US20230109104A1 - Heterocyclic compounds and uses thereof - Google Patents

Heterocyclic compounds and uses thereof Download PDF

Info

Publication number
US20230109104A1
US20230109104A1 US17/716,657 US202217716657A US2023109104A1 US 20230109104 A1 US20230109104 A1 US 20230109104A1 US 202217716657 A US202217716657 A US 202217716657A US 2023109104 A1 US2023109104 A1 US 2023109104A1
Authority
US
United States
Prior art keywords
alkylene
optionally substituted
halogen
oxo
alkyl
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US17/716,657
Inventor
Sarvajit Chakravarty
Son Minh Pham
Jayakanth Kankanala
Anil Kumar AGARWAL
Brahmam Pujala
Sanjeev SONI
Satish K. ARYA
Deepak PALVE
Ashu Gupta
Varun Kumar
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Nuvation Bio Inc
Original Assignee
Nuvation Bio 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 Nuvation Bio Inc filed Critical Nuvation Bio Inc
Priority to US17/716,657 priority Critical patent/US20230109104A1/en
Publication of US20230109104A1 publication Critical patent/US20230109104A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Landscapes

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

Abstract

Heterocyclic compounds as Wee1 inhibitors are provided. The compounds may find use as therapeutic agents for the treatment of diseases and may find particular use in oncology.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a divisional of U.S. application Ser. No. 16/155,838, filed Oct. 9, 2018, which claims priority to U.S. Provisional Application No. 62/570,043, filed Oct. 9, 2017, the contents of which are incorporated herein by reference in their entirety.
  • FIELD OF THE INVENTION
  • This disclosure relates generally to therapeutics engaged in inhibition of the DNA damage checkpoint kinase, Wee1, which potentiates genotoxic chemotherapies by abrogating cell-cycle arrest and proper DNA repair. The invention also provides pharmaceutically acceptable compositions comprising compounds of the present invention and methods of using said compositions in the treatment of diseases associated with this pathway.
  • BACKGROUND OF THE INVENTION
  • Wee1 is a tyrosine kinase that phosphorylates and inactivates Cdc2 and is involved in G checkpoint signaling. More particularly, Wee1 is involved in G2-M checkpoint signaling. Because p53 is a key regulator in the G checkpoint, p53-deficient tumors rely only on the G checkpoint after DNA damage. More particularly, because p53 is a key regulator in the G1-S checkpoint, p53-deficient tumors rely only on the G2-M checkpoint after DNA damage. Hence, such tumors are selectively sensitized to DNA-damaging agents by Wee1 inhibition.
  • Wee1 belongs to a family of protein kinases involved in the terminal phosphorylation and inactivation of cyclin-dependent kinase 1-bound cyclin B, resulting in G cell cycle arrest in response to DNA damage. Wee1 was first identified in fission yeast, where Wee1 deficiency resulted in premature mitotic entry and replication of smaller-sized yeast. It is the major kinase responsible for the inhibitory phosphorylation of the tyrosine.
  • Before cells undergo mitosis, they progress through a tightly controlled cascade of G1-S, intra-S, and G2-M checkpoints. Wee1 kinase has emerged as a key G2-M checkpoint regulator. This tyrosine kinase negatively regulates entry into mitosis by catalyzing an inhibitory phosphorylation of Cdc2 (the human homolog of cyclin-dependent kinase 1 (CDK1) on tyrosine-15 (Y15). This results in inactivation of the Cdc2/cyclin B complex, which arrests cells in G2-M, allowing for DNA repair. Such inhibition also occurs through Chk1-mediated inhibition of Cdc25 phosphatases, which remove the inhibitory phosphorylation on Cdc2. Thus, entry into mitosis rests on a balance between the opposing activities of Wee1 and Chk1/Cdc25. Wee1 inhibition is thus expected to abrogate G2-M arrest and propel cells into premature mitosis, a hypothesis confirmed by studies documenting that Wee1 inhibition by either small molecule inhibitors or small interference RNA leads to premature entry into mitosis and consequent cell death through mitotic catastrophe or apoptosis. (S. Muller, J. Clinical. Oncology, 2015).
  • Recently, a few classes of Wee1 inhibitors have been disclosed. Among them is a selective inhibitor, AZD-1775 (1, 2-allyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((4-(4methylpiperazin-1-yl)phenyl)amino)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one). AZD-1775 exhibited antitumor activity in various preclinical studies as a monotherapy or in potentiating chemo- and radiotherapy, and is currently in phase I/II clinical trials.
  • Wee1 is highly expressed in several cancer types, including hepatocellular carcinoma, breast cancers, cervical cancers, lung cancers, squamous cell carcinoma, diffuse intrinsic pontine glioma (DIPG), glioblastoma, medulloblastoma, leukemia, melanoma, and ovarian cancers. (P. Reigan et al., Trends in Pharmacol. Sci., 2016).
  • There are few Wee1 inhibitors in clinical development. There is scope to improve Wee1 inhibitor selectivity and the properties of the inhibitors to permit targeting of specific cancer types.
  • BRIEF SUMMARY OF THE INVENTION
  • In one aspect, provided is a compound of Formula (I):
  • Figure US20230109104A1-20230406-C00001
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein m, n, X1, X2, A, B, R1, R2, R3 and R4 are as detailed herein.
  • In some embodiments, provided is a compound of Formula (Ia):
  • Figure US20230109104A1-20230406-C00002
  • or a salt thereof, wherein A, B, R1, R2, R3 and R4 are as detailed herein. In some embodiments, provided is a compound of Formula (Ia), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing.
  • In some embodiments, the compound of Formula (I) or a salt thereof, is of the Formula (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, as detailed herein. In some embodiments, the compound of Formula (I) or a salt thereof, is of Formula (Ia), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ia-5), or (Ia-6) as detailed herein.
  • In another aspect, provided is a method of treating cancer in an individual in need thereof comprising administering to the individual a therapeutically effective amount of a compound as detailed herein, such as a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ia-5), or (Ia-6) or a pharmaceutically acceptable salt thereof. In some embodiments, provided is a method of treating cancer in an individual in need thereof comprising administering to the individual a therapeutically effective amount of a compound as detailed herein, such as a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing. Also provided is a method of inhibiting Wee1 in a cell, comprising administering a compound detailed herein, or a salt thereof, to the cell. Also provided is a method of inhibiting Wee1 in a cell, comprising administering a compound detailed herein, or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, to the cell.
  • In another aspect, provided are pharmaceutical compositions comprising a compound detailed herein and a pharmaceutically acceptable carrier or excipient. In another aspect, provided are pharmaceutical compositions comprising a compound, or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, detailed herein and a pharmaceutically acceptable carrier or excipient. Kits comprising a compound detailed herein or a salt thereof are also provided. Kits comprising a compound, or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, detailed herein are also provided. A compound as detailed herein, or a salt thereof, is also provided for the manufacture of a medicament for the treatment of cancer. A compound, or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, as detailed herein is also provided for the manufacture of a medicament for the treatment of cancer.
  • DETAILED DESCRIPTION OF THE INVENTION Definitions
  • “Alkyl” refers to and includes saturated linear and branched univalent hydrocarbon structures and combination thereof, having the number of carbon atoms designated (i.e., C1-C10 means one to ten carbons). Particular alkyl groups are those having 1 to 20 carbon atoms (a “C1-C20 alkyl”). More particular alkyl groups are those having 1 to 8 carbon atoms (a “C1-C5 alkyl”), 3 to 8 carbon atoms (a “C3-C8 alkyl”), 1 to 6 carbon atoms (a “C1-C6 alkyl”), 1 to 5 carbon atoms (a “C1-C5 alkyl”), or 1 to 4 carbon atoms (a “C1-C4 alkyl”). Examples of alkyl include, but are not limited to, groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, homologs and isomers of, for example, n-pentyl, n-hexyl, n-heptyl, n-octyl, and the like.
  • “Alkenyl” as used herein refers to an unsaturated linear or branched univalent hydrocarbon chain or combination thereof, having at least one site of olefinic unsaturation (i.e., having at least one moiety of the formula C═C) and having the number of carbon atoms designated (i.e., C2-C10 means two to ten carbon atoms). The alkenyl group may be in “cis” or “trans” configurations, or alternatively in “E” or “Z” configurations. Particular alkenyl groups are those having 2 to 20 carbon atoms (a “C2-C20 alkenyl”), having 2 to 8 carbon atoms (a “C2-C5 alkenyl”), having 2 to 6 carbon atoms (a “C2-C6 alkenyl”), or having 2 to 4 carbon atoms (a “C2-C4 alkenyl”). Examples of alkenyl include, but are not limited to, groups such as ethenyl (or vinyl), prop-1-enyl, prop-2-enyl (or allyl), 2-methylprop-1-enyl, but-1-enyl, but-2-enyl, but-3-enyl, buta-1,3-dienyl, 2-methylbuta-1,3-dienyl, homologs and isomers thereof, and the like.
  • “Alkylene” as used herein refers to the same residues as alkyl, but having bivalency. Particular alkylene groups are those having 1 to 6 carbon atoms (a “C1-C6 alkylene”), 1 to 5 carbon atoms (a “C1-C5 alkylene”), 1 to 4 carbon atoms (a “C1-C4 alkylene”) or 1 to 3 carbon atoms (a “C1-C3 alkylene”). Examples of alkylene include, but are not limited to, groups such as methylene (—CH2—), ethylene (—CH2CH2—), propylene (—CH2CH2CH2—), butylene (—CH2CH2CH2CH2—), and the like.
  • “Alkynyl” as used herein refers to an unsaturated linear or branched univalent hydrocarbon chain or combination thereof, having at least one site of acetylenic unsaturation (i.e., having at least one moiety of the formula C≡C) and having the number of carbon atoms designated (i.e., C2-C10 means two to ten carbon atoms). Particular alkynyl groups are those having 2 to 20 carbon atoms (a “C2-C20 alkynyl”), having 2 to 8 carbon atoms (a “C2-C8 alkynyl”), having 2 to 6 carbon atoms (a “C2-C6 alkynyl”), or having 2 to 4 carbon atoms (a “C2-C4 alkynyl”). Examples of alkynyl include, but are not limited to, groups such as ethynyl (or acetylenyl), prop-1-ynyl, prop-2-ynyl (or propargyl), but-1-ynyl, but-2-ynyl, but-3-ynyl, homologs and isomers thereof, and the like.
  • “Aryl” refers to and includes polyunsaturated aromatic hydrocarbon groups. Aryl may contain additional fused rings (e.g., from 1 to 3 rings), including additionally fused aryl, heteroaryl, cycloalkyl, and/or heterocyclyl rings. In one variation, the aryl group contains from 6 to 14 annular carbon atoms. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, biphenyl, and the like.
  • “Carbonyl” refers to the group C═O.
  • “Cycloalkyl” refers to and includes cyclic univalent hydrocarbon structures, which may be fully saturated, mono- or polyunsaturated, but which are non-aromatic, having the number of carbon atoms designated (e.g., C1-C10 means one to ten carbons). Cycloalkyl can consist of one ring, such as cyclohexyl, or multiple rings, such as adamantly, but excludes aryl groups. A cycloalkyl comprising more than one ring may be fused, spiro or bridged, or combinations thereof. A preferred cycloalkyl is a cyclic hydrocarbon having from 3 to 13 annular carbon atoms. A more preferred cycloalkyl is a cyclic hydrocarbon having from 3 to 8 annular carbon atoms (a “C3-C5 cycloalkyl”). Examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, 1-cyclohexenyl, 3-cyclohexenyl, cycloheptyl, norbornyl, and the like.
  • “Halo” or “halogen” refers to elements of the Group 17 series having atomic number 9 to 85. Preferred halo groups include fluoro, chloro, bromo and iodo. Where a residue is substituted with more than one halogen, it may be referred to by using a prefix corresponding to the number of halogen moieties attached, e.g., dihaloaryl, dihaloalkyl, trihaloaryl etc. refer to aryl and alkyl substituted with two (“di”) or three (“tri”) halo groups, which may be but are not necessarily the same halo; thus 4-chloro-3-fluorophenyl is within the scope of dihaloaryl. An alkyl group in which each hydrogen is replaced with a halo group is referred to as a “perhaloalkyl.” A preferred perhaloalkyl group is trifluoroalkyl (—CF3). Similarly, “perhaloalkoxy” refers to an alkoxy group in which a halogen takes the place of each H in the hydrocarbon making up the alkyl moiety of the alkoxy group. An example of a perhaloalkoxy group is trifluoromethoxy (—OCF3).
  • “Heteroaryl” refers to and includes unsaturated aromatic cyclic groups having from 1 to 10 annular carbon atoms and at least one annular heteroatom, including but not limited to heteroatoms such as nitrogen, oxygen and sulfur, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom(s) are optionally quaternized. A heteroaryl group can be attached to the remainder of the molecule at an annular carbon or at an annular heteroatom. Heteroaryl may contain additional fused rings (e.g., from 1 to 3 rings), including additionally fused aryl, heteroaryl, cycloalkyl, and/or heterocyclyl rings. Examples of heteroaryl groups include, but are not limited to, pyridyl, pyrimidyl, thiophenyl, furanyl, thiazolyl, and the like.
  • “Heterocycle” or “heterocyclyl” refers to a saturated or an unsaturated non-aromatic group having from 1 to 10 annular carbon atoms and from 1 to 4 annular heteroatoms, such as nitrogen, sulfur or oxygen, and the like, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom(s) are optionally quaternized. A heterocyclyl group may have a single ring or multiple condensed rings, but excludes heteroaryl groups. A heterocycle comprising more than one ring may be fused, spiro or bridged, or any combination thereof. In fused ring systems, one or more of the fused rings can be aryl or heteroaryl. Examples of heterocyclyl groups include, but are not limited to, tetrahydropyranyl, dihydropyranyl, piperidinyl, piperazinyl, pyrrolidinyl, thiazolinyl, thiazolidinyl, tetrahydrofuranyl, tetrahydrothiophenyl, 2,3-dihydrobenzo[b]thiophen-2-yl, 4-amino-2-oxopyrimidin-1(2H)-yl, and the like.
  • “Oxo” refers to the moiety ═O.
  • “Optionally substituted” unless otherwise specified means that a group may be unsubstituted or substituted by one or more (e.g., 1, 2, 3, 4 or 5) of the substituents listed for that group in which the substituents may be the same of different. In one embodiment, an optionally substituted group has one substituent. In another embodiment, an optionally substituted group has two substituents. In another embodiment, an optionally substituted group has three substituents. In another embodiment, an optionally substituted group has four substituents. In some embodiments, an optionally substituted group has 1 to 2, 2 to 5, 3 to 5, 2 to 3, 2 to 4, 3 to 4, 1 to 3, 1 to 4 or 1 to 5 substituents.
  • A “pharmaceutically acceptable carrier” refers to an ingredient in a pharmaceutical formulation, other than an active ingredient, which is nontoxic to a subject. A pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, or preservative.
  • As used herein, “treatment” or “treating” is an approach for obtaining beneficial or desired results including clinical results. For example, beneficial or desired results include, but are not limited to, one or more of the following: decreasing symptoms resulting from the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, delaying the progression of the disease, and/or prolonging survival of individuals. In reference to cancers or other unwanted cell proliferation, beneficial or desired results include shrinking a tumor (reducing tumor size); decreasing the growth rate of the tumor (such as to suppress tumor growth); reducing the number of cancer cells; inhibiting, retarding or slowing to some extent and preferably stopping cancer cell infiltration into peripheral organs; inhibiting (slowing to some extent and preferably stopping) tumor metastasis; inhibiting tumor growth; preventing or delaying occurrence and/or recurrence of tumor; and/or relieving to some extent one or more of the symptoms associated with the cancer. In some embodiments, beneficial or desired results include preventing or delaying occurrence and/or recurrence, such as of unwanted cell proliferation.
  • As used herein, “delaying development of a disease” means to defer, hinder, slow, retard, stabilize, and/or postpone development of the disease (such as cancer). This delay can be of varying lengths of time, depending on the history of the disease and/or individual being treated. As is evident to one skilled in the art, a sufficient or significant delay can, in effect, encompass prevention, in that the individual does not develop the disease. For example, a late stage cancer, such as development of metastasis, may be delayed.
  • As used herein, an “effective dosage” or “effective amount” of compound or salt thereof or pharmaceutical composition is an amount sufficient to effect beneficial or desired results. For prophylactic use, beneficial or desired results include results such as eliminating or reducing the risk, lessening the severity of, or delaying the onset of the disease, including biochemical, histological and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease. For therapeutic use, beneficial or desired results include ameliorating, palliating, lessening, delaying or decreasing one or more symptoms resulting from the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, enhancing effect of another medication such as via targeting, delaying the progression of the disease, and/or prolonging survival. In reference to cancers or other unwanted cell proliferation, an effective amount comprises an amount sufficient to cause a tumor to shrink and/or to decrease the growth rate of the tumor (such as to suppress tumor growth) or to prevent or delay other unwanted cell proliferation. In some embodiments, an effective amount is an amount sufficient to delay development. In some embodiments, an effective amount is an amount sufficient to prevent or delay occurrence and/or recurrence. An effective amount can be administered in one or more administrations, in the case of cancer, the effective amount of the drug or composition may: (i) reduce the number of cancer cells; (ii) reduce tumor size; (iii) inhibit, retard, slow to some extent and preferably stop cancer cell infiltration into peripheral organs; (iv) inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; (v) inhibit tumor growth; (vi) prevent or delay occurrence and/or recurrence of tumor; and/or (vii) relieve to some extent one or more of the symptoms associated with the cancer. An effective dosage can be administered in one or more administrations. For purposes of this disclosure, an effective dosage of compound or a salt thereof, or pharmaceutical composition is an amount sufficient to accomplish prophylactic or therapeutic treatment either directly or indirectly. It is intended and understood that an effective dosage of a compound or salt thereof, or pharmaceutical composition may or may not be achieved in conjunction with another drug, compound, or pharmaceutical composition. Thus, an “effective dosage” may be considered in the context of administering one or more therapeutic agents, and a single agent may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable result may be or is achieved.
  • As used herein, the term “individual” is a mammal, including humans. An individual includes, but is not limited to, human, bovine, horse, feline, canine, rodent, or primate. In some embodiments, the individual is human. The individual (such as a human) may have advanced disease or lesser extent of disease, such as low tumor burden. In some embodiments, the individual is at an early stage of a proliferative disease (such as cancer). In some embodiments, the individual is at an advanced stage of a proliferative disease (such as an advanced cancer).
  • Reference to “about” a value or parameter herein includes (and describes) embodiments that are directed to that value or parameter per se. For example, description referring to “about X” includes description of “X”.
  • It is understood that aspects and variations described herein also include “consisting” and/or “consisting essentially of” aspects and variations.
  • Compounds
  • In one aspect, provided is a compound of Formula (I):
  • Figure US20230109104A1-20230406-C00003
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein:
  • X1 is N or CR;
  • X2 is N or CR, wherein at least one of X1 and X2 is N;
  • each R is independently hydrogen, C1-C6 alkyl, halogen or CN;
  • A is phenyl or 5- to 6-membered heteroaryl, each of which is optionally substituted by R1;
  • B is C6-C14 aryl, 5- to 10-membered heteroaryl, C3-C8 cycloalkyl, or 3- to 12-membered heterocyclyl, each of which is optionally substituted by R4, wherein A is fused to B;
  • m is 0, 1, 2, 3, or 4;
  • n is 0, 1, 2, 3, or 4;
  • R1 is independently halogen, —CN, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, —NR17R18 or —C(O)NR17R18;
  • R2 is independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR10, —(C1-C3 alkylene)NR11R12, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R10, —(C1-C3 alkylene)C(O)NR11R12, —(C2-C3 alkylene)NR10C(O)R11, —(C1-C3 alkylene)S(O)2R10, —(C2-C3 alkylene)NR10S(O)2R11, —(C1-C3 alkylene)S(O)2NR11R12, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3- to 12-membered heterocyclyl), —(C1-C3 alkylene)(5- to 10-membered heteroaryl), or —(C1-C3 alkylene)(C6-C14 aryl), wherein each R2 is independently optionally substituted by halogen, oxo, —OR13, C1-C6 alkyl, or —NR13R14;
  • R3 is independently hydrogen, C1-C6 alkyl, —C(O)R10, —S(O)R10, —S(O)2R10, C3-C6 cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, C6-C14 aryl, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3- to 12-membered heterocyclyl), —(C1-C3 alkylene)(5- to 10-membered heteroaryl), or —(C1-C3 alkylene)(C6-C14 aryl), wherein each R3 is independently optionally substituted by halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that the 5- to 10-membered heteroaryl and C6-C14 aryl of R3 is not substituted by
  • Figure US20230109104A1-20230406-C00004
  • each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR17S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl) or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19,
  • or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19;
  • R10 is independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl, or 3- to 6-membered heterocyclyl, wherein the C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl and 3- to 6-membered heterocyclyl are independently optionally substituted by halogen, oxo, —CN, —OR15, —NR15R16, —P(O)(OR15)(OR16), phenyl optionally substituted by halogen, or C1-C6 alkyl optionally substituted by halogen, —OH or oxo;
  • R11 and R12 are each independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl, or 3- to 6-membered heterocyclyl, wherein the C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl and 3- to 6-membered heterocyclyl are independently optionally substituted by halogen, oxo, —CN, —OR15, —NR15R16, or C1-C6 alkyl optionally substituted by halogen, —OH or oxo,
  • or R11 and R12 are taken together with the atom to which they attached to form a 3- to 6-membered heterocyclyl optionally substituted by halogen, oxo, —OR15, —NR15R16 or C1-C6 alkyl optionally substituted by halogen, oxo or —OH;
  • R13 and R14 are each independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl, 3- to 6-membered heterocyclyl, —C(O)OR15, —(C1-C3 alkylene)OR15 or —C(O)NR15R16, wherein the C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl and 3- to 6-membered heterocyclyl are independently optionally substituted by halogen, oxo, —CN, —OR15, —NR15R16, or C1-C6 alkyl optionally substituted by halogen, —OH or oxo,
  • or R13 and R14 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl optionally substituted by halogen or C1-C6 alkyl optionally substituted by halogen or oxo;
  • R15 and R16 are each independently hydrogen, C1-C6 alkyl optionally substituted by halogen or oxo, C2-C6 alkenyl optionally substituted by halogen or oxo, or C2-C6 alkynyl optionally substituted by halogen or oxo,
  • or R15 and R16 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl optionally substituted by halogen, oxo or C1-C6 alkyl optionally substituted by oxo or halogen; and
  • each R17, R18, R19, and R20 is independently hydrogen, C3-C6 cycloalkyl, 3-6 membered heterocyclyl or C1-C6 alkyl, each of which is optionally substituted by halogen, oxo or —OH,
  • or R17 and R18 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl optionally substituted by halogen, oxo or —OH.
  • In some embodiments of a compound of Formula (I), B is C6 aryl, 5- to 7-membered heteroaryl, C3-C5 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is optionally substituted by R4, wherein B is fused to A.
  • In some embodiments, provided is a compound of Formula (Ia):
  • Figure US20230109104A1-20230406-C00005
  • or a salt thereof, wherein:
  • A is phenyl or 6-membered heteroaryl, each of which is optionally substituted by R1;
  • B is C6 aryl, 5- to 6-membered heteroaryl, C3-C5 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is optionally substituted by R4, wherein A is fused to B;
  • m is 0, 1, 2, 3, or 4;
  • n is 0, 1, 2, 3, or 4;
  • R1 is independently halogen, —CN, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, or —C(O)NR17R18;
  • R2 is independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR10, —(C1-C3 alkylene)NR11R12, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R10, —(C1-C3 alkylene)C(O)NR11R12, —(C2-C3 alkylene)NR10C(O)R11, —(C1-C3 alkylene)S(O)2R10, —(C2-C3 alkylene)NR10S(O)2R11, —(C1-C3 alkylene)S(O)2NR11R12, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3- to 12-membered heterocyclyl), —(C1-C3 alkylene)(5- to 10-membered heteroaryl), or —(C1-C3 alkylene)(C6-C14 aryl), wherein each R2 is independently optionally substituted by halogen, oxo, —OR13, C1-C6 alkyl, or —NR13R14;
  • R3 is independently hydrogen, C1-C6 alkyl, —C(O)R10, —S(O)R10, —S(O)2R10, C3-C6 cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, C6-C14 aryl, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3- to 12-membered heterocyclyl), —(C1-C3 alkylene)(5- to 10-membered heteroaryl), or —(C1-C3 alkylene)(C6-C14 aryl), wherein each R3 is independently optionally substituted by halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)NR13R14, —NR13C(O)R14, —CN, —S(O)R13, —S(O)2R13, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH or halogen;
  • each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, or —C(O)NR17R18, wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19,
  • or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl;
  • R10 is independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl, or 3- to 6-membered heterocyclyl, wherein the C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl and 3- to 6-membered heterocyclyl are independently optionally substituted by halogen, oxo, —CN, —OR15, —NR15R16, —P(O)(OR15)(OR16), phenyl optionally substituted by halogen, or C1-C6 alkyl optionally substituted by halogen, —OH or oxo;
  • R11 and R12 are each independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl, or 3- to 6-membered heterocyclyl, wherein the C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl and 3- to 6-membered heterocyclyl are independently optionally substituted by halogen, oxo, —CN, —OR15, —NR15R16, or C1-C6 alkyl optionally substituted by halogen, —OH or oxo,
  • or R11 and R12 are taken together with the atom to which they attached to form a 3- to 6-membered heterocyclyl optionally substituted by halogen, oxo, —OR15, —NR15R16 or C1-C6 alkyl optionally substituted by halogen, oxo or —OH;
  • R13 and R14 are each independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl, or 3- to 6-membered heterocyclyl, wherein the C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl and 3- to 6-membered heterocyclyl are independently optionally substituted by halogen, oxo, —CN, —OR15, —NR15R16, or C1-C6 alkyl optionally substituted by halogen, —OH or oxo,
  • or R13 and R14 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl optionally substituted by halogen or C1-C6 alkyl optionally substituted by halogen or oxo;
  • R15 and R16 are each independently hydrogen, C1-C6 alkyl optionally substituted by halogen or oxo, C2-C6 alkenyl optionally substituted by halogen or oxo, or C2-C6 alkynyl optionally substituted by halogen or oxo;
  • or R15 and R16 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl optionally substituted by halogen, oxo or C1-C6 alkyl optionally substituted by oxo or halogen; and
  • each R17, R18, R19, and R20 is independently hydrogen or C1-C6 alkyl.
  • In some embodiments, provided is a compound of Formula (Ia), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing.
  • In some embodiments of a compound of Formula (Ia), the the 5- to 10-membered heteroaryl and C6-C14 aryl of R3 is not substituted by
  • Figure US20230109104A1-20230406-C00006
  • In some embodiments, the compound of Formula (I) is of Formula (Ib):
  • Figure US20230109104A1-20230406-C00007
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein m, n, A, B, R1, R2, R3 and R4 are as detailed herein. In some embodiments of a compound of Formula (Ib), B is C6 aryl, 5- to 7-membered heteroaryl, C3-C5 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is optionally substituted by R4, wherein B is fused to A.
  • In some embodiments, the compound of Formula (I) is of Formula (Ic):
  • Figure US20230109104A1-20230406-C00008
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein m, n, A, B, R1, R2, R3 and R4 are as detailed herein. In some embodiments of a compound of Formula (I), X1 is N. In some embodiments of a compound of Formula (Ic), B is C6 aryl, 5- to 7-membered heteroaryl, C3-C5 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is optionally substituted by R4, wherein B is fused to A.
  • In some embodiments of a compound of Formula (I), X1 is CR. In some embodiments of a compound of Formula (I), X1 is CH. In some embodiments, X1 is N. In some embodiments of a compound of Formula (I), X2 is N. In some embodiments of a compound of Formula (I), X2 is CR. In some embodiments of a compound of Formula (I), X2 is CH.
  • In some embodiments of a compound of Formula (I), X1 is N and X2 is CR. In some embodiments of a compound of Formula (I), X1 is N and X2 is CH. In some embodiments of a compound of Formula (I), X1 is CR and X2 is N. In some embodiments of a compound of Formula (I), X1 is CH and X2 is N. In some embodiments of a compound of Formula (I), X1 and X2 both are N.
  • In some embodiments of a compound of Formula (I), R is hydrogen, C1-C6 alkyl, halogen or CN. In some embodiments of a compound of Formula (I), R is hydrogen. In some embodiments of a compound of Formula (I), R is C1-C6 alkyl. In some embodiments of a compound of Formula (I), R is methyl. In some embodiments of a compound of Formula (I), R is halogen. In some embodiments of a compound of Formula (I), R is CN.
  • In some embodiments of a compound of Formula (I), A is phenyl optionally substituted by R1. In some embodiments of a compound of Formula (I), A is unsubstituted phenyl. In some embodiments of a compound of Formula (I), A is phenyl substituted by R1.
  • In some embodiments of a compound of Formula (I), A is 5- to 6-membered heteroaryl optionally substituted by R1. In some embodiments of a compound of Formula (I), A is 6-membered heteroaryl optionally substituted by R1. In some embodiments of a compound of Formula (I), A is unsubstituted 6-membered heteroaryl. In some embodiments of a compound of Formula (I), A is 6-membered heteroaryl substituted by R1. In some embodiments of a compound of Formula (I), A is unsubstituted 5-membered heteroaryl. In some embodiments of a compound of Formula (I), A is 5-membered heteroaryl substituted by R1.
  • In some embodiments of a compound of Formula (I), A is selected from the group consisting of:
  • Figure US20230109104A1-20230406-C00009
  • wherein the wavy lines denote attachment points to the parent molecule and each is fused with B and is optionally substituted by R1.
  • In some embodiments of a compound of Formula (I), A is selected from the group consisting of:
  • Figure US20230109104A1-20230406-C00010
  • wherein the wavy lines denote attachment points to the parent molecule and each is fused with B and is optionally substituted by R1.
  • In some embodiments of a compound of Formula (I), R1 is independently halogen, —CN, oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, —NR17R18 or —C(O)NR17R18. In some embodiments of a compound of Formula (I), R1 is independently halogen, —CN, oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, or —C(O)NR17R18. In some embodiments of a compound of Formula (I), R1 is independently —CN, C1-C6 alkyl, or halogen. In some embodiments of a compound of Formula (I), R1 is —CN. In some embodiments of a compound of Formula (I), R1 is C1-C6 alkyl. In some embodiments of a compound of Formula (I), R1 is halogen.
  • In some embodiments of a compound of Formula (I), A is phenyl optionally substituted by R1, and R1 is independently —CN, C1-C6 alkyl, or halogen. In some embodiments of a compound of Formula (I), A is 6-membered heteroaryl optionally substituted by R1, and R1 is independently —CN, C1-C6 alkyl, or halogen.
  • In some embodiments of a compound of Formula (I), A is selected from the group consisting of:
  • Figure US20230109104A1-20230406-C00011
  • wherein the wavy lines denote attachment points to the parent molecule and each is fused with B and optionally substituted by R1, wherein R1 is independently —CN, C1-C6 alkyl, or halogen.
  • In some embodiments of a compound of Formula (I), B is C6 aryl, 5- to 7-membered heteroaryl, C3-C5 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is unsubstituted. In some embodiments of a compound of Formula (I), B is C6 aryl, 5- to 7-membered heteroaryl, C3-C8 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is substituted with R4. In some embodiments of a compound of Formula (I), B is C6 aryl or 3- to 8-membered heterocyclyl, each of which is unsubstituted. In some embodiments of a compound of Formula (I), B is C6 aryl or 3- to 8-membered heterocyclyl, each of which is optionally substituted with R4.
  • In some embodiments of a compound of Formula (I), B is C6 aryl which is optionally substituted by R4. In some embodiments of a compound of Formula (I), B is C6 aryl which is unsubstituted. In some embodiments of a compound of Formula (I), B is 5- to 7-membered heteroaryl which is optionally substituted by R4. In some embodiments of a compound of Formula (I), B is 5- to 7-membered heteroaryl which is unsubstituted. In some embodiments of a compound of Formula (I), B is C3-C8 cycloalkyl which is optionally substituted by R4. In some embodiments of a compound of Formula (I), B is C3-C5 cycloalkyl which is unsubstituted. In some embodiments of a compound of Formula (I), B is 3- to 7-membered heterocyclyl which is optionally substituted by R4. In some embodiments of a compound of Formula (I), B is 3- to 8-membered heterocyclyl which is optionally substituted by R4. In some embodiments of a compound of Formula (I), B is 3- to 8-membered heterocyclyl which is unsubstituted.
  • In some embodiments of a compound of Formula (I), B is C6 aryl, 5- to 7-membered heteroaryl, C3-C5 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is substituted with R4 and each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR17S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl) or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments of a compound of Formula (I), B is C6 aryl, 5- to 7-membered heteroaryl, C3-C5 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is substituted with R4, wherein R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments of a compound of Formula (I), B is C6 aryl optionally substituted with R4, wherein R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments of a compound of Formula (I), B is 5- to 7-membered heteroaryl optionally substituted with R4, wherein R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R9. In some embodiments of a compound of Formula (I), B is C3-C8 cycloalkyl optionally substituted with R4, wherein R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments of a compound of Formula (I), B is 3- to 8-membered heterocyclyl optionally substituted with R4, wherein R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments of a compound of Formula (I), B is 6-membered heterocyclyl optionally substituted by R4, wherein R4 is independently C1-C6 alkyl. In some embodiments of a compound of Formula (I), B is 6-membered heterocyclyl having one nitrogen atom, wherein the 6-membered heterocyclyl is optionally substituted by R4 and R4 is independently C1-C6 alkyl.
  • In some embodiments of a compound of Formula (I), B is C6 aryl, 5- to 7-membered heteroaryl, C3-C5 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is substituted with R4 and each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, or —C(O)NR17R18, wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl. In some embodiments of a compound of Formula (I), B is C6 aryl, 5- to 7-membered heteroaryl, C3-C5 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is substituted with R4, wherein R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20 or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl. In some embodiments of a compound of Formula (I), B is C6 aryl optionally substituted with R4, wherein R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl. In some embodiments of a compound of Formula (I), B is 5- to 7-membered heteroaryl optionally substituted with R4, wherein R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl. In some embodiments of a compound of Formula (I), B is C3-C8 cycloalkyl optionally substituted with R4, wherein R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl. In some embodiments of a compound of Formula (I), B is 3- to 8-membered heterocyclyl optionally substituted with R4, wherein R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl. In some embodiments of a compound of Formula (I), B is 6-membered heterocyclyl optionally substituted by R4, wherein R4 is independently C1-C6 alkyl. In some embodiments of a compound of Formula (I), B is 6-membered heterocyclyl having one nitrogen atom, wherein the 6-membered heterocyclyl is optionally substituted by R4 and R4 is independently C1-C6 alkyl.
  • In some embodiments of a compound of Formula (I), A is unsubstituted phenyl fused with B, wherein B is C6 aryl, 5- to 7-membered heteroaryl, C3-C5 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is substituted with R4; R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, or —C(O)NR17R18, wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments of a compound of Formula (I), A is unsubstituted phenyl fused with B, wherein B is C6 aryl, 5- to 7-membered heteroaryl, C3-C5 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is substituted with R4; R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, or —C(O)NR17R18, wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl. In some embodiments of a compound of Formula (I), A is unsubstituted phenyl fused with B, wherein B is 3- to 8-membered heterocyclyl optionally substituted with R4; R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments of a compound of Formula (I), A is unsubstituted phenyl fused with B, wherein B is 3- to 8-membered heterocyclyl optionally substituted with R4; R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl. In some embodiments of a compound of Formula (I), A is unsubstituted phenyl fused with B, wherein B is 6-membered heterocyclyl having one nitrogen atom, wherein the 6-membered heterocyclyl is optionally substituted by R4; R4 is independently C1-C6 alkyl.
  • In some embodiments of a compound of Formula (I), A is phenyl optionally substituted by R1 and fused with B, wherein R1 is independently —CN, C1-C6 alkyl, or halogen; B is C6 aryl, 5- to 7-membered heteroaryl, C3-C8 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is substituted with R4; R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR7S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR17S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl) or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments of a compound of Formula (I), A is phenyl optionally substituted by R1 and fused with B, wherein R1 is independently —CN, C1-C6 alkyl, or halogen; B is 3- to 8-membered heterocyclyl optionally substituted with R4; R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19.
  • In some embodiments of a compound of Formula (I), A is phenyl optionally substituted by R1 and fused with B, wherein R1 is independently —CN, C1-C6 alkyl, or halogen; B is C6 aryl, 5- to 7-membered heteroaryl, C3-C8 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is substituted with R4; R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, or —C(O)NR17R18, wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl. In some embodiments of a compound of Formula (I), A is phenyl optionally substituted by R1 and fused with B, wherein R1 is independently —CN, C1-C6 alkyl, or halogen; B is 3- to 8-membered heterocyclyl optionally substituted with R4; R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl.
  • In some embodiments of a compound of Formula (I), A is unsubstituted 6-membered heteroaryl fused with B, wherein B is C6 aryl, 5- to 7-membered heteroaryl, C3-C5 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is substituted with R4; R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR17S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl), or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments of a compound of Formula (I), A is unsubstituted 6-membered heteroaryl fused with B, wherein B is 3- to 8-membered heterocyclyl optionally substituted with R4; R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments of a compound of Formula (I), A is unsubstituted 6-membered heteroaryl fused with B, wherein B is 6-membered heterocyclyl having one nitrogen atom, wherein the 6-membered heterocyclyl is optionally substituted by R4; R4 is independently C1-C6 alkyl.
  • In some embodiments of a compound of Formula (I), A is unsubstituted 6-membered heteroaryl fused with B, wherein B is C6 aryl, 5- to 7-membered heteroaryl, C3-C5 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is substituted with R4; R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, or —C(O)NR17R18, wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl. In some embodiments of a compound of Formula (I), A is unsubstituted 6-membered heteroaryl fused with B, wherein B is 3- to 8-membered heterocyclyl optionally substituted with R4; R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl. In some embodiments of a compound of Formula (I), A is unsubstituted 6-membered heteroaryl fused with B, wherein B is 6-membered heterocyclyl having one nitrogen atom, wherein the 6-membered heterocyclyl is optionally substituted by R4; R4 is independently C1-C6 alkyl.
  • In some embodiments, provided is a compound of Formula (Ia-1):
  • Figure US20230109104A1-20230406-C00012
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein n is 0, 1, 2, 3, or 4, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR17S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl) or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R4 is independently oxo, C1-C6 alkyl or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17, wherein R17 is C1-C6 alkyl optionally substituted with —OH.
  • In some embodiments, provided is a compound of Formula (Ia-1):
  • Figure US20230109104A1-20230406-C00013
  • or a salt thereof, wherein n is 0, 1, 2, 3, or 4, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, or —C(O)NR17R18, wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl. In some embodiments, R4 is independently oxo, C1-C6 alkyl or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17, wherein R17 is C1-C6 alkyl optionally substituted with —OH.
  • In some embodiments, provided is a compound of Formula (Ia-2):
  • Figure US20230109104A1-20230406-C00014
  • or salt thereof, wherein Y is hydrogen or R4, and n is 0, 1, 2, 3, or 4. In some embodiments of a compound of Formula (Ia-2), Y is hydrogen. In some embodiments of a compound of Formula (Ia-2), Y is R4. In some embodiments, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR7S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR7S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl), or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R4 is independently oxo, C1-C6 alkyl or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17, wherein R17 is C1-C6 alkyl optionally substituted with —OH. In some embodiments, Y is hydrogen, R4 is independently C1-C6 alkyl and n is 2. In some embodiments, Y is hydrogen, R4 is methyl and n is 2.
  • In some embodiments, provided is a compound of Formula (Ia-2):
  • Figure US20230109104A1-20230406-C00015
  • or salt thereof, wherein Y is hydrogen or R4, and n is 0, 1, 2, 3, or 4. In some embodiments, Y is hydrogen. In some embodiments, Y is R4. In some embodiments, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, or —C(O)NR17R18, wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl. In some embodiments, R4 is independently oxo, C1-C6 alkyl or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17, wherein R17 is C1-C6 alkyl optionally substituted with —OH. In some embodiments, Y is hydrogen, R4 is independently C1-C6 alkyl and n is 2. In some embodiments, Y is hydrogen, R4 is methyl and n is 2.
  • In some embodiments, provided is a compound of Formula (Ia-3):
  • Figure US20230109104A1-20230406-C00016
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein Y is hydrogen or R4, and n is 0, 1, 2, 3, or 4. In some embodiments of a compound of Formula (Ia-3), Y is hydrogen. In some embodiments of a compound of Formula (Ia-3), Y is R4. In some embodiments, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR17S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl) or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R4 is independently oxo, C1-C6 alkyl or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17, wherein R17 is C1-C6 alkyl optionally substituted with —OH. In some embodiments, Y is hydrogen and n is 0.
  • In some embodiments, provided is a compound of Formula (Ia-3):
  • Figure US20230109104A1-20230406-C00017
  • or a salt thereof, wherein Y is hydrogen or R4, and n is 0, 1, 2, 3, or 4. In some embodiments, Y is hydrogen. In some embodiments, Y is R4. In some embodiments, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, or —C(O)NR17R18, wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl. In some embodiments, R4 is independently oxo, C1-C6 alkyl or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17, wherein R17 is C1-C6 alkyl optionally substituted with —OH. In some embodiments, Y is hydrogen and n is 0.
  • In some embodiments, provided is a compound of Formula (Ia-4):
  • Figure US20230109104A1-20230406-C00018
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein n is 0, 1, 2, 3, or 4, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR7S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR17S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl), or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R4 is independently oxo, C1-C6 alkyl or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17, wherein R17 is C1-C6 alkyl optionally substituted with —OH.
  • In some embodiments, provided is a compound of Formula (Ia-4):
  • Figure US20230109104A1-20230406-C00019
  • or a salt thereof, wherein n is 0, 1, 2, 3, or 4, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, or —C(O)NR17R18, wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl. In some embodiments, R4 is independently oxo, C1-C6 alkyl or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17, wherein R17 is C1-C6 alkyl optionally substituted with —OH.
  • In some embodiments, provided is a compound of Formula (Ia-5):
  • Figure US20230109104A1-20230406-C00020
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein n is 0, 1, 2, 3, or 4, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR7S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR17S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl) or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R4 is independently oxo, C1-C6 alkyl or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl each is optionally substituted by R19. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17, wherein R17 is C1-C6 alkyl optionally substituted with —OH.
  • In some embodiments, provided is a compound of Formula (Ia-5):
  • Figure US20230109104A1-20230406-C00021
  • or a salt thereof, wherein n is 0, 1, 2, 3, or 4, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, or —C(O)NR17R18, wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl. In some embodiments, R4 is independently oxo, C1-C6 alkyl or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17, wherein R17 is C1-C6 alkyl optionally substituted with —OH.
  • In some embodiments, provided is a compound of Formula (Ia-6):
  • Figure US20230109104A1-20230406-C00022
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein n is 0, 1, 2, 3, or 4, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR7S(O)2R′8, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR7S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl) or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R4 is independently oxo, C1-C6 alkyl or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17, wherein R17 is C1-C6 alkyl optionally substituted with —OH.
  • In some embodiments, provided is a compound of Formula (Ia-6):
  • Figure US20230109104A1-20230406-C00023
  • or a salt thereof, wherein n is 0, 1, 2, 3, or 4, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, or —C(O)NR17R18, wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl. In some embodiments, R4 is independently oxo, C1-C6 alkyl or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17, wherein R17 is C1-C6 alkyl optionally substituted with —OH.
  • In some embodiments, provided is a compound of Formula (Ia-7):
  • Figure US20230109104A1-20230406-C00024
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein Y is hydrogen or R4, n and p are independently 0, 1, 2, 3, or 4; R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00025
  • In some embodiments of a compound of Formula (Ia-7), Y is hydrogen. In some embodiments of a compound of Formula (Ia-7), Y is R4. In some embodiments, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —Si(C1-C6 alkyl)3, —CN, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR7S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl), or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4 when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R3a is C3-C5 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00026
  • In some embodiments, R3a is C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen.
  • In some embodiments, provided is a compound of Formula (Ia-8):
  • Figure US20230109104A1-20230406-C00027
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein Y is hydrogen or R4, n and p are independently 0, 1, 2, 3, or 4; R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00028
  • In some embodiments of a compound of Formula (Ia-8), Y is hydrogen. In some embodiments of a compound of Formula (Ia-8), Y is R4. In some embodiments, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR17S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl), or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4 when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R3a is C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR5R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00029
  • In some embodiments, R3a is C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen.
  • In some embodiments, provided is a compound of Formula (Ia-9):
  • Figure US20230109104A1-20230406-C00030
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein Y is hydrogen or R4, n and p are independently 0, 1, 2, 3, or 4; R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00031
  • In some embodiments of a compound of Formula (Ia-9), Y is hydrogen. In some embodiments of a compound of Formula (Ia-9), Y is R4. In some embodiments, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR17S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl), or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4 when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R3a is C3-C5 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted b oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00032
  • In some embodiments, R3a is C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen.
  • In some embodiments, provided is a compound of Formula (Ia-10):
  • Figure US20230109104A1-20230406-C00033
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein Y is hydrogen or R4, n and p are independently 0, 1, 2, 3, or 4; R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00034
  • In some embodiments of a compound of Formula (Ia-10), Y is hydrogen. In some embodiments of a compound of Formula (Ia-10), Y is R4. In some embodiments, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR7S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR7S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl), or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4 when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R3a is C3-C5 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00035
  • In some embodiments, R3a is C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen.
  • In some embodiments, provided is a compound of Formula (Ia-11):
  • Figure US20230109104A1-20230406-C00036
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein Y is hydrogen or R4, n and p are independently 0, 1, 2, 3, or 4; R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00037
  • In some embodiments of a compound of Formula (Ia-11), Y is hydrogen. In some embodiments of a compound of Formula (Ia-11), Y is R4. In some embodiments, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR7S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl), or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4 when bound to the same carbon or two different carbon atoms, are taken together with the carbon to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R3a is halogen, C3-C5 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00038
  • In some embodiments, R3a is halogen. In some embodiments, R3a is C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen.
  • In some embodiments, provided is a compound of Formula (Ia-12
  • Figure US20230109104A1-20230406-C00039
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein Y is hydrogen or R4, n and p are independently 0, 1, 2, 3, or 4; R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00040
  • In some embodiments of a compound of Formula (Ia-12), Y is hydrogen. In some embodiments of a compound of Formula (Ia-12), Y is R4. In some embodiments, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR7S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR7S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl), or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4 when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R3a is halogen, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00041
  • In some embodiments, R3a is halogen; In some embodiments, R3a is C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen.
  • In some embodiments, provided is a compound of Formula (Ib-1):
  • Figure US20230109104A1-20230406-C00042
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein n is 0, 1, 2, 3, or 4, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR17S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl) or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R4 is independently oxo, C1-C6 alkyl or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17, wherein R17 is C1-C6 alkyl optionally substituted with —OH.
  • In some embodiments, provided is a compound of Formula (Ib-2):
  • Figure US20230109104A1-20230406-C00043
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein Y is hydrogen or R4, and n is 0, 1, 2, 3, or 4. In some embodiments of a compound of Formula (Ib-2), Y is hydrogen. In some embodiments of a compound of Formula (Ib-2), Y is R4. In some embodiments, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR17S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl), or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R4 is independently oxo, C1-C6 alkyl or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17, wherein R17 is C1-C6 alkyl optionally substituted with —OH. In some embodiments, Y is hydrogen, R4 is independently C1-C6 alkyl and n is 2. In some embodiments, Y is hydrogen, R4 is methyl and n is 2.
  • In some embodiments, provided is a compound of Formula (Ib-3):
  • Figure US20230109104A1-20230406-C00044
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein Y is hydrogen or R4, and n is 0, 1, 2, 3, or 4. In some embodiments of a compound of Formula (Ib-3), Y is hydrogen. In some embodiments of a compound of Formula (Ib-3), Y is R4. In some embodiments, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR17S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl) or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R4 is independently oxo, C1-C6 alkyl or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17, wherein R17 is C1-C6 alkyl optionally substituted with —OH. In some embodiments, Y is hydrogen and n is 0.
  • In some embodiments, provided is a compound of Formula (Ib-4):
  • Figure US20230109104A1-20230406-C00045
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein Y is hydrogen or R4, n and p are independently 0, 1, 2, 3, or 4; R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00046
  • In some embodiments of a compound of Formula (Ib-4), Y is hydrogen. In some embodiments of a compound of Formula (Ib-4), Y is R4. In some embodiments, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —Si(C1-C6 alkyl)3, —CN, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR17S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl), or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4 when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R3a is C3-C5 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00047
  • In some embodiments, R3a is C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen.
  • In some embodiments, provided is a compound of Formula (Ib-5):
  • Figure US20230109104A1-20230406-C00048
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein Y is hydrogen or R4, n and p are independently 0, 1, 2, 3, or 4; R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00049
  • In some embodiments of a compound of Formula (Ib-5), Y is hydrogen. In some embodiments of a compound of Formula (Ib-5), Y is R4. In some embodiments, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR7S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl), or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4 when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R3a is C3-C5 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00050
  • In some embodiments, R3a is C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen.
  • In some embodiments, provided is a compound of Formula (Ib-6):
  • Figure US20230109104A1-20230406-C00051
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein Y is hydrogen or R4, n and p are independently 0, 1, 2, 3, or 4; R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00052
  • In some embodiments of a compound of Formula (Ib-6), Y is hydrogen. In some embodiments of a compound of Formula (Ib-6), Y is R4. In some embodiments, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR17S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl), or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4 when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R3a is C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00053
  • In some embodiments, R3a is C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen.
  • In some embodiments, provided is a compound of Formula (Ib-7):
  • Figure US20230109104A1-20230406-C00054
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein Y is hydrogen or R4, n and p are independently 0, 1, 2, 3, or 4; R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00055
  • In some embodiments of a compound of Formula (Ib-7), Y is hydrogen. In some embodiments of a compound of Formula (Ib-7), Y is R4. In some embodiments, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, alogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR17S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl), or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4 when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R3a is C3-C5 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00056
  • In some embodiments, R3a is C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen.
  • In some embodiments, provided is a compound of Formula (Ib-8):
  • Figure US20230109104A1-20230406-C00057
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein Y is hydrogen or R4, n and p are independently 0, 1, 2, 3, or 4; R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00058
  • In some embodiments of a compound of Formula (Ib-8), Y is hydrogen. In some embodiments of a compound of Formula (Ib-8), Y is R4. In some embodiments, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR7S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl), or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4 when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R3a is halogen, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00059
  • In some embodiments, R3a is halogen; In some embodiments, R3a is C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen.
  • In some embodiments, provided is a compound of Formula (Ib-9):
  • Figure US20230109104A1-20230406-C00060
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein Y is hydrogen or R4, n and p are independently 0, 1, 2, 3, or 4; R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR5, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00061
  • In some embodiments of a compound of Formula (Ib-9), Y is hydrogen. In some embodiments of a compound of Formula (Ib-9), Y is R4. In some embodiments, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR7S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl), or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4 when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R3a is halogen, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00062
  • In some embodiments, R3a is halogen. In some embodiments, R3a is C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen.
  • In some embodiments, provided is a compound of Formula (Ic-1):
  • Figure US20230109104A1-20230406-C00063
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein n is 0, 1, 2, 3, or 4, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR7S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR17S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl) or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R4 is independently oxo, C1-C6 alkyl or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17, wherein R17 is C1-C6 alkyl optionally substituted with —OH.
  • In some embodiments, provided is a compound of Formula (Ic-2):
  • Figure US20230109104A1-20230406-C00064
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein Y is hydrogen or R4, and n is 0, 1, 2, 3, or 4. In some embodiments of a compound of Formula (Ic-2), Y is hydrogen. In some embodiments of a compound of Formula (Ic-2), Y is R4. In some embodiments, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR17S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl), or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R4 is independently oxo, C1-C6 alkyl or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17, wherein R17 is C1-C6 alkyl optionally substituted with —OH. In some embodiments, Y is hydrogen, R4 is independently C1-C6 alkyl and n is 2. In some embodiments, Y is hydrogen, R4 is methyl and n is 2.
  • In some embodiments, provided is a compound of Formula (Ic-3):
  • Figure US20230109104A1-20230406-C00065
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein Y is hydrogen or R4, and n is 0, 1, 2, 3, or 4. In some embodiments of a compound of Formula (Ic-3), Y is hydrogen. In some embodiments of a compound of Formula (Ic-3), Y is R4. In some embodiments, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR7S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl) or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R4 is independently oxo, C1-C6 alkyl or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17. In some embodiments, R4 is independently C1-C6 alkyl or —C(O)R17, wherein R17 is C1-C6 alkyl optionally substituted with —OH. In some embodiments, Y is hydrogen and n is 0.
  • In some embodiments, provided is a compound of Formula (Ic-4):
  • Figure US20230109104A1-20230406-C00066
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein Y is hydrogen or R4, n and p are independently 0, 1, 2, 3, or 4; R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00067
  • In some embodiments of a compound of Formula (Ic-4), Y is hydrogen. In some embodiments of a compound of Formula (Ic-4), Y is R4. In some embodiments, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —Si(C1-C6 alkyl)3, —CN, —OR17, —NR17R11, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR7S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl), or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4 when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R3a is C3-C5 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00068
  • In some embodiments, R3a is C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen.
  • In some embodiments, provided is a compound of Formula (Ic-5):
  • Figure US20230109104A1-20230406-C00069
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein Y is hydrogen or R4, n and p are independently 0, 1, 2, 3, or 4; R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or
  • Figure US20230109104A1-20230406-C00070
  • halogen, with the proviso that R3a is not 0. In some embodiments of a compound of Formula (Ic-5), Y is hydrogen. In some embodiments of a compound of Formula (Ic-5), Y is R. In some embodiments, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR17S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl), or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4 when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R3a is C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00071
  • In some embodiments, R3a is C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen.
  • In some embodiments, provided is a compound of Formula (Ic-6):
  • Figure US20230109104A1-20230406-C00072
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein Y is hydrogen or R4, n and p are independently 0, 1, 2, 3, or 4; R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00073
  • In some embodiments of a compound of Formula (Ic-6), Y is hydrogen. In some embodiments of a compound of Formula (Ic-6), Y is R4. In some embodiments, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR7S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl), or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4 when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R3a is C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00074
  • In some embodiments, R3a is C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen.
  • In some embodiments, provided is a compound of Formula (Ic-7):
  • Figure US20230109104A1-20230406-C00075
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein Y is hydrogen or R4, n and p are independently 0, 1, 2, 3, or 4; R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00076
  • In some embodiments of a compound of Formula (Ic-7), Y is hydrogen. In some embodiments of a compound of Formula (Ic-7), Y is R4. In some embodiments, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR7S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl), or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4 when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R3a is C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00077
  • In some embodiments, R3a is C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen.
  • In some embodiments, provided is a compound of Formula (Ic-8):
  • Figure US20230109104A1-20230406-C00078
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein Y is hydrogen or R4, n and p are independently 0, 1, 2, 3, or 4; R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00079
  • In some embodiments of a compound of Formula (Ic-8), Y is hydrogen. In some embodiments of a compound of Formula (Ic-8), Y is R4. In some embodiments, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR17S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl), or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4 when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R3a is halogen, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00080
  • In some embodiments, R3a is halogen; In some embodiments, R3a is C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen.
  • In some embodiments, provided is a compound of Formula (Ic-9):
  • Figure US20230109104A1-20230406-C00081
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein Y is hydrogen or R4, n and p are independently 0, 1, 2, 3, or 4; R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00082
  • In some embodiments of a compound of Formula (Ic-9), Y is hydrogen. In some embodiments of a compound of Formula (Ic-9), Y is R4. In some embodiments, each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR7S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl), or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19, or two R4 when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19. In some embodiments, R3a is halogen, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —OR15 or halogen, with the proviso that R3a is not
  • Figure US20230109104A1-20230406-C00083
  • In some embodiments, R3a is halogen. In some embodiments, R3a is C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen.
  • It is understood that for compounds of formula (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) and (IC-1 to Ic-9), the variables R2 and R3 are as defined in formula (I) or in any variation thereof. It is also understood that any combination of R2 and R3 described for formula (I) in one aspect applies to any of formulae (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) and (IC-1 to Ic-9). It is understood that for compounds of formula (Ia), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ia-5) or (Ia-6), the variables R2 and R3 are as defined in formula (I) or in any variation thereof. It is also understood that any combination of R2 and R3 described for formula (I) in one aspect applies to any of formulae (Ia), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ia-5) and (Ia-6).
  • In some embodiments of a compound of Formula (I), A, B, R1 and R4 together are selected from the group consisting of:
  • Figure US20230109104A1-20230406-C00084
    Figure US20230109104A1-20230406-C00085
    Figure US20230109104A1-20230406-C00086
    Figure US20230109104A1-20230406-C00087
    Figure US20230109104A1-20230406-C00088
    Figure US20230109104A1-20230406-C00089
    Figure US20230109104A1-20230406-C00090
    Figure US20230109104A1-20230406-C00091
    Figure US20230109104A1-20230406-C00092
    Figure US20230109104A1-20230406-C00093
    Figure US20230109104A1-20230406-C00094
    Figure US20230109104A1-20230406-C00095
    Figure US20230109104A1-20230406-C00096
  • wherein the wavy lines denote attachment points to the parent molecule.
  • In some embodiments of a compound of Formula (I), A, B, R1 and R1 together are selected from the group consisting of:
  • Figure US20230109104A1-20230406-C00097
    Figure US20230109104A1-20230406-C00098
    Figure US20230109104A1-20230406-C00099
  • and wherein the wavy lines denote attachment points to the parent molecule.
  • In some embodiments of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9), R2 is hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C3-C6 cycloalkyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR10, —(C1-C3 alkylene)S(O)2R10, —(C1-C3 alkylene)(5- to 10-membered heteroaryl), —(C1-C3 alkylene)(C6-C14 aryl), or —(C1-C3 alkylene)(C3-C6 cycloalkyl), each of which is optionally substituted by halogen or C1-C6 alkyl. In some embodiments of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9), R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl). In some embodiments of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9), R2 is C1-C6 alkyl or —(C1-C3 alkylene)(C3-C6 cycloalkyl). In some embodiments of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9), R2 is C1-C6 alkyl or C2-C6 alkenyl. In some embodiments of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9), R2 is —(C1-C3 alkylene)(C3-C6 cycloalkyl) or C2-C6 alkenyl. In some embodiments of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9), R2 is C1-C6 alkyl. In some embodiments of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9), R2 is C2-C6 alkenyl. In some embodiments of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9), R2 is —(C1-C3 alkylene)(C3-C6 cycloalkyl). In some embodiments of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9), R2 is C3-C6 cycloalkyl.
  • In some embodiments of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ia-5) or (Ia-6), R2 is hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C3-C6 cycloalkyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR10, —(C1-C3 alkylene)S(O)2R10, —(C1-C3 alkylene)(5- to 10-membered heteroaryl), —(C1-C3 alkylene)(C6-C14 aryl), or —(C1-C3 alkylene)(C3-C6 cycloalkyl), each of which is optionally substituted by halogen or C1-C6 alkyl. In some embodiments of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ia-5) or (Ia-6), R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl). In some embodiments of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ia-5) or (Ia-6), R2 is C1-C6 alkyl or —(C1-C3 alkylene)(C3-C6 cycloalkyl). In some embodiments of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ia-5) or (Ia-6), or (If), R2 is C1-C6 alkyl or C2-C6 alkenyl. In some embodiments of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ia-5) or (Ia-6), R2 is —(C1-C3 alkylene)(C3-C6 cycloalkyl) or C2-C6 alkenyl. In some embodiments of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ia-5) or (Ia-6), R2 is C1-C6 alkyl. In some embodiments of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ia-5) or (Ia-6), R2 is C2-C6 alkenyl. In some embodiments of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ia-5) or (Ia-6), R2 is —(C1-C3 alkylene)(C3-C6 cycloalkyl). In some embodiments of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ia-5) or (Ia-6), R2 is C3-C6 cycloalkyl.
  • In some embodiments of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9), R2 is selected from the group consisting of:
  • methyl, ethyl, isopropyl, cyclopropyl, cyclobuyl,
  • Figure US20230109104A1-20230406-C00100
    Figure US20230109104A1-20230406-C00101
  • wherein the wavy lines denote attachment points to the parent molecule.
  • In some embodiments of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ia-5) or (Ia-6), R2 is selected from the group consisting of:
  • methyl, ethyl, isopropyl, cyclopropyl,
  • Figure US20230109104A1-20230406-C00102
  • wherein the wavy lines denote attachment points to the parent molecule.
  • In some embodiments of a compound of Formula (I), A is unsubstituted phenyl fused with B, wherein B is C aryl, 5- to 7-membered heteroaryl, C3-C8 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is substituted with R4; R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR7C(O)R18, —S(O)2R17, —NR7S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR7S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl) or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19; R2 is hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C3-C6 cycloalkyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR10, —(C1-C3 alkylene)S(O)2R′0, —(C1-C3 alkylene)(5- to 10-membered heteroaryl), —(C1-C3 alkylene)(C6-C14 aryl), or —(C1-C3 alkylene)(C3-C6 cycloalkyl), each of which is optionally substituted by halogen or C1-C6 alkyl. In some embodiments of a compound of Formula (I), A is unsubstituted phenyl fused with B, wherein B is C6 aryl, 5- to 7-membered heteroaryl, C3-C8 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is substituted with R4; R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, or —C(O)NR17R18, wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl; R2 is hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C3-C6 cycloalkyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR10, —(C1-C3 alkylene)S(O)2R10, —(C1-C3 alkylene)(5- to 10-membered heteroaryl), —(C1-C3 alkylene)(C6-C14 aryl), or —(C1-C3 alkylene)(C3-C6 cycloalkyl), each of which is optionally substituted by halogen or C1-C6 alkyl. In some embodiments of a compound of Formula (I), A is unsubstituted phenyl fused with B, wherein B is 3- to 8-membered heterocyclyl optionally substituted with R4; R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl). In some embodiments of a compound of Formula (I), A is unsubstituted phenyl fused with B, wherein B is 3- to 8-membered heterocyclyl optionally substituted with R4; R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl). In some embodiments of a compound of Formula (I), A is unsubstituted phenyl fused with B, wherein B is 6-membered heterocyclyl having one nitrogen atom, wherein the 6-membered heterocyclyl is optionally substituted by R4; R4 is independently C1-C6 alkyl; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl). In some embodiments of a compound of Formula (I), A is unsubstituted phenyl fused with B, wherein B is 6-membered heterocyclyl having one nitrogen atom, wherein the 6-membered heterocyclyl is optionally substituted by R4; R4 is independently C1-C6 alkyl; R2 is selected from the group consisting of: allyl, cyclopropylmethyl, isopropyl, cyclopropyl, ethyl, and propyl.
  • In some embodiments of a compound of Formula (I), A is phenyl optionally substituted by R1 and fused with B, wherein R1 is independently —CN, C1-C6 alkyl, or halogen; B is C6 aryl, 5- to 7-membered heteroaryl, C3-C8 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is substituted with R4; R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR17S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl) or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl). In some embodiments of a compound of Formula (I), A is phenyl optionally substituted by R1 and fused with B, wherein R1 is independently —CN, C1-C6 alkyl, or halogen; B is 3- to 8-membered heterocyclyl optionally substituted with R4; R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4 are, when bound to the same carbon or two different carbon atoms, are taken together with the carbon to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl). In some embodiments of a compound of Formula (I), A is phenyl optionally substituted by R1 and fused with B, wherein R1 is independently —CN, C1-C6 alkyl, or halogen; B is 3- to 8-membered heterocyclyl optionally substituted with R4; R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19; R2 is selected from the group consisting of: allyl, cyclopropylmethyl, isopropyl, cyclopropyl, ethyl, and propyl.
  • In some embodiments of a compound of Formula (I), A is phenyl optionally substituted by R1 and fused with B, wherein R1 is independently —CN, C1-C6 alkyl, or halogen; B is C6 aryl, 5- to 7-membered heteroaryl, C3-C8 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is substituted with R4; R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, or —C(O)NR17R18, wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl). In some embodiments of a compound of Formula (I), A is phenyl optionally substituted by R1 and fused with B, wherein R1 is independently —CN, C1-C6 alkyl, or halogen; B is 3- to 8-membered heterocyclyl optionally substituted with R4; R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl). In some embodiments of a compound of Formula (I), A is phenyl optionally substituted by R1 and fused with B, wherein R1 is independently —CN, C1-C6 alkyl, or halogen; B is 3- to 8-membered heterocyclyl optionally substituted with R4; R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl; R2 is selected from the group consisting of: allyl, cyclopropylmethyl, isopropyl, cyclopropyl, ethyl, and propyl.
  • In some embodiments of a compound of Formula (I), A is unsubstituted 6-membered heteroaryl fused with B, wherein B is C6 aryl, 5- to 7-membered heteroaryl, C3-C8 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is substituted with R4; R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR17S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl) or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19; R2 is hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C3-C6 cycloalkyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR10, —(C1-C3 alkylene)S(O)2R10, —(C1-C3 alkylene)(5- to 10-membered heteroaryl), —(C1-C3 alkylene)(C6-C14 aryl), or —(C1-C3 alkylene)(C3-C6 cycloalkyl), each of which is optionally substituted by halogen or C1-C6 alkyl. In some embodiments of a compound of Formula (I), A is unsubstituted 6-membered heteroaryl fused with B, wherein B is 3- to 8-membered heterocyclyl optionally substituted with R4; R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl). In some embodiments of a compound of Formula (I), A is unsubstituted 6-membered heteroaryl fused with B, wherein B is 6-membered heterocyclyl having one nitrogen atom, wherein the 6-membered heterocyclyl is optionally substituted by R4; R4 is independently C1-C6 alkyl; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl). In some embodiments of a compound of Formula (I), A is unsubstituted 6-membered heteroaryl fused with B, wherein B is 6-membered heterocyclyl having one nitrogen atom, wherein the 6-membered heterocyclyl is optionally substituted by R4; R4 is independently C1-C6 alkyl; R2 is selected from the group consisting of: allyl, cyclopropylmethyl, isopropyl, cyclopropyl, ethyl, and propyl.
  • In some embodiments of a compound of Formula (I), A is unsubstituted 6-membered heteroaryl fused with B, wherein B is C6 aryl, 5- to 7-membered heteroaryl, C3-C8 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is substituted with R4; R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, or —C(O)NR17R18, wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl; R2 is hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C3-C6 cycloalkyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR10, —(C1-C3 alkylene)S(O)2R10, —(C1-C3 alkylene)(5- to 10-membered heteroaryl), —(C1-C3 alkylene)(C6-C14 aryl), or —(C1-C3 alkylene)(C3-C6 cycloalkyl), each of which is optionally substituted by halogen or C1-C6 alkyl. In some embodiments of a compound of Formula (I), A is unsubstituted 6-membered heteroaryl fused with B, wherein B is 3- to 8-membered heterocyclyl optionally substituted with R4; R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl). In some embodiments of a compound of Formula (I), A is unsubstituted 6-membered heteroaryl fused with B, wherein B is 6-membered heterocyclyl having one nitrogen atom, wherein the 6-membered heterocyclyl is optionally substituted by R4; R4 is independently C1-C6 alkyl; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl). In some embodiments of a compound of Formula (I), A is unsubstituted 6-membered heteroaryl fused with B, wherein B is 6-membered heterocyclyl having one nitrogen atom, wherein the 6-membered heterocyclyl is optionally substituted by R4; R4 is independently C1-C6 alkyl; R2 is selected from the group consisting of: allyl, cyclopropylmethyl, isopropyl, cyclopropyl, ethyl, and propyl.
  • In some embodiments of a compound of Formula (I), A, B, R1 and R4 together are selected from the group consisting of:
  • Figure US20230109104A1-20230406-C00103
    Figure US20230109104A1-20230406-C00104
    Figure US20230109104A1-20230406-C00105
    Figure US20230109104A1-20230406-C00106
    Figure US20230109104A1-20230406-C00107
    Figure US20230109104A1-20230406-C00108
    Figure US20230109104A1-20230406-C00109
    Figure US20230109104A1-20230406-C00110
    Figure US20230109104A1-20230406-C00111
    Figure US20230109104A1-20230406-C00112
    Figure US20230109104A1-20230406-C00113
    Figure US20230109104A1-20230406-C00114
    Figure US20230109104A1-20230406-C00115
  • wherein the wavy lines denote attachment points to the parent molecule; R2 is selected from the group consisting of:
    methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl,
  • Figure US20230109104A1-20230406-C00116
  • wherein the wavy lines denote attachment points to the parent molecule.
  • In some embodiments of a compound of Formula (I), A, B, R1 and R4 together are selected from the group consisting of:
  • Figure US20230109104A1-20230406-C00117
    Figure US20230109104A1-20230406-C00118
    Figure US20230109104A1-20230406-C00119
  • wherein the wavy lines denote attachment points to the parent molecule; R2 is selected from the group consisting of:
    methyl, ethyl, isopropyl, cyclopropyl,
  • Figure US20230109104A1-20230406-C00120
  • In some embodiments of a compound of Formula (I), (Ia), (Ia-1 to Ia-6), (Ib), (Ib-1 to Ib-3), (Ic) or (Ic-1 to Ic-3), R3 is hydrogen, C1-C6 alkyl, —C(O)R10, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3- to 12-membered heterocyclyl), —(C1-C3 alkylene)(5- to 10-membered heteroaryl), —S(O)R10, —S(O)2R10, or 5- to 10-membered heteroaryl, each of which is optionally substituted by halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR5 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that the 5- to 10-membered heteroaryl of R3 is not substituted by
  • Figure US20230109104A1-20230406-C00121
  • In some embodiments of a compound of Formula (I), (Ia), (Ia-1 to Ia-6), (Ib), (Ib-1 to Ib-3), (Ic) or (Ic-1 to Ic-3), R3 is —C(O)R10 or 5- to 10-membered heteroaryl which is unsubstituted. In some embodiments of a compound of Formula (I), (Ia), (Ia-1 to Ia-6), (Ib), (Ib-1 to Ib-3), (Ic) or (Ic-1 to Ic-3), R3 is —C(O)R10 or 5- to 10-membered heteroaryl optionally substituted by halogen, oxo, —OR13, —NR13R14, —C(O)R3, —C(O)NR13R14, —NR13C(O)R14, —CN, —S(O)R13, —SR13, —S(O)2R13, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, or halogen, with the proviso that the 5- to 10-membered heteroaryl of R3 is not substituted by
  • Figure US20230109104A1-20230406-C00122
  • In some embodiments of a compound of Formula (I), (Ia), (Ia-1 to Ia-6), (Ib), (Ib-1 to Ib-3), (Ic) or (Ic-1 to Ic-3), R3 is —C(O)R10 or 5- to 10-membered heteroaryl substituted by 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, —CN, —(C1-C3 alkylene)NR13R14, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, —C(O)R13, —NR13R14, —OR13, or C1-C6 alkyl optionally substituted by —OH or halogen, with the proviso that the 5- to 10-membered heteroaryl of R3 is not substituted by
  • Figure US20230109104A1-20230406-C00123
  • In some embodiments of a compound of Formula (I), (Ia), (Ia-1 to Ia-6), (Ib), (Ib-1 to Ib-3), (Ic) or (Ic-1 to Ic-3), R3 is 5- to 10-membered heteroaryl substituted by 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, —CN, —(C1-C3 alkylene)NR13R14, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, —C(O)R13, —NR13R14, —OR13, or C1-C6 alkyl optionally substituted by —OH or halogen, with the proviso that the 5- to 10-membered heteroaryl of R3 is not substituted by
  • Figure US20230109104A1-20230406-C00124
  • In some embodiments of a compound of Formula (I), (Ia), (Ia-1 to Ia-6), (Ib), (Ib-1 to Ib-3), (Ic) or (Ic-1 to Ic-3), R3 is 5- to 10-membered heteroaryl substituted by —OR13 or C1-C6 alkyl, wherein the C1-C6 alkyl is optionally substituted by —OH or halogen. In some embodiments of a compound of Formula (I), (Ia), (Ia-1 to Ia-6), (Ib), (Ib-1 to Ib-3), (Ic) or (Ic-1 to Ic-3), R3 is 5- to 10-membered heteroaryl substituted by —OR13 or C1-C6 alkyl, wherein the C1-C6 alkyl is unsubstituted.
  • In some embodiments of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ia-5) or (Ia-6), R3 is hydrogen, C1-C6 alkyl, —C(O)R10, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3- to 12-membered heterocyclyl), —(C1-C3 alkylene)(5- to 10-membered heteroaryl), —S(O)R10, —S(O)2R10, or 5- to 10-membered heteroaryl, each of which is optionally substituted by halogen, oxo, —OR13, —NR13R14, —C(O)R13, —CN, —S(O)R13, —S(O)2R13, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, or halogen. In some embodiments of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ia-5) or (Ia-6), or (If), R3 is —C(O)R10 or 5- to 10-membered heteroaryl which is unsubstituted. In some embodiments of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ia-5) or (Ia-6), R3 is —C(O)R10 or 5- to 10-membered heteroaryl optionally substituted by halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)NR13R14, —NR13C(O)R14, —CN, —S(O)R13, —S(O)2R13, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, or halogen. In some embodiments of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ia-5) or (Ia-6), R3 is —C(O)R10 or 5- to 10-membered heteroaryl substituted by 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, —CN, —(C1-C3 alkylene)NR13R14, C3-C8 cycloalkyl, —C(O)R13, —NR13R14, —OR13, or C1-C6 alkyl optionally substituted by —OH or halogen. In some embodiments of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ia-5) or (Ia-6), R3 is 5- to 10-membered heteroaryl substituted by 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, —CN, —(C1-C3 alkylene)NR13R14, C3-C8 cycloalkyl, —C(O)R13, —NR13R14, —OR13, or C1-C6 alkyl optionally substituted by —OH or halogen. In some embodiments of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ia-5) or (Ia-6), R3 is 5- to 10-membered heteroaryl substituted by —OR3 or C1-C6 alkyl, wherein the C1-C6 alkyl is optionally substituted by —OH.
  • In some embodiments of a compound of Formula (I), (Ia), (Ia-1 to Ia-6), (Ib), (Ib-1 to Ib-3), (Ic) or (Ic-1 to Ic-3), R3 is —C(O)R10 or 6-membered heteroaryl optionally substituted by halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR5 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that the 6-membered heteroaryl of R3 is not substituted by
  • Figure US20230109104A1-20230406-C00125
  • In some embodiments of a compound of Formula (I), (Ia), (Ia-1 to Ia-6), (Ib), (Ib-1 to Ib-3), (Ic) or (Ic-1 to Ic-3), R3 is 6-membered heteroaryl optionally substituted by 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, —CN, —(C1-C3 alkylene)NR13R14, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, —C(O)R13, —NR13R14, —OR13, or C1-C6 alkyl optionally substituted by —OH, —NR15R16, —OR15 or halogen, with the proviso that the 6-membered heteroaryl of R3 is not substituted by
  • Figure US20230109104A1-20230406-C00126
  • In some embodiments of a compound of Formula (I), (Ia), (Ia-1 to Ia-6), (Ib), (Ib-1 to Ib-3), (Ic) or (Ic-1 to Ic-3), R3 is 6-membered heteroaryl optionally substituted by —OR13 or C1-C6 alkyl, wherein the C1-C6 alkyl is optionally substituted by —OH or halogen.
  • In some embodiments of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ia-5) or (Ia-6), R3 is —C(O)R10 or 6-membered heteroaryl optionally substituted by halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)NR13R14, —NR13C(O)R14, —CN, —S(O)R13, —S(O)2R13, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, or halogen. In some embodiments of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ia-5) or (Ia-6), R3 is 6-membered heteroaryl optionally substituted by 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, —CN, —(C1-C3 alkylene)NR13R14, C3-C8 cycloalkyl, —C(O)R13, —NR13R14, —OR13, or C1-C6 alkyl optionally substituted by —OH or halogen. In some embodiments of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ia-5) or (Ia-6), R3 is 6-membered heteroaryl optionally substituted by —OR13 or C1-C6 alkyl, wherein the C1-C6 alkyl is optionally substituted by —OH.
  • In some embodiments of a compound of Formula (I), (Ia), (Ia-1 to Ia-6), (Ib), (Ib-1 to Ib-3), (Ic) or (Ic-1 to Ic-3), R3 is pyridinyl optionally substituted by halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)NR13R14, —NR13C(O)R14, —CN, —S(O)R13, —SR13, —S(O)2R13, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —NR15R16, —C(O)OR15, —OR15, —OH, or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen. In some embodiments of a compound of Formula (I), (Ia), (Ia-1 to Ia-6), (Ib), (Ib-1 to Ib-3), (Ic) or (Ic-1 to Ic-3), R3 is pyridinyl optionally substituted by 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, —CN, —(C1-C3 alkylene)NR13R14, C3-C8 cycloalkyl optionally substituted by —OH or halogen, —C(O)R13, —NR13R14, —OR13, or C1-C6 alkyl optionally substituted by —OH or halogen, with the proviso that the pyridinyl of R3 is not substituted by
  • Figure US20230109104A1-20230406-C00127
  • In some embodiments of a compound of Formula (I), (Ia), (Ia-1 to Ia-6), (Ib), (Ib-1 to Ib-3), (Ic) or (Ic-1 to Ic-3), R3 is pyridinyl optionally substituted by —OR13 or C1-C6 alkyl, wherein the C1-C6 alkyl is optionally substituted by —OH. In some embodiments of a compound of Formula (I), (Ia), (Ia-1 to Ia-6), (Ib), (Ib-1 to Ib-3), (Ic) or (Ic-1 to Ic-3), R3 is pyridinyl substituted by C1-C6 alkyl, wherein the C1-C6 alkyl is optionally substituted by —OH. In some embodiments of a compound of Formula (I), (Ia), (Ia-1 to Ia-6), (Ib), (Ib-1 to Ib-3), (Ic) or (Ic-1 to Ic-3), R3 is pyridinyl substituted by C1-C6 alkyl, wherein the C1-C6 alkyl is optionally substituted by halogen. In some embodiments of a compound of Formula (I), (Ia), (Ia-1 to Ia-6), (Ib), (Ib-1 to Ib-3), (Ic) or (Ic-1 to Ic-3), R3 is pyridinyl substituted by C1-C6 alkyl, wherein the C1-C6 alkyl is tertiary butyl.
  • In some embodiments of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ia-5) or (Ia-6), R3 is pyridinyl optionally substituted by halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)NR13R14, —NR13C(O)R14, —CN, —S(O)R13, —S(O)2R13, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, or halogen. In some embodiments of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ia-5) or (Ia-6), R3 is pyridinyl optionally substituted by 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, —CN, —(C1-C3 alkylene)NR13R14, C3-C8 cycloalkyl, —C(O)R13, —NR13R14, —OR13, or C1-C6 alkyl optionally substituted by —OH or halogen. In some embodiments of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ia-5) or (Ia-6), R3 is pyridinyl optionally substituted by —OR13 or C1-C6 alkyl, wherein the C1-C6 alkyl is optionally substituted by —OH.
  • In some embodiments of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9), R2 is selected from the group consisting of:
  • methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl,
  • Figure US20230109104A1-20230406-C00128
    Figure US20230109104A1-20230406-C00129
  • wherein the wavy lines denote attachment points to the parent molecule.
  • In some embodiments of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ia-5) or (Ia-6), R2 is selected from the group consisting of:
  • methyl, ethyl, isopropyl, cyclopropyl,
  • Figure US20230109104A1-20230406-C00130
  • wherein the wavy lines denote attachment points to the parent molecule.
  • In some embodiments of a compound of Formula (I), (Ia), (Ia-1 to Ia-6), (Ib), (Ib-1 to Ib-3), (Ic) or (Ic-1 to Ic-3), R3 is selected from the group consisting of:
  • Figure US20230109104A1-20230406-C00131
    Figure US20230109104A1-20230406-C00132
    Figure US20230109104A1-20230406-C00133
    Figure US20230109104A1-20230406-C00134
    Figure US20230109104A1-20230406-C00135
    Figure US20230109104A1-20230406-C00136
    Figure US20230109104A1-20230406-C00137
    Figure US20230109104A1-20230406-C00138
    Figure US20230109104A1-20230406-C00139
    Figure US20230109104A1-20230406-C00140
    Figure US20230109104A1-20230406-C00141
    Figure US20230109104A1-20230406-C00142
    Figure US20230109104A1-20230406-C00143
    Figure US20230109104A1-20230406-C00144
  • wherein the wavy lines denote attachment points to the parent molecule.
  • In some embodiments of a compound of Formula (I), (Ia), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ia-5) or (Ia-6), R3 is selected from the group consisting of:
  • Figure US20230109104A1-20230406-C00145
    Figure US20230109104A1-20230406-C00146
  • wherein the wavy lines denote attachment points to the parent molecule.
  • In some embodiments of a compound of Formula (I), A is unsubstituted phenyl fused with B, wherein B is C6 aryl, 5- to 7-membered heteroaryl, C3-C8 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is substituted with R4; R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR7C(O)R18, —S(O)2R17, —NR7S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR17S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl) or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl); R3 is hydrogen, C1-C6 alkyl, —C(O)R10, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3- to 12-membered heterocyclyl), —(C1-C3 alkylene)(5- to 10-membered heteroaryl), —S(O)R10, —S(O)2R10, or 5- to 10-membered heteroaryl, each of which is optionally substituted by halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15, or halogen, with the proviso that the 5- to 10-membered heteroaryl of R3 is not substituted by
  • Figure US20230109104A1-20230406-C00147
  • In some embodiments of a compound of Formula (I), A is unsubstituted phenyl fused with B, wherein B is C6 aryl, 3- to 7-membered heteroaryl, C3-C8 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is substituted with R4; R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, or —C(O)NR17R18, wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl); R3 is —C(O)R10 or 5- to 10-membered heteroaryl optionally substituted by halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)NR13R14, —NR13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by oxo, —OH or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, or halogen, with the proviso that the 5- to 10-membered heteroaryl of R3 is not substituted by
  • Figure US20230109104A1-20230406-C00148
  • In some embodiments of a compound of Formula (I), A is unsubstituted phenyl fused with B, wherein B is 3- to 8-membered heterocyclyl optionally substituted with R4; R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl); R3 is 5- to 10-membered heteroaryl substituted by 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, —CN, —(C1-C3 alkylene)NR13R14, C3-C8 cycloalkyl optionally substituted by oxo, —OH or halogen, —C(O)R13, —NR13R14, —OR13, or C1-C6 alkyl optionally substituted by —OH or halogen, with the proviso that the 5- to 10-membered heteroaryl of R3 is not substituted by
  • Figure US20230109104A1-20230406-C00149
  • In some embodiments of a compound of Formula (I), A is unsubstituted phenyl fused with B, wherein B is 6-membered heterocyclyl having one nitrogen atom, wherein the 6-membered heterocyclyl is optionally substituted by R4; R4 is independently C1-C6 alkyl; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl); R3 is 5- to 10-membered heteroaryl substituted by 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, —CN, —(C1-C3 alkylene)NR13R14, C3-C8 cycloalkyl optionally substituted by oxo, —OH or halogen, —C(O)R13, —NR13R14, —OR13, or C1-C6 alkyl optionally substituted by —OH or halogen, with the proviso that the 5- to 10-membered heteroaryl of R3 is not substituted by
  • Figure US20230109104A1-20230406-C00150
  • In some embodiments of a compound of Formula (I), A is unsubstituted phenyl fused with B, wherein B is 6-membered heterocyclyl having one nitrogen atom, wherein the 6-membered heterocyclyl is optionally substituted by R4; R4 is independently C1-C6 alkyl; R2 is selected from the group consisting of: allyl, cyclopropylmethyl, isopropyl, cyclopropyl, ethyl, and propyl; R3 is 5- to 10-membered heteroaryl substituted by —OR13 or C1-C6 alkyl, wherein the C1-C6 alkyl is optionally substituted by —OH or halogen.
  • In some embodiments of a compound of Formula (I), A is unsubstituted phenyl fused with B, wherein B is C6-C14 aryl, 5- to 10-membered heteroaryl, C3-C8 cycloalkyl, or 3- to 12-membered heterocyclyl, each of which is substituted with R4; R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, or —C(O)NR17R18, wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl); R3 is hydrogen, C1-C6 alkyl, —C(O)R10, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3- to 12-membered heterocyclyl), —(C1-C3 alkylene)(5- to 10-membered heteroaryl), —S(O)R10, —S(O)2R10, or 5- to 10-membered heteroaryl, each of which is optionally substituted by halogen, oxo, —OR13, —NR13R14, —C(O)R13, —CN, —S(O)R13, —S(O)2R13, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, or halogen. In some embodiments of a compound of Formula (I), A is unsubstituted phenyl fused with B, wherein B is C6-C14 aryl, 5- to 10-membered heteroaryl, C3-C8 cycloalkyl, or 3- to 12-membered heterocyclyl, each of which is substituted with R4; R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, or —C(O)NR17R18, wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl); R3 is —C(O)R10 or 5- to 10-membered heteroaryl optionally substituted by halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)NR13R14, —NR13C(O)R14, —CN, —S(O)R13, —S(O)2R13, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, or halogen. In some embodiments of a compound of Formula (I), A is unsubstituted phenyl fused with B, wherein B is 3- to 12-membered heterocyclyl optionally substituted with R4; R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl); R3 is 5- to 10-membered heteroaryl substituted by 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, —CN, —(C1-C3 alkylene)NR13R14, C3-C8 cycloalkyl, —C(O)R13, —NR13R14, —OR13, or C1-C6 alkyl optionally substituted by —OH or halogen. In some embodiments of a compound of Formula (I), A is unsubstituted phenyl fused with B, wherein B is 6-membered heterocyclyl having one nitrogen atom, wherein the 6-membered heterocyclyl is optionally substituted by R4; R4 is independently C1-C6 alkyl; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl); R3 is 5- to 10-membered heteroaryl substituted by 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, —CN, —(C1-C3 alkylene)NR13R14, C3-C8 cycloalkyl, —C(O)R13, —NR13R14, —OR13, or C1-C6 alkyl optionally substituted by —OH or halogen. In some embodiments of a compound of Formula (I), A is unsubstituted phenyl fused with B, wherein B is 6-membered heterocyclyl having one nitrogen atom, wherein the 6-membered heterocyclyl is optionally substituted by R4; R4 is independently C1-C6 alkyl; R2 is selected from the group consisting of: allyl, cyclopropylmethyl, isopropyl, cyclopropyl, ethyl, and propyl; R3 is 5- to 10-membered heteroaryl substituted by —OR13 or C1-C6 alkyl, wherein the C1-C6 alkyl is optionally substituted by —OH.
  • In some embodiments of a compound of Formula (I), A is phenyl optionally substituted by R1 and fused with B, wherein R1 is independently —CN, C1-C6 alkyl, or halogen; B is C6 aryl, 5- to 7-membered heteroaryl, C3-C8 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is substituted with R4; R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR7S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR7S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl) or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl); R3 is hydrogen, C1-C6 alkyl, —C(O)R10, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3- to 12-membered heterocyclyl), —(C1-C3 alkylene)(5- to 10-membered heteroaryl), —S(O)R10, —S(O)2R10, or 5- to 10-membered heteroaryl, each of which is optionally substituted by halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15, or halogen, with the proviso that the 5- to 10-membered heteroaryl of R3 is not substituted by
