WO2008150260A1 - 8-oxy-2-aminopyrido (2, 3-d) pyrimidin-7-one derivatives as kinase inhibitors and anticancer agents - Google Patents

8-oxy-2-aminopyrido (2, 3-d) pyrimidin-7-one derivatives as kinase inhibitors and anticancer agents Download PDF

Info

Publication number
WO2008150260A1
WO2008150260A1 PCT/US2007/013299 US2007013299W WO2008150260A1 WO 2008150260 A1 WO2008150260 A1 WO 2008150260A1 US 2007013299 W US2007013299 W US 2007013299W WO 2008150260 A1 WO2008150260 A1 WO 2008150260A1
Authority
WO
WIPO (PCT)
Prior art keywords
pyrido
pyrimidin
methoxy
amino
dichlorophenyl
Prior art date
Application number
PCT/US2007/013299
Other languages
French (fr)
Inventor
Krishna K. Murthi
Rebecca Casaubon
Arthur F. Kluge
Chase C. Smith
Original Assignee
Gpc Biotech, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to PCT/US2007/013299 priority Critical patent/WO2008150260A1/en
Application filed by Gpc Biotech, Inc. filed Critical Gpc Biotech, Inc.
Priority to BRPI0715566-2A priority patent/BRPI0715566A2/en
Priority to JP2009532937A priority patent/JP2010506902A/en
Priority to AU2007311480A priority patent/AU2007311480A1/en
Priority to EP07826759A priority patent/EP2084159A1/en
Priority to PCT/IB2007/054209 priority patent/WO2008047307A1/en
Priority to CA002665384A priority patent/CA2665384A1/en
Publication of WO2008150260A1 publication Critical patent/WO2008150260A1/en
Priority to US13/949,728 priority patent/US20140163223A1/en
Priority to US13/949,508 priority patent/US20140163004A1/en
Priority to US13/949,743 priority patent/US20140163007A1/en
Priority to US13/949,495 priority patent/US20140163003A1/en
Priority to US13/949,690 priority patent/US20140163006A1/en
Priority to US13/949,712 priority patent/US20140163222A1/en
Priority to US13/949,666 priority patent/US20140163005A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention provides derivatives of pyrido [2, 3-d] pyrimidin-7-one. These compounds are kinase inhibitors, including compounds that show anti-proliferative activity, including against tumor cells, and are useful in the treatment of diseases including cancer.

