OA20534A - Pyrazolo[3,4-b]Pyridine compounds as inhibitors of TAM and MET kinases. - Google Patents

Pyrazolo[3,4-b]Pyridine compounds as inhibitors of TAM and MET kinases. Download PDF

Info

Publication number
OA20534A
OA20534A OA1202100068 OA20534A OA 20534 A OA20534 A OA 20534A OA 1202100068 OA1202100068 OA 1202100068 OA 20534 A OA20534 A OA 20534A
Authority
OA
OAPI
Prior art keywords
oxy
amino
pyridin
pyrazolo
fluoro
Prior art date
Application number
OA1202100068
Inventor
Ronald Jay Hinklin
Shelley Allen
Patrick BARBOUR
Adam Cook
Joshua DAHLKE
John Gaudino
Ellen Laird
Oren T. MCNULTY
Qian Zhao
Original Assignee
Array Biopharma 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 Array Biopharma Inc. filed Critical Array Biopharma Inc.
Publication of OA20534A publication Critical patent/OA20534A/en

Links

Abstract

Provided herein are compounds of the Formula I :

Description

PYRAZOLO[3,4-b]PYRIDINE COMPOUNDS AS INHIBITORS OF TAM AND MET KINASES
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims priority to U.S. Provisional Application Serial Nos. 62/724,829, filed on August 30, 2018, and 62/858,686, filed June 7, 2019; the contents of which are hereby incorporated by référencé in their entireties.
BACKGROÜND
[0002] Provided herein are novel inhibitors of TAM and MET kinases, pharmaceutical compositions comprising the compounds, processes for making the compounds, and the use of the compounds in therapy. More particularly, provided herein are pyrazolo[3,4-b]pyridine compounds useful in the treatment and prévention of diseases which can be treated with a TAM kinase inhibitor or MET kinase inhibitor.
[0003] Receptor tyrosine kinases (RTKs) are cell surface proteins that transmit signais from the extracellular environment to the cell cytoplasm and nucléus to regulate cellular events such as survival, growth, prolifération, différentiation, adhesion and migration.
[0004] The TAM subfamily consists of three RTKs including TYRO3, AXL and Mer (Graham étal., 2014, Nature Reviews Cancer 14, 769-785; Lingeret al., 2008, Advances in Cancer Research 100, 35-83). TAM kinases are characterized by an extracellular ligand binding domain consisting of two immunoglobulin-like domains and two fibronectin type III domains. Two ligands, growth arrest spécifie 6 (GAS6) and protein S (PROS1), hâve been identified for TAM kinases. GAS6 can bind to and activate ail three TAM kinases, while PROS1 is a ligand for Mer and TYRO3 (Graham et al., 2014, Nature Reviews Cancer 14, 769-785).
[0005] AXL (also known as UFO, ARK, JTKI1 and TYRO7) was originally identified as a transforming gene from DNA of patients with chronic myelogenous leukemia (O'Bryan et al., 1991, Mol Cell Biol 11, 5016-5031; Graham et al., 2014, Nature Reviews Cancer 14, 769-785; Linger et al., 2008, Advances in Cancer Research 100, 35-83). GAS6 binds to AXL and induces subséquent auto-phosphorylation and activation of AXL tyrosine kinase. AXL activâtes several downstream signaling pathways including PI3K-Akt, Raf-MAPK, PLC-PKC (Feneyrolles et al.,
2014, Molecular Cancer Therapeutics 13, 2141-2148; Linger et al., 2008, Advances in Cancer Research 100, 35-83).
[0006] MER (also known as MERTK, EYK, RYK, RP38, NYK and TYRO 12) was originally identîfied as a phospho-protein from a lymphoblastoid expression library (Graham et 5 al., 1995, Oncogene 10, 2349-2359; Graham et al., 2014, Nature Reviews Cancer 14, 769-785;
Linger et al., 2008, Advances in Cancer Research 100, 35-83). Both GAS6 and PROS1 can bind to Mer and induce the phosphorylation and activation of Mer kinase (Lew et al., 2014). Like AXL, MER activation also conveys downstream signaling pathways including PI3K-Akt and RafMAPK (Linger et al., 2008, Advances in Cancer Research 100, 35-83).
[0007] TYRO3 (also known as DTK, SK Y, RSE, BRT, TIF, ETK2) was originally identifîed through a PCR-based cloning study (Lai et al., Neuron 6, 691-70, 1991; Graham et al., 2014, Nature Reviews Cancer 14, 769-785; Linger et al., 2008, Advances in Cancer Research 100, 35-83). Both ligands, GAS6 and PROS I, can bind to and activate TYRO3. Although the signaling pathways downstream of TYRO3 activation are the least studied among TAM RTKs, it appears that both PI3K-Akt and Raf-MAPK pathways are involved (Linger et al., 2008, Advances in Cancer Research 100, 35-83). AXL, MER and TYRO3 are found to be over-expressed in cancer cells.
[0008] The MET family includes mesenchymal-epithelial transition factor (c-Met), a single pass tyrosine kinase receptor that is expressed on the surface of various épithélial cells; its ligand is hépatocyte growth factor/scatter factor (HGF/SF) (Nakamura et al., Nature 342:440-443, 1989). The binding of HFG to c-Met initiâtes a sériés of intracellular signais that médiate embrogenesis and would healing in normal cells (Organ, Ther. Adv. Med. Oncol. 3(1 Supply):S7-S19, 2011). However, in cancer cells, aberrant HGF/c-Met axis activation, which is closely related to c-Met gene mutations, overexpression, and amplification, promotes tumor development and progression
5 - e.g., by stimulating the PI3K/AKT, Ras/MAPK, JAK/STAT, SRC, and Wnt/p-catenin signal pathways (Zhang et al., Mol. Cancer 17:45, 2018; Mizuno et al., Int. J. Mol. Sci. 14:888-919, 2013). The constitutive activation of the aforementioned c-Met-dependent signaling pathways confers cancer cells with compétitive growth advantage relative to normal cells and increases the likelihood of metastasis - e.g., by enabling access to blood supply and conferring ability to dissociate from tissues (Comoglio et al., Nat. Rev. Drug Dîscov. 7:504-516, 2008; Birchmeier et al, Nat. Rev. Mol. Cell. Biol. 4:915-925, 2003).
[0009] Accordingly, there is a need in the art for compounds and methods of use thereof for the modulation of TAM and MET kinases in treatment of cancer.
SUMMARY OF THE INVENTION
[0010] Provided herein is a compound of the Formula I:
I
[0011] and stéréo isomers, tautomers and pharmaceutically acceptable salts thereof, wherein R1, R2, R9, X1 and G are as defined herein.
[0012] Also provided herein is a phannaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable sait thereof, in admixture with a pharmaceutically acceptable diluent or carrier.
[0013] Also provided herein is a method of inhibiting cell prolifération, in vitro or in vivo, the method comprising contacting a cell with an effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof, or a phannaceutical composition thereof as defined herein.
[0014] Also provided herein is a method of treating cancer and/or inhibiting metastasis associated with a particular cancer in a patient in need of such treatment, the method comprising administering to the patient a therapeutically effective amount of a compound of Formula 1 or a pharmaceutically acceptable sait thereof or a phannaceutical composition thereof as defined herein.
[0015] Also provided herein is a compound of Formula I or a pharmaceutically acceptable sait thereof, or a phannaceutical composition thereof as defined herein for use in therapy.
[0016] Also provided herein is a compound of Formula I or a phannaceutically acceptable sait thereof or a pharmaceutical composition thereof as defined herein for use in the treatment of cancer and/or inhibiting metastasis associated with a particular cancer.
[0017] Also provided herein is a compound of Formula l or a pharmaceutically acceptable sait thereof for use in the inhibition of TAM kinase activity.
[0018] Also provided herein is a compound of Formula I or a pharmaceutically acceptable sait thereof or a pharmaceutical composition thereof as defined herein, for use in the treatment of a TAM-associated disease or disorder such as cancer. In some embodiments, the TAM-associated cancer is a cancer having a chromosomal translocation that results in the expression of a TMEM87B-MERTK fusion protein (e.g., amino acids 1-55 of TMEM87B and amino acids 4331000 of MERTK) or a AXL-MBIP fusion protein.
[0019] Also provided herein is the use of a compound of Formula I or a pharmaceutically acceptable sait thereof, as defined herein in the manufacture of a médicament for the treatment of cancer and/or inhibiting metastasis associated with a particular cancer.
[0020] Also provided herein is a use of a compound of Formula I or a pharmaceutically acceptable sait thereof, as defined herein in the manufacture of a médicament for the inhibition of TAM kinase activity.
[0021] Also provided herein is the use of a compound of Formula I or a pharmaceutically acceptable sait thereof, as defined herein, in the manufacture of a médicament for the treatment of a TAM-associated disease or disorder such as cancer.
[0022] Also provided herein is a pharmaceutical combination which comprises (a) a compound of Formula I or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent. Also provided herein is a pharmaceutical combination which comprises (a) a compound of Formula I or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy. In one embodiment, the compound of Formula I or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Formula 1 or a pharmaceutically acceptable sait thereof and of the additional therapeutic agent are together therapeutically effective. Also provided herein is a pharmaceutical composition comprising such a combination. Also provided herein is a commercial package or product comprising such a combination as a combined préparation for simultaneous, separate or sequential use.
[0023| In one embodiment, the additional therapeutic agent is an anticancer agent (e.g,, any of the additional anticancer agents described herein). Accordingly, provided herein is a pharmaceutical combination for treating cancer (e.g., a TAM-associated cancer) in a patient in need thereof, which comprises (a) a compound of Formula 1 or a pharmaceutically acceptable sait thereof, and (b) an additional anticancer agent (e.g., any of the additional anticancer agents described herein), wherein the compound of Formula I or the pharmaceutically acceptable sait thereof and the additional therapeutic are formulated as separate compositions or dosages for simultaneous, separate or sequential use for the treatment of cancer, wherein the amounts of the compound of Formula 1 or a pharmaceutically acceptable sait thereof and of the additional anticancer agent are together effective in treating the cancer.
[0024J Also provided herein is a pharmaceutical combination for treating cancer (e.g., a TAM-associated cancer) in a patient in need thereof, which comprises (a) a compound of Formula I or a pharmaceutically acceptable sait thereof, and (b) an additional anticancer agent (e.g., any of the additional anticancer agents described herein), wherein the compound of Formula I or the pharmaceutically acceptable sait thereof and the additional therapeutic are formulated as separate compositions or dosages for simultaneous, separate or sequential use for the treatment of cancer, wherein the amounts of the compound of Formula I or a pharmaceutically acceptable sait thereof and of the additional anticancer agent are together effective in treating the cancer. Also provided herein is a pharmaceutical composition comprising such a combination. Also provided herein is the use of such a combination for the préparation of a médicament for the treatment of cancer. Also provided herein is a commercial package or product comprising such a combination as a combined préparation for simultaneous, separate or sequential use; and to a method of treatment of cancer a patient in need thereof.
[0025J Also provided are methods of treating an individual with cancer that include administerîng a compound of Formula I or a pharmaceutically acceptable sait thereof, before, during, or after administration of another anticancer agent (e.g., another anticancer agent to which the subject has previously developed résistance, e.g., any of the additional anticancer agents described herein).
[0026] Also provided herein are methods of treating a patient identified or diagnosed as having a TAM-associated cancer that include administerîng to a patient identified or diagnosed as having a TAM-associated cancer a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof, or pharmaceutical composition including a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof.
[0027] Also provided herein are methods of treating a patient having a cancer that include (a) identifying the patient as having a TAM-associated cancer, and (b) administering to the patient identifîed as having a TAM-associated cancer a therapeutically effective amount of a compound of Formula 1 or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition including a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof.
[0028] Also provided herein are methods of decreasing the risk of developing a metastasis or an additional metastasis in a patient identifîed or diagnosed as having a TAM-associated cancer that include administering to a patient identifîed or diagnosed as having a TAM-associated cancer a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition including a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof.
[0029] Also provided herein are methods of decreasing the risk of developing a metastasis or an additional metastasis in a patient having a cancer that include; (a) identifying a patient having a TAM-associated cancer, and (b) administering to the identifîed as having a TAM-associated cancer a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition including a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof.
[0030] Also provided herein are methods of decreasing migration and/or invasion of a cancer cell in a patient identifîed or diagnosed as having a TAM-associated cancer that include administering to a patient identifîed or diagnosed as having a TAM-associated cancer a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition including a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof.
[0031] Also provided herein are methods of decreasing migration and/or invasion of a cancer cell in a patient having a cancer that include (a) identifying the patient as having a TAMassociated cancer; and (b) administering to the patient identifîed as having a TAM-associated cancer a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition including a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof.
[0032] Also provided herein are methods of selecting a treatment for a patient identified or diagnosed as having a TAM-associated cancer that include selecting a compound of Formula I or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition including a therapeutically effective amount of a compound of Formula 1 or a pharmaceutically acceptable sait thereof, for a patient identified or diagnosed as having a TAM-associated cancer.
[0033] Also provided herein are methods of selecting a treatment for a patient that include (a) identifying the patient as having a TAM-associated cancer, and (b) selecting a compound of Formula I or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition including a therapeutically effective amount of a compound of Formula 1 or a pharmaceutically acceptable sait thereof, for the patient identified as having a TAM-associated cancer.
[0034) Also provided herein are methods of selecting a treatment for a patient identified or diagnosed as having a cancer that include (a) administering an additionaî anticancer agent to the patient, (b) after (a), detecting increased expression and/or activity of a TAM kinase in a cancer cell from the patient, and (c) after (b), selecting a compound of Formula 1 or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition including a therapeutically effective amount of a compound of Formula 1 or a pharmaceutically acceptable sait thereof, for the patient. [0035] Also provided herein are methods of treating a patient identified or diagnosed as having a cancer that include: (a) administering to the patient identified or diagnosed as having a cancer one or more doses of at least one additionaî anticancer agent; (b) after (a), detecting an increase in the expression and/or activity of a TAM kinase in a cancer cell or an immune cell from the subject; and (c) after (b), administering to the patient a compound of Formula I or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition including a therapeutically effective amount of a compound of Formula 1 or a pharmaceutically acceptable sait thereof. In some embodiments of these methods, step (c) further includes administering to the patent the at least one additionaî anticancer agent.
[0036] Also provided are methods of treating a patient identified or diagnosed as having a cancer that include: (a) detecting an increase in the expression and/or activity of a TAM kinase in a cancer cell or an immune cell from a patient identified or diagnosed as having a cancer and previously administered one or more doses of at least one additional anticancer agent; and (b) after (a), administering to the patient a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition including a therapeutically effective amount of a compound of Formula 1 or a pharmaceutically acceptable sait thereof. In some embodiments of these methods, step (b) further includes administering to the patient the at least one additional anticancer agent.
[0037] Also provided herein are methods of treating a patient identified or diagnosed as havîng a cancer that has been previously administered one or more doses of at least one additional anticancer agent and has been identified as having a cancer cell or an immune cell that has increased expression and/or activity of a TAM kinase that include administering a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition including a therapeutically effective amount of a compound of Formula 1 or a pharmaceutically acceptable sait thereof, to the patient. In some embodiments of these methods, step (b) further includes administering to the patient the at least one additional anticancer agent.
[0038] Also provided herein are methods of treating a patient identified or diagnosed as having a cancer that include: (a) selecting a patient identified or diagnosed as having increased expression and/or activity of a TAM kinase in a cancer cell or an immune cell; and (b) after (a) administering to the selected patient a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition including a therapeutically effective amount of a compound of Formula 1 or a pharmaceutically acceptable sait thereof. In some embodiments of these methods, step (b) further includes administering to the patient at least one additional anticancer agent.
[0039] Also provided herein are methods of treating a patient identified or diagnosed as having a cancer that include: (a) selecting a patient identified or diagnosed as having a cancer that has been previously administered one or more doses of an additional anticancer agent and identified as having a cancer cell or an immune cell having increased expression and/or activity of a TAM kinase; and (b) after (a), administering to the selected patient a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition including a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof. In some embodiments of these methods, step (b) further includes administering to the patient at least one additional anticancer agent.
[0040] Also provided herein are methods of treating a patient identified or diagnosed as having a TAM-associated cancer that include; (a) administering to the patient identified or diagnosed as having a TAM-associated cancer one or more doses of a TAM kinase inhibitor; (b) after (a), detecting résistance of the TAM-associated cancer in the patient to the TAM kinase inhibitor; and (c) after (b), administering to the patient a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition including a therapeutically effective amount of a compound of Formula 1 or a pharmaceutically acceptable sait thereof. In some embodiments of these methods, step (c) further includes administering to the patient at least one additional anticancer agent.
[0041] Also provided herein are methods of treating a patient identified or diagnosed as having a TAM-associated cancer that include; (a) detecting résistance of the TAM-associated cancer in the patient to a TAM kinase inhibitor that was previously administered to the patient; and (b) after (a), administering to the patient a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition including a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof. In some embodiments of these methods, step (b) further includes administering to the patient at least one additional anticancer agent.
|0Û42] Also provided herein are methods of treating a patient identified or diagnosed as having a TAM-associated cancer and determined to hâve previously developed résistance to a TAM kinase inhibitor that include administering to the patient a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition including a therapeutically effective amount of a compound of Formula 1 or a pharmaceutically acceptable sait thereof. Some embodiments of these methods further include administering to the patient at least one additional anticancer agent.
[0043] Also provided herein are methods of decreasing immune tolérance in a subject in need thereof that include administering to the subject a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition thereof as defined herein.
[0044] Also provided herein are methods of inhibiting angiogenesis in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of
a compound of Formula I or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition thereof as defined herein,
[0045] Also provided herein are methods of suppressing résistance to a therapeutic agent 5 in a subject in need thereof that include administering to the subject a therapeutically effective amount of (i) a compound of Formula I or a pharmaceutically acceptable sali thereof, or any of the pharmaceutical compositions thereof described herein, and (ii) the therapeutic agent, where the therapeutic agent is selected from the group consîsting of a chemotherapeutic agent, a PI-3 kinase inhibitor, an EGFR inhibitor, a HER2/neu inhibitor, an FGFR inhibitor, an ALK inhibitor, an 10 IGF1R inhibitor, a VEGFR inhibitor, a PDGFR inhibitor, a glucocorticoid, a BRAF inhibitor, a
MEK inhibitor, a HER4 inhibitor, a MET inhibitor (e.g., a Type 1 c-Met inhibitor), a RAF inhibitor, an Akt inhibitor, a FTL-3 inhibitor, and a MAP kinase pathway inhibitor.
[0046] Also provided herein are methods of treating a patient identified or diagnosed as having a TAM-associated cancer that include administering radiation therapy before or after 15 administering to the patient a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition including a therapeutically effective amount of a compound of Formula 1 or a pharmaceutically acceptable sait thereof.
[0047) Also provided herein are methods of treating a patient identified or diagnosed as 20 having a TAM-associated cancer that include administering surgery before or after administering to the patient a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition including a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof.
[0048] Also provided herein are methods for inhibiting a TAM kinase activity in a 2 5 mammalian cell in need thereof that include contacting the mammalian cell with a compound of
Formula 1 or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition including a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof.
[0049] Also provided herein is a process for preparing a compound of Formula I or a 30 pharmaceutically acceptable sait thereof.
[0050] Also provided herein is a compound of Formula I or a pharmaceutically acceptable
II sait thereof obtained by a process of preparing the compound as defïned herein.
[0051] Unless otherwise defïned, ail technical and scientific terms used herein hâve the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Methods and materials are described herein for use in the présent invention; other, suitable methods and materials known in the art can also be used. The materials, methods, and examples are illustrative only and not intended to be limiting, AU publications, patent applications, patents, sequences, database entries, and other référencés mentioned herein are incorporated by reference in their entirety. In case of conflict, the présent spécification, including définitions, will control.
Other features and advantages of the invention will be apparent from the following detailed description and figures, and from the daims.
DETAILED DESCRIPTION OF THE INVENTION
[0052] Provided herein is a compound of the Formula I:
[0053| and stéréo isomers, tautomers and pharmaceutically acceptable salts thereof, wherein:
[0054] X'isCHorN;
[0055] R1 ts hydrogen or C1-C6 alkyl;
[0056] R2 is
[0057] (a) hydrogen,
[0058] (b) Cl-C6 alkyl,
[0059] (c) hydroxyC 1-C6 alkyl,
[0060] (d) dihydroxyC2-C6 alkyl, |0061] (e) C1-C6 fluoroalkyl optionally substituted with OH,
[0062] (f) (di-C I-C6 alkoxy)C2-C6 alkyl-,
[0063] (g) (C1-C6 alkoxy)Cl-C6 alkyl- wherein said alkyl portion is optionally substituted with OH,
[0064] (h) Cyc1, {0065] (i) Cyc2,
[0066] (j) (hetCyc’)C 1-C6 alkyl- wherein said alkyl portion is optionally substituted with
OH,
[0067] (k) (Ar')Cl -C6 alkyl- wherein said alkyl portion is optionally substituted with OH,
[0068] (1) (hetAr')Cl-C6 alkyl- wherein said alkyl portion is optionally substituted with
OH, or
[0069] (m) (HOSO3)C1-C6 alkyl-;
[0070] Cyc1 is a 3-4 membered cycloalkyl ring optionally substituted with 1-2 substituents independently selected from halogen, hydroxy, C1-C3 alkyl, hydroxyCl-C3 alkyl, C1-C3 alkoxy, (C1-C3 alkoxy)Cl-C3 alkyl- and R'RNC(=O)-;
[0071] R' and R are independently hydrogen or C1-C6 alkyl;
[0072] Cyc2 is a 5-membered cycloalkyl ring substituted with 1-2 substituents independently selected from C1-C3 alkyl, (C1-C3 a!koxy)Cl-C3 alkyl- and hydroxyCl-C3 alkyl-; [0073] hetCyc1 is a 5-6 membered saturated heterocyclic ring having 1-2 ring heteroatoms independently selected from O, N and SO2, wherein said ring is optionally substituted with oxo; [0074] Ar1 is phenyl;
[0075] hetAr1 is pyridyl;
|0076] G is
[0077] X2isCorN;
[0078] Ring A, including the atoms at the points of attachment, is a 5-6 membered heterocyclic ring optionally having an additional 1 -2 ring nitrogen atoms when X2 is N and having one ring nitrogen atom when X2 is C;
[0079] R2 is hydrogen, methyl or absent;
[0080] Ring B, including the atoms at the points of attachment, is a 6-membered saturated carbocyclic optionally substîtuted with oxo or a 6-membered aromatic carbocyclic ring optionally substituted with OH;
[0081] R6 is hydrogen, halogen, C1-C6 alkyl, C1-C6 alkoxy, hydroxyCl-C6 alkyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)Cl-C6 alkyi- or hetCyc2, provîded that when R6 is on the ring carbon atom adjacent to the carbon linked to the —NHC(=O)- moiety of Formula I, then R6 is not halogen. and
[0082] R7 is hydrogen, C1-C6 alkyl, oxo or thioxo,
[0083] or optionally when R6 and R7 are on the same carbon atom, R6 and R7 together with the carbon atom to which they are attached form a cyclopropyl ring;
[0084] hetCyc2 is a 4-6 membered saturated heterocyclic ring having a ring nitrogen atom and optionally substîtuted with C1-C6 alkyl;
[0085] Rs is Ar2, hetAr2, C3-C6 cycloalkyl, hetCycJ or C1-C6 alkyl;
[0086] Ar2 is phenyl optionally substituted with one or more substituents independentiy selected frorn halogen, C1-C2 alkyl and C1-C2 alkoxy;
[0087] hetAr2 is a 5-6 membered heteroary! having 1-2 ring nitrogen atoms and optionally substituted with one or more substituents independentiy selected from halogen, C1-C2 alkyl and C1-C2 alkoxy;
[0088] hetCyc3 is a 5-6 membered heterocyclic ring having a ring oxygen atom; and [0089] R9 is hydrogen or halogen.
|0090] For complex Chemical names employed herein, the substituent group is named before the group to which it attaches. For example, methoxyethyl comprises an ethyl backbone with a methoxy substituent.
]0091 ] The term heterocyclic ring when specifically referring to Ring A means that Ring A is a saturated, partially unsaturated or aromatic 5-6 membered heterocyclic ring.
[0092] The term halogen means -F (sometimes referred to herein as fluoro or fluoros), -Cl,-Br and -I.
[0093] The terms C 1-C2 alkyl, C 1-C3 alkyl, C 1-C6 alkyl and C2-C6 alkyl as used herein refer to saturated linear or branched-chain monovalent hydrocarbon radicals of one to two, one to three, one to six or two to six carbon atoms, respectively. Examples include, but are not limited to, methyl, ethyl, 1-propyl, isopropyl, 1-butyl, isobutyl, sec-butyl, tert-butyl, 2-methyl-2propyl, pentyl, neopentyl, and hexyl.
[0094] The tenus C1-C2 alkoxy, C1-C3 alkoxy and C1-C6 alkoxy as used herein refers to a saturated linear or branched-chain monovalent alkoxy radical of one to two, one to three, or one to six carbon atoms, respectively, wherein the radical is on the oxygen atom. Examples include methoxy, ethoxy, propoxy, isopropoxy, butoxy and tert-butoxy.
[0095] The tenu C1-C6 fluoroalkyl as used herein include C1-C6 alkyl and C1-C6 alkoxy groups, respectively, that are substituted with one or more fluorines, such as, but not limited to, fluoromethyl, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, and the like.
[0096] The terni (C1-C6 alkoxy)Cl-C6 alkyl- as used herein refers to saturated linear or branched-chain monovalent radicals of one to six carbon atoms, wherein one ofthe carbon atoms is substituted with a C1-C6 alkoxy group as defined herein. Examples include methoxymethyl (CH3OCH2-) and methoxyethyl (CH3OCH2CH2-).
[0097] The terni (C1-C3 alkoxy)Cl-C3 alkyl- as used herein refers to saturated linear or branched-chain monovalent radicals of one to three carbon atoms, wherein one of the carbon atoms is substituted with a C1-C3 alkoxy group as defined herein.
[0098] The tenu (di-Cl -C6 alkoxy)C2-C6 alkyl- as used herein refers to saturated linear or branched-chain monovalent radicals of two to six carbon atoms, wherein two of the carbon atoms are substituted with a C1-C6 alkoxy group as defined herein.
[0099] The ternis hydroxyCl-C3 alkyl- and hydroxyCl-C6 alkyl- as used herein refers to a saturated linear or branched-chain monovalent alkyl radicals of one to six carbon atoms, respectively, wherein one of the carbon atoms is substituted with a hydroxyl group.
[00100] The terni dihydroxyC2-C6 alkyl- as used herein refers to a saturated linear or branched-chain monovalent alkyl radical of two to six carbon atoms, wherein two of the carbon atoms are substituted with a hydroxyl group, provided two hydroxyl groups are not on the same carbon atom.
[00101] The term (hetCyc'jCl-Cô alkyl- as used herein refers to a saturated linear or branched-chain monovalent alkyl radicals of one to six carbon atoms, respectively, wherein one of the carbon atoms is substituted with a hetCyc1 group as defined herein.
[00102] The terni (Ar'jCl-Cô alkyl- as used herein refers to a saturated linear or branched-chain monovalent alkyl radicals of one to six carbon atoms, respectively, wherein one of the carbon atoms is substîtuted with a Ar1 group as defined herein.
[00103] The term (hetAr'jCl-Cô alkyl- as used herein refers to a saturated linear or branched-chain monovalent alkyl radicals of one to six carbon atoms, respectively, wherein one of the carbon atoms is substîtuted with a hetAr1 group as defined herein.
[00104] The term (HOSChjCl-Cô alkyl- as used herein refers to a saturated linear or branched-chain monovalent alkyl radicals of one to six carbon atoms, respectively, wherein one of the carbon atoms is substîtuted with a HOSO3- group.
[00105] The term (C3-C6 cycloalkyl)Cl-C6 alkyl- as used herein refers to a saturated linear or branched-chain monovalent alkyl radicals of one to six carbon atoms, respectively, wherein one of the carbon atoms is substîtuted with a C3-C6 cycloalkyl group.
[00106] The term C3-C6 cycloalkyl as used herein refers to a saturated carbocyclic ring having three to six ring carbon atoms, that is, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyL [00107] The term oxo or oxo group as used herein means an oxygen atom that is double bonded to a carbon atom, i.e., =O.
[00108] The term thioxo as used herein means a sulfur atom that is double bonded to a carbon atom, i.e., =S.
[00109] The term compound as used herein is meant to include ail stéréo isomers, géométrie isomers, tautomers, and isotopes of the structures depicted. Compounds herein identifïed by name or structure as one particular tautomeric fonn are intended to include other tautomeric forms unless otherwise specified,
[00110] The term tautomer as used herein refers to compounds whose structures differ markedly in arrangement of atoms, but which exist in easy and rapid equilibrium, and it is to be understood that compounds provided herein may be depicted as different tautomers, and when compounds hâve tautomeric forms, ail tautomeric forms are intended to be within the scope of the invention, and the naming of the compounds dues not exclude any tautomer. Exemplary tautomerizations include, but are not iimited to, amide-to-imide; enamine-to-imine; enamine-to-(a different) enamine tautomerizations; and keto-to-enol.
[00111] It will be appreciated that certain compounds provided herein may contain one or more centers of asymmetry and may therefore be prepared and isolated in a mixture of isomers
[6 such as a racemîc mixture, or in an enantiomerically pure form. For compounds of the invention wherein stereochemistry is designated by straight thick bars or straight dashed bars, the straight thick bars or straight dashed bars indicate relative stereochemistry. For compounds of the invention wherein stereochemistry is designated by solid wedges or dashed wedges, the solid wedges or dashed wedges indicate absolute stereochemistry.
[00112| In one embodiment of Formula I, X is CH.
[00113] In one embodiment of Formula 1, X1 is N.
[00114] In one embodiment of Formula I, R9 is hydrogen.
[00115] In one embodiment of Formula I, R9 is halogen.
[00116J In one embodiment of Formula I, X1 is CH and R9 is halogen. In one embodiment
of Formula 1, X1 is CH and R9 is fluoro.
[00117] In one embodiment of Formula I, X1 is N and R9 is hydrogen.
[00118] in one embodiment of Formula 1, R1 is hydrogen.
[00119| In one embodiment of Formula I, R1 is C1-C6 alkyl. In one embodiment of Formula I, R1 is methyL
[00120] In one embodiment of Formula I, R2 is hydrogen. In one embodiment of Formula I, R2 is hydrogen and R! is hydrogen.
[00121] In one embodiment of Formula I, R2 is C1-C6 alkyl. In one embodiment of Formula I, R2 is C1-C6 alkyl and R1 is hydrogen. In one embodiment of Formula I, R2 is tnethyl or ethyl. In one embodiment of Formula I, R2 is methyl or ethyl and R1 is hydrogen.
[00122] In one embodiment of Formula I, R2 is hydroxyCl-C6 alkyl. In one embodiment of Formula I, R2 is hydroxyCl-C6 alkyl and R1 is hydrogen. In one embodiment of Formula I, R2 is hydroxyCl-C6 alkyl and R1 is CI-C6 alkyl. In one embodiment of Formula I, R2 is hydroxyClC6 alkyl and R1 is methyl. In one embodiment of Formula I, R2 is selected from the structures:
and R1 is hydrogen or C1-C6 alkyl. In one of said embodiments, R1 is hydrogen. In one of said 5 embodiments, R1 is C1-C6 alkyl. In one of said embodiments, R1 is methyi.
[00123] In one embodiment of Formula I, R2 is dihydroxyC2-C6 alkyl. In one embodiment of Formula I, R2 is dihydroxyC2-C6 alkyl and R1 is hydrogen. In one embodiment of Formula I, R2 is selected from the structures:
and R1 is hydrogen or C1-C6 alkyl. In one of said embodiments, R1 is hydrogen.
[00124] In one embodiment of Formula I, R2 is CI-C6 fluoroalkyl optionally substituted 15 with OF1. In one embodiment of Formula I, R2 is C1-C6 fluoroalkyl optionally substituted with OH and R1 is hydrogen. In one embodiment of Formula I, R2 is CJ-C6 fluoroalkyl optionally substituted with OH and R1 ts C1-C6 alkyl. In one embodiment of Formula I, R2 is selected from the structures:
and R1 is hydrogen orCl-C6 alkyl. In one ofsaid embodiments, R1 is hydrogen.
[00125] In one embodiment of Formula I, R2 is (di-Cl-C6 alkoxy)C2-C6 alkyl-. In one embodiment of Formula I, R2 is (di-Cl-C6 alkoxy)C2-C6 alkyl- and R1 is hydrogen. In one embodiment of Formula 1, R2 is (di-CI-C6 alkoxy)C2-C6 alkyl- and R1 is C1-C6 alkyl. In one embodiment, R2 is
and R1 îs hydrogen or C1-C6 alkyl. In one of said embodiments, R1 is hydrogen.
[00126] In one embodiment of Formula 1, R2 is (C1-C6 alkoxy)Cl-C6 alkyl- wherein said alkyl portion is optionally substituted with OH. In one embodiment of Formula I, R2 is (C1-C6 10 alkoxy)Cl-C6 alkyl- wherein said alkyl portion is optionally substituted with OH and R1 is hydrogen. In one embodiment of Formula I, R2 is (CI-C6alkoxy)Cl-C6 alkyl- wherein said alkyl portion is optionally substituted with OH and R2 is C1-C6 alkyl. In one embodiment, R2 is selected from the structures:
HO
and R1 is hydrogen or C1-C6 alkyl. In one of said embodiments, R1 is hydrogen. In one of said embodiments, R1 is CI-C6 alkyl. In one of said embodiments, R1 is methyl.
[00127] In one embodiment of Formula I, R2 is Cyc1. In one embodiment of Formula I, R2 is Cyc1 and R1 is hydrogen. In one embodiment of Formula I, R2 is Cyc’ and R1 is C1-C6 alkyl. In one embodiment of Formula I, R2 is selected from the structures:
and R1 is hydrogen or C1-C6 alkyl. In one of said embodiments, R1 is hydrogen.
[00128] In one embodiment of Formula I, R2 is Cyc2. In one embodiment of Formula I, R2 is Cyc2 and R1 is hydrogen. In one embodiment of Formula 1, R2 is Cyc2 and R1 is C1-C6 alkyl. 15 In one embodiment of Formula I, R2 is selected from the structures:
0 and R1 is hydrogen or C1-C6 alkyl. In one of said embodiments, R1 is hydrogen.
[00129] In one embodiment of Formula I, R2 is (hetCyc’)Ct-C6 alkyl- wherein said alkyl portion is optionally substituted with OH. In one embodiment of Formula I, R2 is (hetCyc’)Cl20534
C6 alkyl- wherein said alkyl portion is optionally substituted with OH and R1 is hydrogen. In one embodiment of Formula I, R2 is (hetCycl)Cl-C6 alkyl- wherein said alkyl portion is optionally substituted with OH and R1 is C1-C6 alkyl. In one embedment, R2 is selected from the structures
ηολ,
O-Z and R1 is hydrogen or CI-C6 alkyl. In one of said embodiments, R1 is hydrogen.
[00130| In one embodiment of Formula I, R2 is (Ar’)C I-C6 alkyl- wherein said alkyl portion is optionally substituted with OH. In one embodiment of Formula I, R2 is (Ar')Cl-C6 alkylwherein said alkyl portion is optionally substituted with OH and R1 is hydrogen. In one embodiment of Formula I, R2 is (Ar'jCl-Cô alkyl- wherein said alkyl portion is optionally substituted with OH and R2 is C1-C6 alkyl. In one of said embodiments, R2 is (Ar')Cl-C6 alkylwherein said alkyl portion is substituted with OH. In one embodiment, R2 is
HO~^
O and R1 is hydrogen or C1-C6 alkyl. In one of saîd embodiments, R1 îs hydrogen.
[00131[ In one embodiment of Formula I, R2 is (hetAr'jCl-Cô alkyl- wherein said alkyl portion is optionally substituted with OH. In one embodiment of Formula I, R2 is (hetAr’)Cl-C6 alkyl- wherein said alkyl portion is optionally substituted with OH and R1 is hydrogen. In one embodiment of Formula I, R2 is (hetAr')Cl-C6 alkyl- wherein said alkyl portion is optionally substituted with OH and R1 is C1-C6 alkyl. In one of said embodiments, R2 is (hetAr'jCl-Cé alkyl- wherein said alkyl portion is substituted with OH. In one embodiment, R2 is selected from the structures:
and R1 is hydrogen or C1-C6 alkyl. En one of said embodiments, R1 is hydrogen.
|00132| In one embodiment of Formula I, R2 is (HOSO3)C1-C6 alkyl-. In one embodiment of Formula I, R2 is (HOSOs)Cl-C6 alkyl- and R1 is hydrogen. In one embodiment of Formula I, R2 is (H0S0s)CI-C6 alkyl- and R1 is C1-C6 alkyl. In one embodiment, R2 is
and R1 is hydrogen or C1-C6 alkyl. In one of said embodiments, R1 is hydrogen.
[00133] In one embodiment of Formula I, G is
R6 R7 r3
K A % rB
O wherein X2 is C or N, and Ring A is a 5-6 membered heterocyclic ring optionally having an additionaî 1 -2 ring nitrogen atoms when X2 is N and having one ring nitrogen atom when X2 is C, and R3, R6, R7 and R8 are as defined for Formula I.
[00134] In one embodiment of Formula I, G is R.6 R7 r3
y. A ?x3 rE o
wherein X2 is C or N, Ring A is a 5-6 membered heterocyclic ring having one ring nitrogen atom, and R3, R6, R7 and R8 are as defined for Formula I.
[00135] In one embodiment of Formula I, G is
Ο wherein X2 is N, R3 is absent. Ring A is a 6-membered heterocyclic ring having one additional ring nitrogen atom, and R7 is oxo, wherein G has the formula A-l
A-l wherein R6 is C1-C6 alkyl, hydroxyCl-C6 alkyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)C 1-C6 alkyl- or hetCyc2, and hetCyc2 and Rs are as defined for Formula I. In one embodiment of formula A-l, R8 is Ar2, wherein Ar2 is pheny! optionally substîtuted with one or more substituents independently selected from halogen, Cl -C2 alkyl and C1-C2 alkoxy. In one embodiment, Ar is pheny 1 optionally substîtuted with one or more halogens. In one embodiment G is formula A-l wherein formula A-l is seiected from the structures:
[00136] In one embodiment of Formula I, G is
wherein X2 is N, R3 is absent, Ring A is a 6-membered heterocyclic ring having an additional ring nitrogen atom, and R7 is hydrogen, wherein G has the formula A-2
RÊ
o
A-2 wherein R6 is hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl, R7 is hydrogen, and R8 is as defined for Formula I. In one embodiment of formula A-2, R8 is Ar2, wherein Ar2 is phenyl optionally 10 substituted with one or more substituents independently selected from halogen, C1-C2 alkyl and
C1-C2 alkoxy. In one embodiment, Ar2 is phenyl optionally substituted with one or more halogens. In one embodiment, Ring A is formula A-2 and is selected from the structures:
[00137] In one embodiment of Formula I, G is
wherein X2 is N, R3 is absent, Ring A is a 6-membered heterocyclic ring, wherein G has the formula A-3
R6 R7
A-3 wherein R6 is halogen, C1-C6 alky 1, C1-C6 alkoxy, or C3-C6 cycloalkyl, R7 is hydrogen, provided that when R6 is on the ring carbon atom adjacent to the carbon linked to the -NHC(=O)- moiety 10 of Formula I, then R6 is not halogen, and R8 is as defined for Formula 1. As used herein, the ring carbon atom adjacent to the carbon linked to the -NHC(=O)- moiety of Formula I refers to the carbon identified by the asterîsk in the following structure:
[00138]
In one embodiment of formula A-3, R8 ÎS Ar2, wherein Ar2 is phenyl optionally substituted with one or more substituents independently selected from halogen, CÎ-C2 alkyl and C1-C2 alkoxy. In one embodiment, Ar2 is phenyl optionally substituted with one or more halogens. In one embodiment, G is formula A-3 and is selected from the structures:
wherein X2 is N, R3 is absent, Ring A is a 6-inembered heterocyclic ring having two additional ring nitrogen atoms, and R7 is oxo or thiooxo, wherein G has the formula A-4
A-4 wherein Y is O or S, R6 is C1-C6 alkyl, and R8 is as defined for Formula 1. in one embodiment of formula A-4, R8 is Ar2, wherein Ar2 is phenyi optionally substituted with one or more substituents independently selected from halogen, C1-C2 alkyl and C1-C2 alkoxy. In one embodiment, Ar2 is phenyi optionally substituted with one or more halogens. In one embodiment, 10 G is formula A-4 and is selected from the structures:
[00140]
In one embodiment of Formula I, G is
O
wherein X2 is C, R3 is absent, Ring A is a 6-membered heterocyclic ring having a ring nitrogen atom, wherein G has the formula A-5
R6
I
o
A-5 wherein R6 is C1-C6 alkyl, R7 is hydrogen, and R8 is as defined for Formula 1. In one embodiment of formula A-5, R8 is Ar2, wherein Ar2 is phenyl optionally substituted with substituents independently selected from halogen, CI-C2 alkyl and C1-C2 alkoxy. In one embodiment, Ar2 is phenyl optionally substituted with one or more halogens. In one embodiment, G is formula A-5 and is selected from the structures:
[00141]
In one embodiment of Formula I, G is
Ο wherein X2 is N, R3 is absent, Ring A is a 6-membered heterocyclic ring having two additional ring nitrogen atoms, wherein G has the formula A-6
wherein R6 and R7 are hydrogen and R8 is as defïned for Formula J. In one embodiment of formula A-6, R8 is Ar2, wherein Ar2 is phenyl optionally substituted with substituents independently selected from halogen, C1-C2 alkyl and CI-C2 alkoxy. In one embodiment, Ar2 is phenyl optionally substituted with one or more halogens. In one embodiment, G is formula A-6 and has the structure:
[00142] In one embodiment of Formula I, G is
O wherein X2 is N, R3 is hydrogen or methyl, and Ring A is a 6-membered heterocyclic ring having a ring nitrogen atom, wherein G has the formula A-7
R® R7
O
A-7 wherein R6 and R7 are hydrogen, or R6 and R7 are on the same carbon atom and R6 and R7 together 5 with the carbon atom to which they are attached form a cyclopropyl ring, and R8 is as defïned for
Formula 1. In one embodiment of formula A-7, R8 is Ar2, wherein Ar2 is phenyl optionally substituted with substituents independently selected from halogen, C1-C2 alkyl and C1-C2 alkoxy. In one embodiment, Ar2 is phenyl optionally substituted with one or more halogens. In one embodiment, G is formula A-7 and is selected from the structures:
[00143] In one embodiment of Formula I, G is
wherein X2 is N, R3 is absent, Ring A is a 5-membered heterocyclic ring having an additional ring nitrogen atom, wherein G has the formula A-8
A-8 wherein R6 is C1-C6 alkyl, R7 is hydrogen or CI-C6 alkyl, and Rs is as defïned for Formula I. In one embodiment of formula A-8, R8 is Ar2, wherein Ar2 is phenyl optionally substituted with substituents independently selected from halogen, C1-C2 alkyl and C1-C2 alkoxy. In one embodiment, Ar2 is phenyl optionally substituted with one or more halogens. In one embodiment, G is formula A-8 and is selected from the structures:
[00144]
In one embodiment of Formula I, G is
wherein Ring B and R8 are as defïned for Formula I.
[00145] In one embodiment of Formula I, G is
O wherein Ring B is a 6-membered saturated carbocyclic optionally substituted with oxo. In one embodiment, G has the formula B-l
B-l wherein Rs is as defined for Formula I, In one embodiment of formula B-l, Rs is Ar2, wherein Ar2 5 is phenyl optionally substituted with substituents independently selected from halogen, C1-C2 alkyl and C1-C2 alkoxy. In one embodiment, Ar2 is phenyl optionally substituted with one or more halogens. In one embodiment, G is formula B-l and is has the structure:
[00146]
In one embodiment of Formula I, G is
wherein Ring B is a 6-membered aromatic carbocyclic ring optionally substituted with OH. In one embodiment, G has the formula B-2
B-2 wherein R8 is as defïned for Formula I. In one embodiment of formula B-2, R8 is Ar2, wherein Ar2 is phenyl optionally substituted with substituents independently selected from halogen, C1-C2 alkyl and C1-C2 alkoxy. In one embodiment, Ar2 is phenyl optionally substituted with one or more halogens. In one embodiment of formula B-2, R8 is Ar2, wherein Ar2 is phenyl which is unsubstituted. In one embodiment, G is formula B-2 and is has the structure:
[00147] In one embodiment, compounds of Formula I include Formula I-A, wherein:
[00148] X1 isCHorN;
[00149] R' is hydrogen orCl-C6 alkyl;
[00150] R2 is
[00151] (a)hydrogen,
[00152] (b) Cl-C 6 alkyl,
[00153] (c) hydroxyCI-C6 alkyl,
[00154] (g) (C1-C6 alkoxy)Cl-C6 alkyl-wherein said alkyl portion is optionally substituted with OH, or
[00155] (h)Cyc1;
[00156] Cyc1 is a 3-4 membered cycloalkyl ring optionally substituted with 1-2 substituents independently selected from halogen, hydroxy, CI-C3 alkyl, hydroxyC 1-C3 alkyl, CI-C3 alkoxy, (C1-C3 alkoxy)Cl-C3 alkyl- and R’RNC(=O)-;
[00157] X2 is N, R3 is absent, Ring A is a 6-membered heterocyclic ring having one additional ring nitrogen atom, and R7 is oxo, wherein G has the formula A-l
Rs I
o
A-l;
[00158] R6 is C1-C6 alkyl, hydroxyCl-C6 alkyl, C3-C6 cycloalkyl, (C3-C6 cycioalkyl)ClC6 alkyl- or hetCyc2;
[00159] hetCyc2 is a 4-6 membered saturated heterocyclic ring having a ring nitrogen atom and optionally substituted with C1-C6 alkyl;
[00160] R8 is Ar2, hetAr2, C3-C6 cycloalkyl, hetCyc3 or C1-C6 alkyl;
[00161] Ar2 is phenyl optionally substituted with one or more substituents independently selected from halogen, CI-C2 alkyl and C1-C2 alkoxy;
[00162] hetAr2 is a 5-6 membered heteroaryl having 1-2 ring nitrogen atoms and optionally substituted with one or more substituents independently selected from halogen, CJ-C2 alkyl and C1-C2 alkoxy;
[00163] hetCyc3 is a 5-6 membered heterocyclic ring having a ring oxygen atom; and
[00164] R9 is hydrogen or halogen.
[00165] In one embodiment of Formula I-A, R1 is hydrogen,
[00166] In one embodiment of Formula I-A, R9 is hydrogen.
[00167] In one embodiment of Formula I-A, R9 is halogen. In one embodiment of Formula
I-A, R9 is fluoro.
[00168] In one embodiment of Formula I-A, X1 is CH.
[00169] In one embodiment of Formula I-A, X! is CFI and R9 is halogen. In one embodiment of Formula I-A, X1 is CH and R9 is fluoro.
[00170] In one embodiment of Formula I-A, R1 is H, X1 is CFI and R9 is fluoro.
[00171] In one embodiment of Formula I-A, R1 is H, X1 is CH, Rs is Ar2 and R9 is fluoro.
[00172] In one embodiment of Formula I-A, X1 isN.
[00173] In one embodiment of Formula I-A, X1 is N and R9 is hydrogen.
[00174] In one embodiment of Formula I-A, R1 is hydrogen, X1 is N and R9 is hydrogen.
(00175] In one embodiment of Formula I-A, R1 is hydrogen, X1 is N, R8 is Ar2 and R9 is hydrogen.
[00176] In one embodiment of Formula 1-A, R3 is H.
[00177] In one embodiment of Formula 1-A, R3 is CI-C6 alkyl.
[00178] In one embodiment of Formula I-A, R3 is hydroxyCl-C6 alkyl.
[00179] In one embodiment of Formula 1-A, R3 is (C1-C6 alkoxy)Cl-C6 alkyl- wherein said alkyl portion is optionally substituted with OH.
[00180] In one embodiment of Formula 1-A, R3 is Cyc1.
[00181]
[00182] In one embodiment of Formula I-A, R8 is Ar3. In one embodiment, Ar2 is phenyl optionally substituted with one or more halogens.
[00183] In one embodiment of Formula I-A, R8 is hetAr3. In one embodiment of Formula I-A, R8 is pyrazolyl optionally substituted with C1-C6 alkyl, [00184] In one embodiment of Formula 1-A, R8 is C3-C6 cycloalkyl.
[00185] In one embodiment of Formula 1-A, R8 is hetCyc3.
[00186] In one embodiment of Formula I-A, R8 is C1-C6 alkyl.
[00187] In one embodiment, compounds of Formula I include Formula I-B, wherein:
[00188] X1 isCH or N;
[00189] R1 is hydrogen or C1-C6 alkyl;
[00190] R3 is
[00191] (c) hydroxyCI-C6 alkyl,
[00192] (d) dihydroxyC2-C6 alkyl,
[00193] (e) C1-C6 fluoroalkyl optionally substituted with OH,
[00194] (f) (di-C 1-C6 alkoxy)C2-C6 alkyl-,
[00195] (g) (C1-C6 alkoxy)Cl-C6 alkyl- wherein said alkyl portion is optionally substituted with OH, [00196] (h) Cyc1,
[00197] (i) Cyc3,
[00198] (j) (hetCyc1 )C1-C6 alkyl- wherein said alkyl portion is optionally substituted with
OH,
[00199] (k) (Ar^Cl-Cô alkyl- wherein said alkyl portion is optionally substituted with OH,
[00200] (1) (hetAr’jCl-Cô alkyl- wherein said alkyl portion is optionally substituted with
OH, or
[00201] (ni) (HOSO3)C1-C6 alkyl-;
[00202] Cyc1 is a 3-4 membered cycloalkyl ring optionally substituted with 1 -2 substituents independently selected from halogen, hydroxy, C1-C3 alkyl, hydroxyCl-C3 alkyl, C1-C3 alkoxy, (C1-C3 alkoxy)Cl-C3 alkyl-and R'RNC(=O)-;
[00203] R' and R are independently hydrogen or C1-C6 alkyl;
[00204] Cyc2 is a 5-membered cycloalkyl ring substituted with 1-2 substituents independently selected from Cl -C3 alkyl, (Cl -C3 alkoxy)Cl-C3 alkyl - and hydroxyCl-C3 alkyl[00205] hetCyc1 is a 5-6 membered saturated heterocyclic ring having 1-2 ring heteroatoms independently selected from O, N, and SO2, wherein said ring îs optionally substituted with oxo; [00206] Ar1 is phenyi;
[00207] hetAr1 is pyridyl;
[00208] X2 is N, R3 is absent, Ring A is a 6-membered heterocyclic ring having an additional ring nitrogen atom, and R7 is hydrogen, wherein G has the formula A-2
A-2;
[00209] R6 is hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl;
[00210] R8 is Ar2, hetAr2, C3-C6 cycloalkyl, hetCyc3 or C1-C6 alkyl;
[00211] Ar2 is phenyi optionally substituted with one or more substituents independently selected from halogen, C1-C2 alkyl and C1-C2 alkoxy;
[00212] hetAr is a 5-6 membered heteroaryl having 1-2 ring nitrogen atoms and optionally substituted with one or more substituents independently selected from halogen, CJ-C2 alkyl and C1-C2 alkoxy;
[00213] hetCyc3 is a 5-6 membered heterocyclic ring having a ring oxygen atom; and [00214] R9 is hydrogen or halogen.
[00215] In one embodiment of Formula I-B, X1 is CH.
[00216] In one embodiment of Formula I-B, X1 isN.
[00217] In one embodiment of Formula I-B, R8 is Ar2. In one embodiment, Ar2 is phenyl optionally substîtuted with one or more halogens.
[00218] In one embodiment of Formula I-B, R1 is hydrogen.
[00219] In one embodiment of Formula I-B, R9 is hydrogen.
[00220] In one embodiment of Formula I-B, R9 is halogen. In one embodiment of Formula
I-B, R9 is fluoro.
(00221 ] In one embodiment of Formula I-B, R1 is hydrogen, X1 is CH, R8 is Ar2 wherein Ar2 is as defined for Formula I-B, and R9 is fluoro.
[00222] In one embodiment of Formula I-B, R1 is hydrogen, X1 is CH, R8 is Ar2 wherein Ar2 is phenyl optionally substîtuted with one or more halogens, and R9 is fluoro.
100223] In one embodiment of Formula I-B, R1 is hydrogen, X1 is N, R8 is Ar2 wherein Ar2 is as defined for Formula 1-B, and R9 is hydrogen.
[00224] In one embodiment of Formula I-B, R1 is hydrogen, X1 is N, R8 is Ar2 wherein Ar2 is phenyl optionally substîtuted with one or more halogens, and R9 is hydrogen.
[00225] In one embodiment, compounds of Formula I include Formula I-C, wherein:
[00226] X1 isCHorN;
[00227] R1 is hydrogen;
[00228] R2 is
[00229] (c) hydroxyC I-C6 alkyl, |00230] (e) C1-C6 fluoroalkyl optionally substîtuted with OH, or
[00231] (g) (Cl -C6 alkoxy)Cl-C6 alkyl- wherein said alkyl portion is optionally substîtuted with OH;
[00232] X2 is N, R3 is absent, Ring A is a 6-membered heterocyclic ring, wherein G has the formula A-3
R6 R7
O
A-3;
[00233] R6 is halogen, C1-C6 alkyl, C1-C6 alkoxy, or C3-C6 cycloalkyl;
[00234] R7 is hydrogen;
[00235] R8 is Ar2, hetAr2, C3-C6 cycloalkyl, hetCyc3 or C1-C6 alkyl;
[00236] Ar2 is phenyl optionally substituted with one or more substituents independently selected from halogen, CI-C2 alkyl and C1-C2 alkoxy;
[00237] hetAr2 is a 5-6 membered heteroaryl having 1 -2 ring nitrogen atoms and optionally substituted with one or more substituents independently selected from halogen, C1-C2 alkyl and C1-C2 alkoxy;
[00238] hetCyc3 is a 5-6 membered heterocyclic ring having a ring oxygen atom; and
[00239] R9 is hydrogen or halogen.
[00240] In one embodiment of Formula I-C, R9 is hydrogen.
[00241] In one embodiment of Formula 1-C, R9 is halogen. In one embodiment of Formula
I-C, R9 is fluoro.
[00242] In one embodiment of Formula I-C, X1 is CH.
[00243] InoneembodimentofFormulaI-C,X' is CH and R9 is halogen. In one embodiment of Formula I-C, X1 is CH and R9is fluoro.
[00244] In one embodiment of Formula I-C, X1 is N.
[00245] In one embodiment of Formula I-C, X1 is N and R9 is hydrogen,
[00246] In one embodiment of Formula I-C, Rs is Ar2. In one embodiment, Ar2 is phenyl optionally substituted with one or more halogens.
[00247] In one embodiment of Formula 1-C, X1 is CH, R9 is fluoro, and R8 is Ar2 wherein Ar2 is phenyl optionally substituted with one or more halogens.
[00248] In one embodiment of Formula 1-C, X1 is N, R9 is hydrogen, and R8 is Ar2 wherein Ar2 is phenyl optionally substituted with one or more halogens.
[00249] In one embodiment, compounds of Formula I include Formula 1-D, wherein:
|00250] X'isCH;
[00251] R1 is hydrogen;
[00252] R2 is
[00253] (c) hydroxyC 1-C6 alkyl,
[00254] (e) C1-C6 fluoroalkyl optionally substituted with OH, or
[00255] (g) (C 1-C6 alkoxy)C I-C6 alkyl- wherein said alkyl portion is optionally substituted with OH;
[00256] R3 is absent;
[00257] X2 is N;
[00258] X2 is N, R3 is absent. Ring A is a 6-membered heterocyclic ring having two additional ring nitrogen atoms, and R7 is oxo or thiooxo, wherein G has the formula
A-4 ;
[00259] R6isCl-C6alkyl;
[00260] Y is O or S;
[00261] R8 is Ar2, hetAr2, C3-C6 cycloalkyl, hetCyc3 or C1-C6 alkyl;
[00262] Ar2 is phenyl optionally substituted with one or more substituents independently selected from halogen. Cl-C2 alkyl and C l -C2 alkoxy;
[00263] hetAr2 is a 5-6 membered heteroaryl having 1-2 ring nitrogen atoms and optionally substituted with one or more substituents independently selected from halogen, CI-C2 alkyl and C1-C2 alkoxy;
[00264] hetCyc3 is a 5-6 membered heterocyclic ring having a ring oxygen atom; and
[00265] R9 is hydrogen or halogen.
[00266] In one embodiment of Formula 1-D, R8 is Ar2. In one embodiment, Ar2 is phenyl optionally substituted with one or more halogens.
[00267] In one embodiment of Formula I-D, R9 is halogen. In one embodiment of Formula I-D, R9 is fluoro.
[00268] In one embodiment of Formula I-D, Ar2 is phenyl optionally substituted with one or more halogens and R9 is fluoro.
[00269] In one embodiment, compounds of Formula 1 include Formula I-E, wherein:
[00270] X1 is CH or N;
[00271] R2 îs
[00272] (c) hydroxyC 1-C6 alkyl,
[00273] (e) C1-C6 fluoroalkyl optionally substituted with OH, or
[00274] (g) (C1-C6 alkoxy)Cl-C6 alkyl- wherein said alkyl portion is optionally substituted with OH;
[00275] X2 is C, R3 is absent. Ring A is a 6-membered heterocyclic ring having a ring nitrogen atom, wherein G has the formula A-5
[00276] R6 is C1-C6 alkyl;
[00277] R7 is hydrogen;
[00278] R8 is Ar2, hetAr2, C3-C6 cycloalkyl, hetCyc3 or C1-C6 alkyl;
[00279] Ar3 is phenyl optionally substituted with one or more substituents îndependently selected from halogen, C1-C2 alkyl and C1-C2 alkoxy;
[00280] hetAr2 is a 5-6 membered heteroaryl having 1-2 ring nitrogen atoms and optionally substituted with one or more substituents îndependently selected from halogen, C1-C2 alkyl and C1-C2 alkoxy;
[00281] hetCyc3 is a 5-6 membered heterocyclic ring having a ring oxygen atom; and
[00282] R9 is hydrogen or halogen.
[00283] In one embodiment of Formula I-E, R1 is hydrogen.
[00284] In one embodiment of Formula I-E, X1 is CH.
[00285] In one embodiment of Formula I-E, X1 is N.
[00286] In one embodiment of Formula I-E, R8 is Ar2. In one embodiment, Ar2 is phenyl optionally substituted with one or more halogens.
[00287] In one embodiment of Formula I-E, R9 îs hydrogen.
[00288] In one embodiment of Formula I-E, R9 is fluoro.
|00289] In one embodiment of Formula I-E, R1 is hydrogen, X1 is CH, Ar2 is phenyl optionally substituted with one or more halogens, and R9 is fluoro.
[00290] In one embodiment of Formula I-E, R1 is hydrogen, X1 is N, Ar2 is phenyl optionally substituted with one or more halogens, and R9 is hydrogen.
[00291| In one embodiment, compounds of Formula I include Formula I-F, wherein: [00292] X1 is CH;
[00293] R1 is hydrogen;
[00294] R2 is (c) hydroxyCl-C6 alkyl;
[00295] X2 is N, R3 is absent, Ring A is a 6-membered heterocyclic ring having two additional ring nitrogen atoms, wherein G has the formula A-6
A-6
[00296] R6 is hydrogen;
[00297] R7 is hydrogen;
[00298] Rs is Ar2, hetAr2, C3-C6 cycioalkyl, hetCyc3 or C1-C6 alkyl;
[00299] Ar2 is phenyl optionally substituted with one or more substituents independently selected from halogen, C1-C2 alkyl and C1-C2 alkoxy;
[00300] hetAr2 is a 5-6 membered heteroaryl having 1 -2 ring nitrogen atoms and optionally substituted with one or more substituents independently selected from halogen, C1-C2 alkyl and CI-C2 alkoxy;
[00301] hetCyc3 is a 5-6 membered heterocyclic ring having a ring oxygen atom; and [00302] R9 is hydrogen or halogen.
[00303] In one embodiment of Formula I-F, R8 is Ar2. In one embodiment, Ar2 is phenyl optionally substituted with one or more halogens.
[00304] In one embodiment of Formula I-F, R9 is halogen. In one embodiment of Formula I-F, R9 ts fluoro.
100305| In one embodiment of Formula 1-F, Ar2 is phenyl optionally substituted with one or more halogens, and R9 is fluoro.
|00306] In one embodiment, compounds of Formula I include Formula I-G, wherein:
[00307] X'isCH;
[00308] R! is hydrogen;
[00309] R2 is (c) hydroxyC I-C6 alkyl;
[00310] X2 is N, R5 is hydrogen or methyl, and Ring A is a 6-membered heterocyclic ring having a ring nitrogen atom, wherein G has the formula A-7
Re R7
O
A-7
[00311] R6 is hydrogen and R7 is hydrogen,
[00312] or R6 and R7 are on the same carbon atom and R6 and R7 together with the carbon atom to which they are attached fonn a cyclopropyl ring;
[00313] R8 is Ar2, hetAr2, C3-C6 cycloalkyl, hetCyc3 or C1-C6 alkyl;
[00314] Ar2 is phenyl optionally substituted with one or more substituents independentiy selected from halogen, C1-C2 alkyl and C1-C2 alkoxy;
[00315] hetAr2 is a 5-6 membered heteroaryl having 1-2 ring nitrogen atoms and optionally substituted with one or more substituents independentiy selected from halogen, C1-C2 alkyl and CI-C2 alkoxy;
[00316] hetCyc3 is a 5-6 membered heterocyclic ring having a ring oxygen atom; and [00317] R9 is hydrogen or halogen.
[00318] In one embodiment of Formula I-G, R8 is Ar2. In one embodiment, Ar2 is phenyl optionally substituted with one or more halogens.
[00319] In one embodiment of Formula I-G, R9 is halogen. In one embodiment of Formula I-G, R9 is fluoro.
[00320] In one embodiment of Formula I-G, Ar2 is phenyl optionally substituted with one or more halogens, and R9 is fluoro.
[00321] In one embodiment, compounds of Formula I include Formula 1-H, wherein:
[00322] X' is CH;
[00323] R! is hydrogen;
[00324] R2 is (c) hydroxyCl-C6 alkyl;
[00325] X2 is N, R3 is absent, Ring A is a 5-membered heterocyclic ring having an additional ring nitrogen atom, wherein G has the formula A-8
A-8 ;
[00326] R6 is C1-C6 alkyl;
[00327] R7 is hydrogen or C1-C6 alkyl;
[00328) R8 is Ar2, hetAr2, C3-C6 cycloalkyl, hetCyc3 or C1-C6 alkyl;
[00329] Ar2 is phenyl optionally substituted with one or more substituents îndependently selected from halogen, C1-C2 alkyl and CI-C2 alkoxy;
[00330] hetAr2 is a 5-6 membered heteroaryl having 1-2 ring nitrogen atoms and optionally substituted with one or more substituents îndependently selected from halogen, Cl -C2 alkyl and C1-C2 alkoxy;
[00331] hetCyc3 is a 5-6 membered heterocyclic ring having a ring oxygen atom; and [00332| R9 is hydrogen or halogen.
[00333] In one embodiment of Formula I-H, R8 is Ar. In one embodiment of Formula 1-H, Rs îs Ar2 wherein Ar2 is phenyl optionally substituted with one or more halogens.
[00334] In one embodiment of Formula I-H, R9 is halogen. In one embodiment of Formula 1-H, R9 is fluoro.
[00335] In one embodiment of Formula I-H, R8 is Ar2 wherein Ar2 îs phenyl optionally substituted with one or more halogens, and R9 is fluoro.
[00336] In one embodiment, compounds of Formula I include Formula I-I, wherein:
[00337] X’isCH;
[00338] R1 is hydrogen;
[00339] R2 is (c) hydroxyCl-C6 alkyl;
[00340] G is formula B-I
B-l ;
[003411 R8 is Ar2, hetAr2, C3-C6 cycloalkyl, hetCyc3 or C1-C6 alkyl;
[00342] Ar2 is phenyl optionally substituted with one or more substituents independently selected from halogen, C1-C2 alkyl and C1-C2 alkoxy;
[00343] hetAr2 is a 5-6 membered heteroaiyl having 1 -2 ring nitrogen atoms and optionally substituted with one or more substituents independently selected from halogen, C1-C2 alkyl and C1-C2 alkoxy;
[00344] hetCyc3 is a 5-6 membered heterocyclic ring having a ring oxygen atom; and
[00345] R9 is hydrogen or halogen.
[00346] In one embodiment of Formula 1-1, R8 is Ar2. In one embodiment, Ar2 is phenyl optionally substituted with one or more halogens.
[00347] In one embodiment of Formula I-I, R9 is halogen. In one embodiment of Formula I-I, R9 is fluoro.
[00348] In one embodiment of Formula I-I, Ar2 is phenyl optionally substituted with one or more halogens, and R9 is fluoro.
[00349] In one embodiment, compounds of Formula I include Formula 1-J, wherein:
[00350] X'isCH;
[00351] R1 is hydrogen;
[00352] R2 is (c) hydroxyC 1-C6 alkyl;
[00353] G is formula B-2
B-2 ;
[00354] Rs is Ar2, hetAr2, C3-C6 cycloalkyl, hetCyc2 or C1-C6 alkyl;
[00355] Ar2 is phenyl optionally substituted with one or more substituents independently selected from halogen, C1-C2 alkyl and C1-C2 alkoxy;
[00356] hetAr2 is a 5-6 membered heteroaryl having î-2 ring nitrogen atoms and optionally substituted with one or more substituents independently selected from halogen, C1-C2 alkyl and
C1-C2 alkoxy;
[00357] hetCyc3 is a 5-6 membered heterocyclic ring having a ring oxygen atom; and [00358] R9 is hydrogen or halogen.
[00359] In one embodiment of Formula I-J, R8 is Ar2. In one embodiment, Ar2 is phenyl 5 optionally substituted with one or more halogens.
[00360] In one embodiment of Formula I-J, R9 is halogen. In one embodiment of Formula 1-J, R9 is fluoro.
[00361] In one embodiment of Formula I-J, Ar2 is phenyl optionally substituted with one or more halogens, and R9 is fluoro.
|00362] The compounds of Formula I include pharmaceutically acceptable salts thereof. In addition, the compounds of Formula I also include other salts of such compounds which are not necessarily pharmaceutically acceptable salts, and which may be useful as intermediates for preparing and/or purifying compounds of Formula I and/or for separating enantiomers of compounds of Fonnula I. Non-limiting examples of pharmaceutically acceptable salts of compounds of Formula I include hydrochloride salts.
|00363] It will further be appreciated that the compounds of Formula 1 or their salts may be isolated in the form of solvatés, and accordingly that any such solvaté is included within the scope of the présent invention. For example, compounds of Formula I and salts thereof can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, 20 ethanoL and the like.
[00364] In one embodiment, the compounds of Formula I include the compounds of Examples 1-201 and stereoisomers and pharmaceutically acceptable salts and solvatés thereof. In one embodiment, the compounds of Examples 1-201 are in the free base form. In one embodiment, one or more compounds of Examples 1 -201 are hydrochloride acid salts.
5 [00365] In one embodiment, the compound of fonnula I is a compound of Example No. 25, 37,
46, 48, 55, 58, 72, 76,77,78, 83, 84, 85, 91, 97, 100, 103, 105, 107, 108, 114, 115, 119, 121, 124, 125, 126, 127, 129, 151, 152, 163, 169, 188, 190, 199, 200, or 201, or a pharmaceutically acceptable sait or solvaté thereof.
[00366] In one embodiment, the compound of formula I is a compound of Example No. 25, or 30 a pharmaceutically acceptable sait or solvaté thereof.
[00367] In one embodiment, the compound of formula I is a compound of Example No. 37, or a pharmaceutically acceptable sait or solvaté thereof.
[00368] In one embodiment, the compound of formula I is a compound of Example No. 46, or a pharmaceutically acceptable sait or solvaté thereof.
[00369] In one embodiment, the compound of formula I is a compound of Example No. 48, or a pharmaceutically acceptable sait or solvaté thereof.
[00370] In one embodiment, the compound of formula I is a compound of Example No. 55, or a pharmaceutically acceptable sait or solvaté thereof.
[00371] In one embodiment, the compound of formula I is a compound of Example No. 58, or a pharmaceutically acceptable sait or solvaté thereof.
[00372] In one embodiment, the compound of formula 1 is a compound of Example No. 72, or a pharmaceutically acceptable sait or solvaté thereof.
[00373] In one embodiment, the compound of formula I is a compound of Example No. 76, or a pharmaceutically acceptable sait or solvaté thereof.
[00374] In one embodiment, the compound of formula 1 is a compound of Example No. 77, or a pharmaceutically acceptable sait or solvaté thereof.
[00375] In one embodiment, the compound of formula I is a compound of Example No. 78, or a pharmaceutically acceptable sait or solvaté thereof.
[00376] In one embodiment, the compound of formula I is a compound of Example No. 83, or a pharmaceutically acceptable sait or solvaté thereof.
[00377] in one embodiment, the compound of formula I is a compound of Example No. 84, or a pharmaceutically acceptable sait or solvaté thereof.
[00378] In one embodiment, the compound of formula I is a compound of Example No. 85, or a pharmaceutically acceptable sait or solvaté thereof.
[00379] In one embodiment, the compound of formula I is a compound of Example No. 91, or a pharmaceutically acceptable sait or solvaté thereof.
[00380] In one embodiment, the compound of formula I is a compound of Example No. 97, or a pharmaceutically acceptable sait or solvaté thereof.
[00381] In one embodiment, the compound of formula 1 is a compound of Example No. 100, or a pharmaceutically acceptable sait or solvaté thereof.
[00382] In one embodiment, the compound of formula I is a compound of Example No. 103, or a pharmaceutically acceptable sait or solvaté thereof.
[00383] In one embodiment, the compound of formula I is a compound of Example No. 105, or a pharmaceuticaliy acceptable sait or solvaté thereof.
[00384] In one embodiment, the compound of formulai is a compound of Example No. 107, or a pharmaceuticaliy acceptable sait or solvaté thereof.
[00385] In one embodiment, the compound of formula 1 is a compound of Example No. 108, or a pharmaceuticaliy acceptable sait or solvaté thereof.
[00386| In one embodiment, the compound of formula I is a compound of Example No. 114, or a pharmaceuticaliy acceptable sait or solvaté thereof.
[00387] In one embodiment, the compound of formula I is a compound of Example No. 115, or a pharmaceuticaliy acceptable sait or solvaté thereof.
[00388] In one embodiment, the compound of formula I is a compound of Example No. 119, or a pharmaceuticaliy acceptable sait or solvaté thereof.
[00389] In one embodiment, the compound of formula I is a compound of Example No. 121, or a pharmaceuticaliy acceptable sait or solvaté thereof.
[00390] In one embodiment, the compound of formula I is a compound of Example No. 124, or a pharmaceuticaliy acceptable sait or solvaté thereof.
[00391] In one embodiment, the compound of formula I is a compound of ExampleNo. 125, or a pharmaceuticaliy acceptable sait or solvaté thereof.
[00392] In one embodiment, the compound of formula 1 is a compound of Example No. 126, or a pharmaceuticaliy acceptable sait or solvaté thereof.
[00393] In one embodiment, the compound of formula 1 is a compound of Example No. 127, or a pharmaceuticaliy acceptable sait or solvaté thereof.
[00394] In one embodiment, the compound of formula I is a compound of Example No. 129, or a pharmaceuticaliy acceptable sait or solvaté thereof.
[00395] In one embodiment, the compound of formula I is a compound of Example No. 151, or a pharmaceuticaliy acceptable sait or solvaté thereof.
[00396] In one embodiment, the compound of formulai is a compound of Example No. 152, or a pharmaceuticaliy acceptable sait or solvaté thereof.
[00397] In one embodiment, the compound of formula I is a compound of Example No. 163, or a pharmaceuticaliy acceptable sait or solvaté thereof.
[00398] In one embodiment, the compound of formula I is a compound of Example No. 169, or a pharmaceutically acceptable sait or solvaté thereof.
[00399] In one embodiment, the compound of formula 1 is a compound of Example No. 188, or a pharmaceutically acceptable sait or solvaté thereof.
[00400] In one embodiment, the compound of formula 1 is a compound of Example No. 190, or a pharmaceutically acceptable sait or solvaté thereof.
[00401] In one embodiment, the compound of formula I is a compound of Example No. 199, or a pharmaceutically acceptable sait or solvaté thereof.
[00402] In one embodiment, the compound of formula I is a compound of Example No. 200, or a pharmaceutically acceptable sait or solvaté thereof.
[00403] In one embodiment, the compound of formulai is acompound ofExample No. 201, or a pharmaceutically acceptable sait or solvaté thereof.
[00404] In one embodiment, compounds of Formula 1 include compounds of Formula II and stereoisomers, tautomers and pharmaceutically acceptable salts thereof, wherein:
[00405] X'isCHorN;
[00406] R1 is hydrogen or C1-C6 alkyl;
[00407] R2 is |Û0408] (a) hydrogen,
[00409] (b) Cl-C6 alkyl,
[00410] (c) hydroxyCl-C6 alkyl,
[00411] (d) dihydroxyC2-C6 alkyl,
[00412] (e) C1-C6 fluoroalkyl optionally substîtuted with OH,
[00413] (g) (CI-C6 a!koxy)C J-C6 alkyl- wherein said alkyl portion is optionally substîtuted with OH,
[00414] (h) Cyc1,
[00415] (i) Cyc2,
[00416] (j) (hetCyc')Cl-C6 alkyl- wherein said alkyl portion is optionally substîtuted with
OH,
[00417] (k) (Ar')CI-C6 alkyl-wherein said alkyl portion is optionally substîtuted with OH,
[00418] (l) (hetAr’)CI-C6 alkyl- wherein said alkyl portion is optionally substîtuted with
OH, or
[00419] (m) (HOSOs)Cl-C6 alkyl-;
[00420] Cyc1 is a 3-4 membered cycloalkyl ring optionally substituted with 1 -2 substituents independently selected from halogen, hydroxy, hydroxyCl-C3 alkyl, C1-C3 alkoxy, (C1-C3 alkoxy)Cl-C3 alkyl-, and R'RNC(=O)-;
[00421] R' and R are independently selected from C1-C6 alkyl;
[00422] Cyc2 is a 5-membered cycloalkyl ring substituted with 1-2 substituents independently selected from C1-C3 alkyl, (C1-C3 alkoxy)Cl-C3 alkyl- and hydroxyCl-C3 alkyl-; |00423] hetCyc1 is a 5-6 membered saturated heterocyclic ring having 1-2 ring heteroatoms independently selected from O, N and SO2, wherein said ring is optionally substituted with oxo;
[00424] Ar1 is phenyl;
[00425] hetAr1 is pyridyl;
[00426] G is
[00427] X2isCorN;
[00428] Ring A, includîng the atoms at the points of attachment, is a 5-6 membered heterocyclic ring optionally having an additional 1-2 ring nitrogen atoms when X2 is N and having one ring nitrogen atom when X2 is C;
[00429] R3 is hydrogen or absent;
[00430| R6 is hydrogen, halogen, C1-C6 alkyl, C1-C6 alkoxy, hydroxyCl-C6 alkyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)Cl-C6 alkyl- or hetCyc2, provided that when R6 is halogen and is on the ring carbon atom adjacent to the carbon linked to the -NHC(=O)- moiety of Formula I, then R6 is not halogen;
[00431] R7 is hydrogen, C1-C6 alkyl, oxo or thioxo;
[00432] hetCyc2 is a 4 membered saturated heterocyclic ring having a ring nitrogen atom substituted with C1-C6 alkyl;
[00433] Ring B, including the atoms at the points of attachment, is a 6-membered saturated carbocyclic optionally substituted with oxo or a 6-membered aromatic carbocyclic ring optionally substituted with OH;
[00434] R8 is Ar2, hetAr2, C3-C6 cycloalkyl, hetCyc3 orCI-C6 alkyl;
[00435] Ar2 is phenyl optionally substituted with one or more substituents independently selected from halogen;
[00436] hetAr2 is a 5-6 membered heteroaryî having 1-2 ring nitrogen atoms and optionally substituted with one or more substituents independently selected from C1-C2 alkyl; and
[00437] R9 is hydrogen or halogen.
[00438] In one embodiment of Formula II, G is
O wherein X2 is N, R3 is absent, Ring A is a 6-membered heterocyclic ring having one additional ring nitrogen atom, and R7 is oxo, such that G has the formula A-l
[00439] wherein R6 is C1-C6 alkyl, hydroxyC 1-C6 alkyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)Cl-C6 alkyl- or hetCyc2, and hetCyc2 and R8 are as defined for Formula II.
[00440] In one embodiment of Formula 11, G is
wherein X2 is N, R3 is absent, Ring A is a 6-membered heterocyclic ring having an additional ring nitrogen atom, and R7 is hydrogen, such that G has the formula A-2
[00441] wherein R6 is hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl, R7 is hydrogen, and R8 5 is as defined for Formula IL
[00442J In one embodiment of Formula II, G is
O wherein X2 is N, R3 is absent, Ring A is a 6-membered heterocyclic ring, such that G has the formula A-3
R6 R7
O
A-3 wherein R6 is halogen, C1-C6 alkyl, Cl -C6 alkoxy, orC3-C6 cycloalkyl, R7 is hydrogen, provided that when R6 is on the ring carbon atom adjacent to the carbon linked to the -NHC(=O)- moiety 15 of Formula Π, then R6 is not halogen, and R8 is as defined for Formula II. As used herein, the ring carbon atom adjacent to the carbon linked to the -NHC(=O)- moiety of Formula II refers to the carbon identifîed by the asterisk in the following structure:
[00443] In one embodiment of Formula II, G is
wherein X2 is N, R3 is absent, Ring A is a 6-membered heterocyclic ring having two additionaî ring nitrogen atoms, and R7 is oxo or thiooxo, such that G has the formula A-4
R6
I
o
A-4
[00444] wherein Y is O or S, R6 is C1-C6 alkyl, and R8 is as defined for Formula II.
|00445] In one embodiment of Formula II, G is
wherein X2 is C, R3 is absent, Ring A is a 6-membered heterocyclic ring having a ring nitrogen atom, such that G has the formula A-5
R6
I
o
A-5 |00446]
[00447] wherein R6 is CI-C6 alkyl, R7 is hydrogen, and R8 is as defined for Formula II. In one embodiment of Formula II, G is
Ο wherein X2 is N, R3 is absent. Ring A is a 6-membered heterocyclic ring having two additional ring nitrogen atoms, such that G has the formula A-6
[00448] wherein R6 and R7 are hydrogen and R8 is as defined for Formula II.
[00449] In one embodiment of Formula II, G is
O wherein X2 is N, R3 is hydrogen or methyl, and Ring A is a 6-membered heterocyclic ring having a ring nitrogen atom, such that G has the formula A-7
R6 R7
O
A-7
[00450]
[00451] wherein R6 and R7 are hydrogen, and R8 is as defined for Formula II. In one embodiment of Formula II, G is
Ο wherein X2 is N, R3 is absent. Ring A is a 5-membered heterocyclic ring having an additional ring nitrogen atom, such that G has the formula A-S
A-8
[00452]
Formula IL
[00453] wherein R6 is C1-C6 alkyl, R7 is hydrogen or C1-C6 alkyl, and R8 is as defined for
In one embodiment of Formula II, G is
[00454]
[00455]
[00456]
[00457]
0X0.
wherein Ring B and R8 are as defined for Formula IL In one embodiment of Formula II, G is
wherein Ring B is a 6-membered saturated carbocyclic optionally substituted with
[00458] In one embodiment of Formula II, G has the formula B-l
B-l
[00459] wherein R8 is as defined for Formula II.
[00460] In one embodiment, the compound of Formula II is a compound of Example No. 2,
3, 4, 5, 6, 7, 8, 12, 13, 14, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31,32, 33,34,
35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61,62,
63, 64, 65, 67, 68, 69, 70, 71, 72, 73, 75, 76, 77, 79, 80, 81, 84, 86, 87, 88, 89, 90, 91, 92, 93,94,
95, 96, 97, 98,99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 113, 114, 1J 5, 1 16, 117,
118, 119, 120, 121, 122, 128, 142, 144, 145, 146, 147, 148, 150, 151, 152, 153, 154, 155, 156,
157, 158, 159, 160, 161, 162, 163, 164, 166, 167, 168, 169, 170, 171, 172, 173, 179, 181, 182, 183, 184, 185, 186, 187, 189, 191, 192, 193, 194, 195, 196, or 197, or a pharmaceutically acceptable sait or solvaté thereof.
[00461] In one embodiment, the compound of Formula II is a compound of Example No. 2, 15 or a pharmaceutically acceptable sait or solvaté thereof.
|00462) In one embodiment, the compound of Formula II is a compound of Example No. 3, or a pharmaceutically acceptable sait or solvaté thereof.
[00463] In one embodiment, the compound of Formula II is a compound of Example No. 4, or a pharmaceutically acceptable sait or solvaté thereof.
0 [00464] In one embodiment, the compound of Formula II is a compound of Example No. 5, or a pharmaceutically acceptable sait or solvaté thereof.
|00465] In one embodiment, the compound of Formula II is a compound of Example No. 6, or a pharmaceutically acceptable sait or solvaté thereof.
[00466] In one embodiment, the compound of Formula II is a compound of Example No. 7, 25 or a pharmaceutically acceptable sait or solvaté thereof.
[00467] In one embodiment, the compound of Formula II is a compound of Example No. 8, or a pharmaceutically acceptable sait or solvaté thereof.
[00468] In one embodiment, the compound of Formula II is a compound of Example No. 12, or a pharmaceutically acceptable sait or solvaté thereof.
[00469] In one embodiment, the compound of Fonnula II is a compound of Example No.
13, or a pharmaceutically acceptable sait or solvaté thereof.
[00470] In one embodiment, the compound of Formula II is a compound of Example No.
14, or a pharmaceutically acceptable sait or solvaté thereof.
[00471] In one embodiment, the compound of Formula II is a compound of Example No.
16, or a pharmaceutically acceptable sait or solvaté thereof.
[00472] In one embodiment, the compound of Formula II is a compound of Example No.
, or a pharmaceutically acceptable sait or solvaté thereof.
[00473] In one embodiment, the compound of Formula II is a compound of Example No.
18, or a pharmaceutically acceptable sait or solvaté thereof.
[00474] In one embodiment, the compound of Formula II is a compound of Example No.
19, or a pharmaceutically acceptable sait or solvaté thereof.
[00475] In one embodiment, the compound of Formula II is a compound of Example No.
20, or a pharmaceutically acceptable sait or solvaté thereof.
[00476] In one embodiment, the compound of Formula II is a compound of Example No.
21, or a pharmaceutically acceptable sait or solvaté thereof.
[00477] In one embodiment, the compound of Formula II is a compound of Example No.
22, or a pharmaceutically acceptable sait or solvaté thereof.
[00478] In one embodiment, the compound of Formula II is a compound of Example No.
23, or a pharmaceutically acceptable sait or solvaté thereof.
[00479] In one embodiment, the compound of Formula II is a compound of Example No.
24, or a pharmaceutically acceptable sait or solvaté thereof.
[00480] In one embodiment, the compound of Formula II is a compound of Example No.
25, or a pharmaceutically acceptable sait or solvaté thereof.
[00481| In one embodiment, the compound of Formula II is a compound of Example No.
26, or a pharmaceutically acceptable sait or solvaté thereof.
[00482) In one embodiment, the compound of Formula II is a compound of Example No.
27, or a pharmaceutically acceptable sait or solvaté thereof.
[00483] In one embodiment, the compound of Formula II is a compound of Example No. 28, or a pharmaceutically acceptable sait or solvaté thereof.
[00484] In one embodiment, the compound of Formula II is a compound of Example No. 29, or a pharmaceutically acceptable sait or solvaté thereof.
[00485] In one embodiment, the compound of Formula II is a compound of Example No.
, or a pharmaceutically acceptable sait or solvaté thereof.
[00486] In one embodiment, the compound of Formula II is a compound of Example No.
l, or a pharmaceutically acceptable sait or solvaté thereof.
[00487] In one embodiment, the compound of Formula II is a compound of Example No.
32, or a pharmaceutically acceptable sait or solvaté thereof.
[00488] In one embodiment, the compound of Formula II is a compound of Example No.
33, or a pharmaceutically acceptable sait or solvaté thereof.
[00489] In one embodiment, the compound of Formula II is a compound of Example No.
34, or a pharmaceutically acceptable sait or solvaté thereof.
[00490J In one embodiment, the compound of Formula II is a compound of Example No.
35, or a pharmaceutically acceptable sait or solvaté thereof.
[00491] In one embodiment, the compound of Formula II is a compound of Example No.
36, or a pharmaceutically acceptable sait or solvaté thereof.
|00492) In one embodiment, the compound of Formula II is a compound of Example No.
37, or a pharmaceutically acceptable sait or solvaté thereof.
[00493] In one embodiment, the compound of Formula II is a compound of Example No.
38, or a pharmaceutically acceptable sait or solvaté thereof.
[00494] In one embodiment, the compound of Formula II is a compound of Example No.
39, or a pharmaceutically acceptable sait or solvaté thereof.
5 [00495] in one embodiment, the compound of Formula II is a compound of Example No.
4Ü, or a pharmaceutically acceptable sait or solvaté thereof.
[00496] In one embodiment, the compound of Formula II is a compound of Example No.
41, or a pharmaceutically acceptable sait or solvaté thereof.
[00497] In one embodiment, the compound of Formula II is a compound of Example No.
42, or a pharmaceutically acceptable sait or solvaté thereof.
[00498] In one embodiment, the compound of Formula II is a compound of Example No.
43, or a pharmaceutically acceptable sait or solvaté thereof.
[00499] In one embodiment, the compound of Formula II is a compound of Example No. 44, or a pharmaceutically acceptable sait or solvaté thereof.
[00500] In one embodiment, the compound of Formula II is a compound of Example No.
45, or a pharmaceutically acceptable sait or solvaté thereof.
[00501] In one embodiment, the compound of Formula II is a compound of Example No.
46, or a pharmaceutically acceptable sait or solvaté thereof.
[00502] In one embodiment, the compound of Formula II is a compound of Example No.
47, or a pharmaceutically acceptable sait or solvaté thereof.
[00503| In one embodiment, the compound of Formula II is a compound of Example No.
50, or a pharmaceutically acceptable sait or solvaté thereof.
[00504] In one embodiment, the compound of Formula II is a compound of Example No.
51, or a pharmaceutically acceptable sait or solvaté thereof.
[00505] In one embodiment, the compound of Formula II is a compound of Example No.
52, or a pharmaceutically acceptable sait or solvaté thereof.
[00506] In one embodiment, the compound of Formula II is a compound of Example No.
53, or a pharmaceutically acceptable sait or solvaté thereof.
[00507] In one embodiment, the compound of Formula II is a compound of Example No.
0 54, or a pharmaceutically acceptable sait or solvaté thereof.
[00508] In one embodiment, the compound of Formula II is a compound of Example No.
55, or a pharmaceutically acceptable sait or solvaté thereof.
[00509] In one embodiment, the compound of Formula II is a compound of Example No.
56, or a pharmaceutically acceptable sait or solvaté thereof.
[00510] In one embodiment, the compound of Formula II is a compound of Example No.
57, or a pharmaceutically acceptable sait or solvaté thereof.
[00511) In one embodiment, the compound of Formula II is a compound of Example No.
