 ml). The aqueous was extracted a total of three times with methylene chloride. The combined organic extracts were dried (Na 2
SO
4 and evaporated to yield (-)-2-methoxyethyl 4-chloro-alpha-(3trifluoromethylphenoxy)benzeneacetate as a clear viscous oil (96 mg, 82% yield). Chiral analysis (Ultron ES-OVM reversed phase albumin protein column): 98% ee, LC/MS purity: Method 4: Conversion of (-)-CPTA to acid chloride via oxalyl chloride/DMF and reaction with alcohols. Preparation of (-)-propargyl 4-chloro-alpha-(3trifluoromethylphenoxy)benzeneacetate.
(-)-CPTA (100 mg, 0.302 mmol) was added to a white suspension prepared by the dropwise addition of oxalyl chloride (43 mg, 0.336 mmol) to DMF (33 mg, 0.453 mmol) in 1 ml of a 60/40 acetonitrile/methylene chloride. The reaction was stirred for 30 min at 0 OC to complete the formation of acid chloride. A solution of propargyl alcohol (18.5 mg, 0.330 mmol) and pyridine (52.4 mg, 0.663 mmol) in 200 pJ dry methylene chloride was added dropwise to the cooled solution of the acid chloride.
The temperature was maintained at 0°C for the first hour and then allowed to warm to ambient. After 3 h the reaction was complete (monitored by LC/MS). The reaction mixture was partitioned between methyl chloroform and 0.2 M Na 2
CO
3 The organic phase was washed three times with aqueous base. The organic was then washed once with 1M HC1, dried (MgSO 4 and stripped to yield (-)-propargyl 4-chloro-alpha-(3trifluoromethylphcnoxy)benzeneacetate as a clear viscous oil (96 mg, 86% yield). Chiral analysis on a Shinwa Chemical Ultron ES-OVM (reversed phase albumin protein column): 98% ee, LC/MS purity: 100%.
Method 5: In situ generation of the silver salt of(-)-CPTA by substoichiometric addition ofAg 2 CO3 and reaction with 1-chloroethyl cyclohexyl WO 02/044113 PCT/US01/44603 carbonate. Preparation of -(cyclohexyloxycarbonyloxy)ethyl 4-chloroalpha-(3-trifluoromethylphenoxy)benzeneacetate To a cooled (0-5 oC) solution of(-)-4-chloro-alpha-(3trifluoromethylphenoxy)benzeneacetic acid (0.200 g, 0.605 mmol) in 2 ml acetonitrile was added 1-chloroethyl cyclohexyl carbonate (0.105 g, 0.544 mmol). Ag 2
CO
3 (0.067 g, 0.242 mmol) was then added in one portion. The ice bath was removed and after 6 h at room temperature the reaction was complete. The reaction mixture was partitioned between methylene chloride and 0.2 M NaHCO 3 The organic layer was washed a total of three times with bicarbonate solution and then once with 0.01M HC1, dried (Na 2 SO4) and evaporated to yield 0.126 g -(cyclohexyloxycarbonyloxy)ethyl 4chloro-alpha-(3-trifluoromethylphenoxy)benzeneacetate. Chiral analysis (Ultron ES- OVM reversed phase albumin protein column): 98% ee, LC/MS purity: 100%.
Table 9 provides structures of additional prodrug compounds, along with characterization data and reference to the methods employed for preparation the source of starting materials.
Table 9 Illustrative compounds, source of reagents and coupling methods Coupling Compound Reagent and Structural elucidation Method and number Source (vida Reference: infra): cF 3 ,/Br [ao] 24 D- 55.03 (c 5.23, CHCI 3 Method 1 Aldrich 'HNMR (400MHz, CHC13): Aldrich 57.0-7.5 (8H, m),5.8 (1H, m), o 5.6 (1H, 5.2 (2H, dd), 4.6 (2H, m).
19.1 LCMS, 369, 99% 98%ee WO 02/044113 WO 02/44113PCT/USOI/44603 1
CF
3
S
0 N 0 CI I H 1 19.2 0
N
U.S. Patent No.
5,506,224
I
[a]j 24 D- 27.16 (c 6.99, Gil) 'HNNvR (400MHz, CHCl 3 87.0-7.5 (I11H, m),5.9 (I1H, in), 4.3 (2H, in), 3.6(2H, in).
LCMS, 482, 99% 98%ee I Method 2
<OH
Aldrich [cx] 24 D- 44.46 (c 5.90, CHCl,) 'HNMR (400MHz, CHCl 3 87.0-7.5 (13H, mn), 5.6 (111, s), 4.7 (211, dd), 2.4(111, s).
LCMS, 368, 99% 98%ee Method 4 19.3 0 0 K Aldrich
[CC]
24 D- 35.40 (c 12.6, Cl-Id 3 87.0-7.5 (13H, mn), 6.0 (111, in), 5. 6 (1IH, 4.3 (214, mn), 3.6 (2H, mn).
LCMS, 477, 99% 97.4%ee 19.4 -s 0 C '-A 0 Maybridge 19.5 [lCJ 24 D- 57.39 (c 10.3, CHCl 3 'HNMR (400MHz, CHCl 3 87.1-7.5 (8H, mn), 5.7 (111, s), 4.9 (1IH, 4.6 (11H, 3.6 (8H, in).
LCMS, 457, 99% 97.4%e 24 D- 35.38 (c 7.36, CHC1 3 1 LINMR (400MHz, CHC1 3 87.0-7.5 (13H, mn), 5.6 (lIH, s), 5.1 (2H, dd).
LCMS, 421, 99% 98%ee Method I WO 021044113 WO 02144113PCTIUS01144603 Aldrich 24 D- 56.34 (c 5.32, CHC1 3 'HNMR (400MHz, CHC1 3 57.0-7.5 (8H, in), 5.6 (1H, s), 4.2 (211, mn), 1.1 (3H, t).
LCMS, 358, 98% 98%ee Method 3 19.7 4 i 1- CF6 19.8 rOH 1,110 Aldrich [a] 24 D- 54.13 (c 8.40, CHC1 3 'HNMR (400MHz, CHC1 3 87.0-7.5 (8H, in), 5.6 (IH, s), 4.4 (111, in), 4.2(IH, in), (2H, in), 3.3 (3H, s).
LCMS, 445, 97% 97.7%ee Method 3 19.9 0 [cX] 24 D- 43.30 (c 6.61, CHC1 3 Method 2 Ho- N o 1 HNMR (400MHz,
CHCI
3 67.0-7.5 (8H1, in), 5.6 (11H, s), (1H1, mn), 4.2 (2H, in), U.S. Patent No. (2Hl, in), 3.3 (2H1, in), 1.1 (3H, 5,506,224 in).
LCMS, 445, 97% 96.8%ee 01 [cX] 24 D- 42.33 (c 7.30, CHCl 3 Method 2 N 1 'HNMR (400MHz, CHG1 3 HO0 57.0-7.5 (811, mn), 5.6 (IH, s), U.S. Patent No. 4.3 (2H, in), 4.1 (1 H, in), 3.3 5,506,224 (2H, in), 2.8 (3H1, s).
LCMS, 451, 97% 96.5%ee 19.10 0 C1 24 D- 57.87 (c 2.57, CHC1 3 'HN'MR (400MHz, CHC1 3 57.0-7.5 (8H1, in), 5.6 (1H, s), 4.9 (1IH, 4.6(111, 2.9 (311, 2.8 (3H, s).
LCMS, 415, 98% 97. 1%ee Method I Aldrich 11 19.11 WO 02/044113 WO 02/44113PCT/US01/44603
[CC]
24 D- 62.27 (c 5.35, CflC1 3 'HNMR (400MHz, CHC1 3 67.0-7.5 (8H, in), 5.6 (1H, s), 4.2 (2H, in), 1.1 (3H, t).
LCMS, 416, 99% 98%ee Method 1 19.12 0 24 D- 49.85 (c 11.3, CHCI 3 Method I CI-IA 'HNMR (400MHz, CHC1 3 63 7.0-7.5 (8H, in), 5.7 (1H, s), U.S. Patent No. 4.6 (2H, 4.4 (2H, 3,998,808 (4H, mn), 2.3 (2H, mn).
LCMS, 427, 96% 96. 1%ee 19.13 rOH
OH
Aldrich 4 D- 51.17 (c 6.76, CHC1 3 'HNMR (400IVIz, CHC1 3 67.0-7.5 (8H, in), 5.6 (1 H, s), 5.3 (1 H, 4.3 (2H, in), 3.7 (2H, in).
LCMS, 374, 98% 95.9%ee Method 3 19.14
I
[cC] 24 D- 40.97 (c 8.18, CHCI 3 'HNMR (400MHz, CHC1 2 67.0-7.5 (8H, in), 5.6 (1H, s), 5.3 (1 H, in), 4.1 (2H, in), 3.4 (2H, in), 2.1 (1H1, in), 1.0 (3H, d) LCMS, 443, 99% 97. 1%ee Method 2 U.S. Patent No.
5,506,224 19.15 I I WO 02/044113 WO 02/44113PCT/US01/44603 0
NH
Synthesis (1991) 571 19.16 2 111.2 (c 10.8, CHC1 3 'LINNiR (400MHz, CHC1 3 67.0-7.5 (8H1, in), 6.7 (IH, m) 5.6 (1IH, 5.3 (11H, in), 4.9 (I1H, 4.4 (11H, in), 4.4 (1 H, 3.4 (11H, in), 3.3 (11H, in), 2.4 (1H, in), 2.2 (111, in), LCMS, 484, 99% 96%ee
I
Method 1
HO
0
N-
H
US Pat 5,506,224 24 D- 36.72 (c 8.90, CHC1 3 'HNMR (400MHz, CHC1 3 66.8-7.5 (8H, mn), 6.5 (11, in), 5.6 (1H, 4.3 (2H1, mn), 3.8 (2H1, 3.5 (21-1, in), 3.4 (3H, s).
LCMS, 445, 99% 98%ee Method 2 19.17 HO0 N Aldrich
[(XI]
24 D-21.23 (c 13.2, CHCI 3 'HNIVR (400MHz, CHC1 3 6 8.6 (1H, cr), 7.8 (111, 7.6 (10H1, mn), 5.6 (IH, 5.2 (2H, s).
LCMS, 421, 99% 97%ee Method 2 19.18
CCF
19.19 [a] 24 D- 108.3 (c 9.36, CHC1 3 1 11Nvfl (400MHz, CHCI 3 57.0-7.5 (811, in), 5.8 (1H1, s), (2H1, 4.5 (11H, in), (2H, 3.7 (311, 3.4-3.7 (2H, mn), 1.8-2.3 (4H, mn).
LCMS, 499, 99% 98.3%ee
I
Method 1 J.Med.Chein (1997) 3594 4 1 WO 02/044113 PCT/US01/44603 CF3 0 0 0" 0 'N0
H
N
19.20
HO
N 0
H
N
U.S. Patent No.
5,506,224
CF
3 01 0 C1
H
19.21 Aldrich [a] 2 D- 32.05 (c 6.00, CHC1 3 'HNMR (400MHz, CHC1 3 6 9.4 (1 H, br. 8.7 (1H, br.
8.4 (1H, br. 7.7 (1l, br.s 7.0-7.5 (8H, 5.8 (1H, s), 4.4(2H, 3.7 (2H, i).
LCMS, 479, 99% 97.6%ee [a] 2 4 D- 28.19 (c 14.3, CHC1 3 'HNMR (400M1-Iz, CHC1 3 67.0-7.5 (13H, 5.6 (1H, s), 5.1 (2H, 4.6 (1H, 4.2 (2H, 3.4 (2H, m).
LCMS, 507, 99% 98%ee [a] 24 D- 39.80 (c 5.90, CHC1 3 1 HNMR (400MHz, CHC1 3 87.0-7.5 (8H, 5.6 (lH, s), 4.2 (2H, 4.0 (1H, 3.4 (2H, 3.0 (2H, 1.0 (3H, t0.
LCMS, 444, 99% 98%ee [a] 2 4 D- 44.75 (c 7.60, CHC1 3 'HNMR (400MHz, CHCI 3 87.0-7.5 (8H, 5.6 (11, s), 4.1-4.4 (411, 1.9 (311, s).
LCMS, 416, 99% 98.36%ee Method 2 Method 2 Chem.Soc.Rev (1975) 231 Method 2 19.22
CF
3 01 0 19.23 Method 4 Aldrich 4 [c] 2 4 D- 35.63 (c 5.30, CHC1 3 'INMR (400MHz, CHC1 3 87.0-7.5 (8H, 5.6 (1H, s), 4.9(2H, dd), 2.1 (3H, s).
LCMS, 442, 99% 98.3%ee Method 1 WO 02/044113 PCT/US01/44603 24 D- 38.42 (c 8.78, CudC1 3 Method 2 OH 'HNMR (400MHz,
CHCI
3 56.9-7.7 (13H, 5.8 (1H, s).
Aldrich LCMS, 406, 99% 99%ee 19.25 N 0
H
N
Aldrich [cX] 24 D- 28.50 (c 8.40, CHC 3 'HNIR (400MHz, CHC1 3 8.8(211, 7.5 (2H, 7.4 (8H, in), 6.3 (11H, 5.6 (11, 4.4 (2H, br. 3.8 (2H, br. s).
LCMS, 478, 99% 97%ee Method 2 19.26
CP
3 CK;OyO [OC] 2 4 D- 31.48 (c 5.00, CHCl 3 Method 0 I 'HNMR (400MHz, CHC1 3 ySyn. (1986) 627 57.0-7.5 (811, m),6.8 (11, in), K. 0 00-10 5.6 (1H, 4.5 (1H, 1.2- 19.27 2.0 (13H, m).
LCMS, 500, 99% 99%ee CFI, 0 24 D- 29.52 (c 2.04, CHCI 3 Method 2 no HNH 2 'HNVIR (400MHz, CHC1 3 A -I Nil, Sigma 87.0-7.5 (8H, 5.6 (11, s), 4.2-4.5 (4H, 3.7 (2H, i).
19.28 LCMS, 431, 99% 96.4%ee 0,6 O 0 a]2D- 2.83(c 5.42,C{C1 3 Method2 'HNMR (400MHz,
CHC
3 07.0-7.5 (8H, 5.6 (1H, s), 0 S4.4 (IH, 3.2-4.0 (2H, i), CI NH Sigm1.6-2.2 (4H, m).
LCMS, 413, 98% 19.29 WO 02/044113 PCT/US01/44603 EXAMPLE This example illustrates methodology and conditions for the chiral analysis of prodrug esters of (-)-CPTA.
Since the chiral center of the prodrug esters is adjacent to the carbonyl of the acid, the esters were analyzed to confirm that the optical center had not racemized during the coupling. Both MBX102 and the MBX102 acid have been resolved on a normal phase chiral column, but confirmation of the relative retentions of the enantiomeric esters on such a column would require standards of both enantiomers for all the compounds involved. Alternatively, the esters could be hydrolyzed to the known acids and the ratio of the enantiomeric acids could be analyzed, but some degree of racemization is likely under these conditions. Reverse phase chiral columns offer the ability to be interfaced with an LC-MS system, so that the enantiomers could be positively identified without separate standards. Under negative ion, selected ion monitoring (SIM) conditions, an LC-MS would be expected to be an order of magnitude more sensitive than UV detection as well as being much more specific. The application of chiral-column-LC-MS has proven to be an excellent method for characterizing the optical purity of this series of compounds. The quantitation limit has been shown to be well below the 2.5% limit set for the alternate optical isomer, and the extremely low level of detection gives more flexibility in the separation of the series, as well as establishing the relative retentions of the enantiomeric pairs in most cases without the need to produce racemized material.
The enantiomers of the prodrug esters synthesized were well separated using one of the columns and solvent systems listed below. Most of the enantiomers were separated using the reverse phase ES-OVM column with the mass spectrometer as a detector. Since this gives the added confirmation of the molecular species and isotope ratio, this was the preferred technique. For enantiomeric pairs which were not separated on the reverse phase column, a normal phase system was used and some of the sample was partly racemized to establish the retention time of the enantiomers.
Description of Analytical Conditions: Reversed Phase separation: Column: Shinwa Chemicals Ultron ES-OVM 4.6 x 150mm with matching pre column (part 712111630). Available from Mac Mod Analytical.
WO 02/044113 PCT/US01/44603 Solvents: Acetonitrile, Ethanol or Methanol. Water containing molar NH4 OCOCH3 (Ammonium Acetate) adjusted to pH 4.6 with acetic acid.
Solvent Flow: 1 milliliter per minute Isocratic as indicated.
Detector: UV for method development at 220 or 270 nm or as specified. LC- MS using negative ion electrospray, monitoring two ions, M-H- and the chlorine 37 isotope Mass Spectrometer: Waters Micromass ZMD Bench top LC-MS with Electrospray source.
HPLC: Agilent 1050 or Shimadzu LC-10 as indicated.
Software: HP 3396 integrator for method development and Micromass MassLynx V3.4 for quantitation.
Methods Development: Most prodrug esters separated using between 10 and 40% acetonitrile in water with an ammonium acetate buffer at pH 4.6. The free acid was a contaminant in the crude or racemized samples. The retention times of the acids were pH dependant, but the chromatography of the esters was in general not sensitive to pH. Retention and peak shape were affected by the concentration and injection solvent, and the samples were injected using a minimum concentration and the column solvent mixture for best results.
The desired enantiomer was retained less and with a retention time of approximately minutes, the second enantiomer eluted from 1 to 3 minutes later with complete separation in most cases. There was some peak tailing from the main enantiomer in some cases.
The amount of acetonitrile was adjusted to give a retention time of about 5 minutes for the main component or best peak separation with maximum peak sharpness. Peaks retained more than 10 minutes tended to be too broad to accurately quantify the second enantiomer at levels under 2%.
Detection Limit: Sample 1061-18-05 the benzyl ester, MW 420) was mixed with racemized material to give an expected concentration of 2.47% of the unwanted enantiomer. The sample was analyzed repeatedly by LC-MS (monitoring ions at 419 and 421) giving an average of 2.6% recovery of the second enantiomer with a 10.9% RSD.
See Figure 18.
WO 02/044113 PCT/US01/44603 Normal Phase Separation: Column: Analytical Column: Regis WHELK-0 4.6 x 250mm with Peek Scientific Cyano pre column (part DC5-CN). Preparative Column Regis WHELK-0 20 x 250mm with Cyano pre column.
Solvents: 10% of Solvent B in Hexane.
Isopropanol (IPA), Ethanol or Ethyl Acetate buffered with ammonium acetate or triethylamine when indicated.
Solvent Flow: 1.2, 1.5 or 25 milliliter per minute Isocratic as indicated.
Detector: UV for method development at 270 nm or as specified.
HPLC: Agilent 1050, Gilson 321 or Shimadzu LC-10 as indicated.
Software: Gilson Unipoint, HP 3396 Integrator or Micromass MassLynx V 3.4 as indicated.
Methods Development: Most prodrug esters separated using between 10 and 40% alcohol in Hexane without a modifier. The desired enantiomer was retained less. With a retention time of approximately 5 minutes for the first enantiomer, the second enantiomer eluted from 1 to 3 minutes later with complete separation. The compounds with a basic amine group required ammonium acetate as a modifier to sharpen the peak shape.
Triethyl amine was tried but was not effective as a modifier. IPA was normally the first solvent tried, if the separations were not complete, ethanol or ethylacetate were used.
The enantiomeric excess for each compound was measured using reverse phase chromatography when possible, but in several cases, separation was only possible using normal phase. In these cases, the data were collected and integrated using the MicroMass MassLynx software to be consistent with the other analyses (Figure 19).
EXAMPLE 21 This example illustrates the plasma hydrolysis of various prodrug esters of the (S)-enantiomer of CPTA at physiological conditions. Hydrolysis was monitored and analyzed by HPLC. The hydrolytic product was identified by comparison to authentic CPTA and the hydrolytic rates were calculated.
WO 02/044113 PCT/US01/44603 CF3 C OR 0 CI H Prodrug esters CPTA Human plasma (heparinized, pooled) was obtained from Golden West Biololgicals Inc., USA; Plasma incubation was carried out in a Water Bath Shaker (New Brunswick Scientific, Inc.); Sample analyses were carried out on Agilent HPLC 1100 system.
General hydrolytic procedure: Prepare 31.25 or 62.5 mM prodrug stock solutions in 100% DMSO. Add 4 L of 31.25 or 62.5 mM prodrug stock solutions to 1.0 mL of plasma in a microcentrifuge tube to make plasma concentration at 125 or 250 gM, mix gently.
Aliquots of 50 pL are transferred to 15 microcentrifuge vials. Three samples are immediately removed to a freezer at about -80'C (0 time point), the remaining samples are incubated in a bath shaker at 37°C. Three samples are removed at 30 min, 2, 7 and 24 hr, and stored at -80 0 C until analysis. The relative concentrations of MBX-compounds are determined by HPLC methods. If there are enough data points, the hydrolytic rate can be calculated using WinNonlin software. Table 10 provides a summary of plasma halflives of some pro-drug esters and Figures 20A-20G illustrate the hydrolytic curves of those prodrug esters.
HPLC: Agilent 1100 HPLC System Column: Phenomenex Luna C18(2) 5u 150 x 2 mm lot #105554-2 Phenomenex Luna C18(2) 5u 250 x 4.6 mm lot #103992-8 Solvent Flow Rate: 0.25 or 1 ml minute Injection Volume: 20 or 40 gl1 Run Time: 7 to 15 min Solvent Composition: 45 to 76%ACN/0.1% TFA in Water (V/V) Detection: Diode Array UV-Visible Detector at 220 mn WO 02/044113 WO 02/44113PCT[USOI/44603 Table Plasma Half-Life of CPTA Prodrugs Compound number Plasma Tj 12 (hours) 19.1 19.2 1 19.3 1 19.4 1 19.5 19.6 6.3 19.7 7 19.8 2 19.9 1 19.10 1 19.11 19.12 19.13 19.14 2 19.15 1 19.16 19.17 1 19.18 1 19.19 19.20 19.21 2 19.22 1 19.23 1 19.24 19.25 19.26 1 19.27 <1I WO 02/044113 PCT/US01/44603 19.28 Although the foregoing invention has been described in detail for purposes of clarity of understanding, it will be obvious that certain modifications can be practiced within the scope of the appended claims. All publications and patent documents cited herein are hereby incorporated by reference in their entirety for all purposes to the same extent as if each were so individually denoted.

