WO2022261068A1 - Methods and treatment of viral infection with substituted furo-pyrimidines - Google Patents

Methods and treatment of viral infection with substituted furo-pyrimidines Download PDF

Info

Publication number
WO2022261068A1
WO2022261068A1 PCT/US2022/032464 US2022032464W WO2022261068A1 WO 2022261068 A1 WO2022261068 A1 WO 2022261068A1 US 2022032464 W US2022032464 W US 2022032464W WO 2022261068 A1 WO2022261068 A1 WO 2022261068A1
Authority
WO
WIPO (PCT)
Prior art keywords
optionally substituted
mmol
methyl
4alkyl
alkyl
Prior art date
Application number
PCT/US2022/032464
Other languages
French (fr)
Inventor
Jane RHODES
Michelle MIGHDOLL
Irene Y. Choi
Brian KOPEC
Original Assignee
Verge Analytics, 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 Verge Analytics, Inc. filed Critical Verge Analytics, Inc.
Publication of WO2022261068A1 publication Critical patent/WO2022261068A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses

Definitions

  • the present disclosure relates to methods of i) blocking alpha-coronavirus, beta- coronavirus lineage B, and beta-coronavirus lineage A into a host cell; and ii) preventing and treating an infection caused by alpha-coronavirus, beta-coronavirus lineage B, and beta- coronavirus lineage A with compounds that are phosphoinositide kinase inhibitors, in particular FYVE-type finger-containing phosphoinositide kinase (“PIKfyve”) inhibitors.
  • PIKfyve FYVE-type finger-containing phosphoinositide kinase
  • SARS-CoV-2 which is responsible for the COVID-19 (2019 novel coronavirus (2019-nCoV) disease, is an enveloped, positive-sense, RNA virus that belongs to the Betacoronavirus genus. Bouhaddou et al, Cell 182: 1-28 (2020). Improved understanding of key steps in viral entry and ways to disrupt them can lead to the development of effective antiviral drugs, not only for COVID-19, but for future viral outbreaks as well. Treatments for COVID-19 are greatly needed.
  • Coronavirus entry into susceptible cells is a complex process that requires the concerted action of receptor-binding and proteolytic processing of the coronavirus S protein to promote virus-cell fusion.
  • viral entry into cells may be mediated by a viral glycoprotein (GP), which attaches viral particles to the cell surface, delivers them to endosomes, and catalyzes fusion between viral and endosomal membranes.
  • GP viral glycoprotein
  • SARS-CoV-2 its spike (S) protein binds to an ACE2 receptor on the target cell and is subsequently primed by a serine protease, TMPRSS2, that cleaves the S protein and allows fusion of viral and lysosomal membranes.
  • TMPRSS2 serine protease
  • coronavirus membrane fusion with host cell membrane takes place within acidified endosomes. Kang, et al, PNAS 117(34):20803-20813 (2020).
  • Coronaviruses are known to interact with phosphatidylinositol (PI) kinases, which are distributed across various subcellular compartments.
  • PIKfyve PI kinase phosphatidylinositol 3-phosphate 5 kinase
  • the PIKfyve inhibition with small molecule inhibitors has been shown to inhibit SARS- CoV-2 infection. Kang et al, PNAS 117(34):20803-20813 (2020); Nelson, et al, PLoS Negl. Trop. Dis.11(4):e0005540 (2017).
  • Embodiment 1 is a method of blocking alpha-coronavirus, beta-coronavirus lineage B, and/or beta-coronavirus lineage A entry into a host cell and preventing an infection caused by alpha-coronavirus, beta-coronavirus lineage B, and beta-coronavirus lineage A, comprising administering to a subject in need thereof a compound of (i) Formula (I) wherein: R 1a and R 1b taken together with the nitrogen to which they are attached form: wherein X and Y are independently N or CR a ; wherein R a is H or C 1-4 alkyl; and R b is phenyl, monocyclic cycloalkyl, monocyclic heterocyclyl, monocyclic heterocycloalkyl, or monocyclic heteroaryl, each optionally substituted with one, two, or three R d substituents; or R 1a is H or C1-4alkyl; and R 1b is
  • Embodiment 1a is the method of embodiment 1,wherein: R 1a and R 1b taken together with the nitrogen to which they are attached form: wherein X and Y are independently N or CR a ; wherein R a is H or C 1-4 alkyl; and R b is phenyl, monocyclic cycloalkyl, monocyclic heterocyclyl, monocyclic heterocycloalkyl, or monocyclic heteroaryl, each optionally substituted with one, two, or three R d substituents; or R 1a is H or C 1-4 alkyl; and R 1b is a moncyclic heteroaryl optionally substituted with R c ; wherein R c is C1-4alkyl, phenyl, -C1-4alkyl-phenyl, monocyclic cycloalkyl, -C1-4alkyl-(monocyclic cycloalkyl), monocyclic heterocyclyl, monocyclic heterocycloalkyl, monocyclic heteroaryl, monocycl
  • Embodiment 1b is the method of embodiment 1, wherein R 1b is a monocyclic heteroaryl.
  • Embodiment 1c is the method of embodiment 1, wherein R 2 and R 3 taken together with the nitrogen to which they are attached form a heterocyclyl, optionally substituted with one, two, three, or four R j substituents, or further wherein any of the hydrogens bonded to carbon atoms are optionally replaced by deuterium.
  • Embodiment 1d is the method of embodiment 1, wherein R 4 is halo, -C(O)OH, C1-4alkylNR x R y , or -C(O)NR x R y , or is a cycloalkyl, heterocyclyl, heterocycloalkyl, phenyl or heteroaryl, wherein each cycloalkyl, heterocyclyl, heterocycloalkyl, phenyl or heteroaryl is optionally substituted with one, two, or three R z substituents
  • Embodiment 2 is the method of embodiment 1, wherein a. the alpha-coronavirus is HCoV 229E; b. the beta-coronavirus lineage B is SARS-CoV2; and c. the beta-coronavirus lineage A is HCoV OC43.
  • Embodiment 3 is the method of embodiment 2, wherein the infection is caused by SARS-CoV-2, and wherein the infection is COVID-19.
  • Embodiment 4 is the method of any one of embodiments 1 - 3, wherein R 1a and R 1b are taken together with the nitrogen to which they are attached to form .
  • Embodiment 5 is the method of any one of embodiments 1 - 3, wherein R 1a and R 1b are taken together with the nitrogen to which they are attached to form .
  • Embodiment 6 is the method of any one of embodiments 1 - 5, wherein X is N and Y is CR a .
  • Embodiment 7 is the method of any one of embodiments 1 - 5, wherein X is CR a and Y is N.
  • Embodiment 8 is the method of any one of embodiments 1 - 5, wherein X is N and Y is N.
  • Embodiment 9 is the method of any one of embodiments 1 - 8, wherein R a is H, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, or tert-butyl.
  • Embodiment 10 is the method of any one of embodiments 1 - 8, wherein R a is H or methyl.
  • Embodiment 11 is the method of any one of embodiments 1 - 8, wherein R a is H.
  • Embodiment 12 is the method of any one of embodiments 1 - 11, wherein R b is optionally substituted phenyl.
  • Embodiment 13 is the method of any one of embodiments 1 - 11, wherein R b is tolyl.
  • Embodiment 14 is the method of any one of embodiments 1 - 11, wherein R b is phenyl.
  • Embodiment 15 is the method of any one of embodiments 1 - 11, wherein R b is optionally substituted pyridinyl or pyrimidinyl.
  • Embodiment 16 is the method of any one of embodiments 1 - 11, wherein R b is optionally substituted pyridinyl.
  • Embodiment 17 is the method of any one of embodiments 1 - 11, wherein R b is substituted with one or two R d substituents.
  • Embodiment 18 is the method of any one of embodiments 1 - 11, wherein R b is methylpryridinyl, phenyl, m-tolyl, chlorophenyl, bromophenyl, methoxyphenyl.
  • Embodiment 19 is the method of any one of embodiments 1 - 18, wherein R 1a is H or C1-4alkyl; and R 1b is a 5-membered N-containing heteroaryl optionally substituted with R c .
  • Embodiment 20 is the method of any one of embodiments 1 - 18, wherein R 1a is H.
  • Embodiment 21 is the method of any one of embodiments 1 - 18, wherein R 1a is C 1- 4alkyl.
  • Embodiment 22 is the method of any one of embodiments 1 - 18, wherein R 1a is methyl.
  • Embodiment 23 is the method of any one of embodiments 1 - 22, wherein R 1b is pyrrolyl, pyrazolyl, imidazolyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, oxadiazolyl, thiadiazolyl, pyrazolopyridinyl, or indazolyl, each optionally substituted with R c .
  • Embodiment 23a is the method of any one of embodiments 1 – 22, wherein R 1b is pyrrolyl, pyrazolyl, imidazolyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, oxadiazolyl, or thiadiazolyl, each optionally substituted with R c .
  • Embodiment 24 is the method of any one of embodiments 1 - 22, wherein R 1b is pyrazolyl, imidazolyl, oxazolyl, oxadiazolyl or isoxazolyl, each optionally substituted with R c .
  • Embodiment 25 is the method of any one of embodiments 1 - 22, wherein R 1b is pyrazolyl, optionally substituted with R c .
  • Embodiment 26 is the method of any one of embodiments 1 - 22, wherein R 1b is
  • Embodiment 27 is the method of any one of embodiments 1 - 22, wherein R 1b is c .
  • Embodiment 28 is the method of any one of embodiments 1 - 27, wherein R c is optionally substituted C1-4alkyl.
  • Embodiment 29 is the method of any one of embodiments 1 - 27, wherein R c is methyl, ethyl, isopropyl, or trifluoromethyl.
  • Embodiment 30 is the method of any one of embodiments 1 - 27, wherein R c is optionally substituted phenyl.
  • Embodiment 31 is the method of any one of embodiments 1 - 27, wherein R c is phenyl or o-, m-, p-tolyl, fluorophenyl, methoxyphenyl, or trifluoromethoxyphenyl.
  • Embodiment 32 is the method of any one of embodiments 1 - 27, wherein R c is phenyl.
  • Embodiment 33 is the method of any one of embodiments 1 - 27, wherein R c is optionally substituted monocyclic cycloalkyl.
  • Embodiment 34 is the method of any one of embodiments 1 - 27, wherein R c is optionally substituted cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.
  • Embodiment 35 is the method of any one of embodiments 1 - 27, wherein R c is optionally substituted cyclopropyl.
  • Embodiment 36 is the method of any one of embodiments 1 - 27, wherein R c is optionally substituted monocyclic heterocycloalkyl.
  • Embodiment 37 is the method of any one of embodiments 1 - 27, wherein R c is optionally substituted cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, or cyclohexylmethyl.
  • Embodiment 38 is the method of any one of embodiments 1 - 27, wherein R c is optionally substituted monocyclic heterocyclyl.
  • Embodiment 39 is the method of any one of embodiments 1 - 27, wherein R c is optionally substituted pyrrolidinyl, tetrahydrofuranyl, piperidinyl, morpholinyl, or piperazinyl.
  • Embodiment 40 is the method of any one of embodiments 1 - 27, wherein R c is optionally substituted monocyclic heteroaryl.
  • Embodiment 41 is the method of any one of embodiments 1 - 27, wherein R c is optionally substituted pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, furanyl, thiophenyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, or pyridazinyl.
  • Embodiment 42 is the method of any one of embodiments 1 - 27, wherein R c is optionally substituted pyrazole, thiophenyl, imidazolyl, pyridinyl, or pyrimidinyl.
  • Embodiment 43 is the method of any one of embodiments 1 - 27, wherein R c is optionally substituted pyrazolyl.
  • Embodiment 44 is the method of any one of embodiments 1 - 27, wherein R c is optionally substituted pyridinyl.
  • Embodiment 45 is the method of any one of embodiments 1 - 27, wherein R c is methylpyridinyl.
  • Embodiment 46 is the method of any one of embodiments 1 - 27, wherein R c is optionally substituted -C1-4alkyl-phenyl, -C1-4alkyl-(monocyclic cycloalkyl), monocyclic heterocycloalkyl, or -C1-4alkyl-(monocyclic heteroaryl).
  • Embodiment 48 is the method of any one of embodiments 1 - 47, wherein each R d substituent is independently C1-4alkyl, -O-C1-4alkyl, C1-4haloalkyl, or halo.
  • Embodiment 49 is the method of any one of embodiments 1 - 47, wherein each R d substituent is independently methyl, ethyl, isopropyl, -CF 3 , -OCH 3 , -OCF 3 , or fluoro.
  • Embodiment 50 is the method of any one of embodiments 1 - 49, wherein R g and R h are each independently H or methyl.
  • Embodiment 51 is the method of any one of embodiments 1 - 50, wherein each of R 2 and R 3 are independently selected from H, pyrrolidinyl, piperidinyl, and piperazinyl, wherein each pyrrolidinyl, piperidinyl, and piperazinyl is optionally substituted with one R j substituent.
  • Embodiment 52 is the method of any one of embodiments 1 - 50, wherein R 2 and R 3 taken together with the nitrogen to which they are attached form pyrrolidinyl, piperidinyl, piperazinyl, morpholino, or thiomorpholino, each optionally substituted with one, two, three, or four R j substituents.
  • Embodiment 53 is the method of any one of embodiments 1 - 50, wherein R 2 and R 3 taken together with the nitrogen to which they are attached form morpholino or piperazinyl, optionally substituted with one, two, three, or four R j substituents.
  • Embodiment 54 is the method of any one of embodiments 1 - 50, wherein R 2 and R 3 taken together with the nitrogen to which they are attached form 2,2,6,6-tetrafluoro-morpholino, morpholino-2-one, morpholino-3-one, piperazinyl-2-one, piperazinyl-3-one, thi
  • Embodiment 55 is the method of any one of embodiments 1 - 50, wherein each R j substituent is independently methyl, oxo, hydroxy, NH 2 , -OCH 3 , halo, -CF 3 , or -OCF 3 .
  • Embodiment 56 is the method of any one of embodiments 1 - 50, wherein R 2 and R 3 taken together with the nitrogen to which they are attached form morpholino in which 1 to 8 hydrogens are replaced with deuterium.
  • Embodiment 57 is the method of any one of embodiments 1 - 56, wherein R k and R l are each independently H or methyl.
  • Embodiment 58 is the method of any one of embodiments 1 - 57, wherein R 4 is H.
  • Embodiment 59 is the method of any one of embodiments 1 - 57, wherein R 4 is chloro.
  • Embodiment 60 is the method of any one of embodiments 1 - 57, wherein R 4 is optionally substituted phenyl.
  • Embodiment 61 is the method of any one of embodiments 1 - 57, wherein R 4 is optionally substituted heteroaryl.
  • Embodiment 62 is the method of any one of embodiments 1 - 57, wherein R 4 is optionally substituted monocyclic heteroaryl.
  • Embodiment 63 is the method of any one of embodiments 1 - 57, wherein R 4 is optionally substituted pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, furanyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, or pyridazinyl.
  • Embodiment 64 is the method of any one of embodiments 1 - 57, wherein R 4 is or each optionally substituted with 1 or 2 R z groups.
  • Embodiment 65 is the method of any one of embodiments 1 - 57, wherein R 4 is optionally substituted pyridinyl or pyrimidinyl.
  • Embodiment 66 is the method of any one of embodiments 1 - 57, wherein R 4 is optionally substituted pyridinyl.
  • Embodiment 67 is the method of any one of embodiments 1 - 57, wherein R 4 is pyridinyl.
  • Embodiment 68 is the method of any one of embodiments 1 - 57, wherein R 4 is optionally substituted pyrazolyl.
  • Embodiment 69 is the method of any one of embodiments 1 - 57, wherein R 4 is optionally substituted with one or two R z substituents.
  • Embodiment 70 is the method of any one of embodiments 1 - 57, wherein R 4 is pyrazolyl optionally substituted with one or two R z substituents.
  • Embodiment 71 is the method of any one of embodiments 1 - 57, wherein R 4 is phenyl or pyridyl, each optionally substituted with one or two substituents selected from C 1-4 alkyl, - CF 3 , fluoro, chloro, -OCH 3 , and -OCF 3 .
  • Embodiment 72 is the method of any one of embodiments 1 - 57, wherein R 4 is heterocyclyl, optionally substituted with one or two R z substituents.
  • Embodiment 73 is the method of any one of embodiments 1 - 57, wherein R 4 is pyrrolidinyl, piperidinyl, piperazinyl, morpholino, or thiomorpholino, optionally substituted with one or two R z substituents.
  • Embodiment 74 is the method of any one of embodiments 1 - 57, wherein R 4 is heterocycloalkyl, optionally substituted with one or two R z substituents.
  • Embodiment 75 is the method of any one of embodiments 1 - 57, wherein R 4 is pyrrolidinylmethyl, piperidinylmethyl, piperazinylmethyl, morpholinomethyl, or thiomorpholinomethyl, optionally substituted with one or two R z substituents.
  • Embodiment 76 is the method of any one of embodiments 1 - 57, wherein R 4 is 3- methyl-1H-pyrazol-5-yl, 3-methylisothiazol-5-yl, 2-methyl-1H-imidazol-5-yl, 1-methyl-pyrazol- 4-yl, 1-methylpyrazol-3-yl, 1-((1-acetamido)-eth-2-yl)-5-methyl-pyrazol-3-yl, 1-((1- chloromethylamido)-eth-2-yl)-5-methyl-pyrazol-3-yl, 1-((1-acrylamido)-eth-2-yl)-5-methyl- pyrazol-3-yl, thiazol-2-yl, pyrazol-4-yl, pyrazol-1-yl, oxazol-2-yl, 3-(1-N,N-dimethyl-eth-2-yl)- 4-methyl-pyrazol-1-yl, or
  • Embodiment 77 is the method of any one of embodiments 1 - 57, wherein R 4 is C1- 4alkylNR x R y .
  • Embodiment 78 is the method of any one of embodiments 1 - 57, wherein R 4 is CH 2 NR x R y .
  • Embodiment 79 is the method of any one of embodiments 1 - 57, wherein R 4 is - C(O)NR x R y .
  • Embodiment 80 is the method of any one of embodiments 1 - 79, wherein R x is H.
  • Embodiment 81 is the method of any one of embodiments 1 - 79, wherein R x is methyl or ethyl, optionally substituted with one, two, or three R o substituents.
  • Embodiment 82 is the method of any one of embodiments 1 - 79, wherein R x is methyl.
  • Embodiment 83 is the method of any one of embodiments 1 - 82, wherein R y is H.
  • Embodiment 84 is the method of any one of embodiments 1 - 82, wherein R y is C1- 4alkyl, optionally substituted with one, two, or three R o substituents.
  • Embodiment 85 is the method of any one of embodiments 1 - 82, wherein R y is methyl, ethyl, propyl, or isopropyl, each optionally substituted with one, two, or three R o substituents.
  • Embodiment 86 is the method of any one of embodiments 1 - 82, wherein R y is H, methyl, ethyl, methyoxy, or methoxyethyl.
  • Embodiment 87 is the method of any one of embodiments 1 - 82, wherein R y is methyl.
  • Embodiment 88 is the method of any one of embodiments 1 - 82, wherein R y is -SO2- R r or C 1-4 alkyl-SO 2 -R r .
  • Embodiment 89 is the method of any one of embodiments 1 - 82, wherein R y is -SO 2 - R r , C1-4alkyl-SO2-R r ; and R r is CH3 or NH 2 , NHCH3, or N(CH3)2.
  • Embodiment 90 is the method of any one of embodiments 1 - 82, wherein R y is -SO2- methyl, C 2-4 alkyl-SO 2 -N(CH 3 ) 2 .
  • Embodiment 91 is the method of any one of embodiments 1 - 82, wherein R y is monocyclic cycloalkyl or -C1-2alkyl(monocyclic cycloalkyl), each optionally substituted with one, two, or three R o substituents.
  • Embodiment 92 is the method of any one of embodiments 1 - 82, wherein R y is monocyclic cycloalkyl, optionally substituted with one, two, or three R o substituents.
  • Embodiment 93 is the method of any one of embodiments 1 - 82, wherein R y is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl, each optionally substituted with one, two, or three R o substituents.
  • Embodiment 94 is the method of any one of embodiments 1 - 82, wherein R y is cyclopropyl.
  • Embodiment 95 is the method of any one of embodiments 1 - 82, wherein R y is cyclopropyl, cyclobutyl, cyclopentyl, cyclopropylmethyl, 1-cyclopropylethyl, 2- cyclopropylethyl, cyclobutylmethyl, or cyclopentylmethyl.
  • Embodiment 96 is the method of any one of embodiments 1 - 82, wherein R y is monocyclic heterocyclyl, optionally substituted with one, two, or three R o substituents.
  • Embodiment 97 is the method of any one of embodiments 1 - 82, wherein R y is optionally substituted azetidinyl, oxetanyl, pyrrolidinyl, piperidinyl, morpholinyl, piperazinyl, azepanyl, azocanyl, tetrahydrofuranyl, or tetrahydropyranyl, optionally substituted with methyl.
  • Embodiment 98 is the method of any one of embodiments 1 - 82, wherein R y is monocyclic heterocycloalkyl, optionally substituted with one, two, or three R o substituents.
  • Embodiment 99 is the method of any one of embodiments 1 - 82, wherein R y is optionally substituted azetidinylmethyl, oxetanylmethyl, pyrrolidinylmethyl, piperidinylmethyl, morpholinylmethyl, or piperazinylmethyl, optionally substituted with methyl.
  • Embodiment 100 is the method of any one of embodiments 1 - 79, wherein one of R x and R y is H and the other is -CH3.
  • Embodiment 101 is the method of any one of embodiments 1 - 79, wherein both of R x and R y is H.
  • Embodiment 102 is the method of any one of embodiments 1 - 79, wherein both of R x and R y is -CH 3 .
  • Embodiment 103 is the method of any one of embodiments 1 - 79, wherein R x and R y taken together with the nitrogen to which they are attached form a monocyclic heterocyclyl, optionally substituted with C1-4alkyl.
  • Embodiment 104 is the method of any one of embodiments 1 - 79, wherein R x and R y are taken together with the nitrogen to which they are attached to form azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, or thiomorpholinyl, each optionally substituted with methyl.
  • Embodiment 105 is the method of any one of embodiments 1 - 104, wherein each R z is independently C1-4alkyl, halo, -OH, or -OC1-4alkyl, wherein each alkyl is optionally substituted with -NR m R n .
  • Embodiment 106 is the method of any one of embodiments 1 - 104, wherein each R z is independently -CH3, -OH, halo, or -OCH3.
  • Embodiment 107 is the method of any one of embodiments 1 - 104, wherein R z is C 2- 4 alkyl substituted with -NR m R n or OCH 3 .
  • Embodiment 108 is the method of any one of embodiments 1 - 104, wherein each R z substituent is independently -NR p R q , -C(O)NR p R q .
  • Embodiment 109 is the method of any one of embodiments 1 - 104, wherein each R z substituent is methyl, ethyl, isopropyl, -CF3, fluoro, chloro, -OCH3, -OCF3, methylamino, ethylamino, propylamino, butylamino, aminomethyl, aminoethyl, aminopropyl, aminobutyl, dimethylamino, dimethylaminomethyl, dimethylaminoethyl, dimethylaminopropyl, dimethylaminobutyl, -C(O)methylamino, -C(O)ethylamino, -C(O)propylamino, - C(O)butylamino, -C(O)dimethylamino, -C(O)dimethylaminomethyl, -C(O)dimethylaminoethyl, -C(O)d
  • Embodiment 110 is the method of any one of embodiments 1 - 109, wherein R m and R n are each independently H, C1-4alkyl, C(O)CH3, C(O)CH 2 Cl, or C(O)CH 2 CH 2 .
  • Embodiment 111 is the method of any one of embodiments 1 - 109, wherein R m and R n are each H.
  • Embodiment 112 is the method of any one of embodiments 1 - 109, wherein R m and R n are each methyl.
  • Embodiment 113 is the method of any one of embodiments 1 - 109, wherein R m and R n taken together with the nitrogen to which they are attached form a monocyclic heterocyclyl, optionally substituted with one or two R o substituents.
  • Embodiment 114 is the method of any one of embodiments 1 - 109, wherein R m and R n taken together with the nitrogen to which they are attached form pyrrolidinyl, piperidinyl, piperazinyl, morpholino, thiomorpholino, or thiomorpholino-1,1-dioxide, each optionally substituted with one or two R o substituents.
  • Embodiment 115 is the method of any one of embodiments 1 - 109, wherein R m and R n taken together with the nitrogen to which they are attached form pyrrolidinyl, piperidinyl, piperazinyl, or morpholino, each optionally substituted with methyl.
  • Embodiment 116 is the method of any one of embodiments 1 - 115, wherein each R o substituent is C 1-4 alkyl, or -NR p R q .
  • Embodiment 117 is the method of any one of embodiments 1 - 116, wherein R p and R q are each independently H, methyl, C1-4alkylNH 2 , C1-4alkylNHCH3, or C1-4alkylN(CH3)2.
  • Embodiment 118 is the method of any one of embodiments 1 - 116, wherein R p and R q are each independently H or methyl.
  • Embodiment 119 is the method of any one of embodiments 1 - 118, wherein R 5 is H, methyl, ethyl, chloro, bromo, fluoro, -OH, or -OCH 3 .
  • Embodiment 120 is the method of any one of embodiments 1 - 118, wherein R 5 is H.
  • Embodiment 121 is the method of any one of embodiments 1 - 3, wherein R c1 is phenyl or pyridyl, each optionally substituted with methyl, -CF3, Cl, Br, or OCH3.
  • Embodiment 122 is the method of any one of embodiments 1 - 3, wherein R c1 is phenyl.
  • Embodiment 123 is the method of any one of embodiments 1 - 3, wherein R c1 is tolyl.
  • Embodiment 124 is the method of any one of embodiments 1 - 3, wherein R c1 is pyridyl optionally substituted with methyl or -CF 3 .
  • Embodiment 125 is the method of any one of embodiments 1 - 3, wherein R 4a is pyridyl, optionally substituted with one or two R z groups.
  • Embodiment 126 is the method of any one of embodiments 1 - 3, wherein R 4a is pyridyl.
  • Embodiment 127 is the method of any one of embodiments 1 - 3, wherein R 4a is pyrazolyl optionally substituted with one or two R z groups.
  • Embodiment 128 is the method of any one of embodiments 1 - 3, wherein R 4a is 3- methyl-1H-pyrazol-5-yl, 3-methylisothiazol-5-yl, 2-methyl-1H-imidazol-5-yl, 1-methyl-pyrazol- 4-yl, 1-methylpyrazol-3-yl, 1-((1-acetamido)-eth-2-yl)-5-methyl-pyrazol-3-yl, 1-((1- chloromethylamido)-eth-2-yl)-5-methyl-pyrazol-3-yl, 1-((1-acrylamido)-eth-2-yl)-5-methyl- pyrazol-3-yl, thiazol-2-yl, pyrazol-4-yl, pyrazol-1-yl, oxazol-2-yl, or 3-(1-N,N-dimethyl-eth-2- yl)-4-methyl-pyrazol-1-yl.
  • Embodiment 129 is the method of any one of embodiments 1 - 3, wherein R 4a is - C(O)NR x R y wherein R x is H or C 1-4 alkyl and R y is H, C 1-4 alkyl, -O-C 1-4 alkyl, -SO 2 -R r , C1-4alkyl-SO2-R r monocyclic cycloalkyl, -C1-4alkyl(monocyclic cycloalkyl), monocyclic heterocyclyl, or monocyclic heterocycloalkyl, each optionally substituted with one, two, or three R o substituents; and R r and R o are as defined herein.
  • Embodiment 130 is the method of any one of embodiments 1 - 3, wherein R 4a is - C(O)NR x R y wherein R x is H or methyl; and R y is H, methyl, ethyl, butyl, isopropyl, methoxy, - SO 2 -methyl, C 2-4 alkyl-SO 2 -methyl, C 2-4 alkyl-SO 2 -N(CH 3 ) 2 , cyclopropyl, cyclobutyl, cyclopentyl, cyclopropylmethyl, 1-cyclopropylethyl, 2-cyclopropylethyl, cyclobutylmethyl, cyclopentylmethyl, azetidinyl, oxetanyl, pyrrolidinyl, piperidinyl, morpholinyl, piperazinyl, azepanyl, azocanyl, tetrahydrofuranyl, t
  • Embodiment 131 is the method of any one of the preceding embodiments, wherein the compound is selected from a compound of Table 1 or a pharmaceutically acceptable salt thereof.
  • Embodiment 132 is the method of any one of embodiments 1 to 131, wherein one or more hydrogen atoms attached to carbon atoms of the compound are replaced by deuterium atoms.
  • Embodiment 133 is the method of any one of the preceding embodiments, wherein the compound and/or the pharmaceutically acceptable salt is in a pharmaceutical composition.
  • Embodiment 134 is the method of embodiment 133, wherein the pharmaceutical composition further comprises a pharmaceutically acceptable excipient.
  • Figure 1 shows antiviral effect and cell toxicity data for Compound 101 in Vero-E6 cells. Concentration-dependent antiviral effect (i.e., antiviral activity) is shown as percent cell survival (i.e., percent inhibition) on the left axis (data represented by black squares with a solid line). Concentration-dependent cell toxicity is shown as percent cell survival on the right axis (data represented by white squares with a dotted line).
  • Figure 2 shows antiviral effects of Compound 97 A549-ACE2 cells. Viral titer is shown as Log10 of plaque forming units per mL (PFU/mL) on the left axis (data represented by white circles).
  • FIG. 3A-B show the effects of Compounds 91 and 121 on human alphacoronavirus strain 229E (HCoV 229E).
  • HCV 229E Human bronchial epithelial (16BHE) cells infected with HCoV 229E were compared to uninfected 16HBE cells.
  • Figure 3A shows data for Compound 91;
  • Figure 3B shows data for Compound 121.
  • Figures 4A-C show the effects of Compounds 91, 114 and 121 on human betacoronavirus strain OC43 (HCoV OC43).
  • Human lung mucoepidermoid (H 2 92) cells infected HCoV OC43 were compared to uninfected H 2 92 cells.
  • Figure 4A shows data for Compound 91;
  • Figure 4B shows data for Compound 114;
  • Figure 4C shows data for Compound 121.
  • Figures 5A-C show the activity of Compound 114 (Fig.5A), Compound 163 (Fig.5B), and the remdesivir control (Fig.5C) in a Vero-E6-SARS-CoV-2 cytopathic assay against two SARS-CoV2 strains, Wildtype (WT) and Delta (D). DETAILED DESCRIPTION OF THE INVENTION [0147]
  • the present disclosure provides methods and compositions for the treatment of certain coronavirus infections.
  • the present disclosure provides methods and compositions for blocking alpha-coronavirus, beta-coronavirus lineage B, and/or beta- coronavirus lineage A into a host cell with compounds that are phosphoinositide kinase inhibitors, in particular FYVE-type finger-containing phosphoinositide kinase (“PIKfyve”) inhibitors.
  • the present disclosure provides methods and compositions for preventing and/or treating an infection caused by alpha-coronavirus, beta-coronavirus lineage B, or a beta-coronavirus lineage A with compounds that are phosphoinositide kinase inhibitors, in particular PIKfyve inhibitors.
  • “about” or “approximately” can mean a range of up to 10% (i.e., ⁇ 10%) or more depending on the limitations of the measurement system. For example, about 5 mg can include any number between 4.5 mg and 5.5 mg. Furthermore, particularly with respect to biological systems or processes, the terms can mean up to an order of magnitude or up to 5-fold of a value. When particular values or compositions are provided in the instant disclosure, unless otherwise stated, the meaning of “about” or “approximately” should be assumed to be within an acceptable error range for that particular value or composition. “Or” is used in the inclusive sense, i.e., equivalent to “and/or,” unless the context requires otherwise.
  • the term “and/or” as used in a phrase such as “A, B, and/or C” is intended to encompass each of the following aspects: A, B, and C; A, B, or C; A or C; A or B; B or C; A and C; A and B; B and C; A (alone); B (alone); and C (alone).
  • the terms “or a combination thereof” and “or combinations thereof” as used herein refers to any and all permutations and combinations of the listed terms preceding the term.
  • A, B, C, or combinations thereof is intended to include at least one of: A, B, C, AB, AC, BC, or ABC, and if order is important in a particular context, also BA, CA, CB, ACB, CBA, BCA, BAC, or CAB.
  • expressly included are combinations that contain repeats of one or more item or term, such as BB, AAA, AAB, BBC, AAABCCCC, CBBAAA, CABABB, and so forth.
  • BB Biller Identifier
  • AAA AAA
  • AAB AAA
  • CBA BCA
  • BAC BAC
  • CAB CAB
  • expressly included are combinations that contain repeats of one or more item or term, such as BB, AAA, AAB, BBC, AAABCCCC, CBBAAA, CABABB, and so forth.
  • the skilled artisan will understand that typically there is no limit on the number of items or terms in any combination, unless otherwise apparent from the context.
  • subject and “patient” as used herein refers to human and non-human animals, including vertebrates, mammals and non-mammals.
  • the subject can be human, non-human primates, simian, ape, murine (e.g., mice and rats), bovine, porcine, equine, canine, feline, caprine, lupine, ranine or piscine.
  • administering refers to the physical introduction of an agent to a subject, using any of the various methods and delivery systems known to those skilled in the art.
  • Exemplary routes of administration for the formulations disclosed herein include intravenous, intramuscular, subcutaneous, intraperitoneal, spinal or other parenteral routes of administration, for example by injection or infusion.
  • parenteral administration means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intralymphatic, intralesional, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal injection and infusion, as well as in vivo electroporation.
  • the formulation is administered via a non-parenteral route, e.g., orally.
  • non-parenteral routes include a topical, epidermal or mucosal route of administration, for example, intranasally, vaginally, rectally, sublingually or topically.
  • Administering can also be performed, for example, once, a plurality of times, and/or over one or more extended periods.
  • the terms “treat,” “treating,” and “treatment,” as used herein, covers any administration or application of a therapeutic for disease/disorder in a subject, and includes inhibiting the disease/disorder, arresting its development, relieving one or more symptoms of the disease/disorder, or curing the disease/disorder.
  • prevent means inhibiting or arresting development of a disease/disorder in a subject deemed to be disease/disorder free.
  • block and “blocking” as used herein with reference to viral entry into a host cell refers to stopping the entry of some or all of a virus, such as a coronavirus, into a host cell or host cells. The blocking may be complete or partial. Partial blocking includes preventing at least some virus from entering host cells, for example, enough virus to prevent the subject from displaying at least one symptom associated with such viral infection.
  • a “pharmaceutically acceptable vehicle” for therapeutic purposes is a physical embodiment that can be administered to a subject.
  • Pharmaceutically acceptable vehicles include pills, capsules, caplets, tablets, oral fluids, injection fluids, sprays, aerosols, troches, dietary supplements, creams, lotions, oils, solutions, pastes, powders, steam, Or it may be a liquid, but is not limited to these.
  • a pharmaceutically acceptable vehicle is a buffered isotonic solution such as phosphate buffered saline (PBS).
  • PBS phosphate buffered saline
  • Alkyl means a linear saturated monovalent hydrocarbon radical of one to six carbon atoms or a branched saturated monovalent hydrocarbon radical of three to six carbon atoms, e.g., methyl, ethyl, propyl, 2-propyl, butyl (including all isomeric forms), pentyl (including all isomeric forms), and the like.
  • Alkylene means a linear saturated divalent hydrocarbon radical of one to six carbon atoms or a branched saturated divalent hydrocarbon radical of three to six carbon atoms unless otherwise stated e.g., methylene, ethylene, propylene, 1-methylpropylene, 2-methylpropylene, butylene, pentylene, and the like.
  • Alkylsulfonyl means a –SO 2 R radical where R is alkyl as defined above, e.g., methylsulfonyl, ethylsulfonyl, and the like.
  • Amino means a -NH 2 .
  • Alkoxy means a -OR radical where R is alkyl as defined above, e.g., methoxy, ethoxy, propoxy, or 2-propoxy, n-, iso-, or tert-butoxy, and the like.
  • Alkoxyalkyl means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with an alkoxy group, (in one embodiment one or two alkoxy groups), as defined above, e.g., 2-methoxyethyl, 1-, 2-, or 3-methoxypropyl, 2-ethoxyethyl, and the like.
  • Alkoxycarbonyl means a -C(O)OR radical where R is alkyl as defined above, e.g., methoxycarbonyl, ethoxycarbonyl, and the like.
  • Acyl means a -COR radical where R is alkyl, haloalkyl, or cycloalkyl, e.g., acetyl, propionyl, cyclopropylcarbonyl, and the like. When R is alkyl, the radical is also referred to herein as alkylcarbonyl.
  • Cycloalkyl means a cyclic saturated monovalent hydrocarbon radical of three to ten carbon atoms wherein one or two carbon atoms may be replaced by an oxo group, e.g., cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl, and the like.
  • Carboxy means –COOH.
  • a “coronavirus,” “corona respiratory virus,” and “CoV” are used interchangeably herein to refer to a virus belonging to the family Coronaviridae. Coronaviruses are enveloped, positive-sense RNA viruses of approximately 31 Kb, making these viruses the largest known RNA viruses.
  • Coronaviruses infect a variety of host species, including humans and several other vertebrates. These viruses predominantly cause respiratory and intestinal tract infections and induce a wide range of clinical manifestations. In general, coronaviruses can be classified into low pathogenic CoVs (including human CoVs (hCoVs)) and highly pathogenic CoVs, such as severe acute respiratory syndrome CoV (SARS-CoV) and Middle East respiratory syndrome CoV (MERS-CoV). Low pathogenic hCoV infect upper airways and cause seasonal mild to moderate cold-like respiratory illnesses in healthy individuals.
  • hCoVs human CoVs
  • SARS-CoV severe acute respiratory syndrome CoV
  • MERS-CoV Middle East respiratory syndrome CoV
  • hCoV pathogenic hCoV
  • ALI acute lung injury
  • ARDS acute respiratory distress syndrome
  • Haloalkyl means alkyl radical as defined above, which is substituted with one or one to five halogen atoms (in one embodiment fluorine or chlorine,) including those substituted with different halogens, e.g., -CH 2 Cl, -CF 3 , -CHF 2 , -CH 2 CF 3 , -CF 2 CF 3 , -CF(CH 3 ) 2 , and the like.
  • halogen atoms in one embodiment fluorine or chlorine,
  • Cx-y-haloalkyl “Cx-y” means the number of carbon atoms in the alkyl group ranges from x to y.
  • Haloalkoxy means a –OR radical where R is haloalkyl as defined above e.g., -OCF3, -OCHF 2 , and the like. When R is haloalkyl where the alkyl is substituted with only fluoro, it can be referred to in this disclosure as fluoroalkoxy.
  • “Hydroxyalkyl” means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with one or two hydroxy groups, provided that if two hydroxy groups are present they are not both on the same carbon atom.
  • Representative examples include, but are not limited to, hydroxymethyl, 2-hydroxyethyl, 2-hydroxypropyl, 3-hydroxypropyl, 1-(hydroxymethyl)-2-methylpropyl, 2-hydroxybutyl, 3-hydroxybutyl, 4-hydroxybutyl, 2,3-dihydroxypropyl, 1-(hydroxymethyl)-2- hydroxyethyl, 2,3-dihydroxybutyl, 3,4-dihydroxybutyl and 2-(hydroxymethyl)-3-hydroxypropyl.
  • Further examples include, but are not limited to, 2-hydroxyethyl, 2,3-dihydroxypropyl, and 1- (hydroxymethyl)-2-hydroxyethyl.
  • Heterocyclyl means a saturated or unsaturated monovalent monocyclic or bi-cyclic group (fused bi-cyclic or bridged bi-cyclic) of 4 to 10 ring atoms in which one or two ring atoms are heteroatom selected from N, O, and S(O)n, where n is an integer from 0 to 2, the remaining ring atoms being C. Additionally, one or two ring carbon atoms in the heterocyclyl ring can optionally be replaced by a –CO- group.
  • heterocyclyl includes, but is not limited to, oxetanyl, pyrrolidino, piperidino, homopiperidino, 2-oxopyrrolidinyl, 2-oxopiperidinyl, morpholino, piperazino, tetrahydropyranyl, thiomorpholino, hexahydropyrrolo[1,2-a]pyrazin-6(2H)-one-yl, tetrahydro-1H-oxazolo[3,4-a]pyrazin-3(5H)-one- yl, 5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyrazine-yl, 3-oxa-8-azabicyclo[3.2.1]octane-yl, and the like.
  • heterocyclylalkyl and “heterocycloalkyl” mean an –(alkylene)-R radical where R is heterocyclyl ring as defined above e.g., tetraydrofuranylmethyl, piperazinylmethyl, morpholinylethyl, and the like.
  • Heterocycloamino means a saturated or unsaturated monovalent monocyclic group of 4 to 8 ring atoms in which one or two ring atoms are heteroatom selected from N, O, or S(O)n, where n is an integer from 0 to 2, the remaining ring atoms being C provided that at least one of the ring atoms is N. Additionally, one or two ring carbon atoms in the heterocycloamino ring can optionally be replaced by a –CO- group. When the heterocycloamino ring is unsaturated it can contain one or two ring double bonds provided that the ring is not aromatic.
  • Heterocycloaminoalkyl means a –(alkylene)-R radical where R is heterocycloamino as described above.
  • Heteroaryl means a monovalent monocyclic or bicyclic aromatic radical of 5 to 10 ring atoms where one or more, (in one embodiment one, two, or three), ring atoms are heteroatom selected from N, O, and S, the remaining ring atoms being carbon.
  • Representative examples include, but are not limited to, pyrrolyl, thienyl, thiazolyl, imidazolyl, furanyl, indolyl, isoindolyl, oxazolyl, isoxazolyl, benzothiazolyl, benzoxazolyl, quinolinyl, isoquinolinyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, tetrazolyl, pyrazolopyridinyl, indazolyl, furopyrimidinyl, and the like.
  • “Optional” or “optionally” means that the subsequently described event or circumstance may but need not occur, and that the description includes instances where the event or circumstance occurs and instances in which it does not.
  • “heterocyclyl group optionally substituted with an alkyl group” means that the alkyl may but need not be present, and the description includes situations where the heterocyclyl group is substituted with an alkyl group and situations where the heterocyclyl group is not substituted with alkyl.
  • phrases “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable excipient or “pharmaceutically acceptable carrier” as used herein means a pharmaceutically acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material. Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient.
  • materials which can serve as pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide;
  • PIKfyve inhibitor refers to a molecule that inhibits phosphatidylinositol 3-phosphate 5-kinase (PIKfyve).
  • salt or “pharmaceutically acceptable salt” refers to salts derived from a variety of organic and inorganic counter ions well known in the art. Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids. Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
  • Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like.
  • Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases.
  • Inorganic bases from which salts can be derived include, for example, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like.
  • Organic bases from which salts can be derived include, for example, primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like, specifically such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, and ethanolamine.
  • the pharmaceutically acceptable base addition salt is chosen from ammonium, potassium, sodium, calcium, and magnesium salts. It is understood that the pharmaceutically acceptable salts are non-toxic.
  • compositions [0190] Provided herein are methods of treating an infection caused by a virus, inhibiting entry of a virus into a host cell, or inhibiting fusion of viral membrane with a host cell membrane comprising administering to a subject in need thereof a compound of Formula (I) or any of the embodiments thereof (e.g., compounds of Table 1) described herein.
  • the disclosure relates to a method of blocking alpha-coronavirus entry into a host cell and preventing an infection caused by alpha-coronavirus, comprising administering to a subject in need thereof a compound of Formula (I) or any of the embodiments thereof described herein.
  • the alpha-coronavirus is HCoV 229E.
  • the disclosure relates to a method of blocking beta-coronavirus lineage B entry into a host cell and preventing an infection caused by beta-coronavirus lineage B, comprising administering to a subject in need thereof a compound of Formula (I) or any of the embodiments thereof described herein.
  • the beta-coronavirus lineage B is SARS-CoV2.
  • the infection caused by SARS-CoV-2 is COVID-19.
  • the methods treat or prevent COVID-19 infection.
  • the disclosure relates to a method of blocking beta-coronavirus lineage A entry into a host cell and preventing an infection caused by beta-coronavirus lineage A, comprising administering to a subject in need thereof a compound of Formula (I) or any of the embodiments thereof described herein.
  • the beta-coronavirus lineage A is HCoV OC43.
  • a method of inhibiting coronavirus viral membrane fusion with an early endosomal membrane comprising administering a compound of Formula (I) or any of the embodiments thereof (e.g., compounds of Table 1) described herein.
  • a method of inhibiting coronavirus viral membrane fusion with a maturing endosomal membrane comprising administering a compound of Formula (I) or any of the embodiments thereof (e.g., compounds of Table 1) described herein.
  • a method of inhibiting coronavirus viral membrane fusion with a late endosomal membrane comprising administering a compound of Formula (I) or any of the embodiments thereof (e.g., compounds of Table 1) described herein.
  • a method of inhibiting coronavirus viral membrane fusion with an endo-lysosomal membrane comprising administering a compound of Formula (I) or any of the embodiments thereof (e.g., compounds of Table 1) described herein.
