Title of Invention

"A COMPOUND OF FORMULA (I)"

Abstract A compound of Formula (I) or a pharmaceutically acceptable acid salt or basic salt thereof, for treating hyper-proliferative and angiogenesis disorders, as a sole agent or in combination with cytotoxic therapies.
Full Text molecThe present invention relates to a compound of formula (I)
Related Application?
This application claims priority to serial no. 60/450,323, filed February 28, 2003, serial no. 60/450,324 filed February 28, 2003 and serial no. 60/450,348 filed February 28, 2003
Field of the Invention
This invention relates to novel compounds, pharmaceutical compositions containing such compounds and the use of these compounds or compositions for treating hyper-proliferative and angiogenesis disorders, as a sole agent or in combination with other active ingredients, e.g., cytotoxic therapies.
Background of the Invention
Activation of the ras signal transduction pathway indicates a cascade of events that have a profound impact on cellular proliferation, differentiation, and transformation. Raf kinase, a downstream effector of ras, is recognized as a key mediator of these signals from cell surface receptors to the cell nucleus (Lowy, D. R.; Willumsen, B. M. Ann. Rev. Biochem. 1993, 62, 851; Bos, J. L. Cancer Res. 1989, 49, 4682). It has been shown that inhibiting the effect of active ras by inhibiting the raf kinase signaling pathway by administration of deactivating antibodies to raf kinase or by co-expression of dominant negative raf kinase or dominant negative i\4EK, the substrate of raf kinase, leads to the reversion of transformed cells to the normal growth phenotype (see: Daum et al. Trends Biochem. Sci. 1994, 19, 474-80; Fridman et al. J. Biol. Chem. 1994, 269, 30105-8. Kolch et al. [Nature 1991, 349, 426-28) have further indicated that inhibition of raf expression by antisense RNA blocks cell proliferation in membrane-associated oncogenes, Similarly, inhibition of raf kinase (by antisense oligodeoxynucleotides) has been correlated in vitro and in vivo with inhibition of the growth of a variety of human tumor types (Monia et al., Nat. Med. 1996, 2, 668-75). Some examples of small
ule inhibitors of Raf kinase activity are important agents for the treatment of cancer. (Naumann, U.; Eisenmann-Tappe, [.; Rapp, U. R. Recent Results Cancer Res. 1997, 143, 237; Monia, B. P.; Johnston, J. F,; Geiger, T.; Muiier, M.; Fabbro, D. Nature Medicine 1996, 2,668).
To support progressive tumor growth beyond the size of 1-2 mm3, it is recognized that tumor cells require a functional stroma, a support structure consisting of fibroblast, smooth muscle cells, endotheiial cells, extracellular matrix proteins, and soluble factors (Folkman, J., Semin Oncol, 2002. 29(6 Suppl 16), 15-8). Tumors induce the formation of stromal tissues through the secretion of soluble growth factors such as PDGF and transforming growth factor-beta (TGF-beta), which in turn stimulate the secretion of complimentary factors by host cells such as fibrobfast growth factor (FGF), epidermal growth factor (EGF), and vascular endotheliaf growth factor (VEGF). These stimulatory factors induce the formation of new blood vessels, or angiogenesis, which brings oxygen and nutrients to the tumor and allows it to grow and provides a route for metastasis. It is believed some therapies directed at inhibiting stroma formation will inhibit the growth of epithelial tumors from a wide variety of histological types. (George, D. Semin Oncoi> 2001. 26(5 Supp! 17), 27-33; Shaheen, R.M., et at., Cancer Res, 2001. 61(4), 1464-8; Shaheen, R.M., et al. Cancer Res, 1999. 59(21), 5412-6). However, because of the complex nature and the multiple growth factors involved in angiogenesis process and tumor progression, an agent targeting a single pathway may have limited efficacy. It is desirable to provide treatment against a number of key signaling pathways utilized by tumors to induce angiogenesis in the host stroma. These include PDGF, a potent stimulator of stroma formation (Ostman, A. and C.H. Heldin, Adv Cancer Res, 2001, 80, 1-38), FGF, a chemo-attractant and mitogen for fibrobiasts and endothelia! ceils, and VEGF, a potent regulator of vascularization.
PDGF is another key regulator of stromal formation which is secreted by many tumors in a paracrine fashion and is believed to promote the growth of fibrobiasts, smooth muscle and endothelia! cells, promoting stroma formation and angiogenesis, PDGF was originally identified as the v-sis oncogens product of the simian sarcoma
virus (Heldin, C.H., et al., J Cell Sci Suppl, 1985, 3, 65-76). The growth factor is made up of two peptide chains, referred to as A or B chains which share 60% homology in their primary amino acid sequence. The chains are disulfide cross linked to form the 30 kDa mature protein composed of either AA, 3B or AB homo- or heterodimmers, PDGF is found at high levels in platelets, and is expressed by endotheliai cells and vascular smooth muscle cells. In addition, the production of PDGF is up regulated under low oxygen conditions such as those found in poorly vascularized tumor tissue {Kourembanas, S., et al., Kidney Int, 1997, 51(2), 438-43). PDGF binds with high affinity to the PDGF receptor, a 1106'amino acid 124 kDa transmembrane tyrosine kinase receptor (Heidin, C.H., A. Ostman, and L, Ronnstrand, Biochrm Biophys Acta, 1998. 1378(1), 79-113). PDGFR is found as homo- or heterodimer chains which have 30% homology overall in their amino acid sequence and 64% homology between -their kinase domains (Heldin, C.H., et al.. Embo J, 1988, 7(5), 1387-93). PDGFR is a member of a family of tyrosine kinase receptors with split kinase domains that includes VEGFR2 (KDR). VEGFR3 fFI!4). c-Kit, and FLT3. The PDGF receptor is expressed primarily on fibroblast, smooth muscfe cells, and pericytes and to a lesser extent on neurons, kidney mesangial, Leydig, and Schwann cells of the central nervous system. Upon binding to the receptor, PDGF induces receptor dimerization and undergoes auto- and trans-phosphorylation of tyrosine residues which increase the receptors' kinase activity and promotes the recruitment of downstream effectors through the activation of SH2 protein binding domains. A number of signaling molecules form complexes with activated PDGFR including PI-3-kinase, phospholipase C-gamma, src and GAP (GTPase activating protein for p21-ras) (Soskic, V., et al. Biochemistry, 1999, 38(6), 1757-64). Through the activation of PI-3-kinase, PDGF activates the Rho signaling pathway inducing cell motility and migration, and through the activation of GAP, induces mitogenesis through the activation of p21-ras and the MAPK signaling pathway.
In adults, it is believed the major function of PDGF is to faciiitate and increase the rate of wound healing and to maintain blood vessel homeostasis (Baker, E.A. and D. J. Leaper, Wound Repair Regen, 2000. 8(5), 392-8; Yu, J., A. Moon, and H.R, Kim,
Biochem Biophys Res Commun, 2001. 282(3), 697-700). PDGF is found at high concentrations in platelets and is a potent chemoattractant for fibroblast, smooth muscle cells, neutrophils and macrophages. In addition to its role in wound healing PDGF is known to help maintain vascular homeostasis. During the development of new blood vessels, PDGF recruits pericytes and smooth muscle cells that are needed for the structural integrity of the vessels. PDGF is thought to play a similar role during tumor neovascularization. As part of its role in angiogenesis PDGF controls interstitial fluid pressure, regulating the permeability of vessels through its regulation of the interaction between connective tissue cells-and the extracellular matrix. Inhibiting PDGFR activity can lower interstitial pressure and facilitate the influx of cytotoxics into tumors improving the anti-tumor efficacy of these agents (Pietras, K., et al. Cancer Res, 2002. 62(19), 5476-84; Pietras, K., et al. Cancer Res, 2001. 61(7), 2929-34).
PDGF can promote tumor growth through either the paracrine or autocrine stimulation of PDGFR receptors on stromal cells or tumor cells directly, or through the amplification of the receptor or activation of the receptor by recombination. Over expressed PDGF can transform human melanoma cells and keratinocytes (Forsberg, K., et al. Proc Natl Acad Sci U S A., 1993. 90(2), 393-7; Skobe, M. and N.E. Fusenig, Proc Natl Acad Sci USA, 1998. 95(3), 1050-5), two cell types that do not express PDGF receptors, presumably by the direct effect of PDGF on stroma formation and induction of angiogenesis. This paracrine stimulation of tumor stroma is also observed in carcinomas of the colon, lung, breast, and prostate (Bhardwaj, B., et al. din Cancer Res, 1996, 2(4), 773-82; Nakanishi, K., et al. Mod Pathol, 1997, 10(4), 341-7; Sundberg, C., et al. Am J Pathol, 1997, 151(2), 479-92; Lindmark, G., et al. Lab Invest, 1993, 69(6), 682-9; Vignaud, J.M., et al, Cancer Res, 1994, 54(20), 5455-63) where the tumors express PDGF, but not the receptor. The autocrine stimulation of tumor cell growth, where a large faction of tumors analyzed express both the ligand PDGF and the receptor, has been reported in glioblastomas (Fleming, T.P.;' et al. Cancer Res, 1992, 52(16), 4550-3), soft tissue sarcomas (Wang, J., M.D. Coltrera, and A.M. Gown, Cancer Res, 1994, 54(2), 560-4) and cancers of the ovary (Henriksen, R., et al. Cancer Res,
1993, 53(19), 4550-4), prostate (Fudge, K., C.Y. Wang, and M.E. Stearns, Mod Pathol,
1994, 7(5), 549-54), pancreas (Funa, K., et al. Cancer Res, 1990, 50(3), 748-53) and
lung (Antoniades, H.N., et al., Proc NailAcad Sci USA, 1992, 89(9), 3942-6). Ligand
independent activation of the receptor is found to a lesser extent but has been reported
in chronic myelomonocytic leukemia (CMML) where the a chromosomal translocation
event forms a fusion protein between the Ets-like transcription factor TEL and the PDGF
receptor. In addition, activating mutations in PDGFR have been found in gastrointestinal
stromal tumors in which c-Kit activation is not involved (Heinrich, M.C., et al., Science,
2003, 9, 9).
Certain PDGFR inhibitors will interfere with tumor stromal development and are believed to inhibit tumor growth and metastasis.
Another major regulator of angiogenesis and vasculogenesis in both embryonic development and some angiogenic-dependent diseases is vascular endothelial growth factor (VEGF; also called vascular permeability factor, VPF). VEGF represents a family of isoforms of mitogens existing in homodimeric forms due to alternative RNA splicing. The VEGF isoforms are reported to be highly specific for vascular endothelial cells (for reviews, see: Farrara et al. Endocr. Rev. 1992, 13, 18; Neufield et al. FASEB J. 1999, 13, 9).
VEGF expression is reported to be induced by hypoxia (Shweiki et al. Nature 1992, 359, 843), as well as by a variety of cytokines and growth factors, such as interleukin-1, interleukin-6, epidermal growth factor and transforming growth factor. To date, VEGF and the VEGF family members have been reported to bind to one or more of three transmembrane receptor tyrosine kinases (Mustonen et al. J. Cell Biol., 1995, 129, 895), VEGF receptor-1 (also known as flt-1 (fms-like tyrosine kinase-1)), VEGFR-2 (also known as kinase insert domain containing receptor (KDR); the murine analogue of KDR is known as fetal liver kinase-1 (flk-1)), and VEGFR-3 (also known as flt-4). KDR and flt-1 have been shown to have different signal transduction properties (Waltenberger et al. J. Biol. Chem. 1994, 269, 26988); Park et al. Oncogene 1995, 10,
135). Thus, KDR undergoes strong ligand-dependant tyrosine phosphorylation in intact cells, whereas flt-1 displays a weak response. Thus, binding to KDR is believed to be a critical requirement for induction of the full spectrum of VEGF-mediated biological responses.
In vivo, VEGF plays a central role in vasculogenesis, and induces angiogenesis and permeabilization of blood vessels. Deregulated VEGF expression contributes to the development of a number of diseases that are characterized by abnormal angiogenesis and/or hyperpermeability processes. It is believed regulation of the VEGF-mediated signal transduction cascade by some agents can provide a useful mode for control of abnormal angiogenesis and/or hyperpermeability processes.
Angiogenesis is regarded as an important prerequisite for growth of tumors beyond about 1-2 mm. Oxygen and nutrients may be supplied to cells in tumor smaller than this limit through diffusion. However, it is believed every tumor is dependent on angiogenesis for continued growth after it has reached a certain size. Tumorigenic cells within hypoxic regions of tumors respond by stimulation of VEGF production, which triggers activation of quiescent endothelial cells to stimulate new blood vessel formation. (Shweiki et al. Proc. Nat'l. Acad. Sci., 1995, 92, 768). In addition, VEGF production in tumor regions where there is no angiogenesis may proceed through the ras signal transduction pathway (Grugel et al. J. Biol. Chem., 1995, 270, 25915; Rak et al. Cancer Res, 1995, 55, 4575). In situ hybridization studies have demonstrated VEGF mRNA is strongly upregulated in a wide variety of human tumors, including lung (Mattern et al. Br. J. Cancer 1996, 73, 931), thyroid (Viglietto et al. Oncogens 1995, 11, 1569), breast (Brown et al. Human Pathol. 1995, 26, 86), gastrointestional tract (Brown et al. Cancer Res. 1993, 53, 4727; Suzuki et al. Cancer Res. 1996, 56, 3004), kidney and bladder (Brown et al. Am. J. Pathol. 1993, 1431, 1255), ovary (Olson et al. Cancer Res. 1994, 54, 1255), and cervical (Guidi et al. J. Nat'l Cancer Inst. 1995, 87, 12137) carcinomas, as well as angiosacroma (Hashimoto et al. Lab. Invest. 1995, 73, 859) and several intracranial tumors (Plate et al. A/aft/re 1992, 359, 845; Phillips et al. Int. J. Oncol. 1993,
2, 913; Berkman et al. J. Clin. invest, 1993, 9f, 153). Neutralizing monoclonal antibodies to KDR have been shown to be efficacious in blocking tumor angiogenesis (Kim et al. Nature 1993, 362, 841; Rockwell et al. Mo/. Cell. Differ. 1995, 3, 315).
Overexpression of VEGF, for example under conditions of extreme hypoxia, can lead to intraocular angiogenesis, resulting in hyperproliferation of blood vessels, leading eventually to blindness. Such a cascade of events has been observed for a number of retinopathies, including diabetic retinopathy, ischemic retinal-vein occlusion, and retinopathy of prematurity (Aiello-et al. New Engl. J. Med. 1994, 331, 1480; Peer et al. Lab. Invest 1995, 72, 638), and age-related macular degeneration (AMD; see, Lopez et al. Invest. Opththalmol. Vis. Sci. 1996, 37, 855).
In rheumatoid arthritis (RA), the in-growth of vascular pannus may be mediated by production of angiogenic factors. Levels of immunoreactive VEGF are hig'h in the synovial fluid of RA patients, while VEGF levels were low in the synovial fluid of patients with other forms of arthritis of with degenerative joint disease (Koch et al. J. Immunol. 1994, 152, 4149). The angiogenesis inhibitor AGM-170 has been shown to prevent neovascularization of the joint in the rat collagen arthritis model (Peacock et al. J. Exper. Med. 1992, 775, 1135).
Increased VEGF expression has also been shown in psoriatic skin, as well as bullous disorders associated with subepidermal blister formation, such as bullous pemphigoid, erythema multiforme, and dermatitis herpetiformis (Brown et al. J. Invest. Dermatol. 1995, 104,744).
The vascular endothelial growth factors (VEGF, VEGF-C, VEGF-D) and their receptors (VEGFR2, VEGFR3) are not only key regulators of tumor angiogenesis, but also lymphangiogenesis. VEGF, VEGF-C and VEGF-D are expressed in most tumors, primarily during periods of tumor growth and, often at substantially increased levels. VEGF expression is stimulated by hypoxia, cytokines, oncogenes such as ras, or by

