Novel pyridine compounds, process for their preparation and compositions containing them

Abstract
The present invention provides new heterocyclic compounds, particularly substituted pyridines, methods and compositions for making and using these heterocyclic compounds, and methods for treating a variety of diseases and disease states, including atherosclerosis, arthritis, restenosis, diabetic nephropathy, or dyslipidemia, or disease states mediated by the low expression of Perlecan.
Description
FIELD OF THE INVENTION

The present invention relates to substituted pyridine compounds, methods and compositions for making and using substituted pyridine compounds, and methods for preventing or treating diseases in humans or animals employing such compounds and compositions.


BACKGROUND OF THE INVENTION

Novel compounds for new therapeutic interventions are needed for many areas of medicine and disease treatment. For example, chronic and acute inflammatory conditions form the basis for diseases affecting all organ systems including, but not limited to, asthma, acute inflammatory diseases, vascular inflammatory disease, chronic inflammation, atherosclerosis, angiopathy, myocarditis, nephritis, Crohn's disease, arthritis, type I and II diabetes and associated vascular pathologies. The incidence of these inflammatory conditions is on the rise in the population as a whole, with diabetes alone affecting 16 million people. Therefore, synthesis of novel compounds leads to new possibilities for discovery of novel therapeutic interventions.


While inflammation in and of itself is a normal immune response, chronic inflammation leads to complications and ongoing system damage due to the interactions of unknown cellular factors. In particular, chronic inflammation can cause endothelial damage resulting in vascular complications. Coronary artery, cerbrovascular and peripheral vascular disease resulting from atherosclerotic and thromboembolic macroangiopathy are the primary causes of mortality in chronic inflammatory diseases.


Many humans and animals have limited lifespans and lifestyles because of conditions relating to lifestyle choices, such as diet and exercise, or because of genetic predispositions to develop a disease. For example, vascular smooth muscle cell proliferation is a common consequence of endothelial injury and is believed to be an early pathogenetic event in the formation of atherosclerotic plaques or complications related to vascular injury or as a result surgical interventions. Abnormal vascular smooth muscle cell (SMC) proliferation is thought to contribute to the pathogenesis of vascular occlusive lesions, including arteriosclerosis, atherosclerosis, restenosis, and graft atherosclerosis after organ transplantation.


Percutaneous coronary artery intervention (PTCA) procedures are the most common in-patient hospital procedure in the United States. According to the American Heart Association, about one-third of the patients that undergo balloon angioplasty have restenosis of the widened segment of the vessel within approximately 6 months. It may be necessary to perform another angioplasty or coronary artery bypass surgery on restenosed arteries. A key feature of restenosis is an injury response that results in activation of an inflammatory cascade and remodeling of the cells both inside and outside the carotid artery wall. This includes excessive growth of connective tissue and smooth muscle into the lumen of the artery known as neointimal hyperplasia. Currently there are no effective pharmacological treatments available that control the pathogenesis of vascular occlusive lesions, such as, but not limited to, arteriosclerosis, atherosclerosis, restenosis, and graft atherosclerosis after organ transplantation. Identification of effective therapeutics with minimal side effects will restore quality of life without requiring additional surgical procedures such as coronary artery bypass surgery.


Control or modulation of factors produced by the body in response to injury, surgery, metabolic factors or loss of control of in feedback mechanisms, leading to too much or too little of a factor has long been the goal of administering pharmacological agents. One disease that rapidly growing in the industrialized countries is the occurrence of diabetes and all of its attendant sequellae. One of the factors important in the damage associated with diabetes is the presence of glycated proteins.


Glycated proteins and advanced glycation end products (AGE) contribute to cellular damage, particularly, diabetic tissue injury, by at least by two major mechanisms: modulation of cellular functions through interactions with specific cell surface receptors; and alteration of the extracellular matrix leading to the formation of protein cross-links. Studies suggest that glycated protein and AGE interactions with cells may promote inflammatory processes and oxidative cellular injury. AGE increases lipoprotein oxidizability and atherogenicity. Its binding to matrix proteins induces synthesis of cytokines and activates cellular messengers. Diseases where glycated protein and AGE accumulation is a suspected etiological factor include vascular complications of diabetes, microangiopathies, renal insufficiency and Alzheimer's disease.


The exact mechanisms by which high plasma glucose, as seen in diabetes, causes microvascular damage are not completely understood. One potential mechanism by which hyperglycemia can be linked to microangiopathies is through the process of non-enzymatic glycation of critical proteins. Non-enzymatic glycation, i.e., the linking of proteins with glucose, leads to the formation of glycated proteins. The first step in this glycation pathway involves the non-enzymatic condensation of glucose with free amino groups in the protein, primarily the epsilon-amino groups of lysine residues, forming the Amadori adducts. These early glycation products can undergo further reactions such as rearrangements, dehydration and condensations to form irreversible advanced glycation end products (AGE). These are a highly reactive group of molecules whose interaction with specific receptors on the cell-surface which are thought to lead to pathogenic outcomes.


Other major area of disease of where treatments are needed and for which adequate and effective therapies do not exist are cellular proliferative disorders, or disorders caused by unwanted or unintended cellular growth. As mentioned, smooth muscle cell (SMC) hyperplasia is a major event in the development of atherosclerosis and is also responsible for the significant number of failure rates following vascular procedures such as angioplasty, stent implantation and coronary artery bypass surgery. In the normal vessel, SMC are quiescent, but they proliferate when damage to the endothelium occurs. Naturally occurring growth modulators, many of which are derived from the endothelium, tightly control SMC proliferation in vivo. When the control becomes unregulated, a pathological state is induced in the subject.


Another major area of unwanted cellular growth, that is unchecked by the body's regulatory systems, is cancer or oncological conditions. Many therapies have been used and are being used in an effort to restore health or at least stop the unwanted cell growth. Many times, therapeutic agents can have an effect individually, but often, therapeutic regimes require combinations of different pharmacological agents with treatments such as surgery or radiation.


There is a present need for treatments of chronic or acute diseases, such as atherosclerosis, unwanted cellular growth or cellular proliferation, diabetes, inflammatory conditions and vascular occlusive pathologic conditions. Because of occurrence is frequent, the currently available treatments are costly and the conditions are refractory to many pharmacological therapies. The mechanisms involved in the control or prevention of such diseases are not clear and there exists a need for preventive and therapeutic treatments of these and other diseases. Thus, what is presently needed are novel compounds that find utility in methods and compositions for treatment and prevention of chronic and acute diseases, to which the present invention is directed.


SUMMARY OF THE INVENTION

The present invention is directed to novel pyridines, novel compositions comprising pyridines, and novel methods employing such pyridines and compositions. Disclosed herein are methods for making pyridines, compositions comprising pyridines, and methods and compositions for using pyridines. The pyridine compounds and compositions comprising the pyridine compounds have utility in treating and preventing a variety of diseases.


In one aspect, compounds in accordance with the present invention, and compositions comprising these compounds, comprise nitrogen heterocyclic compounds of formulas (I):
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein one of X and Y is nitrogen and the other of X and Y is CH;


Y1 and Y2, in each occurrence, are independently >NR5, —(CH2)n—, —(CH2)p-(CH═CH)(CH2)q-, >CR5R6, —(CH2)p(C≡C)(CH2)q-, —O—, >CO, —S—, >SO or >SO2; wherein n, p, and q are independently an integer from 0 to 3;


R5 and R6, in each occurrence, are independently: 1) a substituted or an unsubstituted alkyl, aryl, alkoxyalkyl, heteroaryl, cycloalkyl, or heterocyclyl, any of which having up to 10 carbon atoms; wherein any heteroaryl or heterocyclyl comprises at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; or 2) hydrogen;


wherein when Y1 or Y2 is independently >NR5;

    • 1) the corresponding Rz, wherein z is 1 or 2, in each occurrence is independently selected from: a) a substituted or an unsubstituted alkyl, aryl, alkoxyalkyl, cycloalkyl, —COR9, aralkyl, heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; any of which having up to 10 carbon atoms; b) hydrogen; or c) halogen; or
    • 2) the corresponding YzRz, wherein z is 1 or 2, is a cyclic structure selected from: a) a substituted or an unsubstituted cyclic ring, which optionally comprises at least one additional heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; or b) a substituted or an unsubstituted morpholinyl, piperazinyl, thiomorpholinyl, pyrrolidinyl, or piperidinyl; any of which having up to 10 carbon atoms; wherein the optional substituents on the cyclic YzRz structure are independently selected from at least one of: i) hydroxyl or halogen; or ii) alkyl, alkoxy, haloalkyl, cycloalkyl, aryl, or heteroaryl any of which having up to 10 carbon atoms;
    • wherein when YzRz is piperazinyl, the piperazine nitrogen is optionally substituted by an alkyl, a cycloalkyl, an acyl, a haloalkyl, an alkoxyalkyl, SO2R7, SO2NR72, or CO2R7, wherein R7 is independently selected from: a) an alkyl or an aryl having up to 8 carbon atoms; or b) hydrogen;


wherein when Y1 or Y2 is independently —O— or —S—; the corresponding Rz, wherein z is 1 or 2, in each occurrence is independently selected from: 1) a substituted or an unsubstituted alkyl, aryl, alkoxyalkyl, cycloalkyl, —COR9, aralkyl, heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; any of which having up to 10 carbon atoms; or 2) hydrogen;


wherein when Y1 or Y2 is independently —(CH2)n-, —(CH2)p(CH═CH)(CH2)q-, >CR5R6, —(CH2)p(C≡C)(CH2)q-, >CO, >SO or >SO2; the corresponding Rz, wherein z is 1 or 2, in each occurrence is independently selected from: 1) a substituted or an unsubstituted alkyl, haloalkyl, cycloalkyl, —COR9, aralkyl, alkoxy, alkenyl, alkynyl, alkoxyalkyl, aryl, —CO2R5, —COR5, heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; any of which having up to 10 carbon atoms; or 2) hydrogen, halogen, cyano, or hydroxyl;


wherein R3 and R4, in each occurrence, are independently: 1) a substituted or an unsubstituted alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, haloalkyl, haloalkoxy, alkylthio, alkylsufonyl, aryl, —CO2R5, —COR5, —NR5R6, —SO2NR5R6, —SO3R5, heterocycly or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; any of which having up to 10 carbon atoms; 2) hydrogen; halogen; hydroxyl; or cyano; or 3) Y1R1;


wherein any of R1, R2, R5, or R6 is optionally substituted with at least one group independently selected from: 1) alkyl; alkoxy; alkylthio; haloalkyl; cycloalkyls; aryl; heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; haloalkoxy; —OCH2O—; —OCOR9; N(R8)2; —COR9; —CON(8)2; —(CH2)bCO2R8 wherein b is an integer from 0 to 3; —OCO(CH2)bCO2R10 wherein b is an integer from 0 to 3; —SO2R9; —NHSO2R9; or —SO2N(R8)2; any of which having up to 10 carbon atoms; or 2) hydrogen, halogen, hydroxyl, or cyano;


wherein R8, in each occurrence, is independently: 1) an alkyl; a haloalkyl; a heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; or an aryl having up to 10 carbon atoms; or 2) hydrogen;


wherein R9, in each occurrence, is independently an alkyl; a haloalkyl; an aryl; or a heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; having up to 8 carbon atoms; wherein R9 is optionally substituted with: 1) an alkyl, an alkoxy, a carboxylic acid, or a carboxylic acid ester, any of which having up to 8 carbon atoms; 2) halogen; or 3) hydroxyl; and


wherein any of R3 or R4 is optionally substituted with at least one group independently selected from: 1) alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyl, aryl, heteroaryl, heterocyclyl, alkenyl, alkynyl, —COR10, —CO2R10, —CON(R10


)2, —SO2R10, —SO2N(R10)2, or —N(R10)2, any of which having up to 10 carbon atoms; 2) halogen; or 3) hydroxyl; and


wherein R10, in each occurrence, is independently: 1) an alkyl or an aryl having up to 10 carbon atoms; or hydrogen.


In another aspect, compounds in accordance with the present invention, and compositions comprising these compounds, comprise nitrogen heterocyclic compounds of formula (III):
embedded image

or a salt, including a pharmaceutically acceptable or non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein R1, R2, R3, R4, Y1, Y2, as well as other substituents of formula (III), are as disclosed above for the compounds of formula (I).


The present invention is directed to methods and compositions comprising compounds that have utility in treatment of pathological conditions. One aspect of the present invention comprises pyridines and compositions comprising pyridines in methods for treating diseases related to unwanted cellular proliferation. Vascular diseases, such as cardiovascular diseases, organ transplant sequellae, vascular occlusive conditions including, but not limited to, neointimal hyperplasia, restenosis, transplant vasculopathy, cardiac allograft vasculopathy, atherosclerosis, and arteriosclerosis, are caused by or have collateral damage due to unwanted cellular proliferation, such as smooth muscle cell (SMC) hyperplasia. At least one activity of one or more of these compounds is that the compound has the activity of affecting the synthesis of proteoglycans including induction and synthesis of proteoglycans and active fragments of proteoglycans. Methods comprise administration of compositions comprising compounds that have at least the activity of affecting cellular proliferation and affecting proteoglycan synthesis and activity. Further, the pyridines and compositions comprising pyridines disclosed herein can be employed to prevent or to treat the aforementioned diseases.


The present invention also comprises methods and compositions comprising pyridines described herein that have an activity associated with modulation of glycosidase enzymes and thus, affecting the substrates for such enzymes. Glycosidase enzymes and their activity with their substrates, such as proteoglycans or glycated proteins, are aspects of a variety of diseases such as vascular conditions, proteoglycan-associated diseases, kidney disease, autoimmune disease and inflammatory diseases. Pyridines described herein that have an activity that affects the concentrations of substrates of glycosidase enzymes are used in methods of treatment of such vascular, inflammatory, metastatic and systemic diseases.


Another aspect of the present invention comprises methods and compositions comprising pyridines of the present invention for the treatment and prevention of conditions or diseases that have as an aspect of the disease or condition, inflammation. An aspect of the present invention is directed to methods and compositions comprising pyridines that are effective in inhibiting inflammation, particularly inflammation associated with the accumulation or presence of glycated proteins or AGE. Methods of treatment comprise administration of compositions comprising pyridines having at least the activity of modulating inflammatory reactions that are components of biological conditions including, but not limited to, vascular complications of type I and type II diabetic-induced vasculopathies, other vasculopathies, microangiopathies, renal insufficiency, Alzheimer's syndrome, and inflammation-induced diseases such as atherosclerosis. An aspect of the present invention also comprises methods and compositions for the treatment of diseases, preconditions or pathologies associated with inflammatory cytokines and other inflammation related molecules.


Another aspect of the present invention comprises methods and compositions comprising compounds that have at least the activity of causing cellular death or a cessation of cellular activity, referred to herein as cytotoxic activity. This activity can be used in methods for in vitro or in vivo cytotoxicity. For example, compounds having this activity can be selectively delivered to an area within a living organism to selectively kill cells in that area. Such methods are using in treating hyperproliferative cells, such as cancers, or other unwanted cellular growth or cellular activities. One aspect of the invention provides compositions comprising compounds that nonselectively kill cells. Another aspect of the invention provides compounds that selectively kill cells, for example, cells that have a particular cellular marker or other identifying characteristic such as metabolic rate or uptake of a particular compound.


Accordingly, in one aspect, this invention also provides compositions comprising a pharmaceutically acceptable carrier and at least one compound as disclosed herein, and further comprising: optionally, a pharmaceutically acceptable auxiliary; optionally, a pharmaceutically acceptable preservative; optionally, a pharmaceutically acceptable excipient; optionally, a pharmaceutically acceptable diluent; and optionally, a pharmaceutically acceptable solvate. In this aspect, this composition can be in the form of, for example, a tablet, a capsule, a syrup, a cachet, a powder, a granule, a solution, a suspension, an emulsion, a bolus, a lozenge, a suppository, a pessary, a tampon, a cream, a gel, a paste, a foam, a spray, an aerosol, a microcapsule, a liposome, a transdermal patch, a pastille, a paste, or a mouthwash, and the like. Also in this aspect, this composition can further comprise an agent selected from a chemotherapeutic agent, an immunosuppressive agent, a cytokine, a cytotoxic agent, an anti-inflammatory agent, an antirheumatic agent, a cardiovascular agent, or any combination thereof.


The present invention also comprises pharmaceutical compositions comprising the compounds disclosed herein. Routes of administration and dosages of effective amounts of the compounds and pharmaceutical compositions are also disclosed. For example, the compounds of the present invention can be administered in combination with other pharmaceutical agents in a variety of protocols for effective treatment of disease.


In another aspect, the present invention relates to drug delivering or eluting medical devices that contain or are coated with at least one compound disclosed herein. The medical device suitable for use with the compounds of the present invention include, but are not limited to, stents and other medical devices that can provide a substrate for delivery of at least one compound.


Other aspects of the present invention comprise compositions and methods for microarray devices. Such microarray devices and methods comprise a variety of microarrays that may be used, for example, to study and monitor gene expression in response to treatment with the compounds of the present invention. The microarrays may comprise nucleic acid sequences, carbohydrates or proteins that are determinative for specific cells, tissues, species, disease states, prognoses, disease progression, or any other combination of molecules that can be used to determine an effect of one or more of the compounds of the present invention. Other aspects of the present invention comprise methods using databases and computer applications.







DETAILED DESCRIPTION OF THE INVENTION

In accordance with the present invention, novel pyridine compounds, and novel compositions comprising pyridine compounds are described herein. In one aspect, compounds in accordance with the present invention, and compositions comprising these compounds, comprise nitrogen heterocyclic compounds of formula (IIIi):
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


Y1 and Y2, in each occurrence, are independently >NR5, —(CH2)n- wherein n is 0 or 1, —S—, —O—, >CO, or >SO2;


R5, in each occurrence, is independently: 1) a substituted or an unsubstituted alkyl, aryl, alkoxyalkyl, heteroaryl, cycloalkyl, or heterocyclyl, any of which having up to 10 carbon atoms; wherein any heteroaryl or heterocyclyl comprises at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; or 2) hydrogen;


wherein when Y1 or y2 is independently >NR5;

    • 1) the corresponding Rz, wherein z is 1 or 2, in each occurrence is independently selected from: a) a substituted or an unsubstituted alkyl, aryl, alkoxyalkyl, cycloalkyl, —COR9, aralkyl, heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; any of which having up to 10 carbon atoms; b) hydrogen; or c) halogen; or
    • 2) the corresponding YzRz, wherein z is 1 or 2, is selected from a morpholinyl, a piperazinyl, or a piperidinyl; any of which having up to 10 carbon atoms, wherein when YzRz is piperazinyl, the piperazine nitrogen is optionally substituted by an alkyl, a cycloalkyl, an acyl, a haloalkyl, an alkoxyalkyl, SO2R7, SO2NR72, or CO2R7, wherein R7 is independently selected from: a) an alkyl or an aryl having up to 8 carbon atoms; or b) hydrogen;


wherein when Y1 or Y2 is independently —O— or —S—; the corresponding Rz, wherein z is 1 or 2, in each occurrence is independently selected from: 1) a substituted or an unsubstituted alkyl, aryl, alkoxyalkyl, cycloalkyl, —COR9, aralkyl, heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; any of which having up to 10 carbon atoms; or 2) hydrogen;


wherein when Y1 or Y2 is independently —(CH2)n-, >CO, or >SO2; the corresponding Rz, wherein z is 1 or 2, in each occurrence is independently selected from: 1) a substituted or an unsubstituted alkyl, haloalkyl, cycloalkyl, —COR9, aralkyl, alkoxy, alkenyl, alkynyl, alkoxyalkyl, aryl, —CO2R5, —COR5, heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; any of which having up to 10 carbon atoms; or 2) hydrogen, halogen, cyano, or hydroxyl;


wherein R3 and R4, in each occurrence, are independently: 1) haloalkyl having less than 3 carbon atoms; 2) alkyl, haloalkoxy, aryl, cycloalkyl, heteroaryl, or heterocyclyl having up to 10 carbon atoms, wherein any heteroaryl or heterocyclyl comprises at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; 3) hydrogen; or 4) Y1R1;


wherein any of R1, R2, or R5 is optionally substituted with at least one group independently selected from: 1) alkyl, alkoxy, haloalkyl, haloalkoxy, —OCH2O—, N(R8)2, —SO2R9, —OCOR9 or —SO2N(R8)2, any of which having up to 10 carbon atoms; or 2) hydrogen, halogen, or cyano;


wherein R8, in each occurrence, is independently: 1) an alkyl; a haloalkyl; a heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; or an aryl having up to 10 carbon atoms; or 2) hydrogen;


wherein R9, in each occurrence, is independently an alkyl; a haloalkyl; an aryl; or a heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; having up to 8 carbon atoms; wherein R9 is optionally substituted with: 1) an alkyl, an alkoxy, a carboxylic acid, or a carboxylic acid ester, any of which having up to 8 carbon atoms; 2) halogen; or 3) hydroxyl; and


wherein any of R3 or R4 is optionally substituted with at least one group independently selected from: 1) alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyl, aryl, heteroaryl, heterocyclyl, alkenyl, alkynyl, —COR10, —CO2R10, —CON(R10)2, —SO2R10, —SO2N(R10)2, or —N(R10)2, any of which having up to 10 carbon atoms; 2) halogen; or 3) hydroxyl; and


wherein R10, in each occurrence, is independently: 1) an alkyl, a heterocyclyl, or an aryl having up to 10 carbon atoms; or hydrogen.


2. (Revised 17)


In another aspect, the present invention provides for compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


Y1 and Y2 are independently selected from >NR5, —(CH2)n- wherein n is 0 or 1, or —O—;


R5 is selected from: 1) a substituted or an unsubstituted alkyl, aryl, cycloalkyl, or heteroaryl or heterocyclyl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO, any of which having up to 10 carbon atoms; or 2) hydrogen;


wherein when yl or Y2 is independently >NR5;

    • 1) the corresponding Rz, wherein z is 1 or 2, in each occurrence is independently selected from: a) a substituted or an unsubstituted alkyl, aryl, cycloalkyl, or heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO, any of which having up to 10 carbon atoms; or b) hydrogen; or
    • 2) the corresponding YzRz, wherein z is 1 or 2, is a cyclic structure selected from: a) a substituted or an unsubstituted heterocyclic ring, which optionally comprises at least one additional heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; or b) a substituted or an unsubstituted morpholinyl, piperazinyl, thiomorpholinyl, pyrrolidinyl, or piperidinyl; any of which having up to 10 carbon atoms; wherein the optional substituents on the heterocyclic YzRz structure are independently selected from a) hydroxyl; or b) alkyl, alkoxy, haloalkyl, aryl, or heteroaryl; any of which having up to 10 carbon atoms;
    • wherein when YzRz is piperazinyl, the piperazine nitrogen is optionally substituted by an alkyl, a cycloalkyl, an acyl, a haloalkyl, an alkoxyalkyl, SO2R7, SO2NR72, or CO2R7, wherein R7 is independently selected from: a) an alkyl or an aryl having up to 8 carbon atoms; or b) hydrogen;


wherein when Y1 or Y2 is independently —(CH2)n-, the corresponding Rz, wherein z is 1 or 2, in each occurrence is independently selected from: 1) an alkyl, a haloalkyl, —COR9, an alkoxy, an alkenyl, an alkynyl, an alkoxyalkyl, an aryl, —CO2R5, —COR5, or a heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; any of which having up to 10 carbon atoms; or 2) hydrogen, halogen, cyano, or hydroxyl;


wherein R4, in each occurrence, is independently selected from: 1) an alkyl, an alkenyl, an alkynyl, an alkoxy, a haloalkyl, an alkylsufonyl, an aryl, —CO2R5, —COR5, —NR5R6, —SO2NR5R6, or a heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; any of which having up to 10 carbon atoms; or 2) hydrogen, halogen, hydroxyl, or cyano;


wherein R1 and R2 are optionally substituted with at least one group independently selected from: 1) an alkyl, an alkoxy, an alkylthio, a haloalkyl, a cycloalkyl, an aryl, a haloalkoxy, NR82, —COR9, —CONR82, —SO2R9, —NHSO2R9, or —SO2NR82, or a heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; any of which having up to 10 carbon atoms; or 2) hydrogen, halogen, —OCH2O—, hydroxyl, or cyano;


R8, in each occurrence, is selected independently from: 1) an alkyl or a haloalkyl, any of which having up to 10 carbon atoms; or 2) hydrogen;


R9, in each occurrence, is selected independently from: 1) an alkyl or a haloalkyl, any of which having up to 10 carbon atoms; or 2) hydrogen or hydroxyl; and


R4 is optionally substituted with at least one group independently selected from: 1) an alkyl, an alkoxy, a haloalkyl, a haloalkoxy, a cycloalkyl, an aryl, an alkenyl, an alkynyl, —COR10, —CO2R10, —CONR102, —SO2R10, —SO2NR102, —NR102, or a heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO, any of which having up to 10 carbon atoms; or 2) halogen or hydroxyl; and


wherein R10, in each occurrence, is selected independently from: 1) an alkyl or an aryl having up to 10 carbon atoms; or 2) hydrogen.


In still another aspect, the present invention provides for compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


Y1 and Y2 are selected independently from >NR5 or —(CH2)n- wherein n is 0 or 1;


R5 is hydrogen or methyl;


wherein when Y1 or Y2 is independently >NR5;

    • 1) the corresponding Rz, wherein z is 1 or 2, in each occurrence is selected independently from: a) an alkyl, an aryl, a cycloalkyl, or a heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO, any of which having up to 10 carbon atoms; or b) hydrogen; or
    • 2) the corresponding YzRz, wherein z is 1 or 2, is selected independently from a morpholinyl, a piperazinyl, a piperidinyl, or a pyrrolidinyl;
    • wherein when YzRz is a piperazinyl, the piperazine nitrogen is optionally substituted by an alkyl, a cycloalkyl, an acyl, a haloalkyl, an alkoxyalkyl, SO2R7, or SO2NR72, any of which having up to 10 carbon atoms, wherein R7 is selected independently from: a) an alkyl or an aryl having up to 8 carbon atoms; or b) hydrogen;
    • wherein when YzRz is a piperidinyl or a pyrrolidinyl, the ring is optionally substituted by: a) an alkyl or a haloalkyl having up to 10 carbon atoms; or 2) hydroxyl;


wherein when Y1 or Y2 is independently —(CH2)n-, the corresponding Rz, wherein z is 1 or 2, in each occurrence is selected independently from: 1) an alkyl, a cycloalkyl, a haloalkyl, an alkoxy, an aryl, or a heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO, any of which having up to 10 carbon atoms; or 2) hydrogen, halogen, cyano, or hydroxyl;


R4 is selected independently from: 1) a haloalkyl having less than 3 carbon atoms; 2) an alkyl, a haloalkoxy, an aryl, or a heteroaryl or heterocyclyl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO, any of which having up to 10 carbon atoms; 3) hydrogen or halogen; or 4) Y1R1;


wherein any of R1, R2, or R5 is optionally substituted with at least one group independently selected from: 1) an alkyl, an alkoxy, a haloalkyl, a haloalkoxy, NR82, —COR9, —CO2R8, —CONR8, —SO2R9, or —SO2NR82, any of which having up to 10 carbon atoms; or 2) hydrogen, hydroxyl, halogen, —OCH2O—, or cyano;


R8, in each occurrence, is selected independently from: 1) an alkyl, a haloalkyl, or an aryl, any of which having up to 10 carbon atoms; or 2) hydrogen;


R9, in each occurrence, is selected independently from an alkyl, a haloalkyl, an aryl, or a heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, or —S—, any of which having up to 10 carbon atoms;


R4 is optionally substituted with at least one group selected independently from: 1) an alkyl, an alkoxy, a haloalkyl, a haloalkoxy, a cycloalkyl, an aryl, —COR9, —COR10, —CO2R10, —CONR102, —SO2R9, —SO2R10, —SO2NR102, —NR102, or a heteroaryl or heterocyclyl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO, any of which having up to 10 carbon atoms; or 2) halogen, cyano, or hydroxyl; and


wherein R10, in each occurrence, is selected independently from: 1) an alkyl or an aryl having up to 10 carbon atoms; or hydrogen.


In this aspect in the formula (III-Ai), Y1 and Y2 can be —(CH2)n- wherein n is 0, that is, R1 and R2 can be bonded directly to the pyridine core.


In another aspect, the present invention provides for compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


Y1 and Y2 are selected independently from >NR5 or —(CH2)n- wherein n is 0;


R5 is hydrogen or methyl;


wherein when Y1 or Y2 is independently >NR5;

    • 1) the corresponding Rz, wherein z is 1 or 2, in each occurrence is selected independently from: a) a substituted or an unsubstituted alkyl, aryl, cycloalkyl, or heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO, any of which having up to 10 carbon atoms; or b) hydrogen; or
    • 2) the corresponding YzRz, wherein z is 1 or 2, is selected independently from a substituted or an unsubstituted morpholinyl, piperazinyl, piperidinyl, or pyrrolidinyl;
    • wherein when YzRz is a piperazinyl, the piperazine nitrogen is optionally substituted by an alkyl, a cycloalkyl, an acyl, a haloalkyl, an alkoxyalkyl, SO2R7, or SO2NR72, any of which having up to 10 carbon atoms, wherein R7 is selected independently from: a) an alkyl or an aryl having up to 8 carbon atoms; or b) hydrogen;
    • wherein when YzRz is a piperidinyl or a pyrrolidinyl, the ring is optionally substituted by: a) an alkyl or a haloalkyl having up to 10 carbon atoms; or 2) hydroxyl;


wherein when Y1 or Y2 is independently —(CH2)n-, the corresponding Rz, wherein z is 1 or 2, in each occurrence is selected independently from a substituted or an unsubstituted alkyl, cycloalkyl, aryl, or heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO, any of which having up to 10 carbon atoms;


R4 is selected independently from: 1) a substituted or an unsubstituted aryl, or a substituted or an unsubstituted heteroaryl or heterocyclyl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO, any of which having up to 10 carbon atoms; or 2) Y1R1;


wherein any of R1 or R2, is also optionally substituted with at least one group independently selected from: 1) an alkyl, an alkoxy, a haloalkyl, a haloalkoxy, NR82, —COR9, —CO2R8, —OCOCH2CH2CO2R8, —CONR82, —SO2R9, —NHSO2R9, or —SO2NR82, any of which having up to 10 carbon atoms; or 2) hydroxyl, halogen, —OCH2O—, or cyano;


R8, in each occurrence, is selected independently from: 1) an alkyl, a haloalkyl, or an aryl, any of which having up to 10 carbon atoms; or 2) hydrogen;


R9, in each occurrence, is selected independently from an alkyl, a haloalkyl, an aryl, or a heterocyclyl or heteroaryl comprising at least one heteroatom selected from —O—, —S—, or >N—, any of which having up to 10 carbon atoms;


R4 is optionally substituted with at least one group selected independently from: 1) an alkyl, an alkoxy, a haloalkyl, a haloalkoxy, a cycloalkyl, an aryl, —COR9, —CO2R8, —CO2R10, —OCOCH2CH2CO2R10, —CONR82, —CONR102, —SO2R9, —SO2NR82, —SO2NR102, —NHSO2R9, —NR102, or a heteroaryl or heterocyclyl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO, any of which having up to 10 carbon atoms; or 2) halogen, —OCH2O—, cyano, or hydroxyl; and

    • R10, in each occurrence, is selected independently from: 1) an alkyl, an aryl, or a heterocyclyl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms; or 2) hydrogen.


Another aspect of the present invention provides for compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


Y1 and Y2 are independently selected from >NR5, —(CH2)n- wherein n is 0 or 1, —O—, >CO, or >SO2;


R5, in each occurrence, is selected independently from: 1) a substituted or an unsubstituted alkyl, aryl, alkoxyalkyl, cycloalkyl, or heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO, any of which having up to 10 carbon atoms; or 2) hydrogen;


wherein when Y1 or Y2 is independently >NR5;

    • 1) the corresponding Rz, wherein z is 1 or 2, in each occurrence is independently selected from: a) a substituted or an unsubstituted alkyl, aryl, alkoxyalkyl, cycloalkyl, —COR9, aralkyl, heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; any of which having up to 10 carbon atoms; or b) hydrogen; or
    • 2) the corresponding YzRz, wherein z is 1 or 2, is selected from a morpholinyl, a piperazinyl, or a piperidinyl; wherein when YzRz is piperazinyl, the piperazine nitrogen is optionally substituted by an alkyl, a cycloalkyl, an acyl, a haloalkyl, an alkoxyalkyl, SO2R7, SO2NR72, or CO2R7, wherein R7 is independently selected from: a) an alkyl or an aryl having up to 8 carbon atoms; or b) hydrogen;


wherein when Y1 or Y2 is independently —O—; the corresponding Rz, wherein z is 1 or 2, in each occurrence is independently selected from: 1) a substituted or an unsubstituted alkyl, aryl, cycloalkyl, —COR9, aralkyl, heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; any of which having up to 10 carbon atoms; or 2) hydrogen; and


wherein when Y1 or Y2 is independently —(CH2)n-, >CO, or >SO2; the corresponding Rz, wherein z is 1 or 2, in each occurrence is independently selected from: 1) a substituted or an unsubstituted alkyl, haloalkyl, cycloalkyl, —COR9, aralkyl, alkoxy, aryl, or heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; any of which having up to 10 carbon atoms; or 2) hydrogen, halogen, cyano, or hydroxyl;


wherein R3 and R4 in each occurrence, are independently: 1) haloalkyl having less than 3 carbon atoms; 2) alkyl, haloalkoxy, aryl, cycloalkyl, heteroaryl, or heterocyclyl having up to 10 carbon atoms, wherein any heteroaryl or heterocyclyl comprises at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; 3) hydrogen; or 4) Y1R1;


wherein any of R1, R2, or R5 is optionally substituted with at least one group independently selected from: 1) alkyl, alkoxy, haloalkyl, haloalkoxy, —O—CH2—O—, —OCOR9, NR82, —SO2R9, or —SO2NR82, any of which having up to 10 carbon atoms; or 2) hydrogen, halogen, or cyano;


wherein any of R3 or R4 is optionally substituted with at least one group independently selected from alkyl, haloalkyl, —SO2R10, —SO2NR102, or —NR102, any of which having up to 10 carbon atoms; and


wherein R8, R9, and R10, in each occurrence, is independently: 1) an alkyl; an aryl; or a heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; having up to 8 carbon atoms; or 2) hydrogen; wherein each of R8, R9, and R10 are optionally substituted with: 1) an alkyl, an alkoxy, a carboxylic acid, or a carboxylic acid ester, any of which having up to 8 carbon atoms; 2) halogen; or 3) hydroxyl.


Another aspect of the present invention provides for compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


Y1 and Y2 are selected independently from >NR5 or —(CH2)n- wherein n is 0 or 1;


R5 is methyl or hydrogen;


wherein when Y1 or Y2 is independently >NR5;

    • 1) the corresponding Rz, wherein z is 1 or 2, in each occurrence is selected independently from: a) an alkyl, an aryl, or a heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO, any of which having up to 10 carbon atoms; or b) hydrogen; or
    • 2) the corresponding YzRz, wherein z is 1 or 2, is selected independently from a morpholinyl, a piperazinyl, a pyrrolidinyl, or a piperidinyl, wherein YzRz is optionally substituted with: a) an alkyl or an acyl having up to 10 carbon atoms; or b) hydroxyl;


wherein when Y1 or Y2 is independently —(CH2)n-, the corresponding Rz, wherein z is 1 or 2, in each occurrence is selected independently from: 1) a substituted or an unsubstituted aryl, or a substituted or an unsubstituted heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO, any of which having up to 10 carbon atoms; or 2) hydrogen, halogen, cyano, or hydroxyl;


wherein R1 and R2 are optionally substituted with at least one group independently selected from: 1) an alkyl, an alkoxy, a haloalkyl, a haloalkoxy, —OCOR9, NR82, —SO2R9, or —SO2NR82, any of which having up to 10 carbon atoms; or 2) hydrogen, —OCH2O—, halogen, or cyano;


R8 and R9, in each occurrence, are selected independently from: 1) an alkyl or an aryl having up to 10 carbon atoms; or 2) hydrogen; and


R4 is selected independently from: 1) an alkyl, an aryl, —COR5, a cycloalkyl, a haloalkoxy, or a heteroaryl or heterocyclyl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO, any of which having up to 10 carbon atoms; or 2) hydrogen.


In another aspect, the present invention encompasses compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


Y1 and Y2 are selected independently from >NR5, —(CH2)n- wherein n is 0 or 1, or —O—;


R5 is selected independently from: 1) an alkyl, an aryl, a cycloalkyl, or a heteroaryl or a heterocyclyl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; any of which having up to 10 carbon atoms; or 2) hydrogen;


wherein when Y1 or Y2 is independently >NR5;

    • 1) the corresponding Rz, wherein z is 1 or 2, in each occurrence is independently selected from: a) a substituted or an unsubstituted alkyl, aryl, cycloalkyl, —COR9, aralkyl, or heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; any of which having up to 10 carbon atoms; b) hydrogen; or c) halogen; or
    • 2) the corresponding YzRz, wherein z is 1 or 2, is selected from a morpholinyl, a piperazinyl, or a piperidinyl; wherein when YzRz is piperazinyl, the piperazine nitrogen is optionally substituted by an alkyl, a cycloalkyl, a haloalkyl, an alkoxyalkyl, any of which having up to 10 carbon atoms; 2) SO2R7, SO2NR72, or CO2R7, wherein R7 is independently selected from: a) an alkyl or an aryl having up to 8 carbon atoms; or b) hydrogen;


wherein when Y1 or Y2 is independently —(CH2)n- or —O—; the corresponding Rz, wherein z is 1 or 2, in each occurrence is independently selected from: 1) a substituted or an unsubstituted alkyl, aryl, cycloalkyl, —COR9, aralkyl, or heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; any of which having up to 10 carbon atoms; or 2) hydrogen;


wherein any of R1, R2, or R5 is optionally substituted with at least one group independently selected from: 1) an alkyl, an alkoxy, a haloalkyl, a haloalkoxy, —OCOR9, NR82, —SO2R9, or —SO2NR82, any of which having up to 10 carbon atoms; or 2) hydrogen, —OCH2O—, halogen, or cyano;


R8 and R9, in each occurrence, are selected independently from: 1) an alkyl, an aryl, or a heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO, any of which having up to 10 carbon atoms; or 2) hydrogen;


wherein R8 and R9 are optionally substituted with: 1) an alkyl, an alkoxy, a carboxylic acid, or a carboxylic acid ester, any of which having up to 8 carbon atoms; or 2) halogen or hydroxyl; and


wherein R3 and R4 are, in each occurrence, independently: 1) R1 or 2) Y1R1.


In still another aspect, the present invention encompasses compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


Y1 and Y2 are selected independently from >NR5, >CH2, or —O—;


R5 is selected independently from: 1) an alkyl, an aryl, a cycloalkyl, or a heteroaryl or a heterocyclyl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO, any of which having up to 10 carbon atoms; or 2) hydrogen;


wherein when Y1 or Y2 is independently >NR5;

    • 1) the corresponding Rz, wherein z is 1 or 2, in each occurrence is independently selected from: a) an alkyl, an aryl, a cycloalkyl, —COR9, aralkyl, or a heterocyclyl or a heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO, any of which having up to 10 carbon atoms; or b) halogen; or
    • 2) the corresponding YzRz, wherein z is 1 or 2, is selected from a morpholinyl, a piperazinyl, or a piperidinyl;


wherein when Y1 or Y2 is independently >CH2 or —O—; the corresponding Rz, wherein z is 1 or 2, in each occurrence is selected independently from: 1) a substituted or an unsubstituted alkyl, aryl, cycloalkyl, —COR9, aralkyl, or heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO, any of which having up to 10 carbon atoms; or 2) hydrogen; and


R3 and R4 are selected independently from: 1) R1 or 2) Y1R1.


In another aspect, the present invention encompasses compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


Y1 is selected from >NR5 or —(CH2)n-wherein n is 0 or 1;


R1 and R2 are selected independently from: a) a substituted or an unsubstituted alkyl, cycloalkyl, aryl, or heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >CO or >SO2, any of which having up to 10 carbon atoms; or b) hydrogen, halogen, or hydroxy;


R5 is an alkyl having up to 3 carbon atoms or hydrogen;


R4 is selected from: 1) a substituted or an unsubstituted alkyl, aryl, or heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, or >CO, any of which having up to 10 carbon atoms; or 2) hydrogen or halogen;


R1 and R2 are optionally substituted with at least one group independently selected from: 1) an alkyl, an alkoxy, a haloalkyl, a haloalkoxy, —COR9, —CO2R8, —CONR82, —SO2R9, —NHSO2R9, or —SO2NR82, any of which having up to 10 carbon atoms; or 2) halogen, —OCH2O—, hydroxyl, or cyano;


R8, in each occurrence, is selected independently from: 1) an alkyl, a haloalkyl, or an aryl, any of which having up to 10 carbon atoms; or 2) hydrogen;


R9, in each occurrence, is selected independently from an alkyl, a haloalkyl, an aryl, or a heterocyclyl or heteroaryl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms;


R4 is optionally substituted with at least one group selected independently selected from: 1) an alkyl, a haloalkoxy, an alkoxy, —COR9, —COR10, —CONR82,, —CO2R8, —SO2R9, —SO2R10, —SO2NR102, or —NR102, any of which having up to 10 carbon atoms; or 2) halogen or hydroxyl; and


R10, in each occurrence, is selected independently from: 1) an alkyl, an aryl, or a heterocyclyl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms; or 2) hydrogen.


In this aspect in the formula (III-C), Y1 can be —(CH2)n- wherein n is 0, that is, R1 can be bonded directly to the pyridine core. Also in this aspect in the formula (III-C), R5 can be methyl or hydrogen.


In still another aspect, the present invention encompasses compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


R1 and R2 are selected independently from: a) a substituted or an unsubstituted aryl or heteroaryl comprising at least one heteroatom selected from —O—, >N—, or —S—, any of which having up to 10 carbon atoms; or b) hydrogen, halogen, or hydroxy;


R4 is selected from: 1) a substituted or an unsubstituted alkyl, aryl, or heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, or >CO, any of which having up to 10 carbon atoms; or 2) hydrogen or halogen;


R1 and R2 are optionally substituted with at least one group independently selected from: 1) an alkyl, an alkoxy, a haloalkyl, a haloalkoxy, —COR9, —CONR82, SO2R9, —NHSO2R9, or —SO2NR82, any of which having up to 10 carbon atoms; or 2) halogen, —OCH2O—, cyano, or hydroxyl;


R8, in each occurrence, is selected independently from: 1) an alkyl, a haloalkyl, or an aryl having up to 10 carbon atoms; or 2) hydrogen;


R9, in each occurrence, is selected independently from an alkyl, a haloalkyl, an aryl, or a heterocyclyl or heteroaryl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms;


R4 is optionally substituted with at least one group independently selected from: 1) an alkyl, a haloalkoxy, an alkoxy, —COR10, —CONR82,, —SO2R10, —SO2NR102, or —NR102, any of which having up to 10 carbon atoms; or 2) halogen or hydroxyl; and


R10, in each occurrence, is selected independently from: 1) an alkyl, an aryl, or a heterocyclyl comprising at least one heteroatom selected from —O— or >N—, or 2) hydrogen.


Another aspect of the present invention provides compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


R1 and R2 are selected independently from: 1) a substituted or an unsubstituted aryl or a substituted or an unsubstituted heteroaryl or heterocyclyl comprising at least one heteroatom selected from —O—, >N—, or —S—, any of which having up to 10 carbon atoms; or 2) hydrogen, halogen, or hydroxy;


R4 is selected from a substituted or an unsubstituted aryl or heteroaryl comprising at least one heteroatom selected from —O—, >N—, or —S—, any of which having up to 10 carbon atoms;


R5 is an alkyl having up to 3 carbon atoms or hydrogen;


R1 and R2 are optionally substituted with at least one group independently selected from: 1) an alkyl, an alkoxy, a haloalkyl, a haloalkoxy, —COR9, —CO2R8, —CONR82, —SO2R9, —NHSO2R9, or —SO2NR82, any of which having up to 10 carbon atoms; or 2) halogen, —OCH2O—, or hydroxyl;


R8, in each occurrence, is selected independently from: 1) an alkyl, a haloalkyl, or an aryl having up to 10 carbon atoms; or 2) hydrogen;


R9, in each occurrence, is selected independently from an alkyl, a haloalkyl, an aryl, or a heterocyclyl or heteroaryl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms;


R4 is optionally substituted with at least one group independently selected from: 1) an alkyl, a haloalkoxy, an alkoxy, —COR9, —COR10, —CO2R8, —CONR82,, —SO2R9, —SO2R10, —SO2NR102, or —NR102, any of which having up to 10 carbon atoms; or 2) halogen or hydroxyl; and


R10, in each occurrence, is selected independently from: 1) an alkyl, an aryl, or a heterocyclyl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms; or 2) hydrogen.


In this aspect of formula (III-H), R5 can be methyl or hydrogen.


Another aspect of the present invention provides compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


R1 and R2 are selected independently from: a) a substituted or an unsubstituted aryl or heteroaryl comprising at least one heteroatom selected from —O—, >N—, or —S—, any of which having up to 10 carbon atoms; or b) hydrogen, halogen, or hydroxy;


R4 is selected from a substituted or an unsubstituted alkyl or heterocyclyl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, or >CO, any of which having up to 10 carbon atoms;


R1 and R2 are optionally substituted with at least one group independently selected from: 1) an alkyl, an alkoxy, a haloalkyl, a haloalkoxy, —COR9, —CONR82, SO2R9, —NHSO2R9, or —SO2NR82, any of which having up to 10 carbon atoms; or 2) halogen, —OCH2O—, cyano, or hydroxyl;


R8, in each occurrence, is selected independently from: 1) an alkyl, a haloalkyl, or an aryl, any of which having up to 10 carbon atoms; or 2) hydrogen;


R9, in each occurrence, is selected independently from an alkyl, a haloalkyl, an aryl, or a heterocyclyl or heteroaryl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms;


R4 is optionally substituted with at least one group independently selected from: 1) an alkyl, a haloalkoxy, an alkoxy, —COR10, —CONR82,, —SO2R10, —SO10NR2, or —NR102, any of which having up to 10 carbon atoms; or 2) halogen or hydroxyl; and


R10, in each occurrence, is selected independently from: 1) an alkyl, an aryl, or a heterocyclyl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms; or 2) hydrogen.


Still another aspect of the present invention provides compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


R4 is selected from: 1) a substituted or an unsubstituted heterocyclyl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO,


n and m are independently an integer from 0 to 3, inclusive;


R11 and R12, in each occurrence, are selected independently from: 1) an alkyl, an alkoxy, a haloalkyl, a haloalkoxy, —COR9, —CONR82, —CO2R8, —CO2R9, —SO2R9, —NHSO2R9, or —SO2NR82, any of which having up to 10 carbon atoms; or 2) halogen, —OCH2O—, cyano, or hydroxyl;


R8, in each occurrence, is selected independently from: 1) an alkyl, a haloalkyl, or an aryl, any of which having up to 10 carbon atoms; or 2) hydrogen;


R9, in each occurrence, is selected independently from an alkyl, a haloalkyl, an aryl, or a heterocyclyl or heteroaryl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms;


R4 is optionally substituted with at least one group selected independently from: 1) an alkyl, a linear alkyl, a branched alkyl, a cycloalkyl, —COR10, —CONR82,, —OCOCH2CH2CO2R10, —SO2R10, or —SO2NR102, any of which having up to 10 carbon atoms; or 2) hydroxyl; and


R10, in each occurrence, is selected independently from: 1) an alkyl, an aryl, or a heterocyclyl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms; or 2) hydrogen.


In this aspect of the formula (III-I), R4 can be selected from
embedded imageembedded image

wherein X is selected from CH2, O, NH, NMe, NEt, S, SO2, CH(OCOCH2CH2CO2H), or CH(OH);


n and m are independently an integer from 0 to 2, inclusive; and


R11 and R12, in each occurrence, are selected independently from OCF3, OMe, Cl, F, SO2Me, CF3, Me, COMe, CONHMe, NHSO2Me, SO2NH2, SO2NHMe, SO2NMe2, CONH2, CONMe2, CO2Me, —OCH2O—, or OH.


Yet another, aspect of the present invention provides compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


R4 is selected from: 1) a substituted or an unsubstituted aryl, alkoxy, or heteroaryl comprising at least one heteratom selected from —O—, —S—, or >N—, any of which having up to 10 carbon atoms; or 2) hydrogen, chloro, or hydroxyl;


n and m are independently an integer from 0 to 3, inclusive;


R11 and R12, in each occurrence, are selected independently from: 1) an alkyl, an alkoxy, a haloalkyl, a haloalkoxy, —COR9, —CONR82, —CO2R8, —CO2R9, —SO2R9, —NHSO2R9, or —SO2NR82, any of which having up to 10 carbon atoms; or 2) halogen, —OCH2O—, cyano, or hydroxyl;


R8, in each occurrence, is selected independently from: 1) an alkyl, a haloalkyl, or an aryl, any of which having up to 10 carbon atoms; or 2) hydrogen;


R9, in each occurrence, is selected independently from an alkyl, a haloalkyl, an aryl, or a heterocyclyl or heteroaryl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms;


wherein when R4 is optionally substituted with at least one group selected independently selected from: 1) an alkyl, a haloalkoxy, an alkoxy, —COR9, —COR10, —CONR82,, —SO2R9, —SO2R10, —SO2NR102, or —NR102, any of which having up to 10 carbon atoms; or 2) halogen, cyano, or hydroxyl; and


R10, in each occurrence, is selected independently from: 1) an alkyl, an aryl, or a heterocyclyl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms; or 2) hydrogen.


Still another aspect of the present invention provides compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


m, n, and p are independently an integer from 0 to 3, inclusive;


R11, R12 and R13, in each occurrence, are selected independently from: 1) an alkyl, an alkoxy, a haloalkyl, a haloalkoxy, —COR9, —CONR82, —CO2R8, —SO2R9, —NHSO2R9, or —SO2NR82, any of which having up to 10 carbon atoms; or 2) halogen, —OCH2O—, or hydroxyl;


R8, in each occurrence, is selected independently from: 1) an alkyl, a haloalkyl, or an aryl, any of which having up to 10 carbon atoms; or 2) hydrogen; and


R9, in each occurrence, is selected independently from an alkyl, a haloalkyl, an aryl, or a heterocyclyl or heteroaryl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms.


In this aspect of the formula (II-N),


n, m and p can be independently an integer from 0 to 2, inclusive; and


R11, R12 and R13, in each occurrence, can be selected independently from OCF3, OMe, Cl, F, SO2Me, CF3, Me, COMe, CONHMe, NHSO2Me, SO2NH2, SO2NHMe, SO2NMe2, CONH2, CONMe2, CO2Me, —OCH2O—, or OH.