Description

-OXY-2-AMINOPYRIDO (2 , 3-D) PYRIMIDIN-7-ONE DERIVATIVES AS KINASE INHIBITORS AND
ANTICANCER AGENTS
Field of the Invention
The present invention provides derivatives of pyrido[2,3-d]pyrimidin-7-one. These compounds are kinase inhibitors, including compounds that show antiproliferative activity against cells, including against tumor cells, and are useful in the treatment of diseases including cancer.
Background of the Invention
Kinases are important cellular enzymes that perform essential cellular functions such as regulating cell division and proliferation, and appear to play a decisive role in many disease states such as in disease states that are characterized by uncontrolled proliferation and differentiation of cells. These disease states encompass a variety of cell types and maladies such as cancer, atherosclerosis, and restenosis.
Increased activity or temporally abnormal activation of cyclin-dependent kinases has been shown to result in the development of human tumors (Sherr C. J., Science 1996; 274:1672-1677). Indeed, human tumor development is commonly associated with alterations in either the Cdk proteins themselves or their regulators (Cordon-Cardo C, Am. J. Pathol. 1995; 147:545-560; Karp J. E. and Broder S., Nat. Med. 1995; 1 : 309-320; Hall M. et al., Adv. Cancer Res. 1996; 68:67-108). Naturally occurring protein inhibitors of Cdks such as pi 6 and p27 have been shown to cause growth inhibition in vitro in lung cancer cell lines (Kamb A., Curr. Top. Microbiol. Immunol. 1998.227:139-148).
Tyrosine kinases are essential for the propagation of growth factor signal transduction leading to cell cycle progression, cellular proliferation, differentiation, and migration. Tyrosine kinases include cell surface growth factor receptor tyrosine kinases (RTKs) such as FGFr and PDGFr as well as non-receptor tyrosine kinases including c-Src and Lck. Inhibition of these enzymes has been demonstrated to cause antitumor and antiangiogenesis activity (Hamby et al., Pharmacol. Ther, 1999; 82(2-3): 169-193). The molecular mechanisms and signaling pathways that regulate cell proliferation and survival are receiving considerable attention as potential targets for anticancer drug development. Recently, there has been a notable increase in efforts directed at targeting the MAPK pathway, which integrates proliferative signals that are initiated by a wide array of RTKs and G protein-coupled receptors.
The MAPK signal cascade includes a G protein, known as Ras, that works upstream of a core module consisting of three kinases: Raf, MEK1/2 and ERK1/2. hi this signal cascade, Raf (a serine/threonine kinase) phosphorylates and thus activates MEKl /2, which in turn ultimately leads to the activation of ERKl /2. Understanding of Raf function in Ras signaling is complicated by the fact that in mammals Raf is encoded by a gene family consisting of three genes (A-raf, B-raf and C-raf (raf-1)) which encode highly conserved 68 to 74 kD proteins (Daum et al., Trends Biochem. Sci. 1994, 19: 474-480) sharing highly conserved amino-terminal regulatory regions and catalytic domains at the carboxyl terminus. Raf proteins are normally cytosolic but are recruited to the plasma membrane by the small G-protein Ras, with this being an essential step for Raf activation by growth factors, cytokines, and hormones. Raf activation at the membrane occurs through a highly complex process involving conformation changes, binding to other proteins, binding to lipids, and phosphorylation and dephosphorylation of some residues.
Raf kinases, and particularly B-Raf, have long been considered attractive targets for drug discovery and therapeutic intervention due to their importance as potential checkpoints for cancer-related signal transduction (Tuveson et al., Cancer Cell, 2003, 4: 95-98; Strumberg and Seeber, Onkologie, 2005, 28: 101-107; Beeram et al., J. Clin. Oncol. 2005, 23: 6771-6790).
The importance of the MAPK signalling cascade for the proliferation and survival of tumor cells has recently increased following the discovery of a number of activating mutations of B-Raf in human tumors. Activating Raf mutations have been identified in melanoma, thyroid, colon, and other cancers (Davies et al., Nature,
2002, 417: 949-954; Cohen et al., J. Natl. Cancer Inst., 2003, 95: 625-627; Mercer and Pritchard, Biochim Biophys Acta, 2003, 1653: 25-40; Oliveira et al., Oncogene,
2003, 22: 9192-9196; Pollock et al., Nat. Genet. 2003, 33: 19-20; Domingo et al., Genes Chromosomes Cancer 2004, 39: 138-142; Shih and Kurman, Am. J. Pathol., 2004, 164: 1511-1518) Therefore, in addition to a role in controlling tumors with Ras mutations or activated growth factor receptors, inhibitors of Raf kinases harbor therapeutic potential for tumors carrying a B-Raf oncogene (Sharma et al., Cancer Res. 2005, 65: 2412-2421).
A variety of agents have been discovered to interfere with Raf kinases, including antisense oligonucleotides and small molecules. These inhibitors prevent the expression of Raf protein, block Ras/Raf interaction, or obstruct its kinase activity. Down-regulation of B-Raf activity by siRNA led to decreased tumorigenic potential of 1205 Lu cells (Sharma et al., Cancer Research, 2005, 65: 2412-2421), and by the kinase inhibitor BAY43-9006 (Sorafenib) led to inhibition of the growth of melanoma cells (Panka et al., Cancer Research., 2006, 66: 1611-9). Raf inhibitors that are currently undergoing clinical evaluation show promising signs of anti-cancer efficacy with a very tolerable safety profile. Clinically most advanced is the Raf inhibitor BAY 43-9006 (Sorafenib), which has been in phase II clinical testing for the treatment of metastatic renal cell carcinoma (Ratain et al., Proc. Am. Soc. Clin. Oncol. (ASCO Meeting Abstract), 2004, 23: Abstract 4501 ) and which recently entered phase III clinical testing.
The family of mitogen-activated protein (M AP) kinases are proline-directed serine/threonine kinases that activate their substrates by dual phosphorylation. The kinases are activated by a variety of signals including nutritional and osmotic stress, UV light, growth factors, endotoxin and inflammatory cytokines. One group of MAP kinases is the p38 kinase group that includes various isoforms (e. g., p38a, p39ss, p38 or p388).
The p38 kinases are responsible for phosphorylating and activating transcription factors as well as other kinases, and are activated by physical and chemical stress, proinflammatory cytokines, and bacterial lipopolysaccharides.
More importantly, the products of the p38 phosphorylation have been shown to mediate the production of inflammatory cytokines, including TNF, IL-I, and cyclooxygenase-2. These cytokines have been implicated in numerous disease states and conditions. For example, TNF-α is a cytokine produced primarily by activated monocytes and macrophages. Its excessive or unregulated production has been implicated as playing a causative role in the pathogenesis of rheumatoid arthritis. More recently, inhibition of TNF production has been shown to have broad application in the treatment of inflammation, inflammatory bowel disease, multiple sclerosis and asthma.
TNF has also been implicated in viral infections, such as HIV, influenza virus, and herpes virus including herpes simplex virus type-1 (HSV-I), herpes simplex virus type-2 (HSV-2), cytomegalovirus (CMV), varicella-zoster virus (VZV), Epstein Barr virus, human herpes virus-6 (HHV-6), human herpes virus- 7(HHV-7), human herpes virus-8 (HHV-8), pseudorabies and rhiriotracheitis, among others.
Similarly, IL-I is produced by activated monocytes and macrophages, and plays a role in many pathophysiological responses including rheumatoid arthritis, fever and reduction of bone resorption.
Additionally, p38 has been implicated in stroke, Alzheimer's disease, osteoarthritis, lung injury, septic shock, angiogenesis, dermatitis, psoriasis and atopic dermatitis, see, e.g., J. Exp. Opin. Ther. Patents, (2000) 10(1).
The inhibition of the above-mentioned cytokines by inhibition of the p38 kinase can be of benefit in controlling, reducing and/or alleviating one or more of these disease states.
Despite the progress that has been made, the search continues for low molecular weight kinase inhibitor compounds that are useful for treating a wide variety of diseases, including cancer, tumors and other proliferative disorders or diseases including restenosis, angiogenesis, diabetic retinopathy, psoriasis, surgical adhesions, macular degeneration, and atherosclerosis, or other disorders or diseases mentioned above. Thus, a strong need exists to provide compositions, pharmaceuticals and/or medicaments with kinase inhibitory, including antiproliferative activity against cells such as tumour cells. Such compositions, pharmaceuticals and/or medicaments may possess not only such activity, but may also exert tolerable, acceptable or diminished side effects in comparison to other anti -proliferative agents. Furthermore, the spectrum of tumors or other diseases responsive to treatment with such compositions, pharmaceuticals and/or medicaments may be broad. The active ingredients of such compositions, pharmaceuticals and/or medicaments may be suitable in the mentioned indication as single agent, and/or in combination therapy, be it in connection with other therapeutic agents, with radiation, with operative/surgical procedures, heat treatment or any other treatment known in the mentioned indications.
It is known that specific classes of pyrido[2,3-d]pyrimidines, substituted in a specific manner, have pharmacologically useful properties. In particular, specific derivatives of pyrido[2,3-d]pyrimidin-7-one are known to possess anti-proliferative activity. These compounds however are structurally dissimilar from the compounds of the present invention.
WO 96/34867 discloses 2-substituted and 2,8-disubstituted 6-aryl- pyrido[2,3-d]pyrimidin-7-ones and 7-imino derivatives thereof, that are shown to inhibit tyrosine kinases and to have certain activity in tumor models (see also US 5,620,981 and US 5,733,914). WO 01/55147 discloses 5,6-disubstituted 2,7- diamino-pyrido[2,3-d]pyrimidines having similar activities. WO 01/70741 discloses substituted 2-amino-5-(alkyl,aryl)-pyrido[2,3-d]pyrimidin-7-ones, that are shown to have Cdk inhibitor activity. WO 02/18380 discloses 8-unsubstituted and 8- substituted 2-amino-6-aryl-pyrido[2,3-d]pyrimidin-7-ones, that are shown to inhibit protein kinases, including p38. WO 02/18380 and WO 03/088972 disclose 2,4,8- tri substituted pyrido[2,3-d]pyrimidin-7-ones, that are shown to inhibit kinases and are thus are able to inhibit the production of various cytokines. WO 02/064594 discloses 8-unsubstituted and 8-substituted 2-amino-6-(amino,oxy)-pyrido[2,3- d]pyrimidin-7-ones and 7-imino derivatives thereof, that are shown to inhibit protein kinases, including p38. WO 03/062236 discloses 2-(pyrid-2-yl)amino-pyrido[2,3- d]pyrimidin-7-ones (optionally substituted at positions 5, 6 and/or 8), that are shown to be potent inhibitors of Cdk 4. WO 03/066630 discloses 6-(monocyclyl)- pyrido[2,3-d]pyrimidin-7-ones (optionally substituted at positions 2, 4 and/or 5), that are shown to be inhibitors of Cdks, and that showed activity in an ischemic stroke model. WO 2004/063195 discloses derivatives of 2-amino-8-methyl-6-phenyl- pyrido[2,3-d]pyrimidin-7-one that are shown to inhibit Bcr-Abl kinase. In particular, PCT publication WO 03/062236 discloses 2-(pyrid-2-yl)amino- pyrido[2,3-d]pyrimidin-7-ones and 2-(pyrid-2-yl)amino-dihydroρyrido[2,3- d]pyrimidin-7-ones (optionally substituted at positions 5, 6 and/or 8), as potent and selective inhibitors of Cdk 4 (and Cdk 6), and compares certain such compounds to their C2-phenylamino analogues, as disclosed in WO 98/33798 and WO 01/70741. The generic Markush structure disclosed and claimed in WO 03/062236 includes, amongst other suggested substituents at the nitrogen in position 8, a generically described substituent being "Ci to C8 alkoxy".
US patent application 2005/0182078 is directed to 2-(pyrid-3-yl)amino- pyrido[2,3-d]pyrimidin-7-ones, but does not provide for alkoxy substituents in position 8 of the pyrido[2,3-d]pyrimidin-7-one core.
Summary of the Invention
We have invented a class of 8-substitued pyrido[2,3-d]pyrimidin-7-ones that includes compounds that exhibit surprising properties, including activity as inhibitors of a number of protein kinases such as C- and B-Raf, or p38, and antiproliferative activity against cells such as tumour cells. Such derivatives of pyrido[2,3-d]pyrimidin-7-όnes provide an opportunity to develop new and effective therapies for diseases associated with kinase de-regulation or cellular proliferation, such as cancer or inflammatory disorders.
Compounds of the present invention are specific derivatives of pyrido[2,3- d]pyrimidin-7-one, as discussed in greater detail below. In analogy to certain pyrido[2,3-d]pyrimidin-7-ones previously described, the compounds of the present invention are suitable for further pre-clinical or clinical research and development towards the treatment of a variety of disorders and diseases including cancer, proliferative, degenerative, inflammatory and other disorders and diseases. The present invention provides effective therapies and therapeutics for particularly debilitating diseases such as cancer and other diseases and disorders including those listed herein.
One aspect of the invention relates to 8-oxy-pyrido[2,3-d]pyrimidin-7-ones having a structure represented by formula (I) presented below, or tautomeric or stereoisomeric forms thereof, which are useful as kinase inhibitors and thus useful for treating proliferative disorders or diseases such as such as cancer, atherosclerosis, and restenosis, among others.
In another aspect, the invention relates to pharmaceutical compositions, including a pharmaceutically acceptable diluent, excipient or carrier and an amount, such as a therapeutically effective amount, of such kinase inhibitor, e.g., which ameliorates the effects of proliferative disorders or diseases such as those mentioned above.
Another aspect of the invention relates to a pharmaceutical package, including such pharmaceutical composition, and instructions which indicate that said pharmaceutical composition may be used for the treatment of a patient suffering from a proliferative disorder or disease such as cancer.
In another aspect, the invention relates to methods that involve administering to or contacting a subject, a cell, a tissue, an organ or an organism with a therapeutically effective amount of a pharmaceutical composition disclosed herein. These methods include, but are not limited to, prophylaxis and/or treatment of a proliferative disorder or disease such as cancer, atherosclerosis, and restenosis, or prophylaxis and/or treatment of an inflammatory disorder or disease.
In another aspect, the invention relates to uses of the compounds of the present invention for the preparation of a medicament for the treatment of a proliferative disorder or disease, such as cancer.
In another aspect, the invention relates to uses of the compounds of the present invention for the preparation of a medicament for the treatment of an inflammatory disorder or disease.
Another aspect of the invention relates to methods of synthesizing the compounds of the present invention, and to intermediates for such compounds.
Accordingly, the present invention provides compounds having a structure represented by the general formula (I)
Figure imgf000009_0001
(I) or any tautomeric or stereoisomer^ form thereof, wherein
R1 is selected from hydrogen,
Figure imgf000009_0002
-C2-6-alkenyl, -C2-6-alkynyl, -C3-6- cycloalkyl and -C^e-cycloalkenyl;
V is selected from a bond, -O-, -N(R1 ')-, -C(=X)-, -S(O)n-, -C(=X)-O-, - C(=X)-N(Rn)-, -C(=X)-S-, -C(=X)-N(R1 1)-N(R1 1)-, -N(Rn)-C(=X)-, -N(R1 1)- CC=X)-N(R1 ')-, and -N(R1 ')-S(O) „-, with n = 1 or 2;
R2 is selected from hydrogen, -alkyl, -alkenyl, — alkynyl, -cycloalkyl, - cycloalkenyl, -heterocycloalkyl, — heterocycloalkenyl, -aryl and — heteroaryl; or R1 and R2, together with V and the nitrogen atom they are attached to, form a heterocycle;
R is selected from hydrogen, -Ci-6alkyl, -C2-6-alkenyl, -C2-6- alkynyl, -C3-6- cycloalkyl, -Cs-β-cycloalkenyl and halogen;
W is selected from a bond, -C(=O)-, -O-, and -N(R1 ')-;
R4 is selected from hydrogen, halogen, -alkyl, -alkenyl, —alkynyl, - cycloalkyl, -cycloalkenyl, -heterocycloalkyl, —heterocycloalkenyl, -aryl and — heteroaryl;
R5 is selected from hydrogen, -alkyl, -alkenyl, -alkynyl, -cycloalkyl, - cycloalkenyl, -(C-linked— heterocycloalkyl), -(C-linked-heterocycloalkenyl), —aryl, and -heteroaryl;
X is independently selected from =O, =S, =NR12, =N-OR13, =N-N(R! ')2, =N- N(R1 1XR12), and =N-N(R12)2; R10 is independently selected from -Ci^alkyl, -C2-6-alkenyl, -C2-6-alkynyl, - C3_6-cycloalkyl and -Ca-β-cycloalkenyl;
R11 is independently selected from hydrogen and R10;
R12 is independently selected from -alkyl, -alkenyl, -alkynyl, -cycloalkyl, - cycloalkenyl, -heterocycloalkyl, — heterocycloalkenyl, -aryl and — heteroaryl;
R13 is independently selected from hydrogen and R12; wherein R2, R4, R5, R10, and R12 may optionally be substituted; or any pharmaceutically acceptable salt or N-oxide thereof.
In certain embodiments, R4 is selected from hydrogen, -alkyl, -alkenyl, — alkynyl, -cycloalkyl, -cycloalkenyl, -heterocycloalkyl, -heterocycloalkenyl, -aryl and —heteroaryl. In other embodiments, R4 is halogen.
In certain embodiments, R5 is selected from -alkyl, -alkenyl, -alkynyl, - cycloalkyl, -cycloalkenyl, -(C-linked-heterocycloalkyl), -(C-linked- heterocycloalkenyl), —aryl, and -heteroaryl. In other embodiments, R5 is hydrogen.
In yet other certain embodiments, -V-R2, when taken together, does not include pyrid-2-yl, e.g., R2 is not substituted or unsubstituted pyrid-2-yl. In certain other embodiments, -V-R2 does not include pyridyl, e.g., R2 is not substituted or unsubstituted pyridyl.
In further certain embodiments, -O-R5, when taken together, is not Ci-8- alkoxy or an O-linked polyether containing between 2 and 8 carbon atoms in total. In certain such embodiments, R5 is not alkyl or alkoxy-substituted alkyl.
In more particular such embodiments, -V-R2, when taken together, does not include pyrid-2-yl, e.g., R2 is not substituted or unsubstituted pyrid-2-yl, and -O-R5, when taken together, is not a Ci-8-alkoxy or an O-linked polyether containing between 2 and 8 carbon atoms in total, or R is not alkyl or alkoxy-substituted alkyl. In alternative such embodiments, -V-R2, when taken together, does not include pyridyl, e.g., R2 is not substituted or unsubstituted pyridyl, and -O-R5, when taken together, is not a Ci-8-alkoxy or an O-linked polyether containing between 2 and 8 carbon atoms in total, or R5 is not alkyl or alkoxy-substituted alkyl. In certain embodiments, R2 is a substituted or unsubstituted phenyl ring. In other embodiments, R2 is a substituted or unsubstituted 5-membered heteroaryl ring, e.g, thienyl, furanyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, etc. In yet other embodiments, R2 is a substituted or unsubstituted heterocyclic ring, e.g., piperidine, piperazine, or morpholine. In yet other embodiments, R2 is a substituted or unsubstituted carbocyclic ring, e.g., cyclopropyl, cyclopentyl, or cyclohexyl. In yet other embodiments, R2 is a substituted or unsubstituted alkyl group, e.g., aralkyl, heteroaralkyl, hydroxyalkyl, alkoxyalkyl, etc. In yet other embodiments, R2 is acyl, e.g., alkylC(=O), alkenylC(=O), or cycloalkylC(=O). In certain embodiments, R2 is a substituted or unsubstituted 3-pyridyl, 4-pyridyl, pyrazinyl, pyrimidyl, or pyridazyl.
In certain embodiments, compounds disclosed in WO 03/062236 (incorporated by reference herein), e.g., disclosed as discrete compounds or as represented by the general formula (VI), are excluded:
Figure imgf000011_0001
(VI) . wherein:
Xla, X2a, and X3a are in each instance independently selected from hydrogen, halogen, C)-C6 alkyl, Ci-C6 haloalkyl, C(-C8 alkoxy, Ci-C8 alkoxyalkyl, CN, NO2, OR5a, NR5aR6a, CO2R5a, CORSa, S(O)naR5a, CONR5aR6a, NR5aCOR6a, NR5aSO2R6a, SO2NR5aR6a, and P(O)(OR5a)(OR6a); with the proviso that at least one of Xla, X2a, and X3a must be hydrogen; na = 0-2; Rla is, in each instance, independently, hydrogen, halogen, Ci-C6 alkyl, Q- C6 haloalkyl, CpC6 hydroxyalkyl, or C3-C7 cycloalkyl;
R2a and R4a are independently selected from hydrogen, halogen, Ci-C8 alkyl, C3-C7 cycloalkyl, C1-C8 alkoxy, C)-C8 alkoxyalkyl, Ci-C8 haloalkyl, C1-C8 hydroxyalkyl, C2-C8 alkenyl, C2-C8 alkynyl, nitrile, nitro, OR5a, SR5a, NR5aR6a, N(O)R5aR6a, P(O)(ORSa)(OR6a), (CR5aR6a)maNR7aR8a, COR5a, (CR4aR5a)maC(O)R7a, CO2R5a, CONR5aR6a, C(O)NR5aSO2R6a, NR5aSO2R6a, C(O)NR5aOR6a, S(O)naR55a, SO2NR5aR6a, P(O)(OR5a)(OR6a), (CR5aR6a)maP(O)(OR7a)(OR8a), (CR5aR6a)ma-aryl, (CR5aR6a)ma-heteroaryl, -Ta(CH2)maQaR5a, -C(O)Ta(CH2)maQaR5a,
NR5aC(O)Ta(CH2)maQaR5a, and -CR5a=CR6aC(O)R7a; or
Rla and R2a may form a carbocyclic group containing 3-7 ring members, preferably 5-6 ring members, up to four of which can optionally be replaced with a heteroatom independently selected from oxygen, sulfur, and nitrogen, and wherein the carbocyclic group is unsubstituted or substituted with one, two, or three groups independently selected from halogen, hydroxy, hydroxyalkyl, nitrile, lower C]-C8 alkyl, lower Ci-C8 alkoxy, alkoxycarbonyl, alkylcarbonyl, alkyl carbonyl amino, aminoalkyl, trifluoromethyl, N-hydroxyacetamide, trifluoromethylalkyl, amino, and mono or dialkylamino, (CH2)maC(O)NR5aR6a, and O(CH2)maC(O)OR5a, provided, however, that there is at least one carbon atom in the carbocyclic ring and that if there are two or more ring oxygen atoms, the ring oxygen atoms are not adjacent to one another;
Ta is O, S, NR7a, N(O)R7a, NR7aR8aWa, or CR7aR8a;
Qa is O, S, NR7a, N(O)R7a, NR7aR8aWa, CO2, 0(CH2)mn-heteroaryl, O(CH2)maS(O)naR8a, (CH2)-heteroaryl, or a carbocyclic group containing from 3-7 ring members, up to four of which ring members are optionally heteroatoms independently selected from oxygen, sulfur, and nitrogen, provided, however, that there is at least one carbon atom in the carbocyclic ring and that if there are two or more ring oxygen atoms, the ring oxygen atoms are not adjacent to one another, wherein the carbocyclic group is unsubstituted or substituted with one, two, or three groups independently selected from halogen, hydroxy, hydroxyalkyl, lower alkyl, lower alkoxy, alkoxycarbonyl, alkylcarbonyl, alkylcarbonylamino, aminoalkyl, trifluoromethyl, N-hydroxyacetamide, trifluoromethylalkyl, amino, and mono or dialkyl amino;
Wa is an anion selected from the group consisting of chloride, bromide, trifluoroacetate, and triethylammonium; ma = 0-6;
R4a and one of Xla, X2a, and X3a may form an aromatic ring containing up to three heteroatoms independently selected from oxygen, sulfur, and nitrogen, and optionally substituted by up to 4 groups independently selected from halogen, hydroxy, hydroxyalkyl, lower alkyl, lower alkoxy, alkoxy carbonyl, alkylcarbonyl, alkylcarbonylamino, aminoalkyl, aminoalkylcarbonyl, trifluoromethyl, trifluoromethyl alkyl, trifluoromethylalkylaminoalkyl, amino, mono- or dialkylamino, N-hydroxyacetamido, aryl, heteroaryl, carboxyalkyl, nitrile, NR7aSO2R8a, C(O)NR7aR8\ NR7aC(O)R8a, C(O)OR7a, C(O)NR7aSO2R8a, (CH2)maS(O)naR7a, (CH2)ma-heteroaryl, O(CH2)ma-heteroaryl, (CH2)]IiaC(O)NR7aR8a, O(CH2)maC(O)OR7a, (CH2)raaSO2NR7aR8a, and C(O)R7a;
R3a is Ci-C8 alkoxy;
R5a and R6a independently are hydrogen, Ci-C8 alkyl, C2-C8 alkenyl, C2-Cs alkynyl, arylalkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, or heter aryl alkyl; or
R5a and R6a, when attached to the same nitrogen atom, taken together with the nitrogen to which they are attached, form a heterocyclic ring containing from 3-8 ring members, up to four of which members can optionally be replaced with heteroatoms independently selected from oxygen, sulfur, S(O), S(O)2, and nitrogen, provided, however, that there is at least one carbon atom in the heterocyclic ring and that if there are two or more ring oxygen atoms, the ring oxygen atoms are not adjacent to one another, wherein the heterocyclic group is unsubstituted or substituted with one, two or three groups independently selected from halogen, hydroxy, hydroxyalkyl, lower alkyl, lower alkoxy, alkoxycarbonyl, alkylcarbonyl, alkylcarbonylamino, aminoalkyl. aminoalkylcarbonyl, trifluoromethyl, trifluoromethylalkyl, trifluoromethylalkylaminoalkyl, amino, nitrile, mono- or dialkylamino, N-hydroxyacetamido, aryl, heteroaryl, carboxyalkyl, NR7aSO2R8a, C(O)NR7aR8a, NR7aC(O)R8a, C(O)OR73, C(O)NR7aSO2R8a, (CH2)maS(O)naR7a, (CH2)ma-heteroaryl, O(CH2)ma-heteroaryl, (CH2)maC(O)NR7aR8a,
O(CH2)maC(O)OR7a, and (CH2)SO2NR7aR8a;
R7a and R8a are, independently, hydrogen, Cj-C8 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, arylalkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, or heteroarylalkyl; or
R7a and R8a, when attached to the same nitrogen atom, taken together with the nitrogen to which they are attached, may form a heterocyclic ring containing from 3-8 ring members, up to four of which members are optionally heteroatoms independently selected from oxygen, sulfur, S(O), S(O)2, and nitrogen, provided, however, that there is at least one carbon atom in the heterocyclic ring and that if there are two or more ring oxygen atoms, the ring oxygen atoms are not adjacent to one another, wherein the heterocyclic group is unsubstituted or substituted with one, two or three groups independently selected from halogen, hydroxy, hydroxyalkyl , lower alkyl, lower alkoxy, alkoxycarbonyl, alkylcarbonyl, alkylcarbonylamino, aminoalkyl, aminoalkylcarbonyl, trifluoromethyl, trifluoromethylalkyl, trifluoromethylalkylaminoalkyl, amino, nitrile, mono- or dialkylamino, N- hydroxyacetamido, aryl, heteroaryl, carboxyalkyl;
In certain embodiments of Formula VI, Ci-C8 alkoxy (e.g., in R3a) refers to straight or branched chain alkyl groups having 1 -8 carbon atoms and linked through oxygen, such as methoxy, ethoxy, propoxy, isopropoxy, n-butoxy, sec-butoxy, tert- butoxy, pentoxy, 2-pentyloxy, isopentoxy, neopentoxy, hexoxy, 2-hexoxy, 3- hexoxy, and 3-methylpentoxy. In certain embodiments, Ci-C8 alkoxy (e.g., in R3a) includes polyethers, e.g., alkoxys bearing alkyl chains having from 1 to 8 carbon atoms interrupted one or more times by oxygen atoms, such as methoxymethoxy, methoxy ethoxy, butoxyethoxy, methoxyethoxyethoxy, and the like.
Other features and advantages of the invention will be apparent from the following detailed description and from the claims. Detailed Description of the Invention
Definitions
The term "alkyl" refers to straight- or branched-chain saturated hydrocarbon groups having from 1 to about 20 carbon atoms, including groups having from 1 to about 7 carbon atoms. In certain embodiments, alkyl substituents may be lower alkyl substituents. The term "lower alkyl" refers to alkyl groups having from 1 to 6 carbon atoms, and in certain embodiments from 1 to 4 carbon atoms. Examples of alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, i-propyl, n- butyl, s-butyl, t-butyl, n-peπtyl, s-pentyl, and hexyl.
The term "alkenyl" refers to groups having 2 to about 20 carbon atoms, wherein at least one of the carbon-carbon bonds is a double bond, while other bonds may be single bonds or further double bonds. The term "alkynyl" herein refers to groups having 2 to about 20 carbon atoms, wherein at least one of the carbon-carbon bonds is a triple bond, while other bonds may be single, double or further triple bonds. Examples of alkenyl groups include ethenyl, 1-propenyl, 2-propenyl, 1- butenyl, 2-butenyl, 3-butenyl, 2-methyl-l -propenyl, 2-methyl-2-propenyl, and the like. Examples of alkynyl groups include ethynyl, 1-propynyl, 2-propynyl, and so forth.
As used herein, "cycloalkyl" is intended to refer to a ring being part of any stable monocyclic or polycyclic system, where such ring has between 3 and about 12 carbon atoms, but no heteroatom, and where such ring is fully saturated, and the term "cycloalkenyl" is intended to refer to a ring being part of any stable monocyclic or polycyclic system, where such ring has between 3 and about 12 carbon atoms, but no heteroatom, and where such ring is at least partially unsaturated (but excluding any aryl ring). Examples of cycloalkyls include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, adamantyl, cyclooctyl, bicycloalkyls, including bicyclooctanes such as [2.2.2]bicyclooctane or [3.3.0]bicyclooctane, bicyclononanes such as [4.3.0]bicyclononane, and bicyclodecanes such as [4.4.0]bicyclodecane (decalin), or spiro compounds. Examples of cycloalkenyls include, but are not limited to, cyclopentenyl or cyclohexenyl. For the sake of clarity, if a substituent is a polycyclic ring system as described above wherein one ring is a least partially unsaturated, then such substituent will be referred to as "cycloalkenyl", if substitution occurs via the at least partially unsaturated ring, and as "cycloalkyl", if substitution occurs via a fully saturated ring.
The term "Cx-y-", when used in combination with a group as defined herein, is intended to indicate the range of carbon atoms being present in the respective group, excluding substituents. For example, the term "d-6-alkyl" refers to the alkyl groups having between one carbon atom (i.e. a methyl group) and six carbon atoms (e.g. n-hexyl); the term "Cs-β-cycloalkyl" includes cyanocyclohexane, which is a substituted Cβ-cycloalkyl, not a C7-cycloalkyl, because the substituent is not inherently a cycloalkyl.
As used herein, the terms "heterocycloalkyl" and "heterocycloalkenyl", are intended to refer to a ring being part of any stable monocyclic or polycyclic ring system, where such ring has between 3 and about 12 atoms, and where such ring consists of carbon atoms and at least one heteroatom, particularly at least one heteroatom independently selected from the group consisting of N, O and S, with heterocycloalkyl referring to such a ring that is fully saturated, and heterocycloalkenyl referring to a ring that is at least partially unsaturated (but excluding any aryl or heteroaryl ring). For the sake of clarity, if a substituent is a polycyclic ring system as described above wherein one ring contains at least one heteroatom, then such substituent will be referred to as "heterocycloalkyl/-alkenyl", if substitution occurs via the ring containing the heteroatom(s). Heterocycloalkyl and heterocycloalkenyl groups may be linked to other groups via a carbon ring atom ("C-linked-heterocycloalkyl" and "C-linked-heterocycloalkenyl", respectively), or via a nitrogen ring atom ("N-linked-heterocycloalkyl" and "N- linkedheterocycloalkenyl", respectively). Heteroatoms such as nitrogen and sulfur may optionally be oxidized to form N-oxides or sulfoxides and sulfones, respectively. In certain embodiments, a nitrogen in the heterocycle may be quaternized. In certain embodiments, when the total number of S and O atoms in the heterocycle exceeds 1 , then these heteroatoms are not adjacent to one another. In particular embodiments, the total number of S atoms in the heterocycle is not more than 1. Examples of unsubstituted heterocycloalkyls include, but are not limited to, pyrrolidinyl, tetrahydroforanyl, morpholinyl, thiomorpholinyl, piperazinyl, piperidinyl, decahydroquinolinyl (when linked via the piperidinyl moiety), or imidazolidinyl. Examples of heterocycloalkenyls include, but are not limited to, pyrrolinyl, 2H, 6H-1 ,5,2-dithiazinyl, dihydrofuro[2,3-b]tetrahydrofuranyl, imidazolinyl, indolenyl (when linked via the 5-membered ring), indolinyl (when linked via the 5-membered ring), octahydroisoquinolinyl (when linked via the piperidinyl moiety), tetrahydroisoquinolinyl (when linked via the piperidinyl moiety), or tetrahydroquinolinyl (when linked via the piperidinyl moiety). Also included are fused ring and spiro compounds containing, for example, any of the above heterocycles, in each case when linked via the heteratom-containing ring. Also included are substituted heterocycloalkyls and heterocycloalkenyls, where substitution may occur by one or more of the substituents described herein at any heteroatom or carbon atom of the heterocycle, where such substitution results in a stable structure. Examples of substituted heterocycloalkyls include, but are not limited to, 2-pyrrolidonyl, 4-piperidonyl, or 4-alkyl -piperazinyl. Examples of substituted heterocycloalkenyls include, but are not limited to, maleinimido or 1,4- dihydropyridinyl .