Claims (71)

1. A method of treating Type 2 diabetes in a mammal, comprising: administering to said mammal a therapeutically effective amount of the stereoisomer of a compound of Formula I, X R X 2 X4 X 3 (I) wherein: R is a member selected from the group consisting of a hydroxy, alkoxy, heteroalkoxy, aryloxy, heteroaryloxy, lower aralkoxy, di-lower alkylamino-lower alkoxy, lower alkanamido lower alkoxy, benzamido-lower alkoxy, ureido-lower alkoxy, N'-lower alkyl-ureido-lower alkoxy, carbamoyl-lower alkoxy, halophenoxy substituted lower alkoxy, carbamoyl substituted phenoxy, carbonyl-lower alkylamino, N,N-di-lower alkylamino-lower alkylamino, halo substituted lower alkylamino, hydroxy substituted lower alkylamino, lower alkanoyloxy substituted lower alkylamino, ureido, and lower alkoxycarbonylamino; and X' is a halogen; and X 2 X 3 and X 4 are each independently selected from hydrogen and halogen with the proviso that no more than two of X 2 X 3 and X 4 are hydrogen; or a pharmaceutically acceptable salt thereof, wherein the compound is substantially free of its stereoisomer.
2. The method of claim 1, wherein the compound is a compound of Formula II, R 2 0 x o x 2 X 4 X 3 (II) wherein: R 2 is a member selected from the group consisting of: CI-C 5 alkyl, C 1 -Cs-cyclic alkyl, C 2 -C5 alkenyl, and C 2 -C 5 alkynyl, wherein the groups are optionally substituted with one or more halogen atoms; phenyl, naphthyl and pyridyl, wherein the groups are optionally substituted with one or more substituents selected from the group consisting of [R:\LIBH]05820.doc:UG halo, C 1 -C 4 alkyl, C 1 -C 4 alkoxy, -NO 2 -S(O),(Ci-Csalkyl), -OH, -NR 3 R 4 -C0 2 R', -CONR 3 R 4 -NR'COR 4 -NR'CONR 3 R 4 and -(CHR 3 )R 4 -R'0R'; -R 7 O 2 CR'NRR R; -R7COR6; -R 7NR COR 6 -R NRR 6; -(CH 2 ),CH(R)(CH 2 )qO 2 CR 9 -(CH 2 ),CH(R 3 )(CH 2 )qNR 4 COR'; -(CH 2 )oCH(R 3 )(CH 2 )qNR 4 CONR 3 R 4 -(CH 2 3 )(CH,)qNR 4 COOR 10; -(CH 2 )oCH(R 3 )(CH 2 )qNR 4 SO 2 R I; -(CHR 3 ),COR 1 2 -(CHR 3 )PNR 3 R 4 -(CHR 3 ),CONR' 3 R1 4 0 0A 12I _f (CHA) R 3 0 R 15 R 16 -N adR1 wherein mis 0to 2,0 and q are 0to 5, p is I to 5, s is I to 3, t is I to 5, uisOto 1, v is I to 3, and w is 1 to (2v and wherein R 3 and R 4 are independently H, C 1 -C5 alkyl, phenyl or benzyl; and wherein R 5 is H, C I-C 5 alkyl or NR 3 R 4 and wherein R 6 is phenyl, naphthyl, pyridyl, imidazolyl, indoxyl, indolizinyl, oxazolyl, thiazolyl, thienyl, pyrimidyl, or 1-pyrazolyl optionally substituted with one or more substituents selected from the group consisting of halo, C 1 -C 4 alkyl, C 1 -C 4 alkoxy, -N0 2 -S(O)m,(Ci-Csalkyl), -OH, -NR 3 R 4 -C0 2 R 5 -CONR 3 R 4 -NR 3 COR 4 -NR 3 CONR 3 R 4 and and wherein R 7 is a C 1 -C 8 saturated or unsaturated, straight-chain, branched or cyclic alkylene or alkylidene group optionally substituted with one or more groups selected from halo, hydroxyl, thiol, amino, monoalkyl amino, dialkyl amino, acylamino, carboxyl, alkylcarboxyl, acyl, aryl, aroyl, aralkyl, cyano, nitro, alkoxy, alkenyloxy, alkylcarbonyloxy and arylcarbonyloxy; and wherein R 8 is a C 1 -C 8 straight-chain or branched alkylene or alkylidene optionally substituted with one or more groups selected from amino, monoalkyl amino, dialkyl amino, acylamino, hydroxyl, thiol, mcthylthiol, carboxyl and phenyl; and wherein R 9 and R1 0 are independently H, C 1 -C 5 alkyl, optionally substituted with one or more groups consisting Of C 1 -C5 alkoxy aryl and heteroaryl, wherein the aryl is phenyl or naphthyl optionally substituted with one or more substituents selected from the group consisting of halo, C 1 -C 4 alkyl, C 1 -C 4 alkoxy, -NO 2 -S(O)m,(Ci-Csalkyl), -OH, -NR 3 R 4 -C0 2 _CONR 3 R 4 -NR 3 COR 4 -NR 3 CONR 3 R 4 and -CFw, and wherein the heteroaryl is pyridyl optionally substituted with one or more substituents selected from the group consisting of halo, C 1 -C 4 alkyl, CI-C 4 alkoxy, -NO 2 (CI-Csalkyl), -OH, -NR 3 R 4 -C0 2 R 5 -CONR 3 R 4 _NR 3 COR 4 -NR'CONR 3 R 4 and and wherein R" is methyl or phenyl, wherein the phenyl is optionally substituted with methyl and/or -NO 2 and wherein R 12is H, C 1 -C 5 alkyl, phenyl, benzyl, naphthyl or pyridyl, wherein the C 1 -C5 alkyl, phenyl, naphthyl, benzyl and pyridyl are [RALIBH1O5820 doc UG optionally substituted with one or more substituents selected from the group consisting of halo, C 1 -C 4 alkyl, C 1 -C 4 alkoxy, -NO 2 -S(O)mn(Ci-Csalkyl), -OH, -NR 3 R 4 -C0 2 R 5 -CONR 3 R 4 -NR'COR 4 -NR'CONR 3 R 4 and -CFw; and wherein R 1 3 and R 1 4 are independently the following: alkyl, alkenyl, aryl, aralkyl or cycloalkyl, wherein the groups are optionally substituted with one or more substituents selected from the group consisting of halo, C 1 -C 4 alkyl, C 1 -C 4 alkoxy, -NO 2 -S(O)m,(CI'-C 5 alkyl), -OH, -NR 3 R 4 -C0 2 -CONR 3 R 4 -NR'COR 4 -NR 3 CONR 3 R 4 -CH 2 NR 3 R 4 00CR 8 and -CF,; and wherein when R' and R' are included as -(CHR 3 )CONR' 3 R, NR 'R'is -N -N-OR -N -N <NR 3 COOR 3 N NN O H R 3 000 R R NOC -N ?or -0N COOR 3 and wherein R 1 5 is CF, or CI-C 5 alkyl, wherein CI-C 5 alkyl is optionally substituted with the following substituents: CI-C 5 alkoxy; phenyl, optionally substituted with one or more substituents selected from the group consisting of halo, C 1 -C 4 alkyl, C 1 C 4 alkoxy, -N0 2 -S(O)m..(CI-Csalkyl), -OH, -NR 3 R, -C0 2 -CONR 3 R 4 -NR'COR 4 -NR'CONR 3 R 4 and benzyl, optionally substituted with one or more substituents selected from the group consisting of halo, CI-C 4 alkyl, C 1 -C 4 alkoxy, -N0 2 -S(O)M(C 1 Csalkyl), -OH, -NR 3 R 4 -C0 2 -CONR 3 R 4 -NR'COR 4 -NR'CONR 3 R 4 and and wherein R 1 6 is H, C 1 -C 5 alkyl or benzyl; and wherein R 17 is C 1 -C5 alkyl, C 3 -C 8 Cyclic alkyl, phenyl or benzyl; and wherein R 1 8 is H, alkyl, aryl, aralkyl or cycloalkyl, where the group is optionally substituted with one or more substituents selected from the group consisting of halo, CI-C 4 alkyl, CI-C 4 alkoxy, -N0 2 -S(O)mn(Ci-Csalkyl), -OH, -NR 3 R 4 -C0 2 R 5 -CONR 3 R 4 -NR'COR 4 -NR 3 CONR 3 R 4 and -CF,.
3. The method of claim 1, wherein the compound is administered by intravenous infusion, transdermal delivery, or oral delivery.
4. The method of claim 1, wherein the amount administered is about 100 mg to about 3000 mg per day. [R-\LIBH]05820 docUG The method of claim 1, wherein the amount administered is about 500 mg to about 1500 mg per day.
6. The method of claim 1, wherein the amount administered is about 5 to about 250 mg per kg per day. s 7. The method of claim 1, wherein the compound is administered together with a pharmaceutically acceptable carrier.
8. The method of claim 1, wherein the compound modulates hyperglycemia by reducing blood glucose levels in the mammal.
9. The method of claim 1, wherein the compound modulates hemoglobin Alc in the mammal. The method of claim 1, wherein the compound modulates a microvascular and macrovascular complication associated with diabetes.
11. The method of claim 10, wherein the microvascular complication is retinopathy, neuropathy or nephropathy.
12. The method of claim 10, wherein the macrovascular complication is cardiovascular disease or peripheral vascular disease.
13. The method of claim 1, wherein the compound modulates atherosclerosis.
14. The method of claim 1, wherein the compound prevents the development of diabetes in a mammal.
15. The method of claim 1, wherein the compound is administered in combination with a compound selected from the group consisting of: a sulfonylurea or other insulin secretogogue, a thiazolidinedione, a fibrate, a HMG-CoA reductase inhibitor, a biguanide, a bile acid binding resin, nicotinic acid, a a-glucosidase inhibitor, and insulin.
16. A method for treating insulin resistance in a mammal, comprising: administering to said mammal a therapeutically effective amount of the stereoisomer of a compound of Formula I, O X R X 2 X 4 X 3 (1) wherein: R is a member selected from the group consisting of a hydroxy, alkoxy, heteroalkoxy, aryloxy, heteroaryloxy, lower aralkoxy, di-lower alkylamino-lower alkoxy, lower alkanamido lower alkoxy, benzamido-lower alkoxy, ureido-lower alkoxy, N'-lower (R:\LIBH]05820.doc:UG alkyl-ureido-lower alkoxy, carbamoyl-lower alkoxy, halophenoxy substituted lower alkoxy, carbamoyl substituted phenoxy, carbonyl-lower alkylamino, N,N-di-lower alkylamino-lower alkylamino, halo substituted lower alkylamino, hydroxy substituted lower alkylamino, lower alkanoyloxy substituted lower alkylamino, ureido, and lower alkoxycarbonylamino; and X' is a halogen; and X 2 X 3 and X 4 are each independently selected from hydrogen and halogen with the proviso that no more than two of X 2 X 3 and X 4 are hydrogen; or a pharmaceutically acceptable salt thereof, wherein the compound is substantially free of its stereoisomer.
