Anilinopyrimidine derivatives as IKK inhibitors and compositions and methods related thereto

Abstract
Compounds having activity as inhibitors of IKK are disclosed, particularly IKK-2. The compounds of this invention are anilinopyrimidine derivatives having the following structure: wherein R1 and R6 are as defined herein. Such compounds have utility in the treatment of a wide range of conditions that are responsive to IKK inhibition. Thus, methods of treating such conditions are also disclosed, as are pharmaceutical compositions containing one or more compounds of the above compounds.
Description
1. FIELD OF THE INVENTION

This invention is generally directed to anilinopyrimidine derivatives that have utility as IκB kinase (IKK) inhibitors, and particularly as IKK-2 inhibitors, as well to related compositions and methods.


2. BACKGROUND OF THE INVENTION

NF-κB is a heterodimeric transcription transcription factor regulating the expression of multiple inflammatory genes. The expression of more than 70 known proteins is transcriptionally regulated by the binding of NF-κB to specific sequence elements in the promoter region of these genes (Baeuerle and Baichwal, Advances in Immunology 65:111-137, 1997) NF-κB has been implicated in many pathophysiologic processes including angiogenesis (Koch et al., Nature 376:517-519, 1995), atherosclerosis (Brand et al., J Clin Inv. 97:1715-1722, 1996), endotoxic shock and sepsis (Bohrer et al., J. Clin. Inv. 100:972-985, 1997), inflammatory bowel disease (Panes et al., Am J Physiol. 269:H1955-H1964, 1995), ischemia/reperfusion injury (Zwacka et al., Nature Medicine 4:698-704, 1998), and allergic lung inflammation (Gosset et al., Int Arch Allergy Immunol. 106:69-77, 1995). Because of the central role of NF-κB in inflammatory disease, inhibition of NF-κB by targeting regulatory proteins in the NF-κB activation pathway represents an attractive strategy for generating anti-inflammatory therapeutics.


The IκB kinases (IKKs), are key regulatory signaling molecules coordinating the activation of NF-κB. IKK-1 and IKK-2 are structurally unique kinases containing an N-terminal kinase domain with a dual serine activation loop, a leucine zipper domain, and a C-terminal helix-loop-helix domain and serine cluster. IKK enzymes show relatively low sequence homologies with other kinases, and early profiles with known kinase inhibitors have not identified compounds with striking potency. Kinetic analysis shows that IKK-2 binds to and phosphorylates IκBα, IκBβ, and IκBε with high and relatively equal affinities (Heilker et. al. 1999). Recombinant IKK-2 phosphorylates IκBα peptide 26-42 with near equal affinity to full length IκBα, however the native IKK enzyme complex phosphorylates full length IκBα 25,000 fold more efficiently, suggesting important regulatory sequences in the C-terminal region of IκBα, or additional regulatory proteins in the IKK enzyme complex that accelerate the rate of catalysis (Burke et al., Journal of Biological Chemistry 274:36146-36152, 1999). Phosphorylation of IκBα occurs via a random sequential kinetic mechanism, meaning either ATP or IκBα may bind first to IKK-2, t that both must be bound before phosphorylation of IκBα can take place (Peet and Li, Journal of Biological Chemistry 274:32655-32661, 1999). IKK-2 binds ATP with uniquely high affinity (Ki=130 nM) compared to other serine-threonine kinases such as p38 and JNK perhaps indicating a unique ATP binding pocket that reflects the relatively poor activity to many broad specificity kinase inhibitors when tested against IKK-2. To date, no crystal structure of IKK-2 has been reported. However homology modeling has identified 3 structural domains including an N-terminal kinase domain with an activation loop, a leucine zipper domain that likely mediates the formation of IKK-1 and IKK-2 homo/heterodimers, and a C-terminal helix-loop-helix with serine rich tail. Activation of IKK-2 is critically dependent upon phosphorylation of serine 177 and 181 in the activation or T loop. Alanine mutations abolish activity, while glutamate mutations result in a constitutively active enzyme (Mercurio et al. Science 278:860-866, 1997; Delhase et al., Science 284:30 313, 1999).


IKK-1 and IKK-2 occur both as heterodimers and IKK-2 homodimers, and are associated with a 700-900 kDa cytoplasmic enzyme complex called the “IKK Signalsome” (Mercurio et al., Science 278:860-866, 1997). Another component, IKKAP-1 or NEMO/IKKγ has no apparent catalytic function but will associate directly with IKK-2 and is necessary for full activation of NF-κB (Mercurio et al., Mol Cell Biol. 19:1526-1538, 1999). Many immune and inflammatory mediators including TNFα, lipopolysaccharide (LPS), IL-1, anti-CD28, CD40L, FasL, viral infection, and oxidative stress have been shown to lead to NF-κB activation. Although the receptor complexes that transduce these diverse stimuli appear very different in their protein components, it is understood that each of these stimulation events leads to activation of the IKKs and NF-κB.


The IKK complex appears to be the central integrator of diverse inflammatory signals leading to the phosphorylation of I κB. IKKs are activated at dual serine residues by upstream kinases including NF-κB inducing kinase, NIK (Malinin et al., Nature 385:540-544, 1997), and MEKK-1 (Yujiri et al., Science 282:1911-1914, 1998). The differential activities of NIK and MEKK-1 remain unclear although initial data indicates these kinases may preferentially activate IKK-1 and IKK-2, respectively. Activated IKK phosphorylates a cytoplasmic inhibitor protein, IκB which binds NF-κB, thereby masking a nuclear localization signal present in Rel proteins (Cramer et al., Structure 7:R1-R6, 1999). IKK phosphorylation of IκB on serines 32 and 36 forms a structural motif recognized by the E3 ligase, βTRcP (Yaron et al., Nature 396:590-594, 1998). Docking of βTRcP results in the formation of a ligase complex which polyubiquitinates IκB thus targeting it for degradation by the 26S proteosome. Free NF-κB is then identified by nuclear transport proteins which translocate it to the nucleus where it can associate with sequence specific regulatory elements on gene promoters.


Although both kinases can phosphorylate IκB in vitro, early studies using genetic mutants indicated that IKK-2, but not IKK-1, was essential for activation of NF-κB by pro-inflammatory stimuli such as IL-1β P and TNFα. Furthermore, only catalytically inactive mutants of IKK-2 blocked the expression of NF-κB regulated genes such as monocyte chemotactic protein (MCP-1) and intercellular adhesion molecule (ICAM-1) (Mercurio et al , Science 278:860-866, 1997). Studies of knockout animals for IKK-1 and IKK-2 substantiate these initial findings (Hu et al., Science 284:316-320, 1999; Li et al., Genes & Development 13:1322-1328, 1999; Li et al., Science 284:321-324, 1999; Takeda et al., Science 84:313-316, 1999; Tanaka et al., Immunity 10:421-429, 1999). IKK-1−/− animals were born alive but died within hours. Pups showed abnormalities of the skin due to defective proliferation and differentiation, but showed no gross deficiency in cytokine induced activation of NF-κB. In contrast, IKK-2−/− embryos died at day 14-16 of pregnancy from liver degeneration and apoptosis that bore a striking resemblance to that observed in Rel A knock-out animals (Beg et al., Nature 376:167-170, 1995). Furthermore, embryonic fibroblasts from IKK-2−/− animals exhibited markedly reduced NF-κB activation following cytokine stimulation, while IKK-1−/− did not.


Accordingly, cell and animal experiments indicate that IKK-2 is a central regulator of the pro-inflammatory role of NF-κB. IKK-2 is activated in response to multiple inflammatory stimuli and signaling pathways, many of which play an important role in respiratory disease including IL-1β, LPS, TNFα, CD3/CD28 (antigen presentation), CD40L, viral infection, and oxidative stress. The ubiquitous expression of NF-κB, along with its response to multiple stimuli means that almost all cell types present in the lung are potential target for anti-NF-κB/IKK-2 therapy. This includes alveolar epithelium, mast cells, fibroblasts, vascular endothelium, and infiltrating leukocytes; neutrophils, macrophages, lympophocytes, eosinophils and basophils. By inhibiting the expression of genes such as cyclooxygenase-2 and 12-lipoxygenase (synthesis of inflammatory mediators), TAP-1 peptide transporter (antigen processing), MHC class I H-2K and class II invariant chains (antigen presentation), E-selectin and vascular cell adhesion molecule (leukocyte recruitment), interleukins-1, 2, 6, 8 (cytokines), RANTES, eotaxin, GM-CSF (chemokines), and superoxide dismutase and NADPH quinone oxidoreductase (reactive oxygen species), inhibitors of IKK-2 are believed to display broad anti-inflammatory activity.


International Publication No. WO 98/18782 to Celltech Therapeutics Limited discloses 4-pyridyl pyrimidine compounds which are allegedly useful in the prophylaxis and treatment of immune diseases, allergic diseases involving mast cells or eosinophils, and diseases involving inappropriate platelet activation.


Accordingly, there is a need in the art for selective inhibitors of IKK, particularly IKK2 inhibitors. In addition, there is a need for pharmaceutical compositions comprising one or more inhibitors, as well as to methods for treating conditions in animals which are responsive to such inhibitors. The present invention fulfills these needs, and provides further related advantages.


Citation of identification of any reference in Section 2 of this application shall not be construed as an admission that such reference is prior art to the present invention.


3. SUMMARY OF THE INVENTION

In brief, the present invention is directed to compounds having activity as inhibitors, preferably selective inhibitors, of as IκB kinase (IKK), particularly IKK-2, and to compositions an methods related thereto.


The compounds of the present invention are “anilinopyrimidine derivatives” having the following structure (I):
embedded image

wherein R1 though R6 are as defined below, and including isomers, prodrugs and pharmaceutically acceptable salts thereof.


In general, the present invention is directed to methods for treating or preventing a condition responsive to IKK-2 inhibition, comprising administering to a patient in need thereof an effective amount of an anilinopyrimidine derivative.


The present invention is also directed to methods for treating or preventing an inflammatory or autoimmune condition comprising administering to a patient in need thereof an effective amount of an anilinopyrimidine derivative.


The present invention is also directed to methods for treating or preventing a cardiovascular, metabolic or ischemic condition comprising administering to a patient in need thereof an effective amount of an anilinopyrimidine derivative.


The present invention is also directed to methods for treating or preventing an infectious disease comprising administering to a patient in need thereof an effective amount of an anilinopyrimidine derivative.


The present invention is also directed to methods for treating or preventing cancer comprising administering to a patient in need thereof an effective amount of an anilinopyrimidine derivative.


The present invention is also directed to methods for treating or preventing stroke, epilepsy, Alzheimer's disease, or Parkinson's disease comprising administering to a patient in need thereof an effective amount of an anilinopyrimidine derivative.


These and other aspects of this invention will be evident upon reference to the following detailed description and illustrative examples, which are intended to exemplify non-limiting embodiments of the invention. Certain patent and other documents are cited herein to more specifically set forth various aspects of this invention. Each of these documents are hereby incorporated by reference in their entirety.







4. DETAILED DESCRIPTION OF THE INVENTION

The present invention is directed to anilinopyrimidine derivatives having activity as inhibitors, preferably selective inhibitors, of as IκB kinase (IKK), particularly IKK-2, and to compositions an methods related thereto.


The anilinopyrimidine derivatives have the following structure (I):
embedded image

including isomers, prodrugs and pharmaceutically acceptable salts thereof,


wherein:

    • R1 is aryl or heteroaryl optionally substituted with one to four substituents independently selected from R7;
    • R2 is hydrogen;
    • R3 is hydrogen or lower alkyl;
    • R4 represents one to four optional substituents, wherein each substituent is the same or different and independently selected from halogen, hydroxy, lower alkyl and lower alkoxy;
    • R5 and R6 are the same or different and independently —R8, —(CH2)aC(═O)R9, —(CH2)aC(═O)OR9, —(CH2)aC(═O)NR9R10, —(CH2)aC(═O)NR9(CH2)bC(═O)R10, —(CH2)aNR9C(═O)R10, (CH2)aNR11C(═O)NR9R10, —(CH2)aNR9, R10, —(CH2)aOR9, —(CH2)a,SOcR9 or —(CH2)aSO2NR9R10;
    • or R5 and R6 taken together with the nitrogen atom to which they are attached to form a heterocycle or substituted heterocycle;
    • R7 is at each occurrence independently halogen, hydroxy, cyano, nitro, carboxy, alkyl, alkoxy, haloalkyl, acyloxy, thioalkyl, sulfinylalkyl, sulfonylalkyl, hydroxyalkyl, aryl, substituted aryl, aralkyl, substituted aralkyl, heterocycle, substituted heterocycle, heterocyclealkyl, substituted heterocyclealkyl, —C(═O)OR8, —OC(═O)R8, —C(═O)NR8R9, —C(═O)NR8OR9, —SOcR8, —SOcNR8R9, —NR8SOcR9, —NR8R9, —NR8C(═O)R9, —NR8C(═O)(CH2)bOR9, —NR8C(═O)(CH2)bR9, —O(CH2)bNR8R9, or heterocycle fused to phenyl;
    • R8, R9, R10 and R11 are the same or different and at each occurrence independently hydrogen, alkyl, substituted alkyl, aryl, substituted aryl, aralkyl, substituted arylalkyl, heterocycle, substituted heterocycle, heterocyclealkyl or substituted heterocyclealkyl;
    • or R8 and R9 taken together with the atom or atoms to which they are attached to form a heterocycle or substituted heterocycle;
    • a and b are the same or different and at each occurrence independently selected from 0, 1, 2, 3 or 4; and
    • c is at each occurrence 0, 1 or 2.


In one embodiment of the invention, in the anilinopyrimidine derivatives of structure (I), R1 is a substituted or unsubstituted aryl or heteroaryl with the proviso that the heteroaryl is not pyridyl. When R1 is substituted, it is substituted with one or more substituents defined below. Preferably, when substituted, R1 is substituted with a halogen, sulfone or sulfonamide.


In another embodiment of the invention, in the anilinopyrimidine derivatives of structure (I), R1 is substituted or unsubstituted aryl, furyl, benzofuranyl, thiophenyl, benzothiophenyl, quinolinyl, pyrrolyl, indolyl, oxazolyl, benzoxazolyl, imidazolyl, benzimidazolyl, thiazolyl, benzothiazolyl, isoxazolyl, pyrazolyl, isothiazolyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, cinnolinyl, phthalazinyl or quinazolinyl.


In another embodiment of the invention, in the anilinopyrimidine derivatives of structure (I), R1 is substituted or unsubstituted aryl or heteroaryl with the proviso that the heteroaryl is not imidazo[1,2a]pyrid-3-yl or pyrazolo[2,3a]pyrid-3-yl. When R1 is substituted, it is substituted with one or more substituents defined below. Preferably, when substituted, R1 is substituted with a halogen, sulfone or sulfonamide.


In another embodiment of the invention, in the anilinopyrimidine derivatives of structure (I), R1 is substituted or unsubstituted aryl, preferably phenyl. When R1 is a substituted aryl, the aryl is substituted with one or more substituents defined below. Preferably, when substituted, R1 is substituted with a halogen, sulfone or sulfonamide.