  • Figure US20230109104A1-20230406-C00151
  • In some embodiments of a compound of Formula (I), A is phenyl optionally substituted by R1 and fused with B, wherein R1 is independently —CN, C1-C6 alkyl, or halogen; B is C6 aryl, 5- to 7-membered heteroaryl, C3-C8 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is substituted with R4; R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, or —C(O)NR17R18, wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl); R3 is —C(O)R10 or 5- to 10-membered heteroaryl optionally substituted by halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)NR13R14, —NR13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by oxo, —OH or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, or halogen, with the proviso that the 5- to 10-membered heteroaryl of R3 is not substituted by
  • Figure US20230109104A1-20230406-C00152
  • In some embodiments of a compound of Formula (I), A is phenyl optionally substituted by R1 and fused with B, wherein R1 is independently —CN, C1-C6 alkyl, or halogen; B is 3- to 8-membered heterocyclyl optionally substituted with R4; R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl); R3 is 5- to 10-membered heteroaryl substituted by 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, —CN, —(C1-C3 alkylene)NR13R14, C3-C8 cycloalkyl optionally substituted by oxo, —OH or halogen, —C(O)R13, —NR13R14, —OR13, or C1-C6 alkyl optionally substituted by —OH or halogen, with the proviso that the 5- to 10-membered heteroaryl of R3 is not substituted by
  • Figure US20230109104A1-20230406-C00153
  • In some embodiments of a compound of Formula (I), A is phenyl optionally substituted by R1 and fused with B, wherein R1 is independently —CN, C1-C6 alkyl, or halogen; B is 3- to 8-membered heterocyclyl optionally substituted with R4; R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19; R2 is selected from the group consisting of: allyl, cyclopropylmethyl, isopropyl, cyclopropyl, ethyl, and propyl; R3 is 5- to 10-membered heteroaryl substituted by —OR13 or C1-C6 alkyl, wherein the C1-C6 alkyl is optionally substituted by —OH or halogen.
  • In some embodiments of a compound of Formula (I), A is phenyl optionally substituted by R1 and fused with B, wherein R1 is independently —CN, C1-C6 alkyl, or halogen; B is C6-C14 aryl, 5- to 10-membered heteroaryl, C3-C8 cycloalkyl, or 3- to 12-membered heterocyclyl, each of which is substituted with R4; R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, or —C(O)NR17R18, wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl); R3 is hydrogen, C1-C6 alkyl, —C(O)R10, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3- to 12-membered heterocyclyl), —(C1-C3 alkylene)(5- to 10-membered heteroaryl), —S(O)R10, —S(O)2R10, or 5- to 10-membered heteroaryl, each of which is optionally substituted by halogen, oxo, —OR13, —NR13R14, —C(O)R13, —CN, —S(O)R13, —S(O)2R13, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, or halogen. In some embodiments of a compound of Formula (I), A is phenyl optionally substituted by R1 and fused with B, wherein R1 is independently —CN, C1-C6 alkyl, or halogen; B is C6-C14 aryl, 5- to 10-membered heteroaryl, C3-C8 cycloalkyl, or 3- to 12-membered heterocyclyl, each of which is substituted with R4; R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, or —C(O)NR17R18, wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl); R3 is —C(O)R10 or 5- to 10-membered heteroaryl optionally substituted by halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)NR13R14, —NR13C(O)R14, —CN, —S(O)R13, —S(O)2R13, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C5 cycloalkyl, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, or halogen. In some embodiments of a compound of Formula (I), A is phenyl optionally substituted by R1 and fused with B, wherein R1 is independently —CN, C1-C6 alkyl, or halogen; B is 3- to 12-membered heterocyclyl optionally substituted with R4; R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl); R3 is 5- to 10-membered heteroaryl substituted by 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, —CN, —(C1-C3 alkylene)NR13R14, C3-C8 cycloalkyl, —C(O)R13, —NR13R14, —OR13, or C1-C6 alkyl optionally substituted by —OH or halogen. In some embodiments of a compound of Formula (I), A is phenyl optionally substituted by R1 and fused with B, wherein R1 is independently —CN, C1-C6 alkyl, or halogen; B is 3- to 12-membered heterocyclyl optionally substituted with R4; R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl; R2 is selected from the group consisting of: allyl, cyclopropylmethyl, isopropyl, cyclopropyl, ethyl, and propyl; R3 is 5- to 10-membered heteroaryl substituted by —OR13 or C1-C6 alkyl, wherein the C1-C6 alkyl is optionally substituted by —OH.
  • In some embodiments of a compound of Formula (I), A is unsubstituted 6-membered heteroaryl fused with B, wherein B is C6 aryl, 5- to 7-membered heteroaryl, C3-C8 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is substituted with R4; R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR17S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl) or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O) R19; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl); R3 is hydrogen, C1-C6 alkyl, —C(O)R10, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3- to 12-membered heterocyclyl), —(C1-C3 alkylene)(5- to 10-membered heteroaryl), —S(O)R10, —S(O)2R10, or 5- to 10-membered heteroaryl, each of which is optionally substituted by halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15, or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that the 5- to 10-membered heteroaryl of R3 is not substituted by
  • Figure US20230109104A1-20230406-C00154
  • In some embodiments of a compound of Formula (I), A is unsubstituted 6-membered heteroaryl fused with B, wherein B is C6 aryl, 5- to 7-membered heteroaryl, C3-C8 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is substituted with R4; R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, or —C(O)NR17R18, wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl); R3 is —C(O)R10 or 5- to 10-membered heteroaryl optionally substituted by halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)NR13R14, —NR13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by oxo, —OH or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, or halogen, with the proviso that the 5- to 10-membered heteroaryl of R3 is not substituted by
  • Figure US20230109104A1-20230406-C00155
  • In some embodiments of a compound of Formula (I), A is unsubstituted 6-membered heteroaryl fused with B, wherein B is 3- to 12-membered heterocyclyl optionally substituted with R4; R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl); R3 is 5- to 10-membered heteroaryl substituted by 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, —CN, —(C1-C3 alkylene)NR13R14, C3-C8 cycloalkyl optionally substituted by oxo, —OH or halogen, —C(O)R13, —NR13R14, —OR13, or C1-C6 alkyl optionally substituted by —OH or halogen, with the proviso that the 5- to 10-membered heteroaryl of R3 is not substituted by
  • Figure US20230109104A1-20230406-C00156
  • In some embodiments of a compound of Formula (I), A is unsubstituted 6-membered heteroaryl fused with B, wherein B is 6-membered heterocyclyl having one nitrogen atom, wherein the 6-membered heterocyclyl is optionally substituted by R4; R4 is independently C1-C6 alkyl; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl); R3 is 5- to 10-membered heteroaryl substituted by 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, —CN, —(C1-C3 alkylene)NR13R14, C3-C8 cycloalkyl optionally substituted by oxo, —OH or halogen, —C(O)R13, —NR13R14, —OR13, or C1-C6 alkyl optionally substituted by —OH or halogen, with the proviso that the 5- to 10-membered heteroaryl of R3 is not substituted by
  • Figure US20230109104A1-20230406-C00157
  • In some embodiments of a compound of Formula (I), A is unsubstituted 6-membered heteroaryl fused with B, wherein B is 6-membered heterocyclyl having one nitrogen atom, wherein the 6-membered heterocyclyl is optionally substituted by R4; R4 is independently C1-C6 alkyl; R2 is selected from the group consisting of: allyl, cyclopropylmethyl, isopropyl, cyclopropyl, ethyl, and propyl; R3 is 5- to 10-membered heteroaryl substituted by —OR13 or C1-C6 alkyl, wherein the C1-C6 alkyl is optionally substituted by —OH or halogen.
  • In some embodiments of a compound of Formula (I), A is unsubstituted 6-membered heteroaryl fused with B, wherein B is C6-C14 aryl, 5- to 10-membered heteroaryl, C3-C5 cycloalkyl, or 3- to 12-membered heterocyclyl, each of which is substituted with R4; R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, or —C(O)NR17R18, wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl); R3 is hydrogen, C1-C6 alkyl, —C(O)R10, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3- to 12-membered heterocyclyl), —(C1-C3 alkylene)(5- to 10-membered heteroaryl), —S(O)R10, —S(O)2R10, or 5- to 10-membered heteroaryl, each of which is optionally substituted by halogen, oxo, —OR13, —NR13R14, —C(O)R13, —CN, —S(O)R13, —S(O)2R13, —(C1-C3 alkylene)OR19, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C5 cycloalkyl, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, or halogen. In some embodiments of a compound of Formula (I), A is unsubstituted 6-membered heteroaryl fused with B, wherein B is C6-C14 aryl, 5- to 10-membered heteroaryl, C3-C5 cycloalkyl, or 3- to 12-membered heterocyclyl, each of which is substituted with R4; R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, or —C(O)NR17R18, wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl); R3 is —C(O)R10 or 5- to 10-membered heteroaryl optionally substituted by halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)NR13R14, —NR13C(O)R14, —CN, —S(O)R13, —S(O)2R13, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C5 cycloalkyl, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, or halogen. In some embodiments of a compound of Formula (I), A is unsubstituted 6-membered heteroaryl fused with B, wherein B is 3- to 12-membered heterocyclyl optionally substituted with R4; R4 is independently oxo, C1-C6 alkyl, or —C(O)R17, wherein the C1-C6 alkyl or —C(O)R17 is optionally substituted by —OR19 or —NR19R20; or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl); R3 is 5- to 10-membered heteroaryl substituted by 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, —CN, —(C1-C3 alkylene)NR13R14, C3-C8 cycloalkyl, —C(O)R13, —NR13R14, —OR13, or C1-C6 alkyl optionally substituted by —OH or halogen. In some embodiments of a compound of Formula (I), A is unsubstituted 6-membered heteroaryl fused with B, wherein B is 6-membered heterocyclyl having one nitrogen atom, wherein the 6-membered heterocyclyl is optionally substituted by R4; R4 is independently C1-C6 alkyl; R2 is C1-C6 alkyl, C3-C6 cycloalkyl, C2-C6 alkenyl, or —(C1-C3 alkylene)(C3-C6 cycloalkyl); R3 is 5- to 10-membered heteroaryl substituted by 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, —CN, —(C1-C3 alkylene)NR13R14, C3-C8 cycloalkyl, —C(O)R13, —NR13R14, —OR13, or C1-C6 alkyl optionally substituted by —OH or halogen. In some embodiments of a compound of Formula (I), A is unsubstituted 6-membered heteroaryl fused with B, wherein B is 6-membered heterocyclyl having one nitrogen atom, wherein the 6-membered heterocyclyl is optionally substituted by R4; R4 is independently C1-C6 alkyl; R2 is selected from the group consisting of: allyl, cyclopropylmethyl, isopropyl, cyclopropyl, ethyl, and propyl; R3 is 5- to 10-membered heteroaryl substituted by —OR13 or C1-C6 alkyl, wherein the C1-C6 alkyl is optionally substituted by —OH.
  • It is understood that each description of A and B may be combined with each description of R2 and/or R3 the same as if each and every combination were specifically and individually listed. For example, in one aspect, it is understood that each description of A and B
  • may be combined in one aspect with a variation in which R2 is allyl and R3 is
  • Figure US20230109104A1-20230406-C00158
  • In some embodiments, it is understood that each description of A and B may be combined in one aspect with a variation in which R2 is allyl and R3 is
  • Figure US20230109104A1-20230406-C00159
  • In some embodiments, provided is a compound of Formula (I), wherein A, B, R1 and R4 together are selected from the group consisting of:
  • Figure US20230109104A1-20230406-C00160
    Figure US20230109104A1-20230406-C00161
    Figure US20230109104A1-20230406-C00162
    Figure US20230109104A1-20230406-C00163
    Figure US20230109104A1-20230406-C00164
    Figure US20230109104A1-20230406-C00165
    Figure US20230109104A1-20230406-C00166
    Figure US20230109104A1-20230406-C00167
    Figure US20230109104A1-20230406-C00168
    Figure US20230109104A1-20230406-C00169
    Figure US20230109104A1-20230406-C00170
    Figure US20230109104A1-20230406-C00171
    Figure US20230109104A1-20230406-C00172
    Figure US20230109104A1-20230406-C00173
    Figure US20230109104A1-20230406-C00174
    Figure US20230109104A1-20230406-C00175
    Figure US20230109104A1-20230406-C00176
    Figure US20230109104A1-20230406-C00177
  • wherein the wavy lines denote attachment points to the parent molecule;
    R2 is selected from the group consisting of: allyl, cyclopropylmethyl, isopropyl, cyclopropyl, ethyl, and propyl; and R3 is selected from the group consisting of:
  • Figure US20230109104A1-20230406-C00178
    Figure US20230109104A1-20230406-C00179
    Figure US20230109104A1-20230406-C00180
    Figure US20230109104A1-20230406-C00181
    Figure US20230109104A1-20230406-C00182
    Figure US20230109104A1-20230406-C00183
    Figure US20230109104A1-20230406-C00184
    Figure US20230109104A1-20230406-C00185
    Figure US20230109104A1-20230406-C00186
    Figure US20230109104A1-20230406-C00187
    Figure US20230109104A1-20230406-C00188
    Figure US20230109104A1-20230406-C00189
    Figure US20230109104A1-20230406-C00190
    Figure US20230109104A1-20230406-C00191
  • wherein the wavy lines denote attachment points to the parent molecule.
  • In some embodiments, provided is a compound of Formula (I), wherein A, B, R1 and R4 together are selected from the group consisting of:
  • Figure US20230109104A1-20230406-C00192
    Figure US20230109104A1-20230406-C00193
    Figure US20230109104A1-20230406-C00194
  • wherein the wavy lines denote attachment points to the parent molecule;
    R2 is selected from the group consisting of: allyl, cyclopropylmethyl, isopropyl, cyclopropyl, ethyl, and propyl; and
    R3 is selected from the group consisting of:
  • Figure US20230109104A1-20230406-C00195
    Figure US20230109104A1-20230406-C00196
  • and wherein the wavy lines denote attachment points to the parent molecule.
  • In some embodiments, provided is a compound of Formula (Ia-2), wherein Y is hydrogen; R4 is independently C1-C6 alkyl and n is 2; R2 is C1-C6 alkyl; and R3 is 6-membered heteroaryl optionally substituted by C1-C6 alkyl, wherein the C1-C6 alkyl is optionally substituted by —OH or halogen. In some embodiments, provided is a compound of Formula (Ia-2), wherein Y is hydrogen; R4 is methyl and n is 2; R2 is C1-C6 alkyl; and R3 is 6-membered heteroaryl optionally substituted by C1-C6 alkyl, wherein the C1-C6 alkyl is optionally substituted by —OH or halogen. In some embodiments, provided is a compound of Formula (Ia-2), wherein Y is hydrogen; R4 is methyl and n is 2; R2 is ethyl or isopropyl; and R3 is
  • Figure US20230109104A1-20230406-C00197
  • In some embodiments, provided is a compound of Formula (Ia-3), wherein Y is hydrogen and n is 0; R2 is C1-C6 alkyl; and R3 is 6-membered heteroaryl optionally substituted by C1-C6 alkyl, wherein the C1-C6 alkyl is optionally substituted by —OH or halogen. In some embodiments, provided is a compound of Formula (Ia-3), Y is hydrogen and n is 0; R2 is ethyl or isopropyl; and R3 is
  • Figure US20230109104A1-20230406-C00198
  • In some embodiments, provided is a compound of Formula (Ia-2), wherein Y is hydrogen; R4 is independently C1-C6 alkyl and n is 2; R2 is C1-C6 alkyl; and R3 is 6-membered heteroaryl optionally substituted by C1-C6 alkyl, wherein the C1-C6 alkyl is optionally substituted by —OH. In some embodiments, provided is a compound of Formula (Ia-2), wherein Y is hydrogen; R4 is methyl and n is 2; R2 is C1-C6 alkyl; and R3 is 6-membered heteroaryl optionally substituted by C1-C6 alkyl, wherein the C1-C6 alkyl is optionally substituted by —OH. In some embodiments, provided is a compound of Formula (Ia-2), wherein Y is hydrogen; R4 is methyl and n is 2; R2 is ethyl; and R3 is
  • Figure US20230109104A1-20230406-C00199
  • In some embodiments, provided is a compound of Formula (Ia-3), wherein Y is hydrogen and n is 0; R2 is C1-C6 alkyl; and R3 is 6-membered heteroaryl optionally substituted by C1-C6 alkyl, wherein the C1-C6 alkyl is optionally substituted by —OH. In some embodiments, provided is a compound of Formula (Ia-3), Y is hydrogen and n is 0; R2 is ethyl; and R3 is
  • Figure US20230109104A1-20230406-C00200
  • In some embodiments, provided is a compound of Formula (I), wherein: A, B, R1 and R4 together are
  • Figure US20230109104A1-20230406-C00201
  • wherein the wavy lines denote attachment points to the parent molecule; R2 is selected from the group consisting of: allyl, cyclopropylmethyl, isopropyl, cyclopropyl, ethyl, and propyl;
    and R3 is selected from the group consisting of
  • Figure US20230109104A1-20230406-C00202
    Figure US20230109104A1-20230406-C00203
  • In some embodiments, provided is a compound of Formula (I), wherein: A, B, R1 and R4 together are
  • Figure US20230109104A1-20230406-C00204
  • wherein the wavy lines denote attachment points to the parent molecule;
    R2 is selected from the group consisting of: allyl, cyclopropylmethyl, isopropyl, cyclopropyl, ethyl, and propyl; and R3 is
  • Figure US20230109104A1-20230406-C00205
  • Also provided are salts of compounds referred to herein, such as pharmaceutically acceptable salts. The invention also includes any or all of the stereochemical forms, including any enantiomeric or diastereomeric forms, and any tautomers or other forms of the compounds described. In some embodiments, an isomer of a compound detained herein is a stereoisomer.
  • A compound as detailed herein may in one aspect be in a purified form and compositions comprising a compound in purified forms are detailed herein. Compositions comprising a compound as detailed herein or a salt thereof are provided, such as compositions of substantially pure compounds. Compositions comprising a compound as detailed herein or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing are provided, such as compositions of substantially pure compounds. In some embodiments, a composition containing a compound as detailed herein or a salt thereof is in substantially pure form. Unless otherwise stated, “substantially pure” intends a composition that contains no more than 35% impurity, wherein the impurity denotes a compound other than the compound comprising the majority of the composition or a salt thereof. In some embodiments, a composition of substantially pure compound or a salt thereof is provided wherein the composition contains no more than 25%, 20%, 15%, 10%, or 5% impurity. In some embodiments, a composition of substantially pure compound or a salt thereof is provided wherein the composition contains or no more than 3%, 2%, 1% or 0.5% impurity.
  • Representative compounds are listed in Table 1A and Table 1B.
  • TABLE 1A
    Compound
    No. Structure
    1.1
    Figure US20230109104A1-20230406-C00206
    1.2
    Figure US20230109104A1-20230406-C00207
    1.3
    Figure US20230109104A1-20230406-C00208
    1.4
    Figure US20230109104A1-20230406-C00209
    1.5
    Figure US20230109104A1-20230406-C00210
    1.6
    Figure US20230109104A1-20230406-C00211
    1.7
    Figure US20230109104A1-20230406-C00212
    1.8
    Figure US20230109104A1-20230406-C00213
    1.9
    Figure US20230109104A1-20230406-C00214
    1.10
    Figure US20230109104A1-20230406-C00215
    1.11
    Figure US20230109104A1-20230406-C00216
    1.12
    Figure US20230109104A1-20230406-C00217
    1.13
    Figure US20230109104A1-20230406-C00218
    1.14
    Figure US20230109104A1-20230406-C00219
    1.15
    Figure US20230109104A1-20230406-C00220
    1.16
    Figure US20230109104A1-20230406-C00221
    1.17
    Figure US20230109104A1-20230406-C00222
    1.18
    Figure US20230109104A1-20230406-C00223
    1.19
    Figure US20230109104A1-20230406-C00224
    1.20
    Figure US20230109104A1-20230406-C00225
    1.21
    Figure US20230109104A1-20230406-C00226
    1.22
    Figure US20230109104A1-20230406-C00227
    1.23
    Figure US20230109104A1-20230406-C00228
    1.24
    Figure US20230109104A1-20230406-C00229
    1.25
    Figure US20230109104A1-20230406-C00230
    1.26
    Figure US20230109104A1-20230406-C00231
    1.27
    Figure US20230109104A1-20230406-C00232
    1.28
    Figure US20230109104A1-20230406-C00233
    1.29
    Figure US20230109104A1-20230406-C00234
    1.30
    Figure US20230109104A1-20230406-C00235
    1.31
    Figure US20230109104A1-20230406-C00236
    1.32
    Figure US20230109104A1-20230406-C00237
    1.33
    Figure US20230109104A1-20230406-C00238
    1.34
    Figure US20230109104A1-20230406-C00239
    1.35
    Figure US20230109104A1-20230406-C00240
    1.36
    Figure US20230109104A1-20230406-C00241
    1.37
    Figure US20230109104A1-20230406-C00242
    1.38
    Figure US20230109104A1-20230406-C00243
    1.39
    Figure US20230109104A1-20230406-C00244
    1.40
    Figure US20230109104A1-20230406-C00245
    1.41
    Figure US20230109104A1-20230406-C00246
    1.42
    Figure US20230109104A1-20230406-C00247
    1.43
    Figure US20230109104A1-20230406-C00248
    1.44
    Figure US20230109104A1-20230406-C00249
    1.45
    Figure US20230109104A1-20230406-C00250
    1.46
    Figure US20230109104A1-20230406-C00251
    1.47
    Figure US20230109104A1-20230406-C00252
    1.48
    Figure US20230109104A1-20230406-C00253
    1.49
    Figure US20230109104A1-20230406-C00254
    1.50
    Figure US20230109104A1-20230406-C00255
    1.51
    Figure US20230109104A1-20230406-C00256
    1.52
    Figure US20230109104A1-20230406-C00257
    1.53
    Figure US20230109104A1-20230406-C00258
    1.54
    Figure US20230109104A1-20230406-C00259
    1.55
    Figure US20230109104A1-20230406-C00260
    1.56
    Figure US20230109104A1-20230406-C00261
    1.57
    Figure US20230109104A1-20230406-C00262
    1.58
    Figure US20230109104A1-20230406-C00263
    1.59
    Figure US20230109104A1-20230406-C00264
    1.60
    Figure US20230109104A1-20230406-C00265
    1.61
    Figure US20230109104A1-20230406-C00266
    1.62
    Figure US20230109104A1-20230406-C00267
    1.63
    Figure US20230109104A1-20230406-C00268
    1.64
    Figure US20230109104A1-20230406-C00269
    1.65
    Figure US20230109104A1-20230406-C00270
    1.66
    Figure US20230109104A1-20230406-C00271
    1.67
    Figure US20230109104A1-20230406-C00272
    1.68
    Figure US20230109104A1-20230406-C00273
    1.69
    Figure US20230109104A1-20230406-C00274
    1.70
    Figure US20230109104A1-20230406-C00275
    1.71
    Figure US20230109104A1-20230406-C00276
    1.72
    Figure US20230109104A1-20230406-C00277
    1.73
    Figure US20230109104A1-20230406-C00278
    1.74
    Figure US20230109104A1-20230406-C00279
    1.75
    Figure US20230109104A1-20230406-C00280
    1.76
    Figure US20230109104A1-20230406-C00281
    1.77
    Figure US20230109104A1-20230406-C00282
    1.78
    Figure US20230109104A1-20230406-C00283
    1.79
    Figure US20230109104A1-20230406-C00284
    1.80
    Figure US20230109104A1-20230406-C00285
    1.81
    Figure US20230109104A1-20230406-C00286
    1.82
    Figure US20230109104A1-20230406-C00287
    1.83
    Figure US20230109104A1-20230406-C00288
    1.84
    Figure US20230109104A1-20230406-C00289
    1.85
    Figure US20230109104A1-20230406-C00290
    1.86
    Figure US20230109104A1-20230406-C00291
    1.87
    Figure US20230109104A1-20230406-C00292
    1.88
    Figure US20230109104A1-20230406-C00293
    1.89
    Figure US20230109104A1-20230406-C00294
    1.90
    Figure US20230109104A1-20230406-C00295
    1.91
    Figure US20230109104A1-20230406-C00296
    1.92
    Figure US20230109104A1-20230406-C00297
    1.93
    Figure US20230109104A1-20230406-C00298
    1.94
    Figure US20230109104A1-20230406-C00299
    1.95
    Figure US20230109104A1-20230406-C00300
    1.96
    Figure US20230109104A1-20230406-C00301
    1.97
    Figure US20230109104A1-20230406-C00302
    1.98
    Figure US20230109104A1-20230406-C00303
    1.99
    Figure US20230109104A1-20230406-C00304
    1.100
    Figure US20230109104A1-20230406-C00305
    1.101
    Figure US20230109104A1-20230406-C00306
    1.102
    Figure US20230109104A1-20230406-C00307
    1.103
    Figure US20230109104A1-20230406-C00308
    1.104
    Figure US20230109104A1-20230406-C00309
    1.105
    Figure US20230109104A1-20230406-C00310
    1.106
    Figure US20230109104A1-20230406-C00311
    1.107
    Figure US20230109104A1-20230406-C00312
    1.108
    Figure US20230109104A1-20230406-C00313
    1.109
    Figure US20230109104A1-20230406-C00314
    1.110
    Figure US20230109104A1-20230406-C00315
    1.111
    Figure US20230109104A1-20230406-C00316
    1.112
    Figure US20230109104A1-20230406-C00317
    1.113
    Figure US20230109104A1-20230406-C00318
    1.114
    Figure US20230109104A1-20230406-C00319
    1.115
    Figure US20230109104A1-20230406-C00320
    1.116
    Figure US20230109104A1-20230406-C00321
    1.117
    Figure US20230109104A1-20230406-C00322
    1.118
    Figure US20230109104A1-20230406-C00323
    1.119
    Figure US20230109104A1-20230406-C00324
    1.120
    Figure US20230109104A1-20230406-C00325
    1.121
    Figure US20230109104A1-20230406-C00326
    1.122
    Figure US20230109104A1-20230406-C00327
    1.123
    Figure US20230109104A1-20230406-C00328
    1.124
    Figure US20230109104A1-20230406-C00329
    1.125
    Figure US20230109104A1-20230406-C00330
    1.126
    Figure US20230109104A1-20230406-C00331
    1.127
    Figure US20230109104A1-20230406-C00332
    1.128
    Figure US20230109104A1-20230406-C00333
    1.129
    Figure US20230109104A1-20230406-C00334
    1.130
    Figure US20230109104A1-20230406-C00335
    1.131
    Figure US20230109104A1-20230406-C00336
    1.132
    Figure US20230109104A1-20230406-C00337
    1.133
    Figure US20230109104A1-20230406-C00338
    1.134
    Figure US20230109104A1-20230406-C00339
    1.135
    Figure US20230109104A1-20230406-C00340
    1.136
    Figure US20230109104A1-20230406-C00341
    1.137
    Figure US20230109104A1-20230406-C00342
    1.138
    Figure US20230109104A1-20230406-C00343
    1.139
    Figure US20230109104A1-20230406-C00344
    1.140
    Figure US20230109104A1-20230406-C00345
    1.141
    Figure US20230109104A1-20230406-C00346
    1.142
    Figure US20230109104A1-20230406-C00347
    1.143
    Figure US20230109104A1-20230406-C00348
    1.144
    Figure US20230109104A1-20230406-C00349
    1.145
    Figure US20230109104A1-20230406-C00350
    1.146
    Figure US20230109104A1-20230406-C00351
    1.147
    Figure US20230109104A1-20230406-C00352
    1.148
    Figure US20230109104A1-20230406-C00353
    1.149
    Figure US20230109104A1-20230406-C00354
    1.150
    Figure US20230109104A1-20230406-C00355
    1.151
    Figure US20230109104A1-20230406-C00356
    1.152
    Figure US20230109104A1-20230406-C00357
    1.153
    Figure US20230109104A1-20230406-C00358
    1.154
    Figure US20230109104A1-20230406-C00359
    1.155
    Figure US20230109104A1-20230406-C00360
    1.156
    Figure US20230109104A1-20230406-C00361
    1.157
    Figure US20230109104A1-20230406-C00362
    1.158
    Figure US20230109104A1-20230406-C00363
    1.159
    Figure US20230109104A1-20230406-C00364
    1.160
    Figure US20230109104A1-20230406-C00365
    1.161
    Figure US20230109104A1-20230406-C00366
    1.162
    Figure US20230109104A1-20230406-C00367
    1.163
    Figure US20230109104A1-20230406-C00368
    1.164
    Figure US20230109104A1-20230406-C00369
    1.165
    Figure US20230109104A1-20230406-C00370
    1.166
    Figure US20230109104A1-20230406-C00371
    1.167
    Figure US20230109104A1-20230406-C00372
    1.168
    Figure US20230109104A1-20230406-C00373
    1.169
    Figure US20230109104A1-20230406-C00374
    1.170
    Figure US20230109104A1-20230406-C00375
    1.171
    Figure US20230109104A1-20230406-C00376
    1.172
    Figure US20230109104A1-20230406-C00377
    1.173
    Figure US20230109104A1-20230406-C00378
    1.174
    Figure US20230109104A1-20230406-C00379
    1.175
    Figure US20230109104A1-20230406-C00380
    1.176
    Figure US20230109104A1-20230406-C00381
    1.177
    Figure US20230109104A1-20230406-C00382
    1.178
    Figure US20230109104A1-20230406-C00383
    1.179
    Figure US20230109104A1-20230406-C00384
    1.180
    Figure US20230109104A1-20230406-C00385
    1.181
    Figure US20230109104A1-20230406-C00386
    1.182
    Figure US20230109104A1-20230406-C00387
    1.183
    Figure US20230109104A1-20230406-C00388
    1.184
    Figure US20230109104A1-20230406-C00389
    1.185
    Figure US20230109104A1-20230406-C00390
    1.186
    Figure US20230109104A1-20230406-C00391
    1.187
    Figure US20230109104A1-20230406-C00392
    1.188
    Figure US20230109104A1-20230406-C00393
    1.189
    Figure US20230109104A1-20230406-C00394
    1.190
    Figure US20230109104A1-20230406-C00395
    1.191
    Figure US20230109104A1-20230406-C00396
    1.192
    Figure US20230109104A1-20230406-C00397
    1.193
    Figure US20230109104A1-20230406-C00398
    1.194
    Figure US20230109104A1-20230406-C00399
    1.195
    Figure US20230109104A1-20230406-C00400
    1.196
    Figure US20230109104A1-20230406-C00401
    1.197
    Figure US20230109104A1-20230406-C00402
    1.198
    Figure US20230109104A1-20230406-C00403
    1.199
    Figure US20230109104A1-20230406-C00404
    1.200
    Figure US20230109104A1-20230406-C00405
    1.201
    Figure US20230109104A1-20230406-C00406
    1.202
    Figure US20230109104A1-20230406-C00407
    1.203
    Figure US20230109104A1-20230406-C00408
    1.204
    Figure US20230109104A1-20230406-C00409
    1.205
    Figure US20230109104A1-20230406-C00410
    1.206
    Figure US20230109104A1-20230406-C00411
    1.207
    Figure US20230109104A1-20230406-C00412
    1.208
    Figure US20230109104A1-20230406-C00413
    1.209
    Figure US20230109104A1-20230406-C00414
    1.210
    Figure US20230109104A1-20230406-C00415
    1.211
    Figure US20230109104A1-20230406-C00416
    1.212
    Figure US20230109104A1-20230406-C00417
    1.213
    Figure US20230109104A1-20230406-C00418
    1.214
    Figure US20230109104A1-20230406-C00419
    1.215
    Figure US20230109104A1-20230406-C00420
    1.216
    Figure US20230109104A1-20230406-C00421
    1.217
    Figure US20230109104A1-20230406-C00422
    1.218
    Figure US20230109104A1-20230406-C00423
    1.219
    Figure US20230109104A1-20230406-C00424
    1.220
    Figure US20230109104A1-20230406-C00425
    1.221
    Figure US20230109104A1-20230406-C00426
    1.222
    Figure US20230109104A1-20230406-C00427
    1.223
    Figure US20230109104A1-20230406-C00428
    1.224
    Figure US20230109104A1-20230406-C00429
    1.225
    Figure US20230109104A1-20230406-C00430
    1.226
    Figure US20230109104A1-20230406-C00431
    1.227
    Figure US20230109104A1-20230406-C00432
    1.228
    Figure US20230109104A1-20230406-C00433
    1.229
    Figure US20230109104A1-20230406-C00434
    1.230
    Figure US20230109104A1-20230406-C00435
    1.231
    Figure US20230109104A1-20230406-C00436
    1.232
    Figure US20230109104A1-20230406-C00437
    1.233
    Figure US20230109104A1-20230406-C00438
    1.234
    Figure US20230109104A1-20230406-C00439
    1.235
    Figure US20230109104A1-20230406-C00440
    1.236
    Figure US20230109104A1-20230406-C00441
    1.237
    Figure US20230109104A1-20230406-C00442
    1.238
    Figure US20230109104A1-20230406-C00443
    1.239
    Figure US20230109104A1-20230406-C00444
    1.240
    Figure US20230109104A1-20230406-C00445
    1.241
    Figure US20230109104A1-20230406-C00446
    1.242
    Figure US20230109104A1-20230406-C00447
    1.243
    Figure US20230109104A1-20230406-C00448
    1.244
    Figure US20230109104A1-20230406-C00449
    1.245
    Figure US20230109104A1-20230406-C00450
    1.246
    Figure US20230109104A1-20230406-C00451
    1.247
    Figure US20230109104A1-20230406-C00452
    1.248
    Figure US20230109104A1-20230406-C00453
    1.249
    Figure US20230109104A1-20230406-C00454
    1.250
    Figure US20230109104A1-20230406-C00455
    1.251
    Figure US20230109104A1-20230406-C00456
    1.252
    Figure US20230109104A1-20230406-C00457
    1.253
    Figure US20230109104A1-20230406-C00458
    1.254
    Figure US20230109104A1-20230406-C00459
    1.255
    Figure US20230109104A1-20230406-C00460
    1.256
    Figure US20230109104A1-20230406-C00461
    1.257
    Figure US20230109104A1-20230406-C00462
    1.258
    Figure US20230109104A1-20230406-C00463
    1.259
    Figure US20230109104A1-20230406-C00464
    1.260
    Figure US20230109104A1-20230406-C00465
    1.261
    Figure US20230109104A1-20230406-C00466
    1.262
    Figure US20230109104A1-20230406-C00467
    1.263
    Figure US20230109104A1-20230406-C00468
    1.264
    Figure US20230109104A1-20230406-C00469
    1.265
    Figure US20230109104A1-20230406-C00470
    1.266
    Figure US20230109104A1-20230406-C00471
    1.267
    Figure US20230109104A1-20230406-C00472
    1.268
    Figure US20230109104A1-20230406-C00473
    1.269
    Figure US20230109104A1-20230406-C00474
    1.270
    Figure US20230109104A1-20230406-C00475
    1.271
    Figure US20230109104A1-20230406-C00476
    1.272
    Figure US20230109104A1-20230406-C00477
    1.273
    Figure US20230109104A1-20230406-C00478
    1.274
    Figure US20230109104A1-20230406-C00479
    1.275
    Figure US20230109104A1-20230406-C00480
    1.276
    Figure US20230109104A1-20230406-C00481
    1.277
    Figure US20230109104A1-20230406-C00482
    1.278
    Figure US20230109104A1-20230406-C00483
    1.279
    Figure US20230109104A1-20230406-C00484
    1.280
    Figure US20230109104A1-20230406-C00485
    1.281
    Figure US20230109104A1-20230406-C00486
    1.282
    Figure US20230109104A1-20230406-C00487
    1.283
    Figure US20230109104A1-20230406-C00488
    1.284
    Figure US20230109104A1-20230406-C00489
    1.285
    Figure US20230109104A1-20230406-C00490
    1.286
    Figure US20230109104A1-20230406-C00491
    1.287
    Figure US20230109104A1-20230406-C00492
    1.288
    Figure US20230109104A1-20230406-C00493
    1.289
    Figure US20230109104A1-20230406-C00494
    1.290
    Figure US20230109104A1-20230406-C00495
    1.291
    Figure US20230109104A1-20230406-C00496
    1.292
    Figure US20230109104A1-20230406-C00497
    1.293
    Figure US20230109104A1-20230406-C00498
    1.294
    Figure US20230109104A1-20230406-C00499
    1.295
    Figure US20230109104A1-20230406-C00500
    1.296
    Figure US20230109104A1-20230406-C00501
    1.297
    Figure US20230109104A1-20230406-C00502
    1.298
    Figure US20230109104A1-20230406-C00503
    1.299
    Figure US20230109104A1-20230406-C00504
    1.300
    Figure US20230109104A1-20230406-C00505
    1.301
    Figure US20230109104A1-20230406-C00506
    1.302
    Figure US20230109104A1-20230406-C00507
    1.303
    Figure US20230109104A1-20230406-C00508
    1.304
    Figure US20230109104A1-20230406-C00509
    1.305
    Figure US20230109104A1-20230406-C00510
    1.306
    Figure US20230109104A1-20230406-C00511
    1.307
    Figure US20230109104A1-20230406-C00512
    1.308
    Figure US20230109104A1-20230406-C00513
    1.309
    Figure US20230109104A1-20230406-C00514
    1.310
    Figure US20230109104A1-20230406-C00515
    1.311
    Figure US20230109104A1-20230406-C00516
    1.312
    Figure US20230109104A1-20230406-C00517
    1.313
    Figure US20230109104A1-20230406-C00518
    1.314
    Figure US20230109104A1-20230406-C00519
    1.315
    Figure US20230109104A1-20230406-C00520
    1.316
    Figure US20230109104A1-20230406-C00521
    1.317
    Figure US20230109104A1-20230406-C00522
    1.318
    Figure US20230109104A1-20230406-C00523
    1.319
    Figure US20230109104A1-20230406-C00524
    1.320
    Figure US20230109104A1-20230406-C00525
    1.321
    Figure US20230109104A1-20230406-C00526
    1.322
    Figure US20230109104A1-20230406-C00527
    1.323
    Figure US20230109104A1-20230406-C00528
    1.324
    Figure US20230109104A1-20230406-C00529
    1.325
    Figure US20230109104A1-20230406-C00530
    1.326
    Figure US20230109104A1-20230406-C00531
    1.327
    Figure US20230109104A1-20230406-C00532
    1.328
    Figure US20230109104A1-20230406-C00533
    1.329
    Figure US20230109104A1-20230406-C00534
    1.330
    Figure US20230109104A1-20230406-C00535
    1.331
    Figure US20230109104A1-20230406-C00536
    1.332
    Figure US20230109104A1-20230406-C00537
    1.333
    Figure US20230109104A1-20230406-C00538
    1.334
    Figure US20230109104A1-20230406-C00539
    1.335
    Figure US20230109104A1-20230406-C00540
    1.336
    Figure US20230109104A1-20230406-C00541
    1.337
    Figure US20230109104A1-20230406-C00542
    1.338
    Figure US20230109104A1-20230406-C00543
    1.339
    Figure US20230109104A1-20230406-C00544
    1.340
    Figure US20230109104A1-20230406-C00545
    1.341
    Figure US20230109104A1-20230406-C00546
    1.342
    Figure US20230109104A1-20230406-C00547
    1.343
    Figure US20230109104A1-20230406-C00548
    1.344
    Figure US20230109104A1-20230406-C00549
    1.345
    Figure US20230109104A1-20230406-C00550
    1.346
    Figure US20230109104A1-20230406-C00551
    1.347
    Figure US20230109104A1-20230406-C00552
    1.348
    Figure US20230109104A1-20230406-C00553
    1.349
    Figure US20230109104A1-20230406-C00554
    1.350
    Figure US20230109104A1-20230406-C00555
    1.351
    Figure US20230109104A1-20230406-C00556
    1.352
    Figure US20230109104A1-20230406-C00557
    1.353
    Figure US20230109104A1-20230406-C00558
    1.354
    Figure US20230109104A1-20230406-C00559
    1.355
    Figure US20230109104A1-20230406-C00560
    1.356
    Figure US20230109104A1-20230406-C00561
    1.357
    Figure US20230109104A1-20230406-C00562
    1.358
    Figure US20230109104A1-20230406-C00563
    1.359
    Figure US20230109104A1-20230406-C00564
    1.360
    Figure US20230109104A1-20230406-C00565
    1.361
    Figure US20230109104A1-20230406-C00566
    1.362
    Figure US20230109104A1-20230406-C00567
    1.363
    Figure US20230109104A1-20230406-C00568
    1.364
    Figure US20230109104A1-20230406-C00569
    1.365
    Figure US20230109104A1-20230406-C00570
    1.366
    Figure US20230109104A1-20230406-C00571
    1.367
    Figure US20230109104A1-20230406-C00572
    1.368
    Figure US20230109104A1-20230406-C00573
    1.369
    Figure US20230109104A1-20230406-C00574
    1.370
    Figure US20230109104A1-20230406-C00575
    1.371
    Figure US20230109104A1-20230406-C00576
    1.372
    Figure US20230109104A1-20230406-C00577
    1.373
    Figure US20230109104A1-20230406-C00578
    1.374
    Figure US20230109104A1-20230406-C00579
    1.375
    Figure US20230109104A1-20230406-C00580
    1.376
    Figure US20230109104A1-20230406-C00581
    1.377
    Figure US20230109104A1-20230406-C00582
    1.378
    Figure US20230109104A1-20230406-C00583
    1.379
    Figure US20230109104A1-20230406-C00584
    1.380
    Figure US20230109104A1-20230406-C00585
    1.381
    Figure US20230109104A1-20230406-C00586
    1.382
    Figure US20230109104A1-20230406-C00587
    1.383
    Figure US20230109104A1-20230406-C00588
    1.384
    Figure US20230109104A1-20230406-C00589
    1.385
    Figure US20230109104A1-20230406-C00590
    1.386
    Figure US20230109104A1-20230406-C00591
    1.387
    Figure US20230109104A1-20230406-C00592
    1.388
    Figure US20230109104A1-20230406-C00593
    1.389
    Figure US20230109104A1-20230406-C00594
    1.390
    Figure US20230109104A1-20230406-C00595
    1.391
    Figure US20230109104A1-20230406-C00596
    1.392
    Figure US20230109104A1-20230406-C00597
    1.393
    Figure US20230109104A1-20230406-C00598
    1.394
    Figure US20230109104A1-20230406-C00599
    1.395
    Figure US20230109104A1-20230406-C00600
    1.396
    Figure US20230109104A1-20230406-C00601
    1.397
    Figure US20230109104A1-20230406-C00602
    1.398
    Figure US20230109104A1-20230406-C00603
    1.399
    Figure US20230109104A1-20230406-C00604
    1.400
    Figure US20230109104A1-20230406-C00605
    1.401
    Figure US20230109104A1-20230406-C00606
    1.402
    Figure US20230109104A1-20230406-C00607
    1.403
    Figure US20230109104A1-20230406-C00608
    1.404
    Figure US20230109104A1-20230406-C00609
    1.405
    Figure US20230109104A1-20230406-C00610
    1.406
    Figure US20230109104A1-20230406-C00611
    1.407
    Figure US20230109104A1-20230406-C00612
    1.408
    Figure US20230109104A1-20230406-C00613
    1.409
    Figure US20230109104A1-20230406-C00614
    1.410
    Figure US20230109104A1-20230406-C00615
    1.411
    Figure US20230109104A1-20230406-C00616
    1.412
    Figure US20230109104A1-20230406-C00617
    1.413
    Figure US20230109104A1-20230406-C00618
    1.414
    Figure US20230109104A1-20230406-C00619
    1.415
    Figure US20230109104A1-20230406-C00620
    1.416
    Figure US20230109104A1-20230406-C00621
    1.417
    Figure US20230109104A1-20230406-C00622
    1.418
    Figure US20230109104A1-20230406-C00623
    1.419
    Figure US20230109104A1-20230406-C00624
    1.420
    Figure US20230109104A1-20230406-C00625
    1.421
    Figure US20230109104A1-20230406-C00626
    1.422
    Figure US20230109104A1-20230406-C00627
    1.423
    Figure US20230109104A1-20230406-C00628
    1.424
    Figure US20230109104A1-20230406-C00629
    1.425
    Figure US20230109104A1-20230406-C00630
    1.426
    Figure US20230109104A1-20230406-C00631
    1.427
    Figure US20230109104A1-20230406-C00632
    1.428
    Figure US20230109104A1-20230406-C00633
    1.429
    Figure US20230109104A1-20230406-C00634
    1.430
    Figure US20230109104A1-20230406-C00635
    1.431
    Figure US20230109104A1-20230406-C00636
    1.432
    Figure US20230109104A1-20230406-C00637
    1.433
    Figure US20230109104A1-20230406-C00638
    1.434
    Figure US20230109104A1-20230406-C00639
    1.435
    Figure US20230109104A1-20230406-C00640
    1.436
    Figure US20230109104A1-20230406-C00641
    1.437
    Figure US20230109104A1-20230406-C00642
    1.438
    Figure US20230109104A1-20230406-C00643
    1.439
    Figure US20230109104A1-20230406-C00644
    1.440
    Figure US20230109104A1-20230406-C00645
    1.441
    Figure US20230109104A1-20230406-C00646
    1.442
    Figure US20230109104A1-20230406-C00647
    1.443
    Figure US20230109104A1-20230406-C00648
    1.444
    Figure US20230109104A1-20230406-C00649
    1.445
    Figure US20230109104A1-20230406-C00650
    1.446
    Figure US20230109104A1-20230406-C00651
    1.447
    Figure US20230109104A1-20230406-C00652
    1.448
    Figure US20230109104A1-20230406-C00653
    1.449
    Figure US20230109104A1-20230406-C00654
    1.450
    Figure US20230109104A1-20230406-C00655
    1.451
    Figure US20230109104A1-20230406-C00656
    1.452
    Figure US20230109104A1-20230406-C00657
    1.453
    Figure US20230109104A1-20230406-C00658
    1.454
    Figure US20230109104A1-20230406-C00659
    1.455
    Figure US20230109104A1-20230406-C00660
    1.456
    Figure US20230109104A1-20230406-C00661
    1.457
    Figure US20230109104A1-20230406-C00662
    1.458
    Figure US20230109104A1-20230406-C00663
    1.459
    Figure US20230109104A1-20230406-C00664
    1.460
    Figure US20230109104A1-20230406-C00665
    1.461
    Figure US20230109104A1-20230406-C00666
    1.462
    Figure US20230109104A1-20230406-C00667
    1.463
    Figure US20230109104A1-20230406-C00668
    1.464
    Figure US20230109104A1-20230406-C00669
    1.465
    Figure US20230109104A1-20230406-C00670
    1.466
    Figure US20230109104A1-20230406-C00671
    1.467
    Figure US20230109104A1-20230406-C00672
    1.468
    Figure US20230109104A1-20230406-C00673
    1.469
    Figure US20230109104A1-20230406-C00674
    1.470
    Figure US20230109104A1-20230406-C00675
    1.471
    Figure US20230109104A1-20230406-C00676
    1.472
    Figure US20230109104A1-20230406-C00677
    1.473
    Figure US20230109104A1-20230406-C00678
    1.474
    Figure US20230109104A1-20230406-C00679
    1.475
    Figure US20230109104A1-20230406-C00680
    1.476
    Figure US20230109104A1-20230406-C00681
    1.477
    Figure US20230109104A1-20230406-C00682
    1.478
    Figure US20230109104A1-20230406-C00683
    1.479
    Figure US20230109104A1-20230406-C00684
    1.480
    Figure US20230109104A1-20230406-C00685
    1.481
    Figure US20230109104A1-20230406-C00686
    1.482
    Figure US20230109104A1-20230406-C00687
    1.483
    Figure US20230109104A1-20230406-C00688
    1.484
    Figure US20230109104A1-20230406-C00689
    1.485
    Figure US20230109104A1-20230406-C00690
    1.486
    Figure US20230109104A1-20230406-C00691
    1.487
    Figure US20230109104A1-20230406-C00692
    1.488
    Figure US20230109104A1-20230406-C00693
    1.489
    Figure US20230109104A1-20230406-C00694
    1.490
    Figure US20230109104A1-20230406-C00695
    1.491
    Figure US20230109104A1-20230406-C00696
    1.492
    Figure US20230109104A1-20230406-C00697
    1.493
    Figure US20230109104A1-20230406-C00698
    1.494
    Figure US20230109104A1-20230406-C00699
    1.495
    Figure US20230109104A1-20230406-C00700
    1.496
    Figure US20230109104A1-20230406-C00701
    1.497
    Figure US20230109104A1-20230406-C00702
    1.498
    Figure US20230109104A1-20230406-C00703
    1.499
    Figure US20230109104A1-20230406-C00704
    1.500
    Figure US20230109104A1-20230406-C00705
    1.501
    Figure US20230109104A1-20230406-C00706
    1.502
    Figure US20230109104A1-20230406-C00707
    1.503
    Figure US20230109104A1-20230406-C00708
    1.504
    Figure US20230109104A1-20230406-C00709
    1.505
    Figure US20230109104A1-20230406-C00710
    1.506
    Figure US20230109104A1-20230406-C00711
    1.507
    Figure US20230109104A1-20230406-C00712
    1.508
    Figure US20230109104A1-20230406-C00713
    1.509
    Figure US20230109104A1-20230406-C00714
    1.510
    Figure US20230109104A1-20230406-C00715
    1.511
    Figure US20230109104A1-20230406-C00716
    1.512
    Figure US20230109104A1-20230406-C00717
    1.513
    Figure US20230109104A1-20230406-C00718
    1.514
    Figure US20230109104A1-20230406-C00719
    1.515
    Figure US20230109104A1-20230406-C00720
    1.516
    Figure US20230109104A1-20230406-C00721
    1.517
    Figure US20230109104A1-20230406-C00722
    1.518
    Figure US20230109104A1-20230406-C00723
    1.519
    Figure US20230109104A1-20230406-C00724
    1.520
    Figure US20230109104A1-20230406-C00725
    1.521
    Figure US20230109104A1-20230406-C00726
    1.522
    Figure US20230109104A1-20230406-C00727
    1.523
    Figure US20230109104A1-20230406-C00728
    1.524
    Figure US20230109104A1-20230406-C00729
    1.525
    Figure US20230109104A1-20230406-C00730
    1.526
    Figure US20230109104A1-20230406-C00731
    1.527
    Figure US20230109104A1-20230406-C00732
    1.528
    Figure US20230109104A1-20230406-C00733
    1.529
    Figure US20230109104A1-20230406-C00734
    1.530
    Figure US20230109104A1-20230406-C00735
    1.531
    Figure US20230109104A1-20230406-C00736
    1.532
    Figure US20230109104A1-20230406-C00737
    1.533
    Figure US20230109104A1-20230406-C00738
    1.534
    Figure US20230109104A1-20230406-C00739
    1.535
    Figure US20230109104A1-20230406-C00740
    1.536
    Figure US20230109104A1-20230406-C00741
    1.537
    Figure US20230109104A1-20230406-C00742
    1.538
    Figure US20230109104A1-20230406-C00743
    1.539
    Figure US20230109104A1-20230406-C00744
    1.540
    Figure US20230109104A1-20230406-C00745
    1.541
    Figure US20230109104A1-20230406-C00746
    1.542
    Figure US20230109104A1-20230406-C00747
    1.543
    Figure US20230109104A1-20230406-C00748
    1.544
    Figure US20230109104A1-20230406-C00749
    1.545
    Figure US20230109104A1-20230406-C00750
    1.546
    Figure US20230109104A1-20230406-C00751
    1.547
    Figure US20230109104A1-20230406-C00752
    1.548
    Figure US20230109104A1-20230406-C00753
    1.549
    Figure US20230109104A1-20230406-C00754
    1.550
    Figure US20230109104A1-20230406-C00755
    1.551
    Figure US20230109104A1-20230406-C00756
    1.552
    Figure US20230109104A1-20230406-C00757
    1.553
    Figure US20230109104A1-20230406-C00758
    1.554
    Figure US20230109104A1-20230406-C00759
    1.555
    Figure US20230109104A1-20230406-C00760
    1.556
    Figure US20230109104A1-20230406-C00761
    1.557
    Figure US20230109104A1-20230406-C00762
    1.558
    Figure US20230109104A1-20230406-C00763
    1.559
    Figure US20230109104A1-20230406-C00764
    1.560
    Figure US20230109104A1-20230406-C00765
    1.561
    Figure US20230109104A1-20230406-C00766
    1.562
    Figure US20230109104A1-20230406-C00767
    1.563
    Figure US20230109104A1-20230406-C00768
    1.564
    Figure US20230109104A1-20230406-C00769
    1.565
    Figure US20230109104A1-20230406-C00770
    1.566
    Figure US20230109104A1-20230406-C00771
    1.567
    Figure US20230109104A1-20230406-C00772
    1.568
    Figure US20230109104A1-20230406-C00773
    1.569
    Figure US20230109104A1-20230406-C00774
    1.570
    Figure US20230109104A1-20230406-C00775
    1.571
    Figure US20230109104A1-20230406-C00776
    1.572
    Figure US20230109104A1-20230406-C00777
    1.573
    Figure US20230109104A1-20230406-C00778
    1.574
    Figure US20230109104A1-20230406-C00779
    1.575
    Figure US20230109104A1-20230406-C00780
    1.576
    Figure US20230109104A1-20230406-C00781
    1.577
    Figure US20230109104A1-20230406-C00782
    1.578
    Figure US20230109104A1-20230406-C00783
    1.579
    Figure US20230109104A1-20230406-C00784
    1.580
    Figure US20230109104A1-20230406-C00785
    1.581
    Figure US20230109104A1-20230406-C00786
    1.582
    Figure US20230109104A1-20230406-C00787
    1.583
    Figure US20230109104A1-20230406-C00788
    1.584
    Figure US20230109104A1-20230406-C00789
    1.585
    Figure US20230109104A1-20230406-C00790
    1.586
    Figure US20230109104A1-20230406-C00791
    1.587
    Figure US20230109104A1-20230406-C00792
    1.588
    Figure US20230109104A1-20230406-C00793
    1.589
    Figure US20230109104A1-20230406-C00794
    1.590
    Figure US20230109104A1-20230406-C00795
    1.591
    Figure US20230109104A1-20230406-C00796
    1.592
    Figure US20230109104A1-20230406-C00797
    1.593
    Figure US20230109104A1-20230406-C00798
    1.594
    Figure US20230109104A1-20230406-C00799
    1.595
    Figure US20230109104A1-20230406-C00800
    1.596
    Figure US20230109104A1-20230406-C00801
    1.597
    Figure US20230109104A1-20230406-C00802
    1.598
    Figure US20230109104A1-20230406-C00803
    1.599
    Figure US20230109104A1-20230406-C00804
    1.600
    Figure US20230109104A1-20230406-C00805
    1.601
    Figure US20230109104A1-20230406-C00806
    1.602
    Figure US20230109104A1-20230406-C00807
    1.603
    Figure US20230109104A1-20230406-C00808
    1.604
    Figure US20230109104A1-20230406-C00809
    1.605
    Figure US20230109104A1-20230406-C00810
    1.606
    Figure US20230109104A1-20230406-C00811
    1.607
    Figure US20230109104A1-20230406-C00812
    1.608
    Figure US20230109104A1-20230406-C00813
    1.609
    Figure US20230109104A1-20230406-C00814
    1.610
    Figure US20230109104A1-20230406-C00815
    1.611
    Figure US20230109104A1-20230406-C00816
    1.612
    Figure US20230109104A1-20230406-C00817
    1.613
    Figure US20230109104A1-20230406-C00818
    1.614
    Figure US20230109104A1-20230406-C00819
    1.615
    Figure US20230109104A1-20230406-C00820
    1.616
    Figure US20230109104A1-20230406-C00821
    1.617
    Figure US20230109104A1-20230406-C00822
    1.618
    Figure US20230109104A1-20230406-C00823
    1.619
    Figure US20230109104A1-20230406-C00824
    1.620
    Figure US20230109104A1-20230406-C00825
    1.621
    Figure US20230109104A1-20230406-C00826
    1.622
    Figure US20230109104A1-20230406-C00827
    1.623
    Figure US20230109104A1-20230406-C00828
    1.624
    Figure US20230109104A1-20230406-C00829
    1.625
    Figure US20230109104A1-20230406-C00830
    1.626
    Figure US20230109104A1-20230406-C00831
    1.627
    Figure US20230109104A1-20230406-C00832
    1.628
    Figure US20230109104A1-20230406-C00833
    1.629
    Figure US20230109104A1-20230406-C00834
    1.630
    Figure US20230109104A1-20230406-C00835
    1.631
    Figure US20230109104A1-20230406-C00836
    1.632
    Figure US20230109104A1-20230406-C00837
    1.633
    Figure US20230109104A1-20230406-C00838
    1.634
    Figure US20230109104A1-20230406-C00839
    1.635
    Figure US20230109104A1-20230406-C00840
    1.636
    Figure US20230109104A1-20230406-C00841
    1.637
    Figure US20230109104A1-20230406-C00842
    1.638
    Figure US20230109104A1-20230406-C00843
    1.639
    Figure US20230109104A1-20230406-C00844
    1.640
    Figure US20230109104A1-20230406-C00845
    1.641
    Figure US20230109104A1-20230406-C00846
    1.642
    Figure US20230109104A1-20230406-C00847
    1.643
    Figure US20230109104A1-20230406-C00848
    1.644
    Figure US20230109104A1-20230406-C00849
    1.645
    Figure US20230109104A1-20230406-C00850
    1.646
    Figure US20230109104A1-20230406-C00851
    1.647
    Figure US20230109104A1-20230406-C00852
    1.648
    Figure US20230109104A1-20230406-C00853
    1.649
    Figure US20230109104A1-20230406-C00854
    1.650
    Figure US20230109104A1-20230406-C00855
    1.651
    Figure US20230109104A1-20230406-C00856
    1.652
    Figure US20230109104A1-20230406-C00857
    1.653
    Figure US20230109104A1-20230406-C00858
    1.654
    Figure US20230109104A1-20230406-C00859
    1.655
    Figure US20230109104A1-20230406-C00860
    1.656
    Figure US20230109104A1-20230406-C00861
    1.657
    Figure US20230109104A1-20230406-C00862
    1.658
    Figure US20230109104A1-20230406-C00863
    1.659
    Figure US20230109104A1-20230406-C00864
    1.660
    Figure US20230109104A1-20230406-C00865
    1.661
    Figure US20230109104A1-20230406-C00866
    1.662
    Figure US20230109104A1-20230406-C00867
    1.663
    Figure US20230109104A1-20230406-C00868
    1.664
    Figure US20230109104A1-20230406-C00869
    1.665
    Figure US20230109104A1-20230406-C00870
    1.666
    Figure US20230109104A1-20230406-C00871
    1.667
    Figure US20230109104A1-20230406-C00872
    1.668
    Figure US20230109104A1-20230406-C00873
    1.669
    Figure US20230109104A1-20230406-C00874
    1.670
    Figure US20230109104A1-20230406-C00875
    1.671
    Figure US20230109104A1-20230406-C00876
    1.672
    Figure US20230109104A1-20230406-C00877
    1.673
    Figure US20230109104A1-20230406-C00878
    1.674
    Figure US20230109104A1-20230406-C00879
    1.675
    Figure US20230109104A1-20230406-C00880
    1.676
    Figure US20230109104A1-20230406-C00881
    1.677
    Figure US20230109104A1-20230406-C00882
    1.678
    Figure US20230109104A1-20230406-C00883
    1.679
    Figure US20230109104A1-20230406-C00884
    1.680
    Figure US20230109104A1-20230406-C00885
    1.681
    Figure US20230109104A1-20230406-C00886
    1.682
    Figure US20230109104A1-20230406-C00887
    1.683
    Figure US20230109104A1-20230406-C00888
    1.684
    Figure US20230109104A1-20230406-C00889
    1.685
    Figure US20230109104A1-20230406-C00890
    1.686
    Figure US20230109104A1-20230406-C00891
    1.687
    Figure US20230109104A1-20230406-C00892
    1.688
    Figure US20230109104A1-20230406-C00893
    1.689
    Figure US20230109104A1-20230406-C00894
    1.690
    Figure US20230109104A1-20230406-C00895
    1.691
    Figure US20230109104A1-20230406-C00896
    1.692
    Figure US20230109104A1-20230406-C00897
    1.693
    Figure US20230109104A1-20230406-C00898
    1.694
    Figure US20230109104A1-20230406-C00899
    1.695
    Figure US20230109104A1-20230406-C00900
    1.696
    Figure US20230109104A1-20230406-C00901
    1.697
    Figure US20230109104A1-20230406-C00902
    1.698
    Figure US20230109104A1-20230406-C00903
    1.699
    Figure US20230109104A1-20230406-C00904
    1.700
    Figure US20230109104A1-20230406-C00905
    1.701
    Figure US20230109104A1-20230406-C00906
    1.702
    Figure US20230109104A1-20230406-C00907
    1.703
    Figure US20230109104A1-20230406-C00908
    1.704
    Figure US20230109104A1-20230406-C00909
    1.705
    Figure US20230109104A1-20230406-C00910
    1.706
    Figure US20230109104A1-20230406-C00911
    1.707
    Figure US20230109104A1-20230406-C00912
    1.708
    Figure US20230109104A1-20230406-C00913
    1.709
    Figure US20230109104A1-20230406-C00914
    1.710
    Figure US20230109104A1-20230406-C00915
    1.711
    Figure US20230109104A1-20230406-C00916
    1.712
    Figure US20230109104A1-20230406-C00917
    1.713
    Figure US20230109104A1-20230406-C00918
    1.714
    Figure US20230109104A1-20230406-C00919
    1.715
    Figure US20230109104A1-20230406-C00920
    1.716
    Figure US20230109104A1-20230406-C00921
    1.717
    Figure US20230109104A1-20230406-C00922
    1.718
    Figure US20230109104A1-20230406-C00923
    1.719
    Figure US20230109104A1-20230406-C00924
    1.720
    Figure US20230109104A1-20230406-C00925
    1.721
    Figure US20230109104A1-20230406-C00926
    1.722
    Figure US20230109104A1-20230406-C00927
    1.723
    Figure US20230109104A1-20230406-C00928
    1.724
    Figure US20230109104A1-20230406-C00929
    1.725
    Figure US20230109104A1-20230406-C00930
    1.726
    Figure US20230109104A1-20230406-C00931
    1.727
    Figure US20230109104A1-20230406-C00932
    1.728
    Figure US20230109104A1-20230406-C00933
    1.729
    Figure US20230109104A1-20230406-C00934
    1.730
    Figure US20230109104A1-20230406-C00935
    1.731
    Figure US20230109104A1-20230406-C00936
    1.732
    Figure US20230109104A1-20230406-C00937
    1.733
    Figure US20230109104A1-20230406-C00938
    1.734
    Figure US20230109104A1-20230406-C00939
    1.735
    Figure US20230109104A1-20230406-C00940
    1.736
    Figure US20230109104A1-20230406-C00941
    1.737
    Figure US20230109104A1-20230406-C00942
    1.738
    Figure US20230109104A1-20230406-C00943
    1.739
    Figure US20230109104A1-20230406-C00944
    1.740
    Figure US20230109104A1-20230406-C00945
    1.741
    Figure US20230109104A1-20230406-C00946
    1.742
    Figure US20230109104A1-20230406-C00947
    1.743
    Figure US20230109104A1-20230406-C00948
    1.744
    Figure US20230109104A1-20230406-C00949
    1.745
    Figure US20230109104A1-20230406-C00950
    1.746
    Figure US20230109104A1-20230406-C00951
    1.747
    Figure US20230109104A1-20230406-C00952
    1.748
    Figure US20230109104A1-20230406-C00953
    1.749
    Figure US20230109104A1-20230406-C00954
    1.750
    Figure US20230109104A1-20230406-C00955
    1.751
    Figure US20230109104A1-20230406-C00956
    1.752
    Figure US20230109104A1-20230406-C00957
    1.753
    Figure US20230109104A1-20230406-C00958
    1.754
    Figure US20230109104A1-20230406-C00959
    1.755
    Figure US20230109104A1-20230406-C00960
    1.756
    Figure US20230109104A1-20230406-C00961
    1.757
    Figure US20230109104A1-20230406-C00962
    1.758
    Figure US20230109104A1-20230406-C00963
    1.759
    Figure US20230109104A1-20230406-C00964
    1.760
    Figure US20230109104A1-20230406-C00965
    1.761
    Figure US20230109104A1-20230406-C00966
    1.762
    Figure US20230109104A1-20230406-C00967
    1.763
    Figure US20230109104A1-20230406-C00968
    1.764
    Figure US20230109104A1-20230406-C00969
    1.765
    Figure US20230109104A1-20230406-C00970
    1.766
    Figure US20230109104A1-20230406-C00971
    1.767
    Figure US20230109104A1-20230406-C00972
    1.768
    Figure US20230109104A1-20230406-C00973
    1.769
    Figure US20230109104A1-20230406-C00974
    1.770
    Figure US20230109104A1-20230406-C00975
    1.771
    Figure US20230109104A1-20230406-C00976
    1.772
    Figure US20230109104A1-20230406-C00977
    1.773
    Figure US20230109104A1-20230406-C00978
    1.774
    Figure US20230109104A1-20230406-C00979
    1.775
    Figure US20230109104A1-20230406-C00980
    1.776
    Figure US20230109104A1-20230406-C00981
    1.777
    Figure US20230109104A1-20230406-C00982
    1.778
    Figure US20230109104A1-20230406-C00983
    1.779
    Figure US20230109104A1-20230406-C00984
    1.780
    Figure US20230109104A1-20230406-C00985
    1.781
    Figure US20230109104A1-20230406-C00986
    1.782
    Figure US20230109104A1-20230406-C00987
    1.783
    Figure US20230109104A1-20230406-C00988
    1.784
    Figure US20230109104A1-20230406-C00989
    1.785
    Figure US20230109104A1-20230406-C00990
    1.786
    Figure US20230109104A1-20230406-C00991
    1.787
    Figure US20230109104A1-20230406-C00992
    1.788
    Figure US20230109104A1-20230406-C00993
    1.789
    Figure US20230109104A1-20230406-C00994
    1.790
    Figure US20230109104A1-20230406-C00995
    1.791
    Figure US20230109104A1-20230406-C00996
    1.792
    Figure US20230109104A1-20230406-C00997
    1.793
    Figure US20230109104A1-20230406-C00998
    1.794
    Figure US20230109104A1-20230406-C00999
    1.795
    Figure US20230109104A1-20230406-C01000
    1.796
    Figure US20230109104A1-20230406-C01001
    1.797
    Figure US20230109104A1-20230406-C01002
    1.798
    Figure US20230109104A1-20230406-C01003
    1.799
    Figure US20230109104A1-20230406-C01004
    1.800
    Figure US20230109104A1-20230406-C01005
    1.801
    Figure US20230109104A1-20230406-C01006
    1.802
    Figure US20230109104A1-20230406-C01007
    1.803
    Figure US20230109104A1-20230406-C01008
    1.804
    Figure US20230109104A1-20230406-C01009
    1.805
    Figure US20230109104A1-20230406-C01010
    1.806
    Figure US20230109104A1-20230406-C01011
    1.807
    Figure US20230109104A1-20230406-C01012
    1.808
    Figure US20230109104A1-20230406-C01013
    1.809
    Figure US20230109104A1-20230406-C01014
    1.810
    Figure US20230109104A1-20230406-C01015
    1.811
    Figure US20230109104A1-20230406-C01016
    1.812
    Figure US20230109104A1-20230406-C01017
    1.813
    Figure US20230109104A1-20230406-C01018
    1.814
    Figure US20230109104A1-20230406-C01019
    1.815
    Figure US20230109104A1-20230406-C01020
    1.816
    Figure US20230109104A1-20230406-C01021
    1.817
    Figure US20230109104A1-20230406-C01022
    1.818
    Figure US20230109104A1-20230406-C01023
    1.819
    Figure US20230109104A1-20230406-C01024
    1.820
    Figure US20230109104A1-20230406-C01025
    1.821
    Figure US20230109104A1-20230406-C01026
    1.822
    Figure US20230109104A1-20230406-C01027
    1.823
    Figure US20230109104A1-20230406-C01028
    1.824
    Figure US20230109104A1-20230406-C01029
    1.825
    Figure US20230109104A1-20230406-C01030
    1.826
    Figure US20230109104A1-20230406-C01031
    1.827
    Figure US20230109104A1-20230406-C01032
    1.828
    Figure US20230109104A1-20230406-C01033
    1.829
    Figure US20230109104A1-20230406-C01034
    1.830
    Figure US20230109104A1-20230406-C01035
    1.831
    Figure US20230109104A1-20230406-C01036
    1.832
    Figure US20230109104A1-20230406-C01037
    1.833
    Figure US20230109104A1-20230406-C01038
    1.834
    Figure US20230109104A1-20230406-C01039
    1.835
    Figure US20230109104A1-20230406-C01040
    1.836
    Figure US20230109104A1-20230406-C01041
    1.837
    Figure US20230109104A1-20230406-C01042
    1.838
    Figure US20230109104A1-20230406-C01043
    1.839
    Figure US20230109104A1-20230406-C01044
    1.840
    Figure US20230109104A1-20230406-C01045
    1.841
    Figure US20230109104A1-20230406-C01046
    1.842
    Figure US20230109104A1-20230406-C01047
    1.843
    Figure US20230109104A1-20230406-C01048
    1.844
    Figure US20230109104A1-20230406-C01049
    1.845
    Figure US20230109104A1-20230406-C01050
    1.846
    Figure US20230109104A1-20230406-C01051
    1.847
    Figure US20230109104A1-20230406-C01052
    1.848
    Figure US20230109104A1-20230406-C01053
    1.849
    Figure US20230109104A1-20230406-C01054
    1.850
    Figure US20230109104A1-20230406-C01055
    1.851
    Figure US20230109104A1-20230406-C01056
    1.852
    Figure US20230109104A1-20230406-C01057
    1.853
    Figure US20230109104A1-20230406-C01058
    1.854
    Figure US20230109104A1-20230406-C01059
    1.855
    Figure US20230109104A1-20230406-C01060
    1.856
    Figure US20230109104A1-20230406-C01061
    1.857
    Figure US20230109104A1-20230406-C01062
    1.858
    Figure US20230109104A1-20230406-C01063
    1.859
    Figure US20230109104A1-20230406-C01064
    1.860
    Figure US20230109104A1-20230406-C01065
    1.861
    Figure US20230109104A1-20230406-C01066
    1.862
    Figure US20230109104A1-20230406-C01067
    1.863
    Figure US20230109104A1-20230406-C01068
    1.864
    Figure US20230109104A1-20230406-C01069
    1.865
    Figure US20230109104A1-20230406-C01070
    1.866
    Figure US20230109104A1-20230406-C01071
    1.867
    Figure US20230109104A1-20230406-C01072
    1.868
    Figure US20230109104A1-20230406-C01073
    1.869
    Figure US20230109104A1-20230406-C01074
    1.870
    Figure US20230109104A1-20230406-C01075
    1.871
    Figure US20230109104A1-20230406-C01076
    1.872
    Figure US20230109104A1-20230406-C01077
    1.873
    Figure US20230109104A1-20230406-C01078
    1.874
    Figure US20230109104A1-20230406-C01079
    1.875
    Figure US20230109104A1-20230406-C01080
    1.876
    Figure US20230109104A1-20230406-C01081
    1.877
    Figure US20230109104A1-20230406-C01082
    1.878
    Figure US20230109104A1-20230406-C01083
    1.879
    Figure US20230109104A1-20230406-C01084
    1.880
    Figure US20230109104A1-20230406-C01085
    1.881
    Figure US20230109104A1-20230406-C01086
    1.882
    Figure US20230109104A1-20230406-C01087
    1.883
    Figure US20230109104A1-20230406-C01088
    1.884
    Figure US20230109104A1-20230406-C01089
    1.885
    Figure US20230109104A1-20230406-C01090
    1.886
    Figure US20230109104A1-20230406-C01091
    1.887
    Figure US20230109104A1-20230406-C01092
    1.888
    Figure US20230109104A1-20230406-C01093
    1.889
    Figure US20230109104A1-20230406-C01094
    1.890
    Figure US20230109104A1-20230406-C01095
    1.891
    Figure US20230109104A1-20230406-C01096
    1.892
    Figure US20230109104A1-20230406-C01097
    1.893
    Figure US20230109104A1-20230406-C01098
    1.894
    Figure US20230109104A1-20230406-C01099
    1.895
    Figure US20230109104A1-20230406-C01100
    1.896
    Figure US20230109104A1-20230406-C01101
    1.897
    Figure US20230109104A1-20230406-C01102
    1.898
    Figure US20230109104A1-20230406-C01103
    1.899
    Figure US20230109104A1-20230406-C01104
    1.900
    Figure US20230109104A1-20230406-C01105
    1.901
    Figure US20230109104A1-20230406-C01106
    1.902
    Figure US20230109104A1-20230406-C01107
    1.903
    Figure US20230109104A1-20230406-C01108
    1.904
    Figure US20230109104A1-20230406-C01109
    1.905
    Figure US20230109104A1-20230406-C01110
    1.906
    Figure US20230109104A1-20230406-C01111
    1.907
    Figure US20230109104A1-20230406-C01112
    1.908
    Figure US20230109104A1-20230406-C01113
    1.909
    Figure US20230109104A1-20230406-C01114
    1.910
    Figure US20230109104A1-20230406-C01115
    1.911
    Figure US20230109104A1-20230406-C01116
    1.912
    Figure US20230109104A1-20230406-C01117
    1.913
    Figure US20230109104A1-20230406-C01118
    1.914
    Figure US20230109104A1-20230406-C01119
    1.915
    Figure US20230109104A1-20230406-C01120
    1.916
    Figure US20230109104A1-20230406-C01121
    1.917
    Figure US20230109104A1-20230406-C01122
    1.918
    Figure US20230109104A1-20230406-C01123
    1.919
    Figure US20230109104A1-20230406-C01124
    1.920
    Figure US20230109104A1-20230406-C01125
    1.921
    Figure US20230109104A1-20230406-C01126
    1.922
    Figure US20230109104A1-20230406-C01127
    1.923
    Figure US20230109104A1-20230406-C01128
    1.924
    Figure US20230109104A1-20230406-C01129
    1.925
    Figure US20230109104A1-20230406-C01130
    1.926
    Figure US20230109104A1-20230406-C01131
    1.927
    Figure US20230109104A1-20230406-C01132
    1.928
    Figure US20230109104A1-20230406-C01133
    1.929
    Figure US20230109104A1-20230406-C01134
    1.930
    Figure US20230109104A1-20230406-C01135
    1.931
    Figure US20230109104A1-20230406-C01136
    1.932
    Figure US20230109104A1-20230406-C01137
    1.933
    Figure US20230109104A1-20230406-C01138
    1.934
    Figure US20230109104A1-20230406-C01139
    1.935
    Figure US20230109104A1-20230406-C01140
    1.936
    Figure US20230109104A1-20230406-C01141
    1.937
    Figure US20230109104A1-20230406-C01142
    1.938
    Figure US20230109104A1-20230406-C01143
    1.939
    Figure US20230109104A1-20230406-C01144
    1.940
    Figure US20230109104A1-20230406-C01145
    1.941
    Figure US20230109104A1-20230406-C01146
    1.942
    Figure US20230109104A1-20230406-C01147
    1.943
    Figure US20230109104A1-20230406-C01148
    1.944
    Figure US20230109104A1-20230406-C01149
    1.945
    Figure US20230109104A1-20230406-C01150
    1.946
    Figure US20230109104A1-20230406-C01151
    1.947
    Figure US20230109104A1-20230406-C01152
    1.948
    Figure US20230109104A1-20230406-C01153
    1.949
    Figure US20230109104A1-20230406-C01154
    1.950
    Figure US20230109104A1-20230406-C01155
    1.951
    Figure US20230109104A1-20230406-C01156
    1.952
    Figure US20230109104A1-20230406-C01157
    1.953
    Figure US20230109104A1-20230406-C01158
    1.954
    Figure US20230109104A1-20230406-C01159
    1.955
    Figure US20230109104A1-20230406-C01160
    1.956
    Figure US20230109104A1-20230406-C01161
    1.957
    Figure US20230109104A1-20230406-C01162
    1.958
    Figure US20230109104A1-20230406-C01163
    1.959
    Figure US20230109104A1-20230406-C01164
    1.960
    Figure US20230109104A1-20230406-C01165
    1.961
    Figure US20230109104A1-20230406-C01166
    1.962
    Figure US20230109104A1-20230406-C01167
    1.963
    Figure US20230109104A1-20230406-C01168
    1.964
    Figure US20230109104A1-20230406-C01169
    1.965
    Figure US20230109104A1-20230406-C01170
    1.966
    Figure US20230109104A1-20230406-C01171
    1.967
    Figure US20230109104A1-20230406-C01172
    1.968
    Figure US20230109104A1-20230406-C01173
    1.969
    Figure US20230109104A1-20230406-C01174
    1.970
    Figure US20230109104A1-20230406-C01175
    1.971
    Figure US20230109104A1-20230406-C01176
    1.972
    Figure US20230109104A1-20230406-C01177
    1.973
    Figure US20230109104A1-20230406-C01178
    1.974
    Figure US20230109104A1-20230406-C01179
    1.975
    Figure US20230109104A1-20230406-C01180
    1.976
    Figure US20230109104A1-20230406-C01181
    1.977
    Figure US20230109104A1-20230406-C01182
    1.978
    Figure US20230109104A1-20230406-C01183
    1.979
    Figure US20230109104A1-20230406-C01184
    1.980
    Figure US20230109104A1-20230406-C01185
    1.981
    Figure US20230109104A1-20230406-C01186
    1.982
    Figure US20230109104A1-20230406-C01187
    1.983
    Figure US20230109104A1-20230406-C01188
    1.984
    Figure US20230109104A1-20230406-C01189
    1.985
    Figure US20230109104A1-20230406-C01190
    1.986
    Figure US20230109104A1-20230406-C01191
    1.987
    Figure US20230109104A1-20230406-C01192
    1.988
    Figure US20230109104A1-20230406-C01193
    1.989
    Figure US20230109104A1-20230406-C01194
    1.990
    Figure US20230109104A1-20230406-C01195
    1.991
    Figure US20230109104A1-20230406-C01196
    1.992
    Figure US20230109104A1-20230406-C01197
    1.993
    Figure US20230109104A1-20230406-C01198
    1.994
    Figure US20230109104A1-20230406-C01199
    1.995
    Figure US20230109104A1-20230406-C01200
    1.996
    Figure US20230109104A1-20230406-C01201
    1.997
    Figure US20230109104A1-20230406-C01202
    1.998
    Figure US20230109104A1-20230406-C01203
    1.999
    Figure US20230109104A1-20230406-C01204
    2.000
    Figure US20230109104A1-20230406-C01205
    2.001
    Figure US20230109104A1-20230406-C01206
    2.002
    Figure US20230109104A1-20230406-C01207
    2.003
    Figure US20230109104A1-20230406-C01208
    2.004
    Figure US20230109104A1-20230406-C01209
    2.005
    Figure US20230109104A1-20230406-C01210
    2.006
    Figure US20230109104A1-20230406-C01211
    2.007
    Figure US20230109104A1-20230406-C01212
    2.008
    Figure US20230109104A1-20230406-C01213
    2.009
    Figure US20230109104A1-20230406-C01214
    2.010
    Figure US20230109104A1-20230406-C01215
    2.011
    Figure US20230109104A1-20230406-C01216
    2.012
    Figure US20230109104A1-20230406-C01217
    2.013
    Figure US20230109104A1-20230406-C01218
    2.014
    Figure US20230109104A1-20230406-C01219
    2.015
    Figure US20230109104A1-20230406-C01220
    2.016
    Figure US20230109104A1-20230406-C01221
    2.017
    Figure US20230109104A1-20230406-C01222
    2.018
    Figure US20230109104A1-20230406-C01223
    2.019
    Figure US20230109104A1-20230406-C01224
    2.020
    Figure US20230109104A1-20230406-C01225
    2.021
    Figure US20230109104A1-20230406-C01226
    2.022
    Figure US20230109104A1-20230406-C01227
    2.023
    Figure US20230109104A1-20230406-C01228
    2.024
    Figure US20230109104A1-20230406-C01229
    2.025
    Figure US20230109104A1-20230406-C01230
    2.026
    Figure US20230109104A1-20230406-C01231
    2.027
    Figure US20230109104A1-20230406-C01232
    2.028
    Figure US20230109104A1-20230406-C01233
    2.029
    Figure US20230109104A1-20230406-C01234
    2.030
    Figure US20230109104A1-20230406-C01235
    2.031
    Figure US20230109104A1-20230406-C01236
    2.032
    Figure US20230109104A1-20230406-C01237
    2.033
    Figure US20230109104A1-20230406-C01238
    2.034
    Figure US20230109104A1-20230406-C01239
    2.035
    Figure US20230109104A1-20230406-C01240
    2.036
    Figure US20230109104A1-20230406-C01241
    2.037
    Figure US20230109104A1-20230406-C01242
    2.038
    Figure US20230109104A1-20230406-C01243
    2.039
    Figure US20230109104A1-20230406-C01244
    2.040
    Figure US20230109104A1-20230406-C01245
    2.041
    Figure US20230109104A1-20230406-C01246
    2.042
    Figure US20230109104A1-20230406-C01247
    2.043
    Figure US20230109104A1-20230406-C01248
    2.044
    Figure US20230109104A1-20230406-C01249
    2.045
    Figure US20230109104A1-20230406-C01250
    2.046
    Figure US20230109104A1-20230406-C01251
    2.047
    Figure US20230109104A1-20230406-C01252
    2.048
    Figure US20230109104A1-20230406-C01253
    2.049
    Figure US20230109104A1-20230406-C01254
    2.050
    Figure US20230109104A1-20230406-C01255
    2.051
    Figure US20230109104A1-20230406-C01256
    2.052
    Figure US20230109104A1-20230406-C01257
    2.053
    Figure US20230109104A1-20230406-C01258
    2.054
    Figure US20230109104A1-20230406-C01259
    2.055
    Figure US20230109104A1-20230406-C01260
    2.056
    Figure US20230109104A1-20230406-C01261
    2.057
    Figure US20230109104A1-20230406-C01262
    2.058
    Figure US20230109104A1-20230406-C01263
    2.059
    Figure US20230109104A1-20230406-C01264
    2.060
    Figure US20230109104A1-20230406-C01265
    2.061
    Figure US20230109104A1-20230406-C01266
    2.062
    Figure US20230109104A1-20230406-C01267
    2.063
    Figure US20230109104A1-20230406-C01268
    2.064
    Figure US20230109104A1-20230406-C01269
    2.065
    Figure US20230109104A1-20230406-C01270
    2.066
    Figure US20230109104A1-20230406-C01271
    2.067
    Figure US20230109104A1-20230406-C01272
    2.068
    Figure US20230109104A1-20230406-C01273
    2.069
    Figure US20230109104A1-20230406-C01274
    2.070
    Figure US20230109104A1-20230406-C01275
    2.071
    Figure US20230109104A1-20230406-C01276
    2.072
    Figure US20230109104A1-20230406-C01277
    2.073
    Figure US20230109104A1-20230406-C01278
    2.074
    Figure US20230109104A1-20230406-C01279
    2.075
    Figure US20230109104A1-20230406-C01280
    2.076
    Figure US20230109104A1-20230406-C01281
    2.077
    Figure US20230109104A1-20230406-C01282
    2.078
    Figure US20230109104A1-20230406-C01283
    2.079
    Figure US20230109104A1-20230406-C01284
    2.080
    Figure US20230109104A1-20230406-C01285
    2.081
    Figure US20230109104A1-20230406-C01286
    2.082
    Figure US20230109104A1-20230406-C01287
    2.083
    Figure US20230109104A1-20230406-C01288
    2.084
    Figure US20230109104A1-20230406-C01289
    2.085
    Figure US20230109104A1-20230406-C01290
    2.086
    Figure US20230109104A1-20230406-C01291
    2.087
    Figure US20230109104A1-20230406-C01292
    2.088
    Figure US20230109104A1-20230406-C01293
    2.089
    Figure US20230109104A1-20230406-C01294
    2.090
    Figure US20230109104A1-20230406-C01295
    2.091
    Figure US20230109104A1-20230406-C01296
    2.092
    Figure US20230109104A1-20230406-C01297
    2.093
    Figure US20230109104A1-20230406-C01298
    2.094
    Figure US20230109104A1-20230406-C01299
    2.095
    Figure US20230109104A1-20230406-C01300
    2.096
    Figure US20230109104A1-20230406-C01301
    2.097
    Figure US20230109104A1-20230406-C01302
    2.098
    Figure US20230109104A1-20230406-C01303
    2.099
    Figure US20230109104A1-20230406-C01304
    2.100
    Figure US20230109104A1-20230406-C01305
    2.101
    Figure US20230109104A1-20230406-C01306
    2.102
    Figure US20230109104A1-20230406-C01307
    2.103
    Figure US20230109104A1-20230406-C01308
    2.104
    Figure US20230109104A1-20230406-C01309
    2.105
    Figure US20230109104A1-20230406-C01310
    2.106
    Figure US20230109104A1-20230406-C01311
    2.107
    Figure US20230109104A1-20230406-C01312
    2.108
    Figure US20230109104A1-20230406-C01313
    2.109
    Figure US20230109104A1-20230406-C01314
    2.110
    Figure US20230109104A1-20230406-C01315
    2.111
    Figure US20230109104A1-20230406-C01316
    2.112
    Figure US20230109104A1-20230406-C01317
    2.113
    Figure US20230109104A1-20230406-C01318
    2.114
    Figure US20230109104A1-20230406-C01319
    2.115
    Figure US20230109104A1-20230406-C01320
    2.116
    Figure US20230109104A1-20230406-C01321
    2.117
    Figure US20230109104A1-20230406-C01322
    2.118
    Figure US20230109104A1-20230406-C01323
    2.119
    Figure US20230109104A1-20230406-C01324
    2.120
    Figure US20230109104A1-20230406-C01325
    2.121
    Figure US20230109104A1-20230406-C01326
    2.122
    Figure US20230109104A1-20230406-C01327
    2.123
    Figure US20230109104A1-20230406-C01328
    2.124
    Figure US20230109104A1-20230406-C01329
    2.125
    Figure US20230109104A1-20230406-C01330
    2.126
    Figure US20230109104A1-20230406-C01331
    2.127
    Figure US20230109104A1-20230406-C01332
    2.128
    Figure US20230109104A1-20230406-C01333
    2.129
    Figure US20230109104A1-20230406-C01334
    2.130
    Figure US20230109104A1-20230406-C01335
    2.131
    Figure US20230109104A1-20230406-C01336
    2.132
    Figure US20230109104A1-20230406-C01337
    2.133
    Figure US20230109104A1-20230406-C01338
    2.134
    Figure US20230109104A1-20230406-C01339
    2.135
    Figure US20230109104A1-20230406-C01340
    2.136
    Figure US20230109104A1-20230406-C01341
    2.137
    Figure US20230109104A1-20230406-C01342
    2.138
    Figure US20230109104A1-20230406-C01343
    2.139
    Figure US20230109104A1-20230406-C01344
    2.140
    Figure US20230109104A1-20230406-C01345
    2.141
    Figure US20230109104A1-20230406-C01346
    2.142
    Figure US20230109104A1-20230406-C01347
    2.143
    Figure US20230109104A1-20230406-C01348
    2.144
    Figure US20230109104A1-20230406-C01349
    2.145
    Figure US20230109104A1-20230406-C01350
    2.146
    Figure US20230109104A1-20230406-C01351
    2.147
    Figure US20230109104A1-20230406-C01352
    2.148
    Figure US20230109104A1-20230406-C01353
    2.149
    Figure US20230109104A1-20230406-C01354
    2.150
    Figure US20230109104A1-20230406-C01355
    2151
    Figure US20230109104A1-20230406-C01356
    2.152
    Figure US20230109104A1-20230406-C01357
    2.153
    Figure US20230109104A1-20230406-C01358
    2.154
    Figure US20230109104A1-20230406-C01359
    2.155
    Figure US20230109104A1-20230406-C01360
    2.156
    Figure US20230109104A1-20230406-C01361
    2.157
    Figure US20230109104A1-20230406-C01362
    2.158
    Figure US20230109104A1-20230406-C01363
    2.159
    Figure US20230109104A1-20230406-C01364
    2.160
    Figure US20230109104A1-20230406-C01365
    2.161
    Figure US20230109104A1-20230406-C01366
    2.162
    Figure US20230109104A1-20230406-C01367
    2.163
    Figure US20230109104A1-20230406-C01368
    2.164
    Figure US20230109104A1-20230406-C01369
    2.165
    Figure US20230109104A1-20230406-C01370
    2.166
    Figure US20230109104A1-20230406-C01371
    2.167
    Figure US20230109104A1-20230406-C01372
    2.168
    Figure US20230109104A1-20230406-C01373
    2.169
    Figure US20230109104A1-20230406-C01374
    2.170
    Figure US20230109104A1-20230406-C01375
    2.171
    Figure US20230109104A1-20230406-C01376
    2.172
    Figure US20230109104A1-20230406-C01377
    2.173
    Figure US20230109104A1-20230406-C01378
    2.174
    Figure US20230109104A1-20230406-C01379
    2.175
    Figure US20230109104A1-20230406-C01380
    2.176
    Figure US20230109104A1-20230406-C01381
    2.177
    Figure US20230109104A1-20230406-C01382
    2.178
    Figure US20230109104A1-20230406-C01383
    2.179
    Figure US20230109104A1-20230406-C01384
    2.180
    Figure US20230109104A1-20230406-C01385
    2.181
    Figure US20230109104A1-20230406-C01386
    2.182
    Figure US20230109104A1-20230406-C01387
    2.183
    Figure US20230109104A1-20230406-C01388
    2.184
    Figure US20230109104A1-20230406-C01389
    2.185
    Figure US20230109104A1-20230406-C01390
    2.186
    Figure US20230109104A1-20230406-C01391
    2.187
    Figure US20230109104A1-20230406-C01392
    2.188
    Figure US20230109104A1-20230406-C01393
    2.189
    Figure US20230109104A1-20230406-C01394
    2.190
    Figure US20230109104A1-20230406-C01395
    2.191
    Figure US20230109104A1-20230406-C01396
    2.192
    Figure US20230109104A1-20230406-C01397
    2.193
    Figure US20230109104A1-20230406-C01398
    2.194
    Figure US20230109104A1-20230406-C01399
    2.195
    Figure US20230109104A1-20230406-C01400
    2.196
    Figure US20230109104A1-20230406-C01401
    2.197
    Figure US20230109104A1-20230406-C01402
    2.198
    Figure US20230109104A1-20230406-C01403
    2.199
    Figure US20230109104A1-20230406-C01404
    2.200
    Figure US20230109104A1-20230406-C01405
    2.201
    Figure US20230109104A1-20230406-C01406
    2.202
    Figure US20230109104A1-20230406-C01407
    2.203
    Figure US20230109104A1-20230406-C01408
    2.204
    Figure US20230109104A1-20230406-C01409
    2.205
    Figure US20230109104A1-20230406-C01410
    2.206
    Figure US20230109104A1-20230406-C01411
    2.207
    Figure US20230109104A1-20230406-C01412
    2.208
    Figure US20230109104A1-20230406-C01413
    2.209
    Figure US20230109104A1-20230406-C01414
    2.210
    Figure US20230109104A1-20230406-C01415
    2.211
    Figure US20230109104A1-20230406-C01416
    2.212
    Figure US20230109104A1-20230406-C01417
    2.213
    Figure US20230109104A1-20230406-C01418
    2.214
    Figure US20230109104A1-20230406-C01419
    2.215
    Figure US20230109104A1-20230406-C01420
    2.216
    Figure US20230109104A1-20230406-C01421
    2.217
    Figure US20230109104A1-20230406-C01422
    2.218
    Figure US20230109104A1-20230406-C01423
    2.219
    Figure US20230109104A1-20230406-C01424
    2.220
    Figure US20230109104A1-20230406-C01425
    2.221
    Figure US20230109104A1-20230406-C01426
    2.222
    Figure US20230109104A1-20230406-C01427
    2.223
    Figure US20230109104A1-20230406-C01428
    2.224
    Figure US20230109104A1-20230406-C01429
    2.225
    Figure US20230109104A1-20230406-C01430
    2.226
    Figure US20230109104A1-20230406-C01431
    2.227
    Figure US20230109104A1-20230406-C01432
    2.228
    Figure US20230109104A1-20230406-C01433
    2.229
    Figure US20230109104A1-20230406-C01434
    2.230
    Figure US20230109104A1-20230406-C01435
    2.231
    Figure US20230109104A1-20230406-C01436
    2.232
    Figure US20230109104A1-20230406-C01437
    2.233
    Figure US20230109104A1-20230406-C01438
    2.234
    Figure US20230109104A1-20230406-C01439
    2.235
    Figure US20230109104A1-20230406-C01440
    2.236
    Figure US20230109104A1-20230406-C01441
    2.237
    Figure US20230109104A1-20230406-C01442
    2.238
    Figure US20230109104A1-20230406-C01443
    2.239
    Figure US20230109104A1-20230406-C01444
    2.240
    Figure US20230109104A1-20230406-C01445
    2.241
    Figure US20230109104A1-20230406-C01446
    2.242
    Figure US20230109104A1-20230406-C01447
    2.243
    Figure US20230109104A1-20230406-C01448
    2.244
    Figure US20230109104A1-20230406-C01449
    2.245
    Figure US20230109104A1-20230406-C01450
    2.246
    Figure US20230109104A1-20230406-C01451
    2.247
    Figure US20230109104A1-20230406-C01452
    2.248
    Figure US20230109104A1-20230406-C01453
    2.249
    Figure US20230109104A1-20230406-C01454
    2.250
    Figure US20230109104A1-20230406-C01455
    2.251
    Figure US20230109104A1-20230406-C01456
    2.252
    Figure US20230109104A1-20230406-C01457
    2.253
    Figure US20230109104A1-20230406-C01458
    2.254
    Figure US20230109104A1-20230406-C01459
    2.255
    Figure US20230109104A1-20230406-C01460
    2.256
    Figure US20230109104A1-20230406-C01461
    2.257
    Figure US20230109104A1-20230406-C01462
    2.258
    Figure US20230109104A1-20230406-C01463
    2.259
    Figure US20230109104A1-20230406-C01464
    2.260
    Figure US20230109104A1-20230406-C01465
    2.261
    Figure US20230109104A1-20230406-C01466
    2.262
    Figure US20230109104A1-20230406-C01467
    2.263
    Figure US20230109104A1-20230406-C01468
    2.264
    Figure US20230109104A1-20230406-C01469
    2.265
    Figure US20230109104A1-20230406-C01470
    2.266
    Figure US20230109104A1-20230406-C01471
    2.267
    Figure US20230109104A1-20230406-C01472
    2.268
    Figure US20230109104A1-20230406-C01473
    2.269
    Figure US20230109104A1-20230406-C01474
    2.270
    Figure US20230109104A1-20230406-C01475
    2.271
    Figure US20230109104A1-20230406-C01476
    2.272
    Figure US20230109104A1-20230406-C01477
    2.273
    Figure US20230109104A1-20230406-C01478
    2.274
    Figure US20230109104A1-20230406-C01479
    2.275
    Figure US20230109104A1-20230406-C01480
    2.276
    Figure US20230109104A1-20230406-C01481
    2.277
    Figure US20230109104A1-20230406-C01482
    2.278
    Figure US20230109104A1-20230406-C01483
    2.279
    Figure US20230109104A1-20230406-C01484
    2.280
    Figure US20230109104A1-20230406-C01485
    2.281
    Figure US20230109104A1-20230406-C01486
    2.282
    Figure US20230109104A1-20230406-C01487
    2.283
    Figure US20230109104A1-20230406-C01488
    2.284
    Figure US20230109104A1-20230406-C01489
    2.285
    Figure US20230109104A1-20230406-C01490
    2.286
    Figure US20230109104A1-20230406-C01491
    2.287
    Figure US20230109104A1-20230406-C01492
    2.288
    Figure US20230109104A1-20230406-C01493
    2.289
    Figure US20230109104A1-20230406-C01494
    2.290
    Figure US20230109104A1-20230406-C01495
    2.291
    Figure US20230109104A1-20230406-C01496
    2.292
    Figure US20230109104A1-20230406-C01497
    2.293
    Figure US20230109104A1-20230406-C01498
    2.294
    Figure US20230109104A1-20230406-C01499
    2.295
    Figure US20230109104A1-20230406-C01500
    2.296
    Figure US20230109104A1-20230406-C01501
    2.297
    Figure US20230109104A1-20230406-C01502
    2.298
    Figure US20230109104A1-20230406-C01503
    2.299
    Figure US20230109104A1-20230406-C01504
    2.300
    Figure US20230109104A1-20230406-C01505
    2.301
    Figure US20230109104A1-20230406-C01506
    2.302
    Figure US20230109104A1-20230406-C01507
    2.303
    Figure US20230109104A1-20230406-C01508
    2.304
    Figure US20230109104A1-20230406-C01509
    2.305
    Figure US20230109104A1-20230406-C01510
    2.306
    Figure US20230109104A1-20230406-C01511
    2.307
    Figure US20230109104A1-20230406-C01512
    2.308
    Figure US20230109104A1-20230406-C01513
    2.309
    Figure US20230109104A1-20230406-C01514
    2.310
    Figure US20230109104A1-20230406-C01515
    2.311
    Figure US20230109104A1-20230406-C01516
    2.312
    Figure US20230109104A1-20230406-C01517
    2.313
    Figure US20230109104A1-20230406-C01518
    2.314
    Figure US20230109104A1-20230406-C01519
    2.315
    Figure US20230109104A1-20230406-C01520
    2.316
    Figure US20230109104A1-20230406-C01521
    2.317
    Figure US20230109104A1-20230406-C01522
    2.318
    Figure US20230109104A1-20230406-C01523
    2.319
    Figure US20230109104A1-20230406-C01524
    2.320
    Figure US20230109104A1-20230406-C01525
    2.321
    Figure US20230109104A1-20230406-C01526
    2.322
    Figure US20230109104A1-20230406-C01527
    2.323
    Figure US20230109104A1-20230406-C01528
    2.324
    Figure US20230109104A1-20230406-C01529
    2.325
    Figure US20230109104A1-20230406-C01530
    2.326
    Figure US20230109104A1-20230406-C01531
    2.327
    Figure US20230109104A1-20230406-C01532
    2.328
    Figure US20230109104A1-20230406-C01533
    2.329
    Figure US20230109104A1-20230406-C01534
    2.330
    Figure US20230109104A1-20230406-C01535
    2.331
    Figure US20230109104A1-20230406-C01536
    2.332
    Figure US20230109104A1-20230406-C01537
    2.333
    Figure US20230109104A1-20230406-C01538
    2.334
    Figure US20230109104A1-20230406-C01539
    2.335
    Figure US20230109104A1-20230406-C01540
    2.336
    Figure US20230109104A1-20230406-C01541
    2.337
    Figure US20230109104A1-20230406-C01542
    2.338
    Figure US20230109104A1-20230406-C01543
    2.339
    Figure US20230109104A1-20230406-C01544
    2.340
    Figure US20230109104A1-20230406-C01545
    2.341
    Figure US20230109104A1-20230406-C01546
    2.342
    Figure US20230109104A1-20230406-C01547
    2.343
    Figure US20230109104A1-20230406-C01548
    2.344
    Figure US20230109104A1-20230406-C01549
    2.345
    Figure US20230109104A1-20230406-C01550
    2.346
    Figure US20230109104A1-20230406-C01551
    2.347
    Figure US20230109104A1-20230406-C01552
    2.348
    Figure US20230109104A1-20230406-C01553
    2.349
    Figure US20230109104A1-20230406-C01554
    2.350
    Figure US20230109104A1-20230406-C01555
    2.351
    Figure US20230109104A1-20230406-C01556
    2.352
    Figure US20230109104A1-20230406-C01557
    2.353
    Figure US20230109104A1-20230406-C01558
    2.354
    Figure US20230109104A1-20230406-C01559
    2.355
    Figure US20230109104A1-20230406-C01560
    2.356
    Figure US20230109104A1-20230406-C01561
    2.357
    Figure US20230109104A1-20230406-C01562
    2.358
    Figure US20230109104A1-20230406-C01563
    2.359
    Figure US20230109104A1-20230406-C01564
    2.360
    Figure US20230109104A1-20230406-C01565
    2.361
    Figure US20230109104A1-20230406-C01566
    2.362
    Figure US20230109104A1-20230406-C01567
    2.363
    Figure US20230109104A1-20230406-C01568
    2.364
    Figure US20230109104A1-20230406-C01569
    2.365
    Figure US20230109104A1-20230406-C01570
    2.366
    Figure US20230109104A1-20230406-C01571
    2.367
    Figure US20230109104A1-20230406-C01572
    2.368
    Figure US20230109104A1-20230406-C01573
    2.369
    Figure US20230109104A1-20230406-C01574
    2.370
    Figure US20230109104A1-20230406-C01575
    2.371
    Figure US20230109104A1-20230406-C01576
    2.372
    Figure US20230109104A1-20230406-C01577
    2.373
    Figure US20230109104A1-20230406-C01578
    2.374
    Figure US20230109104A1-20230406-C01579
    2.375
    Figure US20230109104A1-20230406-C01580
    2.376
    Figure US20230109104A1-20230406-C01581
    2.377
    Figure US20230109104A1-20230406-C01582
    2.378
    Figure US20230109104A1-20230406-C01583
    2.379
    Figure US20230109104A1-20230406-C01584
    2.380
    Figure US20230109104A1-20230406-C01585
    2.381
    Figure US20230109104A1-20230406-C01586
    2.382
    Figure US20230109104A1-20230406-C01587
    2.383
    Figure US20230109104A1-20230406-C01588
    2.384
    Figure US20230109104A1-20230406-C01589
    2.385
    Figure US20230109104A1-20230406-C01590
    2.386
    Figure US20230109104A1-20230406-C01591
    2.387
    Figure US20230109104A1-20230406-C01592
    2.388
    Figure US20230109104A1-20230406-C01593
    2.389
    Figure US20230109104A1-20230406-C01594
    2.390
    Figure US20230109104A1-20230406-C01595
    2.391
    Figure US20230109104A1-20230406-C01596
    2.392
    Figure US20230109104A1-20230406-C01597
    2.393
    Figure US20230109104A1-20230406-C01598
    2.394
    Figure US20230109104A1-20230406-C01599
    2.395
    Figure US20230109104A1-20230406-C01600
    2.396
    Figure US20230109104A1-20230406-C01601
    2.397
    Figure US20230109104A1-20230406-C01602
    2.398
    Figure US20230109104A1-20230406-C01603
    2.399
    Figure US20230109104A1-20230406-C01604
    2.400
    Figure US20230109104A1-20230406-C01605
    2.401
    Figure US20230109104A1-20230406-C01606
    2.402
    Figure US20230109104A1-20230406-C01607
    2.403
    Figure US20230109104A1-20230406-C01608
    2.404
    Figure US20230109104A1-20230406-C01609
    2.405
    Figure US20230109104A1-20230406-C01610
    2.406
    Figure US20230109104A1-20230406-C01611
    2.407
    Figure US20230109104A1-20230406-C01612
    2.408
    Figure US20230109104A1-20230406-C01613
    2.409
    Figure US20230109104A1-20230406-C01614
    2.410
    Figure US20230109104A1-20230406-C01615
    2.411
    Figure US20230109104A1-20230406-C01616
    2.412
    Figure US20230109104A1-20230406-C01617
    2.413
    Figure US20230109104A1-20230406-C01618
    2.414
    Figure US20230109104A1-20230406-C01619
    2.415
    Figure US20230109104A1-20230406-C01620
    2.416
    Figure US20230109104A1-20230406-C01621
    2.417
    Figure US20230109104A1-20230406-C01622
    2.418
    Figure US20230109104A1-20230406-C01623
    2.419
    Figure US20230109104A1-20230406-C01624
    2.420
    Figure US20230109104A1-20230406-C01625
    2.421
    Figure US20230109104A1-20230406-C01626
    2.422
    Figure US20230109104A1-20230406-C01627
    2.423
    Figure US20230109104A1-20230406-C01628
    2.424
    Figure US20230109104A1-20230406-C01629
    2.425
    Figure US20230109104A1-20230406-C01630
    2.426
    Figure US20230109104A1-20230406-C01631
    2.427
    Figure US20230109104A1-20230406-C01632
    2.428
    Figure US20230109104A1-20230406-C01633
    2.429
    Figure US20230109104A1-20230406-C01634
    2.430
    Figure US20230109104A1-20230406-C01635
    2.431
    Figure US20230109104A1-20230406-C01636
    2.432
    Figure US20230109104A1-20230406-C01637
    2.433
    Figure US20230109104A1-20230406-C01638
    2.434
    Figure US20230109104A1-20230406-C01639
    2.435
    Figure US20230109104A1-20230406-C01640
    2.436
    Figure US20230109104A1-20230406-C01641
    2.437
    Figure US20230109104A1-20230406-C01642
    2.438
    Figure US20230109104A1-20230406-C01643
    2.439
    Figure US20230109104A1-20230406-C01644
    2.440
    Figure US20230109104A1-20230406-C01645
    2.441
    Figure US20230109104A1-20230406-C01646
    2.442
    Figure US20230109104A1-20230406-C01647
    2.443
    Figure US20230109104A1-20230406-C01648
    2.444
    Figure US20230109104A1-20230406-C01649
    2.445
    Figure US20230109104A1-20230406-C01650
    2.446
    Figure US20230109104A1-20230406-C01651
    2.447
    Figure US20230109104A1-20230406-C01652
    2.448
    Figure US20230109104A1-20230406-C01653
    2.449
    Figure US20230109104A1-20230406-C01654
    2.450
    Figure US20230109104A1-20230406-C01655
    2.451
    Figure US20230109104A1-20230406-C01656
    2.452
    Figure US20230109104A1-20230406-C01657
    2.453
    Figure US20230109104A1-20230406-C01658
    2.454
    Figure US20230109104A1-20230406-C01659
    2.455
    Figure US20230109104A1-20230406-C01660
    2.456
    Figure US20230109104A1-20230406-C01661
    2.457
    Figure US20230109104A1-20230406-C01662
    2.458
    Figure US20230109104A1-20230406-C01663
    2.459
    Figure US20230109104A1-20230406-C01664
    2.460
    Figure US20230109104A1-20230406-C01665
    2.461
    Figure US20230109104A1-20230406-C01666
    2.462
    Figure US20230109104A1-20230406-C01667
    2.463
    Figure US20230109104A1-20230406-C01668
    2.464
    Figure US20230109104A1-20230406-C01669
    2.465
    Figure US20230109104A1-20230406-C01670
    2.466
    Figure US20230109104A1-20230406-C01671
    2.467
    Figure US20230109104A1-20230406-C01672
    2.468
    Figure US20230109104A1-20230406-C01673
    2.469
    Figure US20230109104A1-20230406-C01674
    2.470
    Figure US20230109104A1-20230406-C01675
    2.471
    Figure US20230109104A1-20230406-C01676
    2.472
    Figure US20230109104A1-20230406-C01677
    2.473
    Figure US20230109104A1-20230406-C01678
    2.474
    Figure US20230109104A1-20230406-C01679
    2.475
    Figure US20230109104A1-20230406-C01680
    2.476
    Figure US20230109104A1-20230406-C01681
    2.477
    Figure US20230109104A1-20230406-C01682
    2.478
    Figure US20230109104A1-20230406-C01683
    2.479
    Figure US20230109104A1-20230406-C01684
    2.480
    Figure US20230109104A1-20230406-C01685
    2.481
    Figure US20230109104A1-20230406-C01686
    2.482
    Figure US20230109104A1-20230406-C01687
    2.483
    Figure US20230109104A1-20230406-C01688
    2.484
    Figure US20230109104A1-20230406-C01689
    2.485
    Figure US20230109104A1-20230406-C01690
    2.486
    Figure US20230109104A1-20230406-C01691
    2.487
    Figure US20230109104A1-20230406-C01692
    2.488
    Figure US20230109104A1-20230406-C01693
    2.489
    Figure US20230109104A1-20230406-C01694
    2.490
    Figure US20230109104A1-20230406-C01695
    2.491
    Figure US20230109104A1-20230406-C01696
    2.492
    Figure US20230109104A1-20230406-C01697
    2.493
    Figure US20230109104A1-20230406-C01698
    2.494
    Figure US20230109104A1-20230406-C01699
    2.495
    Figure US20230109104A1-20230406-C01700
    2.496
    Figure US20230109104A1-20230406-C01701
    2.497
    Figure US20230109104A1-20230406-C01702
    2.498
    Figure US20230109104A1-20230406-C01703
    2.499
    Figure US20230109104A1-20230406-C01704
    2.500
    Figure US20230109104A1-20230406-C01705
    2.501
    Figure US20230109104A1-20230406-C01706
    2.502
    Figure US20230109104A1-20230406-C01707
    2.503
    Figure US20230109104A1-20230406-C01708
    2.504
    Figure US20230109104A1-20230406-C01709
    2.505
    Figure US20230109104A1-20230406-C01710
    2.506
    Figure US20230109104A1-20230406-C01711
    2.507
    Figure US20230109104A1-20230406-C01712
    2.508
    Figure US20230109104A1-20230406-C01713
    2.509
    Figure US20230109104A1-20230406-C01714
    2.510
    Figure US20230109104A1-20230406-C01715
    2.511
    Figure US20230109104A1-20230406-C01716
    2.512
    Figure US20230109104A1-20230406-C01717
    2.513
    Figure US20230109104A1-20230406-C01718
    2.514
    Figure US20230109104A1-20230406-C01719
    2.515
    Figure US20230109104A1-20230406-C01720
    2.516
    Figure US20230109104A1-20230406-C01721
    2.517
    Figure US20230109104A1-20230406-C01722
    2.518
    Figure US20230109104A1-20230406-C01723
    2.519
    Figure US20230109104A1-20230406-C01724
    2.520
    Figure US20230109104A1-20230406-C01725
    2.521
    Figure US20230109104A1-20230406-C01726
    2.522
    Figure US20230109104A1-20230406-C01727
    2.523
    Figure US20230109104A1-20230406-C01728
    2.524
    Figure US20230109104A1-20230406-C01729
    2.525
    Figure US20230109104A1-20230406-C01730
    2.526
    Figure US20230109104A1-20230406-C01731
    2.527
    Figure US20230109104A1-20230406-C01732
    2.528
    Figure US20230109104A1-20230406-C01733
    2.529
    Figure US20230109104A1-20230406-C01734
    2.530
    Figure US20230109104A1-20230406-C01735
    2.531
    Figure US20230109104A1-20230406-C01736
    2.532
    Figure US20230109104A1-20230406-C01737
    2.533
    Figure US20230109104A1-20230406-C01738
    2.534
    Figure US20230109104A1-20230406-C01739
    2.535
    Figure US20230109104A1-20230406-C01740
    2.536
    Figure US20230109104A1-20230406-C01741
    2.537
    Figure US20230109104A1-20230406-C01742
    2.538
    Figure US20230109104A1-20230406-C01743
    2.539
    Figure US20230109104A1-20230406-C01744
    2.540
    Figure US20230109104A1-20230406-C01745
    2.541
    Figure US20230109104A1-20230406-C01746
    2.542
    Figure US20230109104A1-20230406-C01747
    2.543
    Figure US20230109104A1-20230406-C01748
    2.544
    Figure US20230109104A1-20230406-C01749
    2.545
    Figure US20230109104A1-20230406-C01750
    2.546
    Figure US20230109104A1-20230406-C01751
    2.547
    Figure US20230109104A1-20230406-C01752
    2.548
    Figure US20230109104A1-20230406-C01753
    2.549
    Figure US20230109104A1-20230406-C01754
    2.550
    Figure US20230109104A1-20230406-C01755
    2.551
    Figure US20230109104A1-20230406-C01756
    2.552
    Figure US20230109104A1-20230406-C01757
    2.553
    Figure US20230109104A1-20230406-C01758
    2.554
    Figure US20230109104A1-20230406-C01759
    2.555
    Figure US20230109104A1-20230406-C01760
    2.556
    Figure US20230109104A1-20230406-C01761
    2.557
    Figure US20230109104A1-20230406-C01762
    2.558
    Figure US20230109104A1-20230406-C01763
    2.559
    Figure US20230109104A1-20230406-C01764
    2.560
    Figure US20230109104A1-20230406-C01765
    2.561
    Figure US20230109104A1-20230406-C01766
    2.562
    Figure US20230109104A1-20230406-C01767
    2.563
    Figure US20230109104A1-20230406-C01768
    2.564
    Figure US20230109104A1-20230406-C01769
    2.565
    Figure US20230109104A1-20230406-C01770
    2.566
    Figure US20230109104A1-20230406-C01771
    2.567
    Figure US20230109104A1-20230406-C01772
    2.568
    Figure US20230109104A1-20230406-C01773
    2.569
    Figure US20230109104A1-20230406-C01774
    2.570
    Figure US20230109104A1-20230406-C01775
    2.571
    Figure US20230109104A1-20230406-C01776
    2.572
    Figure US20230109104A1-20230406-C01777
    2.573
    Figure US20230109104A1-20230406-C01778
    2.574
    Figure US20230109104A1-20230406-C01779
    2.575
    Figure US20230109104A1-20230406-C01780
    2.576
    Figure US20230109104A1-20230406-C01781
    2.577
    Figure US20230109104A1-20230406-C01782
    2.578
    Figure US20230109104A1-20230406-C01783
    2.579
    Figure US20230109104A1-20230406-C01784
    2.580
    Figure US20230109104A1-20230406-C01785
    2.581
    Figure US20230109104A1-20230406-C01786
    2.582
    Figure US20230109104A1-20230406-C01787
    2.583
    Figure US20230109104A1-20230406-C01788
    2.584
    Figure US20230109104A1-20230406-C01789
    2.585
    Figure US20230109104A1-20230406-C01790
    2.586
    Figure US20230109104A1-20230406-C01791
    2.587
    Figure US20230109104A1-20230406-C01792
    2.588
    Figure US20230109104A1-20230406-C01793
    2.589
    Figure US20230109104A1-20230406-C01794
    2.590
    Figure US20230109104A1-20230406-C01795
    2.591
    Figure US20230109104A1-20230406-C01796
    2.592
    Figure US20230109104A1-20230406-C01797
    2.593
    Figure US20230109104A1-20230406-C01798
    2.594
    Figure US20230109104A1-20230406-C01799
    2.595
    Figure US20230109104A1-20230406-C01800
    2.596
    Figure US20230109104A1-20230406-C01801
    2.597
    Figure US20230109104A1-20230406-C01802
    2.598
    Figure US20230109104A1-20230406-C01803
    2.599
    Figure US20230109104A1-20230406-C01804
    2.600
    Figure US20230109104A1-20230406-C01805
    2.601
    Figure US20230109104A1-20230406-C01806
    2.602
    Figure US20230109104A1-20230406-C01807
    2.603
    Figure US20230109104A1-20230406-C01808
    2.604
    Figure US20230109104A1-20230406-C01809
    2.605
    Figure US20230109104A1-20230406-C01810
    2.606
    Figure US20230109104A1-20230406-C01811
    2.607
    Figure US20230109104A1-20230406-C01812
    2.608
    Figure US20230109104A1-20230406-C01813
    2.609
    Figure US20230109104A1-20230406-C01814
    2.610
    Figure US20230109104A1-20230406-C01815
    2.611
    Figure US20230109104A1-20230406-C01816