inactivation of tumor suppressor genes (McMahon, G. Oncologist 2000, 5(Suppl. 1), 3-10; McDonald, N.Q.; Hendrickson, W.A. Ce//1993, 73, 421-424).
The biological activities of the VEGFs are mediated through binding to their receptors. VEGFR3 (also called Flt-4) is predominantly expressed on lymphatic endothelium in normal adult tissues. VEGFR3 function is needed for new lymphatic vessel formation, but not for maintenance of the pre-existing lymphatics. VEGFR3 is also upregulated on blood vessel endothelium in tumors. Recently VEGF-C and VEGF-D, ligands for VEGFR3, have been identified as regulators of lymphangiogenesis in mammals. Lymphangiogenesis induced by tumor-associated lymphangiogenic factors could promote the growth of new vessels into the tumor, providing tumor cells access to systemic circulation. Cells that invade the lymphatics could find their way into the bloodstream via the thoracic duct. Tumor expression studies have allowed a direct comparison of VEGF-C, VEGF-D and VEGFR3 expression with clinicopathoiogical factors that relate directly to the ability of primary tumors to spread (e.g., lymph node involvement, lymphatic invasion, secondary metastases, and disease-free survival). In many instances, these studies demonstrate a statistical correlation between the expression of lymphangiogenic factors and the ability of a primary solid tumor to metastasize (Skobe, M. et al. Nature Med. 2001, 7(2), 192-198; Stacker, S.A. et aL Nature Med. 2001, 7(2), 186-191; Makinen, T. et al. Nature Med. 2001, 7(2), 199-205; Mandriota, S.J. et al. EMBO J. 2001, 20(4), 672-82; Karpanen, T. et al. Cancer Res. 2001, 61(5), 1786-90; Kubo, H. et al. Blood 2000, 96(2), 546-53).
Hypoxia appears to be an important stimulus for VEGF production in malignant cells. Activation of p38 MAP kinase is required for VEGF induction by tumor cells in response to hypoxia (Blaschke, F. et al. Biochem. Biophys. Res. Commun. 2002, 296, 890-896; Shemirani, B. et al. Oral Oncology 2002, 38, 251-257). In addition to its involvement in angiogenesis through regulation of VEGF secretion, p38 MAP kinase promotes malignant cell invasion, and migration of different tumor types through regulation of collagenase activity and urokinase plasminogen activator expression
(Laferriere, J. et al. J. Biol. Chem. 2001, 276, 33762-33772; Westermarck, J. et al. Cancer Res. 2000, 60, 7156-7162; Huang, S. et al. J. Biol. Chem. 2000, 275, 12266-12272; Simon, C. et al. Exp. Cell Res. 2001, 277, 344-355).
Some diarylureas have been described as having activity as serine-threonine kinase and/or as tyrosine kinase inhibitors. The ultility of these diarylureas as an active ingredient in pharmaceutical compositions for the treatment of cancer, angiogenesis disorders, and inflammatory disorders has been demonstated. See Redman et al., Bioorg. Med. Chem. Lett 2001, -11, 9-12; Smith et al,, Bioorg. Mod. Chem. Lett. 2001, 11, 2775-2778; Dumas et al., Bioorg. Med. Chem. Lett. 2000, 10, 2047-2050; Dumas et al., Bioorg. Med. Chem. Lett. 2000, 10, 2051-2054; Ranges et al., Book of Abstracts, 220th ACS National Meeting, Washington, DC, USA, MEDl 149; Dumas et al., Bioorg. Med. Chem. Lett. 2002, 12, 1559-1562; Lowinger et al., Clin. Cancer Res. 2000, 6(suppl.), 335; Lyons et al., Endocr.-Relat. Cancer 2001, 8, 219-225, Riedl et al., Book of Abstracts, 92nd AACR Meeting, New Orleans, LA, USA, abstract 4956; Khire et al., Book of Abstracts, 93rdAACR Meeting, San Francisco, CA, USA, abstract 4211; Lowinger et a!., Curr. Pharm. Design 2002, 8, 99-110; Regan et al., J. Med. Chem. 2002, 45, 2994-3008; Pargellis et al., Nature Struct Biol. 2002, 9(4), 268-272; Carter et al., Book of Abstracts, 92nd AACR Meeting, New Orleans, LA, USA, abstract 4954; Vincent et al., Book of Abstracts, 38th ASCO Meeting, Orlando, PL, USA, abstract 1900; Hilger et al., Book of Abstracts, 38th ASCO Meeting, Orlando, PL, USA, abstract 1916; Moore et al., Book of Abstracts, 38th ASCO Meeting, Orlando, FL, USA, abstract 1816; Strurriberg et al., Book of Abstracts, 38th ASCO Meeting, Orlando, FL, USA, abstract 121; Madwed JB: Book of Abstracts, Protein Kinases: Novel Target Identification and Validation for Therapeutic Development, San Diego, CA, USA, March 2002; Roberts et al., Book of Abstracts, 38th ASCO Meeting, Orlando, FL, USA, abstract 473; Tolcher et al., Book of Abstracts, 38th ASCO Meeting, Orlando, FL, USA, abstract 334; and Karp et al., Book of Abstracts, 38th AACR Meeting, San Francisco, CA, USA, abstract 2753.
Despite the advancements in the art, there remains a need for additional treatments.
Summary of the Invention
The present invention pertains to:
(i) novel compounds, salts, metabolites and prodrugs thereof, including diastereoisomeric forms,
(ii) pharmaceutical compositions containing any of such compounds salts, metabolites and prodrugs thereof, including diastereoisomeric forms, and
(iii) use of those compounds or compositions for treating diseases, e.g., hyper-proliferative and angiogenesis disorders, as a sole agent or in combination with other active ingredients, e.g., cytotoxic therapies.
The compounds of formula (I), salts, metabolites and prodrugs thereof, including diastereoisomeric forms (both isolated stereoisomers and mixtures of stereoisomers) are collectively referred to herein as the "compounds of the invention". Formula (I) is as follows:
(Formula Removed)
wherein
A is optionally substituted (unsubstituted and substituted) pyridinyl,
naphthyl,
8-10 membered bicyclic heteroaryl groups having 1-4 heteroatoms which are O, N, S or combinations thereof,
partially saturated C8-C10 bicyclic carbocyclic moieties, bound to the urea moiety through a benzene group, or
partially saturated 8 to 10 membered bicyclic heterocyclic moieties, said heterocyclic moieties having 1-4 heteroatoms which are O, N or S or combinations thereof.
Where A of Formula (I) is a partially saturated 8 to 10 membered bicyclic heterocyclic moiety with at least one oxygen atom, it is preferably substituted, more preferably halosubstituted. The halogen substituents are preferably located on the saturated carbons atoms of the partially saturated 8 to 10 membered bicyclic heterocyclic moieties. These saturated carbons atoms are more preferably per-halosubstituted, most preferably fluoro-substituted.
Structures of optionally substituted pyridinyl moieties for A of formula (I) which are of particular interest include structures of formula 1x:

(Formula Removed)
Structures of optionally substituted naphthyl moieties for A of formula (I) which are of particular interest include structures of formula 1y:
(Formula Removed)

The structure 1y represents that the substituents R3 can appear on any carbon atom in either ring which has a valence that is otherwise complete with a hydrogen atom as a substituent. The bond to the urea group can also be through any carbon atom on
either ring which has a valence that is otherwise complete with a hydrogen atom as a substituent.
Examples of suitable optionally substituted 8-10 membered bicyctic heteroary! groups for A of formula (I) include:
2-, 3-, 4-, 5-, 6-, 7- or 8-quinolinyl, 1-, 3-, 4-, 5-, 6-, 7-, 8- isoquinolinyl, benzimidazol-5-yl, benzimidazol~6-yl,
1,3-benzothiazoi-2-yl, 1,3-benzothiazol-5-yl, 1,3-benzothiazol-6-yl, 1,2,3-ben2otriazol-5-yl, 1,3-benzoxazoi-2-yl, 1,3-benzoxazol-6-yl, quinoxa!in-2-yl, quinoxalin-6-y!, 1 H-indazol-5-yl, 2H-indazol-5-yl, 1H-indazol-6-yl and 1 H-indoi-5-yt.
Structures of optionally substituted 8-10 membered bicyclic heteroaryl groups for A of formula (I) which are of particular interest include structures of formulae1a,1b and
(Formula Removed)
The structure 1a represents that the substituents R3 can appear on any carbon in either ring which has a valence that is otherwise complete with a hydrogen atom as a substituent. The bond to the urea group can also be through any carbon atom on either ring which has a valence that is otherwise complete with a hydrogen atom as a substituent
The structures 1b and 1c represent that the substituents R3 can appear on any atom in the five membered ring which has a valence that is otherwise complete with a hydrogen atom as a substituent. The bond to the urea group can be through any carbon on the six membered ring which has a valence that is otherwise complete with a hydrogen substituent.
Examples of partially saturated C8-C10 bicyclic carbocyclic moieties, bound to the urea moiety through a benzene group include 2,3-dihydro-1 H-inden-4-yl and 2,3-dihydro-1 H-inden-5-yt.
Structures of partially saturated C8-C10 bicyclic carbocyclic moieties for A of formula (I) which are of particular interest include structures 1d and 1e:
(Formula Removed)
The structures 1d and 1e represent that the substituents R3 can appear on any carbon atom in the unsaturated ring which has a valence that is otherwise complete with a hydrogen atom as a substituent. The bond to the urea group can be through any carbon atom on the unsaturated six membered ring which has a valence that is otherwise complete with a hydrogen atom as a substituent.
Examples of partially saturated 8 to 10 membered bicyclic heterocyclic moieties having 1-4 heteroatoms which are N, S or combinations there of include 2,3-dihydro-1 H-indol-5-yl and 2,3-dihydro-1H-indol-6-yl.
Examples of partially saturated 8 to 10 membered- bicyclic heterocyclic moieties having 1-4 heteroatoms which are O, N, S or combinations thereof with at least one oxygen atom include: 2H,3H-benzo[e]1,4-dioxan-6-yl, 1,1-dioxido-2,3-dihydro-1-benzothien-6-yl, 1-oxo-2,3-dihydro-1H-inden-5-yl, 2H-benzo[d]1,3-dioxolen-5-yl, 2H-benzo[d]1,3-dioxolen-4-yl, 2,3-dihydrobenzo[b]fur-5-yl, 2H,4H-benzo[e]1,3-dioxan-6-yl, or 2H,4H-benzo[e]1,3-dioxan-8-yl.
These moieties can be halosubstituted, preferably at the saturated carbon atoms, up to per-halosubstitution.
Structures of partially saturated 8 to 10 membered bicyclic heterocyclic moieties for A of formula (I) which are of particular interest include structures 1f, 1g, 1h and 11:

(Formula Removed)


The structures 1f, 1d, 1h and 1i represent that the substituents R3 can appear on any carbon atom in the unsaturated ring which has a valence that is otherwise complete with a hydrogen atom as a substituent. The bond to the urea group can be through any carbon atom on the unsaturated six membered ring which has a valence that is otherwise complete with a hydrogen atom as a substituent.
B is optionally substituted phenyl or naphthyl. Structures of optionally substituted phenyl or naphthyl moieties for B of formula (I) which are of particular interest include structures- 2a and 2b:
(Formula Removed)
The structures 2a and 2b represent that the substituents R1 can appear on any carbon atom in the structure which has a valence that is otherwise complete with a hydrogen atom as a substituent and the bond to the urea group can be through any
carbon atom in the structure which has a valence that is otherwise complete with a hydrogen atom as a substituent.
In a class of embodiments of this invention, B is substituted by at least one halogen substituent.
L is a bridging group which is -S- or -O-.
The variable p is 0,1, 2, 3, or 4, typically 0 or 1.
The variable n is 0,1, 2, 3, 4, 5 or 6, typically 0,1,2,3 or 4.
The variable m is 0,1,2 or 3, typically 0.
Each R1 is independently:
halogen,
C1-5 haloalkyl
N02, '
C(0)NR4R5,
C1-6 alkyl,
C1-6 dialkylamine,
C1-3 alkylamine,
CN,
amino,
hydroxy or
C1-3 alkoxy.
Where present, R1 is more commonly halogen and of the halogens, typically chlorine or fluorine, and more commonly fluorine.
Each. R2 is independently :
C1-5 alkyl, C1-5 haloalkyl, C 1-3 alkoxy, N-oxo or N-hydroxy.
Where present, R2 is typically methyl or trifluoromethyl. Each R3 is independently selected from halogen ,
R'4
OR4, S(0)R4, C(0)R4 C(0)NR4R5,
0X0,
cyano or nitro (N02).
Preferably, at least one R3 is halogen. In certain embodiments, each R3 is halogen. R4 and R5 are independently selected from
hydrogen,
C 1-6 alkyl, and
up to per-halogenated C 1-6 alkyl.
Other examples of A include:

(Formula Removed)



Other examples of B include
(Formula Removed)
Preferably the urea group -NH-C(O)-NH- and the bridging group, L, are not bound to contiguous ring carbons .of B, but rather have 1 or 2 ring carbons separating them.
Examples of R1 groups include fluorine, chorine, bromine, methyl, NO2, C(O)NH2, methoxy, SCH3, trtfluoromethyl, and methanesulfonyl.
Examples of R2 groups include methyl, ethyl, propyl, oxygen, and cyano.
Examples of R3 groups include trifluoromethyl, methyl, ethyl, propyl, butyl, isopropyl, tert-butyl, chlorine, fluorine, bromine, cyano, methoxy, acetyl, trifluoromethanesulfonyl, trifluoromethoxy, and trifluoromethylthio.
Compounds of formula (I) of interest are:
{[2-chloro-4-(2-cyano(4-pyridyloxy))phenyl3amino}-N-indan-5-ylcarboxamide;
{[4-(2-cyano(4-pyridyloxy))-2-fluorophenyl]amino}-N-indan-5-ylcarboxamide;
{[2-:chloro-4-(2-cyano(4-pyridyloxy))phenyI]amino}-N-(1-oxoindan-5-yl)carboxamide;
{[4-(2-cyano(4-pyridyloxy))-2-fluorophenyl]amino}-N-(2-naphthyl)carboxamide;
N-(2,2-diTluorobenzo[d]1,3-dioxolan-5-yl){[4-(2-cyano(4-
pyridyloxy))phenyl]amino}carboxamide;
N-(2J2-difluorobenzo[d]1,3-dioxolan-5-yl){[4-(2-cyano(4-
pyridyloxy))phenyl]amino}carboxamide;
N-(2,2-difluorobenzo[d]1,3-dioxolan-5-yl){[2-chloro-4-(2-cyano(4-
pyridyloxy))phenyl]amino}carboxamide;
N-(2,2-difluorobenzo[d]1,3-dioxolan-5-yl){[3-chloro-4-(2-cyano(4-
pyridyloxy))phenyl]amino}carboxamide;
N-(2,2-difluorobenzo[d]1,3-dioxolan-5-yl){[3-chloro-4-(2-cyano(4-
pyridyloxy))phenyl]amino}carboxamide;
N-(2,2-difluorobenzo[d]1,3-dioxo[an-5-yl){[4-(2-cyano(4-pyridyloxy))-3-
fluorophenyl]amino}carboxamide;
{[4-(2-cyano(4-pyridyloxy))-2-(trifluoromethyl)phenyl]amtno}-N-(2,2,3)3-
tetrafluorobenzo[e]1,4-dioxan-6-yl)carboxamide;
{[2-chloro-4-(2-cyano(4-pyridyloxy))phenyl]amino}-N-(2,2,3,3-tetrafluorobenzo[e]1,4-
dioxan-6-yl)carboxamide;
{[4-(2-cyano(4-pyridyIoxy))-2-fluorophenyl]amino}-N-(2,2,3,3-tetrafluorobenzo[e]1,4-dioxan-6-yl)carboxamide;
{[4-(2-cyano(4-pyridyloxy))-2)6-difluorophenyl]amino}-N-(2,2,3,3-tetrafluorobenzo[e]1,4-dioxan-6-yl)carboxamide;
{[4-(2-cyano(4-pyridyloxy))-2,5-difluorophenyl]amino}-N-(2I2,3,3-tetrafluorobenzo[e]1I4-
dioxan-6-yl)carboxamide;
{[3-chloro-4-(2-cyano(4-pyridyloxy))phenyl]amino}-N-(2,2,3,3-tetraf]uorobenzo[e]1,4-
dioxan-6-yl)carboxamide;
{[4-(2-cyano(4-pyridyloxy}}-2-methylpheny!]amino}-N-(2,2,3,3-tetrafluorobenzo[e]1,4-
dioxan-6-yl)carboxamide;
{[4-(2-cyano(4-pyridyloxy))-3-methyiphenyl]amino}-N-(2,2,3,3-tetrafluorobenzo[e]1,4-
dioxan-6-yl)carboxamide;
{[4-(2-cyano(4-pyridy!oxy))-2-nitrophenyl]amino}-N-(2,2,3,3-tetraffuorobenzo[e]1,4-
dioxan-6-yl)carboxamide;
{[4-(2-cyano(4-pyridyloxy))pheny!]amino}-N-(2,2,4,4-tetrafluorobenzo[3,4-e]1,3-dioxan-
6-yl)carboxamide;
{[4-(2-cyano(4-pyridyloxy))-2-fluorophenyl]amino}-N-(2,2,4,4-tetraf3uorobenzo[3,4-e]1,3-dioxan-6-yl)carboxamide;
{[3-chloro-4-(2-cyano(4-pyridy[oxy))phenyl3amino}-N-(2,2,4,4-tetraf[uorobenzo[3,4-e]1,3-d ioxa n-6-y l)ca rboxa m ide;
{[2-chIoro-4-(2-cyano(4-pyridyloxy})phenyl3amino}-N-(2,2,4,4-tetrafluorobenzo[3,4-e]1,3-dioxan-6-yl)carboxam(de;
{[4-(2-cyano(4-pyridy!oxy))-3-fluorophenyl]amino}-N-(2,2,4>4-tetrafluorobenzo[3,4-e31,3-dioxan-6-y!)carboxamide;
{[4-(2-cyano(4-pyridyioxy))-2-(trifluoromethyl)phenyl]amino}-N-(2,2,4,4-tetrafluorobenzofS^-ejI.S-dioxan-e-yOcarboxamide;
{[4-(2-cyano(4-pyridyloxy))-2,3-difluorophenyi]amino}-N-(2,2,4(4-tetrafluorobenzo[;3,4-e]1,3-dioxan-6-yl)carboxamide;
{[4-(2-cyano(4-pyridyloxy))-2,5-dif!uorophenyl]amino}-N-(2,214,4-tetrafluorobenzo[3,4-e]1,3-dioxan-6-yl)carboxamide;
{[4-(2-cyano(4-pyridyloxy))-2,6-difluorophenyl]amino}-N-(2,2,4,4-tetrafIuorobenzo[3,4-e]1,3-dioxan-6-y!)carboxamide;
{[4-(2-cyano(4-pyridyl}oxy)-3-methoxyphenyl]amino>-N-(2,2,4,4-tetrafluorobenzo[3,4-e}1,3-dioxan-6-yl)carboxamide;
{[3-bromo-4-(2-cyano(4-pyridyloxy))phenyl]amino}-N-(2,2,4,4-tetrafluorobenzo[3,4-e]1,3-
dioxan-6-yl)carboxamide;
{[4-(2-cyano(4-pyridyioxy})-2-methylphenyl]amino}-N-(2,21414-tetrafluorobenzo[3,4-
e]1,3-dioxan-6-yl)carboxamide;
{[4-(2-cyano(4-pyridy[oxy))-3-methylpheny]]amino}-N-(2,2,4,4-tetrafluorobenzo[3,4:
e]1,3-dioxan-6-yl)carboxamide;
5-(2-cyano(4-pyridyl)oxy)-2-{[N-(2,2,4,4-tetrafluorobenzo[3,4-e]1,3-dioxan-6-
yf)carbamoyl]amino}benzamide
{[4-(2-cyano(4-pyridyloxy))-2-nitrophenyl]amino}-N-(2,2,4,4-tetrafluorobenzo[3,4-e]1,3-
dioxan-6-yl)carboxamide;
{[4-(2-cyano-1-hydroxy(4-pyridyloxy))phenyl]amino}-N-(2,2,4,4-tetrafluorobenzo[3,4-
e}1,3-dioxan-6-y()carboxarnide;
{[4-(2-cyano-1-hydroxy(4-pyridy!oxy))-2-f)uorophenyl3amino}-N-(2,2,4,4-
tetrafluorobenzo[3,4-e]1,3~dioxan~6-yl)carboxamide;
{[4-(2-cyano(4-pyridyl)oxy)-2-rnethylthiophenyl]amino}-N-(2,2,4)4-tetraf!uorobenzo[314-
e]1,3-dioxan-6-yl)carboxamide;
{[4-{2-cyanD(4-pyridyI)oxy)-2-(methylsulfony!)phenyl]amino}-N-(2,2,4I4-
tetrafluorobenzo[3,4-el1,3-dioxan-6-yl)carboxamide;
{[4-(2-cyano(4-pyridyloxy))-2-fluorophenyl]amino}-N-[2-(trifluoromethyl)(4-
pyridyl)]carboxamide;
N-[4-(tert-butyl){2-pyridyl)]{[4-(2-cyano(4-pyridyloxy))phenylJamino}carboxamide;
fM-[4-(tert-butyl)(2-pyridyl)3{[3-chloro-4-(2-cyano(4-
pyridyloxy))pheny!]amino}carboxamide;
N-f4-(fert-butyl)(2-pyn'dyl)I{[4-(2-cyano(4-pyridyloxy))-3-
fluorophenyl]amino}carboxamide;
N-[4-(tert-butyl)(2-pyridyl)]{[4-(2-cyano(4-pyridyloxy))-2-
fiuorophenyl]amino}carboxamide;
N-[4-(tert-butyl)(2-pyridyl)]{[3-bromo-4-(2-cyano(4-
pyridyloxy))phenyl]amino}carboxamide;
2-({N-[4-(terf-butyf)(2-pyridy[)]carbamoy[}am!no)-5-(2-cyano(4-pyridyl)oxy)benzamide
N-[4-(tert-butyl)(2-pyridy[)]{[4-(2-cyano(4-pyridyloxy))-3-
fluorophenyOamino}carboxamide;
{[4-(2-cyano(4-pyridyloxy))-2-(trif!uoromethyl)phenyl]amino}-N-[4-(trifluoromethyl}(2-
pyridyl)]carboxamide;
{[4-(2-cyano(4-pyridyioxy))-2,6-difluorophenyl]amino}-N-[4-(trifluoromethyI)(2-
pyridyJ)]carboxamide;
{[4-(2-cyano(4-pyndy[oxy)}phenyl]amino}-N-[4-(trifluoromethyl)(2-pyridyl)Jcarboxamide;
{[4-(2-cyano{4-pyridyloxy))phenyl]am!no}-N-(4-ethy!(2-pyridyl))carboxamide;
{[4-(2-cyano(4-pyridyloxy))phenyl]amino}-N-(2-methyl(6-quinolyl))carboxamide;
{[3-chloro-4-(2-cyano(4-pyridy!oxy))phenyl]amino}-N-(2-methyJ(6-quinolyl))carboxamide;
{[3-chloro-4-(2-cyano(4-pyridyloxy))phenyl]amino}-N-(6-quinolyl)carboxamide;
{[2-chloro-4-(2-cyano(4-pyridyloxy))phenyOamino}-N-(6-quinoly])carboxamide;
{[4-(2-cyano(4-pyridyloxy))-2-(trifluoromethy()phenyl]amino}-N-(6-quinolyl)carboxamide;
{{4-(2-cyano(4-pyridyloxy)}phenyl]amino}-N-(3-isoquinolyf)carboxamide;
{{4-(2-cyano(4-pyridyfoxy))-2-fluorophenyl]amino}-N-(3-isoquinolyl)carboxamide;
{[3-chloro-4-(2-cyano(4-pyridyloxy))phenyf]amino}-N-(3-isoquinolyl)carboxamide;
{[4-(2-cyano(4-pyridyloxy))phenyl]amino}-N-(1-methy!(1H-indazol-5-yl))carboxamide;
{[4-(2-cyano(4-pyridyloxy))-2-fluoropheny!3amino}-N-(1-methyl(1H-indazo!-5-
yl))carboxamide;
{[2-chioro-4-(2-cyano(4-pyridyloxy))phenyljamino}-N-(1-methy[(1H-indazol-5-yl))carboxamide;
{[4-(2-cyano(4-pyridyloxy))-2-(trifluoromethyl)phenyl]amino}-N-(1-methyl(1H-indazol-5-yl))carboxamide;
{[3-chloro-4-(2-cyano(4-pyridy)oxy))phenyI]amino}-N-(1-methyl(1H-Jndazol-5-yf))carboxamide;
{[4-(2-cyano(4-pyridyloxy))phenyi]amino}-N-[2-(triffuoromethyf)benzimida2oI-5-yljcarboxamfde;
{[3-chloro-4-(2-cyano(4-pyridyloxy))phenyl]amino}-N-[2-(trifluoromethyl)benizmidazol-5-
yljcarboxamide;
N-benzothiazof-5-yI{[4-(2-cyano{4-pyridyloxy)}-2-nitrophenyI]amino}carboxamide;
{[4-(2-cyano(4-pyridyloxy))-3-methyiphenyl]amino}-N-(2-methylbenzothiazol-5-
yl)carboxamide; or
salts thereof and stereoisomers thereof.
A class of compounds of interest are of formula II below
(Formula Removed)