  • a method of inhibiting coronavirus viral membrane fusion with a lysosomal membrane is provided comprising administering a compound of Formula (I) or any of the embodiments thereof (e.g., compounds of Table 1) described herein.
  • a method of inhibiting coronavirus viral membrane fusion with an early macropinosomal membrane comprising administering a compound of Formula (I) or any of the embodiments thereof (e.g., compounds of Table 1) described herein.
  • a method of inhibiting coronavirus viral membrane fusion with a macropinosomal membrane comprising administering a compound of Formula (I) or any of the embodiments thereof (e.g., compounds of Table 1) described herein.
  • a method of inhibiting coronavirus viral membrane fusion with a late macropinosomal membrane comprising administering a compound of Formula (I) or any of the embodiments thereof (e.g., compounds of Table 1) described herein.
  • a method of inhibiting coronavirus viral membrane fusion with the endoplasmic reticulum (ER) comprising administering a compound of Formula (I) or any of the embodiments thereof (e.g., compounds of Table 1) described herein.
  • a method of inhibiting coronavirus viral membrane fusing with the plasma membrane directly comprising administering a compound of Formula (I) or any of the embodiments thereof (e.g., compounds of Table 1) described herein.
  • the coronavirus may be an alpha-coronavirus, a beta-coronavirus lineage B, or a beta-coronavirus lineage A.
  • the compounds of this disclosure will be administered in a therapeutically effective amount by any of the accepted modes of administration for agents that serve similar utilities.
  • compounds of this disclosure will be administered as pharmaceutical compositions by any one of the following routes: oral, systemic (e.g., transdermal, intranasal or by suppository), or parenteral (e.g., intramuscular, intravenous, or subcutaneous) administration.
  • the manner of administration is nasal using a convenient daily dosage regimen, which can be adjusted according to the degree of affliction.
  • compositions can take the form of tablets, pills, capsules, semisolids, powders, sustained release formulations, solutions, suspensions, elixirs, aerosols, or any other appropriate compositions.
  • Pharmaceutical compositions can be formulated using one or more pharmaceutically acceptable carriers comprising excipients and auxiliaries. The formulation can be modified depending upon the route of administration chosen.
  • the pharmaceutical compositions can also include the compounds described herein in a free base form or a pharmaceutically acceptable salt form.
  • Methods for formulation of the pharmaceutical compositions can include formulating any of the compounds described herein with one or more inert, pharmaceutically acceptable excipients or carriers to form a solid, semi-solid, or liquid composition.
  • Solid compositions can include, for example, powders, tablets, dispersible granules and capsules, and in some aspects, the solid compositions further contain nontoxic, auxiliary substances, for example wetting or emulsifying agents, pH buffering agents, and other pharmaceutically acceptable additives.
  • the compositions described herein can be lyophilized or in powder form for re- constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • the active ingredients can be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization (e.g., hydroxymethylcellulose or gelatin microcapsules and poly-(methylmethacylate) microcapsules, respectively), in colloidal drug- delivery systems (e.g., liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
  • the pharmaceutical compositions and formulations can be sterilized. Sterilization can be accomplished by filtration through sterile filtration.
  • the pharmaceutical compositions described herein can be formulated for administration as an injection.
  • Non-limiting examples of formulations for injection can include a sterile suspension, solution, or emulsion in oily or aqueous vehicles.
  • Suitable oily vehicles can include, but are not limited to, lipophilic solvents or vehicles such as fatty oils, synthetic fatty acid esters, or liposomes.
  • Aqueous injection suspensions can contain substances which increase the viscosity of the suspension.
  • the suspension can also contain suitable stabilizers.
  • Injections can be formulated for bolus injection or continuous infusion.
  • the compounds can be formulated in a unit dosage injectable form (e.g., solution, suspension, emulsion) in association with a pharmaceutically acceptable parenteral vehicle.
  • Such vehicles can be inherently nontoxic, and non-therapeutic.
  • a vehicle can be water, saline, Ringer’s solution, dextrose solution, and 5% human serum albumin.
  • Nonaqueous vehicles such as fixed oils and ethyl oleate can also be used.
  • Liposomes can be used as carriers.
  • the vehicle can contain minor amounts of additives such as substances that enhance isotonicity and chemical stability (e.g., buffers and preservatives).
  • Sustained-release preparations can also be prepared.
  • sustained-release matrices can include polyesters, hydrogels (e.g., poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides, copolymers of L-glutamic acid and ⁇ ethyl-L-glutamate, non- degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPO TM (i.e., injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-(–)-3-hydroxybutyric acid.
  • polyesters e.g., poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)
  • polylactides e.g., poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)
  • polylactides e.g., poly(2-
  • compositions described herein can be prepared for storage by mixing a compound with a pharmaceutically acceptable carrier, excipient, and/or a stabilizer.
  • This formulation can be a lyophilized formulation or an aqueous solution.
  • Acceptable carriers, excipients, and/or stabilizers can be nontoxic to recipients at the dosages and concentrations used.
  • Acceptable carriers, excipients, and/or stabilizers can include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives, polypeptides; proteins, such as serum albumin or gelatin; hydrophilic polymers; amino acids; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes; and/or non- ionic surfactants or polyethylene glycol.
  • buffers such as phosphate, citrate, and other organic acids
  • antioxidants including ascorbic acid and methionine
  • preservatives polypeptides
  • proteins such as serum albumin or gelatin
  • hydrophilic polymers amino acids
  • Compounds of the present disclosure may be used in methods of treating in combination with one or more other combination agents (e.g., one, two, or three other drugs) that are used in the prevention, treatment, control, amelioration, or reduction of risk of the diseases or conditions for which compounds of the present disclosure are useful.
  • the combination of the drugs together is safer or more effective than either drug alone.
  • the compounds disclosed herein and the one or more combination agents have complementary activities that do not adversely affect each other. Such molecules can be present in combination in amounts that are effective for the purpose intended.
  • Such other drug(s) may be administered, by a route and in an amount commonly used therefore, contemporaneously or sequentially with a compound of the present disclosure.
  • the agents are administered together in a single pharmaceutical composition in unit dosage form.
  • the pharmaceutical compositions of the present disclosure also include those that contain one or more other active ingredients, in addition to a compound of the present disclosure.
  • the weight ratio of the compound of the present disclosure to the second active agent may be varied and will depend upon the effective dose of each ingredient. Generally, an effective dose of each will be used.
  • combination therapy includes therapies in which the compound of the present disclosure and one or more other drugs are administered separately, and in some cases, the two or more agents are administered on different, overlapping schedules.
  • the compounds, pharmaceutical compositions, and methods of the present disclosure can be useful for treating and preventing infection in a subject such as, but not limited to, a mammal, a human, a non-human mammal, a domesticated animal (e.g., laboratory animals, household pets, or livestock), a non-domesticated animal (e.g., wildlife), a dog, a cat, a rodent, a mouse, a hamster, a cow, a bird, a chicken, a fish, a pig, a horse, a goat, a sheep, or a rabbit.
  • a mammal a human
  • a non-human mammal a domesticated animal (e.g., laboratory animals, household pets, or livestock), a non-domesticated animal (e.g., wildlife), a dog, a cat, a rodent, a mouse, a hamster, a cow, a bird, a chicken, a fish, a pig, a
  • compounds, pharmaceutical compositions, and methods of the present disclosure are used for treating a human.
  • the compounds, pharmaceutical compositions, and methods described herein can be useful as a therapeutic or preventative, for example a treatment or preventative that can be administered to a subject in need thereof.
  • a therapeutic or preventative effect can be obtained in a subject by prevention (complete or partial), reduction, suppression, remission, or eradication of a disease state, including, but not limited to, a symptom thereof.
  • a therapeutic effect in a subject having a disease or condition, or pre-disposed to have or is beginning to have the disease or condition can be obtained by a reduction, a suppression, a prevention, a remission, or an eradication of the condition or disease, or pre-condition or pre-disease state.
  • therapeutically effective amounts of the compounds or pharmaceutical compositions described herein can be administered to a subject in need thereof, often for treating and/or preventing a condition or progression thereof.
  • a pharmaceutical composition can affect the physiology of the subject, such as the immune system, inflammatory response, or other physiologic affect.
  • Treat and/or treating can refer to any indicia of success in the treatment or amelioration of the disease or condition. Treating can include, for example, reducing, delaying or alleviating the severity of one or more symptoms of the disease or condition, or it can include reducing the frequency with which symptoms of a disease, defect, disorder, or adverse condition, and the like, are experienced by a patient. Treat can be used herein to refer to a method that results in some level of treatment or amelioration of the disease or condition and can contemplate a range of results directed to that end, including but not restricted to prevention of the condition entirely.
  • Prevent, preventing, and the like can refer to the prevention of the disease or condition in the patient. For example, if an individual at risk of contracting a disease is treated with the methods of the present disclosure and does not later contract the disease, then the disease has been prevented, at least over a period of time, in that individual.
  • the PIKfyve inhibitors described herein can prevent coronavirus infection.
  • the PIKfyve inhibitors described herein can treat a coronavirus infection.
  • a therapeutically effective amount can be the amount of a compound or pharmaceutical composition or an active component thereof sufficient to provide a beneficial effect or to otherwise reduce a detrimental non-beneficial event to the individual to whom the composition is administered.
  • a therapeutically effective dose can be a dose that produces one or more desired or desirable (e.g., beneficial) effects for which it is administered, such administration occurring one or more times over a given period of time.
  • An exact dose can depend on the purpose of the treatment and can be ascertainable by one skilled in the art using known techniques.
  • the compounds or pharmaceutical compositions described herein that can be used in the methods and uses described herein can be formulated and dosages established in a fashion consistent with good medical practice taking into account the disorder to be treated, the condition of the individual patient, the site of delivery of the compound or pharmaceutical composition, the method of administration and other factors known to practitioners.
  • the compounds or pharmaceutical compositions can be prepared according to the description of preparation described herein.
  • administration of the compounds or pharmaceutical compositions can include routes of administration, non-limiting examples of administration routes include intravenous, intraarterial, subcutaneous, subdural, intramuscular, intracranial, intrasternal, intratumoral, or intraperitoneally.
  • a pharmaceutical composition or compound can be administered to a subject by additional routes of administration, for example, by inhalation, oral, dermal, intranasal, or intrathecal administration.
  • Pharmaceutical compositions or compounds of the present disclosure can be administered to a subject in need thereof in a first administration, and in one or more additional administrations. The one or more additional administrations can be administered to the subject in need thereof minutes, hours, days, weeks, or months following the first administration.
  • any one of the additional administrations can be administered to the subject in need thereof less than 21 days, or less than 14 days, less than 10 days, less than 7 days, less than 4 days or less than 1 day after the first administration.
  • the one or more administrations can occur more than once per day, more than once per week, or more than once per month.
  • the compounds or pharmaceutical compositions can be administered to the subject in need thereof in cycles of 21 days, 14 days, 10 days, 7 days, 4 days, or daily over a period of one to seven days.
  • the compounds, pharmaceutical compositions, and methods provided herein can be useful for the treatment of a plurality of diseases or conditions or preventing a disease or a condition in a subject, or other therapeutic applications for subjects in need thereof. III.
  • the PIKfyve inhibitors described herein can be administered to treat and/or prevent certain coronavirus infection.
  • the coronaviruses belong to the order Nidovirales, family Coronaviridae, and the subfamily Coronavirinae. They are genetically categorized into four genera: the Alphacoronavirus, Betacoronavirus, Gammacoronavirus, and Deltacoronavirus. Mahendra et al., Cureus, 12(3):e7423 (2020).
  • Alphacoronaviruses and the Betacoronaviruses typically infect mammals, whereas the Gammacoronaviruses and the Deltacoronaviruses predominantly infect birds.
  • the coronavirus is an Alphacoronavirus, Betacoronavirus, Gammacoronavirus, or Deltacoronavirus.
  • Alphacoronavirus strains that are associated with human disease include HCoV-229E and HCoV-NL63.
  • alphacoronavirus strains include feline infectious peritonitis virus (FIPV), canine coronavirus (CCoV), porcine respiratory coronavirus (PRCV), porcine epidemic diarrhea virus (PEDV), transmissible gastroenteritis virus (TGEV), Rhinolophus bat coronavirus (Rh-BatCoV) HKU2, Miniopterus bat coronavirus (Mi-BatCoV) HKU8, Mi-BatCoV 1A, Mi-BatCoV 1B, Rousettus bat coronavirus (Ro-BatCoV) HKU10183A, Hipposideros bat coronavirus (Hi-BatCoV) HKU10 LSH5A, and Scotiphilus bat coronavirus (Sc-BatCoV) 512.
  • FIPV feline infectious peritonitis virus
  • CoV canine coronavirus
  • PRCV porcine respiratory coronavirus
  • PEDV porcine epidemic diarrhea virus
  • TGEV transmissible gastroenteriti
  • the coronavirus is HCoV-229E or HCoV-NL63.
  • the coronavirus is FIPV, CCoV, PRCV, PEDV, TGEV, Rh-BatCoV HKU2, Mi-BatCoV HKU8, Mi-BatCoV 1A, Mi-BatCoV 1B, Ro-BatCoV HKU10183A, Hi- BatCoV HKU10 LSH5A, and Sc-BatCoV 512.
  • Betacoronaviruses are divided into four subgroups: Embecovirus (lineage A), Sarbecovirus (lineage B), Merbecovirus (lineage C), and Nobecovirus (lineage D).
  • Embecovirus (lineage A) include bovine coronavirus (BCoV), human coronavirus OC43 (HCoV-OC43), HCoV-HKU1, porcine hemagglutinating encephalomyelitis virus (PHEV), giraffe coronavirus (GiCoV), and murine hepatitis virus (MHV).
  • Sarbecovirus examples include SARS-CoV-1 and SARSr-Rh-BatCoV HKU3.
  • Examples of Merbecovirus (lineage C) include MERS-CoV, Tylonycteris bat coronavirus (Ty-BatCoV) HKU4 and Pipistrellus bat coronavirus (Pi-BatCoV) HKU5.
  • Examples of Nobecovirus (lineage D) include Ro-BatCoV HKU9.
  • Betacoronavirus strains that are associated with human disease include SARS-CoV-1, HCoV- OC43, HCoV-HKU1, and MERS-CoV.
  • the coronavirus causes upper respiratory tract disease, lower respiratory tract disease, fever, sore throat, swollen adenoids, colds with minor or major symptoms, malaise, muscle and joint pains, nausea, vomiting, loss of appetite, pneumonia, secondary bacterial infection, bronchitis, dyspnea, diarrhea, shortness of breath, acute respiratory distress syndrome, cytokine storm, multi-organ failure, septic shock, blood clots, loss of smell, or loss of taste.
  • the coronavirus causes no symptoms at all.
  • the disclosed compounds are administered to block an alpha- coronavirus entry into a host cell.
  • the disclosed compounds are administered to block a beta-coronavirus lineage B entry into a host cell. In some embodiments, the disclosed compounds are administered to block a beta-coronavirus lineage A entry into a host cell. In some embodiments, the disclosed compounds are administered to prevent an alpha- coronavirus infection in a subject. In some embodiments, the disclosed compounds are administered to prevent a beta-coronavirus lineage B infection in a subject. In some embodiments, the disclosed compounds are administered to prevent a beta-coronavirus lineage A infection in a subject. In some embodiments, the disclosed compounds are administered to block an alpha-coronavirus entry into a host cell and prevent an alpha-coronavirus infection in a subject.
  • the disclosed compounds are administered to block a beta- coronavirus lineage B entry into a host cell prevent a beta-coronavirus lineage B infection in a subject. In some embodiments, the disclosed compounds are administered to block a beta- coronavirus lineage A entry into a host cell prevent a beta-coronavirus lineage A infection in a subject.
  • the alpha-coronavirus is HCoV 229E.
  • the beta-coronavirus lineage B is SARS-CoV2.
  • the beta-coronavirus lineage A is HCoV OC43.
  • the infection is caused by SARS-CoV-2, and the infection is COVID-19. IV.
  • PIKfyve inhibitor compounds [0222]
  • the compounds described herein may in some cases exist as diastereomers, enantiomers, or other stereoisomeric forms. All chiral, diastereomeric, racemic forms, as individual forms and mixtures thereof, are within the scope of this disclosure, unless the specific stereochemistry or isomeric form is specifically indicated.
  • Compounds of the present disclosure containing an asymmetrically substituted atom may be isolated in optically active, optically enriched, optically pure, or racemic forms. It is well known in the art how to prepare optically active forms, such as by resolution of materials.
  • Stereoisomers may be performed by chromatography or by forming diastereomers and separating by recrystallization, or chromatography, or any combination thereof. (Jean Jacques, Andre Collet, Samuel H. Wilen, “Enantiomers, Racemates and Resolutions”, John Wiley and Sons, Inc., 1981, herein incorporated by reference for this disclosure). Stereoisomers may also be obtained by stereoselective synthesis. [0223] Certain compounds of Formula (I) (or any of the embodiments thereof described herein) and/or a pharmaceutically acceptable salt or prodrug thereof can exist as tautomers and/or geometric isomers.
  • the compounds described herein include hydrates and solvates of the compounds or pharmaceutically acceptable salts thereof.
  • the present disclosure also includes the prodrugs of compounds of Formula (I) (or any of the embodiments thereof described herein) and/or a pharmaceutically acceptable salt or prodrug thereof.
  • the term prodrug is intended to represent covalently bonded carriers, which are capable of releasing the active ingredient of Formula (I) (or any of the embodiments thereof described herein) when the prodrug is administered to a mammalian subject. Release of the active ingredient occurs in vivo.
  • Prodrugs can be prepared by techniques known to one skilled in the art. These techniques generally modify appropriate functional groups in a given compound. These modified functional groups however regenerate original functional groups in vivo or by routine manipulation.
  • Prodrugs of compounds of Formula (I) include compounds wherein a hydroxy, amino, carboxylic, or a similar group is modified.
  • esters e.g., acetate, formate, and benzoate derivatives
  • carbamates e.g., N,N-dimethylaminocarbonyl
  • amides e.g., trifluoroacet
  • Prodrugs of compounds of Formula (I) (or any of the embodiments thereof described herein) and/or a pharmaceutically acceptable salt or prodrug thereof are also within the scope of this disclosure.
  • the present disclosure also includes polymorphic forms (amorphous as well as crystalline) and deuterated forms of compounds of Formula (I) (or any of the embodiments thereof described herein) and/or a pharmaceutically acceptable salt or prodrug thereof.
  • the compounds disclosed herein, in some embodiments, are used in different enriched isotopic forms, e.g., enriched in the content of 2 H, 3 H, 11 C, 13 C and/or 14 C. In one particular embodiment, the compound is deuterated in at least one position.
  • deuterated forms can be made by the procedure described in U.S. Patent Nos.5,846,514 and 6,334,997. As described in U.S. Patent Nos.5,846,514 and 6,334,997, deuteration can improve the metabolic stability and or efficacy, thus increasing the duration of action of drugs.
  • structures depicted herein are intended to include compounds which differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structures except for the replacement of a hydrogen by a deuterium or tritium, or the replacement of a carbon by 13 C- or 14 C-enriched carbon are within the scope of the present disclosure.
  • the compounds of the present disclosure optionally contain unnatural proportions of atomic isotopes at one or more atoms that constitute such compounds.
  • the compounds may be labeled with isotopes, such as for example, deuterium ( 2 H), tritium ( 3 H), iodine-125 ( 125 I) or carbon-14 ( 14 C).
  • isotopes such as for example, deuterium ( 2 H), tritium ( 3 H), iodine-125 ( 125 I) or carbon-14 ( 14 C).
  • Isotopic substitution with 2 H, 11 C, 13 C, 14 C, 15 C, 12 N, 13 N, 15 N, 16 N, 16 O, 17 O, 14 F, 15 F, 16 F, 17 F, 18 F, 33 S, 34 S, 35 S, 36 S, 35 Cl, 37 Cl, 79 Br, 81 Br, and 125 I are all contemplated.
  • the compounds disclosed herein have some or all of the 1 H atoms replaced with 2 H atoms.
  • the methods of synthesis for deuterium-containing compounds are known in the art and include, by way of non-limiting example only, the following synthetic methods.
  • Deuterium substituted compounds are synthesized using various methods such as described in: Dean, Dennis C.; Editor. Recent Advances in the Synthesis and Applications of Radiolabeled Compounds for Drug Discovery and Development. [In: Curr., Pharm. Des., 2000; 6(10)] 2000, 110 pp; George W.; Varma, Rajender S.
  • R 1a and R 1b taken together with the nitrogen to which they are attached form: wherein X and Y are independently N or CR a ; wherein R a is H or C 1-4 alkyl; and R b is phenyl, monocyclic cycloalkyl, monocyclic heterocyclyl, monocyclic heterocycloalkyl, or monocyclic heteroaryl, each optionally substituted with one, two, or three R d substituents; or R 1a is H or C1-4alkyl; and R 1b is a heteroaryl optionally substituted with R c ; wherein R c is C1-4alkyl, phenyl, -C1-4alkyl-phenyl, monocyclic cycloalkyl, -C 1-4 alkyl-(monocyclic cycloalkyl), monocyclic heterocyclyl, monocyclic heterocycloalkyl, monocyclic heteroaryl, or
  • R 1a and R 1b taken together with the nitrogen to which they are attached form: wherein X and Y are independently N or CR a ; wherein R a is H or C 1-4 alkyl; and R b is phenyl, monocyclic cycloalkyl, monocyclic heterocyclyl, monocyclic heterocycloalkyl, or monocyclic heteroaryl, each optionally substituted with one, two, or three R d substituents; or R 1a is H or C1-4alkyl; and R 1b is a moncyclic heteroaryl optionally substituted with R c ; wherein R c is C1-4alkyl, phenyl, -C1-4alkyl-phenyl, monocyclic cycloalkyl, -C 1-4 alkyl-(monocyclic cycloalkyl), monocyclic heterocyclyl, monocyclic heterocycloalkyl, monocyclic heteroaryl, or -C1-4alkyl-
  • R 1a and R 1b are taken together with the nitrogen to which they are attached to form . In some embodiments, R 1a and R 1b are taken together with the nitrogen to which they are attached to form. In some embodiments, X is N and Y is CR a . In some embodiments, X is CR a and Y is N. In some embodiments, X is N and Y is N. In some embodiments, R a is H, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, or tert-butyl. In some embodiments, R a is H or methyl. In some embodiments, R a is H.
  • R b is optionally substituted phenyl. In some embodiments, R b is optionally substituted monocyclic heteroaryl. In some embodiments, R b is optionally substituted pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, furanyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, or pyridazinyl. In some embodiments, R b is optionally substituted pyridinyl or pyrimidinyl.
  • R b is optionally substituted pyridinyl. In some embodiments, R b is phenyl. In some embodiments, R b is o-, m-, or p-tolyl. In some embodiments, R b is optionally substituted with one or two R d substituents. In some embodiments, R b is optionally substituted with one R d substituent. In some embodiments, R b is methylpryridinyl, phenyl, tolyl, chlorophenyl, bromophenyl, or methoxyphenyl.
  • R 1a is H or C 1-4 alkyl; and R 1b is a 5-membered N-containing heteroaryl optionally substituted with R c .
  • R 1a is H.
  • R 1a is C 1-4 alkyl.
  • R 1a is methyl.
  • R 1b is pyrrolyl, pyrazolyl, imidazolyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, oxadiazolyl, thiadiazolyl, pyrazolopyridinyl, or indazolyl, each optionally substituted with R c .
  • R 1b is a monocyclic heteroaryl.
  • R 1b is pyrrolyl, pyrazolyl, imidazolyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, oxadiazolyl, or thiadiazolyl, each optionally substituted with R c .
  • R 1b is pyrazolyl, imidazolyl, oxazolyl, oxadiazolyl or isoxazolyl, each optionally substituted with R c .
  • R 1b is pyrazolyl, optionally substituted with R c .
  • R 1b is [ 0240] In some embodiments, R 1b is [0241] In some embodiments, R c is optionally substituted C 1-4 alkyl. In some embodiments, R c is methyl, ethyl, isopropyl, or trifluoromethyl. In some embodiments, R c is optionally substituted phenyl. In some embodiments, R c is phenyl or o-, m-, p-tolyl, fluorophenyl, methoxyphenyl, or trifluoromethoxyphenyl. In some embodiments, R c is phenyl. In some embodiments, R c is optionally substituted monocyclic cycloalkyl.
  • R c is optionally substituted cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl. In some embodiments, R c is optionally substituted cyclopropyl. In some embodiments, R c is optionally substituted monocyclic heterocycloalkyl. In some embodiments, R c is optionally substituted cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, or cyclohexylmethyl. In some embodiments, R c is optionally substituted monocyclic heterocyclyl.
  • R c is optionally substituted pyrrolidinyl, tetrahydrofuranyl, piperidinyl, morpholinyl, or piperazinyl. In some embodiments, R c is optionally substituted monocyclic heteroaryl. In some embodiments, R c is optionally substituted pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, furanyl, thiophenyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, or pyridazinyl.
  • R c is optionally substituted pyrazole, thiophenyl, imidazolyl, pyridinyl, or pyrimidinyl. In some embodiments, R c is optionally substituted pyrazolyl. In some embodiments, R c is optionally substituted pyridinyl. In some embodiments, R c is methylpyridinyl. In some embodiments, R c is optionally substituted -C 1-4 alkyl-phenyl, -C 1- 4alkyl-(monocyclic cycloalkyl), monocyclic heterocycloalkyl, or -C1-4alkyl-(monocyclic heteroaryl).
  • R c is optionally substituted benzyl, -CH 2 -(monocyclic cycloalkyl), -CH 2 -(monocyclic heterocycloalkyl), or -CH 2 -(monocyclic heteroaryl). In some embodiments, R c is optionally substituted benzyl or -CH 2 -(monocyclic cycloalkyl), such as -CH 2 -cyclopropyl. In some embodiments, each R c is optionally substituted with one or two R d substituents.
  • each R d substituent is independently C 1-4 alkyl, -O-C 1-4 alkyl, C 1-4 haloalkyl, or halo. In some embodiments, each R d substituent is independently methyl, ethyl, isopropyl, -CF3, -OCH3, -OCF3, or fluoro. [0243] In some embodiments, R g and R h are each independently H or methyl.
  • R 2 and R 3 taken together with the nitrogen to which they are attached form a heterocyclyl, optionally substituted with one, two, three, or four R j substituents, or further wherein any of the hydrogens bonded to carbon atoms are optionally replaced by deuterium.
  • each of R 2 and R 3 are independently selected from H, pyrrolidinyl, piperidinyl, and piperazinyl, wherein each pyrrolidinyl, piperidinyl, and piperazinyl is optionally substituted with one R j substituent.
  • R 2 and R 3 taken together with the nitrogen to which they are attached form pyrrolidinyl, piperidinyl, piperazinyl, morpholino, thiomorpholino, or thiomorpholino-1,1-dioxide, each optionally substituted with one, two, three, or four R j substituents.
  • R 2 and R 3 taken together with the nitrogen to which they are attached form morpholino or piperazinyl, optionally substituted with one, two, three, or four R j substituents.
  • R 2 and R 3 taken together with the nitrogen to which they are attached form 2,2,6,6-tetrafluoro-morpholino, morpholino-2-one, morpholino-3-one, piperazinyl-2-one, piperazinyl-3-one, thiomorpholino-1,1-dioxide.
  • each R j substituent is independently methyl, oxo, hydroxy, -OCH3, NH 2 , halo, -CF3, or -OCF3.
  • R 2 and R 3 taken together with the nitrogen to which they are attached form morpholino in which 1 to 8 hydrogens are replaced with deuterium.
  • R k and R l are each independently H or methyl.
  • R 4 is H.
  • R 4 is chloro.
  • R 4 is halo, -C(O)OH, C 1-4 alkylNR x R y , or -C(O)NR x R y , or is a cycloalkyl, heterocyclyl, heterocycloalkyl, phenyl or heteroaryl, wherein each cycloalkyl, heterocyclyl, heterocycloalkyl, phenyl or heteroaryl is optionally substituted with one, two, or three R z substituents.
  • R 4 is optionally substituted phenyl. In some embodiments, R 4 is optionally substituted heteroaryl. In some embodiments, R 4 is optionally substituted monocyclic heteroaryl. In some embodiments, R 4 is optionally substituted pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, furanyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, or pyridazinyl. In some embodiments, R 4 is optionally substituted pyridinyl or pyrimidinyl.
  • R 4 is each optionally substituted with 1 or 2 R z groups.
  • R 4 is optionally substituted pyridinyl.
  • R 4 is pyridinyl.
  • R 4 is 4-pyridyl, 3-pyridyl, or 2-pyridyl.
  • R 4 is 4-pyridyl.
  • R 4 is optionally substituted with one or two R z substituents.
  • R 4 is phenyl or pyridyl, each optionally substituted with one or two substituents selected from C1-4alkyl, -CF3, fluoro, chloro, -OCH3, and -OCF3.
  • R 4 is heterocyclyl, optionally substituted with one or two R z substituents.
  • R 4 is pyrrolidinyl, piperidinyl, piperazinyl, morpholino, or thiomorpholino, optionally substituted with one or two R z substituents.
  • R 4 is pyrrolidinyl, or piperazinyl, optionally substituted with one C1-4alkyl.
  • R 4 is optionally substituted pyrazolyl. In some embodiments, R 4 is optionally substituted with one or two R z substituents. In some embodiments, R 4 is optionally substituted with one R z substituent. In some embodiments, R 4 is 3-methyl-1H-pyrazol-5-yl, 3- methylisothiazol-5-yl, 2-methyl-1H-imidazol-5-yl, 1-methyl-pyrazol-4-yl, 1-methylpyrazol-3-yl, 1-((1-acetamido)-eth-2-yl)-5-methyl-pyrazol-3-yl, 1-((1-chloromethylamido)-eth-2-yl)-5- methyl-pyrazol-3-yl, 1-((1-acrylamido)-eth-2-yl)-5-methyl-pyrazol-3-yl, thiazol-2-yl, pyrazol-4- yl, pyrazol-1-yl, o
  • R 4 is heterocycloalkyl, optionally substituted with one or two R z substituents.
  • R 4 is pyrrolidinylmethyl, piperidinylmethyl, piperazinylmethyl, morpholinomethyl, or thiomorpholinomethyl, optionally substituted with one or two R z substituents.
  • R 4 is C1-4alkylNR x R y .
  • R 4 is CH 2 NR x R y .
  • R 4 is -C(O)NR x R y .
  • R x is H.
  • R x is methyl or ethyl, optionally substituted with one, two, or three R o substituents. In some embodiments, R x is methyl. [0259] In some embodiments, R y is H, methyl, ethyl, methyoxy, or methoxyethyl. In some embodiments, R y is H. In some embodiments, R y is C 1-4 alkyl, optionally substituted with one, two, or three R o substituents. In some embodiments, R y is methyl, ethyl, propyl, or isopropyl, each optionally substituted with one, two, or three R o substituents.
  • R y is methyl, ethyl, or methoxyethyl. In some embodiments, R y is methoxy. In some embodiments, R y is -SO2-R r or C1-4alkyl-SO2-R r . R y is -SO2-R r , C1-4alkyl-SO2-R r ; and R r is CH3 or NH 2 , NHCH3, or N(CH3)2. In some embodiments, R y is -SO2-methyl, C2-4alkyl-SO2-N(CH3)2. In some embodiments, R y is -SO 2 -methyl.
  • R y is monocyclic cycloalkyl or -C1-2alkyl(monocyclic cycloalkyl), each optionally substituted with one, two, or three R o substituents. In some embodiments, R y is monocyclic cycloalkyl, optionally substituted with one, two, or three R o substituents. In some embodiments, R y is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl, each optionally substituted with one, two, or three R o substituents. In some embodiments, R y is cyclopropyl.
  • R y is cyclopropyl, cyclobutyl, cyclopentyl, cyclopropylmethyl, 1-cyclopropylethyl, 2-cyclopropylethyl, cyclobutylmethyl, or cyclopentylmethyl.
  • R y is monocyclic heterocycloalkyl, optionally substituted with one, two, or three R o substituents.
  • R y is optionally substituted azetidinyl, oxetanyl, pyrrolidinyl, piperidinyl, morpholinyl, piperazinyl, azepanyl, azocanyl, tetrahydrofuranyl, or tetrahydropyranyl, optionally substituted with methyl.
  • R y is optionally substituted azetidinyl, pyrrolidinyl, piperidinyl, morpholinyl, or piperazinyl.
  • R y is monocyclic heterocycloalkyl, optionally substituted with one, two, or three R o substituents.
  • R y is optionally substituted azetidinylmethyl, oxetanylmethyl, pyrrolidinylmethyl, piperidinylmethyl, morpholinylmethyl, or piperazinylmethyl, optionally substituted with methyl.
  • R x and R y is H and the other is -CH3. In some embodiments, both of R x and R y is H. In some embodiments, both of R x and R y is -CH 3 .
  • R x and R y taken together with the nitrogen to which they are attached form a monocyclic heterocyclyl, optionally substituted with C1-4alkyl.
  • R x and R y are taken together with the nitrogen to which they are attached to form azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, or thiomorpholino, each optionally substituted with methyl.
  • each R z is independently C1-4alkyl, halo, -OH, or -OC1-4alkyl, wherein each alkyl is optionally substituted with -NR m R n .
  • each R z is independently -CH 3 , -OH, halo, or -OCH 3 .
  • R z is C 2-3 alkyl substituted with -NR m R n . In some embodiments, R z is C2-4alkyl substituted with -NR m R n or OCH3. In some embodiments, each R z substituent is independently -NR p R q , -C(O)NR p R q .
  • each R z substituent is methyl, ethyl, isopropyl, -CF 3 , fluoro, chloro, -OCH3, -OCF3, methylamino, ethylamino, propylamino, butylamino, aminomethyl, aminoethyl, aminopropyl, aminobutyl, dimethylamino, dimethylaminomethyl, dimethylaminoethyl, dimethylaminopropyl, dimethylaminobutyl, -C(O)methylamino, -C(O)ethylamino, -C(O)propylamino, -C(O)butylamino, -C(O)dimethylamino, -C(O)dimethylaminomethyl, -C(O)dimethylaminoethyl, -C(O)dimethylaminopropyl, or -C(O)
  • R m and R n are each independently H, C1-4alkyl, C(O)CH3, C(O)CH 2 Cl, or C(O)CH 2 CH 2 . In some embodiments, R m and R n are each H. In some embodiments, R m and R n are each methyl. In some embodiments, R m and R n taken together with the nitrogen to which they are attached form a monocyclic heterocyclyl, optionally substituted with one or two R o substituents.
  • R m and R n taken together with the nitrogen to which they are attached form pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholino, or thiomorpholino-1,1-dioxide, each optionally substituted with one or two R o substituents.
  • R m and R n taken together with the nitrogen to which they are attached form pyrrolidinyl, piperidinyl, piperazinyl, or morpholino, each optionally substituted with one or two R o substituents.
  • R m and R n taken together with the nitrogen to which they are attached form pyrrolidinyl, piperidinyl, piperazinyl, or morpholinyl, each optionally substituted with methyl.
  • each R o substituent is C1-4alkyl.
  • each R o substituent is -NR p R q .
  • R p and R q are each independently H or methyl.
  • R p and R q are each independently H, methyl, C 1-4 alkylNH 2 , C1-4alkylNHCH3, or C1-4alkylN(CH3)2.
  • R 5 is H, methyl, ethyl, chloro, bromo, fluoro, -OH, or -OCH 3 . In some embodiments, R 5 is H.
  • the compound of Formula (I) or the pharmaceutically acceptable salt thereof is a compound of Formula (II): wherein R c1 is phenyl or pyridyl, each optionally substituted with one or two substituents selected from C1-4alkyl, -CF3, fluoro, chloro, -OCH3, and -OCF3; and R 4a is C1-4alkylNR x R y or C(O)NR x R y wherein R x and R y are as defined herein; or phenyl, pyrazolyl, or pyridyl, each optionally substituted with one or two R z groups; or a pharmaceutically acceptable salt or prodrug thereof.
  • the compound of Formula (I) or the pharmaceutically acceptable salt thereof is a compound of Formula (III): wherein R c1 is phenyl or pyridyl, each optionally substituted with one or two substituents selected from C1-4alkyl, -CF3, fluoro, chloro, -OCH3, and -OCF3; and R 4a is C1-4alkylNR x R y or -C(O)NR x R y wherein R x and R y are as defined herein; or phenyl, pyrazolyl, or pyridyl, each optionally substituted with one or two R z groups; or a pharmaceutically acceptable salt or prodrug thereof.
  • R c1 is phenyl or pyridyl, each optionally substituted with one or two substituents selected from C1-4alkyl, -CF3, fluoro, chloro, -OCH3, and -OCF3
  • R 4a is C1-4alkylNR x R
  • the compound of Formula (I) or the pharmaceutically acceptable salt thereof is a compound of Formula (IV): wherein R c1 is phenyl or pyridyl, each optionally substituted with one or two substituents selected from C1-4alkyl, -CF3, fluoro, chloro, -OCH3, and -OCF3; and R 4a is C1-4alkylNR x R y or -C(O)NR x R y wherein R x and R y are as defined herein; or phenyl, pyrazolyl, or pyridyl, each optionally substituted with one or two R z groups; or a pharmaceutically acceptable salt or prodrug thereof.
  • R c1 is phenyl or pyridyl, each optionally substituted with one or two substituents selected from C1-4alkyl, -CF3, fluoro, chloro, -OCH3, and -OCF3
  • R 4a is C1-4alkylNR x R
  • R c1 is phenyl or pyridyl, each optionally substituted with methyl, -CF 3 , Cl, Br, or OCH 3. In some embodiments, R c1 is phenyl or m-tolyl. In some embodiments, R c1 is pyridyl. In some embodiments, R c1 is 4-pyridyl. [0272] In some embodiments, R 4a is phenyl or pyridyl, each optionally substituted with methyl or -CF3. In some embodiments, R 4a is phenyl. In some embodiments, R 4a is tolyl. In some embodiments, R 4a is m-tolyl.
  • R 4a is pyridyl. In some embodiments, R 4a is 4-pyridyl. [0273] In some embodiments, R 4a is pyrazolyl optionally substituted with one or two R z groups. [0274] In some embodiments, each R z is independently methyl, ethyl, isopropyl, -CF 3 , fluoro, chloro, -OCH3, -OCF3, methylamino, ethylamino, propylamino, butylamino, aminomethyl, aminoethyl, aminopropyl, aminobutyl, dimethylamino, dimethylaminomethyl, dimethylaminoethyl, dimethylaminopropyl, dimethylaminobutyl, -C(O)methylamino, -C(O)ethylamino, -C(O)propylamino, -C(O)butylamino, -C(O)
  • R 4a is 3-methyl-1H-pyrazol-5-yl, 3-methylisothiazol-5-yl, 2- methyl-1H-imidazol-5-yl, 1-methyl-pyrazol-4-yl, 1-methylpyrazol-3-yl, 1-((1-acetamido)-eth-2- yl)-5-methyl-pyrazol-3-yl, 1-((1-chloromethylamido)-eth-2-yl)-5-methyl-pyrazol-3-yl, 1-((1- acrylamido)-eth-2-yl)-5-methyl-pyrazol-3-yl, thiazol-2-yl, pyrazol-4-yl, pyrazol-1-yl, oxazol-2- yl, or 3-(1-N,N-dimethyl-eth-2-yl)-4-methyl-pyrazol-1-yl.
  • R 4a is -C(O)NR x R y wherein R x is H or C1-4alkyl and R y is H, C1- 4alkyl, -O-C1-4alkyl, -SO2-R r , C1-4alkyl-SO2-R r monocyclic cycloalkyl, -C1-4alkyl(monocyclic cycloalkyl), monocyclic heterocyclyl, or monocyclic heterocycloalkyl, each optionally substituted with one, two, or three R o substituents; and R r and R o are as defined herein.
  • R 4a is -C(O)NR x R y wherein R x is H or methyl; and R y is H, methyl, ethyl, butyl, isopropyl, methoxy, -SO 2 -methyl, C 2-4 alkyl-SO 2 -methyl, C 2-4 alkyl-SO 2 -N(CH 3 ) 2 , cyclopropyl, cyclobutyl, cyclopentyl, cyclopropylmethyl, 1-cyclopropylethyl, 2-cyclopropylethyl, cyclobutylmethyl, cyclopentylmethyl, azetidinyl, oxetanyl, pyrrolidinyl, piperidinyl, morpholinyl, piperazinyl, azepanyl, azocanyl, tetrahydrofuranyl, tetrahydropyranyl, substituted azetidinylmethyl
  • the compound of Formula (I) or the pharmaceutically acceptable salt thereof is a compound of Formula (I): Formula (II): Formula (III): Formula (IV): as defined herein, wherein one or more hydrogen atoms attached to carbon atoms of the compound are replaced by deuterium atoms.
  • one or more hydrogen atoms attached to carbon atoms of R 1 , R 2 , R 3 , R 4 , R 5 , R 1a , R 1b , R 1c or R 4a are replaced by deuterium atoms.
  • one or more hydrogen atoms attached to carbon atoms of R a , R b , R c , R d , R g , R h , R j , R k , R l , R m , R n , R o , R p , R q , R r , R x , R y , or R z are replaced by deuterium atoms.
  • one or more R a , R b , R c , R d , R g , R h , R j , R k , R l , R m , R n , R o , R p , R q , R r , R x , R y , or R z group is a C1-4alkyl group wherein one or more hydrogen atoms attached to carbon atoms are replaced by deuterium atoms.
  • one or more R a , R b , R c , R d , R g , R h , R j , R k , R l , R m , R n , R o , R p , R q , R r , R x , R y , or R z group is a methyl group wherein one or more hydrogen atoms attached to the carbon atom are replaced by deuterium atoms.
  • one or more R a , R b , R c , R d , R g , R h , R j , R k , R l , R m , R n , R o , R p , R q , R r , R x , R y , or R z group is -CD 3 .
  • the compound of Formula (I) - (IV) comprises a -D in place of at least one -H, or a -CD3 substituent in place of at least one CH3.
  • the compound is a compound selected from those of Table 1: Table 1
  • kits for the treatment of a coronavirus infection are SARS-CoV-1, SARS-CoV-2, MERS-CoV, HCoV-OC43, HCoV-HKU1, HCoV-229E, or HCoV-NL63.
  • the kit may contain a compound of Formula I (or any of the embodiments thereof described herein), a pharmaceutically acceptable carrier, a physiologically acceptable carrier, instructions for use, a container, a vessel for administration, or any combination thereof.
  • the disclosure further provides any compounds disclosed herein for use in a method of treatment of the human or animal body by therapy. Therapy may be by any mechanism disclosed herein, such as inhibiting, reducing, or reducing progression of the diseases disclosed herein.
  • the disclosure further provides any compound disclosed herein for prevention or treatment of any condition disclosed herein.
  • the disclosure also provides any compound or pharmaceutical composition thereof disclosed herein for obtaining any clinical outcome disclosed herein for any condition disclosed herein.
  • the disclosure also provides use of any compound disclosed herein in the manufacture of a medicament for preventing or treating any disease or condition disclosed herein.
  • reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 2% MeOH/DCM to provide N,N-dimethyl-5-((4-morpholino-6-(pyridin-4-yl)furo[3,2-d]pyrimidin-2-yl)amino)-3-phenyl- 1H-pyrazole-1-sulfonamide (140 mg, 0.26 mmol) as a white solid.
  • EXAMPLE 2 Compound 10 using General Synthetic Route 2: 1.1) Synthesis of 5-amino-N,N-dimethyl-3-(pyridin-4-yl)-1H-pyrazole-1-sulfonamide [0306] To a solution of 3-(pyridin -4-yl)-1H-pyrazol-5-amine (500 mg, 3.12 mmol) in THF (5 mL) at 0°C was added NaH (374 mg, 9.36 mmol). After stirred at 0°C for 1 h, to the solution was added dimethylsulfamoyl chloride (536 mg, 3.75 mmol). The completion of the reaction was monitored by TLC.
  • reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide N,N-dimethyl-5-((4-morpholinofuro[3,2-d]pyrimidin-2-yl)amino)-3-(pyridin-4-yl)-1H-pyrazole- 1-sulfonamide (27 mg, 0.06 mmol) as a yellow solid.
  • EXAMPLE 3 Compound 11 using General Synthetic Route 3: 1.1) Synthesis of 2-bromo-6-chloro-4-morpholinofuro[3,2-d]pyrimidine [0312] To a solution of 2-bromo-4-morpholinofuro[3,2-d]pyrimidine (1.3 g, 4.59 mmol) in dry THF (4 mL) at -78 °C was added LDA (7.5 mL, 14.7 mmol) dropwise. After addition, the solution was stirred at that temperature for 1 h. Then to the solution was added NCS (733 mg, 5.5 mmol). The completion of the reaction was monitored by TLC. The reaction mixture was quenched with water (30 mL).
  • EXAMPLE 4 Compound 12 using General Synthetic Route 4: 1.1) Synthesis of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carbaldehyde
  • 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carbaldehyde To a solution of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine (50 mg, 0.21 mmol) in THF at -78 °C under N 2 was added n-BuLi ( 0.1 mL, 0.25 mmol). The mixture was stirred at that temperature for 15 min and then DMF (90 mg, 1.23 mmol) was added. The solution was allowed to warm to room temperature for 10 min. The completion of the reaction was monitored by TLC.
