WO2012097196A1 - Pyrazolopyrimidine derivatives and uses as anticancer agents - Google Patents

Pyrazolopyrimidine derivatives and uses as anticancer agents Download PDF

Info

Publication number
WO2012097196A1
WO2012097196A1 PCT/US2012/021144 US2012021144W WO2012097196A1 WO 2012097196 A1 WO2012097196 A1 WO 2012097196A1 US 2012021144 W US2012021144 W US 2012021144W WO 2012097196 A1 WO2012097196 A1 WO 2012097196A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
alkyl
cancer
ring
haloalkyl
Prior art date
Application number
PCT/US2012/021144
Other languages
French (fr)
Inventor
Long Mao
Li Zhao
Jia Liu
Xiaobo Wang
Xiao Xu
Original Assignee
Acea Biosciences Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Acea Biosciences Inc. filed Critical Acea Biosciences Inc.
Publication of WO2012097196A1 publication Critical patent/WO2012097196A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the field of this invention is pharmaceutical compounds, compositions and methods, especially as they are related to compositions and methods for the treatment of cancer and other diseases related to the dysregulation of kinase (such as Aurora A, Aurora B, Aurora C, cMet, JAK2, ROS1, but not limited to) pathways.
  • kinase such as Aurora A, Aurora B, Aurora C, cMet, JAK2, ROS1, but not limited to
  • Protein kinases are a group of enzymes that regulate diverse, important biological processes including cell growth, proliferation, survival, invasion and differentiation, organ formation, tissue repair and regeneration, etc. Protein kinases exert their physiological functions through catalyzing the phosphorylation of protein and thereby modulating the cellular activities. Because protein kinases have profound effects on cells, their activities are highly regulated. Kinases are turned on or off by phosphorylation (sometimes by
  • cMet is a member of a distinct subfamily of heterodimeric receptor tyrosine kinases which include cMet, Ron and Sea.
  • cMet is the cell surface receptor for hepatocyte growth factor (HGF, also known as scatter factor) that conveys a unique combination of pro-migratory, anti-apoptotic and mitogenic signals. Inappropriate activation of cMet can be induced by specific genetic lesions, transcriptional upregulation or ligand- dependent autocrine or paracrine mechanisms.
  • HGF hepatocyte growth factor
  • cMet inhibitors may be useful in treating diseases such as cancer and other disease related to abnormal cell growth and cMet activation.
  • the present invention is directed to various classes of fused pyrazolopyrimidine derivatives and related fused ring systems described herein, pharmaceutical compositions, and methods of using these compounds and compositions to treat cancer and other diseases related to the dysregulation of kinase (such as Aurora A, Aurora B, Aurora C, cMet, JAK2, ROSl, but not limited to) pathways.
  • kinase such as Aurora A, Aurora B, Aurora C, cMet, JAK2, ROSl, but not limited to
  • These compounds have been shown as described herein to possess inhibition to protein kinases including receptor tyrosine kinases such as those of the MET subfamily, and have anti-cancer activity in cell based assays using MET addicted cell lines, which demonstrate antiproliferative and anti-migration activity against such cells.
  • the compounds and compositions comprising the compounds of the invention are useful to treat conditions characterized by undesired cell proliferation and migration.
  • the compounds are useful to treat the following type of cancer, but not limited to: carcinomas ⁇ e.g. , bladder, breast, cervical, ovarian, gastric, colorectal, esophageal, head and neck, lung, kidney, liver, cholangiocarcinoma, nasopharyngeal, pancreas, prostate, thyroid), soft tissue sarcomas ⁇ e.g. , fibrosarcoma, leiomyosarcoma, Kaposi's sarcoma), musculoskeletal sarcomas ⁇ e.g.
  • rhabdomyosarcoma rhabdomyosarcoma, osteosarcoma, synovial sarcoma
  • hematopoietic malignancies ⁇ e.g. , multiple myeloma, lymphomas, acute myelogenous leukemia, chronic myeloid leukemia, adult T cell leukemia), and other neoplasms ⁇ e.g. , glioblastomas, melanoma, mesothelioma, astrocytomas and Wilms' tumor).
  • the pyrazolopyrimidine or similar heterocyclic moiety of the compounds described herein can be further fused with other aryl/ non-aryl ring or substituted with substituted aryl amino, substituted arylthio, substituted aryloxy, substituted heterocyclic amino, substituted heterocyclic thio, and substituted heterocyclic oxy derivatives thereof.
  • the compounds as described herein exhibit anti-tumor, anticancer, anti-inflammation, anti-infectious, and anti- proliferation activity.
  • the present invention also relates to the methods of making and formulating the described compounds and methods to use them therapeutically.
  • the contemplated heterocyclic compounds have a structure according to Formula la and lb:
  • W 1 , W 2 W 3 , W 4 are each independently absent, N, NH, NR 1 , O, S, CH, or CR 2 ,
  • W 1 , W 2 W 3 , and W 4 is CH or CR 2 ,
  • R' is Ci_8 alkyl, C 2 -8 alkenyl, C 2 -8 alkynyl, aryl, or heteroaryl, wherein each alkyl, alkenyl, alkynyl, aryl and heteroaryl is optionally substituted;
  • Z is S, SO, S0 2 , SO 2 NH, SO 2 NR 3 , NHSO 2 , NR 1 , CR 1 ]* 2 , NR 1 , or O;
  • Het is substituted or unsubstituted Ci -8 alkyl, C 2 -8 alkenyl, C 2 -8 alkynyl, aryl, fused aryl, heteroaryl, fused heterocycle, a saturated or unsaturated carbocyclic ring or a heterocyclic ring, or a group of the formula -(CH 2 ) q -Q where q is 1-4 and Q is an aryl, heteroaryl, cycloalkyl or heterocycle and Q is optionally substituted,
  • each Het is optionally substituted and may contain a heteroatom selected from N, O and S in place of a ring or chain carbon atom of Het;
  • each R x is H or C1-C4 alkyl or C1-C4 haloalkyl, or R can be substituted or unsubstituted Ci_g alkyl, C2-8 alkenyl, C2-8 alkynyl, aryl, fused aryl, heteroaryl, fused heterocycle, a C 3 -8 carbocyclic ring or a C 3 -8 heterocyclic ring, saturated or unsaturated, wherein each Ci -8 alkyl, C 3 -8 cyclic alkyl, C2-8 alkenyl, C2-8 alkynyl can optionally contain a heteroatom selected from N, O and S in place of a carbon atom; p is 0-3;
  • each R 10 is H or acyl
  • R, R , R , R , R , R , or R on the same or adjacent atoms can optionally be linked together to form a 3-8 membered ring that can contain up to two heteroatoms selected from N, O and S as ring members and which is optionally substituted;
  • the invention provides a compound of Formula Ila and lib:
  • W 1 is NR ⁇ O or S
  • W 2 is N or CR 2 ;
  • Ar is a 5-6 membered aryl or heteroaryl ring that can be substituted or unsubstituted
  • R 1 , R 2 , R 10 , Z, Y, A, and Het are as defined for Formula la and lb;
  • the compounds can be of Formula Ilia and Illb
  • W 1 is NR ⁇ O or S
  • Z is S, SO, S0 2 , SO 2 NH, SO 2 NR 3 , NHSO 2 , NR 1 , CR ⁇ 2 NR 1 , or O;
  • Y is -CH 2 - or -CH 2 -CH 2 - or -CH(Me)-;
  • R 10 is H
  • R 2 and Het are as defined for Formula la and lb above;
  • Ph is an optionally substituted phenyl group
  • the invention provides a compound of Formula IVa-d:
  • Het is as defined for Formula la and lb
  • n 1-4
  • each R x is H or C1-C4 alkyl or C1-C4 haloalkyl
  • the invention provides a compound of Formula V:
  • Z is S, SO, S0 2 , Ci ⁇ R 2 NR J R 2 , or O;
  • Het is as defined for Formula la and lb;
  • n 1-4;
  • R 1 , R 2 and R 3 is independently selected from H, OH, Halo, NHR', NR'R',
  • OR', SR', COOR', C( 0)R', CN, CF 3 , OCF 3 , N0 2 , OC(0)R', S0 3 R', P0 3 R' 2 ,
  • each R' is independently selected from H, halo, OR x , SR X , C0 2 R x ,
  • each R x is H or C1-C4 alkyl or C1-C4 haloalkyl
  • compositions comprising a compound of the invention and methods of using these compounds and compositions for treating proliferative disorders such as cancers.
  • Figure 1 shows the TCRP (time-dependent cell response profiling) of c-Met addicted H1993 cells in response to the treatment of various heterocyclic compounds.
  • Figure 2 shows the TCRP (time-dependent cell response profiling) of c-Met addicted GTL16 cells in response to the treatment of various heterocyclic compounds.
  • Figure 3 shows the TCRP (time-dependent cell response profiling) of c-Met-non- addicted A549 cells in response to the treatment of various heterocyclic compounds.
  • Figure 4 shows the impedance response profile of HGF- mediated migration of A431 cells to the treatment of various heterocyclic compounds
  • FIG. 5 shows the impedance response profile of HGF-mediated migration of HUVECs (human umbilical vein endothelial cells) to the treatment of various heterocyclic compounds
  • Figure 6 shows one of the heterocyclic compounds (compoundl3) inhibited cMet phosphorylation in A549 and GTL16 cell lines.
  • Figure 7 shows the inhibitory effects of Compoundl3, compound20, compoundl9 and compound21 on ROS1 phosphorylation in U138MG cells.
  • the invention provides a compound of Formula la and lb:
  • W 1 , W 2 W 3 , W 4 are each independently absent, N, NH, NR 1 , O, S, CH, or CR 2 ,
  • W 1 , W 2 W 3 , and W 4 is CH or CR 2 ,
  • R' is Ci-8 alkyl, C 2 -8 alkenyl, C 2 -8 alkynyl, aryl, or heteroaryl, wherein each alkyl, alkenyl, alkynyl, aryl and heteroaryl is optionally substituted;
  • Z is S, SO, S0 2 , S0 2 NH, S0 2 NR 3 , NHSO 2 , NR 1 , CR 1 ]* 2 , NR 1 , or O;
  • Het is substituted or unsubstituted Ci -8 alkyl, C 2 -8 alkenyl, C 2 -8 alkynyl, aryl, fused aryl, heteroaryl, fused heterocycle, a saturated or unsaturated carbocyclic ring or a heterocyclic ring, or a group of the formula -(CH 2 ) q -Q where q is 1-4 and Q is an aryl, heteroaryl, cycloalkyl or heterocycle and Q is optionally substituted,
  • each Het is optionally substituted and may contain a heteroatom selected from N, O and S in place of a ring or chain carbon atom of Het;
  • each R x is H or C1-C4 alkyl or C1-C4 haloalkyl
  • each R 10 is H or acyl
  • R, R , R , R , R , R , or R on the same or adjacent atoms can optionally be linked together to form a 3-8 membered ring that can contain up to two heteroatoms selected from N, O and S as ring members and which is optionally substituted;
  • W 4 is CR 2 and W 3 is a bond.
  • W 1 is NR 1 .
  • Rl for these compounds is often H or a lower alkyl, e.g. , C1-C4 alkyl (Me, Et). In other embodiments, W 1 is O or S.
  • R 10 is H.
  • R 10 is an acyl group, typically comprising a lower alkyl or lower haloalkyl ⁇ e.g. , up to about 4 carbon atoms).
  • Z is S, SO, S0 2 , S0 2 NH, S0 2 NR 3 , NHS0 2 , NR 1 , CR ⁇ 2 NR 1 , or O.
  • Y is (CH 2 )i_ 4 . In many embodiments, Y is CH 2 , CH(Me) or CH 2 CH 2 .
  • Het is a 5 or 6 membered aromatic or heteroaromatic ring. In some such embodiments, Het is phenyl or pyridyl, and is optionally substituted with up to three R groups as described above. In some of these embodiments, the R substituents are selected from Me, OMe, SMe, CF 3 , CN, and halo.
  • Het is phenyl or pyridyl and is optionally substituted with up to three R groups, often 1-2 R groups.
  • W 4 is CR 2 , wherein R 2 is H, Cl-4 alkyl, or Cl-4 haloalkyl.
  • R' is optionally substituted phenyl.
  • this phenyl is optionally substituted with up to three substituents groups as described herein.
  • the substituents are selected from Me, OMe, SMe, CF 3 , CN, and halo.
  • W 2 is N.
  • the compounds of the invention are of Formula Ila and lib:
  • W 1 is NR ⁇ O or S
  • W 2 is N or CR 2 ;
  • Ar is a 5-6 membered aryl or heteroaryl ring that can be substituted or unsubstituted
  • R 1 , R 2 , R 10 , Z, Y, A, and Het are as defined in Formula la and lb above; or a pharmaceutically acceptable salt thereof.
  • R 10 is H.
  • W 1 is NR ⁇ or W 1 is O or S.
  • W 2 is N or CR 2 .
  • Z is S, SO, S0 2 , SO 2 NH, SO 2 NR 3 , NHSO 2 , NR 1 , C ⁇ R 2 NR 1 , or O;
  • Y is (CH 2 ) 1-4 .
  • Het is a 5 or 6 membered aromatic or heteroaromatic ring; for example, Het is phenyl or pyridyl and is optionally substituted with up to three R groups.
  • R 2 is H, Ci-4 alkyl, or C 1 -4 haloalkyl.
  • Ar is optionally substituted phenyl.
  • Het is one of the following
  • wavy line across one non-cyclic bond indicates the bond that connects each Het group to A; where in a ring indicates the ring is an aromatic or heteroaromatic ring;
  • R can be substituted or unsubstituted Ci -8 alkyl, C 2 -8 alkenyl, C 2 -8 alkynyl;
  • each R 12 is H or C1-C4 alkyl or C1-C4 haloalkyl
  • one or more R groups may be selected from: Ci -8 alkyl, C 3 _8 cyclic alkyl, C 2 -8 alkenyl, C 2 -8 alkynyl, an aryl, heteroaryl, a carbocyclic ring or a heterocyclic ring, each of which may contain a heteroatom selected from N, O and S in place of a carbon atom of the alkyl, alkenyl, alkynyl group.
  • the compounds of Formula Ila and lib may be compounds of Formula Ilia and Illb:
  • Z is S, SO, S0 2 , SO 2 NH, SO 2 NR 3 , NHSO 2 , NR 1 , CR ⁇ 2 NR 1 , or O;
  • Y is -CH 2 - or -CH 2 -CH 2 - or -CH(Me)-;
  • R 1 and R 2 are as defined for Formula la or lb above;
  • R 10 is H
  • Ph is an optionally substituted phenyl group
  • Het may be one of the following groups:
  • wavy line across one non-cyclic bond indicates the bond that connects each Het group to A; where in a ring indicates the ring is an aromatic or heteroaromatic ring;
  • each W and W is independently a bond, C, CH, CH 2 , CHR 12 , CR 12 , N, NH, NR 12 , S, O,
  • R can be substituted or unsubstituted Ci -8 alkyl, C2-8 alkenyl, C2-8 alkynyl;
  • each R 12 is H or C1-C4 alkyl or C1-C4 haloalkyl.
  • the invention provides a compound of Formula IVa-d:
  • Het is as defined for Formula la and lb
  • n 1-4
  • each R x is H or C1-C4 alkyl or C1-C4 haloalkyl, or a pharmaceutically acceptable salt thereof.
  • Het may be one of the following groups:
  • wavy line across one non-cyclic bond indicates the bond that connects each Het group to A; where in a ring indicates the ring is an aromatic or heteroaromatic ring;
  • R can be substituted or unsubstituted Ci -8 alkyl, C2-8 alkenyl, C2-8 alkynyl;
  • each R 12 is H or C1-C4 alkyl or C1-C4 haloalkyl.
  • the invention provides a compound of Formula Va or Vb:
  • Z is S, SO, S0 2 , Ci ⁇ R 2 NR J R 2 , or O;
  • Het is as defined for Formula la or lb;
  • n 1-4;
  • each R x is H or C1-C4 alkyl or C1-C4 haloalkyl
  • Het may be one of the following groups:
  • R can be substituted or unsubstituted Ci -8 alkyl, C2-8 alkenyl, C2-8 alkynyl;
  • each R 12 is H or C1-C4 alkyl or C1-C4 haloalkyl.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound according to any of the foregoing embodiments admixed with at least one pharmaceutically acceptable carrier or excipient. Suitable carriers and excipients are described herein.
  • this pharmaceutical composition comprises at least one sterile pharmaceutically acceptable carrier or excipient.
  • the composition comprises at least two pharmaceutically acceptable carriers and/or excipients.
  • the invention provides a compound according to any one of the foregoing embodiments for use in therapy.
  • the compound is for use in therapy to treat cancer, e.g. , a cancer selected from leukemia, lymphoma, lung cancer, colon cancer, CNS cancer, melanoma, ovarian cancer, renal cancer, prostate cancer, breast cancer, head and neck cancers, and pancreatic cancer.
  • the invention provides a method to treat cancer, which comprises administering to a subject in need thereof an effective amount of a compound according to any of the foregoing embodiments, or a pharmaceutical composition comprising one or more of such compounds.
  • the cancer is selected from leukemia, lymphoma, lung cancer, colon cancer, CNS cancer, melanoma, ovarian cancer, renal cancer, prostate cancer, breast cancer, head and neck cancers, and pancreatic cancer.
  • the invention provides use of a compound according to any one of the foregoing embodiments for the manufacture of a medicament.
  • the medicament is one for treating cancer, and in some embodiments the cancer is selected from leukemia, lymphoma, lung cancer, colon cancer, CNS cancer, melanoma, ovarian cancer, renal cancer, prostate cancer, breast cancer, head and neck cancers, and pancreatic cancer.
  • Het may be mono-/di-/tri-substituted or unsubstituted benzene, pyridine, pyridazine, pyrimidine, pyrazine, triazine, oxazole, isoxazole, thiazole, isothiazole, oxadiazole, triazole, thiadiazole, pyrazole, imidazole, benzoxazole, pyrrole, furan, thiophene, indolizine, indole, isoindole, indoline, benzofuran, benzothiophene, indazole, benzimidazole, benzthiazole, purine, quinoxaline, quinoline, isoquinoline, cinnoline, phthalazine, quinazoline, naphthyridine, pteridine, acridine, phenazine,
  • each ring of these compounds will be a 5-6 atom ring, whether aromatic or not, and will contain at least one carbon atom, no more than one O or S atom (except for a dioxane ring having two O atoms), and up to four N atoms as ring members.
  • Non-aromatic rings will typically contain no more than two heteroatoms in place of ring carbon atoms, while aromatic rings containing 3-4 heteroatoms— especially N atoms— such as triazines, triazoles, tetrazines and tetrazoles, are included.
  • other groups such as R would be selected to avoid compounds generally considered to be too reactive for use as pharmaceuticals, e.g. , R would not be Halo in groups such as NHR, NRR, OR, SR, or COOR.
  • Suitable dosages, routes of administration, and administration protocols can be selected by a skilled person based on the information herein.
  • alkyl refers to saturated hydrocarbon groups in a straight, branched, or cyclic configuration or any combination thereof, and particularly contemplated alkyl groups include those having ten or less carbon atoms, especially 1-6 carbon atoms and lower alkyl groups having 1-4 carbon atoms.
  • Exemplary alkyl groups are methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, tertiary butyl, pentyl, isopentyl, hexyl, cyclopropylmethyl, etc.
  • Alkyl groups can be unsubstituted, or they can be substituted to the extent that such substitution makes sense chemically.
  • Alkyl, alkenyl and alkynyl groups can also be substituted by C1-C8 acyl, C2-C8 heteroacyl, C6-C10 aryl or C5-C10 heteroaryl, each of which can be substituted by the substituents that are appropriate for the particular group.
  • a substituent group contains two R a or R b groups on the same or adjacent atoms (e.g., -NR b 2, or -NR b -C(0) R b )
  • the two R a or R b groups can optionally be taken together with the atoms in the substituent group to which the are attached to form a ring having 5-8 ring members, which can be substituted as allowed for the R a or R b itself, and can contain an additional heteroatom (N, O or S) as a ring member.
  • alkenyl refers to an alkyl as defined above having at least two carbon atoms and at least one carbon-carbon double bond.
  • alkenyl groups include straight, branched, or cyclic alkenyl groups having two to ten carbon atoms (e.g., ethenyl, propenyl, butenyl, pentenyl, etc.) or 5-10 atoms for cyclic alkenyl groups.
  • Alkenyl groups are optionally substituted by groups suitable for alkyl groups as set forth herein.
  • alkynyl refers to an alkyl or alkenyl as defined above and having at least two (preferably three) carbon atoms and at least one carbon-carbon triple bond.
  • alkynyls include straight, branched, or cyclic alkynes having two to ten total carbon atoms (e.g. , ethynyl, propynyl, butynyl, cyclopropylethynyl, etc.).
  • Alkynyl groups are optionally substituted by groups suitable for alkyl groups as set forth herein.
  • cycloalkyl refers to a cyclic alkane (i.e. , in which a chain of carbon atoms of a hydrocarbon forms a ring), preferably including three to eight carbon atoms.
  • exemplary cycloalkanes include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl.
  • Cycloalkyls also include one or two double bonds, which form the "cycloalkenyl” groups. Cycloalkyl groups are optionally substituted by groups suitable for alkyl groups as set forth herein.
  • aryl or "aromatic moiety” as used herein refers to an aromatic ring system, which may further include one or more non-carbon atoms. These are typically 5-6 membered isolated rings, or 8-10 membered bicyclic groups, and can be substituted. Thus, contemplated aryl groups include (e.g., phenyl, naphthyl, etc.) and pyridyl. Further,
  • aryl groups may be fused (i.e., covalently bound with 2 atoms on the first aromatic ring) with one or two 5- or 6-membered aryl or heterocyclic group, and are thus termed "fused aryl” or "fused aromatic".
  • Aromatic groups containing one or more heteroatoms (typically N, O or S) as ring members can be referred to as heteroaryl or heteroaromatic groups.
  • Typical heteroaromatic groups include monocyclic C5-C6 aromatic groups such as pyridyl, pyrimidyl, pyrazinyl, thienyl, furanyl, pyrrolyl, pyrazolyl, thiazolyl, oxazolyl, isothiazolyl, isoxazolyl, and imidazolyl and the fused bicyclic moieties formed by fusing one of these monocyclic groups with a phenyl ring or with any of the heteroaromatic monocyclic groups to form a C8-C10 bicyclic group such as indolyl, benzimidazolyl, indazolyl, benzotriazolyl, isoquinolyl, quinolyl, benzothiazolyl, benzofuranyl, pyrazolopy
  • any monocyclic or fused ring bicyclic system which has the characteristics of aromaticity in terms of electron distribution throughout the ring system is included in this definition. It also includes bicyclic groups where at least the ring which is directly attached to the remainder of the molecule has the characteristics of aromaticity. Typically, the ring systems contain 5- 12 ring member atoms.
  • heterocycle As also used herein, the terms “heterocycle”, “cycloheteroalkyl”, and “heterocyclic moieties” are used interchangeably herein and refer to any compound in which a plurality of atoms form a ring via a plurality of covalent bonds, wherein the ring includes at least one atom other than a carbon atom as a ring member.
  • Particularly contemplated heterocyclic rings include 5- and 6-membered rings with nitrogen, sulfur, or oxygen as the non-carbon atom (e.g. , imidazole, pyrrole, triazole, dihydropyrimidine, indole, pyridine, thiazole, tetrazole etc.).
  • these rings typically contain 0-1 oxygen or sulfur atoms, at least one and typically 2-3 carbon atoms, and up to four nitrogen atoms as ring members.
  • heterocycles may be fused (i.e., covalently bound with two atoms on the first heterocyclic ring) to one or two carbocyclic rings or heterocycles, and are thus termed "fused heterocycle" or "fused
  • heterocyclic ring or “fused heterocyclic moieties” as used herein. Where the ring is aromatic, these can be referred to herein as 'heteroaryl' or heteroaromatic groups.
  • Heterocyclic groups that are not aromatic can be substituted with groups suitable for alkyl group substituents, as set forth above.
  • Aryl and heteroaryl groups can be substituted where permitted. Suitable substituents include, but are not limited to, halo, -OR a , -NR a 2 , -SR a , -S0 2 R a , -S0 2 NR a 2 , -NR a S0 2 R a , -NR a CONR a 2 , -NR a COOR a , -NR a COR a , -CN, -COOR a , -CONR a 2 , -OOCR a , -COR a , and -N0 2 , wherein each R a is independently H, Ci-C 8 alkyl, C2-C8 heteroalkyl, C3-C8 heterocyclyl, C4-C10 heterocyclyclalkyl, C1-C8 acyl, C2-C8 heteroacyl, C2-C8 alkenyl, C2-C8
  • Alkyl, alkenyl and alkynyl groups can also be substituted by C1-C8 acyl, C2-C8 heteroacyl, C6-C10 aryl or C5-C10 heteroaryl, each of which can be substituted by the substituents that are appropriate for the particular group.
  • a substituent group contains two R a or R b groups on the same or adjacent atoms (e.g., -NR b 2, or -NR b -C(0) R b )
  • the two R a or R b groups can optionally be taken together with the atoms in the substituent group to which the are attached to
  • the terms "imidazopyridine” or “imidazopyrimidine” or “thiazopyridine” or “thiazopyrimidine” herein refer to any compound in which the two designated heterocyclic rings are fused by any two adjacent atoms on the two heterocyclic rings.
  • alkoxy refers to a hydrocarbon group connected through an oxygen atom, e.g., -O-Hc, wherein the hydrocarbon portion He may have any number of carbon atoms, typically 1-10 carbon atoms, may further include a double or triple bond and may include one or two oxygen, sulfur or nitrogen atoms in the alkyl chains, and can be substituted with aryl, heteroaryl, cycloalkyl, and/or heterocyclyl groups.
  • suitable alkoxy groups include methoxy, ethoxy, propyloxy, isopropoxy, methoxyethoxy, benzyloxy, allyloxy, and the like.
  • alkylthio refers to alkylsulfides of the general formula -S- Hc, wherein the hydrocarbon portion He is as described for alkoxy groups.
  • contemplated alkylthio groups include methylthio, ethylthio, isopropylthio, methoxyethylthio, benzylthio, allylthio, and the like.
  • alkylamino refers to amino groups where one or both hydrogen atoms are replaced by a hydrocarbon group He as described above, wherein the amino nitrogen "N” can be substituted by one or two He groups as set forth for alkoxy groups described above.
  • exemplary alkylamino groups include methylamino, dimethylamino, ethylamino, diethylamino, etc.
  • D can be H, Me, Et, isopropyl, propyl, butyl, C1-C4 alkyl substituted with -OH, -OMe, or N3 ⁇ 4, phenyl, halophenyl, alkylphenyl, and the like.
  • aryloxy refers to an aryl group connecting to an oxygen atom, wherein the aryl group may be further substituted.
  • suitable aryloxy groups include phenyloxy, etc.
  • arylthio refers to an aryl group connecting to a sulfur atom, wherein the aryl group may be further substituted.
  • suitable arylthio groups include phenylthio, etc.
  • hydrocarbon portion of each alkoxy, alkylthio, alkylamino, and aryloxy, etc. can be substituted as appropriate for the relevant hydrocarbon moiety.
  • halogen refers to fluorine, chlorine, bromine and iodine. Where present as a substituent group, halogen or halo typically refers to F or CI or Br, more typically F or CI.
  • substituents may further be substituted with one or more substituents, which may in turn be substituted with hydroxy, amino, cyano, C1-C4 alkyl, halo, or C1-C4 haloalkyl.
  • a hydrogen atom in an alkyl or aryl can be replaced by an amino, halo or Ci- 4 haloalkyl or alkyl group.
  • substituted refers to a replacement of a hydrogen atom of the unsubstituted group with a functional group
  • functional groups include nucleophilic groups (e.g., -NH 2 , -OH, -SH, -CN, etc.), electrophilic groups (e.g. , C(0)OR, C(X)OH, etc.), polar groups (e.g. , -OH), non-polar groups (e.g. , heterocycle, aryl, alkyl, alkenyl, alkynyl, etc.), ionic groups (e.g. , -NH 3 "1" ), and halogens (e.g.
  • substituted also includes multiple degrees of substitution, and where multiple substituents are disclosed or claimed, the substituted compound can be independently substituted by one or more of the disclosed or claimed substituent moieties.
  • Any formula given herein is intended to represent compounds having structures depicted by the structural formula as well as certain variations or forms.
  • compounds of any formula given herein may have asymmetric centers and therefore exist in different enantiomeric forms. All optical isomers and stereoisomers of the compounds of the general formula, and mixtures thereof, are considered within the scope of the formula.
  • any formula given herein is intended to represent a racemate, one or more enantiomeric forms, one or more diastereomeric forms, one or more atropisomeric forms, and mixtures thereof.
  • certain structures may exist as geometric isomers (i.e., cis and trans isomers), as tautomers, or as atropisomers.
  • any formula given herein is intended to refer also to any one of hydrates, solvates, and polymorphs of such compounds, and mixtures thereof, even if such forms are not listed explicitly.
  • the solvent is water and the solvates are hydrates.
  • any formula given herein is also intended to represent unlabeled forms as well as isotopically labeled forms of the compounds.
  • Isotopically labeled compounds have structures depicted by the formulas given herein except that one or more atoms are replaced by an atom having a selected atomic mass or mass number.
  • isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, chlorine, and iodine, such as 2 H, 3 H, n C, 13 C, 14 C, 15 N, 18 0, 17 0, 31 P, 32 P, 35 S, 18 F, 36 C1, and 125 I, respectively.
  • Such isotopically labelled compounds are useful in metabolic studies (preferably with 14 C), reaction kinetic studies (with, for example 2 H or 3 H), detection or imaging techniques [such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT)] including drug or substrate tissue distribution assays, or in radioactive treatment of patients.
  • PET positron emission tomography
  • SPECT single-photon emission computed tomography
  • an 18 F or 11 C labeled compound may be particularly preferred for PET or SPECT studies.
  • substitution with heavier isotopes such as deuterium (i.e., 2 H) may afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements.
  • Isotopically labeled compounds of this invention and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the schemes or in the examples and preparations described below by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent.
  • Ci_3 or C1-C3 refers independently to embodiments that have one carbon member (Ci or CI), embodiments that have two carbon members (C 2 or C2), and embodiments that have three carbon members (C 3 or C3).
  • any disubstituent referred to herein is meant to encompass the various attachment possibilities when more than one of such possibilities are allowed.
  • reference to disubstituent -A-B-, where A ⁇ B, refers herein to such disubstituent with A attached to a first substituted member and B attached to a second substituted member, and it also refers to such disubstituent with A attached to the second substituted member and B attached to the first substituted member.
  • the invention also includes pharmaceutically acceptable salts of the compounds represented by Formula I-V, preferably of those described above and of the specific compounds exemplified herein, and pharmaceutical compositions comprising such salts, and methods of using such salts.
  • a "pharmaceutically acceptable salt” is intended to mean a salt of a free acid or base of a compound represented herein that is non-toxic, biologically tolerable, or otherwise biologically suitable for administration to the subject. See, generally, S.M. Berge, et al., "Pharmaceutical Salts," J. Pharm. Sci., 1977, 66, 1-19.
  • Preferred pharmaceutically acceptable salts are those that are pharmacologically effective and suitable for contact with the tissues of subjects without undue toxicity, irritation, or allergic response.