Another aspect of the present invention provides compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


Y2 is selected from >NR5 or >(CH2)n wherein n is 0 or 1;


R1 and R2 are selected independently from: a) a substituted or an unsubstituted alkyl, cycloalkyl, aryl, or heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >CO or >SO2, any of which having up to 10 carbon atoms; or b) hydrogen, halogen, or hydroxy;


R5 is selected from an alkyl having up to 3 carbon atoms or hydrogen;


R4 is selected from: 1) a substituted or an unsubstituted alkyl, aryl, or heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, or >CO, any of which having up 10 carbon atoms; or 2) hydrogen or halogen;


R1 and R2 are optionally substituted with at least one group independently selected from: 1) an alkyl, an alkoxy, a haloalkyl, a haloalkoxy, —COR9, —CO2R8, —CONR82, —SO2R9, —NHSO2R9, or —SO2NR82, any of which having up to 10 carbon atoms; or 2) halogen, —OCH2O—, cyano, or hydroxyl;


R8, in each occurrence, is selected independently from: 1) an alkyl, a haloalkyl, or an aryl, any of which having up to 10 carbon atoms; or 2) hydrogen;


R9, in each occurrence, is selected independently from an alkyl, a haloalkyl, an aryl, or a heterocyclyl or heteroaryl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms;


R4 is optionally substituted with at least one group independently selected from: 1) an alkyl, a haloalkoxy, an alkoxy, —COR9, —COR10, —CONR82,, —SO2R9, —SO2R10, —SO2NR102, or —NR102, any of which having up to 10 carbon atoms; or 2) halogen, cyano, or hydroxyl; and


R10, in each occurrence, is selected independently from: 1) an alkyl, an aryl, or a heterocyclyl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms; or 2) hydrogen. In this aspect in the formula (III-D), Y2 can be —(CH2)n- wherein n is 0, that is, R2 can be bonded directly to the pyridine core.


Yet another aspect of the present invention provides compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


R1 and R2 are selected independently from: a) a substituted or an unsubstituted aryl or a substituted or an unsubstituted heteroaryl comprising at least one heteroatom selected from —O—, >N—, or —S—, any of which having up to 10 carbon atoms; or b) hydrogen, halogen, or hydroxyl;


R4 is selected from: 1) a substituted or an unsubstituted alkyl, aryl, or heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, or >CO, any of which having up to 10 carbon atoms; or 2) hydrogen or halogen;


R1 and R2 are optionally substituted with at least one group independently selected from: 1) an alkyl, an alkoxy, a haloalkyl, a haloalkoxy, —COR9, —CONR82, —SO2R9, —NHSO2R9, or —SO2NR82, any of which having up to 10 carbon atoms; or 2) halogen, —OCH2O—, cyano, or hydroxyl;


R8, in each occurrence, is selected independently from: 1) an alkyl, a haloalkyl, or an aryl, any of which having up to 10 carbon atoms; or 2) hydrogen;


R9, in each occurrence, is selected independently from an alkyl, a haloalkyl, an aryl, or a heterocyclyl or heteroaryl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms;


R4 is optionally substituted with at least one group independently selected from: 1) an alkyl, a haloalkoxy, an alkoxy, —COR9, —COR10, —CONR82,, —SO2R9, —SO2R10, —SO2NR102, or —NR102, any of which having up to 10 carbon atoms; or 2) halogen or hydroxyl; and


R10, in each occurrence, is selected independently from: 1) an alkyl, an aryl, or a heterocyclyl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms; or 2) hydrogen.


Yet another aspect of the present invention provides compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


R1 and R2 are, in each occurrence, selected independently from: 1) a substituted or an unsubstituted aryl or a substituted or an unsubstituted heteroaryl comprising at least one heteroatom selected from —O—, >N—, or —S—, any of which having up to 10 carbon atoms; or b) hydrogen, halogen, or hydroxyl;


R4 is selected from a substituted or an unsubstituted aryl or a substituted or an unsubstituted heteroaryl comprising at least one heteroatom selected from —O—, >N—, or —S—, any of which having up to 10 carbon atoms;


R5 is selected from an alkyl having up to 3 carbon atoms or hydrogen;


R1 and R2 are optionally substituted with at least one group independently selected from: 1) an alkyl, an alkoxy, a haloalkyl, a haloalkoxy, —COR9, —CONR82, —SO2R9, —NHSO2R9, or —SO2NR82, any of which having up to 10 carbon atoms; or 2) halogen, —OCH2O—, or hydroxyl;


R8, in each occurrence, is selected independently from: 1) an alkyl, a haloalkyl, or an aryl, any of which having up to 10 carbon atoms; or 2) hydrogen;


R9, in each occurrence, is selected independently from an alkyl, a haloalkyl, an aryl, or a heterocyclyl or heteroaryl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms;


R4 is optionally substituted with at least one group independently selected from: 1) an alkyl, a haloalkoxy, an alkoxy, —COR10, —CONR82,, —SO2R10, —SO2NR102, or —NR102, any of which having up to 10 carbon atoms; or 2) halogen or hydroxyl; and


R10, in each occurrence, is selected independently: 1) an alkyl, an aryl, or a heterocyclyl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms; or 2) hydrogen.


In one aspect, the present invention encompasses compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


R1 and R2 are selected independently from: 1) a substituted or an unsubstituted aryl, or a substituted or an unsubstituted heteroaryl comprising at least one heteroatom selected from —O—, >N—, or —S—, any of which having up to 10 carbon atoms; or 2) hydrogen, halogen, or hydroxy;


R4 is selected from a substituted or an unsubstituted alkyl, or a substituted or an unsubstituted heterocyclyl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, or >CO, any of which having up to 10 carbon atoms;


R1 and R2 are optionally substituted with at least one group independently selected from: 1) an alkyl, an alkoxy, a haloalkyl, a haloalkoxy, —COR9, —CONR82, —SO2R9, —NHSO2R9, or —SO2NR82, any of which having up to 10 carbon atoms; or 2) halogen, —OCH2O—, or hydroxyl;


R8, in each occurrence, is selected independently from: 1) an alkyl, a haloalkyl, or an aryl, any of which having up to 10 carbon atoms; or 2) hydrogen;


R9, in each occurrence, is selected independently from an alkyl, a haloalkyl, an aryl, or a heterocyclyl or heteroaryl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms;


R4 is optionally substituted with at least one group independently selected from: 1) an alkyl, a haloalkoxy, an alkoxy, —COR10, —CONR82,, —SO2R10, —SO2NR102, or —NR102, any of which having up to 10 carbon atoms; or 2) halogen or hydroxyl; and


R10, in each occurrence, is selected independently from: 1) an alkyl, an aryl, or a heterocyclyl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms; or 2) hydrogen.


In another aspect, the present invention encompasses compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


R4 is selected from a substituted or an unsubstituted heterocyclyl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, or >CO, any of which having up to 10 carbon atoms;


n and m are independently an integer from 0 to 3, inclusive;


R11 and R12, in each occurrence, are selected independently from: 1) an alkyl, an alkoxy, a haloalkyl, a haloalkoxy, —COR9, —CONR82, —CO2R8, —CO2R9, —SO2R9, —NHSO2R9, or —SO2NR82, any of which having up to 10 carbon atoms; or 2) halogen, —OCH2O—, cyano, or hydroxyl;


R8, in each occurrence, is selected independently from: 1) an alkyl, a haloalkyl, or an aryl, any of which having up to 10 carbon atoms; or 2) hydrogen;


R9, in each occurrence, is selected independently from an alkyl, a haloalkyl, an aryl, or a heterocyclyl or heteroaryl comprising at least one heteroatom selected from —O— or>N—, any of which having up to 10 carbon atoms;


R4 is optionally substituted with at least one group independently selected from: 1) an alkyl, a branched alkyl, a linear alkyl, a cycloalkyl, —COR10, —CONR82,, —SO2R10, or —SO2NR102, any of which having up to 10 carbon atoms; or 2) hydroxyl;


and


R10, in each occurrence, is selected independently from: 1) an alkyl, an aryl, or a heterocyclyl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms; or 2) hydrogen.


In another aspect, the present invention encompasses compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


R4 is selected from: 1) a substituted or an unsubstituted aryl, alkoxy, or heteroaryl comprising at least one heteroatom selected from —O—, —S—, or >N—, any of which having up to 10 carbon atoms; or 2) hydrogen, chloro, or hydroxyl;


n and m are independently an integer from 0 to 3, inclusive;


R11 and R12, in each occurrence, are selected independently from: 1) an alkyl, an alkoxy, a haloalkyl, a haloalkoxy, —COR9, —CONR82, —SO2R9, —NHSO2R9, or —SO2NR82, any of which having up to 10 carbon atoms; or 2) halogen, —OCH2O—, or hydroxyl;


R8, in each occurrence, is selected independently from: 1) an alkyl, a haloalkyl, or an aryl, any of which having up to 10 carbon atoms; or 2) hydrogen;


R9, in each occurrence, is selected independently from an alkyl, a haloalkyl, an aryl, or a heterocyclyl or heteroaryl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms;


R4 is optionally substituted with at least one group independently selected from: 1) an alkyl, a haloalkoxy, an alkoxy, —COR9, —COR10, —CONR82,, —SO2R9, —SO2R10, —SO2NR102, or —NR102, any of which having up to 10 carbon atoms; or 2) halogen or hydroxyl; and


R10, in each occurrence, is selected independently from: 1) an alkyl, an aryl, or a heterocyclyl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms; or 2) hydrogen.


In another aspect, the present invention encompasses compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


R1 is selected from a substituted or an unsubstituted heterocyclyl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO, any of which having up to 10 carbon atoms;


n and m are independently an integer from 0 to 3, inclusive;


R11 and R12, in each occurrence, are selected independently from: 1) an alkyl an alkoxy, a haloalkyl, a haloalkoxy, —COR9, —CO2R8, —CONR82, —SO2R9, —NHSO2R9, or —SO2NR82, any of which having up to 10 carbon atoms; or 2) halogen, —OCH2O—, cyano, or hydroxyl;


R8, in each occurrence, is selected independently from: 1) an alkyl, a haloalkyl, or an aryl, any of which having up to 10 carbon atoms; or 2) hydrogen;


R9, in each occurrence, is selected independently from an alkyl, a haloalkyl, an aryl, or a heterocyclyl or heteroaryl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms;


R1 is optionally substituted with at least one group independently selected from: 1) an alkyl, a branched alkyl, a linear alkyl, a cycloalkyl, a haloalkyl, —COR10, —CONR82,, —OCOCH2CH2CO2R10, —SO2R10, or —SO2NR102, any of which having up to 10 carbon atoms; or 2) hydroxyl; and


R10, in each occurrence, is selected independently from: 1) an alkyl, an aryl, or a heterocyclyl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms; or 2) hydrogen.


In this aspect of the formula (III-M), R1 can be selected from or
embedded imageembedded image

wherein X is selected from CH2, O, NH, NMe, NEt, S, SO2, CH(OCOCH2CH2CO2H), or CH(OH);


n and m are independently an integer from 0 to 2, inclusive; and


R11 and R12, in each occurrence, are selected independently from OCF3, OMe, Cl, F, SO2Me, CF3, Me, COMe, CONHMe, NHSO2Me, SO2NH2, SO2NHMe, SO2NMe2, CONH2, CONMe2, CO2Me, —OCH2O—, or OH.


In another aspect, the present invention encompasses compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


R1 is selected from a substituted or an unsubstituted heterocyclyl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO, any of which having up to 10 carbon atoms;


n and m are independently an integer from 0 to 3, inclusive;


R11 and R12, in each occurrence, are selected independently from: 1) an alkyl, an alkoxy, a haloalkyl, a haloalkoxy, —COR9, —CO2R8, —CONR82, —SO2R9, —NHSO2R9, or —SO2NR82, any of which having up to 10 carbon atoms; or 2) halogen, cyano, —OCH2O—, or hydroxyl;


R8, in each occurrence, is selected independently from: 1) an alkyl, a haloalkyl, or an aryl, any of which having up to 10 carbon atoms; or 2) hydrogen;


R9, in each occurrence, is selected independently from an alkyl, a haloalkyl, an aryl, or a heterocyclyl or heteroaryl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms; and


R1 is optionally substituted with at least one group independently selected from: 1) an alkyl, a cycloalkyl, a haloalkyl, or —OCOCH2CH2CO2R8, any of which having up to 10 carbon atoms; or 2) hydroxyl.


In this aspect of the formula (III-O), R1 can be selected from
embedded imageembedded image

wherein X is selected from CH2, O, NH, NMe, NEt, S, SO2, CH(OCOCH2CH2CO2H), or CH(OH);


n and m can be independently an integer from 0 to 2, inclusive; and


R11 and R12, in each occurrence, can be selected independently from OCF3, OMe, Cl, F, SO2Me, CF3, Me, COMe, CONHMe, NHSO2Me, SO2NH2, SO2NHMe, SO2NMe2, CONH2, CONMe2, CO2Me, —OCH2O—, or OH.


In yet another aspect, the present invention encompasses compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


R1 is selected from a substituted or an unsubstituted heterocyclyl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO, any of which having up to 10 carbon atoms;


n and m are independently an integer from 0 to 3, inclusive;


R11 and R12, in each occurrence, are selected independently from: 1) an alkyl, an alkoxy, a haloalkyl, a haloalkoxy, —COR9, —CO2R8, —CONR82, —SO2R9, —NHSO2R9, or —SO2NR82, any of which having up to 10 carbon atoms; or 2) halogen, cyano, —OCH2O—, or hydroxyl;


R8, in each occurrence, is selected independently from: 1) an alkyl, a haloalkyl, or an aryl, any of which having up to 10 carbon atoms; or 2) hydrogen;


R9, in each occurrence, is selected independently from an alkyl, a haloalkyl, an aryl, or a heterocyclyl or heteroaryl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms; and


R1 is optionally substituted with at least one group independently selected from: 1) an alkyl, a cycloalkyl, a haloalkyl, or —OCOCH2CH2CO2R8, any of which having up to 10 carbon atoms; or 2) hydroxyl.


In this aspect of the formula (III-P), R1 can be selected from
embedded imageembedded image

wherein X is selected from CH2, O, NH, NMe, NEt, S, SO2, CH(OCOCH2CH2CO2H), or CH(OH);


n and m can be independently an integer from 0 to 2, inclusive; and


R11 and R12, in each occurrence, can be selected independently from OCF3, OMe, Cl, F, SO2Me, CF3, Me, COMe, CONHMe, NHSO2Me, SO2NH2, SO2NHMe, SO2NMe2, CONH2, CONMe2, CO2Me, —OCH2O—, or OH.


In still another aspect, the present invention encompasses compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


m, n and p are independently an integer from 0 to 3, inclusive;


R11, R12 and R13, in each occurrence, are selected independently from: 1) an alkyl, an alkoxy, a haloalkyl, a haloalkoxy, —COR9, —CONR82, —CO2R8, —SO2R9, —NHSO2R9, or —SO2NR82, any of which having up to 10 carbon atoms; or 2) halogen, —OCH2O—, or hydroxyl;


R8, in each occurrence, is selected independently from: 1) an alkyl, a haloalkyl, or an aryl, any of which having up to 10 carbon atoms; or 2) hydrogen; and


R9, in each occurrence, is selected independently from an alkyl, a haloalkyl, an aryl, or a heterocyclyl or heteroaryl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms.


In this aspect of the formula (III-Q), n and m can be independently an integer from 0 to 2, inclusive; and R11 and R12, in each occurrence, can be selected independently from OCF3, OMe, Cl, F, SO2Me, CF3, Me, COMe, CONHMe, NHSO2Me, SO2NH2, SO2NHMe, SO2NMe2, CONH2, CONMe2, CO2Me, —OCH2O—, or OH.


Yet another aspect of this invention encompasses compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


Y1 and Y2, in each occurrence, are independently selected from —O—, —S—, >NR5, or >CH2;


R1 and R2 are, in each occurrence, selected independently from: a) a substituted or an unsubstituted alkyl, cycloalkyl, aryl, heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >CO or >SO2, any of which having up to 10 carbon atoms; b) hydrogen, or c) halogen;


R5 is an alkyl having up to 10 carbon atoms or hydrogen;


R3 is hydrogen;


R4 is selected from: 1) a substituted or an unsubstituted alkyl, aryl, heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, or >CO, 2) hydrogen; or 3) halogen;


R1 and R2 are optionally substituted with at least one group independently selected from: 1) alkyl, alkoxy, haloalkyl, haloalkoxy, —OCH2O—, —COR9, —OCOR9, —CON(R8)2, —(CH2)bCO2R8 wherein b is an integer from 0 to 3, —SO2R9, —NHSO2R9, or —SO2N(R8)2, any of which having up to 10 carbon atoms; 2) halogen; or 3) or hydroxyl;


R8, in each occurrence, is selected independently from: 1) an alkyl, a haloalkyl, or an aryl having up to 10 carbon atoms; or 2) hydrogen;


R9, in each occurrence, is independently an alkyl, a haloalkyl, an aryl, a heterocyclyl or heteroaryl comprising at least one heteroatom selected from —O— or >N—; wherein R9 is optionally substituted with: 1) an alkyl, an alkoxy, a carboxylic acid, or a carboxylic acid ester, any of which having up to 8 carbon atoms; 2) halogen; or 3) hydroxyl;


R4 is optionally substituted with at least one group independently selected from: 1) alkyl, haloalkoxy, alkoxy, —COR10, —CON(R8)2, —SR10, —SO2R10, —SO2N(R10)2, or —N(R10)2, any of which having up to 10 carbon atoms; 2) halogen; or 3) hydroxyl; and


R10, in each occurrence, is independently: 1) an alkyl, an aryl, or a heterocyclyl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms; or 2) hydrogen.


Still another aspect of this invention encompasses compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


R1 and R2 are, in each occurrence, selected independently from a substituted or an unsubstituted cycloalkyl; aryl; aralkyl; heterocyclyl or heteroaryl comprising at least one heteroatom selected from —O—, >N—, or —S—; any of which having up to 10 carbon atoms;


R3 is hydrogen;


R4 is a substituted or unsubstituted heterocyclyl or heteroaryl comprising a nitrogen atom directly bonded to the pyridine ring and having up to 10 carbon atoms, wherein the heterocyclyl or heteroaryl optionally comprises at least one additional heteroatom selected from —O—, >N— or —S—;


R1 and R2 are optionally substituted with at least one group independently selected from: 1) alkyl, alkoxy, haloalkyl, haloalkoxy, —COR9, —OCOR9, —CON(R8)2, —SO2R9, —NHSO2R9, or —SO2N(R8)2, any of which having up to 10 carbon atoms; 2) halogen; or 3) or hydroxyl; and


R4 is optionally substituted with at least one group independently selected from: 1) haloalkoxy, —COR10, —CON(R8)2, —SO2R10, or —SO2N(R10)2, any of which having up to 10 carbon atoms; 2) halogen; or 3) hydroxyl.


R8, in each occurrence, is selected independently from: 1) an alkyl, a haloalkyl, or an aryl having up to 10 carbon atoms; or 2) hydrogen;


R9, in each occurrence, is independently an alkyl, a haloalkyl, an aryl, a heterocyclyl or heteroaryl comprising at least one heteroatom selected from —O— or >N—; wherein R9 is optionally substituted with: 1) an alkyl, an alkoxy, a carboxylic acid, or a carboxylic acid ester, any of which having up to 8 carbon atoms; 2) halogen; or 3) hydroxyl;


R10, in each occurrence, is independently: 1) an alkyl, an aryl, or a heterocyclyl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms; or 2) hydrogen.


Still another aspect of this invention encompasses compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


Y1 is >CH2;


R1 and R2 are, in each occurrence, selected independently from a substituted or an unsubstituted aryl, heterocyclyl, or heteroaryl, any of which having up to 10 carbon atoms, wherein the heterocyclyl and heteroaryl comprise at least one heteroatom selected from —O—, >N—, or —S—;


R4 is a substituted or an unsubstituted aryl having up to 10 carbon atoms;


R1 and R2 are optionally substituted with at least one group independently selected from: 1) alkoxy, haloalkoxy, —COR9, —CON(R8)2, —SO2R9, —NHSO2R9, or —SO2N(R8)2, any of which having up to 10 carbon atoms; or 2) halogen;


R4 is optionally substituted with at least one group independently selected from: 1) haloalkoxy, —COR10, —CON(R8)2, —SO2R10, or —SO2N(R10)2, any of which having up to 10 carbon atoms; 2) halogen; or 3) hydroxyl; and


R8, in each occurrence, is selected independently from: 1) an alkyl, a haloalkyl, or an aryl having up to 10 carbon atoms; or 2) hydrogen;


R9, in each occurrence, is selected independently from an alkyl, a haloalkyl, an aryl, or a heteroaryl comprising at least one heteroatom selected from —O— or >N—; any of which having up to 10 carbon atoms; and


R10, in each occurrence, is selected independently from: 1) an alkyl, an aryl, or a heterocyclyl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms; or 2) hydrogen.


Yet a further aspect of this invention encompasses compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


Y1 is >CH2;


R1 is a substituted or unsubstituted aryl or heterocyclyl having up to 10 carbon atoms, wherein the heterocyclyl comprises a nitrogen atom directly bonded to the pyridine ring and optionally comprising at least one additional heteroatom selected from —O—, >N— or —S—;


R2 is a substituted or an unsubstituted aryl having up to 10 carbon atoms;


R4 is a substituted or an unsubstituted aryl, heterocyclyl, or heteroaryl, any of which having up to 10 carbon atoms, wherein the heterocyclyl and heteroaryl comprise at least one heteroatom selected from —O—, >N—, or —S—;


R1 and R2 are optionally substituted with at least one group independently selected from: 1) alkoxy, haloalkoxy, haloalkyl, —COR9, —CON(R8)2, —SO2R9, — or —SO2N(R8)2, any of which having up to 10 carbon atoms; or 2) halogen;


R4 is optionally substituted with at least one group independently selected from: 1) haloalkoxy, —COR10, —CON(R8)2, —SO2R10, or —SO2N(R10)2, any of which having up to 10 carbon atoms; 2) halogen; or 3) hydroxyl; and


R8, in each occurrence, is selected independently from: 1) an alkyl, a haloalkyl, or an aryl having up to 10 carbon atoms; or 2) hydrogen;


R9, in each occurrence, is selected independently from an alkyl, a haloalkyl, an aryl, or a heteroaryl comprising at least one heteroatom selected from —O— or>N—; any of which having up to 10 carbon atoms; and


R10, in each occurrence, is selected independently from: 1) an alkyl, an aryl, or a heterocyclyl comprising at least one heteroatom selected from —O— or>N—, any of which having up to 10 carbon atoms; or 2) hydrogen.


Yet another aspect of this invention encompasses compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


Y2 is >CH2;


R1 and R2 are, in each occurrence, selected independently from a substituted or an unsubstituted aryl, heterocyclyl, or heteroaryl, any of which having up to 10 carbon atoms, wherein the heterocyclyl and heteroaryl comprise at least one heteroatom selected from —O—, >N—, or —S—;


R4 is a substituted or an unsubstituted aryl having up to 10 carbon atoms;


R1 and R2 are optionally substituted with at least one group independently selected from: 1) alkoxy, haloalkoxy, —COR9, —CON(R8)2, —SO2R9, —NHSO2R9, or —SO2N(R8)2, any of which having up to 10 carbon atoms; or 2) halogen;


R4 is optionally substituted with at least one group independently selected from: 1) haloalkoxy, —COR10, —CON(R8)2, —SO2R10, or —SO2N(R10)2, any of which having up to 10 carbon atoms; 2) halogen; or 3) hydroxyl; and


R8, in each occurrence, is selected independently from: 1) an alkyl, a haloalkyl, or an aryl having up to 10 carbon atoms; or 2) hydrogen;


R9, in each occurrence, is selected independently from an alkyl, a haloalkyl, an aryl, or a heteroaryl comprising at least one heteroatom selected from —O— or >N—; any of which having up to 10 carbon atoms; and


R10, in each occurrence, is selected independently from: 1) an alkyl, an aryl, or a heterocyclyl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms; or 2) hydrogen.


Yet a further aspect of this invention encompasses compounds and compositions comprising these compounds, wherein the compounds have the following formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof;


wherein:


Y2 is >CH2;


R1 is a substituted or unsubstituted aryl or heterocyclyl having up to 10 carbon atoms, wherein the heterocyclyl comprises a nitrogen atom directly bonded to the pyridine ring and optionally comprising at least one additional heteroatom selected from —O—, >N— or —S—;


R2 is a substituted or an unsubstituted aryl having up to 10 carbon atoms;


R4 is a substituted or an unsubstituted aryl, heterocyclyl, or heteroaryl, any of which having up to 10 carbon atoms, wherein the heterocyclyl and heteroaryl comprise at least one heteroatom selected from —O—, >N—, or —S—;


R1 and R2 are optionally substituted with at least one group independently selected from: 1) alkoxy, haloalkoxy, haloalkyl, —COR9, —CON(R8)2, —SO2R9, — or —SO2N(R8)2, any of which having up to 10 carbon atoms; or 2) halogen;


R4 is optionally substituted with at least one group independently selected from: 1) haloalkoxy, —COR10, —CON(R8)2, —SO2R10, or —SO2N(R10)2, any of which having up to 10 carbon atoms; 2) halogen; or 3) hydroxyl; and


R8, in each occurrence, is selected independently from: 1) an alkyl, a haloalkyl, or an aryl having up to 10 carbon atoms; or 2) hydrogen;


R9, in each occurrence, is selected independently from an alkyl, a haloalkyl, an aryl, or a heteroaryl comprising at least one heteroatom selected from —O— or >N—; any of which having up to 10 carbon atoms; and


R10, in each occurrence, is selected independently from: 1) an alkyl, an aryl, or a heterocyclyl comprising at least one heteroatom selected from —O— or >N—, any of which having up to 10 carbon atoms; or 2) hydrogen.


DEFINITIONS

The groups defined for various symbols used in the formulas of this disclosure, as well as the optional substituents defined on those groups, may be defined in the detailed manner as follows. Further definitions related to the more biological aspects of this disclosure are provided further below in their respective sections. Unless otherwise specified, any recitation of the number of carbon atoms in a particular group is intended to refer to the unsubstituted “base” group, therefore, any substituent recited on a base group is described by its own definition, including its own limitation of the number of carbon atoms. Unless otherwise specified, all structural isomers of a given structure, for example, all enantiomers, diasteriomers, and regioisomers, are included within this definition.


The terms ‘halogen’ or ‘halo’ includes fluorine, chlorine, bromine, or iodine.


The term ‘alkyl’ group is used to refer to both linear and branched alkyl groups. Exemplary alkyl groups include, but are not limited to, methyl, ethyl, propyl, butyl, pentyl, pentyl, hexyl, heptyl, octyl, nonyl, or decyl, and the like. Unless otherwise specified, an alkyl group has from 1 to 10 carbon atoms. Also unless otherwise specified, all structural isomers of a given structure, for example, all enantiomers and all diasteriomers, are included within this definition. For example, unless otherwise specified, the term propyl is meant to include n-propyl and iso-propyl, while the term butyl is meant to include n-butyl, iso-butyl, t-butyl, sec-butyl, and so forth.


‘Haloalkyl’ is a group containing at least one halogen and an alkyl portion as define above. Unless otherwise specified, all structural isomers of a given structure, for example, all enantiomers and all diasteriomers, are included within this definition. Exemplary haloalkyl groups include fluoromethyl, chloromethyl, fluoroethyl, chloroethyl, trilfluoromethyl, and the like. Unless otherwise specified, a haloalkyl group has from 1 to 10 carbon atoms.


‘Acyl’ is used to refer to an H—CO— or an alkyl-CO— group, where alkyl is defined herein. Exemplary acyl groups include, but are not limited to, acetyl, propionyl, iso-propionyl, tert-butionyl, and the like.


‘Cycloalkyl’ group refers to a cyclic alkyl group which may be mono or polycyclic. Exemplary cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, and cyclodecyl. Unless otherwise specified, a cycloalkyl group has from 3 to 10 carbon atoms.


‘Alkoxy’ refers to an —O(alkyl) group, where alkyl is as defined above. Therefore, unless otherwise specified, all isomers of a given structure are included within a definition. Exemplary alkyl groups include methoxy, ethoxy, n-propoxy, iso-propoxy, n-butoxy, iso-butoxy, t-butoxy, and the like. Unless otherwise specified, an alkoxy group has from 1 to 10 carbon atoms.


‘Alkoxyalkyl’ is an alkyl group with an alkoxy substituent, where alkoxy and alkyl groups are as defined above. Exemplary alkoxyalkyl groups include methoxymethyl, methoxyethyl, methoxypropyl, ethoxymethyl, ethoxyethyl, propoxymethyl, isopropoxymethyl isopropoxyethyl, isopropoxypropyl, t-butoxymethyl, t-butoxyethyl, t-butoxypropyl, and the like. Unless otherwise specified, an alkoxyalkyl group typically has from 1 to 10 carbon atoms.


‘Haloalkoxy’ is an alkoxy group with a halo substituent, where alkoxy and halo groups are as defined above. Exemplary haloalkoxy groups include chloromethoxy, trichloroethoxy, trifloroethoxy, perfluoroethoxy (—OCF2CF3), trifluoro-t-butoxy, hexafluoro-t-butoxy, perfluoro-t-butoxy (—OC(CF3)3), and the like. Unless otherwise specified, an haloalkoxy group typically has from 1 to 10 carbon atoms.


‘Alkylthio’ refers to an —S(alkyl) goup, where alkyl group is as defined above. Exemplary alkyl groups include methylthio, ethylthio, propylthio, butylthio, iso-propylthio, iso-butylthio, and the like. Unless otherwise specified, an alkylthio group typically has from 1 to 10 carbon atoms.


‘Alkylsulfonyl’ refers to a —SO2(alkyl) group, where alkyl group is as defined above. Exemplary alkylsulfonyl groups include methylsulfonyl, ethylsulfonyl and the like. Unless otherwise specified, an alkylsulfonyl group typically has from 1 to 10 carbon atoms.


‘Alkenyl’ is an unsaturated aliphatic group containing a C═C double bond. Exemplary alkenyl groups include ethenyl, propenyl, prop-1-enyl, isopropenyl, butenyl, but-1-enyl, isobutenyl, pentenyl, pent-1-enyl, hexenyl, pent-2-enyl, 2-methyl-but-2-ene, 2-methyl-pent-2-enyl and the like. Unless otherwise specified, an alkenyl group typically has from 2 to 10 carbon atoms.


‘Alkynyl’ is an unsaturated aliphatic group containing a C≡C triple bond. Exemplary alkynyl groups include ethenyl, propynyl, prop-1-ynyl, butynyl, butaynyl and the like. Unless otherwise specified, an alkynyl group typically has from 2 to 10 carbon atoms.


‘Aryl’ is optionally substituted monocylic or polycyclic aromatic ring system of 6 to 14 carbon atoms. Exemplary groups include phenyl, naphthyl and the like. Unless otherwise specified, an aryl group typically has from 6 to 14 carbon atoms.


‘Aralkyl’ is an alkyl group with an aryl substituent, where alkyl and aryl groups are as defined above. Exemplary aralkyl groups include, but are not limited to, benzyl, phenethyl (for example, 2-phenethyl), phenylpropyl (for example, 3-phenylpropyl), naphthylmethyl (for example, 1-naphthylmethyl and 2-naphthylmethyl) and the like.


‘Heteroaryl’ is an aromatic monocyclic or polycyclic ring system of 4 to 10 carbon atoms, having at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >NH or NR, and the like, wherein R is a substituted or unstubstituted alkyl, aryl, or acyl, as defined herein. In this aspect, >NH or NR are considered to be included when the heteroatom or heterogroup can be >N—. Exemplary heteroaryl groups include as pyrazinyl, isothiazolyl, oxazolyl, pyrazolyl, pyrrolyl, pyridazinyl, thienopyrimidyl, furanyl, indolyl, isoindolyl, benzo[1,3]dioxolyl, 1,3-benzoxathiole, quinazolinyl, pyridyl, thiophenyl and the like. Unless otherwise specified, a heteroaryl group typically has from 4 to 10 carbon atoms. Moreover, the heteroaryl group can be bonded to the pyrimidine core structure at a ring carbon atom, or, if applicable for a N-substituted heteroaryl such as pyrrole, can be bonded to the pyrimidine core structure through the heteroatom that is formally deprotonated to form a direct heteroatom-pyrimdine ring bond.


‘Heterocyclyl’ is a non-aromatic saturated monocyclic or polycyclic ring system of 3 to 10 member having at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO. Exemplary heterocyclyl groups include aziridinyl, pyrrolidinyl, piperdinyl, piperazinyl, morpholinyl, thiomorpholinyl, thiazolidinyl, 1,3-dioxolanyl, 1,4-dioxanyl and the like. Unless otherwise specified, a heterocyclyl group typically has from 2 to 10 carbon atoms. A heterocyclyl group can be bonded through a heteroatom that is formally deprotonated or a heterocyclyl group can be bonded through a carbon atom of the heterocyclyl group.


‘Carboxylic acid or its derivatives’ may be amides or esters. Exemplary carboxylic acid groups include CONH2, CONHMe, CONMe2, CONHEt, CONEt2, CONHPh, COOH, COOCH3, COOC2H5 or COOC3H7.


‘Cyclic amines’ means nitrogen containing heteroaryl or heterocyclyl groups.


Accordingly, in one aspect, compounds according to the present invention can have the formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof, wherein:


A is selected from A1, A2, or A3, wherein:
embedded image


n is 0 or 1;


R1 is H or CH3;


X1 is H, F, Cl, OCH3, SO2H, SO2CH3, SO2NHCH3, C(O)NHCH3,, C(O)NHCH2CH3,, C(O)CH3, C(O)N(CH3)2, C(O)(NC4H8), or C(O)(NC5H10); and


X2 is H, F, CH3, OCH3, OCF3, SO2CH3, SO2NHCH3, C(O)CH3, C(O)(morpholino), or X1 and X2 form a fused 1,3-dioxolane ring; and
embedded image

wherein n is 0 or 1; and m is 5, 6, 7, or 8;


B is selected from A1, A2, A3, or I; and


C is selected from A1, A2, A3, —H, —Cl, or —Br.


In another aspect, compounds according to the present invention can have the formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof, wherein:


B is selected from A1, A2′, or A3, wherein:
embedded image


n is 0 or 1;


R1 is H or CH3;


X1 is H, F, Cl, OCH3, SO2H, SO2CH3, SO2NHCH3, C(O)NHCH3,, C(O)NHCH2CH3,, C(O)CH3, C(O)N(CH3)2, C(O)(NC4H8), or C(O)(NC5H10); and


X2 is H, F, CH3, OCH3, OCF3, SO2CH3, SO2NHCH3, C(O)CH3, C(O)(morpholino), or X1 and X2 form a fused 1,3-dioxolane ring;


wherein X1 and X2 are not concurrently H;


and
embedded image

wherein n is 0 or 1; and m is 5, 6, 7, or 8;


A and C are selected independently from A1, A2′, A3, —H, —Cl, or —Br.


In a further aspect, compounds according to the present invention can have the formula:
embedded image

according to claim 2;


or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof, wherein:


B is selected from
embedded image


n is 0 or 1;


X1 is H, F, OCH3, or SO2CH3; and


X2 is H, F, OCH3, or SO2CH3, or X1 and X2 form a fused 1,3-dioxolane ring;


wherein X1 and X2 are not concurrently H; and


A and C are selected independently from —H or —Cl.


In yet another aspect, compounds according to the present invention can have the formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof, wherein:


A and C are selected independently from A1, A2′, or A3, wherein:
embedded image


n is 0 or 1;


R1 is H or CH3;


X1 is H, F, Cl, OCH3, SO2H, SO2CH3, SO2NHCH3, C(O)NHCH3,, C(O)NHCH2CH3,, C(O)CH3, C(O)N(CH3)2, C(O)(NC4H8), or C(O)(NC5H10); and


X2 is H, F, CH3, OCH3, OCF3, SO2CH3, SO2NHCH3, C(O)CH3, C(O)(morpholino), or X1 and X2 form a fused 1,3-dioxolane ring;


wherein X1 and X2 are not concurrently H;


and
embedded image

wherein n is 0 or 1; and m is 5, 6, 7, or 8; and


B is selected from A1, A2′, A3, —H or —I.


In still a further aspect, compounds according to the present invention can have the formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof, wherein:
embedded imageembedded imageembedded image


In another aspect, compounds according to the present invention can have the formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof, wherein:
embedded imageembedded imageembedded image


In a different further aspect, compounds according to the present invention can have the formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof, wherein:


A is selected from A1 or A2, and


B and C are selected from A2, wherein:
embedded image


n is 0 or 1;


X1 is H, SO2CH3, C(O)CH3, or C(O)(NC4H8); and


X2 is H, F, OCF3, SO2CH3, SO2NHCH3, or C(O)CH3.


Compounds of the present invention can, in yet another aspect, have the formula
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof, wherein:
embedded image


Accordingly, in one aspect, compounds according to the present invention can have the formula:
embedded image

or a salt, including a pharmaceutically acceptable or a non-pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof, wherein:
embedded imageembedded imageembedded image


According to another aspect of this invention, and consistent with the definitions provided herein, the present invention also provides for compounds of the following general structure III:
embedded image

wherein within structure III, the substituents Y1, R1, Y2, R2, R3 and R4 can be selected according to the following listings, wherein each substituent is defined in Table 1.


The substituent Y1 and Y2 can be selected independently from YA, YB, YC, YD, YE, YF, YG, YH, YI, or YJ.


The substituent R1 can be selected independently from R1A, R1B, R1C, R1D, R1E, R1F, R1G1, R1G2, R1G3, R1G4, R1G5, R1H1, R1H2, R1H3, R1H4, R1H5, R1I, R1J, R1K, R1L, R1M, R1N, R1O, R1P, R1Q.


The substituent R2 can be selected independently from R2A, R2B, R2C, R2D, R2E, R2F, R2G1, R2G2, R2G3, R2G4, R2G5, R2H1, R2H2, R2H3, R2H4, R2H5, R2I, R2J, R2K, R2L, R2M, R2N, R2O, R2P, or R2Q.


Alternatively, the moieties Y1R1 and Y2R2 can be selected independently from YRA, YRB, YRC, YRD, YRE, YRF, YRG, YRH, YRI, YRJ, or YRK, as defined herein.


The substituent R3 can be selected independently from R3A, R3B, R3C, R3D, R3E, R3F, R3G, R3H, R3I, R3J, R3K, R3L, R3M, R3N, R3O, R3P1, R3P2, R3P3, R3P4, R3P5, R3Q1, R3Q2, R3Q3, R3Q4, R3Q5, R3R, R3S, R3T, R3U, or R3V. The substituent R4 can be selected independently from R4A, R4B, R4C, R4D, R4E, R4F, R4G, R4H, R4I, R4J, R4K, R4L, R4M, R4N, R4O, R4P1, R4P2, R4P3, R4P4, R4P5, R4Q1, R4Q2, R4Q3, R4Q4, R4Q5, R4R, R4S, R4T, R4U, or R4V.


The substituents recited above are defined as follows, consistent with the definitions provided herein.

TABLE 1Substituent abbreviationsYA>NR5, wherein R5 is selected from R5A through R5GYB—(CH2)n—, n is 0 to 3YC—(CH2)p(CH═CH)(CH2)q—, p and q are independently 0 to 3YD>CR5R6, wherein R5 is selected from R5A through R5G, and R6 is selectedfrom R6A through R6GYE—(CH2)p(C≡C)(CH2)q—, p and q are independently 0 to 3YF—O—YG>COYH—S—YI>SOYJ>SO2YRAsaturated or unsaturated carbocyclic or N-heterocyclic ring having up to10 carbon atomsYRBsaturated or unsaturated carbocyclic or N-heterocyclic ring having up to10 carbon atoms, further comprising —O— in the ringYRCsaturated or unsaturated carbocyclic or N-heterocyclic ring having up to10 carbon atoms, further comprising —S— in the ringYRDsaturated or unsaturated carbocyclic or N-heterocyclic ring having up to10 carbon atoms, further comprising >N— in the ringYREsaturated or unsaturated carbocyclic or N-heterocyclic ring having up to10 carbon atoms, further comprising >SO2 in the ringYRFsaturated or unsaturated carbocyclic or N-heterocyclic ring having up to10 carbon atoms, further comprising >CO in the ringYRGsubstituted or an unsubstituted morpholinylYRHsubstituted or an unsubstituted piperazinylYRIsubstituted or an unsubstituted thiomorpholinylYRJsubstituted or an unsubstituted pyrrolidinylYRKsubstituted or an unsubstituted piperidinylR1A, R2AAlkyl having up to 10 carbon atomsR1B, R2BAryl having up to 10 carbon atomsR1C, R2CAlkoxyalkyl having up to 10 carbon atomsR1D, R2DCycloalky having up to 10 carbon atomsR1E, R2E—COR9 having up to 10 carbon atomsR1F, R2FAralkyl having up to 10 carbon atomsR1G1, R2G1Heterocyclyl having up to 10 carbon atoms, comprising —O—R1G2, R2G2Heterocyclyl having up to 10 carbon atoms, comprising >N—R1G3, R2G3Heterocyclyl having up to 10 carbon atoms, comprising —S—R1G4, R2G4Heterocyclyl having up to 10 carbon atoms, comprising >SO2R1G5, R2G5Heterocyclyl having up to 10 carbon atoms, comprising >COR1H1, R2H1Heteroaryl having up to 10 carbon atoms, comprising —O—R1H2, R2H2Heteroaryl having up to 10 carbon atoms, comprising >N—R1H3, R2H3Heteroaryl having up to 10 carbon atoms, comprising —S—R1H4, R2H4Heteroaryl having up to 10 carbon atoms, comprising >SO2R1H5, R2H5Heteroaryl having up to 10 carbon atoms, comprising >COR1I, R2IhydrogenR1J, R2JHalogenR1K, R2KCyanoR1L, R2LHydroxylR1M, R2MAlkoxy having up to 10 carbon atomsR1N, R2NAlkenyl having up to 10 carbon atomsR1O, R2OAlkynyl having up to 10 carbon atomsR1P, R2P—CO2R5 having up to 10 carbon atomsR1Q, R2Q—COR5 having up to 10 carbon atomsR3A, R4AAlkyl having up to 10 carbon atomsR3B, R4BAlkenyl having up to 10 carbon atomsR3C, R4CAlkynyl having up to 10 carbon atomsR3D, R4DAlkoxy having up to 10 carbon atomsR3E, R4ECycloalkyl having up to 10 carbon atomsR3F, R4FHaloalkyl having up to 10 carbon atomsR3G, R4GHaloalkoxy having up to 10 carbon atomsR3H, R4HAlkylthio having up to 10 carbon atomsR3I, R4IAlkylsufonyl having up to 10 carbon atomsR3J, R4JAryl having up to 10 carbon atomsR3K, R4K—CO2R5 having up to 10 carbon atomsR3L, R4L—COR5 having up to 10 carbon atomsR3M, R4M—NR5R6 having up to 10 carbon atomsR3N, R4N—SO2NR5R6 having up to 10 carbon atomsR3O, R4O—SO3R5 having up to 10 carbon atomsR3P1, R4P1Heterocyclyl having up to 10 carbon atoms, comprising —O—R3P2, R4P2Heterocyclyl having up to 10 carbon atoms, comprising >N—R3P3, R4P3Heterocyclyl having up to 10 carbon atoms, comprising —S—R3P4, R4P4Heterocyclyl having up to 10 carbon atoms, comprising >SO2R3P5, R4P5Heterocyclyl having up to 10 carbon atoms, comprising >COR3Q1, R4Q1Heteroaryl having up to 10 carbon atoms, comprising —O—R3Q2, R4Q2Heteroaryl having up to 10 carbon atoms, comprising >N—R3Q3, R4Q3Heteroaryl having up to 10 carbon atoms, comprising —S—R3Q4, R4Q4Heteroaryl having up to 10 carbon atoms, comprising >SO2R3Q5, R4Q5Heteroaryl having up to 10 carbon atoms, comprising >COR3R, R4RHydrogenR3S, R4SHalogenR3T, R4THydroxylR3U, R4UCyanoR3V, R4VY1R1, independent of the selection of Y1R1R5A, R6AAlkyl having up to 10 carbon atomsR5B, R6BAryl having up to 10 carbon atomsR5C, R6CAlkoxyalkyl having up to 10 carbon atomsR5D1, R6D1Heteroaryl having up to 10 carbon atoms, comprising —O—R5D2, R6D2Heteroaryl having up to 10 carbon atoms, comprising >N—R5D3, R6D3Heteroaryl having up to 10 carbon atoms, comprising —S—R5D4, R6D4Heteroaryl having up to 10 carbon atoms, comprising >SO2R5D5, R6D5Heteroaryl having up to 10 carbon atoms, comprising >COR5E, R6ECycloalkyl having up to 10 carbon atomsR5F1, R6F1Heterocyclyl having up to 10 carbon atoms, comprising —O—R5F2, R6F2Heterocyclyl having up to 10 carbon atoms, comprising >N—R5F3, R6F3Heterocyclyl having up to 10 carbon atoms, comprising —S—R5F4, R6F4Heterocyclyl having up to 10 carbon atoms, comprising >SO2R5F5, R6F5Heterocyclyl having up to 10 carbon atoms, comprising >COR5G, R6GHydrogen


In these selections, unless otherwise indicated, the number of carbon atoms on the substituents refers to the carbon atoms on the base chemical moiety, and does not include the carbon atoms in any optional substituent. Again, unless otherwise indicated, any substituents are limited in size by the carbon atoms listed in the definitions of the subsitutents.


In these selections, the following features are applicable. Any carbocyclic ring, N-heterocyclic ring, morpholinyl, piperazinyl, thiomorpholinyl, pyrrolidinyl, or piperidinyl can be optionally substituted with at least one hydroxyl, halogen, alkyl, alkoxy, haloalkyl, cycloalkyl, aryl, or heteroaryl any of which having up to 10 carbon atoms. Further any when a piperazinyl moiety is present in the substituted pyridine compound, the piperazine nitrogen is optionally substituted by an alkyl, a cycloalkyl, an acyl, a haloalkyl, an alkoxyalkyl, SO2R7, SO2NR72, or CO2R7, wherein R7 is independently selected from: a) an alkyl or an aryl having up to 8 carbon atoms; or b) hydrogen.


Any of the R1, R2, R5, or R6 moieties that do not constitute hydrogen, halogen, cyano, or hydroxyl (for example, R1A through R1H, R1M through R1Q, R2A through R2H, R2M through R2Q, R3A through R3Q and R3V, R4A through R4Q and R4V, R5A through R5F, and R6A through R6F) can be optionally substituted with at least one group independently selected from: 1) alkyl; alkoxy; alkylthio; haloalkyl; cycloalkyls; aryl; heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; haloalkoxy; —OCH2O—; —OCOR9; N(R8)2; —COR9; —CON(R8)2; —(CH2)bCO2R8 wherein b is an integer from 0 to 3; —OCO(CH2)bCO2R10 wherein b is an integer from 0 to 3; —SO2R9; —NHSO2R9; or —SO2N(R8)2; any of which having up to 10 carbon atoms; or 2) hydrogen, halogen, hydroxyl, or cyano. In these groups, R8, in each occurrence, is independently: 1) an alkyl; a haloalkyl; a heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; or an aryl having up to 10 carbon atoms; or 2) hydrogen. Further, in these moieties, R9, in each occurrence, is independently an alkyl; a haloalkyl; an aryl; or a heterocyclyl or heteroaryl comprising at least one heteroatom or heterogroup selected from —O—, >N—, —S—, >SO2, or >CO; having up to 8 carbon atoms; wherein R9 is optionally substituted with: 1) an alkyl, an alkoxy, a carboxylic acid, or a carboxylic acid ester, any of which having up to 8 carbon atoms; 2) halogen; or 3) hydroxyl; and


Any of the R3 or R4 moieties that do not constitute hydrogen, halogen, cyano, or hydroxyl can be optionally substituted with at least one group independently selected from: 1) alkyl, alkoxy, haloalkyl, haloalkoxy, cycloalkyl, aryl, heteroaryl, heterocyclyl, alkenyl, alkynyl, —COR10, —CO2R10, —CON(R10)2, —SO2R10, —SO2N(R10)2, or —N(R10)2, any of which having up to 10 carbon atoms; 2) halogen; or 3) hydroxyl; wherein R10, in each occurrence, is independently: 1) an alkyl or an aryl having up to 10 carbon atoms; or hydrogen.


Accordingly, this invention encompasses compounds of the formula III-E, corresponding to formula III in which Y2 is >NR5, and formula III-F, corresponding to formula III in which Y1 is >NR5.
embedded image


According to the various aspects of this invention, Y1, R1, R2, R4, and R5 of formulas III-E and Y2, R1, R2, R4, and R5 III -F can be selected according to the listings of substituent definitions provided herein.


In one aspect, the novel compound of the present invention encompasses any one of the following compounds, and any combination of the following compounds, including salts of the following compounds: [2-(3-methanesulfonyl-phenyl)-6-morpholin-4-yl-pyridin-4-yl]-(4-trifluoromethoxy-phenyl)-amine; 1-{3-[6-morpholin-4-yl-4-(4-trifluoromethoxy-phenylamino)-pyridin-2-yl]-phenyl}-ethanone; (2,6-di-phenyl-pyridin-4-yl)-(4-trifluoromethoxy-phenyl)-amine; [2,6-bis-(3-fluoro-phenyl)-pyridin-4-yl]-(4-trifluoromethoxy-phenyl)-amine; 1-[3-(4-hydroxy-6′-phenyl-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4′-ylamino)-phenyl]-ethanone; [2-(4-fluoro-phenyl)-6-morpholin-4-yl-pyridin-4-yl]-(4-trifluoromethoxy-phenyl)-amine; {3-[6-morpholin-4-yl-4-(4-trifluoromethoxy-phenylamino)-pyridin-2-yl]-phenyl }-pyrrolidin-1-yl-methanone; [6′-(4-Fluoro-phenyl)-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-2′-yl]-(4-trifluoromethoxy-phenyl)-amine; (6′-Phenyl-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-2′-yl)-(4-trifluoromethoxy-phenyl)-amine; [6′-(3-Methanesulfonyl-phenyl)-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-2′-yl]-(4-trifluoromethoxy-phenyl)-amine; 4-[6′-(4-Fluoro-phenyl)-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-2′-ylamino]-N-methyl-benzenesulfonamide; 1-[4-(4-Hydroxy-6′-phenyl-3,4,5,6-tetrahydro-2H-[1,4′]bi-pyridinyl-2′-ylamino)-phenyl]-ethanone; [6-(3-Methanesulfonyl-phenyl)-4-morpholin-4-yl-pyridin-2-yl]-(4-trifluoromethoxy-phenyl)-amine; N-Methyl-4-[4-morpholin-4-yl-6-(4-trifluoromethoxy-phenylamino)-pyridin-2-yl]-benzene-sulfonamide; 1-[4-(2-morpholin-4-yl-6-phenyl-pyridin-4-ylamino)-phenyl]-ethanone; 6′-(4-fluoro-phenyl)-4′-(4-trifluoromethoxy-phenylamino)-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4-ol; 1-[4-(4-Hydroxy-6′-phenyl-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4′-ylamino)-phenyl]-ethanone; 6′-(4-fluoro-phenyl)-4′-(4-methanesulfonyl-phenyl)-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4-ol; N-Methyl-4-[4-pyrrolidin-1-yl-6-(4-trifluoromethoxy-phenyl)-pyridin-2-ylamino]-benzene-sulfonamide; and the like, including any pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof.


Representative compounds in accordance with the present invention are presented in Table 2. This table is not intended to be exclusive of the compounds of the present invention, but rather exemplary of the heterocyclic compounds that are encompassed by this invention.