The term "aryl" is intended to mean a ring or ring system being part of any stable monocyclic or polycyclic system, where such ring or ring system has between 3 and about 20 carbon atoms, but has no heteroatom, which ring or ring system consists of an aromatic moiety as defined by the "2n+2 π electron rule". For the sake of clarity, if a substituent is a polycyclic system as described above wherein one ring or ring system consists of an aromatic ' moiety as defined herein, then such substituent will be referred to as "aryl", if substitution occurs via the aromatic moiety. This includes phenyl and benzene rings fused to, e.g. one or aryl, e.g., to other benzene rings to form, for example, anthracene, phenanthrene, or naphthalene ring systems, or to one or more cycloalkyl moieties to form, for example, indanyl, fluorenyl or tetrahydronaphthyl (tetralin), or fused to heterocycloalklyl rings (provided, in each case, that such fused system is linked as a substituent via the aromatic moiety). As used herein, the term "heteroaryl" refers to a ring or ring system being part of any stable mono- or polycyclic system, where such ring or ring system has between 3 and about 20 atoms, which ring or ring system consists of an aromatic moiety as defined by the "2n+2 π electron rule" and contains carbon atoms and one or more nitrogen, sulfur, and/or oxygen heteroatoms. For the sake of clarity, if a substituent is a polycyclic system as described above wherein one ring or ring system consists of an aromatic moiety containing a heteroatom as defined herein, then such substituent will be referred to as "heteroaryl", if substitution occurs via the aromatic moiety containing the heteroatom. In certain embodiments, the total number of N5 S and O atoms in the heteroaryl is between 1 and about 4. In certain embodiments, the total number of S and O atoms in the aromatic heteroaryl is not more than 1. In certain embodiments, a nitrogen in the heterocycle may be quaternized or oxidized to an N-oxide. Heteroaryl moieties may contain 3 to about 12 members per ring. Examples of heteroaryls include, but are not limited to, IH- indazolyl, 2H-pyrrolyl, 3H-indolyl, 4H-quinolizinyl, 6H-l,2,5-thiadiazinyl, acridinyl, azocinyl, benzimidazolyl, benzofuranyl , benzothiofuranyl, bcnzothienyl, benzoxazolyl, benzthiazolyl, bcnztriazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazalonyl, carbazolyl, 4aH-carbazolyl, P-carbolinyl, chromanyl, chromenyl, cinnoliny], 2H,6H dithiazinyl, furanyl, furazanyl, imidazolyl, lH-indazolyl, indolizinyl, indolyl, isobenzofuranyl, isochromanyl, isoindazolyl, isoindolinyl, isoindolyl, isoquinolinyl, isothiazolyl, isoxazolyl, naphthyridinyl, oxadiazolyl, 1 ,2,3- oxadiazolyl, 1 ,2,4-oxadiazolyl, 1,2,5-oxadiazolyl, 1 ,3,4-oxadiazolyl, oxazolidinyl, oxazolyl, oxazolidinylperimidinyl, phenanthridinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, phenoxathiinyl, phenoxazinyl, phthalazinyl, pteridinyl, pteridinyl, purinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolyl, pyridazyl, pyridazinyl, pyridooxazole, pyridoimidazole, pyridothiazole, pyridinyl, pyridyl, oxo- pyridyl, pyrimidinyl, pyrrolyl, quinazolinyl, quinolinyl, 4H-quinolizinyl, quinoxalinyl, quinuclidinyl, carbolinyl, 6H-l,2,5-thiadiazinyl, 1 ,2,3-thiadiazolyl, 1 ,2,4-thiadiazolyl, 1,2,5-thiadiazolyl, 1,3,4-thiadiazolyl, thianthrenyl, thiazolyl, thienyl, thienothiazolyl, thienooxazolyl, thienoimidazolyl, triazinyl, 1,2,3-triazolyl, 1 ,2,4-triazolyl, 1 ,2,5-triazolyl, 1,3,4-triazolyl, tetrazolyl, xanthenyl. Preferred heterocycles include, but are not limited to, pyridinyl, furanyl, thienyl, pyrrolyl, pyrazolyl, imidazolyl, indolyl, benzimidazolyl, lH-indazolyl, oxazolidinyl, benzotriazolyl, benzisoxazolyl, oxindolyl, benzoxazolinyl, or isatinoyl and substituted versions thereof.
Also included in the term heteroaryl are fused heteroaryls containing, for example, the above heteroaryls fused to cycloalkyls or heterocycloalkyls (provided, in each case, that such fused system is linked as a substituents via the aromatic moiety containing at least one heteroatom), or aryls or other heteroaryls.
Alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, heterocycloalkyl, heterocycloalkenyl, aryl and heteroaryl groups as well as and any other substructure comprising at least one hydrogen in the substructure may optionally be substituted by one or more substituents. "Substituted" is intended to indicate that one or more hydrogens on the atom or group indicated in the expression using "substituted" is replaced with a selection from the indicated group(s), provided that the indicated atom's normal valency, or that of the appropriate atom of the group that is substituted, is not exceeded, and that the substitution results in a stable compound. The term "optionally substituted" is intended to mean that a given compound, or substructure of a compound, is either unsubstituted, or substituted, as defined above, with one or more substituents, as defined herein. When a substituent is keto or oxo (i.e., =O) group, a thio or imino group or the like, then two hydrogens on the atom are replaced. Keto/oxo substituents are not direct substituents of aromatic moieties. Exemplary substituents include, for example, alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, heteroaralkyl, acyl, aroyl, heteroaroyl, carboxyl, alkoxy, aryloxy, acyloxy, aroyloxy, heteroaroyloxy, alkoxycarbonyl, halogen, (thio)ester, cyano, phosphoryl, amino, imino, (thio)amido, sulfhydryl, alkylthio, acylthio, sulfonyl, a sulfate, a sulfonate, a sulfamoyl, a sulfonamido, nitro, azido, haloalkyl, inducing perfluoroalkyl (such as trifiuoromethyl), haloalkoxy, alkylsulfanyl, alkylsulfinyl, alkylsulfonyl, alkylsulfonylamino, arylsulfonoamino, phosphoryl, phosphate, phosphonate, phosphinate, alkylcarboxy, alkylcarboxyamide, oxo, hydroxy, mercapto, amino (optionally mono- or di- substituted, e.g. by alkyl, aryl, or heteroaryl), imino, carboxamide, carbamoyl (optionally mono- or di -substituted, e.g. by alkyl, aryl, or heteroaryl), amidino, aminosulfonyl, acylamino, aroylamino, (thio)ureido, arylthio)ureido, alkyl(thio)ureido, cycloalkyl(thio)ureido, aryloxy, aralkoxy, or -O(CH2)n-OH, - O(CH2)n-NH2, -O(CH2)nCOOH, -(CHa)nCOOH5 -C(O)O(CH2) nR, (CH2)nN(H)C(O)OR, or -N(R)S(O)2R wherein n is 1-4 and R is independently selected from hydrogen, -alkyl, -alkenyl, -alkynyl, -cycloalkyl, -cycloalkenyl, -(C- linked— heterocycloalkyl), -(C-linked-heterocycloalkenyl), — aryl, and — heteroaryl, with multiple degrees of substitution being allowed. It will be understood by those skilled in the art that substituents, such as heterocycloalkyl, aryl, heteroaryl, alkyl, etc., or functional groups such as -OH, -NHR etc., can themselves be substituted, if appropriate. It will also be understood by those skilled in the art that the substituted moieties themselves can be substituted as well when appropriate. Examples of substituted aryl groups include p-tolyl, 4-methoxyphenyl, 4-tert-butoxyphenyl, 3- methyl-1-rnethoxyphenyl, 4-fluorophenyl, 4-chlorophenyl, 3-nitrophenyl, 3- aminophenyl, 3-acetamidophenyl, 4-acetamidophenyl, 2-methyl-l-acetamidophenyl, 2-methyl aminophenyl, 3-methyl-l-aminophenyl, 2-amino-l-methylphenyl, 2,4- dimcthyl-1-aminophcnyl, 4-hydroxyphenyl, 3-methyl-l-hydroxyphenyl, 1-naphthyl, 2-naphthyl, 3-amino-l-naphthyl, 2-methylaminonaphthyl, 6-aminonaphthyl, 4,6- dimethoxynaphthyl and the like.
Nitrogen-containing substructures, such as amines or nitrogen-containing heterocycles, may be substituted as well, by formation of quaternary amines or N- oxides.
A divalent alkyl group being substituted at both ends of the alkyl chain may also be referred to as "alkylene" group, e.g., methylene (-CH2-), or ethylene (-CH2- CH2-).
The terms "alkenylene" and "alkynylene" refer to the corresponding divalent groups with at least one double and triple bond, respectively, as described above for alkylenes. These groups may or may not be branched. Examples of alkenylene groups include ethenylene, 1-propenylene, 2-propenylene, 1-butenylene, 2- butenylene, 3-butenylene, 2-methyl-l-propenylenc, 2-methyl-2-propenylene, and the like. Examples of alkynylene groups include ethynylene, 1-propynylene, 2- propynylene, and so forth. In addition, the terms are intended to include both unsubstituted and substituted alkenylene and alkynylene groups. Substituted alkenylene and alkynylene groups refer to alkenylene and alkynylene moieties having one or more hydrogen substituents replaced by a substituent as indicated above.
If alkylene, alkenylene, alkynylene or similar groups are linked with both ends to the same moiety, cyclic structures will result, where two hydrogens of said moiety are being replaced by the two ends of the alkylene, alkenylene, alkynylene or similar group, thus creating cyclic structures, as in tetralin, macrocycles or spiro compounds.
An alkyl/aryl group that is substituted by an alkoxy/aryloxy group may be termed "ether", e.g. dimethyl ether or methyl phenyl ether.
The term "acyl" refers to a group represented by the general formula -C(=O)- (cyclo)alkyl.
The term "aroyl" refers to a group represented by the general formula - C(=O)-aryl.
The terms "amide" and "amido" are art-recognized as an amino-substituted carbonyl and includes a moiety that can be represented by the general formula - CX=O)NR2. Preferred embodiments of the amide will not include imides which may be unstable.
The term "alkoxy'V'aryloxy" refers to an alkyl/aryl group having an oxygen attached thereto. Representative alkoxy groups include methoxy, ethoxy, propoxy, tert-butoxy and the like, and a representative aryloxy group is phenoxy. The term "lower alkoxy" refers to a lower alkyl group attached to an oxygen atom.
The term "polyether" refers to two or more alkyl groups linked through and separated by oxygen atoms. A representative polyether is -O-(CH2)2-O-CH3.
The term "halogen" or "halo" refers to fluoro, chloro, bromo and iodo substituents.
The terms "stereoisomer" and "tautomer" as used herein include all possible stereoisomeric and tautomeric forms of the compounds of the present invention. Where the compounds of the present invention contain one or more chiral centers, all possible enantiomeric and diastereomeric forms are included. The present invention is intended to. include all isotopes of atoms occurring on the present compounds. Isotopes are atoms having the same atomic number but different mass numbers. By way of general example and without limitation, isotopes of hydrogen include tritium and deuterium. Isotopes of carbon include 12C and 14C.
The term "metabolite", as used herein, refers to any substance produced by the metabolism or by a metabolic process. Metabolism, as used herein, refers to the various physical/chemical/biochemical/pharmacological reactions involved in the transformation of molecules or chemical compounds occurring in the cell, tissue, system, body, animal, individual, patient or human therein.
The term "IC50", as used herein, refers to concentrations at which a measurable activity, phenotype or response, for example growth or proliferation of cells such as tumor cells, is inhibited by 50%. IC50 values can be estimated from an appropriate dose-response curve, for example by eye or by using appropriate curve fitting or statistical software. More accurately, IC50 values may be determined using non-linear regression analysis.
As used herein, an "individual" means a multi-cellular organism, for example an animal such as a mammal, including a primate. In addition to primates, such as humans, a variety of other mammals can be treated according to a method that utilizes one or more compounds of the present invention. For example, mammals including, but not limited to, cows, sheep, goats, horses, dogs, cats, guinea pigs, rats or other bovine, ovine, equine, canine, feline, rodent, or murine species can be used.
As used herein, a "proliferative disorder" or a "proliferative disease" includes a disease or disorder that affects a cellular growth, differentiation, or proliferation process.
As used herein, a "cellular growth, differentiation or proliferation process" is a process by which a cell increases in number, size or content, by which a cell develops a specialized set of characteristics which differ from that of other cells, or by which a cell moves closer to or further from a particular location or stimulus. A cellular growth, differentiation, or proliferation process includes amino acid transport and degradation and other metabolic processes of a cell. A cellular proliferation disorder may be characterized by aberrantly regulated cellular growth, proliferation, differentiation, or migration. Cellular proliferation disorders include tumorigenic diseases or disorders.
As used herein, a "tumorigenic disease or disorder" includes a disease or disorder characterized by aberrantly regulated cellular growth, proliferation, differentiation, adhesion, or migration, which may result in the production of or tendency to produce tumors. As used herein, a "tumor" includes a benign or malignant mass of tissue. Examples of cellular growth or proliferation disorders include, but are not limited to tumors, cancer, autoimmune diseases, viral diseases, fungal diseases, neurodegenerative disorders and cardiovascular diseases.
As used herein, the terms "anti-cancer agent" or "antiproliferative agent" refer to compounds with anti-cancer and anti-proliferative properties, respectively. These compounds include, but are not limited to, altretamine, busulfan, chlorambucil, cyclophosphamide, ifosfamide, mechloreth amine, melphalan, thiotepa, cladribine, fluorouracil, floxuridine, gemcitabine, thioguanine, pentostatin, methotrexate, 6-mercaptopurine, cytarabine, carmustine, lomustine, streptozotocin, carboplatin, cisplatin, oxaliplatin, picoplatin, LA-12, iproplatin, tetraplatin, lobaplatin, JM216, JM335, satraplatin, fludarabine, aminoglutethimide, flutamide, goserelin, leuprolide, megestrol acetate, cyproterone acetate, tamoxifen, anastrozole, bicalutamide, dexamethasone, diethylstilbestrol, prednisone, bleomycin, dactinomycin, daunorubicin, doxirubicin, idarubicin, mitoxantrone, losoxantrone, mitomycin-c, plicamycin, paclitaxel, docetaxel, topotecan, irinotecan, 9-amino camptothecan, 9-nitro camptothecan, GS-21 1, JM 1 18, etoposide, teniposide, vinblastine, vincristine, vinorelbine, procarbazine, asparaginase, pegaspargase, octreotide, cstramustine, and hydroxyurea. Said terms also include, but are not limited to, non-small molecule therapeutics, such as antibodies, e.g., 1D09C3 and other anti-HLA-DR antibodies as described in WO 01/87337 and WO 01/97338, Rituxan as described in US patents 5,736,137, 5,776,456, 5,843,437, 4D5, Mab225, C225, Daclizumab (Zenapax), Antegren, CDP 870, CMB-401, MDX-33, MDX-220, MDX-477, CEA-CIDE, AHM, Vitaxin, 3622W94, Therex, 5G1.1 , IDEC-131, HU- 901, Mylotarg, Zamyl (SMART M195), MDX-210, Humicade, LymphoCIDE, ABX-EGF, 17- IA, Trastuzumab (Herceptin ®, rhuMAb), Epratuzumab, Cetuximab (Erbitux ®), Pertuzumab (Omnitarg®, 2C4), R3, CDP860, Bevacizumab (Avastin ®), tositumomab (Bexxar ®), Ibritumomab tiuxetan (Zevalin ®), M195, IDlO, HuI DlO (Remitogen®, apolizumab), Danton/DN1924, an "HD" antibody such as HD4 or HD8, CAMPATH-I and CAMPATH-IH or other variants, fragments, conjugates, derivatives and modifications thereof, or other equivalent compositions with improved or optimized properties, and proteins or peptides, e.g., those described in Trends in Biotechnology (2003), 21(12), p.556-562.
As used herein, an "inflammatory disorder" or an "inflammatory disease" includes a disease or disorder that is caused or accompanied by inflammatory processes. This includes, but is not limited to, diseases or disorders such as arthritis, including but not limited to rheumatoid arthritis, spondyloarthropathies, gouty arthritis, osteoarthritis, systemic lupus erythematosus and juvenile arthritis, osteoarthritis, gouty arthritis and other arthritic conditions; pulmonary disorders or lung inflammation, including adult respiratory distress syndrome, pulmonary sarcoidosis, asthma, silicosis, and chronic pulmonary inflammatory disease; viral and bacterial infections, including sepsis, septic shock, gram negative sepsis, malaria, meningitis, cachexia secondary to infection or malignancy, cachexia secondary to acquired immune deficiency syndrome (AIDS), AIDS, ARC (AIDS related complex), pneumonia, and herpes virus; bone resorption diseases, such as osteoporosis, endotoxic shock, toxic shock syndrome, reperfusion injury, autoimmune disease including graft vs. host reaction and allograft rejections, cardiovascular diseases including atherosclerosis, thrombosis, congestive heart failure, and cardiac reperfusion injury, renal reperfusion injury, liver disease and nephritis, and myalgias due to infection; Alzheimer's disease, influenza, multiple sclerosis, cancer, diabetes, systemic lupus erythematosus (SLE), skin-related conditions such as psoriasis, eczema, burns, dermatitis, keloid formation, and scar tissue formation; gastrointestinal conditions such as inflammatory bowel disease, Crohn's disease, gastritis, irritable bowel syndrome and ulcerative colitis; ophthalmic diseases, such as retinitis, retinopathies, uveitis, ocular photophobia, and of acute injury to the eye tissue; angiogenesis, including neoplasia; metastasis; ophthalmological conditions such as corneal graft rejection, ocular neovascularization, retinal neovascularization including neovascularization following injury or infection, diabetic retinopathy, retrolental fibroplasia and neovascular glaucoma; ulcerative diseases such as gastric ulcer; pathological, but non-malignant, conditions such as hemangiomas, including infantile hemangiomas, angiofibroma of the nasopharynx and avascular necrosis of bone; diabetic nephropathy and cardiomyopathy; and disorders of the female reproductive system such as endometriosis.
As used herein, "pharmaceutically acceptable salts" refers to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from nontoxic inorganic or organic acids. For example, such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, mcthanesulfonic, ethane disulfonic, oxalic, isethionic, and the like.
The pharmaceutically acceptable salts of the present invention can be synthesized from a parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, EtOAc, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 18th ed., Mack Publishing Company, Easton, PA5 1990, p. 1445, the disclosure of which is hereby incorporated by reference. Any salt that retains the desired biological activity of the compounds contained herein and that exhibits minimal or no undesired or toxicological effects is intended for inclusion here. Pharmaceutically acceptable salts include those derived from pharmaceutically acceptable organic or inorganic acids and bases. Non- pharmaceutically acceptable acids and bases also find use herein, as for example, in the synthesis and/or purification of the compounds of interest. Thus, all "salts" are also encompassed within the scope of the instant invention.
Non-limiting examples of suitable salts include those derived from inorganic acids, such as, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid, bicarbonic acid, carbonic acid; and salts formed with organic acids, such as, for example, formic acid, acetic acid, oxalic acid, tartaric acid, succinic acid, malic acid, malonic acid, ascorbic acid, citric acid, benzoic acid, tannic acid, palmoic acid, alginic acid, polyglutamic acid, tosic acid, methanesulfonic acid, naphthalenesulfonic acid, naphthalenedi sulfonic acid, α- ketoglutaric acid, β-glycerophosphoric acid and polygalacturonic acid. Suitable salts include those derived from alkali metals such as lithium, potassium and sodium, from alkaline earth metals such as calcium and magnesium, as well as from other acids well known to those of skill in the pharmaceutical art. Other suitable salts include those derived from metal cations such as zinc, bismuth, barium, or aluminum, or with a cation formed from an amine, such as ammonia, N,N- dibenzylethylene-diamine, D-glucosamine, tetraethylammonium, or ethyl en ediamine. Moreover, suitable salts include those derived from a combination of acids and bases, such as, for example, a zinc tannate salt.
The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication commensurate with a reasonable benefit/risk ratio.
The term "prodrug", as used herein, refers to an agent that is converted into a pharmacologically active parent drug in vivo, such as a compound as defined herein. The term "prodrug" includes any covalently bonded carriers that release an active parent drug of the present invention in vivo when such prodrug is administered to an animal. Since prodrugs are known to enhance numerous desirable qualities of pharmaceuticals (e.g., solubility, bioavailability, manufacturing, transport, pharmacodynamics, etc.), the compounds of the present invention may be delivered in prodrug form. Prodrugs, for instance, may be bioavailable by oral administration even when the parent drug is not. Thus, the present invention is intended to cover prodrugs of the presently claimed compounds, methods of delivering the same, and compositions containing the same. Prodrugs of the present invention are prepared by modifying functional groups present in the compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound. Prodrugs include compounds of the present invention wherein a hydroxy, amino, or sulfhydryl group is bonded to any group that, when the prodrug of the present invention is administered to a mammalian subject, it cleaves to form a free hydroxyl, free amino, or free sulfydryl group, respectively. Examples of prodrugs include, but are not limited to, acetate, formate, and benzoate derivatives of alcohol and amine functional groups in the compounds of the present invention.
Generally speaking, prodrugs are derivatives of per se drugs that after administration undergo conversion or metabolism to the physiologically active species. The conversion may be spontaneous, such as hydrolysis in the physiological environment, or may be enzyme-catalyzed. Prodrugs include compounds that can be oxidized, reduced, aminated, deaminated, hydroxylated, dehydroxylated, hydrolyzed, esterifϊed, alkylated, dealkylated, acylated, deacylated, phosphorylated, and/or dephosphorylated to produce the active compound.
From among the voluminous scientific literature devoted to prodrugs in general, the foregoing examples are cited: Gangwar et al., "Prodrug, molecular structure and percutaneous delivery", Des. Biopharm. Prop. Prodrugs Analogs, [Symp.] Meeting Date 1976, 409-21. (1977); Nathwani and Wood, "Penicillins: a current review of their clinical pharmacology and therapeutic use", Drugs 45(6): 866-94 (1993); Sinhababu and Thakker, "Prodrugs of anticancer agents", Adv. Drug Delivery Rev. 19(2): 241-273 (1996); Stella et al., "Prodrugs. Do they have advantages in clinical practice?", Drugs 29(5): 455-73 (1985); Tan et al. "Development and optimization of anti-HIV nucleoside analogs and prodrugs: A review of their cellular pharmacology, structure-activity relationships and pharmacokinetics", Adv. Drug Delivery Rev. 39(1-3): 117-151 (1999); Design of Prodrugs (Bundgaard H. ed.) 1985 Elsevier Science Publishers B. V. (Biomedical Division), Chapter 1; Design of Prodrugs: Bioreversible derivatives for various functional groups and chemical entities (Hans Bundgaard); Bundgaard et al. Int. J. of Pharmaceutics 22 (1984) 45 - 56 (Elsevier); Bundgaard et al. Int. J. of Pharmaceutics 29 (1986) 19 - 28 (Elsevier); Bundgaard et al. J. Med. Chem. 32 (1989) 2503 - 2507 Chem. Abstracts 93. 137935y {Bundgaard et al.)', Chem. Abstracts 95, 138493f {Bundgaard et al.); Chem. Abstracts 95, 138592n (Bundgaard et al.); Chem. Abstracts 110, 57664p (Alminger et al.); Chem. Abstracts 115, 64029s (Buur et al.); Chem. Abstracts 1 15, 189582y (Hansen et al.); Chem. Abstracts 117, 14347q (Bundgaard et al.); Chem. Abstracts 117, 5579Ox (Jensen et al); and Chem. Abstracts 123, 17593b (Thomsen et al).
The terms "administered", "administration", or "administering" a compound will be understood to mean providing any compound of the invention to an individual, including an animal, in need of treatment by bringing such individual in contact with, or otherwise exposing such individual to, such compound.
The term "in vitro" refers to a biological entity, a biological process, or a biological reaction outside the body in artificial conditions. For example a cell grown in vitro is to be understood as a cell grown in an environment outside the body, e.g., in a test tube, a culture tray or a microtiter plate.
The term "therapeutically effective amount" means the amount of the subject compound that will elicit the biological, physiological, pharmacological, therapeutic or medical response of a cell, tissue, system, body, animal, individual, patient or human that is being sought by the researcher, scientist, pharmacologist, pharmacist, veterinarian, medical doctor, or other clinician, e.g., lessening of the effects/symptoms of a disorder or disease, such as a proliferative disorder or disease, for example, a cancer or tumor, or killing or inhibiting growth of a proliferating cell, such as a tumor cell. The therapeutically effective amount can be determined by standard procedures, including those described below in the section "Dosages". The term "further treated", "further administer", or "further administered" means that different therapeutic agents or compounds may be administered together, alternatively or intermittently. Such further administration may be temporally or spatially separated, for example, at different times, on different days or via different modes or routes of administration.
Compounds of the present invention
One aspect of the invention relates to a compound having a structure represented by formula (I) or any tautomeric or stereoisomeric form thereof, wherein one or more hydrogen atoms in any one of R2, R4, R5, R10, and R12 are independently substituted with substituents R6, with R6 being independently taken from the list of: Y-R14 and R15; with R14 being independently selected from -R13, -OR13, -SR13, - N(RI3)2, -N(R13)N(R13)2, -N=C(RI3)2, and -N=NR13; with R15 being independently selected from -F, -Cl, -Br, -I, -CN, -NO2, and =Z; with Y being independently selected from a bond, -C(=Z)-, -O-, -O-C(=Z)-, -N(R13)-, -N(R13)-C(=Z)-, -N(R13)- N(R13)-C(=Z)-, -N(Rl3)-S(O) n-, -S-, and -S(O)n-, with n = 1 or 2; provided that if Y is a bond, then R14 is not hydrogen; and with Z being independently selected from =O, =S, =NR12, =N-OR13, and =N-N(Rπ)2.
In another aspect, the invention relates to compounds having a structure represented by formula (I) or any tautomeric or stereoisomeric form thereof, wherein one or more hydrogen atoms in any one of R2, R4, R5, R!0, and R12 are independently substituted with substituents R7, with R being independently taken from R , wherein one or more hydrogens of R6 are substituted by substituents independently taken from the list of: Y-R14 and R15.
One aspect of the invention relates to a compound having a structure represented by formula (I) or any tautomeric or stereoisomeric form thereof, wherein R1 is hydrogen, and wherein R2, R3, R4, R5, V, W, and X are as defined above.
Another aspect of the invention relates to a compound having a structure represented by formula (I) or any tautomeric or stereoisomeric form thereof, wherein V is a bond. In certain embodiments, R2 is selected from -aryl and -heteroaryl, substituted with 0, 1, 2, 3, 4 or 5 substituents R8, wherein R8 is independently selected from R6 and R7, and wherein R1, R3, R4, R5, W, and X are as defined above.
In certain embodiments, R2 is -phenyl substituted with one substituent R8 in position 3 or 4 (i.e., in meta or para position).
In certain embodiments, R8 is independently selected from -O-C|-3-alkyl, — S- Ci-3-alkyl, -Ci.3-alkyl-OH, -SO2-NH2, and -N-linked-heterocycloalkyl. In certain alternative embodiments, R8 is selected from — heteroaryl, -CN, and halogen, e.g., chlorine or fluorine. In certain embodiments, R8 is independently selected from — (CH2)n-N-linked-heterocycloalkyl and -(CH2)n-heteroaryl with n selected from 0, 1 , 2, 3, 4 or 5, including embodiments when the N-linked-heterocycloalkyl is selected from morpholinyl, piperazinyl, pyrrolidinyl and azetidine, and the -heteroaryl is selected from pyrrole, pyrazole and triazole. In particular such embodiments, the alkyl, N-linked-heterocycloalkyl or -heteroaryl is substituted by 1 or 2 substituents. In more particular embodiments, n is 0 or 1.
In certain embodiments, R2 is -phenyl substituted with R8 in position 4 (i.e., in para position), including embodiments where R2 is substituted with one R8. In particular such embodiments, R8 is independently selected from — O-Ci-3-alkyl, — S- C]-3-alkyl, -Ci-3-alkyl-OH, -SO2-NH2, CN, halogen (e.g., chlorine or fluorine), — (CH2)n-N-linked-heterocycloalkyl and — (CH2)n-heteroaryl with n selected from 0, 1 , 2, 3, 4 or 5, including embodiments when the N-linked-heterocycloalkyl is selected from morpholinyl, piperazinyl, pyrrolidinyl and azetidine, and the -heteroaryl is selected from pyrrole, pyrazole and triazole. In particular such embodiments, the alkyl, N-linked-heterocycloalkyl or —heteroaryl is substituted by 1 or 2 substituents. In more particular embodiments, n is 0 or 1, including embodiments where R8 is a — (CH2),,-N-linked-heterocycloalkyl selected from morpholinyl and piperazinyl, optionally substituted by 1 or 2 substituents.
In certain embodiments, R2 is substituted with two substituents R8, including embodiments where R2 is -phenyl. In certain such embodiments, R2 is -phenyl substituted with two substituents R8 in position 3 and 4 (i.e., in meta and para positions). In certain such embodiments, R8 in position 4 (i.e., in para position) is a — (CH2)n-N-linked-heterocycloalkyl selected from morpholinyl and piperazinyl, optionally substituted by 1 or 2 substituents, where n is 0 or 1. In particular such embodiments, R8 in position 3 (i.e., in meta position) is selected from -CN, a halogen (e.g., chlorine or fluorine), — O-Ci-3-alkyl and -Ci-3-alkyl-OH.
In certain embodiments, R2 is a fϊve-membered heteroaryl substituted with 0, 1, 2 or 3 substituents R8, wherein R8 is independently selected from R6 and R7 (both as defined above), and wherein R1, R3, R4, R5, W, and X are as defined above.
In certain such embodiments, R2 is a pyrrolyl, e.g., pyrrol-3-yl. In particular embodiments, R2 is N-alkyl-pyrrol-3-yl. In further embodiments, R2 is N-alkyl- pyrroi-3-yl substituted in position 5 (i.e., adjacent to the pyrrole ring nitrogen) by a substituent taken from the list of: -C(=Z)-OR13, -e(=Z)-N(R13)2, and -C(=Z)- N(R13)N(R13)2 (with both Z and R13 as defined above). In particular such embodiments, R2 is N-alkyl-pyrrol-3-yl substituted in position 5 (i.e., adjacent to the pyrrole ring nitrogen) by a substituent taken from the list of: -C(=O)-OR13, and - C(=O)-N(R13)2.
In certain embodiments, R2 is -pyridyl substituted with 0, 1, 2 or 3 substituents R , wherein R is independently selected from R and R7 (both as defined above) and wherein R1, R3, R4, R5, W, and X are as defined above.
In certain embodiments, R2 is — pyrid-2-yl substituted with one substituent R8 in position 4, that is independently selected from -O-Cι-3-alkyl, -S-Ci-3-alkyl, -Ci-3- alkyl-OH, -SO2-NH2, and -N-linked-heterocycloalkyl, including R8 being piperazinyl with 0, 1, 2 or 3 substituents being independently selected from R6 and R7.
Another aspect of the invention relates to a compound having a structure represented by formula (I) or any tautomeric or stereoisomeric form thereof, wherein R3 is hydrogen, and wherein R1, R2, R4, R5, V, W, and X are as defined above.
Another aspect of the invention relates to a compound having a structure represented by formula (I) or any tautomeric or stereoisomeric form thereof, wherein W is a bond. In certain embodiments, R4 is selected from — aryl and — heteroaryl, and is substituted with 0, 1, 2, 3, 4, or 5 substituents R9, wherein R9 is independently selected from R6 and R7, and wherein R1, R2, R3, R5, V, and X are as defined above.
In certain embodiments, R4 is —phenyl substituted with 0, 1, 2, 3, 4, or 5 substituents R9.
In certain embodiments, R4 is —phenyl that is substituted with one substituent R9 in position 2 or 3 (i.e., in ortho or meta position), including embodiments where R8 is selected from -methyl, -O-Me, -CF3, N(R13)2, -NH-C(=X)-R13 and halogen.
In certain other embodiments, R4 is —phenyl that is substituted with two substituents R9 in positions 2,5 or 2,6, including embodiments where said two substituents R9 are independently selected from -methyl, -O-Me, -CF3, N(R13)2,, - NH-C(=X)-R13 and halogen, for example where R4 is —phenyl that is substituted with two -Cl substituents in positions 2,6 (i.e. where R4 is 2,6-dichlorophenyl).
Another aspect of the invention relates to a compound having a structure represented by formula (I) or any tautomeric or stereoisomeric foπn thereof, wherein R3 is selected from H and C|-3-alkyl, such as methyl, and W-R4 is selected from H, Ci-3-alkyl and -C(=O)-Ci-3-alkyl, and wherein R1, R2, R5, V, and X are as defined above. In certain embodiments, W-R4 is selected froin H, methyl or C(=O)-CH3. In particular embodiments, R3 and W-R4 are both hydrogen. In alternative embodiments, R5 is selected from: a branched C4-C6-alkyl or Cs-C7-cycloalkyl. In particular such embodiments, R5 is selected from: tBu, cyclopentyl, and cyclohexyl, including embodiments where R3 and W-R4 are both hydrogen.
In an alternate aspect of the invention, W-R is a halogen, including bromine, and where R1, R2, R3, R5, V, and X are as defined above
Indeed, it will be well known to a person of ordinary skill in the art in light of the instant invention, that specificity of compounds of the invention to particular kinase classes or kinases can be changed by modification of the substituents attached to the pyridopyrimidone framework (see, for example, the patent applications cited in "Background to the Invention" or Mclnnes et al. "Strategies for the Design of Potent and Selective Kinase Inhibitors" Current Pharmaceutical Design, Volume 11, Number 14, May 2005, pp. 1845-1863(19)). For example, by having small groups or hydrogen at R3 and W-R4, as described for certain embodiments above, specificity for CDKs can be increased.
Another aspect of the invention relates to a compound having a structure represented by formula (I) or any tautomeric or stereoisomer^ form thereof, wherein X is =O, and wherein R1, R2, R3, R4, R5. V, and W are as defined above.
Another aspect of the invention relates to a compound having a structure represented by formula (I) or any tautomeric or stereoisomeric form thereof, wherein Rs is selected from R10 and phenyl, in each case substituted with 0, 1, 2, or 3 substituents R16, wherein R16 is independently selected from R6 and R7, and wherein R1, R2, R3, R4, V, W and X are as defined above.
In certain embodiments, R5 is — C|.6-alkyl, or
Figure imgf000033_0001
substituted with 0, 1 or 2 substituents R16, including embodiments wherein R is —methyl, -butyl or — t- butyl. In certain other embodiments, R5 is — Ci_s-cycloalkyl substituted with 0, 1 or 2 substituents R16, including embodiments wherein R5 is -cyclopentyl.
Those skilled in the art will recognize that all specific combinations of the individual possible residues of the variable regions of the compounds as disclosed herein, i.e. R1, R2, R3, R4, and V, W and X are within the scope of the invention.
In one embodiment of the. present invention, compounds of the invention have a molecular weight of between 190 and 1000, particularly between 270 and 800, more particularly between 370 and 600, and even more particularly between 400 and 550. In certain embodiments, compounds of the invention have one or more of the following characteristics: (i) not more than 5 hydrogen bond donors, (ii) not more than 10 hydrogen bond acceptors, and (iii) not more than 10 rotatable bonds (excluding bonds to terminal atoms). In certain embodiments, the compounds of formula (I) are in accordance with Lipinski's "Rule of Five" (Lipinski, Adv. Drug Del. Rev. 1997; 23: 3), by having a molecular weight below 500, not more than 5 hydrogen bond donors, not more than 10 hydrogen bond acceptors, and a cLogP value between -2 and 5.