  • a compound described herein may possess a sufficiently acidic group, a sufficiently basic group, both types of functional groups, or more than one of each type, and accordingly react with a number of inorganic or organic bases, and inorganic and organic acids, to form a pharmaceutically acceptable salt.
  • Examples of pharmaceutically acceptable salts include sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, phosphates, monohydrogen-phosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates, caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyne-l,4-dioates, hexyne-l,6-dioates, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, hydroxybenzoates, methoxybenzoates, phthalates, sulfonates, methylsulfonates,
  • propylsulfonates besylates, xylenesulfonates, naphthalene- 1- sulfonates, naphthalene-2- sulfonates, phenylacetates, phenylpropionates, phenylbutyrates, citrates, lactates, ⁇ - hydroxybutyrates, glycolates, tartrates, and mandelates.
  • a pharmaceutically acceptable salt may be prepared by any suitable method available in the art, for example, treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, sulfamic acid, nitric acid, boric acid, phosphoric acid, and the like, or with an organic acid, such as acetic acid, phenylacetic acid, propionic acid, stearic acid, lactic acid, ascorbic acid, maleic acid, hydroxymaleic acid, isethionic acid, succinic acid, valeric acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, oleic acid, palmitic acid, lauric acid, a pyranosidyl acid, such as glucuronic acid or galacturonic acid, an alpha-hydroxy acid, such as mandelic acid, citric acid, or
  • an inorganic acid such as hydrochloric acid, hydrobromic acid
  • the invention also relates to pharmaceutically acceptable prodrugs of the compounds of Formula I-V, and treatment methods employing such pharmaceutically acceptable prodrugs.
  • prodrug means a precursor of a designated compound that, following administration to a subject, yields the compound in vivo via a chemical or physiological process such as solvolysis or enzymatic cleavage, or under physiological conditions (e.g., a prodrug on being brought to physiological pH is converted to the compound of Formula I- V).
  • a “pharmaceutically acceptable prodrug” is a prodrug that is non-toxic, biologically tolerable, and otherwise biologically suitable for administration to the subject. Illustrative procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in "Design of Prodrugs", ed. H. Bundgaard, Elsevier, 1985.
  • the present invention also relates to pharmaceutically active metabolites of compounds of Formula I-V, and uses of such metabolites in the methods of the invention.
  • a "pharmaceutically active metabolite” means a pharmacologically active product of metabolism in the body of a compound of Formulas I-V or salt thereof.
  • Prodrugs and active metabolites of a compound may be determined using routine techniques known or available in the art. See, e.g., Bertolini et al., /. Med. Chem. 1997, 40, 2011-2016; Shan et al., /. Pharm. Sci. 1997, 86 (7), 765-767; Bagshawe, Drug Dev. Res. 1995, 34, 220-230; Bodor, Adv. Drug Res. 1984, 13, 255- 331; Bundgaard, Design of Prodrugs (Elsevier Press, 1985); and Larsen, Design and
  • Any suitable formulation of the compounds described herein can be prepared. See generally, Remington's Pharmaceutical Sciences, (2000) Hoover, J. E. editor, 20 th edition, Lippincott Williams and Wilkins Publishing Company, Easton, Pa., pages 780-857.
  • a formulation is selected to be suitable for an appropriate route of administration. Some routes of administration are oral, parenteral, by inhalation, topical, rectal, nasal, buccal, vaginal, via an implanted reservoir, or other drug administration methods. In cases where compounds are sufficiently basic or acidic to form stable nontoxic acid or base salts, administration of the compounds as salts may be appropriate.
  • Examples of pharmaceutically acceptable salts are organic acid addition salts formed with acids that form a physiological acceptable anion, for example, tosylate, methanesulfonate, acetate, citrate, malonate, tartarate, succinate, benzoate, ascorbate, a-ketoglutarate, and a-glycerophosphate.
  • Suitable inorganic salts may also be formed, including hydrochloride, sulfate, nitrate, bicarbonate, and carbonate salts.
  • salts are obtained using standard procedures well known in the art, for example, by a sufficiently basic compound such as an amine with a suitable acid, affording a physiologically acceptable anion.
  • a sufficiently basic compound such as an amine with a suitable acid, affording a physiologically acceptable anion.
  • Alkali metal e.g. , sodium, potassium or lithium
  • alkaline earth metal e.g. , calcium
  • contemplated compounds are administered in a pharmacological composition
  • the compounds can be formulated in admixture with a pharmaceutically acceptable excipient and/or carrier.
  • contemplated compounds can be administered orally as neutral compounds or as pharmaceutically acceptable salts, or intravenously in a physiological saline solution.
  • Conventional buffers such as phosphates, bicarbonates or citrates can be used for this purpose.
  • one of ordinary skill in the art may modify the formulations within the teachings of the specification to provide numerous formulations for a particular route of administration.
  • contemplated compounds may be modified to render them more soluble in water or other vehicle, which for example, may be easily accomplished with minor modifications (salt formulation, esterification, etc.) that are well within the ordinary skill in the art. It is also well within the ordinary skill of the art to modify the route of administration and dosage regimen of a particular compound in order to manage the pharmacokinetics of the present compounds for maximum beneficial effect in a patient.
  • the compounds having formula I-V as described herein are generally soluble in organic solvents such as chloroform, dichloromethane, ethyl acetate, ethanol, methanol, isopropanol, acetonitrile, glycerol, N,N-dimethylformamide, N,N-dimetheylaceatmide, dimethylsulfoxide, etc.
  • organic solvents such as chloroform, dichloromethane, ethyl acetate, ethanol, methanol, isopropanol, acetonitrile, glycerol, N,N-dimethylformamide, N,N-dimetheylaceatmide, dimethylsulfoxide, etc.
  • the present invention provides formulations prepared by mixing a compound having formula I-V with a pharmaceutically acceptable carrier.
  • the formulation may be prepared using a method comprising: a) dissolving a described compound in a water-soluble organic solvent, a non-ionic solvent, a water-soluble lipid, a cyclodextrin, a vitamin such as tocopherol, a fatty acid, a fatty acid ester, a
  • the phospholipid or a combination thereof, to provide a solution; and b) adding saline or a buffer containing 1-10% carbohydrate solution.
  • the carbohydrate comprises dextrose.
  • Illustrative examples of water soluble organic solvents for use in the present methods include and are not limited to polyethylene glycol (PEG), alcohols, acetonitrile, N-methyl-2- pyrrolidone, N,N-dimethylformamide, N,N-dimethylacetamide, dimethyl sulfoxide, or a combination thereof.
  • PEG polyethylene glycol
  • alcohols include but are not limited to methanol, ethanol, isopropanol, glycerol, or propylene glycol.
  • Illustrative examples of water soluble non-ionic surfactants for use in the present methods include and are not limited to CREMOPHOR ® EL, polyethylene glycol modified CREMOPHOR ® (polyoxyethyleneglyceroltriricinoleat 35), hydrogenated CREMOPHOR ® RH40, hydrogenated CREMOPHOR ® RH60, PEG-succinate, polysorbate 20, polysorbate 80, SOLUTOL ® HS (polyethylene glycol 660 12-hydroxystearate), sorbitan monooleate, poloxamer, LABRAFIL ® (ethoxylated persic oil), LABRASOL ® (capryl-caproyl macrogol-8- glyceride), GELUCIRE ® (glycerol ester), SOFTIGEN ® (PEG 6 caprylic glyceride), glycerin, glycol-polysorbate, or a combination thereof.
  • lipid oils include but are not limited to castor oil, polyoxyl castor oil, corn oil, olive oil, cottonseed oil, peanut oil, peppermint oil, safflower oil, sesame oil, soybean oil, hydrogenated vegetable oil, hydrogenated soybean oil, a triglyceride of coconut oil, palm seed oil, and hydrogenated forms thereof, or a combination thereof.
  • Illustrative examples of fatty acids and fatty acid esters for use in the present methods include but are not limited to oleic acid, monoglycerides, diglycerides, a mono- or di- fatty acid ester of PEG, or a combination thereof.
  • cyclodextrins for use in the present methods include but are not limited to alpha-cyclodextrin, beta-cyclodextrin, hydroxypropyl-beta-cyclodextrin, or sulfobutyl ether-beta-cyclodextrin.
  • Illustrative examples of phospholipids for use in the present methods include but are not limited to soy phosphatidylcholine, or distearoyl phosphatidylglycerol, and hydrogenated forms thereof, or a combination thereof.
  • One of ordinary skill in the art may modify the formulations within the teachings of the specification to provide numerous formulations for a particular route of administration.
  • the compounds may be modified to render them more soluble in water or other vehicle. It is also well within the ordinary skill of the art to modify the route of administration and dosage regimen of a particular compound in order to manage the pharmacokinetics of the present compounds for maximum beneficial effect in a patient.
  • compositions for the treatment of a cell proliferative disorder comprising any compound having formula I-V,
  • compounds having formula I-V and pharmaceutical compositions thereof may be administered orally, parenterally, by inhalation, topically, rectally, nasally, buccally, vaginally, via an implanted reservoir, or other drug administration methods.
  • parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intraarticular, intraarterial, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.
  • a sterile injectable composition such as a sterile injectable aqueous or oleaginous suspension, may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent.
  • acceptable vehicles and solvents include mannitol, water, Ringer' s solution and isotonic sodium chloride solution.
  • Suitable carriers and other pharmaceutical composition components are typically sterile.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium (e.g. , synthetic mono- or diglycerides).
  • Fatty acids such as oleic acid and its glyceride derivatives, are useful in the preparation of injectables, as are pharmaceutically acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions can also contain a long-chain alcohol diluent or dispersant, or carboxymethyl cellulose or similar dispersing agents.
  • Various emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms can also be used for the purpose of formulation.
  • a composition for oral administration may be any orally acceptable dosage form including, but not limited to, tablets, capsules, emulsions and aqueous suspensions, dispersions and solutions.
  • commonly used carriers include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, can also be added.
  • useful diluents include lactose and dried corn starch.
  • a nasal aerosol or inhalation compositions can be prepared according to techniques well-known in the art of pharmaceutical formulation and can be prepared as solutions in, for example saline, employing suitable preservatives (for example, benzyl alcohol), absorption promoters to enhance bioavailability, and/or other solubilizing or dispersing agents known in the art.
  • suitable preservatives for example, benzyl alcohol
  • absorption promoters to enhance bioavailability
  • other solubilizing or dispersing agents known in the art.
  • the compounds having formula I-V may be administered alone or in combination with other anticancer agents for the treatment of various cancers or conditions.
  • Combination therapies according to the present invention comprise the administration of at least one compound of the present invention or a functional derivative thereof and at least one other pharmaceutically active ingredient.
  • the active ingredient(s) and pharmaceutically active agents may be administered separately or together.
  • the amounts of the active ingredient(s) and pharmaceutically active agent(s) and the relative timings of administration will be selected in order to achieve the desired combined therapeutic effect.
  • a compound of formula I-V can besides or in addition be administered especially for tumor therapy in combination with chemotherapy, radiotherapy, immunotherapy, surgical intervention, or a combination of these. Long-term therapy and adjuvant therapy in
  • the compounds and compositions described herein can be administered to a subject in need of treatment for a cell proliferation disorder such as cancer, particularly cancers selected from leukemia, lymphoma, lung cancer, colon cancer, CNS cancer, melanoma, ovarian cancer, renal cancer, prostate cancer, breast cancer, head and neck cancers, and pancreatic cancer.
  • a cell proliferation disorder such as cancer, particularly cancers selected from leukemia, lymphoma, lung cancer, colon cancer, CNS cancer, melanoma, ovarian cancer, renal cancer, prostate cancer, breast cancer, head and neck cancers, and pancreatic cancer.
  • the subject is typically a mammal diagnosed as being in need of treatment for one or more of such proliferative disorders, and frequently the subject is a human.
  • the methods comprise administering an effective amount of at least one compound of the invention; optionally the compound may be administered in combination with one or more additional therapeutic agents, particularly therapeutic agents known to be useful for treating the cancer or proliferative disorder afflicting the particular subject.
  • treat or “treating” as used herein is intended to refer to administration of a compound of the invention to a subject for the purpose of creating a therapeutic benefit to a subject suffering from or diagnosed with cancer or cell proliferation disease, disorder or condition. Treating includes preventing, reversing, ameliorating, alleviating, inhibiting the progress of, or lessening the severity of, a cancer or cell proliferation disorder or one or more symptoms of that disorder, as well as the delay of progression of a disease, disorder or condition.
  • subject refers to a mammalian patient in need of such treatment, such as a human.
  • an "effective amount” means an amount sufficient to reduce, slow the progression of, or reverse cancer or cell proliferation disorder. Measuring the amount of cell proliferation may be performed by routine analytical methods such as those described below. Such modulation is useful in a variety of settings, including in vitro assays.
  • the cell is preferably a GTL-16, H1993 or A431 cells.
  • an "effective amount” means an amount or dose sufficient to generally bring about the desired therapeutic benefit in subjects needing such treatment.
  • Effective amounts or doses of the compounds of the invention may be ascertained by routine methods, such as modeling, dose escalation, or clinical trials, taking into account routine factors, e.g., the mode or route of administration or drug delivery, the pharmacokinetics of the agent, the severity and course of the infection, the subject's health status, condition, and weight, and the judgment of the treating physician.
  • the dose may be adjusted for preventative or maintenance treatment.
  • the dosage or the frequency of administration, or both may be reduced as a function of the symptoms, to a level at which the desired therapeutic or prophylactic effect is maintained.
  • treatment may cease. Patients may, however, require intermittent treatment on a long-term basis upon any recurrence of symptoms. Patients may also require chronic treatment on a long-term basis.
  • combination refers to either a fixed combination in one dosage unit form, or a kit of parts for the combined administration where a compound of the formula (I) and a combination partner (e.g. an other drug as explained below, also referred to as “therapeutic agent” or “co-agent”) may be administered independently at the same time or separately within time intervals, especially where these time intervals allow that the combination partners show a cooperative, e.g. synergistic effect.
  • a combination partner e.g. an other drug as explained below, also referred to as “therapeutic agent” or “co-agent”
  • co-administration or “combined administration” or the like as utilized herein are meant to encompass administration of the selected combination partner to a single subject in need thereof (e.g. a patient), and are intended to include treatment regimens in which the agents are not necessarily administered by the same route of
  • the term "pharmaceutical combination” as used herein means a product that results from the mixing or combining of more than one active ingredient and includes both fixed and non-fixed combinations of the active ingredients.
  • the term "fixed combination” means that the active ingredients, e.g. a compound of formula (I) and a combination partner, are both administered to a patient simultaneously in the form of a single entity or dosage.
  • the term “non-fixed combination” means that the active ingredients, e.g. a compound of formula (I) and a combination partner, are both administered to a patient as separate entities either simultaneously, concurrently or sequentially with no specific time limits, wherein such administration provides therapeutically effective levels of the two compounds in the body of the patient.
  • cocktail therapy e.g. the administration of three or more active ingredients.
  • inventive compounds described herein may be used in pharmaceutical compositions or methods in combination with one or more additional active ingredients in the treatment of cancer, cell proliferative disorders or inflammatory conditions.
  • additional active ingredients are those that are known or discovered to be effective in treating cancer, cell proliferative disorders or inflammatory conditions, including those active against another target associated with the disease, or a combination thereof. Such a combination may serve to increase efficacy, ameliorate other disease symptoms, decrease one or more side effects, or decrease the required dose of an inventive compound.
  • the additional active ingredients may be administered in a separate pharmaceutical composition from a compound of the present invention or may be included with a compound of the present invention in a single pharmaceutical composition.
  • the additional active ingredients may be administered simultaneously with, prior to, or after administration of a compound of the present invention.
  • a compound of the formula I-V may be used in combination with other antiproliferative compounds.
  • antiproliferative compounds include, but are not limited to aromatase inhibitors; antiestrogens; topoisomerase I inhibitors; topoisomerase II inhibitors; microtubule active compounds; alkylating compounds; histone deacetylase inhibitors;
  • chemotherapeutic compounds include, but are not limited to, plant alkaloids, hormonal compounds and antagonists; biological response modifiers, preferably lymphokines or interferons; antisense oligonucleotides or oligonucleotide derivatives; shRNA or siRNA; or miscellaneous compounds or compounds with other or unknown mechanism of action.
  • Cyclo A substituted benzenes, pyrindines, heterocyclic compounds
  • Cyclo A substituted benzenes, pyrindines, heterocyclic amines
  • R 1 , R 2 , R 3 , R H, Me, Et, i-Pr, Propyl, Butyl, F, Br, CI, 1, OMe, NMe 2 , NEt 2 , N0 2 , SMe, SEt, CF 3 , CH 2 COOEt, cyclopropyl, NHS0 2 R, alkenyl, alkynyl, saturated/ unsaturated carbocyclic or heterocyclic ring (fused or not fused), etc.
  • R and R' are as set forth for Formula I.
  • Scheme lb Representative substituted and fused pyrazolo pyrimidine embodiments.
  • Step 1 Methyl 5-methylnicotinate (10 mmol, 1.52g) was dissolved in 50 Ml dried THF, then cooled to 0 deg with ice-bath. LiAlH4 (16 mmol, 0.61g) was added in small portions during about 20 minutes. The mixture stirred for another 20 minutes until the reaction was completed, which monitored by TLC. 1.5 mL water was dropwised into the mixture to quench the reaction, then filtered, solution was concentrated to get the (5-methylpyridin-3-yl)methanol, Yield was high to 85%
  • Step 2 Alcohol (10 mmol) were refluxed in 10ml HBr (40% aq) for more than 5 hours, and monitored through TLC or LC-MS. After completed, mixture were heated in order to evaporator solvents (water and excess HBr) until the mixture became sticky. Cool the mixture, add acetone to the mixture, precipitated solid and filtered followed by drying. Yield was high to
  • Compounds prepared according to the schemes described above may be obtained as single enantiomers, diastereomers, or regioisomers, by enantio-, diastero-, or regiospecific synthesis, or by resolution.
  • Compounds prepared according to the schemes above may alternately be obtained as racemic (1 : 1) or non-racemic (not 1 : 1) mixtures or as mixtures of diastereomers or regioisomers.
  • single enantiomers may be isolated using conventional separation methods known to one skilled in the art, such as chiral chromatography, recrystallization, diastereomeric salt formation, derivatization into diastereomeric adducts, biotransformation, or enzymatic transformation.
  • separation methods known to one skilled in the art, such as chiral chromatography, recrystallization, diastereomeric salt formation, derivatization into diastereomeric adducts, biotransformation, or enzymatic transformation.
  • regioisomeric or diastereomeric mixtures are obtained, single isomers may be separated using conventional methods such as chromatography or crystallization.
  • Example 11 Synthesis of 5-amino-3-bromo-lH-pyrazole-4-carbonitrile (42) and 5-amino-3- chloro-lH-pyrazole-4-carbonitrile (43) and analogs A537 (44), A542 (45), A547 (46), A558 (47):
  • heterocyclic compounds in the present invention are developed for the anticancer activities for cancer cells with certain molecular targets, i.e. , cMet.
  • the anticancer efficacy of these heterocyclic compounds and their analogues described above were:
  • RT-CES real time electronic cell sensing
  • microelectrodes having appropriate geometries are fabricated onto the bottom surfaces of microtiter plate or similar device, facing into the wells. Cells are introduced into the wells of the devices, and make contact to and attach to the electrode surfaces. The presence, absence or change of properties of cells affects the electronic and ionic passage on the electrode sensor surfaces. Measuring the impedance between or among electrodes provides important information about biological status of cells present on the sensors. When there are changes to the biological status of the cells analogue, electronic readout signals are measured automatically and in real time, and are converted to digital signals for processing and analysis.
  • a cell index is automatically derived and provided based on measured electrode impedance values.
  • the cell index obtained for a given well reflects: 1) how many cells are attached to the electrode surfaces in this well; 2) how well cells are attached to the electrode surfaces in this well. Thus, the more the cells of same type in similar
  • Impedance- based TCRP time-dependent cell response profiling
  • Oncogene addiction refers to the acquired dependency of cancer cells on a single cellular pathway for survival or sustained proliferation, despite the fact that such cells have accumulated numerous genetic alterations.
  • H1993 and GTL16 have previously been reported as cMET addicted cell lines (Benvenuti, S., et ah, Cancer Res. (2011) 71 : 1945-1955). We used these two cell lines to assay specific inhibitory effects of the heterocyclic compounds described in the examples above on cMet.
  • H1993 and GTL16 were seeded into wells of 96 well E-plate devices (Roche) with initial seeding density of 10,000 cells per well and were pre-incubated in incubator under standard cell culture condition for about 24 hours. The heterocyclic compounds at different concentration in DMSO were added into wells following the incubation period. Negative control wells were treated with buffer solutions containing DMSO only.
  • Figure 1 shows the TCRP (time-dependent cell response profiling) of c-Met addicted HI 993 cells to the treatment of various heterocyclic compounds.
  • the dose-dependent rapid impedance (corresponding to normalized cell index in the y-axis) increase and TCRP are the same as those observed for H1993 cells when treated with cMet inhibitor control compounds (results not shown).
  • the cell index was normalized against the cell index values at a time point just before compound addition.
  • the calculated effective concentrations EC50 (2 hr post compound treatment) in this specific experiment is 220 nM, 179 nM, 217 nM and 154 nM for compound A424 (19),
  • Figure 2 shows the TCRP (time-dependent cell response profiling) of c-Met addicted GTL16 cells to the treatment of various heterocyclic compounds.
  • TCRP TCRP are the same as those observed for GTL16 cells when treated with cMet inhibitor control compounds (results not shown).
  • the calculated effective concentrations EC50 (2 hr post compound treatment) in this specific experiment is 137 nM, 141 nM, 138 nM and 109 nM for compound A424 (19), compound A418 (20), compound A447 (21) and compound A448 (22), respectively.
  • the EC50 calculated based on the short-term TCRP correlates well with the IC 50 calculated based other cell based assay (EXAMPLE 17).
  • Results for the compounds tested in these assays are presented in Table 1-11 as an average of results obtained.
  • the EC5 0 S (in H1993) toward cMet inhibition for selected compounds of this invention are listed in Tables 1-11.
  • Compounds were tested in free base form. Where activity is shown as greater than (>) a particular value, the value is the highest concentration tested.
  • RT-CES real-time cell electronic sensing
  • A549 cells were treated with various heterocyclic compounds described above.
  • A549 were seeded into wells of 96 well E-plate devices (Roche) with initial seeding density of 5,000 cells per well and were pre-incubated in incubator under standard cell culture condition for about 24 hours.
  • the heterocyclic compounds at different concentration in DMSO were added into wells following the incubation period. Negative control wells were treated with buffer solutions containing DMSO only.
  • Figure 3 shows the TCRP of cMet-non- addicted A549 cells to the treatment of various heterocyclic compounds.
  • A431 cells as determined using real-time cell electronic sensing (RT-CES) system
  • HGF-mediated migration of A431 cells was performed in the presence of different heterocyclic compounds.
  • the technical details, instrument, devices of cell migration/invasion assays with real-time cell electronic sensing technology can be found in United States patent number 7,459,303. Briefly, A431 cells were starved for 6 hr and then incubated with compounds for 1 hr before seeding to fibronectin precoated CIM device (Roche). HGF at 50 ng/ml was used to mediate migration of A431 from upper chamber to lower chamber. The time course of the migration was monitored.
  • Table 12 below shows IC 50 values derived from the impedance response profile of HGF-mediated migration of A431 cells in the presence of different compounds described in the present invention.
  • Figure 4 shows that these heterocyclic compounds inhibited HGF-mediated A431 migration in a dose-dependent manner.
  • the positive control compound, PF02341066 (HGFi) inhibited A431 migration in a similar dose-dependent manner.
  • HGFR cMet
  • HUVECs as determined using real-time cell electronic sensing (RT-CES) system
  • HGF-mediated migration of HUVECs was performed in the presence of a series of concentrations of different heterocyclic compounds.
  • the technical details, instrument, devices of cell migration/invasion assays with real-time cell electronic sensing technology can be found in United States patent number 7,459,303. Briefly, HUVECs were starved for 6 hr and then incubated with compounds for 1 hr before seeding to fibronectin precoated CIM device (Roche). HGF at 50 ng/ml was used to mediate migration of HUVECs from upper chamber to lower chamber. The time course of the migration was monitored.
  • Table 13 below lists IC50 values derived from the impedance response profile of HGF-mediated migration of HUVECs in the presence of different compounds described in the present invention.
  • Figure 5 shows that these heterocyclic compounds inhibited HGF-mediated HUVECs migration in a dose-dependent manner.
  • the positive control compound, PF02341066 (HGFi) inhibited HUVECs migration in a similar dose-dependent manner.
  • HGFR cMet
  • Example 17 Heterocyclic compounds show inhibitory effect on cMet phosphorylation both in vivo and in vitro
  • the calculated IC 50 is 38.8 nM, 74.7 nM and 36.2 nM for compound A 447 (21), compound A418 (20) and compound A424 (19) and, respectively. Furthermore, we tested the activities of the heterocyclic compounds on the enzymatic activities of purified cMet protein (experiments were performed at Reaction Biology Corporation). At 10 ⁇ ATP concentration, the IC 50 is 494.9 nM, 10.8 nM, 4.01 nM and 1.21 nM for compound A348 (13), compound A447 (21), compound A418 (20) and compound A424 (19), respectively. The potency ranking agrees well with that derived from our RT-CES based assay. Taken together, there is strong evidence that the heterocyclic compounds described herein are cMet inhibitors.
  • KINOMEscan is the industry's most comprehensive high-throughput system for screening compounds against large number of human kinases (Fabian et al. (2005) Nat.
  • ROSl transcripts have been detected in several glioblastoma cell lines, e.g., U-118 MG, U-138 MG and SW-1088, but not in A-172 and A-382 (Fabian et al. (1987) Proc. Nati. Acad. Sci. USA 84, 9270-9274).
  • the results from Western analyses confirm that ROSl was expressed in U138 MG, not in A-172.
  • the Western results show that at 10 ⁇ concentration, compound A348 (13), compound A418 (20), compound A424 (19) and compound A447 (21) inhibited ROSl phosphorylation after 1 hour compound treatment (Figure 7).