TABLE 2Representative compounds in accordance with the present inventionCmpd. No.StructureNameB1embedded image4-(3-fluoro-4-methoxy-phenyl)- pyridine;B2embedded image2-chloro-4-(3-fluoro-4-methoxy- phenyl)-pyridine;B3embedded image4-(3-fluoro-4-methoxy-phenyl)-2- phenyl-pyridine;B5embedded image2-(3-fluoro-4-methoxy-phenyl)-4- phenyl-pyridine;B6embedded image2-chloro-4-(3,4-difluoro-phenyl)- pyridine;B7embedded image4-(3,4-difluoro-phenyl)-2-phenyl- pyridine;B8embedded image2-chloro-4-(4-methanesulfonyl- phenyl)-pyridine;B9embedded image2-chloro-4-(3-methanesulfonyl- phenyl)-pyridine;B10embedded image4-benzo[1,3]dioxol-5-yl-2-chloro- pyridine;B11embedded image4-benzo[1,3]dioxol-5-yl-2-phenyl- pyridine;B12embedded image2,6-bis-(3-fluoro-4-methoxy-phenyl)- pyridine;B13embedded image(3-fluoro-4-methoxy-phenyl)-(4- phenyl-pyridin-2-yl)-amine;B14embedded imagecycloheptyl-[4-(3-fluoro-4-methoxy- phenyl)-pyridin-2-yl]-amine;B15embedded image[4-(3-fluoro-4-methoxy-phenyl)- pyridin-2-yl]-morpholine;B16embedded imagecyclohexylmethyl-[4-(3-fluoro-4- methoxy-phenyl)-pyridine-2-yl]-amine;B17embedded image[4-(3-fluoro-4-methoxy-phenyl)- pyridin-2-yl]-(4-fluoro-phenyl)-amine;B18embedded image(2-Chloro-pyridin-4-yl)-(4-fluoro-3- methoxy-phenyl)-amine;B19embedded image(3-chloro-4-methoxy-phenyl)-(2- phenyl-pyridin-4-yl)-amine;B20embedded image(4-fluoro-phenyl)-[4-(4- trifluoromethoxy-phenyl)-pyridin-2- yl]-amine;B22embedded image2,4,6-tris-(4-fluoro-phenyl)-pyridine;B23embedded image(3-chloro-4-methoxy-phenyl)-(2- chloro-6-phenyl-pyridin-4-yl)-amine;B24embedded imagebenzo[1,3]dioxol-5-yl-(2,6-dichloro- pyridin-4-yl)-amine;B25embedded image(2,6-dichloro-pyridin-4-yl)-(3-fluoro- phenyl)-amine;B26embedded image(2,6-diphenyl-pyridin-4-yl)-p-tolyl- amine;B27embedded image(2,6-diphenyl-pyridin-4-yl)-(4- trifluoromethoxy-phenyl)-amine;B28embedded image[2,6-bis-(3-fluoro-phenyl)-pyridin-4- yl]-(4-trifluoromethoxy-phenyl)-amine;B29embedded image[2,6-bis-(4-fluoro-phenyl)-pyridin-4- yl]-methyl-(4-trifluoromethoxy- phenyl)-amine;B30embedded image[2,6-bis-(4-methanesulfonyl-phenyl)- pyridin-4-yl]-(4-trifluoromethoxy- phenyl)-amine;B31embedded image[2,6-bis-(3-(methylsulfonyl)-phenyl)- pyridin-4-yl]-(4-trifluoromethoxy- phenyl)-amine;B32embedded imageN-ethyl-3-[6-(3-methanesulfonyl- phenyl)-4-(4-trifluoromethoxy- phenylamino)-pyridin-2-yl]- benzamide;B33embedded image1-{3-[6-(3-acetyl-phenyl)-4-(4- trifluoromethoxy-phenylamino)- pyridin-2-yl]-phenyl}-ethanone;B34embedded image1-{4-[2,6-bis-(4-fluoro-phenyl)- pyridin-4-ylamino]-phenyl}-ethanone;B35embedded image[2,6-bis-(3-N,N-dimethyl-benzamide)- pyridin-2-yl]-(4-trifluoromethoxy- phenyl)-amine;B36embedded image3-{2,6-bis-(4-fluoro-phenyl)-pyridin-4- ylamino]-benzenethiol; compound with acetic acid methyl ester;B37embedded imagethiocarbonic acid 0-methyl ester S-{3- [6-pyrrolidin-1-yl-4-(4- trifluoromethoxy-phenylamino)- pyridin-2-yl]-phenyl}ester;B38embedded image1-{3-[6-pyrrolidin-1-yl-4-(4- trifluoromethoxy-phenylamino)- pyridin-2-yl]-phenyl}-ethanone;B39embedded image[2-(4-fluoro-phenyl)-6-(4-methyl- piperazin-1 -yl)-pyridin-4-yl]-(4- trifluoromethoxy-phenyl)-amine;B40embedded image1-[4,6-bis-(4-fluoro-phenyl)-pyridin-2- yl]-4-methyl-piperazine;B41embedded image1-[4-(4-fluoro-phenyl)-6-(4- methanesulfonyl-phenyl)-pyridin-2-yl]- 4-methyl-piperazine;B42embedded image1-[6-(4-fluoro-phenyl)-4-(4- methanesulfonyl-phenyl)-pyridin-2-yl]- 4-methyl-piperazine;B43embedded image{4-[2-(4-fluoro-phenyl)-6-(4-methyl- piperazin-1-yl)-pyridin-4-yl]-phenyl}- morpholin-4-yl-methanone;B44embedded image1-[6-(4-fluoro-phenyl)-4-(4- trifluoromethoxy-phenyl)-pyridin-2- yl]-4-methyl-piperazine;B45embedded image[4-(2-morpholin-4-yl-6-phenyl- pyridin-4-ylamino)-phenyl]-ethanone;B46embedded image6′-(4-fluoro-phenyl)-4′-(4- trifluoromethoxy-phenylamino)- 3,4,5,6-tetrahydro-2H- [1,2′]bipyridinyl-4-ol;B47embedded image1-[4-(4-hydroxy-6′-phenyl-3,4,5,6- tetrahydro-2H-[1,2′]bipyridinyl-4′- ylamino)-phenyl]-ethanone;B48embedded image1-[3-(4-hydroxy-6′-phenyl-3,4,5,6- tetrahydro-2H-[1,2′]bipyridinyl-4′- ylamino)-phenyl]-ethanone;B49embedded image[2-(4-fluoro-phenyl)-6-morpholin-4-yl- pyridin-4-yl]-(4-trifluoromethoxy- phenyl)-amine;B50embedded image[2-(3-methanesulfonyl-phenyl)-6- morpholin-4-yl-pyridin-4-yl]-(4- trifluoromethoxy-phenyl)-amine;B51embedded image1-{3-[6-morpholin-4-yl-4-(4- trifluoromethoxy-phenylamino)- pyridin-2-yl]-phenyl}-ethanone;B52embedded image{3-[6-morpholin-4-yl-4-(4- trifluoromethoxy-phenylamino)- pyridin-2-yl]-phenyl}-pyrrolidin-1-yl- methanone;B53embedded image6′-chloro-4′-phenyl-3,4,5,6-tetrahydo- 2H-[1,2′]bipyridinyl-4-ol;B54embedded image4′,6′-bis-(4-fluoro-phenyl)-3,4,5,6- tetrahydro-2H-[1,2′]bipyridinyl-4-ol;B55embedded image6′-(4-fluoro-phenyl)-4′-(4- methanesulfonyl-phenyl)-3,4,5,6- tetrahydro-2H-[1,2′]bipyridinyl-4-ol;B56embedded image[6′-(4-fluoro-phenyl)-3,4,5,6-tetra- hydro-2H-[1,4′]bipyridinyl-2′-yl]-(4- trifluoromethoxy-phenyl)-amine;B57embedded image(4-trifluoromethoxy-phenyl)-[6′-(4- trifluoromethoxy-phenyl)-3,4,5,6- tetrahydro-2H-[1,4′]bipyridinyl-2′-yl]- amine;B58embedded image(6′-phenyl-3,4,5,6-tetrahydro-2H- [1,4′]bipyridinyl-2′-yl)-(4-trifluoro- methoxy-phenyl)-amine;B59embedded image[6′-(3-methanesulfonyl-phenyl)-3,4, 5,6-tetrahydro-2H-[1,4′]bipyridinyl-2′- yl]-(4-trifluoromethoxy-phenyl)-amine;B60embedded image4-[6′-(4-fluoro-phenyl)-3,4,5,6- tetrahydro-2H-[1,4′]bipyridinyl-2′- ylamino]-N-methyl-benzene- sulfonamide;B61embedded image1-[4-(4-hydroxy-6′-phenyl-3,4,5,6- tetrahydro-2H-[1,4′]bipyridinyl-2′- ylamino)-phenyl]-ethanone;B62embedded image2′,6′-bis-(4-fluoro-phenyl)-3,4,5,6- tetrahydro-2H-[1,4′]bipyridinyl;B63embedded image2′,6′-bis-(4-trifluoromethoxy-phenyl)- 3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl;B64embedded image(6-phenyl-4-pyrrolidin-1-yl-pyridin-2- yl)-(4-trifluoromethoxy-phenyl)-amine;B65embedded image[6-(4-fluoro-phenyl)-4-pyrrolidin-1-yl- pyridin-2-yl]-(4-trifluoromethoxy- phenyl)-amine;B66embedded imageN-methyl-4-[4-pyrrolidin-1-yl-6-(4- trifluoromethoxy-phenyl)-pyridin-2- ylamino]-benzenesulfonamide;B67embedded imageN-methyl-4-[4-(4-methyl-piperazin-1- yl)-6-(4-trifluoromethoxy- phenylamino)-pyridin-2-yl]- benzenesulfonamide;B68embedded image[6-(3-methanesulfonyl-phenyl)-4- morpholin-4-yl-pyridin-2-yl]-(4- trifluoromethoxy-phenyl)-amine;B69embedded imageN-methyl-4-[4-morpholin-4-yl-6-(4- trifluoromethoxy-phenylamino)- pyridin-2-yl]-benzenesulfonamide;B70embedded image(4-fluoro-phenyl)-[6-(4- trifluoromethoxy-phenyl)-pyridin-2- yl]-amine;B71embedded image2′,6′-bis-(4-trifluoromethoxy- phenylamino)-3,4,5,6-tetrahydro-2H- [1,4′]bipyridinyl-4-ol;B72embedded imageN2-cyclohexylmethyl-N4-(3-fluoro-4- methoxy-phenyl)-pyridine-2,4-diamine;B73embedded image[4-(2,6-dichloro-pyridin-4-yl)-phenyl]- morpholin-4-yl-methanone;B74embedded image2,6-dichloro-4-phenyl-pyridine;B75embedded image2,6-dichloro-4-(4-fluoro-phenyl)- pyridine;B76embedded image2,6-dichloro-4-(4-methanesulfonyl- phenyl)-pyridine;B77embedded image(3-chloro-4-methoxy-phenyl)-(2- chloro-pyridin-4-yl)-amineB78embedded image2-chloro-4-(4-trifluoromethoxy- phenyl)-pyridineB79embedded image(3-chloro-4-methoxy-phenyl)-(2,6- dichloro-pyridin-4-yl)-amineB80embedded image(2,6-dichloro-pyridin-4-yl)-p-tolyl- amineB81embedded image(2,6-dichloro-pyridin-4-yl)-(4- trifluoromethoxy-phenyl)-amineB82embedded image3-[6-chloro-4-(4-trifluoromethoxy- phenylamino)-pyridin-2-yl]-N-ethyl- benzamideB83embedded image1-[4-(2,6-dichloro-pyridin-4-ylamino)- phenyl]-ethanoneB84embedded image(2,6-dichloro-pyridin-4-yl)-(3- methylsulfanyl-phenyl)-amineB85embedded imageS-[3-(2,6-dichloro-pyridin-4-ylamino)- phenyl]ester-O-methyl esterB86embedded image(2-chloro-6-pyrrolidin-1-yl-pyridin-4- yl)-(4-tnfluoromethoxy-phenyl)-amineB87embedded image[2-chloro-6-(4-methyl-piperazin-1-yl)- pyridin-4-yl]-(4-trifluoromethoxy- phenyl)-amineB88embedded image2,6-dichloro-4-(4-fluoro-phenyl)- pyridineB89embedded image1-[6-chloro-4-(4-fluoro-phenyl)- pyridin-2-yl]-4-methyl-piperazineB90embedded image2,6-dichloro-4-(4-methanesulfonyl- phenyl)-pyridineB91embedded image1-[6-chloro-4-(4-methanesulfonyl- phenyl)-pyridin-2-yl]-4-methyl- piperazineB92embedded image[4-(2,6-dichloro-pyridin-4-yl)-phenyl]- morpholin-4-yl-methanoneB93embedded image{4-[2-chloro-6-(4-methyl-piperazin-1- yl)-pyridin-4-yl]-phenyl}-morpholin-4- yl-methanoneB94embedded image2,6-dichloro-4-(4-trifluoromethoxy- phenyl)-pyridineB95embedded image1-[6-chloro-4-(4-trifluoromethoxy- phenyl)-pyridin-2-yl]-4-methyl- piperazineB96embedded image1-[4-(2,6-dichloro-pyridin-4-ylamino)- phenyl]-ethanoneB97embedded image1-[4-(2-chloro-6-morpholin-4-yl- pyridin-4-ylamino)-phenyl]-ethanoneB98embedded image6′-chloro-4′-(4-trifluoromethoxy- phenylamino)-3,4,5,6-tetrahydro-2H- [1,2′]bipyridinyl-4-olB99embedded image1-[4-(6′-chloro-4-hydroxy-3,4,5,6- tetrahydro-2H-[1,2′]bipyridinyl-4′- ylamino)-phenyl]-ethanoneB100embedded image1-[4-(4-hydroxy-6′-phenyl-3,4,5,6- tetrahydro-2H-[1,2′]bipyridinyl-4′- ylamino)-phenyl]-ethanoneB101embedded image1-[3-(6′-chloro-4-hydroxy-3,4,5,6- tetrahydro-2H-[1,2′]bipyridinyl-4′- ylamino)-phenyl]-ethanoneB102embedded image(2-chloro-6-morpholin-4-yl-pyridin-4- yl)-(4-trifluoromethoxy-phenyl)-amineB103embedded image2,6-duchloro-4-phenyl-pyriduneB104embedded image6′-chloro-4′-(4-fluoro-phenyl)-3,4,5,6- tetrahydro-2H-[1,2′]bipyridinyl-4-olB105embedded image6′-chloro-4′-(4-methanesulfonyl- phenyl)-3,4,5,6-tetrahydro-2H- [1,2′]bipyridinyl-4-olB106embedded image(2-chloro-pyridin-4-yl)-(3-fluoro-4- methoxy-phenyl)-amineB107embedded image2′,6′-dichloro-3,4,5,6-tetrahydro-2H- [1,4′]bipyridinyl (alternatively, 2,6- dichloro-4-piperdino pyridine)B108embedded image(6′-chloro-3,4,5,6-tetrahydro-2H- [1,4′]bipyridinyl-2′-yl]-(4- trifluoromethoxyphenyl)-amineB109embedded image4-(6′-chloro-3,4,5,6-tetrahydro-2H- [1,4′]bipyridinyl-2′-ylamino)-N- methyl-benzenesulfonamideB110embedded image2′,6′-dichloro-3,4,5,6-tetrahydro-3H- [1,4′]bipyridinyl-4-olB111embedded image2′-chloro-6′-phenyl-3,4,5,6-tetrahydro- 2H-[1,4′]bipyridinyl-4-olB112embedded image2,6-dichloro-4-pyrrolidino pyridineB113embedded image(6-chloro-4-pyrrolidin-1-yl-pyridin-2- yl)-(4-trifluoromethoxy-phenyl)-amineB114embedded image4-(6-chloro-4-pyrrolidin-1-yl-pyridin- 2-ylamino)-N-methyl- benzenesulfonamideB115embedded image1-(2,6-dichloro-pyridin-4-yl)-4-methyl- piperazineB116embedded image[6-chloro-4-(4-methyl-piperazin-1-yl)- pyridin-2-yl]-(4-trifluoromethoxy- phenyl)-amineB117embedded image4-(2,6-dichloro-pyridin-4-yl)- morpholineB118embedded image(6-chloro-4-morpholin-4-yl-pyridin-2- yl)-(4-trifluoromethoxy-phenyl)-amineB119embedded image(6-chloro-pyridine-2-yl)-(4-fluoro- phenyl)-amine


In this aspect of the present invention, compounds provided herein may be chiral or achiral, or they may exist as racemic mixtures, diastereomers, pure enantiomers, a prodrug, a tautomer, or any mixture thereof. For chiral compounds, separate enantiomers, separate diastereomers, and any mixture of enantiomers, diastereomers, or both are encompassed herein. Further, the present invention also encompasses any combination of compounds provided herein, including any salts, including pharmaceutically acceptable or non-pharmaceutically acceptable salts, or any mixture thereof.


As used herein, the terms “pharmaceutically acceptable” salt or “pharmacologically acceptable” salt refers generally to a salt or complex of the compound or compounds in which the compound can be either anionic or cationic, and have associated with it a counter cation or anion, respectively, that is generally considered suitable for human or animal consumption. For example, a pharmaceutically acceptable salt can refer to a salt of a compound disclosed herein that forms upon reaction or complexation with an acid whose anion is generally considered suitable for human or animal consumption. In this aspect, pharmacologically acceptable salts include salts with organic acids or inorganic acids. Examples of pharmacologically acceptable salts include, but are not limited to, hydrochloride, hydrobromide, hydroiodide, sulfate, phosphate, propionate, lactate, maleate, malate, succinate, tartarate, and the like.


Salts may also be formed by deprotonating acid moiety of compound, such as a carboxylic acid moiety, OH, or NH, and the like, using a base such as an organic base, an inorganic base, an organometallic base, a Lewis base, a BrØnsted base, or any mixture thereof. In cases where compounds carry an acidic moiety, suitable pharmaceutically acceptable salts can include alkali metal salts, alkaline earth metal salts, or salts with organic basis, and the like. In this aspect, examples of alkali metal salts include, but are not limited to, sodium and potassium salts, and examples of salts with organic basis include, but are not limited to, meglumine salts, and the like. The pharmacologically acceptable salts may be prepared by conventional means. Additional examples of pharmaceutically acceptable salts, and methods of preparing such salts, are found, for example, in Berg et.al., J. Pharma. Sci, 66, 1-19 (1977).


In a further aspect, this invention also provides a composition comprising at least one compound as disclosed herein, including a composition comprising a pharmaceutically acceptable carrier and at least one compound as disclosed herein. In this aspect, the at least one compound can be present as a neutral compound, as a salt, or as any combination thereof. This invention also encompasses a composition comprising at least one compound as disclosed herein, and optionally comprising a pharmaceutically acceptable additive selected from a carrier, an auxiliary, a diluent, an excipient, a preservative, a solvate, or any combination thereof.


Further, this invention encompasses a pharmaceutical composition, comprising at least one compound as disclosed herein, and optionally comprising a pharmaceutically acceptable additive selected from a carrier, an auxiliary, a diluent, an excipient, a preservative, a solvate, or any combination thereof, wherein the pharmaceutical composition is in the form of a tablet, a capsule, a syrup, a cachet, a powder, a granule, a solution, a suspension, an emulsion, a bolus, a lozenge, a suppository, a cream, a gel, a paste, a foam, a spray, an aerosol, a microcapsule, a liposome, or a transdermal patch.


In another aspect, this invention encompasses a pharmaceutical composition, comprising at least one compound as disclosed herein, and optionally comprising a pharmaceutically acceptable additive selected from a carrier, an auxiliary, a diluent, an excipient, a preservative, a solvate, or any combination thereof; and further comprising an agent selected from a chemotherapeutic agent, an immunosuppressive agent, a cytokine, a cytotoxic agent, an anti-inflammatory agent, an antirheumatic agent, a cardiovascular agent, or any combination thereof.


Another aspect of this invention is directed to using the compounds and compositions disclosed herein in a method of treating a condition or disease state mediated by the low expression of Perlecan, comprising administering an amount of at least one compound as disclosed herein, effective to induce Perlecan expression.


A further aspect of this invention is directed to using the compounds and compositions disclosed herein in a method of treating atherosclerosis, arthritis, restenosis, diabetic nephropathy, or dyslipidemia, comprising administering an effective amount of at least one compound as disclosed herein.


Preparation of Substituted Pyridine Compounds


One more aspect of the present invention provides a process for the preparation of the compounds of general formulas (I) and (III). Thus, substituted pyridine analogs can be prepared generally using standard synthetic methods and employing starting materials that are readily available commercially. As demonstrated by the general reaction schemes and examples disclosed herein, the general synthetic methods provided will be readily understood by one of ordinary skill in the art, and any variations needed for a particular species are simple and readily understood and appreciated by the skilled artisan. In the following general reaction schemes, variable chemical moieties refer to any chemical group consistent with the description of the compound and substituents on that compound as provided herein. Further, in the schemes that follow, the term “palladium catalyst” refers to a suitable palladium catalyst, typically a complex of Pd(0) or Pd(II), including but not limited to, such compounds as palladium(0) tetrakis-(triphenylphosphine), tris(dibenzylideneacetone)dipalladium(0), palladium(II) acetate, that is known to catalyze the reaction shown. In one aspect, the catalytic system can also include monodentate or chelating ligands, examples of which include, but are not limited to, 2,2′-bis(diphenyl phosphino)-1,1-binapthyl, tri-tert-butyl phosphine, and the like, and can also include a base such as sodium carbonate, sodium or potassium tert-butoxide, or potassium phosphate. Transition metal catalyzed reactions can be typically carried out at ambient temperature or at elevated temperatures using various inert solvents, examples of which include, but are not limited to, toluene, dioxane, DMF, N-methyl pyrrolidine, ethylene glycol, dimethyl ether, diglyme, acetonitrile, or any combination thereof. In one aspect, for example, commonly employed reagent and catalyst pairs include, but are not limited to, aryl boronic acids and palladium(0), (Suzuki reaction, Miyaura and Suzuki, Chem. Rev. 1995, 95, 2457).


The following general reaction schemes provide some of the synthetic methods that can be used to prepare the pyridine compounds disclosed herein.


In one aspect of this invention, as provided in Scheme 1, a compound of formula 1a was aminated with any of a variety of substituted or unsubstituted anilines to provide a compound of the formula 1b, wherein R1 is typically an aryl group such as a substituted or unsubstituted phenyl group and Y is a leaving group, in presence of tris(dibenzylideneacetone)dipalladium(0), 1,3-bis(diphenylphosphino)propane, and sodium tert-butoxide. Compound 1b was converted to compound of the type 1c, where one or both of the R2 substituents can be a substituted or unsubstituted phenyl and one of the R2 is optionally hydrogen, by palladium-catalyzed cross-coupling of substituted or unsubstituted phenyl boronic acids. For example, palladium tetrakis-(triphenyl-phosphine) was used as palladium catalyst in this reaction scheme. These fundamental reactions appear in additional reaction sequences provided throughout.
embedded image


In another aspect of this invention, as provided in Scheme 2, a compound of formula 2a, prepared for example according to Scheme 1, was useful in further amination reactions. For example, compound 2a was treated with various primary (H2NR1, as illustrated in Scheme 2) or secondary amines (HNR12, not illustrated in Scheme 2) in an appropriate solvent such as dimethylformamide, N-methyl pyrollidine, and a base such as potassium carbonate. Examples of secondary amines that were used include, but are not limited to, the heterocyclic compounds such as piperidine, pyrrolidine, and the like. Compounds of the formula 2b were then converted to compounds of the type 2c, where R3, in one aspect, is an aryl group such as a substituted or unsubstituted phenyl, by a palladium-catalyzed cross-coupling of substituted or unsubstituted phenyl boronic acids. For example, palladium tetrakis(triphenylphosphine) was used as palladium catalyst in this reaction scheme.
embedded image


In still another aspect of this invention, a compound of formula 1a, which was prepared, for example, as illustrated in Scheme 1, was converted to a compound of formula 3a, Scheme 3, where R1 represents at least one optional substituent on the aryl group, by reacting 1a with appropriately substituted or unsubstituted phenyl boronic acids. Compound 3a was then converted to compound 3b, wherein R2 is an amino group, by its reaction with, for example, a primary or secondary amine or aniline, in an appropriate solvent such as dimethylformamide, N-methyl pyrollidine, and a base such as potassium carbonate. Compound 3b was converted to a compound of formula 3c, where R3, in one aspect, is an aryl group such as a substituted or unsubstituted phenyl, by a palladium-catalyzed cross-coupling of substituted or unsubstituted phenyl boronic acids, in the presence of a base such as potassium carbonate or sodium carbonate. For example, tris(dibenzylideneacetone)dipalladium(0) was used as palladium catalyst in this step of the reaction scheme.
embedded image


In yet another aspect of this invention, a compound of formula 4a is converted to compound of formula 4b, where R1 was a range of hydrocarbyl groups, such as substituted or unsubstituted alkyl or aryl groups, by the reaction of 4a with the appropriate substituted or unsubstituted primary amine (H2NR1, as illustrated in Scheme 4) or secondary amine (HNR12, not illustrated in Scheme 4), or aniline (also not illustrated in Scheme 4). This reaction was typically effected in a polar solvent, including but not limited to, dimethylsulfoxide, and in the presence of base, for example sodium hydride. In a further amidation reaction, compound 4b was converted to compound 4c by its reaction with a range of substituted or unsubstituted anilines H2NR2 in the presence of a base and a palladium catalyst, where R2 is typically a substituted or unsubstituted aryl group. Compound 4c was converted to compounds of the type 4d, where R3, in one aspect, is an aryl group such as a substituted or unsubstituted phenyl, by a palladium-catalyzed cross-coupling of substituted or unsubstituted phenyl boronic acids. For example, palladium tetrakis(triphenylphosphine) was used as palladium catalyst in this reaction scheme.
embedded image


In a further aspect of this invention and as provided in Scheme 5, pyridines with leaving groups at the 2-, 4-, and 6-positions such as trihalogenated pyridines, were useful in preparing a number of substituted pyridines. For example, 2,4,6-trichloropyridine, formula 5a, was aminated using primary (H2NR1, as illustrated in Scheme 5) or secondary amines (HNR12, not illustrated in Scheme 5) to prepared the 2,6-dihalogenated pyridines of formula 5b. The palladium-catalyzed cross-coupling reaction of substituted or unsubstituted phenyl boronic acids with compound 5b afforded the 2,6-diarylated pyridine compounds of formula 5c. Treatment of 2,6-dihalogenated pyridines 5b with any of a variety of substituted or unsubstituted anilines H2NR2 wherein R2 is typically an aryl group such as a substituted or unsubstituted phenyl group, in the presence of a palladium catalyst such as a palladium (II) catalyst, provided the 2,6-bis(aryl)amino pyridine derivatives 5d. The conversion of 5b to 5d can be accomplished stepwise, such that two different —NHR2 moities can be substituted at the pyridine core.
embedded image


In another aspect, the following reaction Scheme 6 is a general reaction scheme that illustrates one aspect of how the compounds of the present invention can be prepared. The compound of formula (I-A) wherein L represents leaving group selected from halogen, aryloxy, alkylsulfinyl, alkylsulfonyl such as trifluoromethanesulfonyloxy, arylsulfinyl, arylsulfonyl, siloxy, cyano, pyrazolo, triazolo and the like, is converted to a compound of formula (I) by reacting with the compound GR2 wherein G represents hydrogen, NH2, NHR5, OH, SH, B(OH)2, Li or MgZ where Z represents halogen; when G represents NR5, R2 and R5 together may also form an optionally substituted cyclic ring along with adjacent N atom, which may be optionally containing one or more hetero atoms selected from oxygen, nitrogen or sulfur; and all other symbols are as defined earlier, in presence of a base such as sodium hydroxide, potassium hydroxide, potassium carbonate and the like or Lewis acid such as aluminum chloride (AlCl3) or palladium catalyst such as tetrakis-(triphenylphosphine)-palladium(0) [(PPh3)4Pd], bis-(triphenylphosphine)-palladium(II)chloride [(PPh3)2PdCl2] and the like. The reaction is carried out in presence of solvent such as acetone, dimethylformamide (DMF), dimethylacetamide (DMA), benzene, toluene and the like. The temperature of the reaction may be in the range of about 25° C. to about 150° C. The duration of the reaction is variable, but can be, for example, in the range of about 2 to about 48 hours.
embedded image


Thus, in accordance with the reaction schemes provided herein, typical reactions and reaction conditions that can be used to prepare the novel compounds of this invention include, but are not limited to, for example, the reactions provided in Scheme 7. Thus, in Scheme 7, typical reaction conditions include, but by no means are limited to the following. These conditions are provided solely as a guide for one of ordinary skill, such that the skilled artisan will readily appreciate how modifications of these conditions can selected according to the particular chemical moiety being substituted at the pyridine core. Thus, Examples of conditions include, but are not limited to: A, acetonitrile, sodium carbonate (0.4 M), tetrakis(triphenylphosphine)palladium(0), reflux; B. (PhCH═CHCOCH═CHPh)3Pd2 (tris(dibenzylidineacetone)dipalladium(0)), sodium-tert-butoxide, 2,8,9-triisobutyl-2,5,8,9-tetraaza-1-phosphabicyclo[3.3.3]undecane (CAS number 331465-71-5), toluene, reflux; C, (PhCH═CHCOCH═CBPh)3Pd2 (tris(dibenzylidineacetone)dipalladium(0)), 1,3-bis(diphenylphosphino)propane, sodium-tert-butoxide, toluene, reflux; D, dimethoxy ethane, sodium carbonate (2 M), tetrakis(triphenylphosphine)palladium(O), reflux under nitrogen.
embedded image


The following reaction schemes provide additional synthetic methods that can be used to prepare the pyridine compounds disclosed herein.


Synthesis of 2,4,6-trisubstituted pyridines comprising a 4-cycloalkylamino substituent. As illustrated in Scheme 8, the intermediate compound shown as compound 8a can be used to prepare several tri-substituted pyridines such as those exemplified in this scheme. Conditions “a” for the reaction shown are as follows. The heterocyclic amine was reacted with trichloropyridine under basic conditions [NaOH (aq) or NaH (anhydrous)] in a polar organic solvent at temperatures ranging from 0° C. to 60° C. to yield compound 8a. For conditions “b”, the intermediate was coupled with a boronic acid under Suzuki conditions using a Pd catalyst [for example, Pd(PPh3)4] in the presence of a base (for example, Na2CO3 or K2CO3) in a polar solvent under thermal conditions, either traditional thermal conditions or under microwave heating conditions. For conditions “c”, the monochloropyridine was aminated using Buchwald-Hartwig conditions using a Pd catalyst [for example, Pd(OAc)2] and a ligand such as BINAP, under basic conditions (for example, using potassium tert-butoxide) in a toluene solvent, in a laboratory microwave (at about 150° C.).
embedded image


Synthesis of 2,4,6-trisubstituted pyridines comprising a 4-arylamino substituent. As illustrated in Scheme 9, the intermediate compound shown as compound 9a can be used to prepare various tri-substituted pyridines such as those exemplified in this scheme. Conditions “a” for the reaction shown are as follows. The compound 2,6-dichloro-4-iodopyridine was aminated using Buchwald-Hartwig conditions using a Pd catalyst [for example, Pd2(dba)3] and a ligand (for example, dpp), under basic conditions (sodium tert-butoxide) in an toluene solution at reflux. For conditions “b”, the intermediate was coupled with a boronic acid under Suzuki conditions using a Pd catalyst [for example, Pd(PPh3)4] in the presence of a based (for example, Na2CO3) in a polar solvent at reflux. For conditions “c”, the intermediate was aminated with the heterocyclic amine using excess amine or in DMF/K2CO3 at reflux. For conditions “d”, the intermediate was alkylated using a base followed by an alkyl halide.
embedded image


Synthesis of 2,4,6-trisubstituted pyridines comprising a 4-aryl substituent. As illustrated in Scheme 10, the substituted pyridines such as those exemplified in this scheme were prepared as follows. For conditions “a”, 2,6-dichloro-4-iodopyridine was coupled with an boronic acid under Suzuki conditions using a Pd catalyst [for example, Pd(PPh3)4] in the presence of a base (such as Na2CO3) in a polar solvent at reflux to give intermediate 10a. For conditions “b”, the intermediate 10a was aminated with the heterocyclic amine under basic conditions (such as K2CO3) in a DMF at about 90° C. After isolation and another Suzuki coupling, compound 10b was prepared. Compound 10c was prepared under Suzuki conditions.
embedded image


General synthesis of 4-mono-substituted pyridines that can be used for the synthesis of 2,4,6-trisubstituted pyridines. Scheme 11 illustrates the preparation of 4-aryl substituted pyridines that were synthesized under standard Suzuki conditions as described in various other reaction schemes disclosed herein. Compound 11a is drawn to indicate that the chemistry could potentially be expanded if further functionality was in the 2-position (A), the 6-position (B), or both the 2- and 6-positions. Thus, if Compound 11a was appropriately halo-substituted, then other Pd-mediated coupling/amination chemistry could be employed to yield substituted 2,4,6-substituted pyridines.
embedded image


Synthesis of 2,6-disubstituted pyridines that can be used for the synthesis of 2,4,6-trisubstituted amines). Scheme 12 illustrates the typical preparation of 2,6-di-aryl substituted pyridines (12b) that were prepared under standard Suzuki conditions (“a”) as described the various reaction schemes disclosed herein. For the 2-amino-6-aryl substituted pyridines (12c), compound 12a was aminated under microwave conditions (thermal) in the presence of a base such as potassium tert-butoxide (conditions “b”). The intermediate compound shown could then be subjected to conditions “a” as illustrated. For example, for synthesizing compound 12b, 2,6-dibromopyridine was used as the starting compound, and for synthesizing compound 12c, 2,6-dichloropyridine was used as the starting compound. However, X can be at least Cl, Br, or I, as indicated in the compound 12a. Additionally, 12a is shown indicating a further functionality located at the 4 (C) position, which also occurs in compounds 12b and 12c. For example, if the pyridine 12a is halo-substituted at the 4-position (C is halide), then other Pd-mediated coupling/amination chemistry could be employed to yield substituted 2,4,6-substituted pyridines.
embedded image


Synthesis of 2,4-disubstituted pyridines that can be used for the synthesis of 2,4,6-trisubstituted amines. Scheme 13 illustrates the typical preparation of 2,4-di-aryl substituted pyridines that were prepared under sequential Pd-mediated couplings. For conditions “a”, 2,6-dichloro-4-iodopyridine was coupled with an boronic acid under Suzuki conditions using a Pd catalyst [for example, Pd(PPh3)4] in the presence of a base (such as Na2CO3) in a polar solvent at reflux to give intermediates or final products. For conditions “b”, Buchwald or Buchwald-Hartwig Pd-mediated amination conditions [Pd2(dba)3, dpp, sodium tert-butoxide in refluxing toluene] were used. For conditions “c”, the Verkade Pd-mediated amination conditions were used which comprising employing the Verkade ligand, 2,8,9-triisobutyl-2,5,8,9-tetraza-1-phosphabi-cyclo [3.3.3]undecane, dpp, Pd2(dba)3, sodium tert-butoxide in refluxing toluene. Compound 13a is drawn to indicate that the chemistry could potentially be expanded if further functionality was at the 6-position (D). For example, if the pyridine 6-position is halo-substituted, then other Pd-mediated coupling/amination chemistry could be employed to yield substituted 2,4,6-substituted pyridines. Otherwise the functionality could already be in place prior to initiating the reaction sequence.
embedded image

Prodrugs


The compounds alternatively can be formulated and administered in a prodrug form. In general, prodrugs comprise functional derivatives of the claimed compounds which are capable of being enzymatically activated or converted into the more active parent form. Thus, in the treatment methods of the present invention, the term “administering” encompasses the treatment of the various disorders described with the compound specifically disclosed or with a compound which may not be specifically disclosed, but which converts to the specified compound in vivo after administration to the patient. Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in Wihnan, 14 Biochem. Soc. Trans. 375-82 (1986); Stella et al., Prodrugs: A Chemical Approach to Targeted Drug Delivery in Directed Drug Delivery, 247-67 (1985).


The prodrugs of present invention include, but are not limited to, derivatives of carboxylic acid, sulfonamide, amine, hydroxyl, and the like, including other functional groups and including any combination thereof.


In another aspect, this invention provides a pharmaceutical composition, comprising a compound any of the formulas shown above, including any combination thereof, and optionally comprising a pharmaceutically acceptable additive selected from a carrier, an auxiliary, a diluent, an excipient, a preservative, a solvate, and the like, or any combination thereof. In a related aspect, this invention affords a method of treating a condition or disease state mediated by the low expression of Perlecan, comprising administering at least one compound as disclosed herein, in an amount effective to induce Perlecan expression. In a related aspect, this invention also provides a method of treating atherosclerosis, arthritis, restenosis, diabetic nephropathy, or dyslipidemia, comprising administering an effective amount of at least one compound as disclosed herein.


Antiproliferative Activities


One aspect of the present invention comprises methods and compositions comprising the compounds of the present invention for the treatment and prevention of conditions or diseases that have as an aspect of the disease or condition, unwanted cellular proliferation occurring or are the result of cellular proliferation. For example, many vascular diseases, such as cardiovascular diseases, organ transplant sequellae, vascular occlusive conditions including, but not limited to, neointimal hyperplasia, restenosis, transplant vasculopathy, cardiac allograft vasculopathy, atherosclerosis, and arteriosclerosis, are caused by or have collateral damage due to unwanted cellular proliferation.


One aspect of the present invention relates to methods and compositions for the treatment and prevention of SMC proliferation, such compositions comprising compounds having cellular antiproliferative activity. These compounds and compositions comprising such compounds are referred to as antiproliferative compounds or compositions. At least one activity of one or more of these compounds is that the compound has the activity of affecting the synthesis of proteoglycans including induction and synthesis of proteoglycans and active fragments of proteoglycans. Thus, one aspect of the activity of one or more of the compounds and compositions of the present invention comprise molecules that induce HSPG production and that regulate SMC proliferation.


Compounds of the present invention that have at least the activity of affecting cellular proliferation are shown in Table 3.

TABLE 3Compounds of the present invention that have at least the activity of affecting15 cellular proliferationEntryStructureCompound Name 1embedded image3-chloro-4-methoxy-phenyl)-(2-phenyl- pyridin-4-yl)-amine 2embedded image(2,6-diphenyl-pyridin-4-yl)-p-tolyl- amine 3embedded image(2,6-diphenyl-pyridin-4-yl)-(4- trifluoromethoxy-phenyl)-amine 4embedded image[2,6-bis-(3-fluoro-phenyl)-pyridin-4-yl]- (4-trifluoromethoxy-phenyl)-amine 5embedded image[2,6-bis-(4-fluoro-phenyl)-pyridin-4-yl]- methyl-(4-trifluoromethoxy-phenyl)- amine 6embedded image[2,6-bis-(4-methanesulfonyl-phenyl)- pyridin-4-yl]-(4-trifluoromethoxy- phenyl)-amine 7embedded image2,6-Bis-(3-methanesulfonyl-phenyl)- pyridin-4-yl]-(4-trifluoromethoxy- phenyl)-amine 8embedded imageN-ethyl-3-[6-(3-methanesuifonyl- phenylamino)-pyridin-2-yl]-benzamide 9embedded image1-{3-[6-(3-acetyl-phenyl)-4-(4- trifluoromethoxy-phenylamino)-pyridin- 2-yl]-phenyl}-ethanone10embedded image1-{4-[2,6-bis-(4-fluoro-phenyl)-pyridin- 4-ylamino]-phenyl}-ethanone11embedded image[2,6-bis-(3-N,N-dimethyl-benzamide)- pyridin-2-yl]-(4-trifluoromethoxy- phenyl)-amine12embedded image[2,6-Bis-(4-fluoro-phenyl)-pyridin- 4-yl]-(3-methanesulfonyl-phenyl)-amine13embedded image[2-(3-Methanesulfonyl-phenyl)-6-pyr- rolidin-1-yl-pyridin-4-yl]-(4-trifluoro- methoxy-phenyl)-amine14embedded image1-{3-[6-pyrrolidin-1-yl-4-(4- trifluoromethoxy-phenylamino)-pyridin- 2-yl]-phenyl}-ethanone15embedded image[2-(4-fluoro-phenyl)-6-(4-methyl- piperazin-1-yl)-pyridin-4-yl]-(4- trifluoromethoxy-phenyl)-amine16embedded image1-[4-(2-morpholin-4-yl-6-phenyl- pyridin-4-ylamino)-phenyl]-ethanone17embedded image1-[4-(4-hydroxy-6′-phenyl-3,4,5,6- tetrahydro-2H-[1,2′]bipyridinyl-4′- ylamino)-phenyl]-ethanone18embedded image1-[3-(4-hydroxy-6′-phenyl-3,4,5,6- tetrahydro-2H-[1,2′]bipyridinyl-4′- ylamino)-phenyl]-ethanone19embedded image[2-(4-fluoro-phenyl)-6-morpholin-4-yl- pyridin-4-yl]-(4-trifluoromethoxy- phenyl)-amine20embedded image[2-(3-methanesulfonyl-phenyl)-6- morpholin-4-yl-pyridin-4-yl]-(4- trifluoromethoxy-phenyl)-amine21embedded image1-{3-[6-morpholin-4-yl-4-(4- trifluoromethoxy-phenylamino)-pyridin- 2-yl]-phenyl}-ethanone22embedded image{3-[6-morpholin-4-yl-4-(4- trifluoromethoxy-phenylamino)-pyridin- 2-yl]-phenyl}-pyrrolidin-1-yl- methanone23embedded image[6′-(4-Fluoro-phenyl)-3,4,5,6-tetra- hydro-2H-[1,4′]bipyridinyl-2′-yl]- (4-trifluoromethoxy-phenyl)-amine24embedded image(4-Trifluoromethoxy-phenyl)-[6′-(4- trifluoromethoxy-phenyl)-3,4,5,6- tetrahydro-2H-[1,4′]bipyridinyl-2′-yl]- amine25embedded image(6′-Phenyl-3,4,5,6-tetrahydro-2H- [1,4′]bipyridinyl-2′-yl)-(4-trifluoro- methoxy-phenyl)-amine26embedded image3[6′-(3-Methanesulfonyl-phenyl)-3,4, 5,6-tetrahydro-2H-[1,4′]bipyridinyl- 2′-yl]-(4-trifluoromethoxy-phenyl)- amine27embedded image4-[6′-(4-Fluoro-phenyl)-3,4,5,6- tetrahydro-2H-[1,4′]bipyridinyl-2′-yl- amino]-N-methyl-benzenesulfonamide28embedded image1-[4-(4-Hydroxy-6′-phenyl-3,4,5,6- tetrahydro-2H-[1,4′]bipyridinyl-2′-yl- amino)-phenyl]-ethanone29embedded image(6-Phenyl-4-pyrrolidin-1-yl-pyridin-2- yl)-(4-trifluoromethoxy-phenyl)-amine30embedded image[6-(4-Fluoro-phenyl)-4-pyrrolidin-1-yl- pyridin-2-yl]-(4-trifluoromethoxy- phenyl)-amine31embedded imageN-Methyl-4-[4-pyrrolidin-1-yl-6-(4- trifluoromethoxy-phenyl)-pyridin-2- ylamino]-benzenesulfonamide32embedded imageN-Methyl-4-[4-(4-methyl-piperazin-1- yl)-6-(4-trifluoromethoxy- phenylamino)-pyridin-2-yl]- benzenesulfonamide33embedded image[6-(3-Methanesulfonyl-phenyl)-4- morpholin-4-yl-pyridin-2-yl]-(4- trifluoromethoxy-phenyl)-amine34embedded imageN-Methyl-4-[4-morpholin-4-yl-6-(4- trifluoromethoxy-phenylamino)-pyridin- 2-yl]-benzenesulfonamide35embedded image(4-Fluoro-phenyl)-[6-(4- trifluoromethoxy-phenyl)-pyridin-2-yl]- amine36embedded image2′,6′-Bis-(4-trifluoromethoxy- phenylamino)-3,4,5,6-tetrahydro-2H- [1,4′]bipyridinyl-4-ol


Methods for identifying the activity and screening for one or more of these compounds or molecules that induce synthesis of proteoglycans such as HSPG are taught in U.S. patent application Ser. No. 10/091,357, which is incorporated herein in its entirety. Assays of effects of compounds in vivo are also taught in the incorporated references and are known to those skilled in the art. In general, methods comprise the addition of such compounds to assays and measurement of HSPG synthesis including, but not limited to, the production of syndecans, glypicans, and perlecans, for example, syndecans 1, 2 and 4; and glypican-1. Other assays that can be used to determine the activity of the compounds of the present invention include other methods for measuring the induction of perlecan synthesis. For example, in one assay, perlecan is induced in cells by certain inducers, and the response is measured. Compounds of the present invention are then added to a replicate assay and the effect on perlecan induction is determined. Using such methods, compounds are determined that can either inhibit perlecan, elevate induction of perlecan, or have no effect at all. Those compounds that are effective as therapeutic agents can then be used in animals, humans or patients with cellular proliferation disease aspects, such as vascular-associated diseases or SMC (smooth muscle cell) proliferation pathologies.


Another assay for determining compounds having SMC effects comprises adding a composition suspected of effecting SMC proliferation to smooth muscle cells in growth medium or serum-free medium. The change in cell proliferation can be measured by methods known to those skilled in the art, such as incorporation of labeled nucleotides into dividing cells' DNA, and compared to the proliferation of cells which are not treated with the compound. Other measurements include directly determining levels of HSPG synthesis by measuring the amount or change in amount of HSPG such as with ELISA for HSPGs, and compared to the amount of HSPG synthesis in untreated cells. Other indirect or direct measurements are contemplated by the present invention and are known to those skilled in the art. For example, such methods include, but are not limited to, measurement of RNA levels, RT-PCR, Northern blotting, Western blotting promoter-based assays to identify compounds that affect one or more proteoglycans and assays for proteoglycan biological activity shown by recombinant proteins, partially purified proteins, or lysates from cells expressing proteoglycans in the presence or absence of compounds of interest.


An assay for identifying and determining an activity of one or more of the compounds of the present invention comprises identifying compounds that interact with the promoter regions of a gene, or interact and effect proteins that interact with the promoter region, and are important in the transcriptional regulation of the protein's expression. For example, if perlecan were the protein, in general, the method comprises a vector comprising regulatory sequences of the perlecan gene and an indicator region controlled by the regulatory sequences, such as an enzyme, in a promoter-reporter construct. The protein product of the indicator region is referred to herein as a reporter enzyme or reporter protein. The regulatory region of the sequence of perlecan comprises a range of nucleotides from approximately −4000 to +2000 wherein the transcription initiation site is +1, alternatively, from −2500 to +1200, and still alternatively, from −1500 to +800 relative to the transcription initiation site.


Cells are transfected with a vector comprising the promoter-reporter construct and then treated with one or more compositions comprising at least one compound of the present invention. For example, the transfected cells are treated with a composition comprising a compound suspected of effecting the transcription of perlecan and the level of activity of the perlecan regulatory sequences are compared to the level of activity in cells that were not treated with the compound. The levels of activity of the perlecan regulatory sequences are determined by measuring the amount of the reporter protein or determining the activity of the reporter enzyme controlled by the regulatory sequences. An increase in the amount of the reporter protein or the reporter enzyme activity shows a stimulatory effect on perlecan, by positively effecting the promoter, whereas a decrease in the amount or the reporter protein or the reporter enzyme activity shows a negative effect on the promoter and thus, on perlecan.


Additionally, the present invention comprises methods and compositions that can be used with gene therapy methods and composition, such as those gene therapy methods comprising administering compositions comprising nucleic acids that effect the synthesis or expression of HSPGs, particularly perlecan. Such methods and compositions are taught in U.S. patent application Ser. No. 10/091,357, incorporated herein by reference.


The present invention comprises methods and compositions for mediating proteoglycan synthesis, expression and for the maintenance of SMC in a quiescent state. Methods and compositions of the present invention comprise treatment and prevention of vascular diseases and pathologies related to cellular proliferation, such as SMC proliferation. Such methods and compositions comprise methods for inhibition of SMC growth and proliferation, and for induction of quiescence in smooth muscle cells. Aspects of the present invention comprise methods and compositions for inducing proteoglycan synthesis, particularly HSPG synthesis and expression including, but not limited to, the induction of HSPGs such as syndecans, glypicans, and perlecans, and preferably perlecan synthesis and gene expression. Perlecan is a major extracellular HSPG in the blood vessel matrix. It interacts with extracellular matrix proteins, growth factors and receptors. Perlecan is also present in basement membranes other than blood vessels and in other extracellular matrix structures.


The activities of the compounds included in the present invention affect cells or tissues to increase the synthesis of proteoglycans by those cells or tissues or can act directly upon one or more proteoglycans to modulate the biological activity or to increase the biological stability of the proteoglycan itself, for example, of the protein perlecan. Activities also included herein are ones that increase the biosynthesis of one or more proteoglycans by increasing the transcription of the poteoglycan gene, increasing the biological stability of the proteoglycan mRNA or increasing the translation of proteoglycan mRNA into protein. Further activites include activities of compounds that can block or decrease the effects of agents or proteins that inhibit the activity of proteoglycans.


The present invention comprises methods and compositions for the treatment and prevention of smooth muscle cell proliferation, including vascular occlusive pathologies. Such methods comprise administration of compositions comprising compounds capable of inhibiting SMC proliferation, such as compositions comprising compounds disclosed herein that inhibit SMC proliferation. Administration of such compounds that are effective in inhibiting SMC proliferation are administered to humans and animals suspected of having or who have, for example, vasculopathy or who have undergone angioplasty or other procedures damaging to the endothelium. Effective amounts are administered to such humans and animals in dosages that are safe and effective, including, but not limited to, the ranges taught herein. Routes of administration include, but are not limited to, those disclosed herein. As disclosed herein, compositions comprising such compounds may be used in conjunction with other therapeutic agents or in methods comprising steps such as altered patient activities, including, but not limited to, changes in exercise or diet.


Glycosidase Modulation Activity


The present invention also comprises methods and compositions comprising compounds described herein that have an activity associated with modulation of glycosidase enzymes and thus, effecting the substrates for such enzymes. Glycosidase enzymes and their activity with their substrates, such as proteoglycans or glycated proteins, are aspects of a variety of diseases such as vascular conditions, including those conditions discussed supra, proteoglycan-associated diseases, supra, associated diseases with vascular components, including but not limited to, kidney disease, ischemic heart disease, cardiovascular disease, generalized vascular disease, proliferative retinopathy, macroangeopathy, inflammatory diseases, and metastatic diseases such as cancer, cellular proliferative conditions, and solid and blood borne tumors, or other oncological conditions. Compounds described herein that have an activity that affects the concentrations of substrates of glycosidase enzymes are used in methods of treatment of such vascular, inflammatory, metastatic, and systemic diseases.


Compounds or compositions comprising such compounds that are effective in modulating glycosidase enzyme activity are useful in treating and/or preventing cancer including, but not limited to, malignant and non-malignant cell growth, and the like. In another aspect of the present invention, the compounds disclosed herein are useful in modulating heparanase activity or the activity of other glycosidases as a means for treating and preventing autoimmune diseases.