    2.612
    Figure US20230109104A1-20230406-C01817
    2.613
    Figure US20230109104A1-20230406-C01818
    2.614
    Figure US20230109104A1-20230406-C01819
    2.615
    Figure US20230109104A1-20230406-C01820
    2.616
    Figure US20230109104A1-20230406-C01821
    2.617
    Figure US20230109104A1-20230406-C01822
    2.618
    Figure US20230109104A1-20230406-C01823
    2.619
    Figure US20230109104A1-20230406-C01824
    2.620
    Figure US20230109104A1-20230406-C01825
    2.621
    Figure US20230109104A1-20230406-C01826
    2.622
    Figure US20230109104A1-20230406-C01827
    2.623
    Figure US20230109104A1-20230406-C01828
    2.624
    Figure US20230109104A1-20230406-C01829
    2.625
    Figure US20230109104A1-20230406-C01830
    2.626
    Figure US20230109104A1-20230406-C01831
    2.627
    Figure US20230109104A1-20230406-C01832
    2.628
    Figure US20230109104A1-20230406-C01833
    2.629
    Figure US20230109104A1-20230406-C01834
    2.630
    Figure US20230109104A1-20230406-C01835
    2.631
    Figure US20230109104A1-20230406-C01836
    2.632
    Figure US20230109104A1-20230406-C01837
    2.633
    Figure US20230109104A1-20230406-C01838
    2.634
    Figure US20230109104A1-20230406-C01839
    2.635
    Figure US20230109104A1-20230406-C01840
    2.636
    Figure US20230109104A1-20230406-C01841
    2.637
    Figure US20230109104A1-20230406-C01842
    2.638
    Figure US20230109104A1-20230406-C01843
    2.639
    Figure US20230109104A1-20230406-C01844
    2.640
    Figure US20230109104A1-20230406-C01845
    2.641
    Figure US20230109104A1-20230406-C01846
    2.642
    Figure US20230109104A1-20230406-C01847
    2.643
    Figure US20230109104A1-20230406-C01848
    2.644
    Figure US20230109104A1-20230406-C01849
    2.645
    Figure US20230109104A1-20230406-C01850
    2.646
    Figure US20230109104A1-20230406-C01851
    2.647
    Figure US20230109104A1-20230406-C01852
    2.648
    Figure US20230109104A1-20230406-C01853
    2.649
    Figure US20230109104A1-20230406-C01854
    2.650
    Figure US20230109104A1-20230406-C01855
    2.651
    Figure US20230109104A1-20230406-C01856
    2.652
    Figure US20230109104A1-20230406-C01857
    2.653
    Figure US20230109104A1-20230406-C01858
    2.654
    Figure US20230109104A1-20230406-C01859
    2.655
    Figure US20230109104A1-20230406-C01860
    2.656
    Figure US20230109104A1-20230406-C01861
    2.657
    Figure US20230109104A1-20230406-C01862
    2.658
    Figure US20230109104A1-20230406-C01863
    2.659
    Figure US20230109104A1-20230406-C01864
    2.660
    Figure US20230109104A1-20230406-C01865
    2.661
    Figure US20230109104A1-20230406-C01866
    2.662
    Figure US20230109104A1-20230406-C01867
    2.663
    Figure US20230109104A1-20230406-C01868
    2.664
    Figure US20230109104A1-20230406-C01869
    2.665
    Figure US20230109104A1-20230406-C01870
    2.666
    Figure US20230109104A1-20230406-C01871
    2.667
    Figure US20230109104A1-20230406-C01872
    2.668
    Figure US20230109104A1-20230406-C01873
    2.669
    Figure US20230109104A1-20230406-C01874
    2.670
    Figure US20230109104A1-20230406-C01875
    2.671
    Figure US20230109104A1-20230406-C01876
    2.672
    Figure US20230109104A1-20230406-C01877
    2.673
    Figure US20230109104A1-20230406-C01878
    2.674
    Figure US20230109104A1-20230406-C01879
    2.675
    Figure US20230109104A1-20230406-C01880
    2.676
    Figure US20230109104A1-20230406-C01881
    2.677
    Figure US20230109104A1-20230406-C01882
    2.678
    Figure US20230109104A1-20230406-C01883
    2.679
    Figure US20230109104A1-20230406-C01884
    2.680
    Figure US20230109104A1-20230406-C01885
    2.681
    Figure US20230109104A1-20230406-C01886
    2.682
    Figure US20230109104A1-20230406-C01887
    2.683
    Figure US20230109104A1-20230406-C01888
    2.684
    Figure US20230109104A1-20230406-C01889
    2.685
    Figure US20230109104A1-20230406-C01890
    2.686
    Figure US20230109104A1-20230406-C01891
    2.687
    Figure US20230109104A1-20230406-C01892
    2.688
    Figure US20230109104A1-20230406-C01893
    2.689
    Figure US20230109104A1-20230406-C01894
    2.690
    Figure US20230109104A1-20230406-C01895
    2.691
    Figure US20230109104A1-20230406-C01896
    2.692
    Figure US20230109104A1-20230406-C01897
    2.693
    Figure US20230109104A1-20230406-C01898
    2.694
    Figure US20230109104A1-20230406-C01899
    2.695
    Figure US20230109104A1-20230406-C01900
    2.696
    Figure US20230109104A1-20230406-C01901
    2.697
    Figure US20230109104A1-20230406-C01902
    2.698
    Figure US20230109104A1-20230406-C01903
    2.699
    Figure US20230109104A1-20230406-C01904
    2.700
    Figure US20230109104A1-20230406-C01905
    2.701
    Figure US20230109104A1-20230406-C01906
    2.702
    Figure US20230109104A1-20230406-C01907
    2.703
    Figure US20230109104A1-20230406-C01908
    2.704
    Figure US20230109104A1-20230406-C01909
    2.705
    Figure US20230109104A1-20230406-C01910
    2.706
    Figure US20230109104A1-20230406-C01911
    2.707
    Figure US20230109104A1-20230406-C01912
    2.708
    Figure US20230109104A1-20230406-C01913
  • TABLE-1B
    Figure US20230109104A1-20230406-C01914
    2.709
    Figure US20230109104A1-20230406-C01915
    2.710
    Figure US20230109104A1-20230406-C01916
    2.711
    Figure US20230109104A1-20230406-C01917
    2.712
    Figure US20230109104A1-20230406-C01918
    2.713
    Figure US20230109104A1-20230406-C01919
    2.714
    Figure US20230109104A1-20230406-C01920
    2.715
    Figure US20230109104A1-20230406-C01921
    2.716
    Figure US20230109104A1-20230406-C01922
    2.717
    Figure US20230109104A1-20230406-C01923
    2.718
    Figure US20230109104A1-20230406-C01924
    2.719
    Figure US20230109104A1-20230406-C01925
    2.720
    Figure US20230109104A1-20230406-C01926
    2.721
    Figure US20230109104A1-20230406-C01927
    2.722
    Figure US20230109104A1-20230406-C01928
    2.723
    Figure US20230109104A1-20230406-C01929
    2.724
    Figure US20230109104A1-20230406-C01930
    2.725
    Figure US20230109104A1-20230406-C01931
    2.726
    Figure US20230109104A1-20230406-C01932
    2.727
    Figure US20230109104A1-20230406-C01933
    2.728
    Figure US20230109104A1-20230406-C01934
    2.729
    Figure US20230109104A1-20230406-C01935
    2.730
    Figure US20230109104A1-20230406-C01936
    2.731
    Figure US20230109104A1-20230406-C01937
    2.732
    Figure US20230109104A1-20230406-C01938
    2.733
    Figure US20230109104A1-20230406-C01939
    2.734
    Figure US20230109104A1-20230406-C01940
    2.735
    Figure US20230109104A1-20230406-C01941
    2.736
    Figure US20230109104A1-20230406-C01942
    2.737
    Figure US20230109104A1-20230406-C01943
    2.738
    Figure US20230109104A1-20230406-C01944
    2.739
    Figure US20230109104A1-20230406-C01945
    2.740
    Figure US20230109104A1-20230406-C01946
    2.741
    Figure US20230109104A1-20230406-C01947
    2.742
    Figure US20230109104A1-20230406-C01948
    2.743
    Figure US20230109104A1-20230406-C01949
    2.744
    Figure US20230109104A1-20230406-C01950
    2.745
    Figure US20230109104A1-20230406-C01951
    2.746
    Figure US20230109104A1-20230406-C01952
    2.747
    Figure US20230109104A1-20230406-C01953
    2.748
    Figure US20230109104A1-20230406-C01954
    2.749
    Figure US20230109104A1-20230406-C01955
    2.750
    Figure US20230109104A1-20230406-C01956
    2.751
    Figure US20230109104A1-20230406-C01957
    2.752
    Figure US20230109104A1-20230406-C01958
    2.753
    Figure US20230109104A1-20230406-C01959
    2.754
    Figure US20230109104A1-20230406-C01960
    2.755
    Figure US20230109104A1-20230406-C01961
    2.756
    Figure US20230109104A1-20230406-C01962
    2.757
    Figure US20230109104A1-20230406-C01963
    2.758
    Figure US20230109104A1-20230406-C01964
    2.759
    Figure US20230109104A1-20230406-C01965
    2.760
    Figure US20230109104A1-20230406-C01966
    2.761
    Figure US20230109104A1-20230406-C01967
    2.762
    Figure US20230109104A1-20230406-C01968
    2.763
    Figure US20230109104A1-20230406-C01969
    2.764
    Figure US20230109104A1-20230406-C01970
    2.765
    Figure US20230109104A1-20230406-C01971
    2.766
    Figure US20230109104A1-20230406-C01972
    2.767
    Figure US20230109104A1-20230406-C01973
    2.768
    Figure US20230109104A1-20230406-C01974
    2.769
    Figure US20230109104A1-20230406-C01975
    2.770
    Figure US20230109104A1-20230406-C01976
    2.771
    Figure US20230109104A1-20230406-C01977
    2.772
    Figure US20230109104A1-20230406-C01978
    2.773
    Figure US20230109104A1-20230406-C01979
    2.774
    Figure US20230109104A1-20230406-C01980
    2.775
    Figure US20230109104A1-20230406-C01981
    2.776
    Figure US20230109104A1-20230406-C01982
    2.777
    Figure US20230109104A1-20230406-C01983
    2.778
    Figure US20230109104A1-20230406-C01984
    2.779
    Figure US20230109104A1-20230406-C01985
    2.780
    Figure US20230109104A1-20230406-C01986
    2.781
    Figure US20230109104A1-20230406-C01987
    2.782
    Figure US20230109104A1-20230406-C01988
    2.783
    Figure US20230109104A1-20230406-C01989
    2.784
    Figure US20230109104A1-20230406-C01990
    2.785
    Figure US20230109104A1-20230406-C01991
    2.786
    Figure US20230109104A1-20230406-C01992
    2.787
    Figure US20230109104A1-20230406-C01993
    2.788
    Figure US20230109104A1-20230406-C01994
    2.789
    Figure US20230109104A1-20230406-C01995
    2.790
    Figure US20230109104A1-20230406-C01996
    2.791
    Figure US20230109104A1-20230406-C01997
    2.792
    Figure US20230109104A1-20230406-C01998
    2.793
    Figure US20230109104A1-20230406-C01999
    2.794
    Figure US20230109104A1-20230406-C02000
    2.795
    Figure US20230109104A1-20230406-C02001
    2.796
    Figure US20230109104A1-20230406-C02002
    2.797
    Figure US20230109104A1-20230406-C02003
    2.798
    Figure US20230109104A1-20230406-C02004
    2.799
    Figure US20230109104A1-20230406-C02005
    2.800
    Figure US20230109104A1-20230406-C02006
    2.801
    Figure US20230109104A1-20230406-C02007
    2.802
    Figure US20230109104A1-20230406-C02008
    2.803
    Figure US20230109104A1-20230406-C02009
    2.804
    Figure US20230109104A1-20230406-C02010
    2.805
    Figure US20230109104A1-20230406-C02011
    2.806
    Figure US20230109104A1-20230406-C02012
    2.807
    Figure US20230109104A1-20230406-C02013
    2.808
    Figure US20230109104A1-20230406-C02014
    2.809
    Figure US20230109104A1-20230406-C02015
    2.810
    Figure US20230109104A1-20230406-C02016
    2.811
    Figure US20230109104A1-20230406-C02017
    2.812
    Figure US20230109104A1-20230406-C02018
    2.813
    Figure US20230109104A1-20230406-C02019
    2.814
    Figure US20230109104A1-20230406-C02020
    2.815
    Figure US20230109104A1-20230406-C02021
    2.816
    Figure US20230109104A1-20230406-C02022
    2.817
    Figure US20230109104A1-20230406-C02023
    2.818
    Figure US20230109104A1-20230406-C02024
    2.819
    Figure US20230109104A1-20230406-C02025
    2.820
    Figure US20230109104A1-20230406-C02026
    2.821
    Figure US20230109104A1-20230406-C02027
    2.822
    Figure US20230109104A1-20230406-C02028
    2.823
    Figure US20230109104A1-20230406-C02029
    2.824
    Figure US20230109104A1-20230406-C02030
    2.825
    Figure US20230109104A1-20230406-C02031
    2.826
    Figure US20230109104A1-20230406-C02032
    2.827
    Figure US20230109104A1-20230406-C02033
    2.828
    Figure US20230109104A1-20230406-C02034
    2.829
    Figure US20230109104A1-20230406-C02035
    2.830
    Figure US20230109104A1-20230406-C02036
    2.831
    Figure US20230109104A1-20230406-C02037
    2.832
    Figure US20230109104A1-20230406-C02038
    2.833
    Figure US20230109104A1-20230406-C02039
    2.834
    Figure US20230109104A1-20230406-C02040
    2.835
    Figure US20230109104A1-20230406-C02041
    2.836
    Figure US20230109104A1-20230406-C02042
    2.837
    Figure US20230109104A1-20230406-C02043
    2.838
    Figure US20230109104A1-20230406-C02044
    2.839
    Figure US20230109104A1-20230406-C02045
    2.840
    Figure US20230109104A1-20230406-C02046
    2.841
    Figure US20230109104A1-20230406-C02047
    2.842
    Figure US20230109104A1-20230406-C02048
    2.843
    Figure US20230109104A1-20230406-C02049
    2.844
    Figure US20230109104A1-20230406-C02050
    2.845
    Figure US20230109104A1-20230406-C02051
    2.846
    Figure US20230109104A1-20230406-C02052
    2.847
    Figure US20230109104A1-20230406-C02053
    2.848
    Figure US20230109104A1-20230406-C02054
    2.849
    Figure US20230109104A1-20230406-C02055
    2.850
    Figure US20230109104A1-20230406-C02056
    2.851
    Figure US20230109104A1-20230406-C02057
    2.852
    Figure US20230109104A1-20230406-C02058
    2.853
    Figure US20230109104A1-20230406-C02059
    2.854
    Figure US20230109104A1-20230406-C02060
    2.855
    Figure US20230109104A1-20230406-C02061
    2.856
    Figure US20230109104A1-20230406-C02062
    2.857
    Figure US20230109104A1-20230406-C02063
    2.858
    Figure US20230109104A1-20230406-C02064
    2.859
    Figure US20230109104A1-20230406-C02065
    2.860
    Figure US20230109104A1-20230406-C02066
    2.861
    Figure US20230109104A1-20230406-C02067
    2.862
    Figure US20230109104A1-20230406-C02068
    2.863
    Figure US20230109104A1-20230406-C02069
    2.864
    Figure US20230109104A1-20230406-C02070
    2.865
    Figure US20230109104A1-20230406-C02071
    2.866
    Figure US20230109104A1-20230406-C02072
    2.867
    Figure US20230109104A1-20230406-C02073
    2.868
    Figure US20230109104A1-20230406-C02074
    2.869
    Figure US20230109104A1-20230406-C02075
    2.870
    Figure US20230109104A1-20230406-C02076
    2.871
    Figure US20230109104A1-20230406-C02077
    2.872
    Figure US20230109104A1-20230406-C02078
    2.873
    Figure US20230109104A1-20230406-C02079
    2.874
    Figure US20230109104A1-20230406-C02080
    2.875
    Figure US20230109104A1-20230406-C02081
    2.876
    Figure US20230109104A1-20230406-C02082
    2.877
    Figure US20230109104A1-20230406-C02083
    2.878
    Figure US20230109104A1-20230406-C02084
    2.879
    Figure US20230109104A1-20230406-C02085
    2.880
    Figure US20230109104A1-20230406-C02086
    2.881
    Figure US20230109104A1-20230406-C02087
    2.882
    Figure US20230109104A1-20230406-C02088
    2.883
    Figure US20230109104A1-20230406-C02089
    2.884
    Figure US20230109104A1-20230406-C02090
    2.885
    Figure US20230109104A1-20230406-C02091
    2.886
    Figure US20230109104A1-20230406-C02092
    2.887
    Figure US20230109104A1-20230406-C02093
    2.888
    Figure US20230109104A1-20230406-C02094
    2.889
    Figure US20230109104A1-20230406-C02095
    2.890
    Figure US20230109104A1-20230406-C02096
    2.891
    Figure US20230109104A1-20230406-C02097
    2.892
    Figure US20230109104A1-20230406-C02098
    2.893
    Figure US20230109104A1-20230406-C02099
    2.894
    Figure US20230109104A1-20230406-C02100
    2.895
    Figure US20230109104A1-20230406-C02101
    2.896
    Figure US20230109104A1-20230406-C02102
    2.897
    Figure US20230109104A1-20230406-C02103
    2.898
    Figure US20230109104A1-20230406-C02104
    2.899
    Figure US20230109104A1-20230406-C02105
    2.900
    Figure US20230109104A1-20230406-C02106
    2.901
    Figure US20230109104A1-20230406-C02107
    2.902
    Figure US20230109104A1-20230406-C02108
    2.903
    Figure US20230109104A1-20230406-C02109
    2.904
    Figure US20230109104A1-20230406-C02110
    2.905
    Figure US20230109104A1-20230406-C02111
    2.906
    Figure US20230109104A1-20230406-C02112
    2.907
    Figure US20230109104A1-20230406-C02113
    2.908
    Figure US20230109104A1-20230406-C02114
    2.909
    Figure US20230109104A1-20230406-C02115
    2.910
    Figure US20230109104A1-20230406-C02116
    2.911
    Figure US20230109104A1-20230406-C02117
    2.912
    Figure US20230109104A1-20230406-C02118
    2.913
    Figure US20230109104A1-20230406-C02119
    2.914
    Figure US20230109104A1-20230406-C02120
    2.915
    Figure US20230109104A1-20230406-C02121
    2.916
    Figure US20230109104A1-20230406-C02122
    2.917
    Figure US20230109104A1-20230406-C02123
    2.918
    Figure US20230109104A1-20230406-C02124
    2.919
    Figure US20230109104A1-20230406-C02125
    2.920
    Figure US20230109104A1-20230406-C02126
    2.921
    Figure US20230109104A1-20230406-C02127
    2.922
    Figure US20230109104A1-20230406-C02128
    2.923
    Figure US20230109104A1-20230406-C02129
    2.924
    Figure US20230109104A1-20230406-C02130
    2.925
    Figure US20230109104A1-20230406-C02131
    2.926
    Figure US20230109104A1-20230406-C02132
    2.927
    Figure US20230109104A1-20230406-C02133
    2.928
    Figure US20230109104A1-20230406-C02134
    2.929
    Figure US20230109104A1-20230406-C02135
    2.930
    Figure US20230109104A1-20230406-C02136
    2.931
    Figure US20230109104A1-20230406-C02137
    2.932
    Figure US20230109104A1-20230406-C02138
    2.933
    Figure US20230109104A1-20230406-C02139
    2.934
    Figure US20230109104A1-20230406-C02140
    2.935
    Figure US20230109104A1-20230406-C02141
    2.936
    Figure US20230109104A1-20230406-C02142
    2.937
    Figure US20230109104A1-20230406-C02143
    2.938
    Figure US20230109104A1-20230406-C02144
    2.939
    Figure US20230109104A1-20230406-C02145
    2.940
    Figure US20230109104A1-20230406-C02146
    2.941
    Figure US20230109104A1-20230406-C02147
    2.942
    Figure US20230109104A1-20230406-C02148
    2.943
    Figure US20230109104A1-20230406-C02149
    2.944
    Figure US20230109104A1-20230406-C02150
    2.945
    Figure US20230109104A1-20230406-C02151
    2.946
    Figure US20230109104A1-20230406-C02152
    2.947
    Figure US20230109104A1-20230406-C02153
    2.948
    Figure US20230109104A1-20230406-C02154
    2.949
    Figure US20230109104A1-20230406-C02155
    2.950
    Figure US20230109104A1-20230406-C02156
    2.951
    Figure US20230109104A1-20230406-C02157
    2.952
    Figure US20230109104A1-20230406-C02158
    2.953
    Figure US20230109104A1-20230406-C02159
    2.954
    Figure US20230109104A1-20230406-C02160
    2.955
    Figure US20230109104A1-20230406-C02161
    2.956
    Figure US20230109104A1-20230406-C02162
    2.957
    Figure US20230109104A1-20230406-C02163
    2.958
    Figure US20230109104A1-20230406-C02164
    2.959
    Figure US20230109104A1-20230406-C02165
    2.960
    Figure US20230109104A1-20230406-C02166
    2.961
    Figure US20230109104A1-20230406-C02167
    2.962
    Figure US20230109104A1-20230406-C02168
    2.963
    Figure US20230109104A1-20230406-C02169
    2.964
    Figure US20230109104A1-20230406-C02170
    2.965
    Figure US20230109104A1-20230406-C02171
    2.966
    Figure US20230109104A1-20230406-C02172
    2.967
    Figure US20230109104A1-20230406-C02173
    2.968
    Figure US20230109104A1-20230406-C02174
    2.969
    Figure US20230109104A1-20230406-C02175
    2.970
    Figure US20230109104A1-20230406-C02176
    2.971
    Figure US20230109104A1-20230406-C02177
    2.972
    Figure US20230109104A1-20230406-C02178
    2.973
    Figure US20230109104A1-20230406-C02179
    2.974
    Figure US20230109104A1-20230406-C02180
    2.975
    Figure US20230109104A1-20230406-C02181
    2.976
    Figure US20230109104A1-20230406-C02182
    2.977
    Figure US20230109104A1-20230406-C02183
    2.978
    Figure US20230109104A1-20230406-C02184
    2.979
    Figure US20230109104A1-20230406-C02185
    2.980
    Figure US20230109104A1-20230406-C02186
    2.981
    Figure US20230109104A1-20230406-C02187
    2.982
    Figure US20230109104A1-20230406-C02188
    2.983
    Figure US20230109104A1-20230406-C02189
    2.984
    Figure US20230109104A1-20230406-C02190
    2.985
    Figure US20230109104A1-20230406-C02191
    2.986
    Figure US20230109104A1-20230406-C02192
    2.987
    Figure US20230109104A1-20230406-C02193
    2.988
    Figure US20230109104A1-20230406-C02194
    2.989
    Figure US20230109104A1-20230406-C02195
    2.990
    Figure US20230109104A1-20230406-C02196
    2.991
    Figure US20230109104A1-20230406-C02197
    2.992
    Figure US20230109104A1-20230406-C02198
    2.993
    Figure US20230109104A1-20230406-C02199
    2.994
    Figure US20230109104A1-20230406-C02200
    2.995
    Figure US20230109104A1-20230406-C02201
    2.996
    Figure US20230109104A1-20230406-C02202
    2.997
    Figure US20230109104A1-20230406-C02203
    2.998
    Figure US20230109104A1-20230406-C02204
    2.999
    Figure US20230109104A1-20230406-C02205
    3.000
    Figure US20230109104A1-20230406-C02206
    3.001
    Figure US20230109104A1-20230406-C02207
    3.002
    Figure US20230109104A1-20230406-C02208
    3.003
    Figure US20230109104A1-20230406-C02209
    3.004
    Figure US20230109104A1-20230406-C02210
    3.005
    Figure US20230109104A1-20230406-C02211
    3.006
    Figure US20230109104A1-20230406-C02212
    3.007
    Figure US20230109104A1-20230406-C02213
    3.008
    Figure US20230109104A1-20230406-C02214
    3.009
    Figure US20230109104A1-20230406-C02215
    3.010
    Figure US20230109104A1-20230406-C02216
    3.011
    Figure US20230109104A1-20230406-C02217
    3.012
    Figure US20230109104A1-20230406-C02218
    3.013
    Figure US20230109104A1-20230406-C02219
    3.014
    Figure US20230109104A1-20230406-C02220
    3.015
    Figure US20230109104A1-20230406-C02221
    3.016
    Figure US20230109104A1-20230406-C02222
    3.017
    Figure US20230109104A1-20230406-C02223
    3.018
    Figure US20230109104A1-20230406-C02224
    3.019
    Figure US20230109104A1-20230406-C02225
    3.020
    Figure US20230109104A1-20230406-C02226
    3.021
    Figure US20230109104A1-20230406-C02227
    3.022
    Figure US20230109104A1-20230406-C02228
    3.023
    Figure US20230109104A1-20230406-C02229
    3.024
    Figure US20230109104A1-20230406-C02230
    3.025
    Figure US20230109104A1-20230406-C02231
    3.026
    Figure US20230109104A1-20230406-C02232
    3.027
    Figure US20230109104A1-20230406-C02233
    3.028
    Figure US20230109104A1-20230406-C02234
    3.029
    Figure US20230109104A1-20230406-C02235
    3.030
    Figure US20230109104A1-20230406-C02236
    3.031
    Figure US20230109104A1-20230406-C02237
    3.032
    Figure US20230109104A1-20230406-C02238
    3.033
    Figure US20230109104A1-20230406-C02239
    3.034
    Figure US20230109104A1-20230406-C02240
    3.035
    Figure US20230109104A1-20230406-C02241
    3.036
    Figure US20230109104A1-20230406-C02242
    3.037
    Figure US20230109104A1-20230406-C02243
    3.038
    Figure US20230109104A1-20230406-C02244
    3.039
    Figure US20230109104A1-20230406-C02245
    3.040
    Figure US20230109104A1-20230406-C02246
    3.041
    Figure US20230109104A1-20230406-C02247
    3.042
    Figure US20230109104A1-20230406-C02248
    3.043
    Figure US20230109104A1-20230406-C02249
    3.044
    Figure US20230109104A1-20230406-C02250
    3.045
    Figure US20230109104A1-20230406-C02251
    3.046
    Figure US20230109104A1-20230406-C02252
    3.047
    Figure US20230109104A1-20230406-C02253
    3.048
    Figure US20230109104A1-20230406-C02254
    3.049
    Figure US20230109104A1-20230406-C02255
    3.050
    Figure US20230109104A1-20230406-C02256
    3.051
    Figure US20230109104A1-20230406-C02257
    3.052
    Figure US20230109104A1-20230406-C02258
    3.053
    Figure US20230109104A1-20230406-C02259
    3.054
    Figure US20230109104A1-20230406-C02260
    3.055
    Figure US20230109104A1-20230406-C02261
    3.056
    Figure US20230109104A1-20230406-C02262
    3.057
    Figure US20230109104A1-20230406-C02263
    3.058
    Figure US20230109104A1-20230406-C02264
    3.059
    Figure US20230109104A1-20230406-C02265
    3.060
    Figure US20230109104A1-20230406-C02266
    3.061
    Figure US20230109104A1-20230406-C02267
    3.062
    Figure US20230109104A1-20230406-C02268
    3.063
    Figure US20230109104A1-20230406-C02269
    3.064
    Figure US20230109104A1-20230406-C02270
    3.065
    Figure US20230109104A1-20230406-C02271
    3.066
    Figure US20230109104A1-20230406-C02272
    3.067
    Figure US20230109104A1-20230406-C02273
    3.068
    Figure US20230109104A1-20230406-C02274
    3.069
    Figure US20230109104A1-20230406-C02275
    3.070
    Figure US20230109104A1-20230406-C02276
    3.071
    Figure US20230109104A1-20230406-C02277
    3.072
    Figure US20230109104A1-20230406-C02278
    3.073
    Figure US20230109104A1-20230406-C02279
    3.074
    Figure US20230109104A1-20230406-C02280
    3.075
    Figure US20230109104A1-20230406-C02281
    3.076
    Figure US20230109104A1-20230406-C02282
    3.077
    Figure US20230109104A1-20230406-C02283
    3.078
    Figure US20230109104A1-20230406-C02284
    3.079
    Figure US20230109104A1-20230406-C02285
    3.080
    Figure US20230109104A1-20230406-C02286
    3.081
    Figure US20230109104A1-20230406-C02287
    3.082
    Figure US20230109104A1-20230406-C02288
    3.083
    Figure US20230109104A1-20230406-C02289
    3.084
    Figure US20230109104A1-20230406-C02290
    3.085
    Figure US20230109104A1-20230406-C02291
    3.086
    Figure US20230109104A1-20230406-C02292
    3.087
    Figure US20230109104A1-20230406-C02293
    3.088
    Figure US20230109104A1-20230406-C02294
    3.089
    Figure US20230109104A1-20230406-C02295
    3.090
    Figure US20230109104A1-20230406-C02296
    3.091
    Figure US20230109104A1-20230406-C02297
    3.092
    Figure US20230109104A1-20230406-C02298
    3.093
    Figure US20230109104A1-20230406-C02299
    3.094
    Figure US20230109104A1-20230406-C02300
    3.095
    Figure US20230109104A1-20230406-C02301
    3.096
    Figure US20230109104A1-20230406-C02302
    3.097
    Figure US20230109104A1-20230406-C02303
    3.098
    Figure US20230109104A1-20230406-C02304
    3.099
    Figure US20230109104A1-20230406-C02305
    3.100
    Figure US20230109104A1-20230406-C02306
    3.101
    Figure US20230109104A1-20230406-C02307
    3.102
    Figure US20230109104A1-20230406-C02308
    3.103
    Figure US20230109104A1-20230406-C02309
    3.104
    Figure US20230109104A1-20230406-C02310
    3.105
    Figure US20230109104A1-20230406-C02311
    3.106
    Figure US20230109104A1-20230406-C02312
    3.107
    Figure US20230109104A1-20230406-C02313
    3.108
    Figure US20230109104A1-20230406-C02314
    3.109
    Figure US20230109104A1-20230406-C02315
    3.110
    Figure US20230109104A1-20230406-C02316
    3.111
    Figure US20230109104A1-20230406-C02317
    3.112
    Figure US20230109104A1-20230406-C02318
    3.113
    Figure US20230109104A1-20230406-C02319
    3.114
    Figure US20230109104A1-20230406-C02320
    3.115
    Figure US20230109104A1-20230406-C02321
    3.116
    Figure US20230109104A1-20230406-C02322
    3.117
    Figure US20230109104A1-20230406-C02323
    3.118
    Figure US20230109104A1-20230406-C02324
    3.119
    Figure US20230109104A1-20230406-C02325
    3.120
    Figure US20230109104A1-20230406-C02326
    3.121
    Figure US20230109104A1-20230406-C02327
    3.122
    Figure US20230109104A1-20230406-C02328
    3.123
    Figure US20230109104A1-20230406-C02329
    3.124
    Figure US20230109104A1-20230406-C02330
    3.125
    Figure US20230109104A1-20230406-C02331
    3.126
    Figure US20230109104A1-20230406-C02332
    3.127
    Figure US20230109104A1-20230406-C02333
    3.128
    Figure US20230109104A1-20230406-C02334
    3.129
    Figure US20230109104A1-20230406-C02335
    3.130
    Figure US20230109104A1-20230406-C02336
    3.131
    Figure US20230109104A1-20230406-C02337
    3.132
    Figure US20230109104A1-20230406-C02338
    3.133
    Figure US20230109104A1-20230406-C02339
    3.134
    Figure US20230109104A1-20230406-C02340
    3.135
    Figure US20230109104A1-20230406-C02341
    3.136
  • In some embodiments, provided herein is a compound described in Table 1A or Table 1B, or a tautomer thereof, or a salt of any of the foregoing, and uses thereof. In some embodiments, provided herein is a compound described in Table 1A or Table 1B or a pharmaceutically acceptable salt thereof. In some embodiments, provided herein is a compound described in Table 1A or Table 1B, or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing.
  • The embodiments and variations described herein are suitable for compounds of any formulae detailed herein, where applicable. For example, it is understood that all variations and embodiments for moieties A and B are applicable to any variation of Formula (I) in which such moiety is present. To illustrate, in some embodiments of a compound of Formula (I), or other structure described herein containing moieties A and B, in one variation, B is C6 aryl, 5- to 7-membered heteroaryl, C3-C5 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is optionally substituted by R4, wherein B is fused to A.
  • Representative examples of compounds detailed herein, including intermediates and final compounds according to the present disclosure are depicted herein. It is understood that in one aspect, any of the compounds may be used in the methods detailed herein, including, where applicable, intermediate compounds that may be isolated and administered to an individual.
  • The compounds depicted herein may be present as salts even if salts are not depicted and it is understood that the present disclosure embraces all salts and solvates of the compounds depicted here, as well as the non-salt and non-solvate form of the compound, as is well understood by the skilled artisan. In some embodiments, the salts of the compounds provided herein are pharmaceutically acceptable salts. Where one or more tertiary amine moiety is present in the compound, the N-oxides are also provided and described.
  • Where tautomeric forms may be present for any of the compounds described herein, each and every tautomeric form is intended even though only one or some of the tautomeric forms may be explicitly depicted. The tautomeric forms specifically depicted may or may not be the predominant forms in solution or when used according to the methods described herein. It is understood that tautomeric forms of a compound of the formulae described herein may be present, for example, when tautomeric forms of a substituent are present, such as when a substituent embraces a keto-enol tautomer or the like.
  • The present disclosure also includes any or all of the stereochemical forms, including any enantiomeric or diastereomeric forms of the compounds described, such as the compounds of Table 1A or Table 1B. The structure or name is intended to embrace all possible stereoisomers of a compound depicted, and each unique stereoisomer has a compound number bearing a suffix “a”, “b”, etc. All forms of the compounds are also embraced by the invention, such as crystalline or non-crystalline forms of the compounds. Compositions comprising a compound of the invention are also intended, such as a composition of substantially pure compound, including a specific stereochemical form thereof, or a composition comprising mixtures of compounds of the invention in any ratio, including two or more stereochemical forms, such as in a racemic or non-racemic mixture.
  • The invention also intends isotopically-labeled and/or isotopically-enriched forms of compounds described herein. The compounds herein may contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute such compounds. In some embodiments, the compound is isotopically-labeled, such as an isotopically-labeled compound of Formula (I) or variations thereof described herein, where a fraction of one or more atoms are replaced by an isotope of the same element. Exemplary isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, chlorine, such as 2H, 3H, 11C, 13C, 14C 13N, 15O, 17O, 32P, 35S, 8F, 36C1. Certain isotope labeled compounds (e.g. 3H and 14C) is useful in compound or substrate tissue distribution studies. Incorporation of heavier isotopes such as deuterium (2H) can afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life, or reduced dosage requirements and, hence may be preferred in some instances.
  • Isotopically-labeled compounds of the present invention can generally be prepared by standard methods and techniques known to those skilled in the art or by procedures similar to those described in the accompanying Examples substituting appropriate isotopically-labeled reagents in place of the corresponding non-labeled reagent.
  • The invention also includes any or all metabolites of any of the compounds described. The metabolites may include any chemical species generated by a biotransformation of any of the compounds described, such as intermediates and products of metabolism of the compound, such as would be generated in vivo following administration to a human.
  • Articles of manufacture comprising a compound described herein, or a salt or solvate thereof, in a suitable container are provided. The container may be a vial, jar, ampoule, preloaded syringe, i.v. bag, and the like.
  • Preferably, the compounds detailed herein are orally bioavailable. However, the compounds may also be formulated for parenteral (e.g., intravenous) administration.
  • One or several compounds described herein can be used in the preparation of a medicament by combining the compound or compounds as an active ingredient with a pharmacologically acceptable carrier, which are known in the art. Depending on the therapeutic form of the medication, the carrier may be in various forms. In one variation, the manufacture of a medicament is for use in any of the methods disclosed herein, e.g., for the treatment of cancer.
  • General Synthetic Methods
  • The compounds of the invention may be prepared by a number of processes as generally described below and more specifically in the Examples hereinafter (such as the schemes provided in the Examples below). In the following process descriptions, the symbols when used in the formulae depicted are to be understood to represent those groups described above in relation to the formulae herein.
  • Where it is desired to obtain a particular enantiomer of a compound, this may be accomplished from a corresponding mixture of enantiomers using any suitable conventional procedure for separating or resolving enantiomers. Thus, for example, diastereomeric derivatives may be produced by reaction of a mixture of enantiomers, e.g., a racemate, and an appropriate chiral compound. The diastereomers may then be separated by any convenient means, for example by crystallization and the desired enantiomer recovered. In another resolution process, a racemate may be separated using chiral High Performance Liquid Chromatography. Alternatively, if desired a particular enantiomer may be obtained by using an appropriate chiral intermediate in one of the processes described.
  • Chromatography, recrystallization and other conventional separation procedures may also be used with intermediates or final products where it is desired to obtain a particular isomer of a compound or to otherwise purify a product of a reaction.
  • Solvates and/or polymorphs of a compound provided herein or a salt thereof are also contemplated. Solvates contain either stoichiometric or non-stoichiometric amounts of a solvent, and are often formed during the process of crystallization. Hydrates are formed when the solvent is water, or alcoholates are formed when the solvent is alcohol. Polymorphs include the different crystal packing arrangements of the same elemental composition of a compound. Polymorphs usually have different X-ray diffraction patterns, infrared spectra, melting points, density, hardness, crystal shape, optical and electrical properties, stability, and/or solubility. Various factors such as the recrystallization solvent, rate of crystallization, and storage temperature may cause a single crystal form to dominate
  • In some embodiments, compounds of Formula (I), (Ia), (Ia-1 to Ia-12), (1b), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) may be synthesized according to Scheme 1 to Scheme 13.
  • Figure US20230109104A1-20230406-C02342
  • wherein A, B, R1, R2, R3, and R4 are as defined for Formula (I). Particular examples are provided in the Example Section below.
  • Figure US20230109104A1-20230406-C02343
    Figure US20230109104A1-20230406-C02344
  • Figure US20230109104A1-20230406-C02345
    Figure US20230109104A1-20230406-C02346
  • Figure US20230109104A1-20230406-C02347
  • Figure US20230109104A1-20230406-C02348
    Figure US20230109104A1-20230406-C02349
  • Figure US20230109104A1-20230406-C02350
  • Figure US20230109104A1-20230406-C02351
    Figure US20230109104A1-20230406-C02352
  • Figure US20230109104A1-20230406-C02353
  • Figure US20230109104A1-20230406-C02354
    Figure US20230109104A1-20230406-C02355
  • Figure US20230109104A1-20230406-C02356
  • Figure US20230109104A1-20230406-C02357
  • Figure US20230109104A1-20230406-C02358
  • Figure US20230109104A1-20230406-C02359
  • wherein m, n, X1, X2, A, B, R), R2, R3, and R4 are as defined for Formula (I). Particular examples are provided in the Example Section below.
  • Pharmaceutical Compositions and Formulations
  • Pharmaceutical compositions of any of the compounds detailed herein are embraced by this disclosure. Thus, the present disclosure includes pharmaceutical compositions comprising a compound as detailed herein or a salt thereof and a pharmaceutically acceptable carrier or excipient. The present disclosure includes pharmaceutical compositions comprising a compound as detailed herein, or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, and a pharmaceutically acceptable carrier or excipient. In one aspect, the pharmaceutically acceptable salt is an acid addition salt, such as a salt formed with an inorganic or organic acid. Pharmaceutical compositions may take a form suitable for oral, buccal, parenteral, nasal, topical or rectal administration or a form suitable for administration by inhalation.
  • A compound as detailed herein may in one aspect be in a purified form and compositions comprising a compound in purified forms are detailed herein. Compositions comprising a compound as detailed herein or a salt thereof are provided, such as compositions of substantially pure compounds. In some embodiments, a composition containing a compound as detailed herein or a salt thereof is in substantially pure form.
  • In one variation, the compounds herein are synthetic compounds prepared for administration to an individual. In another variation, compositions are provided containing a compound in substantially pure form. In another variation, the present disclosure embraces pharmaceutical compositions comprising a compound detailed herein and a pharmaceutically acceptable carrier. In another variation, methods of administering a compound are provided. The purified forms, pharmaceutical compositions and methods of administering the compounds are suitable for any compound or form thereof detailed herein.
  • A compound detailed herein or salt thereof may be formulated for any available delivery route, including an oral, mucosal (e.g., nasal, sublingual, vaginal, buccal or rectal), parenteral (e.g., intramuscular, subcutaneous or intravenous), topical or transdermal delivery form. A compound or salt thereof may be formulated with suitable carriers to provide delivery forms that include, but are not limited to, tablets, caplets, capsules (such as hard gelatin capsules or soft elastic gelatin capsules), cachets, troches, lozenges, gums, dispersions, suppositories, ointments, cataplasms (poultices), pastes, powders, dressings, creams, solutions, patches, aerosols (e.g., nasal spray or inhalers), gels, suspensions (e.g., aqueous or non-aqueous liquid suspensions, oil-in-water emulsions or water-in-oil liquid emulsions), solutions and elixirs.
  • One or several compounds described herein or a salt thereof can be used in the preparation of a formulation, such as a pharmaceutical formulation, by combining the compound or compounds, or a salt thereof, as an active ingredient with a pharmaceutically acceptable carrier, such as those mentioned above. Depending on the therapeutic form of the system (e.g., transdermal patch vs. oral tablet), the carrier may be in various forms. In addition, pharmaceutical formulations may contain preservatives, solubilizers, stabilizers, re-wetting agents, emulgators, sweeteners, dyes, adjusters, and salts for the adjustment of osmotic pressure, buffers, coating agents or antioxidants. Formulations comprising the compound may also contain other substances which have valuable therapeutic properties. Pharmaceutical formulations may be prepared by known pharmaceutical methods. Suitable formulations can be found, e.g., in Remington's Pharmaceutical Sciences, Mack Publishing Company, Philadelphia, Pa., 20th ed. (2000), which is incorporated herein by reference.
  • Compounds as described herein may be administered to individuals in a form of generally accepted oral compositions, such as tablets, coated tablets, and gel capsules in a hard or in soft shell, emulsions or suspensions. Examples of carriers, which may be used for the preparation of such compositions, are lactose, corn starch or its derivatives, talc, stearate or its salts, etc. Acceptable carriers for gel capsules with soft shell are, for instance, plant oils, wax, fats, semisolid and liquid poly-ols, and so on. In addition, pharmaceutical formulations may contain preservatives, solubilizers, stabilizers, re-wetting agents, emulgators, sweeteners, dyes, adjusters, and salts for the adjustment of osmotic pressure, buffers, coating agents or antioxidants.
  • Any of the compounds described herein can be formulated in a tablet in any dosage form described, for example, a compound as described herein or a salt thereof can be formulated as a 10 mg tablet.
  • Compositions comprising a compound provided herein are also described. In one variation, the composition comprises a compound or salt thereof and a pharmaceutically acceptable carrier or excipient. In another variation, a composition of substantially pure compound is provided.
  • Methods of Use
  • Compounds and compositions detailed herein, such as a pharmaceutical composition containing a compound of any formula provided herein or a salt thereof and a pharmaceutically acceptable carrier or excipient, may be used in methods of administration and treatment as provided herein. Compounds and compositions detailed herein, such as a pharmaceutical composition containing a compound of any formula provided herein or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing and a pharmaceutically acceptable carrier or excipient, may be used in methods of administration and treatment as provided herein. The compounds and compositions may also be used in in vitro methods, such as in vitro methods of administering a compound or composition to cells for screening purposes and/or for conducting quality control assays.
  • Provided herein is a method of treating a disease in an individual comprising administering an effective amount of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or any embodiment, variation or aspect thereof (collectively, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or the present compounds or the compounds detailed or described herein) or a pharmaceutically acceptable salt thereof, to the individual. Further provided herein is a method of treating a proliferative disease in an individual, comprising administering an effective amount of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof, to the individual. Also provided herein is a method of treating cancer in an individual comprising administering an effective amount of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof, to the individual. In some embodiments, the compound is administered to the individual according to a dosage and/or method of administration described herein.
  • In some embodiments, the cancer in the individual has one or more TP53 gene mutations or expresses mutant p53. TP53 is the human gene that encodes p53. In some embodiments, provided herein is a method of treating a cancer in an individual, comprising (a) selecting the individual for treatment based on (i) the presence of one or more mutations of the TP53 gene in the cancer, or (ii) expression of mutant p53 in the cancer, and administering an effective amount of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof, to the individual. In some embodiments, the cancer is assayed for the expression of mutant p53. In some embodiments, the TP53 gene of the cancer is sequenced to detect the one or more mutations. In some embodiments, the TP53 gene is sequenced by biopsying the cancer and sequencing the TP53 gene from the biopsied cancer. In some embodiments, the TP53 gene is sequenced by sequencing circulating-tumor DNA (ctDNA) from the individual.
  • In some embodiments, provided herein is a method of using a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or any embodiment in the manufacture of a medicament for treatment of a disease. In some embodiments, provided herein is a method of using a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or any embodiment in the manufacture of a medicament for treatment of cancer.
  • In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof is used to treat an individual having a proliferative disease, such as cancer as described herein. In some embodiments, the individual is at risk of developing a proliferative disease, such as cancer. In some of these embodiments, the individual is determined to be at risk of developing cancer based upon one or more risk factors. In some of these embodiments, the risk factor is a family history and/or gene associated with cancer.
  • The present compounds or salts thereof are believed to be effective for treating a variety of diseases and disorders. For example, in some embodiments, the present compositions may be used to treat a proliferative disease, such as cancer. In some embodiments the cancer is a solid tumor. In some embodiments the cancer is any of adult and pediatric oncology, myxoid and round cell carcinoma, locally advanced tumors, metastatic cancer, human soft tissue sarcomas, including Ewing's sarcoma, cancer metastases, including lymphatic metastases, squamous cell carcinoma, particularly of the head and neck, esophageal squamous cell carcinoma, oral carcinoma, blood cell malignancies, including multiple myeloma, leukemias, including acute lymphocytic leukemia, acute nonlymphocytic leukemia, chronic lymphocytic leukemia, chronic myelocytic leukemia, and hairy cell leukemia, effusion lymphomas (body cavity based lymphomas), thymic lymphoma, cutaneous T cell lymphoma, Hodgkin's lymphoma, non-Hodgkin's lymphoma, cancer of the adrenal cortex, ACTH-producing tumors, lung cancer, including small cell carcinoma and nonsmall cell cancers, breast cancer, including small cell carcinoma and ductal carcinoma, gastrointestinal cancers, including stomach cancer, colon cancer, colorectal cancer, polyps associated with colorectal neoplasia, pancreatic cancer, liver cancer, urological cancers, including bladder cancer, including primary superficial bladder tumors, invasive transitional cell carcinoma of the bladder, and muscle-invasive bladder cancer, prostate cancer, malignancies of the female genital tract, including ovarian carcinoma, primary peritoneal epithelial neoplasms, cervical carcinoma, uterine endometrial cancers, vaginal cancer, cancer of the vulva, uterine cancer and solid tumors in the ovarian follicle, malignancies of the male genital tract, including testicular cancer and penile cancer, kidney cancer, including renal cell carcinoma, brain cancer, including intrinsic brain tumors, neuroblastoma, astrocytic brain tumors, gliomas, metastatic tumor cell invasion in the central nervous system, bone cancers, including osteomas and osteosarcomas, skin cancers, including melanoma, tumor progression of human skin keratinocytes, squamous cell cancer, thyroid cancer, retinoblastoma, neuroblastoma, peritoneal effusion, malignant pleural effusion, mesothelioma, Wilms's tumors, gall bladder cancer, trophoblastic neoplasms, hemangiopericytoma, and Kaposi's sarcoma.
  • In some embodiments, the compounds and compositions described herein suppress G2-M checkpoint in a cell (such as a cancer cell). In some embodiments, the cancer cell is a cancer cell from any of the cancer types described herein. Suppression of the G2-M DNA damage checkpoint results in premature mitosis of the cell, and consequently apoptosis. In some embodiments, provided herein is a method of suppressing the G2-M DNA damage checkpoint in a cell comprising administering an effective amount of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof, to the cell. In some embodiments, the G2-M DNA damage checkpoint is suppressed in about 40% or more, about 50% or more, about 60% or more, about 70% or more, about 80% or more, about 85% or more, about 90% or more, about 95% or more, about 96% or more, about 97% or more, about 98% or more, or about 99% or more of cells in a cell population. In some embodiments, the G2-M DNA damage checkpoint is suppressed in up to about 99%, up to about 98%, up to about 97%, up to about 96%, up to about 95%, up to about 90%, up to about 85%, or up to about 80% of cells in the cell population.
  • In some embodiments, provided herein is a method of inducing premature mitosis in a cell comprising administering an effective amount of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof, to the cell. In some embodiments, premature mitosis is induced in about 40% or more, about 50% or more, about 60% or more, about 70% or more, about 80% or more, about 85% or more, about 90% or more, about 95% or more, about 96% or more, about 97% or more, about 98% or more, or about 99% or more of cells in a cell population. In some embodiments, premature mitosis is induced in up to about 99%, up to about 98%, up to about 97%, up to about 96%, up to about 95%, up to about 90%, up to about 85%, or up to about 80% of cells in the cell population.
  • In some embodiments, provided herein is a method of inducing apoptosis in a cell comprising administering an effective amount of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof, to the cell. In some embodiments, apoptosis is induced in about 40% or more, about 50% or more, about 60% or more, about 70% or more, about 80% or more, about 85% or more, about 90% or more, about 95% or more, about 96% or more, about 97% or more, about 98% or more, or about 99% or more of cells in a cell population. In some embodiments, apoptosis is induced in up to about 99%, up to about 98%, up to about 97%, up to about 96%, up to about 95%, up to about 90%, up to about 85%, or up to about 80% of cells in the cell population.
  • In some embodiments, provided herein is a method of inhibiting Wee1 in a cell comprising administering an effective amount of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof, to the cell. In some embodiments, Wee1 is inhibited by about 10% or more, about 20% or more, about 30% or more, about 40% or more, about 50% or more, about 60% or more, about 70% or more, about 75% or more, about 80% or more, about 90% or more, about 95% or more, about 96% or more, about 97% or more, about 98% or more, or about 99% or more. In some embodiments, Wee1 is inhibited up to about 99%, up to about 98%, up to about 97%, up to about 96%, up to about 95%, up to about 90%, up to about 85%, up to about 80%, up to about 70%, or up to about 60%. In some embodiments, the activity of Wee1 is measured according to a kinase assay.
  • In some embodiments, provided herein is a method of inhibiting Wee1 comprising contacting Wee1 with an effective amount of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof. In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof binds to Wee1 with an IC50 of less than 1 μM, less than 900 nM, less than 800 nM, less than 700 nM, less than 600 nM, less than 500 nM, less than 400 nM, less than 300 nM, less than 200 nM, less than 100 nM, less than 50 nM, less than 10 nM, less than 5 nM, less than 1 nM, or less than 0.5 nM. In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof binds to Wee1 with an IC50 between 0.1 nM and 1 nM, between 1 nM and 5 nM, between 5 nM and 10 nM, between 10 nM and 50 nM, between 50 nM and 100 nM, between 100 nM and 200 nM, between 200 nM and 300 nM, between 300 nM and 400 nM, between 400 nM and 500 nM, between 500 nM and 600 nM, between 600 nM and 700 nM, between 700 nM and 800 nM, between 800 nM and 900 nM, or between 900 nM and 1 μM. In some embodiments, the IC50 is measured according to a kinase assay. In some embodiments, the IC50 is measured according to a cell cytotoxicity assay.
  • In some embodiments, provided herein is a method of inhibiting the proliferation of a cell, comprising contacting the cell with an effective amount of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof. In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is effective in inhibiting the proliferation of the cell with an IC50 of less than 5 μM, less than 2 μM, less than 1 μM, less than 900 nM, less than 800 nM, less than 700 nM, less than 600 nM, less than 500 nM, less than 400 nM, less than 300 nM, less than 200 nM, less than 100 nM, or less than 50 nM. In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt is effective in inhibiting the proliferation of the cell with an IC50 between 10 nM and 20 nM, between 20 nM and 50 nM, between 50 nM and 100 nM, between 100 nM and 500 nM, between 500 nM and 1 μM, between 1 μM and 2 μM, or between 2 μM and 5 μM. In some embodiments, the IC50 is measured according to a cell proliferation assay.
  • Combination Therapy
  • As provided herein, the presently disclosed compounds or a salt thereof may activate the immune system, for example by inducing apoptosis or suppressing mitosis of cancer cells. Accordingly, the present compounds or a salt thereof may be used in combination with other anti-cancer agents to enhance tumor immunotherapy. In some embodiments, provided herein is a method of treating a disease in an individual comprising administering an effective amount of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or any embodiment, variation or aspect thereof (collectively, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or the present compounds or the compounds detailed or described herein) or a pharmaceutically acceptable salt thereof, and an additional therapeutic agent to the individual. In some embodiments, the disease is a proliferative disease such as cancer.
  • In some embodiments, the additional therapeutic agent is a cancer immunotherapy agent. In some embodiments, the additional therapeutic agent is a chemotherapeutic agent. In some embodiments, the additional therapeutic agent is an immunostimulatory agent. In some embodiments, the additional therapeutic agent targets a checkpoint protein (for example an immune checkpoint inhibitor). In some embodiments, the additional therapeutic agent is effective to stimulate, enhance or improve an immune response against a tumor. In some embodiments, the additional chemotherapeutic agent is a DNA alkylating agent, a platinum-based chemotherapeutic agent, a kinase inhibitor or a DNA damage repair (DDR) pathway inhibitor. In some embodiments, the additional chemotherapeutic agent is a DNA alkylating agent. In some embodiments, the additional chemotherapeutic agent is a platinum-based chemotherapeutic agent. In some embodiments, the additional chemotherapeutic agent is a kinase inhibitor. In some embodiments, the additional chemotherapeutic agent is a DNA damage repair (DDR) pathway inhibitor.
  • In another aspect, provided herein is a combination therapy for the treatment of a disease, such as cancer. In some embodiments, provided herein is a method of treating a disease in an individual comprising administering an effective amount of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or any embodiment, variation or aspect thereof (collectively, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or the present compounds or the compounds detailed or described herein) or a pharmaceutically acceptable salt thereof, in combination with a radiation therapy.
  • In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or any embodiment, variation or aspect thereof (collectively, Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9)) or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of an additional chemotherapeutic agent. In some embodiments, the chemotherapeutic agent is a kinase inhibitor or an agent that inhibits one or more DNA damage repair (DDR) pathways. In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the additional chemotherapeutic agent. In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the additional chemotherapeutic agent.
  • Examples of chemotherapeutic agents that can be used in combination with a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof include DNA-targeted agents, a DNA alkylating agent (such as cyclophosphamide, mechlorethamine, chlorambucil, melphalan, dacarbazine, or nitrosoureas), a topoisomerase inhibitor (such as a Topoisomerase I inhibitor (e.g., irinotecan or topotecan) or a Topoisomerase II inhibitor (e.g., etoposide or teniposide)), an anthracycline (such as daunorubicin, doxorubicin, epirubicin, idarubicin, mitoxantrone, or valrubicin), a histone deacetylase inhibitor (such as vorinostat or romidepsin), a bromodomain inhibitor, other epigenetic inhibitors, a taxane (such as paclitaxel or docetaxel), a kinase inhibitor (such as bortezomib, erlotinib, gefitinib, imatinib, vemurafenib, or vismodegib), an anti-angiogenic inhibitor, a nucleotide analog or precursor analog (such as azacitidine, azathioprine, capecitabine, cytarabine, doxifluridine, 5-fluorouracil, gemcitabine, hydroxyurea, mercaptopurine, methotrexate, or tioguanine), or a platinum-based chemotherapeutic agent (such as cisplatin, carboplatin, or oxaliplatin), pemetrexed, or a combination thereof. In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or any embodiment, variation or aspect thereof (collectively, Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9)) or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a kinase inhibitor (such as bortezomib, erlotinib, gefitinib, imatinib, vemurafenib, or vismodegib). In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the kinase inhibitor. In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the kinase inhibitor.
  • In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or any embodiment, variation or aspect thereof (collectively, Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9)) or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a DNA damaging agent. In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the DNA damaging agent. In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the DNA damaging agent.
  • In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or any embodiment, variation or aspect thereof (collectively, Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9)) or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a DNA alkylating agent (such as cyclophosphamide, mechlorethamine, chlorambucil, melphalan, dacarbazine, or nitrosoureas). In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the DNA alkylating agent. In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the DNA alkylating agent.
  • In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or any embodiment, variation or aspect thereof (collectively, Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9)) or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a topoisomerase inhibitor (such as a Topoisomerase I inhibitor (e.g., irinotecan or topotecan) or a Topoisomerase II inhibitor (e.g., etoposide or teniposide)). In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the topoisomerase inhibitor. In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the topoisomerase inhibitor.
  • In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or any embodiment, variation or aspect thereof (collectively, Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9)) or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of an anthracycline (such as daunorubicin, doxorubicin, epirubicin, idarubicin, mitoxantrone, or valrubicin). In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the anthracycline. In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the anthracycline.
  • In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or any embodiment, variation or aspect thereof (collectively, Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9)) or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a histone deacetylase inhibitor (such as vorinostat or romidepsin). In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the histone deacetylase inhibitor. In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the histone deacetylase inhibitor.
  • In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or any embodiment, variation or aspect thereof (collectively, Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9)) or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a taxane (such as paclitaxel or docetaxel). In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the taxane. In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the taxane.
  • In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or any embodiment, variation or aspect thereof (collectively, Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9)) or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a nucleotide analog or precursor analog (such as azacitidine, azathioprine, capecitabine, cytarabine, doxifluridine, 5-fluorouracil, gemcitabine, hydroxyurea, mercaptopurine, methotrexate, or tioguanine). In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the nucleotide analog or precursor analog. In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the nucleotide analog or precursor analog.
  • In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or any embodiment, variation or aspect thereof (collectively, Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9)) or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a platinum-based chemotherapeutic agent (such as cisplatin, carboplatin, or oxaliplatin). In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the platinum-based chemotherapeutic agent. In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the platinum-based chemotherapeutic agent.
  • In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or any embodiment, variation or aspect thereof (collectively, Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9)) or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of pemetrexed. In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the pemetrexed. In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the pemetrexed.
  • In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or any embodiment, variation or aspect thereof (collectively, Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9)) or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a DDR pathway inhibitor. In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the DDR pathway inhibitor. In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the DDR pathway inhibitor. Examples of inhibitors of the DDR pathway include poly(ADP-ribose) polymerase (PARP) inhibitors (such as olaparib, rucaparib, niraparib, or talazoparib), ataxia telangiectasia mutated (ATM) protein inhibitors, ataxia telangiectasia and Rad3-related (ATR) protein inhibitors, checkpoint kinase 1 (Chk1) inhibitors, or combinations thereof.
  • In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or any embodiment, variation or aspect thereof (collectively, Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9)) or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of a PARP inhibitor (such as olaparib, rucaparib, niraparib, or talazoparib). In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the PARP inhibitor. In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the PARP inhibitor.
  • In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or any embodiment, variation or aspect thereof (collectively, Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9)) or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of an ATM protein inhibitor. In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (1b), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the ATM protein inhibitor. In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the ATM protein inhibitor.
  • In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or any embodiment, variation or aspect thereof (collectively, Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9)) or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of an ATR protein inhibitor. In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the ATR protein inhibitor. In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the ATR protein inhibitor.
  • In some embodiments, provided herein is a method of treating a disease in an individual comprising (a) administering an effective amount of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or any embodiment, variation or aspect thereof (collectively, Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9)) or a pharmaceutically acceptable salt thereof, and (b) administering an effective amount of an Chk1 inhibitor. In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered prior to, after, or simultaneously co-administered with the Chk1 inhibitor. In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a pharmaceutically acceptable salt thereof is administered 1 or more hours (such as 2 or more hours, 4 or more hours, 8 or more hours, 12 or more hours, 24 or more hours, or 48 or more hours) prior to or after the Chk1 inhibitor.
  • In another aspect, provided herein is a combination therapy in which a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof is coadministered (which may be separately or simultaneously) with one or more additional agents that are effective in stimulating immune responses to thereby further enhance, stimulate or upregulate immune responses in a subject. For example, provided is a method for stimulating an immune response in a subject comprising administering to the subject a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof and one or more immunostimulatory antibodies, such as an anti-PD-1 antibody, an anti-PD-L1 antibody and/or an anti-CTLA-4 antibody, such that an immune response is stimulated in the subject, for example to inhibit tumor growth. In one embodiment, the subject is administered a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof and an anti-PD-1 antibody. In another embodiment, the subject is administered a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof and an anti-PD-L1 antibody. In yet another embodiment, the subject is administered a compound of formula (I) or a salt thereof and an anti-CTLA-4 antibody. In another embodiment, the immunostimulatory antibody (e.g., anti-PD-1, anti-PD-L1 and/or anti-CTLA-4 antibody) is a human antibody. Alternatively, the immunostimulatory antibody can be, for example, a chimeric or humanized antibody (e.g., prepared from a mouse anti-PD-1, anti-PD-L1 and/or anti-CTLA-4 antibody).
  • In one embodiment, the present disclosure provides a method for treating a proliferative disease (e.g., cancer), comprising administering a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof and an anti-PD-1 antibody to a subject. In further embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof is administered at a subtherapeutic dose, the anti-PD-1 antibody is administered at a subtherapeutic dose, or both are administered at a subtherapeutic dose. In another embodiment, the present disclosure provides a method for altering an adverse event associated with treatment of a hyperproliferative disease with an immunostimulatory agent, comprising administering a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof and a subtherapeutic dose of anti-PD-1 antibody to a subject. In certain embodiments, the subject is human. In certain embodiments, the anti-PD-1 antibody is a human sequence monoclonal antibody.
  • In one embodiment, the present invention provides a method for treating a hyperproliferative disease (e.g., cancer), comprising administering a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof and an anti-PD-L1 antibody to a subject. In further embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof is administered at a subtherapeutic dose, the anti-PD-L1 antibody is administered at a subtherapeutic dose, or both are administered at a subtherapeutic dose. In another embodiment, the present invention provides a method for altering an adverse event associated with treatment of a hyperproliferative disease with an immunostimulatory agent, comprising administering a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof and a subtherapeutic dose of anti-PD-L1 antibody to a subject. In certain embodiments, the subject is human. In certain embodiments, the anti-PD-L1 antibody is a human sequence monoclonal antibody.
  • In certain embodiments, the combination of therapeutic agents discussed herein can be administered concurrently as a single composition in a pharmaceutically acceptable carrier, or concurrently as separate compositions each in a pharmaceutically acceptable carrier. In another embodiment, the combination of therapeutic agents can be administered sequentially. For example, an anti-CTLA-4 antibody and a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof can be administered sequentially, such as anti-CTLA-4 antibody being administered first and a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof second, or a compound of formula Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof being administered first and anti-CTLA-4 antibody second. Additionally or alternatively, an anti-PD-1 antibody and a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof can be administered sequentially, such as anti-PD-1 antibody being administered first and a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof second, or a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof being administered first and anti-PD-1 antibody second. Additionally or alternatively, an anti-PD-L1 antibody and a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof can be administered sequentially, such as anti-PD-L1 antibody being administered first and a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof second, or a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof being administered first and anti-PD-L1 antibody second.
  • Furthermore, if more than one dose of the combination therapy is administered sequentially, the order of the sequential administration can be reversed or kept in the same order at each time point of administration, sequential administrations can be combined with concurrent administrations, or any combination thereof.
  • Optionally, the combination of a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof can be further combined with an immunogenic agent, such as cancerous cells, purified tumor antigens (including recombinant proteins, peptides, and carbohydrate molecules), cells, and cells transfected with genes encoding immune stimulating cytokines.
  • A compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof can also be further combined with standard cancer treatments. For example, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof can be effectively combined with chemotherapeutic regimes. In these instances, it is possible to reduce the dose of other chemotherapeutic reagent administered with the combination of the instant disclosure (Mokyr et al. (1998) Cancer Research 58: 5301-5304). Other combination therapies with a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof include radiation, surgery, or hormone deprivation. Angiogenesis inhibitors can also be combined with a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof. Inhibition of angiogenesis leads to tumor cell death, which can be a source of tumor antigen fed into host antigen presentation pathways.
  • In another example, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof can be used in conjunction with anti-neoplastic antibodies. By way of example and not wishing to be bound by theory, treatment with an anti-cancer antibody or an anti-cancer antibody conjugated to a toxin can lead to cancer cell death (e.g., tumor cells) which would potentiate an immune response mediated by CTLA-4, PD-1, PD-L1 or a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof. In an exemplary embodiment, a treatment of a hyperproliferative disease (e.g., a cancer tumor) can include an anti-cancer antibody in combination with a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof and anti-CTLA-4 and/or anti-PD-1 and/or anti-PD-L1 antibodies, concurrently or sequentially or any combination thereof, which can potentiate anti-tumor immune responses by the host. Other antibodies that can be used to activate host immune responsiveness can be further used in combination with a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof.
  • In some embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof can be combined with an anti-CD73 therapy, such as an anti-CD73 antibody.
  • In yet further embodiments, a compound of Formula (I), (Ia), (Ia-1 to Ia-12), (Ib), (Ib-1 to Ib-9), (Ic) or (Ic-1 to Ic-9) or a salt thereof is administered in combination with another Wee1 inhibitor.
  • Dosing and Method of Administration
  • The dose of a compound administered to an individual (such as a human) may vary with the particular compound or salt thereof, the method of administration, and the particular disease, such as type and stage of cancer, being treated. In some embodiments, the amount of the compound or salt thereof is a therapeutically effective amount.
  • The effective amount of the compound may in one aspect be a dose of between about 0.01 and about 100 mg/kg. Effective amounts or doses of the compounds of the invention may be ascertained by routine methods, such as modeling, dose escalation, or clinical trials, taking into account routine factors, e.g., the mode or route of administration or drug delivery, the pharmacokinetics of the agent, the severity and course of the disease to be treated, the subject's health status, condition, and weight. An exemplary dose is in the range of about from about 0.7 mg to 7 g daily, or about 7 mg to 350 mg daily, or about 350 mg to 1.75 g daily, or about 1.75 to 7 g daily.
  • Any of the methods provided herein may in one aspect comprise administering to an individual a pharmaceutical composition that contains an effective amount of a compound provided herein or a salt thereof and a pharmaceutically acceptable excipient.
  • A compound or composition of the invention may be administered to an individual in accordance with an effective dosing regimen for a desired period of time or duration, such as at least about one month, at least about 2 months, at least about 3 months, at least about 6 months, or at least about 12 months or longer, which in some variations may be for the duration of the individual's life. In one variation, the compound is administered on a daily or intermittent schedule. The compound can be administered to an individual continuously (for example, at least once daily) over a period of time. The dosing frequency can also be less than once daily, e.g., about a once weekly dosing. The dosing frequency can be more than once daily, e.g., twice or three times daily. The dosing frequency can also be intermittent, including a ‘drug holiday’ (e.g., once daily dosing for 7 days followed by no doses for 7 days, repeated for any 14 day time period, such as about 2 months, about 4 months, about 6 months or more). Any of the dosing frequencies can employ any of the compounds described herein together with any of the dosages described herein.
  • The compounds provided herein or a salt thereof may be administered to an individual via various routes, including, e.g., intravenous, intramuscular, subcutaneous, oral and transdermal. A compound provided herein can be administered frequently at low doses, known as ‘metronomic therapy,’ or as part of a maintenance therapy using compound alone or in combination with one or more additional drugs. Metronomic therapy or maintenance therapy can comprise administration of a compound provided herein in cycles. Metronomic therapy or maintenance therapy can comprise intra-tumoral administration of a compound provided herein.
  • In one aspect, the invention provides a method of treating cancer in an individual by parenterally administering to the individual (e.g., a human) an effective amount of a compound or salt thereof. In some embodiments, the route of administration is intravenous, intra-arterial, intramuscular, or subcutaneous. In some embodiments, the route of administration is oral. In still other embodiments, the route of administration is transdermal.
  • The invention also provides compositions (including pharmaceutical compositions) as described herein for the use in treating, preventing, and/or delaying the onset and/or development of cancer and other methods described herein. In certain embodiments, the composition comprises a pharmaceutical formulation which is present in a unit dosage form.
  • Also provided are articles of manufacture comprising a compound of the disclosure or a salt thereof, composition, and unit dosages described herein in suitable packaging for use in the methods described herein. Suitable packaging is known in the art and includes, for example, vials, vessels, ampules, bottles, jars, flexible packaging and the like. An article of manufacture may further be sterilized and/or sealed.
  • Kits
  • The present disclosure further provides kits for carrying out the methods of the invention, which comprises one or more compounds described herein or a composition comprising a compound described herein. The kits may employ any of the compounds disclosed herein. In one variation, the kit employs a compound described herein or a salt thereof. The kits may be used for any one or more of the uses described herein, and, accordingly, may contain instructions for the treatment of cancer.
  • Kits generally comprise suitable packaging. The kits may comprise one or more containers comprising any compound described herein. Each component (if there is more than one component) can be packaged in separate containers or some components can be combined in one container where cross-reactivity and shelf life permit.
  • The kits may be in unit dosage forms, bulk packages (e.g., multi-dose packages) or sub-unit doses. For example, kits may be provided that contain sufficient dosages of a compound as disclosed herein and/or an additional pharmaceutically active compound useful for a disease detailed herein to provide effective treatment of an individual for an extended period, such as any of a week, 2 weeks, 3 weeks, 4 weeks, 6 weeks, 8 weeks, 3 months, 4 months, 5 months, 7 months, 8 months, 9 months, or more. Kits may also include multiple unit doses of the compounds and instructions for use and be packaged in quantities sufficient for storage and use in pharmacies (e.g., hospital pharmacies and compounding pharmacies).
  • The kits may optionally include a set of instructions, generally written instructions, although electronic storage media (e.g., magnetic diskette or optical disk) containing instructions are also acceptable, relating to the use of component(s) of the methods of the present invention. The instructions included with the kit generally include information as to the components and their administration to an individual.
  • The invention can be further understood by reference to the following examples, which are provided by way of illustration and are not meant to be limiting.
  • EXAMPLES Synthetic Examples Example S1. Synthesis of 2-allyl-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]-6-(1,2,3,4-tetrahydroisoquinolin-7-ylamino)pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02360
  • Step-1: Synthesis of ethyl 4-(2-allyl-2-tert-butoxycarbonyl-hydrazino)-2-methylsulfanyl-pyrimidine-5-carboxylate: To a stirred solution of ethyl 4-chloro-2-methylsulfanyl-pyrimidine-5-carboxylate (1.0 g, 4.29 mmol, 1.0 eq) and tert-butyl N-allyl-N-amino-carbamate (960 mg, 5.57 mmol, 1.3 eq) in THF (15 mL) was added DIPEA (1.86 mL, 10.72 mmol, 2.5 eq) and stirred at reflux for 16 h. Solvent was removed under reduced pressure. Residue was diluted with water (20 mL) and extracted with ethyl acetate (50 mL×3). The organic layer was washed with brine solution (20 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain crude product which was purified by flash chromatography to afford ethyl 4-(2-allyl-2-tert-butoxycarbonyl-hydrazino)-2-methylsulfanyl-pyrimidine-5-carboxylate (1.10 g, 53.6%) as an oily liquid.
  • LCMS: 369 [M+1]+
  • 1H NMR (400 MHz, CDCl3): δ 9.50 (br s, 1H), 8.70 (s, 1H), 5.90 (m, 1H), 5.20 (m, 2H), 4.35 (q, 2H), 4.20 (br s, 2H), 2.50 (s, 3H), 1.50-1.25 (m, 12H).
  • Step-2: Synthesis of 2-allyl-6-methylsulfanyl-1H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of ethyl 4-(2-allyl-2-tert-butoxycarbonyl-hydrazino)-2-methylsulfanyl-pyrimidine-5-carboxylate (3.0 g, 8.19 mmol, 1.0 eq) in CH2Cl2 (10 mL) was added TFA (10 mL) dropwise at 0° C. and allowed to stir at RT for 12 h. After completion of reaction, solvent was removed under reduced pressure. Residue was diluted with EtOH (20 mL) and 6N NaOH solution (10 mL) was added at 0° C. and allowed to stir at RT for 1 h. After completion of reaction, the mixture was acidified by using 6N HCl solution. EtOH was removed under reduced pressure; residue obtained was cooled to RT and extracted with chloroform (50 mL×3). The organic layer was washed with brine solution, dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford 2-allyl-6-methylsulfanyl-1H-pyrazolo[3,4-d]pyrimidin-3-one (1.40 g, 76%) as a yellow solid.
  • LCMS: 223 [M+1]+
  • Step-3: Synthesis of 2-allyl-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]-6-methylsulfanyl-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-allyl-6-methylsulfanyl-1H-pyrazolo[3,4-d]pyrimidin-3-one (200 mg, 0.90 mmol, 1.0 eq) and 2-(6-bromo-2-pyridyl)propan-2-ol (233 mg, 1.08 mmol, 1.20 eq) in 10 mL of dioxane were added copper iodide (171 mg, 0.90 mmol, 1.0 eq), potassium carbonate (186 mg, 1.35 mmol, 1.5 eq) and N,N′-dimethylethylenediamine (87 mg, 0.99 mmol, 1.1 eq) and stirred at 80° C. for overnight. After completion of reaction, solvent was removed under reduced pressure; residue was diluted with water and extracted with ethyl acetate (30 mL×3). Combined organic layer was washed with brine solution, dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography to afford 240 mg (74%) of 2-allyl-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]-6-methylsulfanyl-pyrazolo[3,4-d]pyrimidin-3-one.
  • LCMS: 358 [M+1]+
  • Step-4: Synthesis of tert-butyl 7-[[2-allyl-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]-3-oxo-pyrazolo[3,4-d]pyrimidin-6-yl]amino]-3,4-dihydro-1H-isoquinoline-2-carboxylate: To a stirred solution of 2-allyl-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]-6-methylsulfanyl-pyrazolo[3,4-d]pyrimidin-3-one (240 mg, 0.67 mmol, 1.0 eq) in 5 mL of toluene was added mCPBA (232 mg, 1.34 mmol, 2.0 eq) and allowed to stir at RT for 20 minutes. Tert-butyl 7-amino-3,4-dihydro-1H-isoquinoline-2-carboxylate (216 mg, 0.87 mmol, 1.3 eq) and DIPEA (260 mg, 2.10 mmol, 3.0 eq) were added and allowed to stir at RT for overnight. Solvent was removed under reduced pressure; residue was diluted with water and extracted with ethyl acetate (30 mL×3). Organic layer was washed with brine solution (20 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product which was purified by flash chromatography to afford 200 mg (53%) of tert-butyl 7-[[2-allyl-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]-3-oxo-pyrazolo[3,4-d]pyrimidin-6-yl]amino]-3,4-dihydro-1H-isoquinoline-2-carboxylate.
  • LCMS: 558 [M+1]+
  • Step-5: Synthesis of 2-allyl-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]-6-(1,2,3,4-tetrahydroisoquinolin-7-ylamino)pyrazolo[3,4-d]pyrimidin-3-one: tert-Butyl 7-[[2-allyl-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]-3-oxo-pyrazolo[3,4-d]pyrimidin-6-yl]amino]-3,4-dihydro-1H-isoquinoline-2-carboxylate (200 mg, 0.35 mmol, 1.0 eq) was dissolved in 5 mL of DCM and added TFA (1 mL) and allowed to stir at RT for 3 h. After completion of reaction, solvent was removed under reduced pressure, residue was diluted with aqueous NaHCO3 solution and extracted with ethyl acetate (30 mL×3). Combined organic layer was washed with brine solution (20 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product which was recrystallized with DCM and n-pentane to afford 4 mg (2.5%) of 2-allyl-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]-6-(1,2,3,4-tetrahydroisoquinolin-7-ylamino)pyrazolo[3,4-d]pyrimidin-3-one as white solid. This product was treated with 0.5 mL of 1.25 M ethanolic HCl. Solvent was removed under reduced pressure and product was freeze-dried to afford HCl salt of it.
  • LCMS: 458 [M+1]+
  • 1HNMR (400 MHz, DMSO-d6, HCl salt): δ 10.40 (br s, 1H), 9.50 (br s, 2H), 8.90 (s, 1H), 8.10 (t, J=7.8 Hz, 1H), 7.79 (d, J=8.2 Hz, 1H), 7.75 (br s, 1H) 7.65 (d, J=7.8 Hz, 1H), 7.50 (d, J=7.4 Hz, 1H), 7.18 (d, J=8.2 Hz, 1H), 5.72-5.62 (m, 1H), 5.0 (d, J=10.2 Hz, 1H), 4.80 (d, J=17.2 Hz, 1H), 4.70 (d, J=5.5 Hz, 2H), 4.25 (br s, 2H), 3.39-3.35 (m, 2H), 2.95 (t, J=5.9 Hz, 2H), 1.45 (s, 6H).
  • Example S2. Synthesis of 2-allyl-6-((2-(2-hydroxyacetyl)-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02361
  • Synthesis of 2-allyl-6-[[2-(2-hydroxyacetyl)-3,4-dihydro-1H-isoquinolin-7yl]amino]-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of glycolic acid (22 mg, 0.288 mmol, 1.2 eq) in 3 mL of DMF was added HATU (136 mg, 0.36 mmol, 1.5 eq) and stirred at RT for 5 minutes. 2-allyl-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]-6-(1,2,3,4-tetrahydroisoquinolin-7-ylamino)pyrazolo[3,4-d]pyrimidin-3-one (110 mg, 0.24 mmol, 1.0 eq) and DIPEA (62 mg, 0.48 mmol, 2.0 eq) were added and stirred at RT for 5 h. After completion of reaction, mixture was diluted with ice-cold water and extracted with ethyl acetate (50 mL×2). Combined organic layer was washed with brine solution (20 mL×4), dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain crude product which was purified by reverse phase chromatography to afford 15 mg (12%) of 2-allyl-6-[[2-(2-hydroxyacetyl)-3,4-dihydro-1H-isoquinolin-7-yl]amino]-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]pyrazolo[3,4-d]pyrimidin-3-one as white solid.
  • LCMS: 516 [M+1]+
  • 1H NMR (400 MHz, DMSO-d6): δ 10.30 (br s, 1H), 8.90 (s, 1H), 8.15-8.07 (m, 1H), 7.81-7.79 (m, 2H), 7.64-7.62 (m, 1H), 7.43-7.36 (m, 1H), 7.10 (d, J=8.2 Hz, 1H), 5.71-5.61 (m, 1H), 5.35 (s, 1H), 5.0 (d, J=10.2 Hz, 1H), 4.80 (d, J=16.4 Hz, 1H), 4.70-4.57 (m, 5H), 4.22-4.19 (m, 2H), 3.71-3.59 (m, 1H), 3.59 (t, J=5.5 Hz, 1H), 2.95-2.80 (m, 2H), 1.45 (s, 6H).
  • Example S3. Synthesis of 2-allyl-6-((2-(2-hydroxy-2-methylpropanoyl)-4,4-dimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02362
  • Step-1: Synthesis of tert-butyl 7-[[2-allyl-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]-3-oxo-pyrazolo[3,4-d]pyrimidin-6-yl]amino]-4,4-dimethyl-1,3-dihydroisoquinoline-2-carboxylate: To a stirred solution of 2-allyl-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]-6-methylsulfanyl-pyrazolo[3,4-d]pyrimidin-3-one (500 mg, 1.40 mmol, 1.0 eq) in 5 mL of toluene was added mCPBA (172 mg, 2.80 mmol, 2.0 eq) and allowed to stir at RT for 30 minutes. Tert-butyl 7-amino-4, 4-dimethyl-1, 3-dihydroisoquinoline-2-carboxylate (424 mg, 1.69 mmol, 1.20 eq) and DIPEA (541 mg, 4.2 mmol, 3.0 eq) were added and allowed to stir at RT for overnight. After completion of reaction, solvent was removed under reduced pressure. Residue was diluted with water and extracted with ethyl acetate (50 mL×2). Combined organic layer was washed with brine solution (20 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product which was purified by flash chromatography to afford 340 mg (41.4%) of tert-butyl 7-[[2-allyl-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]-3-oxo-pyrazolo[3,4-d]pyrimidin-6-yl]amino]-4,4-dimethyl-1,3-dihydroisoquinoline-2-carboxylate.
  • LCMS: 586 [M+1]+.
  • Step-2: Synthesis of 2-allyl-6-[(4,4-dimethyl-2,3-dihydro-1H-isoquinolin-7-yl)amino]-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]pyrazolo[3,4-d]pyrimidin-3-one: Tert-butyl 7-[[2-allyl-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]-3-oxo-pyrazolo[3,4-d]pyrimidin-6-yl]amino]-4,4-dimethyl-1,3-dihydroisoquinoline-2-carboxylate (340 mg, 0.581 mmol, 1.0 eq) was dissolved in 5 mL of DCM and added TFA (1 mL) and allowed to stir at RT for 3 h. After completion of reaction, solvent was removed under reduced pressure to obtain crude. Compound was recrystallized with diethyl ether and n-pentane to afford 80 mg (28.3%) of 2-allyl-6-[(4,4-dimethyl-2,3-dihydro-1H-isoquinolin-7-yl)amino]-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]pyrazolo[3,4-d]pyrimidin-3-one as white solid.
  • LCMS: 486 [M+1]+
  • Step-3: Synthesis of 2-allyl-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]-6-[[2-(2-hydroxy-2-methyl-propanoyl)-4,4-dimethyl-1,3-dihydroisoquinolin-7-yl]amino]pyrazolo[3,4 d]pyrimidin-3-one: To a stirred solution of 2-hydroxy-2-methyl-propanoic acid (20.60 mg, 0.197 mmol, 1.2 eq) in 2 mL of DMF was added HATU (93.48 mg, 0.246 mmol, 1.5 eq) and stirred at RT for 5 minutes 2-allyl-6-[(4,4-dimethyl-2,3-dihydro-1H-isoquinolin-7-yl)amino]-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]pyrazolo[3,4-d]pyrimidin-3-one (80 mg, 0.164 mmol, 1.0 eq) and DIPEA (42.31 mg, 0.328 mmol, 2.0 eq) were added and stirred at RT for 5 h. After completion of reaction, mixture was diluted with ice-cold water and extracted with ethyl acetate (50 mL×2). Combined organic layer was washed with brine solution (20 mL×4), dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain crude product which was purified by reverse phase chromatography to afford 16.8 mg (18%) 2-allyl-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]-6-[[2-(2-hydroxy-2-methyl-propanoyl)-4,4-dimethyl-1,3-dihydroisoquinolin-7-yl]amino]pyrazolo[3,4-d]pyrimidin-3-one as white solid.
  • LCMS: 572 [M+1]+
  • 1H NMR (400 MHz, DMSO-d6): δ 10.30 (br s, 1H), 8.90 (s, 1H), 8.20 (br s, 1H), 7.79 (d, J=7.9 Hz, 2H), 7.63 (d, J=7.9 Hz, 1H), 7.40-7.39 (m, 1H), 7.33 (d, d, J=8.0 Hz 1H), 5.72-5.62 (m, 1H), 5.17 (br s, 1H), 5.01-4.98 (m, 2H), 4.84-4.79 (m, 2H), 4.7-4.89 (m, 2H), 1.46 (s, 6H), 1.37 (s, 6H), 1.2 (s, 6H).
  • Example S4. Synthesis of 2-allyl-6-[(4,4-dimethyl-2,3-dihydro-JH-isoquinolin-7-yl)amino]-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02363
  • Synthesis of 2-allyl-6-[(4,4-dimethyl-2,3-dihydro-1H-isoquinolin-7-yl)amino]-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]pyrazolo[3,4-d]pyrimidin-3-one: Tert-butyl 7-[[2-allyl-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]-3-oxo-pyrazolo[3,4-d]pyrimidin-6-yl]amino]-4,4-dimethyl-1,3-dihydroisoquinoline-2-carboxylate (60 mg, 0.102 mmol, 1.0 eq) was dissolved in 1 mL of ethanol and added 2M HCl in methanol and allowed to stir at RT for 16 h. After completion of reaction, solvent was removed under reduced pressure to obtain crude. Product was recrystallized with diethyl ether and n-pentane and dried under reduced pressure. Product was freeze-dried to afford 48 mg (90%) HCl salt of 2-allyl-6-[(4,4-dimethyl-2,3-dihydro-1H-isoquinolin-7-yl)amino]-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]pyrazolo[3,4-d]pyrimidin-3-one as off-white solid.
  • LCMS: 486 [M+1]+
  • 1HNMR (400 MHz, DMSO-d6, HCl Salt): δ10.39 (hr s, 1H), 9.3 (hr s, 1H), 8.90 (s, 1H), 8.07 (t, J=7.89 Hz, 1H), 7.8 (d, J=7.45 Hz 1H), 7.71 (hr s, 1H), 7.64 (d, J=7.89 Hz, 1H), 7.53 (dd, J=8.77, 2.19 Hz, 1H), 7.44 (d, J=8.33 Hz, 1H), 5.67 (dd, J=17.10, 10.52 Hz, 1H), 5.0 (dd, J=10.30, 1.10 Hz, 1H), 4.80 (dd, J=17.10, 1.32 Hz, 1H), 4.70-4.62 (d, J=5.70 Hz, 2H), 4.23 (s, 2H), 3.23 (d, J=5.70 Hz, 2H), 1.46 (s, 6H), 1.34 (s, 6H).
  • Example S5. Synthesis of 2-allyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((2,4,4-trimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02364
  • Synthesis of 2-allyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((2,4,4-trimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-allyl-6-((4,4-dimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (110 mg, 0.226 mmol, 1.0 eq) and 37% formaldehyde in water (1.2 mL) in 5 mL of dichloroethane was added dropwise acetic acid (0.068 mL, 1.132 mmol, 5.0 eq) at 0° C. The resulting mixture was stirred at RT for 1 h followed by addition of NaCNBH4 (56 mg, 0.904 mmol, 4.0 eq) at 0° C. The resulting mixture was stirred at RT for 1 h. The progress of reaction was monitored by LCMS. After completion of reaction, solvent was removed under reduced pressure. Residue was basified with saturated solution of NaHCO3 (100 mL) and extracted with ethyl acetate (100 mL×2). The combined organic layer was washed with water (50 mL) and brine (50 mL), dried over Na2SO4 and concentrated under reduced pressure. Crude was purified by reverse phase chromatography to afford 8 mg free base of 2-allyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((2,4,4-trimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one as brown solid.
  • LCMS: 501 [M+1]+
  • 1H NMR (400 MHz, CDCl3): δ 10.21 (s, 1H), 8.84 (br s, 1H), 8.03-8.0 (m, 1H), 7.73 (d, J=7.89 Hz, 1H), 7.66 (d, J=7.89 Hz, 1H), 7.57-7.52 (m, 1H), 7.40 (d, J=8.77 Hz, 1H), 7.30 (d, J=8.33 Hz, 1H), 5.71-5.61 (m, 1H), 5.35 (br s, 1H), 4.99 (d, J=10.96 Hz, 1H), 4.81 (d, J=18 Hz, 1H), 4.68 (d, J=6.58 Hz, 2H), 2.46-2.31 (m, 3H), 1.60 (s, 3H), 1.49 (br s, 6H), 1.25 (br s, 6H).
  • Example S6. Synthesis of 2-allyl-6-[[2-(2-hydroxyacetyl)-4,4-dimethyl-1,3-dihydroisoquinolin-7-yl]amino]-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02365
  • Synthesis of 2-allyl-6-[[2-(2-hydroxyacetyl)-4,4-dimethyl-1,3-dihydroisoquinolin-7-yl]amino]-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-hydroxyacetic acid (23.50 mg, 0.309 mmol, 1.5 eq) in 2 mL of DMF were added EDC.HCl (39.34 mg, 0.206 mmol, 1.0 eq) & HOBT (2.78 mg, 0.0206 mmol, 0.1 eq) and stirred at RT for 5 min. 2-Allyl-6-[(4,4-dimethyl-2,3-dihydro-1H-isoquinolin-7-yl)amino]-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]pyrazolo[3,4-d]pyrimidin-3-one (100 mg, 0.206 mmol, 1.0 eq) and DIPEA (79.72 mg, 0.618 mmol, 3.0 eq) were added and stirred at RT for 16 h. The progress of reaction was monitored by LCMS. After completion of reaction, mixture was diluted with ice-cold water and extracted with ethyl acetate (50 mL×2). Combined organic layers was washed with brine solution (20 mL×4), dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain crude product which was purified by reverse phase chromatography to afford 8 mg (7%) of 2-allyl-6-[[2-(2-hydroxyacetyl)-4,4-dimethyl-1,3-dihydroisoquinolin-7-yl]amino]-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]pyrazolo[3,4-d]pyrimidin-3-one as off white solid.
  • LCMS: 544 [M+1]+
  • 1H NMR (400 MHz, DMSO-d6): δ 10.30 (br s, 1H), 8.90 (s, 1H), 8.18 (br s, 1H), 7.83-7.79 (m, 1H), 7.65-7.62 (m, 1H), 7.45-7.23 (m, 2H), 5.75-5.6 (m, 1H), 5.35 (br s, 1H), 5.0 (dd, J=10.41, 1.21 Hz, 1H), 4.84-4.79 (m, 1H), 4.71-4.62 (m, 3H), 4.6 (s, 1H), 4.28-4.26 (m, 1H), 4.22-4.20 (m, 1H), 3.52-3.5 (m, 2H), 1.46 (s, 6H), 1.21 (d, J=11.40 Hz, 6H).
  • Example S7. Synthesis of 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02366
    Figure US20230109104A1-20230406-C02367
  • Step-1: Synthesis of tert-butyl (1,3-dioxoisoindolin-2-yl)(ethyl)carbamate: To a stirred solution of tert-butyl (1,3-dioxoisoindolin-2-yl)carbamate (5.0 g, 19.064 mmol, 1.0 eq), in CH3CN (100 mL) was added ethyliodide (1.83 mL, 22.877 mmol, 1.2 eq), BnEt3N+Cl (435 mg, 1.906 mmol, 0.1 eq) and K2CO3 (5.27 g, 38.128 mmol, 2.0 eq) at RT. The resulting mixture was heated at 55° C. for 12 h. The progress of reaction was monitored by LCMS. The reaction mixture was diluted with water (50 mL), extracted with EtOAc (2×100 mL). The combined organic layers were washed with water (50 mL), with brine (50 mL), dried over Na2SO4, concentrated and purified by recrystallization to afford the desired compound, tert-butyl (1,3-dioxoisoindolin-2-yl)(ethyl)carbamate (5.21 g, 94.19%) as light yellow solid.
  • LCMS: 291.1[M+1]+
  • Step-2: Synthesis of tert-butyl 1-ethylhydrazine-1-carboxylate: To a stirred solution of tert-butyl (1,3-dioxoisoindolin-2-yl)(ethyl)carbamate. (5.0 g, 17.22 mmol, 1.0 eq), in CH2Cl2 (100 mL) was added NH2NH2.H2O (2.16 mL, 34.44 mmol, 2.0 eq) at RT. The resulting mixture was stirred at RT for 12 h, after which formation of white precipitates was obr served. Progress of the reaction was monitored by 1H NMR. The reaction mixture was filtered and the filtrate was concentrated and purified by n-pentane to afford the desired compound, tert-butyl 1-ethylhydrazine-1-carboxylate (2.1 g, 75.26%) as light yellow liquid.
  • 1H NMR (400 MHz, CDCl3): δ 3.95 (br s, 2H), 3.40 (q, 2H), 1.47 (s, 9H), 1.12 (t, 3H).
  • Step-3: Synthesis of ethyl 4-(2-(tert-butoxycarbonyl)-2-ethylhydrazinyl)-2-(methylthio) pyrimidine-5-carboxylate: To a stirred solution of ethyl 4-chloro-2-methylsulfanyl-pyrimidine-5-carboxylate (2.0 g, 8.595 mmol, 1.0 eq) and tert-butyl 1-ethylhydrazine-1-carboxylate (2.06 g, 12.893 mmol, 1.5 eq) in (50 mL) of THF was added DIPEA (3.73 mL, 21.487 mmol, 2.5 eq) and stirred at 60° C. for 16 h. Solvent was removed under reduced pressure. Residue was diluted with water (50 mL) and extracted with EtOAc (50 mL×2). Organic layer was washed with brine solution (50 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford the desired compound, ethyl 4-(2-(tert-butoxycarbonyl)-2-ethylhydrazinyl)-2-(methylthio)pyrimidine-5-carboxylate (2.91 g, 95.03%) as light yellow liquid.
  • LCMS: 356.0 [M+1]+
  • Step-4: Synthesis of 2-ethyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of ethyl 4-(2-(tert-butoxycarbonyl)-2-ethylhydrazinyl)-2-(methylthio) pyrimidine-5-carboxylate (2.90 g, 8.136 mmol, 1.0 eq) in CH2C1 (50 mL) was added TFA (8.136 mL) dropwise at 0° C. and allowed to stir at RT for 12 h. After completion of reaction, solvent was removed under reduced pressure. Residue was diluted with EtOH (20 mL). 6N NaOH solution (10 mL) was added in to above at 0° C. and allowed to stir at RT for 1 h. After completion of reaction, mixture was acidified by using 6N HCl solution. EtOH was removed under reduced pressure; residue obtained was cooled to RT and extracted with chloroform (100 mL×3). Organic layer was washed with brine solution, dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford the desired compound, 2-ethyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (1.15 g, 67.25%) as yellow solid.
  • LCMS: 211.1 [M+1]+
  • Step-5: Synthesis of 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-ethyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (250 mg, 1.189 mmol, 1.0 eq) and 2-(6-bromo-2-pyridyl)propan-2-ol (308 mg, 1.426 mmol, 1.20 eq) in dioxane (10 mL) were added K2CO3 (328 mg, 2.378 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min followed by addition of CuI (45 mg, 0.237 mmol, 0.2 eq), and DIPEA (42 mg, 0.475 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 90° C. for 12 h. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography to afford the desired compound, 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (170 mg, 40.24%) as off white viscous.
  • LCMS: 346.2 [M+1]+
  • Step-6: Synthesis of tert-butyl 7-((2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (165 mg, 0.477 mmol, 1.0 eq) in toluene (10 mL) was added m-CPBA (165 mg, 0.954 mmol, 2.0 eq) and allowed to stir at RT for 30 min. Tert-butyl 7-amino-3,4-dihydro-1H-isoquinoline-2-carboxylate (131 mg, 0.525 mmol, 1.3 eq) and DIPEA (0.331 mL, 1.908 mmol, 4.0 eq) were added and allowed to stir at RT for 12 h. The formation of precipitate was obr served which was filtered to afford the desired compound, tert-butyl 7-((2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo [3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (110 mg, 42.30%) as off white solid.
  • LCMS: 546.4 [M+1]+
  • Step-7: Synthesis of 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: tert-butyl 7-((2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (105 mg, 0.192 mmol, 1.0 eq) was dissolved in dioxane (2 mL) and added 4M HCl (2 mL) and allowed to stir at RT for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (65 mg, 75.82%) as HCl salt yellow solid.
  • LCMS: 446.3 [M+1]+
  • 1H NMR (400 MHz, DMSO-d6): δ 10.35 (brs, 1H), 9.36 (brs, 2H), 8.88 (s, 1H), 8.11 (t, J=7.89 Hz, 1H), 7.85 (d, J=7.89 Hz, 1H), 7.75 (brs, 1H), 7.66 (d, J=7.89 Hz, 1H), 7.51 (d, J=8.33 Hz, 1H), 7.18 (d, J=8.77 Hz, 1H), 4.26 (brs, 2H), 4.05 (d, J=7.02 Hz, 2H), 3.36 (brs, 2H), 2.89-3.04 (m, 2H), 1.44 (s, 6H), 0.98 (t, J=7.02 Hz, 3H).
  • Example S8. Synthesis of 2-allyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(1,2,3,4-tetrahydroisoquinolin-6-ylamino)-JH-pyrazolo[3,4-d]pyrimidin-3(2H)-one
  • Figure US20230109104A1-20230406-C02368
  • Step-1: Synthesis of tert-butyl 6-(2-allyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 2-allyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(methylthio)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one (300 mg, 0.84 mmol, 1.0 eq) in toluene (5 mL) was added m-CPBA (361 mg, 2.10 mmol, 2.5 eq) and allowed to stir at RT for 30 min. tert-Butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (208 mg, 0.84 mmol, 1.0 eq) and DIPEA (433 mg, 3.36 mmol, 4.0 eq) were added and allowed to stir at RT for 12 h. Progress of reaction was monitored by LCMS. After completion of reaction, precipitated compound was filtered off, washed with toluene (3 mL) and dried under reduced pressure to obtain tert-butyl 6-(2-allyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (220 mg, 47.00%).
  • LCMS: 558 [M+1]+
  • Step-2: Synthesis of 2-allyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(1,2,3,4-tetrahydroisoquinolin-6-ylamino)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one: tert-Butyl 6-(2-allyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (220 mg, 0.395 mmol, 1.0 eq) was dissolved in 4N HCl in dioxane (5 mL) at 0° C. Reaction was stirred at RT for 30 min. Progress of reaction was monitored by LCMS. After completion of reaction, precipitated compound was filtered off and dried under reduced pressure. Solid was triturated with diethyl ether (10 mL), filtered off and dried under reduced pressure to obtain 2-allyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(1,2,3,4-tetrahydroisoquinolin-6-ylamino)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one (150 mg, 77.3%) as HCl salt.
  • LCMS: 448 [M+1]+
  • 1H NMR (400 MHz, DMSO-d6, HCl salt): δ 10.38 (br s, 1H), 9.25 (s, 1H), 8.90 (s, 1H), 8.05 (t, J=7.89 Hz, 1H), 7.80 (d, J=8.33 Hz, 2H), 7.65 (d, J=7.45 Hz, 1H), 7.50 (d, J=8.33 Hz, 1H), 7.20 (d, J=8.33 Hz, 1H), 5.72-5.62 (m, 1H), 5.00 (d, J=9.21 Hz, 1H), 4.90 (d, J=17.10 Hz, 1H), 4.70 (d, J=6.14 Hz, 2H), 4.25-4.2 (m, 2H), 3.41-3.36 (m, 2H), 3.02-2.99 (m, 2H), 1.50 (s, 6H).
  • Example S9. Synthesis of 2-allyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((2-methyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02369
  • Synthesis of 2-allyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((2-methyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-allyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (170 mg, 0.3715 mmol, 1.0 eq) and HCHO (0.1 mL) in dichloroethane (7 mL) was added acetic acid (111.45 mg, 1.85 mmol, 5.0 eq) dropwise at 0° C. The resulting mixture was stirred at RT for 1 h followed by addition of NaCNBH3 (70.2 mg, 1.1145 mmol, 3.0 eq) at 0° C. The resulting mixture was stirred at RT for 1 h. The progress of reaction was monitored by LCMS. The reaction mixture was concentrated, basified with saturated solution of NaHCO3 (100 mL) extracted with EtOAc (2×100 mL). The combined organic layer extracts were washed with water (50 mL), with brine (50 mL) dried over Na2SO4 and concentrated under reduced and purified by reversed phase chromatography to afford the desired compound, 2-allyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((2-methyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (0.023 g, 13.14%) as off white solid.
  • LCMS: 472 [M+1]+
  • 1H NMR (400 MHz, DMSO-d6): δ 10.22 (br s, 1H), 8.87 (s, 1H), 8.15 (s, 1H), 8.01 (t, J=7.89 Hz, 1H), 7.76 (d, J=8.33 Hz, 1H), 7.64 (d, J=7.89 Hz, 1H), 7.57 (br s, 1H), 7.40 (d, J=8.77 Hz, 1H), 7.05 (d, J=8.77 Hz, 1H), 5.70-5.64 (m, 1H), 5.36-5.29 (m, 1H), 4.99 (d, J=10.52 Hz, 1H), 4.81 (d, 1H), 4.68 (d, J=4.82 Hz, 2H), 3.49 (br s, 2H), 2.77 (br s, 2H), 2.61 (br s, 2H), 2.38 (s, 3H), 1.46 (s, 6H).