Abstract

Novel compounds having a fused pyrazolopyrimidine derivatives and related fused ring systems are disclosed. The compounds inhibit growth of a variety of types of cancer cells, and are thus useful for treating cancer. Pharmaceutical compositions, and methods of using these compounds and compositions to treat cancer and other diseases related to the dysregulation of kinase (such as Aurora A, Aurora B, Aurora C, cMet, JAK2, ROS1, but not limited to) pathways are disclosed. Efficacy of these compounds is demonstrated with a system for monitoring cell growth/migration, which shows they are potent inhibitors of growth and/or migration of cancer cells. Compositions comprising these compounds, and methods to use these compounds and compositions for treatment of cancers, are disclosed.

Description

PYRAZOLOPYRIMIDINE DERIVATIVES AND USES AS
ANTICANCER AGENTS
Cross-reference to related applications
[0001] This application claims priority to U.S. Provisional Patent Application Nos.
61/432,164, filed January 12, 2011, the disclosure of which is hereby incorporated herein by reference in their entirety.
Field of the Invention
[0002] The field of this invention is pharmaceutical compounds, compositions and methods, especially as they are related to compositions and methods for the treatment of cancer and other diseases related to the dysregulation of kinase (such as Aurora A, Aurora B, Aurora C, cMet, JAK2, ROS1, but not limited to) pathways.
Background of the Invention
[0003] Protein kinases are a group of enzymes that regulate diverse, important biological processes including cell growth, proliferation, survival, invasion and differentiation, organ formation, tissue repair and regeneration, etc. Protein kinases exert their physiological functions through catalyzing the phosphorylation of protein and thereby modulating the cellular activities. Because protein kinases have profound effects on cells, their activities are highly regulated. Kinases are turned on or off by phosphorylation (sometimes by
autophosphorylation), by binding of activator proteins or inhibitor proteins, or small molecules, or by controlling their location in the cell relative to their substrates. Dysfunctions in the activities of kinases, arising from genetic abnormalities or environmental factors, are known to be associated with many diseases. Several severe pathological states, including cancer and chronic inflammation, are associated with stimulation of intra-cellular signaling, and since kinases positively relay signaling events, their inhibition offers a powerful way to inhibit or control signal transduction cascades.
[0004] The proto-oncogene cMet is a member of a distinct subfamily of heterodimeric receptor tyrosine kinases which include cMet, Ron and Sea. cMet is the cell surface receptor for hepatocyte growth factor (HGF, also known as scatter factor) that conveys a unique combination of pro-migratory, anti-apoptotic and mitogenic signals. Inappropriate activation of cMet can be induced by specific genetic lesions, transcriptional upregulation or ligand- dependent autocrine or paracrine mechanisms. Amplification of the MET gene, with consequent protein overexpression and constitutive kinase activation, has been reported in a number of human primary tumors, including non-small-cell lung (NSCL) carcinomas, gastric and esophageal carcinomas with acquired resistance to epidermal growth factor receptor (EGFR) inhibitors and medulloblastomas. The MET gene can also carry activating mutations, such as those found in a subset of patients with hereditary and sporadic papillary renal cancer, gastric carcinoma and childhood hepatocellular carcinoma. HGF can also aberrantly activate cMet in an autocrine fashion, as described for glioblastomas, breast carcinomas, osteosarcomas and rhabdomyosarcomas. (Comoglio, P. M., et ah, Nature Reviews Drug Discovery (2008) 7:504- 516) Therefore cMet inhibitors may be useful in treating diseases such as cancer and other disease related to abnormal cell growth and cMet activation.
Summary of the Inventions
[0005] The present invention is directed to various classes of fused pyrazolopyrimidine derivatives and related fused ring systems described herein, pharmaceutical compositions, and methods of using these compounds and compositions to treat cancer and other diseases related to the dysregulation of kinase (such as Aurora A, Aurora B, Aurora C, cMet, JAK2, ROSl, but not limited to) pathways. These compounds have been shown as described herein to possess inhibition to protein kinases including receptor tyrosine kinases such as those of the MET subfamily, and have anti-cancer activity in cell based assays using MET addicted cell lines, which demonstrate antiproliferative and anti-migration activity against such cells. Accordingly, the compounds and compositions comprising the compounds of the invention are useful to treat conditions characterized by undesired cell proliferation and migration. In particular, the compounds are useful to treat the following type of cancer, but not limited to: carcinomas {e.g. , bladder, breast, cervical, ovarian, gastric, colorectal, esophageal, head and neck, lung, kidney, liver, cholangiocarcinoma, nasopharyngeal, pancreas, prostate, thyroid), soft tissue sarcomas {e.g. , fibrosarcoma, leiomyosarcoma, Kaposi's sarcoma), musculoskeletal sarcomas {e.g. , rhabdomyosarcoma, osteosarcoma, synovial sarcoma), hematopoietic malignancies {e.g. , multiple myeloma, lymphomas, acute myelogenous leukemia, chronic myeloid leukemia, adult T cell leukemia), and other neoplasms {e.g. , glioblastomas, melanoma, mesothelioma, astrocytomas and Wilms' tumor).
[0006] The pyrazolopyrimidine or similar heterocyclic moiety of the compounds described herein can be further fused with other aryl/ non-aryl ring or substituted with substituted aryl amino, substituted arylthio, substituted aryloxy, substituted heterocyclic amino, substituted heterocyclic thio, and substituted heterocyclic oxy derivatives thereof. The compounds as described herein exhibit anti-tumor, anticancer, anti-inflammation, anti-infectious, and anti- proliferation activity. The present invention also relates to the methods of making and formulating the described compounds and methods to use them therapeutically.
[0007] In one aspect, the contemplated heterocyclic compounds have a structure according to Formula la and lb:
Figure imgf000004_0001
Formula la Formula lb where in a ring indicates the ring is an aromatic or heteroaromatic ring;
W1, W2 W3 , W4 are each independently absent, N, NH, NR1, O, S, CH, or CR2,
provided at least one of W1, W2 W3 , and W4 is CH or CR2,
not more than one of them is absent,
and not more than one of them is O or S;
R' is Ci_8 alkyl, C2-8 alkenyl, C2-8 alkynyl, aryl, or heteroaryl, wherein each alkyl, alkenyl, alkynyl, aryl and heteroaryl is optionally substituted;
Z is S, SO, S02, SO2NH, SO2NR3, NHSO2, NR1, CR1]*2 , NR1, or O;
Y is (CHR4)n wherein n = 1-5 or a C3-C7 carbocyclic or aromatic divalent moiety;
A is a bond (absent), C(=0), NH, NR5, C(0)NH, C(0)NR6, C(S)NH, C(S)NR6, NHC(O), NR6C(0), NHSO2, NR6S02, SO2NH, SO2NR6, or OP(=0)(OR7);
Het is substituted or unsubstituted Ci-8 alkyl, C2-8 alkenyl, C2-8 alkynyl, aryl, fused aryl, heteroaryl, fused heterocycle, a saturated or unsaturated carbocyclic ring or a heterocyclic ring, or a group of the formula -(CH2)q-Q where q is 1-4 and Q is an aryl, heteroaryl, cycloalkyl or heterocycle and Q is optionally substituted,
where each Het is optionally substituted and may contain a heteroatom selected from N, O and S in place of a ring or chain carbon atom of Het;
each of R1, R2, R3, R4, R5, R6, and R7 is independently selected from H, OH, Halo, NHR, NRR, OR, SR, COOR, C(=0)R, CN, CF3, OCF3, N02, OC(0)R, S03R, PO3R2,
CR(COOR)2;
each R is independently selected from H, halo, ORx, SRX, C02Rx, C(0)NRx 2, C(=0) Rx, CN, CF3, OCF3, N02, NRX2, OCORx, S03Rx, P03Rx 2, and CH(COORx)2;
where each Rx is H or C1-C4 alkyl or C1-C4 haloalkyl, or R can be substituted or unsubstituted Ci_g alkyl, C2-8 alkenyl, C2-8 alkynyl, aryl, fused aryl, heteroaryl, fused heterocycle, a C3-8 carbocyclic ring or a C3-8 heterocyclic ring, saturated or unsaturated, wherein each Ci-8 alkyl, C3-8 cyclic alkyl, C2-8 alkenyl, C2-8 alkynyl can optionally contain a heteroatom selected from N, O and S in place of a carbon atom; p is 0-3;
each R10 is H or acyl;
1 2 3 4 5 6 V
and two R, R , R , R , R , R , R , or R on the same or adjacent atoms can optionally be linked together to form a 3-8 membered ring that can contain up to two heteroatoms selected from N, O and S as ring members and which is optionally substituted;
or a pharmaceutically acceptable salt thereof.
[0008] In another aspect, the invention provides a compound of Formula Ila and lib:
A com ound of Formula Ila and lib:
Figure imgf000005_0001
Ila lib
wherein:
W1 is NR\ O or S;
W2 is N or CR2;
Ar is a 5-6 membered aryl or heteroaryl ring that can be substituted or unsubstituted; and
R1, R2, R10, Z, Y, A, and Het are as defined for Formula la and lb;
or a pharmaceutically acceptable salt thereof.
[0009] In another embodiment the compounds can be of Formula Ilia and Illb
Figure imgf000005_0002
wherein: W1 is NR\ O or S;
Z is S, SO, S02, SO2NH, SO2NR3, NHSO2, NR1, CR^2 NR1, or O; Y is -CH2- or -CH2-CH2- or -CH(Me)-;
A is a bond or C(=0);
R10 is H;
Figure imgf000006_0001
R2 and Het are as defined for Formula la and lb above;
Ph is an optionally substituted phenyl group;
or a pharmaceutically acceptable salt thereof.
[0010] In a further embodiment, the invention provides a compound of Formula IVa-d:
Figure imgf000006_0002
Wherein:
Het is as defined for Formula la and lb
n is 1-4
R1, R2 and R3 is independently selected from H, OH, Halo, NHR' , NR'R' , OR' , SR', COOR', C(=0)R' , CN, CF3, OCF3, N02, OC(0)R', S03R' , P03R'2,
CR'(COOR')2
each R' is independently selected from H, halo, ORx, SRX, C02Rx, C(0)NRX2, C(=0) Rx, CN, CF3, OCF3, N02, NRX 2, OCORx, S03Rx, P03Rx 2, and
CH(COORx)2;
where each Rx is H or C1-C4 alkyl or C1-C4 haloalkyl,
or a pharmaceutically acceptable salt thereof. [0011] In another aspect, the invention provides a compound of Formula V:
A compound of Formula Va or Vb:
Figure imgf000007_0001
Formula Va Formula Vb
Wherein:
Z is S, SO, S02, Ci^R2 NRJR2, or O;
A is a bond, C=0, CONH or NHCO;
Het is as defined for Formula la and lb;
n is 1-4;
R1, R2 and R3 is independently selected from H, OH, Halo, NHR', NR'R',
OR', SR', COOR', C(=0)R', CN, CF3, OCF3, N02, OC(0)R', S03R', P03R'2,
CR'(COOR')2;
each R' is independently selected from H, halo, ORx, SRX, C02Rx,
C(0)NRX2, C(=0) Rx, CN, CF3, OCF3, N02, NRX 2, OCORx, S03Rx, P03Rx 2, and
CH(COORx)2;
where each Rx is H or C1-C4 alkyl or C1-C4 haloalkyl;
or a pharmaceutically acceptable salt thereof.
[0012] Other aspects of the invention provide pharmaceutical compositions comprising a compound of the invention and methods of using these compounds and compositions for treating proliferative disorders such as cancers.
Brief Description of the Figures
[0013] Figure 1 shows the TCRP (time-dependent cell response profiling) of c-Met addicted H1993 cells in response to the treatment of various heterocyclic compounds.
[0014] Figure 2 shows the TCRP (time-dependent cell response profiling) of c-Met addicted GTL16 cells in response to the treatment of various heterocyclic compounds. [0015] Figure 3 shows the TCRP (time-dependent cell response profiling) of c-Met-non- addicted A549 cells in response to the treatment of various heterocyclic compounds.
[0016] Figure 4 shows the impedance response profile of HGF- mediated migration of A431 cells to the treatment of various heterocyclic compounds
[0017] Figure 5 shows the impedance response profile of HGF-mediated migration of HUVECs (human umbilical vein endothelial cells) to the treatment of various heterocyclic compounds
[0018] Figure 6 shows one of the heterocyclic compounds (compoundl3) inhibited cMet phosphorylation in A549 and GTL16 cell lines.
[0019] Figure 7 shows the inhibitory effects of Compoundl3, compound20, compoundl9 and compound21 on ROS1 phosphorylation in U138MG cells.
Detailed Description
[0020] In one aspect, the invention provides a compound of Formula la and lb:
Figure imgf000008_0001
Formula la Formula lb where in a ring indicates the ring is an aromatic or heteroaromatic ring;
W1, W2 W3 , W4 are each independently absent, N, NH, NR1, O, S, CH, or CR2,
provided at least one of W1, W2 W3 , and W4 is CH or CR2,
not more than one of them is absent,
and not more than one of them is O or S;
R' is Ci-8 alkyl, C2-8 alkenyl, C2-8 alkynyl, aryl, or heteroaryl, wherein each alkyl, alkenyl, alkynyl, aryl and heteroaryl is optionally substituted;
Z is S, SO, S02, S02NH, S02NR3, NHSO2, NR1, CR1]*2 , NR1, or O;
Y is (CHR4)n wherein n = 1-5 or a C3-C7 carbocyclic or aromatic divalent moiety;
A is a bond (absent), C(=0), NH, NR5, C(0)NH, C(0)NR6, C(S)NH, C(S)NR6, NHC(O), NR6C(0), NHSO2, NR6S02, S02NH, S02NR6, or OP(=0)(OR7);
Het is substituted or unsubstituted Ci-8 alkyl, C2-8 alkenyl, C2-8 alkynyl, aryl, fused aryl, heteroaryl, fused heterocycle, a saturated or unsaturated carbocyclic ring or a heterocyclic ring, or a group of the formula -(CH2)q-Q where q is 1-4 and Q is an aryl, heteroaryl, cycloalkyl or heterocycle and Q is optionally substituted,
where each Het is optionally substituted and may contain a heteroatom selected from N, O and S in place of a ring or chain carbon atom of Het;
each of R1, R2, R3, R4, R5, R6, and R7 is independently selected from H, OH, Halo, NHR, NRR, OR, SR, COOR, C(=0)R, CN, CF3, OCF3, N02, OC(0)R, S03R, P03R2,
CR(COOR)2;
each R is independently selected from H, halo, ORx, SRX, C02Rx, C(0)NRx 2, C(=0) Rx, CN, CF3, OCF3, N02, NRX2, OCORx, S03Rx, P03Rx 2, and CH(COORx)2;
where each Rx is H or C1-C4 alkyl or C1-C4 haloalkyl,
or R can be substituted or unsubstituted Ci-8 alkyl, C2_8 alkenyl, C2_8 alkynyl, aryl, fused aryl, heteroaryl, fused heterocycle, a C3_8 carbocyclic ring or a C3_8 heterocyclic ring, saturated or unsaturated, wherein each Ci-8 alkyl, C3_8 cyclic alkyl, C2_8 alkenyl, C2_8 alkynyl can optionally contain a heteroatom selected from N, O and S in place of a carbon atom; p is 0-3;
each R10 is H or acyl;
1 2 3 4 5 6 V
and two R, R , R , R , R , R , R , or R on the same or adjacent atoms can optionally be linked together to form a 3-8 membered ring that can contain up to two heteroatoms selected from N, O and S as ring members and which is optionally substituted;
or a pharmaceutically acceptable salt thereof.
[0021] In some embodiments of these compounds, W4 is CR2 and W3 is a bond.
[0022] In some embodiments of any of the foregoing compounds, W1 is NR1. Rl for these compounds is often H or a lower alkyl, e.g. , C1-C4 alkyl (Me, Et). In other embodiments, W1 is O or S.
[0023] In some embodiments of any of the foregoing compounds R10 is H. In other embodiments, R10 is an acyl group, typically comprising a lower alkyl or lower haloalkyl {e.g. , up to about 4 carbon atoms).
[0024] In some embodiments of any of the foregoing compounds, Z is S, SO, S02, S02NH, S02NR3, NHS02, NR1, CR^2 NR1, or O.
[0025] In some embodiments of any of the foregoing compounds, Y is (CH2)i_4. In many embodiments, Y is CH2, CH(Me) or CH2CH2.
[0026] In some embodiments of any of the foregoing compounds, A is a bond or C(=0). [0027] In some embodiments of any of the foregoing compounds, Het is a 5 or 6 membered aromatic or heteroaromatic ring. In some such embodiments, Het is phenyl or pyridyl, and is optionally substituted with up to three R groups as described above. In some of these embodiments, the R substituents are selected from Me, OMe, SMe, CF3, CN, and halo.
[0028] In some embodiments of any of the foregoing compounds, Het is phenyl or pyridyl and is optionally substituted with up to three R groups, often 1-2 R groups.
[0029] In some embodiments of any of the compounds of Formula I, W4 is CR2, wherein R2 is H, Cl-4 alkyl, or Cl-4 haloalkyl.
[0030] In certain embodiments of any of the foregoing compounds, R' is optionally substituted phenyl. Typically this phenyl is optionally substituted with up to three substituents groups as described herein. In some of these embodiments, the substituents are selected from Me, OMe, SMe, CF3, CN, and halo.
[0031] In certain embodiments of any of the foregoing compounds, W2 is N.
[0032] In another as ect, the compounds of the invention are of Formula Ila and lib:
Figure imgf000010_0001
Ila lib
wherein:
W1 is NR\ O or S;
W2 is N or CR2;
Ar is a 5-6 membered aryl or heteroaryl ring that can be substituted or unsubstituted; and
R1, R2, R10, Z, Y, A, and Het are as defined in Formula la and lb above; or a pharmaceutically acceptable salt thereof.
[0033] In the compounds of Formula Ila and lib, any or all of the following further selections may be utilized:
R10 is H.
W1 is NR^ or W1 is O or S.
W2 is N or CR2.
Z is S, SO, S02, SO2NH, SO2NR3, NHSO2, NR1, C^R2 NR1, or O;
Y is (CH2)1-4. A is a bond or C(=0).
Het is a 5 or 6 membered aromatic or heteroaromatic ring; for example, Het is phenyl or pyridyl and is optionally substituted with up to three R groups.
R2 is H, Ci-4 alkyl, or C1-4 haloalkyl.
Ar is optionally substituted phenyl.
[0034] In some embodiments of the compounds of Formula Ila and lib, Het is one of the following
Figure imgf000011_0001
wherein the wavy line across one non-cyclic bond indicates the bond that connects each Het group to A; where in a ring indicates the ring is an aromatic or heteroaromatic ring;
each W and W is independently a bond, C, CH, CH2, CHR12, CR12, N, NH, NR12, S, O, C=0, or C=S,
where each R is independently selected from H, halo, ORx, SRX, C02Rx, C(0)NRx 2, C(=0) Rx, CN, CF3, OCF3, N02, NRX2, OCORx, S03Rx, P03Rx 2, and CH(COORx)2, where each Rx is H or C1-C4 alkyl or C1-C4 haloalkyl,
or R can be substituted or unsubstituted Ci-8 alkyl, C2-8 alkenyl, C2-8 alkynyl;
and each R12 is H or C1-C4 alkyl or C1-C4 haloalkyl,
or a pharmaceutically acceptable salt thereof.
[0035] In these embodiments, one or more R groups may be selected from: H, halo, ORx, S(0)tRx, C02Rx, C(0)NRX2, C(=0) Rx, CN, CF3, OCF3, N02, NRX 2, OCORx, S03Rx, P03Rx 2, CH(COORx)2, where each Rx is H or C1-C4 alkyl or C1-C4 haloalkyl, and each t is 0-2.
[0036] In these embodiments, one or more R groups may be selected from: Ci-8 alkyl, C3_8 cyclic alkyl, C2-8 alkenyl, C2-8 alkynyl, an aryl, heteroaryl, a carbocyclic ring or a heterocyclic ring, each of which may contain a heteroatom selected from N, O and S in place of a carbon atom of the alkyl, alkenyl, alkynyl group.
[0037] In another aspect, the compounds of Formula Ila and lib may be compounds of Formula Ilia and Illb:
Figure imgf000012_0001
wherein:
Figure imgf000012_0002
Z is S, SO, S02, SO2NH, SO2NR3, NHSO2, NR1, CR^2 NR1, or O; Y is -CH2- or -CH2-CH2- or -CH(Me)-;
A is a bond or C(=0);
Het, R1 and R2 are as defined for Formula la or lb above;
R10 is H;
Ph is an optionally substituted phenyl group;
or a pharmaceutically acceptable salt thereof.
[0038] In the com ounds of Formula Ilia and Illb, Het may be one of the following groups:
Figure imgf000012_0003
wherein the wavy line across one non-cyclic bond indicates the bond that connects each Het group to A; where in a ring indicates the ring is an aromatic or heteroaromatic ring;
each W and W is independently a bond, C, CH, CH2, CHR12, CR12, N, NH, NR12, S, O,
C=0, or C=S, where each R is independently selected from H, halo, ORx, SRX, C02Rx, C(0)NRx 2, C(=0) Rx, CN, CF3, OCF3, N02, NRX2, OCORx, S03Rx, P03Rx 2, and CH(COORx)2, where each Rx is H or C1-C4 alkyl or C1-C4 haloalkyl,
or R can be substituted or unsubstituted Ci-8 alkyl, C2-8 alkenyl, C2-8 alkynyl;
and each R12 is H or C1-C4 alkyl or C1-C4 haloalkyl.
[0039] An embodiment of Formula Ilia and Illb may be where Het is phenyl or pyridyl, it is sometimes optionally substituted with up to three groups selected from C1-C4 alkyl, C1-C4 haloalkyl, halo, ORx, SRX, C02Rx, C(0)NRx 2, C(=0) Rx, CN, CF3, OCF3, N02, NRX 2, OCORx, S03Rx, P03RX2, and CH(COORx)2, where each Rx is H or C1-C4 alkyl or C1-C4 haloalkyl.
[0040] In a further embodiment, the invention provides a compound of Formula IVa-d:
Figure imgf000013_0001
wherein:
Het is as defined for Formula la and lb
n is 1-4
R1, R2 and R3 is independently selected from H, OH, Halo, NHR' , NR'R' , OR' , SR', COOR', C(=0)R\ CN, CF3, OCF3, N02, OC(0)R', S03R' , P03R'2,
CR'(COOR')2
each R' is independently selected from H, halo, ORx, SRX, C02Rx, C(0)NRX2, C(=0) Rx, CN, CF3, OCF3, N02, NRX 2, OCORx, S03Rx, P03Rx 2, and
CH(COORx)2;
where each Rx is H or C1-C4 alkyl or C1-C4 haloalkyl, or a pharmaceutically acceptable salt thereof.
[0041] In the compounds of Formula IVa, IVb, IVc or IVd, Het may be one of the following groups:
Figure imgf000014_0001
wherein the wavy line across one non-cyclic bond indicates the bond that connects each Het group to A; where in a ring indicates the ring is an aromatic or heteroaromatic ring;
each W and W is independently a bond, C, CH, CH2, CHR12, CR12, N, NH, NR12, S, O, C=0, or C=S,
where each R is independently selected from H, halo, ORx, SRX, C02Rx, C(0)NRx 2, C(=0) Rx, CN, CF3, OCF3, N02, NRX2, OCORx, S03Rx, P03Rx 2, and CH(COORx)2, where each Rx is H or C1-C4 alkyl or C1-C4 haloalkyl,
or R can be substituted or unsubstituted Ci-8 alkyl, C2-8 alkenyl, C2-8 alkynyl;
and each R12 is H or C1-C4 alkyl or C1-C4 haloalkyl.
[0042] An embodiment of Formula IVa, IVb, IVc or IVd may be where Het is phenyl or pyridyl, it is sometimes optionally substituted with up to three groups selected from C1-C4 alkyl, C1-C4 haloalkyl, halo, ORx, SRX, C02Rx, C(0)NRx 2, C(=0) Rx, CN, CF3, OCF3, N02, NRX2, OCORx, S03Rx, P03RX2, and CH(COORx)2, where each Rx is H or C1-C4 alkyl or C1-C4 haloalkyl.
[0043] In another aspect, the invention provides a compound of Formula Va or Vb:
A compound of Formula Va or Vb:
Figure imgf000015_0001
Wherein:
Z is S, SO, S02, Ci^R2 NRJR2, or O;
A is a bond, C=0, CONH or NHCO;
Het is as defined for Formula la or lb;
n is 1-4;
R1, R2 and R3 is independently selected from H, OH, Halo, NHR' , NR'R' , OR' , SR', COOR', C(=0)R' , CN, CF3, OCF3, N02, OC(0)R', S03R' , P03R'2,