Thus, the inhibition of heparanase or the activity of other glycosidases using the compounds of the present invention finds utitlity in treating arthritis and other autoimmune diseases. More specifically, the compounds of the present invention are useful in the treatment or prophylaxis of at least one autoimmune-related disease in a cell, tissue, organ, animal, or patient including, but not limited to, rheumatoid arthritis, juvenile rheumatoid arthritis, systemic onset juvenile rheumatoid arthritis, psoriatic arthritis, ankylosing spondilitis, gastric ulcer, seronegative arthropathies, osteoarthritis, inflammatory bowel disease, ulcerative colitis, systemic lupus erythematosis, antiphospholipid syndrome, iridocyclitis/uveitis/optic neuritis, idiopathic pulmonary fibrosis, systemic vasculitis/wegener's granulomatosis, sarcoidosis, orchitis/vasectomy reversal procedures, allergic/atopic diseases, asthma, allergic rhinitis, eczema, allergic contact dermatitis, allergic conjunctivitis, hypersensitivity pneumonitis, transplants, organ transplant rejection, graft-versus-host disease, systemic inflammatory response syndrome, sepsis syndrome, gram positive sepsis, gram negative sepsis, culture negative sepsis, fungal sepsis, neutropenic fever, urosepsis, meningococcemia, trauma/hemorrhage, burns, ionizing radiation exposure, acute pancreatitis, adult respiratory distress syndrome, rheumatoid arthritis, alcohol-induced hepatitis, chronic inflammatory pathologies, Crohn's pathology, sickle cell anemia, diabetes, nephrosis, atopic diseases, hypersensitity reactions, allergic rhinitis, hay fever, perennial rhinitis, conjunctivitis, endometriosis, asthma, urticaria, systemic anaphalaxis, dermatitis, pernicious anemia, hemolytic disesease, thrombocytopenia, graft rejection of any organ or tissue, kidney translplant rejection, heart transplant rejection, liver transplant rejection, pancreas transplant rejection, lung transplant rejection, bone marrow transplant (BMT) rejection, skin allograft rejection, cartilage transplant rejection, bone graft rejection, small bowel transplant rejection, fetal thymus implant rejection, parathyroid transplant rejection, xenograft rejection of any organ or tissue, allograft rejection, anti-receptor hypersensitivity reactions, Graves disease, Raynoud's disease, type B insulin-resistant diabetes, asthma, myasthenia gravis, type III hypersensitivity reactions, POEMS syndrome (polyneuropathy, organomegaly, endocrinopathy, monoclonal gammopathy, and skin changes syndrome), polyneuropathy, organomegaly, endocrinopathy, monoclonal gammopathy, skin changes syndrome, anti-phospholipid syndrome, pemphigus, scleroderma, mixed connective tissue disease, idiopathic Addison's disease, autoimmune hemolytic anemia, autoimmune hepatitis, idiopathic pulmonary fibrosis, scleroderma, diabetes mellitus, chronic active hepatitis, vitiligo, vasculitis, post-MI cardiotomy syndrome, type IV hypersensitivity, contact dermatitis, hypersensitivity pneumonitis, allograft rejection, granulomas due to intracellular organisms, drug sensitivity, metabolic/idiopathic, Wilson's disease, hemachromatosis, alpha-1-antitrypsin deficiency, diabetic retinopathy, Hashimoto's thyroiditis, osteoporosis, hypothalamic-pituitary-adrenal axis evaluation, primary biliary cirrhosis, thyroiditis, encephalomyelitis, cachexia, cystic fibrosis, neonatal chronic lung disease, chronic obstructive pulmonary disease (COPD), familial hematophagocytic lymphohistiocytosis, dermatologic conditions, psoriasis, alopecia, nephrotic syndrome, nephritis, glomerular nephritis, acute renal failure, hemodialysis, uremia, toxicity, preeclampsia, ankylosing spondylitis, Behcet's disease, bullous pemphigoid, cardiomyopathy, celiac sprue-dermatitis, chronic fatigue immune dysfunction syndrome (CFIDS), chronic inflammatory demyelinating polyneuropathy, Churg-Strauss syndrome, cicatricial pemphigoid, CREST syndrome, cold agglutinin disease, discoid lupus, essential mixed cryoglobulinemia, fibromyalgia-fibromyositis, Graves' disease, Guillain-Barré, Hashimoto's thyroiditis, idiopathic thrombocytopenia purpura (ITP), IgA nephropathy, insulin dependent diabetes, juvenile arthritis, lichen planus, ménière's disease, multiple sclerosis, pemphigus vulgaris, polyarteritis nodosa, Cogan's syndrome, polychondritis, polyglandular syndromes, polymyalgia rheumatica, polymyositis and dermatomyositis, primary agammaglobulinemia, Raynaud's phenomenon, Reiter's syndrome, rheumatic fever, Sjögren's syndrome, stiff-man syndrome, Takayasu arteritis, temporal arteritis/giant cell arteritis, Wegener's granulomatosis; okt3 therapy, anti-cd3 therapy, cytokine therapy, chemotherapy, radiation therapy (e.g., including but not limited toasthenia, anemia, cachexia, and the like), chronic salicylate intoxication, and the like.


Compounds having heparanase activity inhibition, that are effective for example, in treatment of cancer and autoimmune disease, can be determined using assays such as those disclosed in U.S. patent application Ser. No. 09/952,648, which is incorporated herein in its entirety. Such assays, which are used for measurement of cellular and enzymatic activities, both qualitatively and quantitatively, and in methods for diagnosing metastases, metastatic potential, and inflammatory states, are performed with and without the addition of at least one of the compounds of the present invention to determine the activity of the compound. Existing heparanase assays are taught in Goshen et al., 2 MOL. HUM. REPROD. 679-84 (1996); Nakajima et al., 31 CANCER LETR. 277-83 (1986); and Vlodasky et al., 12 INVASION METASTASIS 112-27 (1992); Freeman and Parish, 325 BIOCHEM. J. 229-37 (1997); Kahn and Newman, 196 ANAL. BIOCHEM. 373-76 (1991). Solid-phase heparanase assays have also been developed where chemically and biosynthetically radiolabeled heparin and HS chains were attached to a solid support, with release of radiolabel from the solid support being a measure of enzyme activity. Assays using such procedures are taught in U.S. Pat. No. 4,859,581, which is incorporated herein by reference in its entirety.


The present invention comprises methods and compositions for the treatment and prevention of diseases or conditions that present or result from glycosidase activity. Such methods comprise administration of compositions comprising compounds capable of modulating heparanase activity, such as compositions comprising compounds disclosed herein that inhibit heparanase activity. Administration of such compounds that are effective in modulating heparanase activity are administered to humans and animals suspected of having or who have, for example, inflammatory conditions, autoimmune disease, or diabetic vasculopathy. Effective amounts are administered to such humans and animals in dosages that are safe and effective, including, but not limited to, the ranges taught herein. Routes of administration include, but are not limited to, those disclosed herein. As disclosed herein, compositions comprising such compounds can be used in conjunction with other therapeutic agents or in methods comprising steps such as altered patient activities.


Inflammation Modulation


One aspect of the present invention comprises methods and compositions comprising compounds of the present invention for the treatment and prevention of conditions or diseases that have as an aspect of the disease or condition, inflammation. An aspect of the present invention is directed to methods and compositions comprising compounds that are effective in inhibiting inflammation, particularly inflammation associated with the accumulation or presence of glycated proteins or AGE. The activity of modulating inflammation includes, but is not limited to, inhibiting inflammation and/or its associated cell activation by glycated proteins or AGE, blocking the glycation of proteins, blocking AGE interactions with receptors, blocking AGE-induced signaling or signaling-associated inflammatory responses, cytokine induction, synthesis, or release, AGE formation, or AGE cross-linking.


The present invention also provides compositions for and methods of treatment of biological conditions including, but not limited to, vascular complications of type I and type II diabetes and atherosclerosis. Other inflammatory related diseases include, but are not limited to, rheumatoid arthritis, osteoarthritis, intraoccular inflammation, psoriasis, and asthma.


The compounds of the present invention have utility in inhibiting inflammation and/or its associated cell activation by glycated proteins or AGE. Pharmacological inhibition of AGE-induced cell activation provides the basis for therapeutic intervention in many diseases, notably in diabetic complications and Alzheimer's disease. Therapeutic approaches for inhibition of AGE-induced inflammation include, but are not limited to, blocking the glycation of proteins, blocking AGE interactions with receptors, and blocking AGE-induced signaling or signaling-associated inflammatory responses.


Compounds of the present invention that have at least the activity of modulating inflammation activity are shown in Table 4. The compounds shown in this Table have the activity of modulating inflammation activity as measured by the assays taught herein.

TABLE 4Compounds of the present invention that have at least the activity ofmodulating inflammation activityEntryStructureCompound Name 1embedded image3-chloro-4-methoxy-phenyl)-(2-phenyl- pyridin-4-yl)-amine 2embedded image(2,6-diphenyl-pyridin-4-yl)-p-tolyl-amine 3embedded image[2,6-bis-(4-fluoro-phenyl)-pyridin-4-yl]-(4- trifluoromethoxy-phenyl)-amine 4embedded image2,6-Bis-(3-methanesulfonyl-phenyl)-pyridin- 4-yl]-(4-trifluoromethoxy-phenyl)-amine 5embedded imageN-ethyl-3-[6-(3-methanesulfonyl-phenyl)-4- (4-trifluoromethoxy-phenylamino)-pyridin-2- yl]-benzamide 6embedded image1-{3-[6-(3-acetyl-phenyl)-4-(4- trifluoromethoxy-phenylamino)-pyridin-2- yl]-phenyl}-ethanone 7embedded image1-{4-[2,6-bis-(4-fluoro-phenyl)-pyridin-4- ylamino]-phenyl}-ethanone 8embedded image[2,6-bis-(3-N,N-dimethyl-benzamide)- pyridin-2-yl]-(4-trifluoromethoxy-phenyl)- amine 9embedded image[2,6-Bis-(4-fluoro-phenyl)-pyridin- 1-yl]-(3-methanesulfonyl-phenyl)-amine10embedded image[2-(3-Methanesulfonyl-phenyl)-6-pyr- rolidin-1-yl-pyridin-4-yl]-(4-trifluoro- methoxy-phenyl)-amine11embedded image1-{3-[6-pyrrolidin-1-yl-4-(4- trifluoromethoxy-phenylamino)-pyridin-2- yl]-phenyl}-ethanone12embedded image[2-(4-fluoro-phenyl)-6-(4-methyl-piperazin- 1-yl)-pyridin-4-yl]-(4-trifluoromethoxy- phenyl)-amine13embedded image1-[4-(2-morpholin-4-yl-6-phenyl-pyridin-4- ylamino)-phenyl]-ethanone14embedded image1-[4-(4-hydroxy-6′-phenyl-3,4,5,6- tetrahydro-2H-[1,2′]bipyridinyl-4′-ylamino)- phenyl]-ethanone15embedded image1-[3-(4-hydroxy-6′-phenyl-3,4,5,6- tetrahydro-2H-[1,2′]bipyridinyl-4′-ylamino)- phenyl]-ethanone16embedded image[2-(4-fluoro-phenyl)-6-morpholin-4-yl- pyridin-4-yl]-(4-trifluoromethoxy-phenyl)- amine17embedded image[2-(3-methanesulfonyl-phenyl)-6-morpholin- 4-yl-pyridin-4-yl]-(4-trifluoromethoxy- phenyl)-amine18embedded image1-{3-[6-morpholin-4-yl-4-(4- trifluoromethoxy-phenylamino)-pyridin-2- yl]-phenyl}-ethanone19embedded image{3-[6-morpholin-4-yl-4-(4-trifluoromethoxy- phenylamino)-pyridin-2-yl]-phenyl}- pyrrolidin-1-yl-methanone20embedded image[6′-(4-Fluoro-phenyl)-3,4,5,6-tetra- hydro-2H-[1,4′]bipyridinyl-2′-yl]- (4-trifluoromethoxy-phenyl)-amine21embedded image(6′-Phenyl-3,4,5,6-tetrahydro-2H- [1,4′]bipyridinyl-2′-yl)-(4-trifluoro- methoxy-phenyl)-amine22embedded image[6′-(3-Methanesulfonyl-phenyl)-3,4, 5,6-tetrahydro-2H-[1,4′]bipyridinyl- 2′-yl]-(4-trifluoromethoxy-phenyl)- amine23embedded image4-[6′-(4-Fluoro-phenyl)-3,4,5,6-tetra- hydro-2H-[1,4′]bipyridmyl-2′-yl- amino]-N-methyl-benzenesulfonamide24embedded image1-[4-(4-Hydroxy-6′-phenyl-3,4,5,6- tetrahydro-2H-[1,4′]bipyridinyl-2′-ylamino)- phenyl]-ethanone25embedded image2′,6′-Bis-(4-trifluoromethoxy-phenyl)- 3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl26embedded image(6-Phenyl-4-pyrrolidin-1-yl-pyridin-2-yl)-(4- trifluoromethoxy-phenyl)-amine27embedded image[6-(4-Fluoro-phenyl)-4-pyrrolidin-l-yl- pyridin-2-yl]-(4-trifluoromethoxy-phenyl)- amine28embedded imageN-Methyl-4-[4-pyrrolidin-1-yl-6-(4- trifluoromethoxy-phenyl)-pyridin-2- ylamino]-benzenesulfonamide29embedded imageN-Methyl-4-[4-(4-methyl-piperazin-1-yl)-6- (4-trifluoromethoxy-phenylamino)-pyridin-2- yl]-benzenesulfonamide30embedded image[6-(3-Methanesulfonyl-phenyl)-4-morpholin- 4-yl-pyridin-2-yl]-(4-trifluoromethoxy- phenyl)-amine31embedded imageN-Methyl-4-[4-morpholin-4-yl-6-(4- trifluoromethoxy-phenylamino)-pyridin-2- yl]-benzenesulfonamide32embedded image(4-Fluoro-phenyl)-[6-(4-trifluoromethoxy- phenyl)-pyridin-2-yl]-amine33embedded image2′,6′-Bis-(4-trifluoromethoxy-phenylamino)- 3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-4-ol


The inclusion of compounds in the categories of the tables disclosed herein are not to be seen as limiting, in that compounds included in such tables have at least the activity shown for inclusion in the table and may have more or other activities. Nor are the tables to be seen as limiting in that these are the only compounds disclosed herein that have that activity, representative compounds are shown in the tables that have at least that particular activity for inclusion in the table. One or more compounds disclosed herein have at least an activity that has utility in treatment of disease states.


The activity of the compounds of the present invention in inhibiting glycated protein- and AGE-induced inflammation can be determined using the assays described herein and in U.S. patent application Ser. No. 10/026,335, which is incorporated by reference herein in its entirety. Such assays comprise measurement of the specific activity of biological components involved in a known cellular response. The assays provide a measurable response in which the activity of the compounds is determined. One assay comprises measurement of the effects of compounds on an inflammatory response by cells to the presence of a stimulating agent. Yet another assay comprises endothelial cells that are stimulated by the addition of a glycated protein, the stimulating agent. The endothelial cells respond by producing specific cytokines. The amount of cytokines produced are determined by measurement protocols known to those skilled in the art. The compounds of the present invention are then added to the assay and the production of cytokines is measured. From the comparison of the assay without the compound with the assay with the compound, the biological effect of the compound can be determined. The compound may have an inhibitory effect, a stimulatory effect, or no effect at all.


The amount and type of cytokine produced can be determined using immunological methods, such as ELISA assays. The methods of the present invention are not limited by the type of assay used to measure the amount of cytokine produced, and any methods known to those skilled in the art and later developed can be used to measure the amount of cytokines produced in response to the stimulating agent and to the compound having unknown activity.


An aspect of the present invention comprises methods and compositions for the treatment of diseases, preconditions, or pathologies associated with inflammatory cytokines and other inflammation related molecules including, but not limited to IL-6, VCAM-1, or AGE-induced MCP-1, (monocyte chemoattractant protein 1).


Assays for determining the activity of compounds capable of modulating inflammation include those taught in U.S. patent application Ser. Nos. 10/026,335 and 09/969,013, which are both expressly incorporated by reference in their entireties. In general, once the baseline response to the stimulating agent for the production of cytokines by the endothelial cells is established, thus comprising the control levels for the screening assay, the methods comprise addition of compounds of the present invention. The effect of the compound on the baseline response is determined by comparing the amount of cytokine produced in the presence of the stimulating agent and the amount of cytokine produced in the presence of the stimulating agent and the compound of the present invention. In one aspect, compounds that have inhibitory effects on the inflammation of the cells in the presence of glycated albumin are then used as therapeutic agents. One or more compounds can be added to the screening assay. Combinations or mixtures of compounds can be added. Different amounts and formulations of the compounds are added to determine the effects on the screening assay. The screening assay can also be used to determine stimulatory compounds or compounds that have no effects in the assay.


The present invention comprises methods and compositions for the treatment and prevention of disease, conditions and pathologies associated with inflammation. Such methods comprise administration of compositions comprising compounds capable of modulating the activity of molecules associated with inflammation such as AGE or cytokines or other cellular factors, including release rates or activity, and include compositions comprising compounds disclosed herein with inflammation modulating activity. Administration of such compounds that are effective in modulating inflammation are administered to humans and animals suspected of having or who have inflammatory diseases, for example, diabetic-induced vasculopathies, autoimmune diseases, renal insufficiency, Alzheimer's syndrome, and inflammation-induced diseases such as atherosclerosis. Effective amounts are administered to such humans and animals in dosages that are safe and effective, including, but not limited to, the ranges taught herein. Routes of administration include, but are not limited to, those disclosed herein. As disclosed herein, compositions comprising such compounds can be used in conjunction with other therapeutic agents or in methods comprising steps such as altered patient activities, including, but not limited to, changes in exercise or diet.


Correlation of Physiological Parameters and Assays to Diseases and Conditions


The following Tables 5-8 provide disclosure and references that relate the various physiological parameters and assays disclosed herein to general and specific diseases, disease states, and conditions. Among other things, the references and citations provided in these tables support the specification as fully enabled for treating or modulating all the diseases or conditions encompassed herein, based on the inhibiting activity of the compounds provided in the specification, and the predictive nature of the tests provided of the disclosed uses. In particular, Tables 5-8 provide specific references that link the parameters measured in the key assays disclosed in the application with a specific physiology, pathophysiology, or medical condition.


Table 5 provides scientific references that demonstrate, among other things, the connection between TNF-α and IL-6 in rheumatoid arthritis, vascular inflammation, and atherosclerosis. For example, these references demonstrate the importance of TNF inhibition in preventing rheumatoid arthritis, the therapeutic benefit of IL-6 inhibition in rheumatoid arthritis as well as its importance in preventing rheumatoid arthritis, the role of AGE in different diabetic vascular diseases, and AGE inhibition as a therapeutic strategy for vascular complications.


Further, Table 6 provides scientific references that demonstrate, among other things, the importance of HSPG in the prevention of atherosclerosis and diabetic vascular disease. For example, these references demonstrate that atherosclerotic vessels have reduced HSPG, and that cholesterol deposition is inversely correlated to HSPG content in the vessel.


Table 7 also provides scientific references that demonstrate, among other things, the connection between smooth muscle cell (SMC) proliferation in contributing to restenosis and atherosclerosis. For example, these references demonstrate that: smooth muscle proliferation contributes to unstable angina and restenosis; inhibition of SMC proliferation by LRP is important for atherosclerosis prevention; and the function of the SMC inhibitor, rapamycin, in preventing restenosis and vein graft disease.


Table 8 provides scientific references that demonstrate, among other things, the role of heparanase and TNF-α in promoting tumor angiogenesis and metastasis, as well as the use of inhibitors of heparanase and TNF-α in treating cancer. For example, these references demonstrate the role of heparanase inhibitors in treating tumor angiogenesis and metastasis, the role of TNF-α as a tumor-promoting agent, and the use of TNF-α inhibitors in the treatment of cancer.


The key assays described herein for screening the compounds in the present invention include, but are not limited to: a) the inhibition of smooth muscle cell (SMC) proliferation, that was used to identify, for example, compounds in Table 3; b) the induction of HSPG in smooth muscle cells; c) the induction of heparanase in endothelial cells; d) the inhibition of AGE-induced inflammatory response in endothelial cells as measured by IL-6 or other inflammatory cytokines, that was used to identify, for example, compounds in Table 4; and e) cytotoxicity effects of the disclosed compounds. By using these disclosed assays, the present disclosure is fully enabled for identification of compounds for the treatment of the diseases disclosed generically and specifically.


Accordingly, this evidence along with the references of Tables 5-8 demonstrate that the parameters measured in the key assays above are associated with and predictive of the specific physiology, pathophysiology, or medical conditions disclosed herein. The physiology, pathophysiology, or medical conditions disclosed include generically disclosed conditions and diseases such as, but are not limited to, unwanted cellular proliferation, inflammation mediated diseases, hyperproliferative diseases, and diseases involving a glycosidase. Specifically disclosed diseases include, but are not limited to, restenosis, vascular occlusive diseases, arthritis, cancer, and the like. Therefore, methods of treating diseases, disease states, or conditions disclosed in the specification, or methods of modulating, for example, the production or uptake of a biologically-active chemical, are disclosed in such as way as to allow the skilled artisan to make and use the invention, the tests provided are predictive of the claimed uses, and therefore are fully enabled for all the diseases or conditions encompassed therein.

TABLE 5The Role of TNF-α, IL-6, and AGE in Rheumatoid Arthritis, Vascular Inflammation, and Atherosclerosis.Pages inreferencearguing orReferencePhysiologicalshowingAuthorTitle of ReferenceCitationParameterDiseaseconnectionOther commentsFeldmann MDiscovery of TNF-α asJoint Bone Spine.TNFArthritisAllReview detailing theRef 1a therapeutic target in2002inhibitionimportance of TNF-αrheumatoid arthritis:Jan; 69(1): 12-8inhibition in preventingpreclinical and clinicalReviewrheumatoid arthritisstudiesChoy et alTherapeutic benefit ofArthritis Rheum.IL-6Arthritis3144Human trial showing theRef 2blocking interleukin-62002inhibition(abstract),therapeutic benefit of IL-6activity with an anti-Dec; 46(12): 3143-503146inhibition in rheumatoidinterleukin-6 receptorarthritismonoclonal antibody inrheumatoid arthritis: arandomized, double-blind, placebo-controlled, dose-escalation trial.Wong et alThe role of theArthritis Rheum.IL-6Arthrits1177 para 4Review detailing theRef 3interleukin-6 family of2003inhibitionimportance of IL-6 incytokines inMay; 48(5): 1177-89.preventing rheumatoidinflammatory arthritisReviewarthritisand bone turnoverBasta et alAdvanced glycationCardiovasc Res.AGE-IL6Diabetic582, 589Highlights the role of AGE inRef 4end products and2004 Sepinhibitionvasculardifferent diabetic vascularvascular inflammation:1; 63(4): 582-92diseasesdiseases and AGE inhibitionimplications foras a therapeutic strategy foracceleratedvascular complicationsatherosclerosis indiabetes









TABLE 6










The Potential Role of HSPG Induction in the Prevention of Atherosclerosis and Diabetic Vascular Disease.


















Pages in








reference







arguing or




Reference
Physiological

showing


Author
Title of Reference
Citation
Parameter
Disease
connection
Other comments





Engelberg H.
Endogenous heparin
Atherosclerosis.
HSPG
Atherosclerosis
All
Review detailing the


Ref 5
activity deficiency: the
2001
induction


importance of HSPG in



‘missing link’ in
Dec; 159(2): 253-60.



preventing events related



atherogenesis?
Review



to atherosclerosis








development


Jensen T
Pathogenesis of
Diabetes. 1997
HSPG
Diabetic
All
Review detailing the


Ref 6
diabetic vascular
Sep; 46 Suppl
induction
vascular

importance of HSPG in



disease: evidence for
2: S98-100

disease

preventing diabetic



the role of reduced




vascular disease



heparan sulfate



proteoglycan


Hollmann J
Relationship of
Artherosclerosis.
HSPG
Atherosclerosis

Data show that


et al,
sulfated
1989; 9: 154-8



atherosclerotic vessels


Ref 7
glycosaminoglycans




have reduced HSPG and



and cholesterol




that cholesterol deposition



content in normal and




is inversely correlated to



atherosclerotic human




HSPG content in the



aorta




vessel


Kruse R et
Cholesterol-dependent
Basic Res
HSPG
Atherosclerosis

Data show that


al
changes of
Cardiol. 1996



atherosclerotic vessels


Ref 8
glycosaminoglycan
Sep-



have reduced HSPG and



pattern in human aorta
Oct; 91(5): 344-52



that cholesterol deposition








is inversely correlated to








HSPG content in the








vessel
















TABLE 7










The Role of Smooth Muscle Cell (SMC) Proliferation in Restenosis and Atherosclerosis.


















Pages in








reference







arguing or




Reference
Physiological

showing


Author
Title of Reference
Citation
Parameter
Disease
connection
Other comments





Chen et al
Electron microscopic
Circulation. 1997
Smooth
Restenosis
1175
Data suggest that smooth


Ref 9
studies of phenotypic
Mar
muscle cell

(Conclusion)
muscle proliferation



modulation of smooth
4; 95(5): 1169-75
(SMC)


contributes to unstable



muscle cells in

proliferation


angina and restenosis



coronary arteries of



patients with unstable



angina pectoris and



postangioplasty



restenosis


Braun-
Cell cycle progression:
Circulation. 1998
Smooth
Restenosis
82
Review detailing the role of


Dullaeus
new therapeutic target
Jul 7; 98(1): 82-9
muscle cell


smooth muscle


et al
for vascular

(SMC)


proliferation in restenosis


Ref 10
proliferative disease

proliferation


and pharmacological








approaches to inhibit cell








cycle progression


Boucher et
LRP: role in vascular
Science. 2003
Smooth
Atherosclerosis
Abstract
Study shows that inhibition


al
wall integrity and
Apr
muscle cell


of SMC proliferation by


Ref 11
protection from
11; 300(5617): 329-32
(SMC)


LRP (lipoprotein receptor-



atherosclerosis

proliferation


related protein) is critical








for atherosclerosis








prevention


Marx et al
Bench to bedside: the
Circulation. 2001
Smooth
Restenosis
852
Review highlighting the


Ref 12
development of
Aug
muscle cell


role of smooth muscle cell



rapamycin and its
21; 104(8): 852-5
(SMC)


proliferation in restenosis



application to stent

proliferation


and the application of



restenosis




smooth muscle cell








inhibitor, rapamycin, in








preventing restenosis and








vein graft disease
















TABLE 8










The Role of Heparanase and TNF-α in Promoting Tumor Angiogenesis and Metastasis and the Use of Heparanase


and TNF-α Inhibitors in Treating Cancer.


















Pages in








reference







arguing or





Physiological

showing


Author
Title of Reference
Reference Citation
Parameter
Disease
connection
Other comments





Vlodavsky I
Molecular properties
J Clin Invest. 2001
Heparanase
Cancer
All
Review detailing the role of


et al
and involvement of
Aug; 108(3): 341-7.
inhibition


heparanase in promoting


Ref 13
heparanase in cancer
Review



tumor angiogenesis and



metastasis and




metastasis



angiogenesis


Goldshmidt
Cell surface
Proc Natl Acad Sci
Heparanase
Cancer
10031, 10036
Study showing that


et al
expression and
U S A. 2002 Jul
inhibition


heparanase promotes


Ref 14
secretion of
23; 99(15): 10031-6



angiogenesis and tumor



heparanase markedly




metastasis in animal



promote tumor




models.



angiogenesis and



metastasis


Simizu et al
Heparanase as a
Cancer Sci. 2004
Heparanase
Cancer
553, 557
Review detailing the role of


Ref 15
molecular target of
Jul; 95(7): 553-8
inhibition


heparanase inhibitors in



cancer chemotherapy




tumor angiogenesis and








metastasis


Szlosarek
Tumour necrosis factor
The Lancet
TNFα
Cancer
565
Review highlighting the role


et al
α: a potential target for
Oncology 2003
inhibition


TNFα as a tumor promoting


Ref 16
the therapy of solid
Sept; 4: 565-73



agent and the use of TNF



tumours




inhibitors in the treatment of








cancer










Compound/Composition-Coated Medical Devices


The compounds of the present invention can be used alone or in combination with other agents along with delivery devices to effectively prevent and treat the diseases described herein, though particular applications are found in vascular disease, and in particular, vascular disease caused by injury and/or by transplantation. Though this example focuses on vascular disease, provision of the compounds of the present invention with medical devices for treatment of the diseases and conditions capable of being treated with the compounds is contemplated by the present invention.


Various medical treatment devices utilized in the treatment of vascular disease can ultimately induce further complications. For example, balloon angioplasty is a procedure utilized to increase blood flow through an artery and is the predominant treatment for coronary vessel stenosis. However, the procedure typically causes a certain degree of damage to the vessel wall, thereby creating new problems or exacerbating the original problem at a point later in time. Although other procedures and diseases may cause similar injury, exemplary aspects of the present invention will be described with respect to the treatment of restenosis and related complications following percutaneous transluminal coronary angioplasty and other similar arterial/venous procedures, including the joining of arteries, veins and other fluid carrying conduits in other organs or sites of the body, such as the liver, lung, bladder, kidney, brain, prostate, neck and legs.


The local delivery of a compound of the present invention and, in some aspects, along with other therapeutic agents, from a stent prevents vessel recoil and remodeling through the scaffolding action of the stent. The activity of compound provided, with or without other therapeutic agents, helps determine for which application, to treat which disease, the coated medical device is being administered. For example, compound-coated stents can prevent multiple components of neointimal hyperplasia or restenosis as well as reduce inflammation and thrombosis. Local administration of a compound of the present invention and other therapeutic agents to stented coronary arteries may also have additional therapeutic benefit. For example, higher tissue concentrations of the compounds of the present invention and other therapeutic agents may be achieved utilizing local delivery rather than systemic administration. In addition, reduced systemic toxicity may be achieved utilizing local delivery rather than systemic administration while maintaining higher tissue concentrations. In utilizing local delivery from a stent rather than systemic administration, a single procedure may suffice with better patient compliance. An additional benefit of combination therapeutic agent and/or compound therapy can be to reduce the dose of each of the therapeutic agents, thereby limiting toxicity, while still achieving a reduction in restenosis, inflammation and thrombosis. Local stent-based therapy is therefore a means of improving the therapeutic ratio (efficacy/toxicity) of anti-restenosis, anti-inflammatory, and anti-thrombotic therapeutic agents.


Although exemplary aspects of the invention will be described with respect to the treatment of restenosis and other related complications, it is important to note that the local delivery of a compound of the present invention, alone or as part of a therapeutic agent combination, can be utilized to treat a wide variety of conditions utilizing any number of medical devices, or to enhance the function and/or life of the device. For example, intraocular lenses, placed to restore vision after cataract surgery is often compromised by the formation of a secondary cataract. The latter is often a result of cellular overgrowth on the lens surface and can be potentially minimized by combining one or more compounds of the present invention having activity that is effective in preventing unwanted cellular growth with the device. Other medical devices that often fail due to tissue in-growth or accumulation of proteinaceous material in, on and around the device, such as shunts for hydrocephalus, dialysis grafts, colostomy bag attachment devices, ear drainage tubes, leads for pace makers, and implantable defibrillators can also benefit from the combinations of the compounds of the present invention, possibly other pharmaceutical agents, and the devices. Other surgical devices, sutures, staples, anastomosis devices, vertebral disks, bone pins, suture anchors, hemostatic barriers, clamps, screws, plates, clips, vascular implants, tissue adhesives and sealants, tissue scaffolds, various types of dressings, bone substitutes, intraluminal devices, and vascular supports can also provide enhanced patient benefit using this compound-device combination approach. Essentially, any type of medical device can be coated in some fashion with at least one compound of the present invention, alone or as part of a therapeutic agent combination, which enhances treatment over the use of the device or therapeutic agent without combination with the compound.


As disclosed supra, the compounds of the present invention can be administered in combinational therapies with other therapeutic agents, and are not limited to only the other therapeutic agents disclosed herein. Thus, the present invention also contemplates, in addition to various medical devices; the coatings on these devices may be used to deliver a compound of the present invention in combination with other therapeutic agents. This illustrative list of therapeutic agents can be administered through pharmeutical means or in association with medical devices and such therapeutic agents include, but are not limited to, antiproliferative/antimitotic agents including natural products such as vinca alkaloids (e.g., vinblastine, vincristine, and vinorelbine), paclitaxel, epidipodophyllotoxins (e.g., etoposide, teniposide), antibiotics [e.g., dactinomycin (actinomycin D) daunorubicin, doxorubicin and idarubicin], anthracyclines, mitoxantrone, bleomycins, plicamycin (mithramycin) and mitomycin, enzymes (L-asparaginase which systemically metabolizes L-asparagine and deprives cells which do not have the capacity to synthesize their own asparagine); antiplatelet agents such as G(GP) IIb/IIIa inhibitors and vitronectin receptor antagonists; antiproliferative/antimitotic alkylating agents such as nitrogen mustards (e.g., mechlorethamine, cyclophosphamide, and analogs, melphalan, chlorambucil), ethylenimines and methylmelamines (hexamethylmelamine and thiotepa), alkyl sulfonates-busulfan, nirtosoureas [carmustine (BCNU) and analogs, streptozocin], trazenes-dacarbazinine (DTIC); antiproliferative/antimitotic antimetabolites such as folic acid analogs (methotrexate), pyrimidine analogs (e.g., fluorouracil, floxuridine, and cytarabine), purine analogs and related inhibitors [mercaptopurine, thioguanine, pentostatin and 2-chlorodeoxyadenosine (cladribine)]; platinum coordination complexes (cisplatin, carboplatin), procarbazine, hydroxyurea, mitotane, aminoglutethimide; hormones (e.g., estrogen); anticoagulants (e.g., heparin, synthetic heparin salts and other inhibitors of thrombin); fibrinolytic agents (such as tissue plasminogen activator, streptokinase and urokinase), aspirin, dipyridamole, ticlopidine, clopidogrel, abciximab; antimigratory; antisecretory (breveldin); anti-inflammatory agents such as adrenocortical steroids (e.g., cortisol, cortisone, fludrocortisone, prednisone, prednisolone, 6α-methylprednisolone, triamcinolone, betamethasone, and dexamethasone), non-steroidal agents (salicylic acid derivatives, i.e., aspirin; para-aminophenol derivatives, i.e., acetominophen; indole and indene acetic acids (indomethacin, sulindac, and etodalac), heteroaryl acetic acids (tolmetin, diclofenac, and ketorolac), arylpropionic acids (ibuprofen and derivatives), anthranilic acids (mefenamic acid, and meclofenamic acid), enolic acids (piroxicam, tenoxicam, phenylbutazone, and oxyphenthatrazone), nabumetone, gold compounds (auranofin, aurothioglucose, gold sodium thiomalate); immunosuppressives, (Cyclosporine, tacrolimus (FK-506), sirolimus (rapamycin), azathioprine, mycophenolate mofetil); angiogenic agents: vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF); angiotensin receptor blockers; nitric oxide donors; anti-sense oligionucleotides and combinations thereof, cell cycle inhibitors; mTOR inhibitors; and growth factor signal transduction kinase inhibitors.


Although any number of stents can be utilized in accordance with the present invention, for simplicity, a limited number of stents will be described in exemplary aspects of the present invention. The skilled artisan will recognize that any number of stents can be utilized in connection with the present invention. In addition, as stated above, other medical devices can be utilized. For example, though stents are described, sleeves outside the vessels are also contemplated, as are other medical devices that can provide a substrate for administration for at least one of the compounds of the present invention.


A stent is commonly used as a tubular structure left inside the lumen of a duct to relieve an obstruction. Typically, stents are inserted into the lumen in a non-expanded form and are then expanded autonomously, or with the aid of a second device in situ. A common method of expansion occurs through the use of a catheter-mounted, angioplasty balloon that is inflated within the stenosed vessel or body passageway in order to shear and disrupt the obstructions associated with the wall components of the vessel and to obtain an enlarged lumen.


A stent may resemble an expandable cylinder and may comprise a fenestrated structure for placement in a blood vessel, duct, or lumen to hold the vessel, duct, or lumen open, more particularly for protecting a segment of artery from restenosis after angioplasty. The stent can be expanded circumferentially and maintained in an expanded configuration that is circumferentially or radially rigid. The stent can be axially flexible and when flexed at a band, for example, the stent avoids any externally protruding component parts.


The stent can be fabricated utilizing any number of methods. For example, the stent can be fabricated from a hollow or formed stainless steel tube that can be machined using lasers, electric discharge milling, chemical etching, or other means. The stent is inserted into the body and placed at the desired site in an unexpanded form. In one aspect, expansion can be effected in a blood vessel by a balloon catheter, where the final diameter of the stent is a function of the diameter of the balloon catheter used. It should be appreciated that a stent in accordance with the present invention can be embodied in a shape-memory material including, for example, an appropriate alloy of nickel and titanium or stainless steel.


Structures formed from stainless steel can be made self-expanding by configuring the stainless steel in a predetermined manner, for example, by twisting it into a braided configuration. In this aspect, after the stent has been formed it can be compressed so as to occupy a space sufficiently small as to permit its insertion in a blood vessel or other tissue by insertion means, wherein the insertion means include a suitable catheter, or flexible rod. Upon emerging from the catheter, the stent can be configured to expand into the desired configuration where the expansion is automatic or triggered by a change in pressure, temperature, or electrical stimulation.


Furthermore, a stent can be modified to comprise one or more reservoirs. Each of the reservoirs can be opened or closed as desired. These reservoirs can be specifically designed to hold the compound or compound/therapeutic agent combination to be delivered. Regardless of the design of the stent, the compound or compound/therapeutic agent combination dosage can be applied with enough specificity and a sufficient concentration to provide an effective dosage in the effected area. In this regard, the reservoir size in the bands is preferably sized to adequately apply the compound or compound/therapeutic agent combination dosage at the desired location and in the desired amount.


In an alternative aspect, the entire inner and outer surface of the stent can be coated with the compound or compound/therapeutic agent combination in therapeutic dosage amounts. The coating techniques can vary depending on the compound or compound/therapeutic agent combination. Also, the coating techniques can vary depending on the material comprising the stent or other intraluminal medical device.


One or more compounds of the present invention and, in some instances, other therapeutic agents as a combination, can be incorporated onto or affixed to the stent in a number of ways. In one aspect, the compound is directly incorporated into a polymeric matrix and sprayed onto the outer surface of the stent. The compound elutes from the polymeric matrix over time and enters the surrounding tissue. The compound can remain on the stent for at least three days up to approximately six months, and, in another aspect, preferably between seven and thirty days.


Any number of non-erodible polymers may be utilized in conjunction with the compound, and such polymeric compositions are well known in the art. In one aspect, the polymeric matrix comprises two layers. The base layer comprises a solution of poly(ethylene-co-vinylacetate) and polybutylmethacrylate. The compound is incorporated into this base layer. The outer layer comprises only polybutylmethacrylate and acts as a diffusion barrier to prevent the compound from eluting too quickly. The thickness of the outer layer or topcoat determines the rate at which the compound elutes from the matrix. Essentially, the compound elutes from the matrix by diffusion through the polymer matrix. Polymers are permeable, thereby allowing solids, liquids and gases to escape therefrom. The total thickness of the polymeric matrix is in the range from about one micron to about twenty microns or greater. It is important to note that primer layers and metal surface treatments can be utilized before the polymeric matrix is affixed to the medical device. For example, acid cleaning, alkaline (base) cleaning, salinization and parylene deposition may be used as part of the overall process described above.


The poly(ethylene-co-vinylacetate), polybutylmethacrylate, and compound solution can be incorporated into or onto the stent in a number of ways. For example, the solution can be sprayed onto the stent or the stent can be dipped into the solution. Other methods include spin coating and plasma polymerization. In one aspect, the solution is sprayed onto the stent and then allowed to dry. In another aspect, the solution can be electrically charged to one polarity and the stent electrically charged to the opposite polarity. In this manner, the solution and stent will be attracted to one another. In using this type of spraying process, waste can be reduced and more precise control over the thickness of the coat may be achieved.


Drug-coated stents are manufactured by a number of companies including Johnson & Johnson, Inc. (New Brunswick, N.J.), Guidant Corp. (Santa Clara, Calif.), Medtronic, Inc. (Minneapolis, Minn.), Cook Group Incorporated (Bloomington, Ind.), Abbott Labs., Inc. (Abbott Park, Ill.), and Boston Scientific Corp. (Natick, Mass.). See e.g., U.S. Pat. No. 6,273, 913; U.S. Patent Application No. 20020051730; WO 02/26271; and WO 02/26139, each expressly entirely incorporated herein by reference.


Pharmaceutical Compositions


In one aspect, the present invention provides a composition comprising at least one compound as disclosed herein.


In another aspect, this invention provides a pharmaceutical composition, comprising:


at least one compound as disclosed herein; and


optionally comprising a pharmaceutically acceptable additive selected from a carrier, an auxiliary, a diluent, an excipient, a preservative, a solvate, or any combination thereof.


In yet another aspect, this invention provides a pharmaceutical composition, comprising:


at least one compound as disclosed herein; and


optionally comprising a pharmaceutically acceptable additive selected from a carrier, an auxiliary, a diluent, an excipient, a preservative, a solvate, or any combination thereof;


wherein the pharmaceutical composition is in the form of a tablet, a capsule, a syrup, a cachet, a powder, a granule, a solution, a suspension, an emulsion, a bolus, a lozenge, a suppository, a cream, a gel, a paste, a foam, a spray, an aerosol, a microcapsule, a liposome, or a transdermal patch.


In still another aspect, this invention provides a pharmaceutical composition, comprising:


at least one compound as disclosed herein;


optionally comprising a pharmaceutically acceptable additive selected from a carrier, an auxiliary, a diluent, an excipient, a preservative, a solvate, or any combination thereof; and


further comprising an agent selected from a chemotherapeutic agent, an immunosuppressive agent, a cytokine, a cytotoxic agent, an anti-inflammatory agent, an antirheumatic agent, a cardiovascular agent, or any combination thereof.


Accordingly, in addition to the compounds disclosed herein, the pharmaceutical compositions of the present invention can further comprise at least one of any suitable auxiliary such as, but not limited to, diluent, binder, stabilizer, buffers, salts, lipophilic solvents, preservative, adjuvant, or the like. In one aspect of the present invention, pharmaceutically acceptable auxiliaries are employed. Examples and methods of preparing such sterile solutions are well known in the art and can be found in well known texts such as, but not limited to, REMINGTON'S PHARMACEUTICAL SCIENCES (Gennaro, Ed., 18th Edition, Mack Publishing Co. (1990)). Pharmaceutically acceptable carriers can be routinely selected that are suitable for the mode of administration, solubility and/or stability of the compound.


Pharmaceutical Compositions for Oral Administration


For oral administration in the form of a tablet or capsule, a compound can be combined with an oral, non-toxic pharmaceutically acceptable inert carrier such as ethanol, glycerol, water, and the like. Moreover, when desired or necessary, suitable binders, lubricants, disintegrating agents, and coloring agents may also be incorporated into the mixture. Suitable binders include, without limitation, starch; gelatin; natural sugars such as glucose or beta-lactose; corn sweeteners; natural and synthetic gums such as acacia, tragacanth, or sodium alginate, carboxymethylcellulose; polyethylene glycol; waxes; and the like. Lubricants used in these dosage forms include, without limitation, sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride, and the like. Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum, and the like.


Formulations of the present invention suitable for oral administration can be presented as discrete units such as capsules, cachets, or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous liquid or a non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil emulsion and as a bolus, and the like.


Routes of Administration


The invention further relates to the administration of at least one compound disclosed herein by the following routes, including, but not limited to oral, parenteral, subcutaneous, intramuscular, intravenous, intrarticular, intrabronchial, intraabdominal, intracapsular, intracartilaginous, intracavitary, intracelial, intracelebellar, intracerebroventricular, intracolic, intracervical, intragastric, intrahepatic, intramyocardial, intraosteal, intrapelvic, intrapericardiac, intraperitoneal, intrapleural, intraprostatic, intrapulmonary, intrarectal, intrarenal, intraretinal, intraspinal, intrasynovial, intrathoracic, intrauterine, intravesical, bolus, vaginal, rectal, buccal, sublingual, intranasal, iontophoretic means, or transdermal means.


Dosages


More specifically, the pharmaceutical compositions can be administered in a single daily dose, or the total daily dosage can be administered in divided doses of two, three, or four times daily. In the case of oral administration, the daily dosage of the compositions can be varied over a wide range from about 0.0001 to about 1,000 mg per patient, per day. Alternatively, the range can be from about 0.001 mg/kg to about 10 mg/kg of body weight per day, about 0.1 to about 100 mg, about 1.0 to about 50 mg or about 1.0 to about 20 mg per day for adults (at about 60 kg).


The daily dosage of the pharmaceutical compositions may be varied over a wide range from about 0.01 to about 1000 mg per adult human per day. For oral administration, the pharmaceutical compositions can be provided in the form of tablets containing from about 0.1 mg to about 1000 mg of the compound or about 0.1, 0.2, 0.5, 1.0, 2.0, 5.0, 10.0, 15.0, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 800, 900, or 1000 milligrams of the active compound for the symptomatic adjustment of the dosage to the patient to be treated. An effective amount of the drug is ordinarily supplied at a dosage level of from about 0.1 mg/kg to about 20 mg/kg of body weight per day. In one aspect, the range is from about 0.2 mg/kg to about 10 mg/kg of body weight per day. In another aspect, the range is from about 0.5 mg/kg to about 10 mg/kg of body weight per day. The compounds may be administered on a regimen of about 1 to about 10 times per day.


In the case of injections, it is usually convenient to give by an intravenous route in an amount of about 0.01 to about 30 mg, about 0.1to about 20 mg or about 0.1 to about 10 mg per day to adults (at about 60 kg). In the case of other animals, the dose calculated for 60 kg may be administered as well.


In addition, co-administration or sequential administration of the compounds of the present invention and other therapeutic agents can be desirable, such as chemotherapeutic agents, immunosuppressive agents, cytokines, cytotoxic agents, nucleolytic compounds, radioactive isotopes, receptors, and pro-drug activating enzymes, which can be naturally occurring or produced by recombinant methods. The combined administration includes co-administration, using separate formulations or a single pharmaceutical formulation, and consecutive administration in either order, wherein there is a time period while both (or all) active therapeutic agents simultaneously exert their biological activities.


It is to be understood that this invention is not limited to the particular methodology, syntheses, formulations, protocols, cell lines, constructs, and reagents described herein and as such can vary. It is also to be understood that the terminology used herein is for the purpose of describing particular aspects only, and is not intended to limit the scope of the present invention.


All publications, patents, and other references mentioned herein are provided for the purpose of describing and disclosing, for example, the constructs and methodologies that are described in these references, which might be used in connection with the presently described invention. The references provided or discussed in the text are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the inventors are not entitled to antedate such disclosure by virtue of prior invention.


To the extent that any definition or usage provided by any document incorporated herein by reference conflicts with the definition or usage provided herein, the definition or usage provided herein controls.


For any particular compound disclosed herein, any general structure presented also encompasses all conformational isomers, regioisomers, stereoisomers and tautomers that can arise from a particular set of substituents. The general structure also emcompasses all enantiomers, diastereomers, and other optical isomers whether in enantiomeric or racemic forms, as well as mixtures of stereoisomers, as the context requires. The general structure also encompasses all salts, including pharmaceutically acceptable and non-pharmaceutically acceptable salts and prodrugs thereof.


When Applicants disclose or claim a range of any type, for example a range of temperatures, a range of numbers of atoms, a molar ratio, or the like, Applicants' intent is to disclose or claim individually each possible number that such a range could reasonably encompass, as well as any sub-ranges and combinations of sub-ranges encompassed therein. For example, when the Applicants disclose or claim a chemical moiety having a certain number of carbon atoms, Applicants' intent is to disclose or claim individually every possible number that such a range could encompass, consistent with the disclosure herein. For example, the disclosure that R is selected independently from an alkyl group having up to 20 carbon atoms, or in alternative language a C1 to C20 alkyl group, as used herein, refers to an R group that can be selected independently from a hydrocarbyl group having 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 carbon atoms, as well as any range between these two numbers for example a C3 to C8 alkyl group, and also including any combination of ranges between these two numbers for example a C3 to C5 and C7 to C10 hydrocarbyl group. In another example, by the disclosure that the molar ratio typically spans the range from about 0.1 to about 1.1, Applicants intend to recite that the molar ratio can be selected from about 0.1:1, about 0.2:1, about 0.3:1, about 0.4:1, about 0.5:1, about 0.6:1, about 0.7:1, about 0.8:1, about 0.9:1, about 1.0:1, or about 1.1:1.


Applicants reserve the right to proviso out or exclude any individual members of any such group, including any sub-ranges or combinations of sub-ranges within the group, that may be claimed according to a range or in any similar manner, if for any reason Applicants choose to claim less than the full measure of the disclosure, for example, to account for a reference that Applicants may be unaware of at the time of the filing of the application. Further, Applicants reserve the right to proviso out or exclude any individual substituents, compounds, ligands, structures, or groups thereof, or any members of a claimed group, if for any reason Applicants choose to claim less than the full measure of the disclosure, for example, to account for a reference that Applicants may be unaware of at the time of the filing of the application.


The following references disclose certain pyridine compounds.