58, or a pharmaceutically acceptable sait or solvaté thereof.
[00512] In one embodiment, the compound of Formula II is a compound of Example No.
59, or a pharmaceutically acceptable sait or solvaté thereof.
[00513] In one embodiment, the compound of Formula II is a compound of Example No. 60, or a pharmaceutically acceptable sait or solvaté thereof.
[00514] In one embodiment, the compound of Formula II is a compound of Example No. 61, or a pharmaceutically acceptable sait or solvaté thereof.
[00515] In one embodiment, the compound of Formula II is a compound of Example No.
, or a pharmaceutically acceptable sait or solvaté thereof.
[00516] In one embodiment, the compound of Formula II îs a compound of Example No.
, or a pharmaceutically acceptable sait or solvaté thereof.
[00517] In one embodiment, the compound of Formula II is a compound of Example No.
64, or a pharmaceutically acceptable sait or solvaté thereof.
[00518] In one embodiment, the compound of Formula 11 is a compound of Example No.
65, or a pharmaceutically acceptable sait or solvaté thereof.
[00519] In one embodiment, the compound of Formula II is a compound of Example No.
66, or a pharmaceutically acceptable sait or solvaté thereof.
[00520] In one embodiment, the compound of Formula II is a compound of Example No.
67, or a pharmaceutically acceptable sait or solvaté thereof.
|00521] In one embodiment, the compound of Formula II is a compound of Example No.
6S, or a pharmaceutically acceptable sait or solvaté thereof.
[00522] In one embodiment, the compound of Formula II is a compound of Example No.
0 69, or a pharmaceutically acceptable sait or solvaté thereof.
[00523| In one embodiment, the compound of Formula II is a compound of Example No.
70, or a pharmaceutically acceptable sait or solvaté thereof.
[00524] In one embodiment, the compound of Formula II is a compound of Example No.
71, or a pharmaceutically acceptable sait or solvaté thereof.
[00525] In one embodiment, the compound of Formula II is a compound of Example No.
, or a pharmaceutically acceptable sait or solvaté thereof.
[00526] In one embodiment, the compound of Formula 11 is a compound of Example No.
73, or a pharmaceutically acceptable sait or solvaté thereof.
[00527] In one embodiment, the compound of Formula II is a compound of Example No.
74, or a pharmaceutically acceptable sait or solvaté thereof.
(00528] In one embodiment, the compound of Formula II is a compound of Example No. 75, or a pharmaceutically acceptable sait or solvaté thereof.
[00529] In one embodiment, the compound of Formula II is a compound of Example No.
76, or a pharmaceutically acceptable sait or solvaté thereof.
|0Û530] In one embodiment, the compound of Formula II is a compound of Example No.
77, or a pharmaceutically acceptable sait or solvaté thereof.
[00531] In one embodiment, the compound of Formula II is a compound of Example No.
79, or a pharmaceutically acceptable sait or solvaté thereof.
[00532] In one embodiment, the compound of Formula II is a compound of Example No.
80, or a pharmaceutically acceptable sait or solvaté thereof.
[00533] In one embodiment, the compound of Formula II is a compound of Example No.
81, or a pharmaceutically acceptable sait or solvaté thereof.
[00534] In one embodiment, the compound of Formula II is a compound of Example No.
84, or a pharmaceutically acceptable sait or solvaté thereof.
[00535] In one embodiment, the compound of Formula II is a compound of Example No.
86, or a pharmaceutically acceptable sait or solvaté thereof.
[00536] In one embodiment, the compound of Formula 11 is a compound of Example No.
, or a pharmaceutically acceptable sait or solvaté thereof.
[00537] In one embodiment, the compound of Formula II is a compound of Example No.
88, or a pharmaceutically acceptable sait or solvaté thereof.
[00538] In one embodiment, the compound of Formula II is a compound of Example No.
89, or a pharmaceutically acceptable sait or solvaté thereof.
[00539] In one embodiment, the compound of Formula II is a compound of Example No.
90, or a pharmaceutically acceptable sait or solvaté thereof.
[00540] In one embodiment, the compound of Formula II is a compound of Example No.
91, or a pharmaceutically acceptable sait or solvaté thereof.
[00541] In one embodiment, the compound of Formula II is a compound of Example No.
92, or a pharmaceutically acceptable sait or solvaté thereof.
[00542] In one embodiment, the compound of Formula II is a compound of Example No.
93, or a pharmaceutically acceptable sait or solvaté thereof.
[00543] In one embodiment, the compound of Formula II is a compound of Example No.
94, or a pharmaceutically acceptable sait or solvaté thereof.
[00544] In one embodiment, the compound of Formula II is a compound of Example No.
95, or a pharmaceutically acceptable sait or solvaté thereof.
[00545] In one embodiment, the compound of Formula II is a compound of Example No.
96, or a pharmaceutically acceptable sait or solvaté thereof.
|00546] In one embodiment, the compound of Formula II is a compound of Example No.
97, or a pharmaceutically acceptable sait or solvaté thereof.
[00547] In one embodiment, the compound of Formula II is a compound of Example No.
98, or a pharmaceutically acceptable sait or solvaté thereof.
[00548] In one embodiment, the compound of Formula II is a compound of Example No.
99, or a pharmaceutically acceptable sait or solvaté thereof.
[00549] In one embodiment, the compound of Formula II is a compound of Example No.
100, or a pharmaceutically acceptable sait or solvaté thereof.
[00550] In one embodiment, the compound of Formula II is a compound of Example No.
101, or a pharmaceutically acceptable sait or solvaté thereof.
[00551] In one embodiment, the compound of Formula II is a compound of Example No.
102, or a pharmaceutically acceptable sait or solvaté thereof.
[00552] In one embodiment, the compound of Formula II is a compound of Example No.
103, or a pharmaceutically acceptable sait or solvaté thereof.
[00553] In one embodiment, the compound of Formula II is a compound of Example No.
104, or a pharmaceutically acceptable sait or solvaté thereof.
[00554] In one embodiment, the compound of Formula II is a compound of Example No.
105, or a pharmaceutically acceptable sait or solvaté thereof.
[00555] In one embodiment, the compound of Formula II is a compound of Example No.
106, or a pharmaceutically acceptable sait or solvaté thereof.
[00556] In one embodiment, the compound of Formula II is a compound of Example No.
107, or a pharmaceutically acceptable sait or solvaté thereof.
[00557] In one embodiment, the compound of Formula II is a compound of Example No.
108, or a pharmaceutically acceptable sait or solvaté thereof.
[00558] In one embodiment, the compound of Fonnula II is a compound of Example No. 109, or a pharmaceutically acceptable sait or solvaté thereof.
[00559| In one embodiment, the compound of Formula II is a compound of Example No. 110, or a pharmaceutically acceptable sait or solvaté thereof.
[00560] In one embodiment, the compound of Formula II is a compound of Example No.
] 13, or a pharmaceutically acceptable sait or solvaté thereof.
[00561] In one embodiment, the compound of Formula II is a compound of Example No.
114 , or a pharmaceutically acceptable sait or solvaté thereof.
[00562] In one embodiment, the compound of Formula II is a compound of Example No.
115, or a pharmaceutically acceptable sait or solvaté thereof.
[00563] In one embodiment, the compound of Formula II is a compound of Example No.
116, or a pharmaceutically acceptable sait or solvaté thereof.
[00564] In one embodiment, the compound of Formula II is a compound of Example No.
117, or a pharmaceutically acceptable sait or solvaté thereof.
[00565] In one embodiment, the compound of Formula II is a compound of Example No.
118, or a pharmaceutically acceptable sait or solvaté thereof.
[00566] In one embodiment, the compound of Formula II is a compound of Example No.
119, or a pharmaceutically acceptable sait or solvaté thereof.
[00567] In one embodiment, the compound of Formula II is a compound of Example No.
120, or a pharmaceutically acceptable sait or solvaté thereof.
[00568] In one embodiment, the compound of Formula II is a compound of Example No.
121, or a pharmaceutically acceptable sait or solvaté thereof.
[00569] In one embodiment, the compound of Formula II is a compound of Example No.
122, or a pharmaceutically acceptable sait or solvaté thereof.
5 [00570] In one embodiment, the compound of Formula II is a compound of Example No.
128, or a pharmaceutically acceptable sait or solvaté thereof.
[00571] In one embodiment, the compound of Formula II is a compound of Example No.
142, or a pharmaceutically acceptable sait or solvaté thereof.
[00572] In one embodiment, the compound of Formula II is a compound of Example No.
144, or a pharmaceutically acceptable sait or solvaté thereof.
[00573] In one embodiment, the compound of Formula II is a compound of Example No. 145, or a pharmaceutically acceptable sait or solvaté thereof.
[00574] In one embodiment, the compound of Formula II is a compound of Example No. 146, or a pharmaceutically acceptable sait or solvaté thereof.
[00575] In one embodiment, the compound of Formula II is a compound of Example No.
147, or a pharmaceutically acceptable sait or solvaté thereof.
[00576] In one embodiment, the compound of Formula II is a compound of Example No.
148, or a pharmaceutically acceptable sait or solvaté thereof.
[00577] In one embodiment, the compound of Formula II is a compound of Example No.
150, or a pharmaceutically acceptable sait or solvaté thereof.
[00578] In one embodiment, the compound of Formula II is a compound of Example No.
151, or a pharmaceutically acceptable sait or solvaté thereof.
[00579| In one embodiment, the compound of Formula II is a compound of Example No.
152, or a pharmaceutically acceptable sait or solvaté thereof.
[00580] In one embodiment, the compound of Formula II is a compound of Example No.
153, or a pharmaceutically acceptable sait or solvaté thereof.
[Ü058Ï ] In one embodiment, the compound of Formula II is a compound of Example No.
154, or a pharmaceutically acceptable sait or solvaté thereof.
[00582] In one embodiment, the compound of Formula II is a compound of Example No.
155, or a pharmaceutically acceptable sait or solvaté thereof.
[00583] In one embodiment, the compound of Formula II îs a compound of Example No.
156, or a pharmaceutically acceptable sait or solvaté thereof.
[00584] In one embodiment, the compound of Formula II is a compound of Example No.
157, or a pharmaceutically acceptable sait or solvaté thereof.
|00585) In one embodiment, the compound of Formula II is a compound of Example No.
158, or a pharmaceutically acceptable sait or solvaté thereof.
[00586] In one embodiment, the compound of Formula II is a compound of Example No.
159, or a pharmaceutically acceptable sait or solvaté thereof.
[00587] In one embodiment, the compound of Formula II is a compound of Example No.
160, or a pharmaceutically acceptable sait or solvaté thereof.
[00588] In one embodiment, the compound of Formula II is a compound of Example No. 161, or a pharmaceutically acceptable sait or solvaté thereof.
[00589] In one embodiment, the compound of Formula II is a compound of Example No. 162, or a pharmaceutically acceptable sait or solvaté thereof.
[00590] In one embodiment, the compound of Formula II is a compound of Example No.
163, or a pharmaceutically acceptable sait or solvaté thereof.
[00591] In one embodiment, the compound of Formula II is a compound of Example No.
164, or a pharmaceutically acceptable sait or solvaté thereof.
[00592] In one embodiment, the compound of Formula II is a compound of Example No.
166, or a pharmaceutically acceptable sait or solvaté thereof.
[00593] In one embodiment, the compound of Formula II is a compound of Example No.
167, or a pharmaceutically acceptable sait or solvaté thereof.
[00594] In one embodiment, the compound of Formula II is a compound of Example No.
168, or a pharmaceutically acceptable sait or solvaté thereof.
[00595] In one embodiment, the compound of Formula II is a compound of Example No.
169, or a pharmaceutically acceptable sait or solvaté thereof.
[00596] In one embodiment, the compound of Formula II is a compound of Example No.
170, or a pharmaceutically acceptable sait or solvaté thereof.
[00597] In one embodiment, the compound of Formula II is a compound of Example No.
171, or a pharmaceutically acceptable sait or solvaté thereof.
[00598] In one embodiment, the compound of Formula II is a compound of Example No.
172, or a pharmaceutically acceptable sait or solvaté thereof.
[00599| In one embodiment, the compound of Formula II is a compound of Example No.
173, or a pharmaceutically acceptable sait or solvaté thereof.
5 [00600] In one embodiment, the compound of Formula II is a compound of Example No.
179, or a pharmaceutically acceptable sait or solvaté thereof.
[00601] In one embodiment, the compound of Formula II is a compound of Example No.
181, or a pharmaceutically acceptable sait or solvaté thereof.
[00602] In one embodiment, the compound of Formula II is a compound of Example No.
182, or a pharmaceutically acceptable sait or solvaté thereof.
[00603] In one embodiment, the compound of Formula II is a compound of Example No, 183, or a pharmaceutically acceptable sait or solvaté thereof.
[00604J In one embodiment, the compound of Formula II is a compound of Example No. 184, or a pharmaceutically acceptable sait or solvaté thereof.
[00605] In one embodiment, the compound of Formula II is a compound of Example No.
185, or a pharmaceutically acceptable sait or solvaté thereof.
[00606] In one embodiment, the compound of Formula II is a compound of Example No. 186, or a pharmaceutically acceptable sait or solvaté thereof.
[00607] In one embodiment, the compound of Formula II is a compound of Example No. 10 187, or a pharmaceutically acceptable sait or solvaté thereof.
[00608] In one embodiment, the compound of Formula II is a compound of Example No.
189, or a pharmaceutically acceptable sait or solvaté thereof.
[00609] In one embodiment, the compound of Formula II is a compound of Example No. 191, or a pharmaceutically acceptable sait or solvaté thereof.
[00610] In one embodiment, the compound of Formula II is a compound of Example No.
192, or a pharmaceutically acceptable sait or solvaté thereof.
[00611] In one embodiment, the compound of Formula II is a compound of Example No. 193, or a pharmaceutically acceptable sait or solvaté thereof.
[00612] In one embodiment, the compound of Formula II is a compound of Example No.
0 194, or a pharmaceutically acceptable sait or solvaté thereof.
[00613] In one embodiment, the compound of Formula II is a compound of Example No.
195, or a pharmaceutically acceptable sait or solvaté thereof.
[00614] In one embodiment, the compound of Formula II is a compound of Example No.
196, or a pharmaceutically acceptable sait or solvaté thereof.
In one embodiment, the compound of Formula II is a compound of Example No. 197, or a pharmaceutically acceptable sait or solvaté thereof.
[00615] The term pharmaceutically acceptable indïcates that the compound, or sait or composition thereof is compatible chemically and/or toxicologically with the other ingrédients comprising a formulation and/or the patient being treated therewith.
|00616] Compounds provided herein may also contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute such compounds. That is, an atom, in particular when mentioned in relation to a compound according to Formula I, comprises ail isotopes and isotopic mixtures of that atom, either naturel ly occurring or syntheticaily produced, either with naturel abundance or in an isotopically enriched form. For example, when hydrogen is mentioned, it is understood to refer to 'H, 2H, 3H or mixtures thereof; when carbon is mentioned, it is understood to refer to 1!C, 12C, l3C, 14C or mixtures thereof; when nitrogen is mentioned, it is understood to refer to 13N, 14N, ,5N or mixtures thereof; when oxygen is mentioned, it is understood to refer to l4O, I5O, 16O, 17O, 18O or mixtures thereof; and when fluoro is mentioned, it is understood to refer to 18F, 19F or mixtures thereof. The compounds provided herein therefore also comprise compounds with one or more isotopes of one or more atoms, and mixtures thereof, including radioactive compounds, wherein one or more non-radioactive atoms has been replaced by one of its radioactive enriched isotopes. Radiolabeled compounds are useful as additional anticancer agents, e.g., cancer therapeutic agents, research reagents, e.g., assay reagents, and diagnostic agents, e.g., in vivo imaging agents. AU isotopic variations of the compounds provided herein, whether radioactive or not, are intended to be encompassed within the scope of the présent invention.
[00617] For illustrative purposes, Schemes 1-24 show general methods for preparing the compounds provided herein as well as key intermediates. For a more detailed description of the indivîdual reaction steps, see the Examples section below. Those skilled in the art will appreciate that other synthetic routes may be used to synthesîze the inventive compounds. Although spécifie starting materials and reagents are depicted in the Schemes and discussed below, other starting materials and reagents can be easily substituted to provide a variety of dérivatives and/or reaction conditions. In addition, many of the compounds prepared by the methods described below can be further modified in lîght of this disclosure using conventional chemistry well known to those skilled in the art.
OCN-R8
DIEA
NaOMe
HCl
Scheme 1
[00618] Scheme 1 shows a general process for the préparation of compound 5 wherein R6 and R8 are as defîned for Formula 1, which is an intermediate useful for the préparation of compounds of Formula I wherein G is Ring A and Ring A is A-l, R7 is hydrogen and R3 is absent. [00619| Diethyl 2-(aminomethyiene)malonate may be reacted with a reagent having the formula OCN-R8 wherein Rs is as defîned for Formula I, to provide compound 2. Compound 2 may be treated with a strong base (e.g, sodium methoxide) to provide compound 3. Compound 3 may be reacted with a reagent having the formula R6-i and a base, such as K2CO3, wherein R6 is as defîned for Formula 1 to provide compound 4. Treatment of compound 4 with aqueous acid provides compound 5.
Re B(OH)2
Cu(OAc)2
HCl
Scheme 2
[00620] Scheme 2 shows a general process for the préparation of compound 7 wherein R6 is cyciopropyl, and R8 is as defined for Formula I, which is an intermédiare useful for the préparation of compounds of Formula I wherein G is Ring A and Ring A is A-l, R6 is cyciopropyl, 5 R7 is hydrogen and R3 is absent.
[00621] Compound 3, wherein R8 is as deflned for Formula I (prepared as in Scheme 1), may be treated with a reagent having the formula R6-B(OH)2, wherein R6 is cyciopropyl, in the presence of Cu(OAc)2 and 2,2'-bipyrtdine to provide compound 6. Treatment of compound 6 with aqueous acid provides compound 7.
Scheme 3
[00622] Scheme 3 shows a general process for the préparation of compound 12 wherein R6 is hetCyc2 and R8 is as defined for Formula I, which is an intermediate useful for the préparation of compounds of Formula 1 wherein G is Ring A and Ring A is A-l, R7 is hydrogen and R3 is absent.
[00623] Compound 3, wherein R8 is as defïned for Formula I (prepared as in Scheme 1 ), may be treated with reagent (8), wherein P1 is an amino protecting group (e.g., Boc) in the presence of a base (e.g., K.2CO3) to provide compound 9. The protecting group P1 of compound 9 may be removed to provide compound 10. Compound 10 may be reacted with a reagent having the formula RC(=O)H wherein R is CI-C6 alkyl, in the presence of a reducing agent (e.g., NaBHfOAch) to provide compound 11. Compound 11 may be treated with aqueous acid to provide compound 12.
H
Η2Ν'%β HCi
acid
[00624] Scheme 4 shows a general process for the préparation of compound 16, wherein R8 is as defined for Formula I, which is an intermediate useful for the préparation of compounds of 5 Formula I wherein G is Ring A and Ring A is A-2, R6 and R7 are hydrogen and R3 is absent.
[00625] Compound 13, wherein R8 is as defined for Formula I, may be treated with oxalaldéhyde in the presence of an acid to provide compound 14. Compound 14 may be treated with 2,2-dimethyl-l,3-dioxane-4,6-dione, in the presence of a carboxylic acid (e.g., acetic acid) and an amine base (e.g., piperidine) to provide compound 15. Compound 15 may be treated with 10 a base (e.g., sodium methoxide) to provide compound 16.
h2n-nhr8
HOAc
° HN'r.8
R8
acid
Piperidine
Scheme 5
[00626] Scheme 5 shows a general process for the préparation of compound 19, wherein R6 15 is hydrogen, C1-C6 alkyl, C1-C6 alkoxy, HOC1-C6 alkyl, C3-C6 cycloalkyl, (C3-C6 cycîoalkyl)Cl -C6 alkyl- or hetCyc2, and R8 is as defined for Formula I, which is an intermediate useful for the préparation of compounds of Formula I wherein G is Ring A and Ring A is A-2, R7 is hydrogen and R3 is absent.
[00627] Compound 17, wherein R6 is hydrogen, C1-C6 alkyl, C1-C6 alkoxy, P2OC1-C6 2 0 alkyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)C I-C6 alkyl- or hetCyc2, and wherein P2 is a hydroxy protecting group, may be reacted with a reagent having the formula FbN-NHR8 wherein R8 is as defined for Formula 1, to provide compound 18, as the minor product. Compound 18 may be reacted with 2,2-dimethyl-l,3-dioxane-4,6-dione in the presence of an acid (e.g., HOAc) and piperidine to provide compound 19, after which, if R6 is P2OC1-C6 alkyl, the hydroxyl protecting group is removed.
Br
O
O
Palladium catalyst
LiOH
$ Scheme6
[00628] Scheme 6 shows a general process for the préparation of compound 22, wherein R8 is as defined for Formula I, and R1 and R2 are independently H or C1-C3 alkyl, which is an intermediate useful for the préparation of compounds of Formula I wherein G is Ring A and Ring A is A-3, R6 is C1-C6 alkyl, R7 is hydrogen and R3 is absent.
100629] Compound 20, wherein R8 is as defined for Formula I, may be reacted with compound 21a, wherein R1 and R2 are independently FI or C1-C3 alkyl, in the presence of a palladium catalyst to provide compound 21. Compound 21 may be treated with lithium hydroxide to provide compound 22.
Re-I
Copper catalyst base quinolînol
acid
Scheme 7
[00630] Scheme 7 shows a general process for the préparation of compound 25, wherein R6 is C1-C6 alkoxy and R8 is as defined for Formula 1, which is an intermediate useful for the préparation of compounds of Formula 1 wherein G is Ring A and Ring A is A-3, R7 is hydrogen and R3 is absent.
|00631] Compound 23, wherein R6 is C1-C6 alkoxy, may be reacted with a reagent having the formula R8-!, wherein R8 is as defined for Formula I, in the presence of a copper catalyst, a base, and quinolinol, to provide compound 24. Treatment of compound 24 with aqueous acid provides compound 25.
O
28
Scheme 8
[00632] Scheme 8 shows a general process for the préparation of compound 28, wherein R6 is C1-C6 alkyl and R8 is as defined for Formula I, which is an intermediate useful for the préparation of compounds of Formula I wherein G is Ring A and Ring A is A-3, R7 is hydrogen, and R3 is absent.
[00633] Ethyl 3-chloro-3-oxopropanoate may be reacted with reagent R8-NH2, wherein R8 is as defined for Formula ï, to provide compound 26. Compound 26 may be reacted with reagent 27, wherein R6 is C1-C6 alkyl, in the presence of a base (e.g., sodium ethoxide), to provide compound 28.
31 32
Scheme 9
[00634] Scheme 9 shows a general process for the préparation of compound 32, wherein R8 is as defined for Formula I, which is an intennediate useful for the préparation of compounds of Formula I wherein G is Ring A and Ring A is A-3, R6 is methyl, R7 is hydrogen and R3 is absent.
[00635] Diethyl 2-(propan-2-ylidene)malonate may be reacted with a reagent having the formula R8-NH2 wherein R8 is as defined for Formula I, in the presence of imidazole at elevated températures (e.g., at about 200 °C), to provide compound 30. Compound 30 may be reacted with
,l-dimethoxy-N,N-dimethylmethanamine to provide compound 31. Treatment of compound 32 with lithium hydroxide provides compound 32,
34
Scheme 10
[00636] Scheme 10 shows a general process for the préparation of compound 34, wherein R8 is as defined for Formula I, which is an intermediate useful for the préparation of compounds of Formula I wherein G is Ring A and Ring A is A-3, R6 is CI-C6 alkyl, R7 is hydrogen, and R3 is absent.
[00637] Compound 34 may be prepared by treating compound 26 (prepared as in Scheme
8), wherein R8 is as defined for Formula 1, with reagent 27a wherein R6 is C1-C6 alkyl, in the presence of sodium ethoxide.
x oh x rS R6
RB BFjK
nu rv I η .,-o J nh , TT Tl copper catalyst II II on ο o ° ° base 35 36 LiOH X η y Ra O O 39 15 ------ ---- HO. AÂ 8 Palladium catalyst Π Π Tl Tl R oo 0 0 37 38 Scheme 11
[00638]
Scheme 11 shows a general process for the préparation of compound 38, wherein
R8 is Ar2, hetAr2, or C3-C6 cycloalkyl, and R6 is C3-C6 cycloalkyl, and compound 39, wherein R8 is as defined for Formula I and X is Cl or Br, which are intermediates useful for the préparation of compounds of Formula I wherein G is Ring A and Ring A is A-3, R7 is hydrogen and R3 is absent.
[00639] Compound 35, wherein X is CI or Br, may be reacted with a boronic acid having the formula (HOfrB-R8, wherein R8 is Ar2, hetAr2, or C3-C6 cycloalkyl, in the presence of a copper catalyst (e.g., copper(II) acetate) and a base (e.g., pyridine) to provide compound 36. Compound 36 may be treated with a compound having the formula R6-BFsK wherein R6 is C3-C6 cycloalkyl to provide compound 37. Treatment of compound 37 with lithium hydroxide provides compound 10 38. Compound 39 may be obtained by treating compound 36 with lithium hydroxide.
H H s (40}
R8
ο o
Schème 12
[00640] Scheme 12 shows a general process for the préparation of compound 43, wherein 15 R6 isCl-C6 alkyl, C1-C6 alkoxy, hydroxyCl-C6 alkyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)ClC6 alkyl- or hetCyc2, and Rs is as defined for Formula I, which is an intermediate useful for the préparation of compounds of Formula I wherein G is Ring A and Ring A is A-4, R7 is thioxo, and R3 is absent.
[00641] Diethyl 2-oxomalonate may be reacted with a compound having formula 40, 20 wherein R8 is as defined for Formula I, to provide compound 4]. Compound 41 may be treated with a reagent having the formula R6-!, wherein R6 is C1-C6 alkyl, C1-C6 alkoxy, P2O-C1-C6 alkyl, C3-C6 cycloalkyl, (C3-C6 cyc!oalkyl)Cl-C6 alkyl- or hetCyc2 and P2 is a hydroxyl protecting group, in the presence of a base (e.g., K2CO3), to provide compound 42. Treatment of compound 42 with aqueous acid provides compound 43, after which, if R6 is P2OC1-C6 alkyl, the 2 5 hydroxyl protecting group is removed.
acid h2o2
46
Scheme 13
[00642] Scheme 13 shows a general process for the préparation of compound 46, wherein R6 isCl-C6 alkyl, CI-C6 alkoxy, hydroxyCl-C6 alkyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)Cl5 C6 alkyl- or hetCyc2 and R8 is as defined for Formula I, which is an intermediate useful for the préparation of compounds of Formula I wherein G is Ring A and Ring A is A-4, R7 is oxo, and R3 is absent.
[00643] Compound 42, prepared according to Scheme 12, may be treated with hydrogen peroxide in the presence of an acid to provide compound 44. Compound 44 may be treated with 10 a compound having the formula R6-I wherein R6 is C1-C6 alkyl, C1-C6 alkoxy, P2O-C1-C6 alkyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)Cl-C6 alkyl- or hetCyc2 and P2 is a hydroxy protecting group, in the presence of a base (e.g., K2CO3) to provide compound 45. Treatment of compound 45 with aqueous acid provides compound 46, after which, if R6 is P2OC1-C6 alkyl, the hydroxyl protecting group is removed.
Scheme 14
[00644] Scheme 14 shows a general process for the préparation of compound 50, wherein R6 is C1-C6 alkyl, C1-C6 alkoxy, hydroxyC 1-C6 alkyl, C3-C6 cycloalkyl, (C3-C6 cycîoalkyl)Cl20 C6 alkyl- or hetCyc2 and Rs is Ar2, hetAr2 or cyclopropyl which is an intermediate useful for the préparation of compounds of Formula I wherein G is Ring A and Ring A is A-5, R7 is hydrogen and R3 is absent.
[00645] Methyl 5-bromo-4-hydroxynicotinate may be treated with a compound having the formula R6-! wherein R6 is C1-C6 alkyl, C1-C6 alkoxy, P2O-C1-C6 alkyl, C3-C6 cycloalkyl, (C3C6 cyc!oalkyl)Cl-C6 alkyl- or hetCyc2 and P2 is a hydroxy protecting group, in the presence of a base (e.g., CsaCOj) to provide compound 47. Compound 47 may be treated with boronic acid 48, wherein R8 is Ar2, hetAr2 or cyclopropyl, to provide compound 49. Treatment of compound 49 5 with aqueous acid provides compound 50, after which, if R6 is P2OC1-C6 alkyl, the hydroxyl protecting group is removed.
Ra‘NH2
KCN
NaOMe
52 hydrogénation
acid
Zn(CN)2
Palladium catalyst
O
POCI3
O
57 56 55
Scheme 15
[00646] Scheme 15 shows a general process for the préparation of compound 58, wherein
R8 îs as defined for Formula I, which is an intermediate useful for the préparation of compounds of Formula I wherein G is Ring A and Ring A is A-6, R6 and R7 are hydrogen, and R3 is absent. [00647] Compound 51, wherein R8 is as defined for Formula I, may be treated with formaldéhyde and potassium cyanide to provide compound 52. Compound 52 may be treated with oxalyl dichloride to provide compound 53. Compound 53 may be treated with sodium methoxide to provide compound 54. Compound 54 may be reduced under standard hydrogénation conditions (e.g., under a hydrogen atmosphère in the presence of a palladium catalyst such as palladium on carbon) to provide compound 55. Compound 55 may be converted to compound 56 upon treatment with phosphoryl chloride. Compound 56 may be treated with zinc cyanide in the presence of a palladium catalyst and a ligand (e.g. Pd^dbaj and dppf) to provide compound 57. The nitrile group
of compound 57 may be hydrolyzed upon treatment with aqueous acid to provide compound 58.
Rs-I
Cul quinolin-8-ol base
R3-l Base
basic hydrolysis
61
Scheme 16
[00648] Scheme 16 shows a general process for the préparation of compound 59, wherein
R8 is Ar2, hetAr, C3-C6 cycloalkyl or hetCyc3, and compound 61, wherein R3 is Cl -C6 alkyl, and R8 is Ar2, hetAr2, C3-C6 cycloalkyl or hetCyc3, which are intermediates useful for the préparation of compounds of Formula I wherein G is Ring A and Ring A is A-7, R7 is hydrogen, and R3 is methyl.
[00649] Ethyl 2-oxopiperidine-3-carboxylate may be treated with reagent R8-!, wherein R8 is Ar2, hetAr2, C3-C6 cycloalkyl or hetCyc3, in the presence of copper (I) iodide, quinlin-8-ol and a base (e.g., CsCO3) to provide compound 58. Compound 58 may be hydrolyzed under basic conditions (e.g., LiOH) to provide compound 59.
[00650] Alternatively, compound 58 may be treated with a compound having the formula 15 R3-! wherein R3 is C1-C6 alkyl to provide compound 60. Compound 60 may be hydrolyzed under basic conditions (e.g., LiOH) to provide compound 61.
R®-l
Cul quinolin-8-ol base
LiOH
Scheme 17
[00651] Scheme 17 shows a general process for the préparation of compound 65, wherein R8 is Ar2, hetAr2, C3-C6 cycloalkyl or hetCyc3, which is an intermediate useful for the préparation 5 of compounds of Formula 1 wherein G is Ring A and Ring A is A-7, R6 and R7 together form a cyclopropyl ring, and R3 is hydrogen.
[00652] (l-Cyanocyclopropyl)methyl methanesulfonate may be treated with dîethyl malonate in the presence of sodium hydride to provide compound 62. Compound 62 may undergo an intramolecular cyclization in the presence of hydrogen and a catalytic amount of platinum(IV) 10 oxide to provide compound 63. Compound 63 may be treated with a compound having the formula R8-!, wherein R8 is Ar2, hetAr2, C3-C6 cycloalkyl or hetCyc3, in the presence of copper (1) iodide, quinlin-8-ol and a base (e.g, CS2CO3) to provide compound 64. Compound 64 may be hydrolyzed under basic conditions (e.g, LiOH) to provide compound 65.
H HCl (66)
2?
f3c o Rs (66)
Na OH
Schéma 18
[006531 Scheme 18 shows a general process for the préparation of compound 70, wherein R8 is as defined for Formula I, which is an intermediate use fui for the préparation of compounds 5 of Formula I wherein G is Ring A and Ring A is A-8, R7 is hydrogen, and R3 is absent.
[00654] Diethyl 2-(ethoxymethylene)malonate may be reacted with a compound 66, wherein Rs is as defined for Formula I, to provide compound 67. Compound 67 may be reacted with compound 68, wherein R6 is C1-C6 alkyl, to provide compound 69. Treatment of compound 69 with sodium hydroxide provides compound 70.
Scheme 19
[00655] Scheme 19 shows a general process for the préparation of compound 72, wherein R8 is as defined for Formula I, which is an intermediate useful for the préparation of compounds 15 of Formula I wherein G is Ring B and Ring B is B-l,
[00656] Ethyl (E)-2-cyano-3-ethoxyacrylate may be reacted with cyclohexane-1,3-dione in the presence of potassium tert-butoxide to provide compound 71, Compound 71 may be reacted with the reagent Rs-I, wherein R8 is as defined for Formula I, to provide compound 72.
η
Schéma 20
[00657] Scheme 20 shows a general process for the préparation of compound 79, wherein R9 is as defined for Fonnula I, and compound 80, wherein R9 and R2 are as defined for Formula 5 I, which are intermediates usefui for the préparation of compounds of Formula I wherein X1 is
CH.
[00658] Compound 73, wherein P3 is an amino protecting group (e.g., para-methoxybenzyl;
PMB), may be reacted with compound 74, wherein R9 is as defined for Formula I, in the presence of a base to provide compound 75. The amino protecting group of compound 75 may be removed 10 under standard conditions (e.g., TFA) to provide compound 76. Compound 76 may be treated with h in the presence of a base (e.g., KOH) to provide compound 77. The amino group of compound 77 may be protected under standard conditions to provide compound 78, wherein P3 is an amino protecting group (e.g., para-methoxybenzyl; PMB). The nitro group of compound 78 may be reduced under standard conditions (e.g., tin(II) chloride) to provide compound 79. 15 Compound 79 may be treated with a compound having the formula H2NR2, wherein R2 is as defined for Formula I, tn the presence of Cul, a ligand (e.g., IH-pyrrole-2-carboxylic acid), and a base (e.g., potassium carbonate) to provide compound 80.
ci
b base
amino protection
R2 NHj
Cul, base
Palladium catalyst
TMS. „TMS N
Li
Schéma 21
[00659] Scheme 21 shows a general process for the préparation of compound 86, wherein 5 R2 is hydrogen, C1-C6 alkyl, C1-C6 fluoroalkyl, (di-Cl-C6 alkoxy)C2-C6 alkyl-, (C1-C6 alkoxy)Cl-C6 alkyl-, Cyc1, Cyc2, (hetCyc'jCl-Cô alkyl-, (Ar'jCl-CÔ alkyl-, (hetAr')Cl-C6 alkyl-, or (HOSOa)Cl-C6 alkyl-, which is an intermediate useful for preparing compounds of Formula I wherein X1 is N, R9 is hydrogen, and R2 is hydrogen, C1-C6 alkyl, C1-C6 fluoroalkyl, (di-Cl-C6 alkoxy)C2-C6 alkyl-, (C1-C6 alkoxy)Cl-C6 alkyl-, Cyc1, Cyc2, (hetCyc')Cl-C6 alkyΙΙΟ , (Ar')C 1-C6 alkyl-, (hetAr')C 1-C6 alkyl-, or (HOSO3)C I-C6 alkyl-,
[00660] Compound 81 may be treated with h in the presence of a base (e.g., KOH) to provide compound 82. The amino group of compound 82 may be protected under standard conditions (e.g., by treatment with l-(chloromethyl)-4-methoxybenzene in the presence of base, e.g., K2CO3) to provide compound 83 wherein P3 is an amino protecting group (e.g., PMB). 15 Compound 83 may be treated with 2-chloro-5-hydroxypyridine in the presence of a base (e.g.,
CS2CO3) to provide compound 84. Compound 84 may be treated with reagent H2NR2, wherein R2 is hydrogen. Cl-C6alkyl, C1-C6 fluoroalkyl, (di-Cl-C6 a!koxy)C2-C6 alkyl-, (C I-C6 a!koxy)ClC6 alkyl-, Cyc1, Cyc2, (hetCyc')C1-C6 alkyl-, (Ar')Cl-C6 alkyl-, (hetAr'jCl-CÔ alkyl-, or (HOSO3)C1-C6 alkyl-,, in the presence ofCuI, ligand (e.g., lH-pyrrole-2-carboxylic acid), and a base (e.g., potassium carbonate) to provide compound 85. Compound 85 may be treated with lithium bis(trimethylsilyl)amide in the presence of a palladium catalyst (e.g., Pd2dba3) and a ligand (e.g., X-Phos) to provide compound 86.
Cul, base
hydroxyl deprotectjon
Schéma 22
[00661] Scheme 22 shows a general process for the préparation of compound 91, wherein
R2 is hydroxyCl-C6 alkyl and P3 is an amino protecting group, which is an intennediate useful for preparîng compounds of Formula 1 wherein X1 is N and R9 is hydrogen.
[00662] Compound 87 (prepared as in Scheme 21), wherein P3 is an amino protecting group, may be treated with a compound having the formula H2NR2, wherein R2 is hydroxyCl-C6 alkyl, 15 in the presence of Cul, ligand (e.g., lH-pyrrole-2-carboxylic acid), and a base (e.g., potassium carbonate) to provide compound 88. The hydroxyl moiety of the R2 group of compound 88 may be protected under standard conditions (e.g., by treatment with tert-butyldimethylsilyl chloride) to provide compound 89 wherein R2a is hydroxyCl-C6 alkyl wherein the hydroxyl moiety is protected. Compound 89 may be treated with tert-butyl carbamate in the presence of a palladium catalyst (e.g., Pd2dba3) and a ligand (e.g., X-Phos) to provide compound 90. Removal of the hydroxyl protecting group under standard conditions provides compound 91.
deprotection
Scheme 23
[00663] Scheme 23 shows a general process for the préparation of compound 96, which is a compound of Formula I wherein R1, R2, X1, R9 and G are as defined for Formula 1.
[00664] Compound 92, wherein R9 and X1 are as defined for Formula I and P3 is an amino protecting group, may be treated with compound 93, wherein G is as defined for Formula I, in the presence of coupling reagents (e.g., in the presence of HATU or EDCI/HOBt and diisoproylethyl amine) to provide compound 94. Compound 94 may be treated with reagent R'R2NH, wherein R1 and R2 are as defined for Formula 1 and wherein if the R2 moiety conta ins a hydroxyl group 15 then the hydroxyl group is optionally protected with a hydroxyl protecting group, in the presence of copper(l) iodide and either in the presence of a base (e.g., K2CO3) and 1 H-pyrrole-2-carboxylic acid, or in the presence of a ligand (e.g,, NI,N2-dimethylethane-l,2-diamine) and a base (e.g., K3PO4) to provide compound 95. Removal of the amino protecting group and the hydroxyl group, if présent, under standard conditions (e.g., trifluoroacetic acid) provides compound 96. In embodiments wherein G is B-l (e.g., as in Scheme 19), a compound 95 wherein G is B-2 is formed 5 as an oxidative by-product during the reaction step converting compound 94 to compound 95.
Scheme 24
[00665] Scheme 24 shows an alternative general process for the préparation of compound 10 96, which is a compound of Formula I wherein R1, R2, X1, R9 and G are as deflned for Formula I.
[00666] Compound 92, wherein R9 and X1 are as defined for Formula I and P3 is an amino protecting group, may be treated with a reagent having the formula R^NH, wherein R1 and R2 are as defined for Formula I and wherein if the R2 moîety contains a hydroxyl group then the hydroxyl group is optionally protected with a hydroxyl protecting group, in the presence of copper(I) iodide and either in the presence of a base (e.g., K2CO3) and lH-pyrrole-2-carboxylic acid, or in the presence of a ligand (e.g., Nl,N2-dimethylethane-l,2-diamine) and a base (e.g., K3PO4) to provide compound 97. Compound 97 may be treated with compound 93, wherein G is as defined for Formula I, in the presence of coupling reagents (e.g., in the presence of HA TU or EDCI/HOBt and diisopropylethyl amine) to provide compound 95. Removal of the amino protecting group and the hydroxyl group, if présent, under standard conditions (e.g., trifluoroacetic acid) provides compound 96.
[00667] The term amino protecting group as used herein refers to a dérivative of the groups commonly employed to block or protect an amino group while reactions are carried out on other functional groups on the compound. Examples of suitable protecting groups for use in any of the processes described herein include carbamates, amides, alkyl and aryl groups, imines, as well as many N-heteroatom dérivatives which can be removed to regenerate the desired amine group. Non-limiting examples of amino protecting groups are para-methoxybenzyl (PMB), tbutyloxycarbonyt (“Boc”), benzyloxycarbonyl (“CBz”) and 9-fluorenylmethyleneoxycarbony! (“Fmoc”). Further examples of these groups, and other protecting groups, are found in T. W. Greene, et al., Greene’s Protective Groups in Organic Synthesis. New York: Wiley Interscience, 2006.
[00668] Flydroxyl groups may be protected with any convenient hydroxyl protecting group, for example as described in T. W. Greene, et al., Greene’s Protective Groups in Organic Synthesis. New York: Wiley Interscience, 2006. Examples include benzyl, trityl, silyl ethers, and the like.
[00669] In one embodiment, provided herein is a process for preparing a compound of Formula I, comprising:
[00670] (a) reactîng a compound having the formula
[006711 wherein X1, G, and R9 are as defined for Formula I and PJ is an amino protecting group, with a reagent having the formula R*R2NH, wherein R1 and R2 are as defined for Formula
I and wherein if the R2 moiety contains a hydroxyl group then the hydroxyl group is optionally protected with a hydroxyl protecting group, in the presence of copper(I) iodide and either in the presence of a base and lH-pyrrole-2-carboxylic acid, or in the presence of a ligand and a base, followed by removal ofthe amîno protecting group and removal of the hydroxyl group, if présent; or
[00672] (b) reacting a compound having the fonnula:
[00673] wherein R9 and X1 are as defined for Formula I, P3 is an amino protecting group, and R1 and R2 are as defîned for Formula I, wherein if the R2 moiety contains a hydroxyl group then the hydroxyl group is optionally protected with a hydroxyl protecting group, with a reagent having the formula
O HO^G
[00674| wherein G is as defîned for Fonnula I, in the presence of coupling reagents, followed by removal of the amino protecting group and removal of the hydroxyl group, if présent; and
[00675] optionally forming a pharmaceutically acceptable sait thereof, |00676] Compounds of Formula I or phannaceutically acceptable salts thereof can modulate or inhibit the activity of one or more TAM kinases. The ability of compounds of Formula I to act as inhibitors of one or more TAM kinases may be demonstrated by the assays described in Examples A, B and C. ICso values are shown in the tables in the Examples.
(00677] Compounds of Formula I or pharmaceutically acceptable salts thereof can modulate or inhibit the activity of c-Met kinase. The ability of compounds of Formula I to act as inhibitors of wild type and certain mutant c-Met kinases may be demonstrated by the assay described in Example D. IC50 values are shown in Table 9.
[00678] As used herein, the term a TAM kinase refers to one, two or ail three of the TAM receptor tyrosine kinases, i.e., TYRO3, AXL and MER.
[00679] As used herein, the term “a TAM kinase inhibitor” refers to any compound exhibiting inhibition activity against one, two or ail three of the TAM receptor kinases, i.e., the compounds exhibit inhibitory activity against AXL and/or MER and/or TYRO3.
[00680] As used herein, the term “a c-Met kinase inhibitor” refers to any compound exhibiting inhibitory activity against wild type or certain mutant c-Met kinases. In one embodiment, the term “a c-met kinase inhibitor” refers to any compound exhibiting inhibitory activity against wild type c-Met kinase or a mutant c-Met kinase selected from Del 14, DI228H, DI228N, F1200I, Ll 195V, Y1230C, Y1230H and Y1230S.
[00681] In some embodiments, compounds of Formula I or pharmaceutically acceptable salts thereof hâve inhibitory activity against AXL. In some embodiments, compounds of Formula 1 or pharmaceutically acceptable salts thereof hâve inhibitory activity against MER. In some embodiments, a compound of Fonnula I bas inhibitory activity against AXL and MER. In some embodiments, compounds of Formula I or pharmaceutically acceptable salts thereof hâve inhibitory activity against AXL, MER and TYRO3. In some embodiments, a compound of Formula I or pharmaceutically acceptable salts thereof has inhibitory activity against c-Met kinase. In some embodiments, a compound of Formula 1 or pharmaceutically acceptable salts thereof has inhibitory activity against one or more receptor tyrosine kinases selected from AXL, MER, TYRO3, and c-Met. In some embodiments, a compound of Formula I or pharmaceutically acceptable salts thereof has inhibitory activity against a c-Met kinase that does not include amino acids encoded by exon 14. In some embodiments, a compound of Formula 1 or pharmaceutically acceptable salts thereof has inhibitory activity against a mutated c-Met (e.g., any of the examples of mutated c-Met proteins described herein or known in the art) (e.g., a mutation in c-Met that causes résistance to a Type I c-Met inhibitor).
[00682] In one embodiment, compounds of Formula I or pharmaceutically acceptable salts thereof exhibit inhibition activity (IC50) against a TAM kinase and/or c-Met of less than about 1000 nM, less than about 500 nM, less than about 200 nM, less than about 100 nM, less than about 50 nM, less than about 25 nM, less than about 10 nM, or less than about I nM as measured in an assay as described herein. In some embodiments, compounds of Formula I or pharmaceutically acceptable salts thereof exhibit inhibition activity (IC50) against a TAM kinase and/or c-Met of less than about 25 nM, less than about 10 nM, less than about 5 nM, or less than about 1 nM as measured in an assay as provided herein.
[00683] In one embodiment, exemplary compounds of Formula 1 or pharmaceutically acceptable salts thereof exhibit inhibition activity (IC50) against AXL of less than about 50 nM, less than about 25 nM, less than about 10 nM, or less than about l nM as measured in an assay as 5 described herein.
[00684] In one embodiment, compounds of Formula I or pharmaceutically acceptable salts thereof exhibit inhibition activity (IC50) against MER of less than about less than about 200 nM, less than about 100 nM, less than about 50 nM, less than about 25 nM, less than about 10 nM, or less than about 1 nM as measured in an assay as described herein. In some embodiments, 10 compounds of Formula 1 or pharmaceutically acceptable salts thereof exhibit inhibition activity (IC50) against MER of less than about 25 nM, less than about 10 nM, less than about 5 nM, or less than about 1 nM as measured in an assay as provided herein.
[00685] In one embodiment, compounds of Formula 1 or pharmaceutically acceptable salts thereof exhibit inhibition activity (IC50) against TYRO3 of less than about 1000 nM, less than 15 about 750 nM, less than about 500 nM, less than about 250 nM, less than about 200 nM, less than about 100 nM, less than about 50 nM, less than about 25 nM, less than about 10 nM, or less than about 1 nM as measured in an assay as described herein.
[00686] In one embodiment, compounds of Formula I or pharmaceutically acceptable salts thereof exhibit inhibition activity (IC50) against c-Metof less than about 1000 nM, less than about 2 0 750 nM, less than about 500 nM, less than about 250 nM, less than about 200 nM, less than about
100 nM, less than about 50 nM, less than about 25 nM, less than about 10 nM, or less than about 1 nM as measured in an assay as described herein.
[00687] In some embodiments, compounds of Formula 1 or pharmaceutically acceptable salts thereof inhibit ail of three of the TAM kinases (i.e., AXL, MER and TYRO3) within about 25 a 5-fold différence.
[00688] In some embodiments, compounds of Formula 1 or pharmaceutically acceptable salts thereof are sélective for AXL over MER. In some embodiments, a compound of Formula I or a pharmaceutically acceptable sait thereof, exhibits at least a 2-fo!d selectivity; at least a 3-fold selectivity; at least a 4-fold selectivity; at least a 5-fold selectivity; at least a 6-fold selectivity; at 30 least a 7-foid selectivity; at least a 8-fold selectivity; at least a 9-fold selectivity; at least a 10-fold selectivity; at least a 11-fold selectivity; at least a 12-fold selectivity; at least a 13-fold selectivity;
at least a I4-fold selectivity; at least a 15-fold selectivity; at least a 20-fold selectivity; at least a 25-fold selectivity; at least a 30-fold selectivity; at least a 35-fold selectivity; at least a 40-fold selectivity; at least a 45-fold selectivity; at least a 50-fold selectivity; or at least a 55-fold selectivity, for AXL over MER. In some embodiments, selectivity for AXL and MER is measured 5 in an enzyme assay (e.g., an enzyme assay as provided herein.
[00689] In some embodiments, a compound of Formula I or a pharmaceutically acceptable sait thereof, exhibits at least a 5-fold selectivity; at least a 10-fold selectivity; at least a 15-fold selectivity; at least a 20-fold selectivity; at least a 25-fold selectivity; at least a 30-fold selectivity; at least a 35-fold selectivity, at least a 40-fold selectivity; at least a 50-fold selectivity; at least a 10 60-fold selectivity; at least a 70-fold selectivity; at least a 80-fold selectivity; at least a 90-fold selectivity; at least a 100-fold selectivity; at least a 125-fold selectivity; at least a 150-fold selectivity; or at least a 200-fold selectivity, for AXL over TYRO3. In some embodiments, selectivity for AXL and TYRO3 is measured in an enzyme assay (e.g., an enzyme assay as provided herein).
[00690] In some embodiments, compounds of Formula I or a pharmaceutically acceptable sait thereof exhibit at least a 5-fold selectivity; at least a 10-fold selectivity; at least a 15-fold selectivity; at least a 20-fold selectivity; at least a 25-fold selectivity; at least a 30-fold selectivity; at least a 35-fold selectivity; or at least a 40-fold selectivity; for MER over TYRO3. In some embodiments, selectivity for MER and TYRO3 is measured in an enzyme assay (e.g., an enzyme 2 0 assay as provided herein.
[00691] In some embodiments, provided herein is a method for inhibiting AXL kinase, which comprises contacting the AXL kinase with compound of Formula I, or a pharmaceutically acceptable sait thereof.
[00692] In some embodiments, provided herein is a method for inhibiting MER kinase, 25 which comprises contacting the MER kinase with compound of Formula I, or a pharmaceutically acceptable sait thereof.
[00693] in some embodiments, provided herein is a method for inhibiting TYRO3 kinase, which comprises contacting the TYRO3 kinase with compound of Formula I, or a pharmaceutically acceptable sait thereof.
[00694] In some embodiments, provided herein is a method for inhibiting c-Met kinase (e.g., any of the exemplary c-Met kinases described herein), which comprises contacting the c-Met kinase with compound of Formula I, or a pharmaceutically acceptable sait thereof.
[00695] Compounds of Formula 1 or pharmaceutically acceptable salts thereof are useful in the treatment of varions diseases associated with increased (e.g., at least 1%, at least 2%, at least 4%, at least 6%, at least 8%, at least 10%, at least 12%, at least 14%, at least 16%, at least 18%, at least 5 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 100%, at least 110%, at least 120%, at least 130%, at least 140%, at least 150%, at least 160%, at least 170%, at least 180%, at least 190%, at least 200%, at least 210%, at least 220%, at least 230%, at least 240%, at least 250%, at least 260%, at least 270%, at least 280%, at 10 least 290%, or at least 300%) expression, level, and/or activity of one or more of the TAM kinases and/or c-Met kinase (e.g., in a cancer cell or in an immune cell) (e.g., as compared to a control, e.g., a non-cancerous tissue or cell, or a corresponding tissue or cell from a control subject that does not hâve cancer). In one embodiment, compounds of Formula I or pharmaceutically acceptable salts thereof are useful in treating or preventing proliférative disorders such as cancers. 15 In one embodiment, tumors with an activating mutation (e.g., a point mutation or a chromosomal translocation) in a gene encoding a receptor tyrosine kinase and/or upregulation of the expression of a receptor tyrosine kinase (e.g., any of the TAM kinases or c-Met kinase described herein) may be particularly sensitive to compounds of Formula 1. In one embodiment, tumors with a mutation in a MET gene that results in exon 14 skipping during mRNA splicing are sensitive to compounds 20 of Formula I. In one embodiment, tumors having a mutation in a MET gene that results in expression of a c-Met protein having résistance to a Type I c-Met inhibitor are sensitive to compounds of Formula 1.
[00696] As used herein, terrns treat or treatment refer to therapeutic or palliative measures. Bénéficiai or desired clinical results include, but are not limited to, alîeviation, in whoie 25 or in part, of symptoms associated with a disease or disorder or condition, diminishment of the extent of disease, stabilîzed (i.e., not worsening) State of disease, delay or slowing of disease progression, amelioration or palliation of the disease State (e.g., one or more symptoms of the disease), and remission (whether partial or total), whether détectable or undetectable. Treatment can also mean prolonging survival as compared to expected survival if not receiving treatment.
[00697] As used herein, the terms subject, individual, or patient, are used interchangeably, refers to any animal, including mammals such as mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, primates, and humans. In some embodiments, the patient is a human. In some embodiments, the subject has experienced and/or exhibited at least one symptom of the disease or disorder to be treated and/or prevented. In some embodiments, the subject has been identifîed or diagnosed as having a TAM-associated disease or disorder (e.g., a 5 TAM-associated cancer) and/or has been identifîed or diagnosed as having a c-Met-associated disease or disorder (e.g., a c-Met-associated cancer) (e.g., as determined using a regulatory agencyapproved, e.g., FDA-approved, assay or kit). In some embodiments, the subject has been identifîed or diagnosed as having a cancer associated with one or more TAM kinases and/or c-Met kinase (e.g., a TAK-associated cancer) (e.g., as determined using a regulatory agency-approved, e.g., 10 FDA-approved, assay or kit). In some embodiments, the subject has a tumor that is associated with one or more TAM kinases and/or c-Met kinase (e.g., an increase in the expression, level, and/or activity of one or more TAM kinases and/or c-Met kinase in a cell (e.g., a cancer celI or an immune cell) as compared to a control, e.g., a non-cancerous tissue or a corresponding tissue from a control subject that does not hâve cancer) (e.g., as determined using a regulatory agency15 approved assay or kit). In some embodiments, the subject is suspected of having a TAMassociated cancer and/or a c-Met-associated cancer. In some embodiments, the subject has a ciinical record indicating that the subject has a tumor is associated with one or more TAM kinases (e.g-, a TAM-associated cancer) and/or c-Met kinase (and optionally the ciinical record indicates that the subject should be treated with any of the compositions provided herein). In some 20 embodiments, the patient is a pédiatrie patient.
[00698] The terni “pédiatrie patient” as used herein refers to a patient under the âge of 21 years at the time of diagnosis or treatment. The terni “pédiatrie” can be further be divided into various subpopulations including: neonates (from birth through the First month of life); infants (1 month up to two years of âge); children (two years of âge up to 12 years of âge); and adolescents 25 (12 years of âge through 21 years of âge (up to, but not including, the twenty-second birthday)).
Berhman RE, Kliegman R, Arvin AM, Nelson WE. Nelson Textbook of Pediatrics, 15th Ed. Philadelphia: W.B. Saunders Company, 1996; Rudolph AM, et al. Rudolph ’s Pediatrics, 21 st Ed. New York: McGraw-Hill, 2002; and Avery MD, First LR. Pédiatrie Medicine, 2nd Ed. Baltimore: Williams & Wilkins; 1994. In some embodiments, a pédiatrie patient is from birth through the 30 First 28 days of life, from 29 days of âge to less than two years of âge, from two years of âge to less than 12 years of âge, or 12 years of âge through 21 years of âge (up to, but not including, the twenty-second birthday). In some embodiments, a pédiatrie patient is from birth through the first 28 days of life, from 29 days of âge to less than 1 year of âge, from one month of âge to less than four months of âge, from three months of âge to less than seven months of âge, from six months of âge to less than 1 year of âge, from 1 year of âge to less than 2 years of âge, from 2 years of âge 5 to less than 3 years of âge, from 2 years of âge to less than seven years of âge, from 3 years of âge to less than 5 years of âge, from 5 years of âge to less than 10 years of âge, from 6 years of âge to less than 13 years of âge, from 10 years of âge to less than 15 years of âge, or from 15 years of âge to less than 22 years of âge.
[00699] The phrase therapeutically effective amount means an amount of compound that, 10 when administered to a patient in need of such treatment, is sufficient to (i) treat a TAM kinaseassociated disease or dîsorder (e.g., a TAM-associated cancer) and/or a c-Met kinase-associated disease or disorder (e.g., a MET-associated cancer), (ii) attenuate, ameliorate, or eliminate one or more symptoms of the particular disease, condition, or disorder, or (iii) delay the onset of one or more symptoms of the particular disease, condition, or disorder described herein. The amount of 15 a compound of Formula I that will correspond to such an amount will vary depending upon factors such as the particular compound, disease condition and its severity, the identity (e.g., weight) of the patient in need of treatment, but can nevertheiess be routinely determined by one skilled in the art.
[00700] The term regulatory agency refers to a country's agency for the approval of the 20 medical use of pharmaceutical agents with the country. For example, a non-limiting example of a regulatory agency is the U.S. Food and Drug Administration (FDA).
[00701] The term TAM-associated disease or disorder as used herein refers to diseases or disorders associated with or having increased expression and/or activity of one or more of the TAM kinases in a cell (e.g., a cancer cell or an immune cell) (e.g., as compared to a control, e.g., 2 5 a non-cancerous tissue or cell, or a corresponding tissue or cell from a control subject that does not hâve cancer) and/or where activation of a TAM kinase expressed on non-cancer cells contributes to disease. Non-limiting examples of a TAM-associated disease or disorder include, for example, cancer (a TAM-associated cancer), e.g., any of the cancers described herein. In one embodiment, the disease is a cancer that overexpresses one or more TAM kinases after treatment 30 with at least one additionaî anticancer agent (e.g., one or more of any of the additionaî anticancer agents described herein), e.g., a kinase-targeted therapeutic agent and/or a chemotherapeutic agent as described herein). In one embodiment, the disease is associated with signaling through one or more TAM kinases expressed in cells of the immune System (e.g., immune cells selected from the group of tumor-associated macrophages, natural killer (NK) cells, and subsets of tumor associated dendritic cells), wherein the expression of one or more TAM kinases in the immune cells may limit 5 the ability of the patient’s immune System to make an effective anti-tumor response.
[00702] The terni “TAM-associated cancer” as used herein refers to cancers associated with or having increased expression and/or activîty of one or more of the TAM kinases in a cancer cell or an immune cell (e.g., as compared to a control, e.g., a non-cancerous tissue or cell, or a corresponding tissue or cell from a control subject that does not hâve cancer). Non-limiting 10 examples of a TAM-associated cancer are described herein. In some embodiments, the TAMassociated cancer is a cancer having a chromosomal translocation that results in the expression of a TMEM87B-MERTK fusion protein (e.g., amino acids 1-55 of TMEM87B and amino acids 4331000 of MERTK) or a AXL-MBIP fusion protein. A description of an exemplary chromosomal translocation that results in the expression of a TMEM87B-MERTK fusion protein is provided in 15 Shaveretal, (Cancer Res. 76(16):4850-4860, 2016). A description ofan exemplary chromosomal translocation that results in the expression of an AXL-MBIP fusion protein is provided in Seo et al. (Genome Res. 22:2109-2119, 2012). Chromosomal trans location s or the resulting expression of TMEM87B-MERTK or AXL-MBIP fusion proteins can be detected using In Situ Hybridization (e.g., Fluorescent In Situ Hybridization (FISH)). Chromosomal translocations that resuit in the 20 expression of TMEM87B-MERTK or AXL-MBIP can be detected by sequencing DNA from a sample obtained from the subject (e.g., blood, plasma, urine, cerebrospînal fluid, saliva, sputum, bronchoalveolar lavage, bile, lymphatic fluid, cyst fluid, stool ascites, or a tumor biopsy obtained from the subject). Exemplary methods that can be used to sequence DNA are known in the art and include, e.g., next-generation sequencing (NGS), traditional PCR, digital PCR, and microarray 25 analysis. Additional methods that can be used to detect chromosomal translocations that resuit in the expression of TMEM87B-MERTK or AXL-MBIP fusion proteins, or the expression of TMEM87B-MERTK or AXL-MBIP fusion proteins, are known in the art.
[00703] The term c-Met-associated disease or disorder as used herein refers to diseases or disorders associated with or having increased expression, level, and/or activity of c-Met kinase in 30 a cell (e.g., a cancer cell or an immune cell) (e.g., as compared to a control, e.g., a non-cancerous tissue or cell, or a corresponding tissue or cell from a control subject that does not hâve cancer) and/or where activation of c-Met kinase expressed in non-cancer cells contributes to disease. Nonlimiting examples of a c-MET-associated disease or dîsorder include, for example, cancer (a cMet-associated cancer), e.g, any of the cancers described herein. In one embodiment, the disease is a cancer that overexpresses c-Met kinase after treatment with at least one additional anticancer 5 agent (e.g, one or more of any of the additional anticancer agents described herein), e.g, a kinasetargeted therapeutic agent and/or a chemotherapeutic agent as described herein). In some embodiments, the disease is a cancer that has a higher protein level of c-Met kinase (e.g, due to mutation in a MET gene that results in decreased protéasome dégradation of c-MET kinase in a mammalian cell). In some embodiments, the disease is a cancer that has a higher level of c-Met 10 kinase activity due to an actîvating mutation in a c-Met gene (e.g, any of the activating mutations in a c-Met gene described herein) or an increase in the expression of a c-Met kinase in a mammalian cell. In some embodiments, the disease is a cancer that expresses a c-Met kinase that is résistant (e.g, to at least some extent as compared to a wildtype c-Met kinase) to a Type I c-Met inhibitor.
[00704] Receptor tyrosine kinases (RTKs) are cell surface proteins that transmit signais from the extracellular environment to the cell cytoplasm and nucléus to regulate cellular events such as survival, growth, prolifération, différentiation, adhesion, and migration. AU RTKs contain an extracellular ligand binding domain and a cytoplasmic protein tyrosine kinase domain. Ligand binding leads to the dimerîzation of RTKs, which triggers the activation of the cytoplasmic kinase and initiâtes downstream signal transduction pathways. RTKs can be classified into distinct subfamilies based on theîr sequence similarity.
[00705] The TAM receptor tyrosine kinases (TYRO3, AXL (also known as UFO) and MER) is an emerging class of innate immune checkpoints that participate in key steps of antitumoral immunity (Akalu, T, et al, Immunological Reviews 2017; 276:165-177). TAM kinases 25 are characterized by an extracellular ligand binding domain consisting of two immunoglobulinlike domains and two fibronectin type III domains. Two ligands, growth arrest spécifie 6 (GAS6) and protein S (ProS), hâve been identified for TAM kinases. GAS6 can bind to and activate ail three TAM kinases, while ProS is a ligand for MER and TYRO3 (Graham et al, 2014, Nature reviews Cancer 14, 769-785).
[00706] TAM kinases are ectopically expressed or over-expressed in a wide variety of cancers, including breast, colon, rénal, skin, iung, liver, brain, ovarian, prostate, and thyroid malignancies (Graham et al., 2014, Nature Reviews Cancer 14, 769-785; and Linger et al., 2008, Oncogene 32, 3420-3431) and play important rôles in tumor initiation and maintenance. When activated, AXL and MER can regulate tumor cell survivai, prolifération, migration and invasion, angiogenesis, and tumor-host interactions (Schoumacher, M. et aL, Curr. Oncol, Rep. 2017;
19(3); 19). Accordingly, blocking TAM signaling may promote engagement of adaptive immunîty and complément T-cell checkpoint blockade (Akalu, T, et al., Immunological Reviews 2017; 276:165-177). Therefore, TAM inhibition represents an attractive approach for targeting another class of oncogenic RTKs (Graham et al., 2014, Nature Reviews Cancer 14, 769-785; and Linger et al., 2008, Oncogene 32, 3420-3431).
[00707] AXL was originally identified as a transfonning gene from DNA of patients with chronic myelogenous leukemia (O'Bryan et al., 1991, Molecular and Cellular Biology 11, 50165031). GAS6 binds to AXL and induces subséquent auto-phosphorylation and activation of AXL tyrosine kinase. AXL activâtes several downstream signaling pathways including PI3K-AKT, RAF-MAPK., PLC-PKC (Feneyrolles et al., 2014, Molecular Cancer Therapeutics 13, 2141-2148;
Linger et al., 2008, Oncogene 32, 3420-3431). Over-expression or overactivation of the AXL protein has been correlated with the promotion of multiple tumorigenic processes. High levels of AXL expression hâve been associâtes with poor prognosis in different cancers such as glioblastoma multiforme (Hutterer, M., et al., Clin. Caner Res. 2008, 14, 130-138), breast cancer (Wang, X., Cancer Res. 2013, 73, 6516-6525), lung cancer (Niederst, M. et al, Sci. Signaling, 2 0 2013, 6, re6), osteosarcoma (Han, J., Biochem. Biophys. Res. Commun. 2013, 435, 493-500), and acute myeloid leukemia (Ben-Batalla, L., et al., Blood 2013, 122, 2443-2452). AXL is overexpressed or amplified in a variety of malignancies including lung cancer, prostate cancer, colon cancer, breast cancer, melanoma, and rénal cell carcinoma (Linger et al., 2008, Oncogene 32, 3420-3431), and over-expression of AXL is correlated with poor prognosis (Linger et al., 2008, 25 Oncogene 32, 3420-3431). AXL activation promûtes cancer cell survivai, prolifération, angiogenesis, metastasis, and résistance to chemotherapy and targeted thérapies. AXL knockdown or AXL antibody can inhibit the migration of breast cancer and NSCLC cancer in vitro, and blocked tumor growth in xenograft tumor models (Li et al., 2009, Oncogene 28, 3442-3455). In pancreatic cancer cells, inhibition of AXL decreased cell prolifération and survivai (Koorstra et 30 al., 2009, Cancer Biology & Therapy 8, 618-626). In prostate cancer, AXL inhibition decreased cell migration, invasion, and prolifération (Tai et al., 2008, Oncogene 27, 4044-4055). In triple20534 négative breast cancer, patients typically présent a significant clinical challenge, as they do not respond to the varions targeted cancer thérapies due to an apparent lack of RTK activation. However, patients with triple-negative breast cancer do show some response to taxane-based chemotherapy and studies hâve suggested that combining anti-mitotic drugs (e.g., docetaxel) with 5 an AXL inhibitor sensitized cancer cells to the anti-mitotic drug, and AXL in combination with an anti-mitotic drug may be an appropriate combination therapy in this disease setting ( Wilson, et al.. Cancer Res. 2014, 74(20), 5878-5890).
[00708] T AM kinases can contribute to therapeutic résistance by at least three mechanisms: intrinsic survival signaling in tumor cells, induction of TAM kinases as an escape mechanism for 10 tumors that hâve been treated with oncogene-targeted agents, and immunosuppression in the tumor microenvironment (Graham, et al, Nature Reviews Cancer, 2014, 14, 769-785).
[00709] TAM kinases were found to promote résistance to cytotoxic chemotherapies (chemoresistance) in leukemia cells and soiid tumor cells (Graham, et al, Nature Reviews Cancer, 2014, 14, 769-785). Transgenic lymphocytes ectopically expressing MER were found to be more 15 résistant to déxaméthasone than wild-type lymphocytes (Keating, A.K., et al., Oncogene, 2006,
25, 6092-6100), and stimulation of B-ALL cells with GAS6 increased résistance to cytarabine (Shiozawa, Y., et al., Neoplasia, 2010, 12, 116-127). AXL is induced in acute myeloid leukemia (AML) cells that hâve been treated with cytotoxic chemotherapies, and it médiates increased chemoresistance (Hong, C.C., et aL, Cancer Lett., 2008, 268, 314-324). Chemotherapy-resistant chronic myeloid leukemia (CML) cell lines hâve upregulated levels of AXL, and shRNA-mediated knockdown of AXL increases chemosensitivity in CML cells and xenograft models (Zhao, Y., et al,, Cancer Invest. 2012, 30, 287-294). Similarly, shRNA-mediated MER knock-down sensitizes B-cell acute lymphoblastic leukemia (B-ALL) and T-lineage acute lymphoblastic leukemia (TALL) cells to a range of chemotherapies (Linger, R.M., et al., Blood, 2013, 122, 1599-1609;
Brandao, L.N., et aL, Blood Cancer J., 2013, 3, el 01). In solid tumors such as non-small cell lung cancer, pancreatic ductal adenocarcinoma, astrocytoma, lung adenocarcinoma, ovarian cancer, melanoma, and glioblastoma multiforme, overexpression of AXL or MER promûtes chemoresistance, and shRNA-mediated inhibition sensitizes cells to treatment with cytotoxic chemotherapies (Linger, R.N., et al., Oncogene, 2013, 32, 3420-3431; Song, X., et al., Cancer,
2011, 117, 734-743; Keating, A.K., et al., Mol. Cancer Ther. 2010, 9, 1298-1307; Lay, J.D., et aL,
Cancer Res. 2007, 67, 3878-3887; Zhao, Y., et al., Cancer Invest, 2012, 30, 287-294; Macleod,
K., Cancer Res. 2005, 65, 6789-6800; Zhu, S., et al., Proc.Natl Acad. Sci. USA, 2009, 106, 1702517030; Wang, Y., et al., Oncogene 2013, 32, 872-882).
[00710] In contrast to chemoresistance, examples of acquired résistance for TAM kinases are currently limited to AXL. AXL is upreguîated in imatînib-resistant CML and gastrointestinal 5 stromal tumor (GIST) cell lines and tumor samples (Mahadevan, D., et al., Oncogene, 2007, 26, 3909-3919; Dufies, M., et al., Oncotarget 2011, 2, 874-885; Gioia, R., et al., Blood, 2011, 118, 2211-2221), and siRNA-mediated knockdown of AXL restored imatinib sensîtîvity to résistant cell lines (Dufies, M., et al,). Simiiariy, AXL is induced in lapatinib-resistant HER2 (also known as ERBB2)-positive breast cancer cell lines, and AXL inhibition restored lapatinib sensitivity (Liu, 10 L., et al., Cancer Res. 2009, 69, 6871-6878). AXL has been associated with acquired résistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (e.g., lapatinib and erlotinib) and therapeutic antibodies (e.g., cetuximab) in triple-negative breast cancer (Meyer, A.S. et al., Sci. Signal 2013, 6, ra66), colorectal cancer (Brand, et al., Cancer Res. 2014, 74:5152-5164), head and neck cancer (Kiles, K.M, et al., Mol. Cancer Ther. 2013, 12, 2541-2558) cell lines, and non15 small cell lung cancer (Zhang, Nat. Genet. 2013, 44(8), 852-860). AXL has also been associated with acquired résistance to inhibitors targeting other kinases, including PI3Ku inhibitors such as apelisib (BYL719) in head and neck and esophageal squamous cell carcinomas (Elkabets, et al., Cancer Cell 2015, 27:533-546), MEK inhibitors (e.g., U0126 (l,4-Diamino-2,3-dicyano-l,4bis(o-aminophenylmercapto)butadiene) and PD 325901 (l,4-Diamino-2,3-dicyano-l,4-bis(o20 aminophenylmercapto)butadiene) in triple-negative breast cancer cell lines and melanoma cell lines (Miller, et al., Cancer Discovery 2016, 6:382-39), fibroblast growth factor (FGFR) (Ware, K.E., Oncogenesis 2013, 2, e39), anaplastic lymphoma kinase (ALK) (Kim, H.R., et al.. Mol. Oncol. 2013, 7, 1093-1102) and insulin-like growth factor 1 receptor (IGF1R) (Huang, R., Cancer Res. 2010, 70, 7221-7231), and AXL inhibition has been demonstrated to overcome or delay 25 résistance to these inhibitors. AXL is upregulated in NSCLC cell lines and xenografts that are résistant to EGFR tyrosine kinase inhibitors (erlotinib) and antibody drugs (cetuximab) (Brad, T.M., et al., Cancer Res. 2014, 74, 5152-5164; Zhang, Z., et al., Nature Genet. 2012, 44, 852-860), and it is induced in 20% of matched tumor samples taken from patients with NSCLC after development of résistance to the EGFR inhibitor erlotinib.
[00711] Regarding MER and AXL dual inhibitors, the normal rôles of MER and AXL in preventing or terminating innate immune-mediated inflammation and natural killer (NK) cell responses are subverted in the tumor microenvironment. MER and AXL decrease NK cell antitumor activity, which allows increased métastasés.
100712] MER was originally identified as a phospho-protein from a lymphoblastoid expression library (Graham et al., 1995, Oncogene 10, 2349-2359). Both GAS6 and ProS can bind 5 to MER and induce the phosphorylation and activation of MER kinase (Lew et al., 2014. eLife, 3:e03385). Like AXL, MER activation also conveys downstream signaling pathways including P13K-Akt and Raf-MAPK (Linger et al., 2008, Oncogene 32, 3420-3431). MER is over-expressed in many cancers including multiple myeloma, gastric, prostate, breast, melanoma and rhabdomyosarcoma (Linger et al., 2008, Oncogene 32, 3420-3431). MER knockdown inhibits 10 multiple myeloma cell growth in vitro and in xenograft models (Waizenegger et al., 2014,
Leukemia, 1-9). In acute myeloid leukemia, MER knockdown induced apoptosis, decreased colony formation, and increased survival in a mouse model (Lee-Sherick et al., 2013, Oncogene 32, 5359-5368). MER inhibition increased apoptosis, decreased colony formation, increased chemo-sensitivity, and decreased tumor growth in NSCLC (Linger et al., 2013, Oncogene 32, 15 3420-3431). Similareffects areobserved for MER knockdown in melanoma (Schlegel étal., 2013) and glioblastoma (Wang et al., 2013, Oncogene 32, 872-882).
[00713] TYRO3 was originally identified through a PCR-based cloning study (Lai and Lemke, 1991, Neuron 6, 691-704). Both ligands, GAS6 and ProS, can bindto and activate Tyro3. TYRO3 also plays a rôle in cancer growth and prolifération. TYRO3 is over-expressed in 2 0 melanoma cetls, and knockdown ofTYRO3 induces apoptosis inthese cells (Demarest et al., 2013, Biochemistry 52, 3102-31 i 8).
[00714] TAM kinases hâve emerged as potential iinmune-oncology targets. The durable clinical responses to immune checkpoint blockade observed in cancer patients clearly indicate that the immune System plays a critical rôle in tumor initiation and maintenance. Genetic mutations 25 from cancer cells can provide a diverse set of antigens that the immune cells can use to distinguish tumor cells from their normal counterpart. However, cancer cells hâve evolved multiple mechanisms to évadé host immune surveillance. In fact, one hallmarkofhuman cancer is its ability to avoid immune destruction. Cancer cells can induce an immune-suppressive microenvironment by promoting the fonnatîon of M2 tumor associated macrophages, myeloid derived suppressor 30 cells (MDSC), and regulatory T cells. Cancer cells can also produce high levels of immune checkpoint proteins such as PD-L1 to induce T cell anergy or exhaustion. It is now clear that tumors co-opt certain immune-checkpoint pathways as a major mechanism of immune résistance (PardoU, 2012, Cancer 12, 252-264). Antagonizing these négative regulators of T-celi fonction with antibodies has shown striking efficacy in clinical trials of a number of malignancies including advanced melanoma, non-smail cell long and bladder cancer. While these thérapies hâve shown encouraging results, not ail patients mount an anti-tumor response suggesting that other immunesuppressive pathways may also be important.
[00715] T AM kinases hâve been shown to fonction as checkpoints for immune activation in the tumor milieu. Ail TAM kinases are expressed in NK cells, and TAM kinases inhibit the antitumor activity of NK cells. LDC1267, a small molécule TAM kinase inhibitor, activâtes NK cells, and blocks metastasis in tumor models with different histologies (Paolino etaL, 2014, Nature 507, 508-512). In addition, MER kinase decreases the activity of tumor associated macrophages through the increased sécrétion of immune suppressive cytokines such as ILIO and IL4, and decreased production of immune activâtingcytokines such as IL 12 (Cook étal., 2013, The Journal of Clinical Investigation 123, 3231-3242). MER inhibition has been shown to reverse this effect. As a resuit, MER knockout mice are résistant to PyVmT tumor formation (Cook et al., 2013, Journal of Clinical Investigation 123, 3231-3242). The rôle of TAM kinases in the immune response is aiso supported by knockout mouse studies. TAM triple knockout mice (TKO) are viable, However, these mice displayed signs of autoimmune disease including enlarged spleen and lymph nodes, autoantibody production, swollen footpad and joints, skin lésions, and systemic lupus erythematosus (Lu and Lemke, 2001, Science 293, 306-311). This is consistent with the knockout phenotype for approved immune-oncology targets such as CTLA4 and PD-L Both CTLA-4 and PD-1 knockout mice showed signs of autoimmune disease, and these mice die within a few weeks after birth (Chambers et al., 1997, Immunity 7, 885-895; and Nishimura et al., 2001, Science 291, 319-322). Therefore, inhibition of TAM kinases alone or in combination with other immune thérapies may increase the ability of the immune System to make a therapeutically bénéficiai immune response against the cancer.
[00716] The MET receptor tyrosine kinases (e.g., c-Met) Controls growth, invasion and metastasis in cancer cells. The c-Met is activated in human cancer by a variety of different molecular mechanisms (see, e.g., Zhang et al., Carcinogenesis 4:345-355, 2016). For example, a c-Met-associated disease or condition (e.g., a c-Met-associated cancer) include: (i) mutations that alter the sequence and increase the activity of c-Met kinase; (ii) mutations in regulatory sequences
100 controlling c-Met expression or regulators of c-Met expression that confer increased expression of c-Met, (iii) mutations that alter the c-Met polypeptide sequence to confer increased c-Met kinase half-life (e.g., a mutation in a MET gene that results in exon 14 skipping during mRNA splicing that results in an increased level of c-Met in a mammalian cell); (iv) méthylation of a MET gene (see, e.g., Nones et al., Int. J. Cancer 135:1110-8, 2014); (v) méthylation of long interspersed nuclear element (Ll) présent in the MET intron between exon 2 and exon 3 (Weber et al., Oncogene 29:5775-5784, 2010); (vi) METgene amplification; or (vii) by simultaneous expression of receptor and ligand, which results in autocrine stimulation of cancer ceils (Birchmeier et al., Nat. Rev. Mol. Cell Biol. 4:915-925, 2003).
[00717] Exemplary mutations in a MET gene that alter the sequence of a c-Met kinase and increase the activity of c-Met kinase (e.g., as compared to wildtype c-Met kinase) include, but are not limited to those listed in Table 1.
Table 1. Exemplary list of mutations in a MET gene that alter the sequence of a c-Met kinase and increase the activity of the c-Met kinase
MET Isoform 1 mutation MET Isoform 2 mutation Reference
VI0921 VI1101 Schmidt et al., Oncogene 18:2343-2350, 1999
H1094L H1112L Schmidt et al., Oncogene 18:2343-2350, 1999
H1094R H1112R Schmidt étal., Cancer Research 58:1719-1722, 1998
H1094Y H1112Y Schmidt et al., Oncogene 18:2343-2350, 1999
H1106D H1124D Schmidt et al., Oncogene 18:2343-2350, 1999
D1228H D1246H Bardelli et al., Proc. Natl. Acad. Sci. 95: 14379-14383, 2002
D1228N D1246N Bardelli et al., Proc. Natl. Acad. Sci. 95: 14379-14383, 2002
Y1230C Y1248C Bardelli et al., Proc. Natl. Acad. Sci. 95: 14379-14383, 2002
101
Y1230D Y1248D Schmidt et al., Oncogene 18:2343-2350, 1999
Y1230H Y1248H Bardelli étal., Proc. Natl. Acad. Sci. 95: 14379-14383, 2002
M1250T MI268T Bardelli et al., Proc. Natl. Acad. Sci. 95: 14379-14383, 2002
Exemplary mutations that alter the c-Met polypeptide sequence to confer increased c-Met kinase half-life (as compared to a wildtype c-Met kinase) include, but are not limited to, the mutations listed in Table 2 that promote skipping of MET exon 14 during mRNA spiicing. Other exemplary mutations that are predicted to promote skipping of MET exon 14 during mRNA spiicing include, but are not limited to, those disclosed in Frampton et al., Cancer Discovery 5(8):850-9, 2015; and Heist et al., (9/7^0/0^/.^21(4):481-6, 2016. The portion of the c-Met protein encoded by exon 14, most prominently Y1003 in a DpYR motif, is required for efficient recruitment of the E3 ubiquitin-proteîn ligase CBL, which targets MET for ubiquitin-mediated dégradation (Lee et al., J. Biol. Chem. 269:19457-61, 1994; Lee et aL, Exp. Mol. Med. 38:565-
73, 2006; Lee et al., Oncogene 33:34-43, 2014). Skipping of MET exon 14 in mRNA spiicing results in a c-Met kinase that ma intains the reading frame and that demonstrates increased c-Met protein stability and prolonged signaling upon HGF stimulation, leading to increased oncogenic potential (Peschard et al., Mol. Cell 8:995-1004, 2001; Abella étal., Mol. Cell. Biol. 25:9632-45, 2005). Other exemplary mutations that alter the c-Met polypeptide sequence to confer increased c-Met kinase half-life include, but are not limited to an amino acid substitution at Y1003 (e.g., a Y1003F ami no acid substitution) (Peschard et al., Mol. Cell 8:995-1004, 2001).
Table 2. Exemplary list of mutations that confer skipping of MET exon 14
Chromosomal
Reference
Altered sequence (‘Reference
102
location sequence ‘ dénotés délétion) 1----
chr7:l 16411875 -116411897 AAGCTCTTT CTTTCTCTCT GTT Kong-Beltran et al., Cancer Res. 66(1):283-289, 2006
chr7:l 16412022 -116412050 ACCGAGCTA CTTTTCCAG AAGGTATAT T Kong-Beltran et al., Cancer Res. 66(1):283-289, 2006
chr7:l 16412043 -116412044 G T Kong-Beltran et al.. Cancer Res. 66(1):283-289, 2006
chr7:116411854 -116411874 CCCATGATA GCCGTCTTT AA Onozato et al., J. Thorac. Oncol. 4:5-11,2009.
chr7:l 16411884 -116411895 CTTTCTCTCT G Onozato et al., J. Thorac. Oncol. 4:5-11,2009.
chr7:116411886 -116411905 TTCTCTCTGT TTTAAGATC - Onozato et al., J. Thorac. Oncol. 4:5-11,2009.
chr7:l 16412043 -116412044 G A Onozato et al., J. Thorac. Oncol. 4:5-11,2009.
chr7:116412043 -116412044 G T Asaoka et al., Biochem. Biophys. Res. Comm. 394:1042-6, 2010.
chr7:l 164! 1884 -116411896 CTTTCTCTCT GT - Jenkins et al., Clin. Lang Cancer 16:e!01-el04,2015.
chr7:116412042 -116412043 G c Waqar et al., J. Thorac. Oncol. 10x29-31,2015.
chr7:l 16412042 -116412043 G c Mendenhall et al., J. Thorac. Oncol. 10x23-34,2015.
Exemplary c-Met-associated cancers include, but are not limited to those listed in Table 3.
103
Table 3, Exemplary c-Met-associated cancers exhibiting increased expression and/or activity of c-Met
Cancer type Type of genetic alterations Reference
Gastrointestinal cancer (GI); METgene amplification; Mo et al., Chronic Dis.
Gastric cancer Amino acid substitution in kinase domain (e.g., an amino acid substitution at position 1108, e.g., an Al 108S amino acid substitution); point mutation conferring skipping of METexon 14 during mRNA splicing (e.g., chr7:116412043-116412044, G to T mutation) Transi. Med. 3(3):148153, 2017; Tovar et al., Ann. Transi. Med. 5(10):205, 2017;Asaoka et al., Biocheni. Biophys. Res. Comm. 394:1042-6, 2010.
Colorectal Adenocarcinoma Amino acid substitution at position 375 (e.g., aN375S amino acid substitution); an amino acid substitution at position 1010 (e.g., a Tl 0101 amino acid substitution); an amino acid substitution at position 988 (e.g., a R988C amino acid substitution); an amino acid substitution at position 1253 (e.g., a Y1253D amino acid substitution); and an amino acid substitution at position 1248 (e.g., a Y1248II amino acid substitution) Zenali étal., Oncoscience 2(5):533-541,2015.
Colorectal carcinoma (CRC) MET gene amplification; MET overexpression; Zeng et al., Cancer Leti. 265:258-269,2008;
104
amino acid substitutions in JM domain of c-Met kinase (e.g., an amino acid substitution at position 970 (e.g., an R970C amino acid substitution) and an amino acid substitution at position 992 (e.g., a T992I amino acid substitution) Kong-Beltran et al., Cancer Res. 66:283-9, 2006; Tovar et al., Arm. Transi. Med. 5(10):205, 2017.
Non-small cell lung cancer (NSCLC) Point mutation conferring skipping of METexon 14 during mRNA splicing; MET gene amplification; amino acid substitutions in cMet kinase domain (e.g., an amino acid substitution at position 970 (e.g., a R970C amino acid substitution), an amino acid substitution at position 988 (e.g., a R988C amino acid substitution); an amino acid substitution at position 1010 (e.g., a Tl010Î amino acid substitution); an amino acid substitution at position 1058 (e.g., a S1058P amino acid substitution)); amino acid substitution in the JM domain of c-Met kinase (e.g., an amino acid substitution at position 988 (e.g., a R988C amino acid Ichimura et al., Jpn J. Cancer Res. 87:1063- 1069, 1996; Ma et al., Cancer Res. 63:6272-81, 2003; Kong-Beltran et al., Cancer Res. 66:2839, 2006; Tovar et al., 2017, Ann. Transi. Med. 5(10):205,2017
105
substitution), an amino acid substitution at position 1010 (e.g., a T10101 amino acid substitution), an amino acid substitution at position 1058 (e.g., a S1058P amino acid substitution), an amino acid substitution at position 970 (e.g., a R970C amino acid substitution), and an amino acid substitution at position 992 (e.g., a T992I amino acid substitution)).