  • the mixture was stirred at room temperature for 3 h. The completion of the reaction was monitored by TLC.
  • the reaction mixture was quenched with a saturated NaHCO3 solution and the pH was adjusted to 8.
  • the aqueous solution was extracted with DCM/MeOH (15/1, 3 x 20 mL).
  • the combined organic phase was dried over anhydrous Na 2 SO 4 , filtrated and concentrated under reduced pressure.
  • the resulting residue was purified by silica gel column chromatography with a gradient elution of 30% EtOAc/PE to EtOAc to provide 2-chloro-4-morpholino-6- (morpholinomethyl)furo[3,2-d]pyrimidine (120 mg, 0.35 mmol).
  • reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide N,N-dimethyl-5-((4- morpholino-6-(morpholinomethyl)furo[3,2-d]pyrimidin-2-yl)amino)-3- phenyl-1H-pyrazole-1-sul-fonamide (23 mg, 0.04 mmol) as a yellow solid.
  • EXAMPLE 5 Compound 22 using General Synthetic Route 5: 1.1) Synthesis of 3-(2-methylpyridin-4-yl)-3-oxopropanenitrile [0322] To a solution of acetonitrile (1 .63 g, 39.7 mmol) in anhydrous THF (40 mL) at -70 °C under N2 was added n-BuLi (15.9 mL, 39.7 mmol) dropwise. After addition, a solution of methyl 2-methylisonicotinate (2.0 g, 13.2 mmol) in THF (10 mL) was added to the above solution over 10 min. The reaction mixture was stirred at that temperature for 2 h.
  • reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide impure N,N-dimethyl-3-(2-methylpyridin-4-yl)-5-((4-morpholino-6- (pyridin-4-yl)furo [3,2-d]pyrimidin-2-yl)amino)-1H-pyrazole-1-sulfonamide (23.2 mg, 0.04 mmol) as a yellow solid.
  • EXAMPLE 6 Compound 28 using General Synthetic Route 6: 1.1) Synthesis of 2-chloro-6-iodo-4-morpholinofuro[3,2-d]pyrimidine [0328] To a solution of 2-chloro -4-morpholinofuro[3,2-d]pyrimidine (2.0 g, 0.83 mmol) in THF (30 mL) at -78 °C under N2 was added LDA (1.33 mL, 2M, 2.66 mmol). After stirred at - 78 °C for 1 h, to the solution was added NIS (2.25 g, 1.0 mmol) in THF (10 mL). The completion of the reaction was monitored by TLC.
  • reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide N,N-dimethyl-5-((4-morpholino-6-(pyridin-2-yl)furo[3,2-d]pyrimidin-2-yl)amino)-3-(pyridin-4- yl)-1H-pyrazole-1-sulfonamide (35.6 mg, 0.065 mmol) as a yellow solid.
  • EXAMPLE 7 Compound 34 using General Synthetic Route 7: 1.1) Synthesis of 2-chloro-6-(1-methyl-1,2,3,6-tetrahydropyridin-4-yl)-4-morpholinofuro[3,2-d] pyrimidine [0334] A suspension of 2-chloro-6-iodo-4-morpholinofuro[3,2-d]pyrimidine (400 mg, 1.1 mmol), 1-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,2,3,6-tetrahydropyridine (244 mg, 1.1 mmol), K2CO3 (454 mg, 3.29 mmol) and Pd(PPh3)4 (127 mg, 0.011 mmol) in 1,4- dioxane/H 2 O (8/1, 40 mL) was heated to 50 °C for 2 h under N 2 .
  • reaction mixture was diluted with water and extracted with DCM/MeOH (15/1, 3 x 30mL).
  • the combined organic phase was dried over anhydrous Na 2 SO 4 , filtrated and concentrated under reduced pressure.
  • the residue was purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 10% MeOH/DCM to provide N,N-dimethyl-5-((6-(1-methyl-1,2,3,6-tetrahydropyridin-4-yl)-4-morpholinofuro[3,2- d]pyrimidin-2-yl)amino)-3-(pyridin-4-yl)-1H-pyrazole-1-sulfonamide (120 mg, 0.21 mmol) as a yellow solid.
  • EXAMPLE 8 Compound 35 using General Synthetic Route 8: 1.1) Synthesis of tert-butyl 3-(2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)-5,6- dihydropyridine-1(2H)-carboxylate [0339] A suspension of 2-chloro-6-iodo-4-morpholinofuro[3,2-d]pyrimidine (400 mg, 1.1 mmol), tert-butyl3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-5,6-dihydropyridine-1(2H)- carboxylate (339 mg, 1.1 mmol), K 2 CO 3 (454 mg, 3.29 mmol) and Pd(PPh 3 ) 4 (127 mg, 0.011 mmol) in 1,4-dioxane/H 2 O (8/1, 40 mL) was heated to 90 °C for 3 h under N 2
  • reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 2% MeOH/DCM to provide N,N-dimethyl-5-((6-(1-methyl-1,2,5,6-tetrahydropyridin-3-yl)-4-morpholinofuro[3,2- d]pyrimidin-2-yl)amino)-3-phenyl-1H-pyrazole-1-sulfonamide (50 mg, 0.089 mmol) as a brown solid.
  • EXAMPLE 9 Compound 39 using General Synthetic Route 9: 1.1) Synthesis of tert-butyl 3-(2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)-2,5-dihydro- 1H-pyrrole-1-carboxylate [0346] A suspension of 2-chloro-6-iodo-4-morpholinofuro[3,2-d]pyrimidine (800 mg, 2.2 mmol), tert-butyl 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2,5-dihydro-1H-pyrrole-1- carboxylate (649 mg, 2.2 mmol), K2CO3 (911 mg, 6.6 mmol) and Pd(PPh3)4 (254 mg, 0.022 mmol) in 1,4-dioxane/H 2 O (2/1, 60 mL) was heated to 90 °C for 3 h under N 2
  • reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide N,N-dimethyl-5-((6-(1-methylpyrrolidin-3-yl)-4-morpholinofuro[3,2- d]pyrimidin-2-yl)amino)-3-(m-tolyl)-1H-pyrazole-1-sulfonamide (60 mg, 0.11 mmol).
  • EXAMPLE 10 Compound 42 using General Synthetic Route 10: General procedure 10: 1.1) Synthesis of tert-butyl 4-(m-tolyl)-1H-pyrazole-1-carboxylate [0353] A solution of tert-butyl 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole - 1-carboxylate (1.5 g, 5.1 mmol) 1-bromo-3-methylbenzene (872 mg, 5.1 mmol), Cs2CO3 (91 mg, 0.28 mmol), PdCl 2 (PPh 3 ) 2 (590 mg, 0.051 mmol) and CsF (1.16 g, 7.65 mmol) in 1,4- dioxane/H 2 O (2/1, 30 mL) was heated to 80 °C overnight under N 2 .
  • EXAMPLE 11 Compound 43 using General Synthetic Route 11: 1.1) Synthesis of 5-phenylisoxazol-3-amine [0357] A solution of 3-oxo-3-phenylpropanenitrile (1.5 g, 10.3 mmol) in EtOH/H 2 O (1/1, 20 mL) was added hydroxylamine hydrochloride (785 mg, 11.3 mmol) and sodium hydroxide (450 mg, 11.3 mmol). The reaction mixture was heated to 80 °C overnight. To the above solution was added conc. HCl aq. (1.3 mL). The resulting mixture was stirred at 80 °C for 2 h. The completion of the reaction was monitored by TLC.
  • EXAMPLE 12 Compound 44 using General Synthetic Route 12: 1.1) Synthesis of Compound 44, 4-morpholino-N-(3-phenylisoxazol-5-yl)-6-(pyridin-4-yl) furo[3,2-d]pyrimidin-2-amine H [0360] A suspension of 2-bromo-4-morpholino-6-(pyridin-4-yl)furo[3,2-d]pyrimidine (76 mg, 0.21 mmol), 3-phenylisoxazol-5-amine (40 mg, 0.25 mmol), Cs2CO3 (158 mg, 0.48 mmol), Pd(OAc) 2 (5 mg, 0.021 mmol) and Xantphos (12 mg, 0.021 mmol) in DMF/1,4-dioxane (1/7, 8 mL) was heated to 80 °C for 25 min under microwave condition.
  • EXAMPLE 13 Compound 50 using General Synthetic Route 13: 1.1) Synthesis of 3-amino-N,N-dimethyl-1H-indazole-1-sulfonamide
  • 1H-indazol-3-amine (1 g, 7.5 mmol) in THF (30 mL) at 0 °C was added NaH (541 mg, 13.53 mmol). After stirred at 0 °C for 1 h, to the solution was added dimethylsulfamoyl chloride (1.61 g, 11.28 mmol). The completion of the reaction was monitored by TLC. The reaction mixture was quenched with a saturated NH4Cl solution.
  • reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide N,N-dimethyl-3-((4- morpholino-6-(pyridin-4-yl)furo[3,2-d]pyrimidin-2-yl)amino)-1H-indazole-1-sulfonamide (50 mg, 0.096 mmol) as a white solid.
  • EXAMPLE 14 Compound 52 using General Synthetic Route 14: 1.1) Synthesis of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid
  • 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid To a solution of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine (2.4 g, 10 mmol) in anhydrous THF (4 mL) at -78 °C under N 2 was added n-BuLi ( 5.2 mL, 2.5 M, 13 mmol) dropwise. The reaction mixture was stirred at that temperature for 1 h. To the solution was added dry ice (4.4 g, 100 mmol) in one portion. The resulting reaction mixture was stirred at that temperature for 3 h.
  • reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide N-(2,4-dimethoxybenzyl)-2-((1-(N,N-dimethylsulfamoyl)-3- phenyl-1H- pyrazol-5-yl)amino)-4-morpholinofuro[3,2-d]pyrimidine-6-carboxamide (43 mg, 0.065 mmol) as a yellow solid.
  • EXAMPLE 15 Compound 53 using General Synthetic Route 15: 1.1) Synthesis of azetidin-1-yl(2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)methanone [0372] A solution of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid (200 mg, 0.71 mmol) in DCM was added oxalyl dichloride (182 mg, 1.4 mmol) and one drop of DMF. The mixture was stirred at room temperature for 6 h. The solution was concentrated, and the resulting residue was dissolved in DCM (15 mL).
  • reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide 5-((6-(azetidine-1-carbonyl)-4-morpholinofuro[3,2-d]pyrimidin-2- yl)amino)-N,N-dimethyl-3-phenyl-1H-pyrazole-1-sulfonamide (143 mg, 0.26 mmol) as a yellow solid.
  • EXAMPLE 16 Compound 55 using General Synthetic Route 16: 1.1) Synthesis of (2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)(piperidin-1-yl)methanone [0376] A solution of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid (200 mg, 0.71 mmol), piperidine (61 mg, 0.71 mmol), HOBT (245 mg, 1.76 mmol), EDCl (340 mg, 1.76 mmol) in DMF (12 mL) was stirred at room temperature overnight. The completion of the reaction was monitored by TLC.
  • reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide N,N-dimethyl-5-((4-morpholino-6-(piperidine-1-carbonyl)furo[3,2- d]pyrimidin-2-yl) amino)-3-phenyl-1H-pyrazole-1-sulfonamide (53 mg, 0.09 mmol) as a colorless oil.
  • EXAMPLE 17 Compound 64 using General Synthetic Route 17: 1.1) Synthesis of methyl 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylate [0380] A solution of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid (1.0 g, 3.5 mmol) in DCM was added oxalyl dichloride (912.0 mg, 7.0 mmol) and one drop of DMF. The mixture was stirred at room temperature for 6 h. The solution was concentrated directly and the residue was dissolved in DCM (15 mL).
  • EXAMPLE 18 Compound 65 using General Synthetic Route 18: 1.1) Synthesis of 2-chloro-N-(methylsulfonyl)-4-morpholinofuro[3,2-d]pyrimidine-6- carboxamide [0386] A solution of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid (200 mg, 0.71 mmol), methanesulfonamide (134 mg, 1.42 mmol), 2-chloro-1-methylpyridin-1-ium iodide (216 mg, 0.85 mmol), Et 3 N (214 mg, 2.12 mmol) and DMAP (4.3 mg, 0.035 mmol) in DCM (25 mL) was stirred at room temperature overnight.
  • 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid 200 mg, 0.71 mmol
  • methanesulfonamide 134 mg, 1.42 mmol
  • the completion of the reaction was monitored by TLC.
  • the reaction was quenched with water (10 ml) and adjusted the pH to 4 using 1 N HCl aqueous solution.
  • the aqueous phase was extracted with DCM/MeOH (15/1, 3 x 20 mL).
  • the combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure.
  • the residue was purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide 2-chloro-N-(methylsulfonyl)-4- morpholinofuro [3,2-d]pyrimidine-6-carboxamide (244 mg, 0.68 mmol) as a yellow solid.
  • reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 2% MeOH/DCM to 10% MeOH/DCM to provide 2-((1-(N,N-dimethylsulfamoyl)-3-phenyl-1H-pyrazol-5-yl)amino)-N- (methylsulfonyl)-4-morpholinofuro[3,2-d]pyrimidine-6-carboxamide (30 mg, 0.051 mmol) as a white solid.
  • EXAMPLE 19 Compound 66 using General Synthetic Route 19: 1.1) Synthesis of 2-chloro-N-(cyclopropylmethyl)-N-methyl-4-morpholinofuro[3,2-d] pyrimidine-6-carboxamide [0390] A solution of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid (270 mg, 0.95 mmol), 1-cyclopropyl-N-methylmethanamine (80 mg, 0.95 mmol), DMAP (292 mg, 2.4 mmol) and EDCl (460 mg, 2.4 mmol) in DCM (20 mL) was stirred at room temperature overnight. The completion of the reaction was monitored by TLC.
  • reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 2% MeOH/DCM to provide 2-chloro-N-(cyclopropylmethyl)-N-methyl- 4-morpholinofuro[3,2-d] pyrimidine-6-carboxamide (272mg, 0.78 mmol) as a white solid.
  • EXAMPLE 20 Compound 69 using General Synthetic Route 20: 1.1) Synthesis of 4-cyclopropyl-3-oxobutanenitrile [0393] To a solution of acetonitrile (1.08 g, 26.3 mmol) and methyl 2-cyclopropylacetate (2.0 g, 17.5 mmol) in anhydrous THF (40 mL) at 0 °C under N 2 was added NaHDMS (13.2 mL, 26.3 mmol) dropwise. After addition, the solution was stirred at room temperature for 2 h. The completion of the reaction was monitored by TLC.
  • reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide 3-(cyclopropylmethyl)-N,N-dimethyl-5-((4-morpholino-6-(pyridin-4- yl)furo[3,2-d] pyrimidin-2-yl)amino)-1H-pyrazole-1-sulfonamide (47 mg, 0.09 mmol) as a white solid.
  • EXAMPLE 21 Compound 78 using General Synthetic Route 21: 1.1) Synthesis of 5-amino-3-cyclopropyl-N,N-dimethyl-1H-pyrazole-1-sulfonamide H N [0399] To a solution of 3-cyclopropyl-1H-pyrazol-5-amine (500 mg, 4.06 mmol) in THF (30 mL) at 0 °C was added NaH (243 mg, 6.1 mmol). After stirred at 0 °C for 1 h, to the solution was added dimethylsulfamoyl chloride (755 mg, 5.28 mmol). The completion of the reaction was monitored by TLC.
  • reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide 2-((3-cyclopropyl-1-(N,N-dimethylsulfamoyl)-1H-pyrazol-5-yl)amino)-N-methyl-4- morpholinofuro[3,2-d]pyrimidine-6-carboxamide (62 mg, 0.13 mmol) as a white solid.
  • EXAMPLE 22 Compound 89 using General Synthetic Route 22: 1.1) Synthesis of (Z)-methyl 2-((2-cyano-1-(pyridin-4-yl)vinyl)oxy)acetate
  • DEAD Diethyl azodicarboxylate
  • 3-oxo-3-(pyridin-4- yl)propanenitrile 1.5 g, 10.3 mmol
  • methyl 2-hydroxyacetate 1.2 g, 13.4 mmol
  • reaction mixture was heated to 120 °C overnight. Upon the completion of the reaction as monitored by TLC, the reaction was cooled to room temperature, a saturated NaHCO3 solution was added to adjust the pH to 8. The aqueous solution was extracted with EtOAc (3 x 50 mL). The combined organic phase was dried over anhydrous Na 2 SO 4 , filtrated and concentrated. The resulting residue was purified by silica gel column chromatography with a gradient elution of 50% EtOAc/PE to 75% EtOAc/PE to provide 2,4-dichloro-6-(pyridin-4-yl) furo[3,2-d]pyrimidine (1.6 g, 6.9 mmol) as a yellow solid.
  • EXAMPLE 23 Compound 90 using General Synthetic Route 23: 1.1) Synthesis of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine [0414] To a solution of 2,4-dichlorofuro[3,2-d]pyrimidine (6.46 g, 34.2 mmol mmol) in 1,4- dioxane (100 mL) was added morpholine (5.95 g, 68.4 mmol). The reaction was stirred at room temperature for 30 min.
  • EXAMPLE 24 Compound 91 using General Synthetic Route 24: 1.1) Synthesis of 2-chloro-4-morpholino-6-(pyridin-2-yl)furo[3,2-d]pyrimidine [0419] A solution of 2-chloro-6-iodo-4-morpholinofuro[3,2-d]pyrimidine (1 g, 2.7 mmol), 2- (tributylstannyl)pyridine (1.2 g, 3.3 mmol) and Pd(PPh3)4 (155 mg, 0.14 mmol) in toluene (5 mL) was heated to 90 °C overnight.
  • EXAMPLE 25 Compound 92 using General Synthetic Route 25: 1.1) Synthesis of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid
  • 2-chloro-4-morpholinofuro[3,2-d]pyrimidine 2.4 g, 10 mmol
  • n-BuLi 5.2 mL, 2.5 M, 13 mmol
  • the reaction mixture was stirred at that temperature for 1 h.
  • To the solution was added excessive amount of dry ice in one portion.
  • the resulting reaction mixture was stirred at that temperature for 3 h.
  • EXAMPLE 26 Compound 98 using General Synthetic Route 26: 1.1) Synthesis of 4-morpholino-2-(3-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d]pyrimidine [0426] To a solution of 2- chloro-4-morpholinofuro[3,2-d]pyrimidine (300 mg, 1.25 mmol) in DMF (4 mL) was added 3-(m-tolyl)-1H-pyrazole (237 mg, 0.35 mmol), Cs 2 CO 3 (815 mg, 2.5 mmol) and CuI (24 mg, 0.125 mmol). The reaction was stirred at 110 °C overnight.
  • EXAMPLE 27 Compound 127 using General Synthetic Route 27: 1.1) Synthesis of 2-chloro-7-iodo-4-morpholinofuro[3,2-d]pyrimidine [0430] To a solution of 2-chloro-6-iodo-4-morpholinofuro[3,2-d]pyrimidine (5.0 g, 13.7 mmol) in anhydrous THF (30 mL) at -78 °C under N2 was added LDA (14 mL, 2 M, 27.4 mmol). The reaction mixture was stirred at -78 °C for 1 h.
  • reaction mixture was stirred at that temperature for 1 h. To the solution was added excessive amount of dry ice in one portion. The resulting reaction mixture was stirred at that temperature for 1 h. Upon the completion of the reaction as monitored by TLC, the reaction was quenched with water and the pH was adjusted to 5 using 1 N HCl aqueous solution. The aqueous solution was extracted with DCM (2 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was slurry in Et 2 O to provide 2-chloro-7-methyl-4- morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid (60 mg, 0.20 mmol) as a yellow solid.
  • reaction solution was concentrated directly and the resulting residue was dissolved in DCM (20 mL). To the solution was added cyclopropanamine (23 mg, 0.4 mmol), followed by Et 3 N (40 mg, 0.4 mmol) dropwise. The completion of the reaction was monitored by TLC. The reaction was quenched with water (20 mL) and extracted with EtOAc (2 x 10 mL). The combined organic phase was dried over anhydrous Na 2 SO 4 , filtrated and concentrated under reduced pressure.
  • EXAMPLE 28 Compound 122 using General Synthetic Route 28: 1.1) Synthesis of 2-chloro-6-iodo-7-met hyl-4-morpholinofuro[3,2-d]pyrimidine [0436] To a solution of 2-chloro-7-methyl-4-morpholinofuro[3,2-d]pyrimidine (350 mg, 1.38 mmol) in anhydrous THF (30 mL) at -78 °C under N2 was added LDA (1.4 mL, 2 M, 2.76 mmol). After stirred at -78 °C for 1 h, to the solution was added a solution of NIS (374 mg, 1.66 mmol) in anhydrous THF (5 mL).
  • EXAMPLE 29 Compound 112 using General Synthetic Route 29: 1.1) Synthesis of 2-chloro-4-morpholino-7-(trifluoromethyl)furo[3,2-d]pyrimidine F C [0440] A solid mixture of 2-chloro-7-iodo-4-morpholinofuro[3,2-d]pyrimidine (300 mg, 0.82 mmol), KF (144 mg, 2.47 mmol), CuI (30 mg, 0.16 mmol) and 1,10-phenanthroline (30 mg, 0.16 mmol) in three neck flask was heated to 100 °C under reduced pressure using oil pump for 1 h.
  • EXAMPLE 30 Compound 134 using General Synthetic Route 30: 1.1) Synthesis of 2,4-dichlorofuro[3,2-d]pyrimidine-6-carboxylic acid
  • LDA LDA
  • reaction mixture was stirred at 80 °C for 1 h. Upon the completion of the reaction as monitored by TLC, the reaction was quenched with water (30 mL) and extracted with EtOAc (2 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 30% EtOAc/PE to provide 2-chloro-N-ethyl-4-(2,2,6,6-tetrafluoromorpholino)furo[3,2-d]pyrimidine-6- carboxamide (83 mg, 0.22 mmol) as a brown solid.
  • EXAMPLE 31 Compound 133 using General Synthetic Route 31: 1.1) Synthesis of 2,4-dichloro-6-iodofuro[3,2-d]pyrimidine [0450] To a solution of 2,4-dichlorofuro[3,2-d]pyrimidine (1.0 g, 5.32 mmol) in anhydrous THF (30 mL) at -78 °C under N 2 was added n-BuLi (5.33 mL, 2.5M, 13.3 mmol). After stirred at -78 °C for 1 h, to the solution was added a solution of NIS (1.44 g, 6.38 mmol) in anhydrous THF (10 mL).
  • the reaction was stirred at 80 °C for 1 h. Upon the completion of the reaction as monitored by TLC, the reaction was cooled to room temperature, quenched with water (30 mL) and extracted with DCM (4 x 10 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 30% EtOAc/PE to provide 2-chloro-6-(pyridin-3-yl)- 4-(2,2,6,6-tetrafluoromorpholino)furo[3,2- d]pyrimidine (97 mg, 0.25 mmol) as a brown solid.
  • the reaction was stirred at 110 °C for 5 h. Upon the completion of the reaction as monitored by TLC, the reaction was cooled to room temperature, quenched with water (10 mL) and extracted with DCM/MeOH (15/1, 3 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by preparative TLC to provide 6-(pyridin-3-yl)-4-(2,2,6,6- tetrafluoromorpholino)-2-(4-(m-tolyl)-1H-pyrazol-1- yl)furo[3,2-d]pyrimidine (7.5 mg, 0.014 mmol) as a white solid.
  • EXAMPLE 32 Compound 143 using General synthetic route 32: N N N Cl Sn N Cl I O N N N O N N P d(PPh3)4 /CuCl/LiCl/THF N O O 1 2 N HN N N N N N O N Cu 2 O/Cs 2 CO 3 /DMF N O
  • Compound 143 1) Synthesis of 2-chloro-4-morpholino-6-(pyrimidin-4-yl)furo[3,2-d]pyrimidine N Cl N N O N N O
  • a suspension of 2-chloro-6-iodo-4-morpholinofuro[3,2-d]pyrimidine 80 mg, 0.22 mmol
  • 4-(tributylstannyl)pyrimidine (128 mg, 0.34 mmol)
  • EXAMPLE 33 Compound 156 using General synthetic route 33: 1) Synthesis of 2-chloro-4-morpholino-6-(pyridin-3-yl)furo[3,2-d]pyrimidine [0458] To a solution of 2-chloro -6-iodo-4-morpholinofuro[3,2-d]pyrimidine (500 mg, 1.37 mmol) in 1,4-dioxane/H 2 O (2/1, 30 mL) was added pyridin-3-ylboronic acid (185 mg, 1.51 mmol), K 2 CO 3 (378 mg, 2.74 mmol) and PdCl 2 (PPh 3 ) 2 (48 mg, 0.068 mmol) under N 2 .
  • EXAMPLE 34 Compounds 145 and 146 using General synthetic route 34: H I 1) Synthesis of tert-butyl 5-(2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)-3-methyl-1H- pyrazole-1-carboxylate N [0460] A suspension of 2-chloro-6-iodo-4-morpholinofuro[3,2-d]pyrimidine (5.72 g, 15.67 mmol), (1-(tert-butoxycarbonyl)-3-methyl-1H-pyrazol-5-yl)boronic acid (3.90 g, 17.24 mmol), Pd(PPh)2Cl2 (2.20 g, 3.13 mmol) and CsF (7.15 g, 47.01 mmol) in 1,4-dioxane/H 2 O (4/1, 330 mL) under N2 was heated to 80 °C for 1 h.
  • the mixture was stirred at 50 °C for 2 h. The completion of the reaction was monitored by TLC.
  • the reaction mixture was quenched with water and extracted with EtOAc (3 x 10 mL). The combined organic phase was dried over anhydrous Na 2 SO 4 , filtrated and concentrated under reduce pressure.
  • EXAMPLE 35 Compound 148 using General synthetic route 35: 1) Synthesis of 2,4-dichloro-6-(pyridin-2-yl)furo[3,2-d]pyrimidine [0467] To a solution of 2,4-dichloro-6-iodofuro[3,2-d]pyrimidine (2.3 g, 7.3 mmol) in DMF (60 mL) under N2 was added 2-(tributylstannyl)pyridine (2.7 g, 7.3 mmol), CuI (416 mg, 2.2 mmol) and PdCl 2 (dppf) (534 mg, 0.73 mmol). The reaction was stirred at 100 °C for 3 h. The completion of the reaction was monitored by TLC.
  • reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 5% MeOH/DCM to 10% MeOH/DCM to provide 6-(pyridin-2-yl)-2-(3-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d]pyrimidin- 4-ol (100 mg, 0.27 mmol) as a green solid.
  • EXAMPLE 36 Compound 150 using General synthetic route 36: 1) Synthesis of (2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)methanol [0474] To a solution of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid (1 g, 3.53 mmol) in THF (10 mL) at 0 °C was added BH 3 /THF (1 mol/L, 14 mL) dropwise. The reaction mixture was stirred at rt overnight. The completion of the reaction was monitored by TLC. The reaction was quenched with 1N HCl. The mixture was heated under reflux for 2h.
  • EXAMPLE 37 Compound 192 using General synthetic route 37: 1)Synthesis of tert-butyl 4-(3-chlorophenyl)-1H-pyrazole-1-carboxylate [0480] To a solution of tert-butyl 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole -1-carboxylate (1 g, 4.20 mmol) in 1,4-dioxane/H 2 O (10/1, 20 mL) was added 1-chloro-3- iodobenzene (1.24 g, 1.51 mmol), CsF (958 mg, 6.30 mmol) and PdCl 2 (PPh 3 ) 2 (295 mg, 0.42 mmol) under N2.
  • the reaction was stirred at 110 °C overnight. The completion of the reaction was monitored by TLC.
  • the reaction mixture was quenched with water (50 mL).
  • the aqueous solution was extracted with DCM (3 x 30 mL).
  • the combined organic phase was dried over anhydrous Na 2 SO 4 , filtrated and concentrated under reduce pressure.
  • EXAMPLE 38 Compound 157 using General synthetic route 38: 1)Synthesis of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid
  • 2-chloro-4-morpholinofuro[3,2-d]pyrimidine 2.4 g, 10 mmol
  • n-BuLi 5.2 mL, 2.5 M, 13 mmol
  • the reaction mixture was stirred at that temperature for 1 h.
  • dry ice 4.4 g, 100 mmol
  • the resulting reaction mixture was stirred at that temperature for 3 h.
  • EXAMPLE 39 Compound 179 using General synthetic route 39: 1)Synthesis of 2-chloro-6-iodo-4-morpholinofuro[3,2-d]pyrimidine [0488] To a solution of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine (2.0 g, 0.83 mmol) in THF (30 mL) at -78 °C under N 2 was added LDA (1.33 mL, 2M, 2.66 mmol). After stirred at - 78 °C for 1 h, to the solution was added a solution of NIS (2.25 g, 1.0 mmol) in THF (10 mL). The completion of the reaction was monitored by TLC.
  • EXAMPLE 40 Compound 147 using General synthetic route 40: 1)Synthesis of 2-chloro-4-morpholino-7-(trifluoromethyl)furo[3,2-d]pyrimidine [0495] To a solution of 2-chloro-7-iodo-4-morpholinofuro[3,2-d]pyrimidine (300 mg, 0.82 mmol) in anhydrous DMSO (6 mL) was added KF (144 mg, 2.47 mmol), CuI (30 mg, 0.16 mmol), 1,10-phenanthroline (30 mg, 0.16 mmol), B(OMe) 3 (252 mg, 2.47 mmol) and TMSCF 3 (348 mg, 2.47 mmol).
  • the reaction was stirred at 80 °C under microwave for 30 min.
  • the reaction mixture was diluted with water and extracted with DCM (3 x 20 mL).
  • the combined organic phase was dried over anhydrous Na 2 SO 4 , filtrated and concentrated under reduce pressure.
  • the resulting residue was purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 2% MeOH/DCM to provide N-cyclopropyl-4- morpholino-2-(4-(m-tolyl)-1H-pyrazol-1-yl)-7-(trifluoromethyl)furo[3,2-d]pyrimidine-6- carboxamide (13 mg, 0.025 mmol) as a yellow solid.
  • EXAMPLE 41 Compound 204 using General synthetic route 41: 1) Synthesis of 4-morpholino-2-(3-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d]pyrimidine [0500] To a solution of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine (300 mg, 1.25 mmol) in DMF (4 mL) was added 3-(m-tolyl)-1H-pyrazole (237 mg, 0.35 mmol), Cs 2 CO 3 (815 mg, 2.5 mmol) and CuI (24 mg, 0.125 mmol). The reaction mixture was stirred at 110 °C overnight. The reaction mixture was quenched with water (10 mL).
  • EXAMPLE 42 Compound 212 using General synthetic route 42: 1) Synthesis of (2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)boronic acid [0504] To a solution of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine (200 mg, 0.84 mmol) in THF (30 mL) at -78 °C under N 2 was added n-BuLi (0.4 mL, 2.5 M, 1.00 mmol). After stirred at -78 °C for 1 h, to the solution was added a solution of triisopropyl borate (190 mg, 1.00 mmol) in THF (2 mL).
  • EXAMPLE 43 Compound 144 using General synthetic route 43: l 1) Synthesis of 2-chloro-6-(3-methyl-1H-pyrazol-5-yl)-4-morpholinofuro[3,2-d]pyrimidine [0509] A suspension of 2-chloro-6 -iodo-4-morpholinofuro[3,2-d]pyrimidine (400 mg, 1.08 mmol), (3-methyl-1H-pyrazol-5-yl)boronic acid (154 mg, 1.1 mmol), Na2CO3 (232 mg, 2.16 mmol) and Pd(PPh 3 ) 4 (12 mg, 0.01 mmol) in 1,4-dioxane/H 2 O (8 mL, 4:1) under N 2 was heated to 50 °C for 2 h.
  • reaction mixture was stirred at 160 °C in a sealed tube overnight. The completion of the reaction was monitored by TLC.
  • the reaction mixture was concentrated directly and purified by preparative TLC with a elution of 10% MeOH/DCM to provide 6-(3-methyl-1H-pyrazol-5-yl)-4-morpholino-2-(4-(m- tolyl)-1H-pyrazol-1-yl)furo[3,2-d]pyrimidine (18 mg, 0.041 mmol) as a yellow solid.
  • PIKfyve Full length human recombinant PIKFYVE expressed in baculovirus expression system as N-terminal GST-fusion protein (265 kDa) was obtained from Carna Biosciences (Kobe, Japan).
  • the kinase substrate was prepared by mixing and sonicating fluorescently-labeled phosphatidylinositol 3-phosphate (PI3P) with phospho-L-serine (PS) at a 1:10 ratio in 50 mM HEPES buffer pH 7.5.
  • PI3P fluorescently-labeled phosphatidylinositol 3-phosphate
  • PS phospho-L-serine
  • Kinase protein was pre-diluted in an assay buffer comprising 25 mM HEPES, pH 7.5, 1 mM DTT, 2.5 mM MgCl2, and 2.5 mM MnCl2, and 0.005% Triton X-100, and dispensed into a 384-well plate (10 ⁇ L per well).
  • Test compounds were serially pre-diluted in DMSO and added to the protein samples by acoustic dispensing (Labcyte Echo). The concentration of DMSO was equalized to 1% in all samples. All test compounds were tested at 12 concentrations. Apilimod was used as a reference compound and was tested in identical manner in each assay plate.
  • Control samples (0%-inhibition, in the absence of inhibitor, DMSO only) and 100%-inhibition (in the absence of enzyme) were assembled in replicates of four and were used to calculate %-inhibition in the presence of compounds.
  • the reactions were initiated by addition of 10 ⁇ L of 2x PI3P/PS substrate supplemented with ATP.
  • the final concentration of enzyme was 2 nM, the final concentration of ATP was 10 mM, and the final concentration of PI3P/PS substrate was 1 ⁇ M (PI3P).
  • the kinase reactions were allowed to proceed for 3 h at room temperature.
  • Terminated plates were analyzed on a microfluidic electrophoresis instrument (Caliper LabChip® 3000, Caliper Life Sciences/Perkin Elmer). The change in the relative fluorescence intensity of the PI(3)P substrate and PI(3,5)P product peaks was measured. The activity in each test sample was determined as the product to sum ratio (PSR): P/(S+P), where P is the peak height of the product, and S is the peak height of the substrate.
  • PSR product to sum ratio
  • IC 50 of test compounds 50%-inhibition
  • the %-inh cdata (P inh versus compound concentration) were fitted by a four-parameter sigmoid dose- response model using XLfit software (IDBS).
  • IC50 values for certain compounds of the disclosure are provided in Table 6 below.
  • Table 6 Biological Example 2 Antiviral effects of PIKfyve inhibitors in a Vero-E6 SARS-CoV-2 cytopathic assay
  • Materials and Methods [0518] An in vitro antiviral assay to test compounds acting against SARS-CoV-2 was designed and run based on cytopathic effect (CPE) in Vero (African green monkey kidney epithelial) cells essentially according to the methods of Ivens et al. (2005) Journal of Virological Methods 129, 56-63.
  • CPE cytopathic effect
  • VeroE6-EGFP cells (provided by Lab of Virology & Chemotherapy, Rega Institute, KU Leuven, Leuven, Belgium) (sometimes referred to herein as VeroE6, Vero-E6, or Vero-E6- GFP) were propagated in growth medium which was prepared by supplementing DMEM (Gibco 41965-039) with 10% v/v heat-inactivated FCS and 5 mL sodium bicarbonate 7.5% (Gibco 25080-060). Cells were cultured in T150 bottles and split 1/4 twice a week. Pen-strep was added directly to the T150 bottle at a 1/100 dilution.
  • Assay medium was prepared by supplementing DMEM (Gibco 41965-039) with 2% v/v heat-inactivated FCS and 5 mL sodium bicarbonate 7.5% (Gibco 25080-060). [0521] Serial dilutions of compounds were prepared in 96-well plates to which cells were added (25,000 cells/well) after which plates were incubated overnight (37°C; 5% CO 2 ).
  • virus inoculum SARS-CoV-2 clinical isolate, Belgian strain: BetaCov/Belgium/GHB- 03021/2020
  • CPE cytopathic effect
  • GFP signal was determined using high-content imaging.
  • Antiviral activity is expressed as the EC50 or concentration required to rescue 50% of the GFP signal from the virus-induced cytopathogenicy. The signal is provided as the logarithm of the surface of the well that is covered with fluorescent pixels which correlates with living cells.
  • cytotoxicity was assessed by growing uninfected cells in the presence of the test compound at the concentrations tested. After a 4 day incubation, cell viability was measured using a commercial kit.
  • Antiviral readout was performed using high-content imagers. Using a 5x objective, almost the entire well of a 384-well plate is captured at once (for an 96-well plate, the well is covered by 4 field of views). A GFP marker located in both the cell cytoplasm as well as the nucleus allowed for the calculation of the surface of the well that is (still) covered by cells (SpotTotalAreaCh2).
  • PIKfyve inhibitors were assessed for anti-viral activity against SARS-COV-2 using A549-ACE2 cells (adenocarcinomic human alveolar basal epithelial cells) transduced to express the human Angiotensin-converting enzyme 2 (ACE2), provided by Institut Pasteur, Paris, France.
  • A549-ACE2 cells were grown in 96-well plates, and ten concentrations of each tested PIKfyve inhibitor was tested in triplicate.
  • Viral replication was measured by quantitative RT-PCR in the presence and absence of the tested compounds.
  • Compounds 171 and 163 were also tested.
  • Compound 171 has EC 50 of 55.85 ⁇ M;
  • Compound 163 has EC 50 of 4.228 ⁇ M.
  • Both compounds 163 and 171 have CC 50 of >10000 nM.
  • Biological Example 4 Reduction of virus-induced cytopathic effect (primary CPE assay) [0534] Materials and Methods [0535] Confluent or near-confluent cell culture monolayers of Vero 76 (African green monkey kidney epithelial cells, are provided by Institute for Antiviral Research, Utah State University, Utah, USA) cells and are prepared in 96-well disposable microplates the day before testing.
  • Cells are maintained in MEM supplemented with 5% FBS.
  • FBS reduced to 2% and supplemented with 50- ⁇ g/ml gentamicin.
  • Compounds are dissolved in DMSO.
  • the test compounds are prepared at four serial log10 concentrations, 0.1, 1.0, 10, and 100 ⁇ g/ml or ⁇ M. Five microwells are used per dilution: three for infected cultures and two for uninfected toxicity cultures. Controls for the experiment consisted of six microwells that are infected and not treated (virus controls) and six that are untreated and uninfected (cell controls) on every plate.
  • Growth media is removed from the cells and the test compound is applied in 0.1 ml volume to wells at 2X concentration.
  • Medium devoid of virus is placed in toxicity control wells and cell control wells. Plates are incubated at 37°C with 5% CO 2 until marked CPE (>80% CPE for most virus strains) is observed in virus control wells.
  • the plates are then stained with 0.011% neutral red for approximately two hours at 37°C in a 5% CO2 incubator.
  • the neutral red medium is removed by complete aspiration, and the cells are optionally rinsed 1X with phosphate buffered solution (PBS) to remove residual dye.
  • PBS phosphate buffered solution
  • the PBS is completely removed, and the incorporated neutral red is eluted with 50% Sorensen’s citrate buffer/50% ethanol for at least 30 minutes.
  • Neutral red dye penetrates into living cells, thus, the more intense the red color, the larger the number of viable cells present in the wells.
  • the dye content in each well is quantified using a spectrophotometer at 540 nm wavelength.
  • the dye content in each set of wells is converted to a percentage of dye present in untreated control wells using a Microsoft Excel computer-based spreadsheet and normalized based on the virus control.
  • the 50% effective (EC50, virus-inhibitory) concentrations and 50% cytotoxic (CC50, cell-inhibitory) concentrations are then calculated by regression analysis.
  • the quotient of CC50 divided by EC50 gives the selectivity index (SI) value. Compounds showing SI values >10 are considered active.
  • Example 5 Anti-viral Activity Against Human Alphacoronavirus Strain 229E (HCoV 229E) and Human Betacoronavirus Strain OC43 (HCoV OC43)
  • HCV 229E Human Alphacoronavirus Strain 229E
  • HoV OC43 Human Betacoronavirus Strain OC43
  • the tested compounds were serially diluted (8-point, 3-fold dose titration) and added prophylactically for an hour, to cells. Cells were then infected with virus for one hour, at a single infective dose (100x median tissue culture infectious dose; TCID50). Additional media was added to the wells, with equivalent concentrations of compound, for the duration of the study. Vehicle and positive control wells were set up to control for any influence on cell viability. Cells were visually inspected daily for the appearance of any cytophathic/cytopathogenic effect (CPE).
  • CPE cytophathic/cytopathogenic effect
  • EC50 values were calculated using non-linear regression (log(agonist) vs. response – Variable slope (four parameters)) using log(concentration).
  • CC50 values were calculated used non-linear regression as above for EC50.
  • HCoV 229E was tested with human bronchial epithelial (16BHE) cells.
  • HCoV OC43 was tested with human lung mucoepidermoid (H 2 92) cells. Remdesivir was used as the control compound.
  • Compound 114 was also tested and had EC50 ⁇ 10 ⁇ M and CC50 of 1.02. Data not shown.
  • Efficacy against HCoV OC43 in H292 cells – Compound 91 showed efficacy against HCoV OC43, with an EC50 of 0.08 ⁇ M ( Figure 4A) and CC50 ⁇ 10 ⁇ M.
  • Compound 91 increased cell viability to levels that were higher than the uninfected cells, leading to percentages above 100%. The viability in infected cells was reduced to below 0% at 10 ⁇ M, 3.16 ⁇ M, and 1 ⁇ M. In order to calculate a more representative EC50, these data points were excluded from the calculation.
  • Compound 114 showed efficacy against HCoV OC43, with an EC 50 of 0.31 ⁇ M ( Figure 4B). Compound 114 increased cell viability to levels that were higher than the uninfected cells, leading to percentages above 100%. The viability in infected cells was reduced to below 0% at 10 ⁇ M and 3.16 ⁇ M. In order to calculate a more representative EC 50 , these data points were excluded from the calculation. The compound had a CC50 ⁇ 10 ⁇ M. [0552] Compound 121 showed efficacy against HCoV OC43, with an EC50 of 0.99 ⁇ M ( Figure 4C).
  • Compound 121 increased cell viability to levels that were higher than the uninfected cells, leading to percentages above 100%. No reduction in viability in infected cells was seen at the tested concentrations. The compound had a CC50 ⁇ 10 ⁇ M.
  • Biological Example 6 PIKfyve inhibition of SARS CoV2–induced cytopathic effect (CPE) in Vero E6 cells [0553] Materials and Methods [0554] Five PIKfyve inhibitors were assessed for anti-viral activity against SARS-CoV-2, strain USA_WA1/2020, using Vero E6 and Vero 6 cells according to the methods of Severson et al. (2007) Journal of Biomolecular Screening 33-40.
  • Results [0556] A summary of the results is shown in Table 7. The tested compounds showed potent antiviral effects with minimal toxicity. Table 7. Biological Example 7: Drug Drug Interaction Study with Cytochrome P450 Enzymes [0557] Preparation of stock solutions [0558] The stock solutions of test compound were prepared in dimethyl sulfoxide (DMSO) at 10 mM concentration, then diluted to 2 mM with DMSO. The final concentration of test compound was 10 ⁇ M. [0559] Preparation of positive inhibitors [0560] The concentration of positive inhibitor was listed in Table 8.
  • DMSO dimethyl sulfoxide
  • Solution B was prepared by adding 3.400 g of potassium dihydrogen phosphate to 250 mL of pure water, followed by sonication. Solution A was placed on a stirrer and Solution B was added slowly into Solution A until the pH reached 7.4. [0565] 10 mM NADPH solution was prepared, fresh prior to use, by dissolving nicotinamide adenine dinucleotide phosphate (NADPH) at 8.334 mg/mL in phosphate buffer. [0566] Preparation of master solution [0567] The master solution was prepared according to Table 10. The incubation was carried out in 96 deep well plates.
  • the following volumes were dispensed into each well of the incubation plate, 179 ⁇ L of the substrate and human liver microsomes (HLM) mixture in phosphate buffer, 1 ⁇ L of the compound working solution, or vehicle (mixture of DMSO and acetonitrile (1:4)).
  • the incubation plate was placed into the water bath and pre-warmed at 37°C for 15 minutes before the reactions were started by the addition of 20 ⁇ L of 10 mM NADPH solution in phosphate buffer. After the addition of NADPH, the incubation plate was incubated at 37°C for the corresponding time. The assay was performed in duplicate. Table 10.
  • HTS Transwell ® plates were incubated at 37 °C, 5% CO 2 for 1 hour before cell seeding.
  • MDCK-MDR1 cells were diluted to 1.56 ⁇ 10 6 cells/mL with culture medium and 50 ⁇ L of cell suspension were dispensed into the filter well of the 96-well HTS Transwell plate. Cells were cultivated for 4-8 days in a cell culture incubator at 37 °C, 5% CO 2 , and 95% relative humidity. Cell culture medium was replaced every other day, beginning no later than 24 hours after initial plating.
  • Preparation of Stock Solutions [0576] Stock solutions of the test compounds and of the positive controls were prepared in dimethyl sulfoxide (DMSO) at the concentration of 10 mM.
  • DMSO dimethyl sulfoxide
  • Metoprolol and Digoxin were used as the control compounds.
  • Assessment of Cell Monolayer Integrity [0578] Medium was removed from the reservoir and each Transwell insert and replaced with prewarmed fresh culture medium. Transepithelial electrical resistance (TEER) across the monolayer was measured using Millicell® Epithelial Volt-Ohm measuring system (Millipore®, USA).
  • TEER value should be greater than 42 ohm•cm 2 , which indicates the well-qualified MDCK-MDR1 monolayer.
  • Assay Procedures [0582] The MDCK-MDR1 plate was removed from the incubator and washed twice with pre- warmed Hanks' Balanced Salt solution (HBSS) (10 mM HEPES, pH 7.4), and then incubated at 37 °C for 30 minutes.
  • HBSS Hanks' Balanced Salt solution
  • the stock solutions of control compounds were diluted in DMSO to get 200 ⁇ M solutions and then diluted with HBSS (10 mM HEPES, pH 7.4) to get 1 ⁇ M working solutions.
  • the test compounds were diluted in DMSO to get 200 ⁇ M solutions and then diluted with HBSS (10 mM HEPES with 4% BSA, pH 7.4) to get 1 ⁇ M working solutions.
  • the final concentration of DMSO in the incubation system was 0.5%.
  • the plates were Incubated at 37 °C for 30 mins.80 ⁇ L samples were removed directly from the apical and basolateral wells (using the basolateral access holes) and transferred to wells of new 96 wells plates.
  • the Lucifer Yellow fluorescence (to monitor monolayer integrity) signal was measured in a fluorescence plate reader at 480 nM excitation and 530 nM emission.
  • %LY leakage 100 ⁇ [LY]acceptor/([LY]donor+[LY]acceptor) LY leakage of ⁇ 1% is acceptable to indicate the well-qualified MDCK-MDR1 monolayer.
  • Results for this Example are shown in Table 15.
  • Biological Example 9 Hepatocyte Drug Metabolism Study [0592] Preparation of Working Solutions [0593] 10 mM stock solutions of test compound and positive controls were prepared in DMSO.
  • the cells were thawed by placing the vial in a 37°C water bath and gently shaking the vials for 2 minutes. After thawing was completed, the vial was sprayed with 70% ethanol, and transferred to a biosafety cabinet. [0597] The hepatocytes were transferred into 50 mL conical tube containing thawing medium. The 50 mL conical tube was placed into a centrifuge and spun at 100 g for 10 minutes. Upon completion of spin, the thawing medium was aspirated and the hepatocytes resuspended in enough incubation medium to yield ⁇ 1.5 ⁇ 106 cells/mL.
  • AO/PI Staining was used to count cells and determine the viable cell density, after which, the cells were diluted with incubation medium to a working cell density of 0.5 ⁇ 106 viable cells/mL.
  • Procedure for Stability Determination [0600] 198 ⁇ L of hepatocytes were pipetted into each wells of a 96-well non-coated plate, which was then placed in the incubator to warm the hepatocytes for 10 minutes.2 ⁇ L of the 100 ⁇ M test compound or positive control were pipetted into respective wells of the 96-well non- coated plate to start the reaction, and the plate was returned to the incubator for the designed time points.
  • Table 12 shows scaling factors for intrinsic clearance prediction in human, monkey, dog, rat and mouse hepatocytes. Table 12. [0607] The rules for data processing are shown in Table 13. Table 13. [0608] Results for this example are shown in Table 14. Table 14. Biological Example 10: Antiviral effects of PIKfyve inhibitors in a Vero-E6 SARS-CoV-2 cytopathic assay [0609] Materials and Methods [0610] To determine the half maximal inhibitory concentration (IC 50 ) of the compounds against SARS-CoV-2, anti-viral screening was performed against two (2) SARS-CoV-2 strains, Wildtype (WT) and Delta (D). [0611] Adherent Vero-E6 cells were seeded in multi-well plates.
  • Cell cultures were inoculated with a standardized amount of virus, in the absence and presence of serial compound dilutions, followed by 18-24 hours of incubation and an appropriate virus detection method (e.g., Immunostaining). Eight 3.16-fold dilutions of the compounds (lowest concentration 1 nM) were added to the assay cells for 1h before infection with each SARS-CoV-2 strain respectively. Virus signals detected in the presence of each compound concentration were used to determine the IC50. After infection, cells were fixed and immunostained for a viral antigen, and the percentage of infected cells quantified relative to an infected, untreated control using high content imaging.
  • an appropriate virus detection method e.g., Immunostaining

Landscapes

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

Abstract

The present disclosure provides compounds that are useful for the treatment of coronavirus infections.

Description

METHODS AND TREATMENT OF VIRAL INFECTION WITH SUBSTITUTED FURO-PYRIMIDINES CROSS-REFERENCE TO RELATED APPLICATIONS [0001] This application claims priority to U.S. Provisional Application No.63/208,459, filed on June 8, 2021, the disclosure of which is hereby incorporated by reference in its entirety. FIELD OF THE INVENTION [0002] The present disclosure relates to methods of i) blocking alpha-coronavirus, beta- coronavirus lineage B, and beta-coronavirus lineage A into a host cell; and ii) preventing and treating an infection caused by alpha-coronavirus, beta-coronavirus lineage B, and beta- coronavirus lineage A with compounds that are phosphoinositide kinase inhibitors, in particular FYVE-type finger-containing phosphoinositide kinase (“PIKfyve”) inhibitors. BACKGROUND OF THE INVENTION [0003] According to the World Health Organization, there have been over 517 million confirmed cases and about 6.3 million deaths worldwide due to COVID-19 as of May 2022. SARS-CoV-2, which is responsible for the COVID-19 (2019 novel coronavirus (2019-nCoV) disease, is an enveloped, positive-sense, RNA virus that belongs to the Betacoronavirus genus. Bouhaddou et al, Cell 182: 1-28 (2020). Improved understanding of key steps in viral entry and ways to disrupt them can lead to the development of effective antiviral drugs, not only for COVID-19, but for future viral outbreaks as well. Treatments for COVID-19 are greatly needed. [0004] Coronavirus entry into susceptible cells is a complex process that requires the concerted action of receptor-binding and proteolytic processing of the coronavirus S protein to promote virus-cell fusion. Heald-Sargent and Gallagher, Viruses 4:557-580 (2012); Millet and Whittaker, Virus Res.202:120-134 (2015). For example, viral entry into cells may be mediated by a viral glycoprotein (GP), which attaches viral particles to the cell surface, delivers them to endosomes, and catalyzes fusion between viral and endosomal membranes. Murray, et al, J. Virology 79:11742-11751 (2005). In the case of SARS-CoV-2, its spike (S) protein binds to an ACE2 receptor on the target cell and is subsequently primed by a serine protease, TMPRSS2, that cleaves the S protein and allows fusion of viral and lysosomal membranes. Hoffmann, et al., Cell 181:271-280.e8 (2020). [0005] Like most viruses, coronavirus membrane fusion with host cell membrane takes place within acidified endosomes. Kang, et al, PNAS 117(34):20803-20813 (2020). Coronaviruses are known to interact with phosphatidylinositol (PI) kinases, which are distributed across various subcellular compartments. Beziau et al, Viruses 12, 1124; doi:10.3390/v12101124 (2020). The PI kinase phosphatidylinositol 3-phosphate 5 kinase (PIKfyve) involved in endosomal acidification is required for SARS-CoV-2 to enter the cell. Ou, et al, Nat. Commun.11(1620):1- 12 (2020). PIKfyve inhibition with small molecule inhibitors has been shown to inhibit SARS- CoV-2 infection. Kang et al, PNAS 117(34):20803-20813 (2020); Nelson, et al, PLoS Negl. Trop. Dis.11(4):e0005540 (2017). Ablation of PIKfyve arrests endosomal maturation (Ikonomov et al, J. Biol. Chem.276(28):26141-26147 (2001); Ikonomov et al, J. Biol. Chem. 277(11):9206-9211 (2002); Jefferies et al, EMBO Rep.9(2):164-170 (2008); Rutherford, et al, J. Cell Sci.119(19):3944-3957 (2006); Sbrissa, et al, J. Biol. Chem.277(8):6073-6079 (2002)), which makes PIKfyve a candidate target for new drugs against viruses that exploit the endosomal pathway. SUMMARY OF THE INVENTION [0006] Embodiment 1 is a method of blocking alpha-coronavirus, beta-coronavirus lineage B, and/or beta-coronavirus lineage A entry into a host cell and preventing an infection caused by alpha-coronavirus, beta-coronavirus lineage B, and beta-coronavirus lineage A, comprising administering to a subject in need thereof a compound of (i) Formula (I)
Figure imgf000004_0001
wherein: R1a and R1b taken together with the nitrogen to which they are attached form:
Figure imgf000004_0002
wherein X and Y are independently N or CRa; wherein Ra is H or C1-4alkyl; and Rb is phenyl, monocyclic cycloalkyl, monocyclic heterocyclyl, monocyclic heterocycloalkyl, or monocyclic heteroaryl, each optionally substituted with one, two, or three Rd substituents; or R1a is H or C1-4alkyl; and R1b is a heteroaryl optionally substituted with Rc; wherein Rc is C1-4alkyl, phenyl, -C1-4alkyl-phenyl, monocyclic cycloalkyl, -C1-4alkyl-(monocyclic cycloalkyl), monocyclic heterocyclyl, monocyclic heterocycloalkyl, monocyclic heteroaryl, or -C1-4alkyl-(monocyclic heteroaryl), wherein each alkyl, phenyl, cycloalkyl, heterocyclyl, heterocycloalkyl or heteroaryl is optionally substituted with one, two, or three Rd substituents; wherein each Rd substituent is independently C1-4alkyl, C1-4alkenyl, C1-4alkynyl, -O-C1-4alkyl, halo, cyano, nitro, azido, C1-4haloalkyl, -O-C1-4-haloalkyl, -NRgRh, -NRgC(=O)Rh, -NRgC(=O)NRgRh, -NRgC(=O)ORh, =NORg, -NRgS(=O)1-2Rh, -NRgS(=O)1-2NRgRh, =NSO2Rg, -C(=O)Rg, -C(=O)ORg, -OC(=O)ORg, -OC(=O)Rg, -C(=O)NRgRh, -OC(=O)NRgRh, -ORg, -SRg, -S(=O)Rg, -S(=O)2Rg, -OS(=O)1-2Rg, -S(=O)1-2ORg, or -S(=O)1-2NRgRh; wherein Rg and Rh are each independently H or C1-4alkyl; each of R2 and R3 is independently chosen from H, C1-4alkyl, cycloalkyl, C1- 4alkylcycloalkyl, heterocyclyl, heterocycloalkyl, and heteroaryl optionally substituted with one, two, or three Rj substituents; or R2 and R3 taken together with the nitrogen to which they are attached form a heterocyclyl, optionally substituted with one, two, three, or four Rj substituents, or further wherein any of the hydrogens bonded to carbon atoms are optionally replaced by deuterium; wherein each Rj substituent is independently C1-4alkyl, -OH, oxo, -NRkRl, halo, C1-4haloalkyl, -O-C1-4alkyl, or -O-C1-4-haloalkyl; where Rk and Rl are each independently H or C1-4alkyl; R4 is H, halo, -C(O)OH, C1-4alkylNRxRy , or -C(O)NRxRy, or is a cycloalkyl, heterocyclyl, heterocycloalkyl, phenyl or heteroaryl, wherein each cycloalkyl, heterocyclyl, heterocycloalkyl, phenyl or heteroaryl is optionally substituted with one, two, or three Rz substituents; wherein Rx is H or C1-4alkyl and Ry is H, C1-4alkyl, -O-C1-4alkyl, -SO2-Rr, C1-4alkyl-SO2- Rr monocyclic cycloalkyl, -C1-4alkyl(monocyclic cycloalkyl), monocyclic heterocyclyl, or monocyclic heterocycloalkyl, each optionally substituted with one, two, or three Ro substituents; or Rx and Ry taken together with the nitrogen to which they are attached form a monocyclic heterocyclyl, optionally substituted with C1-4alkyl or -OC1-4alkyl; and each Rz substituent is independently C1-4alkyl, halo, -NRpRq, -C(O)NRpRq, -OH, or -OC1-4alkyl, wherein each alkyl is optionally substituted with -NRmRn; wherein Rm and Rn are each independently H, C1-4alkyl, C(O)C1-2alkyl, C(O)C1-2haloalkyl, C(O)C1-2alkenyl, or Rm and Rn taken together with the nitrogen to which they are attached form a monocyclic heterocycloalkyl, optionally substituted with one or two Ro substituents; wherein each Ro substituent is independently C1-4alkyl, -OH, -OC1-4alkyl, halo, cyano, methylsulfonyl, -NRpRq, or -C(O)NRpRq; wherein Rp and Rq are each independently H, C1-4alkyl, C1-4alkylNH2, C1-4alkylNH(C1-4alkyl), or C1-4alkylN(C1-4alkyl)2; wherein each Rr is independently C1-4alkyl or NRpRq; and R5 is H, C1-4alkyl, halo, -OH, or -OC1-4alkyl; or a pharmaceutically acceptable salt or prodrug or prodrug thereof; or (ii) Formula (II)
Figure imgf000006_0001
wherein Rc1 is phenyl or pyridyl, each optionally substituted with one or two substituents selected from C1-4alkyl, -CF3, fluoro, chloro, -OCH3, and -OCF3; and R4a is C1-4alkylNRxRy or C(O)NRxRy; or is phenyl, pyrazolyl, or pyridyl, each optionally substituted with one or two Rz groups; or a pharmaceutically acceptable salt or prodrug thereof; or (iii) Formula (III):
Figure imgf000006_0002
wherein Rc1 is phenyl or pyridyl, each optionally substituted with one or two substituents selected from C1-4alkyl, -CF3, fluoro, chloro, -OCH3, and -OCF3; and R4a is C1-4alkylNRxRy or -C(O)NRxRy; or is phenyl, pyrazolyl, or pyridyl, each optionally substituted with one or two Rz groups; or a pharmaceutically acceptable salt or prodrug thereof; or (iv) Formula (IV):
Figure imgf000007_0001
wherein Rc1 is phenyl or pyridyl, each optionally substituted with one or two substituents selected from C1-4alkyl, -CF3, fluoro, chloro, -OCH3, and -OCF3; and R4a is C1-4alkylNRxRy or -C(O)NRxRy; or is phenyl, pyrazolyl, or pyridyl, each optionally substituted with one or two Rz groups; or a pharmaceutically acceptable salt or prodrug thereof. [0007] Embodiment 1a is the method of embodiment 1,wherein: R1a and R1b taken together with the nitrogen to which they are attached form:
Figure imgf000007_0002
wherein X and Y are independently N or CRa; wherein Ra is H or C1-4alkyl; and Rb is phenyl, monocyclic cycloalkyl, monocyclic heterocyclyl, monocyclic heterocycloalkyl, or monocyclic heteroaryl, each optionally substituted with one, two, or three Rd substituents; or R1a is H or C1-4alkyl; and R1b is a moncyclic heteroaryl optionally substituted with Rc; wherein Rc is C1-4alkyl, phenyl, -C1-4alkyl-phenyl, monocyclic cycloalkyl, -C1-4alkyl-(monocyclic cycloalkyl), monocyclic heterocyclyl, monocyclic heterocycloalkyl, monocyclic heteroaryl, or -C1-4alkyl-(monocyclic heteroaryl), wherein each alkyl, phenyl, cycloalkyl, heterocyclyl, heterocycloalkyl or heteroaryl is optionally substituted with one, two, or three Rd substituents; wherein each Rd substituent is independently C1-4alkyl, C1-4alkenyl, C1-4alkynyl, -O-C1-4alkyl, halo, cyano, nitro, azido, C1-4haloalkyl, -O-C1-4-haloalkyl, -NRgRh, -NRgC(=O)Rh, -NRgC(=O)NRgRh, -NRgC(=O)ORh, =NORg, -NRgS(=O)1-2Rh, -NRgS(=O)1-2NRgRh, =NSO2Rg, -C(=O)Rg, -C(=O)ORg, -OC(=O)ORg, -OC(=O)Rg, -C(=O)NRgRh, -OC(=O)NRgRh, -ORg, -SRg, -S(=O)Rg, -S(=O)2Rg, -OS(=O)1-2Rg, -S(=O)1-2ORg, or -S(=O)1-2NRgRh; wherein Rg and Rh are each independently H or C1-4alkyl; R2 and R3 taken together with the nitrogen to which they are attached form a heterocyclyl, optionally substituted with one, two, three, or four Rj substituents, or further wherein any of the hydrogens bonded to carbon atoms are optionally replaced by deuterium; wherein each Rj substituent is independently C1-4alkyl, -OH, oxo, -NRkRl, halo, C1-4haloalkyl, -O-C1-4alkyl, or -O-C1-4-haloalkyl; where Rk and Rl are each independently H or C1-4alkyl; R4 is halo, -C(O)OH, C1-4alkylNRxRy , or -C(O)NRxRy, or is a cycloalkyl, heterocyclyl, heterocycloalkyl, phenyl or heteroaryl, wherein each cycloalkyl, heterocyclyl, heterocycloalkyl, phenyl or heteroaryl is optionally substituted with one, two, or three Rz substituents; wherein Rx is H or C1-4alkyl and Ry is H, C1-4alkyl, -O-C1-4alkyl, -SO2-Rr, C1-4alkyl-SO2-Rr monocyclic cycloalkyl, -C1-4alkyl(monocyclic cycloalkyl), monocyclic heterocyclyl, or monocyclic heterocycloalkyl, each optionally substituted with one, two, or three Ro substituents; or Rx and Ry taken together with the nitrogen to which they are attached form a monocyclic heterocyclyl, optionally substituted with C1-4alkyl or -OC1-4alkyl; and each Rz substituent is independently C1-4alkyl, halo, -NRpRq, -C(O)NRpRq, -OH, or -OC1-4alkyl, wherein each alkyl is optionally substituted with -NRmRn; wherein Rm and Rn are each independently H, C1-4alkyl, C(O)C1-2alkyl, C(O)C1-2haloalkyl, C(O)C1-2alkenyl, or Rm and Rn taken together with the nitrogen to which they are attached form a monocyclic heterocycloalkyl, optionally substituted with one or two Ro substituents; wherein each Ro substituent is independently C1-4alkyl, -OH, -OC1-4alkyl, halo, cyano, methylsulfonyl, -NRpRq, or -C(O)NRpRq; wherein Rp and Rq are each independently H, C1-4alkyl, C1-4alkylNH2, C1-4alkylNH(C1- 4alkyl), or C1-4alkylN(C1-4alkyl)2; wherein each Rr is independently C1-4alkyl or NRpRq; and R5 is H, C1-4alkyl, halo, -OH, or -OC1-4alkyl; or a pharmaceutically acceptable salt or prodrug or prodrug thereof. [0008] Embodiment 1b is the method of embodiment 1, wherein R1b is a monocyclic heteroaryl. [0009] Embodiment 1c is the method of embodiment 1, wherein R2 and R3 taken together with the nitrogen to which they are attached form a heterocyclyl, optionally substituted with one, two, three, or four Rj substituents, or further wherein any of the hydrogens bonded to carbon atoms are optionally replaced by deuterium.Embodiment 1d is the method of embodiment 1, wherein R4 is halo, -C(O)OH, C1-4alkylNRxRy, or -C(O)NRxRy, or is a cycloalkyl, heterocyclyl, heterocycloalkyl, phenyl or heteroaryl, wherein each cycloalkyl, heterocyclyl, heterocycloalkyl, phenyl or heteroaryl is optionally substituted with one, two, or three Rz substituents. [0010] Embodiment 2 is the method of embodiment 1, wherein a. the alpha-coronavirus is HCoV 229E; b. the beta-coronavirus lineage B is SARS-CoV2; and c. the beta-coronavirus lineage A is HCoV OC43. [0011] Embodiment 3 is the method of embodiment 2, wherein the infection is caused by SARS-CoV-2, and wherein the infection is COVID-19. [0012] Embodiment 4 is the method of any one of embodiments 1 - 3, wherein R1a and R1b are taken together with the nitrogen to which they are attached to form
Figure imgf000009_0001
. [0013] Embodiment 5 is the method of any one of embodiments 1 - 3, wherein R1a and R1b are taken together with the nitrogen to which they are attached to form
Figure imgf000009_0002
. [0014] Embodiment 6 is the method of any one of embodiments 1 - 5, wherein X is N and Y is CRa. [0015] Embodiment 7 is the method of any one of embodiments 1 - 5, wherein X is CRa and Y is N. [0016] Embodiment 8 is the method of any one of embodiments 1 - 5, wherein X is N and Y is N. [0017] Embodiment 9 is the method of any one of embodiments 1 - 8, wherein Ra is H, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, or tert-butyl. [0018] Embodiment 10 is the method of any one of embodiments 1 - 8, wherein Ra is H or methyl. [0019] Embodiment 11 is the method of any one of embodiments 1 - 8, wherein Ra is H. [0020] Embodiment 12 is the method of any one of embodiments 1 - 11, wherein Rb is optionally substituted phenyl. [0021] Embodiment 13 is the method of any one of embodiments 1 - 11, wherein Rb is tolyl. [0022] Embodiment 14 is the method of any one of embodiments 1 - 11, wherein Rb is phenyl. [0023] Embodiment 15 is the method of any one of embodiments 1 - 11, wherein Rb is optionally substituted pyridinyl or pyrimidinyl. [0024] Embodiment 16 is the method of any one of embodiments 1 - 11, wherein Rb is optionally substituted pyridinyl. [0025] Embodiment 17 is the method of any one of embodiments 1 - 11, wherein Rb is substituted with one or two Rd substituents. [0026] Embodiment 18 is the method of any one of embodiments 1 - 11, wherein Rb is methylpryridinyl, phenyl, m-tolyl, chlorophenyl, bromophenyl, methoxyphenyl. [0027] Embodiment 19 is the method of any one of embodiments 1 - 18, wherein R1a is H or C1-4alkyl; and R1b is a 5-membered N-containing heteroaryl optionally substituted with Rc. [0028] Embodiment 20 is the method of any one of embodiments 1 - 18, wherein R1a is H. [0029] Embodiment 21 is the method of any one of embodiments 1 - 18, wherein R1a is C1- 4alkyl. [0030] Embodiment 22 is the method of any one of embodiments 1 - 18, wherein R1a is methyl. [0031] Embodiment 23 is the method of any one of embodiments 1 - 22, wherein R1b is pyrrolyl, pyrazolyl, imidazolyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, oxadiazolyl, thiadiazolyl, pyrazolopyridinyl, or indazolyl, each optionally substituted with Rc. [0032] Embodiment 23a is the method of any one of embodiments 1 – 22, wherein R1b is pyrrolyl, pyrazolyl, imidazolyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, oxadiazolyl, or thiadiazolyl, each optionally substituted with Rc. [0033] Embodiment 24 is the method of any one of embodiments 1 - 22, wherein R1b is pyrazolyl, imidazolyl, oxazolyl, oxadiazolyl or isoxazolyl, each optionally substituted with Rc. [0034] Embodiment 25 is the method of any one of embodiments 1 - 22, wherein R1b is pyrazolyl, optionally substituted with Rc. [0035] Embodiment 26 is the method of any one of embodiments 1 - 22, wherein R1b is
Figure imgf000010_0001
[0036] Embodiment 27 is the method of any one of embodiments 1 - 22, wherein R1b is c
Figure imgf000010_0002
. [0037] Embodiment 28 is the method of any one of embodiments 1 - 27, wherein Rc is optionally substituted C1-4alkyl. [0038] Embodiment 29 is the method of any one of embodiments 1 - 27, wherein Rc is methyl, ethyl, isopropyl, or trifluoromethyl. [0039] Embodiment 30 is the method of any one of embodiments 1 - 27, wherein Rc is optionally substituted phenyl. [0040] Embodiment 31 is the method of any one of embodiments 1 - 27, wherein Rc is phenyl or o-, m-, p-tolyl, fluorophenyl, methoxyphenyl, or trifluoromethoxyphenyl. [0041] Embodiment 32 is the method of any one of embodiments 1 - 27, wherein Rc is phenyl. [0042] Embodiment 33 is the method of any one of embodiments 1 - 27, wherein Rc is optionally substituted monocyclic cycloalkyl. [0043] Embodiment 34 is the method of any one of embodiments 1 - 27, wherein Rc is optionally substituted cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl. [0044] Embodiment 35 is the method of any one of embodiments 1 - 27, wherein Rc is optionally substituted cyclopropyl. [0045] Embodiment 36 is the method of any one of embodiments 1 - 27, wherein Rc is optionally substituted monocyclic heterocycloalkyl. [0046] Embodiment 37 is the method of any one of embodiments 1 - 27, wherein Rc is optionally substituted cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, or cyclohexylmethyl. [0047] Embodiment 38 is the method of any one of embodiments 1 - 27, wherein Rc is optionally substituted monocyclic heterocyclyl. [0048] Embodiment 39 is the method of any one of embodiments 1 - 27, wherein Rc is optionally substituted pyrrolidinyl, tetrahydrofuranyl, piperidinyl, morpholinyl, or piperazinyl. [0049] Embodiment 40 is the method of any one of embodiments 1 - 27, wherein Rc is optionally substituted monocyclic heteroaryl. [0050] Embodiment 41 is the method of any one of embodiments 1 - 27, wherein Rc is optionally substituted pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, furanyl, thiophenyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, or pyridazinyl. [0051] Embodiment 42 is the method of any one of embodiments 1 - 27, wherein Rc is optionally substituted pyrazole, thiophenyl, imidazolyl, pyridinyl, or pyrimidinyl. [0052] Embodiment 43 is the method of any one of embodiments 1 - 27, wherein Rc is optionally substituted pyrazolyl. [0053] Embodiment 44 is the method of any one of embodiments 1 - 27, wherein Rc is optionally substituted pyridinyl. [0054] Embodiment 45 is the method of any one of embodiments 1 - 27, wherein Rc is methylpyridinyl. [0055] Embodiment 46 is the method of any one of embodiments 1 - 27, wherein Rc is optionally substituted -C1-4alkyl-phenyl, -C1-4alkyl-(monocyclic cycloalkyl), monocyclic heterocycloalkyl, or -C1-4alkyl-(monocyclic heteroaryl). [0056] Embodiment 47 is the method of any one of embodiments 1 - 27, wherein Rc is optionally substituted with one or two Rd substituents and each Rd substituent is independently C1-4alkyl, C1-4alkenyl, C1-4alkynyl, -O-C1-4alkyl, halo, cyano, nitro, azido, C1-4haloalkyl, -O-C1-4-haloalkyl, -NRgRh, -NRgC(=O)Rh, -NRgC(=O)NRgRh, -NRgC(=O)ORh, =NORg, -NRgS(=O)1-2Rh, -NRgS(=O)1-2NRgRh, =NSO2Rg, -C(=O)Rg, -C(=O)ORg, -OC(=O)ORg, -OC(=O)Rg, -C(=O)NRgRh, -OC(=O)NRgRh, -ORg, -SRg, -S(=O)Rg, -S(=O)2Rg, -OS(=O)1-2Rg, -S(=O)1-2ORg, or -S(=O)1-2NRgRh. [0057] Embodiment 48 is the method of any one of embodiments 1 - 47, wherein each Rd substituent is independently C1-4alkyl, -O-C1-4alkyl, C1-4haloalkyl, or halo. [0058] Embodiment 49 is the method of any one of embodiments 1 - 47, wherein each Rd substituent is independently methyl, ethyl, isopropyl, -CF3, -OCH3, -OCF3, or fluoro. [0059] Embodiment 50 is the method of any one of embodiments 1 - 49, wherein Rg and Rh are each independently H or methyl. [0060] Embodiment 51 is the method of any one of embodiments 1 - 50, wherein each of R2 and R3 are independently selected from H, pyrrolidinyl, piperidinyl, and piperazinyl, wherein each pyrrolidinyl, piperidinyl, and piperazinyl is optionally substituted with one Rj substituent. [0061] Embodiment 52 is the method of any one of embodiments 1 - 50, wherein R2 and R3 taken together with the nitrogen to which they are attached form pyrrolidinyl, piperidinyl, piperazinyl, morpholino, or thiomorpholino, each optionally substituted with one, two, three, or four Rj substituents.Embodiment 53 is the method of any one of embodiments 1 - 50, wherein R2 and R3 taken together with the nitrogen to which they are attached form morpholino or piperazinyl, optionally substituted with one, two, three, or four Rj substituents.Embodiment 54 is the method of any one of embodiments 1 - 50, wherein R2 and R3 taken together with the nitrogen to which they are attached form 2,2,6,6-tetrafluoro-morpholino, morpholino-2-one, morpholino-3-one, piperazinyl-2-one, piperazinyl-3-one, thiomorpholino-1,1-dioxide. [0062] Embodiment 55 is the method of any one of embodiments 1 - 50, wherein each Rj substituent is independently methyl, oxo, hydroxy, NH2, -OCH3, halo, -CF3, or -OCF3. [0063] Embodiment 56 is the method of any one of embodiments 1 - 50, wherein R2 and R3 taken together with the nitrogen to which they are attached form morpholino in which 1 to 8 hydrogens are replaced with deuterium. [0064] Embodiment 57 is the method of any one of embodiments 1 - 56, wherein Rk and Rl are each independently H or methyl. [0065] Embodiment 58 is the method of any one of embodiments 1 - 57, wherein R4 is H. [0066] Embodiment 59 is the method of any one of embodiments 1 - 57, wherein R4 is chloro. [0067] Embodiment 60 is the method of any one of embodiments 1 - 57, wherein R4 is optionally substituted phenyl. [0068] Embodiment 61 is the method of any one of embodiments 1 - 57, wherein R4 is optionally substituted heteroaryl. [0069] Embodiment 62 is the method of any one of embodiments 1 - 57, wherein R4 is optionally substituted monocyclic heteroaryl. [0070] Embodiment 63 is the method of any one of embodiments 1 - 57, wherein R4 is optionally substituted pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, furanyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, or pyridazinyl. [0071] Embodiment 64 is the method of any one of embodiments 1 - 57, wherein R4 is
Figure imgf000013_0001
or each optionally
Figure imgf000013_0002
Figure imgf000013_0003
substituted with 1 or 2 Rz groups. [0072] Embodiment 65 is the method of any one of embodiments 1 - 57, wherein R4 is optionally substituted pyridinyl or pyrimidinyl. [0073] Embodiment 66 is the method of any one of embodiments 1 - 57, wherein R4 is optionally substituted pyridinyl. [0074] Embodiment 67 is the method of any one of embodiments 1 - 57, wherein R4 is pyridinyl. [0075] Embodiment 68 is the method of any one of embodiments 1 - 57, wherein R4 is optionally substituted pyrazolyl. [0076] Embodiment 69 is the method of any one of embodiments 1 - 57, wherein R4 is optionally substituted with one or two Rz substituents. [0077] Embodiment 70 is the method of any one of embodiments 1 - 57, wherein R4 is pyrazolyl optionally substituted with one or two Rz substituents. [0078] Embodiment 71 is the method of any one of embodiments 1 - 57, wherein R4 is phenyl or pyridyl, each optionally substituted with one or two substituents selected from C1-4alkyl, - CF3, fluoro, chloro, -OCH3, and -OCF3. [0079] Embodiment 72 is the method of any one of embodiments 1 - 57, wherein R4 is heterocyclyl, optionally substituted with one or two Rz substituents. [0080] Embodiment 73 is the method of any one of embodiments 1 - 57, wherein R4 is pyrrolidinyl, piperidinyl, piperazinyl, morpholino, or thiomorpholino, optionally substituted with one or two Rz substituents. [0081] Embodiment 74 is the method of any one of embodiments 1 - 57, wherein R4 is heterocycloalkyl, optionally substituted with one or two Rz substituents. [0082] Embodiment 75 is the method of any one of embodiments 1 - 57, wherein R4 is pyrrolidinylmethyl, piperidinylmethyl, piperazinylmethyl, morpholinomethyl, or thiomorpholinomethyl, optionally substituted with one or two Rz substituents. [0083] Embodiment 76 is the method of any one of embodiments 1 - 57, wherein R4 is 3- methyl-1H-pyrazol-5-yl, 3-methylisothiazol-5-yl, 2-methyl-1H-imidazol-5-yl, 1-methyl-pyrazol- 4-yl, 1-methylpyrazol-3-yl, 1-((1-acetamido)-eth-2-yl)-5-methyl-pyrazol-3-yl, 1-((1- chloromethylamido)-eth-2-yl)-5-methyl-pyrazol-3-yl, 1-((1-acrylamido)-eth-2-yl)-5-methyl- pyrazol-3-yl, thiazol-2-yl, pyrazol-4-yl, pyrazol-1-yl, oxazol-2-yl, 3-(1-N,N-dimethyl-eth-2-yl)- 4-methyl-pyrazol-1-yl, or pyridinyl. [0084] Embodiment 77 is the method of any one of embodiments 1 - 57, wherein R4 is C1- 4alkylNRxRy. [0085] Embodiment 78 is the method of any one of embodiments 1 - 57, wherein R4 is CH2NRxRy. [0086] Embodiment 79 is the method of any one of embodiments 1 - 57, wherein R4 is - C(O)NRxRy. [0087] Embodiment 80 is the method of any one of embodiments 1 - 79, wherein Rx is H. [0088] Embodiment 81 is the method of any one of embodiments 1 - 79, wherein Rx is methyl or ethyl, optionally substituted with one, two, or three Ro substituents. [0089] Embodiment 82 is the method of any one of embodiments 1 - 79, wherein Rx is methyl. [0090] Embodiment 83 is the method of any one of embodiments 1 - 82, wherein Ry is H. [0091] Embodiment 84 is the method of any one of embodiments 1 - 82, wherein Ry is C1- 4alkyl, optionally substituted with one, two, or three Ro substituents. [0092] Embodiment 85 is the method of any one of embodiments 1 - 82, wherein Ry is methyl, ethyl, propyl, or isopropyl, each optionally substituted with one, two, or three Ro substituents. [0093] Embodiment 86 is the method of any one of embodiments 1 - 82, wherein Ry is H, methyl, ethyl, methyoxy, or methoxyethyl. [0094] Embodiment 87 is the method of any one of embodiments 1 - 82, wherein Ry is methyl. [0095] Embodiment 88 is the method of any one of embodiments 1 - 82, wherein Ry is -SO2- Rr or C1-4alkyl-SO2-Rr. [0096] Embodiment 89 is the method of any one of embodiments 1 - 82, wherein Ry is -SO2- Rr, C1-4alkyl-SO2-Rr; and Rr is CH3 or NH2, NHCH3, or N(CH3)2. [0097] Embodiment 90 is the method of any one of embodiments 1 - 82, wherein Ry is -SO2- methyl, C2-4alkyl-SO2-N(CH3)2. [0098] Embodiment 91 is the method of any one of embodiments 1 - 82, wherein Ry is monocyclic cycloalkyl or -C1-2alkyl(monocyclic cycloalkyl), each optionally substituted with one, two, or three Ro substituents. [0099] Embodiment 92 is the method of any one of embodiments 1 - 82, wherein Ry is monocyclic cycloalkyl, optionally substituted with one, two, or three Ro substituents. [0100] Embodiment 93 is the method of any one of embodiments 1 - 82, wherein Ry is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl, each optionally substituted with one, two, or three Ro substituents. [0101] Embodiment 94 is the method of any one of embodiments 1 - 82, wherein Ry is cyclopropyl. [0102] Embodiment 95 is the method of any one of embodiments 1 - 82, wherein Ry is cyclopropyl, cyclobutyl, cyclopentyl, cyclopropylmethyl, 1-cyclopropylethyl, 2- cyclopropylethyl, cyclobutylmethyl, or cyclopentylmethyl. [0103] Embodiment 96 is the method of any one of embodiments 1 - 82, wherein Ry is monocyclic heterocyclyl, optionally substituted with one, two, or three Ro substituents. [0104] Embodiment 97 is the method of any one of embodiments 1 - 82, wherein Ry is optionally substituted azetidinyl, oxetanyl, pyrrolidinyl, piperidinyl, morpholinyl, piperazinyl, azepanyl, azocanyl, tetrahydrofuranyl, or tetrahydropyranyl, optionally substituted with methyl. [0105] Embodiment 98 is the method of any one of embodiments 1 - 82, wherein Ry is monocyclic heterocycloalkyl, optionally substituted with one, two, or three Ro substituents. [0106] Embodiment 99 is the method of any one of embodiments 1 - 82, wherein Ry is optionally substituted azetidinylmethyl, oxetanylmethyl, pyrrolidinylmethyl, piperidinylmethyl, morpholinylmethyl, or piperazinylmethyl, optionally substituted with methyl. [0107] Embodiment 100 is the method of any one of embodiments 1 - 79, wherein one of Rx and Ry is H and the other is -CH3. [0108] Embodiment 101 is the method of any one of embodiments 1 - 79, wherein both of Rx and Ry is H. [0109] Embodiment 102 is the method of any one of embodiments 1 - 79, wherein both of Rx and Ry is -CH3. [0110] Embodiment 103 is the method of any one of embodiments 1 - 79, wherein Rx and Ry taken together with the nitrogen to which they are attached form a monocyclic heterocyclyl, optionally substituted with C1-4alkyl. [0111] Embodiment 104 is the method of any one of embodiments 1 - 79, wherein Rx and Ry are taken together with the nitrogen to which they are attached to form azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, or thiomorpholinyl, each optionally substituted with methyl. [0112] Embodiment 105 is the method of any one of embodiments 1 - 104, wherein each Rz is independently C1-4alkyl, halo, -OH, or -OC1-4alkyl, wherein each alkyl is optionally substituted with -NRmRn. [0113] Embodiment 106 is the method of any one of embodiments 1 - 104, wherein each Rz is independently -CH3, -OH, halo, or -OCH3. [0114] Embodiment 107 is the method of any one of embodiments 1 - 104, wherein Rz is C2- 4alkyl substituted with -NRmRn or OCH3. [0115] Embodiment 108 is the method of any one of embodiments 1 - 104, wherein each Rz substituent is independently -NRpRq, -C(O)NRpRq. [0116] Embodiment 109 is the method of any one of embodiments 1 - 104, wherein each Rz substituent is methyl, ethyl, isopropyl, -CF3, fluoro, chloro, -OCH3, -OCF3, methylamino, ethylamino, propylamino, butylamino, aminomethyl, aminoethyl, aminopropyl, aminobutyl, dimethylamino, dimethylaminomethyl, dimethylaminoethyl, dimethylaminopropyl, dimethylaminobutyl, -C(O)methylamino, -C(O)ethylamino, -C(O)propylamino, - C(O)butylamino, -C(O)dimethylamino, -C(O)dimethylaminomethyl, -C(O)dimethylaminoethyl, -C(O)dimethylaminopropyl, or -C(O)dimethylaminobutyl. [0117] Embodiment 110 is the method of any one of embodiments 1 - 109, wherein Rm and Rn are each independently H, C1-4alkyl, C(O)CH3, C(O)CH2Cl, or C(O)CH2CH2. [0118] Embodiment 111 is the method of any one of embodiments 1 - 109, wherein Rm and Rn are each H. [0119] Embodiment 112 is the method of any one of embodiments 1 - 109, wherein Rm and Rn are each methyl. [0120] Embodiment 113 is the method of any one of embodiments 1 - 109, wherein Rm and Rn taken together with the nitrogen to which they are attached form a monocyclic heterocyclyl, optionally substituted with one or two Ro substituents. [0121] Embodiment 114 is the method of any one of embodiments 1 - 109, wherein Rm and Rn taken together with the nitrogen to which they are attached form pyrrolidinyl, piperidinyl, piperazinyl, morpholino, thiomorpholino, or thiomorpholino-1,1-dioxide, each optionally substituted with one or two Ro substituents. [0122] Embodiment 115 is the method of any one of embodiments 1 - 109, wherein Rm and Rn taken together with the nitrogen to which they are attached form pyrrolidinyl, piperidinyl, piperazinyl, or morpholino, each optionally substituted with methyl. [0123] Embodiment 116 is the method of any one of embodiments 1 - 115, wherein each Ro substituent is C1-4alkyl, or -NRpRq. [0124] Embodiment 117 is the method of any one of embodiments 1 - 116, wherein Rp and Rq are each independently H, methyl, C1-4alkylNH2, C1-4alkylNHCH3, or C1-4alkylN(CH3)2. [0125] Embodiment 118 is the method of any one of embodiments 1 - 116, wherein Rp and Rq are each independently H or methyl. [0126] Embodiment 119 is the method of any one of embodiments 1 - 118, wherein R5 is H, methyl, ethyl, chloro, bromo, fluoro, -OH, or -OCH3. [0127] Embodiment 120 is the method of any one of embodiments 1 - 118, wherein R5 is H. [0128] Embodiment 121 is the method of any one of embodiments 1 - 3, wherein Rc1 is phenyl or pyridyl, each optionally substituted with methyl, -CF3, Cl, Br, or OCH3. [0129] Embodiment 122 is the method of any one of embodiments 1 - 3, wherein Rc1 is phenyl. [0130] Embodiment 123 is the method of any one of embodiments 1 - 3, wherein Rc1 is tolyl. [0131] Embodiment 124 is the method of any one of embodiments 1 - 3, wherein Rc1 is pyridyl optionally substituted with methyl or -CF3. [0132] Embodiment 125 is the method of any one of embodiments 1 - 3, wherein R4a is pyridyl, optionally substituted with one or two Rz groups. [0133] Embodiment 126 is the method of any one of embodiments 1 - 3, wherein R4a is pyridyl. [0134] Embodiment 127 is the method of any one of embodiments 1 - 3, wherein R4a is pyrazolyl optionally substituted with one or two Rz groups. [0135] Embodiment 128 is the method of any one of embodiments 1 - 3, wherein R4a is 3- methyl-1H-pyrazol-5-yl, 3-methylisothiazol-5-yl, 2-methyl-1H-imidazol-5-yl, 1-methyl-pyrazol- 4-yl, 1-methylpyrazol-3-yl, 1-((1-acetamido)-eth-2-yl)-5-methyl-pyrazol-3-yl, 1-((1- chloromethylamido)-eth-2-yl)-5-methyl-pyrazol-3-yl, 1-((1-acrylamido)-eth-2-yl)-5-methyl- pyrazol-3-yl, thiazol-2-yl, pyrazol-4-yl, pyrazol-1-yl, oxazol-2-yl, or 3-(1-N,N-dimethyl-eth-2- yl)-4-methyl-pyrazol-1-yl. [0136] Embodiment 129 is the method of any one of embodiments 1 - 3, wherein R4a is - C(O)NRxRy wherein Rx is H or C1-4alkyl and Ry is H, C1-4alkyl, -O-C1-4alkyl, -SO2-Rr, C1-4alkyl-SO2-Rr monocyclic cycloalkyl, -C1-4alkyl(monocyclic cycloalkyl), monocyclic heterocyclyl, or monocyclic heterocycloalkyl, each optionally substituted with one, two, or three Ro substituents; and Rr and Ro are as defined herein. [0137] Embodiment 130 is the method of any one of embodiments 1 - 3, wherein R4a is - C(O)NRxRy wherein Rx is H or methyl; and Ry is H, methyl, ethyl, butyl, isopropyl, methoxy, - SO2-methyl, C2-4alkyl-SO2-methyl, C2-4alkyl-SO2-N(CH3)2, cyclopropyl, cyclobutyl, cyclopentyl, cyclopropylmethyl, 1-cyclopropylethyl, 2-cyclopropylethyl, cyclobutylmethyl, cyclopentylmethyl, azetidinyl, oxetanyl, pyrrolidinyl, piperidinyl, morpholinyl, piperazinyl, azepanyl, azocanyl, tetrahydrofuranyl, tetrahydropyranyl, substituted azetidinylmethyl, oxetanylmethyl, pyrrolidinylmethyl, piperidinylmethyl, morpholinylmethyl, or piperazinylmethyl, each optionally substituted with one, two, or three methyl, methoxy, fluoro or amino groups. [0138] Embodiment 131 is the method of any one of the preceding embodiments, wherein the compound is selected from a compound of Table 1 or a pharmaceutically acceptable salt thereof. [0139] Embodiment 132 is the method of any one of embodiments 1 to 131, wherein one or more hydrogen atoms attached to carbon atoms of the compound are replaced by deuterium atoms. [0140] Embodiment 133 is the method of any one of the preceding embodiments, wherein the compound and/or the pharmaceutically acceptable salt is in a pharmaceutical composition. [0141] Embodiment 134 is the method of embodiment 133, wherein the pharmaceutical composition further comprises a pharmaceutically acceptable excipient. BRIEF DESCRIPTION OF THE DRAWINGS [0142] Figure 1 shows antiviral effect and cell toxicity data for Compound 101 in Vero-E6 cells. Concentration-dependent antiviral effect (i.e., antiviral activity) is shown as percent cell survival (i.e., percent inhibition) on the left axis (data represented by black squares with a solid line). Concentration-dependent cell toxicity is shown as percent cell survival on the right axis (data represented by white squares with a dotted line). [0143] Figure 2 shows antiviral effects of Compound 97 A549-ACE2 cells. Viral titer is shown as Log10 of plaque forming units per mL (PFU/mL) on the left axis (data represented by white circles). Percent cell viability is shown on the right axis (data represented by black circles). [0144] Figures 3A-B show the effects of Compounds 91 and 121 on human alphacoronavirus strain 229E (HCoV 229E). Human bronchial epithelial (16BHE) cells infected with HCoV 229E were compared to uninfected 16HBE cells. Data are presented as mean percentage viral inhibition (EC50; circles) or cell viability (CC50; squares) ± SEM (n=3). Figure 3A shows data for Compound 91; Figure 3B shows data for Compound 121. [0145] Figures 4A-C show the effects of Compounds 91, 114 and 121 on human betacoronavirus strain OC43 (HCoV OC43). Human lung mucoepidermoid (H292) cells infected HCoV OC43 were compared to uninfected H292 cells. Data are presented as mean percentage viral inhibition (EC50; circles) or cell viability (CC50; squares) ± SEM (n=3). Figure 4A shows data for Compound 91; Figure 4B shows data for Compound 114; Figure 4C shows data for Compound 121. [0146] Figures 5A-C show the activity of Compound 114 (Fig.5A), Compound 163 (Fig.5B), and the remdesivir control (Fig.5C) in a Vero-E6-SARS-CoV-2 cytopathic assay against two SARS-CoV2 strains, Wildtype (WT) and Delta (D). DETAILED DESCRIPTION OF THE INVENTION [0147] The present disclosure provides methods and compositions for the treatment of certain coronavirus infections. In some embodiments, the present disclosure provides methods and compositions for blocking alpha-coronavirus, beta-coronavirus lineage B, and/or beta- coronavirus lineage A into a host cell with compounds that are phosphoinositide kinase inhibitors, in particular FYVE-type finger-containing phosphoinositide kinase (“PIKfyve”) inhibitors. In some embodiments, the present disclosure provides methods and compositions for preventing and/or treating an infection caused by alpha-coronavirus, beta-coronavirus lineage B, or a beta-coronavirus lineage A with compounds that are phosphoinositide kinase inhibitors, in particular PIKfyve inhibitors. [0148] Reference will now be made in detail to certain embodiments of the invention. While the invention will be described in conjunction with the described embodiments, it will be understood that such descriptions are not intended to limit the invention to those embodiments. On the contrary, the invention is intended to cover all alternatives, modifications, and equivalents, which may be included within the invention as defined by the appended claims. [0149] Before describing the present teachings in detail, it is to be understood that the disclosure is not limited to specific compositions or process steps, as such may vary. It should be noted that, as used in this specification and the appended claims, the singular form “a,” “an,” and “the” include plural references unless the context clearly dictates otherwise. Thus, for example, reference to “a surfactant” includes a plurality of surfactants and the like. [0150] Numeric ranges are inclusive of the numbers defining the range. Measured and measurable values are understood to be approximate, taking into account significant digits and the error associated with the measurement. Also, the use of “comprise,” “comprises,” “comprising,” “contain,” “contains,” “containing,” “include,” “includes,” “included,” and “including” are not intended to be limiting. It is to be understood that both the foregoing general description and detailed description are exemplary and explanatory only and are not restrictive of the teachings. [0151] Unless specifically noted in the above specification, embodiments in the specification that recite “comprising” various components are also contemplated as “consisting of” or “consisting essentially of” the recited components; embodiments in the specification that recite “consisting of” various components are also contemplated as “comprising” or “consisting essentially of” the recited components; and embodiments in the specification that recite “consisting essentially of” various components are also contemplated as “consisting of” or “comprising” the recited components (this interchangeability does not apply to the use of these terms in the claims). [0152] The section headings used herein are for organizational purposes only and are not to be construed as limiting the desired subject matter in any way. In the event that any literature incorporated by reference contradicts any term defined in this specification, this specification controls. While the present teachings are described in conjunction with various embodiments, it is not intended that the present teachings be limited to such embodiments. On the contrary, the present teachings encompass various alternatives, modifications, and equivalents, as will be appreciated by those of skill in the art. I. Definitions [0153] Unless otherwise defined herein, scientific and technical terms used herein have the meanings that are commonly understood by those of ordinary skill in the art. In the event of any latent ambiguity, definitions provided herein take precedence over any dictionary or extrinsic definition. Unless otherwise required by context, singular terms shall include pluralities and plural terms shall include the singular. [0154] Unless otherwise stated, the following terms used in the specification and claims are defined for the purposes of this disclosure and have the following meanings. [0155] As used herein, the term “about” refers to a value or composition that is within an acceptable error range for the particular value or composition as determined by one of ordinary skill in the art, which will depend in part on how the value or composition is measured or determined, i.e., the limitations of the measurement system. For example, “about” or “approximately” can mean within one or more than one standard deviation per the practice in the art. Alternatively, “about” or “approximately” can mean a range of up to 10% (i.e., ±10%) or more depending on the limitations of the measurement system. For example, about 5 mg can include any number between 4.5 mg and 5.5 mg. Furthermore, particularly with respect to biological systems or processes, the terms can mean up to an order of magnitude or up to 5-fold of a value. When particular values or compositions are provided in the instant disclosure, unless otherwise stated, the meaning of “about” or “approximately” should be assumed to be within an acceptable error range for that particular value or composition. “Or” is used in the inclusive sense, i.e., equivalent to “and/or,” unless the context requires otherwise. [0156] The term “and/or” used herein is to be taken mean specific disclosure of each of the specified features or components with or without the other. For example, the term “and/or” as used in a phrase such as “A and/or B” herein is intended to include “A and B,” “A or B,” “A” (alone), and “B” (alone). Likewise, the term “and/or” as used in a phrase such as “A, B, and/or C” is intended to encompass each of the following aspects: A, B, and C; A, B, or C; A or C; A or B; B or C; A and C; A and B; B and C; A (alone); B (alone); and C (alone).The terms “or a combination thereof” and “or combinations thereof” as used herein refers to any and all permutations and combinations of the listed terms preceding the term. For example, “A, B, C, or combinations thereof” is intended to include at least one of: A, B, C, AB, AC, BC, or ABC, and if order is important in a particular context, also BA, CA, CB, ACB, CBA, BCA, BAC, or CAB. Continuing with this example, expressly included are combinations that contain repeats of one or more item or term, such as BB, AAA, AAB, BBC, AAABCCCC, CBBAAA, CABABB, and so forth. The skilled artisan will understand that typically there is no limit on the number of items or terms in any combination, unless otherwise apparent from the context. [0157] The term “subject” and “patient” as used herein refers to human and non-human animals, including vertebrates, mammals and non-mammals. In one embodiment, the subject can be human, non-human primates, simian, ape, murine (e.g., mice and rats), bovine, porcine, equine, canine, feline, caprine, lupine, ranine or piscine. [0158] The term “administering”, “administered” and grammatical variants refers to the physical introduction of an agent to a subject, using any of the various methods and delivery systems known to those skilled in the art. Exemplary routes of administration for the formulations disclosed herein include intravenous, intramuscular, subcutaneous, intraperitoneal, spinal or other parenteral routes of administration, for example by injection or infusion. The phrase “parenteral administration” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intralymphatic, intralesional, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal injection and infusion, as well as in vivo electroporation. In one embodiment, the formulation is administered via a non-parenteral route, e.g., orally. Other non-parenteral routes include a topical, epidermal or mucosal route of administration, for example, intranasally, vaginally, rectally, sublingually or topically. Administering can also be performed, for example, once, a plurality of times, and/or over one or more extended periods. [0159] The terms “treat,” “treating,” and “treatment,” as used herein, covers any administration or application of a therapeutic for disease/disorder in a subject, and includes inhibiting the disease/disorder, arresting its development, relieving one or more symptoms of the disease/disorder, or curing the disease/disorder. [0160] The terms “prevent” and “preventing” as used herein, means inhibiting or arresting development of a disease/disorder in a subject deemed to be disease/disorder free. [0161] The terms “block” and “blocking” as used herein with reference to viral entry into a host cell refers to stopping the entry of some or all of a virus, such as a coronavirus, into a host cell or host cells. The blocking may be complete or partial. Partial blocking includes preventing at least some virus from entering host cells, for example, enough virus to prevent the subject from displaying at least one symptom associated with such viral infection. [0162] The terms "effective amount", “therapeutically effective amount” or “effective dose” or related terms may be used interchangeably and refer to an amount of a described PIKfyve inhibitor that when administered to a subject, is sufficient to affect a measurable improvement or prevention of a disease or disorder associated with a virus infection. [0163] A “pharmaceutically acceptable vehicle” for therapeutic purposes is a physical embodiment that can be administered to a subject. Pharmaceutically acceptable vehicles include pills, capsules, caplets, tablets, oral fluids, injection fluids, sprays, aerosols, troches, dietary supplements, creams, lotions, oils, solutions, pastes, powders, steam, Or it may be a liquid, but is not limited to these. An example of a pharmaceutically acceptable vehicle is a buffered isotonic solution such as phosphate buffered saline (PBS). [0164] “Alkyl” means a linear saturated monovalent hydrocarbon radical of one to six carbon atoms or a branched saturated monovalent hydrocarbon radical of three to six carbon atoms, e.g., methyl, ethyl, propyl, 2-propyl, butyl (including all isomeric forms), pentyl (including all isomeric forms), and the like. [0165] “Alkylene” means a linear saturated divalent hydrocarbon radical of one to six carbon atoms or a branched saturated divalent hydrocarbon radical of three to six carbon atoms unless otherwise stated e.g., methylene, ethylene, propylene, 1-methylpropylene, 2-methylpropylene, butylene, pentylene, and the like. [0166] “Alkylsulfonyl” means a –SO2R radical where R is alkyl as defined above, e.g., methylsulfonyl, ethylsulfonyl, and the like. [0167] “Amino” means a -NH2. [0168] “Alkoxy” means a -OR radical where R is alkyl as defined above, e.g., methoxy, ethoxy, propoxy, or 2-propoxy, n-, iso-, or tert-butoxy, and the like. [0169] “Alkoxyalkyl” means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with an alkoxy group, (in one embodiment one or two alkoxy groups), as defined above, e.g., 2-methoxyethyl, 1-, 2-, or 3-methoxypropyl, 2-ethoxyethyl, and the like. [0170] “Alkoxycarbonyl” means a -C(O)OR radical where R is alkyl as defined above, e.g., methoxycarbonyl, ethoxycarbonyl, and the like. [0171] “Acyl” means a -COR radical where R is alkyl, haloalkyl, or cycloalkyl, e.g., acetyl, propionyl, cyclopropylcarbonyl, and the like. When R is alkyl, the radical is also referred to herein as alkylcarbonyl. [0172] “Cycloalkyl” means a cyclic saturated monovalent hydrocarbon radical of three to ten carbon atoms wherein one or two carbon atoms may be replaced by an oxo group, e.g., cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl, and the like. [0173] “Carboxy” means –COOH. [0174] A “coronavirus,” “corona respiratory virus,” and “CoV” are used interchangeably herein to refer to a virus belonging to the family Coronaviridae. Coronaviruses are enveloped, positive-sense RNA viruses of approximately 31 Kb, making these viruses the largest known RNA viruses. Coronaviruses infect a variety of host species, including humans and several other vertebrates. These viruses predominantly cause respiratory and intestinal tract infections and induce a wide range of clinical manifestations. In general, coronaviruses can be classified into low pathogenic CoVs (including human CoVs (hCoVs)) and highly pathogenic CoVs, such as severe acute respiratory syndrome CoV (SARS-CoV) and Middle East respiratory syndrome CoV (MERS-CoV). Low pathogenic hCoV infect upper airways and cause seasonal mild to moderate cold-like respiratory illnesses in healthy individuals. In contrast, the highly pathogenic hCoVs (pathogenic hCoV) infect the lower respiratory tract and cause severe pneumonia, which sometimes leads to fatal acute lung injury (ALI) and acute respiratory distress syndrome (ARDS), resulting in high morbidity and mortality. [0175] “Halo” means fluoro, chloro, bromo, or iodo. [0176] “Haloalkyl” means alkyl radical as defined above, which is substituted with one or one to five halogen atoms (in one embodiment fluorine or chlorine,) including those substituted with different halogens, e.g., -CH2Cl, -CF3, -CHF2, -CH2CF3, -CF2CF3, -CF(CH3)2, and the like. In Cx-y-haloalkyl, “Cx-y” means the number of carbon atoms in the alkyl group ranges from x to y. When the alkyl is substituted with only fluoro, it can be referred to in this disclosure as fluoroalkyl. [0177] “Haloalkoxy” means a –OR radical where R is haloalkyl as defined above e.g., -OCF3, -OCHF2, and the like. When R is haloalkyl where the alkyl is substituted with only fluoro, it can be referred to in this disclosure as fluoroalkoxy. [0178] “Hydroxyalkyl” means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with one or two hydroxy groups, provided that if two hydroxy groups are present they are not both on the same carbon atom. Representative examples include, but are not limited to, hydroxymethyl, 2-hydroxyethyl, 2-hydroxypropyl, 3-hydroxypropyl, 1-(hydroxymethyl)-2-methylpropyl, 2-hydroxybutyl, 3-hydroxybutyl, 4-hydroxybutyl, 2,3-dihydroxypropyl, 1-(hydroxymethyl)-2- hydroxyethyl, 2,3-dihydroxybutyl, 3,4-dihydroxybutyl and 2-(hydroxymethyl)-3-hydroxypropyl. Further examples include, but are not limited to, 2-hydroxyethyl, 2,3-dihydroxypropyl, and 1- (hydroxymethyl)-2-hydroxyethyl. [0179] “Heterocyclyl” means a saturated or unsaturated monovalent monocyclic or bi-cyclic group (fused bi-cyclic or bridged bi-cyclic) of 4 to 10 ring atoms in which one or two ring atoms are heteroatom selected from N, O, and S(O)n, where n is an integer from 0 to 2, the remaining ring atoms being C. Additionally, one or two ring carbon atoms in the heterocyclyl ring can optionally be replaced by a –CO- group. More specifically the term heterocyclyl includes, but is not limited to, oxetanyl, pyrrolidino, piperidino, homopiperidino, 2-oxopyrrolidinyl, 2-oxopiperidinyl, morpholino, piperazino, tetrahydropyranyl, thiomorpholino, hexahydropyrrolo[1,2-a]pyrazin-6(2H)-one-yl, tetrahydro-1H-oxazolo[3,4-a]pyrazin-3(5H)-one- yl, 5,6,7,8-tetrahydro-[1,2,4]triazolo[4,3-a]pyrazine-yl, 3-oxa-8-azabicyclo[3.2.1]octane-yl, and the like. When the heterocyclyl ring is unsaturated it can contain one or two ring double bonds provided that the ring is not aromatic. [0180] Heterocyclylalkyl” and “heterocycloalkyl” mean an –(alkylene)-R radical where R is heterocyclyl ring as defined above e.g., tetraydrofuranylmethyl, piperazinylmethyl, morpholinylethyl, and the like. [0181] “Heterocycloamino” means a saturated or unsaturated monovalent monocyclic group of 4 to 8 ring atoms in which one or two ring atoms are heteroatom selected from N, O, or S(O)n, where n is an integer from 0 to 2, the remaining ring atoms being C provided that at least one of the ring atoms is N. Additionally, one or two ring carbon atoms in the heterocycloamino ring can optionally be replaced by a –CO- group. When the heterocycloamino ring is unsaturated it can contain one or two ring double bonds provided that the ring is not aromatic. [0182] “Heterocycloaminoalkyl” means a –(alkylene)-R radical where R is heterocycloamino as described above. [0183] “Heteroaryl” means a monovalent monocyclic or bicyclic aromatic radical of 5 to 10 ring atoms where one or more, (in one embodiment one, two, or three), ring atoms are heteroatom selected from N, O, and S, the remaining ring atoms being carbon. Representative examples include, but are not limited to, pyrrolyl, thienyl, thiazolyl, imidazolyl, furanyl, indolyl, isoindolyl, oxazolyl, isoxazolyl, benzothiazolyl, benzoxazolyl, quinolinyl, isoquinolinyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, tetrazolyl, pyrazolopyridinyl, indazolyl, furopyrimidinyl, and the like. [0184] “Oxo” means an =(O) group and “carbonyl” means a >C(O) group. [0185] “Optional” or “optionally” means that the subsequently described event or circumstance may but need not occur, and that the description includes instances where the event or circumstance occurs and instances in which it does not. For example, “heterocyclyl group optionally substituted with an alkyl group” means that the alkyl may but need not be present, and the description includes situations where the heterocyclyl group is substituted with an alkyl group and situations where the heterocyclyl group is not substituted with alkyl. [0186] The phrase “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio. [0187] The phrase “pharmaceutically acceptable excipient” or “pharmaceutically acceptable carrier” as used herein means a pharmaceutically acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material. Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient. Some examples of materials which can serve as pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen- free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) phosphate buffer solutions; and (21) other non-toxic compatible substances employed in pharmaceutical formulations. [0188] “PIKfyve inhibitor” refers to a molecule that inhibits phosphatidylinositol 3-phosphate 5-kinase (PIKfyve). [0189] The term “salt” or “pharmaceutically acceptable salt” refers to salts derived from a variety of organic and inorganic counter ions well known in the art. Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids. Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like. Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like. Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases. Inorganic bases from which salts can be derived include, for example, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like. Organic bases from which salts can be derived include, for example, primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like, specifically such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, and ethanolamine. In some embodiments, the pharmaceutically acceptable base addition salt is chosen from ammonium, potassium, sodium, calcium, and magnesium salts. It is understood that the pharmaceutically acceptable salts are non-toxic. Additional information on suitable pharmaceutically acceptable salts can be found in Remington’s Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, PA, 1985, which is incorporated herein by reference. II. Methods of Treating, Uses, Administration, and Pharmaceutical Compositions [0190] Provided herein are methods of treating an infection caused by a virus, inhibiting entry of a virus into a host cell, or inhibiting fusion of viral membrane with a host cell membrane comprising administering to a subject in need thereof a compound of Formula (I) or any of the embodiments thereof (e.g., compounds of Table 1) described herein. [0191] In one aspect, the disclosure relates to a method of blocking alpha-coronavirus entry into a host cell and preventing an infection caused by alpha-coronavirus, comprising administering to a subject in need thereof a compound of Formula (I) or any of the embodiments thereof described herein. In some embodiments, the alpha-coronavirus is HCoV 229E. [0192] In another aspect, the disclosure relates to a method of blocking beta-coronavirus lineage B entry into a host cell and preventing an infection caused by beta-coronavirus lineage B, comprising administering to a subject in need thereof a compound of Formula (I) or any of the embodiments thereof described herein. In some embodiments, the beta-coronavirus lineage B is SARS-CoV2. In some embodiments, the infection caused by SARS-CoV-2 is COVID-19. In some embodiments, the methods treat or prevent COVID-19 infection. [0193] In another aspect, the disclosure relates to a method of blocking beta-coronavirus lineage A entry into a host cell and preventing an infection caused by beta-coronavirus lineage A, comprising administering to a subject in need thereof a compound of Formula (I) or any of the embodiments thereof described herein. In some embodiments, the beta-coronavirus lineage A is HCoV OC43. [0194] In some embodiments, a method of inhibiting coronavirus viral membrane fusion with an early endosomal membrane is provided comprising administering a compound of Formula (I) or any of the embodiments thereof (e.g., compounds of Table 1) described herein. In some embodiments, a method of inhibiting coronavirus viral membrane fusion with a maturing endosomal membrane is provided comprising administering a compound of Formula (I) or any of the embodiments thereof (e.g., compounds of Table 1) described herein. In some embodiments, a method of inhibiting coronavirus viral membrane fusion with a late endosomal membrane is provided comprising administering a compound of Formula (I) or any of the embodiments thereof (e.g., compounds of Table 1) described herein. In some embodiments, a method of inhibiting coronavirus viral membrane fusion with an endo-lysosomal membrane is provided comprising administering a compound of Formula (I) or any of the embodiments thereof (e.g., compounds of Table 1) described herein. In some embodiments, a method of inhibiting coronavirus viral membrane fusion with a lysosomal membrane is provided comprising administering a compound of Formula (I) or any of the embodiments thereof (e.g., compounds of Table 1) described herein. In some embodiments, a method of inhibiting coronavirus viral membrane fusion with an early macropinosomal membrane is provided comprising administering a compound of Formula (I) or any of the embodiments thereof (e.g., compounds of Table 1) described herein. In some embodiments, a method of inhibiting coronavirus viral membrane fusion with a macropinosomal membrane is provided comprising administering a compound of Formula (I) or any of the embodiments thereof (e.g., compounds of Table 1) described herein. In some embodiments, a method of inhibiting coronavirus viral membrane fusion with a late macropinosomal membrane is provided comprising administering a compound of Formula (I) or any of the embodiments thereof (e.g., compounds of Table 1) described herein. In some embodiments, a method of inhibiting coronavirus viral membrane fusion with the endoplasmic reticulum (ER) is provided comprising administering a compound of Formula (I) or any of the embodiments thereof (e.g., compounds of Table 1) described herein. In some embodiments, a method of inhibiting coronavirus viral membrane fusing with the plasma membrane directly is provided comprising administering a compound of Formula (I) or any of the embodiments thereof (e.g., compounds of Table 1) described herein. In each such embodiment, the coronavirus may be an alpha-coronavirus, a beta-coronavirus lineage B, or a beta-coronavirus lineage A. [0195] In general, the compounds of this disclosure will be administered in a therapeutically effective amount by any of the accepted modes of administration for agents that serve similar utilities. [0196] In general, compounds of this disclosure will be administered as pharmaceutical compositions by any one of the following routes: oral, systemic (e.g., transdermal, intranasal or by suppository), or parenteral (e.g., intramuscular, intravenous, or subcutaneous) administration. In some embodiments, the manner of administration is nasal using a convenient daily dosage regimen, which can be adjusted according to the degree of affliction. Compositions can take the form of tablets, pills, capsules, semisolids, powders, sustained release formulations, solutions, suspensions, elixirs, aerosols, or any other appropriate compositions. [0197] Pharmaceutical compositions can be formulated using one or more pharmaceutically acceptable carriers comprising excipients and auxiliaries. The formulation can be modified depending upon the route of administration chosen. The pharmaceutical compositions can also include the compounds described herein in a free base form or a pharmaceutically acceptable salt form. [0198] Methods for formulation of the pharmaceutical compositions can include formulating any of the compounds described herein with one or more inert, pharmaceutically acceptable excipients or carriers to form a solid, semi-solid, or liquid composition. Solid compositions can include, for example, powders, tablets, dispersible granules and capsules, and in some aspects, the solid compositions further contain nontoxic, auxiliary substances, for example wetting or emulsifying agents, pH buffering agents, and other pharmaceutically acceptable additives. Alternatively, the compositions described herein can be lyophilized or in powder form for re- constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use. The active ingredients can be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization (e.g., hydroxymethylcellulose or gelatin microcapsules and poly-(methylmethacylate) microcapsules, respectively), in colloidal drug- delivery systems (e.g., liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions. [0199] The pharmaceutical compositions and formulations can be sterilized. Sterilization can be accomplished by filtration through sterile filtration. [0200] The pharmaceutical compositions described herein can be formulated for administration as an injection. Non-limiting examples of formulations for injection can include a sterile suspension, solution, or emulsion in oily or aqueous vehicles. Suitable oily vehicles can include, but are not limited to, lipophilic solvents or vehicles such as fatty oils, synthetic fatty acid esters, or liposomes. Aqueous injection suspensions can contain substances which increase the viscosity of the suspension. The suspension can also contain suitable stabilizers. Injections can be formulated for bolus injection or continuous infusion. [0201] For parenteral administration, the compounds can be formulated in a unit dosage injectable form (e.g., solution, suspension, emulsion) in association with a pharmaceutically acceptable parenteral vehicle. Such vehicles can be inherently nontoxic, and non-therapeutic. A vehicle can be water, saline, Ringer’s solution, dextrose solution, and 5% human serum albumin. Nonaqueous vehicles such as fixed oils and ethyl oleate can also be used. Liposomes can be used as carriers. The vehicle can contain minor amounts of additives such as substances that enhance isotonicity and chemical stability (e.g., buffers and preservatives). [0202] Sustained-release preparations can also be prepared. Examples of sustained-release matrices can include polyesters, hydrogels (e.g., poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides, copolymers of L-glutamic acid and γ ethyl-L-glutamate, non- degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOTM (i.e., injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-(–)-3-hydroxybutyric acid. [0203] Pharmaceutical formulations of the compositions described herein can be prepared for storage by mixing a compound with a pharmaceutically acceptable carrier, excipient, and/or a stabilizer. This formulation can be a lyophilized formulation or an aqueous solution. Acceptable carriers, excipients, and/or stabilizers can be nontoxic to recipients at the dosages and concentrations used. Acceptable carriers, excipients, and/or stabilizers can include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives, polypeptides; proteins, such as serum albumin or gelatin; hydrophilic polymers; amino acids; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes; and/or non- ionic surfactants or polyethylene glycol. [0204] Compounds of the present disclosure may be used in methods of treating in combination with one or more other combination agents (e.g., one, two, or three other drugs) that are used in the prevention, treatment, control, amelioration, or reduction of risk of the diseases or conditions for which compounds of the present disclosure are useful. In some embodiments, the combination of the drugs together is safer or more effective than either drug alone. In some embodiments the compounds disclosed herein and the one or more combination agents have complementary activities that do not adversely affect each other. Such molecules can be present in combination in amounts that are effective for the purpose intended. Such other drug(s) may be administered, by a route and in an amount commonly used therefore, contemporaneously or sequentially with a compound of the present disclosure. When a compound of the present disclosure is used contemporaneously with one or more other drugs, in some embodiments, the agents are administered together in a single pharmaceutical composition in unit dosage form. Accordingly, the pharmaceutical compositions of the present disclosure also include those that contain one or more other active ingredients, in addition to a compound of the present disclosure. The weight ratio of the compound of the present disclosure to the second active agent may be varied and will depend upon the effective dose of each ingredient. Generally, an effective dose of each will be used. In some embodiments, combination therapy includes therapies in which the compound of the present disclosure and one or more other drugs are administered separately, and in some cases, the two or more agents are administered on different, overlapping schedules. It is also contemplated that when used in combination with one or more other active ingredients, the compounds of the present disclosure and the other active ingredients may be used in lower doses than when each is used singly. [0205] The compounds, pharmaceutical compositions, and methods of the present disclosure can be useful for treating and preventing infection in a subject such as, but not limited to, a mammal, a human, a non-human mammal, a domesticated animal (e.g., laboratory animals, household pets, or livestock), a non-domesticated animal (e.g., wildlife), a dog, a cat, a rodent, a mouse, a hamster, a cow, a bird, a chicken, a fish, a pig, a horse, a goat, a sheep, or a rabbit. In preferred embodiments, compounds, pharmaceutical compositions, and methods of the present disclosure are used for treating a human. [0206] The compounds, pharmaceutical compositions, and methods described herein can be useful as a therapeutic or preventative, for example a treatment or preventative that can be administered to a subject in need thereof. A therapeutic or preventative effect can be obtained in a subject by prevention (complete or partial), reduction, suppression, remission, or eradication of a disease state, including, but not limited to, a symptom thereof. A therapeutic effect in a subject having a disease or condition, or pre-disposed to have or is beginning to have the disease or condition, can be obtained by a reduction, a suppression, a prevention, a remission, or an eradication of the condition or disease, or pre-condition or pre-disease state. [0207] In practicing the methods described herein, therapeutically effective amounts of the compounds or pharmaceutical compositions described herein can be administered to a subject in need thereof, often for treating and/or preventing a condition or progression thereof. A pharmaceutical composition can affect the physiology of the subject, such as the immune system, inflammatory response, or other physiologic affect. A therapeutically effective amount can vary widely depending on the severity of the disease, the age and relative health of the subject, the potency of the compounds used, and other factors. [0208] Treat and/or treating can refer to any indicia of success in the treatment or amelioration of the disease or condition. Treating can include, for example, reducing, delaying or alleviating the severity of one or more symptoms of the disease or condition, or it can include reducing the frequency with which symptoms of a disease, defect, disorder, or adverse condition, and the like, are experienced by a patient. Treat can be used herein to refer to a method that results in some level of treatment or amelioration of the disease or condition and can contemplate a range of results directed to that end, including but not restricted to prevention of the condition entirely. [0209] Prevent, preventing, and the like can refer to the prevention of the disease or condition in the patient. For example, if an individual at risk of contracting a disease is treated with the methods of the present disclosure and does not later contract the disease, then the disease has been prevented, at least over a period of time, in that individual. In some embodiments the PIKfyve inhibitors described herein can prevent coronavirus infection. In some embodiments the PIKfyve inhibitors described herein can treat a coronavirus infection. [0210] A therapeutically effective amount can be the amount of a compound or pharmaceutical composition or an active component thereof sufficient to provide a beneficial effect or to otherwise reduce a detrimental non-beneficial event to the individual to whom the composition is administered. A therapeutically effective dose can be a dose that produces one or more desired or desirable (e.g., beneficial) effects for which it is administered, such administration occurring one or more times over a given period of time. An exact dose can depend on the purpose of the treatment and can be ascertainable by one skilled in the art using known techniques. [0211] The compounds or pharmaceutical compositions described herein that can be used in the methods and uses described herein can be formulated and dosages established in a fashion consistent with good medical practice taking into account the disorder to be treated, the condition of the individual patient, the site of delivery of the compound or pharmaceutical composition, the method of administration and other factors known to practitioners. The compounds or pharmaceutical compositions can be prepared according to the description of preparation described herein. [0212] One of ordinary skill in the art would understand that the amount, duration, and frequency of administration of a pharmaceutical composition or compound described herein to a subject in need thereof depends on several factors including, for example but not limited to, the health of the subject, the specific disease or condition of the patient, the grade or level of a specific disease or condition of the patient, the additional therapeutics the subject is being or has been administered, and the like. [0213] The methods, uses, compounds, and pharmaceutical compositions described herein can be for administration to a subject in need thereof. Often, administration of the compounds or pharmaceutical compositions can include routes of administration, non-limiting examples of administration routes include intravenous, intraarterial, subcutaneous, subdural, intramuscular, intracranial, intrasternal, intratumoral, or intraperitoneally. Additionally, a pharmaceutical composition or compound can be administered to a subject by additional routes of administration, for example, by inhalation, oral, dermal, intranasal, or intrathecal administration. [0214] Pharmaceutical compositions or compounds of the present disclosure can be administered to a subject in need thereof in a first administration, and in one or more additional administrations. The one or more additional administrations can be administered to the subject in need thereof minutes, hours, days, weeks, or months following the first administration. Any one of the additional administrations can be administered to the subject in need thereof less than 21 days, or less than 14 days, less than 10 days, less than 7 days, less than 4 days or less than 1 day after the first administration. The one or more administrations can occur more than once per day, more than once per week, or more than once per month. The compounds or pharmaceutical compositions can be administered to the subject in need thereof in cycles of 21 days, 14 days, 10 days, 7 days, 4 days, or daily over a period of one to seven days. [0215] The compounds, pharmaceutical compositions, and methods provided herein can be useful for the treatment of a plurality of diseases or conditions or preventing a disease or a condition in a subject, or other therapeutic applications for subjects in need thereof. III. Viruses Treatable and Preventable with the Disclosed Compositions [0216] The PIKfyve inhibitors described herein can be administered to treat and/or prevent certain coronavirus infection. [0217] The coronaviruses belong to the order Nidovirales, family Coronaviridae, and the subfamily Coronavirinae. They are genetically categorized into four genera: the Alphacoronavirus, Betacoronavirus, Gammacoronavirus, and Deltacoronavirus. Mahendra et al., Cureus, 12(3):e7423 (2020). The Alphacoronaviruses and the Betacoronaviruses typically infect mammals, whereas the Gammacoronaviruses and the Deltacoronaviruses predominantly infect birds. In some embodiments, the coronavirus is an Alphacoronavirus, Betacoronavirus, Gammacoronavirus, or Deltacoronavirus. [0218] Alphacoronavirus strains that are associated with human disease include HCoV-229E and HCoV-NL63. (Id.) Other examples of alphacoronavirus strains include feline infectious peritonitis virus (FIPV), canine coronavirus (CCoV), porcine respiratory coronavirus (PRCV), porcine epidemic diarrhea virus (PEDV), transmissible gastroenteritis virus (TGEV), Rhinolophus bat coronavirus (Rh-BatCoV) HKU2, Miniopterus bat coronavirus (Mi-BatCoV) HKU8, Mi-BatCoV 1A, Mi-BatCoV 1B, Rousettus bat coronavirus (Ro-BatCoV) HKU10183A, Hipposideros bat coronavirus (Hi-BatCoV) HKU10 LSH5A, and Scotiphilus bat coronavirus (Sc-BatCoV) 512. Lau et al., J. Virol.86(21):11906-11918 (2012); Woo et al., Viruses, 2(8):1804-1820 (2010). In some embodiments, the coronavirus is HCoV-229E or HCoV-NL63. In other embodiments, the coronavirus is FIPV, CCoV, PRCV, PEDV, TGEV, Rh-BatCoV HKU2, Mi-BatCoV HKU8, Mi-BatCoV 1A, Mi-BatCoV 1B, Ro-BatCoV HKU10183A, Hi- BatCoV HKU10 LSH5A, and Sc-BatCoV 512. [0219] Betacoronaviruses are divided into four subgroups: Embecovirus (lineage A), Sarbecovirus (lineage B), Merbecovirus (lineage C), and Nobecovirus (lineage D). Woo et al., Viruses, 2(8):1804-1820 (2010). Examples of Embecovirus (lineage A) include bovine coronavirus (BCoV), human coronavirus OC43 (HCoV-OC43), HCoV-HKU1, porcine hemagglutinating encephalomyelitis virus (PHEV), giraffe coronavirus (GiCoV), and murine hepatitis virus (MHV). Id. Examples of Sarbecovirus (lineage B) include SARS-CoV-1 and SARSr-Rh-BatCoV HKU3. Id. Examples of Merbecovirus (lineage C) include MERS-CoV, Tylonycteris bat coronavirus (Ty-BatCoV) HKU4 and Pipistrellus bat coronavirus (Pi-BatCoV) HKU5. Id. Examples of Nobecovirus (lineage D) include Ro-BatCoV HKU9. Id. Betacoronavirus strains that are associated with human disease include SARS-CoV-1, HCoV- OC43, HCoV-HKU1, and MERS-CoV. [0220] In some embodiments, the coronavirus causes upper respiratory tract disease, lower respiratory tract disease, fever, sore throat, swollen adenoids, colds with minor or major symptoms, malaise, muscle and joint pains, nausea, vomiting, loss of appetite, pneumonia, secondary bacterial infection, bronchitis, dyspnea, diarrhea, shortness of breath, acute respiratory distress syndrome, cytokine storm, multi-organ failure, septic shock, blood clots, loss of smell, or loss of taste. In some embodiments, the coronavirus causes no symptoms at all. [0221] In some embodiments, the disclosed compounds are administered to block an alpha- coronavirus entry into a host cell. In some embodiments, the disclosed compounds are administered to block a beta-coronavirus lineage B entry into a host cell. In some embodiments, the disclosed compounds are administered to block a beta-coronavirus lineage A entry into a host cell. In some embodiments, the disclosed compounds are administered to prevent an alpha- coronavirus infection in a subject. In some embodiments, the disclosed compounds are administered to prevent a beta-coronavirus lineage B infection in a subject. In some embodiments, the disclosed compounds are administered to prevent a beta-coronavirus lineage A infection in a subject. In some embodiments, the disclosed compounds are administered to block an alpha-coronavirus entry into a host cell and prevent an alpha-coronavirus infection in a subject. In some embodiments, the disclosed compounds are administered to block a beta- coronavirus lineage B entry into a host cell prevent a beta-coronavirus lineage B infection in a subject. In some embodiments, the disclosed compounds are administered to block a beta- coronavirus lineage A entry into a host cell prevent a beta-coronavirus lineage A infection in a subject. In some embodiments, the alpha-coronavirus is HCoV 229E. In some embodiments, the beta-coronavirus lineage B is SARS-CoV2. In some embodiments, the beta-coronavirus lineage A is HCoV OC43. In some embodiments, the infection is caused by SARS-CoV-2, and the infection is COVID-19. IV. PIKfyve inhibitor compounds [0222] The compounds described herein may in some cases exist as diastereomers, enantiomers, or other stereoisomeric forms. All chiral, diastereomeric, racemic forms, as individual forms and mixtures thereof, are within the scope of this disclosure, unless the specific stereochemistry or isomeric form is specifically indicated. Compounds of the present disclosure containing an asymmetrically substituted atom may be isolated in optically active, optically enriched, optically pure, or racemic forms. It is well known in the art how to prepare optically active forms, such as by resolution of materials. Separation of stereoisomers may be performed by chromatography or by forming diastereomers and separating by recrystallization, or chromatography, or any combination thereof. (Jean Jacques, Andre Collet, Samuel H. Wilen, “Enantiomers, Racemates and Resolutions”, John Wiley and Sons, Inc., 1981, herein incorporated by reference for this disclosure). Stereoisomers may also be obtained by stereoselective synthesis. [0223] Certain compounds of Formula (I) (or any of the embodiments thereof described herein) and/or a pharmaceutically acceptable salt or prodrug thereof can exist as tautomers and/or geometric isomers. All possible tautomers and cis and trans isomers, as individual forms and mixtures thereof, are within the scope of this disclosure. For example, pyrazole tautomers as shown below are equivalent structures. The depiction of one such structure is intended to encompass both structures.
Figure imgf000035_0001
[0224] Additionally, as used herein the term alkyl includes all the possible isomeric forms of said alkyl group albeit only a few examples are set forth. Furthermore, when the cyclic groups such as heteroaryl, heterocyclyl are substituted, they include all the positional isomers. [0225] Pharmaceutically acceptable salts of the compounds of Formula (I) (or any of the embodiments thereof described herein) are within the scope of this disclosure. In addition, the compounds described herein include hydrates and solvates of the compounds or pharmaceutically acceptable salts thereof. [0226] The present disclosure also includes the prodrugs of compounds of Formula (I) (or any of the embodiments thereof described herein) and/or a pharmaceutically acceptable salt or prodrug thereof. The term prodrug is intended to represent covalently bonded carriers, which are capable of releasing the active ingredient of Formula (I) (or any of the embodiments thereof described herein) when the prodrug is administered to a mammalian subject. Release of the active ingredient occurs in vivo. Prodrugs can be prepared by techniques known to one skilled in the art. These techniques generally modify appropriate functional groups in a given compound. These modified functional groups however regenerate original functional groups in vivo or by routine manipulation. Prodrugs of compounds of Formula (I) (or any of the embodiments thereof described herein) include compounds wherein a hydroxy, amino, carboxylic, or a similar group is modified. Examples of prodrugs include, but are not limited to esters (e.g., acetate, formate, and benzoate derivatives), carbamates (e.g., N,N-dimethylaminocarbonyl) or phosphonates (e.g., -OP(=O)(OH)2 ) of hydroxy or amino functional groups in compounds of Formula (I)), amides (e.g., trifluoroacetylamino, acetylamino, and the like), and the like. Prodrugs of compounds of Formula (I) (or any of the embodiments thereof described herein) and/or a pharmaceutically acceptable salt or prodrug thereof are also within the scope of this disclosure. [0227] The present disclosure also includes polymorphic forms (amorphous as well as crystalline) and deuterated forms of compounds of Formula (I) (or any of the embodiments thereof described herein) and/or a pharmaceutically acceptable salt or prodrug thereof. [0228] The compounds disclosed herein, in some embodiments, are used in different enriched isotopic forms, e.g., enriched in the content of 2H, 3H, 11C, 13C and/or 14C. In one particular embodiment, the compound is deuterated in at least one position. Such deuterated forms can be made by the procedure described in U.S. Patent Nos.5,846,514 and 6,334,997. As described in U.S. Patent Nos.5,846,514 and 6,334,997, deuteration can improve the metabolic stability and or efficacy, thus increasing the duration of action of drugs. [0229] Unless otherwise stated, structures depicted herein are intended to include compounds which differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structures except for the replacement of a hydrogen by a deuterium or tritium, or the replacement of a carbon by 13C- or 14C-enriched carbon are within the scope of the present disclosure. [0230] The compounds of the present disclosure optionally contain unnatural proportions of atomic isotopes at one or more atoms that constitute such compounds. For example, the compounds may be labeled with isotopes, such as for example, deuterium (2H), tritium (3H), iodine-125 (125I) or carbon-14 (14C). Isotopic substitution with 2H, 11C, 13C, 14C, 15C, 12N, 13N, 15N, 16N, 16O, 17O, 14F, 15F, 16F, 17F, 18F, 33S, 34S, 35S, 36S, 35Cl, 37Cl, 79Br, 81Br, and 125I are all contemplated. All isotopic variations of the compounds of the present invention, whether radioactive or not, are encompassed within the scope of the present invention. [0231] In certain embodiments, the compounds disclosed herein have some or all of the 1H atoms replaced with 2H atoms. The methods of synthesis for deuterium-containing compounds are known in the art and include, by way of non-limiting example only, the following synthetic methods. [0232] Deuterium substituted compounds are synthesized using various methods such as described in: Dean, Dennis C.; Editor. Recent Advances in the Synthesis and Applications of Radiolabeled Compounds for Drug Discovery and Development. [In: Curr., Pharm. Des., 2000; 6(10)] 2000, 110 pp; George W.; Varma, Rajender S. The Synthesis of Radiolabeled Compounds via Organometallic Intermediates, Tetrahedron, 1989, 45(21), 6601-21; and Evans, E. Anthony. Synthesis of radiolabeled compounds, J. Radioanal. Chem., 1981, 64(1-2), 9-32. [0233] Deuterated starting materials are readily available and are subjected to the synthetic methods described herein to provide for the synthesis of deuterium-containing compounds. Large numbers of deuterium-containing reagents and building blocks are available commercially from chemical vendors, such as Aldrich Chemical Co. [0234] In one aspect is a compound of Formula (I):
Figure imgf000037_0001
wherein: R1a and R1b taken together with the nitrogen to which they are attached form:
Figure imgf000037_0002
wherein X and Y are independently N or CRa; wherein Ra is H or C1-4alkyl; and Rb is phenyl, monocyclic cycloalkyl, monocyclic heterocyclyl, monocyclic heterocycloalkyl, or monocyclic heteroaryl, each optionally substituted with one, two, or three Rd substituents; or R1a is H or C1-4alkyl; and R1b is a heteroaryl optionally substituted with Rc; wherein Rc is C1-4alkyl, phenyl, -C1-4alkyl-phenyl, monocyclic cycloalkyl, -C1-4alkyl-(monocyclic cycloalkyl), monocyclic heterocyclyl, monocyclic heterocycloalkyl, monocyclic heteroaryl, or -C1-4alkyl-(monocyclic heteroaryl), wherein each alkyl, phenyl, cycloalkyl, heterocyclyl, heterocycloalkyl or heteroaryl is optionally substituted with one, two, or three Rd substituents; wherein each Rd substituent is independently C1-4alkyl, C1-4alkenyl, C1-4alkynyl, -O-C1-4alkyl, halo, cyano, nitro, azido, C1-4haloalkyl, -O-C1-4-haloalkyl, -NRgRh, -NRgC(=O)Rh, -NRgC(=O)NRgRh, -NRgC(=O)ORh, =NORg, -NRgS(=O)1-2Rh, -NRgS(=O)1-2NRgRh, =NSO2Rg, -C(=O)Rg, -C(=O)ORg, -OC(=O)ORg, -OC(=O)Rg, -C(=O)NRgRh, -OC(=O)NRgRh, -ORg, -SRg, -S(=O)Rg, -S(=O)2Rg, -OS(=O)1-2Rg, -S(=O)1-2ORg, or -S(=O)1-2NRgRh; wherein Rg and Rh are each independently H or C1-4alkyl; each of R2 and R3 is independently chosen from H, C1-4alkyl, cycloalkyl, C1-4alkylcycloalkyl, heterocyclyl, heterocycloalkyl, and heteroaryl, each optionally substituted with one, two, or three Rj substituents; or R2 and R3 taken together with the nitrogen to which they are attached form a heterocyclyl, optionally substituted with one, two, three, or four Rj substituents, or further wherein any of the hydrogens bonded to carbon atoms are optionally replaced by deuterium; wherein each Rj substituent is independently C1-4alkyl, -OH, -NRkRl, halo, C1-4haloalkyl, -O- C1-4alkyl, or -O-C1-4-haloalkyl; where Rk and Rl are each independently H or C1-4alkyl; R4 is H, halo, -C(O)OH, C1-4alkylNRxRy , or -C(O)NRxRy, or is a cycloalkyl, heterocyclyl, heterocycloalkyl, phenyl or heteroaryl, wherein each cycloalkyl, heterocyclyl, heterocycloalkyl, phenyl or heteroaryl is optionally substituted with one, two, or three Rz substituents; wherein Rx is H or C1-4alkyl and Ry is H, C1-4alkyl, -O-C1-4alkyl, -SO2-Rr, C1-4alkyl-SO2-Rr monocyclic cycloalkyl, -C1-4alkyl(monocyclic cycloalkyl), monocyclic heterocyclyl, or monocyclic heterocycloalkyl, each optionally substituted with one, two, or three Ro substituents; or Rx and Ry taken together with the nitrogen to which they are attached form a monocyclic heterocyclyl, optionally substituted with C1-4alkyl or -OC1-4alkyl; and each Rz substituent is independently C1-4alkyl, halo, -NRpRq, -C(O)NRpRq, -OH, or -OC1- 4alkyl, wherein each alkyl is optionally substituted with -NRmRn; wherein Rm and Rn are each independently H, C1-4alkyl, C(O)C1-2alkyl, C(O)C1-2haloalkyl, C(O)C1-2alkenyl, or Rm and Rn taken together with the nitrogen to which they are attached form a monocyclic heterocycloalkyl, optionally substituted with one or two Ro substituents; wherein each Ro substituent is independently C1-4alkyl, -OH, -OC1-4alkyl, halo, cyano, methylsulfonyl, -NRpRq, or -C(O)NRpRq; wherein Rp and Rq are each independently H, C1-4alkyl, C1-4alkylNH2, C1-4alkylNH(C1- 4alkyl), or C1-4alkylN(C1-4alkyl)2; wherein each Rr substituent is independently C1-4alkyl or NRpRq; and R5 is H, C1-4alkyl, halo, -OH, or -OC1-4alkyl; or a pharmaceutically acceptable salt or prodrug thereof. [0235] In some embodiments, R1a and R1b taken together with the nitrogen to which they are attached form:
Figure imgf000039_0001
wherein X and Y are independently N or CRa; wherein Ra is H or C1-4alkyl; and Rb is phenyl, monocyclic cycloalkyl, monocyclic heterocyclyl, monocyclic heterocycloalkyl, or monocyclic heteroaryl, each optionally substituted with one, two, or three Rd substituents; or R1a is H or C1-4alkyl; and R1b is a moncyclic heteroaryl optionally substituted with Rc; wherein Rc is C1-4alkyl, phenyl, -C1-4alkyl-phenyl, monocyclic cycloalkyl, -C1-4alkyl-(monocyclic cycloalkyl), monocyclic heterocyclyl, monocyclic heterocycloalkyl, monocyclic heteroaryl, or -C1-4alkyl-(monocyclic heteroaryl), wherein each alkyl, phenyl, cycloalkyl, heterocyclyl, heterocycloalkyl or heteroaryl is optionally substituted with one, two, or three Rd substituents; wherein each Rd substituent is independently C1-4alkyl, C1-4alkenyl, C1-4alkynyl, -O-C1-4alkyl, halo, cyano, nitro, azido, C1-4haloalkyl, -O-C1-4-haloalkyl, -NRgRh, -NRgC(=O)Rh, -NRgC(=O)NRgRh, -NRgC(=O)ORh, =NORg, -NRgS(=O)1-2Rh, -NRgS(=O)1-2NRgRh, =NSO2Rg, -C(=O)Rg, -C(=O)ORg, -OC(=O)ORg, -OC(=O)Rg, -C(=O)NRgRh, -OC(=O)NRgRh, -ORg, -SRg, -S(=O)Rg, -S(=O)2Rg, -OS(=O)1-2Rg, -S(=O)1-2ORg, or -S(=O)1-2NRgRh; wherein Rg and Rh are each independently H or C1-4alkyl; R2 and R3 taken together with the nitrogen to which they are attached form a heterocyclyl, optionally substituted with one, two, three, or four Rj substituents, or further wherein any of the hydrogens bonded to carbon atoms are optionally replaced by deuterium; wherein each Rj substituent is independently C1-4alkyl, -OH, oxo, -NRkRl, halo, C1-4haloalkyl, -O-C1-4alkyl, or -O-C1-4-haloalkyl; where Rk and Rl are each independently H or C1-4alkyl; R4 is halo, -C(O)OH, C1-4alkylNRxRy, or -C(O)NRxRy, or is a cycloalkyl, heterocyclyl, heterocycloalkyl, phenyl or heteroaryl, wherein each cycloalkyl, heterocyclyl, heterocycloalkyl, phenyl or heteroaryl is optionally substituted with one, two, or three Rz substituents; wherein Rx is H or C1-4alkyl and Ry is H, C1-4alkyl, -O-C1-4alkyl, -SO2-Rr, C1-4alkyl-SO2- Rr monocyclic cycloalkyl, -C1-4alkyl(monocyclic cycloalkyl), monocyclic heterocyclyl, or monocyclic heterocycloalkyl, each optionally substituted with one, two, or three Ro substituents; or Rx and Ry taken together with the nitrogen to which they are attached form a monocyclic heterocyclyl, optionally substituted with C1-4alkyl or -OC1-4alkyl; and each Rz substituent is independently C1-4alkyl, halo, -NRpRq, -C(O)NRpRq, -OH, or -OC1-4alkyl, wherein each alkyl is optionally substituted with -NRmRn; wherein Rm and Rn are each independently H, C1-4alkyl, C(O)C1-2alkyl, C(O)C1-2haloalkyl, C(O)C1-2alkenyl, or Rm and Rn taken together with the nitrogen to which they are attached form a monocyclic heterocycloalkyl, optionally substituted with one or two Ro substituents; wherein each Ro substituent is independently C1-4alkyl, -OH, -OC1-4alkyl, halo, cyano, methylsulfonyl, -NRpRq, or -C(O)NRpRq; wherein Rp and Rq are each independently H, C1-4alkyl, C1-4alkylNH2, C1-4alkylNH(C1- 4alkyl), or C1-4alkylN(C1-4alkyl)2; wherein each Rr is independently C1-4alkyl or NRpRq; and R5 is H, C1-4alkyl, halo, -OH, or -OC1-4alkyl; or a pharmaceutically acceptable salt or prodrug or prodrug thereof. [0236] In some embodiments, R1a and R1b are taken together with the nitrogen to which they are attached to form . In some embodiments, R1a and R1b are taken together with
Figure imgf000040_0002
the nitrogen to which they are attached to form In some embodiments, X is N and
Figure imgf000040_0001
Y is CRa. In some embodiments, X is CRa and Y is N. In some embodiments, X is N and Y is N. In some embodiments, Ra is H, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, or tert-butyl. In some embodiments, Ra is H or methyl. In some embodiments, Ra is H. [0237] In some embodiments, Rb is optionally substituted phenyl. In some embodiments, Rb is optionally substituted monocyclic heteroaryl. In some embodiments, Rb is optionally substituted pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, furanyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, or pyridazinyl. In some embodiments, Rb is optionally substituted pyridinyl or pyrimidinyl. In some embodiments, Rb is optionally substituted pyridinyl. In some embodiments, Rb is phenyl. In some embodiments, Rb is o-, m-, or p-tolyl. In some embodiments, Rb is optionally substituted with one or two Rd substituents. In some embodiments, Rb is optionally substituted with one Rd substituent. In some embodiments, Rb is methylpryridinyl, phenyl, tolyl, chlorophenyl, bromophenyl, or methoxyphenyl. [0238] In some embodiments, R1a is H or C1-4alkyl; and R1b is a 5-membered N-containing heteroaryl optionally substituted with Rc. In some embodiments, R1a is H. In some embodiments, R1a is C1-4alkyl. In some embodiments, R1a is methyl. [0239] In some embodiments, R1b is pyrrolyl, pyrazolyl, imidazolyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, oxadiazolyl, thiadiazolyl, pyrazolopyridinyl, or indazolyl, each optionally substituted with Rc. In some embodiments, R1b is a monocyclic heteroaryl. In some embodiments, R1b is pyrrolyl, pyrazolyl, imidazolyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, oxadiazolyl, or thiadiazolyl, each optionally substituted with Rc. In some embodiments, R1b is pyrazolyl, imidazolyl, oxazolyl, oxadiazolyl or isoxazolyl, each optionally substituted with Rc. In some embodiments, R1b is pyrazolyl, optionally substituted with Rc. In some embodiments, R1b is
Figure imgf000041_0002
[0240] In some embodiments, R 1b is [0241] In some embodiments, Rc
Figure imgf000041_0001
is optionally substituted C1-4alkyl. In some embodiments, Rc is methyl, ethyl, isopropyl, or trifluoromethyl. In some embodiments, Rc is optionally substituted phenyl. In some embodiments, Rc is phenyl or o-, m-, p-tolyl, fluorophenyl, methoxyphenyl, or trifluoromethoxyphenyl. In some embodiments, Rc is phenyl. In some embodiments, Rc is optionally substituted monocyclic cycloalkyl. In some embodiments, Rc is optionally substituted cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl. In some embodiments, Rc is optionally substituted cyclopropyl. In some embodiments, Rc is optionally substituted monocyclic heterocycloalkyl. In some embodiments, Rc is optionally substituted cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, or cyclohexylmethyl. In some embodiments, Rc is optionally substituted monocyclic heterocyclyl. In some embodiments, Rc is optionally substituted pyrrolidinyl, tetrahydrofuranyl, piperidinyl, morpholinyl, or piperazinyl. In some embodiments, Rc is optionally substituted monocyclic heteroaryl. In some embodiments, Rc is optionally substituted pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, furanyl, thiophenyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, or pyridazinyl. In some embodiments, Rc is optionally substituted pyrazole, thiophenyl, imidazolyl, pyridinyl, or pyrimidinyl. In some embodiments, Rc is optionally substituted pyrazolyl. In some embodiments, Rc is optionally substituted pyridinyl. In some embodiments, Rc is methylpyridinyl. In some embodiments, Rc is optionally substituted -C1-4alkyl-phenyl, -C1- 4alkyl-(monocyclic cycloalkyl), monocyclic heterocycloalkyl, or -C1-4alkyl-(monocyclic heteroaryl). In some embodiments, Rc is optionally substituted benzyl, -CH2-(monocyclic cycloalkyl), -CH2-(monocyclic heterocycloalkyl), or -CH2-(monocyclic heteroaryl). In some embodiments, Rc is optionally substituted benzyl or -CH2-(monocyclic cycloalkyl), such as -CH2-cyclopropyl. In some embodiments, each Rc is optionally substituted with one or two Rd substituents. [0242] In some embodiments, each Rd substituent is independently C1-4alkyl, C1-4alkenyl, C1-4alkynyl, -O-C1-4alkyl, halo, cyano, nitro, azido, C1-4haloalkyl, -O-C1-4-haloalkyl, -NRgRh, -NRgC(=O)Rh, -NRgC(=O)NRgRh, -NRgC(=O)ORh, =NORg, -NRgS(=O)1-2Rh, -NRgS(=O)1-2NRgRh, =NSO2Rg, -C(=O)Rg, -C(=O)ORg, -OC(=O)ORg, -OC(=O)Rg, -C(=O)NRgRh, -OC(=O)NRgRh, -ORg, -SRg, -S(=O)Rg, -S(=O)2Rg, -OS(=O)1-2Rg, -S(=O)1-2ORg, or -S(=O)1-2NRgRh. In some embodiments, each Rd substituent is independently C1-4alkyl, -O-C1-4alkyl, C1-4haloalkyl, or halo. In some embodiments, each Rd substituent is independently methyl, ethyl, isopropyl, -CF3, -OCH3, -OCF3, or fluoro. [0243] In some embodiments, Rg and Rh are each independently H or methyl. [0244] In some embodiments, R2 and R3 taken together with the nitrogen to which they are attached form a heterocyclyl, optionally substituted with one, two, three, or four Rj substituents, or further wherein any of the hydrogens bonded to carbon atoms are optionally replaced by deuterium. [0245] In some embodiments, each of R2 and R3 are independently selected from H, pyrrolidinyl, piperidinyl, and piperazinyl, wherein each pyrrolidinyl, piperidinyl, and piperazinyl is optionally substituted with one Rj substituent. [0246] In some embodiments, R2 and R3 taken together with the nitrogen to which they are attached form pyrrolidinyl, piperidinyl, piperazinyl, morpholino, thiomorpholino, or thiomorpholino-1,1-dioxide, each optionally substituted with one, two, three, or four Rj substituents. In some embodiments, R2 and R3 taken together with the nitrogen to which they are attached form morpholino or piperazinyl, optionally substituted with one, two, three, or four Rj substituents. In some embodiments, R2 and R3 taken together with the nitrogen to which they are attached form 2,2,6,6-tetrafluoro-morpholino, morpholino-2-one, morpholino-3-one, piperazinyl-2-one, piperazinyl-3-one, thiomorpholino-1,1-dioxide. [0247] In some embodiments, each Rj substituent is independently methyl, oxo, hydroxy, -OCH3, NH2, halo, -CF3, or -OCF3. [0248] In some embodiments, R2 and R3 taken together with the nitrogen to which they are attached form morpholino in which 1 to 8 hydrogens are replaced with deuterium. [0249] In some embodiments, Rk and Rl are each independently H or methyl. [0250] In some embodiments, R4 is H. In some embodiments, R4 is chloro. [0251] In some embodiments, R4 is halo, -C(O)OH, C1-4alkylNRxRy , or -C(O)NRxRy, or is a cycloalkyl, heterocyclyl, heterocycloalkyl, phenyl or heteroaryl, wherein each cycloalkyl, heterocyclyl, heterocycloalkyl, phenyl or heteroaryl is optionally substituted with one, two, or three Rz substituents. [0252] In some embodiments, R4 is optionally substituted phenyl. In some embodiments, R4 is optionally substituted heteroaryl. In some embodiments, R4 is optionally substituted monocyclic heteroaryl. In some embodiments, R4 is optionally substituted pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, furanyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, or pyridazinyl. In some embodiments, R4 is optionally substituted pyridinyl or pyrimidinyl. [0253] In some embodiments, R4 is
Figure imgf000043_0001
each optionally substituted with 1 or 2 Rz groups.
Figure imgf000043_0002
[0254] In some embodiments, R4 is optionally substituted pyridinyl. In some embodiments, R4 is pyridinyl. In some embodiments, R4 is 4-pyridyl, 3-pyridyl, or 2-pyridyl. In some embodiments, R4 is 4-pyridyl. In some embodiments, R4 is optionally substituted with one or two Rz substituents. In some embodiments, R4 is phenyl or pyridyl, each optionally substituted with one or two substituents selected from C1-4alkyl, -CF3, fluoro, chloro, -OCH3, and -OCF3. [0255] In some embodiments, R4 is heterocyclyl, optionally substituted with one or two Rz substituents. In some embodiments, R4 is pyrrolidinyl, piperidinyl, piperazinyl, morpholino, or thiomorpholino, optionally substituted with one or two Rz substituents. In some embodiments, R4 is pyrrolidinyl, or piperazinyl, optionally substituted with one C1-4alkyl. In some embodiments, R4 is optionally substituted pyrazolyl. In some embodiments, R4 is optionally substituted with one or two Rz substituents. In some embodiments, R4 is optionally substituted with one Rz substituent. In some embodiments, R4 is 3-methyl-1H-pyrazol-5-yl, 3- methylisothiazol-5-yl, 2-methyl-1H-imidazol-5-yl, 1-methyl-pyrazol-4-yl, 1-methylpyrazol-3-yl, 1-((1-acetamido)-eth-2-yl)-5-methyl-pyrazol-3-yl, 1-((1-chloromethylamido)-eth-2-yl)-5- methyl-pyrazol-3-yl, 1-((1-acrylamido)-eth-2-yl)-5-methyl-pyrazol-3-yl, thiazol-2-yl, pyrazol-4- yl, pyrazol-1-yl, oxazol-2-yl, or 3-(1-N,N-dimethyl-eth-2-yl)-4-methyl-pyrazol-1-yl. [0256] In some embodiments, R4 is heterocycloalkyl, optionally substituted with one or two Rz substituents. In some embodiments, R4 is pyrrolidinylmethyl, piperidinylmethyl, piperazinylmethyl, morpholinomethyl, or thiomorpholinomethyl, optionally substituted with one or two Rz substituents. [0257] In some embodiments, R4 is C1-4alkylNRxRy. In some embodiments, R4 is CH2NRxRy. In some embodiments, R4 is -C(O)NRxRy. [0258] In some embodiments, Rx is H. In some embodiments, Rx is methyl or ethyl, optionally substituted with one, two, or three Ro substituents. In some embodiments, Rx is methyl. [0259] In some embodiments, Ry is H, methyl, ethyl, methyoxy, or methoxyethyl. In some embodiments, Ry is H. In some embodiments, Ry is C1-4alkyl, optionally substituted with one, two, or three Ro substituents. In some embodiments, Ry is methyl, ethyl, propyl, or isopropyl, each optionally substituted with one, two, or three Ro substituents. In some embodiments, Ry is methyl, ethyl, or methoxyethyl. In some embodiments, Ry is methoxy. In some embodiments, Ry is -SO2-Rr or C1-4alkyl-SO2-Rr. Ry is -SO2-Rr, C1-4alkyl-SO2-Rr; and Rr is CH3 or NH2, NHCH3, or N(CH3)2. In some embodiments, Ry is -SO2-methyl, C2-4alkyl-SO2-N(CH3)2. In some embodiments, Ry is -SO2-methyl. In some embodiments, Ry is monocyclic cycloalkyl or -C1-2alkyl(monocyclic cycloalkyl), each optionally substituted with one, two, or three Ro substituents. In some embodiments, Ry is monocyclic cycloalkyl, optionally substituted with one, two, or three Ro substituents. In some embodiments, Ry is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl, each optionally substituted with one, two, or three Ro substituents. In some embodiments, Ry is cyclopropyl. In some embodiments, Ry is cyclopropyl, cyclobutyl, cyclopentyl, cyclopropylmethyl, 1-cyclopropylethyl, 2-cyclopropylethyl, cyclobutylmethyl, or cyclopentylmethyl. In some embodiments, Ry is monocyclic heterocycloalkyl, optionally substituted with one, two, or three Ro substituents. In some embodiments, Ry is optionally substituted azetidinyl, oxetanyl, pyrrolidinyl, piperidinyl, morpholinyl, piperazinyl, azepanyl, azocanyl, tetrahydrofuranyl, or tetrahydropyranyl, optionally substituted with methyl. In some embodiments, Ry is optionally substituted azetidinyl, pyrrolidinyl, piperidinyl, morpholinyl, or piperazinyl. In some embodiments, Ry is monocyclic heterocycloalkyl, optionally substituted with one, two, or three Ro substituents. In some embodiments, Ry is optionally substituted azetidinylmethyl, oxetanylmethyl, pyrrolidinylmethyl, piperidinylmethyl, morpholinylmethyl, or piperazinylmethyl, optionally substituted with methyl. [0260] In some embodiments, Rx and Ry is H and the other is -CH3. In some embodiments, both of Rx and Ry is H. In some embodiments, both of Rx and Ry is -CH3. [0261] In some embodiments, Rx and Ry taken together with the nitrogen to which they are attached form a monocyclic heterocyclyl, optionally substituted with C1-4alkyl. In some embodiments, Rx and Ry are taken together with the nitrogen to which they are attached to form azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, or thiomorpholino, each optionally substituted with methyl. [0262] In some embodiments, each Rz is independently C1-4alkyl, halo, -OH, or -OC1-4alkyl, wherein each alkyl is optionally substituted with -NRmRn. In some embodiments, each Rz is independently -CH3, -OH, halo, or -OCH3. In some embodiments, Rz is C2-3alkyl substituted with -NRmRn. In some embodiments, Rz is C2-4alkyl substituted with -NRmRn or OCH3. In some embodiments, each Rz substituent is independently -NRpRq, -C(O)NRpRq. [0263] In some embodiments, each Rz substituent is methyl, ethyl, isopropyl, -CF3, fluoro, chloro, -OCH3, -OCF3, methylamino, ethylamino, propylamino, butylamino, aminomethyl, aminoethyl, aminopropyl, aminobutyl, dimethylamino, dimethylaminomethyl, dimethylaminoethyl, dimethylaminopropyl, dimethylaminobutyl, -C(O)methylamino, -C(O)ethylamino, -C(O)propylamino, -C(O)butylamino, -C(O)dimethylamino, -C(O)dimethylaminomethyl, -C(O)dimethylaminoethyl, -C(O)dimethylaminopropyl, or -C(O)dimethylaminobutyl. [0264] In some embodiments, Rm and Rn are each independently H, C1-4alkyl, C(O)CH3, C(O)CH2Cl, or C(O)CH2CH2. In some embodiments, Rm and Rn are each H. In some embodiments, Rm and Rn are each methyl. In some embodiments, Rm and Rn taken together with the nitrogen to which they are attached form a monocyclic heterocyclyl, optionally substituted with one or two Ro substituents. In some embodiments, Rm and Rn taken together with the nitrogen to which they are attached form pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholino, or thiomorpholino-1,1-dioxide, each optionally substituted with one or two Ro substituents. In some embodiments, Rm and Rn taken together with the nitrogen to which they are attached form pyrrolidinyl, piperidinyl, piperazinyl, or morpholino, each optionally substituted with one or two Ro substituents. In some embodiments, Rm and Rn taken together with the nitrogen to which they are attached form pyrrolidinyl, piperidinyl, piperazinyl, or morpholinyl, each optionally substituted with methyl. [0265] In some embodiments, each Ro substituent is C1-4alkyl. In some embodiments, each Ro substituent is -NRpRq. In some embodiments, Rp and Rq are each independently H or methyl. [0266] In some embodiments, Rp and Rq are each independently H, methyl, C1-4alkylNH2, C1-4alkylNHCH3, or C1-4alkylN(CH3)2. [0267] In some embodiments, R5 is H, methyl, ethyl, chloro, bromo, fluoro, -OH, or -OCH3. In some embodiments, R5 is H. [0268] In some embodiments, the compound of Formula (I) or the pharmaceutically acceptable salt thereof is a compound of Formula (II):
Figure imgf000046_0001
wherein Rc1 is phenyl or pyridyl, each optionally substituted with one or two substituents selected from C1-4alkyl, -CF3, fluoro, chloro, -OCH3, and -OCF3; and R4a is C1-4alkylNRxRy or C(O)NRxRy wherein Rx and Ry are as defined herein; or phenyl, pyrazolyl, or pyridyl, each optionally substituted with one or two Rz groups; or a pharmaceutically acceptable salt or prodrug thereof. [0269] In some embodiments, the compound of Formula (I) or the pharmaceutically acceptable salt thereof is a compound of Formula (III):
Figure imgf000046_0002
wherein Rc1 is phenyl or pyridyl, each optionally substituted with one or two substituents selected from C1-4alkyl, -CF3, fluoro, chloro, -OCH3, and -OCF3; and R4a is C1-4alkylNRxRy or -C(O)NRxRy wherein Rx and Ry are as defined herein; or phenyl, pyrazolyl, or pyridyl, each optionally substituted with one or two Rz groups; or a pharmaceutically acceptable salt or prodrug thereof. [0270] In some embodiments, the compound of Formula (I) or the pharmaceutically acceptable salt thereof is a compound of Formula (IV):
Figure imgf000047_0001
wherein Rc1 is phenyl or pyridyl, each optionally substituted with one or two substituents selected from C1-4alkyl, -CF3, fluoro, chloro, -OCH3, and -OCF3; and R4a is C1-4alkylNRxRy or -C(O)NRxRy wherein Rx and Ry are as defined herein; or phenyl, pyrazolyl, or pyridyl, each optionally substituted with one or two Rz groups; or a pharmaceutically acceptable salt or prodrug thereof. [0271] In some embodiments, Rc1 is phenyl or pyridyl, each optionally substituted with methyl, -CF3, Cl, Br, or OCH3. In some embodiments, Rc1 is phenyl or m-tolyl. In some embodiments, Rc1 is pyridyl. In some embodiments, Rc1 is 4-pyridyl. [0272] In some embodiments, R4a is phenyl or pyridyl, each optionally substituted with methyl or -CF3. In some embodiments, R4a is phenyl. In some embodiments, R4a is tolyl. In some embodiments, R4a is m-tolyl. In some embodiments, R4a is pyridyl. In some embodiments, R4a is 4-pyridyl. [0273] In some embodiments, R4a is pyrazolyl optionally substituted with one or two Rz groups. [0274] In some embodiments, each Rz is independently methyl, ethyl, isopropyl, -CF3, fluoro, chloro, -OCH3, -OCF3, methylamino, ethylamino, propylamino, butylamino, aminomethyl, aminoethyl, aminopropyl, aminobutyl, dimethylamino, dimethylaminomethyl, dimethylaminoethyl, dimethylaminopropyl, dimethylaminobutyl, -C(O)methylamino, -C(O)ethylamino, -C(O)propylamino, -C(O)butylamino, -C(O)dimethylamino, -C(O)dimethylaminomethyl, -C(O)dimethylaminoethyl, -C(O)dimethylaminopropyl, or -C(O)dimethylaminobutyl. [0275] In some embodiments, R4a is 3-methyl-1H-pyrazol-5-yl, 3-methylisothiazol-5-yl, 2- methyl-1H-imidazol-5-yl, 1-methyl-pyrazol-4-yl, 1-methylpyrazol-3-yl, 1-((1-acetamido)-eth-2- yl)-5-methyl-pyrazol-3-yl, 1-((1-chloromethylamido)-eth-2-yl)-5-methyl-pyrazol-3-yl, 1-((1- acrylamido)-eth-2-yl)-5-methyl-pyrazol-3-yl, thiazol-2-yl, pyrazol-4-yl, pyrazol-1-yl, oxazol-2- yl, or 3-(1-N,N-dimethyl-eth-2-yl)-4-methyl-pyrazol-1-yl. [0276] In some embodiments, R4a is -C(O)NRxRy wherein Rx is H or C1-4alkyl and Ry is H, C1- 4alkyl, -O-C1-4alkyl, -SO2-Rr, C1-4alkyl-SO2-Rr monocyclic cycloalkyl, -C1-4alkyl(monocyclic cycloalkyl), monocyclic heterocyclyl, or monocyclic heterocycloalkyl, each optionally substituted with one, two, or three Ro substituents; and Rr and Ro are as defined herein. In some embodiments, R4a is -C(O)NRxRy wherein Rx is H or methyl; and Ry is H, methyl, ethyl, butyl, isopropyl, methoxy, -SO2-methyl, C2-4alkyl-SO2-methyl, C2-4alkyl-SO2-N(CH3)2, cyclopropyl, cyclobutyl, cyclopentyl, cyclopropylmethyl, 1-cyclopropylethyl, 2-cyclopropylethyl, cyclobutylmethyl, cyclopentylmethyl, azetidinyl, oxetanyl, pyrrolidinyl, piperidinyl, morpholinyl, piperazinyl, azepanyl, azocanyl, tetrahydrofuranyl, tetrahydropyranyl, substituted azetidinylmethyl, oxetanylmethyl, pyrrolidinylmethyl, piperidinylmethyl, morpholinylmethyl, or piperazinylmethyl, each optionally substituted with one, two, or three methyl, methoxy, fluoro or amino groups. [0277] In some embodiments, the compound of Formula (I) or the pharmaceutically acceptable salt thereof is a compound of Formula (I):
Figure imgf000048_0001
Formula (II):
Figure imgf000048_0002
Formula (III):
Figure imgf000048_0003
Formula (IV):
Figure imgf000049_0001
as defined herein, wherein one or more hydrogen atoms attached to carbon atoms of the compound are replaced by deuterium atoms. [0278] In some embodiments, one or more hydrogen atoms attached to carbon atoms of R1, R2, R3, R4, R5, R1a, R1b, R1c or R4a are replaced by deuterium atoms. [0279] In some embodiments, one or more hydrogen atoms attached to carbon atoms of Ra, Rb, Rc, Rd, Rg, Rh, Rj, Rk, Rl, Rm, Rn, Ro, Rp, Rq, Rr, Rx, Ry, or Rz are replaced by deuterium atoms. In some embodiments, one or more Ra, Rb, Rc, Rd, Rg, Rh, Rj, Rk, Rl, Rm, Rn, Ro, Rp, Rq, Rr, Rx, Ry, or Rz group is a C1-4alkyl group wherein one or more hydrogen atoms attached to carbon atoms are replaced by deuterium atoms. In some embodiments, one or more Ra, Rb, Rc, Rd, Rg, Rh, Rj, Rk, Rl, Rm, Rn, Ro, Rp, Rq, Rr, Rx, Ry, or Rz group is a methyl group wherein one or more hydrogen atoms attached to the carbon atom are replaced by deuterium atoms. In some embodiments, one or more Ra, Rb, Rc, Rd, Rg, Rh, Rj, Rk, Rl, Rm, Rn, Ro, Rp, Rq, Rr, Rx, Ry, or Rz group is -CD3. [0280] In some embodiments, the compound of Formula (I) - (IV) comprises a -D in place of at least one -H, or a -CD3 substituent in place of at least one CH3. [0281] In some embodiments, the compound is a compound selected from those of Table 1: Table 1
Figure imgf000049_0002
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
and pharmaceutically acceptable salts thereof. I. Kits and Articles of Manufacture [0282] Any of the aforementioned methods can be implemented via kits for the treatment of a coronavirus infection. In various embodiments, the coronavirus is SARS-CoV-1, SARS-CoV-2, MERS-CoV, HCoV-OC43, HCoV-HKU1, HCoV-229E, or HCoV-NL63. [0283] The kit may contain a compound of Formula I (or any of the embodiments thereof described herein), a pharmaceutically acceptable carrier, a physiologically acceptable carrier, instructions for use, a container, a vessel for administration, or any combination thereof. [0284] The disclosure further provides any compounds disclosed herein for use in a method of treatment of the human or animal body by therapy. Therapy may be by any mechanism disclosed herein, such as inhibiting, reducing, or reducing progression of the diseases disclosed herein. The disclosure further provides any compound disclosed herein for prevention or treatment of any condition disclosed herein. The disclosure also provides any compound or pharmaceutical composition thereof disclosed herein for obtaining any clinical outcome disclosed herein for any condition disclosed herein. The disclosure also provides use of any compound disclosed herein in the manufacture of a medicament for preventing or treating any disease or condition disclosed herein. EXAMPLES [0285] The following preparations of compounds of Formula (I) and intermediates are given to enable those skilled in the art to more clearly understand and to practice the present disclosure. They should not be considered as limiting the scope of the disclosure, but merely as being illustrative and representative thereof. [0286] The starting materials and reagents used in preparing these compounds are either available from commercial suppliers such as Aldrich Chemical Co., (Milwaukee, Wis.), Bachem (Torrance, Calif.), or Sigma (St. Louis, Mo.) or are prepared by methods known to those skilled in the art following procedures set forth in references such as Fieser and Fieser’s Reagents for Organic Synthesis, Volumes 1-17 (John Wiley and Sons, 1991); Rodd’s Chemistry of Carbon Compounds, Volumes 1-5 and Supplementals (Elsevier Science Publishers, 1989); Organic Reactions, Volumes 1-40 (John Wiley and Sons, 1991), March’s Advanced Organic Chemistry, (John Wiley and Sons, 4th Edition) and Larock’s Comprehensive Organic Transformations (VCH Publishers Inc., 1989). These schemes are merely illustrative of some methods by which the compounds of this disclosure can be synthesized, and various modifications to these schemes can be made and will be suggested to one skilled in the art having referred to this disclosure. The starting materials and the intermediates, and the final products of the reaction may be isolated and purified if desired using conventional techniques, including but not limited to filtration, distillation, crystallization, chromatography and the like. Such materials may be characterized using conventional means, including physical constants and spectral data. [0287] Unless specified to the contrary, the reactions described herein take place at atmospheric pressure over a temperature range from about –78 °C to about 150 °C, or from about 0 °C to about 125 °C or at about room (or ambient) temperature, e.g., about 20 °C. [0288] Compounds of Formula (I) and subformulae and species described herein, including those where the substituent groups as defined herein, can be prepared as illustrated and described below. [0289] Unless otherwise noted, all reagents were used without further purification. 1H NMR spectra were obtained in CDCl3, DMSO-d6, or CD3OD at room temperature on a Bruker 300 MHz instrument. When more than one conformer was detected, the chemical shifts for the most abundant one is reported. Chemical shifts of 1H NMR spectra were recorded in parts per million (ppm) on the δ scale from an internal standard of residual solvent. Splitting patterns are designed as s, singlet; d, doublet; t, triplet; q, quartet; m, multiplet; br, broad. LC-MS conditions were as described below: [0290] LCMS Column: Agilent Zorbax XDB C184.6×50 mm, 3.5µm a. Mobile phase: Solvent A: Water (with 0.1% formic acid); Solvent B: MeOH b. Flow rate: 1.0 mL/min, c. Run time: 2 min gradient (20%-90% B), then 3 min @90% B, d. Temperature: 30 °C [0291] HPLC Column: Agilent SB-C184.6×150 mm, 3.5µm a. Mobile phase: Solvent A: water (with 0.02% TFA); Solvent B: MeOH b. Flow rate: 1.0 mL/min, c. Run time: 0.5 min @10% B, 9.5 min gradient (10%-90% B), then 10 min @90% B, d. Temperature: 30 °C [0292] Preparative LC Column: Phenomenex Luna 5u 100A, 21.2×250mm, 5µm a. Mobile phase: Solvent A: Water Solvent B: MeOH b. Flow rate: 10 mL/min, c. Run time: 1 min @20% B, 30 min gradient (20%-80% B), then 10 min @90% B, d. Temperature: Ambient [0293] The following abbreviations are used in the text: PE = petroleum ether, EA or AcOH = acetic acid, EtOAc = ethyl acetate, DMSO = dimethyl sulfoxide, DMF = N, N- dimethylacetamide, MeOH = methanol, i-PrOH = isopropyl alcohol, MTBE = Methyl tert-butyl ether, DCM = dichloromethane, Et3N or TEA = triethylamine, DIPEA = Diisopropylethylamine, DIEA = N,N-Diisopropylethylamine, TFA = trifluoroacetic acid, TLC = thin layer chromatography, (BPin)2 = Bis(pinacolato)diboron, HFIP = 1,1,1,3,3,3-hexafluoropropan-2-ol, DIBAL-H = Diisobutylaluminum hydride, MeI = Iodomethane, n-Hex = n-Hexane, DCE = 1,2- Dichloroethane, TBSCl = tert-Butyldimethylsilyl chloride, Tf2O = Trifluoromethanesulfonic anhydride, n-BuLi = n-Butyllithium, DMAP = 4-Dimethylaminopyridine, KOAc = Potassium acetate, NaOAc = Sodium acetate, TFAA = Trifluoroacetic anhydride, m-CPBA = meta- Chloroperoxybenzoic acid, DME = 1,2-Dimethoxyethane, PS-TPP = polymer supported triphenylphosphine, MSA = methanesulfonic acid, SEMCl = 2-(Trimethylsilyl)ethoxymethyl chloride, Et2O = diethylether, THF = tetrahydrofuran, NIS = N-Iodosuccinimide, LDA = Lithium diisopropylamide, EDCl = 1-Ethyl-3-(3-dimethylaminopropyl)carbodiimide, TMSCF3 = Trifluoromethyltrimethylsilane, Xantphos = 4,5-Bis(diphenylphosphino)-9,9-dimethylxanthene, h and hr = hour, rt = room temperature, Ph = phenyl, dppf = 1,1'-Bis(diphenylphosphino)- ferrocene, dba = dibenzylideneacetone, [0294] EXAMPLE 1: Compound 1 using General Synthetic Route 1:
Figure imgf000089_0001
1.1) Synthesis of (Z)-methyl 2-((2-cyano-1-(pyridin-4-yl)vinyl)oxy)acetate [0295] To a solution of triphenylph
Figure imgf000089_0002
osphine (3.5 g, 13.4 mmol) in dry THF was added diethyl azodicarboxylate (DEAD) (2.3 g, 13.4 mmol), 3-oxo-3-(pyridin-4-yl)propanenitrile (1.5 g, 10.3 mmol) and methyl 2-hydroxyacetate (1.2 g, 13.4 mmol) under N2. The reaction was stirred at ambient temperature overnight. The reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 2% MeOH/DCM to provide 4.4 g of impure (Z)-methyl2-((2-cyano-1-(pyridin-4- yl)vinyl)oxy)acetate containing triphenylphosphine oxide as a yellow solid. LC-MS (ESI+): m/z 219 (MH+). 1.2) Synthesis of methyl 3-amino-5-(pyridin-4-yl)furan-2-carboxylate
Figure imgf000089_0003
[0296] To a solution of impure (Z)-methyl2-((2-cyano-1-(pyridin-4-yl)vinyl)oxy)acetate (4.6 g, 21.1 mmol) in dry THF at 0 °C was added NaH (1.5 g, 31.6 mmol). The reaction was warmed to room temperature and stirred for 2 h. The reaction mixture was quenched with a saturated NH4Cl solution and the pH was adjusted to 3 using 2 N HCl aqueous solutions. The aqueous solution was extracted with ethyl acetate (3 x 50 mL) to remove some impurities. To the remaining aqueous solution was added a saturated Na2CO3 solution to adjust the pH to 11. The resulting aqueous solution was extracted with DCM/MeOH (10/1, 3 x 60 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated to provide 1.35 g of crude methyl 3-amino-5-(pyridin-4-yl)furan-2-carboxylate as an oil. The crude product was used directly for the next step without further purification. LC-MS (ESI+): m/z 219 (MH+).1HNMR (300 MHz, CDCl3) δ 8.66 (dd, J = 4.5, 1.5 Hz, 2H), 7.58 (dd, J = 4.8, 1.2 Hz, 2H), 6.58 (s, 1H), 4.67 (brs, 2H), 3.93 (s, 3H). 1.3) Synthesis of methyl 5-(pyridin-4-yl)-3-ureidofuran-2-carboxylate [0297] To a solution of methyl 3-am
Figure imgf000090_0001
ino-5-(pyridin-4-yl)furan-2-carboxylate (1.94 g, 8.9 mmol) in DCM (40 mL) at -78 °C under N2 was added chlorosulfonyl isocyante (3.79 g, 26.7 mmol) dropwise. After addition, the reaction was warmed to room temperature and stirred for 1 h. After removal of DCM by evaporation, the resulting residue was treated with 6 N HCl (10 mL) aqueous solutions. The mixture was heated to reflux for 30 min. The completion of the reaction was monitored by thin layer chromatography (TLC). The reaction was cooled to room temperature and the pH was adjusted to 9 using a saturated NaHCO3 solution. A large amount of solid was precipitated. After filtration, the filter cake was washed with water and dried to provide 2.7 g of crude methyl5-(pyridin-4-yl)-3-ureidofuran-2-carboxylate as a yellow solid. LC-MS (ESI+): m/z 262 (MH+).1HNMR (300 MHz, CD3OD) δ 8.61 (dd, J = 4.8, 1.5 Hz, 2H), 7.89 (s, 1H), 7.78 (dd, J = 5.1, 1.5 Hz, 2H), 3.95 (s, 3H). 1.4) Synthesis of 6-(pyridin-4-yl)furo[3,2-d]pyrimidine-2,4-diol
Figure imgf000090_0002
[0298] To a solution of crude methyl 5-(pyridin-4-yl)-3-ureidofuran-2-carboxylate (2.7 g, 10.3 mmol) in MeOH (40 mL) was added 1.5 N NaOH (15 mL). The reaction was heated to reflux for 1.5 h. The completion of the reaction was monitored by TLC. The solvent MeOH was removed by evaporation. To the resulting residue were added 6 N HCl solutions until the pH was adjusted to 2. A large amount of solid was precipitated. After filtration, the filter cake was washed with water and dried to provide crude 2.1 g of 6-(pyridin-4-yl)furo[3,2-d]pyrimidine- 2,4-diol as a yellow solid. LC-MS (ESI+): m/z 230 (MH+).1HNMR (300 MHz, DMSO-d6) δ 11.61 (s, 1H), 11.37 (s, 1H), 8.89 (d, J = 6.6 Hz, 2H), 8.24 (d, J = 6.3 Hz, 2H), 7.63 (s, 1H). 1.5) Synthesis of 2,4-dichloro-6-(pyridin-4-yl)furo[3,2-d]pyrimidine
Figure imgf000091_0001
[0299] To a solution of 6-(pyridin-4-yl)furo[3,2-d]pyrimidine-2,4-diol (1.5 g, 6.54 mmol) in phenylphosphonic dichloride (30 mL) was added DIPEA (8.43 g, 65.4 mmol). The reaction was heated to 120 °C overnight. After the reaction mixture was cooled to room temperature, a saturated NaHCO3 solution was added to adjust the pH to 8. The aqueous solution was extracted with EtOAc (3 x 50 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated. The resulting residue was purified by silica gel column chromatography with a gradient elution of 50% EtOAc/PE to 75% EtOAc/PE to provide 2,4- dichloro-6-(pyridin-4-yl)furo[3,2-d]pyrimidine (1.6 g, 6.9 mmol) as a yellow solid. LC-MS (ESI+): m/z 266/268 (MH+).1HNMR (300 MHz, CDCl3) δ 8.85 (dd, J = 4.8, 1.5 Hz, 2H), 7.82 (dd, J = 4.5, 1.5 Hz, 2H), 7.38 (s, 1H). 1.6) Synthesis of 2-chloro-4-morpholino-6-(pyridin-4-yl)furo[3,2-d]pyrimidine
Figure imgf000091_0002
[0300] To a solution of 2,4-dichloro-6-(pyridin-4-yl)furo[3,2-d]pyrimidine (1.6 g, 6.9 mmol) in DCM/EtOH (1/3, 120 mL) was added morpholine (0.91 g, 10.5 mmol) and K2CO3 (1.91 g, 14 mmol). The reaction was stirred at room temperature for 2 h. The reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 2% MeOH/DCM to 3% MeOH/DCM to provide 2-chloro-4-morpholino-6-(pyridin-4-yl) furo[3,2-d]pyrimidine (770 mg, 2.43 mmol) as a yellow solid. LC-MS (ESI+): m/z 317/319 (MH+).1HNMR (300 MHz, DMSO-d6) δ 8.76 (d, J = 6.0 Hz, 2H), 7.97 (d, J = 6.0 Hz, 2H), 7.82 (s, 1H), 4.06-3.97 (m, 4H), 3.82-3.75 (m, 4H). 1.6) Synthesis of 2-bromo-4-morpholino-6-(pyridin-4-yl)furo[3,2-d]pyrimidine
Figure imgf000092_0001
[0301] A solution of 2,4-dichloro-6-(pyridin-4-yl)furo[3,2-d]pyrimidine (770 mg, 2.43 mmol) in HBr/AcOH (33 wt.% in Acetic acid, 10 mL) was heated to refluxed for 3.5 h. The completion of the reaction was monitored by LC-MS. The reaction mixture was quenched with a saturated NaHCO3 solution and the pH was adjusted to 8. The aqueous solution was extracted with DCM/MeOH (15/1, 3 x 50 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure to provide 740 mg of crude 2-bromo- 4-morpholino-6-(pyridin-4-yl)furo[3,2-d]pyrimidine as a brown solid. The crude product was used directly for the next step without further purification. LC-MS (ESI+): m/z 362/364 (MH+). 1HNMR (300 MHz, DMSO-d6) δ 8.76 (d, J = 4.5 Hz, 2H), 7.98 (d, J = 4.5 Hz, 2H), 7.82 (s, 1H), 4.02-3.95 (m, 4H), 3.83-3.76 (m, 4H). 1.7) Synthesis of 5-amino-N,N-dimethyl-3-phenyl-1H-pyrazole-1-sulfonamide [0302] To a solution of 3-pheny
Figure imgf000092_0002
l-1H-pyrazol-5-amine (300 mg, 1.88 mmol) in THF (5 mL) at 0 °C was added NaH (100 mg, 2.82 mmol). After stirring at 0 °C for 1 h, to the solution was added dimethylsulfamoyl chloride (315 mg, 2.20 mmol). The completion of the reaction was monitored by TLC. The reaction mixture was quenched with a saturated NH4Cl solution. The aqueous solution was extracted with ethyl acetate (3 x 50 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 20% EtOAc/PE to 33% EtOAc/PE to provide 5-amino-N,N-dimethyl-3-phenyl-1H-pyrazole-1- sulfonamide (300 mg, 1.13 mmol). LC-MS: m/z 267 (MH+).1HNMR (300 MHz, CDCl3) δ 7.79- 7.76 (m, 2H), 7.42-7.35 (m, 3H), 5.75 (s, 1H), 4.84 (s, 2H), 3.03 (s, 6H). 1.8) Synthesis of N,N-dimethyl-5-((4-morpholino-6-(pyridin-4-yl)furo[3,2-d]pyrimidin-2- yl)amino)-3-phenyl-1H-pyrazole-1-sulfonamide
Figure imgf000093_0002
[0303] A suspension of 2-bromo-4-morpholino-6-(pyridin-4-yl)furo[3,2-d]pyrimidine (500 mg, 1.4 mmol), 3-amino-N,N-dimethyl-5-phenyl-1H-pyrazole-1-sulfonamide (554 mg, 2.1 mmol), Cs2CO3 (906 mg, 2.8 mmol), Pd(OAc)2 (30 mg, 0.1 mmol) and Xantphos (80 mg, 0.1 mmol) in DMF/1,4-dioxane (1/7, 16 mL) was heated to 100 °C for 40 min under microwave conditions. The reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 2% MeOH/DCM to provide N,N-dimethyl-5-((4-morpholino-6-(pyridin-4-yl)furo[3,2-d]pyrimidin-2-yl)amino)-3-phenyl- 1H-pyrazole-1-sulfonamide (140 mg, 0.26 mmol) as a white solid. LC-MS (ESI+): m/z 547 (MH+).1HNMR (300 MHz, CDCl3) δ 8.76-8.71 (m, 3H), 7.91 (d, J = 6.9 Hz, 2H), 7.64 (d, J = 5.7 Hz, 2H), 7.47-7.39 (m, 3H), 7.28-7.22 (m, 2H), 4.14-4.08 (m, 4H), 3.94-3.87 (m, 4H), 3.07 (s, 6H). 1.9) Synthesis of 4-morpholino-N-(3-phenyl-1H-pyrazol-5-yl)-6-(pyridin-4-yl)furo[3,2- d]pyrimidin-2-amine hydrochloride
Figure imgf000093_0001
[0304] To a solution of N,N-dimethyl-5-((4-morpholino-6-(pyridin-4-yl)furo[3,2-d]pyrimidin- 2-yl)amino) -3-phenyl-1H-pyrazole-1-sulfonamide (140 mg, 0.26 mmol) in DCM (4 mL) was added HCl/Et2O (2 mL). The reaction mixture was stirred at room temperature for 2 h. After concentration and slurry in MeOH/Et2O (1/20, 2 mL), 4-morpholino-N-(3-phenyl-1H-pyrazol-5- yl)-6-(pyridin-4-yl)furo[3,2-d] pyrimidin-2-amine hydrochloride (Compound 1, 112 mg, 0.22 mmol) was obtained as a yellow solid. LC-MS (ESI+): m/z 440 (MH+).1HNMR (300 MHz, CD3OD) δ 8.97 (d, J = 6.6 Hz, 2H), 8.55 (d, J = 6.6 Hz, 2H), 8.05 (s, 1H), 7.37 (d, J = 6.9 Hz, 2H), 7.66-7.40 (m, 3H), 6.44 (s, 1H), 4.35-4.27 (m, 4H), 4.01-3.92 (m, 4H). [0305] EXAMPLE 2: Compound 10 using General Synthetic Route 2:
Figure imgf000094_0001
1.1) Synthesis of 5-amino-N,N-dimethyl-3-(pyridin-4-yl)-1H-pyrazole-1-sulfonamide [0306] To a solution of 3-(pyridin
Figure imgf000094_0002
-4-yl)-1H-pyrazol-5-amine (500 mg, 3.12 mmol) in THF (5 mL) at 0°C was added NaH (374 mg, 9.36 mmol). After stirred at 0°C for 1 h, to the solution was added dimethylsulfamoyl chloride (536 mg, 3.75 mmol). The completion of the reaction was monitored by TLC. The reaction mixture was quenched with a saturated NH4Cl solution. The aqueous solution was extracted with ethyl acetate (3 x 50 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 20% EtOAc/PE to 33% EtOAc/PE to provide 5-amino-N,N-dimethyl-3-(pyridin-4-yl)- 1H- pyrazole-1-sulfonamide (94 mg, 0.35 mmol). LC-MS: m/z 268 (MH+).1HNMR (300 MHz, DMSO-d6) δ 8.50 (d, J = 6.0 Hz, 2H), 7.60 (dd, J = 4.5, 1.2 Hz, 2H), 6.04 (s, 2H), 5.79 (s, 1H), 2.81 (s, 6H). 1.2) Synthesis of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine [0307] To a solution of 2,4-dichlorofu
Figure imgf000095_0001
ro[3,2-d]pyrimidine (6.46 g, 34.2 mmol mmol) in methanol (100 mL) was added morpholine (5.95 g, 68.4 mmol). The reaction was stirred at room temperature for 30 min. The completion of the reaction was monitored by TLC. The reaction mixture was concentrated directly and the resulting residue was purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 20% EtOAc/PE to provide 2- chloro-4-morpholinofuro [3,2-d]pyrimidine (7.5 g, 40.1 mmol) as a white solid. LC-MS (ESI+): m/z 240/242 (MH+).1HNMR (300 MHz, CDCl3) δ 7.74 (d, J = 1.8 Hz, 1H), 6.79 (d, J = 2.1 Hz, 1H), 4.05-4.02 (m, 4H), 3.85-3.82 (m, 4H). 1.3) Synthesis of 2-bromo-4-morpholinofuro[3,2-d]pyrimidine
Figure imgf000095_0002
[0308] A solution of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine (180 mg, 0.75 mmol) in HBr/AcOH (33 wt.% in Acetic acid, 3 mL) was heated to reflux for 3.5 h. The completion of the reaction was monitored by LC-MS. The reaction mixture was quenched with a saturated NaHCO3 solution and the pH was adjusted to 8. The aqueous solution was extracted with DCM/MeOH (15/1, 3 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure to provide 175 mg of crude 2-bromo- 4-morpholino- furo[3,2-d]pyrimidine as a yellow solid. The crude product was used directly for the next step without further purification. LC-MS (ESI+): m/z 284/286 (MH+).1HNMR (300 MHz, CDCl3) δ 7.72 (d, J = 2.1 Hz, 1H), 6.79 (d, J = 2.1 Hz, 1H), 4.04-4.01 (m, 4H), 3.85-3.81 (m, 4H). 1.4) Synthesis of N,N-dimethyl-5-((4-morpholinofuro[3,2-d]pyrimidin-2-yl)amino)-3-(pyridin-4- yl)-1H-pyrazole-1-sulfonamide
Figure imgf000096_0001
[0309] A suspension of 2-bromo-4-morpholinofuro[3,2-d]pyrimidine (48 mg, 0.17 mmol), 5- amino-N,N-dimethyl-3-(pyridin-4-yl)-1H-pyrazole-1-sulfonamide (54 mg, 0.20 mmol), Cs2CO3 (126 mg, 0.39 mmol), Pd(OAc)2 (4 mg, 0.017 mmol) and Xantphos (10 mg, 0.017 mmol) in DMF/1,4-dioxane (1/7, 3 mL) was heated to 100 °C for 40 min under microwave conditions. The reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide N,N-dimethyl-5-((4-morpholinofuro[3,2-d]pyrimidin-2-yl)amino)-3-(pyridin-4-yl)-1H-pyrazole- 1-sulfonamide (27 mg, 0.06 mmol) as a yellow solid. LC-MS (ESI+): m/z 471 (MH+).1HNMR (300 MHz, CDCl3) δ 8.69-8.67 (m, 3H), 7.77 (d, J = 6.0 Hz, 2H), 7.70 (d, J = 2.1 Hz, 1H), 7.35 (s, 1H), 6.79 (d, J = 2.1 Hz, 1H), 4.05-4.02 (m, 4H), 3.87-3.84 (m, 4H), 3.08 (s, 6H). 1.5) Synthesis of 4-morpholino-N-(3-(pyridin-4-yl)-1H-pyrazol-5-yl)furo[3,2-d]pyrimidin-2- amine hydrochloride
Figure imgf000096_0002
[0310] To a solution of N,N-dimethyl-5-((4-morpholinofuro[3,2-d]pyrimidin-2-yl)amino)-3- (pyridin-4-yl) -1H-pyrazole-1-sulfonamide (27 mg, 0.06 mmol) in DCM (4 mL) was added HCl/Et2O (2 mL). The reaction mixture was stirred at room temperature for 2 h. After concentration and slurry in MeOH/Et2O (1/20, 2 mL), 4-morpholino-N-(3-(pyridin-4-yl)-1H- pyrazol-5-yl)furo[3,2-d]pyrimidin-2-amine hydrochloride (Compound 10, 21.2 mg, 0.042 mmol) was obtained as a yellow solid. LC-MS (ESI+): m/z 364 (MH+).1HNMR (300 MHz, CD3OD) δ 8.90 (d, J = 6.9 Hz, 2H), 8.42 (d, J = 6.9 Hz, 2H), 8.17 (d, J = 2.1 Hz, 1H), 7.06-7.04 (m, 2H), 4.22-4.10 (m, 4H), 3.92-3.86 (m, 4H). [0311] EXAMPLE 3: Compound 11 using General Synthetic Route 3:
Figure imgf000097_0001
1.1) Synthesis of 2-bromo-6-chloro-4-morpholinofuro[3,2-d]pyrimidine
Figure imgf000097_0002
[0312] To a solution of 2-bromo-4-morpholinofuro[3,2-d]pyrimidine (1.3 g, 4.59 mmol) in dry THF (4 mL) at -78 °C was added LDA (7.5 mL, 14.7 mmol) dropwise. After addition, the solution was stirred at that temperature for 1 h. Then to the solution was added NCS (733 mg, 5.5 mmol). The completion of the reaction was monitored by TLC. The reaction mixture was quenched with water (30 mL). The aqueous solution was extracted with EtOAc (3 x 30 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The residue was purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 25% EtOAc/PE to provide 2-bromo-6-chloro- 4- morpholinofuro[3,2-d]pyrimidine (550 mg, 1.73 mmol) as a light yellow solid. LC-MS (ESI+): m/z 318/320 (MH+).1HNMR (300 MHz, CD3OD) δ6.77 (s, 1H), 3.99-3.95 (m, 4H), 3.82-3.79 (m, 4H). 1.2) Synthesis of 3-((6-chloro-4-morpholinofuro[3,2-d]pyrimidin-2-yl)amino)-N,N-dimethyl-5- phenyl-1H-pyrazole-1-sulfonamide
Figure imgf000097_0003
[0313] A suspension of 2-bromo-6-chloro-4-morpholinofuro[3,2-d]pyrimidine (3 x 50 mg, 0.16 mmol), 3-amino-N,N-dimethyl-5-phenyl-1H-pyrazole-1-sulfonamide (42 mg, 0.16 mmol), Cs2CO3 (118 mg, 0.36 mmol), Pd(OAc)2 (3.5 mg, 0.016 mmol) and Xantphos (9 mg, 0.015 mmol) in DMF/1,4-dioxane (1/7, 3 mL) was heated to 80 °C for 40 min under microwave conditions. The reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 35% EtOAc/PE to provide the impure product. After further preparative HPLC purification, 26 mg of pure 3-((6-chloro-4- morpholinofuro [3,2-d]pyrimidin-2-yl)amino)-N,N-dimethyl-5-phenyl-1H-pyrazole-1- sulfonamide was obtained. LC-MS (ESI+): m/z 504/506 (MH+).1HNMR (300 MHz, CDCl3) δ 8.67 (s, 1H), 7.89 (dd, J = 8.1, 1.5 Hz, 2H), 7.46-7.39 (m, 3H), 7.21 (s, 1H), 6.60 (s, 1H), 3.99- 3.96 (m, 4H), 3.87-3.84 (m, 4H), 3.06 (s, 6H). 1.3) Synthesis of 6-chloro-4-morpholino-N-(5-phenyl-1H-pyrazol-3-yl)furo[3,2-d]pyrimidin-2- amine hydrochloride
Figure imgf000098_0001
[0314] To a solution of 3-((6-chloro-4-morpholinofuro[3,2-d]pyrimidin-2-yl)amino)-N,N- dimethyl-5- phenyl-1H-pyrazole-1-sulfonamide (26 mg, 0.05 mmol) in DCM (4 mL) was added HCl/Et2O (2 mL). The reaction mixture was stirred at room temperature for 2 h. After concentration and slurry in MeOH/Et2O (1/20, 2 mL), 6-chloro-4-morpholino-N-(5-phenyl-1H- pyrazol-3-yl)furo[3,2-d]pyrimidin-2-amine hydrochloride (Compound 11, 18.5 mg, 0.043 mmol) was obtained. LC-MS (ESI+): m/z 397/399 (MH+).1HNMR (300 MHz, CD3OD) δ 7.87- 7.69 (m, 2H), 7.51-7.39 (m, 3H), 7.09 (s, 1H), 6.41 (s, 1H), 4.17-4.12 (m, 4H), 3.89-3.84 (m, 4H). [0315] EXAMPLE 4: Compound 12 using General Synthetic Route 4:
Figure imgf000098_0002
1.1) Synthesis of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carbaldehyde
Figure imgf000099_0001
[0316] To a solution of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine (50 mg, 0.21 mmol) in THF at -78 °C under N2 was added n-BuLi ( 0.1 mL, 0.25 mmol). The mixture was stirred at that temperature for 15 min and then DMF (90 mg, 1.23 mmol) was added. The solution was allowed to warm to room temperature for 10 min. The completion of the reaction was monitored by TLC. The reaction was quenched with water and the aqueous solution was extracted with EtOAc (3 x 10 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 35% EtOAc/PE to provide 2- chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carbaldehyde (26 mg, 0.097 mmol) as a white solid. LC-MS (ESI+): m/z 268/270 (MH+).1HNMR (300 MHz, CDCl3) δ 9.91 (s, 1H), 7.48 (s, 1H), 4.15-4.10 (m, 4H), 3.88-3.85 (m, 4H). 1.2) Synthesis of 2-chloro-4-morpholino-6-(morpholinomethyl)furo[3,2-d]pyrimidine [0317] A solution of 2-chloro-4-
Figure imgf000099_0002
morpholinofuro[3,2-d]pyrimidine-6-carbaldehyde (150 mg, 0.56 mmol) and morpholine (58 mg, 0.67 mmol) in DCM was stirred at room temperature for 15 min. To the solution was added sodium triacetoxyborohydride (356 mg, 1.68 mmol). The mixture was stirred at room temperature for 3 h. The completion of the reaction was monitored by TLC. The reaction mixture was quenched with a saturated NaHCO3 solution and the pH was adjusted to 8. The aqueous solution was extracted with DCM/MeOH (15/1, 3 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 30% EtOAc/PE to EtOAc to provide 2-chloro-4-morpholino-6- (morpholinomethyl)furo[3,2-d]pyrimidine (120 mg, 0.35 mmol). LC-MS (ESI+): m/z 339/341 (MH+).1HNMR (300 MHz, CDCl3) δ 6.63 (s, 1H), 4.13-3.92 (m, 4H), 3.85-3.82 (m, 4H), 3.74- 3.71 (m, 4H), 3.63 (s, 2H), 2.56-2.53 (m, 4H). 1.3) Synthesis of 2-bromo-4-morpholino-6-(morpholinomethyl)furo[3,2-d]pyrimidine
Figure imgf000100_0001
[0318] A solution of 2-chloro-4-morpholino-6-(morpholinomethyl)furo[3,2-d]pyrimidine (120 mg, 0.35 mmol) in HBr/AcOH (33 wt.% in Acetic acid, 5 mL) was heated to refluxed for 3.5 h. The completion of the reaction was monitored by LC-MS. The reaction mixture was quenched with a saturated NaHCO3 solution and the pH was adjusted to 8. The aqueous solution was extracted with DCM/MeOH (15/1, 3 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure to provide 80 mg of crude 2-bromo-4-morpholino-6-(morpholinomethyl)furo[3,2-d]pyrimidine as a brown solid. The crude product was used directly for the next step without further purification. LC-MS (ESI+): m/z 383/385 (MH+).1HNMR (300 MHz, CD3OD) δ 6.71 (s, 1H), 4.03-4.00 (m, 4H), 3.83-3.81 (m, 4H), 3.75 (s, 2H), 3.72-3.68 (m, 4H), 2.57-2.54 (m, 4H). 1.4) Synthesis of N,N-dimethyl-5-((4-morpholino-6-(morpholinomethyl)furo[3,2-d]pyrimidin-2- yl)amino)-3-phenyl-1H-pyrazole-1-sulfonamide
Figure imgf000100_0002
[0319] A suspension of 2-bromo-4-morpholino-6-(morpholinomethyl)furo[3,2-d]pyrimidine (60 mg, 0.16 mmol), 5-amino-N,N-dimethyl-3-phenyl-1H-pyrazole-1-sulfonamide (50 mg, 0.19 mmol), Cs2CO3 (120 mg, 0.37 mmol), Pd(OAc)2 (3 mg, 0.01 mmol) and Xantphos (6 mg, 0.01 mmol) in DMF/1,4-dioxane (1/7, 8 mL) was heated to 90 °C for 30 min under microwave condition. The reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide N,N-dimethyl-5-((4- morpholino-6-(morpholinomethyl)furo[3,2-d]pyrimidin-2-yl)amino)-3- phenyl-1H-pyrazole-1-sul-fonamide (23 mg, 0.04 mmol) as a yellow solid. LC-MS (ESI+): m/z 569 (MH+).1HNMR (300 MHz, CDCl3) δ 8.65 (s, 1H), 7.89 (d, J = 6.9 Hz, 2H), 7.45-7.38 (m, 3H), 7.25 (s, 1H), 6.63 (s, 1H), 4.02-4.00 (m, 4H), 3.87-3.84 (m, 4H), 3.75-3.73 (m, 4H), 3.67 (s, 2H), 3.06 (s, 6H), 2.58-2.55 (m, 4H). 1.5) Synthesis of 4-morpholino-6-(morpholinomethyl)-N-(3-phenyl-1H-pyrazol-5-yl)furo[3,2- d]pyrimidin-2-amine hydrochloride
Figure imgf000101_0001
[0320] To a solution of N,N-dimethyl-5-((4-morpholino-6-(morpholinomethyl)furo[3,2- d]pyrimidin -2-yl)amino)-3-phenyl-1H-pyrazole-1-sulfonamide (23 mg, 0.04 mmol) in DCM (4 mL) was added HCl/Et2O (2 mL). The reaction mixture was stirred at room temperature for 2 h. After concentration and slurry in MeOH/Et2O (1/20, 2 mL), 4-morpholino-6- (morpholinomethyl)-N-(3-phenyl-1H- pyrazol-5-yl)furo[3,2-d]pyrimidin-2-amine hydrochloride (Compound 12, 21.2 mg, 0.042 mmol) was obtained as a yellow solid. LC-MS (ESI+): m/z 462 (MH+).1HNMR (300 MHz, CD3OD) δ 7.79-7.70 (m, 2H), 7.52-7.42 (m, 3H), 7.39 (s, 1H), 6.42 (s, 1H), 4.67 (s, 2H), 4.32-4.12 (m, 4H), 4.10-3.86 (m, 8H), 3.55-3.42 (m, 4H). [0321] EXAMPLE 5: Compound 22 using General Synthetic Route 5:
Figure imgf000101_0002
1.1) Synthesis of 3-(2-methylpyridin-4-yl)-3-oxopropanenitrile [0322] To a solution of acetonitrile (1
Figure imgf000102_0001
.63 g, 39.7 mmol) in anhydrous THF (40 mL) at -70 °C under N2 was added n-BuLi (15.9 mL, 39.7 mmol) dropwise. After addition, a solution of methyl 2-methylisonicotinate (2.0 g, 13.2 mmol) in THF (10 mL) was added to the above solution over 10 min. The reaction mixture was stirred at that temperature for 2 h. The completion of the reaction was monitored by TLC. The reaction was quenched with AcOH (6.9 mL) and the solution was concentrated directly under reduced pressure. The residue was purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide 3-(2-methylpyridin-4-yl)-3-oxopropanenitrile (1.15 g, 7.2 mmol) as a yellow solid. LC-MS (ESI+): m/z 161 (MH+).1HNMR (300 MHz, CDCl3) δ 8.77 (d, J = 5.4 Hz, 1H), 7.58 (s, 1H), 7.51 (d, J = 5.1 Hz, 1H), 4.08 (s, 2H), 2.69 (s, 3H). 1.2) Synthesis of 3-(2-methylpyridin-4-yl)-1H-pyrazol-5-amine
Figure imgf000102_0002
[0323] To a solution of 3-(2-methylpyridin-4-yl)-3-oxopropanenitrile (1.15 g, 7.2 mmol) in EtOH (40 mL) was added NH2NH2.H2O (0.54 g, 10.8 mmol). The mixture was heated to reflux overnight. The completion of the reaction was monitored by TLC. The reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide 3-(2-methylpyridin-4-yl)-1H-pyrazol-5- amine (0.8 g, 4.6 mmol) as a yellow oil. LC-MS (ESI+): m/z 175 (MH+).1HNMR (300 MHz, CDCl3) δ 8.47 (d, J = 5.4Hz, 1H), 7.33 (s, 1H), 7.27 (d, J = 5.1 Hz, 1H), 6.00 (s, 1H), 4.70 (brs, 2H), 2.55 (s, 3H). 1.3) Synthesis of 5-amino-N,N-dimethyl-3-(2-methylpyridin-4-yl)-1H-pyrazole-1-sulfonamide [0324] To a solution of 3
Figure imgf000102_0003
-(2-methylpyridin-4-yl)-1H-pyrazol-5-amine (800 mg, 4.6 mmol) in THF (30 mL) at 0 °C was added NaH (413 mg, 6.9 mmol). After stirred at 0 °C for 1 h, to the reaction solution was added dimethylsulfamoyl chloride (854 mg, 5.98 mmol). The completion of the reaction was monitored by TLC. The reaction mixture was quenched with a saturated NH4Cl solution. The aqueous solution was extracted with DCM/MeOH (15/1, 3 x 50 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 50% EtOAc/PE to 66% EtOAc/PE to provide 5-amino-N,N-dimethyl-3-(2- methylpyridin-4-yl)-1H-pyrazole-1-sulfonamide (220 mg, 0.78 mmol). LC-MS: m/z 282 (MH+). 1HNMR (300 MHz, CDCl3) δ 8.51 (d, J = 5.1Hz, 1H), 7.52 (s, 1H), 7.43 (d, J = 5.4Hz, 1H), 5.78 (s, 1H), 4.91 (brs, 2H), 3.05 (s, 6H), 2.60 (s, 3H). 1.4) Synthesis of N,N-dimethyl-3-(2-methylpyridin-4-yl)-5-((4-morpholino-6-(pyridin-4- yl)furo[3,2-d]pyrimidin-2-yl)amino)-1H-pyrazole-1-sulfonamide
Figure imgf000103_0001
[0325] A suspension of 2-bromo-4-morpholino-6-(pyridin-4-yl)furo[3,2-d]pyrimidine (2 x 50 mg, 0.14 mmol), 5-amino-N,N-dimethyl-3-(2-methylpyridin-4-yl)-1H-pyrazole-1-sulfonamide (46.8 mg, 0.17 mmol), Cs2CO3 (90.6 mg, 0.28 mmol), Pd(OAc)2 (3 mg, 0.014 mmol) and Xantphos (6 mg, 0.014 mmol) in DMF/1,4-dioxane (1/7, 4 mL) was heated to 100 °C for 30 min under microwave condition. The reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide impure N,N-dimethyl-3-(2-methylpyridin-4-yl)-5-((4-morpholino-6- (pyridin-4-yl)furo [3,2-d]pyrimidin-2-yl)amino)-1H-pyrazole-1-sulfonamide (23.2 mg, 0.04 mmol) as a yellow solid. LC-MS (ESI+): m/z 562 (MH+).1HNMR (300 MHz, CDCl3) δ 8.76 (d, J = 6.0 Hz, 2H), 8.71 (s, 1H), 8.57 (d, J = 5.4 Hz, 1H), 7.66-7.61 (m, 3H), 7.58 (d, J = 4.2 Hz, 1H), 7.35 (s, 1H), 4.10-4.08 (m, 4H), 3.93-3.90 (m, 4H), 3.09 (s, 6H), 2.65 (s, 3H). 1.5) Synthesis of N-(3-(2-methylpyridin-4-yl)-1H-pyrazol-5-yl)-4-morpholino-6-(pyridin-4- yl)furo[3,2-d]pyrimidin-2-amine hydrochloride
Figure imgf000103_0002
[0326] To a solution of impure N,N-dimethyl-3-(2-methylpyridin-4-yl)-5-((4-morpholino-6- (pyridin-4-yl) furo[3,2-d]pyrimidin-2-yl)amino)-1H-pyrazole-1-sulfonamide (23 mg, 0.04 mmol) in DCM (4 mL) was added HCl/Et2O (2 mL). The reaction mixture was stirred at room temperature for 2 h. After concentration and slurry in MeOH/Et2O (1/20, 2 mL), N-(3-(2- methylpyridin-4-yl)-1H- pyrazol- 5-yl)-4-morpholino-6-(pyridin-4-yl)furo[3,2-d]pyrimidin-2- amine hydrochloride (Compound 22, 20.8 mg, 0.04 mmol) was obtained as a yellow solid. LC- MS (ESI+): m/z 455 (MH+).1HNMR (300 MHz, CD3OD) δ 8.94 (d, J = 6.6 Hz, 2H), 8.74 (d, J = 6.6 Hz, 1H), 8.51 (d, J = 6.9 Hz, 2H), 8.30 (s, 1H), 8.24 (d, J = 6.3 Hz, 1H), 8.02 (s, 1H), 6.99 (s, 1H), 4.32-4.28 (m, 4H), 3.95-3.94 (m, 4H), 2.85 (s, 3H). [0327] EXAMPLE 6: Compound 28 using General Synthetic Route 6:
Figure imgf000104_0001
1.1) Synthesis of 2-chloro-6-iodo-4-morpholinofuro[3,2-d]pyrimidine [0328] To a solution of 2-chloro
Figure imgf000104_0002
-4-morpholinofuro[3,2-d]pyrimidine (2.0 g, 0.83 mmol) in THF (30 mL) at -78 °C under N2 was added LDA (1.33 mL, 2M, 2.66 mmol). After stirred at - 78 °C for 1 h, to the solution was added NIS (2.25 g, 1.0 mmol) in THF (10 mL). The completion of the reaction was monitored by TLC. The reaction mixture was quenched with water (50 mL). The aqueous solution was extracted with DCM (3 x 50 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 5% EtOAc/PE to 10% EtOAc/PE to provide 2-chloro-6-iodo-4- morpholinofuro[3,2-d]pyrimidine (1.6 g, 4.4 mmol) as yellow solid. LC-MS (ESI+): m/z 366/368 (MH+).1HNMR (300 MHz, CDCl3) δ 6.97 (s, 1H), 4.01-3.98 (m, 4H), 3.85-3.82 (m, 4H). 1.2) Synthesis of 2-chloro-4-morpholino-6-(pyridin-2-yl)furo[3,2-d]pyrimidine
Figure imgf000105_0001
[0329] A solution of 2-chloro-6-iodo-4-morpholinofuro[3,2-d]pyrimidine (1 g, 2.7 mmol), 2- (tributylstannyl)pyridine (1.2 g, 3.3 mmol) and Pd(PPh3)4 (155 mg, 0.14 mmol) in toluene (5 mL) was heated to 90 °C overnight. The completion was monitored by TLC. The reaction mixture was diluted with water and extracted with DCM/MeOH (15/1, 3 x 50mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide 2-chloro-4-morpholino- 6- (pyridin-2-yl)furo[3,2-d]pyrimidine (352 mg, 1.11 mmol) as a yellow solid. LC-MS (ESI+): m/z 317/319 (MH+). 1.3) Synthesis of 2-bromo-4-morpholino-6-(pyridin-2-yl)furo[3,2-d]pyrimidine [0330] A solution of 2-chloro-4-
Figure imgf000105_0002
morpholino-6-(pyridin-2-yl)furo[3,2-d]pyrimidine (350 mg, 1.11 mmol) in HBr/AcOH (33 wt.% in Acetic acid, 5 mL) was heated to reflux for 3.5 h. The completion of the reaction was monitored by LC-MS. The reaction mixture was quenched with a saturated NaHCO3 solution and the pH was adjusted to 8. The aqueous solution was extracted with DCM/MeOH (15/1, 3 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure to provide 290 mg of crude 2-bromo- 4-morpholino-6-(pyridin-2-yl)furo[3,2-d]pyrimidine as a yellow solid. The crude product was used directly for the next step without further purification. LC-MS (ESI+): m/z 361/363 (MH+). 1.4) Synthesis of N,N-dimethyl-5-((4-morpholino-6-(pyridin-2-yl)furo[3,2-d]pyrimidin-2- yl)amino)-3-(pyridin-4-yl)-1H-pyrazole-1-sulfonamide
Figure imgf000106_0001
[0331] A suspension of 2-bromo-4-morpholino-6-(pyridin-2-yl)furo[3,2-d]pyrimidine (2 x 40 mg, 0.13 mmol), 3-amino-N,N-dimethyl-5-(pyridin-4-yl)-1H-pyrazole-1-sulfonamide (41 mg, 0.15 mmol), Cs2CO3 (95 mg, 0.29 mmol), Pd(OAc)2 (3 mg, 0.013 mmol) and Xantphos (7 mg, 0.013 mmol) in DMF/1,4-dioxane (1/7, 4 mL) was heated to 90 °C for 30 min under microwave conditions. The reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide N,N-dimethyl-5-((4-morpholino-6-(pyridin-2-yl)furo[3,2-d]pyrimidin-2-yl)amino)-3-(pyridin-4- yl)-1H-pyrazole-1-sulfonamide (35.6 mg, 0.065 mmol) as a yellow solid. LC-MS (ESI+): m/z 548 (MH+).1HNMR (300 MHz, CDCl3) δ 8.70-8.68 (m, 3H), 7.81-7.77 (m, 4H), 7.41-7.39 (m, 2H), 7.36-7.26 (m, 1H), 4.11-4.09 (m, 4H), 3.92-3.91 (m, 4H), 3.09 (s, 6H). 1.5) Synthesis of Compound 28, 4-morpholino-6-(pyridin-2-yl)-N-(3-(pyridin-4-yl)-s1H-pyrazol- 5-yl)furo[3,2-d]pyrimidin-2-aminehydrochloride [0332] To a solution o
Figure imgf000106_0002
f N,N-dimethyl-5-((4-morpholino-6-(pyridin-2-yl)furo[3,2-d]pyrimidin- 2-yl)amino)- 3-(pyridin-4-yl)-1H-pyrazole-1-sulfonamide (35.6 mg, 0.065 mmol) in DCM (4 mL) was added HCl/Et2O (2 mL). The reaction mixture was stirred at room temperature for 2 h. After concentration and slurry in MeOH/Et2O (1/20, 2 mL), 4-morpholino-6-(pyridin-2-yl)-N- (3-(pyridin-4-yl)-1H-pyrazol-5-yl) furo[3,2-d] pyrimidin-2-aminehydrochloride (Compound 28, 28.8 mg, 0.06 mmol) was obtained as a yellow solid. LC-MS (ESI+): m/z 441 (MH+).1HNMR (300 MHz, CD3OD) δ 8.92 (d, J = 6.3 Hz, 2H), 8.75 (d, J = 4.8 Hz, 1H), 8.45 (d, J = 6.6 Hz, 2H), 8.17 (d, J = 7.2 Hz, 1H), 8.12- 8.07 (m, 1H), 7.65 (s, 1H), 7.62-7.58 (m, 1H), 7.07 (s,1H), 4.47-4.13 (m, 4H), 3.97-3.85 (m, 4H). [0333] EXAMPLE 7: Compound 34 using General Synthetic Route 7:
Figure imgf000107_0002
1.1) Synthesis of 2-chloro-6-(1-methyl-1,2,3,6-tetrahydropyridin-4-yl)-4-morpholinofuro[3,2-d] pyrimidine
Figure imgf000107_0001
[0334] A suspension of 2-chloro-6-iodo-4-morpholinofuro[3,2-d]pyrimidine (400 mg, 1.1 mmol), 1-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,2,3,6-tetrahydropyridine (244 mg, 1.1 mmol), K2CO3 (454 mg, 3.29 mmol) and Pd(PPh3)4 (127 mg, 0.011 mmol) in 1,4- dioxane/H2O (8/1, 40 mL) was heated to 50 °C for 2 h under N2. The completion was monitored by TLC. The reaction was diluted with water and extracted with DCM/MeOH (15/1, 3 x 30 mL). The combined organic phase was dried over Na2SO4, filtrated and concentrated under reduced pressure. The residue was purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 3% MeOH/DCM to provide 2-chloro-6-(1-methyl-1,2,3,6- tetrahydropyridin-4-yl)-4-morpholinofuro[3,2-d]pyrimidine (270 mg, 0.81 mmol) as a yellow solid. LC-MS (ESI+): m/z 335/337 (MH+).1HNMR (300 MHz, CDCl3) δ 6.52 (s, 1H), 6.52-6.47 (m, 1H), 4.03-4.00 (m, 4H), 3.86-3.83 (m, 4H), 3.19-3.18 (m, 2H), 2.70-2.66 (m, 2H), 2.58-2.54 (m, 2H), 2.43 (s, 3H). 1.2) Synthesis of N,N-dimethyl-5-((6-(1-methyl-1,2,3,6-tetrahydropyridin-4-yl)-4- morpholinofuro[3,2-d]pyrimidin-2-yl)amino)-3-(pyridin-4-yl)-1H-pyrazole-1-sulfonamide
Figure imgf000108_0001
[0335] A suspension of 2-chloro-6-(1-methyl-1,2,3,6-tetrahydropyridin-4-yl)-4- morpholinofuro [3,2-d]pyrimidine (120 mg, 0.60 mmol), 5-amino-N,N-dimethyl-3-(pyridin-4- yl)-1H-pyrazole-1- sulfonamide (176 mg, 0.18 mmol), KOAc (176 mg, 1.80 mmol), Pd(OAc)2 (14.8 mg, 0.066 mmol) and Xantphos (80 mg, 0.18 mmol) in DMF (20 mL) was heated to 80 °C for 3 h. The reaction mixture was diluted with water and extracted with DCM/MeOH (15/1, 3 x 30mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The residue was purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 10% MeOH/DCM to provide N,N-dimethyl-5-((6-(1-methyl-1,2,3,6-tetrahydropyridin-4-yl)-4-morpholinofuro[3,2- d]pyrimidin-2-yl)amino)-3-(pyridin-4-yl)-1H-pyrazole-1-sulfonamide (120 mg, 0.21 mmol) as a yellow solid. LC-MS (ESI+): m/z 566 (MH+).1HNMR (300 MHz, CDCl3) δ 8.68 (d, J = 6.0 Hz, 2H), 7.77 (d, J = 6.0 Hz, 2H), 7.32 (s, 1H), 6.58 (s, 1H), 6.47-6.42 (m, 1H), 4.03-4.00 (m, 4H), 3.88-3.85 (m, 4H), 3.30-3.29 (m, 2H), 3.07 (s, 6H), 2.83-2.79 (m, 2H), 2.62-2.58 (m, 2H), 2.50 (s, 3H). 1.3) Synthesis of N,N-dimethyl-5-((6-(1-methylpiperidin-4-yl)-4-morpholinofuro[3,2- d]pyrimidin- 2-yl)amino)-3-(pyridin-4-yl)-1H-pyrazole-1-sulfonamide
Figure imgf000108_0002
[0336] To a solution of N,N-dimethyl-5-((6-(1-methyl-1,2,3,6-tetrahydropyridin-4-yl)-4- morpholinofuro [3,2-d]pyrimidin-2-yl)amino)-3-(pyridin-4-yl)-1H-pyrazole-1-sulfonamide (120 mg, 0.21 mmol) in DCM/MeOH (1/1, 10 mL) was added Pd/C under H2 (balloon). The reaction mixture was stirred at room temperature for 3 h. The completion of the reaction was monitored by LC-MS. After filtration, the filtrate was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 10% MeOH/DCM to provide N,N-dimethyl-5- ((6-(1-methylpiperidin-4-yl)-4-morpholinofuro[3,2-d]pyrimidin-2- yl)amino)-3-(pyridin-4-yl)-1H-pyrazole-1-sulfonamide (71 mg, 0.13 mmol) as a white solid. LC-MS (ESI+): m/z 568 (MH+).1HNMR (300 MHz, CD3OD) δ 8.62 (d, J = 6.3 Hz, 2H), 7.87 (d, J = 6.0 Hz, 2H), 7.29 (s, 1H), 6.54 (s, 1H), 4.03-4.00 (m, 4H), 3.86-3.83 (m, 4H), 3.40-3.34 (m, 2H), 3.06 (s, 6H), 2.82-2.79 (m, 2H), 2.67 (s, 3H), 2.30-2.26 (m, 2H), 1.97-1.95 (m, 2H). 1.4) Synthesis of 6-(1-methylpiperidin-4-yl)-4-morpholino-N-(3-(pyridin-4-yl)-1H-pyrazol- 5- yl)furo[3,2-d]pyrimidin-2-amine hydrochloride
Figure imgf000109_0001
[0337] To a solution of N,N-dimethyl-5-((6-(1-methylpiperidin-4-yl)-4-morpholinofuro[3,2-d] pyrimidin-2-yl)amino)-3-(pyridin-4-yl)-1H-pyrazole-1-sulfonamide (71 mg, 0.13 mmol) in DCM (4 mL) was added HCl/Et2O (2 mL). The reaction mixture was stirred at room temperature for 2 h. After concentration and slurry in MeOH/Et2O (1/20, 2 mL), 6-(1- methylpiperidin-4-yl)- 4-morpholino-N-(3-(pyridin-4-yl)-1H-pyrazol-5-yl)furo[3,2-d]pyrimidin- 2-amine hydrochloride (Compound 34, 55 mg, 0.11 mmol) was obtained as a yellow solid. LC- MS (ESI+): m/z 461 (MH+).1HNMR (300 MHz, CD3OD) δ 8.90 (d, J = 6.6 Hz, 2H), 8.42 (d, J = 6.6 Hz, 2H), 7.03 (s, 1H), 6.88 (s, 1H), 4.25-4.10 (m, 4H), 3.96-3.85 (m, 4H), 3.69-3.64 (m, 2H), 3.30-3.17(m, 3H), 2.93 (s, 3H), 2.48-2.39 (m, 2H), 2.16-2.02 (m, 2H).
[0338] EXAMPLE 8: Compound 35 using General Synthetic Route 8:
Figure imgf000110_0002
1.1) Synthesis of tert-butyl 3-(2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)-5,6- dihydropyridine-1(2H)-carboxylate
Figure imgf000110_0001
[0339] A suspension of 2-chloro-6-iodo-4-morpholinofuro[3,2-d]pyrimidine (400 mg, 1.1 mmol), tert-butyl3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-5,6-dihydropyridine-1(2H)- carboxylate (339 mg, 1.1 mmol), K2CO3 (454 mg, 3.29 mmol) and Pd(PPh3)4 (127 mg, 0.011 mmol) in 1,4-dioxane/H2O (8/1, 40 mL) was heated to 90 °C for 3 h under N2. The completion of the reaction was monitored by TLC. The reaction was diluted with water and extracted with EtOAc (3 x 30 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The residue was purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 33% EtOAc/PE to provide tert- butyl 3-(2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)-5,6-dihydropyridine-1(2H)- carboxylate (155 mg, 0.37 mmol) as a yellow solid. LC-MS (ESI+): m/z 421/423 (MH+). 1HNMR (300 MHz, CDCl3) δ 6.68-6.62 (m, 1H), 6.55 (s, 1H), 4.25 (brs, 2H), 4.03-4.00 (m, 4H), 3.86-3.83 (m, 4H), 3.60-3.56 (m, 2H), 2.42-2.38 (m, 2H), 1.50 (s, 9H). 1.2) Synthesis of 2-chloro-4-morpholino-6-(1,2,5,6-tetrahydropyridin-3-yl)furo[3,2- d]pyrimidine H
Figure imgf000111_0001
[0340] To a solution of tert-butyl3-(2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)-5,6- dihydropyridine-1(2H)-carboxylate (600 mg, 1.43 mmol) in DCM (15 mL) was added TFA (3 mL). The reaction mixture was stirred at room temperature for 2 h. The completion of the reaction was monitored by TLC. The reaction was quenched with Na2CO3 and extracted with MeOH/DCM (1/15, 3 x 40 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The residue was slurried in MeOH/Et2O (1/20, 10 mL) to provide 2-chloro-4-morpholino-6-(1,2,5,6-tetrahydropyridin-3- yl)furo[3,2-d]pyrimidine (385 mg, 1.20 mmol) as a white solid. LC-MS (ESI+): m/z 321/323 (MH+).1HNMR (300 MHz, CD3OD) δ 6.94-6.91 (m, 1H), 6.82 (s, 1H), 4.25-4.05 (m, 6H), 3.90- 3.83 (m, 4H), 3.59-3.54 (m, 2H), 2.87-2.70 (m, 2H). 1.3) Synthesis of 2-chloro-6-(1-methyl-1,2,5,6-tetrahydropyridin-3-yl)-4-morpholinofuro[3,2- d]pyrimidine
Figure imgf000111_0002
[0341] A solution of 2-chloro-4-morpholino-6-(1,2,5,6-tetrahydropyridin-3-yl)furo[3,2- d]pyrimidine (385 mg, 1.2 mmol), formaldehyde solution (488 mg, 37%, 6.02 mmol) and CH3COOH (one drop) in DCM was stirred at room temperature for 30 min. To the solution was added sodium triacetoxyborohydride (1.27 g, 6.02 mmol). The reaction mixture was stirred at room temperature for 3 h. The completion of the reaction was monitored by TLC. The reaction mixture was quenched with a saturated NaHCO3 solution and the pH was adjusted to 8. The aqueous solution was extracted with DCM (3 x 40 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 3% MeOH/DCM to provide 2-chloro-6-(1-methyl-1,2,5,6-tetrahydropyridin-3-yl)-4- morpholinofuro[3,2-d]pyrimidine (375 mg, 1.12 mmol) as a brown solid. LC-MS (ESI+): m/z 335/337 (MH+).1HNMR (300 MHz, CDCl3) δ 6.61-6.58 (m, 1H), 6.46 (s, 1H), 4.03-4.00 (m, 4H), 3.86-3.83 (m, 4H), 3.25-3.24 (m, 2H), 2.63-2.59 (m, 2H), 2.48-2.45 (m, 5H). 1.4) Synthesis of N,N-dimethyl-5-((6-(1-methyl-1,2,5,6-tetrahydropyridin-3-yl)-4- morpholinofuro[3,2-d]pyrimidin-2-yl)amino)-3-phenyl-1H-pyrazole-1-sulfonamide
Figure imgf000112_0001
[0342] A suspension of 2-bromo-4-morpholino-6-(pyridin-4-yl)furo[3,2-d]pyrimidine (2 x 20 mg, 0.06 mmol), 5-amino-N,N-dimethyl-3-phenyl-1H-pyrazole-1-sulfonamide (19 mg, 0.07 mmol), Cs2CO3 (49 mg, 0.15 mmol), Pd(OAc)2 (1 mg, 0.006 mmol) and Xantphos (3 mg, 0.006 mmol) in DMF/1,4-dioxane (1/7, 2 mL) was heated to 100 °C for 25 min under microwave conditions. The reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 2% MeOH/DCM to provide N,N-dimethyl-5-((6-(1-methyl-1,2,5,6-tetrahydropyridin-3-yl)-4-morpholinofuro[3,2- d]pyrimidin-2-yl)amino)-3-phenyl-1H-pyrazole-1-sulfonamide (50 mg, 0.089 mmol) as a brown solid. LC-MS (ESI+): m/z 565 (MH+). 1HNMR (300 MHz, CDCl3) δ 8.64 (s, 1H), 7.89 (d, J = 6.6 Hz, 2H), 7.45-7.37 (m, 4H), 6.60-6.57 (m, 1H), 6.50 (s, 1H), 4.03-4.00 (m, 4H), 3.88-3.85 (m, 4H), 3.30-3.27 (m, 2H), 3.05 (s, 6H), 2.62-2.60 (m, 2H), 2.50-2.40 (m, 5H). 1.5) Synthesis of N,N-dimethyl-5-((6-(1-methylpiperidin-3-yl)-4-morpholinofuro [3,2- d]pyrimidin-2-yl)amino)-3-phenyl-1H-pyrazole-1-sulfonamide [0343] To a solutio
Figure imgf000112_0002
n of N,N-dimethyl-5-((6-(1-methyl-1,2,5,6-tetrahydropyridin-3-yl)-4- morpholinofuro [3,2-d]pyrimidin-2-yl)amino)-3-phenyl-1H-pyrazole-1-sulfonamide (50 mg, 0.089 mmol) in DCM/MeOH (1/1, 4 mL) was added Pd/C under H2 (balloon). The reaction mixture was stirred at room temperature for 3 h. The completion of the reaction was monitored by LC-MS. After filtration, the filtrate was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 2% MeOH/DCM to provide N,N-dimethyl-5-((6-(1-methylpiperidin -3-yl)-4-morpholinofuro[3,2-d]pyrimidin-2- yl)amino)-3-phenyl-1H-pyrazole-1-sulfonamide (28 mg, 0.049 mmol) as a yellow solid. LC-MS (ESI+): m/z 567 (MH+).1HNMR (300 MHz, CDCl3) δ 8.63 (s, 1H), 7.89 (dd, J = 8.1, 1.5 Hz, 2H), 7.46-7.37 (m, 3H), 7.24 (s, 1H), 6.44 (s, 1H), 4.01-3.98 (m, 4H), 3.87-3.84 (m, 4H), 3.09- 3.03 (m, 8H), 2.90-2.80 (m, 1H), 2.35 (s, 3H), 2.13-2.02 (m, 3H), 1.82-1.79 (m, 2H), 1.51-1.46 (m, 1H). 1.6) Synthesis of Compound 35, 6-(1-methylpiperidin-3-yl)-4-morpholino-N-(3-phenyl-1H- pyrazol-5-yl)furo[3,2-d]pyrimidin-2-amine hydrochloride
Figure imgf000113_0001
[0344] To a solution of N,N-dimethyl-5-((6-(1-methylpiperidin-3-yl)-4-morpholinofuro[3,2-d] pyrimidin -2-yl)amino)-3-phenyl-1H-pyrazole-1-sulfonamide (28 mg, 0.049 mmol) in DCM (4 mL) was added HCl/Et2O (2 mL). The reaction mixture was stirred at room temperature for 2 h. After concentration and slurry in MeOH/Et2O (1/20, 2 mL), 6-(1-methylpiperidin-3-yl)-4- morpholino-N-(3-phenyl -1H-pyrazol-5-yl)furo[3,2-d]pyrimidin-2-amine hydrochloride (Compound 35, 22 mg, 0.044 mmol) was obtained as a white solid. LC-MS (ESI+): m/z 460 (MH+).1HNMR (300 MHz, DMSO-d6) δ 11.24 (s, 1H), 10.97 (s, 1H), 7.80 (d, J = 7.5 Hz, 2H), 7.52-7.47 (m, 2H), 7.43-7.38 (m, 1H), 6.93 (s, 1H), 6.60 (s, 1H), 4.20-4.08 (m, 4H), 3.90-3.82 (m, 4H), 3.72-3.61 (m, 1H), 3.53-3.39(m, 2H), 3.22-3.14 (m, 1H), 3.06-2.94 (m, 1H), 2.78 (s, 3H), 2.19-2.08(m,1H), 2.03-1.97(m, 2H),1.69-1.59 (m, 1H).