TABLE 9References disclosing pyridine compounds.Publication orPatent No.TitlePatent AssigneeUS2004/0198728Pyridines and uses thereofHong (inventor)WO 2005007648Preparation of biaryl piperazinyl-pyridineNeurogenanalogues as capsaicin receptor modulatorsCorporation, USAWO 2005033105Preparation of pyridinyl and analogousAmgen Inc., USAvanilloid receptor ligands and their use intreating painWO 2005030714Process for the production of compoundsEisai Co., Ltd.,having 5- to 10-membered aromaticJapanheterocycles with alkylmagnesiummonoamidesWO 2005007646Preparation of arylaminotriazines andNeurogenarylaminopyrimidines as capsaicin receptorCorporation, USAmodulators for the treatment of pain and otherdiseasesGB 2404855Preparation of arylcarboxylates asPantherix Ltd, UKantibacterialsWO 2004094361Cobalt carbonyl and aminopyridineDaiso Co. Ltd.,derivatives catalyzed process for producingJapanβ -hydroxyesterWO 2004089286Preparation of substituted pyrimidinaminesIRM LLC, Bermudaand triazinamines as protein kinase inhibitorsWO 2004085423Preparation of piperidine derivatives for theAstrazeneca AB,treatment of chemokine or H1 mediatedSwed.disease stateWO 2004062665Preparation of heteroaryl-substitutedSB Pharmco Puertopyrrolo[2,3-b]pyridine derivatives as CRFRico Inc., USA;receptor antagonistsNeurocrineBiosciences Inc.;Glaxo GroupLimitedWO 2004050643A preparation of heterocyclic non-nucleosideBoehringerreverse transcriptase inhibitors, useful for theIngelheimtreatment of HIV-1InternationalG.m.b.H., GermanyWO 2004039795Preparation of aryl and heteroaryl amides, inFujisawaparticular benzamides and pyridinyl amides,Pharmaceutical Co.,as apolipoprotein B (Apo B) secretionLtd., Japan; DaisoinhibitorsCo., Ltd.; et al.US 2004082780Preparation of (aryloxy)pyrimidine andAmgen Inc., USA(aryloxy)pyridazine as vanilloid receptorligandsWO 2004014366Preparation of tetrazole derivs. as matrixWarner-Lambertmetalloproteinase inhibitorsCompany Llc, USAUS 2004204584Preparation of 2-acylaminobenzothiazoleHoffmann-La Rochederivatives as adenosine receptor ligandsInc., Switz.JP 2004359642Maleimides as monomers for heat-resistantNippon Shokubaipolymers, and their manufactureCo., Ltd., JapanJP 2004047229Photoelectric conversion devices usingNational Institute ofelectrolyte solutions containingAdvanced Industrialaminopyridines, and dye-sensitized solar cellsScience andusing themTechnology, JapanJP 2004018448Preparation of anhydrous salts of 2′-Mitsui ChemicalsA2 20040122deoxyguanosine derivativesInc., JapanJP 2002-17477020020614WO 2004089910Preparation of arylpyrazoles as serotonin 5-Merck PatentHT2A and 5-HT2C receptor antagonistsGmbH, GermanyWO 2004000820Certain aromatic monocycles, particularlyCellular Genomics,trisubstituted [1,3,5]triazine derivatives, asInc., USAkinase modulators, and their pharmaceuticalcompositions and methods of useWO 2003101989Preparation of pyrazolo[4,3-c]pyridinylVertexsubstituted pyrimidinamines as inhibitors ofPharmaceuticalsJAK and CDK2 protein kinasesIncorporated, USAWO 2003101959Preparation of pyrroles for the treatment ofGlaxo Groupprostaglandin mediated diseasesLimited, UKWO 2003099771Preparation of diarylurea derivatives usefulNovartis A.-G.,for the treatment of protein kinase dependentSwitz.; NovartisdiseasesPharma G.m.b.H.U.S. Pat. No. 6864261Preparation of 1-(pyrid-2-yl)piperazines asEuro-Celtique, S.A.,metabotropic glutamate receptor inhibitorLuxembourgWO 2003091226Preparation of triazole derivatives asEli Lilly andtachykinin receptor antagonistsCompany, USAWO 2003087067Preparation of aryl-alkyne compounds asSyngentaherbicidesParticipations A.-G.,Switz.WO 2003080060Substituted piperazine antithrombotic PAI-1Schering(plasminogen activator inhibitor-1) inhibitors,Aktiengesellschaft,and their preparation, pharmaceuticalGermanycompositions, and use in the treatment ofthrombotic diseases.WO 2003062392Methods using Edg receptor modulators forCeretek LLC, USAthe treatment of Edg receptor-associatedconditionsWO 2003051366Preparation of pyridine derivatives as proteinAbbott Laboratories,kinase inhibitorsUSAWO 2003050087Preparation of pyridylalkynes as herbicidesSyngentaParticipations Ag,Switz.WO 2003048137Preparation of 2-phenylbenzoxazoles asMerck & Co., Inc.,metabotropic glutamate receptor-5 modulatorsUSAfor treatment of pain and CNS disordersWO 2003045941Preparation of pyridine and pyrimidineCelltech R & Dderivatives as p38α kinase inhibitorsLimited, UKWO 2003029226Heterocyclyl-substituted phenoxyalkyl-,Basfphenylthioalkyl-, phenylaminoalkyl- andAktiengesellschaft,phenylalkyl-sulfamoylcarboxamides asGermanyherbicidesWO 2003026664Preparation of 2-phenylamino-4-(5-Bayer Corporation,pyrazolylamino)pyrimidines as kinaseUSAinhibitors, in particular, SRC kinase inhibitorsWO 2003022285Preparation of substituted 2-(4-Euro-Celtique S.A.,phenoxyphenyl)pyridine derivatives andLuxembourgrelated compounds as sodium channelblockers for the treatment of neuronal damageand neurodegenerative conditionsUS 2004248739Preparation of pyridylpropynyloxyphenylSyngentaderivatives as herbicidesParticipations AG,Switz.US 2003139435Preparation of pyridine and pyrimidine N-Bristol-Myersheterocyclic p38 kinase inhibitors for treatingSquibb Company,TNF-α mediated disordersUSA;Pharmacopeia, Inc.US 2003205696Carbazole-based materials for guest-hostCanon Kabushikielectroluminescent systemsKaisha, JapanU.S. Pat. No. 6831175Preparation of heteroaryls for therapeutic useAbbott Laboratories,in pharmaceutical compositions as kinaseUSAinhibitors for treatment of hyperproliferativediseases, including cancerUS 2003187026Preparation of pyridine derivatives as proteinUSAkinase inhibitorsU.S. Pat. No. 6559167Preparation of benzimidazole derivatives asRegents of theprodrugs of proton pump inhibitorsUniversity ofCalifornia, USA;The United StatesDepartment ofVeteran Affairs;WinstonPharmaceuticals,LLCJP 2003344970Composition of bleachable dye and radicalKonica Minoltagenerator, silver halide photographic materialHoldings Inc., Japancontaining it, and dye bleachingJP 2003335754Dibenzopyrrolidine derivatives for organicFuji Photo Film Co.,electroluminescent devicesLtd., JapanJP 2003335753Dibenzopyrrolidine derivatives for organicFuji Photo Film Co.,electroluminescent devicesLtd., JapanJP 2003221518Coloration compositions with good hue andFuji Photo Film Co.,light and ozone resistanceLtd., JapanJP 2003002834Pharmaceutical compositions containingTanabe Seiyaku Co.,heterocyclic compounds as α 1β 2Ltd., Japanintegrin-mediated adhesion inhibitors fortreatment of inflammatory diseasesCN 1405156Preparation of N-hydroxyamidinesWuhan University,Peop. Rep. ChinaCN 1405154Preparation of N-hydroxylamidine derivativesWuhan University,Peop. Rep. ChinaUS 2004/0198728Pyridines and Uses ThereofUS 2004122219Pyrazole azo dyes, their production andFuji Photo Film Co.,coupling agents thereforLtd., JapanUS 2004110757Preparation of aromatic carboxylic acids asChugai SeiyakuFlt-1 ligands.Kabushiki Kaisha,JapanUS 2003176416Aryl- and heteroaryl-substitutedNeurosearch A/S,diazabicycloalkanes as cholinergic ligands forDen.the nicotinic acetylcholine receptorU.S. Pat. No. 6906067Preparation of s-triazines and pyrimidines forBristol-Myerspharmaceutical use as cytokine, especiallySquibb Company,TNF-α inhibitorsUSA;Pharmaceopeia, Inc.U.S. Pat. No. 6693295Novel indole derivative, material for light-Fuji Photo Film Co.,emitting device and light-emitting deviceLtd., Japanusing the sameJP 2002216969White- or blue-emitting organicToyota Centralelectroluminescent (EL) elements withResearch andexcellent emission efficiency and color purityDevelopmentLaboratories, Inc.,JapanJP 2002037777Aliphatic group-substituted aminopyridiniumFuji Photo Film Co.,derivative for controlling of liquid crystal tiltLtd., JapanangleU.S. Pat. No. 6673818Preparation of fluoro-substitutedPharmaciabenzenesulfonyl pyrazoles and isoxazoles forCorporation, USAthe treatment of cyclooxygenase-2 mediateddisorders such as inflammationEP 1202608Organic light-emitting devicesKabushiki KaishaToyota ChuoKenkyusho, JapanUS 2002028329Light emitting element and azole compoundFuji Photo Film Co.,Ltd., JapanU.S. Pat. No. 6767671Nonaqueous electrolytic solution forMitsubishisecondary batteryChemicalCorporation, JapanUS 2004082586Preparation of 3,4-dihydro-2H-pyrroles asBayer A.-G.,pesticidesGermanyUS 2004019190Thrombopoietin mimeticsSmithKlineBeechamCorporation, USA;Glaxo GroupLimitedU.S. Pat. No. 6849618Preparation of uracil substituted N-sulfamoylBasfbenzamides as herbicidesAktiengesellschaft,GermanyUS 2004092552Preparation of 2-fluorobenzenesulfonyl-Pharmaciaheterocycles with COX-1 and COX-2Corporation, USAinhibiting activity for pharmaceutical use inthe treatment of inflammationUS 2001027196Preparation of pyrimidinamines andF. Hoffmann Lapyridinamines as adenosine receptorRoche A.-G., Switz.modulators for treatment of CNS disordersU.S. Pat. No. 6897225Preparation of 3-(hetero)aryl-1,3-Tanabe Seiyaku Co.,diazabicyclo[3.3.0]octane-2,4-diones andLtd., Japananalogs as inhibitors of α 1β 2mediated cell adhesionWO 2001027119Preparation of imidazopyridineamines andGruenenthalanalogs as analgesicsG.m.b.H., GermanyUS 2004220235Method of inhibiting amyloid proteinWarner-Lambertaggregation, treating Alzheimer's disease, andCompany, USA;imaging amyloid deposits usingYamanouchi[[(phenylalkyl)phenyl]amino]be nzoic acidsPharmaceuticaland analogsCompany, Ltd.; etal.WO 2000043385Preparation of benzimidazolyltriazineZenyaku Kogyoderivatives as antitumor agentsKabushiki Kaisha,JapanWO 2000021954Preparation of 1-benzyl-3-(pyrimidin-2-Bayeryl)indazoles and related compounds asAktiengesellschaft,stimulators of soluble guanylate cyclase.GermanyU.S. Pat. No. 6093734Preparation of benzimidazole derivatives asUSAprodrugs of proton pump inhibitorsJP 2000119256Pyrazolylacrylonitriles and their use asNissan ChemicalagrochemicalsIndustries, Ltd.,JapanU.S. Pat. No. 6248892Procedure for the production of arylpyridinesClariant G.m.b.H.,GermanyWO 9957103Preparation of condensed imidazole derivativeNippon Chemipharas therapeutic agents for liver diseaseCo., Ltd., Japan;ZeriaPharmaceutical Co.,Ltd.WO 9920606Preparation of piperidine and piperazineJ. Uriach & Cia.glycoprotein IIb/IIIa antagonistsS.A., SpainU.S. Pat. No. 6251900Preparation of heterocyclic compounds asZenyaku Kogyoantitumor agentsKabushiki Kaisha,JapanU.S. Pat. No. 59771381,4-Disubstituted piperidine ether muscarinicScheringantagonistsCorporation, USAU.S. Pat. No. 5952349Preparation of arylaminopiperidines asScheringmuscarinic M2 antagonists for treatingCorporation, USAmemory lossWO 9844925Calcilytic compoundsSmithklineBeechamCorporation, USAU.S. Pat. No. 6841674Preparation of N-heterocyclic derivatives asBerlex Laboratories,NOS inhibitorsInc., USA;Pharmacopeia, Inc.;et al.U.S. Pat. No. 6043242Preparation of imidazopyridazines for controlByk Guldenof Helicobacter bacteriaLombergChemische FabrikG.m.b.H., GermanyWO 9825912Preparation of herbicidal 1H-tetrazole-1-E. I. Du Pont decarboxamidesNemours & Co.,USA; Rorer, MorrisPadgettU.S. Pat. No. 6020349Preparation of thiourea derivatives and relatedNovo Nordisk A/S,compounds as constrained somatostatinDen.agonists and antagonistsU.S. Pat. No. 6040302Preparation of 1,4-disubstituted piperazinesNovo Nordisk A/S,for the treatment of painful, hyperalgesicDen.and/or inflammatory conditionsU.S. Pat. No. 6008234Benzamidine derivatives substituted by cyclicSchering A.-G.,amino acid or cyclic hydroxy acid derivatives,Germanyand their use as anticoagulantsWO 9806697Preparation of phenyl piperidin-4-yl ethers asScheringmuscarinic antagonistsCorporation, USAWO 9801425Preparation of 1,4-disubstituted piperidines asSchering Corp.,muscarinic antagonistsUSAJP 10324671Amino alcohol esters as ceramide analogs andSeikagaku Kogyopharmaceuticals containing them for treatmentCo., Ltd., Japanof nerve diseasesJP 10310583Pyridoneazo compound and thermal-transferMitsubishiprinting material using itChemical IndustriesLtd., JapanJP 10260512Processing of photographic material usingFuji Photo Film Co.,developer containing silver stain inhibitorLtd., JapanJP 10207019Processing of silver halide photographicFuji Photo Film Co.,material using developer containingLtd., Japanheterocyclic compoundU.S. Pat. No. 6048675Processing of silver halide photographicFuji Photo Film Co.,materials with mercapto compoundsLtd., JapanJP 10153838Processing of silver halide photographicFuji Photo Film Co.,materials containing mercapto compoundsLtd., JapanU.S. Pat. No. 6573278Preparation of arysulfonamides and relatedBayer A.-G.,compounds as cannabinoid CB1 and CB2Germanyreceptor agonists.WO 9826127Multicomponent system for altering,Consortium fuerdegrading, or bleaching lignin, lignin-Elektrochemischecontaining materials, or similar substances,Industrie G.m.b.H.,and method for its useGermanyWO 9816224Pyrazolinones for the management of potencyMerck PatentdisordersG.m.b.H., GermanyU.S. Pat. No. 6156903Preparation of novel pyridonecarboxylic acidWakunagaderivatives as antibacterial agentsPharmaceutical Co.,Ltd., Japan; Yazaki,Akira; Niino,Yoshiko; Ohshita,Yoshihiro; Hirao,Yuzo; Amano,Hirotaka; Hayashi,Norihiro; Kuramoto,YasuhiroU.S. Pat. No. 5696282Process and catalysts for producingPhillips Petroleumorganosulfur compounds by the additionCo., USAreaction of hydrogen sulfide or mercaptanswith α, β-unsaturated carbonyl ornitrile compoundsU.S. Pat. No. 6306884Preparation of benzamidine derivatives asBerlex Laboratories,anticoagulantsInc., USAJP 09194582Manufacture of polycarbonates using nitrogenIdemitsucompound catalystsPetrochemical Co.,Ltd., JapanU.S. Pat. No. 6335444Preparation of diastereomeric 2-acylamino-3-Seikagaku Kogyomorpholino-1-phenyl-1-propanols andKabushiki Kaishaanalogs(SeikagakuCorporation), JapanWO 9715555Preparation of 1-phenylimidazoles as nitricSchering A.-G.,oxide synthase inhibitorsGermanyU.S. Pat. No. 5990123Preparation of vasoconstrictive 2,3-dihydro-Janssen1,4-dioxinopyridinesPharmaceutica N.V.,Belg.U.S. Pat. No. 5559135Preparation of endothelin antagonists bearingMerck and Co., Inc.,pyridylamide groupsUSAU.S. Pat. No. 5929248Substituted aryl- andUpjohn Co., USAheteroarylphenyloxazolidinonesU.S. Pat. No. 5559108Cephalosporin derivativesBristol-MyersSquibb Company,USAU.S. Pat. No. 5514505Method for obtaining improved imageXerox Corp., USAcontrast in migration imaging membersJP 08175993Preparation and formulation of pyridineGreen Cross Corp,derivatives as antioxidantsJapanU.S. Pat. No. 5629134Chemical amplification positive-workingShinetsu Chemicalresist materialIndustry Co., Ltd.,Japan; NipponTelegraph &TelephoneGB 2297747Hydrazide for photographic materialIlford A.G., Switz.U.S. Pat. No. 5559108Cephalosporin derivativesBristol-MyersCompany, USAU.S. Pat. No. 5672708Preparation od N-arylaminoacrylic acids andBayer A.-G.,their use as intermediates for the preparationGermanyof 4-quinolone-3-carboxylic acid derivativesWO 9602532Preparation of 7-piperazinyl-1,4-dihydro-4-Bayer A.-G.,oxo-1-[4-(1H-1,2,4-triazol-1-yl-Germanymethyl)phenyl]quinoline-3-carboxylic acids asvirucidesU.S. Pat. No. 5763438Preparation of 2-acylamino-3-piperidino-1-Seikagaku Corp.,propanol derivatives and analogs for treatmentJapanof viral and nervous diseasesU.S. Pat. No. 6100268Preparation of vasoconstrictiveJanssendihydrobenzopyranpyrimidine derivativesPharmaceutica N.V.,Belg.U.S. Pat. No. 5801179Preparation of vasoconstrictive substitutedJanssenaryloxyalkyl diaminoheterocyclylsPharmaceutica N.V.,Belg.U.S. Pat. No. 5451486Photographic contrast promoting agentsSun ChemicalCorp., USAU.S. Pat. No. 5231094Pyrazolopyrimidine derivatives which areLaboratoires UPSA,angiotensin II receptor antagonistsFr.U.S. Pat. No. 5387747Triazolopyrimidine derivatives which areLaboratoires Upsa,angiotensin II receptor antagonists, theirFr.methods of preparation and pharmaceuticalcompositions in which they are presentJP 07179771Preparation of trimethine dyes useful asFuji Photo Film Cointermediates for recording materialsLtd, JapanJP 07099996Substrate composition for the determination ofFujirebio Kk, JapanperoxidaseU.S. Pat. No. 5332582Cyclodextrin- and polymer-based drugInsite Vision Inc,delivery systemUSAU.S. Pat. No. 5336677Preparation of 4-American Home(biphenylylamino)pyrimidines and analogs asProducts Corp.,angiotensin II antagonistsUSAJP 06240163Manufacture of intermediates for trimethineFuji Photo Film Cocolor formersLtd, JapanU.S. Pat. No. 5374514Photothermographic materials.Minnesota Miningand ManufacturingCo., USAU.S. Pat. No. 5316890Silver halide photographic materialFuji Photo Film Co.,Ltd., JapanU.S. Pat. No. 5510345Preparation of 1-(eburnaminine-14-carbonyl)-Richeter Gedeon4-pyrimidinylpiperazines and analogs as lipidVegyeszeti Gyarperoxidation inhibitorsRT., Hung.U.S. Pat. No. 5550240Preparation ofRichter, Gedeon,piperazinylbis(alkylamino)pyrimidineVegyeszeti Gyarderivatives as intermediates for lipidRt., Hung.peroxidation inhibitorsWO 9322311Fungicidal 1,3,4-oxadiazines and 1,3,4-du Pont dethiadiazinesNemours, E. I., andCo., USAWO 9311134Preparation of (3H,7H)thiazolo(3,4-Boehringera)pyridines having antiasthmatic andMannheim Italiaantiinflammatory activities on the respiratoryS.p.A., ItalytractU.S. Pat. No. 5336677Substituted aminopyrimidine angiotensin IIAmerican Homereceptor antagonistsProducts Corp.,USAJP 05132462Pyridinium salts and cationic polymerizationNippon Soda Co,initiatorsJapanJP 05132461Pyridinium salts and cationic polymerizationNippon Soda Co,initiatorsJapanU.S. Pat. No. 5149699Condensed pyrimidine derivatives and theirAmerican Homeuse as angiotensine II antagonistsProducts Corp.,USAEP 525768Heterocyclyl group-substituted tetralonesUriach, J., e Cia.having antihypertensive and bronochodilatingS.A., SpainactivityWO 9217448Preparation of 3-methyleneisoindolin-1-oneKyowa Hakkoderivatives for treating ischemic cerebralKogyo Co., Ltd.,disordersJapanU.S. Pat. No. 6559186Adrenergic agonists and antagonists forUSAtreatment of sympathetically maintained painWO 9300342Preparation of 1-thiazolylaminocarbonyl-4-Boehringerarylpiperazines and analogs as bronchodilatorsMannheim ItaliaS.p.A., ItalyEP 517542Organic electroluminescent devicesSumitomo ChemicalCo., Ltd., JapanWO 9219615Preparation of pyrazoles, pyrazolines anddu Pont detetrahydropyridazines as agrochemicalNemours, E. I., andfungicidesCo., USAU.S. Pat. No. 5942384Silver halide photographic material and itsFuji Photo Film Co.,processingLtd., JapanEP 493670Preparation of pyridinium betaines asBayer A.-G.,enhancers of plant resistance against infectionGermanyby microorganismsU.S. Pat. No. 5256408Aminosteroids for ophthalmic useInsite Vision, Inc.,USAWO 9106542Preparation of pharmaceutically active amino-Upjohn Co., USAsubstituted heteroaryl aminesU.S. Pat. No. 5023258Preparation of 1-(piperazinylphenoxy)-2-Pfizer Inc., USAtriazolylethanols as fungicidesU.S. Pat. No. 5527914Methine compounds as photographicFuji Photo Film Co.,sensitizers and silver halide emulsionsLtd., Japancontaining themU.S. Pat. No. 5202224pyridine derivative photographic cyanFuji Photo Film Co.,couplersLtd., JapanU.S. Pat. No. 5155166Use of 1-(1-pyrrolidinylcarbonyl)pyridiniumEastman Kodak Co.,salts to attach compounds to carboxylatedUSAparticles and a kit containing sameU.S. Pat. No. 5654298Preparation of 4-aminopyridinium salts asImperial ChemicalbradycardiacsIndustries PLC, UKU.S. Pat. No. 5179123Preparation of alkynoylbenzoic,G.D. Searle and Co.,-thiophenecarboxylic, -furancarboxylic, andUSA-pyridinecarboxylic acids as leukotriene B4(LTB4) synthesis inhibitorsEP 451585Preparation of 2-aryl-6-heteroarylpyridines asBayer A.-G.,herbicidesGermanyJP02235054Silver halide color photographic material withFuji Photo Film Co.,good image sharpner and safety light stabilityLtd., JapanEP 376870Preparation of new organosilyl polyphosphateCentro Marga paralareagents for cyclization ofInvestigacion S.A.,aminomethylenemalonates in the preparationSpainof quinolone and azaquinolone antibacterialsEP 352946Triazolyl(hydroxypropoxy)phenylpiperazinesPfizer Ltd., UK;as medical and agrochemical fungicidesPfizer Inc.U.S. Pat. No. 4980350Preparation of piperazinylpyrimidines andMerck and Co., Inc.,analogs as hypoglycemic agentsUSAWO 2005028467Preparation of 3,5-diaminopiperidine-Anadyssubstituted hetero/aromatic compounds asPharmaceuticals,antibacterial agentsInc., USAUS 2004110031Organic electroluminescent device and displayJapanU.S. Pat. No. 5681954Preparation of piperazines having calmodulinDaiichiinhibitory activityPharmaceutical Co.,Ltd., JapanJP 08225535Preparation of 3-(piperazinoalkyl)indoleDaiichi Seiyaku Co,derivatives as calmodulin antagonistsJapanEP 624584Preparation of piperazine derivatives asDaiichicalmodulin inhibitors.Pharmaceutical Co.Ltd., JapanEP 263213Amino steroids useful for treating a variety ofUpjohn Co., USAconditions, and a process for their preparationHeterocycles (1996),Ring transformation of fused pyridazines. IV.Iwamoto, Ken-ichi;43(1), 199-204Reaction of halo-substituted fused pyridazinesSuzuki, Sumiko;CODEN: HTCYAM;with ynaminesOishi, Etuso;ISSN: 0385-5414Miyashita, Akira;Higashino, Takeo


Applicants reserve the right to proviso out, or to restrict from any claim currently presented, or from any claim that may be presented in this or any further application based upon this disclosure, including claims drawn any genus or subgenus disclosed herein, any compound or group of compounds disclosed in any reference provided herein.


Although methods, syntheses, and materials similar or equivalent to those described herein can be used in the practice or testing of this invention, typical methods, syntheses, and materials are described herein.


Acronyms and Abbreviations


The following abbreviations and acronyms are commonly used throughout this disclosure, including the Examples: DMF, dimethylformamide; BINAP, 2R,3S,2,2′-bis-(diphenylphosphino)-1,1′-binapthyl; DMSO, dimethylsulphoxide; NaH, sodium hydride; CH2Cl2 or DCM, dichloromethane; CDCl3, deuterated chloroform or chloroform-d; POCl3, phosphorus oxychloride; THF, tetrahydrofuran; AlCl3, aluminum chloride; NaOH, sodium hydroxide; Na2CO3, sodium carbonate; MeOH, methanol; NH4OH, ammonium hydroxide; K2CO3, potassium carbonate; TFA, trifluoracetic acid; SiO2, silicon dioxide or silica; KH2PO4, potassium dihydrogen phosphate; n-BuLi, n-butyllithium; (PPh3)4Pd, tetrakis(triphenylphosphine)palladium(0); (PPh3)2PdCl2, bis(triphenylphosphine)palladium(II) chloride; HPLC, high performance liquid chromatography; TLC, thin layer chromatography; mL, milliliters; M.P., melting point; RT, room temperature, typically ranging from about 20° C. to about 40° C.; aq, aqueous; min, minutes; h or hr, hours; g, grams; atm, atmosphere; conc., concentrated; MS or Mass Spec, mass spectroscopy/spectrometry; NMR, nuclear magnetic resonance; TMS, tetramethylsilane; Rf, TLC retention factor; Rt, HPLC retention time; HPFC, high performance flash chromatography; IR, infrared spectroscopy/spectrum; CH3CN, acetonitrile; N2, nitrogen; mg, milligrams; mmol, millimoles; mol, moles; nm, nanometers; HRMS, high resolution mass spectroscopy; and ° C., degrees Centigrade.


Abbreviations especially frequent in the NMR data are as follows: MHz, megahertz; Hz, hertz; br, broad; apt, apparent; s, singlet; d, doublet; t, triplet; q, quartet; dq, doublet of quartets; dd, doublet of doublets; dt, doublet of triplets; and m, multiplet.


The precursor compounds such as halogenated pyridines, phenylboronic acid, and substituted phenylboronic acids were obtained from a variety of commercial sources, including, for example, Sigma-Aldrich Inc., Asymchem Laboratories, and Lancaster Synthesis, Inc.


The following experiments and Examples are merely illustrative, and compounds of the present invention are not limited by the following particular species. The skilled artisan will appreciate how the experiments and Examples may be further implemented as disclosed by variously altering the following examples, substituents, conditions, or reagents. In the following examples, in the disclosure of any measurements, including temperatures, pressures, times, weights, percents, concentrations, ranges, chemical shifts, frequencies, molar ratio, etc., it is to be understood that such measurements are respectively, “about.”


EXAMPLES
Example 1
Synthesis of 2,6-dichloro-4-iodopyridine (B21)



embedded image


4-Amino-2,6-dichloropyridine (11.58 g, 71 mmol) was stirred in 100 mL of concentrated HCl at room temperature for 24 hour. The mixture was transferred to a 2 L Erlenmeyer flask and was cooled in an ice bath. Sodium nitrite (9.85 g, 142 mmol) in water (15 mL) was added dropwise. Potassium iodide (29.85 g, 178 mmol) in water (30 mL) was added slowly, and the reaction was allowed to stir for 5 minutes. (Water was used to rinse down the sides of the flask.) Tetrahydrofuran (60 mL) was added and the solution was neutralized by addition of solid sodium bicarbonate. The reaction mixture was extracted three times with diethyl ether. The combined organic fractions were washed with 10% sodium thiosulfate solution until it turned light orange in color. The organic layer was dried over magnesium sulfate and concentrated on the rotary evaporator, which yielded a light orange solid product (15.4 g, 80%). This material was typically used without further purification. A recrystallization may be performed as needed using 3:1 (hexanes:THF). The solid that was formed was filtered and quickly rinsed with cold acetone.


M.P.: 143-145° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 11.8 min, 95.6% purity.



1H NMR (600 MHz, CDCl3, TMS): δ 7.64 (s, 2H).


Reference: adapted from Mello, J. V.; Finney, N. S. Org. Lett; 2001, 26, 4263-4265.


Example 2
Synthesis of 2,6-di-chloro pyridine-1-oxide



embedded image


A mixture of 2,6-dichloropyridine (2 g, 13.6 mmol) in TFA (6 mL) and 30% H2O2 (2.5 mL) was prepared at 0° C., after which the mixture was heated to reflux at 90-100° C. After 7 hour at this temperature, the mixture was cooled to room temperature and neutralized with IN NaOH solution to pH 8-9. The aqueous layer was extracted with petroleum ether (200 mL×3) and the organic layer was dried over sodium sulfate, filtered, and concentrated to dryness under vacuum. The residue was purified by column chromatography over 100-200 mesh silica gel, eluting with 20:80 acetone: hexanes, to afford the pure compound as a colorless solid (1.75 g, yield 80%).


M.P.: 137-138° C.



1H NMR (200 MHz, CDCl3) δ 7.35-7.49 (d, 2H, J=8.25 Hz), 7.08-7.18 (dd, 1H, J=8.56 Hz).


Mass Spec: (CI-MS) m/z: 164 (M++1, 100%).


Reference: Stan V. D Andrea et.al Tetrahedron 2000, 56, 5687-5698.


Example 3
Synthesis of 2,4,6-trichloropyridine



embedded image


A mixture of 2,6-dichloropyridine-1-oxide (1.60 g, 9.81 mmol), obtained in step (i), distilled POCl3 (10.0 mL), and dry LiCl was heated to reflux with stirring under a nitrogen atmosphere for 6 hour. After this time, the reaction mixture was cooled to 0° C., after which crushed ice was added slowly with stirring. Petroleum ether was added (50 mL) and 1N NaOH solution was added dropwise to the resulting reaction mixture until the pH was basic. The organic layer was separated, the aqueous layer was extracted with petroleum ether, and the combined organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by column chromatography over 100-200 mesh silica gel by elution with 10:90 ethyl acetate:hexanes, to provide the product as pure brown oil (yield 51%).



1H NMR (200 MHz, CDCl3) δ 7.29 (s, 2H).


Mass Spec: (CI-MS) m/z: 184 (M++2, 5%).


Reference: Stan V. D Andrea et.al Tetrahedron 2000, 56, 5687-5698.


Example 4
Synthesis of (2,6-dichloro-pyridin-4-yl)-p-tolyl-amine



embedded image


To 2,6-dichloro-4-iodopyridine (2.737 g, 10 mmol) dissolved in toluene (35 mL) was added p-toluidine (1.346 g, 12 mmol), tris(dibenzylideneacetone)dipalladium (180.3 mg, 0.2 mmol), 1,3-bis(diphenylphosphino)propane (162.3 mg, 0.4 mmol), and sodium-tert-butoxide (1.347 g, 14 mmol). The resulting mixture was allowed to stir at reflux for 12 hour. The sample was diluted in dichloromethane and filtered through Celite™. The Celite™ was washed with CH2Cl2; the filtrate was washed two times with water and one time with brine. The organic phase was dried over potassium carbonate and concentrated by rotary evaporation. The resulting sample was dried for 12 to 14 hours under vacuum. Purification (Biotage Horizon HPFC chromatography system, SiO2, 80:20 hexanes:ethyl acetate) gave a light brown solid (149 mg, 6%).


M.P.: 94° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 12.1 min, 95.1% purity.



1H NMR (300 MHz, CDCl3, TMS) δ 2.37 (s, 3H), 6.15 (s, 1H), 6.63 (s, 2H), 7.07 (apt d, J=8.4 Hz, 2H), 7.21 (apt d, J=8.1 Hz, 2H).


Mass Spec: LC-MSD (ES+): m/z 253 (M+H, 70.89).


Example 5
Synthesis of 1-[4-(2,6-dichloro-pyridin-4-ylamino)-phenyl]-ethanone



embedded image


To 2,6-dichloro-4-iodopyridine (5.42 g, 20 mmol) dissolved in toluene (30 mL) and THF (10 mL) was added 4-amino-acetophenone (3.2412 g, 24 mmol), tris(dibenzylideneacetone)dipalladium (367.1 mg, 0.4 mmol), 1,3-bis(diphenyl-phosphino)propane (331.2 mg, 0.8 mmol), and sodium-tert-butoxide (2.69 g, 28 mmol). The resulting mixture was allowed to stir at reflux for 12-18 hours. The sample was diluted in dichloromethane and filtered through Celite™. The Celite™ was washed with dichloromethane; the filtrate was washed two times with water and one time with brine. The organic phase was dried over potassium carbonate and concentrated by rotary evaporation. The resulting sample was dried for 12-18 hours under vacuum. Recrystallization in dichloromethane gave a yellow solid (2.1 g, 37%).


M.P.: 226° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 6.5 min, 88.4% purity.


Example 6
Synthesis of 1-[4-(4-hydroxy-6′-phenyl-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4′-ylamino)-phenyl]-ethanone



embedded image


To 2,6-dichloro-4-iodopyridine (5.43 g, 20 mmol) and 3-aminoacetophenone (0.3.25 g, 24 mmol) dissolved in dry toluene (10 mL) and THF (2 mL) and tris(dibenzylideneacetone)dipalladium (0) (0.3627 g, 0.0.4 mmol), 1,3-bis(diphenylphospino)propane (0.3257 g, 0.8 mmol), sodium-tert-butoxide (2.68 g, 28 mmol) was added. The reaction mixture was stirred and refluxed for 12-18 hours under N2. The reaction mixture was diluted with CH2Cl2 and filtered through Celite™, then rinsed with CH2Cl2. The filtrate was washed two times with water and one time with brine. The organic phase was collected and dried over potassium carbonate. The filtered sample was concentrated, and the resulting solid was dried for 12-18 hours under vacuum. Biotage Horizon HPFC system chromatography (SiO2, 96:3:1 CH2Cl2:MeOH:NH4OH) yielded a solid (1.2 g, 21%).


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4(0.01M, pH 3.2):CH3CN], 264 nm, Rt 6.6 min, 98.3% purity.



1H NMR: (300 MHz, CDCl3, TMS): δ 2.63 (s, 3H), 6.47 (br s, 1H), 6.72 (s, 2H), 7.26-7.45 (m, 1H), 7.50 (t, J=7.8 Hz, 1H), 7.75-7.81 (m, 2H).


Example 7
Synthesis of benzo[1,3]dioxol-5-yl-(2,6-dichloro-pyridin-4-yl)-amine (B24)



embedded image


To 2,6-dichloro-4-iodopyridine (274.3 mg, 1 mmol) dissolved in toluene (15 mL) were added 3,4-(methylenedioxy)aniline (168.7 mg, 1.2 mmol), tris(dibenzylidene-acetone)-dipalladium (18.0 mg, 0.02 mmol), 1,3-bis(diphenylphosphino)propane (16.8 mg, 0.04 mmol), and sodium-tert-butoxide (134.0 mg, 1.4 mmol). The resulting mixture is allowed to stir at reflux for 12-18 hours. The sample was diluted in dichloromethane and filtered through Celite™. The Celite™ was washed with dichloromethane; the filtrate was washed two times with water and one time with brine. The organic phase was dried over sodium sulfate and concentrated by rotary evaporation. The resulting sample was dried for 12-18 hours under vacuum. Purification (Biotage Horizon HPFC chromatography system, SiO2, 80:20 hexanes:ethyl acetate) gave a light brown solid product (190 mg, 67%).


M.P.: 129° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 8.1 min, 99.4% purity.



1H NMR (600 MHz, CDCl3, TMS): δ 6.011 (s, 2H), 6.09 (br s, 1H), 6.54 (s, 2H), 6.64 (dd, J=2.4, 8.1 Hz, 1H), 6.68 (d, J=1.8 Hz, 1H), 6.81. d, J=7.8 Hz, 1H).


Mass Spec: (TOF MS ES+): m/z 283 (M+H, 100).


Example 8
Synthesis of (2,6-dichloro-pyridin-4-yl)-(3-fluoro-phenyl)-amine (B25)



embedded image


This compound was prepared by an analogous procedure to that disclosed in Example 7.


M.P.: 163° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 9.8 min, 94.5% purity.



1H NMR (600 MHz, CDCl3, TMS): δ 6.271 (s, 1H), 6.73 (s, 2H), 6.88-6.92 (m, 2H), 6.95-6.96 (m, 1H), 7.35 (q, J=7.8 Hz, 1H).


Mass Spec: (TOF MS ES+): m/z 257 (M+H, 100).


Example 9
Synthesis of (2,6-dichloro-pyridin-4-yl)-(3-methylsulfanyl-phenyl)-amine



embedded image


To 2,6-dichloro-4-iodopyridine (1.3715 g, 5 mmol) dissolved in toluene (30 mL) were added 3-methylthioaniline (0.73 mL, 6 mmol), tris(dibenzylidene-acetone)dipalladium (90.6 mg, 0.1 mmol), 1,3-bis(diphenylphosphino)propane (80.0 mg, 0.2 mmol), and sodium-tert-butoxide (672.1 mg, 7 mmol). The resulting mixture was allowed to stir at reflux for 12-18 hours. The sample was diluted in dichloromethane and filtered through Celite™. The Celite™ was washed with dichloromethane and the filtrate was washed two times with water and one time with brine. The organic phase was dried over potassium carbonate and concentrated by rotary evaporation. The resulting sample (9) was dried for 12-18 hours under vacuum. The sample was used without purification.


Example 10
Synthesis of thiocarbonic acid S-[3-(2,6-dichloro-pyridin-4-ylamino)-phenyl] ester-O-methyl ester



embedded image


Unpurified (2,6-dichloro-pyridin-4-yl)-(3-methylsulfanyl-phenyl)-amine (1.8 g, 6.3 mmol) was dissolved in methanol (127 mL) and the resulting solution was cooled to 0° C. with an ice bath. A solution of oxone (17.52 g, 28.4 mmol) in water (285 mL) was added. The resulting mixture was allowed to warm to room temperature and was allowed to stir at room temperature for about 1.5 hours. The reaction mixture was diluted with water and extracted three times with dichloromethane. The organic phase was dried over sodium sulfate and concentrated by rotary evaporation. The resulting sample was dried for 12-18 hours under vacuum. Purification (Biotage Horizon HPFC chromatography system, SiO2, 50:50 hexanes:ethyl acetate) gave a white solid product (905 mg, 45%).


M.P.: 162° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 um, Rt 5.0 min, 93.8% purity.



1H NMR (600 MHz, CDCl3, TMS): δ 3.09 (s, 3H), 6.58 (s, 1H), 6.76 (s, 2H), 7.52-7.52 (m, 1H), 7.61 (apt t, J=7.9, 1H), 7.96 (apt t, J=1.6, 1H), 7.72-7.72 (m, 1H).


Mass Spec: (TOF MS ES+): m/z 317 (M+H, 100).


Example 11
Synthesis of 4-(2,6-dichloro-pyridin-4-yl)-morpholine



embedded image


A mixture of morpholine (0.604 g, 6.04 mmol) in dry DMSO and NaH (0.251 g, 6.6 mmol) was stirred for 30 minutes at 0° C., after which time 2,4,6-trichloropyridine (1.0 g, 5.50 mmol) was added. This reaction mixture was stirred at room temperature for 4 hours, after which time ice cold water was added to quench the reaction (100 mL). The reaction mixture was then extracted with ethyl acetate, and the extract was washed with 3-4 times water. The organic layer was dried over sodium sulphate and concentrated under vacuum. The residue was purified by column chromatography over 100-200 mesh silica gel. Elution of the column with 20% acetone in hexanes gave the pure product as a colorless solid (0.565 g, yield 44%).



1H NMR (200 MHz, CDCl3): δ 6.58 (s, 2H), 3.83 (t, 4H, J=4.83 Hz), 3.28-3.35 (t, 4H, J=5.37 Hz).


Mass Spec: (CI-MS) m/z: 233 (M+, 100%).


Example 12
Synthesis of 2,6-dichloro-4-pyrrolidino pyridine



embedded image


A mixture of pyrrolidine (0.910 g, 10.7 mmol) in dry DMSO and NaH (0.385 g, 16.0 mmol) was stirred for 30 minutes at 0° C., after which a sample of 2,4,6-trichloropyridine (1.75 g, 9.63 mmol) was added. This reaction mixture was stirred at room temperature for 4 hours, after which time ice cold water was added to quench the reaction. The reaction mixture was then extracted with ethyl acetate, and the extract was washed 3-4 times with water. The organic layer was dried over sodium sulphate and concentrated. The residue was purified by column chromatography over 100-200 mesh silica gel. Elution of the column with 20% acetone in hexanes gave the pure product as a colorless solid (yield: 75%).



1H NMR (200 MHz, CDCl3): δ 6.30 (s, 2H), 3.25-3.32 (t, 4H, J=13.34 Hz), 1.86-2.15 (m, 4H).


Mass Spec: (ES-MS) m/z: 216 (M+, 100%).


Example 13
Synthesis of 2,6-dichloro-4-piperdino pyridine



embedded image


A mixture of piperidine (0.910 g, 10.7 mmol) in 25 mL of THF and 5 mL of 1N NaOH was stirred for 30 minutes at 0° C., after which time 2,4,6-trichloropyridine (1.63 g, 8.86 mmol) was added. The reaction mixture was then stirred at 30-40° C. for 12-18 hours, after which the reaction mixture was cooled and neutralized with 5% HCl (aq). This product was extracted with ethyl acetate, and the organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by column chromatography over 100-200 mesh silica gel by elution with 10:90 ethyl acetate: hexanes to afford the product as a pure colorless solid (yield 32%).



1H NMR (200 MHz, CDCl3): δ 6.56 (s, 2H), 3.34-3.37 (t, 4H, J=5.60 Hz), 1.54-1.75 (m, 6H).


Mass Spec: (CI-MS) m/z: 231 (M++1, 100%).


Reference: Stan V. D Andrea et al Tetrahedron 2000, 56, 5687-5698.


Example 14
Synthesis of 2′,6′-dichloro-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-4-ol



embedded image


A solution of piperdine-4-ol (1.2 g, 12.0 mmol) in isopropylalcohol (30 mL) was prepared and 1N NaOH (8 mL) was added with stirring. After stirring 15 min at room temperature, 2,4,6-trichloropyridine (2.0 g, 10.0 mmol) was added, and the resulting reaction mixture was heated at 60° C. for 10 hours. After this time, water was added to the reaction mixture (100 mL), and the product was extracted with ethyl acetate (200 mL×3). The organic extract was washed with brine solution followed by water, after which the organic layer was dried over using sodium sulphate and concentrated under vacuum. The resulting product was purified by column chromatography using 230-400 mesh silica gel, eluted with 30:70 ethyl acetate:petroleum ether, to afford the desired compound as light yellow solid (1 g, yield 37%).



1H NMR (200 MHz, CDCl3): δ 6.58 (s, 1H), 4.05 (m, 1H), 3.7 (m, 2H), 3.19 (m, 2H), 2.0 (s, 1H), 1.8 (m, 2H), 1.6 (m, 2H).


Mass Spec: (ES-MS) m/z: 247 (M++1, 100%).


Example 15
Synthesis of (3-chloro-4-methoxy-phenyl)-(2,6-dichloro-pyridin-4-yl)-amine



embedded image


To tris(dibenzylideneacetone)dipalladium (0.1822 g, 0.2 mmol) and sodium tert-butoxide (1.5515 g, 15.9 mmol) in toluene (40 mL, anhydrous) under a nitrogen atmosphere was added a mixture of 2,6-dichloro-4-iodopyridine (2.4993, 9.13 mmol), 3-chloro-4-methoxy-phenylamine (1.8423 g, 10.5 mmol) and 1,3-bis(diphenyl-phospino)propane (0.1897, 0.45 mmol) in toluene (40 mL, anhydrous)-THF (2 mL, dry) by cannular transfer. The reaction mixture was heated at gentle reflux for approximately 3 h under nitrogen. The mixture was cooled and diluted with ethyl acetate followed by a brine wash. The organic layer was separated, dried over anhydrous potassium carbonate, filtered and then concentrated under vacuum. Purification (Biotage Horizon HPFC chromatography system, SiO2, 70:30 hexanes:ethyl acetate) gave a solid (1.817 g, 65.6%).


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 10.7 min, 98.4% purity.



1H NMR (600 MHz, CDCl3, TMS): δ 3.93 (s, 3H), 6.21 (s, 1H), 6.56 (s, 2H), 6.95 (d, J=9 Hz, 1H), 7.10 (dd, J=9, 3 Hz, 1H), 6.95 (d, J=3.6 Hz, 1H).


Mass Spec (TOF MS ES+): m/z 303 (M+H, 100).


Example 16
Synthesis of (2,6-dichloro-pyridin-4-yl)-(4-trifluoromethoxy-phenyl)-amine



embedded image


To 2,6-dichloro-4-iodopyridine (1.37 g, 5 mmol) dissolved in toluene (25 mL) was added 4-trifluoromethoxyaniline (0.80 mL, 6 mmol), tris(dibenzylidene-acetone)dipalladium (90.1 mg, 0.1 mmol), 1,3-bis(diphenylphosphino)propane (80.8 mg, 0.2 mmol), and sodium-tert-butoxide (672.7 mg, 7 mmol). The resulting mixture is allowed to stir at reflux overnight. The sample was diluted in dichloromethane and filtered through celite. The celite is washed with dichloromethane; the filtrate was washed two times with water and one time with brine. The organic phase was dried over sodium sulfate and concentrated by rotary evaporation. The resulting sample was dried overnight under vacuum. Purification (Biotage Horizon HPFC chromatography system, SiO2, 70:30 hexanes:ethyl acetate) gave a light purple solid (1.186 g, 73%).


M.P.: 124° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 14.5 min, 96.0% purity.



1H NMR (600 MHz, CDCl3, TMS): δ 6.31 (s, 1H), 6.67 (s, 1H), 7.20-7.22 (m, 2H), 7.25-7.26 (m, 2H).


Mass Spec (TOF MS ES+): m/z 323 (M+H, 100).


Example 17
Synthesis of 1-[4-(2,6-dichloro-pyridin-4-ylamino)-phenyl]-ethanone



embedded image


To 2,6-dichloro-4-iodopyridine (0.8177 g, 3 mmol) and 4-aminoacetophenone (0.4891 g, 3.6 mmol) dissolved in dry toluene (10 mL) and THF (2 mL) was added tris(dibenzylideneacetone)dipalladium(0) (0.0571 g, 0.06 mmol), 1,3-bis(diphenyl-phospino)propane (0.0499 g, 0.12 mmol), and sodium-tert-butoxide (0.4042 g, 4.2 mmol). The reaction mixture was stirred and refluxed for 12 to 14 hours under N2. The reaction mixture was diluted with CH2Cl2 and filtered through Celite™, then rinsed with CH2Cl2. The filtrate was washed two times with water and one time with brine. The organic phase was collected and dried over potassium carbonate. The filtered sample was concentrated, and the resulting solid was dried for 12-18 hours under vacuum.


Biotage Horizon HPFC system chromatography (SiO2, 96:3:1 CH2Cl2:MeOH:NH4OH) yielded a light yellow (250 mg, 30%).


M.P.: 210° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 6.3 min, 84.7% purity.



1H NMR (300 MHz, DMSO, TMS): δ 2.50 (s, 3H, overlaps with DMSO peak), 6.96 (s, 2H), 7.28 (d, J=8.7 Hz, 2H), 7.93 (d, J=8.7 Hz, 2H), 9.73 (s, 1H).


Mass Spec: : (TOF MS ES+): m/z 281 (M+, 100).


Example 18
Synthesis of 1-(2,6-dichloro-pyridin-4-yl)-4-methyl-piperazine (I)



embedded image


A mixture of N-methyl piperazine (0.604 g, 6.04 mmol) in dry DMSO (6 mL) was prepared and NaH (0.2637 g, 10.98 mmol) was added. The resulting mixture was stirred for 30 minutes at 0° C., after which time 2,4,6-trichloropyridine (1.0 g) was added. The reaction mixture was stirred at 20 to 40° C. for 4 hours. In the reaction mixture ice cold water was added and extracted with ethyl acetate giving 3-4 times water washing. The organic layer was dried and concentrated. The residue was purified by column chromatography using 100-200 mesh silica gel. Elution of the column with 20% acetone in hexanes gave the pure product as a colorless solid. Yield: 26%.



1H NMR (200 MHz, CDCl3): δ 6.62(s, 2H, J=13.67 Hz), 3.35-3.45 (t, 4H, J=10.34 Hz), 2.56-2.62 (t, 4H, J=10.00 Hz), 2.40 (s, 3H).


Mass Spec: (ES-MS) m/z: 216 (M+, 100%).


Example 19
Synthesis of (3-chloro-4-methoxy-phenyl)-(2-chloro-6-phenyl-pyridin-4-yl)-amine (B23)



embedded image


To (3-chloro-4-methoxy-phenyl)-(2,6-dichloro-pyridin-4-yl)-amine (0.459 g, 1.5 mmol) and phenylboronic acid (0.1861 g, 1.5 mmol) dissolved in acetonitrile (20 mL) and Na2CO3 (20 mL, 0.4 M) was added palladium(0) tetrakis triphenylphosphine (0.0891 g, 0.075 mmol). The reaction mixture was stirred and refluxed for 12-18 hours under N2. The reaction mixture was diluted with CH2Cl2 and filtered through Celite™, then rinsed with CH2Cl2. The filtrate was washed one time with water and one time with brine. The organic phase was collected and dried over potassium carbonate. The filtered sample was concentrated, and the resulting solid was dried for 12-18 hours under vacuum. Biotage Horizon HPFC system chromatography (SiO2, 100% CH2Cl2) yielded an off white solid product (182 mg, 52%).


M.P.: 128° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4(0.01M, pH 3.2):CH3CN], 264 nm, Rt 18.7 min, 96% purity.



1H NMR: (600 MHz, CDCl3, TMS, 55° C.): δ 3.92 (s, 3H), 6.00 (s, 1H), 6.60 (s, 1H), 6.94-6.95 (m, 2H), 7.11 (dd, J=2.4, 8.4 Hz, 1H), 7.25 (s, 1H), 7.36-7.42 (m, 3H), 7.86 (d, J=8.4 Hz, 2H).


Mass Spec: (TOF MS ES+): m/z 345 (M+H, 100).


Example 20
Synthesis of 2′-chloro-6′-phenyl-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-4-ol



embedded image


To a solution of 2′,6′-dichloro-3,4,5,6-tetrahydro-3H-[1,4′]bipyridinyl-4-ol (100 mg, 0.4 mmol) in 1,4-dioxane (8 mL) were added 1N sodium carbonate solution (172 mg, 1.6 mmol) and tetrakis(triphenylphosphine)palladium(0) (46 mg, 0.04 mmol), followed by phenyl boronic acid (74 mg, 0.6 mmol). The resulting reaction mixture was refluxed for 4 hours, after which time water was added 100 mL to the reaction mixture, and the product was extracted with ethyl acetate. The organic layer was dried over sodium sulphate, filtered, and the filtrate was concentrated under vacuum. The resulting product was purified by column chromatography using 230-400 mesh silica gel (5% methanol in dichloromethane) to afford the desired compound as a yellow solid (50 mg, yield 45%.).



1H NMR (400 MHz, CDCl3): δ 7.91 (m, 2H), 7.45-7.38 (m, 3H), 6.99 (s, 1H), 6.64 (s, 1H), 4.01-3.95 (m, 1H), 3.78-3.699 (m, 2H), 3.23-3.168 (m, 2H), 2.02-1.95 (m, 2H), 1.67-1.59 (m, 2H).


Mass Spec: (CI-MS) (m/z): 289 (M+b +1, 100%).


IR (neat) cm−1: 3329, 2926, 2854, 1599, 1526, 1434, 1367, 1224, 1145, 1074, 1042, 982, 810.


Example 21
Synthesis of 3-[6-chloro-4-(4-trifluoromethoxy-phenylamino)-pyridin-2-yl]-N-ethyl-benzamide



embedded image


To (2,6-dichloro-pyridin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (0.1002 g, 0.3 mmol) and 3-(N-ethylaminocarbonyl)phenyl boronic acid (0.1162 g, 0.6 mmol) dissolved in acetonitrile (10 mL) and Na2CO3 (10 mL, 0.4M) was added palladium (0) tetrakis(triphenylphosphine) (0.0179 g, 0.015 mmol). The reaction mixture was stirred and refluxed for 12-18 hours under N2. The reaction mixture was diluted with CH2Cl2 and filtered through Celite™, then rinsed with CH2Cl2. The filtrate was washed two times with water and one time with brine. The organic phase was collected and dried over potassium carbonate. The filtered sample was concentrated, and the resulting solid was dried for 12-18 hours under vacuum. Biotage Horizon HPFC system chromatography (SiO2, 50:50 hexanes:ethyl acetate) yielded a (36.5 mg, 32%).


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4(0.01M, pH 3.2):CH3CN], 264 nm, Rt 11.6 min, 97.8% purity.



1H NMR: (300 MHz, DMSO-d6, TMS): δ 1.13 (t, J=6.9 Hz, 3H), 3.26-3.33 (m, 2H), 6.90 (s, 1H), 7.38-7.41 (br m, 4H), 7.56 (apt t, J=16.8 Hz, 1H), 7.90 (d, J=7.8 Hz, 1H), 8.05 (d, J=7.8 Hz, 1H), 8.35 (s, 1H), 8.62 (apt t, J=4.8 Hz, 1H), 9.38 (s, 1H).


Example 22
Synthesis of 1-[4-(2-chloro-6-morpholin-4-yl-pyridin-4-ylamino)-phenyl]-ethanone



embedded image


A portion of 1-[4-(2,6-Dichloro-pyridin-4-ylamino)-phenyl]-ethanone (600.1 mg, 2.13 mmol) was dissolved in morpholine (10 mL, 115 mmol). The resulting mixture was allowed to stir at reflux for 12-18 hours. The sample was allowed to cool to room temperature and was diluted in dichloromethane. The mixture was washed two times with water and one time with brine. The organic phase was dried over sodium sulfate and concentrated by rotary evaporation. The resulting sample was dried for 12-18 hours under vacuum. Purification (Biotage Horizon HPFC chromatography system, SiO2, 98:2 dichloromethane:methanol) gave a brown solid (22, 554 mg, 78%).