In one embodiment of the present invention, compounds of the invention are inhibitors of the activity of B-Raf. In certain embodiments, the compounds of the present invention inhibit B-Raf activity with IC50 values below 1 μM, such IC50 value being determined in accordance with the B-Raf inhibition assay described in the examples shown below. In certain embodiments, the IC50 value is below 0.5 μM, below 0.2 μM, below 0.1 μM, or below 0.01 μM.
In another embodiment of the present invention, compounds of the invention are inhibitors of the activity of p38. In certain embodiments, the compounds of the present invention inhibit p38 activity with IC50 values below 1 μM. In certain embodiments, the IC50 value is below 0.5 μM, below 0.2 μM, below 0.1 μM, or below 0.01 μM.
In one embodiment of the present invention, compounds of the invention have a purity of more than 90%, more than 95%, more than 98%, or more than 99%. Such compounds may exist in one or more crystalline foπns, including two or more polymorphic forms, and may exist as dry solids or as solvates including defined amounts of solvents, including hydrates including defined amounts of water.
In another embodiment, compounds of the invention are the planned and deliberate products of a synthetic chemistry scheme, i.e., produced by specific and planned chemical processes conducted in reaction vessels, and not by degradation, metabolism or fermentation, or produced as impurities or by-products in the synthesis of other compounds.
In certain embodiments, compounds of the invention are purified or isolated, e.g., to have a purity of at least 80%, preferably at least 90%, more preferably at least 95%, such as at least 97%, at least 98% or even at least 99%. Purity, as used herein, can refer to either absolute or relative purity. Absolute purity refers to the amount of a compound of the invention obtained as the product of a synthetic chemistry scheme, either before or after one or more purification steps. Relative purity refers to the amount of a compound of the invention relative to one or more impurities such as by-products, degradation products (e.g., metabolites, products of oxidation or hydrolysis, etc.) and/or compounds that degrade to form a compound of the invention (e.g., precursors or prodrugs), e.g., that may be present in the product of a synthetic chemistry scheme. Thus, absolute purity refers to the amount of a compound relative to all others, while relative purity is generally unaffected by the addition of unrelated compounds, such as excipients, stabilizers, or other medicaments for conjoint administration. Purity can be assessed based upon weight, volume or molar ratios of one compound relative to others. Purity can be measured by a variety of analytical techniques, including elemental abundance, UV-visible spectrometry, HPLC, GC-MS, NMR, mass spectrometry, and thin layer chromatography, preferably by HPLC, GC-MS, or NMR.
Yet another aspect of the invention relates to prodrugs of a compound described above.
Particular embodiments
In a particular aspect, the invention relates to a compound represented by a structure of formula (Ia)
Figure imgf000035_0001
(Ia) or any tautomeric or stereoisomeric form thereof, wherein I and m are independently selected from 0, 1, 2, 3, 4, and 5; and
R5 is C|.6-alkyl substituted with 0, 1, 2 or 3 substituents R16, wherein R8 and R9 are as defined above.
In certain embodiments, I is 1 or 2, and in particular embodiments, I is 1 , in both cases including embodiments wherein R8 is independently selected from optionally substituted — O-C1-3-alkyl, optionally substituted— S-C ι-3-alkyl, optionally substituted -Ci-3-alkyl-OH, -SO2-NH2, and optionally substituted -N-linked- heterocycloalkyl, including embodiments, where I is 1 with R8 being at the 3- or 4- position of the phenyl ring. Such embodiments include, but are not limited to, compounds represented by formula (Ia) where R is-OMe, -0-CH2-CH2-NMe2, or — CH2-OH in 3-position of the phenyl ring, or where R8 is 4-methyl-piperazine at the 4-position of the phenyl ring.
In other certain embodiments, I is 1 or 2, and R8 is independently selected from -O-Ci-3-alkyl, -S-CI-3-alkyl, -Ci-3-alkyl-OH, -SO2-NH2, CN, halogen (e.g., chlorine or fluorine), — (CH2)n-N-linked-heterocycloalkyl and — (CH2)n-heteroaryl with n selected from 0, 1, 2, 3, 4 or 5, including embodiments when the N-linked- heterocycloalkyl is selected from morpholinyl, piperazinyl, pyrrolidinyl and azetidine, and the -heteroaryl is selected from pyrrole, pyrazole and triazole. In particular such embodiments, the alkyl, N-linked-heterocycloalkyl or -heteroaryl is substituted by 1 or 2 substituents. In more particular embodiments, n is 0 or 1 , including embodiments where R8 is a -(CH2)n-N-linked-heterocycloalkyl selected from morpholinyl and piperazinyl, optionally substituted by 1 or 2 substituents.
In particular other embodiments, I is 2 with R8 in position 3 and 4 (i.e., in meta and para positions). In certain such embodiments, Rs in position 4 (i.e., in para position) is a — (CH2)n-N-linked-heterocycloalkyl selected from morpholinyl and piperazinyl, optionally substituted by 1 or 2 substituents, where n is 0 or 1. In particular such embodiments, R8 in position 3 (i.e., in meta position) is selected from -CN, a halogen (e.g., chlorine or fluorine), — O-Ci-3-alkyl and -C]-3-alkyl-OH.
In certain embodiments, m is 1 or 2, including embodiments wherein R9 is independently selected from optionally substituted -Ci .6— alkyl, optionally substituted -C2-6-alkenyl, optionally substituted -C2-6-alkynyl, optionally substituted — O-Ci-e— alkyl, -CF3, -N(R13)2, -NH-C(=X)-R13, -NO2, and halogen, including embodiments where m is 2 with R9 being in 2,5- or 2,6-positions of the phenyl ring. Such embodiments include, but are not limited to, compounds represented by formula (Ia) where both R9 are independently selected from methyl, -O-Me, -CF3, -N(RI3)2, -NH- C(=X)-R13 and halogen, for example where both R9 are -Cl substituents in 2,6- positions of the phenyl ring. Further such embodiments include, but are not limited to, compounds represented by formula (Ia) where both R9 are independently selected from -Cl or a — F, substituted in the 2 and 6-positions of the phenyl ring, or where R9 in position 2 is chlorine and R9 in position 5 is -NH-C(=X)-R13 (with X and R13 as defined above). In certain other embodiments, m is 1 with R9 being in 2- or 3- position of the phenyl ring (i.e., in ortho or meta position). Such embodiments include, but are not limited to, compounds represented by formula (Ia) where R9 is selected from methyl, -O-Me, -CF3, -N(RI3)2, -NH-C(=X)-R13 and halogen, for example where R9 is -Cl or -F in 2-position of the phenyl ring, or where R9 is -NH- C(=X)-R13 in position 3 (with X and R13 as defined above).
In certain embodiments, R5 is -Ci-4-alkyl substituted with 0 or 1 substituent R16, including embodiments wherein R5 is methyl.
In certain embodiments of Formula I, the compound is selected from:
6-(2,6-Dichlorophenyl)-2-(3-hydroxymethylphenylamino)-8-methoxy- pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(3-methylthiophenylamino)- pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(3-methoxyphenylamino)-pyrido[2,3- d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(3-sulfamoylphenylamino)-pyrido[2,3- d]pyrimidin-7-one,
6-(2-Chlorophenyl)-8-methoxy-2-(3-hydroxvmethylphenylamino)- pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chlorophenyl)-8-methoxy-2-(3-sulfamoylphenylamino)-pyrido[2,3- d]pyrimidin-7-one,
6-(2-Chlorophenyl)-8-methoxy-2-(3-methoxyphenylamino)-pyrido[2,3- d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-8-methoxy-2-(4-(2-dimethylaminoethoxy)- phenylamino)-pyrido[2,3-d]pyrimidin-7-one,
6-(5-Benzoylamino-2-chloro-phenyl)-8-methoxy-2-(4-(4-methylpiperazino)- phenylamino)-pyrido[2,3-d]pyrimidin-7-one, 6-(2.6-Dichlorophenyl)-8-methoxy-2-(3-(2- hydroxyethylsulfonyl)phenylamino)-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(4-methylsulfonylphenylamino)- pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-((4-methoxycarbonyl-3-methylpyrrol- 3 -yl)amino) -pyrido [2 ,3 -d]pyrimidm-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(pyrid-4-ylamino)-pyrido[2,3- d]pyrimidin-7-one5
6-(2,6-Dichlorophenyl)-8-methoxy-2-(4-(4-methylpiρerazino)- phenylamino)-pyrido[2,3-d]pyrimidin-7.-one,
6-(2-Chloro-6-fluorophenyl)-8-methoxy-2-(3-hydroxymethylphenylamino)- pyrido[2,3-d]pyrimidin-7-one,
6-(2,4-Dichlorophenyl)-8-methoxy-2-(4-(2-dimethylaminoethoxy)- phenylamino)-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichloroρhenyl)-8-methoxy-2-(4-(2-hydroxyethyl)-phenylamino)- pyrido[2,3-d]pyrimidin-7-one,
6-(3,4-Dichlorophenyl)-8-methoxy-2-(4-(4-methylpiperazino)- phenylamino)-pyrido[2,3-d]pyrimidin-7-one5
6-(2,4-Dichlorophenyl)-8-methoxy-2-(4-(4-methylpiperazino)- phenylamino)-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chlorophenyl)-8-(2-methoxyethoxy)-2-phenylamino)-pyrido[2,3- d]pyrimidin-7-one5
6-(2,6-Dichlorophenyl)-8-methoxy-2-(3-(pyrrolidin-l - yl)methylphenylamino)-pyrido[2,3-d]pyriτnidin-7-one,
6-(2-Chloτophenyl)-8-methoxy-2-phenylamino-pyrido[2,3-d]ρyrin-ιidin-7- one,
6-(5-Amino-2-chlorophenyl)-8-methoxy-2-phenylaτnino-pyrido[2,3- d]pyriτnidin-7-one, 6-(2,6-Dichlorophenyl)-8-methoxy-2-(4-(2-dimethylaminoethoxy)- phenylamino)-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-8-methoxy-2-(4-(4-methylpiperazino)- phenylamino)-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-5-(pyτid-4-ylcarbonylatnino)phenyl)-8-methoxy-2-(4-(4- methylpiperazino)-phenylamino)-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-(2-fluoro-5-(hydroxymethyl)phenylaπiino)-8- raethoxy-pyrido[2,3-d]pyriraidin-7-one,
6-(3-Benzoylaminophenyl)-8-methoxy-2-phenylamino)-pyrido[2,3- d]pyrimidin-7-one,
6-(5-Benzoylamino-2-chloro-phenyl)-8-methoxy-2-phenylamino)- pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-8-methoxy-2-(3-sulfamoylphenylamino)- pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-5-(pyrid-3-ylcarbonylamino)phenyl)-8-methoxy-2- phenylamino)-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-5-(dimethylacetylamino)phenyl)-8-methoxy-2-phenylamino)- pyrido[2,3-d]pyrimidin-7-one,
6-(5-Benzoylaπαino-2-chloro-phenyl)-8-methoxy-2-(2-methoxyethyl)amino)- pyrido[2,3-d]pyrimidin-7-one,
8-(4-Aminobutoxy)-6-(2,6-dichlorophenyl)-2-phenylamino-pyrido[2,3- d]pyrimidin-7-one,
6-(2-Chloro-5-((3-trifluoromethyl)benzoylamino)phenyl)-8-methoxy-2- phenylamino)-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-5-(3-chlorobenzoylamino)phenyl)-8-methoxy-2-phenylamino)- pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-5-(4-chlorobenzoylamino)phenyl)-8-methoxy-2-phenylamino)- pyrido[2,3-d]pyrimidin-7-one, 6-(2,6-Dimethylphenyl)-8-methoxy-2-(4-(4-methylpiperazino)- phenylamino)-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-methoxyphenyl)-8-methoxy-2-(4-(4-methylpiperazino)- phenylamino)-pyrido[2,3-d]pyrimidin-7-one,
8-(4-Aminobutoxy)-6-(236-dichlorophenyl)-2-(3-sulfamoylphenylamino)- pyrido [2,3 -d]pyrimidin-7-one,
8-(4-Aminobutoxy)-6-(2:,6-dichlorophenyl)-2-(3-methoxyphenylamino)- pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-8-dimethylmethoxy-2-(4-(4-methylpiperazino)- phenylamino)-pyrido[2,3-d]pyrimidin-7-one,
2-(3-Hydroxymethylphenylamino)-8-methoxy-6-phenyl-pyrido[2,3- d]pyrimidin-7-one,
6-(235-Dimethoxyphcnyl)-2-(3-hydroxymcthylphcnylamino)-8-mcthoxy- pyri do [2, 3 -d] pyrimidin-7-onc,
6-(2,6-Dichlorophenyl)-2-((2-methyl-5-hydroxymethylphenyl)-amino)-8- methoxy-pyrido[2,3-d]pyrimidin-7-one,
2-Amino-6-(2,6-dichlorophenyl)-8-methoxy-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(4-methylpiperidino-amino)- pyrido[2,3-d]pyrimidin-7-onc,
6-(2,6-Dichlorophenyl)-8-methoxy-2-methoxyethylamino-pyrido[2,3- d]pyrimidin-7-one,
6-(2-Chlorophenyl)-8-cyclopropylmethoxy-2-phenylamino-pyrido[2,3- d]pyrimidin-7-one,
2-(4-(2-Diτnethylaminocthoxy)-6-(2-mcthoxyphcnyl)-phcnylamino)-8- methoxy-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-8-ethoxy-2-(4-(4-methylpiperazino)- phenylamino)-pyrido[2,3-d]pyrimidin-7-one, 6-(2-Chloro-6-fluorophenyl)-8-cyclopropylmethoxy-2-(4-(4- methylpiperazino)-phenylamino)-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Fluoro-6-trifluoromethyl-phenyl)-8-methoxy-2-(4-(4- methylpiperazino)-phenylamino)-pyrido[2,3-d]pyrimidin-7-one5 and
2-(5-Carboxy-l-methyl-pyrrol-3-yl)-amino-6-(2,6-dichlorophenyl)-8- methoxy-pyrido [2, 3 -d] pyrimidin-7-one,
8-(3-Aminopropyl)oxy-6-(2,6-dichlorophenyl)-2-phenylamino-pyrido[2,3- d]pyrimidin-7-one,
8-(5-Aminopentyl)oxy-6-(2,6-dichlorophenyl)-2-phenylamino-pyrido[2,3- d]pyrimidin-7-one,
8-(3-Acetylaminopropyl)oxy-6-(2,6-dichlorophenyI)-2-phenylamino- pyrido[2,3-d]pyrimidin-7-one,
8-(2-(2-Aminoethyloxy)ethyl)oxy-6-(2,6-dichloroρhenyl)-2-phenylamino- pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-5-acetylaminophenyl)-8-methoxy-2-phenylamino)-pyrido[2,3- d]pyrimidin-7-one,
6-(2>5-Dimethoxyphenyl)-8-methoxy-2-phenylamino)-pyrido[2,3- d]pyrimidin-7-one,
8-Methoxy-2-phenylamino-6-phenylaminocarbonyl-pyrido[2,3-d]pyrimidin- 7-one,
6-(3-Acetylaminophenyl)-8-methoxy-2-phenylaτnino)-pyrido[2,3- d]pyrimidin-7-one,
6-(2-Chlorophenyl)-8-(l ,l-dimethyl)ethyloxy-2-phenylaτnino-pyrido[2,3- d]pyrimidin-7-one,
2-(3-Aminosulfonlyρhenyl)-amino-6-(3,4-dichloiOphenyl)-8-methoxy- pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chlorophenyl)-8-(l -methylethyl)oxy-2-phenylamino)-pyrido[2,3- d]pyrimidin-7-one, 8-(4-Aminobutyl)oxy-6-(2,6-dichlorophenyl)-2-phenylamino-pyrido[2,3- d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-(5-(2-dimethylaminoethyl)aminocarbonyl-l- methyl-pyrrol-3-yl)-amino-8-methoxy-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-(2-methoxyethyl)oxy-2-(3-methoxyphenyl)amino- pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dimethylphenyl)-8-methoxy-2-(3-methoxyphenyl)-amino-pyrido[2,3- d]pyrimidin-7-one,
8-(2-Aminoethyl)oxy-6-(2,6-dichlorophenyl)-2-(3-methoxyphenyl)amino- pyrido[2,3-d]pyrimidin-7-one,
8-(3-Aminopropyl)oxy-6-(2,6-dichlorophenyl)-2-(3-methoxyphenyl)amino- pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dimethylphenyl)-8-τnethoxy-2-(3-sυlfamoylphenylamino)- pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-(2-hydroxyethyl)oxy-2-(3-methoxyphenyl)amino- pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-(2-methylaminoethyl)oxy-2-(3- methoxyphenyl)amino-pyrido[2,3-d]pyrimidin-7-onc,
6-(2-Chloro-6-fluorophenyl)-8-(2-(S)-2,3-dihydroxypropyl)oxy-2-(3- methoxyphenyl)amino-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-8-(2-(R)-2,3-dihydroxypropyl)oxy-2-(3- methoxyphenyl)amino-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-(2-diraethylaminoethyl)oxy-2-(3- methoxyphenyl)aτnino-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-(2-dimethylaminopropyl)oxy-2-(3- methoxyphenyl)amino-pyrido[2,3-d]ρyrimidin-7-one,
6-(2,6-Dimethylphenyl)-8-methoxy-2-(5-(methoxycarbonyl-l-methyl-pyrrol- 3-yl)-amino-pyrido[2,3-d]pyrimidin-7-one, 2-Cyclopropylcarbonylamino-6-(2,6-dichlorophenyl)-8-methoxy-pyrido[2,3- d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-(5-(2-diethylaminoethyl)aminocarbonyl-l-methyl- pyrrol-3 -yl)-amino-8-methoxy-pyrido[2 ,3 -d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-(5-(2-hydroxyethyl)aminocarbonyl-l-methyl- pyrrol-3-yl)-amino-8-methoxy-pyrido[2,3-d]ρyrimidin-7-one,
6-(2,6-Dimethylphenyl)-2-(5-(2-hydroxyethyl)aminocarbonyl-l-methyl- pyrrol-3-yl)-amino-8-methoxy-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dimethylphenyl)-2-(5-(2-diethylaminoethyl)aminocarbonyl-l-methyl- pyrrol-3-yl)-amino-8-methoxy-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-(isoxazol-3-yl)-amino-8-methoxy-pyrido[2,3- d]pyrimidin-7-one,
2-(4-Cyanophenyl)-amino-6-(2,6-dichlorophenyl)-8-methoxy-pyrido[2,3- d]pyrimidin-7-one,
6-(2,6-Dimethylphenyl)-8-methoxy-2-(5-(2-pyrrolidinoethyl)aminocarbonyl- l -methyl-pyrrol-3-yl)-amino-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(pyrazol-3-yl)-amino-pyrido[2,3- d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-(4-(2-hydroxyethyl)oxyphenyl)amino-8-methoxy- pyrido[2,3 -d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(l-thia-3,4-diazol-2-yl)-amino- pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(4-(2- pyrrolidinoethyl)oxyphenyl)amino-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-(4-(2-(3-(S)- hydroxypyrrolidino)ethyl)oxyphenyl)amino-8-methoxy-pyrido[2,3-d]pyrimidin-7- one,
6-(2,6-Dichlorophenyl)-2-(4-(2,3-dihydroxypropyl)oxyphenyl)amino-8- methoxy-pyrido[2,3-d]pyrimidin-7-one, 6-(2,6-Dichloroρhenyl)-8-methoxy-2-(5-(2-pyrrolidinoethyl)aminocarbonyl- l-methyl-pyrrol-3-yl)-amino-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(5-(2- pyrrolidinopropyl)aminocarbonyl-l-methyl-pyrrol-3-yl)-amino-pyrido[2,3- d]pyrimidin-7-one,
2-But-2-enoylamino-6-(2,6-dimethylphenyl)-8-methoxy-pyrido[2,3- d]pyrimidin-7-one,
2-(4-cyanomethylphenyl)-amino-6-(2,6-dichlorophenyl)-8-τnethoxy- pyrido [2,3 -d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(4-morpholinophenyl)-amino- pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-(2-methoxyethyl)oxy-2-(5-(2- pyrrolidinoethyl)aminocarbonyl-l -methyl -pyrrol-3-yl)-amino-pyrido[2,3- d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(4-τnorpholinomethylphenyl)-amino- pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(4-pyrrolidinomethylphenyl)-amino- pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-hydroxyethylamino-8-methoxy-pyrido[2,3- d]pyrimi di n-7- one,
6-(2,6-Dichlorophenyl)-8-(2-(S)-2,3-dihydroxypropyl)oxy-2-(5-(2- pyrrolidinoethyl)aminocarbonyl-l-methyl-pyrrol-3-yl)-amino-pyrido[2,3- d]pyrimidin-7-one,
6-(2,6-DichlorophenyI)-8-methoxy-2-(4-(l ,2,4-triazol-l-yl)methylphenyl)- amino-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(4-pyrrolidinophenyl)-amino- pyrido[2,3-d]pyrimidin-7-one,
6-(5-Benzoylamino-2-chloro-phenyl)-8-(2-methoxyethyl)oxy-2-(4-(4- methylpiperazin.o)-phenylamino)-pyrido[2,3-d]pyrimidin-7-one, 8-(2-Methoxyethyl)oxy-2-(4-(4-methylpipcrazino)-6-(5-(3- trifluoromethylbenzoyl)amino-2-cMoro-phenyl)-phenylamino)-pyrido[2,3- d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-(2-(R)-2,3-dihydroxypropyl)oxy-2-(5-(2- pyrrolidinoethyl)aminocarbonyl- 1 -methyl-pyrrol-3 -yl)-amino-pyrido [2,3- d]pyrimidin-7-one,
2-((2-(S)-2-Amino-3-methylbutanoyloxy)ethyl)-amino-6-(2,6- dichlorophenyl)-8-methoxy-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-raethoxy-2-(4-(2-oxopyrrolidino)phenyl)-amino- pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(4-methylsulfonylaminophenyl)- amino-pyrido[2,3-d]pyritnidin-7-one,
2-(5-(2-(2-(S)-2-Amino-3-mcthylbutanoyloxy)ethyl)aminocarbonyl-l- methyl-pyrrol-3-yl)-amino-6-(2,6-dichlorophenyl)-8-methoxy-pyrido[2,3- d]pyrimidin-7-one,
2-Cyclopropylamino-6-(2,6-dichlorophenyl)-8-methoxy-pyrido[2)3- d]pyrimidin-7-one,
6-(2,6-Dimethylphenyl)-8-methoxy-2-pyrid-3-ylamino-pyrido[2,3- d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(4-(2- pyrrolidinoethylaminocarbonylmethyl)phenyl)-aτnino-pyrido[2;i3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-(5-(N-(2-hydroxyethyl)-N-methyl- amino)carbonyl-l-methyl-pyrrol-3-yl)-amino-8-methoxy-pyrido[2,3-d]pyrimidin-7- one,
6-(2,6-Dichlorophenyl)-2-(5-(2-(R)-2,3-dihydroxyethylamino)carbonyl-l- methyl-pyrrol-3-yl)-amino-8-methoxy-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-(5-(2-(S)-2,3-dihydroxyethylamino)carbonyl-l- methyl-pyrrol-3-yl)-amino-8-methoxy-pyrido[2,3-d]pyrimidin-7-one5 6-(2,6-Dichlorophenyl)-8-methoxy-2-(3-methylsulfonylaminophenyl)- amino-pyrido[2:>3-d]ρyrimidin-7-one!
6-(2,6-Dichlorophenyl)-8-methoxy-2-(4- methylsulfonylaminomethylphenyl)-amino-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-8-(2-methoxyethyl)oxy-2-(4- morpholinophenyl)-amino)-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-(3-ethylaminosulfonylphenyl)-amino-8-methoxy- pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-(3-diethylaminosulfonylphenyl)-amino-8- methoxy-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-dicHorophenyl)-8-methoxy-2-(4-(pyrazol-l-ylmethyl)phenyl)-amino- pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-dichlorophenyl)-8-methoxy-2-(4- (methylaminosulfonylmethyl)phenyl)-amino-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-(3-(2-hydroxyethyl)aminosulfonylphenyl)-amino- 8-methoxy-pyrido[2:3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-(3-morpholinosulfonylphenyl)-amino-8-raethoxy- pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-8-ethoxy-2-(4-morpholinophenyl)-amino)- pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-8-(cyclopropylmethyl)oxy-2-(4- morpholinophenyl)-amino)-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(3-tetrazol-5-ylphenyl)-amino- pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-metlioxy-2-(3-methylaminocarbonylphenyl)- amino-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-8-(pyrid-3-ylmethyl)oxy-2-(4- morpholinophenyl)-amino)-pyrido[2,3-d]pyrimidin-7-one, 2-(3-Chloro-4-trifluoromethylphenyl)-amino-6-(2,6-dichlorophenyl)-8- methoxy-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(3-(l,2,4-triazol-l-ylmethyl)phenyl)- amino-pyrido [2 ,3 -d] pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(pyrimidin-4-yl)-amino-pyrido[2,3- d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-8-metlioxy-2-(4-morpholinoph.enyl)-amino)- pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(5-morpholinocarbonyl-l-methyl- pyrrol-3-yl)-aτnino-pyrido[2,3-d]pyτimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(4-N-(2-hydroxyethyl)-N-methyl- aminocarbonyl-thiazol-2-yl)-amino-pyrido[2,3-d]pyrimidin-7-one,
6-{2,6-Dichlorophenyl)-8-methoxy-2-(5-N-(2-hydroxyethyl)-N-methyl- aminocarbonyl-thiophen-3-yl)-amino-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(5-N-(2-(2,2- dimethylpropanoyl)oxyethyl)-N-methyl-aminocarbonyl- 1 -methyl-pyrrol-3 -yl)- amino-pyiido[2,3-d]pyrimidin-7-one,
2-(5-N-(2-(Benzoyloxyethyl)-N-methyl-arninocarbonyl-l-methyl-pyrrol-3- yl)-amino-6-(2,6-dichlorophenyl)-8-methoxy-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(5-(4-methylpiperazino)carbonyl- thiophen-3-yl)-amino-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-2-(4-moφholinophenyl)-8-(tetrahydropyran-4- ylmethyl)oxy-amino)-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-(4-hydroxymethylphenyl)-amino-8-methoxy- pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-phenylmethylamino-pyrido[2,3- d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-pyrid-3-ylmethylamino-pyrido[2,3- d]pyrimidin-7-one, 6-(2,6-Dichloroρhenyl)-8-methoxy-2-pyrid-4-ylmethylamino-pyrido[233- d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-8-(2-(S)-2,3-dihydroxypropyl)oxy-2-(4- moφholinophenyl)-amino)-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-8-(3-(R)-pyrrolidin-3-ylmethyl)oxy-2-(4- morpholinophenyl)-amino)-pyrido [2 , 3 -d] pyrimi din-7 -one,
6-(2,6-Dichlorophenyl)-2-(4-methylphenyl)-amino-8-methoxy-pyrido[2,3- d]pyrimidin-7~one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(4-(5-methyl-l,2,4-triazol-3- yl)methylphenyl)-amino-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-8-(2-(R)-2,3-dihydroxypropyl)oxy-2-(4- morpholinophenyl)-amino)-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-2-phenylamino-8-(pyrid-3-ylmethyl)oxy- pyrido[2,3 -d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(5-N-(2-methoxyethyl)-N-methyl- aminocarbonyl-l-methyl-pyrrol-3-yl)-amino-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-8-(3-(S)-pyrrolidin-3-ylmethyl)oxy-2-(4- morpholinophenyl)-amino)-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-(3-((2- hydroxyethylamino)sulfonylmethyl)phenyl)-amino-8-methoxy-pyrido[2,3- d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(3-methylsυlfonylphenyl)-amino- pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-(4-(3-hydroxypropyl)thiophenyl)-amino-8- methoxy-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(5-N-(2-(pyridin-3- ylcarbonyloxy)ethyl-N-methyl-aminocarbonyl-l-methyl-pyrrol-3-yl)-amino- pyrido[2,3-d]pyrimidin-7-one, 6-(2>6-Dichlorophenyl)-2-(4-(3-hydroxypropyl)sulfonylphenyl)-amino-8- methoxy-pyrido[2,3-d]pyriraidin-7-one,
6-(2,6-Dichloropheπyl)-8-methoxy-2-(3-methyliminosulfonylphenyl)-amino- pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-(4-methoxycarbonylphenyl)-amino-8-methoxy- pyτido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(5-(4-methylpiperazino)carbonyl-l- methyl-pyrrol-3-yl)-amino-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-2-phenylamino-8-(tetrahydropyran-4- ylmethyl)oxy-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-ρhenylamino-8-(pyrid-3-ylmethyl)oxy-pyrido[2,3- d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(4-(4-(l- methylethyl)piperazinomethylphenyl)-amino-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-(4-diethylaminomethylphenyl)-amino-8-methoxy- pyrido[2,3-d]pyrimidin-7-one,
2-(4-(4-hydroxy- 1 -aza-cyclobutyl)methylphenyl)-amino-6-(2,6- dichlorophenyl)-8-methoxy-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-(4-(2-(S)-hydroxymethyl- pyrrolidinomethyl)phenyl)-amino-8-methoxy-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-(2-methoxyethyl)oxy-2-(5-N-(2-methoxyethyl)-N- methyl-aminocarbonyl-l -methyl-pyrrol-3-yl)-aτnino-pyrido[2;>3-d]pyrimidin-7-one3
6-(2,6-Dichlorophenyl)-8-ethoxy-2-(5-N-(2-methoxyethyl)-N-methyl- aminocarbonyl-l-methyl-pyrrol-3-yl)-aτnino-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-8-cyclobutylmethyloxy-2-(4- morpholinophenyl)-amino)-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-8-cyclopentylmethyloxy-2-(4- morpholinophenyl)-amino)-pyrido[2,3-d]pyrimidin-7-one. 6-(2,6-Dichlorophenyl)-8-methoxy-2-phenylamino-pyrido[2,3-d]pyrimIdin- 7-one,
2-(4-(l,3,4-triazol-l-yl)methylphenyl)-amino-6-(2>6-dichlorophenyl)-8- methoxy-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-cyclopropylmethyloxy-2-(5-N-(2-methoxyethyl)- N-methyl-aminocarbonyl-l-methyl-pyrrol-3-yl)-amino-pyrido[2,3-d]pyrimidin-7- one,
6-(2,6-dichlorophenyl)-2-(5-N-(2-methoxyethyl)-N-methyl-aminocarbonyl- l-methyl-pyrrol-3-yl)-amino-8-(tetrahydropyran-4-ylmethyl)oxy-pyrido[2,3- d]pyrimidin-7-one,
2-(4-(24iydroxyethylamino)methylphenyl)-amino-6-(2,6-dichlorophenyl)-8- methoxy-pyτido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-(4-(3-(R)-hydroxypyrrolidinomethyl)phenyl)- amino-8-methoxy-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-(4-(3-(S)-hydroxypyrrolidinomethyl)phenyl)- amino-8-methoxy-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-(3-(3-(S)-hydroxypyrrolidinomethyl)phenyl)- amino-8-methoxy-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-(3-(3-(R)-hydroxypyrrolidinomethyl)phenyl)- amino-8-methoxy-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-(3-(3,3-difluoropyrrolidinomethyl)phenyl)-amino- 8-methoxy-pyrido[2,3-d]pyriτnidin-7-one,
6-(2,6-Dichlorophenyl)-8-iτιethoxy-2-(4-piperazinophenyl)-amino- pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(5-piperazinocarbonyl-l-methyl- pyrrol-3-yl)-amino-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-phenylamino-8-(tetrahydropyran-4-ylmethyl)oxy- pyrido[2,3-d]pyrimidin-7-one, 6-(2,6-Dichlorophenyl)-8-methoxy-2-(4-(4-(l- methylethyl)piperazinophenyl)-amino-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(5-(4-(2- methoxy)ethyl)piperazinocarbonyl-l-methyl-pyrrol-3-yl)-aτnino-pyrido[2,3- d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-(4-(4-ethylpiperazino)phenyl)-amino-8-methoxy- pyridoPjS-dJpyrimidin-T-one,
6-(2,6-Dichlorophenyl)-2-(2-fluorophenyl)amino-8-methoxy-pyrido[2,3- d]pyrimidin-7-one,
2-(4-Bromophenyl)amino-6-(2,6-dichlorophenyl)-8-methoxy-pyrido[2,3- d]pyrimidin-7-one,
2-(4-Acetylphenyl)amino-6-(2,6-dichlorophenyl)-8-τnethoxy-pyrido[2,3- d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-2-(5-N-(2-hydroxyethyl)-N-methyl- aminocarbonyl-1 -methyl -pyrrol-3-yl)-amino-8-methoxy-pyrido[2,3-d]pyrimidin-7- one,
6-(2-Chloro-6-fluorophenyl)-2-(5-N-(2-methoxyethyl)-N-rnethyl- aminocarbonyl-1 -methyl -pyrrol-3-yl)-amino-8-methoxy-pyτido[2,3-d]pyriτnidin-7- one,
6-(2,6-Dichlorophenyl)-8-(2-methoxyethyl)oxy-2-(4-piperazinophenyl)- amino-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-(4-(3,3-difluoropyrrolidinomethyl)phenyl)-amino- 8-methoxy-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-8-methoxy-2-(5-moφholinocarbonyl-l-methyl- pyrrol-3-yl)-amino-pyrido[2,3-d]pyriraidin-7-one,
6-(2-Chloro-6-fluorophenyl)-8-methoxy-2-(4-piρerazinophenyl)-amino- pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-2-(3-chloro-4-piperazinophenyl)-amino-8- iτiethoxy-pyrido[2,3-d]pyrimidin-7-one, 6-(2,6-Dichlorophenyl)-8-methoxy-2-(4-(4- methylsulfonyl)piperazinophenyl)-amino-pyrido[2,3-d]pyrimidin-7-one,
2-(4-(l-azacyclobutyl)methylphenyl)-amino-6-(2,6-dichlorophenyl)-8- methoxy-pyrido [2,3 -d]pyrimidin-7 -one,
6-(2,6-DichIorophenyl)-8-(tetrahydropyran-4-ylmethyl)oxy-2-(4- piperazinophenyl)-amino-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-8-methoxy-2-(4-(4-( 1 - methylethyl)piperazino)phenyl)-amino-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(4-(4-propylpiperazino)phenyl)-amino- pyrido [2 , 3 - d]pyrimidin-7 -one,
6-(2,6-Dichlorophenyl)-8-(2-methoxyethyl)oxy-2-(4-(4-( 1 - methylethyl)piperazino)phenyl)-amino-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-(tetrahydropyran-4-ylmethyl)oxy-2-(4-(4-(l- methylethyl)piperazino)phenyl)-amino-pyrido[2,3-d]pyrimidin-7-one,
2-(4-(4-propylpiperazino)phenyl)-amino-6-(2,6-dichlorophenyl)-8-(2- methoxyethyl)oxy-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-8-methoxy-2-(5-piperazinocarbonyl-l-methyl- pyrrol-3-yl)-amino-pyrido[2,3-d]pyrimidin-7-one,
2-(4-(methoximino)methylphenyl)-amino-6-(2,6-dichlorophenyl)-8- methoxy-pyrido[2,3-d]pyrimidin-7-one,
2-(4-(hydroximino)methylphenyl)-amino-6-(2,6-dichlorophenyl)-8-τnethoxy- pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-2-(3-fluoro-4-piperazinophenyl)-amino-8- methoxy-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-8-(3-(S)-l-methyl-pyrrolidin-3-yl)methoxy-2- (4-moφholinophenyl)-amino-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-8-methoxy-2-(3-cyano-4-piperazinophenyl)- amino-pyrido[2,3-d]pyriinidin-7-one, 6-(2-Chloro-6-fluorophenyl)-8-methoxy-2-(3-methoxy-4-piperazinophenyl)- amino-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-8-(tetrahydropyran-4-ylmethyl)oxy-2-(3- methoxy-4-piperazinophenyl)-amino-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-8-(2~methoxyethyl)oxy-2-(4- piperazinophenyl)-amino-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-8-methoxy-2-(4-(4-propylpiperazino)phenyl)- amino-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-8-methoxy-2-(3-hydroxymethyl-4- piperazinophenyl)-amino-pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-(3-hydroxyphenyl)-amino-8-methoxy-pyrido[2a3- d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-8-methoxy-2-(5-N-(2-(pyridin-3- ylcarbonyloxy)ethyl-N-methyl-aminocarbonyl-l-metliyl-pyrrol-3-yl)-amino- pyrido[2,3-d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(4-(4-methyl-l ,4- • diazacycloheptyl)phenyl-)-amino-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-8-methoxy-2-(4-(piperidin-4-yl)phenyl)-amino- pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorc»phenyl)-8-methoxy-2-(4-(4- methylsulfonylpiperazino)phenyl)-amino-pyrido[2,3-d]pyrimidin-7-one,
6-(2-Chloro-6-fluorophenyl)-2-(5-(piperazinocarbonyl)-l-methyl-pyrrol-3- yl)-amino-8-(tetrahydropyran-4-ylmethyl)oxy -pyrido[2,3-d]pyrimidin-7-one,
8-Cyclopentyloxy-2-phenylamino)-pyrido[2,3-d]pyrimidin-7-one,
8-Cyclopentyloxy-2-(4-morpholinophenyl)amino)-pyrido[2,3-d]pyrimidin-7- one,
6-(2,6-Dichlorophenyl)-8-methoxy-2-(3-methoxy-4-piperazino-phenyl)- amino-pyrido[2,3-d]pyrimidin-7-one, 2-(4-(2-(2-aminoethoxy)ethoxyphenyl)-amino-6-(2,6-dichlorophenyl)-8- methoxy-pyrido [2, 3 -d] pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-2-(3-hydroxymethyl-4-piperazino-phenyl)-amino-8- methoxy-pyrido [2,3 -d]pyrimidin-7-one,
6-(2,6-Dichlorophenyl)-8-(2-(S)-2,3-dihydroxypropyl)oxy-2-(4- morpholinophenyl)amino-pyrido [2,3 -d]pyrimidin-7-one,
6-(256-Dichlorophenyl)-8-methoxy-2-(5-(4-methylpiperazino)carbonyl-l- methyl-pyrrol-3-yl)-amino-pyrido[253-d]pyrimidin-7-one,
2-Amino-6-(2,6-dichlorophenyl)-8-(pyrid-3-yl)methoxy-pyrido[2,3- d]pyriraidin-7-one.
8-(I5I -Ditnethylethoxy)-2-(4-(4-methylpiperazino)phenyl)amino)- pyrido[2,3-d]pyrimidin-7-one,
8-Cyclopentyloxy-2-(4-(4-methylpiperazino)phenyI)amino)-pyrido[2,3- d]pyrimidin-7-one,
8-Cyclopentyloxy-2-(4-(2-dimethylaminoethoxy)phenyl)amino)-pyrido[2,3- d]pyrimidin-7-one,
8-Cyclohexyloxy-2-(4-(4-methylpiperazino)phenyl)amino)-pyrido[2,3- d]pyrimidin-7-one3
8-Cyclohexyloxy-2-(4-(2-dimethylaminoethoxy)phenyl)amino)-pyrido[2,3- d]pyrimidin-7-one5
8-Cyclopentyloxy-2-(4-piperazinophenyl)amino-pyrido[2,3-d]pyrimidin-7- one,
6-Bromo-8-(l5l-dimethylethoxy)-2-phenylamino)-pyrido[2,3-d]pyriπiidin-7- one,
6-(2-Chlorophenyl)-8-hydroxy-2-phenylamϊno-pyrido[233-d]pyrimidin-7- one, and,
6-(2-Chloro-6-fluorophenyl)-8-hydroxy-2-(4-(4- methylpiperazino)phenyl)amino)-pyrido[2,3-d]pyrimidin-7-one, where such compounds have the structures as disclosed in Table 1 below. In case of a discrepancy between the chemical name given above and the corresponding structure shown in Table 1 , the structure should be regarded as correct, and the name amended accordingly.
Formulations, Dosages and Applications
The present invention further provides a pharmaceutical composition including a compound as described above, or prodrug thereof, and a pharmaceutically acceptable diluent, excipient or carrier, including pharmaceutical compositions including a therapeutically effective amount of such compound or prodrug.
Formulations
The compositions of this invention can be formulated and administered to treat individuals in need by any means that produces contact of the active ingredient with the agent's site of action, such as a cell, in the body of an individual. They can be administered by any conventional means available for use in conjunction with pharmaceuticals, either as individual therapeutic active ingredients or in a combination of therapeutic active ingredients. They can be administered alone, but are generally administered with a pharmaceutically acceptable diluent, excipient or carrier selected on the basis of the chosen route of administration and standard pharmaceutical practice.
A pharmaceutical composition comprising less than a therapeutically effective amount of any of the compounds described above, or a prodrug thereof, may also be used, such as when used in combination with another pharmaceutical composition, such as an anti-cancer agent, so that such combination is therapeutically effective, or may be useful for prophylactic treatment.
Pharmaceutical compositions for use in accordance with the present invention may be formulated in conventional manner using one or more pharmaceutically acceptable diluents, excipients or carriers. The pharmaceutical compositions of the invention can be formulated for a variety of routes of administration, including systemic and topical or localized administration. Techniques and formulations generally may be found in Remington's Pharmaceutical Sciences, Meade Publishing Co., Easton, PA. As described in detail below, the pharmaceutical compositions of the present invention may be specially formulated for administration in solid or liquid form, including those adapted for the following: (1) oral administration, for example, drenches (aqueous or non-aqueous solutions or suspensions), tablets, capsules, boluses, powders, granules, pastes for application to the tongue; (2) parenteral administration, for example, by subcutaneous, intramuscular or intravenous injection as, for example, a sterile solution or suspension; (3) topical application, for example, as a cream, ointment or spray applied to the skin; or (4) intravaginally or intrarectally, for example, as a pessary, cream or foam. In certain embodiments, the pharmaceutical preparations may be non-pyrogenic, i.e., do not substantially elevate the body temperature of a patient.
Wetting agents, emulsifiers and lubricants, such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions.
Examples of pharmaceutically acceptable antioxidants include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
Formulations of the present invention include those suitable for oral, nasal, topical (including buccal and sublingual), rectal, vaginal and/or parenteral administration. The formulations may conveniently be presented in unit dosage foπn and may be prepared by any methods well known in the art of pharmacy. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated, as well as the particular mode of administration. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of inhibitor which produces a therapeutic effect. Generally, out of one hundred percent, this amount will range from about 1 percent to about ninety-nine percent of active ingredient, preferably from about 5 percent to about 70 percent, most preferably from about 10 percent to about 30 percent.
Methods of preparing these formulations or compositions include the step of bringing into association a compound of the present invention with the carrier and, optionally, one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association a compound of the present invention with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product.
For systemic administration, injection is preferred, including intramuscular, intravenous, intraperitoneal, and subcutaneous (i.m., i.v., i.p., and s.c. respectively). The phrases "systemic administration", "administered systemically", "peripheral administration", and "administered peripherally" as used herein mean the administration of a compound, drug or other material other than directly into the central nervous system, such that it enters the patient's system and, thus, is subject to metabolism and other like processes, for example, subcutaneous administration.
For injection, the pharmaceutical compositions of the invention can be formulated in liquid solutions, preferably in physiologically compatible buffers such as Hank's solution or Ringer's solution. In addition, the pharmaceutical compositions may be formulated in solid form and redissolved or suspended immediately prior to use. Lyophilized forms are also included.
Pharmaceutical compositions of the invention may be formulated to be suitable for oral administration may be in the form of capsules, cachets, sachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and/or as mouth washes and the like, each containing a predetermined amount of a compound of the present invention as an active ingredient. A compound of the present invention may also be administered as a bolus, electuary or paste.
In formulating the pharmaceutical compositions of the invention in solid dosage forms for oral (p.o.) administration (capsules, tablets, pills, dragees, powders, granules and the like), a compound of the invention as active ingredient is mixed with one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethyl cellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as, for example, cetyl alcohol and glycerol monostearate; (8) absorbents, such as kaolin and bentonite clay; (9) lubricants, such a talc, calcium stcaratc, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof; and (10) coloring agents. In the case of capsules, tablets and pills, the pharmaceutical compositions may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, high molecular weight polyethylene glycols, and the like.
Gelatin capsules contain a compound of the present invention as active ingredient and powdered carriers, such as lactose, starch, cellulose derivatives, magnesium stearate, stearic acid, and the like. Similar carriers can be used to make compressed tablets. Both tablets and capsules can be manufactured as sustained release products to provide for continuous release of medication over a period of hours. Compressed tablets can be sugar-coated or film-coated to mask any unpleasant taste and protect the tablet from the atmosphere, or enteric coated for selective disintegration in the gastrointestinal tract. Solid compositions of a similar type are also employed as fillers in soft and hard-filled gelatin capsules; preferred materials in this connection also include lactose or milk sugar as well as high molecular weight polyethylene glycols. A preferred formulation is a solution or suspension in an oil, for example olive oil, Miglyol, or Capmul, in a soft gelatin capsule. Antioxidants may be added to prevent long-term degradation as appropriate.
A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared using a binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered inhibitor moistened with an inert liquid diluent.
The tablets and other solid dosage forms of the pharmaceutical compositions of the present invention, such as dragees, capsules, pills and granules, may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulations so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres. They may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved in sterile water, or some other sterile injectable medium immediately before use. These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. The active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients.
Liquid dosage forms for oral administration of the pharmaceutical compositions of the invention include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups, and elixirs. In addition to the active ingredient, the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifϊers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
Besides inert diluents, the pharmaceutical compositions for oral administration can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming, and preservative agents.
Suspensions, in addition to the pharmaceutical composition of the present invention, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar, and tragacanth, and mixtures thereof.
For buccal administration the pharmaceutical compositions may take the form of tablets or lozenges formulated in a conventional manner.
For administration by inhalation, the pharmaceutical compositions of the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of, for example, gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the therapeutic agents and a suitable powder base such as lactose or starch.
The pharmaceutical compositions may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The pharmaceutical compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
The phrases "parenteral administration" and "administered parenterally" as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion.
Pharmaceutical compositions of this invention suitable for parenteral administration comprise one or more inhibitors of the invention in combination with one or more pharmaceutically acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
Examples of suitable aqueous and nonaqueous carriers which may be employed in the pharmaceutical compositions of the invention include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
These pharmaceutical compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the pharmaceutical compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents that delay absorption such as aluminum monostearate and/or gelatin.
In addition to the formulations described previously, the pharmaceutical compositions may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the pharmaceutical compositions may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
Systemic administration can also be by transmucosal or transdermal means. For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration bile salts and fusidic acid derivatives. In addition, detergents may be used to facilitate permeation. Transmucosal administration may be through nasal sprays or using suppositories. For topical administration, the pharmaceutical compositions of the invention are formulated into ointments, salves, gels, or creams as generally known in the art. A wash solution can be used locally to treat an injury or inflammation to accelerate healing.
In some cases, in order to prolong the therapeutic effect of an inhibitor, it is desirable to slow the absorption of the inhibitor from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material having poor water solubility. The rate of absorption of the inhibitor then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered inhibitor form is accomplished by dissolving or suspending the inhibitor in an oil vehicle.
Pharmaceutical compositions of the invention may be formulated for rectal or vaginal administration as a suppository, which may be prepared by mixing one or more compounds of the invention with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active inhibitor.
Formulations of the pharmaceutical compositions of the present invention, which are suitable for vaginal administration, also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate.
Dosage forms for the topical or transdermal administration of a compound of this invention include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. Such compound may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants which may be required.
The ointments, pastes, creams and gels may contain, in addition to a compound of the invention, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
Powders and sprays can contain, in addition to a compound of this invention, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellants, such as chlorofiuorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
Transdermal patches have the added advantage of providing controlled delivery of a compound of the present invention to the body. Such dosage forms can be made by dissolving or dispersing an inhibitor of the present invention in the proper medium. Absorption enhancers can also be used to increase the flux of the drug across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the compound of the present invention in a polymer matrix or gel. Ophthalmic formulations, eye ointments, powders, solutions and the like, are also contemplated as being within the scope of this invention.
The pharmaceutical compositions may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the active ingredient. The pack may for example comprise metal or plastic foil, such as a blister pack. The pack or dispenser device may be accompanied by instructions for administration. In other embodiments, the pack or dispenser may be further packaged in an outer carton.
A pharmaceutical composition of the present invention can also be formulated as a sustained and/or timed release formulation. Such sustained and/or timed release formulations may be made by sustained release means or delivery devices that are well known to those of ordinary skill in the art, such as those described in U.S. Patent Nos.: 3,845,770; 3,916,899; 3,536,809; 3,598,123; 4,008,719; 4,710,384; 5,674,533; 5,059,595; 5,591,767; 5,120,548; 5,073,543; 5,639,476; 5,354,556; and 5,733,566, the disclosures of which are each incorporated herein by reference. The pharmaceutical compositions of the present invention can be used to provide slow or sustained release of one or more of the active ingredients using, for example, hydroxypropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microparticles, liposomes, microspheres, or the like, or a combination thereof to provide the desired release profile in varying proportions. Suitable sustained release formulations known to those of ordinary skill in the art, including those described herein, may be readily selected for use with the pharmaceutical compositions of the invention. Thus, single unit dosage forms suitable for oral administration, such as, but not limited to, tablets, capsules, gelcaps, caplets, powders, and the like, that are adapted for sustained release are encompassed by the present invention.
Injectable depot forms are made by forming microencapsuled matrices of the subject inhibitors in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions that are compatible with body tissue.
When the compounds of the present invention are administered as pharmaceuticals, to individuals, such as humans and animals, they can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99.5% (in certain embodiments, 0.5 to 90%) of active ingredient in combination with a pharmaceutically acceptable carrier.
The present invention provides new methods of treating proliferative, degenerative and other disorders or diseases, including cancer, by administering an amount such as a therapeutically effective amount of at least one of the compounds disclosed herein or a prodrug, tautomeric, pharmaceutically acceptable salt, N-oxide or stereoisomeric form thereof. The present invention further provides methods of treating proliferative, degenerative or other disorders or diseases, including cancer, by administering a therapeutically effective combination of at least one of these compounds and another anti-cancer or anti-proliferative agent.
A compound of the present invention may be administered as a salt or prodrug that, upon administration to the individual, is capable of providing directly or indirectly the parent compound, such as a compound as defined herein, or that exhibits activity itself. Nonlimiting examples include a pharmaceutically acceptable salt, alternatively referred to as a "physiologically acceptable salt". In addition, modifications made to a compound can affect its biological activity, in some cases increasing the activity over the parent compound. This activity can be assessed by preparing a salt or prodrug form of the compound, and testing its activity by using methods described herein or other methods known to those of skill in the art.
As will be apparent to a person skilled in the art, through the use of a prodrug of a given subject compound, an individual such as an animal administered or treated with such prodrug will be exposed to, and hence indirectly administered with, the subject compound. Such a procedure may expose those cells associated with a disease, such as a proliferative disease or disorder including cancer, to the subject compound. • ■ The compounds of the present invention may contain an asymmetrically substituted carbon atom, and may be isolated in optically active or racemic forms. It is well known in the art how to prepare optically active forms, such as by resolution of racemic forms or by synthesis from optically active starting materials. All chiral, diastereomeric, racemic forms and all geometric isomeric forms of a structure are intended, unless the specific stereochemistry or isomer form is specifically indicated. All processes used to prepare compounds of the present invention and intermediates made therein are considered to be part of the present invention.
Dosages
A dosage administered that will be a therapeutically effective amount of the compound sufficient, or reasonably expected by a health-care professional such as a physician, pharmacist or nurse, to result in amelioration of symptoms of, for example, the cancer or tumor will, of course, vary depending upon known factors such as the pharmacodynamic characteristics of the particular active ingredient and its mode and route of administration; age, sex, health and weight of the recipient; nature and extent of symptoms; kind of concurrent treatment, frequency of treatment and the effect desired.
The subject compounds may also be administered in prophylactic treatment. If the compound is administered prior to clinical manifestation of the unwanted condition (e.g., disease or other unwanted state of the host animal) then the treatment is prophylactic (i.e., it protects the individual against initiating, developing or further developing the unwanted condition). The subject compounds may also be administered to prevent a condition, disorder or diseases, such as cancer, or a syndrome complex, such as heart failure or any other medical condition. This includes administration of a compound the intent of which is to reduce the frequency of, or delay the onset of, symptoms of a medical condition in an individual relative to an individual which does not receive the compound. Thus, prevention of cancer includes, for example, reducing the number of detectable cancerous growths, tumors, or malignancies in a population of patients receiving a prophylactic treatment relative to an untreated control population, delaying the appearance of detectable cancerous growths in a treated population versus an untreated control population, and/or delaying disease progression and/or improving the quality of patient life, e.g., by a statistically and/or clinically significant amount.
Toxicity and therapeutic efficacy of pharmaceutical compositions of the present invention can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED5Q. Therapeutic agents that exhibit large therapeutic indices are useful for many circumstances. In certain circumstances, even therapeutic compositions that appear to exhibit debilitating or toxic side effects may be used, including circumstances where care is taken to design a delivery system that targets such therapeutic agents to the site of affected tissue in order to minimize potential damage to unaffected cells and, thereby, reduce or localize side effects.
The data obtained from cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For any agents used in the method of the invention, the therapeutically effective dose can be estimated initially from cell culture assays. A dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (i.e., the concentration of the test therapeutic agent which achieves a half-maximal inhibition of symptoms or inhibition of biochemical activity) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Levels in plasma may be measured, for example, by high performance liquid chromatography.
It is understood that appropriate doses of therapeutic agents depends upon a number of factors known to those or ordinary skill in the art, e.g., a physician. The dose(s) of the subject compounds will vary, for example, depending upon the identity, size, and condition of the subject or sample being treated, further depending upon the route by which the composition is to be administered, if applicable, and the effect which the practitioner desires the therapeutic to have upon the therapeutic target of targets, such as cells, nucleic acid or polypeptides, through with the disease causes, symptoms or effects are mediated.
Exemplary doses include milligram or microgram amounts of the compounds of the present invention per kilogram of subject or sample weight, e.g., about 1 microgram per kilogram to about 500 milligrams per kilogram, about 100 micrograms per kilogram to about 50 milligrams per kilogram, or about 1 milligram per kilogram to about 5 milligrams per kilogram.
A person skilled in the art will appreciate that doses can also be calculated on a body surface basis. A person of 70 kg has an approximate body surface area of 1.8 square meter, and doses can be expressed as milligram or microgram amounts of the compound per body surface area of subject or sample, e.g. about 50 microgram per square meter to about 1 5 grams per square meter, about 5 milligrams per square meter to about 1.5 grams per square meter, or about 50 milligram per square meter to about 150 milligrams per square meter.
Applications
The present invention further provides the compounds as described above for therapy. In other aspects, the invention provides the compounds of the present invention for prophylatic uses.
In certain embodiments, said therapy or prophylactic use is the treatment or prevention of a proliferative disorder or disease, such as a tumor or cancer. In certain embodiments, said treatment is the treatment of a cancer that can be treated by the inhibition of the activity of a protein kinase or mutant thereof, such as the inhibition of the activity of B-Raf or mutants thereof.
In certain other embodiments, said therapy or prophylactic use is the treatment or prevention of an inflammatory disorder or disease. In certain embodiments, said treatment is the treatment of an inflammatory disorder or disease that can be treated by the inhibition of the activity of a protein kinase or mutant thereof, such as the inhibition of the activity of p38 or an isoform thereof. Thus, the present invention additionally provides a method for treating an individual, such as a mammal, having a disease-state selected from the group of proliferative disorders or diseases, or inflammatory disorders or diseases, comprising administering to said individual a therapeutically effective amount of a compound, a prodrug, or a pharmaceutical composition of the invention as described above. In certain embodiments, said individual is a human. In certain embodiments, said proliferative disorder or disease is cancer. In certain embodiments, said treatment is the treatment of a cancer that can be treated by the inhibition of the activity of a protein kinase or mutant thereof, such as the inhibition of the activity of B-Raf or mutants thereof. In certain embodiments, said treatment is the treatment of the treatment of an inflammatory disorder or disease that can be treated by the inhibition of the activity of a protein kinase or mutant thereof, such as the inhibition of the activity of p38 or an isoform thereof.
The present invention also provides a method for prophylactic treatment of an individual such as an animal, including a mammal, particularly a human, the intent of which is to reduce the frequency of, delay the onset of, or the symptoms of a medical condition, such as cancer, in a subject relative to a subject which does not receive the composition.
In a further aspect, the invention provides methods of treating or preventing an individual suffering from a disease, such as a mammal, including a domestic mammal, cat, dog, horse, sheet, cow, rodent, and human, comprising the step of exposing said individual to an amount, including a therapeutically effective amount, of a subject compound. In certain embodiments, the disease is a proliferative disorder or disease, such as a cancer or tumour. In yet another embodiment, cells associated with said proliferative disorder or disease, including tumour cells included in a cancer, are exposed to the subject compound. In certain embodiments, said compound, or a prodrug thereof, is administered to said individual. In certain embodiments, said treatment is the treatment of a cancer that can be treated by the inhibition of the activity of a protein kinase or mutant thereof, such as the inhibition of the activity of B-Raf or mutants thereof. In certain embodiments, the disease is an inflammatory disorder or disease. In yet another embodiment, cells associated with said inflammatory disorder or disease are exposed to the subject compound. In certain embodiments, said compound, or a prodrug thereof, is administered to said individual. In certain embodiments, said treatment is the treatment of an inflammatory disease or disorder that can be treated by the inhibition of the activity of a protein kinase or mutant thereof, such as the inhibition of the activity of p38 or an isoform thereof.
In a further aspect, the invention provides a method of killing or inhibiting proliferation or growth of a cell, comprising contacting the cell with a compound of the invention. In one embodiment, the cell is cultured in-vitro, while in an alternative embodiment the cell is present in an individual. In a particular embodiment the cell is a cancer cell, for example a cell from a tumour cell line or a cell included in a tumour, including cancer cells from a tumour that can be treated by the inhibition of the activity of a protein kinase or mutant thereof, such as the inhibition of the activity of B-Raf or mutants thereof.
Yet another aspect of the invention relates to the use of a compound as described above, or a prodrug thereof, .for the preparation of a medicament for the treatment or prevention of a proliferative disorder or disease, including cancer, including cancers that can be treated by the inhibition of the activity of a protein kinase or mutant thereof, such as the inhibition of the activity of B-Raf or mutants thereof. Additionally, the invention relates to a pharmaceutical composition comprising a compound as described above, or a prodrug thereof, and a pharmaceutically acceptable diluent, excipient or carrier, for the treatment of a proliferative disorder or disease, including cancer, including cancers that can be treated by the inhibition of the activity of a protein kinase or mutant thereof, such as the inhibition of the activity of B-Raf or mutants thereof.
Yet another aspect of the invention relates to the use of a compound as described above, or a prodrug thereof, for the preparation of a medicament for the treatment or prevention of an inflammatory disorder or disease, including inflammatory disorders or diseases that can be treated by the inhibition of the activity of a protein kinase or mutant thereof, such as the inhibition of the activity of p38 or an isoform thereof. Additionally, the invention relates to a pharmaceutical composition comprising a compound as described above, or a prodrug thereof, and a pharmaceutically acceptable diluent, excipient or carrier, for the treatment of an inflammatory disorder or disease, including cancers that can be treated by the inhibition of the activity of a protein kinase or mutant thereof, such as the inhibition of the activity of p38 or an isoform thereof.
The subject compounds are useful to treat various disorders or diseases, including proliferative disorders or diseases, and inflammatory disorders or diseases. The term "proliferative disorder or disease" is also art recognized and includes a disorder or disease affecting an individual, such as an animal, in a manner which is marked by aberrant, or otherwise unwanted, proliferation of a subset of cells of an individual. Cancer and tumors are proliferative disorders or diseases. Cells comprising or derived from a tumor will generally be understood to be a proliferating cell, typically a hyper-proliferating cell, and in other circumstances, a tumor cell may be dysplastic, or may have proliferated, in certain embodiments, said treatment is the treatment of a cancer that can be treated by the inhibition of the activity of a protein kinase or mutant thereof, such as the inhibition of the activity of B-Raf or mutants thereof.
It will be apparent to a person skilled in the art, on reading the disclosure of the instant invention, that the methods, pharmaceutical compositions and packaged pharmaceuticals comprising the subject compounds will be useful for the treatment of other proliferative disorders or diseases, or for killing or inhibiting proliferating cells including tumor cells.
Compounds of the present invention may be useful in the treatment of disease processes which feature abnormal cellular proliferation, such as hyperproliferative diseases, including cancer, benign prostate hyperplasia, familial adenomatosis polyposis, neurofibromatosis, psoriasis, fungal infections, endotoxic shock, hypertrophic scar formation, inflammatory bowel disease, transplant rejection, vascular smooth muscle cell proliferation associated with atherosclerosis, psoriasis, pulmonary fibrosis, arthritis, glomerulonephritis, restenosis following angioplasty or vascular surgery, and other post-surgical stenosis and restenosis. See, for example, U.S. Patent Nos. 6,114,365 and 6,107,305. The compounds disclosed herein are expected to be useful in the therapy of proliferative or hyperproliferative disorders or diseases such as cancer, autoimmune diseases, viral diseases, fungal diseases, neurodegenerative disorders and cardiovascular disease.
In certain embodiments, tumors may be solid tumors, which are cancer of body tissues other than blood, bone marrow, or the lymphatic system. In other embodiments, tumors may be hematological tumors, such as leukemia and lymphomas. Leukemia is a collective term for malignant diseases characterized by a proliferation of malignantly changed white blood cells. Diseases arising from lymphatic tissue are called lymphomas.
Solid tumors may be selected from: liver cancer, stomach cancer, colon cancer, breast cancer, pancreas cancer, prostate cancer, skin cancer, renal cancer, bone cancer, thyroid cancer, skin cancer, including squamous cell carcinoma, esophagus cancer, kidney cancer, bladder cancer, gall cancer, cervical cancer, ovarian cancer, lung cancer, bronchial, small and non-small-cell lung cancer, gastric, and head and neck cancer.
Hematological tumors may be leukemia, such as Acute Myelogenous Leukemia (AML), Acute Lymphoblastic Leukemia (ALL), Acute Lymphocytic Leukemia, Acute Leukemia, Acute Promyelocytic Leukemia, Chronic Granulocytic Leukemia (CGL), Chronic Leukemia, Chronic Lymphocytic Leukemia (CLL), Chronic Myelogenous Leukemia (CML), Chronic Myelomonocytic Leukemia, Common-type Acute Lymphoblastic Leukemia, Eosinophilic Leukemia, Erythroleukemia, Extranodal Lymphoma, Follicular Lymphoma, Hairy Cell Leukemia, Monocytic Leukemia, Prolymphocytic Leukemia.
Hematological tumors may also be lymphoma, such as B Cell Lymphomas, Burkitt Lymphoma, Cutaneous T Cell Lymphoma, High-Grade Lymphoma, Hodgkin's Lymphoma, Non-Hodgkin's Lymphoma, . Low-grade Lymphoma, Lymphoblastic Lymphoma, Mantle Cell Lymphoma, Marginal Zone Lymphoma, Mucosa-Associated Lymphoid Tissue (MALT) Lymphomas, T Cell Lymphomas, peripheral T cell lymphoma, multiple myeloma, Essential Thrombocythemia, Hairy Cell Lymphoma, Extramedullar myeloma, Granulocytic Sarcomae. Hematological tumors may also be tumors of myeloid lineage, including acute and chronic myelogenous leukemias, myelodysplastic syndrome, and promyelocytic leukaemia.
Tumors may also be of mesenchymal origin, such as fibrosarcoma and rhabdomyosarcoma. Furthermore, tumors may be tumors of the central and peripheral nervous system, such as astrocytoma, neuroblastoma, glioma, and schwannomas; and tumors may be other tumors, such as melanoma, seminoma, teratocarcinoma, osteosarcoma, xenoderoma pigmentosum, keratoctanthoma, thyroid follicular cancer, and Kaposi's sarcoma.
Tumors that are resistant or refractory to treatment with other anti-cancer or anti-proliferative agents may also benefit from treatment with the methods and pharmaceutical compositions of the present invention.
Compounds disclosed herein may also be useful in the chemoprevention of cancer. Chemoprevention is defined as inhibiting the development of invasive cancer by either blocking the initiating mutagenic event or by blocking the progression of pre-malignant cells, such as by blocking growth of the tumor, that have already suffered an insult or inhibiting tumor relapse.
Compounds disclosed herein may also be useful in inhibiting tumor angiogenesis and metastasis.
The compounds of this invention may also be useful in combination (administered together or sequentially) with known anti-cancer treatments such as radiation therapy or with anti-cancer, anti-proliferative, cytostatic or cytotoxic agents. Other anti-cancer and anti-proliferative agents which may be used in combination with the compounds of the present invention include those decribed herein. In combination treatment, the compounds of the present invention may be further administered with any other anti-cancer and anti-proliferative agent disclosed herein.
If formulated as a fixed dose, such combination products employ the compounds of this invention within the dosage range described herein and the other pharmaceutically active agent or treatment within its approved dosage range. For example, the cdc2 inhibitor olomucine has been found to act synergistically with known cytotoxic agents in inducing apoptosis (J. Cell Sci., 108, 2897 (1995)). Compounds described herein may also be administered sequentially with known anti-cancer or anti-proliferative agents when a combination formulation is inappropriate. The invention is not limited in the sequence of administration; compounds described herein may be administered either prior to or after administration of the known anti-cancer or anti-proliferative agent. For example, the cytotoxic activity of the cyclin-dependent kinase inhibitor flavopiridol is affected by the sequence of administration with anticancer agents (Cancer Research, 57, 3375 (1997)).