  • Example S10. Synthesis of 1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02370
    Figure US20230109104A1-20230406-C02371
  • Step-1: Synthesis of tert-butyl (1,3-dioxoisoindolin-2-yl)carbamate: A mixture of phthalic anhydride (15 g, 101.2 mmol) and tert-butylcarbazate (13.3 g, 101.2 mmol) in toluene (200 mL) was refluxed for 3 h. The mixture was allowed to cool and filtered. The white solid product obtained was dissolved in ethyl acetate and washed with water and brine (100 mL×3). The organic layer was dried over anhydrous sodium sulphate and concentrated under reduced pressure to give tert-butyl (1,3-dioxoisoindolin-2-yl)carbamate (15 g, 56%) as a white solid.
  • LCMS: 263.1 [M+1]+
  • Step-2: Synthesis of tert-butyl (1,3-dioxoisoindolin-2-yl)(isopropyl)carbamate: To a solution of tert-butyl 1,3-dioxoisoindolin-2-ylcarbamate (10.0 g, 38.12 mmol) in CH3CN (100 mL), K2CO3 (21.07 g, 152.5 mmol), triethylbenzylammonium chloride (1.736 g, 7.62 mmol) was added under stirring. Further, 2-bromopropane (5.62 g, 45.75 mmol) was added dropwise under stirring. The reaction was purged with nitrogen and mixture was heated at 60° C. 12 h. After completion the reaction, mixture was cooled to RT and solvent was evaporated. The residue was diluted with water, and extracted with EtOAc (100 mL×5). The combined extracts were washed with brine, dried over sodium sulphate, and concentrated to give the title compound as a white solid (9.88 g, 85%).
  • LCMS: 305.1 [M+1]+
  • Step-3: Synthesis of tert-butyl 1-isopropylhydrazine-1-carboxylate: To a solution of tert-butyl (1,3-dioxoisoindolin-2-yl)(isopropyl)carbamate (9.88 g, 32.5 mmol) in dry CH2Cl2 (50 ML), NH2NH2.H2O (3.25 g, 65.0 mmol) was added dropwise at 0° C. under inert atmosphere. The resulting mixture was stirred at RT for 72 h. The progress of the reaction was monitored by TLC. After completion, the solvent was removed under reduced pressure and the resulting mass was washed with pentane. The resulting precipitate formed if any was removed by filtration. The filtrate was concentrated to give the title compound as yellow oil (2.62 g, 46%).
  • LCMS: 175.1 [M+1]+
  • Step-4: Synthesis of ethyl 4-(2-(tert-butoxycarbonyl)-2-isopropylhydrazinyl)-2-(methylthio)pyrimidine-5-carboxylate: To a stirred solution of ethyl 4-chloro-2-methylsulfanyl-pyrimidine-5-carboxylate (2 g, 11.47 mmol, 1.0 eq) and tert-butyl 1-isopropylhydrazine-1-carboxylate (2.66 g, 11.47 mmol, 1.0 eq) in dry THF (50 mL), DIPEA (4.99 mL, 28.69 mmol, 2.5 eq) was added and the resulting solution was stirred at reflux temperature for 16 h. After completion of reaction, solvent was removed under reduced pressure. Residue was diluted with water (20 mL) and extracted with ethyl acetate (20 mL×3). The combined organic layer was washed with brine solution (10 mL×2), dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain crude product which was purified by flash chromatography (MeOH:CH2Cl2 1-10%) to afford ethyl 4-(2-(tert-butoxycarbonyl)-2-isopropylhydrazinyl)-2-(methylthio)pyrimidine-5-carboxylate (1.2 g, 38%) as oily liquid.
  • LCMS: 371.1 [M+1]+
  • Step-5: Synthesis of 2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of ethyl 4-(2-(tert-butoxycarbonyl)-2-isopropylhydrazinyl)-2-(methylthio)pyrimidine-5-carboxylate (1 g, 2.96 mmol, 1.0 eq) in CH2Cl2 (10 mL) was added TFA (2.0 mL) dropwise at 0° C. and the resulting solution was allowed to stir at RT for 12 h. After completion of reaction, solvent was removed under reduced pressure. The residue was diluted with EtOH (10 mL) and 6N NaOH solution (3 mL) was added at 0° C. and the resulting solution was allowed to stir at RT for 1 h. After completion of reaction, mixture was acidified with 6N HCl solution (3 mL). Solvent was removed under reduced pressure; residue obtained was cooled to RT and extracted with CH2Cl2 (10 mL×3). The combined organic layer was washed with brine solution, dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford 2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (0.27 g, 45%) as yellow solid.
  • LCMS: 225 [M+1]+
  • Step-6: Synthesis of 1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (0.26 g, 1.16 mmol, 1.0 eq) and 2-(6-bromo-2-pyridyl)propan-2-ol (0.3 g, 1.39 mmol, 1.20 eq) in dioxane (10 mL) were added CuI (0.044 g, 0.23 mmol, 1.0 eq), K2CO3 (0.32 g, 2.32 mmol, 2 eq) and DIPEA (0.041 g, 0.46 mmol, 0.4 eq) and stirred at 80° C. 12 h. After completion of reaction, solvent was removed under reduced pressure; residue was diluted with water and extracted with ethyl acetate (10 mL×3). Combined organic layer was washed with brine solution, dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography to afford 1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (55 mg, 13+%) as yellow oil.
  • LCMS: 360 [M+1]+
  • Step-7: Synthesis of tert-butyl 7-((1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (50 mg, 0.14 mmol, 1.0 eq) in toluene (5 mL) was added m-CPBA (48.0 mg, 027 mmol, 2.0 eq) and allowed to stir at RT for 30 min. Further, tert-butyl 7-amino-3,4-dihydro-1H-isoquinoline-2-carboxylate (38 mg, 0.15 mmol, 1.1 eq) and DIPEA (0.09 mL, 0.55 mmol, 4 eq) were added and allowed to stir at RT for 12 h. After completion of reaction, solvent was removed under reduced pressure; residue was diluted with water and extracted with ethyl acetate (5 mL×3). The combined organic layer was washed with brine solution (10 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford crude product which was purified by flash chromatography to afford tert-butyl 7-[[2-allyl-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]-3-oxo-pyrazolo[3,4-d]pyrimidin-6-yl]amino]-3,4-dihydro-1H-isoquinoline-2-carboxylate (28 mg, 36%).
  • LCMS: 560 [M+1]+
  • Step-8: Synthesis of 1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: 4 M HCl in dioxane (1 mL) was added to tert-butyl 7-[[2-allyl-1-[6-(1-hydroxy-1-methyl-ethyl)-2-pyridyl]-3-oxo-pyrazolo[3,4-d]pyrimidin-6-yl]amino]-3,4-dihydro-1H-isoquinoline-2-carboxylate (28 mg, 0.05 mmol, 1.0 eq) and the resulting solution was allowed to stir at RT for 3 h. After completion of reaction, solvent was removed under reduced pressure, and the resulting solid was filtered and washed with ether, dried to afford HCl salt of 1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (15 mg, 60%) as a white solid.
  • LCMS: 460.2 [M+1]+
  • 1H NMR (400 MHz, DMSO-d6, FB): δ 10.1 (br s, 1H), 8.7 (br s, 1H), 8.07-8.01 (m, 1H), 7.74 (d, J=7.89 Hz, 1H), 7.67 (d, J=7.45 Hz, 1H), 7.51 (br s, 1H), 7.35-7.33 (m, 1H), 6.98 (d, J=8.77 Hz, 1H), 5.33 (br s, 1H), 4.07-4.22 (m, 1H), 3.80 (s, 2H), 2.93 (t, J=5.92 Hz, 2H), 2.59-2.70 (m, 2H), 1.44 (s, 6H), 1.37 (d, J=7.02 Hz, 6H).
  • Example S11. Synthesis of 2-(cyclopropylmethyl)-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride
  • Figure US20230109104A1-20230406-C02372
  • Step-1: tert-butyl (cyclopropylmetyl)(1,3-dioxoisoindolin-2-yl)carbamate: To a stirred solution of tert-butyl (1,3-dioxoisoindolin-2-yl)carbamate (10.0 g, 38.12 mmol, 1.0 eq), in CH3CN (100 mL) was added (bromomethyl)cyclopropane (4.44 mL, 45.75 mmol, 1.2 eq), BnEt3N+Cl (1.73 g, 7.624 mmol, 0.2 eq) and K2CO3 (21.07 g, 152.48 mmol, 4.0 eq) at RT. The resulting mixture was heated at 80° C. for 12 h. The progress of reaction was monitored by LCMS. The reaction mixture was diluted with water (100 mL), extracted with EtOAc (2×200 mL). The combined organic layers were washed with water (100 mL), with brine (50 mL), dried over Na2SO4, concentrated and purified by recrystallization to afford the desired compound, tert-butyl (cyclopropylmethyl)(1,3-dioxoisoindolin-2-yl)carbamate (6.8 g, 56.38%) as light yellow liquid.
  • LCMS: 317.1 [M+1]+
  • Step-2: tert-butyl 1-(cyclopropylmethyl)hydrazine-1-carboxylate: To a stirred solution of tert-butyl (cyclopropylmethyl)(1,3-dioxoisoindolin-2-yl)carbamate (6.8 g, 21.49 mmol, 1.0 eq), in CH2Cl2 (100 mL) was added NH2NH2.H2O (2.04 mL, 42.98 mmol, 2.0 eq) at RT. The resulting mixture was stirred at RT for 12 h, formation of white precipitates was obr served. The progress of reaction was monitored by 1HNMR. The reaction mixture was filtered and the filtrate was concentrated and purified by pentane to afford the desired compound, tert-butyl 1-(cyclopropylmethyl)hydrazine-1-carboxylate (3.90 g, 97.5%) as light yellow liquid.
  • LCMS: 187.1 [M+1]+
  • Step-3: Synthesis of ethyl 4-(2-(tert-butoxycarbonyl)-2-(cyclopropylmethyl)hydrazinyl)-2-(methylthio)pyrimidine-5-carboxylate: To a stirred solution of ethyl 4-chloro-2-methylsulfanyl-pyrimidine-5-carboxylate (3.48 g, 14.99 mmol, 1.0 eq) and tert-butyl 1-(cyclopropylmethyl)hydrazine-1-carboxylate (3.90 g, 20.98 mmol, 1.5 eq) in THF (36 mL) was added DIPEA (6.48 mL, 37.47 mmol, 2.5 eq) and stirred at 80° C. for 16 h. Solvent was removed under reduced pressure. Residue was diluted with water (50 mL) and extracted with EtOAc (50 mL×2). Organic layer was washed with brine solution (50 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford the desired compound, ethyl 4-(2-(tert-butoxycarbonyl)-2-(cyclopropylmethyl)hydrazinyl)-2-(methylthio)pyrimidine-5-carboxylate (3.0 g, 52.34%) as light yellow solid.
  • LCMS: 237.0 [M+1]+
  • Step-4: Synthesis of 2-(cyclopropylmethyl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of ethyl 4-(2-(tert-butoxycarbonyl)-2-(cyclopropylmethyl)hydrazinyl)-2-(methylthio)pyrimidine-5-carboxylate (3.0 g, 7.84 mmol, 1.0 eq) in CH2Cl2 (50 mL) was added TFA (7.84 mL) dropwise at 0° C. and allowed to stir at RT for 12 h. After completion of reaction, solvent was removed under reduced pressure. Residue was diluted with EtOH (20 mL). 6N NaOH solution (10 mL) was added in to above solution at 0° C. and allowed to stir at RT for 1 h. After completion, the reaction mixture was acidified by using 6N HCl solution. EtOH was removed under reduced pressure; residue obtained was cooled to RT and extracted with chloroform (100 mL×3). Organic layer was washed with brine solution, dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford the desired compound, 2-(cyclopropylmethyl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (1.0 g, 54.05%) as yellow solid.
  • LCMS: 237.1 [M+1]+
  • Step-5: Synthesis of 2-(cyclopropylmethyl)-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-(cyclopropylmethyl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (1.0 g, 4.23 mmol, 1.0 eq) and 2-(6-bromo-2-pyridyl)propan-2-ol (1.09 g, 5.07 mmol, 1.20 eq) in dioxane (10 mL) were added K2CO3 (1.16 g, 8.46 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min followed by addition of CuI (161 mg, 0.84 mmol, 0.2 eq), and DIPEA (149 mg, 1.69 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 90° C. for 12 h. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). Combined organic layer was washed with water (50 mL) and brine solution (50 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography to afford the desired compound, 2-(cyclopropylmethyl)-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (500 mg, 31.84%) as an off white solid.
  • LCMS: 372.2 [M+1]+
  • Step-6: Synthesis of tert-butyl 7-((2-(cyclopropylmethyl)-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 2-(cyclopropylmethyl)-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (500 mg, 1.34 mmol, 1.0 eq) in toluene (10 mL) was added m-CPBA (465 mg, 2.692 mmol, 2.0 eq) and allowed to stir at RT for 30 min. tert-Butyl 7-amino-3,4-dihydro-1H-isoquinoline-2-carboxylate (401 mg, 1.615 mmol, 1.3 eq) and DIPEA (0.9 mL, 5.384 mmol, 4.0 eq) were added and allowed to stir at RT for 12 h. The formation of precipitates was obr served which was filtered to afford the desired compound, tert-butyl 7-((2-(cyclopropylmethyl)-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (400 mg, 51.98%) as an off white solid.
  • LCMS: 546.4 [M+1]+
  • Step-7: Synthesis of 2-(cyclopropylmethyl)-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride: Tert-butyl 7-((2-(cyclopropylmethyl)-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (78 mg, 0.136 mmol, 1.0 eq) was dissolved in dioxane (2 mL) and added 4M dioxane-HCl (2 mL) and allowed to stir at RT for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 2-(cyclopropylmethyl)-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride (40 mg, 57.71%) as an off white solid.
  • LCMS: 472.3 [M+1]+
  • 1H NMR (400 MHz, DMSO-d6, HCl salt): δ 10.3 (br s, 1H), 9.1 (br s, 1H), 8.83 (s, 1H), 8.08 (t, J=7.67 Hz, 1H), 7.80 (d, J=7.89 Hz, 1H), 7.60-7.69 (m, 1H), 7.49 (d, 1H), 7.19 (d, J=8.33 Hz, 1H), 5.48-5.65 (m, 1H), 4.78-4.93 (m, 2H), 4.24 (br s, 2H), 4.13 (t, J=7.00 Hz, 2H), 3.37-3.34 (m, 2H), 2.95 (t, J=6.14 Hz, 2H), 2.00-2.16 (m, 2H), 1.42 (s, 6H).
  • Example S12. Synthesis of 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02373
  • Step-1: Synthesis of tert-butyl 6-((2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (200 mg, 0.579 mmol, 1.0 eq) in toluene (10 mL) was added m-CPBA (200 mg, 1.158 mmol, 2.0 eq) and allowed to stir at RT for 30 min. tert-Butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (158 mg, 0.636 mmol, 1.1 eq) and DIPEA (0.404 mL, 2.316 mmol, 4.0 eq) were added and allowed to stir at RT 12 h. The formation of precipitate was obr served which was filtered and purified by flash chromatography [elution 0-30% EtOAc in hexane] to afford the desired compound, tert-butyl 6-((2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (115 mg, 36.50%) as an off white solid.
  • LCMS: 546.5 [M+1]+
  • Step-2: Synthesis of 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride: tert-butyl 6-((2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (110 mg, 0.201 mmol, 1.0 eq) was dissolved in 4M HCl in dioxane (2 mL) and allowed to stir at RT for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (51 mg, 49.03%) as yellow solid.
  • LCMS: 446.4 [M+1]+
  • 1H NMR (400 MHz, DMSO-d6): δ 10.33 (brs, 1H), 9.27 (brs, 2H), 8.88 (s, 1H), 8.09 (t, J=8.11 Hz, 1H), 7.85 (s, 1H), 7.78 (brs, 1H), 7.66 (d, J=7.89 Hz, 1H), 7.49 (d, J=7.45 Hz, 1H), 7.17 (d, J=8.33 Hz, 1H), 4.21 (brs, 2H), 4.05 (d, J=7.02 Hz, 2H), 3.39 (brs, 2H), 3.01 (brs, 2H), 1.44 (s, 6H), 0.98 (t, J=6.80 Hz, 3H).
  • Example S13. Synthesis of 6-((4,4-dimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02374
  • Step-1: Synthesis of tert-butyl 7-((2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-4,4-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (1.1 g, 3.184 mmol, 1.0 eq) in toluene (20 mL) was added m-CPBA (1.57 g, 6.368 mmol, 2.0 eq) and allowed to stir at RT for 30 min. tert-Butyl 7-amino-4,4-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (0.968 g, 3.502 mmol, 1.1 eq) and DIPEA (2.2 mL, 12.736 mmol, 4.0 eq) were added and allowed to stir at RT 12 h. The reaction mixture was diluted with water (50 mL) and extracted with EtOAc (2×50 mL). The combined organic layers were washed with water (50 mL), brine (50 mL), dried over Na2SO4, filtered, concentrated and purified by flash chromatography [elution 0-30% EtOAc in Hexane] to afford the desired compound, tert-butyl 7-((2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-4,4-dimethyl-3,4-dihydroiso quinoline-2(1H)-carboxylate (630 mg, 34.61%) as light yellow solid.
  • LCMS: 574.4 [M+1]+
  • Step-2: Synthesis of 6-((4,4-dimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride: tert-butyl 7-((2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-4,4-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (500 mg, 0.871 mmol, 1.0 eq) was dissolved in 4M HCl in dioxane (5 mL) and allowed to stir at RT for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure and purified by diethyl ether to afford the desired compound, 6-((4,4-dimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (475 mg, 99.99%) as yellow solid.
  • LCMS: 474.4 [M+1]+
  • 1H NMR (400 MHz, DMSO-d6): δ 10.33 (brs, 1H), 9.34 (brs, 2H), 8.87 (s, 1H), 8.11 (t, J=7.89 Hz, 1H), 7.86 (d, J=7.89 Hz, 1H), 7.70 (brs, 1H), 7.66 (d, J=7.45 Hz, 1H), 7.54 (d, J=8.33 Hz, 1H), 7.44 (d, J=8.77 Hz, 1H), 4.25 (brs, 2H), 4.01-4.08 (m, 2H), 3.21 (brs, 2H), 1.44 (s, 6H), 1.35 (s, 6H), 0.98 (t, J=7.02 Hz, 3H).
  • Example S14. Synthesis of 2-ethyl-6-((2-(2-hydroxyacetyl)-4,4-dimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02375
  • Step-1: Synthesis of 2-ethyl-6-((2-(2-hydroxyacetyl)-4,4-dimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of glycolic acid (31 mg, 0.401 mmol, 1.1 eq) in DMF (5 mL) was added HATU (208 mg, 0.547 mmol, 1.5 eq) at RT. The resulting mixture was stirred at RT for 5 min, followed by addition of 6-((4,4-dimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (200 mg, 0.365 mmol, 1.0 eq) and DIPEA (0.128 mL, 0.730 mmol, 2.0 eq) and stirred at RT for 1 h. The reaction mixture was diluted with water (50 mL) and extracted with ethyl acetate (2×50 mL). The combined organic extracts were washed with water (50 mL) and brine (50 mL) dried over Na2SO4, concentrated under reduced and purified by flash chromatography [elution 0-3% MeOH in CH2CL2] to afford the desired compound, 2-ethyl-6-((2-(2-hydroxyacetyl)-4,4-dimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl) amino)-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (70 mg, 35.89%) as off white solid.
  • LCMS: 532.3 [M+2]+
  • 1H NMR (400 MHz, DMSO-d6): δ 10.27 (brs, 1H), 8.86 (s, 1H), 8.15 (brs, 1H), 7.87 (t, J=7.67 Hz, 1H), 7.58-7.79 (m, 2H), 7.43 (d, J=7.89 Hz, 1H), 7.26-7.40 (m, 1H), 5.34 (s, 1H), 4.64-4.76 (m, 2H), 4.61 (brs, 1H), 4.27 (d, J=5.26 Hz, 1H), 4.21 (d, J=5.70 Hz, 1H), 4.05 (d, J=7.02 Hz, 2H), 3.51 (brs, 1H), 3.38 (brs, 1H), 1.44 (s, 6H), 1.23 (s, 3H), 1.20 (s, 3H), 0.91-1.07 (m, 3H).
  • Example S15. Synthesis of 1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-2-propyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02376
  • Step-1: Synthesis of 6-(methylthio)-2-propyl-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-allyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-on (2.0 g, 8.9 mmol, 1.0 eq) in dry MeOH (50 ML), Pd/C (10 wt %) (200 mg) was added under stirring under inert atmosphere in Parr vessel. The reaction was stirred in Parr reactor at 60 psi hydrogen pressure 12 h. The reaction was monitored by LCMS. After completion the mixture was filtered through celite and concentrated to afford, 6-(methylthio)-2-propyl-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one as yellow solid (1.318 g, 65%).
  • LCMS: 225.1 [M+1]+
  • Step-2: Synthesis of 1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(methylthio)-2-propyl-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 6-(methylthio)-2-propyl-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (1.318 g, 5.88 mmol, 1.0 eq) and 2-(6-bromo-2-pyridyl)propan-2-ol (1.525 g, 7.06 mmol, 1.20 eq) in dioxane (50 mL) were added CuI (224 mg, 1.17 mmol, 0.2 eq), K2CO3 (1.626 g, 11.76 mmol, 2 eq) and DIPEA (2.35 g, 2.35 mmol, 0.4 eq) and stirred at 80° C. 12 h. The reaction was monitored by LCMS. After completion of reaction, solvent was removed under reduced pressure; residue was diluted with water and extracted with ethyl acetate (30 mL×3). The combined organic layer was washed with brine solution, dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography (elution: 0-10% MeOH in CH2Cl2) to afford 1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (1.035 g, 49%) as yellow solid.
  • LCMS: 360.1 [M+1]+
  • Step-3: Synthesis of tert-butyl 7-((1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2-propyl-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (1.035 g, 2.87 mmol, 1.0 eq) in toluene (50 mL), m-CPBA (0.99 g, 5.75 mmol, 2.0 eq) was added under stirring and resulting mixture was allowed to stir at RT for 30 min. Further, tert-butyl-7-amino-3,4-dihydro-1H-isoquinoline-2-carboxylate (0.786 g, 3.16 mmol, 1.1 eq) and DIPEA (1.48 g, 11.51 mmol, 4 eq) were added and the reaction was allowed to stir at RT for 12 h. The reaction was monitored by LCMS. After completion reaction was quenched with water (30 mL) and extracted with ethyl acetate (30 mL×3). The combined organic layer was washed with brine solution (50 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford crude product which was purified by flash chromatography (elution: 0-10% MeOH in CH2Cl2) to afford tert-butyl 7-((1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2-propyl-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (2.5 g, 78%) as yellow solid.
  • LCMS: 560.3 [M+1]+
  • Step-4: Synthesis of 1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-2-propyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: 20% HCl in dioxane (2 mL) was added to a stirred solution of tert-butyl 7-((1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2-propyl-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (105 mg, 0.18 mmol, 1.0 eq) at 0° C. and the resulting solution was allowed to stir at RT for 3 h. After completion of reaction, solvent was removed under reduced pressure, and the resulting solid was filtered and washed with ether, dried to afford HCl salt of 1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-2-propyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (60 mg, 65%) as a white solid.
  • LCMS: 460.3 [M+1]+
  • 1H NMR (400 MHz, DMSO-d6, HCl salt): δ 10.36 (br s, 1H), 9.21 (br s, 2H), 8.39 (s, 1H), 8.09 (t, J=7.89 Hz, 1H), 7.84 (d, J=7.89 Hz, 1H), 7.74 (br s, 1H), 7.66 (d, J=7.89 Hz, 1H), 7.46-7.53 (m, 1H), 4.28-4.26 (m, 2H), 4.03 (t, J=7.24 Hz, 2H), 3.41-3.66 (m, 2H), 2.96 (t, J=6.14 Hz, 2H), 1.44 (s, 6H), 1.29-1.41 (m, 2H), 0.68 (t, J=7.45 Hz, 3H).
  • Example S16. Synthesis of 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((2,4,4-trimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02377
  • Synthesis of 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((2,4,4-trimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 6-((4,4-dimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (200 mg, 0.366 mmol, 1.0 eq) and HCHO (0.15 mL, 1.83 mmol, 5.0 eq) in DCE (10 mL) was dropwise added acetic acid (0.11 mL, 1.83 mmol, 5.0 eq) at 0° C. The resulting mixture was stirred at RT for 1 h followed by addition of NaCNBH3 (69 mg, 1.098 mmol, 3.0 eq) at 0° C. The resulting mixture was stirred at RT for 1 h. The progress of reaction was monitored by LCMS. The reaction mixture was concentrated, basified with saturated solution of NaHCO3 (50 mL), extracted with EtOAc (2×50 mL). The combined organic extracts were washed with water (50 mL), with brine (50 mL) dried over Na2SO4 and concentrated under reduced and purified by reversed phase chromatography to afford the desired compound, 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((2,4,4-trimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (0.02 g, 11.23%) as white solid.
  • LCMS: 488.4 [M+1]+
  • 1H NMR (400 MHz, DMSO-d6): δ 10.16 (brs, 1H), 8.84 (s, 1H), 8.05 (t, J=7.89 Hz, 1H), 7.82 (d, J=7.89 Hz, 1H), 7.67 (d, J=7.89 Hz, 1H), 7.52 (brs, 1H), 7.42 (d, J=7.89 Hz, 1H), 7.28 (d, J=8.33 Hz, 1H), 5.33 (s, 1H), 4.03 (d, J=7.02 Hz, 2H), 3.43 (s, 2H), 2.34 (d, J=9.21 Hz, 5H), 1.44 (s, 6H), 1.23 (s, 6H), 0.97 (t, J=7.02 Hz, 3H).
  • Example S17. Synthesis of 2-cyclopropyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02378
  • Step-1: Synthesis of tert-butyl 7-((2-cyclopropyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 2-cyclopropyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (130 mg, 0.364 mmol, 1.0 eq) in toluene (10 mL) was added m-CPBA (126 mg, 0.728 mmol, 2.0 eq) and allowed to stir at RT for 30 min. tert-Butyl-7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (108 mg, 0.436 mmol, 2.0 eq) and DIPEA (0.25 mL, 1.45 mmol, 4.0 eq) were added and allowed to stir at RT for 12 h. The formation of precipitate was obr served which was filtered to afford the desired compound, tert-butyl 7-((2-cyclopropyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (87 mg, 42.89%) as an off white solid.
  • LCMS: 558.3 [M+1]+
  • Step-2: Synthesis of 2-cyclopropyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: Tert-butyl-7-((2-cyclopropyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (35 mg, 0.056 mmol, 1.0 eq) was dissolved in dioxane (0.5 mL) and added 6M dioxane-HCl (1 mL) and allowed to stir at RT for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 2-cyclopropyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (12 mg, 16.81%) as an off white solid.
  • LCMS: 458.3 [M+1]+
  • 1H NMR (400 MHz, DMSO-d6): δ 10.18 (brs, 1H), 8.79 (s, 1H), 8.06 (t, J=7.67 Hz, 1H), 7.75 (d, J=7.89 Hz, 1H), 7.65 (d, J=7.45 Hz, 2H), 7.42 (d, J=8.33 Hz, 1H), 7.09 (d, J=7.89 Hz, 1H), 5.32 (s, 1H), 4.05 (br s, 2H), 3.17 (br s, 4H), 2.80 (br s, 2H), 1.44 (s, 6H), 0.81 (brs, 4H).
  • Example S18. Synthesis of 2-ethyl-1-(6-methoxypyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02379
  • Step-1: Synthesis of 2-ethyl-1-(6-methoxypyridin-2-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-ethyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (450 mg, 2.140 mmol, 1.0 eq) and 2-bromo-6-methoxypyridine (483 mg, 2.568 mmol, 1.20 eq) in dioxane (20 mL) were added K2CO3 (591 mg, 4.28 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min followed by addition of CuI (82 mg, 0.428 mmol, 0.2 eq), and DIPEA (0.092 mL, 0.856 mmol, 0.4 eq). Reaction was again purged with nitrogen for 10 min and stirred at 90° C. for 12 h. After completion of reaction, the reaction mixture was diluted with water (20 mL) and extracted with EtOAc (50 mL×2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography (elution 0-30% EtOAc in Hexane) to afford the desired compound, 2-ethyl-1-(6-methoxypyridin-2-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (300 mg, 44.18%) as an off white solid.
  • LCMS: 318.1 [M+1]+
  • Step-2: Synthesis of tert-butyl 7-((2-ethyl-1-(6-methoxypyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 2-ethyl-1-(6-methoxypyridin-2-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (150 mg, 0.472 mmol, 1.0 eq) in toluene (5.0 mL) was added m-CPBA (233 mg, 0.944 mmol, 2.0 eq) and allowed to stir at RT for 30 min. tert-Butyl-7-amino-3,4-dihydro-1H-isoquinoline-2-carboxylate (130 mg, 0.519 mmol, 1.1 eq) and DIPEA (0.331 mL, 1.888 mmol, 4.0 eq) were added and allowed to stir at RT for 12 h. The formation of precipitate was obr served which was filtered to afford the desired compound, tert-butyl 7-((2-ethyl-1-(6-methoxypyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (125 mg, 49.18%) as off white solid.
  • LCMS: 518.3 [M+1]+
  • Step-3: Synthesis of 2-ethyl-1-(6-methoxypyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride: tert-Butyl 7-((2-ethyl-1-(6-methoxypyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (120 mg, 0.231 mmol, 1.0 eq) was dissolved in 4M HCl in dioxane (2 mL) and allowed to stir at RT for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 2-ethyl-1-(6-methoxypyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride (85 mg, 88.54%) as yellow solid.
  • LCMS: 418.3 [M+1]+
  • 1H NMR (400 MHz, DMSO-d6): δ 10.34 (br s, 1H), 9.37 (br s, 2H), 8.87 (s, 1H), 8.02 (t, J=7.89 Hz 1H), 7.73 (br s, 1H), 7.52 (d, J=7.89 Hz, 2H), 7.17 (d, J=8.77 Hz, 1H), 6.85 (d, J=7.89 Hz, 1H), 4.23 (br s, 2H), 4.04 (q, J=6.72 Hz, 2H), 3.87 (s, 3H), 3.36 (br s, 2H), 2.96 (t, J=6.14 Hz 2H), 1.01 (t, J=6.80 Hz, 3H).
  • Example S19. Synthesis of 2-ethyl-1-(6-methoxypyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02380
  • Step-1: Synthesis of tert-butyl 6-((2-ethyl-1-(6-methoxypyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 2-ethyl-1-(6-methoxypyridin-2-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (150 mg, 0.472 mmol, 1.0 eq) in toluene (5.0 mL) was added m-CPBA (233 mg, 0.944 mmol, 2.0 eq) and allowed to stir at rt for 30 min. tert-Butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (130 mg, 0.519 mmol, 1.1 eq) and DIPEA (0.331 mL, 1.888 mmol, 4.0 eq) were added and allowed to stir at RT for overnight. The formation of precipitates was observed which was filtered to afford the desired compound, tert-butyl 6-((2-ethyl-1-(6-methoxypyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (120 mg, 49%) as off white solid.
  • LCMS: [M+1]+: 518.3
  • Step-2: Synthesis of 2-ethyl-1-(6-methoxypyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: tert-Butyl 6-((2-ethyl-1-(6-methoxypyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl) amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (120 mg, 0.231 mmol, 1.0 eq) was dissolved in (3 mL) 4M HCl in dioxane and allowed to stir at RT for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure and purified by SFC to afford the desired compound, 2-ethyl-1-(6-methoxypyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (40 mg, 42%) as light yellow solid.
  • LCMS [M+1]+: 418.3
  • UPLC @ 254 nm=91.26% and @ 220 nm=93.54%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.17 (br s, 1H), 8.84 (s, 1H), 7.97 (t, J=7.67 Hz, 1H), 7.61 (br s, 1H), 7.50 (d, J=7.45 Hz, 1H), 7.38 (d, J=8.33 Hz, 1H), 6.95 (d, J=8.33 Hz, 1H), 6.85 (d, J=8.33 Hz, 1H), 4.03 (d, J=6.58 Hz, 2H), 3.86 (s, 3H), 3.82 (br s, 2H), 2.97 (br s, 2H), 2.67 (br s, 2H), 1.00 (t, J=6.80 Hz, 3H).
  • Example S20. Synthesis of 1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride
  • Figure US20230109104A1-20230406-C02381
  • Step-1: Synthesis of 1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (0.5 g, 2.23 mmol, 1.0 eq) and 2-(6-bromo-2-pyridyl)propan-2-ol (0.578 g, 2.67 mmol, 1.2 eq) in dioxane (30 mL) were added CuI (0.084 g, 0.44 mmol, 0.2 eq), K2CO3 (0.62 g, 4.46 mmol, 2 eq) and DMEDA (0.078 g, 0.89 mmol, 0.4 eq) and the resulting mixture was stirred at 80° C. for 16 h. After completion of reaction, solvent was removed under reduced pressure; residue was diluted with water (20 mL) and extracted with ethyl acetate (10 mL×3). The combined organic layer was washed with brine (5 mL×3), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography (EtOAc/hexane 10-70%) to afford 1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (556 mg, 69%) as yellow oil.
  • LCMS: 360 [M+1]+
  • Step-2: Synthesis of tert-butyl 6-((1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (250 mg, 0.69 mmol, 1.0 eq) in toluene (3 mL) was added m-CPBA (300.3 mg, 1.74 mmol, 2.5 eq) and allowed to stir at rt for 30 min. Further, tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (190.15 mg, 0.76 mmol, 1.1 eq) and DIPEA (0.49 mL, 2.76 mmol, 4 eq) were added to the above mixture and allowed to stir at rt for 12 h. After completion of reaction, solvent was removed under reduced pressure; residue was diluted with water (10 mL) and extracted with ethyl acetate (5 mL×3). The combined organic layer was washed with brine solution (5 mL×3), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford crude product which was purified by flash chromatography (MeOH/CH2CH2 1-5%) to afford tert-butyl 6-((1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (270 mg, 69%) as yellow solid.
  • LCMS: 560 [M+1]+
  • Step-3: Synthesis of 1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride: 20% HCl in dioxane (2 mL) was added to a solution of tert-butyl 6-((1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (260 mg, 0.46 mmol, 1.0 eq) in dioxane (1 mL) at 0° C. and the resulting solution was allowed to stir at rt for 3 h. After completion of reaction, solvent was removed under reduced pressure and the resulting solid was filtered and washed with diethyl ether (5 mL×3), dried to afford 1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride (165 mg, 71%) as a yellow solid.
  • LCMS: 460 [M+1]+
  • UPLC @ 254 nm=98.23% and @ 220 nm=97.37%
  • 1H NMR (400 MHz, DMSO-d6, HCl salt): δ 10.3 (br s, 1H), 9.27 (br s, 1H), 8.79 (s, 1H), 8.07-8.03 (m, 1H), 7.70-7.63 (m, 3H), 7.42 (d, J=8 Hz, 1H), 7.12 (d, J=8 Hz, 1H) 4.18 (br. s., 1H), 3.36-3.33 (m, 2H), 2.97-2.95 (m, 2H), 1.40 (s, 6H), 1.32 (d, J=6.58 Hz, 6H).
  • Example S21. Synthesis of 6-(2′,3′-dihydro-1′H-spiro[cyclopropane-1,4′-isoquinoline]-7′-ylamino)-2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one
  • Figure US20230109104A1-20230406-C02382
  • Step-1: Synthesis of tert-butyl 7′-(2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-ylamino)-1′H-spiro[cyclopropane-1,4′-isoquinoline]-2′(3′H)-carboxylate: To a stirred solution of 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(methylthio)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one (200 mg, 0.58 mmol, 1.0 eq) in toluene (5 mL) was added m-CPBA (250 mg, 1.45 mmol, 2.5 eq.) and allowed to stir at RT for 30 min. tert-Butyl 7′-amino-1′H-spiro[cyclopropane-1,4′-isoquinoline]-2′(3′H)-carboxylate (160 mg, 0.58 mmol, 1.0 eq) and DIPEA (300 mg, 2.32 mmol, 4.0 eq) were added and allowed to stir at RT for 1 h. Progress of reaction was monitored by LCMS. After completion of reaction solvent was removed under reduced pressure, residue was diluted with water (20 mL) and extracted with ethyl acetate (50 mL×3). Combined organic layer was washed with water (20 mL×3), dried over anhydrous Na2SO4 and concentrated under reduced pressure. Crude was purified by flash chromatography to obtain tert-butyl 7′-(2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-ylamino)-1′H-spiro[cyclopropane-1,4′-isoquinoline]-2′(3′H)-carboxylate (150 mg, 45.3%).
  • LCMS: 572 [M+1]+
  • Step-2: Synthesis of 6-(2′,3′-dihydro-1′H-spiro[cyclopropane-1,4′-isoquinoline]-7′-ylamino)-2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one: tert-butyl 7′-(2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-ylamino)-1′H-spiro[cyclopropane-1,4′-isoquinoline]-2′(3′H)-carboxylate (150 mg, 0.262 mmol, 1.0 eq) was dissolved in 4N HCl in dioxane (5 mL) at 0° C. Reaction was stirred at RT for 30 min. Progress of reaction was monitored by LCMS. After completion of reaction, precipitated compound was filtered off and dried under vacuum to obtain 6-(2′,3′-dihydro-1′H-spiro[cyclopropane-1,4′-isoquinoline]-7′-ylamino)-2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one (35 mg, 28.4%) as HCl salt.
  • LCMS: 472 [M+1]+
  • UPLC @ 254 nm=90.35% and @ 220 nm=94.63%
  • 1H NMR (400 MHz, DMSO-d6): δ 10.31 (br s, 1H), 9.52 (br s, 2H), 8.86 (s, 1H), 8.11 (t, J=7.89 Hz, 1H), 7.84 (d, J=7.89 Hz, 1H), 7.72 (br s, 1H), 7.66 (d, J=7.45 Hz, 1H), 7.48 (d, J=8.77 Hz, 1H), 6.83 (d, J=8.33 Hz, 1H), 4.36 (br s, 2H), 4.04 (d, J=7.02 Hz, 2H), 3.24 (br s, 2H), 1.44 (s, 6H), 1.08 (br s, 4H), 0.97 (t, J=7.02 Hz, 3H).
  • Example S22. Synthesis of 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(1,2,3,4-tetrahydroisoquinolin-5-ylamino)-JH-pyrazolo[3,4-d]pyrimidin-3(2H)-one
  • Figure US20230109104A1-20230406-C02383
  • Step-1: Synthesis of tert-butyl 5-(2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(methylthio)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one (200 mg, 0.58 mmol, 1.0 eq) in 5 mL of Toluene was added m-CPBA (250 mg, 1.45 mmol, 2.5 eq.) and allowed to stir at rt for 30 minutes. tert-butyl 5-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (140 mg, 0.58 mmol, 1.0 eq) and DIPEA (300 mg, 2.32 mmol, 4.0 eq) were added and allowed to stir at rt for 1 h. Progress of reaction was monitored by LCMS. After completion of reaction solvent was removed under reduced pressure, residue was diluted with 20 ml of water and extracted with ethyl acetate (50 mL×3). Combined organic layer was washed with water (20 ml×3), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Crude was purified by flash chromatography to obtain 170 mg (53.79%) of tert-butyl 5-(2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline-2(1H)-carboxylate.
  • LCMS: 546 [M+1]+
  • Step-2: Synthesis of 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(1,2,3,4-tetrahydroisoquinolin-5-ylamino)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one: tert-butyl 5-(2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (170 mg, 0.311 mmol, 1.0 eq) was dissolved in 5 mL of 4N HCl in dioxane solution at 0° C. Reaction was stirred at rt for 30 min. Progress of reaction was monitored by LCMS. After completion of reaction, precipitated compound was filtered off and dried under vacuum. Crude was basified with saturated NaHCO3 solution; precipitated compound was filtered off and dried under vacuum and purified by reverse phase chromatography to obtain 30 mg (21.7%) free base of 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(1,2,3,4-tetrahydroisoquinolin-5-ylamino)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one.
  • LCMS: 446 [M+1]+
  • 1H NMR (400 MHz, DMSO-d6) δ 9.56 (brs, 1H), 8.77 (s, 1H), 7.89 (brs, 1H), 7.69 (d, J=6.14 Hz, 1H), 7.58 (d, J=7.45 Hz, 1H), 7.33 (brs, 1H), 7.19 (t, J=7.67 Hz, 1H), 6.97 (d, J=7.89 Hz, 1H), 5.30 (brs, 1H), 3.96-4.07 (m, 4H), 3.04 (brs, 2H), 2.69 (brs, 2H), 1.37-1.49 (m, 7H), 0.95 (t, J=6.80 Hz, 3H).
  • Example S23. Synthesis of 2-cyclopropyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02384
  • Step-1: synthesis of tert-butyl cyclopropyl(1,3-dioxoisoindolin-2-yl)carbamate: To a stirred solution of tert-butyl (1,3-dioxoisoindolin-2-yl)carbamate (5.0 g, 19.06 mmol, 1.0 eq), in DCM (120 mL) were added cyclopropylboronic acid (4.61 g, 38.12 mmol, 2.0 eq), 2,2-bipyridine, (5.95 g, 38.12 mmol, 2.0 eq), Cu(OAc)2 (6.92 g, 38.12 mmol, 2.0 eq) and Na2CO3 (6.06 g, 57.18 mmol, 3.0 eq) at rt. The resulting mixture was stirred in open bottle for 24 h. The progress of reaction was monitored by LCMS. The reaction mixture was diluted with water (100 mL), extracted with DCM (2×200 mL). The combined organic layers were washed with water (100 mL), with brine (50 mL), dried over Na2SO4, concentrated and purified by column chromatography (Combiflash, elution, 0-15% EtOAC in hexane) to afford the desired compound tert-butyl cyclopropyl(1,3-dioxoisoindolin-2-yl)carbamate (64.3 g, 74.65%) as white solid.
  • LCMS: 303.1 [M+1]+
  • Step-2: Synthesis of tert-butyl 1-cyclopropylhydrazine-1-carboxylate: To a stirred solution of tert-butyl cyclopropyl(1,3-dioxoisoindolin-2-yl)carbamate (4.3 g, 14.22 mmol, 1.0 eq), in DCM (60 mL) was added NH2NH2.H2O (1.35 mL, 28.44 mmol, 2.0 eq) at rt. The resulting mixture was stirred at rt for overnight, formation of white precipitates was observed. The progress of reaction was monitored by 1HNMR. The reaction mixture was filtered and the filtrate was concentrated and purified by pentane to afford the desired compound, tert-butyl 1-cyclopropylhydrazine-1-carboxylate (2.2 g, 90.16%) as light yellow liquid.
  • LCMS: 173.1 [M+1]+
  • Step-3: Synthesis of ethyl 4-(2-(tert-butoxycarbonyl)-2-cyclopropylhydrazinyl)-2-(methylthio)pyrimidine-5-carboxylate: To a stirred solution of tert-butyl 1-cyclopropylhydrazine-1-carboxylate (1.98 g, 8.51 mmol, 1.0 eq) and tert-butyl 1-(cyclopropylmethyl)hydrazine-1-carboxylate (2.2 g, 12.77 mmol, 1.5 eq) in (24 mL) of THF was added DIPEA (3.68 mL, 21.27 mmol, 2.5 eq) and stirred at 80° C. for 16 h. Solvent was removed under reduced pressure. Residue was diluted with water (50 mL) and extracted with EtOAc (50 mL×2). The organic layers were washed with brine solution (50 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure, purified by column chromatography (Combiflash, elution-0-40% EtOAc in Hexane) to afford the desired compound, ethyl 4-(2-(tert-butoxycarbonyl)-2-cyclopropylhydrazinyl)-2-(methylthio)pyrimidine-5-carboxylate (2.2 g, 70.28%) as off white solid.
  • LCMS: 369.1 [M+1]+
  • Step-4: Synthesis of 2-cyclopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of ethyl 4-(2-(tert-butoxycarbonyl)-2-cyclopropylhydrazinyl)-2-(methylthio)pyrimidine-5-carboxylate (2.2 g, 5.97 mmol, 1.0 eq) in 24 mL of DCM was added TFA (5.97 mL) dropwise at 0° C. and allowed to stir at rt for overnight. After completion of reaction, solvent was removed under reduced pressure. Residue was diluted with ethanol (12 mL), 6N NaOH solution (12 mL) was added in to above solution at 0° C. and allowed to stir at rt for 1 h. After completion, the reaction mixture was acidified by using 6N HCl solution. Ethanol was removed under reduced pressure; residue obtained was cooled to rt and extracted with chloroform (100 mL×3). The organic layers were washed with brine solution, dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford the desired compound, 2-cyclopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (1.0 g, 75.75%) as yellow solid.
  • LCMS: 223.1 [M+1]+
  • Step-5: Synthesis of 2-cyclopropyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-(cyclopropylmethyl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (1.0 g, 4.49 mmol, 1.0 eq) and 2-(6-bromo-2-pyridyl)propan-2-ol (1.16 g, 5.39 mmol, 1.20 eq) in (10 mL) of dioxane were added Potassium carbonate (1.24 g, 8.98 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min, followed by addition of copper iodide (171 mg, 0.89 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (158 mg, 1.79 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 90° C. for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layers were washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography to afford the desired compound, 2-cyclopropyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (600 mg, 37.50%) as an off white solid.
  • LCMS: 358.2 [M+1]+
  • Step-6: Synthesis of tert-butyl 6-((2-cyclopropyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 2-cyclopropyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (100 mg, 0.280 mmol, 1.0 eq) in (3 mL) of toluene was added m-CPBA (97 mg, 0.560 mmol, 2.0 eq) and allowed to stir at rt for 30 min. tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (83 mg, 0.336 mmol, 2.0 eq) and DIPEA (0.2 mL, 1.12 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. The formation of precipitates was observed which was filtered to afford the desired compound, tert-butyl 6-((2-cyclopropyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (50 mg, 32.04%) as an off white solid.
  • LCMS: 558.3 [M+1]+
  • Step-7: Synthesis of 2-cyclopropyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: tert-butyl 6-((2-cyclopropyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (50 mg, 0.089 mmol, 1.0 eq) was dissolved in (0.5 mL) of dioxane and added 6M dioxane-HCl (1 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 2-cyclopropyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (12 mg, 29.25%) as an off white solid.
  • LCMS: 458.3 [M+1]+; UPLC @ 254 nm=99.73% and @ 220 nm=99.78%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.09 (br. s., 1H), 8.77 (s, 1H), 8.04-8.13 (m, 1H), 7.64 (br. s., 2H), 7.35 (br. s., 1H), 6.91-7.01 (m, 2H), 4.40 (br. s., 1H), 3.78 (br. s., 2H), 3.52 (br. s., 3H), 3.20 (br. s., 2H), 2.94 (br. s., 2H), 2.66 (br. s., 3H), 1.59 (s, 3H), 1.44 (s, 7H), 0.81 (br. s., 4H).
  • Example S24. Synthesis of 2-ethyl-1-(6-(2-oxopyrrolidin-1-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-indazol-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02385
  • Step-1: Synthesis of 2-ethyl-6-(methylthio)-1-(6-(2-oxopyrrolidin-1-yl)pyridin-2-yl)-1,2-dihydro-3H-indazol-3-one: To a stirred solution of 2-ethyl-6-(methylthio)-1,2-dihydro-3H-indazol-3-one (400 mg, 1.90 mmol, 1.0 eq) and 1-(6-bromopyridin-2-yl)pyrrolidin-2-one (459 mg, 2.28 mmol, 1.20 eq) in (20 mL) of dioxane was added Potassium carbonate (525 mg, 3.80 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min followed by addition of copper iodide (72 mg, 0.38 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (0.08 mL, 0.76 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 90° C. for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-30% EtOAc in hexane] to afford the desired compound 2-ethyl-6-(methylthio)-1-(6-(2-oxopyrrolidin-1-yl)pyridin-2-yl)-1,2-dihydro-3H-indazol-3-one (200 mg, 28.26%) as light yellow solid.
  • LCMS: 371.3 [M+1]+
  • Step-2: Synthesis of tert-butyl 7-((2-ethyl-3-oxo-1-(6-(2-oxopyrrolidin-1-yl)pyridin-2-yl)-2,3-dihydro-1H-indazol-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 2-ethyl-6-(methylthio)-1-(6-(2-oxopyrrolidin-1-yl)pyridin-2-yl)-1,2-dihydro-3H-indazol-3-one (200 mg, 0.539 mmol, 1.0 eq) in (5.0 mL) of toluene was added m-CPBA (283 mg, 1.347 mmol, 2.0 eq) and allowed to stir at rt for 30 minutes. tert-butyl 7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (147 mg, 0.593 mmol, 1.1 eq) and DIPEA (0.37 mL, 2.156 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. The formation of precipitates was observed which was filtered to afford the desired compound tert-butyl 7-((2-ethyl-3-oxo-1-(6-(2-oxopyrrolidin-1-yl)pyridin-2-yl)-2,3-dihydro-1H-indazol-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (200 mg, 64.91%) as white solid. LCMS: 571.4 [M+1]+
  • Step-3: Synthesis of 2-ethyl-1-(6-(2-oxopyrrolidin-1-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-indazol-3-one dihydrochloride: tert-butyl 7-((2-ethyl-3-oxo-1-(6-(2-oxopyrrolidin-1-yl)pyridin-2-yl)-2,3-dihydro-1H-indazol-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (200 mg, 0.350 mmol, 1.0 eq) was dissolved in (0.5 mL) of dioxane and added 6M dioxane-HCl (1 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 2-ethyl-1-(6-(2-oxopyrrolidin-1-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-indazol-3-one dihydrochloride (90 mg, 54.57%) as an off white solid.
  • LCMS: 469.3 [M+1]+; UPLC @ 254 nm=95.67% and @ 220 nm=96.28%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.34 (br. s., 1H), 9.15 (br. s., 2H), 8.88 (s, 1H), 8.29 (d, J=7.89 Hz, 1H), 8.12 (t, J=8.33 Hz, 1H), 7.70-7.76 (m, 1H), 7.51 (d, J=8.33 Hz, 1H), 7.18 (d, J=8.77 Hz, 1H), 4.26 (br. s., 2H), 4.02-4.13 (m, 2H), 3.98 (t, J=7.02 Hz, 2H), 3.57 (s, 2H), 3.39 (br. s., 2H), 2.96 (br. s., 2H), 2.67 (br. s., 1H), 2.59-2.64 (m, 2H), 2.00-2.12 (m, 3H), 0.99 (t, J=7.02 Hz, 3H).
  • Example S25. Synthesis of 2-ethyl-1-(6-(methylamino)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02386
  • Step-1: Synthesis of 6-bromo-N-methylpyridin-2-amine: To a solution of 2,6-dibromopyridine (5 g, 21.1, 1 eq) in EtOH (50 mL), methyl amine hydrochloride (1.71 g, 25.3 mmol, 1.2 eq) and Na2CO3 (6.71 g, 63.3 mmol, 3 eq) were added under stirring at rt. The mixture was heated at 190° C. for 12 h. The progress of the reaction was monitored by LCMS. After completion the mixture was cooled to rt, concentrated under vacuo to remove solvent. Water (10 mL×3) was added followed by extraction using EtOAc (20 mL×3). The combined organic layer was dried over Na2SO4 and concentrated. The residue was purified by flash chromatography (20% EtOAc/pet ether) to give 6-bromo-N-methylpyridin-2-amine (2.8 g, 71%) as colourless oil.
  • LCMS: 187 [M+1]+
  • Step-2: Synthesis of tert-butyl (6-bromopyridin-2-yl)(methyl)carbamate: To a solution of 6-bromo-N-methylpyridin-2-amine (2.3 g, 12.2, 1 eq) in dry DCM (50 mL), Et3N (2.57 mL, 18.4 mmol, 1.5 eq) followed by DMAP (0.3 g, 2.45 mmol, 0.2 eq) were added under stirring at rt under inert atmosphere. The resulting solution was stirred for 15-20 min at rt followed by dropwise addition of Boc-anhydride (4.23 mL, 18.4 mmol, 1.5 eq). The resulting solution was stirred at rt for 16 h. The progress of the reaction was monitored by LCMS. After completion, reaction was quenched with water (20 mL×3) and extracted using DCM (10 mL×3). The combined organic layer was dried over Na2SO4, concentrated and purified by flash chromatography (20% EtOAc/pet ether) to give tert-butyl (6-bromopyridin-2-yl)(methyl)carbamate (2.5 g, 71%) as colourless liquid.
  • LCMS: 287 [M+1]+
  • Step-3: Synthesis of tert-butyl (6-(2-ethyl-6-(methylthio)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)(methyl)carbamate: To a stirring solution of 2-ethyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (400 mg, 1.9 mmol, 1 eq) in dioxane (20 mL), tert-butyl (6-bromopyridin-2-yl)(methyl)carbamate (0.655 g, 2.28 mmol, 1.2 eq) and Cs2CO3 (1.23 g, 3.8 mmol, 2 eq) were added. The mixture was degassed for 10-15 min followed addition of Pd2(dba)3 (174.2 mg, 0.19 mmol, 0.1 eq) and xantphos (165.1 mg, 3.8 mmol, 2 eq). The reaction was heated at 100° C. for 16 h. After completion, the reaction mixture was filtered through celite and water (20 mL) was added to it. Extraction was carried out using MeOH/DCM (20 mL×3). The combined organic layer was dried, concentrated and purified by flash chromatography (MeOH/DCM 1-5%) to give tert-butyl (6-(2-ethyl-6-(methylthio)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)(methyl)carbamate (145 mg, 19%) as yellow solid.
  • LCMS: 416 ([M+1]+
  • Step-4: Synthesis of tert-butyl 7-((1-(6-((tert-butoxycarbonyl)(methyl)amino)pyridin-2-yl)-2-ethyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of tert-butyl (6-(2-ethyl-6-(methylthio)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)(methyl)carbamate (140 mg, 0.33 mmol, 1.0 eq) in toluene (4 mL); m-CPBA (145 mg, 0.84 mmol, 2.5 eq) was added under stirring and resulting mixture was allowed to stir at rt for 30 minutes. tert-butyl 7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (243.1 mg, 0.36 mmol, 1.1 eq) and DIPEA (0.23 mL, 1.34 mmol, 4 eq) were then added and the reaction was allowed to stir at rt for 16 h. The progress of the reaction was monitored by LCMS. After completion reaction was quenched with water and extracted with MeOH/DCM (10 mL×3). The combined organic layer was washed with brine solution (20 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford crude product which was purified by flash chromatography (1-10% MeOH/DCM) to afford tert-butyl 7-((1-(6-((tert-butoxycarbonyl)(methyl)amino)pyridin-2-yl)-2-ethyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (100 mg, 48%) as yellow solid.
  • LCMS: 616 [M+1]+
  • Step-5: Synthesis of 2-ethyl-1-(6-(methylamino)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride: 20% HCl in dioxane (2 mL) was added to a stirred solution of tert-butyl 7-((1-(6-((tert-butoxycarbonyl)(methyl)amino)pyridin-2-yl)-2-ethyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (97 mg, 0.15 mmol, 1.0 eq) in dioxane (2 mL) at 0° C. under inert atmosphere and the resulting solution was allowed to stir at rt for 3 h. After completion, solvent was removed under reduced pressure and the resulting solid was filtered and washed with ether, dried to afford 2-ethyl-1-(6-(methylamino)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (22 mg, 29%) as a yellow solid.
  • LCMS: 416 [M+1]+; UPLC: @220 nm=91.97% @254 nm=90.07% HPLC: @220 nm=97.13% @254 nm=96.03%
  • 1H NMR (400 MHz, DMSO-d6, Formate): δ 10.13 (br s, 1H), 8.81 (s, 1H), 8.24 (br s, 1H), 7.65-7.61 (m, 2H), 7.41-7.61 (m, 1H), 7.01 (d, J=8.33 Hz, 1H), 6.96-6.94 (m, 1H), 6.88-6.84 (m, 1H), 6.44 (d, J=7.89 Hz, 1H), 4 (q, J=7 Hz, 2H), 3.89 (br s, 2H), 3.04-3.01 (m, 2H), 2.75 (d, J=4.82 Hz, 2H), 2.7-2.67 (m, 3H), 1.01 (t, J=7.02 Hz, 3H).
  • Example 526. Synthesis of 6-(2-ethyl-3-oxo-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)-N-methylpicolinamide hydrochloride
  • Figure US20230109104A1-20230406-C02387
  • Step-1: Synthesis of 6-bromopicolinoyl chloride: To a solution of 6-bromopicolinic acid (1 g, 4.95, 1 eq) in dry DCM (100 mL), oxalyl chloride (1.27 mL, 14.85 mmol, 3 eq) was added followed by addition of DMF (0.1 mL) under stirring at 0° C. under inert atmosphere. The resulting mixture was stirred at rt for 3-4 h. After completion the mixture was concentrated under vacuo to remove oxalyl chloride to give 6-bromopicolinoyl chloride (0.95 g, 88%) as yellow solid.
  • LCMS: 220.1 [M+1]+
  • Step-2: Synthesis of 6-bromo-N-methylpicolinamide: To a solution of 6-bromopicolinoyl chloride (0.9 g, 4.08, 1 eq) in dry THF (25 mL), MeNH2 (40 wt % in H2O, 10 mL, 122.4 mmol, 30 eq) in dry THF (25 mL) was added under stirring at 0° C. under inert atmosphere. The resulting solution was stirred at rt for 16 h. The progress of the reaction was monitored by LCMS. After completion, reaction was quenched with water (20 mL×3) and extracted using DCM (10 mL×3). The combined organic layer was dried over Na2SO4, concentrated to give 6-bromo-N-methylpicolinamide (0.8 g, 82%) as yellow liquid which was used without further purification.
  • LCMS: 214 [M+1]+
  • Step-3: Synthesis of 6-(2-ethyl-6-(methylthio)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)-N-methylpicolinamide: To a stirring solution of 2-ethyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (300 mg, 1.42 mmol, 1 eq) in dioxane (10 mL), K2CO3 (349.3 mg, 2.85 mmol, 2 eq) and 6-bromo-N-methylpicolinamide (368.2 mg, 1.71 mmol, 1.2 eq) were added under stirring at rt. The mixture was degassed for 10-15 min followed addition of CuI (54.3 mg, 0.28 mmol, 0.2 eq) and DMEDA (0.06 mL, 0.57 mmol, 0.4 eq). The reaction was heated at 100° C. for 16 h. The progress of the reaction was monitored by LCMS. After completion, the reaction mixture was filtered through celite and water (20 mL) was added to it. Extraction was carried out using MeOH/DCM (10 mL×3). The combined organic layer was dried, concentrated and purified by flash chromatography (MeOH/DCM 1-5%) to give 6-(2-ethyl-6-(methylthio)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)-N-methylpicolinamide (75 mg, 15%) as yellow solid.
  • LCMS: 344 [M+1]+
  • Step-4: Synthesis of tert-butyl 7-((2-ethyl-1-(6-(methylcarbamoyl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 6-(2-ethyl-6-(methylthio)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)-N-methylpicolinamide (70 mg, 0.20 mmol, 1.0 eq) in toluene (3 mL); m-CPBA (87.6 mg, 0.50 mmol, 2.5 eq) was added under stirring and resulting mixture was allowed to stir at rt for 30 minutes. tert-butyl 7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (55.5 mg, 0.22 mmol, 1.1 eq) and DIPEA (0.16 mL, 0.81 mmol, 4 eq) were then added and the reaction was heated at 80° C. for 16 h. The progress of the reaction was monitored by LCMS. After completion reaction was quenched with water (10 mL) and extracted with MeOH/DCM (10 mL×3). The combined organic layer was washed with brine solution (20 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford crude product which was purified by flash chromatography (1-10% MeOH/DCM) to afford tert-butyl 7-((2-ethyl-1-(6-(methylcarbamoyl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (72 mg, 65%) as yellow solid.
  • LCMS: 544 [M+1]+
  • Step-5: Synthesis of 6-(2-ethyl-3-oxo-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)-N-methylpicolinamide hydrochloride: 20% HCl in dioxane (2 mL) was added to a stirred solution of tert-butyl 7-((2-ethyl-1-(6-(methylcarbamoyl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (70 mg, 0.12 mmol, 1.0 eq) in dioxane (1 mL) at 0° C. under inert atmosphere and the resulting solution was allowed to stir at rt for 3 h. The progress of the reaction was monitored by LCMS. After completion, solvent was removed under reduced pressure and the resulting solid was filtered and washed with ether, dried to afford 6-(2-ethyl-3-oxo-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)-N-methylpicolinamide hydrochloride (5 mg, 8%) as a yellow solid.
  • LCMS: 444 [M+1]+; UPLC: @ 220 nm=97.02% @ 254 nm=96.19%
  • 1H NMR (400 MHz, DMSO-d6, Formate): δ 10.32 (br s, 1H), 8.89 (s, 1H), 8.31-8.26 (m, 1H), 8.21 (br s, 1H), 8.14-8.1 (m, 1H), 8.03-8.0 (m, 1H), 7.65-7.62 (m, 1H), 7.47-7.43 (m, 1H), 7.13-7.09 (m, 1H), 4.12 (br s, 2H), 4.04 (q, J=7 Hz, 2H), 3.37-3.29 (m, 2H), 2.97-2.9 (m, 2H), 2.81 (s, 3H), 0.93 (t, J=7.02 Hz, 3H)
  • Example S27. Synthesis of 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(5,6,7,8-tetrahydro-1,6-naphthyridin-3-ylamino)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one
  • Figure US20230109104A1-20230406-C02388
  • Step-1: Synthesis of tert-butyl 3-(2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-ylamino)-7,8-dihydro-1,6-naphthyridine-6(5H)-carboxylate: To a stirred solution of 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(methylthio)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one (200 mg, 0.58 mmol, 1.0 eq) in 5 mL of Toluene was added m-CPBA (250 mg, 1.45 mmol, 2.5 eq.) and allowed to stir at rt for 30 minutes. tert-butyl 3-amino-7,8-dihydro-1,6-naphthyridine-6(5H)-carboxylate (150 mg, 0.58 mmol, 1.0 eq) and DIPEA (300 mg, 2.32 mmol, 4.0 eq) were added and allowed to stir at rt for 12 h. Progress of reaction was monitored by LCMS. After completion of reaction solvent was removed under reduced pressure, residue was diluted with 20 ml of water and extracted with ethyl acetate (50 mL×3). Combined organic layer was washed with water (20 ml×3), dried over anhydrous sodium sulfate and concentrated under reduced pressure. Crude was purified by reverse phase chromatography to obtain 20 mg (6.30%) of tert-butyl 3-(2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-ylamino)-7,8-dihydro-1,6-naphthyridine-6(5H)-carboxylate.
  • LCMS: 547 [M+1]+
  • Step-2: Synthesis of 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(5,6,7,8-tetrahydro-1,6-naphthyridin-3-ylamino)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one: tert-butyl 3-(2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-ylamino)-7,8-dihydro-1,6-naphthyridine-6(5H)-carboxylate (20 mg, 0.037 mmol, 1.0 eq) was dissolved in 1 mL of 4N HCl in dioxane solution at 0° C. Reaction was stirred at rt for 30 min. Progress of reaction was monitored by LCMS. After completion of reaction, precipitated compound was filtered off and dried under vacuum to obtain 10 mg (62.5%) HCl of 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(5,6,7,8-tetrahydro-1,6-naphthyridin-3-ylamino)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one.
  • LCMS: 447 [M+1]+
  • 1H NMR (400 MHz, DMSO-d6) δ 10.62 (br. s., 1H), 9.56 (br. s., 2H), 8.94 (s, 1H), 8.78 (br. s., 1H), 8.23 (br. s., 1H), 8.13 (t, J=7.89 Hz, 1H), 7.84 (d, J=7.89 Hz, 1H), 7.67 (d, J=7.89 Hz, 1H), 4.37 (br. s., 2H), 4.05 (d, J=7.45 Hz, 3H), 3.48 (br. s., 2H), 3.12 (br. s., 2H), 1.44 (s, 3H), 1.23 (br. s., 1H), 0.99 (t, J=7.24 Hz, 2H), 0.84 (d, J=7.45 Hz, 1H).
  • Example S28. Synthesis of 1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02389
  • Step-1: Synthesis of 2-bromo-6-(tert-butyl)pyridine: To a stirred solution of 2,6-dibromopyridine (1.0 g, 4.221 mmol, 1.0 eq) in (30.0 mL) of THF was added CuI (40 mg, 0.211 mmol, 0.05 eq) followed by addition of 2.0 M solution of tert-butylmagnesium bromide (8.44 mL, 16.885 mmol, 4.0 eq) at 0° C. The reaction mixture was stirred at RT for 1 h. The progress of reaction was monitored by LCMS. The reaction mixture was quenched with saturated solution of NH4Cl (50 mL), extracted with EtOAc (2×100 mL), the combined organic layers were washed with water (50 mL), dried over Na2SO4, concentrated to afford the desired compound 2-bromo-6-(tert-butyl)pyridine (790 mg, 87.19%) as Light yellow liquid.
  • LCMS: 214.1 [M+1]+
  • Step-2: Synthesis of 1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-ethyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (600 mg, 2.853 mmol, 1.0 eq) and 2-bromo-6-(tert-butyl)pyridine (734 mg, 3.424 mmol, 1.20 eq) in (20 mL) of dioxane was added Potassium carbonate (788 mg, 5.706 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min followed by addition of copper iodide (109 mg, 0.570 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (0.123 mL, 1.141 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 90° C. for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-30% EtOAc in hexane] to afford the desired compound 1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (380 mg, 38.77%) as light yellow solid.
  • LCMS: 344.3 [M+1]+
  • Step-3: Synthesis of tert-butyl 7-((1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (200 mg, 0.582 mmol, 1.0 eq) in (5.0 mL) of toluene was added m-CPBA (287 mg, 1.164 mmol, 2.0 eq) and allowed to stir at rt for 30 minutes. tert-butyl 7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (174 mg, 0.698 mmol, 1.2 eq) and DIPEA (0.40 mL, 2.328 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. The formation of precipitates was observed which was filtered to afford the desired compound tert-butyl 7-((1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (158 mg, 49.84%) as white solid.
  • LCMS: 544.4 [M+1]+
  • Step-4: Synthesis of 1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride: tert-butyl 7-((1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (158 mg, 0.290 mmol, 1.0 eq) was dissolved in (1 mL) of dioxane and added 4.0 M-HCl (2 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound 1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (125 mg, 83.33%) as yellow solid.
  • LCMS: 444.4 [M+1]+; UPLC @ 254 nm=99.12% and @ 220 nm=99.47%.
  • 1H NMR (400 MHz, DMSO-d6) δ 10.33 (brs, 1H), 9.26 (brs, 2H), 8.88 (s, 1H), 8.06 (t, J=7.67 Hz, 1H), 7.84 (d, J=8.33 Hz, 1H), 7.73 (brs, 1H), 7.52 (d, J=8.33 Hz, 1H), 7.43 (d, J=7.89 Hz, 1H), 7.19 (d, J=8.33 Hz, 1H), 4.27 (brs, 2H), 4.08 (d, J=7.45 Hz, 2H), 3.37 (brs, 2H), 2.96 (brs, 2H), 1.33 (s, 9H), 0.98 (t, J=7.02 Hz, 3H).
  • Example S29. Synthesis of N-(6-(2-ethyl-3-oxo-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-2,3-dihydro-JH-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)acetamide dihydrochloride
  • Figure US20230109104A1-20230406-C02390
  • Step-1: Synthesis of N-(6-bromopyridin-2-yl)acetamide: To a stirred solution of 6-bromopyridin-2-amine (1.0 g, 5.78 mmol, 1.0 eq) in (30.0 mL) of DCM was added TEA (1.61 mL, 11.567 mmol, 2.0 eq) followed by addition of actyl chloride (0.62 mL, 8.67 mmol, 1.5 eq) at 0° C. The reaction mixture was stirred at RT for overnight. The progress of reaction was monitored by LCMS. The reaction mixture was quenched with saturated solution of NaHCO3 (100 mL), extracted with EtOAc (2×100 mL), the combined organic layers were washed with water (50 mL), dried over Na2SO4, concentrated and purified by combiflash chromatography [silica gel 100-200 mesh; elution 0-10% EtOAc in hexane] to afford the desired compound N-(6-bromopyridin-2-yl)acetamide (355 mg, 28.62%) as white solid.
  • LCMS: 215.1 [M+1]+
  • Step-2: Synthesis of N-(6-(2-ethyl-6-(methylthio)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)acetamide: To a stirred solution of 2-ethyl-6-(methylthio)-1,2-dihydro-3H-indazol-3-one (285 mg, 1.356 mmol, 1.0 eq) and N-(6-bromopyridin-2-yl)acetamide (350 mg, 1.627 mmol, 1.20 eq) in (10 mL) of dioxane was added Potassium carbonate (375 mg, 2.712 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min followed by addition of copper iodide (52 mg, 0.271 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (0.06 mL, 0.542 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 90° C. for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-3% MeOH in DCM] to afford the desired compound N-(6-(2-ethyl-6-(methylthio)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)acetamide (1420 mg, 30.47%) as white solid.
  • LCMS: 345.3 [M+1]+
  • Step-3: Synthesis of tert-butyl 7-((1-(6-acetamidopyridin-2-yl)-2-ethyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of N-(6-(2-ethyl-6-(methylthio)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)acetamide (140 mg, 0.407 mmol, 1.0 eq) in (2.0 mL) of toluene was added m-CPBA (200 mg, 0.814 mmol, 2.0 eq) and allowed to stir at rt for 30 minutes. tert-butyl 7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (121 mg, 0.488 mmol, 1.2 eq) and DIPEA (0.285 mL, 1.628 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. The formation of precipitates was observed which was filtered to afford the desired compound tert-butyl 7-((1-(6-acetamidopyridin-2-yl)-2-ethyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (105 mg, 47.51%) as white solid.
  • LCMS: 545.5 [M+1]+
  • Step-4: Synthesis of N-(6-(2-ethyl-3-oxo-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)acetamide dihydrochloride: tert-butyl 7-((1-(6-acetamidopyridin-2-yl)-2-ethyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (105 mg, 0.193 mmol, 1.0 eq) was dissolved in 4.0 M-HCl (3 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound N-(6-(2-ethyl-3-oxo-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)acetamide dihydrochloride (80 mg, 80.80%) as yellow solid.
  • LCMS: 445.3 [M+1]+; UPLC @ 254 nm=94.71% and @ 220 nm=96.31%.
  • 1H NMR (400 MHz, DMSO-d6) δ 10.66 (s, 1H), 10.35 (brs, 1H), 9.33 (brs, 2H), 8.87 (s, 1H), 8.08 (d, J=3.95 Hz, 2H), 7.73 (brs, 1H), 7.58-7.68 (m, 1H), 7.50 (d, J=7.02 Hz, 1H), 7.17 (d, J=7.89 Hz, 1H), 4.23 (brs, 2H), 4.00 (d, J=7.45 Hz, 2H), 3.36 (brs, 2H), 2.96 (brs, 2H), 2.11 (s, 3H), 0.97 (t, J=7.02 Hz, 3H).
  • Example S30. 2-ethyl-6-((2-(2-hydroxyethyl)-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02391
  • Step-1: tert-butyl 7-((2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (0.2 g 0.5790 mmol, 1.0 eq) in (5.0 mL) of toluene was added m-CPBA (0.280 g, 1.158 mmol, 2 eq) and allowed to stir at rt for 30 min. followed by addition of tert-butyl 7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (0.186 g, 0.7527 mmol, 1.3 eq) and DIPEA (0.224 g 1.737 mmol, 3.0 eq) and allowed to stir at rt for overnight. The reaction mixture was diluted with water (20 mL) and extracted with EtOAc (2×20 mL). The combined organic layers were washed with water (30 mL), brine (30 mL), dried over Na2SO4, concentrated and purified by combi flash [silica gel-100-200 mesh; elution 0-35% EtOAc in Hexane] to afford the desired compound, tert-butyl 4-(4-((3-(2,6-dichlorophenyl)-4-oxo-3,4-dihydro-2H-pyrimido[5,4-e][1,3]oxazin-7-yl)amino)phenyl)-2-(hydroxymethyl)piperazine-1-carboxylate (0.110 g, 49.52%) as white solid.
  • LCMS: 546.4 [M+1]+
  • Step-2: Synthesis of 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: tert-butyl 7-((2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (0.110 mg, 0.2017 mmol, 1.0 eq) was dissolved in 4.0 M-HCl in dioxane (3 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure and purified by diethyl ether to afford the desired compound, 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (86 mg, 63.84%) as white solid.
  • LCMS: 446.5 [M+1]+
  • Step-3: 2-ethyl-6-((2-(2-hydroxyethyl)-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (0.086 g, 0.1931 mmol, 1.0 eq) and 2-bromoethan-1-ol (0.026 g, 0.2124 mmol, 1.1 eq) in DMF (2 mL) was added potassium carbonate (0.054 g, 0.3862 mmol, 2.0 eq) at rt. The resulting mixture was stirred at 90° C. for overnight. The progress of reaction was monitored by LCMS. The reaction mixture was poured into ice cold water (100 mL), stirred for 5 min, formation of precipitates was observed, which was filtered and dried under vacuum to afford the desired compound, 2-ethyl-6-((2-(2-hydroxyethyl)-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (15 mg, 15.87%) as white solid.
  • LCMS: 490.3 [M+1]+
  • 1H NMR (400 MHz, DMSO-d6): δ 10.39 (br. s., 2H), 8.95 (s, 1H), 8.20 (t, J=7.67 Hz, 1H), 7.91 (d, J=7.89 Hz, 1H), 7.81 (br. s., 1H), 7.74 (d, J=7.89 Hz, 1H), 7.60 (d, J=8.77 Hz, 1H), 7.27 (d, J=8.77 Hz, 1H), 4.60 (d, J=14.91 Hz, 1H), 4.48 (d, J=7.89 Hz, 1H), 4.12 (d, J=7.45 Hz, 2H), 3.96 (br. s., 2H), 3.80 (br. s., 2H), 3.42 (br. s., 2H), 3.23 (br. s., 2H), 3.05 (d, J=17.10 Hz, 1H), 1.51 (s, 5H), 1.42 (br. s., 1H), 1.30 (br. s., 4H), 0.99-1.10 (m, 3H), 0.91 (d, J=7.02 Hz, 1H)
  • Example S31. 2-(7-((2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-JH-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinolin-2(H)-yl)-N,N-dimethylacetamide
  • Figure US20230109104A1-20230406-C02392
  • Step-1: 2-bromo-N,N-dimethylacetamide: To a stirred solution of 2-bromo-N,N-dimethylacetamide (0.5 g, 3.1768 mmol, 1.0 eq) and dimethylamine (0.026 g, 0.2124 mmol, 1.1 eq) in THF (5 mL) at rt. The resulting mixture was stirred at RT for overnight. The progress of reaction was monitored by LCMS. Solvent was removed under reduce pressure, residue was diluted with water (5 ml) and extracted using DCM (50 ml). The combined organic layer was washed with water (10 ml), brine (10 ml) and concentrated to afford desired compound (120 mg, 22.77%) as white solid.
  • 1H NMR (400 MHz, CDCl3) δ 2.97 (s, 3H), 3.1 (s, 3H), 3.86 (s, 2H).
  • Step-2: 2-(7-((2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinolin-2(1H)-yl)-N,N-dimethylacetamide: To a stirred solution of 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (0.150 g, 0.3368 mmol, 1.0 eq) and 2-bromo-N,N-dimethylacetamide (0.061 g, 0.3705 mmol, 1.1 eq) in DMF (2 mL) was added potassium carbonate (0.093 g, 0.6736 mmol, 2.0 eq) at rt. The resulting mixture was stirred at 90° C. for overnight. The progress of reaction was monitored by LCMS. The reaction mixture was poured into ice cold water (100 mL), stirred for 5 min, formation of precipitates was observed, which was filtered and dried under vacuum to afford the desired compound, 2-(7-((2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinolin-2(1H)-yl)-N,N-dimethylacetamide (13 mg, 07.27%) as white solid.
  • LCMS: 531.4 [M+1]+; UPLC @254 nm=94.98%, @220 nm=95.59%
  • 1H NMR (400 MHz, DMSO-d6): δ 10.22 (br. s., 1H), 8.84 (s, 1H), 8.10 (t, J=7.89 Hz, 1H), 7.82 (d, J=8.33 Hz, 1H), 7.71 (br. s., 1H), 7.65 (br. s., 1H), 7.33 (d, J=7.45 Hz, 1H), 7.05 (d, J=8.33 Hz, 1H), 5.33 (s, 1H), 4.04 (d, J=6.58 Hz, 2H), 3.62 (br. s., 2H), 3.36 (br. s., 2H), 3.07 (s, 3H), 2.88 (s, 3H), 2.76 (br. s., 3H), 1.44 (s, 6H), 1.23 (br. s., 1H), 0.98 (t, J=6.80 Hz, 3H)
  • Example S32. Synthesis of 2-ethyl-1-(pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02393
  • Step-1: Synthesis of 2-ethyl-6-(methylthio)-1-(pyridin-2-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-ethyl-6-(methylthio)-1,2-dihydro-3H-indazol-3-one (500 mg, 2.378 mmol, 1.0 eq) and 2-iodopyridine (585 mg, 2.854 mmol, 1.2 eq) in (10 mL) of dioxane was added Potassium carbonate (657 mg, 4.756 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min followed by addition of copper iodide (91 mg, 0.475 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (0.1 mL, 0.951 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 90° C. for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-30% EtOAc in Hexane] to afford the desired compound 2-ethyl-6-(methylthio)-1-(pyridin-2-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (450 mg, 65.88%) as light yellow viscous.
  • LCMS: 288.2 [M+1]+
  • Step-2: Synthesis of tert-butyl 7-((2-ethyl-3-oxo-1-(pyridin-2-yl)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 2-ethyl-6-(methylthio)-1-(pyridin-2-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (200 mg, 0.696 mmol, 1.0 eq) in (2.0 mL) of toluene was added m-CPBA (343 mg, 1.392 mmol, 2.0 eq) and allowed to stir at rt for 30 minutes. tert-butyl 7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (207 mg, 0.835 mmol, 1.2 eq) and DIPEA (0.485 mL, 2.784 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. The formation of precipitates was observed which was filtered to afford the desired compound tert-butyl 7-((2-ethyl-3-oxo-1-(pyridin-2-yl)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (158 mg, 46.60%) as white solid.
  • LCMS: 488.4 [M+1]+
  • Step-3: Synthesis of 2-ethyl-1-(pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride: tert-butyl 7-((2-ethyl-3-oxo-1-(pyridin-2-yl)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (158 mg, 0.324 mmol, 1.0 eq) was dissolved in 4.0 M-HCl in dioxane (2 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound 2-ethyl-1-(pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (120 mg, 80.0%) as brown solid.
  • LCMS: 388.3 [M+1]+; UPLC @ 254 nm=96.60% and @ 220 nm=98.10%.
  • 1H NMR (400 MHz, DMSO-d6) δ 10.35 (brs, 1H), 9.50 (brs, 2H), 8.88 (s, 1H), 8.57 (d, J=4.38 Hz, 1H), 8.14 (t, J=7.89 Hz, 1H), 7.96 (d, J=8.33 Hz, 1H), 7.73 (brs, 1H), 7.49 (d, J=8.77 Hz, 1H), 7.34-7.46 (m, 1H), 7.17 (d, J=8.33 Hz, 1H), 4.24 (brs, 2H), 3.96 (d, J=7.02 Hz, 2H), 3.35 (brs, 2H), 2.96 (brs, 2H), 0.98 (t, J=7.02 Hz, 3H).
  • Example S33. Synthesis of 1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride
  • Figure US20230109104A1-20230406-C02394
  • Step-1: Synthesis of tert-butyl 6-((1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (100 mg, 0.291 mmol, 1.0 eq) in (2.0 mL) of toluene was added m-CPBA (143 mg, 0.582 mmol, 2.0 eq) and allowed to stir at rt for 30 minutes. tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (87 mg, 0.349 mmol, 1.2 eq) and DIPEA (0.20 mL, 1.164 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water (50 mL) and extracted with EtOAc (50 mL×2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-30% EtOAc in Hexane] to afford the desired compound tert-butyl 6-((1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (80 mg, 50.63%) as brown solid.
  • LCMS: 544.4 [M+1]+
  • Step-2: Synthesis of 1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride: tert-butyl 6-((1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (80 mg, 0.147 mmol, 1.0 eq) was dissolved in 4.0 M-HCl (2 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound 1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride (44 mg, 62.85%) as light brown solid.
  • LCMS: 444.4 [M+1]+; UPLC @ 254 nm=96.64% and @ 220 nm=97.96%.
  • 1H NMR (400 MHz, DMSO-d6) δ 10.32 (brs, 1H), 9.02 (brs, 2H), 8.88 (s, 1H), 8.04 (t, J=7.89 Hz, 1H), 7.75-7.87 (m, 2H), 7.51 (d, J=7.02 Hz, 1H), 7.44 (d, J=7.45 Hz, 1H), 7.17 (d, J=8.33 Hz, 1H), 4.23 (brs, 2H), 4.04-4.13 (m, 2H), 3.40 (brs, 2H), 3.00 (brs, 2H), 1.48 (brs, 1H), 1.33 (s, 7H), 0.98 (t, J=7.02 Hz, 3H).
  • Example S34. Synthesis of 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(isoindolin-5-ylamino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02395
  • Step-1: tert-butyl 5-((2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)isoindoline-2-carboxylate: To a stirred solution of 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (0.2 g 0.5790 mmol, 1.0 eq) in (5.0 mL) of toluene was added m-CPBA (0.280 g, 1.158 mmol, 2 eq) and allowed to stir at rt for 30 min. followed by addition of tert-butyl 5-aminoisoindoline-2-carboxylate (0.176 g, 0.7527 mmol, 1.3 eq) and DIPEA (0.3 ml 1.737 mmol, 3.0 eq) and allowed to stir at rt for overnight. The reaction mixture was diluted with water (20 mL) and extracted with EtOAc (2×20 mL). The combined organic layers were washed with water (30 mL), brine (30 mL), dried over Na2SO4, concentrated and purified by combi flash [silica gel-100-200 mesh; elution 0-35% EtOAc in Hexane] to afford the desired compound, tert-butyl 5-((2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)isoindoline-2-carboxylate (0.100 g, 49.52%) as white solid.
  • LCMS: 532.4 [M+1]+
  • Step-2: Synthesis of 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(isoindolin-5-ylamino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: tert-butyl 5-((2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)isoindoline-2-carboxylate (0.100 mg, 0.1881 mmol, 1.0 eq) was dissolved in 4.0 M-HCl in dioxane (3 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure and purified by diethyl ether to afford the desired compound, 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (70 mg, 63.84%) as white solid.
  • LCMS: 432.3 [M+1]+
  • 1H NMR (400 MHz, DMSO-d6): δ 10.42 (br. s., 1H), 9.66 (br. s., 2H), 8.89 (s, 1H), 8.08 (t, J=7.67 Hz, 1H), 7.90 (br. s., 1H), 7.83 (d, J=8.33 Hz, 1H), 7.61-7.70 (m, 2H), 7.35 (d, J=8.77 Hz, 1H), 4.52 (br. s., 2H), 4.46 (br. s., 2H), 4.05 (d, J=7.02 Hz, 2H), 3.57 (s, 2H), 1.44 (s, 6H), 0.98 (t, J=6.80 Hz, 3H)
  • Example S35. Synthesis of 2-ethyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1-(6-(trifluoromethyl)pyridin-2-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride
  • Figure US20230109104A1-20230406-C02396
  • Step-1: Synthesis of 2-ethyl-6-(methylthio)-1-(6-(trifluoromethyl)pyridin-2-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-ethyl-6-(methylthio)-1,2-dihydro-3H-indazol-3-one (500 mg, 2.375 mmol, 1.0 eq) and 2-bromo-6-(trifluoromethyl)pyridine (644 mg, 2.850 mmol, 1.2 eq) in (30 mL) of dioxane was added Potassium carbonate (656 mg, 4.75 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min followed by addition of copper iodide (91 mg, 0.475 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (0.1 mL, 0.951 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 90° C. for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-30% EtOAc in Hexane] to afford the desired compound 2-ethyl-6-(methylthio)-1-(6-(trifluoromethyl)pyridin-2-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (450 mg, 53.25%) as light yellow viscous.
  • LCMS: 356.1 [M+1]+
  • Step-2: Synthesis of tert-butyl 7-((2-ethyl-3-oxo-1-(6-(trifluoromethyl)pyridin-2-yl)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 2-ethyl-6-(methylthio)-1-(6-(trifluoromethyl)pyridin-2-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (200 mg, 0.562 mmol, 1.0 eq) in (3.0 mL) of toluene was added m-CPBA (277 mg, 1.124 mmol, 2.0 eq) and allowed to stir at rt for 30 minutes. tert-butyl 7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (168 mg, 0.675 mmol, 1.2 eq) and DIPEA (0.39 mL, 2.248 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. The formation of precipitates was observed which was filtered to afford the desired compound tert-butyl 7-((2-ethyl-3-oxo-1-(6-(trifluoromethyl)pyridin-2-yl)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (97 mg, 30.99%) as white solid.
  • LCMS: 556.3 [M+1]+
  • Step-3: Synthesis of 2-ethyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1-(6-(trifluoromethyl)pyridin-2-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride: tert-butyl 7-((2-ethyl-3-oxo-1-(6-(trifluoromethyl)pyridin-2-yl)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (95 mg, 0.171 mmol, 1.0 eq) was dissolved in 4.0 M-HCl in dioxane (2 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound 2-ethyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1-(6-(trifluoromethyl)pyridin-2-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride (60 mg, 71.42%) as light yellow solid.
  • LCMS: 456.3 [M+1]+; UPLC @ 254 nm=98.86% and @ 220 nm=99.25%.
  • 1H NMR (400 MHz, DMSO-d6) δ 10.44 (brs, 1H), 9.37 (brs, 2H), 8.92 (s, 1H), 8.31-8.44 (m, 2H), 7.90 (d, J=7.45 Hz, 1H), 7.65 (brs, 1H), 7.55 (d, J=8.33 Hz, 1H), 7.21 (d, J=8.33 Hz, 1H), 4.28 (brs, 3H), 3.92-4.05 (m, 3H), 3.37 (brs, 2H), 2.93-3.04 (m, 2H), 1.02 (t, J=7.02 Hz, 3H).
  • Example S36. Synthesis of 6-((2-(dimethylamino)-2,3-dihydro-JH-inden-5-yl)amino)-2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02397
  • To a stirred solution of 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (195 mg, 0565 mmol, 1.0 eq) in (3.0 mL) of toluene was added m-CPBA (278 mg, 1.13 mmol, 2.0 eq) and allowed to stir at rt for 30 minutes. N2,N2-dimethyl-2,3-dihydro-1H-indene-2,5-diamine (140 mg, 0.679 mmol, 1.2 eq) and DIPEA (0.395 mL, 2.26 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water (50 mL) and extracted with EtOAc (50 mL×2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by reverse phase purification to afford the desired compound 6-((2-(dimethylamino)-2,3-dihydro-1H-inden-5-yl)amino)-2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (5.0 mg, 1.87%) as brown solid.
  • LCMS: 474.3 [M+1]+; UPLC @ 254 nm=76.60% and @ 220 nm=85.51%.
  • 1H NMR (400 MHz, DMSO-d6) δ 10.18 (brs, 1H), 8.84 (s, 1H), 8.32 (d, J=9.65 Hz, 1H), 7.99-8.07 (m, 1H), 7.82 (d, J=7.89 Hz, 1H), 7.65 (d, J=7.89 Hz, 2H), 7.42 (d, J=7.89 Hz, 1H), 7.13 (d, J=8.33 Hz, 1H), 4.01-4.09 (m, 2H), 3.17 (s, 2H), 2.88-3.11 (m, 4H), 2.60-2.85 (m, 3H), 2.22 (s, 6H), 1.32-1.50 (m, 6H), 1.23 (brs, 1H), 0.97 (t, J=7.02 Hz, 3H).
  • Example S37. Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-2-ethyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02398
  • Step-1: Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-2-ethyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-ethyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (300 mg, 1.42 mmol) and 2-(tert-butyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine (445 mg, 2.85 mmol) in DCM (50 mL) was added 2,2-bipyridine (445 mg, 2.85 mmol), copper acetate (518 mg, 2.85 mmol) and Na2CO3 (451 mg, 4.26 mmol). The reaction mixture was stirred at rt for 24 h in open air. The progress of reaction was monitored by TLC. After completion, the reaction mixture was filtered over celite to remove inorganic impurities. The filtrate was washed with water, dried over Na2SO4, concentrated and purified by column chromatography (Combiflash, Elution: 0-30% EtOAc in Hexane) to afford the desired product, 1-(2-(tert-butyl)pyridin-4-yl)-2-ethyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (180 mg, 36.73%) as an off white solid.
  • LCMS: 344.2 [M+1]+
  • Step-2: Synthesis of tert-butyl 7-((1-(2-(tert-butyl)pyridin-4-yl)-2-ethyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(2-(tert-butyl)pyridin-4-yl)-2-ethyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (100 mg, 0.291 mmol, 1.0 eq) in (6.0 mL) of toluene was added m-CPBA (100 mg, 0.582 mmol, 2.0 eq) and allowed to stir at rt for 1 h. tert-butyl 7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (87 mg, 0.349 mmol, 1.2 eq) and DIPEA (0.2 mL, 1.164 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 7-((1-(2-(tert-butyl)pyridin-4-yl)-2-ethyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (27 mg, 17.05%) as an off white solid.
  • LCMS: 544.3 [M+1]+
  • Step-3: Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-2-ethyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride: tert-butyl 7-((1-(2-(tert-butyl)pyridin-4-yl)-2-ethyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (27 mg, 0.049 mmol, 1.0 eq) was dissolved in dioxane (0.5 mL), followed by dropwise addition of 4.0 M-HCl (0.5 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(2-(tert-butyl)pyridin-4-yl)-2-ethyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (18 mg, 70.20%) as light yellow solid.
  • LCMS: 444.4 [M+1]+; UPLC @ 254 nm=94.74% and @ 220 nm=93.98%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.54 (br. s., 1H), 9.47 (br. s., 2H), 8.94 (s, 1H), 8.82 (d, J=6.14 Hz, 1H), 7.88 (br. s., 1H), 7.59 (d, J=7.45 Hz, 2H), 7.53 (br. s., 1H), 7.22 (d, J=7.89 Hz, 1H), 4.28 (br. s., 4H), 3.83 (d, J=7.02 Hz, 2H), 3.36 (br. s., 2H), 2.99 (br. s., 2H), 1.42 (br. s., 9H), 0.97 (t, J=6.80 Hz, 3H)
  • Example S38. 6-{[7-(dimethylamino)-5,6,7,8-tetrahydronaphthalen-2-yl]amino}-2-ethyl-1-[6-(2-hydroxypropan-2-yl)pyridin-2-yl]-1H, 2H, 3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02399
    Figure US20230109104A1-20230406-C02400
  • Step-1: Synthesis of N,N-dimethyl-1,2,3,4-tetrahydronaphthalen-2-amine: 46 mL of a methanol solution of NaCNBH3 (0.87 g, 13.825 mmol, 1.0 eq) and ZnCl2 (0.94 g, 6.914 mmol, 0.5 eq) and 6.9 mL of a THF solution of 2M dimethyl amine, were added to a THF solution (10 mL) of 3,4-dihydronaphthalen-2(1H)-one (2.02 g, 13.828 mmol, 1.0 eq) at 0° C. The resulting mixture was stirred at rt for overnight. The progress of reaction was monitored by LCMS. The reaction mixture was concentrated, acidified with 1N-HCl (50 mL), washed with EtOAc (2×50 mL), then made alkaline with aqueous 5N NaOH solution, extracted with EtOAc (2×50 mL). The combined organic layers were washed with water (50 mL), with brine (50 mL), dried over Na2SO4 concentrated and purified by combiflash chromatography [basic alumina, elution 0-10% EtOAc in Hexane] to afford the desired compound N,N-dimethyl-1,2,3,4-tetrahydronaphthalen-2-amine (845 mg, 34.77%) as colorless liquid.
  • LCMS: 176.3 4 [M+1]+
  • Step-2: Synthesis of N,N-dimethyl-6-nitro-1,2,3,4-tetrahydronaphthalen-2-amine and N,N-dimethyl-7-nitro-1,2,3,4-tetrahydronaphthalen-2-amine: 1.2 mL of nitric acid (specific gravity, 1.41) to a TFA (5.0 mL) solution of N,N-dimethyl-1,2,3,4-tetrahydronaphthalen-2-amine (0.840 g, 4.792 mmol, 1.0 eq) at 0° C. The resulting mixture was stirred at rt for overnight. The progress of reaction was monitored by LCMS. The reaction mixture was poured into ice cold water (100 mL), then made alkaline with aqueous 5M solution of NaOH added there to and extracted with EtOAc (2×100 mL). The combined organic layers were washed with water (50 mL), with brine (50 mL), dried over Na2SO4 concentrated and purified by combiflash chromatography [basic alumina, elution 0-10% EtOAc in Hexane] to afford the mixture of N,N-dimethyl-6-nitro-1,2,3,4-tetrahydronaphthalen-2-amine and N,N-dimethyl-7-nitro-1,2,3,4-tetrahydronaphthalen-2-amine (900 mg, 85.30%) as brown viscous.
  • LCMS: 221.3 4 [M+1]+
  • Step-3: Synthesis of N2,N2-dimethyl-1,2,3,4-tetrahydronaphthalene-2,6-diamine and N2,N2-dimethyl-1,2,3,4-tetrahydronaphthalene-2,7-diamine: 130 mg of palladium hydroxide-carbon (20%) was added to a solution of N,N-dimethyl-6-nitro-1,2,3,4-tetrahydronaphthalen-2-amine and N,N-dimethyl-7-nitro-1,2,3,4-tetrahydronaphthalen-2-amine (0.900 g, 4.085 mmol, 1.0 eq) in EtOH (10 mL) at rt. The resulting mixture was stirred at rt for overnight in hydrogen atmosphere. The catalyst was removed though filtration, the filtrate was concentrated, and stirred in hexane, filtered and concentrated to afford the mixture of N2,N2-dimethyl-1,2,3,4-tetrahydronaphthalene-2,6-diamine and N2,N2-dimethyl-1,2,3,4-tetrahydro naphthalene-2,7-diamine (730 mg, 93.95%) as brown viscous.
  • LCMS: 191.3 4 [M+1]+
  • Step-4: Synthesis of 6-{[7-(dimethylamino)-5,6,7,8-tetrahydronaphthalen-2-yl]amino}-2-ethyl-1-[6-(2-hydroxypropan-2-yl)pyridin-2-yl]-1H, 2H, 3H-pyrazolo[3,4-d]pyrimidin-3-one (PK-II): To a stirred solution of 2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (1.05 g, 3.065 mmol, 1.0 eq) in (20.0 mL) of toluene was added m-CPBA (1.51 g, 6.13 mmol, 2.0 eq) and allowed to stir at rt for 30 minutes. N2,N2-dimethyl-1,2,3,4-tetrahydronaphthalene-2,6-diamine and N2,N2-dimethyl-1,2,3,4-tetrahydro naphthalene-2,7-diamine (700 mg, 3.678 mmol, 1.2 eq) and DIPEA (2.14 mL, 12.26 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the mixture of 6-((6-(dimethylamino)-5,6,7,8-tetrahydronaphthalen-2-yl)amino)-2-ethyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (PK-I) and 6-((7-(dimethylamino)-5,6,7,8-tetrahydronaphthalen-2-yl)amino)-2-ethyl-1-(6-(2-hydroxypropan-2-yl) pyridin-2-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (PK-II), which was purified by reverse phase purification to obtain peak-II (5.0 mg, 1.87%) as brown solid.
  • LCMS: 474.3 4 [M+1]+; UPLC @ 254 nm=76.60% and @ 220 nm=85.51%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.18 (brs, 1H), 8.84 (s, 1H), 8.32 (d, J=9.65 Hz, 1H), 7.99-8.07 (m, 1H), 7.82 (d, J=7.89 Hz, 1H), 7.65 (d, J=7.89 Hz, 2H), 7.42 (d, J=7.89 Hz, 1H), 7.13 (d, J=8.33 Hz, 1H), 4.01-4.09 (m, 2H), 3.17 (s, 2H), 2.88-3.11 (m, 4H), 2.60-2.85 (m, 3H), 2.22 (s, 6H), 1.32-1.50 (m, 6H), 1.23 (brs, 1H), 0.97 (t, J=7.02 Hz, 3H).
  • Example S39. Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-2-ethyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02401
  • Step-1: Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-2-ethyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-ethyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (300 mg, 1.42 mmol) and 2-(tert-butyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine (445 mg, 2.85 mmol) in DCM (50 mL) was added 2,2-bipyridine (445 mg, 2.85 mmol), copper acetate (518 mg, 2.85 mmol) and Na2CO3 (451 mg, 4.26 mmol). The reaction mixture was stirred at rt for 24 h in open air. The progress of reaction was monitored by TLC. After completion, the reaction mixture was filtered over celite to remove inorganic impurities. The filtrate was washed with water, dried over Na2SO4, concentrated and purified by column chromatography (Combiflash, Elution: 0-30% EtOAc in Hexane) to afford the desired product, 1-(2-(tert-butyl)pyridin-4-yl)-2-ethyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (180 mg, 36.73%) as an off white solid.
  • LCMS: 344.2 [M+1]+
  • Step-2: Synthesis of tert-butyl 6-((1-(2-(tert-butyl)pyridin-4-yl)-2-ethyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(2-(tert-butyl)pyridin-4-yl)-2-ethyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (100 mg, 0.291 mmol, 1.0 eq) in (6.0 mL) of toluene was added m-CPBA (100 mg, 0.582 mmol, 2.0 eq) and allowed to stir at rt for 1 h. tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (87 mg, 0.349 mmol, 1.2 eq) and DIPEA (0.2 mL, 1.164 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 6-((1-(2-(tert-butyl)pyridin-4-yl)-2-ethyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (50 mg, 33.26%) as an off white solid.
  • LCMS: 544.3 [M+1]+
  • Step-3: Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-2-ethyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride: tert-butyl 6-((1-(2-(tert-butyl)pyridin-4-yl)-2-ethyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (100 mg, 0.179 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (2 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(2-(tert-butyl)pyridin-4-yl)-2-ethyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (70 mg, 73.59%) as light yellow solid.
  • LCMS: 444.3 [M+1]+; UPLC @ 254 nm=93.80% and @ 220 nm=93.41%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.48 (br. s., 1H), 9.33 (br. s., 2H), 8.94 (s, 1H), 8.77 (d, J=5.70 Hz, 1H), 7.79 (br. s., 1H), 7.58 (br. s., 3H), 7.21 (s, 1H), 4.23 (br. s., 2H), 3.02 (br. s., 2H), 1.28-1.57 (m, 9H), 0.97 (t, J=6.80 Hz, 3H).
  • Example S40. Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02402
  • Step-1: Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (600 mg, 2.67 mmol) and 2-(tert-butyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine (1.39 g, 5.35 mmol) in DCM (100 mL) was added 2,2-bipyridine (836 mg, 5.35 mmol), copper acetate (972 mg, 5.35 mmol) and Na2CO3 (849 mg, 8.01 mmol). The reaction mixture was stirred at rt for 24 h in open air. The progress of reaction was monitored by TLC. After completion, the reaction mixture was filtered over celite to remove inorganic impurities. The filtrate was washed with water, dried over Na2SO4, concentrated and purified by column chromatography (Combiflash, Elution: 0-30% EtOAc in Hexane) to afford the desired product, 1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (450 mg, 47.05%) as an off white solid.
  • LCMS: 358.2 [M+1]+
  • Step-2: Synthesis of tert-butyl 7-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (225 mg, 0.629 mmol, 1.0 eq) in (6.0 mL) of toluene was added m-CPBA (217 mg, 1.258 mmol, 2.0 eq) and allowed to stir at rt for 1 h. tert-butyl 7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (188 mg, 0.755 mmol, 1.2 eq) and DIPEA (0.43 mL, 2.516 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound tert-butyl 7-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (80 mg, 22.78%) as an off white solid.
  • LCMS: 558.3 [M+1]+
  • Step-3: Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride: tert-butyl 7-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (80 mg, 0.143 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (2 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (45 mg, 59.14%) as light yellow solid.
  • LCMS: 458.4 [M+1]+; UPLC @ 254 nm=95.58% and @ 220 nm=96.94%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.56 (br. s., 1H), 9.57 (br. s., 3H), 8.90 (s, 1H), 8.82 (br. s., 1H), 7.90 (br. s., 1H), 7.52 (br. s., 1H), 7.22 (d, J=8.33 Hz, 1H), 4.28 (br. s., 2H), 3.95 (br. s., 1H), 3.36 (br. s., 2H), 2.99 (br. s., 2H), 1.29-1.53 (m, 15H)
  • Example S41. Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02403
  • Step-1: Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (600 mg, 2.67 mmol) and 2-(tert-butyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine (1.39 g, 5.35 mmol) in DCM (100 mL) was added 2,2-bipyridine (836 mg, 5.35 mmol), copper acetate (972 mg, 5.35 mmol) and Na2CO3 (849 mg, 8.01 mmol). The reaction mixture was stirred at rt for 24 h in open air. The progress of reaction was monitored by TLC. After completion, the reaction mixture was filtered over celite to remove inorganic impurities. The filtrate was washed with water, dried over Na2SO4, concentrated and purified by column chromatography (Combiflash, Elution: 0-30% EtOAc in Hexane) to afford the desired product, 1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (450 mg, 47.05%) as an off white solid.
  • LCMS: 358.2 [M+1]+
  • Step-2: Synthesis of tert-butyl 6-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (225 mg, 0.629 mmol, 1.0 eq) in (6.0 mL) of toluene was added m-CPBA (217 mg, 1.258 mmol, 2.0 eq) and allowed to stir at rt for 1 h. tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (188 mg, 0.755 mmol, 1.2 eq) and DIPEA (0.43 mL, 2.516 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 6-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (100 mg, 28.48%) as an off white solid.
  • LCMS: 558.3 [M+1]+
  • Step-3: Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride: tert-butyl 6-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (100 mg, 0.179 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (2 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (70 mg, 73.59%) as light yellow solid.
  • LCMS: 458.4 [M+1]+; UPLC @ 254 nm=95.47% and @ 220 nm=97.76%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.58 (br. s., 1H), 9.61 (br. s., 2H), 8.91 (s, 1H), 8.81 (d, J=6.14 Hz, 1H), 7.93 (br. s., 1H), 7.66 (br. s., 1H), 7.58 (d, J=7.89 Hz, 1H), 7.52 (br. s., 1H), 7.23 (d, J=8.33 Hz, 1H), 4.23 (br. s., 3H), 3.95 (td, J=6.80, 13.59 Hz, 1H), 3.36 (br. s., 2H), 3.04 (br. s., 2H), 1.32-1.58 (m, 15H)
  • Example S42. Synthesis of 1-(6-(2-fluoropropan-2-yl)pyridin-2-yl)-2-isopropyl-6-(1,2,3,4-tetrahydroisoquinolin-7-ylamino)-JH-pyrazolo[3,4-d]pyrimidin-3(2H)-one dihydrochloride
  • Figure US20230109104A1-20230406-C02404
  • Step-1: Synthesis of tert-butyl 7-((1-(6-(2-fluoropropan-2-yl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(6-(tert-butyl)pyridin-2-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (150 mg, 0.4155 mmol, 1.0 eq) in (5.0 mL) of toluene was added m-CPBA (143 mg, 0.831 mmol, 2.0 eq) and allowed to stir at rt for 30 minutes. tert-butyl 7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (124 mg, 0.5 mmol, 1.2 eq) and DIPEA (0.3 mL, 1.662 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 6-((2-isopropyl-1-(6-(2-fluoropropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (80 mg, 34.32%) as an off white solid.
  • 1H NMR (400 MHz, DMSO-d6): δ10.22 (brs, 1H), 8.81 (s, 1H), 8.10-8.17 (m, 1H), 7.96 (d, J=8.77 Hz, 1H), 7.7-7.3 (m, 3H), 7.06 (d, 1H), 4.5 (brs, 2H), 4.06 (s, 1H), 3.6 (brs, 2H), 2.7 (brs, 2H), 1.7 (d, 6H), 1.5-1.3 (m, 15H).
  • Step-2: Synthesis of 1-(6-(2-fluoropropan-2-yl)pyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: tert-butyl 6-((1-(6-(tert-butyl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (80 mg, 0. mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (2 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 2-isopropyl-1-(6-(2-fluoropropan-2-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (45 mg, 30.16%) as light yellow solid.
  • LCMS: 462.3 [M+1]+; UPLC @ 254 nm=94.68% and @ 220 nm=97.66%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.22 (brs, 1H), 9.22 (brs, 2H), 8.81 (s, 1H), 8.20-8.10 (m, 1H), 7.88 (d, 1H), 7.64 (brs, 1H), 7.55-7.42 (m, 2H), 7.18 (d, 1H), 4.28-4.16 (m, 3H), 3.4 (brs, 2H), 2.98 (brs, 2H), 1.66 (s, 3H), 1.62 (s, 3H), 1.40 (d, 6H).
  • Example S43. Synthesis of 2-isopropyl-1-(6-(2-fluoropropan-2-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02405
  • Step)-1: Synthesis of 2-bromo-6-(2-fluoropropan-2-yl)pyridine: To a stirred solution of 2-(6-bromo-pyridine-2-yl)-propane-2-ol (0.500 g, 2.313 mmol, 1.0 eq) in DCM (10 mL), DAST (0.36 mL, 2.545 mmol, 1.1 eq) was added at −78° C. The reaction mixture was stirred at rt for 12 h. After completion of reaction, the reaction mixture was quenched with saturated sodium bicarbonate solution and was extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 ML), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-10% EtOAc in hexane] to afford the desired compound, tert-butyl 6-((2-cyclopropyl-1-(6-(2-fluoropropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (355 mg, 70.43%) as colorless liquid. LCMS: 217.99 [M+1]+
  • Step-2: Synthesis of 2-isopropyl-1-(6-(2-fluoropropan-2-yl)pyridin-2-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (300 mg, 1.337 mmol, 1.0 eq) and 2-bromo-6-(2-fluoropropan-2-yl)pyridine (294 mg, 1.36 mmol, 1.20 eq) in (8 mL) of dioxane was added potassium carbonate (370 mg, 2.674 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min, followed by addition of copper iodide (51 mg, 0.27 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (47 mg, 0.535 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 90° C. for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-30% EtOAc in hexane] to afford the desired product, 2-isopropyl-1-(6-(2-fluoropropan-2-yl)pyridin-2-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (310 mg, 64%) as an off white solid.
  • LCMS: 362.3 [M+1]+
  • Step-3: Synthesis of tert-butyl 6-((2-isopropyl-1-(6-(2-fluoropropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(6-(tert-butyl)pyridin-2-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (150 mg, 0.4155 mmol, 1.0 eq) in (5.0 mL) of toluene was added m-CPBA (143 mg, 0.831 mmol, 2.0 eq) and allowed to stir at rt for 30 minutes. tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (124 mg, 0.5 mmol, 1.2 eq) and DIPEA (0.3 mL, 1.662 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 6-((2-isopropyl-1-(6-(2-fluoropropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (80 mg, %) as an off white solid.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.22 (brs, 1H), 8.81 (s, 1H), 8.10-8.17 (m, 1H), 7.88 (d, J=8.77 Hz, 1H), 7.6-7.4 (m, 3H), 7.10 (brs, 1H), 4.9 (brs, 1H), 4.10-4.46 (m, 4H), 3.4 (brs, 2H), 1.86 (d, 6H), 1.4 (m, 15H).
  • Step-4: Synthesis of 2-isopropyl-1-(6-(2-fluoropropan-2-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: tert-butyl 6-((1-(6-(tert-butyl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (80 mg, 0. mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (2 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 2-isopropyl-1-(6-(2-fluoropropan-2-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (45 mg, 30.16%) as light yellow solid.
  • LCMS: 462.3 [M+1]+; UPLC @ 254 nm=94.72% and @ 220 nm=96.31%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.22 (brs, 1H), 9.22 (brs, 2H), 8.81 (s, 1H), 8.10-8.17 (m, 1H), 7.88 (d, 1H), 7.69 (brs, 1H), 7.55-7.40 (m, 2H), 7.18 (d, 1H), 4.22-4.16 (m, 3H), 3.4 (brs, 2H), 3.24 (brs, 2H), 1.66 (s, 3H), 1.62 (s, 3H), 1.40 (d, 6H).
  • Example S44. Synthesis of 1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-6-((5,6,7,8-tetrahydro-1,6-naphthyridin-3-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one trihydrochloride
  • Figure US20230109104A1-20230406-C02406
  • Step-1: Synthesis of tert-butyl 3-((1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-7,8-dihydro-1,6-naphthyridine-6(5H)-carboxylate: To a stirred solution of 1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-6-(methylthio)-1,2-dihydro-3H-indazol-3-one (200 mg, 0.582 mmol, 1.0 eq) in (2.0 mL) of toluene was added m-CPBA (287 mg, 1.164 mmol, 2.0 eq) and allowed to stir at rt for 30 minutes. tert-butyl 3-amino-7,8-dihydro-1,6-naphthyridine-6(5H)-carboxylate (174 mg, 0.698 mmol, 1.2 eq) and DIPEA (0.40 mL, 2.328 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. The formation of precipitates were observed which were filtered to afford the desired compound tert-butyl 3-((1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-7,8-dihydro-1,6-naphthyridine-6(5H)-carboxylate (50 mg, 15.82%) as brown solid.
  • LCMS: 545.4 [M+1]+
  • Step-2: Synthesis of 1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-6-((5,6,7,8-tetrahydro-1,6-naphthyridin-3-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one trihydrochloride: tert-butyl 3-((1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-7,8-dihydro-1,6-naphthyridine-6(5H)-carboxylate (45 mg, 0.0826 mmol, 1.0 eq) was dissolved in 4.0 M-HCl (1 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound 1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-6-((5,6,7,8-tetrahydro-1,6-naphthyridin-3-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one trihydrochloride (35 mg, 87.50%) as yellow solid.
  • LCMS: 445.3.4 [M+1]+; UPLC @ 254 nm=92.74% and @ 220 nm=95.09%.
  • 1H NMR (400 MHz, DMSO-d6) δ 10.53 (brs, 1H), 9.27 (brs, 2H), 8.93 (s, 1H), 8.71 (brs, 1H), 8.17 (brs, 1H), 8.05 (t, J=7.89 Hz, 1H), 7.82 (d, J=7.89 Hz, 1H), 7.45 (d, J=7.45 Hz, 1H), 4.35 (brs, 2H), 4.08 (d, J=6.58 Hz, 2H), 3.51 (brs, 2H), 3.07 (brs, 2H), 1.32 (s, 9H), 0.98 (t, J=6.80 Hz, 3H).
  • Example S45. Synthesis of 1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-6-(isoindolin-5-ylamino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02407
  • Step-1: Synthesis of tert-butyl 5-((1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)isoindoline-2-carboxylate: To a stirred solution of 1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-6-(methylthio)-1,2-dihydro-3H-indazol-3-one (200 mg, 0.582 mmol, 1.0 eq) in (2.0 mL) of toluene was added m-CPBA (287 mg, 1.164 mmol, 2.0 eq) and allowed to stir at rt for 30 minutes. tert-butyl 5-aminoisoindoline-2-carboxylate (164 mg, 0.698 mmol, 1.2 eq) and DIPEA (0.40 mL, 2.328 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. The formation of precipitates were observed which were filtered to afford the desired compound tert-butyl 5-((1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)isoindoline-2-carboxylate (135 mg, 43.83%) as brown solid.
  • LCMS: 530.4 [M+1]+
  • Step-2: Synthesis of 1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-6-(isoindolin-5-ylamino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride: tert-butyl 5-((1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)isoindoline-2-carboxylate (130 mg, 1.321 mmol, 1.0 eq) was dissolved in 4.0 M-HCl (1.5 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound 1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-6-(isoindolin-5-ylamino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (105 mg, 94.92%) as yellow solid.
  • LCMS: 430.4 [M+1]+; UPLC @ 254 nm=95.23% and @ 220 nm=96.75%.
  • 1H NMR (400 MHz, DMSO-d6) δ 10.42 (brs, 1H), 9.91 (brs, 2H), 8.89 (s, 1H), 8.05 (t, J=7.45 Hz, 1H), 7.89 (brs, 1H), 7.81 (d, J=7.89 Hz, 1H), 7.62 (d, J=7.89 Hz, 1H), 7.43 (d, J=7.45 Hz, 1H), 7.35 (d, J=7.89 Hz, 1H), 4.51 (brs, 2H), 4.45 (brs, 3H), 4.08 (d, J=6.58 Hz, 3H), 1.23-1.38 (m, 9H), 0.98 (t, J=6.58 Hz, 3H).
  • Example S46 Synthesis of 1-(6-(tert-butyl)pyridin-2-yl)-2-cyclopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02408
  • Step-1: Synthesis of 1-(6-(tert-butyl)pyridin-2-yl)-2-cyclopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-cyclopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (500 mg, 2.242 mmol, 1.0 eq) and 2-bromo-6-(tert-butyl)pyridine (586 mg, 2.690 mmol, 1.20 eq) in (12 mL) of dioxane was added potassium carbonate (619 mg, 4.484 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min followed by addition of copper iodide (85 mg, 0.448 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (80 mg, 0.896 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 90° C. for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 ML), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-30% EtOAc in hexane] to afford the desired product, 1-(6-(tert-butyl)pyridin-2-yl)-2-cyclopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (300 mg, 37.51%) as light yellow viscous.
  • LCMS: 356.2 [M+1]+
  • Step-2: Synthesis of tert-butyl 6-((1-(6-(tert-butyl)pyridin-2-yl)-2-cyclopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(6-(tert-butyl)pyridin-2-yl)-2-cyclopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (140 mg, 0.393 mmol, 1.0 eq) in (2.0 mL) of toluene was added m-CPBA (135 mg, 0.786 mmol, 2.0 eq) and allowed to stir at rt for 30 minutes. tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (195 mg, 0.786 mmol, 1.2 eq) and DIPEA (0.27 mL, 1.572 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 6-((1-(6-(tert-butyl)pyridin-2-yl)-2-cyclopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (76 mg, 34.72%) as white solid.
  • LCMS: 556.3 [M+1]+
  • Step-3: Synthesis of 1-(6-(tert-butyl)pyridin-2-yl)-2-cyclopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride: tert-butyl 6-((1-(6-(tert-butyl)pyridin-2-yl)-2-cyclopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (76 mg, 0.136 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (2 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(6-(tert-butyl)pyridin-2-yl)-2-cyclopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (52 mg, 71.65%) as light yellow solid.
  • LCMS: 456.3 [M+1]+; UPLC @ 254 nm=96.94% and @ 220 nm=96.60%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.25 (brs, 1H), 9.24 (brs, 2H), 8.81 (s, 1H), 8.03 (t, J=7.89 Hz, 1H), 7.70-7.79 (m, 2H), 7.40-7.51 (m, 2H), 7.16 (d, J=8.33 Hz, 1H), 4.21 (brs, 2H), 3.37 (brs, 2H), 3.23 (brs, 2H), 2.95-3.04 (m, 2H), 1.32 (s, 9H), 0.82 (brs, 4H).
  • Example S47. Synthesis of 1-(6-(tert-butyl)pyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02409