M.P.: 174° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 5.9 min, 64.3% purity. (Used as is with no further purification.)



1H NMR (300 MHz, CDCl3, TMS): δ 2.57 (s, 3H), 3.41-3.47 (m, 4H), 3.76-3.781 (m, 4H), 6.12 (d, J=1.5 Hz, 1H), 6.34 (s, 1H), 6.413 (d, J=1.5 Hz, 1H), 7.13-7.26 (m, 2H), 7.90 7.97 (m, 2H).


Mass Spec: LC-MSD (ES+): m/z 332 (M+H, 40.1).


Example 23
Synthesis of 6′-chloro-4′-(4-trifluoromethoxy-phenylamino)-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4-ol



embedded image


Portions of (2,6-Dichloro-pyridin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (0.1987 g, 0.6 mmol) and 4-hydroxypiperidine (0.1258 g, 1.2 mmol) were dissolved in DMF (5 mL) followed by the addition of potassium carbonate (0.0998 g, 0.72 mmol). The reaction mixture was stirred and refluxed for 12-18 hours at 100° C. under N2. The reaction was monitored by TLC, and after 18 hours, potassium carbonate (0.0491 g, 0.36 mmol) was added stirred and refluxed for another 12-18 hours under N2. The reaction mixture was diluted with CH2Cl2. The filtrate was washed two times with water and one time with brine. The organic phase was collected and dried over potassium carbonate. The filtered sample was concentrated, and the resulting solid was dried for 12-18 hours under vacuum. Biotage Horizon HPFC system chromatography (SiO2, 98:2 CH2Cl2:MeOH) yielded a solid (83 mg, 23%).


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4(0.01M, pH 3.2):CH3CN], 264 nm, Rt 8.6 min, 98.9% purity.



1H NMR: (300 MHz, CHCl3, TMS): δ 1.50-1.61 (m, 3H) 1.92-1.97 (m, 2H), 3.07-3.15 (m, 2H), 3.92-3.99 (m, 3H), 5.91 (s, 1H), 6.21 (d, J=1.5 Hz, 1H), 7.14-7.22 (m, 4H), 7.15 (s, 2H), 7.23 (br s, 4H), 7.26 (s, 1H), 7.40-7.49 (m, 4H), 8.02-8.04 (br m, 4H).


Example 24
Synthesis of 1-[4-(6′-chloro-4-hydroxy-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4′-ylamino)-phenyl]-ethanone



embedded image


Portions of 1-[4-(2,6-Dichloro-pyridin-4-ylamino)-phenyl]-ethanone (0.99 g, 3.5 mmol) and 4-hydroxypiperidine (0.7079 g, 7 mmol) were dissolved in DMF (10 mL) followed by the addition of potassium carbonate (0.5810 g, 4.2 mmol). The reaction mixture was stirred and refluxed for 12-18 hours at 100° C. under N2. The reaction mixture was diluted with CH2Cl2. The filtrate was washed two times with water and one time with brine. The organic phase was collected and dried over potassium carbonate. The filtered sample was concentrated, and the resulting solid was dried for 12-18 hours under vacuum.


Biotage Horizon HPFC system chromatography (SiO2, 98:2 CH2Cl2:MeOH) yielded a light yellow solid (552 mg, 49%).


M.P.: 80° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4(0.01M, pH 3.2):CH3CN], 264 nm, Rt 4.2 min, 97.1% purity.



1H NMR: (300 MHz, CHCl3, TMS): δ 1.51-1.62 (m, 3H), 1.91-1.99 (m, 2H), 2.58 (s, 3H), 3.10-3.19 (m, 3H), 3.89-4.01 (m, 3H), 6.15 (d, J=1.8 Hz, 1H), 6.22 (s, 1H), 6.35 (d, J=1.2 Hz, 1H), 7.14-7.17 (m, 2H), 7.93-7.96 (m, 2H).


Mass Spec: LC-MSD (ES+): m/z 346 (M+H, 100).


Example 25
Synthesis of 1-[3-(6′-chloro-4-hydroxy-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4′-ylamino)-phenyl]-ethanone



embedded image


Samples of 1-[4-(4-hydroxy-6′-phenyl-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4′-ylamino)-phenyl]-ethanone (0.99 g, 3.5 mmol) and 4-hydroxypiperidine (0.7079 g, 7 mmol) were dissolved in DMF (10 mL) followed by the addition of potassium carbonate (0.5835 g, 4.2 mmol). The reaction mixture was stirred and refluxed for 12-18 hours at 100° C. under N2. The reaction mixture was diluted with CH2Cl2. The filtrate was washed two times with water and one time with brine. The organic phase was collected and dried over potassium carbonate. The filtered sample was concentrated, and the resulting solid was dried for 12-18 hours under vacuum.


Biotage Horizon HPFC system chromatography (SiO2, 96:3:1 CH2Cl2:MeOH:NH4OH) yielded a light brown solid (450 mg, 37%).


M.P.: 70° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4(0.01M, pH 3.2):CH3CN], 264 nm, Rt 4.5 min, 99.7% purity.



1H NMR: (300 MHz, CHCl3, TMS): δ 1.46-1.61 (m, 3H), 1.90-1.97 (m, 2H), 2.60 (s, 3H), 3.07-3.16 (m, 2H), 3.88-3.99 (m, 3H), 5.99 (s, 1H), 6.03 (d, J=1.5 Hz, 1H), 6.22 (d, J=1.5 Hz, 1H), 7.35-7.48 (m, 2H), 7.65-7.72 (m, 2H).


Mass Spec: LC-MSD (ES+): m/z 346 (M+H, 100).


Example 26
Synthesis of (2-chloro-6-morpholin-4-yl-pyridin-4-yl)-(4-trifluoromethoxy-phenyl)-amine



embedded image


A portion of (2,6-dichloro-pyridin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (0.0513 g, 0.15 mmol) was dissolved in morpholine (0.5 mL, 38 mmol). The reaction mixture was stirred and refluxed for 12-18 hours at 120° C. under N2. The reaction mixture was diluted with ethyl acetate and washed with water two times and one time with brine. The organic phase was collected and dried over sodium sulfate. The filtered sample was concentrated, and the resulting solid was dried for 12-18 hours under vacuum. Flash chromatography (SiO2, 70:30 hexanes:ethyl acetate) yielded a solid (36 mg, 61%).


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 13.8 min, 98.5% purity.



1H NMR: (300 MHz, CHCl3, TMS): δ 3.44 (t, J=4.8 Hz, 4H), 3.77 (t, J=5.1 Hz, 4H), 5.94 (apt t, J=1.5 Hz, 2H), 6.26 (d, J=1.2 Hz, 1H), 7.19 (apt t, J=2.4 Hz, 4H).


Example 27
Synthesis of (6-chloro-4-pyrrolidin-1-yl-pyridin-2-yl)-(4-trifluoromethoxy-phenyl)-amine



embedded image


To a solution of 2,6-dichloro-4-pyrrolidino-pyridine (1.0 g, 4.62 mmol) in toluene (6 mL) were added potassium tert-butoxide (0.1556 g, 1.38 mmol), palladium(II)acetate (26 mg, 0.115 mmol), BINAP (36 mg, 0.057 mmol) and 4-trifluoromethoxyphenylamine (246 mg, 1.38 mmol), in a 10 mL reaction vessel. This reaction mixture was subjected the microwave radiation, in which the microwave power was 250 W, to attain a reaction temperature of 150° C. After this reaction proceeded for 30 min, water (200 mL) was added to the reaction mixture and the reaction product was extracted with ethyl acetate. The organic layer was dried over sodium sulphate, filtered, and the filtrate concentrated under vacuum. The product was purified through column chromatography using 230-400 mesh silica gel to afford the desired compound as light brown solid (220 mg, yield 14%).



1H NMR (200 MHz, DMSO-d6): δ 7.12-7.33 (m, 4H), 6.45 (s, 1H), 6.04 (s, 1H, J=1.66 Hz), 5.75 (s, 1H, J=1.66 Hz), 3.22-3.30 (m, 4H, J=13.34 Hz), 1.95-2.05 (m, 4H).


Mass Spec: (ES-MS) m/z: 358 (M++1, 100%).


Example 28
Synthesis of (6′-chloro-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-2′-yl]-(4-trifluoro-methoxyphenyl)-amine



embedded image


To a solution of 2′,6′-dichloro-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl (1 g, 4.3 mmol) in toluene (6 mL) were added potassium tert-butoxide (973 mg, 8.6 mmol), palladium(II) acetate (48 mg, 0.21 mmol), BINAP (135 mg, 0.21 mmol) and 4-trifluoro-methoxyphenylamine (769 mg, 4.3 mmol) in a 10 mL reaction vessel. This reaction mixture was subjected the microwave irradiation, in which the microwave power was 250 W, at a temperature of 150° C. After this reaction proceeded for 30 min, water was added to the reaction mixture and the reaction product was extracted with ethyl acetate. The organic layer was dried over sodium sulphate, filtered, and the filtrate concentrated under vacuum. The resulting product was purified through column chromatography using 230-400 mesh silica gel (18% ethylacetate in petroleum ether) to afford the desired compound as light yellow solid (820 mg, yield 51%).



1H NMR (400 MHz, CDCl3): δ 7.29-7.17(m, 4H), 6.61 (s, 1H), 6.27 (s, 1H), 6.04 (s, 1H), 3.28-3.26 (m, 4H), 1.66-1.59 (m, 6H).


Mass Spec: (CI-MS) m/z: 372 (MW++1, 100%).


Example 29
Synthesis of 4-(6′-chloro-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-2′-ylamino)-N-methyl-benzenesulfonamide



embedded image


To a solution of 2′,6′-dichloro-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl (500 mg, 2.15 mmol) in toluene (6 mL) were added potassium tert-butoxide (486 mg, 4.3 mmol), palladium(II) acetate (24 mg, 0.1 mmol), BINAP (67 mg, 0.1 mmol), and 4-amino-N-methylbenzenesulfonamide (444 mg, 2.15 mmol) were taken in a 10 mL reaction vessel. This reaction mixture was subjected the microwave irradiation, in which the microwave power was 250 W, to attain a reaction temperature of 1 50° C. After this reaction proceeded for 30 min, water (200 mL) was added to the reaction mixture and the reaction product was extracted with ethyl acetate. The organic layer was dried over sodium sulphate, filtered, and the filtrate concentrated under vacuum. The product was purified through column chromatography using 230-400 mesh silica gel to afford the desired compound as light yellow solid (290 mg, yield, 35%).


Mass Spec: (ES-MS) m/z: 381 (M++1, 100%).


Example 30
Synthesis of 4-(6-chloro-4-pyrrolidin-1-yl-pyridin-2-ylamino)-N-methyl-benzenesulfonamide



embedded image


To a solution of 2,6-dichloro-4-pyrrolidino-pyridine (0.350 g, 1.62 mmol) in toluene (6 mL) were added potassium tert-butoxide (0.132 g, 1.18 mmol), palladium(II)-acetate (36.40 mg, 0.160 mmol), BINAP (32 mg, 0.514 mmol) and 4-amino-N-methyl-benzenesulfonamide (227 mg, 1.22 mmol) in a 10 mL reaction vessel. This reaction mixture was subjected the microwave irradiation, in which the microwave power was 250 W, to attain a reaction temperature of 150° C. After this reaction proceeded for 30 min, water (150 mL) was added to the reaction mixture and the reaction product was extracted with ethyl acetate. The organic layer was dried over sodium sulphate, filtered, and the filtrate concentrated under vacuum. The product was purified through column chromatography using 230-400 mesh silica gel (elution 20:80 acetone:petroleum ether) to afford the desired compound as a light brown solid (110 mg, yield 19%).



1H NMR (200 MHz, DMSO-d6): δ 9.31 (s, 1H), 7.74 (d, 2H, J=9.00 Hz), 7.64 (d, 2H), 7.46 (d, 2H, J=8.33 Hz), 7.18 (br s, 1H), 6.63 (s, 1H), 5.96 (s, 1H), 3.22-3.38 (m, 4H), 2.31-2.40 (d, 3H, J=5.00),1.99-2.06 (m, 4H).


Mass Spec: (ES-MS) m/z: 367 ((M++1, 50%).


Example 31
Synthesis of (6-chloro-4-morpholin-4-yl-pyridin-2-yl)-(4-trifluoromethoxy-phenyl)-amine



embedded image


To a solution of 4-(2,6-dichloro-pyridin-4-yl)-morpholine (0.500 g, 2.15 mmol) in toluene (6 mL) were added potassium tert-butoxide (0.109 g, 0.973 mmol), palladium(II)acetate (24.13 mg, 0.107 mmol), BINAP (67.0 mg, 0.107 mmol) and 4-trifluoromethoxyphenylamine (456 mg, 2.58 mmol), in a 10 mL reaction vessel. This reaction mixture was subjected the microwave radiation, in which the microwave power was 250 W, to attain a reaction temperature of 150° C. After this reaction proceeded for 30 min, water (200 mL) was added to the reaction mixture and the reaction product was extracted with ethyl acetate. The organic layer was dried over sodium sulphate, filtered, and the filtrate concentrated under vacuum. The product was purified through column chromatography using 230-400 mesh silica gel (elution 30:70 ethyl acetate:petroleum ether) to afford the desired compound as a light yellow solid (140 mg, yield 18%).



1H NMR (200 MHz, DMSO-d6): δ7.15-7.32 (m, 4H), 6.45 (s, 2H), 6.28 (s, 1H), 3.77-3.82 (m, 4H), 3.19-3.25 (m, 4H).


Mass Spec: (ES-MS) m/z: 374 (M++1, 100%).


Example 32
Synthesis of (2-chloro-6-pyrrolidin-1-yl-pyridin-4-yl)-(4-trifluoromethoxy-phenyl)-amine



embedded image


To (2,6-dichloro-pyridin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (800.5 mg, 2.5 mmol) dissolved in pyrrolidine (16 mL, 192 mmol) was added potassium carbonate (1.041 g, 7.5 mmol). The resulting mixture is allowed to stir at 86° C. for 12 to 14 hours. The sample was allowed to cool to 20 to 40° C. and was diluted in dichloromethane. The mixture was washed two times with water and one time with brine. The organic phase was dried over potassium carbonate and concentrated by rotary evaporation. The resulting sample was dried for 12-18 hours under vacuum. Purification (Biotage Horizon KPFC chromatography system, SiO2, 70:30 hexanes:ethyl acetate) gave a light brown solid (665 mg, 74%).


M.P.: 87° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 22.7 min, 98.4% purity.



1H NMR (300 MHz, CDCl3, TMS): δ 1.93-1.98 (m, 4H), 3.40 (t, J=6.6 Hz, 4H), 5.70, (d, J=1.8 Hz, 1H), 6.08 (s, 1H), 6.18 (d, J=1.5 Hz, 1H), 7.18 (s, 4H).


Mass Spec (TOF MS ES+): m/z 358 (M+H, 100).


Example 33
Synthesis of [2-chloro-6-(4-methyl-piperazin-1-yl)-pyridin-4-yl]-(4-trifluoromethoxy-phenyl)-amine



embedded image


To (2,6-dichloro-pyridin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (291.1 mg, 0.9 mmol) dissolved in dimethylformamide (5 mL) was added N-methylpiperazine (0.1 mL, 0.9 mmol) and potassium carbonate (152.1 mg, 1.1 mmol). After the resulting mixture was allowed to stir at 90° C. for 12 to 14 hours, a significant amount of starting material remained. Therefore, water (4-5 drops) was added, the temperature was raised to 110° C., and the reaction mixture was allowed to stir at 110° C. for 12 to 18 hours. The sample was diluted in dichloromethane, washed two times with water and one time with brine and filtered through Celite™. The organic phase was dried over potassium carbonate and concentrated by rotary evaporation. The resulting sample was dried for 12-18 hours under vacuum. Purification (Biotage Horizon HPFC chromatography system, SiO2, 93:6:1 dichloromethane:methanol:ammonium hydroxide) gave the product as a solid (121 mg, 35%).


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 2.9 min, 96.1% purity.



1H NMR (600 MHz, CDCl3, TMS): δ 2.33 (s, 3H), 2.47-2.48 (m, 4H), 3.48-3.49 (m, 4H), 5.96 (d, J=1.8 Hz, 1H), 5.98 (s, 1H), 6.23 (d, J=1.2 Hz, 1H), 7.16-7.27 (m, 4H).


Example 34
Synthesis of [6-chloro-4-(4-methyl-piperazin-1-yl)-pyridin-2-yl]-(4-trifluoromethoxy-phenyl)-amine



embedded image


In a 10 mL reaction vessel, a solution of 1-(2,6-dichloro-pyridin-4-yl)-4-methyl-piperazine (0.250 g, 1.02 mmol) in toluene (6 mL) was prepared, and potassium tertiary butoxide (0.136 g, 1.22 mmol), palladium(II)acetate (22.80 mg, 0.101 mmol), BINAP (31.6 mg, 0.0508 mmol) and 4-trifluoromethoxy-phenylamine (216 mg, 1.22 mmol) were added to the solution. This reaction mixture was subjected the microwave irradiation, in which the microwave power was 250 W, to attain a temperature of 150° C. After this reaction proceeded for 30 min, water was added to the reaction mixture and the product extracted with ethyl acetate. The organic layer was dried over sodium sulphate, filtered and concentrated under vacuum and purified through column chromatography using 100-200 mesh silica gel (elution-20% acetone:petroleum ether) to afford the desired compound as light brown solid (98 mg, 25%).



1H NMR (400 MHz, DMSO-d6): δ 7.25-7.32 (d, 4H), 6.05-6.38 (s, 2H), 4.95(s, 1H), 3.25-3.32 (m, 4H), 2.02-2.35 (s, 3H).


Mass Spec: (ES-MS) m/z: 387 ((M++1,100%).


Example 35
Synthesis of (2,6-diphenyl-pyridin-4-yl)-p-tolyl-amine (B26)



embedded image


To a mixture of (2,6-dichloro-pyridin-4-yl)-p-tolyl-amine (100.4 mg, 0.4 mmol) dissolved in acetonitrile (6 mL) and 2 M Na2CO3 (6 mL) was added phenylboronic acid (122.5 mg, 1 mmol) and palladium tetrakis(triphenylphosphine) (46.3 mg, 0.04 mmol). The resulting mixture was allowed to stir at reflux for 12 hours. The sample was diluted in dichloromethane and filtered through Celite™. The Celite™ was washed with dichloromethane, and the filtrate was washed two times with water and one time with brine. The organic phase was dried over potassium carbonate and concentrated by rotary evaporation. The resulting sample was dried for 12-18 hours under vacuum. Purification (Biotage Horizon HPFC chromatography system, SiO2, 70:30 hexanes:ethyl acetate) gave a brown solid (2,6-diphenyl-pyridin-4-yl)-p-tolyl-amine (116 mg, 86%).


M.P.: 125° C.


HPLC: Inertsil ODS-3V C18, 40:10:50 [KH2PO4 (0.01M, pH 3.2):CH3CN:MeOH], 264 nm, Rt 8.5 min, 91.4% purity.



1H NMR (300 MHz, CDCl3, TMS): δ 2.37 (s, 2H), 6.08 (s, 1H), 7.15-7.25 (m, 5H), 7.39-7.48 (m, 5H), 8.04-8.07 (m, 4H).


Mass Spec: (TOF MS ES+): m/z 337 (M+H, 100).


Example 36
Synthesis of (2,6-diphenyl-pyridin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (B29)



embedded image


To (2,6-dichloro-pyridin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (0.201 g, 0.6 mmol) and phenyl boronic acid (0.1476 g, 1.2 mmol) dissolved in acetonitrile (10 mL) and Na2CO3 (10 mL, 0.4M) was added palladium (0) tetrakis(triphenylphosphine) (0.0351 g, 0.03 mmol). The reaction mixture was stirred and refluxed for 12-18 hours under N2. The reaction mixture was diluted with CH2Cl2 and filtered through Celite™, then rinsed with CH2Cl2. The filtrate was washed one time with water and one time with brine. The organic phase was collected and dried over potassium carbonate. The filtered sample was concentrated, and the resulting solid was dried for 12-18 hours under vacuum. Biotage Horizon HPFC system chromatography (SiO2, 80:20 hexanes:ethyl acetate) yielded a pale brown solid (99 mg, 39%).


M.P.: 137° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4(0.01M, pH 3.2):CH3CN], 264 nm, Rt 13.4 min, 93.5% purity.



1H NMR: (300 MHz, CDCl3, TMS): δ 6.33 (s, 1H), 7.26-7.30 (m, 6H), 7.39-7.50 (m, 6H), 8.05 (dd, J=1.2, 7.9 Hz, 4H).


Mass Spec: (TOF MS ES+): m/z 407 (M+H, 100).


Example 37
Synthesis of [2,6-bis-(3-fluoro-phenyl)-pyridin-4-yl]-(4-trifluoromethoxy-phenyl)-amine (B28)



embedded image


To (2,6-dichloro-pyridin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (3.007 g, 9.3 mmol) dissolved in acetonitrile (40 mL) and 2 M Na2CO3 (40 mL, 80 mmol) was added 4-fluorophenyl boronic acid (3.256 g, 23.25 mmol) and palladium (0) tetrakis-(triphenylphosphine) (1.073 g, 0.93 mmol). The resulting mixture is allowed to stir at reflux for 12-18 hours. The sample was diluted in dichloromethane and filtered through Celite™. The Celite™ was washed with dichloromethane; the filtrate was washed two times with water and one time with brine. The organic phase was dried over potassium carbonate and concentrated by rotary evaporation. The resulting sample was dried for 12-18 hours under vacuum. Column chromatography (SiO2, 70:30 hexanes:ethyl acetate) gave a white solid (3.291 g, 80%).


M.P.: 159° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 32.1 min, 98.3% purity.



1H NMR (300 MHz, CDCl3, TMS): δ 6.15 (s, 1H), 7.11-7.25 (m, 6H), 8.00-8.04 (m, 4H).


Mass Spec: (TOF MS ES+): m/z 443 (M+H, 100).


Example 38
Synthesis of [2,6-bis-(4-methanesulfonyl-phenyl)-pyridin-4-yl]-(4-trifluoromethoxy-phenyl)-amine (B30)



embedded image


This compound was prepared by the process disclosed in Example 37.


M.P.: >270° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 8.6 min, 96.1% purity.



1H NMR: (300 MHz, DMSO-d6, TMS): δ 3.27 (s, 3H), 3.25 (s, 3H), 7.46 (quartet, J=9 Hz, 4H), 8.07 (d, J=8.4 Hz, 4H), 8.34 (d, J=8.1 Hz, 4H), 9.34 (s, 1H).


Mass Spec: (TOF MS ES+): m/z 563 (M+, 100); HRMS (TOF MS ES+) calcd for C26H21F3N2O5S2, [M+H] 563.0922, found 563.0911.


Example 39
Synthesis of [2,6-bis-(3-(methylsulfonyl)-phenyl)-pyridin-4-yl]-(4-trifluoromethoxy-phenyl)-amine (B31)



embedded image


This compound was prepared by the process disclosed in Example 37.


M.P.: 231° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 8.9 min, 99.96% purity.



1H NMR (300 MHz, DMSO-d6, TMS): δ 3.32 (s, 6H), 5.76 (s, 1H), 7.38-7.47 (m, 4H), 7.59 (s, 1H), 7.83 (t, J=7.8 Hz, 2H), 8.03-8.48 (m, 4H), 8.59 (apt d, J=1.5 Hz, 2H), 9.36 (s, 1H).


Mass Spec: (TOF MS ES+): m/z 563 (M+H, 100); HRMS (TOF MS ES+) calcd for C26H21F3N2O5S2, [M+H] 563.0922, found 563.0920.


Example 40
Synthesis of 1-{3-[6-(3-acetyl-phenyl)-4-(4-trifluoromethoxy-phenylamino)-pyridin-2-yl]-phenyl}-ethanone (B33)



embedded image


This compound was prepared by the process disclosed in Example 37.


M.P.: 160° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4(0.01M, pH 3.2):CH3CN], 264 nm, Rt 19.9 min, 99.6% purity.



1H NMR: (300 MHz, CHCl3, TMS): δ 2.68 (s, 6H), 6.42 (s, 1H), 7.24-7.30 (m, 6H), 7.57 (t, J=7.8 Hz, 2H), 8.00 (dt, J=1.8, 6.6 Hz, 2H), 8.26-8.29 (m, 1H), 8.62 (br s, 2H).


Mass Spec: (TOF MS ES+): m/z 491 (M+H, 100); HRMS (TOF MS ES+) calcd for C28H21F3N2O3, [M+H] 491.1582, found 491.1582.


Example 41
Synthesis of 1-{4-[2,6-bis-(4-fluoro-phenyl)-pyridin-4-ylamino]-phenyl}-ethanone (B34)



embedded image


This compound was prepared by the process disclosed in Example 37.


M.P.: 220° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4(0.01M, pH 3.2):CH3CN], 264 nm, Rt 17.3 min, 95.6% purity.



1H NMR: (300 MHz, CHCl3, TMS): δ 2.60 (s, 3H), 6.49 (s, 1H), 7.13-7.20 (m, 4H), 7.26 (s, 2H), 7.32 (s, 2H), 7.99-8.08 (m, 6H).


Mass Spec: (TOF MS ES+): m/z 401 (M+, 100); HRMS (TOF MS ES+) calcd for C25H18F2N2O, [M+H] 401.1465, found 401.1466.


Example 42
Synthesis of [2,6-bis-(3-N,N-dimethyl-benzamide)-pyridin-2-yl]-(4-trifluoromethoxy-phenyl)-amine (B35)



embedded image


This compound was prepared by the process disclosed in Example 37.


M.P.: 175° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 4.8 min, 98.1% purity.



1H NMR: (300 MHz, CHCl3, TMS): δ 3.00 (br s, 6H), 3.15 (br s, 6H), 7.15 (s, 2H), 7.23 (br s, 4H), 7.26 (s, 1H), 7.40-7.49 (m, 4H), 8.02-8.04 (br m, 4H).


Mass Spec: (TOF MS ES+): m/z 549 (M+, 100).


Example 43
Synthesis of 3-{2,6-bis-(4-fluoro-phenyl)-pyridin-4-ylamino]-benzenethiol; compound with acetic acid methyl ester (B36)



embedded image


To thiocarbonic acid S-[3-(2,6-dichloro-pyridin-4-ylamino)-phenyl] ester-O-methyl ester (317.7 mg, 1 mmol) dissolved in acetonitrile (10 mL) and 0.4 M Na2CO3 (10 mL, 4 mmol) were added 4-fluorophenyl boronic acid (280.0 mg, 2 mmol) and palladium (0) tetrakis(triphenylphosphine) (58 mg, 0.05 mmol). The resulting mixture was allowed to stir at reflux for 12-18 hours. The sample was diluted in dichloromethane and filtered through Celite™. The Celite™ was washed with dichloromethane, and the filtrate was washed two times with water and one time with brine. The organic phase was dried over potassium carbonate and concentrated by rotary evaporation. The resulting sample was dried for 12-18 hours under vacuum. Purification (Biotage Horizon HPFC chromatography system, SiO2, 98:2 dichloromethane:methanol) gave a white solid product (383 mg, 88%).


M.P.: 180° C.


HPLC: Inertsil ODS-3V C18, 40:10:50 [KH2PO4 (0.01M, pH 3.2):CH3CN:MeOH], 264 nm, Rt 11.2 min, 99.0% purity.



1H NMR (300 MHz, CDCl3, TMS): δ 3.10 (s, 3H), 6.49 (s, 1H), 7.12-7.18 (m, 4H), 7.242 (s, 2H), 7.55-7.64 (m, 3H), 7.79-7.80 (m, 1H), 8.01-8.06 (m, 4H).


Mass Spec: (TOF MS ES+): m/z 437 (M+H, 100); HRMS (TOF MS ES+) calcd for C24H18F2N2O2S, [M+H] 437.1135, found 437.1138.


Example 44
Synthesis of [2,6-bis-(4-fluoro-phenyl)-pyridin-4-yl]-methyl-(4-trifluoromethoxy-phenyl)-amine (B29)



embedded image


To [2,6-bis-(4-fluoro-phenyl)-pyridin-4-yl]-(4-trifluoromethoxy-phenyl)-amine in anhydrous THF (2 mL) under N2 atmosphere was added lithium bis(trimethylsilyl)amide (0.2 mL, 1 M in THF, 0.2 mmol) at room temperature. The reaction was stirred for approximately 19 minutes, after which time iodomethane (0.012 mL, 0.2 mmol) was added, and the reaction was stirred for an additional 3 hours. Upon quenching with water, the mixture was diluted with dichloromethane and washed one time with brine. The organic layer was separated and dried over anhydrous potassium carbonate, filtered, and concentrated under vacuum. Biotage Horizon HPFC system chromatography (SiO2, 20:80 ethyl acetate:hexanes) yielded a solid product (6 mg, 10%).


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 30.5 min, 97% purity.



1H NMR: (300 MHz, CHCl3, TMS): δ 3.45 (s, 3H), 6.92 (s, 2H), 7.13 (apt t, J=8.7 Hz), 4H), 7.32 (s, 4 H), 7.96-8.00 (m, 4 H).


Example 45
Synthesis of 2,6-di(4-fluoro phenyl)-4-piperidino pyridine (B62)



embedded image


A mixture of 2,6-dichloro-4-piperdino pyridine (0.200 g, 1.09 mmol), 4-fluoro boronic acid (0.189 g, 1.30 mmol) and tetrakis(triphenylphosphine)Pd(0) [Pd(PPh3)4] (63 mg, 0.05 mmol), 1N Na2CO3 solution (2.2 mL), dioxane (15 mL) was prepared in a 10 mL vessel. This reaction was maintained in a CEM microwave at a pressure of 250 psi and a temperature of 200° C., and subjected the microwave radiationat 250 W for 30 minutes. Water was added (200 mL) to the reaction mixture and the product was extracted with ethyl acetate. The organic layer was dried over sodium sulfate and concentrated under reduced pressure. The residue was purified by column chromatography over 230-400 mesh silica gel by elution with 10:90 ethyl acetate:hexanes to afford the product as a pure colorless solid (purity 99%, yield 85%).


M.P.: 201-203° C.



1H NMR (200 MHz, CDCl3) δ: 7.90-8.06 (m, 4H), 7.08-7.24 (m, 6H), 3.24-3.56 (m, 4H), 1.55-1.75 (m, 6H).


Mass Spec (CI-MS): m/z 351 (M++1, 100%).


IR (neat) cm−1: 2917, 1603, 1435, 1225, 1152, 987, 955, 824, 779, 562.


Example 46
Synthesis of 2′,6′-bis-(4-trifluoromethoxy-phenyl)-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl (B63)



embedded image


This compound was prepared by at least one of the processes disclosed in Examples 54-55, and could be prepared by both of the processes disclosed in these examples.


M.P.: 119-121° C.



1H NMR (200 MHz, CDCl3): δ 7.99-8.09 (m, 4H), 7.05-7.32 (m, 6H), 3.24-3.56 (m, 4H), 1.55-1.75 (m, 6H).


Mass Spec: (CI-MS) m/z: 483 (M++1, 100%).


IR (neat) cm−1: 3431, 2943, 1603, 1445, 1272, 1151, 830.


Example 47
Synthesis of N-ethyl-3-[6-(3-methanesulfonyl-phenyl)-4-(4-trifluoromethoxy-phenylamino)-pyridin-2-yl]-benzamide (B32)



embedded image


To 3-[6-chloro-4-(4-trifluoromethoxy-phenylamino)-pyridin-2-yl]-N-ethyl-benzamide (0.166 g, 0.4 mmol) and (3-methylsulfonylphenyl) boronic acid (0.1624 g, 0.8 mmol) dissolved in acetonitrile (8 mL) and Na2CO3 (8 mL, 2.0 M) was added palladium (0) tetrakis(triphenylphosphine) (0.0471 g, 0.04 mmol). The reaction mixture was stirred and refluxed for 12-18 hours under N2. The reaction mixture was diluted with CH2Cl2 and filtered through Celite™, then rinsed with CH2Cl2. The filtrate was washed two times with water and one time with brine. The organic phase was collected and dried over potassium carbonate. The filtered sample was concentrated, and the resulting solid was dried for 12-18 hours under vacuum. Biotage Horizon HPFC system chromatography (SiO2, 93:6:1 CH2Cl2:CH3OH:NH4OH) yielded a light yellow solid (70 mg, 33%).


M.P.: 85° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4(0.01M, pH 3.2):CH3CN], 264 nm, Rt 8.3 min, 96.2% purity.



1H NMR: (300 MHz, CHCl3, TMS): δ 1.27 (t, J=7.2 Hz, 3H), 3.09 (s, 3H), 3.48-3.58 (m, 2H), 6.46 (t, J=5.4 Hz, 1H), 7.00 (s, 1H), 7.21-7.28 (m, 5H), 7.42-7.70 (m, 8H), 7.83 (br d, J=7.8 Hz, 1H), 7.95 (dt, J=0.9, 8.4 Hz, 1H), 8.10-8.13 (m, 1H), 8.25 (apt dt, J=1.2, 6.9 Hz, 1H), 8.40 (t, J=1.2 Hz, 1H), 8.56 (t, J=1.5 Hz, 1H).


Mass Spec: LC-MSD (ES+): m/z 556 (M+H, 100).


Example 48
Synthesis of 1-[3-(4-hydroxy-6′-phenyl-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4′-ylamino)-phenyl]-ethanone (B48)



embedded image


To 1-[3-(6′-chloro-4-hydroxy-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4′-ylamino)-phenyl]-ethanone (0.445 g, 1.2 mmol) and phenyl boronic acid (0.2954 g, 2.4 mmol) dissolved in acetonitrile (10 mL) and Na2CO3 (10 mL, 2.0 M) was added palladium(O) tetrakis triphenylphosphine (0.1386 g, 0.12 mmol). The reaction mixture was stirred and refluxed for 12-18 hours under N2. The reaction mixture was diluted with CH2Cl2 and filtered through Celite™, then rinsed with CH2Cl2. The filtrate was washed two times with water and one time with brine. The organic phase was collected and dried over potassium carbonate. The filtered sample was concentrated, and the resulting solid was dried for 12-18 hours under vacuum. Flash chromatography (SiO2, 93:6:1 CH2Cl2:MeOH:NH4OH) yielding a light yellow solid (83 mg, 17%).


M.P.: 95° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 2.9 min, 91.5% purity.



1H NMR: (300 MHz, CHCl3, TMS): δ 1.55-1.67 (m, 4H), 1.7-2.02 (m, 2H), 2.60 (s, 3H), 3.13-3.22 (m, 2H), 3.89-3.93 (m, 1H), 4.15 (dt, J=3.9, 13.5 Hz, 2H), 6.04 (s, 1H), 6.23 (d, J=1.5Hz, 1H), 6.74 (d, J=1.5Hz, 1H), 7.25-7.44 (m, 5H), 7.61-7.65 (m, 1H), 7.77-7.78 (m, 1H), 7.92-7.96 (m, 2H).


Mass Spec: LC-MSD (ES+): m/z 388 (M+H, 92.2).


Example 49
Synthesis of [2-(4-fluoro-phenyl)-6-morpholin-4-yl-pyridin-4-yl]-(4-trifluoromethoxy-phenyl)-amine (B49)



embedded image


To (2-chloro-6-morpholin-4-yl-pyridin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (0.03 g, 0.08 mmol) and 4-fluorophenyl boronic acid (0.0284 g, 0.16 mmol) dissolved in acetonitrile (5 mL) and Na2CO3 (5 mL, 0.4M) was added palladium (0) tetrakis-(triphenylphosphine) (0.0099 g, 0.008 mmol). The reaction mixture was stirred and refluxed for 12-18 hours under N2. The reaction mixture was diluted with CH2Cl2 and filtered through Celite™, then rinsed with CH2Cl2. The filtrate was washed one time with water and one time with brine. The organic phase was collected and dried over potassium carbonate. The filtered sample was concentrated, and the resulting solid was dried for 12-18 hours under vacuum. Flash chromatography (SiO2, 50:50 hexanes:ethyl acetate) yielded a pale yellow solid (30 mg, 86%).


M.P.: 192° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 13.6 min, 97.3% purity.



1H NMR: (300 MHz, DMSO-d6, TMS): δ 3.45-3.46 (br m, 4H), 3.72 (br s, 4H), 6.28 (s, 1H), 6.85 (s, 1H), 7.22-7.30 (m, 6H), 7.95-8.00 (m, 2H), 8.82 (s, 1H).


Mass Spec: (TOF MS ES+): m/z 434 (M+H, 100); HRMS (TOF MS ES+) calcd for C22H19F4N3O2, [M+H] 434.1491, found 434.1481.


Example 50
Synthesis of [2-(3-methanesufonyl-phenyl)-6-morpholin-4-yl-pyridin-4-yl]-(4-trifluoro-methoxy-phenyl)-amine (B50)



embedded image


To (2-chloro-6-morpholin-4-yl-pyridin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (0.1872 g, 0.5 mmol) and (3-methylsulfonyl phenyl) boronic acid (0.2011 g, 1.0 mmol) dissolved in acetonitrile (10 mL) and Na2CO3 (10 mL, 2.0 M) was added palladium (0) tetrakis(triphenylphoshine) (0.0581 g, 0.05 mmol). The reaction mixture was stirred and refluxed for 12-18 hours under N2. The reaction was monitored by TLC, and after 18 h (3-methylsulfonylphenyl) boronic acid (0.2010 g, 1.0 mmol), Pd(PPh3)4 (0.0581 g, 0.05 mmol) was added stirred and refluxed for another 12-18 hours under N2. The reaction mixture was diluted with CH2Cl2 and filtered through Celite™, then rinsed with CH2Cl2. The filtrate was washed two times with water and one time with brine. The organic phase was collected and dried over potassium carbonate. The filtered sample was concentrated, and the resulting solid was dried for 12-18 hours under vacuum. Biotage Horizon HPFC system chromatography (SiO2, 50:50 hexanes:ethyl acetate) yielded a pale yellow solid (199 mg, 80%).


M.P.: 196° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4(0.01M, pH 3.2):CH3CN], 264 nm, Rt 8.4 min, 99.0% purity.



1H NMR: (300 MHz, CHCl3, TMS): δ 3.09 (s, 3H), 3.54 (t, J=4.8 Hz, 4H), 3.84 (t, J=4.8 Hz, 4H), 6.09 (s, 1H), 6.19 (d, J=1.8 Hz, 1H), 6.76 (d, J=1.5 Hz, 1H), 7.22 (s, 4H), 7.62 (t, J=7.8 Hz, 1H), 7.91-7.95 (m, 1H), 8.21-8.23 (m, 1H), 8.5 (t, J=1.8 Hz, 1H).


Mass Spec: LC-MSD (ES+): m/z 494 (M+H, 100).


Example 51
Synthesis of ]-{3-[6-morpholin-4-yl-4-(4-trifluoromethoxy-phenylamino)-pyridin-2-yl]-phenyl}-ethanone (B51)



embedded image


To (2-chloro-6-morpholin-4-yl-pyridin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (0.1862 g, 0.5 mmol) and 3-acetylphenyl boronic acid (0.1621 g, 1.0 mmol) dissolved in acetonitrile (10 mL) and Na2CO3 (10 mL, 2.0 M) was added palladium (0) tetrakis(triphenylphosphine) (0.0579 g, 0.05 mmol). The reaction mixture was stirred and refluxed for 12-18 hours under N2. The reaction was monitored by TLC after 18 h 3-acetyl phenyl boronic acid (0.0821 g, 0.5 mmol), palladium (0) tetrakis(triphenylphosphine) (0.0288 g, 0.025 mmol) was added stirred at reflux for 12-18 hours under N2. The reaction mixture was diluted with CH2Cl2 and filtered through Celite™, then rinsed with CH2Cl2. The filtrate was washed two times with water and one time with brine. The organic phase was collected and dried over potassium carbonate. The filtered sample was concentrated, and the resulting solid was dried for 12-18 hours under vacuum. Biotage Horizon HPFC system chromatography (SiO2, 70:30 hexanes:ethyl acetate) yielded a pale brown solid (160 mg, 70%).


M.P.: 122° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 10.6 min, 99.4% purity.



1H NMR: (300 MHz, CHCl3, TMS): δ 2.65 (s, 3H), 3.55 (apt t, J=4.8 Hz, 4H), 3.84 (apt t, J=5.1 Hz, 4H), 6.05 (s, 1H), 6.18 (d, J=1.5Hz, 1H), 6.79 (d, J=1.5 Hz, 1H), 7.21 (s, 4H), 7.52 (t, J=7.8 Hz, 1H), 7.95 (dd, J=1.5, 6.6 Hz, 1H), 8.06 (dd, J=1.5, 7.7 Hz, 1H), 8.50-8.519 (m, 1H).


Mass Spec: (TOF MS ES+): m/z 458 (M+H, 100); HRMS (TOF MS ES+) calcd for C24H22F3N3O3, [M+H] 458.1691, found 458.1686.


Example 52
Synthesis of {3-[6-morpholin-4-yl-4-(4-trifluoromethoxy-phenylamino)-pyridin-2-yl]-phenyl}-pyrrolidin-1-yl-methanone (B52)



embedded image


To (2-chloro-6-morpholin-4-yl-pyridin-4-yl)-(4-trifluoromethoxy-phenyl)-amine (0.1806 g, 0.5 mmol) and 3-(pyrrolidine-1-carbonyl)phenyl boronic acid (0.2185 g, 1.0 mmol) dissolved in acetonitrile (10 mL) and Na2CO3 (10 mL, 2.0 M) was added palladium (0) tetrakis(triphenylphoshine) (0.0581 g, 0.05 mmol). The reaction mixture was stirred and refluxed for 12-18 hours under N2. The reaction was monitored by TLC, and after 18 h 3-(pyrrolidine-1-carbonyl)phenyl boronic acid (0.108 g, 0.5 mmol), palladium (0) tetrakis(triphenylphoshine) (0.0291 g, 0.025 mmol) was added stirred at reflux for 12-18 hours under N2. The reaction mixture was diluted with CH2Cl2 and filtered through Celite™, then rinsed with CH2Cl2. The filtrate was washed two times with water and one time with brine. The organic phase was collected and dried over potassium carbonate. The filtered sample was concentrated, and the resulting solid was dried for 12-18 hours under vacuum. Biotage Horizon HPFC system chromatography (SiO2, 98:2 CH2Cl2:MeOH) yielded a pale brown solid (185 mg, 75%),


M.P.: 179° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 6.2 min, 96.8% purity.



1H NMR: (300 MHz, CHCl3, TMS): δ 1.62 (br s, 1H), 1.86 (pentet, J=6.6 Hz, 2H), 1.97 (pentet, J=6.9 Hz, 2H), 3.44 (t, J=6.3 Hz, 2H), 3.52 (br t, J=4.8 Hz, 3H), 3.67 (t, J=6.9 Hz, 2H), 3.82 (t, J=5.1 Hz, 3H), 6.14 (d, J=1.8 Hz, 1H), 6.21 (s, 1H), 6.74 (d, J=1.5 Hz, 1H), 7.19 (s, 3H), 7.40-7.55 (m, 3H), 7.63-7.70 (m, 1H), 7.97 (apt d, J=7.5 Hz, 1H), 8.07 (s, 1H).


Mass Spec: (TOF MS ES+): m/z 513 (M+H, 100).


Example 53
Synthesis of thiocarbonic acid O-methyl ester S-{3-[6-pyrrolidin-1-yl-4-(4-trifluoromethoxy-phenylamino)-pyridin-2-yl]-phenyl}ester (B37)



embedded image


This compound was prepared by a method analogous to that disclosed in Example 52.


M.P.: 149° C.


HPLC: Inertsil ODS-3V C18, 40:10:50 [KH2PO4 (0.01M, pH 3.2):CH3CN:MeOH], 264 nm, Rt 4.0 min, 98% purity.



1H NMR (300 MHz, CDCl3, TMS): δ 1.97-2.02 (m, 4H), 3.10 (s, 1H), 3.51 (br s, 4H), 5.92 (d, J=1.5 Hz, 1H), 6.65 (d, J=1.5 Hz, 1H), 7.17-7.21 (m, 4H), 7.58 (t, J=7.8 Hz, 1H), 7.90 (apt d, J=7.8 Hz, 1H), 8.19 (apt d, J=7.8 Hz, 1H), 8.49 (s, 1H).


Mass Spec: (TOF MS ES+): m/z 478 (M+H).


Example 54
Synthesis of 1-(3-[6-pyrrolidin-1-yl-4-(4-trifluoromethoxy-phenylamino)-pyridin-2-yl]-phenyl}-ethanone (B38)



embedded image


This compound was prepared by a method analogous to that disclosed in Example 52.


M.P.: 95° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 3.6 min, 95.2% purity.



1H NMR: (300 MHz, CHCl3, TMS): δ 1.97-2.01 (m, 4H), 2.65 (s, 3H), 3.51 (t, J=6.6 Hz, 4H), 5.93 (d, J=1.8 Hz, 1H), 6.05 (s, 1H), 6.66 (d, J=1.5 Hz, 1H), 7.20 (apt s, 3H), 7.49 (t, J=7.8 Hz, 2H), 7.93 (dt, J=1.5, 7.8 Hz, 1H), 8.18 (dt, J=1.5, 7.8 Hz, 1H), 8.56 (t, J=1.5 Hz, 1H).


Mass Spec: LC-MSD (ES+): m/z 442 (M+H, 100).


Example 55
Synthesis of [2-(4-fluoro-phenyl)-6-(4-methyl-piperazin-1-yl)-pyridin-4-yl]-(4-trifluoromethoxy-phenyl)-amine (B39)



embedded image


This compound was prepared by a method analogous to that disclosed in Example 52.


M.P.: 142° C.


HPLC: Inertsil ODS-3V C18, 40:10:50 [KH2PO4 (0.01M, pH 3.2):CH3CN:MeOH], 264 nm, Rt 6.3 min, 96.7% purity.



1H NMR (600 MHz, CDCl3, TMS): δ 2.34 (s, 3H), 2.51-2.53 (m, 4H), 3.58-3.60 (m, 4H), 5.91 (s, 1H), 6.125 (s, 1H), 6.66 (s, 1H), 7.06-7.09 (t, J=8.4 Hz, 2H), 7.19 (apt s, 4H), 7.90-7.92 (m, 2H).


Mass Spec: LC-MSD (ES+): m/z 447 (M+H, 98.5).


Example 56
Synthesis of (6-phenyl-4-pyrrolidin-1-yl-pyridin-2-yl)-(4-trifluoromethoxy-phenyl)-amine (B64)



embedded image


To a solution of (6-chloro-4-pyrrolidin-1-yl-pyridin-2-yl)-(4-trifluoromethoxy-phenyl)-amine (120 mg, 0.336 mmol) in 1,4-dioxane (8 mL) was added potassium-tert-butoxide (75 mg, 0.670 mmol) and tetrakis(triphenylphosphine)palladium (0) (20 mg, 0.017 mmol), followed by phenyl boronic acid (102 mg, 0.836 mmol). The resulting reaction mixture was refluxed for 10 hours, after which time water was added 100 mL to the reaction mixture, and the product was extracted with ethyl acetate (100 mL×3). The organic layer was dried over sodium sulphate, filtered, and the filtrate was concentrated under vacuum. The resulting product was purified by column chromatography using 230-400 mesh silica gel (20:80 acetone:petroleum ether) to afford the desired compound (75 mg, purity 99%, yield 6%.).


M.P.: 141-143° C.



1H NMR (200 MHz, CDCl3): δ 7.90-7.96 (d, 2H, J=9.67 Hz), 7.40-7.48 (m, 5H), 7.15 (d, 2H, J=25.34), 6.50 (s, 1H), 6.47 (s, 1H), 5.90 (s, 1H), 3.20-3.39 (t, 4H, J=13.00 Hz), 1.99-2.06 (m, 4H).


Mass Spec: (CI-MS) m/z: 400 (M++1, 100%).


IR (neat) cm−1: 2924, 2599, 1555, 1505, 1268, 1157, 983, 814.


Example 57
Synthesis of [6′-(4-fluorophenyl)-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-2′-yl]-(4-trifluoromethoxyphenyl)-amine (B56)



embedded image


To a solution of (6′-chloro-3,4,5,6-tetrahydro-2H[1,4′]bipyridinyl-2′-yl)-(4-trifluoromethoxyphenyl)-amine (200 mg, 0.53 mmol) in 1,4-dioxane (8 mL) were added potassium carbonate (148 mg, 1.06 mmol), and tetrakis(triphenylphosphine)palladium(0) (62 mg, 0.053 mmol), followed by 4-fluorophenyl boronic acid (150 mg, 1.6 mmol). This reaction mixture was refluxed for 8 hours, after which time water was added (10 mL) to the reaction mixture, and the product was extracted with ethyl acetate. The organic layer was dried over sodium sulphate, filtered, and the filtrate was concentrated under vacuum. The resulting product was purified through column chromatography using 230-400 mesh silica gel (15% ethyl acetate in petroleum ether) to afford the desired compound as colorless gummy mass (130 mg, yield 60% ).



1H NMR (400 MHz, CDCl3): δ 7.91-7.86 (m, 2H), 7.4 (d, 2H, J=3.22 Hz), 7.17-7.08 (m, 4H), 6.74-6.71 (br s, 1 H), 6.67 (s, 1H), 6.14 (s, 1H), 3.34-3.33 (m, 4H), 1.66 (m, 6 H).


Mass Spec: (ES-MS) m/z: 432 (M++1, 100%).


IR (neat) cm−1: 3408, 2935, 1607, 1590, 1449, 1261, 1157, 1018, 921, 844, 812.


Example 58
Synthesis of (6′phenyl-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-2′-yl)-(4-trifluoro-methoxyphenyl)-amine (B58)



embedded image


To a solution of (6′-chloro-3,4,5,6-tetrahydro-2H[1,4′]bipyridinyl-2′-yl)-(4-trifluoromethoxyphenyl)-amine (170 mg, 0.45 mmol) in 1,4-dioxane (8 mL) were added potassium carbonate (101 mg, 0.9 mmol), and tetrakis(triphenylphosphine)palladium(0) (52 mg, 0.045 mmol), followed by phenyl boronic acid (111 mg, 0.9 mmol). This reaction mixture was refluxed for 8 hours, after which time water was added (10 mL) to the reaction mixture, and the product was extracted with ethyl acetate (100 mL×3). The organic layer was dried over sodium sulphate, filtered, and the filtrate was concentrated under vacuum. The resulting product was purified through column chromatography using 230-400 mesh silica gel (15% ethyl acetate in petroleum ether) to afford the desired compound as light yellow solid (120 mg, yield 65%).


M.P.: 107-108° C.



1H NMR (400 MHz, CDCl3): δ 7.93 (d, 2H), 7.45-7.349 (m, 5H), 7.169 (d, 2H), 6.75 (s, 1H), 6.5 (s, 1H), 6.16 (s, 1H), 3.348-3.335 (m, 4H), 1.66 (m, 6H).


Mass Spec: (ES-MS) m/z: 414 (M++1, 100%).


IR (neat) cm−1: 2940, 1597, 1556, 1506, 1451, 1263, 1223, 1152, 986, 922.