Heptacellular carcinoma (HCC) MET overexpression; Amino acid substitutions in kinase domain of c-Met (e.g., an amino acid substitution at position 1191 (e.g., a Tl 1911 amino acid substitution), an amino acid substitution at position 1262 (e.g., a J1262R amino acid substitution), or an amino acid substitution at position 1268 (e.g., a M1268T or an M1268I amino acid substitution)); an amino acid substitution at position 375 (e.g., an N375S amino acid substitution); an amino acid substitution at position 988 Goyal et al., Clin. Cancer Res. 19:2310-2318,2013; Tovar et al., Ann. Transi. Med. 5(10):205, 2017; Zenali et al., Oncoscience 2(5):533-541,2015
106
(e.g., a R988C amino acid substitution)
Hereditary papillary rénal carcinoma (1-IPRC) Amino acid substitutions in the kinase domain of c-Met (e.g., an amino acid substitution at position 112 (e.g., a Hl 12R, a H112L, or a Hl 121 amino acid substitution), an amino acid position as position 1230 (e.g., a Y1230C, a Y1230H, or a Y1230D amino acid substitution), an amino acid substitution at position 1246 (e.g., a D1246N amino acid substitution), an amino acid substitution at position 1248 (e.g., a Y1248C amino acid substitution), an amino acid substitution at position 1268 (e.g., a M1268T amino acid substitution or a M12681 amino acid substitution). Tovar et al., Ann. Transi. Med. 5(10):205, 2017
Papillary rénal carcinoma Amino acid substitutions in the kinase domain of c-Met (e.g., those listed in Table 1) Jeffers et al., Proc. Natl. Acad. Sci. U.S.A. 94(21):11445-11450, 1997; Schmidt et al., Nat. Genet. 16:68-73, 1997; Schmidt et al., Oncogene 18:2343-50, 1991.
Melanoma An amino acid substitution at position 375 (e.g., a N375S Zenali et al., Oncoscience 2(5):533-541, 2015.
107
amino acid substitution); an amino acid substitution at position 988 (e.g., a R988C amino acid substitution); an amino acid substitution at position 1010 (e.g., a TI0101 amino acid substitution).
Gastric adenocarcinoma An amino acid substitution at position 375 (e.g., an N375S amino acid substitution). Zenali et al., Oncoscience 2(5):533-541,2015.
Appendiceal adenocarcinoma An amino acid substitution at position 375 (e.g,, an N375S amino acid substitution); an amino acid substitution at position 988 (e.g., a R988C amino acid substitution) Zenali et al., Oncoscience 2(5):533-541,2015.
Duodenal adenocarcinoma An amino acid substitution at position 375 (e.g., an N375S amino acid substitution) Zenali et al., Oncoscience 2(5):533-541,2015.
Pancreatic adenocarcinoma An amino acid substitution at position 375 (e.g., an N375S amino acid substitution) Zenali et al., Oncoscience 2(5):533-541,2015.
Lang adenocarcinoma An amino acid substitution at position 375 (e.g., an N375S amino acid substitution); an amino acid substitution at position 988 (e.g., a R988C amino acid substitution); an amino acid substitution at position 1010 (e.g., a T10101 amino acid substitution) Zenali étal., Oncoscience 2(5):533-541,2015.
108
Thyroid papillary carcinoma An amino acid substitution at position 375 (e.g., an N375S amino acid substitution) Zenali et al., Oncoscience 2(5):533-541,2015.
Thyroid medullary carcinoma An amino acid substitution at position 1010 (e.g., a Tl0101 amino acid substitution) Zenali et al., Oncoscience 2(5):533-541,2015.
Ewing sarcoma An amino acid substitution at position 1010 (e.g., a Tl 0101 amino acid substitution) Zenali et al., Oncoscience 2(5):533-541,2015.
Prostate adenocarcinoma An amino acid substitution at position 375 (e.g., an N375S amino acid substitution) Zenali et al., Oncoscience 2(5):533-541,2015.
Squamous cell carcinoma of the head and neck and cervix An amino acid substitution at position 375 (e.g., an N375S amino acid substitution); an amino acid substitution at position 988 (e.g., a R988C amino acid substitution); an amino acid substitution at position 1010 (e.g., an T10I0I amino acid substitution) Zenali et al,, Oncoscience 2(5):533-541,2015,
Rénal cell carcinoma An amino acid substitution at position 375 (e.g., an N375S amino acid substitution); an amino acid substitution at position 1092 (e.g., a VI0921 amino acid substitution); an amino acid substitution at position 1094 (e.g., a H1094L, aH1094R, or a H1094Y amino acid substitution); an amino Zenali et al., Oncoscience 2(5):533-541,2015; Schmidt et al., Oncogene 18:2343-2350, 1999; Scmidt et al., Cancer Research 58:1719-1722, 1998; Bardelli et al., Proc. Natl. Acad. Sci. 95: 14379-14383,2002.
109
acid substitution at position 1106 (e.g., a H1106D amino acid substitution); an amino · acid substitution at position 1228 (e.g., a DI 228H or a D1228N amino acid substitution); an amino acid substitution at position 1230 (e.g., a Y1230C, a Y1230D, or a Y1230H amino acid substitution); an amino acid substitution at position 1250 (e.g., a M1250T amino acid substitution)
Pheochromocytoma and composite pheochromocytoma An amino acid substitution at position 375 (e.g., an N375S amino acid substitution); an amino acid substitution at position 988 (e.g., an R988C amino acid substitution) Zenali et al., Oncoscience 2(5):533-541,2015.
Ovarian serons carcinoma An amino acid substitution at position 375 (e.g., an N375S amîno acid substitution); an amino acid substitution at position 1010(e.g.,a 10101 amino acid substitution) Zenali et al., Oncoscience 2(5):533-541,2015.
Ovarian clear cell carcinoma An amino acid substitution at position 375 (e.g., an N375S amino acid substitution) Zenali et al., Oncoscience 2(5):533-541,2015,
Ovarian mixed carcinoma An amino acid substitution at position 1010 (e.g., a T1010I Zenali et al., Oncoscience 2(5):533-541,2015.
110
amino acid substitution) -----—
Peritoneal serons carcinoma An amino acid substitution at position 1010 (e.g., a T1010I amino acid substitution) Zenali et al., Oncoscience 2(5):533-541, 2015.
Breast ductal adenocarcinoina An amino acid substitution at position 375 (e.g., an N375S amino acid substitution); an amino acid substitution at position 1010 (e.g., a T1010I amino acid substitution). Zenali et al., Oncoscience 2(5):533-541,2015.
Uterine leiomyosarcoma An amino acid substitution at position 375 (e.g., an N375S amino acid substitution); amino acid substitution at position 1010 (e.g., a Tl0101 amino acid substitution) Zenali étal., Oncoscience 2(5):533-541, 2015.
Uterîne endometrioid adenocarcinoina An amino acid substitution at position 375 (e.g., an N375S amino acid substitution); an amino acid substitution at position 1010 (e.g., an T10101 amino acid substitution). Zenali étal., Oncoscience 2(5):533-541, 2015.
Uterine malignant mixed mullerian tumor An amino acid substitution at position 375 (e.g., an N375S amino acid substitution) Zenali et al., Oncoscience 2(5):533-541,2015.
Glioblastoma An amino acid substitution at position 375 (e.g., anN375S amino acid substitution) Zenali et al., Oncoscience 2(5):533-541,2015.
Anaplastic glioma An amino acid substitution at position 375 (e.g., an N375S amino acid substitution) Zenali étal., Oncoscience 2(5):533-541,2015.
111
Oligodendroglioma An amino acid substitution at position 1010 (e.g., an Tl 010T amino acid substitution) Zenali et al., Oncoscience 2(5):533-541,2015.
Desmoplastic small round cell tumor An amino acid substitution at position 375 (e.g., an N375S amino acid substitution) Zenali et al., Oncoscience 2(5):533-541,2015.
Squamous cell carcinoma of rectum An amino acid substitution at position 375 (e.g., N375S amino acid substitution) Zenali et al., Oncoscience 2(5):533-541,2015.
Salivary gland carcinoma An amino acid substitution at position 375 (e.g., an N375S amino acid substitution) Zenali et al., Oncoscience 2(5):533-541, 2015.
Heart angiosarcoma An amino acid substitution at position 375 (e.g., a N375S amino acid substitution); an amino acid substitution at position 1010 (e.g., a T1010I amino acid substitution) Zenali et aL, Oncoscience 2(5):533-541,2015.
Gastrointestinal stromal tumor (GIST) An amino acid substitution at position 1010 (e.g., a T10 E 01 amino acid substitution); an amino acid substitution at position 988 (e.g., an R988C amino acid substitution) Zenali et al., Oncoscience 2(5):533-541,2015.
Invasive thymoma An amino acid substitution at position 375 (e.g., an N375S amino acid substitution) Zenali et al., Oncoscience 2(5):533-541,2015.
Spindle sarcoma An amino acid substitution at position 375 (e.g., an N375S amino acid substitution) Zenali et al., Oncoscience 2(5):533-541,2015.
112
In some embodiments, compounds of Formula I can be used to treat a c-Met associated cancer expressing a c-Met kinase that is résistant (e.g., to at least some extent as compared to a wildtype c-Met kinase) to a c-Met inhibitor (e.g., a Type I c-Met inhibitor). Non-limiting examples of amino acid substitutions that resuit in résistance of c-Met to a c-Met inhibitor (e.g., a Type 1 c-Met inhibitor) include: an amino acid substitution at position 1092 (e.g., a VI0921 amino acid substitution in isoform 1 of c-Met or a VI1101 amino acid substitution in isoform 2 of c-Met); an amino acid substitution at position 1094 (e.g., a H1094L amino acid substitution in isoform 1 of c-Met or a H1112L amino acid substitution in isoform 2 of c-Met; an H1094Y amino acid substitution in isoform 1 of c-Met or an H1112Y amino acid substitution in isoform 2 of c-Met); an amino acid substitution at position 1155 (e.g., a VI155L amino acid substitution in isoform 1 or a V] 173L amino acid substitution in isoform 2 of c-Met); an amino acid substitution at position 1163 (e.g., a G1163R amino acid substitution in isoform 1 of c-Met or a Gl 181R amino acid substitution in isoform 2 of c-Met); an amino acid substitution at position 1195 (e.g., an L1195F amino acid substitution in isoform 1 ofc-Met or a L1213F amino acîd substitution in isoform 2 of c-Met; an L1195V amino acid substitution in isoform 1 of c-Met or an L1213V amino acid substitution in isoform 2 of c-Met); an amino acid substitution at position 1200 (e.g., an Fl2001 amino acid substitution in isoform 1 ofc-Met or an F1218I amino acid substitution in isoform 2 ofc-Met); an amino acid substitution at position 1211 (e.g., an Ml21 IL amino acid substitution in isoform 1 of c-Met or an Μ1229L amino acid substitution in isoform 2 of c-Met); an amino acid substitution at position 1228 (e.g., a DI228A amino acid substitution in isoform 1 of c-Met or a DI246A amino acid substitution in isoform 2 of c-Met; a D1228G amino acid substitution in isoform 1 of c-Met or a D1246G amino acid substitution in isoform 2 of c-Met; a D1228H amino acid substitution in isoform 1 of c-Met or a D1246H amino acid substitution in isoform 2 of cMet; a D1228N amino acid substitution in isoform 1 ofc-Met or a D1246N amino acid substitution in isoform 2 of c-Met; a D1228V amino acid substitution in isoform 1 of c-Met or a D1246V amino acid substitution in isoform 2 of c-Met; or a D1228Y amino acid substitution in isoform I of cMet or a D1246Y amino acid substitution in isoform 2 of c-Met); an amino acid substitution at position 1230 (e.g., a Y1230C amino acid substitution in isoform 1 of c-Met or a Y1248C amino acid substitution in isoform 2 of c-Met; a YI230H amino acid substitution in isoform 1 of c-Met or a Y1248H amino acid substitution in isoform 2 of c-Met; or a Y1230S amino acid substitution in isoform l of c-Met or a Y1248S amino acid substitution in isoform 2 of c-Met); or an amino
113 acid substitution at position 1250 (e.g., a M125ÛT amino acid substitution in isoform 1 of c-Met or a M1268T amino acid substitution in isoform 2 of c-Met). Non-limiting examples of Type I inhîbitors include crizotinib (PF-02341066), capmatinib, NVP-BVU972, AMG 337, bozitinib, glumetinib, savolitinib, and tepotinib. In some embodiments, amino acid substitutions that resuit in résistance of c-Met to a Type 1 c-Met inhibitor include L1195V, Fl2001, D1228H, D1228N, Y1230C, Y1230H, and Y1230S.
In some embodiments, compounds of Formula I can be used to treat a c-Met associated cancer having a chromosomal translocation that resuit in a fusion protein including the c-Met kinase domain, where the fusion protein has increased c-Met activity as compared to a wildtype c-Met kinase (e.g., a Met-TPR fusion protein (Rodrigues et al.. Mol. Cell. Biol. 13:67116722, 1993) and the fusion protein/chromosomal translocation described in Cooper et al., Nature 311(5981):29-33, 1984, [00718] Accordingly, in one embodiment, provided herein is a method fortreating a TAMassociated disease or disorder (e.g., a TAM-associated cancer), a c-Met-associated disease or disorder (e.g., a c-Met-associated cancer), or both, in a patient in need thereof, the method comprising administering to the patient a therapeutically effective amount of a compound of Formula 1 or a pharmaceutically acceptable sait thereof or a pharmaceutical composition thereof. [00719] Also provided herein are methods of treating a patient identified or diagnosed as having a TAM-associated cancer, a c-Met-associated cancer, or both, that include administering to a patient identified or diagnosed as having a TAM-associated cancer, a c-Met-associated cancer, or both, a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof or a pharmaceutical composition thereof.
[00720] Also provided herein are methods of treating a patient having a cancer that include: (a) identifying the patient as having a TAM-associated cancer, a c-Met-associated cancer, or both, and (b) administering to the patient identified as having a TAM-associated cancer, a c-Met-associated cancer, or both, a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof or a pharmaceutical composition thereof.
[00721] Also provided herein are methods of decreasing the risk of developing a metastasis or an additional metastasis in a patient identified or diagnosed as having a TAM-associated cancer, a c-Met-associated cancer, or both, that include administering to the patient identified or diagnosed as having a TAM-associated cancer, a c-Met-associated cancer, or both, a therapeutically effective
114 amount of a compound of Formula I or a pharmaceutically acceptable sait thereof or a pharmaceutical composition thereof. In some embodiments, the methods resuit in at least a 1% réduction (e.g., at least a 2% réduction, at least a 3% réduction, at least a 4% réduction, at least a 5% réduction, at least a 6% réduction, at least a 8% réduction, at least a 10% réduction, at least a 5 12% réduction, at least a 14% réduction, at least a 16% réduction at least a 18% réduction, at least a 20% réduction, at least a 25% réduction, at least a 30% réduction, at least a 35% réduction, at least a 40% réduction, at least a 45% réduction, at least a 50% réduction, at least a 55% réduction, at least a 60% réduction, at least a 65% réduction, at least a 70% réduction, at least a 75% réduction, at least a 80% réduction, at least a 85% réduction, at least a 90% réduction, at least a 95% réduction, 10 or at least a 99% réduction) in the patient’s risk of developing a metastasis or an addîtional metastasis, e.g., as compared to a population of subjects having a similar TAM-associated cancer and/or c-Met-associated cancer but receiving a different treatment or no treatment.
[00722] Also provided are methods of decreasing the risk of developing a metastasis or an additional metastasis in a patient having a cancer that include: (a) identifying the patient as having 15 a TAM-associated cancer, a c-Met-associated cancer, or both; and (b) administering to the patient identified as having a TAM-associated cancer, a c-Met-associated cancer, or both, a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof or a pharmaceutical composition thereof. In some embodiments, the methods resuit in at least a 1% réduction (e.g., at least a 2% réduction, at least a 3% réduction, at least a 4% réduction, at least a 20 5% réduction, at least a 6% réduction, at least a 8% réduction, at least a 10% réduction, at least a
12% réduction, at least a 14% réduction, at least a 16% réduction, at least a 18% réduction, at least a 20% réduction, at least a 25% réduction, at least a 30% réduction, at least a 35% réduction, at least a 40% réduction, at least a 45% réduction, at least a 50% réduction, at least a 55% réduction, at least a 60% réduction, at least a 65% réduction, at least a 70% réduction, at least a 75% réduction, 25 at least a 80% réduction, at least a 85% réduction, at least a 90% réduction, at least a 95% réduction, or at least a 99% réduction) in the patienfs risk of developing a metastasis or an additional metastasis, e.g., as compared to a population of subjects having a similar TAM-associated cancer and/or c-Met-associated cancer, but receiving a different treatment or no treatment.
[00723] Also provided are methods of decreasing migration and/or invasion of a cancer cell in a patient identified as having a TAM-associated cancer, a c-Met-associated cancer, or both, that include administering to a patient identified or diagnosed as having a TAM-associated cancer, a c-
115
Met-associated cancer, or both, a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof or a pharmaceutical composition thereof. In some embodiments, the methods resuit in at least a 1% decrease (e.g., at least a 2% decrease, at least a 3% decrease, at least a 4% decrease, at least a 5% decrease, at least a 6% decrease, at least a 8% decrease, at least a 10% decrease, at least a 12% decrease, at least a 14% decrease, at least a 16% decrease, at least a 18% decrease, at least a 20% decrease, at least a 25% decrease, at least a 30% decrease, at least a 35% decrease, at least a 40% decrease, at least a 45% decrease, at least a 50% decrease, at least a 55% decrease, at least a 60% decrease, at least a 65% decrease, at least a 70% decrease, at least a 75% decrease, at least a 80% decrease, at least a 85% decrease, at least a 90% decrease, at least a 95% decrease, or at least a 99% decrease) in the migration and/or invasion of a cancer cell in the patient, e.g., as compared to the migration and/or invasion of a cancer cell or a population of cancer cells in a subject having a similar TAM-associated cancer and/or c-Metassociated cancer but receiving a different treatment or no treatment.
[00724] Also provided herein are methods of decreasing migration and/or invasion of a cancer cell in a patient having a cancer that include: (a) identîfying the patient as having a TAM-associated cancer, a c-Met-associated cancer, or both; and (b) administering to the patient identified as having a TAM-associated cancer, a c-Met-associated cancer, or both, a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof or a pharmaceutical composition thereof. In some embodiments, the methods resuit in at least a 1% decrease (e.g., at least a 2% decrease, at least a 3% decrease, at least a 4% decrease, at least a 5% decrease, at least a 6% decrease, at least a 8% decrease, at least a 10% decrease, at least a 12% decrease, at least a 14% decrease, at least a 16% decrease, at least a 18% decrease, at least a 20% decrease, at least a
25% decrease, at least a 30% decrease, at least a 35% decrease, at least a 40% decrease, at leasta
45% decrease, at least a 50% decrease, at least a 55% decrease, at least a 60% decrease, at leasta
65% decrease, at least a 70% decrease, at least a 75% decrease, at least a 80% decrease, at leasta
85% decrease, at least a 90% decrease, at least a 95% decrease, or at least a 99% decrease) in the migration and/or invasion of a cancer cell in the patient, e.g., as compared to the migration and/or invasion of a cancer cell or a population of cancer cells in a subject having a similar TAMassociated cancer and/or a c-Met-associated cancer, but receiving a different treatment or no treatment.
[00725] Some embodiments ofthese methods further include administering to the patient at
116 least one additional anticancer agent (e.g, any of the exemplary additional anticancer agents described herein or known in the art). For example, in some examples, the at least one anticancer agent or therapy can be selected from the group of: an immune checkpoint inhibitor, a kinase inhibitor, a chemotherapy, radiation, and surgery.
[00726] In some embodiments of any of the methods described herein, the patient was previously treated with al least one additional anticancer agent (e.g, any of the additional anticancer agents described herein) and the previous treatment with the at least one additional anticancer agent was unsuccessful (e.g, the patient previously developed résistance to one or more of the at least one additional anticancer agent).
[00727] In some embodiments of any of the methods described herein, the at least one additional anticancer agent is selected from the group of: a chemotherapeutic agent, a Pl-3 kinase inhibitor, an EGFR inhibitor, a HER2/neu inhibitor, an FGFR inhibitor, an ALK inhibitor, an IGF1R inhibitor, a VEGFR inhibitor, a PDGFR inhibitor, a glucocorticoid, a BRAF inhibitor, a MEK inhibitor, a HER4 inhibitor, a MET inhibitor (e.g, a type I c-Met kinase inhibitor), a RAF inhibitor, an Akt inhibitor, a FTL-3 inhibitor, a MAP kinase pathway inhibitor.
[00728] In some embodiments of any of the methods described herein, the at least one additional anticancer agent can include a kinase inhibitor, and the patient previously developed résistance to the kinase inhibitor. In some embodiments of any of the methods described herein, the at least one anticancer agent includes a kinase inhibitor selected from the group of: bozitinib, l-(6,7-Dihydro20 5H-benzo[6,7]cyciohepta[l,2-c]pyridazin-3-yl)-N3-[7(S)-( 1-pyrrolidiny!)-6,7,8,9-tetrahydro-5Hbenzocycloheptene-2-yl]-lH-l,2,4-triazole-3,5-dÎamine (BGB324), crizotinib, foretinib, (N-[4(2-Amino-3-chloropyridin-4-yloxy)-3-fluorophenyl]-4-ethoxy-l-(4-fluorophenyl)-2-oxo-l,2dihydropyridine-3-carboxamide (BMS-777607), , amuvatinib, BMS-796302, cabozantinib, glesatinib (MGCD265), 2-(4-Fluoropheuyl)-N-[3-fluoro-4-(3-phenyl-l H-pyrrolo[2,3-b]pyridin25 4-yloxy)phenyl]-l ,5-dimethyl-3-oxo-2,3-dihydro-lH-pyrazole-4-carboxamide (NPS-I034), N[4-[(6,7-Dimethoxyquino[in-4-yl)oxy]-3-fluorophenyl]-4-ethoxy-l-(4“fluoro-2-methylphenyl)1 H-pyrazole-3-carboxamide hydrochloride (LDCI267), gilteritinib, [3-(2-[[3-Fluoro-4-(4methylpiperazin-l-yl)phenyl]amino]-5-methyl-7H-pynOlo[2,3-d]pyrimidîn-4yl)phenyl]acetonitrile (SG1-7079), dubermatinib (TP-0903), trans-4-[2-(Butylamino)-5-[4-[(430 methylpiperazin-]-yl)methyl]phenyl]-7H-pyrrolo[2,3-d]pyrimidin-7-yl]cyclohexanol (UNC2025), 3-[3-[4-(Morpholin-4-ylmethyl)-lH-pyrrol-2-ylmethylene]-2-oxo-2,3-dihydro-lH20534
117 indol-5-yimethyl]thiazolidine-2,4-dione hydrochloride (S49076), sunitinib, 12AH, Mabl73, YW327.6S2, D9, ES, merestinib, , [3-(2-[[3-Fluoro-4-(4-methylpiperazin-l-yl)phenyl]amino]-5methyl-7H-pyrrolo[2,3-d]pyrimidin-4-yl)phenyl]acetonitrile (SGI-7079), , N-[4-[(6,7Dimethoxyquinolin-4-yl)oxy]-3-fluorophenyl]-4-ethoxy-l-(4-fluoro-2-methyiphenyl)-lHpyrazole-3-carboxamide hydrochloride, capmatinib, NVP-BVU972, AMG 337, bozitinib, glumetinib, savolitinib, and tepotinib.
[00729] In some embodiments of any of the methods described herein, the at least one additional anticancer agent includes dexamethasone, and the patient previously developed résistance to dexamethasone. In some embodiments of any of the methods described herein, the at least one additional anticancer agent includes cytarabine, and the patient previously developed résistance to cytarabine. In some embodiments of any of the methods described herein, the at least one additional anticancer agent includes imatinib, and the patient previously developed résistance to imatinib. In some embodiments of any of the methods described herein, the at least one additional anticancer agent includes lapatinib, and the patient previously developed résistance to lapatinib. In some embodiments of any of the methods described herein, the at least one additional anticancer agent includes cetuximab, and the patient previously developed résistance to cetuximab. In some embodiments of any of the methods described herein, the at least one additional anticancer agent includes erlotinib, and the patient previously developed résistance to erlotinib. In some embodiments of any of the methods described herein, the at least one additional anticancer agent includes alpelisib, and the patient previously developed résistance to alpelisib. In some embodiments of any of the methods described herein, the at least one additional anticancer agent includes cîsplatin, and the patient previously developed résistance to cisplatin. In some embodiments of any of the methods described herein, the at least one additional anticancer agent includes sunitinib, and the patient previously developed résistance to sunitinib. In some embodiments of any of the methods described herein, the at least one additional anticancer agent includes metformin, and the patient previously developed résistance to metformin.
[00730| In some embodiments of any of the methods described herein, the at least one additional anticancer agent includes an anti-PDl antibody, and the patient previously developed résistance to the anti-PDl antibody. In some embodiments of any of the methods described herein, the at least one additional anticancer agent includes docetaxel, and the patient previously developed résistance to docetaxel. In some embodiments of any of the methods described herein,
118 the at least one additional anticancer agent includes an EGFR inhibitor, and the patient previously deveioped résistance to the EGFR inhibitor.
[00731] In some embodiments of any of the methods described herein, the at least one additional anticancer agent is a Type 1 c-Met inhibitor, and the patient previously deveioped résistance to the c-Met inhibitor. In some embodiments of any of the methods described herein, the at least one additional Type 1 c-Met inhibitor includes crizotinib, and the patient previously deveioped résistance to crizotinib. In some embodiments of any of the methods described herein, the at least one additional Type 1 c-Met inhibitor includes capmatinib, and the patient previously deveioped résistance to capmatinib. In some embodiments of any of the methods described herein, the at least one additional Type 1 c-Met inhibitor includes NVP-BVU972, and the patient previously deveioped résistance to NVP-BVU972. In some embodiments of any of the methods described herein, the at least one additional Type 1 c-Met inhibitor includes AMG 337, and the patient previously deveioped résistance to AMG 337. In some embodiments of any of the methods described herein, the at least one additional Type I c-Met inhibitor includes bozitinib, and the patient previously deveioped résistance to bozitinib. In some embodiments of any of the methods described herein, the at least one additional Type 1 c-Met inhibitor includes glumetinib, and the patient previously deveioped résistance to glumetinib. In some embodiments of any of the methods described herein, the at least one additional Type 1 c-Met inhibitor includes savolitinib, and the patient previously deveioped résistance to savolitinib. In some embodiments of any of the methods described herein, the at least one additional Type 1 c-Met inhibitor includes tepotinib, and the patient previously deveioped résistance to tepotinib.
[00732] In some embodiments, the tumor deveioped a résistance mutation after treatment with the Type 1 c-Met inhibitor. In some embodiments, the résistance mutation in c-Met results in the expression of a c-Met protein including one or more of the following amino acid substitutions: LI195V, F1200I, D1228H, D1228N, D1230C, Y1230H, and Y1230S.
[00733] Also provided herein are methods of selecting a treatment for a patient identifîed or diagnosed as having a TAM-associated cancer, a c-Met-associated cancer, or both, that include selecting a compound of Formula I or a pharmaceutically acceptable sait thereof or a pharmaceutical composition thereof for the patient identifîed or diagnosed as having a TAMassociated cancer, a c-Met-associated cancer, or both. Some embodiments further comprise administering the selected compound of Formula I or the pharmaceutically acceptable sait thereof
119 or the pharmaceutical composition thereof to the patient,
[00734] Also provided herein are methods of selecting a treatment for a patient that include: (a) identifying the patient as having a TAM-associated cancer, a c-Met-associated cancer, or both; and (b) selecting a compound of Formula I or a pharmaceutically acceptable sait thereof or a pharmaceutical composition thereof for the patient identified as having a TAM-associated cancer, a c-Met-associated cancer, or both. Some embodiments further comprise administering the selected compound of Formula I or the pharmaceutically acceptable sait thereof or the pharmaceutical composition thereof to the patient identified as having a TAM-associated cancer, a c-Met-associated cancer, or both.
[00735] In some embodiments of any of the methods described herein, the subject is identified or diagnosed as having a TAM-associated cancer (e.g., any of the TAM-associated cancers described herein, e.g., having any of the exemplary TAM mutations described herein). In some embodiments of any of the methods described herein, the subject is identified or diagnosed as havîng both a TAM-associated cancer (e.g., any of the TAM-associated cancers described herein, e.g., having any of the exemplary TAM mutations described herein) and a c-Met-associated cancer (e.g., any of the exemplary c-Met-associated cancers described herein, e.g., having any of the exemplary c-Met mutations described herein). In some embodiments of any of the methods described herein, the subject is identified or diagnosed as having a c-Met-associated cancer (e.g., any of the exemplary c-Met-associated cancers described herein, e.g., having any of the exemplary c-Met-associated mutations described herein).
[00736] In some embodiments of any of the methods described herein, the c-Met-associated cancer is a cancer having a mutation that increases the activity of a c-Met kinase. In some embodiments of any of the methods described herein, the mutation that increases the activity of a c-Met kinase results in one or more amino acid substitutions in the c-Met kinase. In some embodiments of any of the methods described herein, the c-Met-associated cancer is a cancer having a mutation that increases the expression of c-Met in a mammalian cell. In some embodiments of any of the methods described herein, the c-Met-associated cancer is a cancer having a mutation that confers increased half-life of c-Met kinase in a mammalian cell. In some embodiments of any of the methods described herein, the mutation that confers increased half-life of c-Met kinase in a mammalian cell is a mutation that results in c-Met exon 14 skipping during
120 mRNA splicing. In some embodiments of any of the methods described herein, the c-Metassociated cancer is a cancer having a MET gene amplification. In some embodiments of any of the methods described herein, the c-Met-associated cancer is a c-Met-associated cancer that has résistance to a type I c-Met inhibitor.
[00737] In some embodiments of any of the methods described herein, the c-Met-associated cancer is selected from the group of: gastrointestinal cancer (GI), gastric cancer, colorectal adenocarcinoma, colorectal carcinoma (CRC), non-small cell lung cancer (NSCLC), hepatocellular carcinoma (HCC), hereditary papillary rénal carcinoma (HPRC), papillary rénal carcinoma, melanoma, gastric adenocarcinoma, appendiceal adenocarcinoma, duodenal adenocarcinoma, pancreatic adenocarcinoma, lung adenocarcinoma, thyroid papillary carcinoma, thyroid inedullary carcinoma, Ewing sarcoma, prostate adenocarcinoma, squamous cell carcinoma of the head and neck and cervix, rénal cell carcinoma, pheochromocytoma and composite pheochromocytoma, ovarian serous carcinoma, ovarian clear cell carcinoma, ovarian mixed carcinoma, peritoneal serous carcinoma, breast ductal adenocarcinoma, uterine leiomyosarcoma, uterine endometrioid adenocarcinoma, uterine malignant mixed mullerian tumor, glioblastoma, anaplastic glioma, oligodendroglioma, desmoplastic small round cell tumor, squamous cell carcinoma of rectum, salivary gland carcinoma, heart angiosarcoma, gastrointestinai stromal tumor, invasive thymoma, and spindle sarcoma.
[00738] Also provided herein are methods of selecting a treatment for a patient identifîed or diagnosed as having a cancer that include: (a) administering at least one additional anticancer agent to the patient (e.g., any of the additional anticancer agents described herein); (b) after (a), detecting increased expression, level, and/or activity of a TAM kinase and/or c-Met kinase in a cancer cell or an immune cell from the patient; and (c) after (b), selecting a compound of Formula I or a pharmaceutically acceptable sait thereof or a phannaceutical composition thereof for the patient.
[00739| Also provided herein are methods of treating a patient identifîed or diagnosed as having a cancer that include: (a) administering to the patient identifîed or diagnosed as having a cancer one or more doses of at least one additional anticancer agent (e.g., at least one of any of the additional anticancer agents described herein); (b) after (a), detecting an increase in the expression, level, and/or activity of a TAM kinase and/or c-Met kinase in a cancer cell or an immune cell from the patient; and (c) after (b), administering to the patient a therapeutically effective amount of a
121 compound of Formula I or a pharmaceutically acceptable sait thereof or a pharmaceutical composition thereof. In some embodiments, step (c) further includes administering to the patient the at least one additional anticancer agent.
[00740] Also provided herein are methods of treating a patient identified or diagnosed as having a cancer that include: (a) detecting an increase in the expression, level, and/or activity of a TAM kinase and/or c-Met kinase in a cancer cell or an immune cell from a patient identified or diagnosed as having a cancer and previously administered one or more doses of the at least on additional anticancer agent (e.g., any of the additional anticancer agents described herein); and (b) after (a), administering to the patient a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof or a pharmaceutical composition thereof. In some embodiments, step (b) further includes administering to the patient the at least one additional anticancer agent.
[00741] Also provided herein are methods of treating a patient identified or diagnosed as having a cancer that has been previously administered one or more doses of at least one additional anticancer agent and has been identified as having a cancer cell or an immune cell that has increased expression, level, and/or activity of a TAM kinase and/or c-Met kinase that include administering a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof or a pharmaceutical composition thereof to the patient. In some embodiments, the method further includes administering to the patient that at least one additional anticancer agent.
[00742] Also provided herein are methods of treating a patient identified or diagnosed as having a cancer that include: (a) selecting a patient identified or diagnosed as having increased expression, level, and/or activity of a TAM kinase and/or c-Met kinase in a cancer cell or an immune cell; and (b) after (a) administering to the selected patient a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof or a pharmaceutical composition thereof. In some embodiments, step (b) further includes administering to the patient at least one additional anticancer agent (e.g., any of the additional anticancer agents described herein).
100743] Also provided are methods of treating a patient identified or diagnosed as having a cancer that include: (a) selecting a patient identified or diagnosed as having a cancer that has been previously administered one or more doses of an additional anticancer agent (e.g., any of the
122 additionaî anticancer agents described herein) and identified as having a cancer cell or an immune cell having increased expression, level, and/or activity of a TAM kinase and/or c-Met kinase; and (b) after (a), administering to the selected patient a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof or a pharmaceutical composition thereof. In some embodiments, step (b) further includes administering to the patient the at least one additionaî anticancer agent.
[00744] In some embodiments of any of the methods described herein, increased expression, level, and/or activity of a TAM kinase is detected in a cancer cell or an immune cell. In some embodiments of any of the methods described herein, the patient is identified or diagnosed as having a cancer cell or an immune cell having increased expression, level, and/or activity of a TAM kinase.
[00745] In some embodiments of any of the methods described herein, an increased expression, level, and/or activity of a TAM kinase and a c-Met kinase are detected in a cancer cell or an immune cell. In some embodiments of any of the methods described herein, the patient is identified or diagnosed as having a cancer cell or an immune cell having increased expression, level, and/or activity of a TAM kinase and a c-Met kinase.
[00746] In some embodiments of any of the methods described herein, the increased expression, level, and/or activity of a TAM kinase in a cancer cell or an immune cell results from a chromosomal translocation that results in the expression of a TREM87B-MERTK fusion protein or an AXL-MBIP fusion protein.
[00747] In some embodiments of any of the methods described herein, increased expression, level, and/or activity of a c-Met kinase is detected in a cancer cell or an immune cell. In some embodiments of any of the methods described herein, the patient is identified or diagnosed as having a cancer cell or an immune cell having increased expression, level, and/or activity of a cMet kinase.
[00748] Also provided are methods of treating a patient identified or diagnosed as having a TAM-associated cancer that include: (a) administering to the patient identified or diagnosed as having a TAM-associated cancer one or more doses of a TAM kinase inhibitor; (b) after (a), detecting résistance of the TAM-associated cancer in the patient to the TAM kinase inhibitor; and (c) after (b), administering to the patient a therapeutically effective amount of a compound of Formula 1 or a pharmaceutically acceptable sait thereof or a pharmaceutical composition thereof.
123
In some embodiments, step (c) further includes administering to the patient at least one additional anticancer agent (e.g., any of the additional anticancer agents described herein).
[00749] Also provided are methods of treating a patient identified or diagnosed as having a TAM-associated cancer that include: (a) detecting résistance of the TAM-associated cancer in the patient to a TAM kinase inhibitor that was previously administered to the patient; and (b) after (a), administering to the patient a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof or a pharmaceutical composition thereof. In some embodiments, step (b) further includes administering to the patient at least one additional anticancer agent.
[00750] Also provided herein are methods of treating a patient identified or diagnosed as having a TAM-associated cancer and determined to hâve a previously developed résistance to a TAM kinase inhibitor that include administering to the patient a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof or a pharmaceutical composition thereof. Some embodiments of these methods further include administering to the patient at least one additional anticancer agent (e.g., any of the additional anticancer agents described herein or known in the art).
[00751] Also provided herein are methods of treating a patient identified or diagnosed as having a c-Met-associated cancer that include: (a) administering to the patient identified or diagnosed as having a c-Met-associated cancer one or more doses of a c-Met kinase inhibitor; (b) after (a), detecting résistance of the c-Met-associated cancer in the patient to the c-Met kinase inhibitor; and (c), after (b), administering to the patient a therapeutically effective amount of a compound of Formula 1 or a pharmaceutically acceptable sait thereof or a pharmaceutical composition thereof. Some embodiments of these methods further include administering to the patient at least one additional anticancer agent (e.g., any of the additional anticancer agents described herein or known in the art). In one embodiment, the c-Met inhibitor administered in step (a) is a Type 1 c-Met inhibitor. In one embodiment, the Type 1 c-Met inhibitor is crizotinib, capmatinib, NVP-BVU972, AMG 337, bozitinib, glumetinib, savolitinib, or tepotinib.
[00752] Also provided herein are methods of treating a patient identified or diagnosed as having a c-Met-associated cancer that include: (a) detecting résistance of the c-Met-associated cancer in the patient to a c-Met kinase inhibitor that was previously administered to the patient; and (b) after (a), administering to the patient a therapeutically effective amount of a compound of Fonnula I or
124 a pharmaceutically acceptable sait thereof or a pharmaceutical composition thereof. Some embodiments of these methods, step (b) further includes administering to the patient at least one additional anticancer agent. In one embodiment, the c-Met inhibitor administered in step (a) is a Type I c-Met inhibitor. In one embodiment, the Type 1 c-Met inhibitor is crizotinib, capmatinib, NVP-BVU972, AMG 337, bozitinib, glumetinib, savolitinib, ortepotinib.
[00753] Also provided herein are methods of treating a patient identified or diagnosed as having a c-Met-associated cancer and determined to hâve previously developed résistance to a c-Met kinase inhibitor that include administering to the patient a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof or a pharmaceutical composition thereof. Some embodiments of these methods further include administering to the patient at least one additional anticancer agent. In one embodiment, the patient developed résistance to a Type I c-Met inhibitor. In one embodiment, the Type 1 c-Met inhibitor is crizotinib, capmatinib, NVP-BVU972, AMG 337, bozitinib, glumetinib, savolitinib, or tepotînib.
[00754] In some embodiments of any of the methods described herein, the step of identifying the patient as having a TAM-associated cancer and/or a c-Met-associated cancer includes performing an assay on a biopsy sample obtained from the patient. In some embodiments, the assay is selected from the group of sequencing, immunohistochemistry, enzyme-linked immunosorbent assay, and fluorescence in situ hybridization (FISH). In some embodiments, the assay is selected from the group of: denaturing gradient gel electrophoresis (DGGE), température gradient electrophoresis (TGGE), température gradient capillary electrophoresis, a single strand conformational polymorphism assay, a molecular beacon assay, a dynamic hybridization assay, a PCR-based assay and denaturing high performance liquid chromatography. Some embodiments of these methods can further include obtaining the biopsy sample from the patient.
[00755] In some embodiments of any of the methods described herein, a compound of Formula I is selected from the compounds described in Example Nos. 1-201, or pharmaceutically acceptable salts thereof. In some embodiments, a compound of Formula I is selected from i) Example Nos. 1-20; ii) Example Nos. 21-40; iii) Example Nos. 41-60; iv) Example Nos. 61-80; v) Example Nos. 81-100; vi) Example Nos. 101-120; vii) Example Nos. 121-140; viii) Example Nos. 141-160; ix) Example Nos. 161-180; x) Example Nos. 181-201 or pharmaceutically acceptable salts thereof.
[00756] The compounds and methods described herein are useful for the treatment of
125 tumors and cancers (e.g., TAM-associated cancers and/or c-Met-associated cancers). The TAMassociated cancer and/or c-Met-associated cancer treated can be a primary tumor or a metastatic tumor. In one aspect, the methods described herein are used to treat a solid TAM-associated tumor, for example, melanoma, lung cancer (including lung adenocarcinoma, basal cell carcinoma, squamous cell carcinoma, large cell carcinoma, bronchioloalveolar carcinoma, bronchiogenic carcinoma, ποη-small-cell carcinoma, small cell carcinoma, mesothelioma); breast cancer (including ductal carcinoma, lobular carcinoma, inflammatory breast cancer, clear cell carcinoma, mucinous carcinoma, serosal cavities breast carcinoma); colorectal cancer (colon cancer, rectal cancer, colorectal adenocarcinoma); anal cancer; pancreatic cancer (including pancreatic adenocarcinoma, islet cell carcinoma, neuroendocrine tumors); prostate cancer; prostate adenocarcinoma; urinary tract cancer; ovarian cancer or carcinoma (ovarian épithélial carcinoma or surface epithelial-stromal tumor including serous tumor, endometrioid tumor and mucinous cystadenocarcinoma); liver and bile duct carcinoma (including hepatocellular carcinoma, cholangiocarcinoma, hemangioma); esophageal carcinoma or cancer (including esophageal adenocarcinoma and squamous cell carcinoma); oral and oropharyngeal squamous cell carcinoma; salivary gland adenoid cystic carcinoma: bladder cancer; bladder carcinoma; carcinoma of the utérus (including endométrial cancer or endométrial adenocarcinoma, ocular, uterine papillary serous carcinoma, uterine clear-cell carcinoma, uterine sarcomas and leiomyosarcomas, mixed mullerian tumors); glioma, glioblastoma, medulablastoma, and other tumors of the brain; kidney cancers (including rénal cancer, rénal cell carcinoma, clear cell carcinoma, Wilm's tumor); pituitary adenoma; cancer of the head and neck (including squamous cell carcinomas); cancer of the stomach (gastric cancers, stomach adenocarcinoma, gastrointestinal stromal tumor (GIST)); testicular cancer; germ cell tumor; neuroendocrine tumor; cervical cancer; carcinoids of the gastrointestinal tract, breast, and other organs; signet ring cell carcinoma; mesenchymal tumors including sarcomas (e.g., Kaposi’s sarcoma), fibrosarcomas, haemangioma, angiomatosis, haemangiopericytoma, pseudoangiomatous stromal hyperplasia, myofibroblastoma, fïbromatosis, inflammatory myofibroblastic tumor, lipoma, angiolipoma, granular cell tumor, neurofibroina, schwannoma, angiosarcoma, liposarcoma, rhabdomyosarcoma, osteosarcoma, leiomyoma, leiomysarcoma, skin (e.g., squamous cell skin cancer), including melanoma, cervical, retinoblastoma, head and neck cancer, pancreatic, brain, thyroid, testicular, rénal, bladder, soft tissue, adrenal gland, urethra, cancers of the pénis, myxosarcoma, chondrosarcoma, osteosarcoma,
126 chordoma, malignant fibrous histiocytoma, lymphangiosarcoma, mesothelioma, squamous cell carcinoma; epidermoid carcinoma, malignant skin adnexal tumors, adenocarcinoma, hepatoma, hepatocellular carcinoma, rénal cell carcinoma, hypernephroma, cholangiocarcinoma, transîtional cell carcinoma, choriocareinoma, seminoma, embryonal cell carcinoma, glioma anaplastic; glîoblastoma multiforme, neuroblastoma, medulloblastoma, malignant meningioma, malignant schwannoma, neurofibrosarcoma, parathyroid carcinoma, medullary carcinoma of thyroid, bronchial carcinoid, pheochromocytoma, Islet cell carcinoma, malignant carcinoid, malignant paraganglioma, melanoma, Merkel cell neoplasm, eystosarcoma phylloide, salivary cancers, thymie carcinomas, and cancers of the vagina among others.
[00757] The compounds of Formula I or pharmaceutically acceptable salts thereof can also be used for treating lymphoma or lymphocytic or myelocytic prolifération disorder or abnormality (e.g., a TAM-associated lymphoma or lymphocytic or myelocytic prolifération disorder or abnormality). For example, the TAM-associated cancer can be a Hodgkin Lymphoma of a NonHodgkin Lymphoma. For example, the subject can be suffering from a TAM-associated NonHodgkin Lymphoma such as, but not limited to: an AIDS-Related Lymphoma; Anaplastic LargeCell Lymphoma; Angioimmunoblastic Lymphoma; Blastic N -Cell Lymphoma: Burkitt's Lymphoma: Burkitt-like Lymphoma (Small Non-Cleaved Cell Lymphoma); Chrome Lymphocytic Leukemia/Small Lymphocytic Lymphoma: Cutaneous T-Cell Lymphoma; Diffuse Large B-Cell Lymphoma; Entcropathy-Type T-Cell Lymphoma; Follicular Lymphoma; Hepatosplenic Gamma-Delta T-Cell Lymphoma; Lymphoblastic Lymphoma: Mantle Cell Lymphoma; Marginal Zone Lymphoma; Nasal T-Cell Lymphoma; Pédiatrie Lymphoma; Peripheral T-Cell Lymphomas; Primary Central Nervous System Lymphoma; T-Cell Leukemias; Transformed Lymphomas; Treatment-Related T-Cell Lymphomas; or Waldenstrom's Macroglobuliuemia.
[00758] Alternatively, the subject may be suffering from a TAM-associated Hodgkin Lymphoma, such as, but not limited to: Nodular Sclerosis Classical Hodgkin's Lymphoma (CHL); Mixed Cellularity CHL; Lymphocyte-depletion CHL; Lymphocyte-rich CHL; Lymphocyte Prédominant Hodgkin Lymphoma; or Nodular Lymphocyte Prédominant HL.
[00759| In one embodiment, the methods as described herein may be useful to treat a patient suffering from a spécifie TAM-associated T-cell, a B-cell, or a NK-cell based lymphoma, proliférative disorder, or abnormality. For example, the patient can be suffering from a spécifie
127
TAM-associated T-cell or NK-cell lymphoma, for example, but not lirnited to: Peripheral T-cell lymphoma, for example, peripheral T-cell lymphoma and peripheral T-cell lymphoma not otherwise specified (PTCL-NOS); anaplastic large cell lymphoma, for example anaplastic lymphoma kinase (ALK) positive. ALK négative anaplastic large cell lymphoma, mantle cell 5 lymphoma, or primary cutaneous anaplastic large cell lymphoma; angioimmunoblastic lymphoma;
cutaneous T-cell lymphoma, for example mycosis fungoides, Sezary syndrome, primary cutaneous anaplastic large cell lymphoma, primary cutaneous CD30+ T-cell lymphoproliférative disorder;
primary cutaneous aggressive epidermolropic CD8+ cytotoxic T-cell lymphoma; primary cutaneous gamma-delta T-ceil lymphoma; primary cutaneous small/medium CD4+ T-cell 10 lymphoma, and lymphomatoid papulosis; Adult T-cell Leukemia/Lymphoma (ATLL); Blastic
NK-cell Lymphoma: Enteropathy-type T-cell lymphoma; Hematosplenic gamma-delta T-cell Lymphoma: Lymphoblastic Lymphoma; Nasal NKÆ-cell Lymphomas; Treatment-related T-celi lymphomas; for example lymphomas that appear after solid organ or bone marrow transplantation; T-cell prolymphocyte leukemia; T-cell large granular lymphocytic leukemia; Chronic 15 lymphoproliférative disorder of NK-cells; Aggressive NK cell leukemia; Systemic EBV+ T-cell lymphoproliférative disease of childhood (associated with chronic active EBV infection); Hydroa vacciniforme-like lymphoma; Adult T-cell leukemia/ lymphoma; Enteropathy-associated T-cell lymphoma; Hepatosplenic T-cell lymphoma; or Subcutaneous panniculitis-like T-cell lymphoma. [00760] In one embodiment, the methods as described herein may be useful to treat a patient 20 suffering from a spécifie TAM-associated B-cell lymphoma or proliférative disorder such as, but not lirnited to: multiple myeloma; Diffuse large B cell lymphoma; Follicular lymphoma; MucosaAssociated Lymphatic Tissue lymphoma (MALT); Small cell lymphocytic lymphoma; Mantle cell lymphoma (MCL); Burkitt lymphoma; Médiastinal large B cell lymphoma; Waldenstrom macroglobulinemia; Nodal marginal zone B cell lymphoma (NMZL); Splenic marginal zone lymphoma (SMZL); Intravascular large B-cell lymphoma; Primary effusion lymphoma; or Lymphomatoid granulomatosis; Chronic lymphocytic leukemia/small lymphocytic lymphoma; Bcell prolymphocyte leukemia; Hairy cell leukemia; Splenic lymphoma/leukemia, unclassifiable; Splenic diffuse red pulp small B-cell lymphoma; Hairy cell leukemia-variant; Lymphoplasmacytic lymphoma; Heavy chain diseases, for example, Alpha heavy chain disease, Gamma heavy chain disease, Mu heavy chain disease; Plasma cell myeloma; Solitary plasmacytoma of bone;
Extraosseous plasmacytoma; Primary cutaneous follicle center lymphoma; cell/histiocytic rich
128 large B-cell lymphoma; DLBCL associated with chronic inflammation; Epstein-Barr virus (EBV)-i- DLBCL of the elderly; Primary médiastinal (thymie) large B-cell lymphoma; Primary cutaneous DLBCL, leg type; ALK+ large B-cell lymphoma; Plasmablastic lymphoma; Large Bcell lymphoma arising in HHV8-associated multicentric; Castleman disease; B-cell lymphoma, unclassifiable, with features intermediate between diffuse large B-cell lymphoma and Burkitt lymphoma; B-cell lymphoma, unclassifiable, with features intermediate between diffuse large Bcell lymphoma and classical Hodgkin lymphoma; Noduiar sclerosis classical Hodgkin lymphoma; Lymphocyte-rich classical Hodgkin lymphoma; Mixed cellularity classical Hodgkin lymphoma; or Lymphocyte-depleted classical Hodgkin lymphoma.
[007611 In one embodiment, the methods as described herein may be useful to treat a patient suffering from a TAM-associated leukemia. For example, the subject may be suffering from an acute or chronic TAM-associated leukemia of a lymphocytic or myelogenous origin, such as, but not limited to: Acute iymphoblastic leukemia (ALL); Acute myelogenous leukemia (AML); Chronic lymphocytic leukemia (CLL); Chronic myelogenous leukemia (CML); juvénile myelomonocytic leukemia (JMML); hairy cell leukemia (HCL); acute promyélocyte leukemia (a sublype of AML); T-cell prolymphocyte leukemia (TPLL); large granular lymphocytic leukemia; or Adult T-cell chronic leukemia; large granular lymphocytic leukemia (LGL). In one embodiment, the patient suffers from an acute myelogenous leukemia, for example an undifferentiated AML (MO); myeloblasts leukemia (Ml ; with/without minimal cell maturation); myeloblastic leukemia (M2; with cell maturation); promyelocytic leukemia (M3 or M3 variant [M3V]); myelomonocytic leukemia (M4 or M4 variant with eosinophîlîa [M4E]); monocytic leukemia (M5); erythroleukemia (M6); or megakaryoblastic leukemia (M7).
[00762| In one embodiment, the compounds and methods described herein are useful for treating a TAM-associated cancer in a patient, wherein the cancer overexpresses AXL, MER, or TYRO3, or a combination thereof, e.g., as compared to a control non-cancerous tissue or a control cell (e.g., from the same or a different subject). In one embodiment, the cancer overexpresses AXL. In one embodiment, the cancer overexpresses MER. In an alternative embodiment, the cancer ectopically expresses MER. In one embodiment, the TAM-associated cancer is breast, colon, rénal, skin, lung (including non-small cell lung cancer), liver, gastric, brain (including glioblastoma), ovarian, pancreatic, prostate, glioblastoma multiforme, osteosarcoma, thyroid maiignancies, rhabdomyosarcoma, melanoma. acute myeloid leukemia, T-cell acute lymphoid
129 leukemia, B-cell acute lymphoid leukemia, schwannoma, and mantle cell lymphoma.
[00763] In one embodiment, the TAM-associated cancer is selected from breast, colon, rénal, skin, lung (including non-small cell lung cancer), liver, gastric, brain (including glioblastoma), ovarian, pancreatic, prostate, glioblastoma multiforme, osteosarcoma, thyroid 5 malignancies, rhabdomyosarcoma, and melanoma.
[00764] In one embodiment, the TAM-associated cancer is selected from leukemias (including acute myeloid leukemia and chronic myeloid leukemia, B-cell myeloid leukemia (BCLL), B-cell acute lymphoblastic leukemia, erythroid leukemia, and T-lineage acute lymphoblastîc leukemia), non-small cell lung cancer, pancreatic ductal adenocarcinoma, 10 astrocytoma, lung adenocarcinoma, ovarian cancer, melanoma, and glioblastoma multiforme.
[00765] In one embodiment, the TAM-associated cancer is selected from chronic myeloid leukemia, gastrointestinal stromal tumors (GIST), breast cancer (e.g.,HER2 positive breast cancer and triple négative breast cancer), head and neck cancer, and non-small cell lung cancer.
[00766] In some embodiments of any of the methods described herein, the TAM-associated 15 cancer is a cancer having overexpression of a TAM kinase, e.g., as compared to a non-cancerous tissue or cell in the same patient or a different subject. In some embodiments of any of the methods described herein, the TAM-associated cancer is a cancer having ectopic expression of a TAM kinase.
[00767] In some embodiments of any of the methods described herein, the TAM-associated 20 cancer is a cancer having overexpression or ectopic expression of a TYRO3 protein. In some embodiments of any of the methods described herein, the TAM-associated cancer has one or more point mutations in a gene encoding TYRO3 that results in the expression of a TYRO3 that includes one or more amino acid substitutions. In some embodiments of any of the methods described herein, the TAM-associated cancer has a chromosomal translocation which results in the 2 5 expression of a fusion protein including the kinase domain of TYRO3 and a fusion partner. Nonlimiting examples of a TAM-associated cancer having overexpression or ectopic expression of TYRO3, or a mutation in a TYRO3 gene that results in the expression of TYRO3 having one or more point mutations or a TYRO3 fusion protein include: AML, multiple myeloma, lung cancer, melanoma, prostate cancer, endométrial cancer, thyroid cancer, schwannoma, pancreatic cancer, 3 0 and brain cancer. Non-limiting aspects of TAM-associated cancers having increased expression and/or actîvity of TYRO3 are listed in Table 4.
130
Table 4. TAM-Associated Cancers Having with Increased Expression and/or Activity of
TYRO3
Melanoma Amino acid substitutions at: Q67 and/or R462Q, and/or W708fs*5
Lung Cancer Amino acid substitution at E340 or N615K in TYRO3
Pancreatic Cancer Amino acid substitution R514Q in TYRO3
Colon Cancer Amino acid substitution G809D and/or M592I in TYRO3
Brain Cancer Amino acid substitution A709T in TYRO3
AML, multiple myeloma, lung cancer, melanoma, prostate cancer, endométrial cancer, thyroid cancer, and schwannoma Overexpression or ectopic expression of TYRO3
[00768] Additional anticancer agents that are TYRO3 inhibitors include, e.g., 6g, merestinib (LY2801653), ASLAN002 (BMS-777607; (N-[4-(2-Amino-3-chloropyridin-4-yloxy)-3fluorophenyl]-4-ethoxy-l-(4-fluorophenyl)-2-oxo-l,2-dihydropyridine-3-carboxamide),
LDC1267 (N-[4-[(6,7-Dimethoxyqumolin-4-yl)oxy]-3-fluorophenyl]-4-ethoxy-l-(4-fluoro-2methylphenyl)-lH-pyrazole-3-carboxamide hydrochloride, and UNC2025 (trans-4-[210 (Butylamino)-5-[4-[(4-methylpiperazin-l-yl)methyl]phenyl]-7H-pyrrolo[2,3-d]pyrimidin-7yl]cyclohexanol).
[00769] In some embodiments of any of the methods described herein, the TAM-associated cancer is a cancer having overexpression or ectopic expression of a AXL protein. In some embodiments of any of the methods described herein, the TAM-associated cancer has one or more 15 point mutations in a gene encodîng AXL that results in the expression of a AXL that includes one or more amino acid substitutions. In some embodiments of any of the methods described herein, the TAM-associated cancer has a chromosomal translocation which results in the expression of a fusion protein including the kinase domain of AXL and a fusion partner. Non-limiting examples of a TAM-associated cancer having overexpression or ectopic expression of AXL, or a mutation
131 în a AXL gene that results in the expression of AXL having one or more point mutations or a AXL fusion protein include: AML, CML, B-CLL, lung cancer, glioblastoma, breast cancer, colorectal cancer, gastric cancer, pancreatic cancer, oesophageal cancer, melanoma, squamous cell skin cancer, prostate cancer, endométrial cancer, ovarian cancer, oral squamous cell carcinoma, thyroid cancer, bladder cancer, rénal cancer, schwannoma, mesothelioma, Kaposi’s sarcoma, osteosarcoma, erythroid leukemia, colon cancer, liver cancer, rénal cell carcinoma, osteosarcoma, kidney cancer, PH+ CML, non-small cell lung cancer, triple-negative metastatic breast cancer, and HER2+ breast cancer. Non-limiting aspects of TAM-associated cancers having increased expression and/or activity of AXL are Iisted în Table 5.
Table 5. TAM-Associated Cancers Having with Increased Expression and/or Activity of AXL
Ovarian Cancer Amino acid substitutions C24G and/or A358V in AXL
Melanoma One or more of the amino acid substitutions of P36L, R236C, G4I3W, E43IK, A451T, E535K, G829E, I610V, A666T, S685F, and R784Q in AXL
Colon Cancer One or more of the amino acid substitutions of N43T, M58OK, and L684P in AXL
Skin Cancer An amino acid substitution of P238L in AXL
Gastric Cancer One or more of the amino acid substitutions of V289M, R492C, S842F, and P636H in AXL
Lung Cancer One or more of the amino acid substitutions of R295W, L423Q, K526N, and S599F in AXL
Breast Cancer One or more of the amino acid substitutions of T343M, E745K, and S747R in AXL
Prostate Cancer An amino acid substitution of R368Q in AXL
Pancreatic Cancer An amino acid substitution of E484D in AXL
Kidney Cancer An amino acid substitution of P742T in AXL
AML, CML, B-CLL, lung cancer, Overexpression or ectopic expression of AXL
132
glioblastoma, breast cancer, colorectal cancer, gastric cancer, pancreatic cancer, esophageal cancer, melanoma, squamous cell skin cancer, prostate cancer, endométrial cancer, ovarian cancer, oral squamous cell carcinoma, thyroid cancer, bladder cancer, rénal cancer, schwannoma, mesothelioma, Kaposi’s sarcoma, and osteosarcoma
[00770] Additional anticancer agents that are AXL inhibitors include, e.g., bozitinib (SK1606, PF-5208765, Bosulif), Bemcentinib (BGB324; R428), crizotinib (PF-2341066, Xalkon), foretinib (GSK1363089, XL880), (N-[4-(2-Amino-3-chloropyridin-4-yloxy)-3-f1uorophenyl]-45 ethoxy-l-(4-fluorophenyl)-2-oxo-l,2-dihydropyridine-3-carboxamide (BMS-7776Û7;
ASLAN002), LY2801653 (merestinib), amuvatinib (MP-470), cabozantinib (XL184, BMS907351, Cometriq), glesatinib (MGCD265), NPS-1034 (2-(4-Fluorophenyl)-N-[3-fluoro-4-(3phenyl-lH-pyrrolo[2,3-b]pyridin-4-yloxy)phenyl]-l,5-dimethyl-3-oxo-2,3-dihydro-lH-pyrazole4-carboxamîde), LDC1267 (N-[4-[(6,7-Dimethoxyquinolin-4-yl)oxy]-3-fluorophenyI]-4-ethoxy10 l-(4-fluoro-2-methylphenyl)-lH-pyrazole-3-carboxamide hydrochloride), gilteritinib (ASP22I5), SG1-7079 ([3-(2-[[3-Fluoro-4-(4-methylpiperazin-l-yl)phenyl]amino]-5-methyl-7H-pynOlo[2,3d]pyrimidin-4-yl)phenyl]acetonitrile), dubermatinib (TP-0903), trans-4-[2-(Butyiamino)-5-[4-[(4methylpiperazin-l-yl)methyl]phenyl]-7H-pynrolo[2,3-d]pyr!midin-7-yl]cyciohexanol (UNC2025), 3-[3-[4-(Morpholin-4-ylmethyl)-l H-pyrrol-2-ylmethylene]-2-oxo-2,3-dihydro-lH15 indol-5-ylmethyl]thiazolidine-2,4-dione hydrochloride (S49076), sunitinib (SU11248, Sutent), and the monoclonal antibodies of 12A11, Mabl 73, YW327.6S2, D9, and E8.
[00771] In some embodiments of any of the methods described herein, the TAM-associated cancer is a cancer having overexpression or ectopic expression of a MER protein. In some embodiments of any of the methods described herein, the TAM-associated cancer has one or more
0 point mutations in a gene encodîng MER that results in the expression of a MER that includes one or more amino acid substitutions. In some embodiments of any of the methods described herein, the TAM-associated cancer has a chromosomal translocation which results in the expression of a fusion protein including the kinase domain of MER and a fusion partner. Non-Iimiting examples
133 of a TAM-associated cancer having overexpression or ectopic expression of MER, or a mutation in a MER gene that results in the expression of MER having one or more point mutations or a MER fusion protein include: AML, ALL (B-ALL, T-ALL), lung cancer, glioma, melanoma, prostate cancer, schwannoma, mantle cell lymphoma, rhabdomyosarcoma, pancreatic cancer, 5 breast cancer, gastric cancer, pituitary adenoma, urinary tract cancer, kidney cancer, liver cancer, colon cancer, and breast cancer. Non-limiting aspects of MER-associated cancers having increased expression and/or activity of MER are listed in Table 6.
Table 6. TAM-Associated Cancers Having with Increased Expression and/or Activity of MER
Melanoma One or more amino acid substitutions of P40S, V8611, K923R, and P802S in MER
Lung Cancer One or more amino acid substitutions of S159F, 1431 F, S905F, P672S, N718Y, and M790V in MER
Urinary Tract Cancer One or more amino acid substitutions of E204K, L586F, and S626C in MER
Gastric Cancer An amino acid substitutions of S428G in MER
Kidney Cancer Amino acid substitutions of A446G and/or P958L in MER
Liver Cancer One or more amino acid substitutions of N454S, V873I, and D983N in MER
Lymphoma An amino acid substitution of W485S/C in MER
Colon Cancer One or more amino acid substitutions of D990N, L688M, and R722 in MER
Breast Cancer An amino acid substitution of G594R in MER
Head and Neck Cancer An amino acid substitution of A708S in MER
AML, ALL, lung cancer, glioma, melanoma, prostate cancer, schwannoma, mantle cell lymphoma, and rhabdomyosarcoma Overexpression or ectopic expression of MER
134
[00772] Additional anticancer agents that are MER inhibitors include, e.g., foretinib, merestinib (LY2801653), (N-[4-(2-Amino-3-chloropyridin-4-yloxy)-3-fluorophenyl]-4-ethoxy-l(4-fluorophenyl)-2-oxo-l,2-dihydropyridine-3-carboxamide (ASLAN002; BMS-777607), [3-(2[[3-Fluoro-4-(4-methylpiperazin-I-yl)phenyl]amino]-5-methyl-7H-pyrrolo[2,3-d]pyrimidin-4yl)phenyl]acetonitrile (SGI-7079), dubermatinib (TP-0903), trans-4-[2-(Butylamino)-5-[4-[(4methylpiperazin-l-yl)methyl]phenyl]-7I-I-pyrrolo[2,3-d]pynmidîn-7-yl]cyclohexanol (UNC2025), and 3-[3-[4-(Morpholin-4-ylmethyl)-lH-pyrrol-2-ylmethylene]-2-oxo-2,3-dihydro1 H-indoI-5-ylmethyl]thiazolidine-2,4-dione hydrochloride (S49076).
[00773] Also provided are methods for treating a cancer (e.g., a TAM-associated cancer and/or c-Met-associated cancer) in a patient in need thereof, the method comprising: (a) determining if the cancer in the patient is a TAM-associated cancer, a c-Met-associated cancer, or both; and (b) if the cancer is determined to be a TAM-associated cancer, a c-Met-associated cancer, or both, administering to the patient a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof or a pharmaceutical composition thereof. Some embodiments of these methods further include administering to the subject at least one additional anticancer agent (e.g., an immunotherapy). In some embodiments, the subject was previously treated with at least one additional anticancer agent or therapy, e.g., a kinase inhibitor, an immunotherapy, chemotherapy, radiation therapy and/or surgery. In some embodiments the patient has a cancer that is résistant to the at least one additional anticancer agent. In one embodiment, the at least one additional anticancer agent does not include a compound of Formula I.
[00774] Also provided herein is a method for treating a patient diagnosed with or identifîed as having a TAM-associated cancer ( e.g., any of the exemplary TAM-associated cancers disclosed herein), a c-Met-associated cancer (e.g., any of the exemplary c-Met-associated cancers disclosed herein), or both, comprising administering to the patient a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition thereof as defined herein. Some embodiments of these methods further include administering to the subject at least one additional anticancer agent (e.g., an immunotherapy). In some embodiments, the subject was previously treated with at least one additional anticancer agent or therapy, e.g., a kinase inhibitor, an immunotherapy, chemotherapy, radiation therapy and
135 or surgery. In one embodiment the patient has a cancer that is résistant to the previously administered at least one additionaî anticancer agent. In some embodiments, the ai least one additionaî anticancer agent does not include a compound of Formula I.
[007751 In one embodiment, provided herein are methods for treating a patient diagnosed 5 with (or identified as having) a cancer (e.g., a TAM-associated cancer, a c-Met-associated cancer, or both) that include administering to the patient a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof. Also provided herein are methods for treating a patient identified or diagnosed as having a TAM-associated cancer, a c-Met-associated cancer, or both, that include administering to the patient a therapeutically effective amount of a 10 compound of Formula I or a pharmaceutically acceptable sait thereof or a pharmaceutical composition thereof. In some embodiments, the patient that has been identified or diagnosed as having a TAM-associated cancer, a c-Met-associated cancer, or both, through the use of a regulatory agency-approved, e.g., FDA-approved test or assay for identifying abnormal (e.g., increased) expression, level, and/or activity of one or more of the TAM kinases and/or c-Met 15 kinase, in a patient or a biopsy sampie from the patient or by performing any of the non-limiting examples of assays described herein. In some embodiments, the test or assay is provided as a kit. Some embodiments of these methods further include administering to the patient at least one additionaî anticancer agent (e.g., an immunotherapy). In some embodiments, the patient was previously treated with at least one additionaî anticancer agent or therapy, e.g., a kinase inhibitor, 20 an immunotherapy (e.g., an immune checkpoint inhibitor), chemotherapy, radiation therapy and or surgery. In some embodiments, the patient has a cancer that is résistant to the at least one additionaî anticancer agent. In some embodiments, the at least one additionaî anticancer agent does not include a compound of Formula I.
[00776| Also provided is a compound of Formula I or a pharmaceutically acceptable sait 25 thereof, for use in the treatment of a cancer in a patient in need thereof or a patient identified or diagnosed as having a TAM-associated cancer (e.g., any of the TAM-associated cancers described herein), a c-Met-associated cancer (e.g., any of the c-Met-associated cancers described herein), or both. Also provided is the use of a compound of Formula I or a pharmaceutically acceptable sait thereof for the manufacture of a médicament for treating a cancer in a patient identified or 30 diagnosed as having a TAM-associated cancer, a c-Met-associated cancer, or both. In some embodiments, the cancer is a TAM-associated cancer. In some embodiments, the cancer is a c20534
136
Met-associated cancer. In some embodiments, the cancer is both a TAM-associated cancer and a c-Met-associated cancer. In some embodiments, a patient is identified or diagnosed as having a TAM-associated cancer, a c-Met-associated cancer, or both, through the use of a regulatory agency-approved, e.g., FDA-approved, kit for identifying abnormal (e.g., increased) expression, 5 level, and/or activity of one or more of the TAM kinases and/or c-Met kinase, e.g., as compared to a non-cancerous tissue or cell from the same or a different subject. Some embodiments of these methods further include administering to the patient at least one additional anticancer agent (e.g., immunotherapy). In some embodiments, the subject was previously treated with at least one additional anticancer agent or therapy, e.g., an immune checkpoînt inhibitor, a kinase inhibitor, an 10 immunotherapy, chemotherapy, radiation therapy and/or surgery. In some embodiments, the patient has a cancer that is résistant to one or more of the at least one additional anticancer agents. In one embodiment, the at least one additional anticancer agent does not include a compound of Formula I.
[00777] In some embodiments of any of the methods or uses described herein, the patient 15 has been identified or diagnosed as having a cancer associated with or having abnormal (e.g., increased) expression, level, and/or activity of one or more of the TAM kinases and/or c-Met kinase, e.g., as compared to a non-cancerous tissue or cell in the same or a different subject. In some embodiments, provided herein are methods for treating a TAM-associated cancer, a c-Metassociated cancer, or both, in a patient in need of such treatment, the method comprising a) 20 detecting abnormal (e.g., increased) expression and/or activity of one or more of the TAM kinases and/or c-Met kinase, e.g., as compared to a non-cancerous tissue or cell in the same or a different subject; and b) after a), administering a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof. Some embodiments of these methods further include administering to the patient at least one additional anticancer agent (e.g., immunotherapy).
In some embodiments, the patient was previously treated with at least one additional anticancer agent or therapy, e.g., a kinase inhibitor, an immunotherapy, chemotherapy, radiation therapy and/or surgery. In some embodiments, the patient has a cancer that is résistant to one or more of the at least one additional anticancer agent. In one embodiment, the at least one additional anticancer agent does not include a compound of Formula I.
[00778] In some embodiments of any of the methods or uses described herein, the patient has a clinical record indicating that the patient has a tumor associated with or having abnormal
137 (e.g., increased) expression, level, and/or activity of one or more ofthe TAM kinases and/or c-Met kinase, e.g., as compared to a non-cancerous tissue or cell in the same patient or a different subject). In some embodiments, the clinical record indicates that the patient should be treated with one or more of the compounds of Formula I or a pharmaceutically acceptable salts thereof or compositions provided herein. Some embodiments of these methods further include administering to the patient at least one additional anticancer agent (e.g., an immunotherapy). In some embodiments, the subject was previously treated with at least one additional anticancer agent or therapy, e.g., a kinase inhibitor, an immunotherapy, chemotherapy, radiation therapy and/or surgery. In some embodiments, the patient has a cancer that is résistant to one or more of the at least one additional anticancer agent. In one embodiment, the at least one additional anticancer agent does not include a compound of Formula I.
[00779] Also provided are methods of treating a patient that include administering a therapeutically effective amount of a compound of Formula 1 or a pharmaceutically acceptable sait thereof to a patient having a clinical record that indicates that the patient has a cancer associated with or having abnormal (e.g., increased) expression, level, and/or activity of one or more of the TAM kinases and/or c-Met kinase, e.g., as compared to a non-cancerous tissue or cell from the patient or a different subject. Also provided is the use of a compound of Formula I or a pharmaceutically acceptable sait thereof for the manufacture of a médicament for treating a TAMassociated cancer, a c-Met-associated cancer, or both, in a patient having a clinical record that indicates that the patient has a cancer associated with or having abnormal (e.g., increased) expression, level, and/or activity of one or more of the TAM kinases and/or c-Met kinase, e.g., as compared to a non-cancerous tissue or cell from the patient or a different subject. Some embodiments of these methods and uses can further include: a step of performing an assay on a sample (e.g., a biopsy sample) obtained from the patient to détermine whether the patient has abnormal (e.g., increased) expression, level, and/or activity of one or more of the TAM kinases and/or c-Met kinase (e.g., as compared to a non-cancerous tissue or cell from the patient or a different subject), and recording the information in a patient’s clinical file (e.g., a computer readable medium) that the patient has been identified to hâve abnormal (e.g., increased) expression and/or activity of one or more of the TAM kinases and/or c-Met kinase. In some embodiments, the assay is an in vitro assay. Some embodiments of these methods further include administering to the patient at least one additional anticancer agent (e.g., an immunotherapy). In some
138 embodiments, the subject was previously treated with at least one additional anticancer agent or therapy, e.g., a kinase inhibitor, an immunotherapy, chemotherapy, radiation therapy and/or surgery. In some embodiments, the patient has a cancer that is résistant to one or more of the at least one additional anticancer agent. In one embodiment, the at least one additional anticancer agent does not include a compound of Formula I.
[00780] Also provided herein is a method of treating a patient in need thereof. The method includes performing an assay on a sample obtaîned from the patient to déterminé whether the subject has abnormal (e.g., increased) expression, level, and/or activity of one or more ofthe TAM kinases and/or c-Met kinase, e.g., as compared to a non-cancerous tissue or cell from the same patient or a different subject). The method also includes administering to a patient determined to hâve abnormal (e.g., increased) expression, level, and/or activity of one or more of the TAM kinases and/or c-Met kinase (e.g., as compared to a non-cancerous tissue or cell from the same patient or a different subject) a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof. Some embodiments of these methods further include administering to the patient at least one additional anticancer agent (e.g., an immunotherapy). In some embodiments, the patient was previously treated with at least one additional anticancer agent or therapy, e.g., a kinase inhibitor, an immunotherapy, chemotherapy, radiation therapy and/or surgery. In some embodiments, the patient has a cancer that is résistant to one or more of the at least one additional anticancer agent, in one embodiment, the at least one additional anticancer agent does not include a compound of Formula I.
[00781] Also provided are methods (e.g., in vitro methods) of selecting a treatment for a patient identified or diagnosed as having a TAM-associated cancer, a c-Met-associated cancer, or both. Some embodiments can further include administering the selected treatment to the patient identified or diagnosed as having a TAM-associated cancer, a c-Met-associated cancer, or both. For example, the selected treatment can include administration of a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof. Some embodiments can further include a step of performing an assay on a sample (e.g., a biopsy sample) obtained from the patient to déterminé whether the patient has abnormal (e.g., increased) expression, level, and/or activity of one or more of the TAM kinases and/or c-Met kinase (e.g., as compared to a non-cancerous tissue or cell from the same patient or a different subject), and
139 identîfying and diagnosing a patient determined to hâve abnormal (e.g., increased) expression, level, and/or activity of one or more of the TAM kinases and/or c-Met kinase, as having a TAMassociated cancer and/or c-Met-associated cancer, respectively. In some embodiments, the patient has been identified or diagnosed as having a TAM-associated cancer, a c-Met-associated cancer, 5 or both, through the use of a regulatory agency-approved, e.g., FDA-approved, kit for identifying abnormal (e.g., increased) expression, level, and/or activity of one or more of the TAM kinases and/or c-Met kinase in a patient or a biopsy sample from the patient. In some embodiments, the TAM-associated cancer is a cancer described herein or known în the art. In some embodiments, the c-Met-associated cancer is a cancer described herein or known in the art. In some embodiments, 10 the assay is an in vitro assay. For example, an assay that utilizes the next génération sequencing, immunohistochemistry, or break apart FISH analysis. In some embodiments, the assay is a regulatory agency-approved, e.g., FDA-approved, kit. Some embodiments of these methods further include administering to the patient at least one additional anticancer agent (e.g., an immunotherapy). Some embodiments of these methods further include administering to the 15 subject at least one additional anticancer agent (e.g., an immunotherapy). In some embodiments, the patient was previously treated with at least one additional anticancer agent or therapy, e.g., an immune checkpoint inhibitor, a kinase inhibitor, an immunotherapy, chemotherapy, radiation therapy, and/or surgery. In some embodiments, the patient has a cancer that is résistant to one or more of the at least one additional anticancer agent. In one embodiment, the at least one additional
0 anticancer agent does not include a compound of Formula I.
[00782] Also provided herein are methods of selecting a treatment for a patient, wherein the methods include a step of performing an assay on a sample obtained from the patient to détermine whether the patient has abnormal (e.g., increased) expression, level, and/or activity of one or more 2 5 of the TAM kinases and/or c-Met kinase, e.g., as compared to a non-cancerous tissue or cell from the patient or a different subject. Some embodiments further include administering the selected treatment to the patient identified or diagnosed as having a TAM-associated cancer, a c-Metassociated cancer, or both. For example, the selected treatment can include administration of a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait 3 0 thereof to the patient identified or diagnosed as having a TAM-associated cancer, a c-Metassociated cancer, or both. In some embodiments, the assay is an in vitro assay. Some
140 embodiments of these methods further include administering to the patient at least one additional anticancer agent (e.g., an immunotherapy). In some embodiments, the patient was previously treated with at least one additional anticancer agent or therapy, e.g., an immune checkpoint inhibitor, a kinase inhibitor, an immunotherapy, chemotherapy, radiation therapy and/or surgery. In some embodiments, the patient has a cancer that is résistant to one or more of the at least one additional anticancer agent. In one embodiment, the at least one additional anticancer agent does not include a compound of Formula I.
[00783] Also provided are methods of selecting a patient for treatment, wherein the methods include selecting, identifying, or diagnosing a patient having a TAM-associated cancer, a c-Metassociated cancer, or both, and selecting the patient for treatment including administration of a therapeutically-effective amount of a compound of Formula 1 or a pharmaceutically acceptable sait thereof. In some embodiments, identifying or diagnosing a patient as having a TAMassociated cancer, a c-Met-associated cancer, or both, can include a step of performing an assay on a sample obtained from the patient to déterminé whether the patient has abnormal (e.g., increased) expression, level, and/or activity of one or more of the TAM kinases and/or c-Met kinase (e.g., as compared to a non-cancerous tissue or cell from the patient or a different subject), as having a TAM-associated cancer and/or c-Met-associated cancer, respectively. In some embodiments, the method of selecting a treatment can be used as a part of a clinical study that includes administration of various treatments of a TAM-associated cancer, a c-Met-associated cancer, or both. In some embodiments, the assay is an in vitro assay, Some embodiments of these methods further include administering to the subject at least one additional anticancer agent or therapy, e.g., an immune checkpoint inhibitor, a kinase inhibitor, an immunotherapy, chemotherapy, radiation therapy and/or surgery. In some embodiments, the patient has a cancer that is résistant to one or more of the at least one additional anticancer agent. In one embodiment, the at least one additional anticancer agent does not include a compound of Formula I.
[00784] In some embodiments of any of the methods or uses described herein, an assay can be used to détermine whether the patient has abnormal (e.g., increased) expression, level, and/or activity of one or more of the TAM kinases and/or c-Met kinase. In some embodiments, the sample is a biological sample or a biopsy sample (e.g., a paraffin-embedded biopsy sample) from the patient. In some embodiments, the patient is a patient suspected of having a TAM-associated cancer, a c-Met-associated cancer, or both, a patient having one or more symptoms of a TAM20534
141 associated cancer, a c-Met-associated cancer, or both, and/or a patient that has an increased risk of deveioping a TAM-associated cancer, a c-Met-associated cancer, or both),
[00785] In some embodiments of any the methods described herein, the compound of Formula I or a pharmaceutically acceptable sait thereof is administered in combination with a therapeutically effective amount of at least one additional anticancer agent selected from one or more additional thérapies or therapeutic agents, for example an agent that works by the sanie or by a different mechanism of action. In one embodiment, the compound of Formula I is selected from the compounds described in Example Nos. 1-201, or pharmaceutically acceptable salts thereof. In some embodiments, a compound of Formula I is selected from i) Exampîe Nos. 1-20; îi) Example Nos. 21-40; iii) Example Nos. 41-60; iv) Example Nos. 61-80; v) Example Nos. 81100; vi) Example Nos. 101-120; vii) Example Nos. 121-140; viii) Example Nos. 141-160; ix) Example Nos. 161-180; x) Example Nos. 181-201 or pharmaceutically acceptable salts thereof.
[00786] Non-limiting examples of additional anticancer agents include immune-targeted agents including immunotherapy agents, anti-viral agents, kinase-targeted therapeutic agents, anti-viral vaccines, anti-hormonal agents, signal transduction pathway inhibîtors, chemotherapeutics or other anti-cancer agents, angiogenesis inhibitors, and radiotherapy.
[00787] One or more of any of the additional anticancer agents described herein can be combined with the présent compounds in a single dosage form, or the présent compounds and the at least one additional anticancer agents can be administered simultaneously or sequentially as separate dosage forms.