  • Step-1: Synthesis of 1-(6-(tert-butyl)pyridin-2-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (400 mg, 1.78 mmol, 1.0 eq) and 2-bromo-6-(tert-butyl)pyridine (458 mg, 2.14 mmol, 1.20 eq) in (12 mL) of dioxane was added potassium carbonate (492 mg, 3.56 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min followed by addition of copper iodide (68 mg, 0.356 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (63 mg, 0.712 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 90° C. for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 ML), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-30% EtOAc in hexane] to afford the desired product, 1-(6-(tert-butyl)pyridin-2-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (366 mg, 57.40%) as colorless liquid. LCMS: 358.2 [M+1]+
  • Step-2: Synthesis of tert-butyl 7-((1-(6-(tert-butyl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(6-(tert-butyl)pyridin-2-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (180 mg, 0.503 mmol, 1.0 eq) in (6.0 mL) of toluene was added m-CPBA (174 mg, 1.006 mmol, 2.0 eq) and allowed to stir at rt for 30 minutes. tert-butyl 7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (150 mg, 0.604 mmol, 1.2 eq) and DIPEA (0.34 mL, 2.012 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 7-((1-(6-(tert-butyl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (127 mg, 45.22%) as an off white solid.
  • LCMS: 558.3 [M+1]+
  • Step-3: Synthesis of 1-(6-(tert-butyl)pyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride: tert-butyl 7-((1-(6-(tert-butyl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (127 mg, 0.227 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (2 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(6-(tert-butyl)pyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (96 mg, 79.47%) as light yellow solid.
  • LCMS: 458.3 [M+1]+; UPLC @ 254 nm=96.17% and @ 220 nm=96.72%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.31 (brs, 1H), 9.20 (brs, 2H), 8.83 (s, 1H), 8.04 (t, J=7.89 Hz, 1H), 7.76 (d, J=8.33 Hz, 1H), 7.69 (brs, 1H), 7.50 (d, J=7.89 Hz, 1H), 7.44 (d, J=7.45 Hz, 1H), 7.16 (d, J=8.33 Hz, 1H), 4.25 (brs, 2H), 4.12-4.21 (m, 1H), 3.36 (brs, 2H), 2.95 (t, J=5.70 Hz, 2H), 1.38 (d, J=6.58 Hz, 4H), 1.32 (s, 6H).
  • Example S48. Synthesis of 1-(6-(tert-butyl)pyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02410
  • Step-1: Synthesis of 1-(6-(tert-butyl)pyridin-2-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (400 mg, 1.78 mmol, 1.0 eq) and 2-bromo-6-(tert-butyl)pyridine (458 mg, 2.14 mmol, 1.20 eq) in (12 mL) of dioxane was added potassium carbonate (492 mg, 3.56 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min followed by addition of copper iodide (68 mg, 0.356 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (63 mg, 0.712 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 90° C. for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 ML), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-30% EtOAc in hexane] to afford the desired product, 1-(6-(tert-butyl)pyridin-2-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (366 mg, 57.40%) as colorless liquid.
  • LCMS: 358.2 [M+1]+
  • Step-2: Synthesis of tert-butyl 6-((1-(6-(tert-butyl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(6-(tert-butyl)pyridin-2-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (180 mg, 0.503 mmol, 1.0 eq) in (6.0 mL) of toluene was added m-CPBA (174 mg, 1.006 mmol, 2.0 eq) and allowed to stir at rt for 30 minutes. tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (150 mg, 0.604 mmol, 1.2 eq) and DIPEA (0.34 mL, 2.012 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 6-((1-(6-(tert-butyl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (120 mg, 42.73%) as an off white solid.
  • LCMS: 558.3 [M+1]+
  • Step-3: Synthesis of 1-(6-(tert-butyl)pyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride: tert-butyl 7-((1-(6-(tert-butyl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (120 mg, 0.215 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (2 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(6-(tert-butyl)pyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (81 mg, 70.96%) as light yellow solid.
  • LCMS: 458.3 [M+1]+; UPLC @ 254 nm=95.25% and @ 220 nm=97.03%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.30 (brs, 1H), 9.29 (brs, 2H), 8.83 (s, 1H), 8.02 (d, J=7.89 Hz, 1H), 7.74 (d, J=7.45 Hz, 2H), 7.41-7.54 (m, 2H), 7.15 (d, J=8.33 Hz, 1H), 4.20 (brs, 3H), 3.37 (brs, 2H), 2.98 (brs, 2H), 1.38 (d, J=6.14 Hz, 4H), 1.32 (s, 6H)
  • Example S49. Synthesis of 2-cyclopropyl-1-(6-(2-fluoropropan-2-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02411
  • Step-1: Synthesis of 2-bromo-6-(2-fluoropropan-2-yl)pyridine: To a stirred solution of 2-(6-bromo-pyridine-2-yl)-propane-2-ol (0.500 g, 2.313 mmol, 1.0 eq) in DCM (10 mL), DAST (0.36 mL, 2.545 mmol, 1.1 eq) was added at −78° C. The reaction mixture was stirred at rt for 12 h. After completion of reaction, the reaction mixture was quenched with saturated sodium bicarbonate solution and was extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 ML), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-10% EtOAc in hexane] to afford the desired compound, tert-butyl 6-((2-cyclopropyl-1-(6-(2-fluoropropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (355 mg, 70.43%) as colorless liquid.
  • LCMS: 217.99 [M+1]+
  • Step-2: Synthesis of 2-cyclopropyl-1-(6-(2-fluoropropan-2-yl)pyridin-2-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-on: To a stirred solution of 2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (300 mg, 1.345 mmol, 1.0 eq) and 2-bromo-6-(2-fluoropropan-2-yl)pyridine (351 mg, 1.614 mmol, 1.20 eq) in (10 mL) of dioxane was added potassium carbonate (371 mg, 2.690 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min, followed by addition of copper iodide (51 mg, 0.269 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (47 mg, 0.538 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 90° C. for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-30% EtOAc in hexane] to afford the desired product, 2-cyclopropyl-1-(6-(2-fluoropropan-2-yl)pyridin-2-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (260 mg, 52.63%) as an off white solid.
  • LCMS: (M+1): 359.12 [M+1]+
  • Step-3: Synthesis of tert-butyl 6-((2-cyclopropyl-1-(6-(2-fluoropropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(6-(tert-butyl)pyridin-2-yl)-2-cyclopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (260 mg, 0.722 mmol, 1.0 eq) in (5.0 mL) of toluene was added m-CPBA (249 mg, 1.444 mmol, 2.0 eq) and allowed to stir at rt for 30 minutes. tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (358 mg, 1.444 mmol, 2.0 eq) and DIPEA (0.5 mL, 2.888 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 6-((2-cyclopropyl-1-(6-(2-fluoropropan-2-yl)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (100 mg, 24.75%) as an off white solid.
  • LCMS: 560.27 [M+1]+
  • Step-4: Synthesis of 2-cyclopropyl-1-(6-(2-fluoropropan-2-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: tert-butyl 6-((1-(6-(tert-butyl)pyridin-2-yl)-2-cyclopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (100 mg, 0.178 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (2 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 2-cyclopropyl-1-(6-(2-fluoropropan-2-yl)pyridin-2-yl)-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (25 mg, 30.16%) as light yellow solid.
  • LCMS: 460.22 [M+1]+; UPLC @ 254 nm=96.72% and @ 220 nm=98.03%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.22 (brs, 1H), 8.81 (s, 1H), 8.10-8.17 (m, 1H), 7.88 (d, J=8.77 Hz, 2H), 7.69 (brs, 1H), 7.55 (brs, 1H), 7.43 (brs, 1H), 7.10 (brs, 1H), 4.07 (brs, 2H), 3.24 (brs, 2H), 2.88 (brs, 2H), 1.70 (s, 3H), 1.64 (s, 3H), 0.82 (d, J=5.26 Hz, 4H).
  • Example S50. Synthesis of 1-(6-(tert-butyl)pyridin-2-yl)-2-cyclopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02412
  • Step-1: Synthesis of 1-(6-(tert-butyl)pyridin-2-yl)-2-cyclopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-cyclopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (500 mg, 2.242 mmol, 1.0 eq) and 2-bromo-6-(tert-butyl)pyridine (586 mg, 2.690 mmol, 1.20 eq) in (12 mL) of dioxane was added potassium carbonate (619 mg, 4.484 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min followed by addition of copper iodide (85 mg, 0.448 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (80 mg, 0.896 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 90° C. for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 ML), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-30% EtOAc in hexane] to afford the desired product, 1-(6-(tert-butyl)pyridin-2-yl)-2-cyclopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (300 mg, 37.51%) as light yellow viscous.
  • LCMS: 356.2 [M+1]+
  • Step-2: Synthesis of tert-butyl 7-((1-(6-(tert-butyl)pyridin-2-yl)-2-cyclopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(6-(tert-butyl)pyridin-2-yl)-2-cyclopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (140 mg, 0.393 mmol, 1.0 eq) in (2.0 mL) of toluene was added m-CPBA (135 mg, 0.786 mmol, 2.0 eq) and allowed to stir at rt for 30 minutes. tert-butyl 7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (195 mg, 0.786 mmol, 1.2 eq) and DIPEA (0.27 mL, 1.572 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 7-((1-(6-(tert-butyl)pyridin-2-yl)-2-cyclopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (81 mg, 37.01%) as white solid.
  • LCMS: 556.3 [M+1]+
  • Step-3: Synthesis of 1-(6-(tert-butyl)pyridin-2-yl)-2-cyclopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride: tert-butyl 7-((1-(6-(tert-butyl)pyridin-2-yl)-2-cyclopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (81 mg, 0.145 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (2 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(6-(tert-butyl)pyridin-2-yl)-2-cyclopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (58 mg, 75.29%) as light yellow solid.
  • LCMS: 456.3 [M+1]+; UPLC @ 254 nm=96.46% and @ 220 nm=97.00%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.25 (brs, 1H), 9.27 (brs, 3H), 8.81 (s, 1H), 8.04 (t, J=7.89 Hz, 1H), 7.67-7.79 (m, 3H), 7.51 (d, J=10.09 Hz, 1H), 7.43 (d, J=7.89 Hz, 1H), 7.17 (d, J=8.77 Hz, 1H), 4.25 (brs, 2H), 3.36 (brs, 2H), 3.23 (brs, 1H), 2.95 (t, J=5.70 Hz, 3H), 1.33 (s, 9H), 0.82 (brs, 4H).
  • Example S51. Synthesis of 1-(6-cyclopropylpyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02413
  • Step-1: Synthesis of 2-bromo-6-cyclopropylpyridine: To a stirred solution of 2,6-dibromopyridine (1.0 g, 4.221 mmol, 1.0 eq) in (30.0 mL) of dioxane was added Cs2CO3 (8.25 g, 25.329 mmol, 3.0 eq) at rt. The resulting mixture was purged with nitrogen for 10 min, followed by addition of Pd(PPh3)4 (487 mg, 0.422 mmol, 0.05 eq) and again purged with nitrogen for 10 min. The reaction mixture was stirred at 100° C. for 2 h. The progress of reaction was monitored by LCMS. The reaction mixture was diluted with water (50 mL), extracted with EtOAc (2×50 mL), the combined organic layers were washed with water (50 mL), dried over Na2SO4, concentrated and purified by combi flash chromatography [silica gel 100-200 mesh, elution 0-2% EtOAc in hexane] to afford the desired compound, 2-bromo-6-cyclopropylpyridine (500 mg, 29.92%) as colorless liquid.
  • LCMS: 198.9 [M+1]+
  • Step-2: Synthesis of 1-(6-cyclopropylpyridin-2-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (400 mg, 1.783 mmol, 1.0 eq) and 2-bromo-6-cyclopropylpyridine (423 mg, 2.140 mmol, 1.20 eq) in (20 mL) of dioxane was added potassium carbonate (493 mg, 3.566 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min followed by addition of copper iodide (68 mg, 0.356 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (63 mg, 0.713 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 90° C. for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-30% EtOAc in hexane] to afford the desired compound 1-(6-cyclopropylpyridin-2-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (500 mg, 82.10%) as light yellow viscous.
  • LCMS: 342.2 [M+1]+
  • Step-3: Synthesis of tert-butyl 7-((1-(6-cyclopropylpyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(6-cyclopropylpyridin-2-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (200 mg, 0.586 mmol, 1.0 eq) in (2.0 mL) of toluene was added m-CPBA (289 mg, 1.172 mmol, 2.0 eq) and allowed to stir at rt for 30 minutes. tert-butyl 7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (175 mg, 0.702 mmol, 1.2 eq) and DIPEA (0.40 mL, 2.344 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound tert-butyl 7-((1-(6-cyclopropylpyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (170 mg, 53.63%) as white solid.
  • LCMS: 542.5 [M+1]+
  • Step-4: Synthesis of 1-(6-cyclopropylpyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride: tert-butyl 7-((1-(6-cyclopropylpyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (160 mg, 0.295 mmol, 1.0 eq) was dissolved in 4.0 M-HCl (2 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound 1-(6-cyclopropylpyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (140 mg, 92.71%) as yellow solid.
  • LCMS: 442.3 [M+1]+; UPLC @ 254 nm=94.55% and @ 220 nm=97.17%.
  • 1H NMR (400 MHz, DMSO-d6) δ 10.30 (brs, 1H), 9.44 (brs, 2H), 8.81 (s, 1H), 7.95 (t, J=7.67 Hz, 1H), 7.68 (brs, 1H), 7.63 (d, J=7.89 Hz, 1H), 7.49 (d, J=6.58 Hz, 1H), 7.35 (d, J=7.45 Hz, 1H), 7.15 (d, J=8.33 Hz, 1H), 4.22 (brs, 2H), 4.10 (td, J=6.63, 13.48 Hz, 1H), 3.35 (brs, 2H), 2.95 (t, J=5.70 Hz, 2H), 2.17 (dt, J=4.17, 8.44 Hz, 1H), 1.34 (d, J=7.02 Hz, 6H), 0.95-1.03 (m, 2H), 0.87-0.95 (m, 2H).
  • Example S52. Synthesis of 1-(6-cyclopropylpyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02414
  • Step-1: Synthesis of tert-butyl 6-((1-(6-cyclopropylpyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(6-cyclopropylpyridin-2-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (200 mg, 0.586 mmol, 1.0 eq) in (2.0 mL) of toluene was added m-CPBA (289 mg, 1.172 mmol, 2.0 eq) and allowed to stir at rt for 30 minutes. tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (175 mg, 0.702 mmol, 1.2 eq) and DIPEA (0.40 mL, 2.344 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound tert-butyl 6-((1-(6-cyclopropylpyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (150 mg, 47.31%) as white solid.
  • LCMS: 542.5 [M+1]+
  • Step-2: Synthesis of 1-(6-cyclopropylpyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride: tert-butyl 6-((1-(6-cyclopropylpyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (150 mg, 0.276 mmol, 1.0 eq) was dissolved in 4.0 M-HCl (2 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound 1-(6-cyclopropylpyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (110 mg, 77.46%) as yellow solid.
  • LCMS: 442.3 [M+1]+; UPLC @ 254 nm=95.97% and @ 220 nm=94.33%.
  • 1H NMR (400 MHz, DMSO-d6) δ 10.28 (brs, 1H), 9.33 (brs, 2H), 8.82 (s, 1H), 7.93 (t, J=7.67 Hz, 1H), 7.72 (brs, 1H), 7.61 (d, J=7.89 Hz, 1H), 7.48 (d, J=7.45 Hz, 1H), 7.35 (d, J=7.45 Hz, 1H), 7.14 (d, J=8.33 Hz, 1H), 4.20 (brs, 3H), 3.37 (brs, 3H), 2.93-3.02 (m, 2H), 2.12-2.22 (m, 1H), 1.34 (d, J=6.58 Hz, 6H), 0.99 (d, J=7.89 Hz, 2H), 0.92 (brs, 2H).
  • Example S53. Synthesis of 1-(6-tert-butylpyridin-2-yl)-6-(1,2,3,4-tetrahydroisoquinolin-7-ylamino)-2-(1,1,1-trifluoropropan-2-yl)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one hydrochloride
  • Figure US20230109104A1-20230406-C02415
    Figure US20230109104A1-20230406-C02416
  • Step-1: Synthesis of ethyl 4-hydrazinyl-2-(methylthio)pyrimidine-5-carboxylate: To a stirred solution of hydrazine hydrate (3.5 mL, 107.44 mmol, 5 eq) in EtOH (50 mL) was added the solution of ethyl 4-chloro-2-(methylthio)pyrimidine-5-carboxylate (5 g, 21.488 mmol, 1 eq) in EtOH (100 mL) at 0° C. The resulting mixture was stirred at RT for 1 h. The progress of reaction was monitored by TLC. The reaction mixture was diluted with water (100 mL), extracted with EtOAc (2×200 mL). The combined organic layers were washed with water (100 mL), with brine (100 mL), dried over Na2SO4, concentrated under reduced pressure to afford ethyl 4-hydrazinyl-2-(methylthio)pyrimidine-5-carboxylate (4.21 g, 85.71%) as an off white solid.
  • LCMS: 229 [M+1]+
  • Step-2: Synthesis of (E)-ethyl 2-(methylthio)-4-(2-(1,1,1-trifluoropropan-2-ylidene)hydrazinyl)pyrimidine-5-carboxylate: To a stirred solution of ethyl 4-hydrazinyl-2-(methylthio)pyrimidine-5-carboxylate (4.2 g, 21.01 mmol, 1 eq) in EtOH (10 mL) was added 1,1,1-trifluoropropan-2-one (2.35 g, 21.02 mmol, 1 eq) at RT. The resulting mixture was heated at 50° C. for 2 h. The progress of reaction was monitored by TLC. The reaction mixture was diluted with water (100 mL), extracted with EtOAc (2×200 mL). The combined organic layers were washed with water (100 mL), with brine (100 mL), dried over Na2SO4, concentrated under reduced pressure to afford (E)-ethyl 2-(methylthio)-4-(2-(1,1,1-trifluoropropan-2-ylidene)hydrazinyl)pyrimidine-5-carboxylate (3.5 g, 59.32%) as an off-white solid.
  • LCMS: 323 [M+1]+
  • Step-3: Synthesis of ethyl 2-(methylthio)-4-(2-(1,1,1-trifluoropropan-2-yl)hydrazinyl) pyrimidine-5-carboxylate: To a stirred solution of (E)-ethyl 2-(methylthio)-4-(2-(1,1,1-trifluoropropan-2-ylidene)hydrazinyl)pyrimidine-5-carboxylate (2.2 g, 6.825 mmol, 1 eq) in THF (50 mL) was portion wise added LiBH4 (0.178 g, 8.19 mmol, 1.2 eq) at −30° C. The resulting mixture was warmed at 0° C. for 20 min. The progress of reaction was monitored by TLC. The reaction mixture was diluted with water (100 ML), extracted with EtOAc (2×200 ML). The combined organic layers were washed with water (100 mL), with brine (100 mL), dried over Na2SO4, concentrated under reduced pressure to afford crude which was purified using combi flash chromatography (Teledyne Isco Rf+); compound eluting 30% EtOAc/Hexane to afford ethyl 2-(methylthio)-4-(2-(1,1,1-trifluoropropan-2-yl)hydrazinyl) pyrimidine-5-carboxylate (0.28 g, 12.27%) as an off-white solid.
  • LCMS: 325 [M+1]+
  • Step-4: Synthesis of 6-(methylthio)-2-(1,1,1-trifluoropropan-2-yl)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one: To a stirred solution of ethyl 2-(methylthio)-4-(2-(1,1,1-trifluoropropan-2-yl)hydrazinyl) pyrimidine-5-carboxylate (0.28 g, 0.864 mmol, 1 eq) in EtOH (5 mL) was portion wise added NaOEt (0.235 g, 3.456 mmol, 4 eq) at RT. The resulting mixture was heated at 50° C. for 16 h. The progress of reaction was monitored by TLC. The reaction mixture was diluted with water (50 mL), extracted with EtOAc (2×75 mL). The combined organic layers were washed with water (50 mL), with brine (50 mL), dried over Na2SO4, concentrated under reduced pressure to afford crude which was purified using combi flash chromatography (Teledyne Isco Rf+); compound eluting 20% MeOH/DCM to afford 6-(methylthio)-2-(1,1,1-trifluoropropan-2-yl)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one (0.125 g, 52.08%) as an off-white solid.
  • LCMS: 279 [M+1]+
  • Step-5: Synthesis of 1-(6-tert-butylpyridin-2-yl)-6-(methylthio)-2-(1,1,1-trifluoropropan-2-yl)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one: To a stirred solution of 6-(methylthio)-2-(1,1,1-trifluoropropan-2-yl)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one (100 mg, 0.359 mmol, 1.0 eq) and 2-bromo-6-tert-butylpyridine (92.33 mg, 0.431 mmol, 1.2 eq) in 1,4 dioxane (5 mL) was added potassium carbonate (99.2 mg, 0.718 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 30 min followed by addition of copper iodide (13.67 mg, 0.0718 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (12.65 mg, 0.143 mmol, 0.4 eq) and again purged with nitrogen for 10 min. The resultant mixture was heated at 100° C. for 16 h. The reaction was monitored by TLC. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford crude product which was purified by flash chromatography (Teledyne Isco Rf+); to afford 1-(6-tert-butylpyridin-2-yl)-6-(methylthio)-2-(1,1,1-trifluoropropan-2-yl)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one (100 mg, 68.02%) as off white solid.
  • LCMS: 412 [M+1]+
  • Step-6: Synthesis of tert-butyl 7-(1-(6-tert-butylpyridin-2-yl)-3-oxo-2-(1,1,1-trifluoropropan-2-yl)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of -(6-tert-butylpyridin-2-yl)-6-(methylthio)-2-(1,1,1-trifluoropropan-2-yl)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one (90 mg, 0.218 mmol, 1.0 eq) in toluene (2 mL) was added m-CPBA (150.4 mg, 0.872 mmol, 4.0 eq) and allowed to stir at RT for 1 h. tert-butyl 7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (59.75 mg, 0.24 mmol, 1.1 eq) and DIPEA (168.8 mg, 1.308 mmol, 6.0 eq) were then added and the mixture was allowed to stir at RT for 1 h. The reaction was monitored by TLC. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford crude which was purified by reversed-phase chromatography to afford tert-butyl 7-(1-(6-tert-butylpyridin-2-yl)-3-oxo-2-(1,1,1-trifluoropropan-2-yl)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (30 mg, 22.55%) as off white solid.
  • LCMS: 612 [M+1]+
  • Step-7: Synthesis of 1-(6-tert-butylpyridin-2-yl)-6-(1,2,3,4-tetrahydroisoquinolin-7-ylamino)-2-(1,1,1-trifluoropropan-2-yl)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one hydrochloride: The solution of tert-butyl 7-(1-(6-tert-butylpyridin-2-yl)-3-oxo-2-(1,1,1-trifluoropropan-2-yl)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (30 mg, 0.049 mmol, 1.0 eq) in 2 M HCl in methanol (10 mL) was stirred at RT for 72 h. The reaction was monitored by TLC. After completion of reaction, the volatiles are removed under reduced pressure to afford 1-(6-tert-butylpyridin-2-yl)-6-(1,2,3,4-tetrahydroisoquinolin-7-ylamino)-2-(1,1,1-trifluoropropan-2-yl)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one as a hydrochloride salt (11 mg, 42.38%) as off white solid.
  • LCMS: 512 [M+1]+
  • 1H NMR: (400 MHz, MeOD) δ 8.86 (s, 1H), 7.99 (s, 1H), 7.59-7.68 (m, 2H), 7.46-7.55 (m, 2H), 7.19 (d, J=8.33 Hz, 1H), 5.03 (br. s., 1H), 4.31 (br. s., 2H), 3.51 (t, J=6.36 Hz, 2H), 3.09 (t, J=6.36 Hz, 2H), 1.71 (d, J=7.02 Hz, 3H), 1.36 (s, 9H).
  • Example S54. Synthesis of 2-tert-butyl-1-(6-tert-butylpyridin-2-yl)-6-(1,2,3,4-tetrahydroisoquinolin-7-ylamino)-JH-pyrazolo[3,4-d]pyrimidin-3(2H)-one
  • Figure US20230109104A1-20230406-C02417
    Figure US20230109104A1-20230406-C02418
  • Step-1: Synthesis of ethyl 4-(2-tert-butylhydrazinyl)-2-(methylthio)pyrimidine-5-carboxylate: To a stirred solution of ethyl 4-chloro-2-(methylthio)pyrimidine-5-carboxylate (1.66 g, 10.3 mmol, 1.2 eq) in EtOH (50 mL) was added DIPEA (4.48 g, 34.3 mmol, 4 eq) at RT. The resulting reaction mixture was stirred at RT for 30 min, followed by addition of 1 (2 g, 8.621 mmol, 1 eq) at RT. The resulting reaction mixture was stirred at RT for 3 h. The progress of reaction was monitored by TLC. Upon completion, the reaction mixture was diluted with water (100 mL), extracted with EtOAc (2×200 mL). The combined organic layers were washed with water (100 mL), with brine (100 mL), dried over Na2SO4, concentrated under reduced pressure to afford ethyl 4-(2-tert-butylhydrazinyl)-2-(methylthio)pyrimidine-5-carboxylate (2.19 g, 89.38%) as off white solid.
  • LCMS: 285 [M+1]+
  • Step-2: Synthesis of 2-tert-butyl-6-(methylthio)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one: To a stirred solution of ethyl 4-(2-tert-butylhydrazinyl)-2-(methylthio)pyrimidine-5-carboxylate (1.2 g, 4.21 mmol, 1 eq) in MeOH (10 mL) was added NaOMe (0.455 g, 1.68 mmol, 4 eq) at RT. The resulting mixture was irradiated in microwave at 60° C. for 40 min. The progress of reaction was monitored by TLC. Upon completion, the reaction mixture was diluted with water (100 mL), extracted with EtOAc (2×200 mL). The combined organic layers were washed with water (100 mL), with brine (100 mL), dried over Na2SO4, concentrated under reduced pressure to afford crude which was purified using combi flash chromatography (Teledyne Isco Rf+); compound eluting 90% EtOAc/Hexane to afford 2-tert-butyl-6-(methylthio)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one (0.12 g, 12%) as an off-white solid.
  • LCMS: 239 [M+1]+
  • Step-3: Synthesis of 2-tert-butyl-1-(6-tert-butylpyridin-2-yl)-6-(methylthio)-1H-pyrazolo [3,4-d]pyrimidin-3(2H)-one: To a stirred solution of 2-tert-butyl-6-(methylthio)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one (180 mg, 0.756 mmol, 1.0 eq) and 2-bromo-6-tert-butylpyridine (194.3 mg, 0.907 mmol, 1.2 eq) in 1,4 dioxane (5 mL) was added potassium carbonate (208.9 mg, 1.512 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 30 min followed by addition of copper iodide (28.7 mg, 0.151 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (26.05 mg, 0.302 mmol, 0.4 eq) and again purged with nitrogen for 10 min. The resultant mixture was heated at 100° C. for 5 h. The reaction was monitored by TLC. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford crude which was purified by flash chromatography (Teledyne Isco Rf+); compound eluting 30% EtOAc/Hexane to afford 2-tert-butyl-1-(6-tert-butylpyridin-2-yl)-6-(methylthio)-1H-pyrazolo [3,4-d]pyrimidin-3(2H)-one (100 mg, 35.71%) as off white solid.
  • LCMS: 372 [M+H]+
  • Step-4: Synthesis of tert-butyl 7-(2-tert-butyl-1-(6-tert-butylpyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 2-tert-butyl-1-(6-tert-butylpyridin-2-yl)-6-(methylthio)-1H-pyrazolo [3,4-d]pyrimidin-3(2H)-one (100 mg, 0.269 mmol, 1.0 eq) in toluene (2 mL) was added m-CPBA (185.6 mg, 1.076 mmol, 4.0 eq) and allowed to stir at RT for 1 h, followed by addition of 7 (84.1 mg, 0.296 mmol, 1.1 eq) and DIPEA (208.3 mg, 1.614 mmol, 6.0 eq) were added and allowed to stir at 55° C. for 6 h. The reaction was monitored by TLC. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford crude which was purified by flash chromatography (Teledyne Isco Rf+); compound eluting 30% EtOAc/Hexane to afford tert-butyl 7-(2-tert-butyl-1-(6-tert-butylpyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (95 mg, 54.28%) as off white solid.
  • LCMS: 572 [M+1]+
  • Step-5: Synthesis of 2-tert-butyl-1-(6-tert-butylpyridin-2-yl)-6-(1,2,3,4-tetrahydro isoquinolin-7-ylamino)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one: The solution of tert-butyl 7-(2-tert-butyl-1-(6-tert-butylpyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (95 mg, 0.166 mmol, 1.0 eq) in 2 M HCl in methanol (10 mL) was stirred at RT for 72 h. The reaction was monitored by TLC. After completion of reaction, the volatiles are removed under reduced pressure and the residue was taken in saturated NaHCO3 and extracted with EtOAc (150 mL×2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford crude which was purified by reversed-phase chromatography to afford 2-tert-butyl-1-(6-tert-butylpyridin-2-yl)-6-(1,2,3,4-tetrahydro isoquinolin-7-ylamino)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one (13 mg, 16.66%) as off white solid.
  • LCMS: 472 [M+1]+
  • 1H NMR: (400 MHz, MeOD) δ 8.77 (s, 1H), 7.94 (s, 1H), 7.59 (br. s., 1H), 7.52 (d, J=7.89 Hz, 2H), 7.44 (d, J=7.89 Hz, 1H), 7.17 (d, J=8.33 Hz, 1H), 4.33 (s, 2H), 3.51 (t, J=6.58 Hz, 2H), 3.08 (t, J=6.14 Hz, 2H), 1.47 (s, 9H), 1.35 (s, 9H).
  • Example S55. Synthesis of 1-(6-tert-butylpyridin-2-yl)-6-(1,2,3,4-tetrahydroisoquinolin-7-ylamino)-2-(2,2,2-trifluoroethyl)-JH-pyrazolo[3,4-d]pyrimidin-3(2H)-one (Compound No. 1.152)
  • Figure US20230109104A1-20230406-C02419
  • Step-1: Synthesis of ethyl 4-hydrazinyl-2-(methylthio)pyrimidine-5-carboxylate: To a stirred solution of hydrazine hydrate (3.5 mL, 107.44 mmol, 5 eq) in EtOH (50 mL) was added the solution of ethyl 4-chloro-2-(methylthio)pyrimidine-5-carboxylate (5 g, 21.488 mmol, 1 eq) in EtOH (100 mL) at 0° C. The resulting mixture was stirred at RT for 1 h. The progress of reaction was monitored by TLC. The reaction mixture was diluted with water (100 mL), extracted with EtOAc (2×200 mL). The combined organic layers were washed with water (100 mL), with brine (100 mL), dried over Na2SO4, concentrated under reduced pressure to afford ethyl 4-hydrazinyl-2-(methylthio)pyrimidine-5-carboxylate (4.21 g, 85.71%) as an off white solid.
  • LCMS: 229 [M+1]+
  • Step-2: Synthesis of (E)-ethyl 2-(methylthio)-4-(2-(2,2,2-trifluoroethylidene) hydrazinyl) pyrimidine-5-carboxylate: To a stirred solution of (E)-ethyl 2-(methylthio)-4-(2-(2,2,2-trifluoroethylidene) hydrazinyl) pyrimidine-5-carboxylate (4 g, 17.541 mmol, 1 eq) in IPA (20 mL) was added 2,2,2-trifluoroacetaldehyde hydrate (6.1 g, 52.523 mmol, 3 eq) at RT. The resulting mixture was heated at 80° C. for 72 h. The progress of reaction was monitored by TLC. Upon completion, the resulting reaction mixture was quenched in to water to follow out solid, filtered through Buchner funnel washed with water and hexane to afford (E)-ethyl 2-(methylthio)-4-(2-(2,2,2-trifluoroethylidene) hydrazinyl) pyrimidine-5-carboxylate (2.59 g, 48.05%) as an off-white solid.
  • LCMS: 309 [M+1]+
  • Step-3: Synthesis of ethyl 2-(methylthio)-4-(2-(2,2,2-trifluoroethyl)hydrazinyl)pyrimidine-5-carboxylate: To a stirred solution of (E)-ethyl 2-(methylthio)-4-(2-(2,2,2-trifluoroethylidene) hydrazinyl) pyrimidine-5-carboxylate (2 g, 6.425 mmol, 1 eq) in THF (50 mL) was added LiBH4 (0.169 g, 7.79 mmol, 1.2 eq) at 0° C. portion wise. The resulting mixture was stirred at RT for 1 h. The progress of reaction was monitored by TLC. Upon completion, the reaction mixture was diluted with water (100 mL), extracted with EtOAc (2×200 mL). The combined organic layers were washed with water (100 mL), with brine (100 mL), dried over Na2SO4, concentrated under reduced pressure to afford crude which was purified using combi flash chromatography (Teledyne Isco Rf+); compound eluting 30% EtOAc/Hexane to afford ethyl 2-(methylthio)-4-(2-(2,2,2-trifluoroethyl)hydrazinyl)pyrimidine-5-carboxylate (0.6 g, 30%) as an off-white solid.
  • LCMS: 311 [M+1]+
  • Step-4: Synthesis of 6-(methylthio)-2-(2,2,2-trifluoroethyl)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one: To a stirred solution of ethyl 2-(methylthio)-4-(2-(2,2,2-trifluoroethyl)hydrazinyl)pyrimidine-5-carboxylate (0.4 g, 1.29 mmol, 1 eq) in EtOH (7 mL) was added 6N—NaOH (2 mL) at 0° C. The resulting mixture was stirred at 0° C. for 10 min. The progress of reaction was monitored by TLC. Upon completion, the reaction mixture was quenched in 1N HCl solution, extracted with EtOAc (2×200 mL). The combined organic layers were washed with water (100 mL), with brine (100 mL), dried over Na2SO4, concentrated under reduced pressure to afford crude which was purified using combi flash chromatography (Teledyne Isco Rf+); compound eluting 5% MeOH/DCM to afford pure 6-(methylthio)-2-(2,2,2-trifluoroethyl)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one (0.19 g, 55.88%) as an off-white solid.
  • LCMS: 265 [M+1]+
  • Step-5: Synthesis of 1-(6-tert-butylpyridin-2-yl)-6-(methylthio)-2-(2,2,2-trifluoroethyl)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one: To a stirred solution of 6-(methylthio)-2-(2,2,2-trifluoroethyl)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one (90 mg, 0.719 mmol, 1.0 eq) and 2-bromo-6-tert-butylpyridine (184.7 mg, 0.862 mmol, 1.2 eq) in 1,4 dioxane (3 mL) was added potassium carbonate (198.7 mg, 1.438 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 30 min followed by addition of copper iodide (27.38 mg, 0.143 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (25.35 mg, 0.287 mmol, 0.4 eq) and again purged with nitrogen for 10 min. The resulting mixture was heated at 100° C. for 5 h. The reaction was monitored by TLC. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (150 mL×2). The combined organic layers were washed with water (50 mL) brine solution (50 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford crude which was purified by flash chromatography (Teledyne Isco Rf+); compound eluting 30% EtOAc/Hexane to afford pure 1-(6-tert-butylpyridin-2-yl)-6-(methylthio)-2-(2,2,2-trifluoroethyl)-1H-pyrazolo [3,4-d]pyrimidin-3(2H)-one (180 mg, 63.15%) as off white solid.
  • LCMS: 398 [M+1]+
  • Step-6: Synthesis of tert-butyl 7-(1-(6-tert-butylpyridin-2-yl)-3-oxo-2-(2,2,2-trifluoroethyl)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(6-tert-butylpyridin-2-yl)-6-(methylthio)-2-(2,2,2-trifluoroethyl)-1H-pyrazolo [3,4-d]pyrimidin-3(2H)-one (150 mg, 0.377 mmol, 1.0 eq) in toluene (3 mL) was added m-CPBA (260.2 mg, 1.508 mmol, 4.0 eq) and allowed to stir at RT for 1 h, followed by addition of 9 (118.04 mg, 0.415 mmol, 1.1 eq) and DIPEA (292.02 mg, 2.262 mmol, 6.0 eq) were added and allowed to stir at 55° C. for 3 h. The reaction was monitored by TLC. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (100 mL×2). The combined organic layers were washed with water (50 mL) brine solution (50 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford crude tert-butyl 7-(1-(6-tert-butylpyridin-2-yl)-3-oxo-2-(2,2,2-trifluoroethyl)-2,3-dihydro-1H-pyrazolo[3,4-d] pyrimidin-6-ylamino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (200 mg, 88.88%) as off white solid.
  • LCMS: 598 [M+1]+
  • Step-7: Synthesis of 1-(6-tert-butylpyridin-2-yl)-6-(1,2,3,4-tetrahydroisoquinolin-7-ylamino)-2-(2,2,2-trifluoroethyl)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one: The solution of tert-butyl 7-(1-(6-tert-butylpyridin-2-yl)-3-oxo-2-(2,2,2-trifluoroethyl)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (200 mg, 0.335 mmol, 1.0 eq) in 2 M HCl in methanol (20 mL) was stirred at RT for 72 h. The reaction was monitored by TLC. After completion of reaction, the volatiles are removed under reduced pressure and the residue was taken in saturated NaHCO3 and extracted with EtOAc (200 mL×2). Combined organic layer was washed with water (150 mL) brine solution (100 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford crude which was purified by reversed-phase chromatography to afford 1-(6-tert-butylpyridin-2-yl)-6-(1,2,3,4-tetrahydroisoquinolin-7-ylamino)-2-(2,2,2-trifluoroethyl)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one (30 mg, 12.5%) as off white solid.
  • LCMS: 498 [M+1]+
  • 1H NMR: (400 MHz, MeOD) δ 8.90 (s, 1H), 7.99 (s, 1H), 7.85 (d, J=8.33 Hz, 1H), 7.69 (s, 1H), 7.54 (d, J=8.33 Hz, 1H), 7.45 (d, J=7.89 Hz, 1H), 7.22 (d, J=8.33 Hz, 1H), 5.02-5.13 (m, 2H), 4.31 (br. s., 2H), 3.47 (br. s., 2H), 3.07 (br. s., 2H), 1.39 (s, 9H).
  • Example S56 Synthesis of 1-(6-tert-butylpyridin-2-yl)-2-(1-hydroxypropan-2-yl)-6-(1,2,3,4-tetrahydro isoquinolin-7-ylamino)-JH-pyrazolo[3,4-d]pyrimidin-3(2H)-one
  • Figure US20230109104A1-20230406-C02420
    Figure US20230109104A1-20230406-C02421
  • Step-1: Synthesis of ethyl 4-hydrazinyl-2-(methylthio)pyrimidine-5-carboxylate: To a stirred solution of hydrazine hydrate (3.5 mL, 107.44 mmol, 5 eq) in EtOH (50 mL) was added the solution of ethyl 4-chloro-2-(methylthio)pyrimidine-5-carboxylate (5 g, 21.488 mmol, 1 eq) in EtOH (100 mL) at 0° C. The resulting mixture was stirred at RT for 1 h. The progress of reaction was monitored by TLC. Upon completion, the reaction mixture was diluted with water (100 mL), extracted with EtOAc (2×200 mL). The combined organic layers were washed with water (100 mL), with brine (100 mL), dried over Na2SO4, concentrated under reduced pressure to afford ethyl 4-hydrazinyl-2-(methylthio)pyrimidine-5-carboxylate (4.21 g, 85.71%) as off white solid.
  • LCMS: 229 [M+1]+
  • Step-2: Synthesis of (E)-ethyl 4-(2-(1-hydroxypropan-2-ylidene)hydrazinyl)-2-(methylthio) pyrimidine-5-carboxylate: To a stirred solution of ethyl 4-hydrazinyl-2-(methylthio)pyrimidine-5-carboxylate (4.7 g, 26.6 mmol, 1 eq) in EtOH (70 mL) was added 1-hydroxypropan-2-one (7.63 g, 103.6 mmol, 5 eq) at RT. The resulting mixture was heated at 60° C. for 16 h. The progress of reaction was monitored by TLC. Upon completion, the reaction mixture was diluted with water (100 mL), extracted with EtOAc (2×200 mL). The combined organic layers were washed with water (100 mL), with brine (100 mL), dried over Na2SO4, concentrated under reduced pressure to afford crude which was purified using combi flash chromatography (Teledyne Isco Rf+); compound eluting 30% EtOAc/Hexane to afford (E)-ethyl 4-(2-(1-hydroxypropan-2-ylidene)hydrazinyl)-2-(methylthio) pyrimidine-5-carboxylate (4 g, 68.96%) as an off-white solid.
  • LCMS: 284 [M+1]+
  • Step-3: Synthesis of ethyl 4-(2-(1-hydroxypropan-2-yl)hydrazinyl)-2-(methylthio) pyrimidine-5-carboxylate: To a stirred solution of (E)-ethyl 4-(2-(1-hydroxypropan-2-ylidene)hydrazinyl)-2-(methylthio) pyrimidine-5-carboxylate (2 g, 7.425 mmol, 1 eq) in THF (50 mL) was portion wise added LiBH4 (0.199 g, 9.1 mmol, 1.3 eq) at 0° C. The resulting mixture was stirred at 0° C. for 1 h. The progress of reaction was monitored by TLC. Upon completion, the reaction mixture was diluted with water (100 mL), extracted with EtOAc (2×200 mL). The combined organic layers were washed with water (100 mL), with brine (100 mL), dried over Na2SO4, concentrated under reduced pressure to afford crude which was purified using combi flash chromatography (Teledyne Isco Rf+); compound eluting 95% EtOAc/Hexane to afford ethyl 4-(2-(1-hydroxypropan-2-yl)hydrazinyl)-2-(methylthio) pyrimidine-5-carboxylate (0.5 g, 24.87%) as an off-white solid.
  • LCMS: 287 [M+1]+
  • Step-4: Synthesis of 2-(1-hydroxypropan-2-yl)-6-(methylthio)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one: To a stirred solution of ethyl 4-(2-(1-hydroxypropan-2-yl)hydrazinyl)-2-(methylthio) pyrimidine-5-carboxylate (0.45 g, 0.0015 mmol, 1 eq) in EtOH (7 mL) was added 6N NaOH (2.2 mL) at 0° C. The resulting mixture was stirred at 0° C. for 10 min. The progress of reaction was monitored by TLC. Upon completion, the reaction mixture was quenched in 1N HCl solution, extracted with EtOAc (2×200 mL). The combined organic layers were washed with water (100 mL), with brine (100 mL), dried over Na2SO4, concentrated under reduced pressure to afford 2-(1-hydroxypropan-2-yl)-6-(methylthio)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one (0.225 g, 59.36%) as an off-white solid.
  • LCMS: 241 [M+1]+
  • Step-5: Synthesis of 1-(6-tert-butylpyridin-2-yl)-2-(1-hydroxypropan-2-yl)-6-(methylthio)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one: To a stirred solution of 2-(1-hydroxypropan-2-yl)-6-(methylthio)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one (200 mg, 0.833 mmol, 1.0 eq) and 2-bromo-6-tert-butylpyridine (214 mg, 0.999 mmol, 1.2 eq) in 1,4 dioxane (3 mL) was added potassium carbonate (230 mg, 1.666 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 30 min followed by addition of copper iodide (31 mg, 0.666 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (30 mg, 0.333 mmol, 0.4 eq) and again purged with nitrogen for 10 min. The resultant mixture was heated at 100° C. for 5 h. The reaction was monitored by TLC. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (150 mL×2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford crude which was purified by flash chromatography (Teledyne Isco Rf+); compound eluting 95% EtOAc/Hexane to afford 1-(6-tert-butylpyridin-2-yl)-2-(1-hydroxypropan-2-yl)-6-(methylthio)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one (100 mg, 32.25%) as off white solid.
  • LCMS: 374 [M+1]+
  • Step-6: Synthesis of tert-butyl 7-(1-(6-tert-butylpyridin-2-yl)-2-(1-hydroxypropan-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(6-tert-butylpyridin-2-yl)-2-(1-hydroxypropan-2-yl)-6-(methylthio)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one (100 mg, 0.268 mmol, 1.0 eq) in toluene (3 mL) was added m-CPBA (184 mg, 1.072 mmol, 4.0 eq) and allowed to stir at RT for 1 h, followed by addition of tert-butyl 7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (83 mg, 0.294 mmol, 1.1 eq) and DIPEA (207 mg, 1.608 mmol, 6.0 eq) were added and allowed to stir at 80° C. for 4 h. The reaction was monitored by TLC. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (100 mL×2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford crude tert-butyl 7-(1-(6-tert-butylpyridin-2-yl)-2-(1-hydroxypropan-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (130 mg, 84.96%) as off white solid.
  • LCMS: 574 [M+1]+
  • Step-7: Synthesis of 1-(6-tert-butylpyridin-2-yl)-2-(1-hydroxypropan-2-yl)-6-(1,2,3,4-tetrahydro isoquinolin-7-ylamino)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one: The solution of tert-butyl 7-(1-(6-tert-butylpyridin-2-yl)-2-(1-hydroxypropan-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (200 mg, 0.349 mmol, 1.0 eq) in 2 M HCl in methanol (20 mL) was stirred at RT for 72 h. The reaction was monitored by TLC. After completion of reaction, the volatiles are removed under reduced pressure and the residue was taken in saturated NaHCO3 and extracted with EtOAc (200 mL×2). Combined organic layer was washed with water (150 mL) brine solution (100 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford crude which was purified by reversed-phase chromatography to afford 1-(6-tert-butylpyridin-2-yl)-2-(1-hydroxypropan-2-yl)-6-(1,2,3,4-tetrahydroisoquinolin-7-ylamino)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one (14 mg, 8.48%) as off white solid.
  • LCMS: 474 [M+1]+
  • 1H NMR: (400 MHz, MeOD) δ 8.82 (s, 1H), 8.52 (br. s., 1H), 7.98 (t, J=7.89 Hz, 1H), 7.71 (d, J=7.89 Hz, 1H), 7.64 (br. s., 1H), 7.46-7.54 (m, 2H), 7.17 (d, J=8.77 Hz, 1H), 4.29-4.35 (m, 1H), 4.26 (s, 2H), 4.02 (dd, J=7.67, 11.18 Hz, 1H), 3.83 (dd, J=5.70, 10.96 Hz, 1H), 3.41-3.49 (m, 2H), 3.04 (t, J=6.14 Hz, 2H), 1.47 (d, J=7.02 Hz, 3H), 1.38 (s, 9H)
  • Example S57. Synthesis of 1-(6-tert-butylpyridin-2-yl)-2-(2-methoxyethyl)-6-(1,2,3,4-tetrahydro isoquinolin-7-ylamino)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one
  • Figure US20230109104A1-20230406-C02422
    Figure US20230109104A1-20230406-C02423
  • Step-1: Synthesis of tert-butyl 1,3-dioxoisoindolin-2-yl(2-methoxyethyl)carbamate: To a stirred solution of tert-butyl 1,3-dioxoisoindolin-2-ylcarbamate (1 g, 3.8526 mmol, 1.0 eq) in ACN (100 mL) was added 2 (1.06 g, 7.625 mmol, 2 eq), BnEt3N4Cl (347 mg, 1.506 mmol, 0.4 eq) and K2CO3 (1.57 g, 11.258 mmol, 3.0 eq) at RT. The resulting mixture was heated at 55° C. for overnight. The progress of reaction was monitored by TLC. Upon completion, the reaction mixture was diluted with water (100 mL), extracted with EtOAc (2×200 mL). The combined organic layers were washed with water (50 mL), with brine (50 mL), dried over Na2SO4, concentrated and purified by crystallization to afford tert-butyl 1,3-dioxoisoindolin-2-yl(2-methoxyethyl)carbamate (1.1 g, 91.66%) as light yellow solid.
  • LCMS: 321 [M+1]+
  • Step-2: Synthesis of tert-butyl 1-(2-methoxyethyl)hydrazinecarboxylate: To a stirred solution of afford tert-butyl 1,3-dioxoisoindolin-2-yl(2-methoxyethyl)carbamate (1.4 g, 4.375 mmol, 1.0 eq) in EtOH (25 mL) was added NH2NH2.H2O (1.4 mL) at RT. The resulting mixture was stirred at 110° C. for 1 h, formation of white precipitates was observed. The progress of reaction was monitored by TLC. Upon completion, the reaction mixture was diluted with water (100 mL), extracted with EtOAc (2×200 mL). The combined organic layers were washed with water (50 mL), with brine (50 mL), dried over Na2SO4, concentrated and purified by crystallization to afford tert-butyl 1-(2-methoxyethyl)hydrazinecarboxylate (0.41 g, 49.41%) as light yellow solid.
  • LCMS: 191 [M+1]+
  • Step-3: Synthesis of ethyl 4-(2-(tert-butoxycarbonyl)-2-(2-methoxyethyl)hydrazinyl)-2-(methylthio)pyrimidine-5-carboxylate: To a stirred solution of ethyl 4-(2-(tert-butoxycarbonyl)-2-(2-methoxyethyl)hydrazinyl)-2-(methylthio)pyrimidine-5-carboxylate (0.402 g, 2.114 mmol, 1.2 eq) in EtOH (10 mL) was added DIPEA (0.909 g, 7.048 mmol, 4 eq) at RT. The reaction was stirred at RT for 30 min, followed by addition of 5 (0.41 g, 1.762 mmol, 1 eq) and stirred at 60° C. for 4 h. Upon completion, reaction was diluted with water (50 mL) and extracted with EtOAc (50 mL×2). Organic layer was washed with brine solution (50 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography (Teledyne Isco Rf+); compound eluting 30% EtOAc/Hexane to afford ethyl 4-(2-(tert-butoxycarbonyl)-2-(2-methoxyethyl)hydrazinyl)-2-(methylthio)pyrimidine-5-carboxylate (0.5 g, 73.31%).
  • LCMS: 387 [M+1]+
  • Step-4: Synthesis of ethyl 4-(2-(2-methoxyethyl)hydrazinyl)-2-(methylthio)pyrimidine-5-carboxylate: To a stirred solution of ethyl 4-(2-(tert-butoxycarbonyl)-2-(2-methoxyethyl)hydrazinyl)-2-(methylthio)pyrimidine-5-carboxylate (0.5 g, 1.295 mmol, 1.0 eq.) in 10 mL of DCM was added TFA (5 mL) dropwise at 0° C. and allowed to stir at RT for 3 h. Upon completion, solvent was removed under reduced pressure to afford ethyl 4-(2-(2-methoxyethyl)hydrazinyl)-2-(methylthio)pyrimidine-5-carboxylate (0.41 g, 82.99%) as off white solid.
  • LCMS: 383 [M+1]+
  • Step-5: Synthesis of 2-(2-methoxyethyl)-6-(methylthio)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one: To a stirred solution of ethyl 4-(2-(2-methoxyethyl)hydrazinyl)-2-(methylthio)pyrimidine-5-carboxylate (400 mg, 1.044 mmol, 1 eq) in EtOH (10 mL) was added 6N NaOH solution (2 mL) at 0° C. and allowed to stir at 0° C. for 10 min. Upon completion, reaction was acidified by using 1N HCl solution. Ethanol was removed under reduced pressure; residue obtained was cooled to RT and extracted with DCM (100 mL×3). Organic layer was washed with brine solution, dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford 2-(2-methoxyethyl)-6-(methylthio)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one (300 mg, 84.26%) as an off white solid.
  • LCMS: 341 [M+1]+
  • Step-6: Synthesis of 1-(6-tert-butylpyridin-2-yl)-2-(2-methoxyethyl)-6-(methylthio)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one: To a stirred solution of 2-(2-methoxyethyl)-6-(methylthio)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one (300 mg, 1.248 mmol, 1.0 eq) and 2-bromo-6-tert-butylpyridine (320 mg, 1.497 mmol, 1.2 eq) in dioxane (6 mL) was added potassium carbonate (344.7 mg, 2.496 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 30 min followed by addition of copper iodide (47.53 mg, 0.249 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (44 mg, 0.499 mmol, 0.4 eq) and again purged with nitrogen for 30 min. The resultant mixture was heated at 110° C. for 5 h. Upon completion, the reaction mixture was diluted with water and extracted with EtOAc (150 mL×2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography (Teledyne Isco Rf+); compound eluting 50% EtOAc/Hexane to afford pure to afford 1-(6-tert-butylpyridin-2-yl)-2-(2-methoxyethyl)-6-(methylthio)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one (200 mg, 42.91%).
  • LCMS: 373 [M+1]+
  • Step-7: Synthesis of tert-butyl 7-(1-(6-tert-butylpyridin-2-yl)-2-(2-methoxyethyl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 10 (200 mg, 0.535 mmol, 1.0 eq) in toluene (5 mL) was added m-CPBA (369 mg, 2.14 mmol, 4.0 eq) and allowed to stir at RT for 1 h, followed by addition of 11 (146.28 mg, 0.589 mmol, 1.1 eq) and DIPEA (414.4 mg, 3.21 mmol, 6.0 eq) at RT and allowed to stir at 80° C. for 16 h. Upon completion, the reaction mixture was diluted with water (50 mL) and extracted with EtOAc (150 mL×2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude tert-butyl 7-(1-(6-tert-butylpyridin-2-yl)-2-(2-methoxyethyl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (300 mg, 98.68%).
  • LCMS: 574 [M+1]+
  • Step-8: Synthesis of 1-(6-tert-butylpyridin-2-yl)-2-(2-methoxyethyl)-6-(1,2,3,4-tetrahydro isoquinolin-7-ylamino)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one: The solution of tert-butyl 7-(1-(6-tert-butylpyridin-2-yl)-2-(2-methoxyethyl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (300 mg, 0.523 mmol, 1.0 eq) in 2 M HCl in methanol (20 mL) was stirred at RT for 72 h. The reaction was monitored by TLC. Upon completion, the volatiles are removed under reduced pressure and the residue was taken in saturated NaHCO3 and extracted with EtOAc (200 mL×2). Combined organic layer was washed with water (150 mL) brine solution (100 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford crude which was purified by reversed-phase chromatography to afford 1-(6-tert-butylpyridin-2-yl)-2-(2-methoxyethyl)-6-(1,2,3,4-tetrahydro isoquinolin-7-ylamino)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one (34 mg, 13.76%) as off white solid.
  • LCMS: 474 [M+1]+
  • 1H NMR: (400 MHz, MeOD) δ 8.84 (s, 1H), 7.97 (d, J=7.89 Hz, 1H), 7.82 (d, J=7.89 Hz, 1H), 7.72 (br. s., 1H), 7.54 (br. s., 1H), 7.43 (d, J=7.89 Hz, 1H), 7.21 (d, J=8.33 Hz, 1H), 4.44 (t, J=5.48 Hz, 2H), 4.33 (s, 2H), 3.42-3.51 (m, 4H), 3.18 (s, 3H), 3.08 (t, J=6.14 Hz, 2H), 1.38 (s, 9H)
  • Example S58. Synthesis of 1-(3-(tert-butyl)phenyl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02424
  • Step-1: Synthesis of 1-(3-(tert-butyl)phenyl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-(cyclopropylmethyl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (0.400 g, 1.777 mmol, 1.0 eq) and 1-bromo-3-(tert-butyl)benzene (0.36 mL, 2.133 mmol, 1.2 eq) in (5 mL) of dioxane was added Potassium carbonate (0.491 g, 3.554 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min, followed by addition of copper iodide (67 mg, 0.355 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (62 mg, 0.710 mmol, 0.4 eq) and again purged with nitrogen for 10 minutes and stirred at 80° C. for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layers were washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure. The crude product obtained was purified by flash chromatography (Combiflash, Elution: 0-70% EtOAc in Hexane) to afford the desired compound, 1-(3-(tert-butyl)phenyl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (160 mg, 25.27%) as an off white solid.
  • LCMS: 357.2 [M+1]+
  • Step-2: Synthesis of tert-butyl 6-((1-(3-(tert-butyl)phenyl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(3-(tert-butyl)phenyl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (80 mg, 0.224 mmol, 1.0 eq) in (1 mL) of toluene was added m-CPBA (77 mg, 0.448 mmol, 2.0 eq) and allowed to stir at rt for 30 min. tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (66 mg, 0.268 mmol, 1.2 eq) and DIPEA (0.3 mL, 0.896 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. The formation of precipitates was observed which was filtered to afford the desired compound, tert-butyl 6-((1-(3-(tert-butyl)phenyl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (70 mg, 56.45%) as an off white solid.
  • LCMS: 557.3 [M+1]+
  • Step-3: Synthesis of 1-(3-(tert-butyl)phenyl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: tert-butyl 6-((1-(3-(tert-butyl)phenyl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (70 mg, 0.125 mmol, 1.0 eq) was dissolved in (0.9 mL) of dioxane and added 4M dioxane-HCl (0.9 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(3-(tert-butyl)phenyl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (35 mg, 39.32%) as an off white solid.
  • LCMS: 457.3 [M+1]+; UPLC @ 254 nm=90.86% and @ 220 nm=94.62%.
  • 1H NMR (400 MHz, DMSO-d6): δ10.21 (br. s., 1H), 9.22 (br. s., 2H), 8.82 (s, 1H), 7.63 (br. s., 1H), 7.52 (d, J=7.45 Hz, 2H), 7.48 (br. s., 2H), 7.39 (br. s., 1H), 7.08 (d, J=8.77 Hz, 1H), 4.18 (br. s., 2H), 4.06 (br. s., 1H), 3.35 (br. s., 3H), 2.93 (br. s., 2H), 1.20-1.38 (m, 15H)
  • Example S59. Synthesis of 1-(3-(tert-butyl)phenyl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02425
  • Step-1: Synthesis 1-(3-(tert-butyl)phenyl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-(cyclopropylmethyl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (0.400 g, 1.777 mmol, 1.0 eq) and 1-bromo-3-(tert-butyl)benzene (0.36 mL, 2.133 mmol, 1.2 eq) in (5 mL) of dioxane were added Potassium carbonate (0.491 g, 3.554 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min, followed by addition of copper iodide (67 mg, 0.355 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (62 mg, 0.710 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 90° C. for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layers were washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography to afford the desired compound, 1-(3-(tert-butyl)phenyl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (160 mg, 25.27%) as an off white solid.
  • LCMS: 358.2 [M+1]+
  • Step-2: Synthesis of tert-butyl 6-((1-(3-(tert-butyl)phenyl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(3-(tert-butyl)phenyl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (80 mg, 0.224 mmol, 1.0 eq) in (1 mL) of toluene was added m-CPBA (77 mg, 0.448 mmol, 2.0 eq) and allowed to stir at rt for 30 min. tert-butyl 7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (66 mg, 0.268 mmol, 1.2 eq) and DIPEA (0.3 mL, 0.896 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. The formation of precipitates was observed which was filtered to afford the desired compound tert-butyl 7-((1-(3-(tert-butyl)phenyl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (70 mg, 56.45%) as an off white solid.
  • LCMS: 556.3 [M+1]+
  • Step-3: Synthesis of 1-(3-(tert-butyl)phenyl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: tert-butyl 7-((1-(3-(tert-butyl)phenyl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (70 mg, 0.125 mmol, 1.0 eq) was dissolved in (0.9 mL) of dioxane and added 6M dioxane-HCl (0.9 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(3-(tert-butyl)phenyl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (45 mg, 39.32%) as an off white solid.
  • LCMS: 456.4 [M+1]+; UPLC @ 254 nm=98.86% and @ 220 nm=97.62%.
  • 1H NMR (400 MHz, DMSO-d6): δ10.24 (br. s., 1H), 9.34 (br. s., 2H), 8.81 (br. s., 1H), 7.60 (br. s., 1H), 7.52 (br. s., 2H), 7.47 (br. s., 2H), 7.39 (br. s., 1H), 7.10 (br. s., 1H), 4.18 (br. s., 2H), 4.06 (br. s., 1H), 3.34 (br. s., 2H), 2.93 (br. s., 2H), 1.31 (br. s., 15H)
  • Example S60. Synthesis of 2-allyl-1-(2-(tert-butyl)pyridin-4-yl)-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02426
  • Step-1: Synthesis of tert-butyl tert-butyl 6-((2-allyl-1-(2-(tert-butyl)pyridin-4-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 2-allyl-1-(2-(tert-butyl)pyridin-4-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (170 mg, 0.5 mmol, 1.0 eq) in (2.0 mL) of toluene was added m-CPBA (247 mg, 1.0 mmol, 2 eq) and allowed to stir at rt for 1 h. tert-butyl-6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (149 mg, 0.6 mmol, 1.2 eq) and DIPEA (0.36 mL, 2.0 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 6-((2-allyl-1-(2-(tert-butyl)pyridin-4-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (40 mg, 14.38%) as an off white solid. LCMS: 556.6 [M+1]4
  • Step-2: Synthesis of 2-allyl-1-(2-(tert-butyl)pyridin-4-yl)-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: tert-butyl 6-((2-allyl-1-(2-(tert-butyl)pyridin-4-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (30 mg, 0.054 mmol, 1.0 eq) was dissolved in dioxane (0.6 mL), followed by dropwise addition of 4.0 M-HCl (0.6 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 2-allyl-1-(2-(tert-butyl)pyridin-4-yl)-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (12 mg, 50%) as off white solid.
  • LCMS: 456.3 [M+1]+; UPLC @ 254 nm=87.26% and @ 220 nm=90.52%.
  • 1H NMR (400 MHz, DMSO-d6) δ 10.53 (s, 1H), 9.37 (br. s., 2H), 8.95 (s, 1H), 8.76 (d, J=6.14 Hz, 1H), 7.78 (br. s., 1H), 7.57 (d, J=8.33 Hz, 3H), 7.21 (br. s., 1H), 5.68-5.77 (m, 1H), 5.12 (d, J=10.09 Hz, 1H), 5.05 (br. s., 1H), 4.39 (d, J=5.70 Hz, 2H), 4.23 (br. s., 2H), 3.37 (br. s., 2H), 2.97-3.02 (m, 2H), 1.28-1.49 (m, 9H)
  • Example S61. Synthesis of 2-allyl-1-(2-(tert-butyl)pyridin-4-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02427
  • Step-1: 2-allyl-1-(2-(tert-butyl)pyridin-4-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-allyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (540 mg, 2.43 mmol, 1 eq) and 2-(tert-butyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine (1.26 g, 4.84 mmol, 2 eq) in DCM (50 mL) was added 2,2-bipyridine (756 mg, 4.84 mmol, 2 eq), copper acetate (880 mg, 4.84 mmol, 2 eq) and Na2CO3 (773 mg, 7.29 mmol, 3 eq). The reaction mixture was stirred at rt for 24 h in open air. The progress of reaction was monitored by TLC. After completion, the reaction mixture was filtered over celite to remove inorganic impurities. The filtrate was washed with water, dried over Na2SO4, concentrated and purified by column chromatography (Combiflash, Elution: 0-30% EtOAc in Hexane) to afford the desired product, 2-allyl-1-(2-(tert-butyl)pyridin-4-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (380 mg, 44%) as yellow solid.
  • LCMS: 356.3 [M+1]+
  • Step-2: Synthesis of tert-butyl 7-((2-allyl-1-(2-(tert-butyl)pyridin-4-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 2 2-allyl-1-(2-(tert-butyl)pyridin-4-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (170 mg, 0.5 mmol, 1.0 eq) in (2.0 mL) of toluene was added m-CPBA (247 mg, 1.0 mmol, 2 eq) and allowed to stir at rt for 1 h. tert-butyl-7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (149 mg, 0.6 mmol, 1.2 eq) and DIPEA (0.36 mL, 2.0 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 7-((2-allyl-1-(2-(tert-butyl)pyridin-4-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (30 mg, 13.14%) as an off white solid.
  • LCMS: 556.6 [M+1]+
  • Step-3: Synthesis of 2-allyl-1-(2-(tert-butyl)pyridin-4-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: tert-butyl 7-((2-allyl-1-(2-(tert-butyl)pyridin-4-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (30 mg, 0.054 mmol, 1.0 eq) was dissolved in dioxane (0.6 mL), followed by dropwise addition of 4.0 M-HCl (0.6 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 2-allyl-1-(2-(tert-butyl)pyridin-4-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (36 mg, 75.70%) as off white solid.
  • LCMS: 456.3 [M+1]+; UPLC @ 254 nm=91.79% and @ 220 nm=89.02%.
  • 1H NMR (400 MHz, DMSO-d6) δ 10.56 (br. s., 1H), 9.47 (br. s., 2H), 8.96 (s, 1H), 8.74-8.83 (m, 1H), 7.85 (br. s., 1H), 7.49-7.61 (m, 3H), 7.22 (d, J=7.89 Hz, 1H), 6.55 (s, 1H), 5.66-5.78 (m, 1H), 5.12 (d, J=10.52 Hz, 1H), 5.05 (d, J=17.10 Hz, 1H), 4.39 (d, J=5.70 Hz, 2H), 4.28 (br. s., 2H), 3.36 (br. s., 2H), 2.99 (d, J=5.70 Hz, 2H), 1.34-1.49 (m, 9H).
  • Example S62. Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02428
  • Step-1: Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-cyclopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (600 mg, 2.69 mmol, 1 eq) and 2-(tert-butyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine (1.05 g, 4.04 mmol, 1.5 eq) in DCM (100 mL) was added 2,2-bipyridine (840 mg, 5.38 mmol, 2 eq), copper acetate (977 mg, 5.38 mmol, 2 eq) and Na2CO3 (855 mg, 8.07 mmol, 3 eq). The reaction mixture was stirred at rt for 24 h in open air. The progress of reaction was monitored by TLC. After completion, the reaction mixture was filtered over celite to remove inorganic impurities. The filtrate was washed with water, dried over Na2SO4, concentrated and purified by column chromatography (Combiflash, Elution: 0-30% EtOAc in Hexane) to afford the desired product, 1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (392 mg, 40.85%) as an off white solid.
  • LCMS: 356.2 [M+1]+
  • Step-2: Synthesis of tert-butyl 6-((1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate): To a stirred solution of 1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (196 mg, 0.548 mmol, 1.0 eq) in (6.0 mL) of toluene was added m-CPBA (189 mg, 1.096 mmol, 2.0 eq) and allowed to stir at rt for 1 h. tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (163 mg, 0.657 mmol, 1.2 eq) and DIPEA (0.37 mL, 2.192 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 6-((1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (50 mg, 16.31%) as an off white solid.
  • LCMS: 556.3 [M+1]+
  • Step-3: Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride: tert-butyl 6-((1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (50 mg, 0.179 mmol, 1.0 eq) was dissolved in dioxane (0.6 mL), followed by dropwise addition of 4.0 M-HCl (0.6 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (36 mg, 75.70%) as light yellow solid.
  • LCMS: 456.4 [M+1]+; UPLC @ 254 nm=89.33% and @ 220 nm=87.71%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.39 (br. s., 1H), 9.39 (br. s., 2H), 8.87 (s, 1H), 8.76 (d, J=6.14 Hz, 1H), 7.86 (br. s., 1H), 7.71 (br. s., 1H), 7.56 (br. s., 2H), 7.19 (d, J=8.77 Hz, 1H), 4.22 (br. s., 2H), 3.37 (br. s., 2H), 3.21 (br. s., 1H), 3.01 (br. s., 2H), 1.41 (s, 9H), 0.88 (d, J=6.14 Hz, 2H), 0.83 (br. s., 2H).
  • Example S63. Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02429
  • Step-1: Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-cyclopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (600 mg, 2.69 mmol, 1 eq) and 2-(tert-butyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine (1.05 g, 4.04 mmol, 1.5 eq) in DCM (100 mL) was added 2,2-bipyridine (840 mg, 5.38 mmol, 2 eq), copper acetate (977 mg, 5.38 mmol, 2 eq) and Na2CO3 (855 mg, 8.07 mmol, 3 eq). The reaction mixture was stirred at rt for 24 h in open air. The progress of reaction was monitored by TLC. After completion, the reaction mixture was filtered over celite to remove inorganic impurities. The filtrate was washed with water, dried over Na2SO4, concentrated and purified by column chromatography (Combiflash, Elution: 0-30% EtOAc in Hexane) to afford the desired product, 1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (392 mg, 40.85%) as an off white solid.
  • LCMS: 356.2 [M+1]+
  • Step-2: Synthesis of tert-butyl 7-((1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (196 mg, 0.548 mmol, 1.0 eq) in (6.0 mL) of toluene was added m-CPBA (189 mg, 1.096 mmol, 2.0 eq) and allowed to stir at rt for 1 h. tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (163 mg, 0.657 mmol, 1.2 eq) and DIPEA (0.37 mL, 2.192 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 7-((1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (100 mg, 32.63%) as an off white solid.
  • LCMS: 556.3 [M+1]+
  • Step-3: Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride: tert-butyl 7-((1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (100 mg, 0.179 mmol, 1.0 eq) was dissolved in dioxane (1.2 mL), followed by dropwise addition of 4.0 M-HCl (1.2 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (66 mg, 69.40%) as light yellow solid.
  • LCMS: 456.4 [M+1]+; UPLC @ 254 nm=93.54% and @ 220 nm=92.28%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.41 (br. s., 1H), 9.40 (br. s., 2H), 8.87 (s, 1H), 8.78 (d, J=6.14 Hz, 1H), 7.89 (br. s., 1H), 7.72 (br. s., 1H), 7.51-7.59 (m, 2H), 7.21 (d, J=8.77 Hz, 1H), 4.27 (br. s., 2H), 3.37 (br. s., 2H), 3.22 (br. s., 1H), 2.95-3.01 (m, 2H), 1.44 (br. s., 1H), 1.41 (s, 9H), 0.89 (d, J=6.58 Hz, 2H), 0.76-0.84 (m, 2H).
  • Example S64. Synthesis of 1-(4-(tert-butyl)pyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02430
  • Step-1: Synthesis of 1-(4-(tert-butyl)pyridin-2-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (300 mg, 1.337 mmol, 1.0 eq) and 2-bromo-4-(tert-butyl)pyridine (294 mg, 1.36 mmol, 1.20 eq) in (8 mL) of dioxane was added potassium carbonate (370 mg, 2.674 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min, followed by addition of copper iodide (51 mg, 0.27 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (47 mg, 0.535 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 90° C. for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 ML), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-30% EtOAc in hexane] to afford the desired product, 1-(4-(tert-butyl)pyridin-2-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (270 mg, 56.5%) as yellow solid.
  • LCMS: 359.3 [M+1]+
  • Step-2: tert-butyl 7-((1-(4-(tert-butyl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(4-(tert-butyl)pyridin-2-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (120 mg, 0.34 mmol, 1.0 eq) in (2.0 mL) of toluene was added mCPBA (91 mg, 0.37, 1.1 eq) and allowed to stir at rt for 30 minutes. tert-butyl 7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (102 mg, 0.408 mmol, 1.2 eq) and DIPEA (0.30 mL, 1.7 mmol, 5.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography to afford the desired product, tert-butyl 7-((1-(4-(tert-butyl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (68 mg, 35.98%) as brown solid.
  • LCMS: 558.5 [M+1]+
  • Step-3: Synthesis of 1-(4-(tert-butyl)pyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: tert-butyl 7-((1-(4-(tert-butyl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (65 mg, 0.117 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (0.5 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound 1-(4-(tert-butyl)pyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (46 mg, 95.83%) as an off white solid.
  • LCMS: 458.3 [M+1]+; UPLC @ 254 nm=98.87% and @ 220 nm=99.76%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.22 (br. s., 1H), 9.31 (br. s., 2H), 8.83 (s, 1H), 8.50 (d, J=5.26 Hz, 1H), 7.65 (s, 2H), 7.49 (d, J=3.95 Hz, 1H), 7.42 (br. s., 1H), 7.09 (d, J=8.33 Hz, 1H), 4.17 (br. s., 2H), 3.33 (br. s., 2H), 2.90-2.97 (m, 2H), 1.33 (s, 9H), 1.26 (d, J=7.02 Hz, 6H).
  • Example S65. Synthesis of 1-(4-(tert-butyl)pyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02431
  • Step-1: tert-butyl 6-((1-(4-(tert-butyl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(4-(tert-butyl)pyridin-2-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (120 mg, 0.34 mmol, 1.0 eq) in (2.0 mL) of toluene was added mCPBA (91 mg, 0.37, 1.1 eq) and allowed to stir at rt for 30 minutes. tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (102 mg, 0.408 mmol, 1.2 eq) and DIPEA (0.30 mL, 1.7 mmol, 5.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography to afford the desired product, tert-butyl 6-((1-(4-(tert-butyl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (50 mg, 26.24%) as brown solid.
  • LCMS: 558.5 [M+1]+
  • Step-2: Synthesis of 1-(4-(tert-butyl)pyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: tert-butyl 6-((1-(4-(tert-butyl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (50 mg, 0.09 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (0.5 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound 1-(4-(tert-butyl)pyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (40 mg, 97.5%) as light brown solid.
  • LCMS: 458.3 [M+1]+; UPLC @ 254 nm=96.51% and @ 220 nm=99.29%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.22 (br. s., 1H), 9.42 (br. s., 2H), 8.84 (s, 1H), 8.51 (d, J=5.26 Hz, 1H), 7.59-7.69 (m, 2H), 7.44-7.52 (m, 2H), 7.07 (d, J=8.33 Hz, 1H), 4.12-4.21 (m, 3H), 3.33 (br. s., 2H), 2.92 (br. s., 2H), 1.33 (s, 9H), 1.25 (d, J=6.58 Hz, 6H).
  • Example S66. Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-6-((2′,3′-dihydro-1′H-spiro[cyclopropane-1,4′-isoquinolin]-7′-yl)amino)-2-isopropyl-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02432
  • Step-1: Synthesis of tert-butyl 7′-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-1′H-spiro[cyclopropane-1,4′-isoquinoline]-2′(3′H)-carboxylate: To a stirred solution of 1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (150 mg, 0.419 mmol, 1.0 eq) in (6.0 mL) of toluene was added m-CPBA (145 mg, 0.838 mmol, 2.0 eq) and allowed to stir at rt for 1 h. tert-butyl 7′-amino-1′H-spiro[cyclopropane-1,4′-isoquinoline]-2′(3′H)-carboxylate (138 mg, 0.503 mmol, 1.2 eq) and DIPEA (0.28 mL, 1.676 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 7′-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-1′H-spiro[cyclopropane-1,4′-isoquinoline]-2′(3′H)-carboxylate (110 mg, 61.24%) as an off white solid.
  • LCMS: 584.3 [M+1]+
  • Step-2: Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-6-((2′,3′-dihydro-1′H-spiro[cyclopropane-1,4′-isoquinolin]-7′-yl)amino)-2-isopropyl-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride: tert-butyl 7′-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-1′H-spiro[cyclopropane-1,4′-isoquinoline]-2′(3′H)-carboxylate (110 mg, 0.188 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (2 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(2-(tert-butyl)pyridin-4-yl)-6-((2′,3′-dihydro-1′H-spiro[cyclopropane-1,4′-isoquinolin]-7′-yl)amino)-2-isopropyl-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (63 mg, 60.07%) as light yellow solid.
  • LCMS: 484.4 [M+1]+; UPLC @ 254 nm=95.36% and @ 220 nm=97.34%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.53 (br. s., 1H), 9.49 (br. s., 2H), 8.88 (s, 1H), 8.80 (br. s., 1H), 7.80 (br. s., 1H), 7.54 (br. s., 3H), 6.85 (d, J=8.33 Hz, 1H), 4.39 (br. s., 2H), 3.26 (br. s., 3H), 1.25-1.52 (m, 14H), 1.09 (br. s., 4H).
  • Example S67. Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-6-((4,4-dimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-2-isopropyl-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02433
  • Step-1: Synthesis of tert-butyl 7-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-4,4-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (150 mg, 0.419 mmol, 1.0 eq) in (6.0 mL) of toluene was added m-CPBA (145 mg, 0.838 mmol, 2.0 eq) and allowed to stir at rt for 1 h. tert-butyl 7-amino-4,4-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (138 mg, 0.503 mmol, 1.2 eq) and DIPEA (0.28 mL, 1.676 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 7-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-4,4-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (60 mg, 24.41%) as an off white solid.
  • LCMS: 586.3 [M+1]+
  • Step-2: Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-6-((4,4-dimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-2-isopropyl-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride: tert-butyl 7-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-4,4-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (60 mg, 0.102 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (2 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(2-(tert-butyl)pyridin-4-yl)-6-((4,4-dimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-2-isopropyl-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (36 mg, 62.92%) as light yellow solid.
  • LCMS: 486.4 [M+1]+; UPLC @ 254 nm=96.12% and @ 220 nm=97.81%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.48 (br. s., 1H), 9.31 (br. s., 2H), 8.88 (s, 1H), 8.76 (br. s., 1H), 7.72 (br. s., 1H), 7.45-7.58 (m, 3H), 7.43 (br. s., 1H), 4.27 (br. s., 2H), 3.89-4.01 (m, 2H), 3.23 (br. s., 2H), 1.16-1.44 (m, 21H).
  • Example S68. Synthesis of 1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02434
  • Step-1: Synthesis of 4-bromo-2-(2-fluoropropan-2-yl)pyridine: To a stirred solution of 2-(4-bromopyridin-2-yl)propan-2-ol (0.5 g, 2.313 mmol, 1.0 eq) in DCM (7 mL) was added DAST (0.33 mL, 2.545 mmol, 1.1 eq) at −78° C. The reaction mixture was stirred at rt for 12 h. After completion of reaction, the reaction mixture was quenched with NaHCO3 solution and was extracted with DCM (100 mL×2). The combined organic layer were dried over Na2SO4, concentrated under reduced pressure and purified by column chromatography [Combiflash, elution: 0-50% EtOAc in hexane] to afford the desired compound, 4-bromo-2-(2-fluoropropan-2-yl)pyridine (250 mg, 49.54%) as yellow liquid.
  • LCMS: 218.2/220.2 [M+2]+
  • Step-2: Synthesis of 1-(2-(2-fluoropropan-2-yl) pyridin-4-yl)-2-isopropyl-6-(methylthio)-1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one: To a stirred solution of 2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (500 mg, 2.22 mmol, 1.0 eq) and 4-bromo-2-(2-fluoropropan-2-yl)pyridine (580 mg, 2.675 mmol, 1.2 eq) in (8 mL) of dioxane were added Potassium carbonate (620 mg, 4.45 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min, followed by addition of copper iodide (85 mg, 0.44 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (0.1 mL, 0.891 mmol, 2 eq) and again purged with nitrogen for 10 min, stirred at 130° C. for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (30 mL×2). The combined organic layers were washed with water (30 mL), brine solution (30 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography to afford the desired compound, 2-isopropyl-1-(6-(2-hydroxypropan-2-yl)pyridin-2-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (150 mg, 68.19%) as an off white solid.
  • LCMS: 362.3 [M+1]+
  • Step-3: Synthesis of tert-butyl 6-((1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (150 mg, 0.415 mmol, 1.0 eq) in (5 mL) of toluene was added m-CPBA (143.4 mg, 0.830 mmol, 2.0 eq) and allowed to stir at rt for 30 min. tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (123 mg, 0.498 mmol, 1.2 eq) and DIPEA (0.3 mL, 1.662 mmol, 4.0 eq) were added and allowed to stir at 80° C. for overnight. Solvent was evaporated and reaction mass was diluted with water and extracted with EtOAc (30 mL×2). The combined organic layer were dried over sodium sulphate, concentrated under reduced pressure purified by column chromatography (Combiflash, elution-0-70% EtOAc in Hexane) to afford the desired compound, tert-butyl 6-((1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (52 mg, 22.30%) as yellow liquid.
  • LCMS: 562.5 [M+1]+
  • Step-4: Synthesis of 1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride: To a stirred solution of tert-butyl 6-((1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (52 mg, 92.691 mmol, 1.0 eq) in (1 mL) of dioxane was added 4M dioxane-HCl (0.6 mL) and allowed to stir at rt for 2 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (20 mg, 40.42%) as yellow solid.
  • LCMS: 462.3 [M+1]+, UPLC @ 254 nm=86.26% and @ 220 nm=92.47%.
  • 1H NMR (400 MHz, DMSO-d6): 10.48 (br. s., 1H), 9.08 (br. s., 2H), 8.87 (s, 1H), 8.72 (d, J=5.26 Hz, 1H), 7.60 (s, 1H), 7.63 (s, 2H), 7.18 (d, J=7.89 Hz, 1H), 4.23 (br. s., 2H), 3.97 (d, J=6.14 Hz, 2H), 3.63 (br. s., 8H), 3.01 (br. s., 2H), 1.72 (s, 2H), 1.67 (s, 2H), 1.36 (d, J=6.58 Hz, 7H), 1.24 (br. s., 1H)
  • Example S69. Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-6-((2-(dimethylamino)-2,3-dihydro-JH-inden-5-yl)amino)-2-isopropyl-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02435
  • To a stirred solution of 1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (90 mg, 0.251 mmol, 1.0 eq) in (2.0 mL) of toluene was added mCPBA (124 mg, 0.502 mmol, 2.0 eq) and allowed to stir at rt for 30 minutes. N2,N2-dimethyl-2,3-dihydro-1H-indene-2,5-diamine (62 mg, 0.302 mmol, 1.2 eq) and DIPEA (0.175 mL, 1.004 mmol, 4.0 eq) were added and allowed to stir at 100° C. for overnight. After completion of reaction, the reaction mixture was diluted with water (20 mL) and extracted with EtOAc (20 mL×2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by reverse phase chromatography to afford the desired 1-(2-(tert-butyl)pyridin-4-yl)-6-((2-(dimethylamino)-2,3-dihydro-1H-inden-5-yl)amino)-2-isopropyl-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (6.0 mg, 4.91%) as off white solid.
  • LCMS: 486.4 [M+1]+; UPLC @ 254 nm=97.62% and @ 220 nm=96.06%.
  • 1H NMR (400 MHz, DMSO-d6) δ 10.28 (brs, 1H), 8.81 (s, 1H), 8.64 (d, J=5.26 Hz, 1H), 7.68 (brs, 1H), 7.52 (brs, 1H), 7.43 (brs, 2H), 7.12 (d, J=7.89 Hz, 1H), 3.94 (d, J=6.58 Hz, 1H), 3.66 (brs, 1H), 2.96 (brs, 2H), 2.70 (brs, 2H), 2.20 (s, 6H), 1.28-1.43 (m, 15H).
  • Example S70. Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-6-(isoindolin-5-ylamino)-2-isopropyl-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02436
  • Step-1: Synthesis of tert-butyl 5-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)isoindoline-2-carboxylate: To a stirred solution of 1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-indazol-3-one (120 mg, 0.34 mmol, 1.0 eq) in (2.0 mL) of toluene was added m-CPBA (91 mg, 0.37, 1.1 eq) and allowed to stir at rt for 30 minutes. tert-butyl 5-aminoisoindoline-2-carboxylate (102 mg, 408 mmol, 1.2 eq) and DIPEA (0.30 mL, 1.7 mmol, 5.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography to afford the desired product, tert-butyl 5-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)isoindoline-2-carboxylate (40 mg, 21.73%) as brown solid.
  • LCMS: 544.6 [M+1]+
  • Step-2: Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-6-(isoindolin-5-ylamino)-2-isopropyl-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: tert-butyl 5-((1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)isoindoline-2-carboxylate (130 mg, 1.321 mmol, 1.0 eq) was dissolved in 4.0 M-HCl (1.5 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound 1-(6-(tert-butyl)pyridin-2-yl)-2-ethyl-6-(isoindolin-5-ylamino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (11 mg, 27.50%) as brown solid.
  • LCMS: 444.3 [M+1]+; UPLC @ 254 nm=90.44% and @ 220 nm=96.36%.
  • 1H NMR (400 MHz, DMSO-d6) δ 10.62 (br. s., 1H), 9.99 (br. s., 2H), 8.91 (s, 1H), 8.79 (d, J=4.38 Hz, 1H), 7.85 (br. s., 1H), 7.74 (br. s., 1H), 7.56-7.68 (m, 3H), 7.39 (d, J=8.33 Hz, 2H), 4.53 (br. s., 2H), 4.47 (br. s., 2H), 3.96 (m, 1H), 1.35-1.45 (m, 15H).
  • Example S71. Synthesis of 1-(4-fluorophenyl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride
  • Figure US20230109104A1-20230406-C02437
  • Step-1: Synthesis of 1-(4-fluorophenyl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (500 mg, 2.22 mmol) and 2-(4-fluorophenyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (374 g, 2.67 mmol) in DCM (100 mL) was added 2,2-bipyridine (520 mg, 3.33 mmol), copper acetate (605 mg, 3.33 mmol) and Na2CO3 (706 mg, 6.66 mmol). The reaction mixture was stirred at rt for 24 h in open air. The progress of reaction was monitored by TLC. After completion, the reaction mixture was filtered over celite to remove inorganic impurities. The filtrate was washed with water, dried over Na2SO4, concentrated and purified by column chromatography (Combiflash, Elution: 0-30% EtOAc in Hexane) to afford the desired product, 1-(4-fluorophenyl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (270 mg, 38.04%) as an off white solid.
  • LCMS: 319.2 [M+1]+
  • Step-2: Synthesis of tert-butyl 6-((1-(4-fluorophenyl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(4-fluorophenyl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (135 mg, 0.424 mmol, 1.0 eq) in (6.0 mL) of toluene was added m-CPBA (146 mg, 0.848 mmol, 2.0 eq) and allowed to stir at rt for 1 h. tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (126 mg, 0.508 mmol, 1.2 eq) and DIPEA (0.29 mL, 1.69 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 6-((1-(4-fluorophenyl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (50 mg, 22.73%) as an off white solid.
  • LCMS: 519.3 [M+1]+
  • Step-3: Synthesis of 1-(4-fluorophenyl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride: tert-butyl 6-((1-(4-fluorophenyl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (50 mg, 0.179 mmol, 1.0 eq) was dissolved in dioxane (0.6 mL), followed by dropwise addition of 4.0 M-HCl (0.6 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(4-fluorophenyl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride (10 mg, 24.78%) as light yellow solid.
  • LCMS: 419.3 [M+1]+; UPLC @ 254 nm=98.11% and @ 220 nm=98.37%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.29 (brs, 1H), 9.05 (brs, 2H), 8.83 (s, 1H), 7.69 (brs, 1H), 7.61 (d, J=4.82 Hz, 2H), 7.29-7.49 (m, 3H), 7.13 (d, J=8.77 Hz, 1H), 4.20 (brs, 2H), 4.02-4.12 (m, 1H), 2.93 (brs, 2H), 1.35 (brs, 1H), 1.27 (d, J=6.58 Hz, 6H).
  • Example S72. Synthesis of 1-(4-fluorophenyl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02438
  • Step-1: Synthesis of 1-(4-fluorophenyl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (500 mg, 2.22 mmol) and 2-(4-fluorophenyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (374 g, 2.67 mmol) in DCM (100 mL) was added 2,2-bipyridine (520 mg, 3.33 mmol), copper acetate (605 mg, 3.33 mmol) and Na2CO3 (706 mg, 6.66 mmol). The reaction mixture was stirred at rt for 24 h in open air. The progress of reaction was monitored by TLC. After completion, the reaction mixture was filtered over celite to remove inorganic impurities. The filtrate was washed with water, dried over Na2SO4, concentrated and purified by column chromatography (Combiflash, Elution: 0-30% EtOAc in Hexane) to afford the desired product, 1-(4-fluorophenyl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (270 mg, 38.04%) as an off white solid.
  • LCMS: 319.2 [M+1]+
  • Step-2: Synthesis of tert-butyl 7-((1-(4-fluorophenyl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(4-fluorophenyl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (135 mg, 0.424 mmol, 1.0 eq) in (6.0 mL) of toluene was added m-CPBA (146 mg, 0.848 mmol, 2.0 eq) and allowed to stir at rt for 1 h. tert-butyl 7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (126 mg, 0.508 mmol, 1.2 eq) and DIPEA (0.29 mL, 1.69 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 7-((1-(4-fluorophenyl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (50 mg, 22.73%) as an off white solid.
  • LCMS: 519.3 [M+1]+
  • Step-3: Synthesis of 1-(4-fluorophenyl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: tert-butyl 7-((1-(4-fluorophenyl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (50 mg, 0.179 mmol, 1.0 eq) was dissolved in dioxane (0.6 mL), followed by dropwise addition of 4.0 M-HCl (0.6 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(4-fluorophenyl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (10 mg, 24.78%) as light yellow solid.
  • LCMS: 419.3 [M+1]+; UPLC @ 254 nm=98.25% and @ 220 nm=98.37%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.26 (brs, 1H), 9.05 (brs, 2H), 8.83 (s, 1H), 7.70 (brs, 1H), 7.55-7.66 (m, 2H), 7.30-7.47 (m, 3H), 7.11 (d, J=8.33 Hz, 1H), 4.19 (brs, 2H), 4.06-4.14 (m, 1H), 2.92 (brs, 2H), 1.27 (d, J=7.02 Hz, 6H).
  • Example S73. Synthesis of 1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02439
  • Step-1: Synthesis of tert-butyl 7-((1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroquinoline-1(2H)-carboxylate: To a stirred solution of 1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (150 mg, 0.415 mmol, 1.0 eq) in (5 mL) of toluene was added m-CPBA (143.4 mg, 0.830 mmol, 2.0 eq) and allowed to stir at rt for 30 min. tert-butyl 7-amino-3,4-dihydroquinoline-1(2H)-carboxylate, (123 mg, 0.498 mmol, 1.2 eq) and DIPEA (0.3 mL, 1.662 mmol, 4.0 eq) were added and allowed to stir at 80° C. for overnight. Solvent was evaporated and reaction mass was diluted with water and extracted with EtOAc (30 mL×2). The combined organic layer were dried over sodium sulphate, concentrated under reduced pressure purified by column chromatography (Combiflash, elution-0-70% EtOAc in Hexane) to afford the desired compound, tert-butyl 7-((1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroquinoline-1(2H)-carboxylate, (82 mg, 35.17%) as yellow liquid.
  • LCMS: 562.5 [M+1]+
  • Step-2: Synthesis of 1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride: tert-butyl 7-((1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroquinoline-1(2H)-carboxylate (82 mg, 0.146 mmol, 1.0 eq) was dissolved in (1 mL) of dioxane and added 4M dioxane-HCl (0.8 mL) and allowed to stir at rt for 2 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (25 mg, 32.04%) as white solid.
  • LCMS: 462.3 [M+1]+, UPLC @ 254 nm=91.28% and @ 220 nm=99.61%.
  • 1H NMR (400 MHz, DMSO-d6): δ 8.84 (s, 1H), 8.72 (d, J=4.82 Hz, 1H), 7.66 (brs, 1H), 7.58 (brs, 1H), 7.03 (brs, 2H), 3.95 (d, J=5.70 Hz, 2H), 3.28 (brs, 1H), 2.71 (brs, 2H), 1.89 (brs, 2H), 1.72 (s, 3H), 1.67 (s, 3H), 1.36 (d, J=6.58 Hz, 6H).
  • Example S74. Synthesis of 1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02440
  • Step-1: Synthesis of tert-butyl 7-((1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (140 mg, 0.387 mmol, 1.0 eq) in (6 mL) of toluene was added m-CPBA (133.6 mg, 0.775 mmol, 2.0 eq) and allowed to stir at rt for 30 min. tert-butyl 7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate, (115 mg, 0.465 mmol, 1.2 eq) and DIPEA (0.27 mL, 1.551 mmol, 4.0 eq) were added and allowed to stir at 80° C. for overnight. Solvent was evaporated and reaction mass was diluted with water and extracted with EtOAc (30 mL×2). The combined organic layer were dried over sodium sulphate, concentrated under reduced pressure purified by column chromatography (Combiflash, elution-0-70% EtOAc in Hexane) to afford the desired compound, tert-butyl 7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylateAc in Hexane) to afford the desired compound, tert-butyl 7-((1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (70 mg, 32.17%) as yellow liquid.
  • LCMS: 562.5 [M+1]+
  • Step-2: Synthesis of 1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride: tert-butyl 7-((1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (70 mg, 0.124 mmol, 1.0 eq) was dissolved in (2 ML) of dioxane and added 4M dioxane-HCl (0.7 mL) and allowed to stir at rt for 2 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (15 mg, 26.07%) as white solid.
  • LCMS: 462.3 [M+1]+, UPLC @ 254 nm=95.42% and @ 220 nm=97.82%.
  • 1H NMR (400 MHz, DMSO-d6): δ 9.05 (brs, 2H), 8.87 (s, 1H), 8.72 (brs, 1H), 7.63 (brs, 2H), 7.56 (brs, 1H), 7.20 (d, J=8.77 Hz, 1H), 4.29 (brs, 3H), 3.96 (brs., 2H), 2.96 (brs, 2H), 1.72 (s, 3H), 1.66 (s, 3H), 1.36 (d, J=6.58 Hz, 6H).
  • Example S75a. Synthesis of 1-(2-tert-butylpyridin-4-yl)-6-{[6-(dimethylamino)-5,6,7,8-tetrahydronaphthalen-2-yl]amino}-2-(propan-2-yl)-1H, 2H, 3H-pyrazolo[3,4-d]pyrimidin-3-one (Peak-I: Compound No. 1.170); and Example S75b. Synthesis of 1-(2-tert-butylpyridin-4-yl)-6-{[7-(dimethylamino)-5,6,7,8-tetrahydronaphthalen-2-yl]amino}-2-(propan-2-yl)-1H, 2H, 3H-pyrazolo[3,4-d]pyrimidin-3-one (Peak-II: Compound No. 1.171)
  • Figure US20230109104A1-20230406-C02441
  • To a stirred solution of 1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-6-(methylthio)-1 dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (210 mg, 0.587 mmol, 1.0 eq) in (5.0 mL) of toluene was added mCPBA (290 mg, 1.174 mmol, 2.0 eq) and allowed to stir at rt for 30 minutes. N2,N2-dimethyl-1,2,3,4-tetrahydronaphthalene-2,6-diamine and N2,N2-dimethyl-1,2,3,4-tetrahydro naphthalene-2,7-diamine (135 mg, 0.704 mmol, 1.2 eq) and DIPEA (0.41 mL, 2.348 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water (50 mL) extracted with EtOAc (2×50 mL). The combined organic extracts were washed with water (50 mL), with brine (50 mL) dried over Na2SO4 and concentrated under reduced pressure to afford mixture of desired compound 1-(2-(tert-butyl)pyridin-4-yl)-6-((6-(dimethylamino)-5,6,7,8-tetrahydronaphthalen-2-yl)amino)-2-isopropyl-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one and 1-(2-(tert-butyl)pyridin-4-yl)-6-((7-(dimethylamino)-5,6,7,8-tetrahydronaphthalen-2-yl)amino)-2-isopropyl-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one, which was purified by reverse phase purification as PK-I and PK-II (PK-I; 10.0 mg, 3.41%, PK-II; 11.0 mg, 3.75%) both as white solid.
  • LCMS: 500.4 [M+1]+; PK-I; UPLC @ 254 nm=97.35% and @ 220 nm=97.52%.
  • PK-II; UPLC @ 254 nm=99.89% and @ 220 nm=99.87%.
  • PK-I: 1H NMR (400 MHz, DMSO-d6) δ 10.30 (brs, 1H), 8.83 (s, 1H), 8.67 (d, J=5.26 Hz, 1H), 7.44-7.57 (m, 3H), 7.40 (brs, 1H), 7.07 (d, J=8.77 Hz, 1H), 3.95 (brs, 1H), 3.01 (brs, 1H), 2.87 (brs, 3H), 2.67 (brs, 6H), 2.20 (brs, 2H), 1.72 (brs, 1H), 1.49 (brs, 1H), 1.28-1.42 (m, 15H).
  • PK-II: 1H NMR (400 MHz, DMSO-d6) δ 10.29 (brs, 1H), 8.82 (s, 1H), 8.66 (brs, 1H), 7.53 (brs, 1H), 7.48 (brs, 1H), 7.40 (brs, 2H), 7.03 (d, J=8.77 Hz, 1H), 3.95 (brs, 1H), 2.90 (brs, 2H), 2.82 (brs, 2H), 2.73 (brs, 2H), 2.67 (br., 2H), 2.10 (brs, 1H), 1.62 (brs, 1H), 1.25-1.48 (m, 15H).
  • Example S76. Synthesis of 1-(2-(dimethylamino)pyridin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02442
  • Step-1: Synthesis of 4-bromo-N,N-dimethylpyridin-2-amine: To a solution of compound 2-amino-4-bromopyridine (704 mg, 4.0 mmol, 1.0 eq) in THF at 0° C. was added NaH (352 mg, 8.8 mmol, 2.2 eq) and the reaction mixture was stirred for 30 minutes. To this solution was added methyl iodide at 0° C. (0.622 mL, 10 mmol, 2.5 eq) and reaction mixture was warmed to rt. Reaction was allowed to stir at rt for 16 h and the progress of the reaction was monitored by TLC. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, 4-bromo-N,N-dimethylpyridin-2-amine (500 mg, 62.5%) as a brown solid.
  • LCMS: 201.0 [M+1]+
  • Step-2: Synthesis of 1-(2-(dimethylamino)pyridin-4-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (300 mg, 1.337 mmol, 1.0 eq) and -bromo-N,N-dimethylpyridin-2-amine (321 mg, 1.60 mmol, 1.20 eq) in (8 mL) of dioxane was added potassium carbonate (370 mg, 2.674 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min, followed by addition of copper iodide (51 mg, 0.27 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (47 mg, 0.535 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 90° C. for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-30% EtOAc in hexane] to afford the desired product, 1-(2-(dimethylamino)pyridin-4-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (60 mg, 13.1%) as brown solid.
  • LCMS: 345.2 [M+1]+
  • Step-3: Synthesis of tert-butyl 6-((1-(2-(dimethylamino)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(2-(dimethylamino)pyridin-4-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (55 mg, 0.145 mmol, 1.0 eq) in (2.0 mL) of toluene was added mCPBA (40 mg, 0.16, 1.1 eq) and allowed to stir at rt for 30 minutes. tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (43 mg, 0.174 mmol, 1.2 eq) and DIPEA (0.13 mL, 0.725 mmol, 5.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography to afford the desired product, tert-butyl 6-((1-(2-(dimethylamino)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (20 mg, 25.3%) as brown solid.
  • LCMS: 546.3 [M+1]+
  • Step-4: Synthesis of 1-(2-(dimethylamino)pyridin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: tert-butyl 6-((1-(2-(dimethylamino)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (20 mg, 0.036 mmol, 1.0 eq ) was dissolved in dioxane (0.3 mL), followed by dropwise addition of 4.0 M-HCl (0.3 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried. Purified product was obtained by preparative chromatography to give 1-(2-(dimethylamino)pyridin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (10 mg, 62%) as off white solid.
  • LCMS: 445.3 [M+1]+; UPLC @ 254 nm=83.56% and @ 220 nm=88.54%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.53 (brs, 1H), 9.53 (brs, 2H), 8.90 (s, 1H), 8.14 (d, J=6.58 Hz, 1H), 7.49-7.61 (m, 2H), 7.13-7.23 (m, 2H), 6.99 (brs, 1H), 4.22 (brs, 2H), 3.98 (brs, 1H), 3.35 (brs, 2H), 3.23 (brs, 6H), 2.99 (brs, 2H), 1.38 (d, J=6.58 Hz, 6H).
  • Example S77. Synthesis of methyl 6-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-JH-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate
  • Figure US20230109104A1-20230406-C02443
  • To a stirred solution of 1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (110 mg, 0.240 mmol) in DMF (3 mL) was added TEA (0.1 mL, 0.720 mmol), followed by addition of methyl carbonochloridate (0.02 mL, 0.288 mmol) at 0° C. The reaction mixture was stirred at rt for 1 h. The reaction mixture was diluted with water (10 ML), extracted with EtOAc (50 mL×2). The combined organic layers were dried over Na2SO4, concentrated and purified by column chromatography (Combiflash, Elution: 0-70% EtOAc in Hexane) to afford the desired product, 1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (36 mg, 29.04%) as an off white solid.
  • LCMS: 516.4 [M+1]+; UPLC @ 254 nm=95.88% and @ 220 nm=97.00%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.35 (brs, 1H), 8.84 (s, 1H), 8.69 (d, J=5.26 Hz, 1H), 7.47-7.58 (m, 3H), 7.42 (brs, 1H), 7.13 (d, J=7.45 Hz, 1H), 4.51 (brs, 2H), 3.94 (d, J=6.58 Hz, 1H), 3.60-3.67 (m, 4H), 2.79 (brs, 2H), 1.26-1.44 (m, 15H).
  • Example S78. Synthesis of 1-(2-tert-butylpyridin-4-yl)-6-(1,2,3,4-tetrahydroisoquinolin-7-ylamino)-2-(2,2,2-trifluoroethyl)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one
  • Figure US20230109104A1-20230406-C02444
  • Step-1: Synthesis of 1-(2-tert-butylpyridin-4-yl)-6-(methylthio)-2-(2,2,2-trifluoroethyl)-1H-pyrazolo [3,4-d]pyrimidin-3(2H)-one: To a stirred solution of 6-(methylthio)-2-(2,2,2-trifluoroethyl)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one (440 mg, 1.665 mmol, 1.0 eq) and 2-tert-butyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine (652 mg, 2.49 mmol, 1.5 eq) in DCM (20 mL) was added Na2CO3 (529 mg, 4.99 mmol, 3.0 eq), Cu(OAc)2 (604 mg, 3.33 mmol, 2.0 eq) and Bipyridyl (520 mg, 3.33 mmol, 2 eq) and the mixture was stirred in presence of oxygen at Rt for 16 h. The reaction was monitored by TLC. After completion of reaction, the volatiles are removed under reduced pressure and the residue was taken in water and extracted with EtOAc (150 mL×2). The combined organic layers were washed with water (50 mL), brine (50 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford crude which was purified by flash chromatography (Teledyne Isco Rf+); compound eluting 50% EtOAc/Hexane to afford 1-(2-tert-butylpyridin-4-yl)-6-(methylthio)-2-(2,2,2-trifluoroethyl)-1H-pyrazolo [3,4-d]pyrimidin-3(2H)-one (350 mg, 53%) as an off white solid.
  • LCMS: 398 [M+1]+
  • Step-2: Synthesis of tert-butyl 7-(1-(2-tert-butylpyridin-4-yl)-3-oxo-2-(2,2,2-trifluoroethyl)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(2-tert-butylpyridin-4-yl)-6-(methylthio)-2-(2,2,2-trifluoroethyl)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one (150 mg, 0.377 mmol, 1.0 eq) in toluene (3 mL) was added m-CPBA (260.2 mg, 1.508 mmol, 4.0 eq) at 0° C. and the mixture was allowed to stir at RT for 1 h, followed by addition of DIPEA (292.02 mg, 2.262 mmol, 6.0 eq) and tert-butyl 7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (118.04 mg, 0.415 mmol, 1.1 eq) and the mixture was allowed to stir at RT for 1 h. The reaction was monitored by TLC. After completion of reaction, the mixture was diluted with water and extracted with EtOAc (100 mL×2). The combined organic layers were washed with water (50 mL), brine (50 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford a crude which was purified by flash chromatography (Teledyne Isco Rf+); compound eluting 70% EtOAc/Hexane to afford tert-butyl 7-(1-(2-tert-butylpyridin-4-yl)-3-oxo-2-(2,2,2-trifluoroethyl)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (30 mg, 13%) as off white solid.
  • LCMS: 598 [M+1]+
  • Step-3: Synthesis of 1-(2-tert-butylpyridin-4-yl)-6-(1,2,3,4-tetrahydroisoquinolin-7-ylamino)-2-(2,2,2-trifluoroethyl)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one: The tert-butyl 7-(1-(2-tert-butylpyridin-4-yl)-3-oxo-2-(2,2,2-trifluoroethyl)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (30 mg, 0.0502 mmol, 1.0 eq) was added 4N-HCl in 1,4-Dioxane (2 mL) and the mixture was stirred was stirred at RT for 16 h. The reaction was monitored by TLC. After completion, the volatiles are removed under reduced pressure and the residue was taken in saturated NaHCO3 (100 mL) and extracted with EtOAc (200 mL×2). The combined organic layers were washed with water (150 mL), brine (100 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure to afford crude which was purified by reversed-phase chromatography to afford 1-(2-tert-butylpyridin-4-yl)-6-(1,2,3,4-tetrahydroisoquinolin-7-ylamino)-2-(2,2,2-trifluoroethyl)-1H-pyrazolo[3,4-d]pyrimidin-3(2H)-one (3 mg, 13%) as a formate salt (off-white solid).
  • LCMS: 498 [M+1]+
  • 1H NMR (400 MHz, DMSO-d6): δ 8.90 (s, 1H), 8.64 (d, 1H), 7.61-7.42 (m, 4H), 7.22 (d, 1H), 4.26 (s, 2H), 3.23 (m, 2H), 3.03 (m, 2H), 2.16 (s, 1H), 1.40 (s, 6H).
  • Example S79. Synthesis of 1-(3-(tert-butyl)-4-(dimethylamino)phenyl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02445
  • Step-1: Synthesis of 4-bromo-2-(tert-butyl)-N,N-dimethylaniline: To a stirred solution of 4-bromo-2-(tert-butyl)aniline (500 mg, 2.192 mmol, 1.0 eq) and HCHO (0.822 mL, 10.92 mmol, 5.0 eq) in dichloroethane (10 mL) was dropwise added acetic acid (0.65 mL, 10.92 mmol, 5.0 eq) at 0° C. The resulting mixture was stirred at rt for 1 h, followed by addition of Na(COCH3)3BH (1.4 g, 6.576 mmol, 3.0 eq) at 0° C. The resulting mixture was stirred at rt for 1 h. The progress of reaction was monitored by LCMS. The resection mixture was concentrated, basified with saturated solution of NaHCO3(50 mL) extracted with EtOAc (2×50 mL). The combined organic extracts were washed with water (50 mL), with brine (50 mL) dried over Na2SO4 and concentrated under reduced and purified by combi flash chromatography [silica gel, 100-200 mesh; elution 0-1% EtOAc in hexane] to afford the desired compound 4-bromo-2-(tert-butyl)-N,N-dimethylaniline (350 mg, 62.38%) as white solid.
  • LCMS: 256.0 [M+1]+
  • Step-2: Synthesis of 1-(3-(tert-butyl)-4-(dimethylamino)phenyl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (550 mg, 2.452 mmol, 1.0 eq) and 4-bromo-2-(tert-butyl)-N,N-dimethylaniline (700 mg, 2.697 mmol, 1.10 eq) in (20 mL) of dioxane was added Potassium carbonate (678 mg, 4.904 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min., followed by addition of copper iodide (103 mg, 0.539 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (0.13 mL, 1.226 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 90° C. for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound 1-(3-(tert-butyl)-4-(dimethylamino)phenyl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (60 mg, 6.12%) as light yellow solid.
  • LCMS: 400.2 [M+1]+
  • Step-3: Synthesis of tert-butyl 6-((1-(3-(tert-butyl)-4-(dimethylamino)phenyl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(3-(tert-butyl)-4-(dimethylamino)phenyl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (60 mg, 0.150 mmol, 1.0 eq) in (2.0 mL) of toluene was added m-CPBA (74 mg, 0.30 mmol, 2.0 eq) and allowed to stir at rt for 30 min. tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (41 mg, 0.165 mmol, 1.1 eq) and DIPEA (0.104 mL, 0.600 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (20 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 ML), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-60% EtOAc in hexane] to afford the desired compound, tert-butyl 6-((1-(3-(tert-butyl)-4-(dimethylamino)phenyl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (62 mg, 68.88%) as brown viscous.
  • LCMS: 600.4 [M+1]+
  • Step-4: Synthesis of 1-(3-(tert-butyl)-4-(dimethylamino)phenyl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: tert-butyl 6-((1-(3-(tert-butyl)-4-(dimethylamino)phenyl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (60 mg, 1.00 mmol, 1.0 eq) was dissolved in 4.0 M-HCl (1.5 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was basified with NaHCO3 water and extracted with EtOAc (20 mL×2). The combined organic layers were washed with water (50 mL) brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by reverse phase chromatography to afford the desired compound 1-(3-(tert-butyl)-4-(dimethylamino)phenyl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (4.0 mg, 8.16%) as white solid.
  • LCMS: 500.4 [M+1]+; UPLC @ 254 nm=94.68% and @ 220 nm=95.49%.
  • 1H NMR (400 MHz, DMSO-d6) δ 10.08 (s, 1H), 8.77 (s, 1H), 8.29 (s, 1H), 7.63 (d, J=8.33 Hz, 1H), 7.46-7.55 (m, 2H), 7.40 (s, 1H), 7.28 (br. s., 1H), 6.91 (d, J=8.33 Hz, 1H), 4.01 (d, J=6.14 Hz, 1H), 3.83 (brs, 2H), 2.98 (brs, 2H), 2.66 (d, J=5.70 Hz, 2H), 2.59 (s, 6H), 1.42 (s, 9H), 1.30 (d, J=7.02 Hz, 6H).
  • Example S80. Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-6-((1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-2-isopropyl-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02446
    Figure US20230109104A1-20230406-C02447
  • Step-1: Synthesis of 2-(3-nitrophenyl) ethan-1-amine: To a stirred solution of 3-nitrophenylacetonitril (5.0 g, 30.83 mmol, 1 eq), (3.22 mL, 1.5 eq) of borane-dimethyl sulfide complex was added to THF (100 mL) and stirred under reflux for 2 h. The reaction mixture was cooled to room temperature, and 40 mL of 5% HCl/methanol solution was added to it, and heated under reflux for 1 h. The solvent was evaporated, and diethyl ether was added to it to obtain, 2-(3-nitrophenyl) ethan-1-amine (3.5 g, 68.35%) as yellowish solid.
  • LCMS: 166.1 [M+1]+
  • Step-2: Synthesis of tert-butyl (3-nitrophenethyl) carbamate: To a stirred solution of 2-(3-nitrophenyl) ethan-1-amine (3.5 g, 21.08 mmol, 1 eq) in THF (50 mL) were added, 5M sodium hydroxide (7 mL) and di-tert-butyl dicarboxylic anhydride (4.68 g, 21.08 mmol, 1 eq), and stirred overnight at room temperature. The reaction mixture was diluted with water (100 mL), extracted with EtOAc (200 mL×2). The combined organic layers were dried over Na2SO4, concentrated and purified by column chromatography (Combiflash, Elution: 0-70% EtOAc in Hexane) to afford the desired product, tert-butyl (3-nitrophenethyl) carbamate (4.0 g, 71.42%) as colorless oil.
  • LCMS: 266.1 [M+1]+
  • Step-3: Synthesis of tert-butyl (3-aminophenethyl) carbamate: To a stirred solution of tert-butyl (3-nitrophenethyl) carbamate (4.0 g, 15.03 mmol, 1 eq), in THF: Methanol (150 mL, 2:1 ratio), palladium hydroxide-carbon (1.21 g) was added and the reaction mixture was stirred under hydrogen atmosphere overnight. The catalyst was removed through filtration, and the filtrate was concentrated to obtain, tert-butyl (3-aminophenethyl) carbamate (3.0 g, 84.75%) as colorless liquid.
  • LCMS: 237.0 [M+1]+
  • Step-4: Synthesis of 3-(2-aminoethyl) aniline: A solution of tert-butyl (3-aminophenethyl) carbamate (3.0 g, 12.71 mmol, 1 eq) in TFA (10 mL) was stirred at room temperature for 1 h. The reaction liquid was concentrated to obtain, 3-(2-aminoethyl) aniline (2.5 g, crude) as brown liquid.
  • LCMS: 137.0 [M+1]+
  • Step-5: Synthesis of 1, 1-dimethyl-1, 2, 3, 4-tetrahydroisoquinolin-6-amine: A mixture of 3-(2-aminoethyl) aniline (2.5 g, 18.38 mmol, 1 eq), 6.5 mL of 85% phosphoric acid and 1.8 mL of 2, 2-dimethoxypropane was stirred at 70° C. for 3 days. Further, 1 mL of 2, 2-dimethoxypropane was added to it, and reacted at 140° C. for 3 h using a microwave reactor. The reaction mixture was diluted with water (50 mL), basified with K2CO3 and extracted with 10% methanol in dichloromethane (100 mL×2), dried with anhydrous Na2SO4, and the solvent was evaporated to obtain, 1, 1-dimethyl-1, 2, 3, 4-tetrahydroisoquinolin-6-amine (1.24 g, crude) as brown solid.
  • LCMS: 176.9 [M+1]+
  • Step-6: Synthesis of tert-butyl 6-amino-1,1-dimethyl-3, 4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1, 1-dimethyl-1, 2, 3, 4-tetrahydroisoquinolin-6-amine (1.24 g, 7.045 mmol, 1.0 eq) in (10.0 mL) of THF was added di-tert-butyl dicarboxylic anhydride (1.84 g, 8.45 mmol, 1.2 eq) and allowed to stir at rt for 12 h and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water (50 mL) and extracted with EtOAc (50 mL×2). The combined organic layers were washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-40% EtOAc in hexane] to afford the desired compound (0.7 g) as brown solid.
  • LCMS: 276.9 [M+1]+
  • Step-7: Synthesis of tert-butyl 6-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of tert-butyl 6-amino-1, 1-dimethyl-3, 4-dihydroisoquinoline-2(1H)-carboxylate (200 mg, 0.560 mmol, 1.0 eq) in (6.0 mL) of toluene was added m-CPBA (194 mg, 1.120 mmol, 2.0 eq) and allowed to stir at rt for 1 h. 1, 1-dimethyl-1, 2, 3, 4-tetrahydroisoquinolin-6-amine (186 mg, 0.672 mmol, 1.2 eq) and DIPEA (0.43 mL, 2.2408 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 6-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (50 mg, 15.25%) as an off white solid.
  • LCMS: 586.4 [M+1]+
  • Step-8: Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-6-((1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-2-isopropyl-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride: tert-butyl 6-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (50 mg, 0.0854 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (2 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(2-(tert-butyl)pyridin-4-yl)-6-((1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-2-isopropyl-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (21 mg, 44.05%) as light yellow solid.
  • LCMS: 486.4 [M+1]+; UPLC @ 254 nm=91.54% and @ 220 nm=93.66%.
  • 1H NMR (400 MHz, DMSO-ds): δ 10.48 (br. s., 1H), 9.75 (br. s., 1H), 8.87 (s, 1H) 8.58 (d, J=5.26 Hz, 1H) 7.90 (br. s., 1H) 7.58 (d, J=4.82 Hz, 2H) 7.48 (s, 1H) 7.19 (d, J=8.33 Hz, 1H) 3.94-3.99 (m, 1H) 3.35 (t, 2H) 3.03 (t, 2H), 1.62 (s, 6H), 1.42 (d, 6H), 1.37 (s, 9H).
  • Example S81. Synthesis of N-(4-(2-isopropyl-3-oxo-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)-N-methylmethanesulfonamide dihydrochloride
  • Figure US20230109104A1-20230406-C02448
  • Step-1: Synthesis of 4-bromo-N-methylpyridin-2-amine: To a stirred solution of 4-bromopyridin-2-amine (3.0 g, 17.34 mmol, 1 eq), in DMF (10 mL) and NaH (0.5 g, 19.07 mmol, 1 eq), was added at 0° C. & stirred for 30 min. Mel (1.62 mL, 26.01 mmol, 1.5 eq) was added dropwise & stirred for 1 h. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layers were washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, 4-bromo-N-methylpyridin-2-amine (600 mg, 18.51%) as white solid.
  • LCMS: 187.0 [M+1]+