17. The method of claim 16, wherein the compound is a compound of Formula 11, 0 1 0 0 )X 4 X 3 wherein: R 2 is a member selected from the group consisting of: CI-C 5 alkyl, C 1 -C 8 -cyclic alkyl, C 2 -C 5 alkenyl, and C 2 -C 5 alkynyl, wherein the groups are optionally substituted with one or more halogen atoms; phenyl, naphthyl and pyridyl, wherein the groups are optionally substituted with one or more substituents selected from the group consisting of halo, CI-C 4 alkyl, C 1 -C 4 alkoxy, -NO 2 -S(O)mn(CI-Csalkyl), -OH, -NR 3 R 4 -C0 2 R', -CONR 3 R 4 -NR'COR 4 -NR'CONR 3 R 4 and -(CHR 3 )R 4 -R 7 0R'; -R 7 O 2 CR'NR 3 R 4 -R 7 COR'; -R'NR'COR'; -R'NR 3 R 6 -(CH 2 ).CH(R 3 )(CH 2 )qO 2 CR 9 -(CH 2 3 )(CH 2 )qNR 4 COR'; -(CH 2 )oCH(R 3 )(CH 2 )q NR 4 CONR 3 R 4 -(CH 2 ),CH(R 3 )(CH 2 )qNR 4 COOR' 0 -(CH 2 )oCH(R 3 )(CH 2 )qNR 4 SO 2 R 11 -(CHR 3 )pCO 2 R 12 -(CHR 3 )PNR 3 R 4 -(CHR 3 ),CONR3 3 R1 4 0 \A 1( H1 2 )t (C H 2 R 3 Q 1 2 R15 R 16 -N 1 )and O wherein m is 0 to 2,0o and q are 0 to 5, p is 1ito 5, s is I to 3, t is I to 5, u isO0 to 1, v is I to 3, and w is 1 to (2v and wherein R 3 and R 4 are independently H, CI-C 5 alkyl, phenyl or benzyl; and wherein R 5 is H, C 1 -C 5 alkyl or NR 3 R 4 and wherein R 6 is phenyl, I R:LI BH)05820.docUG naphthyl, pyridyl, imidazolyl, indoxyl, indolizinyl, oxazolyl, thiazolyl, thienyl, pyrimidyl, or 1-pyrazolyl optionally substituted with one or more substituents selected from the group consisting of halo, CI-C 4 alkyl, CI-C 4 alkoxy, -NO 2 -S(O)m(CI-Csalkyl), -OH, -NRR 4 -C0 2 R 5 -CONRR 4 -NR 3 COR 4 -NR 3 CONR 3 R 4 and and wherein R 7 s is a CI-Cs saturated or unsaturated, straight-chain, branched or cyclic alkylene or alkylidene group optionally substituted with one or more groups selected from halo, hydroxyl, thiol, amino, monoalkyl amino, dialkyl amino, acylamino, carboxyl, alkylcarboxyl, acyl, aryl, aroyl, aralkyl, cyano, nitro, alkoxy, alkenyloxy, alkylcarbonyloxy and arylcarbonyloxy; and wherein R is a CI-Cs straight-chain or to branched alkylene or alkylidene optionally substituted with one or more groups selected from amino, monoalkyl amino, dialkyl amino, acylamino, hydroxyl, thiol, methylthiol, carboxyl and phenyl; and wherein R 9 and R' 0 are independently H, C 1 -Cs alkyl, optionally substituted with one or more groups consisting of C,-Cs alkoxy aryl and heteroaryl, wherein the aryl is phenyl or naphthyl optionally substituted with one or more substituents selected from the group consisting of halo, C,-C 4 alkyl, CI-C 4 alkoxy, -NO 2 -S(O)m(CI-Csalkyl), -OH, -NR 3 R 4 -C0 2 -CONR 3 R 4 -NR 3 COR 4 -NR 3 CONR 3 R 4 and and wherein the heteroaryl is pyridyl optionally substituted with one or more substituents selected from the group consisting of halo, C,-C 4 alkyl, C,-C 4 alkoxy, -NO 2 -S(O)m(Ci-Csalkyl), -OH, -NR 3 R 4 -C0 2 R, -CONRR 4 -NR 3 COR 4 -NR 3 CONR 3 R 4 and and wherein R" is methyl or phenyl, wherein the phenyl is optionally substituted with methyl and/or -NO 2 and wherein R12 is H, C,-Cs alkyl, phenyl, benzyl, naphthyl or pyridyl, wherein the CI-C 5 alkyl, phenyl, naphthyl, benzyl and pyridyl are optionally substituted with one or more substituents selected from the group consisting of halo, C 1 -C 4 alkyl, C,-C 4 alkoxy, -NO 2 -S(O)m(Ci-Csalkyl), -OH, -NRR 4 -C0 2 Rs, -CONRR 4 -NR 3 COR 4 -NRCONRR 4 and and wherein R' 3 and R' 4 are independently the following: alkyl, alkenyl, aryl, aralkyl or cycloalkyl, wherein the groups are optionally substituted with one or more substituents selected from the group consisting of halo, CI-C 4 alkyl, C,-C 4 alkoxy, -NO 2 -S(O)m(Ci-C 5 alkyl), -OH, -NRR 4 -C0 2 R, -CONRR 4 -NR 3 COR 4 -NR 3 CONR 3 R 4 -CH 2 NRR 4 00CR' 8 and -CF,; and wherein when R13 and R14 are included as -(CHR 3 )CONR' R 4, NR IR4 is 3 -N 0 -N OR 3 -N NR 3 -N NR 3 COOR 3 COOR [R:\LIBHOSZ05820.doc UG NP N NN OH R 3 00C R 4 R 3 NOC -N or -N COOR 3 and wherein R 1 5 is CF, or CI-C 5 alkyl, wherein CI-C 5 alkyl is optionally substituted with the following substituents: CI-C 5 alkoxy; phenyl, optionally substituted with one or more substituents selected from the group consisting of halo, Ci-C 4 alkyl, CI- C 4 alkoxy, -NO 2 -S(O)m(Ci-Csalkyl), -OH, -NR 3 R 4 -CO 2 R 5 -CONRR 4 -NR 3 COR 4 -NR 3 CONR 3 R 4 and -CvFw; benzyl, optionally substituted with one or more substituents selected from the group consisting of halo, C 1 -C 4 alkyl, CI-C 4 alkoxy, -NO 2 -S(O)m(Ci- Csalkyl), -OH, -NR 3 R 4 -C0 2 R 5 -CONR 3 R 4 -NR 3 COR 4 -NR 3 CONR 3 R 4 and -CyFw; o0 and wherein R 1 6 is H, CI-C 5 alkyl or benzyl; and wherein R' 7 is C 1 -C 5 alkyl, C 3 -C 8 cyclic alkyl, phenyl or benzyl; and wherein R 1 8 is H, alkyl, aryl, aralkyl or cycloalkyl, where the group is optionally substituted with one or more substituents selected from the group consisting of halo, Ci-C 4 alkyl, Ci-C 4 alkoxy, -NO 2 -S(O)m(Ci-Csalkyl), -OH, -NR 3 R 4 -C0 2 R 5 -CONR 3 R 4 -NR 3 COR 4 -NR 3 CONR 3 R 4 and -CFw.
18. The method of claim 16, wherein the compound is administered by intravenous infusion, transdermal delivery, or oral delivery.
19. The method of claim 16, wherein the amount administered is about 100 mg to about 3000 mg per day. The method of claim 16, wherein the amount administered is about 500 mg to about 1500 mg per day.
21. The method of claim 16, wherein the amount administered is about 5 to about 250 mg per kg per day.
22. The method of claim 16, wherein the compound is administered together with a pharmaceutically acceptable carrier.
23. The method of claim 16, wherein the compound prevents the development of insulin resistance in a mammal.
24. The method of claim 16, wherein the compound modulates polycystic ovarian syndrome. The method of claim 16, wherein the compound modulates Impaired Glucose Tolerance.
26. The method of claim 16, wherein the compound modulates obesity. R:LIBH]05820.doc:UG
27. The method of claim 16, wherein the compound modulates gestational diabetes.
28. The method of claim 16, wherein the compound modulates Syndrome X.
29. The method of claim 16, wherein the compound modulates atherosclerosis.
30. The method of claim 16, wherein the compound is administered in combination with a compound selected from the group consisting of: a sulfonylurea or other insulin secretogogue, a thiazolidinedione, a fibrate, a HMG-CoA reductase inhibitor, a biguanide, a bile acid binding resin, nicotinic acid, a a-glucosidase inhibitor, and insulin.
31. A pharmaceutical composition when used for treating insulin resistance, type 2 diabetes, or hyperuricemia, said composition comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of the stereoisomer of a compound of Formula I, XR x 2 X 4 X 3 (I) wherein: R is a member selected from the group consisting of a hydroxy, alkoxy, heteroalkoxy, aryloxy, heteroaryloxy, lower aralkoxy, di-lower alkylamino-lower alkoxy, lower alkanamido lower alkoxy, benzamido-lower alkoxy, ureido-lower alkoxy, N'-lower alkyl-ureido-lower alkoxy, carbamoyl-lower alkoxy, halophenoxy substituted lower alkoxy, carbamoyl substituted phenoxy, carbonyl-lower alkylamino, N,N-di-lower alkylamino-lower alkylamino, halo substituted lower alkylamino, hydroxy substituted lower alkylamino, lower alkanoyloxy substituted lower alkylamino, ureido, and lower alkoxycarbonylamino; and X' is a halogen; and X 2 X 3 and X 4 are each independently selected from hydrogen and halogen with the proviso that no more than two of X 2 X 3 and X 4 are hydrogen; or a pharmaceutically acceptable salt thereof, wherein the compound is substantially free of its stereoisomer.
32. The pharmaceutical composition when used according to claim 31, wherein the pharmaceutical composition treats Type 2 diabetes. [R\I.IBH)05820.doc:LUG
33. The pharmaceutical composition when used according to claim 31, wherein the pharmaceutical composition treats insulin resistance.
34. The pharmaceutical composition when used according to claim 31, wherein the pharmaceutical composition treats hyperuricemia.