In another embodiment of the invention, in anilinopyrimidine derivatives of structure (I), R5 and R6, taken together with the nitrogen atom to which they are attached form a substituted or unsubstituted nitrogen-containing non-aromatic heterocycle, preferably piperazinyl, piperidinyl or morpholinyl.


When R5 and R6, taken together with the nitrogen atom to which they are attached form substituted piperazinyl, piperadinyl or morpholinyl, the piperazinyl, piperadinyl or morpholinyl is substituted with one or more substituents defined below. Preferably, when substituted, the substituent is alkyl, amino, alkylamino, alkylether, acyl, pyrrolidinyl or piperidinyl.


In one embodiment of the invention, in the anilinopyrimidine derivatives of structure (I), R3 is hydrogen and R4 is not present, and the compounds of this invention have the following structure (II):
embedded image


In a more specific embodiment of the invention, in the anilinopyrimidine derivatives of structure (II), R1 is phenyl optionally substituted with R7, and having the following structure (III):
embedded image


In still a further embodiment of the invention, in the anilinopyrimidine derivatives of structure (II), R7 is at the para position relative to the pyrimidine, as represented by the following structure (IV):
embedded image


As used herein, the terms used above having following meaning:


“Alkyl” means a straight chain or branched, saturated or unsaturated alkyl, cyclic or non-cyclic hydrocarbon having from 1 to 10 carbon atoms, while “lower alkyl” has the same meaning but only has from 1 to 6 carbon atoms. Representative saturated straight chain alkyls include methyl, ethyl, n-propyl, n-butyl, n-pentyl, n-hexyl, and the like; while saturated branched alkyls include isopropyl, sec-butyl, isobutyl, tert-butyl, isopentyl, and the like. Unsaturated alkyls contain at least one double or triple bond between adjacent carbon atoms (also referred to as an “alkenyl” or “alkynyl”, respectively). Representative straight chain and branched alkenyls include ethylenyl, propylenyl, 1-butenyl, 2-butenyl, isobutylenyl, 1-pentenyl, 2-pentenyl, 3-methyl-1-butenyl, 2-methyl-2-butenyl, 2,3-dimethyl-2-butenyl, and the like; while representative straight chain and branched alkynyls include acetylenyl, propynyl, 1-butynyl, 2-butynyl, 1-pentynyl, 2-pentynyl, 3-methyl-1 butynyl, and the like. Representative saturated cyclic alkyls include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like; while unsaturated cyclic alkyls include cyclopentenyl and cyclohexenyl, and the like. Cycloalkyls are also referred to herein as “carbocyclic” rings systems, and include bi- and tri-cyclic ring systems having from 8 to 14 carbon atoms such as a cycloalkyl (such as cyclopentane or cyclohexane) fused to one or more aromatic (such as phenyl) or non-aromatic (such as cyclohexane) carbocyclic rings.


“Halogen” means fluorine, chlorine, bromine or iodine.


“Keto” means a carbonyl group (i.e., ═O).


“Aryl” means an aromatic carbocyclic moiety such as—phenyl or naphthyl.


“Arylalkyl” means an alkyl having at least one alkyl hydrogen atom replaced with an aryl moiety, such as benzyl, —(CH2)2phenyl, —(CH2)3phenyl, —CH(phenyl)2, and the like.


“Heteroaryl” means an aromatic heterocycle ring of 5- to 10 members and having at least one heteroatom selected from nitrogen, oxygen and sulfur, and containing at least 1 carbon atom, including both mono- and bicyclic ring systems. Representative heteroaryls are pyridyl, furyl, benzofuranyl, thiophenyl, benzothiophenyl, quinolinyl, pyrrolyl, indolyl, oxazolyl, benzoxazolyl, imidazolyl, benzimidazolyl, thiazolyl, benzothiazolyl, isoxazolyl, pyrazolyl, isothiazolyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, cinnolinyl, phthalazinyl, and quinazolinyl.


“Heteroarylalkyl” means an alkyl having at least one alkyl hydrogen atom replaced with a heteroaryl moiety, such as —CH2pyridinyl, —CH2pyrimidinyl, and the like.


“Heterocycle” means a heterocyclic ring containing from 5 to 10 ring atoms


“Heterocycle” means a 5- to 7-membered monocyclic, or 7- to 10-membered bicyclic, heterocyclic ring which is either saturated, unsaturated, or aromatic, and which contains from 1 to 4 heteroatoms independently selected from nitrogen, oxygen and sulfur, and wherein the nitrogen and sulfur heteroatoms may be optionally oxidized, and the nitrogen heteroatom may be optionally quaternized, including bicyclic rings in which any of the above heterocycles are fused to a benzene ring. The heterocycle may be attached via any heteroatom or carbon atom. Heterocycles include heteroaryls as defined above. Thus, in addition to the heteroaryls listed above, heterocycles also include morpholinyl, pyrrolidinonyl, pyrrolidinyl, piperidinyl, piperazinyl, hydantoinyl, valerolactamyl, oxiranyl, oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydropyrindinyl, tetrahydroprimidinyl, tetrahydrothiophenyl, tetrahydrothiopyranyl, tetrahydropyrimidinyl, tetrahydrothiophenyl, tetrahydrothiopyranyl, and the like.


“Heterocyclealkyl” means an alkyl having at least one alkyl hydrogen atom replaced with a heterocycle, such as —CH2morpholinyl, and the like.


The term “substituted” as used herein means any of the above groups (i.e., aryl, arylalkyl, heterocycle and heterocyclealkyl) wherein at least one hydrogen atom is replaced with a substituent. In the case of a keto substituent (“C(═O)”) two hydrogen atoms are replaced. Substituents include halogen, hydroxy, alkyl, substituted alkyl (such as haloalkyl, mono- or di-substituted aminoalkyl, alkyloxyalkyl, and the like, aryl, substituted aryl, arylalkyl, substituted arylalkyl, heterocycle, substituted heterocycle, heterocyclealkyl, substituted heterocyclealkyl, —NRaRb, —NRaC(═O)Rb, —NRaC(═O)NRaRb, —NRaC(═O)ORb—NRaSO2Ra, —ORa, —C(═O)Ra—C(═O)ORa—C(═O)NRaRb, —OC(═O)Ra, —OC(═O)ORa, —OC(═O)NRaRb—NRaSO2Rb, or a radical of the formula —Y—Z—Ra where Y is alkanediyl, substitute alkanediyl, or a direct bond, Z is —O—, —S—, —S(═O)—, —S(═O)2—, —N(Rb)—, —C(═O)—, —C(═O)O—, —OC(═O)—, —N(Rb)C(═O)—, —C(═O)N(Rb)— or a direct bond, wherein Ra and Rb are the same or different and independently hydrogen, amino, alkyl, substituted alkyl (including halogenated alkyl), aryl, substituted aryl, arylalkyl, substituted arylalkyl, heterocycle, substituted heterocycle, heterocylealkyl or substituted heterocyclealkyl, or wherein Ra and Rb taken together with the nitrogen atom to which they are attached form a heterocycle or substituted heterocycle.


“Haloalkyl” means alkyl having one or more hydrogen atoms replaced with halogen, such as —CF3.


“Hydroxyalkyl” means alkyl having one or more hydrogen atoms replaced with hydroxy, such as —CH2OH


“Sulfonylalkyl” means —SO2-(alkyl);


“Sulfinylalkyl” means —SO-(alkyl);


“Thioalkyl” means —S-(alkyl);


“Carboxyl” means —COOH.


“Alkoxy” means —O-(alkyl), such as methoxy, ethoxy, n-propyloxy, iso-propyloxy, n-butyloxy, iso-butyloxy, and the like.


“Patient” means an animal, including, but not limited to, an animal such as a cow, monkey, horse, sheep, pig, chicken, turkey, quail, cat, dog, mouse, rat, rabbit, and guinea pig, and is more preferably a mammal, and most preferably a human.


“Acyl” means alkyl(C═O)


“ClH” means the hydrochloride salt of compounds depicted by their chemical structure.


“Nitrogen-containing non-aromatic heterocycle” means morpholinyl, thiomorpholinyl, pyrrolidinonyl, pyrrolidinyl, piperidinyl, homopiperidinyl, piperazinyl, homopiperazinyl, hydantoinyl, tetrahydropyrindinyl, tetrahydropyrimidinyl, oxazolidinyl, thiazolidinyl, indolinyl, isoindolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl and the like.


The anilinopyrimidine derivatives can generally be obtained using organic synthesis techniques known to those skilled in the art, as well as by the following general techniques and the procedures set forth in the Examples. To that end, the anilinopyrimidine derivatives can be made according to the following Reaction Schemes 1 through 9:
embedded image


Appropriately substituted methylketones may be treated with a dimethylformamide acetal, such as dimethylformamide dimethylacetal or dimethylformamide diethylacetal, to afford the corresponding β-dimethylaminobutenones. Treatment of the aminobutenones with thiourea in the presence of a base such as sodium methoxide, followed by alkylation with an alkyl halide, such as methyl iodide, gives 4-substituted 2-alkylthiopyrimidines. Oxidation of the thioether with organic and inorganic oxidizing agents, such as m-chloroperbenzoic acid or oxone, yields the sulfones which, upon condensation with p-aminocarbonylanilines, give rise to the formation of the desired anilinopyrimidine derivatives.
embedded image


Similarly, the anilinopyrimidine derivatives may be prepared from the 2-chloropyrimidine derivatives. Thus, condensation of the β-dimethylaminobutenones with urea followed y the treatment with chlorinating agent such as phosphorus oxychloride gives 4-substituted 2-chloropyrimidines. Further treatment with substituted anilines affords the desired anilinopyrimidine derivatives.
embedded image


The anilinopyrimidine derivatives can also be prepared by condensation of the β-dimethylaminobutenones with appropriately substituted guanidines. The requisite guanidines may be synthesized by the reaction of the aniline with cyanamide in the presence of an acid, or with a pyrazoloamidine.
embedded image


Cyclization of alkoxycarbonylphenylguanidines with the b-aminoketones gives 4-substituted 2-(4-carboxyphenyl)aminopyrimidines. Condensation of the benzoic acid derivatives with appropriate amines affords the desired amides.
embedded image


Condensation of the benzoic acids with N-Boc-piperazine followed by deprotection of the tert-butoxycarbonyl group with an acid such as hydrochloric acid yields piperazineamides. Subsequent condensation with carboxylic acid derivatives yields bisacylpiperazines.
embedded image


Similar reaction with sulfonyl chlorides gives the corresponding sulfonamides.
embedded image


Acetophenones with p-alkyl- and arylthio groups may be prepared by the reaction of p-chloroacetophenone with alkyl and arylthiols.
embedded image


Anilinopyrimidine derivatives having the p-alkyl- and arylsulfenyl groups may be prepared by controlled oxidation of the sulfides with an oxidizing agent such as oxone.
embedded image


Anilinopyrimidine derivatives having p-alkyl- and arylsulfonyl groups may be prepared by oxidation of the sulfides with an oxidizing agent such as oxone.


The anilinopyrimidine derivatives can be in the form of a pharmaceutically acceptable salt or free base. Acid addition salts of the free base can be prepared by methods well known in the art, and may be formed from organic and inorganic acids. Suitable organic acids include maleic, fumaric, benzoic, ascorbic, succinic, methanesulfonic acetic, oxalic, propionic, tartaric, salicylic, citric, gluconic, lactic, mandelic, cinnamic, aspartic, stearic, palmitic, glycolic, glutamic, and benzenesulfonic acids. Suitable inorganic acids include hydrochloric, hydrobromic, sulfuric, phosphoric, and nitric acids. Additional salts include sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, sulfate, bisulfate, phosphate, acid phosphate, isonicotinate, acetate, lactate, salicylate, citrate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucaronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate, and pamoate (i.e., 1,1′-methylene-bis-(2-hydroxy-3-naphthoate)) salts. The term “pharmaceutically acceptable salt” is intended to encompass any and all acceptable salt forms.


Pharmaceutically acceptable salts can be formed by conventional and known techniques, such as by reacting a compound of this invention with a suitable acid as disclosed above. Such salts are typically formed in high yields at moderate temperatures, and often are prepared by merely isolating the compound from a suitable acidic wash in the final step of the synthesis. The salt-forming acid may dissolved in an appropriate organic solvent, or aqueous organic solvent, such as an alkanol, ketone or ester. On the other hand, if the anilinopyrimidine derivative is desired in the free base form, it may be isolated from a basic final wash step, according to known techniques. For example, a typical technique for preparing hydrochloride salt is to dissolve the free base in a suitable solvent, and dry the solution thoroughly, as over molecular sieves, before bubbling hydrogen chloride gas through it.


The anilinopyrimidine derivatives can also exist in various isomeric forms, including configurational, geometric and conformational isomers, as well as existing in various tautomeric forms, particularly those that differ in the point of attachment of a hydrogen atom. As used herein, the term “isomer” is intended to encompass all isomeric forms of a compound, including tautomeric forms of the compound.


As used herein, the term “prodrug” refers to any derivative of the anilinopyrimidine derivatives that are metabolized or otherwise converted into an active form upon introduction into the body of an animal. Prodrugs are well known to those skilled in the art of pharmaceutical chemistry, and provide benefits such as increased adsorption and half-life. Prodrugs of this invention may be formed when, for example, hydroxy groups are esterified or alkylated, or when carboxyl groups are esterified. Those skilled in the art of drug delivery will readily appreciate that the pharmacokinetic properties of anilinopyrimidine derivatives may be controlled by an appropriate choice of moieties to produce prodrug derivatives.


In another embodiment, the present invention provides a method for treating or preventing a condition responsive to IKK-2 inhibition, comprising administering to a patient in need thereof an effective amount of an anilinopyrimidine derivative having the formula of structure (I):
embedded image

including isomers, prodrugs and pharmaceutically acceptable salts thereof,


wherein

    • R1 is aryl or heteroaryl optionally substituted with one to four substituents independently selected from R7;
    • R2 and R3 are the same or different and are independently hydrogen or lower alkyl;
    • R4 represents one to four optional substituents, wherein each substituent is the same or different and independently selected from halogen, hydroxy, lower alkyl and lower alkoxy;
    • R5 and R6 are the same or different and independently —R8, —(CH2)aC(═O)R9, —(CH2)aC(═O)OR9, —(CH2)aC(═O)NR9R10, —(CH2)C(═O)NR9(CH2)bC(═O)R10, —(CH2)aNR9C(═O)R10, (CH2)aNR11C(═O)NR9R10, —(CH2)aNR9R10, —(CH2)aOR9, —(CH2)aSOcR9 or —(CH2)aSO2NR9R10;
    • or R5 and R6 taken together with the nitrogen atom to which they are attached to form a heterocycle or substituted heterocycle;
    • R7 is at each occurrence independently halogen, hydroxy, cyano, nitro, carboxy, alkyl, alkoxy, haloalkyl, acyloxy, thioalkyl, sulfinylalkyl, sulfonlyalkyl, aryl, substituted aryl, aralkyl, substituted aralkyl, heterocycle, substituted heterocycle, heterocyclealkyl, substituted heterocyclealkyl, —C(═O)OR8, —OC(═O)R8, —C(═O)NR8R9, —C(═O)NR8OR9, —SOcR8, —SOcNR8R9, —NR8SOcR9, —NR8R9, —NR8C(═O)R9, —NR8C(═O)(CH2)bOR9, —NR8C(═O)(CH2)bR9, —O(CH2)bNR8R9, or heterocycle fused to phenyl;
    • R8, R9, R10 and R11 are the same or different and at each occurrence independently hydrogen, alkyl, substituted alkyl, aryl, substituted aryl, aralkyl, substituted arylalkyl, heterocycle, substituted heterocycle, heterocyclealkyl or substituted heterocyclealkyl;
    • or R8 and R9 taken together with the atom or atoms to which they are attached to form a heterocycle or substituted heterocycle;
    • a and b are the same or different and at each occurrence independently selected from 0, 1, 2, 3 or 4; and
    • c is at each occurrence 0, 1 or 2.