  • Step-2: Synthesis of N-(4-bromopyridin-2-yl)-N-methylmethanesulfonamide: To a stirred solution of 4-bromo-N-methylpyridin-2-amine (0.6 g, 3.22 mmol, 1 eq), in (50 mL) of DCM, was added TEA (1 mL, 7.09 mmol, 2.2 eq), followed by addition of methane sulphonyl chloride (0.4 mL, 4.83 mmol, 1.5 eq) and stirred overnight at room temperature. The reaction mixture was basified with saturated sodium bicarbonate solution and extracted with EtOAc (50 mL×2). The combined organic layers were washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and the solvent was evaporated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, N-(4-bromopyridin-2-yl)-N-methylmethanesulfonamide (563 mg, 66.19%) as white solid.
  • LCMS: 265.0 [M+1]+
  • Step-3: Synthesis of N-(4-(2-isopropyl-6-(methylthio)-3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-1-yl) pyridin-2-yl)-N-methylmethanesulfonamide: To a stirred solution of 2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (400 mg, 1.77 mmol, 1.0 eq) and N-(4-bromopyridin-2-yl)-N-methylmethanesulfonamide (563 mg, 2.13 mmol, 1.2 eq) in (10 mL) of dioxane was added Potassium carbonate (490 mg, 3.55 mmol, 2 eq) and the resulting mixture was purged with nitrogen for 10 min, followed by addition of copper iodide (68 mg, 0.35 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (62 mg, 0.710 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 90° C. for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). Combined organic layers were washed with water (50 mL) brine solution (50 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography to afford the desired compound, N-(4-(2-isopropyl-6-(methylthio)-3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-1-yl) pyridin-2-yl)-N-methylmethanesulfonamide (120 mg, 16.47%) as an off white solid.
  • LCMS: 409.1 [M+1]+
  • Step-4: Synthesis of tert-butyl 6-((2-isopropyl-1-(2-(N-methylmethylsulfonamido)pyridin-4-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of N-(4-(2-isopropyl-6-(methylthio)-3-oxo-2, 3-dihydro-1H-pyrazolo [3, 4-d] pyrimidin-1-yl) pyridin-2-yl)-N-methylmethanesulfonamide (120 mg, 0.294 mmol, 1.0 eq) in (3.0 mL) of toluene was added m-CPBA (102 mg, 0.5882 mmol, 2.0 eq) and allowed to stir at rt for 1 h. tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (102 mg, 0.3529 mmol, 1.2 eq) and DIPEA (0.2 mL, 1.18 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 6-((2-isopropyl-1-(2-(N-methylmethylsulfonamido)pyridin-4-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (40 mg, 22.37%) as an off white solid.
  • LCMS: 609.3 [M+1]+
  • Step-5: Synthesis of N-(4-(2-isopropyl-3-oxo-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)-N-methylmethanesulfonamide dihydrochloride: tert-butyl 6-((2-isopropyl-1-(2-(N-methylmethylsulfonamido)pyridin-4-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (30 mg, 0.0495 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (2 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, N-(4-(2-isopropyl-3-oxo-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)-N-methylmethanesulfonamide dihydrochloride (11 mg, 38.39%) as light yellow solid.
  • LCMS: 509.3 [M+1]+; UPLC @ 254 nm=85.04% and @ 220 nm=87.09%.
  • 1H NMR (400 MHz, DMSO-ds): δ 10.48 (br. s., 1H), 9.17 (br. s., 1H), 8.87 (s, 1H) 8.58 (d, J=5.26 Hz, 1H) 7.74 (br. s., 1H) 7.58 (d, J=4.82 Hz, 2H) 7.48 (s, 1H) 7.19 (d, J=8.33 Hz, 1H) 4.22 (br. s., 2H) 3.94-3.99 (m, 1H) 3.35 (t, 2H) 3.25 (br. s., 3H) 3.17 (t, 2H) 3.03 (br. s., 3H) 1.37 (d, J=7.02 Hz, 6H).
  • Example S82. Synthesis of 1-(2-(2-hydroxypropan-2-yl)pyridin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02449
  • Step-1: Synthesis of tert-butyl 6-((1-(2-(2-hydroxypropan-2-yl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(2-(2-hydroxypropan-2-yl)pyridin-4-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (200 mg, 0.5571 mmol, 1.0 eq) in (3.0 mL) of toluene was added m-CPBA (270 mg, 1.1142 mmol, 2.0 eq) and allowed to stir at rt for 1 h. tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (166 mg, 0.6685 mmol, 1.2 eq) and DIPEA (0.4 mL, 2.2284 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 6-((1-(2-(2-hydroxypropan-2-yl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (110 mg, 35.32%) as an off white solid.
  • LCMS: 560.4[M+1]+
  • Step-2: Synthesis of 1-(2-(2-hydroxypropan-2-yl)pyridin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: tert-butyl 6-((1-(2-(2-hydroxypropan-2-yl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (50 mg, 0.089 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (1 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure and purified by prep purification to afford the desired compound, 1-(2-(2-hydroxypropan-2-yl)pyridin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (10 mg, 24.35%) as light yellow solid.
  • LCMS: 460.4 [M+1]+; UPLC @ 254 nm=97.95% and @ 220 nm=98.46%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.29 (br. s. 1H), 8.82 (s, 1H) 8.63 (d, J=5.26 Hz, 1H) 7.72 (br. s., 1H) 7.55 (d, J=4.38 Hz, 2H) 7.46 (br. s., 1H) 7.01 (d, J=7.45 Hz, 1H) 5.35 (br. s., 1H) 3.96 (d, J=6.14 Hz, 2H) 3.80 (t, 2H) 2.95 (br. s., 1H) 2.67 (t, 2H) 1.75 (s, 1H) 1.46 (s, 6H) 1.35 (d, J=6.58 Hz, 6H).
  • Example S83. Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-6-((1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02450
  • Step-1: Synthesis of tert-butyl 6-((1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (150 mg, 0.421 mmol, 1.0 eq) in (6.0 mL) of toluene was added m-CPBA (145 mg, 0.842 mmol, 2.0 eq) and allowed to stir at rt for 1 h. tert-butyl 6-amino-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (140 mg, 0.506 mmol, 1.2 eq) and DIPEA (0.29 mL, 1.684 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 6-((1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (60 mg, 24.35%) as an off white solid.
  • LCMS: 584.3 [M+1]+
  • Step-2: Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-6-((1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride: tert-butyl 6-((1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (60 mg, 0.102 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (2 ML) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-6-((1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (36 mg, 62.93%) as an off white solid.
  • LCMS: 486.4 [M+1]+; UPLC @ 254 nm=96.12% and @ 220 nm=97.81%.
  • 1H NMR (400 MHz, DMSO-d6): δδ 10.33 (brs, 1H), 9.34 (brs, 2H), 8.86 (brs, 1H), 8.73 (brs, 1H), 7.58 (brs, 3H), 7.36 (d, J=7.45 Hz, 1H), 3.17 (brs, 2H), 3.04 (brs, 3H), 1.65 (s, 6H), 1.38 (s, 9H), 0.84 (brs, 4H)
  • Example S84. Synthesis of 1-(2-(tert-butyl)pyrimidin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride
  • Figure US20230109104A1-20230406-C02451
  • Step 1: Synthesis of 1-(2-(tert-butyl)pyrimidin-4-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (417 mg, 1.85 mmol, 1.0 eq.) and 4-bromo-2-(tert-butyl)pyrimidine (400 mg, 1.85 mmol, 1.2 eq.) in 30 mL of dioxane were added copper iodide (71 mg, 0.37 mmol, 0.2 eq.), Potassium carbonate (514 mg, 3.71 mmol, 2 eq.) and DMEDA (0.08 mL, 0.74 mmol, 0.4 eq.) and stirred at 110° C. overnight. After completion of reaction, solvent was removed under reduced pressure; residue was diluted with water and extracted with ethyl acetate (100 mL×3). The combined organic layer were washed with brine solution, dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography to afford 1-(2-(tert-butyl)pyrimidin-4-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (352 mg, 52.81%) as an off white solid.
  • LCMS: 359.2 [M+1]+
  • Step 2: Synthesis of tert-butyl 6-((1-(2-(tert-butyl)pyrimidin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(2-(tert-butyl)pyrimidin-4-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (176 mg, 0.49 mmol, 1.0 eq.) in 3 mL of toluene was added m-CPBA (169 mg, 0.98 mmol, 2 eq.) and allowed to stir at rt for 1 h. Further, tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (146 mg, 0.58 mmol, 1.2 eq.) and DIPEA (0.33 mL, 1.96 mmol, 4 eq.) were added and allowed to stir at rt for overnight. After completion of reaction, solvent was removed under reduced pressure; residue was diluted with water and extracted with ethyl acetate (50 mL×3). The combined organic layer was washed with brine solution (10 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product which was purified by flash chromatography to afford tert-butyl 6-((1-(2-(tert-butyl)pyrimidin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (120 mg, 43.74%) as yellow solid.
  • LCMS: 559.3 [M+1]+
  • Step 3: Synthesis of 1-(2-(tert-butyl)pyrimidin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride: tert-butyl 7-((1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-4,4-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (120 mg, 0.214 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (2 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(2-(tert-butyl)pyrimidin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride (64 mg, 60.19%) as an off white solid.
  • LCMS: 459.4 [M+1]+; UPLC @ 254 nm=96.17% and @ 220 nm=95.77%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.48 (brs, 1H), 9.48 (brs, 2H), 8.84-8.95 (m, 2H), 7.99 (brs, 1H), 7.72 (brs, 1H), 7.47 (brs, 1H), 7.23 (d, J=8.33 Hz, 1H), 4.20-4.31 (m, 3H), 3.39 (brs, 2H), 3.06 (brs, 2H), 1.39-1.55 (m, 6H), 1.38 (s, 9H).
  • Example S85. Synthesis of 1-(2-(tert-butyl)pyrimidin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride
  • Figure US20230109104A1-20230406-C02452
  • Step 1: Synthesis of 1-(2-(tert-butyl)pyrimidin-4-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (417 mg, 1.85 mmol, 1.0 eq.) and 4-bromo-2-(tert-butyl)pyrimidine (400 mg, 1.85 mmol, 1.2 eq.) in 30 mL of dioxane were added copper iodide (71 mg, 0.37 mmol, 0.2 eq.), Potassium carbonate (514 mg, 3.71 mmol, 2 eq.) and DMEDA (0.08 mL, 0.74 mmol, 0.4 eq.) and stirred at 110° C. overnight. After completion of reaction, solvent was removed under reduced pressure; residue was diluted with water and extracted with ethyl acetate (100 mL×3). The combined organic layer were washed with brine solution, dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography to afford 1-(2-(tert-butyl)pyrimidin-4-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (352 mg, 52.81%) as an off white solid.
  • LCMS: 359.2 [M+1]+
  • Step 2: Synthesis of tert-butyl 7-((1-(2-(tert-butyl)pyrimidin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(2-(tert-butyl)pyrimidin-4-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (176 mg, 0.49 mmol, 1.0 eq.) in 3 mL of toluene was added m-CPBA (169 mg, 0.98 mmol, 2 eq.) and allowed to stir at rt for 1 h. Further, tert-butyl 7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (146 mg, 0.58 mmol, 1.2 eq.) and DIPEA (0.33 mL, 1.96 mmol, 4 eq.) were added and allowed to stir at rt for overnight. After completion of reaction, solvent was removed under reduced pressure; residue was diluted with water and extracted with ethyl acetate (50 mL×3). The combined organic layer was washed with brine solution (10 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product which was purified by flash chromatography to afford tert-butyl 7-((1-(2-(tert-butyl)pyrimidin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (120 mg, 43.74%) as yellow solid.
  • LCMS: 559.3 [M+1]+
  • Step 3: Synthesis of 1-(2-(tert-butyl)pyrimidin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride: tert-butyl 7-((1-(2-(tert-butyl)pyrimidin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (120 mg, 0.214 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (2 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(2-(tert-butyl)pyrimidin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride (98 mg, 92.17%) as an off white solid.
  • LCMS: 459.4 [M+1]+; UPLC @ 254 nm=95.93% and @ 220 nm=96.06%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.49 (brs, 1H), 9.19 (brs, 2H), 8.84-8.95 (m, 2H), 8.00 (brs, 1H), 7.71 (brs, 1H), 7.47 (brs, 1H), 7.24 (d, J=8.33 Hz, 1H), 4.18-4.39 (m, 3H), 3.40 (brs, 3H), 2.99 (brs, 2H), 1.47 (d, J=7.02 Hz, 6H), 1.38 (s, 9H).
  • Example S86 Synthesis of 1-(4-fluoro-3-(2-hydroxypropan-2-yl)phenyl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02453
  • Step-1: Synthesis of 1-(5-bromo-2-fluorophenyl)ethan-1-ol: To a stirred solution of 5-bromo-2-fluorobenzaldehyde (5.0 g, 24.63 mmol, 1.0 eq) in diethylether (50 mL) was dropwise added methylmagnesium bromide (12.3 mL, 36.94 mmol, 1.5 eq) at 0° C. The resulting mixture was stirred at 0° C. for 15 min. The progress of reaction was monitored by LCMS. The resection mixture was quenched with saturated solution of NH4Cl (50 mL) extracted with EtOAc (2×50 mL). The combined organic extracts were washed with water (50 mL), with brine (50 mL) dried over Na2SO4 and concentrated under reduced to afford the desired compound 1-(5-bromo-2-fluorophenyl)ethan-1-ol (5.31 g, 98.51%) as yellow liquid.
  • 1H NMR (400 MHz, CDCl3) 7.64 (dd, J=2.41, 6.36 Hz, 1H), 7.35 (ddd, J=2.63, 4.60, 8.55 Hz, 1H), 6.91 (dd, J=8.77, 9.65 Hz, 1H), 5.16 (d, J=5.26 Hz, 1H), 1.88 (brs, 1H), 1.50 (d, J=6.14 Hz, 3H).
  • Step-2: Synthesis of 1-(5-bromo-2-fluorophenyl)ethan-1-one: To a stirred solution of 1-(5-bromo-2-fluorophenyl)ethan-1-ol (5.3 g, 24.195 mmol, 1.0 eq) in THF (50 mL) was added Dess-martin peridionane (12.314 g, 29.034 mmol, 1.2 eq) at 0° C. The resulting mixture was stirred at rt for 15 min. The progress of reaction was monitored by LCMS. The resection mixture was quenched with saturated solution of NaHCO3 (50 mL) extracted with EtOAc (2×50 mL). The combined organic extracts were washed with water (50 mL), with brine (50 mL) dried over Na2SO4 and concentrated under reduced pressure and purified by flash chromatography [silica gel 100-200 mesh; elution 0-5% EtOAc in hexane] to afford the desired compound 1-(5-bromo-2-fluorophenyl)ethan-1-one (3.78 g, 72.00%) as yellow liquid.
  • 1H NMR (400 MHz, CDCl3) δ 7.99 (dd, J=2.63, 6.58 Hz, 1H), 7.61 (ddd, J=2.63, 4.39, 8.77 Hz, 1H), 7.05 (dd, J=8.77, 10.09 Hz, 1H), 2.64 (d, J=4.82 Hz, 3H).
  • Step-3: Synthesis of 2-(5-bromo-2-fluorophenyl)propan-2-ol: To a stirred solution of 1-(5-bromo-2-fluorophenyl)ethan-1-one (3.70 g, 17.047 mmol, 1.0 eq) in diethylether (30 mL) was dropwise added methylmagnesium bromide (8.53 mL, 25.571 mmol, 1.5 eq) at 0° C. The resulting mixture was stirred at 0° C. for 15 min. The progress of reaction was monitored by LCMS. The resection mixture was quenched with saturated solution of NH4Cl (50 mL) extracted with EtOAc (2×50 mL). The combined organic extracts were washed with water (50 mL), with brine (50 mL) dried over Na2SO4 and concentrated under reduced pressure and purified by flash chromatography [silica gel 100-200 mesh; elution 0-5% EtOAc in hexane] to afford the desired compound 2-(5-bromo-2-fluorophenyl)propan-2-ol (3.2 g, 93.02%) as yellow liquid.
  • 1H NMR (400 MHz, CDCl3) δ 7.73 (dd, J=2.63, 7.45 Hz, 1H), 7.34 (ddd, J=2.63, 4.38, 8.77 Hz, 1H), 6.91 (dd, J=8.33, 11.40 Hz, 1H), 2.02 (d, J=17.54 Hz, 1H), 1.61-1.64 (m, 6H).
  • Step-4: Synthesis of 2-(2-fluoro-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)propan-2-ol: To a stirred solution of 2-(5-bromo-2-fluorophenyl)propan-2-ol (1.0 g, 4.290 mmol, 1.0 eq) and 4,4,4′,4′,5,5,5′,5′-octamethyl-2,2′-bi(1,3,2-dioxaborolane) (1.30 g, 5.148 mmol, 1.2 eq) in dioxane (30 mL) was added KoAc (1.05 g, 10.725 mmol, 2.5 eq) at rt. The resulting mixture was purged with N2 for 10 min. followed by addition of Pd(dppf)Cl2.DCM (175 mg, 0.214 mmol, 0.05 eq) and again purged with N2 for 10 min. The resulting mixture was stirred at 70° C. for overnight. The progress of reaction was monitored by LCMS. The resection mixture was diluted with water (50 mL) extracted with EtOAc (2×50 mL). The combined organic extracts were washed with water (50 mL), with brine (50 mL) dried over Na2SO4 and concentrated under reduced pressure and purified by flash chromatography [silica gel 100-200 mesh; elution 0-5% EtOAc in hexane] to afford the desired compound 2-(2-fluoro-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)propan-2-ol (705 mg, 58.75%) as yellow viscous.
  • 1H NMR (400 MHz, CDCl3) δ 7.98 (dd, J=1.53, 8.99 Hz, 1H), 7.64-7.74 (m, 1H), 7.02 (dd, J=8.11, 12.50 Hz, 1H), 2.14 (brs, 1H), 1.61-1.70 (m, 6H), 1.34 (s, 12H).
  • Step-5: Synthesis of 1-(4-fluoro-3-(2-hydroxypropan-2-yl)phenyl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (280 mg, 1.249 mmol, 1.0 eq) and 2-(2-fluoro-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)propan-2-ol (700 mg, 2.498 mmol, 2.0 eq) in (50 mL) of CH2Cl2 was added Na2CO3 (397 mg, 3.747 mmol, 3.0 eq), Cu(OAc)2 (442 mg, 2.498 mmol, 2.0 eq), Py2 (390 mg, 2.498 mmol, 2.0 eq), and the resulting mixture was stirred at rt for 48 h. After completion of reaction, the reaction mixture was diluted with water (50.0 mL) and extracted with EtOAc (50 mL×2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound 1-(4-fluoro-3-(2-hydroxypropan-2-yl)phenyl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (220 mg, 46.80%) as light yellow viscous.
  • LCMS: 377.2 [M+1]+
  • Step-6: Synthesis of tert-butyl 6-((1-(4-fluoro-3-(2-hydroxypropan-2-yl)phenyl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydro isoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(4-fluoro-3-(2-hydroxypropan-2-yl)phenyl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (100 mg, 0.265 mmol, 1.0 eq) in (2.0 mL) of toluene was added mCPBA (130 mg, 0.530 mmol, 2.0 eq) and allowed to stir at rt for 30 minutes. tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (80 mg, 0.318 mmol, 1.2 eq) and DIPEA (0.180 mL, 1.06 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (20 mL×2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-60% EtOAc in hexane] to afford the desired compound tert-butyl 6-((1-(4-fluoro-3-(2-hydroxypropan-2-yl)phenyl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydro isoquinoline-2(1H)-carboxylate (60 mg, 39.21%) as brown viscous.
  • LCMS: 577.4 [M+1]+
  • Step-7: Synthesis of 1-(4-fluoro-3-(2-hydroxypropan-2-yl)phenyl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: tert-butyl 6-((1-(4-fluoro-3-(2-hydroxypropan-2-yl)phenyl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydro isoquinoline-2(1H)-carboxylate (60 mg, 1.00 mmol, 1.0 eq) was dissolved in 4.0 M-HCl (1.5 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was basified with NaHCO3 water and extracted with EtOAc (20 mL×2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by reverse phase chromatography to afford the desired compound 1-(4-fluoro-3-(2-hydroxypropan-2-yl)phenyl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (7.0 mg, 14.28%) as white solid.
  • LCMS: 477.4 [M+1]+; UPLC @ 254 nm=95.78% and @ 220 nm=96.10%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.26 (brs, 1H), 9.10 (brs, 2H), 8.84 (s, 1H), 7.66 (brs, 1H), 7.60 (brs, 1H), 7.54 (d, J=7.02 Hz, 2H), 7.28-7.38 (m, 1H), 7.12 (d, J=8.77 Hz, 1H), 5.47 (brs, 1H), 4.19 (brs, 2H), 4.05-4.13 (m, 1H), 2.92 (brs, 2H), 1.50 (s, 6H), 1.26 (d, J=6.58 Hz, 6H).
  • Example S87. Synthesis of 1-(4-fluoro-3-(2-hydroxypropan-2-yl)phenyl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02454
  • Step-1: Synthesis of tert-butyl 7-((1-(4-fluoro-3-(2-hydroxypropan-2-yl)phenyl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydro isoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(4-fluoro-3-(2-hydroxypropan-2-yl)phenyl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (100 mg, 0.265 mmol, 1.0 eq) in (2.0 mL) of toluene was added mCPBA (130 mg, 0.530 mmol, 2.0 eq) and allowed to stir at rt for 30 minutes. tert-butyl 7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (80 mg, 0.318 mmol, 1.2 eq) and DIPEA (0.180 mL, 1.06 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (20 mL×2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-60% EtOAc in hexane] to afford the desired compound tert-butyl 7-((1-(4-fluoro-3-(2-hydroxypropan-2-yl)phenyl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydro isoquinoline-2(1H)-carboxylate (40 mg, 26.14%) as brown viscous.
  • LCMS: 577.4 [M+1]+
  • Step-2: Synthesis of 1-(4-fluoro-3-(2-hydroxypropan-2-yl)phenyl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: tert-butyl 7-((1-(4-fluoro-3-(2-hydroxypropan-2-yl)phenyl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydro isoquinoline-2(1H)-carboxylate (40 mg, 0.069 mmol, 1.0 eq) was dissolved in 4.0 M-HCl (1.5 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was basified with NaHCO3 water and extracted with EtOAc (20 mL×2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by reverse phase chromatography to afford the desired compound 1-(4-fluoro-3-(2-hydroxypropan-2-yl)phenyl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (9.0 mg, 27.27%) as white solid.
  • LCMS: 477.4 [M+1]+; UPLC @ 254 nm=98.89% and @ 220 nm=98.54%.
  • 1H NMR (400 MHz, DMSO-d6) δ 10.28 (brs, 1H), 9.12 (brs, 2H), 8.83 (s, 1H), 7.66 (brs, 1H), 7.55 (s, 1H), 7.58 (s, 2H), 7.36 (d, J=10.96 Hz, 1H), 7.14 (d, J=7.89 Hz, 2H), 5.48 (brs, 1H), 4.21 (brs, 2H), 4.05-4.11 (m, 1H), 2.93 (brs, 2H), 1.51 (s, 6H), 1.26 (d, J=6.58 Hz, 6H).
  • Example S88. Synthesis of 6-((1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-2-isopropyl-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02455
  • Step-1: Synthesis of tert-butyl 6-((1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (95 mg, 0.262 mmol, 1.0 eq) in (6.0 mL) of toluene was added m-CPBA (90 mg, 0.524 mmol, 2.0 eq) and allowed to stir at rt for 1 h. tert-butyl 6-amino-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (87 mg, 0.315 mmol, 1.2 eq) and DIPEA (0.18 mL, 1.048 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 6-((1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (60 mg, 38.70%) as an off white solid.
  • LCMS: 590.3 [M+1]+
  • Step-2: Synthesis of 6-((1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-2-isopropyl-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride: tert-butyl 6-((1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (60 mg, 0.102 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (2 ML) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 6-((1, 1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-2-isopropyl-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (36 mg, 62.90%) as an off white solid.
  • LCMS: 490.3 [M+1]+; UPLC @ 254 nm=96.29% and @ 220 nm=96.60%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.47 (brs, 1H), 9.53 (brs, 2H), 8.87 (s, 1H), 8.74 (d, J=5.70 Hz, 1H), 7.63 (d, J=7.02 Hz, 2H), 7.53 (brs, 1H), 7.34 (d, J=8.77 Hz, 1H), 3.41 (brs, 2H), 3.05 (br., 2H), 1.72 (s, 3H), 1.53-1.67 (m, 9H), 1.36 (d, J=6.58 Hz, 4H).
  • Example S89. Synthesis of 1-(1-cyclopropyl-1H-pyrazol-3-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride
  • Figure US20230109104A1-20230406-C02456
  • Step-1: Synthesis of 1-(1-cyclopropyl-1H-pyrazol-3-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (1.0 g, 4.45 mmol, 1.0 eq) and 3-bromo-1-cyclopropyl-1H-pyrazole (1.0 g, 5.34 mmol, 1.2 eq) in (30 mL) of dioxane was added Potassium carbonate (1.23 g, 8.9 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min, followed by addition of copper iodide (0.17 g, 0.89 mmol, 0.2 eq) and N,N′-dimethylethylenediamine (DMEDA) (0.20 mL, 1.78 mmol, 2 eq) and again purged with nitrogen for 10 min, stirred at 130° C. for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layers were washed with water (30 mL), brine solution (30 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography to afford the desired compound, 1-(1-cyclopropyl-1H-pyrazol-3-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (180 mg, 12.24%) as an off white solid.
  • LCMS: 331.1 [M+1]+
  • Step-2: synthesis of tert-butyl 6-((1-(1-cyclopropyl-1H-pyrazol-3-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(1-cyclopropyl-1H-pyrazol-3-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (180 mg, 0.545 mmol, 1.0 eq) in (2 mL) of toluene was added m-CPBA (188 mg, 1.09 mmol, 2.0 eq) and allowed to stir at rt for 1 h. Tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (162.4 mg, 0.654 mmol, 1.2 eq) and DIPEA (0.377 mL, 2.18 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. The reaction mixture was diluted with water (30 mL) and extracted with EtOAc (30 mL×2). The combined organic layer were dried over sodium sulphate, concentrated under reduced pressure purified by column chromatography (Combiflash, elution-0-60% EtOAc in Hexane) to afford the desired compound, tert-butyl 6-((1-(1-cyclopropyl-1H-pyrazol-3-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (95 mg, 32.86%) as yellow liquid.
  • LCMS: 531.4 [M+1]+
  • Step-3: Synthesis of 1-(1-cyclopropyl-1H-pyrazol-3-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride: To a stirred solution of tert-butyl 6-((1-(1-cyclopropyl-1H-pyrazol-3-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (95 mg, 0.17 mmol, 1.0 eq) in (1 mL) of dioxane was added 4M dioxane-HCl (1 mL) and allowed to stir at rt for 2 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(1-cyclopropyl-1H-pyrazol-3-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride (64 mg, 76.55%) as light yellow solid.
  • LCMS: 431.2 [M+1]t, UPLC @ 254 nm=97.16% and @ 220 nm=98.13%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.23 (brs, 1H), 9.30 (brs, 2H), 8.81 (s, 1H), 7.97 (d, J=2.19 Hz, 1H), 7.65 (brs, 1H), 7.51 (d, J=7.89 Hz, 1H), 7.11 (d, J=8.77 Hz, 1H), 6.46 (d, J=2.19 Hz, 1H), 4.24-4.33 (m, 1H), 4.18 (brs, 2H), 3.81 (dd, J=3.51, 7.45 Hz, 1H), 3.34 (brs, 2H), 2.88-2.97 (m, 2H), 1.24 (d, J=6.58 Hz, 6H), 1.00-1.09 (m, 4H).
  • Example S90. Synthesis of 1-(1-cyclopropyl-JH-pyrazol-3-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02457
  • Step-1: synthesis of tert-butyl 7-((1-(1-cyclopropyl-1H-pyrazol-3-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(1-cyclopropyl-1H-pyrazol-3-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (85 mg, 0.257 mmol, 1.0 eq) in (1.5 mL) of toluene was added m-CPBA (88.70 mg, 0.514 mmol, 2.0 eq) and allowed to stir at rt for 1 h. Tert-butyl 7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (76.6 mg, 0.308 mmol, 1.2 eq) and DIPEA (0.17 mL, 2.18 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. The reaction mixture was diluted with water (30 mL) and extracted with EtOAc (30 mL×2). The combined organic layer were dried over sodium sulphate, concentrated under reduced pressure purified by column chromatography (Combiflash, elution-0-60% EtOAc in Hexane) to afford the desired compound, tert-butyl 7-((1-(1-cyclopropyl-1H-pyrazol-3-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (36 mg, 26.37%) as yellow liquid.
  • LCMS: 531.4 [M+1]+
  • Step-2: Synthesis of 1-(1-cyclopropyl-1H-pyrazol-3-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of tert-butyl 7-((1-(1-cyclopropyl-1H-pyrazol-3-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (36 mg, 0.067 mmol, 1.0 eq) in (0.6 mL) of dioxane was added 4M dioxane-HCl (0.6 mL) and allowed to stir at rt for 2 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, of 1-(1-cyclopropyl-1H-pyrazol-3-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (18 mg, 58.59%) as light yellow solid.
  • LCMS: 431.2 [M+1]+, UPLC @ 254 nm=95.48% and @ 220 nm=96.05%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.23 (brs, 1H), 9.23 (brs, 2H), 8.81 (s, 1H), 7.95 (d, J=2.63 Hz, 1H), 7.63 (brs, 1H), 7.53 (d, J=8.33 Hz, 1H), 7.12 (d, J=7.89 Hz, 1H), 6.46 (d, J=2.19 Hz, 1H), 4.22-4.31 (m, 1H), 4.19 (brs, 2H), 3.74-3.87 (m, 2H), 3.35 (brs, 2H), 2.93 (t, J=6.14 Hz, 2H), 1.24 (d, J=6.58 Hz, 6H), 0.93-1.10 (m, 4H).
  • Example S91. Synthesis of 2-isopropyl-1-(1-methyl-JH-pyrazol-3-yl)-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride
  • Figure US20230109104A1-20230406-C02458
  • Step-1: Synthesis of (1-methyl-1H-pyrazol-3-yl)boronic acid: To a stirred solution of 3-bromo-1-methyl-1H-pyrazole (10 g, 62.11 mmol, 1 eq.) and 4,4,4′4′,5,5,5′,5′-octamethyl-2,2′-bi(1,3,2-dioxaborolane) (23.6 g, 93.167 mmol, 1.5 eq.) in 120 mL of dioxane was added potassium acetate (18.28 g, 186.33 mmol, 3.0 eq.). The reaction mixture was purged with N2 for about 5 min and Pd(dppf)Cl2-DCM complex (2.5 g, 3.105 mmol, 0.05 eq.) was added. The reaction mixture was re-purged with N2 and heated at 100° C. for 16 h. Following this reaction mixture was allowed to cool to rt and filtered through celite bed and washed with ethyl acetate (200 mL). The obtained organic layer was concentrated under reduced pressure to get desired product, (1-methyl-1H-pyrazol-3-yl)boronic acid (6.5 g, 89.28%) as an off white solid.
  • LCMS: 127.0 [M+1]+
  • Step-2: Synthesis of 2-isopropyl-1-(1-methyl-1H-pyrazol-3-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (300 mg, 1.337 mmol, 1 eq.) and (1-methyl-1H-pyrazol-3-yl)boronic acid (340 mg, 2.675 mmol, 2.0 eq) in DCM (10 mL) was added 2,2-bipyridine (417 mg, 2.675 mmol, 2.0 eq.), copper acetate (474 mg, 2.675 mmol, 2.0 eq.) and Na2CO3 (425 mg, 4.012 mmol, 3.0 eq). The reaction mixture was stirred at rt for 24 h in open air. The progress of reaction was monitored by TLC. The reaction mass was diluted with water (200 mL) and extracted with ethyl acetate (200 mL×2) and the organic layer was dried over Na2SO4, concentrated and purified by column chromatography (Combiflash, Elution: 0-30% EtOAc in Hexane) to afford the desired product, 2-isopropyl-1-(1-methyl-1H-pyrazol-3-yl)-6-(methylthio)-1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (100 mg, 24.6%) as brown solid.
  • LCMS: 305.2 [M+1]+
  • Step-3: Synthesis of tert-butyl 6-((2-isopropyl-1-(1-methyl-1H-pyrazol-3-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 2-isopropyl-1-(1-methyl-1H-pyrazol-3-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (100 mg, 0.328 mmol, 1.0 eq) in (6 mL) of toluene was added m-CPBA (113 mg, 0.657 mmol, 2.0 eq) and allowed to stir at rt for 1 h. Tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (98 mg, 0.394 mmol, 1.2 eq) and DIPEA (0.23 mL, 1.315 mmol, 4.0 eq) were added and allowed to stir at 80° C. for overnight. Toluene was evaporated and the reaction mass was diluted with water (200 mL) and extracted with ethyl acetate (200 mL×2). The organic layer was dried over Na2SO4, concentrated and purified by column chromatography (Combiflash, elution-0-70% EtOAc in Hexane) to afford the desired compound, tert-butyl 6-((2-isopropyl-1-(1-methyl-1H-pyrazol-3-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (33 mg, 19.95%) as brown solid.
  • LCMS: 504.26 [M+1]+
  • Step-4: Synthesis of 2-isopropyl-1-(1-methyl-1H-pyrazol-3-yl)-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride: To a stirred solution of tert-butyl 6-((2-isopropyl-1-(1-methyl-1H-pyrazol-3-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (33 mg, 65.476 mmol, 1.0 eq) was dissolved in (1 mL) of dioxane and added 4M dioxane-HCl (0.6 mL) and allowed to stir at rt for 3 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 2-isopropyl-1-(1-methyl-1H-pyrazol-3-yl)-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride (10 mg, 37.80%) as an off white solid.
  • LCMS: 404.26 [M+1]+, UPLC @ 254 nm=96.95% and @ 220 nm=97.27%.
  • 1H NMR (400 MHz, DMSO-d6): δ ppm 10.22 (br. s., 1H), 9.14 (br. s., 1H), 8.82 (s, 1H), 7.86 (s, 1H), 7.67 (br. s., 1H), 7.50 (d, J=7.89 Hz, 1H), 7.11 (d, J=8.77 Hz, 1H), 6.45 (d, J=2.19 Hz, 1H), 4.27 (m, 1H), 4.19 (d, 2H), 3.91 (s, 3H), 3.40 (d, 2H), 2.92 (d, 2H) 1.26 (d, J=6.58 Hz, 6H).
  • Example S92. Synthesis of 1-(5-fluoropyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02459
  • Step-1: Synthesis of 1-(5-fluoropyridin-2-yl)-2-isopropyl-6-(methylthio)-1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one: To a stirred solution of 2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one, (300 mg, 1.337 mmol, 1.0 eq) and 2-bromo-5-fluoropyridine (282 mg, 1.605 mmol, 1.2 eq) in (6 mL) of dioxane was added Potassium carbonate (370 mg, 2.675 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min, followed by addition of copper iodide (51 mg, 0.267 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (0.06 mL, 0.535 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 130° C. for overnight. After completion of reaction, the reaction mixture was diluted with water (50 mL) and extracted with EtOAc (30 mL×2). The combined organic layers were washed with water (30 mL), brine solution (30 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography to afford the desired compound, 1-(5-fluoropyridin-2-yl)-2-isopropyl-6-(methylthio)-1, 2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (265 mg, 62.10%) as an sticky yellow liquid.
  • LCMS: 320.10 [M+1]+
  • Step-2: Synthesis of tert-butyl 6-((1-(5-fluoropyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(5-fluoropyridin-2-yl)-2-isopropyl-6-(methylthio)-1, 2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (130 mg, 0.407 mmol, 1.0 eq) in (6 mL) of toluene was added m-CPBA (141 mg, 0.814 mmol, 2.0 eq) and allowed to stir at rt for 1 h. tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (121.4 mg, 0.489 mmol, 1.2 eq) and DIPEA (0.3 mL, 1.628 mmol, 4.0 eq) were added and allowed to stir at 80° C. for overnight. Toluene was evaporated and reaction mixture was diluted with water (50 mL) and extracted with EtOAc (30 mL×2), the organic layer was dried over sodium sulphate, concentrated under reduced pressure purified by column chromatography (Combiflash, elution-0-70% EtOAc in Hexane) to afford the desired compound. tert-butyl 6-((1-(5-fluoropyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (100 mg, 47.31%) as yellow semi solid.
  • LCMS: 520.24 [M+1]+
  • Step-3: Synthesis of 1-(5-fluoropyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride: To a stirred solution of tert-butyl 6-((1-(5-fluoropyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (100 mg, 0.192 mmol, 1.0 eq) in (1 mL) of dioxane and added 4M dioxane-HCl (0.8 mL) and allowed to stir at rt for 3 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(5-fluoropyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (63 mg, 78.3%) as an off white solid.
  • LCMS: 420.3 [M+1]+, UPLC @ 254 nm=95.96% and @ 220 nm=96.53%.
  • 1H NMR (400 MHz, DMSO-d6): δ ppm 10.32 (br. s., 1H) 9.23 (br. s., 1H) 8.85 (s, 1H) 8.62 (d, J=2.63 Hz, 2H) 8.08 (d, J=8.55 Hz, 1H) 7.45 (d, J=7.89 Hz, 1H) 7.14 (d, J=8.33 Hz, 2H) 4.09-4.30 (m, 1H), 3.48 (s, 2H) 3.36 (d, 2H) 2.96 (d, 2H) 1.16-1.32 (m, 6H).
  • Example S93. Synthesis of 1-(5-fluoropyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02460
  • Step-1: Synthesis of 1-(5-fluoropyridin-2-yl)-2-isopropyl-6-(methylthio)-1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one: To a stirred solution of 2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one, (300 mg, 1.337 mmol, 1.0 eq) and 2-bromo-5-fluoropyridine (282 mg, 1.605 mmol, 1.2 eq) in (6 mL) of dioxane was added Potassium carbonate (370 mg, 2.675 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min, followed by addition of copper iodide (51 mg, 0.267 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (0.06 mL, 0.535 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 130° C. for overnight. After completion of reaction, the reaction mixture was diluted with water (30 mL) and extracted with EtOAc (30 mL×2). The combined organic layers were washed with water (30 mL), brine solution (30 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography to afford the desired compound, 1-(5-fluoropyridin-2-yl)-2-isopropyl-6-(methylthio)-1, 2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (265 mg, 62.10%) as an sticky yellow liquid.
  • LCMS: 320.10 [M+1]+
  • Step-2: Synthesis of tert-butyl 7-((1-(5-fluoropyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(5-fluoropyridin-2-yl)-2-isopropyl-6-(methylthio)-1, 2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (130 mg, 0.407 mmol, 1.0 eq) in (6 mL) of toluene was added m-CPBA (141 mg, 0.814 mmol, 2.0 eq) and allowed to stir at rt for 30 min. tert-butyl 7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (121.4 mg, 0.489 mmol, 1.2 eq) and DIPEA (0.3 mL, 1.628 mmol, 4.0 eq) were added and allowed to stir at 80° C. for overnight. Toluene was evaporated and reaction mixture was diluted with water (30 mL) and extracted with EtOAc (30 mL×2), the organic layer was dried over sodium sulphate, concentrated under reduced pressure purified by column chromatography (Combiflash, elution-0-70% EtOAc in Hexane) to afford the desired compound. tert-butyl 7-((1-(5-fluoropyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate. (70 mg, 33.12%) as yellow semi solid.
  • LCMS: 520.24 [M+1]+
  • Step-3: Synthesis of 1-(5-fluoropyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride: To a stirred solution of tert-butyl 7-((1-(5-fluoropyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate. (70 mg, 0.134 mmol, 1.0 eq) was dissolved in (3 mL) of dioxane and added 4M dioxane-HCl (0.9 mL) and allowed to stir at rt for 3 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(5-fluoropyridin-2-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (33 mg, 58.7%).
  • LCMS: 420.3 [M+1]+, UPLC @ 254 nm=95.51% and @ 220 nm=94.28%.
  • 1H NMR (400 MHz, DMSO-d6): δ ppm 10.32 (br. s., 1H) 9.23 (br. s., 1H) 8.85 (s, 1H) 8.62 (d, J=2.63 Hz, 2H) 8.08 (d, J=8.55 Hz, 1H) 7.45 (d, J=7.89 Hz, 1H) 7.14 (d, J=8.33 Hz, 2H) 4.09-4.30 (m, 1H), 3.48 (s, 2H) 3.36 (d, 2H) 2.96 (d, 2H) 1.16-1.32 (m, 6H).
  • Example S94. Synthesis of 2-isopropyl-1-(1-methyl-1H-pyrazol-3-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride
  • Figure US20230109104A1-20230406-C02461
  • Step-1: synthesis of tert-butyl 7-((2-isopropyl-1-(1-methyl-1H-pyrazol-3-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 2-isopropyl-1-(1-methyl-1H-pyrazol-3-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (100 mg, 0.328 mmol, 1.0 eq) in (6 mL) of toluene was added m-CPBA (113 mg, 0.657 mmol, 2.0 eq) and allowed to stir at rt for 30 min. tert-butyl 7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (98 mg, 0.394 mmol, 1.2 eq) and DIPEA (0.23 mL, 1.315 mmol, 4.0 eq) were added and allowed to stir at 80° C. for overnight. Tolene was evaporated and reaction mass was diluted with ethyl acetate and water, organic layer was dried over sodium sulphate, concentrated under reduced pressure purified by column chromatography (Combiflash, elution-0-70% EtOAc in Hexane) to afford the desired compound, tert-butyl 7-((2-isopropyl-1-(1-methyl-1H-pyrazol-3-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (35 mg, 21.16%) as brown solid.
  • LCMS: 504.26 [M+1]+
  • Step-2: Synthesis of (2-isopropyl-1-(1-methyl-1H-pyrazol-3-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride: To a stirred solution of tert-butyl 7-((2-isopropyl-1-(1-methyl-1H-pyrazol-3-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (35 mg 69.44 mmol, 1.0 eq) was dissolved in (1 mL) of dioxane and added 4M dioxane-HCl (0.6 mL) and allowed to stir at rt for 3 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 2-isopropyl-1-(1-methyl-1H-pyrazol-3-yl)-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride (15 mg, 53.47%) as an off white solid.
  • LCMS: 404.26 [M+1]+, UPLC @ 254 nm=98.84% and @ 220 nm=99.25%.
  • 1H NMR: (400 MHz, DMSO-d6): δ ppm 10.22 (br. s., 1H), 9.14 (br. s., 1H), 8.82 (s, 1H), 7.86 (s, 1H), 7.67 (br. s., 1H), 7.50 (d, J=7.89 Hz, 1H), 7.11 (d, J=8.77 Hz, 1H), 6.45 (d, J=2.19 Hz, 1H), 4.27 (m, 1H), 4.19 (d., 2H), 3.91 (s, 3H), 3.40 (d, 2H), 2.92 (d., 2H) 1.26 (d, J=6.58 Hz, 6H).
  • Example S95. Synthesis of 2-isopropyl-1-(2-(2-methoxypropan-2-yl)pyridin-4-yl)-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02462
  • Step-1: Synthesis of tert-butyl 6-((2-isopropyl-1-(2-(2-methoxypropan-2-yl)pyridin-4-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of tert-butyl 6-((1-(2-(2-hydroxypropan-2-yl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (50 mg, 0.0892 mmol, 1.0 eq) in (3.0 mL) of DMF was added K2CO3 (14 mg, 0.0982 mmol, 1.1 eq) and allowed to stir at 0° for 15 min. to that methyl iodide (0.1 mL, 0.0982 mmol, 1.1 eq) were added and allowed to stir at rt for 2 h. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-80% EtOAc in hexane] to afford the desired compound, tert-butyl 6-((1-(2-(2-hydroxypropan-2-yl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (35 mg, 68.62%) as an off white solid.
  • LCMS: 574.4 [M+1]+
  • Step-2: Synthesis of 2-isopropyl-1-(2-(2-methoxypropan-2-yl)pyridin-4-yl)-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: tert-butyl 6-((1-(2-(2-hydroxypropan-2-yl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (35 mg, 0.0609 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (1 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure and purified by prep purification to afford the desired compound, 2-isopropyl-1-(2-(2-methoxypropan-2-yl)pyridin-4-yl)-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (2 mg, 7.14%) as an off white solid.
  • LCMS: 474.4 [M+1]+; UPLC @ 254 nm=97.91% and @ 220 nm=97.81%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.29 (br. s. 1H), 8.82 (s, 1H) 8.63 (d, J=5.26 Hz, 1H) 7.72 (br. s., 1H) 7.55 (d, J=4.38 Hz, 2H) 7.46 (br. s., 1H) 7.01 (d, J=7.45 Hz, 1H) 3.96 (d, J=6.14 Hz, 2H) 3.80 (t, 2H), 3.45 (s, 3H), 2.95 (br. s., 1H) 2.67 (t, 2H) 1.75 (s, 1H) 1.46 (s, 6H) 1.35 (d, J=6.58 Hz, 6H).
  • Example S96 Synthesis of N-(6-(6-((1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)-N-methylmethanesulfonamide dihydrochloride
  • Figure US20230109104A1-20230406-C02463
  • Step-1: Synthesis of 6-bromo-N-methylpyridin-2-amine: To a stirred solution of 6-bromopyridin-2-amine (5.0 g, 28.90 mmol, 1 eq), in DMF (50 mL) and NaH (1.2 g, 31.79 mmol, 1.1 eq), was added at 0° C. & stirred for 30 min. Mel (2.7 mL, 43.35 mmol, 1.5 eq) was added dropwise & stirred for 1 h. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layers were washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, 6-bromo-N-methylpyridin-2-amine (2.0 g, 37.03%) as white solid.
  • LCMS: 187.0 [M+1]+
  • Step-2: Synthesis of N-(6-bromopyridin-2-yl)-N-methylmethanesulfonamide: To a stirred solution of 6-bromo-N-methylpyridin-2-amine (2.0 g, 10.75 mmol, 1 eq), in (50 mL) of DCM, was added TEA (3.31 mL, 23.65 mmol, 2.2 eq), followed by addition of methane sulphonyl chloride (1.25 mL, 16.12 mmol, 1.5 eq) and stirred overnight at room temperature. The reaction mixture was basified with saturated sodium bicarbonate solution and extracted with EtOAc (50 mL×2). The combined organic layers were washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and the solvent was evaporated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, N-(6-bromopyridin-2-yl)-N-methylmethanesulfonamide (2.8 g, 98.93%) as white solid.
  • LCMS: 265.0 [M+1]+
  • Step-3: Synthesis of N-(6-(2-isopropyl-6-(methylthio)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)-N-methylmethanesulfonamide: To a stirred solution of N-(6-bromopyridin-2-yl)-N-methylmethanesulfonamide (1.0 g, 4.45 mmol, 1.0 eq) and N-(4-bromopyridin-2-yl)-N-methylmethanesulfonamide (1.41 g, 5.34 mmol, 1.2 eq) in (10 mL) of dioxane was added Potassium carbonate (1.23 g, 8.9 mmol, 2 eq) and the resulting mixture was purged with nitrogen for 10 min, followed by addition of copper iodide (170 mg, 0.89 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (0.2 mL, 1.78 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 90° C. for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). Combined organic layers were washed with water (50 mL) brine solution (50 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography to afford the desired compound, N-(6-(2-isopropyl-6-(methylthio)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)-N-methylmethanesulfonamide (1.2 g, 65.93%) as an off white solid.
  • LCMS: 409.1 [M+1]+
  • Step-4: Synthesis of tert-butyl 6-((2-isopropyl-1-(6-(N-methylmethylsulfonamido)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of N-(6-(2-isopropyl-6-(methylthio)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)-N-methylmethanesulfonamide (200 mg, 0.489 mmol, 1.0 eq) in (3.0 mL) of toluene was added m-CPBA (169 mg, 0.978 mmol, 2.0 eq) and allowed to stir at rt for 1 h. tert-butyl 6-amino-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (135 mg, 0.489 mmol, 1.0 eq) and DIPEA (0.33 mL, 1.95 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 6-((2-isopropyl-1-(6-(N-methylmethylsulfonamido)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (90 mg, 28.86%) as an off white solid.
  • LCMS: 637.3 [M+1]+
  • Step-5: Synthesis of N-(6-(6-((1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)-N-methylmethanesulfonamide dihydrochloride: tert-butyl 6-((2-isopropyl-1-(6-(N-methylmethylsulfonamido)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (90 mg, 0.141 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (1 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, N-(6-(6-((1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)-N-methylmethanesulfonamide dihydrochloride (29 mg, 33.66%) as an off white solid.
  • LCMS: 537.5 [M+1]+; UPLC @ 254 nm=98.68% and @ 220 nm=99.14%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.32 (brs, 1H), 9.37 (brs, 2H), 8.84 (s, 1H), 8.14 (t, J=7.83 Hz, 1H), 7.72 (d, J=7.83 Hz, 1H), 7.67 (brs, 1H), 7.51 (d, J=7.83 Hz, 1H), 7.43 (d, J=8.31 Hz, 1H), 7.35 (d, J=8.31 Hz, 1H), 4.17-4.22 (m, 1H), 3.43 (brs, 2H), 3.32 (s, 3H), 3.21 (s, 3H), 3.01 (brs, 2H), 1.64 (s, 6H), 1.36 (d, J=6.85 Hz, 6H).
  • Example S97. Synthesis of 1-(1-cyclopropyl-JH-pyrazol-3-yl)-6-((1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-2-isopropyl-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride
  • Figure US20230109104A1-20230406-C02464
  • Step-1: synthesis of tert-butyl 6-((1-(1-cyclopropyl-1H-pyrazol-3-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(1-cyclopropyl-1H-pyrazol-3-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (95 mg, 0.287 mmol, 1.0 eq) in (1.5 mL) of toluene was added m-CPBA (99 mg, 0.574 mmol, 2.0 eq) and allowed to stir at rt for 1 h. Tert-butyl 6-amino-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (95.18 mg, 0.344 mmol, 1.2 eq) and DIPEA (0.19 mL, 1.14 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. Reaction mixture was diluted with water and extracted with EtOAc (30 mL×2). The combined organic layer were dried over sodium sulphate, concentrated under reduced pressure purified by column chromatography (Combiflash, elution-0-60% EtOAc in Hexane) to afford the desired compound, tert-butyl 6-((1-(1-cyclopropyl-1H-pyrazol-3-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (20 mg, 12.46%) as yellow liquid.
  • LCMS: 559.4 [M+1]+
  • Step-2: Synthesis of 1-(1-cyclopropyl-1H-pyrazol-3-yl)-6-((1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-2-isopropyl-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride: To a stirred solution of tert-butyl 6-((1-(1-cyclopropyl-1H-pyrazol-3-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (20 mg, 0.035 mmol, 1.0 eq) in (0.3 mL) of dioxane was added 4M dioxane-HCl (0.3 mL) and allowed to stir at rt for 2 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, of 1-(1-cyclopropyl-1H-pyrazol-3-yl)-6-((1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-2-isopropyl-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride (7.8 mg, 44.01%) as an off white solid.
  • LCMS: 459.4 [M+1]+, UPLC @ 254 nm=90.87% and @ 220 nm=91.88%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.21 (brs, 1H), 9.21 (brs, 2H), 8.81 (s, 1H), 7.97 (brs, 1H), 7.62 (brs, 1H), 7.49 (brs, 1H), 7.29 (brs, 1H), 6.46 (d, J=2.19 Hz, 1H), 4.26 (d, J=6.58 Hz, 1H), 3.81 (d, J=4.39 Hz, 2H), 2.94 (brs, 2H), 1.62 (s, 6H), 1.24 (d, J=6.58 Hz, 6H), 0.84-1.10 (m, 4H).
  • Example S98. Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-6-((2-methyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02465
  • To a stirred solution of 1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (50 mg, 0.094 mmol, 1.0 eq) and HCHO (8.49 mg, 0.282 mmol, 3.0 eq) in dichloroethane (10 mL) acetic acid (5.6 mg, 0.094 mmol, 1.0 eq) was added drop-wise at 0° C. The resulting mixture was stirred at rt for 1 h, followed by addition of NaBH(OAc)3 (20 mg, 0.094 mmol, 1.0 eq) at 0° C. The resulting mixture was stirred at rt for 1 h. The progress of reaction was monitored by LCMS. After completion of reaction, solvent was removed under reduced pressure, residue was basified with saturated solution of NaHCO3 (50 mL) and extracted with ethyl acetate (100 mL×2). The combined organic layers were washed with water (50 mL) and brine (50 mL), dried over anhydrous Na2SO4 and concentrated under reduced pressure. Crude residue was purified by flash chromatography to afford, 1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-6-((2-methyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (3 mg, 5.04%) as an off white solid. HCl salt of this compound was synthesized using 4M dioxane/HCl.
  • LCMS: 472.5 [M+1]+, UPLC @ 254 nm=95.82% and @ 220 nm=95.59%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.47 (brs, 1H), 10.26 (brs, 1H), 8.88 (s, 1H), 8.73 (d, J=4.82 Hz, 1H), 7.51-7.72 (m, 3H), 7.43 (brs, 1H), 7.15 (d, J=8.77 Hz, 1H), 4.45 (d, J=14.47 Hz, 2H), 4.24 (brs, 2H), 3.97 (brs, 2H), 3.66 (brs, 3H), 3.19 (brs, 2H), 3.04 (d, J=16.66 Hz, 2H), 2.93 (d, J=4.82 Hz, 3H), 1.35 (s, 15H).
  • Example S99. Synthesis of N-(6-(2-isopropyl-3-oxo-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)-N-methylmethanesulfonamide dihydrochloride
  • Figure US20230109104A1-20230406-C02466
  • Step-1: Synthesis of tert-butyl 6-((2-isopropyl-1-(6-(N-methylmethylsulfonamido)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of N-(6-(2-isopropyl-6-(methylthio)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)-N-methylmethanesulfonamide (200 mg, 0.489 mmol, 1.0 eq) in (3.0 mL) of toluene was added m-CPBA (169 mg, 0.978 mmol, 2.0 eq) and allowed to stir at rt for 1 h tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (146 mg, 0.58 mmol, 1.2 eq) and DIPEA (0.33 mL, 1.95 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 6-((2-isopropyl-1-(6-(N-methylmethylsulfonamido)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (90 mg, 30.19%) as an off white solid.
  • LCMS: 609.3 [M+1]+
  • Step-2: Synthesis of N-(6-(2-isopropyl-3-oxo-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)-N-methylmethanesulfonamide dihydrochloride: tert-butyl 6-((2-isopropyl-1-(6-(N-methylmethylsulfonamido)pyridin-2-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (90 mg, 0.147 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (1 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, N-(6-(2-isopropyl-3-oxo-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)-N-methylmethanesulfonamide dihydrochloride (78 mg, 90.72%) as light yellow solid.
  • LCMS: 509.4 [M+1]+; UPLC @ 254 nm=97.44% and @ 220 nm=97.61%.
  • 1H NMR (400 MHz, DMSO-d6): δ10.34 (brs, 1H), 9.33 (brs, 2H), 8.84 (s, 1H), 8.11-8.16 (m, 1H), 7.73 (d, J=7.89 Hz, 2H), 7.39-7.49 (m, 2H), 7.16 (d, J=8.77 Hz, 1H), 4.20 (brs, 3H), 3.36 (brs, 2H), 3.32 (s, 3H), 3.21 (s, 3H), 2.96-3.04 (m, 2H), 1.36 (d, J=6.58 Hz, 6H).
  • Example S100. Synthesis of 1-(1-cyclopropyl-1H-pyrazol-3-yl)-6-((4,4-dimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-2-isopropyl-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride
  • Figure US20230109104A1-20230406-C02467
  • Step-1: Synthesis of tert-butyl 7-((1-(1-cyclopropyl-1H-pyrazol-3-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-4,4-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(1-cyclopropyl-1H-pyrazol-3-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (95 mg, 0.287 mmol, 1.0 eq) in (1.5 mL) of toluene was added m-CPBA (99 mg, 0.574 mmol, 2.0 eq) and allowed to stir at rt for 1 h. Tert-butyl 7-amino-4,4-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (95.18 mg, 0.344 mmol, 1.2 eq) and DIPEA (0.19 mL, 1.14 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. The reaction mixture was diluted with water (25 mL) and extracted with EtOAc (30 mL×2). The combined organic layer were dried over sodium sulphate, concentrated under reduced pressure purified by column chromatography (Combiflash, elution-0-60% EtOAc in Hexane) to afford the desired compound, tert-butyl 7-((1-(1-cyclopropyl-1H-pyrazol-3-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-4,4-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (50 mg, 31.12%) as yellow liquid.
  • LCMS: 559.2 [M+1]+
  • Step-2: Synthesis of 1-(1-cyclopropyl-1H-pyrazol-3-yl)-6-((4,4-dimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-2-isopropyl-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride: To a stirred solution of tert-butyl 7-((1-(1-cyclopropyl-1H-pyrazol-3-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-4,4-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (50 mg, 0.089 mmol, 1.0 eq) in (0.6 mL) of dioxane was added 4M dioxane-HCl (0.6 mL) and allowed to stir at rt for 2 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(1-cyclopropyl-1H-pyrazol-3-yl)-6-((4,4-dimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-2-isopropyl-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride (36 mg, 81.26%) as light yellow solid.
  • LCMS: 459.4 [M+1]+, UPLC @ 254 nm=92.32% and @ 220 nm=89.51%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.23 (brs, 1H), 9.35 (brs, 2H), 8.80 (s, 1H), 7.95 (d, J=2.19 Hz, 1H), 7.60 (brs, 1H), 7.54 (brs, 1H), 7.37 (d, J=8.77 Hz, 2H), 6.46 (d, J=2.19 Hz, 1H), 4.23-4.31 (m, 1H), 4.16 (brs, 2H), 3.82 (d, J=3.95 Hz, 2H), 3.18 (brs, 2H), 1.12-1.40 (m, 18H), 0.99-1.09 (m, 4H).
  • Example S10. Synthesis of 2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1-(thiophen-3-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride
  • Figure US20230109104A1-20230406-C02468
  • Step-1: Synthesis of 2-isopropyl-6-(methylthio)-1-(thiophen-3-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (300 mg, 1.33 mmol) and 3-bromothiophene (221 g, 1.36 mmol) in Dioxane (10 mL) was added CuI (51 mg, 0.26 mmol), DMEDA (47 mg, 0.53 mmol). The reaction mixture was stirred at 130° C. for 14 h. The progress of reaction was monitored by TLC. After completion, the reaction mixture was extracted with EtOAc (3×50 mL) dried over Na2SO4, concentrated and purified by column chromatography (Combiflash, Elution: 0-30% EtOAc in Hexane) to afford the desired product, 2-isopropyl-6-(methylthio)-1-(thiophen-3-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (130 mg, 31.71%) as an off white solid.
  • LCMS: 307.1 [M+1]+
  • Step-2: Synthesis of tert-butyl 6-((2-isopropyl-3-oxo-1-(thiophen-3-yl)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 2-isopropyl-6-(methylthio)-1-(thiophen-3-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (130 mg, 0.424 mmol, 1.0 eq) in (6.0 mL) of toluene was added m-CPBA (146 mg, 0.84 mmol, 2 eq) and allowed to stir at rt for 1 h. Tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (125 mg, 0.51 mmol, 1.2 eq) and DIPEA (0.36 mL, 2.1 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (20 mL×3). The organic layer was dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 6-((2-isopropyl-3-oxo-1-(thiophen-3-yl)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (40 mg, 18.60%) as an off white solid.
  • LCMS: 507.2 [M+1]+
  • Step-3: Synthesis of 2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1-(thiophen-3-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride: tert-butyl 6-((2-isopropyl-3-oxo-1-(thiophen-3-yl)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (40 mg, 0.079 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl in dioxane (1 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1-(thiophen-3-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride (3 mg, 8.57%) as an off white solid.
  • LCMS: 407.3 [M+1]+; UPLC @ 254 nm=97.46% and @ 220 nm=98.32%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.26 (s, 1H), 8.99 (brs, 2H), 8.82 (s, 1H), 7.82 (brs, 1H), 7.72-7.79 (m, 1H), 7.44 (brs, 1H), 7.29 (d, J=4.38 Hz, 1H), 7.11 (d, J=8.77 Hz, 1H), 4.08-4.24 (m, 3H), 2.94 (brs, 2H), 1.29 (d, J=6.58 Hz, 6H).
  • Example S102. Synthesis of 1-(1-(tert-butyl)-JH-pyrazol-3-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride
  • Figure US20230109104A1-20230406-C02469
  • Step-1: Synthesis of 1-(1-(tert-butyl)-1H-pyrazol-3-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (442 mg, 1.96 mmol, 1.0 eq) and 3-bromo-1-(tert-butyl)-1H-pyrazole (400 mg, 1.96 mmol, 1.0 eq) in (12 mL) of dioxane was added potassium carbonate (542 mg, 3.92 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min followed by addition of copper iodide (75 mg, 0.392 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (0.08 mL, 0.784 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 90° C. for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-30% EtOAc in hexane] to afford the desired product, 1-(1-(tert-butyl)-1H-pyrazol-3-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (366 mg, 14.64%) as colorless liquid.
  • LCMS: 347.2 [M+1]+
  • Step-2: Synthesis of tert-butyl 6-((1-(1-(tert-butyl)-1H-pyrazol-3-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(1-(tert-butyl)-1H-pyrazol-3-yl)-2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (100 mg, 0.288 mmol, 1.0 eq) in (6.0 mL) of toluene was added m-CPBA (99 mg, 0.576 mmol, 2.0 eq) and allowed to stir at rt for 30 minutes. tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (86 mg, 0.604 mmol, 1.2 eq) and DIPEA (0.2 mL, 1.15 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 6-((1-(1-(tert-butyl)-1H-pyrazol-3-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (17 mg, 10.77%) as an off white solid. LCMS: 547.3 [M+1]+
  • Step-3: Synthesis of 1-(1-(tert-butyl)-1H-pyrazol-3-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride: tert-butyl 6-((1-(1-(tert-butyl)-1H-pyrazol-3-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (17 mg, 0.031 mmol, 1.0 eq) was dissolved in dioxane (0.3 mL), followed by dropwise addition of 4.0 M-HCl (0.3 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(1-(tert-butyl)-1H-pyrazol-3-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one hydrochloride (96 mg, 46.60%) as light yellow solid.
  • LCMS: 447.3 [M+1]+; UPLC @ 254 nm=90.92% and @ 220 nm=88.16%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.22 (br. s., 1H) 9.21 (br. s., 2H) 8.82 (s, 1H) 7.99 (br. s., 1H) 7.63 (br. s., 1H) 7.54 (d, J=8.80 Hz, 1H) 7.08 (d, J=8.31 Hz, 1H) 6.47 (d, J=1.96 Hz, 1H) 4.30-4.39 (m, 1H) 4.18 (br. s., 2H) 3.15-3.20 (d, 2H) 2.91 (br. s., 2H) 1.44-1.61 (m, 9H) 1.19-1.32 (m, 6H).
  • Example S103. Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-6-((4,4-dimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride
  • Figure US20230109104A1-20230406-C02470
  • Step-1: Synthesis of tert-butyl 7-((1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-4,4-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (100 mg, 0.281 mmol, 1.0 eq) in (6.0 mL) of toluene was added m-CPBA (97 mg, 0.562 mmol, 2.0 eq) and allowed to stir at rt for 1 h. tert-butyl 7-amino-4,4-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (93 mg, 0.337 mmol, 1.2 eq) and DIPEA (0.2 mL, 1.124 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 7-((1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-4,4-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (60 mg, 36.53%) as an off white solid.
  • LCMS: 584.3 [M+1]+
  • Step-2: Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-6-((4,4-dimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride: tert-butyl 7-((1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-4,4-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (60 mg, 0.102 mmol, 1.0 eq) was dissolved in dioxane (0.6 mL), followed by dropwise addition of 4.0 M-HCl (0.6 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(2-(tert-butyl)pyridin-4-yl)-2-cyclopropyl-6-((4,4-dimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one dihydrochloride (36 mg, 52.44%) as an off white solid.
  • LCMS: 486.5 [M+1]+; UPLC @ 254 nm=93.71% and @ 220 nm=94.78%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.40(br. s., 1H) 9.60 (br. s., 2H) 8.80-8.92 (m, 2H) 7.99 (br. s., 1H) 7.80 (br. s., 1H) 7.62 (d, J=8.31 Hz, 1H) 7.38-7.49 (m, 2H) 4.25 (br. s., 2H), 4.10(s., 2H), 3.90-3.99 (m, 1H) 3.10-3.27 (d, 4H) 1.39-1.55 (m, 9H) 1.37 (m, 6H).
  • Example S104. Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[4,3-c]pyridin-3-one trihydrochloride
  • Figure US20230109104A1-20230406-C02471
  • Step-1: Synthesis of methyl 6-chloro-4-hydrazinylnicotinate: Hydrazine hydrate (4.76 mL, 97.06 mmol, 2 eq) was dissolved in methanol (50 mL) and cooled to 0° C. methyl 4,6-dichloronicotinate (10.0 g, 48.53 mmol, 1 eq) was added. The reaction mixture was stirred at same temperature for 12 h. Solid obtained was filtered under vacuum to get the desired product, methyl 6-chloro-4-hydrazinylnicotinate (6.19 g, 63.29%) as an off white solid.
  • LCMS: 202.1 [M+1]+
  • Step-2: Synthesis of methyl 6-chloro-4-(2-isopropylhydrazinyl)nicotinate: Methyl 6-chloro-4-hydrazinylnicotinate (6.19 g, 3.07 mmol, 1.0 eq) was suspended in Acetone and reflex for 4 h, after the completion of reaction, the reaction mixture was evaporated under reduced pressure to afford 6.20 g of methyl 6-chloro-4-(2-(propan-2-ylidene)hydrazinyl)nicotinate. The crude product obtained, 6-chloro-4-(2-(propan-2-ylidene)hydrazinyl)nicotinate (6.20 g, 25.65 mmol, 1 eq) was dissolved in Methanol:Dichloromethane (3:1) (160 mL) and cooled at 0° C. A solution of Sodium cynoborohydride (9.67 g, 153.92 mmol, 6.0 eq) in Methanol (40 mL) and concentrated Hydrochloric acid (1.2 mL) solution was added drop wise to the reaction mixture. The reaction mixture was stirred at ambient temperature for 24 h. Saturated aqueous solution of sodium bicarbonate was added to reaction mixture till pH 7-8 and organic phase was extracted with ethyl acetate (500 mL×3). The combined organic layers were dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford, methyl 6-chloro-4-(2-isopropylhydrazinyl)nicotinate (5.91 g, 79.01%) as an off white solid.
  • LCMS: 244.1 [M+1]+
  • Step-3: Synthesis of 6-chloro-2-isopropyl-1,2-dihydro-3H-pyrazolo[4,3-c]pyridin-3-one: Methyl 6-chloro-4-(2-isopropylhydrazinyl)nicotinate (5.91 g, 24.25 mmol, 1 eq) was dissolved in Methanol (55 mL) and solution was cooled to 0° C. A solution of sodium hydroxide in water (5N, 173 mL) was added drop wise to reaction solution and reaction mixture was stirred at same temperature for 2 h. Hydrochloric acid in water (5N) was added drop wise to make pH ˜2 and stirred for 3 h at ambient temperature. After completion of reaction, dichloromethane (500 mL×2) was added in to reaction mixture and organic phase was extracted. Organic layer was dried over anhydrous Sodium sulfate and concentrated under reduced pressure. Solid obtained was washed with pentane (100 mL×2) to afford 6-chloro-2-isopropyl-1,2-dihydro-3H-pyrazolo[4,3-c]pyridin-3-one (1.5 g, 24.25%) as yellow solid.
  • LCMS: 212.2 [M+1]+
  • Step-4: Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-6-chloro-2-isopropyl-1,2-dihydro-3H-pyrazolo[4,3-c]pyridin-3-one: To a stirred solution of 6-chloro-2-isopropyl-1,2-dihydro-3H-pyrazolo[4,3-c]pyridin-3-one (1.5 g, 7.08 mmol, 1 eq) and 2-(tert-butyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridine (2.77 g, 10.63 mmol, 1.5 eq) in DCM (100 mL) was added 2,2-bipyridine (2.21 g, 14.16 mmol, 2 eq), copper acetate (2.57 g, 14.16 mmol, 2 eq) and Na2CO3 (2.25 g, 21.24 mmol, 3 eq). The reaction mixture was stirred at rt for 24 h in open air. The progress of reaction was monitored by TLC. After completion, the reaction mixture was filtered over celite to remove inorganic impurities. The filtrate was washed with water, dried over Na2SO4, concentrated and purified by column chromatography (Combiflash, Elution: 0-30% EtOAc in Hexane) to afford the desired product, 1-(2-(tert-butyl)pyridin-4-yl)-6-chloro-2-isopropyl-1,2-dihydro-3H-pyrazolo[4,3-c]pyridin-3-one (900 mg, 37.50%) as an off white solid.
  • LCMS: 345.2 [M+1]+
  • Step-5: Synthesis of tert-butyl 6-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[4,3-c]pyridin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirring solution of 1-(2-(tert-butyl)pyridin-4-yl)-6-chloro-2-isopropyl-1,2-dihydro-3H-pyrazolo[4,3-c]pyridin-3-one (200 mg, 0.579 mmol, 1 eq) in dioxane (20 mL), tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (173 mg, 0.695 mmol, 1.2 eq) and Cs2CO3 (377 mg, 1.158 mmol, 2 eq) were added. The mixture was degassed for 10-15 min followed addition of Pd2(dba)3 (64 mg, 0.069 mmol, 0.12 eq) and xantphos (50 mg, 0.086 mmol, 1.5 eq). The reaction was heated at 110° C. for 1 h under microwave irradiation. After completion, the reaction mixture was filtered through celite using EtOAc. The combined organic layer was dried, concentrated and purified by flash chromatography (EtOAc-Hexane 0-70%) to give tert-butyl 6-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[4,3-c]pyridin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (60 mg, 18.58%) as yellow solid.)
  • LCMS: 557.3 [M+1]+
  • Step-6: Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[4,3-c]pyridin-3-one trihydrochloride: tert-butyl 6-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[4,3-c]pyridin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (60 mg, 0.107 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (1 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1,2-dihydro-3H-pyrazolo[4,3-c]pyridin-3-one trihydrochloride (42 mg, 68.86%) as light yellow solid.
  • LCMS: 457.5 [M+1]+; UPLC @ 254 nm=96.90% and @ 220 nm=98.32%.
  • 1H NMR (400 MHz, DMSO-d6): δ 9.86 (br. s., 1H) 9.40 (br. s., 1H) 8.75 (d, J=5.87 Hz, 1H) 8.63 (s, 1H) 7.60 (s, 2H) 7.64 (m, 2H) 7.14 (d, J=8.80 Hz, 1H) 6.81 (br. s., 1H) 4.18 (br. s., 2H) 3.96 (m, 1H) 3.33 (br. s., 2H) 2.92-3.01 (m, 2H) 2.81 (d, J=4.40 Hz, 2H) 1.45 (s, 9H) 1.28-1.41 (m, 6H).
  • Example S105. Synthesis of 2-cyclopropyl-6-((1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02472
  • Step-1: Synthesis of 2-cyclopropyl-1-(2-(2-fluoropropan-2-yl) pyridin-4-yl)-6-(methylthio)-1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one: To a stirred solution of 2-cyclopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (250 mg, 1.124 mmol, 1.0 eq) and 4-bromo-2-(2-fluoropropan-2-yl)pyridine (293 mg, 1.349 mmol, 1.2 eq) in (6 mL) of dioxane was added potassium carbonate (310 mg, 2.249 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min followed by addition of copper iodide (43 mg, 0.224 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (0.04 mL, 0.449 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 130° C. for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired product 2-cyclopropyl-1-(2-(2-fluoropropan-2-yl) pyridin-4-yl)-6-(methylthio)-1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one. (50 mg, 12.3%) as yellow semi solid.
  • LCMS: 359.2 [M+1]+
  • Step-2: Synthesis of tert-butyl 6-((2-cyclopropyl-1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 2-cyclopropyl-1-(2-(2-fluoropropan-2-yl) pyridin-4-yl)-6-(methylthio)-1, 2-dihydro-3H-pyrazolo [3, 4-d] pyrimidin-3-one (50 mg, 0.139 mmol, 1.0 eq) in (2.0 mL) of toluene was added m-CPBA (48 mg, 0.278 mmol, 2.0 eq) and allowed to stir at rt for 60 minutes. tert-butyl 6-amino-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (46 mg, 0.167 mmol, 1.2 eq) and DIPEA (0.09 mL, 0.557 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 6-((2-cyclopropyl-1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (12 mg, 14.70%) as light yellow solid.
  • LCMS: 587.25 [M+1]+
  • Step-3: Synthesis of 2-cyclopropyl-6-((1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: tert-butyl 6-((2-cyclopropyl-1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (15 mg, 25.55 mmol, 1.0 eq) was dissolved in dioxane (1.5 mL), followed by dropwise addition of 4.0 M-HCl (0.4 mL) and allowed to stir at rt for 2 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 2-cyclopropyl-6-((1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (3.5 mg, 28.11%) as light yellow solid.
  • LCMS: 487.25 [M+1]+; UPLC @ 254 nm=93.48% and @ 220 nm=92.49%.
  • 1H NMR (400 MHz, DMSO-d6): δ ppm 10.35 (br. s., 1H) 9.43 (br. s., 1H) 8.84 (s, 1H) 8.72 (d, J=5.38 Hz, 2H) 7.59-7.79 (m, 2H) 7.51 (s, 1H) 7.33 (d, J=8.31 Hz, 2H) 3.42 (s., 6H) 3.11 (s., 2H) 3.03 (s, 2H) 3.0 (d 2H) 1.74 (s, 2H) 1.49-1.71 (m, 6H).
  • Example S106 Synthesis of 2-cyclopropyl-6-((4,4-dimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one
  • Figure US20230109104A1-20230406-C02473
  • Step-1: Synthesis of 2-cyclopropyl-1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: To a stirred solution of 2-cyclopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (250 mg, 1.124 mmol, 1.0 eq) and 4-bromo-2-(2-fluoropropan-2-yl)pyridine (293 mg, 1.349 mmol, 1.2 eq) in (5 mL) of dioxane was added potassium carbonate (310 mg, 2.249 mmol, 2.0 eq) and the resulting mixture was purged with nitrogen for 10 min followed by addition of copper iodide (43 mg, 0.224 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (0.04 mL, 0.449 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 130° C. for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-30% EtOAc in hexane] to afford the desired product, 2-cyclopropyl-1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (50 mg, 12.39%) as light yellow viscous.
  • LCMS: 359.2 [M+1]+
  • Step-2: Synthesis of tert-butyl 7-((2-cyclopropyl-1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-4,4-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 2-cyclopropyl-1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (50 mg, 0.139 mmol, 1.0 eq) in (2.0 mL) of toluene was added m-CPBA (48 mg, 0.278 mmol, 2.0 eq) and allowed to stir at rt for 60 minutes. tert-butyl 7-amino-4,4-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (46 mg, 0.167 mmol, 1.2 eq) and DIPEA (0.09 mL, 0.557 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 7-((2-cyclopropyl-1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-4,4-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (18 mg, 22.06%) as white solid. LCMS: 587.25(M+1)+
  • Step-3: Synthesis of 2-cyclopropyl-6-((4,4-dimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one: tert-butyl 7-((2-cyclopropyl-1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-4,4-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (18 mg, 30.66 mmol, 1.0 eq) was dissolved in dioxane (1.5 mL), followed by dropwise addition of 4.0 M-HCl (0.5 mL) and allowed to stir at rt for 2 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 2-cyclopropyl-6-((4,4-dimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1-(2-(2-fluoropropan-2-yl)pyridin-4-yl)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (5 mg, 33.48%) as light yellow solid.
  • LCMS: 487.26 [M+1]+; UPLC @ 254 nm=92.57% and @ 220 nm=91.91%.
  • 1H NMR (400 MHz, DMSO-d6): δ ppm 10.36 (br. s., 1H) 9.06 (d., 2H) 8.84 (s, 1H) 8.71 (d, J=5.38 Hz, 2H) 7.72 (br. s., 1H) 7.67 (d, J=3.91 Hz, 2H) 7.27-7.56 (m, 3H) 4.26 (s., 2H) 3.23 (s., 3H) 3.11 (s., 3H) 1.74 (m, 4H) 1.69 (s, 3H) 1.34 (s, 3H).
  • Example S107. Synthesis of 2-(6-(2-isopropyl-3-oxo-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)-2-methylpropanenitrile dihydrochloride
  • Figure US20230109104A1-20230406-C02474
  • Step-1: Synthesis of tert-butyl 7-((1-(6-(2-cyanopropan-2-yl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 2-(6-(2-isopropyl-6-(methylthio)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)-2-methylpropanenitrile (125 mg, 0.339 mmol, 1.0 eq) in (3.0 ML) of toluene was added m-CPBA (117 mg, 0.6792 mmol, 2.0 eq) and allowed to stir at rt for 1 h. tert-butyl 7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (101 mg, 0.4076 mmol, 1.2 eq) and DIPEA (0.25 mL, 1.3584 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane]to afford the desired compound, tert-butyl 7-((1-(6-(2-cyanopropan-2-yl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (55 mg, 28.49%) as an off white solid.
  • LCMS: 569.2 [M+1]+
  • Step-2: Synthesis of 2-(6-(2-isopropyl-3-oxo-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)-2-methylpropanenitrile dihydrochloride: tert-butyl 7-((1-(6-(2-cyanopropan-2-yl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (55 mg, 0.149 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (2 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 2-(6-(2-isopropyl-3-oxo-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)-2-methylpropanenitrile dihydrochloride (30 mg, 66.66%) as light yellow solid.
  • LCMS: 469.5 [M+1]+; UPLC @ 254 nm=95.20% and @ 220 nm=96.27%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.28 (br. s., 1H), 9.17 (br. s., 1H), 8.87 (s, 1H) 8.18 (t, J=5.26 Hz, 1H) 8.0 (d, 1H) 7.70 (br. s., 1H) 7.58 (d, J=4.82 Hz, 1H) 7.48 (d, 1H) 7.19 (d, J=8.33 Hz, 1H) 4.22 (br. s., 2H) 3.94-3.99 (m, 1H) 3.35 (t, 2H) 2.95 (t, 2H) 1.70 (s, 6H) 1.37 (d, J=7.02 Hz, 6H).
  • Example S108. Synthesis of 2-(6-(2-isopropyl-3-oxo-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)-2-methylpropanenitrile dihydrochloride
  • Figure US20230109104A1-20230406-C02475
  • Step-1: Synthesis of 2-(6-bromopyridin-2-yl)-2-cyanopropan-1-ylium: To a stirred solution of isobutyronitrile (1 g, 14.46 mmol, 1 eq), in toluene (10 mL) was added KHMDS (32 ml, 15.91 mmol, 1.1 eq), at 0° C. & the reaction mixture was stirred for 1 h. 2, 6-dibromopyridine (already dissolving in toluene 5 ml), (3.42 g, 14.46 mmol, 1.0 eq) was added dropwise & stirred for 18 h. After completion of reaction, the reaction mixture was diluted with saturated NH4Cl solution and extracted with diethyl ether (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound 2-(6-bromopyridin-2-yl)-2-cyanopropan-1-ylium (600 mg, 18.52%) as colorless liquid.
  • LCMS: 224.9 [M+1]+
  • Step-2: Synthesis of 2-(6-(2-isopropyl-6-(methylthio)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)-2-methylpropanenitrile: To a stirred solution of 2-isopropyl-6-(methylthio)-1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one (126 mg, 0.5582 mmol, 1.0 eq) and 2-(6-bromopyridin-2-yl)-2-cyanopropan-1-ylium (150 mg, 0.6696 mmol, 1.2 eq) in (10 mL) of dioxane were added Potassium carbonate (154 mg, 1.116 mmol, 2 eq) and the resulting mixture was purged with nitrogen for 10 min followed by addition of copper iodide (22 mg, 0.11 mmol, 0.2 eq), and N,N′-dimethylethylenediamine (DMEDA) (100 mg, 0.22 mmol, 0.4 eq) and again purged with nitrogen for 10 min, stirred at 90° C. for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). Combined organic layer was washed with water (50 mL) brine solution (50 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography to afford the desired compound 2-(6-(2-isopropyl-6-(methylthio)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)-2-methylpropanenitrile (200 mg 80.97%) as an off white solid.
  • LCMS: 369.2 [M+1]+
  • Step-3: Synthesis of tert-butyl 6-((1-(6-(2-cyanopropan-2-yl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirred solution of 2-(6-(2-isopropyl-6-(methylthio)-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)-2-methylpropanenitrile (125 mg, 0.339 mmol, 1.0 eq) in (3.0 ML) of toluene was added m-CPBA (117 mg, 0.6792 mmol, 2.0 eq) and allowed to stir at rt for 1 h. tert-butyl 6-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (101 mg, 0.4076 mmol, 1.2 eq) and DIPEA (0.25 mL, 1.3584 mmol, 4.0 eq) were added and allowed to stir at rt for overnight. After completion of reaction, the reaction mixture was diluted with water and extracted with EtOAc (50 mL×2). The combined organic layer was washed with water (50 mL), brine solution (50 mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure to afford crude product, which was purified by flash chromatography [silica gel 100-200 mesh; elution 0-50% EtOAc in hexane] to afford the desired compound, tert-butyl 6-((1-(6-(2-cyanopropan-2-yl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (80 mg, 41.45%) as an off white solid.
  • LCMS: 569.4 [M+1]+
  • Step-4: Synthesis of 2-(6-(2-isopropyl-3-oxo-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)-2-methylpropanenitrile dihydrochloride: tert-butyl 6-((1-(6-(2-cyanopropan-2-yl)pyridin-2-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (80 mg, 0.140 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (2 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 2-(6-(2-isopropyl-3-oxo-6-((1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-2,3-dihydro-1H-pyrazolo[3,4-d]pyrimidin-1-yl)pyridin-2-yl)-2-methylpropanenitrile dihydrochloride (50 mg, 75.75%) as light yellow solid.
  • LCMS: 469.5 [M+1]+; UPLC @ 254 nm=95.81% and @ 220 nm=96.05%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.28 (br. s., 1H), 9.17 (br. s., 1H), 8.87 (s, 1H) 8.18 (t, J=5.26 Hz, 1H) 8.0 (d, 1H) 7.70 (br. s., 1H) 7.58 (d, J=4.82 Hz, 1H) 7.48 (d, 1H) 7.19 (d, J=8.33 Hz, 1H) 4.22 (br. s., 2H) 3.94-3.99 (m, 1H) 3.35 (t, 2H) 2.95 (t, 2H) 1.70 (s, 6H) 1.37 (d, J=7.02 Hz, 6H).
  • Example S109. Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[4,3-c]pyridin-3-one trihydrochloride
  • Figure US20230109104A1-20230406-C02476
  • Step-1: Synthesis of tert-butyl 7-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[4,3-c]pyridin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirring solution of 1-(2-(tert-butyl)pyridin-4-yl)-6-chloro-2-isopropyl-1,2-dihydro-3H-pyrazolo[4,3-c]pyridin-3-one (200 mg, 0.579 mmol, 1 eq) in dioxane (20 mL), tert-butyl 7-amino-3,4-dihydroisoquinoline-2(1H)-carboxylate (173 mg, 0.695 mmol, 1.2 eq) and Cs2CO3 (377 mg, 1.158 mmol, 2 eq) were added. The mixture was degassed for 10-15 min followed addition of Pd2(dba)3 (64 mg, 0.069 mmol, 0.12 eq) and xantphos (50 mg, 0.086 mmol, 1.5 eq). The reaction was heated at 110° C. for 1 h under microwave irradiation. After completion, the reaction mixture was filtered through celite using EtOAc. The combined organic layer was dried, concentrated and purified by flash chromatography (EtOAc-Hexane 0-70%) to give tert-butyl 7-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[4,3-c]pyridin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (60 mg, 18.58%) as yellow solid.)
  • LCMS: 557.3 [M+1]+
  • Step-2: Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[4,3-c]pyridin-3-one trihydrochloride: tert-butyl 7-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[4,3-c]pyridin-6-yl)amino)-3,4-dihydroisoquinoline-2(1H)-carboxylate (60 mg, 0.107 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (1 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-6-((1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-1,2-dihydro-3H-pyrazolo[4,3-c]pyridin-3-one trihydrochloride (27 mg, 44.26%) as light yellow solid.
  • LCMS: 457.5 [M+1]+; UPLC @ 254 nm=96.96% and @ 220 nm=98.33%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.03 (br. s., 1H) 9.54 (br. s., 2H) 8.79 (d, J=6.36 Hz, 1H) 8.63 (s, 1H) 7.72 (br. s., 2H) 7.44-7.50 (m, 2H) 7.15 (d, J=8.80 Hz, 1H) 6.92 (s, 1H) 4.23 (br. s., 2H) 3.93-3.99 (m, 1H) 3.33 (br. s., 2H) 2.95 (t, J=5.87 Hz, 2H) 1.49 (s, 9H) 1.36 (s, 6H)
  • Example S110. Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-6-((1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-2-isopropyl-1,2-dihydro-3H-pyrazolo[4,3-c]pyridin-3-one trihydrochloride
  • Figure US20230109104A1-20230406-C02477
  • Step-1: Synthesis of tert-butyl 6-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[4,3-c]pyridin-6-yl)amino)-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirring solution of 1-(2-(tert-butyl)pyridin-4-yl)-6-chloro-2-isopropyl-1,2-dihydro-3H-pyrazolo[4,3-c]pyridin-3-one (200 mg, 0.579 mmol, 1 eq) in dioxane (20 mL), tert-butyl 6-amino-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (160 mg, 0.695 mmol, 1.2 eq) and Cs2CO3 (377 mg, 1.158 mmol, 2 eq) were added. The mixture was degassed for 10-15 min followed addition of Pd2(dba)3 (64 mg, 0.069 mmol, 0.12 eq) and xantphos (50 mg, 0.086 mmol, 1.5 eq). The reaction was heated at 110° C. for 1 h under microwave irradiation. After completion, the reaction mixture was filtered through celite using EtOAc. The combined organic layer was dried, concentrated and purified by flash chromatography (EtOAc-Hexane 0-70%) to give tert-butyl 6-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[4,3-c]pyridin-6-yl)amino)-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (60 mg, 23.58%) as yellow solid.)
  • LCMS: 557.3 [M+1]+
  • Step-2: Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-6-((1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-2-isopropyl-1,2-dihydro-3H-pyrazolo[4,3-c]pyridin-3-one trihydrochloride: tert-butyl 6-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[4,3-c]pyridin-6-yl)amino)-1,1-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (60 mg, 0.107 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (1 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(2-(tert-butyl)pyridin-4-yl)-6-((1,1-dimethyl-1,2,3,4-tetrahydroisoquinolin-6-yl)amino)-2-isopropyl-1,2-dihydro-3H-pyrazolo[4,3-c]pyridin-3-one trihydrochloride (12 mg, 19.68%) as light yellow solid.
  • LCMS: 485.6 [M+1]+; UPLC @ 254 nm=97.88% and @ 220 nm=97.77%.
  • 1H NMR (400 MHz, DMSO-d6): δ 10.01 (br. s., 1H) 9.70 (br. s., 2H) 8.78 (d, J=5.87 Hz, 1H) 8.64 (s, 1H) 7.66-7.80 (m, 2H) 7.51 (d, J=8.80 Hz, 1H) 7.42 (s, 1H) 7.31 (d, J=8.80 Hz, 1H) 6.91 (s, 1H) 3.89-3.96 (m, 1H) 3.35 (br. s., 2H) 3.02 (t, J=5.38 Hz, 2H) 1.65 (s, 6H) 1.49 (s, 9H) 1.36 (s, 6H).
  • Example S111. Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-6-((4,4-dimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-2-isopropyl-1,2-dihydro-3H-pyrazolo[4,3-c]pyridin-3-one trihydrochloride
  • Figure US20230109104A1-20230406-C02478
  • Step-1: Synthesis of tert-butyl 7-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[4,3-c]pyridin-6-yl)amino)-4,4-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate: To a stirring solution of 1-(2-(tert-butyl)pyridin-4-yl)-6-chloro-2-isopropyl-1,2-dihydro-3H-pyrazolo[4,3-c]pyridin-3-one (200 mg, 0.579 mmol, 1 eq) in dioxane (20 mL), tert-butyl 7-amino-4,4-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (160 mg, 0.695 mmol, 1.2 eq) and Cs2CO3 (377 mg, 1.158 mmol, 2 eq) were added. The mixture was degassed for 10-15 min followed addition of Pd2(dba)3 (64 mg, 0.069 mmol, 0.12 eq) and xantphos (50 mg, 0.086 mmol, 1.5 eq). The reaction was heated at 110° C. for 1 h under microwave irradiation. After completion, the reaction mixture was filtered through celite using EtOAc. The combined organic layer was dried, concentrated and purified by flash chromatography (EtOAc-Hexane 0-70%) to give tert-butyl 7-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[4,3-c]pyridin-6-yl)amino)-4,4-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (60 mg, 23.58%) as yellow solid.)
  • LCMS: 557.3 [M+1]+
  • Step-2: Synthesis of 1-(2-(tert-butyl)pyridin-4-yl)-6-((4,4-dimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-2-isopropyl-1,2-dihydro-3H-pyrazolo[4,3-c]pyridin-3-one trihydrochloride: tert-butyl 7-((1-(2-(tert-butyl)pyridin-4-yl)-2-isopropyl-3-oxo-2,3-dihydro-1H-pyrazolo[4,3-c]pyridin-6-yl)amino)-4,4-dimethyl-3,4-dihydroisoquinoline-2(1H)-carboxylate (60 mg, 0.107 mmol, 1.0 eq) was dissolved in dioxane (1 mL), followed by dropwise addition of 4.0 M-HCl (1 mL) and allowed to stir at rt for 1 h. After completion of reaction, the reaction mixture was filtered and dried under reduced pressure to afford the desired compound, 1-(2-(tert-butyl)pyridin-4-yl)-6-((4,4-dimethyl-1,2,3,4-tetrahydroisoquinolin-7-yl)amino)-2-isopropyl-1,2-dihydro-3H-pyrazolo[4,3-c]pyridin-3-one trihydrochloride (21 mg, 34.45%) as light yellow solid.
  • LCMS: 485.6 [M+1]+; UPLC @ 254 nm=98.86% and @ 220 nm=98.90%.
  • 1H NMR (400 MHz, DMSO-d6): δ 9.97 (br. s., 1H) 9.57 (br. s., 2H) 8.79 (d, J=5.87 Hz, 1H) 8.62 (s, 1H) 7.64-7.76 (m, 2H) 7.52 (d, J=8.31 Hz, 1H) 7.30-7.47 (m, 2H) 6.88 (br. s., 1H) 4.20-4.30(S, 4H) 3.90-3.99 (m, 1H) 3.17 (br. s., 2H) 1.48 (s, 9H) 1.24-1.39 (m, 12H).
  • Compounds 1.19-1.68 and 1.70-1.125 can be prepared according to the experimental details exemplified in Examples S1-S21 and Scheme 1, using the appropriate starting materials and reagents.
  • Other compounds can be prepared according to the experimental details exemplified in Examples S1-S111 and Scheme 1 to Scheme 13, using the appropriate starting materials and reagents.
  • BIOLOGICAL EXAMPLES Example B1. WEE1 IC50 Determination
  • IC50 values of compounds against WEE1 kinase enzyme were determined by LanthaScreen™ Terbium Labeled TR-FRET assay. Kinase assays were performed in 1× kinase buffer (#PV6135, Invitrogen, Life Technologies Grand Island, N.Y.) where total reaction volume was 10 μL in low-volume 384-well plates (#4511, Corning). Serially diluted compounds (3-fold) were incubated with WEE1 Enzyme (1 nM) (#PR7373A, Invitrogen, Life Technologies Grand Island, N.Y.) for 10 min, following which a mixture of ATP (10 μM) (#A1852, Sigma, St-Louis, Mo.) and fluorescent-PolyGT substrate (200 nM) (#PV3610, Invitrogen, Life Technologies Grand Island, N.Y.) was added and incubated in dark at room temperature for 1 h. After 1 h, 10 μL stop solution containing Terbium labeled antibody (4 nM) (#PV3529, Invitrogen, Life Technologies Grand Island, N.Y.) and EDTA (#E5134, Sigma, St-Louis, Mo.) (20 mM) in TR-FRET dilution buffer (# PV3574, Invitrogen, Life Technologies Grand Island, N.Y.) was added. Readings were taken in a Synergy Neo Plate reader (BioTek, Winooski) at single excitation of 340 nm and Dual emission at 495 nm and 520 nm respectively.
  • The % activity of test samples was calculated as (Sample−Min)*100/(Max−M). [Max: DMSO control, complete reaction with enzyme & DMSO and Min: No enzyme & DMSO]. Percent inhibition (100−% activity) was filled to the “four-parameter logistic model” in XLfit for determination of IC50 values. The results are shown in Table 2.
  • TABLE 2
    Synthesis Enzyme Activity
    Example No Compound No. Weel IC50 (uM)
    S-1 1.1 0.0024
    S-2 1.2 0.004
    S-3 1.3 <0.002
    S-4 1.4 0.0016
    S-5 1.5 0.0031
    S-6 1.6 0.0031
    S-7 1.7 0.003
    S-8 1.8 0.001
    S-9 1.9 0.001
    S-10 1.10 0.002
    S-11 1.11 0.002
    S-12 1.12 0.004
    S-13 1.13 0.0015
    S-14 1.14 0.001
    S-15 1.15 0.0015
    S-16 1.16 0.0045
    S-17 1.17 0.0095
    S-18 1.18 0.025
    S-19 1.23 0.040
    S-20 1.69 0.011
    S-21 1.70 0.015
    S-22 1.125 >5.00
    S-23 1.24 0.023
    S-24 1.31 0.005
    S-25 1.27 0.010
    S-26 1.25 0.099
    S-27 1.127 0.058
    S-28 1.126 <0.001
    S-29 1.47 0.005
    S-30 1.128 0.006
    S-31 1.129 0.006
    S-32 1.130 0.018
    S-33 1.131 0.0006
    S-34 1.132 0.004
    S-35 1.41 0.044
    S-36 1.133 0.006
    S-37 1.134 0.009
    S-38 1.135 0.020
    S-39 1.136 0.007
    S-40 1.137 0.004
    S-41 1.138 0.006
    S-42 1.139 0.004
    S-43 1.140 0.005
    S-44 1.141 0.024
    S-45 1.142 0.004
    S-46 1.143 0.003
    S-47 1.144 0.0006
    S-48 1.145 0.003
    S-49 1.146 0.009
    S-50 1.147 0.007
    S-51 1.148 0.002
    S-52 1.149 0.003
    S-53 1.150 0.013
    S-54 1.151 2.540
    S-55 1.152 <0.001
    S-56 1.153 0.016
    S-57 1.154 0.008
    S-58 1.155 0.002
    S-59 1.156 0.001
    S-60 1.157 0.005
    S-61 1.158 0.004
    S-62 1.159 0.030
    S-63 1.160 0.030
    S-64 2.287 0.120
    S-65 2.288 0.217
    S-66 1.161 0.004
    S-67 1.162 0.011
    S-68 1.163 0.015
    S-69 1.164 0.008
    S-70 1.165 0.014
    S-71 1.166 0.029
    S-72 1.167 0.041
    S-73 1.168 4.150
    S-74 1.169 0.016
    S-75a 1.170 0.012
    S-75b 1.171 0.006
    S-76 1.172 0.012
    S-77 1.173 0.343
    S-78 1.174 0.108
    S-79 1.175 0.156
    S-80 1.176 0.008
    S-81 1.177 0.012
    S-82 1.178 0.014
    S-83 2.182 0.062
    S-84 2.183 0.004
    S-85 2.184 0.002
    S-86 2.185 0.034
    S-87 2.186 0.024
    S-88 2.187 0.012
    S-89 2.188 0.053
    S-90 2.189 0.043
    S-91 2.289 0.049
    S-92 2.290 0.090
    S-93 2.291 0.046
    S-94 2.292 0.043
    S-95 2.293 >3.65
    S-96 2.642 0.006
    S-97 2.643 0.026
    S-98 2.644 0.005
    S-99 2.645 0.005
    S-100 2.646 0.015
    S-101 2.647 0.010
    S-102 2.454 0.048
    S-103 2.600 0.025
    S-104 2.732 0.179
    S-105 2.555 0.153
    S-106 2.605 0.123
    S-107 3.035 0.002
    S-108 3.036 0.005
    S-109 2.744 ND
    S-110 2.792 ND
    S-111 2.797 ND
    ND: Not Determined
  • Example B2. Determination of Potency of Compounds in Cytotoxicity Assay in A427, A549, As-Pc-1, Panc 10.05 and A172 Cell Lines
  • A549 (CCL-185; ATCC) and A427 (HTB-53; ATCC), both lung epithelial cell lines were seeded in their respective medium (DMEM/MEM, 10569044/41090101; Gibco) at a cell count of 1500 cells per 100 L per well in a 96 well edge plate (167425; ThermoFisher). Cells were allowed to grow at 37° C. for 24 hr in 5% CO2 environment (culture conditions) in a Nuaire incubator (humidified). Serially diluted test compounds (100 μL) within the desired testing concentration ranges were added to the culture plate were further incubated in culture conditions for 72 hr and 96 hr for A427 and A549 respectively. The experiment was terminated at the designated incubation time by replacing the medium with 100 μL of 1 mM of resazurin (R7017; Sigma) prepared in respective culture medium, and the plates were further incubated in culture conditions for 4-6 hr. Fluorescence was recorded using a multimodal plate reader (Biotek Synergy Neo) at an excitation wavelength of 535 nm and emission wavelength of 590 nm to obtain relative fluorescence units. Data analysis was done by subtracting the background fluorescence (only medium blank) value from each reading and then normalizing with the vehicle control (DMSO treated cells) to obtain percent survival/proliferation. Percent survival was then subtracted by 100 to get the percent inhibition of proliferation which was used to calculate IC50 values. Potency of compounds in other cell lines (As-Pc-1, Panc 10.05, A172) was determined in an analogous manner. The results are shown in Table 3.
  • TABLE 3
    Cell Cell Cell Cell Cell
    Viability Viability Viability Viability Viability
    Synthesis IC50 A427 IC50 A549 IC50 As-Pc- IC50 Panc IC50 A172
    Example No Compound No. (μM) (μM) 1 (μM) 10.05 (μM) (μM)
    S-1 1.1 0.095 0.46 ND ND ND
    S-2 1.2 0.33 0.842 ND ND ND
    S-3 1.3 0.62 1.22 ND ND ND
    S-4 1.4 0.065 0.272 ND ND ND
    S-5 1.5 0.12 0.585 ND ND ND
    S-6 1.6 0.355 0.972 ND ND ND
    S-7 1.7 0.8 0.66 ND ND ND
    S-8 1.8 ND 0.545 ND ND ND
    S-9 1.9 ND 0.52 ND ND ND
    S-10 1.10 0.34 0.51 ND ND ND
    S-11 1.11 0.43 1.18 ND ND ND
    S-12 1.12 1.715 1.055 ND ND ND
    S-13 1.13 1 0.895 ND ND ND
    S-14 1.14 ND 1.62 ND ND ND
    S-15 1.15 0.56 1.195 ND ND ND
    S-16 1.16 1.42 1.55 ND ND ND
    S-17 1.17 2.323 9.065 ND ND ND
    S-18 1.18 7.06 ND ND ND ND
    S-19 1.23 3.21 ND ND ND ND
    S-20 1.69 0.485 ND ND ND ND
    S-21 1.70 0.640 ND ND ND ND
    S-22 1.125 >30 ND ND ND ND
    S-23 1.24 6.03 ND ND ND ND
    S-24 1.31 1.32 ND ND ND ND
    S-25 1.27 4.14 ND ND ND ND
    S-26 1.25 >30 ND ND ND ND
    S-27 1.127 >23 ND ND ND ND
    S-28 1.126 0.275 ND ND ND ND
    S-29 1.47 3.02 ND ND ND ND
    S-30 1.128 1.55 ND ND ND ND
    S-31 1.129 0.865 ND ND ND ND
    S-32 1.130 9.07 ND ND ND ND
    S-33 1.131 0.285 ND ND ND ND
    S-34 1.132 1.68 ND ND ND ND
    S-35 1.41 5.12 ND ND ND ND
    S-36 1.133 0.583 ND ND ND ND
    S-37 1.134 0.820 ND ND ND ND
    S-38 1.135 0.440 ND ND ND ND
    S-39 1.136 1.086 ND ND ND ND
    S-40 1.137 0.415 ND ND ND ND
    S-41 1.138 0.993 2.42 0.20 2.82  4.09 
    S-42 1.139 0.355 ND ND ND ND
    S-43 1.140 0.555 ND ND 1.52  ND
    S-44 1.141 2.97 ND ND ND ND
    S-45 1.142 0.390 ND ND ND ND
    S-46 1.143 0.470 2.85 0.27 1.85  3.625
    S-47 1.144 0.260 ND ND ND ND
    S-48 1.145 0.255 ND ND ND ND
    S-49 1.146 0.965 6.685 0.92 5.695 6.815
    S-50 1.147 0.370 ND ND ND ND
    S-51 1.148 1.085 ND ND ND ND
    S-52 1.149 1.130 ND ND ND ND
    S-53 1.150 3.0 ND ND ND ND
    S-54 1.151 3.38 ND ND ND ND
    S-55 1.152 1.02 ND ND ND ND
    S-56 1.153 2.54 ND ND ND ND
    S-57 1.154 2.37 ND ND ND ND
    S-58 1.155 1.98 ND ND ND ND
    S-59 1.156 1.77 ND ND ND ND
    S-60 1.157 0.460 ND ND ND ND
    S-61 1.158 0.200 ND ND ND ND
    S-62 1.159 3.32 ND ND ND ND
    S-63 1.160 3.98 ND ND ND ND
    S-64 2.287 1.96 ND ND ND ND
    S-65 2.288 2.58 ND ND ND ND
    S-66 1.161 0.240 ND ND ND ND
    S-67 1.162 0.385 ND ND ND ND
    S-68 1.163 0.720 ND ND ND ND
    S-69 1.164 0.485 ND ND ND ND
    S-70 1.165 1.57 ND ND ND ND
    S-71 1.166 4.13 ND ND ND ND
    S-72 1.167 2.06 ND ND ND ND
    S-73 1.168 4.0 ND ND ND ND
    S-74 1.169 2.74 ND ND ND ND
    S-75a 1.170 0.775 ND ND ND ND
    S-75b 1.171 0.735 ND ND ND ND
    S-76 1.172 1.35 ND ND ND ND
    S-77 1.173 2.33 ND ND ND ND
    S-78 1.174 4.0 ND ND ND ND
    S-79 1.175 1.16 ND ND ND ND
    S-80 1.176 0.595 ND ND ND ND
    S-81 1.177 1.97 ND ND ND ND
    S-82 1.178 6.20 ND ND ND ND
    S-83 2.182 0.720 7.75 1.74 8.6  10.64 
    S-84 2.183 0.135 1.805  0.095 0.805 1.635
    S-85 2.184 0.090 0.655  0.085 0.545 0.985
    S-86 2.185 1.54 ND ND ND ND
    S-87 2.186 0.905 ND ND ND ND
    S-88 2.187 0.435 ND ND ND ND
    S-89 2.188 0.815 0.91  0.965 3.105 1.525
    S-90 2.189 1.99 ND ND ND ND
    S-91 2.289 8.76 ND ND ND ND
    S-92 2.290 3.37 ND ND ND ND
    S-93 2.291 7.99 ND ND ND ND
    S-94 2.292 13.30 ND ND ND ND
    S-95 2.293 >30 ND ND ND ND
    S-96 2.642 2.46 ND ND ND ND
    S-97 2.643 2.18 ND ND ND ND
    S-98 2.644 2.33 ND ND ND ND
    S-99 2.645 4.01 ND ND ND ND
    S-100 2.646 4.37 ND ND ND ND
    S-101 2.647 2.65 ND ND ND ND
    S-102 2.454 ND ND ND ND ND
    S-103 2.600 ND ND ND ND ND
    S-104 2.732 ND ND ND ND ND
    S-105 2.555 ND ND ND ND ND
    S-106 2.605 5.96 ND ND ND ND
    S-107 3.035 0.420 ND ND ND ND
    S-108 3.036 0.250 ND ND ND ND
    S-109 2.744 10.37 ND ND ND ND
    S-110 2.792 ND ND ND ND ND
    S-111 2.797 ND ND ND ND ND
    ND: Not Determined
  • Example B3. Determination of Potency of Compounds in Cell Proliferation Assay in Selected Cancer Cell Lines and Cellular PD Effects
  • The effects of test compounds were studied in five cell lines with various histotypes. The cancer cells (Table 4) were harvested during the logarithmic growth period and counted. Adjust cell concentrations to the appropriated number with respective medium, and add 90 μL cell suspensions to 96-well plates. After cells were seeded, the plates were shaken gently to distribute cells evenly and incubated at 37° C., 5% CO2 on day 1.
  • TABLE 4
    Cell Culture Conditions
    No. Cell Line Histopathology Medium
    1 A427 Lung adenocarcinoma MEM + 10% FBS +
    NEAA + Sodium
    Pyruvate
    2 LoVo Colorectal adenocarcinoma Ham's F12K + 10% FBS
    3 NCI-H460 Large-cell lung carcinoma RPMI1640 + 10% FBS
    4 HCT-116 Colorectal carcinoma McCoy's 5a + 10% FBS
    5 A2780 Ovarian cancer RPMI1640 + 10% FBS
  • Cells were treated with test compounds at 9 concentrations within a desired concentration range (e.g. 1.5 nM-10 μM) on day 2 by series diluting the test compound stock solution (10 mMv in DMSO) with culture medium. Cell viability was assessed by Cell Titer-Glo® as recommended by Promega (Cat. No.: G7572, Promega) typically 72 h post-treatment.
  • Cell viability data were plotted using GraphPad Prism (version 5, GraphPad Software, Inc., San Diego, Calif.). In addition, a nonlinear regression model with a sigmoidal dose response and variable slope within GraphPad Prism was used to calculate the IC50 value of individual test compounds.
  • TABLE 5
    IC50 (μM)
    Compound No. LOVO HCT116 NCI-H460 A427 A2780
    1.1 0.258 1.040 >10 0.064 ND
    1.2 ND ND ND 0.269 ND
    1.3 ND ND ND 0.483 ND
    1.4 ND ND ND 0.044 ND
    1.5 ND ND ND 0.070 0.140
    1.6 ND ND ND 0.277 ND
    1.7 ND ND ND ND 1.096
    ND: Not Determined
  • Additional test compounds will be studied in the same and/or other cancer cell lines with various sensitivities to reported Wee1 compounds using similar proliferation method with possible variables, such as cell seeding densities and/or incubation durations. Example B4. Determination of potency of compounds by assay of cellular PD effects.
  • pCDC2 and γ-H2AX are two clinical relevant biomarkers associated with Wee1 inhibition. CDC2Y15 phosphorylation in cells was reported to be abolished by Wee1 inhibitors (Gavory G et. al., Almac Discovery, AACR poster, 2016). γ-H2AX, a DNA double-strand break marker, was upregulated by Wee1 treatment in Wee1 sensitive cell lines (Guertin A D et al., Molecular Cancer Therapeutics, 2013). The effects of selected test compounds on pCDC2 and γ-H2AX will be assessed in selected cancer cell lines post 24 or 48 hr treatment using Western blotting methods with selective antibodies (Guertin A D et al., Molecular Cancer Therapeutics, 2013).
  • Changes in the levels of phospho-CDC2 following treatment of cells with test compounds are assessed by enzyme-linked immunosorbent assay (ELISA) or Western blotting. A427 cells (or other suitable cell line) are plated in 6-well plates and cultured for 24 hr to approximately 80-90% confluency. Medium is then replaced, and the cells are treated with test compound at several different concentrations as well as vehicle control. After incubation of treated cells in cell culture conditions for a specified time (e.g., 24 hr), cells are rinsed with ice-cold PBS and lysed in 1× cell lysis buffer containing protease inhibitors and phosphatase inhibitors. The cells are scraped from the plate with a cell scraper after a brief incubation on ice and transferred to a centrifuge tube, and then subjected to three freeze-thaw cycles in liquid nitrogen and a 37° C. water bath for further lysis. The lysates are centrifuged to pellet cell debris (using, for example, a 10 min centrifugation of 2000× g at 4° C.) and the supernatants transferred to fresh tubes on ice. The protein concentrations of the samples are estimated by the Bradford method or equivalent. The ELISA is carried out with the PathScan® Phospho-CDC2 (Tyr15) Sandwich ELISA Kit (Cat. #7176, Cell Signaling Technology, Danvers, Mass.) or similar product according to the manufacturer's instructions. Changes in the levels of phospho-CDC2 may alternatively or additionally be analyzed by Western blotting of the samples using a primary antibody to phospho-CDC2 such as phospho-CDC2 (Tyr15) (10A11) rabbit mAb (Cat. #4539, Cell Signaling Technology) or rabbit polyclonal anti-CDK1 (phospho Y15) antibody (Cat. #ab47594, Abcam, Cambridge, United Kingdom).
  • Example B5. Determination of Activity of Compounds in Cancer Cells in Combination with Various DNA-Damaging Agents
  • The activity of test compounds in combination with cisplatin in A427 cells was determined. Cells were seeded in a 96 well plate at 2000 cells/well. The next day, cells were treated with 2.5 μM cisplatin or vehicle (1× PBS) and incubated in culture conditions (37° C., 5% CO2) for 24 hr. Following the incubation, culture medium was replaced with medium containing test compound and cisplatin/vehicle, and cells were further incubated in culture conditions for another 72 hr. By this procedure, cisplatin was either continued or discontinued after an initial 24-hr incubation. Addition of test compound was in concentrations needed to obtain an 8-point dose response curve, with concentrations prepared by 3-fold dilution. The assay was terminated upon addition of resazurin, incubation for 4 hr in culture conditions and measurement of fluorescence at excitation and emission wavelengths of 535 and 590 nm, respectively. Assay results are shown in Table 6.
  • It was reported that MK-1775 (aka AZD-1775), a potent and selective small molecule inhibitor of Wee1, in combination with gemcitabine, carboplatin, or cisplatin abolished the phosphorylation of CDC2 at Tyr15 residue and abrogate the DNA damage checkpoint, leading to apoptosis (Hirai H et al. Mol Cancer Ther 2009; 8:2992-3000, Small-molecule inhibition of Wee1 kinase by MK-1775 selectively sensitizes p53-deficient tumor cells to DNA-damaging agents).
  • Various cancer cell lines with p53 mutation will be studied by co-incubation of test compound and one of the various DNA-damaging agents, such as pemetrexed, doxorubicin, camptothecin, mitomycin C, gemcitabine, and 5-FU, etc. The anti-proliferation effects of the DNA-damaging agents will be evaluated in the presence or absence of individual test compound using CTG assays described in Example B3. The concentrations of test compounds in the combination studies will be selected based on the anti-cell proliferation effects of test compounds in cancer cell lines as monotherapy using CTG assays. Incubation time will be optimized prior to the combination treatment for individual test compound. In vitro mechanism based studies using histology staining and/or flow cytometry methods as described by Hirai H. et al. (Hirai H 2010, MK-1775 enhances antitumor efficacy 5-FU) may be used.
  • In addition, the sensitization of test compounds in drug-induced resistance cell lines (e.g., A2780cis) will be studied in vitro as combination therapy (e.g., cisplatin+Wee1 inhibitor) with the anti-proliferation assays, histology staining and/or flow cytometry methods mentioned therebefore.
  • TABLE 6
    Cell viability IC50 (μM) in A427 cells with 2.5 μM cisplatin
    Compound 24 hr cisplatin 24 hr cisplatin then
    No. then compound compound + cisplatin
    1.4 0.04 <0.014
    1.5 0.05 <0.014
    1.12 0.12 0.07
    1.16 0.1 <0.014
  • Example B6. Determination of Synergistic Activity of Compounds Combined with Chemotherapeutics in Cancer Cells
  • Determination of synergistic activity of compounds in combination with a chemotherapeutic drug on cell viability is determined by a combination matrix method. Chemotherapeutics that may be used in the combination include but are not limited to a platinum-based chemotherapeutic agent, a DNA alkylating agent, a topoisomerase inhibitor, an anthracycline, a histone deacetylase inhibitor, a bromodomain inhibitor, a kinase inhibitor, a mTOR inhibitor, a PARP inhibitor, an ATM inhibitor, an ATR inhibitor, a Wee1 inhibitor, a proteasome inhibitor, and a nucleotide analog or precursor analog. Cancer cell lines that may be used in the assay include but are not limited to lung cancer, leukemia, lymphoma, multiple myeloma, ovarian cancer, breast cancer, pancreatic cancer, stomach cancer, colon cancer, liver cancer, head and neck cancer, kidney cancer, skin cancer and brain cancer cell lines. Cells are seeded in a 96-well plate and incubated at 37° C. in cell culture conditions for 24 hr. Drugs are added and then the cells are incubated further at 37° C. in cell culture conditions for 72 hr. Cells are treated with single agents to obtain a dose response curve for each agent. Cells are also treated with combinations of the drugs, based on a matrix generated by combining the two drugs at all different combinations of the doses used in the dose response curves. The assay is terminated by addition of Resazurin, incubation for 4 hr at 37° C., 5% CO2. Measurement of fluorescence at an excitation and emission wavelength of 535 and 590 nm respectively. Synergy is evaluated with the combination index (CI) value using the Chou-Talalay method in which additive effect (CI=1), synergism (CI<1), and antagonism (CI>1) in drug combinations is determined (Chou T C. Cancer Res 2010; 70:440-6). A fixed drug ratio dilution method in which drugs are combined in a fixed ratio which is diluted to 5 or more dilutions, may also be used in place of the combination matrix method.
  • Example B7. Determination of Synergistic Activity of Compound 1.138 Combined with Gemcitabine in Pancreatic Cancer Cells
  • An assay testing the combination of compound 1.138 with the chemotherapeutic drug gemcitabine was conducted according to the method of Example B6. The pancreatic cancer cell line Panc 10.05 was seeded in a 96-well plate at 2000 cells/well. In a separate experiment, the pancreatic cancer cell line AsPC-1 was seeded in a 96-well plate at 2000 cells/well. Drug concentrations ranged from about 14 nM to 30 mM for compound 1.138 and 0.4 nM to 300 nM for gemcitabine in the dose response curves for the single agents. Each dose used in the dose response curve for compound 1.138 was combined with each dose used in the dose response curve for gemcitabine to generate the matrix combinations, which included all possible combinations of the doses. Combination of compound 1.138 and gemcitabine in Panc 10.05 cells resulted in combination index values less than 1 at a fractional effect level of about 0.5, indicating synergistic effects of the drugs in reducing cell viability. Combination of compound 1.138 and gemcitabine in AsPC-1 cells also resulted in combination index values less than 1 at a fractional effect level of about 0.5, indicating synergistic effects of the drugs in reducing cell viability.
  • Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it is apparent to those skilled in the art that certain minor changes and modifications will be practiced in light of the above teaching. Therefore, the description and examples should not be construed as limiting the scope of the invention.