35. The pharmaceutical composition when used according to claim 3 1, comprising a therapeutically effective amount of the stereoisomer of a compound of Formula 11, 0 f 0 ri4 3 x x to wherein: Ris a member selected from the group consisting of: CI-C5 alkyl, Cl-C 8 -cyclic alkyl, C 2 -C 5 alkenyl, and C 2 -C 5 alkynyl, wherein the groups are optionally substituted with one or more halogen atoms; phenyl, naphthyl and pyridyl, wherein the groups are optionally substituted with one or more substituents selected from the group consisting of halo, C 1 -C 4 alkyl, CI-C 4 alkoxy, -NO 2 -S(O)m(C,-Csalkyl), -OH, -NR 3 R -C0 2 R -CONR 3 R 4 -NR'COR 4 -NR'CONR 3 R 4 and -(CHR 3 )R 4 -ROR; -R 7 O 2 CR'NR -R'COR'; -R'NR'COR'; -R'NR -(CH 2 ),CH(R 3 )(CH 2 )qO 2 CR 9 -(CH 2 )oCH(R 3 )(CH 2 )qNR 4 COR 9 -(CH 2 ).CH(R 3 )(CH 2 )q NR 4 CONR 3 R 4 -(CH 2 )oCH(R 3 )(CH 2 )q NR 4 COOR 10 -(CH 2 ),oCH(R 3 )(CH 2 )qNR 4 SO 2 -(CHR')pCO 2 R' 2 -(CHR' )PNR 3 R 4 -(CHR 3 ),CONR'R 1R 4 0 oA ~o 112I y (CHA) R 3 0 1 R 15 R 16,N and ,0 R wherein mnis 0 to 2,0o and q are 0 to 5, p is I to 5, s is I to 3, t is I to 5, u isO0 to 1, v is I to 3, and w is 1 to (2v and wherein R 3 and R 4 are independently H, C 1 -C 5 alkyl, phenyl or benzyl; and wherein R 5 is H, C I-C 5 alkyl or NR 3 R 4 and wherein R 6is phenyl, naphthyl, pyridyl, imidazolyl, indoxyl, indolizinyl, oxazolyl, thiazolyl, thienyl, pyrimidyl, or 1-pyrazolyl optionally substituted with one or more substituents selected from the group consisting of halo, C 1 -C 4 alkyl, C 1 -C 4 alkoxy, -NO 2 -S(O)nm(Ci-C 5 alkyl), -OH, -NR 3 R 4 -C0 2 -CONR 3 R 4 -NR'COR 4 -NR'CONR 3 R 4 and and wherein R 7 (R.\LIBH]05820 doc UG alkylidene group optionally substituted with one or more groups selected from halo, hydroxyl, thiol, amino, monoalkyl amino, dialkyl amino, acylamino, carboxyl, alkylcarboxyl, acyl, aryl, aroyl, aralkyl, cyano, nitro, alkoxy, alkenyloxy, alkylcarbonyloxy and arylcarbonyloxy; and wherein R 8 is a C,-C 8 straight-chain or CN s branched alkylene or alkylidene optionally substituted with one or more groups selected from amino, monoalkyl amino, dialkyl amino, acylamino, hydroxyl, thiol, methylthiol, carboxyl and phenyl; and wherein R9 and Ro 10 are independently H, CI-Cs alkyl, optionally substituted with one or more groups consisting of C 1 -C 5 alkoxy aryl and heteroaryl, wherein the aryl is phenyl or naphthyl optionally substituted with one or more 1o substituents selected from the group consisting of halo, Ci-C 4 alkyl, C,-C 4 alkoxy, -NO 2 -S(O)m(Ci-Csalkyl), -OH, -NR 3 R 4 -C0 2 R 5 -CONR 3 R 4 -NR 3 COR 4 -NR 3 CONR 3 R 4 and and wherein the heteroaryl is pyridyl optionally substituted with one or more substituents selected from the group consisting of halo, CI-C 4 alkyl, CI-C 4 alkoxy, -NO 2 -S(O)m(Cj-Csalkyl), -OH, -NR 3 R 4 -C0 2 R, -CONR 3 R 4 -NR 3 COR 4 -NR 3 CONR 3 R 4 is and and wherein R" is methyl or phenyl, wherein the phenyl is optionally substituted with methyl and/or -NO 2 and wherein R' 2 is H, C 1 -C 5 alkyl, phenyl, benzyl, naphthyl or pyridyl, wherein the C 1 -C 5 alkyl, phenyl, naphthyl, benzyl and pyridyl are optionally substituted with one or more substituents selected from the group consisting of halo, C,-C 4 alkyl, CI-C 4 alkoxy, -NO 2 -S(O)m'(C 1 -Csalkyl), -OH, -NR 3 R 4 -C0 2 R 5 -CONR 3 R 4 -NR 3 COR 4 -NR 3 CONR 3 R 4 and and wherein R' 3 and R 4 are independently the following: alkyl, alkenyl, aryl, aralkyl or cycloalkyl, wherein the groups are optionally substituted with one or more substituents selected from the group consisting of halo, CI-C 4 alkyl, CI-C 4 alkoxy, -NO 2 -S(O)m(CI-Csalkyl), -OH, -NR 3 R 4 -C0 2 R 5 -CONR 3 R 4 -NR 3 COR 4 -NR 3 CONR 3 R 4 CH 2 NR 3 R 4 00CR'18 and and wherein when R' 3 and R' 4 are included as -(CHR 3 )CONR' 3 R' 4 NR' 3 R' 4 is 3 3 -N 0 -N OR 3 -N NR 3 -N NR 3 3 COOR N N N N OH R 3 00 C R 4 R 3 NOC -N or -0 COOR 3 [RA\LIBHO05820doc:UG -N or -N COOR 3 and wherein RS 15 is CFw or C 1 -C 5 alkyl, wherein CI-Cs alkyl is optionally substituted with the following substituents: CI-Cs alkoxy; phenyl, optionally substituted with one or more substituents selected from the group consisting of halo, C 1 -C 4 alkyl, C 1 C 4 alkoxy, -NO 2 -S(O)m(Ci-Csalkyl), -OH, -NRR 4 -C0 2 -CONR 3 R 4 -NR 3 COR 4 -NR 3 CONRR 4 and benzyl, optionally substituted with one or more substituents selected from the group consisting of halo, C 1 -C 4 alkyl, C 1 -C 4 alkoxy, -NO 2 -S(O)m(C- Csalkyl), -OH, -NR 3 R 4 -C0 2 R 5 -CONRR 4 -NR 3 COR 4 -NR 3 CONR 3 R 4 and -CF,; and wherein R16 is H, C 1 -C 5 alkyl or benzyl; and wherein R17 is C 1 -C 5 alkyl, C 3 -C 8 cyclic alkyl, phenyl or benzyl; and wherein R' 8 is H, alkyl, aryl, aralkyl or cycloalkyl, where the group is optionally substituted with one or more substituents selected from the group consisting of halo, CI-C 4 alkyl, C 1 -C 4 alkoxy, -NO 2 -S(O)m(Ci-Csalkyl), -OH, -NRR 4 -C0 2 -CONR 3 R 4 -NR 3 COR 4 -NR 3 CONR 3 R 4 and -CFw.
36. The pharmaceutical composition when used according to claim 31 wherein is the composition is in the form of a tablet or capsule.
37. A method of treating hyperuricemia in a mammal, comprising administering to said mammal a therapeutically effective amount of the stereoisomer of a compound of Formula I, X R x 1 X 2 X 4 X 3 (I) wherein: R is a member selected from the group consisting of a hydroxy, alkoxy, heteroalkoxy, aryloxy, heteroaryloxy, lower aralkoxy, di-lower alkylamino-lower alkoxy, lower alkanamido lower alkoxy, benzamido-lower alkoxy, ureido-lower alkoxy, N'-lower alkyl-ureido-lower alkoxy, carbamoyl-lower alkoxy, halophenoxy substituted lower alkoxy, carbamoyl substituted phenoxy, carbonyl-lower alkylamino, N,N-di-lower alkylamino-lower alkylamino, halo substituted lower alkylamino, hydroxy substituted lower alkylamino, lower alkanoyloxy substituted lower alkylamino, ureido, and lower alkoxycarbonylamino; and X' is a halogen; and [R:\LIBH05820.doc UG wherein the compound is substantially free of its stereoisomer.
38. The method of claim 37, wherein the compound is a compound of Formula II, 06 x 2 wherein: R 2is a member selected from the group consisting of: CI-C 5 alkyl, CI-C8-cyclic alkyl, C 2 -C 5 alkenyl, and C 2 -C5 alkynyl, wherein the groups are optionally substituted with one or more halogen atoms; phenyl, naphthyl and pyridyl, wherein the groups are optionally substituted with one or more substituents selected from the group consisting of io halo, C 1 -C 4 alkyl, CI-C 4 alkoxy, -NO 2 -S(O)m(Ci-Csalkyl), -OH, -NR 3 R 4 -C0 2 R 5 -CONR 3 R 4 -NR'COR 4 -NR'CONR 3 R 4 and -(CHR 3 )R 4 -R'0R'; -R 7 O 2 CR 8NR R4; -R'COR'; -R 7 NR 3 COR'; -R 7NR R6; -(CH 2 )oCH(R)(CH 2 )qO 2 CR 9 -(CH 2 )oCH(R 3 )(CH 2 )qNR 4 COR'; -(CH 2 )oCH(R 3 )(CH 2 )q NR 4 CONR 3 R 4 -(CH 2 )oCH(R 3 )(CH 2 )q NR 4 COOR" -(CH 2 )oCH(R 3 )(CH 2 )qNR 4 SO 2 R"; -(CHR'),CO 2 R 1 2 -(CHiR),NR 3 R 4 -(CHR 3 ),CONR' 3 R1 4 0 OX 1 1 2t(C HA) R 3 0 1 R 15 R 16 -N and \L ko ~J R1 wherein m isO0 to 2, o and q are 0 to 5, p is I to 5, s is I to 3, t is I to 5, u isO0 to 1, v is 1 to 3, and w is I to (2v and wherein R 3 and R 4 are independently H, C 1 -C 5 alkyl, phenyl or benzyl; and wherein R 5is H, C I-C5 alkyl or NR 3R 4 and wherein R 6is phenyl, naphthyl, pyridyl, imidazolyl, indoxyl, indolizinyl, oxazolyl, thiazolyl, thienyl, pyrimidyl, or 1 -pyrazolyl optionally substituted with one or more substituents selected from the group consisting of halo, CI-C 4 alkyl, C 1 -C 4 alkoxy, -N0 2 -S(O)mn(Cp-Csalkyl), -OH, -NR 3 R 4 -C0 2 R 5 -CONR 3 -NR 3 COR 4 -NR 3 CONR 3 R 4 and and wherein R 7 is a C 1 -C 8 saturated or unsaturated, straight-chain, branched or cyclic alkylene or alkylidene group optionally substituted with one or more groups selected from halo, hydroxyl, thiol, amino, monoalkyl amino, dialkyl amino, acylamino, carboxyl, alkylcarboxyl, acyl, aryl, aroyl, aralkyl, cyano, nitro, alkoxy, alkenyloxy, atkylcarbonyloxy and arylcarbonyloxy; and wherein R 8is a C 1 -C8 straight-chain or [R \LIBHJOS820.doc:UG branched alkylene or alkylidene optionally substituted with one or more groups selected from amino, monoalkyl amino, dialkyl amino, acylamino, hydroxyl, thiol, methylthiol, carboxyl and phenyl; and wherein R 9 and R10 are independently H, C 1 -Cs alkyl, optionally substituted with one or more groups consisting of C,-Cs alkoxy aryl and heteroaryl, C s wherein the aryl is phenyl or naphthyl optionally substituted with one or more substituents selected from the group consisting of halo, Ci-C 4 alkyl, C 1 -C 4 alkoxy, -NO 2 -S(O)m(Ci-Csalkyl), -OH, -NRR 4 -C0 2 R 5 -CONRR 4 -NR 3 COR 4 _NR 3 CONRR 4 and and wherein the heteroaryl is pyridyl optionally substituted with one or more substituents selected from the group consisting of halo, C 1 -C 4 alkyl, C,-C 4 alkoxy, -NO 2 -S(O)m(CI-Csalkyl), -OH, -NR3R4, -C0 2 R 5 -CONR 3 R 4 -NR 3 COR 4 -NR 3 CONR 3 R 4 and and wherein R" is methyl or phenyl, wherein the phenyl is optionally substituted with methyl and/or -NO 2 and wherein R' 2 is H, C 1 -Cs alkyl, phenyl, benzyl, naphthyl or pyridyl, wherein the CI-C 5 alkyl, phenyl, naphthyl, benzyl and pyridyl are optionally substituted with one or more substituents selected from the group consisting of is halo, CI-C 4 alkyl, CI-C 4 alkoxy, -NO 2 -S(O)..