wherein B of formula II is(Formula Removed)
wherein the urea group, -NH-C(O)-NH-, and the bridging group, L, are not bound to contiguous ring carbons of B, but rather have 1 or 2 ring carbons separating them,
and A of formula (I!) is
(Formula Removed)
wherein the variable n is 0, 1, 2, 3 or 4,
R3 is trifluoromethyl, methyl, ethyl, propyl, butyl, isopropyl, tert-butyl, chlorine, fluorine, bromine, cyano, methoxy, acetyl, trifluoromethanesulfonyl, trifluoromethoxy, or trifluoromethylthio.
In a subclass of such compounds, each R3 substituent on A of formula II is fluorine. In another subclass of such compounds, A of formula II is
(Formula Removed)and B of formula II is phenylene, fluoro substituted phenylene or difluoro substituted phenylene.Another class of compounds of interest includes compounds having the structures of formulae X and Y below wherein phenyl ring "B" and rnoiety Q each have at least one halogen substttuent, preferably Ci or F, more preferably F, In a subclass of these compounds, phenyl ring "B" and moiety Q each have 2-4 halogen substituents, preferably CI or F, more preferably F.
(Formula Removed)For the compounds of formula X, R2, m and A are as defined above for formula I. The variable "m" is preferably zero, leaving CN as the only substituent on the pyridtnyl moiety. Preferred values for A are substituted pyridinyl and partially unsaturated 8-10 member heterocyclics which have at (east one oxygen heteroatom and.at least one halogen substituent.
For the compounds of formula Y, R2 and m are as defined above for the compounds of formula I with "m" preferably being zero.
The moiety Q represents a 8 to 10 membered bicyclic heterocydic moiety having 1-4 heteroatoms which are O, N, S or combinations thereof. Preferably the bicyclic structure is partially unsaturated and has at least one oxygen hetero atom. In preferred embodiments, the unsaturated carbon atoms are per-halogenated.
Examples of such structures include :
2,2,4,4-tetrafluorobenzo[3,4-e]1,3-dioxan-6-yl, 2,2,3,3-tetrafluorobenzo[e]1,4-dioxan-6-yl and 2,2-difluorobenzo[d]1,3-dioxolan-5-yl.
A subclass of compounds of interest includes compounds having the structure of formulas Za, Zb , Zc and Zd below :
(Formula Removed)Each R1 is independently halogen or trifluoromethyl and each R3 is independently halogen, R4, OR4, S(O)R4,C(O)R4, C(0)NR4R5, oxo or cyano ornitro (NO2) and is preferably fluoro, trifluoromethyl, methyl and t-butyl.
The variable n is 0,1, 2, 3 or 4 and the variable p is 0 or 1.
When any moiety is "substituted", it can have up to the highest number of indicated substltuents, and each substituent can be located at any available position on the moiety and can be attached through any available atom on the substituent, "Any available position" means any position on the moiety that is chemically accessible
through means known in the art or taught herein and that does not create an unstable molecule, e.g., incapable of administration to a human. When there are two or more substituents on any moiety, each substituent is defined independently of any other substituent and can, accordingly, be the same or different.
The term "optionally substituted" means that the moiety so modified may be either unsubstituted, or substituted with the identified substituent(s).
It is understood that the term "hydroxy" as a pyridine substituent includes 2-, 3-, and 4-hydroxypyridine, and also includes those structures referred to in the art as 1-oxo-pyridine, 1 -hydroxy-pvridine or pyridine N-oxide.
Where the plural form of the word compounds, salts, and the like, is used herein, this is taken to mean also a single compound, salt, or the like.
The term C1-6 alkyl, unless indicated otherwise, means straight, branched chain or cyclic alkyl groups having from one to six carbon atoms, which may be cyclic, linear or branched with single or multiple branching. Such groups include for example methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, cyclopropyl, cyclobutyl and the like.
The term C1-6 haloalkyl, unless indicated otherwise, means a saturated hydrocarbon radical having up to six carbon atoms, which is substituted with a least one halogen atom, up to perhalo. The radical may be cyclic, linear or branched with single or multiple branching. The halo substituent(s) include fluoro, chloro, bromo, or iodo. Fluoro, chloro and bromo are preferred, and fluoro and chloro are more preferred. The halogen substituent(s) can be located on any available carbon. When more than one halogen substituent is present on this moiety, they may be the same or different. Examples of such halogenated alkyl substituents include but are not limited to
chloromethyl, dichloromethyl, trichloromethyl, fiuoromethyl, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, and 1,1,2,2-tetrafluoroethyl, and the like.
The term C1-6 alkoxy, unless indicated otherwise, means a cyclic, straight or branched chain alkoxy group having from one to six saturated carbon atoms which may be cyclic, linear or branched with single or multiple branching, and includes such groups as methoxy, ethoxy, n-propoxy, isopropoxy, butoxy, pentoxy and the like. It also includes halogenated groups such as 2,2-dichloroethoxy, trifluoromethoxy, and the like.
.Halo or halogen means fluoro, chloro, bromo, or iodo. Fluoro, chloro and bromo are preferred, and fluoro and chloro are more preferred.
C1-3 alkylamine, unless indicated otherwise, means methylamino, ethylamino, propylamino or isopropylamino.
Examples of C1-6 dialkylamine include but are not limited to diethylamino, ethyl-isopropylamino, methyl-isobutylamino and dihexylamino.
The term heteroaryl refers to both monocyclic and bicyclic heteroaryl rings. Monocyclic heteroaryl means an aromatic monocyclic ring having 5 to 6 ring atoms and 1-4 hetero atoms selected from N, 0 and S, the remaining atoms being carbon. When more than one hetero atom is present in the moiety, they are selected independently from the other(s) so that they may be the same or different. Monocycfic heteroaryl rings include, but are not limited to pyrrole, furan, thiophene, imidazole, pyrazole, thiazole, oxazole, isoxazole, isothiazole, triazole, tetrazole, thiadiazole, oxadiazole, pyridine, pyrimidine, pyridazine, pyrazine, and triazine.
Bicyclic heteroaryl means fused bicyclic moieties where one of the rings is chosen from the monocyclic heteroaryl rings described above and the second ring is either benzene or another monocyclic heteroaryl ring described above. When both
rings in the bicyclic moiety are heteroaryl rings, they may be the same or different, as long as they are chemicaliy accessible by means known in the art. Bicyclic heteroaryl rings include synthetically accessible 5-5, 5-6, or 6-6 fused bicyclic aromatic structures including, for example but not by way of limitation, benzoxazole (fused phenyl and oxazole), quinoline (fused phenyl and pyridine), imidazopyrimidine (fused imidazole and pyrimidine), and the like.
Where indicated, the bicyclic heteroaryl moieties may be partially saturated. When partially saturated either the monocyclic heteroaryl ring as described above is fully or partially saturated, the second ring as described above is either fully or partially saturated or both rings are partially saturated.
The term "5 or 6 membered heterocyclic ring, containing at least one atom selected from oxygen, nitrogen and sulfur, which is saturated, partially saturated, or aromatic" includes, by no way of limitation, tetrahydropyran, tetrahydrofuran, 1,3-dioxolane, 1,4-dioxane, morpholine, thiomorpholine, piperazine, piperidine, piperidinone, tetrahydropyrimidone, pentamethyiene sulfide, tetramethylene sulfide, dihydropyrane, dihydrofuran, dihydrothiophene, pyrrole, furan, thiophene, imidazole, pyrazole, thiazole, oxazole, isoxazole, isothiazole, triazole, pyridine, pyrimidine, pyridazine, pyrazine, triazine, and the like.
The term "C1-3 alkyl-phenyl" includes, for example, 2-methylphenyl, isopropylphenyl, 3-phenylpropyl, or 2-phenyl-1-methylethyl. Substituted examples include 2-[2-chlorophenyl]ethyl, 3,4-dimethylphenylmethyl, and the like.
Unless otherwise stated or indicated, the term "aryl" includes 6-12 membered mono or bicyclic aromatic hydrocarbon groups (e.g., phenyl, naphthalene, azulene, indene group ) having 0, 1,2, 3, 4, 5 or 6 substituents.
The compounds of Formula (1) may contain one or more asymmetric centers, depending upon the location and nature of the various substituents desired. Asymmetric carbon atoms may be present in the {R) or (S) configuration or (R,S) configuration. In certain instances, asymmetry may also be present due to restricted rotation about a given bond, for example, the central bond adjoining two substituted aromatic rings of the specified compounds. Substituents on a ring may also be present in either cis or trans form. It is intended that all such configurations (including enantiomers and diastereomers), are included within the scope of the present invention. Preferred compounds are those- with the absolute configuration of the compound of Formula (I) which produces the more desirabfe biological activity. Separated, pure or partially purified isomers or racemic mixtures of the compounds of this invention are also included within the scope of the present invention. The purification of said isomers and the separation of said isomeric mixtures can be accomplished by standard techniques known in the art.
The optical isomers can be obtained by resolution of the racemic mixtures according to conventional processes, for example, by the formation of diastereoi'someric salts using an optically active acid or base or formation of covaient diastereomers. Examples of appropriate acids are tartaric, diacetyltartaric, ditoluoyitartaric and camphorsulfonic acid. Mixtures of diastereoisomers can be separated into their individual diastereomers on the basis of their physical and/or chemical differences by methods known in the art, for example, by chrornatography or fractional crystallization. The optically active bases or acids are then (iberated from the separated diastereomeric salts. A different process for separation of optical isomers involves the use of chirai chrornatography (e.g., chira! HPLC columns), with or without conventional derivation, optimally chosen to maximize the separation of the enantiomers. Suitable chirai HPLC columns are manufactured by Diacel, e.g., Chiracel OD and Chiracel OJ among many others, all routinely selectable. Enzymatic separations, with or without derivitization, are also useful. The optically active compounds of Formula I can likewise be obtained by chirai syntheses utilizing opticafly active starting materials.
The present invention also relates to useful forms of the compounds as disclosed herein, such as pharmaceutically acceptable salts, metabolites and prodrugs of all the compounds Formula (I). The term "pharmaceuticaliy acceptable salt" refers to a relatively non-toxic, inorganic or organic acid addition salt of a compound of the present invention. For example, see S. M. Berge, et a/, "Pharmaceutical Salts," J. Pharm. Sci 1977, 66, 1-19. Pharmaceuticaliy acceptable salts include those obtained by reacting the main compound, functioning as a base, with an inorganic or organic acid to form a salt, for example, salts of hydrochloric acid, sulfuric acid, phosphoric acid, methane sulfonic acid, camphor sulfonic acid, oxalic acid, maleic acid, succintc acid and citric acid. Pharmaceutically acceptable salts also include those in which the main compound functions as an acid and is reacted with an-appropriate base to form, e.g., sodium, potassium, calcium, mangnesium, ammonium, and choline salts. Those skilled in the art will further recognize that acid addition salts of the claimed compounds may be prepared by reaction of the compounds with the appropriate inorganic or organic acid via any of a number of known methods. Alternatively, alkali and alkaline earth metal saits are prepared by reacting the compounds of the invention with the appropriate base via a variety of known methods.
Representative salts of the compounds of this invention include the conventional non-toxic salts and the quaternary ammonium salts which are formed, for example, from inorganic or organic acids or bases by means well known in the art. For example, such acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, carnphorsulfonate, cinnamate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glueoheptanoate, glycerophosphate, hemisuifate, heptanoate, hexanoate, hydrochforide, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, itaconate, lactate, maleate, mandelate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oxalate, pamoate, pectinate, persulfate, 3-phenylpropionate, picrate, pivalate, propionate, succinate, sulfonate, tartrate, thiocyanate, tosylate,
trifiuoromethanesulfonate, and undecanoate.
Base salts include alkali metal salts such as potassium and sodium salts, alkaline earth metal salts such as cafcium and magnesium salts, and ammonium salts with organic bases such as dicyclohexylamine and N-methyl-D-g!ucamine. Additionally, basic nitrogen containing groups may be quaternized with such agents as lower alkyl hafides such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides; dialkyl sulfates like dimethyl, diethyl, and dibutyl sulfate; and diamyf sulfates, long chain hafides such as decyl, lauryl, myristyl and strearyl. chlorides, bromides and iodides, aryl or aralkyl halides like benzyl and phenethyl bromides, and others monosubstituted aralkyl halides or polysubstituted aralkyf halides.
Certain pharmacologically active agents can be further modified with labile functional groups that are cleaved after in vivo administration to furnish the parent active agent and the pharmacologically inactive derivatizing group, These derivatives, commonly referred to as prodrugs, can be used, for example, to alter the physicochemical properties of the active agent, to target the active agent to a specific tissue, to alter the pharmacokinetic and pharmacodynamic properties of the active agent, and to reduce undesirable side effects, Prodrugs of the invention include, e.g., the esters of appropriate compounds of this invention that are well-tolerated, pharmaceutically acceptable esters such as alkyl esters including methyl, ethyl, propyl, isopropyl, butyl, isobutyl or pentyl esters. Additional esters such as phenyl-C1-C5 alkyl may be used, although methy! ester is preferred.
Methods which can be used to synthesize other prodrugs are described in the following reviews on the subject, which are incorporated herein by reference for their description of these synthesis methods:
• Higuchi, T.; Stella, V. eds. Prodrugs As Novel Drug Delivery Systems. ACS Symposium
Series. American Chemical Society: Washington, DC (1975).
• Roche, E. B. Design of Biopharmaceutical Properties through Prodrugs and Analogs,
American Pharmaceutical Association: Washington, DC (1977).
• Sinkula, A. A.; Yalkowsky, S. H. JPharm Sci, 1975, 64,181-210.
• Stella, V. L; Charman, W. N. Naringrekar, V. H. Drugs 1985, 29,455-473.
• Bundgaard, H., ed. Design of Prodrugs. Elsevier: New York (1985).
• Stella, V. J,; Himmelstein, K. J. J, Med. Chem. 1980, 23,1275-1282.
• Han, H-K; Amidon, G. L. AAPS Pharmsci 2000, 2, 1 - 11,
• Denny, W. A. Eur. J. Med. Chem. 2001, 36, 577-595.
• Wermuth, C. G. in Wemiuth, C. G. ed. The Practice of Medicinal Chemistry Academic
Press: San Diego (1996), 697-715.
• Balant, L. P.; Doelker, E. in Wolff, M. E. ed. Burgers Medicinal Chemistry And Drug
Discovery John Wiley & Sons: New York (1997), 949-982.
The metabolites of the compounds of this invention include oxidized derivatives of the compounds of Formula I, II, X, Y, Za, Zb, Zc and Zd, wherein one or more of the nitrogens are substituted with a hydroxy group; which includes derivatives where the nitrogen atom of the pyridine group is in the oxide form, referred to in the art as 1-oxo-pyridine or has a hydroxy substituent, referred to in the art as 1-hydroxy-pyridine.
General Preparative Methods
The particular process to be utilized in the preparation of the compounds used in this embodiment of the invention depends upon the specific compound desired. Such factors as the selection of the specific substituents play a role in the path to be followed in the preparation of the specific compounds of this invention. Those factors are readily recognized by one of ordinary skill in the art.
The compounds of the invention may be prepared by use of known chemical reactions and procedures. Nevertheless, the following general preparative methods are presented to aid the reader in synthesizing the compounds of the present invention, with more detailed particular examples being presented below in the experimental section describing the working examples.
All variable groups of these methods are as described in the generic description if they are not specifically defined below. When a variable group or substituent with a given symbol is used more than- once in a given structure, it is to be understood that each of these groups or substituents may be independently varied within the range of definitions for that symbol. It is recognized that compounds of the invention with each claimed optional functional group cannot be prepared with each of the below-listed methods. Within the scope of each method optional substituents are used which are stable to the reaction conditions, or the functional groups which may participate in the reactions are present in protected form where necessary, and the removal of such protective groups is completed at appropriate stages by methods well known to those skilled in the art.
The compounds of the invention can be made according to conventional chemical methods, and/or as disclosed below, from starting materials which are either commercially available or producible according to routine, conventional chemical methods. General methods for the preparation of the compounds, are given below, and the preparation of representative compounds is specifically illustrated in exampfes.
General Methods
Reaction Scheme 1: Synthesis of Ureas of formula
(Formula Removed)Aryl amines of formulas (III) or (V) are commercially available, or can be synthesized according to Method A or B, or methods commonly known to those skilled in the art. Aryl amines are commonly synthesized by reduction of nitroaryls using a metal catalyst, such as Ni, Pd, or Pt, and H2 or a hydride transfer agent, such as formate, cyclohexadiene, or a borohydride (Rylander. Hydrogenation Methods; Academic Press: London, UK (1985)). Nitroaryls may also be directly reduced using a strong hydride source, such as LiAlH4 (Seyden-Penne. Reductions by the Alumina- and horohydrides in Organic Synthesis; VCH Publishers: New York (1991)), or using a zero valent metal, such as Fe, Sn or Ca, often in acidic media. Many methods exist for the synthesis of nitroaryis (March. Advanced Organic Chemistry, 3rd Ed.; John Wiley: New York (1985). Larock. Comprehensive Organic Transformations; VCH Publishers: New York (1989)). Nitro aryls are commonly formed by electrophilic aromatic nitration using HNO3, or an alternative NO2+ source.
For the synthesis of compounds of formula (II) where L represents -O- or • -S-, and B, R2 and m are broadly defined as above, the nitroaryls are further elaborated prior to reduction. In Reaction Scheme 2 - method D, nitro aryls substituted with potential leaving groups such as F or Cl undergo substitution reactions on treatment with nucleophiles, such as phenoxide or thiolate, under basic conditions.
Another method for the preparation of the intermediate of Formula (II) is depicted in. Reaction Scheme 2 - Method C. The condensation of amine (V) with an appropriate substituted choropyridine has been previously described in the patent literatures, and can be adapted to the compounds of the present invention. For example, PCT Int. AppL, WO 99 32111, Dumas, J. et al., "Method for the Treatment of Neoplasm by inhibition of raf Kinase using N-Heteroaryl-N'-(hetero)arylureas", PCT Int. AppL, WO 99 32110, Dumas, J., et al., "Inhibition of raf Kinase using Aryl- and Heteroaryl- Substituted Heterocyclic Ureas",
Reactfon Scheme 3: Alternate Synthesis of Ureas of formula (I)
(Formula Removed)
The compounds of the invention may also be prepared from compounds of formula (VII) according to the reaction sequence shown in the General Methods G and H above. Using Method G, ureas of Formula (VI) are treated with a lewis acid such as magnesium chloride and the appropriate substituted amines, in a solvent such as THF
at room temperature, to provide substituted amides. In Method H, ureas of Formula (VI) are deesterified with a base such as potassium hydroxide, lithium hydroxide, or sodium hydroxide, Carboxylic acids of formula (VII) are coupled with the appropriate amines according to methods commonly known to those skilled in the art [e.g. from treatment of a carboxylic acid with DCC/DMAP or EDC!/ HOST], in a solvent such as THF, AcCN, or DMF. In addition, compounds of formula (I) where R4 and R5 are hydrogens may be synthesized according to the reaction scheme shown in Method I. The cyano compound (VIII) can be hydrolyzed in the presence of NaOH or sodium percarbonate, in aqueous solvent such as acetone-water, and at temperature from 20 to 100°C. Compounds of formula (VI) and (VIII) are synthesized according to methods A to F, or methods commonly known to those skilled in the art.
Pyridine-1 -oxides or Formula (I) where the pyridine ririg carries a hydroxy substituent on its nitrogen atom, and-A, 8, L are broadly defined as above can be prepared from the corresponding pyridines using oxidation conditions know in the art. Some examples are as follows:
• peracids such as meta chloroperbenzoic acids in chlorinated solvents such as