CR'(COOR')2;
each R' is independently selected from H, halo, ORx, SRX, C02Rx, C(0)NRX2, C(=0) Rx, CN, CF3, OCF3, N02, NRX 2, OCORx, S03Rx, P03Rx 2, and
CH(COORx)2;
where each Rx is H or C1-C4 alkyl or C1-C4 haloalkyl;
or a pharmaceutically acceptable salt thereof.
[0044] In the compounds of Formula Va or Vb, Het may be one of the following groups:
Figure imgf000015_0002
wherein the wavy line across one non-cyclic bond indicates the bond that connects each Het group to A; where ι--' in a ring indicates the ring is an aromatic or heteroaromatic ring;
each W and W is independently a bond, C, CH, CH2, CHR12, CR12, N, NH, NR12, S, O, C=0, or C=S,
where each R is independently selected from H, halo, ORx, SRX, C02Rx, C(0)NRx 2, C(=0) Rx, CN, CF3, OCF3, N02, NRX2, OCORx, S03Rx, P03Rx 2, and CH(COORx)2, where each Rx is H or C1-C4 alkyl or C1-C4 haloalkyl,
or R can be substituted or unsubstituted Ci-8 alkyl, C2-8 alkenyl, C2-8 alkynyl;
and each R12 is H or C1-C4 alkyl or C1-C4 haloalkyl.
[0045] An embodiment of Formula Va or Vb may be where Het is phenyl or pyridyl, it is sometimes optionally substituted with up to three groups selected from C1-C4 alkyl, C1-C4 haloalkyl, halo, ORx, SRX, C02Rx, C(0)NRx 2, C(=0) Rx, CN, CF3, OCF3, N02, NRX 2, OCORx, S03Rx, P03Rx 2, and CH(COORx)2, where each Rx is H or C1-C4 alkyl or C1-C4 haloalkyl.
[0046] In another aspect, the invention provides a pharmaceutical composition comprising a compound according to any of the foregoing embodiments admixed with at least one pharmaceutically acceptable carrier or excipient. Suitable carriers and excipients are described herein. In some embodiments, this pharmaceutical composition comprises at least one sterile pharmaceutically acceptable carrier or excipient. In some embodiments, the composition comprises at least two pharmaceutically acceptable carriers and/or excipients.
[0047] In another aspect, the invention provides a compound according to any one of the foregoing embodiments for use in therapy. In particular embodiments of interest, the compound is for use in therapy to treat cancer, e.g. , a cancer selected from leukemia, lymphoma, lung cancer, colon cancer, CNS cancer, melanoma, ovarian cancer, renal cancer, prostate cancer, breast cancer, head and neck cancers, and pancreatic cancer.
[0048] In one aspect, the invention provides a method to treat cancer, which comprises administering to a subject in need thereof an effective amount of a compound according to any of the foregoing embodiments, or a pharmaceutical composition comprising one or more of such compounds. In some embodiments, the cancer is selected from leukemia, lymphoma, lung cancer, colon cancer, CNS cancer, melanoma, ovarian cancer, renal cancer, prostate cancer, breast cancer, head and neck cancers, and pancreatic cancer.
[0049] Similarly, the invention provides use of a compound according to any one of the foregoing embodiments for the manufacture of a medicament. In some embodiments, the medicament is one for treating cancer, and in some embodiments the cancer is selected from leukemia, lymphoma, lung cancer, colon cancer, CNS cancer, melanoma, ovarian cancer, renal cancer, prostate cancer, breast cancer, head and neck cancers, and pancreatic cancer.
[0050] In the above formula I - V, "Het" may be mono-/di-/tri-substituted or unsubstituted benzene, pyridine, pyridazine, pyrimidine, pyrazine, triazine, oxazole, isoxazole, thiazole, isothiazole, oxadiazole, triazole, thiadiazole, pyrazole, imidazole, benzoxazole, pyrrole, furan, thiophene, indolizine, indole, isoindole, indoline, benzofuran, benzothiophene, indazole, benzimidazole, benzthiazole, purine, quinoxaline, quinoline, isoquinoline, cinnoline, phthalazine, quinazoline, naphthyridine, pteridine, acridine, phenazine, phenothiazine, indene, naphthalene or benzoxadiazole. Illustrated examples of "Het", include but are not limited to the fo
Figure imgf000017_0001
[0051] It will be understood that the selections of W1, W2, W3, W4, W and Z for any of the above cyclic groups would of course be made by a person of ordinary skill in the art in accordance with well known limitations associated with valence rules and stability. These atoms or bonds would thus be selected to provide only a stable ring or ring system consistent with well-known bonding and valence principles; thus these rings would not be constructed with -O-O- or -S-S- linkages in the rings, for example, or with inappropriate numbers of O or N or S atoms causing instability in aqueous media. Typically each ring of these compounds will be a 5-6 atom ring, whether aromatic or not, and will contain at least one carbon atom, no more than one O or S atom (except for a dioxane ring having two O atoms), and up to four N atoms as ring members. Non-aromatic rings will typically contain no more than two heteroatoms in place of ring carbon atoms, while aromatic rings containing 3-4 heteroatoms— especially N atoms— such as triazines, triazoles, tetrazines and tetrazoles, are included. Likewise, other groups such as R would be selected to avoid compounds generally considered to be too reactive for use as pharmaceuticals, e.g. , R would not be Halo in groups such as NHR, NRR, OR, SR, or COOR. [0052] Suitable dosages, routes of administration, and administration protocols can be selected by a skilled person based on the information herein.
General Definitions:
[0053] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of ordinary skill in the art to which this invention belongs. All patents, applications, published applications and other publications referred to herein are incorporated by reference in their entireties. If a definition set forth in this section is contrary to or otherwise inconsistent with a definition set forth in a patent, application, or other publication that is herein incorporated by reference, the definition set forth in this section prevails over the definition incorporated herein by reference.
[0054] As used herein, "a" or "an" means "at least one" or "one or more".
[0055] The term "alkyl" as used herein refers to saturated hydrocarbon groups in a straight, branched, or cyclic configuration or any combination thereof, and particularly contemplated alkyl groups include those having ten or less carbon atoms, especially 1-6 carbon atoms and lower alkyl groups having 1-4 carbon atoms. Exemplary alkyl groups are methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, tertiary butyl, pentyl, isopentyl, hexyl, cyclopropylmethyl, etc.
[0056] Alkyl groups can be unsubstituted, or they can be substituted to the extent that such substitution makes sense chemically. Typical substituents include, but are not limited to, halo, =0, =N-CN, =N-ORa, =NRa, -ORa, -NRa 2, -SRa, -S02Ra, -S02NRa 2, -NRaS02Ra, -NRaCONRa 2, -NRaCOORa, -NRaCORa, -CN, -COORa, -CONRa 2, -OOCRa, -CORa, and -N02, wherein each Ra is independently H, C1-C8 alkyl, C2-C8 heteroalkyl, C3-C8 heterocyclyl,
C4-C10 heterocyclyclalkyl, C1-C8 acyl, C2-C8 heteroacyl, C2-C8 alkenyl,
C2-C8 heteroalkenyl, C2-C8 alkynyl, C2-C8 heteroalkynyl, C6-C10 aryl, or C5-C10 heteroaryl, and each Ra is optionally substituted with halo, =0, =N-CN, =N-ORb, =NRb, ORb, NRb 2, SRb, S02Rb, S02NRb 2, NRbS02Rb, NRbCONRb 2, NRbCOORb, NRbCORb, CN, COORb, CONRb 2, OOCRb, CORb, and N02, wherein each Rb is independently H, C1-C8 alkyl, C2-C8 heteroalkyl, C3-C8 heterocyclyl, C4-C10 heterocyclyclalkyl, C1-C8 acyl, C2-C8 heteroacyl, C6-C10 aryl or C5-C10 heteroaryl. Alkyl, alkenyl and alkynyl groups can also be substituted by C1-C8 acyl, C2-C8 heteroacyl, C6-C10 aryl or C5-C10 heteroaryl, each of which can be substituted by the substituents that are appropriate for the particular group. Where a substituent group contains two Ra or Rb groups on the same or adjacent atoms (e.g., -NRb2, or -NRb-C(0) Rb), the two Ra or Rb groups can optionally be taken together with the atoms in the substituent group to which the are attached to form a ring having 5-8 ring members, which can be substituted as allowed for the Ra or Rb itself, and can contain an additional heteroatom (N, O or S) as a ring member. [0057] The term "alkenyl" as used herein refers to an alkyl as defined above having at least two carbon atoms and at least one carbon-carbon double bond. Thus, particularly contemplated alkenyl groups include straight, branched, or cyclic alkenyl groups having two to ten carbon atoms (e.g., ethenyl, propenyl, butenyl, pentenyl, etc.) or 5-10 atoms for cyclic alkenyl groups. Alkenyl groups are optionally substituted by groups suitable for alkyl groups as set forth herein.
[0058] Similarly, the term "alkynyl" as used herein refers to an alkyl or alkenyl as defined above and having at least two (preferably three) carbon atoms and at least one carbon-carbon triple bond. Especially contemplated alkynyls include straight, branched, or cyclic alkynes having two to ten total carbon atoms (e.g. , ethynyl, propynyl, butynyl, cyclopropylethynyl, etc.). Alkynyl groups are optionally substituted by groups suitable for alkyl groups as set forth herein.
[0059] The term "cycloalkyl" as used herein refers to a cyclic alkane (i.e. , in which a chain of carbon atoms of a hydrocarbon forms a ring), preferably including three to eight carbon atoms. Thus, exemplary cycloalkanes include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Cycloalkyls also include one or two double bonds, which form the "cycloalkenyl" groups. Cycloalkyl groups are optionally substituted by groups suitable for alkyl groups as set forth herein.
[0060] The term "aryl" or "aromatic moiety" as used herein refers to an aromatic ring system, which may further include one or more non-carbon atoms. These are typically 5-6 membered isolated rings, or 8-10 membered bicyclic groups, and can be substituted. Thus, contemplated aryl groups include (e.g., phenyl, naphthyl, etc.) and pyridyl. Further
contemplated aryl groups may be fused (i.e., covalently bound with 2 atoms on the first aromatic ring) with one or two 5- or 6-membered aryl or heterocyclic group, and are thus termed "fused aryl" or "fused aromatic".
[0061] Aromatic groups containing one or more heteroatoms (typically N, O or S) as ring members can be referred to as heteroaryl or heteroaromatic groups. Typical heteroaromatic groups include monocyclic C5-C6 aromatic groups such as pyridyl, pyrimidyl, pyrazinyl, thienyl, furanyl, pyrrolyl, pyrazolyl, thiazolyl, oxazolyl, isothiazolyl, isoxazolyl, and imidazolyl and the fused bicyclic moieties formed by fusing one of these monocyclic groups with a phenyl ring or with any of the heteroaromatic monocyclic groups to form a C8-C10 bicyclic group such as indolyl, benzimidazolyl, indazolyl, benzotriazolyl, isoquinolyl, quinolyl, benzothiazolyl, benzofuranyl, pyrazolopyridyl, pyrazolopyrimidyl, quinazolinyl, quinoxalinyl, cinnolinyl, and the like. Any monocyclic or fused ring bicyclic system which has the characteristics of aromaticity in terms of electron distribution throughout the ring system is included in this definition. It also includes bicyclic groups where at least the ring which is directly attached to the remainder of the molecule has the characteristics of aromaticity. Typically, the ring systems contain 5- 12 ring member atoms.
[0062] As also used herein, the terms "heterocycle", "cycloheteroalkyl", and "heterocyclic moieties" are used interchangeably herein and refer to any compound in which a plurality of atoms form a ring via a plurality of covalent bonds, wherein the ring includes at least one atom other than a carbon atom as a ring member. Particularly contemplated heterocyclic rings include 5- and 6-membered rings with nitrogen, sulfur, or oxygen as the non-carbon atom (e.g. , imidazole, pyrrole, triazole, dihydropyrimidine, indole, pyridine, thiazole, tetrazole etc.).
Typically these rings contain 0-1 oxygen or sulfur atoms, at least one and typically 2-3 carbon atoms, and up to four nitrogen atoms as ring members. Further contemplated heterocycles may be fused (i.e., covalently bound with two atoms on the first heterocyclic ring) to one or two carbocyclic rings or heterocycles, and are thus termed "fused heterocycle" or "fused
heterocyclic ring" or "fused heterocyclic moieties" as used herein. Where the ring is aromatic, these can be referred to herein as 'heteroaryl' or heteroaromatic groups.
[0063] Heterocyclic groups that are not aromatic can be substituted with groups suitable for alkyl group substituents, as set forth above.
[0064] Aryl and heteroaryl groups can be substituted where permitted. Suitable substituents include, but are not limited to, halo, -ORa, -NRa 2, -SRa, -S02Ra, -S02NRa 2, -NRaS02Ra, -NRaCONRa 2, -NRaCOORa, -NRaCORa, -CN, -COORa, -CONRa 2, -OOCRa, -CORa, and -N02, wherein each Ra is independently H, Ci-C8 alkyl, C2-C8 heteroalkyl, C3-C8 heterocyclyl, C4-C10 heterocyclyclalkyl, C1-C8 acyl, C2-C8 heteroacyl, C2-C8 alkenyl, C2-C8
heteroalkenyl, C2-C8 alkynyl, C2-C8 heteroalkynyl, C6-C10 aryl, or C5-C10 heteroaryl, and each Ra is optionally substituted with halo, =0, =N-CN, =N-ORb, =NRb, ORb, NRb 2, SRb, S02Rb, S02NRb 2, NRbS02Rb, NRbCONRb 2, NRbCOORb, NRbCORb, CN, COORb, CONRb 2, OOCRb, CORb, and N02, wherein each Rb is independently H, C1-C8 alkyl, C2-C8 heteroalkyl, C3-C8 heterocyclyl, C4-C10 heterocyclyclalkyl, C1-C8 acyl, C2-C8 heteroacyl, C6-C10 aryl or C5-C10 heteroaryl. Alkyl, alkenyl and alkynyl groups can also be substituted by C1-C8 acyl, C2-C8 heteroacyl, C6-C10 aryl or C5-C10 heteroaryl, each of which can be substituted by the substituents that are appropriate for the particular group. Where a substituent group contains two Ra or Rb groups on the same or adjacent atoms (e.g., -NRb2, or -NRb-C(0) Rb), the two Ra or Rb groups can optionally be taken together with the atoms in the substituent group to which the are attached to
a b
form a ring having 5-8 ring members, which can be substituted as allowed for the R or R itself, and can contain an additional heteroatom (N, O or S) as a ring member. [0065] As also used herein, the terms "imidazopyridine" or "imidazopyrimidine" or "thiazopyridine" or "thiazopyrimidine" herein refer to any compound in which the two designated heterocyclic rings are fused by any two adjacent atoms on the two heterocyclic rings.
[0066] The term "alkoxy" as used herein refers to a hydrocarbon group connected through an oxygen atom, e.g., -O-Hc, wherein the hydrocarbon portion He may have any number of carbon atoms, typically 1-10 carbon atoms, may further include a double or triple bond and may include one or two oxygen, sulfur or nitrogen atoms in the alkyl chains, and can be substituted with aryl, heteroaryl, cycloalkyl, and/or heterocyclyl groups. For example, suitable alkoxy groups include methoxy, ethoxy, propyloxy, isopropoxy, methoxyethoxy, benzyloxy, allyloxy, and the like. Similarly, the term "alkylthio" refers to alkylsulfides of the general formula -S- Hc, wherein the hydrocarbon portion He is as described for alkoxy groups. For example, contemplated alkylthio groups include methylthio, ethylthio, isopropylthio, methoxyethylthio, benzylthio, allylthio, and the like.
[0067] The term 'amino' as used herein refers to the group -N¾. The term "alkylamino" refers to amino groups where one or both hydrogen atoms are replaced by a hydrocarbon group He as described above, wherein the amino nitrogen "N" can be substituted by one or two He groups as set forth for alkoxy groups described above. Exemplary alkylamino groups include methylamino, dimethylamino, ethylamino, diethylamino, etc.
[0068] The term 'acyl' as used herein refers to a group of the formula -C(=0)-D, where D represents an alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl, or heterocycle as described above. Typical examples are groups wherein D is a C1-C10 alkyl, C2-C10 alkenyl or alkynyl, or phenyl, each of which is optionally substituted. In some embodiments, D can be H, Me, Et, isopropyl, propyl, butyl, C1-C4 alkyl substituted with -OH, -OMe, or N¾, phenyl, halophenyl, alkylphenyl, and the like.
[0069] The term "aryloxy" as used herein refers to an aryl group connecting to an oxygen atom, wherein the aryl group may be further substituted. For example suitable aryloxy groups include phenyloxy, etc. Similarly, the term "arylthio" as used herein refers to an aryl group connecting to a sulfur atom, wherein the aryl group may be further substituted. For example suitable arylthio groups include phenylthio, etc.
[0070] The hydrocarbon portion of each alkoxy, alkylthio, alkylamino, and aryloxy, etc., can be substituted as appropriate for the relevant hydrocarbon moiety.
[0071] The term "halogen" as used herein refers to fluorine, chlorine, bromine and iodine. Where present as a substituent group, halogen or halo typically refers to F or CI or Br, more typically F or CI. [0072] It should further be recognized that all of the above-defined groups may further be substituted with one or more substituents, which may in turn be substituted with hydroxy, amino, cyano, C1-C4 alkyl, halo, or C1-C4 haloalkyl. For example, a hydrogen atom in an alkyl or aryl can be replaced by an amino, halo or Ci-4 haloalkyl or alkyl group.
[0073] The term "substituted" as used herein refers to a replacement of a hydrogen atom of the unsubstituted group with a functional group, and particularly contemplated functional groups include nucleophilic groups (e.g., -NH2, -OH, -SH, -CN, etc.), electrophilic groups (e.g. , C(0)OR, C(X)OH, etc.), polar groups (e.g. , -OH), non-polar groups (e.g. , heterocycle, aryl, alkyl, alkenyl, alkynyl, etc.), ionic groups (e.g. , -NH3 "1"), and halogens (e.g. , -F, -CI), NHCOR, NHCONH2, OCH2COOH, OCH2CONH2, OCH2CONHR, NHCH2COOH, NHCH2CONH2, NHSO2R, OCH2-heterocycles, PO3H, SO3H, amino acids, and all chemically reasonable combinations thereof. Moreover, the term "substituted" also includes multiple degrees of substitution, and where multiple substituents are disclosed or claimed, the substituted compound can be independently substituted by one or more of the disclosed or claimed substituent moieties.
[0074] Any formula given herein is intended to represent compounds having structures depicted by the structural formula as well as certain variations or forms. In particular, compounds of any formula given herein may have asymmetric centers and therefore exist in different enantiomeric forms. All optical isomers and stereoisomers of the compounds of the general formula, and mixtures thereof, are considered within the scope of the formula. Thus, any formula given herein is intended to represent a racemate, one or more enantiomeric forms, one or more diastereomeric forms, one or more atropisomeric forms, and mixtures thereof. Furthermore, certain structures may exist as geometric isomers (i.e., cis and trans isomers), as tautomers, or as atropisomers. Additionally, any formula given herein is intended to refer also to any one of hydrates, solvates, and polymorphs of such compounds, and mixtures thereof, even if such forms are not listed explicitly. In some embodiments, the solvent is water and the solvates are hydrates.
[0075] Any formula given herein is also intended to represent unlabeled forms as well as isotopically labeled forms of the compounds. Isotopically labeled compounds have structures depicted by the formulas given herein except that one or more atoms are replaced by an atom having a selected atomic mass or mass number. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, chlorine, and iodine, such as 2H, 3H, nC, 13C, 14C, 15N, 180, 170, 31P, 32P, 35S, 18F, 36C1, and 125I, respectively. Such isotopically labelled compounds are useful in metabolic studies (preferably with 14C), reaction kinetic studies (with, for example 2H or 3H), detection or imaging techniques [such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT)] including drug or substrate tissue distribution assays, or in radioactive treatment of patients. In particular, an 18F or 11 C labeled compound may be particularly preferred for PET or SPECT studies. Further, substitution with heavier isotopes such as deuterium (i.e., 2H) may afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements. Isotopically labeled compounds of this invention and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the schemes or in the examples and preparations described below by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent.
[0076] The nomenclature "Ci_j" or "Ci-Cj" with j > i, when applied herein to a class of substituents, is meant to refer to embodiments of this invention for which each and every one of the number of carbon members, from i to j including i and j, is independently realized. By way of example, the term Ci_3 or C1-C3 refers independently to embodiments that have one carbon member (Ci or CI), embodiments that have two carbon members (C2 or C2), and embodiments that have three carbon members (C3 or C3).
[0077] Any disubstituent referred to herein is meant to encompass the various attachment possibilities when more than one of such possibilities are allowed. For example, reference to disubstituent -A-B-, where A≠ B, refers herein to such disubstituent with A attached to a first substituted member and B attached to a second substituted member, and it also refers to such disubstituent with A attached to the second substituted member and B attached to the first substituted member.
[0078] The invention also includes pharmaceutically acceptable salts of the compounds represented by Formula I-V, preferably of those described above and of the specific compounds exemplified herein, and pharmaceutical compositions comprising such salts, and methods of using such salts.
[0079] A "pharmaceutically acceptable salt" is intended to mean a salt of a free acid or base of a compound represented herein that is non-toxic, biologically tolerable, or otherwise biologically suitable for administration to the subject. See, generally, S.M. Berge, et al., "Pharmaceutical Salts," J. Pharm. Sci., 1977, 66, 1-19. Preferred pharmaceutically acceptable salts are those that are pharmacologically effective and suitable for contact with the tissues of subjects without undue toxicity, irritation, or allergic response. A compound described herein may possess a sufficiently acidic group, a sufficiently basic group, both types of functional groups, or more than one of each type, and accordingly react with a number of inorganic or organic bases, and inorganic and organic acids, to form a pharmaceutically acceptable salt.