(CI-Csalkyl), -OH, -NR 3 R 4 -C0 2 R, -CONR 3 R 4 -NR 3 COR 4 -NR 3 CONR 3 R 4 and and wherein R' 3 and R' 4 are independently the following: alkyl, alkenyl, aryl, aralkyl or cycloalkyl, wherein the groups are optionally substituted with one or more substituents selected from the group consisting of halo, Ci-C 4 alkyl, CI-C 4 alkoxy, -NO 2 -S(O)m(Ci-Csalkyl), -OH, -NR 3 R 4 -C0 2 R 5 -CONR 3 R 4 -NR 3 COR 4 -NR 3 CONR 3 R 4 -CH 2 NR 3 R 4 00CR' 8 and -CF,; and wherein when R' 3 and R' 4 are included as -(CHR 3 )CONRI' 3 R' 4 NR 3 R 14 is 3 3 -N 0 -N OR -N NR 3 -N NR 3 COOR N N N N OH P 4 3 Q R 3 00C R 4 R 3 NOC -N or -N COOR 3 and wherein R' 5 is CF, or C 1 -C 5 alkyl, wherein Ci-C 5 alkyl is optionally substituted with the following substituents: Ci-C 5 alkoxy; phenyl, optionally substituted with one or more substituents selected from the group consisting of halo, C 1 -C 4 alkyl, C 1 C 4 alkoxy, -NO 2 -S(O)m(Ci-Calkyl), -OH, -NR 3 R 4 -C0 2 R 5 -CONR 3 R 4 -NR 3 COR 4 -NR 3 CONR 3 R 4 and benzyl, optionally substituted with one or more substituents [RALIBH]05820.docUG selected from the group consisting of halo, CI-C 4 alkyl, CI-C 4 alkoxy, -NO 2 -S(O)m(CI- C 5 alkyl), -OH, -NR 3 R 4 -C0 2 R 5 -CONR3R 4 -NR 3 COR 4 -NR 3 CONR 3 R 4 and -CyFw; and wherein R 16 is H, C 1 -C 5 alkyl or benzyl; and wherein R 17 is CI-C 5 alkyl, C 3 -C 8 cyclic alkyl, phenyl or benzyl; and wherein R 1 8 is H, alkyl, aryl, aralkyl or cycloalkyl, where the group is optionally substituted with one or more substituents selected from the group consisting of halo, Ci-C 4 alkyl, CI-C 4 alkoxy, -NO 2 -S(O)m(Ci-Csalkyl), -OH, -NRR 4 -C0 2 R 5 -CONR 3 R 4 -NR 3 COR 4 -NR 3 CONR 3 R 4 and -CyFw.
39. The method of claim 37, wherein the compound is administered together with a pharmaceutically acceptable carrier.
40. A method for treating insulin resistance in a mammal, comprising administering to said mammal a therapeutically effective amount of the stereoisomer of a compound of Formula I, 0 R X 1 Z x 2 X 4 X 3 (I) wherein: R is hydroxy or comprises a hydroxy group in an ester linkage which can be hydrolyzed in vivo to provide a pharmacologically effective amount of a compound of formula I wherein R is hydroxy; X' is a halogen; and X 2 X 3 and X 4 are each independently selected from hydrogen and halogen with the proviso that no more than two of X 2 X 3 and X 4 are hydrogen; or a pharmaceutically acceptable salt thereof, wherein the compound is substantially free of its stereoisomer.
41. The method of claim 40, wherein the compound is a compound of Formula II, R 2 0 x x X x 3 (II) wherein: |R:\LIBH]05820 docLUG R 2 is a member selected from the group consisting of CI-Cs alkyl, Ci-Cs-cyclic alkyl, C 2 -C5 alkenyl, and C 2 -C 5 alkynyl, wherein the groups are optionally substituted with one or more halogen atoms; heteroalkyl; phenyl-lower alkyl; benzamido-lower alkyl; di-lower alkylamino-lower alkyl; ureido-lower alkyl; N'-lower alkyl-ureido-lower alkyl; carbamoyl-lower alkyl; halophenoxy substituted lower alkyl;- (CHR 3 )R 4 -(CH 2 )oCH(R 3 )(CH 2 )qO(O)CR 9 -(CH 2 )oCH(R 3 )(CH 2 )qNR 4 C(O)R 9 -(CH 2 )oCH(R 3 )(CH 2 )qNR 4 C(O)NR 3 R 4 -(CH 2 )oCH(R 3 )(CH 2 )qNR 4 C(O)ORi; wherein o and q are each independently an integer from 0 to 5, and wherein R 3 and R 4 are independently H, unsubstituted Ci-C 5 alkyl, phenyl or benzyl; and wherein R 9 and t0 Ro 0 are independently H or unsubstituted CI-C 5 alkyl.
42. The method of claim 40, wherein the enantiomer is in an enantiomeric excess of at least
43. The method of claim 40, wherein the enantiomer is in an enantiomeric excess of at least 98%.
44. The method of claim 40, wherein the mammal is a human. The method of claim 40, wherein X 1 is chloro and each of X 2 X 3 and X 4 are fluoro.
46. The method of claim 40, wherein X' is fluoro or bromo.
47. A method for treating type 2 diabetes in a mammal, comprising administering to said mammal a therapeutically effective amount of the stereoisomer of a compound of Formula I, O X 2 X 4 X 3 (I) wherein: R is hydroxy or comprises a hydroxy group in an ester linkage which can be hydrolyzed in vivo to provide a pharmacologically effective amount of a compound of formula I wherein R is hydroxy; X' is a halogen; and X 2 X 3 and X 4 are each independently selected from hydrogen and halogen with the proviso that no more than two of X 2 X 3 and X 4 are hydrogen; or a pharmaceutically acceptable salt thereof, and wherein the compound is substantially free of its stereoisomer. [R:ALIBH]O5820doc: UG
48. The method of claim 47, wherein the compound is a compound of Formula II, R 2 0 0 X 4 X 3 (II) wherein: s R 2 is a member selected from the group consisting of CI-C 5 alkyl, Ci-Cs-cyclic alkyl, C 2 -C 5 alkenyl, and C 2 -C 5 alkynyl, wherein the groups are optionally substituted with one or more halogen atoms; heteroalkyl; phenyl-lower alkyl; benzamido-lower alkyl; di-lower alkylamino-lower alkyl; ureido-lower alkyl; N'-lower alkyl-ureido-lower alkyl; carbamoyl-lower alkyl; halophenoxy substituted lower alkyl;- (CHR 3 )R 4 -(CH 2 )oCH(R 3 )(CH 2 )qO(O)CR 9 -(CH 2 )oCH(R 3 )(CH 2 )qNR 4 C(O)R 9 -(CH 2 )oC((C(R)(CH 2 )qNR 4 C(O)NRR 4 -(CH 2 )oCH(R 3 )(CH 2 )qNR 4 C(O)ORi; wherein o and q are each independently an integer from 0 to 5, and wherein R 3 and R 4 are independently H, unsubstituted CI-C 5 alkyl, phenyl or benzyl; and wherein R 9 and R 1 0 are independently H or unsubstituted CI-Cs alkyl.
49. The method of claim 48, wherein the enantiomer is in an enantiomeric excess of at least The method of claim 48, wherein the enantiomer is in an enantiomeric excess of at least 98%.
51. The method of claim 48, wherein the mammal is a human.
52. The method of claim 48, wherein X' is chloro and each of X 2 X 3 and X 4 are fluoro.
53. The method of claim 48, wherein X' is fluoro or bromo.
54. A method for treating hyperuricemia in a mammal, comprising administering to said mammal a therapeutically effective amount of the stereoisomer of a compound of Formula I, 0 R Y-X 2 X 4 X 3 (I) wherein: R:\LIBHI05820.doc:UG R is hydroxy or comprises a hydroxy group in an ester linkage which can be hydrolyzed in vivo to provide a pharmacologically effective amount of a compound of formula I wherein R is hydroxy; X' is a halogen; and X 2 X 3 and X 4 are each independently selected from hydrogen and halogen with the proviso that no more than two of X 2 X 3 and X 4 are hydrogen; or a pharmaceutically acceptable salt thereof, and wherein the compound is substantially free of its stereoisomer. The method of claim 54, wherein the compound is a compound of Formula II, R 2 X 0 O 0 X2 X X (II) wherein: R 2 is a member selected from the group consisting of Ci-C 5 alkyl, Ci-Cs-cyclic alkyl, C 2 -C 5 alkenyl, and C 2 -C 5 alkynyl, wherein the groups are optionally substituted with one or more halogen atoms; heteroalkyl; phenyl-lower alkyl; benzamido-lower alkyl; di-lower alkylamino-lower alkyl; ureido-lower alkyl; N'-lower alkyl-ureido-lower alkyl; carbamoyl-lower alkyl; halophenoxy substituted lower alkyl; -(CHR 3 )R 4 -(CH 2 )oCH(R 3 )(CH 2 )qO(O)CR 9 -(CH 2 )oCH(R 3 )(CH 2 )qNR 4 C(O)R 9 -(CH 2 )oCH(R 3 )(CH 2 )qNR 4 C(O)NR 3 R 4 -(CH 2 )oCH(R 3 )(CH 2 )qNR 4 C(O)ORo0; wherein o and q are each independently an integer from 0 to 5, and wherein R 3 and R 4 are independently H, unsubstituted CI-C 5 alkyl, phenyl or benzyl; and wherein R 9 and R 1 0 are independently H or unsubstituted Ci-C 5 alkyl.
56. The method of claim 54, wherein the enantiomer is in an enantiomeric excess of at least
57. The method of claim 54, wherein the enantiomer is in an enantiomeric excess of at least 98%.
58. The method of claim 54, wherein the mammal is a human.
59. The method of claim 54, wherein X' is chloro and each of X 2 X 3 and X 4 are fluoro.
60. The method of claim 54, wherein X' is fluoro or bromo. [R:\LIBH]05820 doc:UG The method of claim 54, wherein X' is fluoro or bromo.
