WO2012065019A2 - Pyridopyrimidinone inhibitors of p13k alpha - Google Patents

Pyridopyrimidinone inhibitors of p13k alpha Download PDF

Info

Publication number
WO2012065019A2
WO2012065019A2 PCT/US2011/060293 US2011060293W WO2012065019A2 WO 2012065019 A2 WO2012065019 A2 WO 2012065019A2 US 2011060293 W US2011060293 W US 2011060293W WO 2012065019 A2 WO2012065019 A2 WO 2012065019A2
Authority
WO
WIPO (PCT)
Prior art keywords
pyrimidin
ethyl
methyl
optionally substituted
compound
Prior art date
Application number
PCT/US2011/060293
Other languages
French (fr)
Other versions
WO2012065019A3 (en
Inventor
Dana T. Aftab
Arthur Decillis
Original Assignee
Exelixis, 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
Application filed by Exelixis, Inc. filed Critical Exelixis, Inc.
Publication of WO2012065019A2 publication Critical patent/WO2012065019A2/en
Publication of WO2012065019A3 publication Critical patent/WO2012065019A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia

Definitions

  • This invention relates to the field of protein kinases and inhibitors thereof.
  • the invention relates to inhibitors of phosphatidylinositol 3-kinase (PI3K) signaling pathways, and methods of their use.
  • PI3K phosphatidylinositol 3-kinase
  • the protein kinases are a large and diverse family of enzymes that catalyze protein phosphorylation and play a critical role in cellular signaling. Protein kinases may exert positive or negative regulatory effects, depending upon their target protein. Protein kinases are involved in specific signaling pathways which regulate cell functions such as, but not limited to, metabolism, cell cycle progression, cell adhesion, vascular function, apoptosis, and angiogenesis. Malfunctions of cellular signaling have been associated with many diseases, the most characterized of which include cancer and diabetes. The regulation of signal transduction by cytokines and the association of signal molecules with protooncogenes and tumor suppressor genes have been well documented.
  • Phosphatidylinositol 3-kinase a dual specificity protein kinase, is composed of an 85 kDa regulatory subunit and a 1 10 kDa catalytic subunit.
  • the protein encoded by this gene represents the catalytic subunit, which uses ATP to phosphorylate Ptdlns, PtdIns4P and PtdIns(4,5)P2.
  • PTEN a tumor suppressor which inhibits cell growth through multiple mechanisms, can dephosphorylate PIP3, the major product of PIK3CA.
  • PIP3 in turn, is required for translocation of protein kinase B (AKT1, PKB) to the cell membrane, where it is phosphorylated and activated by upstream kinases.
  • AKT1 protein kinase B
  • PKB protein kinase B
  • PI3Ko has been implicated in the control of cytoskeletal reorganization, apoptosis, vesicular trafficking, proliferation and differentiation processes.
  • Increased copy number and expression of PIK3CA is associated with a number of malignancies such as ovarian cancer (Campbell et al., Cancer Res 2004, 64, 7678-7681 ; Levine et al., Clin Cancer Res 2005, 1 1, 2875-2878; Wang et al., Hum Mutat 2005, 25, 322; Lee et al., Gynecol Oncol 2005, 97, 26-34), cervical cancer, breast cancer (Bachman, et al.
  • the invention provides compounds that inhibit, regulate, and/or modulate PI3K that are useful in the treatment of hyperproliferative diseases, such as cancer, in humans.
  • This invention also provides methods of making the compound, methods of using such compounds in the treatment of hyperproliferative diseases in humans and to pharmaceutical compositions containing such compounds.
  • a first aspect of the invention provides a compound of Formula I:
  • R 1 is hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted arylalkyl, optionally substituted heterocycloalkyl, optionally substituted
  • heterocycloalkylalkyl optionally substituted heteroaryl or optionally substituted heteroarylalkyl;
  • R 2 is hydrogen or alkyl where the alkyl is optionally substituted with 1, 2, 3, 4, or 5 R 8 groups;
  • X is -NR 3 -;
  • R 4 is optionally substituted alkyl
  • R 5 is hydrogen
  • R 6 is phenyl, acyl, or heteroaryl wherein the phenyl and heteroaryl are optionally substituted with 1, 2, 3, 4, or 5 R 9 groups;
  • each R 8 when present, is independently hydroxy, halo, alkoxy, haloalkoxy, amino,
  • alkylamino dialkylaminoalkyl, or alkoxyalkylamino
  • each R 9 when present, is independently halo, alkyl, haloalkyl, alkoxy, haloalkoxy, cyano, amino, alkylamino, dialkylamino, alkoxyalkyl, carboxyalkyl, alkoxycarbonyl, aminoalkyl, cycloalkyl, aryl, arylalkyl, aryloxy, heterocycloalkyl, or heteroaryl and where the cycloalkyl, aryl, heterocycloalkyl, and heteroaryl, each either alone or as part of another group within R 9 , are independently optionally substituted with 1, 2, 3, or 4 groups selected from halo, alkyl, haloalkyl, hydroxy, alkoxy, haloalkxy, amino, alkylamino, and dialkylamino.
  • a second aspect of the invention provides a compound of Formula II:
  • R 1 is hydrogen, optionally substituted alkyl, optionally substituted C 3 -C 7 cycloalkyl,
  • optionally substituted aryl optionally substituted arylalkyl, optionally substituted heterocycloalkyl, optionally substituted heterocycloalkylalkyl, optionally substituted heteroaryl or optionally substituted heteroarylalkyl;
  • X is S, S0 2 , or -NR 3 -;
  • R is hydrogen, haloalkyl, optionally substituted alkyl, optionally substituted C3-O7
  • R 2 is optionally further substituted with one or more R 8 groups;
  • R 3 , R 3a , and R 3b are independently hydrogen, optionally substituted alkyl, optionally
  • substituted C3-C7 cycloalkyl optionally substituted aryl, optionally substituted heterocycloalkyl or optionally substituted heteroaryl;
  • R 4 is hydrogen, halo, haloalkyl, haloalkoxy, -NR 3a -, optionally substituted alkyl, optionally substituted C1-C6 alkoxy, optionally substituted C
  • R 5 is hydrogen, halo, haloalkyl, haloalkoxy, optionally substituted Q-C6 alkyl, optionally substituted Ci-C 6 alkoxy, optionally substituted Ci-C 6 alkoxyalkyl, optionally substituted aminoaikyl, optionally substituted C3-C7 cycloalkyl, optionally substituted aryl, optionally substituted aryl Ci-C 6 alkyl or optionally substituted heteroaryl; and
  • R 6 is hydrogen, halo, haloalkyl, haloalkoxy, -NR 3b -, optionally substituted Ci-C 6 alkyl, optionally substituted Q-C6 alkoxy, optionally substituted C
  • R 6 groups are optionally further substituted with 1 , 2, 3, 4, or 5 R 9 groups; each R 8 , when present, is independently hydroxy, halo, haloalkyl, haloalkoxy, optionally substituted alkyl, optionally substituted Ci-C 6 alkoxy, optionally substituted C
  • alkylcarboxyheterocycloalkyl oxy Ci-C 6 alkylheterocycloalkyl, optionally substituted aminoalkyl, optionally substituted C3-C7 cycloalkyl, optionally substituted aryl, optionally substituted aryl Ci-C 6 alkyl, optionally substituted heterocycloalkyl, optionally substituted heterocycloalkylalkyl, optionally substituted heteroaryl or optionally substituted heteroarylalkyl;
  • each R 9 when present, is independently halo, haloalkyl, haloalkoxy, optionally substituted C
  • the invention is directed to a pharmaceutical composition which comprises a compound of Formula I or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier, excipient, or diluent.
  • the invention comprises a method of inhibiting PI3K, comprising contacting a cell with a compound of Formula I or II or a pharmaceutically acceptable salt or solvate thereof, or with a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula I or II and a pharmaceutically acceptable carrier, excipient, or diluent.
  • a fifth aspect of the invention is a method of inhibiting the in vivo activity of PDKa, the method comprising administering to a patient an effective PBKcc-inhibiting- inhibiting amount of a compound of Formula I or ⁇ , or a pharmaceutically acceptable salt, solvate, or a pharmaceutical composition thereof.
  • the Invention provides a method for treating a disease, disorder, or syndrome which method comprises administering to a patient a therapeutically effective amount of a compound of Formula I or II or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula I or II and a pharmaceutically acceptable carrier, excipient, or diluent.
  • a seventh aspect of the invention is directed to a process of preparing a compound of Formula I, comprising:
  • R 6 is phenyl or heteroaryl each optionally substituted with I, 2, 3, 4, or 5 R 9 groups (as defined in the Summary of the Invention) and R 1 and R 4 are as defined in the
  • R 1 and R 4 are as defined in the Summary of the Invention; with tributyl-1- ethylvinyltin or with an intermediate of formula R 6 B(OH) 2 where R 6 is phenyl or heteroaryl each optionally substituted with 1, 2, 3, 4, or 5 R 9 groups (as defined in the Summary of the Invention) to yield, respectively, a Compound of Formula 1(a) or 1(b):
  • R 1 and R 4 are as defined in the Summary of the Invention; with an intermediate of R 2 H 2 (where R 2 is as defined in the Summary of the Invention) to yield a Compound of Formula 1(a);
  • Compound A is the Compound of Formula I in each of the above aspects.
  • An additional aspect relates to a method for treating a disease, disorder, or syndrome which method comprises administering to a patient a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula I or II or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula I, la, or II and a pharmaceutically acceptable carrier, excipient, or diluent, in combination with one or more agent.
  • Compound A is the Compound of Formula I in this aspect.
  • the additional agent or agents are listed below:
  • FK228 (Depsipeptide; Romidepsin) Celgene Corporation
  • MDX-010 MDX-CTLA4; Hybridoma- Medarex Inc
  • GM-CSF (sargramostim, Leukine) Genzyme
  • the additional agent or agents are selected from the following agents:
  • the additional agent or agents are selected from ABT-888, AZD6244, CCI-779, erlotinib, gefitinib, GW572016, GW786034, pertuzumab, or sunitinib.
  • An additional aspect relates to a method for treating a disease selected from anaplastic thyroid cancer, chronic lymphocytic leukemia, mantel cell lymphoma, non- Hodgkins lymphoma, glioblastoma multiforme, pediatric brain tumors, pontine glioma, HPV- related head and neck cancer, Hormone Receptive positve (HR+) breast cancer, triple negative breast cancer, and HER-2 overexpressing breast cancer, comprising adminstering a therapeutically effective amount of a compound of Formula I or II or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition comprising a
  • Compound A is the Compound of Formula I in this aspect.
  • An additional aspect relates to a method for treating a disease selected from anaplastic thyroid cancer, chronic lymphocytic leukemia, mantel cell lymphoma, non- Hodgkins lymphoma, glioblastoma multiforme, pediatric brain tumors, pontine glioma, HPV- related head and neck cancer, Hormone Receptive positve (HR+) breast cancer, triple negative breast cancer, and HER-2 overexpressing breast cancer, comprising adminstering a therapeutically effective amount of a compound of Formula I or ⁇ or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition comprising a
  • Compound A is the Compound of Formula I in this aspect.
  • Compound A is the Compound of Formula I and the additional agent or agents is selected from the group consisting of ABT-888, AZD6244, CCI-779, erlotinib, gefitinib, GW572016, GW786034, pertuzumab, and sunitinib.
  • An additional aspect relates to a capsule formulation of a compound of formula I.
  • Compound A is the Compound of Formula I in this aspect.
  • the capsule formulation is one of the following formulations or an equivalent thereof.
  • the capsule formulation is a 90 mg "powder-in-capsule" (PiC) formulation qd or a 50 mg capsule
  • the symbol "-" means a single bond
  • " " means a single or double bond.
  • the symbol “ ⁇ ” refers to a group on a double-bond as occupying either position on the terminus of a double bond to which the symbol is attached; that is, the geometry, E- or Z-, of the double bond is ambiguous. When a group is depicted removed from its parent formula, the symbol will be used at the end of the bond which was theoretically cleaved in order to separate the group from its parent structural formula.
  • a group "R” is depicted as "floating" on a ring system, as for example
  • a substituent "R” may reside on any atom of the ring system, assuming replacement of a depicted, implied, or expressly defined hydrogen from one of the ring atoms, so long as a stable structure is formed.
  • the "R” group may reside on either the 5-membered or the 6-membered ring of the fused ring system.
  • the two "R's" may reside on any two atoms of the ring system, again assuming each replaces a depicted, implied, or expressly defined hydrogen on the ring.
  • Acyl means a -C(0)R radical where R is optionally substituted alkyl, optionally substituted alkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, heterocycloalkyl, or heterocycloalkylalkyl, as defined herein, e.g., acetyl,
  • Acylamino means a -NRR' radical where R is hydrogen, hydroxy, alkyl, or alkoxy and R' is acyl, as defined herein.
  • Acyloxy means an -OR radical where R is acyl, as defined herein, e.g.
  • administering and variants thereof (e.g., “administering” a compound) in reference to a compound of the invention means introducing the compound or a prodrug of the compound into the system of the animal in need of treatment.
  • a compound of the invention or prodrug thereof is provided in combination with one or more other active agents (e.g., surgery, radiation, and chemotherapy, etc.)
  • “administration” and its variants are each understood to include concurrent and sequential introduction of the compound or prodrug thereof and other agents.
  • Alkenyl means a means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to 6 carbon atoms which radical contains at least one double bond, e.g., ethenyl, propenyl, l-but-3-enyl, and l-pent-3-enyl, and the like.
  • Alkoxy means an -OR group where R is alkyl group as defined herein.
  • Examples include methoxy, ethoxy, propoxy, isopropoxy, and the like.
  • Alkoxyalkyl means an alkyl group, as defined herein, substituted with at least one, preferably one, two, or three, alkoxy groups as defined herein. Representative examples include methoxymethyl and the like.
  • Alkoxyalkylamino means an -NRR' group where R is hydrogen, alkyl, or alkoxyalkyl and R' is alkoxyalkyl, as defined herein.
  • Alkoxyalkylaminoalkyl means an alkyl group substituted with at least one, specifically one or two, alkoxyalkylamino group(s), as defined herein.
  • Alkoxycarbonyl means a -C(0)R group where R is alkoxy, as defined herein.
  • Alkyl means a linear saturated monovalent hydrocarbon radical of one to six carbon atoms or a branched saturated monovalent hydrocarbon radical of three to 6 carbon atoms, e.g., methyl, ethyl, propyl, 2-propyl, butyl (including all isomeric forms), or pentyl (including all isomeric forms), and the like.
  • Alkylamino means an -NHR group where R is alkyl, as defined herein.
  • Alkylaminoalkyl means an alkyl group substituted with one or two alkylamino groups, as defined herein.
  • Alkylaminoalkyloxy means an -OR group where R is alkylaminoalkyl, as defined herein.
  • Alkylcarbonyl means a -C(0)R group where R is alkyl, as defined herein.
  • Alkynyl means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to 6 carbon atoms which radical contains at least one triple bond, e.g., ethynyl, propynyl, butynyl, pentyN-2-yl and the like.
  • Amino means -NH 2 .
  • aminoalkyl means an alkyl group substituted with at least one, specifically one, two or three, amino groups.
  • aminoalkyloxy means an -OR group where R is aminoalkyl, as defined herein.
  • Aryl means a monovalent six- to fourteen-membered, mono- or bi-carbocyclic ring, wherein the monocyclic ring is aromatic and at least one of the rings in the bicyclic ring is aromatic. Unless stated otherwise, the valency of the group may be located on any atom of any ring within the radical, valency rules permitting. Representative examples include phenyl, naphthyl, and indanyl, and the like.
  • Arylalkyl means an alkyl radical, as defined herein, substituted with one or two aryl groups, as defined herein, e.g., benzyl and phenethyl, and the like.
  • Aryloxy means an -OR group where R is aryl, as defined herein.
  • Carboxyalkyl means an alkyl group, as defined herein, substituted with at least one, specifically one or two, -C(0)OH group(s).
  • Cycloalkyl means a monocyclic or fused bicyclic, saturated or partially unsaturated (but not aromatic), monovalent hydrocarbon radical of three to ten carbon ring atoms.
  • Fused bicyclic hydrocarbon radical includes bridged ring systems.
  • the valency of the group may be located on any atom of any ring within the radical, valency rules permitting.
  • cycloalkyl includes, but is not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexyl, or cyclohex-3-enyl, and the like.
  • Cycloalkylalkyl means an alkyl group substituted with at least one, specifically one or two, cycloalkyl group(s) as defined herein.
  • Dialkylamino means a -NRR' radical where R and R' are alkyl as defined herein, or an N-oxide derivative, or a protected derivative thereof, e.g., dimethylamino, diethylamino, N,N-methylpropylamino or N,N-methylethylamino, and the like.
  • Dialkylaminoalkyl means an alkyl group substituted with one or two dialkylamino groups, as defined herein.
  • Dialkylaminoalkyloxy means an -OR group where R is dialkylaminoalkyl, as defined herein. Representative examples include 2-(N,N-diethylamino)-ethyloxy, and the like.
  • fused-polycyclic or "fused ring system” means a polycyclic ring system that contains bridged or fused rings; that is, where two rings have more than one shared atom in their ring structures.
  • fused-polycyclics and fused ring systems are not necessarily all aromatic ring systems.
  • fused-polycyclics share a vicinal set of atoms, for example naphthalene or 1,2,3,4-tetrahydro-naphthalene.
  • a spiro ring system is not a fused-polycyclic by this definition, but fused polycyclic ring systems of the invention may themselves have spiro rings attached thereto via a single ring atom of the fused-polycyclic.
  • two adjacent groups on an aromatic system may be fused together to form a ring structure.
  • the fused ring structure may contain heteroatoms and may be optionally substituted with one or more groups. It should additionally be noted that saturated carbons of such fused groups ⁇ i.e. saturated ring structures) can contain two substitution groups.
  • Halogen or "halo” refers to fluorine, chlorine, bromine or iodine.
  • Haloalkoxy means an -OR' group where R' is haloalkyl as defined herein, e.g., trifluoromethoxy or 2,2,2-trifluoroethoxy, and the like.
  • Haloalkyl mean an alkyl group substituted with one or more halogens, specifically one to five halo atoms, e.g., trifluoromethyl, 2-chloroethyl, and 2,2-difluoroethyl, and the like.
  • Heteroaryl means a monocyclic, fused bicyclic, or fused tricyclic, monovalent radical of 5 to 14 ring atoms containing one or more, specifically one, two, three, or four ring heteroatoms independently selected from -0-, -S(0)yy. (n is 0, 1, or 2), -N-, -N(R X )-, and the remaining ring atoms being carbon, wherein the ring comprising a monocyclic radical is aromatic and wherein at least one of the fused rings comprising a bicyclic or tricyclic radical is aromatic.
  • R x is hydrogen, alkyl, hydroxy, alkoxy, acyl, or alkylsulfonyl.
  • Fused bicyclic radical includes bridged ring systems. Unless stated otherwise, the valency may be located on any atom of any ring of the heteroaryl group, valency rules permitting. When the point of valency is located on the nitrogen, R x is absent.
  • heteroaryl includes, but is not limited to, 1,2,4-triazolyl, 1,3,5-triazolyl, phthalimidyl, pyridinyl, pyrrolyl, imidazolyl, thienyl, furanyl, indolyl, 2,3-dihydro-lH-indolyl (including, for example, 2,3-dihydro- lH-indol-2-yl or 2,3-dihydro-lH-indol-5-yl, and the like), isoindolyl, indolinyl, isoindolinyl, benzimidazolyl, benzodioxol-4-yl, benzofuranyl, cinnolinyl, indolizinyl, naphthyridiV-3-yl, phthalazi/V-3-yl, phthaIazi/V-4-yl, pteridinyl, purin
  • pyrrolo[3,2- c]pyridinyl including, for example, pyrrolo[3,2-c]pyridiN-2-yl or pyrrolo[3,2-c]pyridiN-7-yl, and the like
  • benzopyranyl thiazolyl, isothiazolyl, thiadiazolyl, benzothiazolyl,
  • benzothienyl and the derivatives thereof, or N-oxide or a protected derivative thereof.
  • Heteroarylalkyl means an alkyl group, as defined herein, substituted with at least one, specifically one or two heteroaryl group(s), as defined herein.
  • Heteroatom refers to O, S, N, or P.
  • Heterocycloalkyl means a saturated or partially unsaturated (but not aromatic) monovalent monocyclic group of 3 to 8 ring atoms or a saturated or partially unsaturated (but not aromatic) monovalent fused bicyclic group of 5 to 12 ring atoms in which one or more, specifically one, two, three, or four ring heteroatoms independently selected from O, S(0) Struktur (n is 0, 1, or 2), N, N(R y ) (where R y is hydrogen, alkyl, hydroxy, alkoxy, acyl, or alkylsulfonyl), the remaining ring atoms being carbon.
  • Fused bicyclic radical includes bridged ring systems. Unless otherwise stated, the valency of the group may be located on any atom of any ring within the radical, valency rules permitting. When the point of valency is located on a nitrogen atom, R y is absent.
  • heterocycloalkyl includes, but is not limited to, azetidinyl, pyrrolidinyl, 2-oxopyrrolidinyl, 2,5-dihydro-lH-pyrrolyl, piperidinyl, 4-piperidonyl, morpholinyl, piperazinyl, 2-oxopiperazinyl, tetrahydropyranyl,
  • Heterocycloalkylalkyl means an alkyl radical, as defined herein, substituted with one or two heterocycloalkyl groups, as defined herein, e.g., morpholinylmethyl, N-pyrrolidinylethyl, and 3-(N-azetidinyl)propyl, and the like.
  • Heterocycloalkylalkyloxy means an -OR group where R is heterocycloalkylalkyl, as defined herein.
  • saturated bridged ring system refers to a bicyclic or polycyclic ring system that is not aromatic. Such a system may contain isolated or conjugated unsaturation, but not aromatic or heteroaromatic rings in its core structure (but may have aromatic substitution thereon). For example, hexahydro-furo[3,2-b]furan, 2,3,3a,4,7,7a-hexahydro-lH-indene, 7-aza-bicyclo[2.2.1]heptane, and l,2,3,4,4a,5,8,8a-octahydro-naphthalene are all included in the class "saturated bridged ring system.
  • Spirocyclyl or "spirocyclic ring” refers to a ring originating from a particular annular carbon of another ring.
  • a ring atom of a saturated bridged ring system (rings B and B'), but not a bridgehead atom, can be a shared atom between the saturated bridged ring system and a spirocyclyl (ring A) attached thereto.
  • a spirocyclyl can be carbocyclic or heteroalicyclic.
  • Optionally substituted alkoxy means an -OR group where R is optionally substituted alkyl, as defined herein.
  • Optionally substituted alkyl means an alkyl radical, as defined herein, optionally substituted with one or more group(s), specifically one, two, three, four, or five groups, independently selected from alkylcarbonyl, alkenylcarbonyl, cycloalkylcarbonyl, alkylcarbonyloxy, alkenylcarbonyloxy, amino, alkylamino, dialkylamino, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, cyano, cyanoalkylaminocarbonyl, alkoxy, alkenyloxy, hydroxy, hydroxyalkoxy, halo, carboxy, alkylcarbonylamino, alkylcarbonyloxy, alkyl-S(0)o- 2 -, alkenyl-S(0)o-2-, aminosulfonyl, alkylaminosulfonyl, dialkylaminosulfonyl, alkylsulfonyl-
  • alkoxyalkyloxy and -C(0)NR a R b (where R a and R are independently hydrogen, alkyl, optionally substituted alkenyl, hydroxy, alkoxy, alkenyloxy, or cyanoalkyl).
  • Optionally substituted alkenyl means an alkyl radical, as defined herein, optionally substituted with one or more group(s), specifically one, two, three, four, or five groups, independently selected from alkylcarbonyl, alkenylcarbonyl, cycloalkylcarbonyl, alkylcarbonyloxy, alkenylcarbonyloxy, amino, alkylamino, dialkylamino, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, cyano, cyanoalkylaminocarbonyl, alkoxy, alkenyloxy, hydroxy, hydroxyalkoxy, halo, carboxy, alkylcarbonylamino, alkylcarbonyloxy, alkyl-S(0)o- 2 - > alkenyl-S(0)o- 2 -, aminosulfonyl, alkylaminosulfonyl, dialkylaminosulfonyl, alkylsulfon
  • alkoxy alky loxy and -C(0)NR a R b (where R a and R b are independently hydrogen, alkyl, optionally substituted alkenyl, hydroxy, alkoxy, alkenyloxy, or cyanoalkyl).
  • Optionally substituted amino refers to the group -N(H)R or -N(R)R where each R is independently selected from the group: optionally substituted alkyl, optionally substituted alkoxy, optionally substituted aryl, optionally substituted heterocycloalkyl, optionally substituted heteroaryl, acyl, carboxy, alkoxycarbonyl, -S(0) 2 -(optionally substituted alkyl), -S(0) 2 -optionally substituted aryl), -S(0) 2 -(optionally substituted heterocycloalkyl), -S(0) 2 -(optionally substituted heteroaryl), and -S(0)2-(optionally substituted heteroaryl).
  • optionally substituted amino includes diethylamino, methylsulfonylamino, and furanyl-oxy-sulfonamino.
  • Optionally substituted aminoalkyl means an alkyl group, as defined herein, substituted with at least one, specifically one or two, optionally substituted amino group(s), as defined herein.
  • Optionally substituted aryl means an aryl group, as defined herein, optionally substituted with one, two, or three substituents independently selected from acyl, acylamino, acyloxy, optionally substituted alkyl, optionally substituted alkenyl, alkoxy, alkenyloxy, halo, hydroxy, alkoxycarbonyl, alkenyloxycarbonyl, amino, alkylamino, dialkylamino, nitro, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, carboxy, cyano, alkylthio, alkylsulfinyl, alkylsulfonyl, aminosulfonyl, alkylaminosulfonyl, dialkylaminosulfonyl, alkylsulfonylamino, aminoalkoxy, or aryl is pentafluorophenyl. Within the optional substituents on "aryl", the optional substituents
  • alkyl in alkoxycarbonyl are independently optionally substituted with one, two, three, four, or five halo.
  • Optionally substituted arylalkyl means an alkyl group, as defined herein, substituted with optionally substituted aryl, as defined herein.
  • Optionally substituted cycloalkyl means a cycloalkyl group, as defined herein, substituted with one, two, or three groups independently selected from acyl, acyloxy, acylamino, optionally substituted alkyl, optionally substituted alkenyl, alkoxy, alkenyloxy, alkoxycarbonyl, alkenyloxycarbonyl, alkylthio, alkylsulfinyl, alkylsulfonyl, aminosulfonyl, alkylaminosulfonyl, dialkylaminosulfonyl, alkylsulfonylamino, halo, hydroxy, amino, alkylamino, dialkylamino, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, nitro, alkoxyalkyloxy, aminoalkoxy, alkylaminoalkoxy, dialkylaminoalkoxy, carboxy, and
  • alkyl and alkenyl are independently optionally substituted with one, two, three, four, or five halo, e.g. haloalkyl, haloalkoxy, haloalkenyloxy, or haloalkylsulfonyl.
  • Optionally substituted cycloalkylalkyl means an alkyl group substituted with at least one, specifically one or two, optionally substituted cycloalkyl groups, as defined herein.
  • Optionally substituted heteroaryl means a heteroaryl group optionally substituted with one, two, or three substituents independently selected from acyl, acylamino, acyloxy, optionally substituted alkyl, optionally substituted alkenyl, alkoxy, alkenyloxy, halo, hydroxy, alkoxycarbonyl, alkenyloxycarbonyl, amino, alkylamino, dialkylamino, nitro, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, carboxy, cyano, alkylthio, alkylsulfinyl, alkylsulfonyl, aminosulfonyl, alkylaminosulfonyl, dialkylaminosulfonyl, alkylsulfonylamino, aminoalkoxy, alkylaminoalkoxy, and dialkylaminoalkoxy.
  • alkyl and alkenyl are independently optionally substituted with one, two, three, four, or five halo.
  • Optionally substituted heteroarylalkyl means an alkyl group, as defined herein, substituted with at least one, specifically one or two, optionally substituted heteroaryl group(s), as defined herein.
  • Optionally substituted heterocycloalkyl means a heterocycloalkyl group, as defined herein, optionally substituted with one, two, or three substituents independently selected from acyl, acylamino, acyloxy, optionally substituted alkyl, optionally substituted alkenyl, alkoxy, alkenyloxy, halo, hydroxy, alkoxycarbonyl, alkenyloxycarbonyl, amino, alkylamino, dialkylamino, nitro, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, carboxy, cyano, alkylthio, alkylsulfinyl, alkylsulfonyl, aminosulfonyl, alkylaminosulfonyl, dialkylaminosulfonyl, alkylsulfonylamino, aminoalkoxy, or aryl is pentafluorophenyl.
  • heterocycloalkyl Within the optional substituents on “heterocycloalkyl”, the alkyl and alkenyl, either alone or as part of another group (including, for example, the alkyl in alkoxycarbonyl), are independently optionally substituted with one, two, three, four, or five halo.
  • Optionally substituted heterocycloalkylalkyl means an alkyl group, as defined herein, substituted with at least one, specifically one or two, optionally substituted heterocycloalkyl group(s) as defined herein.
  • Yield for each of the reactions described herein is expressed as a percentage of the theoretical yield.
  • Patient for the purposes of the present invention includes humans and other animals, particularly mammals, and other organisms. Thus the methods are applicable to both human therapy and veterinary applications. In a preferred embodiment the patient is a mammal, and in a most preferred embodiment the patient is human.
  • Kinase-dependent diseases or conditions refer to pathologic conditions that depend on the activity of one or more protein kinases.
  • Kinases either directly or indirectly participate in the signal transduction pathways of a variety of cellular activities including proliferation, adhesion, migration, differentiation and invasion.
  • Diseases associated with kinase activities include tumor growth, the pathologic neovascularization that supports solid tumor growth, and associated with other diseases where excessive local vascularization is involved such as ocular diseases (diabetic retinopathy, age-related macular degeneration, and the like) and inflammation (psoriasis, rheumatoid arthritis, and the like).
  • phosphatases can also play a role in "kinase-dependent diseases or conditions" as cognates of kinases; that is, kinases
  • phosphorylate and phosphatases dephosphorylate, for example protein substrates. Therefore compounds of the invention, while modulating kinase activity as described herein, may also modulate, either directly or indirectly, phosphatase activity. This additional modulation, if present, may be synergistic (or not) to activity of compounds of the invention toward a related or otherwise interdependent kinase or kinase family.
  • the compounds of the invention are useful for treating diseases characterized in part by abnormal levels of cell proliferation (i.e. tumor growth), programmed cell death (apoptosis), cell migration and invasion and angiogenesis associated with tumor growth.
  • Compound A refers to a compound having the structure
  • “Therapeutically effective amount” is an amount of a compound of the invention, that when administered to a patient, ameliorates a symptom of the disease.
  • the amount of a compound of the invention which constitutes a “therapeutically effective amount” will vary depending on the compound, the disease state and its severity, the age of the patient to be treated, and the like. The therapeutically effective amount can be determined routinely by one of ordinary skill in the art having regard to their knowledge and to this disclosure.
  • Cardiac sarcoma (angiosarcoma, fibrosarcoma,
  • rhabdomyosarcoma liposarcoma
  • myxoma rhabdomyoma
  • rhabdomyoma fibroma
  • lipoma and teratoma Lung: bronchogenic carcinoma (squamous cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hanlartoma, mesothelioma
  • Gastrointestinal esophagus
  • stomach cancer squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma
  • stomach cancer squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma
  • Nervous system skull (osteoma, hemangioma, granuloma, xanthoma, osteitis defomians), meninges (meningioma, meningiosarcoma, gliomatosis), brain (astrocytoma, medulloblastoma, glioma, ependymoma, germinoma [pinealoma], glioblastoma multiform, oligodendroglioma, schwannoma, retinoblastoma, congenital tumors), spinal cord neurofibroma, meningioma, glioma, sarcoma); Gynecological: uterus (endometrial carcinoma), cervix (cervical carcinoma, pre-tumor cervical dysplasia), ovaries (ovarian
  • a "pharmaceutically acceptable salt” of a compound means a salt that is pharmaceutically acceptable and that possesses the desired pharmacological activity of the parent compound. It is understood that the pharmaceutically acceptable salts are non-toxic. Additional information on suitable pharmaceutically acceptable salts can be found in
  • Examples of pharmaceutically acceptable acid addition salts include those formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; as well as organic acids such as acetic acid, trifluoroacetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, 3-(4-hydroxybenzoyl)benzoic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid,
  • 2-naphthalenesulfonic acid 4-toluenesulfonic acid, camphorsulfonic acid, glucoheptonic acid, 4,4'-methylenebis-(3-hydroxy-2-ene-l-carboxylic acid), 3-phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, p-toluenesulfonic acid, and salicylic acid and the like.
  • Examples of a pharmaceutically acceptable base addition salts include those formed when an acidic proton present in the parent compound is replaced by a metal ion, such as sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Preferable salts are the ammonium, potassium, sodium, calcium, and magnesium salts.
  • Salts derived from pharmaceutically acceptable organic non-toxic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins. Examples of organic bases include isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine,
  • Exemplary organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline, and caffeine.
  • Prodrug refers to compounds that are transformed (typically rapidly) in vivo to yield the parent compound of the above formulae, for example, by hydrolysis in blood.
  • Common examples include, but are not limited to, ester and amide forms of a compound having an active form bearing a carboxylic acid moiety.
  • Examples of pharmaceutically acceptable esters of the compounds of this invention include, but are not limited to, alkyl esters (for example with between about one and about six carbons) the alkyl group is a straight or branched chain. Acceptable esters also include cycloalkyl esters and arylalkyl esters such as, but not limited to benzyl.
  • Examples of pharmaceutically acceptable amides of the compounds of this invention include, but are not limited to, primary amides, and secondary and tertiary alkyl amides (for example with between about one and about six carbons).
  • Amides and esters of the compounds of the present invention may be prepared according to conventional methods. A thorough discussion of prodrugs is provided in T. Higuchi and V. Stella, "Pro-drugs as Novel Delivery Systems," Vol 14 of the A.C.S.
  • Methodabolite refers to the break-down or end product of a compound or its salt produced by metabolism or biotransformation in the animal or human body; for example, biotransformation to a more polar molecule such as by oxidation, reduction, or hydrolysis, or to a conjugate (see Goodman and Gilman, "The Pharmacological Basis of Therapeutics” 8.sup.th Ed., Pergamon Press, Gilman et al. (eds), 1990 for a discussion of biotransformation).
  • the metabolite of a compound of the invention or its salt may be the biologically active form of the compound in the body.
  • a prodrug may be used such that the biologically active form, a metabolite, is released in vivo.
  • a biologically active metabolite is discovered serendipitously, that is, no prodrug design per se was undertaken.
  • An assay for activity of a metabolite of a compound of the present invention is known to one of skill in the art in light of the present disclosure.
  • Treating" or "treatment” of a disease, disorder, or syndrome includes (i) preventing the disease, disorder, or syndrome from occurring in a human, i.e. causing the clinical symptoms of the disease, disorder, or syndrome not to develop in an animal that may be exposed to or predisposed to the disease, disorder, or syndrome but does not yet experience or display symptoms of the disease, disorder, or syndrome; (ii) inhibiting the disease, disorder, or syndrome, i.e., arresting its development; and (iii) relieving the disease, disorder, or syndrome, i.e., causing regression of the disease, disorder, or syndrome.
  • the compounds disclosed herein also include all pharmaceutically acceptable isotopic variations, in which at least one atom is replaced by an atom having the same atomic number, but an atomic mass different from the atomic mass usually found in nature.
  • isotopes suitable for inclusion in the disclosed compounds include, without limitation, isotopes of hydrogen, such as 2 H and 3 H; isotopes of carbon, such as 13 C and l4 C; isotopes of nitrogen, such as 15 N; isotopes of oxygen, such as l7 0 and I8 0; isotopes of phosphorus, such as 31 P and 32 P; isotopes of sulfur, such as .sup. 35 S; isotopes of fluorine, such as 18 F; and isotopes of chlorine, such as 36 C1.
  • isotopes of hydrogen such as 2 H and 3 H
  • isotopes of carbon such as 13 C and l4 C
  • isotopes of nitrogen such as 15 N
  • isotopes of oxygen such as l7 0 and I8 0
  • isotopes of phosphorus such as 31 P and 32 P
  • isotopes of sulfur such as .sup. 35 S
  • isotopic variations e.g., deuterium, 2 H
  • isotopic variations of the disclosed compounds may incorporate a radioactive isotope (e.g., tritium, 3 H, or l4 C), which may be useful in drug and/or substrate tissue distribution studies.
  • a radioactive isotope e.g., tritium, 3 H, or l4 C
  • One embodiment (A) of a Compound of Formula I is where hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted arylalkyl, optionally substituted
  • R 1 is hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted arylalkyl, or optionally substituted heterocycloalkylalkyl. More specifically, R 1 is hydrogen, alkyl, alkyl substituted with one or two hydroxy, alkyl substituted with alkoxy, cycloalkyl, arylalkyl, or heterocycloalkylalkyl. Even more specifically, R 1 is hydrogen, methyl, ethyl, propyl, isopropyl, 2-hydroxypropyl, 3-hydroxypropyl, 2-ethoxyethyl, 3-methoxypropyl,
  • R 1 is ethyl, isopropyl, cyclopentyl, or cyclohexyl. Yet even more specifically, R 1 is ethyl.
  • R 2 is hydrogen or alkyl where the alkyl is optionally substituted with 1 , 2, 3, 4, or 5 R 8 groups.
  • R 2 is hydrogen or alkyl where the alkyl is optionally substituted with one, two, or three R 8 groups. More specifically, R 2 is hydrogen or alkyl where the alkyl is optionally substituted with one, two, or three R 8 groups; and each R 8 , when present, is independently selected from amino, alkylamino, dialkylamino, and halo.
  • R 2 is hydrogen, methyl, ethyl, propyl, isopropyl, ferf-butyl, 3-aminopropyl, 3-(N-methylamino)-propyl, 3-(N,N- dimethylamino)-propyl, 2-fluoroethyl, or 2,2,2-trifluoroethyl. Yet even more specifically, R 2 is hydrogen or ethyl. Yet even more preferably, R 2 is hydrogen.
  • R 2 is hydrogen
  • R 2 is alkyl optionally substituted with 1, 2, 3, 4, or 5, R 8 groups.
  • R 2 is alkyl where the alkyl is optionally substituted with one, two, or three R 8 groups; and each R 8 , when present, is independently selected from amino, alkylamino, dialkylamino, and halo.
  • R 2 is methyl, ethyl, propyl, isopropyl, tert-butyl, 3-aminopropyl, 3-(N-methylamino)-propyl, 3-(N,N-dimethylamino)- propyl, 2-fluoroethyl, or 2,2,2-trifluoroethyl.
  • R 2 is ethyl.
  • R 4 is optionally substituted alkyl. Specifically, R 4 is methyl or ethyl. More specifically, R 4 is methyl.
  • R 6 is acyl. More specifically, R 6 is alkylcarbonyl. Even more specifically, R 6 is acetyl.
  • R 6 is phenyl optionally substituted with 1 , 2, 3, 4, or 5 R groups.
  • R is phenyl optionally substituted with one or two R 9 groups; and each R 9 , when present, is independently selected from aryl, halo, alkoxy, aryloxy, and haloalkyl.
  • R 6 is phenyl optionally substituted with one or two R 9 groups; and each R 9 , when present, is independently selected from phenyl, fluoro, chloro, methoxy, phenyloxy, and trifluoromethyl.
  • R 6 is phenyl, phenyl substituted with phenyl, fluorophenyl, difluorophenyl, chlorophenyl, dichlorophenyl, phenyl substituted with chloro and fluoro, methoxyphenyl, dimethoxyphenyl, phenyloxyphenyl, or trifluoromethylphenyl.
  • R 6 is phenyl, 2-phenyl-phenyl, 3-phenyl-phenyl, 4-phenyl-phenyl, 2-fluorophenyl, 3- fluorophenyl, 4-fluorophenyl, 2,3-difluorophenyl, 2,4-difluorophenyl, 2,5-difluorophenyl, 2,6-difluorophenyl, 3,4-difluorophenyI, 3,5-difluorophenyl, 2-chlorophenyl, 3-chlorophenyl, 4-chlorophenyl, 2,3-dichlorophenyl, 2,4-dichlorophenyl, 2,5-dichlorophenyl,
  • Another embodiment (F) of a Compound of Formula I is where R 6 is phenyl substituted with 1, 2, 3, 4, or 5 R 9 groups.
  • Another embodiment (G) of a Compound of Formula I is where R 6 is heteroaryl optionally substituted with 1, 2, 3, 4, or 5 R 9 groups.
  • a more specific embodiment (Gl) of embodiment G is a Compound of Formula I where R 6 is a 6-membered heteroaryl optionally substituted with one or two R 9 . More specifically, R 6 is pyridinyl, pyrazinyl, pyrimidinyl, or pyridazinyl each of which is optionally substituted with one R 9 where R 9 , when present, is halo.
  • R 6 is pyridiN-2-yl, pyridiN-3-yl, pyridiN-4-yl, 3-fluoropyridi/v ' -4-yl, pyrazin-2-yl, pyrazin-3- yl, pyrimidin-2-yl, pyrimidin-4-yl, pyrimidin-5-yl, pyridazin-3-yl, or pyridazin-4-yl, each of which is optionally substituted with one or two R 9 .
  • embodiment G2 is a Compound of Formula I where R 6 is pyrazinyl, pyrimidinyl, or pyridazinyl each of which is optionally substituted with one R 9 where R 9 , when present, is halo.
  • R 6 is pyrazin-2-yl, pyrazin-3-yl, pyrimidin-2-yl, pyrimidin-4-yl, pyrimidin-5-yl, pyridazin-3-yl, or pyridazin-4-yl.
  • a more specific embodiment (G3) of embodiment G is a Compound of Formula I where R 6 is 5-membered heteroaryl optionally substituted with one or two R 9 .
  • R 6 is pyrazolyl, imidazolyl, thienyl, thiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, furanyl, pyrrolyl, triazolyl, or tetrazolyl, each of which is optionally substituted with one R 9 where R 9 , when present, is alkyl, arylalkyl, cyano, aryl, alkoxycarbonyl, or halo.
  • R 6 is pyrazol-l-yl, pyrazol-3-yl, pyrazol-4-yl, pyrazol-5-yl, imidazol-l-yl, imidazoI-2-yl, imidazol-4-yl, imidazol-5-yl, thien-2-yl, thien-3-yl, thiazol-2-yl, thiazol-4-yl, thiazol-5-yl, oxazol-2-yl, oxazol-4-yl, oxazol-5-yl, isoxazol-3-yl, isoxazol-4-yl, isoxazol-5-yl,
  • R 6 is pyrazol-3-yl, pyrazol-4-yl, pyrazol-5-yl, imidazol-2-yl, imidazol-4-yl, imidazol-5-yl, thien-2-yl, thien-3-yl, thiazol-2-yI, thiazol-4-yl, thiazol-5-yl, oxazol-2-yl, oxazol-4-yl, oxazol-5-yl, isoxazol-3-yl, isoxazol-4-yl, isoxazol-5-yl, l,2,3-oxadiazol-4-yl, 1 ,2,3-oxadiazol-5-yl, l,3,4-oxadiazol-2-yl, l,2,4-oxadiazol-3-yl, l ,2,4-oxadiazol-5-yl, furan-2-yl, furan-3-yl,
  • a more specific embodiment (G4) of embodiment G is a Compound of Formula I where R 6 is thienyl, pyrrolyl, furanyl, pyrazolyl, thiazolyl, isoxazolyl, imidazolyl, triazolyl, or tetrazolyl, each of which is optionally substituted with one R 9 where R 9 , when present, is methyl, benzyl, cyano, phenyl, N-rerr-butoxycarbonyl, or chloro.
  • R 6 is thien-2- yl, thien-3-yl, pyrrol-2-yl, furan-2-yl, furan-3-yl, pyrazoI-3-yl, pyrazol-4-yl, pyrazoI-5-yl, thiazol-2-yl, thiazol-5-yl, isoxazol-4-yl, imidazol-5-yl, triazol-5-yI, tetrazol-5-yl, each of which is optionally substituted with one R 9 where R 9 , when present, is methyl, benzyl, cyano, phenyl, N-rerr-butoxycarbonyl, or chloro.
  • R 6 is thien-2-yl, thien-3-yl, 5-cyano-thien-2-yl, 4-methyl-thien-2-yl, 4-methyl-thien-3-yl, 5-chloro-thien-5-yl, 5-phenyl- thien-2-yl, pyrrol-2-yl, N-iert-butoxycarbonyl-pyrrol-2-yl, N-methyl-pyrrol-2-yl, furan-2-yl, furan-3-yl, pyrazol-3-yl, pyrazol-4-yl, N-benzyl-pyrazol-4-yl, pyrazol-5-yl, thiazol-2-yl, thiazol-5-yl, isoxazol-4-yl, imidazol-5-yl, triazol-5-yl, tetrazol-5-yl,
  • a more specific embodiment (G5) of embodiment G is a Compound of Formula I where R 6 is thien-2-yl, thien-3-yl, pyrrol-2-yl, furan-2-yl, furan-3-yl, pyrazol-3-yl, pyrazol-4- yl, pyrazol-5-yl, thiazol-2-yl, thiazol-5-yl, isoxazol-4-yl, imidazol-5-yl, triazol-5-yl, or tetrazoI-5-yl, each of which is optionally substituted with one R 9 where R 9 , when present, is methyl, benzyl, cyano, phenyl, N-terf-butoxycarbonyl, or chloro.
  • a more specific embodiment (G6) of embodiment G is a Compound of Formula I where R 6 is indolyl, benzimidazolyl, benzofuranyl, benzoxazolyl, or benzoisoxazolyl each of which is optionally substituted with 1, 2, 3, 4, or 5 R 9 groups.
  • R 6 is indol-2-yl, indol-3-yl, indol-4-yl, indol-5-yl, indol-6-yl, indol-7-yl, benzimidazol-2-yl, benzimidazol-4- yl, benzimidazol-5-yl, benzimidazol-6-yl, benzimidazol-7-yl, benzofuran-2-yl, benzofuran-3- yl, benzofuran-4-yl, benzofuran-5-yl, benzofuran-6-yl, benzofuran-7-yl, benzoxazol-2-yl, benzoxazol-4-yl, benzoxazol-5-yl, benzoxazol-6-yl, benzoxazol-7-yl, benzoisoxazol-3-yI, benzoisoxazol-4-yl, benzoisoxazol-5-yl,
  • R 1 is hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkylalkyl, or optionally substituted arylalkyl
  • X is -NH-
  • R 2 is hydrogen or alkyl where the alkyl is optionally substituted with one or two R 8 groups
  • R 4 is alkyl
  • R 5 is hydrogen
  • R 6 is phenyl or heteroaryl wherein the phenyl and heteroaryl are optionally substituted with one, two, or three R groups
  • each R when present, is independently amino, alkylamino, dialkylamino, or halo
  • each R 9 when present, is independently alkyl, arylalkyl, cyano, aryl, alkoxycarbonyl, or halo.
  • R 6 is pyrazol-3-yl, pyrazol-4-yl, pyrazol-5-yl, imidazol-2-yl, imidazol-4-yl, imidazol-5-yl, thien-2-yl, thien-3-yl, thiazol-2-yl, thiazoI-4-yl, thiazol-5-yl, oxazol-2-yl, oxazol-4-yl, oxazol-5-yl, isoxazol-3-yl, isoxazol-4-yl, isoxazol-5-yl, l,2,3-oxadiazol-4-yl, l,2,3-oxadiazol-5-yl, l,3,4-oxadiazol-2-yl, 1 ,2,4-oxadiazol-3-yl, l,2,4-oxadiazol-5-yl, fur
  • Another embodiment (K) is a Compound of Formula I is where R 1 is alkyl or cycloalkyl; R 4 is methyl; and R 6 is heteroaryl optionally substituted with one or two R 9 groups. Specifically, each R 9 , when present, is independently alkyl, arylalkyl, cyano, aryl, alkoxycarbonyl, or halo.
  • R 6 is pyrazol-3-yI, pyrazol-4-yl, pyrazol-5-yl, imidazol-2-yl, imidazol-4-yl, imidazol-5-yl, thien-2-yl, thien-3-yl, thiazol-2-yl, thiazol-4-yl, thiazol-5-yl, oxazol-2-yl, oxazol-4-yl, oxazol-5-yl, isoxazol-3-yl, isoxazol-4-yl, isoxazol-5-yl,
  • a more specific embodiment (Kl) of embodiment K is a Compound of Formula I where R 2 is hydrogen.
  • a more specific embodiment (K2) of embodiment K is a Compound of Formula I where R 2 is methyl or ethyl.
  • R 1 is alkyl or cycloalkyl
  • R 4 is methyl
  • R 6 is phenyl optionally substituted with one or two R 9 groups.
  • each R 9 when present, is independently halo, alkoxy, or haloalkyl.
  • R 1 is alkyl or cycloalkyl
  • R 4 is methyl
  • R 2 is hydrogen
  • R 1 is alkyl or cycloalkyl
  • R 4 is methyl
  • R 2 is optionally substituted alkyl
  • the compound of formula I is a compound of formula IA.
  • R 1 is alkyl, cycloalkyl, cycloalkylalkyl, aryl, arylalkyl, heterocycloalkyl,
  • heterocycloalkylalkyl heteroaryl, or heteroarylalkyl
  • R 2 is hydrogen or alkyl
  • R 4 is alkyl
  • R 5 is hydrogen
  • R 6 is phenyl, acyl, or heteroaryl wherein the phenyl and heteroaryl are is optionally substituted with 1, 2, 3, 4, or 5 R 9 groups;
  • each R 9 when present, is independently halo, alkyl, haloalkyl, alkoxy, haloalkoxy, cyano, amino, alkylamino, dialkylamino, alkoxyalkyl, carboxyalkyl, alkoxycarbonyl, aminoalkyl, cycloalkyl, aryl, arylalkyl, aryloxy, heterocycloalkyl, or heteroaryl and where the cycloalkyl, aryl, heterocycloalkyl, and heteroaryl, each either alone or as part of another group within R 9 , are independently optionally substituted with 1, 2, 3, or 4 groups selected from halo, alkyl, haloalkyl, hydroxy, alkoxy, haloalkoxy, amino, alkylamino, and
  • R 1 is alkyl, cycloalkyl, heterocycloalkylalkyl, or arylalkyl;
  • X is -NH-;
  • R 2 is hydrogen or alkyl;
  • R 4 is alkyl;
  • R 5 is hydrogen;
  • R 6 is phenyl or heteroaryl wherein the phenyl and heteroaryl are is_optionally substituted with one, two, or three R groups;
  • each R 8 when present, is independently amino, alkylamino, dialkylamino, or halo; and each R 8 , when present, is independently alkyl, arylalkyl, cyano, aryl, or alkoxycarbonyl.
  • R 4 is methyl
  • R 1 is alkyl, cycloalkyl, or heterocycloalkyl.
  • R 1 is alkyl
  • R 6 is heteroaryl optionally substituted with 1 , 2, or 3 R 9 groups.
  • each R 9 when present, is independently alkyl, arylalkyl, cyano, aryl, alkoxycarbonyl, or halo.
  • R 6 is pyrazolyl, imidazolyl, thienyl, thiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, furanyl, pyrrolyl, triazolyl, or tetrazolyl; each of which is optionally substituted with 1, 2, or 3 R 9 groups.
  • R 6 is pyrazol-3-yl, pyrazol-4-yl, pyrazol-5-yl, imidazol-2- yl, imidazol-4-yl, imidazol-5-yl, thien-2-yl, thien-3-yl, thiazol-2-yl, thiazol-4-yl, thiazol-5-yl, oxazol-2-yl, oxazol-4-yl, oxazol-5-yl, isoxazol-3-yl, isoxazol-4-yl, isoxazol-5-yl, 1,2,3- oxadiazol-4-yl, 1 ,2,3-oxadiazol-5-yl, l,3,4-oxadiazol-2-yl, l ,2,4-oxadiazol-3-yl,
  • R 6 is pyrazinyl, pyrimidinyl, or pyridazinyl each of which is optionally substituted with 1, 2, or 3 R 9 groups and R 4 is methyl.
  • R 2 is hydrogen
  • R 4 is methyl
  • R 1 is optionally substituted alkyl, cycloalkyl, or heterocycloalkyl
  • R 6 is heteroaryl optionally substituted with 1, 2, or 3 R 9 groups.
  • the compound of formula III is selected from:
  • the compound of formula ⁇ is selected from:
  • the compound of formula IA is selected form:
  • Another embodiment (P) of the Invention is a method of treating disease, disorder, or syndrome where the disease is associated with uncontrolled, abnormal, and/or unwanted cellular activities effected directly or indirectly by PI3Ka which method comprises administering to a human in need thereof a therapeutically effective amount of a compound of Formula I or II or a pharmaceutically acceptable salt, solvate, or a pharmaceutical composition thereof.
  • the Compound is of Formula I.
  • Another embodiment (Q) of the invention is directed to a method of treating a disease, disorder, or syndrome which method comprises administering to a patient a therapeutically effective amount of a compound of Formula I, ⁇ , or III or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition comprising a
  • the disease is cancer. More
  • the cancer is breast cancer, colon cancer, rectal cancer, endometrial cancer, gastric carcinoma, glioblastoma, hepatocellular carcinoma, small cell lung cancer, non-small cell lung cancer, melanoma, ovarian cancer, cervical cancer, pancreatic cancer, prostate carcinoma, acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), or thyroid carcinoma.
  • the cancer is ovarian cancer, cervical cancer, breast cancer, colon cancer, rectal cancer, or glioblastoma.
  • Another embodiment (R) of the Invention is directed to a method of treating a disease, disorder, or syndrome which method comprises administering to a patient a therapeutically effective amount of a compound of Formula I, ⁇ . ⁇ or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition comprising a
  • the disease is cancer. More
  • the cancer is breast cancer, colon cancer, rectal cancer, endometrial cancer, gastric carcinoma, glioblastoma, hepatocellular carcinoma, small cell lung cancer, non-small cell lung cancer, melanoma, ovarian cancer, cervical cancer, pancreatic cancer, prostate carcinoma, acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), or thyroid carcinoma. Even more specifically, the cancer is ovarian cancer, cervical cancer, breast cancer, colon cancer, rectal cancer, or glioblastomas.
  • Another aspect of the invention is a method of inhibiting proliferative activity in a cell, the method comprising administering to a cell or a plurality of cells an effective amount of a compound of Formula I or ⁇ , or a pharmaceutically acceptable salt, solvate, or prodrug thereof, or a pharmaceutical composition thereof.
  • the Compound is of Formula I.
  • Another aspect of the invention is directed to employing the compounds of the invention in a method of screening for candidate agents that bind to, for example PBKoc.
  • the protein is bound to a support, and a compound of the invention is added to the assay.
  • the compound of the invention is bound to the support and the protein is added.
  • Classes of candidate agents among which novel binding agents may be sought include specific antibodies, non-natural binding agents identified in screens of chemical libraries, peptide analogs, etc. Of particular interest are screening assays for candidate agents that have a low toxicity for human cells.
  • assays may be used for this purpose, including labeled in vitro protein-protein binding assays, electrophoretic mobility shift assays, immunoassays for protein binding, functional assays (phosphorylation assays, etc.) and the like.
  • the determination of the binding of the candidate agent to, for example, PBKcc can be done in a number of ways.
  • the candidate agent (the compound of the invention) is labeled, for example, with a fluorescent or radioactive moiety and binding determined directly.
  • this may be done by attaching all or a portion of the PI3Ka protein to a solid support, adding a labeled agent (for example a compound of the invention in which at least one atom has been replaced by a detectable isotope), washing off excess reagent, and determining whether the amount of the label is that present on the solid support.
  • a labeled agent for example a compound of the invention in which at least one atom has been replaced by a detectable isotope
  • washing off excess reagent for example a compound of the invention in which at least one atom has been replaced by a detectable isotope
  • Various blocking and washing steps may be utilized as is known in the art.
  • label as used herein is meant to include both direct and indirect labeling with a compound that provides a detectable signal, for example, radioisotope, fluorescent tag, enzyme, antibodies, particles such as magnetic particles, chemiluminescent tag, or specific binding molecules, and the like.
  • Specific binding molecules include pairs, such as biotin and streptavidin, digoxin and antidigoxin, and the like.
  • the complementary member would normally be labeled with a molecule which provides for detection, in accordance with known procedures, as outlined above.
  • the label can directly or indirectly provide a detectable signal.
  • PBKcc protein may be labeled at tyrosine positions using 125 I, or with fluorophores.
  • more than one component may be labeled with different labels; using 125 I for the proteins, for example, and a fluorophor for the candidate agents.
  • the compounds of the invention may also be used as competitors to screen for additional drug candidates.
  • candidate bioactive agent or “drug candidate” or grammatical equivalents as used herein describe any molecule, e.g., protein, oligopeptide, small organic molecule, polysaccharide, polynucleotide, etc., to be tested for bioactivity. They may be capable of directly or indirectly altering the cellular proliferation phenotype or the expression of a cellular proliferation sequence, including both nucleic acid sequences and protein sequences. In other cases, alteration of cellular proliferation protein binding and/or activity is screened. In the case where protein binding or activity is screened, some embodiments exclude molecules already known to bind to that particular protein. Exemplary embodiments of assays described herein include candidate agents, which do not bind the target protein in its endogenous native state, termed herein as "exogenous" agents. In one example, exogenous agents further exclude antibodies to PI3Ka.
  • Candidate agents can encompass numerous chemical classes, though typically they are organic molecules having a molecular weight of more than about 100 and less than about 2,500 daltons.
  • Candidate agents comprise functional groups necessary for structural interaction with proteins, particularly hydrogen bonding and lipophilic binding, and typically include at least an amine, carbonyl, hydroxyl, ether, or carboxyl group, for example at least two of the functional chemical groups.
  • the candidate agents often comprise carbocyclic or heterocyclic structures and/or aromatic or polyaromatic structures substituted with one or more of the above functional groups.
  • Candidate agents are also found among biomolecules including peptides, saccharides, fatty acids, steroids, purines, pyrimidines, derivatives, structural analogs, or combinations thereof.
  • Candidate agents are obtained from a wide variety of sources including libraries of synthetic or natural compounds. For example, numerous means are available for random and directed synthesis of a wide variety of organic compounds and biomolecules, including expression of randomized oligonucleotides. Alternatively, libraries of natural compounds in the form of bacterial, fungal, plant and animal extracts are available or readily produced. Additionally, natural or synthetically produced libraries and compounds are readily modified through conventional chemical, physical and biochemical means. Known pharmacological agents may be subjected to directed or random chemical modifications, such as acylation, alkylation, esterification, amidification to produce structural analogs.
  • the binding of the candidate agent is determined through the use of competitive binding assays.
  • the competitor is a binding moiety known to bind to PBKot, such as an antibody, peptide, binding partner, ligand, etc. Under certain circumstances, there may be competitive binding as between the candidate agent and the binding moiety, with the binding moiety displacing the candidate agent.
  • the candidate agent is labeled. Either the candidate agent, or the competitor, or both, is added first to PI3Ka protein for a time sufficient to allow binding, if present. Incubations may be performed at any temperature that facilitates optimal activity, typically between 4°C and 40°C. [00150] Incubation periods are selected for optimum activity, but may also be optimized to facilitate rapid high throughput screening. Typically between 0.1 and 1 hour will be sufficient. Excess reagent is generally removed or washed away. The second component is then added, and the presence or absence of the labeled component is followed, to indicate binding.
  • the competitor is added first, followed by the candidate agent.
  • Displacement of the competitor is an indication the candidate agent is binding to ⁇ and thus is capable of binding to, and potentially modulating, the activity of the PBKoe.
  • either component can be labeled.
  • the presence of label in the wash solution indicates displacement by the agent.
  • the presence of the label on the support indicates displacement.
  • the candidate agent is added first, with incubation and washing, followed by the competitor.
  • the absence of binding by the competitor may indicate the candidate agent is bound to PDKcc with a higher affinity.
  • the candidate agent is labeled, the presence of the label on the support, coupled with a lack of competitor binding, may indicate the candidate agent is capable of binding to PI3Ka.
  • PI3Koc it may be of value to identify the binding site of PI3Koc. This can be done in a variety of ways. In one embodiment, once PI3Koc is identified as binding to the candidate agent, the PI3Ko is fragmented or modified and the assays repeated to identify the necessary components for binding.
  • Modulation is tested by screening for candidate agents capable of modulating the activity of PDKcc comprising the steps of combining a candidate agent with PI3Kcc, as above, and determining an alteration in the biological activity of the PI3Koc
  • the candidate agent should both bind to (although this may not be necessary), and alter its biological or biochemical activity as defined herein.
  • the methods include both in vitro screening methods and in vivo screening of cells for alterations in cell viability, morphology, and the like.
  • differential screening may be used to identify drug candidates that bind to native PI3Koc, but cannot bind to modified PI3Koc.
  • Positive controls and negative controls can be used in the assays. For example, all control and test samples are performed in at least triplicate to obtain statistically significant results. Incubation of samples is for a time sufficient for the binding of the agent to the protein. Following incubation, samples are washed free of non-specifically bound material and the amount of bound, generally labeled agent determined. For example, where a radioiabel is employed, the samples can be counted in a scintillation counter to determine the amount of bound compound.
  • a variety of other reagents can be included in the screening assays. These include reagents like salts, neutral proteins, e.g., albumin, detergents, etc which may be used to facilitate optimal protein-protein binding and/or reduce non-specific or background interactions. Also reagents that otherwise improve the efficiency of the assay, such as protease inhibitors, nuclease inhibitors, anti-microbial agents, etc., may be used. The mixture of components can be added in any order that provides for the requisite binding.
  • Such suitable x-ray quality crystals can be used as part of a method of identifying a candidate agent capable of binding to and modulating the activity of kinases.
  • Such methods may be characterized by the following aspects: a) introducing into a suitable computer program, information defining a ligand binding domain of a kinase in a conformation (e.g.
  • aspects a-d are not necessarily carried out in the aforementioned order. Such methods may further entail: performing rational drug design with the model of the three-dimensional structure, and selecting a potential candidate agent in conjunction with computer modeling.
  • Such methods may further entail: employing a candidate agent, so-determined to fit spatially into the ligand binding domain, in a biological activity assay for kinase modulation, and determining whether said candidate agent modulates kinase activity in the assay. Such methods may also include administering the candidate agent, determined to modulate kinase activity, to a mammal suffering from a condition treatable by kinase modulation, such as those described above.
  • compounds of the invention can be used in a method of evaluating the ability of a test agent to associate with a molecule or molecular complex comprising a ligand binding domain of a kinase.
  • a method may be characterized by the following aspects: a) creating a computer model of a kinase binding pocket using structure coordinates obtained from suitable x-ray quality crystals of the kinase, b) employing computational algorithms to perform a fitting operation between the test agent and the computer model of the binding pocket, and c) analyzing the results of the fitting operation to quantify the association between the test agent and the computer model of the binding pocket.
  • the invention provides pharmaceutical compositions comprising an inhibitor of PI3K according to the invention and a pharmaceutically acceptable carrier, excipient, or diluent.
  • administration is by the oral route.
  • Administration of the compounds of the invention, or their pharmaceutically acceptable salts, in pure form or in an appropriate pharmaceutical composition, can be carried out via any of the accepted modes of administration or agents for serving similar utilities.
  • administration can be, for example, orally, nasally, parenterally (intravenous, intramuscular, or subcutaneous), topically, transdermally, intra vaginally, intra vesically, intracistemally, or rectally, in the form of solid, semi-solid, lyophilized powder, or liquid dosage forms, such as for example, tablets, suppositories, pills, soft elastic and hard gelatin capsules, powders, solutions, suspensions, or aerosols, or the like, specifically in unit dosage forms suitable for simple administration of precise dosages.
  • the compositions will include a conventional pharmaceutical carrier or excipient and a compound of the invention as the/an active agent, and, in addition, may include carriers and adjuvants, etc.
  • Adjuvants include preserving, wetting, suspending, sweetening, flavoring, perfuming, emulsifying, and dispensing agents. Prevention of the action of microorganisms can be ensured by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, and the like. It may also be desirable to include isotonic agents, for example sugars, sodium chloride, and the like. Prolonged absorption of the injectable pharmaceutical form can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • a pharmaceutical composition of the invention may also contain minor amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents, antioxidants, and the like, such as, for example, citric acid, sorbitan monolaurate, triethanolamine oleate, butylalted hydroxytoluene, etc.
  • auxiliary substances such as wetting or emulsifying agents, pH buffering agents, antioxidants, and the like, such as, for example, citric acid, sorbitan monolaurate, triethanolamine oleate, butylalted hydroxytoluene, etc.
  • formulation depends on various factors such as the mode of drug administration (e.g., for oral administration, formulations in the form of tablets, pills or capsules) and the bioavailability of the drug substance.
  • pharmaceutical formulations have been developed especially for drugs that show poor bioavailability based upon the principle that bioavailability can be increased by increasing the surface area i.e., decreasing particle size.
  • U.S. Pat. No. 4,107,288 describes a pharmaceutical formulation having particles in the size range from 10 to 1,000 nm in which the active material is supported on a crosslinked matrix of macromolecules.
  • 5,145,684 describes the production of a pharmaceutical formulation in which the drug substance is pulverized to nanoparticles (average particle size of 400 nm) in the presence of a surface modifier and then dispersed in a liquid medium to give a pharmaceutical formulation that exhibits remarkably high bioavailability.
  • compositions suitable for parenteral injection may comprise physiologically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions.
  • aqueous and nonaqueous carriers, diluents, solvents or vehicles examples include water, ethanol, polyols (propylenegiycol, polyethyleneglycol, glycerol, and the like), suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate.
  • a coating such as lecithin
  • surfactants for example, water, alcohol, polyols (propylenegiycol, polyethyleneglycol, glycerol, and the like
  • suitable mixtures thereof examples include 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 a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants.
  • One specific route of administration is oral, using a convenient daily dosage regimen that can be adjusted according to the degree of severity of the disease-state to be treated.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the active compound is admixed with at least one inert customary excipient (or carrier) such as sodium citrate or dicalcium phosphate or
  • fillers or extenders as for example, starches, lactose, sucrose, glucose, mannitol, and silicic acid
  • binders as for example, cellulose derivatives, starch, alignates, gelatin, polyvinylpyrrolidone, sucrose, and gum acacia
  • humectants as for example, glycerol
  • disintegrating agents as for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, croscarmellose sodium, complex silicates, and sodium carbonate
  • solution retarders as for example paraffin
  • absorption accelerators as for example,
  • Solid dosage forms as described above can be prepared with coatings and shells, such as enteric coatings and others well known in the art. They may contain pacifying agents, and can also be of such composition that they release the active compound or compounds in a certain part of the intestinal tract in a delayed manner. Examples of embedded compositions that can be used are polymeric substances and waxes. The active compounds can also be in microencapsulated form, if appropriate, with one or more of the above-mentioned excipients.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs. Such dosage forms are prepared, for example, by dissolving, dispersing, etc., a compound(s) of the invention, or a
  • a carrier such as, for example, water, saline, aqueous dextrose, glycerol, ethanol and the like;
  • solubilizing agents and emulsifiers as for example, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propyleneglycol,
  • 1,3-butyleneglycol, dimethylformamide oils, in particular, cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil and sesame oil, glycerol, tetrahydrofurfuryl alcohol, polyethyleneglycols and fatty acid esters of sorbitan; or mixtures of these substances, and the like, to thereby form a solution or suspension.
  • oils in particular, cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil and sesame oil, glycerol, tetrahydrofurfuryl alcohol, polyethyleneglycols and fatty acid esters of sorbitan; or mixtures of these substances, and the like, to thereby form a solution or suspension.
  • Suspensions in addition to the active compounds, may contain suspending agents, as for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, or mixtures of these substances, and the like.
  • suspending agents as for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, or mixtures of these substances, and the like.
  • compositions for rectal administrations are, for example, suppositories that can be prepared by mixing the compounds of the present invention with for example suitable non- irritating excipients or carriers such as cocoa butter, polyethyleneglycol or a suppository wax, which are solid at ordinary temperatures but liquid at body temperature and therefore, melt while in a suitable body cavity and release the active component therein.
  • suitable non- irritating excipients or carriers such as cocoa butter, polyethyleneglycol or a suppository wax, which are solid at ordinary temperatures but liquid at body temperature and therefore, melt while in a suitable body cavity and release the active component therein.
  • Dosage forms for topical administration of a compound of this invention include ointments, powders, sprays, and inhalants.
  • the active component is admixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants as may be required.
  • Ophthalmic formulations, eye ointments, powders, and solutions are also contemplated as being within the scope of this invention.
  • Compressed gases may be used to disperse a compound of this invention in aerosol form.
  • Inert gases suitable for this purpose are nitrogen, carbon dioxide, etc.
  • compositions will contain about 1 % to about 99% by weight of a compound(s) of the invention, or a pharmaceutically acceptable salt thereof, and 99% to 1 % by weight of a suitable pharmaceutical excipient.
  • the composition will be between about 5% and about 75% by weight of a compound(s) of the invention, or a pharmaceutically acceptable salt thereof, with the rest being suitable pharmaceutical excipients.
  • composition to be administered will, in any event, contain a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof, for treatment of a disease-state in accordance with the teachings of this invention.
  • the compounds of the invention are administered in a therapeutically effective amount which will vary depending upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of the compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular disease-states, and the host undergoing therapy.
  • the compounds of the present invention can be administered to a patient at dosage levels in the range of about 0.1 to about 1 ,000 mg per day. For a normal human adult having a body weight of about 70 kilograms, a dosage in the range of about 0.01 to about 100 mg per kilogram of body weight per day is an example.
  • the specific dosage used can vary.
  • the dosage can depend on a number of factors including the requirements of the patient, the severity of the condition being treated, and the pharmacological activity of the compound being used.
  • the determination of optimum dosages for a particular patient is well known to one of ordinary skill in the art.
  • Certain compounds of this invention have been tested using the assay described in Biological Example I and have been determined to be PI3K inhibitors. As such compounds of Formula I are useful for treating diseases, particularly cancer in which PI3K activity contributes to the pathology and/or symptomatology of the disease.
  • cancer in which PI3K activity contributes to its pathology and/or symptomatology include breast cancer, colon cancer, rectal cancer, endometrial cancer, gastric carcinoma, glioblastoma, hepatocellular carcinoma, small cell lung cancer, non-small cell lung cancer, melanoma, ovarian cancer, cervical cancer, pancreatic cancer, prostate carcinoma, acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), or thyroid carcinoma
  • AML acute myelogenous leukemia
  • CML chronic myelogenous leukemia
  • Suitable in vitro assays for measuring PI3K activity and the inhibition thereof by compounds are known in the art.
  • an in vitro assay for measuring PI3K activity see Biological Examples, Example 1 infra. Following the examples disclosed herein, as well as that disclosed in the art, a person of ordinary skill in the art can determine the inhibitory activity of a compound of this invention.
  • Assays for measurement of in vitro efficacy in treatment of cancer are known in the art.
  • cell-based tumor models are described in Biological Examples, Example 2, 3, and 4 infra.
  • Suitable in vivo models for cancer are known to those of ordinary skill in the art.
  • in vivo models for prostate adenocarcinoma, glioblastoma, lung carcinoma, and melanoma see Biological Examples 5, 6, 7, 8, 9, and 10, infra.
  • Compounds of this invention can be made by the synthetic procedures described below.
  • the starting materials and reagents used in preparing these compounds are either available from commercial suppliers such as Aldrich Chemical Co. (Milwaukee, Wis.), or Bachem (Torrance, Calif.), or are prepared by methods known to those skilled in the art following procedures set forth in references such as Fieser and Fieser's Reagents for Organic Synthesis, Volumes 1-17 (John Wiley and Sons, 1991); Rodd's Chemistry of Carbon Compounds, Volumes 1-5 and Supplemental (Elsevier Science Publishers, 1 89); Organic Reactions, Volumes 1-40 (John Wiley and Sons, 1991), March's Advanced Organic Chemistry, (John Wiley and Sons, 4 lh Edition) and Larock's Comprehensive Organic Transformations (VCH Publishers Inc., 1989).
  • the reactions described herein take place at atmospheric pressure and over a temperature range from about -78 °C to about 150 °C, more specifically from about 0°C. to about 125 °C and more specifically at about room (or ambient) temperature, e.g., about 20 °C. Unless otherwise stated (as in the case of an hydrogenation), all reactions are performed under an atmosphere of nitrogen.
  • Prodrugs can be prepared by techniques known to one skilled in the art. These techniques generally modify appropriate functional groups in a given compound. These modified functional groups regenerate original functional groups by routine manipulation or in vivo. Amides and esters of the compounds of the present invention may be prepared according to conventional methods. A thorough discussion of prodrugs is provided in T. Higuchi and V. Stella, "Pro-drugs as Novel Delivery Systems," Vol 14 of the A.C.S.
  • the compounds of the invention may have asymmetric carbon atoms or quaternized nitrogen atoms in their structure.
  • Compounds of Formula I that may be prepared through the syntheses described herein may exist as single stereoisomers, racemates, and as mixtures of enantiomers and diastereomers.
  • the compounds may also exist as geometric isomers. All such single stereoisomers, racemates and mixtures thereof, and geometric isomers are intended to be within the scope of this invention.
  • Some of the compounds of the invention may exist as tautomers.
  • the molecule may exist in the enol form; where an amide is present, the molecule may exist as the imidic acid; and where an enamine is present, the molecule may exist as an imine. All such tautomers are within the scope of the invention.
  • imidazol-5-yl and pyrazol-5-yl each can also exist in their respective tautomeric forms imidazol-4-yl and pyrazol-3-yI. Regardless of which structure or which terminology is used, each tautomer is included within the scope of the Invention.
  • the present invention also includes N-oxide derivatives and protected derivatives of compounds of Formula I.
  • compounds of Formula I when compounds of Formula I contain an oxidizable nitrogen atom, the nitrogen atom can be converted to an N-oxide by methods well known in the art.
  • compounds of Formula I When compounds of Formula I contain groups such as hydroxy, carboxy, thiol or any group containing a nitrogen atom(s), these groups can be protected with a suitable "protecting group” or "protective group”.
  • a comprehensive list of suitable protective groups can be found in T.W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, Inc. 1991, the disclosure of which is incorporated herein by reference in its entirety.
  • the protected derivatives of compounds of Formula I can be prepared by methods well known in the art.
  • stereoisomers from racemic mixtures or non-racemic mixtures of stereoisomers are well known in the art.
  • optically active (R)- and (S)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques.
  • Enantiomers may be resolved by methods known to one of ordinary skill in the art, for example by: formation of diastereoisomeric salts or complexes which may be separated, for example, by crystallization; via formation of diastereoisomeric derivatives which may be separated, for example, by crystallization, selective reaction of one enantiomer with an enantiomer-specific reagent, for example enzymatic oxidation or reduction, followed by separation of the modified and unmodified enantiomers; or gas-liquid or liquid
  • enantiomeric form Alternatively, specific enantiomer may be synthesized by asymmetric synthesis using optically active reagents, substrates, catalysts or solvents or by converting on enantiomer to the other by asymmetric transformation.
  • enantiomers enriched in a particular enantiomer, the major component enantiomer may be further enriched (with concomitant loss in yield) by recrystallization.
  • the compounds of the present invention can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like.
  • the solvated forms are considered equivalent to the unsolvated forms for the purposes of the present invention.
  • a compound of the invention where R 1 is optionally substituted alkyl, R 2 is hydrogen or optionally substituted alkyl, R 4 is methyl or ethyl, R 6 is phenyl or heteroaryl each of which is optionally substituted with 1, 2, 3, 4, or 5 R 9 groups (as defined in the Summary of the Invention), and R 2 is hydrogen can be prepared according to Scheme 1.
  • An intermediate of formula 2 is prepared by reacting an intermediate of formula 1 with a primary amine R'NH 2 in a solvent such as water and with heating. 2 is then treated with iodine monochloride in a solvent such as methanol at around 0 °C and allowed to react for approximately overnight or less as needed for the reaction to go to completion to form 3. After completion the residue is triturated with acetone. The intermediate 3 is then reacted in a solvent, such as DMA, with ethyl acrylate in the presence of a base, such as triethylamine, and in the presence of a catalyst, such as Pd(OAc) 2 , and (+)BINAP. The reaction is heated to approximately 100 °C and allowed to react for approximately overnight or less as needed for the reaction to go to completion to form 4. 4 is then optionally purified by column chromatography.
  • a solvent such as water and with heating. 2 is then treated with iodine monochloride in a solvent such as methanol at around 0
  • [00196] 5 is prepared by treating 4 with DBU in the presence of a base such as DIPEA at room temperature. Then the reaction mixture is heated to reflux and reacted for
  • a Suzuki coupling is then performed using 6 reacting with a boronic acid (or ester) of formula R 6 B(OH) 2 in a solvent(s) such as a DME-H 2 0 mixture, in the presence of a catalyst such as Pd(dpppf ) and a base such as triethylamine at room temperature.
  • a catalyst such as Pd(dpppf )
  • a base such as triethylamine
  • R 1 is optionally substituted alkyl
  • R 4 is methyl or ethyl
  • R 6 is phenyl or heteroaryl each of which is optionally substituted with 1, 2, 3, 4, or 5 R 9 groups (as defined in the Summary of the Invention)
  • R 2 is hydrogen
  • An intermediate of formula 9 is prepared by reacting an intermediate of formula 8 with neat POCl 3 and heating. 9 is then treated with a primary amine R'NH 2 in a solvent such as water or THF and triethylamine at 0 °C to form 10. After removal of the solvent under reduced pressure, the intermediate 10 is then reacted with lithium aluminum hydride in a solvent such as THF at 0 °C. After quenching and aqueous workup, solvent removal provided crystalline 11 without further purification. Treatment of 11 with manganese (II) dioxide in a solvent such as methylene chloride or chloroform at room temperature provided aldehyde 12 upon Filtration and solvent removal.
  • a solvent such as water or THF and triethylamine
  • a Wittig reaction with aldehyde 12 can be employed with (carbethoxymethylene)triphenylphosphorane in refluxing THDF to provide the common intermediate 4. 4 can then be used to prepare a Compound of Formula I using the procedures described in Scheme 1.
  • a compound of the invention where R 1 is optionally substituted alkyl, R 4 is methyl or ethyl, R 6 is phenyl or heteroaryl each of which is optionally substituted with 1, 2, 3, 4, or 5 R 9 groups (as defined in the Summary of the Invention), and R 2 is hydrogen can be prepared according to Scheme 3.
  • An intermediate of formula 14 is prepared by reacting an intermediate of formula 13 with a primary amine R'NHa in a solvent such as water and with heating. 14 is then treated with iodine monochloride in a solvent such as methanol at around 0 °C and allowed to react for approximately overnight or less as needed for the reaction to go to completion to form IS. After completion the residue is triturated with acetone. The intermediate 15 is then reacted in a solvent, such as DMA, with ethyl acrylate in the presence of a base, such as triethylamine, and in the presence of a catalyst, such as Pd(OAc) 2 , and (+)BINAP.
  • a solvent such as DMA
  • ethyl acrylate in the presence of a base, such as triethylamine
  • a catalyst such as Pd(OAc) 2 , and (+)BINAP.
  • reaction is heated to approximately 100 °C and allowed to react for approximately overnight or less as needed for the reaction to go to completion to form 16.
  • 16 is then optionally purified by column chromatography.
  • a Compound of Formula I can then be prepared from 16 by using the same reaction conditions as described in Scheme 1 (starting at the point of the preparation of 5 from 4).
  • R 1 is optionally substituted alkyl
  • R 4 is methyl or ethyl
  • R 6 is phenyl or heteroaryl each of which is optionally substituted with 1, 2, 3, 4, or 5 R 9 groups (as defined in the Summary of the Invention)
  • R 2 is hydrogen
  • An intermediate of formula 20 is prepared by reacting an intermediate of formula 19 with neat POCl 3 and heating. 20 is then treated with a primary amine R*NH 2 in a solvent such as water or THF and triethylamine at 0 °C to form 21. After removal of the solvent under reduced pressure, the intermediate 21 is then reacted with lithium aluminum hydride in a solvent such as THF at 0 °C. After quenching and aqueous workup, solvent removal provided crystalline 22 without further purification. Treatment of 22 with manganese (II) dioxide in a solvent such as methylene chloride or chloroform at room temperature provided aldehyde 23 upon filtration and solvent removal.
  • a solvent such as water or THF and triethylamine
  • a Knovenegal-type condensation with 23 and an arylacetonitrile in the presence of a base such as potassium carbonate or sodium hydroxide in a protic solvent provides the cyclized imine 24.
  • Acetylation of the imine with acetic anhydride is required prior to hydrolysis which takes place in the presence of aqueous acid and heating to afford 25.
  • 25 can be oxidized to the corresponding sulfone with m-CPBA at room temperature and displaced with ammonium to provide I.
  • reaction mixture was heated to reflux and reacted for 4 h. After cooling down to room temperature, the reaction mixture was partitioned with water and ethyl acetate. After separation, the organic layer was dried with Na 2 S0 4 , and the product 8-ethyl-4-methyl- 2-(methylthio)-6-(lH-pyrazol-5-yl)pyrido[2,3-d]pyrimidin-7(8H)-one (0.567 g, 77% yield) was obtained by silica gel column chromatography.
  • N,N-Dimethyl acetamide dimethyl acetal (75 g, 0.56 mole) was added to a suspension of thiourea (33.0 g, 0.43 mole) in methylene chloride. The mixture was heated under reflux for 4 h. The solvent was removed and the residue was crystallized from 5% MeOH and diethyl ether affording (lE)-N'-(aminocarbonothioyl)-N,N- dimeth lethanimidamide (47.8 g, 76% yield).
  • reaction mixture was heated to 95 °C and stirred for 3 h (monitored by LC MC). After reaction completion, the reaction mixture was evaporated about to 1/10 of original volume and partitioned with 500 mL of ethyl acetate and 1000 mL of water. The aqueous layer was extracted with ethyl acetate 5 times.
  • reaction mixture including precipitate, was reduced to near dryness, filtered and washed with a minimal volume of cold THF, affording 2.88 g of 2-amino-8-isopropyl-4-methylpyrido[2,3- d]pyrimidin-7(8H)-one.
  • Example 4b 6-bromo-2-(te ⁇ butylamino)-8-ethyl-4-methylpyrido[2,3- ⁇ pyriraidin-7(8H)- one: ⁇ NMR (400 MHz, CDC1 3 ): ⁇ 8.08 (s, IH), 5.47 (bs, IH), 4.48 (m, 2H), 2.50 (s, 3H), 1.58 (bs, 3H), 1.49 (s, 9H), MS (EI) for C l4 Hi 9 BrN 4 0: 339.2 (MlF)
  • Example 4c 6-Bromo-2-(cyclopentylamino)-8-ethyl-4-methylpyrido[2,3--i]pyrimidin-7(8H)- one: ⁇ NMR (400 MHz, CDC1 3 ): 6 8.07 (s, IH), 5.89 (bs, IH), 4.49 (bd, 2H), 2.51 (s, 3H), 2.07 (m, 2H), 1.71 (m, 2H), 1.58 (m, 2H), 1.31 (t, 3H), MS (EI) for C 15 H, 9 BrN 4 0: 351.2 (MH*)
  • Example 4d 6-Bromo-2-(cyclohexylamino)-8-ethyl-4-methylpyrido[2,3-ii
  • pyrimidin-7(8H)- one: ⁇ NMR (400 MHz, CDC1 3 ): ⁇ 8.07 (s, IH), 5.41 (bs, IH), 4.47 (bd, 2H), 3.84 (bs, IH), 2.51 (s, 3H), 2.05 (d, 7 12.4 Hz, 2H), 1.77 (m, 2H), 1.64 (br m, 4H), 1.39 (m, 2H), 1.30 (m, 3H), MS (EI) for C
  • Example 4f 6-B ⁇ omo-8-ethyl-4-nlethyl-2-(2-mo holinoethylamino
  • Ci 7 Hi 7 BrN 4 0: 375.1 (M2H ⁇ .Example 4p. 6-Bromo-8-emyl-2-(emylamino)-4- methylpyrido[2,3-ii]pyrimidin-7(8H)-one: ⁇ NMR (400 MHz, CDC1 3 ): ⁇ 8.09 (s, IH), 5.71 (bs, IH), 4.48 (bs, 2H), 3.54 (q, J 6.8 Hz, 2H), 2.53 (s, 3H), 1.16 (m, 6H); MS (EI) for C,2H, 5 BrN 4 0: 31 1.9 (MH + ).
  • Pd(dppf) dichloromethane adduct (0.077 g, 0.095 mmol) was added to a suspension of 6-bromo-2-(ethylamino)-4-methyl-8-(l -methylethyl)pyrido[2,3-if
  • Example 5f 6-(2,4-Difluorophenyl)-8-ethyl-2-(ethylamino)-4-methylpyrido[2,3- i/]pyrimidin-7(8H)-one: ⁇ NMR (400 MHz, CDC1 3 ): ⁇ 7.78 (d, 2H), 7.52 (m, IH), 6.85 (m, 2H), 5.38 (bs, IH), 4.48 (m, 2H), 3.56 (m, 2H), 2.57 (s, 3H), 1.39 (m, 6H); MS (EI) for
  • Example 5h 6-(2,4-Dichlorophenyl)-8-ethyI-2-(ethylamino)-4-methylpyrido[2,3- df]pyrimidin-7(8H)-one: ⁇ NMR (400 MHz, CDC1 3 ): ⁇ 7.75 (s, IH), 7.42 (d, IH), 7.38 (m, 2H), 5.38 (bs, IH), 4.42 (m, 2H), 3.59 (m, 2H), 2.56 (s, 3H), 1.24 (m, 6H); MS (EI) for C, 8//I8 C1 2 N 4 0: 377.0 (M + ), 379.0 (M+2)
  • Example 5i 6-(3,4-Difluorophenyl)-8-ethyl-2-(ethylamino)-4-methylpyrido[2,3- i/]pyrimidin-7(8H)-one: ⁇ NMR (400 MHz, CDC1 3 ): ⁇ 7.79 (s, I H), 7.59 (m, IH), 7.39 (m, IH), 7.18 (m, IH), 5.39 (bs, IH), 4.46 (m, 2H), 3.58 (m, 2H), 2.59 (s, 3H), 1.27 (m, 6H); MS (EI) for C, 8W i 8 F 2 N 4 0: 345.1 (MIT).
  • Example 5j 8-Ethyl-2-(ethylamino)-4-methyl-6-[4-(phenyloxy)phenyl]pyrido[2,3- iflpyriniidin-7(8H)-one: ⁇ NMR (400 MHz, CDC1 3 ): ⁇ 7.78 (s, IH), 7.63 (d, 2H), 7.39 (t, 2H), 7.16 (t, IH), 7.04 (d, 4H), 5.38 (bs, IH), 4.47 (m, 2H), 3.57 (m, 2H), 2.59 (s, 3H), 1.26 (m, 6H); MS (EI) for C 24 H2 4 4 0 2 : 401.1 (MH + ).
  • Example 5k 8-Ethyl-2-(ethylamino)-4-methyl-6-naphthaleN- 1 -ylpyrido[2,3-i ]pyrimidin- 7(8H)-one: ⁇ NMR (400 MHz, CDC1 3 ): ⁇ 7.84 (d, 2H), 7.80 (s, IH), 7.73 (d, IH), 7.48 (m, 4H), 539 (bs, IH), 4.55 (bs, 2H), 3.59 (m, 2H), 2.54 (s, 3H), 1.37 (m, 6H); MS (EI) for C 22 H 22 N 4 0: 359.1 (MIT).
  • Example 5aa 8-Ethyl-2-(ethylamino)-4-methyl-6-(lH-pyrazol-5-yl)pyrido[2,3-ii
  • Example 5bb 8- ⁇ 1-4- ⁇ 6 1-6-(1 ⁇ 3 ⁇ 1-5- 1)-2-[(2,2,2- trifluoroethyl)arnino]pyrido[2,3--i]pyrimidin-7(8H)-one: ⁇ NMR (400 MHz, CDC1 3 ): ⁇ 8.18 (s, IH), 7.63 (d, IH), 6.73 (d, IH), 5.62 (bs, IH), 4.58 (m, 2H), 4.30 (m, 2H), 2.74 (s, 3H), 1.35 (t, 3H); MS (EI) for C 15 Hi5F 3 6 0: 353.0 (MH + ).
  • Example 6a 8-Ethyl-2-(ethylamino)-4-methyl-6-[4-(methyloxy)phenyl]pyrido[2,3- £/]pyridimidW-7(8H)-one: ⁇ NMR (400 MHz, CDC1 3 ): 6 7.81 (s, IH), 7.60 (d, 2H), 6.96 (d, 2H), 4.50 (q, 2H), 3.82 (s, 3H), 3.58 (q, 2H), 2.58 (s, 3H), 1.30 (m, 6H); MS (EI) for Ci 9 H 22 N 4 0 2 : 339.1 (MH*).
  • Example 6b 8-Ethyl-2-(ethylamino)-4-methyl-6-[4-(methyloxy)phenyl]pyrido[2,3- £/]pyridimidW-7(8H)-one: ⁇ NMR (400 MHz, CDC1 3 ): 6 7.81 (s, IH), 7.
  • Example 6c 6-[2,4-Bis(methyloxy)phenyl]-8-ethyl-2-(ethylamino)-4-methylpyrido[2,3- ⁇ /]pyrimidin-7(8H)-one: ⁇ NMR (400 MHz, CDCI3): 5 7.70 (s, IH), 7.30 (s, IH), 6.60 (m, 2H), 4.50 (q, 2H), 3.82 (s, 3H), 3.80 (s, 3H), 3.45 (q, 2H), 2.50 (s, 3H), 1.30 (m, 6H); MS (EI) for C 2 oH 24 N 03: 369.1 (MH + ).
  • Example 6d 8-Ethy l-2-(ethylamino)-4-methyl-6-[3-(methyloxy)phenyl]pyrido[2,3- .flpyridimidiN-7(8H)-one: ⁇ NMR (400 MHz, CDC1 3 ): ⁇ 7.81 (s, IH), 7.60 (d, 2H), 6.96 (d, 2H), 4.50 (q, 2H), 3.80 (s, 3H), 3.58 (q, 2H), 2.50 (s, 3H), 1.30 (m, 6H); MS (EI) for
  • Example 6e 8-(5-Chloro-2-thienyl)-8-ethyl-2-(ethylamino)-4-methylpyrido[2,3-rf]pyrimidin- 7(8H)-one: ⁇ NMR (400 MHz, CDC1 3 ): ⁇ 8.00 (s, IH), 7.38 (d, 2H), 6.96 (d, 2H), 4.50 (q, 2H), 3.58 (q, 2H), 2.60 (s, 3H), 1.30 (m, 6H); MS (EI) for C 16 H
  • Example 6f 8-(5-Chloro-2-thienyl)-8-ethyl-2-(ethylamino)-4-methylpyrido[2,3-rf]pyrimidin- 7(8H)-one: ⁇ NMR (400 MHz, CDC1 3 ): ⁇ 8.00 (s, IH), 7.38 (d, 2H
  • Example 6h 8-Ethy l-2-(ethylamino)-6-( lH-indole-6-yl)-4-methylpyrido[2,3-i/
  • Example 6i 8-Ethy 1 -2-(ethylamino)-4-methyl-6-(5-phenyl-2-thienyl)pyrido[2,3- ⁇ flpyrimidin-7(8H)-one: ⁇ NMR (400 MHz, DMSO-rf 6 ): ⁇ 8.40 (s, IH), 7.81 (d, IH), 7.70 (d, 2H), 7.50 (d, IH), 7.42 (m, 2H), 7.30 (m, IH), 4.40 (q, 2H), 3.40 (q, 2H), 2.42 (s, 3H), 1.30 (m, 6H); MS (EI) for C 22 H 22 N 4 OS: 3 1.3 (MH + ).
  • Example 6j 8-Ethy 1 -2-(ethylamino)-4-methyl-6-(5-phenyl-2-thienyl)pyrido[2,3- ⁇ flpyrimidin-7(8H)-one: ⁇ NMR (400
  • Example 6k 8-Ethyl-2-(ethylamino)-6-(3-fluorophenyl)-4-methylpyrido[2,3-ii
  • Triethylamine (3.4 mL, 24.6 mmol) was added to a suspension of 2-amino-4- chloro-6-methylpyrimidine (Aldrich, 1.77 g, 12.3 mmol) and benzylamine ( 1.98 g, 18.5 mmol) in anhydrous dioxane (20 mL). The reaction was heated to 80 °C and allowed to run for 12 h. Upon cooling to room temperature, a white precipitate formed which was collected by vacuum filtration. The solid was recrystallized from acetone: hexanes to afford N'-benzyl- 6-methylpyrimidine-2, -diamine (2.33 g, 89 % yield) as a white solid.
  • Triethylamine (7.60 mL, 54.5 mmol) was added to a suspension of N*-benzyl- 5-iodo-6-methylpyrimidine-2,4-diamine (3.14 g, 10.9 mmol), ethyl acrylate (3.55 mL, 32.7 mmol) and Pd(PPh 3 ) 4 (629 mg, 0.545 mmol) in anhydrous DMF (20 mL). The reaction was heated to 95 °C under nitrogen. After 24 h, the reaction was allowed to cool to room temperature and concentrated in vacuo. The residue was poured into a 10% solution of LiCl and washed with ethyl acetate (100 mL).
  • a 10 1 solution of dioxane and water ( 1 1 mL) was added to a flask charged with 2-amino-6-bromo-4-methy l-(8-pheny lmethyl)pyrido[2,3-ti] pyrimidin-7(8H)-one (0.435 g, 1.27 mmol), lH-pyrazole-5-boronic acid (0.284 g, 2.54 mmol), Pd(PPh ) 4 (0.073 mg, 0.063 mmol), and K 2 C0 3 (0.527 g, 3.81 mmol). The flask was flushed with nitrogen and fitted with a reflux condenser and heated to 1 10 °C.
  • a 3 1 solution of dioxane and water (4 mL) was added to a flask charged with 2-amino-6-bromo-8-e I-4-methyIpyrido[2,3-
  • Example 9b 2-Amino-8-emyl-6-furan-3-yl-4-methylpyrido[2,3-£f
  • Example 9j 2-Amino-8-ethyl-4-methyl-6-phenylpyrido[2,3-ii
  • Trimethylsilylethyne ( 1.44 mL, 10.2 mmol) was added to a pressure tube charged with 2-amino-6-bromo-8-emyl-4-methylpyrido[2,3-if
  • Acetic anhydride 10.0 mL was added to a flask charged with 8-ethyl-7- imino-4-memyl-2-(memylthio)-7,8-dihydropyrido[2,3-iilpyrimidine-6-carbonitrile (0.506 g, 1.95 mmol) and heated to 100 °C. After one h, the reaction was allowed to cool to room temperature and concentrated in vacuo. The acetylated residue was then treated with 6 N HCl (40 mL) and heated to 95 °C for one hour then transferred to a large flask.
  • Tributyltin azide (660 uL, 2.41 mmol) was added to a flask charged with 2-amino-8-emyl-4-memyl-7-oxo-7,8-dihydropyrido[2,3-(i]pyrimidine-6-carbomtrile (0.184 g, 0.803 mmol) in anhydrous toluene (5.0 mL). The reaction was fitted with a reflux condenser and heated to 140 °C under a nitrogen atmosphere.
  • Example 13a 6-bromo-8-(2-ethoxyethyl)-2-(ethylamino)-4-methylpyrido[2,3-d]pyrimidin- 7(8H)-one: ⁇ NMR (400MHz, CDC1 3 ) ⁇ 8.09 (s, 1H), 5.37 (Br. s, 1H), 4.67 (m, 2H), 3.74 (m, 2H), 3.61-3.56 (t, 2H), 3.51 (m, 2H), 2.53 (s, 3H), 1.29-1.25 (t, 3H), 1.19-1.15 (t, 3H); MS (EI) for C l4 H, 9 BrN 0 2 : 355 (MH+).
  • Example 13b 6-bromo-8-(3-ethoxypropyl)-2-(ethylamino)-4-methylpyrido[2,3-d]pyrimidin- 7(8H)-one: ⁇ NMR (400MHz, CDC1 3 ) ⁇ 8.09 (s, 1H), 5.37 (Br. s, 1H), 4.53 (m, 2H), 3.52 (m, 4H), 3.48-3.43 (m, 2H), 2.53 (s, 3H), 2.04-2.00 (m, 2H), 1.29-1.25 (t, 3H), 1.19-1.15 (t, 3H); MS (EI) for C 15 H 2 iBrN 4 0 2 : 369 (MH+).
  • Example 13c 6-bromo-2-(ethyIamino)-8-(3-isopropoxypropyl)-4-methylpyrido[2,3- d]pyrimidin-7(8H)-one: ⁇ NMR (400MHz, CDC1 3 ) ⁇ 8.09 (s, 1H), 5.37 (Br. s, IH), 4.53 (m, 2H), 3.59-3.49 (m, 5H), 2.52 (s, 3H), 2.01-1.98 (m, 2H), 1 .28-1.25 (t, 3H), 1.13- 1.1 1 (t, 6H); MS (EI) for C, 6 H23BrN 4 0 2 : 383 (MH+).
  • Example 14a 6-bromo-8-cyclopropyl-2-(ethyIamino)-4-methylpyrido[2,3-d] pyrimidin- 7(8H)-one: ⁇ NMR (400 MHz, CDC1 3 ) ⁇ 8.06 (s, 1 H), 5.37 (br s, 1 H), 3.54 (m, 2 H), 2.94 (br s, 1H), 2.51 (s, 3 H), 1.31-1.25 (m, 5 H), 0.91 (br s, 2 H); MS (EI) for C, 3 H, 5 BrN 4 0: 323 (MH+).
  • Example 15a 6-bromo-2-(ethylamino)-8-(2-hydroxyethyl)-4-methylpyrido[2,3-d]pyrimidin- 7(8H)-one: ⁇ NMR (400 MHz, DMSO-d6) ⁇ 8.38 (s, 1 H), 4.82 (br s, 1 H), 4.40 (br s, 2 H), 3.62-3.55 (m, 2 H), 3.40-3.20 (m, 3 H), 2.55 (s, 3 H), 1.15 (t, 3 H); MS (EI) for C, 2 Hi 5 BrN 4 0 2 : 327 (MH+).
  • Example 15b 6-bromo-2-(ethylamino)-4-methyl-8-(2-(piperidin- 1 -yl)ethyl)pyrido[2,3- d]pyrimidin-7(8H)-one: ⁇ NMR (400 MHz, CDC1 3 ) ⁇ 8.08 (s, 1 H), 5.39 (br s, 1 H), 4.59 (br s, 2 H), 3.55-3.40 (m, 2 H), 2.70-2.50 (m, 6 H), 2.52 (s, 3 H), 1.62-1.58 (m, 4 H), 1.46- 1.40 (m, 2 H), 1.27 (t, 3 H); MS (EI) for C 17 H 24 BrN 5 0: 394 (MH+).
  • PI3Ka activity is measured as the percent of ATP consumed following the kinase reaction using luciferase-luciferin-coupled chemiluminescence. Reactions were conducted in 384-well white, medium binding microtiter plates (Greiner). Kinase reactions were initiated by combining test compounds, ATP, substrate (PIP2), and kinase in a 20 ⁇ . volume in a buffer solution.
  • the standard PDKalpha assay buffer is composed 50 mM Tris, pH 7.5, 1 mM EGTA, 10 mM MgCl 2 , 1 mM DTT and 0.03% CHAPS. The standard assay
  • the PI3K inhibitor is selected from the compounds in Table 1 having a PI3K-binding affinity of about 9 ⁇ or less.
  • the PI3K inhibitor is selected from the compounds in Table 1 having a PI3K-binding affinity of about 5 ⁇ or less.
  • the PI3K inhibitor is selected from the compounds in Table 1 having a PI3K-binding affinity of about 3 ⁇ or less.
  • the PI3K inhibitor is selected from the compounds in Table 1 having a PI3K-binding affinity of about 1.5 ⁇ or less.
  • the PI3K inhibitor is selected from the compounds in Table 1 having a PI3K-binding affinity of about 1 ⁇ or less. In another embodiment, the PI3K inhibitor is selected from the compounds in Table 1 having a PI3K-binding affinity of about 0.6 ⁇ or less. In another embodiment, the PI3K inhibitor is selected from the compounds in Table 1 having a PI3K- binding affinity of about 0.3 ⁇ or less. In another embodiment, the PI3K inhibitor is selected from the compounds in Table 1 having a PI3K-binding affinity of about 0.2 ⁇ or less. In another embodiment, the PI3K inhibitor is selected from the compounds in Table 1 having a PI3K-binding affinity of about 0.1 ⁇ or less.
  • the PI3K inhibitor is selected from the compounds in Table 1 having a PI3K-binding affinity of about 0.04 ⁇ or less. In another embodiment, the PI3K inhibitor is selected from the compounds in Table 1 having a PI3K-binding affinity of about 0.020 ⁇ or less.
  • PC3 cells were seeded on 6-well plates at 150,000 cells/well. Cells were cultured for 3 days, then treated with compounds in serum-free medium for 3 hr. EGF (100 ng mL) was added for the last 10 min. Cells were lysed in TENN buffer. Phospho T308 Akt and total Akt were quantified by ELISA performed according to the Biosource assay protocol. The readings of phospho Akt were normalized to total Akt readings.
  • PC3 cells were seeded on 96-well plates at 8,000 cells/well. For each experiment, cells were seeded and treated in duplicated plates: one plate for phospho S6 CellELISA, and one plate for total S6 CellELISA. Cells were cultured on the plates for 3 days, then treated with compounds in serum-free medium for 3 hr in triplicate. Cells were fixed with 4% formaldehyde, quenched with 0.6% H 2 0 2 , blocked with 5% BSA, incubated with either phospho S6 antibody or total S6 antibody overnight, incubated with goat-anti-rabbit-IgG- HRP for 1 hr, and developed in chemiluminescent substrate. Biological Example 4
  • MCF-7 cells grown in 10-cm dishes were starved for 3 hours in DMEM, and then treated with compounds for 20 minutes. In the last 2 minutes of the incubation with the compounds, EGF (100 ng/mL) was added to stimulate the production of PDP3. The medium was aspirated and the cells were scraped with 10% trichloroacetic acid. The lipids were extracted from the pellet after the cell lysates were centrifuged. PIP3 in the cellular lipid extraction was quantified with the AlphaScreen assay in which Grpl-PH is used as the PIP3 specific probe. The amount of cellular PIP3 was calculated from the standard curve of diC 8 PI (3,4,5) P3.
  • mice Female and male athymic nude mice (NCr) 5-8 weeks of age and weighing approximately 20 g were used in the following model. Prior to initiation of a study, the animals were allowed to acclimate for a minimum of 48 h. During these studies, animals were provided food and water ad libitum and housed in a room conditioned at 70-75 °F and 60% relative humidity. A 12 h light and 12 h dark cycle was maintained with automatic timers. All animals were examined daily for compound-induced or tumor-related deaths.
  • PC-3 human prostate adenocarcinoma cells were cultured in vitro in DMEM (Mediatech) supplemented with 20% Fetal Bovine Serum (Hyclone), PenicilliN-Streptomycin and non-essential amino acids at 37°C in a humidified 5% C0 2 atmosphere. On day 0, cells were harvested by trypsinization and 3x l0 6 cells (passage 13, 99% viability) in 0.1 mL of ice-cold Hank's balanced salt solution were implanted subcutaneously into the hindflank of 5-8 week old male nude mice. A transponder was implanted in each mouse for identification, and animals were monitored daily for clinical symptoms and survival. Body weights were recorded daily.
  • U-87 MG human glioblastoma cells were cultured in vitro in DMEM (Mediatech) supplemented with 10% Fetal Bovine Serum (Hyclone), PenicilliN-Streptomycin and nonessential amino acids at 37°C in a humidified 5% C0 2 atmosphere.
  • DMEM Mediatech
  • Fetal Bovine Serum Hyclone
  • PenicilliN-Streptomycin nonessential amino acids at 37°C in a humidified 5% C0 2 atmosphere.
  • cells were harvested by trypsinization and 2xl0 6 cells (passage 5, 96% viability) in 0.1 mL of ice-cold Hank's balanced salt solution were implanted intradermally into the hindflank of 5-8 week old female nude mice.
  • a transponder was implanted in each mouse for identification, and animals were monitored daily for clinical symptoms and survival. Body weights were recorded daily.
  • A549 human lung carcinoma cells were cultured in vitro in DMEM (Mediatech) supplemented with 10% Fetal Bovine Serum (Hyclone), PenicilliN-Streptomycin and nonessential amino acids at 37°C in a humidified 5% C0 2 atmosphere. On day 0, cells were harvested by trypsinization and lOxlO 6 cells (passage 12, 99% viability) in 0.1 mL of ice-cold Hank's balanced salt solution were implanted intradermally into the hindflank of 5-8 week old female nude mice. A transponder was implanted in each mouse for identification, and animals were monitored daily for clinical symptoms and survival. Body weights were recorded daily.
  • A2058 human melanoma cells were cultured in vitro in DMEM (Mediatech) supplemented with 10% Fetal Bovine Serum (Hyclone), PenicilliN-Streptomycin and nonessential amino acids at 37°C in a humidified, 5% C0 2 atmosphere. On day 0, cells were harvested by trypsinization and 3xl0 6 cells (passage 3, 95% viability) in 0.1 mL ice-cold Hank's balanced salt solution were implanted intradermally in the hind-flank of 5-8 week old female athymic nude mice. A transponder was implanted in each mouse for identification, and animals were monitored daily for clinical symptoms and survival. Body weights were recorded daily.
  • WM-266-4 human melanoma cells were cultured in vitro in DMEM (Mediatech) supplemented with 10% Fetal Bovine Serum (Hyclone), PenicilliN-Streptomycin and nonessential amino acids at 37°C in a humidified, 5% C0 2 atmosphere. On day 0, cells were harvested by trypsinization and 3xl0 6 cells (passage 5, 99% viability) in 0.1 mL ice-cold Hank's balanced salt solution were implanted intradermally in the hind-flank of 5-8 week old female athymic nude mice. A transponder was implanted in each mouse for identification, and animals were monitored daily for clinical symptoms and survival. Body weights were recorded daily.
  • Tumor weight was determined by measuring perpendicular diameters with a caliper, using the following formula:
  • TGI Percent inhibition of tumor growth
  • Tumor size is calculated individually for each tumor to obtain a mean ⁇ SEM value for each experimental group. Statistical significance is determined using the 2-tailed Student's t-test (significance defined as P ⁇ 0.05).
  • the following ingredients are mixed to form a suspension for oral administration.
  • Veegum K (Vanderbilt Co.) 1.0 g
  • the following ingredients are mixed to form an injectable formulation.
  • a suppository of total weight 2.5 g is prepared by mixing the compound of the invention with Witepsol.RTM. H-15 (triglycerides of saturated vegetable fatty acid; Riches- Nelson, Inc., New York), and has the following composition:

Abstract

The invention relates to methods, combinations, and formulations of a compound of Formula I.

Description

PYRIDOPYRIMIDINO E INHIBITORS OF PI3K Alpha
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit of priority to U.S. Provisional Application No. 61/413,356, filed November 12, 2010, which is incorporated herein by reference.
BACKGROUND OF THE INVENTION
[0002] This invention relates to the field of protein kinases and inhibitors thereof. In particular, the invention relates to inhibitors of phosphatidylinositol 3-kinase (PI3K) signaling pathways, and methods of their use.
[0003] The connection between abnormal protein phosphorylation and the cause or consequence of diseases has been known for over 20 years. Accordingly, protein kinases have become a very important group of drug targets. See Cohen, Nature, 1:309-315 (2002). Various protein kinase inhibitors have been used clinically in the treatment of a wide variety of diseases, such as cancer and chronic inflammatory diseases, including diabetes and stroke. See Cohen, Eur. J. Biochem., 268:5001-5010 (2001).
[0004] The protein kinases are a large and diverse family of enzymes that catalyze protein phosphorylation and play a critical role in cellular signaling. Protein kinases may exert positive or negative regulatory effects, depending upon their target protein. Protein kinases are involved in specific signaling pathways which regulate cell functions such as, but not limited to, metabolism, cell cycle progression, cell adhesion, vascular function, apoptosis, and angiogenesis. Malfunctions of cellular signaling have been associated with many diseases, the most characterized of which include cancer and diabetes. The regulation of signal transduction by cytokines and the association of signal molecules with protooncogenes and tumor suppressor genes have been well documented. Similarly, the connection between diabetes and related conditions, and deregulated levels of protein kinases, has been demonstrated. See e.g., Sridhar et al. Pharmaceutical Research, 17(1 1): 1345-1353 (2000). Viral infections and the conditions related thereto have also been associated with the regulation of protein kinases. Park et al. Cell 101 (7), 777-787 (2000).
[0005] Phosphatidylinositol 3-kinase (PI3Ka), a dual specificity protein kinase, is composed of an 85 kDa regulatory subunit and a 1 10 kDa catalytic subunit. The protein encoded by this gene represents the catalytic subunit, which uses ATP to phosphorylate Ptdlns, PtdIns4P and PtdIns(4,5)P2. PTEN, a tumor suppressor which inhibits cell growth through multiple mechanisms, can dephosphorylate PIP3, the major product of PIK3CA. PIP3, in turn, is required for translocation of protein kinase B (AKT1, PKB) to the cell membrane, where it is phosphorylated and activated by upstream kinases. The effect of PTEN on cell death is mediated through the PIK3CA/AKT1 pathway.
[0006] PI3Ko has been implicated in the control of cytoskeletal reorganization, apoptosis, vesicular trafficking, proliferation and differentiation processes. Increased copy number and expression of PIK3CA is associated with a number of malignancies such as ovarian cancer (Campbell et al., Cancer Res 2004, 64, 7678-7681 ; Levine et al., Clin Cancer Res 2005, 1 1, 2875-2878; Wang et al., Hum Mutat 2005, 25, 322; Lee et al., Gynecol Oncol 2005, 97, 26-34), cervical cancer, breast cancer (Bachman, et al. Cancer Biol Ther 2004, 3, 772-775; Levine, et al., supra; Li et al., Breast Cancer Res Treat 2006, 96, 91-95; Saal et al., Cancer Res 2005, 65, 2554-2559; Samuels and Velculescu, Cell Cycle 2004, 3, 1221-1224), colorectal cancer (Samuels, et al. Science 2004, 304, 554; Velho et al. Eur J Cancer 2005, 41, 1649-1654), endometrial cancer (Oda et al. Cancer Res. 2005, 65, 10669-10673), gastric carcinomas (Byun et al., Int J Cancer 2003, 104, 318-327; Li et al., supra; Velho et al., supra; Lee et al., Oncogene 2005, 24, 1477-1480), hepatocellular carcinoma (Lee et al., id.), small and non-small cell lung cancer (Tang et al., Lung Cancer 2006, 1, 181-191 ; Massion et al., Am J Respir Crit Care Med 2004, 170, 1088-1094), thyroid carcinoma (Wu et al., J Clin Endocrinol Metab 2005, 90, 4688-4693), acute myelogenous leukemia (AML) (Sujobert et al., Blood 1997, 106, 1063-1066), chronic myelogenous leukemia (CML) (Hickey and Cotter J Biol Chem 2006, 281, 2441-2450), and glioblastomas (Hartmann et al. Acta Neuropathol {Berl) 2005, 109, 639-642; Samuels et al., supra).
[0007] In view of the important role of PI3 a in biological processes and disease states, inhibitors of this protein kinase are desirable.
SUMMARY OF THE INVENTION
[0008] The following only summarizes certain aspects of the invention and is not intended to be limiting in nature. These aspects and other aspects and embodiments are described more fully below. All references cited in this specification are hereby incorporated by reference in their entirety. In the event of a discrepancy between the express disclosure of this specification and the references incorporated by reference, the express disclosure of this specification shall control.
[0009] The invention provides compounds that inhibit, regulate, and/or modulate PI3K that are useful in the treatment of hyperproliferative diseases, such as cancer, in humans. This invention also provides methods of making the compound, methods of using such compounds in the treatment of hyperproliferative diseases in humans and to pharmaceutical compositions containing such compounds.
[0010] A first aspect of the invention provides a compound of Formula I:
Figure imgf000004_0001
or a pharmaceutically acceptable salt or solvate thereof, wherein
R1 is hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted arylalkyl, optionally substituted heterocycloalkyl, optionally substituted
heterocycloalkylalkyl, optionally substituted heteroaryl or optionally substituted heteroarylalkyl;
R2 is hydrogen or alkyl where the alkyl is optionally substituted with 1, 2, 3, 4, or 5 R8 groups;
X is -NR3-;
R3 hydrogen;
R4 is optionally substituted alkyl;
R5 is hydrogen; and
R6 is phenyl, acyl, or heteroaryl wherein the phenyl and heteroaryl are optionally substituted with 1, 2, 3, 4, or 5 R9 groups;
each R8, when present, is independently hydroxy, halo, alkoxy, haloalkoxy, amino,
alkylamino, dialkylaminoalkyl, or alkoxyalkylamino; and
each R9, when present, is independently halo, alkyl, haloalkyl, alkoxy, haloalkoxy, cyano, amino, alkylamino, dialkylamino, alkoxyalkyl, carboxyalkyl, alkoxycarbonyl, aminoalkyl, cycloalkyl, aryl, arylalkyl, aryloxy, heterocycloalkyl, or heteroaryl and where the cycloalkyl, aryl, heterocycloalkyl, and heteroaryl, each either alone or as part of another group within R9, are independently optionally substituted with 1, 2, 3, or 4 groups selected from halo, alkyl, haloalkyl, hydroxy, alkoxy, haloalkxy, amino, alkylamino, and dialkylamino.
[0011] A second aspect of the invention provides a compound of Formula II:
Figure imgf000005_0001
II
or a pharmaceutically acceptable salt or solvate thereof, wherein
R1 is hydrogen, optionally substituted alkyl, optionally substituted C3-C7 cycloalkyl,
optionally substituted aryl, optionally substituted arylalkyl, optionally substituted heterocycloalkyl, optionally substituted heterocycloalkylalkyl, optionally substituted heteroaryl or optionally substituted heteroarylalkyl;
X is S, S02, or -NR3-;
R is hydrogen, haloalkyl, optionally substituted alkyl, optionally substituted C3-O7
cycloalkyl, optionally substituted aryl, optionally substituted arylalkyl, optionally substituted heterocycloalkyl, optionally substituted heterocycloalkylalkyl, optionally substituted heterocycloalkyl-aryl- or optionally substituted heteroaryl; R2 is optionally further substituted with one or more R8 groups;
R3, R3a, and R3b are independently hydrogen, optionally substituted alkyl, optionally
substituted C3-C7 cycloalkyl, optionally substituted aryl, optionally substituted heterocycloalkyl or optionally substituted heteroaryl;
R4 is hydrogen, halo, haloalkyl, haloalkoxy, -NR3a-, optionally substituted alkyl, optionally substituted C1-C6 alkoxy, optionally substituted C|-C6 alkoxyalkyl, optionally substituted aminoaikyl, optionally substituted C3-C7 cycloalkyl, optionally substituted aryl, or optionally substituted heteroaryl;
R5 is hydrogen, halo, haloalkyl, haloalkoxy, optionally substituted Q-C6 alkyl, optionally substituted Ci-C6 alkoxy, optionally substituted Ci-C6 alkoxyalkyl, optionally substituted aminoaikyl, optionally substituted C3-C7 cycloalkyl, optionally substituted aryl, optionally substituted aryl Ci-C6 alkyl or optionally substituted heteroaryl; and
R6 is hydrogen, halo, haloalkyl, haloalkoxy, -NR3b-, optionally substituted Ci-C6 alkyl, optionally substituted Q-C6 alkoxy, optionally substituted C|-C6 alkoxyalkyl, optionally substituted acyl, optionally substituted aminoaikyl, optionally substituted C3-C7 cycloalkyl, optionally substituted aryl, optionally substituted arylalkyl, optionally substituted heterocycloalkyl, or optionally substituted heteroaryl;
substitutable R6 groups are optionally further substituted with 1 , 2, 3, 4, or 5 R9 groups; each R8, when present, is independently hydroxy, halo, haloalkyl, haloalkoxy, optionally substituted alkyl, optionally substituted Ci-C6 alkoxy, optionally substituted C|-C6 alkoxyalkyl, optionally substituted C C6 alkoxyalkylaminoalkyl, C1-C6
alkylcarboxyheterocycloalkyl, oxy Ci-C6alkylheterocycloalkyl, optionally substituted aminoalkyl, optionally substituted C3-C7 cycloalkyl, optionally substituted aryl, optionally substituted aryl Ci-C6 alkyl, optionally substituted heterocycloalkyl, optionally substituted heterocycloalkylalkyl, optionally substituted heteroaryl or optionally substituted heteroarylalkyl;
each R9, when present, is independently halo, haloalkyl, haloalkoxy, optionally substituted C|-C6 alkyl, optionally substituted C|-C6 alkoxy, optionally substituted Ci-C6 alkoxyalkyl, optionally substituted Ci-C6 carboxyalkyl, optionally substituted alkoxycarbonyl, optionally substituted aminoalkyl, optionally substituted C3-C7 cycloalkyl, optionally substituted aryl, optionally substituted aryl Ci-C6 alkyl, optionally substituted aryloxy, optionally substituted heterocycloalkyl, or optionally substituted heteroaryl.
[0012] The compounds of formula I and II and Compound A are disclosed in WO
07/044813 and WO 08/124161, the entire contents of each of which are incorporated herein by reference.
[0013] In a third aspect, the invention is directed to a pharmaceutical composition which comprises a compound of Formula I or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier, excipient, or diluent.
[0014] In a fourth aspect, the invention comprises a method of inhibiting PI3K, comprising contacting a cell with a compound of Formula I or II or a pharmaceutically acceptable salt or solvate thereof, or with a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula I or II and a pharmaceutically acceptable carrier, excipient, or diluent.
[0015] In a fifth aspect of the invention is a method of inhibiting the in vivo activity of PDKa, the method comprising administering to a patient an effective PBKcc-inhibiting- inhibiting amount of a compound of Formula I or Π, or a pharmaceutically acceptable salt, solvate, or a pharmaceutical composition thereof.
[0016] In a sixth aspect, the Invention provides a method for treating a disease, disorder, or syndrome which method comprises administering to a patient a therapeutically effective amount of a compound of Formula I or II or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula I or II and a pharmaceutically acceptable carrier, excipient, or diluent.
[0017] A seventh aspect of the invention is directed to a process of preparing a compound of Formula I, comprising:
(a) reacting an intermediate of formula 7(a):
Figure imgf000007_0001
7(a)
where R6 is phenyl or heteroaryl each optionally substituted with I, 2, 3, 4, or 5 R9 groups (as defined in the Summary of the Invention) and R1 and R4 are as defined in the
Summary of the Invention; with an intermediate of formula R2NH2 (where R2 is as defined in the Summary of the Invention) to yield a Compound of Formula 1(a):
Figure imgf000007_0002
1(a);
(b) reacting an intermediate of formula 18:
Figure imgf000007_0003
18
where R1 and R4 are as defined in the Summary of the Invention; with tributyl-1- ethylvinyltin or with an intermediate of formula R6B(OH)2 where R6 is phenyl or heteroaryl each optionally substituted with 1, 2, 3, 4, or 5 R9 groups (as defined in the Summary of the Invention) to yield, respectively, a Compound of Formula 1(a) or 1(b):
Figure imgf000007_0004
1(b); or
(c) reacting an intermediate of formula 25(a):
Figure imgf000008_0001
25(a)
where R1 and R4 are as defined in the Summary of the Invention; with an intermediate of R2 H2 (where R2 is as defined in the Summary of the Invention) to yield a Compound of Formula 1(a); and
(d) optionally further resolving individual isomers; and
(e) optionally further modifying one of the R', R2, R , and R6 groups.
[0018] In an embodiment, Compound A is the Compound of Formula I in each of the above aspects.
[0019] An additional aspect relates to a method for treating a disease, disorder, or syndrome which method comprises administering to a patient a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula I or II or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula I, la, or II and a pharmaceutically acceptable carrier, excipient, or diluent, in combination with one or more agent. In an embodiment, Compound A is the Compound of Formula I in this aspect. The additional agent or agents are listed below:
Figure imgf000008_0002
Agent Name Source
CC-5013 (lenalidomide, Revlimid) Celgene Corporation ch 14.18 National Cancer Institute
Decitabine (5-aza-2'-deoxycytidine) Eisai Inc
E7389 (Halichondrin B Analog) Eisai Inc
EMD 121974 (Cilengitide) Merck gaA
Entinostat (MS-275, SNDX-275) Syndax
FK228 (Depsipeptide; Romidepsin) Celgene Corporation
Flavopiridol (alvocidib) Sanofi Aventis
GDC-0449 *** Genentech Inc
C-A12 Imclone
MDX-010 (MDX-CTLA4; Hybridoma- Medarex Inc
derived and Transfectoma-derived)
MK-2206 Merck and Company Inc
O-6-Benzylguanine NCI
Obatoclax mesylate (GX15-070MS) GeminX Pharmaceuticals
OSI-906 OSI Pharmaceuticals
Pertuzumab Genentech Inc
Rl 15777 (tipifarnib, Zarnestra) Johnson & Johnson
Reolysin Oncolytics Biotech Inc
RO4929097 Roche
SB-715992 (ispinesib) Cytokinetics
SCH727965 Schering-Plough
SJG-136 Spirogen
Thalidomide (Thalomid) Celgene Corporation
Triapine
UCN-01 Kyowa Hakko Kirin
VEGF-Trap (aflibercept) Sanofi Aventis
Vorinostat (suberoylanilide hydroxamic Merck and Company Inc acid; SAHA)
XK469R National Cancer Institute
17-AAG BMS
17-DMAG BMS
Alemtuzumab (Campath) Genzyme
AZD2171 (cediranib; Recentin™) AstraZeneca Pharmaceuticals
AZD6244 AstraZeneca Pharmaceuticals
BAY 43-9006 tosylate (BAY 54-9085; Bayer Corporation sorafenib tosylate)
Bevacizumab (rhuMAb VEGF, Avastin) Genentech Inc
BMS-354825 (dasatinib, Sprycel) Bristol-Myers Squibb
CCI-779 (temsirolimus, Torisel) Pfizer, inc
Erlotinib (OSI-774; Tarceva) OSI Pharmaceuticals
Gefitinib (ZD 1839, Iressa) AstraZeneca Pharmaceuticals
GM-CSF (sargramostim, Leukine) Genzyme
GW572016 (lapatinib) GlaxoSmithKline
GW786034 (pazopanib) GlaxoSmithKline
MLN 518 Millennium Pharmaceuticals
Oxaliplatin (Eloxatin) Sanofi Aventis
Perifosine Keryx Biopharmaceuticals Agent Name Source
Rituximab (MoAb C2B8 anti CD20, Biogen Idee
chimeric)
STI571 (imatinib, Gleevec) Novartis Pharmaceuticals
Corporation
Sunitinib malate (SU011248 L-malate; Pfizer Inc
Sutent)
Trastuzumab (Herceptin) Genentech Inc
[0020] More preferably, the additional agent or agents are selected from the following agents:
Figure imgf000010_0001
SB-715992 (ispinesib) Cytokinetics
SCH727965 Schering-Plough
SJG-136 Spirogen
Thalidomide (Thalomid) Celgene Corporation
Triapine
UCN-01 Kyowa Hakko Kirin
VEGF-Trap (aflibercept) Sanofi Aventis
Vorinostat (suberoylanilide hydroxamic acid; Merck and Company Inc
SAHA)
XK469R National Cancer Institute
[0021] More preferably, the additional agent or agents are selected from ABT-888, AZD6244, CCI-779, erlotinib, gefitinib, GW572016, GW786034, pertuzumab, or sunitinib.
[0022] An additional aspect relates to a method for treating a disease selected from anaplastic thyroid cancer, chronic lymphocytic leukemia, mantel cell lymphoma, non- Hodgkins lymphoma, glioblastoma multiforme, pediatric brain tumors, pontine glioma, HPV- related head and neck cancer, Hormone Receptive positve (HR+) breast cancer, triple negative breast cancer, and HER-2 overexpressing breast cancer, comprising adminstering a therapeutically effective amount of a compound of Formula I or II or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition comprising a
therapeutically effective amount of a compound of Formula I, la, or lit. In an embodiment, Compound A is the Compound of Formula I in this aspect.
[0023] An additional aspect relates to a method for treating a disease selected from anaplastic thyroid cancer, chronic lymphocytic leukemia, mantel cell lymphoma, non- Hodgkins lymphoma, glioblastoma multiforme, pediatric brain tumors, pontine glioma, HPV- related head and neck cancer, Hormone Receptive positve (HR+) breast cancer, triple negative breast cancer, and HER-2 overexpressing breast cancer, comprising adminstering a therapeutically effective amount of a compound of Formula I or Π or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition comprising a
therapeutically effective amount of a compound of Formula I, la, or II and a pharmaceutically acceptable carrier, excipient, or diluent, in combination with another agent. In an
embodiment, Compound A is the Compound of Formula I in this aspect. In another embodiment, Compound A is the Compound of Formula I and the additional agent or agents is selected from the group consisting of ABT-888, AZD6244, CCI-779, erlotinib, gefitinib, GW572016, GW786034, pertuzumab, and sunitinib.
[0024] An additional aspect relates to a capsule formulation of a compound of formula I. In an embodiment, Compound A is the Compound of Formula I in this aspect. The capsule formulation is one of the following formulations or an equivalent thereof. For example, the capsule formulation is a 90 mg "powder-in-capsule" (PiC) formulation qd or a 50 mg capsule
BID.
Figure imgf000012_0001
qsp, quantity sufficient for preparation.
DETAILED DESCRIPTION OF THE INVENTION
Abbreviations and Definitions
The following abbreviations and terms have the indicated meanings throughout:
Figure imgf000012_0002
Abbreviation Meaning
h or hr hour(s)
HPLC high pressure liquid chromatography
L liter(s)
M molar or molarity
m Multiplet
mg milligram(s)
MHz megahertz (frequency)
Min minute(s)
rriL milliliter(s)
μΐ, microliter(s)
μΜ Micromole(s) or micromolar
mM Millimolar
mmol millimole(s)
mol mole(s)
MS mass spectral analysis
N normal or normality
nM Nanomolar
NM nuclear magnetic resonance spectroscopy
q Quartet
RT Room temperature
s Singlet
t or tr Triplet
TFA trifluoroacetic acid
THF tetrahydrofuran
TLC thin layer chromatography
[0026] The symbol "-" means a single bond, "=" means a double bond, "= ' means a triple bond, " " means a single or double bond. The symbol "ΛΛΛΤ" refers to a group on a double-bond as occupying either position on the terminus of a double bond to which the symbol is attached; that is, the geometry, E- or Z-, of the double bond is ambiguous. When a group is depicted removed from its parent formula, the symbol will be used at the end of the bond which was theoretically cleaved in order to separate the group from its parent structural formula.
[0027] When chemical structures are depicted or described, unless explicitly stated otherwise, all carbons are assumed to have hydrogen substitution to conform to a valence of four. For example, in the structure on the left-hand side of the schematic below there are nine hydrogens implied. The nine hydrogens are depicted in the right-hand structure. Sometimes a particular atom in a structure is described in textual formula as having a hydrogen or hydrogens as substitution (expressly defined hydrogen), for example, -CH2CH2-. It is understood by one of ordinary skill in the art that the aforementioned descriptive techniques are common in the chemical arts to provide brevity and simplicity to description of otherwise complex structures.
Figure imgf000014_0001
a group "R" is depicted as "floating" on a ring system, as for example
Figure imgf000014_0002
then, unless otherwise defined, a substituent "R" may reside on any atom of the ring system, assuming replacement of a depicted, implied, or expressly defined hydrogen from one of the ring atoms, so long as a stable structure is formed.
[0029] If a group "R" is depicted as floating on a fused ring system, as for example in the formulae:
Figure imgf000014_0003
then, unless otherwise defined, a substituent "R" may reside on any atom of the fused ring system, assuming replacement of a depicted hydrogen (for example the -NH- in the formula above), implied hydrogen (for example as in the formula above, where the hydrogens are not shown but understood to be present), or expressly defined hydrogen (for example where in the formula above, "Z" equals =CH-) from one of the ring atoms, so long as a stable structure is formed. In the example depicted, the "R" group may reside on either the 5-membered or the 6-membered ring of the fused ring system. In the formula depicted above, when y is 2 for example, then the two "R's" may reside on any two atoms of the ring system, again assuming each replaces a depicted, implied, or expressly defined hydrogen on the ring.
[0030] When a group "R" is depicted as existing on a ring system containing saturated carbons, as for example in the formula:
Figure imgf000015_0001
where, in this example, "y" can be more than one, assuming each replaces a currently depicted, implied, or expressly defined hydrogen on the ring; then, unless otherwise defined, where the resulting structure is stable, two "R's" may reside on the same carbon. A simple example is when R is a methyl group; there can exist a geminal dimethyl on a carbon of the depicted ring (an "annular" carbon). In another example, two R's on the same carbon, including that carbon, may form a ring, thus creating a spirocyclic ring (a "spirocyclyl" group) structure with the depicted ring as for example in the formula:
Figure imgf000015_0002
[0031] "Acyl" means a -C(0)R radical where R is optionally substituted alkyl, optionally substituted alkenyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, heterocycloalkyl, or heterocycloalkylalkyl, as defined herein, e.g., acetyl,
trifluoromethylcarbonyl, or 2-methoxyethylcarbonyl, and the like.
[0032] "Acylamino" means a -NRR' radical where R is hydrogen, hydroxy, alkyl, or alkoxy and R' is acyl, as defined herein.
[0033] "Acyloxy" means an -OR radical where R is acyl, as defined herein, e.g.
cyanomethylcarbonyloxy, and the like.
[0034] "Administration" and variants thereof (e.g., "administering" a compound) in reference to a compound of the invention means introducing the compound or a prodrug of the compound into the system of the animal in need of treatment. When a compound of the invention or prodrug thereof is provided in combination with one or more other active agents (e.g., surgery, radiation, and chemotherapy, etc.), "administration" and its variants are each understood to include concurrent and sequential introduction of the compound or prodrug thereof and other agents.
[0035] "Alkenyl" means a means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to 6 carbon atoms which radical contains at least one double bond, e.g., ethenyl, propenyl, l-but-3-enyl, and l-pent-3-enyl, and the like.
[0036] "Alkoxy" means an -OR group where R is alkyl group as defined herein.
Examples include methoxy, ethoxy, propoxy, isopropoxy, and the like.
[0037] "Alkoxyalkyl" means an alkyl group, as defined herein, substituted with at least one, preferably one, two, or three, alkoxy groups as defined herein. Representative examples include methoxymethyl and the like.
[0038] "Alkoxyalkylamino" means an -NRR' group where R is hydrogen, alkyl, or alkoxyalkyl and R' is alkoxyalkyl, as defined herein.
[0039] "Alkoxyalkylaminoalkyl" means an alkyl group substituted with at least one, specifically one or two, alkoxyalkylamino group(s), as defined herein.
[0040] "Alkoxycarbonyl" means a -C(0)R group where R is alkoxy, as defined herein.
[0041] "Alkyl" means a linear saturated monovalent hydrocarbon radical of one to six carbon atoms or a branched saturated monovalent hydrocarbon radical of three to 6 carbon atoms, e.g., methyl, ethyl, propyl, 2-propyl, butyl (including all isomeric forms), or pentyl (including all isomeric forms), and the like.
[0042] "Alkylamino" means an -NHR group where R is alkyl, as defined herein.
[0043] "Alkylaminoalkyl" means an alkyl group substituted with one or two alkylamino groups, as defined herein.
[0044] "Alkylaminoalkyloxy" means an -OR group where R is alkylaminoalkyl, as defined herein.
[0045] "Alkylcarbonyl" means a -C(0)R group where R is alkyl, as defined herein.
[0046] "Alkynyl" means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to 6 carbon atoms which radical contains at least one triple bond, e.g., ethynyl, propynyl, butynyl, pentyN-2-yl and the like.
[0047] "Amino" means -NH2.
[0048] "Aminoalkyl" means an alkyl group substituted with at least one, specifically one, two or three, amino groups.
[0049] "Aminoalkyloxy" means an -OR group where R is aminoalkyl, as defined herein.
[0050] "Aryl" means a monovalent six- to fourteen-membered, mono- or bi-carbocyclic ring, wherein the monocyclic ring is aromatic and at least one of the rings in the bicyclic ring is aromatic. Unless stated otherwise, the valency of the group may be located on any atom of any ring within the radical, valency rules permitting. Representative examples include phenyl, naphthyl, and indanyl, and the like.
[0051] "Arylalkyl" means an alkyl radical, as defined herein, substituted with one or two aryl groups, as defined herein, e.g., benzyl and phenethyl, and the like.
[0052] "Aryloxy" means an -OR group where R is aryl, as defined herein.
[0053] "Carboxyalkyl" means an alkyl group, as defined herein, substituted with at least one, specifically one or two, -C(0)OH group(s).
[0054] "Cycloalkyl" means a monocyclic or fused bicyclic, saturated or partially unsaturated (but not aromatic), monovalent hydrocarbon radical of three to ten carbon ring atoms. Fused bicyclic hydrocarbon radical includes bridged ring systems. Unless stated otherwise, the valency of the group may be located on any atom of any ring within the radical, valency rules permitting. One or two ring carbon atoms may be replaced by a -C(O)- , -C(S)-, or -C(=NH)- group. More specifically, the term cycloalkyl includes, but is not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexyl, or cyclohex-3-enyl, and the like.
[0055] "Cycloalkylalkyl" means an alkyl group substituted with at least one, specifically one or two, cycloalkyl group(s) as defined herein.
[0056] "Dialkylamino" means a -NRR' radical where R and R' are alkyl as defined herein, or an N-oxide derivative, or a protected derivative thereof, e.g., dimethylamino, diethylamino, N,N-methylpropylamino or N,N-methylethylamino, and the like.
[0057] "Dialkylaminoalkyl" means an alkyl group substituted with one or two dialkylamino groups, as defined herein.
[0058] "Dialkylaminoalkyloxy" means an -OR group where R is dialkylaminoalkyl, as defined herein. Representative examples include 2-(N,N-diethylamino)-ethyloxy, and the like.
[0059] "Fused-polycyclic" or "fused ring system" means a polycyclic ring system that contains bridged or fused rings; that is, where two rings have more than one shared atom in their ring structures. In this application, fused-polycyclics and fused ring systems are not necessarily all aromatic ring systems. Typically, but not necessarily, fused-polycyclics share a vicinal set of atoms, for example naphthalene or 1,2,3,4-tetrahydro-naphthalene. A spiro ring system is not a fused-polycyclic by this definition, but fused polycyclic ring systems of the invention may themselves have spiro rings attached thereto via a single ring atom of the fused-polycyclic. In some examples, as appreciated by one of ordinary skill in the art, two adjacent groups on an aromatic system may be fused together to form a ring structure. The fused ring structure may contain heteroatoms and may be optionally substituted with one or more groups. It should additionally be noted that saturated carbons of such fused groups {i.e. saturated ring structures) can contain two substitution groups.
[0060] "Halogen" or "halo" refers to fluorine, chlorine, bromine or iodine.
[0061] "Haloalkoxy" means an -OR' group where R' is haloalkyl as defined herein, e.g., trifluoromethoxy or 2,2,2-trifluoroethoxy, and the like.
[0062] "Haloalkyl" mean an alkyl group substituted with one or more halogens, specifically one to five halo atoms, e.g., trifluoromethyl, 2-chloroethyl, and 2,2-difluoroethyl, and the like.
[0063] "Heteroaryl" means a monocyclic, fused bicyclic, or fused tricyclic, monovalent radical of 5 to 14 ring atoms containing one or more, specifically one, two, three, or four ring heteroatoms independently selected from -0-, -S(0)yy. (n is 0, 1, or 2), -N-, -N(RX)-, and the remaining ring atoms being carbon, wherein the ring comprising a monocyclic radical is aromatic and wherein at least one of the fused rings comprising a bicyclic or tricyclic radical is aromatic. One or two ring carbon atoms of any nonaromatic rings comprising a bicyclic or tricyclic radical may be replaced by a -C(O)-, -C(S)-, or -C(=NH)- group. Rx is hydrogen, alkyl, hydroxy, alkoxy, acyl, or alkylsulfonyl. Fused bicyclic radical includes bridged ring systems. Unless stated otherwise, the valency may be located on any atom of any ring of the heteroaryl group, valency rules permitting. When the point of valency is located on the nitrogen, Rx is absent. More specifically, the term heteroaryl includes, but is not limited to, 1,2,4-triazolyl, 1,3,5-triazolyl, phthalimidyl, pyridinyl, pyrrolyl, imidazolyl, thienyl, furanyl, indolyl, 2,3-dihydro-lH-indolyl (including, for example, 2,3-dihydro- lH-indol-2-yl or 2,3-dihydro-lH-indol-5-yl, and the like), isoindolyl, indolinyl, isoindolinyl, benzimidazolyl, benzodioxol-4-yl, benzofuranyl, cinnolinyl, indolizinyl, naphthyridiV-3-yl, phthalazi/V-3-yl, phthaIazi/V-4-yl, pteridinyl, purinyl, quinazolinyl, quinoxalinyl, tetrazoyl, pyrazolyl, pyrazinyl, pyrimidinyl, pyridazinyl, oxazolyl, isooxazolyl, oxadiazolyl, benzoxazolyl, quinolinyl, isoquinolinyl, tetrahydroisoquinolinyl (including, for example,
tetrahydroisoquinoli V-4-yl or tetrahydroisoquinoliN-6-yl, and the like), pyrrolo[3,2- c]pyridinyl (including, for example, pyrrolo[3,2-c]pyridiN-2-yl or pyrrolo[3,2-c]pyridiN-7-yl, and the like), benzopyranyl, thiazolyl, isothiazolyl, thiadiazolyl, benzothiazolyl,
benzothienyl, and the derivatives thereof, or N-oxide or a protected derivative thereof.
[0064] "Heteroarylalkyl" means an alkyl group, as defined herein, substituted with at least one, specifically one or two heteroaryl group(s), as defined herein.
[0065] "Heteroatom" refers to O, S, N, or P. [0066] "Heterocycloalkyl" means a saturated or partially unsaturated (but not aromatic) monovalent monocyclic group of 3 to 8 ring atoms or a saturated or partially unsaturated (but not aromatic) monovalent fused bicyclic group of 5 to 12 ring atoms in which one or more, specifically one, two, three, or four ring heteroatoms independently selected from O, S(0)„ (n is 0, 1, or 2), N, N(Ry) (where Ry is hydrogen, alkyl, hydroxy, alkoxy, acyl, or alkylsulfonyl), the remaining ring atoms being carbon. One or two ring carbon atoms may be replaced by a -C(O)-, -C(S)-, or -C(=NH)- group. Fused bicyclic radical includes bridged ring systems. Unless otherwise stated, the valency of the group may be located on any atom of any ring within the radical, valency rules permitting. When the point of valency is located on a nitrogen atom, Ry is absent. More specifically the term heterocycloalkyl includes, but is not limited to, azetidinyl, pyrrolidinyl, 2-oxopyrrolidinyl, 2,5-dihydro-lH-pyrrolyl, piperidinyl, 4-piperidonyl, morpholinyl, piperazinyl, 2-oxopiperazinyl, tetrahydropyranyl,
2-oxopiperidinyl, thiomorpholinyl, thiamorpholinyl, perhydroazepinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, dihydropyridinyl, tetrahydropyridinyl, oxazolinyl, oxazolidinyl, isoxazolidinyl, thiazolinyl, thiazolidinyl, quinuclidinyl, isothiazolidinyl, octahydroindolyl, octahydroisoindolyl, decahydroisoquinolyl, tetrahydrofuryl, and tetrahydropyranyl, and the derivatives thereof and N-oxide or a protected derivative thereof.
[0067] "Heterocycloalkylalkyl" means an alkyl radical, as defined herein, substituted with one or two heterocycloalkyl groups, as defined herein, e.g., morpholinylmethyl, N-pyrrolidinylethyl, and 3-(N-azetidinyl)propyl, and the like.
[0068] "Heterocycloalkylalkyloxy means an -OR group where R is heterocycloalkylalkyl, as defined herein.
[0069] "Saturated bridged ring system" refers to a bicyclic or polycyclic ring system that is not aromatic. Such a system may contain isolated or conjugated unsaturation, but not aromatic or heteroaromatic rings in its core structure (but may have aromatic substitution thereon). For example, hexahydro-furo[3,2-b]furan, 2,3,3a,4,7,7a-hexahydro-lH-indene, 7-aza-bicyclo[2.2.1]heptane, and l,2,3,4,4a,5,8,8a-octahydro-naphthalene are all included in the class "saturated bridged ring system.
[0070] "Spirocyclyl" or "spirocyclic ring" refers to a ring originating from a particular annular carbon of another ring. For example, as depicted below, a ring atom of a saturated bridged ring system (rings B and B'), but not a bridgehead atom, can be a shared atom between the saturated bridged ring system and a spirocyclyl (ring A) attached thereto. A spirocyclyl can be carbocyclic or heteroalicyclic.
Figure imgf000020_0001
[0071] "Optional" or "optionally" means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not. One of ordinary skill in the art would understand that with respect to any molecule described as containing one or more optional substituents, only sterically practical and/or synthetically feasible compounds are meant to be included. "Optionally substituted" refers to all subsequent modifiers in a term. So, for example, in the term "optionally substituted arylCi-g alkyl," optional substitution may occur on both the "C\.g alkyl" portion and the "aryl" portion of the molecule may or may not be substituted. A list of exemplary optional substitutions is presented below in the definition of "substituted."
[0072] "Optionally substituted alkoxy" means an -OR group where R is optionally substituted alkyl, as defined herein.
[0073] "Optionally substituted alkyl" means an alkyl radical, as defined herein, optionally substituted with one or more group(s), specifically one, two, three, four, or five groups, independently selected from alkylcarbonyl, alkenylcarbonyl, cycloalkylcarbonyl, alkylcarbonyloxy, alkenylcarbonyloxy, amino, alkylamino, dialkylamino, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, cyano, cyanoalkylaminocarbonyl, alkoxy, alkenyloxy, hydroxy, hydroxyalkoxy, halo, carboxy, alkylcarbonylamino, alkylcarbonyloxy, alkyl-S(0)o-2-, alkenyl-S(0)o-2-, aminosulfonyl, alkylaminosulfonyl, dialkylaminosulfonyl, alkylsulfonyl-NRc- (where Rc is hydrogen, alkyl, optionally substituted alkenyl, hydroxy, alkoxy, alkenyloxy, or cyanoalkyl), alkylaminocarbonyloxy, dialkylaminocarbonyloxy, alkylaminoalkyloxy, dialkylaminoalkyloxy, alkoxycarbonyl, alkenyloxycarbonyl, alkoxycarbonylamino, alkylaminocarbonylamino, dialkylaminocarbonylamino,
alkoxyalkyloxy, and -C(0)NRaRb (where Ra and R are independently hydrogen, alkyl, optionally substituted alkenyl, hydroxy, alkoxy, alkenyloxy, or cyanoalkyl).
[0074] "Optionally substituted alkenyl" means an alkyl radical, as defined herein, optionally substituted with one or more group(s), specifically one, two, three, four, or five groups, independently selected from alkylcarbonyl, alkenylcarbonyl, cycloalkylcarbonyl, alkylcarbonyloxy, alkenylcarbonyloxy, amino, alkylamino, dialkylamino, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, cyano, cyanoalkylaminocarbonyl, alkoxy, alkenyloxy, hydroxy, hydroxyalkoxy, halo, carboxy, alkylcarbonylamino, alkylcarbonyloxy, alkyl-S(0)o-2-> alkenyl-S(0)o-2-, aminosulfonyl, alkylaminosulfonyl, dialkylaminosulfonyl, alkylsulfonyl-NRc- (where Rc is hydrogen, alkyl, optionally substituted alkenyl, hydroxy, alkoxy, alkenyloxy, or cyanoalkyl), alkylaminocarbonyloxy, dialkylaminocarbonyloxy, alkylaminoalkyloxy, dialkylaminoalkyloxy, alkoxycarbonyl, alkenyloxycarbonyl, alkoxycarbonylamino, alkylaminocarbonylamino, dialkylaminocarbonylamino,
alkoxy alky loxy, and -C(0)NRaRb (where Ra and Rb are independently hydrogen, alkyl, optionally substituted alkenyl, hydroxy, alkoxy, alkenyloxy, or cyanoalkyl).
[0075] "Optionally substituted amino" refers to the group -N(H)R or -N(R)R where each R is independently selected from the group: optionally substituted alkyl, optionally substituted alkoxy, optionally substituted aryl, optionally substituted heterocycloalkyl, optionally substituted heteroaryl, acyl, carboxy, alkoxycarbonyl, -S(0)2-(optionally substituted alkyl), -S(0)2-optionally substituted aryl), -S(0)2-(optionally substituted heterocycloalkyl), -S(0)2-(optionally substituted heteroaryl), and -S(0)2-(optionally substituted heteroaryl). For example, "optionally substituted amino" includes diethylamino, methylsulfonylamino, and furanyl-oxy-sulfonamino.
[0076] "Optionally substituted aminoalkyl" means an alkyl group, as defined herein, substituted with at least one, specifically one or two, optionally substituted amino group(s), as defined herein.
[0077] "Optionally substituted aryl" means an aryl group, as defined herein, optionally substituted with one, two, or three substituents independently selected from acyl, acylamino, acyloxy, optionally substituted alkyl, optionally substituted alkenyl, alkoxy, alkenyloxy, halo, hydroxy, alkoxycarbonyl, alkenyloxycarbonyl, amino, alkylamino, dialkylamino, nitro, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, carboxy, cyano, alkylthio, alkylsulfinyl, alkylsulfonyl, aminosulfonyl, alkylaminosulfonyl, dialkylaminosulfonyl, alkylsulfonylamino, aminoalkoxy, or aryl is pentafluorophenyl. Within the optional substituents on "aryl", the alkyl and alkenyl, either alone or as part of another group
(including, for example, the alkyl in alkoxycarbonyl), are independently optionally substituted with one, two, three, four, or five halo.
[0078] "Optionally substituted arylalkyl" means an alkyl group, as defined herein, substituted with optionally substituted aryl, as defined herein.
[0079] "Optionally substituted cycloalkyl" means a cycloalkyl group, as defined herein, substituted with one, two, or three groups independently selected from acyl, acyloxy, acylamino, optionally substituted alkyl, optionally substituted alkenyl, alkoxy, alkenyloxy, alkoxycarbonyl, alkenyloxycarbonyl, alkylthio, alkylsulfinyl, alkylsulfonyl, aminosulfonyl, alkylaminosulfonyl, dialkylaminosulfonyl, alkylsulfonylamino, halo, hydroxy, amino, alkylamino, dialkylamino, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, nitro, alkoxyalkyloxy, aminoalkoxy, alkylaminoalkoxy, dialkylaminoalkoxy, carboxy, and cyano. Within the above optional substitutents on "cycloalkyl", the alkyl and alkenyl,, either alone or as part of another substituent on the cycloalkyl ring, are independently optionally substituted with one, two, three, four, or five halo, e.g. haloalkyl, haloalkoxy, haloalkenyloxy, or haloalkylsulfonyl.
[0080] "Optionally substituted cycloalkylalkyl" means an alkyl group substituted with at least one, specifically one or two, optionally substituted cycloalkyl groups, as defined herein.
[0081] "Optionally substituted heteroaryl" means a heteroaryl group optionally substituted with one, two, or three substituents independently selected from acyl, acylamino, acyloxy, optionally substituted alkyl, optionally substituted alkenyl, alkoxy, alkenyloxy, halo, hydroxy, alkoxycarbonyl, alkenyloxycarbonyl, amino, alkylamino, dialkylamino, nitro, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, carboxy, cyano, alkylthio, alkylsulfinyl, alkylsulfonyl, aminosulfonyl, alkylaminosulfonyl, dialkylaminosulfonyl, alkylsulfonylamino, aminoalkoxy, alkylaminoalkoxy, and dialkylaminoalkoxy. Within the optional substituents on "heteroaryl", the alkyl and alkenyl, either alone or as part of another group (including, for example, the alkyl in alkoxycarbonyl), are independently optionally substituted with one, two, three, four, or five halo.
[0082] "Optionally substituted heteroarylalkyl" means an alkyl group, as defined herein, substituted with at least one, specifically one or two, optionally substituted heteroaryl group(s), as defined herein.
[0083] "Optionally substituted heterocycloalkyl" means a heterocycloalkyl group, as defined herein, optionally substituted with one, two, or three substituents independently selected from acyl, acylamino, acyloxy, optionally substituted alkyl, optionally substituted alkenyl, alkoxy, alkenyloxy, halo, hydroxy, alkoxycarbonyl, alkenyloxycarbonyl, amino, alkylamino, dialkylamino, nitro, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, carboxy, cyano, alkylthio, alkylsulfinyl, alkylsulfonyl, aminosulfonyl, alkylaminosulfonyl, dialkylaminosulfonyl, alkylsulfonylamino, aminoalkoxy, or aryl is pentafluorophenyl. Within the optional substituents on "heterocycloalkyl", the alkyl and alkenyl, either alone or as part of another group (including, for example, the alkyl in alkoxycarbonyl), are independently optionally substituted with one, two, three, four, or five halo. [0084] "Optionally substituted heterocycloalkylalkyl" means an alkyl group, as defined herein, substituted with at least one, specifically one or two, optionally substituted heterocycloalkyl group(s) as defined herein.
[0085] "Yield" for each of the reactions described herein is expressed as a percentage of the theoretical yield.
[0086] "Patient" for the purposes of the present invention includes humans and other animals, particularly mammals, and other organisms. Thus the methods are applicable to both human therapy and veterinary applications. In a preferred embodiment the patient is a mammal, and in a most preferred embodiment the patient is human.
[0087] "Kinase-dependent diseases or conditions" refer to pathologic conditions that depend on the activity of one or more protein kinases. Kinases either directly or indirectly participate in the signal transduction pathways of a variety of cellular activities including proliferation, adhesion, migration, differentiation and invasion. Diseases associated with kinase activities include tumor growth, the pathologic neovascularization that supports solid tumor growth, and associated with other diseases where excessive local vascularization is involved such as ocular diseases (diabetic retinopathy, age-related macular degeneration, and the like) and inflammation (psoriasis, rheumatoid arthritis, and the like).
[0088] While not wishing to be bound to theory, phosphatases can also play a role in "kinase-dependent diseases or conditions" as cognates of kinases; that is, kinases
phosphorylate and phosphatases dephosphorylate, for example protein substrates. Therefore compounds of the invention, while modulating kinase activity as described herein, may also modulate, either directly or indirectly, phosphatase activity. This additional modulation, if present, may be synergistic (or not) to activity of compounds of the invention toward a related or otherwise interdependent kinase or kinase family. In any case, as stated previously, the compounds of the invention are useful for treating diseases characterized in part by abnormal levels of cell proliferation (i.e. tumor growth), programmed cell death (apoptosis), cell migration and invasion and angiogenesis associated with tumor growth.
[0089] "Compound A" refers to a compound having the structure
Figure imgf000023_0001
and is disclosed in WO 07/044813 and WO 08/124161, the entire contents of each of which are incorporated herein by reference. [0090] "Therapeutically effective amount" is an amount of a compound of the invention, that when administered to a patient, ameliorates a symptom of the disease. The amount of a compound of the invention which constitutes a "therapeutically effective amount" will vary depending on the compound, the disease state and its severity, the age of the patient to be treated, and the like. The therapeutically effective amount can be determined routinely by one of ordinary skill in the art having regard to their knowledge and to this disclosure.
[0091] "Cancer" refers to cellular-proliferative disease states as described hereinabove, and including but not limited to: Cardiac: sarcoma (angiosarcoma, fibrosarcoma,
rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma and teratoma; Lung: bronchogenic carcinoma (squamous cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hanlartoma, mesothelioma; Gastrointestinal: esophagus
(squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), stomach
(carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel (adenocarcinoma, lymphoma, carcinoid tumors, Karposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel (adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma); Genitourinary tract: kidney (adenocarcinoma, Wilms' tumor (nephroblastoma), lymphoma, leukemia), bladder and urethra (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma); Liver: hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, hemangioma; Bone: osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochronfroma (osteocartilaginous exostoses), benign chondroma,
chondroblastoma, chondromyxofibroma, osteoid osteoma and giant cell tumors; Nervous system: skull (osteoma, hemangioma, granuloma, xanthoma, osteitis defomians), meninges (meningioma, meningiosarcoma, gliomatosis), brain (astrocytoma, medulloblastoma, glioma, ependymoma, germinoma [pinealoma], glioblastoma multiform, oligodendroglioma, schwannoma, retinoblastoma, congenital tumors), spinal cord neurofibroma, meningioma, glioma, sarcoma); Gynecological: uterus (endometrial carcinoma), cervix (cervical carcinoma, pre-tumor cervical dysplasia), ovaries (ovarian carcinoma [serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma], granulosa- thecal cell tumors, SertoliLeydig cell tumors, dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, Fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal rhabdomyosarcoma], fallopian tubes (carcinoma); Hematologic: blood (myeloid leukemia [acute and chronic], acute lymphoblastic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplastic syndrome), HodgkiN's disease, non-Hodgkin's lymphoma [malignant lymphoma]; Skin: malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Karposi's sarcoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, psoriasis; and Adrenal glands: neuroblastoma. Thus, the term "cancerous cell" as provided herein, includes a cell afflicted by any one of the above- identified conditions.
[0092] A "pharmaceutically acceptable salt" of a compound means a salt that is pharmaceutically acceptable and that possesses the desired pharmacological activity of the parent compound. It is understood that the pharmaceutically acceptable salts are non-toxic. Additional information on suitable pharmaceutically acceptable salts can be found in
Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, PA, 1985, which is incorporated herein by reference or S. M. Berge, et al., "Pharmaceutical Salts," J. Pharm. Sci., 1977;66:1-19 both of which are incorporated herein by reference.
[0093] Examples of pharmaceutically acceptable acid addition salts include those formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; as well as organic acids such as acetic acid, trifluoroacetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, 3-(4-hydroxybenzoyl)benzoic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid,
2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid,
2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, glucoheptonic acid, 4,4'-methylenebis-(3-hydroxy-2-ene-l-carboxylic acid), 3-phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, p-toluenesulfonic acid, and salicylic acid and the like.
[0094] Examples of a pharmaceutically acceptable base addition salts include those formed when an acidic proton present in the parent compound is replaced by a metal ion, such as sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Preferable salts are the ammonium, potassium, sodium, calcium, and magnesium salts. Salts derived from pharmaceutically acceptable organic non-toxic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins. Examples of organic bases include isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine,
2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purines, piperazine, piperidine, N-ethylpiperidine, tromethamine, N-methylglucamine, polyamine resins, and the like. Exemplary organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline, and caffeine.
[0095] "Prodrug" refers to compounds that are transformed (typically rapidly) in vivo to yield the parent compound of the above formulae, for example, by hydrolysis in blood. Common examples include, but are not limited to, ester and amide forms of a compound having an active form bearing a carboxylic acid moiety. Examples of pharmaceutically acceptable esters of the compounds of this invention include, but are not limited to, alkyl esters (for example with between about one and about six carbons) the alkyl group is a straight or branched chain. Acceptable esters also include cycloalkyl esters and arylalkyl esters such as, but not limited to benzyl. Examples of pharmaceutically acceptable amides of the compounds of this invention include, but are not limited to, primary amides, and secondary and tertiary alkyl amides (for example with between about one and about six carbons). Amides and esters of the compounds of the present invention may be prepared according to conventional methods. A thorough discussion of prodrugs is provided in T. Higuchi and V. Stella, "Pro-drugs as Novel Delivery Systems," Vol 14 of the A.C.S.
Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated herein by reference for all purposes.
[0096] "Metabolite" refers to the break-down or end product of a compound or its salt produced by metabolism or biotransformation in the animal or human body; for example, biotransformation to a more polar molecule such as by oxidation, reduction, or hydrolysis, or to a conjugate (see Goodman and Gilman, "The Pharmacological Basis of Therapeutics" 8.sup.th Ed., Pergamon Press, Gilman et al. (eds), 1990 for a discussion of biotransformation). As used herein, the metabolite of a compound of the invention or its salt may be the biologically active form of the compound in the body. In one example, a prodrug may be used such that the biologically active form, a metabolite, is released in vivo. In another example, a biologically active metabolite is discovered serendipitously, that is, no prodrug design per se was undertaken. An assay for activity of a metabolite of a compound of the present invention is known to one of skill in the art in light of the present disclosure.
[0097] "Treating" or "treatment" of a disease, disorder, or syndrome, as used herein, includes (i) preventing the disease, disorder, or syndrome from occurring in a human, i.e. causing the clinical symptoms of the disease, disorder, or syndrome not to develop in an animal that may be exposed to or predisposed to the disease, disorder, or syndrome but does not yet experience or display symptoms of the disease, disorder, or syndrome; (ii) inhibiting the disease, disorder, or syndrome, i.e., arresting its development; and (iii) relieving the disease, disorder, or syndrome, i.e., causing regression of the disease, disorder, or syndrome. As is known in the art, adjustments for systemic versus localized delivery, age, body weight, general health, sex, diet, time of administration, drug interaction and the severity of the condition may be necessary, and will be ascertainable with routine experimentation by one of ordinary skill in the art.
[0098] The compounds disclosed herein also include all pharmaceutically acceptable isotopic variations, in which at least one atom is replaced by an atom having the same atomic number, but an atomic mass different from the atomic mass usually found in nature.
Examples of isotopes suitable for inclusion in the disclosed compounds include, without limitation, isotopes of hydrogen, such as 2H and 3H; isotopes of carbon, such as 13C and l4C; isotopes of nitrogen, such as 15N; isotopes of oxygen, such as l70 and I80; isotopes of phosphorus, such as 31 P and 32P; isotopes of sulfur, such as .sup.35S; isotopes of fluorine, such as 18F; and isotopes of chlorine, such as 36C1. Use of isotopic variations (e.g., deuterium, 2H) may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements. Additionally, certain isotopic variations of the disclosed compounds may incorporate a radioactive isotope (e.g., tritium, 3H, or l4C), which may be useful in drug and/or substrate tissue distribution studies.
Embodiments of the Invention
[0099] One embodiment (A) of a Compound of Formula I is where hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted cycloalkylalkyl, optionally substituted aryl, optionally substituted arylalkyl, optionally substituted
heterocycloalkyl, optionally substituted heterocycloalkylalkyl, optionally substituted heteroaryl or optionally substituted heteroarylalkyl. Specifically, R1 is hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted arylalkyl, or optionally substituted heterocycloalkylalkyl. More specifically, R1 is hydrogen, alkyl, alkyl substituted with one or two hydroxy, alkyl substituted with alkoxy, cycloalkyl, arylalkyl, or heterocycloalkylalkyl. Even more specifically, R1 is hydrogen, methyl, ethyl, propyl, isopropyl, 2-hydroxypropyl, 3-hydroxypropyl, 2-ethoxyethyl, 3-methoxypropyl,
3-ethoxypropyl, 3-isopropoxypropyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, benzyl, or 2-piperidin-l-ylethyl. Yet even more specifically, R1 is ethyl, isopropyl, cyclopentyl, or cyclohexyl. Yet even more specifically, R1 is ethyl.
[00100] Another embodiment (B) of a Compound of Formula I where R2 is hydrogen or alkyl where the alkyl is optionally substituted with 1 , 2, 3, 4, or 5 R8 groups. Specifically, R2 is hydrogen or alkyl where the alkyl is optionally substituted with one, two, or three R8 groups. More specifically, R2 is hydrogen or alkyl where the alkyl is optionally substituted with one, two, or three R8 groups; and each R8, when present, is independently selected from amino, alkylamino, dialkylamino, and halo. Even more specifically, R2 is hydrogen, methyl, ethyl, propyl, isopropyl, ferf-butyl, 3-aminopropyl, 3-(N-methylamino)-propyl, 3-(N,N- dimethylamino)-propyl, 2-fluoroethyl, or 2,2,2-trifluoroethyl. Yet even more specifically, R2 is hydrogen or ethyl. Yet even more preferably, R2 is hydrogen.
[00101] In another embodiment, R2 is hydrogen.
[00102] In another embodiment, R2 is alkyl optionally substituted with 1, 2, 3, 4, or 5, R8 groups. Specifically, R2 is alkyl where the alkyl is optionally substituted with one, two, or three R8 groups; and each R8, when present, is independently selected from amino, alkylamino, dialkylamino, and halo. Even more specifically, R2 is methyl, ethyl, propyl, isopropyl, tert-butyl, 3-aminopropyl, 3-(N-methylamino)-propyl, 3-(N,N-dimethylamino)- propyl, 2-fluoroethyl, or 2,2,2-trifluoroethyl. Yet even more specifically, R2 is ethyl.
[00103] Another embodiment (C) of a Compound of Formula I is where R4 is optionally substituted alkyl. Specifically, R4 is methyl or ethyl. More specifically, R4 is methyl.
[00104] Another embodiment (D) of a Compound of Formula I where R1 is where R6 is acyl. More specifically, R6 is alkylcarbonyl. Even more specifically, R6 is acetyl.
[00105] Another embodiment (E) of a Compound of Formula I is where R6 is phenyl optionally substituted with 1 , 2, 3, 4, or 5 R groups. Specifically, R is phenyl optionally substituted with one or two R9 groups; and each R9, when present, is independently selected from aryl, halo, alkoxy, aryloxy, and haloalkyl. More specifically, R6 is phenyl optionally substituted with one or two R9 groups; and each R9, when present, is independently selected from phenyl, fluoro, chloro, methoxy, phenyloxy, and trifluoromethyl. Even more specifically, R6 is phenyl, phenyl substituted with phenyl, fluorophenyl, difluorophenyl, chlorophenyl, dichlorophenyl, phenyl substituted with chloro and fluoro, methoxyphenyl, dimethoxyphenyl, phenyloxyphenyl, or trifluoromethylphenyl. Yet even more specifically, R6 is phenyl, 2-phenyl-phenyl, 3-phenyl-phenyl, 4-phenyl-phenyl, 2-fluorophenyl, 3- fluorophenyl, 4-fluorophenyl, 2,3-difluorophenyl, 2,4-difluorophenyl, 2,5-difluorophenyl, 2,6-difluorophenyl, 3,4-difluorophenyI, 3,5-difluorophenyl, 2-chlorophenyl, 3-chlorophenyl, 4-chlorophenyl, 2,3-dichlorophenyl, 2,4-dichlorophenyl, 2,5-dichlorophenyl,
2,6-dichlorophenyl, 3,4-dichlorophenyl, 3,5-dichlorophenyl, 3-chloro-4-fluoro-phenyl, 2-methoxyphenyl, 3-methoxyphenyl, 4-methoxyphenyl, 2,3 -dimethoxyphenyl,
2.4- dimethoxyphenyI, 2,5-dimethoxyphenyl, 2,6-dimethoxyphenyl, 3,4-dimethoxyphenyl,
3.5- dimethoxyphenyl, 4-phenyloxyphenyl, 2-trifluoromethylphenyl, 3-trifluoromethylphenyl, or 4-trifluoromethylphenyl.
[00106] Another embodiment (F) of a Compound of Formula I is where R6 is phenyl substituted with 1, 2, 3, 4, or 5 R9 groups.
[00107] Another embodiment (G) of a Compound of Formula I is where R6 is heteroaryl optionally substituted with 1, 2, 3, 4, or 5 R9 groups.
[00108] A more specific embodiment (Gl) of embodiment G is a Compound of Formula I where R6 is a 6-membered heteroaryl optionally substituted with one or two R9. More specifically, R6 is pyridinyl, pyrazinyl, pyrimidinyl, or pyridazinyl each of which is optionally substituted with one R9 where R9, when present, is halo. Even more specifically, R6 is pyridiN-2-yl, pyridiN-3-yl, pyridiN-4-yl, 3-fluoropyridi/v'-4-yl, pyrazin-2-yl, pyrazin-3- yl, pyrimidin-2-yl, pyrimidin-4-yl, pyrimidin-5-yl, pyridazin-3-yl, or pyridazin-4-yl, each of which is optionally substituted with one or two R9.
[00109] In an even more specific embodiment (G2) of embodiment G is a Compound of Formula I where R6 is pyrazinyl, pyrimidinyl, or pyridazinyl each of which is optionally substituted with one R9 where R9, when present, is halo. Even more specifically, R6 is pyrazin-2-yl, pyrazin-3-yl, pyrimidin-2-yl, pyrimidin-4-yl, pyrimidin-5-yl, pyridazin-3-yl, or pyridazin-4-yl.
[00110] A more specific embodiment (G3) of embodiment G is a Compound of Formula I where R6 is 5-membered heteroaryl optionally substituted with one or two R9. Specifically R6 is pyrazolyl, imidazolyl, thienyl, thiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, furanyl, pyrrolyl, triazolyl, or tetrazolyl, each of which is optionally substituted with one R9 where R9, when present, is alkyl, arylalkyl, cyano, aryl, alkoxycarbonyl, or halo. More specifically, R6 is pyrazol-l-yl, pyrazol-3-yl, pyrazol-4-yl, pyrazol-5-yl, imidazol-l-yl, imidazoI-2-yl, imidazol-4-yl, imidazol-5-yl, thien-2-yl, thien-3-yl, thiazol-2-yl, thiazol-4-yl, thiazol-5-yl, oxazol-2-yl, oxazol-4-yl, oxazol-5-yl, isoxazol-3-yl, isoxazol-4-yl, isoxazol-5-yl,
1.2.3- oxadiazol-4-yl, l,2,3-oxadiazol-5-yl, l,3,4-oxadiazol-2-yl, l,2,4-oxadiazol-3-yl,
1.2.4- oxadiazol-5-yl, furan-2-yl, furan-3-yl, pyrrol- 1 -yl, pyrrol-2-yl, pyrrol-3-yl, triazol- l-yl, triazol-4-yl, triazol-5-yl, tetrazol-l-yl, or tetrazol-5-yl; each of which is optionally substituted with one R9 where R9, when present, is methyl, benzyl, cyano, phenyl, N-terf-butoxycarbonyl, or chloro. Even more specifically, R6 is pyrazol-3-yl, pyrazol-4-yl, pyrazol-5-yl, imidazol-2-yl, imidazol-4-yl, imidazol-5-yl, thien-2-yl, thien-3-yl, thiazol-2-yI, thiazol-4-yl, thiazol-5-yl, oxazol-2-yl, oxazol-4-yl, oxazol-5-yl, isoxazol-3-yl, isoxazol-4-yl, isoxazol-5-yl, l,2,3-oxadiazol-4-yl, 1 ,2,3-oxadiazol-5-yl, l,3,4-oxadiazol-2-yl, l,2,4-oxadiazol-3-yl, l ,2,4-oxadiazol-5-yl, furan-2-yl, furan-3-yl, pyrrol-2-yl, pyrrol-3-yl, (riazol-4-yl, triazol-5-yl, or tetrazol-5-yl; each of which is optionally substituted with one R9 where R9, when present, is methyl, benzyl, cyano, phenyl, N-teri-butoxycarbonyl, or chloro.
[00111] A more specific embodiment (G4) of embodiment G is a Compound of Formula I where R6 is thienyl, pyrrolyl, furanyl, pyrazolyl, thiazolyl, isoxazolyl, imidazolyl, triazolyl, or tetrazolyl, each of which is optionally substituted with one R9 where R9, when present, is methyl, benzyl, cyano, phenyl, N-rerr-butoxycarbonyl, or chloro. Specifically, R6 is thien-2- yl, thien-3-yl, pyrrol-2-yl, furan-2-yl, furan-3-yl, pyrazoI-3-yl, pyrazol-4-yl, pyrazoI-5-yl, thiazol-2-yl, thiazol-5-yl, isoxazol-4-yl, imidazol-5-yl, triazol-5-yI, tetrazol-5-yl, each of which is optionally substituted with one R9 where R9, when present, is methyl, benzyl, cyano, phenyl, N-rerr-butoxycarbonyl, or chloro. More specifically, R6 is thien-2-yl, thien-3-yl, 5-cyano-thien-2-yl, 4-methyl-thien-2-yl, 4-methyl-thien-3-yl, 5-chloro-thien-5-yl, 5-phenyl- thien-2-yl, pyrrol-2-yl, N-iert-butoxycarbonyl-pyrrol-2-yl, N-methyl-pyrrol-2-yl, furan-2-yl, furan-3-yl, pyrazol-3-yl, pyrazol-4-yl, N-benzyl-pyrazol-4-yl, pyrazol-5-yl, thiazol-2-yl, thiazol-5-yl, isoxazol-4-yl, imidazol-5-yl, triazol-5-yl, tetrazol-5-yl,
[00112] A more specific embodiment (G5) of embodiment G is a Compound of Formula I where R6 is thien-2-yl, thien-3-yl, pyrrol-2-yl, furan-2-yl, furan-3-yl, pyrazol-3-yl, pyrazol-4- yl, pyrazol-5-yl, thiazol-2-yl, thiazol-5-yl, isoxazol-4-yl, imidazol-5-yl, triazol-5-yl, or tetrazoI-5-yl, each of which is optionally substituted with one R9 where R9, when present, is methyl, benzyl, cyano, phenyl, N-terf-butoxycarbonyl, or chloro.
[00113] A more specific embodiment (G6) of embodiment G is a Compound of Formula I where R6 is indolyl, benzimidazolyl, benzofuranyl, benzoxazolyl, or benzoisoxazolyl each of which is optionally substituted with 1, 2, 3, 4, or 5 R9 groups. Specifically, R6 is indol-2-yl, indol-3-yl, indol-4-yl, indol-5-yl, indol-6-yl, indol-7-yl, benzimidazol-2-yl, benzimidazol-4- yl, benzimidazol-5-yl, benzimidazol-6-yl, benzimidazol-7-yl, benzofuran-2-yl, benzofuran-3- yl, benzofuran-4-yl, benzofuran-5-yl, benzofuran-6-yl, benzofuran-7-yl, benzoxazol-2-yl, benzoxazol-4-yl, benzoxazol-5-yl, benzoxazol-6-yl, benzoxazol-7-yl, benzoisoxazol-3-yI, benzoisoxazol-4-yl, benzoisoxazol-5-yl, benzoisoxazol-6-yl, or benzoisoxazol-7-yl; each of which is optionally substituted with 1, 2, 3, 4, or 5 R9 groups. More specifically, R6 is indol- 6-yl.
[00114] Another embodiment (H) is a Compound of Formula 1 is where R1 is hydrogen, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkylalkyl, or optionally substituted arylalkyl; X is -NH-; R2 is hydrogen or alkyl where the alkyl is optionally substituted with one or two R8 groups; R4 is alkyl; R5 is hydrogen; R6 is phenyl or heteroaryl wherein the phenyl and heteroaryl are optionally substituted with one, two, or three R groups; each R , when present, is independently amino, alkylamino, dialkylamino, or halo; and each R9, when present, is independently alkyl, arylalkyl, cyano, aryl, alkoxycarbonyl, or halo.
[00115] Another embodiment (J) is a Compound of Formula 1 is where R6 is pyrazol-3-yl, pyrazol-4-yl, pyrazol-5-yl, imidazol-2-yl, imidazol-4-yl, imidazol-5-yl, thien-2-yl, thien-3-yl, thiazol-2-yl, thiazoI-4-yl, thiazol-5-yl, oxazol-2-yl, oxazol-4-yl, oxazol-5-yl, isoxazol-3-yl, isoxazol-4-yl, isoxazol-5-yl, l,2,3-oxadiazol-4-yl, l,2,3-oxadiazol-5-yl, l,3,4-oxadiazol-2-yl, 1 ,2,4-oxadiazol-3-yl, l,2,4-oxadiazol-5-yl, furan-2-yl, furan-3-yl, pyrrol-2-yl, pyrrol-3-yl, triazol-4-yl, triazol-5-yl, or tetrazol-5-yl; each of which is optionally substituted with 1, 2, 3, 4, or 5 R9 groups.
[00116] Another embodiment (K) is a Compound of Formula I is where R1 is alkyl or cycloalkyl; R4 is methyl; and R6 is heteroaryl optionally substituted with one or two R9 groups. Specifically, each R9, when present, is independently alkyl, arylalkyl, cyano, aryl, alkoxycarbonyl, or halo. Specifically, R6 is pyrazol-3-yI, pyrazol-4-yl, pyrazol-5-yl, imidazol-2-yl, imidazol-4-yl, imidazol-5-yl, thien-2-yl, thien-3-yl, thiazol-2-yl, thiazol-4-yl, thiazol-5-yl, oxazol-2-yl, oxazol-4-yl, oxazol-5-yl, isoxazol-3-yl, isoxazol-4-yl, isoxazol-5-yl,
1.2.3- oxadiazol-4-yl, 1 ,2,3-oxadiazol-5-yl, l,3,4-oxadiazol-2-yl, l,2,4-oxadiazol-3-yl,
1.2.4- oxadiazol-5-yl, furan-2-yl, furan-3-yl, pyrrol-2-yl, pyrrol-3-yl, triazol-4-yl, triazol-5-yl, or tetrazol-5-yl; each of which is optionally substituted with one R9 where R9, when present, is methyl, benzyl, cyano, phenyl, or N-teri-butoxycarbonyl.
[00117] A more specific embodiment (Kl) of embodiment K is a Compound of Formula I where R2 is hydrogen. [00118] A more specific embodiment (K2) of embodiment K is a Compound of Formula I where R2 is methyl or ethyl.
[00119] Another embodiment (L) of a Compound of Formula I is where R1 is alkyl or cycloalkyl; R4 is methyl; and R6 is phenyl optionally substituted with one or two R9 groups. Specifically each R9, when present, is independently halo, alkoxy, or haloalkyl.
[00120] Another embodiment (M) of a Compound of Formula I is where R1 is alkyl or cycloalkyl; R4 is methyl; and R2 is hydrogen.
[00121] Another embodiment (N) of a Compound of Formula I is where R1 is alkyl or cycloalkyl; R4 is methyl; and R2 is optionally substituted alkyl.
Compounds of Formula IA
[00122] In another embodiment, the compound of formula I is a compound of formula IA.
Figure imgf000032_0001
IA
or a pharmaceutically acceptable salt thereof, wherein:
R1 is alkyl, cycloalkyl, cycloalkylalkyl, aryl, arylalkyl, heterocycloalkyl,
heterocycloalkylalkyl, heteroaryl, or heteroarylalkyl;
R2 is hydrogen or alkyl;
R4 is alkyl;
R5 is hydrogen;
R6 is phenyl, acyl, or heteroaryl wherein the phenyl and heteroaryl are is optionally substituted with 1, 2, 3, 4, or 5 R9 groups; and
each R9, when present, is independently halo, alkyl, haloalkyl, alkoxy, haloalkoxy, cyano, amino, alkylamino, dialkylamino, alkoxyalkyl, carboxyalkyl, alkoxycarbonyl, aminoalkyl, cycloalkyl, aryl, arylalkyl, aryloxy, heterocycloalkyl, or heteroaryl and where the cycloalkyl, aryl, heterocycloalkyl, and heteroaryl, each either alone or as part of another group within R9, are independently optionally substituted with 1, 2, 3, or 4 groups selected from halo, alkyl, haloalkyl, hydroxy, alkoxy, haloalkoxy, amino, alkylamino, and
dialkylamino.
[00123] In one embodiment, R1 is alkyl, cycloalkyl, heterocycloalkylalkyl, or arylalkyl; X is -NH-; R2 is hydrogen or alkyl; R4 is alkyl; R5 is hydrogen; R6 is phenyl or heteroaryl wherein the phenyl and heteroaryl are is_optionally substituted with one, two, or three R groups; each R8, when present, is independently amino, alkylamino, dialkylamino, or halo; and each R8, when present, is independently alkyl, arylalkyl, cyano, aryl, or alkoxycarbonyl.
[00124] In another embodiment, R4 is methyl.
[00125] In another embodiment, R1 is alkyl, cycloalkyl, or heterocycloalkyl.
[00126] In another embodiment, R1 is alkyl.
[00127] In another embodiment, R6 is heteroaryl optionally substituted with 1 , 2, or 3 R9 groups.
[00128] In another embodiment, each R9, when present, is independently alkyl, arylalkyl, cyano, aryl, alkoxycarbonyl, or halo.
[00129] In another embodiment, R6 is pyrazolyl, imidazolyl, thienyl, thiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, furanyl, pyrrolyl, triazolyl, or tetrazolyl; each of which is optionally substituted with 1, 2, or 3 R9 groups.
[00130] In another embodiment, R6 is pyrazol-3-yl, pyrazol-4-yl, pyrazol-5-yl, imidazol-2- yl, imidazol-4-yl, imidazol-5-yl, thien-2-yl, thien-3-yl, thiazol-2-yl, thiazol-4-yl, thiazol-5-yl, oxazol-2-yl, oxazol-4-yl, oxazol-5-yl, isoxazol-3-yl, isoxazol-4-yl, isoxazol-5-yl, 1,2,3- oxadiazol-4-yl, 1 ,2,3-oxadiazol-5-yl, l,3,4-oxadiazol-2-yl, l ,2,4-oxadiazol-3-yl,
l,2,4-oxadiazol-5-yl, furan-2-yl, furan-3-yl, pyrrol-2-yI, pyrrol-3-yI, triazol-4-yl, triazol-5-yl, or tetrazol-5-yI; each of which is optionally substituted with 1, 2, or 3 R9 groups.
[00131] In another embodiment, R6 is pyrazinyl, pyrimidinyl, or pyridazinyl each of which is optionally substituted with 1, 2, or 3 R9 groups and R4 is methyl.
[00132] In another embodiment, R2 is hydrogen, R4 is methyl, R1 is optionally substituted alkyl, cycloalkyl, or heterocycloalkyl, and R6 is heteroaryl optionally substituted with 1, 2, or 3 R9 groups.
[00133] In another embodiment, the compound of formula III is selected from:
8-ethyl-2-(ethylamino)-4-methyl-6-phenylpyrido[2,3-d]pyrimidin-7(8H)-one;
6-biphenyl-4-yl-8-emyl-2-(emylamino)-4-methylpyrido[2,3-d]pyrimidin-7(8H)-one;
6-(2,4-difluorophenyl)-8-ethyl-2-(ethylamino)-4-methylpyrido[2,3-d]pyrimidin-7(8H)-one; 6-(3-chloro-4-fluorophenyl)-8-ethyl-2-(ethylamino)-4-methylpyrido[2,3-d]pyrimidin- 7(8H)-one;
8-ethyl-2-(ethylamino)-4-methyI-6-[4-(methyloxy)phenyl]pyrido[2,3-d]pyrimidin-7(8H)- one;
6-(2,4-dichlorophenyl)-8-emyl-2-(emylamino)-4-methylpyrido[2,3-d]pyrimidin-7(8H)- one;
6-(3,4-difluorophenyl)-8-ethyl-2-(ethylamino)-4-methylpyrido[2,3-d]pyrimidin-7(8H)-one;
8-ethyl-2-(ethylamino)-4-methyl-6-[2-(methyloxy)phenyl]pyrido[2,3-d]pyrimidin-7(8H)- one;
8-ethyl-2-(ethylamino)-4-methyl-6-[4-(phenyloxy)phenyl]pyrido[2,3-d]pyrimidin-7(8H)- one;
6-[2,4-bis(rnethyloxy)phenyl]-8-ethyl-2-(ethylamino)-4-methylpyrido[2,3-d]pyrimidin- 7(8H)-one;
8-ethyl-2-(e^ylamino)-6-(3-fluorophenyl)-4-methylpyrido[2,3-d]pyrimidin-7(8/¾-one; 8-ethyl-2-(ethylamino)-6-(2-fluorophenyl)-4-methylpyrido[2,3-d]pyrirnidin-7(8H)-one; 8-ethyl-2-(ethylamino)-4-methyl-6-[3-(trifluoromethyl)phenyl]pyrido[2,3-d]pyrimidin- 7(8H)-one;
8-ethyl-2-(ethylamino)-6-(4-fluorophenyl)-4-methyIpyrido[2,3-d]pyrimidin-7(8H)-one; 8-ethyl-2-(ethylamino)-4-methyl-6-(2-thienyl)pyrido[2,3-d]pyrimidin-7(8H)-one;
8-ethyl-2-(ethylamino)-4-methyl-6-[3-(methyloxy)phenyl]pyrido[2,3-d]pyrimidin-7(8H)- one;
6-(3-chlorophenyl)-8-ethyl-2-(ethylamino)-4-methylpyrido[2,3-d]pyrimidin-7(8H)-one;
6-(4-chlorophenyl)-8-ethyl-2-(ethylamino)-4-rnethylpyrido[2,3-d]pyrimidin-7(8H)-one;
8-ethyl-2-(ethylamino)-4-methyl-6-(3-thienyl)pyrido[2,3-d]pyrirnidin-7(8H)-one;
8-ethyl-2-(ediylamino)-4-methyl-6-(4-methyl-2-thienyI)pyrido[2,3-d]pyriniidin-7(8H)-one;
8-ethyl-2-(ethylamino)-4-methyl-6-(4-methyl-3-thienyl)pyrido[2,3-d]pyrimidin-7(8H)-one;
1,1-dimethylethyl 2-[8-ethyl-2-(ethylamino)-4-methyl-7-oxo-7,8-dihydropyrido[2,3- d]pyrimidin-6-yl]- lH-pyrrole- 1 -carboxylatei
5- ethyl- -iethylamino^-methyl-e-ilH-pyrrol^-y pyridoP^-dlpyrimidin^iSHi-one;
6- (5-chloro-2-thienyl)-8-ethyl-2-(ethyIamino)-4-methylpyrido[2,3-d]pyrimidin-7(8H)-one; 8-ethyl-2-(e^ylamino)-4-methyl-6-pyrimidin-5-ylpyrido[2,3-d]pyrimidin-7(8H)-one; 8-ethyl-2-(e^ylamino)-6-(3-fluoropyridn-4-yl)-4-methylpyrido[2,3-d]pyrimidin-7(8H)^ one;
8-ethyl-2-(ethylamino)-6-furan-3-yl-4-methylpyrido[2,3-d]pyrimidin-7(8H)-one;
8-ethyl-2-(ethylamino)-4-methyl-6-[ 1 -(phenylmethyl)- lH-pyrazol-4-yl]pyrido[2,3- d]pyrimidin-7(8H)-one;
2-(ethyiamino)-4-methyl-8-(l-methylethyl)-6-(2-thienyl)pyrido[2,3-d]pyrimidin-7(8H)- one; 8-ethyl-2-(ethylamino)-6-(lH-indol-6-yl)-4-methylpyrido[2,3-d]pyrimidin-7(8H)-one; 8-ethyl-2-(ethylamino)-4-methyl-6-(5-phenyl-2-thienyl)pyrido[2,3-d]pyrimidin-7(8H)-one; 2-(ethylamino)-6-furan-3-yl-4-methyl-8-(l-methylethyl)pyrido[2,3-d]pyrimidin-7(8H)- one;
8-ethyl-2-(ethylamino)-4-methyl-6-(lH-pyrazol-5-yl)pyrido[2,3-d]pyrimidin-7(8H)-one; 8-cyclohexyl-2-(ethylamino)-4-methyl-6-(2-thienyl)pyrido[2,3-d]pyrimidin-7(8H)-one; 8-e l-2-(ethylamino)-4-methyl-6-(l,3-thiazol-2-yl)pyrido[2,3-d]pyrimidin-7(8H)-one; 8-cyclopentyl-2-(ethylamino)-4-methy l-6-( 1 H-pyrazol-3-y l)pyrido[2,3-d] pyrimidin-7(8H)- one;
2-(ethylamino)-4-methyl-8-(l-methylethyl)-6-(lH-pyrazol-5-yl)pyrido[2,3-d]pyrimidin- 7(8H)-one;
8-ethyl-2-(ethylamino)-4-methyl-6-( lH-pyrazol- 1 -yl)pyrido[2,3-d]pyrimidin-7(8H)-one;
2-(ethylaminoM-methyl-8-(l-methylethyl)-6-0
7(8H)-one;
8-cyclopentyl-2-(ethylamino)-4-methyl-6-(lH-pyrazol-l-yl)pyrido[2,3-d]pyrimidin-7(8H)- one;
8-ethyl-4-methyl-6-(lH-pyrazol-5-yl)-2-[(2,2,2-trifluoroethyl)amino]pyrido[2,3- d] py rimidin-7(8H)-one ;
2-amino-8-ethyl-4-methyl-6-(lH-pyrazol-5-yl)pyrido[2,3-ii|pyrimidin-7(8H)-one;
2-(ethylamino)-4-methyl-6-(lH-pyrazol-3-yl)pyrido[2,3-i ]pyrimidin-7(8H)-one;
8-ethyl-4-methyl-2-(methylamino)-6-(lH-pyrazol-5-yl)pyrido[2,3-iflpyrimidin-7(8H)-one;
2-amino-8-cyclopentyl-4-methyl-6-(lH-pyrazol-3-yl)pyrido[2,3-^pyrimidin-7(8H)-one;
8-ethyl-2-[(2-fluoroethyl)amino]-4-methyl-6-(lH-pyrazol-5-yl)pyrido[2,3-^pyrimidin-
7(8H)-one;
2-amino-4-methyl-8-(l-me letoyl)-6-(lH-pyrazoW
one;
2-amino-4-methyl-8-(phenylmethyl)-6-(lH-pyrazol-3-yl)pyrido[2,3-i/]pyrimidin-7(8H)- one;
2-amino-8-ethyl-4-metoyl-6-(4-methyl-3-thtenyl)pyrido[2,^^
2-amino-8-ethyl-4-methyl-6-(2-thienyI)pyrido[2,3-i ]pyrimidin-7(8H)-one;
2-amino-8-ethyl-6-(4-fluorophenyl)-4-methylpyrido[2,3-i ]pyrimidin-7(8H)-one;
2-amino-8-ethyl-6-(3-fluorophenyl)-4-methylpyrido[2,3-iilpyrimidin-7(8H)-one;
2-amino-8-ethyl-6-(2-fluorophenyl)-4-methylpyrido[2,3-^]pyrimidin-7(8H)-one; 2-amino-8-ethyl-4-methyl-6-(3-thienyl)pyrido[2,3-^pyrimidin-7(8H)-one;
2-amino-8-ethyl-6-furan-3-yl-4-methylpyrido[2,3-i ]pyrimidin-7(8H)-one;
2-amino-8-ethyl-4-methyl-6-phenylpyrido[2,3-ii|pyrimidin-7(8H)-one;
2-amino-8-ethyl-4-me l-6-[4-(methyloxy)phenyl]pyrido[2,3-ii]pyrimidin-7(8H)-one;
2-amino-6-(4-chlorophenyl)-8-e l-4-methylpyrido[2,3-iilpyrirnidin-7(8H)-one;
2-amino-6-(3-chlorophenyI)-8-ethyl-4-methylpyrido[2,3- !pyrimidin-7(8H)-one;
2-amino-8-ethyl-6-isoxazol-4-yl-4-methylpyrido[2,3-iilpyrimidin-7(8H)-one;
2-amino-8-ethyl-6-furan-2-yl-4-methylpyrido[2,3-i ]pyriniidin-7(8H)-one;
2-amino-6-(2,4-dichlorophenyl)-8-ethyl-4-methylpyrido[2,3-iflpyrimidin-7(8H)-one;
5-(2-amino-8-ethyl-4-methyl-7-oxo-7,8-dihydropyrido[2,3-^pyrimidin-6-yl)thiophene-2- carbonitrile;
2-amino-8-ethyl-4-methyl-6-pyrimidin-5-ylpyrido[2,3-^pyrimidin-7(8H)-one;
2-amino-8-ethyl-6-(lH-imidazol-5-yl)-4-methylpyrido[2,3-if]pyrimidin-7(8H)-one;
2-amino-8-ethyl-4-methyl-6-( 1H- 1 ,2,3-triazol-5-yl)pyrido[2,3-i/]pyrimidin-7(8H)-one; 2-amino-8-ethyl-4-me^yl-6-(lH-pyrazol-4-yl)pyrido[2,3-ii|pyrimidin-7(8H)-one;
2-amino-8-ethyl-4-methyl-6-(l,3-tniazol-2-yl)pyrido[2,3-iflpyrimidin-7(8H)-one;
2-amino-8-ethyl-4-methyl-6-(lH-tetrazol-5-yl)pyrido[2,3-ii]pyrimidin-7(8H)-one;
2-amino-8-ethyl-4-methyl-6-(l-memyl-lH-pyrrol-2-yl)pyrido[2,3-^pyrimidin-7(8H)-one; 2-amino-4,8-diethyl-6-(lH-pyrazol-5-yl)pyrido[2,3-i ]pyrimidin-7(8H)-one; and
2-amino-8-cyclopentyl-4-methy l-6-( 1 ,3-thiazol-5-y l)pyrido[2,3-i/] pyrimidin-7(8H)-one.
[00134] In another embodiment, the compound of formula ΠΙ is selected from:
8-ethyl-2-(emylamino)-4-methyl-6-(2-thienyl)pyrido[2,3-d]pyrimidin-7(8H)-one;
8-ethyl-2-(ethylamino)-4-methyl-6-(3-thienyl)pyrido[2,3-d]pyrimidin-7(8H)-one;
8-ethyl-2-(ethylamino)-4-methyl-6-(4-methyl-2-thienyl)pyrido[2,3-d]pyrimidin-7(8H)-one; 8-ethyl-2-(e lamino)-4-methyl-6-(4-methyl-3-thienyl)pyrido[2,3-d]pyrimidin-7(8H)-one; 1,1-dimethylethyl 2-[8-ethyl-2-(ethylamino)-4-methyl-7-oxo-7,8-dihydropyrido[2,3- d]pyrimidin-6-yl]- lH-pyrrole- 1 -carboxylate
8-et yl-2-(emylamino)-4-methyl-6-(lH-pyn-ol-2-yl)pyrido[2,3-d]pyrimidin-7(8H)-one;
6-(5-chloro-2-thienyl)-8-ethyl-2-(ethylamino)-4-methylpyrido[2,3-d]pyrimidin-7(8H)-one;
8-ethyl-2-(ethylamino)-4-methyl-6-pyrimidin-5-ylpyrido[2,3-d]pyrimidin-7(8H)-one;
8-etJiyl-2-(ethylamino)-6-(3-fluoropyridn-4-yl)-4-methylpyrido[2,3-d]pyrimidin-7(8H)- one; 8-ethyl-2-(ethylamino)-6-furan-3-yl-4-methylpyrido[2,3-d]pyrimidin-7(8H)-one;
2-(ethylamino)-4-methyl-8-(l-methylethyl)-6-(2-thienyl)pyrido[2,3-d]pyrimidin-7(8H)- one;
8-ethyl-2-(ethylamino)-6-(lH-indol-6-yl)-4-methylpyrido[2,3-d]pyrimidin-7(8H)-one; 8-ethyl-2-(ethylamino)-4-methyl-6-(5-phenyl-2-thienyl)pyrido[2,3-d]pyrimidin-7(8H)-one; 2-(ethylamino)-6-furan-3-yl-4-methyl-8-(l-methylethyl)pyrido[2,3-d]pyrimidin-7(8H)- one;
8-ethyl-2-(ethylamino)-4-methyl-6-(lH-pyrazol-5-yl)pyrido[2,3-d]pyrimidin-7(8H)-one;
8-cyclohexyl-2-(ethylamino)-4-methyl-6-(2-thienyl)pyrido[2,3-d]pyrimidin-7(8H)-one;
8-ethyl-2-(ethylamino)-4-methyl-6-(l,3-thiazol-2-yl)pyrido[2,3-d]pyrimidin-7(8H)-o^^
8-cyclopentyl-2-(ethylamino)-4-methyl-6-(lH-pyrazol-3-yl)pyrido[2,3-d]pyrimidin-7(8H)- one;
2-(ethylamino)-4-methyl-8-( 1 -methy lethyl)-6-( 1 H-pyrazol-5-yl)pyrido[2,3-d]pyrimidin- 7(8H)-one;
8-ethyl-2-(ethylamino)-4-methyl-6-(lH-pyrazol-l-yl)pyrido[2,3-d]pyrimidin-7(8H)-one;
2-(ethylamino)-4-methyl-8-(l-methylethyl)-6-(lH-pyrazol-l-yl)pyrido[2,3-d]pyrim
7(8H)-one;
8-cyclopentyl-2-(ethylamino)-4-methyl-6-(lH-pyrazol-l-yl)pyrido[2,3-d]pyrimidin-7(8H)- one;
8-ethyl-4-methyl-6-(lH-pyrazol-5-yl)-2-[(2,2,2-trifluoroethyl)amino]pyrido[2,3- i/]pyrimidin-7(8H)-one;
2-(ethylamino)-4-methyl-6-(lH-pyrazol-3-yl)pyrido[2,3- |pyrimidin-7(8H)-one;
8-ethyl-4-methyl-2-(methylamino)-6-(lH-pyrazol-5-yl)pyrido[2,3-^pyrimidin-7(8H)-one; and
8-ethyl-2-[(2-fluoroethyl)amino]-4-methyl-6-(lH-pyrazol-5-yl)pyrido[2,3-ii]pyrimidin- 7(8H)-one.
[00135] In another embodiment, the compound of formula IA is selected form:
2-ammo-8-ethyl-4-methyl-6-(lH-pyrazol-5-yl)pyrido[2,3-ii]pyrimidin-7(8H)-one;
2-amino-8-cyclopentyl-4-methyl-6-(lH-pyrazol-3-yl)pyrido[2,3-iijpyrimidin-7(8H)-one; 2-amino-4-me l-8-(l-methylemyl)-6-(lH-pyriizol-3-yl)pyrido[2,3-ii|pyrimidin-7(8H)- one; :
2-amino-4-memyl-8-(phenylmethyl)-6-(lH-pyrazol-3-yl)pyrido[2,3-(i]pynmidin-7(8H)- one;
2-amino-8-emyl-4-methyl-6-(4-met^
2-amino-8-emyl-4-me l-6-(2-thienyl)pyrido[2,3-^pyrimidin-7(8H)-one;
2-amino-8-emyl-4-methyl-6-(3-thienyl)pyrido[2,3-i/]pyrimidin-7(8H)-one;
2-amino-8-ethyl-6-furan-3-yl-4-methylpyrido[2,3-i/]pyrimidin-7(8H)-one
2-amino-8-emyl-4-methyl-6-phenylpyrido[2,3--i|pyrimidin-7(8H)-one;
2-amino-8-emyl-6-isoxazol-4-yl-4-methylpyrido[2,3-ii]pyrimidin-7(8H)-one;
2-amino-8-emyl-6-furan-2-yl-4-methylpyrido[2,3-i/]pyrimidin-7(8H)-one;
5-(2-amino-8-emyl-4-methyl-7-oxo-7,8-dihydropyrido[2,3-c%y
carbonitrile;
2-amino-8-emyl-4-memyl-6-pyrimidin-5-ylpyrido[2,3--i]pyrimidin-7(8H)-one;
2-amino-8-emyl-6-(lH-imidazol-5-yl)-4-methylpyrido[2,3-(i]pyrimidin-7(8H)-one;
2-amino-8-ethyl-4-methyl-6-( 1H- 1 ,2,3-triazol-5-yl)pyrido[2,3-i/]pyrimidin-7(8H)-one;
2-amino-8-emyl-4-memyl-6-(lH-pyrazol-4-yl)pyrido[2,3-rf]pyrimidin-7(8H)-one;
2-amino-8-ethyl-4-methyl-6-(l ,3-thiazol-2-yl)pyrido[2,3-ci]pyrirnidin-7(8H)-one;
2-amino-8-emyl-4-methyl-6-( lH-tetrazol-5-yl)pyrido[2,3-^pyrimidin-7(8H)-one;
2-amino-8-emyl-4-memyl-6-(l-memyl-lH-pyrrol-2-yl)pyri^^
2-amino-4,8-diethyl-6-(lH-pyrazol-5-yl)pyrido[2,3-i/]pyrirnidin-7(8H)-one; and
2-amino-8-cyclopentyl-4-methyl-6-( l ,3-thiazol-5-yl)pyrido[2,3-ii|pyrimidin-7(8H)-one.
[00136] In another embodiment, the compound of formula IA is
Figure imgf000038_0001
2-amino-8-ethyl-4-methyl-6-(lH-pyrazol-5-yl)pyrido[2,3-d]pyrimidin-7(8H)-one (Compound A) or a pharmaceutically acceptable salt thereof.
[00137] Another embodiment (P) of the Invention is a method of treating disease, disorder, or syndrome where the disease is associated with uncontrolled, abnormal, and/or unwanted cellular activities effected directly or indirectly by PI3Ka which method comprises administering to a human in need thereof a therapeutically effective amount of a compound of Formula I or II or a pharmaceutically acceptable salt, solvate, or a pharmaceutical composition thereof. Specifically, the Compound is of Formula I.
[00138] Another embodiment (Q) of the invention is directed to a method of treating a disease, disorder, or syndrome which method comprises administering to a patient a therapeutically effective amount of a compound of Formula I, Π, or III or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition comprising a
therapeutically effective amount of a compound of Formula I and a pharmaceutically acceptable carrier, excipient, or diluent. Specifically, the disease is cancer. More
specifically, the cancer is breast cancer, colon cancer, rectal cancer, endometrial cancer, gastric carcinoma, glioblastoma, hepatocellular carcinoma, small cell lung cancer, non-small cell lung cancer, melanoma, ovarian cancer, cervical cancer, pancreatic cancer, prostate carcinoma, acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), or thyroid carcinoma. Even more specifically, the cancer is ovarian cancer, cervical cancer, breast cancer, colon cancer, rectal cancer, or glioblastoma.
[00139] Another embodiment (R) of the Invention is directed to a method of treating a disease, disorder, or syndrome which method comprises administering to a patient a therapeutically effective amount of a compound of Formula I, Π. ΙΠ or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition comprising a
therapeutically effective amount of a compound of Formula II and a pharmaceutically acceptable carrier, excipient, or diluent. Specifically, the disease is cancer. More
specifically, the cancer is breast cancer, colon cancer, rectal cancer, endometrial cancer, gastric carcinoma, glioblastoma, hepatocellular carcinoma, small cell lung cancer, non-small cell lung cancer, melanoma, ovarian cancer, cervical cancer, pancreatic cancer, prostate carcinoma, acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), or thyroid carcinoma. Even more specifically, the cancer is ovarian cancer, cervical cancer, breast cancer, colon cancer, rectal cancer, or glioblastomas.
[00140] Another aspect of the invention is a method of inhibiting proliferative activity in a cell, the method comprising administering to a cell or a plurality of cells an effective amount of a compound of Formula I or Π, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, or a pharmaceutical composition thereof. Specifically, the Compound is of Formula I.
[00141] Another aspect of the invention is directed to employing the compounds of the invention in a method of screening for candidate agents that bind to, for example PBKoc. The protein is bound to a support, and a compound of the invention is added to the assay. Alternatively, the compound of the invention is bound to the support and the protein is added. Classes of candidate agents among which novel binding agents may be sought include specific antibodies, non-natural binding agents identified in screens of chemical libraries, peptide analogs, etc. Of particular interest are screening assays for candidate agents that have a low toxicity for human cells. A wide variety of assays may be used for this purpose, including labeled in vitro protein-protein binding assays, electrophoretic mobility shift assays, immunoassays for protein binding, functional assays (phosphorylation assays, etc.) and the like.
[00142] The determination of the binding of the candidate agent to, for example, PBKcc can be done in a number of ways. In one example, the candidate agent (the compound of the invention) is labeled, for example, with a fluorescent or radioactive moiety and binding determined directly. For example, this may be done by attaching all or a portion of the PI3Ka protein to a solid support, adding a labeled agent (for example a compound of the invention in which at least one atom has been replaced by a detectable isotope), washing off excess reagent, and determining whether the amount of the label is that present on the solid support. Various blocking and washing steps may be utilized as is known in the art.
[00143] The term "labeled" as used herein is meant to include both direct and indirect labeling with a compound that provides a detectable signal, for example, radioisotope, fluorescent tag, enzyme, antibodies, particles such as magnetic particles, chemiluminescent tag, or specific binding molecules, and the like. Specific binding molecules include pairs, such as biotin and streptavidin, digoxin and antidigoxin, and the like. For the specific binding members, the complementary member would normally be labeled with a molecule which provides for detection, in accordance with known procedures, as outlined above. The label can directly or indirectly provide a detectable signal.
[00144] In some embodiments, only one of the components is labeled. For example, PBKcc protein may be labeled at tyrosine positions using 125I, or with fluorophores.
Alternatively, more than one component may be labeled with different labels; using 125I for the proteins, for example, and a fluorophor for the candidate agents.
[00145] The compounds of the invention may also be used as competitors to screen for additional drug candidates. The terms "candidate bioactive agent" or "drug candidate" or grammatical equivalents as used herein describe any molecule, e.g., protein, oligopeptide, small organic molecule, polysaccharide, polynucleotide, etc., to be tested for bioactivity. They may be capable of directly or indirectly altering the cellular proliferation phenotype or the expression of a cellular proliferation sequence, including both nucleic acid sequences and protein sequences. In other cases, alteration of cellular proliferation protein binding and/or activity is screened. In the case where protein binding or activity is screened, some embodiments exclude molecules already known to bind to that particular protein. Exemplary embodiments of assays described herein include candidate agents, which do not bind the target protein in its endogenous native state, termed herein as "exogenous" agents. In one example, exogenous agents further exclude antibodies to PI3Ka.
[00146] Candidate agents can encompass numerous chemical classes, though typically they are organic molecules having a molecular weight of more than about 100 and less than about 2,500 daltons. Candidate agents comprise functional groups necessary for structural interaction with proteins, particularly hydrogen bonding and lipophilic binding, and typically include at least an amine, carbonyl, hydroxyl, ether, or carboxyl group, for example at least two of the functional chemical groups. The candidate agents often comprise carbocyclic or heterocyclic structures and/or aromatic or polyaromatic structures substituted with one or more of the above functional groups. Candidate agents are also found among biomolecules including peptides, saccharides, fatty acids, steroids, purines, pyrimidines, derivatives, structural analogs, or combinations thereof.
[00147] Candidate agents are obtained from a wide variety of sources including libraries of synthetic or natural compounds. For example, numerous means are available for random and directed synthesis of a wide variety of organic compounds and biomolecules, including expression of randomized oligonucleotides. Alternatively, libraries of natural compounds in the form of bacterial, fungal, plant and animal extracts are available or readily produced. Additionally, natural or synthetically produced libraries and compounds are readily modified through conventional chemical, physical and biochemical means. Known pharmacological agents may be subjected to directed or random chemical modifications, such as acylation, alkylation, esterification, amidification to produce structural analogs.
[00148] In one example, the binding of the candidate agent is determined through the use of competitive binding assays. In this example, the competitor is a binding moiety known to bind to PBKot, such as an antibody, peptide, binding partner, ligand, etc. Under certain circumstances, there may be competitive binding as between the candidate agent and the binding moiety, with the binding moiety displacing the candidate agent.
[00149] In some embodiments, the candidate agent is labeled. Either the candidate agent, or the competitor, or both, is added first to PI3Ka protein for a time sufficient to allow binding, if present. Incubations may be performed at any temperature that facilitates optimal activity, typically between 4°C and 40°C. [00150] Incubation periods are selected for optimum activity, but may also be optimized to facilitate rapid high throughput screening. Typically between 0.1 and 1 hour will be sufficient. Excess reagent is generally removed or washed away. The second component is then added, and the presence or absence of the labeled component is followed, to indicate binding.
[00151] In one example, the competitor is added first, followed by the candidate agent. Displacement of the competitor is an indication the candidate agent is binding to ΡΒΚα and thus is capable of binding to, and potentially modulating, the activity of the PBKoe. In this embodiment, either component can be labeled. Thus, for example, if the competitor is labeled, the presence of label in the wash solution indicates displacement by the agent.
Alternatively, if the candidate agent is labeled, the presence of the label on the support indicates displacement.
[00152] In an alternative embodiment, the candidate agent is added first, with incubation and washing, followed by the competitor. The absence of binding by the competitor may indicate the candidate agent is bound to PDKcc with a higher affinity. Thus, if the candidate agent is labeled, the presence of the label on the support, coupled with a lack of competitor binding, may indicate the candidate agent is capable of binding to PI3Ka.
[00153] It may be of value to identify the binding site of PI3Koc. This can be done in a variety of ways. In one embodiment, once PI3Koc is identified as binding to the candidate agent, the PI3Ko is fragmented or modified and the assays repeated to identify the necessary components for binding.
[00154] Modulation is tested by screening for candidate agents capable of modulating the activity of PDKcc comprising the steps of combining a candidate agent with PI3Kcc, as above, and determining an alteration in the biological activity of the PI3Koc Thus, in this embodiment, the candidate agent should both bind to (although this may not be necessary), and alter its biological or biochemical activity as defined herein. The methods include both in vitro screening methods and in vivo screening of cells for alterations in cell viability, morphology, and the like.
[00155] Alternatively, differential screening may be used to identify drug candidates that bind to native PI3Koc, but cannot bind to modified PI3Koc.
[00156] Positive controls and negative controls can be used in the assays. For example, all control and test samples are performed in at least triplicate to obtain statistically significant results. Incubation of samples is for a time sufficient for the binding of the agent to the protein. Following incubation, samples are washed free of non-specifically bound material and the amount of bound, generally labeled agent determined. For example, where a radioiabel is employed, the samples can be counted in a scintillation counter to determine the amount of bound compound.
[00157] A variety of other reagents can be included in the screening assays. These include reagents like salts, neutral proteins, e.g., albumin, detergents, etc which may be used to facilitate optimal protein-protein binding and/or reduce non-specific or background interactions. Also reagents that otherwise improve the efficiency of the assay, such as protease inhibitors, nuclease inhibitors, anti-microbial agents, etc., may be used. The mixture of components can be added in any order that provides for the requisite binding.
[00158] One of ordinary skill in the art would understand that certain crystallized, protein- ligand complexes, in particular PI3Kcc-ligand-ligand complexes, and their corresponding x- ray structure coordinates can be used to reveal new structural information useful for understanding the biological activity of kinases as described herein. As well, the key structural features of the aforementioned proteins, particularly, the shape of the ligand binding site, are useful in methods for designing or identifying selective modulators of kinases and in solving the structures of other proteins with similar features. Such protein- ligand complexes, having compounds of the invention as their ligand component, are an aspect of the invention.
[00159] As well, one of ordinary skill in the art would appreciate that such suitable x-ray quality crystals can be used as part of a method of identifying a candidate agent capable of binding to and modulating the activity of kinases. Such methods may be characterized by the following aspects: a) introducing into a suitable computer program, information defining a ligand binding domain of a kinase in a conformation (e.g. as defined by x-ray structure coordinates obtained from suitable x-ray quality crystals as described above) wherein the computer program creates a model of the three dimensional structures of the ligand binding domain, b) introducing a model of the three dimensional structure of a candidate agent in the computer program, c) superimposing the model of the candidate agent on the model of the ligand binding domain, and d) assessing whether the candidate agent model fits spatially into the ligand binding domain. Aspects a-d are not necessarily carried out in the aforementioned order. Such methods may further entail: performing rational drug design with the model of the three-dimensional structure, and selecting a potential candidate agent in conjunction with computer modeling. [00160] Additionally, one skilled in the art would appreciate that such methods may further entail: employing a candidate agent, so-determined to fit spatially into the ligand binding domain, in a biological activity assay for kinase modulation, and determining whether said candidate agent modulates kinase activity in the assay. Such methods may also include administering the candidate agent, determined to modulate kinase activity, to a mammal suffering from a condition treatable by kinase modulation, such as those described above.
[00161] Also, one skilled in the art would appreciate that compounds of the invention can be used in a method of evaluating the ability of a test agent to associate with a molecule or molecular complex comprising a ligand binding domain of a kinase. Such a method may be characterized by the following aspects: a) creating a computer model of a kinase binding pocket using structure coordinates obtained from suitable x-ray quality crystals of the kinase, b) employing computational algorithms to perform a fitting operation between the test agent and the computer model of the binding pocket, and c) analyzing the results of the fitting operation to quantify the association between the test agent and the computer model of the binding pocket.
Representative Compounds
[00162] Representative compounds of Formula I and/or II are depicted below. The examples are merely illustrative and do not limit the scope of the invention in any way. Compounds of the invention are named according to systematic application of the nomenclature rules agreed upon by the International Union of Pure and Applied Chemistry (IUPAC), International Union of Biochemistry and Molecular Biology (IUBMB), and the Chemical Abstracts Service (CAS). Names were generated using ACD Labs naming software 8.00 release, product version 8.08.
Table 1
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
H-
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
General Administration
[00163] In one aspect, the invention provides pharmaceutical compositions comprising an inhibitor of PI3K according to the invention and a pharmaceutically acceptable carrier, excipient, or diluent. In certain other specific embodiments, administration is by the oral route. Administration of the compounds of the invention, or their pharmaceutically acceptable salts, in pure form or in an appropriate pharmaceutical composition, can be carried out via any of the accepted modes of administration or agents for serving similar utilities. Thus, administration can be, for example, orally, nasally, parenterally (intravenous, intramuscular, or subcutaneous), topically, transdermally, intra vaginally, intra vesically, intracistemally, or rectally, in the form of solid, semi-solid, lyophilized powder, or liquid dosage forms, such as for example, tablets, suppositories, pills, soft elastic and hard gelatin capsules, powders, solutions, suspensions, or aerosols, or the like, specifically in unit dosage forms suitable for simple administration of precise dosages. [00164] The compositions will include a conventional pharmaceutical carrier or excipient and a compound of the invention as the/an active agent, and, in addition, may include carriers and adjuvants, etc.
[00165] Adjuvants include preserving, wetting, suspending, sweetening, flavoring, perfuming, emulsifying, and dispensing agents. Prevention of the action of microorganisms can be ensured by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, and the like. It may also be desirable to include isotonic agents, for example sugars, sodium chloride, and the like. Prolonged absorption of the injectable pharmaceutical form can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin.
[00166] If desired, a pharmaceutical composition of the invention may also contain minor amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents, antioxidants, and the like, such as, for example, citric acid, sorbitan monolaurate, triethanolamine oleate, butylalted hydroxytoluene, etc.
[00167] The choice of formulation depends on various factors such as the mode of drug administration (e.g., for oral administration, formulations in the form of tablets, pills or capsules) and the bioavailability of the drug substance. Recently, pharmaceutical formulations have been developed especially for drugs that show poor bioavailability based upon the principle that bioavailability can be increased by increasing the surface area i.e., decreasing particle size. For example, U.S. Pat. No. 4,107,288 describes a pharmaceutical formulation having particles in the size range from 10 to 1,000 nm in which the active material is supported on a crosslinked matrix of macromolecules. U.S. Pat. No. 5,145,684 describes the production of a pharmaceutical formulation in which the drug substance is pulverized to nanoparticles (average particle size of 400 nm) in the presence of a surface modifier and then dispersed in a liquid medium to give a pharmaceutical formulation that exhibits remarkably high bioavailability.
[00168] Compositions suitable for parenteral injection may comprise physiologically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions.
Examples of suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols (propylenegiycol, polyethyleneglycol, glycerol, and the like), 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 a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants.
[00169] One specific route of administration is oral, using a convenient daily dosage regimen that can be adjusted according to the degree of severity of the disease-state to be treated.
[00170] Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is admixed with at least one inert customary excipient (or carrier) such as sodium citrate or dicalcium phosphate or (a) fillers or extenders, as for example, starches, lactose, sucrose, glucose, mannitol, and silicic acid, (b) binders, as for example, cellulose derivatives, starch, alignates, gelatin, polyvinylpyrrolidone, sucrose, and gum acacia, (c) humectants, as for example, glycerol, (d) disintegrating agents, as for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, croscarmellose sodium, complex silicates, and sodium carbonate, (e) solution retarders, as for example paraffin, (f) absorption accelerators, as for example, quaternary ammonium compounds, (g) wetting agents, as for example, cetyl alcohol, and glycerol monostearate, magnesium stearate and the like (h) adsorbents, as for example, kaolin and bentonite, and (i) lubricants, as for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, or mixtures thereof. In the case of capsules, tablets, and pills, the dosage forms may also comprise buffering agents.
[00171] Solid dosage forms as described above can be prepared with coatings and shells, such as enteric coatings and others well known in the art. They may contain pacifying agents, and can also be of such composition that they release the active compound or compounds in a certain part of the intestinal tract in a delayed manner. Examples of embedded compositions that can be used are polymeric substances and waxes. The active compounds can also be in microencapsulated form, if appropriate, with one or more of the above-mentioned excipients.
[00172] Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs. Such dosage forms are prepared, for example, by dissolving, dispersing, etc., a compound(s) of the invention, or a
pharmaceutically acceptable salt thereof, and optional pharmaceutical adjuvants in a carrier, such as, for example, water, saline, aqueous dextrose, glycerol, ethanol and the like;
solubilizing agents and emulsifiers, as for example, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propyleneglycol,
1,3-butyleneglycol, dimethylformamide; oils, in particular, cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil and sesame oil, glycerol, tetrahydrofurfuryl alcohol, polyethyleneglycols and fatty acid esters of sorbitan; or mixtures of these substances, and the like, to thereby form a solution or suspension.
[00173] Suspensions, in addition to the active compounds, may contain suspending agents, as for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, or mixtures of these substances, and the like.
[00174] Compositions for rectal administrations are, for example, suppositories that can be prepared by mixing the compounds of the present invention with for example suitable non- irritating excipients or carriers such as cocoa butter, polyethyleneglycol or a suppository wax, which are solid at ordinary temperatures but liquid at body temperature and therefore, melt while in a suitable body cavity and release the active component therein.
[00175] Dosage forms for topical administration of a compound of this invention include ointments, powders, sprays, and inhalants. The active component is admixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants as may be required. Ophthalmic formulations, eye ointments, powders, and solutions are also contemplated as being within the scope of this invention.
[00176] Compressed gases may be used to disperse a compound of this invention in aerosol form. Inert gases suitable for this purpose are nitrogen, carbon dioxide, etc.
[00177] Generally, depending on the intended mode of administration, the
pharmaceutically acceptable compositions will contain about 1 % to about 99% by weight of a compound(s) of the invention, or a pharmaceutically acceptable salt thereof, and 99% to 1 % by weight of a suitable pharmaceutical excipient. In one example, the composition will be between about 5% and about 75% by weight of a compound(s) of the invention, or a pharmaceutically acceptable salt thereof, with the rest being suitable pharmaceutical excipients.
[00178] Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see Remington's Pharmaceutical Sciences, 18th Ed., (Mack Publishing Company, Easton, Pa., 1 90). The composition to be administered will, in any event, contain a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof, for treatment of a disease-state in accordance with the teachings of this invention.
[00179] The compounds of the invention, or their pharmaceutically acceptable salts or solvates, are administered in a therapeutically effective amount which will vary depending upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of the compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular disease-states, and the host undergoing therapy. The compounds of the present invention can be administered to a patient at dosage levels in the range of about 0.1 to about 1 ,000 mg per day. For a normal human adult having a body weight of about 70 kilograms, a dosage in the range of about 0.01 to about 100 mg per kilogram of body weight per day is an example. The specific dosage used, however, can vary. For example, the dosage can depend on a number of factors including the requirements of the patient, the severity of the condition being treated, and the pharmacological activity of the compound being used. The determination of optimum dosages for a particular patient is well known to one of ordinary skill in the art.
[00180] If formulated as a fixed dose, such combination products employ the compounds of this invention within the dosage range described above and the other pharmaceutically active agent(s) within its approved dosage range. Compounds of the instant invention may alternatively be used sequentially with known pharmaceutically acceptable agent(s) when a combination formulation is inappropriate.
[00181] Representative pharmaceutical formulations containing a compound of Formula I are described below in the Pharmaceutical Composition Examples.
UTILITY
[00182] Certain compounds of this invention have been tested using the assay described in Biological Example I and have been determined to be PI3K inhibitors. As such compounds of Formula I are useful for treating diseases, particularly cancer in which PI3K activity contributes to the pathology and/or symptomatology of the disease. For example, cancer in which PI3K activity contributes to its pathology and/or symptomatology include breast cancer, colon cancer, rectal cancer, endometrial cancer, gastric carcinoma, glioblastoma, hepatocellular carcinoma, small cell lung cancer, non-small cell lung cancer, melanoma, ovarian cancer, cervical cancer, pancreatic cancer, prostate carcinoma, acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), or thyroid carcinoma
[00183] Suitable in vitro assays for measuring PI3K activity and the inhibition thereof by compounds are known in the art. For further details of an in vitro assay for measuring PI3K activity see Biological Examples, Example 1 infra. Following the examples disclosed herein, as well as that disclosed in the art, a person of ordinary skill in the art can determine the inhibitory activity of a compound of this invention. [00184] Assays for measurement of in vitro efficacy in treatment of cancer are known in the art. In addition, cell-based tumor models are described in Biological Examples, Example 2, 3, and 4 infra.
[00185] Suitable in vivo models for cancer are known to those of ordinary skill in the art. For further details of in vivo models for prostate adenocarcinoma, glioblastoma, lung carcinoma, and melanoma, see Biological Examples 5, 6, 7, 8, 9, and 10, infra.
General Synthesis
[00186] Compounds of this invention can be made by the synthetic procedures described below. The starting materials and reagents used in preparing these compounds are either available from commercial suppliers such as Aldrich Chemical Co. (Milwaukee, Wis.), or Bachem (Torrance, Calif.), or are prepared by methods known to those skilled in the art following procedures set forth in references such as Fieser and Fieser's Reagents for Organic Synthesis, Volumes 1-17 (John Wiley and Sons, 1991); Rodd's Chemistry of Carbon Compounds, Volumes 1-5 and Supplemental (Elsevier Science Publishers, 1 89); Organic Reactions, Volumes 1-40 (John Wiley and Sons, 1991), March's Advanced Organic Chemistry, (John Wiley and Sons, 4lh Edition) and Larock's Comprehensive Organic Transformations (VCH Publishers Inc., 1989). These schemes are merely illustrative of some methods by which the compounds of this invention can be synthesized, and various modifications to these schemes can be made and will be suggested to one skilled in the art having referred to this disclosure. The starting materials and the intermediates of the reaction may be isolated and purified if desired using conventional techniques, including but not limited to filtration, distillation, crystallization, chromatography and the like. Such materials may be characterized using conventional means, including physical constants and spectral data.
[00187] Unless specified to the contrary, the reactions described herein take place at atmospheric pressure and over a temperature range from about -78 °C to about 150 °C, more specifically from about 0°C. to about 125 °C and more specifically at about room (or ambient) temperature, e.g., about 20 °C. Unless otherwise stated (as in the case of an hydrogenation), all reactions are performed under an atmosphere of nitrogen.
[00188] Prodrugs can be prepared by techniques known to one skilled in the art. These techniques generally modify appropriate functional groups in a given compound. These modified functional groups regenerate original functional groups by routine manipulation or in vivo. Amides and esters of the compounds of the present invention may be prepared according to conventional methods. A thorough discussion of prodrugs is provided in T. Higuchi and V. Stella, "Pro-drugs as Novel Delivery Systems," Vol 14 of the A.C.S.
Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated herein by reference for all purposes.
The compounds of the invention, or their pharmaceutically acceptable salts, may have asymmetric carbon atoms or quaternized nitrogen atoms in their structure. Compounds of Formula I that may be prepared through the syntheses described herein may exist as single stereoisomers, racemates, and as mixtures of enantiomers and diastereomers. The compounds may also exist as geometric isomers. All such single stereoisomers, racemates and mixtures thereof, and geometric isomers are intended to be within the scope of this invention. Some of the compounds of the invention may exist as tautomers. For example, where a ketone or aldehyde is present, the molecule may exist in the enol form; where an amide is present, the molecule may exist as the imidic acid; and where an enamine is present, the molecule may exist as an imine. All such tautomers are within the scope of the invention. In particular, imidazol-5-yl and pyrazol-5-yl each can also exist in their respective tautomeric forms imidazol-4-yl and pyrazol-3-yI. Regardless of which structure or which terminology is used, each tautomer is included within the scope of the Invention.
[00189] The present invention also includes N-oxide derivatives and protected derivatives of compounds of Formula I. For example, when compounds of Formula I contain an oxidizable nitrogen atom, the nitrogen atom can be converted to an N-oxide by methods well known in the art. When compounds of Formula I contain groups such as hydroxy, carboxy, thiol or any group containing a nitrogen atom(s), these groups can be protected with a suitable "protecting group" or "protective group". A comprehensive list of suitable protective groups can be found in T.W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, Inc. 1991, the disclosure of which is incorporated herein by reference in its entirety. The protected derivatives of compounds of Formula I can be prepared by methods well known in the art.
[00190] Methods for the preparation and/or separation and isolation of single
stereoisomers from racemic mixtures or non-racemic mixtures of stereoisomers are well known in the art. For example, optically active (R)- and (S)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques. Enantiomers (R- and S-isomers) may be resolved by methods known to one of ordinary skill in the art, for example by: formation of diastereoisomeric salts or complexes which may be separated, for example, by crystallization; via formation of diastereoisomeric derivatives which may be separated, for example, by crystallization, selective reaction of one enantiomer with an enantiomer-specific reagent, for example enzymatic oxidation or reduction, followed by separation of the modified and unmodified enantiomers; or gas-liquid or liquid
chromatography in a chiral environment, for example on a chiral support, such as silica with a bound chiral ligand or in the presence of a chiral solvent. It will be appreciated that where a desired enantiomer is converted into another chemical entity by one of the separation procedures described above, a further step may be required to liberate the desired
enantiomeric form. Alternatively, specific enantiomer may be synthesized by asymmetric synthesis using optically active reagents, substrates, catalysts or solvents or by converting on enantiomer to the other by asymmetric transformation. For a mixture of enantiomers, enriched in a particular enantiomer, the major component enantiomer may be further enriched (with concomitant loss in yield) by recrystallization.
[00191] In addition, the compounds of the present invention can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. In general, the solvated forms are considered equivalent to the unsolvated forms for the purposes of the present invention.
[00192] The chemistry for the preparation of the compounds of this invention is known to those skilled in the art. In fact, there may be more than one process to prepare the compounds of the invention. For specific examples, see M. Barvian et al. J. Med. Chem. 2000, 43, 4606-4616; S. N. VanderWei et al. J. Med. Chem. 2005, 48, 2371-2387; P. L. Toogood et al. J. Med. Chem. 2005, 48, 2388-2406; J. Kasparec et al. Tetrahedron Letters 2003, 44, 4567-4570; and references cited therein. See also U.S. Pre-grant publication US2004/0009993 Al (M. Angiolini et al.), which is incorporated herein by reference, and references cited therein. The following examples illustrate but do not limit the invention. All references cited herein are incorporated by reference in their entirety.
[00193] A compound of the invention where R1 is optionally substituted alkyl, R2 is hydrogen or optionally substituted alkyl, R4 is methyl or ethyl, R6 is phenyl or heteroaryl each of which is optionally substituted with 1, 2, 3, 4, or 5 R9 groups (as defined in the Summary of the Invention), and R2 is hydrogen can be prepared according to Scheme 1. Scheme 1
Figure imgf000065_0001
[00194] To a solution of commercially available 2-methyl-2-thiopseudourea sulfate in a solvent such as water is added a base such as sodium carbonate and an intermediate of formula 10 at room temperature. The reaction mixture is stirred for overnight or less. After neutralizing, 11 is collected through filtration and followed by drying under vacuum. 11 is then treated with POCI3 and the reaction is heated to reflux for approximately 2 h and then concentrated under vacuum to dryness. 1 can be used directly in the next reaction without further purification.
[00195] An intermediate of formula 2 is prepared by reacting an intermediate of formula 1 with a primary amine R'NH2 in a solvent such as water and with heating. 2 is then treated with iodine monochloride in a solvent such as methanol at around 0 °C and allowed to react for approximately overnight or less as needed for the reaction to go to completion to form 3. After completion the residue is triturated with acetone. The intermediate 3 is then reacted in a solvent, such as DMA, with ethyl acrylate in the presence of a base, such as triethylamine, and in the presence of a catalyst, such as Pd(OAc)2, and (+)BINAP. The reaction is heated to approximately 100 °C and allowed to react for approximately overnight or less as needed for the reaction to go to completion to form 4. 4 is then optionally purified by column chromatography.
[00196] 5 is prepared by treating 4 with DBU in the presence of a base such as DIPEA at room temperature. Then the reaction mixture is heated to reflux and reacted for
approximately 15 h. After evaporation of solvent, the residue is triturated with acetone and collected by filtration to yield 5. [00197] 6 is prepared by reacting 5 with a brominating agent such as Br2 in a solvent such as DCM at room temperature. Then the reaction mixture is stirred for approximately overnight. The resulting product is filtered and then suspended in a solvent such as DCM and treated with a base such as triethylamine. The mixture is then washed with water and dried over a drying agent such as Na2S04 to yield 6.
[00198] A Suzuki coupling is then performed using 6 reacting with a boronic acid (or ester) of formula R6B(OH)2 in a solvent(s) such as a DME-H20 mixture, in the presence of a catalyst such as Pd(dpppf ) and a base such as triethylamine at room temperature. The reaction mixture is heated to reflux for approximately 4 h. After cooling to room temperature, the reaction mixture is partitioned with water and ethyl acetate. After separation, the organic layer is dried over a drying agent such as Na2S04 to yield 7.
[00199] The methylthio group of 7 is then oxidized with m-CPBA in a solvent such as DCM at room temperature allowing to stir for approximately 4 h. After removal of the solvent under reduced pressure, the product is treated with an amine of formula R2NH2 in a solvent such as dioxane and stirred at room temperature for approximately overnight to yield a Compound of Formula I.
[00200] Alternatively, a compound of the invention where R1 is optionally substituted alkyl, R4 is methyl or ethyl, R6 is phenyl or heteroaryl each of which is optionally substituted with 1, 2, 3, 4, or 5 R9 groups (as defined in the Summary of the Invention), and R2 is hydrogen can be prepared according to Scheme 2.
Scheme 2
Figure imgf000066_0001
11 12
[00201] An intermediate of formula 9 is prepared by reacting an intermediate of formula 8 with neat POCl3 and heating. 9 is then treated with a primary amine R'NH2 in a solvent such as water or THF and triethylamine at 0 °C to form 10. After removal of the solvent under reduced pressure, the intermediate 10 is then reacted with lithium aluminum hydride in a solvent such as THF at 0 °C. After quenching and aqueous workup, solvent removal provided crystalline 11 without further purification. Treatment of 11 with manganese (II) dioxide in a solvent such as methylene chloride or chloroform at room temperature provided aldehyde 12 upon Filtration and solvent removal. A Wittig reaction with aldehyde 12 can be employed with (carbethoxymethylene)triphenylphosphorane in refluxing THDF to provide the common intermediate 4. 4 can then be used to prepare a Compound of Formula I using the procedures described in Scheme 1.
[00202] A compound of the invention where R1 is optionally substituted alkyl, R4 is methyl or ethyl, R6 is phenyl or heteroaryl each of which is optionally substituted with 1, 2, 3, 4, or 5 R9 groups (as defined in the Summary of the Invention), and R2 is hydrogen can be prepared according to Scheme 3.
Scheme 3
Figure imgf000067_0001
[00203] An intermediate of formula 14 is prepared by reacting an intermediate of formula 13 with a primary amine R'NHa in a solvent such as water and with heating. 14 is then treated with iodine monochloride in a solvent such as methanol at around 0 °C and allowed to react for approximately overnight or less as needed for the reaction to go to completion to form IS. After completion the residue is triturated with acetone. The intermediate 15 is then reacted in a solvent, such as DMA, with ethyl acrylate in the presence of a base, such as triethylamine, and in the presence of a catalyst, such as Pd(OAc)2, and (+)BINAP. The reaction is heated to approximately 100 °C and allowed to react for approximately overnight or less as needed for the reaction to go to completion to form 16. 16 is then optionally purified by column chromatography. A Compound of Formula I can then be prepared from 16 by using the same reaction conditions as described in Scheme 1 (starting at the point of the preparation of 5 from 4).
[00204] A compound of the invention where R1 is optionally substituted alkyl, R4 is methyl or ethyl, R6 is phenyl or heteroaryl each of which is optionally substituted with 1, 2, 3, 4, or 5 R9 groups (as defined in the Summary of the Invention), and R2 is hydrogen can alternatively be prepared according to Scheme 4.
Scheme 4
Figure imgf000068_0001
23 24 25 I
[00205] An intermediate of formula 20 is prepared by reacting an intermediate of formula 19 with neat POCl3 and heating. 20 is then treated with a primary amine R*NH2 in a solvent such as water or THF and triethylamine at 0 °C to form 21. After removal of the solvent under reduced pressure, the intermediate 21 is then reacted with lithium aluminum hydride in a solvent such as THF at 0 °C. After quenching and aqueous workup, solvent removal provided crystalline 22 without further purification. Treatment of 22 with manganese (II) dioxide in a solvent such as methylene chloride or chloroform at room temperature provided aldehyde 23 upon filtration and solvent removal. A Knovenegal-type condensation with 23 and an arylacetonitrile in the presence of a base such as potassium carbonate or sodium hydroxide in a protic solvent provides the cyclized imine 24. Acetylation of the imine with acetic anhydride is required prior to hydrolysis which takes place in the presence of aqueous acid and heating to afford 25. Subsequently, 25 can be oxidized to the corresponding sulfone with m-CPBA at room temperature and displaced with ammonium to provide I.
Synthetic Examples
Example 1
2-anii -8-ethyl-4-methyl-6-(lH-pyrazol-5-yl)pyrido[2,3-iflpyrimidin-7(8H)-one
Figure imgf000068_0002
[00206] To a solution of 2-methyl-2-thiopseudourea sulfate (Aldrich, 58.74 g, 0.422 mol) in water (1000 mL) were added sodium carbonate (81.44 g, 0.768 mol) and ethyl acetoacetate (50 g, 0.384 mol) at room temperature. The reaction mixture was stirred overnight. After neutralizing to pH = 8, the solid was collected through filtration followed by drying under vacuum overnight to afford 6-methyl-2-(methylthio)pyrimidin-4(3H)-one (57.2 g, 95% yield) of product. Ή NMR (400 MHz, DMSO-d6): δ 12.47 (bs, 1H), 5.96 (bs, 1H), 2.47(s, 3H), 2.17 (s, 3H).
Figure imgf000069_0001
[00207] To the round bottom flask containing 6-methyl-2-(methylthio)pyrimidin-4(3H)- one (19 g, 121.6 mmol) was added POCl3 (30 mL). The reaction mixture was heated to reflux for 2 h and then concentrated on a rotary evaporator to dryness. The crude 4-chloro-6- methyl-2-(methylthio)pyrimidine was used directly in the next reaction without further purification.
Figure imgf000069_0002
[00208] To the 4-chloro-6-methyl-2-(methylthio)pyrimidine from above was added 30 mL of a solution of 70% ethylamine in water. The reaction mixture was heated to 50 °C for 3 h. After completion, excess ethylamine was evaporated on rotary evaporator under vacuum. The solid was filtered and dried under vacuum to afford N-ethyl-6-methyl-2- (methylthio)pyrimidin-4-amine (20 g, 90% yield).
Figure imgf000069_0003
[00209] To the solution of N-ethyl-6-methyl-2-(methylthio)pyrimidin-4-amine (20 g, 121.6 mmol) in methanol was added iodine monochloride (26.58 g, 163.7 mmol) in small portions at 0 °C. Then the reaction mixture was stirred overnight. After evaporation of solvent, the residue was triturated with acetone. The product N-ethyl-5-iodo-6-methyl-2- (methylthio)pyrimin-4-amine (25.2 g, 75% yield) was collected by filtration. 1H NMR (400 MHz, CDCI3): δ 5.37 (bs, 1H), 3.52 (q, J = 7.2 Hz, 1H), 2.50 (s, 3H), 1.26 (t, J = 7.2 Hz, 3H).
Figure imgf000070_0001
[00210] To the solution of N-ethyl-5-iodo-6-methyl-2-(methylthio)pyrimin-4-amine (25.2 g, 81.48 mmol) in DMA (260 mL) were added ethyl acrylate (12.23 g, 122.2 mmol), Pd(OAc)2 (3.65 g, 16.25 mmol), (+)BINAP and triethyl amine (24.68 g, 244.4 mmol). Then the reaction mixture was heated to 100 °C and reacted overnight. After evaporation of solvent, the residue was diluted with water and the aqueous layer was extracted with ethyl acetate. The product (E)-ethyl-3-(4-(ethylamino)-6-methyl-2-(methylthio)pyrimidin-5- yl)acrylate ( 16.8 g, 73% yield) was isolated by silica gel column chromatography with 6-8% ethyl acetate in hexane as eluent. Ή NMR (400 MHz, CDC13): 5 7.65 (d, J = 16.4Hz, 1H), 6.20 (d, J = 16.4Hz, 1H), 5.15 (bs, 1 H), 4.28(q, J = 7.2 Hz, 2H), 3.54 (q, J = 7.2 Hz, 2H), 2.53 (s, 3H), 2.37 (s, 3H), 1.35 (t, J = 7.2 Hz, 3H), 1.24 (t, J = 7.2 Hz, 3H).
Figure imgf000070_0002
[00211] To a solution of (E)-ethyl-3-(4-(ethylamino)-6-methyl-2-(methylthio)pyrimidin-5- yl)acrylate (16.8 g, 59.8 mmol) in DIPEA was added l,8-diazabicyclo[5.4.0]undec-7-ene (DBU, 18.21 g, 1 19.6 mmol) at room temperature. Then the reaction mixture was heated to reflux and reacted for 15 h. After evaporation of solvent, the residue was triturated with acetone. The product 8-ethyl-4-methyl-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (10.77 g, 77% yield) was collected by filtration. Ή NMR (400 MHz, CDC13): 8 7.78 (d, J = 9.6 Hz, 1H), 6.63 (d, J = 9.6 Hz, 1H), 4.5(q, J = 7.2 Hz, 2H), 2.67 (s, 3H), 2.62 (s, 3H), 1.33 (t, J = 7.2 Hz, 3H).
Figure imgf000070_0003
[00212] To a solution of 8-ethyl-4-methyl-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)- one (6.31 g, 26.84 mmol) in DCM was added Br2 (4.79 g, 29.52 mmol) dropwise at room temperature. Then the reaction mixture was stirred at room temperature overnight. After filtration the solid was suspended in DCM ( 100 mL), and triethylamine (20 mL) was added. The mixture was washed with water and dried with Na2S04, and the product 6-bromo-8- emyl-4-methyl-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (6.96 g, 83 % yield) was obtained after evaporation of DCM. Ή NMR (400 MHz, CDC13): δ 8.22 (s, 1H), 4.56 (q, J = 7.2 Hz, 2H), 2.6 = 7.2Hz, 3H).
Figure imgf000071_0001
[00213] To a solution of 6-bromo-8-ethyl-4-methyl-2-(methylthio)pyrido[2,3-d]pyrimidin- 7(8H)-one (0.765 g, 2.43 mmol) in DME-H20 (10: 1 11 mL) was added lH-pyrazol-5- ylboronic acid (Frontier, 0.408 g, 3.65 mmol), [ Ι , - bis(diphenylphosphino)ferrocene]dichloropalladium(II) complex with CH2C12
(Pd(dpppf),0.198 g, 0.243 mmol) and triethylamine (0.736 g, 7.29 mmol) at room
temperature. Then the reaction mixture was heated to reflux and reacted for 4 h. After cooling down to room temperature, the reaction mixture was partitioned with water and ethyl acetate. After separation, the organic layer was dried with Na2S04, and the product 8-ethyl-4-methyl- 2-(methylthio)-6-(lH-pyrazol-5-yl)pyrido[2,3-d]pyrimidin-7(8H)-one (0.567 g, 77% yield) was obtained by silica gel column chromatography. Ή NMR (400 MHz, CDC13): 6 13.3 (bs, 1H), 8.54 (s, 1H), 7.82-7.07 (m, 2H), 4.45 (q, J = 7.2 Hz, 2H), 2.71 (s, 3H), 2.60 (s, 3H), 1.26 (t, J = 7.2Hz, 3H).
Figure imgf000071_0002
[00214] To the solution of 8-ethyl-4-methy l-2-(methylthio)-6-( 1 H-pyrazol-5- yI)pyrido[2,3-d]pyrimidin-7(8H)-one (0.123 g, 0.41mmol) in DCM (2 mL) was added MCPBA (0.176 g, 77%, 0.785 mmol) in a small portion at room temperature. Then the reaction mixture was stirred for 4 h. After evaporation of DCM, dioxane (1 mL) and liquid ammonia ( 1 mL) were introduced. The reaction was stirred at room temperature overnight. The product 2-amino-8-ethyl-4-methyl-6-( lH-pyrazol-5-yl)pyrido[2,3-d]pyrimidin-7(8H)- one (50.4 mg) was obtained by silica gel column chromatography. Ή NMR (400 MHz, CD3OD): 5 8.41 (s, IH), 7.62 (d, J = 2.0 Hz, IH), 6.96 (d, J = 2.0Hz, IH), 4.51 (q, J = 7.2Hz, 2H), 2.64 (s, 3H), 1.29 (t, J = 7.2Hz, 3H); MS (EI) for C13Hi4N60: 271.3 (MH+).
[00215] Using the same or analogous synthetic techniques and substituting with appropriate reagents, the following compounds were prepared:
Example la. 2-(amino)-8-ethyl-4-ethyl-6-(lH-pyrazol-5-yl)pyrido[2,3-d]pyrimidin-7(8H)- one: Ή NMR (400 MHz, DMSO-D6): δ 8.40 (s, IH), 7.27 (bs, IH), 7.00 (s, IH), 4.40 (q, J = 7.2 Hz, 2H), 2.95 (d, J = 7.20 Hz, 2H), 1.14 (t, J = 7.2 Hz, 3H), 1.08 (t, J = 7.2Hz, 3H), 0.89 (m, IH), 0.24 (m, 2H), 0.01 (m, 2H); MS (EI) for Cl4Hi6N60: 285.2 (MH+).
Example lb. 8-ethyl-4-methyl-2-(methylamino)-6-( lH-pyrazol-5-yl)pyrido[2,3--i]pyrimidin- 7(8H)-one: Ή NMR (400 MHz, CH3OH-i/4): δ 8.39 (s, IH), 7.60 (bs, IH), 6.93 (bs, IH), 4.53 (bs, 2H), 3.02 (s, 3H), 2.84 (bs, 3H), 1.33 (bs, 3H); MS (EI) for Ci4Hi6N60: 285.3 (MH+).
Example lc. 8-Ethyl-2-[(2-fluoroethyl)amino]-4-methyl-6-( lH-pyrazol-5-yl)pyrido[2,3- <f|pyrirnidin-7(8H)-one: Ή NMR (400 MHz, CH3OH-i/4): δ 8.34 (bs, IH), 7.25 (bs, IH), 6.90 (bs, IH), 4.60 (dt, J = 5.2, 2.2 Hz, 2H), 4.49 (q, J = 7.20 Hz, 2H), 3.78 (dt, J = 5.2, 2.2 Hz, 2H), 2.64 (s, 3H), 1.30 (t, J = 7.2 Hz, 3H); MS (EI) for CsHnFNeO: 317.3 (MH+).
Example Id. 2-Amino-8-cyclopentyl-4-methyl-6-( lH-pyrazol-3-yl)pyrido[2,3-i/]pyrimidin- 7(8H)-one: Ή NMR (400 MHz, OMSO-d6): δ 13.10 (s, IH), 8.42 (d, IH), 7.70 (s, IH), 7.20 (bs, 2H), 6.01 (m, IH), 2.61 (s, 3H), 2.30 (m, 2H), 2.10 (m, 2H), 1.80 (m, 2H), 1.60 (m, 2H); MS (EI) for Ci6Hi8N60: 31 1.8 (M+H).
Intermediate 1
Alternate route to (E)-ethyl-3-(4-(ethylamino)-6-methyl-2-(methylthio)pyrimidin-5- yl)acr late
Figure imgf000072_0001
[00216] N,N-Dimethyl acetamide dimethyl acetal (75 g, 0.56 mole) was added to a suspension of thiourea (33.0 g, 0.43 mole) in methylene chloride. The mixture was heated under reflux for 4 h. The solvent was removed and the residue was crystallized from 5% MeOH and diethyl ether affording (lE)-N'-(aminocarbonothioyl)-N,N- dimeth lethanimidamide (47.8 g, 76% yield).
Figure imgf000073_0001
[00217] A suspension of ( lE)-N'-(atninocarbonothioyl)-N,N-dimethylethanimidamide (47.8 g, 0.33 mole) in methyl iodide (150 mL) and THF (350 mL) was stirred for 18 h at room temperature. The mixture was evaporated under reduced pressure. After addition of 5% MeOH and diethyl ether, the compound precipitated and was collected by filtration affording (lE)-N'-[amino(methylthio)methyl]-N,N-dimethylethanimidamide hydrogen iodide salt (91.0 g, 96% yield).
Figure imgf000073_0002
[00218] To a solution of (lE)-N'-[amino(methylthio)methyl]-N,N- dimethylethanimidamide hydrogen iodide salt (73.0 g, 0.26 mole) in dry dichloromethane (900 mL), was added ethyl 3-chloro-3-oxopropanoate (44 mL, 95% Lancaster, 0.34 mole) was added under a nitrogen atmosphere. The mixture was stirred for 4 h at room temperature, cooled to 0 °C then triethylamine (107 mL, 0.78 mole) was added. The reaction mixture was stirred overnight. The solvent was removed and ¾0 was added. The pH was adjusted to pH = 5.0 with acetic acid and extracted with ethylacetate then evaporated and crystallized from the appropriate solvent (Ethylacetate-Hexanes mixture solvent, approximately 20% ethylacetate-Hexanes). This afforded ethyl 4-methyl-2-(methylthio)-6-oxo-l,6- dihydropyrimidine-5-carboxylate (36.5 g, 62% yield) after drying under vacuum.
Figure imgf000073_0003
[00219] A solution of ethyl 4-methyl-2-(methylthio)-6-oxo- 1 ,6-dihydropyrimidine-5- carboxylate (60 g, 0.26 mole) and phosphorous oxychloride (POCl3, 320 mL) was heated under reflux for 4 to 5 h (monitor reaction by TLC using 30% ethylacetate and hexanes). After completion of reaction, phosphorous oxychloride was removed on a rotary evaporator. The residue was poured on to ice water and extracted with ethylacetate several times. The combined organic layers were evaporated, on a rotary evaporator, to give crude ethyl 4-chloro-6-methyl-2-(methylthio)pyrimidine-5-carboxyIate (65 g). This compound was used without purification.
Figure imgf000074_0001
[00220] To a solution of ethyl 4-chloro-6-methyl-2-(methylthio)pyrimidine-5-carboxylate (65 g) in THF (1000 mL) and triethylamine ( 1 10 mL, 0.81 mole) was added ethylamine (2.0 M in THF, 0.81 mole) at 0 °C. This reaction mixture was stirred at room temperature overnight and then solvents were removed on a rotary evaporator. H20 was added and the mixture extracted with ethyl acetate several times. Solvents from the combined organic layers were removed on a rotary evaporator affording 58 g (86% yield) of ethyl 4- (ethylamino)-6-methyl-2-(methylthio)pyrimidine-5-carboxylate. This material was used as such without further purification.
Figure imgf000074_0002
[00221] To a lithium aluminum hydride solution (LAH, 1.0 M solution in THF, Aldrich, 450 mL) was added a solution of ethyl 4-(ethylamino)-6-methyl-2-(methylthio)pyrimidine-5- carboxylate (57 g) in THF (1000 mL). The reaction mixture was stirred overnight. After cooling to 0 °C, the reaction mixture was cautiously quenched with a 1:9 mixture of
H20/THF until gas evolution has ceased, then diluted with ¾0 (500 mL) and stirred well for 2 h. The resulting slurry was extracted with ethylacetate several times. The aqueous layer was then filtered through Celite and washed with ethylacetate again. The combined organic layers were washed with brine, dried and concentrated under reduced pressure to give 41.0 g (85% yield) of ^-(ethylaminoi-e-methyl^-imethylthioJpyrimidin-S-yUmethanol as a light yellow crystal, which was used without purification in the next step.
Figure imgf000075_0001
[00222] To a solution of [4-(ethylamino)-6-memyl-2-(memylthio)pyrimidin-5-yl]methanol (41.0 g) in chloroform (4000 mL) was added manganese oxide (125 g, 1.4 mole) and stirred for 4 h at room temperature. More manganese oxide was added until the disappearance of alcohol compound was observed. The reaction mixture was filtered through Celite and washed with some chloroform and evaporated all organic solvents to give 38 g (92 % yield) of 4-(emylamino)-6-memyl-2-(memylthio)pyrimidine-5-carbaldehyde as a colorless solid, which was used without purification in the next step.
Figure imgf000075_0002
[00223] To a solution of 4-(ethylamino)-6-methyl-2-(methylthio)pyrimidine-5- carbaldehyde (38 g, 180 mmol) in THF (500 mL) was added (Carbethoxymethylene) triphenylphosphorane (95%, Aldrich, 85.18 g, 244 mmol). The reaction mixture was heated to reflux for 1.5 h and was monitored by TLC (4: 1 hexanes/ethylacetate). The reaction was cooled to room temperature and was concentrated on a rotary evaporator. It was directly subjected to column chromatography (4:1 hexanes/ethylacetate) to give (E)-ethyl-3-(4- (ethylamino)-6-methyl-2-(methylthio)pyrimidin-5-yl)acrylate as a white crystal, 46.14 g (91% yield).
Example 2
2-Amino-6-bromo-8-eth l-4-meth l rido 23-d riniidin-7 8H-one
Figure imgf000075_0003
[00224] To a 3-necked 3-L flask, that was equipped with an overhead stirrer, was added in order 2-amino-4-chloro-6-methylpyrimidine (Aldrich, 100 g, 0.696 mol, 1 equiv.), ethylamine (70% ethylamine in water, Lancaster, 625 mL), 625 mL H20, and 125 mL TEA (0.889 mol, 1.28 equiv.). The mixture was stirred and heated at reflux for 20 h, during which time the reaction turned homogeneous. The reaction was allowed to cool to room
temperature. The volatile ethylamine was removed on a rotary evaporator. A precipitate formed. The aqueous mixture containing the precipitate was allowed to stand at room temperature for 2 h and then filtered. After drying under vacuum, 106 g (100% yield) of 2- amino-6-ethylaminopyrimidine was obtained as a colorless solid. This material was used as such in the following reaction.
Figure imgf000076_0001
[00225] To a solution of 2-amino-6-ethylaminopyrimidine (98 g, 0.64 mol) in methanol (1.6 L) was added IC1 (115.0 g, 0.71 mol) in a small portion at 15 °C. Then the reaction mixture was stirred at room temperature for 3 h (monitored by LC/MS). After evaporation of solvent by rotary evaporator, the residue was triturated with acetone. 2-amino-6-ethylamino- 4-iodopyrimidine hydrochloride ( 188.5 g, 93% isolated yield) was obtained by vacuum filtration and drying. Ή NMR (400 MHz, CD3OD) δ 3.58 (q, 2H), 2.14 (s, 3H), 1.1 1 (t, 3H); MS (EI) for C7HnN4ClI: 279.1 (MH+).
Figure imgf000076_0002
[00226] To a three-neck round bottom flask equipped with over-head mechanic stirrer were added 2-amino-6-ethylamino-4-iodopyrimidine hydrochloride (188.5 g, 0.60 mol), ethyl acrylate (221 mL, 2.0 mol), triethylamine (285 mL, 2.0 mol), DMF (1.3 L), and
tetrakis(triphenylphosphine)palladium(0) (Pd(PPh3)4, 31.3 g, 0.027 mol). The reaction mixture was heated to 95 °C and stirred for 3 h (monitored by LC MC). After reaction completion, the reaction mixture was evaporated about to 1/10 of original volume and partitioned with 500 mL of ethyl acetate and 1000 mL of water. The aqueous layer was extracted with ethyl acetate 5 times. (E)-Ethyl 3-(2-amino-4-(ethyIamino)-6- methylpyrimidin-5-yl)acrylate (100 g, 67% yield) was obtained by recrystalization from acetone after evaporation of ethyl acetate. Ή NMR (400 MHz, CD3OD) δ 7.48 (dd, Jl = 16.0 Hz, 72 = 4.0 Hz, 1H), 6.20 (dd, Jl = 16 Hz, J2 = 4 Hz, 1H), 4.25 (q, J = 7.2 Hz, 2H), 3.51 (q, J = 7.6 Hz, 2H), 2.39 (s, 3H), 1.3 (t, J = 7.2 Hz, 3H), 1.2 (t, / = 7.6 Hz, 3H). MS (EI) for C,2Hi8N402: 251.3 (MH+).
Figure imgf000077_0001
[00227] (E)-Ethyl 3-(2-amino-4-(ethylamino)-6-rnethylpyrimidin-5-yl)acrylate (4.50 g, 18.0 mmol) was added to DBU ( 10.95 g, 4.0 equiv.) and the mixture was heated to 165 °C and stirred for 24 h. After that, the mixture was cooled to 70 °C followed by the addition of H20 (20 mL) to precipitate crystal and stirred for 1 h at room temperature. The crystal was collected and washed with H20 and acetone and dried under vacuum to afford 2.70 g (73.5% yield of 2-amino-8-ethyl-4-methylpyrido[2,3-d]pyrimidin-7(8H)-one as a light yellowish brown solid. LC MS: Calculated for C|0H|2N4O (204.2). Found: 205.31 (M+l); HPLC analytical purity: 98.5%. Ή NMR (400 MHz, OMSO-d6): δ 7.9 (d, 1H), 7.20 (bs, 2H), 6.20 (m, 1H), 4.20 (q, 2H), 2.50 (s, 3H), 1.20 (t, 3H); MS (EI) for C,oH,2N40: 205.1 1 (MlF).
Figure imgf000077_0002
[00228] 2-Amino-8-ethyl-4-methylpyrido[2,3-d]pyrimidin-7(8H)-one (2.70 g, 13.2 mmol) was added to dichloromethane ( 100 mL), and then bromine (0.75 mL, 1.10 equiv.) was added slowly. This reaction mixture was stirred for 3 h at room temperature. After that, the solvent was evaporated nearly 80% volume of reaction mixture under vacuum, and then acetone was added to give 3.54 g 2-Amino-6-bromo-8-ethyl-4-methylpyrido[2,3-d]pyrimidin-7(8H)-one as a tan solid. LC/MS: Calculated for Ci0H| tBrN40 (283.12). Found: 285.15 (M+2). HPLC analytical purity: 97.7%. Example 3
2-Amino-4-methyl-8 methylethyl)-6 ^
one
Figure imgf000078_0001
[00229] To a crude solution of N-isopropyl-6-methyl-2-(methylthio)pyrimidin-4-amine (44.6 g, 224 mmol), prepared using analogous procedures as described in Example 1, in 400 mL of methanol was added IC1 (40.0 g, 246 mmol) in small portions at room temperature. The reaction mixture was then stirred at for 3 h monitoring by LC/MS. After evaporation of solvent by rotary evaporator, the residue was triturated with acetone to yield 5-iodo-N- isopropyl-6-methyl-2-(methylthio)pyrimidin-4-amine. 'H NMR (400 MHz, CDC13) δ 6.37 (br m, 1H), 4.47 (m, 1H), 2.78 (s, 3H), 2.67 (s, 3H), 1.41 (d, J = 6.4, 6H).
Figure imgf000078_0002
[00230] 5-Iodo-N-isopropyl-6-methyl-2-(methylthio)pyrimidin-4-amine (8.1 g, 26.2 mmol), ethyl acrylate (5.24 g, 52.4 mmol), triethylamine (10.6 g, 105 mmol), palladium (II) acetate (1.17 g, 5.23 mmol), and tri-o-tolyl phosphine (1.59 g, 5.23 mmol) were added in that order to 10.8 mL of DMA in a pressure tube and sealed. The reaction mixture was heated to 100 °C and allowed to stir overnight. The reaction was quenched by filtration through a short silica plug washing with ACN. The solvent was evaporated and diluted with ethyl acetate then extracted with 10 % aqueous LiCI, followed by water and brine. NOTE: Extraction is necessary to remove all DMA giving resolution in chromatography. The sample was purified by silica gel column chromatography using 20 % ethyl acetate hexane as eluent. Desired fractions were combined and reduced to afford 2.5 g (34 % yield) of ethyl (2E)-3-[4- (isopropylamino)-6-methyl-2-(methylthio)pyrimidin-5-yl]acrylate as a yellow/orange oil.
Figure imgf000079_0001
[00231] (E)-Ethyl 3-(4-(isopropylamino)-6-methyl-2-(rnethylthio)pyrirnidin-5- yl)acrylate (2.5 g, 8.46 mmol) was dissolved in acetic acid by gentle warming. Sample was placed in microwave reactor for 6 h at 180 °C, 300 W, and 200 PSI. The product was purified by silica gel column chromatography eluting with 20 % ethyl acetate/hexane. Desired fractions were combined and reduced into 8-isopropyl-4-methyl-2-(methylthio)pyrido[2,3- d]pyrimidin-7(8H)-one as a yellow powder (1.20 g, 57 % yield) which was then dried under heavy vacuum overnight. Ή NMR (400MHz, CDC13) δ 7.74 (d, J = 9.6, 1H), 6.58 (d, = 9.6, 1H), 5.84 (br s, 1H), 2.65 (s, 3H), 2.63 (s, 3H), 1.63 (d, J = 6.8, 6H).
Figure imgf000079_0002
[00232] 8-Isopropyl-4-methyl-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (5.38 g, 21.59 mmol) was dissolved in 100 mL DCM. To the stirring solution, m-CPBA ( 13.97 g, 64.78 mmol) was added. The reaction was allowed to stir for 2.5 h at room temperature. LCMS indicated reaction had gone to completion. Sample was diluted with 300 mL of DCM and 300 mL K2C03, upon addition of base a white precipitate formed that dissolved in excess H20. Organic layer was extracted further with H20 and brine, and then dried over Na2C03. The solvent was evaporated to afford the product8-isopropyl-4-methyl-2- (methylsulfonyl)pyrido[2,3-d]pyrimidin-7(8H)-one (6.0 g, 99 % yield) as a light yellow oil that was used immediately in the next reaction.
Figure imgf000079_0003
[00233] 8-isopropyl-4-methyI-2-(methylsulfonyl)pyrido[2,3-d]pyrimidin-7(8H)-one (approximately 3.0 g) was dissolved in 50 mL THF, in a 350 mL pressure tube. While stirring, NH3 (g) was bubbled in through solution for 1.5 minutes. A color change was observed form light yellow to olive green in about 120 seconds. The tube was sealed and stirred at room temperature overnight. A precipitate had formed. The reaction mixture, including precipitate, was reduced to near dryness, filtered and washed with a minimal volume of cold THF, affording 2.88 g of 2-amino-8-isopropyl-4-methylpyrido[2,3- d]pyrimidin-7(8H)-one.
Figure imgf000080_0001
[00234] To a solution of 2-amino-8-isopropyl-4-methylpyrido[2,3-d]pyrimidin-7(8H)- one (2.88 g, 13.19 mmol) dissolved in 80 mL of DCM at 0 °C, (4.21 g, 26.39 mmol) bromine was added. Reaction vessel was removed from ice bath and allowed to react at room temperature over night. LCMS indicated complete conversion of starting material to product. Sample was evaporated to remove DCM and excess bromine. Orange solid was diluted in ethyl acetate and extracted with 10 % NaHS03, H20, and brine. Organic layer was dried over Na2S04, filtered, and reduced to dryness yielding 2-amino-6-bromo-8-isopropyl-4- methylpyrido[2,3-d]pyrimidin-7(8H)-one as a light yellow powder (2.2 g, 56% yield). Ή NMR (400MHz, CDC13) 5 8.08 (s, IH), 5.83 (m, IH), 5.69 (br s, 2H), 2.60 (s, 3H), 1.58 (d, J = 6.8, 6H).
Figure imgf000080_0002
[00235] In a 350 mL pressure tube 2-amino-6-bromo-8-isopropyl-4-methylpyrido[2,3- d]pyrimidin-7(8H)-one ( 1.50 g, 5.05 mmol), lH-pyrazol-3-yl boronic acid ( 1.12 g, 10.09 mmol), K2C03 (336 mg, 15.1 mmol), and tetrakis(triphenylphosphine) palladium (0) (583 mg, 0.0504 mmol) were dissolved in 50 mL dioxane and 5 mL H20. The tube was sealed, heated to 100 °C and allowed to react overnight. A color change was observed. LCMS indicated no presence of starting material. Sample was filtered through a syringe filter and evaporated to dryness. Compound was dissolved in ethyl acetate and triturated in hexane. Light yellow powder of 2-amino-8-isopropyl-4-methyl-6-(lH-pyrazol-5-yl)pyrido[2,3- d]pyrimidin-7(8H)-one (195 mg, 13.7% yield) was found to be 98% pure by HPLC. Ή NMR (400MHz, CDC ) δ 12.97 (br s, IH), 8.35 (s, IH), 7.60 (br s, IH), 7.21 (s, 2H), 6.94 (s, IH), 5.86 (br s, IH), 2.50 (m, 6H), 1.54 (s, 3H), MS (EI) for Ci4H,6N60: 285.0 (MH+).
Example 4
Figure imgf000081_0001
[00236] 3-Chloroperbenzoic acid (0.565 g, 3.27 mmol) was added to a solution of 6-bromo-8-ethyl-4-memyl-2-(methylthio)pyrido[2,3-i/]pyrimidin-7(8H)-one (0.308 g, 0.980 mmol) in dichloromethane (5.0 mL) at room temperature. After 30 minutes, the reaction was diluted with dichloromethane (50 mL) and washed twice with saturated NaHC03, followed by brine. The organic phase was separated and dried over Na2S04, filtered, and concentrated in vacuo. The residue was precipitated with ethyl acetate to provide 8-ethyl-4-methyl-2- (methylsulfonyl)pyrido[2,3-d]pyrimidin-7(8H)-one (302 mg, 89 % yield) as a yellow solid.
Figure imgf000081_0002
[00237] To a stirred solution of (76.5 mg, 0.221 mmol) in 1.5 mL of CH2C12 was added isopropyl amine (709.9 mg, 12.0 mmol, 54 eq.) The reaction was stirred for 15 h at room temperature. The reaction was diluted with CH2C12 and extracted with 2N NaOH, H20, and brine. The organic layer was dried over Na2S04, filtered and concentrated. The crude material was purified using preparative HPLC. Lyophillization of the product containing fractions affored 19.9 mg (27.6 %yield) of 6-bromo-8-ethyl-2-(isopropylamino)-4- methylpyrido[2,3-i/]pyrimidin-7(8H)-one: Ή NMR (400 MHz, CDC13): 5 8.08 (s, 1H), 5.30 (bs, IH), 4.48 (bd, 2H), 4.18 (bs, IH), 2.52 (s, 3H), 1.62 (bs, 3H), 1.29 (m, 9H), MS (EI) for C,3H,7BrN40: 325.2 (MH+).
[00238] Using the same or analogous synthetic techniques and substituting with appropriate reagents, the following compounds were prepared: Example 4b. 6-bromo-2-(te^butylamino)-8-ethyl-4-methylpyrido[2,3-^pyriraidin-7(8H)- one: Ή NMR (400 MHz, CDC13): δ 8.08 (s, IH), 5.47 (bs, IH), 4.48 (m, 2H), 2.50 (s, 3H), 1.58 (bs, 3H), 1.49 (s, 9H), MS (EI) for Cl4Hi9BrN40: 339.2 (MlF)
Example 4c. 6-Bromo-2-(cyclopentylamino)-8-ethyl-4-methylpyrido[2,3--i]pyrimidin-7(8H)- one: Ή NMR (400 MHz, CDC13): 6 8.07 (s, IH), 5.89 (bs, IH), 4.49 (bd, 2H), 2.51 (s, 3H), 2.07 (m, 2H), 1.71 (m, 2H), 1.58 (m, 2H), 1.31 (t, 3H), MS (EI) for C15H,9BrN40: 351.2 (MH*)
Example 4d. 6-Bromo-2-(cyclohexylamino)-8-ethyl-4-methylpyrido[2,3-ii|pyrimidin-7(8H)- one: Ή NMR (400 MHz, CDC13): δ 8.07 (s, IH), 5.41 (bs, IH), 4.47 (bd, 2H), 3.84 (bs, IH), 2.51 (s, 3H), 2.05 (d, 7 = 12.4 Hz, 2H), 1.77 (m, 2H), 1.64 (br m, 4H), 1.39 (m, 2H), 1.30 (m, 3H), MS (EI) for C|6H21BrN40: 365.2 (MH+)
Example 4e. 6-BΓomo-8-ethyl-4-nlethyl-2-(2-mo holinoethylamino)pyΓido[2,3- i ]pyrimidin-7(8H)-one: Ή NMR (400 MHz, CDC13): δ 8.08 (s, IH), 6.22 (bs, IH), 4.48 (q, J = 6.4 Hz, 2H), 3.74 (t, J = 4.4 Hz, IH), 3.57 (q, J = 4.8 Hz, 3H), 2.98 (bs, 2H), 2.63 (t, J = 6.0 Hz, 2H), 2.53 (s, 3H), 1.30 (t, J = 6.8 Hz, 2H), MS (EI) for Ci6H22BrN50: 396.2 (MH+) Example 4f. 6-Bromo-8-ethyl-4-methyl-2-[(3-morpholino-4-ylpropyl)amino]pyrido[2,3- </]pyrimidin-7(8H)-one: Ή NMR (400 MHz, CDC13): 5 8.07 (s, IH), 6.23 (bs, IH), 4.47 (bs, IH), 3.75 (m, 4H), 3.57 (m, 2H), 2.52 (m, 4H), 2.48 (m, 2H), 1.82 (m, 2H), 1.28 (s, 3H), MS (EI) for Cl7H24BrN50: 410.2 (MH4)
Example 4g. 6-Bromo-2-{ [3-(dimethylamino)propyl]amino}-8-ethyl-4-methylpyrido[2,3- £flpyrimidin-7(8H)-one: Ή NMR (400 MHz, CDC13): δ 8.08 (s, IH), 7.26 (bs, IH), 4.47 (m, 2H), 3.54 (m, 2H), 2.78 (t, / = 7.6 Hz, 2H), 2.52 (s, 3H), 2.50 (s, 3H), 2.04 (s, 3H), 2.00 (m, 2H), 1.29 (t, / = 7.2 Hz, 3H), MS (EI) for C,5H22BrN50: 369.2 (MH+)
Example 4h. 8-E l-2-(ethylamino)-4-methylpyrido[2,3-iflpyrirnidin-7(8H)-one: Ή NMR (400 MHz, CDC13): δ 7.67 (d, J = 9.2 Hz, IH), 6.39 (d, J = 9.2 Hz, IH ), 5.31 (bs, IH), 2.54 (s, 3H), 4.32 (q, J = 6.8 Hz, 2H), 3.52 (q, J = 6.8 Hz, 2H), 2.53 (s, 3H), 1.15 (m, 6H); MS (EI) for C12H16N40: 233.2 (MH+).
Example 4j. 6-Bromo-2-{ [2-(dimethylarnino)ethyl]arnino }-8-ethyl-4-rnethylpyrido[2,3- <flpyrirnidin-7(8H)-one: Ή NMR (400 MHz, OMSO-d6): δ 8.37 (s, IH), 7.83 (bt, J = 8.0 Hz, IH), 4.34 (q, J = 8.0 Hz, 2H), 3.42 (q, J = 4.0 Hz, 2H), 2.51 (s, 3H), 2.45 (t, J = 4.0 Hz, 2H), 1.83 (s, 6H), 1.20 (t, J = 8.0 Hz, 3H); MS (EI) for Ci4H20BrN5O: 354.3 (M+).
Example 4k. 6-bromo-2-(ethylamino)-4-methyl-8-( l-methylethyl)pyrido[2,3-i/]pyrimidin- 7(8H)-one: Ή NMR (400 MHz, CDC13): δ 8.04 (s, IH), 6.66 (bs, IH), 5.83 (sept, J = 6.8 Hz, IH), 3.54 (dq, / = 12.8, 7.6 Hz, 2H), 2.62 (s, 3H), 1.60 (d, / = 6.8 Hz, 6H), 1.34 (t, / = 7.2 Hz, 3H); MS (EI) for Ci3H17BrN40: 324.9 (M*).
Example 4m. 6-Bromo-8-e-hyI-4-methyl-2-mo holW-4- lpyrido[2,3-ίflp rimidi one: Ή NMR (400 MHz, CDC13): δ 8.09 (s, IH), 4.45 (q, J = 6.8 Hz, 2H), 3.92 (s, 3H), 3.79 (s, 3H), 2.55 (s, 3H), 1.30 (t, J = 6.8 Hz, 3H); MS (EI) for C,4Hi7Br N402: 355.1 (M2H+). Example 4n. 6-Bromo-8-ethyl-4-methyl-2-[(phenylmethyl)amino]pyrido[2,3-^pyrirnidin- 7(8H)-one: Ή NMR (400 MHz, CDC13): δ 8.09 (s, IH), 7.32 (m, 5H), 5.86 (bs, IH), 4.68 (s, 2H), 4.43 (q, J = 7.2 Hz, 2H), 2.54 (s, 3H), 1.13 (t, J = 7.2 Hz, 3H); MS (EI) for
Ci7Hi7BrN40: 375.1 (M2H^.Example 4p. 6-Bromo-8-emyl-2-(emylamino)-4- methylpyrido[2,3-ii]pyrimidin-7(8H)-one: Ή NMR (400 MHz, CDC13): δ 8.09 (s, IH), 5.71 (bs, IH), 4.48 (bs, 2H), 3.54 (q, J = 6.8 Hz, 2H), 2.53 (s, 3H), 1.16 (m, 6H); MS (EI) for C,2H,5BrN40: 31 1.9 (MH+).
Example 5
2-(Ethylamino)-4-methyl-8 l-methylethylH
one
Figure imgf000083_0001
[00239] Pd(dppf) dichloromethane adduct (0.077 g, 0.095 mmol) was added to a suspension of 6-bromo-2-(ethylamino)-4-methyl-8-(l -methylethyl)pyrido[2,3-if|pyrimidin- 7(8H)-one (0.154 g, 0.474 mmol), 2-thiophene boronic acid (0.079 g, 0.616 mmol), and tnethylamine (165 L, 1.19 mmol) in 10: 1 DME: water ( 1.5 mL). The reaction was heated to 100 °C. After 5 h, the reaction was cooled to room temperature, filtered though a Celite plug and concentrated in vacuo. The residue was purified on Si02 (3:2 hexanes: ethyl acetate) to give 2-(ethylamino)-4-methyl-8-( 1 -methylethyl)-6-(2-thienyl)pyrido[2,3-ii|pyrimidin-7(8H)- one (28 mg, 18 % yield) as a light yellow solid: Ή NMR (400 MHz, CDC13): δ 8.06 (s, IH), 7.60 (dd, J = 4.0, 1.2 Hz, IH), 7.38 (dd, J = 52 , 0.8 Hz, IH), 7.10 (dd, J = 4.8, 3.2 Hz, IH), 5.93 (bsept, IH), 5.13 (bs, IH), 3.54 (pent, / = 7.2 Hz, 2H), 2.61 (s, 3H), 1.66 (d, J = 6.8 Hz, 6H), 1.28 (t, J = 7.6 Hz, 3H); MS (EI) for C17H20N4OS: 329.0 (MIT). [00240] Using the same or analogous synthetic techniques and substituting with appropriate reagents, the following compounds were prepared:
Example 5a. 2-(Ethylamino)-6-furan-2-yl-4-methyl-8-( 1 -methylethyl)pyrido[2,3- </]pyrimidin-7(8H)-one: Ή NMR (400 MHz, CDC13): 5 8.43 (s, IH), 7.81 (s, IH), 7.47 (t, / = 2 Hz, IH), 6.75 (dd, / = 2.0 , 0.8 Hz, IH), 5.92 (bsept, IH), 5.25 (bs, IH), 3.53 (dq, / = 12.5, 7.6 Hz, 2H), 2.60 (s, 3H), 1.65 (d, / = 6.8 Hz, 6H), 1.29 (t, J = 7.2 Hz, 3H); MS (EI) for C17H2oN402: 313.1 (MIT).
Example 5b. 2-(Ethylamino)-4-methyl-8-( 1 -methylethyl)-6-( lH-pyrazol-3-yl)pyrido[2,3- iflpyrimidin-7(8H)-one: Ή NMR (400 MHz, CDC13): δ 8.08 (s, IH), 7.61 (d, J = 2.0 Hz, IH), 6.65 (bs, IH), 5.93 (bs, IH), 5.44 (bs, IH), 3.55 (dq, J = 12.8, 6.4 Hz, 2H), 2.62 (s, 3H), 1.66 (d, J = 6.4 Hz, 6H), 1.30 (t, J = 7.6 Hz, 3H); MS (EI) for C,6H2oN60: 313.3 (MH+). Example 5c. 2-(Ethylamino)-4-methyl-6-( 1 H-pyrazol-3-yl)pyrido[2,3-iflpyrimidin-7(8H)- one: Ή NMR (400 MHz, MeOH^TFA-d, 10: 1): δ 8.59 (s, IH), 8.07 (s, IH), 7.30 (s, IH), 3.59 (q, J = 8.0 Hz, 2H), 2.88 (s, 3H), 1.28 (t, / = 8.0 Hz, 3H); MS (EI) for CBHI^O: 271.0 (MH*).
Example 5e. 8-Cyclopentyl-2-(ethylamino)-4-methyl-6-( lH-pyrazol-3-yl)pyrido[2,3- </]pyrimidin-7(8H)-one: Ή NMR (400MHz, DMSO-^): δ 8.32 (s, IH), 7.80 (s, IH), 7.59 (s, IH), 6.916 (s, IH), 5.95 (m, IH), 2.35 (bs, 2H), 1.95 (bs, 2H), 1.73 (bs, 2H), 1.61 (bs, 2H), 1.12 (t, J = 6.8 Hz, 3H), MS (EI) for C,8w22N60: 339.1 (MH*)
Example 5f. 6-(2,4-Difluorophenyl)-8-ethyl-2-(ethylamino)-4-methylpyrido[2,3- i/]pyrimidin-7(8H)-one: Ή NMR (400 MHz, CDC13): δ 7.78 (d, 2H), 7.52 (m, IH), 6.85 (m, 2H), 5.38 (bs, IH), 4.48 (m, 2H), 3.56 (m, 2H), 2.57 (s, 3H), 1.39 (m, 6H); MS (EI) for
C,8Wi8F2N40: 345.1 (MlF).
Example 5g. 6-(3-Chloro-4-fluorophenyl)-8-ethyl-2-(ethylamino)-4-methylpyrido[2,3- i/]pyrimidin-7(8H)-one: Ή NMR (400 MHz, CDCI3): δ 7.79 (s, 2H), 7.57 (m, IH), 7.19 (m, I H), 5.41 (bs, IH), 4.45 (bs, 2H), 3.58 (m, 2H), 2.59 (m, 3H), 1.36 (m, 6H); MS (EI) for Ci8Hi8ClFN40: 361.0 (MIT).
Example 5h. 6-(2,4-Dichlorophenyl)-8-ethyI-2-(ethylamino)-4-methylpyrido[2,3- df]pyrimidin-7(8H)-one: Ή NMR (400 MHz, CDC13): δ 7.75 (s, IH), 7.42 (d, IH), 7.38 (m, 2H), 5.38 (bs, IH), 4.42 (m, 2H), 3.59 (m, 2H), 2.56 (s, 3H), 1.24 (m, 6H); MS (EI) for C,8//I8C12N40: 377.0 (M+), 379.0 (M+2)
Example 5i. 6-(3,4-Difluorophenyl)-8-ethyl-2-(ethylamino)-4-methylpyrido[2,3- i/]pyrimidin-7(8H)-one: Ή NMR (400 MHz, CDC13): δ 7.79 (s, I H), 7.59 (m, IH), 7.39 (m, IH), 7.18 (m, IH), 5.39 (bs, IH), 4.46 (m, 2H), 3.58 (m, 2H), 2.59 (s, 3H), 1.27 (m, 6H); MS (EI) for C,8Wi8F2N40: 345.1 (MIT).
Example 5j. 8-Ethyl-2-(ethylamino)-4-methyl-6-[4-(phenyloxy)phenyl]pyrido[2,3- iflpyriniidin-7(8H)-one: Ή NMR (400 MHz, CDC13): δ 7.78 (s, IH), 7.63 (d, 2H), 7.39 (t, 2H), 7.16 (t, IH), 7.04 (d, 4H), 5.38 (bs, IH), 4.47 (m, 2H), 3.57 (m, 2H), 2.59 (s, 3H), 1.26 (m, 6H); MS (EI) for C24H24 402: 401.1 (MH+).
Example 5k. 8-Ethyl-2-(ethylamino)-4-methyl-6-naphthaleN- 1 -ylpyrido[2,3-i ]pyrimidin- 7(8H)-one: Ή NMR (400 MHz, CDC13): δ 7.84 (d, 2H), 7.80 (s, IH), 7.73 (d, IH), 7.48 (m, 4H), 539 (bs, IH), 4.55 (bs, 2H), 3.59 (m, 2H), 2.54 (s, 3H), 1.37 (m, 6H); MS (EI) for C22H22N40: 359.1 (MIT).
Example 5m. 8-Ethyl-2-(ethylamino)-4-methyl-6-[3-(trifluoromethyl)phenyl]pyrido[2,3- ii]pyrimidin-7(8H)-one: Ή NMR (400 MHz, CDCI3): 5 7.82 (m, 3H), 7.56 (m, 2H), 5.59 (bs, IH), 4.47 (d, 2H), 3.51 (m, 2H), 2.58 (s, 3H), 1.30 (m, 6H); MS (EI) for Ci9Hi9F3N40: 377.1 (MH+).
Example 5n. 8-Ethyl-2-(ethylamino)-4-methyl-6-(2-t_hienyl)pyrido[2,3-rf]pyrimidin-7(8H)- one: Ή NMR (400 MHz, CDC13): δ 8.09 (s, IH), 7.64 (dd, J = 3.60, 1.20 Hz, IH), 7.38 (dd, / = 5.20, 1.20 Hz, IH), 7.10 (dd, / = 4.78, 3.60 Hz, 2H), 3.54 (qn, 2H), 2.62 (s, 3H), 1.30 (m, 6H); MS (EI) for C|6H18N4OS: 315.0 (MIT).
Example 5p. 6-(3-Chlorophenyl)-8-ethyl-2-(ethylamino)-4-methylpyrido[2,3-<flpyrimidin- 7(8H)-one: Ή NMR (400 MHz, CDC13): δ 7.78 (s, IH), 7.65 (s, IH), 7.56 (dd, I H), 7.34 (m, 2H), 5.39 (bs, IH), 4.43 (m, 2H0, 3.57 (m, 2H), 2.59 (s, 3H), 1.32 (m, 6H); MS (EI) for C18m9ClN40: 343.0 (MH+).
Example 5q. 6-(4-Chlorophenyl)-8-ethyl-2-(ethylarnino)-4-methylpyrido[2,3-if|pyrimidin- 7(8H)-one: Ή NMR (400 MHz, CDCI3): δ 7.77 (s, IH), 7.62 (dd, 2H0, 7.40 (dd, 2H), 5.38 (bs, IH), 4.47 (m, 2H), 3.58 (m, 2H), 2.59 (s, 3H), 1.39 (m, 6H); MS (EI) for C,8Wi9ClN40: 343.0 (MH+).
Example 5r. 8-E l-2-(ethylamino)-4-methyl-6-[4-(trifluorornethyl)phenyI]pyrido[2,3- <*]pyrimidin-7(8H)-one: Ή NMR (400 MHz, CDC13): δ 7.80 (m, 3H), 7.63 (dd, 2H), 5.39 (bs, IH), 4.51 (m, 2H), 3.58 (m, 2H), 2.58 (s, 3H), 1.33 (m, 6H); MS (EI) for C|9H,9F3N40: 343.0 (MH+).
Example 5s. 8-Ethyl-2-(ethylamino)-4-methyl-6-(3-thienyl)pyrido[2,3-^pyrimidin-7(8H)- one: Ή NMR (400 MHz, CDCI3): 6 8.1 1 (dd, J = 2.10, 0.90 Hz, IH), 7.94 (s, IH), 7.52 (dd, J = 3.90, 1.20 Hz, IH), 7.35 (qr, I H), 5.33 (bs, IH), 4.52 (qr, 2H), 3.54 (m, 2H), 2.58 (s, 3H), 1.28 (m, 6H); MS (EI) for Ci6H18N4OS: 315.0 (MH+).
Example 5t. 8-Ethyl-2-(ethylamino)-4-methyl-6-(4-methyl-2-thienyl)pyrido[2,3- ^pyrimidin-7(8H)-one: Ή NMR (400 MHz, CDC13): δ 8.01 (s, IH), 7.52 (s, IH), 6.93 (s, IH), 5.38 (bs, IH), 4.58 (qr, 2H), 3.57 (m, 2H), 2.61 (s, IH), 2.33 (s, IH), 1.60 (s, 3H); MS (EI) for C|7H20N4OS: 329.0 (MH+).
Example 5u. 8-Ethyl-2-(ethylamino)-4-methyl-6-(4-methyl-3-thienyl)pyrido[2,3- rf]pyrimidin-7(8H)-one: Ή NMR (400 MHz, CDC13): δ 7.69 (s, IH), 7.38 (d, IH), 6.99 (m, IH), 5.35 (bs, IH), 4.51 (qr, 2H), 3.57 (m, 2H), 2.58 (s, 3H), 2.22 (s, 3H), 1.32 (m, 6H); MS (EI) for C17H20N4OS: 329.0 (MH+).
Example 5v. 1,1-Dimethylethyl 2-[8-ethyl-2-(ethylamino)-4-methyl-7-oxo-7,8- dihydropyrido[2,3-rf]pyrimidin-6-yl]-lH-pyrrole-l-carboxylate: Ή NMR (400 MHz, CDC13): 67.65 (s, IH), 7.38 (d, IH), 6.22 (m, 2H), 5.29 (bs, IH), 4.41 (m, 2H), 3.57 (m, 2H), 2.56 (s, 3H), 1.41 (s, 9H), 1.22 (m, 6H); MS (EI) for C2,H27N503: 398.0 (MH+).
Example 5w. 8-Ethyl-2-(ethylamino)-4-methyl-6-( 1 H-pyrrol-2-yl)pyrido[2,3-ii]pyrimidin- 7(8H)-one: *H NMR (400 MHz, CDC13): 6 1 1.1 (bs, IH), 7.99 (s, IH), 6.85 (d, IH), 6.62 (d, IH), 6.29 (d, IH), 5.28 (bs, IH), 4.57 (m, 2H), 3.56 (m, 2H), 2.61 (s, 3H), 1.35 (m, 6H); MS (EI) for C|6Hi9N50: 298.1 (MH+).
Example 5x. 8-Ethyl-2-(ethylamino)-6-furan-3-yl-4-methylpyrido[2,3-i/]pyrimidin-7(8H)- one: Ή NMR (400 MHz, CDC13): 6 8.42 (s, IH), 7.83 (s, IH), 7.43 (s, 1H),6.76 (s, IH), 5.37 (bs, IH), 4.52 (m, 2H), 3.58 (m, 2H), 2.61 (s, 3H), 1.30 (m, 6H); MS (EI) for C16Hi8N402: 299.1 (MH+).
Example 5y. 8-Ethyl-2-(ethylamino)-4-methyl-6-[ l-(phenylmethyl)-lH-pyrazol-4- yl]pyrido[2,3-i/]pyrimidin-7(8H)-one: Ή NMR (400 MHz, CDCI3): 6 8.39 (s, IH), 7.98 (d, IH), 7.96 (d, IH), 7.35 (m, 5H), 5.39 (s, 2H), 5.35 (bs, IH), 4.52 (m, 2H), 3.58 (m, 2H), 2.62 (s, 3H), 1.35 (m, 6H) ; MS (EI) for C22H24 60: 389.3 (MH+).
Example 5z. 6-(3,5-Dirnethylisoxazol-4-yl)-8-ethyl-2-(ethylamino)-4-methylpyrido[2,3- iflpyrimidin-7(8H)-one: Ή NMR (400 MHz, CDC13): 6 7.59 (s, IH), 7.24 (s, IH), 5.43 (bs, I H), 4.47 (bs, 2H), 3.56 (m, 2H), 2.58 (s, 3H), 2.39 (s, 3H), 2.25 (s, 3H), 1.29 (m, 6H) ; MS (EI) for C17H21N502: 328.1 (MH+).
Example 5aa. 8-Ethyl-2-(ethylamino)-4-methyl-6-(lH-pyrazol-5-yl)pyrido[2,3-ii|pyrirriidin- 7(8H)-one: Ή NMR (400 MHz, CDC13): 6 8.1 1 (s, IH), 7.62 (s, IH), 6.65 (d, IH), 5.43 (bs, IH), 4.58 (m, 2H), 3.59 (m, 2H), 2.62 (s, 3H), 1.38 (m, 6H); MS (EI) for d5H18N60: 299.1 (MH+).
Example 5bb. 8-Ε 1-4-Γη6 1-6-(1^ργΓ3ζο1-5- 1)-2-[(2,2,2- trifluoroethyl)arnino]pyrido[2,3--i]pyrimidin-7(8H)-one: Ή NMR (400 MHz, CDC13): δ 8.18 (s, IH), 7.63 (d, IH), 6.73 (d, IH), 5.62 (bs, IH), 4.58 (m, 2H), 4.30 (m, 2H), 2.74 (s, 3H), 1.35 (t, 3H); MS (EI) for C15Hi5F3 60: 353.0 (MH+).
Example 5cc 8-Ethyl-2-(ethylammo)-4-methyl-6-(1
7(8H)-one: 1H NMR (400 MHz, CDC13): δ 8.87 (s, IH), 7.98 (s, IH), 7.43 (s, IH), 7.22 (s, IH), 5.56 (bs, IH), 4.58 (bs, 2H), 2.72 (s, 3H0, 1.36 (m, 6H); MS (EI) for Ci5Hi7N5OS: 316.0 (MET").
Example 6
6-BiphenyI-4-vI-8-ethyl-2-(ethylamino)^^
Figure imgf000087_0001
[00241] 2-Ethylamino-6-bromo-8-ethyl-4-methylpyrido[2,3-i/]pyrimidin-7(8H)-one (60 mg, 0.194 mmol), K2C03 (81.0 mg, 3.0 equiv.), biphenyl boronic acid (17.8 mg, 1.5 equiv.) and Pd(PPh3)4 (10 mol %, 225 mg) were added to dioxane / H20 ( 10 mL / 3 mL). The reaction was heated to 95 °C and stirred for 2 h. The reaction mixture was partitioned between organic and aqueous layers with ethyl acetate (20 mL) and H20 ( 10 mL) and saturated aqueous NaCl (5 mL). The organic layer was dried over anhydrous magnesium sulfate, filtered and evaporated to give 6-Biphenyl-4-yl-8-ethyl-2-(ethylamino)-4- methylpyrido[2,3-i ]pyridimiduV-7(8H)-one (48.42 mg, 65 % yield): Ή NMR (400 MHz, CDC13): δ 7.81 (s, IH), 7.74 (m, 2H), 7.60 (m, 4H), 7.42 (m, 2H), 7.38 (m, IH), 4.50 (q, 2H), 3.60 (q, 2H), 2.60 (s, 3H), 1.30 (m, 6H); MS (EI) for C24H24N40: 385.1 (MH+).
[00242] Using the same or analogous synthetic techniques and substituting with appropriate reagents, the following compounds were prepared:
Example 6a. 8-Ethyl-2-(ethylamino)-4-methyl-6-[4-(methyloxy)phenyl]pyrido[2,3- £/]pyridimidW-7(8H)-one: Ή NMR (400 MHz, CDC13): 6 7.81 (s, IH), 7.60 (d, 2H), 6.96 (d, 2H), 4.50 (q, 2H), 3.82 (s, 3H), 3.58 (q, 2H), 2.58 (s, 3H), 1.30 (m, 6H); MS (EI) for Ci9H22N402: 339.1 (MH*). Example 6b. 8-Ethy l-2-(ethylamino)-4-methyl-6-[2-(methyloxy)phenyl]pyrido[2,3- rf]pyridimidiN-7(8H)-one: Ή NMR (400 MHz, CDC13): δ 7.81 (s, IH), 7.60 (d, 2H), 6.96 (d, 2H), 4.50 (q, 2H), 3.80 (s, 3H), 3.58 (q, 2H), 2.50 (s, 3H), 1.30 (m, 6H); MS (EI) for
Ci9H22N402: 339.1 (MlT).
Example 6c. 6-[2,4-Bis(methyloxy)phenyl]-8-ethyl-2-(ethylamino)-4-methylpyrido[2,3- </]pyrimidin-7(8H)-one: Ή NMR (400 MHz, CDCI3): 5 7.70 (s, IH), 7.30 (s, IH), 6.60 (m, 2H), 4.50 (q, 2H), 3.82 (s, 3H), 3.80 (s, 3H), 3.45 (q, 2H), 2.50 (s, 3H), 1.30 (m, 6H); MS (EI) for C2oH24N 03: 369.1 (MH+).
Example 6d. 8-Ethy l-2-(ethylamino)-4-methyl-6-[3-(methyloxy)phenyl]pyrido[2,3- .flpyridimidiN-7(8H)-one: Ή NMR (400 MHz, CDC13): δ 7.81 (s, IH), 7.60 (d, 2H), 6.96 (d, 2H), 4.50 (q, 2H), 3.80 (s, 3H), 3.58 (q, 2H), 2.50 (s, 3H), 1.30 (m, 6H); MS (EI) for
Ci9H22N402: 339.1 (MET).
Example 6e. 8-(5-Chloro-2-thienyl)-8-ethyl-2-(ethylamino)-4-methylpyrido[2,3-rf]pyrimidin- 7(8H)-one: Ή NMR (400 MHz, CDC13): δ 8.00 (s, IH), 7.38 (d, 2H), 6.96 (d, 2H), 4.50 (q, 2H), 3.58 (q, 2H), 2.60 (s, 3H), 1.30 (m, 6H); MS (EI) for C16H|7ClN4OS: 349.2 (MH+). Example 6f. 8-Ethy 1 -2-(ethylamino)-4-methyl-6-pyrirnidin-5-ylpyrido[2,3-ii|pyridimidiN- 7(8H)-one: Ή NMR (400 MHz, DMSO-d6) δ 9.19 (s, IH), 9.16 (s, IH), 8.23 (s, IH), 8.00 (m, IH), 4.38 (q, 2H), 3.40 (q, 2H), 2.50 (s, 3H), 1.30 (m, 6H); MS (EI) for C16H,8N60: 311.3 (MET).
Example 6g. 8-Ethy 1 -2-(ethylamino)-6-(3-fluoropyridiN-4-yl)-4-methylpyrido[2,3- d]pyrimidin-7(8H)-one: Ή NMR (400 MHz, CDC13): δ 8.58 (s, IH), 8.42 (d, IH), 7.98 (s, IH), 7.60 (t, IH), 4.50 (q, 2H), 3.58 (q, 2H), 2.60 (s, 3H), 1.30 (m, 6H); MS (EI) for
C17H18FN50: 328.3 (MH+).
Example 6h. 8-Ethy l-2-(ethylamino)-6-( lH-indole-6-yl)-4-methylpyrido[2,3-i/|pyrimidin- 7(8H)-one: Ή NMR (400 MHz, DMSO-<¾: δ 1 1.2 (s, IH), 7.90 (s, IH), 7.88 (s, IH), 7.42 (s, 2H), 7.38 (s, IH), 6.50 (s, IH), 4.40 (q, 2H), 3.40 (q, 2H), 2.42 (s, 3H), 1.30 (m, 6H); MS (EI) for C20H2iN5O: 348.3 (MH+).
Example 6i. 8-Ethy 1 -2-(ethylamino)-4-methyl-6-(5-phenyl-2-thienyl)pyrido[2,3- <flpyrimidin-7(8H)-one: Ή NMR (400 MHz, DMSO-rf6): δ 8.40 (s, IH), 7.81 (d, IH), 7.70 (d, 2H), 7.50 (d, IH), 7.42 (m, 2H), 7.30 (m, IH), 4.40 (q, 2H), 3.40 (q, 2H), 2.42 (s, 3H), 1.30 (m, 6H); MS (EI) for C22H22N4OS: 3 1.3 (MH+). Example 6j. 8-Ethyl-2-(ethylamino)-4-me l-6-phenylpyrido[2,3-^pyrimidin-7(8H)-one: Ή NMR (400 MHz, CDC13): 57.78 (s, 1H), 7.46 (m, 5H), 5.41 (bs, 1H), 4.50 (q, J = 6.8 Hz, 2H), 3.60 (m, 2H), 2.57 (s, 3H), 1.30 (m, 6H); MS (EI) for Cl SH20 N40: 309.2 (MH+).
Example 6k. 8-Ethyl-2-(ethylamino)-6-(3-fluorophenyl)-4-methylpyrido[2,3-ii|pyrimidin- 7(8H)-one: Ή NMR (400 MHz, CDCI3): 5 7.79 (s, 1H), 7.46-7.02 (m, 4H), 5.41 (bs, 1H),
4.51 (q, J = 6.4 Hz, 2H), 3.55 (q, J = 6.8 Hz, 2H), 2.58 (s, 3H), 1.34 (t, J = 6.80 Hz, 3H), 1.29 (t, J = 6.40 Hz, 3H); MS (EI) for C18H19FN40: 327.3 (MH+).
Example 6m. 8-ethyl-2-(ethylamino)-6-(2-fluorophenyl)-4-methylpyrido[2,3-£ilpyrimidin- 7(8H)-one: Ή NMR (400 MHz, CDC13): 5 7.80 (s, 1H), 7.52-7.12 (m, 4H), 5.33 (bs, 1H), 4.49 (q, J = 6.8 Hz, 2H), 3.53 (q, J = 1.2 Hz, 2H), 2.55 (s, 3H), 1.34 (t, J = 7.20 Hz, 3H),
1.28 (t, / = 6.80 Hz, 3H); MS (EI) for C18//19FN4O: 327.3 (MH+).
Example 6n. 8-ethyl-2-(etJiylamino)-6-(4-fluorophenyl)-4-methylpyrido[2,3-if|pyrimidin- 7(8H)-one: Ή NMR (400 MHz, CDC13): 5 7.75 (s, 1H), 7.66-7.08 (m, 4H), 5.30 (bs, 1H),
4.52 (q, J = 6.4 Hz, 2H), 3.54 (q, J = 6.8 Hz, 2H), 2.58 (s, 3H), 1.34 (t, J = 6.80 Hz, 3H),
1.29 (t, J = 6.40 Hz, 3H); MS (EI) for Ci8/,i9FN40: 327.3 (MH+).
Intermediate 2
Figure imgf000089_0001
[00243] 3-Chloroperbenzoic acid (1.78 g, 10.4 mmol) was added to a solution of 6-bromo-4-methyl-8-( l-me lethyl)-2-(metnylthio)pyrido[2,3-<i]pyrimidin-7(8H)-one ( 1.33 g, 4.14 mmol), prepared using procedures similar to those described in Example 1, in dichloromethane (30.0 mL) at room temperature. After 1, the reaction was diluted with dichloromethane (50 mL) and washed twice with saturated NaHCC>3, followed by brine. The organic phase was separated and dried over Na2S04, filtered, and concentrated in vacuo. The residue was precipitated with ethyl acetate/hexanes to provide the corresponding sulfone ( 1.31 g, 93 % yield) as an off-white solid. Example 8
2-Amino-4-methyl-8-(phenylmethyl)-6 lH^
one
Figure imgf000090_0001
[00244] Triethylamine (3.4 mL, 24.6 mmol) was added to a suspension of 2-amino-4- chloro-6-methylpyrimidine (Aldrich, 1.77 g, 12.3 mmol) and benzylamine ( 1.98 g, 18.5 mmol) in anhydrous dioxane (20 mL). The reaction was heated to 80 °C and allowed to run for 12 h. Upon cooling to room temperature, a white precipitate formed which was collected by vacuum filtration. The solid was recrystallized from acetone: hexanes to afford N'-benzyl- 6-methylpyrimidine-2, -diamine (2.33 g, 89 % yield) as a white solid.
Figure imgf000090_0002
[00245] Iodine (3.04 g, 12.0 mmol) was added to a solution of N*-benzyl-6- methylpyrimidine-2,4-diamine (2.33 g, 10.9 mmol) in anhydrous MeOH (50 mL) at 0 °C. The reaction was allowed to warm to room temperature overnight. After 12 hours, an additional 0.5 equiv of iodine was added, and the reaction warmed to 50 °C. After four hours, the reaction was cooled to room temperature and concentrated in vacuo. The residue was diluted with ethyl acetate (200 mL) and washed with 10% NaHS03 (200 mL). The aqueous phase was separated and washed once more with ethyl acetate (200 mL). The organic phases were combined, washed with brine, separated and dried over Na2S04, The filtrate was concentrated in vacuo to afford the product A/4-benzyl-5-iodo-6-methylpyrimidine-2,4- diamine (3.14 g, 85 % yield).
Figure imgf000091_0001
[00246] Triethylamine (7.60 mL, 54.5 mmol) was added to a suspension of N*-benzyl- 5-iodo-6-methylpyrimidine-2,4-diamine (3.14 g, 10.9 mmol), ethyl acrylate (3.55 mL, 32.7 mmol) and Pd(PPh3)4 (629 mg, 0.545 mmol) in anhydrous DMF (20 mL). The reaction was heated to 95 °C under nitrogen. After 24 h, the reaction was allowed to cool to room temperature and concentrated in vacuo. The residue was poured into a 10% solution of LiCl and washed with ethyl acetate (100 mL). The organic phase was separated and washed with brine, separated and dried over Na2SC>4. The filtrate was concentrated in vacuo and purified on Si02 (3:2 methylene chloride: ethyl acetate) to afford (E)-ethyl-3-(2-amino-4- (benzylamino)-6-methyIpyrimidin-5-yl)acrylate (0.954 g, 28 % yield) as a light yellow solid.
Figure imgf000091_0002
[00247] 2-amino-4-methyl-8-(phenylmethyl)pyrido[2,3-£Qpyrimidin-7(8H)-one Diazabicyclo[5.4.0]undec-7-ene (DBU) (1.83 mL, 12.2 mmol) was added to a flask charged with (£)-ethyl-3-(2-amino-4-(benzylamino)-6-methylpyrimidin-5-yl)acrylate (0.954 g, 3.05 mmol) and the reaction refluxed at 160 °C under a nitrogen atmosphere. After 20 hours, the reaction was cooled to room temperature and concentrated in vacuo. Purification on Si02 (1 : 1 methylene chloride: ethyl acetate) afforded the product (0.508 g, 62 % yield) as an off-white solid.
Figure imgf000092_0001
[00248] Bromine (72 |iL, 1.40 mmol) was added to a suspension of 2-amino-4-methyl-
8-(phenylmethyl)pyrido[2,3-ii|pyrimidin-7(8H)-one (0.340 g, 1.27 mmol) in methylene chloride (20 mL) at 0 °C. The reaction was allowed to warm to room temperature over one hour and the resulting precipitate collected by vacuum filtration to afford 2-amino-6-bromo- 4-methyl-(8-phenylmethyl)pyrido[2,3-ii]pyrimidin-7(8H)-one (0.435 g, 99 % yield) after drying. The yellow solid was used in the next step without further purification.
Figure imgf000092_0002
[00249] A 10: 1 solution of dioxane and water ( 1 1 mL) was added to a flask charged with 2-amino-6-bromo-4-methy l-(8-pheny lmethyl)pyrido[2,3-ti] pyrimidin-7(8H)-one (0.435 g, 1.27 mmol), lH-pyrazole-5-boronic acid (0.284 g, 2.54 mmol), Pd(PPh )4 (0.073 mg, 0.063 mmol), and K2C03 (0.527 g, 3.81 mmol). The flask was flushed with nitrogen and fitted with a reflux condenser and heated to 1 10 °C. After 12 h the reaction was cooled to room temperature and diluted with ethyl acetate (100 mL) and washed with water. The aqueous phase was acidified to pH 1.0 and washed with ethyl acetate (100 mL). The organic phases were combined and washed with brine, separated and dried over Na2SC>4, filtered and concentrated in vacuo. The residue was precipitated with ethyl acetate to give 2-Amino-4- methyl-8-(phenylmethyl)-6-(lH-pyrazol-3-yI)pyrido[2,3-i/]pyrimidin-7(8H)-one (0.062 g, 15 % yield) as a yellow solid: Ή NMR (400 MHz, DMSO- 6): δ 13.10 (bs, IH), 12.93 (bs, IH), 8.47 (s, IH), 7.76 (bs, IH), 7.51 (bs, IH), 7.28 (m, 5H), 6.97 (s, IH), 5.55 (s, 2H), 2.55 (bs, 3H); MS (EI) for Ci8H16N60: 333.1 (MH+). Example 9
2-Amino- -ethyl-4-methyl-6 4-methyl-3-thienyI)pyrido[2 Ipyrimidi
Figure imgf000093_0001
[00250] A 3: 1 solution of dioxane and water (4 mL) was added to a flask charged with 2-amino-6-bromo-8-e I-4-methyIpyrido[2,3- |pyrimidin-7(8H)-one (0.140 g, 0.495 mmol) from above, 4-methylthiophene-3-boronic acid (0.140 g, 0.989 mmol), Pd(PPh3)4 (0.057 mg, 0.050 mmol), and K2C03 (0.205 g, 1.48 mmol). The flask was flushed with nitrogen and Fitted with a reflux condenser and heated to 100 °C. After 12 hours the reaction was cooled to room temperature and diluted with ethyl acetate (70 mL) and washed with water. The aqueous phase was separated and washed with an additional amount of ethyl acetate (70 mL). The organic phases were combined and washed with brine, separated and dried over Na2S04, filtered and concentrated in vacuo. The residue was purified on Si02 (1 : 1 methylene chloride: ethyl acetate) to give 2-Amino-8-ethyl-4-methyl-6-(4-methyl-3-thienyI)pyrido[2,3- i/]pyrimidin-7(8H)-one (0.081 g, 55 % yield) as an off-white solid: Ή NMR (400 MHz, DMSO-rfo): δ 7.84 (s, 1H), 7.46 (d, J = 4.0 Hz, 1H), 7.19 (m, 3H), 4.32 (q, J = 8.0 Hz, 2H), 2.52 (s, 3H), 2.1 1 (bs, 3H), 1.19 (t, J = 8.0 Hz, 3H); MS (EI) for C,5H16N4OS: 301.1 (MH+).
[00251] Using the same or analogous synthetic techniques and substituting with appropriate reagents, the following compounds were prepared:
Example 9a. 2-Amino-8-emyl-4-memyl-6-(3-thienyl)pyrido[2,3-ii]pyrimidin-7(8H)- one: Ή NMR (400 MHz, CDC13): 5 8.1 1 (dd, / = 2.8, 1.2 Hz, 1H), 7.95 (s, 1H), 7.51 (dd, J = 5.2, 1.2 Hz, 1H), 7.37 (dd, / = 4.8, 3.2 Hz, 1H), 5.21 , (bs, 2H), 4.48 (q, / = 6.8 Hz, 2H), 2.63 (s, 3H), 1.32 (t, / = 7.2 Hz, 3H); MS (EI) for C14H14N4OS: 287.0 (MH* .
Example 9b. 2-Amino-8-emyl-6-furan-3-yl-4-methylpyrido[2,3-£f|pyrimidin-7(8H)-one: Ή NMR (400 MHz, CDC13): δ 8.47 (bs, 1H), 7.85 (s, 1H), 7.49 (t, J = 1.6 Hz, 1H), 6.77 (dd, J = 2.0, 0.8 Hz, 1H), 5.19, (bs, 2H), 4.48 (q, J = 6.8 Hz, 2H), 2.64 (s, 3H), 1.31 (t, / = 7.2 Hz, 3H); MS (EI) for d4H14N402: 271.1 (MH+).
Example 9c. 2-Amino-6-(3,5-dimethylisoxazol-4-yl)-8-ethyl-4-methylpyrido[2,3- i/]pyrimidin-7(8H)-one: Ή NMR (400 MHz, CDC13): δ 7.62 (s, 1H), 5.27, (bs, 2H), 4.44 (q, J = 7.2 Hz, 2H), 2.59 (s, 3H), 2.38 (s, 3H), 2.25 (s, 3H), 1.31 (t, J = 6.8 Hz, 3H); MS (EI) for C,5HnN502: 300.1 (MH+).
Example 9d. 2-Amino-8-ethyl-6-isoxazol-4-yl-4-methylpyridot2,3-iflpyrimidin-7(8H)-one: Ή NMR (400 MHz, CDC13): δ 9.36 (s, 1 H), 8.71 (s, 1H), 7.91 (s, 1H), 5.30, (bs, 2H), 4.48 (q, / = 7.2 Hz, 2H), 2.67 (s, 3H), 1.32 (t, J = 6.8 Hz, 3H); MS (EI) for C13H! 3N502: 272.0 (MH+).
Example 9e. 2-Amino-8-ethyl-6-furan-2-yl-4-methylpyrido[2,3-iiIpyrirnidin-7(8H)-one: Ή NMR (400 MHz, CDC13): δ 8.19 (s, 1H), 7.48 (d, 7 = 0.8 Hz, 1H), 7.37 (d, J = 3.6 Hz, 1H), 6.53 (dd, J = 3.6, 2.0 Hz 1H), 5.21 , (bs, 2H), 4.48 (q, / = 7.2 Hz, 2H), 2.66 (s, 3H), 1.32 (t, J = 6.8 Hz, 3H); MS (EI) for Ci4H|4N402: 271.0 (MH+).
Example 9f. 5-(2-Amino-8-ethyl-4-memyl-7-oxo-7,8-dihydropyrido[2,3-rf]pyrirnidin-6- yl)thiophene-2-carbonitrile: Ή NMR (400 MHz, CDC13): 5 8.24 (s, 1H), 7.61 (d, J = 4.4 Hz, 1H), 7.55 (d, 7 = 4.4 Hz, 1H), 5.33, (bs, 2H), 4.48 (q, / = 7.2 Hz, 2H), 2.68 (s, 3H), 1.33 (t, J = 6.8 Hz, 3H); MS (EI) for C15H,3N5OS: 312.0 (MH+).
Example 9g. 2-Amino-8-ethyl-4-methy l-6-( lH-pyrazol-4-yl)pyrido[2,3-iflpyrimidin-7(8H)- one: Ή NMR (400 MHz, DMSO-J6): δ 12.88 (s, 1H), 8.38 (s, 1H), 8.17 (s, 2H), 7.10 (bs, 2H), 4.35 (q, J = 7.2 Hz, 2H), 2.59 (s, 3H), 1.20 (t, J = 7.2 Hz, 3H); MS (EI) for C^Hi^O: 271.0 (MH+).
Example 9h. 2-Amino-8-ethyl-4-methyl-6-( 1 ,3-thiazol-2-yl)pyrido[2,3-iflpyrimidin-7(8H)- one: Ή NMR (400 MHz, CDC13): 6 8.94 (s, 1H), 7.94 (d, J = 3.2 Hz, 1H), 7.46 (d, J = 3.2 Hz, 1H), 5.34 (bs, 2H), 4.54 (q, J = 7.2 Hz, 2H), 2.73 (s, 3H), 1.35 (t, J = 7.2 Hz, 3H); MS (EI) for C|3Hl3N5OS: 288.0 (MH+).
Example 9i. 2-Amino-8-ethyl-4-methyl-6-( 1 -methyl- lH-pyrroI-2-yl)pyrido[2,3-if]pyrimidin- 7(8H)-one: [H NMR (400 MHz, DMSO-i 6): δ 7.81 (s, 1H), 7.20 (bs, 2H), 6.81 6.1 1 (dd, J = 3.6, 2 .0Hz, 1H), 6.02 (t, J = 3.2 Hz, 1H), 4.32 (q, J = 7.2 Hz, 2H), 3.49 (s, 3H), 2.52 (s, 3H), 1.19 (t, / = 7.2 Hz, 3H); MS (EI) for C|5H17N50: 284.1 (MH+).
Example 9j. 2-Amino-8-ethyl-4-methyl-6-phenylpyrido[2,3-ii|pyrimidin-7(8H)-one: 1H NMR (400MHz, CDC13): δ 7.79 (s, 1H), 7.65 (d, J = 6.8 Hz, 2H), 7.43 (d, J = 7.2 Hz, 2H), 7.36 (d, J = 7.2 Hz, 1H), 5.24 (bs, 2H), 4.47 (q, J = 7.2 Hz, 2H), 2.60 (s, 3H), 1.31 (d, / = 7.2 Hz, 3H), MS (EI) for Ci6H,6N40: 281.2 (MH+)
Example 9k 2-Amino-8-ethyl-6-(4-methoxyphenyl)-4-methylpyrido[2,3-i/]pyrimidin-7(8H)- one: Ή NMR (400MHz, CDC13): δ 7.75 (s, 1H), 7.62 (d, / = 8.8 Hz, 2H), 6.96 (d, J = 8.8 Hz, 2H), 5.17 (bs, 2H), 4.47 (q, J = 6.8 Hz, 2H), 3.85 (s, 3H), 2.60 (s, 3H), 1.31 (d, J = 7.2 Hz, 3H), MS (EI) for C17H18N402: 31 1.2 (MH+)
Example 9m 2-Amino-8-ethyl-6-(2-methoxyphenyl)-4-methylpyrido[2,3-^pyrimidin-7(8H)- one: Ή NMR (400MHz, CDC13): 5 7.75 (m, 1H), 7.36 (m, 2H), 7.01 (m, 2H), 5.20 (bs, 2H), 4.45 (m, 2H), 3.82 (s, 3H), 2.56 (s, 3H), 1.31 (m, 3H), MS (EI) for Cl7Hl8N402: 31 1.2 (MH+) Example 9n 2-Amino-6-(4-chlorophenyl)-8-ethyl-4-methylpyrido[2,3-iflpyrirnidin-7(8H)- one: Ή NMR (400MHz, CDC13): δ 7.78 (s, 1H), 7.61 (d, J = 8.8 Hz, 2H), 7.39 (d, / = 8.8 Hz, 2H), 5.23 (bs, 2H), 4.46 (q, J = 7.2 Hz, 2H), 2.61 (s, 3H), 1.31 (d, J = 6.8 Hz, 3H), MS (EI) for CeH^CUS O: 315.1 (MIT)
Example 9p 2-Amino-6-(3-chlorophenyl)-8-ethyl-4-methylpyrido[2,3-if]pyriniidin-7(8H)- one: Ή NMR (400MHz, CDC13): δ 7.79 (s, 1H), 7.66 (m, 1H), 7.56 (m, 1H), 7.35 (m, 2H), 5.25 (bs, 2H), 4.46 (q, J = 5.6 Hz, 2H), 2.61 (s, 3H), 1.31 (d, J = 7.2 Hz, 3H), MS (EI) for Ci6Hi5ClN40: 315.1 (MH+)
Example 9q 2-Amino-6-(2-chlorophenyl)-8-ethyl-4-methylpyrido[2,3-<i]pyrirnidin-7(8H)- one: Ή NMR (400MHz, CDCI3): 5 7.75 (s, 1H), 7.67 (m, 1H), 7.54 (m, 2H), 7.38 (m, 1H), 7.333 (m, 1H), 5.22 (bs, 2H), 4.46 (q, J = 6.8 Hz, 2H), 2.57 (s, 3H), 1.31 (d, J = 6.8 Hz, 3H), MS (EI) for C16H15CIN4O: 315.1 (MlT)
Example 9r 2-Amino-6-(2,4-dichlorophenyl)-8-ethyl-4-methylpyrido[2,3-ii|pyrirnidin- 7(8H)-one: Ή NMR (400MHz, CDC13): 5 7.77 (s, 1H), 7.67 (m, 1H), 7.49 (m, 1H), 7.32 (m, 1H), 5.24 (bs, 2H), 4.45 (q, J = 6.8 Hz, 2H), 2.58 (s, 3H), 1.30 (d, / = 7.2 Hz, 3H), MS (EI) for C16H,4Cl2N40: 349.1 (MH+)
Example 9t 2-Amino-8-ethyl-4-methyl-6-(2-thienyl)pyrido[2,3--i]pyrimidin-7(8H)-one: Ή NMR (400 MHz, OMSO-d6): 5 8.39 (s, 1H), 7.85-7.13 (m, 5H), 4.37 (q, J = 7.2 Hz, 2H), 2.62 (s, 3H), 1.18 (t, = 7.2 Hz, 3H); MS (EI) for C4H14N4OS: 287.1 (MH+).
Example 9u 2-Amino-8-ethyl-6-(4-fluorophenyl)-4-methylpyrido[2,3-rf]pyrirnidin-7(8H)- one: 'Η NMR (400 MHz, DMSO- ): δ 7.99 (s, 1H), 7.76-7.22 (m, 6H), 4.34 (q, J = 7.2Hz, 2H), 2.56 (s, 3H), 1.20 (t, J = 7.2 Hz, 3H); MS (EI) for C16H,5FN40: 299.2 (MtT).
Example 9v 2-Amino-8-ethyl-6-(3-fluorophenyl)-4-methylpyrido[2,3-flnpyrimidin-7(8H)- one: 1H NMR (400 MHz, DMSO- ): δ 8.06 (s, 1H), 7.61-7.44 (m, 3H), 7.29 (bs, 2H), 7.20- 7.15 (m, 1H), 4.34 (q, / = 7.2Hz, 2H), 2.58 (s, 3H), 1.20 (t, J = 7.2 Hz, 3H); MS (EI) for C16H15FN40: 299.2 (MH+). Example 9w 2-Amino-8-emyl-6-(2-fluorophenyl)-4-memylpyrido[2,3-./|pyrirnidin-7(8H)- one: Ή NMR (400 MHz, DMSO- ): δ 7.96 (s, tH), 7.50-7.23 (m, 6H), 4.32 (q, J = 6.8 Hz, 2H), 2.52 (s, 3H), 1.19 (t, J = 6.8 Hz, 3H); MS (EI) for C,6Hi5FN40: 299.2 (MH+).
Example 9x Methyl 3-(2-amino-8-ethyl-4-methyl-7-oxo-7,8-dihydropyrido[2,3-ii]pyrimidin- 6-yl)benzoate: Ή NMR (400 MHz, DMSO-<¼): δ 8.34 (s, 1 H), 8.06 (s, 1H), 7.95-7.55 (m, 3H), 7.28 (bs, 1H), 4.35 (q, J = 6.8 Hz, 2H), 3.89 (s, 3H), 2.58 (s, 3H), 1.21 (t, J = 6.8 Hz, 3H); MS (EI) for Ci8W18N40 3: 339.2 (MH+).
Example 9y 2-Amino-8-ethyl-4-methyl-6-pyrimidin-5-ylpyrido[2,3--i]pyrimidin-7(8H)-one: Ή NMR (400 MHz, DMSO-d6): δ 8.39 (s, 1H), 7.65-7.30 (m, 5H), 4.31 (q, J = 7.2 Hz, 2H), 2.50 (s, 3H), 1.17 (t, J = 7.2 Hz, 3H); MS (EI) for Ci4H,4N60: 283.2 (MH1).
Example 10
2-Amino-8-ethyl-6 lH-imidazol-5-yl)-4-methylpyrido[2,3^pyrimidin-7(8H)-one
Figure imgf000096_0001
[00252] A solution of potassium hydroxide (0.139 g, 2.48 mmol) in absolute ethanol (3.0 mL) was added to a pressure tube charged with 4-(ethylamino)-6-methyl-2- (methylthio)pyrimidine-5-carbaldehyde (0.229 g, 1.08 mmol), prepared using procedures similar to those described for Intermediate 1, and 2-( lH-imidazol-5-yl)acetonitrile (0.174 g, 162 mmol) and heated to 70 °C. After 12 h, the reaction was allowed to cool to room temperature and concentrated in vacuo affording 8-ethyl-6-( lH-imidazol-5-yl)-4-methyl-2- (methylthio)pyrido[2,3-i/]pyrimidin-7(8H)-imine as a solid. The product was used in the subsequent step without further purification.
Figure imgf000096_0002
[00253] Acetic anhydride (15.0 mL) was added to a flask charged with crude 8-ethyl- 6-(lH-imidazol-5-yl)-4-memyl-2-(methylthio)pyrido[2,3-<i]pyrimidin-7(8H)-imine and heated to 100 °C. After 30 minutes, the reaction was allowed to cool to room temperature and concentrated in vacuo. The acetylated residue was then treated with 6 N HC1 (16 mL) and heated to 95 °C for 30 minutes then transferred to a large flask. A saturated solution of NaHCO3 (150 mL) was added at 0 °C to about pH = 8.0. The aqueous phase was washed thrice with ethyl acetate (100 mL) and the organic layers combined, then washed with brine and dried over Na2S04. The drying agent was filtered off and the organic layers were concentrated in vacuo to afford crude 8-ethyl-6-(lH-imidazol-5-yl)-4-methyl-2- (methyIthio)pyrido[2,3-ii|pyrimidin-7(8H)-one which was used in the subsequent step without further purification.
Figure imgf000097_0001
[00254] 3-Chloroperbenzoic acid (0.299 g, 1.73 mmol) was added to a solution of cmde 8-emyl-6-(lH-imidazol-5-yl)-4-methyl-2-(memylthio)pyrido[2,3-<flpyrimidin-7(8H)- one (0.260g, 0.866 mmol) in dichloromethane (10.0 mL) at room temperature. After 1.5 h, the reaction was diluted with dichloromethane (50 mL) and washed twice with saturated NaHC03, followed by brine. The organic phase was separated and dried over Na2S04, filtered, and concentrated in vacuo. The corresponding sulfone was used in the subsequent step without further purification.
Figure imgf000097_0002
[00255] Concentrated aqueous ammonium hydroxide (400 |JL) was added to a solution of the sulfone in dioxane ( 10 mL) at 0 °C. The reaction flask sealed, and allowed to warm to room temperature upon standing overnight. The reaction was concentrated in vacuo and purified on reverse phase HPLC (acetonitrile: water 0.1 % TFA, 20-60% gradient). The fractions containing product were collected and concentrated to one half volume and poured into saturated NaHC03 (50 mL). The aqueous phase was washed trice with ethyl acetate (50 mL) and dried over Na2S04, filtered, and concentrated in vacuo. The residue was triturated with methylene chloride and ethyl acetate to afford 2-amino-8-ethyl-6-( lH-imidazol-5-yl)- 4-memylpyrido[2,3-i ]pyrimidin-7(8H)-one (29 mg, 12 % yield) as a light yellow solid: Ή MR (400 MHz, CH3OH-d<): 5 8.52 (bs, 1H), 7.88 (bs, 1H), 7.76 (s, 1H), 4.30 (q, / = 6.8 Hz, 2H), 2.65 (s, 3H), 1.29 (t, J = 6.8 Hz, 3H); MS (EI) for C13Hi4 60: 271.0 (MH*).
Example 11
2-AnMno-8-ethyl-4-methyl-6-(lH-l,2,34^^
Figure imgf000098_0001
[00256] Trimethylsilylethyne ( 1.44 mL, 10.2 mmol) was added to a pressure tube charged with 2-amino-6-bromo-8-emyl-4-methylpyrido[2,3-if|pyrimidin-7(8H)-one (1.58 g, 5.59 mmol) from above, Cul (0.053 g, 0.279 mmol), and PdCl2(PPh3)2 (0.21 1 g, 0.279 mmol) in triethylamine (20 mL). The pressure tube was sealed under nitrogen and heated to 50 °C 96 h. The reaction was cooled to room temperature and poured into a saturated solution of NaHC03 (150 mL), then washed four times with ethyl acetate (50 mL). The organic layers were pooled and dried over Na2S04, filtered and concentrated in vacuo. The residue was purified on Si02 (2: 1 , methylene chloride: ethyl acetate) to afford 2-amino-8-ethyl-4-methyl- 6-((trimemylsilyl)ethynyl)pyrido[2,3- ]pyrimidin-7(8H)-one (1.09 g, 65 % yield) as an off white solid.
Figure imgf000098_0002
[00257] Potassium carbonate ( 1.00 g, 7.28 mmol) was added to a flask charged with 2-amino-8-e l-4-memyl-6-((trimethylsilyl)ethynyl)pyrido[2,3-^pyrimidin-7(8H)-one ( 1.09 g, 3.64 mmol) in anhydrous methanol ( 15 mL). The reaction was stirred at room temperature under nitrogen for 16 h. The reaction was concentrated to one half volume and the yellow precipitate collected by vacuum filtration to afford 2-amino-8-ethyl -6-ethynyl-4- methylpyrido[2,3-i ]pyrimidin-7(8H)-one.
Figure imgf000099_0001
[00258] Anhydrous DMF (5.0 mL) was added to a flask charged with 2-amino-8-ethyl
-6-ethynyl-4-methylpyrido[2,3-i ]pyrimidin-7(8H)-one (0.204 g, 0.894 mmol), sodium azide (0.070 g, 1.07 mmol), and ammonium chloride (0.057 g, 1.07 mmol). The reaction was capped under nitrogen and heated to 120 °C. After 48 h, the reaction was cooled to room temperature and concentrated in vacuo. The residue was purified on reverse phase HPLC (acetonitrile: water 0.1 % TFA, 20-60% gradient). The fractions containing product were collected and concentrated to one half volume and poured into saturated NaHC03 (50 mL). The aqueous phase was washed trice with ethyl acetate (50 mL) and dried over Na2S04, filtered, and concentrated in vacuo. The residue was triturated with methylene chloride and ethyl acetate to afford 2-amino-8-ethyl-4-methyl-6-( lH-l,2,3-triazol-5-yl)pyrido[2,3- i/]pyrimidin-7(8H)-one ( 14 mg, 6 % yield) as a light yellow solid: Ή NMR (400 MHz, DMSO- ): δ 8.55 (bs, 1H), 8.41 (bs, 1H), 7.32 (bs, 2H), 4.37 (q, / = 7.2 Hz, 2H), 2.60 (s, 3H), 1.21 (t, J = 7.2 Hz, 3H); MS (EI) for C,2H,3N70: 272.0 (MH+).
Example 12
2-Amino-8-emyl-4-methyl-6-(lH-tetrazol-5-yl)pyrido[2,3--i]pyrimidin-7(8H)-one
Figure imgf000099_0002
[00259] Potassium carbonate (0.539 g, 3.90 mmol) was added to a suspension of 4-(emylamino)-6-memyl-2-(methylthio)pyrimidine-5-carbaldehyde (0.413 g, 1.95 mmol) from above, and malononitrile (0.194 g, 2.93 mmol) in absolute ethanol (15.0 mL) and heated to 70 °C. After one h, the reaction was allowed to cool to room temperature and concentrated in vacuo. The residue was diluted with ethyl acetate (50 mL) and washed with saturated NaHC03 (50 mL), and brine. The organic phase was separated and concentrated in vacuo. The residue was precipitated with ethyl acetate and hexanes to give 8-ethyl-7-imino-4- methyl-2-(memylthio)-7,8-dihydropyrido[2,3-ii]pyrirnidine-6-carbonitrile as a brown solid that was used in the subsequent step without further purification.
Figure imgf000100_0001
[00260] Acetic anhydride ( 10.0 mL) was added to a flask charged with 8-ethyl-7- imino-4-memyl-2-(memylthio)-7,8-dihydropyrido[2,3-iilpyrimidine-6-carbonitrile (0.506 g, 1.95 mmol) and heated to 100 °C. After one h, the reaction was allowed to cool to room temperature and concentrated in vacuo. The acetylated residue was then treated with 6 N HCl (40 mL) and heated to 95 °C for one hour then transferred to a large flask. A saturated solution of NaHC03 (500 mL) was added slowly at 0 °C until a ~pH 8.0 was achieved. The aqueous phase was washed thrice with ethyl acetate (100 mL) and the organic layers combined, then washed with brine and dried over Na2S04. The drying agent was filtered and concentrated in vacuo to afford crude 8-ethyl-4-methyl-2-(methylthio)-7-oxo-7,8- dihydropyrido[2,3-i/]pyrimidine-6-carbonitrile which was used in the subsequent step without further purification.
Figure imgf000100_0002
[00261] 3-Chloroperbenzoic acid (1.00 g, 5.85 mmol) was added to a solution of crude 8-emyl-4-methyl-2-(methylthio)-7-oxo-7,8-dihydropyrido[2,3-rf]pyrimidine-6-carbonitrile (0.507 g, 1.95 mmol) in dichloromethane (30.0 mL) at room temperature. After 2.5 hours, the reaction was diluted with dichloromethane (50 mL) and washed twice with saturated
NaHC03, followed by brine. The organic phase was separated and dried over Na2S04, filtered, and concentrated in vacuo. 2-Amino-8-ethyl-4-methyl-7-oxo-7,8-dihydropyrido[2,3- d]pyrimidine-6-carbonitrile was used in the subsequent step without further purification.
[00262] Ammonium hydroxide (500 |iL) was added to a solution of the above sulfone in dioxane ( 10 mL) at 0 °C. The reaction flask sealed, and allowed to warm to room temperature upon standing overnight. The reaction was concentrated in vacuo triturated with ethyl acetate to afford the product which was used in the subsequent step without further purification.
Figure imgf000101_0001
[00263] Tributyltin azide (660 uL, 2.41 mmol) was added to a flask charged with 2-amino-8-emyl-4-memyl-7-oxo-7,8-dihydropyrido[2,3-(i]pyrimidine-6-carbomtrile (0.184 g, 0.803 mmol) in anhydrous toluene (5.0 mL). The reaction was fitted with a reflux condenser and heated to 140 °C under a nitrogen atmosphere. After 20 h, the reaction was cooled to room temperature and the precipitate collected by vacuum filtration and washed with absolute ethanol to give 2-amino-8-emyl-4-methyl-6-(lH-tetrazoI-5-yl)pyrido[2,3-rf]pyrimidin-7(8H)- one (98 mg, 45 % yield) as a light brown solid: Ή NMR (400 MHz, 20 % DC1 in D20): 8 6.97 (s, 1H), 2.42 (q, J = 7.2 Hz, 2H), 0.953 (s, 3H), -0.73 (t, J = 7.2 Hz, 3H); MS (EI) for CnHnNgO: 271.0 (MIT).
Example 13
Figure imgf000101_0002
[00264] A mixture of 8-(3-methoxypropyl)-4-methyl-2-(methylthio)pyrido[2,3- d]pyrimidin-7(8H)-one (0.36 g, 1.29 mmol), prepared using procedures similar to those described in Example 1, dichloromethane (10 mL), and 77 % 3-chloroperbenzoic acid with water (0.723 g, 3.23 mmol) was stirred for 1 h. The mixture was diluted with dichloromethane, washed with sat. sodium bicarbonate (3 times), brine, dried over sodium sulfate, and DCM was removed under reduced pressure. The crude 8-(3-methoxypropyl)-4- methyl-2-(methylsulfonyl)pyrido[2,3-d]pyrimidin-7(8H)-one was used without further purification for subsequent step.
Figure imgf000102_0001
[00265] 8-(3-me^oxypropyl)-4-rnethyl-2-(rnethylsulfonyl)pyrido[2,3-d]pyrimidin- 7(8H)-one, and a solution of 2M ethyiamine in THF (20 mL) was stirred for 2 h. THF was removed under reduced pressure and the crude product was purified by flash column chromatography to give 2-(ethylamino)-8-(3-methoxypropyl)-4-methylpyrido[2,3- d]pyrimidi -7(8H)-one (0.18 g, 50 % yield over 2 steps).
Figure imgf000102_0002
[00266] To a solution of 2-(ethylamino)-8-(3-methoxypropyl)-4-methylpyrido[2,3- d]pyrimidin-7(8H)-one (0.18 g, 0.65 mmol), acetic acid (5 mL) and dichloromethane (3 mL) was added bromine (36 ul, 0.7 mmol). The mixture was stirred for 5 minutes, and then diluted with DCM and water. The organic layer was washed with sat. sodium bicarbonate (3 times), brine, dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude product was purified by flash column chromatography to gave 0.13 g (56 % yield) of 6-bromo-2-(ethylamino)-8-(3-methoxypropyl)-4-methylpyrido[2,3-d]pyrimidin-7(8H)- one. Ή NMR (400MHz, CDC13) 5 8.09 (s, 1H), 5.44 (Br. s, 1H), 4.55 (m, 2H), 3.54-3.47 (m, 4H), 3.33 (s, 3H), 2.53 (s, 3H), 2.05-2.00 (m, 2H), 1.30- 1.23 (m, 3H); MS (EI) for Cl4Hi9BrN402: 355 (MH+).
[00267] Using the same or analogous synthetic techniques and substituting with appropriate reagents, the following compounds were prepared:
Example 13a. 6-bromo-8-(2-ethoxyethyl)-2-(ethylamino)-4-methylpyrido[2,3-d]pyrimidin- 7(8H)-one: Ή NMR (400MHz, CDC13) δ 8.09 (s, 1H), 5.37 (Br. s, 1H), 4.67 (m, 2H), 3.74 (m, 2H), 3.61-3.56 (t, 2H), 3.51 (m, 2H), 2.53 (s, 3H), 1.29-1.25 (t, 3H), 1.19-1.15 (t, 3H); MS (EI) for Cl4H,9BrN 02: 355 (MH+).
Example 13b. 6-bromo-8-(3-ethoxypropyl)-2-(ethylamino)-4-methylpyrido[2,3-d]pyrimidin- 7(8H)-one: Ή NMR (400MHz, CDC13) δ 8.09 (s, 1H), 5.37 (Br. s, 1H), 4.53 (m, 2H), 3.52 (m, 4H), 3.48-3.43 (m, 2H), 2.53 (s, 3H), 2.04-2.00 (m, 2H), 1.29-1.25 (t, 3H), 1.19-1.15 (t, 3H); MS (EI) for C15H2iBrN402: 369 (MH+).
Example 13c. 6-bromo-2-(ethyIamino)-8-(3-isopropoxypropyl)-4-methylpyrido[2,3- d]pyrimidin-7(8H)-one: Ή NMR (400MHz, CDC13) δ 8.09 (s, 1H), 5.37 (Br. s, IH), 4.53 (m, 2H), 3.59-3.49 (m, 5H), 2.52 (s, 3H), 2.01-1.98 (m, 2H), 1 .28-1.25 (t, 3H), 1.13- 1.1 1 (t, 6H); MS (EI) for C,6H23BrN402: 383 (MH+).
Example 14
Figure imgf000103_0001
[00268] A mixture of 2,4-dichloro-6-methylpyrimidine (Aldrich, 5 g, 30 mmol), cyclohexylamine (3 g, 30 mmol) and DIEA (10 mL) was stirred at 80 °C for 12 h. The volatile material was removed under reduced pressure. The residue was loaded on a silica gel column, and was eluted with hexanes/ethyl acetate (3: 1 ). 8-cyclohexyl-2-(ethylamino)-4- methyl-6-(thiopheN-2-yl)pyrido[2,3-d]pyrimidin-7(8H)-one was obtained as colorless oil (2.8 g, 41% yield).
Figure imgf000103_0002
[00269] The pro uct was reacted with a solution of ethylamine (10 equiv.) in THF at 100 °C for 12 h. The crude 2-ethylamino-4-cyclohexylamino-6-methylpyrimidine was obtained from a standard workup and was used in the next step.
Figure imgf000103_0003
[00270] To a so ution of 2-ethylamino-4-cyclohexy amino-6-methylpyrimidine (600 mg, 2.56 mmol) in CH3CN (10 mL) was added N-iodosuccinimide (MS, 658 mg, 2.92 mmol). The reaction was stirred for 2 h at room temperature. After removal of the solvent, the residue was dissolved in EtOAc. The organic phase was then washed with sodium bisulfite, brine, and dried over Na2S04. Purification by flash column chromatography gave 660 mg (73% yield) of 2-ethylamino-4-cyclohexylamino-5-iodo-6-methylpyrimidine.
Figure imgf000104_0001
[00271] To a solution of 2-ethylamino-4-cyclohexylamino-5-iodo-6-methylpyrimidine (660 mg, 1.83 mmol) in DMA (7 mL) was added ethyl acrylate (458 mg, 4.58 mmol), Pd(OAc)2 (121 mg, 0.18 mmol), (o-Tol)3P (1 10 mg, 0.37 mmol), and Et3N (740 mg, 7.32 mmol). The mixture was then stirred at 100 °C for 12 h under N2. Standard workup and purification by column chromatography gave 411 mg (67% yield) of (E)-ethyl 3-(4- (cyclohexylamino)-2-(ethylamino)-6-methylpyrimidin-5-yl)acrylate
Figure imgf000104_0002
[00272] (E)-ethyl 3-(4-(cyclohexylamino)-2-(ethylamino)-6-methylpyrimidin-5- yl)acrylate (200 mg, 0.6 mmol) was dissolved in AcOH (2 mL). This solution was heated in a sealed tube at 186 °C for 17 h. Standard workup and purification by column
chromatography gave 65 mg (38 % yield) of 8-cyclohexyl-2-(ethylamino)-4- methylpyrido[2,3-d]pyrimidin-7(8H)-one.
Figure imgf000104_0003
[00273] To 8-cyclohexyl-2-(ethylamino)-4-methylpyrido[2,3-d]pyrimidin-7(8H)-one in AcOH and CH2C12 was added Br2 (22 uL, 0.42 mmol) at 80 °C. Standard workup and purification by column chromatography gave 65 mg (0.17 mmol, 80 % yield) of 6-bromo-8- cyclohexyl-2-(ethylamino)-4-methylpyrido[2,3-d]pyrimidin-7(8H)-one.
Figure imgf000105_0001
[00274] The bromide (65 mg, 0.17 mmol) obtained above was reacted with 2-thiopheneboronic acid (45 mg, 0.36 mmol) in the presence of Pd(PPh3)4 (20 mg, 0.018 mmol) and Na2C03 (38 mg, 0.36 mmol) in 1 ,4-dioxane/H20 (1 : 1 ) at 100 °C for 2 h. Removal of solvents and purification by column chromatography gave 33 mg (50% yield) of 8-cyclohexyl-2-(emylamino)-4-memyl-6-(miopheN-2-yl)pyrido[2,3-d]pyrimidin-7(8H)-one. Ή NMR (400 MHz, DMSO-d6) 5 8.01. (br s, 1 H), 7.60 (m, 1 H), 7.37 (m, 1 H), 7.10 (m, 1H), 5.60-5.40 (m, 1 H), 3.55 (m, 2 H), 2.85 (m, 1 H), 2.61 (s, 3 H), 1.90 (m, 2 H), 1.71 (m, 4 H), 1.43 (m, 2 H), 1.30-1.2 (m, 2 H),1.30 (t, 3 H); MS (EI) for C20H24N4OS: 369 (MH+).
[00275] Using the same or analogous synthetic techniques and substituting with appropriate reagents, the following compound was prepared:
Example 14a. 6-bromo-8-cyclopropyl-2-(ethyIamino)-4-methylpyrido[2,3-d] pyrimidin- 7(8H)-one: Ή NMR (400 MHz, CDC13) δ 8.06 (s, 1 H), 5.37 (br s, 1 H), 3.54 (m, 2 H), 2.94 (br s, 1H), 2.51 (s, 3 H), 1.31-1.25 (m, 5 H), 0.91 (br s, 2 H); MS (EI) for C,3H,5BrN40: 323 (MH+).
Example 15
Figure imgf000105_0002
[00276] To a solution of 6-bromo-2-(ethylamino)-4-methylpyrido[2,3-d]pyrimidin- 7(8H)-one (100 mg, 0.35 mmol) in DMF (2 mL), prepared using procedures analogous to those described in Example 14, was added NaH (30 mg, 60%, 0.7 mmol). The mixture was stirred for 30 min at room temperature and was warmed to 70 °C. 3-Bromopropanol (48 mg, 0.35 mmol) was then added. The stirring was continued for 12 h. Standard workup and purification by column chromatography gave 33 mg (27% yield) of 6-bromo-2-(ethylamino)- 8-(3-hydroxypropyl)-4-methylpyrido[2,3-d]pyrimidin-7(8H)-one. Ή NMR (400 MHz, CDC13) δ 8.13 (s, 1 H), 5.42 (br s, 1 H), 4.59 (br s, 2 H), 3.50-3.47 (m, 5 H), 2.55 (s, 3 H), 2.02 (br s, 2 H), 1.28 (t, 3 H); MS (EI) for C,3H,7BrN402: 341 (MH+).
[00277] Using the same or analogous synthetic techniques and substituting with appropriate reagents, the following compounds were prepared:
Example 15a. 6-bromo-2-(ethylamino)-8-(2-hydroxyethyl)-4-methylpyrido[2,3-d]pyrimidin- 7(8H)-one: Ή NMR (400 MHz, DMSO-d6) δ 8.38 (s, 1 H), 4.82 (br s, 1 H), 4.40 (br s, 2 H), 3.62-3.55 (m, 2 H), 3.40-3.20 (m, 3 H), 2.55 (s, 3 H), 1.15 (t, 3 H); MS (EI) for C,2Hi5BrN402: 327 (MH+).
Example 15b. 6-bromo-2-(ethylamino)-4-methyl-8-(2-(piperidin- 1 -yl)ethyl)pyrido[2,3- d]pyrimidin-7(8H)-one: Ή NMR (400 MHz, CDC13) δ 8.08 (s, 1 H), 5.39 (br s, 1 H), 4.59 (br s, 2 H), 3.55-3.40 (m, 2 H), 2.70-2.50 (m, 6 H), 2.52 (s, 3 H), 1.62-1.58 (m, 4 H), 1.46- 1.40 (m, 2 H), 1.27 (t, 3 H); MS (EI) for C17H24BrN50: 394 (MH+).
Biological Examples
Biological Example 1
PI3KaIpha Luciferase-Coupled Chemiluminescence Assay Protocol
[00278] PI3Ka activity is measured as the percent of ATP consumed following the kinase reaction using luciferase-luciferin-coupled chemiluminescence. Reactions were conducted in 384-well white, medium binding microtiter plates (Greiner). Kinase reactions were initiated by combining test compounds, ATP, substrate (PIP2), and kinase in a 20 μΐ. volume in a buffer solution. The standard PDKalpha assay buffer is composed 50 mM Tris, pH 7.5, 1 mM EGTA, 10 mM MgCl2, 1 mM DTT and 0.03% CHAPS. The standard assay
concentrations for enzyme, ATP, and substrate are 0.5-1.1 nM, ΙμΜ, and 7.5 μΜ, respectively. The reaction mixture was incubated at ambient temperature for approximately 2 h. Following the kinase reaction, a 10 μΐ-, aliquot of luciferase-luciferin mix (Promega Kinase-Glo) was added and the chemiluminescence signal measured using a Victor2 plate reader (Perkin Elmer). Total ATP consumption was limited to 40-60% and IC50 values of control compounds correlate well with literature references.
[00279] Certain compounds of the invention were tested in this assay and demonstrated the ability to bind to PI3K. For example, in one embodiment of the invention, the PI3K inhibitor is selected from the compounds in Table 1 having a PI3K-binding affinity of about 9 μΜ or less. In another embodiment, the PI3K inhibitor is selected from the compounds in Table 1 having a PI3K-binding affinity of about 5 μΜ or less. In another embodiment, the PI3K inhibitor is selected from the compounds in Table 1 having a PI3K-binding affinity of about 3 μΜ or less. In another embodiment, the PI3K inhibitor is selected from the compounds in Table 1 having a PI3K-binding affinity of about 1.5 μΜ or less. In another embodiment, the PI3K inhibitor is selected from the compounds in Table 1 having a PI3K-binding affinity of about 1 μΜ or less. In another embodiment, the PI3K inhibitor is selected from the compounds in Table 1 having a PI3K-binding affinity of about 0.6 μΜ or less. In another embodiment, the PI3K inhibitor is selected from the compounds in Table 1 having a PI3K- binding affinity of about 0.3 μΜ or less. In another embodiment, the PI3K inhibitor is selected from the compounds in Table 1 having a PI3K-binding affinity of about 0.2 μΜ or less. In another embodiment, the PI3K inhibitor is selected from the compounds in Table 1 having a PI3K-binding affinity of about 0.1 μΜ or less. In another embodiment, the PI3K inhibitor is selected from the compounds in Table 1 having a PI3K-binding affinity of about 0.04 μΜ or less. In another embodiment, the PI3K inhibitor is selected from the compounds in Table 1 having a PI3K-binding affinity of about 0.020 μΜ or less.
Biological Example 2
Phospho AKT assay
[00280] PC3 cells were seeded on 6-well plates at 150,000 cells/well. Cells were cultured for 3 days, then treated with compounds in serum-free medium for 3 hr. EGF (100 ng mL) was added for the last 10 min. Cells were lysed in TENN buffer. Phospho T308 Akt and total Akt were quantified by ELISA performed according to the Biosource assay protocol. The readings of phospho Akt were normalized to total Akt readings.
Biological Example 3
Phospho S6 assay
[00281] PC3 cells were seeded on 96-well plates at 8,000 cells/well. For each experiment, cells were seeded and treated in duplicated plates: one plate for phospho S6 CellELISA, and one plate for total S6 CellELISA. Cells were cultured on the plates for 3 days, then treated with compounds in serum-free medium for 3 hr in triplicate. Cells were fixed with 4% formaldehyde, quenched with 0.6% H202, blocked with 5% BSA, incubated with either phospho S6 antibody or total S6 antibody overnight, incubated with goat-anti-rabbit-IgG- HRP for 1 hr, and developed in chemiluminescent substrate. Biological Example 4
PIP3 assay
[00282] MCF-7 cells grown in 10-cm dishes were starved for 3 hours in DMEM, and then treated with compounds for 20 minutes. In the last 2 minutes of the incubation with the compounds, EGF (100 ng/mL) was added to stimulate the production of PDP3. The medium was aspirated and the cells were scraped with 10% trichloroacetic acid. The lipids were extracted from the pellet after the cell lysates were centrifuged. PIP3 in the cellular lipid extraction was quantified with the AlphaScreen assay in which Grpl-PH is used as the PIP3 specific probe. The amount of cellular PIP3 was calculated from the standard curve of diC8 PI (3,4,5) P3.
Biological Example 5-10
In vivo models
[00283] Female and male athymic nude mice (NCr) 5-8 weeks of age and weighing approximately 20 g were used in the following model. Prior to initiation of a study, the animals were allowed to acclimate for a minimum of 48 h. During these studies, animals were provided food and water ad libitum and housed in a room conditioned at 70-75 °F and 60% relative humidity. A 12 h light and 12 h dark cycle was maintained with automatic timers. All animals were examined daily for compound-induced or tumor-related deaths.
[00284] PC-3 human prostate adenocarcinoma cells were cultured in vitro in DMEM (Mediatech) supplemented with 20% Fetal Bovine Serum (Hyclone), PenicilliN-Streptomycin and non-essential amino acids at 37°C in a humidified 5% C02 atmosphere. On day 0, cells were harvested by trypsinization and 3x l06 cells (passage 13, 99% viability) in 0.1 mL of ice-cold Hank's balanced salt solution were implanted subcutaneously into the hindflank of 5-8 week old male nude mice. A transponder was implanted in each mouse for identification, and animals were monitored daily for clinical symptoms and survival. Body weights were recorded daily.
[00285] U-87 MG human glioblastoma cells were cultured in vitro in DMEM (Mediatech) supplemented with 10% Fetal Bovine Serum (Hyclone), PenicilliN-Streptomycin and nonessential amino acids at 37°C in a humidified 5% C02 atmosphere. On day 0, cells were harvested by trypsinization and 2xl06 cells (passage 5, 96% viability) in 0.1 mL of ice-cold Hank's balanced salt solution were implanted intradermally into the hindflank of 5-8 week old female nude mice. A transponder was implanted in each mouse for identification, and animals were monitored daily for clinical symptoms and survival. Body weights were recorded daily. [00286] A549 human lung carcinoma cells were cultured in vitro in DMEM (Mediatech) supplemented with 10% Fetal Bovine Serum (Hyclone), PenicilliN-Streptomycin and nonessential amino acids at 37°C in a humidified 5% C02 atmosphere. On day 0, cells were harvested by trypsinization and lOxlO6 cells (passage 12, 99% viability) in 0.1 mL of ice-cold Hank's balanced salt solution were implanted intradermally into the hindflank of 5-8 week old female nude mice. A transponder was implanted in each mouse for identification, and animals were monitored daily for clinical symptoms and survival. Body weights were recorded daily.
[00287] A2058 human melanoma cells were cultured in vitro in DMEM (Mediatech) supplemented with 10% Fetal Bovine Serum (Hyclone), PenicilliN-Streptomycin and nonessential amino acids at 37°C in a humidified, 5% C02 atmosphere. On day 0, cells were harvested by trypsinization and 3xl06 cells (passage 3, 95% viability) in 0.1 mL ice-cold Hank's balanced salt solution were implanted intradermally in the hind-flank of 5-8 week old female athymic nude mice. A transponder was implanted in each mouse for identification, and animals were monitored daily for clinical symptoms and survival. Body weights were recorded daily.
[00288] WM-266-4 human melanoma cells were cultured in vitro in DMEM (Mediatech) supplemented with 10% Fetal Bovine Serum (Hyclone), PenicilliN-Streptomycin and nonessential amino acids at 37°C in a humidified, 5% C02 atmosphere. On day 0, cells were harvested by trypsinization and 3xl06 cells (passage 5, 99% viability) in 0.1 mL ice-cold Hank's balanced salt solution were implanted intradermally in the hind-flank of 5-8 week old female athymic nude mice. A transponder was implanted in each mouse for identification, and animals were monitored daily for clinical symptoms and survival. Body weights were recorded daily.
[00289] For subcutaneous or intradermal tumors, the mean tumor weight of each animal in the respective control and treatment groups was determined twice weekly during the study. Tumor weight (TW) was determined by measuring perpendicular diameters with a caliper, using the following formula:
tumor weight (mg) = [tumor volume = length (mm) x width (mm )]/2
[00290] These data were recorded and plotted on a tumor weight vs. days
post-implantation line graph and presented graphically as an indication of tumor growth rates. Percent inhibition of tumor growth (TGI) is determined with the following formula:
Figure imgf000110_0001
where Xo = average TW of all tumors on group day Xf = TW of treated group on Day f
Yf = TW of vehicle control group on Day f
[00291] If tumors regress below their starting sizes, then the percent tumor regression is determined with the following formula:
( Xf - Xo )
00
[00292] Tumor size is calculated individually for each tumor to obtain a mean ± SEM value for each experimental group. Statistical significance is determined using the 2-tailed Student's t-test (significance defined as P<0.05).
Pharmaceutical Composition Examples
[00293] The following are representative pharmaceutical formulations containing a compound of Formula I.
Tablet Formulation
[00294] The following ingredients are mixed intimately and pressed into single scored tablets.
Ingredient Quantity per tablet, mg compound of this invention 400
Cornstarch 50
croscarmellose sodium 25
Lactose 120
magnesium stearate 5
Capsule Formulation
[00295] The following ingredients are mixed intimately and loaded into a hard-shell gelatin capsule. Ingredient Quantity per tablet, compound of this invention
lactose, spray-dried
magnesium stearate
Suspension Formulation
The following ingredients are mixed to form a suspension for oral administration.
Ingredient Amount compound of this invention 1.0 g
fumaric acid 0.5 g
sodium chloride 2.0 g
methyl paraben 0.15 g
propyl paraben 0.05 g
granulated sugar 25.5 g
sorbitol (70% solution) 12.85 g
Veegum K (Vanderbilt Co.) 1.0 g
Flavoring 0.035 mL
Colorings 0.5 mg
distilled water q.s. to lOO mL
Injectable Formulation
The following ingredients are mixed to form an injectable formulation.
Ingredient Amount compound of this invention 1.2 g
sodium acetate buffer solution 0.4 M 2.0 mL
HC1 ( 1 N) or NaOH ( 1 M) q.s. to suitable pH
water (distilled, sterile) q.s.to 20 mL [00298] All of the above ingredients, except water, are combined and heated to 60- 70.degree. C. with stirring. A sufficient quantity of water at 60.degree. C. is then added with vigorous stirring to emulsify the ingredients, and water then added q.s. to 100 g.
Suppository Formulation
[00299] A suppository of total weight 2.5 g is prepared by mixing the compound of the invention with Witepsol.RTM. H-15 (triglycerides of saturated vegetable fatty acid; Riches- Nelson, Inc., New York), and has the following composition:
Ingredient Quantity per tablet, mg compound of this invention 500
Witepsol®H-15 balance
[00300] The foregoing invention has been described in some detail by way of illustration and example, for purposes of clarity and understanding. The invention has been described with reference to various specific embodiments and techniques. However, it should be understood that many variations and modifications may be made while remaining within the spirit and scope of the invention. It will be obvious to one of skill in the art that changes and modifications may be practiced within the scope of the appended claims. Therefore, it is to be understood that the above description is intended to be illustrative and not restrictive. The scope of the invention should, therefore, be determined not with reference to the above description, but should instead be determined with reference to the following appended claims, along with the full scope of equivalents to which such claims are entitled. All patents, patent applications and publications cited in this application are hereby incorporated by reference in their entirety for all purposes to the same extent as if each individual patent, patent application or publication were so individually denoted.
I l l

Claims

What is claimed is:
1. A method for treating a disease, disorder, or syndrome which method comprises administering to a patient a pharmaceutical composition comprising a therapeutically effective amount of
Figure imgf000113_0002
or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition comprising a therapeutically effective amount of a Compound A and a pharmaceutically acceptable carrier, excipient, or diluent, in combination with another agent.
2. The method of claim 1, wherein the disease, disoreder, or syndrome is selected from anaplastic thyroid cancer, chronic lymphocytic leukemia, mantel cell lymphoma, non- Hodgkins lymphoma, glioblastoma multiforme, pediatric brain tumors, pontine glioma, HPV- related head and neck cancer, Hormone Receptive positve (HR+) breast cancer, and HER-2 overexpressing breast cancer.
3. The method of claim 1, wherein Compound A is adminstered in combination with one or more agents selected from:
Figure imgf000113_0001
Agent Name
E7389 (Halichondrin B Analog)
EMD 121974 (Cilengitide)
Entinostat (MS-275, SNDX-275) FK228 (Depsipeptide; Romidepsin)
Flavopiridol (alvocidib)
GDC-0449 ***
IMC-A12
MDX-010 (MDX-CTLA4; Hybridoma- derived and Transfectoma-derived)
MK-2206
Q-6-Benzylguanine
Obatoclax mesylate (GX15-070MS)
OSI-906
Pertuzumab
Rl 15777 (tipifarnib, Zarnestra)
Reolysin
RO4929097
SB-715992 (ispinesib)
SCH727965
SJG-136
Thalidomide (Thalomid)
Triapine
UCN-01
VEGF-Trap (aflibercept)
Vorinostat (suberoylanilide hydroxamic acid; SAHA)
XK469R
17-AAG
17-DMAG
Alemtuzumab (Campath)
AZD2171 (cediranib; Recentin™)
AZD6244
BAY 43-9006 tosylate (BAY 54-9085; sorafenib tosylate)
Bevacizumab (rhuMAb VEGF, Avastin) BMS-354825 (dasatinib, Sprycel)
CCI-779 (temsirolimus, Torisel)
Erlotinib (OSI-774; Tarceva)
Gefitinib (ZD 1839, Iressa)
GM-CSF (sargramostim, Leukine)
GW572016 (lapatinib)
GW786034 (pazopanib)
MLN 518
Oxaliplatin (Eloxatin)
Perifosine
Rituximab (MoAb C2B8 and CD20, chimeric)
STI571 (imatinib, Gleevec) Agent Name
Sunitinib malate (SUOI 1248 L-malate;
Sutent)
Trastuzumab (Herceptin)
4. The method of claim 3, wherein wherein Compound A is adminstered in combination with one or more agents selected from:
Figure imgf000115_0001
Agent Name
Vorinostat (suberoylanilide hydroxamic
acid; SAHA)
XK469R
17-AAG
5. The method of claim 3, wherein wherein Compound A is adminstered in combination with one or more agents selected from ABT-888, AZD6244, CCI-779, erlotinib, gefitinib, GW572016, GW786034, pertuzumab, and sunitinib.
6. The method of claim 1 , wherein Compound A is administered as a capsule formulation.
Gray White Gray/Orange Gray Opaque Swedish Opaque Opaque Capsule Capsule Orange Capsule1 Capsule (for 30-mg (for 40-mg Capsule (for 5-mg (for 10-mg strength) strength) (for 50-mg
Component strength) strength) strength)
2.7074%
(Body)
FDA/E171 titanium dioxide 2.7074% 2.9079% 2.7074% 0.4902%
0.4902%
(Cap)
FDA E172 black iron dioxide 0.3075%
0.3075% — 0.3075% —
(Body)
FDA/E172 red iron dioxide — — 1.4706% — 1.4706%
(Cap)
Gelatin qsp 100% qsp 100% qsp 100% qsp 100% qsp 100% qsp, quantity sufficient for preparation.
PCT/US2011/060293 2010-11-12 2011-11-11 Pyridopyrimidinone inhibitors of p13k alpha WO2012065019A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US41335610P 2010-11-12 2010-11-12
US61/413,356 2010-11-12

Publications (2)

Publication Number Publication Date
WO2012065019A2 true WO2012065019A2 (en) 2012-05-18
WO2012065019A3 WO2012065019A3 (en) 2012-10-04

Family

ID=45044729

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/060293 WO2012065019A2 (en) 2010-11-12 2011-11-11 Pyridopyrimidinone inhibitors of p13k alpha

Country Status (1)

Country Link
WO (1) WO2012065019A2 (en)

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012149308A1 (en) * 2011-04-29 2012-11-01 Exelixis, Inc. Method of treating lymphoma using pyridopyrimidinone inhibitors of pi3k/mtor
WO2012174327A1 (en) * 2011-06-15 2012-12-20 Exelixis, Inc. Combination therapies for treating hematologic malignancies using pyridopyrimidinone inhibitors of pi3k/mtor with bendamustine and/or rituximab
WO2014058947A1 (en) * 2012-10-12 2014-04-17 Sanofi Compositions and methods for treating cancer using pi3k inhibitor and anti-cd19 maytansinoid immunoconjugate
WO2014135572A1 (en) * 2013-03-05 2014-09-12 Sanofi Tablet formulation of a pi3kalpha inhibitor
WO2014145986A1 (en) * 2013-03-15 2014-09-18 Verseon, Inc. Multisubstituted aromatic compounds as serine protease inhibitors
US8940742B2 (en) 2012-04-10 2015-01-27 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9359365B2 (en) 2013-10-04 2016-06-07 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9533967B2 (en) 2010-03-30 2017-01-03 Verseon Corporation Multisubstituted aromatic compounds as inhibitors of thrombin
US9708348B2 (en) 2014-10-03 2017-07-18 Infinity Pharmaceuticals, Inc. Trisubstituted bicyclic heterocyclic compounds with kinase activities and uses thereof
US9751888B2 (en) 2013-10-04 2017-09-05 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9775844B2 (en) 2014-03-19 2017-10-03 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9951025B2 (en) 2013-03-15 2018-04-24 Verseon Corporation Halogenopyrazoles as inhibitors of thrombin
US10160761B2 (en) 2015-09-14 2018-12-25 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
US10189810B2 (en) 2014-09-17 2019-01-29 Verseon Corporation Pyrazolyl-substituted pyridone compounds as serine protease inhibitors
US10322192B2 (en) 2016-03-02 2019-06-18 Eisai R&D Management Co., Ltd. Eribulin-based antibody-drug conjugates and methods of use
US10532995B2 (en) 2015-02-27 2020-01-14 Verseon Corporation Substituted pyrazole compounds as serine protease inhibitors
US10759806B2 (en) 2016-03-17 2020-09-01 Infinity Pharmaceuticals, Inc. Isotopologues of isoquinolinone and quinazolinone compounds and uses thereof as PI3K kinase inhibitors
US10919914B2 (en) 2016-06-08 2021-02-16 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
CN115348963A (en) * 2021-03-08 2022-11-15 暨南大学 Pyridopyrimidine compound and application thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4107288A (en) 1974-09-18 1978-08-15 Pharmaceutical Society Of Victoria Injectable compositions, nanoparticles useful therein, and process of manufacturing same
US5145684A (en) 1991-01-25 1992-09-08 Sterling Drug Inc. Surface modified drug nanoparticles
US20040009993A1 (en) 2001-12-11 2004-01-15 Pharmacia Italia S.P.A. Pyridopyrimidinones derivatives as telomerase inhibitors
WO2007044813A1 (en) 2005-10-07 2007-04-19 Exelixis, Inc. PYRIDOPYRIMIDINONE INHIBITORS OF PI3Kα
WO2008124161A1 (en) 2007-04-10 2008-10-16 Exelixis, Inc. Methods of treating cancer using pyridopyrimidinone inhibitors of pi3k alpha

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2006302148B2 (en) * 2005-10-07 2012-12-06 Exelixis, Inc. Pyridopyrimidinone inhibitors of PI3Kalpha
US20100150827A1 (en) * 2007-04-11 2010-06-17 Exelixis, Inc Pyrido [2, 3-d] pyrimidin-7-one compounds as inhibitors of p13k-alpha for the treatment of cancer
WO2009017838A2 (en) * 2007-08-01 2009-02-05 Exelixis, Inc. Combinations of jak-2 inhibitors and other agents

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4107288A (en) 1974-09-18 1978-08-15 Pharmaceutical Society Of Victoria Injectable compositions, nanoparticles useful therein, and process of manufacturing same
US5145684A (en) 1991-01-25 1992-09-08 Sterling Drug Inc. Surface modified drug nanoparticles
US20040009993A1 (en) 2001-12-11 2004-01-15 Pharmacia Italia S.P.A. Pyridopyrimidinones derivatives as telomerase inhibitors
WO2007044813A1 (en) 2005-10-07 2007-04-19 Exelixis, Inc. PYRIDOPYRIMIDINONE INHIBITORS OF PI3Kα
WO2008124161A1 (en) 2007-04-10 2008-10-16 Exelixis, Inc. Methods of treating cancer using pyridopyrimidinone inhibitors of pi3k alpha

Non-Patent Citations (40)

* Cited by examiner, † Cited by third party
Title
"Bioreversible Carriers in Drug Design", 1987, AMERICAN PHARMACEUTICAL ASSOCIATION AND PERGAMON PRESS
"Larock's Comprehensive Organic Transformations", 1989, VCH PUBLISHERS INC.
"March's Advanced Organic Chemistry", JOHN WILEY AND SONS
"Organic Reactions", vol. 1-40, 1991, JOHN WILEY AND SONS
"Remington's Pharmaceutical Sciences", 1985, MACK PUBLISHING COMPANY
"Remington's Pharmaceutical Sciences", 1990, MACK PUBLISHING COMPANY
"Rodd's Chemistry of Carbon Compounds", vol. 1-5, 1989, ELSEVIER SCIENCE PUBLISHERS
BACHMAN ET AL., CANCER BIOL THER, vol. 3, 2004, pages 772 - 775
BYUN ET AL., INT J CANCER, vol. 104, 2003, pages 318 - 327
CAMPBELL ET AL., CANCER RES, vol. 64, 2004, pages 7678 - 7681
COHEN, EUR. J. BIOCHEM., vol. 268, 2001, pages 5001 - 5010
COHEN, NATURE, vol. 1, 2002, pages 309 - 315
FIESER; FIESER: "Reagents for Organic Synthesis", vol. 1-17, 1991, JOHN WILEY AND SONS
GOODMAN; GILMAN ET AL.: "The Pharmacological Basis of Therapeutics", 1990, PERGAMON PRESS
HARTMANN ET AL., ACTA NEUROPATHOL (BERL, vol. 109, 2005, pages 639 - 642
HICKEY; COTTER, J BIOL CHEM, vol. 281, 2006, pages 2441 - 2450
J. KASPAREC ET AL., TETRAHEDRON LETTERS, vol. 44, 2003, pages 4567 - 4570
LEE ET AL., GYNECOL ONCOL, vol. 97, 2005, pages 26 - 34
LEE ET AL., ONCOGENE, vol. 24, 2005, pages 1477 - 1480
LEVINE ET AL., CLIN CANCER RES, vol. 11, 2005, pages 2875 - 2878
LI ET AL., BREAST CANCER RES TREAT, vol. 96, 2006, pages 91 - 95
M. BARVIAN ET AL., J. MED. CHEM., vol. 43, 2000, pages 4606 - 4616
MASSION ET AL., AM J RESPIR CRIT CARE MED, vol. 170, 2004, pages 1088 - 1094
ODA ET AL., CANCERRES., vol. 65, 2005, pages 10669 - 10673
P. L. TOOGOOD ET AL., J. MED. CHEM., vol. 48, 2005, pages 2388 - 2406
PARK ET AL., CELL, vol. 101, no. 7, 2000, pages 777 - 787
S. M. BERGE ET AL.: "Pharmaceutical Salts", J. PHARM. SCI., vol. 66, 1977, pages 1 - 19, XP002675560, DOI: doi:10.1002/jps.2600660104
S. N. VANDERWEI ET AL., J. MED. CHEM., vol. 48, 2005, pages 2371 - 2387
SAAL ET AL., CANCER RES, vol. 65, 2005, pages 2554 - 2559
SAMUELS ET AL., SCIENCE, vol. 304, 2004, pages 554
SAMUELS; VELCULESCU, CELL CYCLE, vol. 3, 2004, pages 1221 - 1224
SRIDHAR ET AL., PHARMACEUTICAL RESEARCH, vol. 17, no. 11, 2000, pages 1345 - 1353
SUJOBERT ET AL., BLOOD, vol. 106, 1997, pages 1063 - 1066
T. HIGUCHI; V. STELLA: "Pro-dmgs as Novel Delivery Systems", A.C.S. SYMPOSIUM SERIES, vol. 14
T. HIGUCHI; V. STELLA: "Pro-drugs as Novel Delivery Systems", vol. 14, A.C.S. SYMPOSIUM SERIES
T.W. GREENE: "Protective Groups in Organic Synthesis", 1991, JOHN WILEY & SONS, INC.
TANG ET AL., LUNG CANCER, vol. 51, 2006, pages 181 - 191
VELHO ET AL., EUR J CANCER, vol. 41, 2005, pages 1649 - 1654
WANG ET AL., HUM MUTAL, vol. 25, 2005, pages 322
WU ET AL., J CLIN ENDOCRINOL METAB, vol. 90, 2005, pages 4688 - 4693

Cited By (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9533967B2 (en) 2010-03-30 2017-01-03 Verseon Corporation Multisubstituted aromatic compounds as inhibitors of thrombin
WO2012149308A1 (en) * 2011-04-29 2012-11-01 Exelixis, Inc. Method of treating lymphoma using pyridopyrimidinone inhibitors of pi3k/mtor
WO2012174327A1 (en) * 2011-06-15 2012-12-20 Exelixis, Inc. Combination therapies for treating hematologic malignancies using pyridopyrimidinone inhibitors of pi3k/mtor with bendamustine and/or rituximab
US9255108B2 (en) 2012-04-10 2016-02-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US8940742B2 (en) 2012-04-10 2015-01-27 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
WO2014058947A1 (en) * 2012-10-12 2014-04-17 Sanofi Compositions and methods for treating cancer using pi3k inhibitor and anti-cd19 maytansinoid immunoconjugate
WO2014135572A1 (en) * 2013-03-05 2014-09-12 Sanofi Tablet formulation of a pi3kalpha inhibitor
WO2014145986A1 (en) * 2013-03-15 2014-09-18 Verseon, Inc. Multisubstituted aromatic compounds as serine protease inhibitors
US9951025B2 (en) 2013-03-15 2018-04-24 Verseon Corporation Halogenopyrazoles as inhibitors of thrombin
CN105324117A (en) * 2013-03-15 2016-02-10 维颂公司 Multisubstituted aromatic compounds as serine protease inhibitors
US9533970B2 (en) 2013-03-15 2017-01-03 Verseon Corporation Multisubstituted aromatic compounds as serine protease inhibitors
US9687479B2 (en) 2013-03-15 2017-06-27 Verseon Corporation Multisubstituted aromatic compounds as serine protease inhibitors
US10251872B2 (en) 2013-03-15 2019-04-09 Verseon Corporation Multisubstituted aromatic compounds as serine protease inhibitors
US10058541B2 (en) 2013-03-15 2018-08-28 Verseon Corporation Multisubstituted aromatic compounds as serine protease inhibitors
CN108354933A (en) * 2013-03-15 2018-08-03 维颂公司 The pharmaceutical applications of polysubstituted aromatic compounds as serpin
US9359365B2 (en) 2013-10-04 2016-06-07 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10329299B2 (en) 2013-10-04 2019-06-25 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9751888B2 (en) 2013-10-04 2017-09-05 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9828377B2 (en) 2013-10-04 2017-11-28 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9775844B2 (en) 2014-03-19 2017-10-03 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US11541059B2 (en) 2014-03-19 2023-01-03 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10675286B2 (en) 2014-03-19 2020-06-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10189810B2 (en) 2014-09-17 2019-01-29 Verseon Corporation Pyrazolyl-substituted pyridone compounds as serine protease inhibitors
US10253047B2 (en) 2014-10-03 2019-04-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9708348B2 (en) 2014-10-03 2017-07-18 Infinity Pharmaceuticals, Inc. Trisubstituted bicyclic heterocyclic compounds with kinase activities and uses thereof
US10941162B2 (en) 2014-10-03 2021-03-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10532995B2 (en) 2015-02-27 2020-01-14 Verseon Corporation Substituted pyrazole compounds as serine protease inhibitors
US11247995B2 (en) 2015-09-14 2022-02-15 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
US10160761B2 (en) 2015-09-14 2018-12-25 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
US11939333B2 (en) 2015-09-14 2024-03-26 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
US10322192B2 (en) 2016-03-02 2019-06-18 Eisai R&D Management Co., Ltd. Eribulin-based antibody-drug conjugates and methods of use
US10548986B2 (en) 2016-03-02 2020-02-04 Eisai R&D Management Co., Ltd. Eribulin-based antibody-drug conjugates and methods of use
US10759806B2 (en) 2016-03-17 2020-09-01 Infinity Pharmaceuticals, Inc. Isotopologues of isoquinolinone and quinazolinone compounds and uses thereof as PI3K kinase inhibitors
US10919914B2 (en) 2016-06-08 2021-02-16 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
CN115348963A (en) * 2021-03-08 2022-11-15 暨南大学 Pyridopyrimidine compound and application thereof

Also Published As

Publication number Publication date
WO2012065019A3 (en) 2012-10-04

Similar Documents

Publication Publication Date Title
EP2322523B1 (en) Process for the preparation of Pyridopyrimidinone Inhibitors of PI3Kalpha
WO2012065019A2 (en) Pyridopyrimidinone inhibitors of p13k alpha
JP6043754B2 (en) PI3Kα pyridopyrimidinone type inhibitor
EP2142543B1 (en) Pyrido [2, 3-d]pyrimidin-7-one compounds as inhibitors of pi3k-alpha for the treatment of cancer
EP2139484A1 (en) Methods of treating cancer using pyridopyrimidinone inhibitors of pi3k alpha
AU2008239596A1 (en) Pyrido [2,3-D] pyrimidin-7-one compounds as inhibitors of PI3K-alpha for the treatment of cancer
AU2013200931B2 (en) Pyridopyrimidinone inhibitors of PI3K alpha

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: 11788286

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11788286

Country of ref document: EP

Kind code of ref document: A2