Further Aspects of the Invention
Another aspect the invention provides a pharmaceutical package, wherein said package includes a compound of any of the formulae of the present invention. In certain embodiments, the package comprises instructions which indicate that said composition may be used for the treatment of an individual in need thereof, including a human. In certain other embodiments, the pharmaceutical package includes one or more compounds of the present invention formulated together with another pharmaceutical ingredient such as an anti-cancer or anti-proliferative agent. In this case, the compound(s) of the present invention and the other pharmaceutical ingredient may be formulated separately and in individual dosage amounts.
Other pharmaceutical ingredients that may be formulated together or separately with the compounds of the present invention include but are not limited to other anti-cancer and anti-proliferative agents such as described above. In certain still further embodiments, the pharmaceutical package comprises instructions to treat a patient in need of such treatment. In yet another aspect the invention provides a pharmaceutical package for treating an individual suffering from a proliferative disorder or disease, such as a tumor or a cancer, wherein said package includes at least one compound of the present invention. In certain still further embodiments, the pharmaceutical package comprises instructions to treat the disorder.
As used herein the term "pharmaceutical package" or "pharmaceutical pack" refer to any packaging system for storing and dispensing individual doses of medication. Preferably the pharmaceutical package contains sufficient daily dosage units appropriate to the treatment period or in amounts which facilitate the patient's compliance with the regimen. In certain embodiments, the pharmaceutical pack comprises one or more vessels that include the active ingredient, e.g., a compound of the present invention. Such vessel can be a container such as a bottle, vial, syringe, or capsule, or may be a unit dosage form such as a pill. The active ingredient may be provided in the vessel in a pharmaceutically acceptable form or may be provided, for example, as a lyophilized powder. In further embodiments, the pharmaceutical pack may further include a solvent to prepare the active ingredient for administration. In certain embodiments, the active ingredient may be already provided in a delivery device, such as a syringe, or a suitable delivery device may be included in the pack. The pharmaceutical package may comprise pills, liquids, gels, tablets, dragees or the pharmaceutical preparation in any other suitable form. The package may contain any number of daily pharmaceutical dosage units. The package may be of any shape, and the unit dosage forms may be arranged in any pattern, such as circular, triangular, trapezoid, hexagonal or other patterns. One or more of the doses or subunits may be indicated, for example to aid the doctor, pharmacist or patient, by identifying such dose or subunits, such as by employing color-coding, labels, printing, embossing, scorings or patterns. The pharmaceutical package may also comprise instructions for the patient, the doctor, the pharmacist or any other related person.
Some embodiments comprise the administration of more than one active ingredient, including compounds as disclosed herein. Such administration may occur concurrently or sequentially. The active ingredients may be formulated together such that one administration delivers both components. Alternatively the active ingredients may be formulated separately. The pharmaceutical package may comprise the compound of the present invention and the other pharmaceutical ingredient in a single formulation, i.e., they are formulated together, or the compound of the present invention and the other pharmaceutical ingredient in individual formulations, i.e., they are formulated separately. Each formulation may comprise the compound of the present invention and the other pharmaceutical ingredient in individual dosage amounts (in approximately equal or unequal amounts). Administration of the compound of the present invention and the other pharmaceutical ingredient results in a concentration that results in a therapeutically effective amount of the combination.
As used herein, the term "instructions" means a product label and/or documents or other information describing relevant materials or methodologies pertaining to assembly, preparation or use of a kit or packaged pharmaceutical. These materials may include any combination of the following: background information, steps or procedures to follow, list of components, proposed dosages, warnings regarding possible side effects, instructions for administering the drug, technical support, and any other related documents. Instructions can be supplied in printed form, such as a package label or a package insert. Instructions for a packaged pharmaceutical or a pharmaceutical composition can be inserted in a delivery carton or finished package, e.g., as a package insert, and the text of such has been approved by a competent regulatory authority such as the Food and Drug Administration (FDA) of the United States. Alternatively or complementarily, instruction may also be stored in electronic form, e.g., on a computer-readable storage medium such as a computer-readable memory device, a centralized database, magnetic media such as hard disks, floppy disks, and magnetic tape; optical media such as compact discs, CD-ROMs and holographic devices; magneto-optical media such as floptical disks; and hardware devices that are specially configured to store and execute program code, such as application-specific integrated circuits (ASICs), programmable logic devices (PLDs) and ROM (read only memory) and RAM (random access memory) devices. Instructions may comprise a web address of an internet website from which more detailed instructions may be downloaded, or a recorded presentation. Instructions can contain one or multiple documents or future updates.
The invention further relates to a method of synthesizing a compound according to the present invention, comprising the step of reacting a compound having a structure represented by formula (II) with a compound having a structure represented by formula (III)
Figure imgf000077_0001
(II) (III)
wherein R17 is independently selected from — C1-6- alkyl, -CH2-aryl, or— aryl, with R3 being as defined above.
The invention further relates to a method of synthesizing a compound according to the present invention, comprising the step of reacting a compound having a structure represented by formula (II) as described above with a compound having a structure represented by formula (IV)
Figure imgf000077_0002
(IV)
with R4 and R5 being as defined above.
In certain embodiments, -O-R5, when taken together, is not a Ci-s-alkoxy or an O-linked polyether containing between 2 and 8 carbon atoms in total. In certain such embodiments, R5 is not alkyl or alkoxy-substiruted alkyl.
Furthermore, the invention relates to a compound having a structure represented by formula (V)
Figure imgf000077_0003
(V) or any tautomeric or stereoisomeric form thereof, wherein R7 is selected from -S(O)m-R17, with m = 0, 1 or 2, and -N(RVV-R2;
R18 is taken from the list of -W-R4, -COOH, -COOR17, and -Br; R17 is independently selected from — Ci-6- alkyl, -CH2-aryl, or -aryl; with R1, R2, R3, R4, R5, W, and X being as defined above, provided that if R7 is -N(R^-V-R2, then R18 is not -W-R4.
In further certain embodiments, -O-Rs, when taken together, is not a Ci-8- alkoxy or an O-linked polyether containing between 2 and 8 carbon atoms in total. In certain such embodiments, R5 is not alkyl or alkoxy-substituted alkyl.
Examples
A selection of compounds within the scope of the present invention are listed in Table 1. The compounds in Table 1 were synthesized according to examples 1 to 10 below, and the surprising inhibitory activities in biochemical assays, and antiproliferative activities in cellular assays of these compounds are shown in Tables 2 and 3, respectively, as determined according to examples 11 to 13.
A. Synthesis
Compounds of the invention may be prepared by the synthetic sequence shown in Schemes 1 or 2. For example, examples 1 -7 show in detail the synthesis steps of Scheme 1. As depicted in Scheme 1, 8-oxy pyrido[2,3-d]pyriτnidones may be formed in one step from malonate derivatives and 6-chloro-5-formyl-2- methylthiopyrimidine. The compounds of examples 8-10 show in detail the synthesis steps of Scheme 2. As depicted in Scheme 2, 8-oxy pyrido[253- d]pyrimidones may be formed in one step from phenylacetic acid hydroxylamide derivatives and 6-chloro-5-formyl-2-methylthiopyrimidine. A skilled artisan will appreciate that other routes of synthesis may be employed as well. In particular, other routes of synthesis may in fact be applied to certain aspects of the present invention. The skilled artisan is referred to general textbooks, such as March's Advanced Organic Chemistry (Michael B. Smith & Jerry March, Wiley- Interscience, 2000), The Practice of Medicinal Chemistry (Camile G. Wermuth, Academia Press, 2003) and Protective Groups in Organic Synthesis (Theosora W. Greene & Peter G.M. Wuts; John Wiley & Sons Inc, 1999).
A: Synthesis of 8-hydroxy-pyrido[2,3-d]pyrimidin-7-one derivatives (examples 1-8)
Scheme 1:
Figure imgf000079_0001
AcOH
Figure imgf000079_0002
69%
Figure imgf000079_0003
TΓA
CH2Cl2
Figure imgf000079_0004
Example 1
Figure imgf000080_0001
THF, MeOH b. 2M aq. HCl, 34%
Ester 1 (5.83 g, 28.6 mmol) and aldehyde 2 (4.50 g, 23.9 mmol) were mixed in 300 mL of anhydrous DMF and cooled to 0 0C. Potassium carbonate (9.90 g, 71.7 mmol) was added and the reaction was stirred at 0 0C for 1 hour before being allowed to warm to room temperature for 1.5 hours. Half of the DMF was removed under vacuum and the remainder was diluted with 500 mL of ethyl acetate and poured into a separatory funnel. The organic layer was washed twice with 500 mL of water. The water layer was then extracted with 250 mL of ethyl acetate. The organic layers were combined and washed with 500 mL of brine. The organic layer was separated, dried with anhydrous sodium sulfate, filtered and concentrated under vacuum to give 9.29 g of crude product. The material was filtered through silica gel, eluting with 5% methanol/methylene chloride to give 6.77 g of slightly impure product.
The material was then dissolved in 200 mL of 1 :1 THF:MeOH followed by the addition of 30 mL of 1 M aqueous NaOH (30.0 mmoles). The reaction was stirred for 10 minutes, quenched with 30 mL of 1 M aqueous HCl, and then concentrated under vacuum. The crude slurry was triturated with 50 mL of anhydrous ethanol and filtered. The solid was washed with cold anhydrous EtOH and Et2O to give 2.53 g (35% overall yield) of clean product 3.
Example 2
Figure imgf000080_0002
Acid 3 (2.53 g, 8.17 mmol) was placed in 15OmL of anhydrous pyridine at room temperature followed by bromine (0.42 mL, 8.17 mmol). The mixture was stirred for 15 minutes at room temperature and then heated to 80 0C for 30 minutes. The solution was briefly heated to reflux and then allowed to cool to room temperature. The pyridine was removed under vacuum. The solid was triturated with a 1 :1 mixture of ethyl acetate / diethyl ether and filtered. The mother liquor was concentrated to give 2.05 g of product 4. The solid was dissolved with ethyl acetate and washed with 1 M aqueous HCl to yield a further 0.81 g of product after extraction.
Example 3
Figure imgf000081_0001
Bromide 4 (2.04 g, 5.92 mmol) was slurried in 50 mL of glacial acetic acid followed by the addition OfNa2WO4 (195 mg, 0.59 mmol) and 30% H2O2 (2.30 mL, 20.72 mmol). The slurry was stirred overnight at room temperature. The solution was then concentrated to approximately 25mL of acetic acid and diluted with 75 mL of water. The solid was filtered and washed with water. The filter cake was allowed to air dry to give 1.53 g (69% yield) of product 5.
Example 4
Figure imgf000081_0002
Bromide 5 (1.53 g, 4.06 mmol) was place in 20 mL of aniline and heated to 80 0C for 2 hours. The reaction was cooled and diluted with 20 mL of glacial acetic acid and 100 mL of 2 M aqueous HCl. The solid was filtered and washed with water. The filter cake was allowed to air dry to give 1.36 g of product 6 (86% yield).
Example 5
Figure imgf000082_0001
Bromide 6 (1.29 g, 3.31 mmol), 2-chlorophenylboronic acid (570 mg, 3.65 mmol), K3PO4 (1.55 g, 7.28 mmol) and [1,1'-
Bis(diphenylphosphino)ferrocene]dichloropalladiurn(II) complex with dichloromethane (270 mg, 0.33 mmole) were mixed as solids and placed under argon. Argon was then bubbled through a 1 : 1 mixture of anhydrous DMF (25 mL) and DME (25 mL) for 15 minutes. The solvent was then added to the solid mix and the solution was heated to 90 0C overnight. The solution was cooled, diluted with 250 mL of ethyl acetate and washed with 250 mL of saturated aqueous "NaHCO3 and 250 mL of brine. The organic layer was separated, dried with anhydrous sodium sulfate, filtered and concentrated under vacuum to give 2.08 g of crude product. The material was purified via silica gel column chromatography eluting with 5% methanol/methylene chloride to give 1.54 g (100%) of product.
Example 6
Figure imgf000082_0002
Compound 7 (1.54 g, 3.65 mmol) was dissolved in 40 mL of a 1:1 mixture of methylene chloride and trifluoroacetic acid. The solution was stirred at room temperature for 24 hours and then concentrated under vacuum. 50 mL of anhydrous toluene was then added and the solution was concentrated again under vacuum to give 1.94 g of product which was used directly with no further purification.
Example 7
Figure imgf000083_0001
Compound 8 (205 mg, 0.56 mmol) was placed in DMF with 2- bromopropane (0.07 mL, 0.70 mmol) followed by the addition of K2CO3 (232 mg, 1.68 mmol). The reaction was heated to 80 0C for 2 hours. The solution was cooled, filtered to remove the solid carbonate and purified via reverse phase preparative chromatography (63.8 mg isolated after purification.)
Scheme 2:
Figure imgf000084_0001
Example 8
Figure imgf000084_0002
To an ice cold DMF solution (200 mL) of compound 1 (2.97 g, 12.7 mmol) and compound II (2.0 g, 10.6 mmol) was added potassium carbonate (2.2 g, 15.9 mmol). After stirring for 2 hours reaction was poured into water (500 mL), extracted with ethyl acetate (2 x 100 mL), dried over sodium carbonate, and solvent was removed under reduced pressure. White solid was triturated with ether and collected by vacuum filtration yielding III (2.0 g, 55%).
Example 9
Figure imgf000085_0001
A dichloromethane solution (9 mL) of compound III (300 mg, 0.81 mmol) and (+)-(8,8-Dichlorocamphorsulfonyl)-oxaziridine (730 mg, 2.44 mmol) was irradiated with UV at 80 0C for 90 minutes. Solvent was removed and solid was flash chrornatographed on silica (2 % methanol/dichloromethane) recovering IV (300 mg, 95%).
Example 10
Figure imgf000085_0002
A dioxane solution (0.9 mL) of compound IV (175 mg, 0.46 mmol) and compound V (829 mg, 4.6 mmol) was irradiated with UV at 150 0C for 10 minutes. Solvent was removed under reduced pressure and crude VI was dissolved in DMSO (4 mL). DMSO solution was purified on preparative HPLC recovering VI (140 mg) as TFA salt.
B: Biological Activity Assays (examples 11-13)
The biological activity and utility of the compounds of the invention are demonstrated by one or more assays including those described in more detail below. Example 11: Determination of IC50 values for inhibition of kinase activity
We characterized the in vitro inhibitory activity against protein kinases of compounds of the present invention by determining their IC50 values (see Table 2).
a) C-Raf (MAPH assay).
Kinase: C-Raf Raf-1 (truncated), active (Upstate; cat. no. 14-
352)
Reaction Volume: 40 μl Reaction Time: 60 min Reaction Temperature: room temperature Assay Plate: 96 well U bottom plate (Greiner, 650161)
MultiScreen-PH Plate: 96 well MAPH Filter Plates (Millipore, MAPHNOB50)
Filter Washing Solution: 0.75% H3PO4 Scintillation Liquid: Supermix Liquid Scintillator (PerkinElmer,
1200-439)
Controls:
Negative Control (C-): 100 mM EDTA, no inhibitor
Positive Control (C+): no inhibitor
Reaction Buffer ffinal concentration):
20 mM Tris, pH 7.5
2 mM MnCl2
1 mM DTT
0.01% Tween20
Final Assay Concentrations: Kinase: Use kinase cone, yielding 10% ATP turnover as determined in titration experiment.
ATP: 5.78 μM
Adenosine 5'-[γ- P]triphosphate: 12.5 μCi/ml (Amersham Biosciences,
BFlOOO)
Substrate: Myelin Basic Protein 57.8 μM (Invitrogen, 13228-
010)
Pipetting Sequence:
1) Add 10 μl 4-fold concentrated substrate + 4-fold concentrated ATP in 3- fold concentrated reaction buffer to each well of assay plate
2) Add 10 μl 4-fold concentrated inhibitor in 4% DMSO in H2O to each well except to C- and C+ wells (starting point: final inhibitor concentration 10 μM; IC50 determination based on dilution series)
3) Add 10 μl 4% DMSO in H2O to C- and C+ wells
4) Add 10 μl 500 mM EDTA in H2O to C- wells
5) Add 10 μl 50 μCi/ml adenosine 5'-[γ-33P]triphosphate in H2O to each well
6) Add 10 μl 4- fold concentrated kinase in Reaction Buffer to each well
7) Incubate 1 hr at room temperature
8) Add 10 μl 50 mM EDTA in H2O to each well except to C- wells
9) Prepare MAPH plates by adding 200 μl 0.75% H3PO4 to each well
10) Exhaust 0.75% H3PO4 using Millipore vacuum station
11) Add 60 μl 0.75% H3PO4 to each well of MAPH filter plate
12) Transfer 30 μl sample per well from assay plate to corresponding well of MAPH filter plate
13) Incubate 30 min at room temperature 14) Wash each well of MAPH filter plates 3x with 200 μl 0.75% H3PO4 using Millipore vacuum station
15) Add 20 μl scintillation liquid to each well of MAPH filter plate
16) Seal MAPH filter plate
17) Store MAPH filter plate 30 min in darkness
18) Quantify radioactivity using scintillation counter (MicroBeta, Perkin- Elmer)
b) C-Raf (IMAP assay)
Kinase: C-Raf Raf-1 (truncated), active (Upstate; cat. no. 14-
352)
IMAP Assay:
Reaction Volume: 8.0108 μl Reaction Time: 60 min Reaction Temperature: room temperature IMAP Incubation Time: 60 min
Assay Plate: 384 well U bottom, PP, black, low volume (Corning, 3676)
Compound Plate: 384 well U bottom, PS (Falcon, 3995)
IMAP Binding Buffer A: Molecular Devices, R7282
IMAP Binding Buffer B: Molecular Devices, R7283
IMAP Binding Reagent: Molecular Devices, R7207
Controls:
Negative Control (C-): no kinase, no inhibitor
Positive Control (C+): no inhibitor Reaction buffer: 20 mM Hepes, pH 7.5
1 mM DTT
1O mM MnCl2
Figure imgf000089_0001
Final Assay Concentrations:
Kinase: Kinase cone, yielding 50% substrate turnover as determined in titration experiment c-Raf (Upstate 14-352) ATP: 4.87 μM
Substrate: 5F1-SGQLIDSMANSFV-NH2 400 nM (jpt Peptide
Technologies GmbH, Berlin, Germany)
IMAP Binding Solution: 75 % IMAP Binding Buffer A
25 % IMAP Binding Buffer B IMAP Binding Reagent 1 :800
Pipetting Sequence:
1) Add 6 μl 1.33-fold concentrated substrate + 1.33-fold concentrated ATP in 1-fold concentrated reaction buffer to each well of assay plate
2) Add 10.8 nl 740-fold concentrated inhibitor in 100% DMSO to each well except to C- and C+ wells using pintool (CyBio, Jena, Germany) (starting point: final inhibitor concentration 10 μM; IC50 determination based on dilution series)
3) Add 10.8 nl 100% DMSO to C- and C+ wells using pintool
4) Add 2 μl reaction buffer to C- wells
5) Add 2 μl 4-fold concentrated kinase in reaction buffer to each well except C- wells
6) Incubate according to reaction time at room temperature
7) Add 15 μl IMAP binding solution to each well
8) Incubate according to IMAP incubation time at room temperature 9) Measure fluorescence polarization (Analyst GT, Molecular Devices)
c) B-Raf
Kinase: B-Raf deltal-415 fe.g.. Upstate Cat.No. 14-53O
Reaction Volume: 40 μl Reaction Time: 60 min Reaction Temperature: room temperature Assay Plate: 96 well U bottom plate (Greiner, 650161)
MultiScreen-PH Plate: 96 well MAPH Filter Plates (Millipore, MAPHNOB50)
Filter Washing Solution: 0.75% H3PO4
Szintilation Liquid: Supermix Liquid Szintillator (PerkinElmer, 1200-439)
Controls:
Negative Control (C-): 100 mM EDTA, no Inhibitor Positive Control (C+): no Inhibitor Reaction Buffer: 20 mM Mops, pH 7.0
TO mM MgCl2
0.4 mM MnCl2
10O mM NaCl
1 mM DTT
0.01 % NP40
Final Assay Concentrations: Kinase: Kinase cone, yielding 50% substrate turnover as determined in titration experiment. ATP: 27.15 μM
Adenosine 5'-[γ-33P]triphosphate: 12.5 μCi/ml (Amersham Biosciences, BFlOOO)
Substrate: MEKl inactive 2 μM (Upstate 14-420)
Pipetting Sequence:
1) Add 10 μl 4-fold concentrated substrate in 3 -fold concentrated reaction buffer to each well of assay plate
2) Add 10 μl 4-fold concentrated inhibitor in 4% DMSO in H2O to each well except to C- and C+ wells (starting point: final inhibitor concentration 10 μM; ICso determination based on dilution series)
3) Add 10 μl 4% DMSO in H2O to C- and C+ wells
4) Add 10 μl 500 mM EDTA in H2O to C- wells
5) Add 10 μl 50 μCi/ml adenosine 5'-[γ-33P]triphosphate in H2O + 4-fold cold ATP to each well
6) Add 10 μl 4-fold concentrated kinase in reaction buffer to each well
7) Incubate 1 hr at room temperature
8) Add 10 μl 50 mM EDTA in H2O to each well except to C- wells
9) Prepare MAPH plates by adding 200 μl 0.75% H3PO4 to each well
10) Exhaust 0.75% H3PO4 using Millipore vacuum station
11 ) Add 60 μl 0.75% H3PO4 to each well of MAPH filter plate
12) Transfer 30 μl sample per well from assay plate to corresponding well of MAPH filter plate
13) Incubate 30 min at room temperature
14) Wash each well of MAPH filter plates 3x with 200 μl 0.75% H3PO4 using Millipore vacuum station
15) Add 20 μl scintillation liquid to each well of MAPH filter plate
16) Seal MAPH filter plate
17) Store MAPH filter plate 30 min in darkness
18) Quantify radioactivity using scintillation counter (MicroBeta, Perkin- Elmer) d) Other kinases
Compounds of the present invention were shown to be inhibitors of other kinases, including p38 and KDR, as well by using standard kinase inhibition assays.
Compounds of the present invention are tested for inhibitory activity against other kianses, including tyrosine kinases and serine-threonine kinases, using assays known in the art, such as those described in WO 06/002119. Selectivity of specific compounds or compound-classes between various kinase enzymes or kinase families are investigated by comparison of the IC50 values obtained from such assays.-
Example 12: In vitro anti-proliferative activity of compounds of the present invention against proliferation of a cancer cell line
We observed the surprising finding that compounds of the invention were useful in inhibiting proliferation of HT-29 tumor cells (see Table 3).
3,000-15,000 cells/well were exposed to the test compounds at various concentrations appropriate to determine an IC50, for 72 hours, and cell proliferation was measured using the SRB assay according to Shekan et al (J Natl Cancer Inst (1990) 82, 1107-112) in order to estimate the IC50 values shown in Table 3. Briefly, cells were plated in 96 well dishes 24 hours prior to compound addition. The assay was terminated with the addition of cold TCA to a final concentration of 10% and the plates were incubated for one hour at 4°C. The plates were then washed 5 times with water and lOOμl of a Sulforhodamine B solution (4%) was added to each well. The plate was then incubated for 10 minutes at room temperature before removal of unbound dye by washing with 1% acetic acid. The bound dye was solubilized with 10 mJVl Trizma base and the absorbance read at OD570. Inhibitory activity of the compounds was calculated as % inhibition of cell proliferation compared to cells treated with the solvent (DMSO). Table 3 represents the IC50 values for inhibition of cell proliferation for certain compounds of the invention showing that the compounds demonstrated a clear and pronounced anti-proliferative activity towards HT-29 cells. Example 13: Activity of compounds in xenograft tumor models
With this assay we demonstrated the activity of compounds of the present invention against tumor cells in an in-vivo xenograft model.
METHODS: Compound preparation
Compounds 4 and 24 (see Table 1) were prepared for i.p. administration in a biocompatible vehicle (compound 4: in 10% DMA/50% PEG300/water; compound 24: in 5% Cremophor EL/5% ethanol/90% saline). All preparations were made freshly and injection volumes were adjusted to body weight (resulting in compound dosages of between 40 and 80mg/kg mouse).
Mice/Husbandry.
Mice were obtained from Charles River Laboratory (CRL), housed in static microisolators, and provided ad libitum with water and an irradiated standard rodent diet (Purina Pico-Lab Rodent Diet 20).
Standard Protocol. Experiments in Athymic mice.
Athymic nu/nu female mice (6-8 weeks old) from CRL were allowed to acclimate for at least 4 days. Human HT-29 cells (ATCC) colon carcinoma cell line were cultured in McCoy's medium (ATCC) supplied with 10% FCS and 1% Pen/Strep. The 2nd passage of cells with approximately 80% confluence was used in the study. Briefly, on Day 0, mice were inoculated with 0.1 ml (total 5xlO6 cells) of HT-29 cell suspension (50xl0δ cells/ml in McCoy's medium without supplements mixed 1 :1 with Matrigel) by a subcutaneous injection into the lower right flank under light anesthesia. When the average tumor weights reached above lOOmg (day 7), 70 animals with an appropriate tumor size with (average of about 182 mg) were selected and were randomly divided into 7 groups (10 animals each). Tumor growth and body weight are monitored and recorded twice a week. Tumors are measured by determining the length and width of the tumor with a digital caliper. Tumor weight is estimated using the following formula: Tumor Weight (mg) = (w2 x 1) / 2 where w = width and 1 = length in mm of the tumor.
Tumor Growth Inhibition (TGI) % is calculated as follows: %TGI = 100(1- T/C) where T is the mean tumor size of a compound treated group on a given day, and C is the mean tumor size of the vehicle control group on the same day.
Toxic deaths are defined as deaths caused by compound treatment and not by advanced disease state. A death is considered toxic if the animal dies within 1 week after the final compound treatment and the tumor size has not reached 1000 mg non- tumor related deaths after this point are recorded, but not considered toxic deaths.
Group 1 was treated with vehicle only (10%DM A/50% PEG300/40% ddwater, pό), groups 2 to 4 were treated with compound 4 (40, 60 and 80 mg/kg doses dosed for five days), groups 5 to 7 were treated with compound 24 (40, 60 and 80 mg/kg doses dosed for ten days).
Mice were sacrificed when their tumors reached the 1000 mm3 endpoint volume. Treatment efficacy was determined as Log Cell Kill (LCK). LCK is a calculation that determines the percentage of tumor cells that are presumably killed after the initiation of treatment and can be used as a quantitative measure of efficacy: LCK = (T-C) / (3.32)(Td) where T = is the mean time required for the treatment group of mice to reach 1000 mg in size, C = the mean time for the control group tumors to reach 1000 mg in size, Td = is the Tumor Doubling time estimated from the linear regression analysis from a semi-log growth plot of the control group tumors during exponential growth and 3.32 = the number of doublings required for a population to increase 1-loglO unit. Each LCK unit represents 1-loglO unit of cell killing (e.g., 1 LCK = 90% kill, 2 LCK = 99% kill, etc.).
Results.
No toxic deaths were observed for compounds 4 and 24. Both compounds showed LCK values above 0.4 in all dosages tested. Further xenograft experiments were conducted as above, to investigate compounds of the invention, including compounds 91, 98, 104, 111, 121, 136 and 143. Compounds were dosed at 30, 60 & 90 mg/Kg except for compound 91 which was dosed at amounts 20, 40 and 80 mg/Kg. As a positive control, the Raf inhibitor sorafinib (NEXAVAR) was dosed at 60 mg/Kg. In these experiments, compounds were administered as a qdxlO schedule.
All compounds showed LCKs of between 0.2 to 0.9 at the lowest doses, and between 0.3 to 1.3 at the highest doses. When compared to the positive control, compounds showed LCKs of between -1A of to around the same LCK value as the positive control. Activity of these compounds in these model experiments was further demonstrated by considering the TGI (compared to tumour growth in the vehicle control), with compounds typically showing a TGI of over 50% at the higher concentrations, and in some cases with a TGI of between 80% and 95%. Under these-non-optimised formulation and dosage conditions, a number of compounds showed toxic deaths of individual mice, typically at the higher dosage of compound.
C: Selection and development of drug candidates
Example 14.
In order to select the most appropriate compound to enter further experiments and to assess its suitability for use in a therapeutic composition for the treatment of disorders and diseases, such as cancers, additional data are collected. Such data can include the in vitro inhibition of the target molecule, such as a kinase, as measured by IC50, or inhibition of proliferation across a panel of tumor cell lines, and tumor growth inhibition or reduction data and survival data from in vivo animal models. Furthermore, such experiments may also include the elucidation and/or determination of the mechanism of action of the subject compound, the target or target profile of the subject compound, and other characteristics of the subject compound, such as the binding affinity of the compound to the target(s) or the binding site of the compound on the target(s) and pharmacokinetic properties. Such experiments may also include molecular modelling of the drug-target interaction and the identification of metabolites formed after administration.
The compound that shows the most appropriate results for IC50 for target inhibition, inhibition of cell proliferation, spectrum across various tumor cell lines, inhibition of tumour growth or tumour reduction data and/or animal-survival data, and/or other features, including ADME, pharmacokinetic and pharmacodynamic properties, may be chosen to enter further experiments. Such experiments may include, for example, therapeutic profiling and toxicology in animals, phase I clinical trials in humans and other clinical trails.
***
One skilled in the art readily appreciates that the present invention is well adapted to carry out the objects and obtain the ends and advantages mentioned, as well as those inherent therein. The methods and reagents described herein are representative of preferred embodiments, are exemplary, and are not intended as limitations on the scope of the invention. Modifications therein and other uses will occur to those skilled in the art. These modifications are encompassed within the spirit of the invention and are defined by the scope of the claims.
Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims. Those skilled in the art will also recognize that all combinations of embodiments, combination of aspects or features of the claims described herein are within the scope of the invention. Table 1 : Exemplary compounds of the present invention
Compound
Structure Compound name number
6-(2,6-Dichlorophenyl)-2-(3- hydroxymethylphenylamino)- 8 -methoxy-pyrido [2 , 3 -
Figure imgf000097_0001
d]pyrimidin-7-one
6-(2,6-Dichlorophenyl)-8- methoxy-2-(3- methylthiophenylamino)- pyrido[2,3-d]pyrimidin-7-one
Figure imgf000097_0002
6-(2,6-Dichlorophenyl)-8- methoxy-2-(3- methoxyphenylamino)-
Figure imgf000097_0003
pyrido[2,3-d]pyrimidin-7-one
6-(2,6-Dichlorophcnyl)-8- methoxy-2-(3- sulfamoylphenylamino)- pyrido[2,3-d]pyrimidin-7-one
6-(2-Chlorophenyl)-8- methoxy-2-(3- hydroxymethylphenylamino)-
Figure imgf000097_0004
pyrido[2,3-d]pyrimidin-7-one
Figure imgf000098_0001
91
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
6-(2-Chloro-5-((3- trifluoromethyl)benzoylamin o) phenyl)-8-methoxy-2- phenyl amino) -pyrido [2 ,3 - d]pyrimidin-7-one
6-(2-Chloro-5-(3- chlorobenzoylamino)phenyl)- 8-methoxy-2-phenylamino)- pyrido[2,3-d]pyrimidin-7-one
6-(2-Chloro-5-(4- chlorobenzoylamino)phenyl)- 8-methoxy-2-phenylamino)- pyrido[2,3-d]pyrimidin-7-one
6-(2,6-Dimethylphenyl)-8- methoxy-2-(4-(4- methylpiperazino)- phenylamino)-pyrido[2,3- d]pyrimidin-7-one
6-(2-Chloro-6- methoxyphenyl)-8-methoxy-
2-(4-(4-methylpiperazino)- phenylamino)-pyrido[2,3- d]pyrimidin-7-one
8-(4-Aminobutoxy)-6-(2,6- dichlorophenyl)-2-(3- sulfamoylphenyl amino)- pyrido[2,3-d]pyrimidin-7-one
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000111_0001
Figure imgf000112_0001
Figure imgf000113_0001
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
Figure imgf000131_0001
Figure imgf000133_0001
Figure imgf000134_0001
Figure imgf000135_0001
Figure imgf000136_0001
Figure imgf000137_0001
Figure imgf000138_0001
Figure imgf000139_0001
Figure imgf000140_0001
Figure imgf000141_0001
Figure imgf000142_0001
Figure imgf000143_0001
6-(2-Chloro-6-fluorophenyl)-
8-methoxy-2-(4-(4-
217 propylpiperazino)phenyl)- amino-pyrido[2,3- d]pyrimidin-7-one
6-(2-Chloro-6-fluorophenyl)-
8-methoxy-2-(3-
218 hydroxymethyl-4- piperazinophenyl)-amino- pyrido[2,3-d]pyrimidin-7-one
6-(2,6-Dichlorophenyl)-2-(3- hydroxyphenyl)-amino-8-
219 methoxy-pyrido[2,3- d]pyrimidin-7-one
6-(2-Chloro-6-fluorophenyl)-
8-methoxy-2-(5-N-(2-
(pyridin-3-
220 ylcarbonyloxy)ethyl-N- τnethyl-aminocarbonyl-1 - methyl-pyrrol-3-yl)-amino-
Figure imgf000144_0001
pyrido[2,3-d]pyrimidin-7-one
Figure imgf000145_0001
Figure imgf000146_0001
Figure imgf000147_0001
Figure imgf000148_0001
Figure imgf000149_0001
Table 2/Table 3: IC5O values of exemplary compounds (columns 2 and 3); and inhibition of tumor cell growth (column 4)
Figure imgf000150_0001
Figure imgf000151_0001
Figure imgf000152_0001
Figure imgf000153_0001
Figure imgf000154_0001
Figure imgf000155_0001
Figure imgf000156_0001
Figure imgf000157_0001
Figure imgf000158_0001
Figure imgf000159_0001
Figure imgf000160_0001
Equivalents
Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.
All of the above-cited references and publications are hereby incorporated by reference.

Claims

CLAIMS:
1. A compound having a structure represented by formula (I)
Figure imgf000161_0001
(I) or any tautomeric or stereoisomer^ form thereof, wherein
R1 is selected from hydrogen, -Ci-6alkyl, -C2-6-alkenyl, -C2-6-alkynyl, -C3-6- cycloalkyl and -C3-6-cycloalkenyl;
V is selected from a bond, -O-, -N(R1 1)-, -C(=X)-, -S(O)n-, -C(=X)-O-, - C(=X)-N(R")-, -C(=X)-S-, -C(^X)-N(R1 ')-N(Rn)-, -N(RΠ)-C(=X)-, -N(R1 1)- CC=X)-N(R1 ')-, and -N(R1 ')-S(O) „-, with n = 1 or 2;
R2 is selected from hydrogen, -alkyl, -alkenyl, — alkynyl, -cycloalkyl, - cycloalkenyl, -heterocycloalkyl, — heterocycloalkenyl, -aryl and —heteroaryl; or, R1 and R2, together with V and the nitrogen atom they arc attached to, form a heterocycle;
R3 is selected from hydrogen, -Ci-6alkyl, -C2-6-alkenyl, -C2-6-alkynyl, -C3-6- cycloalkyl, -C3-6-cycloalkenyl and halogen;
W is selected from a bond, -C(=O)-, -O-, and -N(R1 ')-;
R4 is selected from hydrogen, —alkyl, -alkenyl, -alkynyl, -cycloalkyl, - cycloalkenyl, -heterocycloalkyl, -heterocycloalkenyl, -aryl and -heteroaryl;
R5 is selected from -alkyl, -alkenyl, -alkynyl, -cycloalkyl, -cycloalkenyl, - (C-linked-heterocycloalkyl), -(C-linked-heterocycloalkenyl), -aryl, and -heteroaryl;
X is independently selected from =O, =S, =NR12, =N-OR13, =N-N(R! ')2, =N- N(R1 1XR12), and =N-N(R12)2; R10 is independently selected from -C^aHcyl, -C2-6-alkenyl, -C2-6-alkynyl, - C3-6-cycloalkyl and -Cs^-cycloalkenyl;
R1 1 is independently selected from hydrogen and R10;
R12 is independently selected from -alkyl, -alkenyl, -alkynyl, -cycloalkyl, - cycloalkenyl, -heterocycloalkyl, -heterocycloalkenyl, -aryl and -heteroaryl;
R13 is independently selected from hydrogen and R12; wherein R2, R4, R5, R10, and R12 may optionally be substituted; or any pharmaceutically acceptable salt or N-oxide thereof.
2. A compound of claim 1 , wherein one or more hydrogen atoms in any of R2, R4, R5, R10, and R12 are independently substituted with substituents R6, with R6 being independently taken from the list of: Y-R14 and R15; with R14 being independently selected from -R13, -OR13, -SR13, -N(R13)2, - N(R13)N(R13)2, -N=C(R13)2, and -N=NR13; with R15 being independently selected from — F, -Cl, -Br5 -I, -CN, -NO2, and
with Y being independently selected from a bond, -C(=Z)-, -O-, -O-C(=Z)-, - N(R'3)-, -N(RI 3)-C(=Z)-, -N(RI 3)-N(R13)-C(=Z)-, -N(R13)-S(O) „-, -S-, and -S(O)n-, with n = 1 or 2; provided that if Y is a bond, then R14 is not hydrogen; and with Z being independently selected from =O, =S, =NR12, =N-OR13, and =N- N(R1 ')2.
3. A compound of claim 1 , wherein one or more hydrogen atoms in any of R2, R4, R5, R10, and R12 are independently substituted with substituents R7, with R7 being independently taken from R6, wherein one or more hydrogens of R6 are substituted by substituents independently taken from the list of: Y-R14 and R15.
4. A compound of any one of claims 1 to 3, wherein R1 is hydrogen.
5. A compound of any one of claims 1 to 4, wherein V is a bond.
6. A compound of claim 5, wherein R is selected from — aryl and — heteroaryl, substituted with 0, 1, 2, 3, 4 or 5 substituents R8, wherein R8 is independently selected from R6 and R7.
7. A compound of claim 6, wherein R is —phenyl substituted with one substituent R8 in position 3 or 4.
8. A compound of claim 6 or 7, wherein any R8 is independently selected from -O-Ci-3-alkyl, -S-Ci-3-alkyl, -Ci-3-alkyl-OH, -SO2-NH2, and -N-linked- heterocycloalkyl.
9. A compound of any one of claims 1 to 8, wherein R3 is hydrogen.
10. A compound of any one of claims 1 to 9, wherein W is a bond.
11. A compound of claim 10, wherein R4 is selected from —aryl and —heteroaryl, and is substituted with 0, 1, 2, 3, 4, or 5 substituents R9, wherein R9 is independently selected from R6 and R7.
12. A compound of claim 11, wherein R4 is -phenyl that is substituted with 0, 1, 2, 3, 4, or 5 substituents R9.
13. A compound of claim 12, wherein R4 is —phenyl that is substituted with one substituent R9 in position 2 or 3.
14. A compound of claim 13, wherein R9 is selected from —methyl, -O-Me, -CF3, N(R13)2, -NH-C(=X)-R13 and halogen.
15. A compound of claim 11 , wherein R4 is -phenyl that is substituted with two substituents R9 in positions 2 and 5 or 2 and 6.
16. A compound of claim 15, wherein said two substituents R9 are independently selected from -methyl, -O-Me, -CF3, N(R13)2, -NH-C(=X)-R13 and halogen.
17. A compound of claim 16, wherein R4 is -phenyl that is substituted with two -Cl substituents in positions 2 and 6.
18. A compound of any one of claims 1 to 17, wherein X is =O.
19. A compound of any one of claims 1 to 18, wherein R5 is selected from R10 and phenyl, in each case substituted with 0, 1, 2, or 3 substituents R , wherein R is independently selected from R and R .
20. A compound of claim 19, wherein R5 is -Ci^-alkyl substituted with 0 or 1 substituent R16.
21. A compound of claim 20, wherein R5 is —methyl.
22. A compound of claim 1 having a structure represented by formula (Ia)
Figure imgf000164_0001
(Ia) or any tautomeric or stereoisomeric form thereof, wherein 1 and m are independently selected from 0, 1, 2, 3, 4, and 5; and
R5 is Ci-6-alkyl substituted with 0, 1, 2 or 3 substituents R16.
23. A compound of claim 22, wherein 1 is 1 or 2, and R8 is independently selected from -O-C,-3-alkyl, -S-C,-3-alkyl, -Ci_3-alkyl-OH, -SO2-NH2, and -N- linked-heterocycloalkyl.
24. A compound of claim 22 or 23, wherein m is 1 or 2, and wherein R9 is independently selected from -Ci-β-alkyl, -C^-alkenyl, -C2-6-alkynyl, — O-Ci-6- alkyl, -CF3, -N(RI3)2, -NH-C(=X)-R13, -NO2, and halogen.
25. A compound of claim 24, wherein m is two, and wherein the two substituents R9 are in positions 2,5 or 2,6, and are independently selected from methyl, -O-Me, -CF3, -N(R13)2, -NH-C(=X)-R13 and halogen.
26. A compound of claim 25, wherein both substituents R9 are -Cl substituents in positions 2,6.
27. A compound of any one of claims 22 to 26, wherein R5 is C^-alkyl substituted with 0 or 1 substituent R16.
28. The compound of claim 27, wherein R5 is methyl.
29. A compound of claim 1, wherein the compound is selected from: 6-(2,6-Dichlorophenyl)-2-(3-hydroxymethylphenylamino)-8-methoxy-pyrido[2,3- d]pyrimidin-7-one; 6-(2,6-Dichlorophenyl)-8-mcthoxy-2-(3-methylthiophenylamino)-pyrido[2,3- d]pyrimidin-7-one; 6-(2,6-Dichlorophenyl)-8-methoxy-2-(3-methoxyphenylamino)-pyrido[2,3- d]pyrimidin-7-one; 6-(2,6-Dichlorophenyl)-8-methoxy-2-(3-sulfamoylphenylamino)-pyrido[2,3- d]pyrimidin-7-one; 6-(2-Chlorophenyl)-8-methoxy-2-(3-hydroxymethylphenylamino)-pyrido[2,3- d]pyrimidin-7-one; 6-(2-Chlorophenyl)-8-methoxy-2-(3-sulfamoylphenylamino)-pyrido[2,3- d]pyrirnidin-7-one; 6-(2-Chlorophenyl)-8-methoxy-2-(3-methoxyphenylamino)-pyrido[2,3-d]pyrimidin-
7-one; 6-(2-Chloro-6-fluorophenyl)-8-methoxy-2-(4-(2-dimethylaminoethoxy)- phcnylamino)-pyrido[2,3-d]pyrimidin-7-one; -(5-Benzoylamino-2-chloro-phenyl)-8-methoxy-2-(4-(4-methylpiperazino)- phenylamino)-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(3-(2-hydroxyethylsulfonyl)phenylamino)- pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(4-methylsulfonylphenylamino)-pyrido[2,3- d]pyriτnidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-((4-methoxycarbonyl-3-methylpyrrol-3- yl)amino)-pyτido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(pyrid-4-ylamino)-pyrido[2,3-d]pyrimidin-7- one; -(2,6-Dichlorophenyl)-8-methoxy-2-(4-(4-methylpiperazino)-phenylamino)- pyrido[2,3-d]pyrimidin-7-one; -(2-Chloro-6-fluorophenyl)-8-methoxy-2-(3-hydroxymethylphenylamino)- pyrido[2,3-d]pyrimidin-7-one; -(2,4-Dichlorophenyl)-8-methoxy-2-(4-(2-dimethylaminoethoxy)-phenylamino)- pyrido[2.,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(4-(2-hydroxyethyl)-phenylamino)-pyrido[2,3- d]pyrimidin-7-one; -(3,4-Dichlorophenyl)-8-methoxy-2-(4-(4-methylpiperazino)-phenylamino)- pyrido[2,3-d]pyrimidin-7-one; -(2,4-Dichlorophenyl)-8-methoxy-2-(4-(4-methylpiperazino)-phenylamino)- pyrido[2,3-d]pyrimidin-7-one; -(2-Chlorophenyl)-8-(2-meth'oxyethoxy)-2-phenylamino)-pyrido[2,3-d]pyrimidin-
7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(3-(pyrrolidin-l-yl)methylphenylamino)- pyrido[2,3-d]pyrimidin-7-one; -(2-Chlorophenyl)-8-methoxy-2-phenylamino-pyrido[2,3-d]pyrimidin-7-one; -(5-Amino-2-chlorophenyl)-8-methoxy-2-phenylamino-pyrido[2,3-d]pyrimidin-7- one; -(2,6-Dichlorophenyl)-8-methoxy-2-(4-(2-dimethylaminoethoxy)-phenylamino)- pyrido[2,3-d]pyrimidin-7-one; -(2-Chloro-6-fluorophenyl)-8-methoxy-2-(4-(4-methylpiperazino)-phenylamino)- pyrido[2,3-d]pyrimidin-7-one; -(2-Chloro-5-(pyrid-4-ylcarbonylamino)phenyl)-8-methoxy-2-(4-(4- methylpiperazino)-phenylamino)-pyrido[2)3-d]ρyrimidin-7-one; -(2,6-Dichlorophenyl)-2-(2-fluoro-5-(hydroxymethyl)phenylamino)-8-methoxy- pyrido[2,3-d]pyrimidin-7-one; -(3-Benzoylaminophenyl)-8-methoxy-2-phenylamino)-pyrido[2,3-d]pyrimidin-7- one; -(5-Benzoylamino-2-chloro-phenyl)-8-methoxy-2-phenylamino)-pyrido[2,3- d]pyrirnidin-7-one; -(2-Chloro-6-fluorophenyl)-8-methoxy-2-(3-sulfamoylphenylamino)-pyrido[2,3- d]pyrimidin-7-one; -(2-Chloro-5-(pyrid-3-ylcarbonylamino)phenyl)-8-methoxy-2-phenylamino)- pyrido[2,3-d]pyrimidin-7-one; -(2-Cliloro-5-(dimethylacetylamino)phenyl)-8-methoxy-2-phenylamino)- pyrido[2,3-d]pyrimidin-7-one; -(5-Benzoylamino-2-chloro-phenyl)-8-methoxy-2-(2-methoxyethyl)amino)- pyrido[2,3-d]pyrimidin-7-one; -(4-Aminobutoxy)-6-(2,6-dichlorophenyl)-2-phenylamino-pyrido[2,3-d]pyrimidin-
7-one; -(2-Chloro-5-((3-trifluoromethyl)benzoylamino) phenyl)-8-methoxy-2- phenylamino)-pyrido[2,3-d]pyrimidin-7-one; -(2-Chloro-5-(3-chlorobenzoylamino)phenyl)-8-methoxy-2-phenylamino)- pyrido[2,3-d]pyrimidin-7-one; -(2-Chloro-5-(4-chlorobenzoylamino)phenyl)-8-methoxy-2-phenylamino)- pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dimethylphenyl)-8-mcthoxy-2-(4-(4-mcthylpipcrazino)-phcnylamino)- pyrido[2,3-d]pyrimidin-7-one; -(2-Chloro-6-methoxyphenyl)-8-methoxy-2-(4-(4-methylpiperazino)- phenylamino)-pyrido[2,3-d]pyrimidin-7-one; -(4-Aminobutoxy)-6-(2,6-dichlorophenyl)-2-(3-sulfamoylphenylamino)- pyrido[2,3-d]pyrimidin-7-one; -(4-Aminobutoxy)-6-(2,6-dichlorophenyl)-2-(3-methoxyphenylamino)-pyrido[2,3- d]pyrimidin-7-one; -(2-Chloro-6-fluorophenyl)-8-dimethylmethoxy-2-(4-(4-methylpiperazino)- phenylamino)-pyrido[2,3-d]pyrimidin-7-one; -(3-Hydroxyinethylphenylarnino)-8-πiethoxy-6-phenyl-pyrido[2,3-d]pyriniidin-7- one; -(2,5-Dimethoxyphenyl)-2-(3-hydroxymethylphenylamino)-8-methoxy-pyrido[2,3- d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-2-((2-iτiethyl-5-hydroxymetliylphenyl)-amino)-8-methoxy- pyrido [2,3 -d]pyrimidin-7-one; -Amino-6-(2,6-dichlorophenyl)-8-methoxy-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(4-methylpiperidino-amino)-pyrido[2,3- d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-methoxyethylamino-pyrido[2,3-d]pyrimidin-7- one; -(2-Chlorophenyl)-8-cyclopropylmethoxy-2-phenylamino-pyrido[2,3-d]pyrimidin-
7-one; -(4-(2-Dimethylaminoethoxy)-6-(2-methoxyphenyl)-phenylamino)-8-methoxy- pyrido[2,3-d]pyrimidin-7-one; -(2-Chloro-6-fluorophenyl)-8-ethoxy-2-(4-(4-methylpiperazino)-phenylamino)- pyrido[2,3-d]pyrimidin-7-one; -(2-Chloro-6-fluoroρhenyl)-8-cyclopropylmethoxy-2-(4-(4-methylpiperazino)- phenylamino)-pyrido[2,3-d]pyrimidin-7-one; -(2-Fluoro-6-trifluoromethyl-phenyl)-8-methoxy-2-(4-(4-methylpiperazino)- phenylamino)-pyrido[2,3-d]pyrimidin-7-one; and -(5-Carboxy-l-methyl-pyrrol-3-yl)-amino-6-(2,6-dichlorophenyl)-8-methoxy- pyrido[2,3-d]pyrimidin-7-one. -(3-Aminopropyl)oxy-6-(2,6-dichlorophenyl)-2-phenylamino-ρyrido[2,3- d]pyrimidin-7-one; -(5-Aminopentyl)oxy-6-(2;6-dichlorophenyl)-2-phenylamino-pyrido[2,3- d]pyrimidin-7-one; -(3-Acetylaminopropyl)oxy-6-(2,6-dichlorophenyl)-2-phenylamino-pyrido[2,3- d]pyrimidin-7-one; -(2-(2-Aminoethyloxy)ethyl)oxy-6-(2,6-dichlorophenyl)-2-phenylamino- pyrido[2 ,3 -d]pyrimidin-7-one; -(2-Chloro-5-acetylaminophenyl)-8-methoxy-2-phenylamino)-pyrido[2,3- d]pyrimidin-7-one; -(2,5-Dimethoxyphenyl)-8-methoxy-2-phenylamino)-pyrido[2,3-d]pyrimidin-7- one; -Methoxy-2-phenylamino-6-phenylaτninocarbonyl-pyrido[2,3-d]pyrimidin-7-one;-(3-Acetylaminophenyl)-8-methoxy-2-phenylamino)-pyrido[2-3-d]pyrimidin-7- one; -(2-Chlorophenyl)-8-(l , 1 -dimethyl)ethyloxy-2-phenylamino-pyrido[2,3- d]pyrimidin-7-one; -(3-Aminosulfonlyphenyl)-amino-6-(3,4-dichlorophenyl)-8-methoxy-pyrido[2,3- d]pyrimidin-7-one; -(2-Chlorophenyl)-8-(l-methylethyl)oxy-2-phenylamino)-pyrido[2,3-d]pyrimidin-
7-one; -(4-Aminobutyl)oxy-6-(2,6-dichlorophenyl)-2-phenylamino-pyrido[2,3- d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-2-(5-(2-dimethylaminoethyl)aminocarbonyl-l-methyl- pyrrol-3-yl)-amino-8-mcthoxy-pyrido[2,3-d]pyrimidin-7-onc; -(2,6-Dichlorophenyl)-8-(2-methoxyethyl)oxy-2-(3-methoxyphenyl)amino- pyrido[2 ,3 -d]pyrimidin-7-onc; -(2,6-Dimethylphenyl)-8-methoxy-2-(3-methoxyphenyl)-amino-pyrido[2,3- d]pyrimidin-7-one; -(2-Aminoethyl)oxy-6-(2,6-dichlorophenyl)-2-(3-methoxyphenyl)amino- pyrido[2,3-d]pyrimidin-7-one; -(3-Aminopropyl)oxy-6-(2,6-dichlorophenyl)-2-(3-methoxyphenyl)amino- pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dimethylphenyl)-8-methoxy-2-(3-sulfamoylphenylamino)- pyrido[2,3- d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-(2-hydroxyethyl)oxy-2-(3-methoxyphenyl)amino- pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-(2-methylaminoethyl)oxy-2-(3-methoxyphenyl)amino- pyrido[2,3 -d]pyrimidin-7-one; -(2-Chloro-6-fluorophenyl)-8-(2-(S)-2,3-dihydroxypropyl)oxy-2-(3- methoxyphenyl)amino-pyrido[2,3-d]pyrimidin-7-one; -(2-Chloro-6-fluorophenyl)-8-(2~(R)-2,3-dihydroxyproρyl)oxy-2-(3- methoxyphenyl)amino-pyrido[2,3-d]pyrimidϊn-7-one; -(2,6-Dichlorophenyl)-8-(2-dimethylaminoethyl)oxy-2-(3-methoxyphenyl)amino- pyrido [2 , 3 -d] pyrimidin-7-one; -(2J6-Dichlorophenyl)-8-(2-dimethylaminopropyl)oxy-2-(3-methoxyphenyl)amino- pyrido[2,3-d]pyτimidin-7-one; -(2,6-Dimethylphenyl)-8-methoxy-2-(5-(methoxycarbonyl-l-methyl-pyrrol-3-yl)- amino-pyrido[2,3-d]pyrimidin-7-one; -Cyclopropylcarbonylamino-6-(2,6-dichlorophenyl)-8-mcthoxy-pyrido[2,3- d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-2-(5-(2-diethylaminoethyl)aminocarbonyl-l -methyl-pyrrol-
3-yl)-amino-8-mcthoxy-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-2-(5-(2-hydroxyethyl)aminocarbonyl-l-methyl-pyrrol-3-yl)- amino-8-methoxy-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dimethylphenyl)-2-(5-(2-hydroxyethyl)aminocarbonyl-l-methyl-pyrrol-3- yl)-amino-8-methoxy-pyrido[2,3-d]pyrimidin-7-one; -(256-Dimethylphenyl)-2-(5-(2-diethylaminoethyl)aniinocarbonyl-l -methyl-pyrrol-
3-yl)-amino-8-rnethoxy-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-2-(isoxazol-3-yl)-amino-8-methoxy-pyrido[2,3-d]pyrimidin-
7-one; -(4-Cyanophenyl)-amino-6-(2,6-dichlorophenyl)-8-methoxy-pyrido[2,3- d]pyrimidin-7-one; -(2,6-Dimethylphenyl)-8-methoxy-2-(5-(2-pyrrolidinoethyl)aminocarbonyl-l- methyl-pyrrol-3-yl)-amino-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(pyrazol-3-yl)-amino-pyrido[2,3-d]pyrimidin-
7-one; 6-(2,6-Dichlorophenyl)-2-(4-(2-hydroxyethyl)oxyphenyl)amino-8-methoxy- pyrido[2,3-d]pyrimidin-7-one; 6-(2,6-Dichlorophenyl)-8-methoxy-2-(l-thia-3,4-diazol-2-yl)-amino-pyrido[2,3- d]pyrimidin-7-one; •6-(2,6-Dichlorophenyl)-8-raethoxy-2-(4-(2-pyrrolidinoethyl)oxyphenyl)amino- pyrido[2,3-d]pyrimidin-7-one; 6-(2,6-Dichlorophenyl)-2-(4-(2-(3-(S)-hydroxypyiTolidino)ethyl)oxyphenyl)amino-
8-methoxy-pyrido[2,3-d]pyrimidin-7-one; 6-(2,6-Dichlorophenyl)-2-(4-(2,3-dihydroxypropyl)oxyphenyl)amino-8-methoxy- pyrido[2,3-d]pyrimidin-7-one; 6-(2,6-Dichlorophenyl)-8-methoxy-2-(5-(2-pyrrolidinoethyl)aminocarbonyl-l- niethyl-pyrrol-3-yl)-amino-pyrido[2,3-d]pyrimidin-7-one; 6-(2,6-Dichlorophenyl)-8-methoxy-2-(5-(2-pyrrolidinopropyl)aminocarbonyl-l- methyl-pyrrol-3-yl)-amino-pyrido[2,3-d]pyrimidin-7-one; 2-But-2-enoylamino-6-(2,6-dimethylphenyl)-8-methoxy-pyrido[2,3-d]pyrimidin-7- one; 2-(4-cyanomethylphenyl)-aτnino-6-(2,6-dichlorophenyl)-8-methoxy-pyrido[2,3- d]pyrimidin-7-one; 6-(2,6-Dichlorophenyl)-8-methoxy-2-(4-morphoiinophenyl)-amino-pyrido[2,3- d]pyrimi din-7-one; 6-(2,6-Dichlorophenyl)-8-(2-methoxyethyl)oxy-2-(5-(2- pyrrolidinoethyl)aminocarbonyl-l-methyl-pyrrol-3-yl)-amino-pyrido[2,3- d]pyrimidin-7-one; 6-(2,6-Dichlorophenyl)-8-methoxy-2-(4-morpholinomethylphenyl)-amino- pyrido[2,3-d]pyrimidi n-7-one; 6-(2,6-Dichlorophenyl)-8-methoxy-2-(4-pyrrolidinomethylphenyl)-amino- pyrido[2,3-d]pyrimidin-7-one; 6-(2,6-Dichlorophenyl)-2-hydroxyethylamino-8-methoxy-pyrido[2,3-d]pyrimidin-7- one; 6-(2,6-Dichloroρhenyl)-8-(2-(S)-2,3-dihydroxypropyl)oxy-2-(5-(2- pyrrolidinoethyl)aminocarbonyl-l-methyl-pyrrol-3-yl)-amino-pyrido[2,3- d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(4-(l,2,4-triazol-l-yl)methylphenyl)-amino- pyrido [2,3 -d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-raethoxy-2-(4-pyrrolidinophenyl)-amino-pyrido[2,3- d]pyrimidin-7-one; -(5-Benzoylamino-2-chloro-phenyl)-8-(2-methoxyethyl)oxy-2-(4-(4- methylpiperazino)-phenylamino)-pyrido[2,3-d]pyτimidin-7-one; -(2-Methoxyethyl)oxy-2-(4-(4-methylpiperazino)-6-(5-(3- trifluoromethylbenzoyl)aτnino-2-chloro-phenyl)-phenylamino)-pyrido[2,3- d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-(2-(R)-2,3-dihydroxypropyl)oxy-2-(5-(2- pyrrolidinoethyl)aminocarbonyl-l-methyl-ρyrrol-3-yl)-aπiino-pyrido[2,3- d]pyrimidin-7-one; -((2-(S)-2-Amino-3-methylbutanoyloxy)ethyl)-amino-6-(2,6-dichlorophenyl)-8- methoxy-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(4-(2-oxopyrrolidino)phenyl)-amino- pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-mcthoxy-2-(4-methylstjlfonylaminophenyl)-amino- pyτido[2,3-d]pyrimidin-7-one; -(5-(2-(2-(S)-2-Amino-3-methylbutanoyloxy)ethyl)aminocarbonyl-l-methyl- ρyrrol-3-yl)-amino-6-(2,6-dichlorophenyl)-8-methoxy-pyrido[2,3- d]pyrimidin~7-one; -Cyclopropylamino-6-(2,6-dichlorophenyl)-8-τnethoxy-pyrido[2,3-d]pyrimidin-7- one; -(2,6-Dimcthylphcnyl)-8-methoxy-2-pyrid-3-ylamino-pyrido[2,3-d]pyrimidin-7- one; -(2,6-Dichlorophcnyl)-8-methoxy-2-(4-(2- pyrrolidinoetbylaminocarbonylmethyl)phenyl)-amino-pyrido[2,3- d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-2-(5-(lM-(2-hydroxyethyl)-N -methyl- amino)carbonyl- 1 - methyl-pyrrol-3-yl)-amino-8-methoxy-pyrido[2,3-d]pyrimidin-7-one;-(2,6-Dichlorophenyl)-2-(5-(2-(R)-2,3-dihydroxyethylamino)carbonyl-l-methyl- pyrrol-3-yl)-amino-8-methoxy-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-2-(5-(2-(S)-2,3-dihydroxyethylamino)carbonyl-l-τnethyl- pyrrol-3-yl)-amino-8-methoxy-pyrido[2,3-d]pyriraidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(3-methylsulfonylaminophenyl)-amino- pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(4-methylsυlfonylaininomethylphenyl)-amino- pyrido[2,3-d]pyrirnidin-7-one; -(2-Chloro-6-fluorophenyl)-8-(2-methoxyethyl)oxy-2-(4-morpholinophenyl)- amino)-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-2-(3-ethylaminosulfonylphenyl)-amino-8-methoxy- pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichloroρhenyl)-2-(3-diethylaminosulfonylphenyl)-amino-8-methoxy- pyrido[2,3-d]pyrimidin-7-one; -(2,6-dichlorophenyl)-8-methoxy-2-(4-(pyrazol-l-ylmethyl)phenyl)-amino- pyrido[2,3-d]pyrimidin-7-one; -(2,6-dichlorophenyl)-8-methoxy-2-(4-(τnethylaminosulfonylmethyl)phenyl)- amino-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-2-(3-(2-hydroxyethyl)aminosulfonylphenyl)-amino-8- methoxy-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-2-(3-morpholinosulfonylphenyl)-amino-8-methoxy- pyrido[2,3-d]pyrimidin-7-one; -(2-Chloro-6-fluorophenyl)-8-ethoxy-2-(4-moφholinophenyl)-amino)-pyrido[2,3- d]pyrimidin-7-one; -(2-Chloro-6-fluorophenyl)-8-(cyclopropylmethyl)oxy-2-(4-morpholinophenyl)- amino)-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(3-tetrazol-5-ylphenyl)-amino-pyrido[2,3- d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(3-methylaτninocarbonylphenyl)-amino- pyrido[2,3-d]pyrimidin-7-one; -(2-Chloro-6-fluorophenyl)-8-(pyrid-3-ylmethyl)oxy-2-(4-moφholinophenyl)- amino)-pyrido[2,3-d]pyrimidin-7-one; -(3-Chloro-4-trifluoromethylphenyl)-aτnino-6-(2,6-dichlorophcnyl)-8-methoxy- pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(3 -( 1 ,2,4-triazol- 1 -ylmethyl)phenyl)-amino- pyrido [2,3 -d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(pyrimidin-4-yl)-amino-pyrido[2>3- d]pyrimidin-7-one; -(2-Chloro-6-fluorophenyl)-8-methoxy-2-(4-morpholinophenyl)-amino)- pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(5-morpholinocarbonyl-l-methyl-pyrrol-3-yl)- ami no-pyrido [2,3 -d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(4-N-(2-hydroxyethyl)-N-methyl- aminocarbonyl-thiazol-2-yl)-amino-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-raethoxy-2-(5-N-(2-hydroxyethyl)-N-methyl- • aminocarbonyl-thiophen-3-yl)-amino-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(5-N-(2-(2,2-dimethylpropanoyl)oxyethyl)-N- methyl-aminocarbonyl-l -methyl-pyrrol-3-yl)-amino-pyrido[2,3-d]pyrimidin-
7-one; -(5-N-(2-(Benzoyloxyethyl)-N-methyl-aminocarbonyl-l-methyl-pyrrol-3-yl)- amino-6-(2,6-dichlorophenyl)-8-methoxy-pyrido[2,3-d]pyrimidin-7-one;-(2,6-Dichlorophenyl)-8-methoxy-2-(5-(4-methylρiperazino)carbonyl-thiophen-3- yl)-amino-pyrido [2,3 -d]pyrimidin-7-one; -(2-Chloro-6-fluorophenyl)-2-(4-morpholinophenyl)-8-(tetrahydropyran-4- ylmethyl)oxy-amino)-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-2-(4-hydroxymethylphenyl)-aminor8-methoxy-pyrido[2,3- d] pyrimidi n-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-phenylmethylamino-pyrido[2,3-d]pyrimidin-7- one; -(2,6-Dichlorophenyl)-8-methoxy-2-pyrid-3-ylmethylamino-pyrido[2,3- d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-pyrid-4-ylmethylamino-pyrido[2,3- d]pyrimidin-7-one; -(2-Chloro-6-fluorophenyl)-8-(2-(S)-2,3-dihydroxypropyl)oxy-2-(4- morpholinophenyl)-amino)-pyrido[2,3-d]pyrimidin-7-one; -(2-Chloro-6-fluorophenyl)-8-(3-(R)-pyrrolidin-3-ylmethyl)oxy-2-(4- moφholinophenyl)-amino)-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-2-(4-methylphenyl)-amino-8-methoxy-pyrido[2,3- d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(4-(5-methyl-l,2,4-triazol-3-yl)methylphenyl)- amino-pyrido[2,3-d]pyrimidin-7-one; -(2-Chloro-6-fluoroρhenyl)-8-(2-(R)-2,3-dihydroxypropyl)oxy-2-(4- morpholinophenyl)-amino)-pyrido[2,3-d]pyrimidin-7-one; -(2-Chloro-6-fluorophenyl)-2-phenylamino-8-(pyrid-3-ylmethyl)oxy-pyrido[2,3- d] pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(5-N-(2-methoxyethyl)-N-methyl- aminocarbonyl-l-methyl-pyrrol-3-yl)-amino-pyrido[2,3-d]pyrimidin-7-one;-(2-Chloro-6-fluorophenyl)-8-(3-(S)-pyrrolidin-3-ylmethyl)oxy-2-(4- moφholinophenyl)-amino)-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-2-(3-((2-hydroxyethylamino)sulfonylmethyl)phenyl)-amino-
8-methoxy-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(3-methylsulfonylphenyl)-amino-pyrido[2,3- d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-2-(4-(3-hydroxypropyl)thiophenyl)-amino-8-methoxy- pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(5-N-(2-(pyridin-3-ylcarbonyloxy)ethyl-N- methyl-aminocarbonyl-l-methyl-pyrrol-3-yl)-amino-pyrido[2,3-d]pyrimidin-
7-one; -(2,6-Dichlorophenyl)-2-(4-(3-hydroxypropyl)sulfonylphenyl)-amino-8-methoxy- pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(3-methyliminosulfonylphenyl)-amino- pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-2-(4-methoxycarbonylphenyl)-amino-8-methoxy- pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(5-(4-methylpiperazino)carbonyl-l-methyl- pyrrol-3-yl)-amino-pyrido[2,3-d]pyrimidin-7-one; -(2-Chloro-6-fluoroρhenyl)-2-ρhenylamino-8-(tetrahydropyran-4-yltnethyl)oxy- pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-2-phenylamino-8-(pyrid-3-ylmethyl)oxy-pyrido[2,3- d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(4-(4-(l- methylethyl)piρerazinomethylphenyl)-amino-pyrido[2,3-d]pyrimidin-7-one;-(2,6-Dichlorophenyl)-2-(4-diethylaminomethylphenyl)-amino-8-methoxy- pyrido[2,3-d]pyrimidin-7-one; -(4-(4-hydroxy-l-aza-cyclobutyl)methylphenyl)-amino-6-(2,6-dichlorophenyl)-8- methoxy-pyrido[2,3-d]ρyrimidin-7-one; -(2,6-Dichlorophenyl)-2-(4-(2-(S)-hydroxymethyl-pyrrolidinomethyl)phenyl)- amino-8-methoxy-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-(2-methoxyethyl)oxy-2-(5-N-(2-methoxyethyl)-N-methyl- aminocarbonyl-l-methyl-pyrrol-3-yl)-amino-pyrido[2,3-d]pyrimidin-7-one;-(2,6-Dichlorophenyl)-8-ethoxy-2-(5-N-(2-methoxyethyl)-N-methyl- aminocarbonyl-l-methyl-pyrrol-3-yl)-amino-pyrido[2,3-d]pyrimidin-7-one;-(2-Chloro-6-fluorophenyl)-8-cyclobutylmethyloxy-2-(4-moipholinophenyl)- amino)-pyrido[2,3-d]pyrimidin-7-one; -{2-Chloro-6-fluorophenyl)-8-cyclopentylmethyloxy-2-(4-morpholinophenyl)- amino)-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-phenylamino-pyrido[2,3-d]pyrimidin-7-one;-(4-(l,3,4-triazol-l-yl)methylphenyl)-amino-6-(2,6-dichlorophenyl)-8-methoxy- pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-cyclopropylmethyloxy-2-(5-N-(2-methoxyethyl)-N- methyl-aminocarbonyl-l-methyl-pyrrol-3-yl)-amino-pyrido[2,3-d]pyrimidin-
7-one; -(2,6-dichlorophenyl)-2-(5-N-(2-methoxyethyl)-N-methyl-aminocarbonyl-l- methyl-pyrrol-3-yl)-amino-8-(tetrahydropyran-4-ylmethyl)oxy-pyrido[253- d]pyrimidin-7-one; -(4-(2-hydroxyethylamino)methylphenyl)-amino-6-(2,6-dichlorophenyl)-8- methoxy-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-2-(4-(3-(R)-hydroxypyrrolidinomethyl)phenyl)-amino-S- methoxy-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-2-(4-(3-(S)-hydroxypyrrolidinomethyl)phenyl)-amino-8- methoxy-pyrido[2.,3-d]pyrirnidin-7-one; -(2,6-Dichlorophenyl)-2-(3-(3-(S)-hydroxypyrrolidinomethyl)phenyl)-amino-8- methoxy-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-2-(3-(3-(R)-hydroxypyrrolidinomethyl)phenyl)-amino-8- methoxy-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-2-(3-(3,3-difluoropyrrolidinomethyl)phenyl)-amino-8- methoxy-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(4-piperazinophenyl)-amino-pyrido[2,3- d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(5-piperazinocarbonyl-l-methyl-pyrrol-3-yl)- amino-pyrido[2 ,3 -d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-2-phenylamino-8-(tetrahydropyran-4-ylmethyl)oxy- pyrido[2 ,3 -d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(4-(4-(l-methylethyl)piperazinophenyl)- aτnino-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(5-(4-(2-methoxy)ethyl)piperazinocarbonyI-l- methyl-pyrrol-3-yl)-amino-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophcnyl)-2-(4-(4-ethylpiperazino)phenyl)-amino-8-methoxy- pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-2-(2-fluorophenyl)amino-8-methoxy-pyrido[2,3- d]pyrimidin-7-one; -(4-Bromophenyl)amino-6-(2,6-dichlorophenyl)-8-methoxy-pyrido[2,3- d]ρyrimidin-7-one; -(4-Acetylphenyl)amino-6-(2,6-dichlorophenyl)-8-methoxy-pyrido[2,3- d]pyrimidin-7-one; -(2-Chloro-6-fluorophenyl)-2-(5-N-(2-hydroxyethyl)-N-methyl-aminocarbonyl-l- methyl-pyrrol-3-yl)-amino-8-methoxy-pyrido[2,3-d]pyrimidin-7-one; -(2-Chloro-6-fluorophenyl)-2-(5-N-(2-methoxyethyl)-N-methyl-aminocarbonyl-l- methyl-pyrrol-3-yl)-amino-8-methoxy-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-(2-methoxyethyl)oxy-2-(4-piperazinophenyl)-amino- pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-2-(4-(3,3-difluoropyrrolidinomethyl)phenyl)-amino-8- methoxy-pyrido[2,3-d]pyrimidin-7-one; -(2-Chloro-6-fluorophenyl)-8-methoxy-2-(5-morpholinocarbonyl-l-methyl-pyrrol-
3-yl)-amino-pyrido[2,3-d]pyrimϊdin-7-one; -(2-Chloro-6-fluorophenyl)-8-methoxy-2-(4-piperazinophenyl)-amino-pyrido[2,3- d]pyrimidin-7-one; -(2-Chloro-6-fluorophenyl)-2-(3-chloro-4-piperazinophenyl)-amino-8-methoxy- pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(4-(4-methylsulfonyl)piperazinophenyl)- amino-pyrido[2,3-d]pyrimidin-7-one; -(4-(l-azacyclobutyl)methylphenyl)-amino-6-(2,6-dichlorophenyl)-8-methoxy- pyrido [2 ,3-d]pyrimidin-7 -one; -(2,6-Dichlorophenyl)-8-(tetrahydropyτan-4-ylmethyl)oxy-2-(4-piperazinophenyl)- amino-pyrido[2,3-d]pyrimidin-7-one; -(2-Chloro-6-fluorophenyl)-8-methoxy-2-(4-(4-(l-methylethyl)piperazino)phenyl)- amino-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(4-(4-propylpiperazino)phenyl)-amino- pyrido [2 ,3 -d] pyrimidin-7 -one; -(2,6-Dichlorophenyl)-8-(2-methoxyethyl)oxy-2-(4-(4-(l- methylethyl)piperazino)phenyl)-amino-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-(tetrahydropyran-4-ylmethyl)oxy-2-(4-(4-(l- methylethyl)piperazino)phenyl)-amino-pyrido[2,3-d]pyrimidin-7-one; -(4-(4-propylpiperazino)phenyl)-amino-6-(2,6-dichlorophenyl)-8-(2- τnethoxyethyl)oxy-pyrido[2,3-d]pyrimidin-7-one; -(2-Chloro-6-fluorophenyl)-8-methoxy-2-(5-piperazinocarbonyl-l-methyl-pyrrol-
3-yl)-amino-pyrido[2,3-d]pyrimidin-7-one; -(4-(methoximino)methylphenyl)-amino-6-(2,6-dichlorophenyl)-8-methoxy- pyrido[2,3-d]pyrimidin-7-one; -(4-(hydroximino)methylphenyl)-amino-6-(2,6-dichlorophenyl)-8-methoxy- pyrido[2,3-d]pyrimidin-7-one; -(2-Chloro-6-fluorophenyl)-2-(3-fluoro-4-piperazinophenyl)-aπiino-8-methoxy- pyrido[2,3 -d]pyrimidin-7-one; -(2-Chloro-6-fluorophenyl)-8-(3-(S)-l-methyl-pyrrolidin-3-yl)methoxy-2-(4- morpholinophenyl)-amino-pyrido[2,3-d]pyrimidin-7-one; -(2-Chloro-6-fluorophenyl)-8-methoxy-2-(3-cyano-4-piperazinophenyl)-amino- pyrido[2,3 -d]pyrimidin-7-one; -(2-Chloro-6-fluorophenyl)-8-methoxy-2-(3-methoxy-4-piρerazinophenyl)-amino- pyrido [2,3 -d]ρyrimidin-7-one; -(2-Chloro-6-fltiorophenyl)-8-(tetrahydropyran-4-ylπiethyl)oxy-2-(3-methoxy-4- piperazinophenyl)-amino-pyrido[2,3-d]pyrimidin-7-one; -(2-Chloro-6-fluorophenyl)-8-(2-methoxyethyl)oxy-2-(4-piperazinophenyl)-amino- pyrido[2,3-d]pyrimidin-7-one; -(2-Chloro-6-fluorophenyl)-8-methoxy-2-(4-(4-propylpiperazino)phenyl)-amino- pyrido[2,3-d]pyritnidin-7-one; -(2-Chloro-6-fluorophenyl)-8-methoxy-2-(3-hydroxymethyl-4-piperazinophenyl)- amino-pyrido[2,3-d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-2-(3-hydroxyphenyl)-amino-8-methoxy-pyrido[2,3- d]pyrimidin-7-one; -(2-Chloro-6-fluorophenyl)-8-methoxy-2-(5-N-(2-(pyridin-3-ylcarbonyloxy)ethyl-
N-methyl-aminocarbonyl-1 -methyl -pyrrol-3-yl)-amino-pyrido[2,3- d]pyrimidin-7-one; -(2,6-Dichlorophenyl)-8-methoxy-2-(4-(4-methyl-l,4-diazacycloheptyl)phenyl-)- amino-pyrido[2,3-d]pyrimidin-7-one; -(2-Chloro-6-flυorophenyl)-8-n:iethoxy-2-(4-(piperidin-4-yl)phenyl)-amino- pyrido[2,3-d]pyrimidin-7-one; -(2-Chloro-6-fluorophenyl)-8-methoxy-2-(4-(4-methylsulfonylpiperazino)phenyl)- amino-pyrido[2,3 -d]pyrimidin-7-one; -(2-Chloro-6-fluorophenyl)-2-(5-(piperazinocarbonyl)-l-methyl-pyrrol-3-yl)- amino-8-(tetrahydropyran-4-ylmethyl)oxy -pyrido[2,3-d]pyrimidin-7-one;-Cyclopentyloxy-2-phenylamino)-pyrido[2,3-d]pyrimidin-7-one; -Cyclopentyloxy-2-(4-moφholinoρhenyl)amino)-ρyrido[2,3-d]pyrimidin-7-one; 6-(2,6-Dichlorophenyl)-8-methoxy-2-(3-methoxy-4-piperazino-phenyl)-amino- pyrido[2,3-d]pyrimidin-7-one; 2-(4-(2-(2-aminoethoxy)ethoxyphenyl)-amino-6-(2,6-dichlorophenyl)-8-methoxy- pyrido[2,3-d]pyrimidin-7-one; 6-(2,6-Dichlorophenyl)-2-(3-hydroxymethyl-4-piperazino-phenyl)-amino-8- methoxy-pyrido[2,3-d]pyrimidin-7-one; 6-(2,6-Dichlorophenyl)-8-(2-(S)-2,3-dihydroxypropyl)oxy-2-(4- moφholinophenyl)amino-pyrido[2,3-d]pyrimidin-7-one; 6-(2,6-Dichlorophenyl)-8-methoxy-2-(5-(4-methylpiperazino)carbonyl-l-methyl- pyrrol-3-yl)-amino-pyrido[2,3-d]pyrimidin-7-one; 2-Amino-6-(2,6-dichlorophenyl)-8-(pyrid-3-yl)methoxy-pyrido[2,3-d]pyrimidin-7- one; 8-(l,l-Dimethylethoxy)-2-(4-(4-methylpiperazino)phenyl)amino)-pyrido[2,3- d]pyrimidin-7-one; 8-Cyclopentyloxy-2-(4-(4-methylpiperazino)phenyl)amino)-pyrido[2,3-d]pyrimidin-
7-one; 8-Cyclopentyloxy-2-(4-(2-dimethylaminoethoxy)phenyl)amino)-pyrido[2,3- d]pyrimidin-7-one; 8-Cyclohexyloxy-2-(4-(4-methylpiperazino)phenyl)amino)-pyrido[2,3-d]pyrimidin-
7-one; 8-Cyclohexyloxy-2-(4-(2-dimethylaminoethoxy)phenyl)amino)-pyrido[233- d]pyrimidin-7-one; and 8-Cyclopentyloxy-2-(4-piperazinophenyl)amino-pyrido[2,3-d]pyrimidin-7-one.
30. A prodrug of a compound of any one of claims 1 to 29.
31. A pharmaceutical composition, including a compound of any one of claims 1 to 29, or a prodrug of claim 30, and a pharmaceutically acceptable diluent, excipient or carrier.
32. The pharmaceutical composition of claim 31 , comprising a therapeutically effective amount of said compound or prodrug.
33. The pharmaceutical composition of claim 31 or 32, for the treatment of an individual in need thereof.
34. The pharmaceutical composition of claim 33, wherein said individual is a human.
35. A pharmaceutical package, including a pharmaceutical composition of any one of claims 31 to 34, and instructions which indicate that said pharmaceutical composition may be used for the treatment of an individual in need thereof.
36. The pharmaceutical package of claim 35, wherein said instructions indicate that said pharmaceutical composition may be used for the treatment of a human.
37. The pharmaceutical package of claim 35, wherein said instructions indicate that said pharmaceutical composition may be used for the treatment of an individual suffering from cancer.
38. The pharmaceutical package of claim 37, wherein said instructions indicate that said pharmaceutical composition may be used for the treatment of a human suffering from cancer
39. A method for treating cancer in an individual, comprising administering a therapeutically effective amount of a compound of any one of claims 1 to 29, a prodrug of claim 30, or the pharmaceutical composition of any one of claims 31 to 34.
40. The method of claim 39, wherein said individual is a mammal selected from: domestic mammal, cat, dog, horse, sheet, cow, rodent, and human.
41. The method of claim 40, wherein said mammal is a human.
42. A method for treating cancer in an individual, comprising exposing tumour cells included in said cancer to a compound of any one of claims 1 to 29.
43. The method of claim 42, wherein said compound, or a prodrug thereof, is administered to said individual.
44. The method of claim 42 or 43, wherein said individual is a mammal is selected from: domestic mammal, cat, dog, horse, sheet, cow, rodent, and human.
45. The method of claim 44, wherein said mammal is a human.
46. A method for inhibiting cell proliferation, comprising contacting a cell with a compound of any one of claims 1 to 29.
47. Use of a compound of any one of claims 1 to 29, or a prodrug of claim 30, for the preparation of a medicament for the treatment of cancer.
48. A pharmaceutical composition comprising a compound of any one of claims 1 to 29, or a prodrug according to claim 30, and a pharmaceutically acceptable carrier, diluent or excipient, for the treatment of cancer.
49. A method of synthesizing a compound of any one of claims 1 to 29, comprising the step of reacting a compound having a structure represented by formula (II) with a compound having a structure represented by formula (III).
Figure imgf000182_0001
(H) (HI) wherein R17 is independently selected from -Ci-6-alkyl, -CH2-aryl, or — aryl, with R3 being as defined in claim 1.
50. A method of synthesizing a compound of any one of claims 1 to 29, comprising the step of reacting a compound having a structure represented by formula (II) with a compound having a structure represented by formula (IV).
Figure imgf000183_0001
(IV) with R4 and R5 being as defined in claim 1.
51. A compound having a structure represented by formula (V)
Figure imgf000183_0002
(V) or any tautomeric or stereoisomer^ form thereof, wherein R7 is selected from -S(O)1n-R17, with m = 0, 1 or 2, and -N(R^-V-R2; R18 is taken from the list of -W-R4, -COOH, -COOR17, and -Br; and R17 is independently selected from -Ci-6- alkyl, -CH2-aryl, or — aryl; with R1, R2, R3, R4, R5, W and X being as defined in claim 1 , provided that if R7 is -N(R1)- V-R2, then R18 is not -W-R4.
52. A compound of claim 51 , wherein X is =O.
PCT/US2007/013299 2006-10-16 2007-06-06 8-oxy-2-aminopyrido (2, 3-d) pyrimidin-7-one derivatives as kinase inhibitors and anticancer agents WO2008150260A1 (en)

Priority Applications (14)

Application Number Priority Date Filing Date Title
PCT/US2007/013299 WO2008150260A1 (en) 2007-06-06 2007-06-06 8-oxy-2-aminopyrido (2, 3-d) pyrimidin-7-one derivatives as kinase inhibitors and anticancer agents
BRPI0715566-2A BRPI0715566A2 (en) 2006-10-16 2007-10-16 compound, prodrug, pharmaceutical composition, use of a compound, method for inhibiting cell proliferation and method for synthesizing a compound
JP2009532937A JP2010506902A (en) 2006-10-16 2007-10-16 Pyrido [2,3-D] pyrimidines and their use as kinase inhibitors
AU2007311480A AU2007311480A1 (en) 2006-10-16 2007-10-16 Pyrido [2, 3-d] pyrimidines and their use as kinase inhibitors
EP07826759A EP2084159A1 (en) 2006-10-16 2007-10-16 Pyrido ý2, 3-d¨pyrimidines and their use as kinase inhibitors
PCT/IB2007/054209 WO2008047307A1 (en) 2006-10-16 2007-10-16 Pyrido [2, 3-d] pyrimidines and their use as kinase inhibitors
CA002665384A CA2665384A1 (en) 2006-10-16 2007-10-16 Pyrido [2, 3-d] pyrimidines and their use as kinase inhibitors
US13/949,728 US20140163223A1 (en) 2006-10-16 2013-07-24 Pyrido-[2,3-D] Pyrimidines and Their Use as Kinase Inhibitors
US13/949,666 US20140163005A1 (en) 2006-10-16 2013-07-24 Pyrido-[2,3-D] Pyrimidines and Their Use as Kinase Inhibitors
US13/949,508 US20140163004A1 (en) 2006-10-16 2013-07-24 Pyrido-[2,3-D] Pyrimidines and Their Use as Kinase Inhibitors
US13/949,743 US20140163007A1 (en) 2006-10-16 2013-07-24 Pyrido-[2,3-D] Pyrimidines and Their Use as Kinase Inhibitors
US13/949,495 US20140163003A1 (en) 2006-10-16 2013-07-24 Pyrido-[2,3-D] Pyrimidines and Their Use as Kinase Inhibitors
US13/949,690 US20140163006A1 (en) 2006-10-16 2013-07-24 Pyrido-[2,3-D] Pyrimidines and Their Use as Kinase Inhibitors
US13/949,712 US20140163222A1 (en) 2006-10-16 2013-07-24 Pyrido-[2,3-D] Pyrimidines and Their Use as Kinase Inhibitors

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US2007/013299 WO2008150260A1 (en) 2007-06-06 2007-06-06 8-oxy-2-aminopyrido (2, 3-d) pyrimidin-7-one derivatives as kinase inhibitors and anticancer agents

Publications (1)

Publication Number Publication Date
WO2008150260A1 true WO2008150260A1 (en) 2008-12-11

Family

ID=38668983

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/013299 WO2008150260A1 (en) 2006-10-16 2007-06-06 8-oxy-2-aminopyrido (2, 3-d) pyrimidin-7-one derivatives as kinase inhibitors and anticancer agents

Country Status (1)

Country Link
WO (1) WO2008150260A1 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2580217A2 (en) * 2010-06-10 2013-04-17 Afraxis, Inc. 6-(ethynyl)pyrido[2,3-d]pyrimidin-7(8h)-ones for the treatment of cns disorders
WO2014182829A1 (en) * 2013-05-09 2014-11-13 Principia Biopharma Inc. Quinolone derivatives as fibroblast growth factor inhibitors
US8889696B2 (en) 2009-12-18 2014-11-18 Temple University—Of the Commonwealth System of Higher Education Substituted pyrido[2,3-d]pyrimidin-7(8H)-ones and therapeutic uses thereof
US8987267B2 (en) 2010-08-05 2015-03-24 Temple University—Of the Commonwealth System of Higher Education 2-substituted-8-alkyl-7-OXO-7,8-dihydropyrido[2,3-D]pyrimidine-6-carbonitriles and uses thereof in treating proliferative disorders
US9321786B2 (en) 2013-03-15 2016-04-26 Celgene Avilomics Research, Inc. Heteroaryl compounds and uses thereof
US9663524B2 (en) 2013-03-15 2017-05-30 Celgene Car Llc Substituted pyrido[2,3-d]pyrimidines as protein kinase inhibitors
US10065966B2 (en) 2013-03-15 2018-09-04 Celgene Car Llc Substituted pyrido[2,3-d]pyrimidines as inhibitors of protein kinases
US10538517B2 (en) 2015-05-22 2020-01-21 Principia Biopharma, Inc. Quinolone derivatives as FGFR inhibitors
US10899760B2 (en) 2015-08-11 2021-01-26 Principia Biopharma, Inc. Processes for preparing an FGFR inhibitor
US11078199B2 (en) 2014-02-07 2021-08-03 Principia Biopharma, Inc. Quinolone derivatives as fibroblast growth factor receptor inhibitors

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998033798A2 (en) * 1997-02-05 1998-08-06 Warner Lambert Company Pyrido[2,3-d]pyrimidines and 4-amino-pyrimidines as inhibitors of cell proliferation
WO2002064594A2 (en) * 2001-02-12 2002-08-22 F. Hoffmann-La Roche Ag 6-substituted pyrido-pyrimidines
WO2003062236A1 (en) * 2002-01-22 2003-07-31 Warner-Lambert Company Llc 2-(PYRIDIN-2-YLAMINO)-PYRIDO[2,3d]PYRIMIDIN-7-ONES

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998033798A2 (en) * 1997-02-05 1998-08-06 Warner Lambert Company Pyrido[2,3-d]pyrimidines and 4-amino-pyrimidines as inhibitors of cell proliferation
WO2002064594A2 (en) * 2001-02-12 2002-08-22 F. Hoffmann-La Roche Ag 6-substituted pyrido-pyrimidines
WO2003062236A1 (en) * 2002-01-22 2003-07-31 Warner-Lambert Company Llc 2-(PYRIDIN-2-YLAMINO)-PYRIDO[2,3d]PYRIMIDIN-7-ONES

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8889696B2 (en) 2009-12-18 2014-11-18 Temple University—Of the Commonwealth System of Higher Education Substituted pyrido[2,3-d]pyrimidin-7(8H)-ones and therapeutic uses thereof
EP2580217A4 (en) * 2010-06-10 2014-06-18 Afraxis Holdings Inc 6-(ethynyl)pyrido[2,3-d]pyrimidin-7(8h)-ones for the treatment of cns disorders
EP2580217A2 (en) * 2010-06-10 2013-04-17 Afraxis, Inc. 6-(ethynyl)pyrido[2,3-d]pyrimidin-7(8h)-ones for the treatment of cns disorders
US8987267B2 (en) 2010-08-05 2015-03-24 Temple University—Of the Commonwealth System of Higher Education 2-substituted-8-alkyl-7-OXO-7,8-dihydropyrido[2,3-D]pyrimidine-6-carbonitriles and uses thereof in treating proliferative disorders
US9695132B2 (en) 2013-03-15 2017-07-04 Celgene Car Llc Heteroaryl compounds and uses thereof
US9321786B2 (en) 2013-03-15 2016-04-26 Celgene Avilomics Research, Inc. Heteroaryl compounds and uses thereof
US9663524B2 (en) 2013-03-15 2017-05-30 Celgene Car Llc Substituted pyrido[2,3-d]pyrimidines as protein kinase inhibitors
US10065966B2 (en) 2013-03-15 2018-09-04 Celgene Car Llc Substituted pyrido[2,3-d]pyrimidines as inhibitors of protein kinases
US10189794B2 (en) 2013-03-15 2019-01-29 Celgene Car Llc Heteroaryl compounds and uses thereof
US10618902B2 (en) 2013-03-15 2020-04-14 Celgene Car Llc Substituted pyrido[2,3-d]pyrimidines as inhibitors of protein kinases
US10774052B2 (en) 2013-03-15 2020-09-15 Celgene Car Llc Heteroaryl compounds and uses thereof
US9630963B2 (en) 2013-05-09 2017-04-25 Principia Biopharma, Inc. Quinolone derivatives as fibroblast growth factor inhibitors
WO2014182829A1 (en) * 2013-05-09 2014-11-13 Principia Biopharma Inc. Quinolone derivatives as fibroblast growth factor inhibitors
US11078199B2 (en) 2014-02-07 2021-08-03 Principia Biopharma, Inc. Quinolone derivatives as fibroblast growth factor receptor inhibitors
US10538517B2 (en) 2015-05-22 2020-01-21 Principia Biopharma, Inc. Quinolone derivatives as FGFR inhibitors
US10899760B2 (en) 2015-08-11 2021-01-26 Principia Biopharma, Inc. Processes for preparing an FGFR inhibitor

Similar Documents

Publication Publication Date Title
US8518958B2 (en) Pyrido [2,3-D] pyrimidines and their use as kinase inhibitors
EP2084159A1 (en) Pyrido ý2, 3-d¨pyrimidines and their use as kinase inhibitors
WO2008150260A1 (en) 8-oxy-2-aminopyrido (2, 3-d) pyrimidin-7-one derivatives as kinase inhibitors and anticancer agents
ES2715611T3 (en) Novel compounds of 3H-imidazo [4,5-b] pyridine-3,5-disubstituted and 3H- [1,2,3] triazolo [4,5-b] pyridine 3,5-disubstituted as protein kinase modulators
US10689378B2 (en) Triazolopyridine compounds and uses thereof
JP2019504901A (en) Positive allosteric modulator of muscarinic acetylcholine receptor M1
TW200948813A (en) Plk inhibitors
EP3697781B1 (en) Antagonists of the muscarinic acetylcholine receptor m4
CN105793254B (en) Azaquinazoline carboxamide derivatives
EP2112150B1 (en) Improved raf inhibitors
AU2018300980A1 (en) Antagonists of the muscarinic acetylcholine receptor M4
US10577362B2 (en) Substituted 2, 4-diamino-quinoline derivatives for use in the treatment of proliferative diseases
US8232283B2 (en) Raf inhibitors
US20130035349A1 (en) Pyrazolopyrimidinone kinase inhibitor
WO2023288305A1 (en) Small molecule cyclin dependent kinase 4/6 (cdk4/6) and ikzf2 (helios) degraders and methods of use thereof
US20140038991A1 (en) Protein Kinase Inhibitors
EP2096107A1 (en) Derivatives of squaric acid with anti-proliferative activity
WO2023239846A1 (en) Heterocyclic compounds as pi3kα inhibitors
EP2325185A1 (en) Plk inhibitor

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07795794

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 23.03.2010)

122 Ep: pct application non-entry in european phase

Ref document number: 07795794

Country of ref document: EP

Kind code of ref document: A1