61. A pharmaceutical composition when used to treat insulin resistance in a mammal, said composition comprising a therapeutically effective amount of the stereoisomer of a compound of Formula I, 0 R x 2 X 4 X 3 (I) wherein: R is hydroxy or comprises a hydroxy group in an ester linkage which can be hydrolyzed in vivo to provide a pharmacologically effective amount of a compound of to formula I wherein R is hydroxy; X 1 is a halogen; and X 2 X 3 and X 4 are each independently selected from hydrogen and halogen with the proviso that no more than two of X 2 X 3 and X 4 are hydrogen; or a pharmaceutically acceptable salt thereof, and wherein the compound is substantially free of its stereoisomer.
62. The composition when used according to claim 61, wherein the compound is a compound of Formula II, R 2 SX 2 x 4 X 3 (II) wherein: R 2 is a member selected from the group consisting of Ci-Cs alkyl, Ci-C8-cyclic alkyl, C 2 -C 5 alkenyl, and C 2 -Cs alkynyl, wherein the groups are optionally substituted with one or more halogen atoms; heteroalkyl; phenyl-lower alkyl; benzamido-lower alkyl; di-lower alkylamino-lower alkyl; ureido-lower alkyl; N'-lower alkyl-ureido-lower alkyl; carbamoyl-lower alkyl; halophenoxy substituted lower alkyl; -(CHR)R 4 -(CH 2 )oCH(R 3 )(CH 2 )qO(O)CR 9 -(CH 2 )oCH(R 3 )(CH 2 )qNR 4 C(O)R 9 -(CH 2 )oCH(R 3 )(CH 2 )qNR 4 C(O)NR 3 R 4 -(CH 2 )oCH(R 3 )(CH 2 )qNR 4 C(O)OR 0; [R \.IBHj05820.docLUG wherein o and q are each independently an integer from 0 to 5, and wherein R 3 and R 4 are independently H, unsubstituted CI-C 5 alkyl, phenyl or benzyl; and wherein R 9 and R 10 are independently H or unsubstituted Ci-Cs alkyl.
63. The composition when used according to claim 61, wherein the enantiomer is in an enantiomeric excess of at least
64. The composition when used according to claim 61, wherein the enantiomer is in an enantiomeric excess of at least 98%. The composition when used according to claim 61, wherein the mammal is a human.
66. The composition when used according to claim 61, wherein X' is chloro and each of X 2 X 3 and X 4 are fluoro.
67. The composition when used according to claim 61, wherein X' is fluoro or bromo.
68. A pharmaceutical composition when used to treat type 2 diabetes in a mammal, said composition comprising a therapeutically effective amount of the stereoisomer of a compound of Formula I, X R X 4 X 3 (I) wherein: R is hydroxy or comprises a hydroxy group in an ester linkage which can be hydrolyzed in vivo to provide a pharmacologically effective amount of a compound of formula I wherein R is hydroxy; X' is a halogen; and X 2 X 3 and X 4 are each independently selected from hydrogen and halogen with the proviso that no more than two of X 2 X 3 and X 4 are hydrogen; or a pharmaceutically acceptable salt thereof, and wherein the compound is substantially free of its stereoisomer.
69. The composition when used according to claim 68, wherein the compound is a compound of Formula II, (R\LIBIl]05820 doc UG O R2 X 0O (N o0 x 2 X 4 X 3 (II) wherein: ¢C R 2 is a member selected from the group consisting of Ci-Cs alkyl, Ci-C 8 -cyclic C s alkyl, C 2 -C5 alkenyl, and C 2 -C 5 alkynyl, wherein the groups are optionally substituted N with one or more halogen atoms; heteroalkyl; phenyl-lower alkyl; benzamido-lower alkyl; Sdi-lower alkylamino-lower alkyl; ureido-lower alkyl; N'-lower alkyl-ureido-lower alkyl; carbamoyl-lower alkyl; halophenoxy substituted lower alkyl;--(CHR 3 )R 4 -(CH 2 )oCH(R 3 )(CH 2 )qO(O)CR 9 -(CH 2 )oCH(R )(CH 2 )qNRC(O)R 9 -(CH 2 )oCH(R 3 )(CH 2 )qNR 4 C(O)NR 3 R 4 -(CH 2 )oCH(R 3 )(CH 2 )qNR 4 C(O)ORI; wherein o and q are each independently an integer from 0 to 5, and wherein R 3 and R 4 are independently H, unsubstituted Ci-C 5 alkyl, phenyl or benzyl; and wherein R 9 and R' i are independently H or unsubstituted Ci-C 5 alkyl. The composition when used according to claim 68, wherein the enantiomer is in an enantiomeric excess of at least
71. The composition when used according to claim 68, wherein the enantiomer is in an enantiomeric excess of at least 98%.
72. The composition when used according to claim 68, wherein the mammal is a human.
73. The composition when used according to claim 68, wherein X' is chloro and each of X 2 X 3 and X 4 are fluoro.
74. The composition when used according to claim 68, wherein X' is fluoro or bromo. A pharmaceutical composition when used to treat hyperuricemia in a mammal, said composition comprising a therapeutically effective amount of the stereoisomer of a compound of Formula I, X R X1 r [R:\LIBH]05820 docLJG wherein: R is hydroxy or comprises a hydroxy group in an ester linkage which can be hydrolyzed in vivo to provide a pharmacologically effective amount of a compound of formula I wherein R is hydroxy; s X' is a halogen; and X 2 X 3 and X 4 are each independently selected from hydrogen and halogen with the proviso that no more than two of X 2 X 3 and X 4 are hydrogen; or a pharmaceutically acceptable salt thereof, and wherein the compound is substantially free of its stereoisomer.
76. The composition when used according to claim 75, wherein the compound is a compound of Formula II, R 2 0 1 0 X OZ x 2 X 4 x 3 (II) wherein: R 2 is a member selected from the group consisting of C 1 -Cs alkyl, CI-C8-cyclic alkyl, C 2 -C 5 alkenyl, and C 2 -C 5 alkynyl, wherein the groups are optionally substituted with one or more halogen atoms; heteroalkyl; phenyl-lower alkyl; benzamido-lower alkyl; di-lower alkylamino-lower alkyl; ureido-lower alkyl; N'-lower alkyl-ureido-lower alkyl; carbamoyl-lower alkyl; halophenoxy substituted lower alkyl;- (CHR)R 4 -(CH 2 )oCH(R)(CH 2 )qO(O)CR 9 -(CH 2 )oCH(R 3 )(CH 2 )qNR 4 C(O)R 9 -(CH 2 )oCH(R 3 )(CH 2 )qNR 4 C(O)NR 3 R 4 -(CH 2 )oCH(R 3 )(CH 2 )qNR 4 C(O)OR' O wherein o and q are each independently an integer from 0 to 5, and wherein R 3 and R 4 are independently H, unsubstituted CI-C 5 alkyl, phenyl or benzyl; and wherein R 9 and R 1 0 are independently H or unsubstituted C 1 -C 5 alkyl.
77. The composition when used according to claim 75, wherein the enantiomer is in an enantiomeric excess of at least
78. The composition when used according to claim 75, wherein the enantiomer is in an enantiomeric excess of at least 98%.
79. The composition when used according to claim 75, wherein the mammal is a human. [R:\LIBH]05820 doc UG The composition when used according to claim 75, wherein X' is chloro and each of X 2 X 3 and X 4 are fluoro.
81. The composition when used according to claim 75, wherein X' is fluoro or bromo.
82. A method of treating insulin resistance, type 2 diabetes, or hyperuricemia in a mammal, comprising: administering to said mammal a therapeutically effective amount of the stereoisomer of a compound of Formula I substantially as hereinbefore described with reference to any one of the examples.
83. Use of the stereoisomer of a compound of Formula I, SR x 2 X4 X3 (I) wherein: R is a member selected from the group consisting of a hydroxy, alkoxy, heteroalkoxy, aryloxy, heteroaryloxy, lower aralkoxy, di-lower alkylamino-lower alkoxy, lower alkanamido lower alkoxy, benzamido-lower alkoxy, ureido-lower alkoxy, N'-lower alkyl-ureido-lower alkoxy, carbamoyl-lower alkoxy, halophenoxy substituted lower alkoxy, carbamoyl substituted phenoxy, carbonyl-lower alkylamino, N,N-di-lower alkylamino-lower alkylamino, halo substituted lower alkylamino, hydroxy substituted lower alkylamino, lower alkanoyloxy substituted lower alkylamino, ureido, and lower alkoxycarbonylamino; and X' is a halogen; and X 2 X 3 and X 4 are each independently selected from hydrogen and halogen with the proviso that no more than two of X 2 X 3 and X 4 are hydrogen; or a pharmaceutically acceptable salt thereof, wherein the compound is substantially free of its stereoisomer, in the manufacture of a medicament for treating insulin resistance, Type 2 diabetes, or hyperuricemia. Dated 24 May, 2006 Metabolex, Inc. Patent Attorneys for the Applicant/Nominated Person SPRUSON FERGUSON [R:\LIBH)05820doc:UG
AU2002239371A 1999-06-04 2001-11-28 Use of(-) (3-halomethylphenoxy) (4-halophenyl) acetic acid derivatives for treatment of insulin resistance, type 2 diabetes, hyperlipidemia and hyperuricemia Ceased AU2002239371B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2002239371A AU2002239371B2 (en) 1999-06-04 2001-11-28 Use of(-) (3-halomethylphenoxy) (4-halophenyl) acetic acid derivatives for treatment of insulin resistance, type 2 diabetes, hyperlipidemia and hyperuricemia