[0080] Examples of pharmaceutically acceptable salts include sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, phosphates, monohydrogen-phosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates, caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyne-l,4-dioates, hexyne-l,6-dioates, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, hydroxybenzoates, methoxybenzoates, phthalates, sulfonates, methylsulfonates,
propylsulfonates, besylates, xylenesulfonates, naphthalene- 1- sulfonates, naphthalene-2- sulfonates, phenylacetates, phenylpropionates, phenylbutyrates, citrates, lactates, γ- hydroxybutyrates, glycolates, tartrates, and mandelates.
[0081] For a compound of Formula I-V that contains a basic nitrogen, a pharmaceutically acceptable salt may be prepared by any suitable method available in the art, for example, treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, sulfamic acid, nitric acid, boric acid, phosphoric acid, and the like, or with an organic acid, such as acetic acid, phenylacetic acid, propionic acid, stearic acid, lactic acid, ascorbic acid, maleic acid, hydroxymaleic acid, isethionic acid, succinic acid, valeric acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, oleic acid, palmitic acid, lauric acid, a pyranosidyl acid, such as glucuronic acid or galacturonic acid, an alpha-hydroxy acid, such as mandelic acid, citric acid, or tartaric acid, an amino acid, such as aspartic acid or glutamic acid, an aromatic acid, such as benzoic acid, 2-acetoxybenzoic acid, naphthoic acid, or cinnamic acid, a sulfonic acid, such as laurylsulfonic acid, p-toluenesulfonic acid, methanesulfonic acid, or ethanesulfonic acid, or any compatible mixture of acids such as those given as examples herein, and any other acid and mixture thereof that are regarded as equivalents or acceptable substitutes in light of the ordinary level of skill in this technology.
[0082] The invention also relates to pharmaceutically acceptable prodrugs of the compounds of Formula I-V, and treatment methods employing such pharmaceutically acceptable prodrugs. The term "prodrug" means a precursor of a designated compound that, following administration to a subject, yields the compound in vivo via a chemical or physiological process such as solvolysis or enzymatic cleavage, or under physiological conditions (e.g., a prodrug on being brought to physiological pH is converted to the compound of Formula I- V). A "pharmaceutically acceptable prodrug" is a prodrug that is non-toxic, biologically tolerable, and otherwise biologically suitable for administration to the subject. Illustrative procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in "Design of Prodrugs", ed. H. Bundgaard, Elsevier, 1985.
[0083] The present invention also relates to pharmaceutically active metabolites of compounds of Formula I-V, and uses of such metabolites in the methods of the invention. A "pharmaceutically active metabolite" means a pharmacologically active product of metabolism in the body of a compound of Formulas I-V or salt thereof. Prodrugs and active metabolites of a compound may be determined using routine techniques known or available in the art. See, e.g., Bertolini et al., /. Med. Chem. 1997, 40, 2011-2016; Shan et al., /. Pharm. Sci. 1997, 86 (7), 765-767; Bagshawe, Drug Dev. Res. 1995, 34, 220-230; Bodor, Adv. Drug Res. 1984, 13, 255- 331; Bundgaard, Design of Prodrugs (Elsevier Press, 1985); and Larsen, Design and
Application of Prodrugs, Drug Design and Development (Krogsgaard-Larsen et al., eds., Harwood Academic Publishers, 1991).
Formulations
[0084] Any suitable formulation of the compounds described herein can be prepared. See generally, Remington's Pharmaceutical Sciences, (2000) Hoover, J. E. editor, 20 th edition, Lippincott Williams and Wilkins Publishing Company, Easton, Pa., pages 780-857. A formulation is selected to be suitable for an appropriate route of administration. Some routes of administration are oral, parenteral, by inhalation, topical, rectal, nasal, buccal, vaginal, via an implanted reservoir, or other drug administration methods. In cases where compounds are sufficiently basic or acidic to form stable nontoxic acid or base salts, administration of the compounds as salts may be appropriate. Examples of pharmaceutically acceptable salts are organic acid addition salts formed with acids that form a physiological acceptable anion, for example, tosylate, methanesulfonate, acetate, citrate, malonate, tartarate, succinate, benzoate, ascorbate, a-ketoglutarate, and a-glycerophosphate. Suitable inorganic salts may also be formed, including hydrochloride, sulfate, nitrate, bicarbonate, and carbonate salts.
Pharmaceutically acceptable salts are obtained using standard procedures well known in the art, for example, by a sufficiently basic compound such as an amine with a suitable acid, affording a physiologically acceptable anion. Alkali metal (e.g. , sodium, potassium or lithium) or alkaline earth metal (e.g. , calcium) salts of carboxylic acids also are made.
[0085] Where contemplated compounds are administered in a pharmacological composition, it is contemplated that the compounds can be formulated in admixture with a pharmaceutically acceptable excipient and/or carrier. For example, contemplated compounds can be administered orally as neutral compounds or as pharmaceutically acceptable salts, or intravenously in a physiological saline solution. Conventional buffers such as phosphates, bicarbonates or citrates can be used for this purpose. Of course, one of ordinary skill in the art may modify the formulations within the teachings of the specification to provide numerous formulations for a particular route of administration. In particular, contemplated compounds may be modified to render them more soluble in water or other vehicle, which for example, may be easily accomplished with minor modifications (salt formulation, esterification, etc.) that are well within the ordinary skill in the art. It is also well within the ordinary skill of the art to modify the route of administration and dosage regimen of a particular compound in order to manage the pharmacokinetics of the present compounds for maximum beneficial effect in a patient.
[0086] The compounds having formula I-V as described herein are generally soluble in organic solvents such as chloroform, dichloromethane, ethyl acetate, ethanol, methanol, isopropanol, acetonitrile, glycerol, N,N-dimethylformamide, N,N-dimetheylaceatmide, dimethylsulfoxide, etc. In one embodiment, the present invention provides formulations prepared by mixing a compound having formula I-V with a pharmaceutically acceptable carrier. In one aspect, the formulation may be prepared using a method comprising: a) dissolving a described compound in a water-soluble organic solvent, a non-ionic solvent, a water-soluble lipid, a cyclodextrin, a vitamin such as tocopherol, a fatty acid, a fatty acid ester, a
phospholipid, or a combination thereof, to provide a solution; and b) adding saline or a buffer containing 1-10% carbohydrate solution. In one example, the carbohydrate comprises dextrose. The pharmaceutical compositions obtained using the present methods are stable and useful for animal and clinical applications.
[0087] Illustrative examples of water soluble organic solvents for use in the present methods include and are not limited to polyethylene glycol (PEG), alcohols, acetonitrile, N-methyl-2- pyrrolidone, N,N-dimethylformamide, N,N-dimethylacetamide, dimethyl sulfoxide, or a combination thereof. Examples of alcohols include but are not limited to methanol, ethanol, isopropanol, glycerol, or propylene glycol.
[0088] Illustrative examples of water soluble non-ionic surfactants for use in the present methods include and are not limited to CREMOPHOR® EL, polyethylene glycol modified CREMOPHOR® (polyoxyethyleneglyceroltriricinoleat 35), hydrogenated CREMOPHOR® RH40, hydrogenated CREMOPHOR® RH60, PEG-succinate, polysorbate 20, polysorbate 80, SOLUTOL® HS (polyethylene glycol 660 12-hydroxystearate), sorbitan monooleate, poloxamer, LABRAFIL® (ethoxylated persic oil), LABRASOL® (capryl-caproyl macrogol-8- glyceride), GELUCIRE® (glycerol ester), SOFTIGEN® (PEG 6 caprylic glyceride), glycerin, glycol-polysorbate, or a combination thereof. [0089] Illustrative examples of water soluble lipids for use in the present methods include but are not limited to vegetable oils, triglycerides, plant oils, or a combination thereof.
Examples of lipid oils include but are not limited to castor oil, polyoxyl castor oil, corn oil, olive oil, cottonseed oil, peanut oil, peppermint oil, safflower oil, sesame oil, soybean oil, hydrogenated vegetable oil, hydrogenated soybean oil, a triglyceride of coconut oil, palm seed oil, and hydrogenated forms thereof, or a combination thereof.
[0090] Illustrative examples of fatty acids and fatty acid esters for use in the present methods include but are not limited to oleic acid, monoglycerides, diglycerides, a mono- or di- fatty acid ester of PEG, or a combination thereof.
[0091] Illustrative examples of cyclodextrins for use in the present methods include but are not limited to alpha-cyclodextrin, beta-cyclodextrin, hydroxypropyl-beta-cyclodextrin, or sulfobutyl ether-beta-cyclodextrin.
[0092] Illustrative examples of phospholipids for use in the present methods include but are not limited to soy phosphatidylcholine, or distearoyl phosphatidylglycerol, and hydrogenated forms thereof, or a combination thereof.
[0093] One of ordinary skill in the art may modify the formulations within the teachings of the specification to provide numerous formulations for a particular route of administration. In particular, the compounds may be modified to render them more soluble in water or other vehicle. It is also well within the ordinary skill of the art to modify the route of administration and dosage regimen of a particular compound in order to manage the pharmacokinetics of the present compounds for maximum beneficial effect in a patient.
Methods of Using the Invented Compounds and Pharmaceutical Compositions thereof
[0094] The present invention also provides pharmaceutical compositions for the treatment of a cell proliferative disorder, comprising any compound having formula I-V,
[0095] To practice the method of the present invention, compounds having formula I-V and pharmaceutical compositions thereof may be administered orally, parenterally, by inhalation, topically, rectally, nasally, buccally, vaginally, via an implanted reservoir, or other drug administration methods. The term "parenteral" as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intraarticular, intraarterial, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.
[0096] A sterile injectable composition, such as a sterile injectable aqueous or oleaginous suspension, may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent. Among the acceptable vehicles and solvents that may be employed include mannitol, water, Ringer' s solution and isotonic sodium chloride solution. Suitable carriers and other pharmaceutical composition components are typically sterile.
[0097] In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium (e.g. , synthetic mono- or diglycerides). Fatty acids, such as oleic acid and its glyceride derivatives, are useful in the preparation of injectables, as are pharmaceutically acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions can also contain a long-chain alcohol diluent or dispersant, or carboxymethyl cellulose or similar dispersing agents. Various emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms can also be used for the purpose of formulation.
[0098] A composition for oral administration may be any orally acceptable dosage form including, but not limited to, tablets, capsules, emulsions and aqueous suspensions, dispersions and solutions. In the case of tablets for oral use, commonly used carriers include lactose and corn starch. Lubricating agents, such as magnesium stearate, can also be added. For oral administration in a capsule form, useful diluents include lactose and dried corn starch. When aqueous suspensions or emulsions are administered orally, the active ingredient can be suspended or dissolved in an oily phase combined with emulsifying or suspending agents. If needed, certain sweetening, flavoring, or coloring agents can be added. A nasal aerosol or inhalation compositions can be prepared according to techniques well-known in the art of pharmaceutical formulation and can be prepared as solutions in, for example saline, employing suitable preservatives (for example, benzyl alcohol), absorption promoters to enhance bioavailability, and/or other solubilizing or dispersing agents known in the art.
[0099] In addition, the compounds having formula I-V may be administered alone or in combination with other anticancer agents for the treatment of various cancers or conditions. Combination therapies according to the present invention comprise the administration of at least one compound of the present invention or a functional derivative thereof and at least one other pharmaceutically active ingredient. The active ingredient(s) and pharmaceutically active agents may be administered separately or together. The amounts of the active ingredient(s) and pharmaceutically active agent(s) and the relative timings of administration will be selected in order to achieve the desired combined therapeutic effect. [0100] A compound of formula I-V can besides or in addition be administered especially for tumor therapy in combination with chemotherapy, radiotherapy, immunotherapy, surgical intervention, or a combination of these. Long-term therapy and adjuvant therapy in
combination with the treatment strategies, as described above. Other possible treatments are therapy to maintain the patient's status after tumor regression, or even chemopreventive therapy, for example in patients at risk.
[0101] The compounds and compositions described herein can be administered to a subject in need of treatment for a cell proliferation disorder such as cancer, particularly cancers selected from leukemia, lymphoma, lung cancer, colon cancer, CNS cancer, melanoma, ovarian cancer, renal cancer, prostate cancer, breast cancer, head and neck cancers, and pancreatic cancer. The subject is typically a mammal diagnosed as being in need of treatment for one or more of such proliferative disorders, and frequently the subject is a human. The methods comprise administering an effective amount of at least one compound of the invention; optionally the compound may be administered in combination with one or more additional therapeutic agents, particularly therapeutic agents known to be useful for treating the cancer or proliferative disorder afflicting the particular subject.
[0102] The term "treat" or "treating" as used herein is intended to refer to administration of a compound of the invention to a subject for the purpose of creating a therapeutic benefit to a subject suffering from or diagnosed with cancer or cell proliferation disease, disorder or condition. Treating includes preventing, reversing, ameliorating, alleviating, inhibiting the progress of, or lessening the severity of, a cancer or cell proliferation disorder or one or more symptoms of that disorder, as well as the delay of progression of a disease, disorder or condition. The term "subject" refers to a mammalian patient in need of such treatment, such as a human.
[0103] In the inhibitory methods of the invention, an "effective amount" means an amount sufficient to reduce, slow the progression of, or reverse cancer or cell proliferation disorder. Measuring the amount of cell proliferation may be performed by routine analytical methods such as those described below. Such modulation is useful in a variety of settings, including in vitro assays. In such methods, the cell is preferably a GTL-16, H1993 or A431 cells.
[0104] In treatment methods according to the invention, an "effective amount" means an amount or dose sufficient to generally bring about the desired therapeutic benefit in subjects needing such treatment. Effective amounts or doses of the compounds of the invention may be ascertained by routine methods, such as modeling, dose escalation, or clinical trials, taking into account routine factors, e.g., the mode or route of administration or drug delivery, the pharmacokinetics of the agent, the severity and course of the infection, the subject's health status, condition, and weight, and the judgment of the treating physician.
[0105] Once improvement of the patient's disease has occurred, the dose may be adjusted for preventative or maintenance treatment. For example, the dosage or the frequency of administration, or both, may be reduced as a function of the symptoms, to a level at which the desired therapeutic or prophylactic effect is maintained. Of course, if symptoms have been alleviated to an appropriate level, treatment may cease. Patients may, however, require intermittent treatment on a long-term basis upon any recurrence of symptoms. Patients may also require chronic treatment on a long-term basis.
[0106] The term "combination" refers to either a fixed combination in one dosage unit form, or a kit of parts for the combined administration where a compound of the formula (I) and a combination partner (e.g. an other drug as explained below, also referred to as "therapeutic agent" or "co-agent") may be administered independently at the same time or separately within time intervals, especially where these time intervals allow that the combination partners show a cooperative, e.g. synergistic effect. The terms "co-administration" or "combined administration" or the like as utilized herein are meant to encompass administration of the selected combination partner to a single subject in need thereof (e.g. a patient), and are intended to include treatment regimens in which the agents are not necessarily administered by the same route of
administration or at the same time. The term "pharmaceutical combination" as used herein means a product that results from the mixing or combining of more than one active ingredient and includes both fixed and non-fixed combinations of the active ingredients. The term "fixed combination" means that the active ingredients, e.g. a compound of formula (I) and a combination partner, are both administered to a patient simultaneously in the form of a single entity or dosage. The term "non-fixed combination" means that the active ingredients, e.g. a compound of formula (I) and a combination partner, are both administered to a patient as separate entities either simultaneously, concurrently or sequentially with no specific time limits, wherein such administration provides therapeutically effective levels of the two compounds in the body of the patient. The latter also applies to cocktail therapy, e.g. the administration of three or more active ingredients.
Drug Combinations
[0107] The inventive compounds described herein may be used in pharmaceutical compositions or methods in combination with one or more additional active ingredients in the treatment of cancer, cell proliferative disorders or inflammatory conditions. For example, additional active ingredients are those that are known or discovered to be effective in treating cancer, cell proliferative disorders or inflammatory conditions, including those active against another target associated with the disease, or a combination thereof. Such a combination may serve to increase efficacy, ameliorate other disease symptoms, decrease one or more side effects, or decrease the required dose of an inventive compound. The additional active ingredients may be administered in a separate pharmaceutical composition from a compound of the present invention or may be included with a compound of the present invention in a single pharmaceutical composition. The additional active ingredients may be administered simultaneously with, prior to, or after administration of a compound of the present invention.
[0108] Thus, a compound of the formula I-V may be used in combination with other antiproliferative compounds. Such antiproliferative compounds include, but are not limited to aromatase inhibitors; antiestrogens; topoisomerase I inhibitors; topoisomerase II inhibitors; microtubule active compounds; alkylating compounds; histone deacetylase inhibitors;
compounds which induce cell differentiation processes; cyclooxygenase inhibitors; MMP inhibittors; mTOR inhibitors; antineoplastic antimetabolites; platin compounds; compounds targeting/decreasing a protein or lipid kinase activity; anti- angiogenic compounds; compounds which target, decrease or inhibit the activity of a protein or lipid phosphatase; gonadorelin agonists; anti-androgens; methionine aminopeptidase inhibitors; bisphosphonates; biological response modifiers; antiproliferative antibodies; heparanase inhibitors; inhibitors of Ras oncogenic isoforms; telomerase inhibitors; proteasome inhibitors; compounds used in the treatment of hematologic malignancies; compounds which target, decrease or inhibit the activity of Fit- 3; Hsp90 inhibitors; kinesin spindle protein inhibitors; MEK inhibitors; leucovorin; EDG binders; antileukemia compounds; ribonucleotide reductase inhibitors; S-adenosylmethionine decarboxylase inhibitors; angiostatic steroids; corticosteroids; other chemotherapeutic compounds (as defined below); photosensitizing compounds.
[0109] Other chemotherapeutic compounds" include, but are not limited to, plant alkaloids, hormonal compounds and antagonists; biological response modifiers, preferably lymphokines or interferons; antisense oligonucleotides or oligonucleotide derivatives; shRNA or siRNA; or miscellaneous compounds or compounds with other or unknown mechanism of action.