dichloromethane, dichloroethane, or chloroform (Markgraf et al,, Tetrahedron 1991,
47,183);
• (MeaSiO)2 in the presence of a catalytic amount of perrhenic acid in chlorinated
solvents such as dichloromethane (Coperet et al., Terahedron Lett. 1998,39, 761);
« Perfluoro-cis-2-butyl-3-propyloxaziridine in several combinations of halogenated
solvents (Amone et al., Tetrahedron 1998, 54, 7831); . Hypofluoric acid - acetonitrife complex in chloroform (Dayan et al., Synthesis 1999,
1427);
• Oxone, in the presence of a base such as KOH, in water (Robker et al., J. Chem.
Res., Synop. 1993, 1O, 412);
. Magnesium monoperoxyphthalate, in the presence of glacial acetic acid (Klemm et al., J. Heterocylic Chem. 1990, 6, 1537);
. Hydrogen peroxide, in the presence of water and acetic acid (Lin A.J., Org. Prep.
proced. Int. 1991,23(1), 114); • Dimethyldioxirane in acetone (Boyd et al., J. Chem., Soc., Perkin Trans. 1991, 9,
2189).
In addition, specific methods for preparing diaryl ureas and intermediate compounds (II) are already described in the patent literature, and can be adapted to the compounds of the present invention. For example, Miller S. et al, "Inhibition of p38 Kinase using Symmetrical and Unsymmetrical Diphenyl Ureas" PCT Int. Appl. WO 99 32463, Miller, S et al. "Inhibition of raf Kinase using Symmetrical and Unsymmetrical Substituted Diphenyl Ureas" PCT int. Appl., WO 99 32436, Dumas, J. et al., "Inhibition of p38 Kinase Activity using Substituted Heterocyclic Ureas" PCT Int. Appl., WO 99 32111, Dumas, J. et al., "Method for the Treatment of Neoplasm by Inhibition of raf Kinase using A/-Heteroaryl-A/'-(hetero)arylureas" PCT Int. Appl., WO 99 32106, Dumas, J. et al., "Inhibition of p38 Kinase Activity using Aryl- and Heteroaryl- Substituted Heterocyclic Ureas" PCT Int. Appl., WO 99 32110, Dumas, J., et al., "Inhibition of raf Kinase using Aryl- and Heteroaryl- Substituted Heterocyclic Ureas" PCT Int. Appl., WO 99 32455, Riedi, B., et al., "0-Carboxy Aryl Substituted Diphenyl Ureas as raf Kinase Inhibitors" PCT Int. Appl., WO 00 42012, Riedl, B., et al., "O-Carboxy Aryl Substituted Diphenyl Ureas as p38 Kinase Inhibitors" PCT Int. 'Appl., WO 00 41698, Dumas, J. et al. "Heteroaryl ureas containing nitrogen hetero-atoms as p38 kinase inhibitors" U.S. Pat. Appl. Publ., US 20020065296, Durnas, J. et al. "Preparation of N-aryl-N'-[(acylphenoxy) phenyl]ureas as raf kinase inhibitors" PCT Int. Appl., WO 02 62763, Dumas, J. et al. "Inhibition of raf kinase using quinolyl, isoquinolyl or pyridyl ureas" PCT Int. Appl., WO 02 85857, Dumas, J. et al. "Preparation of quinolyl, isoquinolyl or pyridyl-ureas as inhibitors of raf kinase for the treatment of tumors and/or cancerous cell growth" U.S. Pat. Appl. Publ., US 20020165394. All the preceding patent applications are hereby incorporated by reference.
The reaction of the compounds (III) or (IV) with (II) is carried out preferably in a solvent. Suitable solvents comprise the customary organic solvents which are inert under the reaction conditions. Non-limiting examples include ethers such as diethyl ether, dioxane, tetrahydrofuran, 1,2-dimethoxy ethane; hydrocarbons such as benzene, toluene, xylene, hexane, cyclohexane, mineral oil fractions; halogenated hydrocarbons such as dichloromethane, trichloromethane, carbon tetrachlbride, dichloroethane, trichioroethylene, chlorobenzene; alcohols such as methanol, ethanol, n-propanol, isopropanol; esters such as ethyl acetate; ketones such as acetone; nitriles such as acetonitrile; heteroaromatics such as pyridine; polar solvents such as dimethyl formamide and hexamethyl phosphoric acid tris-amide; and mixtures of the above-mentioned solvents, Toluene, benzene, and dichloromethane are preferred.
The compounds (111) are generally employed in an amount of from about 1 to 3 mol per mol of compounds (II); an equimolar amount or slight excess of compounds (III) is preferred.
The reaction of the compounds (II) with (III) is generally carried out within a relatively wide temperature range, ln general, they are carried out in a range of from about -20 to 200°C, preferably from about 0 to 100°C, and more preferably from about 25 to 50°C. The steps of this reaction are generally carried out under atmospheric pressure. However, it is also possible to carry them out under superatmospheric pressure or under reduced pressure (for example, in a range of from about 0.5 to 5 bar). The reaction time can generally be varied within a relatively wide range. In general, the. reaction is finished after a period of from about 2 to 24 hours, preferably from about 6 to 12 hours.
Synthetic transformations that may be employed in the synthesis of compounds of Formula (I) and in the synthesis of intermediates involved in the synthesis of compounds of Formula (I) are known by or accessible to one skilled in the art, Collections of synthetic transformations may be found in compilations, such as:
• J. March. Advanced Organic Chemistry, 4th ed.; John Wiley: New York (1992);
• R.G. Larock. Comprehensive Organic Transformations, 2nd ed.; Wiiey-VCH: New
York (1999);
. FA Carey; R.J. Sundberg. Advanced Organic Chemistry, 2nd ed.; Plenum Press: New York (1984);
• T.W. Greene; P:G.M. Wuts. Protective Groups in Organic Synthesis, 3rd ed.; John
Wiley: New York (1999);
• L.S. Hegedus. Transition Metals in the Synthesis of Complex Organic Molecules,
2nd ed.; University Science Books: Mill Valley, CA (1994);
• L.A. Paquette, Ed. The Encyclopedia of Reagents for Organic Synthesis; John
Wiley: New York (1994);
• A.R. Katritzky; Q. Meth-Cohn; C.W. Rees, Eds. Comprehensive Organic Functional
Group Transformations', Pergamon Press: Oxford, UK (1995);
• G. Wilkinson; F.G A. Stone; E.W. Abel, Eds. Comprehensive Organometallic
Chemistry; Pergamon Press: Oxford, UK (1982);
• B.M. Trost; I. Fleming. Comprehensive Organic Synthesis', Pergamon Press:
Oxford, UK (1991);
• A.R. Katritzky; C.W. Rees Eds. Comprehensive Heterocylic Chemistry; Pergamon
Press: Oxford, UK (1984);
. A.R. Katritzky; C.W. Rees; E.F.V, Scriven, Eds.. Comprehensive Heterocylic
Chemistry II; Pergamon Press: Oxford, UK (1996); and o C. Hansch; P.G. Samrries; J.B. Taylor, Eds. Comprehensive Medicinal Chemistry:
Pergamon Press: Oxford, UK (1990).
In addition, recurring reviews of synthetic methodology and related topics include Organic Reactions; John Wiley: New York; Organic Syntheses; John Wiley: New York; Reagents for Organic Synthesis: John Wiley: New York; The Total Synthesis of Natural Products; John Wiley: New York; The Organic Chemistry of Drug Synthesis; John Wiley: New York; Annual Reports in Organic Synthesis; Academic Press: San Diego CA; and Methoden der Organischen Chemie (Houben-Wey'l); Thieme: Stuttgart,
Germany. Furthermore, databases of synthetic transformations include Chemical Abstracts, which may be searched using either CAS Online or SciFinder, Hahdbuch der Organischen Chemie (Beilstein), which may be searched using SpotFire, and REACCS.
Compositions of the compounds of this invention
This invention also relates to pharmaceutical compositions containing one or more compounds of the present invention. These compositions can be utilized to achieve the desired pharmacological effect by administration to a patient in need thereof. A patient, for the purpose of this invention, is a mammal, including a human, in need of treatment for the particular condition or disease. Therefore, the present invention includes pharmaceutical compositions which are comprised of a pharmaceutically acceptable carrier and a pharmaceutically effective amount of a compound of the present invention, (the compounds of formula 1, their salts, prodrugs and metabolites, including diastereoisomeric forms). A pharmaceutically acceptable carrier is preferably a carrier which is relatively non-toxic and innocuous to a patient at concentrations consistent with effective activity of the active ingredient so that any side effects ascribable to the carrier do not vitiate the beneficial effects of the active ingredient. A pharmaceutically effective amount of compound is preferably that amount which produces a result or exerts an influence on the particular condition being treated. The compounds of the present invention can be administered with pharmaceutically-acceptable carriers well known in the art using any effective conventional dosage unit forms, including immediate, slow and timed release preparations, orally, parenteraily, topically, nasally, ophthalmically, optically, sublinguatly, rectally, vaginally, and the like.
For oral administration, the compounds can be formulated into solid or liquid preparations such as capsules, pills, tablets, troches, lozenges, melts, powders, solutions, suspensions, or emulsions, and may be prepared according to methods known to the art for the manufacture of pharmaceutical compositions. The solid unit
dosage forms can be a capsule which can be of the ordinary hard- or soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers such as lactose, sucrose, calcium phosphate, and corn starch.
In another embodiment, the compounds of this invention may be tableted with conventional tablet bases such as lactose, sucrose and cornstarch in combination with binders such as acacia, corn starch or gelatin, disintegrating agents intended to assist the break-up and dissolution of the tablet following administration such as potato starch, alginic acid, corn starch, and guar gum, gum tragacanth, acacia, lubricants intended to improve the flow of tablet granulation and to prevent the adhesion of tablet material to the surfaces of the tablet dies and punches, for example talc, stearic acid,, or magnesium, calcium or zinc stearate, dyes, coloring agents, and flavoring agents such as peppermint, oil of wintergreen, or cherry flavoring, intended to enhance the aesthetic qualities of the- tablets .and make them more acceptable to the patient. Suitable excipients for use in oral liquid dosage forms include dicalcium phosphate and diluents such as water and alcohols, for example, ethanol, benzyl alcohol, and polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent or emulsifying agent. Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance tablets, pills or capsules may be coated with shellac, sugar or both.
Dispersible powders and granules are suitable for the preparation of an aqueous suspension. They provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example those sweetening, flavoring and coloring agents described above, may also be present.
The pharmaceutical compositions of this invention may also be in the form of oil-in-water emulsions. The oily phase may be a vegetable oil such as liquid paraffin
mixture of vegetable oils. Suitable emulsifying agents may be (1) naturally occurring gums such as gum acacia and gum tragacanth, (2) naturally occurring phosphatides such as soy bean and lecithin, (3) esters or partial esters derived form fatty acids and hexitol anhydrides, for example, sorbitan monooleate, (4) condensation products of said partial esters with ethylene oxide, for example, polyoxyethylene sorbitan monooleate. The emulsions may also contain sweetening and flavoring agents.
Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil such as, for example, arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oily suspensions may contain a thickening agent such as, for. example, beeswax, hard paraffin, or cetyl alcohol. The suspensions may also contain one or more preservatives, for example, ethyl or n-propyl p-hydroxybenzoate; one or more coloring agents; one or more flavoring agents; and one or more sweetening agents such as sucrose or saccharin.
Syrups and elixirs may be formulated with sweetening agents such as, for example, glycerol,.propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, and preservative, such as methyl and propyl parabens and • flavoring and coloring agents.
The compounds of this invention may also be administered parenterally, that is, subcutaneously, intravenously, intraocularly, intrasynovially, intramuscularly, or interperitoneally, as injectable dosages of the compound, preferably in a physiologically acceptable diluent with a pharmaceutical carrier which can be a sterile liquid or mixture of liquids such as water, saline, aqueous dextrose and related sugar solutions, an alcohol such as ethanol, isopropanol, or hexadecyl alcohol, glycols such as propylene glycol or polyethylene giycol, glycerol ketals such as 2,2-dimethyl-1,1-dioxolane-4-methanol, ethers such as poly(ethylene glycol) 400, an oil, a fatty acid, a fatty acid ester or, a fatty acid glyceride, or an acetylated fatty acid glyceride, with or without the addition of a pharmaceutically acceptable surfactant such as a soap .or a detergent,
suspending agent such as pectin, carbomers, methycellulose, hydroxypropylmethylcellulose, or carboxymethylcellulose, or emulsifying agent and other pharmaceutical adjuvants.
Illustrative of oils which can be used in the parenteral formulations of this invention are those of petroleum, animal, vegetable, or synthetic origin, for example, peanut oil, soybean oil, sesame oil, cottonseed oil, corn oil, olive oil, petrolatum and mineral oil. Suitable fatty acids include oleic acid, stearic acid, isostearic acid and myristic acid. Suitable fatty acid esters are, for example, ethyl oleate and isopropyl myristate. Suitable soaps include fatty acid alkali metal, ammonium, and triethanolamine salts and suitable detergents include cationic detergents, for example dimethyl dialkyl ammonium halides, alkyl pyridinium halides, and alkylamine acetates; anionic detergents, for example, alkyl, aryl, and olefin sulfonates, alky], olefin, ether, and monoglyceride sulfates, and sulfosuccinates; non-ionic detergents, for example, fatty amine oxides, fatty acid alkanolamides, and poly(oxyethylene-oxypropylene)s or ethylene oxide or propylene oxide copolymers; and amphoteric detergents, for example, alkyl-beta-aminopropionates, and 2-alkylimidazoline quarternary ammonium salts, as well as mixtures.
The parenteral compositions of this invention will typically contain from about 0.5% to about 25% by weight of the active ingredient in solution. Preservatives and buffers may also be used advantageously. In order to minimize or eliminate irritation at the site of injection, such compositions may contain a non-ionic surfactant having a hydrophile-lipophile balance (HLB) preferably of from about 12 to about 17. The quantity of surfactant in such formulation preferably ranges from about 5% to about 15% by weight. The surfactant can be a single component having the above HLB or can be a mixture of two or more components having the desired HLB.
Illustrative of surfactants used in parenteral formulations are the class of polyethylene sorbitan fatty acid esters, for example, sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the
condensation of propylene oxide with propylene glycol.
The pharmaceutical compositions may be in the form of sterile injectable aqueous suspensions. Such suspensions may be formulated according to known methods using suitable dispersing or wetting agents and suspending agents such as, for example, sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethyl-cellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents which may be a naturally occurring phosphatide such as lecithin, a condensation product of an alkylene oxide with a fatty acid, for example, polyoxyethylene stearate, a condensation product of ethylene oxide with a long chain aliphatic alcohol, for example, heptadeca-ethyleneoxycetanol, a condensation product of ethylene oxide with a partial ester derived form a fatty acid and a hexitol such as polyoxyethylene sorbitol monooleate, or a condensation product of an ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride, for example polyoxyethylene sorbitan monooleate.
The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent. Diluents and solvents that may be employed are, for example, water, Ringer's solution, isotonic sodium chloride solutions and isotonic glucose solutions. In addition, sterile fixed oils are conventionally employed as solvents or suspending media. For this purpose, any bland, fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid can be used in the preparation of injectables.
A composition of the invention may also be administered in the form of suppositories for rectal administration of the drug. These compositions can be prepared by mixing the drug with a suitable non-irritation excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such material are, for example, cocoa butter and polyethylene glycol.
Another formulation employed in the methods of the present invention employs
transdermal delivery devices ("patches"). Such transdermal patches may be used to provide continuous or discontinuous infusion of the compounds of the present invention in controlled amounts. The construction and use of transdermal patches for the delivery of pharmaceutical agents is well known in the art (see, e.g., US Patent No. 5,023,252, issued June 11, 1991, incorporated herein by reference). Such patches may be constructed for continuous,- pulsatile, or on demand delivery of pharmaceutical agents.
Controlled release formulations for parenteral administration include liposomal, polymeric microsphere and polymeric gel formulations which are known in the art.
It may be desirable or necessary to introduce the pharmaceutical composition to the patient via a mechanical delivery device. The construction and use of mechanical delivery devices for the delivery of pharmaceutical agents is well known in the art. Direct techniques for, for example, administering a drug directly to the brain usually involve placement of a drug delivery catheter into the patient's ventricular system to bypass the blood-brain barrier. One such implantable delivery system, used for the transport of agents to specific anatomical regions of the body, is described in US Patent No. 5,011,472, issued April 30, 1991.
The compositions of the invention can also contain other conventional pharmaceutically acceptable compounding ingredients, generally referred to as carriers or diluents, as necessary or desired. Conventional procedures for preparing such composition's in appropriate dosage forms can be utilized. Such ingredients and procedures include those described in the following references, each of which is incorporated herein by reference: Powell, M.F. et al, "Compendium of Excipients for Parenteral Formulations" PDA Journal of Pharmaceutical Science & Technology 1998, 52(5), 238-311; StrickJey, R.G "Parenteral Formulations of Small Molecule Therapeutics Marketed in the United States (1999)-Part-1" PDA Journal of Pharmaceutical Science & Technology 1999, 53(6), 324-349; and Nema, S. et al,
"Excipients and Their Use in Injectable Products" PDA Journal of Pharmaceutical Science & Technology 1997, 51(4), 166-171.
Commonly used pharmaceutical ingredients which can be used -as appropriate to formulate the composition for its intended route of administration include:
acidifying agents (examples include but are not limited to acetic acid, citric acid, fumaric acid, hydrochloric acid, nitric acid);
alkalinizing agents (examples include but are not limited to ammonia solution, ammonium carbonate, diethanolamine, monoethanolamine, potassium hydroxide, sodium borate, sodium carbonate, sodium hydroxide, triethanolamine, trolamine);
adsorbents (examples include but are not limited to powdered cellulose and activated charcoal);
aerosol propellants (examples include but are not limited to carbon dioxide, CCI2F2, F2CIC-CCIF2 and CCIF3);
air displacement agents (examples include but are not limited to nitrogen and argon);
antifungal preservatives (examples include but are not limited to benzoic acid, butylparaben, ethylparaben, methyl paraben, propylparaben, sodium benzoate);
antimicrobial preservatives (examples include but are not limited to benzalkonium chloride, benzethonium chloride, benzyl alcohol, cetylpyridinium chloride, chlorobutanol, phenol, phenylethyl alcohol, phenyimercuric nitrate and thimerosal);
antioxidants (examples include but are not limited to ascorbic acid, ascorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, hypophosphorus acid, monothioglycerol, propyl gallate, sodium ascorbate, sodium bisulfite, sodium formaldehyde sulfoxylate, sodium metabisulfite);
binding materials (examples include but are not limited to block polymers, natural and synthetic rubber, polyacrylates, polyurethanes, silicones, polysiloxanes and styrene-butadiene copotymers);
buffering agents (examples include but are not limited to potassium metaphosphate, dipotassium phosphate, sodium acetate, sodium citrate anhydrous and sodium citrate dihydrate);
carrying agents (examples include but are not limited to acacia syrup, aromatic syrup, aromatic elixir, cherry syrup, cocoa syrup, orange syrup, syrup, corn oil, mineral oil, peanut oil, sesame oil, bacteriostatic sodium chloride injection and bacteriostatic water for injection);
chelating agents (examples include but are not limited to edetate disodium and edetic acid);
colorants (examples include but are not limited to FD&C Red No. 3, FD&C Red No. 20, FD&C Yellow No. 6, FD&C Blue No. 2, D&C Green No. 5, D&C Orange No. 5, D&C Red No. 8, caramel and ferric oxide red);
clarifying agents (examples include but are not limited to bentonite); emulsifying agents (examples include but are not limited to acacia, cetomacrogol, cetyl alcohol, giyceryl monostearate, lecithin, sorbitan monooleate, polyoxyethylene 50 monostearate);
encapsulating agents (examples include but are not limited to gelatin and cellulose acetate phthalate);
flavorants (examples include but are not limited to anise oil, cinnamon oil, cocoa, menthol, orange oil, peppermint oil and vanillin);