In another embodiment, the present invention provides a method for treating or preventing an inflammatory or autoimmune condition comprising administering to a patient in need thereof an effective amount of an anilinopyrimidine derivative.


In another embodiment, the present invention provides a method for treating or preventing a cardiovascular, metabolic or ischemic condition comprising administering to a patient in need thereof an effective amount of an anilinopyrimidine derivative.


In another embodiment, the present invention provides a method for treating or preventing an infectious disease comprising administering to a patient in need thereof an effective amount of an anilinopyrimidine derivative.


In another embodiment, the present invention provides a method for treating or preventing cancer comprising administering to a patient in need thereof an effective amount of an anilinopyrimidine derivative.


In another embodiment, the present invention provides a method for treating or preventing stroke, epilepsy, Alzheimer's disease comprising administering to a patient in need thereof an effective amount of an anilinopyrimidine derivative.


In another embodiment of the present methods, in the anilinopyrimidine derivatives of structure (I), R1 is a substituted or unsubstituted aryl or heteroaryl with the proviso that the heteroaryl is not pyridyl. When R1 is substituted, it is substituted with one or more substituents defined above. Preferably, when substituted, R1 is substituted with a halogen, sulfone or sulfonamide.


In another embodiment of the present methods, in the anilinopyrimidine derivatives of structure (I), R1 is substituted or unsubstituted aryl, furyl, benzofuranyl, thiophenyl, benzothiophenyl, quinolinyl, pyrrolyl, indolyl, oxazolyl, benzoxazolyl, imidazolyl, benzimidazolyl, thiazolyl, benzothiazolyl, isoxazolyl, pyrazolyl, isothiazolyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, cinnolinyl, phthalazinyl or quinazolinyl.


In another embodiment of the present methods, in the anilinopyrimidine derivatives of structure (I), R1 is substituted or unsubstituted aryl or heteroaryl with the proviso that the heteroaryl is not imidazo[1,2a]pyrid-3-yl or pyrazolo[2,3a]pyrid-3-yl. When R1 is substituted, it is substituted with one or more substituents defined above. Preferably, when substituted, R1 is substituted with a halogen, sulfone or sulfonamide.


In another embodiment of the present methods, in the anilinopyrimidine derivatives of structure (I), R1 is substituted or unsubstituted aryl, preferably phenyl or naphthyl. When R1 is a substituted aryl, it is substituted with one or more substituents defined above. Preferably, when substituted, R1 is substituted with a halogen, sulfone or sulfonamide.


In another embodiment of the present methods, in the anilinopyrimidine derivatives of structure (I), R5 and R6 taken together with the nitrogen atom to which they are attached form a substituted or unsubstituted nitrogen-containing non-aromatic heterocycle.


In another embodiment of the present methods, the nitrogen-containing non-aromatic heterocycle is piperazinyl, piperadinyl or morpholinyl. When the nitrogen-containing non-aromatic heterocycle is a substituted piperazinyl, piperadinyl or morpholinyl ring, the substituent is defined above. Preferably, when substituted, the substituent is alkyl, amino, alkylamino, alkylether, acyl, pyrrolidinyl or piperidinyl.


When used in the present methods, the anilinopyrimidine derivatives can be administered as a component of a composition that optionally comprises a pharmaceutically acceptable carrier or vehicle.


Conditions that may be treated using an anilinopyrimidine derivative, or using a pharmaceutical composition containing the same, include any condition that is responsive to IKK inhibition, particularly IKK-2 inhibition, and thereby benefit from administration of such an inhibitor. In general, the anilinopyrimidine derivatives of this invention may be used for the prevention and/or treatment of an inflammatory or autoimmune condition, a cardiovascular, metabolic or ischemic condition, an infectious disease or cancer. Representative conditions in this regard include (but not limited to) rheumatoid arthritis, rheumatoid spondylitis, osteoarthritis, gout, asthma, bronchitis, allergic rhinitis, chronic obstructive pulmonary disease, cystic fibrosis, inflammatory bowel disease, irritable bowel syndrome, mucous colitis, ulcerative colitis, Crohn's disease, Huntington's disease, gastritis, esophagitis, hepatitis, pancreatitis, nephritis, multiple sclerosis, lupus erythematosus, Type II diabetes, osteoporosis, erectile dysfunction, atherosclerosis, restenosis following angioplasty, left ventricular hypertrophy, myocardial infarction, stroke, ischemic diseases of heart, kidney, liver, and brain, organ transplant rejection, graft versus host disease, endotoxin shock, multiple organ failure, psoriasis, eczema, dermatitis, epilepsy, Alzheimer's disease, Parkinson's disease, Lou Gehrig's disease, sepsis, conjunctivitis, acute respiratory distress syndrome, purpura, nasal polip, viral infections (e.g., those caused by human immunodeficiency virus, hepatitis B virus, hepatitis C virus, human papillomavirus, human T-cell leukemia virus or Epstein-Bar virus), cachexia, and cancers of a variety of tissues such as colon, rectum, prostate, liver, lung, bronchus, pancreas, brain, head, neck, stomach, skin, kidney, cervix, blood, larynx, esophagus, mouth, pharynx, urinary bladder, ovary, bone marrow, thymus, breast, bone and uterine.


The anilinopyrimidine derivatives can also be used in cancer adjuvant therapy in combination with a cytotoxic agent or with radiation therapy.


The anilinopyrimidine derivatives are particularly useful in the treatment and/or prevention of bronchitis, multiple sclerosis, nasal polip and viral infections such as that caused by human immunodeficiency virus, hepatitis B virus, hepatitis C virus, human papillomavirus, human T-cell leukemia virus or Epstein-Barr virus.


The anilinopyrimidine derivatives can be administered to a patient orally or parenterally in conventional and well known preparations, such as capsules, microcapsules, tablets, granules, powder, troches, pills, suppositories, injections, suspensions and syrups. Prior to administration, the anilinopyrimidine derivatives are typically formulated as a pharmaceutical composition that contains an effective dosage amount of one or more of such compounds in combination with one (or more) pharmaceutically acceptable carrier(s). Suitable formulations in this regard may be prepared by methods commonly employed using conventional, organic or inorganic additives, such as an excipient (e.g., sucrose, starch, mannitol, sorbitol, lactose, glucose, cellulose, talc, calcium phosphate or calcium carbonate), a binder (e.g., cellulose, methylcellulose, hydroxymethyl cellulose, polypropylpyrrolidone, polyvinylpyrrolidone, gelatin, gum arabic, polyethyleneglycol, sucrose or starch), a disintegrator (e.g., starch, carboxymethylcellulose, hydroxypropylstarch, low substituted hydroxypropylcellulose, sodium bicarbonate, calcium phosphate or calcium citrate), a lubricant (e.g., magnesium stearate, light anhydrous sicilic acid, talc or sodium lauryl sulfate), a flavoring agent (e.g., citric acid, menthol, glycine or orange powder) a preservative (e.g., sodium benzoate, sodium bisulfite, methylparaben or propylparaben), a stabilizer (e.g., citric acid, sodium citrate or acetic acid), a suspending agent (e.g., methylcellulose, polyvinyl pyrroliclone or aluminum stearate), a dispersing agent (e.g., hydroxypropylmethylcellulose), a diluent (e.g., water), and/or a base wax (e.g., cocoa butter, white petrolatum or polyethylene glycol).


The dose of an anilinopyrimidine derivative to be administered to a patient, such as a human, is rather widely variable and subject to the judgment of the attending physician. The general range of effective administration rates of the anilinopyrimidine derivatives are from about 0.05 mg/day to about 250 mg/day, and typically from about 0.25 mg/day to 60 mg/day. Of course, it is often practical to administer the daily dose of compound in portions, at various hours of the day. However, in any given case, the amount of compound administered will depend on such factors as the solubility of the active component, the formulation use, subject condition (such as weight), and/or the route of administration.


Further, the effect of the anilinopyrimidine derivatives can be delayed or prolonged by proper formulation. For example, a slowly soluble pellet of the anilinopyrimidine derivative may be prepared and incorporated in a tablet or capsule. The technique may be improved by making pellets of several different dissolution rates and filling capsules with a mixture of the pellets. Tablets or capsules may be coated with a film which resists dissolution for a predictable period of time. Even the parenteral preparations may be made long-acting, by dissolving or suspending the compound in oily or emulsified vehicles which allow it to disperse only slowly in the serum.


In certain embodiments, the anilinopyrimidine derivatives can be used in combination, e.g., as an adjunct therapy, with at least one other therapeutic agent. An anilinopyrimidine derivative and the other therapeutic agent can act additively or, more preferably, synergistically. In a preferred embodiment, an anilinopyrimidine derivative is administered concurrently with the administration of another therapeutic agent, which can be part of the same composition as or in a different composition from that comprising the anilinopyrimidine derivative. In another embodiment, an anilinopyrimidine derivative is administered prior or subsequent to administration of another therapeutic agent. As many of the disorders for which the anilinopyrimidine derivatives are useful in treating are chronic, in one embodiment combination therapy involves alternating between administering an anilinopyrimidine derivative and another therapeutic agent. The duration of administration of the anilinopyrimidine derivative or the other therapeutic agent can be, e.g., one month, three months, six months, a year, or for more extended periods, such as the patient's lifetime. In certain embodiments, when a composition of the invention is administered concurrently with another therapeutic agent that potentially produces adverse side effects including, but not limited to, toxicity, the other therapeutic agent can advantageously be administered at a dose that falls below the threshold at which the adverse side effect is elicited.


The other therapeutic agent can be an anti-inflammatory agent. Useful anti-inflammatory agents include, but are not limited to, non-steroidal anti-inflammatory drugs such as salicylic acid, acetylsalicylic acid, methyl salicylate, diflunisal, salsalate, olsalazine, sulfasalazine, acetaminophen, indomethacin, sulindac, etodolac, mefenamic acid, meclofenamate sodium, tolmetin, ketorolac, dichlofenac, ibuprofen, naproxen, naproxen sodium, fenoprofen, ketoprofen, flurbinprofen, oxaprozin, piroxicam, meloxicam, ampiroxicam, droxicam, pivoxicam, tenoxicam, nabumetome, phenylbutazone, oxyphenbutazone, antipyrine, aminopyrine, apazone and nimesulide; leukotriene antagonists including, but not limited to, zileuton, aurothioglucose, gold sodium thiomalate and auranofin; and other anti-inflammatory agents including, but not limited to, colchicine, allopurinol, probenecid, sulfinpyrazone and benzbromarone. Anti-inflammatory agents particularly useful for treating arthritis, including rhumatiod arthritis, include enbrel, infliximab, anarkinra, celecoxib and rofecoxib.


The other therapeutic agent can be an anti-cancer agent. Useful anti-cancer agents include, but are not limited to, nitrogen mustards, such as cyclophosphamide, Ifosfamide, trofosfamide and Chlorambucil; nitrosoureas, such as carmustine (BCNU) and Lomustine (CCNU); alkylsulphonates, such as busulfan and Treosulfan; triazenes, such as Dacarbazine; platinum-containing compounds, such as Cisplatin and carboplatin; vinca alkaloids, such as vincristine, Vinblastine, Vindesine and Vinorelbine; taxoids, such as paclitaxel and Docetaxol; epipodophyllins, such as etoposide, Teniposide, Topotecan, 9-aminocamptothecin, camptoirinotecan and crisnatol; mytomycins, such as mytomycin C; DHFR inhibitors, such as methotrexate and Trimetrexate; IMP-dehydrogenase inhibitors, such as mycophenolic acid, Tiazofurin, Ribavirin and EICAR; ribonuclotide-reductase inhibitors, such as hydroxyurea and deferoxamine; uracil analogs, such as 5-fluorouracil, Floxuridine, Doxifluridine and Ratitrexed; cytosine analogs, such as cytarabine (ara C), cytosine arabinoside and fludarabine; purine analogs, such as mercaptopurine and thioguanine; anti-estrogens, such as Tamoxifen, Raloxifene and megestrol; LHRH agonists, such as goserclin and Leuprolide acetate; anti-androgens, such as flutamide and bicalutamide; vitamin D3 analogs, such as B 1089, CB 1093 and KH 1060; photodynamic therapeutic agents, such as vertoporfin (BPD-MA), Phthalocyanine, photosensitizer Pc4 and demethoxyhypocrellin A (2BA-2-DMHA); cytokines, such as interferon-α, interferon-γ and tumor-necrosis factor; isoprenylation inhibitors, such as Lovastatin; dopaminergic neurotoxins, such as 1-methyl-4-phenylpyridinium ion; cell-cycle inhibitors, such as staurosporine; actinomycins, such as Actinomycin D and Dactinomycin; bleomycins, such as bleomycin A2, Bleomycin B2 and Peplomycin; anthracyclines, such as daunorubicin, Doxorubicin (adriamycin), Idarubicin, Epirubicin, Pirarubicin, Zorubicin and Mitoxantrone; MDR inhibitors, such as verapamil; and Ca2+ ATPase inhibitors, such as thapsigargin.


The following examples are offered by way of illustration, not limitation. To this end, it should be noted that one or more hydrogen atoms may be omitted from the drawn structure consistent with accepted shorthand notation of such organic compounds, and that one skilled in the art would readily appreciate their presence.


Retention time data for the following examples was obtained by one of two methods detailed as follows:


Method A




  • Column: YMC Pro C-18, 3.0 μ spherical silica gel, 4.0×50 mm, pore size 120 Å.

  • Gradient: 0-10 min, 20% A-90% A linear binary gradient.

  • Flow rate: 2.0 mL/min.

  • Mobile Phase: A, 0.1% formic acid in acetonitrile; B, 0.1% trifluoroacetic acid in water.


    Method B

  • Column: YMC ODS-A, 5.0 μ spherical silica gel, 4.6×250 mm, pore size 120 Å.

  • Gradient: 0-10 min, 20% A -90% A linear binary gradient followed by 10-25 min, 100% A.