[0110] Further, they may be used in combination with other cancer treatment approaches, including chemotherapy, radiotherapy, immunotherapy, surgical intervention, implants, or a combination of these. Chemical Synthesis
[0111] Exemplary chemical entities useful in methods of the invention will now be described by reference to illustrative synthetic schemes for their general preparation below and the specific examples that follow. Artisans will recognize that, to obtain the various compounds herein, starting materials may be suitably selected so that the ultimately desired substituents will be carried through the reaction scheme with or without protection as appropriate to yield the desired product. Alternatively, it may be necessary or desirable to employ, in the place of the ultimately desired substituent, a suitable group that may be carried through the reaction scheme and replaced as appropriate with the desired substituent. Each of the reactions depicted in Schemes is preferably run at a temperature from about room temperature (rt) to the reflux temperature of the organic solvent used. Unless otherwise specified, the variables are as defined above in reference to Formulas I-V.
Exemplary Synthesis:
[0112] The compounds described above can be synthesized as illustrated in Schemes 1. Some representative compounds are illustrated in Scheme lb, which depicts some suitable Het
Figure imgf000032_0001
Cyclo A: substituted benzenes, pyrindines, heterocyclic compounds
Cyclo A : substituted benzenes, pyrindines, heterocyclic amines
Cyclo B: substituted benzenes, pyrindine
Cyclo C: heterocyclic compounds
3 Reaction conditions: (a) substituted-o-aminonitrile, isothiocyanate 1.05 eq, reflux in the pyridine for 0.5-2 hours; (b) thiourea intermediates, benzylbromides 1.1 eq,
K2C03 2eq, stir in acetone at 50~60 deg for 2~3 hours.
Scheme 1: Generalized synthetic scheme
Figure imgf000033_0001
Z = S, O; n = 1 -5; A = bond, OO; CONH; or NHCO; etc.
R1, R2, R3, R = H, Me, Et, i-Pr, Propyl, Butyl, F, Br, CI, 1, OMe, NMe2, NEt2, N02, SMe, SEt, CF3, CH2COOEt, cyclopropyl, NHS02R, alkenyl, alkynyl, saturated/ unsaturated carbocyclic or heterocyclic ring (fused or not fused), etc. R and R' are as set forth for Formula I.
Scheme lb: Representative substituted and fused pyrazolo pyrimidine embodiments.
Detail procedures of target compounds and key intermediates
[0113] All cyclo-C compounds and isothiocyanates used were obtained from commercial sources (Sigma-Aldrich, Alfa Aesar). Most bromides were synthesized by our group member. Every final compound was characterized by LC-MS(API 150), and some active structures were analyzed by ^-NMR, 13C-NMR.
General Procedure for the Synthesis of Thiourea Intermediates:
Figure imgf000034_0001
[0114] A mixture of substituted-o-aminonitrile (lOmmol) and isothiocycanates (10.5mmol) were refluxed in pyridine (15mL) for over than half an hour, then light- yellow solids were appeared, meanwhile solvent decreased in a short time. The mixture were stirred for another 10 minutes, cooled to roomtemperature, filtered with Buchner funnel, wshed with 50ml EtOH, and dried in vacuum drying oven. The yield of pure products was high to 95%.
General Synthetic scheme:
Figure imgf000034_0002
[0115] Mixture of thiourea intermediates, bromides and K2CO3 (mol ratio, 0.5 mmol : 0.52 mmol : 1 mmol) in 3mL acetone, stirred and heated in 65 deg water bath for 2 hours. The reaction was completed indicated by TLC. Cool, add water 20mL and EA (ethyl acetate) 20 mL, extract, separate, collect and concentrate, the solid will crystallize, the last dry it after filtered. Yield was high to 85%.
General Procedure for the Synthesis of Intermediate Benzylbromo bromides [0116] Typical romides
Figure imgf000035_0001
[0117] Step 1. Methyl 5-methylnicotinate (10 mmol, 1.52g) was dissolved in 50 Ml dried THF, then cooled to 0 deg with ice-bath. LiAlH4 (16 mmol, 0.61g) was added in small portions during about 20 minutes. The mixture stirred for another 20 minutes until the reaction was completed, which monitored by TLC. 1.5 mL water was dropwised into the mixture to quench the reaction, then filtered, solution was concentrated to get the (5-methylpyridin-3-yl)methanol, Yield was high to 85%
[0118] Step 2. Alcohol (10 mmol) were refluxed in 10ml HBr (40% aq) for more than 5 hours, and monitored through TLC or LC-MS. After completed, mixture were heated in order to evaporator solvents (water and excess HBr) until the mixture became sticky. Cool the mixture, add acetone to the mixture, precipitated solid and filtered followed by drying. Yield was high to
80%.
[0119] Similar intermediate pyridine compounds can be synthesized using the above protocol. Some examples are provided in the Table below.
Figure imgf000035_0002
Figure imgf000036_0001
Synthesis of sulfone derivatives through oxidation (A429, A430, A434 and A484):
Figure imgf000036_0002
A372 A429
[0120] Mixture of A372, Na2W04, H202 (mol ratio 1:2:2) were stirred in CH3COOH at RT for3 hours, then add water and EA (ethyl acetate), extract, separate, collect, column chromatography and concentrate, the solid will crystallize, the last dry it after filtered. A429 was given in proper yields. This method were also applied in the preparation of A430, A434 and A484 from oxidation of A419, A348 and A424. Synthesis of sulfoxide derivatives through oxidation, A431, A433 and A485
Figure imgf000037_0001
A419 A431
[0121] Mixture of A419, NaB03.4H20 (mol ratio 1:1.1) were stirred in CH3COOH at 40deg for3 hours, then add water and EA (ethyl acetate), extract, separate, collect and concentrate, the solid will crystallize, the last dry it after filtered. A431 was given in proper yields. This method was also applied in the preparation of A433 and A485 from oxidation of A348 and A424.
Synthesis of 2-bromo-l-(pyrrolidin-l-yl)ethanone
Figure imgf000037_0002
[0122] To 2-bromoacetyl bromide (10 mmol) in dry DCM (10 mL), pyrrolidine (10 mmol) in DCM (5 mL) was added drop- wise at -10 - -5°C (ice bath) with stirring. The addition was completed in ~ 50 min. TEA was then slowly added into the reaction mixture for 10 min. At this point, the ice bath was removed. The reaction mixture was warmed up to room temperature (rt) and continued stirring for 1 h. After removal of solid, the filtrate was collected and
concentrated. Acetone (20 mL) was added to this concentrated residue, giving a DCM/ Acetone solution of 2-bromo-l-(pyrrolidin-l-yl) ethanone (-0.35 mmol/ mL), which was stored at -20 °C for future use.
[0123] Similar intermediates can be prepared using the above protocol. Some examples are provided in the Table below.
Figure imgf000038_0001
Figure imgf000038_0002
Scheme lb Synthesis of substituted and fused pyrazolo pyrimidine derivatives
[0124] Substituted 5-amino pyrazole-4-carbonitrile 1 reacted with substituted
isothiocyanatobenzene 2 in pyridine stirred at refluxing temperature for 2 h providing the corresponding 4-imino-5 -substituted phenyl-4,5-dihydro-pyrazolo-[J,4-ii|pyrimidine-6(7H)- thione 3 in good yield. Thione 3 was alkylated with alkylation reagent halide R-X in acetone and K2CO3 refluxing for 3 h giving the final desired product IV. Below is the synthesis of some representatives.
[0125] Compounds prepared according to the schemes described above may be obtained as single enantiomers, diastereomers, or regioisomers, by enantio-, diastero-, or regiospecific synthesis, or by resolution. Compounds prepared according to the schemes above may alternately be obtained as racemic (1 : 1) or non-racemic (not 1 : 1) mixtures or as mixtures of diastereomers or regioisomers. Where racemic and non-racemic mixtures of enantiomers are obtained, single enantiomers may be isolated using conventional separation methods known to one skilled in the art, such as chiral chromatography, recrystallization, diastereomeric salt formation, derivatization into diastereomeric adducts, biotransformation, or enzymatic transformation. Where regioisomeric or diastereomeric mixtures are obtained, single isomers may be separated using conventional methods such as chromatography or crystallization.
[0126] The following specific examples are provided to further illustrate the invention and various preferred embodiments.
Example 1 : Synthesis of 2-(4-imino-3-methyl-5-phenyl-4,5-dihydro- lH-pyrazolor3,4-d1
pyrimidin-6-ylthio-)- l-(pyrrolidin-l-yl)ethanone (A311 (9)):
[0127] The synthesis of 2-(4-imino-3-methyl-5-phenyl-4,5-dihydro-lH-pyrazolo[3,4-d] pyrimidin-6-ylthio-)-l-(pyrrolidin- l-yl)ethanone 8 was shown in Scheme 2.
Scheme 2 Synthesis of 2-(4-imino-3-methyl-5-phenyl-4,5-dihydro-lH-pyrazolo[3,4-d] pyr
Figure imgf000040_0001
9
[0128] Synthesis of 4-imino-3-methyl-5-phenyl-4,5-dihydro-lH-pyrazolo-[J,4- ii]pyrimidine-6(7H)-thione (7): Isothiocyanatobenzene 6 (1.16g, 8.6 mmol) was dissolved in 5 ml of dry pyridine. To this solution 3-methyl-5-amino pyrazole-4-carbonitrile 5 (1.0 g, 8.2 mmol) was added. The reaction mixture was heated up, stirred at refluxing for 2 h. A lot of yellow solid was precipitated. The reaction mixture was cooled to room temperature (rt). The yellow precipitation was collected, washed with warm ethanol and dried to yield the desired product 7 (1.45 g, 69%).
[0129] Synthesis of 2-(4-imino-3-methyl-5-phenyl-4,5-dihydro-lH-pyrazolor3,4-dl pyrimidin-6-ylthio-)-l-(pyrrolidin-l-yl)ethanone (A311 (9)): A mixture of 7 (200 mg, 0.78 mmol) and 2-bromo-l-(pyrrolidin-l-yl)ethanone (0.82 mmol, synthesized from 2-bromoacetyl bromide and pyrrolidine) in acetone (6 mL) with K2CO3 (216 mg, 1.56 mmol) was heated to refluxing and stirred for 3 h. The reaction mixture was cooled to room temperature and then water (10 mL) was added. White precipitation was collected, washed 3 times with water (to remove all inorganic salts), re-crystallized from MeOH to yield the desired product A311 (9) (M+l)+ = 369.5 (182 mg, white crystals, 63%). Example 2: Synthesis of 5-(2,6-difluorophenyl)-3-methyl-6-(pyridine-3-ylmethylthio)-lH- pyrimidin-4(5H)-imine (A348 (13)):
[0130] The synthesis of 5-(2,6-difluorophenyl)-3-methyl-6-(pyridine-3-ylmethylthio)-lH- pyrimidin-4
Figure imgf000041_0001
Acetone, refluxing, 3h A348 (13)
Scheme 3 Synthesis of 5-(2,6-difluorophenyl)-3-methyl-6-(pyridine-3-ylmethylthio)-lH- pyrimidin-4(5H)-imine A348 (13)
[0131] Synthesis of 5-(2,6-difluorophenyl)-4-imino-3-methyl-4,5-dihydro-lH-pyrazolo- [3,4-if]pyrimidine-6(7H)-thione (11): l,3-difluoro-2-isothiocyanatobenzene 10 (4.4 mmol) was dissolved in 3 ml of dry pyridine. To this solution 3-methyl-5-amino pyrazole-4-carbonitrile 5 (4.0 mmol) was added. The reaction mixture was heated up, stirred at refluxing for 2 h. A lot of yellow solid was precipitated. The reaction mixture was cooled to room temperature. The yellow precipitation was collected, washed with warm ethanol and dried to yield the desired product 11 (880 mg, 75%).
[0132] Synthesis of 5-(2,6-difluorophenyl)-3-methyl-6-(pyridine-3-ylmethylthio)- 1H- pyrimidin-4(5H)-imine (A348 (13)): A mixture of 11 (147 mg, 0.5 mmol) and commercially available 3-(bromomethyl)pyridine 12 (89 mg, 0.52 mmol) in acetone (3 mL) with K2CO3 (208 mg, 1.5 mmol) was heated to refluxing and stirred for 3 h. The reaction mixture was cooled to rt and then water (20 mL) was added. The mixture was extracted with ethyl acetate (3x 20 mL). The combined organic layers were washed with saturated NaHCC>3 solution and water, dried over Na2S04 and concentrated down under reduced pressure to give crude product, which was further crystallized from MeOH to yield the desired product A348 (13) (M+l)+ = 385.5 (135 mg, white crystals, 70%). Example 3: Synthesis of 5-(2,6-difluorophenyl)-3-methyl-6-(pyridine-3-ylmethylsulfinyl) -1H- pyrimidin-4(5H)-imine (A433 (14)):
[0133] The Synthesis of 5-(2,6-difluorophenyl)-3-methyl-6-(pyridine-3-ylmethylsulfinyl) - lH-pyrimidin-4(5H)-imine (A433 (14)) was shown in Scheme 4.
Figure imgf000042_0001
Scheme 4 Synthesis of 5-(2,6-difluorophenyl)-3-methyl-6-(pyridine-3-ylmethylsulfinyl) - lH-pyrimidin-4(5H)-imine A433 (14)
[0134] Synthesis of 5-(2,6-difluorophenyl)-3-methyl-6-(pyridine-3-ylmethylsulfinyl) -1H- pyrimidin-4(5H)-imine (A433 (14)): A mixture of A348 (13) (54 mg, 0.14 mmol) and
NaB034H20 (0.28 mmol) in CH3COOH (3 mL) was stirred at 40°C for 3 h. The reaction mixture was cooled to rt, water (20 mL) was then added, and adjusted pH with 15% NaOH (aq) to pH >10. The mixture was extracted with ethyl acetate (3x 20 mL). The combined organic layers were washed with saturated NaCl aqueous solution and water, dried over Na2S04 and concentrated down under reduced pressure to give crude product, which was crystallized from EtOAc/ Ether to yield the desired product A433 (14) (M+l)+ = 401.5 (20 mg, 36%).
Example 4: Synthesis of 5-(2,6-difluorophenyl)-3-methyl-6-(pyridine-3-ylmethylsulfonyl) -1H- pyrimidin-4(5H)-imine (A434(15)):
[0135] The Synthesis of 5-(2,6-difluorophenyl)-3-methyl-6-(pyridine-3-ylmethylsulfonyl ) - lH-pyrimidin-4(5H)-imine (A434(15)) was shown in Scheme 5.
Figure imgf000042_0002
Scheme 5 Synthesis of 5-(2,6-difluorophenyl)-3-methyl-6-(pyridine-3-ylmethylsulfonyl) - lH-pyrimidin-4(5H)-imine A434(15)
[0136] Synthesis of 5-(2,6-difluorophenyl)-3-methyl-6-(pyridine-3-ylmethylsulfony -1H- pyrimidin-4(5H)-imine (A434(15)): A mixture of A348 (13) (54 mg, 0.14 mmol), Na2W04 (0.02 mmol) and ¾(¾ (0.2 mL ) in CH3OH (3 mL) was stirred at room temperature for 3 h. Water (20 mL) was added. The mixture was extracted with ethyl acetate (3x 20 mL). The combined organic layers were washed with saturated NaHCC>3 solution and water, dried over Na2S04 and concentrated down under reduced pressure to give crude product, which was crystallized from EtOAc/ Ether to yield the desired product A434(15) (M+l)+ = 417.5 (40 mg,
69 ).
Example 5: Synthesis of 5-(2,3,6-trifluorophenyl)-3-methyl-6-(5-methylpyridine-3- ylmethylthio)-lH-pyrimidin-4(5H)-imine (A424 (19)):
[0137] The synthesis of 5-(2,3,6-trifluorophenyl)-3-methyl-6-(5-methylpyridine-3- ylmethylthio)-lH-pyrimidin-4(5H)-imine (A424 (19)) was shown in scheme 6.
Figure imgf000043_0001
Scheme 6 Synthesis of 5-(2,3,6-difluorophenyl)-3-methyl-6-(5-methylpyridine-3- ylmethylthio)-lH-pyrimidin-4(5H)-imine A424 (19)
[0138] Synthesis of 5-(2,3,6-difluorophenyl)-4-imino-3-methyl-4,5-dihydro-lH-pyrazolo- r3,4-ti|pyrimidine-6(7H)-thione (17): l,2,4-trifluoro-3-isothiocyanatobenzene 16 (4.4 mmol) was dissolved in 3 ml of dry pyridine. To this solution 3 -methyl- 5 -amino pyrazole-4- carbonitrile 5 (4.0 mmol) was added. The reaction mixture was heated up, stirred at refluxing for 2 h. A lot of yellow solid was precipitated. The reaction mixture was cooled to room temperature. The yellow precipitation was collected, washed with warm ethanol and dried to yield the desired product 17 (890 mg, 71%).
[0139] Synthesis of 5-(2,3,6-trifluorophenyl)-3-methyl-6-(5-methylpyridine-3- ylmethylthio)-lH-pyrimidin-4(5H)-imine (A424 (19)): A mixture of 17 (156 mg, 0.5 mmol) and 3-(bromomethyl)-5-methyl pyridine 18 (97 mg, 0.52 mmol) (which was synthesized from commercially available methyl 5-methylnicotinate by reduction and bromination in a traditional way) in acetone (3 mL) with K2CO3 (208 mg, 1.5 mmol) was heated to refluxing and stirred for 3 h. The reaction mixture was cooled to rt and then water (20 mL) was added. The mixture was extracted with ethyl acetate (3x 20 mL). The combined organic layers were washed with saturated NaHC(¾ solution and water, dried over Na2S04 and concentrated down under reduced pressure to give crude product, which was further crystallized from MeOH to yield the desired product A424 (19) (M+l)+ = 417.5 (145 mg, white crystals, 70%).
[0140] Using similar chemistry, the following compounds (A418 (20), A447 (21), and A448 (22)) were also synthesized:
Figure imgf000044_0001
Example 6: Synthesis of N-(5-((5-(2,6-difluorophenyl)-4-imino-3-methyl-4,5-dihydro-lH- pyrazolor3,4-dlpyrimidin-6-ylthio)methyl)pyridin-3-yl)formamide (A512 (23)):
Figure imgf000044_0002
A447 (21) A512 (23) Scheme 7 Synthesis of N-(5-((5-(2,6-difluorophenyl)-4-imino-3-methyl-4,5-dihydro-lH- pyrazolo[3,4-d]pyrimidin-6-ylthio)methyl)pyridin-3-yl)formamide (A512 (23))
[0141] Synthesis of N-(5 -((5 -( 2, 6-difluorophenyl)-4-imino-3 -methyl-4,5 -dihydro- 1 H- pyrazolor3,4-dlpyrimidin-6-ylthio)methyl)pyridin-3-yl)formamide (A512 (23)): A mixture of 21 (40 mg, 0.1 mmol), diiodine (5 mg, 0.04 mmol) and formic acid (5 mL) stirred on 70°C oil bath for 3 hours. The reaction mixture was cooled to rt followed by adding Na2S2C>3 (aq) 5mL and Na2C(¾(aq) 20 mL, then extracted with ethyl acetate (25 mL x 3). The combined organic layers were collected, dried over Na2S04 and concentrated down under reduced pressure to give crude product, which was further crystallized from MeOH to yield the desired product A512 (23) (M+l)+ = 428.1 (35 mg, white crystals, 82%).
Example 7: Synthesis of 5,6-bis(4-methoxyphenyl)-3-((pyridin-3-yl)methylthio)-l,2,4- triazine (A535 (25)):
Figure imgf000045_0001
24 A535 (25)
Scheme 8 Synthesis of 5,6-bis(4-methoxyphenyl)-3-((pyridin-3-yl)methylthio)-l,2,4- triazine (A535 (25)):
[0142] Synthesis of 5,6-bis(4-methoxyphenyl)-3-((pyridin-3-yl)methylthio)-l,2,4-triazine (A535 (25)): A mixture of 24 (98 mg, 0.3 mmol) and 3-(bromomethyl) pyridine 12 (55 mg, 0.33 mmol) in acetone (3 mL) with K2CO3 (138 mg, 1 mmol) was heated to refluxing and stirred for 3 h. The reaction mixture was cooled to rt and then water (20 mL) was added. The mixture was extracted with ethyl acetate (3x 20 mL). The combined organic layers were washed with saturated NaHC(¾ solution and water, dried over Na2S04 and concentrated down under reduced pressure to give crude product, which was further crystallized from MeOH to yield the desired product A535 (25) (M+l)+ = 417 (145 mg, white crystals, 70%). Example 8: Synthesis of 5-(2, 6-difluorophenyl)-6-(pyridin-3-ylmethylthio)-lH-pyrazolor3,4- dlpyrimidin-4(5H)-one (A565 (27)):
Figure imgf000046_0001
A565 (27)
Scheme 9: 5-(2,6-difluorophenyl)-6-(pyridin-3-ylmethylthio)-lH-pyrazolo[3,4- d]pyrimidin-4(5H)-one (A565 (27))
[0143] Synthesis of 5-(2,6-difluorophenyl)-6-thioxo-6,7-dihydro-lH-pyrazolor3,4- dlpyrimidin-4(5H)-one(26). A mixture of ethyl 5-amino-lH-pyrazole-4-carboxylate (755 mg, 5 mmol) and l,3-difluoro-2-isothiocyanatobenzene (855 mg, 5 mmol) in dry toluene 5mL was refluxed for 6h, then cooled. Intermediate was precipitated spontaneously and were collected as pure products. A suspension of intermediate in 4% aqueous sodium hydroxide (4 mL) was refluxed until no more of the starting material could be detected by TLC or LC-MS (l-2h). After cooling, the solution was neutralized with 4 hydrochloricacid and then extracted with ethyl acetate (3 x20mL). After drying over anhydrous magnesium sulfate, the solvent was removed and the white solid residue was purified by flash column chromatography to give pure product (M+l)+ = 281 (1.226g, white powder, 87.6%).
[0144] The procedure for the Synthesis of 5-(2, 6-difluorophenyl)-6-(pyridin-3- ylmethylthio)-lH-pyrazolo[3,4-d]pyrimidin-4(5H)-one (A565 (27)) [(M+l)+ = 372.5] is the same as that of A512 (13).
[0145] Using similar chemistry, the following compound A566 (28), A567 (29), A568 (30), A569 (31), A570 (32) was also synthesized.
Figure imgf000047_0001
A569 (31) A570 (32)
Example 9: Synthesis of 4-imino-l-phenyl-5-(2,3,6-trifluorophenyl)-4,5-dihydro-lH- pyrazolor3,4-dlpyrimidine-6(7H)-thione (A575 (33)):
Synthesis of 4-imino-l-phenyl-5-(2,3,6-trifluorophenyl)-4,5-dihydro-lH-pyrazolor3,4- dlpyrimidine-6(7H)-thione (A575 (33)). A mixture of 5-amino-l -phenyl- lH-pyrazole-4 carbonitrile (218 mg, 1 mmol) and l,3,4-trifluoro-2-isothiocyanatobenzene (189 mg, 1 mmol) was refluxed for 2h,and then cooled. Intermediate was precipitated spontaneously and were collected as pure products (M+l)+ = 408 (325 mg, white solid, 80%). Then using the same procedure as that of A348 (13) to synthesis compound (A575 (33)) (M+l)+ = 498.
Figure imgf000047_0002
n erme a e
Figure imgf000047_0003
A575 (33) Example 10: Synthesis of 3-methyl-6-(p-tolylethvnyl)-5-(2,3,6-trifluorophenyl)-lH- pyrazolor3,4-dlpyrimidin-4(5H)-imine (A571 (36)):
Figure imgf000048_0001
Scheme 10, Synthesis of 3-methyl-6-(p-tolylethynyl)-5-(2,3,6-trifluorophenyl)-lH- pyrazolo[3,4-d]pyrimidin-4(5H)-imine (A571 (36))
[0146] Synthesis of 3-methyl-6-(p-tolylethynyl)-5-(2,3,6-trifluorophenyl)-lH-pyrazolor3,4- dlpyrimidin-4(5H)-imine (A571 (36)): A mixture of Ar-Br (107mg, 0.3 mmol), l-ethynyl-4- methylbenzene (38 mg, 0.33 mmol), Pd(PPh3)4 (35 mg, 0.03 mmol), Cul (6 mg, 0.03 mmol) and DIPA (8mg, 0.6 mmol) were refluxed in dry DMF 2mL for about 5 hours. The reaction was detected by TLC/LC-MS. After completed, extract with EA, remove solvent and purified through column chromatography on silica to give pure product (A571 (36)) (M+l)+ = 394 (65 mg, 55%).
Example 11: Synthesis of 5-amino-3-bromo-lH-pyrazole-4-carbonitrile (42) and 5-amino-3- chloro-lH-pyrazole-4-carbonitrile (43) and analogs A537 (44), A542 (45), A547 (46), A558 (47):
[0147] Synthesis of 5-amino-3-bromo-lH-pyrazole-4-carbonitrile (42) and 5-amino-3- chloro- 1 H-pyrazole-4-carbonitrile (43) : To a solution of 5-amino-lH-pyrazole-4-carbonitrile (540mg, 5 mmol) in dry MeCN 20mL, NBS (1209mg, 6.5 mmol) or NCS ( 920mg, 6.5 mmol) was added bathwise at room-temperature. The mixture was stirred forl2 hours, and detected by TLC/LC-MS. After completed, extract with EA, remove solvent and purified through column chromatography (EA:Hex, 1;1) on silica to give pure product(42) (M+l)+ = 187 (460mg, yellow solid, 49% yield), (43) (M+l)+ = 143 (320mg, white solid, 45% yield).
[0148] Using similar chemistry as Scheme 11 shows, the following compound A537 (44), A542 (45), A547 (46), A558 (47) were also synthesized.
Figure imgf000049_0001
Scheme 11: Synthesis of 5-((3-bromo-4-imino-5-(2,3,6-trifluorophenyl)-4,5-dihydro-lH- pyrazolo[3,4-d]pyrimidin-6-ylthio)methyl)pyridin-3-amine and derivates A537 (44), A542 (45), A547 (46), A558 (47)
Example 12: Synthesis of 5-(2,6-difluorophenyl)-6-((5-methylpyridin-3-yl)methylsulfonyl)-lH- pyrazolor3.4-dlpyrimidin-4(5H)-imine (A514 (48)) and (A513 (49)):
Figure imgf000049_0002
Scheme 12: Synthesis of 5-(2,6-difluorophenyl)-6-((5-methylpyridin-3-yl)methylsulfonyl)- lH-pyrazolo[3,4-d]pyrimidin-4(5H)-imine (A514 (48)) and 5-(2,6-difluorophenyl)- 6-((5-methylpyridin-3-yl)methylsulfinyl)-lH^yrazolo[3,4-d]pyrimidin-4(5H)-imine (A513 (49))
[0149] Synthesis of 5-(2,6-difluorophenyl)-6-((5-methylpyridin-3-yl)methylsulfonyl)-lH- pyrazolor3,4-dlpyrimidin-4(5H)-imine (A514 (48)) and (A513 (49)): Using similar chemistry as Schemes 4 and 5, compounds A514 (48), A513 (49) were also synthesized.
Biological screening and anticancer activity:
[0150] Examples demonstrating the anti-proliferation and anti-migration effects of the compounds of the invention are described as below.
In vitro cell-based screening using real-time cell electronic sensing (RT-CES) system
[0151] The heterocyclic compounds in the present invention are developed for the anticancer activities for cancer cells with certain molecular targets, i.e. , cMet. The anticancer efficacy of these heterocyclic compounds and their analogues described above were
preliminarily screened in vitro using a penal of cMet- addicted cancer cell lines by real time electronic cell sensing (RT-CES) system from ACEA Biosciences, Inc. (or xCELLigence system from Roche Applied Sciences/ ACEA Biosciences Inc.) , which provides dynamic cell response information after exposing to cMet inhibitors.
[0152] The details of this cell electronic sensing technology, called real-time cell electronic sensing (RT-CES®) and associated devices, systems and methods of use are described in United States patent number 7,732,127; patent number 7,192,752; patent number 7,459,303; patent number 7,468,255; patent number 7,470,533; patent number 7,560,269; United States provisional application number 60/397,749, filed on July 20, 2002; United States provisional application number 60/435,400, filed on December 20, 2002; United States Provisional application 60/469,572, filed on May 9, 2003, PCT application number PCT/US03/22557, filed on July 18, 2003; PCT application number PCT/US03/22537, filed on July 18, 2003; PCT application number PCT/US04/37696, filed on November 12, 2004; PCT application number PCT/US05/04481, filed on February 9, 2005; United States patent application number
10/705,447, filed on November 10, 2003; United States patent application number 10/705,615, filed on November 10, 2003; United States patent application number 10/987,732, filed on November 12, 2004; United States patent application number 11/055,639, filed on
February 9, 2005, each of which is incorporated by reference. Additional details of RT-CES technology is further disclosed in United States provisional application number 60/519,567, filed on November 12, 2003, and United States provisional application number 60/542,927, filed on February 9, 2004, United States provisional application number 60/548,713, filed on February 27, 2004, United States provisional application number 60/598,608, filed on
August 4, 2004; United States provisional application number 60/598,609, filed on
August 4, 2004; United States provisional application number 60/613,749, filed on
September 27, 2004; United States provisional application number 60/613,872, filed on September 27, 2004; United States provisional application number 60/614,601, filed on September 29, 2004; United States provisional application number 60/630,071, filed on November 22, 2004; United States provisional application number 60/630,131, filed on
November 22, 2004, each of which is incorporated herein by reference.
[0153] For measurement of cell-substrate or cell-electrode impedance using RT-CES technology, microelectrodes having appropriate geometries are fabricated onto the bottom surfaces of microtiter plate or similar device, facing into the wells. Cells are introduced into the wells of the devices, and make contact to and attach to the electrode surfaces. The presence, absence or change of properties of cells affects the electronic and ionic passage on the electrode sensor surfaces. Measuring the impedance between or among electrodes provides important information about biological status of cells present on the sensors. When there are changes to the biological status of the cells analogue, electronic readout signals are measured automatically and in real time, and are converted to digital signals for processing and analysis.
[0154] In a RT-CES system, a cell index is automatically derived and provided based on measured electrode impedance values. The cell index obtained for a given well reflects: 1) how many cells are attached to the electrode surfaces in this well; 2) how well cells are attached to the electrode surfaces in this well. Thus, the more the cells of same type in similar
physiological conditions attach the electrode surfaces, the larger the cell index. And, the better the cells attach to the electrode surfaces (e.g., the cells spread-out more to have larger contact areas, or the cells attach tighter to electrode surfaces), the larger the cell index. Impedance- based TCRP (time-dependent cell response profiling) has shown to be extremely useful in predicting the mechanism of compound action (Abassi, Y. A., et ah, Chem. Biol. (2009) 16:712- 723). Oncogene addiction refers to the acquired dependency of cancer cells on a single cellular pathway for survival or sustained proliferation, despite the fact that such cells have accumulated numerous genetic alterations. We have discovered that the c-Met-addicted cell lines would produce a signature impedance-based TCRP, when treated with positive-control c-Met inhibitors.
[0155] Through the use of the RT-CES system, the heterocyclic compounds described in the examples above have been shown to produce a similar impedance-based TCRP on RT-CES system to that generated by positive control inhibitors (Figure 1-2). The EC50 calculated based on the short-term TCRP correlates well with the IC50 calculated based other cell based assay (e.g. , Western and ELISA analysis evaluating cMet inhibition). These compounds have also been shown to inhibit HGF (hepatocyte growth factor) -induced cell migration (Figure 4-5). In addition, these compounds showed no or negligible effects on non-cMet addicted cancer cell lines (Figure 3).
Example 13 Bioactivity of heterocyclic compounds on cMet- addicted cell lines as determined using real-time cell electronic sensing (RT-CES) system
[0156] H1993 and GTL16 have previously been reported as cMET addicted cell lines (Benvenuti, S., et ah, Cancer Res. (2011) 71 : 1945-1955). We used these two cell lines to assay specific inhibitory effects of the heterocyclic compounds described in the examples above on cMet. H1993 and GTL16 were seeded into wells of 96 well E-plate devices (Roche) with initial seeding density of 10,000 cells per well and were pre-incubated in incubator under standard cell culture condition for about 24 hours. The heterocyclic compounds at different concentration in DMSO were added into wells following the incubation period. Negative control wells were treated with buffer solutions containing DMSO only. Figure 1 shows the TCRP (time- dependent cell response profiling) of c-Met addicted HI 993 cells to the treatment of various heterocyclic compounds. The dose-dependent rapid impedance (corresponding to normalized cell index in the y-axis) increase and TCRP are the same as those observed for H1993 cells when treated with cMet inhibitor control compounds (results not shown). The cell index was normalized against the cell index values at a time point just before compound addition. The calculated effective concentrations EC50 (2 hr post compound treatment) in this specific experiment is 220 nM, 179 nM, 217 nM and 154 nM for compound A424 (19),
compound A418 (20), compound A447 (21) and compound A448 (22), respectively. Figure 2 shows the TCRP (time-dependent cell response profiling) of c-Met addicted GTL16 cells to the treatment of various heterocyclic compounds. The dose-dependent rapid impedance
(corresponding to normalized cell index in the y-axis) increase and TCRP are the same as those observed for GTL16 cells when treated with cMet inhibitor control compounds (results not shown). The calculated effective concentrations EC50 (2 hr post compound treatment) in this specific experiment is 137 nM, 141 nM, 138 nM and 109 nM for compound A424 (19), compound A418 (20), compound A447 (21) and compound A448 (22), respectively. The EC50 calculated based on the short-term TCRP correlates well with the IC50 calculated based other cell based assay (EXAMPLE 17).
[0157] Results for the compounds tested in these assays are presented in Table 1-11 as an average of results obtained. The EC50S (in H1993) toward cMet inhibition for selected compounds of this invention are listed in Tables 1-11. Compounds were tested in free base form. Where activity is shown as greater than (>) a particular value, the value is the highest concentration tested.
Figure imgf000054_0001
Compound No. Cyclo A Cyclo B Cyclo C EC50
Figure imgf000054_0002
A348 (13) H 1,5-di-F R=Me 0.5-1.3uM
A351 H 1,4-di-F R=Me >20uM
A352 H H R=Et >20uM
A353 H 3-OMe R=Me 12.0uM
A354 H 1,3,4-tri-F R=Me >20uM
A356 H 1-F R=Et >20uM
A357 1,4-di-F R=Me >20uM
A358 1,4-di-F R=Me >20uM
A359 1,3,4-tri-F R=Me >20uM
A361 2,3,4-tri-F R=Me >20uM
A362 2,3,4-tri-F R=Me >20uM
A363 2,4-di-F R=Me >20uM
A364 2,4-di-F R=Me >20uM
A365 2,4-di-F R=Me >20uM
A366 2,3-di-F R=Me >20uM
A367 2,3-di-F R=Me >20uM
Figure imgf000054_0003
Figure imgf000055_0001
Compound Cyclo A Cyclo B Cyclo C EC50
No.
A368 2-F,4-Me R=Me >20uM
Figure imgf000055_0002
A373 H 1-F R=Me >20uM
A374 H 2-F R=Me >20uM
A375 H 3-Me R=Me >20uM
A376 H 1,3-di-F R=Me >20uM
A377 H l,5-2Me R=Me >20uM
A379 H 1-Cl R=Me >20uM
A382 2-Cl 1,5-di-F R=Me 12.7uM
A383 H 1-F,4-Me R=Me 18.4uM
A385 H 1-F.4-C1 R=Me >20uM
A386 H 1-Me,3-F R=Me 11.7uM
A389 H N=\ "^L R=Me >20uM
A391 H 1-C1.3-F R=Me >20uM
A393 H 1-OMe R=Me >20uM
A395 H 1-N02 R=Me >20uM
A396 H 3-Cl R=Me >20uM
Figure imgf000055_0003
Figure imgf000056_0001
Compound Cyclo A Cyclo B Cyclo C EC50
No.
Figure imgf000056_0002
A409 H 1,2,5-tri-F R=Me 0.25-0.55uM
A411 H 1.5-2C1 R=Me 4.4uM
Figure imgf000056_0003
A413 H 1-F.4-C1 R=Me >20uM
A414 H l,4-20Me R=Me >20uM
A415 H 1-F,4-Br R=Me >20uM
A416 H 1-F.5-C1 R=Me 4.6uM
A417 ¾ rF 1-F.5-C1 R=Me >20uM
A418 (20) 3-Me 1,5-di-F R=Me 0.16-0.51uM
A419 3-Me H R=Me 5.9uM
Figure imgf000056_0004
A422 1,5-di-F R=Me 12.4uM
A423 1,5-di-F R=Me >20uM
Figure imgf000056_0005
A424 (19) 3-Me 1,2,5-tri-F R=Me 0.13-0.64uM TABLE 4
Figure imgf000057_0001
Compound Cyclo A Cyclo B Cyclo C EC50
No.
A425 1,2,5-tri-F R=Me 18.5uM
Figure imgf000057_0002
A437 3-F 1,5-di-F R=Me 3.4uM
A438 3-Br 1,5-di-F R=Me 2.8uM
A439 3-Cl 1,5-di-F R=Me 2.7uM
A440 3-CF3 1,5-di-F R=Me 11.7uM
\ /
A441 1,5-di-F R=Me >20uM
A442 3-OMe 1,5-di-F R=Me lO.luM
A443 3-OH 1,5-di-F R=Me 4.4uM
A444 3-cyclopropyl 1,5-di-F R=Me >20uM
A445 3-Et 1,2,5-tri-F R=Me 2.9uM
A446 3-Et 1,5-di-F R=Me 3.5uM
Figure imgf000058_0001
Compound Cyclo A Cyclo B Cyclo C EC50 No.
Figure imgf000058_0002
2,3-CH==CH-
A452 1,5-di-F R= =Me >20uM
CH==CH
2,3-CH==CH- 1,2,5-tri-
A453 R= =Me >20uM
CH==CH F
Figure imgf000058_0003
A458 3-NH2,4-Me 1,5-di-F R= =Me >20uM A459 2- NH2 1,5-di-F R= =Me >20uM
A460 1,5-di-F R= =Me >20uM
Figure imgf000058_0004
A461 2,3-2Me 1,5-di-F R= =Me >20uM A462 1-NH2,3-Me 1,5-di-F R= =Me >20uM
1,2,5-tri-
A463 2,3-2Me R= =Me >20uM
F
A464 3- C02Me 1,5-di-F R= =Me >20uM
1,2,5-tri-
A465 1- NH2,3-Me R= =Me 9.5uM
F
A467 2- Me 1,5-di-F R= =Me >20uM
1,2,5-tri-
A468 2- Me R= =Me >20uM
F
A469 4- Me 1,5-di-F R= =Me >20uM A470 3- NHMe 1,5-di-F R= =Me 3.6uM A471 1,5-di-F R= =Me >20uM
A472 1,5-di-F R= =Me 14.9uM
Figure imgf000058_0005
TABLE 6
Figure imgf000059_0001
Compound Cyclo A Cyclo Cyclo EC50
No. B C
A473 3-Ph 1,5-di-F R=Me >20uM A474 3-NHCOCH3 1,5-di-F R=Me 1.5uM
A475 1,5-di-F R=Me 2.6uM
A476 1,5-di-F R= =Me >20uM
Figure imgf000059_0002
A477 3-COOH 1,5-di-F R= =Me >20uM
A479 3-CN 1,5-di-F R= =Me >20uM
A480 3-NHCONH2 1,5-di-F R= =Me 2.4uM
A481 H 1,5-di-F R= =H 8.8uM
A482 3-Me 1,5-di-F R= =H 3.5uM
A483 3-CH2NH2 1,5-di-F R= =Me >20uM
Figure imgf000059_0003
TABLE 7
Figure imgf000060_0001
Compound Cyclo A Cyclo B Cyclo C
No.
A299 CH- H R=H >20uM
A300 H R=H 13.4uM
A301 t>i- 3-Cl R=H >20uM
Figure imgf000060_0002
A303 3-Me R=H >20uM
A304 Ol- 3-Me R=H >20uM
Figure imgf000060_0003
A306 Oh H R=H >20uM
Figure imgf000060_0004
A313 MeO-Q-f- H R=Me >20uM
>20uM
A314 H R=Me
TABLE 8
Figure imgf000061_0001
Compound Cyclo A Cyclo B Cyclo C EC50
No.
A315 3-Br R=Me >20uM
A316 H R=Me >20uM
A377 CM- 3-Br R=Me >20uM
A318 σι- 3-Br R=Me >20uM
A319 CN- - 1-F R=Me 10.8uM
A320 CN-I- 1-F R=Me >20uM
A321 CN- - 2-F R=Me 15.0uM
A322 CN-I- 3-F R=Me 15.1uM
A323 CN-I- 2-F R=Me >20uM
A324 H R=Me >20uM
A325 ipr !- 3-F R=Me >20uM
A326 3-Cl R=Me 4.4uM
A327 ipr -h 3-Cl R=SMe >20uM
'Pr s
2- Cl,
A328 CN-I- R=Me About 1
3- F
A329 2-Cl, 3-F R=Me >20uM
Pr 5
A330 CN-I- 1,3-di-F R=Me >20uM
TABLE 9
Figure imgf000062_0001
Compound Cyclo A Cyclo B Cyclo C EC50 No.
A331 1,3-di-F R= :Me >20uM
Pr 6
Et
A332 1,3-di-F R= =Me >20uM
E.- -
A333 ipr -h 1,5-di-F R= =Me >20uM
Et
A334 1,5-di-F R= =Me >20uM
Ε,·Ν- A335 0-4- 1,5-di-F R= =Me 11.9uM A336 0.4- 1,4-di-F R= =Me 13.1uM
Figure imgf000062_0002
A338 Q^N- - H R= =Me >20uM A341 0-4- 3-Me R= =Me >20uM A342 0-4- 2-CF3 R= =Me >20uM A343 0-4- 3-CF3 R= =Me >20uM A344 0-4- 3-OMe R= =Me 9.4uM
Figure imgf000062_0003
A350 0-4- H R= =Et >20uM
Figure imgf000062_0004
TABLE 10
Figure imgf000063_0001
Compound Cyclo A Cyclo B Cyclo C EC50 No.
A370 Oh 2,3-di-F R=Me 12.1uM
A378 Oh l,5-2Me R=Me >20uM
A380 1-2C1 R=Me 15.6uM
A381 1,5-di-F R=Me >20uM
A384 CN-I- 1-F.4-C1 R=Me >20uM
A387 CN-I- 3-Br R=Me >20uM
Figure imgf000063_0002
A390 Oh 1-C1.3-F R=Me >20uM
A392 Oh 1-OMe R=Me 15.1uM
A394 CN-I- 2-N02 R=Me >20uM
A397 Oi- 3-Cl >20uM
V-
/
A398 0-4- 3-Cl >20uM
A404 1,5-di-F >20uM
V-
A405 Oi- 1,5-di-F R=Me >20uM TABLE 11
Figure imgf000064_0001
Compound Cyclo A Cyclo B Cyclo C EC50
Figure imgf000064_0002
Example 14 Bioactivity of heterocyclic compounds on cMet-non- addicted cell lines as
determined using real-time cell electronic sensing (RT-CES) system
[0158] To examine whether the heterocyclic compounds produce any bioactivity effects on cMet-non-addicted cell lines and to verify that the effect of these compounds on cMet- addicted cells was specific, A549 cells were treated with various heterocyclic compounds described above. A549 were seeded into wells of 96 well E-plate devices (Roche) with initial seeding density of 5,000 cells per well and were pre-incubated in incubator under standard cell culture condition for about 24 hours. The heterocyclic compounds at different concentration in DMSO were added into wells following the incubation period. Negative control wells were treated with buffer solutions containing DMSO only. Figure 3 shows the TCRP of cMet-non- addicted A549 cells to the treatment of various heterocyclic compounds. No obvious effects were observed for A549 cells when treated with these compounds (compound A348 (13), compound A424 (19), compound A418 (20), compound A447 (21) and compound A448 (22)). This suggests that their effect on cMet addicted cells was specific to cMet inhibition.
Example 15 Inhibitory effect of heterocyclic compounds on HGF-mediated migration of
A431 cells as determined using real-time cell electronic sensing (RT-CES) system
[0159] To validate and confirm the cMet-inhibitory effects of various heterocyclic compounds described above, HGF-mediated migration of A431 cells was performed in the presence of different heterocyclic compounds. The technical details, instrument, devices of cell migration/invasion assays with real-time cell electronic sensing technology can be found in United States patent number 7,459,303. Briefly, A431 cells were starved for 6 hr and then incubated with compounds for 1 hr before seeding to fibronectin precoated CIM device (Roche). HGF at 50 ng/ml was used to mediate migration of A431 from upper chamber to lower chamber. The time course of the migration was monitored.
[0160] Table 12 below shows IC50 values derived from the impedance response profile of HGF-mediated migration of A431 cells in the presence of different compounds described in the present invention. Figure 4 shows that these heterocyclic compounds inhibited HGF-mediated A431 migration in a dose-dependent manner. The positive control compound, PF02341066 (HGFi) inhibited A431 migration in a similar dose-dependent manner. These results support the hypothesis that these compounds are cMet (HGFR) inhibitors. The IC50 values were derived from the dose responses at 10, 20 and 30 hrs after migration experiments started.
Table 12
Figure imgf000065_0001
Example 16 Inhibitory effect of heterocyclic compounds on HGF-mediated migration of
HUVECs as determined using real-time cell electronic sensing (RT-CES) system
[0161] To further validate and confirm the cMet-inhibitory effects of various heterocyclic compounds described above, HGF-mediated migration of HUVECs (human umbilical vein endothelial cells) was performed in the presence of a series of concentrations of different heterocyclic compounds. The technical details, instrument, devices of cell migration/invasion assays with real-time cell electronic sensing technology can be found in United States patent number 7,459,303. Briefly, HUVECs were starved for 6 hr and then incubated with compounds for 1 hr before seeding to fibronectin precoated CIM device (Roche). HGF at 50 ng/ml was used to mediate migration of HUVECs from upper chamber to lower chamber. The time course of the migration was monitored. [0162] Table 13 below lists IC50 values derived from the impedance response profile of HGF-mediated migration of HUVECs in the presence of different compounds described in the present invention. Figure 5 shows that these heterocyclic compounds inhibited HGF-mediated HUVECs migration in a dose-dependent manner. The positive control compound, PF02341066 (HGFi) inhibited HUVECs migration in a similar dose-dependent manner. These results further support the notion that these compounds are cMet (HGFR) inhibitors. The IC50 values were derived from the dose responses at 6 hrs and 12 hrs after migration experiments started.
Table 13
Figure imgf000066_0001
Example 17 Heterocyclic compounds show inhibitory effect on cMet phosphorylation both in vivo and in vitro
[0163] To further validate and confirm the cMet-inhibitory effects of various heterocyclic compounds described above, we employed other approaches (not RT-CES based assays). We first tested if compound 13 inhibited cMet phosphorylation in A549 cells stimulated by HGF. Briefly, A549 cells were first treated with compounds in serum free medium for 2 hours, then stimulated with 100 ng/mL HGF for 30 min. Cells were lysed and the lysates were used for detection of the total phospho-cMet using an ELISA kit (R&D system, DYC2480-2). The results (Figure 6a) show that compound A348 (13) inhibited cMet phosphorylation in a dose dependent manner. The calculated IC50 is 332.5 nM. In comparison, the calculated IC50 for the positive control (HGFi, PF02341066) is 2.25 nM.
[0164] We also evaluated the inhibitory effect of the heterocyclic compounds on constitutively active cMet in GTL16 (one of cell lines used in RT-CES assay) using Western analysis. Figure 6b shows that cMet phosphorylation at Y1234/1235 was inhibited by compound 13 treatment in a dose-dependent fashion. The intensities of the phospho-cMet bands were quantified and used for IC50 calculation. The calculated IC50 for compound A348 (13) is 563 nM, similar to that derived from ELISA assay, as well as migration assay (Table 12, compound A348 (13)). In addition, we evaluated the potency of compound A447 (21), compound A418 (20) and compound A424 (19) in GTL16 using Western analyses. The calculated IC50 is 38.8 nM, 74.7 nM and 36.2 nM for compound A 447 (21), compound A418 (20) and compound A424 (19) and, respectively. Furthermore, we tested the activities of the heterocyclic compounds on the enzymatic activities of purified cMet protein (experiments were performed at Reaction Biology Corporation). At 10 μΜ ATP concentration, the IC50 is 494.9 nM, 10.8 nM, 4.01 nM and 1.21 nM for compound A348 (13), compound A447 (21), compound A418 (20) and compound A424 (19), respectively. The potency ranking agrees well with that derived from our RT-CES based assay. Taken together, there is strong evidence that the heterocyclic compounds described herein are cMet inhibitors.
EXAMPLE 18 Heterocyclic compounds show inhibitory effect on other kinase targets in vitro
[0165] KINOMEscan is the industry's most comprehensive high-throughput system for screening compounds against large number of human kinases (Fabian et al. (2005) Nat.
Biotechnol. 23, 329; Karaman et al. (2008) Nat. Biotechnol. 26, 127). To evaluate the activities of these heterocyclic compounds on other kinase targets, we have performed KINOMEscan on all 442 available kinases at Ambit Biosciences. Table 14 lists the 42 kinases that were inhibited by cpd20 (10 μΜ) by more than 65%.
Table 14
Figure imgf000068_0001
[0166] To confirm these results using activity based assay, we also tested the effects of compound A418 (20), compound A424 (19) and compound A447 (21) on the kinase activities of Aurora kinases, Jak2 and ROS l . At 10 uM concentration, all these kinase are effectively inhibited. Table 15 lists the % enzyme activities of these kinases in the presence of 10 μΜ compound A418 (20), compound A424 (19) and compound A447 (21) at a given ATP concentration.
Table 15
Figure imgf000069_0001
EXAMPLE 19 Heterocyclic compounds show inhibitory effect on RQS 1
phosphorylation in vivo
[0167] ROSl transcripts have been detected in several glioblastoma cell lines, e.g., U-118 MG, U-138 MG and SW-1088, but not in A-172 and A-382 (Fabian et al. (1987) Proc. Nati. Acad. Sci. USA 84, 9270-9274). We have tested the effect of these heterocyclic compounds on ROSl phosphorylation in these glioblastoma cell lines. The results from Western analyses confirm that ROSl was expressed in U138 MG, not in A-172. In addition, the Western results show that at 10 μΜ concentration, compound A348 (13), compound A418 (20), compound A424 (19) and compound A447 (21) inhibited ROSl phosphorylation after 1 hour compound treatment (Figure 7).

Claims

CLAIMS:
1.
Figure imgf000070_0001
Formula la Formula lb where in a ring indicates the ring is an aromatic or heteroaromatic ring;
W1, W2 W3 , W4 are each independently absent, N, NH, NR1, O, S, CH, or CR2,
provided at least one of W1, W2 W3 , and W4 is CH or CR2,
not more than one of them is absent,
and not more than one of them is O or S;
R' is Ci-8 alkyl, C2-8 alkenyl, C2-8 alkynyl, aryl, or heteroaryl, wherein each alkyl, alkenyl, alkynyl, aryl and heteroaryl is optionally substituted;
Z is S, SO, S02, SO2NH, SO2NR3, NHSO2, NR1, CR1]*2, NR1, or O;
Y is (CHR4)n wherein n = 1-5 or a C3-C7 carbocyclic or aromatic divalent moiety;
A is a bond (absent), C(=0), NH, NR5, C(0)NH, C(0)NR6, C(S)NH, C(S)NR6, NHC(O), NR6C(0), NHSO2, NR6S02, SO2NH, SO2NR6, or OP(=0)(OR7);
Het is substituted or unsubstituted Ci-8 alkyl, C2-8 alkenyl, C2-8 alkynyl, aryl, fused aryl, heteroaryl, fused heterocycle, a saturated or unsaturated carbocyclic ring or a heterocyclic ring, or a group of the formula -(CH2)q-Q where q is 1-4 and Q is an aryl, heteroaryl, cycloalkyl or heterocycle and Q is optionally substituted,
where each Het is optionally substituted and may contain a heteroatom selected from N, O and S in place of a ring or chain carbon atom of Het;
each of R1, R2, R3, R4, R5, R6, and R7 is independently selected from H, OH, Halo, NHR, NRR, OR, SR, COOR, C(=0)R, CN, CF3, OCF3, N02, OC(0)R, S03R, PO3R2,
CR(COOR)2;
each R is independently selected from H, halo, ORx, SRX, C02Rx, C(0)NRx 2, C(=0) Rx, CN, CF3, OCF3, N02, NRX2, OCORx, S03Rx, P03Rx 2, and CH(COORx)2;
where each Rx is H or C1-C4 alkyl or C1-C4 haloalkyl,
or R can be substituted or unsubstituted Ci-8 alkyl, C2-8 alkenyl, C2-8 alkynyl, aryl, fused aryl, heteroaryl, fused heterocycle, a C3-8 carbocyclic ring or a C3-8 heterocyclic ring, saturated or unsaturated, wherein each Ci-8 alkyl, C3_g cyclic alkyl, C2-8 alkenyl, C2-8 alkynyl can optionally contain a heteroatom selected from N, O and S in place of a carbon atom; p is 0-3;
each R10 is H or acyl;
and two R, R , R , R , R , R , R , or R on the same or adjacent atoms can optionally be linked together to form a 3-8 membered ring that can contain up to two heteroatoms selected from N, O and S as ring members and which is optionally substituted;
or a pharmaceutically acceptable salt thereof.
2. The compound of claim 1, wherein W4 is CR2 and W3 is a bond.
3. The compound of any one of the preceding claims, wherein W1 is NR1.
4. The compound of any of the preceding claims, wherein R10 is H.
5. The compound of any of the preceding claims, wherein Z is S, SO, SO2, SO2NH, SO2NR3, NHSO2, NR1, CR^2 NR1, or O;
6. The compound of any one of the preceding claims, wherein Y is (0¾)ι^.
7. The compound of any of the preceding claims, wherein A is a bond or C(=0).
8. The compound of any one of the preceding claims, Het is a 5 or 6 membered aromatic or heteroaromatic ring.
9. The compound of any one of the preceding claims, wherein Het is phenyl or pyridyl and is optionally substituted with up to three R groups.
10. The compound of any one of the preceding claims, wherein W4 is CR2, wherein R2 is H, CI -4 alkyl, or CI -4 haloalkyl.
11. The compound of any of the preceding claims, wherein R' is optionally substituted phenyl.
12. The compound of any one of the preceding claims, wherein W2 is N. A com ound of Formula Ila and Formula lib
Figure imgf000072_0001
Ila lib
wherein:
W1 is NR1, O or S;
W2 is N or CR2;
Ar is a 5-6 membered aryl or heteroaryl ring that can be substituted or unsubstituted; and
Figure imgf000072_0002
R2, R10, Z, Y, A, and Het are as defined in claim 1 ;
or a pharmaceutically acceptable salt thereof.
14. The compound of claim 13, wherein R10 is H.
15. The compound of claim 13 or 14, wherein W1 is NR1.
16. The compound of any one of the claims 13-15, wherein W1 is O or S.
17. The compound of any one of the claims 13-16, wherein W2 is N or CR2.
18. The compound of any one of the claims 13-17, wherein Z is S, SO, SO2, SO2NH, SO2NR3, NHSO2, NR1, CR^2 NR1, or O;
19. The compound of any one of the claims 13-18, wherein Y is (CH^i^.
20. The compound of any one of the claims 13-19, wherein A is a bond or C(=0).
21. The compound of any one of the claims 13-21, Het is a 5 or 6 membered aromatic or heteroaromatic ring.
22. The compound of any one of the claims 13-21, wherein Het is phenyl or pyridyl and is optionally substituted with up to three R groups.
23. The compound of any one of the claims 13-22, wherein R2 is H, Ci_4 alkyl, or Ci_ 4 haloalkyl.
24. The compound of any one of the claims 13-21, wherein Ar is optionally substituted phenyl.
25. The compound of claim 13,
whe
Figure imgf000073_0001
wherein the wavy line across one non-cyclic bond indicates the bond that connects each Het group to A; where in a ring indicates the ring is an aromatic or heteroaromatic ring;
each W and W is independently a bond, C, CH, CH2, CHR12, CR12, N, NH, NR12, S, O, C=0, or C=S,
where each R is independently selected from H, halo, ORx, SRX, C02Rx, C(0)NRx 2, C(=0) Rx, CN, CF3, OCF3, N02, NRX2, OCORx, S03Rx, P03Rx 2, and CH(COORx)2, where each Rx is H or C1-C4 alkyl or C1-C4 haloalkyl,
or R can be substituted or unsubstituted Ci_g alkyl, C2_g alkenyl, C2_g alkynyl;
and each R12 is H or C1-C4 alkyl or C1-C4 haloalkyl,
or a pharmaceutically acceptable salt thereof.
26. The compound of claim 25, wherein R is selected from H, halo, ORx, S(0)tRx, C02Rx, C(0)NRX2, C(=0) Rx, CN, CF3, OCF3, N02, NRX 2, OCORx, S03Rx,
P03Rx 2, CH(COORx)2,
where each Rx is H or C1-C4 alkyl or C1-C4 haloalkyl, and each t is 0-2.
27. The compound of claim 25, wherein R is Ci_g alkyl, C3_g cyclic alkyl, C2-8 alkenyl, C2-8 alkynyl, an aryl, heteroaryl, a carbocyclic ring or a heterocyclic ring, each of which may contain a heteroatom selected from N, O and S in place of a carbon atom of the alkyl, alkenyl, alkynyl group.
28. A com ound of Formula Ilia and Formula Illb:
Figure imgf000074_0001
Ilia Illb
wherein:
W1 is NR1, O or S;
Z is S, SO, S02, SO2NH, SO2NR3, NHSO2, NR1, C^R2 NR1,
Y is -CH2- or -CH2-CH2- or -CH(Me)-;
A is a bond or C(=0);
R10 is H;
Ph is an optionally substituted phenyl group;
R, R1, R2, R3, and Het are as defined in claim 1 ;
or a pharmaceutically acceptable salt thereof.
29. The compound of claim 28,
where
Figure imgf000074_0002
wherein the wavy line across one non-cyclic bond indicates the bond that connects each Het group to A; where in a ring indicates the ring is an aromatic or heteroaromatic ring; each W and W is independently a bond, C, CH, CH2, CHR , CR , N, NH, NR , S, O, C=0, or C=S,
where each R is independently selected from H, halo, ORx, SRX, C02Rx, C(0)NRx 2, C(=0) Rx, CN, CF3, OCF3, N02, NRX2, OCORx, S03Rx, P03Rx 2, and CH(COORx)2, where each Rx is H or C1-C4 alkyl or C1-C4 haloalkyl,
or R can be substituted or unsubstituted Ci-8 alkyl, C2-8 alkenyl, C2-8 alkynyl;
and each R12 is H or C1-C4 alkyl or C1-C4 haloalkyl,
or a pharmaceutically acceptable salt thereof.
30. The compound of claim 29, wherein Het is phenyl or pyridyl and is optionally substituted with up to three groups selected from C1-C4 alkyl, C1-C4 haloalkyl, halo, ORx, SRX, C02Rx, C(0)NRX2, C(=0) Rx, CN, CF3, OCF3, N02, NRX 2, OCORx, S03Rx, P03Rx 2, and CH(COORx)2, where each Rx is H or C1-C4 alkyl or C1-C4 haloalkyl.
A c
Figure imgf000075_0001
wherein:
Het is as defined in claim 1 ;
n is 1-4;
R1, R2 and R3 is independently selected from H, OH, Halo, NHR', NR'R', OR', SR', COOR', C(=0)R', CN, CF3, OCF3, N02, OC(0)R', S03R', P03R'2,
CR'(COOR')2; each R' is independently selected from H, halo, ORx, SRX, C02Rx, C(0)NRX2, C(=0) Rx, CN, CF3, OCF3, N02, NRX 2, OCORx, S03Rx, P03Rx 2, and
CH(COORx)2;
where each Rx is H or C1-C4 alkyl or C1-C4 haloalkyl;
or a pharmaceutically acceptable salt thereof.
32. The compound of claim 31 ,
where
Figure imgf000076_0001
wherein the wavy line across one non-cyclic bond indicates the bond that connects each Het group to A; where in a ring indicates the ring is an aromatic or heteroaromatic ring;
each W and W is independently a bond, C, CH, CH2, CHR12, CR12, N, NH, NR12, S, O, C=0, or C=S,
where each R is independently selected from H, halo, ORx, SRX, C02Rx, C(0)NRx 2, C(=0) Rx, CN, CF3, OCF3, N02, NRX 2, OCORx, S03Rx, P03Rx 2, and CH(COORx)2, where each Rx is H or C1-C4 alkyl or C1-C4 haloalkyl,
or R can be substituted or unsubstituted Ci-8 alkyl, C2_8 alkenyl, C2_8 alkynyl;
and each R12 is H or C1-C4 alkyl or C1-C4 haloalkyl,
or a pharmaceutically acceptable salt thereof.
33. The compound of claim 32, wherein Het is phenyl or pyridyl and is optionally substituted with up to three groups selected from C1-C4 alkyl, C1-C4 haloalkyl, halo, ORx, SRX, C02Rx, C(0)NRX2, C(=0) Rx, CN, CF3, OCF3, N02, NRX 2, OCORx, S03Rx, P03Rx 2, and CH(COORx)2, where each Rx is H or C1-C4 alkyl or C1-C4 haloalkyl.
34. A compound of Formula Va and Formula
Figure imgf000077_0001
Formula Va Formula Vb wherein:
Z is S, SO, S02, Ci^R2 NRJR2, or O;
A is a bond, C=0, CONH or NHCO;
Het is as defined for Formula I;
n is 1-4;
R1, R2 and R3 is independently selected from H, OH, Halo, NHR', NR'R',
OR', SR', COOR', C(=0)R', CN, CF3, OCF3, N02, OC(0)R', S03R', P03R'2,
CR'(COOR')2;
each R' is independently selected from H, halo, ORx, SRX, C02Rx,
C(0)NRX2, C(=0) Rx, CN, CF3, OCF3, N02, NRX 2, OCORx, S03Rx, P03Rx 2, and
CH(COORx)2;
where each Rx is H or C1-C4 alkyl or C1-C4 haloalkyl;
or a pharmaceutically acceptable salt thereof.
35. The compound of claim 34,
where
Figure imgf000077_0002
wherein the wavy line across one non-cyclic bond indicates the bond that connects each Het group to A; where in a ring indicates the ring is an aromatic or heteroaromatic ring;
each W and W is independently a bond, C, CH, CH2, CHR12, CR12, N, NH, NR12, S, O, C=0, or C=S,
where each R is independently selected from H, halo, ORx, SRX, C02Rx, C(0)NRx 2, C(=0) Rx, CN, CF3, OCF3, N02, NRX2, OCORx, S03Rx, P03Rx 2, and CH(COORx)2, where each Rx is H or C1-C4 alkyl or C1-C4 haloalkyl,
or R can be substituted or unsubstituted Ci-8 alkyl, C2-8 alkenyl, C2-8 alkynyl;
and each R12 is H or C1-C4 alkyl or C1-C4 haloalkyl,
or a pharmaceutically acceptable salt thereof.
36. The compound of claim 35, wherein Het is phenyl or pyridyl and is optionally substituted with up to three groups selected from C1-C4 alkyl, C1-C4 haloalkyl, halo, ORx, SRX, C02Rx, C(0)NRX2, C(=0) Rx, CN, CF3, OCF3, N02, NRX 2, OCORx, S03Rx, P03Rx 2, and CH(COORx)2, where each Rx is H or C1-C4 alkyl or C1-C4 haloalkyl.
37. A pharmaceutical composition comprising a compound of any one of the claims 1-36 admixed with at least one pharmaceutically acceptable carrier or excipient.
38. The pharmaceutical composition of claim 37, which comprises at least one sterile pharmaceutically acceptable carrier or excipient.
39. The pharmaceutical composition of claim 37, which comprises at least two pharmaceutically acceptable carriers and/or excipients.
40. A compound according to any one of claims 1-36 for use in therapy.
41. The compound of claim 40, wherein the use in therapy is a use to treat cancer.
42. The compound of claim 41, wherein the cancer is selected from leukemia, lymphoma, lung cancer, colon cancer, CNS cancer, melanoma, ovarian cancer, renal cancer, prostate cancer, breast cancer, head and neck cancers, and pancreatic cancer.
43. A method to treat cancer, which comprises administering to a subject in need thereof an effective amount of a compound according to any one of claims 1-36.
44. The method of claim 43, wherein the cancer is selected from leukemia, lymphoma, lung cancer, colon cancer, CNS cancer, melanoma, ovarian cancer, renal cancer, prostate cancer, breast cancer, head and neck cancers, and pancreatic cancer.
45. Use of a compound according to any one of claims 1-36 for the manufacture of a medicament.
46. The use of claim 45, wherein the cancer is selected from leukemia, lymphoma, lung cancer, colon cancer, CNS cancer, melanoma, ovarian cancer, renal cancer, prostate cancer, breast cancer, head and neck cancers, and pancreatic cancer.
PCT/US2012/021144 2011-01-12 2012-01-12 Pyrazolopyrimidine derivatives and uses as anticancer agents WO2012097196A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161432164P 2011-01-12 2011-01-12
US61/432,164 2011-01-12

Publications (1)

Publication Number Publication Date
WO2012097196A1 true WO2012097196A1 (en) 2012-07-19

Family

ID=45562445

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/021144 WO2012097196A1 (en) 2011-01-12 2012-01-12 Pyrazolopyrimidine derivatives and uses as anticancer agents

Country Status (1)

Country Link
WO (1) WO2012097196A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103601675A (en) * 2013-10-08 2014-02-26 南京复兴生物科技有限公司 Novel preparation method of 5-aminomethyl nicotinic acid
CN104163786A (en) * 2014-06-10 2014-11-26 上海应用技术学院 Method for preparing 5-methyl-3-bromomethylpyridine hydrobromide
EP3471712A4 (en) * 2016-06-20 2020-02-19 The Regents of The University of Michigan Small molecule inhibitors of aldh and uses thereof
CN113264872A (en) * 2021-05-19 2021-08-17 北京嘉林药业股份有限公司 Preparation method of rupatadine fumarate intermediate 5-methyl-3-hydroxymethylpyridine

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002076986A1 (en) * 2001-03-22 2002-10-03 Abbott Gmbh & Co. Kg Pyrazolopyrimidines as therapeutic agents
US20060111435A1 (en) * 2003-12-29 2006-05-25 President And Fellows Of Harvard College Compositions for treating or preventing obesity and insulin resistance disorders
WO2007008541A2 (en) * 2005-07-08 2007-01-18 Kalypsys, Inc. Cellular cholesterol absorption modifiers
WO2008024759A2 (en) * 2006-08-21 2008-02-28 University Of Maryland, Baltimore Immunomodulatory compounds that target and inhibit the py+3 binding site of tyrosene kinase p56 lck sh2 domain
WO2009086303A2 (en) * 2007-12-21 2009-07-09 University Of Rochester Method for altering the lifespan of eukaryotic organisms
WO2011127192A2 (en) * 2010-04-06 2011-10-13 Brigham Young University Antimetastatic compounds

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002076986A1 (en) * 2001-03-22 2002-10-03 Abbott Gmbh & Co. Kg Pyrazolopyrimidines as therapeutic agents
US20060111435A1 (en) * 2003-12-29 2006-05-25 President And Fellows Of Harvard College Compositions for treating or preventing obesity and insulin resistance disorders
WO2007008541A2 (en) * 2005-07-08 2007-01-18 Kalypsys, Inc. Cellular cholesterol absorption modifiers
WO2008024759A2 (en) * 2006-08-21 2008-02-28 University Of Maryland, Baltimore Immunomodulatory compounds that target and inhibit the py+3 binding site of tyrosene kinase p56 lck sh2 domain
WO2009086303A2 (en) * 2007-12-21 2009-07-09 University Of Rochester Method for altering the lifespan of eukaryotic organisms
WO2011127192A2 (en) * 2010-04-06 2011-10-13 Brigham Young University Antimetastatic compounds

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103601675A (en) * 2013-10-08 2014-02-26 南京复兴生物科技有限公司 Novel preparation method of 5-aminomethyl nicotinic acid
CN103601675B (en) * 2013-10-08 2015-10-28 南京复兴生物科技有限公司 A kind of preparation method of 5-aminomethyl nicotinic acid
CN104163786A (en) * 2014-06-10 2014-11-26 上海应用技术学院 Method for preparing 5-methyl-3-bromomethylpyridine hydrobromide
EP3471712A4 (en) * 2016-06-20 2020-02-19 The Regents of The University of Michigan Small molecule inhibitors of aldh and uses thereof
US11944626B2 (en) 2016-06-20 2024-04-02 The Regents Of The University Of Michigan Small molecule inhibitors of ALDH and uses thereof
CN113264872A (en) * 2021-05-19 2021-08-17 北京嘉林药业股份有限公司 Preparation method of rupatadine fumarate intermediate 5-methyl-3-hydroxymethylpyridine
CN113264872B (en) * 2021-05-19 2023-09-08 北京嘉林药业股份有限公司 Preparation method of rupatadine fumarate intermediate 5-methyl-3-hydroxymethylpyridine

Similar Documents

Publication Publication Date Title
EP2880035B1 (en) Novel pyrrolopyrimidine compounds as inhibitors of protein kinases
KR101297497B1 (en) Protein kinase inhibitors
EP3312177A2 (en) Tricyclic derivative compound, method for preparing same, and pharmaceutical composition comprising same
TWI707855B (en) Novel imidazopyridazine compounds and their use
EP3684772B1 (en) Cyclic iminopyrimidine derivatives as kinase inhibitors
BR112014023460B1 (en) COMPOUND, PHARMACEUTICAL FORMULATION, USE OF A COMPOUND
Kawakita et al. Design and synthesis of pyrrolo [3, 2-d] pyrimidine HER2/EGFR dual inhibitors: Improvement of the physicochemical and pharmacokinetic profiles for potent in vivo anti-tumor efficacy
TWI523856B (en) BCR-ABL kinase inhibitor and its application
EP3590941A1 (en) Urea-substituted aromatic ring-linked dioxane-quinazoline and -linked dioxane-quinoline compounds, preparation method therefor and use thereof
TW200916458A (en) Heterocyclic compounds and methods of use thereof
CA3139285A1 (en) Casein kinase 1e inhibitor, pharmaceutical composition and application thereof
JP6979595B2 (en) Aminothiazole compounds as protein kinase inhibitors
WO2012097196A1 (en) Pyrazolopyrimidine derivatives and uses as anticancer agents
EP3418277B1 (en) Substituted amino six-membered nitric heterocyclic ring compound and preparation and use thereof
BG103370A (en) 2-methoxyphenylpiperazine derivatives
TWI546304B (en) Protein tyrosine kinase inhibitors and their use
AU2017217663B2 (en) Tetrahydroisoquinoline derivatives
CN106467540A (en) Pteridine ketone derivatives are as the application of FLT3 inhibitor
TW202110801A (en) Novel amide compounds and uses thereof
CN112876420B (en) Thiobenzoyl derivative and application thereof
EP1433789A1 (en) Pyrrolopyrazines and their use as selective apoptosis inducers
CA3195511A1 (en) Heteroaromatic compounds and uses thereof
NZ759327B2 (en) Aminothiazole compounds as protein kinase inhibitors
JP2022549678A (en) Heterocyclic derivative and use thereof
CN111978318A (en) Imidazopyridine MNK1/MNK2 kinase inhibitor and preparation method and application thereof

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: 12702347

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12702347

Country of ref document: EP

Kind code of ref document: A1