Example 59
Synthesis of [6′-(3-methanesulfonylphenyl)-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-2′-yl]-(4-trifluoromethoxyphenyl)-amine (B59)



embedded image


To a solution of (6′-chloro-3,4,5,6-tetrahydro-2H[1,4′]bipyridinyl-2′-yl)-(4-trifluoromethoxyphenyl)-amine (200 mg, 0.53 mmol) in 1,4-dioxane (8 mL) were added sodium carbonate (1N solution; 228 mg, 2.12 mmol), and tetrakis(triphenylphosphine)palladium(0) (62 mg, 0.053 mmol), followed by 3-(methanesulphonyl)-phenyl boronic acid (215 mg, 1.06 mmol). This reaction mixture was refluxed for 12 hours, after which time water was added (10 mL) to the reaction mixture, and the product was extracted with ethyl acetate. The organic layer was dried over sodium sulphate, filtered, and the filtrate was concentrated under vacuum. The resulting product was purified through column chromatography using 230-400 mesh silica gel (15% ethyl acetate in petroleum ether) to afford the desired compound as a colorless solid (132 mg, yield 50%).


M.P.: 176-177° C.



1H NMR (400 MHz, CDCl3): δ 8.5 (s, 1H), 8.269 (d, 1H, J=5.1 Hz), 7.95 (d, 1H), 7.65 (t, 2H, J=8.32 Hz), 7.44-7.40 (d, 2H, J=3.49 Hz), 7.19 (d, 2H), 6.78 (s, 1H), 6.45 (s, 1H), 6.17 (s, 1H), 3.36 (m, 4H), 1.68 (m, 6H).


Mass Spec: (ES-MS) m/z: 492 (M++1, 100%), 493 (M++2, 38%).


IR (neat) cm−1: 3362, 2926, 1619, 1595, 1533, 1506, 1464, 1416, 1296, 1251, 1244, 1196, 1149, 1128, 966, 824, 795.


Example 60
Synthesis of 4-[6′-(4-fluorophenyl)-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-2′-ylamino]-N-methyl-benzenesulfonamide (B60)



embedded image


To a solution of 4-(6′-chloro-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-2′-ylamino)-N-methyl-benzenesulfonamide (70 mg, 0.18 mmol) in 1,4-dioxane (8 mL) was added sodium carbonate (1N, 80 mg, 0.72 mmol), and tetrakis(triphenyl-phosphine)palladium(0) (21 mg, 0.018 mmol), followed by 4-fluorophenyl boronic acid (49 mg, 0.36 mmol). This reaction was refluxed for 8 hours, after which time water (100 mL) added to the reaction mixture, and the product was extracted with ethyl acetate (100 mL×3). The organic layer was dried over sodium sulphate, filtered, and the filtrate was and concentrated under vacuum. The resulting product was purified through column chromatography using 230-400 mesh silica gel (5% methanol in dichloromethane) to afford the desired compound as A yellow solid (48 mg, Yield 60%). Purity: 98.2%.


M.P.: 191-192° C.



1H NMR (400 MHz, CDCl3): δ 7.91-7.88 (m, 2H), 7.7 (d, 2H, J=6.9 Hz), 7.578 (d, 2H), 7.13-7.096 (t, 2H), 6.8 (br s, 1H), 6.75 (s, 1H), 6.2 (s, 1H), 4.39 (br s, 1H), 3.38 (m, 4H), 2.63 (s, 3H), 1.68 (m, 6H).


Mass Spec: (CI-MS) m/z: 441 (M++1, 100%).


IR (neat) cm−1: 3387, 2934, 1613, 1584, 1508, 1462, 1331, 1223, 1153, 1093, 819.


Example 61
Synthesis of N-methyl-4-[4-Pyrrolidin-1-yl-6-(4-trifluoromethoxy-phenyl)-pyridin-2-ylamino]-benzenesulfonamide (B66)



embedded image


To a solution of 4-(6-chloro-4-pyrrolidin-1-yl-pyridin-2-ylamino)-N-methyl-benzenesulfonamide (100 mg, 0.27 mmol) in 1,4-dioxane (8 mL) was added 1N sodium carbonate solution (0.3 mL) and tetrakis(triphenylphosphine)palladium(0) (15.8 mg, 0.013 mmol), followed by 4-trifluoromethoxyphenyl boronic acid (67.2 mg, 0.32 mmol). The resulting reaction mixture was refluxed for 8 hours, after which time water was added (100 mL) to the reaction mixture, and the product was extracted with ethyl acetate (100 mL×3). The organic layer was dried over sodium sulphate, filtered, and the filtrate was concentrated under vacuum. The resulting product was purified by column chromatography using 230-400 mesh silica gel (20:80 acetone:petroleum ether) to afford the desired compound pale yellow solid (46 mg, yield 34%.).


M.P.: 83-85° C.



1H NMR (200 MHz, DMSO-d6): δ 9.31 (s, 1H), 8.18 (d, 2H, J=8.67 Hz), 7.94 (d, 2H, J=8.67 Hz), 7.64 (d, 2H, J=8.67 Hz), 7.46 (d, 2H, J=8.33 Hz), 7.12 (br s, 1H), 6.70 (s, 1H), 5.96 (s, 1H), 3.22-3.39 (m, 4H), 2.40 (d, 3H, J=5.00 Hz), 1.99-2.06 (m, 4H).


Mass Spec: (ES-MS) m/z: 493 (M++1, 100%).


IR (neat) cm−1: 3254, 2925, 1618, 1600, 1524, 1326, 1220, 1153, 1092, 979, 917, 856.


Example 62
Synthesis of [6-(3-methanesulfonyl-phenyl)-4-morpholin-4-yl-pyridin-2-yl]-(4-trifluoromethoxy-phenyl)-amine (B68)



embedded image


To a solution of (6-chloro-4-morpholin-4-yl-pyridin-2-yl)-(4-trifluoromethoxy-phenyl)-amine (75 mg, 0.20 mmol) in 1,4-dioxane (8 mL) were added 2N sodium carbonate solution (0.5 mL) and tetrakis(triphenylphosphine)palladium(0) (11.6 mg, 0.010 mmol), followed by 3-methanesulfonyl phenyl boronic acid (48 mg, 0.24 mmol). The resulting reaction mixture was refluxed for 8 hours, after which time water was added (100 mL) to the reaction mixture, and the product was extracted with ethyl acetate (100 mL×3). The organic layer was dried over sodium sulphate, filtered, and the filtrate was concentrated under vacuum. The resulting product was purified by column chromatography using 230-400 mesh silica gel (elution 2:98 methanol:DCM) to afford the desired compound pale yellow solid (30 mg, yield 30%.).


M.P.: 233-235° C.



1H NMR (200 MHz, DMSO-d6): δ 9.12 (s, 1H), 8.45 (d, 2H), 7.74-7.96 (m, 4H), 7.27 (d, 2H, J=11.00 Hz), 7.15 (s, 1H), 6.25 (s, 1H), 3.60-3.85 (m, 4H), 3.15-3.42 (m, 4H), 3.25 (s,3H).


Mass Spec: (ES-MS) m/z: 494 (M++1, 100%).


IR (neat) cm−1: 3386, 2920, 1615, 1450, 1302, 1153, 1015, 959, 832.


Example 63
Synthesis of N-methyl-4-[4-morpholin-4-yl-6-(4-trifluoromethoxy-phenylamino)-pyridin-2-yl]-benzenesulfonamide (B69)



embedded image


To a solution of (6-chloro-4-morpholin-4-yl-pyridin-2-yl)-(4-trifluoromethoxy-phenyl)-amine (50 mg, 0.13 mmol) in 1,4-dioxane (8 mL) were added 2N sodium carbonate solution (0.5 mL), and tetrakis(triphenylphosphine)palladium(0) (7.74 mg, 0.0067 mmol), followed by 3-methyl-4-boron benzenesulfonamide (34.6 mg, 0.16 mmol). The resulting reaction mixture was refluxed for 8 hours, after which time water was added (100 mL) to the reaction mixture, and the product was extracted with ethyl acetate. The organic layer was dried over sodium sulphate, filtered, and the filtrate was concentrated under vacuum. The resulting product was purified by column chromatography using 230-400 mesh silica gel (elution 2:98 methanol:DCM) to afford the desired compound colorless solid (25 mg, yield 36%.).


M.P.: 234-236° C.



1H NMR (200 MHz, DMSO-d6): δ 9.12 (s, 1H), 8.26 (d, 2H, J=8.60 Hz), 7.74-7.87 (m, 4H), 7.47 (d, 1H), 7.25 (d, 2H), 7.15 (s, 1H), 6.24 (s, 1H), 3.78-3.82 (m, 4H), 2.28-2.38 (m, 4H), 2.56 (d, 3H, J=1.60 Hz).


Mass Spec: (CI-MS) m/z: 508 (M+, 100%).


IR (neat) cm−1: 3374, 2925, 1770, 1597, 1450, 1265, 1162, 929.


Example 64
Synthesis of [6-(4-fluoro-phenyl)-4-pyrrolidin-1-yl-pyridin-2-yl]-(4-trifluoromethoxy-phenyl)-amine (B65)



embedded image


This compound was prepared by a process analogous to that disclosed in Example 63.


M.P.: 83-85° C.



1H NMR (200 MHz, CDCl3) δ 7.90-7.96 (d, 2H, J=9.67 Hz), 7.40-7.48 (m, 4H), 6.40 (s, 1H), 5.90 (s, 1H), 5.30 (s, 1H), 3.22-3.39 (t, 4H, J=13.00 Hz), 1.99-2.06 (m, 4H).


Mass Spec: (ES-MS) m/z: 418 (M++1, 100%).


IR (neat) cm−1: 2925, 2854, 1613, 1459, 1352, 1262, 1156, 981, 843, 808.


Example 65
Synthesis of N-methyl-4-[4-(4-methyl-piperazin-1-yl)-6-(4-trifluoromethoxy-phenyl-amino)-pyridin-2-yl]-benzenesulfonamide (B67)



embedded image


This compound was prepared by a process analogous to that disclosed in Example 63.


M.P.: 184-186° C.



1H NMR (200 MHz, CDCl3) δ 8.01 (d, 4H, J=8.67 Hz), 7.17-7.45 (d, 4H, J=9.00 Hz), 6.740 (s, 1H), 6.70 (s, 1H), 6.20 (s, 1H), 4.90 (m, 1H), 3.25-3.42 (m, 4H), 2.46-2.68 (m, 4H), 2.24 (s, 3H), 1.25 (d, 3H, J=7.04 Hz).


Mass Spec: (ES-MS) m/z: 522(M++1, 100%).


IR (neat) cm−1: 3380, 2925, 1595, 1506, 1452, 1261, 1160, 813.


Example 66
Synthesis of (4-trifluoromethoxy-phenyl)-[6′(4-trifluoromethoxy-phenyl)-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-2′-yl]-amine (B57)



embedded image


This compound was prepared by a process analogous to that disclosed in Example 63.



1H NMR (400 MHz, CDCl3): δ 7.95 (d, 2H, J=4.83 Hz), 7.4 (d, 2H, J=4.83 Hz), 7.28 (d, 2H), 7.17 (d, 2H, J=8.32 Hz), 6.7 (s, 1H), 6.49 (s, 1H), 6.15 (s, 1H), 3.35 (m, 4H), 1.66 (m, 6H).


Mass Spec: (ES-MS) m/z: 498 (M++1, 100%).


IR (neat) cm−1: 2938, 1607, 1550, 1506, 1449, 1260, 1203, 1162, 1017, 921, 807.


Example 67
Synthesis of 1-[4-(4-hydroxy-6′-phenyl-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-2′ylamino)-phenyl]-ethanone (B44)



embedded image


To a solution of 2′-chloro-6′-phenyl-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-4-ol (40 mg, 0.13 mmol) in toluene (4 mL) were added potassium tert-butoxide (31 mg, 0.26 mmol), palladium(II)acetate (3 mg, 0.156 mmol), BINAP (4 mg, 0.0065 mmol) and 1-(4-aminophenyl)-ethanone (22 mg, 0.156 mmol) in a 10 mL reaction vessel. This reaction mixture was subjected the microwave radiation, in which the microwave power was 250 W, to attain a reaction temperature of 150° C. After this reaction proceeded for 30 min, water (5 mL) was added to the reaction mixture and the reaction product was extracted with ethyl acetate. The organic layer was dried over sodium sulphate, filtered, and the filtrate concentrated under vacuum. The product was purified through column chromatography using 230-400 mesh silica gel (5% methanol in dichloromethane) to afford the desired compound as a yellow solid (29 mg, yield 55%, purity 96%).


M.P.: 150-152° C.



1H NMR (400 MHz, CDCl3): δ 7.94-7.92 (m, 4H), 7.5-7.39 (m, 5H), 6.82 (m, 2H), 6.29 (s, 1H), 3.98-3.94 (m, 1H), 3.78 (m, 2H), 3.19 (m, 2H), 2.56 (s, 3H), 2.01 (m, 2H), 1.69 (m, 2H).


Mass Spec: (ES-MS) m/z: 388 (M++1, 100%).


Example 68
Synthesis of 1-[4-(2-morpholin-4-yl-6-phenyl-pyridin-4-ylamino)-phenyl]-ethanone (B45)



embedded image


To 1-[4-(2-chloro-6-morpholin-4-yl-pyridin-4-ylamino)-phenyl]-ethanone (251.7 mg, 0.75 mmol) dissolved in acetonitrile (10 mL) and 2 M Na2CO3 (10 mL, 20 mmol) was added phenyl boronic acid (184.9 mg, 1.5 mmol) and palladium tetrakis(triphenylphosphine) (87.3 mg, 0.075 mmol). The resulting mixture was allowed to stir at reflux for 12-18 hours. The sample was diluted in dichloromethane and filtered through Celite™. The Celite™ was washed with dichloromethane; the filtrate was washed two times with water and one time with brine. The organic phase was dried over potassium carbonate and concentrated by rotary evaporation. The resulting sample was dried for 12-18 hours under vacuum. Column chromatography (SiO2, 96:3:1 dichloro-methane:methanol:ammonium hydroxide) gave a light yellow solid (78 mg, 28%).


M.P.: 148° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 7.1 min, 94.8% purity.



1H NMR (300 MHz, CDCl3, TMS): δ 2.57 (s, 3H), 3.57 (t, J=5.1 Hz, 4H), 3.84 (t, J=4.5 Hz, 4H), 6.29 (d, J=1.2 Hz, 1H), 6.54 (br s, 1H), 6.91 (d, J=1.5 Hz, 1H), 7.20 (apt d, J=8.7 Hz, 2H), 7.37-7.45 (m, 3H), 7.92-7.96 (m, 4H) [D2O exchange; peak at 6.54 for 1H disappeared].


Mass Spec: LC-MSD (ES+): m/z 374 (M+H, 94.49).


Example 69
Synthesis of 6′-(4-fluoro-phenyl)-4′-(4-trifluoromethoxy-phenylamino)-3,4,5,6-tetra-hydro-2H-[1,2′]bipyridinyl-4-ol (B46)



embedded image


6′-Chloro-4′-(4-trifluoromethoxy-phenylamino)-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4-ol (0.080 g, 0.2 mmol) and 4-fluorophenyl boronic acid (0.0562 g, 0.4 mmol) dissolved in acetonitrile (5 mL) and Na2CO3 (5 mL, 2.0 M) was added palladium (0) tetrakis(triphenylphosphine) (0.0235 g, 0.02 mmol). The reaction mixture was stirred and refluxed for 12-18 hours under N2. The reaction was monitored by TLC, and after 18 h 4-fluorophenyl boronic acid (0.0381 g, 0.4 mmol), palladium (0) tetrakis (triphenylphosphine) (0.012 g, 0.01 mmol) was added stirred and refluxed for 12-18 hours under N2. The reaction mixture was diluted with CH2Cl2 and filtered through Celite™, then rinsed with CH2Cl2. The filtrate was washed two times with water and one time with brine. The organic phase was collected and dried over potassium carbonate. The filtered sample was concentrated, and the resulting solid was dried for 12-18 hours under vacuum. Biotage Horizon HPFC system chromatography (SiO2, 93:6:1 CH2Cl2:MeOH:NH4OH) yielded a pale brown solid (34 mg, 37%).


M.P. 134° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 4.6 min, 96.1% purity.



1H NMR (300 MHz, CHCl3, TMS): δ 1.48-1.59 (m, 3H), 1.89-1.94 (m, 2H), 3.03-3.01 (m, 2H), 3.81-3.87 (m, 1H), 4.03-4.07 (m, 2H), 5.90 (s, 1H), 6.09 (s, 1H), 6.57 (s, 1H), 7.01 (apt t, J=8.4 Hz, 2H), 7.12 (s, 3H), 7.82-7.87 (m, 2H).


Mass Spec: (TOF MS ES+): m/z 448 (M+, 100); HRMS (TOF MS ES+) calcd for C23H21F4N3O2, [M+H] 448.1648, found 448.1640.


Example 70
Synthesis of 1-[4-(4-hydroxy-6′-phenyl-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4′-ylamino)-phenyl]-ethanone (B47)



embedded image


1-[4-(6′-chloro-4-hydroxy-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4′-ylamino)-phenyl]-ethanone (0.497 g, 1.4 mmol) and phenylboronic acid (0.3421 g, 2.8 mmol) dissolved in acetonitrile (15 mL), THF (5 mL) and Na2CO3 (20 mL, 2.0 M) was added palladium(0) tetrakis(triphenylphosphine) (0.1621 g, 0.14 mmol). The reaction mixture was stirred and refluxed for 12-18 hours under N2. The reaction mixture was diluted with CH2Cl2 and filtered through Celite™, then rinsed with CH2Cl2. The filtrate was washed two times with water and one time with brine. The organic phase was collected and dried over potassium carbonate. The filtered sample was concentrated, and the resulting solid was dried for 12-18 hours under vacuum. Flash chromatography (SiO2, 93:6:1 CH2Cl2:MeOH:NH4OH) yielded a pale brown solid (188 mg, 34%).


M.P.: 70° C.


HPLC: Inertsil ODS-3V C18, 40:10:50 [KH2PO4(0.01M, pH 3.2):CH3CN], 264 nm, Rt 4.1 min, 97.4% purity.



1H NMR: (300 MHz, CHCl3, TMS): δ 1.57-1.68 (m, 3H), 1.69-2.04 (m, 2H), 2.57 (s, 3H), 3.16-3.25 (m, 2H), 3.93 (octet, J=4.2 Hz, 1H), 4.16 (dt, J=4.5, 13.5 Hz, 2H), 6.23 (s, 1H), 6.33 (d, J=1.8 Hz, 1H), 6.85 (d, J=1.8 Hz, 1H), 7.16-7.21 (m, 2H), 7.34-7.45 (m, 3H), 7.92-7.81 (m, 4H).


Mass Spec: LC-MSD (ES+): m/z 388 (M+H, 100).


Example 71
Synthesis of 2′,6′-bis-(4-trifluoromethoxy-phenylamino)-3,4,5,6-tetrahydro-2H[1,4′]bipyridinyl-4-ol (B71



embedded image


To a solution of 2′,6′-dichloro-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-4-ol (280 mg, 1.1 mmol) in toluene (6 mL) were added potassium tert-butoxide (254 mg, 2.2 mmol), palladium(II)acetate (25 mg, 0.11 mmol), BINAP (49 mg, 0.079 mmol) and 4-trifluoromethoxyphenylamine (0.2 mL, 1.134 mmol), in a 10 mL reaction vessel. This reaction mixture was irradiated with microwave radiation at 250 W, to attain a reaction temperature of 160° C. After this reaction proceeded for 15 min, water (5-6 mL) was added to the reaction mixture and the reaction product was extracted with ethyl acetate. The organic layer was dried over sodium sulphate, filtered, and the filtrate concentrated under vacuum. The product was purified through column chromatography using 230-400 mesh silica gel, eluting with 3:97 MeOH:CHCl3, to afford the desired compound as a colorless solid (100 mg, yield 16.7%, purity 96%).


M.P.: 119-120° C.



1H NMR (200 MHz, CDCl3): δ 7.3 (d, 4H), 7.2 (m, 4H), 6.39 (s, 1H), 5.79 (s, 2H), 3.9 (m, 1H, J=4.3 Hz), 3.6 (m, 2H), 3.0 (m, 2H), 1.8 (m, 2H), 1.6 (m, 2H).


Mass Spec: (ES-MS) m/z: 529 (M++1, 100%).


IR (neat) cm−1: 3408, 2929, 1602, 150, 1266, 1201, 1051, 793.


Example 72
Synthesis of 2,6-dichloro-4-(4-methanesulfonyl-phenyl)-pyridine



embedded image


To 2,6-dichloro-4-iodopyridine (0.8296 g, 3 mmol) and 4-methanesulfonylphenyl boronic acid (0.6124 g, 3 mmol) dissolved in acetonitrile (30 mL) and Na2CO3 (30 mL, 0.4M) was added palladium (0) tetrakis(triphenylphosphine) (0.1733 g, 0.15 mmol). The reaction mixture was stirred and refluxed for 12-18 hours under N2. The reaction mixture was diluted with CH2Cl2 and filtered through Celite™, then rinsed with CH2Cl2. The filtrate was washed two times with water and one time with brine. The organic phase was collected and dried over potassium carbonate. The filtered sample was concentrated, and the resulting solid was dried for 12-18 hours under vacuum. Biotage Horizon HPFC system chromatography (SiO2, 50:50 hexanes:ethyl acetate) yielded a off white solid (640 mg, 70%).


M.P.: 165° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 6.2 min, 99.4% purity.



1H NMR: (600 MHz, CDCl3, TMS, 55° C.): δ 3.09 (s, 3H), 7.47 (s, 4H), 7.76-7.78 (m, 2H), 8.07-8.08 (m, 2H).


Mass Spec: (EI+): m/z 301 (M+, 100).


Example 73
Synthesis of [4-(2,6-dichloro-pyridin-4-yl)-phenyl]-morpholin-4-yl-methanone (B73)



embedded image


To 2,6-dichloro-4-iodopyridine (0.8238 g, 3 mmol) and [(4-morpholine-4-carbonyl)-phenyl]boronic acid (0.7051 g, 3 mmol) dissolved in acetonitrile (23 mL), THF (7 mL), and Na2CO3 (30 mL, 0.4M) was added palladium (0) tetrakis(triphenylphosphine) (0.1758 g, 0.15 mmol). The reaction mixture was stirred and refluxed for 2.5 h under N2. The reaction mixture was diluted with CH2Cl2 and filtered through Celite™, then rinsed with CH2Cl2. The filtrate was washed one time with water and one time with brine. The organic phase was collected and dried over potassium carbonate. The filtered sample was concentrated, and the resulting solid was dried for 12-18 hours under vacuum. Biotage Horizon HPFC system chromatography (SiO2, 50:50 hexanes:ethyl acetate) yielded a pale yellow product (570 mg, 56%).


M.P.: 187° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4(0.01M, pH 3.2):CH3CN], 264 nm, Rt 5.6 min, 99.2% purity.



1H NMR: (600 MHz, CDCl3, TMS, 55° C.): δ 3.44 (br s, 2H), 3.64 (br s, 2H), 3.79 (br s, 4H), 7.45 (s, 2H), 7.54 (d, J=8.4 Hz, 2H), 7.63 (apt d, J=8.4 Hz, 2H).


MS (EI+): m/z 337 (M+1, 30), 250 (100).


Examples 74
Synthesis of 2,6-dichloro-4-phenyl-pyridine (B74)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 73.


M.P.: 35° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 15.6 min, 97.3% purity.



1H NMR (600 MHz, CDCl3, TMS): δ 7.54 (s, 1H), 7.48-7.50 (m, 3H), 7.56-7.58 (m, 2H).


Mass Spec: (EI+): m/z 223 (M+H, 100).


Example 75
Synthesis of 2,6-dichloro-4-(4-fluoro-phenyl)-pyridine (B75)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 73.


M.P.: 147° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4(0.01M, pH 3.2):CH3CN], 264 nm, Rt 13.8 min, 92.4% purity.



1H NMR: (600 MHz, CDCl3, TMS, 55° C.): δ 7.17-7.20 (m, 2H), 7.41 (s, 2H), 7.56-7.59 (m, 2H).


Mass Spec: (EI+): m/z 241 (M+1, 100).


Example 76
Synthesis of 2,6-dichloro-4-(4-methanesulfonyl-phenyl)-pyridine (B76)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 73.


M.P.: 165° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 6.2 min, 99.4% purity.



1H NMR: (600 MHz, CDCl3, TMS, 55° C.): δ 3.09 (s, 3H), 7.47 (s, 4H), 7.76-7.78 (m, 2H), 8.07-8.08 (m, 2H).


Mass Spec: (EI+): m/z 301 (M+, 100).


Example 77
Synthesis of 2,6-dichloro-4-(4-fluoro-phenyl)-pyridine (13)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 73.


M.P.: 147° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 13.8 min, 92.4% purity.



1H NMR (600 MHz, CDCl3, TMS, 55° C.): δ 7.17-7.20 (m, 2H), 7.41 (s, 2H), 7.56-7.59 (m, 2H).


Mass Spec (EI+): m/z 241 (M+1, 100).


Example 78
Synthesis of [4-(2,6-dichloro-pyridin-4-yl)-phenyl]-morpholin-4-yl-methanone (17)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 73.


M.P.: 187° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 5.6 min, 99.2% purity.



1H NMR (600 MHz, CDCl3, TMS, 55° C.): δ 3.44 (br s, 2H), 3.64 (br s, 2H), 3.79 (br s, 4H), 7.45 (s, 2H), 7.54 (d, J=8.4 Hz, 2H), 7.63 (apt d, J=8.4 Hz, 2H).


MS (EI+): m/z 337 (M+1, 30), 250 (100).


Example 79
Synthesis of 2,6-dichloro-4-(4-trifluoromethoxy-phenyl)-pyridine (19)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 73.


Example 80
Synthesis of 2,6-dichloro-4-phenyl-pyridine (27)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 73.


M.P.: 35° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 15.6 min, 97.3% purity.



1H NMR (600 MHz, CDCl3, TMS): δ 7.54 (s, 1H), 7.48-7.50 (m, 3H), 7.56-7.58 (m, 2H).


MS (EI+): m/z 223 (M+H, 100).


Example 81
Synthesis of 1-[4,6-bis-(4-fluoro-phenyl)-pyridin-2-yl]-4-methyl-piperazine (B40)



embedded image


1-[6-Chloro-4-(4-fluoro-phenyl)-pyridin-2-yl]-4-methyl-piperazine (0.157 g, 0.5 mmol) and 4-fluorophenyl boronic acid (0.0862 g, 0.6 mmol) were dissolved in acetonitrile (5 mL) and Na2CO3 (5 mL, 0.4M) followed by addition of palladium (0) tetrakis(triphenylphoshine) (0.0292 g, 0.025 mmol). The reaction mixture was allowed to stir and reflux for 12-18 hours. The reaction mixture was diluted with CH2Cl2 and filtered through Celite™, then rinsed with CH2Cl2. The filtrate was washed one time with water and one time with brine. The organic phase was collected and dried over potassium carbonate. The filtered sample was concentrated, and the resulting solid was dried for 12-18 hours under vacuum. Biotage Horizon HPFC system chromatography (SiO2, 93:6:1 CH2Cl2:MeOH:NH4OH) yielded a pale brown solid product (125 mg, 67%).


M.P.: 102° C.


HPLC: Inertsil ODS-3V C18, 40:10:50 [KH2PO4 (0.01M, pH 3.2):CH3CN:MeOH], 264 nm, Rt 5.1 min, 96.5% purity.



1H NMR: (600 MHz, CDCl3, TMS, 55° C.): δ 2.38 (s, 3H), 2.58 (t, J=4.8 Hz, 4H), 3.73 (t, J=4.8 Hz, 4H), 6.72 (s, 1H), 7.11-7.18 (m, 4H), 7.21 (s, 1H), 7.6-7.63 (m, 2H), 8.03-8.06 (m, 2H).


Mass Spec: (TOF MS ES+): m/z 366 (M+H, 100); HRMS (TOF MS ES+) calcd for C22H22F2N3, [M+H] 366.1782, found 366.1793.


Example 82
Synthesis of 6′-(4-fluoro-phenyl)-4′-(4-methanesulfonyl-phenyl)-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4-ol (B55)



embedded image


To 6′-chloro-4′-(4-methanesulfonyl-phenyl)-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4-ol (0.2197 g, 0.6 mmol) and 4-fluorophenyl boronic acid (0.1040 g, 0.72 mmol) dissolved in acetonitrile (4 mL) and THF (6 mL) and Na2CO3 (10 mL, 0.4M) was added palladium (0) tetrakis(triphenylphosphine) (0.0352 g, 0.03 mmol). The reaction mixture was stirred and refluxed for 12-18 hours under N2. The reaction mixture was diluted with CH2Cl2 and filtered through Celite™, then rinsed with CH2Cl2. The filtrate was washed two times with water and one time with brine. The organic phase was collected and dried over potassium carbonate. The filtered sample was concentrated, and the resulting solid was dried for 12-18 hours under vacuum. Biotage Horizon HPFC system chromatography (SiO2, 98:2 CH2Cl2:MeOH) yielded a yellow solid (235 mg, 92%).


M.P.: 111° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4(0.01M, pH 3.2):CH3CN], 264 nm, Rt 6.1 min, 97.3% purity;



1H NMR: (600 MHz, CDCl3, TMS, 55° C.): δ 1.61-1.67 (m, 3H), 2.01-2.04 (m, 2H), 3.09 (s, 3H), 3.97 (septet, J=3.6 Hz, 1H), 3.28-3.32 (m, 2H), 4.24 (dt, J=4.2, 13.8 Hz, 2H), 6.75 (dt, J=4.2, 13.8 Hz, 2H), 7.11-7.14 (m, 2H), 7.15 (s, 1H), 7.8 (d, J=9.0 Hz, 2H) 8.01-8.04 (m, 4H).


Mass Spec: (TOF MS ES+): m/z 427 (M+H, 100); HRMS (TOF MS ES+) calcd for C23H23FN2O3S, [M+H] 427.1491, found 427.1497.


Examples 83
Synthesis of 1-[4-(4-fluoro-phenyl)-6-(4-methanesulfonyl-phenyl)-pyridin-2-yl]-4-methyl-piperazine (B41)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 82.


M.P. 151° C.


HPLC: Inertsil ODS-3V C18, 40:10:50 [KH2PO4 (0.01M, pH 3.2):CH3CN:MeOH], 264 nm, Rt 3.2 min, 97.1% purity.



1H NMR: (600 MHz, CDCl3, TMS, 55° C.): δ 2.39 (s, 3H), 2.59 (t, J=4.8 Hz, 4H), 3.09 (s, 3H), 3.75 (t, J=4.8 Hz, 4H), 6.81 (s, 1H), 7.18 (apt t, J=9.0 Hz, 2H), 7.3 (s, 1H), 7.61-7.63 (m, 2H), 8.02 (d, J=9.0 Hz, 2H), 8.24 (d, J=9.0 Hz, 2H).


Mass Spec: HRMS (TOF MS ES+) calcd for C23H24FN3O2S3, [M+H] 426.1651, found 426.1648.


Example 84
Synthesis of 1-[6-(4-fluoro-phenyl)-4-(4-methanesulfonyl-phenyl)-pyridin-2-yl]-4-methyl-piperazine (B42)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 82.


M.P.: 202° C.


HPLC: Inertsil ODS-3V C18, 40:10:50 [KH2PO4 (0.01M, pH 3.2):CH3CN:MeOH], 264 nm, Rt 3.3 min, 99.4% purity.



1H NMR: (600 MHz, CDCl3, TMS, 55° C.): δ 2.37(s, 3H), 2.57 (apt t, J=6.0 Hz, 4H), 3.09 (s, 3H), 3.73 (apt t, J=4.2 Hz, 4H), 6.73 (br s, 1H), 7.11-7.14 (m, 2H), 7.20 (br s, 1H), 7.80-7.81 (m, 2H), 8.02-8.04 (m, 4H).


Mass Spec: (TOF MS ES+): m/z 426 (M+H, 100); HRMS (TOF MS ES+) calcd for C23H24FN3O2S, [M+H] 426.1651, found 426.1662.


Example 85
Synthesis of {4-[2-(4-fluoro-phenyl)-6-(4-methyl-piperazin-1-yl)-pyridin-4-yl]-phenyl}-morpholin-4-yl-methanone (B43)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 82.


M.P.: 151° C.


HPLC: Inertsil ODS-3V C18, 40:10:50 [KH2PO4 (0.01M, pH 3.2):CH3CN:MeOH], 264 nm, Rt 0.1 min, 98.6% purity.



1H NMR (600 MHz, CDCl3, TMS, 55° C.): δ 2.86 (s, 3H), 2.58 (apt t, J=5.4 Hz, 4H), 3.51 (br s, 2H), 3.66 (br s, 2H), 3.74 (apt t, J=4.8 Hz, 4H), 3.81 (br s, 4H), 6.75 (s, 1H), 7.14 (d, J=8.4 Hz, 2H), 7.23 (s, 1H), 7.52 (d, J=7.8 Hz, 2H), 7.69 (d, J=8.4 Hz, 2H), 8.05 (dd, J=8.1, 5.4 Hz, 2H).


Mass Spec: (TOF MS ES+): m/z 461 (M+H, 100); HRMS (TOF MS ES+) calcd for C27H29FN4O2, [M+H] 461.2353, found 461.2373.


Example 86
Synthesis of 1-[6-(4-fluoro-phenyl)-4-(4-trifluoromethoxy-phenyl)-pyridin-2-yl]-4-methyl-piperazine (B44)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 82.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 3.0 min, 95.7% purity.



1H NMR: (600 MHz, CDCl3, TMS, 55° C.): δ 2.63 (s, 3H), 2.81 (apt t, J=4.8 Hz, 4H), 3.92 (apt t, J=5.4 Hz, 4H), 6.89 (s, 1H), 7.07 (s, 1H), 7.59 (d, J=7.8 Hz, 2H), 7.85-7.87 (m, 2H).


Mass Spec: (TOF MS ES+): m/z 372 (M+H, 100).


Example 87
Synthesis of 4′,6′-bis-(4-fluoro-phenyl)-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4-ol (B54)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 82.


M.P.: 127° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 14.3 min, 97.2% purity.



1H NMR: (600 MHz, CDCl3, TMS, 55° C.): δ 1.49 (apt s, 1H), 1.61-1.67 (m, 2H), 2.01-2.04 (m, 2H), 3.25-3.29 (m, 2H), 3.95 (br s, 1H), 4.25 (dt, J=4.2, 13.2 Hz, 2H), 6.73 (s, 1H), 7.10-7.16 (m, 5H), 7.58-7.60 (m, 2H), 8.01-8.04 (m, 2H).


Mass Spec: (ES+): m/z 367 (M+H, 100); HRMS (TOF MS ES+) calcd for C22H20F2N2O, [M+H] 367.1622, found 367.1615.


Example 88
Synthesis of 2,4,6-tris-(4-fluoro-phenyl)-pyridine (B22)



embedded image


2,6-Dichloro-4-iodopyridine (0.13629 g, 0.5 mmol) and 4-fluorophenyl boronic acid (0.2098 g, 1.5 mmol) were dissolved in acetonitrile (20 mL) and Na2CO3 (20 mL, 0.4M) followed by addition of palladium (0) tetrakis(triphenylphosphine) (0.086 g, 0.075 mmol) to the mixture. The reaction mixture was allowed to stir and reflux for 12-18 hours. The reaction mixture was diluted with CH2Cl2 and filtered through Celite™, then rinsed with CH2Cl2. The filtrate was washed one time with water and one time with brine. The organic phase was collected and dried over potassium carbonate. The filtered sample was concentrated, and the resulting solid was dried for 12-18 hours under vacuum. Biotage Horizon HPFC system chromatography (SiO2, 90:10 hexanes:ethyl acetate) yielded a dark brown solid product (169 mg, 94%).


M.P.: 198° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 71.8 min, 96.7% purity.



1H NMR: (600 MHz, CDCl3, TMS, 55° C.): δ 7.17-7.25 (m, 6H), 7.68-7.71 (m, 2H), 7.68 (s, 2H), 8.14-8.17 (m, 4H).


Mass Spec: (EI+): m/z 361 (M+, 100).


Example 89
Synthesis of 6′-chloro-4′-(4-methanesulfonyl-phenyl)-3,4,5,6-tetrahydro-2H-[1,2′]-bipyridinyl-4-ol



embedded image


2,6-Dichloro-4-(4-methanesulfonyl-phenyl)-pyridine (0.3015 g, 1 mmol) and 4-hydroxypiperidine (0.1024 g, 1 mmol) were dissolved in DMF (5 mL) and potassium carbonate (0.1702 g, 1.2 mmol) was added. The reaction mixture was stirred and refluxed for 12-18 hours at 90° C. under N2. The reaction mixture was diluted with CH2Cl2. The filtrate was washed three times with water and one time with brine. The organic phase was collected and dried over potassium carbonate. The filtered sample was concentrated, and the resulting solid was dried for 12-18 hours under vacuum. Biotage Horizon HPFC system chromatography (SiO2, 93:6:1 CH2Cl2:MeOH:NH4OH) yielded a pale yellow solid (185 mg, 51%).


M.P.: 169° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 4.5 min, 96.5% purity.



1H NMR: (600 MHz, CDCl3, TMS, 55° C.): δ 1.53 (br s, 1H), 1.58-1.63 (m, 2H), 1.96-2.04 (m, 2H), 3.08 (s, 3H), 3.25-3.30 (m, 2H), 3.95-3.98 (m, 1H), 4.08 (dt, J=4.8, 9 Hz, 2H), 6.63 (d, J=1.2 Hz), 6.75 (d, J=1.2 Hz), 7.71-7.72 (m, 2H), 8.00-8.02 (m, 2H).


Mass Spec: (TOF MS ES+): m/z 367 (M+H, 100); HRMS (TOF MS ES+) calcd for C17H19ClN2O3S, [M+H] 367.0883, found 367.0883.


Example 90
Synthesis of 6′-chloro-4′-phenyl-3,4,5,6-tetrahydo-2H-[1,2′]bipyridinyl-4-ol (B53)



embedded image


To 2,6-dichloro-4-phenyl-pyridine (448.7 mg, 2 mmol) dissolved in N,N-dimethylformamide (10 mL) was added 4-hydroxypiperidine (202.7 mg, 2 mmol) and potassium carbonate (332.5 mg, 2.4 mmol). The resulting mixture is allowed to stir at 90° C. for 12-18 hours. The sample was diluted in dichloromethane, washed two times with water, and washed one time with brine. The organic phase was dried over sodium sulfate and concentrated by rotary evaporation. The resulting sample was dried for 12-18 hours under vacuum. Purification (Biotage Horizon HPFC chromatography system, SiO2, 50:50 hexanes:ethyl acetate) gave a beige solid product (377 mg, 65%).


M.P.: 82° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 8.8 min, 99.4% purity.



1H NMR (600 MHz, CDCl3, TMS): δ 1.59-1.63 (m, 2H), 1.97-2.00 (m, 2H), 3.22-3.26 (m, 2H), 3.95 (br s, 1H), 4.10 (apt dt, J=4.2, 9.0 Hz, 2H).


Mass Spec: (TOF MS ES+): m/z 289 (M+H).


Example 91
Synthesis of 1-[6-chloro-4-(4-fluoro-phenyl)-pyridin-2-yl]-4-methyl-piperazine



embedded image


A solution of 2,6-dichloro-4-(4-fluorophenyl)-pyridine (0.480 g, 2 mmol) and 1-methylpiperizine (0.22 mL, 2 mmol) was prepared in DMF (10 mL) and potassium carbonate (0.3420 g, 2.4 mmol) was added to this solution. The reaction mixture was stirred and refluxed for 12-18 hours at 90° C. under an inert atmosphere (N2). The reaction mixture was then diluted with CH2Cl2 and filtered. The filtrate was washed three times with water and one time with brine. The organic phase was collected and dried over potassium carbonate. This sample was filtered, the filtrate was concentrated, and the resulting solid was dried for 12-18 hours under vacuum. Biotage Horizon HPFC system chromatography (SiO2, 93:6:1 CH2Cl2:MeOH:NH4OH) yielded a pale green solid (173 mg, 29%).


M.P.: 60° C.


HPLC: Inertsil ODS-3V C18, 40:10:50 [KH2PO4 (0.01M, pH 3.2):CH3CN:MeOH], 264 nm, Rt 3.6 min, 98.9% purity.



1H NMR: (600 MHz, CDCl3, TMS, 55° C.): δ 2.33 (s, 3H), 2.5 (t, J=4.8 Hz, 4H), 3.61 (dt, J=5.4 Hz, 4H), 6.58 (s, 1H), 6.76 (s, 1H), 7.10-7.14 (m, 2H), 7.5-7.53 (m, 2H).


Mass Spec: (TOF MS ES+): m/z 306 (M+H, 100); HRMS (TOF MS ES+) calcd for C16H17ClFN3, [M+H] 306.1173, found 306.1165.


Example 92
Synthesis of 1]-[6-chloro-4-(4-methanesulfonyl-phenyl)-pyridin-2-yl]-4-methyl-piperazine (16)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 91.


M.P.: 125° C:


HPLC: Zorbax Eclipse C18, 30:70 [Formic acid (0.01M):CH3CN], 264 nm, Rt 0.9 min, 100% purity.



1H NMR: (600 MHz, CDCl3, TMS, 55° C.): δ 2.35 (s, 3H), 2.52 (apt t, J=5.4 Hz, 4H), 3.09 (s, 3H), 4.8 (apt t, J=4.8 Hz, 4H), 6.62 (s, 1H), 6.79 (s, 1H), 7.73 (d, J=8.4 Hz, 2H), 8.02 (d, J=7.8 Hz, 2H).


Mass Spec: (TOF MS ES+): m/z 366 (M+H, 100).


Example 93
Synthesis of {4-[2-chloro-6-(4-methyl-piperazin-1-yl)-pyridin-4-yl]-phenyl}-morpholin-4-yl-methanone (18)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 91.


M.P.: 137° C.


HPLC: Inertsil ODS-3V C18, 40:10:50 [KH2PO4 (0.01M, pH 3.2):CH3CN:MeOH], 264 nm, Rt 2.4 min, 99.8% purity;



1H NMR (600 MHz, CDCl3, TMS, 55° C.): δ 2.64 (s, 3H), 2.81 (t, J=5.4 Hz, 4H), 2.8-4.09 (br m, 12H), 6.92 (s, 1H), 7.09 (s, 1H), 7.78 (d, J=9 Hz, 2H), 7.89 (d, J=8.4 Hz, 2H).


Mass Spec: (TOF MS ES+): m/z 401 (M+H, 100).


Example 94
Synthesis of 1-[6-chloro-4-(4-trifluoromethoxy-phenyl)-pyridin-2-yl]-4-methyl-piperazine (20)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 91.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 3.0 min, 95.7% purity.



1H NMR: (600 MHz, CDCl3, TMS, 55° C.): δ 2.63 (s, 3H), 2.81 (apt t, J=4.8 Hz, 4H), 3.92 (apt t, J=5.4 Hz, 4H), 6.89 (s, 1H), 7.07 (s, 1H), 7.59 (d, J=7.8 Hz, 2H), 7.85-7.87 (m, 2H).


Mass Spec: (TOF MS ES+): m/z 372 (M+H, 100).


Example 95
Synthesis of 6′-chloro-4′-(4-fluoro-phenyl)-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4-ol (28)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 91.


M.P.: 145° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 8.5 min, 99.4% purity.



1H NMR: (600 MHz, CDCl3, TMS, 55° C.): δ 1.49 (apt s, 1H), 1.56-1.62 (m, 2H), 1.95-1.99 (m, 2H), 3.21-3.26 (m, 2H), 3.94 (septet, J=3.6 Hz, 1H), 4.08 (dt, J=4.8, 13.2 Hz, 2H), 6.60 (s, 1H), 6.73 (d, J=1.2 Hz, 1H), 7.10-7.14 (m, 2H), 7.49-7.53 (m, 2H).


Mass Spec: (ES+): m/z 307 (M+100).


Example 96
Synthesis of 1-[4,6-bis-(4-fluoro-phenyl)-pyridin-2-yl]-4-methyl-piperazine (B40)



embedded image


To 1-[6-chloro-4-(4-fluoro-phenyl)-pyridin-2-yl]-4-methyl-piperazine (0.157 g, 0.5 mmol) and 4-fluorophenyl boronic acid (0.0862 g, 0.6 mmol) dissolved in acetonitrile (5 mL) was added Na2CO3 (5 mL, 0.4M) followed by addition of palladium (0) tetrakis(triphenylphoshine) (0.0292 g, 0.025 mmol). This reaction was allowed to stir and reflux for 12-18 hours. The resulting mixture was diluted with CH2Cl2 and filtered through Celite™, then rinsed with CH2Cl2. The filtrate was washed one time with water and one time with brine. The organic phase was collected and dried over potassium carbonate. This sample was filtered, the filtrate was concentrated, and the resulting solid was dried for 12-18 hours under vacuum. Biotage Horizon HPFC system chromatography (SiO2, 93:6:1 CH2Cl2:MeOH:NH4OH) yielded a pale brown solid (125 mg, 67%).


M.P.: 102° C.


HPLC: Inertsil ODS-3V C18, 40:10:50 [KH2PO4 (0.01M, pH 3.2):CH3CN:MeOH], 264 nm, Rt 5.1 min, 96.5% purity.



1H NMR: (600 MHz, CDCl3, TMS, 55° C.): δ 2.38 (s, 3H), 2.58 (t, J=4.8 Hz, 4H), 3.73 (t, J=4.8 Hz, 4H), 6.72 (s, 1H), 7.11-7.18 (m, 4H), 7.21 (s, 1H), 7.6-7.63 (m, 2H), 8.03-8.06 (m, 2H).


Mass Spec: (TOF MS ES+): m/z 366 (M+H, 100); HRMS (TOF MS ES+) calcd for C22H21F2N3, [M+H] 366.1782, found 366.1793.


Example 97
Synthesis of 4-(3-fluoro-4-methoxy-phenyl)-pyridine (B1)



embedded image


4-Iodopyridine (0.1031 g, 0.5 mmol) and 3-fluoro-4-methoxy phenyl boronic acid were (0.0852 g, 0.5 mmol) were dissolved in acetonitrile (5 mL) and Na2CO3 (5 mL, 0.4M) and then Pd(PPh3)4 (0.0291 g, 0.025 mmol) were added. The reaction mixture was stirred at reflux for 2.5 hours. The resulting suspension was filtered, and the filtrate was concentrated to about half the original volume. The precipitate was collected and washed with CH2Cl2 and water. The organic phase was collected and dried over potassium carbonate. The filtered sample was concentrated, and the resulting solid was dried for 12-18 hours under vacuum. Purification (Biotage Horizon HPFC chromatography system, SiO2, 50:50 hexanes:ethyl acetate) yielded a light yellow solid (25 mg, 20%).


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 4.0 min, 99.4% purity.


M.P.: 57° C.



1H NMR (600 MHz, CDCl3, 55° C.): δ 3.95 (s, 3H), 7.06 (t, J=8.4 Hz, 1H), 7.38-7.40 (m, 2H), 7.43-7.44 (m, 2H), 8.63 (d, J=5.6 Hz, 2H).


Mass Spec: m/z (EI) 203 (M+, 100).


Example 98
Synthesis of 2,6-bis-(3-fluoro-4-methoxy-phenyl)-pyridine (B12)



embedded image


To 2,6-dibromopyridine (0.2405 g, 1.0 mmol) and 3-fluoro-4-methoxy phenyl boronic acid (0.3392 g, 2.0 mmol) dissolved in dimethoxy ethane (15 mL) and 2 M sodium carbonate (5 mL) was added tetrakis(triphenylphosphine)palladium(0) (0.0995 g, 0.086 mmol). The reaction mixture was stirred at reflux for 12-18 hours under nitrogen. The reaction was cooled, the reaction volatiles were evaporated, and dichloromethane and water were added to the residue. The organic extract layer was collected and dried over potassium carbonate, filtered, and the filtrate concentrated under vacuum. The resulting solid was collected and dried for 12-18 hours under vacuum. Purification (Biotage Horizon HPFC chromatography system, SiO2, 70:30 hexanes:ethyl acetate) yielded a yellow solid (310 mg, 93%); HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 20.7 min, 98.0% purity; to give the product as a yellow solid (yield, 93%).


M.P.: 108° C.



1H NMR (600 MHz, CDCl3, 55° C.): δ 3.95 (s, 6H), 7.05 (t, J=8.4 Hz, 2H), 7.56 (d, J=13.2 Hz, 2H), 7.74 (t, J=7.8 Hz, 1H), 7.84 (d, J=7.84 Hz, 2H), 7.92 (dd, J=13.2, 1.8 Hz, 2H).


Mass Spec: (EI) m/z: 327 (M+, 100).


Example 99
Synthesis of (6-chloro-pyridine-2-yl)-(4-fluoro-phenyl)-amine



embedded image


A solution of 2,6-dichloropyridine (2 g, 13.5 mmol) in toluene (24 mL) was prepared and 4-flouro-phenylamine (1.43 mL, 14.8 mmol) and potassium tert-butoxide (6.05 g, 54 mmol) were added to the mixture. This reaction mixture was subjected to microwave radiation at 1000 W for 1 minute, after which the mixture was cooled to room temperature. The reaction product was extracted into the ethyl acetate, which was washed with water. The organic layer was dried over sodium sulphate, filtered, and concentrated under vacuum. The product was purified by column chromatography using 230-400 mesh silica gel, eluting with 6:94 ethyl acetate:petroleum ether, to afford the desired compound as light yellow solid (1.24 g, yield, 41.6%).



1H NMR (200 MHz, DMSO-d6): δ 9.33 (s, 1H), 7.58 (m, 3H), 6.77 (d, 2H, J=7.2 Hz), 6.75 (d, 2H, J=8.0 Hz).


Mass Spec: (ES-MS) m/z: 223 (M++1, 100%).


IR (neat) cm−1: 3258, 3173, 3063, 1595, 1418, 1214, 1160, 1096, 787.


Example 100
Synthesis of (4-fluoro-phenyl)-[6-trifluoromethoxy-phenyl)-pyridin-2-yl]-amine (B70)



embedded image


To a solution of (6-chloro-pyridine-2-yl)-(4-fluoro-phenyl)amine (2 g, 9 mmol) in 1,4-dioxane (25 mL) was added potassium carbonate (4.98 g, 36 mol) and tetrakis (triphenylphosphine)palladium(0) (11.6 mg, 0.45 mmol) followed by 4-trifluoro methoxy phenylboronicacid (2.04 g, 9.0 mmol). This reaction mixture was refluxed for 12 hours, after which water added to the mixture (200 mL) and the product was extracted with ethyl acetate. The organic layer was dried over sodium sulphate, filtered, and the filtrate was concentrated under vacuum. The resulting product was purified through column chromatography using 230-400 mesh silica gel, eluting with 20:80 ethyl acetate:DCM, to afford the desired compound as a light yellow solid (1.78 g, yield 56.7%, purity 98.62%).


M.P.: 72-74° C.



1H NMR(400 MHz, DMSO-d6): δ 9.16 (s, 1H), 8.15 (d, 2H, J=6.8, 2.1 Hz due to flourocoupling), 7.75 (d, 2H, J=7.9, 2.1 Hz due to fluorocoupling), 7.74 (t, 1H, J=3.0 Hz), 7.3 (d, 1H, J=6.98 Hz), 7.4 (d, 1H, J=8.8 Hz), 7.12 (d, 2H, J=4.9, 2.4 Hz due to fluoro coupling), 6.83 (d, 2H, J=6.7, 2.4 Hz due to fluoro coupling).


Mass Spec: CI-MS m/z: 349 (M++1, 100%).


IR (neat) cm−1: 3417, 2927, 1578, 1509, 1455, 1220, 1165, 1016, 829, 790.