[00788] In one embodiment, the compound of Formula I or a pharmaceutically acceptable sait thereof is administered daily for 28 consecutive days in a 28 days cycle.
[00789] In one embodiment, compounds of Formula I or pharmaceutically acceptable salts thereof may be combined with immune-targeted agents including immunotherapy drugs.
[00790] The term “immunotherapy agents” refers to an agent that modulâtes the immune System. In some embodiments, an immunotherapy can increase the expression and/or activity of a regulator of the immune System. In some embodiments, an immunotherapy can decrease the expression and/or activity of a regulator of the immune system. In some embodiments, an immunotherapy can recruit and/or enhance the activity of an immune cell.
[00791] In some embodiments, the immunotherapy agent is an immune checkpoint
142 inhibitor. As used herein, the terni immune checkpoint inhibitor or checkpoint inhibitor refers to molécules that totally or partially reduce, inhibit, interféré with, or modulate the expression and/or activity of one or more checkpoint proteins. In some embodiments, the immunotherapy includes one or more immune checkpoint inhibitors. In some embodiments, the immune checkpoint inhibitor is a CTLA-4 inhibitor (e.g., an anti-CTLA-4 antibody), a PD-1 inhibitor (e.g., an anti-PD-1 monoclonal antibody) or a PD-L1 inhibitor (e.g., an anti-PD-Ll monoclonal antibody). In some embodiments, the CTLA-4 inhibitor is ipilimumab (Yervoy®) or tremelimumab (CP-675,206). In some embodiments, the PD-1 inhibitor is pembrolizumab (Keytruda®), nivolumab (Opdivo®), or pidilizumab. In some embodiments, the anti-PD-l monoclonal antibody is nivolumab or pembrolizumab. In some embodiments, the anti-PDI antibody is pembrolizumab. In some embodiments, the PD-L1 inhibitor is atezolizumab (Tecentriq®), avelumab (Bavençîo®), durvalumab (Imfinzî™), MEDI4736, or MPDL3280A. In some embodiments, the PD-1 or PD-L1 inhibitor is a small molécule (e.g., those disclosed in US 2018/305313 and WO 2018/195321). In some embodiments, the PD-L1 inhibitor is atezolizumab (Tecentriq®), avelumab (Bavencio®), or durvalumab (Imfmzi™). In some embodiments, a checkpoint inhibitor can target 4-1BB (e.g., urelumab (BMS-663513) and PF-05082566 (PF2566)), CD27 (e.g., varlilumab (CDX-1127), CD40 (e.g., CP-870,893), OX40, TIM-3, ICOS, BTLA, A2AR, B7-H3, B7-H4, BTLA, IDO, KIR, LAG3, T1M-3, and VISTA. Additional nonlimiting examples of immune checkpoint inhibitors include ulocuplumab, urelumab, PF 05082566, TRX518, varlilumab, CP 870893, PDR001MEDI4736, avelumab, , BMS 986016, MGA271, IPH2201, emactuzumab, INCB024360, MEDI6469, galunisertib, BKT140, bavituximab, lirilumab, bevacizumab, MNRP1685A, lambroizumab, CC 90002, BMS-936559, and MGA271. [00792| In some embodiments, a compound of Formula I or pharmaceutically acceptable sait thereof is combined with an immune checkpoint inhibitor, wherein the immune checkpoint inhibitor is administered on one or more days in a 28 days cycle. In one embodiment the compound of Formula I or a pharmaceutically acceptable sait thereof is administered daily for 28 consecutive days in a 28 days cycle.
[00793] In some embodiments, a compound of Formula 1 or pharmaceutically acceptable sait thereof is combined with an immune checkpoint inhibitor, wherein the immune checkpoint inhibitor is administered one a week. In one embodiment, the immune checkpoint inhibitor is administered every two weeks. In one embodiment, the immune checkpoint inhibitor is
143 administered every three weeks. In one embodiment, the immune checkpoint inhibitor is administered every 4 weeks. In one embodiment, the immune checkpoint inhibitor is administered on day 1 of a 28 day cycle. In one embodiment, the immune checkpoint inhibitor is administered on days 1 and 7 in a 28 day cycle. In one embodiment, the immune checkpoint inhibitor is administered in days I, 7 and 14 in a 28 day cycle. In one embodiment, the immune checkpoint inhibitor is administered on days 1, 7,14 and 21 in a 28 day cycle. In one embodiment, the immune checkpoint inhibitor is administered on days 1, 7, 14 and 28 in a 28 day cycle. In one embodiment the compound of Formula I or a pharmaceutically acceptable sait thereof is administered daily for 28 consecutive days in a 28 days cycle. In one embodiment, the immune checkpoint inhibitor is administered by intravenous infusion.
[00794] In some embodiments, a compound of Formula I or pharmaceutically acceptable sait thereof is combined with an immune checkpoint inhibitor, wherein the immune checkpoint inhibitor is administered on day 1 of cycles 1 through 13. In one embodiment the compound of Formula I or a pharmaceutically acceptable sait thereof is administered daily for 28 consecutive days in a 28 days cycle.
[00795] In some embodiments, the immunotherapy agent is a cellular immunotherapy (e.g., adoptive T-cell therapy, dendritic cell therapy, or a natural killer cell therapy). In some embodiments, the cellular immunotherapy is sipuleucel-T (APC8015; Provenge™; Plosker(2011) Drugs 71(1): 101-108). In some embodiments, the cellular immunotherapy includes cells that express a chimeric antigen receptor (CAR). In some embodiments, the cellular immunotherapy is a CAR-T cell therapy. In some embodiments, the CAR-T cell therapy ts tisagenlecleucel (Kymriah™).
[00796] In some embodiments, the immunotherapy agent is an antibody therapy (e.g., a monoclonal antibody, a conjugated antibody). In some embodiments, the antibody therapy is bevacizumab (Mvasti™, Avastin®), trastuzumab (Herceptin®), avelumab (Bavencio®), rituximab (MabThera™, Rituxan®), edrecoiomab (Panorex), daratumuab (Darzalex®), olaratumab (Lartruvo™), ofatumumab (Arzerra®), alemtuzumab (Campath®), cetuximab (Erbitux®), oregovomab, pembrolizumab (Keytruda®), dinutiximab (Unituxin®), obinutuzumab (Gazyva®), tremelimumab (CP-675,206), ramucirumab (Cyramza®), ublituximab (TG-1101), panitumumab (Vectibix®), elotuzumab (Empliciti™), avelumab (Bavencio®), necitumumab (Portrazza™), cirmtuzumab (UC-961), ibritumomab (Zevalin®), isatuximab (SAR650984),
144 nimotuzumab, fresolimumab (GC1008), lirilumab (INN), mogamulizumab (Potelîgeo®), ficlatuzumab (AV-299), denosumab (Xgeva®), ganitumab, urelumab, pidilizumab, or amatuximab.
[00797] In some embodiments, the îmmunotherapy agent is an antibody-drug conjugate. In some embodiments, the antibody-drug conjugate is gemtuzumab ozogamicin (Mylotarg™), inotuzumab ozogamicin (Besponsa®), brentuximab vedotin (Adcetris®), ado-trastuzumab emtansine (TDM-1; Kadcyla®), mirvetuximab soravtansine (IMGN853), or anetumab ravtansine [00798] In some embodiments, the îmmunotherapy includes blinatumomab (AMG103; Blincyto®) or midostaurin (Rydapt).
[00799] In some embodiments, the îmmunotherapy agent includes a toxin. In some embodiments, the îmmunotherapy is denileukin diftitox (Ontak®).
[00800] In some embodiments, the îmmunotherapy agent is a cytokine therapy. In some embodiments, the cytokine therapy is an interleukin 2 (IL-2) therapy, an interferon alpha (IFNa) therapy, a granulocyte colony stimulating factor (G-CSF) therapy, an interleukin 12 (IL-12) therapy, an interleukin 15 (IL-15) therapy, an interleukin 7 (IL-7) therapy or an erythropoietinalpha (EPO) therapy. In some embodiments, the IL-2 therapy is aldesleukin (Proleukin®). In some embodiments, the IFNct therapy is IntronA® (Roferon-A®). In some embodiments, the GCSF therapy is filgrastim (Neupogen®).
[00801] In some embodiments, the îmmunotherapy agent is an inhibitory nucleic acid-based îmmunotherapy agent (e.g., antisense oligonucleotides, small interfering RNAs (siRNAs), and short hairpîn RNAs (shRNAs). In some embodiments, the inhibitory nucleic acid-based îmmunotherapy is CV9104 (see, e.g., Rausch et al, (2014) Human Vaccine Immunother. 10(11): 3146-52; and Kubler et al. (2015) J, Immunother. Cancer 3:26).
[00802] In some embodiments, the îmmunotherapy agent îs bacillus Calmette-Guerin (BCG) therapy. In some embodiments, the îmmunotherapy agent is an oncolytîc virus therapy. In some embodiments, the oncolytîc virus therapy is talimogene alherparepvec (T-VEC; Imlygic®). [00803] In some embodiments, the îmmunotherapy agent is a cancer vaccine. In some embodiments, the cancer vaccine is a human papillomavirus (HPV) vaccine. In some embodiments, the HPV vaccine is Gardasil®, Gardasil9® or Cervarix®. in some embodiments, the cancer vaccine is a hepatîtis B virus (HBV) vaccine. In some embodiments, the HBV vaccine is Engerix-B®, Recombivax HB® or GI-13020 (Tarmogen®). In some embodiments, the cancer
145 vaccine is Twinrix® or Pediarîx®. In some embodiments, the cancer vaccine îs BiovaxID®, Oncophage®, GVAX, ADXS11-001, AL VAC-CEA, PROSTVAC®, Rindopepimut®, CimaVaxEGF, lapuleucel-T (APC8024; Neuvenge™), GRNVAC1, GRNVAC2, GRN-1201, hepcortespenlisimut-L (Hepko-V5), DCVAX®, SC1B1, BMTCTN 1401, PrCa VB1R, PANVAC, ProstAtak®, DPX-Survivac, or viagenpumatucel-L (HS-110).
[00804] In some embodiments, the immunotherapy agent is a peptide vaccine. In some embodiments, the peptide vaccine is nelipepimut-S (E75) (NeuVax™), 1MA901, or SurVaxM (SVN53-67). In some embodiments, the cancer vaccine is an immunogenic personal neoantigen vaccine (see, e.g., Ott et al. (2017) Nature 547: 217-221; Sahin et al. (2017) Nature 547: 222-226). In some embodiments, the cancer vaccine is RGSH4K orNEO-PV-01. In some embodiments, the cancer vaccine is a DNA-based vaccine. In some embodiments, the DNA-based vaccine is a mammagiobin-A DNA vaccine (see, e.g., Kim et al. (2016) Oncolmmunology 5(2): el069940). [00805] In some embodiments, immune-targeted agents are selected from aldesleukin, interferon alfa-2b, ipilimumab, lambrolizumab, nivolumab, prednisone, and sipuleucel-T.
[00806] Suitable antiviral agents contemplated for use in combination with a compound of Formula I or a pharmaceutically acceptable sait thereof can comprise nucleoside and nucléotide reverse transcriptase inhibitors (RTIs), non-nucleoside reverse transcriptase inhibitors (NRTIs), protease inhibitors, and other antiviral drugs.
[00807] Example suitable NRTIs include zidovudine (AZT); didanosine (ddl); zalcitabine (ddC); stavudine (d4T); lamivudine (3TC); abacavir (1592U89); adefovir dipivoxil [bis(POM)PMEA]; lobucavir (BMS-180194); BCH-10652; emitricitabine [(-)-FTCJ; beta-L-FD4 (also called beta-L-D4C and named beta-L-2', 3'-dicleoxy-5-fluoro-cytidene); DAPD, ((-)-beta-D-2,6,diamino-purine dioxolane); and lodenosine (FddA). Typical suitable NNRTIs include nevirapine (BI-RG-587); delaviradine (BHAP, U-90152); efavirenz (DMP-266); PNU- 142721; AG-1549; MKC-442 (l-(ethoxy-methyl)-5-(l-methylethyl)-6-(phenylmethy 1)-(2,4(IH,3H)-pyrimidinedione); and (+)-calanolide A (NSC-675451) and B. Typical suitable protease inhibitors include saquinavir (Ro 31-8959); ritonavir (ABT-538); indinavir (MK-639); nelfnavir (AG-1343); amprenavir (14IW94); lasinavir (BMS-234475); DMP-450; BMS-2322623; ABT-378; and AG-1 549. Other antiviral agents include hydroxyurea, ribavirin, 1L-2, IL-12, pentafuside, and Yissum Project No. 11607.
[00808] Compounds of Formula I and pharmaceutically acceptable salts thereof can be used
146 in combination with one or more other kinase inhibitors for the treatment of diseases, such as cancer, that are impacted by one or more signaling pathways.
[00809] In certain embodiments, the patient to be treated with a combination therapy described herein has not been treated with an additional anticancer agent prior to the administration the combination therapy. In certain embodiments, the patient to be treated with a combination therapy described herein has been treated with at least one additional anticancer agent prior to administration of a compound of Formula 1 for use alone or in a combination therapy described herein. In certain embodiments, the patient to be treated with a compound of Formula I as monotherapy or in a combination therapy described herein has developed drug résistance to, or has a cancer that is refractory to, at least one additional anticancer agent.
[00810] In one embodiment, compounds of Formula I and pharmaceuticaliy acceptable salts thereof can be combined with one or more inhibitors of the following kinases for the treatment of cancer: PIM (PIM 1, PIM 2, PIM 3), IDO, AKT 1, AKT2 and AKT3, TGFR, PKA, PKG, PKC, CaM-kinase, phosphorylase kinase, MEKK, ERK, MAPK, mTOR, EGFR, HER2, HER3, HER4, INS-R, IGF-1R, IR-R, PDGFaR, PDGF R, CSFIR, KIT, FLK-Π, KDR/FLK-1, FLK-4, flt-1, FGFR, FGFR1, FGFR2, FGFR3, FGFR4, c-MET, Ron, Sea, TRKA, TRKB, TRKC, FLT3, VEGFR/Fit2, FIt4, EphAI, EphA2, EphA3, EphB2, EphB4, Tie2, Src, Fyn, Lck, Fgr, Btk, FAK, SYK, FRK, JAK, ABL, ALK, and B-Raf.
[00811] Compounds of Formula I and pharmaceuticaliy acceptable salts thereof can also be used in combination with one or more additional anticancer agents, such as a chemotherapeutics. Example chemotherapeutics include any of: abarelix, aldesleukin, alemtuzumab, alitretinoin, allopurinol, altretamîne, anastrozole, arsenic tri oxide, asparaginase, azacitidine, bevacizumab, bexarotene, bleomycin, bortezomib, busulfan intravenous, busulfan oral, calusterone, capecitabine, carboplatin, carmustine, cetuximab, chlorambucil, cisplatin, cladribine, clofarabine, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, dalteparin sodium, dasatinib, daunorubicin, decitabine, denileukin, denileukin diftitox, dexrazoxane, docetaxel, doxorubîcin, dromostanolone propionate, eculizumab, epirubicin, erlotinib, estramustine, etoposide phosphate, etoposide, exemestane, fentanyl citrate, fllgrastim, floxuridine, fludarabine, fluorouracil, fulvestrant, gefitinib, gemcitabine, gemtuzumab ozogamicin, goserelin acetate, histrelin acetate, ibritumomab tiuxetan, idarubicin, ifosfamide, imatinib mesylate, interferon alfa 2a, irinotecan, lapatinib ditosylate, lenalidomide, letrozole, leucovorin, leuprolide acetate, levamisole, lomustine,
147 meclorethamine, megestrol acetate, melphalan, mercaptopurine, methotrexate, methoxsalen, mitomycin C, mitotane, mîtoxantrone, nandrolone phenpropionate, nelarabine, nofetumomab, oxaliplatin, paclitaxel, pamidronate, panitumumab, pegaspargase, pegfilgrastim, pemetrexed disodium, pentostatin, pipobroman, plicamycin, procarbazine, quinacrine, rasburicase, rituximab, 5 ruxolitinib, sorafenib, streptozocin, sunitinib, sunitinib maleate, tamoxifen, temozolomide, tentposide, teste iactone, thalidomide, thioguanine, thiotepa, topotecan, toremifene, tositumomab, trastuzumab, tretînoin, uracîl mustard, valrubicin, Vinblastine, vincristine, vinorelbine, vorinostat and zolédronate.
[00812] In some embodiments, signal transduction pathway inhibitors include kinase 10 inhibitors of the Ras-Raf-MEK-ERK. pathway (e.g., binimetinib, selumetinib, encorafenib, sorafenib, trametinib, cobimetinib, dabrafenib, and vemurafenib), kinase inhibitors of the PI3KAKT-mT0R-S6K pathway (e.g. everolimus, rapamycin, perifosine, temsirolimus), and other kinase inhibitors, such as baricitinib, brigatinib, capmatinib, danusertib, ibrutinib, milciclib, quercetin, regorafenib, ruxolitinib, semaxanib, ((R)-amino-N-[5,6-dihydro-2-( 1-methyl-lH15 pyrazol-d-yO-ô-oxo-lH-pyrrolo^jS^-eflP^Jbenzodiazepin-S-y^-cyclohexaneacetamide), and TGI 01209 (N-tert-butyl-3-(5-methyl-2-(4-(4-methylpiperazin-l -yl)phenylamino)pyrimidin-4ylamino)benzenesulfonamide).
[00813] A combination of a compound of Formula I in combination with binimetinib, selumetinib, encorafenib, sorafenib, trametinib, or vemurafenib results in sensitization of tumors 20 that are résistant to binimetinib, selumetinib, encorafenib, sorafenib, trametinib, or vemurafenib, respective ly.
[00814] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Formula I or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinib, 25 (iiî) encorafenib, (iv) sorafenib, (v) trametinib, and (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00815] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Formula I or a pharmaceutically acceptable sali or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) 30 encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
148
[00816] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 25, 37, 46, 48, 55, 58, 72, 76, 77, 78, 83, 84, 85, 91, 97, 100, 103, 105, 107, 108, 114, 115, 119, 121, 124, 125, 126, 127, 129, 151, 152, 163, 169, 188, 190, 199, 200, or 201, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional 5 anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00817] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Fonnula I or a pharmaceutically acceptable sait or solvaté thereof, and (b) 10 binimetinib or a pharmaceutically acceptable sait or solvaté thereof.
[00818] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 25, 37, 46,48, 55, 58, 72, 76, 77, 78, 83, 84, 85,91,97, 100, 103, 105, 107, 108, 114, 115, 119, 121, 124, 125, 126, 127, 129, 151, 152, 163, 169, 188, 190, 199, 200, or 201, or a pharmaceutically acceptable sait or solvaté thereof, and (b) binimetinib or a 15 pharmaceutically acceptable sait or solvaté thereof.
[00819] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Formula I or a pharmaceutically acceptable sait or solvaté thereof, and (b) encorafenib or a pharmaceutically acceptable sait or solvaté thereof.
[00820] In one embodiment, there îs provided a pharmaceutical combination which comprises 20 (a) a compound of Example No. 25, 37,46,48, 55, 58, 72, 76, 77, 78, 83, 84, 85,91, 97, 100, 103,
105, 107, 108, 114, 115, 119, 121, 124, 125, 126, 127, 129, 151, 152, 163, 169, 188, 190, 199, 200, or 201, or a pharmaceutically acceptable sait or solvaté thereof, and (b) encorafenib or a pharmaceutically acceptable sait or solvaté thereof.
[00821] In one embodiment, there is provided a pharmaceutical combination which comprises 2 5 (a) a compound of Formula I or a pharmaceutically acceptable sait or solvaté thereof, and (b) selumetinib or a pharmaceutically acceptable sait or solvaté thereof.
[00822] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 25, 37, 46, 48, 55, 58, 72, 76, 77, 78, 83, 84, 85, 91,97, 100, 103, 105, 107, 108, 114, 115, 119, 121, 124, 125, 126, 127, 129, 151, 152, 163, 169, 188, 190, 199, 30 200, or 201, or a pharmaceutically acceptable sait or solvaté thereof, and (b) selumetinib or a pharmaceutically acceptable sait or solvaté thereof.
149
[00823] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Formula I or a pharmaceutically acceptable sait or solvaté thereof, and (b) sorafenib or a pharmaceutically acceptable sait or solvaté thereof
[00824] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 25, 37, 46, 48, 55, 58, 72, 76, 77, 78, 83, 84, 85, 91, 97, 100, 103, 105, 107, 108, 114, 115, 119, 121, 124, 125, 126, 127, 129, 151, 152, 163, 169, 188, 190, 199, 200, or 201, or a pharmaceutically acceptable sait or solvaté thereof, and (b) sorafenib or a pharmaceutically acceptable sait or solvaté thereof.
[00825] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Formula I or a pharmaceutically acceptable sait or solvaté thereof, and (b) trametinib or a pharmaceutically acceptable sait or solvaté thereof.
[00826] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 25, 37,46,48, 55, 58, 72, 76, 77, 78, 83, 84, 85, 91, 97, 100, 103, 105, 107, 108, 114, 115, 119, 121, 124, 125, 126, 127, 129, 151, 152, 163, 169, 188, 190, 199, 200, or 201, or a pharmaceutically acceptable sait or solvaté thereof, and (b) trametinib or a pharmaceutically acceptable sait or solvaté thereof.
[00827] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Formula I or a pharmaceutically acceptable sait or solvaté thereof, and (b) vemurafenib or a pharmaceutically acceptable sait or solvaté thereof.
[00828] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 25, 37, 46, 48, 55, 58, 72, 76, 77, 78, 83, 84, 85, 91, 97, 100, 103, 105, 107, 108, 114, 115, 119, 121, 124, 125, 126, 127, 129, 151, 152, 163, 169, 188, 190, 199, 200, or 201, or a pharmaceutically acceptable sait or solvaté thereof, and (b) vemurafenib or a pharmaceutically acceptable sait or solvaté thereof.
[00829] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 25, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00830] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 25, or a pharmaceutically acceptable sait or solvaté thereof, and
150 (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00831] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 37, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinîb, (iii) encorafenib, (iv) sorafenib, (v) trametînib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. |00832] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 37, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00833] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 46, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinîb, (iîi) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00834] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 46, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00835] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 48, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinîb, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00836] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 48, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii)
151 encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00837] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 55, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00838] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 55, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00839] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 58, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00840] in one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 58, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00841] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 72, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00842] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 72, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof.
152 and a combination thereof.
[00843] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 76, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00844] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 76, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00845] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 77, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00846] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 77, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00847] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 78, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00848] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 78, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
153
[00849] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 83, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00850] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 83, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00851] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 84, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00852] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 84, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00853] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 85, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00854] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 85, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
154
[00855] In one embodiment, there is provided a phannaceutical combination which comprises (a) a compound of Example No. 91, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00856] In one embodiment, there is provided a phannaceutical combination which comprises (a) a compound of Example No. 91, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00857] In one embodiment, there is provided a phannaceutical combination which comprises (a) a compound of Example No. 97, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00858] In one embodiment, there is provided a phannaceutical combination which comprises (a) a compound of Example No. 97, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00859] In one embodiment, there is provided a phannaceutical combination which comprises (a) a compound of Example No. 100, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00860] In one embodiment, there is provided a phannaceutical combination which comprises (a) a compound of Example No. 100, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
155
[00861] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 103, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in 5 the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00862] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 103, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, 10 and a combination thereof.
[00863] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 105, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in 15 the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00864] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 105, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, 2 0 and a combination thereof.
[00865] in one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 107, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) 2 5 selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00866] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 107, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) 30 encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
156
[00867] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 108, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consistîng of (i) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00868] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 108, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consistîng of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00869] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 114, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consistîng of (i) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00870] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 114, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consistîng of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00871] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 115, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consistîng of (i) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00872] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 115, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consistîng of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
157
[00873] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 119, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00874] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 119, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00875] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 121, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [008761 In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 121, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00877] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 124, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [008781 In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 124, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
158
[00879] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 125, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00880] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 125, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00881] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 126, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00882] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 126, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00883] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 127, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (î) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00884] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 127, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (t) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
159
[00840] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 129, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00885] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 129, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00886] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 151, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00887] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 151, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
|00888] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 152, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00889] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 152, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
160
[00890] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 163, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00891| In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 163, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00892] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 169, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00893] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 169, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00894] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 188, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (î) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00895] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 188, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
161
[00896] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 190, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetînib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00897] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 190, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00898] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 199, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinîb, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00899] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 199, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00900] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 200, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinîb, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof. [00901] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 200, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
162
[00902] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 201, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib, (ii) selumetinib, (iii) encorafenib, (iv) sorafenib, (v) trametinib, (vi) vemurafenib, each optionally in 5 the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
|00903| in one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 201, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) binimetinib and (ii) encorafenib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, 10 and a combination thereof.
[00904] In each of the above combinations, the compound of Formula 1 or the pharmaceutically acceptable sait thereof and the additional anticancer agent may be formulated as separate compositions or dosages for simultaneous, separate or sequential use for the treatment of cancer, wherein the amounts of the compound of Formula I or a pharmaceutically acceptable sait thereof 15 and of the additional anticancer agent are together effective in treating the cancer. Also provided herein is a pharmaceutical composition comprising such a combination. Also provided herein is the use of such a combination for the préparation of a médicament for the treatment of cancer (e.g., a TAM-associated cancer or a c-Met-associated cancer). Also provided herein is a commercial package or product comprising such a combination as a combined préparation for simultaneous, 2 0 separate or sequential use; and to a method of treatment of cancer a patient in need thereof.
[00905] Also provided are methods of treating an individual with cancer that include administering that include administering to a patient identified or diagnosed as having cancer (e.g., a TAM-associated cancer or a c-Met-associated cancer) a therapeutically effective amount of any 25 ofthe combinations.
(00906] Also provided herein are methods of treating a patient identified or diagnosed as having a TAM-associated cancer or a c-Met-associated that include administering to a patient identified or diagnosed as having a TAM-associated cancer or a c-Met-associated a therapeutically effective amount of a therapeutically effective amount of any of the combinations.
A combination of a compound of Formula 1 in combination with an EGFR, inhibitor (e.g., any of the EGFR inhibitors described herein) results in effective réduction in prolifération of
163 cancer cetls having résistance to EGFR inhibitors or cancer cells having résistance to c-Met inhibîtors).
In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Formula I or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) gefitinib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Formula 1 or a pharmaceutically acceptable sait or solvaté thereof, and (b) cetuximab (or a biosimilar thereof) or a pharmaceutically acceptable sait or solvaté thereof.
In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Formula 1 or a pharmaceutically acceptable sait or solvaté thereof, and (b) panitumumab (or a biosimilar thereof) or a pharmaceutically acceptable sait or solvaté thereof.
In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Formula I or a pharmaceutically acceptable sait or solvaté thereof, and (b) erlotinib or a pharmaceutically acceptable sait or solvaté thereof.
In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Formula T or a pharmaceutically acceptable sait or solvaté thereof, and (b) lapatinib or a pharmaceutically acceptable sait or solvaté thereof.
In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Formula I or a pharmaceutically acceptable sait or solvaté thereof, and (b) gefitinib or a pharmaceutically acceptable sait or solvaté thereof.
In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 25, 37, 46, 48, 55, 58, 72, 76, 77, 78, 83, 84, 85, 91, 97, 100, 103, 105, 107, 108, 114, 115, 119, 121, 124, 125, 126, 127, 129, 151, 152, 163, 169, 188, 190, 199, 200, or 201, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) gefitinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations
164 of any thereof.
In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 25, 37, 46, 48, 55, 58, 72, 76, 77, 78, 83, 84, 85, 91, 97, 100, 103, 105, 107, 108, 114, 1 15, 1 19, 121, 124, 125, 126, 127, 129, 151, 152, 163, 169, 188, 190, 199, 200, or 201, or a pharmaceutically acceptable sait or solvaté thereof and (b) cetuximab (or a bîosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
in one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 25, 37, 46, 48, 55, 58, 72, 76, 77, 78, 83, 84, 85, 91, 97, 100, 103, 105, 107, 108, 114, 115, 119, 121, 124, 125, 126, 127, 129, 151, 152, 163, 169, 188, 190, 199, 200, or 201, or a pharmaceutically acceptable sait or solvaté thereof and (b) panitumumab (or a bîosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 25, 37, 46, 48, 55, 58, 72, 76, 77, 78, 83, 84, 85, 91, 97, 100, 103, 105, 107, 108, 114, 115, 119, 121, 124, 125, 126, 127, 129, 151, 152, 163, 169, 188, 190, 199, 200, or 201, or a pharmaceutically acceptable sait or solvaté thereof and (b) erlotinib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 25, 37, 46, 48, 55, 58, 72, 76, 77, 78, 83, 84, 85, 91, 97, 100, 103, 105, 107, 108, 114, 115, 119, 121, 124, 125, 126, 127, 129, 151, 152, 163, 169, 188, 190, 199, 200, or 201, or a pharmaceutically acceptable sait or solvaté thereof and (b) lapatinib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 25, 37, 46, 48, 55, 58, 72, 76, ΊΊ, Ί&, 83, 84, 85, 91, 97, 100, 103, 105, 107, 108, 114, 115, 119, 121, 124, 125, 126, 127, 129, 151, 152, 163, 169, 188, 190, 199, 200, or 201, or a pharmaceutically acceptable sait or solvaté thereof and (b) gefitinib, each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
165
[00907] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 25, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additionaî anticancer agent selected from the group consisting of (i) cetuximab (or a biosîmilar thereof), (ii) panitumumab (or a biosîmilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) gefitinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00908] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 37, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additionaî anticancer agent selected from the group consisting of (i) cetuximab (or a biosîmilar thereof), (ii) panitumumab (or a biosîmilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) gefitinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00909] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 46, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additionaî anticancer agent selected from the group consisting of (i) cetuximab (or a biosîmilar thereof), (ii) panitumumab (or a biosîmilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) gefitinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00910] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 48, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additionaî anticancer agent selected from the group consisting of (i) cetuximab (or a biosîmilar thereof), (ii) panitumumab (or a biosîmilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) gefitinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00911] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 55, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additionaî anticancer agent selected from the group consisting of (i) cetuximab (or a biosîmilar thereof), (ii) panitumumab (or a biosîmilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) gefltinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
100912] In one embodiment, there is provided a pharmaceutical combination which comprises
166 (a) a compound of Example No. 58, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) cetuximab (or a biosimîlar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) gefitinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00913] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 72, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) gefitinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00914] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 76, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) gefitinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00915] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 77, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (î) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) gefitinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00916] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 78, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) gefitinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00917] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 83, or a pharmaceutically acceptable sait or solvaté thereof and
167 (b) an additional anticancer agent selected from the group consisting of (i) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) gefitinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00918] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 84, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) gefitinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and 10 combinations of any thereof.
[00919] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 85, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) 15 gefitinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00920] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 91, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) cetuximab (or a 20 biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) gefitinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
(00921] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 97, or a pharmaceutically acceptable sait or solvaté thereof and 2 5 (b) an additional anticancer agent selected from the group consisting of (i) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) gefitinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00922] In one embodiment, there is provided a pharmaceutical combination which comprises 30 (a) a compound of Example No. 100, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) cetuximab (or a
168 biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) gefitinib each optionally in the form ofa pharmaceuticaliy acceptable sait or solvaté thereof, and combinations of any thereof.
[00923] In one embodiment, there is provided a pharmaceutical combination which comprises 5 (a) a compound of Example No. 103, or a pharmaceuticaliy acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) gefitinib each optionally in the form of a pharmaceuticaliy acceptable sait or solvaté thereof, and combinations of any thereof.
[00924] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 105, or a pharmaceuticaliy acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) gefitinib each optionally in the form of a pharmaceuticaliy acceptable sait or solvaté thereof, and 15 combinations of any thereof.
[00925] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 107, or a pharmaceuticaliy acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) 20 gefitinib each optionally in the form of a pharmaceuticaliy acceptable sait or solvaté thereof, and combinations of any thereof.
[00926] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 108, or a pharmaceuticaliy acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) cetuximab (or a 25 biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) gefitinib each optionally in the form of a pharmaceuticaliy acceptable sait or solvaté thereof, and combinations ofany thereof.
[00927] in one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 114, or a pharmaceuticaliy acceptable sait or solvaté thereof and 30 (b) an additional anticancer agent selected from the group consisting of (i) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotinib, (iv) lapatinib, and (v)
169 gefitinib each optionally in the form of a phannaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00928] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 115, or a phannaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) cetuximab (or a biosimtlar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) gefitinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00929] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 119, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) gefitinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00930] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 121, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) gefitinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00931] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 124, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) gefitinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00932] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 125, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotinib, (iv) lapatinib, and (v)
170 gefïtinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00933] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 126, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consistîng of (i) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) gefïtinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00934] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 127, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consistîng of (i) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) gefïtinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00935] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 129, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consistîng of (i) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilarthereof), (iii) erlotinib, (iv) lapatinib, and (v) gefïtinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00936] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 151, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consistîng of (i) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) gefïtinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00937] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 152, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consistîng of (i) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotinib, (iv) lapatinib, and (v)
171 gefitinib each optionally in the fonn of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00938] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 163, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (î) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotînib, (iv) lapatinib, and (v) gefitînib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00939] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 169, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotînib, (iv) lapatinib, and (v) gefitinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00940] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 188, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (ï) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotînib, (iv) lapatinib, and (v) gefitinib each optionally in the form of a phannaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00941] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 190, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (t) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotînib, (iv) lapatinib, and (v) gefitinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00942] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 199, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotînib, (iv) lapatinib, and (v)
172 gefîtinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00943] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 200, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) gefîtinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00944] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 201, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) cetuximab (or a biosimilar thereof), (ii) panitumumab (or a biosimilar thereof), (iii) erlotinib, (iv) lapatinib, and (v) gefîtinib each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00945] A combination of a compound of Formula I in combination with an immune checkpoint inhibitor (e.g., any of the checkpoint inhibitors described herein, e.g., a PD-I or a PDL1 inhibitor) results in sensitization of tumors to immune checkpoint inhibitor therapy. For example, a compound of Formula I in combination with an immune checkpoint inhibitor can resuit în one or more (e.g., two, three, four, or five) of an increase in dendritic cell-dépendent antigen présentation, an increase in NK cell response, an increase in T-cell trafficking, an increase in Type 1 macrophages which results in production of immune stimulating cytokines, and an enhancement of both innate and adaptive immune response.
In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Formula I or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iii) cemîplimab (or a biosimilar thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 114IPDCA-170 (or a biosimilar thereof), (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00946] In one embodiment, there is provided a pharmaceutical combination which comprises
173 (a) a compound of Formula 1 or a pharmaceutically acceptable sait or solvaté thereof, and (b) nivolumab (or a biosimilar thereof) or a pharmaceutically acceptable sait or solvaté thereof.
[00947] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Formula I or a pharmaceutically acceptable sait or solvaté thereof, and (b) pembrolîzumab (or a biosimilar thereof) or a pharmaceutically acceptable sait or solvaté thereof.
[00948] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Formula I or a pharmaceutically acceptable sait or solvaté thereof, and (b) cemiplimab (or a biosimilar thereof) or a pharmaceutically acceptable sait or solvaté thereof.
[00949] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Formula I or a pharmaceutically acceptable sait or solvaté thereof, and (b) pidilizumab (or a biosimilar thereof) or a pharmaceutically acceptable sait or solvaté thereof.
[00950] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Formula I or a pharmaceutically acceptable sait or solvaté thereof, and (b) 1141PDCA-170 (or a biosimilar thereof) or a pharmaceutically acceptable sait or solvaté thereof. [00951] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Formula I or a pharmaceutically acceptable sait or solvaté thereof, and (b) atezolizumab (or a biosimilar thereof) or a pharmaceutically acceptable sait or solvaté thereof.
[00952] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Formula I or a pharmaceutically acceptable sait or solvaté thereof, and (b) aveiumab (or a biosimilar thereof) or a pharmaceutically acceptable sait or solvaté thereof.
[00953| In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Formula I or a pharmaceutically acceptable sait or solvaté thereof, and (b) durvalumab (or a biosimilar thereof) or a pharmaceutically acceptable sait or solvaté thereof.
[00954] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 25, 37, 46, 48, 55, 58, 72, 76, ΊΊ, 78, 83, 84, 85, 91, 97, 100, 103, 105, 107, 108, 114, 115, 119, 121, 124, 125, 126, 127, 129, 151, 152, 163, 169, 188, 190, 199, 200, or 201, or a pharmaceutically acceptable sait or solvaté thereof and (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a biosimilar thereof), (ii) pembrolîzumab (or a biosimilar thereof), (iii) cemiplimab (or a biosimilar thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 1I41PDCA-I70 (or a biosimilar thereof), (vi)
174 atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00955] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 25, 37, 46, 48, 55, 58, 72, 76, 77, 78, 83, 84, 85, 91, 97, 100, 103, 105, 107, 108, 114, 115, 119, 121, 124, 125, 126, 127, 129, 151, 152, 163, 169, 188, 190, 199, 200, or 201, or a pharmaceutically acceptable sait or solvaté thereof, and (b) nivolumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00956] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 25, 37, 46, 48, 55, 58, 72, 76, 77, 78, 83, 84, 85, 91, 97, 100, 103, 105, 107, 108, 114, 115, 119, 121, 124, 125, 126, 127, 129, 151, 152, 163, 169, 188, 190, 199, 200, or 201, or a pharmaceutically acceptable sait or solvaté thereof, and (b) pembrolizumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00957] In one embodiment, there is provided a phannaceutical combination which comprises (a) a compound of Example No. 25, 37, 46, 48, 55, 58, 72, 76, 77, 78, 83, 84, 85, 91, 97, 100, 103, 105, 107, 108, 114, 115, 119, 121, 124, 125, 126, 127, 129, 151, 152, 163, 169, 188, 190, 199, 200, or 201, or a pharmaceutically acceptable sait or solvaté thereof, and (b) cemiplimab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00958] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 25, 37, 46, 48, 55, 58, 72, 76, 77, 78, 83, 84, 85, 91,97, 100, 103, 105, 107, 108, 114, 115, 119, 121, 124, 125, 126, 127, 129, 151, 152, 163, 169, 188, 190, 199, 200, or 201, or a pharmaceutically acceptable sait or solvaté thereof, and (b) pidilizumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00959] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 25, 37, 46, 48, 55, 58, 72, 76, 77, 78, 83, 84, 85, 91, 97, 100, 103, 105, 107, 108, 114, 115, 119, 121, 124, 125, 126, 127, 129, 151, 152, 163, 169, 188, 190, 199, 200, or 201, or a pharmaceutically acceptable sait or solvaté thereof, and (b) 1141PDCA-170 (or
175 a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00960] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 25, 37, 46, 48, 55, 58, 72, 76, 77, 78, 83, 84, 85, 91, 97, 100, 103, 105, 107, 108, 114, 115, 119, 121, 124, 125, 126, 127, 129, 151, 152, 163, 169, 188, 190, 199, 200, or 201, or a pharmaceutically acceptable sait or solvaté thereof, and (b) atezolizumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00961] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 25, 37, 46, 48, 55, 58, 72, 76, 77, 78, 83, 84, 85, 91, 97, 100, 103, 105, 107, 108, 114, 115, 119, 121, 124, 125, 126, 127, 129, 151, 152, 163, 169, 188, 190, 199, 200, or 201, or a pharmaceutically acceptable sait or solvaté thereof, and (b) avelumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00962] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 25, 37,46, 48, 55, 58, 72, 76, 77, 78, 83, 84, 85, 91,97, 100, 103, 105, 107, 108, 114, 115, 119, 121, 124, 125, 126, 127, 129, 151, 152, 163, 169, 188, 190, 199, 200, or 201, or a pharmaceutically acceptable sait or solvaté thereof, and (b) durvalumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and a combination thereof.
[00963] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 25, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iii) cemiplimab (or a biosimilar thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 1141PDCA-170 (or a biosimilar thereof), (vi) atezolizumab (or a biosimilar thereof), (vîi) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00964] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 37, or a pharmaceutically acceptable sait or solvaté thereof, and
176 (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iii) cemiplimab (or a biosimilar thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 1141PDCA-170 (or a biosimilar thereof), (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00965] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 46, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iii) cemiplimab (or a biosimilar thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 1141PDCA-170 (or a biosimilar thereof), (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00966] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 48, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iii) cemiplimab (or a biosimilar thereof), (îv) pidilizumab (or a biosimilar thereof), (v) 1141PDCA-170 (or a biosimilar thereof), (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00967] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 55, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iii) cemiplimab (or a biosimilar thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 114IPDCA-17Û (or a biosimilar thereof), (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00968] In one embodiment, there is provided a pharmaceutical combination which comprises
177 (a) a compound of Example No, 58, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iii) cemiplimab (or a biosimilar thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 1141PDCA-I70 (or a biosimilar thereof), (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00969] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No, 72, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iii) cemiplimab (or a biosimilar thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 1141PDCA-170 (or a biosimilar thereof), (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00970] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 76, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iii) cemiplimab (or a biosimilar thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 1141PDCA-170 (or a biosimilar thereof), (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally in the form ofa pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00971] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 77, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iii) cemiplimab (or a biosimilar thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 1141PDCA-170 (or a biosimilar thereof), (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
178
[00972] In one embodiment there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 78, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iii) cemiplimab (or a biosimilar thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 114IPDCA-170 (or a biosimilar thereof), (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally in the fonn of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00973] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 83, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iii) cemiplimab (or a biosimilar thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 1141PDCA-170 (or a biosimilar thereof), (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally în the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00974] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 84, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iii) cemiplimab (or a biosimilar thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 1141PDCA-170 (or a biosimilar thereof), (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00975] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 85, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iii) cemiplimab (or a biosimilar thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 114IPDCA-I70 (or a biosimilar thereof), (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally în the form of a pharmaceutically acceptable
179 sait or solvaté thereof, and combinations of any thereof
[00976] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 91, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a bîosimilar thereof), (ii) pembrolizumab (or a bîosimilar thereof), (iii) cemiplîmab (or a bîosimilar thereof), (iv) pidilizumab (or a bîosimilar thereof), (v) 1141PDCA-170 (or a bîosimilar thereof), (vi) atezolizumab (or a bîosimilar thereof), (vii) avelumab (or a bîosimilar thereof), and (viii) durvalumab (or a bîosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00977] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 97, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a bîosimilar thereof), (ii) pembrolizumab (or a bîosimilar thereof), (iii) cemiplîmab (or a bîosimilar thereof), (iv) pidilizumab (or a bîosimilar thereof), (v) 1141PDCA-170 (or a bîosimilar thereof), (vi) atezolizumab (or a bîosimilar thereof), (vii) avelumab (or a bîosimilar thereof), and (viii) durvalumab (or a bîosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00978] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 100, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a bîosimilar thereof), (ii) pembrolizumab (or a bîosimilar thereof), (iii) cemiplîmab (or a bîosimilar thereof), (iv) pidilizumab (or a bîosimilar thereof), (v) 1141PDCA-170 (or a bîosimilar thereof), (vi) atezolizumab (or a bîosimilar thereof), (vii) avelumab (or a bîosimilar thereof), and (viii) durvalumab (or a bîosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00979] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 103, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a bîosimilar thereof), (ii) pembrolizumab (or a bîosimilar thereof), (iii) cemiplîmab (or a bîosimilar thereof), (iv) pidilizumab (or a bîosimilar thereof), (v) 1141PDCA-170 (or a bîosimilar thereof), (vi) atezolizumab (or a bîosimilar thereof), (vii) avelumab (or a bîosimilar thereof), and (viii)
180 durvalumab (or a biosîmilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00980] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 105, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additionaî anticancer agent selected from the group consisting of (i) nivolumab (or a biosîmilar thereof), (ii) pembrolizumab (or a biosîmilar thereof), (iii) cemiplimab (or a biosîmilar thereof), (iv) pidilizumab (or a biosîmilar thereof), (v) 1141PDCA-170 (or a biosîmilar thereof), (vi) atezolizumab (or a biosîmilar thereof), (vii) avelumab (or a biosîmilar thereof), and (viii) durvalumab (or a biosîmilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00981] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 107, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additionaî anticancer agent selected from the group consisting of (i) nivolumab (or a biosîmilar thereof), (ii) pembrolizumab (or a biosîmilar thereof), (iii) cemiplimab (or a biosîmilar thereof), (iv) pidilizumab (or a biosîmilar thereof), (v) 1141PDCA-I70 (or a biosîmilar thereof), (vi) atezolizumab (or a biosîmilar thereof), (vii) avelumab (or a biosîmilar thereof), and (viii) durvalumab (or a biosîmilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00982] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 108, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additionaî anticancer agent selected from the group consisting of (i) nivolumab (or a biosîmilar thereof), (ii) pembrolizumab (or a biosîmilar thereof), (iii) cemiplimab (or a biosîmilar thereof), (iv) pidilizumab (or a biosîmilar thereof), (v) 1141PDCA-170 (or a biosîmilar thereof), (vi) atezolizumab (or a biosîmilar thereof), (vii) avelumab (or a biosîmilar thereof), and (viii) durvalumab (or a biosîmilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00983| In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 114, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additionaî anticancer agent selected from the group consisting of (i) nivolumab (or a biosîmilar thereof), (ii) pembrolizumab (or a biosîmilar thereof), (iii) cemiplimab (or a biosîmilar thereof), (iv) pidilizumab (or a biosîmilar thereof), (v) 1141PDCA-170 (or a biosîmilar thereof),
18] (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof,
[00984] In one embodiment, there is provided a pharmaceutical combination which comprises 5 (a) a compound of Example No. 115, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iii) cemiplîmab (or a biosimilar thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 1141PDCA-170 (or a biosimilar thereof), (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) 10 durvalumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00985] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 119, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a 15 biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iii) cemiplîmab (or a biosimilar thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 1141PDCA-170 (or a biosimilar thereof), (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00986] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 121, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (î) nivolumab (or a biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iii) cemiplîmab (or a biosimilar thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 1141PDCA-170 (or a biosimilar thereof), 25 (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00987] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 124, or a pharmaceutically acceptable sait or solvaté thereof, and 3 0 (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iîi) cemiplîmab (or a biosimilar
182 thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 1141PDCA-170 (or a biosimilar thereof), (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00988] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 125, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iii) cemiplimab (or a biosimilar thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 1141PDCA-170 (or a biosimilar thereof), 10 (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00989] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 126, or a pharmaceutically acceptable sait or solvaté thereof, and 15 (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iii) cemiplimab (or a biosimilar thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 1141PDCA-170 (or a biosimilar thereof), (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable 20 sait or solvaté thereof, and combinations of any thereof.
[00990] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 127, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a 2 5 biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iii) cemiplimab (or a biosimilar thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 1141PDCA-170 (or a biosimilar thereof), (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00991 ] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 129, or a pharmaceutically acceptable sait or solvaté thereof, and
183 (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iii) cemiplimab (or a biosimilar thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 1141PDCA-170 (or a biosimilar thereof), (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00992] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 151, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iii) cemiplimab (or a biosimilar thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 1141PDCA-170 (or a biosimilar thereof), (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00993] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 152, or a pharmaceutically acceptable sait or solvaté thereof, and ((b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iii) cemiplimab (or a biosimilar thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 1141PDCA-170 (or a biosimilar thereof), (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00994] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 163, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iii) cemiplimab (or a biosimilar thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 1141PDCA-I70 (or a biosimilar thereof), (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00995] In one embodiment, there is provided a pharmaceutical combination which comprises
184 (a) a compound of Example No. 169, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iii) cemiplimab (or a biosimilar thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 1141PDCA-170 (or a biosimilar thereof), 5 (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00 996] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 188, or a pharmaceutically acceptable sait or solvaté thereof, and 10 (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iii) cemiplimab (or a biosimilar thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 1141PDCA-170 (or a biosimilar thereof), (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable 15 sait or solvaté thereof, and combinations of any thereof.
[00997] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 190, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iii) cemiplimab (or a biosimilar 20 thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 1141PDCA-Î70 (or a biosimilar thereof), (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[00 998] In one embodiment, there is provided a pharmaceutical combination which comprises 2 5 (a) a compound of Example No. 199, or a pharmaceutically acceptable sali or solvaté thereof, and (b) an additional anticancer agent selected from the group consisting of (i) nivolumab (or a biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iii) cemiplimab (or a biosimilar thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 1141PDCA-170 (or a biosimilar thereof), (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) 30 durvalumab (or a biosimilar thereof), each optionally in the fonn of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
185
[00999] In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 200, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consistîng of (i) nivolumab (or a biosimilar thereof), (ii) pembrolizumab (or a biosimilar thereof), (iii) cemiplimab (or a biosimilar thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 1141PDCA-170 (or a biosimilar thereof), (vi) atezolizumab (or a biosimilar thereof), (viî) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
In one embodiment, there is provided a pharmaceutical combination which comprises (a) a compound of Example No. 201, or a pharmaceutically acceptable sait or solvaté thereof, and (b) an additional anticancer agent selected from the group consistîng of (i) nivolumab (or a biosimilar thereof), (it) pembrolizumab (or a biosimilar thereof), (iii) cemiplimab (or a biosimilar thereof), (iv) pidilizumab (or a biosimilar thereof), (v) 1141PDCA-170 (or a biosimilar thereof), (vi) atezolizumab (or a biosimilar thereof), (vii) avelumab (or a biosimilar thereof), and (viii) durvalumab (or a biosimilar thereof), each optionally in the form of a pharmaceutically acceptable sait or solvaté thereof, and combinations of any thereof.
[001000] Angiogenesis inhibitors may be efficacious in some tumors in combination with compounds of Formula 1 or pharmaceutically acceptable salts thereof. These include antibodies against VEGF or VEGFR or kinase inhibitors of VEGFR. Antibodies or other therapeutic proteins against VEGF include bevacizumab and aflibercept. Inhibitors of VEGFR kinases and other antiangiogenesis inhibitors include but are not limited to sunitinib, sorafenib, axitinib, cediranib, pazopanib, regorafenib, brivanib, and vandetanib.
[001001J Non-limiting examples of radiotherapy include radioiodide therapy, external-beam radiation, and radium 223 therapy.
[001002] Non-limiting examples of surgery include, e.g., open surgery or minimally invasive surgery. Surgery can include, e.g., removing an entire tumor, debulking of a tumor, or removing a tumor that is causing pain or pressure in the subject. Methods for performing open surgery and minimally invasive surgery on a subject having a cancer are known in the art.
[001003J Accordingly, also provided herein is a method of treating cancer, comprising administering to a patient in need thereof a pharmaceutical combination for treating cancer which comprises (a) a compound of Formula 1 or a pharmaceutically acceptable sait thereof, and (b) an
186 additional anticancer agent, for simultaneous, separate or sequential use for the treatment of cancer, wherein the amounts ofthe compound of Formula 1 or a pharmaceuticaliy acceptable sait thereof and the additional anticancer agent are together effective in treating the cancer.
[001004] Also provided herein is a method of treating cancer, comprising administering to a 5 patient in need thereof a pharmaceutical combination for treating cancer which comprises (a) a compound of Formula I or a pharmaceuticaliy acceptable sait thereof, and (b) an additional anticancer therapy, wherein the therapy is selected from radiation therapy and surgery. In one embodiment, the additional anticancer therapy is radiation therapy. In one embodiment, the additional anticancer therapy is surgery.
[001005] In some embodiments, the additional anticancer agent(s) includes any one of the above listed thérapies or therapeutic agents which are standards of care in cancers wherein the cancer is a TAM-associated cancer. In one embodiment, the compound of Formula I the additional anticancer agent is an immunotherapy agent. In one embodiment, the immunotherapy agent is a CTLA-4 inhibitor (e.g., an anti-CTLA-4 antibody), a PD-1 inhibitor (e.g., an anti-PD-1 monoclonal antibody) or a PD-L1 inhibitor (e.g., an anti-PD-Ll monoclonal antibody).
[001006] In one embodiment, provided herein is a method for treating cancer, comprising administering a compound of Formula I in combination with an immune checkpoint inhibitor. In some embodiments, the immunotherapy includes one or more immune checkpoint inhibitors (e.g., PDR001 or any of the other exemplary immune checkpoint inhibitors described herein). In some
0 embodiments, the immune checkpoint inhibitor is a CTLA-4 inhibitor (e.g., an anti-CTLA-4 antibody), a PD-1 inhibitor (e.g., an anti-PD-1 monoclonal antibody), a PD-L1 inhibitor (e.g., an antî-PD-Ll monoclonal antibody), a NOX2 inhibitor, an A2A4 inhibitor, a B7-H3 inhibitor (e.g., MGA271), a B7-H4 inhibitor (e.g,, an anti-B7-H4 antibody, e.g., those described in Dangaj et al., Cancer Res. 73(15):4820-4829, 2013), an IDO inhibitor (e.g., coptisine, 1-methyl-D-tryptophan, 25 NLG-919, indoximod, 1-DL-methyl tryptophan, or the inhibitors described in Brastianos et al.,
JACS 128(50:16046-16047, 2006), a TIM3 inhibitor, a LAG3 inhibitor (e.g., BMS-986016), TIGIT inhibitor, a BTLA inhibitor, a V1STA inhibitor (e.g., 1141PDCA-170), a ICOS inhibitor, a KIR inhibitor (e.g., iirilumab), a CD39 inhibitor, a SIGLEC7 inhibitor, or a SIGLEC9 inhibitor. In some embodiments, the CTLA-4 inhibitor is ipilimumab (Yervoy®), tremelimumab (CP30 675,206), or the aptamers described in Santulli-Marotto et al., Cancer Res. 63(21):7483-7489,
2003. In some embodiments, the PD-1 inhibitor is pembrolizumab (Keytruda®), nivolumab
187 (Opdivo®), cemiplimab (Libtayo®), pidilizumab, or 1141PDCA-170. In some embodiments, the anti-PD-1 monoclonal antibody ts nivolumab or pembrolizumab. In some embodiments, the antiPD-1 antibody is pembrolizumab. In some embodiments, the PD-L1 inhibitor is atezolizumab (Tecentriq®), avelumab (Bavencio®), durvalumab (Imfinzi™). In some embodiments, the PDL1 inhibitor is atezolizumab (Tecentriq®), avelumab (Bavencio®), or durvalumab (Imfinzi™). In one embodiment, the compound of Formula I is selected from the compounds described in Example Nos. 1-201, or pharmaceutically acceptable salts thereof. In some embodiments, a compound of Formula I is selected from i) Example Nos. 1-20; ii) Example Nos. 21-40; iii) Example Nos. 41-60; iv) Example Nos. 61-80; v) Example Nos. 81-100; vi) Example Nos. 101120; vii) Example Nos. 121-140; viii) Example Nos. 141-160; ix) Example Nos. 161-180; x) Example Nos. 181-201 or pharmaceutically acceptable salts thereof. In some embodiments, provided herein is a method for treating cancer, comprising administering to a patient in need thereof a compound of Formula I in combination with an immune checkpoint inhibitor, wherein the patient is further treated with ionizing radiation. In one embodiment, the cancer overexpresses AXL. In one embodiment, the cancer does not hâve a B-RAF mutation. In one embodiment, the cancer has a B-RAF mutation. In one embodiment, the cancer has a RAS mutation. In one embodiment, the cancer has a EGFR mutation. In one embodiment, the cancer overexpresses MER. In one embodiment, the cancer is lung cancer. In one embodiment. the cancer is non-small cell lung carcinoma (NSCLC). In one embodiment, the cancer is colon cancer. In one embodiment, the cancer is prostate cancer. In one embodiment, the cancer is melanoma. In one embodiment, the cancer is Acute Lymphoblastic Leukemia (ALL). In one embodiment, the cancer is Acute Myeloid Leukemia (AML).
[001007] Combination thérapies as described herein may be administered without restriction on the order in which thérapies are administered to a patient with a disease or disorder described herein. Thus, in one embodiment, a compound of Formula I or pharmaceutically acceptable sait thereof can be administered priorto (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 36 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks before), concomitantly with, or subséquent to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, l hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 36 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks after) the administration of a second therapeutic
188 agent (e.g., any of the additional anticancer agents described herein) to the subject. In another embodiment, a compound of Formula I or pharmaceutically acceptable sait thereof can be administered prier to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 36 hours, 48 hours, 72 hours, 96 hours, l week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks before), concomitantly with, or subséquent to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 36 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks after) the administration of a second therapeutic agent (e.g., any of the anticancer agents described herein).
[001008] In one embodiment, provided herein is a method for treating cancer, comprising sensitizing said cancer to an anti-mitotic drug by administration of a compound of Formula I. In one embodiment, the anti-mitotic drug is a taxane-based chemotherapeutic, such as docetaxel.
[001009] In one embodiment, compounds of Formula I may be used in combination with other agents to treat patients who hâve primary or acquired résistance to at least one additional anticancer agent.
[001010] In one embodiment of methods disclosed herein for treating cancer, compounds of Formula I may be used as monotherapy to treat patients who hâve developed primary or acquired résistance to at least one additional anticancer agent.
[001011] In one embodiment, compounds of Formula I may be used to overcome résistance to at least one additional anticancer agent in a cancer. In one embodiment, a compound of Formula I is used in combination with the at least one additional anticancer agent to which the cancer has developed résistance.
[001012] In one embodiment, compounds of Formula I may be used to delay résistance to at least one additional anticancer agent. In one embodiment, a compound of Fonnula 1 is used in combination with the at least one additional anticancer agent.
[001013] As used herein, the term “résistance” refers to a clinical scénario where a cancer fails to respond to a targeted therapy or immunotherapy. For example, résistance of a cancer can be observed by, e.g., a decrease in the rate of increase of tumor burden in the subject, a lack of a decrease in the tumor burden in the subject, an increase in the dosage of a therapeutic agent over time required to achieve the same therapeutic effect in a patient, and the requirement of coadministration of an additional anticancer agent to achieve the same therapeutic effect as the
189 previous administration of the therapeutic agent as a monotherapy.
[001014] As used herein, the term primary résistance, also known as intrinsic résistance, refers to a clinical scénario where a cancer fails to respond to a targeted therapy or immunotherapy, that is, the cancer is résistant to a therapy without having been previously exposed to the therapy.
[001015] As used herein, the term acquired résistance refers to a clinical scénario in which a cancer initially responded to a targeted therapy or immunotherapy but after a period of time the cancer stops responding to the treatment (e.g., the cancer relapses and progresses).
[001016] In one embodiment of methods disclosed herein for treating cancer, compounds of Formula I may be used as monotherapy to treat patients who hâve developed primary or acquired 10 résistance to at least one additional anticancer agent.
[001017] In one embodiment of methods disclosed herein for treating cancer, compounds of Formula I may be used as in combination with at least one additional anticancer agent to treat patients who hâve developed primary or acquired résistance to one or more of the at least one additional anticancer agent (e.g., a targeted therapeutic agent).
[001018] Targeted therapeutic agents include inhibitors or antibodies against EGFR, HER2,
VEGFR, c-Met, Ret, IGFR1, PDGFR, FGFR1, FGFR2, FGFR3, FGFR4, TrkA, TrkB, TrkC, ROS, c-Kit, or Fit-3 and against cancer-associated fusion protein kinases such as Bcr-Abl and EML4Alk. Inhibitors against EGFR include gefîtinib, erlotinib, and nazartinib (see, e.g., U.S. Patent No. 10,195,208 and J. Med. Chem. 59(14):6671-6689, 2016), and inhibitors against EGFR/Her2 include but are not limited to dacomitinib, afatinib, lapatinib and neratinib. Antibodies against the EGFR include but are not limited to cetuximab, panitumumab and necitumumab. Inhibitors of cMet may be used in combination with compounds of Formula I of pharmaceutically acceptable salts thereof. c-MET inhibitors include onartumzumab, tivantinib, and INC-280. Inhibitors against FGFRs include but not limited to AZD4547, BAY1187982, ARQ087, BGJ398, BIBF1120,
5 TKI258, iucitanib, dovitinib, TAS-120, JNJ-42756493, and Debiol347. Inhibitors against Trks include but not limited to larotrectinib (LOXO-101), and entrectinib (RXDX-101). Inhibitors against Abl (or Bcr-Abl) include imatinib, dasatinib, nilotinib, and ponatinib and those against Alk (or EML4-ALK) include crizotinib.
[001019] In one embodiment, provided herein are methods of treating a patient having cancer 30 who has been previously treated with a First kinase inhibitor, wherein the first kinase inhibitor is not a compound of Formula I, comprising administering to said patient a therapeutically effective
190 amount of a compound of Formula I or a pharmaceutically acceptable sait thereof. In one embodiment, the patient is treated with a compound of Formula I as a single agent. In one embodiment, the patient is treated with a combination of a compound of Formula I and the previously administered first kinase inhibitor. In one embodiment, the patient is treated with a 5 combination of a compound of Formula I and the previously administered first kinase inhibitor.
In one embodiment, the compound of Formula I and the previously administered first kinase inhibitor are administered as separate dosages sequentially in any order. In one embodiment, the kinase inhibitor is an EGFR inhibitor. In one embodiment, the EGFR inhibitor is erlotinib or lapatinib. In one embodiment, the kinase inhibitor is a PI3Ka inhibitor. In one embodiment, the 10 PI3Ka inhibitor is alpelisib. In one embodiment, the kinase inhibitor is a MEK inhibitor. In one embodiment, the MEK inhibitor is binimetinib, U0126, or PD 325901. In one embodiment, the kinase inhibitor is an FGFR inhibitor. In one embodiment, the kinase inhibitor ts an ALK inhibitor. In one embodiment, the kinase inhibitor is an IGFR] inhibitor. In one embodiment, the cancer is breast cancer (e.g., triple négative breast cancer), head and neck cancer (e.g., squamous cell head 15 and neck cancer), non-small cell lung cancer, colorectal cancer, esophageal squamous cell carcinoma, or melanoma.
[001020] In one embodiment, provided herein are methods of treating a patient having cancer who has been previously treated with an EGFR antibody, comprising administering to said patient a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable 20 sait thereof. In one embodiment, the patient is treated with a compound of Formula I as a single agent. In one embodiment, the patient is treated with a combination of a compound of Formula I and the previously administered EGFR antibody. In one embodiment, the compound of Formula I and the previously administered EGFR antibody are administered as separate dosages sequentially in any order. In one embodiment, the EGFR antibody is cetuximab. In one 25 embodiment, the cancer is breast cancer, head and neck cancer, or non-small cell lung cancer
[001021] In one embodiment, provided herein are methods of treating a patient having cancer who has been previously treated with a first kinase inhibitor, wherein the first kinase inhibitor is not a compound of Formula I, comprising (a) determining that said cancer overexpresses a TAM kinase and/or c-Met kinase (e.g., as compared to a non-cancerous tissue or a cell in the patient or 30 a different subject), and (b) after (a), administering to said patient a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof. In one
191 embodiment, the step of determining if the cancer overexpresses a TAM kinase and/or a c-Met kinase includes a step of performing an assay on a sample obtained from the patient to détermine whether the patient has abnormal (e.g., increased) expression, level, and/or activity of one or more of the TAM kinases and/or c-Met kinase (e.g., as compared to a non-cancerous tissue or cell in the patient or a different subject), e.g., AXL and/or MER. and/or TYRO3 and/or c-Met. In one embodiment, the cancer that was previously treated with the first kinase inhibitor overexpresses AXL. In one embodiment, the cancer that was previously treated with the first kinase inhibitor overexpresses MER. In one embodiment, the cancer that was previously treated with the first kinase inhibitor overexpresses TYRO3. In one embodiment, the cancer that was previously treated with the first kinase inhibitor overexpresses c-Met kinase. In one embodiment, the method further comprises obtaining a sample from the patient. In one embodiment, the sample is a biopsy sample. In one embodiment, the assay is selected from the group consisting of sequencing, immunohistochemistry, enzyme-lînked immunosorbent assay, and fluorescence in situ hybridization (FISH). In one embodiment, the first kinase inhibitor is an EGFR inhibitor. In one embodiment, the EGFR inhibitor is erlotînib or lapatinib. In one embodiment, the first kinase inhibitor is a PI3Ka inhibitor. In one embodiment, the PI3Ka inhibitor is alpelisîb. In one embodiment, the First kinase inhibitor is a MEK inhibitor. In one embodiment, the MEK inhibitor is binimetinib, LJO126, or PD 325901. In one embodiment, the first kinase inhibitor is an FGFR inhibitor. In one embodiment, the fîrst kinase inhibitor is an ALK inhibitor. In one embodiment, the first kinase inhibitor is an IGFR1 inhibitor. In one embodiment, the cancer is breast cancer (e.g., triple négative breast cancer), head and neck cancer (e.g., squamous cell head and neck cancer), non-small cell lung cancer, colorectal cancer, esophageal squamous cell carcinoma, or melanoma. In one embodiment, the patient is treated with a compound of Formula I as a single agent. In one embodiment, the patient is treated with a combination of a compound of Formula I and the first kinase inhibitor. In one embodiment, the compound of Formula I and the previously prescribed kinase inhibitor are administered as separate dosages sequentially in any order.
[001022] In one embodiment, provided herein is a method oftreating a subject having cancer, wherein the method comprises (a) determining that a cancer cell in a sample obtained from a subject having a cancer and previously administered one or more doses of a first kinase inhibitor, wherein the fîrst kinase inhibitor is not a compound of Formula I, overexpresses one or more TAM kinases and/or c-Met kinase (e.g., as compared to a non-cancerous tissue or cell in the subject or a
192 different subject); and (b) administering a compound of Formula 1 or a pharmaceuticaliy acceptable sait or solvaté thereof as a monotherapy or in conjunction with the previously admînistered first kinase inhibitor to the subject. In one embodiment, the cancer that was previously treated with the first kinase inhibitor overexpresses AXL. In one embodiment, the cancer that was previously treated with the first kinase inhibitor overexpresses MER. In one embodiment, the cancer that was previously treated with the first kinase inhibitor overexpresses TYRO3. In one embodiment, the cancer that was previously treated with the first kinase inhibitor overexpresses c-Met kinase. In one embodiment, the first kinase inhibitor is an EGFR inhibitor. In one embodiment, the EGFR inhibitor is eriotinib or lapatinib. In one embodiment, the first kinase inhibitor is a P13Ka inhibitor. In one embodiment, the PI3Ka inhibitor is alpelisib. In one embodiment, the first kinase inhibitor is a MEK inhibitor. In one embodiment, the MEK inhibitor is binimetinib, U0126, or PD 325901. In one embodiment, the first kinase inhibitor is an FGFR inhibitor. In one embodiment, the first kinase inhibitor is an ALK inhibitor. In one embodiment, the first kinase inhibitor is an IGFR1 inhibitor. In one embodiment, the cancer is breast cancer (e.g, triple négative breast cancer), head and neck cancer (e.g, squamous cell head and neck cancer), non-small cell lung cancer, colorectal cancer, esophageal squamous cell carcinoma, or melanoma. In one embodiment, the patient is treated with a compound of Formula I as a single agent. In one embodiment, the patient is treated with a combination of a compound of Formula I and the first kinase inhibitor. In one embodiment, the compound of Formula 1 and the previously prescribed kinase inhibitor are admînistered as separate dosages sequentially in any order.
[001023] In one embodiment of methods disclosed herein for treating cancer, compounds of Formula I may be used as monotherapy to treat patients who hâve developed primary or acquired résistance to chemotherapy.
[001024] In one embodiment of methods disclosed herein for treating cancer, compounds of Formula I may be used as in combination with a chemotherapeutic agent to treat patients who hâve developed primary or acquired résistance to the chemotherapeutic agent.
[001025] In one embodiment, provided herein are methods of treating a patient having cancer who has been previously treated with a chemotherapeutic, comprising administering to said patient a therapeutically effective amount of a compound of Formula I or a pharmaceuticaliy acceptable sait thereof. In one embodiment, the patient is treated with a compound of Formula I as a single agent. In one embodiment, the patient is treated with a combination of a compound of Formula I
193 and the previously administered chemotherapeutic. In one embodiment, the chemotherapeutic is selected from taxane-based chemotherapies (e.g., docetaxel), dexamethasone, and cytarabine. In one embodiment, the patient is treated with a compound of Formula I as a single agent. In one embodiment, the patient is treated with a compound of Formula I in combination with the previously administered chemotherapeutic. In one embodiment, the compound of Formula I and the previously administered chemotherapeutic are administered as separate dosages sequentially in any order. In one embodiment, the cancer is selected from leukemias (including acute myeloid leukemia and chronic myeloid leukemia, B-cell acute iymphoblastic leukemia, and T-lineage acute lymphoblastic leukemia), non-small cell lung cancer, pancreatic ductal adenocarcinoma, astrocytoma, lung adenocarcinoma, ovarian cancer, melanoma, and glioblastoma multifonne.
|001026] In one embodiment, provided herein are methods of treating a patient having cancer who has been previously treated with a chemotherapeutic, comprising (a) determining that said cancer overexpresses a TAM kinase and/or c-Met kinase (e.g., as compared to a non-cancerous tissue or cell in the patient or a different subject), and (b) after (a), administering to said patient a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof. In one embodiment, the step of determining if the cancer overexpresses a TAM kinase and/or c-Met kinase includes a step of performing an assay on a sample obtained from the patient to détermine whether the patient has abnormal expression, level, and/or activity of one or more of the TAM kinases and/or c-Met kinase, e.g., AXL and/or MER and/or TYRO3 and/or c-Met kinase. In one embodiment, the method further comprises obtaining a sample from the patient. In one embodiment, the sample is a biopsy sample. In one embodiment, the assay is selected from the group consisting of sequencing, immunohistochemistry, enzyme-lînked immunosorbent assay, and fluorescence in situ hybridization (F1SH). In one embodiment, the patient is treated with a compound of Formula I as a single agent. In one embodiment, the patient is treated with a combination of a compound of Formula 1 and the previously administered chemotherapeutic. In one embodiment, the chemotherapeutic is selected from taxane-based chemotherapies (e.g., docetaxel), dexamethasone, and cytarabine. In one embodiment, the patient is treated with a compound of Formula I as a single agent. In one embodiment, the patient is treated with a compound of Formula I in combination with the previously administered chemotherapeutic. In one embodiment, the compound of Formula I and the previously administered chemotherapeutic are administered as separate dosages sequentially in any order. In one embodiment, the cancer
194 is selected from leukemias (including acute myeloid leukemia and chronic myeloid leukemia, Bcell acute lymphoblastic leukemia, and T-lineage acute lymphoblastic leukemia), non-small cell lung cancer, pancreatic ductal adenocarcinoma, astrocytoma, lung adenocarcinoma, ovarian cancer, melanoma, and glîoblastoma multiforme.
[001027] In one embodiment, provided herein is amethod of treating asubject having cancer, wherein the method comprises (a) determining that a cancer cell in a sample obtained from a subject having a cancer and previously administered one or more doses of a chemotherapeutic, overexpresses one or more TAM kinases and/or c-Met kinase (e.g., as compared to a noncancerous tissue or cell in the subject or a different subject); and (b) administering a compound of Formula I or a pharmaceutically acceptable sait or solvaté thereof as a monotherapy or in conjunction with the previously administered chemotherapeutic or a different chemotherapeutic. In one embodiment, the cancer that was previously treated with the chemotherapeutic overexpresses AXL. In one embodiment, the cancer that was previously treated with the chemotherapeutic overexpresses MER. In one embodiment, the cancer that was previously treated with the chemotherapeutic overexpresses TYRO3. In one embodiment, the cancer that was previously treated with the chemotherapeutic overexpresses c-Met kinase. In one embodiment, the patient is treated with a compound of Formula I as a single agent. In one embodiment, the patient is treated with a combination of a compound of Formula 1 and the previously administered chemotherapeutic. In one embodiment, the chemotherapeutic is selected from taxane-based chemotherapies (e.g., docetaxel), dexamethasone, and cytarabine. In one embodiment, the patient is treated with a compound of Formula I as a single agent. In one embodiment, the patient is treated with a compound of Formula I in combination with the previously administered chemotherapeutic. In one embodiment, the compound of Formula I and the previously administered chemotherapeutic are administered as separate dosages sequentially in any order. In one embodiment, the cancer is selected from leukemias (including acute myeloid leukemia and chronic myeloid leukemia, B-cell acute lymphoblastic leukemia, and T-lineage acute lymphoblastic leukemia), non-small cell lung cancer, pancreatic ductal adenocarcinoma, astrocytoma, lung adenocarcinoma, ovarian cancer, melanoma, and glîoblastoma multiforme.
[001028] Also provided herein is (î) a pharmaceutical combination for treating a cancer in a patient in need thereof, which comprises (a) a compound of Formula I or a pharmaceutically acceptable sait thereof, and (b) at least one additional anticancer agent (e.g., any of the exemplary
195 additional anticancer agents described herein or known in the art), for simultaneous, separate or sequential use for the treatment of cancer, wherein the amounts ofthe compound of Formula I or pharmaceutically acceptable sait thereof and of the additional anticancer agent are together effective in treating the cancer; (ii) a pharmaceutical composition comprising such a combination; (iii) the use of such a combination for the préparation of a médicament for the treatment of cancer; and (iv) a commercial package or product comprising such a combination as a combined préparation for simultaneous, separate or sequential use; and to a method of treatment of cancer in a patient in need thereof. In one embodiment the patient is a human. In some embodiments, the cancer is a TAM-associated cancer.
[001029] The term pharmaceutical combination, as used herein, refers to a pharmaceutical therapy resulting from the mixing or combining of more than one active ingrédient and includes both fixed and non-fixed combinations of the active ingrédients. The term fixed combination” means that a compound of Formula I or a pharmaceutically acceptable sait thereof and at least one additional anticancer agent (e.g., a chemotherapeutic agent), are both administered to a patient simultaneously in the form of a single composition or dosage. The term non-fixed combination means that a compound of Formula I or a pharmaceutically acceptable sait thereof and at least one additional anticancer agent (e.g., chemotherapeutic agent) are formulated as separate compositions or dosages such that they may be administered to a patient in need thereof simultaneously, separately or sequentially with variable intervening time limits, wherein such administration provides effective levels of the two or more compounds in the body of the patient. These also apply to cocktail thérapies, e.g. the administration of three or more active ingrédients
[001030] Accordingly, also provided herein is a method of treating a cancer, comprising administering to a patient in need thereof a pharmaceutical combination for treating cancer which comprises (a) a compound of Formula I or pharmaceutically acceptable sait thereof, and (b) an additional anticancer agent for simultaneous, separate or sequential use for the treatment of cancer, wherein the amounts ofthe compound of Formula I or pharmaceutically acceptable sait thereof and the additional anticancer agent are together effective in treating the cancer. In one embodiment, the compound of Formula I or pharmaceutically acceptable sait thereof, and the additional anticancer agent are administered simultaneously as separate dosages. In one embodiment, the compound of Formula I or pharmaceutically acceptable sait thereof, and the additional anticancer agent are administered as separate dosages sequentially in any order, in
196 jointly therapeutically effective amounts, e.g. in daily or intermittently dosages. In one embodiment, the compound of Formula I or pharmaceutically acceptable sait thereof, and the additional anticancer agent are administered simultaneously as a combined dosage.
[001031] Accordingly, also provided herein are methods for inhibiting, preventing, aiding in the prévention, or decreasing the symptoms of metastasis of a cancer in a patient in need thereof, the method comprising administering to the patient a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof or a pharmaceutical composition thereof. Such methods can be used în the treatment of one or more of the cancers described herein. In some embodiments, the cancer is a TAM-associated cancer, a c-Metassociated cancer, or both. In some embodiments, the compound of Formula I or a pharmaceutically acceptable sait thereof is used in combination with an additional anticancer agent, including an immunotherapy.
[001032] The term “metastasis” is an art known term and means the formation of an additional tumor (e.g., a solid tumor) at a site distant from a primary tumor in a subject or patient, where the additional tumor includes the same or similar cancer eells as the primary tumor.
[001033] Also provided are methods of decreasing the risk of developing a metastasis or an additional metastasis in a patient having a TAM-associated cancer, a c-Met-associated cancer, or both, that include: selecting, identifying, or diagnosing a patient as having a TAM-associated cancer, a c-Met-associated cancer, or both, and administering a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof to the patient selected, identifîed, or diagnosed as having a TAM-associated cancer, a c-Met-associated cancer, or both. Also provided are methods of decreasing the risk of developing a metastasis or an additional metastasis in a patient having a TAM-associated cancer, a c-Met-associated cancer, or both, that includes administering a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait or solvent thereof to a patient having a TAM-associated cancer, a c-Met-associated cancer, or both. The decrease in the risk of developing a metastasis or an additional metastasis in a patient having a TAM-associated cancer, a c-Met-associated cancer, or both can be compared to the risk of developing a metastasis or an additional metastasis in the patient prior to treatment, or as compared to a patient or a population of patients having a similar or the same TAM-associated cancer, c-Met-associated cancer, or both, that has received no treatment or a different treatment.
197
[001034] The phrase “risk of developing a metastasis” means the risk that a subject or patient having a primary tumor will develop an additional tumor (e.g., a solid tumor) at a site distant from a primary tumor in a subject or patient over a set period of time, where the additional tumor includes the same or similar cancer cells as the primary tumor. Methods for reducing the risk of developing a metastasis in a subject or patient having a cancer are described herein.
[001035] The phrase “risk of developing additional métastasés” means the risk that a subject or patient having a primary tumor and one or more additional tumors at sites distant from the primary tumor (where the one or more additional tumors include the same or similar cancer cells as the primary tumor) will develop one or more further tumors distant from the primary tumor, where the further tumors include the same or similar cancer cells as the primary tumor. Methods for reducing the risk of developing additional metastasis are described herein.
[001036] Also provided is a method for inhibiting TAM kinase activity and/or inhibiting cMet kinase activity in a cell (e.g., a mammalian cell), comprising contacting the cell with a compound of Formula 1 or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition thereof. In one embodiment, the contacting is in vitro. Tn one embodiment, the contacting is in vivo. In one embodiment, the contacting is in vivo, wherein the method comprises administering an effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof to a subject having a cell having TAM kinase activity and/or c-Met kinase activity. In some embodiments, the cell is a cancer cell (e.g., a human cancer cell). In one embodiment, the cancer cell is any cancer as described herein. In some embodiments, the cancer cell is a TAMassociated cancer cell. In some embodiments, the cancer cell is a c-Met-associated cancer cell. In some embodiments, the cancer cell is both a TAM-associated cancer cell and a c-Met-associated cancer cell.
[001037] In some embodiments, the mammalian cell is in vitro. In some embodiments, the mammalian cell is in vivo. In some embodiments, the mammalian cell is ex vivo.
[001038] Also provided herein is a method of inhibiting cell prolifération, in vitro or in vivo, the method comprising contacting a cell with an effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition thereof as defïned herein.
[001039] Also provided herein are methods of decreasing immune tolérance in a subject in need thereof that include administering to the subject a therapeutically effective amount of a
198 compound of Formula I or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition thereof as defîned herein. As used herein, the term “immune tolérance” refers to a decrease (e.g., a 1 % to about 99% decrease, or any of the subranges of this range described herein) in one or more of: the processing of tumor-associated antigens by antigen-presenting cells (e.g., dendritic cells), présentation of antigens to tumor antigen-specific T cells, activation and prolifération of tumor antigen-specific T cells, and maintenance of the T-celi response in a subject (e.g., in a solid tumor in a subject), e.g., as compared to a contre! (e.g., a corresponding level in a similar subject that does not hâve a cancer)). In some embodiments of these methods, the subject has been identified or diagnosed as having a cancer (e.g., a TAM-associated cancer (e.g., any of the exemplary TAM-associated cancers described herein), a c-Met-associated cancer (e.g., any of the exemplary c-Met-associated cancers described herein), or both). In some examples, a decrease in immune tolérance in a subject can be detected by observing an about 1% to about 99% (e.g., about 1% to about 95%, about 1% to about 90%, about 1% to about 85%, about 1% to about 80%, about 1% to about 75%, about 1% to about 70%, about 1% to about 65%, about 1% to about 60%, about 1% to about 55%, about 1% to about 50%, about 1% to about 45%, about 1% to about 40%, about 1% to about 35%, about 1% to about 30%, about 1% to about 25%, about 1% to about 20%, about 1% to about 15%, about 1% to about 10%, about 1% to about 5%, about 5% to about 99%, about 5% to about 90%, about 5% to about 85%, about 5% to about 80%, about 5% to about 75%, about 5% to about 70%, about 5% to about 65%, about 5% to about 60%, about 5% to about 55%, about 5% to about 50%, about 5% to about 45%, about 5% to about 40%, about 5% to about 35%, about 5% to about 30%, about 5% to about 25%, about 5% to about 20%, about 5% to about 10%, about 10% to about 99%, about 10% to about 95%, about 10% to about 90%, about 10% to about 85%, about 10% to about 80%, about 10% to about 75%, about 10% to about 70%, about 10% to about 65%, about 10% to about 60%, about 10% to about 55%, about 10% to about 50%, about 10%to about 45%, about 10% to about 40%, about 10% to about 35%, about 10%to about 30%, about 10% to about 25%, about 10% to about 20%, about 10% to about 15%, about 15% to about 99%, about 15% to about 95%, about 15% to about 90%, about 15% to about 85%, about 15% to about 80%, about 15% to about 75%, about 15% to about 70%, about 15% to about 65%, about 15% to about 60%, about 15% to about 55%, about 15% to about 50%, about 15% to about 45%, about 15% to about 40%, about 15% to about 35%, about 15% to about 30%, about 15% to about 25%, about 15% to about 20%, about 20% to about 99%, about 20% to about 95%, about 20% to
199 about 90%, about 20% to about 85%, about 20% to about 80%, about 20% to about 75%, about 20% to about 70%, about 20% to about 65%, about 20% to about 60%, about 20% to about 55%, about 20% to about 50%, about 20% to about 45%, about 20% to about 40%, about 20% to about 35%, about 20% to about 30%, about 20% to about 25%, about 25% to about 99%, about 25% to 5 about 95%, about 25% to about 90%, about 25% to about 85%, about 25% to about 80%, about
25% to about 75%, about 25% to about 70%, about 25% to about 65%, about 25% to about 60%, about 25% to about 55%, about 25% to about 50%, about 25% to about 45%, about 25% to about 40%, about 25% to about 35%, about 25% to about 30%, about 30% to about 99%, about 30% to about 95%, about 30% to about 90%, about 30% to about 85%, about 30% to about 80%, about 10 30% to about 75%, about 30% to about 70%, about 30% to about 65%, about 30% to about 60%, about 30% to about 55%, about 30% to about 50%, about 30% to about 45%, about 30% to about 40%, about 30% to about 35%, about 35% to about 99%, about 35% to about 95%, about 35% to about 90%, about 35% to about 85%, about 35% to about 80%, about 35% to about 75%, about 35% to about 70%, about 35% to about 65%, about 35% to about 60%, about 35% to about 55%, 15 about 35% to about 50%, about 35% to about 45%, about 35% to about 40%, about 40% to about
99%, about 40% to about 95%, about 40% to about 90%, about 40% to about 85%, about 40% to about 80%, about 40% to about 75%, about 40% to about 70%, about 40% to about 65%, about 40% to about 60%, about 40% to about 55%, about 40% to about 50%, about 40% to about 45%, about 45% to about 99%, about 45% to about 95%, about 45% to about 90%, about 45% to about 20 85%, about 45% to about 80%, about 45% to about 75%, about 45% to about 70%, about 45% to about 65%, about 45% to about 60%, about 45% to about 55%, about 45% to about 50%, about 50% to about 99%, about 50% to about 95%, about 50% to about 90%, about 50% to about 85%, about 50% to about 80%, about 50% to about 75%, about 50% to about 70%, about 50% to about 65%, about 50% to about 60%, about 50% to about 55%, about 55% to about 99%, about 55% to 25 about 95%, about 55% to about 90%, about 55% to about 85%, about 55% to about 80%, about
55% to about 75%, about 55% to about 70%, about 55% to about 65%, about 55% to about 60%, about 60% to about 99%, about 60% to about 95%, about 60% to about 90%, about 60% to about 85%, about 60% to about 80%, about 60% to about 75%, about 60% to about 70%, about 60% to about 65%, about 65% to about 99%, about 65% to about 95%, about 65% to about 90%, about 3 0 65% to about 85%, about 65% to about 80%, about 65% to about 75%, about 65% to about 70%, about 70% to about 99%, about 70% to about 95%, about 70% to about 90%, about 70% to about
200
85%, about 70% to about 80%, about 70% to about 75%, about 75% to about 99%, about 75% to about 95%, about 75% to about 90%, about 75% to about 85%, about 75% to about 80%, about 80% to about 99%, about 80% to about 95%, about 80% to about 90%, about 80% to about 85%, about 85% to about 99%, about 85% to about 95%, about 85% to about 90%, about 90% to about 99%, about 90% to about 95%, or about 95% to about 99%) decrease in the level of myeloidderived suppressor cells (MDSCs) (e.g., cells characterized by expression of CD33, CD 14, and low levels of HLA DR) in the subject (e.g., in a sample comprising blood or a biopsy sample obtained from the subject) (e.g., as compared to the level of MDSCs in the subject prior to administration of treatment (e.g., prior to administration of any of the compounds of Formula I or any of the pharmaceutical compositions described herein).
[001040] In some examples, a decrease in immune tolérance in a subject can be detected by observîng an about 1% to about 99% (or any of the subranges of this range described herein) decrease in the level of Treg cells (e.g., cells characterized by expression of CD4, FOXP3, and CD25) in the subject (e.g., in a sample comprising blood or a biopsy sample obtained from the subject) (e.g., as compared to the level of Tregs in the subject prior to administration of treatment (e.g., prior to administration of any of the compounds of Formula I or any of the pharmaceutical compositions described herein).
[001041] In some examples, a decrease in immune tolérance in a subject can be detected by observîng an about 1% to about 99% (or any of the subranges of this range described herein) decrease in the level of dendritic cells with reduced expression of CD80/CD86 in the subject (e.g., in a sample comprising blood or a biopsy sample obtained from the subject) (e.g., as compared to the level of dendritic cells with reduced expression of CD80/CD86 in the subject prior to administration of treatment (e.g., prior to administration of any of the compounds of Formula I or any of the pharmaceutical compositions described herein). Exemplary methods for detecting the levels of MDSCs, Tregs, and dendritic cells with reduced expression of CD80/CD86 include, fluorescence-assisted cell sorting and immunofluorescence microscopy.
[001042] Also provided herein are methods of inhibiting angiogenesis în a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition thereof as defined herein. In some embodiments, the angiogenesis is tumor angiogenesis and the subject has been identified or diagnosed as having a cancer (e.g., a TAM
201 associated cancer, a c-Met-associated cancer, or both). In some embodiments, these methods resuit in a decrease (e.g., a 1% to about 99% decrease, or any of the subranges of this range described herein) in the rate of development of new blood vessels (e.g., as compared to the rate of development of new blood vessels in a similar subject administered a placebo or a different treatment over a similar period of time). Exemplary methods for detecting the formation of new blood vessels include Doppler ultrasound (e.g., Color Dopler Flow Imaging), Ultrasound-Guided Diffus Optical Tomography, MRI, perfusion CT (also called functional multi-detector row CT (fMDCT)), positron émission tomography (PET), dynamic MRI, dynamic susceptibility contrast enhanced MRI (DSC-MRI), and Tl-weighted dynamic MRI (DCE-MRI). Non-limiting methods that can be used to detect the formation of new blood vessels (angiogenesis) are described in Jeswani et al„ Cancer Imaging 5(1):131-138, 2005.
[001043] Also provided herein are methods of suppressing (e.g., decreasing, e.g., a 1% to about 99% decrease, or any of the subranges of this range described herein) résistance to a therapeutic agent in a subject in need thereof that include administering to the subject a therapeutically effective amount of (i) a compound of Formula I or a pharmaceutically acceptable sait thereof, or any of the pharmaceutical compositions thereof described herein, and (ii) the therapeutic agent, where the therapeutic agent is selected from the group consisting of a chemotherapeutîc agent, a PI-3 kinase inhibitor, an EGFR inhibitor, a HER2/neu inhibitor, an FGFR inhibitor, an ALK inhibitor, an IGF! R inhibitor, a VEGFR inhibitor, a PDGFR inhibitor, a glucocorticoid, a BRAF inhibitor, a MEK inhibitor, a HER4 inhibitor, a MET inhibitor, a RAF inhibitor, an Akt inhibitor, a FTL-3 inhibitor, and a MAP kinase pathway inhibitor. In some examples of these methods, the c-Met inhibitor is a Type 1 c-Met inhibitor, e.g., crizotinib, capmatinib, NVP-BVU972, AMG 337, bozitinib, glumetinib, savolitinib, or tepotinib. In some examples of these methods, the compound of Formula I or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition thereof, and the therapeutic agent, are administered to the subject at substantially the same time. In some embodiments of these methods, the compound of Formula I or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition thereof, and the therapeutic agent, are formulated in a single dosage form. In some embodiments of these methods, (î) the compound of Formula I or a pharmaceutically sait thereof, or any of the pharmaceutical compositions thereof described herein is administered to the subject priorto administration of (ii) the therapeutic agent to the subject. In some embodiments of these methods, (ii) the therapeutic
202 agent is administered to the subject prior to administration of (i) the compound of Formula I or a pharmaceutically sait thereof, or any of the pharmaceutical compositions thereof described herein. [ÛÛ1044| In some embodiments of these methods, the compound of Formula I or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition thereof, is administered to the subject prior to administration of the therapeutic agent to the subject. In some embodiments of these methods, the therapeutic agent is administered to the subject prior to administration of the compound of Formula I or a pharmaceutically acceptable sait thereof, or a pharmaceutical composition thereof, to the subject.
[001045] As used herein, the term “résistance to a therapeutic agent” refers to a reduced or decreased level of sensitivity to treatment with a therapeutic agent (e.g., a chemotherapeutic agent, a PI-3 kinase inhibitor, an EGFR inhibitor, a HER2/neu inhibitor, an FGFR inhibitor, an ALK inhibitor, an IGF1R inhibitor, a VEGFR inhibitor, a PDGFR inhibitor, a glucocorttcoid, a BRAF inhibitor, a MEK inhibitor, a HER4 inhibitor, a MET inhibitor (e.g., a Type l c-Met kinase inhibitor, e.g., crizotinib, capmatinib, and NVP-BVLJ972), a RAF inhibitor, an Akt inhibitor, a FTL-3 inhibitor, and a MAP kinase pathway inhibitor) in a subject (e.g., as compared to a similar subject or as compared to the level of sensitivity to the therapeutic agent at an earlier time point). For example, résistance to an therapeutic agent in a subject can be observed by a physician, e.g., by observing the requirement of a increasing dosage amounts of a therapeutic agent over time in orderto achievethe same therapeutic effect in a subject, observing the requirement for an increased number of doses and/or an increased frequency of doses of a therapeutic agent over time in order to achieve the same therapeutic effect in a subject, a decrease in the observed therapeutic response to treatment with the same dosage of a therapeutic agent over time, or an observed progression of dîsease or dîsease relapse in a subject administered a therapeutic agent.
[001046] When employed as pharmaceuticals, the compounds of Formula I can be administered in the form of pharmaceutical compositions. These compositions can be prepared in a manner well known in the pharmaceutical art, and can be administered by a variety of routes, depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration may be topical (including transdermal, epidermai, ophthalmic and to mucous membranes including intranasal, vaginal and rectal delivery), pulmonary (e.g., by inhalation or insufflation of powders or aérosols, including by nebulizer; intratracheal or intranasal), oral or parenteraL Oral administration can include a dosage form formulated for once-daily or twice-daily
203 (BID) administration. Parentéral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal intramuscular or injection or infusion; or intracranial, e.g., intrathecal or intraventricular, administration. Parentéral administration can be in the form of a single bolus dose, or may be, for example, by a continuous perfusion pump. Pharmaceutical compositions and formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders. Conventional pharmaceutical carriers, aqueous, powder or oîly bases, thickeners and the like may be necessary or désirable. In one embodiment, a compound of Formula I is formulated as a tablet. In one embodiment, a compound of Formula I is formulated as a capsule. In one embodiment, a compound of Formula I is administered orally. In one embodiment, a compound of Formula I is administered orally once a day. In one embodiment, a compound of Formula 1 is administered orally twice a day.
[001047] Also provided herein are pharmaceutical compositions which contain, as the active ingrédient, a compound of Formula I or a pharmaceutically acceptable sait thereof, in combination with one or more pharmaceutically acceptable carriers (excipients). In some embodiments, the composition is suitable for topical administration. In making the compositions provided herein, the active ingrédient is typically mixed with an excipient, diluted by an excipient or enclosed within such a carrier in the form of, for example, a capsule, sachet, paper, or other container. When the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingrédient. Thus, the compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, élixirs, suspensions, émulsions, solutions, syrups, aérosols (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, stérile injectable solutions, and stérile packaged powders. In one embodiment, the composition is formulated for oral administration. In one embodiment, the composition is formulated as a tablet or capsule.
[001048] The compositions comprising a compound of Formula I or a pharmaceutically acceptable sait thereof can be formulated in a unit dosage form, each dosage containing from about 5 to about 1,000 mg (1 g), more usually about 100 mg to about 500 mg, of the active ingrédient. The term unit dosage form refers to physically discrète units suitable as unitary dosages for human subjects and other patients, each unît containing a predetermined quantity of active material (i.e., a compound for Formula I as provided herein) calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
204
[001049] In some embodiments, the compositions provided herein contain from about 5 mg to about 50 mg of the active ingrédient. One having ordinary skill in the art will appreciate that this embodies compounds or compositions containing about 5 mg to about 10 mg, about 10 mg to about 15 mg, about 15 mg to about 20 mg, about 20 mg to about 25 mg, about 25 mg to about 30 mg, about 30 mg to about 35 mg, about 35 mg to about 40 mg, about 40 mg to about 45 mg, or about 45 mg to about 50 mg of the active ingrédient.
[001050] In some embodiments, the compositions provided herein contain from about 50 mg to about 500 mg of the active ingrédient. One having ordinary skill in the art will appreciate that this embodies compounds or compositions containing about 50 mg to about 100 mg, about 100 mg to about 150 mg, about 150 mg to about 200 mg, about 200 mg to about 250 mg, about 250 mg to about 300 mg, about 350 mg to about 400 mg, or about 450 mg to about 500 mg of the active ingrédient.
[001051] In some embodiments, the compositions provided herein contain from about 500 mg to about 1,000 mg of the active ingrédient. One having ordinary skill in the art will appreciate that this embodies compounds or compositions containing about 500 mg to about 550 mg, about 550 mg to about 600 mg, about 600 mg to about 650 mg, about 650 mg to about 700 mg, about 700 mg to about 750 mg, about 750 mg to about 800 mg, about 800 mg to about 850 mg, about 850 mg to about 900 mg, about 900 mg to about 950 mg, or about 950 mg to about 1,000 mg of the active ingrédient.
|001052] The active compound may be effective over a wide dosage range and is generally administered in a pharmaceutically effective amount. It will be understood, however, that the amount of the compound actually administered will usually be determined by a physician, according to the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the âge, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
[001053] In some embodiments, the compounds provided herein can be administered in an amount ranging from about 1 mg/kg to about 100 mg/kg. In some embodiments, the compound provided herein can be administered in an amount of about 1 mg/kg to about 20 mg/kg, about 5 mg/kg to about 50 mg/kg, about 10 mg/kg to about 40 mg/kg, about 15 mg/kg to about 45 mg/kg, about 20 mg/kg to about 60 mg/kg, or about 40 mg/kg to about 70 mg/kg. For example, about 5 mg/kg, about 10 mg/kg, about 15 mg/kg, about 20 mg/kg, about 25 mg/kg, about 30 mg/kg, about
205 mg/kg, about 40 mg/kg, about 45 mg/kg, about 50 mg/kg, about 55 mg/kg, about 60 mg/kg, about 65 mg/kg, about 70 mg/kg, about 75 mg/kg, about 80 mg/kg, about 85 mg/kg, about 90 mg/kg, about 95 mg/kg, or about 100 mg/kg. In some embodiments, such administration can be once-daily or twice-daily (B1D) administration.
[001054] One skilled in the art will recognize that, both in vivo and in vitro trials using suitable, known and generally accepted cell and/or animal tnodels are prédictive of the ability of a test compound to treat or prevent a given disorder.
[001055] One skilled in the art will further recognize that human clinical trials including firstin-human, dose ranging and effîcacy trials, in healthy patients and/or those suffering from a given disorder, may be completed according to methods well known in the clinical and medical arts.
Examples
[001056] The following examples illustrate the invention.
Biological Examples
[0Ü1057] Example A
[001058] AXL Enzvme Assay
[001059] Compounds of Formula I were screened fortheir ability to inhibit AXL kinase using Invitrogen’s LanthaScreen™ Eu Kinase Binding technology. His-tagged recombinant human AXL cytoplasmic domain was incubated with 20 nM Alexa-Fluor® Tracer 236 (PR9078A), 2 nM biotinylated anti-His (Cat. No. M4408), and 2 nM europium-labeled Streptavidin (Cat. No. PV5899) along with test compound in a buffer consisting of 25 mM HEPES, pH 7.4, 10 mM MgCh, 0.01% Triton X-IÛ0, and 2% DMSO. Compounds were typically prepared in a threefold serial dilution in DMSO and added to the assay to give the appropriate final concentration. After a 60-minute incubation at 22°C, the reaction was measured using a PerkinElmer EnVision multimode plate reader via TR-FRET dual wavelength détection, and the percent of control (POC) was calculated using a ratiometric émission factor. 100 POC was determined using no test compounds and 0 POC was determined using a concentration of control compound that completely inhibits the enzyme. The POC values are fît to a 4 parameter logistic curve and the ICso value is point where the curve crosses 50 POC.
[001060] Example B
[001061] MER Enzvme Assay
206
[001062] Compounds of Formula I were screened fortheirability to inhibit AXL kinase using Invitrogen’s LanthaScreen™ Eu Kinase Binding technology. His-tagged recombinant human MER cytoplasmic domain (5 nM) was incubated with 20 nM Alexa-Fluor® Tracer 236 (PR9078A), 2 nM biotinylated anti-His (Cat. No. M4408), and 2 nM europium-labeled Streptavidin (Cat. No. PV5899) along with test compound in a buffer consisting of 25 rnM HEPES, pH 7.4, 10 mM MgCh, 0.01% Triton X-100, and 2% DMSO. Compounds were typically prepared in a threefold serial dilution in DMSO and added to the assay to give the appropriate final concentration. After a 60-minute incubation at 22°C, the réaction was measured using a PerkinElmer EnVision multimode plate reader via TR-FRET dual wavelength détection, and the percent of control (POC) was calculated using a ratiometric émission factor. 100 POC was determined using no test compounds and 0 POC was determined using a concentration of control compound that completeiy inhibits the enzyme. The POC values are fit to a 4 parameter logistic curve and the ICso value is point where the curve crosses 50 POC.
[001063] Example C
[001064] TYRO3 Enzyme Assay
[001065] Compounds of Formula I were screened for their ability to inhibit TYRO3 kinase using Invitrogen’s LanthaScreen™ Eu Kinase Binding technology. GST-tagged recombinant human TYRO3 kinase domain from Carna (5 nM; Cat. No. PR7480A) was incubated with 20 nM Alexa-Fluor® Tracer 236 (PR9078A) and 2 nM Europium-anti-G ST (Cat. No. A 151 16) along with test compound in a buffer consisting of 25 rnM HEPES, pH 7.4, 10 mM MgCb, 0.01% Triton X-100, and 2% DMSO. Compounds are typically prepared in a threefold serial dilution in DMSO and added to the assay to give the appropriate final concentration. After a 60-minute incubation at 22°C, the reaction was measured using a PerkinElmer EnVision multimode plate reader via TRFRET dual wavelength détection, and the percent of control (POC) calculated using a ratiometric émission factor. 100 POC was determined using no test compounds and 0 POC was determined using a concentration of control compound that completeiy inhibits the enzyme. The POC values were fit to a 4 parameter logistic curve and the IC50 value is point where the curve crosses 50 POC.
[001066] The averaged ICso's of compounds tested in the assays of Examples A, B and C are shown in Table 7.
207
Table 7
Ex. AXL enzyme 1C5O MER enzyme IC 50 TYRO3 enzyme IC50
1 6.1 13.2 21.6
2 2.7 3.9 10.7
3 1.8 4.7 46.7
4 1.8 3.6 18.2
5 4.6 8.3 30.2
6 5.6 5.1 15.4
7 3.6 5.1 16.5
8 3.4 5.1 13.8
9 4.4 8.3 57.9
10 7.1 11.4 45.7
11 9.4 18.3 86
12 18.3 36.8 295.4
13 4 7.2 16.9
14 5.4 11.6 65.7
15 4.4 10,4 48.1
16 2 4.1 34.1
17 2.8 8.4 56.8
18 3.8 7.3 131.1
19 16.1 24.6 85.6
20 18.9 51.9 535.8
21 12.7 18.1 187.7
22 18.3 29.5 317.9
23 4 6.5 29.6
24 7.1 13.1 119.6
25 1.4 2.3 7.7
208
Ex. AXL enzyme IC50 MER enzyme ICso TYRO3 enzyme IC50
26 3.9 4.9 41.1
27 3.3 11.3 64.6
28 19.1 37.9 374.9
29 5.4 11.9 51.9
30 6.1 12.1 79.3
31 4.1 7.5 76.8
32 2.2 5.7 37.3
33 0.8 2.1 14.9
34 1.2 2.8 19.6
35 3.8 8.8 26.7
36 2.8 4.1 13.1
37 2.4 3.5 8.4
38 1.1 3.1 29.1
39 3.8 11.6 96.3
40 2.5 7.6 98.3
41 0.9 3.9 13.5
42 1.1 7.4 53.1
43 2.8 2.6 18.5
44 7.3 19.4 109.1
45 1.3 2.8 12.1
46 2 2.7 9
47 1.1 2.4 16.9
48 2 2.7 9.8
49 2.2 5.8 23.9
50 4.4 9.7 40.3
51 1.3 4.1 18
209
Ex. AXL enzyme 1C5O MER enzyme IC50 TYRO3 enzyme IC50
52 1.8 3.9 28.4
53 1.8 4.6 31.7
54 3.5 4 41.2
55 1.1 2 7.4
56 1.5 3.5 20.7
57 1.9 4.1 20.8
58 2.2 4.3 9.7
59 1.8 3.7 25.4
60 5.9 6.1 28.1
61 2.6 4.1 22.5
62 3.3 7 41.6
63 5.6 8.2 37.7
64 3.8 5.1 19.2
65 1 5.4 65.6
66 1.3 1.9 10.9
67 1.1 3.4 10.8
68 1.3 5.5 32.3
69 1.4 5.7 26.5
70 1 5.1 25.7
71 5.2 8.5 192.1
72 0.9 2.3 6
73 1.1 10.8 23.8
74 1.6 32.5 97.5
75 0.6 3.8 22.2
76 0.5 2.5 4.2
77 0.7 2.8 7.2
210
Ex. AXL enzyme IC50 MER enzyme IC50 TYRO3 enzyme IC50
78 1 3.1 8.2
79 1.2 2.9 19.7
80 1.6 4.1 38
81 1.8 4 21.8
82 1.6 5.3 19.3
83 0.5 1.6 7.2
84 0.6 1.9 4.3
85 1 1.7 4.4
86 4.8 11 104.1
87 12.6 33.4 432.5
88 6.5 30.2 363.5
89 0.7 19.1 74.3
90 0.8 13.1 76.6
91 1.1 4.7 4
92 1.5 4.4 16.3
93 4.7 259.6 766.6
94 2.9 8.7 19
95 15.4 176.5 874.8
96 2.9 9.6 19.7
97 1.2 3.1 4.8
98 1.3 5.6 12.4
99 5.7 92.8 545.8
100 1.6 1.6 3.9
101 49.7 60.4 198.2
102 2 3.3 44.1
103 0.6 0.9 3.2
211
Ex. AXL enzyme IC50 MER enzyme IC50 TYRO3 enzyme IC5o
104 1.8 2.6 14.1
105 1.1 4 9.8
106 1.6 6.8 16.1
107 2.3 3.3 9.2
108 0.9 2 6.1
109 0.9 5.2 18.7
110 1.3 6.6 38.5
111 4.4 12.5 136.9
112 1.6 6.4 70
113 1.8 5.6 60.4
114 1.5 5.2 7.1
115 1.2 4.3 6.4
116 1.4 4.5 11.4
117 0.8 3.1 13.6
118 1.8 6.1 26.2
119 1.8 3.1 4.7
120 5 6.2 27.3
121 1.6 2.1 4.6
122 3.4 9 44.7
123 1.3 3.8 15.3
124 1.8 3.5 8.3
125 1.3 2.7 7.1
126 1.5 2.4 6.7
127 1.1 2.3 3.7
128 1.2 5.3 23
129 0.8 1.8 3.2
212
Ex. AXL enzyme IC50 MER enzyme IC50 TYRO3 enzyme IC50
130 1.3 5.8 27.1
131 2.1 9.1 47.4
132 8.6 70 347.9
133 5.3 37.8 129.9
134 4.6 32.8 169.2
135 3.5 17.2 64.4
136 3.3 26.7 88.8
137 5.8 27.4 74.3
138 6.2 36.7 159
139 1.7 10.8 86.2
140 354.6 161.2 1000
141 484.6 178.1 1000
142 3.2 5.7 15.6
143 3.1 5.7 41.2
144 2.4 9.6 88.8
145 0.8 3.3 20.9
146 1.8 7 46.5
147 5.6 45.9 91.2
148 1.3 3.1 12.2
149 2 4.1 20
150 1.6 4.4 13.6
151 2 2.2 2.3
152 1.3 4.9 8.7
153 2 7.9 28.2
154 1.6 5.9 14.4
155 1.6 5.6 59.5
213
Ex. AXL enzyme IC50 MER enzyme ICso TYRO3 enzyme IC50
156 1.2 5.2 59.5
157 1.4 6.2 118.3
158 2 13.4 104
159 2.2 18.7 420.2
160 1.3 2.7 15.4
161 1.9 4 28
162 1.3 5.4 37.9
163 1.1 2.6 5.5
164 1.7 11.8 68.9
165 3.3 13.7 90
166 22.7 72.5 475.9
167 3.4 19.8 224.5
168 4.9 11.9 248
169 0.7 1.5 4.4
170 1.9 3.9 36.2
171 3 27.1 333.3
172 4.2 8.3 132.7
173 7.1 15.7 114.1
174 1.4 4.6 98.6
175 5.6 23.2 292.2
176 4.2 22 728
177 1.6 3.5 106.1
178 3.6 11.5 439.2
179 1.3 6 80.6
ISO 2 11.9 140
181 2.5 4.1 30.8
214
Ex. AXL enzyme IC50 MER enzyme ÎC50 TYRO3 enzyme IC50
182 8.6 16.7 706.8
183 5.4 18 108.7
184 1.8 2.7 35.2
185 1.3 2.3 18.4
186 6.6 18.9 217.4
187 1.9 3.5 11.9
188 1.9 1.7 7.8
189 5.8 7.5 69.6
190 1.2 1.8 5
191 4.8 8.9 73.2
192 3.5 10.8 61.4
193 2.2 6.5 12.1
194 4.8 11.2 97.4
195 1.5 3.8 13.4
196 1 10.5 22
197 1.1 12.7 52.3
198 1.3 5.5 23.3
199 1.6 2.7 4.5
200 1.3 1.9 3.4
201 1.3 2.2 3.8
Example D.
c-Met Enzyme Assay
Experimental
The affinity of compound binding to wild type and mutant human MET kinases is measured
215 using Invitrogen’s LanthaScreen™ Eu Kinase Binding technology. Briefly, GST-tagged recombinant human MET kinase domain from Signal Chem (see Table 8 below for concentration in assay) is incubated with 50 nM Alexa-Fluor® Tracer 236 (Invitrogen Cat No. PR9078A) and 2 nM Europium-anti-GST (Invitrogen Cat. No. Al 5116) along with test compound in a buffer consisting of 25 mM HEPES, pH 7.4, 10 mM MgC12, 0.01% Triton X-100, ImM DTT, and 2% DMSO. Compounds are typically prepared in a three-fold serial dilution in DMSO and added to the assay to give the appropriate final concentration. After a 60-minute incubation at 22 °C, the reaction is measured using a PerkinElmer EnVision multimode plate reader via TR-FRET dual wavelength détection, and the percent of control (POC) calculated using a ratiometric émission factor. 100 POC is determined using no test compounds and 0 POC is determined using a concentration of control compound that completely inhibits the enzyme. The POC values are fit to a 4 parameter logistic curve and the IC50 value is point where the curve crosses 50 POC.
216
Table 8. Concentration of Wild Type and Mutant MET kinases in binding assay
Met Mutant Enzyme Source Catalog Number MET Amino Acids Enzyme Concentration in Binding Assay (nM)
de] Ex 14 SignalChem M52-12PG 956-1390 (end) 5
L1195V SignalChem NP-18-156G 956-1390 (end) 10
Fl 2001 SignalChem M52-12GG 956-1390 (end) 2
D1228H SignalChem M52-12HG 956-1390 (end) 2
D1228N SignalChem M52-121G 956-1390 (end) 2
Y1230C SignalChem M52-12KG 956-1390 (end) 2
Y1230H SignalChem M52-12MG 956-1390 (end) 5
Y1230S SignalChem NP18-157G 956-1390 (end) 8
MET (wt) SignalChem M52-18G 956-1390 (end) 10
Res u Its
Table 9. IC50 of Inhibition of Wild Type and Mutation MET kinases of Exemplary Tested
Compounds
Ex.# SigChem WT IC5O (nM) Dell4 ICS0 (nM) D1228H IC 50 (nM) D1228N IC50 (nM) F1200I IC50 (nM) L1195V IC 50 (nM) Y1230C IC50 (nM) Y123OH IC50 (nM) Y1230S ICSO (nM)
2 3.9 13.0 11.2 7.6 6.8 125.9 3.9 15.8 8.2
3 3.7 9.6 16.6 5.2 3.6 133.4 3.9 3.9 3.9
4 3.0 9.3 19.7 3.8 4.9 116.7 10.1 9.4 6.3
5 3.3 7.6 21.2 4.3 5.1 116.1 3.1 7.7 5.0
6 9.2 31.4 61.5 13.5 11.3 244.1 11.2 22.1 13.8
7 12.4 28.2 51.6 15.4 18.8 452.8 15.4 26.5 16.0
8 3.0 12.8 19.7 5.4 6.2 145.2 4.2 11.8 4.8
12 17.6 92.1 161.5 19.8 24.0 828.8 50.0 53.5 39.9
13 3.7 9.7 22.7 3.2 4.4 121.0 4.6 7.9 4.2
14 17.5 104.4 181.7 24.1 15.8 961.5 31.5 48.4 41.3
16 3.2 8.9 22.6 5.3 4.4 101.6 3.5 10.9 4.5
17 26.5 107.7 183.6 38.5 34.2 1383.4 44.4 59.1 30.9
18 49.8 116.4 435.0 87.4 75.2 3787.7 147.6 173.9 123.0
19 40.1 130.3 266.9 56.5 81.0 2060.5 121.4 125.2 100.4
20 19.8 85.4 169.7 48.6 41.3 919.2 30.8 81.8 52.2
217
Ex. tt SigChem WTIC50 (nM) Dell4 IC5Q (nM) D1228H IC50 (nM) D1228N IC50 (nM) F1200I IC50 (nM) L1195V IC5O (nM) Y1230C IC50 (nM) Y1230H 1C50 (nM) Y1230S IC50 (nM)
21 21.7 80.0 143.4 31.0 36.8 2122.0 68.9 92.8 59.5
22 22.1 78.1 141.8 63.3 48.1 2861.3 41.3 94.8 30.8
23 7.7 23.3 36.9 12.2 13.1 514.7 13.7 18.3 15.7
24 13.9 101.7 132.1 19.2 25.0 1520.4 29.5 56.9 26.0
25 2.5 6.9 11.5 4.2 3.4 84.4 2.9 7.3 3.3
26 21.2 63.7 113.2 31.4 31.4 911.9 31.8 61.8 44.8
27 2.7 7.6 14.1 5.0 3.3 77.1 3.7 8.5 5.0
28 1.2 22.9 49.0 16.3 11.9 508.1 11.5 22.0 22.6
29 3.9 14.6 30.9 7.5 7.1 6.9 16.1 8.8
30 4.4 19.6 28.6 6.2 4.9 166.1 6.3 12.4 6.1
31 10.1 31.8 52.9 15.7 15.1 415.7 16.6 22.0 15.4
32 3.4 7.2 9.6 5.4 4.3 101.7 3.8 5.6 3.5
33 23.6 62.5 132.2 35.8 28.2 1107.5 29.6 64.3 26.4
34 1.5 3.9 6.8 2.3 2.6 60.7 2.9 5.0 2.4
35 10.6 31.0 51.7 16.5 17.2 347.7 19.3 26.6 15.6
36 3.4 10.2 14.2 4.4 4.1 83.3 2.4 9.2 4.8
37 1.1 4.6 6.1 1.9 1.3 44.6 1.3 3.9 2.2
38 9.4 21.5 42.7 13.0 13.2 280.4 12.8 25.0 10.7
39 9.5 25.4 48.7 13.8 13.2 451.2 19.5 26.5 10.5
40 4.9 39.5 59.1 8.7 15.6 487.8 17.2 19.8 9.4
41 2.6 9.3 14.3 3.3 3.6 89.5 3.6 7.6 2.7
42 8.5 31.6 38.5 11.1 9.5 297.8 6.6 19.4 6.0
43 6.3 12.0 23.8 9.1 7.4 292.6 10.5 13.9 5.4
44 9.7 42.1 57.3 12.0 15.0 641.0 16.3 24.0 12.0
45 1.9 3.8 4.9 3.1 1.4 75.3 1.4 2.6 1.8
46 12.2 13.9 21.5 12.5 9.7 102.2 9.5 15.9 10.2
47 4.7 9.0 16.3 5.8 8.0 236.8 6.3 12.0 15.0
50 8.0 23.3 26.7 11.9 13.5 446.4 8.2 22.2 15.6
51 3.8 7.0 7.8 6.7 4.9 68.8 5.4 5.2 5.4
52 5.9 10.7 11.7 12.0 8.1 141.3 7.2 7.8 6.3
53 18.1 76.0 109.6 32.0 23.6 772.6 21.3 48.8 28.1
54 7.3 33.5 42.8 9.5 12.9 516.5 15.4 21.6 11.3
218
Ex.# SigChem WTIC50 |nM) Dell4 IC50 (nM) D1228H IC50 (nM) D1228N IC50 (nM) F1200I IC50 (nM) 11195V IC50 (nM) Y1230C IC50 (nM) Y1230H IC50 (nM) Y1230S IC50 (nM)
55 2.6 6.0 7.7 5.4 3.2 34.4 2.6 4.3 2.8
56 2.9 8.4 14.8 3.4 3.6 75.8 3.0 4.9 3.2
57 18.2 33.0 120.0 15.9 15.4 575.7 10.7 43.9 18.3
58 7.2 12.7 34.8 7.6 7.5 256.8 9.1 19.9 9.7
59 5.7 31.0 38.1 9.6 7.8 347.0 9.3 7.9 8.4
60 8.2 21.3 39.2 15.3 11.5 363.1 21.2 17.4 7.9
61 3.0 4.6 11.7 3.4 3.8 75.9 2.9 5.8 4.7
62 29.4 81.0 129.4 30.9 31.9 1187.8 37.2 50.6 42.6
63 6.9 15.3 36.6 5.3 8.9 321.3 9.0 13.0 6.8
64 15.5 28.2 46.6 23.6 20.3 18.3 37.2 21.1
65 3.5 4.6 16.3 10.1 5.0 194.6 3.6 10.1 4.8
67 2.6 1.8 3.4 4.0 1.4 9.9 1.1 3.2 2.6
68 2.7 4.9 6.6 4.2 4.6 33.4 2.3 4.6 3.2
69 5.1 5.7 7.1 7.5 7.0 55.9 3.9 5.9 5.3
70 7.0 12.1 13.6 12.7 9.4 67.9 6.4 7.6 4.5
71 38.5 183.1 302.8 38.5 62.3 2169.7 79.3 88.8 89.8
72 2.0 1.7 2.0 2.3 2.3 8.4 1.2 2.5 1.0
73 4.3 4.6 4.3 5.6 4.1 27.1 2.6 4.6 2.8
75 3.5 7.2 7.6 4.8 12.8 40.7 3.1 4.9 2.7
76 2.2 6.0 4.9 3.8 3.1 29.7 1.7 4.6 1.8
77 2.5 4.8 4.1 4.3 2.2 36.0 1.5 5.8
79 3.9 4.0 3.6 3.6 2.2 18.1 1.5 4.6 2.7
80 4.3 4.9 6.2 4.9 3.0 19.5 3.0 4.0 3.6
81 3.2 1.7 2.7 3.4 2.2 5.9 2.7 2.5 1.4
84 1.5 1.0 1.5 2.2 2.0 6.0 1.7 1.2 1.2
86 19.0 81.8 96.4 36.5 36.9 1558.5 23.2 86.6 41.5
87 37.2 105.9 73.2 67.9 42.7 838.9 44.8 118.1 84.6
88 7.3 63.5 39.1 21.3 562.5 12.4 50.3 22.5
89 3.3 9.0 12.6 8.2 5.4 115.5 4.6 9.1 6.4
90 19.3 101.3 110.0 35.0 43.2 2453.5 58.2 93.3 49.8
91 1.2 0.9 1.8 2.3 1.6 3.6 1.1 1.1 1.8
92 1.3 3.4 4.0 3.0 3.0 45.8 1.9 2.3 1.7
219
Ex.# SigChem WTIC50 (nM) Del 14 IC50 (nM| D1228H IC50 (nM) D1228N IC50 (nM) F1200I IC50 (nM) L119SV IC50 (nM) Y1230C IC50 (nM) Y1230H IC50 (nM) Y1230S IC50 (nM)
93 23.4 66.3 107.4 35.5 19.6 2636.3 32.9 63.3 35.7
94 10.2 7.2 8.9 17.5 8.4 46.0 9.3 12.1 6.3
95 98.8 163.4 294.2 85.8 94.6 2206.4 89.1 267.9 78.4
96 10.0 16.2 26.1 13.0 202.2 13.8 23.4 13.8
97 4.1 10.1 19.2 5.9 4.7 85.8 3.8 12.4 4.0
98 4.0 4.8 6.8 5.0 2.9 29.6 2.3 5.7 4.1
99 51.0 202.3 370.1 109.0 105.9 3226.1 106.7 234.3 120.0
100 1.1 1.2 1.7 1.7 1.4 9.9 0.7 2.1 1.2
101 18.9 40.7 36.8 31.9 26.4 290.4 15.6 66.1 35.6
102 1.3 7.1 4.0 1.4 2.4 79.8 1.6 4.6 1.2
103 0.8 0.9 1.5 1.8 0.6 11.6 0.6 1.5 0.8
104 1.3 3.6 6.0 3.0 2.0 36.2 1.6 4.0 1.6
105 1.7 2.8 3.9 4.9 2.5 28.3 3.1 4.1 2.3
106 3.5 3.0 4.0 5.8 3.9 20.5 3.3 6.4 3.8
107 1.5 3.7 4.1 4.9 3.7 25.6 2.8 4.5 2.9
108 1.3 2.3 3.5 2.8 2.3 33.0 1.2 4.1 1.8
109 4.0 7.2 18.0 6.6 4.2 114.3 4.4 9.5 4.2
110 2.8 6.5 6.7 3.6 6.0 50.4 2.7 5.7 2.2
113 5.0 20.5 23.0 10.9 10.4 130.6 8.6 17.4 8.2
114 3.6 2.7 3.4 6.7 3.3 7.2 3.2 4.5 1.7
115 3.9 5.0 4.7 6.7 3.9 10.0 4.3 6.8 2.7
116 2.8 6.4 7.7 5.4 2.7 29.2 4.1 6.4 2.4
117 2.4 3.8 6.2 3.1 1.5 46.7 1.3 6.6 1.3
118 4.3 18.5 17.8 8.4 12.1 31.0 6.3 12.4 5.5
119 1.9 1.5 1.6 3.2 2.1 4.3 1.9 2.4 1.2
120 1.7 6.5 5.0 4.8 5.0 41.8 3.4 5.7 2.2
121 1.0 2.2 1.8 2.4 2.2 6.0 1.5 2.1 0.8
122 3.1 8.3 6.4 5.4 10.4 57.3 5.5 10.2 3.6
128 2.6 12.0 12.5 4.6 8.2 79.8 4.5 10.3 4.1
142 5.4 4.7 7.5 6.8 8.9 97.3 6.0 8.3 4.9
144 111.0 408.4 289.5 68.9 163.6 1819.2 117.2 483.0 289.8
145 4.2 9.6 14.7 8.4 6.8 184.3 4.9 12.1 9.1
220
Ex.# SigChem WTIC5O (nM) Dell4 IC50 (nM) D1228H IC5Q (nM) D1228N IC50 (nM) F1200I IC50 (nM) L1195V IC50 (nM) Y1230C IC 50 (nM) Y1230H IC50 (nM) Y1230S IC50 (nM)
146 2.4 5.6 7.9 5.1 5.3 31.5 2.7 6.3 3.6
147 73.3 277.4 256.5 154.4 133,8 2247,3 234.9 279.0 314.3
148 1.2 2.6 3.5 2.1 1.7 22.0 1.7 2.3 1.4
150 2.8 2.6 3.3 5.7 4.5 10.8 2.6 2.7 2.5
151 2.5 3.4 6.4 5.4 3.3 26,5 4,1 5,0 3.2
152 3.2 2.4 2.9 5.1 2.7 9.0 2.9 3.6 3.0
153 4.5 6.6 11.0 8.9 5.3 132.2 6.6 9.4 6.6
154 4.2 3.0 4.3 3.9 3.2 21.5 3.8 5.6 3.2
155 6.8 10.7 20.9 9.5 6.7 104.0 7.0 10,3 5.9
156 4.3 15.0 27.5 9.1 6.7 177.2 6.1 16.7 5.9
157 8.9 26.6 60.0 13.7 6,8 329.6 6.1 26.7 7.1
158 18.0 78.6 136,6 25.0 27.1 671.5 32.1 82.0 39.8
159 19.2 72.7 94.6 25.3 23.4 2007.9 20.1 54.6 20.8
160 1.5 3.3 8.3 3.2 1.8 38.7 2.3 4.4 1.6
161 3.2 3.4 4.1 5.2 2,9 22.4 3.1 4.7 3.2
162 3.1 4.6 6.3 5.4 2.1 43.3 3.4 4.3 3.5
163 2.5 5.9 8.0 3.0 4.0 50.5 4.5 6.2 3.5
164 4,2 6.2 7.3 6.7 4.5 35.2 6.1 6.6 4.3
166 29.8 50.7 78.8 38.8 28.5 329.2 33.6 44.4 18.1
167 33.6 156.4 134.7 55.7 74.1 598.9 86.3 128.4 71,0
168 9.4 52.0 39.8 12.5 20.6 285.2 10.3 32.8 10.4
169 2.8 3.8 3.9 4.0 1.6 27.4 2.1 4.5 1,9
170 8,7 37.9 14.9 10.9 4.8 143.0 5.1 16.6 5.5
171 9.0 40.9 16.5 9.7 17.3 114.9 10.4 18.9 8.9
172 3.6 16.5 16.1 5.7 4.7 42.9 3.8 7.1 2.8
173 27.3 106.2 153.9 44.6 51.2 913.9 70.8 63.8 41.6
179 2.4 3.1 4.5 5.3 4.1 17.8 3.0 4.2 3.4
181 6.7 12.2 14.9 8.1 8.7 110.3 6.5 12.0 10.0
182 65.1 393.2 624.9 60.1 97,9 4109.7 96.8 308.2 150.6
183 13.1 26.4 34,1 14.3 14.8 338.8 10.5 24.2 16.6
184 14.1 37.0 52.8 20.0 16.2 394.0 16.1 37.4 10.9
185 2.0 4.7 5.0 3,2 2.3 2.5 4.4 2.2
221
Ex.# SigChem WT IC5Q (nM) Dell4 l€50 (nM) O1228H IC5Q (nM) D1228N IC50 (nM) F1200I IC50 (nM) L1195V 1C50 (nM) Y1230C IC50 (nM) Y1230H IC50 (nM) Y1230S IC50 (nM)
186 24.0 41.0 139.8 33.2 27.8 1182.7 46.8 74.1 35.1
187 8.5 14.8 46.9 18.4 8.9 259.5 8.6 22.3 13.6
189 14.0 33.4 61.3 11.9 15.5 22.6 27.9 16.4
191 10.3 29.1 63.9 12.5 14.2 505.9 14.4 32.1 17.3
192 5.0 19.2 39.5 10.2 5.0 263.3 9.2 17.4 9.2
193 1.5 2.6 4.9 3.7 2.4 45.0 1.9 3.9 1.7
194 3.8 23.8 44.5 6.0 6.0 191.4 9.4 21.2 9.3
195 1.5 2.7 3.5 3.2 2.5 31.0 1.7 4.1 1.8
196 2.6 10.9 7.0 4.2 5.7 44.4 4.8 7.9 4.8
197 2.7 10.1 7.8 6.1 5.6 54.2 3.1 10.6 4.5
[001067] Synthetic Examples
[001068] Synthesis of Synthetic Intermediates
[001069] Préparation 1
[001070] 3-F luoro-4-((3 -iodo-1 -(4-methoxvbenzv D-1 H-pyrazo io [ 3,4-b1 p vrid in-4-
[001072] Step A: A mixture of l-(4-methoxybenzyl)-lH-pyrazolo[3,4-b]pyridin-4-ol (23,5 g, 92.1 mmol), l,2-difluoro-4-nitrobenzene (14.6 g, 92.1 mmol) and césium carbonate (30.0 g, 92.1 mmol) in DMF (300 mL) was heated to 100 °C for 1 hour. After cooiing to room température, 10 the reaction mixture was poured into water (750 mL) and diluted with EtOAc (750 mL). The organic layer was separated. The aqueous phase was re-extracted with EtOAc (1 x 300 mL, 1 x f 00 mL). The combined organic phases were washed with water (2 x 300 mL) and brine (300 mL), dried over sodium sulfate, filtered and concentrated to afford 4-(2-fluoro-4-nitrophenoxy)-l-(4methoxybenzyl)-lH-pyrazolo[3,4-b]pyridine (36.4 g, 100%).
[001073] Step B: A stirred mixture of 4-(2-fluoro-4-nitrophenoxy)-!-(4-methoxybenzyl)1H-pyrazolo[3,4-b]pyridine (36 g, 91.3 mmol) in TFA (250 mL) was heated to 60 °C for 18 h
222 under N2 with attached reflux condenser. After cooling to room température, the reaction mixture was concentrated in vacuo. Toluene (3 x 75 mL) was utilized to azeotrope residual TFA. The dark mixture was carefully treated with aqueous NaHCCh (150 mL total) with stirring. The biphasic mixture was diluted with DCM (50 mL). A tan suspension resulted, which was stirred for 10 min. The suspension was then filtered. The solid (product) was washed with water (50 mL), then DCM (25 mL). The residual water in the solid was removed by toluene azeotrope by rotary évaporation (3 x 100 mL) at 60 °C to afford 4-(2-fluoro-4-nitrophenoxy)-lH-pyrazolo[3,4-b]pyridine (23 g, 88%).
[001074] Step C: To a stirred mixture of 4-(2-f1uoro-4-nitrophenoxy)-lH-pyrazolo[3,4b]pyridine (22 g, 80 mmol) and KOH (14 g, 241 mmol) (crushed by mortar and pestle) in DMF (250 mL) was added I2 (41 g, 160 mmol). The resulting mixture was heated to 60 °C for 1 h under N2. The reaction mixture was poured into a saturated aqueous solution of sodium thiosulfate (100 mL). The mixture was extracted with EtOAc (I x 250 mL, 2 x 50 mL). The combined organic phases were washed with 10% LiCI (2 x 250 mL), dried over sodium sulfate, filtered, and concentrated to obtain crude 4-(2-fluoro-4-nitrophenoxy)-3-iodo-lH-pyrazolo[3,4-b]pyridine (29 g, 70%).
[001075] Step D: To a stirred mixture of 4-(2-fluoro-4-nitrophenoxy)-3-iodo-lHpyrazolo[3,4-b]pyridine (29 g, 72.5 mmol) and l-(chloromethyl)-4-methoxybenzene (13.6 g, 87.0 mmol) in DMF (250 mL) was added K2CO3 (12.0 g, 87.0 mmol). The reaction mixture was stirred for 18 h and then poured into EtOAc (500 mL) and diluted with water (500 mL). The phases were separated, and the aqueous phase was extracted with EtOAc (2 x 250 mL). The combined organic phases were washed with water (250 mL) and brine (250 mL), dried over sodium sulfate, filtered, and concentrated. The dark brown oil was purified over silica gel (30% EtOAc in hexanes) to afford 4-(2-fluoro-4-nitrophenoxy)-3-iodo-1 -(4-methoxybenzyl)-1 H-pyrazolo[3,4-b]pyridine (24.2 g, 61%).
[001076] Step E: To a stirred mixture of 4-(2'fluoro-4-nitrophenoxy)-3-iodo-I-(4methoxybenzyl)-lH-pyrazolo[3,4-b]pyridine (30 g, 57.7 mmol) in EtOH (500 mL) was added SnCl2-2H2O (59.9 g, 288 mmol). The mixture was heated to 65 °C for Ih under N2. After cooling to room température, the mixture was concentrated. The thick mixture was diluted with DCM (500 mL), stirred until solids dissolved, and then basified with 5N aqueous NaOH (100 mL). The resulting suspension was filtered through Ceiite®, rinsing with DCM (3 x 50 mL). The filtrate was
223 trausferred to a separatory funnel, and the phases were separated. The aqueous phase was reextracted with DCM (50 mL). The combined organic phases were dried overNa2SO4, filtered, and concentrated to afford 3-fluoro-4-(3-iodo-l-(4-methoxybenzyl)-lH-pyrazolo[3,4-b]pyridin-4yloxy)aniline (26.9 g, 90%) as a yellow solid.
[001077] Préparation 2
[001078] 2-(4-fluorophenvl)-3-oxo-2.3-dihvdropvridazine-4-carboxvlic acid ο o <
[001079] F
[001080] Step A: To a 2 L round bottom flask was added a 40% aqueous solution of oxalaldéhyde (218 g, 1504 mmol) and whîle stirring at room température, added a mixture of 1(4-fluorophenyl)hydrazine hydrochloride (48.9 g, 301 mmol), acetic acid (86.1 ml, 1504 mmol), and water (200 mL). The dark reddish brown mixture was stirred at room température for I h. The solid was removed by filtration and washed with water. The solid was placed in a vacuum oven overnight to afford (E)-2-(2-(4-fluorophenyl)hydrazono)acetaldehyde (48 g, 97.5%) as a brick-red solid.
[001081] Step B: Added (E)-2-(2-(4-fluorophenyl)hydrazono)acetaldehyde (48 g, 289 mmol) and 2,2-dimethyl-l,3-dioxane-4,6-dione (43.7 g, 303 mmol) to a 1 L round bottom flask and suspended in toluene (400 mL). Acetic acid (1.65 mL, 28.9 mmol) and piperidine (2.85 mL, 28.9 mmol) were added and mixture was stirred at room température overnight. The resulting precipitate was filtered, washed with hexanes and dried in a vacuum oven to afford (E)-5-(2-(2(4-fluorophenyl)hydrazono)ethylidene)-2,2-dimethyl-l,3-dioxane-4,6'dione (84.8 g, 99%) as a bright red solid.
[001082] Step C: To a 3 L 4-neck round bottom equipped with température probe, mechanical stirrer and 2 reflux condensers added (E)-5-(2-(2-(4fluorophenyl)hydrazono)ethylidene)-2,2-dimethyl-l,3-dioxane-4,6-dione (84.8 g, 290.2 mmol) and dissolved in MeOH (IL). Sodium methoxide (79.62 mL, 348.2 mmol) in MeOH (25% by weight solution) was added and the dark brown solution was stirred and heated to reflux for 1 h. The volume of MeOH was reduced by évaporation and added 500 mL IN HCl. The precipitate was removed by filtration, washed with water and ether, and dried on full vacuum (oven 60-70°C) overnight to afford 2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4’Carboxylie acid (56.4 g,
224
82%) as a yellow solid.
[001083] The following compounds were also made using the procedure according to
Préparation 2.
Préparation Structure Name
3 Il ' ο o 2-(4-fluorophenyl)-6-methyl-3oxo-2,3-dihydropyridazine-4carboxylic acid
[001084] Préparation 4
[001085] 2.5-dioxo-l-phenvl-l1215.617,8-hexahvdroquinoline-3-carboxylic acid
[001086]
[001087]
Step A: To a stirred solution of cyclohexane-l,3-dione (5.0 g, 45 mmol) in DMF (100 mL) at room température under nitrogen was added KOtBu (5.0 g, 45 mmol) followed by ethyl (E)-2-cyano-3-ethoxyacrylate (7.5 g, 45 mmol). The reaction mixture was stirred at room température overnight. The mixture was diluted with EtOAc (400 mL) and stirred while 2N aq. HCl (250 mL) was added. The aqueous layer was extracted with EtOAc (2 x 200 mL) and the 15 combined organic layers were washed with water (4 x 200 mL) and brine (200 mL), dried over sodium sulfate, filtered and concentrated. The residue was purified over silica gel (0-60% EtOAc/hexanes) to afford ethyl 2,5-dioxo-5,6,7,8-tetrahydro-2H-chromene-3-carboxylate (6.2g, 59%) as a dark pink oil.
[001088] Step B: To a stirred solution of ethyl 2,5-dioxo-5,6,7,8-tetrahydro-2H-chromene-320 carboxylate (543 mg, 2.3 mmol) in EtOH (10 mL) was added aniline (210 pL, 2.3 mmol). The mixture was stirred at room température overnight. The résultant solids were filtered, washed with EtOH and dried in vacuo to afford 2,5-dioxo-l-pheny!-l,2,5,6,7,8-hexahydroquinoline-3carboxylic acid (160 mg, 25%) as a white solid.
225
[001089] The following compounds were also made using the procedure according to Préparation 4.
Préparation Structure Name
5 N ο o I -(4-fluorophenyl)-2,5-dioxo- 1,2,5,6,7,8hexahydroqu i no line-3 carboxylic acid
6 T ίΐ Ii ] O O F I -(5-f1uoropyridin-2-yl)-2,5dioxo-1,2,5,6,7,8hexahydroqui nol îne-3 carboxylic acid
[001090] Préparation 7
[001091| 3-(4-fluorophenvl)-l-isopropvl-2,4-dioxo-l,2,3,4-tetrahydropyrimidine-5carboxylic acid H°ATn
O O T
[001092] — A
[001093] Step A: To a solution of diethyl 2-(aminomethylene)malonate (2.5 g, 13.4 mmol) 10 in dichloroethane (10 mL) was added 1-fluoro-4-isocyanatobenzene (1.59 mL, 14.0 mmol) followed by DIEA (2.57 mL, 14.7 mmol). The reaction mixture was stirred at 100 °C for 6 h and then cooled to room température overnight. The résultant solids were filtered, washed with Et:O and dried in vacuo to afford diethyl 2-((3-(4-fluorophenyl)ureido)methylene)malonate (3.38g, 78%) as a white solid.
[001094] Step B: To a suspension of diethyl 2-((3-(4fluorophenyl)ureido)methylene)malonate (3.38 g, 10.4 mmol) in éthanol (15 mL) was added sodium ethoxide (6.23 mL, 21%, 16.7 mmol) dropwise via syringe. The mixture was stirred for 2 h, then concentrated and partitioned between EtOAc (150 mL) and IM Citric acid (100 mL). The
226 aqueous layer was extracted with EtOAc (2 x 100 mL) and the combined organic phases were washed with brine (50 mL), dried over Na2SÛ4, filtered and concentrated to afiford ethyl 3-(4fluorophenyl)-2,4-dioxo-l,2,3,4-tetrahydropyrimidine-5-carboxylate (2.83 g, 98%) as a pale yellow solid.
[001095] Step C: To a suspension of ethyl 3-(4-fluorophenyl)-2,4-dioxo-l,2,3,4tetrahydropyrimidine-5-carboxylate (1-0 g, 3.6 mmol) and K2CO3 (993 mg, 7.2 mmol) in DMF (5 mL) was added 2-iodopropane (719 pL, 7.2 mmol). The mixture was heated in a sealed tube at 70 °C overnight. The cooled mixture was partitioned between water (50 mL) and EtOAc (50 mL) and the aqueous layer was extracted with EtOAc (2 x 30 mL). The combined organic phases were 10 washed with water (5 x 20 mL) and brine (20 mL) then dried over Na2SÛ4, filtered and concentrated to afford ethyl 3-(4-fluorophenyl)-l-isopropy 1-2,4-dioxo-l ,2,3,4tetrahydropyrimidine-5-carboxylate (1.14g, 99%) a pale yellow foam.
[001096] Step D: To a solution of ethyl 3-(4-fluorophenyl)-l-isopropy 1-2,4-dioxo-l,2,3,4tetrahydropyrimidine-5-carboxylate (1.14 g, 3.56 mmol) in 4N HCI / dioxanes (10 mL) was added 15 water (2 mL). The mixture was stirred at 70 °C overnight. The cooled mixture was treated with water (20 mL) and the resulting solids filtered, washed with water and dried in vacuo to afford 3(4-fluorophenyl)-I-isopropyl-2,4-dioxo-l,2,3,4-tetrahydropyrimidine-5-carboxylic acid as a fluffy white solid.
[001097] The following compounds were also made using the procedure according to 2 0 Préparation 7.
Préparation Structure Name
8 LL 0 —Z }=o 0 X 3-(4-fluorophenyl)-1 -methyl- 2,4-dioxo-1,2,3,4tetrahydropyrimidine-5carboxylic acid
9 X 0 ο=ζ O0 \z— 70 Π l-ethyl-3-(4-fluoropheny 1)-2,4dioxo-1,2,3,4tetrahydropyrimidine-5carboxylic acid
227
10 Η0Λνύ^ ο ο 0^^00. F 1 -(cyclopropylmethyl)-3-(4fIuorophenyl)-2,4-dîoxo- 1,2,3,4-tetrahydropyrimidine-5carboxylic acid
U OH ΗοΛΝη Ο Ο 3-(4-fluorophenyl)-l-(2hydroxyethyl)-2,4-dioxol,2,3,4-tetrahydropyriniidine-5carboxylic acid
12 zr Ο ο=/ Ο=( Ζ—/ ν_ 0 Π 3-(4-fluoropheny l)-2,4-d ioxo1 -(pentan-3-yl)-l,2,3,4tetrahydropy ri m i d i ne-5 carboxylic acid
13 0 ο Γ l-cyclobutyl-3-(4fluorophenyl)-2,4-dioxol,2,3,4-tetrahydropynmidine-5carboxylic acid
14 Η0γντν 3-(3,4-difluorophenyi)-1 isopropyl-2,4-dioxo-1,2,3,4ΐetrahydropyrinlidίne-5carboxylic acid
15 ΗοΛ“η ο ο 0^0 3-cyclohexyl-l -isopropyl-2,4dioxo-1,2,3,4tetrahydropyrimidine-5carboxylic acid
228
16 ηοΛ^ 0 0 L-/ 3-cyclopentyl-l -isopropyl-2,4dioxo-1,2,3,4tetrahydropy rimid i ne-5 carboxylic acid
17 . 0 ) Q 2=0 ο X 1 -isopropy 1-3-( I -methyl-1Hpyrazol-4-yl)-2,4-dioxo- 1,2,3,4-tetrahydropy ri midi ne-5 carboxylic acid
18 ο \λ\=>° 2=0 ο τ l,3-diisopropyl-2,4-dioxo1,2,3,4-tetrahydropyrimidine-5carboxylic acid
19 ηοΛνυί 0 0 k^O ]-isopropyl-2,4-dioxo-3(tetrahydro-2H-pyran-4-yl)1,2,3,4-tetrahydropy ri midi ne-5 carboxylic acid
20 HoANri 0 0 k^k ' Cl 3-(4-chlorophenyl)-lisopropyl-2,4-dioxo-l,2,3,4tetrahyd ropy ri m id ine-5 carboxylic acid
21 LL· LL· .0 2=Q 2=o O X 3-(3,4-difliioropheny[)-1 -ethyl2,4-dioxo-l,2,3,4tetrahyd ropy ri m i d ine-5 carboxylic acid
229
[001098] Préparation 22
[001099] 3-(4-fluorophenyl)-l-(l-methvlazetidin-3-vl)-2.4-dioxo-i.2,3,4tetrahydropvrimidine-5-carboxylic acid hydrochloride
[001100]
[001101] Step A: To a suspension of ethyl 3-(4-fluorophenyl)-2,4-dioxo-l,2,3,4tetrahydropyrimidine-5-carboxylate (1.0 g, 3.6 mmol) and K2CO3 (993 mg, 7.2 mmol) in DMF (5 mL) was added tert-butyl 3-îodoazetidine-l-carboxylate (2.04 g, 7.2 mmol). The mixture was heated in a sealed tube at 70 °C overnight. The cooled mixture was partitioned between water (50 mL) and EtOAc (50 mL) and the aqueous layer was extracted with EtOAc (2 x 30 mL). The combined organic phases were washed with water (5 x 20 mL) and brine (20 mL) then dried over Na2SO4, filtered and concentrated. The residue was purtfied over silica gel (0-4% MeOH in DCM) to afford ethyl 1 -(l-(tert-butoxycarbonyl)azetidin-3-yl)-3-(4-fluorophenyl)-2,4-dioxo-l,2,3,4tetrahydropyrimidine-5-carboxylate (0.579 g, 37%).
[001102] Step B: To a solution of ethyl l-(l-(tert-butoxycarbonyl)azetidin-3-yl)-3-(4fluorophenyl)-2,4-dioxo-l,2,3,4-tetrahydropyrimidine-5-carboxylaie (400 mg, 0.92 mmol) in DCM (8 mL) was added TFA (2 mL). The mixture was stirred for 2 h and concentrated to afford crude ethyl l-(azetidin-3-yl)-3-(4-fluorophenyl)-2,4-dioxo-l,2,3,4-tetrahydropyrimidine-5carboxylate 2,2,2-trifluoroacetate (413 mg, 100%).
[001103] Step C: To a suspension of ethyl l-(azetidin-3-yl)-3-(4-fluorophenyl)-2,4-dioxol,2,3,4-tetrahydropyrimidine-5-carboxylate 2,2,2-trifluoroacetate (200 mg, 0.45 mmol) in dichloroethane (5 mL) was added formaldéhyde (37% in water, 181 mg, 2.24 mmol) followed by NaBH(OAc)3 (237 mg, 1.12 mmol). The mixture was stirred vigorously for 4 h. The mixture was treated with 2N NasCCL (10 mL), stirred for 30 min, then extracted with DCM (3 x 10 mL). The combined organic phases were washed with brine (10 mL), dried over Na2SO4, filtered and concentrated to afford crude ethyl 3-(4-fluorophenyl)-l-(l-methylazetidin-3-yl)-2,4-dioxol,2,3,4-tetrahydropyrimidine-5-carboxylate (127 mg, 82%) as a white solid.
230
[001104] Step D: To a solution of ethyl 3-(4-fluorophenyl)-l-(l-methylazetidin-3-yl)-2,4dioxo-l,2,3,4-tetrahydropyrimidine-5-carboxylate (174 mg, 0.50 mmol) in 4N HCl / dioxanes (5 mL) was added water (I mL). The mixture was stirred at 70 °C overnight. The cooled mixture was treated with water (10 mL) and extracted with EtOAc (10 mL). The aqueous layer was concentrated to afford crude 3-(4-fliiorophenyl)-l-(l-methylazetÎdin-3-y[)-2,4-dioxo-l,2,3,4tetrahydropyrimidine-5-carboxylic acid hydrochloride sait (116 mg, 65%).
[001105] Préparation 23
[001106] l-cyclopropyl-3-(4-fluorophenyl)-2,4-dioxo-l,2,3,4-tetrahydropyrimidine-5-
[001108] Step A: To a suspension of ethyl 3-(4-fluorophenyl)-2,4-dioxo-l,2,3,4tetrahydropyrimidine-5-carboxylate (900 mg, 3.23 mmol), cyclopropylboronic acid (834 mg, 9.70 mmol) and Na2CO3 (1028 mg, 9.70 mmol) in dichloroethane (6mL) was added a suspension of diacetoxycopper (176 mg, 0.970 mmol) and 2,2'-bipyridine (505 mg, 3.23 mmol) in hot dichloroethane (6mL). The mixture was heated to 70 °C for 2 h. Additional Na2CO3 (1028 mg, 9.70 mmol), cyclopropylboronic acid (834 mg, 9.70 mmol), diacetoxycopper (176 mg, 0.970 mmol) and 2,2'-bipyridine (505 mg, 3.23 mmol) were added and the reaction mixture was stirred at 70 °C overnight. The reaction mixture was cooled to room température and diluted with saturated NH4CI solution (20mL). The aqueous layer was extracted with DCM (2 x 20 mL), dried over sodium sulfate, filtered and concentrated. The residue was purified over silica gel (0-100% EtOAc in hexanes) to afford ethyl l-cyclopropyl-3-(4-fluoropheny[)-2,4-dioxo-l,2,3,4tetrahydropyrimidine-5-carboxylate (180 mg, 14%).
[001109] Step B: To ethyl l-cyclopropyl-3-(4-fluorophenyl)-2,4-dioxo-l,2,3,4tetrahydropyrimidine-5-carboxylate (180 mg, 0.565 mmol) was added hydrogen chloride (1414 μΐ, 5.65 mmol, 4M in dioxane) and water (0.3 mL). The mixture was heated to 70 °C for 3 h, The reaction mixture was cooled to room température and diluted with EtOAc (20 mL) and water (10 mL). The aqueous layer was extracted with EtOAc (2 x 10 mL). The combined organic was dried over MgSO4, filtered and concentrated afford l-cyclopropyl-3-(4-fluorophenyl)-2,4-dioxo
231 l,2,3f4-tetrahydropyrimidine-5-carboxylic acid (180 mg, 110%).
[001110] Préparation 24
[001111] 4-ethoxv-l-(4-fluorophenvl)-2-oxo-L2-dihvdropvridine-3-carboxvlic acid ΗοΧλπ O O <
[001112] F
[001113] Step A: Ethyl 4-ethoxy-2-oxo-l,2-dihydropyridine-3-carboxylate (1.0 g, 4.73 mmol), quinolin-8-ol (275 mg, 1.89 mmol) and césium carbonate (3.09 g, 9.47 mmol) were combined in DMF (10 mL) and purged with Ar for 5 min. Cu(I)iodide (271 mg, 1.42 mmol) and l-fluoro-4-iodobenzene (819 pL, 7.1 mmol) were added and the mixture stirred in a sealed vessel at 100 °C overnight. The mixture was partitioned between water (100 mL) and EtOAc (100 mL) and the aqueous layer was extracted with EtOAc (2 x 50 mL). The combined organic phases were washed with water (30 mL), 2N HCl (30 mL), water (2 x 30 mL) and brine (30 mL), dried over NajSOi, filtered and concentrated to afford ethyl 4-ethoxy-l-(4-fluorophenyl)-2-oxo-l,2dihydropyridine-3-carboxylate (1.38 g, 95%) as a yellow solid.
[001114] Step B : To a solution of ethyl 4-ethoxy-l-(4-fluorophenyl)-2-oxo-l,2dihydropyridine-3-carboxylate (1.38 g, 4.52 mmol) in EtOH (40 mL) was added 2N HCl (9 mL, 18.1 mmol). The mixture was stirred at 65 °C overnight. After cooling, the solids were filtered, washed with EtsO and dried in vacuo to afford 4-ethoxy-l-(4-fluorophenyl)-2-oxo-l,2dihydropyridine-3-carboxylic acid (0.418 g, 33%) as a white solid.
[001115] The following compound was also made using the procedure according to Préparation 24.
Préparation Structure Name
25 HoïYn ο o 1 ~(4-fluorophenyl)-2oxopiperidine-3-carboxylic acid
[001116] Préparation 26
[001117] 2-(4-fluorophenyl)-l-methvl-3-oxo-2,3-dihvdro-1 H-pyrazole-4-carboxyIic acid
232
J O HO'A-A /=\ h nV Vf N X— [001118] \
[001119] Step A: (4-Fluorophenyl)hydrazine hydrochloride (3.13 g, 19.2 mmol) was suspended in EtOH (40 mL) and K2CO3 (5.32 g, 38.5 mmol) was added, followed by the addition of diethyl 2-(ethoxymethylene)malonate (3.85 ml, 19.2 mmol) and the reaction mixture was heated to 80 °C overnight. The cooled reaction was concentrated, diluted with water (100 mL) and acidified with 3N aq. HCl (50 mL) to bring the pH to 3. The aqueous layer was extracted with EtOAc (2 x 100 mL). The combined organic layers were dried overNa2SO4 and concentrated. The residue was triturated with MeOH (2x10 mL) and the solid was dried to afford ethyl 2-(4fluorophenyl)-3-oxo-2,3-dihydro-lH-pyrazole-4-carboxylate (2.88g, 60%).
[001120] Step B: To 20 mL vial, ethyl 2-(4-fluorophenyl)-3-oxo-2,3-dihydro-lH-pyrazole4-carboxylate (1.56 g, 6.23 mmol) and methyl trifluoromethanesulfonate (2.11 ml, 18.7 mmol) was added and heated to 80 °C for 1 h. The reaction mixture was diluted with DCM (50 mL) and washed with saturated NaHCO3 (30 mL). The aqueous layer was extracted with DCM (2 x 20 mL). The combined organic layers were dried overNaaSCL and concentrated under vacuum. The residue was purified over silica gel (70-100% EtOAc in hexanes) to afford (4-(ethoxycarbonyl)-2-(4fluorophenyl)-1-methyl-1,2-dihydro-3H-pyrazol-3-yhdene)(methyl)oxonium trifluoromethanesulfonate (2.143 g, 100%).
[001121] Step C: To a 40 mL vial (4-(ethoxycarbonyl)-2-(4-f]uorophenyl)-l-methyl-1,2dihydro-3H-pyrazol-3-ylidene)(methyl)oxonium (1.74 g, 6.23 mmol) added and diluted with THF (6 mL) and MeOH (5 mL). 4N aq. NaOH (9.35 ml, 37.4 mmol) was added and the reaction mixture was heated to 60 °C. After 1 h, the reaction mixture was cooled to room température. The reaction mixture was concentrated to remove volatile solvents. To the solution was 3N aq. HCl added to bring the pH to 1. The solution was extracted with EtOAc (3x10 mL). The combined organic layers were concentrated under reduced pressure to afford 2-(4-fluorophenyl)-l-methyl-3-oxo-2,3dihydro-lH-pyrazole-4-carboxylic acid (1.26 g, 85.8%).
[001122] The following compound was also made using the procedure according to
Préparation 26.
233
Préparation Structure Name
27 9 ° ΗΟ^ν-Λ /=\ L 'N “A ? 'N \ 1 -methyl-3-oxo-2-pheny 1-2,3dihydro-lH-pyrazole-4carboxylic acid
[001123] Préparation 28
[001124] l-(4-fluorophenvl)-6-methyl-2-oxo-l,2-dihydropvridine-3-carboxvlic acid
[001125]
[001126] Step A: A solution of 4-fluoroaniline (1.72 ml, 18.0 mmol) and triethy lamine (3.01 ml, 21.6 mmol) in DCM (90 mL) was prepared and purged with an argon balloon. The solution was cooled to 0 °C in an ice bath. Ethyl 3-chloro-3-oxopropanoate (2.719 mL, 21.60 mmol) was dissolved in DCM (8 mL) and this solution was added dropwise to the reaction over the course of fïve minutes. The reaction mixture was allowed to slowly warm to room température overnight.
The reaction mixture was diluted with water (100 mL) and separated, washed with NaHCOj (50 mL x 4) and brine (50 mL), dried over sodium sulfate, filtered and concentrated to afford crude ethyl 3-((4-fluorophenyl)amino)-3-oxopropanoate (4.32 g, 106%).
[001127] Step B: Ethyl 3-((4-fluoro pheny l)amino)-3-oxopropanoate (2 g, 8.88 mmol) was dissolved in éthanol (22 mL). This solution was stirred while (E)-4~methoxybut-3-en-2-one (1.36 mL, 13.3 mmol) was added. After stirring together for 10 min, sodium ethanolate (4.97 mL, 13.3 mmol) was added and heated to reflux for 2 h. The reaction mixture was concentrated and diluted with DCM (200 mL) and washed with IN HCl (50 mL x 2) and brine (50 mL), dried over sodium sulfate, filtered and concentrated. The residue was purified over silica gel (5% MeOH in DCM) to afford 1 -(4-fluorophenyl)-6-methyl-2-oxo-l ,2-dihydropyridine-3-carboxylic acid (2.2 g, 100%).
234
[001128] Préparation 29
[001129] l-(4-fluoroDhenvl)-4-methyl-2-oxo-I,2-dihvdropvridine-3-carboxvlÎc acid
[001130]
[001131] Step A: A solution of 4-fluoroanîline (1.73 mL, 18.0 mmol) and IH-imidazole (0.249 g, 3.67 mmol) in diethyl 2-(propan-2-ylidene)malonate (9.80 mL, 50 mmol) were combined in a sealed tube and heated to 200 °C underargon for 3 h. The reaction mixture was cooted to room température and purîfîed over silica gel (25% EtOAc in hexanes) to afford ethyl 2-((4fluorophenyl)carbamoyl)-3-methylbut-2-enoate (2.13 g, 31%).
[001132] Step B: Ethyl 2-((4-fluorophenyl)carbamoyl)-3-methylbut-2-enoate (2.13 g, 8.03 mmol) was dissolved in l,l-dimethoxy-N,N-dimethylmethanamine (0.957 g, 8.03 mmol) and heated to 90 °C for 90 minutes in a sealed tube. The reaction mixture was cooled to room température and diiuted with 30 mL EtOAc and 30 mL saturated NH4CI and stirred for 15 minutes. The phases were separated and the organic phase was washed with NH4CI (2x30 mL), dried over Na^SOi, fütered and concentrated. The residue was purified over silica gel (5% MeOH in DCM) to afford ethyl l-(4-fluorophenyl)-4-methyl-2-oxo-l,2-dihydropyridine-3-carboxylate (320 mg, 15%).
[001133] Step C: Ethyl l-(4-fluorophenyl)-4-methyl-2-oxo-l,2-dihydropyridine-3carboxylate (290 mg, 1.05 mmol) was dissolved in MeOH (3 mL) and water (2 mL). LiOH (88 mg, 2.1 mmol) was added and the reaction mixture was heated to 37 °C overnight. The reaction mixture was diiuted with water (10 mL) and the methanol removed under reduced pressure. The aqueous layer was extracted with EtzO (2 x 50 mL) and the pH was decreased to 1 with IM HCl. The résultant solids were fütered, washed with water and dried to afford l-(4-fluorophenyl)-4methyl-2-oxo-l,2-dihydropyridine-3-carboxylic acid (121 mg, 46%).
[001134] Préparation 30
[001135] l-(4-fluorophenvl)-5-methyl-2-oxo-1.2-dihvdropvridine-3-carboxylic acid
[001136]
235
[001137] Ethyl 3-((4-fluorophenyl)amino)-3-oxopropanoate (18 g, 79.9 mmol) was dissolved in éthanol (200 mL) and (E)-3-ethoxy-2-methylacrylaldehyde (13.7, 119.8 mmol) was added. After 5 min, sodium ethanolate (44.7 ml, 119.8 mmol, 21 wt% in EtOH) was added and the reaction mixture was heated at reflux for 3 h. The reaction mixture was cooled to room 5 température and added to a stirred aqueous solution of2N HCl (2 L) and the mixture was stirred overnîght at room température. The solids were filtered and washed with 2N HCl (2 x 500 mL) and water (500 mL). The filter cake and filter paper were transferred to a large Buchi vacuum flask and dried overnight to afford I-(4-fluorophenyl)-5-methyl-2-oxo-l,2-dihydropyridine-3carboxyiic acid (15.9 g, 80%).
[001138] The following compounds were also made using the procedure according to
Préparation 30.
Préparation Structure Name
31 HWnF O O X F 1 -(3,4-difluorophenyl)-5methyl-2-oxo-l ,2d ihydropyrid ine-3-carboxy lie acid
32 LL p r- Z O X l-(3-fluorophenyl)-5-methyl-2oxo-l,2-dihydropyridine-3carboxylic acid
[001139]
[001140]
Préparation 33
5-cyclopropvl-l-i4-fiuorophenvl)-2-oxo-l.2-dîhvdropyndine-3-carboxvlic acid
[001141]
[001142]
Step A: A 250 tnL round bottomed flask was charged with methyl 5-bromo-2-oxol,2-dihydropyrîdine-3-carboxylate (2.84 g, 12.2 mmol), (4-fluorophenyl)boronic acid (4.62 g, 33.0 mmol), diacetoxycopper (4.34 g, 23.9 mmol) and DCM (50 mL). Activated molecular sieves (1g) were added followed by pyridine (3.86 ml, 47.7 mmol). The reaction mixture was stirred for
236 days at room température under normal atmosphère. The reaction mixture was diluted to 200 mL with DCM and filtered through Celite®. The filtrate was washed with water (3 x 250 mL) and brine, dried over sodium sulfate, filtered and concentrated. The residue was purified over silica gel (25% EtOAc in DCM) to afford methyl 5-bromo-1-(4-fluorophenyl)-2-oxo-l,2-dihydropyridine3-carboxylate (3.53 g, 10.8 mmol, 88.4 % yield).
[001143] Step B: Methyl 5-bromo- i-(4-fluorophenyl)-2-oxo-l,2-dihydropyridine-3carboxylate (0.400 g, 1.23 mmol) was dissolved în 10:1 toluene:water (16 mL) and nitrogen was bubbled through this solution for 5 min. Potassium cyclopropyltrifluoro borate (0.726 g, 4.91 mmol), diacetoxypalladium (0.038 g, 0.169 mmol), and K3PO4 (0.260 g, 1.23 mmol) were added with nitrogen bubbling through the reaction mixture for l min. Dicyclohexyl(2,,6'-diisopropoxy[l,r-biphenyl]-2-yl)phosphane (0.172 g, 0.368 mmol) was added and the reaction mixture was bubbled with nitrogen for 1 min, then sealed and heated to 105 °C overnight. The reaction mixture was partit!oned between water and EtOAc, washed with water and brine, dried over sodium sulfate, filtered and concentrated. The residue was purified over silica gel (80:20 DCM: EtOAc) to afford methyl 5-cyclopropyl-l-(4-fluorophenyl)-2-oxo-l,2-dihydropyridine-3-carboxylate (0.240 g, 0.835 mmol, 68.1 % yield).
[0011441 Step C: Methyl 5-cyclopropyl-l-(4-fiuorophenyl)-2-oxo-l,2-dihydropyridine-3carboxylate (240 mg, 0.835 mmol) was dissolved in THF (20 mL) and MeOH (5 mL). To this was added a solution of lithium hydroxide hydrate (550 mg, 13.1 mmol) in a minimal amount of water (1 mL). After 2.5 h, the reaction mixture was concentrated, diluted with water (5 mL) and pH adjusted to 1 with concentrated HCl. The resulting solid was filtered, washed with 2N HCl and dried to afford 5-cyclopropyl-l-(4-fluorophenyl)-2-oxo-l,2-dihydropyridine-3-carboxylic acid (160 mg, 0.586 mmol, 70.1 % yield).
[001145] Préparation 34
[001146] 5-bromo-l-(4-fluorophenvl)-2-oxo-L2-dihydropyridine-3-carboxylie acid
Br
[001147]
[001148] Step A: A round bottomed flask was charged with methyl 5-bromo-2-oxo-l,2dihydropyndine-3-carboxylate (1 g, 4.31 mmol), (4-fluorophenyl)boronic acid (1.63 g, 11.6
237 mmol), diacetoxycopper (1.53 g, 8.40 mmol) and DCM (50 mL). Activated molecular sieves (1 g) were added followed by pyridine (1.36 ml, 16.8 mmol). The reaction mixture was stirred overnight at room température under air. The b lue slurry was filtered through Celite® and the washed with DCM and concentrated. The concentrate was partitioned between DCM and water, dried over sodium sulfate, filtered and concentrated. The residue was purified over silica gel (25% EtOAc in DCM) to afford methyl 5-bromo-l-(4-fluorophenyl)-2-oxo-l,2-dihydropyridine-3carboxylate (0.84 g, 60% yield).
[001149] Step B: Methyl 5-bromo-l-(4-fiuorophenyl)-2-oxo-l,2-dihydropyridine-3carboxylate (382.3 mg, i .172 mmol) was dissolved in THF (20 mL) and MeOH (5 mL). To this was added a solution of lithium hydroxide hydrate (491.9 mg, 11.72 mmol) in a minimal amount of water -1 mL. After 2.5 h, the reaction mixture was concentrated, diluted with water (5mL) and pH adjusted to 1 with concentrated HCl. The resulting solid was filtered, washed with 2N HCl and dried to afford 5-bromo-l-(4-fluorophenyl)-2-oxo-l,2-dihydropyridine-3-carboxylic acid (287.6 mg, 0.9215 mmol, 78.61 % yield).
[001150] The following compound was also made using the procedure according to Préparation 34.
Préparation Structure Name
35 Cl Ο Ο 5-chloro-l-(4-fluorophenyl)-2oxo-l,2-dihydropyndine-3carboxylic acid
[001151] Préparation 36
[001152] 4-(4-fluorophenyl)-3-oxo-3,4-dihydropyrazîne-2-carboxylic acid
[001154] Step A: To a stirred mixture of 4-fluorobenzenamine (41 g, 369.0 mmol) in acetic acid (211.2 ml, 3690 mmol) and KCN (31.23 g, 479.7 mmol) in water (50 mL) at 0 °C was added formaldéhyde (41.2 ml, 553.5 mmol, 37% in water) dropwise. The dark solution was stirred at 0
238 °C for 15 minutes and allowed to warm to room température overnight. The reaction mixture was diluted with water (500 mL), extracted with EtOAc, washed with IN NaOH, water, and brine, dried over MgSCU, fiItered and concentrated to afford crude 2-(4-fluoropheny[amino)acetonitrile (52 g, 93%).
[001155] Step B: To a stirred mixture of 2-(4-fluorophenylamino)acetonitrile (52 g, 346 mmol) în 1,2-dîchlorobenzene (139 ml, 346 mmol) was added slowly oxalyl dichloride (132 g, 1039 mmol). The reaction mixture was heated to 100 °C and stirred for 5.5 h. The dichlorobenzene was removed by vacuum distillation (10 mm Hg at 75°C) and the crude material was purified over silica gel (0-1% acetonitrile in DCM) to afford 3,5-dichloro-l-(4-fluorophenyl)pyrazin-2(I H)-one (18g, 20%).
[001156] Step C: 3,5-Dichioro-1-(4-fluorophenyl)pyrazin-2(lH)-one (18 g, 69.5 mmol) was suspended in MeOH (280 mL) and slowly treated with sodium methoxide (23.8 ml, 104 mmol) at 0 °C. The reaction mixture was allowed to warm to room température and stirred for 5 h. The reaction mixture was neutralized with HCl (17.4 mL, 34.7 mmol, 2N EtsO solution), and concentrated. The residue was taken up in EtOAc (800 mL), washed with aqueous 0.5 N HCl (600 mL), dried over MgSOi, filtered and concentrated to afford 5-chloro-l-(4-fluorophenyl)-3methoxypyrazin-2(lH)-one (16.1 g, 91%).
[001157] Step D: 5-Chloro-l-(4-fluorophenyl)-3-methoxypyrazin-2(lH)-one (43 g, 169 mmol) was suspended in MeOH (2 L) and heated to 50°C to dissolve ail ofthe solids. The réaction mixture was cooled to room température and K2CO3 (23.3 g, 169 mmol) and 10% (dry basis: about 50% water) Pd/C (18,0 g, 8.44 mmol) were added tothe mixture at room température. The mixture was stirred under a hydrogen atmosphère. After 4 h, the mixture was fîltered and concentrated. The residue was taken up in DCM (1500 mL) and washed with water (1000 mL) and brine, dried over MgSÛ4, fîltered and concentrated to afford l-(4-fluorophenyl)-3-methoxypyrazin-2(lH)-one (22.7 g, 61%).
[001158] Step E: l-(4-Fluorophenyl)-3-methoxypyrazin-2(lH)-one (22.0 g, 100 mmol) was suspended in DMF (200 mL). The mixture was cooled to 0 °C and POCls (22.9 ml, 250 mmol) was added dropwise. After addition, the mixture was heated to 90 °C for 2 h and then cooled to 0 °C, The reaction mixture was quenched by adding saturated sodium acetate solution (40 mL) and cooling to 0°C. After 20 min, a fine precipitate formed. The solid was removed by filtration and washed with a small portion (50 mL) of saturated sodium acetate solution followed by îce water
239 ( 100 mL). The solid was dried overnight to afford 3-chloro- ! -(4-fluorophenyl)pyrazin-2( 1 H)-one (14.7 g).
[001159] Step F: 3-Chloro-l-(4-fluorophenyl)pyrazin-2(lH)-one (17.9 g, 79.9 mmol), zinc cyanide (5.63 g, 47.9 mmol), dppf (4.43 g, 7.99 mmol), Pd2dba2 (3.65 g, 3.99 mmol) were suspended in NMP (360 mL). The reaction mixture was stirred under nitrogen at 120 °C overnight. The reaction mixture was cooled, diluted with EtOAc (3000 mL), washed with a 4:1:4 mixture of saturated NLUChconc. NH4OH: water and brine, dried 0verMgSO4, filtered and concentrated. The residue was purified over silica gel (0-1% MeOH in DCM) to afford 4-(4-fluorophenyl)-3-oxo3,4-dihydropyrazine-2-carbonitrile (10.76 g, 62%).
[001160] Step G: 4-(4-Fluorophenyl)-3-oxo-3,4-dihydropyrazine-2-carbonitrile (10.50 g, 48.80 mmol) was dissolved in concentrated H2SO4 (104.0 ml, 1952 mmol) and stirred for 2 h. The reaction mixture was slowly poured into methanol (700 mL) at 0 °C. After complété addition, the mixture was heated to 70 °C and stirred for 2.5 h. The reaction mixture was cooled to room température and poured into 1500 mL of ice. The pH was adjusted to 12 with 5N NaOH. The pH was then adjusted to 2 with 2N HCl, extracted with EtOAc, dried over MgSOi, filtered and concentrated. The residue was triturated with ether and filtered to afford 4-(4-fluorophenyl)-3-oxo3,4-dîhydropyrazine-2-carboxylic acid (9.2 g, 78%).
[001161] Préparation 37
[001162] 6-cvclopropyl-2-(4-fluorophenyl)-3-oxo-2,3-dihvdropvridazine-4-carboxylic acid
[001164J Step A: To 2-cyclopropyl-2-oxoacetaldehyde (0.424 mL, 5.10 mmol) was added a mixture of 4-fluorophenylhydrazine hydrochloride (0.829 g, 5.10 mmol) in AcOH (4.25 mL, 5.10 mmol) and water (4.25 mL, 5.10 mmol) at room température while stirring for 20 minutes. The mixture was added to cold water (200 mL) and stirred for 10 minutes. The mixture was filtered and the isolated solids were washed with water (50 mL). The residue was purified over silica gel (1-10% EtOAc in DCM) to obtain (E)-2-cyclopropyl-2-(2-(4fluoropheny])hydrazono)acetaldehyde (0.080 g, 0.388 mmol, 7.61 % yield) the minor isomer.
240
[001165] Step B: To a solution of (E)-2-cycIopropyl-2-(2-(4fluorophenyl)hydrazono)acetaldehyde (0.080 g, 0.388 mmol) in toluene (3.88 mL) was added 2,2dimethyl-l,3-dioxane-4,6-dione (0.0671 g, 0.466 mmol), AcOH (0.00222 mL, 0.0388 mmol) and piperidine (0.00383 ml, 0.0388 mmol). After 1 h, the mixture was added to water (50 mL) and the organics were extracted with EtOAc (25 mL). The organic layer was washed with water (25 mL) and brine (25 mL), dried with Na2SO4, filtered and concentrated. The residue was purified over silica gel (1-10% EtOAc in DCM) to obtain 6-cyclopropy 1-2-(4-fluorophenyl)-3-oxo-2,3dihydropyridazine-4-carboxylic acid (0.025 g, 0.0912 mmol, 23.5 % yield) as a tan solid.
[001166] The following compound was also made using the procedure according to Préparation 37.
Préparation Structure Name
38 Λ ο Ο ί xÀ. 2-(4-fiuorophenyl)-6isopropyl-3-oxo-2,3dihydropyridazine-4carboxylic acid
[001167] Préparation 39
[001168] 4-(4-fluorophenvl)-2-isopropvl-5-oxo-3-thioxo-2.3A5-tetrahvdro-L2,4-triazine6-carboxvlic acid
[001169]
[001170]
Step A: A mixture of diethyl 2-oxomalonate (4.12 mL, 26.99 mmol) and N-(4fluorophenyl)hydrazinecarbothioamide (5.0 g, 26.99 mmol) in éthanol (100 mL) was heated at reflux for 2 days. The mixture was cooled to room température and filtered. The solids were washed with cold EtOH and dried to afford ethyl 4-(4-fluorophenyl)-5-oxo-3-thioxo-2,3,4,5tetrahydro-l,2,4-triazine-6-carboxylate (3.85 g, 48%) as a yeliow powder.
[001171] Step B: To a suspension of ethyl 4-(4-fluorophenyl)-5-oxo-3-thioxo-2,3,4,5tetrahydro-l,2,4-triazine-6-carboxylate (400 mg, 1.433 mmol) and K2CCh (396 mg, 2.865 mmol) in DMF (5 mL) was added 2-iodopropane (287 pL, 2.86 mmol). The mixture was heated in a
241 sealed tube at 65 °C for 1 h. The mixture was cooled to room température and partitioned between water (20 mL) and EtOAc (20 mL), The aqueous layer was extracted with EtOAc (2x15 mL), The combined organic phases were washed with water (5x15 mL) and brine (20 mL), dried over Na?SO4, filtered and concentrated. The residue was purified over silica gel (0-20% EtOAc in hexanes) to afford ethyl 4-(4-fluorophenyl)-2-isopropyl-5-oxo-3-thioxo-2,3,4,5-tetrahydro-l,2,4triazine-6-carboxylate (211 mg, 46% yield).
[001172] Step C: Sulfuric acid (5 mL) was carefully added to a mixture of ethyl 4-(4fluorophenyl)-2-isopropyl-5-oxo-3-tbioxo-2,3,4,5-tetrahydro- l,2,4-triazine-6-carboxylate (211.1 mg, 0.657 mmol) and water (1 mL). The mixture became homogenous after a few minutes. The reaction mixture was stirred at 40 °C overnight, cooled to room température, and then carefully added to ice. The mixture was saturated with solid NaCl and was extracted with EtOAc (3x40 mL) The combined EtOAc layers were dried over sodium sulfate, filtered and concentrated to afford 4(4-fluorophenyl)-2-isopropyl-5-oxo-3-thioxo-2,3,4,5-tetrahydro-l,2,4-triazine-6-carboxylic acid (195 mg, 100%).
[001173] Préparation 40
[001174] 4-(4-fluorophenvn-2-isopropyl-3,5-dioxo-2,3,4,5-tetrahvdro-l,2,4-triazine-6carboxylic acid
[001175]
[001176]
Step A: To a solution of ethyl 4-(4-fluorophenyl)-5-oxo-3-thioxo-2,3,4,5tetrahydro-l,2,4-trîazine-6-carboxylate (3.85 g, 13.0 mmol) in DMF (35 mL) and acetic acid (14.2 mL, 248.0 mmol) was added 30% aq. H2O2 (2 mL, 65.2 mmol). The mixture was stirred for 3 days. The reaction mixture was extracted between water (200 mL) and EtOAc(120 mL). The aqueous layer was washed with EtOAc (2 x 120 mL). The combined organic layers were washed with water (100 mL), sodium bicarbonate (3 x 100 mL), water (100 mL), and brine (200 mL). The organic layer was then dried with sodium sulfate, filtered and concentrated in vacuo to afford a pale yellow solid. The solid obtained was triturated with ether and filtered. The filtrate was concentrated and purified over silica (0-40% EtOAc in hexanes) to afford ethyl 4-(4-fluoropheny])-3,5-dîoxo2,3,4,5-tetrahydro-l,2,4-triazine-6-carboxylate (1.6 g, 45%).
242
[001177] Step B: To a suspension of ethyl 4-(4-fIuorophenyl)-3,5-dioxo-2,3,4,5-tetrahydrol,2,4-triazine-6-carboxylate (800 mg, 2.87 mmol) and K2CO3 (792 mg, 5.73 mmol) in DMF (10 mL) was added 2-iodopropane (573 pL, 5.73 mmol). The mixture was heated in a sealed tube at 65 °C for 1 h. The cooled mixture was partitioned between water (50 mL) and EtOAc (25 mL) and the aqueous layer was extracted with EtOAc (2 x 25 mL). The combined organic phases were washed with water (5 x 30 mL) and brine (50 mL), then dried over Na2SO4, filtered and concentrated. The residue was purified over silica gel (0-20% EtOAc in hexanes) to afford ethyl 4-(4-fluorophenyl)-2-isopropyl-3,5-dioxo-2,3,4,5-tetrahydro-l,2,4-triazine-6-carboxylate (763 mg, 83%).
[001178] Step C: Sulfuric acid (10 mL) was carefully added to a mixture of ethyl 4-(4fluorophenyl)-2-isopropyl-3,5-dioxo-2,3,4,5-tetrahydro-l ,2,4-triazine-6-carboxylate (763.7 mg, 2.377 mmol) and water (2 mL). The mixture became homogenous after a few minutes. The reaction mixture was stirred at 40 °C overnight, cooled to room température, and carefully added to ice. The mixture was saturated with solid NaCl and was extracted from EtOAc (3x40 mL) The combined organic layers were dried over sodium sulfate, filtered and concentrated to afford 4-(4fluorophenyl)-2-isopropyl-3,5-dioxo-2,3,4,5-tetrahydro-l ,2,4-triazine-6-carboxylic acid (713 mg, 102%).
[001179] The following compounds were also made using the procedure according to Préparation 40.
243
Préparation Structure Name
41 n'nÇ° HoWNrrF 0 0 4-(3,4-d ifluoropheny l)-2ethy l-3,5-dioxo-2,3,4,5tetrahydro-1,2,4-triazine-6carboxylic acid
42 hoyVtV O O 4-(3,4-difluorophenyl)-2isopropy 1-3,5-di oxo-2,3,4,5tetrahydro-1,2,4-triazine-6carboxylic acid
43 N Wn 0 O Γ 2-ethyl-4-(4-fluorophenyl)3,5-dioxo-2,3,4,5-tetrahydro1,2,4-triazine-6-carboxylic acid
[001180]
[001181]
[001182]
[001183]
Préparation 41
5-(4-fluorophenyl)-l-methYl-4-oxo-L4-dihvdropvridine-3-carboxvlic acid
Step A: A mixture of methyl 5-bromo-4-hydroxynicotinate (100 mg, 0.43 I mmol) and CS2CO3 (211 mg, 0.646 mmol) was diluted with DMF (2 mL), placed under a nitrogen atmosphère and heated to 75 °C for 10 min. The reaction mixture was allowed to cool to room température. Mel (40.4 μΐ, 0.646 mmol) was added and the reaction mixture was stirred for 3 h.
The reaction mixture was diluted with water and extracted with DCM/IPA (3:1). The combined organic layers were dried over MgSOi, filtered and concentrated. The residue was purified over silica gel (1-10% methanol in DCM with 1% NH4OH) to afford methyl 5-bromo-l-methyl-4-oxol,4-dihydropyridine-3-carboxylate (92 mg, 0.374 mmol, 86.8 %yie!d).
[001184] Step B: A mixture of methyl 5-bromo-l-methyl-4-oxo-l,4-dihydropyridine-315 carboxylate (92 mg, 0.37 mmol), (4-fluorophenyl)boronic acid (105 mg, 0.75 mmol) and
244
Pd(PPh3)4 (22 mg, 0.019 mmol) was diiuted with dioxane (I mL) followed by the addition of NasCOs (561 μΐ, 1.1 mmol, 2.0 M). The reaction mixture was purged with argon, sealed and heated to 90 °C overnight. The reaction mixture was allowed to cool, and the cooled mixture was diiuted with water and adjusted to pH 2 with IN HCl. The mixture was extracted with three times with 5 DCM/IPA (3/1). The combined organic layers were dried over MgSCL, filtered and concentrated.
The product was triturated with diethyl ether to afford 5-(4-fluorophenyl)-l-methyl-4-oxo-l,4dihydropyridine-3-carboxylîc acid.
[001185] The following compounds were also made using the procedure according to Préparation 41.
Préparation Structure Naine
42 ο o L F 5-(4-fluorophenyl)-lisopropyl-4-oxo-1,4dihydropyridine-3-carboxylic acid
43 nT ο o F 1 -ethyl-5-(4-fluorophenyl)-4oxo-l,4-dihydropyridine-3carboxylic acid
44 X o o=/ T[ Tl 5-(3,4-difluorophenyl)-1 isopropyl-4-oxo-l,4dihydropyridine-3-carboxylic acid
45 ° ° VL-c, 5-(4-chloropheny 1)-1 isopropyl-4-oxo-1,4dihydropyridine-3-carboxylic acid
245
1001186] Préparation 47
[001187] 1 -(4-fluorophenyl)-2-oxo-5-vinyl-L2-dihvdropyridine-3-carboxvlic acid
1001188]
[001189]
Step A: In a 125 mL screw-top pressure vial, methyl 5-bromo-l-(4-fIuorophenyl)2-oxo-l,2-dihydropyridine-3-carboxylate (0.400 g, L227 mmol) was dissolved in 10:1 toluene:water (16 mL) and nitrogen was bubbled through this solution for 5 minutes. Potassium trifluoro(vinyl (borate (0.6572 g. 4.906 mmol), diacetoxypalladium (0.04131 g, 0.1840 mmol), and K3PO4 (0.7811 g, 3.680 mmol) were added with nitrogen bubbling through the reaction for 1 minute. Dicyclohexyl(2',6'-diisopropoxy-[l,r-biphenyl]-2-yl)phosphane (0.1717 g, 0.3680 mmol) was added and the reaction mixture was bubbled with nitrogen for 1 minute, then sealed and heated to 110 °C overnight. The reaction mixture was cooled and partitioned between water and ethyl acetate. The organic layer was washed with water and brine, dried over sodium sulfate, filtered and concentrated. The residue was purified over silica gel (20% EtOAc in DCM) to afford methyl l-(4-fluorophenyl)-2-oxo-5-vinyl-l,2-dihydropyridine-3-carboxylate.
[001190] Step B: Methyl I-(4-fluorophenyl)-2-oxo-5-vinyl-l,2-dihydropyndine-3carboxylate was dissolved in MeOH (50 mL) and water (5 mL). LiOH (500 mg) was added and the reaction mixture was heated to 40 °C for 1.5 h. The MeOH was removed by rotary évaporation and diîuted with water (50 mL). Concentrated HCl was added until pH 1. The résultant precipitate was filtered and dried to afford l-(4-fluorophenyl)-2-oxo-5-vinyl-l ,2-dihydropyridine-3carboxylic acid (209.6 mg, 66% yield).
[001191] The following compounds were also made using the procedure according to Préparation 47.
246
Préparation Structure Name
48 T O A0 b F 1 -(4-fluoro pheny l)-2-oxo-5(prop-1 -en-2-y 1)-1,2dihydropyridine-3-carboxylic acid
49 O O (E)-1 -(4-fluorophenyl)-2-oxo5-(prop-l-en-1-y 1)-1,2dihydropyridine-3-carboxylic acid
[001192] Préparation 50
[001193] 5-(4-t1uorophenyl)-6-oxo-5-azaspiro[2.5]octane-7-carboxylic acid 0 ° Îl 1
[001194] △
[001195] Step A: A solution of l-(hydroxymethyl)cyclopropane-1 -carbonitrile (541 mg, 5.57 mmol) in DCM (10 mL) was cooled to 0 °C in an ice bath and methanesulfonyl chloride (684 pL,
8.36 mmol) was added. followed by triethylamine (1708 μΐ, 12.3 mmol), and the reaction mixture was stirred at 0 °C for l h and warmed to room température. The reaction mixture was diluted 10 with DCM (10 mL), washed with brine, dried over sodium sulfate and concentrated to afford crude ( l-cyanocyclopropyl)methyl methanesulfonate (976 mg, 100% yield).
[001196] Step B: Diethyl malonate (765 μΐ, 5.0] mmol) was added to a solution of sodium hydride (243 mg, 6.08 mmol) in THF (25 mL) followed by the addition of (1cyanocyclopropyl)methyl methanesulfonate (976 mg, 5.57 mmol) and the reaction mixture was 15 refluxed for 4 days. The reaction mixture was cooled and partitioned between MTBE (50mL) and 2N HCl (50 mL), washed of MTBE with 2N HCl (25 mL) and brine (25 mL), dried over sodium sulfate, filtered and concentrated. The crude material was purified over silica gel (5-95% EtOAc in hexanes) to afford diethyl 2-((1-cyanocyclopropyl)methyl)malonate (430 mg, 1.80 mmol, 32.3
247 % yield).
[001197] Step C: Diethyl 2-((1-cyanocyclopropyl)methyl)malonate (275 mg, 1.15 mmol) was dîssolved in EtOH (50 mL) in a 250 mL Parr shaker bottle. PtO? (14 mg, 0.06 mmol) was added and the mixture was maintained under 50 psi H2 for 48 hours. Additional PtÛ2 ( 14 mg, 0.06 mmol) was added to the Parr shaker and the reaction was maintained under at 50 psi H2 overnight. The reaction mixture was filtered, the solids were washed with éthanol and the filtrats was concentrated. The residue was purified over silica gel (0-20% MeOH in EtOAc) to afford ethyl 6oxo-5-azaspiro[2.5]octane-7-carboxylate (98 mg, 43.2% yield).
[001198] Step D: Ethyl 6-oxo-5-azaspiro[2.5]octane-7-carboxylate (0.098 g, 0.4969 mmol), césium carbonate (0.4857 g, 1.491 mmol) and quinolin-8-ol (0.02885 g, 0.1987 mmol) were suspended in DMF (2 mL) and purged under argon for 5 minutes. Copper (I) iodide (0.03785 g, 0.1987 mmol) and l-fluoro-4-iodobenzene (0.08595 mL, 0.7453 mmol) were added and the reaction mixture was heated to 100 7C overnight in a sealed tube under argon. The reaction mixture was cooled and partitioned between water and EtOAc (3 x 25), dried over sodium sulfate, filtered and concentrated. The residue was purified over silica gel (0-100% EtOAc in DCM) to afford ethyl 5-(4-fluorophenyl)-6-oxo-5-azaspiro[2.5]octane-7-carboxylate (40 mg, 0.1373 mmol, 27.63 % yield).
[001199] Step E: Ethyl 5-(4-fluorophenyl)-6-oxo-5-azaspiro[2.5]octane-7-carboxylate (32.7 mg, 0.112 mmol) was dîssolved in EtOH (25 mL) and lithium hydroxide hydrate (9.42 mg, 0.224 mmol) was added as a solid. Water (2 mL) was added and the reaction mixture was heated to 35 °C. After ! h, additional lithium hydroxide hydrate (9.42 mg, 0.224 mmol) was added. After an additional hour, EtOH was removed under reduced pressure. Water (25 mL) was added to the residue and the pH was lowered to <2 with the addition of concentrated HCl. The cloudy suspension was extracted with EtOAc (3x15 mL), dried over sodium sulfate, fîltered and concentrated to afford 5-(4-fluorophenyl)-6-oxo-5-azaspiro[2.5]octane-7-carboxylic acid (23 mg, 0.0874 mmol, 77.8 % yield) as a white solid.
[001200]
[001201]
Préparation 51 l-(4-fluoropheny])-3-methyl-2-oxopiperidine-3-carboxylic acid
[001202]
248
[001203| Step A: Ethyl 2-oxopiperidine-3-carboxylate (500 mg, 2.921 mmol), césium carbonate (2855 mg, 8.762 mmol) and quinolin-8-ol (169.6 mg, 1.168 mmol) were suspended in DMF (6 mL) and purged under argon for 5 minutes. To this solution were added copper(I) iodide (222.5 mg, 1.168 mmol) and 1-fluoro-4-iodobenzene (505.2 pL, 4.381 mmol) and the reaction mixture was heated to 100 °C overnight in a sealed tube. The reaction mixture was cooled and partitioned between EtOAc and water, extracted with EtOAc, washed with brine, dried over sodium sulfate, filtered and concentrated, The residue was purified over silica gel (0-100% EtOAc in DCM) to afford ethyl l-(4-fluorophenyl)-2-oxopiperidine-3-carboxylate (468 mg, 1.76 mmol, 60.4% yield).
[001204] Step B: Ethyl [-(4-fluorophenyl)-2-oxopiperidine-3-carboxylate (468 mg, 1.76 mmol) was dissolved in DCM (10 mL). Césium carbonate (2299 mg, 7.06 mmol) was added and the reaction mixture was stirred for 5 min. lodomethane (659 pi, 10.6 mmol) was added and the reaction mixture was stirred overnight at room température. Additional iodomethane (659 pl, 10.6 mmol) was added and the reaction mixture was heated to 35 °C for 5 h and then at room température over the weekend. The reaction mixture was diluted with DCM (50 mL), filtered through Celite® and concentrated. The residue was purified over silica gel (0-40% EtOAc in hexanes) to afford ethyl l-(4-fluorophenyl)-3-methyl-2-oxopiperidine-3-carboxylate (420 mg, 1.50 mmol, 85.2 % yield) as a clear oil that was used dîrectly in the next step without purification. [001205] Step C: Ethyl l-(4-fluorophenyl)-3-methyl-2-oxopiperidine-3-carboxylate (420 mg, 1.50 mmol) was dissolved in EtOH (3 mL), and lithium hydroxide hydrate (63.1 mg, 1.50 mmol) was added, foliowed by water (3 mL) and the reaction mixture was heated to 35 °C. After 2 h, additional lithium hydroxide hydrate (63.1 mg, 1.50 mmol) was added and the reaction mixture was allowed to stir at room température overnight. The reaction mixture was concentrated, diluted with water (25 mL) and the pH was lowered to <2 by the addition of concentrated HCl. The cloudy mixture was extracted with EtOAc, washed with brine, dried over sodium sulfate, filtered and concentrated to afford l-(4-fluorophenyl)-3-methyl-2-oxopiperidine-3-carboxylic acid (390 mg, 1.55 mmol, 103 % yield).
[001206] Préparation 52
[001207] 4-((6-aminopyridin-3-vl)oxv)-N-(l-methoxv-2-methvlpropan-2-Yl)-l-(4methoxvbenzyl)-IH-pvrazoloi3,4-b1pvridin-3-amine
249
[001208) PMB
1001209] Step A: 4-Chloro-l H-pyrazolo[3,4-b]pyridine (11.6 g, 75.5 mmol) was dissolved in DMF (100 mL) and ground potassium hydroxide (12.7 g, 227 mmol) was added, followed by h (34.5 g, 136 mmol). The resultîng dark solution was stirred under nitrogen at 50 °C for 2 h. The reaction mixture was quenched by addition of aqueous 10% NaHSOs (75 mL). The suspension was further diluted by addition of water (100 mL) and filtered. The resulting precipitate was washed with water (3x30 mL), dried by toluene azeotrope (3 x 50 mL) by rotary évaporation to afford 4-chloro-3-iodo-lH-pyrazolo[3,4-b]pyridine (21.0 g, 88%).
[001210] Step B: 4-Chloro-3-iodo-lH-pyrazolo[3,4-b]pyridine (21.0 g, 75.1 mmol) was dissolved in DMF (100 mL). K2CO3 (20.8 g, 150 mmol) and l-(chloromethyl)-4-methoxybenzene (12.3 ml, 90.2 mmol) were added and the reaction mixture was stirred under nitrogen overnight. The reaction mixture was diluted with water (100 mL) and filtered. The isolated solids were washed with water (3 x 30 mL) and purified over silica gel (25% EtOAc in hexanes) to afford 4chloro-3-iodo-l-(4-methoxybenzyl)-lH-pyrazolo[3,4-b]pyridine.
[0012111 Step C: To a mixture of 2-chloro-5-hydroxypyridine (1.69 g, 13.1 mmol) and 4chloro-3-iodo-l-(4-methoxybenzyl)-lH-pyrazolo[3,4-b]pyridine (2.61 g, 6.53 mmol) in DMA (32 mL) was added CS2CO3 (6.38 g, 19.6 mmol) and the reaction mixture was heated to 120 °C for 1 h. The reaction mixture was cooled to room température and poured into water (500 mL). The resulting solid was filtered and purified over silica gel (25-100% EtOAc in hexanes) to afford 4((6-chloropyridin-3-yl)oxy)-3-iodo-l-(4-methoxybenzyl)-lH-pyrazolo[3,4-b]pyridine (1.87 g, 3.80 mmol, 58.1 % yield) as a white solid.
[001212] Step D: (2-Methoxy-l,l-dimethylethyl)amine (0.185 mL, L52 mmol), 4-((6chloropyridin-3-yl)oxy)-3-iodo-l-(4-methoxybenzyl)-lH-pyrazolo[3,4-b]pyndine (0.250 g, 0.507 mmol), K2CO3 (0.561 g, 4.06 mmol) and pyrrole-2-carboxylic acid (0.0282 g, 0.254 mmol) were suspended in DMSO (2.5 mL) and the reaction mixture was degassed for 5 min with Ar. Copper(I) iodide (0.0483 g, 0.254 mmol) was added and the reaction mixture was heated to 70 °C under argon for 66 h. The cooled reaction mixture was added to 50 mL water and extracted with DCM,
250 washed with saturated NaHCOa (15 mL) and brine (15 mL), dried over sodium sulfate, fîltered and concentrated. The residue was purifîed over silica gel (10-100% EtOAc in hexanes) to afford 4-((6-chloropyridin-3-yl)oxy)-N-(i -methoxy-2-methylpropan-2-yl)-l-(4-methoxybenzyl)-lHpyrazolo[3,4-b]pyridin-3-amine (0.147 g, 0.276 mmol, 54.5 % yield).
[001213] Step E: To a solution of 4-((6-chloropyridin-3-yl)oxy)-N-(l-methoxy-2methylpropan-2-yl)-l-(4-methoxybenzyl)-lH-pyrazolo[3,4-b]pyridin-3-amine (0.050 g, 0.11 mmol) in toluene (0.5 mL) was added 2-dicyclohexylphosphino-2',6'-dimethoxy-l,r-biphenyl (0.0066 g, 0.016 mmol) followed by tris(dibenzylideneacetone)dipalladium (0) (0.0049 g, 0.0053 mmol) and lithium bis(trimethylsîlyl)amide (0.040 mL, 0.21 mmol). The solution was degassed with Ar for 10 min and heated to 80 °C over the weekend. The cooled reaction mixture was poured into water (25 mL) and extracted with DCM. The combined organîc layers were washed with saturated NaHCO3 (15 mL) and brine (15 mL), dried over sodium sulfate, fîltered and concentrated. The residue was purifîed over silica gel (10-50% EtOAc in hexanes) to obtain 4-((6aminopyridin-3-yl)oxy)-N-(l-methoxy-2-methylpropan-2-yl)-l-(4-methoxybenzyl)-IHpyrazolo[3,4-b]pyndin-3-amine (0.033 g, 0,037 mmol, 34 % yield).
[001214] Préparation 53
[001215] (R)-2-((4-(4-amino-2-f]uorophenoxv)-l-(4-methoxvbenzvl)-lH-pvrazolof3,4b1pyridin-3-vl)amino)propan-l-ol
[001216]
N N PMB
[001217| 3-Fluoro-4-((3-iodo-l-(4-methoxybenzyl)-l H-pyrazolo[3,4-b]pyridin-4yl)oxy)aniline (Préparation 1; 1.0 g, 2.04 mmol), Κ:ΟΟ3 (2.26 g, 16.3 mmol) and pyrrole-2carboxylic acid (113 mg, 1.02 mmol) were suspended in DMSO (20 mL) and the mixture was degassed for 5 min with Ar. Cu(I)iodide (194 mg, 1.02 mmol) and D-alaninol (476 pL, 6.12 mmol) were added and the mixture heated to 60 °C in a sealed tube overnight. The mixture was cooled and partitioned between water (100 mL) and EtOAc ( 100 mL) and the aqueous layer was extracted with EtOAc (2 x 50 mL). The combined organîc phases were washed with water (5 x 30 mL) and brine (30 mL), dried over NaaSOi, fîltered and concentrated. The residue was purifîed over silica
251 gel (0-3% MeOH in DCM)to afford (R)-2-((4-(4-amino-2-fluorophenoxy)-l-(4-methoxybenzyl)lH-pyrazolo[3,4-b]pyridin-3-yl)amino)propan-I-ol (0.716 g, 80%) as a light brown foam.
[001218] The following compounds were also made using the procedure according to Préparation 53.
Préparation Structure Name
54 X O u V S z 1 \ Z 1 Z X (S)-2-((4-(4-amino-2fluorophenoxy)-1 -(4methoxybenzyl)-1Hpyrazolo[3,4-b]pyridin-3yl)amino)propan-l -ol
55 λ-\ XX /^NH θ ΛΑ PMB (S)-4-(4-amino-2fluorophenoxy)-l-(4methoxybenzyl)-N-(lmethoxypropan-2-yl)-1Hpyrazolo[3,4-b]pyrtdin-3amine
56 v XJ /NH θ N^JO PMB (R)-4-(4-amino-2fluorophenoxy)-1 -(4m ethoxy benzy 1)-N -( 1 methoxypropan-2-y])-] Hpyrazolo[3,4-b]pyridin-3amine
57 F. ,;-. ζ°Λ XJ 9 Άυ PMB 4-(4-amino-2-fluorophenoxy)- N-(l-methoxy-2methylpropan-2-yl)-l-(4methoxybenzyl)-l Hpyrazolo[3,4-b]pyridin-3amîne
252
4-(4-amino-2-fluorophenoxy)N-(4,4-difluorobutan-2-yl)-l(4-methoxybenzyl)-1Hpyrazolo[3,4-b]pyrîdîn-3amine (S)-4-(4-amino-2fl uorophenoxy)-N -( 1 ethoxypropan-2-yl)-1 -(4methoxybenzyl)-1Hpyrazoio[3,4-b]pyridin-3amine
[001219] Préparation 60
[001220] (R)-2-((4-((6-aminopyridin-3-yl)oxy)-l-(4-methoxybenzyl)-l H-pyrazolo[3,45 b]pyridin-3-yl)amino)propan-l-ol
[001221]
[001222]
Step A: D-Alaninol (0.142 ml, 1.83 mmol), 4-((6-chloropyridin-3-yl)oxy)-3-iodoI-(4-methoxybenzyl)-IH-pyrazolo[3,4-b]pyridine (0.300 g, 0.609 mmol), K2CO3 (0.673 g, 4.87 mmol) and pyrrole-2-carboxylic acid (0.0338 g, 0.304 mmol) were suspended in DMSO (6.09 ml, 10 0.609 mmol) and then this mixture was degassed for 5 min with Ar. Copper(I) iodide (0.0483 g,
0.254 mmol) was added and heated to 60 °C under argon overnight. The cooled reaction mixture was added to 25 mL cold water while stirring, the résultant solids were filtered and washed with 10 mL hexanes. The solids were purified over silica gel (10-100% EtOAc in hexanes) to afford (R)-2-((4-((6-chloropyridin-3-yl)oxy)-l-(4-methoxybenzyl)-lH-pyrazolo[3,4-b]pyridin-315 yl)amino)propan-l-ol (0.155 g, 0.352 mmol, 57.9 % yield) as a clear oil.
[001223] Step B: (R)-2-((4-((6-chloropyridin-3-yl)oxy)-l-(4-methoxybenzyl)-l H20534
253 pyrazolo[3,4-b]pyridin-3-yl)amino)propan-]-ol (0.155 g, 0.352 mmol) was dissolved in DMF (3.52 ml, 0.352 mmol), cooled to 0 °C and then treated with imidazole (0.0360 g, 0.529 mmol) followed by tert-butyldimethylsilyl chloride (0.0797 g, 0.529 mmol). The mixture was allowed to warm to room température over 1 h. The reaction mixture was partitioned between water and EtOAc, washed with water and brine, dried over sodium sulfate, filtered and concentrated to afford (R)-N-( 1 -((tert-butyldimethylsilyi)oxy)propan-2-yl)-4-((6-chloropyridin-3-yl)oxy)-l-(4methoxybenzyl)-lH-pyrazolo[3,4-b]pyridin-3-amme (0.218 g, 0.393 mmol, 112 % yield) as a clear oil.
[001224] Step C: To a mixture of tert-butyl carbamate (0.277 g, 2.36 mmol), CS2CO3 (0.256 g, 0.787 mmol), tris(dibenzylideneacetone)dipalladium (0) (0.0360 g, 0.0393 mmol), and 2(dicyclohexylphosphino)-2’,4',6'-tri-i-propyl-l,r-biphenyl (0.0375 g, 0.0787 mmol) was added a solution of (R)-N-(l-((tert-butyldimethylsilyl)oxy)propan-2-yl)-4-((6-chloropyridin-3-yl)oxy)-l(4-methoxybenzyl)-lH-pyrazoîo[3,4-b]pyridin-3-amine(0.218 g, 0.393 mmol) in l,4-dioxane(3.9 ml). This mixture was degassed for 2 min with Ar and heated to 100 °C for 16 h in a sealed tube. The reaction mixture was cooled and partitioned between EtOAc (2x25 mL) and water (25 mL), washed with brine (25 mL), dried over sodium sulfate, filtered and concentrated. The residue was purified over silica gel (10-50% EtOAc in hexanes) to afford tert-butyl (R)-(5-((3-((l-((tertbutyldimethylsilyl)oxy)propan-2-yl)amino)-1 -(4-methoxybenzyl)-1 H-pyrazolo[3,4-b]pyridin-4yl)oxy)pyridin-2-yl)carbamate (0.147 g, 0.232 mmol, 58.9 % yield) as a white solid.
[001225] Step D: To tert-butyl (R)-(5-((3-((l-((tert-butyldimethylsilyl)oxy)propan-2yl)amino)-1-(4-methoxybenzyl)-1 H-pyrazolo[3,4-b]pyridm-4-yl)oxy)pyridin-2-yl)carbamate (0.147 g, 0.232 mmol) was added 4N HCI in 1,4-dioxane (9.26 ml, 0.232 mmol) and the reaction mixture was stirred overnight. The reaction mixture was concentrated and partitioned between DCM and IN NaOH, washed with water and brine, dried over sodium sulfate, filtered and concentrated to afford (R)-2-((4-((6-aminopyridin-3-y!)oxy)-l-(4-methoxybenzyl)-lHpyrazolo[3,4-b]pyridin-3-yl)amino)propan-l-ol (0.091 g, 0.216 mmol, 93.5 % yield).
[001226] The following compound was also made using the procedure according to Préparation 60.
Préparation
Structure
Name
254
61 F Λ-y \ lï F /NH PMB 4-((6-aminopyridin-3-y l)oxy)N-(4,4-difluorobutan-2-yl)-l (4-methoxybenzyl)-1 Hpyrazolo[3,4-b]pyridin-3amine
62 \ ^_nh2 [H / J'NH ° œ PMB (R)-4-((6-aminopyridin-3yl)oxy)-l-(4-methoxybenzyl)N-( 1 -methoxypropan-2-yI)1 H-pyrazolo[3,4-b]pyridin-3amine
[001227] Préparation 63
[001228] N-(3-fluoro-4-((3-iodo-l-(4-methoxybenzyl)-lH-pyrazolo[3,4-b]pyridin-4 yl)oxy)phenyI)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide
[001229] PMB
[001230] To a 200 mL flask was added 3-fluoro-4-((3-iodo-l-(4-methoxybenzyl)-lH pyrazolo[3,4-b]pyridin-4-yl)oxy)anîline (5 g, 10.2 mmol), 2-(4-fluorophenyl)-3-oxo-2,3dihydropyridazine-4-carboxylic acid (2.87 g, 12.2 mmol), 3-(((ethylimino)methylene)amino)N ,N-dim ethyl propan-1 -amine hydrochloride (2.93 g, 15.3 mmol), and lH-benzo[d][l,2,3]triazoll-ol (2.07 g, 15.3 mmol). DMF (50 mL) was stirred at room température for 1 h. The reaction mixture was poured into ice water (200 mL) and allowed to stir for 30 min. The precipitate was filtered and washed with water (100 mL) and hexanes (100 mL). The product was trîturated with MeOH and dried to afford N-(3-fluoro-4-((3-iodo-l-(4-methoxybenzyl)-lH-pyrazolo[3,4b]pyridin-4-yI)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide (6.68 g, 92%) as a yellow brown solid.
[001231] The following compound was also made using the procedure according to
255
Préparation 63.
Préparation
Name
N-(3-fluoro-4-((3-iodo-1 -(4methoxybenzyl)-1Hpyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-2-(4fluorophenyl)-6-methyl-3oxo-2,3-dihydropyridazine-4carboxamide
[001232] Préparation 65
[001233] N-(3-fluoro-4-((3-iodo-l-(4-methoxybenzy!)-IH-pyrazoIo[3,4-b]pyridin-4yl)oxy)phenyl)-3-(4-fluorophenyl)-l-isopropy 1-2,4-dioxo-l,2,3,4-tetrahydropyrimidine-5carboxamide
[001234]
[001235] To a stirred solution of 3-(4-fluorophenyl)-i-isopropyl-2,4-dioxo-l,2,3,410 tetrahydropyrimidine-5-carboxylic acid (500 mg, 1.71 mmol) and HATU (0.887 g, 2.33 mmol) in DMF (10 mL) at room température was added DIEA (813 pL, 4.67 mmol) followed by 3-fluoro4-((3-iodo-l-(4-methoxybenzyl)-lH-pyrazolo[3,4-b]pyridin-4-y])oxy)aniline (762 mg, 1.56 mmol). The réaction mixture was stirred at room température overnight and partitioned between water (30 mL) and EtOAc (30 mL) and the aqueous layer was extracted with EtOAc (2 x 20 mL).
The combined organic phases were washed with water (5x10 mL) and brine (10 mL), dried over Na2SO4, filtered and concentrated. The residue was purified over silica gel (30-100% EtOAc in hexanes) to afford N-(3-fluoro-4-((3-iodo-l -(4-methoxybenzyl)-IH-pyrazolo[3,4-b]pyridin-4
256 yl)oxy)phenyl)-3-(4-fluorophenyl)-l-isopropyl-2,4-dioxo-l,2,3,4-tetrahydropyrimidine-5carboxamide (1,13 g, 95%).
[001236] The following compounds were also made using the procedure according to Préparation 65.
Préparation Structure Name
66 H ni χτΓίΌ PMB N-(3-fluoro-4-((3-iodo-l-(4methoxybenzyl)-1Hpy razo lo[3,4-b] py ri d in-4yl)oxy)phenyl)-2,5-dioxo-l phenyl-1,2,5,6,7,8hexahydroquinoline-3carboxamide
67 _XJ 0 0 । O F Nbo PMB N-(3-fluoro-4-((3-iodo-l-(4methoxybenzyl)-1Hpyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-l-(4fluorophenyl)-2,5-dioxo1,2,5,6,7,8hexahydroquinoline-3carboxamide
257
Example 1
[001237] N-(3-fluoro-4-(ï3-((2-hvdroxyeth¥l)amino)-lH-pvrazolof3.4-b]pvridin-4¥l)oxv)phenvl)-2-(4-fluorophenyl)-3-oxo-2,3-dihvdropvridazine-4-carboxamide
[0012381
1001239] Step A: Ethanolamine (0.0213 mL, 0.354 mmol), N-(3-fluoro-4-((3-iodo-l-(4methoxybenzyl)-lH-pyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3dihydropyridazine-4-carboxamide (Préparation 63; 0.050 g, 0.0708 mmol), K2CO3 (0.0489 g, 0.354 mmol) and L-proline (0.00407 g, 0.0354 mmol) were suspended in DMSO (0.708 mL) and degassed for 5 min with Ar. Copper(l) iodide (0.00674 g, 0.0354 mmol) was added and the reaction mixture was heated to 80 °C under argon overnight. The reaction mixture was partitioned between water (15 mL) and EtOAc (15 mL) and the combined the organic layers were washed with brine, dried over NazSCU, filtered and concentrated to afford crude N-(3-fluoro-4-((3-((2hydroxyethyl)amino)-l-(4-methoxybenzyl)-lH-pyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-2-(4fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide, which was taken in the next step without further purification.
[001240] Step B: To a solution of crude N-(3-fluoro-4-((3-((2-hydroxyethyl)amino)-l-(4methoxybenzyl)-lH-pyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3dihydropyridazine-4-carboxamide (0.0453 g, 0.0708 mmol) in 2 mL DCM was added 4 mL TFA and the reaction mixture was stirred for 2 hr at 50 °C. The reaction mixture was concentrated in vacuo and resuspended in 2 mL of a solution of 60:40 ACN:water with 2% TFA. The product was purified by Cl8 HPLC (5-95% ACN in water with 0.2% TFA). The fractions containing the product were free-based with saturated NaHCOs ( 15 mL) and the water layer was extracted with DCM (2X15 mL). The pooled organic layer was washed with brine (15 mL), dried over NasSOi, filtered and concentrated in vacuo to afford N-(3-fluoro-4-((3-((2-hydroxyethyl)amino)-lHpyrazoto[3,4-b]pyndin-4-yl)oxy)pheny 1)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4carboxamide (0.0022 g, 0.00424 mmol, 5.98 % yield) as an off-white solid. Mass spectrum: m/z
258 = 520.1 (M+H). 'H NMR (d&-DMSO) δ 12.20 (s, 1H), 11.70 (s, 1H), 8.39 (d, 1H), 8.27 (d, IH), 8.16 (s, 1H), 8.04 (d, 1H), 7.69 (m, 1H), 7.61 (m, 1H), 7.51 (t, 1H), 7.42 (m, 2H), 6.04 (d, IH), 3.64 (t, 2H), 3.36 (t, 2H).
[001241] The following compounds were also synthesized using the procedure according to 5 Example 1.
Ex. No. Structure Name Mass spectrum (apci) m/z
2 θ' Vz Z Λ— Ο τι -y zz 0=( z-z N-(3-fluoro-4-((3-((2methoxy ethy l)am ino)lH-pyrazoio[3,4b]pyridin-4yl)oxy)pheny 1)-2-(4fluorophenyl)-3-oxo2,3-dihydropyridazine4-carboxamide 534.1 (M+H)
3 H O ho-, T ΙΓ π π ]| ] -Λ ο O N H 0 F Λϊί N N N-(3-fluoro-4-((3-((lhydroxy-2methylpropan-2yl)amino)-l Hpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-2-(4fluorophenyl)-3-oxo2,3-dihydropyridazine4-carboxamîde 548.1 (M+H)
259
4 Η ηο-ΔΖ T II Π II T ] \ ο ο ί Ar ^NH θ F ΛΑ H N N-(3-fluoro-4-((3-((2- hydroxy-2methy[propyl)amino)lH-pyrazolo[3,4b]pyridin-4yl)oxy)phenyl)-2-(4fluorophenyl)-3-oxo2,3-dihydropyridazîne4-carboxamide 548.2 (M+H)
5 u_ z-z C /^° )=o IZ ô °O ,. 5^ -ZI x Z ^_- y (R)-N-(3-fluoro-4-((3- ((2hydroxypropyl)amino)lH-pyrazolo[3,4b]pyridin-4yI)oxy)phenyl)-2-(4fluoropheny[)-3-oxo2,3-dihydropyridazine4-carboxamide 534.1 (M+H)
6 z-z^~ ^=O A0 IZ Q „ /\„ _yF^ U- O—(' Z : .K °< J (S)-N-(3-fluoro-4-((3((2hydroxypropyl)amino)lH-pyrazolo[3,4b]pyrîdin-4yl)oxy)pheny 1)-2-( 4fluorophenyl)-3-oxo2,3-dihydropyridazine4-carboxamide 534.1 (M+H)
260
7 H HO^ T il ïï π Ύ ] /^NH 9 F M H N (S)-N-(3-fluoro-4-((3- (( 1 -hydroxypropan-2yl)ammo)-lHpyrazolo[3,4-b]pyridin4-y l)oxy)pheny 1)-2-(4fluorophenyl)-3-oxo2,3-dihydropyridazine4-carboxamide 534.1 (M+H)
8 F-^γΝγθ^ ho^ T ï Π TT ]i i \ «A/ ο o /‘‘NH θ F NAj H N (R)-N-(3-fluoro-4-((3- (( 1 -hydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-2-(4fluoropheny l)-3-o xo2,3-dihydropyridazine4-carboxamide 534.1 (M+H)
9 Η°νλ/ 'f O O XL ^NH O F M H N N-(3-fluoro-4-((3-((3hydroxy-2,2dimethylpropyl)amino) -lH-pyrazolo[3,4b]pyridin-4yl)oxy)phenyi)-2-(4fluorophenyl)-3-oxo2,3-dihydropyridazine4-carboxamide 562.2 (M+H)
10 hVh yLX?ti VNH 0 F nAj N N (S>N-(4-((3-((253dihy d roxy propy l)am i no )-1 H-pyrazolo[3,4b]pyridin-4-yl)oxy)-3fluorophenyl)-2-(4fluorophenyl)-3-oxo- 550.1 (M+H)
261
2,3-dihydropyridazine4-carboxamide
11 .9 « zz o z-z 0 N-(4-((3- (cyclobutylamino)-IHpyrazolo[3,4-b]pyridin4-yl)oxy)-3fl uoropheny 1)-2-(4fluorophenyl)-3-oxo2,3-dihydropyridazine4-carboxamide 530.2 (M+H)
12 F-\Xh 0^ ° ° Nbô H N-(4-((3-((3,3d i fluorocyc lo b uty l)am i no)-lH-pyrazo!o[3,4b]pyridin-4-yl)oxy)-3fluorophenyl)-2-(4fluorophenyl)-3-oxo2,3-dihydropyridazine4-carboxamide 566.2 (M+H)
13 hVh Y/rÇvN \ 0 Ο ^NH 9 F 0TO ü N (R)-N-(4-((3-((2,3dihydroxypropyl)amino )-114-pyrazolo[3,4b]pyridin-4-yl)oxy)-3fluoropheny 1)-2-(4fl uoropheny l)-3-oxo2,3-dihydropyridazine4-carboxamide 550.2 (M+H)
262
14 xo .9 iz' 'Vi Z /)—Ο τι zz °=o> z-z Tl N-(3-fluoro-4-((3- (((lr,3r)-3methoxycyclobutyl)ami no)-lH-pyrazolo[3,4b]pyridin-4yl)oxy)phenyI)-2-(4fluoropheny l)-3 -oxo2,3-dihydropyridazine4-carboxamide 560.2 (M+H)
15 xo J iz T^z Z —0 71 zz 0=/ z-z Tl N-(3-fluoro-4-((3- (((ls,3s)-3methoxycyc lobuty l)ain i no)-1 H-pyrazolo[3,4b]pyridin-4yl)oxy )pheny [)-2-(4fl uoropheny l)-3 -oxo2,3-dihydropyridazine4-carboxamide 560.2 (M+H)
16 v/yÇyn o-.xX/ 0 0 z \x^”NH θ F Λο N N N-(3-fluoro-4-((3(((I s,3s)-3-hydroxy-l methylcyc[obuiyl)amtn o)-l H-pyrazolo[3,4b]pyridin-4y l)oxy)pheny 1)-2-(4fluorophenyl)-3-oxo2,3-dihydropyridazine4-carboxainide 560.2 (M+H)
Example 17
[001242] N-(3-fluotO-4-((3-((l-(methoxvmeth¥l)cvclopropvDamino)-1 H-pyrazoloI3.4
263
b]pvridÎn-4-y[)oxv)phenvl)-2-(4-fluoronhenvl)-3-oxo-2.3-dihvdropvridazine-4-carboxamide
NH o-M ° ° Λο
[001243] H
[001244] I-(Methoxymethyl)cyclopropan-l -amine hydrochloride (0.087 g, 0.637 mmol),N(3-fluoro-4-((3-iodo-l-(4-methoxybenzyl)-1H-pyrazolo[3,4-b]pyridin-4-yl)oxy)pheny 1)-2-(4fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide (Préparation 63; 0.150 g, 0.212 mmol), K2CO3 (0.235 g, 1.70 mmol) and pyrrole-2-carboxylic acid (0.0118 g, 0.106 mmol) were suspended in DMSO (2.12 ml, 0.212 mmol) and the reaction mixture was degassed for 5 min with Ar. Copper(I) iodide (0.0202 g, 0.106 mmol) was added and the reaction mixture was heated to 60 °C under argon for 22 h. The mixture was partitioned between water and EtOAc, the aqueous phase was extracted with EtOAc and the combined organic layers were washed with brine (2X15 mL), dried over sodium sulfate, filtered and concentrated. The resulting oil was purified over silica gel (0-10% MeOH in DCM). The isolated product was suspended tn 5 mL DCM and 10 mL TFA and stirred for 16 h at 50 °C. The mixture was concentrated in vacuo and the residue was suspended in 2 mL of a solution of 60:40 ACN:water with 2% TFA. The product was purified by C18 HPLC (5-95% ACM in water with 0.2% TFA). The fractions containing the product were free-based with saturated NaHCOj (15 mL) and the aqueous phase was extracted with DCM (2X15 mL). The combined organic layers were washed with brine (15 mL), dried over sodium sulfate, filtered and concentrated to afford N-(3-fluoro-4-((3-((]-(methoxyniethyl)cyclopropyl)amino)-lHpyrazolo[3,4-b]pyridin-4-yl)oxy)pheny 1)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4carboxamide (0.0221 g, 0.0395 mmol, 18.6 %yield) as ayellow solid. Mass spectrum: m/z = 560.2 (M+H). Ή NMR (ds-DMSO) δ 12.23 (s, 1H), 11.69 (s, 1H), 8.38 (d, 1H), 8.26 (d, 1H), 8.14 (d, 1H), 8.02 (dd, 1H), 7.68 (m, 2H), 7.59 (m, 1H), 7.48 (t, 1H), 7.41 (m, 2H), 6.03 (m, 1H), 5.72 (s, 1 H), 3.55 (s, 2H), 3.25 (s, 3H), 0.82 (m, 2H), 0.72 (m, 2H).
[001245] The following compounds were also synthesized using the procedure according to Example 17.
264
Ex. No. Structure Name Mass spectrum (apci) m/z
18 LL· 0 z-z γο )=° IZ P Λ Y z /•O N-(3-fluoro-4-((3-((lmethoxy-2methylpropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yI)oxy)phenyl)-2-(4fl uoropheny l)-3-o xo2,3-dihydropyridazine4-carboxamide 562.2 (M+H)
19 ύ z-z c )=° Y° IZ h.. /Λ^ζ N-(3-fluoro-4-((3-((2methoxyethyl)(methyl) amino)-1Hpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-2-(4fl uoropheny!) -3-oxo2,3-dihydropyridazine4-carboxamide 548.1 (M+H)
20 Anh Y J ° ° nYj H N N-(3-fluoro-4-((3-((l(methoxymethyl)cyclo pentyl)amino)-lHpyrazolo[3,4-b]pyridin4-y l)oxy)pheny l)-2 -(4fl uoropheny 1)-3-oxo2,3-dihydropyridazine4-carboxamide 588.2 (M+H)
265
21 LL· 0 zz 0 y=o IZ p ,, U. 0—0 z N-(3-fluoro-4-((3-((l(methoxymethyl)cyclo butyl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-2-(4fluorophenyl)-3-oxo2,3-dihydropyrîdazine4-carboxamide 574.2 (M+H)
22 1 ^o “ô^ dr<w 0 ZI o=n 0Z η N-(3-fkioro-4-((3-((4- methoxy-2methylbutan-2yl)amîno)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-2-(4fluorophenyl)-3-oxo2,3-dihydropyridazine4-carboxamide 576.2 (M+H)
23 . N „¥ Z /)—O 71 ZI °A o=c# z-z m N-(3-fluoro-4-((3(((tetrahydrofuran-2yl)metbyl)amino)-l Hpyrazolo[354-b]pyridin4-yl)oxy)phenyl)-2-(4fluorophenyl)-3-oxo2,3-dihydropyridazine4-carboxamide 560.2 (M+H)
24 ,V/A° Q ZI °A z-z -n N-(4-((3-(((l,ldioxidotetrahydrothiop hen-3yl)methyl)amino)-1HpyrazoloP^-blpyridin4-yl)oxy)-3- 608.1 (M+H)
266
fluoropheny 1 )-2-(4fluorophenyl)-3-oxo2,3-dihydropyridazine4-carboxamide
25 > NH ° ° N' 'N H M (S)-N-(3-fluoro-4-((3- (( 1 -methoxypropan-2yi)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-2-(4fluoropheny 1) -3 -oxo2,3-dihydropyridazine4-carboxamide 548.2 (M+H)
26 % ^NH ° ° ;' Γ H N N -(4-((3-((1,3dimethoxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)-3fluorophenyl)-2-(4Ω uoropheny 1)-3-0 xo2,3-dihydropyridazine4-carboxamide 578.2 (M+H)
27 LL Z-Z c 2=0 xz PLn UX LL N-(4-((3-((4,4difluorobutan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)-3fluoropheny 1)-2-(4fl uoropheny l)-3-o xo2,3-dÎhydropyridazine4-carboxamide 568.2 (M+H)
267
28 Ο / ΖΤ ζ-ζ N-(3-fluoro-4-((3-((cis2(methoxymethyl)cyclo pentyl)amino)-l Ηpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-2-(4fl uoropheny 1)-3-oxo2,3-dihydropyridazine4-carboxamide 588,2 (M+H)
29 LL 2’2 )=ο τζ °·^ζ N-(3-fluoro-4-((3(((tetrahydrofuran-3yl)methyl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-2-(4fl uoropheny l)-3-o xo2,3-dihydropyridazine4-carboxamide 560.2 (M+H)
30 0 ζ-ζ 0 ^ο /=ο τζ Λ & xzÇ^y^ ο N-(3-fluoro-4-((3-(((2oxopiperidin-4yl)inethyl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)pheny 1)-2-(4fluorophenyl)-3-oxo2,3-dihydropyridazine4-carboxamide 587.2 (M+H)
31 Η C? w ΤΤ ϊϊ ΊΡι N V^NH R F ' N^Ô Ν 'Ν' Η N-(4-((3-(((ls,3s)-3(dimethyicarbamoyl)cy clobutyl)amino)-l Hpyrazolo[3,4-b]pyridin4-yl)oxy)-3fluoropheny 1)-2-(4- 601.2 (M+H)
268
f1uorophenyl)-3-oxo2,3-dihydropyridazine4-carboxamide
Ex ample 32
[001246] N-i4-(ü-i(L3-dÎhvdrox¥-2-methylpronan-2-vl)arnÎno)-lH-pyrazolor3,4b1pvridin-4-vl)oxy)-3-fluorophenvl)-2-Î4-nLiorophenvl)-3-oxo-2,3-dihydropvridazine-4carboxamide
[001248] 2-Amino-2-methyl-l,3-propanediol (0.0670 g, 0.637 mmol), N-(3-fluoro-4-((3iodo-l-(4-methoxybenzy])-]H-pyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3oxo-2,3-dihydropyridazine-4-carboxamide (Préparation 63; 0.150 g, 0.212 mmol), K2CO3 (0.235 g, 1.70 mmol) and pyrrole-2-carboxylic acid (0.0118 g, 0.106 mmol) were suspended in DMSO (2.12 ml, 0.212 mmol) and degassed for 5 min with Ar. Copper(I) iodide (0.0202 g, 0.106 mmol) was added and the reaction mixture was heated to 60 °C under argon for 22 h. To this mixture were added Cul Û.5 eq., pyrrole-2-carboxylic acid (0.5 eq.) andl eq. of 2-amino-2-methyl-l,3propanediol and the reaction mixture was stirred at 70 °C for 16 h. The reaction mixture was cooled, then partitioned between water and EtOAc. The aqueous phase was extracted with EtOAc and the combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated in vacuo. The resulting oil was purified over silica gel (30-100% EtOAc in hexanes). The isolated product was suspended in 2 mL DCM and 4 mL TFA and stirred for 16 h at 50 °C. The mixture was concentrated in vacuo and the residue was suspended in 5 mL DCM and 5 mL IM LiOH and stirred at room température for 30 min. The mixture was partitioned between saturated NaHCO3 (15 mL) and DCM (15 mL). The aqueous phase was extracted with EtOAc (2 X 15 mL). The combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated. The residue was suspended in 3 mL of a solution of 60:40 ACN:water
269 with 2% TFA. The product was purified via Cl 8 chromatography (5-95% ACN in water with 0.2% TFA). The fractions containing the product were free-based with saturated NaHCOs (15 mL) and the aqueous phase was extracted with DCM. The combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated to afford N-(4-((3-((l,3-dihydroxy-25 methylpropan-2-yl)amino)-1 H-pyrazolo[3,4-b]pyridin-4-yl)oxy)-3-fluorophenyl)-2-(4fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide (0.0127 g, 0.0225 mmol, 10.6 % yield) as a yellow solid. Mass spectrum: m/z = 564.2 (M+H). 'H NMR (dô-DMSO) δ 12.21 (s, 1H), 11.70 (s, 1H), 8.38 (d, 1H), 8.26 (d, 1H), 8.15 (d, 1H), 8.03 (dd, 1H), 7.68 (m, 2H), 7.59 (m, 1H), 7.50 (t, 1H), 7.41 (m, 2H), 6.04 (dd, 1H), 5.24 (s, 1H), 4.87 (t, 2H), 3.65 (dd, 2H), 3.50 (dd, 10 2H), 1.32 (s, 3H).
270
[001249] The following compounds were also synthesized using the procedure according to Example 32.
Ex. No. Structure Name Mass spectrum (apci) m/z
33 HO\ p ' ° ° nTÎj B n N-(3-fluoro-4-((3-((4hydroxy-2-methylbutan-2yl)amino)-IHpyrazolo[3,4-b]pyridin-4yl)oxy)phenyî)-2-(4fluorophenyl)-3-oxo-2,3dihydropyridazine-4carboxamide 562.2 (M+H)
34 h o ηοΛ/ T Y Y Y Y | /-NH ° F nYj H N N-(3-fluoro-4-((3-((3hydroxy-3-methylbutan-2yt)amino)-lHpyrazolo[3,4-b]pyridin-4yî)oxy)phenyl)-2-(4fluorophenyl)-3 -oxo-2,3dihydropyridazine-4carboxamide 562.1 (M+H)
35 Z-Z^^ C /^° )=o IZ Y- LL O—V Z N Y, ZI O X N-(3-fluoro-4-((3-((2hy droxyethy l)(methy l)am î no)-lH-pyrazolo[3,4b]pyridin-4yI)oxy)phenyl)-2-(4fluoropheny])-3-oxo-2,3dihydropyridazine-4carboxamide 534.1 (M+H)
271
36 H Γ0 H0\ Tï Y0r Ύ0 'j-<A ο o LAr V^NH ? F 0] fi n N-(3-fluoro-4-((3(((lr,3r)-3(hydroxymethyl)cyclobuty I)amino)-1Hpyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-2-(4fluorophenyl)-3-oxo-2,3dihydropyridazine-4carboxamide 560.1 (M+H)
37 h 1 / ΥΎ ni 10 ^JX ° 0 \Ar \/—NH 0 F »15 B N N-(3-fluoro-4-((3- (((Is,3s)-3(hydroxymethyl)cyclobuty I)amino)-1Hpyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-2-(4tluoropheny l)-3 -oxo-2,3 d îhy dropy ridazi ne-4carboxam ide 560.1 (M+H)
38 0 1 z-z 0° 0° IL O—F Z ^000 07^ Z O I N-(3-fluoro-4-((3-((3hy droxy-2-methy 1 butan-2yl)amino)-l Hpyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-2-(4fluoropheny l)-3 -oxo-2.3 dihydropyridazine-4carboxamide 562.2 (M+H)
272
39 Η Ηο-γ T Y Y Y y i YnH O'M ° 0 s N N-(3-fluoro-4-((3-((3hydroxy-2,3dimethylbutan-2yl)amino)-l Hpyrazolo[3,4-b]pyridin-4yI)oxy)pbeny])-2-(4fluorophenyI)-3-oxo-2,3dihydropyridazine-4carboxamide 576.2 (M+H)
40 HO N γ^γ- N y^ ΥΆ -AJ 0 0 Λ]Μ H N N-(3-fluoro-4-((3-((4hydroxy-2-methylpentan2-yl)amino)-lHpyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-2-(4fluoropheny])-3-oxo-2,3dihydropyridazine-4carboxamide 576.2 (M+H)
41 H Π HO% T T Y Y II 1 \ nY/' 0 0 /'NH 9 F kHo H N (R)-N-(3-fluoro-4-((3-((lhydroxypropan-2y[)amino)-l Hpy razo lo[3,4-b] pyridin-4yl)oxy)phenyl)-2-(4fluorophenyl)-6-methyl-3oxo-2,3dihydropyridazine-4carboxamide 548.2 (M+H)
273
42 Η ί| ? N N HO-~, T ίΤ Π Π H 1 NH O O H N-(3-fiuoro-4-((3-((lhydroxy-2-methylpropan2-yl)amino)-lHpy razo lo[3,4-b]pyridin-4yl)oxy)phenyl)-2-(4fluorophenyl)-6-methyl-3oxo-2,3dihydropyridazine-4carboxamide 562.2 (M+H)
43 I o^ “N Q ZI o=/ z-z 0 m N-(3-fluoro-4-((3-((4hydroxy-1 -methoxy-2methylbutan-2-yl)amino)lH-pyrazolo[3,4b]pyrtdin-4yl)oxy)pheny 1)-2-(4fluorophenyl)-3-oxo-2,3dihydropyridazine-4carboxamide 592.2 (M+H)
44 H O oh VynyVy^ f3c^ T II n T 1 VNH O^ ° ° ^F n^Tj H “ (S)-N-(3-fluoro-4-((3((3,3,3-trifluoro-2hydroxypropyl)amino)lH-pyrazolo[3,4b]pyridin-4yl)oxy)phenyl)-2-(4fluorophenyl)-3-oxo-2,3dihydropyridazine-4carboxamide 588.1 (M+H)
274
45 LL· 0 z-z 0 )=o Yo IZ 0 LL Ο—ΐ7 Z Λ Y 2 S g N-(4-((3-((l,3- d îhyd roxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridîn-4yl)oxy)-3-fluorophenyl)2-(4-fluorophenyl)-3-oxo2,3-dihydropyridazine-4carboxamide 550.2 (M+H)
46 b Yr^^n \ aAJ o o YY. ηο^Λ™ ? F ΛΑ H N N-(3-fluoro-4-((3-((lhydroxy-3methoxypropan-2yl)amino)-lHpy razo lo [ 3,4-b]pyrid in-4yl)oxy)pheny 1)-2-(4fluorophenyl)-3-oxo-2,3dihydropyridazine-4carboxamide 554.1 (M+H)
47 LL· 0 z-z 0 Y° Y -° i T N-(3-fluoro-4-((3-((lhydroxy-3-methoxy-2methylpropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-2-(4fluorophenyl)-3 -oxo-2,3dihydropyridazine-4carboxamide 578.2 (M+H)
275
48 X 0‘ T ZT o=( z-z 0 N-(3-fluoro-4-((3-((4hydroxy-1 -methoxybutan2-y])amino)-lHpy razo lo [3,4-b] py rid i n-4yl)oxy)phenyl)-2-(4fluoropheny l)-3 -oxo-2,3 dihydropyridazine-4carboxamide 578.2 (M+H)
49 LL 0 z-z 0 )=o 2=0 xz 0 U- 0—0 Z / Z XO^ O 1 N-(3-fluoro-4-((3-((1hydroxy-4-methoxybutan2-yl)amino)-lHpyrazolo[3s4-b]pyridin-4yl)oxy)phenyl)-2-(4fluorophenyl)-3-oxo-2,3dihydropyridazine-4carboxamide 578.2 (M+H)
50 T <° Z IZzi H ) z-z -Π N-(4-((3-((2,2-difluoro-3hydroxypropyl)amino)lH-pyrazolo[3,4b]pyridin-4-yl)oxy)-3fluoropheny 1)-2-(4fluorophenyl)-3 -oxo-2,3dihydropyridazine-4carboxamide 570.2 (M+H)
51 X -Π o^y-^ 00 0 zx o=z 0’ N-(4-((3-((4,4-difluoro-lhydroxybutan-2yl)amino)-lHpyrazolo[3,4-b]pyrîdin-4yl)oxy)-3-fluorophenyl)2-(4-fluorophenyl)-3-oxo2,3-dihydropyridazine-4- 584.2 (M+H)
276
carboxamide
52 H FsCa XJ I S XX η°^Λνη ? F nXj 9 N N-(3-fluoro-4-((3-((4,4,4trifluoro-l-hydroxybutan2-yl)amino)-lHpyrazoIo[3,4-b]pyrîdin-4y l)oxy)pheny 1)-2-(4fluorophenyl)-3-oxo-2,3dihydropyridazine-4carboxamide 602.2 (M+H)
53 h X^ hoy T T ¥ ¥ T ] ° ° ¥0 H N N-(3-fluoro-4-((3-((l(hydroxymethyl)cyclobuty l)amino)-l Hpyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-2-(4fl uoropheny l)-3-oxo-2,3dihydropyridazine-4carboxatnide 560.2 (M+H)
54 z Λ—ο -Π ZI ο=ζ °x z-z N-(3-fluoro-4-((3(((lS,2R)-2(hydroxymethyl)-1 methylcyclopentyl)amino) -lH-pyrazolo[3,4b]pyridin-4yl)oxy)phenyl)-2-(4fl uoropheny 1)-3-0 xo-2,3dihydropyrîdazine-4carboxamide 588.2 (M+H)
277
55 V ZI o=( z-z H (R)-N-(3-fluoro-4-((3 -(( 1 hydroxybutan-2yl)amino)-l Hpyrazolo[3,4-b]pyridin-4yl)oxy)pheny 1)-2-(4fl uoropheny l)-3 -oxo-2,3 dihydropyridazine-4carboxamide 548.2 (M+H)
56 LL Z-Z ? ^=0 )=o IZ Λ Z \ J· X (S)-N-(3-fluoro-4-((3-((lhydroxybutan-2yl)amino)-lHpy razo lo [3,4-b] pyrid in-4 yl)oxy)phenyl)-2-(4fluorophenyl)-3-oxo-2,3dihydropyridazine-4carboxamide 548.1 (M+H)
Example 57
[001250 ] N-(3-fliioro-4-((3-(( l-(hydroxvmethyl)cyclopropvl)am!no)-lH-pvrazolo[3.4b1pvridin-4-yl)oxy)phenyl)-2-(4-fluorophenvl')-3-oxo-213-dihvdropvridazine-4-carboxamid
[001251]
[001252]
Step A: N-(3-fluoro-4-((3-iodo-l-(4-methoxybenzyl)-l H-pyrazolo[3,4-b]pyridin
4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide (Préparation 63; 150 mg, 0.212 mmol), (l-aminocyclopropyl)tnethanol hydrochloride (26.2 mg, 0.212 mmol), lH-pyrrole-2-carboxylic acid (11.8 mg, 0.106 mmol), and K2CO3 (235 mg, 1.70 mmol) were
278 suspended in DMSO (2 mL) and nitrogen bubbled through the mixture for 5 min, Copper(I) iodide (20.2 mg, 0.106 mmol) was added and the reaction mixture was heated to 60 °C overnight. The reaction mixture was cooled to room température, diluted with water (10 mL), stirred for 10 min, and then filtered. The isolated solids were suspended in DCM/MeOH, dried with sodium sulfate, filtered through Celite® and the filtrate was concentrated. The residue was purified over silica gel (10-100% EtOAc in hex) to afford N-(3-fluoro-4-((3-((l-(hydroxymethyl)cyclopropyl)amino)-l(4-methoxybenzyi)-lH-pyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3dihydropyridazine-4-carboxamide (75 mg, 0.113 mmol, 53.1 % yield) as a yellow solid.
[001253] Step B: N-(3-fluoro-4-((3-((l-(hydroxymethyl)cyclopropyl)amino)-l-(4methoxybenzyl)-lH-pyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3dihydropyridazine-4-carboxamide (75 mg, 0.11 mmol) was dissolved in DCM (0.5 mL) and TFA (0.5 mL). The reaction mixture was heated to 50 °C overnight and then concentrated. The residue was dissolved in MeOH and excess was K.2CO3 added. The mixture was stirred for 10 min, then partit ioned between water and DCM. The organic layer was dried over sodium sulfate, filtered and concentrated. The residue was purified over silica gel (0 to 10% MeOH in DCM) to afford N-(3fluoro-4-((3-((l-(hydroxymethyl)cyclopropyl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-2-(4-fluorophenyI)-3-oxo-2,3~dihydropyridazine-4-carboxamide (33 mg, 0.060 mmol, 54 % yield) as an orange solid. Mass spectrum: m/z = 546.2 (M+H). 'H NMR (dô-DMSO) Ô 12.22 (s, IH), 11.70 (s, 1H), 8.38 (d, 1H), 8.26 (d, 1H), 8.14 (d, 1H), 8.03 (dd, 1H), 7.68 (m, 2H), 7.59 (m, IH), 7.48 (t, 1H), 7.41 (m, 2H), 6.03 (m, IH), 5.76 (s, 1H), 3.59 (s, 2H), 0.75 (m, 4H).
279
[001254] The following compounds were also synthesized using the procedure according to Example 57.
Ex. No. Structure Naine Mass spectrum (apci) m/z
58 z ^=O )=O zz u. O—v z 1 -Γ / ' N-(3-fluoro-4-((3-((cis2(hydroxymethyl)cyclop entyl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-2-(4fl uorophenyl)-3-oxo2,3-dihydropyridazîne4-carboxamide 574.2 (M+H)
59 u_ z-z )=° U- O~+ Z ï ZX O-M z T r-A N-(3-fluoro-4-((3-((l(hydroxymethyl)cyclop entyl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)pbeny 1)-2-(4fluorophenyl)-3-oxo2,3-dihydropyridazine4-carboxamide 574.2 (M+H)
60 H ho^. Y T Y Y Y ] \ A+ ο o >LA ,,Υ-νη 9 F Cy1 n^1îj '[Ύγ H N (R)-N-(3-fluoro-4-((3((2-hydroxy-lphenylethyl)amino)lH-pyrazolo[3,4b]pyridin-4yl)oxy)phenyl)-2-(4fluorophenyl)-3-oxo- 596.2 (M+H)
280
2,3-dihydropyridazine4-carboxamide
61 η X* ho^ \ 1 II Π Π II ] V „ΑΑ ο ο <./,r ηο^/Νη ? f ΛΑ Αν N-(3-fluoro-4-((3-((lhydroxy-2(hydroxymethyl)butan2-yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)pheny 1)-2-(4fluorophenyl)-3-oxo2,3-dihydropyridazîne4-carboxamide 578.2 (M+H)
62 .ζ Γ \ VA J Ζ 4 ΤΖ VÎ Q zi oV 00 z-z 0 Tl N-(3-fluoro-4-((3-((2hydroxy-l-(pyridin-3y l)ethy l)am ino)-1Hpyrazolo[3,4-b]pyrîdin4-yl)oxy)pheny 1)-2-(4nuorophenyl)-3-oxo2,3-dihydropyridazine4-carboxamide 597.2 (M+H)
63 h A* HCv I T Y A Y Ί Anh Λ ° ° 0A 0 N00 A N-(3-fluoro-4-((3-((2hydroxy-1 -(pyridin-4yl)ethyl)amino)-lHpyrazolo[3,4-b]pyrîdin4-yl)oxy)phenyl)-2-(4f1uorophenyl)-3-oxo2,3-dihydropyridazine4-carboxamide 597.2 (M+H)
281
64 Η HO^ T ï ï ï ï 1 NH ? F C) <O H IN N-(3-fluoro-4-((3-((2hydroxy-1 -(tetrahydro2H-pyran-4yi)ethyl)amino)-1Hpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-2-(4fluorophenyl)-3-oxo2,3’dihydropyridazine4-carboxamide 604.2 (M+H)
65 H ίΊΤ ho^ T II π n ji Ί /NH γ H N (R)-N-(3-fluoro-4-((3(( 1-hydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyndin4-yl)oxy)phenyl)-2,5dioxo-l'phenyl1,2,5,6,7,8hexahydroquînoline-3carboxamide 583.2 (M+H)
66 LL ( )—Z M O 0=< /=° IZ / C U- Ο-c Z - Z cC^ T N-(3-fluoro-4-((3-iodo1 -(4-methoxybenzyl)- 1 H-pyrazolo[3,4b]pyridin-4yl)oxy)pheny 1)-1 -(4fluorophenyl)-2,5dioxo-1,2,5,6,7,8hexahydroquinoline-3carboxamide 601.2 (M+H)
282
67 / υυβτ9π ° ° nA,. Λνη θ F ν^Π H (R)-N-(3-fluoro-4-((3(( 1 -hydroxy butan-2yl)amino)-lHpyrazolo[3,4-b]pyridîn4-yl)oxy)phenyl)-3-(4fluorophenyl)-lisopropyi-2,4-dioxo1,2,3,4tetrahydropy rim id ine5-carboxamide 606.2 (M+H)
68 o-a I >--x z - + Â As ZI ο=ζ Op Z—( /=< O “Π (S)-N-(3-fluoro-4-((3- (( 1 -hydroxybutan-2yl)amino)-i Hpyrazolo[3,4-b]pyridin4-yl)oxy)pheny 1)-3-(4fluorophenyl)-!isopropyl-2,4-dioxo1,2,3,4tetrahydropyrimidine5-carboxamide 606.2 (M+H)
69 y-z /=o )=o IZ O /wa O I N-(3-fluoro-4-((3-((l(hydroxymethyl)cyclob utyl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-3-(4fluorophenyl)-!isopropyl-2,4-dioxo1,2,3,4tetrahydropyrimidine5-carboxamide 618.2 (M+H)
283
70 I O z Vi z 4—0 -n ZI O=/ O z—( /=( O w T? N-(3-fluoro-4-((3-((l- hydroxy-2methylpropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-3-(4fluorophenyl)-!isopropyl-2,4-dioxo1,2,3,4tetrahydropy rim îd ine5-carboxamide 606.2 (M+H)
Example 71
[001255] 2-f4-fluorophenyl)-N-(5-((3-(f 1-methoxy-2-methvlpropan-2-yl)amino)-lHPVrazolor3.4-b1pvridin-4-vl)oxy)pvridin-2-vl)-3-oxo-2,3-dihvdropyridazine-4-carboxamide
[001256]
[001257]
To a mixture of 4-((6-aminopyridin-3-yl)oxy)-N-(l-methoxy-2-methylpropan-2 yl)-l-(4-methoxybenzyl)-lH-pyrazolo[3,4-b]pyridin-3-amine (Préparation 52; 0.033 g, 0.074 mmol), 2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxylic acid (0.034 g, 0.15 mmol), EDC1 (0.085 g, 0.44 mmol) and HOBt (0.060 g, 0.44 mmol) was added DMF (0.7 mL), and the reaction mixture was stirred for 2 h. The reaction mixture was diluted with water, stirred for 30 min and then filtered. The isolated solids were washed with water and hexanes. The solids were dissolved in TFA (2 mL) and heated to 50 °C overnight. The reaction mixture was concentrated and purified by Cl8 chromatography (5-95% ACN in water with 0.2% TFA). The fractions containing the product were free-based with saturated NaHCOs (30 mL) and the aqueous phase
284 was extracted with DCM (2 X 15 inL). The combined organic layers were washed with saturated NaHCOj ( 15 mL) and brine ( 15 mL), dried over sodium sulfate, filtered and concentrated to afford 2-(4-fluorophenyl)-N-(5-((3-(( l-methoxy-2-methylpropan-2-yl)amino)-lH-pyrazolo[3,4b]pyridin-4-y])oxy)pyridin-2-yl)-3-oxo-2,3-dihydropyridazine-4-carboxamide (0,0020 g, 0.0037 mmol, 5.0 % yield). Mass spectrum: m/z = 545.2 (M+H), Ή NMR (CDCh) δ 12.20 (s, 1H), 8.47 (d, 1H), 8.40 (d, IH), 8.33 (d, 1H), 8.22 (d, IH), 8.17 (m, 1H), 7.67-7.60 (m, 3H), 7.22 (m, 2H), 6.10 (d, IH), 5.02 (br s, IH), 3.52 (s, 2H), 3.36 (s, 3H), 1.47 (s, 6H).
[001258] Example 72
[001259] (R)-N-(3-fluoro-4-i(3-(( 1 -hvdroxvpropan-2-vl)amino)- 1 H-pyrazolor3,4blpyrÎdtn-4-yl)oxv)phenylL3-(4-fluorophenvl)-l-isopropvl-2,4-dioxo-L2,3,4tetrahy dropyrim id ine- 5 -carboxam i de
[001260]
[001261] Step A: N-(3-fluoro-4-((3-iodo-l-(4-methoxybenzyl)-lH-pyrazoio[3,4-b]pyridin4-yl)oxy)phenyl)-3-(4-fluorophenyl)-l-isopropyl-2,4-dioxo-l,2,3,4-tetrahydropyrimidine-5carboxamide (Préparation 65; 200 mg, 0.26 mmol), D-Alaninol (61 pL, 0.785 mmol), K2CO3 (289 mg, 2.09 mmol) and pyrrole-2-carboxylic acid (0.0145 g, 0.131 mmol) were suspended in DMSO (5 mL) and the mixture was degassed for 5 min with Ar. Cu(I)iodide was added and the mixture was heated to 60 °C in a sealed tube overnight and then cooled to room température. The cooled mixture was partitioned between water (10 mL) and EtOAc (10 mL) and the aqueous layer was extracted with EtOAc (2x10 mL). The combined organic phases were washed with water (5 x 10 mL) and brine (10 mL), then dried over Na2SO4, filtered and concentrated. The residue was purified oversilica gel (0-5% MeOH in DCM) to afford (R)-N-(3-fluoro-4-((3-((l-hydroxypropan2-y l)amino)-l-(4-methoxybenzyl)-lH-pyrazolo[3,4-b]pyridin-4-yl)oxy)pheny 1)-3-(4fIuorophenyl)-l-isopropyl-2,4-dioxo-I,2,3,4-tetrahydropyrimidine-5-carboxamide (98 mg, 53%) as a white solid.
285
[001262] Step B: To a solution of (R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-l(4-methoxybenzyl)-lH-pyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-3-(4-fluorophenyl)-l-isopropyl2,4-dioxo-l,2,3,4-tetrahydropyrimidine-5-carboxamide (98 mg, 0.14 mmol) in DCM (2 mL) was added TFA (4 mL). The mixture was stirred at 35 °C overnight. The mixture was concentrated and 5 dried in vacuo. The residue was dissolved in MeOH/DCM, treated with K2CO3 (57 mg, 0.4] mmol) and stirred at RT for 4 h. The mixture was filtered and concentrated. The residue was purified over silica gel (0-7.5% MeOH in DCM) to afford (R)-N-(3-fkioro-4-((3-((lhydroxypropan-2-yl)amino)-lH-pyrazoIo[3,4-b]pyridin-4-yi)oxy)phenyl)-3-(4-fluorophenyl)-lisopropyl-2,4-dioxo-l,2,3,4-tetrahydropyrimidine-5-carboxamide (68 mg, 83%) as ayellow solid.
Mass spectrum: m/z= 592.2 (M+H). !H NMR (CDCh) δ 1 1.00 (s, 1H), 8.69 (s, lH),8.16(d, IH), 7.90 (dd, 1H), 7.30 (m, 1H), 7.27 (s, 211), 7.25 (s, 2H), 7.22 (d, 1H), 6.12 (d, IH), 4.98 (m, 1H), 4.02 (m, IH), 3.83 (dd, 1H), 3.71 (dd, 1H), 1.51 (d, 6H), 1.34 (d, 3H).
[001263] The following compounds were also synthesized using the procedure according to Example 72.
Ex. No. Structure Naine Mass Spectrum
73 . 0 y—Z Yo y=o IZ X LL 0—0 Z 0 T (R)-N-(3-fluoro-4-((3((l-hydroxypropan-2yl)(methyl)amino)-lHpyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-3-(4fluorophenyl)-!isopropyl-2,4-dioxo1,2,3,4tetrahydropyrimidine-5carboxamide 606.2 (M+H)
286
74 Ο Ο )=7 νΚ 2—ζ >=ο 2=° ΙΖ 0 Λ, ο I (S)-N-(3-fluoro-4-((3(( 1 -hydroxypropan-2yl)(methyi)amino)-lHpyrazolo[3,4-b]pyridin-4y l)oxy)pheny 1)-3-(4fluorophenyl)-!isopropyl-2,4-dioxo1,2,3,4tetrahydropyrimidine-5carboxamide 606.2 (M+H)
75 LL ο )=^ 5—Ζ 2=0 2=ο ΙΖ o^j (R)-N-(3-fluoro-4-((3- ((2hydroxypropyl)amino)lH-pyrazolo[3,4b]pyridin-4yl)oxy)pheny 1)-3-(4fluorophenyl)-!isopropyl-2,4-dioxo1,2,3,4tetrahydropyrimidine-5carboxamide 592,2 (M+H)
76 LL Ο Ο )=/ \ Fz, 5— Ζ 2=0 2=ο ΙΖ ζχΐζ O^J ζ (S)-N-(3-fluoro-4-((3- ((2hydroxypropyl)amino)lH-pyrazolo[3,4b]pyrîdin-4yl)oxy)phenyl)-3-(4fluorophenyl)-!isopropyl-2,4-dioxo1,2,3,4tetrahydropyrimidine-5- 592.2 (M+H)
287
carboxamide
77 -, Γό Ζ τ τζ yi ζ 0—0 -π W Q ΖΤ 1 °Ρ 0 πι N-(3-fluoro-4-((3-((2hydroxy-2methylpropyl)amino)IH-pyrazolo[3,4b]pyridin-4yl)oxy)pheny 1)-3-(4fluorophenyl)-!isopropyl-2,4-dioxo1,2,3,4tetrahydropyrimidine-5carboxamide 606.2 (M+H)
78 λα ο >=^ \ Τζ )—ζ >ο )=° τζ 0 ΛΑ #Α, ll 0—0 Ζ ^Α ,ζτ τ N-(3-fluoro-4-((3(((2R,3S)-3hydroxybutan-2yl)amino)-lHpyrazolo[3,4-b]pyridin-4y l)oxy)pheny 1)-3-(4fluorophenyl)-! isopropyi-2,4-dioxo1,2,3,4tetrahydropyrimidine-5carboxamide 606.2 (M+H)
79 Ll ο // λ2ν>° )=ο τζ 0 °Ρ ό τ N-(3-fluoro-4-((3(((2R,3R)-3hydroxybutan-2yl)amino)-lHpyrazolo[3,4-b]pyridin-4y l)oxy)pheny 1)-3-(4fluorophenyl)-! isopropyl-2,4-dioxo- 606.2 (M+H)
288
1,2,3,4- tetrahy dropy ri m id i ne-5 carboxamide
80 HOY XJ s s Λ /NH θ F H (R)-N-(3-fluoro-4-((3((3-hydroxy-3methylbutan-2yl)amino)-lHpyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-3-(4fluorophenyl)-!isopropyl-2,4-dioxo1,2,3,4tetrahydropyrimidine-5carboxamide 620.2 (M+H)
81 I o Q zi Z—/ JM \ /=\ ° w n (S)-N-(3-fluoro-4-((3- ((3-hydroxy-3methylbutan-2yl)amino)-lHpy razo lo [3,4-b] pyrid in-4yl)oxy)pheny 1)-3-(4fluoropheny!)-!isopropyl-2,4-dioxo1,2,3,4tetrahydropyrimidine-5carboxamide 620.2 (M+H)
Example 82
[001264] (R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)atnmo)-lH-pyrazolor3,4b1pYridin-4-yl)oxy)phenvl)-3-(4-fluorophenyl')-l-methyl-2.4-dioxo-l.2.3.45 tetrahydropyrimidine-5-carboxamide hydrochloride
289
[001265]
[001266]
Step A: To a stirred solution of 3-(4-fluorophenyl)-l-methyl-2,4-dioxo-1,2,3,4 tetrahydropyrimidine-5-carboxylic acid (66.4 mg, 0.251 mmol) and HATU (0.1304 g, 0.3429 mmol) în DMF (3 mL) at room température was added DIEA (119 pL, 0.686 mmol) followed by (R)-2-((4-(4-aminO“2-fluorophenoxy)-l -(4-methoxybenzyl)-lH-pyrazolo[3,4-b]pyridin-3yl)amino)propan-l-ol (Préparation 53; 100 mg, 0.229 mmol) and stirred overnight, The reaction mixture was partîtioned between water (20 mL) and EtOAc (20 mL) and the aqueous layer was extracted with EtOAc (2x10 mL). The combined organîc phases were washed with water (5x10 mL) and brine (10 mL), dried over Na2SÜ4, fîltered and concentrated. The residue was purifîed over silica gel (0-5% MeOH in DCM) to afford (R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2yl)amino)-I-(4-methoxybenzyl)-lH-pyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-3-(4fluorophenyl)-l-methyl-2,4-dioxo-l,2,3,4-tetrahydropyrimidine-5-carboxamîde (123 mg, 79%). [001267] Step B: To a solution of (R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-l(4-methoxybenzyl)-lH-pyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-3-(4-fluorophenyl)-l-methy[2,4-dioxo-l,2,3,4-tetrahydropyrimidine-5-carboxamide (123.0 mg, 0.180 mmol) in DCM (J mL) was added TFA (2 mL). The mixture was stirred at 35 °C for 7 h and then cooled to room température overnight. The mixture was concentrated and the residue dissolved in MeOH/DCM, treated with K2CO3 (74.6 mg, 0.540 mmol) and stirred for 5.5 h. The mixture was fîltered and concentrated, and the residue was and purifîed over silica gel (0-5% MeOH in DCM). The purifîed compound was dissolved in DCM (5 mL), treated with 4N HCl/dioxanes (0.5 mL), concentrated and dried in vacuo to afford (R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lH pyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-3-(4-fluorophenyl)-l-methyl-2,4-dioxo-l,2,3,4tetrahydropyrimidine-5-carboxamide hydrochloride (20.3 mg, 18%) as a yellow solid. Mass spectrum: m/z = 564.2 (M+H-HC1). 'HNMR(CDCh)6 11.05 (s, 1 H), 8.67 (s, IH), 8.26 (d, 1H), 7.95 (dd, 1H), 7.36-7.22 (m, 6H), 6.22 (d, 1H), 4.02 (m, 1H), 3.76 (m, 1H), 3.71 (s, 3H), 3.65 (m, 1H), 1.35 (d, 3H).
290
[001268] The following compounds were also synthesized using the procedure according to Example 82.
Ex. No. Structure Name Mass Spectrum
83 ^N^.0 ηολ nXJ ο ° XX / J'NH 0 F N^XfS n'n H (R)-l-ethyl-N-(3-fluoro4-((3-((1hydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-3-(4fluoropheny 1)-2,4dioxo-1,2,3,4tetrahydropyrimidine-5carboxamide 578.2 (M+H)
84 ho-. T II x x 11 1 \ «XsX 0 0 Z γΖρ. /NH θ F H (R)-l(cyclopropylmethyl)-N(3-fluoro-4-((3-((lhydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)pheny 1)-3-(4fluorophenyl)-2,4dioxo-1,2,3,4tetrahy d ropyrimidine-5 carboxamide 604.2 (M+H)
291
85 h°^ T ΤΓ π π η η Y Y ° ° Y n^xS NAr H (R)-4-ethoxy-N-(3fluoro-4-((3-((lhydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyrtdin4-yl)oxy)phenyl)-l-(4fluorophenyl)-2-oxol,2-dihydropyridîne-3carboxamide 577.2 (M+H)
86 A-n^ ,__ x^n/A-O HO—, T T Π Y \ AY1 ° 0 / NH 0 n/A H (R)-N-(3-fluoro-4-((3(( 1 -hydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yi)oxy)phenyl)-1,5dimethyl-3-oxo-2phenyl-2,3-dihydro-lHpyrazole-4-carboxamide 532.2 (M+H)
87 / r-N z— HO-. T Y Y \ ° ° /NH θ nYj NS/ H (R)-N-(3-fluoro-4-((3((l-hydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-y l)oxy)phenyl)-l methy 1-3 -oxo-2-pheny 12,3-dihydro-IHpyrazole-4-carboxamide 518.2 (M+H)
88 / z— ηολ \ ^AJ O 0 /NH θ nM N 'nx H (R)-N-(3-fluoro-4-((3- (( l-hydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yI)oxy)phenyl)-2-(4fluorophenyl)-! -methyl- 536.2 (M+H)
292
3-oxo-2,3-dihydro-lHpyrazole-4-carboxamide
89 I Z 0—0 Tl T ZI o=( o=< z^C z— 0 ° Π (R)-N-(3-fluoro-4-((3(( 1 -hydroxypropan -2yl)amino)-lHpyrazolo[3,4-b]pyridin4-y!)oxy)phenyl)-3-(4fluorophenyl)-l-(lmethylazetidin-3-yl)2,4-dioxo-ls2,3s4tetrahydropy ri m td i ne-5 carboxamide 619.2 (M+H)
90 I O VV Q ZI θ4 o=( Z—/ 0 ° __________ (R)-3-cyclopentyl-N-(3fluoro-4-((3-((lhydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)phenyi)-1 isopropy 1-2,4-d ioxo1,2,3,4tetrahydropyri midi ne-5 carboxamide 566.2 (M+H)
91 I ΖΖΟ^ Z 0—0 Tl ^0 ZI O=Z o=( z—<Q> 7¼ /=/ O T| (R)-l-cyclobutyl-N-(3fluoro-4-((3-((lhydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-3-(4fluorophenyl)-2,4dioxo-1,2,3,4tetrahydropyrimid i ne-5 - 604.2 (M+H)
293
carboxamide
92 H°^ T Π π π η T >NH ° ° ^F H (R)-3-(3,4- dîfluoropheny!)-N-(3fhioro-4-((3-((lhydroxypropan-2- yl)amino)-lH- pyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-lisopropyl-2,4-dioxo1,2,3,4- tetrahydropyrim id ine-5 carboxamide 610,2 (M+H)
93 Z O z / y-i \-O u X ZI o=\ < Π · z 1 (R)-N-(3-f1uoro-4-((3(( 1 -hydroxypropan-2yl)amino)-l Hpyrazolo[3,4-b]pyridin4-y l)oxy)phenyl)-1 isopropyl-3-(l -methyllH-pyrazol-4-yl)-2,4dioxo-1,2,3,4tetrahy d ropy rimidine-5 carboxamide 578.2 (M+H)
94 N VL, N v nXJ I s Ta / NH 9 F nTîj Ν'Ά' H (R)-N-(3-fluoro-4-((3- (( 1 -hydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyrîdin4-yl)oxy)phenyl)-3-(4fluorophenyl)-2,4d ioxo-1 -(pentan-3 -yl)1,2,3,4tetrahydropyrimidine-5- 620.2 (M+H)
294
carboxamide
95 ΗΟ^ Y T Y T T >NH 0^ ° ° ’ n^Xj H (R)-N-(3-fluoro-4-((3- (( 1-hydroxypropan-2- yl)amino)-1H- pyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-l,3diisopropyl-2,4-dioxo1,2,3,4- tetrahydropy ri m îd i ne-5 carboxamide 540.2 (M+H)
96 jCL^o V+Vv'/', }-NH OJU ° ° nYj H (R)-3 -cy c lohexy l-N -(3 fluoro-4-((3-((lhydroxypropan-2yl)amino)-IH- pyrazolo[3,4-b]pyrîdin4-yl)oxy)phenyl)-1 isopropyl-2,4-dioxo1,2,3,4- tetrahydropyrimidine-5carboxamide 580.3 (M+H)
97 X O Z + irZ /)—Ο “Π Y zz o=/ oY Z-/ /Μλ γ ° g (R)-3-(4-chlorophenyl)N-(3-fluoro-4-((3-((lhydroxypropan-2yl)amino)-IHpyrazoio[3,4-b]pyridin4-yl)oxy)phenyl)-Iisopropyl-2,4-dioxo1,2,3,4tetrahydropyrimidîne-5carboxamide 608.2 (M+H)
295
98 LL ο 0 \ U2. λζ\=>° /=o IZ O UY O X (S)-N-(3-fluoro-4-((3- (( 1-hyd roxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridtn4-y l)oxy)pheny 1)-3-(4fluorophenyl)-!isopropyl-2,4-dioxo1,2,3,4- tetrahydropyrimidine-5carboxamîde 592.2 (M+H)
99 I ζζ/νί zx Y° 7 Y zi o=/ oY Z Y /ZYÎ 0 ° o—7 (R)-N-(3-fluoro-4-((3- (( 1 -hydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-lisopropyl-2,4-dioxo-3(tetrahydro-2H-pyran-4- yl)-U2,3,4tetrahydropyrimidine-5carboxamide 582.2 (M+H)
100 H if^V N - YY N H0% T II ” π II 1 \ «AJ o o / NH θ F Ύ5 N^Y H (R)-N-(3-fluoro-4-((3(( 1 -hydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-l-(4fl uoropheny 1) -6~methy 12-oxo-1,2d ihydropyridine-3carboxamide 547.2 (M+H)
296
101 ηολ XJ Π XX /NH y F »15 H N-(3-fluoro-4-((3-(((R)1 -hydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-l-(4fluorophenyl)-2oxopiperidine-3carboxamide 537.3 (M+H)
102 H N 'süx N ho-, T II n Y Y | \ θ o XJ. /'NH 9 F N^0fj Ν-Ί^ H (R)-N-(3-fluoro-4-((3- (( 1-hydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-l-(4fluorophenyl)-4-methyl2-oxo-1,2dihydropyridine-3carboxamide 547.2 (M+H)
103 LL 0 -£>o 0O IZ 0 / \ U. O—C Z ^00z X c0x X (R)-N-(3-fluoro-4-((3(( 1 -hy droxy propan-2 yl)amino)-lHpyrazoloPJ-bJpyridin4-yl)oxy)phenyl)-l-(4fluorophenyl)-5-methyl2-oxo-1,2dihydropyridine-3carboxamide 547.2 (M+H)
297
104 I Ο zY°y_7 0 (R)-N-(3-fluoro-4-((3- (( 1 -hy d roxy p ropan-2yl)amino)-lHpyrazolo[3,4-b]pyridîn4-yl)oxy)phenyl)-4-(4fluorophenyl)-3-oxo3,4-dihydropyrazine-2carboxamide 534.2 (M+H)
105 I 0* Y ZI °γ oY^Y z-z 0 (R)-6-cyc lo propy 1 -N-(3 fluoro-4-((3-((lhydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyrîdin4-yl)oxy)phenyl)-2-(4fluorophenyl)-3-oxo2,3-dihydropyridazine4-carboxamide 574.2 (M+H)
106 I o Y z^”°^ zx °γ z-z x n (R)-N-(3-fluoro-4-((3(( 1 -hydroxypropan-2yl)amino)-lHpyrazolo[3f4-b]pyridin4-yl)oxy)pheny 1)-2-(4fluorophenyl)-6isopropyl-3-oxo-2,3dihydropyridazine-4carboxamide 576.2 (M+H)
298
107 H 'V N J yX ho-, T lT π π η η ) 'nh ° ° p Nho H (R)-N-(3-fluoro-4-((3((l-hydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)pheny 1)-4-(4fl uoropheny l)-2isopropyl-5-oxo-3thioxo-2,3,4,5tetrahydro-1,2,4triazine-6-carboxam ide 609.2 (M+H)
108 I O I z Y-o 71 zz 0=^ o¥\Y 7+ ' /=\ 0 w (R)-N-(3-fluoro-4-((3- (( 1-hydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-4-(4fluorophenyl)-2isopropyl-3,5-dioxo2,3,4,5-tetrahydro-1,2,4triazine-6-carboxamide 593.2 (M+H)
109 Z 0 ¥ zz 0=0 ° w ' 0 ° “Π (S)-N-(3-fluoro-4-((3- (( 1 -hydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)pheny 1)-4-(4fiuorophenyl)-2isopropyl-3,5-dioxo2,3,4,5-tetrahydro-1,2,4triazine-6-carboxamide 593.2 (M+H)
299
110 Y Ν^,Ο ηολ XJ SS XX /NH 9 F ΝΆ^ H (R)-l-cyclopropyi-N-(3fluoro-4-((3-((lhydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yI)oxy)pheny 1)-3-(4fluorophenyl)-2,4dioxo-1,2,3,4tetrahydropy ri m i d ine-5 carboxamide 590.2 (M+H)
111 O;W h il Ά F'W^N -AAN -v, ho^ Y Y Y Y Y | \ ΛΑ-Α 0 0 N',+V /NH 9 F «A H N (R)-N-(3-fluoro-4-((3- (( 1 -hydroxypropan-2yl)amino)-l Hpyrazolo[3f4-b]pyridin4-yl)oxy)phenyl)-1-(5fl uoropyridin-2-y 1)-2,5 dioxo-1,2,5,6,7,8- hexahydroquinoline-3carboxamide 602.2 (M+H)
112 1 N h ii il W-N Av'vA ho—, \ Y Y Y Y | >,NH VN ο o ΝΆ^ H (R)-5-(4-fluorophenyl)N-(5-((3-((1hydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)pyridin-2-yl)1 -methy ί-4-οχο-1,4dihydropyridine-3carboxamide 530.2 (M+H)
300
113 η ίι ho—, Γ il Y Y [i | A ° ° ^bo H (R)-1 -(4-fluoropheny 1)N-(5-((3-((lhydroxypropan-2yl)amtno)-lHpyrazolo[3,4-b]pyrîdin4-yl)oxy)pyridin-2-yl)5-methyl-2-oxo-l,2dihydropyridine-3carboxamide 530.2 (M+H)
114 X o 'Zx z XZ ’^χ /5 7 zx o=/ ° \H/Z— Tl (R)-N-(3-fluoro-4-((3(( 1 -hydroxypropan-2yi)amino)-lHpyrazolo[3,4-b]pyridin4-y l)oxy)pheny 1)-5-(4fltiorophenyl)-! isopropyl-4-oxo-l,4dihydropyridine-3carboxamide 575.2 (M+H)
115 LL AZ Z \=o )=o xz ?Azi x^ I (R)-l-ethyl-N-(3-fluoro4-((3-((1hydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)pheny 1)-5-(4fluoropheny l)-4-o xo1,4-dihydropyridine-3carboxamide 561.2 (M+H)
301
116 1 ,Ν. H il II ho-, || η il il [i । Λ o O ΑΧ / NH θ F nAj] H N (R)-N-(3-fluoro-4-((3(( 1 -hydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-5-(4fluorophenyl)-I-methyl4-o xo-1,4dihydropyridine-3carboxamide 547.2 (M+H)
117 h ÎÎ3 N -AA N ho^ II T X H H 1 /^NH θ F ΧχΊι H N (S)-N-(3-fluoro-4-((3- (( 1 -hydroxy propan -2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-l-(4fluorophenyl)-5-inethyl2-oxo-1,2dihydropyridine-3carboxamide 547.2 (M+H)
118 LL —z 2=0 'b,-. - «P O z (S)-N-(3-flLioro-4-((3(( 1 -hydroxypropan-2yl)amîno)-lHpyrazolo(3,4-b]pyridin4-yl)oxy)phenyl)-5-(4fluorophenyl)-1 -methyl4-oxo-1,4dihydropyridîne-3carboxamide 547.2 (M+H)
302
119 I iz y-É Z 0—0 τι ^0 zz o=/ Z—J <0* (R)-5 -cyclopropy l-N-(3 fluoro-4-((3-((lhyd roxypropan-2yl)amino)-l Hpyrazcdo[3,4-b]pyridin4-yl)oxy)phenyl)-1 -(4fluorophenyl)-2-oxo1,2-dihydropyridine-3carboxamide 573.2 (M+H)
120 Z O Z 4 0 Z 0—0 Tl zz Ο=ζ O0>- Tl T} (R)-l-(3,4diflaorophenyl)-N-(3fluoro-4-((3-((lhydroxypropan-2yl)amino)-lHpyrazolo[3,4“b]pyridin4-yl)oxy)phenyl)-5methyl-2-oxo-L2dihydropyridine-3carboxamide 565.1 (M+H)
121 Br H H0^ I 11 Y Y Y 1 /'NH θ F nYY H N (R)-5-bromo-N-(3fluoro-4-((3-((lhydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-y l)oxy)phenyl)-1 -(4fluorophenyl)-2-oxo1,2-dihydropyridine-3carboxamide 611.1, 613.1 (M+H)
303
122 H il Ί F/_^/ N vA/ N F T ï n T ¥ T \ „/AA ° ° AA /NH 9 n¥o H N (R)-N-(3-fluoro-4-((3- (( 1 -hydroxypropan-2yl)amino)-lHpyrazolo[3:4-b]pyridin4-yl)oxy)phenyl)-l-(3nuorophenyl)-5-methyl2-oxo-1,2dihydropyridine-3carboxamide 547.2 (M+H)
123 AA h il II ho-^ [i j T TA T A ΛΑ ο o AA^P /'NH F Λχι] H N (R)-5-(3,4- difluorophenyl)-N-(3fluoro-4-((3-((lhydroxypropan-2yl)amîno)-lHpy razo 1 o[3,4-b] py ri d i n4-y l)oxy)phenyl)-1 isopropyl-4-oxo-1,4dihydropyridine-3carboxamide 593.2 (M+H)
124 T O z 4 x z' y—O n P ZI °K Ah O (R)-5-(4-chlorophenyl)N-(3-fluoro-4-((3-((ihydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-y l)oxy)pheny !)-1 isopropy 1-4-oxo-1,4dihydropyridine-3carboxamide 591.2 (M+H)
304
125 LL· Μ O T (S)-N-(3-fluoro-4-((3- (( 1 -hydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-5-(4fluorophenyl)-] - i sopropy 1 -4 -oxo-1,4dihydropyrîdine-3carboxamide 575.2 (M+H)
126 A\_y^° Αθ IZ Ά y/ zA/i O I (S)-l-ethyI-N-(3-fluoro- 4-((3-((1hydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-5-(4fluorophenyl)-4-oxol,4-dihydropyridine-3carboxamide 561.2 (M+H)
127 Xi F h N H 7 ho-, if T Y Y Y Ί \ JI A ο o /•NH 9 F Xo m n (R)-5-(2,4- difluorophenyI)-N-(3fluoro-4-((3-((lhydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-Iisopropyl-4-oxo-1,4dihydropyridine-3carboxamide 593.2 (M+H)
305
128 * Y^V N yV ho—, T il X π T T 2-nh 9 n^O n^y H (R)-3-(3,4difluorophenyl)-]-ethylN-(3-fluoro-4-((3-((Ihydroxypropan-2yl)amino)-IHpyrazoto[3,4-b]pyridin4-yl)oxy)phenyl)-2,4dioxo-1,2,3,4tetrahydropy ri m id i ne-5 carboxamîde 596.2 (M+H)
129 Cl H [|Ί FNYY- N YV n ΎΥ ho-, T II Π Π Π ] \ λ,ΑΧ 0 0 J+X /NH θ F N^ÎJ H (R)-5-chloro-N-(3fluoro-4-((3-((lhydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yi)oxy)phenyl)-l-(4fl uoropheny l)-2-oxo1,2-dihydropyridine-3carboxamide 567.2 (M+H)
130 mC y XJ Π XX /Anh o F N^O n^Y H (S)-i-ethyl-N-(3-fluoro- 4-((3-((1hydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-3-(4fluoropheny 1)-2,4dioxo-1,2,3,4- tetrahyd ropy rim id ine-5 carboxamîde 578.2 (M+H)
306
131 LL LL . 0’ \ y—Z )=° IZ 0 & O T (S)-3-(3,4difIuorophenyl)-N-(3fluoro-4-((3-((lhydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yI)oxy)phenyl)-lisopropy 1 -2,4-d i oxo- 1,2,3,4tetrahydropyrimidine-5carboxamide 610.2 (M+H)
132 H fV^yMy^Y00f hoy τ T Π Π II | \ „0w ο o pNH Nho H (S)-4-(3,4- d ifl uoropheny 1 )-2-ethy 1 N-(3-fluoro-4-((3-((lhydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-3,5dioxo-2,3,4,5tetrahydro-1,2,4triazine-6-carboxamide 597.2 (M+H)
133 H hoy T ï T ” Y | Λ ωΑΑ 0 0 AA /NH Y F n^yS N^A H (R)-4-(3,4difluorophenyl)-2-ethylN-(3-fluoro-4-((3-((lhydroxypropan-2yl)amîno)-l Hpyrazolo[3,4-b]pyridin4-yl)oxy)pheny[)-3,5dioxo-2,3,4,5tetrahydro-1,2,4triazine-6-carboxamide 597.2 (M+H)
307
134 I Ο VV ΖΙ Μ - Q “η -τι (S)-4-(3,4- d ifluoropheny Ι)-Ν-(3 fluoro-4-((3-(( 1 hydroxypropan-2yl)amino)-lHpy razo lo [3,4-b] pyridin 4-yl)oxy)phenyl)-2isopropyl-3,5-dioxo2,3,4,5-tetrahydro-1,2,4triazine-6-carboxamide 611.2 (M+H)
135 Ν^Ο0 Η Ν 1 N ΥΥ N /¥ F ho-. T Τ Y Y Y T \ Υ+Υ ο ο /'ΝΗ 0^^ ^^ F /rS Ν 'Υ Η (R)-4-(3,4- difluorophenyl)-N-(3fluoro-4-((3-((lhydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)phenyI)-2isopropyl-3,5-dioxo2,3,4,5-tetrahydro-l,2,4triazine-6-carboxam ide 611.2 (M+H)
136 Η ηο^ r T Y Y Τ Ί Ανη ? F Ν^Ο νΑΥ Η (S)-2-ethyl-N-(3-fluoro- 4-((3-((1hydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-y l)oxy)pheny 1)-4-(4fluorophenyl)-3,5dioxo-2,3,4,5tetrahydro-1,2,4triazine-6-carboxamide 579.2 (M+H)
308
137 H N'NÇ° hoy T T T π Ύ X Λ rAX ° ° /'NH 9 F nXo H (R)-2-ethyl-N-(3-fluoro- 4-((3-((1hydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridiii4-yl)oxy)phenyl)-4-(4fluorophenyl)-355dioxo-2,3,4,5tetrahydro-1,2,4triazine-6-carboxamide 579,2 (M+H)
138 tC^o n v^vN F hoy T il x x II T N H O F Nbo n-^n^ H (S)-3-(3,4difluorophenyl)-l-ethylN-(3-fluoro-4-((3-((lhydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-2,4dioxo-1,2,3,4tetrahydropy ri m id i ne-5 carboxatnide 596.2 (M+H)
139 Y ,Ν,^Ο fVy“XyY η°λ XJ Y s XX /Anh θ F N^ZO H (S)-l-cyclopropyl-N-(3fluoro-4-((3-((lhydroxypropan-2yl)atnino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-3-(4fluorophenyl)-2,4dioxo-1,2,3,4tetrahydropy ri mi d i ne-5 carboxamide 590.2 (M+H)
309
140 H ho-^ T ii T \ -Y/ Y-NH θ I Y Y.Y vY^N. il Ή +’ N-(3-fluoro-4-((3-(((R)l-hydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-y l)oxy)pheny 1)-5-(4fl uorophenyl)-6-oxo-5azaspiro[2.5]octane-7carboxamide 563.2 (M+H)
14] H HCY | Y γ z 'nh । Γ Y Y^Y N n J J X 1 n N-(3-fluoro-4-((3-(((R)1 -hydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-l-(4fluorophenyl)-3-methyl2-oxopiperidine-3carboxamide 551.2 (M+H)
Example 142
[001269] ÎR)-N-(3-fluoro-4-((3-((l-hvdroxypropan-2-yl)amino)-IH-pvrazolor3.4blp¥ridin-4-yl)oxy)phenyQ-5-hydroxy-2-oxo-l-phenyl-l,2-dihydroquinoline-3-carboxamide
[001270]
[001271]
Step A: N-(3-fluoro-4-((3-iodo-I-(4-methoxybenzyl)-l H-pyrazolo[3,4-b]pyridîn
4-yl)oxy)phenyl)-2,5-dioxo-l-phenyl-l,2,5,6,7,8-hexahydroquino!ine-3-carboxamide (Préparation 66; 400 mg, 0.53 mmol), D-alaninol (124 pL, 1.59 mmol), K2CO3 (585 mg, 4.24
310 mmol) and pyrrole-2-carboxylic acid (0.0294 g, 0.265 mmol) were suspended in DMSO (5 mL) and the mixture was degassed for 5 min with Ar. Cu(I)iodide was added and the mixture heated to 60 °C in a sealed tube overnight. The cooled mixture was partitioned between water (30 mL) and EtOAc (30 mL) and the aqueous layer was extracted with EtOAc (2 x 20 mL). The combined organic phases were washed with water (5 x 20 mL) and brine (20 mL), dried over NajSOj, filtered and concentrated. The residue was purified over silica gel to afford 2 products. The lower Rf material was isolated to afford (R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-l-(4methoxybenzyl)-lH-pyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-5-hydroxy-2-oxo-1-phenyl-1,2dihydroquinoline-3-carboxamide (41 mg, i 1%).
| Û01272J Step B: (R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-l-(4methoxybenzyl)-lH-pyrazolo[3,4-b]pyridin-4~yl)oxy)phenyl)-5-hydroxy-2-oxo-1-phenyl-1,2dihydroquinoline-3-carboxamide (41 mg, 0.06 mmol) was dissolved in DCM (0.5 mL) and TFA (1 mL) was added and the reaction was wanned to 35 °C for 4 h. The cooled mixture was concentrated, diluted with DCM and concentrated again. The residue was dissolved in MeOH with a small amount of DCM to aid solubility and K2CO3 (20 mg) was added. After stirring for 2 h, the reaction mixture was filtered and concentrated. The residue was purified over silica gel (0-12% MeOH in DCM) to afford (R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lHpyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-5-hydroxy-2-oxo-1-phenyl-l,2-dihydroquinoline-3carboxamide (11.2 mg, 33%). Mass spectrum: m/z = 581.2 (M+H). 'H NMR (CD3OD) δ 12.23 (s, IH), 9.58(d, 1H), 8.13 (d, 1H), 8.00 (dd, 1H), 7.71-7.57 (m, 3H), 7.40 (ddd, IH), 7.35-7.29 (m, 6H), 7.23 (t, IH), 6.73 (d, IH), 6.14 (d, IH), 6.09 (dd, IH), 3.93 (m, IH), 3.70 (d, 2H), 1.34 (d, 3H).
Example 143
[001273] (R)-2-(4-fluorophenyl)-N-(5-((3-((1 -hydroxvpropan-2-vl)amino)-lHpyrazolo[3.4-b)pvridin-4-vl)oxv)pvridin-2-vl)-3-oxo-2.3-dihvdropyridazine-4-carboxamide hydrochloride
311
[001274]
[001275]
To a mixture of (R)-2-((4-((6-aminopyridin-3-yl)oxy)-l-(4-methoxybenzyt)-lHpyrazolo[3,4-b]pyridin-3-yl)amino)propan-l-ol (Préparation 60; 0,030 g, 0,071 mmol), 2-(4fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxylic acid (0.033 g, 0.14 mmol), EDC1 (0.082 g, 0.43 mmol) and HOBt (0.058 g, 0,43 mmol) was added DMF (0.7 ml). The reaction mixture was allowed to stir for 4 h and was then added to 50 mL of water while stirring. The résultant solids were filtered and washed with water (15 mL) and hexanes (15 mL) and purified via C18 chromatography (5-95% ACN in water with 0.2% TFA). The purified material was dissolved in TFA (2 mL) and heated to 50 °C for 4 h. The reaction mixture was concentrated and partitioned between DCM and IM LiOH. The combined organic layers were dried over sodium sulfate, filtered and concentrated. The residue was purified by Cl8 chromatography (5-95% ACN in water with 0.2% TFA). The fractions containing the product were concentrated in vacuo and then treated with 4N HCl in dioxanes. A stnall amount of methanol (5 mL) was added to make sure everything was in solution. The mixture was concentrated in vacuo. The isolated solids were dried overnight to provide (R)-2-(4-fluorophenyl)-N-(5-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4b]pyridin-4-yl)oxy)pyridin-2-yl)-3-oxo-2,3-dihydropyridazine-4-carboxamide hydrochlorîde (0.0054 g, 0.0092 mmol, 13% yield) as a yellow solid. Mass spectrum: m/z = 517.2 (M+H-HC1). 'HNMR (de-DMSO) δ 12.13 (s, 1H), 8.45 (d, 1H), 8.42 (d, 1H), 8.40 (d, 1H), 8.34 (d, 1H), 8.22 (s, 1 H), 7.96 (m, 1 H), 7.69 (m,2H),7.41 (m, 2H), 6.13 (d, 1 H), 3.80 (m, 1 H), 3.73-3.65 (m, 2H), 3.55-3.45 (m, 3H), 1.22 (d, 3H).
[001276] The following compounds were also made using the procedure according to Example 143.
Ex. No. Structure Name Mass Spectrum
312
144 w ii N H i n n ho-. f T T T J | \ ο ο /''ΝΗ θ F n^Y H (R)-2-(4-f1uorophenyl)N-(5-((3-((lhydroxypropan-2yl)amîno)-lHpyrazolo[3,4-b]pyridin-4yI)oxy)pyridin-2-yl)-6methyl-3-oxo-2,3dihydropyridazine-4carboxamide 531.2 (M+H)
145 ho—, \ T Y T T । O^N ° ° ^F Nbô H (R)-3-(4-fluorophenyl)N-(5-((3-((lhydroxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin-4yl)oxy)pyridin-2-yl)-lisopropyi-2,4-dioxo1,2,3,4tetrahydropyrimidine-5carboxamide 575.2 (M+H)
Example 146
[001277] ('R)-N-(3-fluoiO-4-([3-((l-methoxvpropan-2-vl)amino)-lH-pvrazolof3,4b1pvridin-4-vl)oxv)phenvl)-3-(4-fluorophenvl)-l-isopropyl-2,4-dioxo-1,2,3,45 tetrahvdropyrimidine-5-carboxamide hydrochloride
313
[001278]
[001279]
Step A: 3-(4-Fluoropheny 1)-1 -isopropyl-2,4-dioxo-1,2,3,4-tetrahydropyrimidine5-carboxylic acid (Préparation 7; 11.3 g, 38.5 mmol) was dissolved in DMF (150 mL) and 2-(3H[1,2,3]triazolo[4,5-b]pyridin-3-y 1)-1,1,3,3-tetramethylisouronium hexafluorophosphate(V) (14.7 g, 38.5 mmol) was added. The reaction mixture was stirred for 10 min, and then triethylamine (13.4 ml, 96.3 mmol) was added to the mixture. (R)-4-(4-amîno-2-fluorophenoxy)-l-(4methoxybenzy])-N-(l-methoxypropan-2-yl)-lH-pyrazolo[3,4-b]pyridin-3-amine (14.5 g, 32.1 mmol), dissolved in DMF (50 mL) was added to the reaction and the reaction mixture was stirred overnight. The reaction mixture was partitioned between 80% brine and MTBE. The combined organic layers were dried over NazSOi, filtered and concentrated, during which solids formed. The concentrated solution was filtered to afford pure product. The filtrate was purified over silîca gel (40-80% EtOAc in hexanes) to afford additional product. The combined lots afforded (R)-N(3-fluoro-4-(( l-(4-methoxybenzy 1)-3-(( l-methoxypropan-2-y l)amino)-1 H-pyrazolo[3,4b]pyridin-4-yl)oxy)phenyl)-3-(4-fluorophenyl)-l-isopropyl-2,4-dioxo-l,2,3,4tetrahydropyrimidine-5-carboxamide (22.3 g, 30.7 mmol, 95.7 % yield).
1001280] Step B: (R)-N-(3-fluoro-4-(( l-(4-methoxybenzy 1)-3-(( l -methoxypropan-2yl)amino)-l H-pyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-3-(4-f]uorophenyl)-l-isopropyl-2,4dioxo-l,2,3,4-tetrahydropyrimidine-5-carboxamide (22.3 g, 30.7 mmol) was dissolved in TFA (40 mL) and heated to 50 °C. After 4 h, additional TFA (10 mL) added. After 8 h, the reaction mixture was cooled and carefully poured into MTBE (150 mL) with vigorous stirring. The solids were dissolved in EtOAc and washed with saturated NaHCOj, dried over sodium sulfate, filtered and concentrated. The residue was purified over silîca gel (70-100% EtOAc in hexanes) to afford a solid. The solid was dissolved in DCM (100 mL), and 5M HCl in IPA added (1.5 eq). The solution was added slowly to 700 mL Et2O with stirring. The mixture was stirred for 10 min, then filtered and the solids were washed with Et2O. The isolated solids were dried in vacuum oven to afford
314 (R)-N-(3-fluoro-4-((3-((l -methoxypropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-3-(4-fluoropheny 1)-1-isopropyl-2,4-dioxo-1,2,3,4-tetrahydropyrimidine-5carboxamide hydrochloride (13.9 g, 21.6 mmol, 70.5 % yield) as a bright yellow solid. Mass spectrum: m/z = 606.2 (M+H-HC1). Ή NMR (d6-DMSO) δ 11.07 (s, 1H), 8.68 (s, 1H), 8.24 (d,
1H), 8.03 (dd, 1H), 7.57 (dd, 1H), 7.50 (t, 1H), 7.43 (m, 2H), 7.36 (m, 2H), 6.11 (dd, 1H), 4.78 (m, 1H), 3.98 (m, 1 H), 3.51 (dd, 1 H), 3.39 (dd, 1H), 1.43 (d, 6H), 1.23 (d, 3H).
|0012811 The following compounds were also made using the procedure according to Example 146.
Ex. No. Structure Naine Mass Spectrum
147 O zt 0—° 71 zz ο=ζ O=( Z—( 0 ° (S)-3-cyclopentyl-N-(3fluoro-4-((3-((lmethoxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-lisopropyl-2,4-dioxo1,2,3,4tetrahydropyrimidine-5carboxamide 580.3 (M+H)
148 >nh yu ° ° H (S)-l-ethyl-N-(3-fluoro- 4-((3-((1methoxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin-4y l)oxy )pheny 1 )-3-(4fluorophenyl)-2,4-dioxo1,2,3,4tetrahydropyrimidine-5carboxamide 592.2 (M+H)
315
149 LL 0Z Z \=O 5=0 IZ 0 o^z z0z x (S)-N-(3-fluoro-4-((3((1 -methoxypropan-2yl)amino)-lHpy razo lo[3,4-b]pyrid in-4 yl)oxy)phenyl)-3-(4fluorophenyl)-l-methyl2,4-dioxo-l,2,3,4tetrahydropyrimidine-5carboxamide 578.2 (M+H)
150 X Υτ^^π >NH ° ° + f Nw bT H (S)-l(cyclopropylmethyl)-N(3-fluoro-4-((3-((lmethoxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-3-(4fluorophenyl)-2,4-dioxo1,2,3,4tetrahydropyrimidine-5carboxamide 618.2 (M+H)
151 L γ//?Υ0 0ΝΗ θ F n1j N N (S)-N-(3-fluoro-4-((3(( 1 -methoxypropan-2yl)amino)-lHpy razo lo [3,4-b]pyridin-4yl)oxy)phenyl)-l-(4fluorophenyl)-6-methyl2-oxo-1,2dihydropyridine-3carboxatnide 561.2 (M+H)
316
152 ’Ûj ZI °x 0K° n (S)-l-cyclobutyl-N-(3fluoro-4-((3-((lmethoxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-3-(4fluorophenyl)-2,4-dioxo1,2,3,4tetrahydropyrimidine-5carboxamide 618.2 (M+H)
153 LL LL· \ / \ / ° 0° IZ A '1 —O (S)-3-(3,4difluorophenyl)-N-(3ftuoro-4-((3-((lmeÎhoxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyndin-4yl)oxy)phenyl)-lîsopropyl-2,4-dioxo1,2,3,4tetrahydropyrimidine-5carboxamide 624.2 (M+H)
154 % yA^n \ 0 0 AiAr- /-N H θ F Lîj H N (S)-N-(3-fluoro-4-((3- (( 1 -methoxypropan-2yl)amino)-l Hpyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-3-(4fluorophenyl)-!isopropyl-2,4-dioxo1,2,3,4tetrahydropyrimidine-5carboxamide 606.2 (M+H)
317
155 / wXÇi \ ^YY 0 0 YY //NH θ F n^Îj H N N-(3-fluoro-4-((3-((lmethoxy-2methylpropan-2yl)amino)-l Hpyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-3-(4fluoropheny])-!isopropyl-2,4-dioxo1,2,3,4tetrahydropyrimidine-5carboxamide 620.2 (M+H)
156 \ ^Y^ Y IZ h /\ Y U- o—C Z z-YZI —o N-(3-fluoro-4-((3-((l- methoxy-2methylpropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-4-(4fluorophenyl)-2îsopropy 1-3,5 -d ioxo2,3,4,5-tetrahydro-l ,2,4triazine-6-carboxamide 621.2 (M+H)
157 o O YY \ F Y —z /o y=o xz Ά \—/ TYzx — 1 -ethy 1-N-(3 -fl uoro-4((3-((1-methoxy-2methylpropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin-4yl)oxy)pheny 1)-3-(4fluorophenyl)-2,4-dioxo1,2,3,4tetrahydropyrimidine-5carboxamide 606.2 (M+H)
318
158 ο )=/ F* —z 2=o 2=o IZ Λ z —ο N-(3-fluoro-4-((3-((l- methoxy-2methylpropan-2yl)amino)-lHpyrazo lo[3,4-b]pyridin-4yl)oxy)phenyl)-3-(4fluorophenyl)-l-methyl2,4-dioxo-l,2,3,4tetrahydropyrimidine-5carboxamide 592.2 (M+H)
159 LL Ο T °Λ / O= /~Z '-z Ao )=o IZ X-Vz & —O N-(3-fluoro-4-((3-((1- methoxy-2methyipropan-2yl)amino)-lHpyrazoio[3,4-b]pyridin-4yl)oxy)phenyi)-3-(4fluoropheny 1)-1-(2hydroxy ethy l)-2,4-d ioxo1,2,3,4tetrahydropyrimidine-5carboxamide 622.2 (M+H)
160 T υ/τΧύγ \ XJ ο o CAc /NH 9 F nXj il N (R)-N-(3-fluoro-4-((3- (( 1 -methoxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin-4yl)oxy)phenyI)-l-(4fluorophenyl)-6-methyl2-oxo-1,2dihydropyridine-3carboxamide 561.2 (M+H)
319
161 ^^O— 0 ZI o=/ o=< Z^k > /M /=/ O Π (R)-l-cyclobutyl-N-(3fluoro-4-((3-((lmethoxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin-4y l)oxy)pheny 1)-3-(4fluorophenyl)-2,4-dioxo1,2,3,4tetrahy dropyrimid ine-5 carboxamide 618.3 (M+H)
162 H % γγγΥγγ \ ° ° k^Y /'NH 0 F Y) H N (R)-N-(3-fiuoro-4-((3- (( 1 -methoxypropan-2yl)amino)-l Hpyrazolo[3,4-b]pyridin-4y l)o xy)pheny 1)-4-(4fluorophenyl)-2isopropyl-3,5-dioxo2,3,4,5-tetrahydro-l,2,4triazine-6-carboxamide 607.2 (M+H)
163 yÇ° H H ' 1 N YY N o^ Y Y Y Y Y Ί Y NH Y^ ° ° ^k (S)-N-(3-fluoro-4-((3(( 1 -methoxypropan-2yl)amino)-l Hpyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-4-(4fluorophenyl)-2isopropyl-3,5-dioxo2,3,4,5-tetrahydro-l ,2,4triazine-6-carboxamide 607.2 (M+H)
320
164 Z Z'A--? Αχ ζζ □γ οΑ ΖΑ 0^° ΤΊ N-(4-((3-((4,4difluorobutan-2yl)amino)-lHpyrazolo[3,4-b]pyridin-4yl)oxy)-3-fluorophenyl)3-(4-fluorophenyl)-lisopropyl-2.4-dioxo1,2,3,4tetrahydropyrimidine-5carboxamide 626.2 (M+H)
165 1 Ν 1 ^/ΑΑ, Α\ οΑΤ OoA Xnh θ F Ao H N-(4-((3-((4,4difluorobutan-2yl)amîno)-lHpyrazolo[3,4-b]pyridin-4yl)oxy)-3-fluorophenyl)5 -(4-fl uoropheny 1)-1 methyl-4-oxo-1,4dihydropyridine-3carboxamîde 581.2 (M+H)
166 Z Ζ^γξ vv 0 zz oY z-z ' 0 Π N-(4-((3-((4,4difluorobutan-2yl)amtno)-lHpyrazolo[3,4-b]pyridin-4yl)oxy)-3-fluorophenyl)2-(4-fl uoropheny l)-6isopropyl-3-oxo-2,3dihydropyridazine-4carboxamide 610.2 (M+H)
321
167 1 e [ Y Y Y Ύ ] Y-NH i ’ F Yjîj H N-(5-((3-((4,4difluorobutan-2yl)amino)-l Hpyrazolo[3,4-b]pyridin-4yl)oxy)pyridin-2-yl)-5(4-f]uorophenyl)-Imethyl-4-oxo-l,4dihydropyridine-3carboxamide 564.2 (M+H)
168 LL Cy /=o IZ 0 Y^Z N-(5-«3-((4,4difluorobutan-2yl)amino)-lHpyrazolo[3,4-b]pyridin-4yl)oxy)pyridin-2-yl)-l(4-fluorophenyl)-5methyl-2-oxo-l,2dihydropyridîne-3carboxamide 564.2 (M+H)
169 X Άχ· 0 ZI o=/ O=<Jh- 0 “Π (S)-N-(3-fluoro-4-((3- (( 1 -methoxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyrîdin-4yl)oxy)phenyl)-1 -(4fluorophenyI)-5-methyl2-oxo-1,2dihydropyridine-3carboxamide 561.2 (M+H)
322
170 Η ί|¥ F N - AAN -/¼. /v ¥ ¥ π ¥ Y ] / A nA/ 0 0 AAC A~nh θ F •A N N N-(3-fluoro-4-((3-((lmethoxy-2methylpropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-l~(4fluorophenyl)-5-methyl2-oxo-1,2dihydropyridine-3carboxamide 575.2 (M+H)
171 LL IL . Y \ K Z 2=0 X Hp A- ,Z X rà (S)-3-(3,4- d îfluoropheny 1)-N-(4-((3 (( 1 -ethoxypropan -2yl)a(nino)-lH- pyrazolo[3,4-b]pyridin-4y l)oxy)-3 -fl uoropheny ΟΙ -isopropyl-2,4-dioxo1,2,3,4tetrahydropyrimidine-5carboxamide 638.3 (M+H)
172 h ¥0 A N - AA N -vv, ¥ ¥ T ¥ Ύ Ί \ A- JJ ο o JA Anh VJ H (S)-N-(4-((3-((lethoxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin-4yl)oxy)-3-fluorophenyl)1 -(4-fluorophenyl)-5methyl-2-oxo-l,2dihydropyridine-3carboxamide 575.2 (M+H)
323
173 LL 0 z-z 0 k A r° (S)-N-(4-((3-((1ethoxypropan-2yl)amino)-!Hpyrazolo[3,4-b]pyridin-4yl)oxy)-3-fluorophenyf)2-(4-fluorophenyl)-3oxo-2,3dihydropyridazine-4carboxamide 562.2 (M+H)
174 h il il \ a^naaa Γ x 7 1 \ rv-AA ° 0 AA-c /'NH 9 F n^A H (R)-5-(4-fluorophenyl)-lisopropyl-N-(5-((3-((lmethoxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin-4yl)oxy)pyridin-2-yl)-4oxo-l,4-dihydropyridine3-carboxamide 572.2 (M+H)
175 H il \ N AXN °~λ T T π T 7 1 \ „AA ° ° AA.. /NH θ F 7o νΆ^ H (R)-N-(3-fluoro-4-((3(( 1 -methoxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-2-(4fluorophenyl)-6isopropyl-3-oxo-2,3dihydropyridazine-4carboxamide 590.2 (M+H)
324
176 h ii n °3 Γ Y Y | \ Υαν ο o AzA / NH θ F ΝΧθ ΝΆ^ H (R)-2-(4-fluorophenyl)-6isopropyi-N-(5-((3-(( 1 methoxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin-4yl)oxy)pyridîn-2-yl)-3oxo-2,3dihydropyridazine-4carboxamide 573,2 (M+H)
177 h |A1 \ N Axz N -zA- °a | T Y Y Y | }NH AN ° ° F NXÔ N^A H (R)-5-cyclopropyl-l-(4fluoropheny l)-N -(5-((3 (( 1 -methoxypropan-2y])amino)-lHpyrazolo[3,4-b]pyridin-4y[)oxy)pyridîn-2-yl)-2oxo-l,2-dihydropyridine3-carboxamide 570.3 (M+H)
178 -\ z° A° IZ A o—e Z ?Azi (R)-1 -(4-fluorophenyl)N-(5-((3-((lmethoxypropan-2yl)amino)-l Hpyrazolo[3,4-b]pyridin-4yl)oxy)pyrîdin-2-yl)-5methyl-2-oxo-l,2dihydropyrîdine-3carboxamide 544,2 (M+H)
Example 179
[001282] (R)-l-(cvclopropvlmeth¥l)-N-(3-fluoro-4-((3-«l-methoxypropan-2-yl)amino)1 H-pyrazolof3,4-b |pyridin-4-Yl)oxv)phenv 1)-3-(4-fluorophenvl)-2,4-dioxo-1,2.3,420534
325 tetrahydrop vrim idine-5 -carboxam ide
[001283]
[001284]
To a mixture of l-(cyclopropylmethyl)-3-(4-fluorophenyl)-2,4-dioxo-l,2,3,4tetrahydropyrimidine-5-carboxylic acid (Préparation 10; 0.0438 g, 0.144 mmol), (R)-4-(4-amino2-fluorophenoxy)-l-(4-methoxybenzyl)-N-(!-methoxypropan-2-yl)-lH-pyrazolo[3,4-b]pyridin3-amine (0.050 g, 0.111 mmol), EDCI (0.0637 g, 0.332 mmol) and HOBt (0.0449 g, 0.332 mmol) was added DMF ( 1.1 ml) and stirred overnight. The reaction mixture was added to 30 mL of cold water while stirring. The resulting solids were filtered and washed with water (15 mL) and hexanes (5 mL). The isolated solids were suspended in 5 mL DCM, treated with 5 mL TFA and left to stir for 2 hr at 50 °C. The reaction mixture was concentrated and partitioned between saturated
NaHCCh and DCM. The organic layer was washed with water and then brine, dried overNasSOi, filtered and concentrated. The residue was purified over silica gel (5-50% EtOAc in DCM) to afford (R)-t-(cyclopropylmethyl)-N-(3-fluoro-4-((3-((l-methoxypropan-2-yl)amino)-ÎHpyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-3-(4-fluorophenyl)-2,4-dioxo-l,2,3,4tetrahydropyrimidine-5-carboxamide (54 mg, 71%). Mass spectrum: m/z = 618.2 (M+H). ‘HNMR (dû-DMSO) δ 12.19 (s, IH), 11.03 (s, 1H), 8.92 (s, 1H), 8.13 (d, 1H), 8.00 (dd, 1H), 7.53 (ddd, IH), 7.45 (m, 3H), 7.36 (m, 2H), 6.02 (dd, IH), 5.09 (d, 1H), 3.96 (m, 1H), 3.86 (d, 2H), 3.50 (dd, 1H), 3.37 (dd, IH), 3.27 (s, 3H), 1.22 (d, 3H), 0.57 (m, 2H), 0.44 (m, 2H).
[001285] The foliowing compounds were also made using the procedure according to
Example 179.
Ex. No. Structure Name Mass Spectrum
326
180 A nXJ π Xa /NH θ F Xn K N (R)-l-ethyl-N-(3-fluoro4-((3-((1methoxypropan-2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)pheny 1)-3-(4fluorophenyl)-2,4dioxo-1,2,3,4' tetrahydropy rim id i ne-5 carboxamide 592.2 (M+H)
181 O— ztzV Q zi n (R)-4-ethoxy-N-(3fluoro-4-((3-((lmethoxy propan -2yl)amino)-lHpyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-l-(4fluoropheny l)-2-o xo1,2-dihydropyridine-3carboxamide 591.2 (M+H)
Example 182
[001286) N-(3-fluoro-4-((3-((l-(2-hvdroxypropan-2-yl)c¥clopentyl)amino)-lHP¥razolor3.4-blp¥ridin-4-vl)oxy)phenvD-2-(4-fluorophenyl)-3-oxo-2,3-dihydrop¥ridazine-45 carboxamide
[0012871
327
[001288] Step A: N-(3-fluoro-4-((3-iodo-l-(4-methoxybenzyl)-lH-pyrazolo[3,4-b]pyridin4-yî)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide (Préparation 63; 75 mg, 0.11 mmol), methyl 1-aminocyclopentane-l-carboxylate hydrochloride (57 mg, 0.32 mmol), lH-pyrrole-2-carboxylic acid (5.9 mg, 0.053 mmol) and K2CO3 (117 mg, 0.85 mmol) were suspended in DMSO (1 mL) and nitrogen bubbled through for 5 min. Copper(I) iodide (10 mg, 0.053 mmol) was added and the reaction was heated to 70 °C ovemight. The reaction mixture was cooled, diluted with EtOAc (10 mL), stirred for 10 min and filtered. The filtrate was washed with water and brine, dried over Na2SO4, filtered through Celite® and concentrated. The residue was purified over silica gel (0-10% MeOH in DCM) to afford methyl l-((4-(2-fluoro-4-(2-(4fhiorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamido)phenoxy)-l-(4-methoxybenzyl)-1 Hpyrazolo[3,4-b]pyridin-3-yl)amino)cyclopentane-l-carboxylate (25 mg, 0.035 mmol, 33 % yield). [001289] Step B: Methyl 1-((4-(2-fl uoro-4-(2-(4-fluoro pheny l)-3-oxo-2,3dihydropyridazine-4-carboxamido)phenoxy)-l -(4-methoxybenzyl)-1 H-pyrazolo[3,4-b]pyridin-3yl)amino)cyclopentane-l-carboxylate (25 mg, 0.035 mmol) was dissolved in THF (1 mL) and méthylmagnésium bromide (99 μΐ, 0.14 mmol) was added. The reaction mixture was stirred for 10 min. The reaction mixture was partitioned between saturated NH4CI and EtOAc, and the organic layer was washed with brine, dried over sodium sulfate, filtered and concentrated to afford N-(3-fluoro-4-((3-(( 1-(2-hydroxypropan-2-y l)cyclopentyl)amino)-l-(4-methoxybenzyl)-1 Hpyrazolo[3,4-b]pyridin-4-yI)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4carboxamide (17 mg, 0.024 mmol, 68 % yield).
[001290] Step C; N-(3-fluoro-4-((3-((l -(2-hydroxypropan-2-yl)cyclopentyl)amino)-] -(4methoxybenzyl)-lH-pyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3dihydropyridazine-4-carboxamide (17 mg, 0.024 mmol) was dissolved in DCM (1 mL) and TFA (2 mL) was added. The reaction mixture was stirred at 37 °C overnight. The reaction mixture was concentrated and partitioned between DCM and 1N NaOH. The organic layer was dried over sodium sulfate, filtered and concentrated. The residue was purified over silica gel (1-15% MeOH in DCM) to afford N-(3-fluoro-4-((3-((l-(2-hydroxypropan-2-yl)cyclopentyl)amino)-lHpyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-2-(4-fluoropheny[)-3-oxo-2,3-dihydropyridazine-4carboxamide (9.4 mg, 0.016 mmol, 66 % yield) as a yellow solid. Mass spectrum: m/z = 602.2 (M+H). *H NMR(CDCh) δ 11.85 (s, 1H), 9.26 (s, 1H), 8.42 (d, I H), 8.18 (d, lH),7.97(dd, 1H), 7.60 (m, 2H), 7.42 (ddd, 1H), 7.32-7.22 (m, 3H), 6.10 (dd, 1H), 5.02 (s, 1H), 2.36 (m, 2H), 2.16
328 (m, 2H), 1.85 (m,4H), 1.55 (s, 6H).
[001291] Example 183
[001292] 2-((4-(2-fluoro-4-(2-(4-fluorophenyl)-3-oxo-2.3-dihydropyrtdazine-4carboxamido)phenoxv)-iH-pvrazolof3,4-b]pyridin-3-yhamino)-2-methylpropvl hydrogen su I laie
[001293] n
[001294] N-(3-fluoro-4-((3-((l-hydroxy-2-methylpropan-2-yl)amino)-l H-pyrazolo[3,4b]pyridin-4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide (Example 3; 50 mg, 0.0913 mmol) was suspended in DMF (1 mL) and warmed to 50 °C. Sulfurochloridic acid (9 μΙ, 0.14 mmol) was added and the reaction mixture was stirred for 10 min.
The reaction mixture was cooled to RT, MTBE (2 mL) was added, and the reaction mixture was stirred vigorously for 10 min. The reaction mixture was decanted and the remaining oil was treated with water (2 mL) with vigorous stirring. The resulting solids were fîltered and dried to afford 2((4-(2-fluoro-4-(2-(4-fkiorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamido)phenoxy)-lHpyrazolo[3,4-b]pyndin-3-yl)amino)-2-methylpropyl hydrogen sulfate (44 mg, 0.0701 mmol, 76.8 % yield) as a yellow solid. Mass spectrum: m/z = 548.2 (M+H-SO3). 'H NMR (CDCh) δ 12.38 (br s, 1H), 11.70 (s, 1H), 8.37 (d, 1H), 8.27 (d, 1H), 8.18 (m, 1H), 8.03 (dd, lh), 7.69 (m, 2H), 7.61-7.49 (m, 2H), 7.41 (m, 2H), 6.08 (d, 1H), 3.86 (s, 2H), 1.40 (s, 6H).
Examples 184 and 185
[001295] (S)-N-(3-fluoro-4-((3-((3-hydroxy-3-methylbutan-2-yl)amino)-lH-pyrazolo[3,42 0 b]pyridin-4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide (Example 184) and (R)-N-(3-fluoro-4-((3-((3-hydroxy-3-methylbutan-2-yl)amino)-ÎHpyrazolo[3,4-b]pyndin-4-y])oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4carboxamîde (Example 185)
329
Example 185
[001296] N-(3-fluoro-4-((3-((3-hydroxy-3-methylbutan-2-yl)amino)-lH-pyrazolo[3,4b]pyridin-4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2}3-dihydropyridazine-4-carboxamide was purified via SFC on a Chiral Tech IA column (5-70% MeOH:IPA:DJEA 80:20:0.1 ) to afford each purified enantiomer. Absolute configuration is not known. Peak 1: Mass spectrum: m/z = 562.1 (M+H). Peak 2: Mass spectrum: m/z = 562.1 (M+H).
330
[001297] Following the procedure in Example 184, the following single enantiomers, prepared as racemates as described above, were isolated from their racemic mixtures.
Ex. No. Structure Name Mass Spectru m
186 HO H O J Χ^ϊΥΌς \Ành θ F H N N-(3-fluoro-4-((3(((lS,2R)-2(hydroxymethyl)cyclopent yl)amino)-l Hpyrazolo[3,4-b]pyridin-4y l)oxy )pheny 1)-2-(4fluorophenyl)-3-oxo-2,3dihydropyridazine-4carboxamide 574.2 (M+H)
187 HO H YY Tï TïΊ / \ AY ο ο YY^ Y/' N H ^^F H N N-(3-fluoro-4-((3(((lR,2S)-2(hydroxymethy 1 )cyc lopent yl)amino)-lHpyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-2-(4fluorophenyl)-3-oxo-2,3dihydropyridazine-4carboxamide 574.2 (M+H)
188 \ I “ζΎτ Y< U ZI oY z-z 0 ~n (S)-N-(3-fluoro-4-((3-((4hydroxy-1 -methoxy-2methylbutan-2-yl)amino)lH-pyrazolo[3,4b]pyridin-4yl)oxy)phenyl)-2-(4fluorophenyl)-3-oxo-2,3- 592.2 (M+H)
331
dihydropyridazine-4carboxamide
189 Η ΓΝ ho N Yv- N \z J ο o O-ZNH F λή ν-Ύ^ H (R)-N-(3-fluoro-4-((3-((4hydroxy-1 -methoxy-2methy Ibutan -2-y 1 )am ino)lH-pyrazoio[3,4b]pyridin-4yl)oxy)pheny 1)-2-(4fluorophenyl)-3-oxo-2,3dihydropyridazine-4carboxamide 592.2 (M+H)
190 0 YY^?Yl \ .Yy ° ° \Yr HO^' Y I Y M N (S)-N-(3 -fluoro-4-((3-(( 1 hydroxy-3methoxypropan-2yl)amino)-l Hpyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-2-(4fluorophenyl)-3 -oxo-2,3dihydropyridazine-4carboxamide 564.2 (M+H)
191 I Y ζγ V 0 ZI °γ O^Y Z-z Tl (R)-N-(3-fiuoro-4-((3-((lhydroxy-3methoxypropan-2yi)amino)-l Hpyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-2-(4fluorophenyl)-3-oxo-2,3dihydropyridazine-4carboxamide 564.1 (M+H)
332
192 X fyykXX Λ ΥΥ ο ο Ί Ar ηοΧ·νη ? F NOÙ Η π (R)-N-(3-fluoro-4-((3-((lhydroxy-4-methoxybutan2-yl)amino)-IHpyrazolo[3,4-b]pyridin-4y i)oxy)pheny 1)-2-(4fluoropheny 1)-3-oxo-2,3dihydropyridazine-4carboxamide 578.2 (M+H)
193 x ν/ΧΧ \ YJ ° ° YYC ΗθΥ Ν» ? F «Y Η Ν (S)-N-(3-fluoro-4-((3-((lhy droxy-4-m ethoxy butan2-yl)amino)-ÎHpyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-2-(4fluorophenyl)-3-oxo-2,3dihydropyridazine-4carboxainide 578.2 (M+H)
194 Uζ-ζ C χ° )=° rz 5 γΓζι i ο U- χ (R)-N-(4-((3-((4,4difluoro-1 -hydroxybutan2-yl)amino)-lHpyrazolo[3s4-b]pyridin-4yl)oxy)-3-fluorophenyl)-2(4-fluorophenyl)-3-oxo2,3-dihydropyridazine-4carboxamide 584.2 (M+H)
195 0 ζ-ζ 0 Υο χ° rz ^'Χ^ο U- τ (S)-N-(4-((3-((4,4d ifluoro-1 -hydroxy butan2-yl)amino)-lHpyrazolo[3,4-b]pyndin-4yl)oxy)-3-fluorophenyl)-2(4-fl uoropheny l)-3-o xo2,3-dihydropyridazine-4- 584.2 (M+H)
333
carboxamide
Example 196
[001298] N-(4-((3-amino-lH-pvrazolo[3.4-blPvridin-4-vl)oxvÎ-3-fluorophenvl)-3-(4fluorophenvl)-l-isopropvl-2,4-dioxo-L2,3.4-tetrahvdropyrimidine-5-carboxamide
[001299]
[001300]
Step A: A suspension of N-(3-fluoro-4-((3-iodo-l-(4-methoxybenzyl)-IHpyrazolo[3,4-b]pyridin-4-yl)oxy)pheny 1)-3-(4-fluorophenyl)-l-isopropyl-2,4-dioxo-l,2,3,4tetrahydropyrimidine-5-carboxamide (Préparation 65; 100 mg, 0.13 mmol), tert-butyl carbamate (0.153 g, 1.3 mmol), Nl,N2-dimethylethane-l,2-diamine (0.032 ml, 0.26 mmol), K3PO4 (0.16 g, 0.78 mmol) and copper(I) iodide (0.024 g, 0.13 mmol) in dioxane (2 mL) was stirred at 60 °C for
h. The reaction mixture was diluted with water (30 mL) and extracted with DCM (3x 20 mL).
The organic layer was washed with brine (15 mL), dried over sodium sulfate, filtered and concentrated. The residue was purified over silica gel (20-60% EtOAc in hexanes) to afford tertbutyl (4-(2-fluoro-4-(3-(4-fluorophenyl)-l-isopropyl-2,4-dîoxo-l,2,3,4-tetrahydropyrimidine-5carboxamido)phenoxy)-l-(4-methoxybenzyl)-lH-pyrazolo[3,4-b]pyridin-3-yl)carbamate (45.8 mg, 46%).
[001301] Step B: To a solution of tert-butyl (4-(2-fluoro-4-(3-(4-fluorophenyl)-l-isopropyl2,4-dÎoxo-l,2,3,4-tetrahydropyrimidine-5-carboxamido)phenoxy)-l-(4-methoxybenzyl)-IH pyrazolo[3,4-b]pyridin-3-yl)carbamate (45.8 mg, 0.061 mmol) in DCM (I mL) was added TFA (2 mL). The mixture was stirred at 35°C for 6 h, then at room température o verni ght. The mixture was concentrated and the residue was dissolved in MeOH/DCM, treated with K2CO3 (25.2 mg, 0.182 mmol) and stirred for 3 h. The mixture was filtered and concentrated. The residue was purified over silica gel (0-5% MeOH in DCM) to afford N-(4-((3-amino-l H-pyrazoIo[3,420534
334
b]pyridin-4-yl)oxy)-3-fluorophenyl)-3-(4-fluorophenyl)-]-isopropyl-2,4-dioxo-l,2,3,4tetrahydropyrimidine-5-carboxamide (20.3 mg, 60%). Mass spectrum: m/z = 534.2 (M+H). ’H NMR(CDCh) δ 10.97 (s, 1H), 9.65 (s, 1H), 8.69 (s, 1H), 8.21 (d, 1H), 7.89 (dd, 1H), 7.29 (ddd, 1H), 7.27-7.20 (m, 4H), 6.11 (dd, 1H),4.98 (m, 1H), 4.50 (s, 2H), 1.51 (d, 6H).
[001302] The following compounds were also synthesized using the procedure according to
Example 196.
Ex. No. Structure Name Mass Spectrum
197 z 2 Z z)-O m + Q ZI 0=7 7=ς 0 N-(3-fluoro-4-((3- (methylamino)-1Hpyrazolo[3,4-b]pyridin4-yl)oxy)pheny 1)-3-(4fiuorophenyl)-!- ï sopropy 1 -2,4-dioxo- 1,2,3,4- tetrahydropyrimidine-5carboxamide 548.2 (M+H)
198 wAxn XJ1 0 0 L/X ^-NH θ F Ν0Π H N-(4-((3-(ethylamino)- 1 H-pyrazolo[3,4b]pyridin-4-yl)oxy)-3fluorophenyl)-3-(4Huorophenyl)-!isopropyl-2,4-dioxo1,2,3,4tetrahydropyrimidine-5carboxamide 562.2 (M+H)
335
Example 199
[001303] (R)-N-(3-fluoro-4-((3-((t-hvdroxvpropan-2-vl)amino)-lH-pvrazolo[3.4b1pyridin-4-vÎ)oxy)phenyl)-l-(4-fluorophenyl)-5-isopropyL2-oxo-1.2-dihvdropvridine-3carboxamide hydrochloride
[001305] Step A: l-(4-Fluorophenyl)-2-oxo-5-(prop-l-en-2-yl)-l,2-dihydropyridine-3carboxylic acid (Préparation 48; 93.7 mg, 0.343 mmol) was dissolved in DMF (3 mL). 2-(3H[ 1,2,3]triazolo[4,5-b]pyridin-3-yl)-l ,1,3,3-tetramethylisouronium hexafluorophosphate(V) (130 mg, 0.343 mmol) was added and the solution was stirred for 5 minutes followed by the addition of N-ethyl-N-isopropylpropan-2-amine (119 μΐ, 0.686 mmol) and (R)-2-((4-(4-amino-2fluorophenoxy)-l-(4-methoxybenzyl)-lH-pyrazolo[3,4-b]pyridin-3-yl)amino)propan-l-ol (Préparation 53; 100 mg, 0.229 mmol). After stirring overnight, the reaction mixture was partitioned between water and EtOAc, washed with water and brine, dried over sodium sulfate, filtered and concentrated. The residue was purified over silica gel to afford (R)-N-(3-fluoro-4-((3((l-hydroxypropan-2-y!)amino)-l-(4-methoxybenzyl)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-i -(4-fluorophenyl)-2-oxo-5-(prop-l-en-2-yl)-L2-dihydropyridine-3-carboxamide (194 mg, 0.280 mmol, 123 % yield).
[001306] Step B: (R)-N-(3-fluoro-4-((3-(( 1 -hydroxypropan-2-yl)amino)-1 -(4methoxybenzyl)-lH-pyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-l-(4-fluorophenyl)-2-oxo-5-(propl-en-2-yl)-l,2-dihydropyridine-3-carboxamide (190 mg, 0.274 mmol) was dissolved in 25 mL of methanol and 5 mL of EtOAc. 10% Pd/C (30 mg, 0.253 mmol) was added and the solution was purged with hydrogen balloon and stirred under balloon pressure of hydrogen for 1 h. The reaction mixture was filtered and concentrated. The residue was purified over silica gel (20% DCM in EtOAc) to afford (R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-l-(4-methoxybenzyl)lH-pyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-1 -(4-fluorophenyl)-5-isopropyl-2-oxo-l,220534
336 dihydropyridine-3-carboxamide (138mg, 0.199 mmol, 72.4 % yield).
[001307| Step C: (R)-N-(3-f1uoro-4-((3-((l-hydroxypropan-2-yl)amino)-l-(4methoxybenzyl)-lH-pyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-l-(4-fluorophenyl)-5-isopropyl-2oxo-l,2-dihydropyridine-3-carboxamide (138mg, 0.199 mmol) was slurried in DCM (1 mL) and
TFA (20 mL) added and heated to 50 C for 4 h. The reaction mixture was concentrated, partitioned between EtOAc and IM NaOH, dried over sodium sulfate, filtered and concentrated. The residue was purifîed over silica gel and the isolated product was converted to the HCl sait to afford (R)N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyndin-4-yl)oxy)phenyl)l-(4-fluorophenyl)-5-isopropyl-2-oxo-l,2-dihydropyridine-3-carboxamide hydrochloride as a 10 yellow solid. Mass spectrum: m/z = 575.2 (M+H-HCI). *H NMR (CDCE) δ 12.30 (s, IH), 8.72 (d, IH), 8.68 (d, IH), 8.06 (dd, IH), 7.46-7.39 (m, 4H), 7.31-7.24 (m, 3H), 6.12 (d, IH), 5.32 (br s, IH), 4.17 (m, 1 H), 3.92 (dd, 1 H), 3.72 (dd, 1 H), 2.87 (m, IH), 1.38 (d, 3H), 1.30 (d, 6H).
[001308] The foliowing compounds were also synthesized using the procedure according to Example 199.
Ex. No. Structure Name Mass Spectrum
200 H ί|Ί N xAA N ηοά T II π ¥ Y ] \ As Λ ο ο C ,A Y'NH θ ArS (R)-5-ethyl-N-(3-fluoro4-((3-(( 1-hydroxypropan2-yI)amino)-lHpyrazolo[3,4-b]pyridin-4yl)oxy)pheny 1)-1-(4fluorophenyl)-2-oxo-1,2dihydropyridine-3carboxamide 561.2 (M+H)
337
201
(R)-N-(3 -fluoro-4-((3 (( 1 -hydroxy propan-2 yl)amîno)-IHpyrazolo[3,4-b]pyridin-4y 1 )oxy)pheny I)-1 -(4fluorophenyl)-2-oxo-5propyl-1,2dihydropyridîne-3carboxamide
575.2 (M+H)
Abbrevîations:
ACN acetonitrile
AcOH acetic acid
Boc, BOC tert-butyl carboxylate group
DCM Dichloromethane
DIEA N ,N-Diisopropylethy lamine
DMA N ,N -Dimethy lacetam ide
DMF N,N-Dimethylformamide
DMSO Dimethylsulfoxide
EtsO Diethyl Ether
EtOAc Ethyl Acetate
EtOH Ethanol
eq équivalent
h hour, hours
HATU 1 -[Bis(dimethy lamino)methylene]-l H-1,2,3-triazolo[4,5b]pyridinium 3-oxide hexafluorophosphate or 2-(7-Aza-lH- benzotriazole-l-yl)-l,l,3,3-tetramethyluronium hexafluorophosphate
HOAc Acetic Acid
338
ΙΡΑ Isopropyl alcohol
min minute, minutes
MTBE Methyl tert-Butyl Ether
10%Pd/C Palladium 10 wt. % (dry basis), active carbon, wet, Degussa
Pd(PPh3)4 Tetrakis(triphenylphosphine)paHadium (0)
Pd3(dba)3 tris(dibenzylideneacetone)dipalladium (0)
TFA Trifluoroacetic acid
THF tetrahydrofuran
X-PHOS dicycIohexyl(2’,4’,6’-triisopropyl-[ï,l’-biphenyl]-2yl)phosphine
339
Exemplary Embodiments
Embodiment 1. A compound of formula I, wherein the compound is a compound of Example No, 25, 37, 46, 48, 55, 58, 72, 76, 77, 78, 83, 84, 85, 91, 97, 100, 103, 105, 107, 108, 114, 115, 119, 121, 124, 125, 126, 127, 129, 151, 152, 163, 169, 188, 190, 199,200, or 201, or a pharmaceutically acceptable sait or solvaté thereof.
Embodiment 2. A pharmaceutical combination which comprises (a) a compound of Formula I or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 3. A pharmaceutical combination which comprises (a) a compound of Formula I or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 4. The pharmaceutical combination of Embodiment 2, or for use of Embodiment 3, wherein the compound of Formula 1 or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Formula I or a pharmaceutically acceptable sait thereof and of the additional therapeutic agent are together therapeutically effective.
Embodiment 5. A pharmaceutical composition, comprising (a) a compound of Formula 1 or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 6. The pharmaceutical combination of Embodiment 2, or for use of Embodiment 3, or the pharmaceutical composition of Embodiment 5, wherein the additional therapeutic agent is an anticancer agent.
Embodiment 7. The pharmaceutical combination of Embodiment 2, or for use of Embodiment 3, or the pharmaceutical composition of Embodiment 5, wherein the additional therapeutic agent is
340 binimetinib and encorafenib.
Embodiment 8. The pharmaceutical combination of Embodiment 2, or for use of Embodiment 3, or the pharmaceutical composition of Embodiment 5, wherein the additional therapeutic agent is 5 binimetinib.
Embodiment 9. The pharmaceutical combination of Embodiment 2, or for use of Embodiment 3, or the pharmaceutical composition of Embodiment 5, wherein the additional therapeutic agent is encorafenib.
Embodiment 10. The pharmaceutical combination of Embodiment 2, or for use of Embodiment 3, or the pharmaceutical composition of Embodiment 5, wherein the additional therapeutic agent is selumetinib.
Embodiment 1 l. The pharmaceutical combination of Embodiment 2, or for use of Embodiment 3, or the pharmaceutical composition of Embodiment 5, wherein the additional therapeutic agent is sorafenib.
Embodiment 12. The pharmaceutical combination of Embodiment 2, or for use of Embodiment 3, 20 or the pharmaceutical composition of Embodiment 5, wherein the additional therapeutic agent is trametinib.
Embodiment 13. The pharmaceutical combination of Embodiment 2, or for use of Embodiment 3, or the pharmaceutical composition of Embodiment 5, wherein the additional therapeutic agent is 25 vemurafenib.
Embodiment 14. The pharmaceutical combination of Embodiment 2, or for use of Embodiment 3, or the pharmaceutical composition of Embodiment 5, wherein the additional therapeutic agent is an EGFR inhibitor.
341
Embodiment 15. The pharmaceutical combination of Embodiment 2, or for use of Embodiment 3, or the pharmaceutical composition of Embodiment 5, wherein the additional therapeutic agent is cetuximab or a bîosimilar thereof.
Embodiment 16. The pharmaceutical combination of Embodiment 2, or for use of Embodiment 3, or the pharmaceutical composition of Embodiment 5, wherein the additional therapeutic agent is panitumumab or a bîosimilar thereof.
Embodiment 17. The pharmaceutical combination of Embodiment 2, or for use of Embodiment 3, 10 or the pharmaceutical composition of Embodiment 5, wherein the additional therapeutic agent is erlotinib.
Embodiment 18. The pharmaceutical combination of Embodiment 2, or for use of Embodiment 3, or the pharmaceutical composition of Embodiment 5, wherein the additional therapeutic agent is 15 lapatinib.
Embodiment 19. The pharmaceutical combination of Embodiment 2, or for use of Embodiment 3, or the pharmaceutical composition of Embodiment 5, wherein the additional therapeutic agent is gefilinib.
Embodiment 20. The pharmaceutical combination of Embodiment 2, or for use of Embodiment 3, or the pharmaceutical composition of Embodiment 5, wherein the additional therapeutic agent is a checkpoint inhibitor.
Embodiment 21. The pharmaceutical combination of Embodiment 2, or for use of Embodiment 3, or the pharmaceutical composition of Embodiment 5, wherein the additional therapeutic agent is nivolumab or a bîosimilar thereof.
Embodiment 22. The pharmaceutical combination of Embodiment 2, or for use of Embodiment 3, 30 or the pharmaceutical composition of Embodiment 5, wherein the additional therapeutic agent îs pembrolizumab or a bîosimilar thereof.
342
Embodiment 23. The pharmaceutical combination of Embodiment 2, or for use of Embodiment 3, or the pharmaceutical composition of Embodiment 5, wherein the additionaî therapeutic agent is cemiplimab or a biosîmilar thereof.
Embodiment 24. The pharmaceutical combination of Embodiment 2, or for use of Embodiment 3, or the pharmaceutical composition of Embodiment 5, wherein the additionaî therapeutic agent is pidilizumab or a biosîmilar thereof.
Embodiment 25. The pharmaceutical combination of Embodiment 2, or for use of Embodiment 3, 10 or the pharmaceutical composition of Embodiment 5, wherein the additionaî therapeutic agent is atezolizumab or a biosîmilar thereof.
Embodiment 26. The pharmaceutical combination of Embodiment 2, or for use of Embodiment 3, or the pharmaceutical composition of Embodiment 5, wherein the additionaî therapeutic agent is 15 avelumab or a biosîmilar thereof.
Embodiment 27. The pharmaceutical combination of Embodiment 2, or for use of Embodiment 3, or the pharmaceutical composition of Embodiment 5, wherein the additionaî therapeutic agent is durvalumab or a biosîmilar thereof.
Embodiment 28. A pharmaceutical combination which comprises (a) a compound of Example No. 25, 37, 46, 48, 55, 58, 72, 76, 77, 78, 83, 84, 85, 91, 97, 100, 103, 105, 107, 108, 114, 115, 119, 121, 124, 125, 126, 127, 129, 151, 152, 163, 169, 188, 190, 199, 200, or 201, or a pharmaceutically acceptable sait thereof, and (b) an additionaî therapeutic agent .
Embodiment 29. A pharmaceutical combination which comprises (a) a compound of Example No. 25, 37, 46, 48, 55, 58, 72, 76, 77, 78, 83, 84, 85, 91, 97, 100, 103, 105, 107, 108, 114, 115, 119, 121, 124, 125, 126, 127, 129, 151, 152, 163, 169, 188, 190, 199, 200, or 201, or a pharmaceutically acceptable sait thereof, and (b) an additionaî therapeutic agent, for use in therapy.
343
Embodiment 30. The pharmaceutical combination of Embodiment 28, or for use of Embodiment 29, wherein the compound of Example No. 25, 37, 46, 48, 55, 58, 72, 76, 77, 78, 83, 84, 85, 91, 97, 100, 103, 105, 107, 108, 114, 115, 119, 121, 124, 125, 126, 127, 129, 151, 152, 163, 169, 188, 190, 199,200, or 201, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 25, 37, 46, 48, 55, 58, 72,76, 77, 78, 83, 84, 85,91,97, 100, 103, 105, 107, 108, 114, 115, 119, 121, 124, 125, 126, 127, 129, 151, 152, 163, 169, 188, 190, 199, 200, or 201, or a pharmaceutically acceptable sait thereof and of the additional therapeutic agent are together therapeutically effective.
Embodiment 31. A pharmaceutical composition, comprising (a) a compound of Example No. 25, 37,46,48,55, 58, 72,76, 77, 78, 83, 84,85,91,97, 100, 103, 105, 107, 108, 114, 115, 119, 121, 124, 125, 126, 127, 129, 151, 152, 163, 169, 188, 190, 199, 200, or 201, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 32. The pharmaceutical combination of Embodiment 28, or for use of Embodiment 29, or the pharmaceutical composition of Embodiment 31, wherein the additional therapeutic agent is an anticancer agent.
Embodiment 33. The pharmaceutical combination of Embodiment 28, or for use of Embodiment 29, or the pharmaceutical composition of Embodiment 31, wherein the additional therapeutic agent is binimetinib and encorafenib.
Embodiment 34. The pharmaceutical combination of Embodiment 28, or for use of Embodiment 29, or the pharmaceutical composition of Embodiment 31, wherein the additional therapeutic agent is binimetinib.
Embodiment 35. The pharmaceutical combination of Embodiment 28, or for use of Embodiment 29, or the pharmaceutical composition of Embodiment 31, wherein the additional therapeutic agent is encorafenib.
344
Embodiment 36. The pharmaceutical combination of Embodiment 28, or for use of Embodiment 29, or the pharmaceutical composition of Embodiment 31, wherein the additional therapeutic agent is selumetinib.
Embodiment 37. The pharmaceutical combination of Embodiment 28, or for use of Embodiment 29, or the pharmaceutical composition of Embodiment 31, wherein the additional therapeutic agent is sorafenib.
Embodiment 38. The pharmaceutical combination of Embodiment 28, or for use of Embodiment 29, or the pharmaceutical composition of Embodiment 31, wherein the additional therapeutic agent is trametinib.
Embodiment 39. The pharmaceutical combination of Embodiment 28, or for use of Embodiment 29, or the pharmaceutical composition of Embodiment 31, wherein the additional therapeutic agent is vemurafenib.
Embodiment 40. The pharmaceutical combination of Embodiment 28, or for use of Embodiment 29, or the pharmaceutical composition of Embodiment 31, wherein the additional therapeutic agent is an EGFR inhibitor.
Embodiment 41. The pharmaceutical combination of Embodiment 28, or for use of Embodiment 29, or the pharmaceutical composition of Embodiment 31, wherein the additional therapeutic agent is cetuximab or a biosimilar thereof.
Embodiment 42. The pharmaceutical combination of Embodiment 28, or for use of Embodiment 29, or the pharmaceutical composition of Embodiment 31, wherein the additional therapeutic agent is panitumumab or a biosimilar thereof.
Embodiment 43. The pharmaceutical combination of Embodiment 28, or for use of Embodiment 29, or the pharmaceutical composition of Embodiment 31, wherein the additional therapeutic agent is erlotinib.
345
Embodiment 44. The pharmaceutical combination of Embodiment 28, or for use of Embodiment 29, or the pharmaceutical composition of Embodiment 31, wherein the additional therapeutic agent is iapatinib.
Embodiment 45. The pharmaceutical combination of Embodiment 28, or for use of Embodiment 29, or the pharmaceutical composition of Embodiment 31, wherein the additional therapeutic agent is gefitinib.
Embodiment 46. The pharmaceutical combination of Embodiment 28, or for use of Embodiment 10 29, or the pharmaceutical composition of Embodiment 31, wherein the additional therapeutic agent is a checkpoint inhibitor.
Embodiment 47. The pharmaceutical combination of Embodiment 28, or for use of Embodiment 29, or the pharmaceutical composition of Embodiment 31, wherein the additional therapeutic agent 15 is nivolumab or a biosimilar thereof.
Embodiment 48. The pharmaceutical combination of Embodiment 28, or for use of Embodiment 29, or the pharmaceutical composition of Embodiment 31, wherein the additional therapeutic agent is pembrolizumab or a biosimilar thereof.
Embodiment 49. The pharmaceutical combination of Embodiment 28, or for use of Embodiment 29, or the pharmaceutical composition of Embodiment 31, wherein the additional therapeutic agent is cemiplimab or a biosimilar thereof.
Embodiment 50. The pharmaceutical combination of Embodiment 28, or for use of Embodiment 29, or the pharmaceutical composition of Embodiment 31, wherein the additional therapeutic agent is pidilizumab or a biosimilar thereof.
Embodiment 5 I. The pharmaceutical combination of Embodiment 28, or for use of Embodiment 30 29, or the pharmaceutical composition of Embodiment 31, wherein the additional therapeutic agent is atezolizumab or a biosimilar thereof.
346
Embodiment 52. The pharmaceutical combination of Embodiment 28, or for use of Embodiment 29, or the pharmaceutical composition of Embodiment 31, wherein the additional therapeutic agent is avelumab or a biosimilar thereof.
Embodiment 53. The pharmaceutical combination of Embodiment 28, or for use of Embodiment 29, or the pharmaceutical composition of Embodiment 31, wherein the additional therapeutic agent is durvalumab or a biosimilar thereof.
Embodiment 54. A pharmaceutical combination which comprises (a) a compound of Example No. 10 25, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 55. A pharmaceutical combination which comprises (a) a compound of Example No. 25, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 56. The pharmaceutical combination of Embodiment 54, or for use of Embodiment 55, wherein the compound of Example No. 25, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts ofthe compound 20 of Example No. 25 or a pharmaceutically acceptable sait thereof and of the additional therapeutic agent are together therapeutically effective.
Embodiment 57. A pharmaceutical composition, comprising (a) a compound of Example No. 25, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a 25 pharmaceutically acceptable diluent or carrier.
Embodiment 58. The pharmaceutical combination of Embodiment 54, or for use of Embodiment 55, or the pharmaceutical composition of Embodiment 57, wherein the additional therapeutic agent is an anticancer agent.
347
Embodiment 59. The pharmaceutical combination of Embodiment 54, or for use of Embodiment 55, or the pharmaceutical composition of Embodiment 57, wherein the additional therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, erlotinib, lapatinib, gefltinib, nivolumab or a biosimilar thereof, pembrolizumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 60. A pharmaceutical combination which comprises (a) a compound of Example No. 37, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 61. A pharmaceutical combination which comprises (a) a compound of Example No. 37, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 62. The pharmaceutical combination of Embodiment 60, or for use of Embodiment 61, wherein the compound ofExample No. 37, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound ofExample No. 37, or a pharmaceutically acceptable sait thereof and of the additional therapeutic agent are together therapeutically effective.
Embodiment 63. A pharmaceutical composition, comprising (a) a compound ofExample No. 37, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 64. The pharmaceutical combination of Embodiment 60, or for use of Embodiment 61, or the pharmaceutical composition of Embodiment 63, wherein the additional therapeutic agent is an anticancer agent.
348
Embodiment 65. The pharmaceutical combination of Embodiment 60, or for use of Embodiment 61, or the pharmaceutical composition of Embodiment 63, wherein the additional therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, erlotinib, lapatinib, gefitinib, nivolumab or a biosimilar thereof, pembrolizumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 66. A pharmaceutical combination which comprises (a) a compound of Example No. 46, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 67. A pharmaceutical combination which comprises (a) a compound of Example No. 46, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 68. The pharmaceutical combination of Embodiment 66, or for use of Embodiment 67, wherein the compound of Example No. 46, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 46, or a pharmaceutically acceptable sait thereof and of the additional therapeutic agent are together therapeutically effective.
Embodiment 69. A pharmaceutical composition, comprising (a) a compound of Example No. 46, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 70. The pharmaceutical combination of Embodiment 66, or for use of Embodiment 67, or the pharmaceutical composition of Embodiment 69, wherein the additional therapeutic agent is an anticancer agent.
349
Embodiment 71. The pharmaceutical combination of Embodiment 66, or for use of Embodiment 67, or the pharmaceutical composition of Embodiment 70, wherein the additional therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a bîosimilar thereof, panitumumab or a bîosimilar thereof, erlotinîb, lapatinib, gefitinib, nivolumab or a bîosimilar thereof, pembrolizumab or a bîosimilar thereof, cemiplîmab or a bîosimilar thereof, pidilizumab or a bîosimilar thereof, atezolizumab or a bîosimilar thereof, avelumab or a bîosimilar thereof, and durvalumab or a bîosimilar thereof.
Embodiment 72. A pharmaceutical combination which comprises (a) a compound of Example No. 48, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 73. A pharmaceutical combination which comprises (a) a compound of Example No. 48, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 74. The pharmaceutical combination of Embodiment 72, or for use of Embodiment 73, wherein the compound of Example No. 48, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for sîmultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 48, or a pharmaceutically acceptable sait thereof and of the additional therapeutic agent are together therapeutically effective.
Embodiment 75. A pharmaceutical composition, comprising (a) a compound of Example No. 48, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 76. The pharmaceutical combination of Embodiment 72, or for use of Embodiment 73, or the pharmaceutical composition of Embodiment 75, wherein the additional therapeutic agent is an anticancer agent.
350
Embodiment 77. The pharmaceutical combination of Embodiment 72, or for use of Embodiment 73, or the pharmaceutical composition of Embodiment 75, wherein the additional therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, erlotinib, lapatinib, gefïtînib, nivolumab or a biosimilar thereof, pembrolizumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 78. A pharmaceutical combination which comprises (a) a compound of Example No. 55, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 79. A pharmaceutical combination which comprises (a) a compound of Example No. 55, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 80. The pharmaceutical combination of Embodiment 78, or for use of Embodiment 79, wherein the compound of Example No. 55, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 55, or a pharmaceutically acceptable sait thereof and of the additional therapeutic agent are together therapeutically effective.
Embodiment 81. A pharmaceutical composition, comprising (a) a compound of Example No. 55, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 82. The pharmaceutical combination of Embodiment 78, or for use of Embodiment 79, or the pharmaceutical composition of Embodiment 81, wherein the additional therapeutic agent is an anticancer agent.
351
Embodiment 83. The pharmaceutical combination of Embodiment 78, or for use of Embodiment 79, or the pharmaceutical composition of Embodiment 81, wherein the additionaî therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosîmilar thereof, panitumumab or a biosîmilar thereof, erlotinib, lapatinib, gefitinib, nivolumab or a biosîmilar thereof, pembrolizumab or a biosîmilar thereof, cemiplimab or a biosîmilar thereof, pidilizumab or a biosîmilar thereof, atezolizumab or a biosîmilar thereof, avelumab or a biosîmilar thereof, and durvalumab or a biosîmilar thereof.
Embodiment 84. A pharmaceutical combination which comprises (a) a compound of Example No. 58, or a pharmaceutically acceptable sait thereof, and (b) an additionaî therapeutic agent.
Embodiment 85. A pharmaceutical combination which comprises (a) a compound of Example No. 58, or a pharmaceutically acceptable sait thereof, and (b) an additionaî therapeutic agent, for use in therapy.
Embodiment 86. The pharmaceutical combination of Embodiment 84, or for use of Embodiment 85, wherein the compound of Example No. 58, or the pharmaceutically acceptable sait thereof and the additionaî therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 58, or a pharmaceutically acceptable sait thereof and of the additionaî therapeutic agent are together therapeutically effective.
Embodiment 87. A pharmaceutical composition, comprising (a) a compound of Example No. 58, or a pharmaceutically acceptable sait thereof, (b) an additionaî therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 88. The pharmaceutical combination of Embodiment 84, or for use of Embodiment 85, or the pharmaceutical composition of Embodiment 87, wherein the additionaî therapeutic agent is an anticancer agent.
352
Embodiment 89. The pharmaceutical combination of Embodiment 84, or for use of Embodiment 85, or the phannaceutical composition of Embodiment 87, wherein the additional therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, erlotinib, lapatinib, gefitinib, nivolumab or a biosimilar thereof, pembrolizumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 90. A pharmaceutical combination which comprises (a) a compound of Example No. 72, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 91. A pharmaceutical combination which comprises (a) a compound of Example No. 72, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 92, The pharmaceutical combination of Embodiment 90, or for use of Embodiment 91, wherein the compound of Example No. 72, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 72, or a pharmaceutically acceptable sait thereof and of the additional therapeutic agent are together therapeutically effective.
Embodiment 93. A pharmaceutical composition, comprising (a) a compound of Example No. 72, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 94. The pharmaceutical combination of Embodiment 90, or for use of Embodiment 91, or the pharmaceutical composition of Embodiment 93, wherein the additional therapeutic agent is an anticancer agent.
353
Embodiment 95. The pharmaceutical combination of Embodiment 90, or for use of Embodiment 91, or the pharmaceutical composition of Embodiment 93, wherein the additional therapeutic agent is selected from the group consistîng of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, erlotinib, lapatinib, gefïtinib, nivolumab or a biosimilar thereof, pembrolizumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 96. A pharmaceutical combination which comprises (a) a compound of Example No. 76, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 97. A pharmaceutical combination which comprises (a) a compound of Example No. 76, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 98. The pharmaceutical combination of Embodiment 96, or for use of Embodiment 97, wherein the compound of Example No. 76, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 76, or a pharmaceutically acceptable sait thereof and of the additional therapeutic agent are together therapeutically effective.
Embodiment 99. A pharmaceutical composition, comprising (a) a compound of Example No. 76, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 100. The pharmaceutical combination of Embodiment 96, or for use of Embodiment 97, or the pharmaceutical composition of Embodiment 99, wherein the additional therapeutic agent is an anticancer agent.
354
Embodiment 101. The pharmaceutical combination of Embodiment 96, or for use of Embodiment 97, or the pharmaceutical composition of Embodiment 99, wherein the additional therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinîb, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, erlotinib, lapatinib, gefitinib, nivolumab or a biosimilar thereof, pembrolizumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 102. A pharmaceutical combination which comprises (a) a compound of Example No. 77, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 103. A pharmaceutical combination which comprises (a) a compound of Example No. 77, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 104. The pharmaceutical combination of Embodiment 102, or for use of Embodiment 103, wherein the compound of Example No. 77, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 77, or a pharmaceutically acceptable sait thereof and of the additional therapeutic agent are together therapeutically effective.
Embodiment 105. A pharmaceutical composition, comprising (a) a compound of Example No. 77, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 106. The pharmaceutical combination of Embodiment 102, or for use of Embodiment 103, or the pharmaceutical composition of Embodiment 105, wherein the additional therapeutic agent is an anticancer agent.
355
Embodiment 107. The pharmaceutical combination of Embodiment 102, or for use of Embodiment 103, or the pharmaceutical composition of Embodiment 105, wherein the additional therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, erlotinib, lapatinib, gefitinib, nivolumab or a biosimilar thereof, pembrolizumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 108. A pharmaceutical combination which comprises (a) a compound of Example No. 78, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 109. A pharmaceutical combination which comprises (a) a compound of Example No. 78, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 110. The pharmaceutical combination of Embodiment 108, or for use of Embodiment 109, wherein the compound of Example No. 78, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequentiai use for use in therapy, wherein the amounts of the compound of Example No. 78, or a pharmaceutically acceptable sait thereof and of the additional therapeutic agent are together therapeutically effective.
Embodiment 1 i 1. A pharmaceutical composition, comprising (a) a compound of Example No. 78, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 112. The pharmaceutical combination of Embodiment 108, or for use of Embodiment 109, or the pharmaceutical composition of Embodiment 111, wherein the additional therapeutic agent is an anticancer agent.
356
Embodiment 113. The pharmaceutical combination of Embodiment 108, or for use of Embodiment 109, or the pharmaceutical composition of Embodiment 111, wherein the additional therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, erlotinib, lapatinib, gefitinib, nivolumab or a biosimilar thereof, pembrolizumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 114. A pharmaceutical combination which comprises (a) a compound of Example No. 83, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent .
Embodiment 115. A pharmaceutical combination which comprises (a) a compound of Example No. 83, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 116. The pharmaceutical combination of Embodiment 114, or for use of Embodiment 115, wherein the compound of Example No. 83, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 83, or a pharmaceutically acceptable sait thereof and of the additional therapeutic agent are together therapeutically effective.
Embodiment 117. A pharmaceutical composition, comprising (a) a compound of Example No. 83, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 118. The pharmaceutical combination of Embodiment 114, or for use of Embodiment 115, or the pharmaceutical composition of Embodiment 117, wherein the additional therapeutic agent is an anticancer agent.
357
Embodiment 119. The pharmaceutical combination of Embodiment 114, or for use of Embodiment 115, or the pharmaceutical composition of Embodiment 117, wherein the additional therapeutic agent is selected from the group consisting of bînimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, erlotinib, lapatinib, gefîtinib, nivolumab or a biosimilar thereof, pembrolizumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 120. A pharmaceutical combination which comprises (a) a compound of Example No. 84, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 121. A pharmaceutical combination which comprises (a) a compound of Example No. 84, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 122. The pharmaceutical combination of Embodiment 120, or for use of Embodiment 121, wherein the compound of Example No. 84, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 84, or a pharmaceutically acceptable sait thereof and of the additional therapeutic agent are together therapeutically effective.
Embodiment 123. A pharmaceutical composition, comprising (a) a compound of Example No. 84, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 124. The pharmaceutical combination of Embodiment 120, or for use of Embodiment 121, or the pharmaceutical composition of Embodiment 123, wherein the additional therapeutic agent is an anticancer agent.
358
Embodiment 125. The pharmaceutical combination of Embodiment 120, or for use of Embodiment 121, or the pharmaceutical composition of Embodiment 123, wherein the additionaî therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosîmilar thereof, panitumumab or a biosîmilar thereof, erlotinib, lapatinib, gefitinib, nivolumab or a biosîmilar thereof, pembrolizumab or a biosîmilar thereof, cemiplimab or a biosîmilar thereof, pidilizumab or a biosîmilar thereof, atezolizumab or a biosîmilar thereof, avelumab or a biosîmilar thereof, and durvalumab or a biosîmilar thereof.
Embodiment 126. A pharmaceutical combination which comprises (a) a compound of Example No. 85, or a pharmaceutically acceptable sait thereof, and (b) an additionaî therapeutic agent.
Embodiment 127. A pharmaceutical combination which comprises (a) a compound of Exampie No. 85, or a pharmaceutically acceptable sait thereof, and (b) an additionaî therapeutic agent, for use in therapy.
Embodiment 128. The pharmaceutical combination of Embodiment 126, or for use of Embodiment 127, wherein the compound of Exampie No. 85, or the pharmaceutically acceptable sait thereof and the additionaî therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 85, or a pharmaceutically acceptable sait thereof and of the additionaî therapeutic agent are together therapeutically effective.
Embodiment 129. A pharmaceutical composition, comprising (a) a compound of Example No. 85, or a pharmaceutically acceptable sait thereof, (b) an additionaî therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 130. The pharmaceutical combination of Embodiment 126, or for use of Embodiment 127, or the pharmaceutical composition of Embodiment 129, wherein the additionaî therapeutic agent is an anticancer agent.
359
Embodiment 131. The pharmaceutical combination of Embodiment 126, or for use of Embodiment 127, or the pharmaceutical composition of Embodiment 129, wherein the additional therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinib, sorafenib, trametînib, vemurafenib, cetuximab or a bîosimilar thereof, panitumumab or a bîosimilar thereof, erlotinib, lapatinib, gefitinib, nivolumab or a bîosimilar thereof, pembrolizumab or a bîosimilar thereof, cemiplîmab or a bîosimilar thereof, pidilizumab or a bîosimilar thereof, atezolizumab or a bîosimilar thereof, avelumab or a bîosimilar thereof, and durvalumab or a bîosimilar thereof.
Embodiment 132. A pharmaceutical combination which comprises (a) a compound of Example No. 91, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 133. A pharmaceutical combination which comprises (a) a compound of Example No. 91, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 134. The pharmaceutical combination of Embodiment 132, or for use of Embodiment 133, wherein the compound of Example No. 91, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 91, or a pharmaceutically acceptable sait thereof and of the additional therapeutic agent are together therapeutically effective.
Embodiment 135. A pharmaceutical composition, comprising (a) a compound of Example No. 91, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 136. The pharmaceutical combination of Embodiment 132, or for use of Embodiment 133, or the pharmaceutical composition of Embodiment 135, wherein the additional therapeutic agent is an anticancer agent.
360
Embodiment 137. The pharmaceutical combination of Embodiment 132, or for use of Embodiment 133, or the pharmaceutical composition of Embodiment 135, wherein the additional therapeutic agent îs selected from the group consisting of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, erlotinib, lapatinib, gefîtinib, nivolumab or a biosimilar thereof, pembrolizumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 138. A pharmaceutical combination which comprises (a) a compound ofExample No. 97, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 139. A pharmaceutical combination which comprises (a) a compound ofExample No. 97, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 140. The pharmaceutical combination of Embodiment 138, or for use of Embodiment 139, wherein the compound of Example No. 97, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound ofExample No. 97, or a pharmaceutically acceptable sait thereof and of the additional therapeutic agent are together therapeutically effective.
Embodiment 141. A pharmaceutical composition, comprising (a) a compound ofExample No. 97, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 142. The pharmaceutical combination of Embodiment 138, or for use of Embodiment 139, or the pharmaceutical composition of Embodiment 141, wherein the additional therapeutic agent is an anticancer agent.
36J
Embodiment 143. The pharmaceutical combination of Embodiment 138, or for use of Embodiment 139, or the pharmaceutical composition of Embodiment 141, wherein the additional therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, erlotinib, lapatinib, gefitinib, nivolumab or a biosimilar thereof, pembrolizumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 144. A pharmaceutical combination which comprises (a) a compound of Example No. 100, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 145. A pharmaceutical combination which comprises (a) a compound of Example No. 100, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 146. The pharmaceutical combination of Embodiment 144, or for use of Embodiment 145, wherein the compound of Example No. 100, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 100, or a pharmaceutically acceptable sait thereof and ofthe additional therapeutic agent are together therapeutically effective.
Embodiment 147. A pharmaceutical composition, comprising (a) a compound of Example No. 100, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 148. The pharmaceutical combination of Embodiment 144, or for use of Embodiment 145, or the pharmaceutical composition of Embodiment 147, wherein the additional therapeutic agent is an anticancer agent.
362
Embodiment 149. The pharmaceutical combination of Embodiment 144, or for use of Embodiment 145, or the pharmaceutical composition of Embodiment 147, wherein the additional therapeutic agent is selected from the group consistîng of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, erlotinib, lapatinib, gefïtinib, nivolumab or a biosimilar thereof, pembrolizumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 150. A pharmaceutical combination which comprises (a) a compound of Example No. 103, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 151. A pharmaceutical combination which comprises (a) a compound of Example No. 103, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 152. The pharmaceutical combination of Embodiment 150, or for use of Embodiment 151, wherein the compound of Example No. 103, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequentîal use for use in therapy, wherein the amounts of the compound of Example No. 103, or a pharmaceutically acceptable sait thereof and of the additional therapeutic agent are together therapeutically effective.
Embodiment 153. A pharmaceutical composition, comprising (a) a compound of Example No. 103, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 154. The pharmaceutical combination of Embodiment 150, or for use of Embodiment 151, or the pharmaceutical composition of Embodiment 153, wherein the additional therapeutic agent is an anticancer agent.
363
Embodiment 155. The pharmaceutical combination of Embodiment 150, or for use of Embodiment 151, or the pharmaceutical composition of Embodiment 153, wherein the additional therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinîb, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, erlotinib, lapatinib, gefitinib, nivolumab or a biosimilar thereof, pembrolizumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 156. A pharmaceutical combination which comprises (a) a compound of Example No. 105, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 157. A pharmaceutical combination which comprises (a) a compound of Example No. 105, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 158. The pharmaceutical combination of Embodiment 156, or for use of Embodiment 157, wherein the compound of Exampie No. 105, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 105, or a pharmaceutically acceptable sait thereof and of the additional therapeutic agent are together therapeutically effective.
Embodiment 159. A pharmaceutical composition, comprising (a) a compound of Example No. 105, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 160. The pharmaceutical combination of Embodiment 156, or for use of Embodiment 157, or the pharmaceutical composition of Embodiment 159, wherein the additional therapeutic agent is an anticancer agent.
364
Embodiment 16 1. The pharmaceutical combination of Embodiment 156, or for use of Embodiment 157, or the pharmaceutical composition of Embodiment 159, wherein the additional therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, erlotinib, lapatinib, gefitinib, nivolumab or a biosimilar thereof, pembrolizumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 162. A pharmaceutical combination which comprises (a) a compound of Example No. 107, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 163. A pharmaceutical combination which comprises (a) a compound of Example No. 107, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 164. The pharmaceutical combination of Embodiment 162, or for use of Embodiment 163, wherein the compound of Example No. 107, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequentiai use for use in therapy, wherein the amounts of the compound of Example No. 107, or a pharmaceutically acceptable sait thereof and of the additional therapeutic agent are together therapeutically effective.
Embodiment 165. A pharmaceutical composition, comprising (a) a compound of Example No. 107, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 166. The pharmaceutical combination of Embodiment 162, or for use of Embodiment 163, or the pharmaceutical composition of Embodiment 165, wherein the additional therapeutic agent is an anticancer agent.
365
Embodiment 167. The pharmaceutical combination of Embodiment 162, or for use of Embodiment 163, or the pharmaceutical composition of Embodiment 165, wherein the additional therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, erlotinib, lapatinib, gefitinib, nivolumab or a biosimilar thereof, pembrolizumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 168. A pharmaceutical combination which comprises (a) a compound of Example No. 108, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 169. A pharmaceutical combination which comprises (a) a compound of Example No. 108, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 170. The pharmaceutical combination of Embodiment 168, or for use of Embodiment 169, wherein the compound of Example No. 108, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 108, or a pharmaceutically acceptable sait thereof and ofthe additional therapeutic agent are together therapeutically effective.
Embodiment 171. A pharmaceutical composition, comprising (a) a compound of Example No. 108, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 172. The pharmaceutical combination of Embodiment 168, or for use of Embodiment 169, or the pharmaceutical composition of Embodiment 171, wherein the additional therapeutic agent is an anticancer agent.
366
Embodiment 173. The pharmaceutical combination of Embodiment 168, or for use of Embodiment 169, or the pharmaceutical composition of Embodiment 171, wherein the additionaî therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosîmilar thereof, erlotinib, lapatinib, gefitinib, nivolumab or a biosimilar thereof, pembrolizumab or a biosîmilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 174. A pharmaceutical combination which comprises (a) a compound of Example No. 114, or a pharmaceutically acceptable sait thereof, and (b) an additionaî therapeutic agent.
Embodiment 175. A pharmaceutical combination which comprises (a) a compound of Example No. 114, or a pharmaceutically acceptable sait thereof, and (b) an additionaî therapeutic agent, for use in therapy.
Embodiment 176. The pharmaceutical combination of Embodiment 174, or for use of Embodiment 175, wherein the compound of Example No. 114, or the pharmaceutically acceptable sait thereof and the additionaî therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts ofthe compound of Example No. 114, or a pharmaceutically acceptable sait thereofand ofthe additionaî therapeutic agent are together therapeutically effective.
Embodiment 177. A pharmaceutical composition, comprising (a) a compound of Example No. 114, or a pharmaceutically acceptable sait thereof, (b) an additionaî therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 178. The pharmaceutical combination of Embodiment 174, or for use of Embodiment 175, or the pharmaceutical composition of Embodiment 177, wherein the additionaî therapeutic agent is an anti cancer agent.
367
Embodiment 179. The pharmaceutical combination of Embodiment 174, or for use of Embodiment 175, or the pharmaceutical composition of Embodiment 177, wherein the additional therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a bîosimilar thereof, panitumumab or a bîosimilar thereof, erlotinib, lapatinib, gefitinib, nivolumab or a bîosimilar thereof, pembrolizumab or a bîosimilar thereof, cemiplîmab or a bîosimilar thereof, pidilizumab or a bîosimilar thereof, atezolizumab or a bîosimilar thereof, avelumab or a bîosimilar thereof, and durvalumab or a bîosimilar thereof.
Embodiment 180. A pharmaceutical combination which comprises (a) a compound of Example No. 115, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 181. A pharmaceutical combination which comprises (a) a compound of Example No. 115, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 182. The pharmaceutical combination of Embodiment 180, or for use of Embodiment 181, wherein the compound of Example No. 115, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 115, or a pharmaceutically acceptable sait thereof and of the additional therapeutic agent are together therapeutically effective.
Embodiment 183. A pharmaceutical composition, comprising (a) a compound of Example No. 115, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 184. The pharmaceutical combination of Embodiment 180, or for use of Embodiment 181, or the pharmaceutical composition of Embodiment 183, wherein the additional therapeutic agent is an anticancer agent.
368
Embodiment 185. The pharmaceutical combination of Embodiment 180, or for use of Embodiment 181, or the pharmaceutical composition of Embodiment 183, wherein the additional therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinib, sorafenib, trametînib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, erlotinib, lapatinib, gefitinib, nivolumab or a biosimilar thereof, pembrolizumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 186. A pharmaceutical combination which comprises (a) a compound of Example No. 119, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 187. A pharmaceutical combination which comprises (a) a compound of Example No. 119, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 188. The pharmaceutical combination of Embodiment 186, or for use of Embodiment 187, wherein the compound of Example No. 119, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 119, or a pharmaceutically acceptable sait thereof and of the additional therapeutic agent are together therapeutically effective.
Embodiment 189. A pharmaceutical composition, comprising (a) a compound of Example No. 119, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 190. The pharmaceutical combination of Embodiment 186, or for use of Embodiment 187, or the pharmaceutical composition of Embodiment 189, wherein the additional therapeutic agent is an anticancer agent.
369
Embodiment 191. The pharmaceutical combination of Embodiment 186, or for use of Embodiment 187, or the pharmaceutical composition of Embodiment 189, wherein the additional therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, erlotinib, lapatinib, gefîtinib, nivolumab or a biosimilar thereof, pembrolizumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 192. A pharmaceutical combination which comprises (a) a compound ofExample No. 121, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 193. A pharmaceutical combination which comprises (a) a compound of Example No. 121, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 194. The pharmaceutical combination of Embodiment 192, or for use of Embodiment 193, wherein the compound ofExample No. 121, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound ofExample No. 121, or a pharmaceutically acceptable sait thereof and of the additional therapeutic agent are together therapeutically effective.
Embodiment 195. A pharmaceutical composition, comprising (a) a compound of Example No. 121, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 196. The pharmaceutical combination of Embodiment 192, or for use of Embodiment 193, or the pharmaceutical composition of Embodiment 195, wherein the additional therapeutic agent is an anticancer agent.
370
Embodiment 197. The pharmaceutical combination of Embodiment 192, or for use of Embodiment 193, or the pharmaceutical composition of Embodiment 195, wherein the additional therapeutic agent is selected from the group consistîng of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, 5 erlotinib, lapatinib, gefïtinib, nivolumab or a biosimilar thereof, pembrolizumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 198. A pharmaceutical combination which comprises (a) a compound of Example 10 No. 124, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 199. A pharmaceutical combination which comprises (a) a compound of Example No. 124, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 200. The pharmaceutical combination of Embodiment 198, or for use of Embodiment 199, wherein the compound of Example No. 124, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound 2 0 of Example No. 124, or a pharmaceutically acceptable sait thereof and ofthe additional therapeutic agent are together therapeutically effective.
Embodiment 201. A pharmaceutical composition, comprising (a) a compound of Example No.
124, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a 25 pharmaceutically acceptable diluent or carrier.
Embodiment 202. The pharmaceutical combination of Embodiment 198, or for use of Embodiment 199, or the pharmaceutical composition of Embodiment 201, wherein the additional therapeutic agent is an anticancer agent.
371
Embodiment 203. The pharmaceutical combination of Embodiment 198, or for use of Embodiment 199, or the pharmaceutical composition of Embodiment 201, wherein the additional therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, erlotinib, lapatinib, gefitinib, nivolumab or a biosimilar thereof, pembrolizumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 204. A pharmaceutical combination which comprises (a) a compound of Example No. 125, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 205. A pharmaceutical combination which comprises (a) a compound of Example No. 125, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use în therapy.
Embodiment 206. The pharmaceutical combination of Embodiment 204, or for use of Embodiment 205, wherein the compound of Example No. 125, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 125, or a pharmaceutically acceptable sait thereof and ofthe additional therapeutic agent are together therapeutically effective.
Embodiment 207. A pharmaceutical composition, comprising (a) a compound of Example No. 125, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 208. The pharmaceutical combination of Embodiment 204, or for use of Embodiment 205, or the pharmaceutical composition of Embodiment 207, wherein the additional therapeutic agent is an anticancer agent.
372
Embodiment 209. The phannaceutical combination of Embodiment 204, or for use of Embodiment 205, or the pharmaceutical composition of Embodiment 207, wherein the additional therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, erlotinib, lapatinib, gefitinib, nivolumab or a biosimilar thereof, pembrolîzumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, aveiumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 210. A pharmaceutical combination which comprises (a) a compound of Example No. 126, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 211. A pharmaceutical combination which comprises (a) a compound of Example No. 126, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 212. The pharmaceutical combination of Embodiment 210, or for use of Embodiment 211, wherein the compound of Example No. 126, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 126, or a pharmaceutically acceptable sait thereof and ofthe additional therapeutic agent are together therapeutically effective.
Embodiment 213. A pharmaceutical composition, comprising (a) a compound of Example No. 126, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 214. The pharmaceutical combination of Embodiment 210, or for use of Embodiment 211, or the pharmaceutical composition of Embodiment 213, wherein the additional therapeutic agent is an anticancer agent.
373
Embodiment 215. The pharmaceutical combination of Embodiment 210, or for use of Embodiment 211, or the pharmaceutical composition of Embodiment 213, wherein the additional therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, erlotinib, lapatinib, gefitinib, nivolumab or a biosimilar thereof, pembrolizumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 216. A pharmaceutical combination which comprises (a) a compound of Example No. 127, or a pharmaceuticaliy acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 217. A pharmaceutical combination which comprises (a) a compound of Example No. 127, or a pharmaceuticaliy acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 218. The pharmaceutical combination of Embodiment 216, or for use of Embodiment 217, wherein the compound of Example No. 127, or the pharmaceuticaliy acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 127, or a pharmaceuticaliy acceptable sait thereof and ofthe additional therapeutic agent are together therapeutically effective.
Embodiment 219. A pharmaceutical composition, comprising (a) a compound of Example No. 127, or a pharmaceuticaliy acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceuticaliy acceptable diluent or carrier.
Embodiment 220. The pharmaceutical combination of Embodiment 216, or for use of Embodiment 217, or the pharmaceutical composition of Embodiment 219, wherein the additional therapeutic agent is an anticancer agent.
374
Embodiment 221. The pharmaceutical combination of Embodiment 216, or for use of Embodiment 217, or the pharmaceutical composition of Embodiment 219, wherein the additional therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, erlotinib, lapatinib, gefitinib, nivolumab or a biosimilar thereof, pembrolizumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 222. A pharmaceutical combination which comprises (a) a compound of Example No. 129, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 223. A pharmaceutical combination which comprises (a) a compound of Example No. 129, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 224. The pharmaceutical combination of Embodiment 222, or for use of Embodiment 223, wherein the compound of Example No. 129, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 129, or a pharmaceutically acceptable sait thereof and of the additional therapeutic agent are together therapeutically effective.
Embodiment 225. A pharmaceutical composition, comprising (a) a compound of Example No. 129, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 226. The pharmaceutical combination of Embodiment 222, or for use of Embodiment 223, or the pharmaceutical composition of Embodiment 225, wherein the additional therapeutic agent is an anticancer agent.
375
Embodiment 227. The pharmaceutical combination of Embodiment 222, or for use of Embodiment 223, or the pharmaceutical composition of Embodiment 225, wherein the additional therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, erlotinib, lapatinib, gefitinib, nivolumab or a biosimilar thereof, pembrolizumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 228. A pharmaceutical combination which comprises (a) a compound of Example No. 151, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 229. A pharmaceutical combination which comprises (a) a compound of Example No. 151, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 230. The pharmaceutical combination of Embodiment 228, or for use of Embodiment 229, wherein the compound of Example No. 151, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 151, or a pharmaceutically acceptable sait thereof and of the additional therapeutic agent are together therapeutically effective.
Embodiment 231. A pharmaceutical composition, comprising (a) a compound of Example No. 151, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 232. The pharmaceutical combination of Embodiment 228, or for use of Embodiment 229, or the pharmaceutical composition of Embodiment 231, wherein the additional therapeutic agent is an anticancer agent.
376
Embodiment 233, The pharmaceutical combination of Embodiment 228, or for use of Embodiment 229, or the pharmaceutical composition of Embodiment 231, wherein the additional therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinîb, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, erlotinib, lapatinib, gefitinib, nivolumab or a biosimilar thereof, pembrolizumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof,
Embodiment 234. A pharmaceutical combination which comprises (a) a compound of Example No. 1 52, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent .
Embodiment 235. A pharmaceutical combination which comprises (a) a compound of Exampie No. 152, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 236. The pharmaceutical combination of Embodiment 234, or for use of Embodiment 235, wherein the compound of Example No. 152, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 152, or a pharmaceutically acceptable sait thereof and ofthe additional therapeutic agent are together therapeutically effective.
Embodiment 237. A pharmaceutical composition, comprising (a) a compound of Example No. 152, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 238. The pharmaceutical combination of Embodiment 234, or for use of Embodiment 235, or the pharmaceutical composition of Embodiment 237, wherein the additional therapeutic agent is an anticancer agent.
377
Embodiment 239. The pharmaceutical combination of Embodiment 234, or for use of Embodiment 235, or the pharmaceutical composition of Embodiment 237, wherein the additionaî therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, erlotinib, lapatinib, gefitinib, nivolumab or a biosimilar thereof, pembrolizumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 240. A pharmaceutical combination which comprises (a) a compound of Example No. 163, or a pharmaceutically acceptable sait thereof, and (b) an additionaî therapeutic agent.
Embodiment 241. A pharmaceutical combination which comprises (a) a compound of Exampie No. 163, or a pharmaceutically acceptable sait thereof, and (b) an additionaî therapeutic agent, for use in therapy.
Embodiment 242. The pharmaceutical combination of Embodiment 240, or for use of Embodiment 241, wherein the compound of Example No. 163, or the pharmaceutically acceptable sait thereof and the additionaî therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 163, or a pharmaceutically acceptable sait thereof and ofthe additionaî therapeutic agent are together therapeutically effective.
Embodiment 243. A pharmaceutical composition, comprising (a) a compound of Example No. 163, or a pharmaceutically acceptable sait thereof, (b) an additionaî therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 244. The pharmaceutical combination of Embodiment 240, or for use of Embodiment 241, or the pharmaceutical composition of Embodiment 243, wherein the additionaî therapeutic agent is an anticancer agent.
378
Embodiment 245. The pharmaceutical combination of Embodiment 240, or for use of Embodiment 241, or the pharmaceutical composition of Embodiment 243, wherein the additional therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a bîosimilar thereof, panitumumab or a bîosimilar thereof, erlotinib, lapatinib, gefitinib, nivolumab or a bîosimilar thereof, pembrolizumab or a bîosimilar thereof, cemiplîmab or a bîosimilar thereof, pidilizumab or a bîosimilar thereof, atezolizumab or a bîosimilar thereof, avelumab or a bîosimilar thereof, and durvalumab or a bîosimilar thereof.
Embodiment 246. A pharmaceutical combination which comprises (a) a compound of Example No. 169, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 247. A pharmaceutical combination which comprises (a) a compound of Example No. 169, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 248. The pharmaceutical combination of Embodiment 246, or for use of Embodiment 247, wherein the compound of Example No. 169, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 169, or a pharmaceutically acceptable sait thereof and ofthe additional therapeutic agent are together therapeutically effective.
Embodiment 249. A pharmaceutical composition, comprising (a) a compound of Example No. 169, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 250. The pharmaceutical combination of Embodiment 246, or for use of Embodiment 247, or the pharmaceutical composition of Embodiment 249, wherein the additional therapeutic agent is an anticancer agent.
379
Embodiment 251. The pharmaceutical combination of Embodiment 246, or for use of Embodiment 247, or the pharmaceutical composition of Embodiment 249, wherein the additional therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, erlotinib, lapatinib, gefîtinib, nivolumab or a biosimilar thereof, pembrolizumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 252. A pharmaceutical combination which comprises (a) a compound ofExample No. 188, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 253. A pharmaceutical combination which comprises (a) a compound of Example No. 188, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 254. The pharmaceutical combination of Embodiment 252, or for use of Embodiment 253, wherein the compound ofExample No. 188, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 188, or a pharmaceutically acceptable sait thereof and of the additional therapeutic agent are together therapeutically effective.
Embodiment 255. A pharmaceutical composition, comprising (a) a compound of Example No. 188, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 256. The pharmaceutical combination of Embodiment 252, or for use of Embodiment 253, or the pharmaceutical composition of Embodiment 255, wherein the additional therapeutic agent is an anticancer agent.
380
Embodiment 257. The pharmaceutical combination of Embodiment 252, or for use of Embodiment 253, or the pharmaceutical composition of Embodiment 255, wherein the additional therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, eriotinib, lapatinib, gefitinib, nivolumab or a biosimilar thereof, pembrolizumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 258. A pharmaceutical combination which comprises (a) a compound of Example No. 190, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 259. A pharmaceutical combination which comprises (a) a compound of Example No. 190, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 260. The pharmaceutical combination of Embodiment 258, or for use of Embodiment 259, wherein the compound of Example No. 190, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 190, or a pharmaceutically acceptable sait thereof and of the additional therapeutic agent are together therapeutically effective.
Embodiment 261. A pharmaceutical composition, comprising (a) a compound of Example No. 190, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 262. The pharmaceutical combination of Embodiment 258, or for use of Embodiment 259, or the pharmaceutical composition of Embodiment 26i, wherein the additional therapeutic agent is an anticancer agent.
381
Embodiment 263. The pharmaceutical combination of Embodiment 258, or for use of Embodiment 259, or the pharmaceutical composition of Embodiment 261, wherein the additional therapeutic agent is selected from the group consistîng of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, erlotinib, lapatinib, gefïtinib, nivolumab or a biosimilar thereof, pembrolizumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 264. A pharmaceutical combination which comprises (a) a compound of Example No. 199, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 265. A pharmaceutical combination which comprises (a) a compound of Example No. 199, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 266. The pharmaceutical combination of Embodiment 264, or for use of Embodiment 265, wherein the compound of Example No. 199, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 199, or a pharmaceutically acceptable sait thereof and of the additional therapeutic agent are together therapeutically effective.
Embodiment 267. A pharmaceutical composition, comprising (a) a compound of Example No. 199, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 268. The pharmaceutical combination of Embodiment 264, or for use of Embodiment 265, or the pharmaceutical composition of Embodiment 267, wherein the additional therapeutic agent is an anticancer agent.
382
Embodiment 269. The pharmaceutical combination of Embodiment 264, or for use of Embodiment 265, or the pharmaceutical composition of Embodiment 267, wherein the additional therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, erlotinib, lapatinib, gefitinib, nivolumab or a biosimilar thereof, pembrolizumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 270. A pharmaceutical combination which comprises (a) a compound of Example No. 200, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 271. A pharmaceutical combination which comprises (a) a compound of Example No. 200, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 272. The pharmaceutical combination of Embodiment 270, or for use of Embodiment 271, wherein the compound of Example No. 200, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 200, or a pharmaceutically acceptable sait thereof and ofthe additional therapeutic agent are together therapeutically effective.
Embodiment 273. A pharmaceutical composition, comprising (a) a compound of Example No. 200, or a pharmaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 274. The pharmaceutical combination of Embodiment 270, or for use of Embodiment 271, or the pharmaceutical composition of Embodiment 273, wherein the additional therapeutic agent is an anticancer agent.
383
Embodiment 275. The pharmaceutical combination of Embodiment 270, or for use of Embodiment 271, or the pharmaceutical composition of Embodiment 273, wherein the additional therapeutic agent is selected from the group consisting of binimetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, erlotinib, lapatinib, gefltinib, nivolumab or a biosimilar thereof, pembrolîzumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.
Embodiment 276. A pharmaceutical combination which comprises (a) a compound of Example No. 201, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent.
Embodiment 277. A pharmaceutical combination which comprises (a) a compound of Example No. 201, or a pharmaceutically acceptable sait thereof, and (b) an additional therapeutic agent, for use in therapy.
Embodiment 278. The pharmaceutical combination of Embodiment 276, or for use of Embodiment 277, wherein the compound of Example No. 201, or the pharmaceutically acceptable sait thereof and the additional therapeutic agent are formulated as separate compositions or dosages for simultaneous, separate or sequential use for use in therapy, wherein the amounts of the compound of Example No. 201, or a pharmaceutically acceptable sait thereof and ofthe additional therapeutic agent are together therapeutically effective.
Embodiment 279. A pharmaceutical composition, comprising (a) a compound of Example No. 201, or a phannaceutically acceptable sait thereof, (b) an additional therapeutic agent, and (c) a pharmaceutically acceptable diluent or carrier.
Embodiment 280. The phannaceutical combination of Embodiment 276, or for use of Embodiment 277, or the pharmaceutical composition of Embodiment 279, wherein the additional therapeutic agent is an anticancer agent.
384
Embodiment 281. The pharmaceutical combination of Embodiment 276, or for use of Embodiment 277, or the pharmaceutical composition of Embodiment 279, wherein the additional therapeutic agent is selected from the group consisting of biniinetinib, encorafenib, selumetinib, sorafenib, trametinib, vemurafenib, cetuximab or a biosimilar thereof, panitumumab or a biosimilar thereof, 5 erlotinib, lapatinib, gefitinib, nivolumab or a biosimilar thereof, pembrolizumab or a biosimilar thereof, cemiplimab or a biosimilar thereof, pidilizumab or a biosimilar thereof, atezolizumab or a biosimilar thereof, avelumab or a biosimilar thereof, and durvalumab or a biosimilar thereof.