  • Flow rate: 1.0 mL/min.

  • Mobile Phase: A, 0.1% trifluoroacetic acid in acetonitrile; B, 0.1% trifluoroacetic acid in water.



EXAMPLES
Example 1
Synthesis of 4-{[4-(4-chlorophenyl)pyrimidin-2-yl]amino} benzamide



embedded image



(2E)-3-(Dimethylamino)-1-(4-chlorophenyl)prop-2-en-1-one


A solution of 1-(4-chlorophenyl)ethan-1-one (3.0 g, 19.3 mmol) and N,N, dimethylformamide diisopropylacetal (20 ml) was heated at 150° C. for 16 hours. The reaction mixture was cooled to 0° C. and treated with hexanes (20 ml). The resulting solid was collected via filtration and washed with hexanes to provide the title compound: EI-MS (m/z) 209 [M+1]+.


4-(4-Chlorophenyl)pyrimidine-2-thiol


To a solution of (2E)-3-(dimethylamino)-1-(4-chlorophenyl)prop-2-en-1-one (1.5 g, 7.2 mmol) in ethanol (25 ml) was added thiourea (0.60 g, 7.9 mmol) and potassium carbonate (K2CO3) (1.19 g. 8.63 mmol). The resulting suspension was heated to 85° C. for 12 hours then cooled to ambient temperature. The resulting solid was collected and thoroughly washed with water and hexanes to provide a beige solid: EI-MS (m/z) 222 [M+1]+.


4-(4-Chlorophenyl)-2-methylthiopyrimidine


4-(4-Chlorophenyl)pyrimidine-2-thiol (1.2 g, 5.39 mmol) was taken in 10 ml of an aqueous potassium hydroxide (0.453 g, 5.39 mmol) solution. Iodomethane (503 μl, 5.39 mmol) was added at ambient temperature and the reaction mixture was allowed to stir for 30 minutes. The resulting white solid was collected via filtration and washed with minimal water and hexanes to provide the title compound: EI-MS (m/z) 237 [M+1]+.


4-(4-chlorophenyll)-2-(methylsulfonyl)pyrimidine


To a solution of 4-(4-chlorophenyl)-2-methylthiopyrimidine (1.1 g, 4.65 mmol) in acetone (30 ml) and water (10 ml) was added oxone (7.14 g, 11.62 mmol). The reaction mixture was stirred for 18 hours then diluted with water and extracted into dichloromethane. The extracts were dried over magnesium sulfate, filtered and concentrated to provide a white solid: EI-MS (m/z) 269 [M+1]+.


4-{[4- (4-chlorophenyl)pyrimidin-2-yl]amino}benzamide


To a solution of 4-(4-chlorophenyl)-2-(methylsulfonyl)pyrimidine (0.10 g, 0.37 mmol) and 4-aminobenzamide in 2-propanol (3 ml) was heated to 120° C. in a sealed vessel for 14 hours. The crude material was concentrated and purified by preparative HPLC to provide the title compound as a beige solid: LC/MS Retention Time; 6.30 min (Method A), M+1; 325.


Example 2
Alternative Synthesis of 4-{[4-(4-chlorophenyl)pyrimidin-2-yl]amino}benzamide



embedded image



N-{(4-Aminocarbonyl)phenyl}guanidine nitrate


To a stirred suspension of 4-aminocarbonylaniline (20 g, 147 mmol) and cyanamide (14.2 g, 338 mmol) in 70 mL of ethanol was added concentrated nitric acid (20 mL) dropwise. The reaction mixture was heated at reflux overnight, and cooled. Volatile matters were evaporated to give a thick oil. The residue was taken up in methylene chloride and methanol to afford yellow solid. This material was filtered, washed with ether and water and dried in vacuo at 50° C. to afford the desired product (17.5 g, 66% yield): LC/MS Retention Time; 0.63 min (Method A), M+1; 179.


4-{[4-(4-Chlorophenyl)pyrimidin-2-yl]amino}benzamide


To a solution of (2E)-3-(dimethylamino)-1-(4-chlorophenyl)prop-2-en-1-one (0.10 g, 0.48 mmol), 4-(amidinoamino)benzamide nitrate (0.116 g, 0.48 mmol), and potassium carbonate (0.132 g, 0.96 mmol) in ethanol (10 ml) with was heated to 120° C. overnight in a sealed vessel. The reaction mixture was cooled to room temperature and the resulting solid was collected then washed with ethanol, water, and diethyl ether to provide the title compound as a beige solid, identical in all respects with the compound prepared in Example 1.


Example 3
Synthesis of Representative Compounds

The compounds listed below were prepared according to the procedure of Example 2 using the appropriate methylketone as the starting material.

CompoundMOL.RT,NumberStructureWEIGHTminM + 13-1 embedded image315.3355.673163-2 embedded image296.3535.532963-3 embedded image324.3145.933253-4 embedded image290.3255.772913-5 embedded image320.356.073213-6 embedded image279.3024.82803-7 embedded image464.9316.474.653-8 embedded image431.4745.534323-9 embedded image431.4745.584323-10embedded image449.5764.624503-11embedded image407.5394.624083-12embedded image462.6194.474633-13embedded image431.4745.534323-14embedded image380.475.553813-15embedded image412.4685.044133-16embedded image565.571.974523-17embedded image452.5375.484533-18embedded image390.3887.183913-19embedded image346.4327.433473-20embedded image398.4887.43993-21embedded image430.4866.644313-22embedded image369.2216.883693-23embedded image335.3655.83363-24embedded image321.3395.53223-25embedded image334.3814.043353-26embedded image373.4585.573743-27embedded image335.3225.873363-28embedded image362.4316.773633-29embedded image333.3935.073343-30embedded image375.435.473763-31embedded image359.2156.573593-32embedded image359.2156.473593-33embedded image374.3216.433753-34embedded image340.3846.333413-35embedded image411.4876.734123-36embedded image356.3874.273573-37embedded image338.7976.373393-38embedded image377.2056.503773-39embedded image393.666.673933-40embedded image334.3344.73353-41embedded image330.34611.1763313-42embedded image346.41310.2883473-43embedded image500.57710.48501.33-44embedded image467.539.956468.33-45embedded image468.51511.268469.33-46embedded image477.537212.74478.33-47embedded image443.548111.292444.63-48embedded image485.463811.396486.33-49embedded image486.5738.548487.33-50embedded image401.46779.6644023-51embedded image450.34288.684378.43-52embedded image469.464811.36470.33-53embedded image521.496812.204522.33-54embedded image501.530812.072502.33-55embedded image444.53628.696445.43-56embedded image500.34989.74428.43-57embedded image480.363811.084482.23-58embedded image457.574912.344458.33-59embedded image500.59989.924501.53-60embedded image368.822310.624369.23-61embedded image564.64286.49565.43-62embedded image415.494510.268416.33-63embedded image470.357912.05470.3


Example 4
Synthesis of 4-[(4-{4-[(4-chlorophenyl)sulfonyl]phenyl}pyrimidin-2-yl)amino]benzamide



embedded image


To a stirred solution of p-chlorobenzenethiol (1) (3.2 g, 0.022 mol) in DMF (40 mL) was added NaH (60% dispersion in mineral oil, 0.8 g). After the effervescence had ceased, p-chlorobenzenethiol (0.011 mol, 0.55 equiv) was added. The solution was then stirred at 110° C. for 3 h. The mixture was cooled to room temperature and then diluted with ether (150 mL). The ethereal suspension was washed with 5% NaOH (aq, 50 mL), 3% HCl (aq, 2×50 mL), filtered, and concentrated to afford 2.88 g of p-chlorophenylthioacetophenone (2) (100%). Biarylsulfide (2) was then dissolved in acetone/water (4:1, v/v, 100 mL). OXONE (13.5 g, 2.2 equiv) was added to the solution. The reaction was stirred 4 h at room temperature. After this time, the acetone was removed in vacuo. The mixture was diluted in ether (100 mL) and water (100 mL). The mixture was shaken and the layers separated. The ether layer was dried (MgSO4 ), filtered, and concentrated to afford 2.02 g (62%) of sulfone 3. Sulfone (3) was then dissolved in dimethylformamide dimethyl acetal (15 mL) and heated to 110° C. for 12 h. The reaction mixture was then concentrated to remove excess in dimethylformamide dimethyl acetal. A portion of the intermediate ene-amino ketone (0.38 g, 1.09 mmol) was taken up in ethanol (20 mL). To this solution was added K2CO3 (0.45 g, 3 equiv) and 4-guanadinobenzamide (4) (0.26 g, 1 equiv). The reaction mixture was heated in a sealed tube at 100° C. for 12 h. The mixture was then cooled to room temperature, diluted with water (30 mL), and then filtered. The solid was washed with water and ethanol. A portion of the material was purified by preparatory HPLC to afford 15 mg of the desired compound, which was found to be 100% pure by analytical HPLC. LCMS (M+H=465.0@ 6.47 min. (Method A)).


Example 5
Synthesis of 4-({4-[4-(4-pyridylsulfonyl)phenyl]pyrimidin-2-yl}amino)benzamide



embedded image


The above compound was made according to the procedure of Example 4 from 2-mercaptopyridine and the appropriate thiol as the starting materials. LCMS: (M+H=432.1, @5.50 min. (Method B)).


Example 6
Synthesis of 4-({4-[4-(2-pyridylsulfonyl)phenyl]}pyrimidin-2-yl}amino)benzamide



embedded image


The above compound was made according to the procedure of Example 4 from 2-mercaptopyridine and the appropriate thiol as the starting materials. LCMS (M+H=432.0@ 5.58 min. (Method B)).


Example 7
Synthesis of 4-({4-[4-(3-pyridylsulfonyl)phenyl]pyrimidin-2-yl}amino)benzamide



embedded image


The above compound was made according to the procedure of Example 4 from 3-mercaptopyridine and the appropriate thiol as the starting materials. LCMS (M+H=432.1@ 5.55 min. (Method B)).


Example 8
Synthesis of 4-({4-[4-(3-hydroxypropylthio)phenyl]pyrimidin-2-yl}amino)benzamide



embedded image


The above compound was made according to the procedure of Example 4 from 3-mercaptopropanol and the appropriate thiol as the starting materials. LCMS (M+H=381.0@ 5.55 min. (Method B)).


Example 9
Synthesis of 4-[(4-{4-[(3-hydroxypropyl)sulfonyl]phenyl}pyrimidin-2-yl)amino]benzamide



embedded image


To a solution of 3-mercaptopropanol (5 g, 0.054 mol) in DMF (40 mL) was added NaH (2.2 g, 60% dispersion in mineral oil). After the bubbling had ceased, p-chloroacetophenone (5.25 mL, 0.041 mol, 0.75 equiv) was added and the mixture was stirred at 100° C. for 3 h. The reaction was cooled, diluted with ether (200 mL), and washed with 5% HCl (aq) (2×30 mL), water (2×50 mL), and then brine (40 mL). The ether layer was dried (MgSO4 ), filtered, and concentrated to afford thioaryl ketone (5) (6.1 g, 0.29 mol, 72%). Ketone (5) (0.72 g, 3.4 mmol) was dissolved in acetone/water (4:1 v/v, 20 mL). OXONE® (4.2 g) was added and the mixture was stirred for 2 h. The mixture was then concentrated, diluted with ether (75 mL), washed with water (3×50 mL), and then brine (50 mL). The ether layer was then dried (MgSO4), filtered, and concentrated to afford to aryl sulfone (6). The title compound was prepared as previously described in Example 4 from ketone (6) to afford 39 mg (3%) of analytically pure material. LCMS: (M+H=413.0@5.04 min. (Method A)).


Example 10
Synthesis of 4-({4-[4-(3-morpholin-4-ylpropylthio)phenyl]pyrimidin-2-yl}amino)benzamide



embedded image


Acetophenone (5) was then taken up on toluene (50 mL). To this solution was added ethylene glycol (2.6 mL, 2 equiv) and p-toluenesulfonic acid (0.7 g). The reaction was refluxed with a Dean Stark trap for 2-3 h. After azeotropic removal of water, the reaction was cooled and then washed with 10% NaHCO3 (aq, 50 mL), water (50 mL), and brine (50 mL). The organic extract was dried (MgSO4), filtered, and concentrated. The crude acetal was then taken up in CH2CL2 (20 mL). In a separate flask, (COCl)2 (2.26 mL, 26.0 mmol) was dissolved in CH2CL2 (20 mL) and cooled to −78° C. DMSO (3.7 mL, 52.0 mmol) in CH2CL2 (5 mL) was then added to the cold solution dropwise. This mixture was stirred for 2 min, after which the crude acetal was added in CH2CL2 (20 mL). After stirring 15 min, Et3N (16.5 mL, 5 equiv) was added slowly. The resulting mixture was stirred 5 min, and then let warm to room temperature over 1 h. The mixture was then poured into a separatory funnel and washed with 5% NaHCO3 (100 mL). The organic layer was then washed with brine (50 mL), dried (Na2SO4), filtered, and concentrated to afford crude aldehyde (7). Aldehyde (7)(0.5 g) was then taken up in MeOH/AcOH (10 mL). To this solution was added morpholine (0.21 mL). The mixture was stirred 10 min, after which time NaBH3CN (0.19 g) was added. After 30 min, the reaction mixture was concentrated, basified with 3 M NaOH, and extracted with CH2CL2 (3×15 mL). The organic extracts were concentrated and then taken up in acetone/water (9:1 v/v, 20 mL). P-TsOH (0.1 g) was then added to the solution and the mixture was stirred 12 h. After this time, the mixture was concentrated, basified with 1 M NaOH, and extracted with CH2Cl2 (3×15 mL). The organic extracts were then dried (Na2SO4), filtered, and concentrated to afford crude aryl ketone (8), which was taken up in dimethylformamide dimethyl acetal (15 mL) and heated to 100° C. for 12 h. The mixture was then concentrated down and taken up in EtOH (15 mL). To this solution was added K2CO3 (0.31 g) and 4-guanadinobenzamide (4) (0.14). The reaction mixture was heated in a sealed tube at 100° C. for 12 h. The mixture was then cooled to room temperature, diluted with water (30 mL), and then filtered. The solid was washed with water and ethanol. The material was purified by preparatory HPLC to afford the titled compound (33 mg, 4%): LCMS 4.62 min. (Method A), M+H=450.


Example 11
Synthesis of 4-[(4-{4-[3-(dimethylamino)propylthio]phenyl}pyrimidin-2-yl)amino]benzamide



embedded image


The titled compound was prepared by the procedure of Example 10, except dimethylamine was used in place of morpholine during the reductive amination of aldehyde (7). LCMS (M+H=408.0@ 4.62 min. (Method B)).


Example 12
Synthesis of 4-[(4-{4-[3-(4-methylpiperazinyl)propylthio]phenyl}pyrimidin-2-yl)amino]benzamide



embedded image


The titled compound was prepared by the procedure of Example 10, except N-methylpiperizine was used in place of morpholine in the reductive amination of aldehyde (7). LCMS (M+H=463.0@ 4.47 min. (Method B)).


Example 13
Synthesis of 4-[4-{4-[(1-methyl-4-piperidyl)sulfonyl]phenyl}pyrimidin-2-yl)amino]benzamide



embedded image


4-mercaptopyridine (2.8 g, 25.0 mmol) was dissolved in DMF (25 mL). NaH (1 g, 60% dispersion in mineral oil) was then added to the solution. After the effervescence had ceased, p-chloroacetophenone (1.4 mL, 11 mmol) was added and the mixture was heated to 110° C. for 14 h. After this time, the mixture was cooled, diluted with ether (100 mL). The mixture was washed with 5% NaOH (2×50 mL), water (2×50 mL), and brine (50 mL). The ethereal extract was dried (MgSO4), filtered, and concentrated. The resulting oil was purified by flash chromatography (9:1 to 7:3 hexanes/ethyl acetate gradient). Concentration of the desired fractions afforded 1.37 g (54%) of thioacetophenone (9). Sulfide (9) (1.37 g) was then dissolved in acetone/water (9:1 v/v, 35 mL). To this solution was added OXONE® (7.4 g, 2 equiv). The mixture was stirred for 2 h. The mixture was then concentrated, neutralized with 10% NaHCO3, and extracted with CH2Cl2 (3×50 mL). The organic extracts were dried (Na2SO4), filtered, and concentrated to afford diarylsulfone (10) (1.25 g, 80%). Sulfone (10) (0.53 g. 2.0 mmol) was dissolved in THF (7 mL). To this solution was added Super Hydride® (6.3 mL, 1 M in THF) at room temperature. The solution was stirred at room temperature for 1 h, followed by quenching with MeOH (0.6 mL). The mixture was then concentrated. The residue was taken up in 1 N HCl (50 mL). The aqueous mixture was extracted with ether (3×50 mL). The organic layers were discarded. The aqueous layer was basified and extracted with CH2Cl2 (3×15 mL). The organic layers were concentrated. The residue was taken up in AcOH/MeOH (1:1 v/v, 10 mL). CH2O (37% aq, 1 mL) and NaBH3CN (0.1 g) were added. The mixture was stirred 30 min. The mixture was then concentrated, basified with 10% NaOH (aq) and extracted with CH2Cl2 (3×15 mL). The organic extracts were dried (Na2SO4), filtered, and concentrated to afford crude ketone (11). Aryl ketone (10) was refluxed in dimethylformamide dimethyl acetal (15 mL) and heated to 100° C. for 12 h. The mixture was then concentrated down and taken up in EtOH (15 mL). To this solution was added K2CO3 (0.31 g) and 4-guanadinobenzamide (4) (0.14 g). The reaction mixture was heated in a sealed tube at 100° C. for 12 h. The mixture was then cooled to room temperature, diluted with water (30 mL), and then filtered. The solid was washed with water and ethanol. The material was purified by preparatory HPLC to afford 6.0 mg (0.5% from sulfone (10)) of the title compound. LCMS (M+H=452@ 6.13 min. (Method A)).


Example 14
Synthesis of 4-[(4-{4-[(4-methylpiperazinyl)sulfonyl]phenyl}pyrimidin-2-yl)amino]benzamide



embedded image


N-Methylpiperizine (1.16 mL, 0.01 mol) was dissolved in CH2Cl2 (30 mL) and Et3N (4.4 mL, 0.033 mol). The solution was cooled to 0° C. and 4-acetylbenzenesulfonyl chloride (2.29 g, 0.01 mol) was added at once. The reaction was stirred for 15 min., poured into a separatory funnel, and extracted with water (3×20 mL) and then brine (10 mL). The organic layer was dried (Na2SO4), filtered, and concentrated to afford aryl ketone (12). Ketone (12) was carried on without purification to make the title compound as described in Example 13. An analytical sample was purified by preparatory HPLC (0.028 mg, 0.6%): LCMS (M+H=453.2@5.48 min. (Method A)).


Example 15
Synthesis of 4-{2-[(4-carbamoylphenyl)amino]pyrimidin-4-yl}benzoic acid



embedded image


A mixture of ethyl 4-acetylbenzoate (3.00 g, 15.62 mmol) and N,N-dimethylformamide dimethyl acetal (6.2 g, 52.10 mmol) was refluxed for 18 hours, cooled and concentrated to give ethyl 4-[(2E)-3-(dimethylamino)prop-2-enoyl]benzoate quantitatively. A solution of ethyl 4-[(2E)-3-(dimethylamino)prop-2-enoyl]benzoate, potassium carbonate (3.55 g, 25.74 mmol), and 4-(amidinoamino)benzamide (3.10 g, 12.87 mmol) in ETOH (120 mL) was refluxed for 18 hours. The mixture was cooled, filtered, and washed with ETOH, water, then ether respectively to give ethyl 4-{2-[(4-carbamoylphenyl)amino]pyrimidin-4-yl}benzoate (2.60 g, 46% yield). This compound was refluxed for 2 hours in ETOH (30 mL), water (20 mL), and NaOH (0.640 g, 16 mmol). The reaction mixture was cooled, acidified to pH 3, and filtered to give 1.00 gram (42% yield) of the titled compound. HPLC/ES-MS (20-100% acetonitrile): R.T. 4.7 min. (Method A); (m/z) 335 [M+1]+.


Example 16
Synthesis of (4-{[4-(4-chlorophenyl)pyrimidin-2-yl]amino}phenyl)-N,N-dimethyl carboxamide



embedded image



4-Guanidino-benzoic Acid Methyl Ester


To a stirred suspension of 4-guanidino benzoic acid (20.0 g, 93 mmol) in methanol (600 mL) was added thionyl chloride (12 mL) drop wise. The reaction mixture was stirred at room temperature overnight. The reaction was concentrated in vacuo to give a white powder. The crude material was dissolved in dichloromethane and evaporated to provide the title compound as a white powder (17.95 g, 100% yield): HPLC Retention Time; 1.27 min (Method A). M+1; 193.


(2E)-3-Dimethylamino-1-(4-chlorophenyl)prop-2-en-1-one


A solution of 1-(4-chlorophenyl)ethane-1-one (35.0 g, 226 mmol) and N,N Dimethylformamide diisopropylacetal (35 mL) was heated to reflux for 16 hours. The reaction mixture was cooled to room temperature and treated with hexanes (50 mL). The resulting solid was collected via filtration and washed with hexanes to provide the title compound as a yellow solid (47.12 g, 99% yield): HPLC Retention Time; 6.45 min (Method B). M+1; 209.


4-[4-(4-Cholorophenyl)-pyrimidin-2-ylamino]benzoic Acid


A Solution of 4-guanidino-benzoic acid methyl ester (17.95 g, 93 mmol), (2E) 3-dimethylamino-1-(4-chlorophenyl)prop-2-en-1-one (19.44 g, 93 mmol, and potassium carbonate (38.50 g, 279 mmol) in 1-propanol was heated to reflux for 24 hours. The reaction mixture was cooled to room temperature. The resulting solid was collected via filtration and washed with ethanol to provide the title compound which was used without further purification. EI MS(m/z) 339 [M+1]+. To a suspension of 4-[4-(4-chlorophenyl)-pyrimidin-2-ylamino]benzoic acid methyl ester in methanol (100 mL) was added 5N NaOH (100 mL). The reaction mixture was heated to reflux for 4 hours and then cooled to room temperature. The resulting solid was collected via filtration, washed with hexanes, and dried in vacuo to provide the title compound as a yellow solid (27.36 g, 100% yield): HPLC Retention Time; 7.29 min (Method A). M+1; 325.


(4-{[4-(4-Chlorophenyl)-pyrimidin-2-yl]amino}phenyl)-N,N-dimethyl carboxamide


To 4-{[4-(4-chlorophenyl)pyrimidin-2-yl]amino} benzoic acid (200 mg, 0.615 mmol) is added thionyl chloride (4 mL) along with a catalytic amount of DMF at room temperature. The resulting suspension is then refluxed for a period of 1 hour resulting in a clear pale yellow solution which was concentrated in vacuo. To the flask was then added a solution of dimethylamine (615 μL of a 2.0 M solution in THF, 1.23 mmol) and triethylamine (124 mg, 1.23 mmol) in tetrahydrofuran (4.5 mL). The solution was then stirred for 18 hours at room temperature, diluted with water (5 mL) and filtered. Purification of the remaining solid by preparative HPLC yielded the title compound. HPLC/ES-MS: RT 6.74 min. (Method A); (m/z) 353 [M+1]+.


Example 17
Synthesis of Further Representative Compounds



embedded image


Compounds listed below were prepared according to the above procedure:

CompoundMOL.RT,NumberStructureWEIGHTminM + 117-1embedded image366.857.0236717-2embedded image352.82367435317-3embedded image338.7976.4333917-4embedded image442.9487.9744317-5embedded image428.9217.8342917-6embedded image418.8577.5341917-7embedded image435.3127.8043617-8embedded image435.3127.8043617-9embedded image401.8556.8240217-10embedded image401.8556.8240217-11embedded image414.8947.6741517-12embedded image416.8666.8741717-13embedded image400.8677.5340117-14embedded image444.927.4044517-15embedded image430.8937.5043117-16embedded image460.9197.6046117-17embedded image443.9365.9744417-18embedded image397.2746.7739717-19embedded image429.9095.0743017-20embedded image408.8876.140917-21embedded image432.9134.5343317-22embedded image409.8755.5741017-23embedded image449.9834.7345017-24embedded image382.8496.1738317-25embedded image382.8496.138317-26embedded image382.8496.1738317-27embedded image408.8876.2840917-28embedded image394.865.8739517-29embedded image542.6175.954317-30embedded image594.6495.8659517-31embedded image408.5245.5840917-32embedded image548.7085.8954917-33embedded image491.6135.3249217-34embedded image543.6456.7354417-35embedded image421.9225.9242217-36embedded image493.9928.0449417-37embedded image449.93311.245017-38embedded image420.9227.742117-39embedded image414.8947.841517-40embedded image482.8918.148317-41embedded image442.9488.0744317-42embedded image493.79849517-43embedded image422.9578.442317-44embedded image406.9157.940717-45embedded image428.9217.842917-46embedded image458.9037.745917-47embedded image5086.250817-48embedded image456.9747.545717-49embedded image474.9466.747517-50embedded image467.9546.746817-51embedded image488.9737.648917-52embedded image550.8888.555117-53embedded image505.0187.850517-54embedded image449.945.945017-55embedded image420.9418.242117-56embedded image442.948844317-57embedded image432.9538.243317-58embedded image404.8557.540517-59embedded image482.8918.148317-60embedded image504.9717.650517-61embedded image432.8847.843317-62embedded image463.3668.146317-63embedded image428.9217.942917-64embedded image458.9037.846017-65embedded image472.937.847317-66embedded image420.9418.142117-67embedded image474.9467.847517-68embedded image483.7848.248317-69embedded image438.9137.843917-70embedded image432.8847.143317-71embedded image392.8887.839317-72embedded image396.8767.239717-73embedded image474.9467.847517-74embedded image463.3668.246317-75embedded image442.9488.144317-76embedded image444.927.844517-77embedded image428.9217.942917-78embedded image444.925.744517-79embedded image493.79849517-80embedded image446.9117.944717-81embedded image456.9748.245717-82embedded image460.9197.346117-83embedded image471.0018.547117-84embedded image511.788.251317-85embedded image463.366846317-86embedded image451.9555.945217-87embedded image420.9418.142117-88embedded image449.3397.944917-89embedded image472.937.847317-90embedded image521.1459.852117-91embedded image396.8326.339717-92embedded image481.9817.648217-93embedded image471.9897.747217-94embedded image366.856.636717-95embedded image500.8817.550117-96embedded image432.8847.143317-97embedded image438.9137.543917-98embedded image444.927.744517-99embedded image537.8437.453917-100embedded image428.9217.342917-101embedded image442.9487.444317-102embedded image420.9417.542117-103embedded image440.9327.344117-104embedded image451.9156.245317-105embedded image431.8814.943217-106embedded image396.8765.7139717-107embedded image422.9577.742317-108embedded image465.0388.646517-109embedded image483.7847.848317-110embedded image456.9747.745717-111embedded image456.9747.645717-112embedded image511.787.451317-113embedded image449.3397.444917-114embedded image483.7847.848517-115embedded image392.8887.139317-116embedded image446.9117.244717-117embedded image378.8616.837917-118embedded image429.9094.943017-119embedded image440.8926.544117-120embedded image408.8726.540917-121embedded image440.8926.444117-122embedded image415.8824.941617-123embedded image422.8986.642317-124embedded image439.9047.144017-125embedded image418.8827.241917-126embedded image364.8346.436517-127embedded image407.9034.840817-128embedded image528.0095.352817-129embedded image435.9136.843617-130embedded image492.027.449217-131embedded image421.8866.842217-132embedded image366.857.436717-133embedded image394.867.239517-134embedded image512.017.651217-135embedded image499.9997.850017-136embedded image516.9877.951517-137embedded image465.9397.446617-138embedded image407.8847.240817-139embedded image450.9247.445117-140embedded image468.9868.346917-141embedded image493.0087.149317-142embedded image437.9294.643817-143embedded image537.9718.353817-144embedded image390.8727.739117-145embedded image437.9294.643817-146embedded image465.0388.446517-147embedded image443.9366.344417-148embedded image470.9626.347317-149embedded image487.964848817-150embedded image486.0166.348617-151embedded image443.9366.344417-152embedded image435.9564.643617-153embedded image437.9724.743817-154embedded image409.9194.641017-155embedded image458.9477.436517-156embedded image364.8347.236517-157embedded image428.9217.942917-158embedded image469.974847017-159embedded image487.9456.348817-160embedded image449.945.845017-161embedded image484.9884.448517-162embedded image463.966646417-163embedded image449.945.845017-164embedded image464.9984.846517-165embedded image443.9365.644417-166embedded image349.787.335017-167embedded image422.91412.167423.017-168embedded image392.8886.983393.217-169embedded image476.0218.92476.217-170embedded image421.88610.436422.217-171embedded image461.9948.717462.217-172embedded image465.98228.45466.917-173embedded image407.9039.3840817-174embedded image449.98310.2745017-175embedded image421.939.3742217-176embedded image407.9039.3740817-177embedded image407.9039.4240817-178embedded image436.9019.0943717-179embedded image490.6298.0249117-180embedded image489.5978.1749017-181embedded image491.6138.4249217-182embedded image407.85910.2340817-183embedded image407.9039.4240817-184embedded image449.9411.0745017-185embedded image405.8879.340617-186embedded image435.9569.8643617-187embedded image476.02110.6647717-188embedded image421.929610.6342217-189embedded image469.973610.5747017-190embedded image421.929617-191embedded image491.03599.03491.317-192embedded image465.98229.88466.317-193embedded image461.994210.48462.317-194embedded image451.95549.7452.317-195embedded image451.95549.7452.317-196embedded image505.0627505.411.97617-197embedded image476.0214.82476.317-198embedded image481.9814.3548217-199embedded image465.9824.66466.317-200embedded image433.9414.5943417-201embedded image477.9934.63478.317-202embedded image479.0250.79479.317-203embedded image491.0363.53491.317-204embedded image478.9817.19479.417-205embedded image545.0156.86553.417-206embedded image556.0677.23556.417-207embedded image508.0197.9508.417-208embedded image574.3815.89465.417-209embedded image630.4443.56631.317-210embedded image614.4455.64505.417-211embedded image406.8715.86436.417-212embedded image477.9932478.57.58317-213embedded image492.028.05492.517-214embedded image476.0218.817476.517-215embedded image437.92438


Example 18
Synthesis of 4-{[4-(4-chlorophenyl)pyrimidin-2-yl]amino}benzoic acid piperazine amide hydrochloride



embedded image


Hydrogen chloride gas was bubbled slowly in a solution of tert-butyl 4-{[4-(4-chlorophenyl)pyrimidin-2-yl]amino}benzoic acid piperazine amide (3.0 g, 6.1 mmol) in acetic acid (61 mL) for 20 minutes. The solution was concentrated and dried on a vacuum pump to give 2.6 g (99%) of the title compound; ES-MS, m/z 394 (M+1)+ LC/MS Retention Time, 5.84 min. (Method A).