humectants (examples include but are not limited to glycerol, propylene glycol and sorbitol);
levigating agents (examples include but are not limited to mineral oil and glycerin);
oils (examples include but are not limited to arachis oil, mineral oil, olive oif, peanut oil, sesame oil and vegetable oil);
ointment bases (examples include but are not limited to lanolin, hydrophilic ointment, polyethylene glycol ointment, petrolatum, hydrophilic petrolatum, white ointment, yellow ointment, and rose water ointment);
penetration enhancers (transdermal delivery) (examples include but are not limited to monohydroxy or polyhydroxy alcohols, mono-or polyvalent alcohols, saturated or unsaturated fatty alcohols, saturated or unsaturated fatty esters, saturated or unsaturated dicarboxyiic acids, essential oils, phosphatidyl derivatives, cephalin, terpenes, amides, ethers, ketones and ureas);
plasticizers (examples include but are not limited to diethyl phthalate and glycerol);
solvents (examples include but are not limited to ethanol, corn oil, cottonseed oil, glycerol, isopropanol, mineral oil, oleic acid, peanut oil, purified water, water for injection, sterile water for injection and sterile water for irrigation);
stiffening agents (examples include but are not limited to cetyi alcohol, cetyl esters wax, microcrystalline wax, paraffin, stearyl alcohol, white wax and yellow wax);
suppository bases (examples include but are not limited to cocoa butter and polyethylene glycols (mixtures));
surfactants (examples include but are not limited to benzalkonium chloride, nonoxynol 10, oxtoxynol 9, polysorbate 80, sodium lauryl sulfate and sorbitan mono-palmitate);
suspending agents (examples include but are not limited to agar, bentonite, carbomers, carboxymethylcellulose sodium, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, kaolin, methylcellulose, tragacanth and veegum);
sweetening agents (examples include but are not limited to aspartame, dextrose, glycerol, mannitol, propylene glycol, saccharin sodium, sorbitol and sucrose);
tablet anti-adherents (examples include but are not limited to magnesium stearate and talc);
tablet binders (examples include but are not limited to acacia, alginic acid, carboxymethylcellulose sodium, compressible sugar, ethylcellulose, gelatin, liquid glucose, methylcellulose, non-crosslinked polyvinyl pyrrolidone, and pregelatinized starch);
tablet and capsule diluents (examples include but are not limited to dibasic calcium phosphate, kaolin, lactose, mannitol, microcrystalline cellulose, powdered cellulose, precipitated calcium carbonate, sodium carbonate, sodium phosphate, sorbitol and starch);
tablet coating agents (examples include but are not limited to liquid glucose, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, methylcellulose, ethylcellulose, cellulose acetate phthalate and shellac);
tablet direct compression excipients (examples include but are not limited to dibasic calcium phosphate);
tablet disintegrants (examples include but are not limited to alginic acid, carboxymethylcellulose calcium, microcrystalline cellulose, polacrillin potassium, cross-linked polyvinylpyrrolidone, sodium alginate, sodium starch glycollate and starch);
tablet glidants (examples include but are not limited to colloidal silica, corn starch and talc);
tablet lubricants (examples include but are not limited to calcium stearate, magnesium stearate, mineral oil, stearic acid and zinc stearate);
tablet/capsule opaquants (examples include but are not limited to titanium dioxide);
tablet polishing agents (examples include but are not limited to carnauba wax and white wax);
thickening agents (examples include but are not limited to beeswax, cetyl alcohol and paraffin);
tonicity agents (examples include but are not limited to dextrose and sodium chloride);
viscosity increasing agents (examples include but are not limited to alginic acid, bentonite, carbomers, carboxymethylcellulose sodium, methylcellulose, polyvinyi pyrrolidone, sodium alginate and tragacanth); and
wetting agents (examples include but are not limited to heptadecaethylene oxycetanol, lecithins, sorbitol monooleate, polyoxyethylene sorbitol monooleate, and polyoxyethylene stearate).
Pharmaceutical, compositions according to the present invention can be illustrated as follows:
Sterile IV Solution: A 5 mg/ml solution of the desired compound of this invention can be made using sterile, injectable water, and the pH is adjusted if necessary. The solution is diluted for administration to 1 - 2 mg/ml with sterile 5% dextrose and is administered as an IV infusion over 60 minutes.
Lyophilized powder for IV administration: A sterile preparation can be prepared with (I) 100 ~ 1000 mg of the desired compound of this invention as a iypholized powder, (ii) 32- 327 mg/ml sodium citrate, and (iii) 300 - 3000 mg Dextran 40. The formulation is reconstituted with sterile, injectable saline or dextrose 5% to a concentration of 10 to 20 mg/m!, which is further diluted with saline or dextrose 5% to 0.2 - 0.4 mg/ml, and is administered either IV bolus or by IV infusion over 15-60 minutes.
.Intramuscular suspension: The following solution or suspension can be prepared, for intramuscular injection:
50 mg/mi of the desired, water-insoluble compound of this invention
5 mg/ml sodium carboxymethylcellulose
4 mg/ml TWEEN 80
9 mg/ml sodium chloride
9 mg/ml benzyl alcohol
Hard Shell Capsules: A large number of unit capsules are prepared by filling standard two-piece hard galantine capsules each with 100 mg of powdered active ingredient, 150 mg of lactose, 50 mg of cellulose and 6 mg of magnesium stearate.
Soft Gelatin Capsules: A mixture of active ingredient in a digestible oil such as soybean oil, cottonseed oil or olive oil is prepared and injected by means of a positive
displacement pump into molten gelatin to form soft gelatin capsules containing 100 mg of the active ingredient. The capsules are washed and dried. The active ingredient can be dissolved in a mixture of polyethylene glycol, glycerin and sorbitof to prepare a water miscible medicine mix.
Tablets; A large number of tablets are prepared by conventional procedures so that the dosage unit is 100 mg of active ingredient, 0.2 mg. Of colloidal silicon dioxide, 5 mg of magnesium stearate, 275 mg of microcrystalline cellulose, 11 mg of starch, and 98.8 mg of iactose. Appropriate aqueous, and non-aqueous coatings may be applied to increase payability, improve elegance and stability or delay absorption.
Immediate Release Tablets/Capsules: These are solid oral dosage forms made by conventionai and novel processes. These units are taken oraiiy without water for immediate dissolution and delivery of the medication. The active ingredient is mixed in a liquid containing ingredient such as sugar, gelatin, pectin and sweeteners. These liquids are solidified into solid tablets or caplets by freeze drying and solid state extraction techniques. The drug compounds may be compressed with viscoelastic and thermoelastfc sugars and polymers or effervescent components to produce porous matrices intended for immediate release, without the need of water.
Method of treating hyper-proliferative disorders
The present invention relates to a method for using the compounds of Formula (I) as described above, including salts and esters thereof and compositions thereof, to treat mammalian hyper-proliferative disorders. This method comprises administering to a mammal in need thereof, including a human, an amount of a compound of this invention, or a pharmaceutically acceptable salt or ester thereof, which is effective to treat the disorder, Hyper-proliferative disorders include but are not limited to solid tumors, such as cancers of the breast, respiratory tract, brain, reproductive organs, digestive tract, urinary tract, eye, liver, skin, head and neck, thyroid, parathyroid and
their distant metastases. Those disorders also include lymphomas, sarcomas, and leukemias.
Examples of breast cancer include, but are not limited to invasive ducta! carcinoma, invasive lobuiar carcinoma, ductal carcinoma in situ, and lobular carcinoma in situ.
Examples of cancers of the respiratory tract include, but are not limited to small-cell and non-small-cell lung -carcinoma, as well as bronchial adenoma and pleuropulmonary blastoma.
Examples of brain cancers include,. but are not limited to brain stem and hypophtalmic glioma, cerebellar and cerebral astrocytoma, medulloblastoma, ependymoma, as well as neuroectodermal and pineal'tumor.
Tumors of the male reproductive organs include, but are not limited to prostate and testicular cancer. Tumors of the female reproductive organs include, but are not limited to endometrial, cervical, ovarian, vaginal, and vulvar cancer, as well as sarcoma of the uterus.
Tumors of the digestive tract include, but are not limited to anal, colon, colorectal, esophageal, gallbladder, gastric, pancreatic, rectal, small-intestine, and salivary gland cancers.
Tumors of the urinary tract include, but are not limited to bladder, penile, kidney, renal pelvis, ureter, and urethral cancers.
Eye cancers include, but are not limited to intraocular melanoma and retinoblastoma.
Examples of liver cancers include, but are not limited to hepatocellular carcinoma (liver cell carcinomas with or without fibrolamellar variant), cholangiocafcinoma (intrahepatic bile duct carcinoma), and mixed hepatocellular cholangiocarcinoma.
Skin cancers include, but are not limited to squamous cell carcinoma, Kaposi's sarcoma, malignant melanoma, Merkel cell skin cancer, and non-melanoma skin cancer.
Head-and-neck cancers include, but are not limited to laryngeal / hypopharyngeal nasopharyngeaf / oropharyngeal cancer, and lip and oral cavity cancer.
Lymphomas include, but are not limited to AIDS-related lymphoma, non-Hodgkin's lymphoma, cutaneous T-cell lymphoma, Hodgkin's disease, and lymphoma of the central nervous system.
Sarcomas include, but are not limited to sarcoma of the soft tissue, osteosarcoma, malignant fibrous histiocytoma, fymphosarcoma, and rhabdomyosarcoma.
Leukemias include, but are not limited to acute myeloid leukemia, acute, lymphoblastic leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, and hairy cell leukemia.
These disorders have been well characterized in humans, but also exist with a similar etiology in other mammals, and can be treated by administering pharmaceutical compositions of the present invention.
Based upon standard laboratory techniques known to evaluate compounds useful for the treatment of hyper-proliferative disorders, by standard toxicity tests and by standard pharmacological assays for the determination of treatment of the conditions
identified above in mammals, and by comparison of these results with the results of known medicaments that are used to treat these conditions, the effective dosage of the compounds of this invention can readily be determined for treatment of each desired indication. The amount of the active ingredient to be administered in the treatment of one of these conditions can vary widely according to such considerations as the particular compound and dosage unit employed, the mode of administration, the period of treatment, the age and sex of the patient treated, and the nature and extent of the condition treated.
The total amount of the active ingredient to be administered will generally range from about 0.001 mg/kg to about 200 mg/kg, and preferably from about 0.01 mg/kg to about 20 mg/kg body weight per day. A unit dosage may contain from about 0.5 mg to about 1500 mg of active ingredient, and can be administered for example, one or more times per day. In some cases every other day or weekly administration is suitable. Clinically useful dosing schedules will range from three times a day dosing to once every four weeks dosing. In addition, "drug holidays" in which a patient is not dosed with a drug for a certain period of time, may be beneficial to the overall balance between pharmacological effect and tolerability. The average daily dosage for administration by injection, including intravenous, intramuscular, subcutaneous and parenteral injections, and use of infusion techniques will preferably be from about 0.01 to 200 mg/kg of total body weight. The average daily rectal dosage regimen will preferably be from about 0.01 to 200 mg/kg of total body weight. The average daily vaginal dosage regimen will preferably be from about 0.01 to 200 mg/kg of total body weight. The average daily topical dosage regimen will preferably be from about 0.1 to 200 mg administered between one to four times daily. The transdermal concentration will preferably be that required to maintain an average daily dose of from about 0.01 to 200 mg/kg, The average daily inhalation dosage regimen will preferably be from about 0.01 to 100 mg/kg of total body weight.
The specific initial and continuing dosage regimen for each patient will vary
according to the nature and severity of the condition as determined by the attending diagnostician, the activity of the specific compound employed, the age and general condition of the patient, time of administration, route of administration, rate of excretion of the drug, drug combinations, and the like. The desired mode of treatment and number of doses of a compound of the present invention or a pharmaceuticaliy acceptable salt or ester or composition thereof can be ascertained by those skilled in the art using conventional treatment tests.
The compounds of this invention can be administered as the sole pharmaceutical agent or in combination with one or more other pharmaceutical agents where the combination causes no unacceptable adverse effects. For example, the compounds of this invention can be combined with known anti-hyper-proliferative or other indication agents, and the like, as well as with admixtures and combinations thereof. Examples of known anti-hyper-proliferative or other indication agents, and the like, include, but are not limited to aldesleukin, alendronic acid, alfaferone, alitretinoin, allopurinol, aloprim, aloxi, altretamine, aminoglutethimide, amifostine, amrubicin, amsacrine, anastrozole, anzmet, aranesp, arglabin, arsenic trioxide, aromasin, 5-azacytidine, azathioprine, BCG or tice BCG, bestat/n, betamethasone acetate, betamethasone sodium phosphate, bexarotene, bleomycin sulfate, broxuridine , bortezomib, busulfan, calcitonin, campath, capecitabine, carboplatin, casodex, cefesone, celmoleukin, cerubidine, chlorambucil, cisplatin, cladribine, cladribine, clodronic acid, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, DaunoXome, decadron, decadron phosphate, delestrogen, denileukin diftitox, depo-medrol, deslorelin, dexrazoxane, diethylstilbestrol, diflucan, docetaxel, doxifluridine, doxorubicin, dronabinol, DW-166HC, eligard, elitek, ellence, emend, epirubicin, epoetin alfa, epogen, eptaplatin, ergamisol, estrace, estradiol, estramustine phosphate sodium, ethinyl estradiol, ethyol, etidronic acid, etopophos, etoposide, fadrozole, farston, filgrastim, finasteride, fligrastim, floxuridine, fluconazole, fludarabine, 5-fluorodeoxyuridine monophosphate, 5-fluorouracil (5-FU), fluoxymesterone, flutamide, formestane, fosteabine, fotemustine, fulvestrant, gammagard, gemcitabine, gemtuzumab, gleevec, gliadel, goserelin, granisetron HCI,
histrelin, hycamtin, hydrocortone, eyrthro-hydroxynonyladenine, hydroxyurea, ibritumomab tiuxetan, idarubicin, ifosfamide, interferon alpha, interferon-alpha 2, interferon alfa-2A, interferon a!fa-2B, interferon aifa-n1, interferon alfa-n3, interferon beta, interferon gamma-la, interleukin-2, intron A, iressa, irinotecan, kytril, lentinan sulphate, letrozole, leucovorin, leuproiide, leuprolide acetate, levamisole, levofolinic ac. calcium salt, levothroid, ievoxyl, lomustine, lonidamine, marinol, mechlorethamine, mecobalamin, medroxyprogesterone acetate, megestrol acetate, melphalan, menest, 6-mercaptopurine, Mesna, methotrexate, metvix, miltefosine, minocycline, mitomycin C, mitotane, mitoxantrone, Modrenal, Myocet, nedaplatin, neulasta, neumega, neupogen, nitutamide, nolvadex, NSC-631570, OCT-43, octreotide, ondansetron HCI, orapred, oxaliplatin, paclitaxel, pediapred, pegaspargase, Pegasys, pentostatin, picibanil, pilocarpine HCI, pirarubicin, plicamycin, porfimer sodium, prednimustine, prednisolone, prednisone, premarin, procarbazine, procrit, raltitrexed, rebif, rheniurn-186 etidronate, rituximab, roferon-A, romurtide, salagen, sandostatin, sargramostim, semustine, sizofiran, sobuzoxane, solu-medrol, sparfosic acid, stem-cell therapy, streptozocin, strontium-89 chloride, synthroid, tamoxifen, tamsulosin, tasonermin, tastolactone, taxotere, teceleukin, temozolomide, teniposide, testosterone propionate, testred, thioguanine, thiotepa, thyrotropin, tiludronic acid, topotecan, toremifene, tositumomab, trastuzumab, treosulfan, tretinoin, trexali, trimethylmelamine, trimetrexate, triptorelin acetate, triptorelin pamoate, UFT, uridine, valrubicin, vesnarinone, vinblastine, vincristine, vindesine, vinorelbine, virulizin, zinecard, zinostatin stimalamer, and zofran.
In addition, compounds of this invention can be administered in combination with one or more other pharmaceutical agents currently being studied for use as anti-hyper-proliferative or other indication agents, and the like, as well as with admixtures and combinations thereof. Examples of pharmaceutical agents currently being studied for use as anti-hyper-proliferative or other indication agents, and the like, include, but are not limited to ABl-007, acolbifene, actimmune, affinitak, aminopterin, arzoxifene, asoprisnil, atamestane, atrasentan, BAY 43-9006, avastin, CCI-779, CDC-501, celebrex, cetuximab, crisnatbl, cyproterone acetate, decitabine, DN-101, doxorubicin-MTC, dSLIM, dutasteride, edotecarin, eflomithine, exatecan, fenretinide, nistamine
dihydrochloride, histrelin hydrogel implant, holmium-166 DOTMP, ibandronic acid, interferon gamma, intron-PEG, ixabepilone, keyhole limpet hemocyanin, L-651582, lanreotide, lasofoxifene, libra, lonafarnib, miproxifene, minodronate, MS-209, liposomal MTP-PE, MX-6, nafarelin, nemorubicin, neovastat, nolatrexed, oblimersen, onco-TCS, osidem, paclitaxel polyglutamate, pamidronate disodium, PN-401, QS-21, quazepam, R-1549, raloxifene, ranpirnase, 13-cis -retinoic acid, satraplatin, seocalcitol, T-138067, tarceva, taxoprexin, thymosin alpha 1, tiazofurine, tipifarnib, tirapazamine, TLK-286, toremifene, TransMID-107R, valspodar, vapreotide, vatalanib, verteporfin, vinflunine, Z-100, and zoledronic acid.
Generally, the use of cytotoxic and/or cytostatic agents in combination with a compound or composition of the present invention will serve to:
(1) yield better efficacy in reducing the growth of a tumor or even eliminate the
tumor as compared to administration of either agent alone,
(2) provide for the administration of lesser amounts of the administered
chemotherapeutic agents,
(3) provide for a chemotherapeutic treatment that is well tolerated in the patient
with fewer deleterious pharmacological complications than observed with single agent
chemotherapies and certain other combined therapies,
(4) provide for treating a broader spectrum of different cancer types in mammals,
especially humans,
(5) provide for a higher response rate among treated patients,
(6) provide for a longer survival time among treated patients compared to
standard chemotherapy treatments,
(7) provide a longer time for tumor progression, and/or
(8) yield efficacy and tolerability results at least as good as those of the agents
used alone, compared to known instances where other cancer agent combinations
produce antagonistic effects.
It is believed that one skilled in the art, using the preceding information and information available in the art, can utilize the present invention to its fullest extent.
It should be apparent to one of ordinary skill in the art that changes and modifications can be made to this invention without departing from the spirit or scope of the invention as it is set forth herein.
The topic headings set forth above and below are meant as guidance where certain information can be found in the application, but are not intended to be the only source in the application where information on such topic can be found.
All publications and patents cited above and below are incorporated herein by reference.
The present invention provides, but is not limited, to the embodiments defined in the following paragraphs:
EXAMPLES
Unless otherwise stated the reaction conditions, methods, abbreviations and reagents are listed below. All temperatures are in degrees Celsius (°C) and all parts and percentages are by weight. Commercial grade reagents and solvents were used without further purification.
Abbreviations used in this specification
DBU 1,8-diazabicyclo[5.4.0]undec-7-ene
DMF N,N-dimethyl formamide
DCM dichloromethane
DCE 1,2-dichloroethane
DMSO dimethyl sulphoxide
HPLC High pressure liquid chromatography
MPLC Medium pressure liquid chromatography
LC-MS liquid chromatography - coupled mass
spectroscopy
RT retention time
MP melting point
NMR nuclear resonance spectroscopy
TLC thin layer chromatography
ES electrospray
DMAC N,N-dimethylacetamide
HRMS high resolution mass spectroscopy
GDI 1,1'-carbonyldiimidazole
HOBT 1-hydroxybenzotriazole
DCC 1,3-dicyclohexylcarbodiimide
EDCI 1-[3-(dimethy!amino) propyl]-3-ethylcarbodnmide
hydrochloride
DMAP 4-dimethylaminopyridine
TMSCI Trimethylsilyl chloride
m-CPBA 3-chloroperbenzoic acid
HEPES N-(2-hydroxyethyl)-piperazine-N'-(2-ethane
sulphonic acid)
Tris/hydrochloric acid tris(hydroxymethyl)-aminomethane
hydrochloride
™Triton X-100® tert.-octyl-phenoxypo!yethoxyethanol, Rohm &
Haas, USA
The yield percentages of the following examples refer to the starting component which was used in the lowest molar amount.
LC-MS Methods

LC-MS (Method 1):
MS equipment: Micromass Quattro LCZ
ionization mode: ESI positive / negative

HPLC equipment: HP 1100
UV detection: temperature:

40 °C

208-400 nm



Column: Supplier: Gradient:

™ Symmetry C 18
50 mm x 2.1 mm 3.5 µm
Waters
Time A: % B: % Flow
[min.] [mL/min.]
0.00 90.0 10.0 0.50
4.00 10.0 90.0 0.50
6.00 10.0 90.0 0.50

A: 0,05% strength solution of formic acid in water B: 0.05% strength formic acid in acetonitrile
LC-MS (Method 2):
MS equipment: Micromass LCZ
ionization mode: ESI

HPLC equipment: Gilson 215 UV detection:

254 nm

Column: Supplier: Gradient


120A
YMC pro C-18 23 mm x 2 mm YMC
Time A: % B: % Flow
[min.] [mL/min.]
0.50 90,0 10.0 1.0
3.50 5.0 -95.0 1.0
4.00 5.0 95.0 1.0
4.01 90.0 10.0 1.0
4.80 90.0 10.0 1.0

A: . 0.02 % strength solution of trifluoroacetic acid in 2 % acetonitrile / 98 % water
B: 0.02 % strength solution of trifluoroacetic acid in 98 % acetonitrile / 2 % water
HPLC (Method 3):

HPLC Equipment: Gilson 215
UV Detection: Temperature:

220 and 254 nM 25 °C



Column:
Supplier: Gradient:

YMC-Pack Pro C18
50 mm x 4.6 mm 5Qm
Waters
Time A: % B: % Flow
[min.] [mL/min]
0.00 10.0 90.0 4.00


3.50 90.0 10.0 4.00
4.50 90.0 10.0 4.00
4.60 10.0 90.0 4.00
5.00 10.0 90.0 4.00
A: 0.1 % strength solution of TFA in acetonitrile B: 0.1 % strength aqueous TFA
HPLC (Method 4):

HPLC Equipment: Gilson 215
UV Detection: Temperature:

220 and 254 nM 25 °C



Column:
Supplier: Gradient:

YMC-Pack Pro C18
75 mm x 30 mm 5|4.m
Waters
Time A: % B: % Flow
[min.] [mL/min]
0.00 20.0 80.0 25,00
20.00 80.0 20.0 25.00

A: acetonitrile • B: 0.1% strength aqueous TFA
Preparation of Starting Materials and Intermediates

General Method A: Preparations of Aminophenols
Aminophenols are either commercially available or may be prepared as described in one or more of the Examples below.
Method A-1 Preparation of 5-Nitroindazole-1-carboxvlic acid terf-butvl ester

(Figure Removed)


Step 1: Preparation of 5-Nitroindazole-1-carboxylic acid ferf-butyl ester

(Figure Removed)
To a 0 °C slurry of 5-nitroindazole (5 g, 30.6 mmol), Et3N (4.7 mL, 33.7 mrnol) and 4-dimethylaminopyridine (0.75 g, 6.1 mmol) in acetonitrile (60 mL) was added dropwise to a solution of di-tert-butyl dicarbonate (8 g, 36.8 mmol) in acetonitrile (40 mL). The resulting mixture was stirred for 30 min, then concentrated under reduced pressure. The residue was dissolved in Et20 (200 mL) and H2O (100 mL). The pH of the aqueous, layer was adjusted to 2 using a 1N HCI solution. The organic phase was separated, dried (Na2SO4) and concentrated under reduced pressure to give 5-nitroindazole-1-carboxylic acid tert-butyl ester (7.8 g, 96%) as a yellow solid: TLC (30% EtOAc/hex), Rf = 0.70; ES-LCMS (rel abundance) m/z 264 (MH+, 100%).
Step 2: Preparation of the title compounds 5-Aminoindazole-1-carboxvlic acid tert-butyl ester

Palladium on carbon (780 mg) was placed under an inert atmosphere and suspended in EtOH (15 mL). A solution of 5-nitroindazole-1-carboxylic acid tert-butyl ester (7.78 g, 29.5) in EtOH (100 ml) and EtOAc (100 ml) was added. The reaction mixture was placed under H2 atmosphere (1 Atm pressure) and stirred overnight. The resulting mixture was filtered through a pad of Celite®. The filtrate was concentrated under reduced pressure to yield a greenish foamy solid. The. crude product was dissolved in CH2Cl2 and purified by Biotage Flash 40M (gradient from 30% to 50% EtOAc/hex) to give the title compound (6.55 g, 95%) as a white solid: TLC (50%EtOAc/hex), Rf = 0.41; ES-LCMS (rel abundance) m/z 234 (MH+, 66%).
A. Methods for making aminophenols Method A. 1.
Preparation of 4-amino-3-metliylthiophenol Step 1. Preparation of 3-methylthio-4-nitrophenol
(Figure Removed)

To a mixture of 3-fluoro-4-nitro-phenol (3.00 g, 19.10 nunol) and sodium thiomethoxide (2.68 g, 38.19 mmol) in DMF (100 ml) was added K2CO3 (7.92, 57.3 mmol) and the mixture was allowed to stir for 12 hours, the next day all the sm had been consumed. Water (500 mL) was added then to the solution and the product was extracted using EtOAc (3x200 mL). The combined organic fractions were dried (Na2SO4 and concentrated under reduced pressure. The residue was purified using column chromatography (20% EtOAc/hex) to yield 3-methylthio-4-nitrophenol as a bright yellow solid (3.25 g, 92%): TLC Rf 0.34 (35% EtOAc/hex).
Step 2. Preparation of 4-amino-3-methylthiophenol
(Figure Removed)

Iron powder (754 mg, 13.5 mmol) was added slowly to a solution of 3-methylthio-4-nitrophenol (ZZZ) in acetic acid (25 mL), The mixture was stirred at room temperature overnight to afford a white precipitate. The remaining solid iron was removed with a magnet, and the slurry was filtered through filter paper. The filtrate was diluted with HaO (100 mL), neutralized with a saturated Na2CO3 solution, then extracted with CH2Cl2 (3xl00mL). The combined organic fractions were dried (Na2SO4) and concentrated under reduced pressure to give 4-amino-3-methylthiophenol as a purple/brown solid (260 mg, 62%): TLC R/0.33 (35% EtOAc/hex).
B Methods for making B-extended anilines
Method B.I. Preparation of 4-(4-Aminophenoxy)pyridine-2-carbonitrile
(Figure Removed)

A solution of 4-aminophenol (1.0 g, 9.16 mmol) in anhydrous DMF (9.2 mL) was treated with potassium te7t-butoxide (1.08 g, 9.62 inmol, 1.05 eq), and the orange-brown reaction mixture was stirred at RT for Ih. The contents were treated with 2-cyano-4-chloropyridine (1.27 g, 9.16 mmol, 1.0 eq) and K2CO3 (497 mg, 5.04 mmol, 0.55 eq) and then heated at 90 °C for 17h. The mixture was cooled to RT and partitioned between BtOAc (250 mL) and a saturated NaCl solution (100 rnL). The aqueous phase was back-extracted with EtOAc' (300 mL), The combined organic layers were washed with brine, dried over Na2SO4, and concentrated under reduced pressure. Purification on MPLC (Biotage; 30% EtOAc / hex) afforded 1.83 g (94.6%) of 4-(4-aniinophenoxy)pyridine-2-carbonitrile as a yellow solid: TLC (50% EtOAc/hex), R/ = 0.28; 'H-NMR (DMSO d6) 6 8.52 (d, J = 6.3 Hz, IH), 7.54 (d, J = 2.4 Hz, IH), 7.07 (dd, J Method B.2. Preparation of 4-(4-Amino-3-methylphenoxy)pyridine-2-carbonitrile
(Figure Removed)

The title compound was prepared in the same manner described for 4-(4-ammophenoxy)pyiidine-2-carbonitrile, substituting 4-amino-3-fluorophenol for 4-aminophenol.
H-NMR (DMSO-D6) 5 8.54 (d, J = 6.0 HZ, IH), 7.61 (d,J = 2.7 HZ, IH), 7.12 (dd, J = 5.7,2.7
Hz, IH), 7.02 (dd, J = 11.7,2.1 Hz, IH), 6.86 to 6.78 (m, 2H), 5.25 (s, 2H).
Method B.3. Preparation of 4-(4-Amino-3-trifluoromethylphenoxy)pyridine-2-carbonitrile

(Figure Removed)
The title, compound was prepared in the same manner described for 4-(4-animophenoxy)pyridine-2-carboriitrile, substituting 4-amino-3-trifIuoromethylphenol for 4-aniinophenol. 'H-NMR (DMSO-d6) 8 8.54 (d, J = 6 Hz, IH), 7.64 (d, J = 2.7 Hz, IH), 7.22 to 7.17 (m, 2H), 7.12 (dd, J - 5.7,2.4 Hz, IH), 6'.92 (d, J = 8.7 Hz, IH), 5.74 (s, 2H).
Method B.4. Preparation of 4-(4-Amino-3-methylphenoxy)pyridine-2-carbonitrile
(Figure Removed)
The title compound was prepared in the same manner described for 4-(4-aminophenoxy)pyiidine-2-carbouitrile, substituting 4-amino-3-methylphenol for 4-aminophenol: ES-LCMS (rei abundance) iti/z 226 (M+H)+.
Method B.5. Preparation of 4-(4-Amino-2-methylplienoxy)pyridine-2-carbonitiile

(Figure Removed)


The title compound was prepared in the same manner described for 4-(4-aminophenoxy)pyridine-2-carbonitrile, substituting 4-artiino-2-m.ethylpb.enol for 4-aminophenol: ES-LCMS (rel abundance) m/z 226 (M+H)+.
Method B.6. Preparation of 4-(4-Amino-3-nitrophenoxy)pyridine-2-carbonitrile


(Figure Removed)
The title compound was prepared in the same manner described for 4-(4-aminophenoxy)p3'ridine-2-carbonitrile) substituting 4-amino-3-nitrophenol for 4-aminophenol: ES-LCMS (rel abundance) m/z 257 (M+H)+.
Method B.7. Preparation of 4-(4-amino-2-fluorophenoxy)pyridine-2-carbonitriIe
(Figure Removed)
A solution of potassium tert-butoxide (1.83 g, 16.36 mmol) in DMA (5 ml) was degassed purge) and cooled to 0 °C. Separately, DMA (5 mL) was added to 4-amino-2-fluorophenol was
added and the resulting mixture degassed (N2 purge) and stirred until all the solid had dissolved, then was added slowly by cannula to the potassium tert-butoxide at 0 °C. To the resulting dark mixture was added dropwise a solution of 4-chloropyridine-2-carbonitrile in DMA (5 mL). The solution was stirred for 1 hr at 0 °C and then heated to 80 °C overnight. Water was added to the resulting mixture, and the aqueous mixture was concentrated under reduced pressure. The residue was treated with a NaOH solution (1:1:1 IN NaOH/water/saturated NaCl solution; 200 mL), then extracted with EtOAc (3X100 mL). The combined organic fractions were dried (Na2SO4) and concentrated reduced pressure. The residue was purified using column chromatography (40% EtOAc/hex) to yield 4-(4-amino-2-fluorophenoxy)pyridine-2-carbonitiile (2.31 g; 64%); TLC R/0.83 (100% EtOAc); 'HNMR (DMSO-d6) 5 5.51 (s, 2H), 6.38/d, J=3 Hz, 1H), 6.50 (dd, J = 3,14 Hz, 1H), 6.98 (app t, J - 9 Hz, 1H), 7.10 (dd, 3,6 Hz, 1 H), 7.63 (d, J = 3 Hz, 1 H), 8.53 (d, J = 6 Hz, 1 H); ES-LCMS (rel abundance) m/z 230 (M+H)+.
Method B .8. Preparation of 4-Aimno-3,5-difluoTo-phenol
Step 1: Preparation of 5-Benzyloxy-1,3-difluono-2-nitrobenzene
(Figure Removed)

A mixture of l,3,5-trifluoro-2-nitrobenzene (6.1 g, 34 mmol), benzyl alcohol (3.7g, 34 mmol) and potassium carbonate (7.1g, 52 mmol) in DMF (10 ml) was stirred at room temp overnight. The reaction mixture was added to water (30 ml) and put into refrigerator for several hours. The resulting yellow precipitate was removed, washed with water, and dried under reduced pressure, providing (6.5 g, 71%) of the 5-benzyloxy-l,3-difluoro-2-nitroberizene and l-benzyloxy-3,5-difluoro-2-nitrobenzene as approximate 1:1 mixture. The mixture was used directly for the next step without further purification: TLC Rf = 0.48 (10% ethyl acetate - hexane); 1H-NMR (CD2C12) 5 7.46-7.36 (m, 5H), 6.75-6.60 (m, 2H), 5.19 (s, 1H), 5.11 (s, 1H).
Step 2. Preparation of 4-Amino-3,5-difluorophenol
(Figure Removed)


A solution of compounds (6.4 g, 24 mmol) obtained from step 1 in methanol (250 ml) was added to a flask with palladium on carbon (10 % wt, 720 mg) under nitrogen atmosphere. The mixture was stirred at room temp, under hydrogen atmosphere overnight. The hydrogen was introduced by 1 Gallon balloon. Upon completion, the reaction mixture was nitrated through a pad of Celite under nitrogen atmosphere and dried under reduced pressure, providing (3.4 g, 97%) of a mixture of 4-amino-3,5-difluorophenol and 2-amino-3,5-difluoro-phenol as a mixture. The mixture was used directly for the next step without further purification, 1H-NMR (DMSO) 8 9.50 (bs, 1H), 6.54-6.22 (m, 2H), 4.36 (s, 2H); ES-LCMS (rel abundance) m/z 146 (M+H)+
Step 3. Preparation of 4-(4-Amino-3,5-difluoro-phenoxy)-pyridine-2-carbonitrile
(Figure Removed)

A solution of compounds (1.5 g, 10 mmol) obtained from step 2 in dry DMF (12 mi) was treated with potassium tert-butoxide (1.2 g, 11 mmol), and the mixture was stirred at room temp, for 2h. The contents were treated with 4-chloropyridine-2-carbonitrile (1,4 g, 10 mmol) in dry DMF (S ml) and potassium carbonate (0.76 g, 5.5 mmol) and then heated to 80 °C under nitrogen atmosphere for 60 h. The mixture was cooled to room temp, and evaporated under vacuum. The residue was purified by silica column (gradient from 10% EtOAc/hex to 20% EtOAc/hex) to afford 0.55 g (22 %) of the title compound. 'H-NMR (DMSO) 5 8.51 (d, J = 5.7 Hz, 1H),'7.19 (d, J = 2.5 Hz, 1H), 7.00 (dd, J « 5.7 Hz, J = 2.4 Hz, 1H), ), 6.64 (dd, J - 6.7 Hz, J = 1.2 Hz, 2H), 3.57 (s, 2H); ES-LCMS (rel abundance) m/z 248 (M+H)+.
C. Methods for making ureas
Method C.I. GDI-mediated urea formation. Synthesis of N-[4-(2-cyano(4-pyridyloxy))-2~ fluorophenyl] {[2-(trifluoroniethyl)(4-pyridyl)] amino } carboxamide

(Figure Removed)
Step 1: To a slurry of CDI (300 mg, 1.85 mmol) in CH2C12 (0.5 mL) was added a solution of 2-(trifiuoTornethyl)-4-pvridylamme (300 mg, 1.85 mmol) in CH2C12 (1 mL). The reaction mixture was stirred for 20 h. The resulting pale yellow solution was diluted to a total volume of 10 rnL and used without further purification.
Step 2: To a solution of CDI-2-(trifluoromethyl)-4-pyiidylamine in CH2C12 ( 1 mL, 0.19 mmol) was added 4-(4-amino-3-fluorophenoxy)pvridine-2-carbonitrile (42 mg, 0.19 mmol). The reaction mixture was stirred for 1 day. The resulting mixture was purified using a Biotage Flash 12M (Si02; 80% EtOAc/hex) to obtain N-[4-(2-cyano(4-pyridyloxy))-2-fluorophenyl]{[2-(trifluoromethyl)(4-pyridyl)]amino}carboxamide as a white solid (58 mg, 75%): mp 192-193; TLC (50% EtOAc/hex) R/0.33; 'H NMR (DMSO-d6) 7.08-7.12 (m, 1H), 7.25 (dd, J~ 2.7, 5.9 Hz, 1H), 7.37 (dd, /= 2.7,11.5 Hz, 1H), 7.55 (dd, J= 2.1, 5.5 Hz, 1H), 7.74 (d, J= 2.5 Hz), 8.06 (d, J= 1.9 Hz, 1H), 8.15 (t, J= 9.0 Hz, 1H), 8.54 (d, J= 5.6 Hz, 1H), 8.60 (d, J~ 5.6 Hz, 1H), 8.95 (d, /= 1.5 Hz, 1H), 9.85 s, 1H); ES-LCMS (rel abundance) m/z 418 ((M+H)+, 100%).
Method C.2. Isocyanate coupling. Preparation of A'"-[4-(2-Cyano(4-pyridyloxy))-2-fluorophenyl][(2.,2,4>4-tetraf;iprpbnemzp]l,3'dioxin-6-yl)amino]carboxamide
(Figure Removed)
2,2,4,4-Tetrafluorobenzo[e]l,3-dioxan-6-isocyanate (5.47 g, 21.9 mmol) was added as a solid to a solution of 4-(4-amino-3-metliylphenoxy)pyiidine-2-carbonitrile (5.03 g, 21.9 mmol) in 30 mL CH2Cl2, and the mixture left to stir at room temp. After 15 min a precipitate began to form, soon becoming thick enough to make stirring difficult. After 1 h the solid was removed and washed with 50 % hexanes in CH2Cl2 . The crude product was dissolved EtOAc:heptane and
allowed to crystallize slowly by cooling in an insulated container overnight The resulting crystals were ground in a mortar and pestle and placed under vacuum (0.1mm, 100 C)'to yield 5.47g (52%) as white crystals: mp. =174 °C; 'H-NMR (DMSO-d6) 6 9.43 (s, IH), 8.76 (d, J = 2.5 Hz, IH), 8.59 (dd, J = 5.8 Hz, IH), 8.17 (dd, J = 9.1 Hz, IH), 8.12 (d, J = 2.3 Hz, IH), 7.71 (dd, J = 2.5 Hz, 0.5 Hz), 7.60 ((dd, J = 9 Hz, 2.5 Hz, 1 H), 7.42 (d, J = 9Hz, IH), 7.33 (dd, J = 11.5 Hz, 2.5 Hz, IH), 7.23 (dd, J = 5.8 Hz, IH), 7.06 (ddd, J-l .3, 2.5, 9 Hz, IH); ES-LCMS (rel abundance) m/z 479 (M+H)+.
D. Synthetic modifications of ureas.
Method D.I. Synthesis of 2-cyanopyridine N-oxides. Preparation of N-[4-(2-cyano-l-
hydroxy(4-pyridyloxy))prienyl][(2,2,4,4-tetrafluorobenzo[3,4-e]l,3-dioxin-6-
yl)amino] carboxamide
(Figure Removed)

To a solution of N-[4-(2-cyano(4-pyridyloxy))phenyl][(252,4,4-tetrafluorobenzo[3,4-e]l,3-dioxin-6-yl)amino]carboxamide (150 mg, 0.33 inmol) in CH2Cl2 (5 mL) and THF (5 mL) was added mCPBA (50% pure, 450 mg, 1.3 mmol). The reaction mixture was stirred for 2 days. The resulting mixture was purified by Biotage Flash 12M (EtOAc) to give N-[4-(2-cyano-l-liydroxy(4-pyridyloxy))pheayl][(2,2,4,4-tetrafluorobenzo[3,4-e]l,3-dioxm-6-yl)amino]carboxarmde as a white solid (33 mg, 21%); TLC (EtOAe) R/-0.30; ]H NMR (DMSO-d6) 7.11-7.16 (m, 2H), 7.24 (dd, J= 3.6, 7.5 Hz, IH), 7.42 (d, J= 9.2 Hz, IH), 7.52, 7.57 (m, 2H), 7.63-7.68 (m, IH), 7.78 (dd, J= 0.2, 3.4 Hz, IH), 8.09 (d, /= 2.5 Hz, IH), 8.33 (d, J= 1.5, 7.3 Hz, IH), 8.99 (s, IH), 9.12 (s, IH); ES-LCMS (rel abundance) m/z 477 ((M+H)+, 100%).
Method D.2. N-Oxide to Cyanopyridine Transformation. Preparation of N-[4-(2-Cyano(4~ pyridyloxy))-2-fluorophenyl] [(2,2,4,4-tetrafluorobenzo[3,4-e] 1,3-dioxin-6-yl)amino] carboxamide
(Figure Removed)

A suspension of N-[2-fluoro-4-(l-oxy(4-pyridyloxy))phenyl][(2,2,4,4-tetrafluorobenzo[3,4-e]l,3-dioxin-6-yl)amino]carboxamide (150 mg, 0.32 mmol) in 2 mL CH2C12 was treated with trimethylsilylcyanide (50 j-iL, 0.35 mmoL). After 5 min stirring dimethylcarbamoyl chloride (30 µL, 0.32 mmol, 1.1 equiv.) was added, and the mixture left to stir at room temp. The reaction gradually became homogeneous, but proceeded very slowly at this concentration. After 48 -lir, the mixture was diluted with CH2C12 (10 mL) and 10% K2CO3 (10 mL) and left to stir for 10 min. The resulting aqueous layer was washed with CH2C12 two times. The combined organic fractions were dried (Na2SO4) and concentrated in vacuo. Purification by flash column (Biotage) provided 102 mg (67%) of N-[4-(2-cyano(4-pyridyloxy))-2-fluorophenyl] [(2,2,4,4-tefrafiuorobenzo[3,4-e]l,3-dioxm-6-yl)amino]carboxamide as a white solid.
Method D.3. Conversion of Thiomethyl Analogues into Methylsulfonyl Analogues. Preparation ofN-[4-(2-Cyano(4-pyridyl)oxy)-2-(methylsulfonyl)phenyl][(2,2,4,4-tetrafluorobenzo[3,4-e]l,3-dioxin-6-yl)amino]carboxamide(Figure Removed)
To a solution of N-[4-(2-cyano(4-pyridyl)oxy)-2-methylthiophenyl][(2,2,4,4-tetrafluorobenzo[3,4-e]l,3-dioxin-6-yl)amino]carboxamide (97.0 mg, 0.19 mmol) in CH2Cl2 at 0 °C was added mCPBA (132.21 mg, 0.42 mmol). The resulting slurry mixture was stirred overnight. TLC indicated a mixture of sulfoxide and sulfone. Therefore, additional mCPBA (3 equiv.) was added and the reaction mixture was allowed to stir 4 h. The reaction mixture was treated with a saturated sodium Na2S2O4 solution (50 mL), then was extracted with EtOAc (3x50
mL). The combined organic phases were washed with a saturated NaCl solution (100 mL), dried (Na2SO4), and concentrated under reduced pressure to give N-[4-(2-cyano(4-p3iidyl)oxy)-2-(methylsulfonyl)phenyl][(2,2,4)4-tetrafluorobenzo[3,4-e]l,3-dioxin-6-yI)amino]carboxaniide: TLC R/0.20 (40% EtOAc/hex); ES-LCMS (rel abundance) m/z 539 ((M+H)+).
Additional compounds illustrated in Table 1 were prepared as described above by choosing the appropriate starting materials that are readily available and/or the synthesis of which is taught herein, and using the processes of Method E described above or other standard chemical processes known in the art.
Table 1: Examples of preferred compounds of formula (I)
(Table Removed 1)
Table 2: Characterization of preferred compounds of formula (I)
(Table Removed 2)

*The following are the LCMS conditions: HPLC - etectrospray mass spectra (HPLC ES-MS) were obtained using a Gilson HPLC system equipped with two Gilson 306 pumps, a Gifson 215 Autosampler, a Gilson diode array detector, a YMC Pro C-18 column (2 x 23mm, 120 A), and a Micromass LCZ single quadrupole mass spectrometer with z-spray eiectrospray ionization. Spectra were scanned from 120-1000 arnu over 2 seconds. ELSD (Evaporative Light Scattering Detector) data was also acquired as an analog channel. Gradient elution was used with Buffer A as 2% acetonitrile in water with 0.02% TFA and Buffer B as 2% water in Acetonitrile with 0.02% TFA at 1.5 mL/min. Samples were eluted as follows: 90% A for 0,5 min ramped to 95% B over 3.5 min and held at 95% B for 0.5 min and then the column is brought back to initial conditions over 0.1 min. Total run time is 4.8 min.
**comm refers to commercially available materials.
Other compounds of Formula (I) may be prepared using the methods described herein or other methods known in the art, and using the appropriate starting materials and/or intermediates that would be readily recognized by those skilled in the art.
Biological Tests
fn-vitro p38 kinase assay No. 1
Purified and His-tagged p38 α2 (expressed in E. Coll) was activate in vitro by MMK-6 to a high specific activity. Using a microtiter format, all reac. is were conducted in 100 µL volumes with reagents diluted to yieid 0.05 pg/weli of activated p38 α2 and 10 µg/well of myelin basic protein in assay buffer (25 mM HEPES 7.4, 20 mM MgCI2, 150 mM NaCf). Test compounds (5 µL of a 10% DMSO solution in water) were
prepared and diluted into the assay to cover a final concentration range from 5 nM to 2.5 µM. The kinase assay was initiated by addition of 25 pi. of an ATP cocktail to give a final concentration of 10 µM cold ATP and 0.2 µCi [y-33P] ATP per well (200-400 dpm/pmol of ATP). The plate was incubated at 32 °C for 35 min., and the reaction quenched with 7 µL of a 1 N aq HC! solution, The samples were harvested onto a P30 Filtermat (Wallac, Inc.) using a TomTec 1295 Harvester (Wallac, Inc.), and counted in a LKB 1205 Betaplate liquid Scintillation Counter (Wallac, Inc.). Negative controls included substrate plus ATP alone. SW1353 cellular assay; SW1353 cells (human chondro-sarcoma) are seeded (1000 cells/100 µL DMEM 10% FCS/well) into 96-well plates and incubated overnight. After medium replacement, cells are exposed to test compounds for 1 h at 37 °C, at which time human lL-1 (1 ng/mL, Endogen, Woburn, WA) and recombinant human TNFα (10 ng/mL) are added. Cultures are incubated for 48 h at 37 °C, then supernatant IL-6 values are determined by ELISA. The compound of this invention shows significant inhibition of p38 kinase.
Murine VEGFR-2 Biochemical Assay
This assay was performed in 96-well opaque plates (Costar 3915) in the TR-FRET
format. Reaction conditions are as follows: 10 µM ATP , 25 nM poly GT-biotin , 2 nM
Eu-labelled phospho-Tyr Ab, 10 nM APC, 7 nM Flk-1 (kinase domain), 1% DMSO, 50
mM HEPES pH 7.5, 10 mM MgCI2, 0.1 mM EDTA, 0.015% BRIJ, 0.1 mg/mL BSA, 0.1%
mercapto-ethanol). Reaction is initiated upon addition of enzyme. Final reaction
volume in each well is 100 µL. Plates are read at both 615 and 665 nM on a Perkin
Elmer Victor V Multilabel counter at about 1.5- 2.0 hours after reaction initiation. Signal
is calculated as a ratio: (665 nm / 615 nm) * 10000 for each well.
The compound of this invention shows significant inhibition of VEGFR2 kinase.
Murine PDGFR FRET biochemical assay
This assay was formatted in a 96-well black plate (Costar 3915). The following reagents are used: Europium-labeled anti-phosphotyrosine antibody pY20 (Perand streptavidin-APC; poly GT-biotin from, and mouse PDGFR. The reaction conditions are as follows: 1
nM mouse PDGFR is combined with 20 pM ATP, 7nM poly GT-biotin, 1 nM pY20 antibody, 5 nM streptavidin-APC, and 1% DMSO in assay buffer (50 mM HEPES pH 7.5, 10 mM MgCI2, 0.1 mM EDTA, 0.015% BR1J 35, 0.1 mg/mL BSA, 0.1% mercaptoethanol). Reaction is initiated upon addition of enzyme. Final reaction volume in each well is 100 µL. After 90 minutes, the reaction is stopped by addition of 10 pi/well of 5 p,M staurosporine. Plates are read at both 615 and 665 nm on a Perkin Elmer VictorV Multilabel counter at about 1 hour after the reaction is stopped. Signal is calculated as a ratio: (665 nm / 615 nm) * 10000 for each well. The compound of this invention shows significant inhibition of PDGFR kinase.
For IC50 generation for both PDGFR and Flk-1, compounds were added prior to the enzyme initiation. A 50-fold stock plate was made with compounds serially diluted 1:3 in a 50% DMSO/50% dH2O solution. A 2 µL. addition of the stock to the assay gave final compound concentrations ranging from 10 µM - 4.56 nM in 1% DMSO. The data were expressed as percent inhibition: % inhibition = 100-((Signal with inhibitor-background)/(Signal without inhibitor - background)) * 100
pPDGFR-b sandwich ELISA in AoSMC cells
100K P3-P6 Aortic SMC were plated in each well of 12-well cluster in 1000 pi. volume/ well of SGM-2 using standard cell culture techniques. Next day, cells were rinsed with 1000 pL D-PBS once, then serum starved in 500 pL SBM (smooth muscle cell basal media) with 0,1% BSA overnight. Compounds were diluted at a dose range from (10 \M to 1 nM in 10-fold dilution steps in DMSO, final DMSO concentration 0.1%). Remove old media by inversion into the sink quickiy then add 100 µl of each dilution to corresponding well of cells for 1 hr at 37 °C. Cells were then stimulated with 10 ng/mL PDGF BB ligand for 7 minutes at 37 °C. The media is decanted and 150 pL of isotonic lysis buffer with protease inhibitor tablet (Complete; EDTA-free) and 0.2 mM Na vanadate is added. Cells are lysed for 15 min at 4 °C on shaker in cold room. Lysates are put in eppendorf tubes to which 15 µL. of agarose-conjugated anti-PDGFR-b
antibody is added and incubated at 4 °C overnight. Next day, beads are rinsed in 50-volumes of PBS three times and boiled in 1x IDS sample buffer for 5 minutes. Samples were run on 3-8% gradient Tris-Acetate gels and transferred onto Nitrocellulose. Membranes were blocked in 1% BSA/TBS-T for 1 hr. before incubation in anti-phospho-PDGFR-b (Tyr-857) antibody in blocking buffer (1:1000 dilution) for 1 hour. After three washes in TBS-T, membranes were incubated in Goat anti-rabbit HRP IgG (1:25000 dilution) for 1 hr. Three more washes followed before addition of ECL substrate. Membranes were exposed to Hyperfilm-ECL. Subsequently, membranes were stripped and reprobed with anti-PDGFR-b antibody for total PDGFR-b.
c-Raf (Raf-1.) Biochemical Assay
Purification of proteins used in the assay
The c-Raf biochemical assay was performed with a c-Raf enzyme that was activated (phosphorylated) by Lck kinase. Lck-activated c-Raf (Lck/c-Raf) was produced in Sf9 insect cells by co-infecting cells with baculoviruses expressing, under the control of the polyhedrin promoter, GST-c-Raf (from amino acid 302 to amino acid 648) and Lck (full-length). Both baculoviruses were used at the multiplicity of infection of 2.5 and the cells were harvested 48 hours post infection.
MEK-1 protein was produced in Sf9 insect cells by infecting cells with the baculovirus expressing GST-MEK-1 (full-length) fusion protein at the multiplicity of infection of 5 and harvesting the cells 48 hours post infection. Similar purification procedure was used for GST-c-Raf 302-648 and GST-MEK-1.
Transfected cells were suspended at 100 mg of wet cell biomass per ml in a buffer containing 10 mM sodium phosphate, 140 mM sodium chloride pH 7.3, 0.5% Triton X-100 and the protease inhibitor cocktail. The cells were disrupted with Poiytron
homogenizer and centrifuged 30,000g for 30 minutes, The 30,000g supernatant was applied onto GSH-Sepharose. The resin was washed with a buffer containing 50 mM Tris, pH 8,0, 150 mM NaCI and 0.01% Triton X-100. The GST-tagged proteins were eluted with a solution containing 100 mM Glutathione, 50 mM Tris, pH 8.0, 150 mM NaCI and 0.01% Triton X-100. The purified proteins were dialyzed' into a buffer containing 20 mM Tris, pH 7.5,150 mM NaCI and 20% Glycerol.
BiochemicaLassay protocol and results,
The compounds were serially diluted in DMSO using three-fold dilutions to stock concentrations ranging typically from 50 µM to 20 nM (final concentrations in the assay range from 1 µM to 0.4 nM). The c-Raf biochemical assay was performed as a radioactive filtermat assay in 96-well Costar polypropylene plates (Costar 3365). The plates were loaded with 75 µL solution containing 50 mM HEPES pH 7.5, 70 mM NaCI, 80 ng of Lck/c-Raf and 1 µg MEK-1. Subsequently, 2 µL of the serially diluted individual compounds were added to the reaction, prior to the addition of ATP. The reaction was initiated with 25 (xL ATP solution containing 5 µM ATP and 0.3 µCi [33P]-ATP. The plates were sealed and incubated at 32 °C for 1 hour. The reaction was quenched with the addition of 50 µl of 4 % Phosphoric Acid and harvested onto P30 filtermats (PerkinElmer) using a Wallac Tomtec Harvester. Filtermats were washed with 1 % Phosphoric Acid first and deinonized H2O second. The filters were dried in a microwave, soaked in scintillation fluid and read in a Wallac 1205 Betaplate Counter (Wallac Inc., Atlanta, GA, U.S.A.). The results were expressed as percent inhibition.
% Inhibition = [100-(Tib/TO] x 100 where.
Tib - (counts per minute with inhibitor)-(background) TI = (counts per minute without inhibitor)-(background)
Assay results
The biological inhibition activity of the compounds of the present invention was tested in various inhibition assays such as those described above. The compounds exhibited a range of inhibitory activity described below:
a) h-FIt4 v2 assay IC50 (nM) values for the preferred compounds of the present
invention range from 0.66 to 3000,
b) m-Flt4 assay IC50 (nM) values for the preferred compounds of the present
invention range from 114 to > 10,000,
c) Flk1 FRET assay IC50 (nM) values for the preferred compounds of the present
invention range from 6.97 to 186,
d) c-RAF-1 assay lC50 (nM) values for the preferred compounds of the
present invention range from 7,86 to >1600,
e) cRaf v2 assay IC50 (nM) values for the preferred compounds of the present
invention range from 7.9 to 1000.
Overall, compounds of the present invention provide a unique combination of angiogenesis and tumor cell proliferation, through an improved inhibition profile against several key kinase targets such as raf, p38, PDGFR, VEGFR3 and VEGFR2, which are all molecular targets of interest for the treatment of diseases, including cancer.
The preceding examples can be repeated with similar success by substituting the generically or specifically described reactants and/or operating conditions of this invention for those used in the preceding examples. From the foregoing description, one skilled in the art can easily ascertain the essential characteristics of this invention and, without departing from the spirit and scope thereof, can make various changes and modifications of the invention to adapt it to various usages and conditions.





WE CLAIM
1. A compound of Formula (I)
(Formula Removed)

or a pharmaceutically acceptable acid salt or basic salt thereof,
wherein A is unsubstituted or substituted 2H,3H-benzo[e]l,4-dioxan-6-yl, 2H,4H-benzo[e]l,3-dioxan-6-yl, or 2H,4H-benzo[e]l,3-dioxan-8-yl wherein the substituents of the substituted 2H,3H-benzo[e]l,4-dioxan-6-yl, 2H,4H-benzo[e]l,3-dioxan-6-yl, or 2H,4H-benzo[e]l,3-dioxan-8-yl are substituted by (R3)n wherein n is an integer 0, 1,2, 3, 4, 5, or 6 and
each R3 is independently
halogen,
R4
OR4,
S(O)R4,
C(O)R4,
C(O)NR4R5,
oxo.

cyano or
NO2; and
R4 and R5 are independently hydrogen, C1-6alkyl, or up to per-halogenated C1-6alkyl,
B is selected from the group consisting of
(Formula Removed)




p is an integer 0, 1,2, 3, or 4, each R1 is independently
halogen,
C1-5haloalkyl,
NO2;
C(O)NR4R5
C1-6alkyl,
C1-6dialkylamine,
C1 -3alkylamine,
CN,

amino,
hydroxyl or C1-3alkoxy
SCH3,
methylsulfonyl,
R4 and R5 are independently hydrogen,C1-6alkyl, or up to per-halogenated C1-6alkyl,
L is O or S,
m is an integer 0, 1,2, or 3, and
each R2 is independently C1-5 alkyl, C1-5haloalkyl, C1-3alkoxy, N-oxo or N-hydroxy.
2, A compound as claimed in claim 1, wherein L is oxygen.
3, A compound as claimed in claim 1, wherein R1 is fluorine, chlorine, bromine, methyl, NO2, C(=O)NH2, methoxy, or trifluoromethyl.

4. A compound as claimed in claim 1, wherein R2 is methyl, ethyl, propyl, or oxygen.
5. A compound as claimed in claim 1, wherein R3 is trifluoromethyl, methyl, ethyl, propyl, butyl, isopropyl, tert-butyl, chlorine, fluorine, bromine, cyano, methoxy, acetyl, trifluromethyl sulfonyl, trifuoromethoxy, or trifluoromethylthio.

6. A compound as claimed in claim 1 wherein each R3 substituent is fluorine.





Documents:

3802-DELNP-2005-Abstract-(13-03-2009).pdf

3802-DELNP-2005-Abstract-(27-03-2009).pdf

3802-delnp-2005-abstract.pdf

3802-DELNP-2005-Claims-(13-03-2009).pdf

3802-DELNP-2005-Claims-(27-03-2009).pdf

3802-delnp-2005-claims.pdf

3802-delnp-2005-complete specification (granted).pdf

3802-delnp-2005-Correspondence Others-(15-03-2011).pdf

3802-DELNP-2005-Correspondence-Others-(13-03-2009).pdf

3802-delnp-2005-correspondence.pdf

3802-DELNP-2005-Description (Complete)-(13-03-2009).pdf

3802-delnp-2005-description (complete)-(27-03-2009).pdf

3802-delnp-2005-description (complete).pdf

3802-DELNP-2005-Form-1-(13-03-2009).pdf

3802-delnp-2005-form-1.pdf

3802-delnp-2005-form-18.pdf

3802-DELNP-2005-Form-2-(13-03-2009).pdf

3802-delnp-2005-form-2.pdf

3802-DELNP-2005-Form-3-(13-03-2009).pdf

3802-delnp-2005-form-3.pdf

3802-delnp-2005-form-5.pdf

3802-delnp-2005-From-15-(15-03-2011).pdf

3802-DELNP-2005-GPA-(13-03-2009).pdf

3802-delnp-2005-GPA-(15-03-2011).pdf

3802-delnp-2005-gpa.pdf

3802-delnp-2005-pct-101.pdf

3802-delnp-2005-pct-210.pdf

3802-delnp-2005-pct-237.pdf

3802-delnp-2005-pct-304.pdf

3802-DELNP-2005-Petition-137-(13-03-2009).pdf

3802-DELNP-2005-Petition-138-(13-03-2009).pdf

abstract.jpg


Patent Number 233627
Indian Patent Application Number 3802/DELNP/2005
PG Journal Number 18/2009
Publication Date 01-May-2009
Grant Date 31-Mar-2009
Date of Filing 26-Aug-2005
Name of Patentee BAYER PHARMACEUTICALS CORPORATION
Applicant Address 400 MORGAN LANE, WEST HAVEN, CT 06516-4175, USA.
Inventors:
# Inventor's Name Inventor's Address
1 STEPHEN BOYER 233 COLONY STREET, FAIRFIELD, CT 06430,USA.
2 WENDY LEE 282 EVERGREEN AVENUE, HAMDEN,CT 06518, USA.
3 YUANWEI CHEN 15 BLUE RIDE LANE, NORTH HAVEN, CT 06473, USA.
4 BARTON PHILLIPS 498 WHITNEY AVENUE, APT. 3D, NEW HAVEN, CT 06511, USA.
5 SHARAD VERMA 29 HARBOUR CLOSE, NEW HAVEN,CT 06519, USA.
6 JIANQING CHEN 117 FREDERICK STREET, APT. 2-L, NEW HAVEN, CT 06515,USA.
7 ZHI CHEN 865 MIX AVENUE #409, HAMDEN, CT 06514, USA.
8 WILLIAM J. SCOTT 210 SADDLE HILL DRIVE, GUILDFORD, CT 06473, USA.
9 JACQUES DUMAS 98 FARMVIEW ROAD,BETHANY, CT 06524,USA.
10 JIANMEI FAN 121 OCTOBER HILL ROAD, HAMDEN, CT 06518, USA.
11 BRIAN RAUDENBUSH 45 DANIELS ROAD, CHARLTON, MA 01507, USA.
12 ANIKO REDMAN 66 E. STREET, DERBY, CT 06418, USA.
13 LIN YI 58 MAGNOLIA ROAD, MILFORD, CT 06460, USA.
14 QINGMING ZHU 11 BASSETT STREET, APT. D4, WEST HAVEN, CT 06516, USA.
PCT International Classification Number C07D 401/12
PCT International Application Number PCT/US2004/006286
PCT International Filing date 2004-03-01
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 60/350,348 2003-02-28 U.S.A.
2 60/450,323 2003-02-28 U.S.A.
3 60/450,324 2003-02-28 U.S.A.