Claims (22)

1. A compound of Formula I
I and stéréo isomers, tautomers and pharmaceutically acceptable salts thereof, wherein:
X1 isCHorN;
R1 is hydrogen or C1-C6 alkyl;
R2 is (a) hydrogen, (b)Cl-C6 alkyl, (c) hydroxyCl-C6 alkyl, (d) dihydroxyC2-C6 alkyl, (e) CI-C6 fluoroalkyl optionally substituted with OH, (f) (di-C 1-C6 alkoxy)C2-C6 alkyl-, (g) (CI-C6 alkoxy)Cl-C6 alkyl- wherein said alkyl portion is optionally substituted with OH, (h) Cyc1, (i) Cyc2, (j) (hetCyc!)Cl-C6 alkyl- wherein said alkyl portion is optionally substituted with OH, (k) (Ar')C 1-C6 alkyl- wherein said alkyl portion is optionally substituted with OH, (1) (hetAr')Cl-C6 alkyl- wherein said alkyl portion is optionally substituted with OH, or (m) (HOSO3)C1-C6 alkyl-;
Cyc1 is a 3-4 membered cycloalkyl ring optionally substituted with 1-2 substituents independently selected from halogen, hydroxy, CI-C3 alkyl, hydroxyC 1-C3 alkyl, C1-C3 alkoxy,
386 (C1-C3 alkoxy)Cl-C3 alkyl- and R'RNC(=O)-;
R* and R are independentiy hydrogen or C1-C6 alkyl;
Cyc2 is a 5-membered cycloalkyl ring substituted with 1-2 substituents independentiy selected from C1-C3 alkyl, (C1-C3 alkoxy)Cl-C3 alkyl- and hydroxyCl-C3 alkyl-;
hetCyc1 is a 5-6 membered saturated heterocyclic ring having 1-2 ring heteroatoms independentiy selected from O, N, S and SO2, wherein said ring is optionally substituted with oxo;
Ar1 is phenyl;
hetAr1 is pyridyl;
G is
O or θ
X2 is C or N;
Ring A, including the atoms at the points of attachment, is a 5-6 membered heterocyclic ring optionally having an additional 1-2 ring nitrogen atoms when X2 is N and having one ring nitrogen atom when X2 is C;
R3 is hydrogen, methyl, or absent;
R6 is hydrogen, halogen, C1-C6 alkyl, C1-C6 alkoxy, hydroxyCl-C6 alkyl, C3-C6 cycloalkyl, (C3-C6 cycloaikyl)Cl-C6 alkyl- or hetCyc2, provided that when R6 is halogen and is on the ring carbon atom adjacent to the carbon linked to the -NHC(=O)- moiety of Formula I, then R6 is not halogen, and
R7 is hydrogen, C1-C6 alkyl, oxo or thioxo, or optionally when R6 and R7 are on the same carbon atom, R6 and R7 together with the carbon atom to which they are attached form a cyclopropyl ring;
hetCyc2 is a 4-6 membered saturated heterocyclic ring having a ring nitrogen atom and optionally substituted with C1-C6 alkyl;
Ring B, including the atoms at the points of attachment, is a 6-membered saturated carbocyclic optionally substituted with oxo or a 6-membered aromatic carbocyclic ring optionally
387 substituted with OH;
R8 is Ar2, hetAr, C3-C6 cycloalkyl, hetCyc3 or C1-C6 alkyl;
Ar2 is phenyl optionally substituted with one or more substituents independently selected from halogen, C1-C2 alkyl and C1-C2 alkoxy;
hetAr2 is a 5-6 membered heteroaryl having 1-2 ring nitrogen atoms and optionally substituted with one or more substituents independently selected from halogen, C1-C2 alkyl and CI-C2 alkoxy;
hetCyc3 is a 5-6 membered heterocyclic ring having a ring oxygen atom;
and
R9 is hydrogen or halogen.
2. A compound of claim 1 or a stereoisomer, tautomer or pharmaceutically acceptable sait thereof, wherein G is
3. A compound of claim 2 or a stereoisomer, tautomer or pharmaceutically acceptable sait thereof, wherein X2 is N, R3 is absent, Ring A is a 6-membered heterocyclic ring having one additionaî ring nitrogen atom, and R7 is oxo, wherein G has the formula A-l and wherein R6 is C1-C6 alkyl, hydroxyCl-C6 alkyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)ClC6 alkyl- or hetCyc2.
388
4. A compound according to any one of daims 1-3 or a stereoisomer, tautomer or pharmaceuticaliy acceptable sait thereof, wherein R2 is hydroxyCl-C6 alkyl.
5. A compound according to any one of daims 1-4 or a stereoisomer, tautomer or pharmaceuticaliy acceptable sait thereof, wherein R8 is Ar2.
6. A compound according to daim 5 or a stereoisomer, tautomer or pharmaceuticaliy acceptable sait thereof, wherein Ar2 is phenyl optionally substituted with one or more halogens.
7. A compound according to any one of daims 1-3 or a stereoisomer, tautomer or pharmaceuticaliy acceptable sait thereof, wherein X1 is CH.
8. A compound according to any one of daims 1-3 or a stereoisomer, tautomer or pharmaceuticaliy acceptable sait thereof, wherein R9 is halogen.
9. A compound according to daim 8 or a stereoisomer, tautomer or pharmaceuticaliy acceptable sait thereof, wherein R9 is fluoro.
10. A compound according to any one of daims 1 -9 or a stereoisomer, tautomer or pharmaceuticaliy acceptable sait thereof, wherein R1 is hydrogen.
11. A compound according to daim 1, selected from
N-(3-fIuoro-4-((3-((2-liydroxyethyi)amino)-l H-pyrazolo[3,4-b]pyridin-4“yl)oxy)phenyi)2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(3-fluoro-4-((3-((2-methoxyethyl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(3-fluoro-4-((3-((l-hydroxy-2-methylpropan-2-yl)amino)-IH-pyrazolo[3,4~b]pyridin4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(3-fluoro-4-((3-((2-hydroxy-2-methylpropyl)amino)-lH-pyrazolo[3,4-b]pyrîdin-4yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
389 (R)-N-(3-fluoro-4-((3-((2-hydroxypropyl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)pheny[)-2-(4-fluorophenyl)-3-oxo-2,3-dihydiOpyridazine-4-carboxamide;
(S)-N-(3-fluoro-4-((3-((2-hydroxypropyl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyI)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyrÎdazine-4-carboxamide;
(S)-N-(3-fluoro-4-((3-((l -hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
(R)'N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4’b]pyridin-4yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(3-fluoro-4-((3-((3-hydroxy-2,2-dimethylpropyl)amino)-lH-pyrazolo[3f4-b]pyridin-4yl)oxy)phenyl)-2-(4-fluorophenyî)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
(S)-N-(4-((3-((2,3-dihydroxypropyl)amino)-lH-pyrazolo[3,4-b]pyridin-4-yl)oxy)-3fluoropheny[)'2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(4-((3-(cyclobutylamino)-lH-pyrazolo[3,4-b]pyridin-4-yl)oxy)-3-fluorophenyl)-2-(4fluorophenyl)-3-oxo-253-dihydropyridazine-4-carboxamide;
N-(4-((3-((3;3-dîfluorocyclobutyl)ainino)-lH-pyrazolo[3f4-b]pyridin-4-yI)oxy)-3fluorophenyl)-2-(4-fluorophenyI)-3'Oxo-2,3-dihydropyridazine-4-carboxamide;
(R)-N-(4-((3-((2,3-dihydroxypropyl)amino)-1H-pyrazolo[3,4-b]pyrÎdin-4-yl)oxy)-3fluorophenyl)-2-(4-fluorophenyI)“3“Oxo-23-dihydropyrîdazine-4-carboxamide;
N-(3-fluoro-4-((3-(((l r,3r)-3-rnethoxycyclobutyl)arnino)-] H-pyrazo!o[3,4-b]pyridin-4yl)oxy)phenyl)-2-(4-fluorophenyÎ)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(3-nuoro-4-((3-(((ls,3s)-3-methoxycyclobutyl)amino)-l H-pyrazolo[3,4-b]pyridîn-4yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(3-f1uoro-4-((3-(((ls,3s)-3-hydroxy-l-methylcyclobutyl)amino)-lH-pyrazolo[354b]pyridin'4-yl)oxy)phenyl)-2-(4-fluoiOphenyl)-3-oxo-2,3-dihydropyridazine~4-carboxaniide;
N-(3-fluoro-4-((3-((l-(methoxymethyl)cyclopropyl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyJ)-2-(4-tluoropheny 103-0X0-2,3-dihydropyridazine-4-carboxamide;
N'(3-fluoro-4-((3-((l-methoxy-2-methylpropan-2-yl)amino)-l H-pyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(3-f]uoiO-4-((3-((2-inethoxyethyl)(methyl)amino)-]H-pyrazoIo[3,4-b]pyridin-4yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(3-fluoro-4-((3-((l-(methoxymethyl)cyclopentyl)amino)-lH-pyrazolo[3f4-b]pyridin-420534
390 yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2;3-dihydropyridazine-4-carboxamide;
N-(3-fluoro-4-((3-((l-(methoxyniethyl)cyclobutyl)amino)-]H-pyrazolo[3,4-b]pyndin-4yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(3-fluoro-4-((3-((4-methoxy-2-methylbutan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-45 yl)oxy)phenyl)-2-(4-flLiorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(3-fluoro-4-((3-(((tetrahydrofuran-2-yl)methyl)amino)-l H-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-çarboxamide;
N-(4-((3-((( 1, l -dioxidotetrahydrothiophen-3-yl)methyi)amino)-l I-l-pyrazolo[3,4b]pyridin-4-yl)oxy)-3-fluorophenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-410 carboxamide;
(S)-N-(3-fluoro-4-((3-((l-methoxypropan-2-y[)amino)-IH-pyrazolo[3,4-b]pyridin-4yl)oxy)pheny 1)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(4-((3-((l,3-dimethoxypropan-2-yl)amino)-lH-pyrazolo[3f4-b]pyridin-4-yl)oxy)-3fluorophenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
15 N-(4-((3-((4,4-difluorobutan-2-y l)amino)-1 H-pyrazolo[3,4-b]pyridin-4-yl)oxy)-3fluorophenyl)-2-(4-iluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(3-fluoro-4-((3-((cis-2-(methoxymethyl)cyclopentyl)amino)-IH-pyrazolo[3,4b]pyridin-4-yl)oxy)pheny 1)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(3-fluoro-4-((3-(((tetrahydrofuran-3-yl)methyl)amino)-lH-pyrazolo[3,4-b]pyridin-420 yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(3-fluoro-4-((3-(((2'OXopiperidin-4-yl)methyl)ammo)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(4-((3-(((1 s,3s)-3-(dimethylcarbamoyl)cyclobuty l)amino)-l H-pyrazolo[3,4-b]pyrid in4-yl)oxy)-3-fluorophenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
2 5 N-(4-((3-((l;3-dihydroxy-2-methyipropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)-3-fluorophenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(3-fluoro-4-((3-((4-hydroxy-2-methylbutan-2-yl)amino)-l H-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyndazine-4-carboxamide;
N-(3-fluoro-4-((3-((3-hydroxy-3-methylbulan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-430 yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(3-fluoro-4-((3-((2-hydroxyethyl)(methyl)amino)-lH-pyrazolo[3,4-b]pyndin-420534
391 yi)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dthydropyridazine-4-carboxam!de;
N-(3-fluoro-4-((3-((( lr,3r)-3-(hydroxymethyl)cyclobutyl)amino)-lH-pyrazolo[3,4b]pyridin-4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(3-fluoro-4-((3-(((ls,3s)-3-(hydroxymethyl)cyclobuty])amino)-lH-pyrazolo[3,4h]pyridin-4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(3-fluoro-4-((3-((3-hydroxy-2-methylbutan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(3-fluoro-4-((3-((3-hydroxy-2,3-dimethylbutan-2-yl)amino)-l H-pyrazolo[3,4b]pyrjdin-4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(3-fluoro-4-((3-((4-hydroxy-2-methyipentan-2-yl)amino)-iH-pyrazoÎo[3,4-b]pyridin4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
(R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yt)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-2-(4-fluorophenyl)-6-methyl-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(3 -fluoro-4-((3 -(( 1 -hyd roxy-2-methy I propan-2-y l)am ino)-1H -py razolo [3,4-b] py rid i n 4-yl)oxy)phenyl)-2-(4-fluorophenyl)-6-methyl-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N -(3 -fl uoro-4-((3 -((4-hy d roxy -1 -methoxy-2-methy lbutan-2 -y i )am ino)-1H -pyrazol o[3,4b]pyridin-4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
(S)-N-(3-fluoro-4-((3-((3,3,3-trifluoro-2-hydroxypropyl)amino)-lH-pyrazolo[3,4b]pyridin-4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(4-((3-(( l,3-dihydroxypropan-2-y l)amino)-lH-pyrazolo[3,4-b]pyridîn-4-yl)oxy)-3fluoropheny 1)-2-(4-fluoropheny l)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(3-fluoro-4-((3-((l-hydroxy-3-methoxypropan-2-yl)amino)-ÎH-pyrazolo[3,4b]pyridin-4-yl)oxy)pheny 1)-2-(4-fluoropheny l)-3 -oxo-2,3-dihydropyridazine-4-carboxamide;
N-(3-fluoro-4-((3-((l-hydroxy-3-methoxy-2-methylpropan-2-yl)ainino)-lH-pyrazolo[3,4b]pyridin-4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3 -oxo-2,3-dihydropyridazine-4-carboxamide;
N-(3-fluoro-4-((3-((4-hydroxy-l-methoxybutan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-2-(4-fluoropheny l)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(3-fluoro-4-((3-((l-hydroxy-4-methoxybutan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(4-((3-((2,2-difluoro-3-hydroxypropyl)amÎno)-IH-pyrazolo[3,4-b]pyridin-4-yl)oxy)-3fluorophenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
392
N-(4-((3-((454-difluoro-l-hydroxybutan-2-yl)amino)-lH-pyrazolo[3.,4-b]pyridin-4yl)oxy)-3-fluorophenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(3-fluoro-4-((3-((4,4,4-trifluoro-l-hydroxybutan-2-yl)amino)-lH-pyrazolo[3,4b]pyrîdin-4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazîne-4-carboxamide;
N-(3-fluorO'4-((3-((l-(hydroxymethyl)cyclobuty])amino)-lH-pyrazolo[3,4-b]pyridin-4y[)oxy)pbenyl)-2-(4-fluorophenyl)-3-oxo-2h3-dihydropyridazine-4-carboxamide;
N-(3-fluoro-4-((3-(((lS52R)-2-(hydroxymethyl)-l-methyicyciopentyl)amino)-lHpyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4carboxamide;
(R)-N-(3-fluoro-4-((3-((l-hydroxybutan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2i3-dihydropyrîdazine-4-carboxamide;
(S)-N-(3-fluoro-4-((3-((l-hydroxybutan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4yî)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(3-fluoro-4-((3-((l-(hydroxymethyl)cyclopropyl)amino)-lH-pyrazoio[3,4-b]pyridin-4yl)oxy)phenyl)-2-(4-fluorophenyl)’3-oxo-2J3’dihydropyridazine-4-carboxamide;
N-(3-fluoro-4-((3-((cis-2-(hydroxymethyl)cyclopentyl)amino)-l H-pyrazolo[3,4b]pyridin-4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyrîdazine-4-carboxamide;
N-(3-6uoro-4-((3-((l -(hydroxymethyl)cyclopentyl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3'dihydropyridazine-4-carboxamide;
(R)-N-(3-fliioro-4-((3-((2-hydroxy-l-phenylethyl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyi)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(3-fluoro-4-((3-((l-hydroxy-2-(hydroxymethyl)butan-2-yl)amino)-tH-pyrazolo[3,4b]pyridin-4-y!)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
bl-(3-fluoiO-4-((3-((2-hydroxy-l-(pyridin-3-yl)ethyl)amino)-lH-pyrazolo[354-b]pyndin4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3“Oxo-2}3-dihydropyridazine-4-carboxamide;
N-(3-f]uorO'4-((3-((2-hydroxy-l-(pyridin-4-yl)ethyl)amino)-lH-pyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)'2-(4-fluorophenyl)-3-oxo-2,3-dihydropyndazine-4-carboxamide;
N-(3-fluoro-4-((3-((2-hydroxy-l-(tetrahydro-2H-pyran-4-yl)ethy!)amino)-1 Hpyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4carboxamide;
(R)-N-(3-fluoro-4-((3-(( l-hydroxypropan-2-y l)amino)-lH-pyrazolo[3,4-b]pyridin-420534
393 y l)oxy)pheny 1)-2,5-dioxo-l -phenyi-1,2,5,6,7,8-hexahydroquinoiine-3-carboxamide;
N-(3-fluoro-4-((3-iodo-l-(4-methoxybenzyl)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-l-(4-fluorophenyl)-2,5-dioxo-l,2,5,6,7,8-hexahydroquinoline-3-carboxamide;
(R.)-N-(3-fluoro-4-((3-((l -hydroxybutan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-45 yl)oxy)phenyl)-3-(4-fluorophenyl)-l-isopropyl-2,4-dioxo-1,2,3,4-tetrahydropyrimidine-5carboxamide;
(S)-N-(3-fluoro-4-((3-((l-hydroxybutan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)pheny 1)-3-(4-fluorophenyl)-!-isopropyl-2,4-dioxo-l,2,3,4-tetrahydropyrimidine-5carboxamide;
10 N-(3-fluoro-4-((3-((l -(hydroxymethyl)cyclobutyl)amino)-1 H-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-3-(4-fluorophenyl)-l-isopropyl-2,4-dioxo-l,2,3,4-tetrahydropyrimidine-5carboxamide;
N-(3-fluoro-4-((3-((l-hydroxy-2-methylpropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin4-yl)oxy)pheny 1)-3-(4-fluorophenyl)-!-isopropyl-2,4-dioxo-1,2,3,4-tetrahydropyrimidine-515 carboxamide;
2-(4-fluorophenyl)-N-(5-((3-((l-methoxy-2-methylpropan-2-yl)amino)-lH-pyrazolo[3,4b]pyridin-4-yl)oxy)pyridin-2-yl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
(R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4y l)oxy)phenyl)-3-(4-fluorophenyl)-l-isopropyl-2,4-dioxo-1,2,3,4-tetrahydropy ri midine-520 carboxamide;
(R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-y])(methyl)amino)-lH-pyrazolo[3,4b]pyridin-4-yl)oxy)phenyl)-3-(4-fluorophenyl)-l-isopropyl-2,4-dioxo-1,2,3,4tetrahydropyrimidine-5-carboxamide;
(S)-N-(3-fluoro-4-((3-((l -hydroxypropan-2-yl)(methyl)amino)-lH-pyrazolo[3,42 5 b]pyridin-4-yl)oxy)phenyl)-3-(4-fluorophenyl)- l -isopropyl-2,4-dioxo-l ,2,3,4tetrahydropyrimidine-5-carboxamide;
(R)-N-(3-fluoro-4-((3-((2-hydroxypropyl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-3-(4-fluorophenyl)-[-isopropyl-2,4-dioxo-1,2,3,4-tetrahydropyrimidine-5carboxamide;
30 (S)-N-(3-fluoro-4-((3-((2-hydroxypropyî)amino)-lH-pyrazolo[3,4-b]pyridin-4y l)oxy)phenyl)-3-(4-fluorophenyl)-l-isopropyl-2,4-dioxo-1,2,3,4-tetrahydropyrimidîne-520534
394 carboxamide;
N-(3-fluoro-4-((3-((2-hydroxy-2-methylpropyl)amino)-lH-pyrazolo[3,4-b]pyridin-4y l)oxy)phenyl)-3-(4-fluoropheny 1)-1-isopropy 1-2,4-dioxo-1,2,3,4-tefrahydropyrimidine-5carboxamide;
N-(3-fluoro-4-((3-(((2R,3S)-3-hydroxybutan-2-yl)ainino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-3-(4-fluorophenyl)-l-isopropyl-2,4-dioxo-l,2s3,4-tetrahydropyrimidine-5carboxamide;
N-(3-f1uoro-4-((3-(((2R,3R)-3-hydroxybutan-2-yl)amino)-l H-pyrazo lo[3,4-b]pyridin-4yl)oxy)phenyl)-3-(4-fluorophenyl)-l-isopropyl-2,4-dioxo-l,2,3,4-tetrahydropyrimidine-5carboxam ide;
(R)-N-(3-fluoro-4-((3-((3-hydroxy-3-methylbutan-2-yl)amino)-lH-pyrazolo[3,4b]pyridin-4-yl)oxy)phenyl)-3-(4-fluorophenyl)-l-isopropy 1-2,4-dioxo-1,2,3,4tetrahydropyrimidine-5-carboxamide;
(S )-N-(3 -fl uoro-4-((3-((3 -hy droxy-3-methy lbutan-2-y l)am i no)-1 H-pyrazo lo [3,4b]pyridin-4-yl)oxy)phenyl)-3-(4-fluorophenyl)-l-isopropy 1-2,4-dioxo-1,2,3,4tetrahydropyrimidîne-5-carboxamide;
(R)-N-(3-fluoro-4-((3-((l -hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyrÎdin-4yl)oxy)phenyl)-3-(4-fluorophenyl)-l-methy 1-2,4-dioxo-1,2,3,4-tetrahydropyrimidine-5carboxamide;
(R)-l -ethyl-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-l H-pyrazolo[3,4b]pyridin-4-yl)oxy)phenyl)-3-(4-fluorophenyl)-2,4-dioxo-1,2,3,4-tetrahydropyrimidine-5carboxamide;
(R)-i-(cyclopropylmethyl)-N-(3-fluoro-4-((3-(( l-hydroxypropan-2-yl)amino)-lHpyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-3-(4-fluorophenyl)-2,4-dioxo-l,2,3,4tetrahydropyrimidine-5-carboxamide;
(R)-4-ethoxy-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazoio[3,4b]pyridin-4-yl)oxy)phenyl)-l-(4-fluorophenyl)-2-oxo-l,2-dihydropyridine-3-carboxamide;
(R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-l,5-dimethyl-3-oxo-2-pheny 1-2,3-dihydro-l H-pyrazo le-4-carboxamide;
(R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-l-methyl-3-oxo-2-pheny 1-2,3-dihydro-l H-pyrazole-4-carboxamide;
395 (R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-2-(4-f1uorophenyl)-l-methyl-3-oxo-2,3-dihydro-lH-pyrazoie-4-carboxamide;
(R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazoLo[3,4-b]pyridin-4yl)oxy)phenyl)-3 -(4-fluorophenyl)-l-(l-methylazetidin-3-yl)-2,4-dioxo-l,2,3,4tetrahydropy ri mid ine-5 -carboxam i de;
(R)-3-cycIopentyl-N-(3-fluoro-4-((3-((l -hydroxypropan-2-yl)amîno)-lH-pyrazolo[3,4b]pyridin-4-yl)oxy)phenyl)-1-isopropy 1-2,4-dioxo-1,2,3,4-tetrahydropyrimidine-5-carboxamide;
(R)-l-cyclobutyl-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4b]pyridin-4-yl)oxy)phenyl)-3-(4-fluorophenyl)-2,4-dioxo-1,2,3,4-tetrahy dropyrimid ine-5carboxamide;
(R)-3-(3,4-difluorophenyl)-bI-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lHpyrazoio[3,4-b]pyridin-4-yl)oxy)phenyl)-l-isopropy 1-2,4-dioxo-1,2,3,4-tetrahyd ropy ri mid ine-5carboxamide;
(R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-l -isopropy 1-3-( 1 -methyl-1 H-pyrazol-4-yl)-2,4-dîoxo-I,2,3,4tetrahydropyrimid ine-5-carboxamide;
(R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4y l)oxy)phenyl)-3-(4-fluorophenyl)-2,4-dioxo-l-(pentan-3-yl)-1,2,3,4-tetrahydropyrimidine-5carboxamide;
(R)-N-(3-fluoro-4-((3-((l -hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-l,3-di isopropy 1-2,4-dioxo-1,2,3,4-tetrahydropyrimidine-5-carboxamide;
(R)-3-cyclohexyl-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4b]pyridin-4-yl)oxy)phenyl)-l-isopropy i-2,4-dioxo-1,2,3,4-tetrahydropyrimidine-5-carboxamide;
(R)-3-(4-chlorophenyl)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lHpyrazolo[3,4-b]pyrîdin-4-yl)oxy)phenyl)-l-isopropy 1-2,4-dioxo-1,2,3,4-tetrahydropyrimidine-5carboxamide;
(S)-N-(3-nLioro-4-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4y l)oxy)phenyl)-3-(4-fluorophenyl)-l-isopropy 1-2,4-dioxo-1,2,3,4-tetrahydropyrim idine-5carboxamide;
(R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)ammo)-lH-pyrazolo[3,4-b]pyndin-4yl)oxy)phenyl)-l -isopropy l-2,4-dîoxo-3-(tetrahydro-2H-pyran-4-yl)-1,2,3,420534
396 tetrahydropyrimidine-5-carboxamîde;
(R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-l-(4-fluorophenyl)-6-methyl-2-oxo-l,2-dihydropyridine-3-carboxamide;
N-(3-fluoro-4-((3-(((R)-l -hydroxypropan-2-yl)amino)-H-I-pyrazolo[3,4-b]pyridin-45 yl)oxy)phenyl)-I-(4-f]uorophenyl)-2-oxopiperidine-3-carboxamide;
(R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-H-l-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-1 -(4-fluorophenyl)-4-methyl-2-oxo-l ,2-dihvdropyridine-3-carboxamide;
(R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-J -(4-fluorophenyl)-5-methyl'2-oxo-l,2-dihydropyridine-3-carboxamide;
10 (R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-4-(4-fluorophenyl)-3-oxo-3s4-dihydropyrazine-2-carboxamide;
(R)-6-cyclopropyl-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4b] pyridin-4-y l)o xy)pheny 1)-2-(4-fl uoropheny l)-3-o xo-2,3-dihydropy ri dazine-4-carboxamide;
(R)-N-(3-fluoro-4-( (3-((1 -hydroxypropan-2-yl)amino)-l H-pyrazolo[3,4-b]pyridin-415 yl)oxy)phenyl)-2-(4-fluorophenyl)-6-isopropyl-3-oxo-2,3-dihydropyridazine-4-carboxamide;
(R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-l H-pyrazolo[3,4-b]pyridin-4yi)oxy)phenyl)-4-(4-fluoropheny!)-2-isopropyl-5-oxo-3-thioxo-2,3,4,5-tetrahydtO-l,2,4-triazine6-carboxamide;
(R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazolo[3s4-b]pyridin-420 y])oxy)phenyl)-4-(4-fluorophenyl)-2-isopropyl-3,5-dioxo-2,3,4}5-tetrahydro-l,2,4-triazine-6carboxamîde;
(S)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-4-(4-fluorophenyl)-2-isopropyl-3,5-dioxo-2,3,4,5-tetrahydrD-L2,4-triazine-6carboxamîde;
25 (R)-l-cyclopropyl-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4b]pyridin-4-yl)oxy)phenyl)-3-(4-f]uorophenyI)-2;4-dioxo-l,2,3!4-tetrahydropyrimidine-5carboxamide;
(R)-N-(3-fIuoro-4-((3-((l-hydroxypropan-2-yl)amino)-l H-pyrazo]o[3,4-b]pyridin-4yl)oxy)phenyt)-l-(5-fluoropyridin-2-yi)-2,5-dioxo-l}2,5;6,758-hexahydroquinoline-330 carboxamîde;
(R)-5-(4-fluorophenyl)-N-(5-((3-((ï-hydroxypropan-2-yl)amino)-lH-pyrazo[o[3,420534
397 b ] py ri d i η-4 -y l)oxy )py rid in-2-y I)-1 -m ethy 1 -4-oxo-1,4-d ihyd ropyri d ine-3 -carboxam ide ;
(R)-l-(4-fluorophenyl)-N-(5-((3-((I-hydroxypropan-2-yl)amino)-lH-pyrazo[o[3,4b Jpy ridin-4-y l)o xy)py rid in-2-y l)-5-m ethy 1-2-oxo-1,2-dihydropy rid ine-3-carboxam ide;
(R)-N-(3 -fl uoro-4-((3 -(( 1 -hydroxy propan -2-y l)am ino)-1H -py razol o[3,4-b]py rid i n-45 yl)oxy)phenyl)-5-(4-fluorophenyl)-Î-isopropyl-4-oxo-l,4-dihydropyridine-3-carboxamide;
(R)-l-ethyl-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4b]pyridÎn-4-yl)oxy)phenyl)-5-(4-fluorophenyl)-4-oxo-l,4-dihydropyridine-3-carboxamide;
(R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-5-(4-fluorophenyl)-l-methyl-4-oxo-l ,4-dîhydropyridine-3-carboxamide;
10 (S)-N-(3-fluoro-4-((3-(( 1 -hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-l-(4-fluorophenyl)-5-methyl-2-oxo-l,2-dihydropyridine-3-carboxamide;
(S)-N-(3-fluoro-4-((3-(( 1 -hydroxypropan-2-yl)amino)-1 H-pyrazolo[3,4-b]pyridin-4yi)oxy)phenyl)-5-(4-nuorophenyl)-l-methyl-4-oxo-l,4-dihydropyridine-3-carboxamide;
(R)-5-cyclopropyl-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)armno)-lH-pyrazolo[3,415 b]pyridÎn-4-yl)oxy)phenyl)-l-(4-f]uoropheny!)-2-oxo-l,2-dihydropyridine-3-carboxamide;
(R)-l-(3,4-difluorophenyl)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lHpyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-5-methyi-2-oxo-l,2-dihydropyridine-3-carboxamide;
(R)-5-bromo-N-(3-fluoro-4-((3-((l-hydroxypropan-2-y0amino)-lH-pyrazolo[354b]pyridin-4-yl)oxy)phenyl)-l-(4-fluorophenyl)-2-oxo-li2-dihydropyridine-3-carboxamide;
20 (R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-l-(3-fluorophenyl)-5-methyl-2-oxo-l,2-dihydropyridine-3-carboxamide;
(R)-5-(3,4-difluorophenyl)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lHpyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-1 -isopropyI-4-oxo-l,4-dihydropyridine-3-carboxamide;
(R)-5-(4-chlorophenyl)-N-(3-fluoro-4-((3-(( 1 -hydroxypropan-2-yl)amino)-l H25 pyrazolo[354-b]pyridin-4-yl)oxy)phenyl)-l -isopropyl-4-oxo-l ,4-dihydropyridine-3-carboxamide;
(S)-N-(3-fluoro-4-((3-(( 1 -hydroxypropan-2-yl)amino)-1 H-pyrazolo[3,4-b]pyridin-4y l)oxy)pheny 1)-5-(4-fluoropheny I)-1 -isopropyl-4-oxo-l,4-dihydropyridîne-3-carboxamide;
(S)-I-ethyl-N-(3-flLioro-4-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4b]pyridin-4-yl)oxy)phenyl)-5-(4-flLiorophenyl)-4-oxo-t,4-dihydropyridine-3-carboxamide:
30 (R)-5-(2,4-difluorophenyl)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-l HpyrazoJo[3,4-b]pyndin-4-yl)oxy)phenyl)-l-isopropyl-4-oxo-l,4-dihydropyridine-3-carboxamide;
398 (R)-3-(3,4-difluorophenyl)-1 -ethyl-N-(3-fluoro-4-((3-(( I -hydroxypropan-2-yl)amino)lH-pyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-2,4-dioxo-1,2,3,4-tetrahydropyrimidine-5carboxamide;
(R)-5-chloro-N-(3-fluoro-4-((3-(( l-hydroxypropan-2-y l)amino)-lH-pyrazolo[3,4b]pyridin-4-yl)oxy)phenyl)-l-(4-fl no rophenyl)-2-oxo-l,2-dihydropyridine-3-carboxamide;
(S)-l-ethyl-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4b]pyridin-4-yl)oxy)phenyl)-3-(4-fluorophenyi)-2,4-dioxo-l,2,3,4-tetrahydropyrimidine-5carboxamide;
(S)-3-(3,4-difluorophenyl)-N-(3-fluoro-4-((3-(( l-hydroxypropan-2-y !)amino)-l Hpyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-l-isopropyl-2,4-dioxo-1,2,3,4-tetrahydropyrimidine-5carboxamide;
(S)-4-(3,4-difluorophenyl)-2-ethyl-N-(3-fluoro-4-((3-(( l-hydroxypropan-2-y l)amino)lH-pyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-l,2,4-triazine-6carboxamide;
(R)-4-(3,4-difluorophenyl)-2-ethyl-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)1 H-pyrazolo[3,4-b]pyridÎn-4-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6carboxamide;
(S)-4-(3,4-difluorophenyl)-N-(3-fl uoro-4-((3-(( l-hydroxypropan-2-y l)ami no)-1 Hpyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-2-isopropyl-3,5-dioxo-2,3,4,5-tetrahydro-l,2,4-triazine6-carboxamide;
(R)-4-(3,4-d i fluoropheny l)-N-(3-fluoro-4-((3 -(( 1 -hyd roxypropan-2-y l)am i no)-1Hpyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-2-isopropyl-3,5-dioxo-2,3,4,5-tetrahydro-l,2,4-triazine6-carboxamide;
(S)-2-ethyl-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-l H-pyrazolo[3,4b]pyridin-4-yl)oxy)phenyl)-4-(4-fiuorophenyI)-3,5-dioxo-2,3,4,5-tetrahydro-l,2,4-triazine-6carboxamide;
(R)-2-ethyl-N-(3-flLioro-4-((3-(( l-hydroxypropan-2-y l)amino)-lH-pyrazolo[3,4b]pyridin-4-yl)oxy)pheny])-4-(4-fluorophenyl)-3,5-dioxo-2,3,4,5-tetrahydro-l,2,4-triazine-6carboxamide;
(S)-3-(3,4-difluorophenyl)-l-ethy]-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)lH-pyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-2,4-dioxo-l,2,3,4-tetrahydropyrîmidine-520534
399 carboxamide;
(S)-l-cyclopropyl-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4b]pyridin-4-yl)oxy)phenyl)-3-(4-fluorophenyl)-2,4-dioxo-l,2,3,4-tetrahydropyrimidine-5carboxamide;
N-(3-fluoro-4-((3-(((R)-l-hydroxypropan-2-yl)amino)-IH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-5-(4-fluorophenyl)-6-oxo-5-azaspiro[2.5]octane-7-carboxamîde;
N-(3-fluoro-4-((3-(((R)-l-hydroxypropan-2-yl)ammo)-l H-pyrazolo[3,4-b]pyridin-4yl)oxy)pheny])-l-(4-fluorophenyL)-3-methyl-2-oxopiperidine-3-carboxamide;
(R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)pheny])-5-hydroxy-2-oxo-l-phenyl-l,2-dihydroquinoline-3-carboxamide;
(R)-2-(4-fluorophenyl)-N-(5-((3-(( 1 -hydroxypropan-2-yl)amino)-1 H-pyrazolo[3,4b]pyridin-4-y])oxy)pyridin-2-yl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
(R)-2-(4-fluoropheny!)-N-(5-((3-((l -hydroxypropan-2-yl)amino)-1 H-pyrazolo[3,4b]pyridin-4-yl)oxy)pyridin-2-yl)-6-methyl-3-oxo-2,3-dihydropyridazine-4-carboxamide;
(R)-3-(4-fluorophenyl)-N-(5-((3-(( 1 -hydroxypropan-2-yl)amino)-1 H-pyrazolo[3,4b]pyridin-4-yl)oxy)pyridin-2-yl)-l-isopropyl-2,4-dioxo-l,2,3,4-tetrahydropyrimîdine-5carboxamide;
(R)-N-(3-fluoro-4-((3-((l-methoxypropan-2-y[)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-3-(4-fluorophenyl)-l-isopropyl-2,4-dîoxo-l,2,3,4-tetrahydropyrimidine-5carboxamide;
(S)-3-cyclopentyl-N-(3-fluoro-4-((3-((l -methoxypropan-2-yl)amino)-î H-pyrazolo[3,4b]pyridin-4-yl)oxy)phenyl)-l-isopropyl-2,4-dioxo-i,2,3,4-tetrahydropyrimidine-5-carboxamide;
(S)-l-ethyl-N-(3-fluoro-4-((3-((l-methoxypropan-2-yl)amino)-IH-pyrazolo[3,4b]pyridin-4-y])oxy)phenyl)-3-(4-fluorophenyl)-2,4-dioxo-1,2,3,4-tetrahydropyrimidine-5carboxamide;
(S)-N-(3-fluoro-4-((3-((l-methoxypiOpan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-3-(4-fIuorophenyl)-l-methyl-2,4-dioxo-l,2,3,4-tetrahydropyrimidine-5carboxamide;
(S)-l -(cyclopropylmethyi)-N-(3-fluoro-4-((3-((l-methoxypropan-2-yl)amino)-]Hpyrazolo[3,4-b]pyridin-4-y])oxy)phenyl)-3-(4-fluorophenyl)-2,4-dioxo-I,2,3,4tetrahydropyrimidine-5-carboxamide;
400 (S)-N-(3-fluoro-4-((3-((l-methoxypropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)pheny 1)-1-(4-fluorophenyl)-6-methyl-2-oxo-l,2-dihydropyridine-3-carboxamide;
(S)-l-cyclobutyl-N-(3-fluoro-4-((3-((l-methoxypropan-2-yl)amino)-lH-pyrazolo[3,4b]pyridin-4-y!)oxy)phenyl)-3-(4-fluorophenyI)-2,4-dioxo-l,2,3,4-tetrahydropyrimidine-5carboxamide;
(S)-3-(3,4-dif]uorophenyl)-N-(3-fluoro-4-((3-((]-rnethoxypropan-2-yl)amino)-l Hpyrazolo[3,4-b]pyridin-4-yl)oxy)phenyi)-l-isopropyl-2,4-dioxo-l,2,3,4-tetrahydropyrimidine-5carboxamide;
(S)-N-(3-fluoro-4-((3-((l-methoxypropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-3-(4-fluorophenyl)-l-isopropyl-2,4-dioxo-l,2,3,4-tetrahydropyrimidine-5carboxamide;
N-(3-fluoro-4-((3-((l-methoxy-2-methylpropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-3-(4-fluorophenyl)-l-isopropyl-2,4-dioxo-l,2,3,4-tetrahydropyrimidine-5carboxamide;
N-(3-fluoro-4-((3-((l-methoxy-2-methylpropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin4-yl)oxy)phenyl)-4-(4-fluorophenyl)-2-isopropyl-3,5-dioxo-2,3,4,5-tetrahydro-l,2,4-triazine-6carboxamide;
1-ethyl-N-(3-f1uoro-4-( (3-((1-methoxy-2-methylpropan-2-yl)amino)-l H-pyrazolo[3,4b]pyridin-4-yl)oxy)pbenyl)-3-(4-fluorophenyl)-2,4-dioxo-1,2,3,4-tetrahydropyrimidine-5carboxamide;
N-(3-fluoro-4-((3-((l-methoxy-2-methylpropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin4-yl)oxy)pheny 1)-3-(4-fluorophenyl)-l-methyl-2,4-dioxo-1,2,3,4-tetrahydropyrimidine-5carboxamide;
N-(3-fluoro-4-((3-(( 1 -methoxy-2-methylpropan-2-yl)amino)-1 H-pyrazolo[3,4-b]pyndin4-yl)oxy)pheny 1)-3-(4-fl uoropheny 1)-1-(2-hydroxyethyl)-2,4-dioxo-1,2,3,4tetrahydropyrimidine-5-carboxamide;
(R)-N-(3-fluoro-4-((3-((1 -methoxypropan-2-y!)amino)-l H-pyrazolo[3,4-b]pyridin-4yl)oxy)pheny])-l-(4-fluorophenyl)-6-methyl-2-oxo-l,2-dihydropyridine-3-carboxamide;
(R)-l-cyclobutyl-N-(3-fluoro-4-((3-((l-methoxypropan-2-yl)amino)-lH-pyrazolo[3,4b]pyridin-4-y[)oxy)phenyl)-3-(4-fluoropheny[)-2,4-dioxo-l,2,3,4-tetrahydropyrimidine-5carboxamide;
401 (R)-N-(3-fluoro-4-((3-((l-methoxypropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-4-(4-fluorophenyl)-2-îsopropyl-3,5-dioxo-2,3,4,5-tetrahydro-l,2,4-triazine-6carboxamide;
(S)-N-(3 -fl uoro-4-((3 -(( 1 -methoxy propan-2 -y 1 )am i no )-1 H-pyrazo lo[3,4-b]pyridin-4yl)oxy)phenyI)-4-(4-fluorophenyl)-2-isopropyl-3,5-dioxo-2,3,4,5-tetrahydro-I,2,4-triazine-6carboxamide;
N-(4-((3-((4,4-difltiorobutan-2-yl)amino)-IH-pyrazolo[3,4-b]pyridin-4-yl)oxy)-3fluorophenyl)-3-(4-fluorophenyl)-l-isopropyl-2,4-dioxo-l,2,3,4-tetrahydropyrimidîne-5carboxamide;
N-(4-((3-((4,4-difluorobutan-2-yI)amino)-l H-pyrazo lo[3,4-b]pyridin-4-yl)oxy)-3f]uorophenyl)-5-(4-flitorophenyl)-l-methyl-4-oxo-l,4-dihydropyridîne-3-carboxamide;
N-(4-((3-((4,4-difluorobutan-2-yl)amino)-l H-pyrazolo[3,4-b]pyridin-4-yl)oxy)-3fluorophenyl)-2-(4-fluorophenyl)-6-isopropyl-3-oxo-2,3-dihydropyndazine-4-carboxamÎde;
N-(5-((3-((4,4-difluorobutan-2-yl)aniino)-lH-pyrazolo[3,4-b]pyridÎn-4-yl)oxy)pyridin-2yl)-5-(4-fluorophenyl)-l-methyl-4-oxo-l,4-dîhydropyridine-3-carboxamide;
N-(5-((3-((4,4-difluorobutan-2-yl)amino)-I H-pyrazolo[3,4-b]pyridin-4-yl)oxy)pyridin-2y])-l-(4-fluorophenyl)-5-methyl-2-oxo-l,2-dihydropyridine-3-carboxamide;
(S)-N-(3-fluoro-4-((3-(( 1 -methoxypropan-2-yl)amino)-1 H-pyrazo lo [3,4-b]pyridin-4yl)oxy)phenyl)-1 -(4-fluorophenyl)-5-methyl-2-oxo-1,2-dihydropyridine-3-carboxamide;
N-(3-fluoro-4-((3-((l-methoxy-2-methylpropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin4-y[)oxy)phenyl)-l-(4-fluorophenyl)-5-methyl-2-oxo-l,2-dihydropyridine-3-carboxamide;
(S)-3-(3,4-difluorophenyl)-N-(4-((3-((l-ethoxypropan-2-yl)amino)-lH-pyrazolo[3,4b]pyridin-4-yl)oxy)-3-fluorophenyl)-l-Îsopropyl-2,4-dioxo-l,2,3,4-tetrahydropyrimÎdine-5carboxamide;
(S)-N-(4-((3-((1-ethoxy propan-2-yl)am ino)-1 H-pyrazolo[3,4-b]pyridin-4-y l)oxy)-3nuorophenyl)-l-(4-fluorophenyi)-5-methyl-2-oxo-l,2-dihydropyridine-3-carboxamide;
(S)-N-(4-((3 -((1-ethoxypropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4-yl)oxy)-3fiuorophenyl)-2-(4-f1uorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
(R)-5-(4-fluorophenyl)-l-isopropyl-N-(5-((3-((l-methoxy propan-2-y l)amino)-l Hpyrazolo[3,4-b]pyridin-4-yl)oxy)pyridin-2-yl)-4-oxo-l,4-dihydropyridine-3-carboxamide;
(R)-N-(3-fîuoro-4-( (3-((1-methoxy propan-2-y l)ami no)-IH-pyrazolo [3,4-b]pyridin-420534
402 yl)oxy)phenyl)-2-(4-fluorophenyl)-6-isopropyl-3-oxo-2,3-dihydropyridazine-4-carboxamide;
(R)-2-(4-fluorophenyl)-6-îsopropyI-N-(5-((3-((l-methoxypropan-2-yl)amino)-IHpyrazolo[3,4-b]pyridin-4-yl)oxy)pyridin-2-yl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
(R)-5-cyclopropyl-l-(4-fluorophenyl)-N-(5-((3-((l-methoxypropan-2-yl)amino)-lHpyrazolo[3,4-b]pyridin-4-y])oxy)pyridin-2-yl)-2-oxo-l,2-dihydropyridine-3-carboxamide;
(R)-l-(4-fluorophenyl)-N-(5-((3-((l-methoxypropan-2-yi)amino)-lH-pyrazolo[3,4b]pyridin-4-y])oxy)pyridin-2-yl)-5-methyl-2-oxo-l,2-dihydropyridine-3-carboxamide;
(R)-l-(cyclopropylmethyl)-N-(3-fluoro-4-((3-((l-methoxypropan-2-yl)amino)-lHpyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-3-(4-fluorophenyl)-2,4-dioxo-l,2,3,4tetrahydropyrimidine-5-carboxamide;
(R)-l-ethyl-N-(3-fluoro-4-((3-((l-methoxypropan-2-yl)amino)-lH-pyrazolo[3,4b]pyridin-4-yl)oxy)phenyl)-3-(4-fluorophenyl)-2,4-dioxo-l,2,3,4-tetrahydropyrimidine-5carboxamide;
(R)-4-ethoxy-N-(3-fluoro-4-((3-((l-methoxypropan-2-yl)amÎno)-] H-pyrazolo[3,4b]pyridin-4-yl)oxy)phenyl)-l -(4-fl uoropheny l)-2-oxo-l,2-dihydropyridine-3-carboxamide;
N-(3-fluoro-4-((3-((]-(2-hydroxypropan-2-yl)cyclopentyl)amino)-lH-pyrazolo[3,4b]pyridin-4-yi)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
2-((4-(2-fl uoro-4-(2-(4-fl uoropheny l)-3-oxo-2,3-dihydropy ridazine-4carboxatn ido)phenoxy)-1 H-py razo lo[3,4-b] py rid î n-3-y l)ami no)-2 -methy Ipropy 1 hydrogen sulfate;
(S)-N-(3-fluoro-4-((3-((3-hydroxy-3-methylbutan-2-yl)amino)-lH-pyrazolo[3,4b]pyridin-4-yl)oxy)pheny 1)-2-( 4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
(R)-N-(3-fluoro-4-((3-((3-hydroxy-3-niethylbutan-2-yl)amino)-lH-pyrazolo[3,4b]pyridin-4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(3-fluoro-4-((3-(((lS,2R)-2-(hydroxymethyl)cyclopentyl)amino)-lH-pyrazolo[3,4b]pyrîdin-4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazÎne-4-carboxamide;
N-(3-fluoro-4-((3-(((lR,2S)-2-(hydroxymethyl)cyclopentyl)aniino)-lH-pyrazolo[3,4b]pyridin-4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
(S)-N-(3-fluoro-4-((3-((4-hydroxy-l-methoxy-2-methylbutan-2-yl)amino)-l Hpyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4carboxamide;
403 (R)-N-(3-fluoro-4-((3-((4-hydroxy-l-methoxy-2-methylbutan-2-yl)amino)-lHpyrazolo[3,4-b]pyridin-4-yl)oxy)phenyl)-2~(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4carboxamide;
(S)-N-(3-fluoro-4-((3-((l-hydroxy-3-methoxypropan-2-yl)amino)-lH-pyrazolo[3,4b]pyridin-4-yl)oxy)phenyI)-2-(4-fluorophenyl)-3-oxo-2,3-dîhydropyridazine-4-carboxamide;
(R)-N-(3-fluoro-4-((3-((l-hydroxy-3-methoxypropan-2-yl)amino)-lH-pyrazolo[3,4b]pyridîn-4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
(R)-N-(3-fluoro-4-((3-((l-hydroxy-4-methoxybutan-2-yl)amino)-lH-pyrazolo[3,4b]pyridin-4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
(S)-N-(3-fluoro-4-((3-((l-hydroxy-4-methoxybutan-2-yl)amino)-l H-pyrazolo[3,4b]pyrîdin-4-yl)oxy)phenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
(R)-N-(4-((3-((4,4-difluoro-l-hydroxybutan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)-3-fluorophenyl)-2-(4-fluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
(S)-N-(4-((3-((4,4-difluoro-l-hydroxybutan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)-3-fluorophenyl)-2-(4-tluorophenyl)-3-oxo-2,3-dihydropyridazine-4-carboxamide;
N-(4-((3-amino-lH-pyrazolo[3,4-b]pyridin-4-yl)oxy)-3-fluorophenyl)-3-(4fluorophenyl)-1 -isopropyl-2,4-dioxo-1,2,3,4-tetrahydropy ri midine-5-carboxamide;
’N-(3-fluoro-4-((3-(methylamino)-lH-pyrazolo[3,4-b]pyridin-4-yl)oxy)pheny 1)-3-(4fluorophenyl)-!-isopropyl-2,4-dioxo-1,2,3,4-tetrahydropyrimÎdine-5-carboxamide;
N-(4-((3-(ethylamino)-l H-pyrazolo[3,4-b]pyridin-4-yl)oxy)-3-fluorophenyl)-3-(4fluorophenyl)-l-isopropyl-2,4-dioxo-l,2,3,4-tetrahydropyrimidine-5-carboxamide;
(R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-l-(4-fluorophenyl)-5-isopropyl-2-oxo-l,2-dihydropyridine-3-carboxamide;
(R)-5-ethyl-N-(3-flitoro-4-((3-((l-hydroxypropan-2-yl)amino)-l H-pyrazolo[3,4b]pyridin-4-yl)oxy)phenyl)-l-(4-fluoropheny[)-2-oxo-l,2-dihydropyridine-3-carboxamide;
(R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-1 H-pyrazolo[3,4-b]pyridin-4yl)oxy)phenyl)-l-(4-fluorophenyl)-2-oxo-5-propyl-l,2-dihydropyridine-3-carboxamide;
and stéréo isomers, tautomers and pharmaceutically acceptable salts thereof.
12. A compound according to claim 1 or a stereoisomer, tautomer or pharmaceutically acceptable sait thereof, wherein:
404
X1 isCHorN;
R1 is hydrogen or C1-C6 alkyl;
R2 is (a) hydrogen, (b) Cl -C6 alkyl, (c) hydroxyCl-C6 alkyl, (d) dihydroxyC2-C6 alkyl, (e) C1-C6 fluoroalkyl optionally substituted with OH, (g) (C1-C6 alkoxy)CI-C6 alkyl- wherein said alkyl portion is optionally substituted with OH, (h) Cyc1, (i) Cyc2, (j) (hetCyc’)Cl-C6 alkyl- wherein said alkyl portion is optionally substituted with OH, (k) (Ar')C I-C6 alkyl- wherein said alkyl portion is optionally substituted with OH, (1) (hetAr')Cl-C6 alkyl- wherein said alkyl portion is optionally substituted with OH, or (m) (HOSO3)C i -C6 alkyl-;
Cyc1 is a 3-4 membered cycloalkyl ring optionally substituted with 1-2 substituents independently selected from halogen, hydroxy, hydroxyCl-C3 alkyl, C1-C3 alkoxy, (CI-C3 alkoxy)CI-C3 alkyl-, and R'RNC(=O)-;
R' and R are independently selected from C1-C6 alkyl;
Cyc2 is a 5-membered cycloalkyl ring substituted with 1-2 substituents independently selected from C1-C3 alkyl, (CI-C3 alkoxy)Cl-C3 alkyl- and hydroxyCI-C3 alkyl-;
hetCyc1 is a 5-6 membered saturated heterocyclic ring having 1-2 ring heteroatoms independently selected from O, N and SO2, wherein said ring is optionally substituted with oxo;
Ar1 is phenyl;
hetAr1 is pyridyl;
G is
405
X2 is C or N;
Ring A, including the atoms at the points of attachment, is a 5-6 membered heterocyclic ring optionally having an additional 1-2 ring nitrogen atoms when X2 is N and having one ring 5 nitrogen atom when X2 is C;
R3 is hydrogen or absent;
R6 is hydrogen, halogen, C1-C6 alkyl, C1-C6 alkoxy, hydroxyC 1-C6 alkyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)Cl-C6 alkyl- or hetCyc2, provided that when R6 is halogen and is on the ring carbon atom adjacent to the carbon linked to the -NHC(=O)- moiety of Formula I, then 10 R6 is not halogen;
R7 is hydrogen, C1-C6 alkyl, oxo or thioxo;
hetCyc2 is a 4 membered saturated heterocyclic ring having a ring nitrogen atom substituted with Cl -C6 alkyl;
Ring B, including the atoms at the points of attachment, is a 6-membered saturated 15 carbocyclic optionally substituted with oxo or a 6-membered aromatic carbocyclic ring optionally substituted with OH;
R8 is Ar2, hetAr2, C3-C6 cycloalkyl, hetCyc3 or C1-C6 alkyl;
Ar2 is phenyl optionally substituted with one or more substituents independently selected from halogen;
20 hetAr2 is a 5-6 membered heteroaryl having 1-2 ring nitrogen atoms and optionally substituted with one or more substituents independently selected from Cl -C2 alkyl; and
R9 is hydrogen or halogen.
13. A compound of Formula 1-A
406
and stereoisomers, tautomers and pharmaceutically acceptable salts thereof, wherein:
X1 is CH or N;
R1 is hydrogen or C1-C6 alkyl;
R2 is (a) hydrogen, (b) C1-C6 alkyl, (c) hydroxyCl-C6 alkyl, (g) (C1-C6 alkoxy)Cl-C6 alkyl- wherein said alkyl portion is optionally substituted with OH, or (h) Cyc1;
Cyc1 is a 3-4 membered cycloalkyl ring optionally substituted with 1-2 substituents independently selected from halogen, hydroxy, CI-C3 alkyl, hydroxyC 1-C3 alkyl, C1-C3 alkoxy, (C1-C3 alkoxy)Cl-C3 alkyl-and R'RNC(=O)-;
X2 is N, R3 is absent, Ring A is a 6-membered heterocyclic ring having one additional ring nitrogen atom, and R7 is oxo, wherein G has the formula A-l
R6 is C1-C6 alkyl, hydroxyCI-C6 alkyl, C3-C6 cycloalkyl, (C3-C6 cycloalkyl)Cl-C6 alkyl- or hetCyc2;
hetCyc2 is a 4-6 membered saturated heterocyclic ring having a ring nitrogen atom and
407 optionally substituted with C1-C6 alkyl;
RK is Ar2, hetAr2, C3-C6 cycloalkyl, hetCyc3 or C1-C6 alkyl;
Ar2 is phenyl optionally substituted with one or more substituents independently selected from halogen, C1-C2 alkyl and C1-C2 alkoxy;
hetAr2 is a 5-6 membered heteroaryl having 1-2 ring nitrogen atoms and optionally substituted with one or more substituents independently selected from halogen, CI-C2 alkyl and C1-C2 alkoxy;
hetCyc3 is a 5-6 membered heterocyclic ring having a ring oxygen atom; and
R9 is hydrogen or halogen.
14. (R)-N-(3-fluoro-4-((3-((l-hydroxypropan-2-yl)amino)-lH-pyrazolo[3,4b]pyridin-4-yl)oxy)phenyl)-3-(4-fluorophenyl)-l-isopropyl-2,4-dioxo-l ,2,3,4tetrahydropyrimidine-5-carboxamide having the structure
or a stereoisomer, tautonrer or pharmaceutically acceptable sait thereof.
15. A pharmaceutical composition, comprising a compound of Formula I as defined in any one of daims 1 to 14 or a stereoisomer, tautomer or pharmaceutically acceptable sait thereof, and a pharmaceutically acceptable diluent or carrier.
16. A compound of any one of daims 1-14 or a stereoisomer, tautomer or pharmaceutically acceptable sait thereof, or a pharmaceutical composition according to daim 15 for use in a method for treating cancer in a patient in need thereof.
408
17. Use of a compound of any one of claims 1-14 or a stereoisomer, tautomer or pharmaceutically acceptable sait thereof, or a pharmaceutical composition according to claim 15 in the manufacture of a médicament for treating cancer in a patient in need thereof.
18. The compound for use of claim 16 or the use of claim 17, wherein compound is for coadministration to the patient with at least one additional anticancer agent.
19. The compound for use or the use of claim 18, wherein the at least one additional anticancer agent or therapy is selected from the group consisting of: an immune checkpoint inhibitor, a kinase inhibitor, a chemotherapy, radiation, and surgery.
20. The compound for use or the use of claim 18 or claim 19, wherein the at least one additional anticancer agent is selected from the group consisting of: a chemotherapeutic agent, a PI-3 kinase inhibitor, an EGFR inhibitor, a HER2/neu inhibitor, an FGFR inhibitor, an ALK inhibitor, an IGF! R inhibitor, a VEGFR inhibitor, a PDGFR inhibitor, a glucocorticoid, a BRAF inhibitor, a MEK inhibitor, a HER4 inhibitor, a MET inhibitor, a RAF inhibitor, an Akt inhibitor, aFTL-3 inhibitor, and a MAP kinase pathway inhibitor.
21. The compound for use or the use of any one of claims 16-20, wherein the subject is identified or diagnosed as having a TAM-associated cancer.
22. The compound for use or the use of claim 21, wherein the TAM-associated cancer is selected from the group consisting of: gastrointestinal stromal tumor (GIST), acute myeloid leukemia (AML), acute lymphocytic leukemia (ALL), chronic myeloid leukemia (CML), B-cell chronic myeloid leukemia (B-CLL), lung cancer, glioblastoma, breast cancer, colorectal cancer, gastric cancer, glioma, pancreatic cancer, esophageal cancer, mande cell lymphoma, melanoma, squamous cell skin cancer, prostate cancer, endométrial cancer, ovarian cancer, oral squamous cell carcinoma, thyroid cancer, bladder cancer, rénal cancer, schwannoma, mesothelioma, Kaposi’s sarcoma, osteosarcoma, rhabdomyosarcoma, erythroid leukemia, colon cancer, liver cancer, rénal cell carcinoma, pituitary adenoma, urinary tract cancer, kidney cancer, colon cancer, head and neck cancer, brain cancer, and non-small cell lung cancer.
OA1202100068 2018-08-30 2019-08-29 Pyrazolo[3,4-b]Pyridine compounds as inhibitors of TAM and MET kinases. OA20534A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US62/724,829 2018-08-30
US62/858,686 2019-06-07

Publications (1)

Publication Number Publication Date
OA20534A true OA20534A (en) 2022-10-27

Family

ID=

Similar Documents

Publication Publication Date Title
AU2019330017B2 (en) Pyrazolo(3,4-b)pyridine compounds as inhibitors of TAM and MET Kinases
AU2016341445B2 (en) 2-aryl- and 2-heteroaryl-substituted 2-pyridazin-3(2H)-one compounds as inhibitors of FGFR tyrosine kinases
JP2020533277A (en) Inhibitors of KRas G12C and methods of using them
KR20240004960A (en) RAS inhibitors
US11247990B1 (en) Bicyclic fused pyridine compounds as inhibitors of TAM kinases
TWI745824B (en) Quinoline compounds as inhibitors of tam and met kinases
OA20534A (en) Pyrazolo[3,4-b]Pyridine compounds as inhibitors of TAM and MET kinases.
RU2773611C1 (en) PYRAZOLO[3,4-b]PYRIDINE COMPOUNDS AS TAM AND MET KINASE INHIBITORS