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US09/325997 1999-06-04
AU53162/00A AU775909C (en) 1999-06-04 2000-06-02 Use of (-) (3-trihalomethylphenoxy) (4-halophenyl) acetic acid derivatives for treatment of insulin resistance, type 2 diabetes, hyperlipidemia and hyperuricemia
US09/724,788 US6624194B1 (en) 1999-06-04 2000-11-28 Use of (−) (3-trihalomethylphenoxy) (4-halophenyl) acetic acid derivatives for treatment of insulin resistance, type 2 diabetes, hyperlipidemia and hyperuricemia
US09/724,788 2000-11-28
AU2002239371A AU2002239371B2 (en) 1999-06-04 2001-11-28 Use of(-) (3-halomethylphenoxy) (4-halophenyl) acetic acid derivatives for treatment of insulin resistance, type 2 diabetes, hyperlipidemia and hyperuricemia
PCT/US2001/044603 WO2002044113A2 (en) 2000-11-28 2001-11-28 Use of(-) (3-halomethylphenoxy) (4-halophenyl) acetic acid derivatives for treatment of insulin resistance, type 2 diabetes, hyperlipidemia and hyperuricemia

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU53162/00A Division AU775909C (en) 1999-06-04 2000-06-02 Use of (-) (3-trihalomethylphenoxy) (4-halophenyl) acetic acid derivatives for treatment of insulin resistance, type 2 diabetes, hyperlipidemia and hyperuricemia