Claims (26)

1-61. (canceled)
62. A method of treating a cancer in an individual in need thereof comprising administering to the individual a therapeutically effective amount of a compound of Formula (I):
Figure US20230109104A1-20230406-C02479
or a tautomer or stereoisomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein:
X1 is N or CR;
X2 is N or CR, wherein at least one of X1 and X2 is N;
each R is independently hydrogen, C1-C6 alkyl, halogen or CN;
A is phenyl or 5- to 6-membered heteroaryl, each of which is optionally substituted by R1;
B is C6-C14 aryl, 5- to 10-membered heteroaryl, C3-C8 cycloalkyl, or 3- to 12-membered heterocyclyl, each of which is optionally substituted by R4, wherein A is fused to B;
m is 0, 1, 2, 3, or 4;
n is 0, 1, 2, 3, or 4;
R1 is independently halogen, —CN, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, —NR17R18 or —C(O)NR17R18;
R2 is independently hydrogen, C1-C6 alkyl, C3-C6 cycloalkyl, —(C1-C3 alkylene)CN, —(C1-C3alkylene)OR10, —(C1-C3 alkylene)NR11R12, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R10, —(C1-C3 alkylene)C(O)NR11R12, —(C2-C3 alkylene)NR10C(O)R11, —(C1-C3 alkylene)S(O)2R10, —(C2-C3 alkylene)NR10S(O)2R11, —(C1-C3 alkylene)S(O)2NR11R12, —(C1-C3 alkylene)(3- to 12-membered heterocyclyl), —(C1-C3 alkylene)(5- to 10-membered heteroaryl), or —(C1-C3 alkylene)(C6-C14 aryl), wherein each R2 is independently optionally substituted by halogen, oxo, —OR13, C1-C6 alkyl, or —NR13R14;
R3 is independently hydrogen, C1-C6 alkyl, —C(O)R10, —S(O)R10, —S(O)2R10, C3-C6 cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, C6-C14 aryl, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3- to 12-membered heterocyclyl), —(C1-C3 alkylene)(5- to 10-membered heteroaryl), or —(C1-C3 alkylene)(C6-C14 aryl), wherein each R3 is independently optionally substituted by halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that the 5- to 10-membered heteroaryl and C6-C14 aryl of R3 is not substituted by
Figure US20230109104A1-20230406-C02480
each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR17S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl) or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19,
or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19;
R10 is independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl, or 3- to 6-membered heterocyclyl, wherein the C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl and 3- to 6-membered heterocyclyl are independently optionally substituted by halogen, oxo, —CN, —OR1, —NR15R16, —P(O)(OR15)(OR16), phenyl optionally substituted by halogen, or C1-C6 alkyl optionally substituted by halogen, —OH or oxo;
R11 and R12 are each independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl, or 3- to 6-membered heterocyclyl, wherein the C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl and 3- to 6-membered heterocyclyl are independently optionally substituted by halogen, oxo, —CN, —OR1, —NR15R16, or C1-C6 alkyl optionally substituted by halogen, —OH or oxo,
or R11 and R12 are taken together with the atom to which they attached to form a 3- to 6-membered heterocyclyl optionally substituted by halogen, oxo, —OR1, —NR15R16 or C1-C6 alkyl optionally substituted by halogen, oxo or —OH;
R13 and R14 are each independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl, 3- to 6-membered heterocyclyl, —C(O)OR15, —(C1-C3 alkylene)OR15 or —C(O)NR15R16, wherein the C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl and 3- to 6-membered heterocyclyl are independently optionally substituted by halogen, oxo, —CN, —OR15, —NR15R16, or C1-C6 alkyl optionally substituted by halogen, —OH or oxo,
or R13 and R14 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl optionally substituted by halogen or C1-C6 alkyl optionally substituted by halogen or oxo;
R15 and R16 are each independently hydrogen, C1-C6 alkyl optionally substituted by halogen or oxo, C2-C6 alkenyl optionally substituted by halogen or oxo, or C2-C6 alkynyl optionally substituted by halogen or oxo,
or R15 and R16 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl optionally substituted by halogen, oxo or C1-C6 alkyl optionally substituted by oxo or halogen; and
each R17, R18, R19, and R20 is independently hydrogen, C3-C6 cycloalkyl, 3-6 membered heterocyclyl or C1-C6 alkyl, each of which is optionally substituted by halogen, oxo or —OH,
or R17 and R18 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl optionally substituted by halogen, oxo or —OH.
63-66. (canceled)
67. The method of claim 62, wherein the cancer comprises a mutant TP53 gene.
68-71. (canceled)
72. A method of inhibiting Wee1 in a cell, comprising administering a compound of Formula (I):
Figure US20230109104A1-20230406-C02481
or a tautomer or stereoisomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, to the cell, wherein:
X1 is N or CR;
X2 is N or CR, wherein at least one of X1 and X2 is N;
each R is independently hydrogen, C1-C6 alkyl, halogen or CN;
A is phenyl or 5- to 6-membered heteroaryl, each of which is optionally substituted by R1;
B is C6-C14 aryl, 5- to 10-membered heteroaryl, C3-C8 cycloalkyl, or 3- to 12-membered heterocyclyl, each of which is optionally substituted by R4, wherein A is fused to B;
m is 0, 1, 2, 3, or 4;
n is 0, 1, 2, 3, or 4;
R1 is independently halogen, —CN, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, —NR17R18 or —C(O)NR17R18;
R2 is independently hydrogen, C1-C6 alkyl, C3-C6 cycloalkyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR10, —(C1-C3 alkylene)NR11R12, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R10, —(C1-C3 alkylene)C(O)NR11R12, —(C2-C3 alkylene)NR10C(O)R11, —(C1-C3 alkylene)S(O)2R10, —(C2-C3 alkylene)NR10S(O)2R11, —(C1-C3 alkylene)S(O)2NR11R12, —(C1-C3 alkylene)(3- to 12-membered heterocyclyl), —(C1-C3 alkylene)(5- to 10-membered heteroaryl), or —(C1-C3 alkylene)(C6-C14 aryl), wherein each R2 is independently optionally substituted by halogen, oxo, —OR13, C1-C6 alkyl, or —NR13R14;
R3 is independently hydrogen, C1-C6 alkyl, —C(O)R10, —S(O)R10, —S(O)2R10, C3-C6 cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, C6-C14 aryl, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3- to 12-membered heterocyclyl), —(C1-C3 alkylene)(5- to 10-membered heteroaryl), or —(C1-C3 alkylene)(C6-C14 aryl), wherein each R3 is independently optionally substituted by halogen, oxo, —OR3, —NR13R14, —C(O)R13, —C(O)OR1, —C(O)NR13R14, —NR13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R1, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR5R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that the 5- to 10-membered heteroaryl and C6-C14 aryl of R3 is not substituted by
Figure US20230109104A1-20230406-C02482
each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —C(O)R17, —C(O)OR17, —C(O)NR17R18, —CN, —Si(C1-C6 alkyl)3, —OR17, —NR17R18, —OC(O)NR17R18, —NR17C(O)R18, —S(O)2R17, —NR17S(O)2R18, —S(O)2NR17R18, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR17, —(C1-C3 alkylene)NR17R18, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R17, —(C1-C3 alkylene)C(O)NR17R18, —(C1-C3 alkylene)NR17C(O)R18, —(C1-C3 alkylene)S(O)2R17, —(C1-C3 alkylene)NR17S(O)2R18, —(C1-C3 alkylene)S(O)2NR17R18, —(C1-C3 alkylene)(C3-C6 cycloalkyl) or —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19,
or two R4, when bound to the same carbon or two different carbon atoms, are taken together with the carbon or carbons to which they are attached to form a C3-C6 cycloalkyl or 3- to 7-membered heterocyclyl, each is optionally substituted by R19;
R10 is independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl, or 3- to 6-membered heterocyclyl, wherein the C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl and 3- to 6-membered heterocyclyl are independently optionally substituted by halogen, oxo, —CN, —OR15, —NR15R16, —P(O)(OR15)(OR16), phenyl optionally substituted by halogen, or C1-C6 alkyl optionally substituted by halogen, —OH or oxo;
R11 and R12 are each independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl, or 3- to 6-membered heterocyclyl, wherein the C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl and 3- to 6-membered heterocyclyl are independently optionally substituted by halogen, oxo, —CN, —OR15, —NR15R16, or C1-C6 alkyl optionally substituted by halogen, —OH or oxo,
or R11 and R12 are taken together with the atom to which they attached to form a 3- to 6-membered heterocyclyl optionally substituted by halogen, oxo, —OR15, —NR15R16 or C1-C6 alkyl optionally substituted by halogen, oxo or —OH;
R13 and R14 are each independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl, 3- to 6-membered heterocyclyl, —C(O)OR15, —(C1-C3 alkylene)OR15 or —C(O)NR15R16, wherein the C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl and 3- to 6-membered heterocyclyl are independently optionally substituted by halogen, oxo, —CN, —OR15, —NR15R16, or C1-C6 alkyl optionally substituted by halogen, —OH or oxo,
or R13 and R14 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl optionally substituted by halogen or C1-C6 alkyl optionally substituted by halogen or oxo;
R15 and R16 are each independently hydrogen, C1-C6 alkyl optionally substituted by halogen or oxo, C2-C6 alkenyl optionally substituted by halogen or oxo, or C2-C6 alkynyl optionally substituted by halogen or oxo,
or R15 and R16 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl optionally substituted by halogen, oxo or C1-C6 alkyl optionally substituted by oxo or halogen; and
each R17, R18, R19, and R20 is independently hydrogen, C3-C6 cycloalkyl, 3-6 membered heterocyclyl or C1-C6 alkyl, each of which is optionally substituted by halogen, oxo or —OH,
or R17 and R18 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl optionally substituted by halogen, oxo or —OH.
73-75. (canceled)
76. The method of claim 62, comprising administering to the individual a therapeutically effective amount of a compound of Formula (Ia),
Figure US20230109104A1-20230406-C02483
or a tautomer or stereoisomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein:
A is phenyl or 6-membered heteroraryl, each of which is optionally substituted by R1;
B is C6 aryl, 5- to 6-membered heteroaryl, C3-C8 cycloalkyl, or 3- to 8-membered heterocyclyl, each of which is optionally substituted by R4, wherein A is fused with B;
m is 0, 1, 2, 3, or 4;
n is 0, 1, 2, 3, or 4;
R1 is independently halogen, —CN, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, or —C(O)NR17R18;
R2 is independently hydrogen, C1-C6 alkyl, C3-C6 cycloalkyl, —(C1-C3 alkylene)CN, —(C1-C3alkylene)OR10, —(C1-C3 alkylene)NR11R12, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)C(O)R10, —(C1-C3 alkylene)C(O)NR11R12, —(C2-C3 alkylene)NR10C(O)R11, —(C1-C3 alkylene)S(O)2R10, —(C2-C3alkylene)NR10S(O)2R11, —(C1-C3 alkylene)S(O)2NR11R12, —(C1-C3 alkylene)(3- to 12-membered heterocyclyl), —(C1-C3 alkylene)(5- to 10-membered heteroaryl), or —(C1-C3 alkylene)(C6-C14 aryl), wherein each R2 is independently optionally substituted by halogen, oxo, —OR13, C1-C6 alkyl, or —NR13R14;
R3 is independently hydrogen, C1-C6 alkyl, —C(O)R10, —S(O)R10, —S(O)2R10, C3-C6 cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, C6-C14 aryl, —(C1-C3alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3- to 12-membered heterocyclyl), —(C1-C3 alkylene)(5- to 10-membered heteroaryl), or —(C1-C3 alkylene)(C6-C14 aryl), wherein each R3 is independently optionally substituted by halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)NR13R14, —NR 13C(O)R14, —CN, —S(O)R13, —S(O)2R13, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH or halogen, with the proviso that the 5- to 10-membered heteroaryl and C6-C14 aryl of R3 is not substituted by
Figure US20230109104A1-20230406-C02484
each R4 is independently oxo, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —C(O)R17, —C(O)OR17, or —C(O)NR17R18, wherein each R4 is independently optionally substituted by halogen, oxo, —OR19, —NR19R20, or —C(O)R19;
or two R4 are taken together with the carbon to which they attach to form a C3-C6 cycloalkyl;
R10 is independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl, or 3- to 6-membered heterocyclyl, wherein the C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl and 3- to 6-membered heterocyclyl are independently optionally substituted by halogen, oxo, —CN, —OR15, —NR15R16, —P(O)(OR15)(OR16), phenyl optionally substituted by halogen, or C1-C6 alkyl optionally substituted by halogen, —OH or oxo;
R11 and R12 are each independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl, or 3- to 6-membered heterocyclyl, wherein the C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl and 3- to 6-membered heterocyclyl are independently optionally substituted by halogen, oxo, —CN, —OR15, —NR15R16, or C1-C6 alkyl optionally substituted by halogen, —OH or oxo,
or R11 and R12 are taken together with the atom to which they attached to form a 3- to 6-membered heterocyclyl optionally substituted by halogen, oxo, —OR15, —NR15R16 or C1-C6 alkyl optionally substituted by halogen, oxo or —OH;
R13 and R14 are each independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl, or 3- to 6-membered heterocyclyl, wherein the C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C6-C14 aryl, 5- to 6-membered heteroaryl and 3- to 6-membered heterocyclyl are independently optionally substituted by halogen, oxo, —CN, —OR1, —NR15R16, or C1-C6 alkyl optionally substituted by halogen, —OH or oxo,
or R13 and R14 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl optionally substituted by halogen or C1-C6 alkyl optionally substituted by halogen or oxo;
R15 and R16 are each independently hydrogen, C1-C6 alkyl optionally substituted by halogen or oxo, C2-C6 alkenyl optionally substituted by halogen or oxo, or C2-C6 alkynyl optionally substituted by halogen or oxo,
or R15 and R16 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl optionally substituted by halogen, oxo or C1-C6 alkyl optionally substituted by oxo or halogen; and
each R17, R18, R19, and R20 is independently hydrogen or C1-C6 alkyl.
77. The method of claim 62, wherein the compound is of Formula (Ib) or Formula (Ic);
Figure US20230109104A1-20230406-C02485
78. The method of claim 62, wherein A is phenyl fused with B and is optionally substituted by R1.
79. The method of claim 62, wherein A is 5- to 6-membered heteroaryl fused with B and is optionally substituted by R1.
80. The method of claim 62, wherein A is 6-membered heteroaryl fused with B and is optionally substituted by R1.
81. The method of claim 62, wherein A is selected from the group consisting of:
Figure US20230109104A1-20230406-C02486
wherein the wavy lines denote attachment points to the parent molecule and each is fused with B and is optionally substituted by R1.
82. The method of claim 62, wherein B is C6 aryl optionally substituted by R4.
83. The method of claim 62, wherein B is 5- to 6-membered heteroaryl optionally substituted by R4.
84. The method of claim 62, wherein B is 3- to 8-membered heterocyclyl optionally substituted by R4.
85. The method of claim 62, wherein B is C3-C8 cycloalkyl optionally substituted by R4.
86. The method of claim 62, wherein the compound is of:
Figure US20230109104A1-20230406-C02487
wherein Y is hydrogen or R4;
Figure US20230109104A1-20230406-C02488
wherein Y is hydrogen or R4;
Figure US20230109104A1-20230406-C02489
wherein:
Y is hydrogen or R4;
n and p are independently 0, 1, 2, 3, or 4; and
R3a is independently hydrogen, halogen, oxo, —OR1, —NR13R14, —C(O)R13, —C(O)OR1, —C(O)NR13R14, —NR13C(O)R14, —CN, —SR 13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR1R16, —OR1 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
Figure US20230109104A1-20230406-C02490
Figure US20230109104A1-20230406-C02491
wherein:
Y is hydrogen or R4;
n and p are independently 0, 1, 2, 3, or 4; and
R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR 13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
Figure US20230109104A1-20230406-C02492
Figure US20230109104A1-20230406-C02493
wherein:
Y is hydrogen or R4;
n and p are independently 0, 1, 2, 3, or 4; and
R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR 13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
Figure US20230109104A1-20230406-C02494
wherein:
Y is hydrogen or R4;
n and p are independently 0, 1, 2, 3, or 4; and
R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR 13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
Figure US20230109104A1-20230406-C02495
wherein:
Y is hydrogen or R4;
n and p are independently 0, 1, 2, 3, or 4; and
R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR 13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
Figure US20230109104A1-20230406-C02496
wherein:
Y is hydrogen or R4;
n and p are independently 0, 1, 2, 3, or 4; and
R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR 13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
Figure US20230109104A1-20230406-C02497
Figure US20230109104A1-20230406-C02498
wherein Y is hydrogen or R4;
Figure US20230109104A1-20230406-C02499
wherein Y is hydrogen or R4;
Figure US20230109104A1-20230406-C02500
wherein:
Y is hydrogen or R4;
n and p are independently 0, 1, 2, 3, or 4; and
R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR 13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
Figure US20230109104A1-20230406-C02501
wherein:
Y is hydrogen or R4;
n and p are independently 0, 1, 2, 3, or 4; and
R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR 13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
Figure US20230109104A1-20230406-C02502
wherein:
Y is hydrogen or R4;
n and p are independently 0, 1, 2, 3, or 4; and
R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR 13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
Figure US20230109104A1-20230406-C02503
wherein:
Y is hydrogen or R4;
n and p are independently 0, 1, 2, 3, or 4; and
R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR 13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR5, —OR15 or halogen, with the proviso that R3a is not
Figure US20230109104A1-20230406-C02504
wherein:
Y is hydrogen or R4;
n and p are independently 0, 1, 2, 3, or 4; and
R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR 13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
Figure US20230109104A1-20230406-C02505
wherein:
Y is hydrogen or R4;
n and p are independently 0, 1, 2, 3, or 4; and
R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR 13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR1R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
Figure US20230109104A1-20230406-C02506
wherein Y is hydrogen or R4;
Figure US20230109104A1-20230406-C02507
or a salt thereof, wherein Y is hydrogen or R4
Figure US20230109104A1-20230406-C02508
wherein:
Y is hydrogen or R4;
n and p are independently 0, 1, 2, 3, or 4; and
R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR5, —OR15 or halogen, with the proviso that R3a is not
Figure US20230109104A1-20230406-C02509
wherein:
Y is hydrogen or R4;
n and p are independently 0, 1, 2, 3, or 4; and
R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR 13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
Figure US20230109104A1-20230406-C02510
wherein
Y is hydrogen or R4;
n and p are independently 0, 1, 2, 3, or 4; and
R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR 13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3 alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR1R16, —C(O)OR5, —OR15 or halogen, with the proviso that R3a is not
Figure US20230109104A1-20230406-C02511
wherein:
Y is hydrogen or R4;
n and p are independently 0, 1, 2, 3, or 4; and
R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR 13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
Figure US20230109104A1-20230406-C02512
wherein:
Y is hydrogen or R4;
n and p are independently 0, 1, 2, 3, or 4;
R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR 13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
Figure US20230109104A1-20230406-C02513
wherein:
Y is hydrogen or R4;
n and p are independently 0, 1, 2, 3, or 4; and
R3a is independently hydrogen, halogen, oxo, —OR13, —NR13R14, —C(O)R13, —C(O)OR13, —C(O)NR13R14, —NR13C(O)R14, —CN, —SR13, —S(O)R13, —S(O)2R13, —Si(C1-C6 alkyl)3, —P(O)R13R14, —NR13S(O)2R14, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3alkylene)C(O)R13, C3-C8 cycloalkyl optionally substituted by C1-C6 alkyl, CN, CF3, oxo, —OH, —NR15R16, —OR15 or halogen, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, —NR15R16, —C(O)OR15, —OR15 or halogen, with the proviso that R3a is not
Figure US20230109104A1-20230406-C02514
87. The method of claim 62, wherein A, B, R1 and R4 together are selected from the group consisting of:
Figure US20230109104A1-20230406-C02515
Figure US20230109104A1-20230406-C02516
Figure US20230109104A1-20230406-C02517
Figure US20230109104A1-20230406-C02518
Figure US20230109104A1-20230406-C02519
Figure US20230109104A1-20230406-C02520
Figure US20230109104A1-20230406-C02521
Figure US20230109104A1-20230406-C02522
Figure US20230109104A1-20230406-C02523
Figure US20230109104A1-20230406-C02524
Figure US20230109104A1-20230406-C02525
Figure US20230109104A1-20230406-C02526
Figure US20230109104A1-20230406-C02527
wherein the wavy lines denote attachment points to the parent molecule.
88. The method of claim 62, wherein R2 is C1-C6 alkyl.
89. The method of claim 62, wherein R2 is C3-C6 cycloalkyl.
90. The method of claim 62, wherein R2 is selected from the group consisting of: methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl,
Figure US20230109104A1-20230406-C02528
91. The method of claim 62, wherein R3 is C1-C6 alkyl, —C(O)R10, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3- to 12-membered heterocyclyl), —(C1-C3alkylene)(5- to 10-membered heteroaryl), —S(O)R10, —S(O)2R10, or 5- to 10-membered heteroaryl, each of which is optionally substituted by halogen, oxo, —OR13, —NR13R14, —C(O)R13, —CN, —S(O)R13, —S(O)2R13, —(C1-C3 alkylene)OR13, —(C1-C3 alkylene)NR13R14, —(C1-C3alkylene)C(O)R13, C3-C8 cycloalkyl, 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, or C1-C6 alkyl optionally substituted by oxo, —OH, or halogen, with the proviso that the 5- to 10-membered heteroaryl of R3 is not substituted by
Figure US20230109104A1-20230406-C02529
92. The method of claim 62, wherein R3 is 5- to 10-membered heteroaryl substituted by 3- to 12-membered heterocyclyl optionally substituted by oxo, —OH, or halogen, —CN, —(C1-C3alkylene)NR13R14, C3-C8 cycloalkyl, —C(O)R13, —NR13R14, —OR13, or C1-C6 alkyl optionally substituted by —OH or halogen, with the proviso that the 5- to 10-membered heteroaryl of R3 is not substituted by
Figure US20230109104A1-20230406-C02530
93. The method of claim 62, wherein R3 is selected from the group consisting of:
Figure US20230109104A1-20230406-C02531
Figure US20230109104A1-20230406-C02532
Figure US20230109104A1-20230406-C02533
Figure US20230109104A1-20230406-C02534
Figure US20230109104A1-20230406-C02535
Figure US20230109104A1-20230406-C02536
Figure US20230109104A1-20230406-C02537
Figure US20230109104A1-20230406-C02538
Figure US20230109104A1-20230406-C02539
Figure US20230109104A1-20230406-C02540
Figure US20230109104A1-20230406-C02541
94. The method of claim 62, wherein the compound is selected from the group consisting of Compound Nos. 1.2, 1.3, 1.6-1.8, and 1.10-3.136 in Tables 1A and 1B, or a tautomer or stereoisomer thereof, or a pharmaceutically acceptable salt of any of the foregoing.
US17/716,657 2017-10-09 2022-04-08 Heterocyclic compounds and uses thereof Abandoned US20230109104A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/716,657 US20230109104A1 (en) 2017-10-09 2022-04-08 Heterocyclic compounds and uses thereof

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201762570043P 2017-10-09 2017-10-09
US16/155,838 US11299493B2 (en) 2017-10-09 2018-10-09 Heterocyclic compounds and uses thereof
US17/716,657 US20230109104A1 (en) 2017-10-09 2022-04-08 Heterocyclic compounds and uses thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US16/155,838 Division US11299493B2 (en) 2017-10-09 2018-10-09 Heterocyclic compounds and uses thereof

Publications (1)

Publication Number Publication Date
US20230109104A1 true US20230109104A1 (en) 2023-04-06

Family

ID=65992970

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/155,838 Active US11299493B2 (en) 2017-10-09 2018-10-09 Heterocyclic compounds and uses thereof
US17/716,657 Abandoned US20230109104A1 (en) 2017-10-09 2022-04-08 Heterocyclic compounds and uses thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US16/155,838 Active US11299493B2 (en) 2017-10-09 2018-10-09 Heterocyclic compounds and uses thereof

Country Status (3)

Country Link
US (2) US11299493B2 (en)
EP (1) EP3694861A4 (en)
WO (1) WO2019074979A1 (en)

Families Citing this family (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019074981A1 (en) 2017-10-09 2019-04-18 GiraFpharma LLC Heterocyclic compounds and uses thereof
EP3762385A4 (en) 2018-03-09 2021-11-24 Recurium IP Holdings, LLC Substituted 1,2-dihydro-3h-pyrazolo[3,4-d]pyrimidin-3-ones
WO2020210375A1 (en) * 2019-04-09 2020-10-15 Nuvation Bio Inc. Heterocyclic compounds and uses thereof
EP3952876A4 (en) * 2019-04-09 2023-04-19 Nuvation Bio Inc. Heterocyclic compounds and uses thereof
EP3952875A4 (en) * 2019-04-09 2022-12-28 Nuvation Bio Inc. Heterocyclic compounds and uses thereof
WO2020210320A1 (en) * 2019-04-11 2020-10-15 Recurium Ip Holdings, Llc Substituted l,2-dihydro-3h-pyrazolo[3,4-d]pyrimidin-3-ones
CN112142748B (en) * 2019-06-28 2023-07-04 上海医药集团股份有限公司 Pyrazolopyrimidine compound, and preparation method and application thereof
BR112021026618A2 (en) * 2019-06-28 2022-06-07 Shanghai Pharmaceuticals Holding Co Ltd Pyrazolpyrimidine compound, method of preparation and applications thereof
US20220389037A1 (en) * 2019-07-04 2022-12-08 Qilu Regor Therapeutics Inc. Hpk1 inhibitors and uses thereof
IL293211A (en) * 2019-11-22 2022-07-01 Medshine Discovery Inc Pyrimidopyrrole spiro compounds and derivatives thereof as dna-pk inhibitors
JP2023507797A (en) * 2019-12-20 2023-02-27 リキュリウム アイピー ホールディングス リミテッド ライアビリティー カンパニー combination
AU2020407070A1 (en) * 2019-12-20 2022-07-14 Recurium Ip Holdings, Llc Combinations
AU2021290701A1 (en) 2020-06-17 2023-02-02 Wigen Biomedicine Technology (shanghai) Co., Ltd. Pyrazolo(3,4-d)pyrimidine-3-ketone derivative as Wee-1 inhibitor
WO2022082174A1 (en) * 2020-10-12 2022-04-21 Nuvation Bio Operating Company Inc. Heterocyclic compounds and uses thereof
US20240124440A1 (en) * 2020-12-23 2024-04-18 Sudo Biosciences Limited Tyk2 inhibitors and uses thereof
WO2022171128A1 (en) * 2021-02-09 2022-08-18 微境生物医药科技(上海)有限公司 Pyrazolo[3,4-d]pyrimidine-3-ketone derivative serving as wee-1 inhibitor
CN116848117A (en) * 2021-02-09 2023-10-03 微境生物医药科技(上海)有限公司 Fused ring compounds as Wee-1 inhibitors
WO2022171088A1 (en) * 2021-02-09 2022-08-18 微境生物医药科技(上海)有限公司 Pyrazolo[3,4-d]pyrimidin-3-one derivative
IL307230A (en) * 2021-03-26 2023-11-01 Orphagen Pharmaceuticals Inc Pyrazolopyrimidinone compounds
EP4332104A1 (en) 2021-04-30 2024-03-06 Wigen Biomedicine Technology (Shanghai) Co., Ltd. Fused ring compound as wee-1 inhibitor, and preparation method therefor and use thereof
AU2022280213A1 (en) * 2021-05-28 2023-08-31 Jiangsu Tasly Diyi Pharmaceutical Co., Ltd. Wee1 inhibitor and use thereof
WO2023280180A1 (en) * 2021-07-05 2023-01-12 微境生物医药科技(上海)有限公司 Fused ring compound as wee-1 inhibitor
WO2023016417A1 (en) * 2021-08-11 2023-02-16 微境生物医药科技(上海)有限公司 1,2-dihydro-3h-pyrazol[3,4-d]pyrimidin-3-one compound as wee-1 inhibitor
CN115838375A (en) * 2021-09-18 2023-03-24 优领医药科技(香港)有限公司 Pyrimido-dihydro-pyrazolone derivative, pharmaceutically acceptable salt thereof, and preparation method and application thereof
WO2023045942A1 (en) * 2021-09-22 2023-03-30 微境生物医药科技(上海)有限公司 1,2-dihydro-3h-pyrazole[3,4-d]pyrimidin-3-one compound serving as wee-1 inhibitor
TW202322812A (en) * 2021-11-01 2023-06-16 大陸商正大天晴藥業集團股份有限公司 Pyrazolo[3,4-d]pyrimidin-3-one compound and medical use thereof
CN114790146B (en) * 2022-05-19 2024-03-29 河南省科学院高新技术研究中心 Preparation method of 3- (methylamino) -1-aryl acetone

Family Cites Families (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6995153B2 (en) 2002-01-18 2006-02-07 Kyorin Pharmaceutical. Co., Ltd. Fused bicyclic pyrimidine derivatives
PT1631295E (en) 2003-06-06 2010-05-24 Arexis Ab Use of condensed heterocyclic compounds as scce inhibitors for the treatment of skin diseases
NZ554211A (en) 2004-09-29 2010-10-29 Portola Pharm Inc Substituted 2H-1,3-Benzoxazin-4(3H)-ones
AR060635A1 (en) 2006-04-27 2008-07-02 Banyu Pharma Co Ltd DERIVATIVES OF 1,2-DIHIDRO-3H-PIRAZOLO [3,4-D] PIRIMIDIN-3-ONA, PHARMACEUTICAL COMPOSITIONS THAT INCLUDE THEM AND THEIR USE IN THE TREATMENT OF CANCER
ES2608940T3 (en) 2007-06-15 2017-04-17 Msd K.K. Bicycloaniline derivative
AU2008315048A1 (en) * 2007-10-23 2009-04-30 Msd K.K. Pyridone-substituted-dihydropyrazolopyrimidinone derivative
US8575179B2 (en) 2008-12-12 2013-11-05 Msd K.K. Dihydropyrazolopyrimidinone derivatives
DK2477628T3 (en) * 2009-09-15 2014-11-24 Merck Sharp & Dohme Preparation of crystalline hemihydratformer of dihydropyrazolopyrimidinon
US8796289B2 (en) 2011-07-19 2014-08-05 Abbvie Inc. Pyridazino[4,5-D]pyrimidin-5(6H)-one inhibitors of kinases
JP2014530867A (en) 2011-10-20 2014-11-20 アッヴィ・インコーポレイテッド Pyridopyrimidinone inhibitor of kinase
MX2014010176A (en) 2012-02-23 2014-11-10 Abbvie Inc Pyridopyrimidinone inhibitors of kinases.
MX363243B (en) 2012-11-28 2019-03-14 Merck Sharp & Dohme Compositions and methods for treating cancer.
GB201306610D0 (en) 2013-04-11 2013-05-29 Almac Discovery Ltd Pharmaceutical compounds
WO2015019037A1 (en) 2013-08-05 2015-02-12 Almac Discovery Limited Pharmaceutical compounds
GB201322602D0 (en) 2013-12-19 2014-02-05 Almac Discovery Ltd Pharmaceutical compounds
AU2016344040B2 (en) 2015-11-01 2020-12-10 The Regents Of The University Of Colorado, A Body Corporate Wee 1 kinase inhibitors and methods of making and using the same
GB201612095D0 (en) 2016-07-12 2016-08-24 Almac Discovery Ltd Pharmaceutical compounds
GB201612092D0 (en) 2016-07-12 2016-08-24 Almac Discovery Ltd Pharmaceutical compounds
AU2017328026A1 (en) 2016-09-15 2019-05-02 Axon Enterprise, Inc. Systems and methods for recovering information from a recording device
WO2018090939A1 (en) 2016-11-16 2018-05-24 上海瑛派药业有限公司 8,9-dihydroimidazole[1,2-a]pyrimido[5,4-e]pyrimidine-5(6h)-ketone compound
JP6717457B2 (en) 2017-01-23 2020-07-01 シージャーズォアン サガシティ ニュー ドラッグ デベロップメント カンパニー リミテッド 1,2-Dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-one derivatives as Wee1 inhibitors
GB201703881D0 (en) 2017-03-10 2017-04-26 Almac Discovery Ltd Pharmaceutical compounds
CN108623615B (en) 2017-03-23 2022-12-13 上海迪诺医药科技有限公司 Macrocyclic derivatives of pyrazolo [3,4-d ] pyrimidin-3-one, pharmaceutical compositions and uses thereof
SG11201908788YA (en) 2017-03-31 2019-10-30 Seattle Genetics Inc Combinations of chk1- and wee1 - inhibitors
WO2019011228A1 (en) 2017-07-10 2019-01-17 上海瑛派药业有限公司 Imidazo[1,2-b]pyrimido[4,5-d]pyridazin-5(6h)-one compound and use thereof
WO2019028008A1 (en) 2017-08-01 2019-02-07 Zeno Royalties & Milestones, LLC 1, 2 - dihydro- 3h- pyrazolo [3, 4 - d] pyrimidin -3 - one analogs
CN109422754A (en) 2017-08-24 2019-03-05 上海迪诺医药科技有限公司 Pyrazolo [3,4-d] pyrimidine -3- ketone derivatives, its pharmaceutical composition and application
WO2019074981A1 (en) 2017-10-09 2019-04-18 GiraFpharma LLC Heterocyclic compounds and uses thereof
CN109810111B (en) 2017-11-20 2023-10-27 上海医药集团股份有限公司 Pyrazolopyrimidine compound, and preparation method and application thereof
WO2019134539A1 (en) 2018-01-05 2019-07-11 四川科伦博泰生物医药股份有限公司 Dihydropyrazolone and pyrimidine compound, preparation method and use therefor
WO2019165204A1 (en) 2018-02-23 2019-08-29 Newave Pharmaceutical Inc. 1,2-dihydro-3h-pyrazolo[3,4-d]pyrimidine-3-one compounds as inhibitors of wee-1 kinase
US20200405723A1 (en) 2018-02-28 2020-12-31 The Regents Of The University Of Colorado, A Body Corporate Wee1 kinase inhibitors and methods of treating cancer using the same
EP3762385A4 (en) 2018-03-09 2021-11-24 Recurium IP Holdings, LLC Substituted 1,2-dihydro-3h-pyrazolo[3,4-d]pyrimidin-3-ones
WO2020210375A1 (en) 2019-04-09 2020-10-15 Nuvation Bio Inc. Heterocyclic compounds and uses thereof

Also Published As

Publication number Publication date
US11299493B2 (en) 2022-04-12
EP3694861A1 (en) 2020-08-19
WO2019074979A1 (en) 2019-04-18
EP3694861A4 (en) 2021-05-19
US20190106427A1 (en) 2019-04-11

Similar Documents

Publication Publication Date Title
US11299493B2 (en) Heterocyclic compounds and uses thereof
US10807994B2 (en) Heterocyclic compounds and uses thereof
US11174252B2 (en) Heterocyclic compounds as kinase inhibitors
KR102483020B1 (en) Pyrrolotriazine compounds as TAM inhibitors
AU2009259867A1 (en) Triazolopyridine JAK inhibitor compounds and methods
US10793561B2 (en) 1,8-naphthyridinone compounds and uses thereof
US11332473B2 (en) Substituted pyrazolo[3,4-d]pyrimidines as Wee1 inhibitors
US20220220115A1 (en) Heterocyclic compounds and uses thereof
US20220273659A1 (en) Heterocyclic compounds as kinase inhibitors
US20220169646A1 (en) Heterocyclic compounds and uses thereof
US20200231589A1 (en) 1,8-naphthyridinone compounds and uses thereof
WO2022082174A1 (en) Heterocyclic compounds and uses thereof
US20210395222A1 (en) Fused Bicyclic Heterocycles as Therapeutic Agents
US20220162229A1 (en) Heterocyclic compounds and uses thereof
US20220168313A1 (en) Heterocyclic compounds and uses thereof
US11633415B2 (en) Compounds and methods useful for treating or preventing cancers
US20220169634A1 (en) Aromatic derivatives, preparation methods, and medical uses thereof

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION