WO2012135009A1 - Pyrimidine-4,6-diamine derivatives as pi3k inhibitors - Google Patents

Pyrimidine-4,6-diamine derivatives as pi3k inhibitors Download PDF

Info

Publication number
WO2012135009A1
WO2012135009A1 PCT/US2012/030310 US2012030310W WO2012135009A1 WO 2012135009 A1 WO2012135009 A1 WO 2012135009A1 US 2012030310 W US2012030310 W US 2012030310W WO 2012135009 A1 WO2012135009 A1 WO 2012135009A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
compound
amino
pharmaceutically acceptable
acceptable salt
Prior art date
Application number
PCT/US2012/030310
Other languages
French (fr)
Inventor
Andrew P. Combs
Yun-Long Li
Eddy W. Yue
Original Assignee
Incyte Corporation
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 Incyte Corporation filed Critical Incyte Corporation
Priority to US14/007,061 priority Critical patent/US9126948B2/en
Publication of WO2012135009A1 publication Critical patent/WO2012135009A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/48Two nitrogen atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C53/00Saturated compounds having only one carboxyl group bound to an acyclic carbon atom or hydrogen
    • C07C53/15Saturated compounds having only one carboxyl group bound to an acyclic carbon atom or hydrogen containing halogen
    • C07C53/16Halogenated acetic acids
    • C07C53/18Halogenated acetic acids containing fluorine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • 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

Definitions

  • the present invention provides pyrimidine-4,6-diamine derivatives that modulate the activity of phosphoinositide 3-kinases (PBKs) and are useful in the treatment of diseases related to the activity of PBKs including, for example, inflammatory disorders, immune- based disorders, cancer, and other diseases.
  • PBKs phosphoinositide 3-kinases
  • PBKs The phosphoinositide 3-kinases (PBKs) belong to a large family of lipid signaling kinases that phosphorylate phosphoinositides at the D3 position of the inositol ring (Cantley, Science, 2002, 296(5573): 1655-7). PBKs are divided into three classes (class I, II, and III) according to their structure, regulation and substrate specificity.
  • Class I PBKs which include ⁇ , ⁇ , ⁇ , and ⁇ , are a family of dual specificity lipid and protein kinases that catalyze the phosphorylation of phosphatidylinosito-4,5-bisphosphate (PIP 2 ) giving rise to phosphatidylinosito-3,4,5-trisphosphate (PIP 3 ).
  • PIP 3 functions as a second messenger that controls a number of cellular processes, including growth, survival, adhesion and migration. All four class I PI3K isoforms exist as heterodimers composed of a catalytic subunit (pi 10) and a tightly associated regulatory subunit that controls their expression, activation, and subcellular localization.
  • ⁇ , ⁇ , and ⁇ associate with a regulatory subunit known as p85 and are activated by growth factors and cytokines through a tyrosine kinase-dependent mechanism (Jimenez, et al., J Biol Chem., 2002, 277(44):41556- 62) whereas ⁇ associates with two regulatory subunits (pi 01 and p84) and its activation is driven by the activation of G-protein-coupled receptors (Brock, et al., J Cell Biol, 2003, 160(l):89-99).
  • PBKcc and ⁇ are ubiquitously expressed. In contrast, ⁇ and ⁇ are predominantly expressed in leukocytes (Vanhaesebroeck, et al., Trends Biochem Sci., 2005, 30(4): 194-204).
  • mice which lack ⁇ 3 ⁇ and PI3K8 are viable, fertile and have a normal life span although they show an altered immune system.
  • ⁇ 3 ⁇ deficiency leads to impaired recruitment of macrophages and neutrophils to sites of inflammation as well as impaired T cell activation (Sasaki, et al., Science, 2000, 287(5455): 1040-6).
  • mice have specific defects in B cell signaling that lead to impaired B cell development and reduced antibody responses after antigen stimulation (Clayton, et al, J Exp Med. 2002, 196(6):753-63; Jou, et al, Mol Cell Biol. 2002, 22(24):8580-91 ; Okkenhaug, et al. Science, 2002, 297(5583): 1031-4).
  • mice The phenotypes of the ⁇ 3 ⁇ and ⁇ -mutant mice suggest that these enzymes may play a role in inflammation and other immune-based diseases and this is borne out in preclinical models. ⁇ -mutant mice are largely protected from disease in mouse models of rheumatoid arthritis (RA) and asthma (Camps, et al, Nat Med. 2005, l l(9):936-43;
  • RA rheumatoid arthritis
  • asthma Camps, et al, Nat Med. 2005, l l(9):936-43;
  • PBK isoforms may play a role in cancer.
  • the gene encoding pi 10a is mutated frequently in common cancers, including breast, prostate, colon and endometrial (Samuels, et al. Science, 2004, 304(5670):554; Samuels, et al, Curr Opin Oncol. 2006, 18(l):77-82). Eighty percent of these mutations are represented by one of three amino acid substitutions in the helical or kinase domains of the enzyme and lead to a significant upregulation of kinase activity resulting in oncogenic transformation in cell culture and in animal models (Kang, et al, Proc Natl Acad Sci U S A. 2005, 102(3):802-7; Bader, et al, Proc Natl Acad Sci U S A. 2006,
  • autoimmune diseases e.g., multiple sclerosis, rheumatoid arthritis, asthma, type I diabetes, inflammatory bowel disease, Crohn's disease, autoimmune thyroid disorders, Alzheimer's disease, nephritis
  • diseases involving a hyperactive inflammatory response e.g. , eczema
  • allergies e.g., eczema
  • lung diseases cancer (e.g., prostate, breast, leukemia, multiple myeloma), and some immune reactions (e.g., skin rash or contact dermatitis or diarrhea) caused by other therapeutics.
  • cancer e.g., prostate, breast, leukemia, multiple myeloma
  • immune reactions e.g., skin rash or contact dermatitis or diarrhea
  • the present invention provides, inter alia, a compound of Formula I:
  • the present invention further provides compositions comprising a compound of the invention, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier.
  • the present invention also provides methods of modulating an activity of a PI3K kinase, comprising contacting the kinase with a compound of the invention, or a
  • the present invention further provides methods of treating a disease in a patient, wherein said disease is associated with abnormal expression or activity of a PI3K kinase, comprising administering to said patient a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof.
  • the present invention further provides methods of treating an immune-based disease in a patient, comprising administering to said patient a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof.
  • the present invention also provides methods of treating a cancer in a patient, comprising administering to said patient a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof.
  • the present invention further provides methods of treating a lung disease in a patient, comprising administering to said patient a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof.
  • the present invention also provides a compound of invention, or a pharmaceutically acceptable salt thereof, for use in any of the methods described herein.
  • the present invention further provides use of a compound, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for use in any of the methods described herein.
  • the present invention provides, inter alia, a compound of Formula I:
  • R 1 is Ci-6 alkyl, C 2- 6 alkenyl, C 2 -6 alkynyl, or Cue haloalkyl, wherein said Cue alkyl, C 2 -6 alkenyl, and C 2 _6 alkynyl are each optionally substituted by 1, 2, 3, or 4 groups independently selected from halo, OH, CN, NR l a R l b , Ci_6 alkoxy, and Ci- 6 haloalkoxy;
  • R 2 is H, halo, CN, C 6 alkyl, C 1-6 haloalkyl, -L l -(C U6 alkyl), or - -(C 6 haloalkyl), wherein said C 1-6 alkyl in said C e alkyl and -L l -(Cue alkyl) is optionally substituted by 1, 2, 3, or 4 independently selected R 2a groups;
  • Ci -6 alkyl, C 2-6 alkenyl, and C 2-6 alkynyl are each optionally substituted by 1, 2, 3, or 4 independently selected R 3a groups;
  • R 4 is H, halo, OH, CN, CM alkyl, CM haloalkyl, C alkoxy, or C 1-4 haloalkoxy;
  • R 5 is halo, OH, CN, CM alkyl, CM alkoxy, or CM haloalkoxy;
  • R 6 is H, halo, OH, CN, C alkyl, C 1-4 haloalkyl, C alkoxy, or CM haloalkoxy;
  • R 8 is H, CM alkyl, C M haloalkyl, or C(0)R 4a ;
  • L 1 is O, NR B , S, S(O), S(0) 2 , C(O), C(0)NR B , S(0)NR B , or S(0) 2 NR B ;
  • R A , R B , R c , and R D are each independently selected from H, Cue alkyl, and Ci. 6 haloalkyl;
  • Cy is selected from C3-7 cycloalkyl, 4-11 membered heterocycloalkyl, 6-10 membered aryl and 5-10 membered heteroaryl, each of which is substituted with 0, 1, 2, 3, or 4 independently selected R 3 groups;
  • each R l a and R lb is independently selected from H and CM alkyl
  • R la and R 2b together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group, which is optionally substituted with 1, 2, 3, or 4 substituents independently selected from C 1-6 alkyl;
  • each R 2 is independently selected from OH, N0 2 , CN, halo, C 1-6 alkyl, C 2- 6 alkenyl, C 2-
  • each R 2b and R 2c is independently selected from H, C] _6 alkyl, and Ci -6 haloalkyl;
  • each R 3 is independently selected from halo, CN, N0 2 , Ci_ alkyl, C 2 -6 alkenyl, C 2 -6 alkynyl, Ci_ 6 haloalkyl, C 3-7 cycloalkyl, 4-11 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, (C3.7 cycloalkyl)-Ci.4 alkyl, (4-11 membered
  • R a is Ci-6 alkyl, C3-7 cycloalkyl, 4-1 1 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, (C3.7 cycloalkyl)-Ci. 4 alkyl, (4-11 membered heterocycloalkyl)-C i_ 4 alkyl, (6-10 membered aryl)-C 1 -4 alkyl, and (5-10 membered heteroaryl)-Ci_ 4 alkyl;
  • each R a , R b , R c , and R d is independently selected from H, Ci -6 alkyl, C 1-6 haloalkyl, C 2-6 alkenyl, and C 2 - 6 alkynyl; wherein said C 1-6 alkyl, C 2- 6 alkenyl, and C 2 _6 alkynyl are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from OH, N0 2 , CN, halo, C alkyl, C 2- 6 alkenyl, C 2 _e alkynyl, C 1-6 haloalkyl, cyano-Ci_ 6 alkyl, HO-C1.6 alkyl, Ci ⁇ alkoxy-Ci-g alkyl, C 3-7 cycloalkyl, C 1-6 haloalkyl, C ⁇ alkoxy, Ci -6 haloalkoxy, amino, Ci -6 alkylamino, di(Ci_ 6 alkyl)amin
  • Ci_6 alkoxy C 1-6 haloalkoxy, amino, Ci -6 alkylamino, di(Q.6 alkyl)amino, thio, C 1-6 alkylthio, Ci -6 alley lsulfinyl, C 1 -6 alkylsulfonyl, carbamyl, C 1-6 alkylcarbamyl, di(C].6 alkyl)carbamyl, carboxy, C] -6 alkylcarbonyl, C .
  • Ci_6 alkylcarbonylamino Ci_ 6 alkylsulfonylamino, aminosulfonyl, Ci ⁇ alkylaminosulfonyl, di(Ci_6 alkyl)aminosulfonyl, aminosulfonylamino, Ci -6 alkylaminosulfonylamino, di(Ci_6
  • alkyl)aminosulfonylamino aminocarbonylamino, Ci-6 alkylaminocarbonylamino, and di(Ci_6 alkyl)aminocarbonylamino;
  • each R E is independently selected from H, Q.4 alkyl, CN, OH, CM alkoxy, C1-4 alkylsulfonyl, carbamyl, C 1.4 alkylcarbamyl, di(Ci_4 alkyl)carbamyl, and Q.4 alkylcarbonyl; each R AL , R BL , R CL , and R D L is independently selected from H, Ci_6 alkyl, Ci_ 6 haloalkyl, C2-6 alkenyl, C 2 _e alkynyl, C 3 -7 cycloalkyl, 4-1 1 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, (C 3 - 7 cycloalkyl)-Ci. alkyl, (4-1 1 membered
  • heterocycloalkyl -Ci-4 alkyl, (6-10 membered aryl)-Ci_4 alkyl, and (5-10 membered heteroaryl)-Ci-4 alkyl, wherein said Q.6 alkyl, C 2 . 6 alkenyl, C 2- 6 alkynyl, C3.7 cycloalkyl, 4-11 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, (03,7
  • cycloalkyl)-Ci_4 alkyl, (4-11 membered heterocycloalkyl)-Ci_4 alkyl, (6-10 membered aryl)- C1.4 alkyl, and (5-10 membered heteroaryl)-Ci_4 alkyl are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from OH, N0 2 , CN, halo, Ci -6 alkyl, C 2- 6 alkenyl, C 2 _6 alkynyl, Ci_6 haloalkyl, cyano-Ci_6 alkyl, HO-Ci-6 alkyl, C 1-4 alkoxy-Ci.6 alkyl, C 3 -7 cycloalkyl, C 1-6 haloalkyl, Ci- ⁇ alkoxy, Ci -6 haloalkoxy, amino, C 1-6 alkylamino, di(C] _6 alkyl)amino, thio, Ci -6 alkylthio
  • alkyl)aminosulfonylamino aminocarbonylamino, C i -6 alkylaminocarbonylamino, and di(Ci -6 alkylaminocarbonylamino;
  • a 0, 1, 2, 3, or 4;
  • b is 0, 1, 2, 3, or 4;
  • n 1, 2, 3, 4, 5, or 6.
  • R 1 is C 1-6 alkyl.
  • R 1 is methyl
  • R 1 is ethyl
  • R 2 is -L 1 - ⁇ ] ⁇ alkyl
  • R 2 is C 1-6 alkoxy.
  • R 2 is methoxy
  • R 3 is Cy or -(CR 2b R 2c ) a -L 2 -(CR 2b R 2c ) b -Cy.
  • R 3 is Cy
  • R 3 is CN, N0 2 , C 2 _ 6 alkenyl, C 2- 6 alkynyl, Ci -6 haloalkyl,
  • R 3 is 5-10 membered heteroaryl substituted with 1, 2, or 3 groups independently selected from halo, CN, N0 2 , C 2- 6 alkenyl, C 2 - 6 alkynyl, Ci -6 haloalkyl, C3-7 cycloalkyl, 4-11 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, (C 3 .7 cycloalkyl)-C 1.4 alkyl, (4-1 1 membered heterocycloalkyl)-C 1 -4 alkyl, (6-10 membered aryl)-Ci_ 4 alkyl, and (5-10 membered heteroaryl)-Ci_ 4 alkyl, OR al , SR al ,
  • R 3 is 6-10 membered aryl substituted with 1, 2, or 3 groups independently selected from N0 2 , C 2 _6 alkenyl, C 2-6 alkynyl, Ci -6 haloalkyl, C 3-7 cycloalkyl, 4-11 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, (C 3- 7 cycloalkyl)-Ci-4 alkyl, (4-11 membered heterocycloalkyl)-C 1-4 alkyl, (6-10 membered aryl)- C alkyl, and (5-10 membered heteroaryl)-Ci. 4 alkyl, SR al , C(0)R bl , C(0)NR cl R dl ,
  • Cy is selected from 6-10 membered aryl and 5-10 membered heteroaryl, each of which is substituted with 0, 1, 2, 3, or 4 independently selected R 3a groups.
  • Cy is phenyl substituted with 0, 1, 2, 3, or 4 independently selected R 3a groups.
  • Cy is 5-6 membered heteroaryl substituted with 0, 1, 2, 3, or 4 independently selected R 3a groups. In some embodiments, Cy is pyridyl or pyrimidinyl substituted with 0, 1, 2, 3, or 4 independently selected R 3 groups.
  • Cy is pyridyl substituted with 0, 1, 2, 3, or 4 independently selected R 3a groups.
  • Cy is pyrimidinyl substituted with 0, 1, 2, 3, or 4 independently selected R 3a groups.
  • Cy is substituted with 1, 2, or 3 independently selected R 3a groups.
  • R 3a is independently selected from halo, CN, NO2, C 2 _6 alkenyl, C 2 -6 alkynyl, Ci_6 haloalkyl, C3-7 cycloalkyl, 4-1 1 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, (C3-7 cycloalkyl)-C] _4 alkyl, (4-1 1 membered heterocycloalkyl)-Ci_4 alkyl, (6-10 membered aryl)-C 1.4 alkyl, and (5-10 membered heteroaryl)-C, _4 alkyl, OR al , SR al , C(0)NR cl R dl OC(0)R bl , OC(0)NR cl R dl , NR cl C(0)R bl , NR cl C(0)OR bl , NR cl C(0)NR cl R dl
  • R 3a is independently selected from N0 2 , C 2 -6 alkenyl, C 2 -6 alkynyl, Ci -6 haloalkyl, C 3-7 cycloalkyl, 4-11 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, (C 3 _ 7 cycloalkyl)-Ci. 4 alkyl, (4-11 membered
  • heterocycloalkyl -C 1 -4 alkyl, (6-10 membered aryl)-C 1 -4 alkyl, and (5-10 membered heteroaryl)-C M alkyl, SR al , C(0)R bl , C(0)NR cl R dl , OC(0)R bl , OC(0)NR cl R dl ,
  • R 3a is independently selected from halo, CN, C(0)NR cl R dl , NR cl R dl , and S(0) 2 R bl .
  • R 3a is independently selected from halo, CN, C(0)NR cl R dl , and S(0) 2 R bl .
  • R 3a is S(0) 2 R bl .
  • R 3a is S(0) 2 -(Ci_4 alkyl).
  • R 3a is S(0) 2 -CH 3 .
  • R 3a is NH 2 .
  • R 3a is other than CN, halo, OH, Ci -6 alkyl, Ci -6 alkoxy, -Nt3 ⁇ 4, -NH(C 1-6 alkyl), -N(C 1-6 alkyl) 2 , -(C 1-6 alkyl)-NH 2 , -(C 1-6 alkyl)-NH(C 1-6 alkyl), -(C 1-6 alkyl)- N(C 1-6 alkyl) 2 , or -C(0)0-(C 1-6 alkyl);
  • R 3a is other than Ci_6 alkyl, -(Ci_6 alkyl)-NH 2 , -(Ci -6 alkyl)- NH(Ci. 6 alkyl), -(Ci_ 6 alkyl)-N(Ci -6 alkyl) 2 , -C(0)-(C,. 6 alkyl), -C(0)OH, or -C(0)0-(Ci -6 alkyl);
  • R 4 is halo, CN, C1.4 alkyl, or Q ⁇ haloalkyl.
  • R 4 is halo
  • R 4 is methyl
  • R 4 is F.
  • R 4 is CI
  • R 5 is CI, F, or methyl.
  • R 5 is CI
  • R 6 is H.
  • R 7 is CN
  • R 8 is H.
  • R A is H.
  • Cy when Cy is 6-10 membered aryl, then said 6-10 membered aryl is substituted by at least one group selected from N0 2 , C 2 _6 alkenyl, C 2- 6 alkynyl, Ci_6 haloalkyl, C 3-7 cycloalkyl, 4-11 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, (C 3 .
  • the compound has Formula II:
  • the compound has Formula III:
  • each divalent linking substituent include both the forward and backward forms of the linking substituent.
  • -NR(CR'R") n - includes both -NR(CR'R") n - and -(CR'R") n NR-.
  • the Markush variables listed for that group are understood to be linking groups.
  • n-membered where n is an integer typically describes the number of ring- forming atoms in a moiety where the number of ring-forming atoms is n.
  • piperidinyl is an example of a 6-membered heterocycloalkyl ring
  • pyrazolyl is an example of a 5-membered heteroaryl ring
  • pyridyl is an example of a 6-membered heteroaryl ring
  • 1,2,3,4-tetrahydro-naphthalene is an example of a 10-membered cycloalkyl group.
  • C n . m indicates a range which includes the endpoints, wherein n and m are integers and indicate the number of carbons. Examples include C , C 1 -6 , and the like.
  • C N - M alkyl refers to a saturated hydrocarbon group that may be straight-chain or branched, having n to m carbons.
  • the alkyl group contains from 1 to 6 carbon atoms, from 1 to 4 carbon atoms, from 1 to 3 carbon atoms, or 1 to 2 carbon atoms.
  • alkyl moieties include, but are not limited to, chemical groups such as methyl, ethyl, n- propyl, isopropyl, w-butyl, te -butyl, isobutyl, ec-butyl; higher homologs such as 2-methyl- 1-butyl, «-pentyl, 3-pentyl, «-hexyl, 1,2,2-trimethylpropyl, and the like.
  • C N-M alkenyl refers to an alkyl group having one or more double carbon-carbon bonds and having n to m carbons. In some embodiments, the alkenyl moiety contains 2 to 6 or to 2 to 4 carbon atoms.
  • Example alkenyl groups include, but are not limited to, ethenyl, «-propenyl, isopropenyl, «-butenyl, sec-butenyl, and the like.
  • C N _ M alkynyF' refers to an alkyl group having one or more triple carbon-carbon bonds and having n to m carbons.
  • Example alkynyl groups include, but are not limited to, ethynyl, propyn-l-yl, propyn-2-yl, and the like.
  • the alkynyl moiety contains 2 to 6 or 2 to 4 carbon atoms.
  • C N _ M alkoxy refers to a group of formula -O-alkyl, wherein the alkyl group has n to m carbons.
  • Example alkoxy groups include methoxy, ethoxy, propoxy (e.g., n-propoxy and isopropoxy), t-butoxy, and the like.
  • the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
  • C n -m alkylamino refers to a group of formula -NH(alkyl), wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
  • C N . M alkoxycarbonyl refers to a group of formula -C(0)0- alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
  • C N-M alkylcarbonyl refers to a group of formula -C(O)- alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
  • C n-m alkylcarbonylamino refers to a group of formula -NHC(0)-alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
  • C n . m alkylsulfonylamino refers to a group of formula -NHS(0) 2 -alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
  • aminosulfonyl refers to a group of formula -S(0) 2 NH 2 , wherein the alkyl group has n to m carbon atoms.
  • C n-m alkylaminosulfonyl refers to a group of formula -S(0) 2 NH(alkyl), wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
  • di(C n _ m alkyl)aminosulfonyl refers to a group of formula -S(0) 2 N(alkyl) 2 , wherein each alkyl group independently has n to m carbon atoms. In some embodiments, each alkyl group has, independently, 1 to 6 or 1 to 4 carbon atoms.
  • aminosulfonylamino refers to a group of formula -
  • C n . m alkylaminosulfonylamino refers to a group of formula -NHS(0) 2 NH(alkyl), wherein the alkyl group has n to m carbon atoms.
  • the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
  • di(C n . m alkyl)aminosulfonylamino refers to a group of formula -NHS(0) 2 N(alkyl) 2 , wherein each alkyl group independently has n to m carbon atoms. In some embodiments, each alkyl group has, independently, 1 to 6 or 1 to 4 carbon atoms.
  • aminocarbonylamino employed alone or in combination with other terms, refers to a group of formula -NHC(0)NH 2 .
  • C n - m alkylaminocarbonylamino refers to a group of formula -NHC(0)NH(alkyl), wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
  • di(C n-m alkyl)aminocarbonylamino refers to a group of formula -NHC(0)N(alkyl) 2 , wherein each alkyl group independently has n to m carbon atoms. In some embodiments, each alkyl group has, independently, 1 to 6 or 1 to 4 carbon atoms.
  • C n-m alkylcarbamyl refers to a group of formula -C(O)- NH(alkyl), wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
  • thio refers to a group of formula -S-H.
  • C n . m alkylthio refers to a group of formula -S-alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
  • C n - m alkylsulfinyl refers to a group of formula -S(0)-alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
  • C n - m alkylsulfonyl refers to a group of formula -S(0) 2 - alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
  • amino refers to a group of formula -NH 2 .
  • carbonyl employed alone or in combination with other terms, refers to a -C(O)- group.
  • carboxy refers to a group of formula -C(0)OH.
  • di(C n-m -alkyl)amino refers to a group of formula - N(alkyl) 2 , wherein the two alkyl groups each has, independently, n to m carbon atoms. In some embodiments, each alkyl group independently has 1 to 6 or 1 to 4 carbon atoms.
  • di(C n - m -alkyl)carbamyl refers to a group of formula - C(0)N(alkyl) 2 , wherein the two alkyl groups each has, independently, n to m carbon atoms. In some embodiments, each alkyl group independently has 1 to 6 or 1 to 4 carbon atoms.
  • halo refers to F, CI, Br, or I. In some embodiments, the halo group is F or CI.
  • C n-m haloalkoxy refers to a group of formula -O-haloalkyl having n to m carbon atoms.
  • An example haloalkoxy group is OCF 3 .
  • the haloalkoxy group is fluorinated only.
  • the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
  • C n _ m haloalkyl refers to an alkyl group having from one halogen atom to 2s+l halogen atoms which may be the same or different, where "s" is the number of carbon atoms in the alkyl group, wherein the alkyl group has n to m carbon atoms.
  • the haloalkyl group is fluorinated only.
  • the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
  • phenyl-Ci-4 alkyl refers to a group of formula -C
  • 6-10 membered aryl refers to a monocyclic or polycyclic aromatic hydrocarbon ring having 6-10 ring members.
  • the aryl group is phenyl.
  • the aryl group is naphthyl.
  • (6-10 membered aryl)-C n-m alkyl refers to a C n . m alkyl group substituted by an aryl group.
  • cycloalkyl refers to non-aromatic cyclic hydrocarbons including cyclized alkyl and/or alkenyl groups.
  • Cycloalkyl groups can include mono- or polycyclic (e.g., having 2, 3 or 4 fused rings) groups and spirocycles. Cycloalkyl groups can have 3, 4, 5, 6, or 7 ring-forming carbons (C 3 . 7 ). Ring-forming carbon atoms of a cycloalkyl group can be optionally substituted by oxo or sulfido. Cycloalkyl groups also include cycloalkylidenes.
  • Example cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclopentenyl, cyclohexenyl, cyclohexadienyl, cycloheptatrienyl, norbornyl, norpinyl, norcarnyl, adamantyl, and the like.
  • cycloalkyl is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.
  • cycloalkyl moieties that have one or more aromatic rings fused (i.e., having a bond in common with) to the cycloalkyl ring, for example, benzo or thienyl derivatives of cyclopentane, cyclohexane, and the like.
  • a cycloalkyl group containing a fused aromatic ring can be attached through any ring-forming atom including a ring-forming atom of the fused aromatic ring.
  • (C3-7 cycloalkyl)-C n - m alkyl refers to a C n-m alkyl group substituted by a C3-7 cycloalkyl group.
  • 5-10 membered heteroaryl refers to a monocyclic or polycyclic aromatic heterocycle having at least one heteroatom ring member selected from sulfur, oxygen, and nitrogen.
  • the heteroaryl ring has 1, 2, 3, 4, or 5 heteroatom ring members independently selected from nitrogen, sulfur and oxygen.
  • the heteroaryl ring has 1, 2, 3 or 4 N heteroatom ring members.
  • any ring-forming N in a heteroaryl moiety can be an N-oxide.
  • the 5-10 membered heteroaryl group is a five-membered heteroaryl ring.
  • the 5-10 membered heteroaryl is a 6-membered heteroaryl ring.
  • a five-membered heteroaryl ring is a heteroaryl with a ring having five ring atoms wherein one or more (e.g., 1, 2, or 3) ring atoms are independently selected from N, O, and S.
  • Exemplary five-membered ring heteroaryls are thienyl, furyl, pyrrolyl, imidazolyl, thiazolyl, oxazolyl, pyrazolyl, isothiazolyl, isoxazolyl, 1,2,3-triazolyl, tetrazolyl, 1,2,3-thiadiazolyl, 1,2,3-oxadiazolyl, 1,2,4-triazolyl, 1,2,4-thiadiazolyl, 1,2,4-oxadiazolyl, 1,3,4-triazolyl, 1,3,4-triazolyl, 1,3,4- thiadiazolyl, and 1,3,4-oxadiazolyl.
  • a six-membered heteroaryl ring is a heteroaryl with a ring having six ring atoms wherein one or more (e.g., 1, 2, or 3) ring atoms are independently selected from N, O, and S.
  • Exemplary six-membered ring heteroaryls are pyridyl, pyrazinyl, pyrimidinyl, triazinyl and pyridazinyl.
  • (5-10 membered heteroaryl)-C n-m alkyl refers to a C n-m alkyl group substituted by a 5-10 membered heteroaryl group.
  • 4-1 1 membered heterocycloalkyi refers to non-aromatic monocyclic or polycyclic heterocycles having one or more ring-forming heteroatoms selected from O, N, or S and having 4-11 ring members. Included in 4-1 1 membered heterocycloalkyi groups are monocyclic 4-, 5-, 6-, and 7-membered heterocycloalkyi groups. Heterocycloalkyi groups can also include spirocycles.
  • Example "4-1 1 membered heterocycloalkyi" groups include pyrrolidin-2-one, l,3-isoxazolidin-2-one, pyranyl, azetidinyl, morpholino, thiomorpholino, piperazinyl, tetrahydrofuranyl, tetrahydrothienyl, piperidinyl, pyrrolidinyl, isoxazolidinyl, isothiazolidinyl, pyrazolidinyl, oxazolidinyl, thiazolidinyl, imidazolidinyl, azepanyl, benzazapene, and the like.
  • Ring-forming carbon atoms and heteroatoms of a heterocycloalkyi group can be optionally substituted by oxo or sulfido (e.g., C(O), S(O), C(S), or S(0) 2 , etc.).
  • the heterocycloalkyi group can be attached through a ring-forming carbon atom or a ring- forming heteroatom.
  • the heterocycloalkyi group contains 0 to 3 double or triple bonds.
  • the heterocycloalkyi group contains 0 to 2 double bonds.
  • heterocycloalkyi moieties that have one or more aromatic rings fused (i.e., having a bond in common with) to the cycloalkyl ring, for example, benzo or thienyl derivatives of piperidine, morpholine, azepine, etc.
  • a heterocycloalkyi group containing a fused aromatic ring can be attached through any ring-forming atom including a ring-forming atom of the fused aromatic ring.
  • (4-11 membered heterocycloalkyl)-C n-m alkyl refers to a C n-m alkyl group substituted by a 4-11 membered heterocycloalkyi.
  • the compounds described herein can be asymmetric (e.g., having one or more stereocenters). All stereoisomers, such as enantiomers and diastereomers, are intended unless otherwise indicated.
  • the compound has the ( ⁇ -configuration. In some embodiments, the compound has the ( ⁇ -configuration.
  • the compound has the ( ⁇ -configuration.
  • An example method includes fractional recrystallizaion using a chiral resolving acid which is an optically active, salt-forming organic acid.
  • Suitable resolving agents for fractional recrystallization methods are, for example, optically active acids, such as the D and L forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or the various optically active camphorsulfonic acids such as ⁇ -camphorsulfonic acid.
  • resolving agents suitable for fractional crystallization methods include stereoisomerically pure forms of a-methylbenzylamine (e.g., S and R forms, or diastereomerically pure forms), 2-phenylglycinol, norephedrine, ephedrine, N- methylephedrine, cyclohexylethylamine, 1,2-diaminocyclohexane, and the like.
  • Resolution of racemic mixtures can also be carried out by elution on a column packed with an optically active resolving agent (e.g., dinitrobenzoylphenylglycine).
  • an optically active resolving agent e.g., dinitrobenzoylphenylglycine
  • Suitable elution solvent composition can be determined by one skilled in the art.
  • Tautomeric forms result from the swapping of a single bond with an adjacent double bond together with the concomitant migration of a proton.
  • Tautomeric forms include prototropic tautomers which are isomeric protonation states having the same empirical formula and total charge.
  • Example prototropic tautomers include ketone - enol pairs, amide - imidic acid pairs, lactam - lactim pairs, enamine - imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, for example, IH- and 3H-imidazole, 1H-, 2H- and 4H- 1,2,4-triazole, IH- and 2H- isoindole, and IH- and 2H-pyrazole. Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution.
  • Compounds of the invention can also include all isotopes of atoms occurring in the intermediates or final compounds. Isotopes include those atoms having the same atomic number but different mass numbers. For example, isotopes of hydrogen include tritium and deuterium.
  • compound as used herein is meant to include all stereoisomers, geometric iosomers, tautomers, and isotopes of the structures depicted. Compounds herein identified by name or structure as one particular tautomeric form are intended to include other tautomeric forms unless otherwise specified.
  • All compounds, and pharmaceutically acceptable salts thereof, can be found together with other substances such as water and solvents (e.g. hydrates and solvates) or can be isolated.
  • the compounds of the invention, or salts thereof are substantially isolated.
  • substantially isolated is meant that the compound is at least partially or substantially separated from the environment in which it was formed or detected.
  • Partial separation can include, for example, a composition enriched in the compounds of the invention.
  • Substantial separation can include compositions containing at least about 50%, at least about 60%, at least about 70%, at least about 80%o, at least about 90%>, at least about 95%, at least about 97%, or at least about 99% by weight of the compounds of the invention, or salt thereof. Methods for isolating compounds and their salts are routine in the art.
  • phrases "pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • ambient temperature and “room temperature,” as used herein, are understood in the art, and refer generally to a temperature, e.g. a reaction temperature, that is about the temperature of the room in which the reaction is carried out, for example, a temperature from about 20 °C to about 30 °C.
  • the present invention also includes pharmaceutically acceptable salts of the compounds described herein.
  • pharmaceutically acceptable salts refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form.
  • examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • the pharmaceutically acceptable salts of the present invention include the conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods.
  • such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, non-aqueous media like ether, ethyl acetate, alcohols (e.g., methanol, ethanol, iso-propanol, or butanol) or acetonitrile (ACN) are preferred.
  • non-aqueous media like ether, ethyl acetate, alcohols (e.g., methanol, ethanol, iso-propanol, or butanol) or acetonitrile (ACN) are preferred.
  • non-aqueous media like ether, ethyl acetate, alcohols (e.g., methanol, ethanol, iso-propanol, or butanol) or acetonitrile (ACN) are preferred.
  • ACN acetonitrile
  • the reactions for preparing compounds of the invention can be carried out in suitable solvents which can be readily selected by one of skill in the art of organic synthesis.
  • suitable solvents can be substantially non-reactive with the starting materials (reactants), the intermediates, or products at the temperatures at which the reactions are carried out, e.g., temperatures which can range from the solvent's freezing temperature to the solvent's boiling temperature.
  • a given reaction can be carried out in one solvent or a mixture of more than one solvent.
  • suitable solvents for a particular reaction step can be selected by the skilled artisan.
  • Reactions can be monitored according to any suitable method known in the art.
  • product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., ⁇ or 13 C), infrared spectroscopy, spectrophotometry (e.g., UV-visible), mass spectrometry, or by chromatographic methods such as high performance liquid chromatography (HPLC), liquid chromatography-mass spectroscopy (LCMS), or thin layer chromatography (TLC).
  • HPLC high performance liquid chromatography
  • LCMS liquid chromatography-mass spectroscopy
  • TLC thin layer chromatography
  • Compounds can be purified by those skilled in the art by a variety of methods, including high performance liquid chromatography (HPLC) ( "Preparative LC-MS Purification: Improved Compound Specific Method
  • Compounds of Formula I can be formed as shown in Scheme I.
  • the halo group of (ii) can be coupled to R 3 -M, where M is a boronic acid, boronic ester or an appropriately substituted metal (e.g., R 3 -M is R 3 -B(OH) 2 or R -Sn(Bu)4), under standard Suzuki conditions or standard Stille conditions (e.g., in the presence of a palladium(O) catalyst, such as
  • R 3 -M can be a cyclic amine (where M is H and attached to the amine nitrogen) with coupling to compound (ii) being performed by heating in base or under Buchwald conditions (e.g., in the presence of a palladium(O) catalyst, such as tetrakis(triphenylphosphine)palladium(0) and a base (e.g., an alkoxide base)) to afford ketone (iii).
  • a palladium(O) catalyst such as tetrakis(triphenylphosphine)palladium(0) and a base (e.g., an alkoxide base)
  • Reductive amination of the ketone (iii) can furnish the amine intermediate (v).
  • ketone (iii) can be reduced to give an alcohol which can be converted to the mesylate and reacted with sodium azide to give an azide derivative (iv).
  • the azide of compound (iv) can be converted to an amine (v) under appropriate reducing conditions, such as trimethylphosphine or TMSI.
  • the amine (v) can be reacted with an appropriate alkylating agent R A X (e.g., Mel) or reacted under reductive amination conditions to give compound (vi).
  • R A X e.g., Mel
  • compound (vi) can be reacted with a heteroaryl halide compound (e.g., Ar-X, such as 4-amino-6-chloropyrimidine-5-carbonitrile) to give a compound of Formula I.
  • a heteroaryl halide compound e.g., Ar-X, such as 4-amino-6-chloropyrimidine-5-carbonitrile
  • the reaction of amine (v) with R A -X can be skipped to give compounds of Formula I, wherein R A is H.
  • Ketone (ii) can be reduced to give an alcohol (iii) which can be converted to the mesylate and reacted with sodium azide to give an azide derivative (iv).
  • the azide of compound (iv) can be converted to an amine (v) under appropriate reducing conditions, such as
  • the amine (v) can be protected with a suitable protecting group (e.g., by reacting with Boc 2 0) and purified by chiral chromatography to afford a single enantiomer of amine compound (v).
  • R A X e.g., Mel
  • a heteroaryl halide compound e.g., Ar-X
  • the reaction of amine (v) with R A -X can be eliminated to give compounds (vi), wherein R A is H.
  • the halo group of (vi) can be coupled to R 3 -M, where M is a boronic acid, boronic ester or an appropriately substituted metal (e.g., R 3 -M is R 3 -B(OH) 2 or R 3 -Sn(Bu) 4 ), under standard Suzuki conditions or standard Stille conditions (e.g., in the presence of a palladium(O) catalyst, such as tetrakis(triphenylphosphine)-palladium(0) and a base (e.g., a bicarbonate or carbonate base)) to give a derivative of formula (vii).
  • M is a boronic acid, boronic ester or an appropriately substituted metal
  • R 3 -M is R 3 -B(OH) 2 or R 3 -Sn(Bu) 4
  • a palladium(O) catalyst such as tetrakis(triphenylphosphine)-palladium(0) and a base (e.g., a bi
  • R 3 -M can be a cyclic amine (where M is H and attached to the amine nitrogen) with coupling to compound (vi) being performed by heating in base or under Buchwald conditions (e.g., in the presence of a palladium(O) catalyst, such as
  • Mitsunobu conditions e.g., R OH, DEAD, Ph 3 P
  • R -Lg, Lg leaving group
  • M is a boronic acid, boronic ester or an appropriately substituted metal
  • R 3 -M is R 3 -B(OH) 2 or R 3 -Sn(Bu) 4
  • a palladium(O) catalyst such as tetrakis(triphenylphosphine)-pal
  • R 3 -M can be a cyclic amine (where M is H and attached to the amine nitrogen) with coupling to compound (ii) being performed by heating in base or under Buchwald conditions (e.g., in the presence of a palladium(O) catalyst, such as tetralds(triphenylphosphine)-palladium(0) and a base (e.g., an alkoxide base)) to afford compounds of formula (iii).
  • the ketone (iii) can be transformed using similar methods as shown in Scheme I and II to afford compounds of Formula I (iv).
  • the halo- ketone (ii) can be transformed using similar methods as shown in Scheme I and II to afford halo intermediate (v). Suzuki, Stille, Negishi or Buchwald coupling of R 3 -M with halo intermediate (v) by similar methods described in Schemes I and II can also afford compounds of Formula I (iv).
  • Compounds of Formula I can be formed as shown in Scheme IV.
  • Compound (i) can be acylated with a suitable acylating reagent (e.g., R'-COCl) to form an ester which can be rearranged under Lewis acid conditions e.g., BF 3 /HOAC complex) to afford ketone (ii).
  • a suitable acylating reagent e.g., R'-COCl
  • ketone (ii) e.g., R'-COCl
  • X CI, Br, or I.
  • the phenol can be converted to the triflate (iv) using standard conditions (e.g.,Tf 2 0).
  • the triflate group of (iv) can be coupled to R 2 -M, where M is a boronic acid, boronic ester or an appropriately substituted metal (e.g., R 2 -M is R 2 -B(OH) 2 or R 2 -Sn(Bu) 4 ), under standard Suzuki conditions or standard Stille conditions (e.g., in the presence of a palladium(O) catalyst, such as tetrakis(triphenylphosphine)palladium(0) and a base (e.g., a bicarbonate or carbonate base)) to give a derivative of formula (v).
  • a palladium(O) catalyst such as tetrakis(triphenylphosphine)palladium(0) and a base (e.g., a bicarbonate or carbonate base)
  • R 2 -M can be a cyclic amine (where M is H and attached to the amine nitrogen) with coupling to compound (iv) being performed by heating in base or under Buchwald conditions (e.g., in the presence of a palladium(O) catalyst, such as tetrakis(triphenylphosphine)palladium(0) and a base (e.g., an alkoxide base)) to afford ketone (v).
  • a palladium(O) catalyst such as tetrakis(triphenylphosphine)palladium(0) and a base (e.g., an alkoxide base)
  • the halo group of (v) can be coupled to R 3 -M, where M is a boronic acid, boronic ester or an appropriately substituted metal (e.g., R 3 -M is R 3 -B(OH) 2 or R 3 -Sn(Bu) 4 ), under standard Suzuki conditions or standard Stille conditions (e.g., in the presence of a palladium(O) catalyst, such as tetrakis(triphenylphosphine)palladium(0) and a base (e.g., a bicarbonate or carbonate base)) to give a derivative of formula (vi).
  • M is a boronic acid, boronic ester or an appropriately substituted metal
  • R 3 -M is R 3 -B(OH) 2 or R 3 -Sn(Bu) 4
  • a palladium(O) catalyst such as tetrakis(triphenylphosphine)palladium(0) and a base (e.g., a bicarbonate
  • R 3 -M can be a cyclic amine (where M is H and attached to the amine nitrogen) with coupling to compound (iv) being performed by heating in base or under Buchwald conditions (e.g., in the presence of a palladium(O) catalyst, such as
  • ketone (vi) tetrakis(triphenylphosphine)palladium(0) and a base (e.g., an alkoxide base)) to afford ketone (vi).
  • a base e.g., an alkoxide base
  • the ketone (vi) can be transformed using similar methods as shown in Scheme I and II to afford compounds of Formula I (viii).
  • halo-ketone (v) can be transformed using similar methods as shown in Scheme I and II to afford halo intermediate (viii). Suzuki, Stille, Negishi or Buchwald coupling of M-R 3 with compound (viii) by similar methods described in Schemes I and II can also afford compounds of Formula I (vii).
  • Ketones which can be used in the processes of Scheme I, II and III can be formed as shown in Scheme V below.
  • the carboxylic acid (i) can be activated with a coupling agent (e.g., HBTU, HATU or EDC) and then reacted with N, 0-dimethylhydroxylamine to give a N- methoxy-N-methylcarboxamide derivative (ii).
  • the ketone (iii) can be transformed using similar methods as shown in Schemes I, II and III to afford compounds of Formula I.
  • Ketones which can be used in the processes of Scheme I, II and III, can also be formed as shown in Scheme VI below.
  • the carboxylic acid (i) can be activated with a coupling agent (e.g. HBTU or HATU) and then reacted with N, O-dimethylhydroxylamine to give a N-methoxy-N-methylcarboxamide.
  • M is a boronic acid, boronic ester or an appropriately substituted metal
  • R 3 -M is R 3 -B(OH) 2 or R 3 -Sn(Bu) 4
  • a palladium(O) catalyst such as tetrakis(triphenylphosphine)palla
  • R 3 -M can be a cyclic amine (where M is H and attached to the amine nitrogen) with coupling to compound (ii) being performed by heating in base or under Buchwald conditions (e.g., in the presence of a palladium(O) catalyst, such as tetrakis(triphenylphosphine)palladium(0) and a base (e.g., an alkoxide base)) to afford amides (iii) of Formula I.
  • a palladium(O) catalyst such as tetrakis(triphenylphosphine)palladium(0) and a base (e.g., an alkoxide base)
  • a base e.g., an alkoxide base
  • the ketone (iv) can be transformed using similar methods as shown in Schemes I
  • Hydrolysis of the cyano group of (ii) under acid or base conditions can give the carboxylic acid which can be coupled to amines using a coupling agent (e.g., HATU, HBTU, EDC) and appropriate amines (HNR R y ) to give amide (iii) (R x and R y are various optionally substituted cyclic groups and non-cyclic groups, or R x and R y , along with the nitrogen atom to which they are attached can cyclize to form a heterocycloalkyl group).
  • the ketone of amide (iii) can be transformed using similar methods as shown in Schemes I, II and III to afford compounds of Formula I.
  • ketone (i) can be converted to the nitro-ketone (ii) using standard nitration conditions (e.g., HN0 3 ).
  • the ether (i) can be converted to a phenol (ii) using standard nitration conditions (e.g., BBr 3 ).
  • the halo-phenol (ii) can be converted to the cyano-phenol (iii) using standard cyanation conditions (e.g., CuCN or Pd(0) and Zn(CN) 2 ).
  • the phenol (iii) can be converted to the triflate (iv) using Tf 2 0.
  • the triflate group of (iv) can be coupled to R 2 -M, where M is a boronic acid, boronic ester or an appropriately substituted metal (e.g., R 2 - M is R 2 -B(OH) 2 or R 2 -Sn(Bu) 4 ), under standard Suzuki conditions or standard Stille conditions (e.g., in the presence of a palladium(O) catalyst, such as
  • R 2 -M can be a cyclic amine (where M is H and attached to the amine nitrogen) with coupling to compound (iv) being performed by heating in base or under Buchwald conditions (e.g., in the presence of a palladium(O) catalyst, such as tetrakis(triphenylphosphine)palladium(0) and a base (e.g., an alkoxide base)) to afford ketone (v).
  • a palladium(O) catalyst such as tetrakis(triphenylphosphine)palladium(0) and a base (e.g., an alkoxide base)
  • the compounds of the invention can modulate activity of one or more of various kinases including, for example, phosphoinositide 3-kinases (PBKs).
  • PBKs phosphoinositide 3-kinases
  • modulate is meant to refer to an ability to increase or decrease the activity of one or more members of the PI3K family.
  • the compounds of the invention can be used in methods of modulating a PI3K by contacting the PI3K with any one or more of the compounds or compositions described herein.
  • compounds of the present invention can act as inhibitors of one or more PBKs.
  • the compounds of the invention can be used to modulate activity of a PI3K in an individual in need of modulation of the receptor by administering a modulating amount of a compound of the invention, or a pharmaceutically acceptable salt thereof. In some embodiments, modulating is inhibiting.
  • the present invention is useful for treating disease states characterized by drug resistant kinase mutants.
  • different kinase inhibitors exhibiting different preferences in the kinases which they modulate the activities of, may be used in combination. This approach could prove highly efficient in treating disease states by targeting multiple signaling pathways, reduce the likelihood of drug-resistance arising in a cell, and reduce the toxicity of treatments for disease.
  • Kinases to which the present compounds bind and/or modulate (e.g., inhibit) include any member of the PI3K family.
  • the PI3K is ⁇ 3 ⁇ , ⁇ , ⁇ , or ⁇ .
  • the PBK is ⁇ or ⁇ .
  • the PBK is ⁇ .
  • the PBK is ⁇ .
  • the PBK includes a mutation.
  • a mutation can be a replacement of one amino acid for another, or a deletion of one or more amino acids. In such embodiments, the mutation can be present in the kinase domain of the PBK.
  • more than one compound of the invention is used to inhibit the activity of one kinase (e.g., ⁇ or ⁇ ).
  • more than one compound of the invention is used to inhibit more than one kinase, such as at least two kinases (e.g., ⁇ and ⁇ ).
  • one or more of the compounds is used in combination with another kinase inhibitor to inhibit the activity of one kinase (e.g., ⁇ or ⁇ ).
  • another kinase inhibitor e.g., ⁇ or ⁇ .
  • one or more of the compounds is used in combination with another kinase inhibitor to inhibit the activities of more than one kinase (e.g., ⁇ or ⁇ ), such as at least two kinases.
  • another kinase inhibitor to inhibit the activities of more than one kinase (e.g., ⁇ or ⁇ ), such as at least two kinases.
  • the compounds of the invention can be selective.
  • selective is meant that the compound binds to or inhibits a kinase with greater affinity or potency, respectively, compared to at least one other kinase.
  • the compounds of the invention are selective inhibitors of ⁇ or ⁇ over PBKa and/or ⁇ .
  • the compounds of the invention are selective inhibitors of ⁇ (e.g., over ⁇ , ⁇ and ⁇ ).
  • the compounds of the invention are selective inhibitors of ⁇ (e.g., over ⁇ , ⁇ and ⁇ ).
  • selectivity can be at least about 2-fold, 5-fold, 10-fold, at least about 20-fold, at least about 50-fold, at least about 100-fold, at least about 200-fold, at least about 500-fold or at least about 1000-fold.
  • Selectivity can be measured by methods routine in the art. In some embodiments, selectivity can be tested at the K m ATP concentration of each enzyme. In some embodiments, the selectivity of compounds of the invention can be determined by cellular assays associated with particular PBK kinase activity.
  • a PI3K-associated disease can include any disease, disorder or condition that is directly or indirectly linked to expression or activity of the PI3K, including overexpression and/or abnormal activity levels.
  • the disease can be linked to Akt (protein kinase B), mammalian target of rapamycin (mTOR), or phosphoinositide-dependent kinase 1 (PDK1).
  • Akt protein kinase B
  • mTOR mammalian target of rapamycin
  • PDK1 phosphoinositide-dependent kinase 1
  • the mTOR-related disease can be inflammation, atherosclerosis, psoriasis, restenosis, benign prostatic hypertrophy, bone disorders, pancreatitis, angiogenesis, diabetic retinopathy, atherosclerosis, arthritis, immunological disorders, kidney disease, or cancer.
  • a PI3K-associated disease can also include any disease, disorder or condition that can be prevented, ameliorated, or cured by modulating PI3K activity.
  • the disease is characterized by the abnormal activity of PI3K.
  • the disease is characterized by mutant PI3K.
  • the mutation can be present in the kinase domain of the PI3K.
  • PI3K-associated diseases include immune-based diseases involving the system including, for example, rheumatoid arthritis, allergy, asthma, glomerulonephritis, lupus, or inflammation related to any of the above.
  • PI3K-associated diseases include cancers such as breast, prostate, colon, endometrial, brain, bladder, skin, uterus, ovary, lung, pancreatic, renal, gastric, or hematological cancer.
  • the hematological cancer is acute myeloblasts leukemia (AML) or chronic myeloid leukemia (CML), or B cell lymphoma.
  • AML acute myeloblasts leukemia
  • CML chronic myeloid leukemia
  • PI3K-associated diseases include lung diseases such as acute lung injury (ALI) and adult respiratory distress syndrome (ARDS).
  • ALI acute lung injury
  • ARDS adult respiratory distress syndrome
  • PI3K-associated diseases include osteoarthritis, restenosis, atherosclerosis, bone disorders, arthritis, diabetic retinopathy, psoriasis, benign prostatic hypertrophy, inflammation, angiogenesis, pancreatitis, kidney disease, inflammatory bowel disease, myasthenia gravis, multiple sclerosis, or Sjogren's syndrome, and the like.
  • contacting refers to the bringing together of indicated moieties in an in vitro system or an in vivo system.
  • "contacting" a PI3K with a compound of the invention includes the administration of a compound of the present invention to an individual or patient, such as a human, having a PI3K, as well as, for example, introducing a compound of the invention into a sample containing a cellular or purified preparation containing the PI3K.
  • individual or patient used interchangeably, refers to any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates, and most preferably humans.
  • the phrase "therapeutically effective amount” refers to the amount of active compound or pharmaceutical agent that elicits the biological or medicinal response that is being sought in a tissue, system, animal, individual or human by a researcher, veterinarian, medical doctor or other clinician.
  • the dosage of the compound, or a pharmaceutically acceptable salt thereof, administered to a patient or individual is about 1 mg to about 2 g, about 1 mg to about 1000 mg, about 1 mg to about 500 mg, about 1 mg to about 100 mg, about 1 mg to 50 mg, or about 50 mg to about 500 mg.
  • the term "treating" or “treatment” refers to one or more of (1) preventing the disease; for example, preventing a disease, condition or disorder in an individual who may be predisposed to the disease, condition or disorder but does not yet experience or display the pathology or symptomatology of the disease; (2) inhibiting the disease; for example, inhibiting a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., arresting further development of the pathology and/or symptomatology); and (3) ameliorating the disease; for example, ameliorating a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., reversing the pathology and/or symptomatology) such as decreasing the severity of disease.
  • preventing the disease for example, preventing a disease, condition or disorder in an individual who may be predisposed to the disease, condition or disorder but does not yet experience or display the pathology or symptomatology of
  • One or more additional pharmaceutical agents such as, for example,
  • chemotherapeutics anti-inflammatory agents, steroids, immunosuppressants, as well as Bcr- Abl, Flt-3, EGFR, HER2, JAK, c-MET, VEGFR, PDGFR, cKit, IGF-1R, RAF, FAK, and mTOR kinase inhibitors such as, for example, those described in WO 2006/056399, or other agents such as, therapeutic antibodies can be used in combination with the compounds of the present invention for treatment of PBK-associated diseases, disorders or conditions.
  • the one or more additional pharmaceutical agents can be administered to a patient simultaneously or sequentially.
  • Example antibodies for use in combination therapy include but are not limited to Trastuzumab (e.g. anti-HER2), Ranibizumab (e.g. anti-VEGF-A), Bevacizumab (trade name Avastin, e.g. anti-VEGF, Panitumumab (e.g. anti-EGF ), Cetuximab (e.g. anti-EGFR), Rituxan (anti-CD20) and antibodies directed to c-MET.
  • Trastuzumab e.g. anti-HER2
  • Ranibizumab e.g. anti-VEGF-A
  • Bevacizumab trade name Avastin, e.g. anti-VEGF, Panitumumab (e.g. anti-EGF )
  • Cetuximab e.g. anti-EGFR
  • Rituxan anti-CD20
  • a cytostatic agent cisplatin, doxorubicin, taxotere, taxol, etoposide, irinotecan, camptostar, topotecan, paclitaxel, docetaxel, epothilones, tamoxifen, 5-fluorouracil, methoxtrexate, temozolomide, cyclophosphamide, SCH 66336, Rl 15777, L778,123, BMS 214662, Iressa, Tarceva, antibodies to EGFR, GleevecTM, intron, ara-C, adriamycin, Cytoxan, gemcitabine, Uracil mustard, Chlormethine, Ifosfamide, Melphalan, Chlorambucil, Pipobroman,
  • Triethylenemelamine Triethylenethiophosphoramine, Busulfan, Carmustine, Lomustine, Streptozocin, dacarbazine, Floxuridine, Cytarabine, 6-Mercaptopurine, 6-Thioguanine, Fludarabine phosphate, oxaliplatin, leucovirin, ELOXATINTM, Pentostatine, Vinblastine, Vincristine, Vindesine, Bleomycin, Dactinomycin, Daunorubicin, Doxorubicin, Epirubicin, Idarubicin, Mithramycin, Deoxycoformycin, Mitomycin-C, L-Asparaginase, Teniposide 17.alpha.-Ethinylestradiol, Diethylstilbestrol, Testosterone, Prednisone, Fluoxymesterone, Dromostanolone propionate, Testolactone, Megestrolacetate, Methylprednisolone,
  • Example chemotherapeutics include proteosome inhibitors (e.g., bortezomib), thalidomide, revlimid, and DNA-damaging agents such as melphalan, doxorubicin, cyclophosphamide, vincristine, etoposide, carmustine, and the like.
  • proteosome inhibitors e.g., bortezomib
  • thalidomide thalidomide
  • revlimid thalidomide
  • DNA-damaging agents such as melphalan, doxorubicin, cyclophosphamide, vincristine, etoposide, carmustine, and the like.
  • Example steroids include coriticosteroids such as dexamethasone or prednisone.
  • Example Bcr-Abl inhibitors include the compounds, and pharmaceutically acceptable salts thereof, of the genera and species disclosed in U.S. Pat. No. 5,521, 184, WO 04/005281, and U.S. Ser. No. 60/578,491.
  • Example suitable Flt-3 inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 03/037347, WO 03/099771, and WO 04/046120.
  • Example suitable RAF inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 00/09495 and WO 05/028444.
  • Example suitable FAK inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 04/080980, WO 04/056786, WO 03/024967, WO 01/064655, WO 00/053595, and WO 01/014402.
  • Example suitable mTOR inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 201 1/025889.
  • the compounds of the invention can be used in combination with one or more other kinase inhibitors including imatinib, particularly for treating patients resistant to imatinib or other kinase inhibitors.
  • the compounds of the invention can be used in combination with a chemotherapeutic in the treatment of cancer, such as multiple myeloma, and may improve the treatment response as compared to the response to the chemotherapeutic agent alone, without exacerbation of its toxic effects.
  • additional pharmaceutical agents used in the treatment of multiple myeloma can include, without limitation, melphalan, melphalan plus prednisone [MP], doxorubicin, dexamethasone, and Velcade (bortezomib).
  • Further additional agents used in the treatment of multiple myeloma include Bcr-Abl, Flt-3, RAF and FAK kinase inhibitors.
  • Additive or synergistic effects are desirable outcomes of combining a PI3K inhibitor of the present invention with an additional agent. Furthermore, resistance of multiple myeloma cells to agents such as dexamethasone may be reversible upon treatment with the PI3K inhibitor of the present invention.
  • the agents can be combined with the present compound in a single or continuous dosage form, or the agents can be administered simultaneously or sequentially as separate dosage forms.
  • a corticosteroid such as dexamethasone is administered to a patient in combination with the compounds of the invention where the dexamethasone is administered intermittently as opposed to continuously.
  • combinations of the compounds of the invention with other therapeutic agents can be administered to a patient prior to, during, and/or after a bone marrow transplant or stem cell transplant.
  • the compounds of the invention can be administered in the form of pharmaceutical compositions.
  • These compositions can be prepared in a manner well known in the pharmaceutical art, and can be administered by a variety of routes, depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration may be topical (including transdermal, epidermal, ophthalmic and to mucous membranes including intranasal, vaginal and rectal delivery), pulmonary (e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal or intranasal), oral or parenteral.
  • topical including transdermal, epidermal, ophthalmic and to mucous membranes including intranasal, vaginal and rectal delivery
  • pulmonary e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal or intranasal
  • oral or parenteral e.g., by inhal
  • Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal intramuscular or injection or infusion; or intracranial, e.g., intrathecal or intraventricular, administration.
  • Parenteral administration can be in the form of a single bolus dose, or may be, for example, by a continuous perfusion pump.
  • Pharmaceutical compositions and formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders. Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable.
  • compositions which contain, as the active ingredient, the compound of the invention or a pharmaceutically acceptable salt thereof, in combination with one or more pharmaceutically acceptable carriers (excipients).
  • the composition is suitable for topical administration.
  • the active ingredient is typically mixed with an excipient, diluted by an excipient or enclosed within such a carrier in the form of, for example, a capsule, sachet, paper, or other container.
  • the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient.
  • compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders.
  • the active compound can be milled to provide the appropriate particle size prior to combining with the other ingredients. If the active compound is substantially insoluble, it can be milled to a particle size of less than 200 mesh. If the active compound is substantially water soluble, the particle size can be adjusted by milling to provide a substantially uniform distribution in the formulation, e.g. about 40 mesh.
  • the compounds of the invention may be milled using known milling procedures such as wet milling to obtain a particle size appropriate for tablet formation and for other formulation types. Finely divided (nanoparticulate) preparations of the compounds of the invention can be prepared by processes known in the art, e.g., see International App. No. WO 2002/000196.
  • excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, and methyl cellulose.
  • the formulations can additionally include: lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propylhydroxy-benzoates; sweetening agents; and flavoring agents.
  • the compositions of the invention can be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the patient by employing procedures known in the art.
  • compositions can be formulated in a unit dosage form, each dosage containing from about 5 to about 1000 mg (1 g), more usually about 100 to about 500 mg, of the active ingredient.
  • unit dosage forms refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
  • compositions of the invention contain from about 5 to about 50 mg of the active ingredient.
  • the compositions of the invention contain from about 5 to about 50 mg of the active ingredient.
  • One having ordinary skill in the art will appreciate that this embodies compositions containing about 5 to about 10, about 10 to about 15, about 15 to about 20, about 20 to about 25, about 25 to about 30, about 30 to about 35, about 35 to about 40, about 40 to about 45, or about 45 to about 50 mg of the active ingredient.
  • compositions of the invention contain from about 50 to about 500 mg of the active ingredient.
  • the compositions of the invention contain from about 500 to about 1000 mg of the active ingredient.
  • One having ordinary skill in the art will appreciate that this embodies compositions containing about 500 to about 550, about 550 to about 600, about 600 to about 650, about 650 to about 700, about 700 to about 750, about 750 to about 800, about 800 to about 850, about 850 to about 900, about 900 to about 950, or about 950 to about 1000 mg of the active ingredient.
  • the active compound can be effective over a wide dosage range and is generally administered in a pharmaceutically effective amount. It will be understood, however, that the amount of the compound actually administered will usually be determined by a physician, according to the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
  • the principal active ingredient is mixed with a pharmaceutical excipient to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention.
  • a solid preformulation composition containing a homogeneous mixture of a compound of the present invention.
  • the active ingredient is typically dispersed evenly throughout the composition so that the composition can be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules.
  • preformulation is then subdivided into unit dosage forms of the type described above containing from, for example, about 0.1 to about 1000 mg of the active ingredient of the present invention.
  • the tablets or pills of the present invention can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action.
  • the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former.
  • the two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release.
  • enteric layers or coatings such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol, and cellulose acetate.
  • compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders.
  • the liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described supra.
  • the compositions are administered by the oral or nasal respiratory route for local or systemic effect.
  • compositions can be nebulized by use of inert gases. Nebulized solutions may be breathed directly from the nebulizing device or the nebulizing device can be attached to a face mask, tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions can be administered orally or nasally from devices which deliver the formulation in an appropriate manner.
  • Topical formulations can contain one or more conventional carriers.
  • ointments can contain water and one or more hydrophobic carriers selected from, for example, liquid paraffin, polyoxyethylene alkyl ether, propylene glycol, white Vaseline, and the like.
  • Carrier compositions of creams can be based on water in combination with glycerol and one or more other components, e.g. glycerinemonostearate, PEG- glycerinemonostearate and cetylstearyl alcohol.
  • Gels can be formulated using isopropyl alcohol and water, suitably in combination with other components such as, for example, glycerol, hydroxyethyl cellulose, and the like.
  • topical formulations contain at least about 0.1, at least about 0.25, at least about 0.5, at least about 1, at least about 2, or at least about 5 wt % of the compound of the invention.
  • the topical formulations can be suitably packaged in tubes of, for example, 100 g which are optionally associated with instructions for the treatment of the select indication, e.g., psoriasis or other skin condition.
  • compositions can be administered to a patient already suffering from a disease in an amount sufficient to cure or at least partially arrest the symptoms of the disease and its complications. Effective doses will depend on the disease condition being treated as well as by the judgment of the attending clinician depending upon factors such as the severity of the disease, the age, weight and general condition of the patient, and the like.
  • compositions administered to a patient can be in the form of pharmaceutical compositions described above. These compositions can be sterilized by conventional sterilization techniques, or may be sterile filtered. Aqueous solutions can be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration.
  • the pH of the compound preparations typically will be between 3 and 1 1, more preferably from 5 to 9 and most preferably from 7 to 8. It will be understood that use of certain of the foregoing excipients, carriers, or stabilizers will result in the formation of pharmaceutical salts.
  • the therapeutic dosage of a compound of the present invention can vary according to, for example, the particular use for which the treatment is made, the manner of administration of the compound, the health and condition of the patient, and the judgment of the prescribing physician.
  • the compounds of the invention can be provided in an aqueous physiological buffer solution containing about 0.1 to about 10% w/v of the compound for parenteral administration.
  • Some typical dose ranges are from about 1 ⁇ g/kg to about 1 g/kg of body weight per day. In some embodiments, the dose range is from about 0.01 mg/kg to about 100 mg/kg of body weight per day.
  • the dosage is likely to depend on such variables as the type and extent of progression of the disease or disorder, the overall health status of the particular patient, the relative biological efficacy of the compound selected, formulation of the excipient, and its route of administration. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • compositions of the invention can further include one or more additional pharmaceutical agents such as a chemotherapeutic, steroid, anti-inflammatory compound, or immunosuppressant, examples of which are listed herein.
  • additional pharmaceutical agents such as a chemotherapeutic, steroid, anti-inflammatory compound, or immunosuppressant, examples of which are listed herein.
  • Another aspect of the present invention relates to labeled compounds of the invention (radio-labeled, fluorescent-labeled, etc.) that would be useful not only in imaging techniques but also in assays, both in vitro and in vivo, for localizing and quantitating PI3K in tissue samples, including human, and for identifying PI3K ligands by inhibition binding of a labeled compound.
  • the present invention includes PI3K assays that contain such labeled compounds.
  • the present invention further includes isotopically-labeled compounds of the invention.
  • An "isotopically" or “radio-labeled” compound is a compound of the invention where one or more atoms are replaced or substituted by an atom having an atomic mass or mass number different from the atomic mass or mass number typically found in nature (i.e., naturally occurring).
  • Suitable radionuclides that may be incorporated in compounds of the present invention include but are not limited to 3 H (also written as T for tritium), n C, 13 C, 14 C, 13 N, 15 N, , 5 0, 17 0, 18 0, 18 F, 35 S, 36 C1, 82 Br, 75 Br, 76 Br, 77 Br, 123 I, 12 I, 125 I and 131 I.
  • the radionuclide that is incorporated in the instant radio-labeled compounds will depend on the specific application of that radio-labeled compound. For example, for in vitro PI3K labeling
  • a “radio-labeled " or “labeled compound” is a compound that has incorporated at least one radionuclide.
  • the radionuclide is selected from the group consisting of 3 H, 14 C, 125 1 , 35 S and 82 Br.
  • one or more H atoms for any compound described herein is each replaced by a deuterium atom.
  • the present invention can further include synthetic methods for incorporating radioisotopes into compounds of the invention. Synthetic methods for incorporating radio-isotopes into organic compounds are well known in the art, and an ordinary skill in the art will readily recognize the methods applicable for the compounds of invention.
  • a newly synthesized or identified compound ⁇ i.e., test compound) which is labeled can be evaluated for its ability to bind a PI3K by monitoring its concentration variation when contacting with the PI3K, through tracking of the labeling.
  • a test compound (labeled) can be evaluated for its ability to reduce binding of another compound which is known to bind to a PI3K ⁇ i.e., standard compound).
  • test compound to compete with the standard compound for binding to the PI3K directly correlates to its binding affinity.
  • standard compound is labeled and test compounds are unlabeled.
  • kits useful for example, in the treatment or prevention of PI3K-associated diseases or disorders, such as cancer, which include one or more containers containing a pharmaceutical composition comprising a therapeutically effective amount of a compound of the invention.
  • kits can further include, if desired, one or more of various conventional pharmaceutical kit components, such as, for example, containers with one or more pharmaceutically acceptable carriers, additional containers, etc., as will be readily apparent to those skilled in the art.
  • Instructions, either as inserts or as labels, indicating quantities of the components to be administered, guidelines for administration, and/or guidelines for mixing the components, can also be included in the kit.
  • Example 1 The example compounds below containing one or more chiral centers were obtained in racemate form or as isomeric mixtures, unless otherwise specified. Salt stoichiometry which is indicated any of the products below is meant only to indicate a probable stoichiometry, and should not be construed to exclude the possible formation of salts in other stoichiometrics.
  • Example 1 Salt stoichiometry which is indicated any of the products below is meant only to indicate a probable stoichiometry, and should not be construed to exclude the possible formation of salts in other stoichiometrics.
  • Example 1 Example 1
  • Step 1 l-(3-Bromo-5-chloro-2-hydroxy-4-methylphenyl)ethanone To a stirred solution of l-(5-chloro-2-hydroxy-4-methylphenyl)ethanone (10.0 g, 54.2 mmol, from Aldrich) in acetic acid (100 mL) was added N-bromosuccinimide (11.6 g, 65.0 mmol) and the resulting mixture was stirred at rt for 18 h. The reaction mixture was concentrated in vacuo, then neutralized with sat. sodium bicarbonate, and the insoluble succinimide was removed by filtration. The filtrate was extracted with ethyl acetate (EtOAc).
  • EtOAc ethyl acetate
  • Step 4 l- ⁇ 5-chloro-2-methoxy-4-methyl-3-[5-(methylsulfonyl)pyridin-3- yl]phenyl ⁇ ethanamine
  • Step 5 4-amino-6-[(l- ⁇ 5-chloro-2-methoxy-4-methyl-3-[5-(methylsulfonyl)pyridin-3- yl]phenyl ⁇ ethyl)amino]pyrimidine-5-carbonitrile trifluoroacetic acid salt
  • Step 1 l-(5-Chloro-4-fluoro-2-hydroxy henyl)ethanone
  • Step 3 l-(3-Bromo-5-chloro-4-fluoro-2-methoxyphenyl)ethanone
  • Step 5 4-Amino-6- ⁇ [l-(3-bromo-5-chloro-4-fluoro-2- methoxyphenyl)ethyl]amino ⁇ pyrimidine-5-carbonitrile
  • Step A l-(4, 5-Dichloro-2-hydroxyphen l)ethanone
  • Step B l-(4,5-Dichloro-2-hydroxy-3-iodophenyl)ethanone
  • Step C l-(4,5-Dichloro-3-iodo-2-methox henyl)ethanone
  • Step F l-(4,5-Dichloro-3-iodo-2-methox henyl)ethanamine
  • Step G 4-Amino-6- ⁇ fl-(4,5-dichloro-3-iodo-2-methoxyphenyl)ethyl]amino ⁇ pyrimidine-5- carbonitrile
  • Step H 4-Amino-6-[(l- ⁇ 4,5-dichloro-2-methoxy-3-[5-(methylsulfonyl)pyridin-3- yl]phenyl ⁇ ethyl)amino]pyrimidine-5-carbonitrile
  • the desired compound was prepared according to the procedure of Example 6, Step 6, using 4-amino-6- ⁇ [l -(4,5-dichloro-3-iodo-2-methoxyphenyl)ethyl]amino ⁇ -pyrimidine-5- carbonitrile as the starting material in 6% yield.
  • LCMS for C20H19CI2N6O3S (M+H) + : m/z 493.1, 495.1; Found: 493.0, 495.0.
  • PI3-Kinase luminescent assay kit including lipid kinase substrate, D-myo- phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P2)D (+)-sn-l,2-di-0-octanoylglyceryl, 3- O-phospho linked (PIP2), biotinylated I(1,3,4,5)P4, PI(3,4,5)P3 Detector Protein, was purchased from Echelon Biosciences (Salt Lake City, UT). AlphaScreenTM GST Detection Kit including donor and acceptor beads was purchased from PerkinElmer Life Sciences
  • the kinase reaction was conducted in 384-well REMP plate from Thermo Fisher Scientific in a final volume of 40 Inhibitors were first diluted serially in DMSO and added to the plate wells before the addition of other reaction components. The final concentration of DMSO in the assay was 2%.
  • the PI3K assays were carried out at room temperature in 50 mM HEPES, pH 7.4, 5mM MgCl 2 , 50 mM NaCl, 5mM DTT and CHAPS 0.04%. Reactions were initiated by the addition of ATP, the final reaction mixture consisted of 20 ⁇ PIP2, 20 ⁇ ATP, 1.2nM ⁇ 3 ⁇ were incubated for 20 min. 10 ⁇ of reaction mixture was then transferred to 5 ⁇ .
  • Lipid kinase substrate phosphoinositol-4,5-bisphosphate (PIP2)
  • PIP2 phosphoinositol-4,5-bisphosphate
  • PI3K isoforms ⁇ , ⁇ , ⁇ and ⁇ were purchased from Millipore (Bedford, MA).
  • ATP, MgCl 2 , DTT, EDTA, MOPS and CHAPS were purchased from Sigma-Aldrich (St. Louis, MO).
  • the kinase reaction was conducted in clear-bottom 96-well plate from Thermo Fisher Scientific in a final volume of 24 Inhibitors were first diluted serially in DMSO and added to the plate wells before the addition of other reaction components. The final concentration of DMSO in the assay was 0.5%.
  • the PI3K assays were carried out at room temperature in 20 mM MOPS, pH 6.7, 10 mM MgCl 2 , 5 mM DTT and CHAPS 0.03%.
  • the reaction mixture was prepared containing 50 ⁇ PIP2, kinase and varying concentration of inhibitors. Reactions were initiated by the addition of ATP containing 2.2 ⁇ [ ⁇ - 33 ⁇ ] ⁇ to a final concentration of 1000 uM.
  • the final concentration of PI3K isoforms ⁇ , ⁇ , ⁇ and ⁇ in the assay were 1.3, 9.4, 2.9 and 10.8 nM respectively.
  • Reactions were incubated for 180 min and terminated by the addition of 100 ⁇ of 1 M potassium phosphate pH 8.0, 30 mM EDTA quench buffer. A 100 ⁇ , aliquot of the reaction solution was then transferred to 96-well Millipore MultiScreen IP 0.45 ⁇ PVDF filter plate (The filter plate was prewetted with 200 ⁇ , 100% ethanol, distilled water, and 1 M potassium phosphate pH 8.0, respectively).
  • the filter plate was aspirated on a Millipore Manifold under vacuum and washed with 18 x 200 ⁇ ⁇ wash buffer containing 1 M potassium phosphate pH 8.0 and 1 mM ATP. After drying by aspiration and blotting, the plate was air dried in an incubator at 37 °C overnight. Packard TopCount adapter (Millipore) was then attached to the plate followed with addition of 120 ⁇ ⁇ Microscint 20 scintillation cocktail (Perkin Elmer) in each well. After the plate sealing, the radioactivity of the product was determined by scintillation counting on Topcount (Perkin- Elmer). IC 50 determination was performed by fitting the curve of percent control activity versus the log of the inhibitor concentration using the GraphPad Prism 3.0 software.
  • [ ⁇ - 33 ⁇ ] ⁇ (lOmCi/mL) was purchased from Perkin-Elmer (Waltham, MA).
  • ⁇ 3 ⁇ (pi 10 ⁇ / ⁇ 85 ⁇ ) was purchased from Millipore (Bedford, MA).
  • ATP MgCl 2 , DTT, EDTA, MOPS and CHAPS were purchased from Sigma-Aldrich (St. Louis, MO).
  • the kinase reaction was conducted in polystyrene 384-well matrix white plate from Thermo Fisher Scientific in a final volume of 25 ⁇ . Inhibitors were first diluted serially in DMSO and added to the plate wells before the addition of other reaction components. The final concentration of DMSO in the assay was 0.5%.
  • the PI3K assays were carried out at room temperature in 20 mM MOPS, pH 6.7, 10 mM MgCl 2 , 5 mM DTT and CHAPS 0.03%. Reactions were initiated by the addition of ATP, the final reaction mixture consisted of 20 ⁇ PIP2, 20 ⁇ ATP, 0.2 ⁇ [ ⁇ - 33 ⁇ ] ATP, 4 nM PI3K5.
  • human PBMC are isolated from the peripheral blood of normal, drug free donors by standard density gradient centrifugation on Ficoll-Hypague (GE).
  • the purified B cells (2x l0 5 /well/200 ⁇ ,) are cultured in 96-well ultra-low binding plates (Corning, Corning, NY) in RPMI1640, 10% FBS and goat F(ab')2 anti-human IgM (10 ⁇ g/ml) (Invitrogen, Carlsbad, CA) in the presence of different amount of test compounds for three days.
  • [ 3 H]-thymidine (1 ⁇ ) (PerkinElmer, Boston, MA) in PBS is then added to the B cell cultures for an additional 12 hours before the incorporated radioactivity is separated by filtration with water through GF B filters (Packard Bioscience, Meriden, CT) and measured by liquid scintillation counting with a TopCount (Packard Bioscience).
  • Pfeiffer cell line (diffuse large B cell lymphoma) was purchased from ATCC
  • the Pfeiffer cells were plated with the culture medium (2x10 3 cells / well/ per 200 ⁇ ) into 96-well ultra-low binding plates (Corning, Corning, NY), in the presence or absence of a concentration range of test compounds.
  • Ramos cells B lymphocyte from Burkitts lymphoma
  • ATCC Manassas, VA
  • RPMI1640 and 10% FBS.
  • the cells (3x l0 7 cells /tube/3 mL in RPMI) are incubated with different amounts of test compounds for 2 hrs at 37 °C and then stimulated with goat F(ab')2 anti-human IgM (5 ⁇ g/mL) (Invitrogen) for 17 minutes in a 37 °C water bath.
  • the stimulated cells are spun down at 4 °C with centrifugation and whole cell extracts are prepared using 300 ⁇ L lysis buffer (Cell Signaling Technology, Danvers, MA).
  • the resulting lysates are sonicated and supernatants are collected.

Abstract

The present invention provides pyrimidine-4,6-diamine derivatives of Formula (I): wherein the variables are defined herein, that modulate the activity of phosphoinositide 3-kinases (PI3Ks) and are useful in the treatment of diseases related to the activity of PI3Ks including, for example, inflammatory disorders, immune-based disorders, cancer, and other diseases.

Description

PYRIMIDINE-4,6-DIAMINE DERIVATIVES AS PI3K INHIBITORS
This application claims the benefit of priority of U.S. Provisional Application No. 61/467,787, filed March 25, 201 1, which is incorporated herein by reference in its entirety.
FIELD OF THE INVENTION
The present invention provides pyrimidine-4,6-diamine derivatives that modulate the activity of phosphoinositide 3-kinases (PBKs) and are useful in the treatment of diseases related to the activity of PBKs including, for example, inflammatory disorders, immune- based disorders, cancer, and other diseases.
BACKGROUND OF THE INVENTION
The phosphoinositide 3-kinases (PBKs) belong to a large family of lipid signaling kinases that phosphorylate phosphoinositides at the D3 position of the inositol ring (Cantley, Science, 2002, 296(5573): 1655-7). PBKs are divided into three classes (class I, II, and III) according to their structure, regulation and substrate specificity. Class I PBKs, which include ΡΒΚα, ΡΒΚβ, ΡΒΚγ, and ΡΒΚδ, are a family of dual specificity lipid and protein kinases that catalyze the phosphorylation of phosphatidylinosito-4,5-bisphosphate (PIP2) giving rise to phosphatidylinosito-3,4,5-trisphosphate (PIP3). PIP3 functions as a second messenger that controls a number of cellular processes, including growth, survival, adhesion and migration. All four class I PI3K isoforms exist as heterodimers composed of a catalytic subunit (pi 10) and a tightly associated regulatory subunit that controls their expression, activation, and subcellular localization. ΡΒΚα, ΡΒΚβ, and ΡΒΚδ associate with a regulatory subunit known as p85 and are activated by growth factors and cytokines through a tyrosine kinase-dependent mechanism (Jimenez, et al., J Biol Chem., 2002, 277(44):41556- 62) whereas ΡΒΚγ associates with two regulatory subunits (pi 01 and p84) and its activation is driven by the activation of G-protein-coupled receptors (Brock, et al., J Cell Biol, 2003, 160(l):89-99). PBKcc and ΡΒΚβ are ubiquitously expressed. In contrast, ΡΒΚγ and ΡΒΚδ are predominantly expressed in leukocytes (Vanhaesebroeck, et al., Trends Biochem Sci., 2005, 30(4): 194-204).
The differential tissue distribution of the PI3K isoforms factors in their distinct biological functions. Genetic ablation of either PBKcc or ΡΒΚβ results in embryonic lethality, indicating that PI3Ka and ΡΙ3Κβ have essential and non-redundant functions, at least during development (Vanhaesebroeck, et al., 2005). In contrast, mice which lack ΡΙ3Κγ and PI3K8 are viable, fertile and have a normal life span although they show an altered immune system. ΡΙ3Κγ deficiency leads to impaired recruitment of macrophages and neutrophils to sites of inflammation as well as impaired T cell activation (Sasaki, et al., Science, 2000, 287(5455): 1040-6). ΡΒΚδ-mutant mice have specific defects in B cell signaling that lead to impaired B cell development and reduced antibody responses after antigen stimulation (Clayton, et al, J Exp Med. 2002, 196(6):753-63; Jou, et al, Mol Cell Biol. 2002, 22(24):8580-91 ; Okkenhaug, et al. Science, 2002, 297(5583): 1031-4).
The phenotypes of the ΡΙ3Κγ and ΡΒΚδ-mutant mice suggest that these enzymes may play a role in inflammation and other immune-based diseases and this is borne out in preclinical models. ΡΒΚγ-mutant mice are largely protected from disease in mouse models of rheumatoid arthritis (RA) and asthma (Camps, et al, Nat Med. 2005, l l(9):936-43;
Thomas, et al, Eur J Immunol. 2005, 35(4): 1283-91). In addition, treatment of wild-type mice with a selective inhibitor of ΡΒΚγ was shown to reduce glomerulonephritis and prolong survival in the MRL-lpr model of systemic lupus nephritis (SLE) and to suppress joint inflammation and damage in models of RA (Barber, et al, Nat Med. 2005, 1 1(9):933-5; Camps, et al, 2005). Similarly, both ΡΒΚδ-mutant mice and wild-type mice treated with a selective inhibitor of ΡΒΚδ have been shown to have attenuated allergic airway
inflammation and hyper-responsiveness in a mouse model of asthma (Ali, et al. Nature.
2004, 431(701 1): 1007-11 ; Lee, et al, FASEB J. 2006, 20(3):455-65) and to have attenuated disease in a model of RA (Randis, et al, Eur. J. Immunol, 2008, 38(5): 1215-24).
In addition to their potential role in inflammatory diseases, all four class I PBK isoforms may play a role in cancer. The gene encoding pi 10a is mutated frequently in common cancers, including breast, prostate, colon and endometrial (Samuels, et al. Science, 2004, 304(5670):554; Samuels, et al, Curr Opin Oncol. 2006, 18(l):77-82). Eighty percent of these mutations are represented by one of three amino acid substitutions in the helical or kinase domains of the enzyme and lead to a significant upregulation of kinase activity resulting in oncogenic transformation in cell culture and in animal models (Kang, et al, Proc Natl Acad Sci U S A. 2005, 102(3):802-7; Bader, et al, Proc Natl Acad Sci U S A. 2006,
103 (5): 1475 -9). No such mutations have been identified in the other PBK isoforms although there is evidence that they can contribute to the development and progression of
malignancies. Consistent overexpression of ΡΒΚδ is observed in acute myeloblastic leukemia (Sujobert, et al., Blood, 2005, 106(3): 1063-6) and inhibitors of PI3K5 can prevent the growth of leukemic cells (Billottet, et al, Oncogene. 2006, 25(50):6648-59). Elevated expression of ΡΙ3Κγ is seen in chronic myeloid leukemia (Hickey, et al., J Biol Chem. 2006, 281(5):2441-50). Alterations in expression of ΡΒΚβ, ΡΙ3Κγ and PI3K8 have also been observed in cancers of the brain, colon and bladder (Benistant, et al., Oncogene, 2000, 19(44):5083-90; Mizoguchi, et al. Brain Pathol. 2004, 14(4):372-7; Knobbe, et al,
Neuropathol Appl Neurobiol. 2005, 31(5):486-90). Further, these isoforms have all been shown to be oncogenic in cell culture (Kang, et al, 2006).
Thus, new or improved agents which inhibit kinases such as PI3K are continually needed for developing new and more effective pharmaceuticals that are aimed at
augmentation or suppression of the immune and inflammatory pathways (such as
immunosuppressive agents for organ transplants), as well as agents for the prevention and treatment of autoimmune diseases (e.g., multiple sclerosis, rheumatoid arthritis, asthma, type I diabetes, inflammatory bowel disease, Crohn's disease, autoimmune thyroid disorders, Alzheimer's disease, nephritis), diseases involving a hyperactive inflammatory response (e.g. , eczema), allergies, lung diseases, cancer (e.g., prostate, breast, leukemia, multiple myeloma), and some immune reactions (e.g., skin rash or contact dermatitis or diarrhea) caused by other therapeutics. The compounds, compositions, and methods described herein are directed toward these needs and others.
SUMMARY
The present invention provides, inter alia, a compound of Formula I:
Figure imgf000004_0001
I
or a pharmaceutically acceptable salt thereof, wherein the variables are defined infra.
The present invention further provides compositions comprising a compound of the invention, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier. The present invention also provides methods of modulating an activity of a PI3K kinase, comprising contacting the kinase with a compound of the invention, or a
pharmaceutically acceptable salt thereof.
The present invention further provides methods of treating a disease in a patient, wherein said disease is associated with abnormal expression or activity of a PI3K kinase, comprising administering to said patient a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof.
The present invention further provides methods of treating an immune-based disease in a patient, comprising administering to said patient a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof.
The present invention also provides methods of treating a cancer in a patient, comprising administering to said patient a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof.
The present invention further provides methods of treating a lung disease in a patient, comprising administering to said patient a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof.
The present invention also provides a compound of invention, or a pharmaceutically acceptable salt thereof, for use in any of the methods described herein.
The present invention further provides use of a compound, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for use in any of the methods described herein.
DETAILED DESCRIPTION
The present invention provides, inter alia, a compound of Formula I:
Figure imgf000005_0001
I
harmaceutically acceptable salt thereof, wherein: R1 is Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, or Cue haloalkyl, wherein said Cue alkyl, C2-6 alkenyl, and C2_6 alkynyl are each optionally substituted by 1, 2, 3, or 4 groups independently selected from halo, OH, CN, NRl aRl b, Ci_6 alkoxy, and Ci-6 haloalkoxy;
R2 is H, halo, CN, C 6 alkyl, C1-6 haloalkyl, -Ll-(CU6 alkyl), or - -(C 6 haloalkyl), wherein said C1-6 alkyl in said C e alkyl and -Ll-(Cue alkyl) is optionally substituted by 1, 2, 3, or 4 independently selected R2a groups;
R3 is Cy, -(CR2bR2c)a-L2-(CR2bR2c)b-Cy, halo, CN, N02, C 6 alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci_6 haloalkyl, ORa, SRa, C(0)Rb, C(0)NRcRd, C(0)ORa, OC(0)Rb, OC(0)NRcRd, NRcRd, NRcC(0)Rb, NRcC(0)ORb, NRcC(0)NRcRd, C(=NRe)Rb, C(=NRe)NRcRd,
NRcC(=NRe)NRcRd, NRcS(0)Rb, NRcS(0)2Rb, NRcS(0)2NRcRd, S(0)Rb, S(0)NRcRd, S(0)2Rb, or S(0)2NRcRd, wherein said Ci-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted by 1, 2, 3, or 4 independently selected R3a groups;
R4 is H, halo, OH, CN, CM alkyl, CM haloalkyl, C alkoxy, or C1-4 haloalkoxy;
R5 is halo, OH, CN, CM alkyl, CM alkoxy, or CM haloalkoxy;
R6 is H, halo, OH, CN, C alkyl, C1-4 haloalkyl, C alkoxy, or CM haloalkoxy;
R7 is F, CN, Ci.3 haloalkyl, CONH2, CM alkyl-NHC(=0)- or (CM alkyl)2NC(=0)-;
R8 is H, CM alkyl, CM haloalkyl, or C(0)R4a;
L1 is O, NRB, S, S(O), S(0)2, C(O), C(0)NRB, S(0)NRB, or S(0)2NRB;
L2 is (CRcRD)n, O, NRB, S, S(O), S(0)2, C(O), C(0)0, C(0)NRB, S(0)NRB, S(0)2NRB, OC(0)NRB, NRBC(0)NRB, C(=NRe), C(=NRe)NRB, or NRBC(=NRe)NRB;
RA, RB, Rc, and RD are each independently selected from H, Cue alkyl, and Ci.6 haloalkyl;
Cy is selected from C3-7 cycloalkyl, 4-11 membered heterocycloalkyl, 6-10 membered aryl and 5-10 membered heteroaryl, each of which is substituted with 0, 1, 2, 3, or 4 independently selected R3 groups;
each Rl a and Rlb is independently selected from H and CM alkyl;
or any Rla and R2b together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group, which is optionally substituted with 1, 2, 3, or 4 substituents independently selected from C1-6 alkyl;
each R2 is independently selected from OH, N02, CN, halo, C1-6 alkyl, C2-6 alkenyl, C2-
6 alkynyl, C3-7 cycloalkyl, Cj_6 haloalkyl, Ci_6 alkoxy, Ci-6 haloalkoxy, amino, Ci_6 alkylamino, di(Ci-6 alkyl)amino, thio, Ci-6 alkylthio, C 1-6 alky lsulfinyl, Ci.6 alkylsulfonyl, carbamyl, C1-6 alkylcarbamyl, di(Ci-6 alkyl)carbamyl, carboxy, C1-6 alkylcarbonyl, Ci-6 alkoxycarbonyl, Cue alkylcarbonylamino, Ci^ alkylsulfonylamino, aminosulfonyl, Ci_6 alkylaminosulfonyl, di(Ci_6 alkyl)aminosulfonyl, aminosulfonylamino, Ci^ alkylaminosulfonylamino, di(C] _6
alkyl)aminosulfonylamino, aminocarbonylamino, Ci-6 alkylaminocarbonylamino, and di(Q_6 alkyl)aminocarbonylamino;
each R2b and R2c is independently selected from H, C] _6 alkyl, and Ci-6 haloalkyl;
each R3 is independently selected from halo, CN, N02, Ci_ alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci_6 haloalkyl, C3-7 cycloalkyl, 4-11 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, (C3.7 cycloalkyl)-Ci.4 alkyl, (4-11 membered
heterocycloalkyl)-C i_4 alkyl, (6-10 membered aryl)-C1 -4 alkyl, (5-10 membered heteroaryl)- C alkyl, ORal, SRal, C(0)Rbl, C(0)NRclRdl, C(0)ORal, OC(0)Rbl, OC(0)NRclRdl, NRclRdl, NRclC(0)Rbl, NRclC(0)ORbl, NRc1C(0)NRclRdl, C(=NRe)Rbl, C(=NRe)NRclRdl, NRclC(=NRe)NRclRdl, NRCIS(0)Rbl, NRclS(0)2Rbl, NRclS(0)2NRclRdl, S(0)Rbl,
S(O)NR0lRdl, S(0)2Rbl, and S(0)2NRclRdl; wherein said C 6 alkyl, C2.6 alkenyl, C2-6 alkynyl, C3.7 cycloalkyl, 4-11 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, (C3.7 cycloalkyl)-Ci_4 alkyl, (4-11 membered heterocycloalkyl)-C 1-4 alkyl, (6-10 membered ary^-Ci^ alkyl, and (5-10 membered heteroaryl)-C]-4 alkyl are each optionally substituted by 1, 2, 3, or 4 groups independently selected from ORaI, SRal, C(0)Rbl, C(0)NRclRdl, C(0)ORal, OC(0)Rbl, OC(0)NRclRdl, NRclRdl, NRclC(0)RM, NRclC(0)ORbl, NRclC(0)NRclRdl, C(=NRe)Rbl, C(=NRe)NRclRdl, NRclC(=NRe)NRclRdl, NRcl S(0)Rbl, NRclS(0)2Rbl, NRclS(0)2NRclRdl, S(0)Rbl, S(0)NRclRdl, S(0)2Rbl, and S(0)2NRclRdl ;
R a is Ci-6 alkyl, C3-7 cycloalkyl, 4-1 1 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, (C3.7 cycloalkyl)-Ci.4 alkyl, (4-11 membered heterocycloalkyl)-C i_ 4 alkyl, (6-10 membered aryl)-C 1 -4 alkyl, and (5-10 membered heteroaryl)-Ci_4 alkyl;
each Ra, Rb, Rc, and Rd is independently selected from H, Ci-6 alkyl, C1-6 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl; wherein said C1-6 alkyl, C2-6 alkenyl, and C2_6 alkynyl are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from OH, N02, CN, halo, C alkyl, C2-6 alkenyl, C2_e alkynyl, C1-6 haloalkyl, cyano-Ci_6 alkyl, HO-C1.6 alkyl, Ci^ alkoxy-Ci-g alkyl, C3-7 cycloalkyl, C1-6 haloalkyl, C^ alkoxy, Ci-6 haloalkoxy, amino, Ci-6 alkylamino, di(Ci_6 alkyl)amino, thio, Ci_6 alkylthio, Ci-6 alkylsulfinyl, Ci-6 alkylsulfonyl, carbamyl, Ci-6 alkylcarbamyl, di(C]_6 alkyl)carbamyl, carboxy, Ci_6 alkylcarbonyl, Ci_6 alkoxycarbonyl, C1 -6 alkylcarbonylamino, Ci-6 alkylsulfonylamino, aminosulfonyl, C^e alkylaminosulfonyl, di(Ci.6 alkyl)aminosulfonyl, aminosulfonylamino, Ci_6 alkylaminosulfonylamino, di(Ci.6 alkyl)aminosulfonylamino, aminocarbonylamino, Ci_6 alkylaminocarbonylamino, and di(C] _6 alkyl)aminocarbonylamino; or RC and RD together with the N atom to which they are attached form a 4-, 5-, 6-, or 7- membered heterocycloalkyl group, which is optionally substituted with 1, 2, 3, or 4 substituents independently selected from OH, N02, CN, halo, alkyl, C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, cyano-Ci_6 alkyl, HO-C]_6 alkyl, Ci.4 alkoxy-Ci_6 alkyl, C3-7
cycloalkyl, C e haloalkyl, Ci_6 alkoxy, C1-6 haloalkoxy, amino, Ci-6 alkylamino, di(Q.6 alkyl)amino, thio, C1-6 alkylthio, Ci-6 alley lsulfinyl, C1 -6 alkylsulfonyl, carbamyl, C1-6 alkylcarbamyl, di(C].6 alkyl)carbamyl, carboxy, C]-6 alkylcarbonyl, C . alkoxycarbonyl, Ci_6 alkylcarbonylamino, Ci_6 alkylsulfonylamino, aminosulfonyl, Ci^ alkylaminosulfonyl, di(Ci_6 alkyl)aminosulfonyl, aminosulfonylamino, Ci-6 alkylaminosulfonylamino, di(Ci_6
alkyl)aminosulfonylamino, aminocarbonylamino, Ci-6 alkylaminocarbonylamino, and di(Ci_6 alkyl)aminocarbonylamino;
each RE is independently selected from H, Q.4 alkyl, CN, OH, CM alkoxy, C1-4 alkylsulfonyl, carbamyl, C 1.4 alkylcarbamyl, di(Ci_4 alkyl)carbamyl, and Q.4 alkylcarbonyl; each RAL, RBL , RCL , and RD L is independently selected from H, Ci_6 alkyl, Ci_6 haloalkyl, C2-6 alkenyl, C2_e alkynyl, C3-7 cycloalkyl, 4-1 1 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, (C3-7 cycloalkyl)-Ci. alkyl, (4-1 1 membered
heterocycloalkyl)-Ci-4 alkyl, (6-10 membered aryl)-Ci_4 alkyl, and (5-10 membered heteroaryl)-Ci-4 alkyl, wherein said Q.6 alkyl, C2.6 alkenyl, C2-6 alkynyl, C3.7 cycloalkyl, 4-11 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, (03,7
cycloalkyl)-Ci_4 alkyl, (4-11 membered heterocycloalkyl)-Ci_4 alkyl, (6-10 membered aryl)- C1.4 alkyl, and (5-10 membered heteroaryl)-Ci_4 alkyl are each optionally substituted with 1, 2, 3, 4, or 5 substituents independently selected from OH, N02, CN, halo, Ci-6 alkyl, C2-6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, cyano-Ci_6 alkyl, HO-Ci-6 alkyl, C1-4 alkoxy-Ci.6 alkyl, C3-7 cycloalkyl, C1-6 haloalkyl, Ci-β alkoxy, Ci-6 haloalkoxy, amino, C1-6 alkylamino, di(C] _6 alkyl)amino, thio, Ci-6 alkylthio, Ci_6 alkylsulfmyl, C1-6 alkylsulfonyl, carbamyl, C1-6 alkylcarbamyl, di(Ci-6 alkyl)carbamyl, carboxy, Ci -e alkylcarbonyl, Ci-6 alkoxycarbonyl, Ci-6 alkylcarbonylamino, Ci-6 alkylsulfonylamino, aminosulfonyl, C]_6 alkylaminosulfonyl, di(Ci_e alkyl)aminosulfonyl, aminosulfonylamino, Ci_6 alkylaminosulfonylamino, di(Ci_6
alkyl)aminosulfonylamino, aminocarbonylamino, C i-6 alkylaminocarbonylamino, and di(Ci-6 alkylaminocarbonylamino;
or RCL and RDL together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group, which is optionally substituted with 1, 2, 3, or 4 substituents independently selected from OH, N02, CN, halo, C1-6 alkyl, C2_6 alkenyl, C2_6 alkynyl, C1-6 haloalkyl, cyano-Q.6 alkyl, HO-C1-6 alkyl, Ci-4 alkoxy-Ci-6 alkyl, C3-7 cycloalkyl, C1-6 haloalkyl, Ci^ alkoxy, Ci_6 haloalkoxy, amino, Ci-6 alkylamino, di(Ci_6 alkyl)amino, thio, Ci_6 alkylthio, Ci^ alkylsulfmyl, Ci-6 alkylsulfonyl, carbamyl, Ci-6 alkylcarbamyl, di(Ci-6 alkyl)carbamyl, carboxy, Ci.6 alkylcarbonyl, Ci-6 alkoxycarbonyl, Ci-6 alkylcarbonylamino, Ci_6 alkylsulfonylamino, aminosulfonyl, Ci-6 lkylaminosulfonyl, di(Ci_6 alkyl)aminosulfonyl, aminosulfonylamino, Ci.6 alkylaminosulfonylamino, di(Ci_6 alkyl)aminosulfonylamino, aminocarbonylamino, Ci-6 alkylaminocarbonylamino, and di(Ci-6 alkyl)aminocarbonylamino;
a is 0, 1, 2, 3, or 4;
b is 0, 1, 2, 3, or 4; and
n is 1, 2, 3, 4, 5, or 6.
In some embodiments, R1 is C1-6 alkyl.
In some embodiments, R1 is methyl.
In some embodiments, R1 is ethyl.
In some embodiments, R2 is -L1-^]^ alkyl).
In some embodiments, R2 is C1-6 alkoxy.
In some embodiments, R2 is methoxy.
In some embodiments, R3 is Cy or -(CR2bR2c)a-L2-(CR2bR2c)b-Cy.
In some embodiments, R3 is Cy.
In some embodiments, R3 is halo, CN, N02, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, Ci-6 haloalkyl, ORa, SRa, C(0)Rb, C(0)NRcRd, C(0)ORa, OC(0)Rb, OC(0)NRcRd, NRcRd, NRcC(0)Rb, NRcC(0)ORb, RcC(0)NRcRd, C(=NRe)Rb, C(=NRe)NRcRd,
NRcC(=NRe)NRcRd, NRcS(0)Rb, NRcS(0)2Rb, NRcS(0)2NRcRd, S(0)Rb, S(0)NRcRd, S(0)2Rb, or S(0)2NRcRd.
In some embodiments, R3 is CN, N02, C2_6 alkenyl, C2-6 alkynyl, Ci-6 haloalkyl,
OC(0)Rb, OC(0)NRcRd, NRcC(0)Rb, NRcC(0)ORb, NRcC(0)NRcRd, C(=NRe)Rb,
C(=NRe)NReRd, NR C(=NRe)NRcRd, NRcS(0)Rb, NRcS(0)2Rb, NRcS(0)2NRcRd, S(0)Rb, S(0)NRcRd or S(0)2Rb.
In some embodiments, R3 is 5-10 membered heteroaryl substituted with 1, 2, or 3 groups independently selected from halo, CN, N02, C2-6 alkenyl, C2-6 alkynyl, Ci-6 haloalkyl, C3-7 cycloalkyl, 4-11 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, (C3.7 cycloalkyl)-C 1.4 alkyl, (4-1 1 membered heterocycloalkyl)-C 1 -4 alkyl, (6-10 membered aryl)-Ci_4 alkyl, and (5-10 membered heteroaryl)-Ci_4 alkyl, ORal, SRal,
C(0)NRclRdI, OC(0)Rbl, OC(0)NRclRdl, NRclRdl, NRclC(0)Rbl, NRclC(0)ORbl, NRclC(0)NRclRdl, C(=NRe)Rbl, C(=NRe)NRclRdl, NRclC(=NRe)NRclRdl, NRclS(0)Rbl, NRclS(0)2Rbl, NRclS(0)2NRclRdl, S(0)Rbl, S(0)NRclRdl, S(0)2Rbl, and S(0)2NRclRdl; wherein said C2-6 alkenyl, C2_6 alkynyl, C3-7 cycloalkyl, 4-11 membered heterocycloalkyl, 6- 10 membered aryl, 5-10 membered heteroaryl, (C3-7 cycloalkyl)-Ci_4 alkyl, (4-11 membered heterocycloalkyI)-Ci-4 alkyl, (6-10 membered aryl)-Ci-4 alkyl, and (5-10 membered heteroaryl)-Ci-4 alkyl are each optionally substituted by 1, 2, 3, or 4 groups independently selected from ORal, SRal, C(0)Rbl, C(0)NRclRdl, C(0)ORal, OC(0)Rbl, OC(0)NRclRdl, NRclRdl, NRclC(0)Rbl, NRc,C(0)ORbl, NRclC(0)NRclRdl, C(=NRe)Rbl, C(=NRe)NRclRdl, NRclC(=NRe)NRclRdl, NRclS(0)Rbl, NRclS(0)2Rbl, NRclS(0)2NRclRdl, S(0)Rbl,
S(0)NRclRdl, S(0)2Rbl, and S(0)2NRclRdl.
In some embodiments, R3 is 6-10 membered aryl substituted with 1, 2, or 3 groups independently selected from N02, C2_6 alkenyl, C2-6 alkynyl, Ci-6 haloalkyl, C3-7 cycloalkyl, 4-11 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, (C3-7 cycloalkyl)-Ci-4 alkyl, (4-11 membered heterocycloalkyl)-C 1-4 alkyl, (6-10 membered aryl)- C alkyl, and (5-10 membered heteroaryl)-Ci.4 alkyl, SRal, C(0)Rbl, C(0)NRclRdl,
C(0)ORal, OC(0)Rbl, OC(0)NRclRdl, NRclC(0)Rbl, NRclC(0)ORbl, NRclC(0)NRclRdl, C(=NRe)Rbl, C(=NRe)NRclRdl, NRclC(=NRe)NRclRdl, NRclS(0)Rbl, NRclS(0)2Rbl, NRclS(0)2NRclRdl, S(0)Rbl, S(0)NRclRdl, S(0)2Rbl, and S(0)2NRdRdl ; wherein said C2-6 alkenyl, C2_6 alkynyl, C3-7 cycloalkyl, 4-11 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, (C3_7 cycloalkyl)-C 1.4 alkyl, (4-11 membered heterocycloalkyl)-C ]. 4 alkyl, (6-10 membered aryl)-Ci-4 alkyl, and (5-10 membered heteroaryl)-Ci-4 alkyl are each optionally substituted by 1, 2, 3, or 4 groups independently selected from ORal, SRal, C(0)Rbl, C(0)NRclRdl, C(0)ORal, OC(0)Rbl, OC(0)NRclRdl, NRclRdl, NRclC(0)Rbl, NRc,C(0)ORbl, NRclC(0)NRclRdl, C(=NRe)Rbl, C(=NRe)NRclRdl, NRclC(=NRe)NRclRdl, NRc!S(0)Rbl, NRclS(0)2Rbl, NRclS(0)2NRclRdl, S(0)Rbl, S(0)NRclRdl, S(0)2Rbl, and S(0)2NRclRdl.
In some embodiments, Cy is selected from 6-10 membered aryl and 5-10 membered heteroaryl, each of which is substituted with 0, 1, 2, 3, or 4 independently selected R3a groups.
In some embodiments, Cy is phenyl substituted with 0, 1, 2, 3, or 4 independently selected R3a groups.
In some embodiments, Cy is 5-6 membered heteroaryl substituted with 0, 1, 2, 3, or 4 independently selected R3a groups. In some embodiments, Cy is pyridyl or pyrimidinyl substituted with 0, 1, 2, 3, or 4 independently selected R3 groups.
In some embodiments, Cy is pyridyl substituted with 0, 1, 2, 3, or 4 independently selected R3a groups.
In some embodiments, Cy is pyrimidinyl substituted with 0, 1, 2, 3, or 4 independently selected R3a groups.
In some embodiments, Cy is substituted with 1, 2, or 3 independently selected R3a groups.
In some embodiments, R3a is independently selected from halo, CN, NO2, C2_6 alkenyl, C2-6 alkynyl, Ci_6 haloalkyl, C3-7 cycloalkyl, 4-1 1 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, (C3-7 cycloalkyl)-C] _4 alkyl, (4-1 1 membered heterocycloalkyl)-Ci_4 alkyl, (6-10 membered aryl)-C 1.4 alkyl, and (5-10 membered heteroaryl)-C, _4 alkyl, ORal, SRal, C(0)NRclRdlOC(0)Rbl, OC(0)NRclRdl, NRclC(0)Rbl, NRclC(0)ORbl, NRclC(0)NRclRdl, C(=NRe)Rbl, C(=NRe)NRclRdl, NRclC(=NRe)NRclRdl, NRcl S(0)Rbl, NRCIS(0)2Rbl, NRcl S(0)2NRclRdl, S(0)Rbl, S(0)NRclRdl, S(0)2Rbl, and S(0)2NRclRdl; wherein said C2-6 alkenyl, C2-6 alkynyl, C3.7 cycloalkyl, 4-1 1 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, (C3..7 cycloalkyl)-Ci_4 alkyl, (4-11 membered heterocycloalkyl)-C 1.4 alkyl, (6-10 membered aryl)-C 1-4 alkyl, and (5- 10 membered heteroaryl)-Ci.4 alkyl are each optionally substituted by 1, 2, 3, or 4 groups independently selected from ORal, SR l, C(0)Rbl, C(0) RclRdl, C(0)ORal, OC(0)Rbl, OC(0)NRclRdl, NRclRdl, NRclC(0)R l, NR0lC(O)ORbl, NRclC(0)NRclRdl, C(=NRe)Rbl, C(=NRe)NRelRdl, NRclC(=NRe)NRclRdl, NRclS(0)Rbl, NRcl S(0)2Rbl, NRcl S(0)2NRclRdl, S(0)Rbl, S(0)NRclRdl, S(0)2Rbl, and S(0)2NRclRdl.
In some embodiments, R3a is independently selected from N02, C2-6 alkenyl, C2-6 alkynyl, Ci-6 haloalkyl, C3-7 cycloalkyl, 4-11 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, (C3_7 cycloalkyl)-Ci.4 alkyl, (4-11 membered
heterocycloalkyl)-C 1 -4 alkyl, (6-10 membered aryl)-C 1 -4 alkyl, and (5-10 membered heteroaryl)-CM alkyl, SRal, C(0)Rbl, C(0)NRclRdl, OC(0)Rbl, OC(0)NRclRdl,
NRclC(0)Rbl, NRclC(0)ORbl, NRclC(0)NRclRdl, C(=NRe)Rbl, C(=NRe)NRclRdl,
NRclC(=NRe)NRclRdl, NRclS(0)Rbl, NRclS(0)2Rbl, NRclS(0)2NRclRdl, S(0)Rbl,
S(0)NRclRdl, S(0)2Rbl, and S(0)2NRclRdl ; wherein said C2-6 alkenyl, C2_6 alkynyl, C3.7 cycloalkyl, 4-11 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, (C3-7 cycloalkyl)-C 1.4 alkyl, (4-11 membered heterocycloalkyl)-Ci-4 alkyl, (6-10 membered aryl)-C1-4 alkyl, and (5-10 membered heteroaryl)-Ci_4 alkyl are each optionally substituted by 1, 2, 3, or 4 groups independently selected from ORal, SRal, C(0)Rbl, C(0)NRclRdl, C(0)ORal, OC(0)Rbl, OC(0)NRclRdl, NRclRdl, NRclC(0)Rbl,
NRclC(0)ORbl, NRclC(0)NRclRdl, C(=NRe)Rbl, C(=NRe)NRclRdl, NRclC(=NRe)NRclRdl, NRcl S(0)Rbl, NRclS(0)2Rbl, NRclS(0)2NRclRdl, S(0)Rbl, S(0)NRclRdl, S(0)2Rbl, and S(0)2NRclRdl.
In some embodiments, R3a is independently selected from halo, CN, C(0)NRclRdl, NRclRdl, and S(0)2Rbl.
In some embodiments, R3a is independently selected from halo, CN, C(0)NRclRdl, and S(0)2Rbl .
In some embodiments, R3a is S(0)2Rbl.
In some embodiments, R3a is S(0)2-(Ci_4 alkyl).
In some embodiments, R3a is S(0)2-CH3.
In some embodiments, R3a is NH2.
In some embodiments, R3a is other than CN, halo, OH, Ci-6 alkyl, Ci-6 alkoxy, -Nt¾, -NH(C1-6 alkyl), -N(C1-6 alkyl)2, -(C1-6 alkyl)-NH2, -(C1-6 alkyl)-NH(C1-6 alkyl), -(C1-6 alkyl)- N(C1-6 alkyl)2, or -C(0)0-(C1-6 alkyl);
In some embodiments, R3a is other than Ci_6 alkyl, -(Ci_6 alkyl)-NH2, -(Ci-6 alkyl)- NH(Ci.6 alkyl), -(Ci_6 alkyl)-N(Ci-6 alkyl)2, -C(0)-(C,.6 alkyl), -C(0)OH, or -C(0)0-(Ci-6 alkyl);
In some embodiments, R4 is halo, CN, C1.4 alkyl, or Q^ haloalkyl.
In some embodiments, R4 is halo.
In some embodiments, R4 is methyl.
In some embodiments, R4 is F.
In some embodiments, R4 is CI.
In some embodiments, R5 is CI, F, or methyl.
In some embodiments, R5 is CI.
In some embodiments, R6 is H.
In some embodiments, R7 is CN.
In some embodiments, R8 is H.
In some embodiments, RA is H.
In some embodiments, when Cy is 6-10 membered aryl, then said 6-10 membered aryl is substituted by at least one group selected from N02, C2_6 alkenyl, C2-6 alkynyl, Ci_6 haloalkyl, C3-7 cycloalkyl, 4-11 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, (C3.7 cycloalkyl)-Ci_4 alkyl, (4-11 membered heterocycloalkyl)-C]_4 alkyl, (6-10 membered aryl)-Ci-4 alkyl, (5-10 membered heteroaryl)-C 1-4 alkyl, SRa , C(0)Rbl, C(0)NRclRdl, OC(0)Rbl, OC(0)NRclRdl, NRclC(0)Rbl, NRclC(0)ORbl,
Figure imgf000013_0001
NRclS(0)2Rbl, NRc,S(0)2NRclRdl, S(0)Rbl, S(0)NRc,Rdl, S(0)2Rbl, and S(0)2NRc,Rd' ; wherein said C2-e alkenyl, C2_6 alkynyl, C3.7 cycloalkyl, 4-1 1 membered heterocycloalkyl, 6- 10 membered aryl, 5-10 membered heteroaryl, (C3-7 cycloalkyl)-C 1 ,4 alkyl, (4-1 1 membered heterocycloalkyl)-Ci-4 alkyl, (6-10 membered aryl)-C 1-4 alkyl, and (5-10 membered heteroaryl)-Ci_4 alkyl are each optionally substituted by 1 , 2, 3, or 4 groups independently selected from ORal, SRal, C(0)Rbl, C(0)NRclRdI, C(0)ORal, OC(0)Rbl, OC(0)NRclRdl, NRclRdl, NRclC(0)Rbl, NRclC(0)ORbl, NRclC(0)NRclRdl, C(=NRe)Rbl, C(=NRe)NRclRdl, NRclC(=NRe)NRclRdl, NRclS(0)Rbl, NRclS(0)2Rbl, NRc lS(0)2NRclRdl, S(0)Rbl,
S(0)NRclRdl, S(0)2Rbl, and S(0)2NRclRdl; and
when Cy is 5-10 membered heteroaryl, then said 5-10 membered heteroaryl is substituted by at least one group selected from halo, CN, N02, C2-6 alkenyl, C2.6 alkynyl, Ci-6 haloalkyl, C3.7 cycloalkyl, 4-11 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, (C3.7 cycloalkyl)-C 1.4 alkyl, (4-11 membered heterocycloalkyl)-C 1 ,4 alkyl, (6-10 membered aryl)-Ci_4 alkyl, and (5-10 membered heteroaryl)-Ci_4 alkyl, ORal, SRa1, C(0)NRclRdl, OC(0)Rbl, OC(0)NRclRdl, NRclRdl, NRclC(0)Rbl, NRclC(0)ORbl, NRc1C(0)NRclRdl, C(=NRe)Rbl, C(=NRe)NRclRdl, NRelC(=NRe)NRclRdl, NRclS(0)Rbl, NRclS(0)2Rbl, NRclS(0)2NRclRdl, S(0)Rbl, S(0)NRclRdl, S(0)2Rbl, and S(0)2NRclRdl; wherein said C2_6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, 4-11 membered heterocycloalkyl, 6- 10 membered aryl, 5-10 membered heteroaryl, (C3.7 cycloalkyl)-Ci_4 alkyl, (4-1 1 membered heterocycloalkyl)-C 1.4 alkyl, (6-10 membered aryl)-C 1.4 alkyl, and (5-10 membered heteroaryl)-Ci„4 alkyl are each optionally substituted by 1, 2, 3, or 4 groups independently selected from ORal, SRal, C(0)R l, C(0)NRclRdl, C(0)ORal, OC(0)Rbl, OC(0)NRclRdl, NRclRdl, NRc,C(0)Rbl, NRclC(0)ORbl, NRclC(0)NRclRdl, C(=NRe)Rbl, C(=NRe)NRclRdl, NRclC(=NRe)NRclRdl, NRclS(0)Rbl, NRclS(0)2Rbl, NRclS(0)2NRclRdl, S(0)Rbl,
S(0)NRclRdl, S(0)2Rbl, and S(0)2NRclRdl. In some embodiments, the compound has Formula II:
Figure imgf000014_0001
or a pharmaceutically acceptable salt thereof.
In some embodiments, the compound has Formula III:
Figure imgf000014_0002
or a pharmaceutically acceptable salt thereof.
It is further appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, can also be provided in combination in a single embodiment. Conversely, various features of the invention which are, for brevity, described in the context of a single embodiment, can also be provided separately or in any suitable subcombination.
At various places in the present specification, divalent linking substituents are described. It is specifically intended that each divalent linking substituent include both the forward and backward forms of the linking substituent. For example, -NR(CR'R")n- includes both -NR(CR'R")n- and -(CR'R")nNR-. Where the structure clearly requires a linking group, the Markush variables listed for that group are understood to be linking groups.
The term "n-membered" where n is an integer typically describes the number of ring- forming atoms in a moiety where the number of ring-forming atoms is n. For example, piperidinyl is an example of a 6-membered heterocycloalkyl ring, pyrazolyl is an example of a 5-membered heteroaryl ring, pyridyl is an example of a 6-membered heteroaryl ring, and 1,2,3,4-tetrahydro-naphthalene is an example of a 10-membered cycloalkyl group. Throughout the definitions, the term "Cn.m" indicates a range which includes the endpoints, wherein n and m are integers and indicate the number of carbons. Examples include C , C1 -6, and the like.
As used herein, the term "CN-M alkyl", employed alone or in combination with other terms, refers to a saturated hydrocarbon group that may be straight-chain or branched, having n to m carbons. In some embodiments, the alkyl group contains from 1 to 6 carbon atoms, from 1 to 4 carbon atoms, from 1 to 3 carbon atoms, or 1 to 2 carbon atoms. Examples of alkyl moieties include, but are not limited to, chemical groups such as methyl, ethyl, n- propyl, isopropyl, w-butyl, te -butyl, isobutyl, ec-butyl; higher homologs such as 2-methyl- 1-butyl, «-pentyl, 3-pentyl, «-hexyl, 1,2,2-trimethylpropyl, and the like.
As used herein, "CN-M alkenyl" refers to an alkyl group having one or more double carbon-carbon bonds and having n to m carbons. In some embodiments, the alkenyl moiety contains 2 to 6 or to 2 to 4 carbon atoms. Example alkenyl groups include, but are not limited to, ethenyl, «-propenyl, isopropenyl, «-butenyl, sec-butenyl, and the like.
As used herein, "CN_M alkynyF'refers to an alkyl group having one or more triple carbon-carbon bonds and having n to m carbons. Example alkynyl groups include, but are not limited to, ethynyl, propyn-l-yl, propyn-2-yl, and the like. In some embodiments, the alkynyl moiety contains 2 to 6 or 2 to 4 carbon atoms.
As used herein, the term "CN_M alkoxy", employed alone or in combination with other terms, refers to a group of formula -O-alkyl, wherein the alkyl group has n to m carbons.
Example alkoxy groups include methoxy, ethoxy, propoxy (e.g., n-propoxy and isopropoxy), t-butoxy, and the like. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
As used herein, the term "Cn-m alkylamino" refers to a group of formula -NH(alkyl), wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
As used herein, the term "CN.M alkoxycarbonyl" refers to a group of formula -C(0)0- alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
As used herein, the term "CN-M alkylcarbonyl" refers to a group of formula -C(O)- alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms. As used herein, the term "Cn-m alkylcarbonylamino" refers to a group of formula -NHC(0)-alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
As used herein, the term "Cn.m alkylsulfonylamino" refers to a group of formula -NHS(0)2-alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
As used herein, the term "aminosulfonyl" refers to a group of formula -S(0)2NH2, wherein the alkyl group has n to m carbon atoms.
As used herein, the term "Cn-m alkylaminosulfonyl" refers to a group of formula -S(0)2NH(alkyl), wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
As used herein, the term "di(Cn_m alkyl)aminosulfonyl" refers to a group of formula -S(0)2N(alkyl)2, wherein each alkyl group independently has n to m carbon atoms. In some embodiments, each alkyl group has, independently, 1 to 6 or 1 to 4 carbon atoms.
As used herein, the term "aminosulfonylamino" refers to a group of formula -
NHS(0)2NH2.
As used herein, the term "Cn.m alkylaminosulfonylamino" refers to a group of formula -NHS(0)2NH(alkyl), wherein the alkyl group has n to m carbon atoms. In some
embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
As used herein, the term "di(Cn.m alkyl)aminosulfonylamino" refers to a group of formula -NHS(0)2N(alkyl)2, wherein each alkyl group independently has n to m carbon atoms. In some embodiments, each alkyl group has, independently, 1 to 6 or 1 to 4 carbon atoms.
As used herein, the term "aminocarbonylamino", employed alone or in combination with other terms, refers to a group of formula -NHC(0)NH2.
As used herein, the term "Cn-m alkylaminocarbonylamino" refers to a group of formula -NHC(0)NH(alkyl), wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
As used herein, the term "di(Cn-m alkyl)aminocarbonylamino" refers to a group of formula -NHC(0)N(alkyl)2, wherein each alkyl group independently has n to m carbon atoms. In some embodiments, each alkyl group has, independently, 1 to 6 or 1 to 4 carbon atoms. As used herein, the term "Cn-m alkylcarbamyl" refers to a group of formula -C(O)- NH(alkyl), wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
As used herein, the term "thio" refers to a group of formula -S-H.
As used herein, the term "Cn.m alkylthio" refers to a group of formula -S-alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
As used herein, the term "Cn-m alkylsulfinyl" refers to a group of formula -S(0)-alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
As used herein, the term "Cn-m alkylsulfonyl" refers to a group of formula -S(0)2- alkyl, wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
As used herein, the term "amino" refers to a group of formula -NH2.
As used herein, the term "carbamyl" to a group of formula -C(0)NH2.
As used herein, the term "carbonyl", employed alone or in combination with other terms, refers to a -C(O)- group.
As used herein, the term "carboxy" refers to a group of formula -C(0)OH.
As used herein, the term "di(Cn-m-alkyl)amino" refers to a group of formula - N(alkyl)2, wherein the two alkyl groups each has, independently, n to m carbon atoms. In some embodiments, each alkyl group independently has 1 to 6 or 1 to 4 carbon atoms.
As used herein, the term "di(Cn-m-alkyl)carbamyl" refers to a group of formula - C(0)N(alkyl)2, wherein the two alkyl groups each has, independently, n to m carbon atoms. In some embodiments, each alkyl group independently has 1 to 6 or 1 to 4 carbon atoms.
As used herein, "halo" refers to F, CI, Br, or I. In some embodiments, the halo group is F or CI.
As used herein, "Cn-m haloalkoxy" refers to a group of formula -O-haloalkyl having n to m carbon atoms. An example haloalkoxy group is OCF3. In some embodiments, the haloalkoxy group is fluorinated only. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
As used herein, the term "Cn_m haloalkyl", employed alone or in combination with other terms, refers to an alkyl group having from one halogen atom to 2s+l halogen atoms which may be the same or different, where "s" is the number of carbon atoms in the alkyl group, wherein the alkyl group has n to m carbon atoms. In some embodiments, the haloalkyl group is fluorinated only. In some embodiments, the alkyl group has 1 to 6 or 1 to 4 carbon atoms.
As used herein, the term "phenyl-Ci-4 alkyl" refers to a group of formula -C
alkylene-phenyl.
As used herein, "6-10 membered aryl" refers to a monocyclic or polycyclic aromatic hydrocarbon ring having 6-10 ring members. In some embodiments, the aryl group is phenyl. In some embodiments, the aryl group is naphthyl.
As used herein "(6-10 membered aryl)-Cn-m alkyl" refers to a Cn.m alkyl group substituted by an aryl group.
As used herein, "cycloalkyl" refers to non-aromatic cyclic hydrocarbons including cyclized alkyl and/or alkenyl groups. Cycloalkyl groups can include mono- or polycyclic (e.g., having 2, 3 or 4 fused rings) groups and spirocycles. Cycloalkyl groups can have 3, 4, 5, 6, or 7 ring-forming carbons (C3.7). Ring-forming carbon atoms of a cycloalkyl group can be optionally substituted by oxo or sulfido. Cycloalkyl groups also include cycloalkylidenes. Example cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclopentenyl, cyclohexenyl, cyclohexadienyl, cycloheptatrienyl, norbornyl, norpinyl, norcarnyl, adamantyl, and the like. In some embodiments, cycloalkyl is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl. Also included in the definition of cycloalkyl are moieties that have one or more aromatic rings fused (i.e., having a bond in common with) to the cycloalkyl ring, for example, benzo or thienyl derivatives of cyclopentane, cyclohexane, and the like. A cycloalkyl group containing a fused aromatic ring can be attached through any ring-forming atom including a ring-forming atom of the fused aromatic ring.
As used herein, "(C3-7 cycloalkyl)-Cn-m alkyl" refers to a Cn-m alkyl group substituted by a C3-7 cycloalkyl group.
As used herein, "5-10 membered heteroaryl" refers to a monocyclic or polycyclic aromatic heterocycle having at least one heteroatom ring member selected from sulfur, oxygen, and nitrogen. In some embodiments, the heteroaryl ring has 1, 2, 3, 4, or 5 heteroatom ring members independently selected from nitrogen, sulfur and oxygen. In some embodiments, the heteroaryl ring has 1, 2, 3 or 4 N heteroatom ring members. In some embodiments, any ring-forming N in a heteroaryl moiety can be an N-oxide. In some embodiments, the 5-10 membered heteroaryl group is a five-membered heteroaryl ring. In some embodiments, the 5-10 membered heteroaryl is a 6-membered heteroaryl ring. A five-membered heteroaryl ring is a heteroaryl with a ring having five ring atoms wherein one or more (e.g., 1, 2, or 3) ring atoms are independently selected from N, O, and S. Exemplary five-membered ring heteroaryls are thienyl, furyl, pyrrolyl, imidazolyl, thiazolyl, oxazolyl, pyrazolyl, isothiazolyl, isoxazolyl, 1,2,3-triazolyl, tetrazolyl, 1,2,3-thiadiazolyl, 1,2,3-oxadiazolyl, 1,2,4-triazolyl, 1,2,4-thiadiazolyl, 1,2,4-oxadiazolyl, 1,3,4-triazolyl, 1,3,4- thiadiazolyl, and 1,3,4-oxadiazolyl.
A six-membered heteroaryl ring is a heteroaryl with a ring having six ring atoms wherein one or more (e.g., 1, 2, or 3) ring atoms are independently selected from N, O, and S. Exemplary six-membered ring heteroaryls are pyridyl, pyrazinyl, pyrimidinyl, triazinyl and pyridazinyl.
As used herein, the term "(5-10 membered heteroaryl)-Cn-m alkyl" refers to a Cn-m alkyl group substituted by a 5-10 membered heteroaryl group.
As used herein, "4-1 1 membered heterocycloalkyi" refers to non-aromatic monocyclic or polycyclic heterocycles having one or more ring-forming heteroatoms selected from O, N, or S and having 4-11 ring members. Included in 4-1 1 membered heterocycloalkyi groups are monocyclic 4-, 5-, 6-, and 7-membered heterocycloalkyi groups. Heterocycloalkyi groups can also include spirocycles. Example "4-1 1 membered heterocycloalkyi" groups include pyrrolidin-2-one, l,3-isoxazolidin-2-one, pyranyl, azetidinyl, morpholino, thiomorpholino, piperazinyl, tetrahydrofuranyl, tetrahydrothienyl, piperidinyl, pyrrolidinyl, isoxazolidinyl, isothiazolidinyl, pyrazolidinyl, oxazolidinyl, thiazolidinyl, imidazolidinyl, azepanyl, benzazapene, and the like. Ring-forming carbon atoms and heteroatoms of a heterocycloalkyi group can be optionally substituted by oxo or sulfido (e.g., C(O), S(O), C(S), or S(0)2, etc.). The heterocycloalkyi group can be attached through a ring-forming carbon atom or a ring- forming heteroatom. In some embodiments, the heterocycloalkyi group contains 0 to 3 double or triple bonds. In some embodiments, the heterocycloalkyi group contains 0 to 2 double bonds. Also included in the definition of heterocycloalkyi are moieties that have one or more aromatic rings fused (i.e., having a bond in common with) to the cycloalkyl ring, for example, benzo or thienyl derivatives of piperidine, morpholine, azepine, etc. A heterocycloalkyi group containing a fused aromatic ring can be attached through any ring-forming atom including a ring-forming atom of the fused aromatic ring.
As used herein, the term "(4-11 membered heterocycloalkyl)-Cn-m alkyl" refers to a Cn-m alkyl group substituted by a 4-11 membered heterocycloalkyi.
The compounds described herein can be asymmetric (e.g., having one or more stereocenters). All stereoisomers, such as enantiomers and diastereomers, are intended unless otherwise indicated. Compounds of the present invention that contain asymmetrically substituted carbon atoms can be isolated in optically active or racemic forms. Methods on how to prepare optically active forms from optically inactive starting materials are known in the art, such as by resolution of racemic mixtures or by stereoselective synthesis. Many geometric isomers of olefins, C=N double bonds, and the like can also be present in the compounds described herein, and all such stable isomers are contemplated in the present invention. Cis and trans geometric isomers of the compounds of the present invention are described and may be isolated as a mixture of isomers or as separated isomeric forms.
In some embodiments, the compound has the (^-configuration. In some
embodiments, the compound has the (^-configuration.
Resolution of racemic mixtures of compounds can be carried out by any of numerous methods known in the art. An example method includes fractional recrystallizaion using a chiral resolving acid which is an optically active, salt-forming organic acid. Suitable resolving agents for fractional recrystallization methods are, for example, optically active acids, such as the D and L forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or the various optically active camphorsulfonic acids such as β-camphorsulfonic acid. Other resolving agents suitable for fractional crystallization methods include stereoisomerically pure forms of a-methylbenzylamine (e.g., S and R forms, or diastereomerically pure forms), 2-phenylglycinol, norephedrine, ephedrine, N- methylephedrine, cyclohexylethylamine, 1,2-diaminocyclohexane, and the like.
Resolution of racemic mixtures can also be carried out by elution on a column packed with an optically active resolving agent (e.g., dinitrobenzoylphenylglycine). Suitable elution solvent composition can be determined by one skilled in the art.
Compounds of the invention also include tautomeric forms. Tautomeric forms result from the swapping of a single bond with an adjacent double bond together with the concomitant migration of a proton. Tautomeric forms include prototropic tautomers which are isomeric protonation states having the same empirical formula and total charge. Example prototropic tautomers include ketone - enol pairs, amide - imidic acid pairs, lactam - lactim pairs, enamine - imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, for example, IH- and 3H-imidazole, 1H-, 2H- and 4H- 1,2,4-triazole, IH- and 2H- isoindole, and IH- and 2H-pyrazole. Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution. Compounds of the invention can also include all isotopes of atoms occurring in the intermediates or final compounds. Isotopes include those atoms having the same atomic number but different mass numbers. For example, isotopes of hydrogen include tritium and deuterium.
The term, "compound," as used herein is meant to include all stereoisomers, geometric iosomers, tautomers, and isotopes of the structures depicted. Compounds herein identified by name or structure as one particular tautomeric form are intended to include other tautomeric forms unless otherwise specified.
All compounds, and pharmaceutically acceptable salts thereof, can be found together with other substances such as water and solvents (e.g. hydrates and solvates) or can be isolated.
In some embodiments, the compounds of the invention, or salts thereof, are substantially isolated. By "substantially isolated" is meant that the compound is at least partially or substantially separated from the environment in which it was formed or detected. Partial separation can include, for example, a composition enriched in the compounds of the invention. Substantial separation can include compositions containing at least about 50%, at least about 60%, at least about 70%, at least about 80%o, at least about 90%>, at least about 95%, at least about 97%, or at least about 99% by weight of the compounds of the invention, or salt thereof. Methods for isolating compounds and their salts are routine in the art.
The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
The expressions, "ambient temperature" and "room temperature," as used herein, are understood in the art, and refer generally to a temperature, e.g. a reaction temperature, that is about the temperature of the room in which the reaction is carried out, for example, a temperature from about 20 °C to about 30 °C.
The present invention also includes pharmaceutically acceptable salts of the compounds described herein. As used herein, "pharmaceutically acceptable salts" refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. The pharmaceutically acceptable salts of the present invention include the conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, non-aqueous media like ether, ethyl acetate, alcohols (e.g., methanol, ethanol, iso-propanol, or butanol) or acetonitrile (ACN) are preferred. Lists of suitable salts are found in Remington's
Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418 and Journal of Pharmaceutical Science, 66, 2 (1977), each of which is incorporated herein by reference in its entirety.
Synthesis
Compounds of the invention, including salts thereof, can be prepared using known organic synthesis techniques and can be synthesized according to any of numerous possible synthetic routes.
The reactions for preparing compounds of the invention can be carried out in suitable solvents which can be readily selected by one of skill in the art of organic synthesis. Suitable solvents can be substantially non-reactive with the starting materials (reactants), the intermediates, or products at the temperatures at which the reactions are carried out, e.g., temperatures which can range from the solvent's freezing temperature to the solvent's boiling temperature. A given reaction can be carried out in one solvent or a mixture of more than one solvent. Depending on the particular reaction step, suitable solvents for a particular reaction step can be selected by the skilled artisan.
Preparation of compounds of the invention can involve the protection and
deprotection of various chemical groups. The need for protection and deprotection, and the selection of appropriate protecting groups, can be readily determined by one skilled in the art. The chemistry of protecting groups can be found, for example, in T. W. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, 3rd Ed., Wiley & Sons, Inc., New York (1999), which is incorporated herein by reference in its entirety.
Reactions can be monitored according to any suitable method known in the art. For example, product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., Ή or 13C), infrared spectroscopy, spectrophotometry (e.g., UV-visible), mass spectrometry, or by chromatographic methods such as high performance liquid chromatography (HPLC), liquid chromatography-mass spectroscopy (LCMS), or thin layer chromatography (TLC). Compounds can be purified by those skilled in the art by a variety of methods, including high performance liquid chromatography (HPLC) ( "Preparative LC-MS Purification: Improved Compound Specific Method
Optimization " Karl F. Blom, Brian Glass, Richard Sparks, Andrew P. Combs J. Combi. Chem. 2004, 6(6), 874-883, which is incorporated herein by reference in its entirety) and normal phase silica chromatography.
Compounds of Formula I can be formed as shown in Scheme I. The compound (i) can be halogenated with N-chlorosuccinamide, N-bromosuccinamide or N-iodosuccinamide to give compound (ii) where X = CI, Br, or I. The halo group of (ii) can be coupled to R3-M, where M is a boronic acid, boronic ester or an appropriately substituted metal (e.g., R3-M is R3-B(OH)2 or R -Sn(Bu)4), under standard Suzuki conditions or standard Stille conditions (e.g., in the presence of a palladium(O) catalyst, such as
tetrakis(triphenylphosphine)palladium(0) and a base (e.g., a bicarbonate or carbonate base) to give a derivative of formula (iii). Alternatively, R3-M can be a cyclic amine (where M is H and attached to the amine nitrogen) with coupling to compound (ii) being performed by heating in base or under Buchwald conditions (e.g., in the presence of a palladium(O) catalyst, such as tetrakis(triphenylphosphine)palladium(0) and a base (e.g., an alkoxide base)) to afford ketone (iii). Reductive amination of the ketone (iii) can furnish the amine intermediate (v). Alternatively, ketone (iii) can be reduced to give an alcohol which can be converted to the mesylate and reacted with sodium azide to give an azide derivative (iv). The azide of compound (iv) can be converted to an amine (v) under appropriate reducing conditions, such as trimethylphosphine or TMSI. The amine (v) can be reacted with an appropriate alkylating agent RAX (e.g., Mel) or reacted under reductive amination conditions to give compound (vi). Finally compound (vi) can be reacted with a heteroaryl halide compound (e.g., Ar-X, such as 4-amino-6-chloropyrimidine-5-carbonitrile) to give a compound of Formula I. The reaction of amine (v) with RA-X can be skipped to give compounds of Formula I, wherein RA is H.
Figure imgf000024_0001
Figure imgf000024_0002
Alternatively, compounds of Formula I can also be formed as shown in Scheme II.
The ketone compound (i) can be halogenated with N-chlorosuccinamide, N- bromosuccinamide or N-iodosuccinamide to give compound (ii) where X = CI, Br, or I. Ketone (ii) can be reduced to give an alcohol (iii) which can be converted to the mesylate and reacted with sodium azide to give an azide derivative (iv). The azide of compound (iv) can be converted to an amine (v) under appropriate reducing conditions, such as
trimethylphosphine or TMSI. The amine (v) can be protected with a suitable protecting group (e.g., by reacting with Boc20) and purified by chiral chromatography to afford a single enantiomer of amine compound (v). The amino group can be deprotected (e.g., TFA when P = Boc) and reacted with an appropriate alkylating agent RAX (e.g., Mel) and the resulting secondary amine can be reacted with a heteroaryl halide compound (e.g., Ar-X) to give a compound (vi). The reaction of amine (v) with RA-X can be eliminated to give compounds (vi), wherein RA is H. Finally, the halo group of (vi) can be coupled to R3-M, where M is a boronic acid, boronic ester or an appropriately substituted metal (e.g., R3-M is R3-B(OH)2 or R3-Sn(Bu)4), under standard Suzuki conditions or standard Stille conditions (e.g., in the presence of a palladium(O) catalyst, such as tetrakis(triphenylphosphine)-palladium(0) and a base (e.g., a bicarbonate or carbonate base)) to give a derivative of formula (vii).
Alternatively, R3-M can be a cyclic amine (where M is H and attached to the amine nitrogen) with coupling to compound (vi) being performed by heating in base or under Buchwald conditions (e.g., in the presence of a palladium(O) catalyst, such as
tetrakis(triphenylphosphine)-palladium(0) and a base (e.g., an alkoxide base)) to afford compounds of Formula I (vii).
Scheme II
Figure imgf000025_0001
Compounds of Formula I, wherein L is O, N, or S, can be formed as shown in Scheme
III. The thiols, phenols or amines (i) can be alkylated using Mitsunobu conditions (e.g., R OH, DEAD, Ph3P) or standard alkylating conditions (R -Lg, Lg = leaving group) to afford thioether, ether, or alkylamine derivatives (ii), respectively. The halo group (e.g., X = Br, I) of (ii) can be coupled to R3-M, where M is a boronic acid, boronic ester or an appropriately substituted metal (e.g., R3-M is R3-B(OH)2 or R3-Sn(Bu)4), under standard Suzuki conditions or standard Stille conditions (e.g., in the presence of a palladium(O) catalyst, such as tetrakis(triphenylphosphine)-palladium(0) and a base (e.g., a bicarbonate or carbonate base)) to give a derivative of formula (iii). Alternatively, R3-M can be a cyclic amine (where M is H and attached to the amine nitrogen) with coupling to compound (ii) being performed by heating in base or under Buchwald conditions (e.g., in the presence of a palladium(O) catalyst, such as tetralds(triphenylphosphine)-palladium(0) and a base (e.g., an alkoxide base)) to afford compounds of formula (iii). The ketone (iii) can be transformed using similar methods as shown in Scheme I and II to afford compounds of Formula I (iv). Alternatively, the halo- ketone (ii) can be transformed using similar methods as shown in Scheme I and II to afford halo intermediate (v). Suzuki, Stille, Negishi or Buchwald coupling of R3-M with halo intermediate (v) by similar methods described in Schemes I and II can also afford compounds of Formula I (iv).
Scheme III
Figure imgf000026_0001
Compounds of Formula I can be formed as shown in Scheme IV. Compound (i) can be acylated with a suitable acylating reagent (e.g., R'-COCl) to form an ester which can be rearranged under Lewis acid conditions e.g., BF3/HOAC complex) to afford ketone (ii). Halogenation of ketone (ii) using NXS (e.g., NXS = N-chlorosuccinamide, N- bromosuccinamide or N-iodosuccinamide) can give compound (iii) where X = CI, Br, or I. The phenol can be converted to the triflate (iv) using standard conditions (e.g.,Tf20). The triflate group of (iv) can be coupled to R2-M, where M is a boronic acid, boronic ester or an appropriately substituted metal (e.g., R2-M is R2-B(OH)2 or R2-Sn(Bu)4), under standard Suzuki conditions or standard Stille conditions (e.g., in the presence of a palladium(O) catalyst, such as tetrakis(triphenylphosphine)palladium(0) and a base (e.g., a bicarbonate or carbonate base)) to give a derivative of formula (v). Alternatively, R2-M can be a cyclic amine (where M is H and attached to the amine nitrogen) with coupling to compound (iv) being performed by heating in base or under Buchwald conditions (e.g., in the presence of a palladium(O) catalyst, such as tetrakis(triphenylphosphine)palladium(0) and a base (e.g., an alkoxide base)) to afford ketone (v). The halo group of (v) can be coupled to R3-M, where M is a boronic acid, boronic ester or an appropriately substituted metal (e.g., R3-M is R3-B(OH)2 or R3-Sn(Bu)4), under standard Suzuki conditions or standard Stille conditions (e.g., in the presence of a palladium(O) catalyst, such as tetrakis(triphenylphosphine)palladium(0) and a base (e.g., a bicarbonate or carbonate base)) to give a derivative of formula (vi).
Alternatively, R3-M can be a cyclic amine (where M is H and attached to the amine nitrogen) with coupling to compound (iv) being performed by heating in base or under Buchwald conditions (e.g., in the presence of a palladium(O) catalyst, such as
tetrakis(triphenylphosphine)palladium(0) and a base (e.g., an alkoxide base)) to afford ketone (vi). The ketone (vi) can be transformed using similar methods as shown in Scheme I and II to afford compounds of Formula I (viii).
Alternatively, the halo-ketone (v) can be transformed using similar methods as shown in Scheme I and II to afford halo intermediate (viii). Suzuki, Stille, Negishi or Buchwald coupling of M-R3 with compound (viii) by similar methods described in Schemes I and II can also afford compounds of Formula I (vii).
Scheme IV
Figure imgf000028_0001
Ar (vii) Ar (viii)
Ketones which can be used in the processes of Scheme I, II and III can be formed as shown in Scheme V below. The carboxylic acid (i) can be activated with a coupling agent (e.g., HBTU, HATU or EDC) and then reacted with N, 0-dimethylhydroxylamine to give a N- methoxy-N-methylcarboxamide derivative (ii). Amide (ii) may then be reacted with a Grignard reagent of formula R^-MgX (X = halo) to give a ketone (iii). The ketone (iii) can be transformed using similar methods as shown in Schemes I, II and III to afford compounds of Formula I.
Scheme V
Figure imgf000028_0002
Ketones which can be used in the processes of Scheme I, II and III, can also be formed as shown in Scheme VI below. The carboxylic acid (i) can be activated with a coupling agent (e.g. HBTU or HATU) and then reacted with N, O-dimethylhydroxylamine to give a N-methoxy-N-methylcarboxamide. The thiols, phenols or amines can be alkylated using Mitsunobu conditions (e.g., R OH, DEAD, Ρ1¾Ρ) or standard alkylating conditions (R - Lg, Lg = leaving group) to afford thioether, ether or alkylamine derivatives (ii), respectively. The halo group (e.g., X = Br, or I) of (ii) can be coupled to R3-M, where M is a boronic acid, boronic ester or an appropriately substituted metal (e.g., R3-M is R3-B(OH)2 or R3-Sn(Bu)4), under standard Suzuki conditions or standard Stille conditions (e.g., in the presence of a palladium(O) catalyst, such as tetrakis(triphenylphosphine)palladium(0) and a base (e.g., a bicarbonate or carbonate base)) to give a derivative of formula (iii). Alternatively, R3-M can be a cyclic amine (where M is H and attached to the amine nitrogen) with coupling to compound (ii) being performed by heating in base or under Buchwald conditions (e.g., in the presence of a palladium(O) catalyst, such as tetrakis(triphenylphosphine)palladium(0) and a base (e.g., an alkoxide base)) to afford amides (iii) of Formula I. Reaction of compound (iii) with a Grignard reagent of formula R'-MgX (X = halo) can give ketone (iv). The ketone (iv) can be transformed using similar methods as shown in Schemes I, II and III to afford compounds of Formula I.
Scheme V I
Figure imgf000029_0001
Compounds which can be used in the processes of Schemes I-III can also be formed as shown in Scheme VII. The halo-ketone (i) can be converted to the cyano-ketone (ii) using standard cyanation conditions (e.g., Pd(0) and Zn(CN)2). Hydrolysis of the cyano group of (ii) under acid or base conditions can give the carboxylic acid which can be coupled to amines using a coupling agent (e.g., HATU, HBTU, EDC) and appropriate amines (HNR Ry) to give amide (iii) (Rx and Ry are various optionally substituted cyclic groups and non-cyclic groups, or Rx and Ry, along with the nitrogen atom to which they are attached can cyclize to form a heterocycloalkyl group). The ketone of amide (iii) can be transformed using similar methods as shown in Schemes I, II and III to afford compounds of Formula I.
Scheme VII
Figure imgf000030_0001
Additional compounds which can be used in the processes of Schemes I-III can be formed as shown in Scheme VIII. The ketone (i) can be converted to the nitro-ketone (ii) using standard nitration conditions (e.g., HN03). Reduction of the nitro group of (ii) under standard conditions (e.g., Fe, Zn, H2 over Pd/C) can give the amino compound which can be derivatized, including acylated with appropriate acylating agents (e.g., RzC=OCl, ROC=OCl, S02C1, and R'R"NC=OCl) to give ketone (iii) (Rz, R, R', and R", for example, can be various optionally substituted cyclic groups and non-cyclic groups as defined in the claims and throughout). The ketone (iii) can be transformed using similar methods as shown in Schemes I, II and III to afford compounds of Formula I. Scheme VIII
Figure imgf000031_0001
Further compounds which can be used in the processes of Schemes I-III can be formed as shown in Scheme IX. The ether (i) can be converted to a phenol (ii) using standard nitration conditions (e.g., BBr3). The halo-phenol (ii) can be converted to the cyano-phenol (iii) using standard cyanation conditions (e.g., CuCN or Pd(0) and Zn(CN)2). The phenol (iii) can be converted to the triflate (iv) using Tf20. The triflate group of (iv) can be coupled to R2-M, where M is a boronic acid, boronic ester or an appropriately substituted metal (e.g., R2- M is R2-B(OH)2 or R2-Sn(Bu)4), under standard Suzuki conditions or standard Stille conditions (e.g., in the presence of a palladium(O) catalyst, such as
tetrakis(triphenylphosphine)palladium(0) and a base (e.g., a bicarbonate or carbonate base)) to give a derivative of formula (v). Alternatively, R2-M can be a cyclic amine (where M is H and attached to the amine nitrogen) with coupling to compound (iv) being performed by heating in base or under Buchwald conditions (e.g., in the presence of a palladium(O) catalyst, such as tetrakis(triphenylphosphine)palladium(0) and a base (e.g., an alkoxide base)) to afford ketone (v). Hydrolysis of the cyano group of (v) under acid or base conditions can give the carboxylic acid which can be coupled to amines using a coupling agent (e.g., HATU, HBTU, EDC) and an appropriate amine (HNRxRy) to give amide (vi). The ketone group of amide (vi) can be transformed using similar methods as shown in Schemes I, II and III to afford compounds of Formula I. Scheme IX
Figure imgf000032_0001
Methods
The compounds of the invention can modulate activity of one or more of various kinases including, for example, phosphoinositide 3-kinases (PBKs). The term "modulate" is meant to refer to an ability to increase or decrease the activity of one or more members of the PI3K family. Accordingly, the compounds of the invention can be used in methods of modulating a PI3K by contacting the PI3K with any one or more of the compounds or compositions described herein. In some embodiments, compounds of the present invention can act as inhibitors of one or more PBKs. In further embodiments, the compounds of the invention can be used to modulate activity of a PI3K in an individual in need of modulation of the receptor by administering a modulating amount of a compound of the invention, or a pharmaceutically acceptable salt thereof. In some embodiments, modulating is inhibiting.
Given that cancer cell growth and survival is impacted by multiple signaling pathways, the present invention is useful for treating disease states characterized by drug resistant kinase mutants. In addition, different kinase inhibitors, exhibiting different preferences in the kinases which they modulate the activities of, may be used in combination. This approach could prove highly efficient in treating disease states by targeting multiple signaling pathways, reduce the likelihood of drug-resistance arising in a cell, and reduce the toxicity of treatments for disease.
Kinases to which the present compounds bind and/or modulate (e.g., inhibit) include any member of the PI3K family. In some embodiments, the PI3K is ΡΙ3Κα, ΡΒΚβ, ΡΒΚγ, or ΡΒΚδ. In some embodiments, the PBK is ΡΒΚγ or ΡΒΚδ. In some embodiments, the PBK is ΡΒΚγ. In some embodiments, the PBK is ΡΒΚδ. In some embodiments, the PBK includes a mutation. A mutation can be a replacement of one amino acid for another, or a deletion of one or more amino acids. In such embodiments, the mutation can be present in the kinase domain of the PBK.
In some embodiments, more than one compound of the invention is used to inhibit the activity of one kinase (e.g., ΡΒΚγ or ΡΒΚδ).
In some embodiments, more than one compound of the invention is used to inhibit more than one kinase, such as at least two kinases (e.g., ΡΒΚγ and ΡΒΚδ).
In some embodiments, one or more of the compounds is used in combination with another kinase inhibitor to inhibit the activity of one kinase (e.g., ΡΒΚγ or ΡΒΚδ).
In some embodiments, one or more of the compounds is used in combination with another kinase inhibitor to inhibit the activities of more than one kinase (e.g., ΡΒΚγ or ΡΒΚδ), such as at least two kinases.
The compounds of the invention can be selective. By "selective" is meant that the compound binds to or inhibits a kinase with greater affinity or potency, respectively, compared to at least one other kinase. In some embodiments, the compounds of the invention are selective inhibitors of ΡΒΚγ or ΡΒΚδ over PBKa and/or ΡΒΚβ. In some embodiments, the compounds of the invention are selective inhibitors of ΡΒΚδ (e.g., over ΡΒΚα, ΡΒΚβ and ΡΒΚγ). In some embodiments, the compounds of the invention are selective inhibitors of ΡΒΚγ (e.g., over ΡΒΚα, ΡΒΚβ and ΡΒΚδ). In some embodiments, selectivity can be at least about 2-fold, 5-fold, 10-fold, at least about 20-fold, at least about 50-fold, at least about 100-fold, at least about 200-fold, at least about 500-fold or at least about 1000-fold.
Selectivity can be measured by methods routine in the art. In some embodiments, selectivity can be tested at the Km ATP concentration of each enzyme. In some embodiments, the selectivity of compounds of the invention can be determined by cellular assays associated with particular PBK kinase activity.
Another aspect of the present invention pertains to methods of treating a kinase (such as PI3K)-associated disease or disorder in an individual (e.g., patient) by administering to the individual in need of such treatment a therapeutically effective amount or dose of one or more compounds of the present invention or a pharmaceutical composition thereof. A PI3K- associated disease can include any disease, disorder or condition that is directly or indirectly linked to expression or activity of the PI3K, including overexpression and/or abnormal activity levels. In some embodiments, the disease can be linked to Akt (protein kinase B), mammalian target of rapamycin (mTOR), or phosphoinositide-dependent kinase 1 (PDK1). In some embodiments, the mTOR-related disease can be inflammation, atherosclerosis, psoriasis, restenosis, benign prostatic hypertrophy, bone disorders, pancreatitis, angiogenesis, diabetic retinopathy, atherosclerosis, arthritis, immunological disorders, kidney disease, or cancer. A PI3K-associated disease can also include any disease, disorder or condition that can be prevented, ameliorated, or cured by modulating PI3K activity. In some embodiments, the disease is characterized by the abnormal activity of PI3K. In some embodiments, the disease is characterized by mutant PI3K. In such embodiments, the mutation can be present in the kinase domain of the PI3K.
Examples of PI3K-associated diseases include immune-based diseases involving the system including, for example, rheumatoid arthritis, allergy, asthma, glomerulonephritis, lupus, or inflammation related to any of the above.
Further examples of PI3K-associated diseases include cancers such as breast, prostate, colon, endometrial, brain, bladder, skin, uterus, ovary, lung, pancreatic, renal, gastric, or hematological cancer.
In some embodiments, the hematological cancer is acute myeloblasts leukemia (AML) or chronic myeloid leukemia (CML), or B cell lymphoma.
Further examples of PI3K-associated diseases include lung diseases such as acute lung injury (ALI) and adult respiratory distress syndrome (ARDS).
Further examples of PI3K-associated diseases include osteoarthritis, restenosis, atherosclerosis, bone disorders, arthritis, diabetic retinopathy, psoriasis, benign prostatic hypertrophy, inflammation, angiogenesis, pancreatitis, kidney disease, inflammatory bowel disease, myasthenia gravis, multiple sclerosis, or Sjogren's syndrome, and the like.
As used herein, the term "contacting" refers to the bringing together of indicated moieties in an in vitro system or an in vivo system. For example, "contacting" a PI3K with a compound of the invention includes the administration of a compound of the present invention to an individual or patient, such as a human, having a PI3K, as well as, for example, introducing a compound of the invention into a sample containing a cellular or purified preparation containing the PI3K. As used herein, the term "individual" or "patient," used interchangeably, refers to any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates, and most preferably humans.
As used herein, the phrase "therapeutically effective amount" refers to the amount of active compound or pharmaceutical agent that elicits the biological or medicinal response that is being sought in a tissue, system, animal, individual or human by a researcher, veterinarian, medical doctor or other clinician. In some embodiments, the dosage of the compound, or a pharmaceutically acceptable salt thereof, administered to a patient or individual is about 1 mg to about 2 g, about 1 mg to about 1000 mg, about 1 mg to about 500 mg, about 1 mg to about 100 mg, about 1 mg to 50 mg, or about 50 mg to about 500 mg.
As used herein, the term "treating" or "treatment" refers to one or more of (1) preventing the disease; for example, preventing a disease, condition or disorder in an individual who may be predisposed to the disease, condition or disorder but does not yet experience or display the pathology or symptomatology of the disease; (2) inhibiting the disease; for example, inhibiting a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., arresting further development of the pathology and/or symptomatology); and (3) ameliorating the disease; for example, ameliorating a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., reversing the pathology and/or symptomatology) such as decreasing the severity of disease.
Combination Therapies
One or more additional pharmaceutical agents such as, for example,
chemotherapeutics, anti-inflammatory agents, steroids, immunosuppressants, as well as Bcr- Abl, Flt-3, EGFR, HER2, JAK, c-MET, VEGFR, PDGFR, cKit, IGF-1R, RAF, FAK, and mTOR kinase inhibitors such as, for example, those described in WO 2006/056399, or other agents such as, therapeutic antibodies can be used in combination with the compounds of the present invention for treatment of PBK-associated diseases, disorders or conditions. The one or more additional pharmaceutical agents can be administered to a patient simultaneously or sequentially.
Example antibodies for use in combination therapy include but are not limited to Trastuzumab (e.g. anti-HER2), Ranibizumab (e.g. anti-VEGF-A), Bevacizumab (trade name Avastin, e.g. anti-VEGF, Panitumumab (e.g. anti-EGF ), Cetuximab (e.g. anti-EGFR), Rituxan (anti-CD20) and antibodies directed to c-MET.
One or more of the following agents may be used in combination with the compounds of the present invention and are presented as a non limiting list: a cytostatic agent, cisplatin, doxorubicin, taxotere, taxol, etoposide, irinotecan, camptostar, topotecan, paclitaxel, docetaxel, epothilones, tamoxifen, 5-fluorouracil, methoxtrexate, temozolomide, cyclophosphamide, SCH 66336, Rl 15777, L778,123, BMS 214662, Iressa, Tarceva, antibodies to EGFR, Gleevec™, intron, ara-C, adriamycin, Cytoxan, gemcitabine, Uracil mustard, Chlormethine, Ifosfamide, Melphalan, Chlorambucil, Pipobroman,
Triethylenemelamine, Triethylenethiophosphoramine, Busulfan, Carmustine, Lomustine, Streptozocin, Dacarbazine, Floxuridine, Cytarabine, 6-Mercaptopurine, 6-Thioguanine, Fludarabine phosphate, oxaliplatin, leucovirin, ELOXATIN™, Pentostatine, Vinblastine, Vincristine, Vindesine, Bleomycin, Dactinomycin, Daunorubicin, Doxorubicin, Epirubicin, Idarubicin, Mithramycin, Deoxycoformycin, Mitomycin-C, L-Asparaginase, Teniposide 17.alpha.-Ethinylestradiol, Diethylstilbestrol, Testosterone, Prednisone, Fluoxymesterone, Dromostanolone propionate, Testolactone, Megestrolacetate, Methylprednisolone,
Methyltestosterone, Prednisolone, Triamcinolone, Chlorotrianisene, Hydroxyprogesterone, Aminoglutethimide, Estramustine, Medroxyprogesteroneacetate, Leuprolide, Flutamide, Toremifene, goserelin, Cisplatin, Carboplatin, Hydroxyurea, Amsacrine, Procarbazine, Mitotane, Mitoxantrone, Levamisole, Navelbene, Anastrazole, Letrazole, Capecitabine, Reloxafine, Droloxafine, Hexamethylmelamine, Avastin, herceptin, Bexxar, Velcade, Zevalin, Trisenox, Xeloda, Vinorelbine, Porfimer, Erbitux, Liposomal, Thiotepa,
Altretamine, Melphalan, Trastuzumab, Lerozole, Fulvestrant, Exemestane, Fulvestrant, Ifosfomide, Rituximab, C225, Campath, Clofarabine, cladribine, aphidicolon, rituxan, sunitinib, dasatinib, tezacitabine, Smll, fludarabine, pentostatin, triapine, didox, trimidox, amidox, 3-AP, MDL-101 ,731, and bendamustine (Treanda).
Example chemotherapeutics include proteosome inhibitors (e.g., bortezomib), thalidomide, revlimid, and DNA-damaging agents such as melphalan, doxorubicin, cyclophosphamide, vincristine, etoposide, carmustine, and the like.
Example steroids include coriticosteroids such as dexamethasone or prednisone.
Example Bcr-Abl inhibitors include the compounds, and pharmaceutically acceptable salts thereof, of the genera and species disclosed in U.S. Pat. No. 5,521, 184, WO 04/005281, and U.S. Ser. No. 60/578,491. Example suitable Flt-3 inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 03/037347, WO 03/099771, and WO 04/046120.
Example suitable RAF inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 00/09495 and WO 05/028444.
Example suitable FAK inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 04/080980, WO 04/056786, WO 03/024967, WO 01/064655, WO 00/053595, and WO 01/014402.
Example suitable mTOR inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 201 1/025889.
In some embodiments, the compounds of the invention can be used in combination with one or more other kinase inhibitors including imatinib, particularly for treating patients resistant to imatinib or other kinase inhibitors.
In some embodiments, the compounds of the invention can be used in combination with a chemotherapeutic in the treatment of cancer, such as multiple myeloma, and may improve the treatment response as compared to the response to the chemotherapeutic agent alone, without exacerbation of its toxic effects. Examples of additional pharmaceutical agents used in the treatment of multiple myeloma, for example, can include, without limitation, melphalan, melphalan plus prednisone [MP], doxorubicin, dexamethasone, and Velcade (bortezomib). Further additional agents used in the treatment of multiple myeloma include Bcr-Abl, Flt-3, RAF and FAK kinase inhibitors. Additive or synergistic effects are desirable outcomes of combining a PI3K inhibitor of the present invention with an additional agent. Furthermore, resistance of multiple myeloma cells to agents such as dexamethasone may be reversible upon treatment with the PI3K inhibitor of the present invention. The agents can be combined with the present compound in a single or continuous dosage form, or the agents can be administered simultaneously or sequentially as separate dosage forms.
In some embodiments, a corticosteroid such as dexamethasone is administered to a patient in combination with the compounds of the invention where the dexamethasone is administered intermittently as opposed to continuously.
In some further embodiments, combinations of the compounds of the invention with other therapeutic agents can be administered to a patient prior to, during, and/or after a bone marrow transplant or stem cell transplant. Pharmaceutical Formulations and Dosage Forms
When employed as pharmaceuticals, the compounds of the invention can be administered in the form of pharmaceutical compositions. These compositions can be prepared in a manner well known in the pharmaceutical art, and can be administered by a variety of routes, depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration may be topical (including transdermal, epidermal, ophthalmic and to mucous membranes including intranasal, vaginal and rectal delivery), pulmonary (e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal or intranasal), oral or parenteral. Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal intramuscular or injection or infusion; or intracranial, e.g., intrathecal or intraventricular, administration. Parenteral administration can be in the form of a single bolus dose, or may be, for example, by a continuous perfusion pump. Pharmaceutical compositions and formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders. Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable. This invention also includes pharmaceutical compositions which contain, as the active ingredient, the compound of the invention or a pharmaceutically acceptable salt thereof, in combination with one or more pharmaceutically acceptable carriers (excipients). In some embodiments, the composition is suitable for topical administration. In making the compositions of the invention, the active ingredient is typically mixed with an excipient, diluted by an excipient or enclosed within such a carrier in the form of, for example, a capsule, sachet, paper, or other container. When the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient. Thus, the compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders.
In preparing a formulation, the active compound can be milled to provide the appropriate particle size prior to combining with the other ingredients. If the active compound is substantially insoluble, it can be milled to a particle size of less than 200 mesh. If the active compound is substantially water soluble, the particle size can be adjusted by milling to provide a substantially uniform distribution in the formulation, e.g. about 40 mesh. The compounds of the invention may be milled using known milling procedures such as wet milling to obtain a particle size appropriate for tablet formation and for other formulation types. Finely divided (nanoparticulate) preparations of the compounds of the invention can be prepared by processes known in the art, e.g., see International App. No. WO 2002/000196.
Some examples of suitable excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, and methyl cellulose. The formulations can additionally include: lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propylhydroxy-benzoates; sweetening agents; and flavoring agents. The compositions of the invention can be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the patient by employing procedures known in the art.
The compositions can be formulated in a unit dosage form, each dosage containing from about 5 to about 1000 mg (1 g), more usually about 100 to about 500 mg, of the active ingredient. The term "unit dosage forms" refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
In some embodiments, the compositions of the invention contain from about 5 to about 50 mg of the active ingredient. One having ordinary skill in the art will appreciate that this embodies compositions containing about 5 to about 10, about 10 to about 15, about 15 to about 20, about 20 to about 25, about 25 to about 30, about 30 to about 35, about 35 to about 40, about 40 to about 45, or about 45 to about 50 mg of the active ingredient.
In some embodiments, the compositions of the invention contain from about 50 to about 500 mg of the active ingredient. One having ordinary skill in the art will appreciate that this embodies compositions containing about 50 to about 100, about 100 to about 150, about 150 to about 200, about 200 to about 250, about 250 to about 300, about 350 to about 400, or about 450 to about 500 mg of the active ingredient.
In some embodiments, the compositions of the invention contain from about 500 to about 1000 mg of the active ingredient. One having ordinary skill in the art will appreciate that this embodies compositions containing about 500 to about 550, about 550 to about 600, about 600 to about 650, about 650 to about 700, about 700 to about 750, about 750 to about 800, about 800 to about 850, about 850 to about 900, about 900 to about 950, or about 950 to about 1000 mg of the active ingredient.
Similar dosages may be used of the compounds described herein in the methods and uses of the invention.
The active compound can be effective over a wide dosage range and is generally administered in a pharmaceutically effective amount. It will be understood, however, that the amount of the compound actually administered will usually be determined by a physician, according to the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
For preparing solid compositions such as tablets, the principal active ingredient is mixed with a pharmaceutical excipient to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention. When referring to these preformulation compositions as homogeneous, the active ingredient is typically dispersed evenly throughout the composition so that the composition can be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules. This solid
preformulation is then subdivided into unit dosage forms of the type described above containing from, for example, about 0.1 to about 1000 mg of the active ingredient of the present invention.
The tablets or pills of the present invention can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action. For example, the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former. The two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release. A variety of materials can be used for such enteric layers or coatings, such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol, and cellulose acetate.
The liquid forms in which the compounds and compositions of the present invention can be incorporated for administration orally or by injection include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles. Compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders. The liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described supra. In some embodiments, the compositions are administered by the oral or nasal respiratory route for local or systemic effect. Compositions can be nebulized by use of inert gases. Nebulized solutions may be breathed directly from the nebulizing device or the nebulizing device can be attached to a face mask, tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions can be administered orally or nasally from devices which deliver the formulation in an appropriate manner.
Topical formulations can contain one or more conventional carriers. In some embodiments, ointments can contain water and one or more hydrophobic carriers selected from, for example, liquid paraffin, polyoxyethylene alkyl ether, propylene glycol, white Vaseline, and the like. Carrier compositions of creams can be based on water in combination with glycerol and one or more other components, e.g. glycerinemonostearate, PEG- glycerinemonostearate and cetylstearyl alcohol. Gels can be formulated using isopropyl alcohol and water, suitably in combination with other components such as, for example, glycerol, hydroxyethyl cellulose, and the like. In some embodiments, topical formulations contain at least about 0.1, at least about 0.25, at least about 0.5, at least about 1, at least about 2, or at least about 5 wt % of the compound of the invention. The topical formulations can be suitably packaged in tubes of, for example, 100 g which are optionally associated with instructions for the treatment of the select indication, e.g., psoriasis or other skin condition.
The amount of compound or composition administered to a patient will vary depending upon what is being administered, the purpose of the administration, such as prophylaxis or therapy, the state of the patient, the manner of administration, and the like. In therapeutic applications, compositions can be administered to a patient already suffering from a disease in an amount sufficient to cure or at least partially arrest the symptoms of the disease and its complications. Effective doses will depend on the disease condition being treated as well as by the judgment of the attending clinician depending upon factors such as the severity of the disease, the age, weight and general condition of the patient, and the like.
The compositions administered to a patient can be in the form of pharmaceutical compositions described above. These compositions can be sterilized by conventional sterilization techniques, or may be sterile filtered. Aqueous solutions can be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration. The pH of the compound preparations typically will be between 3 and 1 1, more preferably from 5 to 9 and most preferably from 7 to 8. It will be understood that use of certain of the foregoing excipients, carriers, or stabilizers will result in the formation of pharmaceutical salts.
The therapeutic dosage of a compound of the present invention can vary according to, for example, the particular use for which the treatment is made, the manner of administration of the compound, the health and condition of the patient, and the judgment of the prescribing physician. The proportion or concentration of a compound of the invention in a
pharmaceutical composition can vary depending upon a number of factors including dosage, chemical characteristics (e.g., hydrophobicity), and the route of administration. For example, the compounds of the invention can be provided in an aqueous physiological buffer solution containing about 0.1 to about 10% w/v of the compound for parenteral administration. Some typical dose ranges are from about 1 μg/kg to about 1 g/kg of body weight per day. In some embodiments, the dose range is from about 0.01 mg/kg to about 100 mg/kg of body weight per day. The dosage is likely to depend on such variables as the type and extent of progression of the disease or disorder, the overall health status of the particular patient, the relative biological efficacy of the compound selected, formulation of the excipient, and its route of administration. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.
The compositions of the invention can further include one or more additional pharmaceutical agents such as a chemotherapeutic, steroid, anti-inflammatory compound, or immunosuppressant, examples of which are listed herein.
Labeled Compounds and Assay Methods
Another aspect of the present invention relates to labeled compounds of the invention (radio-labeled, fluorescent-labeled, etc.) that would be useful not only in imaging techniques but also in assays, both in vitro and in vivo, for localizing and quantitating PI3K in tissue samples, including human, and for identifying PI3K ligands by inhibition binding of a labeled compound. Accordingly, the present invention includes PI3K assays that contain such labeled compounds.
The present invention further includes isotopically-labeled compounds of the invention. An "isotopically" or "radio-labeled" compound is a compound of the invention where one or more atoms are replaced or substituted by an atom having an atomic mass or mass number different from the atomic mass or mass number typically found in nature (i.e., naturally occurring). Suitable radionuclides that may be incorporated in compounds of the present invention include but are not limited to 3H (also written as T for tritium), nC, 13C, 14C, 13N, 15N, , 50, 170, 180, 18F, 35S, 36C1, 82Br, 75Br, 76Br, 77Br, 123I, 12 I, 125I and 131I. The radionuclide that is incorporated in the instant radio-labeled compounds will depend on the specific application of that radio-labeled compound. For example, for in vitro PI3K labeling
3 14 82 125 131 35
and competition assays, compounds that incorporate H, C, Br, I , I, S or will generally be most useful. For radio-imaging applications nC, 18F, 125I, !231, 12 I, 1311, 75Br, 76Br or 77Br will generally be most useful.
It is understood that a "radio-labeled " or "labeled compound" is a compound that has incorporated at least one radionuclide. In some embodiments the radionuclide is selected from the group consisting of 3H, 14C, 1251 , 35S and 82Br. In some embodiments, one or more H atoms for any compound described herein is each replaced by a deuterium atom.
The present invention can further include synthetic methods for incorporating radioisotopes into compounds of the invention. Synthetic methods for incorporating radio-isotopes into organic compounds are well known in the art, and an ordinary skill in the art will readily recognize the methods applicable for the compounds of invention.
A labeled compound of the invention can be used in a screening assay to
identify/evaluate compounds. For example, a newly synthesized or identified compound {i.e., test compound) which is labeled can be evaluated for its ability to bind a PI3K by monitoring its concentration variation when contacting with the PI3K, through tracking of the labeling. For example, a test compound (labeled) can be evaluated for its ability to reduce binding of another compound which is known to bind to a PI3K {i.e., standard compound).
Accordingly, the ability of a test compound to compete with the standard compound for binding to the PI3K directly correlates to its binding affinity. Conversely, in some other screening assays, the standard compound is labeled and test compounds are unlabeled.
Accordingly, the concentration of the labeled standard compound is monitored in order to evaluate the competition between the standard compound and the test compound, and the relative binding affinity of the test compound is thus ascertained. Kits
The present invention also includes pharmaceutical kits useful, for example, in the treatment or prevention of PI3K-associated diseases or disorders, such as cancer, which include one or more containers containing a pharmaceutical composition comprising a therapeutically effective amount of a compound of the invention. Such kits can further include, if desired, one or more of various conventional pharmaceutical kit components, such as, for example, containers with one or more pharmaceutically acceptable carriers, additional containers, etc., as will be readily apparent to those skilled in the art. Instructions, either as inserts or as labels, indicating quantities of the components to be administered, guidelines for administration, and/or guidelines for mixing the components, can also be included in the kit.
The invention will be described in greater detail by way of specific examples. The following examples are offered for illustrative purposes, and are not intended to limit the invention in any manner. Those of skill in the art will readily recognize a variety of non- critical parameters which can be changed or modified to yield essentially the same results. The compounds of the Examples have been found to be PI3K inhibitors according to at least one assay described herein.
EXAMPLES
The example compounds below containing one or more chiral centers were obtained in racemate form or as isomeric mixtures, unless otherwise specified. Salt stoichiometry which is indicated any of the products below is meant only to indicate a probable stoichiometry, and should not be construed to exclude the possible formation of salts in other stoichiometrics. Example 1.
4-Amino-6-[(l-{5-chloro-2-methoxy-4-methyl-3-[5-(methyIsulfonyl)pyridin-3- yI]phenyl}ethyI)amino] cid salt
Figure imgf000044_0001
Step 1. l-(3-Bromo-5-chloro-2-hydroxy-4-methylphenyl)ethanone
Figure imgf000044_0002
To a stirred solution of l-(5-chloro-2-hydroxy-4-methylphenyl)ethanone (10.0 g, 54.2 mmol, from Aldrich) in acetic acid (100 mL) was added N-bromosuccinimide (11.6 g, 65.0 mmol) and the resulting mixture was stirred at rt for 18 h. The reaction mixture was concentrated in vacuo, then neutralized with sat. sodium bicarbonate, and the insoluble succinimide was removed by filtration. The filtrate was extracted with ethyl acetate (EtOAc). The combined organic layers were washed with brine, dried over sodium sulfate, and then concentrated to dryness under reduced pressure. The crude product was recrystalized from a mixture of EtOAc and hexane (1 1.4 g, 80%). Step 2. l-(3-Bromo-5-chloro-2-methoxy-4-methylphenyl)ethanone
Figure imgf000045_0001
A mixture of l-(3-bromo-5-chloro-2-hydroxy-4-methylphenyl)ethanone (10.0 g, 37.9 mmol), dimethyl sulfate (4.31 mL, 45.5 mmol) and potassium carbonate (10.5 g, 75.9 mmol) in acetone (200 mL) was heated at reflux overnight. After evaporation to dryness, the mixute was diluted with water, and extracted with EtOAc. The combined organic layers were washed with brine, dried over sodium sulfate, and evaporated to dryness. The residue was purified on silica gel, eluting with 0 to 20% EtOAc in hexane, to yield the desired product (8.8 g, 84%). LCMS calculated for Ci0HnBrClO2 (M+H)+: m/z = 227.0; found: 277.0. Step 3. l-{5-chloro-2-methoxy-4- ethyl-3-[5-(methylsulfonyl)pyridin-3-yl]phenyl}ethanone
Figure imgf000045_0002
Into a microwave vial was added l-(3-bromo-5-chloro-2-methoxy-4- methylphenyl)ethanone (0.040 g, 0.14 mmol), [5-(methylsulfonyl)pyridin-3-yl]boronic acid (0.035 g, 0.17 mmol), 1.0 M sodium carbonate (0.35 mL, 0.35 mmol), 1,4-dioxane (0.87 mL) and tetrakis(triphenylphosphine)palladium(0) (0.010 g, 0.0086 mmol). The mixture was bubbled with N2 for 5 min, then heated at 100 °C overnight. The cooled reaction mixture was purified on silica gel to give the desired product (0.035 g, 71%). LCMS calculated for C16H17C1N04S (M+H)+: m/z = 354.1 ; found: 354.0
Step 4. l-{5-chloro-2-methoxy-4-methyl-3-[5-(methylsulfonyl)pyridin-3- yl]phenyl}ethanamine
Figure imgf000046_0001
A mixture of l-{5-chloro-2-methoxy-4-methyl-3-[5-(methylsulfonyl)pyridin-3- yl]phenyl}ethanone (0.035 g, 0.099 mmol), ammonium acetate (0.08 g, 1 mmol) and 1.0 M sodium cyanoborohydride in tetrahydrofuran (THF) (0.25 mL, 0.25 mmol) in methanol (0.2 mL) and acetonitrile (0.2 mL) was heated at 65 °C overnight. The mixture was cooled to room temperature (rt), quenched with sat. NaHC(¾ solution, and extracted with
dichloromethane. The combined organic layers were dried over MgS04 and concentrated to give the desired product, which was used in the next step directly (0.024 g, 69%). LCMS calculated for Ci6H,7ClN03S (M-NH2)+: m/z = 338.1; found: 338.0.
Step 5. 4-amino-6-[(l-{5-chloro-2-methoxy-4-methyl-3-[5-(methylsulfonyl)pyridin-3- yl]phenyl}ethyl)amino]pyrimidine-5-carbonitrile trifluoroacetic acid salt
A mixture of l-{5-chloro-2-methoxy-4-methyl-3-[5-(methylsulfonyl)pyridin-3- yl]phenyl}ethanamine (24 mg, 0.068 mmol), 4-amino-6-chloropyrimidine-5-carbonitrile (1 1 mg, 0.074 mmol) and NN-diisopropylethylamine (0.035 mL, 0.20 mmol) in 1-butanol (0.68 mL) was heated at 120 °C overnight. The mixture was filtered and purified on prep- LCMS (XBridge CI 8 Column, eluting with a gradient of acetonitrile in water with 0.05% trifluoroacetic acid, at flow rate of 30 mL/min) to give the desired product as the TFA salt. LCMS calculated for
Figure imgf000046_0002
(M+H)+: m/z = 473.1 ; found: 473.1. !H NMR (DMSO- ck, 300 MHz) δ 9.11 (1H, s), 8.92 (0.5 H, m), 8.81 (0.5 H, m), 8.37 (0.5 H, m), 8.25 (0.5H, m), 8.00 (2H, m), 7,71 (1H, s), 7.51 (2H, br s), 5.62 (1H, m), 3.38 (3H, s), 3.32 (3H, s), 2.04 (3H, s), 1.45 (3H, d, J = 6.9 Hz) ppm. Examples 2-5
The compounds of Examples 2-5, set out in Table 1 below, were prepared by methods analogous to those of Example 1.
Table 1
Figure imgf000047_0001
Figure imgf000047_0002
Ή NMR data (Varian Inova 500 spectrometer, a Mercury 400 spectrometer, or a Varian 300 spectrometer, DMSO-c ) and Mass spectral data (MS) for the compounds of Examples 2-5 are provided below in Table 2.
Table 2
Figure imgf000047_0003
Figure imgf000048_0001
Example 6
4-Amino-6-({l-[5-chloro-3-(5-cyanopyridin-3-yl)-4-fluoro-2-methoxyphenyl]ethyl}- amino)pyrimidine-5-carboni
Figure imgf000048_0002
Step 1: l-(5-Chloro-4-fluoro-2-hydroxy henyl)ethanone
Figure imgf000048_0003
To 4-chloro-3-fluorophenol (Oakwood #004459, 5.1 g, 35 mmol) was added acetyl chloride (3.6 mL, 51 mmol) and the resulting mixture was heated at 60° C for 2 hours.
Aluminum trichloride (7.0 g, 52 mmol) was added and the mixture was heated at 180° C for 30 minutes. The reaction mixture was then cooled to room temperature and slowly quenched with 1M HCl while cooling in an ice-bath and stirred for 30 minutes. The precipitate was washed well with water and dried under vacuum to give the desired product (quantitative). This material was used without further purification. lR MR (300 MHz, CDC13): δ 12.41 (s, 1 H), 7.80 (m, 1 H), 6.79 (m, 1 H), 2.60 (s, 3 H). Step 2; l-(3-Bromo-5-chloro-4-fluoro-2-hydroxyphenyl)ethanone
Figure imgf000049_0001
To a stirred solution of l-(5-chloro-4-fluoro-2-hydroxyphenyl)ethanone (7.9 g, 42 mmol) in acetic acid (80 mL, 1.0 mol) was added N-bromosuccinimide (9.0 g, 50 mmol) and the resulting mixture was stirred at rt for 18 h. The reaction mixture was concentrated in vacuo, neutralized with saturated sodium bicarbonate and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, and concentrated to dryness under reduced pressure. The residue was purified on silica gel, eluting with 0 to 20% ethyl acetate in hexane, to yield the desired product (93% yield). [U NMR (300 MHz, CDC13): 5 13.1 (s, 1 H), 7.79 (m, 1 H), 2.62 (s, 3 H).
Step 3: l-(3-Bromo-5-chloro-4-fluoro-2-methoxyphenyl)ethanone
Figure imgf000049_0002
A mixture of l -(3-bromo-5-chloro-4-fluoro-2-hydroxyphenyl)ethanone (5.3 g, 20 mmol), potassium carbonate (7.2 g, 52mmol) and methyl iodide (2.7 mL, 44 mmol)i N,N- dimethylformamide (20 mL) was heated at 60 °C for 1 hour. The mixture was filtered and washed with ethyl acetate. The filtrate was diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, and evaporated to dryness. The residue was purified on silica gel, eluting with 0 to 20% ethyl acetate in hexane, to yield the desired product (39% yield). LCMS calculated for
C9H8BrClF02 (M+H)+: m/z = 280.9, 282.9; found: 280.8, 282.8 Step 4: l-(3-Bromo-5-chloro-4-fluoro-2-methoxyphenyl)ethanamine
Figure imgf000049_0003
A mixture of l-(3-bromo-5-chloro-4-fluoro-2-methoxyphenyl)ethanone (0.44 g, 1.6 mmol) and ammonium acetate (1.8 g, 23 mmol) in acetonitrile (3.0 mL) and methanol (3.0 mL) was heated at 65 °C for an hour. Sodium cyanoborohydride (0.244 g, 3.88 mmol) was added and the mixture was heated at 65 °C overnight. Purification by preparative LCMS (pH=l 0) gave the desired compound (23% yield). LCMS calculated for CgHsBrClFO (M-
NH2)+: m/z = 264.9, 266.9; found: 264.9, 266.9. ¾ NMR (300 MHz, CDC13): δ 8.81 (br s, 2 H), 7.80 (s, 1 H), 4.80 (m, 1 H), 3.97 (s, 3 H), 1.69 (m, 3 H).
Step 5: 4-Amino-6-{[l-(3-bromo-5-chloro-4-fluoro-2- methoxyphenyl)ethyl]amino}pyrimidine-5-carbonitrile
Figure imgf000050_0001
A mixture of l -(3-bromo-5-chloro-4-fluoro-2-methoxyphenyl)ethanamine (40 mg, 0.14 mmol), 4-amino-6-chloropyrimidine-5-carbonitrile (J & W PharmLab, #70-0156 33 mg, 0.21 mmol) and N,N-diisopropylethylamine (74 μί, 0.43 mmol) in ethanol (1.6 mL, 27 mmol) was heated at 100 °C overnight. Purification by preparative LC MS (pHIO) gave the desired compound (54% yield). LCMS calculated for
Figure imgf000050_0002
(M+H)+: m/z =
400.0, 402.0; found: 400.0, 402.0. *H NMR (300 MHz, DMSO-<¾): δ 7.95 (s, 1 H), 7.77 (m, 1 H), 7.28 (br s, 2 H), 5.56 (m, 1 H), 3.92 (s, 1 H), 1.40 (m, 3 H). Step 6: 4-Amino-6-({l-[5-chloro-3-(5-cyanopyridin~3-yl)-4-fluoro-2- methoxyphenyl] ethyl} amino)pyrimidine-5-carbonitrile
Into a microwave vial was added 4-amino-6-{[l -(3-bromo-5-chloro-4-fluoro-2- methoxyphenyl)ethyl]amino}pyrimidine-5-carbonitrile (8.0 mg, 0.020 mmol), 5-(4,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)nicotinonitrile (5.51 mg, 0.0240 mmol), a solution of sodium carbonate (48 uL, 0.050 mmol) in water (48
Figure imgf000050_0003
1,4-dioxane (0.2 mL, 2 mmol) and tetrakis(triphenylphosphine)palladium(0) (1.4 mg, 0.0012 mmol). The mixture was bubbled with N2 for 5 minutes and heated at 90 °C for 2h. Purification by preparative LCMS (pH 10) gave the desired compound (37% yield). LCMS calculated for C20H1 6CIFN7O
(M+H)+: m/z = 424.1 ; found: 424.1. ]H NMR (DMSO-i&, 500 MHz) δ 9.12 (s, 1 H), 8.97 (s, 1 H), 8.50 (s, 1 H), 7.95 (s, 1 H), 7.88 (m, 1 H), 7.70 (m, 1 H), 7.22 (br s, 2 H), 5.63 (m, 1 H), 3.41 (s, 3 H), 1.42 (m, 3 H).
Examples 7-10
The compounds of Examples 7-10, set out in Table 3 below, were prepared by methods analogous to those of Example 6.
Figure imgf000051_0001
Figure imgf000051_0002
JH NMR data (Varian Inova 500 spectrometer, a Mercury 400 spectrometer, or a Varian 300 spectrometer, DMSO-i¾) and Mass spectral data (MS) for the compounds of Examples 7-10 are provided below in Table 4. Table 4
Figure imgf000052_0004
Example 11
4-Amino-6-[(l-{4,5-dichloro-2-methoxy-3-[5-(methylsulfonyI)pyridin-3- yl]phenyI}ethyl)amino]pyrimidine-5-carbonitrile
Figure imgf000052_0001
Step A: l-(4, 5-Dichloro-2-hydroxyphen l)ethanone
Figure imgf000052_0002
The desired compound was prepared according to the procedure of Example 6, Step 1, using 3,4-dichlorophenol as the starting material in 96% yield. lH NMR (300 MHz, CDCI3): δ 12.17 (s, 1 H), 7.79 (s, 1 H), 7.12 (s, 1 H), 2.62 (s, 3 H).
Step B: l-(4,5-Dichloro-2-hydroxy-3-iodophenyl)ethanone
Figure imgf000052_0003
The desired compound was prepared according to the procedure of Example 6, Step 2, g l-(4,5-dichloro-2-hydroxyphenyl)ethanone and N-iodosuccinimide (instead of N- bromosuccinimide) as the starting materials in 71% yield. LCMS for CgHgC^IC^ (M+H) m/z = 330.9, 332.9; Found: 330.8, 332.9.
Step C: l-(4,5-Dichloro-3-iodo-2-methox henyl)ethanone
Figure imgf000053_0001
A solution of l-(4,5-dichloro-2-hydroxy-3-iodophenyl)ethanone (4.9 g, 15 mmol), triphenylphosphine (5.5 g, 21 mmol), and methanol (0.91 mL, 22 mmol) in tetrahydrofuran (44 mL) at 0 °C was treated with diisopropyl azodicarboxylate (4.1 mL, 21 mmol) dropwise and stirred at 20 °C for 16 h. The reaction mixture was concentrated, diluted with ethyl acetate, and poured into water. The organic layer was separated, washed with brine, dried with magnesium sulfate, filtered, and concentrated to a crude residue. Purification by flash column chromatography using ethyl acetate in hexanes (5% - 30%) gave the desired product
(4.1 g, 79%). LCMS for C9H8C12I02 (M+H)+: m/z = 344.9, 346.9; Found: 344.9, 346.7. Step D: l-(4,5-Dichloro-3-iodo-2-methox henyl)ethanol
Figure imgf000053_0002
A solution of l-(4,5-dichloro-3-iodo-2-methoxyphenyl)ethanone (4.1 g, 12 mmol) in methanol (59 mL) at 0 °C was treated with sodium tetrahydroborate (0.45 g, 12 mmol) and stirred at 0 °C for 1 h. The reaction mixture was quenched with water (50 mL) 0 °C, and diluted with saturated sodium bicarbonate (50 mL) and ethyl acetate (100 mL). The aqueous layer was separated and extracted with additional ethyl acetate (50 mL). The combined organic layers were washed with brine (50 mL), dried with magnesium sulfate, filtered, and concentrated to give the desired product (quantitative). This material was used without further purification. LCMS for CgHgC^IO (M-OH)+: m/z = 328.9, 330.9; Found: 328.9, 330.9. Step E: l-(l-Azidoethyl)-4, 5-dichloro-3-iodo-2-methoxybenzene
Figure imgf000054_0001
A solution of l-(4,5-dichloro-3-iodo-2-methoxyphenyl)ethanol (4.09 g, 12 mmol) in methylene chloride (47 mL) at 0 °C was treated with N,N-diisopropylethylamine (3.3 mL, 19 mmol) followed by methanesulfonyl chloride (1.4 mL, 18 mmol) and stirred at 0 °C for 30 min. The reaction mixture was diluted with water (100 mL) and extracted with methylene chloride (2 x 150 mL). The combined organic extracts were dried with magnesium sulfate, filtered, and concentrated to give the intermediate mesylate. This material was used immediately without further purification. A solution of the crude mesylate in N,N- dimethylformamide (29 mL) was treated with sodium azide (2.3 g, 35 mmol) and stirred at 60 °C for 1 h. The reaction mixture was diluted with ethyl acetate (150 mL) and washed with a mixture of saturated sodium bicarbonate (50 mL) and water (50 mL), brine (100 mL), dried with magnesium sulfate, filtered, and concentrated to a crude residue. Purification by flash column chromatography using ethyl acetate in hexanes (0% - 30%) gave the desired product (3.3 g, 75%). LCMS for C9H8C12I0 (M-N3)+: m/z = 328.9, 330.9; Found: 328.9, 330.9.
Step F: l-(4,5-Dichloro-3-iodo-2-methox henyl)ethanamine
Figure imgf000054_0002
A solution of l-(l-azidoethyl)-4,5-dichloro-3-iodo-2-methoxybenzene (3.3 g, 8.9 mmol) in tetrahydrofuran (20 mL) and water (6.4 mL) was treated with 1.0 M
trimethylphosphine in tetrahydrofuran (11 mL, 11 mmol) and stirred at 20 °C for 1.5 hours. The reaction mixture was concentrated, diluted with ethyl acetate and extracted with 1 M HCl (2x). The combined aqueous layers were neutralized with solid sodium bicarbonate, and extracted with ethyl acetate (2x). The combined organic extracts were washed with brine, dried with magnesium sulfate, filtered, and concentrated to give the desired product (2.2 g, 72%). This material was used without further purification. LCMS for CgHgC^IO (M-
NH2)+: m/z = 328.9, 330.9; Found: 328.8, 330.8. Step G: 4-Amino-6-{fl-(4,5-dichloro-3-iodo-2-methoxyphenyl)ethyl]amino}pyrimidine-5- carbonitrile
Figure imgf000055_0001
The desired compound was prepared according to the procedure of Example 6, Step 5, using l-(4,5-dichloro-3-iodo-2-methoxyphenyl)ethanamine as the starting material in 55% yield. LCMS for C14H13CI2IN5O (M+H)+: m/z = 464.0, 465.9; Found: 463.9, 465.9.
Step H: 4-Amino-6-[(l-{4,5-dichloro-2-methoxy-3-[5-(methylsulfonyl)pyridin-3- yl]phenyl}ethyl)amino]pyrimidine-5-carbonitrile
The desired compound was prepared according to the procedure of Example 6, Step 6, using 4-amino-6-{[l -(4,5-dichloro-3-iodo-2-methoxyphenyl)ethyl]amino}-pyrimidine-5- carbonitrile as the starting material in 6% yield. LCMS for C20H19CI2N6O3S (M+H)+: m/z = 493.1, 495.1; Found: 493.0, 495.0.
Examples 12-14
The compounds of Examples 12-14, set out in Table 5 below, were prepared by methods analogous to those described in Example 11.
Table 5
Figure imgf000055_0002
Figure imgf000055_0003
Figure imgf000056_0001
5- carbonitrile
1
Ή NMR data (Varian Inova 500 spectrometer, a Mercury 400 spectrometer, or a Varian 300 spectrometer, DMSO-i 6) and Mass spectral data (MS) for the compounds of Examples 12-14 are provided below in Table 6.
Table 6
Figure imgf000056_0002
Example Al : PI3K Enzyme Assay
PI3-Kinase luminescent assay kit including lipid kinase substrate, D-myo- phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P2)D (+)-sn-l,2-di-0-octanoylglyceryl, 3- O-phospho linked (PIP2), biotinylated I(1,3,4,5)P4, PI(3,4,5)P3 Detector Protein, was purchased from Echelon Biosciences (Salt Lake City, UT). AlphaScreen™ GST Detection Kit including donor and acceptor beads was purchased from PerkinElmer Life Sciences
(Waltham, MA). ΡΙ3Κδ (ρΐ ΐθδ /ρ85α) was purchased from Millipore (Bedford, MA). ATP, MgCl2, DTT, EDTA, HEPES and CHAPS were purchased from Sigma-Aldrich (St. Louis, MO). AlphaScreen Assay for ΡΒΚδ
The kinase reaction was conducted in 384-well REMP plate from Thermo Fisher Scientific in a final volume of 40
Figure imgf000057_0001
Inhibitors were first diluted serially in DMSO and added to the plate wells before the addition of other reaction components. The final concentration of DMSO in the assay was 2%. The PI3K assays were carried out at room temperature in 50 mM HEPES, pH 7.4, 5mM MgCl2, 50 mM NaCl, 5mM DTT and CHAPS 0.04%. Reactions were initiated by the addition of ATP, the final reaction mixture consisted of 20 μΜ PIP2, 20 μΜ ATP, 1.2nM ΡΙ3Κδ were incubated for 20 min. 10 μΕ of reaction mixture was then transferred to 5 μΐ. 50nM biotinylated I(1,3,4,5)P4 in quench buffer: 50 mM HEPES pH 7.4, 150 mM NaCl, 10 mM EDTA, 5 mM DTT, 0.1 % Tween-20, followed with the addition of 10 iL AlphaScreen™ donor and acceptor beads suspended in quench buffer containing 25nM PI(3,4,5)P3 detector protein. The final concentration of both donor and acceptor beads is 20 mg/ml. After plate sealing, the plate was incubated in a dark location at room temperature for 2 hours. The activity of the product was detennined on Fusion-alpha microplate reader (Perkin-Elmer). IC50 determination was performed by fitting the curve of percent control activity versus the log of the inhibitor concentration using the GraphPad Prism 3.0 software.
Example A2: PI3K Enzyme Assay
Materials: Lipid kinase substrate, phosphoinositol-4,5-bisphosphate (PIP2), was purchased from Echelon Biosciences (Salt Lake City, UT). PI3K isoforms α, β, δ and γ were purchased from Millipore (Bedford, MA). ATP, MgCl2, DTT, EDTA, MOPS and CHAPS were purchased from Sigma-Aldrich (St. Louis, MO).
The kinase reaction was conducted in clear-bottom 96-well plate from Thermo Fisher Scientific in a final volume of 24 Inhibitors were first diluted serially in DMSO and added to the plate wells before the addition of other reaction components. The final concentration of DMSO in the assay was 0.5%. The PI3K assays were carried out at room temperature in 20 mM MOPS, pH 6.7, 10 mM MgCl2, 5 mM DTT and CHAPS 0.03%. The reaction mixture was prepared containing 50 μΜ PIP2, kinase and varying concentration of inhibitors. Reactions were initiated by the addition of ATP containing 2.2 μθ [γ-33Ρ]ΑΤΡ to a final concentration of 1000 uM. The final concentration of PI3K isoforms α, β, δ and γ in the assay were 1.3, 9.4, 2.9 and 10.8 nM respectively. Reactions were incubated for 180 min and terminated by the addition of 100 μί of 1 M potassium phosphate pH 8.0, 30 mM EDTA quench buffer. A 100 μΐ, aliquot of the reaction solution was then transferred to 96-well Millipore MultiScreen IP 0.45 μιη PVDF filter plate (The filter plate was prewetted with 200 μΐ, 100% ethanol, distilled water, and 1 M potassium phosphate pH 8.0, respectively). The filter plate was aspirated on a Millipore Manifold under vacuum and washed with 18 x 200 μΐ^ wash buffer containing 1 M potassium phosphate pH 8.0 and 1 mM ATP. After drying by aspiration and blotting, the plate was air dried in an incubator at 37 °C overnight. Packard TopCount adapter (Millipore) was then attached to the plate followed with addition of 120 μΐ^ Microscint 20 scintillation cocktail (Perkin Elmer) in each well. After the plate sealing, the radioactivity of the product was determined by scintillation counting on Topcount (Perkin- Elmer). IC50 determination was performed by fitting the curve of percent control activity versus the log of the inhibitor concentration using the GraphPad Prism 3.0 software.
Example A3: PI3K6 scintillation proximity assay
Materials
[γ-33Ρ]ΑΤΡ (lOmCi/mL) was purchased from Perkin-Elmer (Waltham, MA). Lipid kinase substrate, D-myo-Phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P2)D (+)-sn-l,2- di-O-octanoylglyceryl, 3-O-phospho linked (PIP2), CAS 204858-53-7, was purchased from Echelon Biosciences (Salt Lake City, UT). ΡΙ3Κδ (pi 10δ /ρ85α) was purchased from Millipore (Bedford, MA). ATP, MgCl2, DTT, EDTA, MOPS and CHAPS were purchased from Sigma-Aldrich (St. Louis, MO). Wheat Germ Agglutinin (WGA) YSi SPA Scintillation Beads was purchased from GE healthcare life sciences (Piscataway, NJ).
The kinase reaction was conducted in polystyrene 384-well matrix white plate from Thermo Fisher Scientific in a final volume of 25 μΤ. Inhibitors were first diluted serially in DMSO and added to the plate wells before the addition of other reaction components. The final concentration of DMSO in the assay was 0.5%. The PI3K assays were carried out at room temperature in 20 mM MOPS, pH 6.7, 10 mM MgCl2, 5 mM DTT and CHAPS 0.03%. Reactions were initiated by the addition of ATP, the final reaction mixture consisted of 20 μΜ PIP2, 20 μΜ ATP, 0.2 μθί [γ-33Ρ] ATP, 4 nM PI3K5. Reactions were incubated for 210 min and terminated by the addition of 40 SPA beads suspended in quench buffer: 150mM potassium phosphate pH 8.0, 20% glycerol. 25 mM EDTA, 400 μΜ ATP. The final concentration of SPA beads was l .Omg/mL. After the plate sealing, plates were shaken overnight at room temperature and centrifuged at 1800 rpm for 10 minutes, the radioactivity of the product was determined by scintillation counting on Topcount (Perkin-Elmer). IC50 determination was performed by fitting the curve of percent control activity versus the the inhibitor concentration using the GraphPad Prism 3.0 software. Data for certain
Examples is provided below in Table 7.
Table 7
Figure imgf000059_0001
Example Bl : B cell proliferation assay
To acquire B cells, human PBMC are isolated from the peripheral blood of normal, drug free donors by standard density gradient centrifugation on Ficoll-Hypague (GE
Healthcare, Piscataway, NJ) and incubated with anti-CD 19 microbeads (Miltenyi Biotech, Auburn, CA). The B cells are then purified by positive immunosorting using an autoMacs (Miltenyi Biotech) according to the manufacture's instruction.
The purified B cells (2x l05/well/200 μΐ,) are cultured in 96-well ultra-low binding plates (Corning, Corning, NY) in RPMI1640, 10% FBS and goat F(ab')2 anti-human IgM (10 μg/ml) (Invitrogen, Carlsbad, CA) in the presence of different amount of test compounds for three days. [3H]-thymidine (1 μΰΛνβΙι) (PerkinElmer, Boston, MA) in PBS is then added to the B cell cultures for an additional 12 hours before the incorporated radioactivity is separated by filtration with water through GF B filters (Packard Bioscience, Meriden, CT) and measured by liquid scintillation counting with a TopCount (Packard Bioscience).
Example B2: Pfeiffer cell proliferation assay
Pfeiffer cell line (diffuse large B cell lymphoma) was purchased from ATCC
(Manassas, VA) and maintained in the culture medium recommended (RPMI and 10% FBS). To measure the anti-proliferation activity of the compounds, the Pfeiffer cells were plated with the culture medium (2x103 cells / well/ per 200 μΐ) into 96-well ultra-low binding plates (Corning, Corning, NY), in the presence or absence of a concentration range of test compounds. After 3-4 days, [3H] -thymidine (1 μ ΛνεΙΙ) (PerkinElmer, Boston, MA) in PBS wass then added to the cell culture for an additional 12 hours before the incorporated radioactivity was separated by filtration with water through GF/B filters (Packard Bioscience, Meridenj, CT) and measured by liquid scintillation counting with a TopCount (Packard Bioscience). Pfeiffer cell proliferation data for certain compounds described herein is provided below in Table 8.
Table 8
Figure imgf000060_0001
Example C: Akt phosphorylation assay
Ramos cells (B lymphocyte from Burkitts lymphoma) are obtained from ATCC (Manassas, VA) and maintained in RPMI1640 and 10% FBS. The cells (3x l07 cells /tube/3 mL in RPMI) are incubated with different amounts of test compounds for 2 hrs at 37 °C and then stimulated with goat F(ab')2 anti-human IgM (5 μg/mL) (Invitrogen) for 17 minutes in a 37 °C water bath. The stimulated cells are spun down at 4 °C with centrifugation and whole cell extracts are prepared using 300 \\L lysis buffer (Cell Signaling Technology, Danvers, MA). The resulting lysates are sonicated and supernatants are collected. The
phosphorylation level of Akt in the supernatants are analyzed by using PathScan phospho- Aktl (Ser473) sandwich ELISA kits (Cell Signaling Technology) according to the manufacturer's instruction. Various modifications of the invention, in addition to those described herein, will be apparent to those skilled in the art from the foregoing description. Such modifications are also intended to fall within the scope of the appended claims. Each reference, including all patent, patent applications, and publications, cited in the present application is incorporated herein by reference in its entirety.

Claims

What is claimed is:
1 . A compound of Formula I:
Figure imgf000062_0001
or a pharmaceutically acceptable salt thereof, wherein:
R1 is Ci_6 alkyl, C2-6 alkenyl, C2-6 alkynyl, or Ci-6 haloalkyl, wherein said Ci_6 alkyl, C2-e alkenyl, and C2-6 alkynyl are each optionally substituted by 1, 2, 3, or 4 groups independently selected from halo, OH, CN, NRlaRlb, C^ alkoxy, and C,_6 haloalkoxy;
R2 is H, halo, CN, Ci_6 alkyl, C 6 haloalkyl, -V-(C 6 alkyl), or -V-(d„6 haloalkyl), wherein said C1-6 alkyl in said Ci-6 alkyl and -ΐ ( ι-6 alkyl) is optionally substituted by 1, 2, 3, or 4 independently selected R2a groups;
R3 is Cy, -(CR2bR2c)a-L2-(CR2bR2c)b-Cy, halo, CN, N02, C,_6 alkyl, C2-6 alkenyl, C2.6 alkynyl, C,,6 haloalkyl, ORa, SRa, C(0)Rb, C(0)NRcRd, C(0)ORa, OC(0)Rb, OC(0)NRcRd, NR°Rd, NRCC(0)Rb, NRCC(0)ORb, NRCC(0)NRcRd, C(=NRe)Rb, C(=NRe)NRcRd,
NRCC(=NRe)NRcRd, NRcS(0)Rb, NRcS(0)2Rb, NRcS(0)2NRcRd, S(0)Rb, S(0)NRcRd, S(0)2Rb, or S(0)2NRcRd, wherein said Ci-6 alkyl, C2.6 alkenyl, and C2.6 alkynyl are each optionally substituted by 1 , 2, 3, or 4 independently selected R3a groups;
R4 is H, halo, OH, CN, C1-4 alkyl, C 1-4 haloalkyl, C^ alkoxy, or C1-4 haloalkoxy;
R5 is halo, OH, CN, C1 -4 alkyl, C^ alkoxy, or C1-4 haloalkoxy;
R6 is H, halo, OH, CN, C alkyl, C haloalkyl, C1-4 alkoxy, or C 1 haloalkoxy;
R7 is F, CN, d-3 haloalkyl, CONH2, CM alkyl-NHC(=0)- or (Ci_4 alkyl)2NC(=0)-;
R8 is H, CM alkyl, CM haloalkyl, or C(0)R4a;
L1 is O, NRB, S, S(O), S(0)2, C(0), C(0)NRB, S(0)NRB, or S(0)2NRB;
L2 is (CRcRD)n, O, NRB, S, S(0), S(0)2, C(O), C(0)0, C(0)NRB, S(0)NRB, S(0)2NRB, OC(0)NRB, NRBC(0)NRB, C(=NRe), C(=NRe)NRB, or NRBC(=NRe)NRB;
RA, RB, Rc, and RD are each independently selected from H, Ci_6 alkyl, and Q-6 haloalkyl; Cy is selected from C3-7 cycloalkyl, 4-11 membered heterocycloalkyl, 6-10 membered aryl and 5-10 membered heteroaryl, each of which is substituted with 0, 1, 2, 3, or 4 independently selected R3a groups;
each Rl a and Rlb is independently selected from H and Ci-6 alkyl;
or any R, a and Rlb together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group, which is optionally substituted with 1, 2, 3, or 4 substituents independently selected from C1-6 alkyl;
each R2 is independently selected from OH, N02, CN, halo, Ci_6 alkyl, C2-6 alkenyl, C2_ 6 alkynyl, C3.7 cycloalkyl, Ci_6 haloalkyl, Ci-6 alkoxy, Ci_6 haloalkoxy, amino, Ci^ alkylamino, di(Ci_6 alkyl)amino, thio, Ci^ alkylthio, C]_6 alkylsulfinyl, Ci_6 alkylsulfonyl, carbamyl, Ci-6 alkylcarbamyl, di(Ci.6 alkyl)carbamyl, carboxy, Ci^ alkylcarbonyl, Ci^ alkoxycarbonyl, Ci-6 alkylcarbonylamino, Ci_6 alkylsulfonylamino, aminosulfonyl, C1-6 alkylaminosulfonyl, di(Ci_6 alkyl)aminosulfonyl, aminosulfonylamino, Ci.6 alkylaminosulfonylamino, di(C1-6
alkyl)aminosulfonylamino, aminocarbonylamino, Ci_6 alkylaminocarbonylamino, and di(C] _6 alkyl)aminocarbonylamino;
each R2b and R2c is independently selected from H, Ci-6 alkyl, and Q_6 haloalkyl;
each R3a is independently selected from halo, CN, N02, Ci-6 alkyl, C2-6 alkenyl, C2_6 alkynyl, C\.6 haloalkyl, C3-7 cycloalkyl, 4-1 1 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, (C3_7 cycloalkyl)-Ci_4 alkyl, (4-11 membered
heterocycloalkyl)-Ci_4 alkyl, (6-10 membered aryl)-Ci_4 alkyl, (5-10 membered heteroaryl)- Ci_4 alkyl, ORal, SRal, C(0)Rbl, C(0)NRclRdl, C(0)OR l, OC(0)Rbl, OC(0)NRclRdl, NRclRdl, NRclC(0)Rbl, NRclC(0)ORbl, NRclC(0)NRclRdl, C(=NRe)Rbl, C(=NRe)NRclRdl, NRclC(=NRe)NRclRdl, NRclS(0)Rbl, NRclS(0)2Rbl, NRclS(0)2NRclRdl, S(0)Rbl,
S(0)NRclRdl, S(0)2Rbl, and S(0)2NRclRdl; wherein said Ci-6 alkyl, C2-6 alkenyl, C2.6 alkynyl, C37 cycloalkyl, 4-11 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, (C3_7 cycloalkyl)-C 1-4 alkyl, (4-1 1 membered heterocycloalkyl)-C 1 -4 alkyl, (6-10 membered aryl)-C 1.4 alkyl, and (5-10 membered heteroaryl)-Ci_4 alkyl are each optionally substituted by 1, 2, 3, or 4 groups independently selected from ORal, SRal, C(0)Rbl, C(0)NRclRdl, C(0)ORal, OC(0)Rbl, OC(0)NRclRdl, NRclRdl, NRclC(0)Rbl, NRclC(0)ORbl, NRclC(0)NRc,Rdl, C(=NRe)Rbl, C(=NRe)NRclRdl, NRclC(=NRe)NRclRdl, NRcl S(0)Rbl, NRclS(0)2Rbl, NRcl S(0)2NRc,Rdl, S(0)Rbl, S(0)NRclRdl, S(0)2Rbl, and S(0)2NRclRdl ; R is C]_6 alkyl, C3-7 cycloalkyl, 4-1 1 membered heterocycloalkyl, 6-10 membered aryl, 5- 10 membered heteroaryl, (C3-7 cycloalkyl)-Ci_4 alkyl, (4-1 1 membered heterocycloalkyl)-C i- 4 alkyl, (6-10 membered aryl)-Ci_4 alkyl, and (5-10 membered heteroaryl)-Ci_4 alkyl;
each Ra, Rb, Rc, and Rd is independently selected from H, C1-6 alkyl, Ci_6 haloalkyl, C2-6 alkenyl, and C2-6 alkynyl; wherein said C .e alkyl, C2-6 alkenyl, and C2-6 alkynyl are each optionally substituted with 1 , 2, 3, 4, or 5 substituents independently selected from OH, N02, CN, halo, Ci-6 alkyl, C2-6 alkenyl, C2_6 alkynyl, Ci_6 haloalkyl, cyano-Q-6 alkyl, HO-C]_6 alkyl, Ci_4 alkoxy-Ci-6 alkyl, C3.7 cycloalkyl, Ci-6 haloalkyl, C1-6 alkoxy, Ci-6 haloalkoxy, amino, Ci-6 alkylamino, di(Ci_6 alkyl)amino, thio, Ci-6 alkylthio, Ci.6 alkylsulfmyl, C e alkylsulfonyl, carbamyl, Ci-6 alkylcarbamyl, di(C1-6 alkyl)carbamyl, carboxy, Ci-6 alkylcarbonyl, Ci-6 alkoxycarbonyl, Ci_6 lkylcarbonylamino, Ci^ alkylsulfonylamino, aminosulfonyl, Ci-6 alkylaminosulfonyl, di(C1 -6 alkyl)aminosulfonyl, aminosulfonylamino, Ci_6 alkylaminosulfonylamino, di(Ci.6 alkyl)aminosulfonylamino, aminocarbonylamino, Ci_6 alkylaminocarbonylamino, and di(Ci-6 alkyl)aminocarbonylamino;
or Rc and Rd together with the N atom to which they are attached form a 4-, 5-, 6-, or 7- membered heterocycloalkyl group, which is optionally substituted with 1 , 2, 3, or 4 substituents independently selected from OH, N02, CN, halo, Ci-6 alkyl, C2-6 alkenyl, C2-e alkynyl, C1-6 haloalkyl, cyano-Ci-6 alkyl, HO-Ci-6 alkyl, Ci-4 alkoxy-Ci.6 alkyl, C3-7 cycloalkyl, C1-6 haloalkyl, Ci-6 alkoxy, Ci_6 haloalkoxy, amino, Ci_6 alkylamino, di(Ci_6 alkyl)amino, thio, Q-6 alkylthio, Cj.6 alkylsulfinyl, Ci -6 alkylsulfonyl, carbamyl, Ci-6 alkylcarbamyl, di(Ci-6 alkyl)carbamyl, carboxy, Ci - alkylcarbonyl, Ci_6 alkoxycarbonyl, C1-6 alkylcarbonylamino, Ci_6 alkylsulfonylamino, aminosulfonyl, Ci_6 alkylaminosulfonyl, di(Ci_6 alkyl)aminosulfonyl, aminosulfonylamino, Ci -6 alkylaminosulfonylamino, di(Ci_6 alkylaminosulfonylamino, aminocarbonylamino, C1-6 alkylaminocarbonylamino, and di(Ci-6 alkyl)aminocarbonylamino;
each Re is independently selected from H, Ci_4 alkyl, CN, OH, CM alkoxy, Ci-4 alkylsulfonyl, carbamyl, Ci-4 alkylcarbamyl, di(C1-4 alkyl)carbamyl, and Ci_4 alkylcarbonyl; each R l, Rbl, Rcl , and Rdl is independently selected from H, C]_6 alkyl, Ci_6 haloalkyl, C2-6 alkenyl, C2-6 alkynyl, C3.7 cycloalkyl, 4- 1 1 membered heterocycloalkyl, 6- 10 membered aryl, 5-10 membered heteroaryl, (C3_7 cycloalkyl)-Ci.4 alkyl, (4-1 1 membered
heterocycloalkyl)-Ci,4 alkyl, (6-10 membered aryl)-Ci-4 alkyl, and (5-10 membered heteroaryl)-Ci_4 alkyl, wherein said C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3.7 cycloalkyl, 4-1 1 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, (C3_7 cycloalkyl)-Ci-4 alkyl, (4-1 1 membered heterocycloalkyl)-Ci.4 alkyl, (6-10 membered aryl)- Ci-4 alkyl, and (5- 10 membered heteroaryl)-Ci-4 alkyl are each optionally substituted with 1 , 2, 3, 4, or 5 substituents independently selected from OH, N02, CN, halo, C1-6 alkyl, C2-e alkenyl, C2_6 alkynyl, Ci-6 haloalkyl, cyano-Q.6 alkyl, HO-C1-6 alkyl, Ci_4 alkoxy-Ci_6 alkyl, C3.7 cycloalkyl, Ci-6 haloalkyl, C1-6 alkoxy, Ci_6 haloalkoxy, amino, Ci-6 alkylamino, di(C1-6 alkyl)amino, thio, Ci-6 alkylthio, Ci_6 alkylsulfinyl, Ci^ alkylsulfonyl, carbamyl, Ci-6 alkylcarbamyl, di(Ci_6 alkyl)carbamyl, carboxy, Ci-6 alkylcarbonyl, Ci-6 alkoxycarbonyl, Ci_6 alkylcarbonylamino, Ci.6 alkylsulfonylamino, aminosulfonyl, Ci_6 alkylaminosulfonyl, di(C1-6 alkyl)aminosulfonyl, aminosulfonylamino, Ci_6 alkylaminosulfonylamino, di(C1-6 alkyl)aminosulfonylamino, aminocarbonylamino, Ci_6 alkylaminocarbonylamino, and di(C1-6 alkyl)aminocarbonylamino;
or Rcl and Rdl together with the N atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group, which is optionally substituted with 1 , 2, 3, or 4 substituents independently selected from OH, N02, CN, halo, C1-6 alkyl, C2.6 alkenyl, C2-6 alkynyl, Ci_6 haloalkyl, cyano-Ci-6 alkyl, HO-C1-6 alkyl, Ci-4 alkoxy-Ci-6 alkyl, C3-7 cycloalkyl, Ci_6 haloalkyl, Ci-6 alkoxy, C1-6 haloalkoxy, amino, C1-6 alkylamino, di(Ci_6 alkyl)amino, thio, Ci-6 alkylthio, Ci_6 alkylsulfmyl, Ci_6 alkylsulfonyl, carbamyl, Ci-6 alkylcarbamyl, di(Ci-6 alkyl)carbamyl, carboxy, Ci-6 alkylcarbonyl, Ci-6 alkoxycarbonyl, Ci-6 alkylcarbonylamino, Ci-6 alky lsulfony lam ino, aminosulfonyl, Ci-6 alkylaminosulfonyl, di(Ci _6 alkyl)aminosulfonyl, aminosulfonylamino, Ci.6 alkylaminosulfonylamino, di(C1-6 alkyl)aminosulfonylamino, aminocarbonylamino, C].6 alkylaminocarbonylamino, and di(Ci-6 alkyl)aminocarbonylamino;
a is 0, 1, 2, 3, or 4;
b is 0, 1, 2, 3, or 4; and
n is 1 , 2, 3, 4, 5, or 6.
2. The compound of claim 1 , or a pharmaceutically acceptable salt thereof, wherein R1 is Ci-6 alkyl.
3. The compound of claim 1 , or a pharmaceutically acceptable salt thereof, wherein R1 is methyl.
4. The compound of any one of claims 1 to 3, or a pharmaceutically acceptable salt thereof, wherein R2 is - -(C\,e alkyl).
5. The compound of any one of claims 1 to 3, or a pharmaceutically acceptable salt thereof, wherein R2 is C1-6 alkoxy.
6. The compound of any one of claims 1 to 3, or a pharmaceutically acceptable salt thereof, wherein R2 is methoxy.
7. The compound of any one of claims 1 to 6, or a pharmaceutically acceptable salt thereof, wherein R3 is Cy or -(CR2bR2c)a-L2-(CR2bR2c)b-Cy.
8. The compound of any one of claims 1 to 6, or a pharmaceutically acceptable salt thereof, wherein R3 is Cy,
9. The compound of any one of claims 1 to 8, or a pharmaceutically acceptable salt thereof, wherein Cy is selected from 6-10 membered aryl and 5-10 membered heteroaryl, each of which is substituted with 0, 1, 2, 3, or 4 independently selected R3a groups.
10. The compound of any one of claims 1 to 8, or a pharmaceutically acceptable salt thereof, wherein Cy is phenyl substituted with 0, 1, 2, 3, or 4 independently selected R3a groups.
11. The compound of any one of claims 1 to 8, or a pharmaceutically acceptable salt thereof, wherein Cy is 5-6 membered heteroaryl substituted with 0, 1, 2, 3, or 4 independently selected R3a groups.
12. The compound of any one of claims 1 to 8, or a pharmaceutically acceptable salt thereof, wherein Cy is pyridyl or pyrimidinyl substituted with 0, 1, 2, 3, or 4 independently selected R3a groups.
13. The compound of any one of claims 1 to 12, or a pharmaceutically acceptable salt thereof, wherein Cy is substituted with 1, 2, or 3 independently selected R3a groups.
14. The compound of any one of claims 1 to 13, or a pharmaceutically acceptable salt thereof, wherein R3a is independently selected from halo, CN, N02, C2-6 alkenyl, C2-6 alkynyl, Ci-6 haloalkyl, C3-7 cycloalkyl, 4-1 1 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, (C3.7 cycloalkyl)-Ci_4 alkyl, (4-1 1 membered heterocycloalkyl)-Ci-4 alkyl, (6-10 membered aryl)-C alkyl, (5-10 membered heteroaryl)-C alkyl, ORal, SRal, C(0)NRclRdl, OC(0)Rbl, OC(0)NRclRdl, NRclC(0)Rbl, NRclC(0)ORbl, NRc1C(0)NRclRdl, C(=NRe)Rbl, C(=NRe)NRclRdl, NRclC(=NRe)NRclRdl, NRclS(0)Rbl, NRcl S(0)2Rbl, NRclS(0)2NRclRdl, S(0)Rbl, S(0)NRclRdl, S(0)2Rbl, and S(0)2NRc,Rdl ; wherein said C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, 4-1 1 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, (C3-7 cycloalkyl)-C i-4 alkyl, (4-11 membered heterocycloalkyl)-Ci_ 4 alkyl, (6-10 membered aryl)-Ci-4 alkyl, and (5-10 membered heteroaryl)-Ci-4 alkyl are each optionally substituted by 1, 2, 3, or 4 groups independently selected from ORal, SRal, C(0)Rbl, C(0)NRclRdl, C(0)ORal, OC(0)Rbl , OC(0)NRclRdl, NRclRdl, NRclC(0)Rbl, NRclC(0)ORbl, NRclC(0)NRclRdl, C(=NRe)Rbl, C(=NRe)NRclRdl, NRclC(=NRe)NRclRdl, NRcl S(0)Rbl, NRcl S(0)2Rbl, NRcl S(0)2NRclRdl, S(0)Rbl, S(0)NRclRdl, S(0)2Rbl, and S(0)2NRclRdl .
15. The compound of any one of claims 1 to 13, or a pharmaceutically acceptable salt thereof, wherein R3a is independently selected from N02, C2-6 alkenyl, C2_6 alkynyl, Ci-6 haloalkyl, C3-7 cycloalkyl, 4-11 membered heterocycloalkyl, 6-10 membered aryl, 5-10 membered heteroaryl, (C3-7 cycloalkyl)-Ci_4 alkyl, (4-1 1 membered heterocycloalkyl)-Ci_4 alkyl, (6-10 membered aryl)-Ci_4 alkyl, and (5-10 membered heteroaryl)-Ci_4 alkyl, SRal, C(0)Rbl, C(0)NRclRdl, OC(0)Rb1, OC(0)NRclRdl, NRclC(0)Rbl, NRclC(0)ORbl,
NRclC(0)NRclRdl, C(=NRe)Rbl, C(=NRe)NRclRdl, NRclC(=NRe) RclRdl, NRcl S(0)Rbl, NRclS(0)2Rbl, NRcl S(0)2NRclRdl, S(0)Rbl, S(0)NRclRdl, S(0)2Rbl, and S(0)2 RclRdl; wherein said C2_6 alkenyl, C2_6 alkynyl, C3.7 cycloalkyl, 4-11 membered heterocycloalkyl, 6- 10 membered aryl, 5-10 membered heteroaryl, (C3.7 cycloalkyl)-C] -4 alkyl, (4-1 1 membered heterocycIoaIkyl)-C 1.4 alkyl, (6-10 membered aryl)-C 1.4 alkyl, and (5-10 membered heteroaryl)-Ci_4 alkyl are each optionally substituted by 1, 2, 3, or 4 groups independently selected from ORal, SRal, C(0)Rbl, C(0)NRclRdl, C(0)ORal, OC(0)Rbl, OC(0)NRc1Rdl, NRclRdl, NRclC(0)Rbl, NRclC(0)ORbl, NRclC(0)NRclRdl, C(=NRe)Rbl, C(=NRe)NRclRdl, NRclC(=NRe)NRc,Rdl, NRclS(0)Rbl, NRclS(0)2Rbl, NRclS(0)2NRclRdl, S(0)Rbl,
S(0)NRclRdl, S(0)2Rbl , and S(0)2NRclRdl .
16. The compound of any one of claims 1 to 13, or a pharmaceutically acceptable salt thereof, wherein each R3a is independently selected from halo, CN, C(0)NRclRdl, NRclRdl, and S(0)2Rbl.
17. The compound of any one of claims 1 to 13, or a pharmaceutically acceptable salt thereof, wherein each R3a is independently selected from halo, CN, C(0)NRclRdl, and
S(0)2R'
18. The compound of any one of claims 1 to 17, or a pharmaceutically acceptable salt thereof, wherein R4 is halo, CN, Ci-4 alkyl, or Ci^ haloalk l.
19. The compound of any one of claims 1 to 17, or a pharmaceutically acceptable salt thereof, wherein R4 is halo.
20. The compound of any one of claims 1 to 19, or a pharmaceutically acceptable salt thereof, wherein R5 is CI, F, or methyl.
21. The compound of any one of claims 1 to 19, or a pharmaceutically acceptable salt thereof, wherein R5 is CI.
22. The compound of any one of claims 1 to 21, or a pharmaceutically acceptable salt thereof, wherein R6 is H.
23. The compound of any one of claims 1 to 22, or a pharmaceutically acceptable salt thereof, wherein R7 is CN.
24. The compound of any one of claims 1 to 23, or a pharmaceutically acceptable salt thereof, wherein R8 is H.
25. The compound of any one of claims 1 and 4 to 21, having Formula II:
Figure imgf000068_0001
II or a pharmaceutically acceptable salt thereof.
26. The compound of any one of claims 1 and 7 to 19, having Formula III:
Figure imgf000069_0001
III
or a pharmaceutically acceptable salt thereof.
27. A compound of claim 1 selected from:
4-amino-6-[(l -{5-chloro-2-methoxy-4-methyl-3-[5-(methylsulfonyl)pyridin-3- yl]phenyl}ethyl)amino]pyrimidine-5-carbonitrile (1);
4-amino-6-( { 1 -[5 -chloro-3 -(5 -cyanopyridin-3 -y l)-2-methoxy-4- methylphenyl]ethyl}amino)-pyrimidine-5-carbonitrile (2);
4-amino-6-( { 1 - [3 -(2-aminopyrimidin-5 -yl)-5 -chloro-2-methoxy-4- methylphenyl]ethyl}-amino)pyrimidine-5-carbonitrile (3);
4-amino-6-({ l-[5-chloro-2-methoxy-6-methyl-4'-(methylsulfonyl)biphenyl-3- yl]ethyl } amino)pyrimidine-5 -carbonitrile (4) ;
4-amino-6-[(l-{5-chloro-2-methoxy-4-methyl-3-[5-(morpholin-4-ylcarbonyl)pyridin- 3-yl]phenyl}ethyl)-amino]pyrimidine-5-carbonitrile (5);
4-amino-6-({ l-[5-chloro-3-(5-cyanopyridin-3-yl)-4-fluoro-2-methoxyphenyl]ethyl}- amino)pyrimidine-5 -carbonitrile (6) ;
4-amino-6-({ l-[5-chloro-4-fluoro-3-(5-fluoropyridin-3-yl)-2-methoxyphenyl]ethyl}- amino)pyrimidine-5-carbonitrile (7);
4-amino-6-[( 1 - {5-chloro-4-fluoro-2-methoxy-3 -[5 -(methylsulfonyl)pyridin-3 - yl]phenyl}ethyl)amino]-pyrimidine-5 -carbonitrile (8);
4-amino-6-({ l-[5-chloro-6-fluoro-2-methoxy-4'-(methylsulfonyl)biphenyl-3- yl]ethyl} amino) pyrimidine-5-carbonitrile (9);
4-amino-6-({ l-[3-(2-aminopyrimidin-5-yl)-5-chloro-4-fluoro-2-methoxyphenyl]- ethyl}amino)pyrimidine-5-carbonitrile (10); 4-amino-6- [( 1 - {4, 5-dichloro-2-methoxy-3 -[5 -(methy lsulfonyl)pyridin-3 - yl]phenyl}ethyl)amino]pyrimidine-5-carbonitrile (1 1);
4-amino-6-( { 1 -[4, 5 -dichloro-3 -(5 -fluoropyridin-3 -yl)-2- methoxypheny l]ethy 1 } amino)pyrimidine-5 -carbonitrile (12);
4-amino-6-({ l -[5,6-dichloro-2-methoxy-4'-(methylsulfonyl)biphenyl-3- yl]ethyl}amino)pyrimidine-5-carbonitrile (13); and
4-amino-6-({ l-[3-(2-aminopyrimidin-5-yl)-4,5-dichloro-2- methoxypheny 1] ethyl } amino)pyrimidine- 5 -carbonitrile (14);
or a pharmaceutically acceptable salt thereof.
28. A pharmaceutical composition comprising a compound of any one of claims 1 to 27, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier.
29. A method of modulating an activity of a PI3K kinase, comprising contacting the kinase with a compound of any one of claims 1 to 27, or a pharmaceutically acceptable salt thereof.
30. The method of claim 29, wherein the PI3K is selected from the group consisting of ΡΒΚα, ΡΒΚβ, ΡΙ3Κγ, and ΡΙ3Κδ.
31. The method of claim 29, wherein the PI3K comprises a mutation.
32. The method of any one of claims 29 to 31 , wherein said modulating is inhibiting.
33. The method of any one of claims 29 to 31, wherein said compound is a selective inhibitor for PI3K5 over one or more of ΡΙ3Κα, ΡΒΚβ, and ΡΒΚγ.
34. A method of treating a disease in a patient, wherein said disease is associated with abnormal expression or activity of a PBK kinase, comprising administering to said patient a therapeutically effective amount of a compound of any one of claims 1 to 27, or a pharmaceutically acceptable salt thereof.
35. The method of claim 34, wherein said disease is osteoarthritis, restenosis, atherosclerosis, bone disorders, arthritis, diabetic retinopathy, psoriasis, benign prostatic hypertrophy, inflammation, angiogenesis, pancreatitis, kidney disease, inflammatory bowel disease, myasthenia gravis, multiple sclerosis, or Sjogren's syndrome.
36. The method of any one of claims 34 to 35, wherein more than one of said compounds is administered.
37. The method of claim 34, wherein the compound is administered in combination with a kinase inhibitor that inhibits a kinase other than a PI3K kinase.
38. A method of treating an immune-based disease in a patient, comprising administering to said patient a therapeutically effective amount of a compound of any one of claims 1 to 27, or a pharmaceutically acceptable salt thereof.
39. The method of claim 38, wherein said immune-based disease is rheumatoid arthritis, allergy, asthma, glomerulonephritis, lupus, or inflammation related to any of the
aforementioned.
40. A method of treating a cancer in a patient, comprising administering to said patient a therapeutically effective amount of a compound of any one of claims 1 to 27, or a pharmaceutically acceptable salt thereof.
41. The method of claim 40, wherein said cancer is breast, prostate, colon, endometrial, brain, bladder, skin, uterus, ovary, lung, pancreatic, renal, gastric, or a hematological cancer.
42. The method of claim 41 wherein said hematological cancer is acute myeloblastic leukemia, chronic myeloid leukemia, or B cell lymphoma.
43. A method of treating a lung disease in a patient, comprising administering to said patient a therapeutically effective amount of a compound of any one of claims 1 to 27, or a pharmaceutically acceptable salt thereof.
44. The method of claim 43, wherein said lung disease is acute lung injury (ALI) or adult respiratory distress syndrome (ARDS).
PCT/US2012/030310 2011-03-25 2012-03-23 Pyrimidine-4,6-diamine derivatives as pi3k inhibitors WO2012135009A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/007,061 US9126948B2 (en) 2011-03-25 2012-03-23 Pyrimidine-4,6-diamine derivatives as PI3K inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161467787P 2011-03-25 2011-03-25
US61/467,787 2011-03-25

Publications (1)

Publication Number Publication Date
WO2012135009A1 true WO2012135009A1 (en) 2012-10-04

Family

ID=45922850

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/030310 WO2012135009A1 (en) 2011-03-25 2012-03-23 Pyrimidine-4,6-diamine derivatives as pi3k inhibitors

Country Status (2)

Country Link
US (1) US9126948B2 (en)
WO (1) WO2012135009A1 (en)

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013088404A1 (en) 2011-12-15 2013-06-20 Novartis Ag Use of inhibitors of the activity or function of PI3K
US8680108B2 (en) 2009-12-18 2014-03-25 Incyte Corporation Substituted fused aryl and heteroaryl derivatives as PI3K inhibitors
US8940752B2 (en) 2009-06-29 2015-01-27 Incyte Corporation Pyrimidinones as PI3K inhibitors
US9018221B2 (en) 2012-12-21 2015-04-28 Gilead Calistoga, Llc Phosphatidylinositol 3-kinase inhibitors
US9029384B2 (en) 2012-12-21 2015-05-12 Gilead Calistoga, LLC. Phosphatidylinositol 3-kinase inhibitors
US9062055B2 (en) 2010-06-21 2015-06-23 Incyte Corporation Fused pyrrole derivatives as PI3K inhibitors
US9096600B2 (en) 2010-12-20 2015-08-04 Incyte Corporation N-(1-(substituted-phenyl)ethyl)-9H-purin-6-amines as PI3K inhibitors
US9108984B2 (en) 2011-03-14 2015-08-18 Incyte Corporation Substituted diamino-pyrimidine and diamino-pyridine derivatives as PI3K inhibitors
US9193721B2 (en) 2010-04-14 2015-11-24 Incyte Holdings Corporation Fused derivatives as PI3Kδ inhibitors
US9199982B2 (en) 2011-09-02 2015-12-01 Incyte Holdings Corporation Heterocyclylamines as PI3K inhibitors
WO2015191677A1 (en) * 2014-06-11 2015-12-17 Incyte Corporation Bicyclic heteroarylaminoalkyl phenyl derivatives as pi3k inhibitors
US9221795B2 (en) 2013-06-14 2015-12-29 Gilead Sciences, Inc. Phosphatidylinositol 3-kinase inhibitors
US9309251B2 (en) 2012-04-02 2016-04-12 Incyte Holdings Corporation Bicyclic azaheterocyclobenzylamines as PI3K inhibitors
US9403847B2 (en) 2009-12-18 2016-08-02 Incyte Holdings Corporation Substituted heteroaryl fused derivatives as P13K inhibitors
WO2016196244A1 (en) 2015-05-29 2016-12-08 Incyte Corporation Pyridineamine compounds useful as pim kinase inhibitors
US9550765B2 (en) 2013-01-15 2017-01-24 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
WO2017044730A1 (en) 2015-09-09 2017-03-16 Incyte Corporation Salts of a pim kinase inhibitor
WO2017059251A1 (en) 2015-10-02 2017-04-06 Incyte Corporation Heterocyclic compounds useful as pim kinase inhibitors
US9676750B2 (en) 2013-01-14 2017-06-13 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as pim kinase inhibitors
US9732097B2 (en) 2015-05-11 2017-08-15 Incyte Corporation Process for the synthesis of a phosphoinositide 3-kinase inhibitor
US9822124B2 (en) 2014-07-14 2017-11-21 Incyte Corporation Bicyclic heteroaromatic carboxamide compounds useful as Pim kinase inhibitors
US9890162B2 (en) 2014-07-14 2018-02-13 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as pim kinase inhibitors
US9988401B2 (en) 2015-05-11 2018-06-05 Incyte Corporation Crystalline forms of a PI3K inhibitor
US10000507B2 (en) 2013-08-23 2018-06-19 Incyte Corporation Furo- and thieno-pyridine carboxamide compounds useful as pim kinase inhibitors
US10336759B2 (en) 2015-02-27 2019-07-02 Incyte Corporation Salts and processes of preparing a PI3K inhibitor
US10596161B2 (en) 2017-12-08 2020-03-24 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI657090B (en) 2013-03-01 2019-04-21 英塞特控股公司 USE OF PYRAZOLOPYRIMIDINE DERIVATIVES FOR THE TREATMENT OF PI3Kδ RELATED DISORDERS
KR20220066179A (en) 2014-04-08 2022-05-23 인사이트 코포레이션 Treatment of b-cell malignancies by a combination jak and pi3k inhibitor
WO2016130501A1 (en) 2015-02-09 2016-08-18 Incyte Corporation Aza-heteroaryl compounds as pi3k-gamma inhibitors
TWI744256B (en) 2015-11-06 2021-11-01 美商英塞特公司 HETEROCYCLIC COMPOUNDS AS PI3K-γ INHIBITORS
ES2833955T3 (en) 2016-01-05 2021-06-16 Incyte Corp Pyridines substituted with pyrazole / imidazole as PI3K-Gamma inhibitors
CN109311901A (en) * 2016-04-07 2019-02-05 达纳-法伯癌症研究所有限公司 The method for the lesion that pyrimido-diazepine ketone kinases framework compound and treatment PI3K are mediated
WO2017223414A1 (en) 2016-06-24 2017-12-28 Incyte Corporation HETEROCYCLIC COMPOUNDS AS PI3K-γ INHIBITORS
SG11202003428VA (en) 2017-10-18 2020-05-28 Incyte Corp Condensed imidazole derivatives substituted by tertiary hydroxy groups as pi3k-gamma inhibitors
CR20210165A (en) 2018-09-05 2021-10-01 Incyte Corp Crystalline forms of a phosphoinositide 3-kinase (pi3k) inhibitor

Citations (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5521184A (en) 1992-04-03 1996-05-28 Ciba-Geigy Corporation Pyrimidine derivatives and processes for the preparation thereof
WO2000009495A1 (en) 1998-08-11 2000-02-24 Novartis Ag Isoquinoline derivatives with angiogenesis inhibiting activity
WO2000053595A1 (en) 1999-03-06 2000-09-14 Astrazeneca Ab Pyrimidine compounds
WO2001014402A1 (en) 1999-08-19 2001-03-01 Isis Pharmaceuticals, Inc. Antisense modulation of focal adhesion kinase expression
WO2001064655A1 (en) 2000-03-01 2001-09-07 Astrazeneca Ab 2, 4-di(hetero-)arylamino (-oxy)-5-substituted pyrimidines as antineoplastic agents
WO2002000196A2 (en) 2000-06-28 2002-01-03 Smithkline Beecham P.L.C. Wet milling process
WO2003024967A2 (en) 2001-09-19 2003-03-27 Aventis Pharma S.A. Indolizines as kinase protein inhibitors
WO2003037347A1 (en) 2001-10-30 2003-05-08 Novartis Ag Staurosporine derivatives as inhibitors of flt3 receptor tyrosine kinase activity
WO2003099771A2 (en) 2002-05-29 2003-12-04 Novartis Ag Diaryl urea derivatives useful for the treatment of protein kinase dependent diseases
WO2004005281A1 (en) 2002-07-05 2004-01-15 Novartis Ag Inhibitors of tyrosine kinases
WO2004046120A2 (en) 2002-11-15 2004-06-03 Vertex Pharmaceuticals Incorporated Diaminotriazoles useful as inhibitors of protein kinases
WO2004048365A1 (en) * 2002-11-21 2004-06-10 Chiron Corporation 2,4,6-trisubstituted pyrimidines as phosphotidylinositol (pi) 3-kinase inhibitors and their use in the treatment of cancer
WO2004056786A2 (en) 2002-12-20 2004-07-08 Pfizer Products Inc. Pyrimidine derivates for the treatment of abnormal cell growth
WO2004080980A1 (en) 2003-03-14 2004-09-23 Novartis Ag 2, 4- di (phenylamino) pyrimidines useful in the treatment of neoplastic diseases, inflammatory and immune system disorders
WO2005028444A1 (en) 2003-09-24 2005-03-31 Novartis Ag 1,4-disubstituted isoquinilone derivatives as raf-kinase inhibitors useful for the treatment of proliferative diseases
WO2006056399A2 (en) 2004-11-24 2006-06-01 Novartis Ag Combinations of jak inhibitors and at least one of bcr-abl, flt-3, fak or raf kinase inhibitors
WO2007042806A1 (en) * 2005-10-11 2007-04-19 Ludwig Institute For Cancer Research Pyrimidine derivatives for the treatment of cancer
WO2008032033A1 (en) * 2006-09-14 2008-03-20 Astrazeneca Ab 4-benzimidazolyl-2-morpholino-6-piperazinylpyrimidine derivatives as pi3k and mtor inhibitors for the treatment of proliferative disorders
WO2011025889A1 (en) 2009-08-28 2011-03-03 Takeda Pharmaceutical Company Limited HEXAHYDROOXAZINOPTERINE COMPOUNDS FOR USE AS mTOR INHIBITORS
WO2011092198A1 (en) * 2010-01-26 2011-08-04 Boehringer Ingelheim International Gmbh 5-alkynyl-pyrimidines

Family Cites Families (229)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3169967A (en) 1957-11-14 1965-02-16 Ciba Geigy Corp Methyl o-lower alkanoyl-reserpates
US3037980A (en) 1955-08-18 1962-06-05 Burroughs Wellcome Co Pyrrolopyrimidine vasodilators and method of making them
DE1770420U (en) 1958-02-27 1958-07-17 Tara Union G M B H FLOWER POT MADE OF PLASTIC.
US3506643A (en) 1966-12-09 1970-04-14 Max Thiel N**6-aralkyl-adenosine derivatives
DE2139107A1 (en) 1971-08-04 1973-02-15 Merck Patent Gmbh N(6)-subst adenosine derivs - with coronary circulatory etc activity
US3814251A (en) 1972-08-09 1974-06-04 Sperry Rand Corp Power transmission
US3962443A (en) 1972-08-14 1976-06-08 Dainippon Pharmaceutical Co., Ltd. Antibacterial pharmaceutical compositions and processes for preparation thereof
DE2248232A1 (en) 1972-10-02 1974-04-11 Basf Ag 4-THIOPYRIMIDO SQUARE BRACKET ON 4.5 D SQUARE BRACKET FOR PYRIMIDINE
US3862189A (en) 1973-08-14 1975-01-21 Warner Lambert Co Aralkyl-substituted purines and pyrimidines as antianginal bronchodilator agents
US3936454A (en) 1973-08-14 1976-02-03 Warner-Lambert Company 5-Amino-4-chloro-6-(substituted amino)-pyrimidines
DK3375A (en) 1974-01-25 1975-09-15 Ciba Geigy Ag
JPS50111080U (en) 1974-02-21 1975-09-10
JPS5359663U (en) 1976-10-25 1978-05-20
JPS52106897A (en) 1977-01-10 1977-09-07 Dainippon Pharmaceut Co Ltd Synthesis of piperazine derivatives
JPS56123981U (en) 1980-02-20 1981-09-21
JPS5883698U (en) 1981-11-27 1983-06-06 石川島播磨重工業株式会社 Heat exchanger
JPS58162949A (en) 1982-03-20 1983-09-27 Konishiroku Photo Ind Co Ltd Color photographic sensitive silver halide material
JPS6190153A (en) 1984-10-09 1986-05-08 Fuji Photo Film Co Ltd Treatment of silver halide photosensitive material
JPS62103640U (en) 1985-12-18 1987-07-02
JPH07119970B2 (en) 1986-04-18 1995-12-20 富士写真フイルム株式会社 Image forming method
JPS6310746A (en) 1986-07-01 1988-01-18 Tanabe Seiyaku Co Ltd Naphthalene derivative
CA1324609C (en) 1986-07-30 1993-11-23 Eastman Kodak Company Photographic element and process
US4861701A (en) 1987-10-05 1989-08-29 Eastman Kodak Company Photographic element and process comprising a compound which comprises two timing groups in sequence
AT388372B (en) 1987-10-08 1989-06-12 Tanabe Seiyaku Co Novel naphthalene derivatives and pharmaceuticals containing them
EP0364598A4 (en) 1988-03-02 1992-01-15 Yoshitomi Pharmaceutical Industries, Ltd. 3,4-dihydrothieno 2,3-d¨pyrimidine compounds and pharmaceutical application thereof
US5208250A (en) 1988-05-25 1993-05-04 Warner-Lambert Company Known and selected novel arylmethylenyl derivatives of thiazolidinones, imidazolidinones and oxazolidinones useful as antiallergy agents and anti-inflammatory agents
JPH07107055B1 (en) 1990-04-25 1995-11-15
SU1712359A1 (en) 1990-05-07 1992-02-15 Уфимский Нефтяной Институт 8'-hydroxyquinoline-7-carboxylic acid 8-hydroxyquinoline ester hydrochloride as a bactericidal agent for inhibition of sulfate-reducing bacteria and cultures pseudomonas and arthrobacter
EP0464612B1 (en) 1990-06-28 1998-05-13 Fuji Photo Film Co., Ltd. Silver halide photographic materials
EP0481614A1 (en) 1990-10-01 1992-04-22 Merck & Co. Inc. Substituted pyridopyrimidinones and related heterocycles as angiotensin II antagonists
US5480883A (en) 1991-05-10 1996-01-02 Rhone-Poulenc Rorer Pharmaceuticals Inc. Bis mono- and bicyclic aryl and heteroaryl compounds which inhibit EGF and/or PDGF receptor tyrosine kinase
AU1625192A (en) 1991-05-31 1992-12-03 Zeneca Limited Heterocyclic derivatives
JP3108483B2 (en) 1991-09-30 2000-11-13 日清製粉株式会社 Indole derivatives and anti-ulcer drugs containing the same as active ingredients
HUT64064A (en) 1992-02-13 1993-11-29 Chinoin Gyogyszer Es Vegyeszet Process for producing puyrido/1,2-a/pyrimidine derivatives and pharmaceutical compositions comprising same as active ingredient
AU3933493A (en) * 1992-04-24 1993-11-29 E.I. Du Pont De Nemours And Company Arthropodicidal and fungicidal aminopyrimidines
TW229140B (en) 1992-06-05 1994-09-01 Shell Internat Res Schappej B V
FR2714907B1 (en) 1994-01-07 1996-03-29 Union Pharma Scient Appl New adenosine derivatives, their preparation processes, pharmaceutical compositions containing them.
US6342501B1 (en) 1994-02-25 2002-01-29 The Regents Of The University Of Michigan Pyrrolo[2,3-d] pyrimidines as antiviral agents
JPH09176162A (en) 1995-12-22 1997-07-08 Toubishi Yakuhin Kogyo Kk Thiazolidinedione derivative, its production and medicinal composition containing the same
JPH1025294A (en) 1996-03-26 1998-01-27 Akira Matsuda Condensed heterocyclic derivative, its production and malignant tumor therapeutic agent containing the same
EP2223920A3 (en) 1996-06-19 2011-09-28 Aventis Pharma Limited Substituted azabicyclic compounds
ES2232871T3 (en) 1996-07-03 2005-06-01 Sumitomo Pharmaceuticals Company, Limited NEW DERIVATIVES OF PURINA.
US5866702A (en) 1996-08-02 1999-02-02 Cv Therapeutics, Incorporation Purine inhibitors of cyclin dependent kinase 2
US6630496B1 (en) 1996-08-26 2003-10-07 Genetics Institute Llc Inhibitors of phospholipase enzymes
JPH10231297A (en) 1997-02-20 1998-09-02 Japan Energy Corp New adenine-1-noxide derivative, and use of the same as medicine
CN1130363C (en) 1997-11-12 2003-12-10 三菱化学株式会社 Purine derivatives and medicine containing the same as the active ingredient
TW572758B (en) 1997-12-22 2004-01-21 Sumitomo Pharma Type 2 helper T cell-selective immune response inhibitors comprising purine derivatives
EP1056719A2 (en) 1998-02-25 2000-12-06 Genetics Institute, Inc. Inhibitors of phospholipase enzymes
CN1298404A (en) 1998-02-25 2001-06-06 遗传研究所有限公司 Inhibitors of phospholipase A2
US6828344B1 (en) 1998-02-25 2004-12-07 Genetics Institute, Llc Inhibitors of phospholipase enzymes
US6479487B1 (en) 1998-02-26 2002-11-12 Aventis Pharmaceuticals Inc. 6, 9-disubstituted 2-[trans-(4-aminocyclohexyl)amino] purines
CA2330756C (en) 1998-05-04 2007-10-02 Asta Medica Aktiengesellschaft Indole derivatives and their use for the treatment of malignant and other diseases based on pathological cell proliferation
AU3851199A (en) 1998-05-26 1999-12-13 Chugai Seiyaku Kabushiki Kaisha Heterocyclic indole derivatives and mono- or diazaindole derivatives
JP3997651B2 (en) 1998-06-24 2007-10-24 コニカミノルタホールディングス株式会社 Novel dye, image recording material, thermal transfer material and ink jet recording liquid
DE69913712T2 (en) 1998-08-25 2004-10-07 Uab Res Foundation Birmingham INHIBITORS OF BACTERIAL NAD SYNTHETASE
TR200102714T2 (en) 1999-02-01 2002-01-21 Cv Therapeutics, Inc. Purine inhibitors of cyclin dependent kinase 2 and IkB - &
JP2000281654A (en) 1999-03-26 2000-10-10 Tanabe Seiyaku Co Ltd Isoquinoline dertivative
DE19932571A1 (en) 1999-07-13 2001-01-18 Clariant Gmbh Process for the preparation of biarylene using palladophosphacyclobutane catalysis
JP2001151771A (en) 1999-09-10 2001-06-05 Kyowa Hakko Kogyo Co Ltd Nitrogen-containing aromatic heterocyclic derivative
US6436965B1 (en) 2000-03-02 2002-08-20 Merck Frosst Canada & Co. PDE IV inhibiting amides, compositions and methods of treatment
EP1138328A1 (en) 2000-03-29 2001-10-04 Eli Lilly And Company Limited Naphthalene derivatives as CNS drugs
US6667300B2 (en) 2000-04-25 2003-12-23 Icos Corporation Inhibitors of human phosphatidylinositol 3-kinase delta
CN1331340A (en) 2000-06-30 2002-01-16 上海博德基因开发有限公司 Polypeptide-human topoisomerase 12.1 and polynucleotide for coding it
PT1300398E (en) 2000-07-05 2006-07-31 Astellas Pharma Inc PROCESSES AND COMPOSITIONS FOR CANCER THERAPIES, USING GENES THAT CODE THE BETA INTERFERENCE
FR2814073B1 (en) 2000-09-21 2005-06-24 Yang Ji Chemical Company Ltd ANTIFUNGAL AND / OR ANTIPARASITIC PHARMACEUTICAL COMPOSITION AND NOVEL INDOLE DERIVATIVES AS ACTIVE INGREDIENTS OF SUCH A COMPOSITION
DOP2002000334A (en) 2001-02-14 2002-08-30 Warner Lambert Co BICYCLE PYRIMIDINES AS MATRIX METALOPROTEINASE INHIBITORS
SE0100568D0 (en) 2001-02-20 2001-02-20 Astrazeneca Ab Compounds
JP3616628B2 (en) 2001-03-01 2005-02-02 塩野義製薬株式会社 Nitrogen-containing aromatic heterocyclic compounds having HIV integrase inhibitory activity
UA76977C2 (en) 2001-03-02 2006-10-16 Icos Corp Aryl- and heteroaryl substituted chk1 inhibitors and their use as radiosensitizers and chemosensitizers
US20050107400A1 (en) 2001-03-30 2005-05-19 Boyd Leslie F. Use of pyrazolopyridines as therapeutic compounds
EP1372643A1 (en) 2001-03-30 2004-01-02 Smithkline Beecham Corporation Pyrazolopyridines, process for their preparation and use as therapeutic compounds
WO2002088124A2 (en) 2001-04-27 2002-11-07 Smithkline Beecham Corporation Pyrazolo'1,5-a!pyridine derivatives
CZ294535B6 (en) 2001-08-02 2005-01-12 Ústav Experimentální Botaniky Avčr Heterocyclic compounds based on N6-substituted adenine, processes of their preparation, their use in the preparation of medicaments, cosmetic compositions and growth regulators, as well as pharmaceutical preparations, cosmetic compositions and growth regulators in which these compounds are comprised
GB0121033D0 (en) 2001-08-30 2001-10-24 Novartis Ag Organic compounds
US8124625B2 (en) 2001-09-14 2012-02-28 Shionogi & Co., Ltd. Method of enhancing the expression of apolipoprotein AI using olefin derivatives
MXPA04002136A (en) 2001-09-26 2005-03-07 Bayer Pharmaceuticals Corp 1,6-naphthyridine derivatives as antidiabetics.
JP4459619B2 (en) 2001-10-02 2010-04-28 グラクソスミスクライン・リミテッド・ライアビリティ・カンパニー Compound
AU2002359364A1 (en) 2001-11-09 2003-05-26 Enzon, Inc. Polymeric thiol-linked prodrugs employing benzyl elimination systems
EP1314733A1 (en) 2001-11-22 2003-05-28 Aventis Pharma Deutschland GmbH Indole-2-carboxamides as factor Xa inhibitors
DE60231439D1 (en) 2001-12-06 2009-04-16 Merck & Co Inc MITOTIC KINESINE HEMMER
CA2467722A1 (en) 2001-12-06 2003-06-19 Merck & Co., Inc. Thienopyrimidinone derivatives as mitotic kinesin inhibitors
TW200301135A (en) 2001-12-27 2003-07-01 Otsuka Maryland Res Inst Inc Pharmaceutical compositions comprising a multifunctional phosphodiesterase inhibitor and an adenosine uptake inhibitor
US7402595B2 (en) 2002-02-13 2008-07-22 Takeda Pharmaceutical Company Limited JNK inhibitor
AU2003225668A1 (en) 2002-03-01 2003-09-16 Pintex Pharmaceutical, Inc. Pin1-modulating compounds and methods of use thereof
ES2291628T3 (en) 2002-04-03 2008-03-01 Bristol-Myers Squibb Company THYROPHENE BASED THYMPHENE COMPOUNDS AND PHARMACEUTICAL COMPOSITIONS THAT INCLUDE SUCH COMPOUNDS.
MXPA04010983A (en) 2002-05-06 2005-02-14 Genelabs Tech Inc Nucleoside derivatives for treating hepatitis c virus infection.
EP2045242A1 (en) 2002-08-13 2009-04-08 Shionogi&Co., Ltd. Heterocyclic compounds having inhibitory activity against HIV integrase
JP4190232B2 (en) 2002-08-26 2008-12-03 富士通株式会社 How to perform mechanical polishing
BR0314761A (en) 2002-09-27 2005-07-26 Sumitomo Pharma Adenine Compound and its Use
CA2507509A1 (en) * 2002-11-25 2004-06-10 Mochida Pharmaceutical Co., Ltd. Therapeutic agent for respiratory disease containing 4-hydroxypiperidine derivative as active ingredient
JP4500689B2 (en) 2002-12-26 2010-07-14 エーザイ・アール・アンド・ディー・マネジメント株式会社 Selective estrogen receptor modulator
CZ294538B6 (en) 2002-12-30 2005-01-12 Ústav Experimentální Botaniky Akademie Vědčeské Re Substituting derivatives of N6-benzyladenosine, process of their preparation, their use in the preparation of medicaments, cosmetic compositions and growth regulators, as well as pharmaceutical preparations, cosmetic compositions and growth regulators in which these compounds are comprised
RU2233842C1 (en) 2003-01-13 2004-08-10 Петров Владимир Иванович Derivatives of purine eliciting antiviral activity
AR043692A1 (en) 2003-02-06 2005-08-10 Novartis Ag 2-CYANOPIRROLOPIRIMIDINAS AND ITS PHARMACEUTICAL USES
GB0304640D0 (en) 2003-02-28 2003-04-02 Novartis Ag Organic compounds
SE0300908D0 (en) 2003-03-31 2003-03-31 Astrazeneca Ab Azaindole derivatives, preparations thereof, uses thereof and compositions containing them
US7129264B2 (en) 2003-04-16 2006-10-31 Bristol-Myers Squibb Company Biarylmethyl indolines and indoles as antithromboembolic agents
US20060247245A1 (en) 2003-05-05 2006-11-02 Yuelian Xu Substituted imidazolopyrazine and triazolopyrazine derivatives: gabaa receptor ligands
AU2004249730A1 (en) 2003-06-20 2004-12-29 Novartis Vaccines And Diagnostics, Inc. Pyridino(1,2-A)pyrimidin-4-one compounds as anticancer agents
WO2005000309A2 (en) 2003-06-27 2005-01-06 Ionix Pharmaceuticals Limited Chemical compounds
JP4570015B2 (en) 2003-07-14 2010-10-27 クミアイ化学工業株式会社 2-Isoxazoline derivatives and herbicides containing them as active ingredients
MXPA06001758A (en) 2003-08-15 2006-08-11 Irm Llc 6-substituted anilino purines as rtk inhibitors.
BRPI0414533A (en) 2003-09-18 2006-11-07 Conforma Therapeutics Corp compound, pharmaceutical composition, and methods for inhibiting an hsp90 and treating an individual having an hsp90 mediated disorder
US7709476B2 (en) 2003-09-23 2010-05-04 Merck Sharp & Dohme Corp. Isoquinolinone potassium channel inhibitors
CA2553724A1 (en) 2004-02-03 2005-08-18 Abbott Laboratories Aminobenzoxazoles as therapeutic agents
EP1717238A4 (en) 2004-02-16 2008-03-05 Daiichi Seiyaku Co Fungicidal heterocyclic compounds
WO2005091857A2 (en) 2004-03-12 2005-10-06 Bayer Pharmaceuticals Corporation 1,6-naphthyridine and 1,8-naphthyridine derivatives and their use to treat diabetes and related disorders
CN1560035A (en) 2004-03-12 2005-01-05 沈阳药科大学 5-hydroxylic indole-3-carboxylic ester kind derivantion
US20070225303A1 (en) 2004-03-26 2007-09-27 Haruhisa Ogita 8-Oxoadenine Compound
US7217702B2 (en) 2004-04-02 2007-05-15 Adenosine Therapeutics, Llc Selective antagonists of A2A adenosine receptors
DK1761540T3 (en) 2004-05-13 2016-11-21 Icos Corp Quinazolinones as inhibitors of human phosphatidylinositol 3-kinase DELTA
WO2005110410A2 (en) 2004-05-14 2005-11-24 Abbott Laboratories Kinase inhibitors as therapeutic agents
CA2574204A1 (en) 2004-07-22 2006-01-26 Astrazeneca Ab Fused pyrimidones useful in the treatment and the prevention of cancer
DE102004044221A1 (en) 2004-09-14 2006-03-16 Boehringer Ingelheim Pharma Gmbh & Co. Kg New 3-methyl-7-butynyl xanthines, their preparation and their use as pharmaceuticals
GB0420719D0 (en) 2004-09-17 2004-10-20 Addex Pharmaceuticals Sa Novel allosteric modulators
MX2007004699A (en) 2004-10-19 2007-06-14 Novartis Vaccines & Diagnostic Indole and benzimidazole derivatives.
EP1807417A2 (en) 2004-11-04 2007-07-18 Neurogen Corporation Pyrazolylmethyl heteroaryl derivatives
JP2008520744A (en) 2004-11-19 2008-06-19 ザ・レジェンツ・オブ・ザ・ユニバーシティ・オブ・カリフォルニア Anti-inflammatory pyrazolopyrimidine
WO2006089106A2 (en) 2005-02-17 2006-08-24 Icos Corporation Phosphoinositide 3-kinase inhibitors for inhibiting leukocyte accumulation
EP1917250B1 (en) 2005-06-27 2010-07-21 Amgen, Inc Anti-inflammatory aryl nitrile compounds
FR2889192A1 (en) 2005-07-27 2007-02-02 Cytomics Systems Sa ANTIFUNGAL COMPOUNDS, COMPOSITIONS CONTAINING THESE COMPOUNDS, AND USES THEREOF
US20070066624A1 (en) 2005-08-16 2007-03-22 Anormed, Inc. Chemokine receptor binding compounds
US7642270B2 (en) 2005-09-14 2010-01-05 Janssen Pharmaceutica N.V. 5-oxo-5,8-dihydro-pyrido-pyrimidine as inhibitors of c-fms kinase
WO2007034817A1 (en) 2005-09-22 2007-03-29 Dainippon Sumitomo Pharma Co., Ltd. Novel adenine compound
DK1951684T3 (en) * 2005-11-01 2016-10-24 Targegen Inc BIARYLMETAPYRIMIDIN kinase inhibitors
EP1783114A1 (en) 2005-11-03 2007-05-09 Novartis AG N-(hetero)aryl indole derivatives as pesticides
ES2353549T3 (en) 2005-11-10 2011-03-03 Chemocentryx, Inc. REPLACED KINOLONES AND METHODS OF USE.
US7989461B2 (en) 2005-12-23 2011-08-02 Amgen Inc. Substituted quinazolinamine compounds for the treatment of cancer
WO2007087548A2 (en) 2006-01-25 2007-08-02 Smithkline Beecham Corporation Chemical compounds
PE20071025A1 (en) 2006-01-31 2007-10-17 Mitsubishi Tanabe Pharma Corp TRISUSTITUTED AMINE COMPOUND
PE20070978A1 (en) 2006-02-14 2007-11-15 Novartis Ag HETEROCICLIC COMPOUNDS AS INHIBITORS OF PHOSPHATIDYLINOSITOL 3-KINASES (PI3Ks)
JPWO2007102392A1 (en) 2006-03-03 2009-07-23 塩野義製薬株式会社 MMP-13 selective inhibitor
BRPI0709699A2 (en) 2006-03-29 2011-07-26 Foldrx Pharmaceuticals Inc inhibition of alpha synuclein toxicity
ES2436028T3 (en) 2006-06-23 2013-12-26 Radius Health, Inc. Treatment of vasomotor symptoms with selective estrogen receptor modulators
EP2044086A2 (en) 2006-06-30 2009-04-08 Janssen Pharmaceutica N.V. Thiazolopyrimidine modulators of trpv1
US20080009508A1 (en) 2006-07-10 2008-01-10 Lucie Szucova 6,9-Disubstituted Purine Derivatives And Their Use For Treating Skin
WO2008006540A1 (en) 2006-07-12 2008-01-17 Syngenta Participations Ag Triazolopyridine derivatives as herbicides
CA2662074A1 (en) 2006-08-30 2008-03-06 Cellzome Limited Triazole derivatives as kinase inhibitors
EP1972631A1 (en) 2007-03-23 2008-09-24 Novartis AG Imidazopyridazines as PI3K lipid kinase inhibitors
US20080114007A1 (en) 2006-10-31 2008-05-15 Player Mark R 5-oxo-5,8-dihydro-pyrido-pyrimidines as inhibitors of c-fms kinase
CN101605797A (en) 2006-11-13 2009-12-16 伊莱利利公司 The Thienopyrimidinones of treatment inflammatory disease and cancer
JP5572388B2 (en) 2006-11-22 2014-08-13 インサイト・コーポレイション Imidazotriazines and imidazopyrimidines as kinase inhibitors
CN101631786A (en) 2006-12-20 2010-01-20 先灵公司 Novel jnk inhibitor
ATE486877T1 (en) 2006-12-29 2010-11-15 Hoffmann La Roche AZASPIRO DERIVATIVES
CA2677493A1 (en) 2007-02-05 2008-08-14 Xenon Pharmaceuticals Inc. Pyridopyrimidinone compounds useful in treating sodium channel-mediated diseases or conditions
US20090047246A1 (en) 2007-02-12 2009-02-19 Intermune, Inc. Novel inhibitors of hepatitis c virus replication
PE20081887A1 (en) 2007-03-20 2009-01-16 Dainippon Sumitomo Pharma Co NEW ADENINE COMPOUND
US20080233127A1 (en) 2007-03-21 2008-09-25 Wyeth Imidazolopyrimidine analogs and their use as pi3 kinase and mtor inhibitors
PT2137186E (en) 2007-03-23 2016-03-30 Amgen Inc Heterocyclic compounds and their uses
CA2680783C (en) 2007-03-23 2012-04-24 Amgen Inc. Heterocyclic compounds and their uses
US8039505B2 (en) 2007-04-11 2011-10-18 University Of Utah Research Foundation Compounds for modulating T-cells
US8633186B2 (en) 2007-06-08 2014-01-21 Senomyx Inc. Modulation of chemosensory receptors and ligands associated therewith
US9603848B2 (en) 2007-06-08 2017-03-28 Senomyx, Inc. Modulation of chemosensory receptors and ligands associated therewith
JP2010531304A (en) 2007-06-18 2010-09-24 ユニバーシティ オブ ルイビル リサーチ ファウンデーション、インコーポレイテッド PFKFB3 inhibitor family with antineoplastic activity
TWI434849B (en) 2007-06-29 2014-04-21 Gilead Sciences Inc Modulators of toll-like receptor 7
US20090053192A1 (en) 2007-08-10 2009-02-26 Burnham Institute For Medical Research Tissue-nonspecific alkaline phosphatase (tnap) activators and uses thereof
JP2009076865A (en) 2007-08-29 2009-04-09 Fujifilm Corp Organic electroluminescence device
WO2009034386A1 (en) 2007-09-13 2009-03-19 Astrazeneca Ab Derivatives of adenine and 8-aza-adenine and uses thereof-796
JP2009080233A (en) 2007-09-26 2009-04-16 Kyocera Mita Corp Electrophotographic photoreceptor
CZ300774B6 (en) 2007-10-05 2009-08-05 Univerzita Palackého Substituted 6-(alkylbenzylamino)purine derivatives for use as cytokinin receptor antagonists and compositions in which these compounds are comprised
JP2011503103A (en) 2007-11-07 2011-01-27 フォールドアールエックス ファーマシューティカルズ インコーポレーティッド Methods for regulating protein transport
WO2009063235A1 (en) 2007-11-13 2009-05-22 Astrazeneca Ab Derivatives of 1,9-dihydro-6h-purin-6-one and uses thereof-018
JP2009120686A (en) 2007-11-14 2009-06-04 Toyo Ink Mfg Co Ltd Photopolymerization initiator, polymerizable composition, and method for producing polymer
EP2231661A1 (en) 2007-12-19 2010-09-29 Amgen, Inc. Inhibitors of pi3 kinase
WO2009086303A2 (en) 2007-12-21 2009-07-09 University Of Rochester Method for altering the lifespan of eukaryotic organisms
CA2710452A1 (en) 2007-12-21 2009-07-09 Wyeth Llc Imidazo [1,2-a] pyridine compounds
US8399483B2 (en) 2007-12-21 2013-03-19 Ucb Pharma S.A. Quinoxaline and quinoline derivatives as kinase inhibitors
US7960397B2 (en) 2007-12-28 2011-06-14 Institute Of Experimental Botany, Academy Of Sciences Of The Czech Republic 6,9-disubstituted purine derivatives and their use as cosmetics and cosmetic compositions
US8193182B2 (en) 2008-01-04 2012-06-05 Intellikine, Inc. Substituted isoquinolin-1(2H)-ones, and methods of use thereof
NZ586642A (en) 2008-01-11 2012-04-27 Natco Pharma Ltd Novel pyrazolo [3, 4 -d] pyrimidine derivatives as anti -cancer agents
US9089572B2 (en) 2008-01-17 2015-07-28 California Institute Of Technology Inhibitors of p97
WO2009097446A1 (en) 2008-01-30 2009-08-06 Genentech, Inc. Pyrazolopyrimidine pi3k inhibitor compounds and methods of use
WO2009128520A1 (en) 2008-04-18 2009-10-22 塩野義製薬株式会社 Heterocyclic compound having inhibitory activity on p13k
US8119647B2 (en) 2008-04-23 2012-02-21 Glenmark Pharmaceuticals S.A. Fused pyrimidineone compounds as TRPV3 modulators
WO2009140215A2 (en) 2008-05-11 2009-11-19 Geraghty, Erin Method for treating drug-resistant bacterial and other infections with clioquinol, phanquinone, and related compounds
WO2009151972A1 (en) 2008-05-28 2009-12-17 , The United States Of America, As Represented By The Secretary Of The Army, On Behalf Of U.S. Army Medical Research And Materiel Command Small molecule inhibitors of botulinum neurotoxins
MX2010013876A (en) 2008-06-20 2011-03-04 Metabolex Inc Aryl gpr119 agonists and uses thereof.
US8026271B2 (en) 2008-07-11 2011-09-27 National Health Research Institutes Formulations of indol-3-yl-2-oxoacetamide compounds
WO2010018458A2 (en) 2008-08-12 2010-02-18 Crystalgenomics, Inc. Phenol derivatives and methods of use thereof
CA2738429C (en) 2008-09-26 2016-10-25 Intellikine, Inc. Heterocyclic kinase inhibitors
PT2355828T (en) 2008-11-13 2018-07-02 Gilead Calistoga Llc Therapies for hematologic malignancies
WO2010075068A1 (en) 2008-12-16 2010-07-01 Schering Corporation Pyridopyrimidine derivatives and methods of use thereof
TWI469979B (en) 2008-12-24 2015-01-21 Bial Portela & Ca Sa Faah inhibitor, and pharmaceutical composition and use thereof
WO2010083444A1 (en) 2009-01-15 2010-07-22 Anvyl, Llc Alpha7 nicotinic acetylcholine receptor allosteric modulators, their derivatives and uses thereof
US8513284B2 (en) 2009-02-13 2013-08-20 Ucb Pharma, S.A. Fused pyridine and pyrazine derivatives as kinase inhibitors
CA2756808A1 (en) 2009-03-31 2010-10-07 Arqule, Inc. Substituted indolo-piperidine compounds
WO2010118367A2 (en) 2009-04-10 2010-10-14 Progenics Pharmaceuticals, Inc. Antiviral pyrimidines
ES2548253T3 (en) 2009-04-20 2015-10-15 Gilead Calistoga Llc Methods for the treatment of solid tumors
WO2010127208A1 (en) 2009-04-30 2010-11-04 Forest Laboratories Holdings Limited Inhibitors of acetyl-coa carboxylase
JP5789252B2 (en) 2009-05-07 2015-10-07 インテリカイン, エルエルシー Heterocyclic compounds and uses thereof
JP2012531435A (en) 2009-06-25 2012-12-10 アムジエン・インコーポレーテツド 4H-pyrido [1,2-a] pyrimidin-4-one derivatives as PI3K inhibitors
TW201111362A (en) 2009-06-25 2011-04-01 Amgen Inc Heterocyclic compounds and their uses
AR077252A1 (en) 2009-06-29 2011-08-10 Xenon Pharmaceuticals Inc ESPIROOXINDOL COMPOUND ENANTIOMERS AND THEIR USES AS THERAPEUTIC AGENTS
UA107667C2 (en) 2009-06-29 2015-02-10 Аджиос Фармасьютікалз, Інк. Medical compounds stimulating activity of pyruvate kinase m2, compositions on their basis and use in the treatment of cancer
AR077280A1 (en) 2009-06-29 2011-08-17 Incyte Corp PYRIMIDINONES AS PI3K INHIBITORS, AND PHARMACEUTICAL COMPOSITIONS THAT UNDERSTAND THEM
FR2947269B1 (en) 2009-06-29 2013-01-18 Sanofi Aventis NEW ANTICANCER COMPOUNDS
WO2011011550A1 (en) 2009-07-21 2011-01-27 Calistoga Pharmaceuticals Inc. Treatment of liver disorders with pi3k inhibitors
MX2012004020A (en) 2009-10-20 2012-05-08 Cellzome Ltd Heterocyclyl pyrazolopyrimidine analogues as jak inhibitors.
NZ598933A (en) 2009-10-22 2013-04-26 Gilead Sciences Inc Derivatives of purine or deazapurine useful for the treatment of (inter alia) viral infections
MX352661B (en) 2009-11-05 2017-12-04 Rhizen Pharmaceuticals S A Star Novel benzopyran kinase modulators.
PE20121469A1 (en) 2009-11-10 2012-11-01 Pfizer N1- PYRAZOLOSPIROKETONE ACETYL-COA CARBOXYLASE INHIBITORS
EP2499126B1 (en) 2009-11-12 2015-01-07 UCB Pharma, S.A. Fused bicyclic pyridine and pyrazine derivatives as kinase inhibitors
WO2011058111A1 (en) 2009-11-12 2011-05-19 Ucb Pharma S.A. Aminopurine derivatives as kinase inhibitors
JP2013513551A (en) 2009-12-10 2013-04-22 中国医学科学院葯物研究所 Use of N6-substituted adenosine derivatives and N6-substituted adenine derivatives for sedation, hypnosis, antidepressant, anticonvulsant, antiepileptic, antiparkinson's disease and recognition / prevention treatment
AU2010330875B2 (en) 2009-12-18 2013-08-01 Amgen Inc. Heterocyclic compounds and their uses
WO2011075630A1 (en) 2009-12-18 2011-06-23 Incyte Corporation Substituted fused aryl and heteroaryl derivatives as pi3k inhibitors
WO2011075643A1 (en) 2009-12-18 2011-06-23 Incyte Corporation Substituted heteroaryl fused derivatives as pi3k inhibitors
JP2011136925A (en) 2009-12-28 2011-07-14 Dainippon Sumitomo Pharma Co Ltd Nitrogen-containing bicyclic compound
EA201290888A1 (en) 2010-03-22 2013-04-30 Гленмарк Фармасьютикалс С.А. PHARMACEUTICAL COMPOSITION CONTAINING DERIVATIVE PYRIMIDINONE
UY33304A (en) 2010-04-02 2011-10-31 Amgen Inc HETEROCYCLIC COMPOUNDS AND THEIR USES
EP2558463A1 (en) 2010-04-14 2013-02-20 Incyte Corporation Fused derivatives as i3 inhibitors
JP5951600B2 (en) 2010-05-21 2016-07-13 インフィニティー ファーマシューティカルズ, インコーポレイテッド Compounds, compositions and methods for kinase regulation
US20110306622A1 (en) 2010-06-11 2011-12-15 Calitoga Pharmaceuticals, Inc. Methods of treating hematological disorders with quinazolinone compounds in selected subjects
US9062055B2 (en) 2010-06-21 2015-06-23 Incyte Corporation Fused pyrrole derivatives as PI3K inhibitors
US20130085131A1 (en) 2010-07-01 2013-04-04 Amgen Inc. Heterocyclic compounds and their uses
WO2012003262A1 (en) 2010-07-01 2012-01-05 Amgen Inc. Heterocyclic compounds and their use as inhibitors of pi3k activity
MX2012015134A (en) 2010-07-02 2013-05-06 Amgen Inc Heterocyclic compounds and their use as inhibitors of pi3k activity.
EP2619209A1 (en) 2010-09-24 2013-07-31 Gilead Calistoga LLC Atropisomers of pi3k-inhibiting compounds
AU2011323243A1 (en) * 2010-11-04 2013-05-23 Amgen Inc. Heterocyclic compounds and their uses
CA2817577A1 (en) 2010-11-10 2012-05-18 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
WO2012069343A1 (en) 2010-11-25 2012-05-31 Henkel Ag & Co. Kgaa Laundry article having improved dirt scavenging properties
CN103298460B (en) 2010-12-14 2016-06-01 电泳有限公司 Casein kinase 1 �� (CK1 ��) inhibitor
TW201249844A (en) 2010-12-20 2012-12-16 Incyte Corp N-(1-(substituted-phenyl)ethyl)-9H-purin-6-amines as PI3K inhibitors
US20130267526A1 (en) 2010-12-23 2013-10-10 Amgen, Inc. Heterocyclic compounds and their uses
US8809349B2 (en) 2011-01-10 2014-08-19 Infinity Pharmaceuticals, Inc. Processes for preparing isoquinolinones and solid forms of isoquinolinones
WO2012125629A1 (en) 2011-03-14 2012-09-20 Incyte Corporation Substituted diamino-pyrimidine and diamino-pyridine derivatives as pi3k inhibitors
AU2012284088B2 (en) 2011-07-19 2015-10-08 Infinity Pharmaceuticals Inc. Heterocyclic compounds and uses thereof
SI2751109T1 (en) 2011-09-02 2017-03-31 Incyte Holdings Corporation Heterocyclylamines as pi3k inhibitors
AR090548A1 (en) 2012-04-02 2014-11-19 Incyte Corp BICYCLIC AZAHETEROCICLOBENCILAMINS AS PI3K INHIBITORS

Patent Citations (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5521184A (en) 1992-04-03 1996-05-28 Ciba-Geigy Corporation Pyrimidine derivatives and processes for the preparation thereof
WO2000009495A1 (en) 1998-08-11 2000-02-24 Novartis Ag Isoquinoline derivatives with angiogenesis inhibiting activity
WO2000053595A1 (en) 1999-03-06 2000-09-14 Astrazeneca Ab Pyrimidine compounds
WO2001014402A1 (en) 1999-08-19 2001-03-01 Isis Pharmaceuticals, Inc. Antisense modulation of focal adhesion kinase expression
WO2001064655A1 (en) 2000-03-01 2001-09-07 Astrazeneca Ab 2, 4-di(hetero-)arylamino (-oxy)-5-substituted pyrimidines as antineoplastic agents
WO2002000196A2 (en) 2000-06-28 2002-01-03 Smithkline Beecham P.L.C. Wet milling process
WO2003024967A2 (en) 2001-09-19 2003-03-27 Aventis Pharma S.A. Indolizines as kinase protein inhibitors
WO2003037347A1 (en) 2001-10-30 2003-05-08 Novartis Ag Staurosporine derivatives as inhibitors of flt3 receptor tyrosine kinase activity
WO2003099771A2 (en) 2002-05-29 2003-12-04 Novartis Ag Diaryl urea derivatives useful for the treatment of protein kinase dependent diseases
WO2004005281A1 (en) 2002-07-05 2004-01-15 Novartis Ag Inhibitors of tyrosine kinases
WO2004046120A2 (en) 2002-11-15 2004-06-03 Vertex Pharmaceuticals Incorporated Diaminotriazoles useful as inhibitors of protein kinases
WO2004048365A1 (en) * 2002-11-21 2004-06-10 Chiron Corporation 2,4,6-trisubstituted pyrimidines as phosphotidylinositol (pi) 3-kinase inhibitors and their use in the treatment of cancer
WO2004056786A2 (en) 2002-12-20 2004-07-08 Pfizer Products Inc. Pyrimidine derivates for the treatment of abnormal cell growth
WO2004080980A1 (en) 2003-03-14 2004-09-23 Novartis Ag 2, 4- di (phenylamino) pyrimidines useful in the treatment of neoplastic diseases, inflammatory and immune system disorders
WO2005028444A1 (en) 2003-09-24 2005-03-31 Novartis Ag 1,4-disubstituted isoquinilone derivatives as raf-kinase inhibitors useful for the treatment of proliferative diseases
WO2006056399A2 (en) 2004-11-24 2006-06-01 Novartis Ag Combinations of jak inhibitors and at least one of bcr-abl, flt-3, fak or raf kinase inhibitors
WO2007042806A1 (en) * 2005-10-11 2007-04-19 Ludwig Institute For Cancer Research Pyrimidine derivatives for the treatment of cancer
WO2008032033A1 (en) * 2006-09-14 2008-03-20 Astrazeneca Ab 4-benzimidazolyl-2-morpholino-6-piperazinylpyrimidine derivatives as pi3k and mtor inhibitors for the treatment of proliferative disorders
WO2011025889A1 (en) 2009-08-28 2011-03-03 Takeda Pharmaceutical Company Limited HEXAHYDROOXAZINOPTERINE COMPOUNDS FOR USE AS mTOR INHIBITORS
WO2011092198A1 (en) * 2010-01-26 2011-08-04 Boehringer Ingelheim International Gmbh 5-alkynyl-pyrimidines

Non-Patent Citations (28)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences, 17th ed.,", 1985, MACK PUBLISHING COMPANY, pages: 1418
ALI ET AL., NATURE, vol. 431, no. 7011, 2004, pages 1007 - 11
BADER ET AL., PROC NATL ACAD SCI U S A., vol. 103, no. 5, 2006, pages 1475 - 9
BARBER ET AL., NAT MED., vol. 11, no. 9, 2005, pages 933 - 5
BENISTANT ET AL., ONCOGENE, vol. 19, no. 44, 2000, pages 5083 - 90
BILLOTTET ET AL., ONCOGENE, vol. 25, no. 50, 2006, pages 6648 - 59
BROCK ET AL., J CELL BIOL., vol. 160, no. 1, 2003, pages 89 - 99
CAMPS ET AL., NAT MED., vol. 11, no. 9, 2005, pages 936 - 43
CANTLEY, SCIENCE, vol. 296, no. 5573, 2002, pages 1655 - 7
CLAYTON ET AL., J EXP MED., vol. 196, no. 6, 2002, pages 753 - 63
HICKEY ET AL., J BIOL CHEM., vol. 281, no. 5, 2006, pages 2441 - 50
JIMENEZ ET AL., J BIOL CHEM., vol. 277, no. 44, 2002, pages 41556 - 62
JOU ET AL., MOL CELL BIOL., vol. 22, no. 24, 2002, pages 8580 - 91
JOURNAL OF PHARMACEUTICAL SCIENCE, vol. 66, 1977, pages 2
KANG ET AL., PROC NATL ACAD SCI U S A., vol. 102, no. 3, 2005, pages 802 - 7
KARL-F. B-LOM; BRIAN-GLASS; RICHARD--SPARKS; ANDREW-P.: "Preparative LC-MS Purification: Improved Compound Specific Method Optimization", COMBS J. COMBI. CHEM., vol. 6, no. 6, 2004, pages 874 - 883
KNOBBE, NEUROPATHOL APPL NEUROBIOL., vol. 31, no. 5, 2005, pages 486 - 90
LEE ET AL., FASEB J., vol. 20, no. 3, 2006, pages 455 - 65
MIZOGUCHI ET AL., BRAIN PATHOL., vol. 14, no. 4, 2004, pages 372 - 7
OKKENHAUG ET AL., SCIENCE, vol. 297, no. 5583, 2002, pages 1031 - 4
RANDIS ET AL., EUR. J. IMMUNOL., vol. 38, no. 5, 2008, pages 1215 - 24
SAMUELS ET AL., CURR OPIN ONCOL., vol. 18, no. L, 2006, pages 77 - 82
SAMUELS ET AL., SCIENCE, vol. 304, no. 5670, 2004, pages 554
SASAKI ET AL., SCIENCE, vol. 287, no. 5455, 2000, pages 1040 - 6
SUJOBERT ET AL., BLOOD, vol. 106, no. 3, 2005, pages 1063 - 6
T. W. GREENE; P. G. M. WUTS: "Protective Groups in Organic Synthesis, 3rd Ed.,", 1999, WILEY & SONS, INC.
THOMAS ET AL., EUR J FMMUNOT., vol. 35, no. 4, 2005, pages 1283 - 91
VANHAESEBROECK, TRENDS BIOCHEM SCI., vol. 30, no. 4, 2005, pages 194 - 204

Cited By (63)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10428087B2 (en) 2009-06-29 2019-10-01 Incyte Corporation Pyrimidinones as PI3K inhibitors
US9975907B2 (en) 2009-06-29 2018-05-22 Incyte Holdings Corporation Pyrimidinones as PI3K inhibitors
US8940752B2 (en) 2009-06-29 2015-01-27 Incyte Corporation Pyrimidinones as PI3K inhibitors
US11401280B2 (en) 2009-06-29 2022-08-02 Incyte Holdings Corporation Pyrimidinones as PI3K inhibitors
US10829502B2 (en) 2009-06-29 2020-11-10 Incyte Corporation Pyrimidinones as PI3K inhibitors
US9434746B2 (en) 2009-06-29 2016-09-06 Incyte Corporation Pyrimidinones as PI3K inhibitors
US8680108B2 (en) 2009-12-18 2014-03-25 Incyte Corporation Substituted fused aryl and heteroaryl derivatives as PI3K inhibitors
US9403847B2 (en) 2009-12-18 2016-08-02 Incyte Holdings Corporation Substituted heteroaryl fused derivatives as P13K inhibitors
US9193721B2 (en) 2010-04-14 2015-11-24 Incyte Holdings Corporation Fused derivatives as PI3Kδ inhibitors
US9062055B2 (en) 2010-06-21 2015-06-23 Incyte Corporation Fused pyrrole derivatives as PI3K inhibitors
US9096600B2 (en) 2010-12-20 2015-08-04 Incyte Corporation N-(1-(substituted-phenyl)ethyl)-9H-purin-6-amines as PI3K inhibitors
US9815839B2 (en) 2010-12-20 2017-11-14 Incyte Corporation N-(1-(substituted-phenyl)ethyl)-9H-purin-6-amines as PI3K inhibitors
US9527848B2 (en) 2010-12-20 2016-12-27 Incyte Holdings Corporation N-(1-(substituted-phenyl)ethyl)-9H-purin-6-amines as PI3K inhibitors
US9108984B2 (en) 2011-03-14 2015-08-18 Incyte Corporation Substituted diamino-pyrimidine and diamino-pyridine derivatives as PI3K inhibitors
US10376513B2 (en) 2011-09-02 2019-08-13 Incyte Holdings Corporation Heterocyclylamines as PI3K inhibitors
US11819505B2 (en) 2011-09-02 2023-11-21 Incyte Corporation Heterocyclylamines as PI3K inhibitors
US11433071B2 (en) 2011-09-02 2022-09-06 Incyte Corporation Heterocyclylamines as PI3K inhibitors
US10092570B2 (en) 2011-09-02 2018-10-09 Incyte Holdings Corporation Heterocyclylamines as PI3K inhibitors
US9199982B2 (en) 2011-09-02 2015-12-01 Incyte Holdings Corporation Heterocyclylamines as PI3K inhibitors
US9707233B2 (en) 2011-09-02 2017-07-18 Incyte Holdings Corporation Heterocyclylamines as PI3K inhibitors
US10646492B2 (en) 2011-09-02 2020-05-12 Incyte Corporation Heterocyclylamines as PI3K inhibitors
US9730939B2 (en) 2011-09-02 2017-08-15 Incyte Holdings Corporation Heterocyclylamines as PI3K inhibitors
WO2013088404A1 (en) 2011-12-15 2013-06-20 Novartis Ag Use of inhibitors of the activity or function of PI3K
US9309251B2 (en) 2012-04-02 2016-04-12 Incyte Holdings Corporation Bicyclic azaheterocyclobenzylamines as PI3K inhibitors
US10259818B2 (en) 2012-04-02 2019-04-16 Incyte Corporation Bicyclic azaheterocyclobenzylamines as PI3K inhibitors
US9944646B2 (en) 2012-04-02 2018-04-17 Incyte Holdings Corporation Bicyclic azaheterocyclobenzylamines as PI3K inhibitors
US9018221B2 (en) 2012-12-21 2015-04-28 Gilead Calistoga, Llc Phosphatidylinositol 3-kinase inhibitors
US9029384B2 (en) 2012-12-21 2015-05-12 Gilead Calistoga, LLC. Phosphatidylinositol 3-kinase inhibitors
US9266878B2 (en) 2012-12-21 2016-02-23 Gilead Calistoga Llc Phosphatidylinositol 3-kinase inhibitors
US9676750B2 (en) 2013-01-14 2017-06-13 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as pim kinase inhibitors
US10828290B2 (en) 2013-01-15 2020-11-10 Incyte Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as pim kinase inhibitors
US9849120B2 (en) 2013-01-15 2017-12-26 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US10517858B2 (en) 2013-01-15 2019-12-31 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as PIM kinase inhibitors
US11229631B2 (en) 2013-01-15 2022-01-25 Incyte Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US9550765B2 (en) 2013-01-15 2017-01-24 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US10265307B2 (en) 2013-01-15 2019-04-23 Incyte Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US9221795B2 (en) 2013-06-14 2015-12-29 Gilead Sciences, Inc. Phosphatidylinositol 3-kinase inhibitors
US10000507B2 (en) 2013-08-23 2018-06-19 Incyte Corporation Furo- and thieno-pyridine carboxamide compounds useful as pim kinase inhibitors
US10479803B2 (en) 2014-06-11 2019-11-19 Incyte Corporation Bicyclic heteroarylaminoalkyl phenyl derivatives as PI3K inhibitors
WO2015191677A1 (en) * 2014-06-11 2015-12-17 Incyte Corporation Bicyclic heteroarylaminoalkyl phenyl derivatives as pi3k inhibitors
US10077277B2 (en) 2014-06-11 2018-09-18 Incyte Corporation Bicyclic heteroarylaminoalkyl phenyl derivatives as PI3K inhibitors
US11130767B2 (en) 2014-06-11 2021-09-28 Incyte Corporation Bicyclic heteroarylaminoalkyl phenyl derivatives as PI3K inhibitors
US9822124B2 (en) 2014-07-14 2017-11-21 Incyte Corporation Bicyclic heteroaromatic carboxamide compounds useful as Pim kinase inhibitors
US9890162B2 (en) 2014-07-14 2018-02-13 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as pim kinase inhibitors
US11084822B2 (en) 2015-02-27 2021-08-10 Incyte Corporation Salts and processes of preparing a PI3K inhibitor
US10336759B2 (en) 2015-02-27 2019-07-02 Incyte Corporation Salts and processes of preparing a PI3K inhibitor
US9988401B2 (en) 2015-05-11 2018-06-05 Incyte Corporation Crystalline forms of a PI3K inhibitor
US9732097B2 (en) 2015-05-11 2017-08-15 Incyte Corporation Process for the synthesis of a phosphoinositide 3-kinase inhibitor
US10125150B2 (en) 2015-05-11 2018-11-13 Incyte Corporation Crystalline forms of a PI3K inhibitor
US9540347B2 (en) 2015-05-29 2017-01-10 Incyte Corporation Pyridineamine compounds useful as Pim kinase inhibitors
WO2016196244A1 (en) 2015-05-29 2016-12-08 Incyte Corporation Pyridineamine compounds useful as pim kinase inhibitors
US9802918B2 (en) 2015-05-29 2017-10-31 Incyte Corporation Pyridineamine compounds useful as Pim kinase inhibitors
US10336728B2 (en) 2015-09-09 2019-07-02 Incyte Corporation Salts of a Pim kinase inhibitor
US11066387B2 (en) 2015-09-09 2021-07-20 Incyte Corporation Salts of a Pim kinase inhibitor
US9862705B2 (en) 2015-09-09 2018-01-09 Incyte Corporation Salts of a pim kinase inhibitor
WO2017044730A1 (en) 2015-09-09 2017-03-16 Incyte Corporation Salts of a pim kinase inhibitor
US11505540B2 (en) 2015-09-09 2022-11-22 Incyte Corporation Salts of a Pim kinase inhibitor
US11053215B2 (en) 2015-10-02 2021-07-06 Incyte Corporation Heterocyclic compounds useful as Pim kinase inhibitors
WO2017059251A1 (en) 2015-10-02 2017-04-06 Incyte Corporation Heterocyclic compounds useful as pim kinase inhibitors
US10450296B2 (en) 2015-10-02 2019-10-22 Incyte Corporation Heterocyclic compounds useful as Pim kinase inhibitors
US9920032B2 (en) 2015-10-02 2018-03-20 Incyte Corporation Heterocyclic compounds useful as pim kinase inhibitors
US10596161B2 (en) 2017-12-08 2020-03-24 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms
US11278541B2 (en) 2017-12-08 2022-03-22 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms

Also Published As

Publication number Publication date
US9126948B2 (en) 2015-09-08
US20140057912A1 (en) 2014-02-27

Similar Documents

Publication Publication Date Title
US9126948B2 (en) Pyrimidine-4,6-diamine derivatives as PI3K inhibitors
US11401280B2 (en) Pyrimidinones as PI3K inhibitors
US11130767B2 (en) Bicyclic heteroarylaminoalkyl phenyl derivatives as PI3K inhibitors
US9108984B2 (en) Substituted diamino-pyrimidine and diamino-pyridine derivatives as PI3K inhibitors
US11084822B2 (en) Salts and processes of preparing a PI3K inhibitor
US9944646B2 (en) Bicyclic azaheterocyclobenzylamines as PI3K inhibitors
US9403847B2 (en) Substituted heteroaryl fused derivatives as P13K inhibitors
US9062055B2 (en) Fused pyrrole derivatives as PI3K inhibitors
US20190202840A1 (en) Salts of (s)-7-(1-(9h-purin-6-ylamino)ethyl)-6-(3-fluorophenyl)-3-methyl-5h-thiazolo[3,2-a]pyrimidin-5-one
WO2011130342A1 (en) FUSED DERIVATIVES AS ΡI3Κδ INHIBITORS
IL301180A (en) Use of pyrazolopyrimidine derivatives for the treatment of pi3k related disorders
US9732097B2 (en) Process for the synthesis of a phosphoinositide 3-kinase inhibitor
IL299533A (en) Heterocyclylamines as pi3k inhibitors
US20180258105A1 (en) Crystalline forms of a pi3k inhibitor
BR112017018312B1 (en) PI3K INHIBITOR SALTS, COMPOSITIONS INCLUDING THEM, THEIR USE, PROCESSES FOR THEIR PREPARATION AND METHOD FOR INHIBITING AN ACTIVITY OF A PI3K KINASE

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: 12711541

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 14007061

Country of ref document: US

122 Ep: pct application non-entry in european phase

Ref document number: 12711541

Country of ref document: EP

Kind code of ref document: A1