Example 19
Synthesis of 4-{[4-(4-chlorophenyl)pyrimidin-2-yl]amino}benzoic acid 4-ethyl piperazine amide



embedded image


A solution of 4-{[4-(4-chlorophenyl)pyrimidin-2-yl]amino}phenyl piperazine ketone (0.5 g, 1.54 mmol), N-ethylpiperazine (0.18 g, 1.54 mmol), 1-(3-dimethylaminopropyl)-3-ethyl carbodiimide hydrochloride (0.44 g, 2.31 mmol) and hydroxybenzotriazole (0.31 g, 2.31 mmol) in dimethylformamide (15 mL) was stirred for 18 h. Water (50 mL) was added and the solid was filtered. The solid was purified on preparatory HPLC (C-18 column, 30% acetonitrile to 100% acetonitrile in water—both containing 0.1% trifluoracetic acid) to give the titled compound, 0.27 g (42%) yield; ES-MS, m/z 422 (M+1)+ LC/MS Retention Time, 5.92 min. (Method A).


Example 20
Synthesis of 4-acylaminopiperidines



embedded image



4-Aminopiperidyl 4-{[4-(4-chlorophenyl)pyrimidin-2-yl]amino}phenyl Ketone Hydrochloride


(tert-Butoxy)-N-{1-[(4-{[4-(4-chlorophenyl)pyrimidin-2-yl]amino}phenyl)carbonyl](4-piperidyl)}carboxamide (4.00 g, 7.87 mmol) was stirred in 50 mL EtOH followed by addition of anhydrous HCl gas. The reaction was stirred for 30 min. then concentrated down to a residue. To this was added a small amount of EtOH followed by dilution with ether. A yellow solid formed which was filtered and dried to give 3.00 grams of 4-aminopiperidyl 4-{[4-(4-chlorophenyl)pyrimidin-2-yl]amino}phenyl ketone hydrochloride: HPLC Retention time; 5.89 min. (Method B) M+1; 408.4


N-{1-[(4-{[4-(4-Chlorophenyl)pyrimidin-2-yl]amino}phenyl)carbonyl]-4-piperidyl}acetamide


Stirred 4-aminopiperidyl 4-{[4-(4-chlorophenyl)pyrimidin-2-yl]amino}phenyl ketone hydrochloride (300 mg, 0.582 mmol) in 10 mL THF with triethylamine (0.293 mg, 2.91 mmol). Acetic anhydride (89 mg, 0.873 mmol) was added and the reaction was stirred for 40 minutes. The solution was concentrated down and purified by preparative HPLC to give N-{1-[(4-{[4-(4-chlorophenyl)pyrimidin-2-yl]amino}phenyl)carbonyl]-4-piperidyl}acetamide (0.120 g, 46% yield): HPLC Retention time; 6.92 min. (Method B) M+1; 450.4


Compounds listed below were prepared according to the above procedure.

CompoundNumberStructureMWRT, minM + 120-1embedded image449.946.92450.420-2embedded image531.0137.49531.420-3embedded image518.0397.6518.420-4embedded image521.0187.19521.420-5embedded image478.9817.18479.420-6embedded image479.9657.3480.220-7embedded image541.0527.68541.4


Example 21
Synthesis of Piperazineacetic Acid Amides



embedded image



Ethyl 2-{4-[(4-{[4(4-Chlorophenyl)pyrimin-2-yl]amino}phenyl)carbonyl]piperazinyl}acetate


4-{[4-(4-chlorophenyl)pyrimidin-2-yl]amino}benzoic acid (5 g, 15.3 mmol) was dissolved in dimethylformamide. The HOBT (2.82 g, 18.4 mmo)] and EDCI (3.53 g, 18.4 mmol) were then added. The reaction stirred for 15 minutes then ethyl-2-piperazinylacetate (2.14 mL, 18.4 mmol) was added. The reaction was stirred overnight at room temperature. Water (150 mL) was added. The solid was collected by filtration, and purified by silica-gel column chromatography (90% EtOAc/Hexane, Rt=0.25) to yield 4.3 g (45% yield) of ethyl 2-{4-[(4-{[4(4-chlorophenyl)pyrimin-2-yl]amino}phenyl)carbonyl]piperazinyl}acetate: HPLC Retention time; 9.932 min. (Method B) M+1;480.2


2-{4-[(4-{[4-(4-Chlorophenyl)pyrimidin-2-yl]amino}phenyl)carbonyl]piperazinyl}acetic Acid


To ethyl 2-{4-[(4-{[4(4-chlorophenyl)pyrimin-2-yl]amino}phenyl)carbonyl]piperazinyl}acetate (5.0 g, 15.3 mmol) was added ethanol (69 mL) and NaOH (1.14 g, 29.2 mmol, 4.1 eq) in 46 mL water. The reaction was heated at 75° C. for 1.5 hours. The reaction was acidified to pH=3, filtered, and dried, affording 4.3 g of the acid 2-{4-[(4-{[4-(4-chlorophenyl)pyrimidin-2-yl]amino}phenyl)carbonyl]piperazinyl}acetic acid (83.3%): HPLC Retention time; 9.260 min. (Method B) M+1; 452.3


2-{4-[(4-{[4-(4-Chlorophenyl)pyrimidin-2-yl]amino}phenyl)carbonyl]piperazinyl}N-ethylacetamide


2-{4-[(4-{[4-(4-Chlorophenyl)pyrimidin-2-yl]amino}phenyl)carbonyl]piperazinyl}acetic acid (0.200 g, 0.44 mmol) was dissolved in DMF then stirred for 15 minutes in ice-brine solution, then the HOBT (0.072 g, 0.53 mmol] then EDCI (0.102 g, 0.53 mmol) were added and stirred for another 30 minutes. Ethylamine (0.030 mL, 0.53 mmol) was added and the reaction was left to stir at room temp overnight. The reaction was quenched with 10 mL of water and a precipitate formed. The precipitate was colleted by filtration, and purified by preparative HPLC to yield 2-{4-[(4-{[4-(4-chlorophenyl)pyrimidin-2-yl]amino}phenyl)carbonyl]piperazinyl}N-ethylacetamide: HPLC Retention time; 9.508 min. (Method B) M+1; 479.2


Compounds listed below were prepared according to the above procedure.

CompundNumberStructureMWRT, minM + 121-1embedded image522.058.648522.321-2embedded image478.9819.508479.321-3embedded image493.0089.79493.221-4embedded image478.9819.472479.321-5embedded image464.9549.268465.321-6embedded image505.0199.676505.221-7embedded image450.9287.933451.021-8embedded image521.01819.644521.621-9embedded image579.9576.1507.4


Example 22
Reductive Amination



embedded image



4-{[4-(4-chlorophenyl)pyrimidin-2-yl]amino}phenyl 4-[(methylethyl)amino]piperidyl ketone hydrochloride


1-[(4-{[4-(4-chlorophenyl)pyrimidin-2yl]amino}phenyl)carbonyl]piperidin-4-one (400 mg, 0.980 mmol) was dissolved in 10 mL EtOH along with isopropylamine (58 mg, 0.980 mmol). Sodium cyanoborohydride (62 mg, 0.986 mmol) was added and the mixture was stirred at room temperature for 18 hours. The reaction was quenched with water, extracted with ethyl acetate followed by flash chromatography (EtOAc/MeOH; 90:10) to give a residue. This was taken up in ETOH saturated with HCl(g), diluted with ether, filtered to give 4-{[4-(4-chlorophenyl)pyrimidin-2-yl]amino}phenyl 4-[(methylethyl)amino]piperidyl ketone hydrochloride (0.150 g, 30% yield): HPLC Retention time; 6.02 min. (Method B) M+1; 450.4.


Compounds listed below were prepared according to the above procedure.

CompoundNumberStructureMWRT, minM + 122-1embedded image522.9056.02450.422-2embedded image490.047810.612490.322-3embedded image465.98229.644466.322-4embedded image465.98229.604466.322-5embedded image465.98229.52466.422-6embedded image465.98229.584466.422-7embedded image480.0099.604480.222-8embedded image519.08959.172519.422-9embedded image517.2865.89408.422-10embedded image588.40765.43479.422-11embedded image451.95546.12452.422-12embedded image480.0099.291480.422-13embedded image447.96749.976448.4


Example 23
Synthesis of Reverse Sulfonamides



embedded image



(2E)-1-(4-nitrophenyl)-3-dimethylamino)prop-2-en-1-one


A mixture of 4-nitroacetophenone (20.0 g, 121 mmol) and N,N-dimethylformamide dimethylacetal (200 ml) was refluxed for 18 hours, cooled and concentrated to give (2E)-1-(4-nitrophenyl)-3-dimethylamino)prop-2-en-1-one quantitatively.


1-Acetyl-4-[(4-{[4-(4-nitrophenyl)pyrimidin-2-yl}amino}phenyl)carbonyl}piperazine


To a mixture of (2E)-1-(4-nitrophenyl)-3-dimethylamino)prop-2-en-1-one (250 mg, 1.14 mmol) and {4-{(4-acetylpiperazinyl)carbonyl]phenyl}aminocarboxamidine (394 mg, 1.36 mmol) in methanol (6 ml) is added 2 mL of a 2.0M solution of sodium methoxide in methanol. The reaction mixture is then refluxed for 18 hours then acidified to pH ˜4 using 1N HCl. The solid which formed at this time was then flittered and purified by column chromatography using 10% methanol in chloroform to give 320 mg (69%) of the desired product.


1-Acetyl-4-[(4-{[4-(4-aminophenyl)pyrimidin-2-yl}amino}phenyl)carbonyl}piperazine


To a solution of 1-acetyl-4-[(4-{[4-(4-nitrophenyl)pyrimidin-2-yl}amino}phenyl)carbonyl}piperazine (150 mg, 0.34 mmol) in methanol (5 mL) containing a few drops of acetic acid, is added 100 mg of 10% Palladium-Charcoal. The solution is then hydrogenated at 50 psi for 6 h at which time there remains no starting material. The solution is then filtered through a pad of Celite which gives 135 mg (95%) of essentially pure reduced material as a brown oil.


1-Acetyl-4-{[4-({4-[4-(phenylsulfonyl)aminophenyl]pyrimidin-2-yl}amino)phenyl]carbonyl}piperazine


To a solution of 1-acetyl-4-[(4-{[4-(4-aminophenyl)pyrimidin-2-yl}amino}phenyl)carbonyl}piperazine (100 mg, 0.24 mmol) in pyridine (5 mL) containing a catalytic amount of DMAP is added benzenesulfonyl chloride (50 mg, 0.29 mmol) and the solution is stirred overnight at room temperature. The pyridine is removed under vacuum and the residue extracted into methylene chloride and washed with 1N HCl. Evaporation of solvent provides the crude piperazine which is purified by preparative HPLC (10-60% CH3CN over 25 min.)to give an analytically pure sample as a yellow solid: M+1; 557.3. HPLC Retention Time; 9.59 min (Method B).


Compounds listed below were prepared according to the above procedure.

CompoundNumberStructureMWRT, minM + 123-1embedded image5868.03587.323-2embedded image624.64139.53625.323-3embedded image570.6718.46571.323-4embedded image586.679587.523-5embedded image556.6449.62557.323-6embedded image494.57348.35495.323-7embedded image591.089310.14591.323-8embedded image598.724610.25599.523-9embedded image624.641310.58625.323-10embedded image562.67249.56563.323-11embedded image570.67110.02571.323-12embedded image570.6719.79571.323-13embedded image601.64137.15602.523-14embedded image601.64138.57602.323-15embedded image614.72368.23615.523-16embedded image514.60744.55515.323-17embedded image523.61518.85524.323-18embedded image586.679.72587.323-19embedded image570.6719.82571.323-20embedded image570.67110.68571.523-21embedded image520.59029.89521.323-22embedded image535.60517.58536.323-23embedded image582.6829.18583.523-24embedded image596.70889.76597.523-25embedded image637.71799.8638.323-26embedded image623.69119.2624.523-27embedded image528.63425.92529.3


Example 24
Synthesis of Further Representative Compounds



embedded image


The compounds of Example 18, with the desired R1 moiety, may be modified according to the above procedures to yield further representative compounds of this invention. For example, the following compounds were made according to the above procedures.

CompoundNumberStructureMWRT, minM + 124-1embedded image498.9639.749924-2embedded image471.9677.1947224-3embedded image512.9906.2451324-4embedded image478.9745.9247924-5embedded image497.9757.4149824-6embedded image526.0377.6652624-7embedded image512.99858.350513.424-8embedded image478.98137.533479.424-9embedded image552.0287.33552.324-10embedded image559.0487.17559.324-11embedded image585.925.15513.324-12embedded image585.924.7851324-13embedded image516.9876.43517.324-14embedded image477.9936.95478.324-15embedded image489.8837.12490.324-16embedded image504.0126.77504.324-17embedded image490.0047.2504.324-18embedded image475.9776.58476.324-19embedded image476.9385.55479.324-20embedded image533.0734.63533.324-21embedded image506.9911.1507.324-22embedded image507.0354.61508.324-23embedded image465.9395.99466.324-24embedded image461.9516.41462.324-25embedded image482.0066.57496.324-26embedded image492.027.14492.324-27embedded image503.916.69504.324-28embedded image548.0437.27548.324-29embedded image565.935.99493.424-30embedded image476.9667.16477.424-31embedded image648.9938.56649.424-32embedded image449.946.92450.424-33embedded image464.9546.09465.324-34embedded image519.0466.87519.324-35embedded image522.997.19524.424-36embedded image537.0174.52537.424-37embedded image537.0217.79537.224-38embedded image504.9756.72505.424-39embedded image486.9616.92487.424-40embedded image487.9496.08488.424-41embedded image486.9617.27487.424-42embedded image502.967.27503.424-43embedded image502.95977.27503.424-44embedded image533.05357.19533.224-45embedded image488.93297.09489.424-46embedded image588.40763.25478.324-47embedded image515.01437.16515.4


Example 25
Synthesis of Sulfides



embedded image



3-Dimethylamino-1-[4-(4-hydroxybutylsulfanyl)phenyl]propenone


To a stirred solution of 4-hydroxybutanethiol (5.0 g, 47 mmol) in DMF (100 mL) was added NaH (60% dispersion in mineral oil, 2.1 g). After the effervescence had ceased, p-chloroacetophenone (4.3 mL, 33 mmol) was added. The solution was then stirred at 110° C. for 3 h. The mixture was cooled to RT and then diluted with ether (200 mL). The ethereal suspension was washed with 5% HCl (aq, 2×100 mL), water (100 mL), and then brine (50 mL). The ether extract was dried (MgSO4), filtered and concentrated to afford crude 1-[4-(4-hydroxybutylsulfanyl)phenyl]ethanone, which was used without purification. 1-[4-(4-hydroxybutylsulfanyl)phenyl]ethanone was taken up in dimethylformamide dimethylacetal (100 mL) and stirred at reflux for 12 h. The mixture was cooled and then concentrated to about one half of the original volume. Hexane was added to precipitate 3-Dimethylamino-1-[4-(4-hydroxybutylsulfanyl)phenyl]propenone. The mixture was filtered, washed with hexanes (50 mL), and dried to afford 3-Dimethylamino-1-[4-(4-hydroxy-butylsulfanyl)phenyl]propenone (6.4 g, 23 mmol): HPLC Retention Time; 5.58 min. (Method B) M+1; 279.8.


4-{4-[4-(4-Hydroxybutylsulfanyl)phenyl]pyrimidin-2-ylamino}benzoic Acid


3-Dimethylamino-1-[4-(4-hydroxybutylsulfanyl)-phenyl]propenone (6.4 g, 23 mmol) was, then taken up in nPrOH (150 mL). To this solution was added 4-guanidinobenzoic acid, methyl ester, hydrochloride salt (1.1 equiv, 5.4 g) and K2CO3 (3 equiv, 9.5 g). The mixture was stirred at reflux for 24 h. After this time, 10% NaOH (aq, 50 mL) was added, and the mixture was stirred at reflux for another 1 h. The mixture was then cooled to RT and concentrated to about half of the original volume. The pH of the mixture was then adjusted to pH 4-5 to 4-{4-[4-(4-Hydroxybutylsulfanyl)phenyl]pyrimidin-2-ylamino}benzoic acid. The acid was immediately filtered and washed with water (50 mL), cold EtOH (50 mL), and then dried (8.6 g, 21 mmol, 88%): HPLC Retention Time; 6.37 min. (Method B) M+1; 396.0.


[4-(Furan-2-carbonyl)piperazin-1-yl]-(4-{4-[4-(4-hydroxybutylsulfanyl)phenyl]pyrimidin-2-ylamino}phenyl)methanone


4-{4-[4-(4-Hydroxybutylsulfanyl)phenyl]pyrimidin-2-ylamino}benzoic acid (0.34 g, 0.86 mmol) was dissolved in THF (5 mL). To this solution was added 1-furoylpiperazine (0.170 g), EDCI (0.180 g), and HOBt (0.127 g). The mixture was stirred 12 h. The mixture was then diluted with CH2Cl2 (20 mL) and washed with 2% NaOH (aq, 30 mL), water (30 mL), and then brine (30 mL). The organic layer was dried (Na2SO4), filtered, and concentrated. The crude solid was subjected to preparatory HPLC (30-80 acetonitrile/water gradient, 20 min). The desired fractions were concentrated to remove most of the acetonitrile, and then the aqueous mixture was extracted with CH2Cl2/2% NaOH (aq). The organic layer was dried (Na2SO4), filtered, and concentrated to afford [4-(Furan-2-carbonyl)-piperazin-1-yl]-(4-{4-[4-(4-hydroxybutylsulfanyl)phenyl]pyrimidin-2-ylamino}phenyl)methanone (0.042 g, 9%): HPLC Retention Time; 10.07 min. (Method B) M+H=558.3.


Compounds listed below were prepared according to the above procedure.

CompoundNumberStructureMWRT, minM + 125-1embedded image557.67210.07558.325-2embedded image505.649.26506.325-3embedded image562.7358.81563.325-4embedded image500.0648.37464.425-5embedded image571.69912.04572.325-6embedded image519.66711.13520.325-7embedded image576.76210.24577.225-8embedded image514.0919.7478.325-9embedded image529.6189.5530.325-10embedded image477.5868.66478.225-11embedded image534.6827.32535.325-12embedded image472.016.88436.225-13embedded image571.69910.62572.325-14embedded image519.6679.76520.225-15embedded image477.638.77478.325-16embedded image491.6578.9492.325-17embedded image576.7629.25577.325-18embedded image492.6419.59493.325-19embedded image562.7798.42563.325-20embedded image588.7738.51589.325-21embedded image571.69910.85572.325-22embedded image519.66710.05520.325-23embedded image477.639478.325-24embedded image576.7629.46577.325-25embedded image491.6579.1492.325-26embedded image562.7798.58563.325-27embedded image588.7739.39589.525-28embedded image492.6429.84493.3


Example 26
Synthesis of Sulfonamides



embedded image



1-[4-(Morpholine-4-sulfonyl)phenyl]ethanone


To a suspension of 4-acetylbenzenesulfonyl chloride (5.5 g, 25 mmol) in CH2Cl2 (75 mL) and Et3N (2 equiv, 7.0 mL, 50 mmol) was added morpholine (1.5 equiv, 3.3 mL, 38 mmol) dropwise. The mixture was stirred at room temperature for 30 min. The mixture was then diluted with CH2Cl2 (100 mL) and washed with 5% HCl (2×50 mL), water (50 mL), and then brine (50 mL). The organic layer was dried (Na2SO4), filtered, and concentrated to afford crude 1-[4-(morpholine-4-sulfonyl)phenyl]ethanone (2) (4.78 g, 18 mmol, 71%): HPLC Retention Time; 5.82 min. (Method B) M+1, 270.0.


4-{4-[4-(Morpholine-4-sulfonyl)-phenyl]-pyrimidin-2-ylamino}benzoic Acid


Crude 1-[4-(morpholine-4-sulfonyl)phenyl]ethanone (4.78 g, 18 mmol) was suspended in dimethyformamide dimethylacetal (50 mL) and refluxed for 12 h. The reaction was allowed to cool and the mixture was concentrated to about half of the original volume. The solution was then titurated with hexanes to precipitate the eneamino ketone intermediate. The eneamino ketone was filtered and washed with hexanes (2×50 mL), dried under vacuum, and then taken up in nPrOH (150 mL). To this solution was added added 4-guanidinobenzoic acid, methyl ester, hydrochloride salt (1.1 equiv, 3.7 g) and K2CO3 (3 equiv, 6.4 g). The mixture was stirred at reflux for 24 h. After this time, 10% NaOH (aq, 50 mL) was added, and the mixture was stirred at reflux for another 1 h. The mixture was then cooled to RT and concentrated to about half of the original volume. The pH of the mixture was then adjusted to pH 4-5 to precipitate the acid. 4-{4-[4-(morpholine-4-sulfonyl)phenyl]pyrimidin-2-ylamino}benzoic acid was immediately filtered and washed with water (50 mL), cold EtOH (50 mL), and then dried (4.6 g, 10.5 mmol, 68%): HPLC Retention Time; 6.6 min. (Method B) M+1, 441.0.


[4-(Furan-2-carbonyl)-piperazin-1-yl](4-{4-[4-(morpholine-4-sulfonyl)phenyl]pyrimidin-2-ylamino}phenyl)methanone


4-{4-[4-(Morpholine-4-sulfonyl)-phenyl]-pyrimidin-2-ylamino}-benzoic acid (0.25 g, 0.57 mmol) was dissolved in THF (5 mL). To this solution was added 1-furoylpiperazine (0.123 g), EDCI (0.131 g), and HOBt (0.092 g). The mixture was stirred 12 h. The mixture was then diluted with CH2Cl2 (20 mL) and washed with 2% NaOH (aq, 30 mL), water (30 mL), and then brine (30 mL). The organic layer was dried (Na2SO4), filtered, and concentrated. The crude solid was subjected to preparatory HPLC (20-70 acetonitrile/water gradient, 20 min). The desired fractions were concentrated to remove most of the acetonitrile, and then the aqueous mixture was extracted with CH2Cl2/2% NaOH (aq). The organic layer was dried (Na2SO4), filtered, and concentrated to afford [4-(furan-2-carbonyl)piperazin-1-yl](4-{4-[4-(morpholine-4-sulfonyl)-phenyl]pyrimidin-2-ylamino}phenyl)methanone (0.177 g, 52%): HPLC Retention Time; 9.62 min. (Method B) M+H=603.3


Compounds listed below were prepared according to the above procedure.

CompoundNumberStructureMWRT, minM + 126-1 embedded image602.6699.62603.326-2 embedded image550.6378.88551.326-3 embedded image508.67.6509.326-4 embedded image607.7328.34608.326-5 embedded image522.6277.9523.326-6 embedded image593.7496.33594.326-7 embedded image619.7438.28620.326-8 embedded image523.6118.76524.326-9 embedded image576.7188.21577.326-10embedded image576.67510.26577.326-11embedded image592.71712.12593.326-12embedded image564.66410.04565.326-13embedded image578.69110.51579.326-14embedded image631.71110.33632.426-15embedded image466.56310.4467.326-16embedded image508.611.35509.326-17embedded image560.63212561.326-18embedded image616.6969.72617.326-19embedded image564.6648.93565.526-20embedded image522.6277.99523.326-21embedded image590.7458.34591.326-22embedded image563.67978.05564.326-23embedded image591.68979.01592.326-24embedded image619.74339.25620.326-25embedded image548.664810.88549.526-26embedded image534.63810535.326-27embedded image552.65286.82553.326-28embedded image522.62710.18523.326-29embedded image617.77118.31618.526-30embedded image556.644210.29557.226-31embedded image494.57348.96495.326-32embedded image562.691611.36563.426-33embedded image562.691611.2563.426-34embedded image562.691611.52563.426-35embedded image562.691611.5563.426-36embedded image564.66389.14565.426-37embedded image549.65298.04550.426-38embedded image565.65198.26566.326-39embedded image538.6269.14539.326-40embedded image551.66877.77552.326-41embedded image506.6289.64507.426-42embedded image492.60129.08493.426-43embedded image534.68169.9535.326-44embedded image591.77699.16592.526-45embedded image578.734210.25579.526-46embedded image520.65489.32521.526-47embedded image564.70749.7565.526-48embedded image577.75018.66578.526-49embedded image563.72338.77564.526-50embedded image577.75019.28578.526-51embedded image536.65388.89537.526-52embedded image580.70649.29581.426-53embedded image579.72238.4580.526-54embedded image538.66299.44539.326-55embedded image494.6179.06495.326-56embedded image537.68558.56538.526-57embedded image551.71238.47552.526-58embedded image536.653810.6453726-59embedded image570.67110.6357126-60embedded image576.718411.4357726-61embedded image596.705410.0159726-62embedded image550.680611.7555126-63embedded image564.707411.8256526-64embedded image571.65918.1157226-65embedded image536.653810.2853726-66embedded image536.653810.2453726-67embedded image579.67878.7158026-68embedded image591.089311.0759126-69embedded image562.691610.956326-70embedded image560.632210.74561


Example 27
Synthesis of Sulfones



embedded image



1-[4-(Tetrahydropyran-4-sulfanyl)phenyl]ethanone


To a stirred solution of Na2S (17.4 g, 0.22 mol) in water (26 mL) was added CS2 (14.7 mL, 0.24 mol). The mixture was stirred at 60-70° C. for 6 h. To the resultant red solution of Na2CS3 was added 4-chlorotetrahydropyran (0.074 mol). The mixture was stirred for 12 h at 60-70° C. The mixture was then cooled to ˜10° C. H2SO4 (conc.) was added to the mixture dropwise with stirring until a cloudy yellow color persisted. The mixture was then extracted with CH2Cl2 (3×50 mL). The aqueous layer was discarded and the CH2Cl2 layer was dried (Na2SO4), filtered, and concentrated. The crude thiol (47.5 mmol, ˜64%) was dissolved in DMF (100 mL) and treated with NaH (1.9 g, 48 mmol). After the effervescence had ceased, p-chloroacetophenone (4.3 mL, 33 mmol) was added. The solution was then stirred at 110° C. for 3 h. The mixture was cooled to RT and then diluted with ether (200 mL). The ethereal suspension was washed with 5% HCl (aq, 2×100 mL), water (100 mL), and then brine (50 mL). The ether extract was dried (MgSO4), filtered and concentrated to afford crude 1-[4-(tetrahydro-pyran-4-sulfanyl)-phenyl]-ethanone 1, which was purified by chromatography (SiO2, 9:1 hex/EtOAc) to afford pure 1-[4-(tetrahydropyran-4-sulfanyl)phenyl]ethanone 1 (7.4 mmol, 16% from 4-chlorotetrahydropyran): HPLC Retention Time; 5.41 min. (Method B) M+1; 269.0.


3-Dimethylamino-1-[4-(tetrahydropyran-4-sulfonyl)phenyl]propenone


1-[4-(Tetrahydro-pyran-4-sulfanyl)-phenyl]-ethanone 1 (7.4 mmol) was dissolved in acetone/water (9:1 v/v, 100 mL). Oxone® (2.1 equiv, 9.1 g) was added to the solution. The reaction was stirred at room temperature for 5 h. The mixture was filtered and the majority of acetone was removed in vacuo. The solution was then diluted with water (50 mL) and extracted with CH2Cl2 (3×50 mL). The organic layer was dried (Na2SO4), filtered, and concentrated to afford the intermediate tetrahydropyranyl sulfone, which was taken up in dimethylformamide dimethylacetal (100 mL) and stirred at reflux for 12 h. The mixture was cooled and then concentrated to about one half of the original volume. Hexane was added to precipitate eneamino ketone intermediate. The mixture was filtered, washed with hexanes (50 mL), and dried to afford 3-dimethylamino-1-[4-(tetrahydro-pyran-4-sulfonyl)-phenyl]-propenone (2.2 g, 7 mmol): HPLC Retention Time; 5.18 min. (Method B) M+1; 324.0.


4-{4-[4-(Tetrahydropyran-4-sulfonyl)-phenyl]pyrimidin-2-ylamino}benzoic Acid


3-Dimethylamino-1-[4-(tetrahydro-pyran-4-sulfonyl)-phenyl]-propenone was then taken up in nPrOH (80 mL). To this solution was added 4-guanidinobenzoic acid, methyl ester, hydrochloride salt (1.1 equiv, 1.7 g) and K2CO3 (3 equiv, 2.9 g). The mixture was stirred at reflux for 24 h. After this time, 10% NaOH (aq, 50 mL) was added, and the mixture was stirred at reflux for another 1 h. The mixture was then cooled to RT and concentrated to about half of the original volume. The pH of the mixture was then adjusted to pH 4-5 to precipitate 4-{4-[4-(tetrahydro-pyran-4-sulfonyl)-phenyl]-pyrimidin-2-ylamino}-benzoic acid 4. The acid was immediately filtered and washed with water (50 mL), cold EtOH (50 mL), and then dried (2.4 g, 5.5 mmol, 79% yield): HPLC Retention Time; 6.07 min. (Method B) M+1; 593.3.


[4-(3-Dimethylamino-propyl)-piperazin-1-yl]-(4-{4-[4-(tetrahydropyran-4-sulfonyl)phenyl]pyrimidin-2-ylamino}phenyl)methanone


4-{4-[4-(Tetrahydropyran-4-sulfonyl)-phenyl]pyrimidin-2-ylamino}benzoic acid 4 (0.26 g, 0.6 mmol) was dissolved in THF (5 mL). To this solution was added 1-(N,N-dimethylaminopropyl)piperazine (0.130 g), EDCI (0.136 g), and HOBt (0.096 g). The mixture was stirred 12 h. The mixture was then diluted with CH2Cl2 (20 mL) and washed with 2% NaOH (aq, 30 mL), water (30 mL), and then brine (30 mL). The organic layer was dried (Na2SO4), filtered, and concentrated. The crude solid was subjected to preparative HPLC (20-70 acetonitrile/water gradient, 20 min). The desired fractions were concentrated to remove most of the acetonitrile, and then the aqueous mixture was extracted with CH2Cl2/2% NaOH (aq). The organic layer was dried (Na2SO4), filtered, and concentrated to afford [4-(3-dimethylamino-propyl)piperazin-1-yl]-(4-{4-[4-(tetrahydropyran-4-sulfonyl)phenyl]pyrimidin-2-ylamino}phenyl)methanone 5 (0.079 g, 22%): HPLC Retention Time; 7.93 min. (Method B) M+1=593.3


Compounds listed below were prepared according to the above procedure.

CompoundNumberStructureMWRT, minM + 127-1 embedded image612.66410.25595.327-2 embedded image542.6178.7543.327-3 embedded image515.5918.57516.327-4 embedded image623.69119.36624.327-4 embedded image601.68110.06602.427-5 embedded image606.7448.64607.427-6 embedded image507.6128.37508.327-7 embedded image521.6398.57522.327-8 embedded image592.7617.93593.327-9 embedded image575.738.57576.327-10embedded image522.6238.95523.327-11embedded image630.72310.25631.327-12embedded image549.6499.555027-13embedded image500.58068.8501.327-14embedded image571.6999.78572.327-15embedded image583.719.736584.527-16embedded image541.62910.484542.327-17embedded image593.66111.264594.327-18embedded image513.6199.336514.327-19embedded image572.5149.20450027-20embedded image584.7418.692585.227-21embedded image528.6310.648529.227-22embedded image458.5411.44458.9


Example 28
Activity of Representative Compounds

The compounds of this invention may be assayed for IKK-2 inhibitory activity according to the following procedures.


IKK-2 Enzyme Assay


To 10 μl of the test compound in 20% DMSO in “Dilution Buffer” (20 mM HEPES pH 7.6, 0.1 mM EDTA, 2.5 mM MgCl2, 0.004% Triton X100, 2 μg/ml Leupeptin, 20 mM β-glycero-phosphate, 0.1 mM Na3VO4, 2 mM DTT) is added 30 μl of 167 μg/ml GST-IκBα in “HBB” (20 mM HEPES pH 7.6, 50 mM NaCl, 0.1 mM EDTA, 2.5 mM MgCl2, 0.05% Triton X100) and 30 μl IKK2EE(his6) at 1.33 μg/ml (40 ng/well). The mixture is preincubated for 15 minutes at room temperature. Then 30 μl of “Kinase Buffer” (20 mM HEPES pH 7.6, 6.67 mM MgCl2, 6.67 mM MnCl2, 0.02% Triton X100, 20 mM β-glycerolphosphate, 2 mM NaF, 2 mM DTT, 2 mM benzamidine, 16 mM para-nitrophenylphosphate, 5 μM ATP, 16.67 μCi/ml γ33P-ATP) is added and the reaction is allowed to proceed for 1 hour at room temperature. The IκBα is precipitated and phosphorylation terminated by addition of 150 μl 12.5% trichloroacetic acid. After 30 minutes the precipitate is harvested onto a filter plate to which 50 μl of scintillation fluid is added and then quantified by a scintillation counter. The IC50 values are calculated as the extrapolated concentration of the test compound at which the IκBα phosphorylation was reduced to 50% of the control value.


Detection of IκBα Degradation


Human umbilical vein endothelial cells (HUVEC) are cultured to 80% confluency and then pre-treated with compound (30 μM) at a final concentration of 0.5% DMSO. After 30 minutes, cells are stimulated with TNFα (30 ng/ml) for 20 minutes. Cells are washed, scraped from the plate, lyzed with 2× Laemmli buffer and heated to 100° C. for 5 minutes. Whole cell lysate (approx. 30 μg) is fractionated on Tris-glycine buffered 10% SDS-polyacrylamide gels (Novex, San Diego, Calif.) and transferred to nitrocellulose membrane (Amersham, Piscataway, N.J.). Membranes are blocked with 5% non-fat milk powder (BioRad, Hercules, Calif.) and incubated with antibody to IκBα (0.2 ug/ml #sc-371) (Santa Cruz Biotechnology, Santa Cruz, Calif.) and then donkey anti-rabbit horse radish peroxidase conjugated antibody (1:2500) (Amersham) in phosphate buffered saline with 0.1% Tween-20 and 5% non-fat milk powder. Immunoreactive proteins are detected with chemiluminescence and autoradiography (Amersham).


Inhibition of Cell Adhesion Molecule Expression


Enzyme Linked Immunosorbent Assay (ELISA) to determine endothelial cell adhesion molecule expression is performed as described by (Bennett et al., J. Biol Chem. 272:10212-12219, 1997). Briefly, HUVEC are plated in 96 well microtiter plates and grown to confluence. Cells are pre-treated with compound (30 μM) at a final concentration of 0.5% DMSO. After 30 minutes, cells are stimulated with TNFα (30 ng/ml) for 5 hours. Following experimental treatment, cells are washed once with phosphate buffered saline (PBS) and incubated with freshly prepared 4% paraformaldehyde solution, pH 7, for 60 min. Plates are then washed once with PBS, blocked overnight at 4° C. with 2% bovine serum albumin (BSA) in PBS, washed once with PBS and incubated with 1 μg/ml primary antibody in 0.1% BSA in PBS at 37° C. for 2 hours. Monoclonal antibodies used are to E-selectin (BBA16; R&D Systems, Minneapolis, Minn.), VCAM-1 (MA10620; Endogen, Woburn, Mass.), ICAM-1 (BBA3; R&D Systems), and ICAM-2 (AHT0201; Biosource, Camarillo, Calif.). After incubation with primary antibody, the cells are washed three times with 0.05% Tween-20 in PBS, incubated with alkaline phosphatase-conjugated goat anti-mouse IgG (AMI3405; Biosource) in 0.1% BSA in PBS at 37° C. for 1 hour, washed three times with 0.05% Tween-20 in PBS and once with PBS. The cells are then incubated in chromogenic substrate (1 mg/ml ρ-nitrophenyl phosphate in 1 M diethanolamine, 0.5 mM MgCl2, pH 9.8) at 37° C. for 30 min and absorbance measured at 405 nm using a ThermoMax microplate reader (Molecular Devices, Menlo Park, Calif.).


Rat in Vivo LPS-Induced TNF-α Production Assay


Male CD rats procured from Charlese River Laboratories at 7 weeks of age are allowed to acclimate for one week prior to use. A lateral tail vein is cannulated percutaneously with a 22-gage over-the-needle catheter under brief isoflurane anesthesia. Rats are administered test compound either by intravenous injection via the tail vein catheter or oral gavage 15 to 180 min prior to injection of 0.05 mg/kg LPS (E. Coli 055:B5). Catheters are flushed with 2.5 mL/kg of normal injectable saline. Blood is collected via cardiac puncture 90 minutes After LPS challenge. Plasma is prepared using lithium heparin separation tubes and frozen at −80° C. until analyzed. TNF-α levels are determined using a rat specific TNF-α ELISA kit (Biosource). The ED50 values are calculated as the dose of the test compound at which the TNF-α production is reduced to 50% of the control value. Preferred compounds of the present invention have an ED50 value ranging 1-30 mg/kg in this assay.


Example 29
Activity of Representative Compounds

Representative compounds of this invention may be assayed for their ability to inhibit IKK-2 by the assays set forth in Example 21. In this regard, preferred compounds of this invention have an IC50 value in the IKK-2 Enzyme Assay of Example 21 of 1 μM or less. To this end, preferred compounds of this invention are 1, 3-8, 3-9, 3-13, 3-14, 3-15, 3-21, 3-34, 17-2, 17-3, 17-18, 17-20, 17-21, 17-22, 17-23, 17-25, 17-27, 17-28, 17-29, 17-30, 17-31, 17-32, 17-33, 17-34, 17-35, 17-36, 17-54, 17-71, 17-72, 17-86, 17-91, 17-118, 17-127, 17-128, 17-129, 17-131, 17-132, 17-133, 17-136, 17-137, 17-139, 17-141, 17-142, 17-144, 17-147, 17-150, 17-151, 17-152, 17-153, 17-154, 17-158, 17-159, 17-160, 17-161, 17-162, 17-163, 17-169, 17-171, 17-190, 17-215, 18, 20-1, 20-2, 20-3, 20-4, 20-5, 20-6, 22-10, 22-11, 25-52. More preferably, compounds of this invention have IC50 value in the IKK-2 Enzyme Assay of Example 21 of 500 nM or less. In this regard, more preferred compounds of this invention are 3-8, 3-14, 3-21, 17-18, 17-2, 17-20, 17-27, 17-28, 17-29, 17-30, 17-31, 17-32, 17-33, 17-34, 17-35, 17-36, 17-37, 17-86, 17-91, 17-127, 17-129, 17-131, 17-133, 17-137, 17-139, 17-141, 17-150, 17-154, 17-159, 17-160, 17-161, 17-162, 17-163, 17-169, 17-171, 17-190, 17-215, 18, 20-1, 20-2, 20-3, 20-4, 20-5, 20-6, 22-10, 22-11, 25-52.


The present invention is not to be limited in scope by the specific embodiments disclosed in the examples which are intended as illustrations of a few aspects of the invention and any embodiments which are functionally equivalent are within the scope of this invention. Indeed, various modifications of the invention in addition to those shown and described herein will become apparent to those skilled in the art and are intended to fall within the appended claims.

Claims
  • 1. A compound having the formula:
  • 2. A compound of claim 1, having the structure:
  • 3. A compound of claim 2, having the structure:
  • 4. A pharmaceutical composition comprising a compound of claim 1 and a pharmaceutically acceptable carrier.
  • 5. A method for treating a condition responsive to IKK-2 inhibition, comprising administering to a patient in need thereof and effective amount of a compound of claim 1.
  • 6. The method of claim 5 wherein the condition is an inflammatory or autoimmune condition.
  • 7. The method of claim 6 wherein the inflammatory or autoimmune condition is rheumatoid arthritis, rheumatoid spondylitis, osteoarthritis, gout, asthma, bronchitis, allergic rhinitis, chronic obstructive pulmonary disease, cystic fibrosis, inflammatory bowel disease, irritable bowel syndrome, mucous colitis, ulcerative colitis, Crohn's disease, gastritis, esophagitis, hepatitis, pancreatitis, nephritis, psoriasis, eczema, dermatitis, multiple sclerosis, Lou Gehrig's disease, sepsis, conjunctivitis, acute respiratory distress syndrome, purpura, nasal polip or lupus erythematosus.
  • 8. The method of claim 5 wherein the condition is a cardiovascular, metabolic or ischemic condition.
  • 9. The method of claim 8 wherein the condition is atherosclerosis, restenosis following angioplasty, left ventricular hypertrophy, Type II diabetes, osteoporosis, erectile dysfunction, cachexia, myocardial infraction, ischemic diseases of heart, kidney, liver, and brain, organ transplant rejection, graft versus host disease, endotoxin shock, or multiple organ failure.
  • 10. The method of claim 5 wherein the condition is an infectious disease.
  • 11. The method of claim 10 wherein the infectious disease is a viral infection.
  • 12. The method of claim 11 wherein the viral infection is caused by human immunodeficiency virus, hepatitis B virus, hepatitis C virus, human papillomavirus, human T-cell leukemia virus or Epstein-Barr virus.
  • 13. The method of claim 5 wherein the condition is cancer.
  • 14. The method of claim 13 wherein the cancer is of the colon, rectum, prostate, liver, lung, bronchus, pancreas, brain, head, neck, stomach, skin, kidney, cervix, blood, larynx, esophagus, mouth, pharynx, testes, urinary bladder, ovary or uterus.
  • 15. The method of claim 5 wherein the condition is stroke, epilepsy, Alzheimer's disease, or Parkinson's disease.
  • 16. A method for treating an inflammatory or an autoimmune condition comprising administering to a patient in need thereof an effective amount of a compound or pharmaceutically acceptable salt of the compound of claim 1.
  • 17. The method of claim 16 wherein the inflammatory or autoimmune condition is rheumatoid arthritis, rheumatoid spondylitis, osteoarthritis, gout, asthma, bronchitis, allergic rhinitis, chronic obstructive pulmonary disease, cystic fibrosis, inflammatory bowel disease, irritable bowel syndrome, mucous colitis, ulcerative colitis, Crohn's disease, gastritis, esophagitis, hepatitis, pancreatitis, nephritis, psoriasis, eczema, dermatitis, multiple sclerosis, Lou Gehrig's disease, sepsis, conjunctivitis, acute respiratory distress syndrome, purpura, nasal polip or lupus erythematosus.
  • 18. A method for treating cancer comprising administering to a patient in need thereof an effective amount of a compound or pharmaceutically acceptable salt of the compound of claim 1.
  • 19. The method of claim 18 wherein the cancer is of the colon, rectum, prostate, liver, lung, bronchus, pancreas, brain, head, neck, stomach, skin, kidney, cervix, blood, larynx, esophagus, mouth, pharynx, testes, urinary bladder, ovary or uterus.
  • 20. A compound, or a pharmaceutically acceptable salt thereof, having the structure:
  • 21. A pharmaceutical composition comprising a compound of claim 20 and a pharmaceutically acceptable carrier.
Parent Case Info

This application claims the benefit of U.S. Provisional Application No. 60/251,816, filed Dec. 6, 2000, incorporated by reference herein in its entirety.

Provisional Applications (1)
Number Date Country
60251816 Dec 2000 US
Continuations (1)
Number Date Country
Parent 10004642 Dec 2001 US
Child 11211383 Aug 2005 US