Related Child Applications (1)

Application Number Title Priority Date Filing Date
AU2006225263A Division AU2006225263A1 (en) 1999-06-04 2006-10-05 Use of (-) (3-halomethylphenoxy) (4-halophenyl) acetic acid derivatives for treatment of insulin resistance, type 2 diabetes, hyperlipidemia and hyperuricemia

Publications (2)

Publication Number Publication Date
AU2002239371A1 AU2002239371A1 (en) 2002-08-15
AU2002239371B2 true AU2002239371B2 (en) 2006-07-06

Family

ID=39338558

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2002239371A Ceased AU2002239371B2 (en) 1999-06-04 2001-11-28 Use of(-) (3-halomethylphenoxy) (4-halophenyl) acetic acid derivatives for treatment of insulin resistance, type 2 diabetes, hyperlipidemia and hyperuricemia

Country Status (1)

Country Link
AU (1) AU2002239371B2 (en)

Similar Documents

Publication Publication Date Title
US6613802B1 (en) Use of (-) (3-trihalomethylphenoxy) (4-halophenyl) acetic acid derivatives for treatment of insulin resistance, type 2 diabetes, hyperlipidemia and hyperuricemia
US8481597B2 (en) Use of (-) (3-trihalomethylphenoxy) (4-halophenyl) acetic acid derivatives for treatment of insulin resistance, type 2 diabetes, hyperlipidemia and hyperuricemia
AU2006225263A1 (en) Use of (-) (3-halomethylphenoxy) (4-halophenyl) acetic acid derivatives for treatment of insulin resistance, type 2 diabetes, hyperlipidemia and hyperuricemia
JP2003501383A5 (en)
AU2002239371B2 (en) Use of(-) (3-halomethylphenoxy) (4-halophenyl) acetic acid derivatives for treatment of insulin resistance, type 2 diabetes, hyperlipidemia and hyperuricemia
AU2002239371A1 (en) Use of(-) (3-halomethylphenoxy) (4-halophenyl) acetic acid derivatives for treatment of insulin resistance, type 2 diabetes, hyperlipidemia and hyperuricemia

Legal Events

Date Code Title Description
DA3 Amendments made section 104

Free format text: THE NATURE OF THE AMENDMENT IS: ADD PRIORITY DETAILS 53162/00 2 JUN 2000 AU

FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired