Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
INDAZOLE COMPOUNDS FOR TREATING INFLAMMATORY DISORDERS, DEMYELINATING DISORDERS AND CANCERS
Document Type and Number:
WIPO Patent Application WO/2009/017795
Kind Code:
A1
Abstract:
Compounds of formula (I) or formula (Ia) and a method of treating a patient suffering from certain inflammatory disorders, demyelinating disorders, FLT3- mediated disorders, CSF-lR-mediated disorders, cancers and leukemias, comprising administering to said patient a therapeutically effective amount of a compound of formula (I) or formula (Ia) or a pharmaceutically acceptable salt thereof. (Ia). Definitions for the variables are provided herein.

Inventors:
AJAMI ALFRED M (US)
DUNCAN KENNETH (US)
Application Number:
PCT/US2008/009263
Publication Date:
February 05, 2009
Filing Date:
July 30, 2008
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
XANTHUS PHARMACEUTICALS INC (US)
AJAMI ALFRED M (US)
DUNCAN KENNETH (US)
International Classes:
C07D471/16; A61K31/4745; A61P29/00; A61P31/00; A61P35/00
Domestic Patent References:
WO2006081431A22006-08-03
WO2007092436A22007-08-16
Other References:
CHOLODY WIESLAW M ET AL: "Structure-activity relationship for antineoplastic imidazoacridinones: Synthesis and antileukemic activity in vivo", JOURNAL OF MEDICINAL CHEMISTRY, US AMERICAN CHEMICAL SOCIETY. WASHINGTON, vol. 39, no. 5, 1 January 1996 (1996-01-01), pages 1028 - 1032, XP002468996, ISSN: 0022-2623
CHOLODY W M ET AL: "5-Ä(AMINOALKYL)AMINOÜIMIDAZOÄ4,5,1-DEÜACRIDIN-6-ONES AS A NOVEL CLASS OF ANTINEOPLASTIC AGENTS. SYNTHESIS AND BIOLOGICAL ACTIVITY", JOURNAL OF MEDICINAL CHEMISTRY, US AMERICAN CHEMICAL SOCIETY. WASHINGTON, vol. 33, 1 January 1990 (1990-01-01), pages 49 - 52, XP001093764, ISSN: 0022-2623
Attorney, Agent or Firm:
DAVIS, Steven, G. et al. (Brook Smith & Reynolds, P.C.,530 Virginia Road,P.O. Box 913, Concord MA, US)
Download PDF:
Claims:
CLAIMS

What is claimed is:

1. A compound of formula (I) or a pharmaceutically acceptable salt thereof:

wherei

R 2 is H or Cl-C4 alkyl;

R 3 is H or Cl-C4 alkyl;

R 4 is H or Cl-C4 alkyl; n is an integer from 2 to 5; and

R a and R b , each independently are hydrogen or an optionally substituted alkyl; or

R a and R b , taken together with the nitrogen to which they are attached, form a non-aromatic heterocycle, optionally substituted at one or more substitutable carbon atoms with methyl, hydroxyl, or methoxy, and optionally N'-substituted with C1-C4 alkyl or C1-C4 alkyl substituted with -NR c R d ; wherein R c and R d are independently H, methyl or ethyl.

2. The compound of Claim 1, wherein R 1 is H.

3. The compound of Claim 1, wherein R 1 is NR a R b .

4. The compound of Claim 3, wherein R a and R b , each independently are hydrogen or an optionally substituted alkyl.

5. The compound of Claim 3, wherein R a and R b , taken together with the nitrogen to which they are attached, form a non-aromatic heterocycle, optionally substituted at one or more substitutable carbon atoms with methyl, hydroxyl, or methoxy, and optionally N'-substituted with C1-C4 alkyl or Cl- C4 alkyl substituted with NR c R d .

6. The compound of Claim 1 , wherein the compound is represented by formula (Ia) or a pharmaceutically acceptable salt thereof:

— -NR a R b ( n Ia N ).

7. The compound of Claim 1, wherein

R a and R b , taken together with the nitrogen to which they are attached, form a 5-7 member non-aromatic heterocycle, optionally substituted at one or more substitutable carbon atoms with methyl, hydroxyl, or methoxy, and optionally N'-substituted with C1-C4 alkyl or C1-C4 alkyl substituted with -NR c R d ; or

R a and R b or individually are hydrogen or a C1-C3 alkyl optionally substituted with -OH, -SH, halogen, cyano, nitro, amino, -COOH, a C1-C3 alkyl.

8. The compound of Claim 7, wherein

R a and R b , taken together with the nitrogen to which they are attached, form a 5-7 member non-aromatic heterocycle, optionally substituted at one or more substitutable carbon atoms with methyl, hydroxyl, or methoxy, and

optionally N'-substituted with C1-C4 alkyl or C1-C4 alkyl substituted with

-NR c R d .

9. The compound of Claim 8, represented by the following formula:

10. The compound of Claim 9 wherein n is 2 or 3.

11. The compound of Claim 10, wherein R a and R b , taken together with the nitrogen to which they are attached, form a 5-7 member non-aromatic heterocycle selected form a group consisting of

wherein Q is S, O, CH 2 , NH, or NR 102 , and R 102 is methyl or ethyl.

12. The compound of Claim 11 , wherein R a and R , taken together with the nitrogen to which they are attached, form N-morpholinyl or N-piperidinyl.

13. The compound of Claim 7, wherein R a and R b individually are hydrogen or a C1-C3 alkyl optionally substituted with -OH, -SH, halogen, cyano, nitro, amino, -COOH, or C1-C3 alkyl.

14. The compound of Claim 13, represented by the following structure:

15. The compound of Claim 14, wherein n is 2 or 3.

16. The compound of Claim 15, wherein R a and R b individually are H, methyl or ethyl.

17. The compound of Claim 1 selected from

or a pharmaceutically acceptable salts thereof.

18. The compound of Claim 1 selected from

or a pharmaceutically acceptable salts thereof.

19. A method of treating an inflammatory disorder, a demyelinating disorder, a cancer, or a leukemia in a patient, comprising administering to said patient a therapeutically effective amount of a compound of any one of Claims 1-18.

20. The method of Claim 19, wherein the disorder is a cancer selected from brest cancer, colorectal cancer, non-small cell lung cancer, ovarian, renal, sarcoma, melanoma, head & neck, hepatocellular, thyroid, multidrug-resistant leukemia, lymphoma, multiple myeloma, esophageal, large bowel, pancreatic, mesothelioma, carcinoma, sarcoma and melanoma.

21. The method of Claim 19, wherein the disorder is an inflammatory disorder selected from systemic lupus, inflammatory bowl disease, psoriasis, Crohn's disease, rheumatoid arthritis, sarcoid, Alzheimer's disease, insulin dependent diabetes mellitus, atherosclerosis, asthma, spinal cord injury, stroke, a chronic inflammatory demyelinating neuropathy, multiple sclerosis, a congenital metabolic disorder, a neuropathy with abnormal myelination, drug-induced demyelination, radiation induced demyelination, a hereditary demyelinating condition, a prion-induced demyelination, and encephalitis-induced demyelination.

22. The method of Claim 19, wherein the disorder is a demyelinating condition selected from multiple sclerosis, a congenital metabolic disorder, a neuropathy with abnormal myelination, drug-induced demyelination, radiation induced demyelination, a hereditary demyelination condition, a prion-induced demyelination, encephalitis-induced demyelination, a spinal cord injury, Alzheimer's disease, Chronic Immune Demyelinating Polyneuropathy (CIDP); multifocal CIDP; multifocal motor neuropathy (MMN); anti-MAG Syndrome (Neuropathy with IgM binding to Myelin-Associated Glycoprotein); GALOP Syndrome (Gait disorder Autoantibody Late-age Onset Polyneuropathy); anti-sulfatide antibody syndrome; anti-GM2

gangliosides antibody syndrome; POEMS syndrome (Polyneuropathy Organomegaly Endocrinopathy or Edema M-protein Skin changes); perineuritis; and IgM anti-GDlb ganglioside antibody syndrome.

23. The method of claim 19,wherein the disorder is rheumatoid arthritis or multiple sclerosis.

24. The method of claim 19,wherein the disorder is Crohn's disease, ulcerative colitis, or inflammatory bowel disease.

25. The method of claim 19,wherein the disorder is leukemia.

26. The method of claim 25, wherein the leukemia is selected from acute myeloid leukemia (AML), acute lymphocytic leukemia (ALL), chronic myeloid leukemia (CML) and chronic lymphocytic leukemia (CLL).

27. The method of claim 25, wherein the leukemia is acute myeloid leukemia characterized by a FLT3 mutation.

28. A method of treating a disease or disorder mediated by one or more of a kinase selected from FLT3, CSF-IR, and MYLK2, comprising administering to said patient a therapeutically effective amount of a compound of any one of Claims 1-18.

29. The method of Claim 28, wherein the disorder is a FLT3-mediated disorder selected from axonal degeneration, acute transverse myelitis, amyotrophic lateral sclerosis, infantile spinal muscular atrophy, juvenile spinal muscular atrophy, Creutzfeldt- Jakob disease, subacute sclerosing panencephalitis, organ rejection, bone marrow transplant rejection, non-myeloablative bone marrow transplant rejection, ankylosing spondylitis, aplastic anemia, Behcet's disease, graft-versus-host disease, Graves' disease, autoimmune hemolytic anemia,

Wegener's granulomatosis, hyper IgE syndrome, idiopathic thrombocytopenia purpura, and Myasthenia gravis.

30. The method of Claim 28, wherein the disorder is a CSF-lR-mediated disorder selected from a cardiovascular disease, diseases with an inflammatory component including glomerulonephritis, prosthesis failure, sarcoidosis, congestive obstructive pulmonary disease, asthma, pancreatitis, HIV infection, psoriasis, diabetes, tumor related angiogenesis, age-related macular degeneration, diabetic retinopathy, restenosis, schizophrenia, skeletal pain caused by tumor metastasis or osteoarthritis, or visceral, inflammatory, and neurogenic pain, osteoporosis, Paget's disease, prosthesis failure, osteolytic sarcoma, myeloma, and tumor metastasis to bone, uterine cancer, stomach cancer, hairy cell leukemia, Sjogren's syndrom, uveitis, osteolytic sarcoma, uterine cancer, and stomach cancer.

31. The method of Claim 28, wherein the disorder is a MYLK2-mediated barrier dysfunction disorder selected from diseases with an inflammatory component including ulcerative colitis, Crohn's disease, bowel ischemia, colonic ileus, vasogenic ischemia, focal cerebral ischemia, hemorrhagic or septic shock, virus associated myelopathy, septic encephalopathy, glomerulonephritis, prosthesis failure, graft-versus-host disease, sarcoidosis, congestive obstructive pulmonary disease, asthma, pancreatitis, HIV infection, HIV- associated dementia, psoriasis, atopic dermatitis, diabetes, tumor related angiogenesis, restenosis, skeletal pain caused by tumor metastasis or osteoarthritis, or visceral, inflammatory, and neurogenic pain, osteolytic sarcoma, myeloma, and tumor metastasis to bone, uterine cancer, stomach cancer, hairy cell leukemia, Sjogren's syndrom, uveitis, uterine cancer, and stomach cancer.

32. The method of Claim 19 or Claim 28, wherein one or more additional pharmaceutical agents is co-administered with a compound of formula (I).

33. A compound represented by the following formula:

or a salt thereof.

34. A pharmaceutical composition comprising the compound of any one of claims 1-18 or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier or excipient.

35. The pharmaceutical composition of claim 32, further comprising an additional therapeutic agent.

36. Use of a compound as defined in any one of claims 1 to 18 in the preparation of a medicament for the treatment of an inflammatory disorder, a demyelinating disorder, a FLT3-mediated disorder, a cancer, a leukemia, a MYLK2-mediated barrier dysfunction disorder, or a CSF-lR-mediated disorder.

37 A method of modulating the activity of FLT3 in a subject, comprising administering to a subject an effective amount of a compound or pharmaceutically acceptable salt as defined in any one of claims 1 to 18.

38. A method of modulating the activity of MYLK2 in a subject, comprising administering to a subject an effective amount of a compound or pharmaceutically acceptable salt as defined in any one of claims 1 to 18.

Description:

INDAZOLE COMPOUNDS FOR TREATING INFLAMMATORY DISORDERS, DEMYELINATING DISORDERS AND CANCERS

RELATED APPLICATIONS

This application claims the benefit of U.S. Provisional Application Serial No. 60/963,144, filed August 2, 2007, the entire teachings of which are incorporated herein by reference.

BACKGROUND OF THE INVENTION

There is a need for new pharmaceutically acceptable therapies for an inflammatory disorder, a demyelinating disorder, a FLT3-mediated disorder, a cancer, a leukemia or a CSF-lR-mediated disorder in a patient.

SUMMARY OF THE INVENTION

The present invention is directed to a class of novel compounds that can be used for treatment of an inflammatory disorder, a demyelinating disorder, a FLT3- mediated disorder, a cancer, a leukemia, MYLK2-mediated barrier dysfunction disorder, or a CSF-lR-mediated disorder in a patient.

In one embodiment, the present invention is a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a compound of formula (Ia) or a pharmaceutically acceptable salt thereof:

(Ia),

wherein R 1 is H or NR a R b ; R a and R b , each independently are hydrogen or an optionally substituted alkyl; or R a and R b , taken together with the nitrogen to which

they are attached, form a non-aromatic heterocycle, optionally substituted at one or more substitutable carbon atoms with methyl, hydroxyl, or methoxy, and optionally N'-substituted with C1-C4 alkyl or C1-C4 alkyl substituted with -NR c R d ; wherein R c and R d are individually H, methyl or ethyl; R 2 is H or C1-C4 alkyl; R 3 is H or C1-C4 alkyl; n is an integer from 2 to 5 and R 4 is H or C1-C4 alkyl.

In another embodiment, the present invention is a method of treating an inflammatory disorder, a demyelinating disorder, a FLT3 -mediated disorder, a cancer, a leukemia or a CSF-lR-mediated disorder in a patient, comprising administering to said patient a therapeutically effective amount of a compound.

In another embodiment, the present invention is a pharmaceutical composition comprising a therapeutically effective amount of a compound of formula (I) or formula (Ia) or a pharmaceutically acceptable salt thereof in a pharmaceutically acceptable carrier.

In another embodiment, the present invention is a use of a compound of formula (I) or formula (Ia) for the manufacture of a medicament for treatment of an inflammatory disorder, a demyelinating disorder, a FLT3-mediated disorder, a cancer, a leukemia or a CSF-lR-mediated disorder in a patient.

DETAILED DESCRIPTION OF THE INVENTION

It has now been discovered that administration of certain indazole compounds can be used to treat an inflammatory disorder, a demyelinating disorder, a FLT3- mediated disorder, a cancer, a leukemia or a CSF-lR-mediated disorder in a patient.

In one embodiment, the present invention is a compound of formula (I) below or a pharmaceutically acceptable salt thereof or compound of formula (Ia) or a pharmaceutically acceptable salt thereof.

(Ia).

Values and preferred values for the variables in formula (I) or formula (Ia) are defined below.

R 1 is H or NR a R b . In one embodiment R 1 is H, alternatively R 1 is NR a R b .

R 2 is H or an optionally substituted C1-C4 alkyl. Preferably, R 2 is H..

R 3 is H or C1-C4 alkyl; preferably, R 3 is methyl.

R 4 is H or C1-C4 alkyl; preferably, R 4 is methyl or ethyl.

Integer n is from 2 to 5, preferably, 2 or 3. In another embodiment, n is 2; alternatively, n is 3. In another embodiment, n is 4; alternatively, n is 5.

R a and R b , each independently, are hydrogen or an optionally substituted alkyl, Alternatively, R a and R b , taken together with the nitrogen to which they are attached, form a non-aromatic heterocycle, each optionally substituted at one or more substitutable carbon atoms with methyl, hydroxyl, or methoxy, and optionally N'-substituted with C1-C4 alkyl or C1-C4 alkyl substituted with -NR c R d .

Preferably, R a and R b , taken together with the nitrogen to which they are attached, form a 5-7 member non-aromatic heterocycle, optionally substituted at one or more substitutable carbon atoms with methyl, hydroxyl, or methoxy, and optionally N'-substituted with C1-C4 alkyl or C1-C4 alkyl substituted with -NR c R d . Alternatively, R a and R b or individually are hydrogen or a C1-C3 alkyl optionally substituted with -OH, -SH, halogen, cyano, nitro, amino, -COOH, a C1-C3 alkyl.

More preferably, R a and R b , taken together with the nitrogen to which they are attached, form a 5-7 member non-aromatic heterocycle, optionally substituted at one or more substitutable carbon atoms with methyl, hydroxyl, or methoxy, and optionally N'-substituted with C1-C4 alkyl or C1-C4 alkyl substituted with -NR c R d .

More preferably, R a and R b , taken together with the nitrogen to which they are attached, form a 5-7 member non-aromatic heterocycle selected form a group

consisting of: ; ; and .

Q is S, O, CH 2 , NH, or NR 102 , and R 102 is methyl or ethyl. More preferably, R a and R b , taken together with the nitrogen to which they are attached, form N- morpholinyl or N-piperidinyl.

More preferably, R a and R b individually are hydrogen or a C1-C3 alkyl optionally substituted with -OH, -SH, halogen, cyano, nitro, amino, -COOH, a C1-C3 alkyl. More preferably, R a and R b individually are H, methyl or ethyl.

R c and R d are individually H, methyl or ethyl.

In a first preferred embodiment, R a and R b , taken together with the nitrogen to which they are attached, form a 5-7 member non-aromatic heterocycle, optionally substituted at one or more substitutable carbon atoms with methyl, hydroxyl, or methoxy, and optionally N'-substituted with C1-C4 alkyl or C1-C4 alkyl substituted with -NR c R d ; or R a and R b or both are hydrogen or a C1-C3 alkyl optionally substituted with -OH, -SH, halogen, cyano, nitro, amino, -COOH, a C1-C3 alkyl. Preferably, R a and R b , taken together with the nitrogen to which they are attached, form a 5-7 member non-aromatic heterocycle, optionally substituted at one or more substitutable carbon atoms with methyl, hydroxyl, or methoxy, and optionally N'- substituted with C1-C4 alkyl or C1-C4 alkyl substituted with -NR c R d . Values and preferred values for the remainder of variables are as defined above in formula (I) and (Ia).

In another embodiment, the compound of the invention is represented by formula (II) or a formula (Ha):

Values and preferred values for formula (II) and (Ha) are as described above for formula (I) and (Ia).

In another embodiment, the compound of the invention is represented by formula (II) or formula (Ha), wherein: R a and R b , taken together with the nitrogen to which they are attached, form a 5-7 member non-aromatic heterocycle, optionally substituted at one or more substitutable carbon atoms with methyl, hydroxyl, or methoxy, and optionally N'-substituted with C1-C4 alkyl or C1-C4 alkyl substituted with -NR c R d . Values and preferred values for the remainder of variables are as defined above in formula (I) and (Ia).

In another embodiment, the compound of the invention is represented by formula (II), or formula (Ha), wherein: n is 2 or 3; and R a and R b , taken together with the nitrogen to which they are attached, form a 5-7 member non-aromatic heterocycle, optionally substituted at one or more substitutable carbon atoms with methyl, hydroxyl, or methoxy, and optionally N'-substituted with C1-C4 alkyl or C1-C4 alkyl substituted with -NR c R d , and values and preferred values for the remainder of variables are as defined above in formula (I) and (Ia). Preferably, R a and R b , taken together with the nitrogen to which they are attached, form a 5-7 member non- aromatic heterocycle selected form a group consisting of:

wherein Q is S, O, CH 2 , NH, or NR 102 , and R 102 is methyl or ethyl. More preferably, R a and R b , taken together with the nitrogen to which they are attached, form N- morpholinyl or N-piperidinyl.

In another embodiment, the compound of the invention is represented by formula (II) or formula (Ha), wherein: R a and R b individually are hydrogen or a Cl- C3 alkyl optionally substituted with -OH, -SH, halogen, cyano, nitro, amino, -COOH, or C1-C3 alkyl; and the values and preferred values for the remainder of the variables are as described for formula (I) and (Ia).

In another embodiment, the compound of the invention is represented by formula (II) or formula (Ha), wherein: n is 2 or 3 and R a and R b individually are hydrogen or a C1-C3 alkyl optionally substituted with -OH, -SH, halogen, cyano, nitro, amino, -COOH, or C1-C3 alkyl; and preferred values for the variables are as described for formula (I). More preferably, R a and R b both are H, methyl or ethyl.

Alternatively, for embodiments described in the previous seven paragraphs, R 1 is H and the reminder of the variables are as described above. In another alternative, in the embodiments described in the previous seven paragraphs, R 1 is NR a R b and the reminder of the variables are as described.

Examples of compounds of the invention include:

The term "alkyl", as used herein, unless otherwise indicated, includes straight or branched saturated monovalent hydrocarbon radicals, typically Cl-ClO, preferably C1-C6. Examples of alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, and t-butyl. Suitable substituents for a substituted alkyl include -OH, -SH, halogen, cyano, nitro, amino, -COOH, a C1-C3 alkyl, C1-C3 haloalkyl,

C1-C3 alkoxy, C1-C3 haloalkoxy or C1-C3 alkyl sulfanyl, or -(CH 2 ) p -(CH 2 ) q - C(O)OH, where p and q are independently an integer from 1 to 6.

The term "haloalkyl", as used herein, includes an alkyl substituted with one or more F, Cl, Br, or I, wherein alkyl is defined above.

The terms "alkoxy", as used herein, means an "alkyl-O-" group, wherein alkyl, is defined above.

The term "haloalkoxy", as used herein, means "haloalkyl-O-", wherein haloalkyl is defined above.

As used herein, an amino group may be a primary (-NH 2 ), secondary (-NHR x ), or tertiary (-NR x Ry), wherein R x and R y may be any of the optionally substituted alkyls described above.

The term "non-aromatic heterocycle" refers to non-aromatic carbocyclic ring systems typically having four to eight members, preferably five to six, in which one or more ring carbons, preferably one to four, are each replaced by a heteroatom such as N, O, or S. Non-aromatic heterocycles can be optionally unsaturated. Examples of non-aromatic heterocyclic rings include 3-tetrahydrofuranyl, 2-tetrahydropyranyl, 3- tetrahydropyranyl, 4-tetrahydropyranyl, [l,3]-dioxalanyl, [l,3]-dithiolanyl, [1,3]- dioxanyl, 2-tetrahydrothiophenyl, 3-tetrahydrothiophenyl, 2-morpholinyl, 3- morpholinyl, 4-morpholinyl, 2-thiomorpholinyl, 3-thiomorpholinyl, 4- thiomorpholinyl, 1-pyrrolidinyl, 2-pyrrolidinyl, 3-pyrorolidinyl, 1-piperazinyl, 2- piperazinyl, 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-piperidinyl, 4-thiazolidinyl, diazolonyl, and N-substituted diazolonyl.

The non-aromatic heterocyclic group may be C-attached or N-attached (where such is possible). For instance, a group derived from pyrrole may be pyrrol- 1-yl (N-attached) or pyrrol-3-yl (C-attached).

Suitable substituents for a non-aromatic heterocyclic group are those that do not substantially interfere with the pharmaceutical activity of the disclosed compound. One or more substituents can be present, which can be identical or different. Examples of suitable substituents for a substitutable carbon atom in aryl, heteroaryl or a non-aromatic heterocyclic group include -OH, halogen (-F, -Cl, -Br, and -I), -R', haloalkyl, -OR', -CH 2 R', -CH 2 OR', -CH 2 CH 2 OR', -CH 2 OC(O)R', -O-COR', -COR',

-SR', -SCH 2 R', - CH 2 SR', -SOR', -SO 2 R', -CN, -NO 2 , -COOH, -SO 3 H, -NH 2 , -NHR', -N(R') 2 , -COOR', -CH 2 COOR', -CH 2 CH 2 COOR', -CHO, -CONH 2 , -CONHR', -C0N(R') 2 , -NHCOR', -NR'COR', -NHCONH 2 , -NHCONR'H, -NHCON(R') 2 , -NR 5 CONH 2 , -NR'CONR'H, -NR'CON(R') 2 , -C(^NH)-NH 2 , -C(=NH)-NHR', -C(=NH)-N(R') 2 , -C(=NR')-NH 2 , -C(=NR')-NHR', -C(=NR')-N(R') 2 , -NH-C(=NH)-NH 2 , -NH-C(=NH)-NHR\ -NH-C(=NH)-N(R') 2 , -NH-C(=NR')-NH 2 , -NH-C(=NR')-NHR', -NH-C(=NR')-N(R') 2 , -NR'H-C(=NH)-NH 2 , -NR'-C(=NH)-NHR', -NR'-C(=NH)-N(R') 2 , -NR'-C(=NR')-NH 2 , -NR'-C(=NR')-NHR', -NR'-C(=NR')-N(R') 2 , -SO 2 NH 2 , -SO 2 NHR', -SO 2 NR' 2 , -SH, -SO k R' (k is O, 1 or 2) and -NH-C(=NH)-NH 2 . Each R' is independently an alkyl group. Oxo (C==0) and thio (C==S) are also suitable substituents for a non-aromatic heterocycle.

Suitable substituents on the nitrogen of a non-aromatic heterocyclic group include -R", -N(R") 2 , -C(O)R", -CO 2 R", -C(O)C(O)R", -C(O)CH 2 C(O)R", -SO 2 R", -SO 2 N(R") 2 , -C(=S)N(R") 2 , -C(=NH)-N(R") 2 , and -NR" SO 2 R". R" is hydrogen, an alkyl or alkoxy group.

At various places in the present specification, substituents of compounds are disclosed in groups or in ranges. It is specifically intended that the description include each and every individual subcombination of the members of such groups and ranges. For example, the term "C 1-6 alkyl" is specifically intended to individually disclose Cl, C 2 , C 3 , C 4 , C 5 , C 6 , Cl-C 6 , Cl-C 5 , Cl -C 4 , Cl-C 3 , Cl-C 2 , Ci--C 6 , C 2 -C 5 , C 2 -C 4 , C 2 - C 3 , C 3 -C 6 , C 3 -C 5 , C 3 -C 4 , C 4 -C 6 , C 4 -C 5 , and C 5 -C 6 alkyl. By way of another example, the term "5-7 member non-aromatic heterocycle" is specifically intended to individually disclose a heterocycle having 5, 6, 7, 5-7, 5-6, and 6-7 ring atoms.

Throughout the specification, structures may or may not be presented with chemical names. Where any question arises as to nomenclature, the structure prevails.

Disorders Treatable by the Compounds of the Invention

It has now been discovered, that compounds of formula (I) or formula (Ia) or formula (II) or formula (Ha) can be used to treat an inflammatory disorder, a demyelinating disorder, a FLT3-mediated disorder, a cancer, a leukemia or a CSF-IR- mediated disorder in a patient. The term "patient" means a warm blooded animal, i.e., a mammal, such as for example rat, mice, dogs, cats, guinea pigs, and primates including humans. The terms "treat" or "treating" include any treatment, including, but not limited to, alleviating symptoms, eliminating the causation of the symptoms either on a temporary or permanent basis, or preventing or slowing the appearance of symptoms and progression of the named disorder or condition.

1. Cancers

In one embodiment, the present invention is a method of treating a patient suffering from a cancer. The method comprises administering to a patient a therapeutically effective amount of a compound of formula (I) or formula (Ia) or formula (II) or formula (Ha) or a pharmaceutically acceptable salt thereof.

In another embodiment, the present invention is a method of treating a subject suffering from a cancer. As used herein, the term "cancer" refers to the uncontrolled growth of abnormal cells that have mutated from normal tissues. A cancerous tumor (malignancy) is of potentially unlimited growth and expands locally by invasion and systemically by metastasis. Examples of cancers that can be treated by the compounds of the present invention include: breast cancer, colorectal cancer, non- small cell lung cancer, ovarian, renal, sarcoma, melanoma, head & neck, hepatocellular, thyroid, multidrug-resistant leukemia, lymphoma, multiple myeloma, esophageal, large bowel, pancreatic, mesothelioma, carcinoma (e.g. adenocarcinoma, including esophageal adenocarcinoma), sarcoma (e.g. spindle cell sarcoma, liposarcoma, leiomyosarcoma, abdominal leiomyosarcoma, sclerosing epithelioid sarcoma) and melanoma (e.g. metastatic malignant melanoma). In one embodiment, the patient can be treated for bone metastases. Treatment of subtypes of the aforementioned cancers is also included. Subtypes are described in the following paragraphs.

"Treating a subject suffering from cancer" includes achieving, partially or substantially, one or more of the following: arresting the growth or spread of a cancer, reducing the extent of a cancer (e.g., reducing size of a tumor or reducing the number of affected sites), inhibiting the growth rate of a cancer, and ameliorating or improving a clinical symptom or indicator associated with a cancer (such as tissue or serum components).

"Treating a bone metastases", as used herein, refers to reducing (partially or completely) the size of the bone metastases, slowing the growth of the metastases relative to the absence of treatment and reducing the extent of further spread of the cancer. "Treating a bone metastases" also includes pain reduction, decreased incidents of fractures, relief of spinal cord compression, control of hypercalcaemia, and/or restoration of normal blood cell counts.

"Breast cancer" includes, but is not limited to, ductal carcinoma, lobular carcinoma, inflammatory carcinoma, medullary carcinoma, colloid or mucinous carcinoma, papillary carcinoma, tubular carinoma, triple negative breast cancer, inflammatory breast cancer, metaplastic carcinoma, Paget's disease, and Phyllodes tumor.

As used herein "ovarian cancer", is cancer of the ovaries or fallopian tubes, including cancers of germ cells, stromal cells, and epithelial cells. Examples of ovarian cancers include but are not limited to:

Epithelial Ovarian Tumors, which include but are not limited to, serous adenomas, mucinous adenomas, and Brenner tumors, tumors of low malignant potential (LMP tumors), borderline epithelial ovarian cancer, epithelial ovarian cancers, carcinomas and undifferentiated epithelial ovarian carcinomas;

Germ Cell tumors which include but are not limited to, teratoma, dysgerminoma, endodermal sinus tumor, and choriocarcinoma; and

Stromal tumors, which include but are not limited to, granulosa cell tumors, granulosa-theca tumors, and Sertoli-Leydig cell tumors.

"Renal cancer" or "kidney cancer", as used herein, includes but is not limited to, transitional cell cancer (TCC) of the renal pelvis, Wilms Tumour and renal cell cancer.

Renal cell cancer is also called renal adenocarcinoma or hypernephroma. In renal cell cancer, the cancerous cells are found in the lining of the tubules (the smallest tubes inside the nephrons that help filter the blood and make urine).

There are several types of renal cell cancer including but not limited to clear cell, chromophilic, chromophobic, oncocytic, collecting duct and sarcomatoid.

Renal cancer also includes cancers containing more than one of the cell types described above.

As used herein, "melanoma" is a type of skin cancer that occurs in the cells that color the skin, called melanocytes. Types of melanoma include but are not limited to:

Cutaneous melanoma, superficially spreading melanoma, nodular malignant melanoma, lentiginous malignant melanoma, acral lentiginous melanoma, demoplastic malignant melanomas, giant melanocyte nevus, amelanotic malignant melanoma, acral lentiginous melanoma unusual melanoma variants, including mucosal malignant melanoma and ocular malignant melanoma.

"Sarcomas", as used herein, include but are not limited to, fibrosarcomas from fibrous body tissues, leiomyosarcomas and rhabdomyosarcomas from muscle tissues, liposarcomas from fat, synovial sarcomas, angiosarcomas from blood vessels, MPNST - malignant peripheral nerve sheath tumours (PNSTs), GIST - gastrointestinal stromal sarcoma, osteosarcoma, myosarcoma, chondrosarcoma, bile duct sarcoma, brain sarcoma, breast sarcoma, soft tissue sarcoma, uterine sarcoma, endocardial sarcoma, stromal sarcomas from supporting tissues (endometrial stromal sarcoma), granuloytic, histiolytic, hemangioendothelial, Kupffer-cell, neurogenic, round-cell, reticulum cell, spindle cell, Kaposi's sarcoma of the skin, Ewing's sarcomas and PNETs. In certain embodiments, the sarcoma is leiomyosarcoma or liposarcoma.

"Thyroid cancer", as used herein, includes but is not limited to, papillary and/or mixed papillary/follicular, follicular and/or Hurthle cell, lymphoma, medullary, anaplastic and combinations thereof.

The term "head and neck cancer" as used herein, encompasses tumors that occur in several areas of the head and neck region, including the nasal passages,

sinuses, mouth, throat, larynx (voice box), swallowing passages, salivary glands, and skin cancers that develop on the scalp, face, or neck may also be considered head and neck cancers. These cancers include but are not limited to squamous cell carcinoma, mucoepidermoid carcinoma, adenoid cystic carcinoma, lymphoma, adenocarcinoma, esthesioneuroblastoma, tumors of the nasal cavity and paranasal sinuses, nasopharyngeal cancer, cancers of the oral cavity (including all the various parts of the mouth: the lips; the lining inside the lips and cheeks (the buccal mucosa); the bottom of the mouth; the front of the tongue; the front part of the top of the mouth (the hard palate); the gums; and the area behind the wisdom teeth (the retromolar trigone)), tumors of the oropharynx, hypopharyngeal tumors, laryngeal cancer and salivary gland cancer (including malignant salivary gland tumor).

As used herein "hepatocellular cancer" or "liver cancer" includes but is not limited to: hepatocellular carcinoma (also sometimes called hepatoma or HCC) "carcinoma", fϊbrolamellar HCC, cholangiocarcinoma, angiosarcoma (also be called haemangiosarcoma) and hepatoblastoma.

As used herein, "non-small cell lung cancer" includes, squamous cell carcinoma, adenocarcinoma and undifferentiated non-small cell lung cancer (undeveloped cancer cells are known as undifferentiated cells) and large cell carcinoma.

"Colorectal cancer" as used herein, includes any type of colon or rectal cancer, including but not limited to, adenoscarcinoma, sarcoma, melanoma, stromal, carcinoid, and lymphoma.

2. Inflammatory conditions

In one embodiment, the present invention is a method of treating a patient suffering from an inflammatory condition. The method comprises administering to a patient a therapeutically effective amount of a compounds of formula (I) or formula (Ia) or formula (II) or formula (Ha) or a pharmaceutically acceptable salt thereof. The condition can be systemic lupus, psoriasis, Crohn's disease, inflammatory bowel disease (IBD), ulcerative colitis, rheumatoid arthritis, sarcoid, Alzheimer's disease, insulin dependent diabetes mellitus, atherosclerosis, asthma, spinal cord injury, stroke,

a chronic inflammatory demyelinating neuropathy, multiple sclerosis, a congenital metabolic disorder, a neuropathy with abnormal myelination, drug-induced demyelination, radiation induced demyelination, a hereditary demyelinating condition, a prion-induced demyelination, encephalitis-induced demyelination. Examples of chronic inflammatory demyelinating neuropathies include: Chronic Immune Demyelinating Polyneuropathy (CIDP); multifocal CIDP; multifocal motor neuropathy (MMN); anti-MAG Syndrome (Neuropathy with IgM binding to Myelin-Associated Glycoprotein); GALOP Syndrome (Gait disorder Autoantibody Late-age Onset Polyneuropathy); anti-sulfatide antibody syndrome; anti-GM2 gangliosides antibody syndrome; POEMS syndrome (Polyneuropathy Organomegaly Endocrinopathy or Edema M-protein Skin changes); perineuritis; and IgM anti-GDlb ganglioside antibody syndrome.

3. Demyelinating conditions

In another embodiment, the present invention is a method of treatment of a patient suffering from a demyelinating condition. The method comprises administering to a patient a therapeutically effective amount of a compound of formula (I) or formula (Ia) or formula (II) or formula (Ha) or a pharmaceutically acceptable salt thereof. As used herein, a "demyelinating condition" is a condition that destroys, breaks the integrity of or damages a myelin sheath. As used herein, the term "myelin sheath" refers to an insulating layer surrounding vertebrate peripheral neurons, that increases the speed of conduction and formed by Schwann cells in the peripheral or by oligodendrocytes in the central nervous system. Such condition can be multiple sclerosis, a congenital metabolic disorder, a neuropathy with abnormal myelination, drug-induced demyelination, radiation induced demyelination, a hereditary demyelination condition, a prion-induced demyelination, encephalitis- induced demyelination, a spinal cord injury, Alzheimer's disease as well as chronic inflammatory demyelinating neuropathies, examples of which are given above. In one embodiment, the condition is multiple sclerosis. The method comprises administering to a patient a therapeutically effective amount of a compound of

formula (I) or formula (Ia) or formula (II) or formula (Ha) or a pharmaceutically acceptable salt thereof.

In another embodiment, the present invention is a method of promoting remyelination of nerve cells in a patient, comprising administering to the patient in need thereof a therapeutically effective amount of a compound of formula (I) or formula (Ia). The patient can be suffering from any of the demyelinating conditions listed above.

In another embodiment, the present invention is a method of preventing demyelination and promoting remyelination in a patient in need thereof, comprising administering a combination of a therapeutically effective amount of a compound of formula (I) or formula (Ia) or formula (II) or formula (Ha) or pharmaceutically acceptable salt thereof, and an anti-inflammatory agent as described below.

In another embodiment, the present invention is a method of reversing paralysis in a subject in need thereof with a demyelinating disease, comprising administering to the subject a compound in an amount sufficient to inhibit lymphocyte infiltration of immune cells in the spinal cord to promote remyelination of nerve cells in the spinal cord and thereby treating paralysis in said subject, wherein the compound is of formula formula (I) or formula (Ia) or formula (II) or formula (Ha) or a pharmaceutically acceptable salt thereof.

Diseases such as, for example, multiple sclerosis (MS), systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), inflammatory bowel disease (IBD) and psoriasis are considered autoimmune diseases which represent assaults by the body's immune system which may be systemic in nature, or else directed at individual organs in the body. They appear to be diseases in which the immune system makes mistakes and, instead of mediating protective functions, becomes the aggressor. In one aspect, the compounds of the present invention are used for the treatment of autoimmune diseases.

The compounds of the present invention can be used in models for the treatment of autoimmune diseases such as multiple sclerosis and rheumatoid arthritis. One method of showing the utility of a pharmaceutical compound for the treatment of

various conditions associated with multiple sclerosis (MS) is its ability to inhibit effects of experimental allergic encephalomyelitis in laboratory animals.

Experimental allergic encephalomyelitis (EAE) is an animal model for MS, which entails inducing a T-cell-mediated autoimmune disease against myelin basic protein in certain susceptible mammalian species. The EAE model is an appropriate method for studying the inflammation of the brain and spinal cord associated with MS (see Bolton, C. Mult, Scler, 1995;l(3);143-9).

In rodents, injection of whole spinal cord or spinal cord components such as myelin basic protein induces an autoimmune response based on the activation of T- lymphocytes. Clinical disease typically becomes manifest around day 8-10 after inoculation, observed as a broad spectrum of behavioral anomalies ranging from mild gait disturbances and tail atony to complete paralysis and death. Weight loss typically occurs. In animals that survive, spontaneous recovery occurs, accompanied by variable recovery of most motor function. Depending on the species, allergen, and methodology used, animals tested by the EAE model may experience a single (acute EAE) or several (chronic relapsing EAE) attacks.

Treatments of EAE come in many structural forms: treatment can be prophylactic or preventative, whereby the therapeutic composition is administered before immunization; treatment can be initiated during the first week of induction; and treatment can be interventious, initiated after clinical symptoms are extent (acute or chronic). Prevention protocols are very common in the literature, treatment after disease is rarer, and treatment after weeks of disease are the most infrequent.

Rheumatoid Arthritis (RA) is an autoimmune disorder characterized by the chronic erosive inflammation in joints leading to the destruction of cartilage and bones. Several disease modifying antirheumatic drugs (DMARDS) are used in the treatment of RA. Currently, the two most important DMARDS are inhibitors of tumor necrosis factor α (TNF-α) and methotrexate (MTX). One method for demonstrating the utility of a pharmaceutical compound for the treatment of various conditions associated with RA is its ability to inhibit the induction of arthritis by collagen monoclonal antibodies (mABs) in mice.

Collagen-induced Arthritis (CIA) is an experimental autoimmune disease that can be elicited in susceptible strains of rodents (rat and mouse) and nonhuman primates by immunization with type II collagen, the major constituent protein of articular cartilage. CIA manifests as swelling and erythema in the limbs of the mouse. This model of autoimmunity shares several clinical and pathological features with rheumatoid arthritis (RA) and has become the most widely studied model of RA. CIA in the mouse model was first described by Courtenay et al. in 1980 (Courtnay, J. S., Dallman, M. J., Dayman, A.D., Martin A., and Mosedale, B. (1980) Immunisation against heterologous type II collagen induces arthritis in mice. Nature 283, 666-668). Like RA, susceptibility to CIA is regulated by the class II molecules of the major histocompatibility complex (MHC), indicating the crucial role played by T cells.

4. FLT 3 -Mediated Disorders

In one embodiment, the present invention is a method of treating a patient suffering from a FLT3-mediated disorder. The method comprises administering to a patient a therapeutically effective amount of a compound of formula (I) or formula (Ia) or formula (II) or formula (Ha) or a pharmaceutically acceptable salt thereof.

As used herein, the term "FLT3 -mediated disorder" is a disorder in which one or more symptoms can be inhibited, alleviated, reduced or whose onset can be delayed by inhibiting completely or partially the FLT3 protein kinase. Inhibition of FLT3 has been shown to suppress immune response, possibly via inhibition of DC- induced stimulation of T cells, and may be considered for the treatment of autoimmune diseases (see, WO 2006/020145 A2; Whartenby, et al, PNAS, 2005, 102, 16741-16746).

The terms "treat" or "treating", when used with reference to a FLT3 -mediated condition, include any treatment, including, but not limited to, alleviating symptoms, eliminating the causation of the symptoms associated with a FLT3-mediated condition either on a temporary or permanent basis, or preventing or slowing the appearance of symptoms and progression of the named disorder or condition.

As used herein the term "therapeutically effective amount", when used with reference to a FLT3-mediated condition, is the amount of a compound disclosed

herein that will achieve a partial or total inhibition or delay of the progression of a FLT3 -mediated disorder in a patient.

FLT3 -mediated disorders and conditions include axonal degeneration, acute transverse myelitis, amyotrophic lateral sclerosis, infantile spinal muscular atrophy, juvenile spinal muscular atrophy, Creutzfeldt- Jakob disease, subacute sclerosing panencephalitis, organ rejection, bone marrow transplant rejection, non-myeloablative bone marrow transplant rejection, ankylosing spondylitis, aplastic anemia, Behcet's disease, graft-versus-host disease, Graves' disease, autoimmune hemolytic anemia, Wegener's granulomatosis, hyper IgE syndrome, idiopathic thrombocytopenia purpura, and Myasthenia gravis.

5. Leukemias

In one embodiment, the compounds of the present invention, for example the compounds of formula (I) or formula (Ia) or formula (II) or formula (Ha), can be used to treat certain leukemias, including FLT3-mediated leukemias.

In one embodiment, the present invention is a method of treating a patient suffering from an acute myeloid leukemia characterized by a FLT3 mutation. The method comprises administering to a patient a therapeutically effective amount of a compound of formula (I) or formula (Ia) or formula (II) or formula (Ha) or a pharmaceutically acceptable salt thereof.

As used herein, the term "leukemia" is a cancer of the blood or bone marrow characterized by an abnormal proliferation of blood cells, usually white blood cells (leukocytes). It is part of the broad group of diseases called hematological neoplasms.

Leukemias are selected from acute myeloid leukemia (AML), acute lymphocytic leukemia (ALL), chronic myeloid leukemia (CML) and chronic lymphocytic leukemia (CLL). In one embodiment, the present invention is a method of treating a patient suffering from a hairy cell leukemia (HCL).

Acute lymphocytic leukemia (also known as Acute Lymphoblastic Leukemia, or ALL) is the most common type of leukemia in young children. This disease also affects adults, especially those age 65 and older.

Acute myelogenous leukemia (also known as Acute Myeloid Leukemia, or AML) occurs more commonly in adults than in children. This type of leukemia was previously called acute nonlymphocytic leukemia.

Chronic lymphocytic leukemia (CLL) most often affects adults over the age of 55. It sometimes occurs in younger adults, but it almost never affects children.

Chronic myelogenous leukemia (CML) occurs mainly in adults. A very small number of children also develop this disease.

Hairy Cell Leukemia (HCL) leukemia is an incurable, indolent blood disorder in which mutated, partly matured B cells accumulate in the bone marrow. Its name is derived from the shape of the cells, which look like they are covered with short, fine, hair-shaped projections. Unlike any other leukemia, HCL is characterized by low white blood cell counts.

6. CSF-I R-mediated disorders

In one embodiment, the present invention is a method of treating a CSF-IR- mediated condition in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of a compound of formula (I) or formula (Ia) or formula (II) or formula (Ha) or a pharmaceutically acceptable salt thereof.

Colony Stimulating Factor -1 Receptor (CSF-IR) signaling play an important role in the etiology of the disorders and conditions described above and is described, for example in Simoncic et al, Mol.Cel. Biol., Vol. 26, No. 11 (2006), pp. 41-49- 4160.; Yang et al, Ann. Rheum. Dis. (2006); 65, pp. 1671-1672; Irving et al, The FASEB J., Vol. 20 (2006): pp.E1315-E1326; Irving et al, The FASEB J., Vol. 20 (2006), pp. 1921-1923; Heinonen et al, PNAS (2006), vol. 103, no. 8, pp. 2776-2781; Conway et al, PNAS (2006), vol. 102, no. 44, pp. 16078-16083; Himes et al, The J. Immunol. (2006), 176: 2219-2228; Pixley et al, Trends in Cell Biol. (2004), Vol. 14, No. 11, pp.628-638; U.S. Pat. App. Pub. No. 2006/0094081; U.S. Pat. App. Pub. No. 2006/0189623; U.S. Pat. App. Pub. No. 2006/0148812; U.S. Pat. App. Pub. No. 2006/0100201 ; U.S. Pat. 5,714,493; U.S. RE37,650. The relevant portions of all of these publications are incorporated herein by reference.

CSF-lR-mediated disorders include cardiovascular disease (e.g. artherial sclerosis), diseases with an inflammatory component including glomerulonephritis, prosthesis failure, sarcoidosis, congestive obstructive pulmonary disease, asthma, pancreatitis, HIV infection, psoriasis, diabetes, tumor related angiogenesis, age- related macular degeneration, diabetic retinopathy, restenosis, schizophrenia, skeletal pain caused by tumor metastasis or osteoarthritis, or visceral, inflammatory, and neurogenic pain, osteoporosis, Paget's disease, prosthesis failure, osteolytic sarcoma, myeloma, and tumor metastasis to bone, uterine cancer, stomach cancer, hairy cell leukemia, Sjogren's syndrom, or uveitis. The disorders include cancers such as osteolytic sarcoma, myeloma, and tumor metastasis to bone, uterine cancer, stomach cancer, hairy cell leukemia.

Kinases Inhibited by the Compounds of the Invention

FLT3 (Fms-like tyrosine kinase; other names include CDl 35, FLK2 (Fetal liver kinase 2), STKl (Stem cell kinase I)) is a class III receptor tyrosine kinase (RTK) structurally related to the receptors for platelet derived growth factor (PDGF), colony stimulating factor 1 (CSFl), and KIT ligand (KL). These RTKs contain five immunoglobulin-like domains in the extracellular region and an intracelular tyrosine kinase domain split in two by a specific hydrophilic insertion (kinase insert). FLT3, closely related to PDGF receptors and c-Kit is, however, not inhibited by the small molecule inhibitors of PDGF and c-Kit; (G Del Zotto et al., J Biol. Regulators Homeostatic Agents 15: 103-106, 2001).

The compounds of the present invention demonstrate inhibition of FLT3 at low nanomolar concentrations. In addition, the compounds of the present invention are highly specific for FLT3, and to a lesser extent V561D mutant PDGFRA (60-80 nM EC 50) and CSF-IR (200-400 nM EC50) within the broader class of tyrosine kinases. Inhibition of FLT3 is associated with modulation of transmigrating inflammatory cells by changing the antigen-presenting and signaling pathways that are mediated through dendritic cells and their recruitment, interaction and retraining of T-cells (Ajami et al. A.M., Boss, M. A. and Paterson, J. Compounds for treating

autoimmune and demyelinating diseases. US Patent Appl. 2006/0189546A1, the disclosure of which is incorporated herein by reference).

In one embodiment, the present invention is a method of modulating the activity of FLT3 in a subject, comprising administering to the subject an effective amount of a compound of formula (I) or formula (Ia) or formula (II) or formula (Ha) or a pharmaceutically acceptable salt thereof.

The compounds of the present invention further demonstrate an unexpected and off target inhibitory effect on the MYLK2, light chain myosin kinase, a calcium dependent, serine-threonine kinase with no active site relationship to the Flt3 kinase class.

Although MYLK2 is structurally unrelated to Flt3, the inhibition of both kinases suggests a fortuitous and relevant functional connection in the mode of action of compounds in the present invention.

A related hallmark of invasive cells, shared by myeloid, lymphoid and cancer cells in their metastatic phase is the ability to breech cellular barriers, such as basement membranes, and therefore to invade and destroy tissues e.g. myelin, in the case of multiple sclerosis, or intestinal epithelial cells in Crohn's disease or ulcerative colitis (Behanna HA, Watterson DM and Ranaivo HR (2006) Biochim. Biophys. Acta 1763:1266-1274; Shen L, Black ED, Witkowski ED, Lencer WI, Guerriero V, Schneeberger EE and Turner JR (2006), J Cell. Sci. 119:2095-2106). Transmigration from blood vessels into synovium and bone in inflammatory arthritis serves as another case in point as does the recognized behavior of transmigrating tumor-associated macrophages and osteoclasts in metastatic cancers.

These so-called diapedesing cells have an overexpression of the molecular machinery for intracellular contractile force governed by the actomyosin regulatory complexes and the contraction cycle of the intracellular myosin complex. By the same token, the underlying barrier dysfunction, responsible for the leaky structures that permit destructive trafficking by invading immune system cells, is itself the result of an imbalance between intercellular adhesive forces, such as adherence junctions, and intracellular contractile forces that are linked to these adherence structures. These contractile forces also are mediated by actin-myosin cytoskeletal function. Activation

of this cytoskeletal function requires phosphorylation of the myosin regulatory light chains (MLC) by myosin light chain kinase (MYLK2) (Kuhlmann CRW, Tamaki R, Gamerdinger M, Lessman V, Behl C, Kempski OS and Luhmann HJ, J. Neurochem. 102:501-507).

Thus, it follows that inhibition of elevated levels of MYLK2 would prove beneficial in both suppressing the ability of activated immune cells to traffic and in suppressing the propensity of immunologically challenged barrier cells to pull themselves apart and exacerbate the impact of invasion into the underlying tissues. As such, the low nM inhibitory EC50 values reached with Compound (IV), an exemplar of the present invention, represents an unexpected and novel result, with significantly greater potency than the aryl sulfonamides known heretofore as MLCK (myosin light chain kinase) inhibitors (at micromolar EC50 levels) (Saitoh M, Ishikawa T, Matsushima S, Naka M, and Hidaka H (1987), J. Biol. Chem. 262(16):7796-7801).

MYLK2-mediated barrier dysfunction disorders include diseases with an inflammator component such as ulcerative colitis, Crohn's disease, bowel ischemia, colonic ileus, vasogenic ischemia, focal cerebral ischemia, hemorrhagic or septic shock, virus associated myelopathy, septic encephalopathy, glomerulonephritis, prosthesis failure, graft-versus-host disease, sarcoidosis, congestive obstructive pulmonary disease, asthma, pancreatitis, HIV infection, HIV-associated dementia, psoriasis, atopic dermatitis, diabetes, tumor related angiogenesis, restenosis, skeletal pain caused by tumor metastasis or osteoarthritis, or visceral, inflammatory, and neurogenic pain, osteolytic sarcoma, myeloma, and tumor metastasis to bone, uterine cancer, stomach cancer, hairy cell leukemia, Sjogren's syndrom, uveitis, uterine cancer, and stomach cancer.

In one embodiment, the present invention is a method of modulating the activity of MYLK2 in a subject, comprising administering to the subject an effective amount of a compound of formula (I) or formula (Ia) or formula (II) or formula (Ha) or a pharmaceutically acceptable salt thereof.

Modes of Administration

The term "therapeutically effective amount" means an amount of the compound, which is effective in treating the named disorder or condition. When administered for the treatment or inhibition of a particular condition or disorder, it is understood that an effective dosage can vary depending upon the particular compound utilized, the mode of administration, and severity of the condition being treated, as well as the various physical factors related to the individual being treated. In therapeutic applications, a compound of the present teachings can be provided to a patient already suffering from a disease in an amount sufficient to treat the symptoms of the disease and its complications. The dosage to be used in the treatment of a specific individual typically must be subjectively determined by the attending physician. The variables involved include the specific condition and its state as well as the size, age and response pattern of the patient.

In certain embodiments, therapeutically effective amount means an amount sufficient to effect remyelination of nerve cells in a patient.

In treating a patient afflicted with a conditions described above, all of the disclosed compounds can be administered in any form or mode which makes the compound bioavailable in therapeutically effective amounts. For example, compounds of formula (I) or formula (Ia) or formula (II) or formula (Ha) can be administered in a form of a pharmaceutically acceptable salt. The term "pharmaceutically acceptable salt" means either an acid addition salt or a basic addition salt, whichever is possible to make with the compounds of the present invention. "Pharmaceutically acceptable acid addition salt" is any non-toxic organic or inorganic acid addition salt of the base compounds represented by formula (I) or formula (Ia). Illustrative inorganic acids which form suitable salts include hydrochloric, hydrobromic, sulfuric and phosphoric acid and acid metal salts such as sodium monohydrogen orthophosphate and potassium hydrogen sulfate. Illustrative organic acids which form suitable salts include the mono-, di- and tri-carboxylic acids. Illustrative of such acids are, for example, acetic, glycolic, lactic, pyruvic, malonic, succinic, glutaric, fumaric, malic, tartaric, citric, ascorbic, maleic, hydroxymaleic, benzoic, hydroxybenzoic, phenylacetic, cinnamic, salicyclic, 2- phenoxybenzoic, p-toluenesulfonic acid and sulfonic acids such as methanesulfonic

acid and 2-hydroxyethanesulfonic acid. Either the mono- or di-acid salts can be formed, and such salts can exist in either a hydrated or substantially anhydrous form. In general, the acid addition salts of these compounds are more soluble in water and various hydrophilic organic solvents and which in comparison to their free base forms, generally demonstrate higher melting points. "Pharmaceutically acceptable basic addition salts" means non-toxic organic or inorganic basic addition salts of the compounds of formula (I) or formula (Ia) or formula (II) or formula (Ha). Examples are alkali metal or alkaline-earth metal hydroxides such as sodium, potassium, calcium, magnesium or barium hydroxides; ammonia, and aliphatic, alicyclic, or aromatic organic amines such as methylamine, trimethylamine and picoline. The selection of the appropriate salt may be important so that the ester is not hydrolyzed. The selection criteria for the appropriate salt will be known to one skilled in the art.

Compounds of the present invention can be administered by a number of routes including orally, sublingually, buccally, subcutaneously, intramuscularly, intravenously, transdermally, intranasally, rectally, topically, and the like. One skilled in the art of preparing formulations can determine the proper form and mode of administration depending upon the particular characteristics of the compound selected for the condition or disease to be treated, the stage of the disease, the condition of the patient and other relevant circumstances. For example, see Remington's Pharmaceutical Sciences, 18 th Edition, Mack Publishing Co. (1990), incorporated herein by reference.

The disclosed compounds are administered by any suitable route, including, for example, orally in capsules, suspensions or tablets.

Pharmaceutical compositions of the invention that are suitable for oral administration can be presented as discrete dosage forms, such as, but are not limited to, tablets (e.g., chewable tablets), caplets, capsules, and liquids (e.g., flavored syrups). Such dosage forms contain predetermined amounts of active ingredients, and may be prepared by methods of pharmacy well known to those skilled in the art. See generally, Remington's Pharmaceutical Sciences (1990) 18th ed., Mack Publishing, Easton PA.

Typical oral dosage forms of the invention are prepared by combining the active ingredient(s) in an admixture with at least one excipient according to conventional pharmaceutical compounding techniques. Excipients can take a wide variety of forms depending on the form of preparation desired for administration. For example, excipients suitable for use in oral liquid or aerosol dosage forms include, but are not limited to, water, glycols, oils, alcohols, flavoring agents, preservatives, and coloring agents. Examples of excipients suitable for use in solid oral dosage forms (e.g., powders, tablets, capsules, and caplets) include, but are not limited to, starches, sugars, micro-crystalline cellulose, diluents, granulating agents, lubricants, binders, and disintegrating agents.

The solutions or suspensions may also include one or more of the following adjuvants: sterile diluents such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl paraben; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylene diaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. The parenteral preparation can be enclosed in ampules, disposable syringes or multiple dose vials.

The compound of formula (I) or formula (Ia) or formula (II) or formula (Ha) of this invention may also be administered topically, and when done so the carrier may suitably comprise a solution, ointment or gel base. The base, for example, may comprise one or more of petrolatum, lanolin, polyethylene glycols, bee wax, mineral oil, diluents such as water and alcohol, and emulsifiers and stabilizers.

In the embodiments in which the compounds of the invention are used to treat cancer, the dosage range at which the disclosed compounds, for example, compounds of formula (I) or formula (Ia) or formula (II) or formula (Ha), including the above- mentioned examples thereof, exhibit their ability to act therapeutically can vary depending upon the severity of the condition, the patient, the formulation, other underlying disease states that the patient is suffering from, and other medications that may be concurrently administered to the patient. Generally, the compounds described herein will exhibit their therapeutic activities at dosages of between about 0.1 mg/m 2

free base equivalent per square meter of body surface area/single dose to about 1000 mg/m 2 free base equivalent per square meter of body surface area/single dose. These dosages can be administered, for example, once per week, once every other week, once every third week or once per week for three out of every four weeks.

Alternatively, when used to treat cancer, the disclosed compounds can be administered daily (typically orally). Daily dose of administration of the compounds of the present invention can be repeated, in one embodiment, for one week. In other embodiments, daily dose can be repeated for one month to six months; for six months to one year; for one year to five years; and for five years to ten years. Representative total daily doses would include those in the range of 10-2000 mg. The total daily dose can be divided into equal doses and administered twice daily, thrice daily, or four times daily. In other embodiments, the length of the treatment by repeated administration is determined by a physician.

When used to treat other indications, the dosage range at which the disclosed compounds of formula (I) or formula (Ia) or formula (II) formula (Ha) exhibit their ability to act therapeutically can vary depending upon the severity of the condition, the patient, the formulation, other underlying disease states that the patient is suffering from, and other medications that may be concurrently administered to the patient. Generally, the inventive compounds of the invention will exhibit their therapeutic activities at dosages of between about 0.001 mg/kg of patient body weight/day to about 100 mg/kg of patient body weight/day. For example, the dosage can be 0.1-100 mg/kg per every other day or per week.

Combination Therapies

The compounds used in the present invention can be administered alone or in combination with one or more other pharmaceutically active agents that are effective against the inflammatory condition and/or the demyelinating disorder being treated.

As used herein, the term "combination" with reference to pharmaceutically active agents and the term "co-administering" and "co-administration" refer to administering more than one pharmaceutically active agent to a patient during one treatment cycle and not necessarily simultaneous or in a mixture.

In one embodiment, the compounds of the present invention are administered in combination with an anti-inflammatory agent. The anti-inflammatory agent can be adrenocorticotropic hormone, a corticosteroid, an interferon, glatiramer acetate, or a non-steroidal anti-inflammatory drug (NSAID).

Examples of suitable anti-inflammatory agents include corticosteroid such as prednisone, methylprednisolone, dexamethasone Cortisol, cortisone, fludrocortisone, prednisolone, 6α-methylprednisolone, triamcinolone, or betamethasone.

Other examples of suitable anti-inflammatory agents include NSAIDs such as aminoarylcarboxylic acid derivatives (e.g., Enfenamic Acid, Etofenamate, Flufenamic Acid, Isonixin, Meclofenamic Acid, Niflumic Acid, Talniflumate, Terofenamate and Tolfenamic Acid), arylacetic acid derivatives (e.g., Acematicin, Alclofenac, Amfenac, Bufexamac, Caprofen, Cinmetacin, Clopirac, Diclofenac, Diclofenac Sodium, Etodolac, Felbinac, Fenclofenac, Fenclorac, Fenclozic Acid, Fenoprofen, Fentiazac, Flubiprofen, Glucametacin, Ibufenac, Ibuprofen, Indomethacin, Isofezolac, Isoxepac, Ketoprofen, Lonazolac, Metiazinic Acid, Naproxen, Oxametacine, Proglumrtacin, Sulindac, Tenidap, Tiramide, Tolectin, Tolmetin, Zomax and Zomepirac), arylbutyric acid ferivatives (e.g., Bumadizon, Butibufen, Fenbufen and Xenbucin) arylcarboxylic acids (e.g., Clidanac, Ketorolac and Tinoridine), arylproprionic acid derivatives (e.g., Alminoprofen, Benoxaprofen, Bucloxic Acid, Carprofen, Fenoprofen, Flunoxaprofen, Flurbiprofen, Ibuprofen, Ibuproxam, Indoprofen, Ketoprofen, Loxoprofen, Miroprofen, Naproxen, Oxaprozin, Piketoprofen, Piroprofen, Pranoprofen, Protinizinic Acid, Suprofen and Tiaprofenic Acid), pyrazoles (e.g., Difenamizole and Epirizole), pyrazolones (e.g., Apazone, Benzpiperylon, Feprazone, Mofebutazone, Morazone, Oxyphenbutazone, Phenylbutazone, Pipebuzone, Propyphenazone, Ramifenazone, Suxibuzone and Thiazolinobutazone), salicyclic acid derivatives (e.g., Acetaminosalol, 5-Aminosalicylic Acid, Aspirin, Benorylate, Biphenyl Aspirin, Bromosaligenin, Calcium Acetylsalicylate, Diflunisal, Etersalate, Fendosal, Flufenisal, Gentisic Acid, Glycol Salicylate, Imidazole Salicylate, Lysine Acetylsalicylate, Mesalamine, Morpholine Salicylate, 1-Naphthyl Sallicylate, Olsalazine, Parsalmide, Phenyl Acetylsalicylate, Phenyl Salicylate, 2- Phosphonoxybenzoic Acid, Salacetamide, Salicylamide O-Acetic Acid, Salicylic

Acid, Salicyloyl Salicylic Acid, Salicylsulfuric Acid, Salsalate and Sulfasalazine), thiazinecarboxamides (e.g., Droxicam, Isoxicam, Piroxicam and Tenoxicam), e- Acetamidocaproic Acid, S-Adenosylmethionine, 3-Amino-4-hydroxybutyric Acid, Amixetrine, Bendazac, Benzydamine, Bucolome, Difenpiramide, Ditazol, Emorfazone, Guaiazulene, Ketorolac, Meclofenamic Acid, Mefenamic Acid, Nabumetone, Nimesulide, Orgotein, Oxaceprol, Paranyline, Perisoxal, Pifoxime, Piroxicam, Proquazone, Tenidap and a COX-2 inhibitor (e.g., Rofecoxib, Valdecoxib and Celecoxib).

Further examples of anti-inflammatory agents include aspirin, a sodium salicylate, choline magnesium trisalicylate, salsalate, diflunisal, sulfasalazine, olsalazine, a para-aminophenol derivatives, an indole, an indene acetic acid, a heteroaryl acetic acid, an anthranilic acid, an enolic acid, an alkanones, a diaryl- substituted furanone, a diaryl-substituted pyrazoles, an indole acetic acids, or a sulfonanilide.

In some embodiments, the compounds of the present invention can be administered in combination with immunotherapeutic agents such as interferons and anti-integrin blocking antibodies like natalizumab.

Examples of agents suitable for treating demyelinating disorders include Pirfenidone, Epalrestat, Nefazodone hydrochloride, Memantine hydrochloride, Mitoxantrone hydrochloride, Mitozantrone hydrochloride, Thalidomide, Roquinimex, Venlafaxine hydrochloride, Intaxel, Paclitaxel, recombinant human nerve growth factor; nerve growth factor, ibudilast, Cladribine, Beraprost sodium, Levacecarnine hydrochloride; Acetyl-L-carnitine hydrochloride; Levocarnitine acetyl hydrochloride, Droxidopa, interferon alfa, natural interferon alpha, human lymphoblastoid interferon, interferon beta- Ib, interferon beta-Ser, Alemtuzumab, Mycophenolate mofetil, Zoledronic acid monohydrate, Adapalene, Eliprodil, Donepezil hydrochloride, Dexanabinol, Dexanabinone, Xaliproden hydrochloride, interferon alfa-n3, lipoic acid, thioctic acid, Teriflunomide, Atorvastatin, Pymadin, 4-Aminopyridine, Fampridine, Fidarestat, Priliximab, Pixantrone maleate, Dacliximab, Daclizumab, Glatiramer acetate, Rituximab, Fingolimod hydrochloride, interferon beta- Ia, Natalizumab, Abatacept, Temsirolimus, Lenercept, Ruboxistaurin mesilate hydrate,

Dextromethorphan/quinidine sulfate, Capsaicin, Dimethylfumarate or Dronabinol/cannabidiol.

In some embodiments, the compounds of the present invention can be administered in combination with one or more other pharmaceutically active agents that are effective against multiple sclerosis. Examples of such agents include the interferons (interferon beta 1-a, beta 1-b, and alpha), glatiramer acetate or corticosteroids such as methylprednisolone and prednisone as well as chemotherapeutic agents such as mitoxantrone, methotrexate, azathioprine, cladribine cyclophosphamide, cyclosporine and tysabri.

Further examples of pharmaceutically active agents that are effective against multiple sclerosis and are suitable to be administered in combination with compounds of the present invention include compounds of the following structural formulae:

Further examples of pharmaceutical agents that can be co-administered with the compounds of formula (I) or formula (Ia) or formula (II) or formula (Ha) include:

T-cell receptor (TCR) Vβ6 CDR2 peptide vaccine consisting of TCR Vβ6, amino acid sequence 39-58, Leu - GIy - GIn - GIy - Pro - GIu - Phe - Leu - Thr - Tyr Phe - GIn - Asn - GIu - Ala - GIn - Leu - GIu - Lys - Ser (SEQ ID NO: 1);

Myelin basic protein immunogen peptide, aminoacid sequence 75-95, Lys - Ser - His - GIy - Arg - Thr - GIn - Asp - GIu - Asn - Pro - VaI - VaI - His - Phe - Phe • Lys - Asn - He - VaI - Thr (SEQ ID N0:2);

Tiplimotide, myelin basic protein immunogen vaccine peptide, aminoacid sequence 83-99, D - Ala - lys - pro - val - val - his - leu - phe - ala - asp - ile - val - thr - pro - arg - thr - pro, (SEQ ID NO:3);

Myelin basic protein immunogen peptide, aminoacid sequence 82-98, Asp - glu - asp - pro - val - val - his - phe - phe - lys - asp - ile - val - thr - pro - arg - thr, (SEQ ID NO:4);

Adrenocorticotropic hormone (ACTH), Ser - Tyr - Ser - met - glu - his - phe - arg - try - gly - lys - pro - val - gly - lys- lys - arg - arg - pro - val - lys - val - tyr- pro - asp - gly - ala - glu - asp - glu - leu - ala - glu - ala - phe - pro - leu - glut - phe, (SEQ ID N0:5).

Further examples of pharmaceutically active agents that are effective against multiple sclerosis and are suitable to be administered in combination with compounds of the present invention include:

3-4 diaminopyridine; ABT-874; Actos® (pioglitazone); ALCAR (acetyl-L- camitine); Alpha lipoic acid; AndroGel® (testosterone gel); combination of trimethoprim and vitamin C; combination of azithromycin and rifampin; minocycline; donezepil HCL; Avandia® (rosiglitazone maleate; combination of IFN beta- Ia) and acetaminophen, ibuprofen or prednisone; combination of Avonex® (interferon beta- Ia) + CellCept®

(mycophenolate mofetil); combination of Avonex® (interferon beta- Ia) and Copaxone® (glatiramer acetate); combination of Avonex® (interferon beta- Ia) and doxycycline; combination of Avonex® (interferon beta-1 a) and EMLA (lidocaine and prilocaine) anesthetic cream; Avonex® (interferon beta- Ia) and estrogen and progesterone; combination of Avonex® (interferon beta- Ia) + Fludara® (fludarabine phosphate); combination of Avonex® (interferon beta- Ia) and methotrexate and leucovorin rescue; combination of Avonex® (interferon beta- Ia) and methotrexate and methylprednisolone; combination of Avonex® (interferon beta- Ia) and Novantrone® (mitoxantrone); combination of Avonex® (interferon beta- Ia) and Prozac® (fluoxetine); combination of Avonex® (interferon beta- Ia) and Topamax® (topiramate); combination of Avonex® (interferon beta- Ia) and Zocor® (simvastatin); AVP-923 (dextromethorphan/quinidine); combination of Betaseron®

(interferon beta- Ib) and Imuran® (azathioprine); combination of Betaseron® (interferon beta-lb) and Copaxone® (glatiramer acetate); combination of BHT-3009- 01 and Lipitor® (atorvastatin); Bone marrow/peripheral stem cell transplant; CellCept® (mycophenolate mofetil); combination of CellCept® (mycophenolate mofetil) and Avonex® (interferon beta- Ia); Oral cladribine; CNTO 1275 (monoclonal antibody); combination of Copaxone® (glatiramer acetate) and Antibiotic therapy (minocycline); combination of Copaxone® (glatiramer acetate) and Novantrone® (mitoxantrone); combination of Copaxone® (glatiramer acetate) and prednisone; combination of Copaxone® (glatiramer acetate) and Proventil® (albuterol); Cyclophosphamide; Daclizumab; Deskar® (pirfenidone); Estriol; Fumaric acid esters; Gabitril® (tiagabine HCL); Ginkgo biloba; IDEC-131 (anti-CD40L or anti-CD 154); the combination of Immunoglobulin and methylprednisolone; Inosine; Interferon tau; Lamictal® (lamotrigine); Lexapro® (escitalopram); Lipitor® (atorvastatin); combination of Lipitor® (atorvastatin) and Rebif® (interferon beta- Ia); combination of Lymphocytapheresis (removal of immune cells), Imuran® (azathioprine) and prednisone; MBP8298; Methylprednisolone; combination of Methylprednisolone and Avonex (interferon beta-la); Modiodal (modafinil); NBI-5788 (altered peptide ligand); combination of Novantrone® (mitoxantrone for injection concentrate) and Avonex® (Interferon beta-la) or Copaxone® (glatiramer acetate); Omega-3 Fatty Acid Supplementation; Pixantrone (BBR 2778); combination of Provigil® (modafinil) and Avonex® (interferon beta- Ia); Rapamune® (sirolimus); RG2077; Rituxan® (rituximab); Rolipram (phosphodiesterase-4 inhibitor); SAIK-MS (laquinimod, ABR- 215062); T cell vaccination; Teriflunomide; Tetrahydrocannabinol; Tetrahydrocannabinol (dronabinol); Thalamic stimulation; combination of Tysabri® (natalizumab) and Avonex® (interferon beta- Ia); combination of Tysabri® (natalizumab) and Copaxone® (glatiramer acetate); and Viagra® (sildafenil citrate).

Further example of pharmaceutically active agents that are effective against multiple sclerosis and are suitable to be administered in combination with compounds of the present invention is Copaxone (Glatiramer), which can be orally coadministered with the compounds of the present invention.

In other embodiments, pharmaceutically active agents that are effective against multiple sclerosis and are suitable to be administered in combination with compounds of the present invention include compounds include: Mylinax, an oral formulation of cladrlbine used in leukaemia treatment, developed by Serono/Ivex; Teriflunomide, a metabolite of Arava, an oral immunosuppressant, developed by Sanofl-Aventis; FTY 720, an oral immunomodulator (Sphingosine-1 -phosphate receptor agonist), developed by Novartis; MBP 8298, a synthetic myelin basis protein designed to reduce the emergence of antibodies directed against the myelin, developed by Bio MS Medical; an orphan drug 4-aminopyridline (4-AP), a potassium channel blocker, developed by Acorda; Gamunex, an intravenous immunoglobulin formulation, developed by Bayer; BG- 12 fumarate, a second generation oral futnarate, developed by Biogen Idec/Fumapharm; Temsirolimus, a T-lymphocytes proliferation blocker, developed by Wyeth; E-2007, an AMPA receptor agonist, developed by Eisal; Campath, a humanized antibody directed against CD52, developed by Genzyme; Neuro Vax, a vaccine, developed by Immune Response; Zocor, a statin, developed by Merck; NBI 5788, a myelin-mimicking peptide ligand, developed by Neurocrine; Tauferon, Interferon tau, developed by Pepgen; Zenapax, a humanized anti-CD25 immunosuppressive antibody, developed by Protein Design; a combination of MS- IET and EMZ 701, a methyl donator, developed by Transition Therapeutics; Laquinlmod, an oral formulation of a derivative of linomide, developed by Active Biotech/Teva; deskar pirfenidone, a TNF-alpha inhibitor, developed by Mamac; ATL- 1102, a second generation antisense inhibitor targeting VLA4, developed by Antisense Therapeutics.

In some embodiments, compounds of formula (A) can be administered in combination with antivascular agents, in particular agents inhibiting the growth factor receptors, Epidermal Growth Factor Receptor (EGFR), Vascular Epidermal Growth Factor Receptor (VEGFR), and Fibroblast Growth Factor Receptor (FGFR). Examples of such agents include, Iressa, Tarceva, Erbitux, Pelitinib, AEE-788, CP- 547632, CP-547623, Tykerb (GW-2016), INCB-7839, ARRY-334543, BMS-599626, BIBW-2992, Falnidamol, AGl 517, E-7080, KRN-951, GFKI-258, BAY-579352, CP- 7055, CEP-5214, Sutent, Macugen, Nexavar, Neovastat, Vatalanib succinate, GW-

78603413, Lucentis, Teavigo, AG-13958, AMG-706, Axitinib, ABT-869, Evizon, Aplidin, NM-3, PI-88, Coprexa, AZD-2171, XL-189, XL-880, XL-820, XL-647, ZK- CDK, VEGFTrap, OSI-930, Avastin, Revlimid, Endostar, Linomide, Xinlay, SU-668, BIBF-1120, BMS-5826624, BMS-540215.

In some embodiments, compounds of formula (I) or formula (Ia) or formula (II) or formula (Ha) can be administered in combination with agents that affect T-cell homing, extravastion and transmigration. Examples of such agents include, FTY- 720PKI-166, PTK-787, SU-11248.

In some embodiments, compounds of formula (I) or formula (Ia) or formula (II) or formula (Ha) can be administered in combination with agents inhibiting VLA- 4. Examples of such agents include, Tysabri, Bio-1211. HMR-1031, SB-683698, RBx-4638,RO-0272441, RBx-7796,SB-683699, DW-908e, AJM-300, and PS- 460644.

In certain embodiments, the compound of formula (I) or formula (Ia) or formula (II) or formula (Ha) can be administered alone or in combination with an anticancer agent.

As used herein, the term "combination" with reference to pharmaceutically active agents and the term "co-administering" and "co-administration" refer to administering more than one pharmaceutically active agent to a patient during one treatment cycle and not necessarily simultaneous or in a mixture.

Anti-cancer agents that can be employed in combination with the compounds of the invention include Taxol™ (also referred to as "paclitaxel", and compounds that have the basic taxane skeleton), Adriamycin, Dactinomycin, Bleomycin, Vinblastine, Cisplatin, acivicin; aclarubicin; acodazole hydrochloride; acronine; adozelesin; aldesleukin; altretamine; ambomycin; ametantrone acetate; aminoglutethimide; amsacrine; anastrozole; anthramycin; asparaginase; asperlin; azacitidine; azetepa; azotomycin; batimastat; benzodepa; bicalutamide; bisantrene hydrochloride; bisnafide dimesylate; bizelesin; bleomycin sulfate; brequinar sodium; bropirimine; busulfan; cactinomycin; calusterone; caracemide; carbetimer; carboplatin; carmustine; carubicin hydrochloride; carzelesin; cedefingol; chlorambucil; cirolemycin; cladribine; crisnatol mesylate; cyclophosphamide; cytarabine; dacarbazine; daunorubicin hydrochloride;

decitabine; dexormaplatin; dezaguanine; dezaguanine mesylate; diaziquone; doxorubicin; doxorubicin hydrochloride; droloxifene; droloxifene citrate; dromostanolone propionate; duazomycin; edatrexate; eflornithine hydrochloride; elsamitrucin; enloplatin; enpromate; epipropidine; epirubicin hydrochloride; erbulozole; esorubicin hydrochloride; estramustine; estramustine phosphate sodium; etanidazole; etoposide; etoposide phosphate; etoprine; fadrozole hydrochloride; fazarabine; fenretinide; floxuridine; fludarabine phosphate; fluorouracil; flurocitabine; fosquidone; fostriecin sodium; gemcitabine; gemcitabine hydrochloride; hydroxyurea; idarubicin hydrochloride; ifosfamide; ilmofosine; interleukin II (including recombinant interleukin II, or rIL2), interferon alfa-2a; interferon alfa-2b; interferon alfa-nl ; interferon alfa-n3; interferon beta-I a; interferon gamma-I b; iproplatin; irinotecan hydrochloride; lanreotide acetate; letrozole; leuprolide acetate; liarozole hydrochloride; lometrexol sodium; lomustine; losoxantrone hydrochloride; masoprocol; maytansine; mechlorethamine hydrochloride; megestrol acetate; melengestrol acetate; melphalan; menogaril; mercaptopurine; methotrexate; methotrexate sodium; metoprine; meturedepa; mitindomide; mitocarcin; mitocromin; mitogillin; mitomalcin; mitomycin; mitosper; mitotane; mitoxantrone hydrochloride; mycophenolic acid; nocodazole; nogalamycin; ormaplatin; oxisuran; pegaspargase; peliomycin; pentamustine; peplomycin sulfate; perfosfamide; pipobroman; piposulfan; piroxantrone hydrochloride; plicamycin; plomestane; porfimer sodium; porfiromycin; prednimustine; procarbazine hydrochloride; puromycin; puromycin hydrochloride; pyrazofurin; riboprine; rogletimide; safingol; safingol hydrochloride; semustine; simtrazene; sparfosate sodium; sparsomycin; spirogermanium hydrochloride; spiromustine; spiroplatin; streptonigrin; streptozocin; sulofenur; talisomycin; tecogalan sodium; tegafur; teloxantrone hydrochloride; temoporfin; teniposide; teroxirone; testolactone; thiamiprine; thioguanine; thiotepa; tiazofurin; tirapazamine; toremifene citrate; trestolone acetate; triciribine phosphate; trimetrexate; trimetrexate glucuronate; triptorelin; tubulozole hydrochloride; uracil mustard; uredepa; vapreotide; verteporfin; vinblastine sulfate; vincristine sulfate; vindesine; vindesine sulfate; vinepidine sulfate; vinglycinate sulfate; vinleurosine

sulfate; vinorelbine tartrate; vinrosidine sulfate; vinzolidine sulfate; vorozole; zeniplatin; zinostatin and zorubicin hydrochloride.

Other anti-cancer drugs that can be employed in combination with the compounds described herein include: 20-epi-l,25 dihydroxyvitamin D3; 5- ethynyluracil; abiraterone; aclarubicin; acylfulvene; adecypenol; adozelesin; aldesleukin; ALL-TK antagonists; altretamine; ambamustine; amidox; amifostine; aminolevulinic acid; amrubicin; amsacrine; anagrelide; anastrozole; andrographolide; angiogenesis inhibitors; antagonist D; antagonist G; antarelix; anti-dorsalizing moφhogenetic protein- 1; antiandrogen, prostatic carcinoma; antiestrogen; antineoplaston; antisense oligonucleotides; aphidicolin glycinate; apoptosis gene modulators; apoptosis regulators; apurinic acid; ara-CDP-DL-PTBA; arginine deaminase; asulacrine; atamestane; atrimustine; axinastatin 1 ; axinastatin 2; axinastatin 3; azasetron; azatoxin; azatyrosine; baccatin III derivatives; balanol; batimastat; BCR/ ABL antagonists; benzochlorins; benzoylstaurosporine; beta lactam derivatives; beta-alethine; betaclamycin B; betulinic acid; bFGF inhibitor; bicalutamide; bisantrene; bisaziridinylspermine; bisnafide; bistratene A; bizelesin; breflate; bropirimine; budotitane; buthionine sulfoximine; calcipotriol; calphostin C; camptothecin derivatives; canarypox IL-2; capecitabine; carboxamide-amino-triazole; carboxyamidotriazole; CaRest M3; CARN 700; cartilage derived inhibitor; carzelesin; casein kinase inhibitors (ICOS); castanospermine; cecropin B; cetrorelix; chlorlns; chloroquinoxaline sulfonamide; cicaprost; cis-porphyrin; cladribine; clomifene analogues; clotrimazole; collismycin A; collismycin B; combretastatin A4; combretastatin analogue; conagenin; crambescidin 816; crisnatol; cryptophycin 8; cryptophycin A derivatives; curacin A; cyclopentanthraquinones; cycloplatam; cypemycin; cytarabine ocfosfate; cytolytic factor; cytostatin; dacliximab; decitabine; dehydrodidemnin B; deslorelin; dexamethasone; dexifosfamide; dexrazoxane; dexverapamil; diaziquone; didemnin B; didox; diethylnorspermine; dihydro-5- azacytidine; 9- dioxamycin; diphenyl spiromustine; docosanol; dolasetron; doxifluridine; droloxifene; dronabinol; duocarmycin SA; ebselen; ecomustine; edelfosine; edrecolomab; eflornithine; elemene; emitefur; epirubicin; epristeride; estramustine analogue; estrogen agonists; estrogen antagonists; etanidazole; etoposide

phosphate; exemestane; fadrozole; fazarabine; fenretinide; filgrastim; finasteride; flavopiridol; flezelastine; fluasterone; fludarabine; fluorodaunorunicin hydrochloride; forfenimex; formestane; fostriecin; fotemustine; gadolinium texaphyrin; gallium nitrate; galocitabine; ganirelix; gelatinase inhibitors; gemcitabine; glutathione inhibitors; hepsulfam; heregulin; hexamethylene bisacetamide; hypericin; ibandronic acid; idarubicin; idoxifene; idramantone; ilmofosine; ilomastat; imidazoacridones; imiquimod; immunostimulant peptides; insulin-like growth factor- 1 receptor inhibitor; interferon agonists; interferons; interleukins; iobenguane; iododoxorubicin; ipomeanol, 4-; iroplact; irsogladine; isobengazole; isohomohalicondrin B; itasetron; jasplakinolide; kahalalide F; lamellarin-N triacetate; lanreotide; leinamycin; lenograstim; lentinan sulfate; leptolstatin; letrozole; leukemia inhibiting factor; leukocyte alpha interferon; leuprolide+estrogen+progesterone; leuprorelin; levamisole; liarozole; linear polyamine analogue; lipophilic disaccharide peptide; lipophilic platinum compounds; lissoclinamide 7; lobaplatin; lombricine; lometrexol; lonidamine; losoxantrone; lovastatin; loxoribine; lurtotecan; lutetium texaphyrin; lysofylline; lytic peptides; maitansine; mannostatin A; marimastat; masoprocol; maspin; matrilysin inhibitors; matrix metalloproteinase inhibitors; menogaril; merbarone; meterelin; methioninase; metoclopramide; MIF inhibitor; mifepristone; miltefosine; mirimostim; mismatched double stranded RNA; mitoguazone; mitolactol; mitomycin analogues; mitonafide; mitotoxin fibroblast growth factor-saporin; mitoxantrone; mofarotene; molgramostim; monoclonal antibody, human chorionic gonadotrophin; monophosphoryl lipid A+myobacterium cell wall sk; mopidamol; multiple drug resistance gene inhibitor; multiple tumor suppressor 1 -based therapy; mustard anticancer agent; mycaperoxide B; mycobacterial cell wall extract; myriaporone; N-acetyldinaline; N-substituted benzamides; nafarelin; nagrestip; naloxone+pentazocine; napavin; naphterpin; nartograstim; nedaplatin; nemorubicin; neridronic acid; neutral endopeptidase; nilutamide; nisamycin; nitric oxide modulators; nitroxide antioxidant; nitrullyn; O6-benzylguanine; octreotide; okicenone; oligonucleotides; onapristone; ondansetron; ondansetron; oracin; oral cytokine inducer; ormaplatin; osaterone; oxaliplatin; oxaunomycin; palauamine; palmitoylrhizoxin; pamidronic acid; panaxytriol; panomifene; parabactin;

pazelliptine; pegaspargase; peldesine; pentosan polysulfate sodium; pentostatin; pentrozole; perflubron; perfosfamide; perillyl alcohol; phenazinomycin; phenylacetate; phosphatase inhibitors; picibanil; pilocarpine hydrochloride; pirarubicin; piritrexim; placetin A; placetin B; plasminogen activator inhibitor; platinum complex; platinum compounds; platinum-triamine complex; porfimer sodium; porfiromycin; prednisone; propyl bis-acridone; prostaglandin J2; proteasome inhibitors; protein A-based immune modulator; protein kinase C inhibitor; protein kinase C inhibitors, microalgal; protein tyrosine phosphatase inhibitors; purine nucleoside phosphorylase inhibitors; purpurins; pyrazoloacridine; pyridoxylated hemoglobin polyoxyethylene conjugate; raf antagonists; raltitrexed; ramosetron; ras farnesyl protein transferase inhibitors; ras inhibitors; ras-GAP inhibitor; retelliptine demethylated; rhenium Re 186 etidronate; rhizoxin; ribozymes; RII retinamide; rogletimide; rohitukine; romurtide; roquinimex; rubiginone Bl ; ruboxyl; safingol; saintopin; SarCNU; sarcophytol A; sargramostim; Sdi 1 mimetics; semustine; senescence derived inhibitor 1 ; sense oligonucleotides; signal transduction inhibitors; signal transduction modulators; single chain antigen-binding protein; sizofiran; sobuzoxane; sodium borocaptate; sodium phenylacetate; solverol; somatomedin binding protein; sonermin; sparfosic acid; spicamycin D; spiromustine; splenopentin; spongistatin 1 ; squalamine; stem cell inhibitor; stem-cell division inhibitors; stipiamide; stromelysin inhibitors; sulfinosine; superactive vasoactive intestinal peptide antagonist; suradista; suramin; swainsonine; synthetic glycosaminoglycans; tallimustine; tamoxifen methiodide; tauromustine; tazarotene; tecogalan sodium; tegafur; tellurapyrylium; telomerase inhibitors; temoporfin; temozolomide; teniposide; tetrachlorodecaoxide; tetrazomine; thaliblastine; thiocoraline; thrombopoietin; thrombopoietin mimetic; thymalfasin; thymopoietin receptor agonist; thymotrinan; thyroid stimulating hormone; tin ethyl etiopurpurin; tirapazamine; titanocene bichloride; topsentin; toremifene; totipotent stem cell factor; translation inhibitors; tretinoin; triacetyluridine; triciribine; trimetrexate; triptorelin; tropisetron; turosteride; tyrosine kinase inhibitors; tyrphostins; UBC inhibitors; ubenimex; urogenital sinus-derived growth inhibitory factor; urokinase receptor antagonists; vapreotide; variolin B; vector system, erythrocyte gene therapy; velaresol; veramine;

verdins; verteporfin; vinorelbine; vinxaltine; vitaxin; vorozole; zanoterone; zeniplatin; zilascorb; and zinostatin stimalamer. Preferred anti-cancer drugs are 5-fluorouracil and leucovorin.

Other chemotherapeutic agents that can be employed in combination with the compounds of the invention include but are not limited to alkylating agents, antimetabolites, natural products, or hormones.

The invention is illustrated by the following examples, which are not intended to be limiting in any way. The skilled practioner will be able to exercise routine judgment for the selection of suitable starting materials in order to prepare specific products, the order of synthetic steps, and the need for protecting groups for remote functionalities.

EXEMPLIFICATION

Example 1 Synthesis of the Compounds of the Invention

Compounds of formula (I) or formula (Ia) and formula (II) or formula (Ha) can be synthesized according to the following examples. It is understood by those skilled in the art of organic synthesis that the substitution patterns of the starting materials determines the substitution patterns of the products, and the skilled practioner will be able to exercise routine judgment for the selection of suitable starting materials in order to prepare specific products, the order of synthetic steps, and the need for protecting groups for remote functionalities.

The chemistry of protecting groups can be found, for example, in Greene, et al., Protective Groups in Organic Synthesis, 2d. Ed., Wiley & Sons, 1991 , the entire disclosure of which is herein incorporated by reference.

Standard synthetic methods and procedures for the preparation of organic molecules and functional group transformations and manipulations can be readily obtained from the relevant scientific literature or from standard textbooks in the field. It will be appreciated that where typical or preferred process conditions (i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given, other process conditions can also be used unless otherwise stated. Optimum reaction

conditions may vary with the particular reactants or solvent used, but one skilled in the art can determine such conditions by routine optimization procedures.

(A) 2-Fluoro-5-nitro-6-(lH-indazol-5-ylamino)benzoic acid

The starting material 3-nitro-2,6-difluorobenzoic acid was prepared by nitration of 2,6-difluorobenzoic acid with potassium nitrate in sulfuric acid as described in part by Kuramoto Y, Ohshita Y, Yoshida J, Yazaki A, Shiro M. and Koike T, J. Med. Chem. (2003), 46:1905-1917) and isolated as described in a similar preparation by Yoshida Y, Barrett D, Azami H, Morinaga C, Matsumoto S, Matsumoto Y, and Takasugi H, (1999), Bioorg. Med. Chem. 7:2647-2666. 3-Nitro- 2,6-difluorobenzoic acid (20.Og, 98.44mmol) was added to a solution of ethanol (10OmL) and water (10OmL). The acid solution was cooled to 1O 0 C and triethylamine (25.09mL) added by drops under rapid stirring to ensure the temperature did not exceed 40 0 C. 5-Aminoindazole (13.01g, 98.44mmol) was then added in portions and the combined mixture heated to 70 0 C for 16 hours. A solution of water (10OmL) and concentrated HCl (10OmL) was prepared, heated to 60 0 C and placed under vigorous stirring. The reaction mixture, still at 7O 0 C, was transferred to the HCl/water solution in small portions and allowed to cool to room temperature. The mixture was stirred for a further 4 hours to ensure maximum precipitation. The resulting precipitate was filtered off and washed with water (2x60mL) and dried in a vacuum oven overnight (28.3g, 89.56mmol, 94%). 1 H δ (rfd-DMSO): 6.97 (IH, dd, ArH, J=8.8, 2.4Hz), 7.18 (IH, d, ArH, J=2.4ηz), 7.43 (IH, d, ArH, J=8.8ηz), 7.45 (IH, d, ArH, J=8.8ηz), 8.10 (IH, d, ArH, J=8.8ηz), 8.99 (IH, s, ArH) 5 9.15 (1η, s, NH), 9.87 (1η, s, NH); ηPLC: R t =3.52 min.; LRMS: m/z=315.4 (M-H)

(B) 1 -Fluoro-4-nitropyrazolo[7,8a]-acridin-9(l 0H)-one

A suspension of 2-fluoro-5-nitro-6-(lH-indazol-5-ylamino)benzoic acid (20.Og, 65.096mmol) in chloroform under argon had freshly distilled POCl 3 (24.3mL, 260.383mmol) added to it. The mixture was heated to 8O 0 C for 16 hours. Once cooled, ethanol (5OmL) was added slowly to quench the excess POCl 3 reaction and then stirred for 30 minutes at room temperature. At this point, the mixture was reduced to dryness under vacuum. Water (10OmL) was added to the residue and saturated sodium bicarbonate solution added until the pH=8. The mixture was then stirred for 60 minutes at room temperature and the precipitate filtered off, washed with water (2x80mL) and dried overnight in a vacuum oven (19.219g, 64.445mmol, 99%). 1 H δ (</6-DMSO): NMR unavailable since compound very insoluble; HPLC: R t =6.22 min.; LRMS: m/z=299.1 (M+H)

(C) l-Fluoro-4-nitropyrazolo[7,8a]-imidazo[4,5,l-de]-acridin-9(1 0H)-one (Compound XVIII)

(XVIII)

To a slurry of starting material (7.1 12g, 23.846mmol) in formic acid (75mL), was added SnCl 2 .2H 2 O (23.07g, 102.25mmol) in concentrated HCl (15mL). The combined mixture was stirred at room temperature for 30 minutes then heated to 95 0 C for 20 hours. Once cooled, the solid was filtered off and partially dried before being slurried in saturated sodium bicarbonate solution and stirred for 60 mins. The precipitate was then filtered off and washed with water (2x10OmL) and dried in a vacuum oven overnight (6.303g, 22.654mmol, 95%).

(D) 4-Amino- 1 -fluoropyrazolo[7,8a]-acridin-9(l 0H)-one

l-Fluoro-4-nitropyrazolo[7,8a]-acridin-9(10H)-one (1Og, 33.5mmol) in ethanol (10OmL) had a solution of tin chloride dihydrate (22.7g, 100.5mmol) in concentrated HCl (3OmL). The mixture was heated to 80°C for 25 hours until the t.l.c. showed no starting material remained. The mixture was cooled, the solid filtered off and washed with water (4x10OmL) then dried under vacuum. 1 H δ (</6-DMSO): 5.60-6.20 (4H, br, NH . HCl), 6.96 (1η, dd, ArH, J=8.8, 8.4ηz), 7.50 (IH, d, ArH, J=4.4ηz), 7.90 (IH, d, ArH, J=9.2ηz), 8.03 (IH, d, ArH, J=9.2ηz), 8.81 (IH, s, ArH), 1 1.89 (1η, s, NH); ηPLC: R,=3.32 min.; LRMS: m/z=277.2 (M-H).

(E) l-Fluoro-4-nitropyrazolo[7,8a]-imidazo[4,5,l-de]-acridin-9(1 0H)-one (Compound XVIII)

(XVIII)

4-Amino-l-fluoropyrazolo[7,8a]-acridin-9(10H)-one (8g, 29.8mmol) in formic acid (10OmL) and concentrated HCl (2OmL) was heated to 95°C for 35 hours. The mixture was cooled and the solid filtered off, washed with water (4x10OmL) and dried under vacuum. No further purification was carried out.

(F) Compound (V)

(XVIII)

Compound XVIII (0.294g, 1.057mmol), 2-morρholine ethylamine (0.22Og, 1.961mmol, 0.222mL), diisopropylethylamine (0.273g, 2.1 14mmol, 0.368mL) and dimethylacetamide (2.OmL) were combined in a 5mL reaction tube and heated to 15O 0 C under microwave irradiation for 20 minutes. Once cooled, the solvent was removed and the residue columned over silica gel (0-20% MeOH in CHCl 3 ). The desired fractions were pooled and the solvent removed to yield the target compound. 1 U δ (</6-DMSO): 2.41 (2H, m, CH 2 NCH 2 ), 2.68 (2H, m, CH 2 ), 3.19 (2H, m, CH 2 NCH 2 ), 3.36 (2H, m, NHCH 2 ), 3.63 (4H, m, CH 2 O CH 2 ), 6.89 (1η, d, ArH, J=8.6ηz), 8.01 (IH, d, ArH, J=8.6ηz), 8.16 (IH, d, ArH, J=9.0), 8.44 (1η, d, ArH, J=9.0ηz), 9.00 (IH, s, Pyrazole-H), 9.07 (1η, m, NH), 9.29 (1η, s, Imidazole-H), 13.69 (1η, s, N=NH); ηPLC: R,=3.55min.; LRMS: 389.0 (M+l).

(G) l-(3-Dimethylamino-propylamino)-4-nitropyrazolo[7,8a]-acridi n-9(10η)-one

To a stirred slurry of l-fluoro-4-nitropyrazolo[7,8a]-acridin-9(10H)-one

(8.75g, 29.34mmol) in DMF (22OmL) was added N,N-dimethyl-l,3-propanediamine (8.99g, 88.02mmol, 3 equiv). The mixture was stirred at room temperature for 18 hours and then concentrated to dryness. The residue was treated with aqueous sodium bicarbonate solution (40OmL) and stirred for 15 minutes. The precipitate was collected by filtration, rinsed with water and ethyl acetate. The filter cake was vacuum-dried to afford the title compound (8.1 Ig, 72% yield) as a yellow solid. 1 H δ

(400 MHz, DMSO-(I 6 ): 13.60 (IH, broad s), 12.68 (IH, s), 12.13 (IH, t, J= 6.5 Hz), 8.78 (IH, s), 8.35 (IH, d, J= 9.8 Hz), 7.97 (IH, m), 7.91 (IH, m), 6.58 (IH, d, J= 9.8 Hz), 3.49 (2H, m), 2.37 (2H, t, J = 6.7 Hz), 2.18 (6H, s), 1.83 (2H, m); LRMS: m/z = 381.4 (M+H).

(H) Compound (IV)

To a stirred solution of l-(3-dimethylamino-propylamino)-4- nitropyrazolo[7,8a]-acridin-9(10H)-one (8.05g, 21.10mmol) in formic acid (25OmL) was added a solution of tin (II) chloride dihydrate (21.43g, 94.95mmol) in 37% hydrochloric acid (28mL) at room temperature. The mixture was heated at a gentle reflux for 22 hours and then concentrated to dryness. The resulting solid was dissolved in water, basified with an excess of aqueous sodium bicarbonate (1.2L), and stirred 30 minutes for granulation. The precipitate was collected by filtration, rinsed with water, and vacuum-dried to give the crude product which contained inorganic impurities (tin hydroxides). To remove the inorganic impurities, the crude product was filtered through a silica gel pad, eluting with DMF, affording the title compound (3.12g, 41% yield) as an orange-yellow solid.

Representative compounds with the fused pyrazole scaffold are prepared in a similar manner. For example compounds (III) and (IV) were prepared by the addition of 5-N,N-dimethylamino pentylamine and 3-N,N-dimethylamino propylamine to compound (VI), following the method used for (V), namely, by treating compound (XVIII) with dialkylamino-alkyl amines using the same reaction stoichiometry shown in the model reaction with 2-morpholine ethylamine.

The resulting compounds conformed to theory as evidenced by their NMR and mass spectroscopic features, shown in the Table 1 that provides the analytical properties for compound (XVIII), and its derivatives.

TABLE 1 : Analytical characterization of representative compounds

Example 2: Determination of FLT3 and related protein tyrosine kinase inhibitory targeting in vitro

Representative compounds of formula (I) or formula (Ia) are screened for activity against FLT3 and related protein tyrosine kinase in standard pharmacological test procedures. Based on the activity shown in the standard pharmacological test procedures, the compounds of the present teachings can be useful as FLT-3 inhibitors.

The terms "IC50" and "EC50" are used interchangeably. As used herein, "EC50" refers to nMolar concentration at median percent inhibition determined by dose respose (DR) assay. As used herein, the term "ElOOO" refers to percent inhibition at 1000 nMolar determined by assay. EC50 was calculated based on dose response curve was fitted to 4 parameter Hill equation.

Testing of the compounds described in this invention was carried out using the SelectScreen™ platform from Invitrogen, Inc. (Carlsbad, CA, USA) and the details of its performance are readily viewed via the web by linking to: http://www.invitrogen.com/downloads/SelectScrn_Brochure.pdf.

Briefly, the approach is based on treating each specific kinase with a unique substrate and optical reporter system in the presence of ATP at 100 micromolar. In controls, the substrate is phosphorylated and a baseline optimal response is recorded.

Compounds were initially tested at 1000 nanomolar concentrations and the % inhibition of enzyme activity determined (ElOOO). The compounds were then re- tested by adding graded amounts of putative inhibitor which were added in 5 separate increments to generate a dose response curve. The latter is obtained by fitting to a 4 parameter Hill equation, a sigmoid saturation equation. The concentration which causes 50% enzyme inhibition (EC50) was then calculated from the dose response equation.

An effective level of inhibition in the low nanomolar range is considered to qualify the test compound as potential drug or targeting agent against the specific kinase that it has inhibited. The EC 50 value is, therefore, a measure of potency. Another important feature is specificity. It is considered a desirable property when claiming efficacy to determine how many kinases are inhibited by the same molecule.

The fewer number inhibited points toward specificity; the greater to inhibitory promiscuity.

For the compounds of this invention, the experimental conditions were as follows. The 2X FLT3/Tyr 02 peptide mixture was prepared in 50 mM HEPES pH 7.5, 0.01 BRIJ-35, 1OmM MgC12, ImM EGTA. The final 10 uL kinase reaction consists of 0.6-76.0 ng FLT3 and 2 uM Tyr 02 peptide in 50 mM HEPES pH 7.5, 0.01% BRIJ-35, 10 mM Mg C12, 1 mM EGTA. After 1 hour kinase reaction incubation, 5 uL of a 1:64 dilution of development reagent A was added.

When tested in in vitro dose response assays against FLT3, the representative compounds identified in Table 1 exhibited the EC50 and ElOOO assay values shown in Table 2. The data was derived in the SelectScreen assay and demonstrates that low nanomolar inhibitory potency is readily achieved with this new class of compounds, in comparison to five known positive controls taken from the current pharmacopoeia of compounds recognized as active inhibitors of Flt3. Notably, the compounds of the present invention have at least comparable if not 10 fold greater potency than Symadex; the properties of Symadex as a Flt3 inhibitor and immune system modulator have been described previously by Ajami, A.M., Boss, M.A. and Paterson, J. Compounds for treating autoimmune and demyelinating diseases. US Patent Appl. 2006/0189546A1. As used herein, "Symadex" and "C-131 1" are both names for the compound 5-(2-(diethylamino)ethylamino)-8-hydroxy-6H-imidazo[4,5, 1 -de]acridin- 6-one.

We have discovered that the side chain decoration of the planar core scaffold is a key feature in determining inhibitory activity.

TABLE 2: Comparative potency of indazole class compounds of Formula I and positive controls

Example 3 : Determination of FLT3 and related protein tyrosine kinase specificity in vitro

The same assay system described in Example 2 can be applied in the determination of kinase specificity. It is well understood by medicinal chemists in the development of kinase inhibitors that the resulting molecules often lack specificity. Not only are homologous kinases inhibited by the same class of compounds, but also non-homologous kinases can be affected by allosteric, induced, activated vs inactivated conformation, and protein folding interactions with the putative inhibitor, which otherwise had been designed to be specific. These off target effects are difficult to predict and can only be ruled in or out by empirically surveying a large set of kinases. It is also understood that changes to scaffolds and variation in their chemical decorations may influence the extent of promiscuity in the patterns of kinase inhibition across kinase classes.

For the compounds of this invention, a high degree of specificity as modulators of Flt3 mediated biological processes would be desirable. It would also be desirable to insure that the replacement of the phenol in Symadex with the indazole moiety should invest the new genus of molecules with similar specificity as that of their antecedent imidazoacridinine.

The following experiment was conducted to address this issue. The indazole compound (IV), described in Example 2 and representative of the new genus was

tested along with Symadex against a panel of 230 kinases representative of all known kinases classes. Included in the survey were the Flt3, its mutant, and close relatives in the Class III receptor tyrosine kinase class, together with serine-threonine kinases and atypical kinases many of which share no homology of active site with Flt3 as shown, for example, in the respective "genecard" for each kinase and available for inspection at on the web at http://www.genecards.org/.

Table 3 lists the estimated EC50 values from an abridged dose response curve for Symadex (C-131 1) and CF- 123. The kinases are listed alphabetically by Group and Family . The results show that both the molecule representative of a new molecular class embodying the indazole pharmacophore show the same pattern of kinase inhibitory specificity as the molecule with the phenolic pharmacophore. Remarkably, the compounds of the present invention are highly specific for FLT3, and to a lesser extent V561D mutant PDGFRA (60-80 nM EC 50) and CSF-IR (200- 400 nM EC50) within the broader class of tyrosine kinases identified as the "TK Group" in Table 3. Off target, and therefore unexpected activities, are also similar for both compounds, notably the inhibitory effect on the MYLK2, light chain myosin kinase, a calcium dependent, serine-threonine kinase with no active site relationship to the Flt3 kinase class.

Table 3: Kinase inhibitory activity (EC50 nM) survey for specificity: Symadex (C-131 1) compared to the indazole compound (IV).

Kinase Group Symadex (IV)

(C-1311)

AKT1 (PKB alpha) AGC >5k >5k

AKT2 (PKB beta) AGC >5k >5k

AKT3 (PKB gamma) AGC >5k >5k

CDC42 BPA (MRCKA) AGC >5k >5k

CDC42 BPB (MRCKB) AGC >5k 3632

ROCK1 AGC >5k 487

ROCK2 AGC >5k 963

ADRBK1 (BARK1. GRK2) AGC >5k >5k

ADRBK2 (BARK2, GRK3) AGC >5k >5k

GRK4 AGC >5k >5k

GRK5 AGC >5k >5k

GRK6 AGC >5k >5k

GRK7 AGC >5k >5k

PRKX AGC 3560 1399

PDK1 AGC >5k 2898

PRKACA (PKA 1 PKACA) AGC >5k >5k

PRKCA (PKC alpha) AGC >5k >5k

PRKCB1 (PKC beta I) AGC >5k >5k

PRKCB2 (PKC beta II) AGC >5k >5k

PRKCD (PKC delta) AGC >5k >5k

PRKCE (PKC epsilon) AGC >5k 2517

PRKCG (PKC gamma) AGC 1895 >5k

PRKCH (PKC eta) AGC >5k 2126

PRKCI (PKC iota) AGC >5k >5k

PRKCQ (PKC theta) AGC >5k >5k

PRKCZ (PKC zeta) AGC >5k >5k

PRKG1 (PKG1) AGC >5k >5k

PRKG2 (PKG2) AGC 2851 1908

PKN1 (PRK1) AGC 4754 1388

RPS6KA1 (RSK1 ) AGC 2071 1735

RPS6KA2 (RSK3) AGC 460 840

RPS6KA3 (RSK2) AGC 692 1489

RPS6KA4 (MSK2) AGC >5k >5k

RPS6KA5 (MSK1 ) AGC >5k >5k

RPS6KA6 (RSK4) AGC 182 655

RPS6KB1 (p70S6K) AGC >5k 2370

SGK (SGKI) AGC 2712 3453

SGK2 AGC >5k >5k

SGKL (SGK3) AGC >5k >5k

EEF2K Atypical >5k >5k

FRAP 1 (mTOR) Atypical >5k >5k

CAMKI D (CaMKI delta) CAMK 3996 2782

CAMK4 (CaMKIV) CAMK >5k >5k

CAMK2A (CaMKII alpha) CAMK >5k 789

CAMK2B (CaMKII beta) CAMK >5k >5k

CAMK2D (CaMKII delta) CAMK 2063 218

AMPK A1/B1/G1 CAMK 3478 >5k

AMPK A2/B1/G1 CAMK 1787 1572

BRSK1 (SAD1) CAMK 1934 1395

CHEK1 (CHK1) CAMK >5k >5k

MARK1 (MARK) CAMK 3191 >5k

MARK2 CAMK 2023 3488

PASK CAMK 4320 1226

DAPK3 (ZIPK) CAMK >5k 973

DCAMKL2 (DCK2) CAMK >5k >5k

MAPKAPK2 CAMK >5k >5k

MAPKAPK3 CAMK >5k >5k

MAPKAPK5 (PRAK) CAMK >5k >5k

MYLK2 (skMLCK) CAMK 41 15

PHKG1 CAMK 2204 1011

PHKG2 CAMK >5k 3716

PIM1 CAMK 959 3422

PIM2 CAMK >5k >5k

PRKCN (PKD3, PRKD3) CAMK 221 302

PRKD1 (PKC mu, PKD1) CAMK 217 494

PRKD2 (PKD2) CAMK 292 759

CHEK2 (CHK2) CAMK >5k >5k

STK22B (TSSK2) CAMK >5k >5k

STK22 D (TSSKI) CAMK 1948 2016

CSNK1A1 (CK1 alpha 1) CK1 3479 >5k

CSNK1 D (CKI delta) CK1 673 4805

CSNK1 E (CKI epsilon) CK1 1089 >5k

CSNK1G1 (CK1 gamma 1 ) CK1 >5k >5k

CSNK1G2 (CK1 gamma 2) CK1 >5k >5k

CSNK1G3 (CK1 gamma 3) CK1 >5k >5k

CDK1/cyclin B (CDC2) CMGC >5k 3947

CDK2/cyclin A CMGC >5k 822

CDK5/p25 CMGC >5k 1582

CDK5/p35 CMGC >5k 2802

CLK1 CMGC 2178 444

CLK2 CMGC 315 55

CLK3 CMGC >5k >5k

DYRK1A CMGC 708 468

DYRK1 B CMGC 991 820

DYR K3 CMGC 611 380

DYR K4 CMGC >5k >5k

HIPK1 (Myak) CMGC >5k 1215

HIPK4 CMGC 1825 1310

GSK3A (GSK3 alpha) CMGC >5k 3620

GSK3B (GSK3 beta) CMGC >5k >5k

MAPK1 (ERK2) CMGC >5k >5k

MAPK10 (JNK3) CMGC >5k 2810

MAPK11 (p38 beta) CMGC >5k >5k

MAPK12 (p38 gamma) CMGC >5k >5k

MAPK13 (p38 delta) CMGC >5k >5k

MAPK14 (p38 alpha) CMGC 2967 3787

MAPK3 (ERK1) CMGC >5k >5k

MAPK8 (JNK1) CMGC >5k >5k

MAPK9 (JNK2) CMGC >5k >5k

SRPK1 CMGC >5k >5k

SRPK2 CMGC >5k >5k

STK23 (MSSK1) CMGC >5k >5k

AURKA (Aurora A) Other >5k >5k

AURKB (Aurora B) Other >5k >5k

AURKC (Aurora C) Other >5k >5k

CSNK2A1 (CK2 alpha 1) Other >5k >5k

CSNK2A2 (CK2 alpha 2) Other 4803 >5k

IKBKB (IKK beta) Other >5k 2803

TBK1 Other >5k >5k

NEK1 Other 3381 >5k

NEK2 Other >5k >5k

NEK4 Other >5k >5k

NEK6 Other >5k >5k

NEK9 Other >5k 2927

PLK1 Other >5k >5k

PLK2 Other >5k >5k

PLK3 Other >5k >5k

MAP3K8 (COT) STE 4628 >5k

MAP4K2 (GCK) STE 4926 >5k

MAP4K4 (HGK, ZC1) STE 3132 >5k

MAP4K5 (KHS1) STE 2429 >5k

MINK1 (ZC3) STE 1088 1364

MST4 STE 1580 >5k

PAK2 (PAK65) STE >5k >5k

PAK3 STE >5k >5k

PAK4 STE >5k >5k

PAK6 STE >5k >5k

PAK7 (KIAA1264, PAK5) STE >5k >5k

STK24 (MST3) STE 733 1041

STK25 (YSK1 ) STE >5k >5k

STK3 (MST2) STE >5k >5k

STK4 (MST1 ) STE >5k 2166

TAOK2 (TAO1 ) STE >5k >5k

MAP2K1 (MEK1) STE >5k >5k

MAP2K2 (MEK2) STE >5k >5k

MAP2K6 (MKK6) STE >5k >5k

ABL1 TK >5k 3777

ABL1 E255K TK >5k 3720

ABL1 G250E TK >5k 4050

ABL1 T315I TK >5k >5k

ABL1 Y253F TK 4485 2524

ABL2 (Arg) TK 3207 2874

ALK TK 2475 4629

LTK (TYKI ) TK >5k >5k

AXL TK 3772 4442

MERTK (cMER) TK 3601 >5k

TYRO3 (RSE) TK 2438 3573

CSK TK >5k >5k

MATK (HYL, CTK) TK >5k >5k

EGFR (ErbBI) TK >5k >5k

EGFR (ErbB1 ) L858R TK >5k >5k

EGFR (ErbB1 ) L861Q TK >5k >5k

EGFR (ErbB1) T790M TK 2899 4007

ERBB2 (HER2) TK >5k >5k

ERBB4 (HER4) TK >5k >5k

EPHA1 TK >5k >5k

EPHA2 TK >5k >5k

EPHA3 TK >5k >5k

EPHA4 TK >5k >5k

EPHA5 TK >5k >5k

EPHA8 TK 3346 4151

EPHB1 TK >5k >5k

EPHB2 TK >5k >5k

EPHB3 TK >5k >5k

EPHB4 TK >5k >5k

PTK2B (FAK2, PYK2) TK >5k >5k

FER TK >5k >5k

FES (FPS) TK 1050 2254

FGFR1 TK 2388 3575

FGFR2 TK 4498 2855

FGFR3 TK >5k 3211

FGFR3 K650E TK >5k >5k

FGFR4 TK >5k >5k

IGF1 R TK >5k 1561

INSR TK >5k 2396

INSRR (IRR) TK >5k 1149

JAK1 TK >5k >5k

JAK2 TK >5k >5k

JAK2 JH1 JH2 TK >5k >5k

JAK2 JH1 JH2 V617F TK >5k >5k

JAK3 TK >5k >5k

MET (cMet) TK >5k >5k

MET M1250T TK >5k >5k

MSTI R (RON) TK >5k >5k

MUSK TK 3713 1432

CSFI R (FMS) TK 428 205

FLT3 TK 15 8

FLT3 D835Y TK 3 1

KIT TK 2250 1100

KIT T670I TK >5k >5k

PDGFRA (PDGFR alpha) TK 1234 562

PDGFRA D842V TK 1057 370

PDGFRA T674I TK >5k >5k

PDGFRA V561 D TK 83 60

PDGFRB (PDGFR beta) TK 1793 278

RET TK 1123 587

RET V804L TK 735 864

RET Y791 F TK 1237 627

ROS1 (ROS) TK >5k 2657

BLK TK 220 481

FGR TK 1268 367

FRK (PTK5) TK 2110 389

FYN TK >5k 754

HCK TK 3983 2282

LCK TK 824 345

LYN A TK 791 389

LYN B TK 812 426

PTK6 (Brk) TK >5k >5k

SRC TK 4411 437

SRC N1 TK 3154 511

SRMS (Srm) TK >5k >5k

YES1 (YES) TK >5k 1575

SYK TK 3910 >5k

ZAP70 TK >5k >5k

BMX TK >5k 3225

BTK TK 3021 1440

ITK TK >5k 637

TEK (Tie2) TK >5k >5k

NTRK1 (TRKA) TK 1434 438

NTRK2 (TRKB) TK >5k 780

NTRK3 (TRKC) TK 3725 240

FLT1 (VEGFR1) TK >5k >5k

FLT4 (VEGFR3) TK 3962 1495

KDR (VEGFR2) TK 1601 830

IRAK4 TKL 847 3321

MAP3K9 (MLK1) TKL >5k >5k

BRAF TKL 4451 1443

BRAF V599E TKL 3404 1167

RAF1 (cRAF) Y340D Y341 D TKL >5k >5k

ACVR1 B (ALK4) TKL >5k >5k

Example 4: Determination of growth inhibitory activity against FLT3 expressing cells by cellular proliferation assay.

The viability of Flt3 expressing cells can be evaluated using a tetrazolium salt reduction cell-based assay. In viable cells, notably among them various immortalized lymphoid and myelioid lines, this colorimetric assay can measure mitochondrial reduction of a tetrazolium component (MTS) into an insoluble formazan product. BaF3, both wild type and mutant, as well as transfected strains, MV4-1 1 and MOLM 14 are well-characterized Flt3 -dependent humanized or human cell lines which express active Flt3 receptors (see, Yee et al. Blood (2002)100 (8):2941-2949; Levis et al. (2002) 99(11): 3885-3891). These cell lines were used to determine the ability of the compounds provided herein to inhibit Flt3 in intact cells. In each case, and by methods known to those practiced in the art of cell culture, proliferation was measured after 72 hour incubation with the compounds provided herein using a standard MTS protocol (Promega Cat #5430 "Cell Titer 96 Aqueous Non- 1 radioactive Cell Proliferation Assay").

In general, cells cells were plated at 10,000-20,000 cells per well in DMEM medium with 0.5% serum. The compound plate was set up by aliquoting into column 1 of a 96 well 30OuI polypropylene plate, the negative control (DMSO), aliquoting into column 12 the positive control and titrating the test compound in serial dilutions into columns 2-1 1. An aliquot from each well of the compound plate was transferred to the plated cells and then incubated @ 370C in 5% CO2. [00538] MTS tetrazolium compound (Owen's reagent) was thawed in a H2O bath. 20 μl of MTS tetrazolium was added to each well of optical plate and the cells were incubated @ 37°C in 5%

CO2 for 2 hours. The absorbance measured at 490 nm using a microplate reader. Cell proliferation values are measured in terms of concentration of test compound that achieves 50% inhibition of cellular proliferation compared to control (IC50) and are reported in Table 4.

These results show that the compounds of this invention inhibit the growth of Flt3 expressing cells in vivo. Once again note is drawn to the efficacy of (IV), a representative indazole class molecule, and its favorable comparison to the phenolic substituted imidazoacridinone, Symadex.

TABLE 4: Growth inhibitory activity (in vivo EC50 nM) on Flt3 expressing cells

DRUG BaF3- BaF3 BaF3- BaF3- MV-4-11 MOLIV

FLT3-WT -ITD D835Y D835N

+IL-3

(V) 246 90 172 — — 333

(IV) 6 20 13 12 12 —

(III) 752 223 887 316 184 —

(Vl) 1391 74 171 — — 84

(VII) 356 36 158 — — 60

(VIII) 341 30 129 — — 627

(IX) >2000 80 723 — — 82

(X) 492 35 79 — — 90

Symadex 13.2 11.2 8.9 14.3 13.1 —

While this invention has been particularly shown and described with references to preferred embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the invention encompassed by the appended claims.