Example 101
Synthesis of 2-chloro-4-(3-fluoro-4-methoxy-phenyl)-pyridine (B2)



embedded image


2-Chloro-4-iodopyridine (2.392 g, 10 mmol) and 3-fluoro-4-methoxy phenyl boronic acid (1.710 g, 10 mmol) were dissolved in acetonitrile (60 mL) and 0.4 M sodium carbonate (60 mL), followed by the addition of tetrakis(triphenylphosphine)palladium(0) (0.577 g, 5.0 mmol). The reaction mixture was stirred at reflux for 2.5 hour. The resulting suspension was filtered and the filtrate was concentrated to about half its original volume. The precipitate that formed was collected and washed with dichloromethane and water. The organic phase was collected and dried over potassium carbonate. The filtered sample was concentrated, and the resulting solid was dried for 12-18 hours under vacuum. Purification (Biotage Horizon HPFC chromatography system, SiO2, 80:20 hexanes:ethyl acetate) yielding an off-white solid (2.03 g, 86%); HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 7.9 min, 99% purity.


M.P.: 104° C.



1H NMR (600 MHz, CDCl3): δ 3.94 (s, 3H), 7.05 (t, J=8.4 Hz, 1H), 7.33-7.36 (m, 3H), 7.46 (s, 1H), 8.38 (d, J=4.8 Hz, 1H).


Mass Spec: m/z (EI) 237(M+, 100).


Example 102
Synthesis of 2-chloro-4-phenyl-pyridine (B4)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 101. The filtered sample was concentrated, and the resulting solid was dried overnight under vacuum. Biotage Horizon HPFC chromatography system (SiO2, 80:20 hexanes:ethyl acetate) yielded an off-white solid (260 mg, 63%); HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 8.7 min, 99.9% purity.


M.P.: 61° C.



1H NMR (600 MHz, CDCl3, 55° C.): δ 7.41 (dd, J=4.8, 0.6 Hz, 1H), 7.44-7.49 (m, 3H), 7.53 (s, 1H), 7.60 (d, J=7.2 Hz, 2H), 8.42 (d, J=5.4 Hz, 1H).


Mass Spec: m/z (EI): 189 (M+, 100).


Example 103
Synthesis of 2-chloro-4-(3,4-difluoro-phenyl)-pyridine (B6)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 101. Purification (Biotage Horizon HPFC chromatography system, SiO2, 80:20 hexanes:ethyl acetate) yielded a light yellow solid (94 mg, 82%); HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 8.9 min, 98.9% purity.


M.P.: 167° C.



1H NMR (600 MHz, CDCl3): δ 7.25-7.30 (m, 1H), 7.33-7.35 (m, 2H), 7.40-7.43 (m, 1H), 7.46 (apt s, 1H), 8.43 (d, J=5.4 Hz, 1H).


Mass Spec: EI (m/z) 225 (M+, 100).


Example 104
Synthesis of 2-chloro-4-(4-methanesulfonyl-phenyl)-pyridine (B8)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 101. Purification (Biotage Horizon HPFC chromatography system, SiO2, 50:50 hexanes:ethyl acetate) yielded a off white solid (79 mg, 58%); HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 4.4 min, 99.8% purity.


M.P.: 112° C.



1H NMR (600 MHz, CDCl3): δ 3.09 (s, 3H), 7.43 (dd, J=5.4, 1.8 Hz, 1H), 7.55 (apt s, 1H), 7.78-7.80 (m, 2H), 8.06-8.07 (m, 2H), 8.50 (d, J=5.4 Hz, 1H).


Mass Spec: EI (m/z) 267 (M+, 100).


Example 105
Synthesis of 2-chloro-4-(3-methanesulfonyl-phenyl)-pyridine (B9)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 101. The filtered sample was concentrated, and the resulting solid was dried overnight under vacuum. Purification (Biotage Horizon HPFC chromatography system, SiO2, 50:50 hexanes:ethyl acetate) yielded a light brown solid (72 mg, 53%); HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 4.5 min, 99.7% purity.


M.P.: 130° C.



1H NMR (600 MHz, CDCl3, 55° C.): δ 3.00 (s, 3H), 7.44 (dd, J=5.4, 1.2 Hz, 1H), 7.56 (app d, J=0.6 Hz, 1H), 7.71 (t, J=7.2 Hz, 1H), 7.86-7.88 (m, 1H), 8.02-8.04 (m, 1H), 8.171-8.177 (m, 1H), 8.48 (d, J=5.4 Hz, 1H).


Mass Spec: EI (m/z): 267 (M+, 100).


Example 106
Synthesis of 4-benzo[1,3]dioxol-5-yl-2-chloro-pyridine (B10)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 101. Purification by column chromatography (Biotage Horizon HPFC system, SiO2, 80:20 hexanes:ethyl acetate) gave a colorless solid (0.275 g, 78%); HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 7.475 min, 99.86% purity.


M.P.: 148-149° C.



1H NMR (600 MHz, CDCl3): δ 6.03 (s, 2H), 6.9 (d, J=7.8 Hz, 1H), 7.07 (d, J=1.2 Hz, 1H), 7.10 (dd, J=7.8, 1.8 Hz, 1H), 7.33 (dd, J=5.4, 1.2 Hz, 1H), 7.45 (d, J=1.2 Hz, 1H), 8.37 (d, J=4.8 Hz, 1H).


Mass Spec: EI (m/z) 233 (M+H, 100), 232 (64), 234(40), 235(42), 140 (23), 113 (11), 99(23).


Example 107
Synthesis of 2-chloro-4-(4-trifluoromethoxy-phenyl)-pyridine (3)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 101. Biotage Horizon HPFC system chromatography (SiO2, 80:20 hexanes:ethyl acetate) yielded a pale yellow solid (1.02 g, 74%).


M.P.: 44° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 12.8 min, 99.9% purity.



1H NMR (600 MHz, CDCl3, TMS, 55° C.): δ 7.33 (d, J=7.8 Hz, 2H), 7.39 (dd, J=4.8, 1.8 Hz, 1H), 7.5 (apt d, J=1.2 Hz, 1H), 7.61-7.64 (m, 2H), 8.44 (d, J=5.4 Hz, 1H).


Mass: (EI+): m/z 273 (M+, 100).


Example 108
Synthesis of 4-(3-fluoro-4-methoxy-phenyl)-2-phenyl-pyridine (B3)



embedded image


2-Chloro-4-(3-chloro-4-methoxy-phenyl)-pyridine (0.118 g, 0.5 mmol) and phenyl boronic acid (0.0655 g, 0.5 mmol) were dissolved in acetonitrile (5 mL), and 0.4M sodium carbonate (5 mL) and tetrakis(triphenylphosphine)palladium(0) (0.0288 g, 0.025 mmol) were added thereto. The reaction mixture was stirred at reflux for 12-18 hours. The resulting suspension was filtered and the filtrate was concentrated to about half its original volume. The precipitate was collected and washed with dichloromethane and water, and the organic phase was collected and dried over potassium carbonate. The filtered sample was then concentrated, and the resulting solid was dried for 12-18 hours under vacuum. Purification (Biotage Horizon HPFC chromatography system, SiO2, 80:20 hexanes:ethyl acetate) yielded an off-white solid (90 mg, 65%); HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 11.3 min, 98.9% purity; to give the product as an off-white solid (yield 65%).


M.P.: 84° C.



1H NMR (600 MHz, CDCl3, 55° C.): δ 3.95 (s, 3H), 7.07 (t, J=8.4 Hz, 1H), 7.37 (dd, J=5.4, 1.8 Hz, 1H), 7.41-7.45 (m, 3H), 7.48 (t, J=7.2 Hz, 2H), 7.85 (s, 1H), 8.04 (d, J=7.2 Hz, 2H), 8.70 (d, J=5.4 Hz, 1H).


Mass Spec: EI (m/z): 279 (M+, 100).


Example 109
Synthesis of 2-(3-fluoro-4-methoxy-phenyl)-4-phenyl-pyridine (B5)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 108. Purification (Biotage Horizon HPFC chromatography system, SiO2, 80:20 hexanes:ethyl acetate) yielded a white solid (95 mg, 53%); HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4(0.01M, pH 3.2):CH3CN], 264 nm, Rt 13.2 min, 98.9% purity.


M.P.: 58° C.



1H NMR (600 MHz, CDCl3, 55° C.): δ 3.95 (s, 3H), 7.06 (t, J=9 Hz, 1H), 7.41 (dd, J=5.4, 1.2 Hz, 1H), 7.44-7.46 (m, 1H), 7.49-5.51 (m, 2H), 7.67-7.68 (m, 2H), 7.80-7.82 (m, 1H), 7.83-7.86 (m, 2H), 8.70 (d, J=5.4 Hz, 1H).


Mass Spec: EI (m/z): 279 (M+, 100).


Example 110
Synthesis of 4-(3,4-difluoro-phenyl)-2-phenyl-pyridine (B7)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 108. Purification (Biotage Horizon HPFC chromatography system, SiO2, 100% CH2 Cl2) yielded a white solid (42 mg, 51%); HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 14.3 min, 99.7% purity.


M.P.: 92° C.



1H NMR (600 MHz, CDCl3): δ 7.27-7.31 (m, 1H), 7.37 (dd, J=5.4, 2.4 Hz, 1H), 7.40-7.45 (m, 2H), 7.48-7.51 (m, 3H), 7.841-7.843 (m, 1H), 8.02-8.03 (m, 2H), 8.74 (d, J=4.8 Hz, 1H).


Mass Spec: EI (m/z): 267 (M+, 100).


Example 111
Synthesis of 4-benzo[1,3]dioxol-5-yl-2-phenyl-pyridine (B11)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 108. Purification by column chromatography (Biotage Horizon HPFC system, SiO2, 80:20 hexanes:ethyl acetate) gave a colorless solid (0.110 g, 62.5%); HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 10.2 min, 97.72% purity.


M.P.: 87-88° C.



1H NMR (600 MHz, CDCl3): δ 6.04 (s, 2H), 6.94 (d, J=7.8 Hz, 1H), 7.17 (apt d, J=1.8 Hz, 1H), 7.20 (dd, J=8.4, 2.4 Hz, 1H), 7.37 (dd, J=7.1, 1.8 Hz, 1H), 7.42-7.46 (m, 1H, 7.49 (t, J=10.2 Hz, 2H), 7.85 (s, 1H), 8.03 (d, J=7.2 Hz, 2H), 8.7 (d, J=5.4 Hz, 1H).


Mass Spec: EI (m/z): 275 (M+, 100).


Example 112
Synthesis of (3-chloro-4-methoxy-phenyl)-(2-chloro-pyridin-4-yl)-amine



embedded image


To 2-chloro-4-iodo pyridine (1.4429 g, 6 mmol) and 3-chloro-p-anisidine (1.13 g, 7.2 mmol) dissolved in dry toluene (20 mL) were added tris(dibenzylideneacetone)dipalladium (0) (0.1095 g, 0.12 mmol), 1,3-bis(diphenylphospino)propane (0.0991 g, 0.24 mmol), and sodium-tert-butoxide (0.8093 g, 8.4 mmol). The reaction mixture was stirred at reflux for 12-18 hours under N2. The reaction mixture was diluted with CH2Cl2 and filtered through Celite™, then rinsed with CH2Cl2. The filtrate was washed two times with water and one time with brine. The organic phase was collected and dried over potassium carbonate. The filtered sample was concentrated, and the resulting solid was dried for 12-18 hours under vacuum. Biotage Horizon HPFC system chromatography (SiO2, 50:50 hexanes:ethyl acetate) yielded a brown solid (780 mg, 48%).


M.P.: 135° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 6.2 min, 99.2% purity.



1H NMR: (300 MHz, CDCl3, TMS,): δ 3.92 (s, 3H), 6.02 (s, 1H), 6.57 (dd, J=2.1, 5.7 Hz, 1H), 6.65 (d, J=2.1 Hz, 1H), 6.95 (d, J=8.7 Hz, 1H), 7.09 (dd, J=2.4, 6.3 Hz, 1H), 7.25 (t, J=2.7 Hz, 1H), 8.01 (t, J=5.7 Hz, 1H).


Mass Spec: LC-MSD (ES+): m/z 269 (M+H, 100).


Example 113
Synthesis of (2-chloro-pyridin-4-yl)-(4-fluoro-3-methoxy-phenyl)-amine (B18)



embedded image


2-Chloro-4-iodopyridine (340.4 mg, 1.42 mmol) was dissolved in anhydrous toluene (15 mL), after which 4-fluoro-3-methoxyaniline (241.4 mg, 1.7 mmol), tris(dibenzylidineacetone)dipalladium(0) (26.5 mg, 0.028 mmol), 1,3-bis(diphenylphosphino)propane (23 mg, 0.06 mmol), and sodium-tert-butoxide (191.9 mg, 2.0 mmol) were added to the solution. This reaction mixture was stirred at reflux for 12-18 hours, after which the reaction was diluted in dichloromethane and filtered through Celite™. The Celite™ was washed with dichloromethane, and the resulting solution was concentrated to afford the product. This solid was collected and dried for 12-18 hours under vacuum. Purification (Biotage Horizon HPFC chromatography system, SiO2, 90:9:1 dichloromethane:methanol:ammonium hydroxide) yielded a light brown solid (18, 169 mg, 47%); HPLC: Inertsil ODS-3V C18, 30:70[KH2PO4 (0.01M, pH 3.2): CH3CN], 264 nm, Rt 5.2 min, 93.6% purity.


M.P.: 89° C.



1H NMR (600 MHz, CDCl3): δ 3.68 (s, 3H), 6.08 (s, 1H), 6.60 (dd, J=5.6, 2.4 Hz, 1H), 6.69 (d, J=1.8 Hz, 1H), 6.71 (dt, J=8.4, 3.6 Hz, 1H), 6.78 (dd, J=7.8, 1.8 Hz, 1H) 7.08 (dd, J=10.8, 8.4 Hz, 1H), 8.01 (d, J=6 Hz, 1H).


Mass Spec: ES (m/z): 256 (45.9), 255 (24.4), 253 (M+H, 100).


Example 114
Synthesis of (2-chloro-pyridin-4-yl)-(3-fluoro-4-methoxy-phenyl)-amine (30)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 113.


Example 115
Synthesis of 3-chloro-4-methoxy-phenyl)-(2-phenyl-pyridin-4-yl)-amine (B19)



embedded image


To (3-chloro-4-methoxy-phenyl)-(2-chloro-pyridin-4-yl)-amine (0.3241 g, 1.2 mmol) and phenyl boronic acid (0.1762 g, 1.44 mmol) dissolved in acetonitrile (15 mL) and Na2CO3 (15 mL, 0.4M) was added palladium (0) tetrakis(triphenylphosphine) (0.0699 g, 0.06 mmol). The reaction mixture was stirred and refluxed for 12-18 hours under N2. The reaction was monitored by TLC; then additional palladium (0) tetrakis(triphenylphosphate) (0.0699 g, 0.06 mmol) was added and refluxed for 12-18 hours. The reaction mixture was diluted with CH2Cl2 and filtered through Celite™, then rinsed with CH2Cl2. The filtrate was washed one time with water and one time with brine. The organic phase was collected and dried over potassium carbonate. The filtered sample was concentrated, and the resulting solid was dried for 12-18 hours under vacuum. Biotage Horizon HPFC system chromatography (SiO2, 50:50 hexanes:ethyl acetate) yielded a white solid product (270 mg, 72% yield).


M.P.: 160° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4(0.01M, pH 3.2):CH3CN], 264 nm, Rt 3.3 min, 97.4% purity.



1H NMR: (300 MHz, CDCl3, TMS,): δ 3.92 (s, 3H), 5.99 (s, 1H), 4.43 (dd, J=2.0, 5.7 Hz, 1H), 6.95(d, J=8.7 Hz, 1H), 7.08-7.15 (m, 2H), 7.29 (dd, J=2.7 Hz, 1H), 7.38-7.43 (m, 3H), 7.86-7.90 (m, 2H), 8.36 (d, J=5.7 Hz, 1H).


Mass Spec: HRMS (TOF MS ES+), calcd for C18H15ClN2O [M+H] 311.0951, found 311.0948.


Example 116
Synthesis of N2-cyclohexylmethyl-N4-(3-fluoro-4-methoxy-phenyl)-pyridine-2,4-diamine (B72)



embedded image


In a dry round bottom flask, tris(dibenzylideneacetone)dipalladium (18.7 mg, 0.16 mmol), sodium-tert-butoxide (144.0 mg, 1.5 mmol) and (2-chloro-pyridin-4-yl)-(3-fluoro-4-methoxy-phenyl)-amine (238.8 mg, 0.95 mmol) were dissolved in 10 mL of anhydrous toluene. Nitrogen was blown over the mixture for about 10 minutes. The ligand, 2,8,9-triisobutyl-2,5,8,9-tetraaza-1-phosphabicyclo-[3.3.3]undecane (0.03 mL, 0.08 mmol) and cyclohexanemethylamine (0.16 mL, 1.3 mmol) were dissolved in 3 mL of dry toluene. The ligand/amine mixture was added to the reaction flask, and the resulting mixture was allowed to stir at reflux for 12-18 hours. The reaction mixture was diluted with dichloromethane and filtered through Celite™. The Celite™ was washed with dichloromethane and the sample was concentrated by rotary evaporation. The resulting solid was dried for 12-18 hours under vacuum. Flash column chromatography (SiO2, 50:50 hexanes:ethyl acetate) gave a light yellow solid (94 mg, 30%).


M.P.: 58° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 um, Rt 3.5 min, 98.8% purity.



1H NMR (600 MHz, CDCl3, TMS): δ 0.96-1.03 (m, 2H), 1.15-1.26 (m, 3H), 1.57-1.83 (m, 6H), 3.14 (t, J=6.0 Hz, 2H), 3.92 (s, 3H), 4.65 (br s, 1H), 6.45 (s, 1H), 6.70 (d, J=5.4 Hz, 1H), 7.00-7.03 (m, 1H), 7.29-7.34 (m, 2H), 8.07 (d, J=5.4 Hz, 1H).


Mass Spec: (TOF MS ES+): m/z 316 (44.3), 315 (M+H, 100).


Example 117
Synthesis of (3-fluoro-4-methoxy-phenyl)-(4-phenyl-pyridin-2-yl)-amine



embedded image


To a dry 50 mL round bottomed flask were added tris(dibenzylidineacetone)dipalladium(0) (0.019 g, 0.021 mmol) and sodium-tert-butoxide (0.151 g, 1.57 mmol), under a nitrogen atmosphere. 2,8,9-Triisobutyl-2,5,8,9-tetraaza-1-phospha-bicyclo[3.3.3]undecane (0.028 g, 0.084 mmol), 2-chloro-4-phenyl pyridine (0.2 g, 1.05 mmol), and 3-fluoro-p-anisidine (0.177 g, 1.26 mmol) were added to the reaction consecutively, followed by dry toluene (8 mL). This reaction mixture was heated at 80° C. for 15 hours, and then at reflux for an additional 22 hours. The crude reaction mixture was filtered through Celite™, which was washed with toluene. The resulting organic layer was concentrated to dryness and the product was purified by column chromatography (Biotage Horizon HPFC system, SiO2, 80:20 hexanes:ethyl acetate) gave a pale brown solid (0.142 g, 46%); HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 8.3 min, 99.3% purity.


M.P.: 139-140° C.



1H NMR (600 MHz, CDCl3): δ 3.89 (s, 3H), 6.61 (s, 1H), 6.91 (s, 1H), 6.94-6.96 (m, 2H), 7.05 (apt d, J=9 Hz, 1H), 7.26 (dd, J=7.2, 3 Hz, 1H), 7.41-7.46 (m, 3H), 7.56 (d, J=6.6 Hz, 2H), 8.23 (d, J=5.4 Hz, 1H).


Mass Spec: ES (m/z): 295 (M+H, 100).


Example 118
Synthesis of cycloheptyl-[4-(3-fluoro-4-methoxy-phenyl)-pyridin-2-yl]-amine (B14)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 117. Purification (Biotage Horizon HPFC chromatography system, SiO2, 50:50 hexanes:ethyl acetate) yielded an off-white solid (203.6 mg, 65%); HPLC: Inertsil ODS-3V C18, 30:70[KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 3.2 min, 97.5% purity.


M.P.: 94° C.



1H NMR (600 MHz, CDCl3, 55° C.): δ 1.54-1.72 (m, 10H), 2.03-2.07 (m, 2H), 3.82-3.85 (m, 1H), 3.94 (s, 3H), 6.43 (s, 1H), 6.69 (dd, J=5.4, 1.2 Hz, 1H), 7.03 (t, J=8.4 Hz, 1H), 7.31-7.34 (m, 2H), 8.09 (d, J=5.4 Hz, 1H).


Mass Spec: ES (m/z): 316 (28.2), 315 (M+H, 100).


Example 119
Synthesis of 4-[4-(3-fluoro-4-methoxy-phenyl)-pyridin-2-yl]-morpholine (B15)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 117. Purification (Biotage Horizon HPFC chromatography system, SiO2, 96:3:1 dichloromethane:methanol:ammonium hydroxide) yielded a mustard yellow solid (206 mg, 71%); HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 5.0 min, 98.9% purity.


M.P.: 92° C.



1H NMR (600 MHz, CDCl3): δ 3.54-3.56 (m, 4H), 3.83-3.85 (m, 4H), 3.92 (s, 3H), 6.72 (s, 1H), 6.82 (dd, J=4.8, 1.2 Hz, 1H), 7.02 (t, J=7.8 Hz, 1H), 7.31-7.35 (m, 2H), 8.21 (d, J=5.4 Hz, 1H).


Mass Spec: ES (m/z): 290 (20.9), 289 (M+H, 100).


Example 120
Synthesis of cyclohexylmethyl-[4-(3-fluoro-4-methoxy-phenyl)-pyridin-2-yl]-amine (B16)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 117. The Celite™ was washed with dichloromethane, and the sample was concentrated and dried overnight under vacuum. Flash column chromatography (SiO2, 50:50 hexanes:ethyl acetate) yielded an off-white solid (94 mg, 30%); HPLC: Inertsil ODS-3V C18, 30:70[KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 3.5 min, 98.8% purity.


M.P.: 62° C.



1H NMR (600 MHz, CDCl3): δ 0.96-1.03 (m, 1H), 1.13-1.28 (m, 2H), 1.55-1.83 (m, 7H), 3.14 (t, J=6.6 Hz, 2H), 3.92 (s, 3H), 4.65 (br s, 1H), 6.46 (s, 1H), 6.70 (d, J=5.4 Hz, 1H), 7.01 (t, J=7.8 Hz, 1H), 7.31-7.34 (m, 2H), 8.07 (d, J=4.8H, 1H).


Mass Spec: ES (m/z): 316 (44.3), 315 (M+H, 100).


Example 121
Synthesis of [4-(3-fluoro-4-methoxy-phenyl)-pyridin-2-yl]-(4-fluoro-phenyl)-amine (B17)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 117. Purification (Biotage Horizon HPFC chromatography system, SiO2, 50:50 hexanes:ethyl acetate) yielded a yellow solid (89 mg, 57%); HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 8.1 min, 99.5% purity;


M.P.: 122° C.



1H NMR (600 MHz, CDCl3, TMS): δ 3.91 (s, 3H), 6.55 (s, 1H), 6.83 (s, 1H), 6.88 (d, J=5.4 Hz, 1H), 6.99-7.06 (m, 3H), 8.19 (d, J=5.4 Hz, 1H).


Mass Spec: ES (m/z): 314 (27.7), 313 (M+H, 100).


Example 122
Synthesis of (4-fluoro-phenyl)-[4-(4-trifluoromethoxy-phenyl)-pyridin-2-yl]-amine (B20)



embedded image


This compound was prepared by a procedure analogous to that disclosed in Example 117. Biotage Horizon HPFC system chromatography (SiO2, 80:20 hexanes:ethyl acetate) yielded a light brown colored solid (410 mg, 78%).


M.P.: 105° C.


HPLC: Inertsil ODS-3V C18, 30:70 [KH2PO4 (0.01M, pH 3.2):CH3CN], 264 nm, Rt 17.2 min, 99.7% purity.



1H NMR (600 MHz, CDCl3, TMS, 55° C.): δ 6.7 (s, 1H), 6.85(s, 1H), 6.9 (dd, J=5.4, 1.8 Hz, 1H), 7.03-7.07 (m, 2H), 7.27 (d, J=7.8 Hz, 2H), 7.31-7.34 (m, 2H), 7.55-7.57 (m, 2H), 8.22 (d, J=5.4 Hz, 1H).


Mass Spec: (TOF MS ES+): m/z 349 (M+H, 100).


In another aspect of the present invention, this invention encompasses salts of the compounds disclosed herein, including pharmaceutically acceptable and non-pharmaceutically acceptable salts. It is envisioned that the compounds, compositions, and all the salts disclosed therein, including the non-pharmaceutically acceptable salts, can have uses and applications beyond pharmaceutical applications. For example, the pyrimidine compounds and compositions comprising pryimidine compounds of this invention can be used in a variety of agricultural uses or applications such as herbicides and pesticides, hardness stabilizers in rubber processing, ultraviolet light absorbers, and other uses.


The foregoing description has been presented for purposes of illustration and description. It is not intended to be exhaustive or to limit the invention to the precise examples or embodiments disclosed. Obvious modifications or variations are possible in light of the above teachings. The embodiment or embodiments discussed were chosen and described to provide the best illustration of the principles of the invention and its practical application to enable one of ordinary skill in the art to utilize the invention in various embodiments and with various modifications as are suited to the particular use contemplated. All such modifications and variations are within the scope of the invention as determined by the appended claims when interpreted in accordance with the breadth to which they are fairly and legally entitled.

Claims
  • 1. A compound having the formula:
  • 2. A compound according to claim 1, having the formula:
  • 3. A compound according to claim 1, having the formula:
  • 4. A compound according to claim 1, having the formula:
  • 5. A compound according to claim 4, wherein: R4 is selected from wherein X is selected from CH2, O, NH, NMe, NEt, S, SO2, CH(OCOCH2CH2CO2H), or CH(OH); n and m are independently an integer from 0 to 2, inclusive; and R11 and R12, in each occurrence, are selected independently from OCF3, OMe, Cl, F, SO2Me, CF3, Me, COMe, CONHMe, NHSO2Me, SO2NH2, SO2NHMe, SO2NMe2, CONH2, CONMe2, CO2Me, —OCH2O—, or OH.
  • 6. A compound according to claim 1, having the formula:
  • 7. A compound according to claim 1, having the formula:
  • 8. A compound according to claim 7, wherein: n, m and p are independently an integer from 0 to 2, inclusive; and R11, R12 and R13, in each occurrence, are selected independently from OCF3, OMe, Cl, F, SO2Me, CF3, Me, COMe, CONHMe, NHSO2Me, SO2NH2, SO2NHMe, SO2NMe2, CONH2, CONMe2, CO2Me, —OCH2O—, or OH.
  • 9. A compound according to claim 1, having the formula:
  • 10. A compound according to claim 1, having the formula:
  • 11. A compound according to claim 1, having the formula:
  • 12. A compound according to claim 1, having the formula:
  • 13. A compound according to claim 1, having the formula:
  • 14. A compound according to claim 13, wherein: R1 is selected from wherein X is selected from CH2, O, NH, NMe, NEt, S, SO2, CH(OCOCH2CH2CO2H), or CH(OH); n and m are independently an integer from 0 to 2, inclusive; and R11 and R12, in each occurrence, are selected independently from OCF3, OMe, Cl, F, SO2Me, CF3, Me, COMe, CONHMe, NHSO2Me, SO2NH2, SO2NHMe, SO2NMe2, CONH2, CONMe2, CO2Me, —OCH2O—, or OH.
  • 15. A compound according to claim 1, having the formula:
  • 16. A compound according to claim 15, wherein: R1 is selected from wherein X is selected from CH2, O, NH, NMe, NEt, S, SO2, CH(OCOCH2CH2CO2H), or CH(OH); n and m are independently an integer from 0 to 2, inclusive; and R11 and R12, in each occurrence, are selected independently from OCF3, OMe, Cl, F, SO2Me, CF3, Me, COMe, CONHMe, NHSO2Me, SO2NH2, SO2NHMe, SO2NMe2, CONH2, CONMe2, CO2Me, —OCH2O—, or OH.
  • 17. A compound according to claim 1, having the formula:
  • 18. A compound according to claim 17, wherein: R1 is selected from wherein X is selected from CH2, O, NH, NMe, NEt, S, SO2, CH(OCOCH2CH2CO2H), or CH(OH); n and m are independently an integer from 0 to 2, inclusive; and R11 and R12, in each occurrence, are selected independently from OCF3, OMe, Cl, F, SO2Me, CF3, Me, COMe, CONHMe, NHSO2Me, SO2NH2, SO2NHMe, SO2NMe2, CONH2, CONMe2, CO2Me, —OCH2O—, or OH.
  • 19. A compound according to claim 1, having the formula:
  • 20. A compound according to claim 19, wherein: n and m are independently an integer from 0 to 2, inclusive; and R11 and R12, in each occurrence, are selected independently from OCF3, OMe, Cl, F, SO2Me, CF3, Me, COMe, CONHMe, NHSO2Me, SO2NH2, SO2NHMe, SO2NMe2, CONH2, CONMe2, CO2Me, —OCH2O—, or OH.
  • 21. A compound having the formula:
  • 22. A compound selected from: 2,4,6-tris-(4-fluoro-phenyl)-pyridine; (2,6-diphenyl-pyridin-4-yl)-p-tolyl-amine; (2,6-diphenyl-pyridin-4-yl)-(4-trifluoromethoxy-phenyl)-amine; [2,6-bis-(3-fluoro-phenyl)-pyridin-4-yl]-(4-trifluoromethoxy-phenyl)-amine; [2,6-bis-(4-fluoro-phenyl)-pyridin-4-yl]-methyl-(4-trifluoromethoxy-phenyl)-amine; [2,6-bis-(4-methanesulfonyl-phenyl)-pyridin-4-yl]-(4-trifluoromethoxy-phenyl)-amine; [2,6-bis-(3-(methylsulfonyl)-phenyl)-pyridin-4-yl]-(4-trifluoromethoxy-phenyl)-amine; N-ethyl-3-[6-(3-methanesulfonyl-phenyl)-4-(4-trifluoromethoxy-phenylamino)-pyridin-2-yl]-benzamide; 1-{3-[6-(3-acetyl-phenyl)-4-(4-trifluoromethoxy-phenylamino)-pyridin-2-yl]-phenyl}-ethanone; 1-{4-[2,6-bis-(4-fluoro-phenyl)-pyridin-4-ylamino]-phenyl}-ethanone; [2,6-bis-(3-N,N-dimethyl-benzamide)-pyridin-2-yl]-(4-trifluoromethoxy-phenyl)-amine; 3-{2,6-bis-(4-fluoro-phenyl)-pyridin-4-ylamino]-benzenethiol; compound with acetic acid methyl ester; thiocarbonic acid O-methyl ester S-{3-[6-pyrrolidin-1-yl-4-(4-trifluoromethoxy-phenylamino)-pyridin-2-yl]-phenyl}ester; 1-{3-[6-pyrrolidin-1-yl-4-(4-trifluoromethoxy-phenylamino)-pyridin-2-yl]-phenyl}-ethanone; [2-(4-fluoro-phenyl)-6-(4-methyl-piperazin-1-yl)-pyridin-4-yl]-(4-trifluoro-methoxy-phenyl)-amine; 1-[4,6-bis-(4-fluoro-phenyl)-pyridin-2-yl]-4-methyl-piperazine; 1-[4-(4-fluoro-phenyl)-6-(4-methanesulfonyl-phenyl)-pyridin-2-yl]-4-methyl-piperazine; 1-[6-(4-fluoro-phenyl)-4-(4-methanesulfonyl-phenyl)-pyridin-2-yl]-4-methyl-piperazine; {4-[2-(4-fluoro-phenyl)-6-(4-methyl-piperazin-1-yl)-pyridin-4-yl]-phenyl}-morpholin-4-yl-methanone; 1-[6-(4-fluoro-phenyl)-4-(4-trifluoromethoxy-phenyl)-pyridin-2-yl]-4-methyl-piperazine; 1-[4-(2-morpholin-4-yl-6-phenyl-pyridin-4-ylamino)-phenyl]-ethanone; 6′-(4-fluoro-phenyl)-4′-(4-trifluoromethoxy-phenylamino)-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4-ol; 1-[4-(4-hydroxy-6′-phenyl-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4′-ylamino)-phenyl]-ethanone; 1-[3-(4-hydroxy-6′-phenyl-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4′-ylamino)-phenyl]-ethanone; [2-(4-fluoro-phenyl) -6-morpholin-4-yl -pyridin-4-yl]-(4-trifluoromethoxy-phenyl) -amine; [2-(3-methanesulfonyl-phenyl)-6-morpholin-4-yl-pyridin-4-yl]-(4-trifluoromethoxy-phenyl)-amine; 1-{3-[6-morpholin-4-yl-4-(4-trifluoromethoxy-phenylamino)-pyridin-2-yl]-phenyl}-ethanone; {3-[6-morpholin-4-yl-4-(4-trifluoromethoxy-phenylamino)-pyridin-2-yl]-phenyl}-pyrrolidin-1-yl-methanone; 4′,6′-bis-(4-fluoro-phenyl)-3,4,5,6-tetrahydro-2H-[1,240 ]bipyridinyl-4-ol; 6′-(4-fluoro-phenyl)-4′-(4-methanesulfonyl-phenyl)-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4-ol; [6′-(4-Fluoro-phenyl)-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-2′-yl]-(4trifluoromethoxy-phenyl)-amine; (4-Trifluoromethoxy-phenyl)-[6′-(4-trifluoromethoxy-phenyl)-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-2′-yl]-amine; (6′-Phenyl-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-2′-yl)-(4-trifluoro-methoxy-phenyl)-amine; [6′-(3-Methanesulfonyl-phenyl)-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-2′-yl]-(4-trifluoromethoxy-phenyl)-amine; 4-[6′-(4-Fluoro-phenyl)-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-2′-ylamino]-N-methyl-benzenesulfonamide; 1-[4-(4-Hydroxy-6′-phenyl-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-2′-ylamino)-phenyl]-ethanone; 2′,6′-Bis-(4-fluoro-phenyl)-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl; 2′,6′-Bis-(4-trifluoromethoxy-phenyl)-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl; (6-Phenyl-4-pyrrolidin-1-yl-pyridin-2-yl)-(4-trifluoromethoxy-phenyl)-amine; [6-(4-Fluoro-phenyl)-4-pyrrolidin-1-yl-pyridin-2-yl]-(4-trifluoromethoxy-phenyl)-amine; N-Methyl-4-[4-pyrrolidin-1-yl-6-(4-trifluoromethoxy-phenyl)-pyridin-2-ylamino]-benzenesulfonamide; N-Methyl-4-[4-(4-methyl-piperazin-1-yl)-6-(4-trifluoromethoxy-phenylamino)-pyridin-2-yl]-benzenesulfonamide; [6-(3-Methanesulfonyl-phenyl)-4-morpholin-4-yl-pyridin-2-yl]-(4-trifluoromethoxy-phenyl)-amine; N-Methyl-4-[4-morpholin-4-yl-6-(4-trifluoromethoxy-phenylamino)-pyridin-2-yl]-benzenesulfonamide; or 2′,6′-Bis-(4-trifluoromethoxy-phenylamino)-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-4-ol; or any combination thereof; or a salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof.
  • 23. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound selected from:
  • 24. The composition as claimed in claim 23, further comprising: optionally, a pharmaceutically acceptable auxiliary; optionally, a pharmaceutically acceptable preservative; optionally, a pharmaceutically acceptable excipient; optionally, a pharmaceutically acceptable diluent; and optionally, a pharmaceutically acceptable solvate.
  • 25. The composition as claimed in claim 23, further comprising an agent selected from an immunosuppressive agent, a cytotoxic agent, an anti-inflammatory agent, an antirheumatic agent, a cardiovascular agent, or any combination thereof.
  • 26. The composition as claimed in claim 23, wherein the composition is in the form of a tablet, a capsule, a cachet, a powder, a granule, a solution, a suspension, an emulsion, a bolus, a lozenge, a suppository, a pessary, a tampon, a cream, a gel, a paste, a foam, a spray, an aerosol, a microcapsule, a liposome, a transdermal patch, a pastille, a paste, or a mouthwash.
  • 27. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound selected from:
  • 28. The composition as claimed in claim 27, further comprising: optionally, a pharmaceutically acceptable auxiliary; optionally, a pharmaceutically acceptable preservative; optionally, a pharmaceutically acceptable excipient; optionally, a pharmaceutically acceptable diluent; and optionally, a pharmaceutically acceptable solvate.
  • 29. The composition as claimed in claim 27, further comprising an agent selected from an immunosuppressive agent, a cytotoxic agent, an anti-inflammatory agent, an antirheumatic agent, a cardiovascular agent, or any combination thereof.
  • 30. The composition as claimed in claim 27, wherein the composition is in the form of a tablet, a capsule, a cachet, a powder, a granule, a solution, a suspension, an emulsion, a bolus, a lozenge, a suppository, a pessary, a tampon, a cream, a gel, a paste, a foam, a spray, an aerosol, a microcapsule, a liposome, a transdermal patch, a pastille, a paste, or a mouthwash.
  • 31. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound selected from: 4-(3 -Fluoro-4-methoxy-phenyl)-pyridine; 2-Chloro-4-(3-fluoro-4-methoxy-phenyl)-pyridine; 4-(3-Fluoro-4-methoxy-phenyl)-2-phenyl-pyridine; 2-(3-Fluoro-4-methoxy-phenyl)-4-phenyl-pyridine; 2-Chloro-4-(3,4-difluoro-phenyl)-pyridine; 4-(3,4-Difluoro-phenyl)-2-phenyl-pyridine; 2-Chloro-4-(4-methanesulfonyl-phenyl)-pyridine; 2-Chloro-4-(3-methanesulfonyl-phenyl)-pyridine; 4-Benzo[1,3]dioxol-5-yl-2-chloro-pyridine; 4-Benzo[1,3]dioxol-5-yl-2-phenyl-pyridine; 2,6-Bis-(3-fluoro-4-methoxy-phenyl)-pyridine; (3-Fluoro-4-methoxy-phenyl)-(4-phenyl-pyridin-2-yl)-amine; Cycloheptyl-[4-(3-fluoro-4-methoxy-phenyl)-pyridin-2-yl]-amine; 4-[4-(3-Fluoro-4-methoxy-phenyl)-pyridin-2-yl]-morpholine; Cyclohexylmethyl-[4-(3-fluoro-4-methoxy-phenyl)-pyridin-2-yl]-amine; [4-(3-Fluoro-4-methoxy-phenyl)-pyridin-2-yl]-(4-fluoro-phenyl)-amine; (2-Chloro-pyridin-4-yl)-(4-fluoro-3-methoxy-phenyl)-amine; (3-chloro-4-methoxy-phenyl)-(2-phenyl-pyridin-4-yl)-amine; (4-fluoro-phenyl)-[4-(4-trifluoromethoxy-phenyl)-pyridin-2-yl]-amine; 2,4,6-tris-(4-fluoro-phenyl)-pyridine; (3-chloro-4-methoxy-phenyl)-(2-chloro-6-phenyl-pyridin-4-yl)-amine; benzo[1,3]dioxol-5-yl-(2,6-dichloro-pyridin-4-yl)-amine; (2,6-dichloro-pyridin-4-yl)-(3-fluoro-phenyl)-amine; (2,6-diphenyl-pyridin-4-yl)-p-tolyl-amine; (2,6-diphenyl-pyridin-4-yl)-(4-trifluoromethoxy-phenyl)-amine; [2,6-bis-(3-fluoro-phenyl)-pyridin-4-yl]-(4-trifluoromethoxy-phenyl)-amine; [2,6-bis-(4-fluoro-phenyl)-pyridin-4-yl]-methyl-(4-trifluoromethoxy-phenyl)-amine; [2,6-bis-(4-methanesulfonyl-phenyl)-pyridin-4-yl]-(4-trifluoromethoxy-phenyl)-amine; [2,6-bis-(3-(methylsulfonyl)-phenyl)-pyridin-4-yl]-(4-trifluoromethoxy-phenyl)-amine; N-ethyl-3-[6-(3 -methanesulfonyl-phenyl)-4-(4-trifluoromethoxy-phenylamino)-pyridin-2-yl]-benzamide; 1-{3-[6-(3-acetyl-phenyl)-4-(4-trifluoromethoxy-phenylamino)-pyridin-2-yl]-phenyl}-ethanone; 1-{4-[2,6-bis-(4-fluoro-phenyl)-pyridin-4-ylamino]-phenyl}-ethanone; [2,6-bis-(3-N,N-dimethyl-benzamide)-pyridin-2-yl]-(4-trifluoromethoxy-phenyl)-amine; 3-{2,6-bis-(4-fluoro-phenyl)-pyridin-4-ylamino]-benzenethiol; compound with acetic acid methyl ester; thiocarbonic acid O-methyl ester S-{3-[6-pyrrolidin-1-yl-4-(4-trifluoromethoxy-phenylamino)-pyridin-2-yl]-phenyl}ester; 1-{3-[6-pyrrolidin-1-yl-4-(4-trifluoromethoxy-phenylamino)-pyridin-2-yl]-phenyl}-ethanone; [2-(4-fluoro-phenyl)-6-(4-methyl-piperazin-1-yl)-pyridin-4-yl]-(4-trifluoromethoxy-phenyl)-amine; 1-[4,6-bis-(4-fluoro-phenyl)-pyridin-2-yl]-4-methyl-piperazine; 1-[4-(4-fluoro-phenyl)-6-(4-methanesulfonyl-phenyl)-pyridin-2-yl]-4-methyl-piperazine; 1-[6-(4-fluoro-phenyl)-4-(4-methanesulfonyl-phenyl)-pyridin-2-yl]-4-methyl-piperazine; {4-[2-(4-fluoro-phenyl)-6-(4-methyl-piperazin-1-yl)-pyridin-4-yl]-phenyl}-morpholin-4-yl-methanone; 1-[6-(4-fluoro-phenyl)-4-(4-trifluoromethoxy-phenyl)-pyridin-2-yl]-4-methyl-piperazine; 1-[4-(2-morpholin-4-yl-6-phenyl-pyridin-4-ylamino)-phenyl]-ethanone; 6′-(4-fluoro-phenyl)-4′-(4-trifluoromethoxy-phenylamino)-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4-ol; 1-[4-(4-hydroxy-6′-phenyl-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4′-ylamino)-phenyl]-ethanone; 1-[3-(4-hydroxy-6′-phenyl-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4′-ylamino)-phenyl]-ethanone; [2-(4-fluoro-phenyl)-6-morpholin-4-yl-pyridin-4-yl]-(4-trifluoromethoxy-phenyl)-amine; [2-(3-methanesulfonyl-phenyl)-6-morpholin-4-yl-pyridin-4-yl]-(4-trifluoro-methoxy-phenyl)-amine; 1-{3-[6-morpholin-4-yl-4-(4-trifluoromethoxy-phenylamino)-pyridin-2-yl]-phenyl}-ethanone; {3-[6-morpholin-4-yl-4-(4-trifluoromethoxy-phenylamino)-pyridin-2-yl]-phenyl}-pyrrolidin-1-yl-methanone; 6′-chloro-4′-phenyl-3,4,5,6-tetrahydo-2H-[1,2′]bipyridinyl-4-ol; 4′,6′-bis-(4-fluoro-phenyl)-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4-ol; 6′-(4-fluoro-phenyl)-4′-(4-methanesulfonyl-phenyl)-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4-ol; [6′-(4-Fluoro-phenyl)-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-2′-yl]-(4-trifluoromethoxy-phenyl)-amine; (4-Trifluoromethoxy-phenyl)-[6′-(4-trifluoromethoxy-phenyl)-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-2′-yl]-amine; (6′-Phenyl-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-2′-yl)-(4-trifluoro-methoxy-phenyl)-amine; [6′-(3-Methanesulfonyl-phenyl)-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-2′-yl]-(4-trifluoromethoxy-phenyl)-amine; 4-[6′-(4-Fluoro-phenyl)-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-2′-ylamino]-N-methyl-benzenesulfonamide; 1-[4-(4-Hydroxy-6′-phenyl-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-2′-ylamino)-phenyl]-ethanone; 2′,6′-Bis-(4-fluoro-phenyl)-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl; 2′,6′-Bis-(4-trifluoromethoxy-phenyl)-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl; (6-Phenyl-4-pyrrolidin-1-yl-pyridin-2-yl)-(4-trifluoromethoxy-phenyl)-amine; [6-(4-Fluoro-phenyl)-4-pyrrolidin-1-yl-pyridin-2-yl]-(4-trifluoromethoxy-phenyl)-amine; N-Methyl-4-[4-pyrrolidin-1-yl-6-(4-trifluoromethoxy-phenyl)-pyridin-2-ylamino]-benzenesulfonamide; N-Methyl-4-[4-(4-methyl-piperazin-1-yl)-6-(4-trifluoromethoxy-phenylamino)-pyridin-2-yl]-benzenesulfonamide; [6-(3-Methanesulfonyl-phenyl)-4-morpholin-4-yl-pyridin-2-yl]-(4-trifluoromethoxy-phenyl)-amine; N-Methyl-4-[4-morpholin-4-yl-6-(4-trifluoromethoxy-phenylamino)-pyridin-2-yl]-benzenesulfonamide; (4-Fluoro-phenyl)-[6-(4-trifluoromethoxy-phenyl)-pyridin-2-yl]-amine; 2′,6′-Bis-(4-trifluoromethoxy-phenylamino)-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-4-ol; N2-Cyclohexylmethyl-N4-(3-fluoro-4-methoxy-phenyl)-pyridine-2,4-diamine; [4-(2,6-dichloro-pyridin-4-yl)-phenyl]-morpholin-4-yl-methanone; 2,6-Dichloro-4-phenyl-pyridine; 2,6-Dichloro-4-(4-fluoro-phenyl)-pyridine; 2,6-Dichloro-4-(4-methanesulfonyl-phenyl)-pyridine; (3-chloro-4-methoxy-phenyl)-(2-chloro-pyridin-4-yl)-amine; 2-chloro-4-(4-trifluoromethoxy-phenyl)-pyridine; (3-chloro-4-methoxy-phenyl)-(2,6-dichloro-pyridin-4-yl)-amine; (2,6-dichloro-pyridin-4-yl)-p-tolyl-amine; (2,6-dichloro-pyridin-4-yl)-(4-trifluoromethoxy-phenyl)-amine; 3-[6-chloro-4-(4-trifluoromethoxy-phenylamino)-pyridin-2-yl]-N-ethyl-benzamide; 1-[4-(2,6-dichloro-pyridin-4-ylamino)-phenyl]-ethanone; (2,6-dichloro-pyridin-4-yl)-(3-methylsulfanyl-phenyl)-amine; S-[3-(2,6-dichloro-pyridin-4-ylamino)-phenyl]ester-O-methyl ester; (2-chloro-6-pyrrolidin-1-yl-pyridin-4-yl)-(4-trifluoromethoxy-phenyl)-amine; [2-chloro-6-(4-methyl-piperazin-1-yl)-pyridin-4-yl]-(4-trifluoromethoxy-phenyl)-amine; 2,6-dichloro-4-(4-fluoro-phenyl)-pyridine; 1-[6-chloro-4-(4-fluoro-phenyl)-pyridin-2-yl]-4-methyl-piperazine; 2,6-dichloro-4-(4-methanesulfonyl-phenyl)-pyridine; 1-[6-chloro-4-(4-methanesulfonyl-phenyl)-pyridin-2-yl]-4-methyl-piperazine; [4-(2,6-dichloro-pyridin-4-yl)-phenyl]-morpholin-4-yl-methanone; {4-[2-chloro-6-(4-methyl-piperazin-1-yl)-pyridin-4-yl]-phenyl}-morpholin-4-yl-methanone; 2,6-dichloro-4-(4-trifluoromethoxy-phenyl)-pyridine; 1-[6-chloro-4-(4-trifluoromethoxy-phenyl)-pyridin-2-yl]-4-methyl-piperazine; 1-[4-(2,6-dichloro-pyridin-4-ylamino)-phenyl]-ethanone; 1-[4-(2-chloro-6-morpholin-4-yl-pyridin-4-ylamino)-phenyl]-ethanone; 6′-chloro-4′-(4-trifluoromethoxy-phenylamino)-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4-ol; 1-[4-(6′-chloro-4-hydroxy-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4′-ylamino)-phenyl]-ethanone; 1-[4-(4-hydroxy-6′-phenyl-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4′-ylamino)-phenyl]-ethanone; 1-[3-(6′-chloro-4-hydroxy-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4′-ylamino)-phenyl]-ethanone; (2-chloro-6-morpholin-4-yl-pyridin-4-yl)-(4-trifluoromethoxy-phenyl)-amine; 2,6-dichloro-4-phenyl-pyridine; 6′-chloro-4′-(4-fluoro-phenyl)-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4-ol; 6′-chloro-4′-(4-methanesulfonyl-phenyl)-3,4,5,6-tetrahydro-2H-[1,2′]bipyridinyl-4-ol; (2-chloro-pyridin-4-yl)-(3-fluoro-4-methoxy-phenyl)-amine; 2′,6′-dichloro-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl; (6′-chloro-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-2′-yl]-(4-trifluoromethoxyphenyl)-amine; 4-(6′-chloro-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-2′-ylamino)-N-methyl-benzenesulfonamide; 2′,6′-dichloro-3,4,5,6-tetrahydro-3H-[1,4′]bipyridinyl-4-ol; 2′-chloro-6′-phenyl-3,4,5,6-tetrahydro-2H-[1,4′]bipyridinyl-4-ol; 2,6-dichloro-4-pyrrolidino pyridine; (6-chloro-4-pyrrolidin-1-yl-pyridin-2-yl)-(4-trifluoromethoxy-phenyl)-amine; 4-(6-chloro-4-pyrrolidin-1-yl-pyridin-2-ylamino)-N-methyl-benzenesulfonamide; 1-(2,6-dichloro-pyridin-4-yl)-4-methyl-piperazine; [6-chloro-4-(4-methyl-piperazin-1-yl)-pyridin-2-yl]-(4-trifluoromethoxy-phenyl)-amine; 4-(2,6-dichloro-pyridin-4-yl)-morpholine; (6-chloro-4-morpholin-4-yl-pyridin-2-yl)-(4-trifluoromethoxy-phenyl)-amine; (6-chloro-pyridine-2-yl)-(4-fluoro-phenyl)-amine; or any combination thereof; or a pharmaceutically acceptable salt, a prodrug, a diastereomeric mixture, an enantiomer, a tautomer, or a racemic mixture thereof.
  • 32. The composition as claimed in claim 31, further comprising: optionally, a pharmaceutically acceptable auxiliary; optionally, a pharmaceutically acceptable preservative; optionally, a pharmaceutically acceptable excipient; optionally, a pharmaceutically acceptable diluent; and optionally, a pharmaceutically acceptable solvate.
  • 33. The composition as claimed in claim 31, further comprising an agent selected from an immunosuppressive agent, a cytotoxic agent, an anti-inflammatory agent, an antirheumatic agent, a cardiovascular agent, or any combination thereof.
  • 34. The composition as claimed in claim 31, wherein the composition is in the form of a tablet, a capsule, a cachet, a powder, a granule, a solution, a suspension, an emulsion, a bolus, a lozenge, a suppository, a pessary, a tampon, a cream, a gel, a paste, a foam, a spray, an aerosol, a microcapsule, a liposome, a transdermal patch, a pastille, a paste, or a mouthwash.
RELATED APPLICATION DATA

This application claims the benefit of U.S. Provisional Patent Application Ser. No. 60/612,374, filed Sep. 23, 2004, which is incorporated herein by reference in its entirety.

Provisional Applications (1)
Number Date Country
60612374 Sep 2004 US