[0345] EXAMPLE 9: Compound 39 using General Synthetic Route 9:
Figure imgf000114_0002
1.1) Synthesis of tert-butyl 3-(2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)-2,5-dihydro- 1H-pyrrole-1-carboxylate
Figure imgf000114_0001
[0346] A suspension of 2-chloro-6-iodo-4-morpholinofuro[3,2-d]pyrimidine (800 mg, 2.2 mmol), tert-butyl 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2,5-dihydro-1H-pyrrole-1- carboxylate (649 mg, 2.2 mmol), K2CO3 (911 mg, 6.6 mmol) and Pd(PPh3)4 (254 mg, 0.022 mmol) in 1,4-dioxane/H2O (2/1, 60 mL) was heated to 90 °C for 3 h under N2. The completion of the reaction was monitored by TLC. The reaction was diluted with water and extracted with EtOAc (3 x 30 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The residue was purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 50% EtOAc/PE to provide tert- butyl 3-(2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)-2,5-dihydro-1H-pyrrole-1- carboxylate (400 mg, 0.99 mmol) as a yellow solid. LC-MS (ESI+): m/z 407/409 (MH+). 1HNMR (300 MHz, CDCl3) δ 6.60 (s, 0.5H), 6.50 (s, 0.5H), 6.40 (s, 0.5H), 6.35 (s, 0.5H), 4.53- 4.35 (m, 4H), 4.05-4.01 (m, 4H), 3.86-3.82 (m, 4H), 1.51 (s, 9H). 1.2) Synthesis of tert-butyl 3-(2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)pyrrolidine-1- carboxylate
Figure imgf000115_0001
[0347] To a solution of tert-butyl 3-(2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)-2,5- dihydro-1H- pyrrole-1-carboxylate (50 mg, 0.089 mmol) in DCM/MeOH (1/1, 20 mL) was added Pd/C under H2 (balloon). The reaction mixture was stirred at room temperature overnight. The completion of the reaction was monitored by LC-MS. After filtration, the filtrate was concentrated directly and purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 33% EtOAc/PE to provide tert-butyl 3-(2-chloro-4-morpholinofuro[3,2- d]pyrimidin-6-yl)pyrrolidine-1- carboxylate (300 mg, 0.74 mmol) as a white solid. LC-MS (ESI+): m/z 409/411 (MH+). 1.3) Synthesis of 2-chloro-4-morpholino-6-(pyrrolidin-3-yl)furo[3,2-d]pyrimidine [0348] To a solution of tert-bu
Figure imgf000115_0002
tyl 3-(2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)-5,6- dihydropyridine-1(2H)-carboxylate (300 mg, 0.74 mmol) in DCM (15 mL) was added TFA (3 mL). The reaction mixture was stirred at room temperature for 2 h. The completion of the reaction was monitored by TLC. The solution was quenched with Na2CO3 and the pH was adjusted to 8. A large amount of solid was precipitated. After filtration, 2-chloro-4-morpholino- 6- (pyrrolidin-3-yl)furo[3,2-d]pyrimidine (200 mg, 0.65 mmol) was obtained as a white solid. LC-MS (ESI+): m/z 309/311 (MH+). 1.4) Synthesis of 2-chloro-6-(1-methylpyrrolidin-3-yl)-4-morpholinofuro[3,2-d]pyrimidine
Figure imgf000115_0003
[0349] A solution of 2-chloro-4-morpholino-6-(pyrrolidin-3-yl)furo[3,2-d]pyrimidine (200 mg, 0.65 mmol), formaldehyde solution (264 mg, 37%, 3.25 mmol) and CH3COOH (one drop) in DCM was stirred at room temperature for 30 min. Then to the reaction was added sodium triacetoxyborohydride (690 mg, 6.02 mmol). The reaction mixture was stirred at room temperature for 3 h. The completion of the reaction was monitored by TLC. The reaction mixture was quenched with a saturated NaHCO3 solution and the pH was adjusted to 8. The aqueous solution was extracted with DCM (3 x 40 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide 2-chloro-6-(1-methylpyrrolidin-3-yl)-4-morpholinofuro[3,2- d]pyrimidine (180 mg, 0.56 mmol) as a white solid. LC-MS (ESI+): m/z 323/325 (MH+).1HNMR (300 MHz, CDCl3) δ6.80 (s, 1H), 3.92-3.89 (m, 4H), 3.75-3.74 (m, 4H), 3.69- 3.66 (m, 1H), 3.12-3.05 (m, 1H), 2.90-2.81 (m, 3H), 2.50 (s, 3H), 2.37-2.30 (m, 1H), 2.09-2.01 (m, 1H). 1.5) Synthesis of N,N-dimethyl-5-((6-(1-methylpyrrolidin-3-yl)-4-morpholinofuro[3,2- d]pyrimidin -2-yl)amino)-3-(m-tolyl)-1H-pyrazole-1-sulfonamide
Figure imgf000116_0001
[0350] A suspension of 2-chloro-6-(1-methylpyrrolidin-3-yl)-4-morpholinofuro[3,2- d]pyrimidine (40 mg, 0.12 mmol), 5-amino-N,N-dimethyl-3-(m-tolyl)-1H-pyrazole-1- sulfonamide (45 mg, 0.16 mmol), Cs2CO3 (100 mg, 0.31 mmol), Pd(OAc)2 (3 mg, 0.012 mmol) and Xantphos (7 mg, 0.012 mmol) in DMF/1,4-dioxane (1/7, 5 mL) was heated to 90 °C for 30 min under microwave condition. The reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide N,N-dimethyl-5-((6-(1-methylpyrrolidin-3-yl)-4-morpholinofuro[3,2- d]pyrimidin-2-yl)amino)-3-(m-tolyl)-1H-pyrazole-1-sulfonamide (60 mg, 0.11 mmol). LC-MS (ESI+): m/z 567 (MH+).1HNMR (300 MHz, CD3OD) δ7.67-7.62 (m, 2H), 7.35-7.30 (m, 1H), 7.23-7.21 (m, 1H), 7.16 (s, 1H), 6.56 (s, 1H), 4.04-4.01 (m, 4H), 3.87-3.84 (m, 4H), 3.69-3.54 (m, 1H), 3.15-3.05 (m, 2H), 2.90 (s, 6H), 2.85-2.75 (m, 2H), 2.46 (s, 3H), 2.41 (s, 3H), 2.37- 2.30 (m, 1H), 2.19-2.10 (m, 1H). 1.6) Synthesis of Compound 39, 6-(1-methylpyrrolidin-3-yl)-4-morpholino-N-(3-(m-tolyl)-1H- pyrazol-5-yl)furo[3,2-d]pyrimidin-2-amine hydrochloride [0351] To a solut
Figure imgf000117_0001
ion of N,N-dimethyl-5-((6-(1-methylpyrrolidin-3-yl)-4-morpholinofuro[3,2- d]pyrimidin -2-yl)amino)-3-(m-tolyl)-1H-pyrazole-1-sulfonamide (60 mg, 0.11 mmol) in DCM (4 mL) was added HCl/Et2O (2 mL). The reaction mixture was stirred at room temperature for 2 h. After concentration, the crude product was purified by preparative HPLC to provide 6-(1- methylpyrrolidin -3-yl)-4-morpholino- N-(3-(m-tolyl)-1H-pyrazol-5-yl)furo[3,2-d]pyrimidin-2- amine hydrochloride (Compound 39, 9 mg, 0.018 mmol) as a yellow solid. LC-MS (ESI+): m/z 460 (MH+).1HNMR (300 MHz, D2O) δ 7.17-7.04 (m, 4H), 6.64 (d, J = 3.0 Hz, 1H), 6.03 (s, 1H), 6.41 (s, 1H), 3.99-3.94 (m, 1H), 3.92-3.76 (m, 8H), 3.75-3.68 (m, 1H), 3.66-3.42 (m, 1H), 3.28-3.09 (m, 2H), 2.92 (d, J = 5.1 Hz, 3H), 2.64-2.44 (m, 1H), 2.32-2.22 (m, 1H), 2.17 (s, 3H). [0352] EXAMPLE 10: Compound 42 using General Synthetic Route 10: General procedure 10:
Figure imgf000117_0002
1.1) Synthesis of tert-butyl 4-(m-tolyl)-1H-pyrazole-1-carboxylate [0353] A solution of tert-butyl
Figure imgf000117_0003
4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole - 1-carboxylate (1.5 g, 5.1 mmol) 1-bromo-3-methylbenzene (872 mg, 5.1 mmol), Cs2CO3 (91 mg, 0.28 mmol), PdCl2(PPh3)2 (590 mg, 0.051 mmol) and CsF (1.16 g, 7.65 mmol) in 1,4- dioxane/H2O (2/1, 30 mL) was heated to 80 °C overnight under N2. The completion of the reaction was monitored by TLC. The reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide tert-butyl 4-(m-tolyl)-1H-pyrazole-1-carboxylate (300 mg, 1.16 mmol) as a yellow solid. LC-MS (ESI+): m/z 259 (MH+).1HNMR (300 MHz, CDCl3) δ 8.29 (s, 1H), 7.99 (s, 1H), 7.34-7.28 (m, 3H), 7.13-7.10 (m, 1H), 2.39 (s, 3H), 1.68 (s, 9H). Synthesis of 4-(m-tolyl)-1H-pyrazole hydrochloride
Figure imgf000118_0001
[0354] To a solution of tert-butyl 4-(m-tolyl)-1H-pyrazole-1-carboxylate (300 mg, 1.16 mmol) in DCM (8 mL) was added HCl/Et2O (3 mL). The reaction mixture was stirred at room temperature for 2 h. A large amount of solid was precipitated. After concentration and slurry in MeOH/Et2O (1/20, 2 mL), 4-(m-tolyl)-1H-pyrazole hydrochloride (153 mg, 0.79 mmol) was obtained as a yellow solid. LC-MS (ESI+): m/z 159 (MH+).1HNMR (300 MHz, CDCl3) δ 8.13 (s, 2H), 7.38-7.20 (m, 4H), 2.42 (s, 3H). 1.2) Synthesis of Compound 42, 4-morpholino-6-(pyridin-4-yl)-2-(4-(m-tolyl)-1H-pyrazol -1- yl)furo[3,2-d] pyrimidine
Figure imgf000118_0002
[0355] A suspension of 2-bromo-4-morpholino-6-(pyridin-4-yl)furo[3,2-d]pyrimidine (60 mg, 0.17 mmol) and 4-(m-tolyl)-1H-pyrazole hydrochloride (40 mg, 0.20 mmol ) in anhydrous THF (10 mL) was added NaH (12 mg, 0.49 mmol). The reaction mixture was stirred at 70 °C overnight. The completion was monitored by TLC. The reaction was quenched with water (10 mL) and the aqueous solution was extracted with DCM/MeOH (10/1, 2 x 20mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The residue was purified by preparative TLC purification to provide 4-morpholino-6- (pyridin-4-yl)-2-(4-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d]pyrimidine (Compound 42, 31.2 mg, 0.049 mmol) as a yellow solid. LC-MS (ESI+): m/z 439 (MH+).1HNMR (300 MHz, CDCl3) δ 8.80-8.78 (m, 2H), 8.71 (s, 1H), 8.08 (s, 1H), 7.68 (d, J = 5.7 Hz, 2H), 7.42 (d, J = 7.5 Hz, 2H), 7.34-7.31 (m, 2H), 7.11 (d, J = 3.9 Hz, 1H), 4.22-4.16 (m, 4H), 3.98-3.91 (m, 4H), 2.41 (s, 3H). [0356] EXAMPLE 11: Compound 43 using General Synthetic Route 11:
Figure imgf000119_0001
1.1) Synthesis of 5-phenylisoxazol-3-amine
Figure imgf000119_0002
[0357] A solution of 3-oxo-3-phenylpropanenitrile (1.5 g, 10.3 mmol) in EtOH/H2O (1/1, 20 mL) was added hydroxylamine hydrochloride (785 mg, 11.3 mmol) and sodium hydroxide (450 mg, 11.3 mmol). The reaction mixture was heated to 80 °C overnight. To the above solution was added conc. HCl aq. (1.3 mL). The resulting mixture was stirred at 80 °C for 2 h. The completion of the reaction was monitored by TLC. The reaction mixture was quenched with a saturated NaHCO3 solution and the pH was adjusted to 10. The aqueous solution was extracted with EtOAc (3 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The residue was purified by silica gel column chromatography with a gradient elution of 15% EtOAc/hex to 35% EtOAc/hex to provide 5- phenylisoxazol-3-amine (0.68 g, 1.25 mmol) as a yellow solid. LC-MS (ESI+): m/z 161 (MH+). 1HNMR (300 MHz, CDCl3) δ 7.72-7.68 (m, 2H), 7.47-7.35 (m, 3H), 6.08 (s, 1H), 4.08 (brs, 2H). 1.2) Synthesis of Compound 43, 4-morpholino-N-(5-phenylisoxazol-3-yl)-6-(pyridin-4-yl) furo[3,2-d]pyrimidin-2-amine
Figure imgf000120_0001
[0358] A suspension of 2-bromo-4-morpholino-6-(pyridin-4-yl)furo[3,2-d]pyrimidine (50 mg, 0.14 mmol), 5-phenylisoxazol-3-amine (35 mg, 0.21 mmol), Cs2CO3 (91 mg, 0.28 mmol), PdCl2(PPh3)2 (10 mg, 0.014 mmol) and Xantphos (24 mg, 0.042 mmol) in 1,4-dioxane (4 mL) was heated to 90 °C for 30 min under microwave condition. The reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide 4-morpholino-N-(5-phenylisoxazol-3-yl)-6- (pyridin-4-yl)furo[3,2-d]pyrimidin-2-amine (Compound 43, 15 mg, 0.034 mmol) as a white solid. LC-MS (ESI+): m/z 441 (MH+). 1HNMR (300 MHz, DMSO-d6) δ 9.93 (s, 1H), 8.74 (d, J = 6.0 Hz, 2H), 7.93 (d, J = 6.0 Hz, 2H), 7.83-7.81 (m, 2H), 7.69 (s, 1H), 7.56-7.53 (m, 3H), 7.40 (s, 1H), 4.08-4.02 (m, 4H), 3.85-3.79 (m, 4H). [0359] EXAMPLE 12: Compound 44 using General Synthetic Route 12:
Figure imgf000120_0003
1.1) Synthesis of Compound 44, 4-morpholino-N-(3-phenylisoxazol-5-yl)-6-(pyridin-4-yl) furo[3,2-d]pyrimidin-2-amine H
Figure imgf000120_0002
[0360] A suspension of 2-bromo-4-morpholino-6-(pyridin-4-yl)furo[3,2-d]pyrimidine (76 mg, 0.21 mmol), 3-phenylisoxazol-5-amine (40 mg, 0.25 mmol), Cs2CO3 (158 mg, 0.48 mmol), Pd(OAc)2 (5 mg, 0.021 mmol) and Xantphos (12 mg, 0.021 mmol) in DMF/1,4-dioxane (1/7, 8 mL) was heated to 80 °C for 25 min under microwave condition. The reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide 4-morpholino-N-(3-phenylisoxazol-5-yl)-6- (pyridin-4-yl)furo[3,2-d]pyrimidin-2-amine (Compound 44, 61 mg, 0.14 mmol) as a yellow solid. LC-MS (ESI+): m/z 441 (MH+). 1HNMR (300 MHz, DMSO-d6) δ 10.86 (s, 1H), 8.75 (d, J = 4.5 Hz, 2H), 7.93 (d, J = 5.7 Hz, 2H), 7.84-7.81 (m, 2H), 7.75 (s, 1H), 7.53-7.51 (m, 3H), 6.70 (s, 1H), 4.08-4.03 (m, 4H), 3.87-3.82 (m, 4H). [0361] EXAMPLE 13: Compound 50 using General Synthetic Route 13:
Figure imgf000121_0002
1.1) Synthesis of 3-amino-N,N-dimethyl-1H-indazole-1-sulfonamide
Figure imgf000121_0001
[0362] To a solution of 1H-indazol-3-amine (1 g, 7.5 mmol) in THF (30 mL) at 0 °C was added NaH (541 mg, 13.53 mmol). After stirred at 0 °C for 1 h, to the solution was added dimethylsulfamoyl chloride (1.61 g, 11.28 mmol). The completion of the reaction was monitored by TLC. The reaction mixture was quenched with a saturated NH4Cl solution. The aqueous solution was extracted with DCM/MeOH (15/1, 3 x 50 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 20% EtOAc/PE to 50% EtOAc/PE to provide 3-amino-N,N-dimethyl-1H-indazole -1- sulfonamide (600 mg, 2.5 mmol) as a yellow solid. LC-MS: m/z 241 (MH+).1HNMR (300 MHz, CDCl3) δ 7.97 (d, J = 8.4 Hz, 1H), 7.55-7.48 (m, 2H), 7.29-7.24 (m, 1H), 4.46 (brs, 2H), 2.92 (s, 6H). 1.2) Synthesis of N,N-dimethyl-3-((4-morpholino-6-(pyridin-4-yl)furo[3,2-d]pyrimidin-2-yl) amino)-1H-indazole-1-sulfonamide
Figure imgf000122_0001
[0363] A suspension of 2-bromo-4-morpholino-6-(pyridin-4-yl)furo[3,2-d]pyrimidine (60 mg, 0.17 mmol), 3-amino-N,N-dimethyl-1H-indazole-1-sulfonamide (48 mg, 0.20 mmol), Cs2CO3 (125 mg, 0.38 mmol), Pd(OAc)2 (4 mg, 0.017 mmol) and Xantphos (10 mg, 0.017 mmol) in DMF/1,4-dioxane (1/7, 4 mL) was heated to 90 °C for 25 min under microwave condition. The reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide N,N-dimethyl-3-((4- morpholino-6-(pyridin-4-yl)furo[3,2-d]pyrimidin-2-yl)amino)-1H-indazole-1-sulfonamide (50 mg, 0.096 mmol) as a white solid. LC-MS (ESI+): m/z 521 (MH+).1HNMR (300 MHz, CDCl3) δ 8.74 (d, J = 6.0 Hz, 2H), 8.04 (d, J = 8.7 Hz, 1H), 7.9 (d, J = 8.4 Hz, 1H), 7.65 (d, J = 6.0 Hz, 2H), 7.60-7.48 (m, 2H), 7.16 (s, 1H), 4.02-3.97 (m,4H), 3.83-3.80 (m, 4H), 2.99 (s, 6H). 1.3) Synthesis of Compound 50, N-(1H-indazol-3-yl)-4-morpholino-6-(pyridin-4-yl)furo[3,2-d] pyrimidin-2-amine hydrochloride
Figure imgf000122_0002
[0364] To a solution of N,N-dimethyl-3-((4-morpholino-6-(pyridin-4-yl)furo[3,2-d]pyrimidin- 2-yl)amino) -1H-indazole-1-sulfonamide (50 mg, 0.096 mmol) in DCM (4 mL) was added HCl/Et2O (2 mL). The reaction mixture was stirred at room temperature for 2 h. After concentration and slurry in MeOH/Et2O (1/20, 2 mL), N-(1H-indazol-3-yl)-4-morpholino-6- (pyridin-4-yl)furo[3,2-d]pyrimidin-2-amine hydrochloride (Compound 50, 33.8 mg, 0.06 mmol) was obtained as a yellow solid. LC-MS (ESI+): m/z 414 (MH+).1HNMR (300 MHz, CD3OD) δ 8.94 (d, J = 6.6 Hz, 2H), 8.48 (d, J = 5.7 Hz, 2H), 7.99 (d, J = 9.0 Hz, 2H), 7.58-7.48 (m, 2H), 7.23 (t, J = 7.5 Hz, 1H), 4.44-4.15 (m, 4H), 3.96-3.87 (m, 4H). [0365] EXAMPLE 14: Compound 52 using General Synthetic Route 14:
Figure imgf000123_0001
1.1) Synthesis of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid
Figure imgf000123_0002
[0366] To a solution of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine (2.4 g, 10 mmol) in anhydrous THF (4 mL) at -78 °C under N2 was added n-BuLi ( 5.2 mL, 2.5 M, 13 mmol) dropwise. The reaction mixture was stirred at that temperature for 1 h. To the solution was added dry ice (4.4 g, 100 mmol) in one portion. The resulting reaction mixture was stirred at that temperature for 3 h. The completion of the reaction was monitored by TLC. The reaction was quenched with water and the pH was adjusted to 5. The aqueous solution was extracted with DCM (3 x 80 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was slurry in Et2O to provide 2- chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid ( 2.92 g, 10.3 mmol) as a yellow solid. LC-MS (ESI+): m/z 284/286 (MH+).1HNMR (300 MHz, CDCl3) δ 7.58 (s, 1H), 4.04-3.95 (m, 4H), 3.81-3.76 (m, 4H). 1.2) Synthesis of 2-chloro-N-(2,4-dimethoxybenzyl)-4-morpholinofuro[3,2-d]pyrimidine -6- carboxamide
Figure imgf000124_0001
[0367] A solution of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid (112 mg, 0.41 mmol), (2,4-dimethoxyphenyl)methanamine (68 mg, 0.41 mmol), HBOT (137 mg, 1.02 mmol) and EDCl (195 mg, 1.02 mmol) in DMF was stirred at room temperature overnight. The completion of the reaction was monitored by TLC. The solution was diluted with water (10 mL) and extracted with DCM/MeOH (10/1, 3 x 30mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure to provide crude 2-chloro- N- (2,4-dimethoxybenzyl)-4-morpholinofuro[3,2-d]pyrimidine-6-carboxamide (140 mg, 0.32 mmol) as a yellow oil. LC-MS (ESI+): m/z 433/435 (MH+). 1.3) Synthesis of N-(2,4-dimethoxybenzyl)-2-((1-(N,N-dimethylsulfamoyl)-3-phenyl-1H-pyrazol - 5-yl)amino)-4-morpholinofuro[3,2-d]pyrimidine-6-carboxamide [0368] A suspens
Figure imgf000124_0002
ion of 2-chloro-N-(2,4-dimethoxybenzyl)-4-morpholinofuro[3,2- d]pyrimidine-6- carboxamide (2 x 50 mg, 0.12 mmol), 5-amino-N,N-dimethyl-3-phenyl-1H- pyrazole-1- sulfonamide (37 mg, 0.14 mmol), Cs2CO3 (90 mg, 0.27 mmol), Pd(OAc)2 (3 mg, 0.012 mmol) and Xantphos (6.5 mg, 0.012 mmol) in DMF/1,4-dioxane (1/7, 4 mL) was heated to 90 °C for 30 min under microwave conditions. The reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide N-(2,4-dimethoxybenzyl)-2-((1-(N,N-dimethylsulfamoyl)-3- phenyl-1H- pyrazol-5-yl)amino)-4-morpholinofuro[3,2-d]pyrimidine-6-carboxamide (43 mg, 0.065 mmol) as a yellow solid. LC-MS (ESI+): m/z 663 (MH+). 1.4) Synthesis of Compound 52, 4-morpholino-2-((3-phenyl-1H-pyrazol-5-yl)amino)furo[3,2-d] pyrimidine-6-carboxamide
Figure imgf000125_0001
[0369] To a solution of N-(2,4-dimethoxybenzyl)-2-((1-(N,N-dimethylsulfamoyl)-3-phenyl- 1H- pyrazol-5-yl)amino)-4-morpholinofuro[3,2-d]pyrimidine-6-carboxamide (43 mg, 0.065 mmol) in DCM (4 mL) was added TFA (2 mL). The reaction mixture was stirred at room temperature overnight. The completion was monitored by LC-MS. The reaction mixture was quenched with a saturated NaHCO3 solution and the pH was adjusted to 10. The aqueous solution was extracted with DCM/MeOH (15/1, 3 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated. After concentration and slurry in MeOH, the impure azetidin-1-yl(4-morpholino-2-((3-phenyl-1H-pyrazol-5-yl)amino)furo[3,2- d]pyrimidin-6-yl)methanone (Compound 52, 21 mg, 0.048 mmol) was obtained as a yellow solid. LC-MS (ESI+): m/z 406 (MH+). 1.5) Synthesis of Compound 52, 4-morpholino-2-((3-phenyl-1H-pyrazol-5-yl)amino)furo[3,2-d] pyrimidine-6-carboxamide hydrochloride
Figure imgf000125_0002
[0370] To a solution of impure azetidin-1-yl(4-morpholino-2-((3-phenyl-1H-pyrazol-5- yl)amino)furo [3,2-d]pyrimidin-6-yl)methanone (21 mg, 0.048 mmol) in DCM (4 mL) was added HCl/Et2O (2 mL). The reaction mixture was stirred at room temperature for 2 h. After concentration and slurry in MeOH/Et2O (1/20, 2 mL), 4-morpholino-2-((3-phenyl-1H-pyrazol-5- yl)amino)furo[3,2-d]pyrimidine-6- carboxamide hydrochloride (Compound 52, 17.1 mg, 0.039 mmol) was obtained as a yellow solid. LC-MS (ESI+): m/z 406 (MH+).1HNMR (300 MHz, CD3OD) δ 7.74 (d, J = 9.9 Hz, 2H), 7.70 (s, 1H), 7.60-7.42 (m, 3H), 6.42 (s, 1H), 4.48-4.14 (m, 4H), 3.91-3.85 (m, 4H). [0371] EXAMPLE 15: Compound 53 using General Synthetic Route 15:
Figure imgf000126_0001
1.1) Synthesis of azetidin-1-yl(2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)methanone
Figure imgf000126_0002
[0372] A solution of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid (200 mg, 0.71 mmol) in DCM was added oxalyl dichloride (182 mg, 1.4 mmol) and one drop of DMF. The mixture was stirred at room temperature for 6 h. The solution was concentrated, and the resulting residue was dissolved in DCM (15 mL). To the solution was added azetidine (60 mg, 1.06 mmol) and followed by triethylamine (107 mg, 1.06 mmol). The completion of the reaction was monitored by TLC. The reaction mixture was quenched with water and extracted with DCM (3 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The residue was purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide azetidin-1-yl(2-chloro- 4-morpholinofuro [3,2-d]pyrimidin-6-yl)methanone (75.8 mg, 0.25 mmol) as a yellow solid. LC-MS (ESI+): m/z 323/325 (MH+). 1HNMR (300 MHz, CDCl3) δ 7.19 (s, 1H), 4.58-4.52 (m, 2H), 4.35-4.27 (m, 2H), 4.07-4.04 (m, 4H), 3.86-3.83 (m, 4H), 2.52- 2.45 (m, 2H). 1.2) Synthesis of 5-((6-(azetidine-1-carbonyl)-4-morpholinofuro[3,2-d]pyrimidin-2-yl)amino) - N,N-dimethyl-3-phenyl-1H-pyrazole-1-sulfonamide
Figure imgf000126_0003
[0373] A suspension of azetidin-1-yl(2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6- yl)methanone (3 x 50 mg, 0.47 mmol), 5-amino-N,N-dimethyl-3-phenyl-1H-pyrazole-1- sulfonamide (148 mg, 0.56 mmol), Cs2CO3 (344 mg, 1.06 mmol), Pd(OAc)2 (10 mg, 0.047 mmol) and Xantphos (25 mg, 0.047 mmol) in DMF/1,4-dioxane (7/1, 4 mL) was heated to 50 °C for 50 min under microwave condition. The reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide 5-((6-(azetidine-1-carbonyl)-4-morpholinofuro[3,2-d]pyrimidin-2- yl)amino)-N,N-dimethyl-3-phenyl-1H-pyrazole-1-sulfonamide (143 mg, 0.26 mmol) as a yellow solid. LC-MS (ESI+): m/z 553 (MH+). 1HNMR (300 MHz, CDCl3) δ 8.70 (s, 1H), 7.91 (d, J = 6.9 Hz, 2H), 7.46-7.36 (m, 4H), 7.22 (s, 1H), 4.07-4.04 (m, 4H), 3.87-3.76 (m, 6H), 3.73-3.67 (m, 2H), 3.06 (s, 6H), 2.11-1.96 (m, 2H). 1.3) Synthesis of Compound 53, azetidin-1-yl(4-morpholino-2-((3-phenyl-1H-pyrazol-5- yl)amino) furo[3,2-d]pyrimidin-6-yl)methanone hydrochloride
Figure imgf000127_0001
[0374] To a solution of 5-((6-(azetidine-1-carbonyl)-4-morpholinofuro[3,2-d]pyrimidin-2- yl)amino)- N,N-dimethyl-3-phenyl-1H-pyrazole-1-sulfonamide (65 mg, 0.11 mmol) in DCM (4 mL) was added HCl/Et2O (2 mL). The reaction mixture was stirred at room temperature for 2 h. After concentration and slurry in MeOH/Et2O (1/20, 2 mL), azetidin-1-yl(4-morpholino-2-((3- phenyl-1H-pyrazol-5-yl) amino)furo[3,2-d] pyrimidin-6-yl)methanone hydrochloride (Compound 53, 30.1 mg, 0.06 mmol) was obtained as a yellow solid. LC-MS (ESI+): m/z 446 (MH+).1HNMR (300 MHz, CD3OD) δ 7.71 (d, J = 7.2 Hz, 2H), 7.64 -7.40 (m, 4H), 6.42 (s, 1H), 4.87-4.65 (m, 2H), 4.38-4.13 (m, 6H), 3.91-3.83 (m, 4H), 2.54-2.43 (m, 2H).
[0375] EXAMPLE 16: Compound 55 using General Synthetic Route 16:
Figure imgf000128_0001
1.1) Synthesis of (2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)(piperidin-1-yl)methanone
Figure imgf000128_0003
[0376] A solution of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid (200 mg, 0.71 mmol), piperidine (61 mg, 0.71 mmol), HOBT (245 mg, 1.76 mmol), EDCl (340 mg, 1.76 mmol) in DMF (12 mL) was stirred at room temperature overnight. The completion of the reaction was monitored by TLC. The reaction mixture was diluted with water and extracted with DCM/MeOH (15/1, 3 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The residue was purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide (2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)(piperidin-1-yl)methanone (171 mg, 0.49 mmol) as a white solid. LC-MS (ESI+): m/z 351/353 (MH+).1HNMR (300 MHz, CDCl3) δ 6.99 (s, 1H), 4.08-4.03 (m, 4H), 3.85-3.79 (m, 4H), 3.75-3.58 (m, 4H), 1.78-1.66 (m, 6H). 1.2) Synthesis of N,N-dimethyl-5-((4-morpholino-6-(piperidine-1-carbonyl)furo[3,2- d]pyrimidin- 2-yl)amino)-3-phenyl-1H-pyrazole-1-sulfonamide
Figure imgf000128_0002
[0377] A suspension of (2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)(piperidin-1- yl)methanone (100 mg, 0.29 mmol), 5-amino-N,N-dimethyl-3-phenyl-1H-pyrazole-1- sulfonamide (91 mg, 0.34mmol), Cs2CO3 (214 mg, 0.66 mmol), Pd(OAc)2 (6.4 mg, 0.029 mmol) and Xantphos (16.5 mg, 0.029 mmol) in DMF/1,4-dioxane (7/1, 4 mL) was heated to 90 °C for 45 min under microwave condition. The reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide N,N-dimethyl-5-((4-morpholino-6-(piperidine-1-carbonyl)furo[3,2- d]pyrimidin-2-yl) amino)-3-phenyl-1H-pyrazole-1-sulfonamide (53 mg, 0.09 mmol) as a colorless oil. LC-MS (ESI+): m/z 581 (MH+).1HNMR (300 MHz, CDCl3) δ 8.70 (s, 1H), 7.90 (d, J = 6.9 Hz, 2H), 7.45-7.34 (m, 4H), 7.02 (s, 1H), 4.04-4.02 (m, 4H), 3.87-3.85 (m, 4H), 3.79- 3.70 (m, 4H), 3.06 (s, 6H), 1.76-1.68 (m, 6H). 1.3) Synthesis of Compound 55, (4-morpholino-2-((3-phenyl-1H-pyrazol-5-yl)amino)furo[3,2-d] pyrimidin-6-yl)(piperidin-1-yl)methanone hydrochloride [0378] To a solution of
Figure imgf000129_0001
N,N-dimethyl-5-((4-morpholino-6-(piperidine-1-carbonyl)furo[3,2-d] pyrimidin-2-yl)amino)-3-phenyl-1H-pyrazole-1-sulfonamide (53 mg, 0.09 mmol) in DCM (4 mL) was added HCl/Et2O (2 mL). The reaction mixture was stirred at room temperature for 2 h. After concentration and slurry in MeOH/Et2O (1/20, 2 mL), (4-morpholino-2-((3-phenyl-1H- pyrazol-5-yl)amino)furo [3,2-d]pyrimidin-6-yl)(piperidin-1-yl)methanone hydrochloride (Compound 55, 17.6 mg, 0.35 mmol) was obtained as a white solid. LC-MS (ESI+): m/z 474 (MH+).1HNMR (300 MHz, DMSO-d6) δ 7.78 (d, J = 7.5 Hz, 2H), 7.59-7.45 (m, 2H), 7.40-7.36 (m, 1H), 7.29 (s, 1H), 6.61 (s, 1H), 4.19-4.03 (m, 4H), 3.88-3.79 (m, 4H), 3.59-3.47 (m, 4H), 1.76-1.47 (m, 6H). [0379] EXAMPLE 17: Compound 64 using General Synthetic Route 17:
Figure imgf000130_0002
1.1) Synthesis of methyl 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylate
Figure imgf000130_0001
[0380] A solution of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid (1.0 g, 3.5 mmol) in DCM was added oxalyl dichloride (912.0 mg, 7.0 mmol) and one drop of DMF. The mixture was stirred at room temperature for 6 h. The solution was concentrated directly and the residue was dissolved in DCM (15 mL). To the solution was added methanol (80 mL) and followed by triethylamine (1.07 g, 10.5 mmol). The completion of the reaction was monitored by TLC. The reaction was quenched with water (10 ml) and a large amount of yellow solid was precipitated. After filtration, 800 mg of methyl 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6- carboxylate was obtained. LC-MS (ESI+): m/z 298/300 (MH+).1HNMR (300 MHz, CDCl3) δ 7.40 (s, 1H), 4.09-4.01 (m, 4H), 3.99 (s, 3H), 3.87-3.84 (m, 4H). 1.2) Synthesis of methyl 2-((1-(N,N-dimethylsulfamoyl)-3-phenyl-1H-pyrazol-5-yl)amino)-4- morpholinofuro[3,2-d]pyrimidine-6-carboxylate
Figure imgf000131_0001
[0381] A suspension of methyl 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylate (400 mg, 1.35 mmol), 5-amino-N,N-dimethyl-3-phenyl-1H-pyrazole-1-sulfonamide (430 mg, 1.620 mmol), Cs2CO3 (1.1 g, 3.38 mmol), Pd(OAc)2 (30 mg, 0.135 mmol) and Xantphos (80 mg, 0.135 mmol) in DMF/1,4-dioxane (1/7, 40 mL) was heated to 80 °C for 2 h. The reaction mixture was diluted with water and extracted with DCM (3 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The residue was purified by silica gel column chromatography with a gradient elution of 50% EtOAc/PE to DCM to provide 2-((1-(N,N-dimethylsulfamoyl)-3-phenyl-1H-pyrazol-5- yl)amino) -4-morpholinofuro[3,2-d]pyrimidine-6-carboxylate (250 mg, 0.47 mmol) as a yellow solid. LC-MS (ESI+): m/z 528 (MH+). 1.3) Synthesis of 2-((1-(N,N-dimethylsulfamoyl)-3-phenyl-1H-pyrazol-5-yl)amino)-4- morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid
Figure imgf000131_0002
[0382] To a solution of 2-((1-(N,N-dimethylsulfamoyl)-3-phenyl-1H-pyrazol-5-yl)amino)-4- morpholinofuro[3,2-d]pyrimidine-6-carboxylate (250 mg, 0.47 mmol) in MeOH/DCM (1/2, 30 mL) was added NaOH aqueous solution (2M, 0.3 mL). The completion of the reaction was monitored by TLC. The reaction mixture was quenched with HCl aqueous solution and adjusted the pH to 5. The aqueous phase was extracted with DCM (3 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The residue was purified by slurry using Et2O to provide 2-((1-(N,N- dimethylsulfamoyl)-3-phenyl- 1H-pyrazol-5-yl)amino)-4-morpholinofuro[3,2-d]pyrimidine-6- carboxylic acid (200 mg, 0.39 mmol) as a yellow solid. LC-MS (ESI+): m/z 514 (MH+). 1.4) Synthesis of (R)-N-(1-cyclopropylethyl)-2-((1-(N,N-dimethylsulfamoyl)-3-phenyl-1H- pyrazol-5-yl)amino)-4-morpholinofuro[3,2-d]pyrimidine-6-carboxamide
Figure imgf000132_0001
[0383] A solution of 2-((1-(N,N-dimethylsulfamoyl)-3-phenyl-1H-pyrazol-5-yl)amino)-4- morpholinofuro [3,2-d]pyrimidine-6-carboxylic acid (150 mg, 0.29 mmol), (R)-1- cyclopropylethanamine (30 mg, 0.35 mmol), HOBT (99 mg, 0.73 mmol) and EDCl (140 mg, 0.73 mmol) in DMF (3 mL) was stirred at room temperature overnight. The completion of the reaction was monitored by TLC. The reaction was quenched with water (10 ml) and extracted with DCM/MeOH (15/1, 3 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The residue was purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide (R)-N-(1-cyclopropylethyl)-2- ((1-(N,N-dimethylsulfamoyl)-5-phenyl-1H-pyrazol-3- yl)amino)-4-morpholinofuro[3,2-d]pyrimidine-6-carboxamide (80 mg, 0.14 mmol). LC-MS (ESI+): m/z 581 (MH+).1HNMR (300 MHz, CDCl3) δ 8.72 (s, 1H), 7.90 (d, J = 6.6 Hz, 2H), 7.47-7.39 (m, 3H), 7.35 (s, 1H), 6.24 (d, J = 8.1 Hz, 1H), 4.10-4.04 (m, 4H), 3.89-3.84 (m, 4H), 3.59-3.56 (m, 1H), 3.07 (s, 6H), 1.36 (d, J = 6.6 Hz, 3H), 0.97-0.95 (m, 1H), 0.60-0.52 (m, 2H), 0.47-0.32 (m, 2H). 1.5) Synthesis of Compound 64, (R)-N-(1-cyclopropylethyl)-4-morpholino-2-((5-phenyl- 1H- pyrazol-3-yl)amino)furo[3,2-d]pyrimidine-6-carboxamide hydrochloride [0384] To a solut
Figure imgf000132_0002
ion of (R)-N-(1-cyclopropylethyl)-2-((1-(N,N-dimethylsulfamoyl)-5-phenyl- 1H- pyrazol-3-yl)amino)-4-morpholinofuro[3,2-d]pyrimidine-6-carboxamide (80 mg, 0.14 mmol) in DCM (4 mL) was added HCl/Et2O (2 mL). The reaction mixture was stirred at room temperature for 2 h. After concentration and slurry in MeOH/Et2O (1/20, 2 mL), (R)-N-(1- cyclopropylethyl)-4-morpholino -2-((5-phenyl-1H-pyrazol-3-yl)amino)furo[3,2-d]pyrimidine-6- carboxamide hydrochloride (Compound 64, 30 mg, 0.06 mmol) was obtained as a white solid. LC-MS (ESI+): m/z 474 (MH+).1HNMR (300 MHz, DMSO-d6) δ 10.79 (s, 1H), 8.93 (d, J = 8.1 Hz, 1H), 7.80 (d, J = 7.5 Hz, 2H), 7.59 (s, 1H), 7.54-7.42 (m, 2H), 7.39-7.37 (m, 1H), 6.61(s, 1H), 4.16-4.05 (m, 4H), 3.86-3.79 (m, 4H), 3.40-3.30 (m, 1H), 1.28 (d, J = 6.9 Hz, 3H), 1.11- 1.04 (m, 1H), 0.49-0.40 (m, 2H), 0.39-0.25 (m, 2H). [0385] EXAMPLE 18: Compound 65 using General Synthetic Route 18:
Figure imgf000133_0001
1.1) Synthesis of 2-chloro-N-(methylsulfonyl)-4-morpholinofuro[3,2-d]pyrimidine-6- carboxamide
Figure imgf000133_0002
[0386] A solution of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid (200 mg, 0.71 mmol), methanesulfonamide (134 mg, 1.42 mmol), 2-chloro-1-methylpyridin-1-ium iodide (216 mg, 0.85 mmol), Et3N (214 mg, 2.12 mmol) and DMAP (4.3 mg, 0.035 mmol) in DCM (25 mL) was stirred at room temperature overnight. The completion of the reaction was monitored by TLC. The reaction was quenched with water (10 ml) and adjusted the pH to 4 using 1 N HCl aqueous solution. The aqueous phase was extracted with DCM/MeOH (15/1, 3 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The residue was purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide 2-chloro-N-(methylsulfonyl)-4- morpholinofuro [3,2-d]pyrimidine-6-carboxamide (244 mg, 0.68 mmol) as a yellow solid. LC- MS (ESI+): m/z 361/363 (MH+). 1.2) Synthesis of 2-((1-(N,N-dimethylsulfamoyl)-3-phenyl-1H-pyrazol-5-yl)amino)-N- (methylsulfonyl)-4-morpholinofuro[3,2-d]pyrimidine-6-carboxamide [0387] A suspension of 2-chlo
Figure imgf000133_0003
ro-N-(methylsulfonyl)-4-morpholinofuro[3,2-d]pyrimidine-6- carboxamide (60 mg x3, 0.17 mmol), 5-amino-N,N-dimethyl-3-phenyl-1H-pyrazole-1- sulfonamide (53 mg, 0.2 mmol), Cs2CO3 (127 mg, 0.39 mmol), Pd(OAc)2 (3.8 mg, 0.017 mmol) and Xantphos (9.6 mg, 0.017 mmol) in DMF/1,4-dioxane (7/1, 8 mL) was heated to 100 °C for 40 min under microwave condition. The reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 2% MeOH/DCM to 10% MeOH/DCM to provide 2-((1-(N,N-dimethylsulfamoyl)-3-phenyl-1H-pyrazol-5-yl)amino)-N- (methylsulfonyl)-4-morpholinofuro[3,2-d]pyrimidine-6-carboxamide (30 mg, 0.051 mmol) as a white solid. LC-MS (ESI+): m/z 591 (MH+).1HNMR (300 MHz, CD3OD) δ 7.86 (d, J = 6.9 Hz, 2H), 7.48-7.40 (m, 3H), 7.28 (s, 1H), 7.21 (s, 1H), 4.13-4.07 (m, 4H), 3.89-3.84 (m, 4H), 3.15 (s, 3H), 3.03 (s, 6H). 1.3) Synthesis of Compound 65, N-(methylsulfonyl)-4-morpholino-2-((3-phenyl-1H-pyrazol-5-yl) amino)furo[3,2-d]pyrimidine-6-carboxamide hydrochloride [0388] To a solution of
Figure imgf000134_0001
2-((1-(N,N-dimethylsulfamoyl)-3-phenyl-1H-pyrazol-5-yl)amino)-N- (methylsulfonyl)-4-morpholinofuro[3,2-d]pyrimidine-6-carboxamide (30 mg, 0.05 mmol) in DCM (4 mL) was added HCl/Et2O (2 mL). The reaction mixture was stirred at room temperature for 2 h. After concentration and slurry in MeOH/Et2O (1/20, 2 mL), N- (methylsulfonyl)-4-morpholino-2- ((3-phenyl-1H-pyrazol-5-yl)amino)furo[3,2-d]pyrimidine-6- carboxamide hydrochloride (Compound 65, 23.3 mg, 0.082 mmol) was obtained as a white solid. LC-MS (ESI+): m/z 484 (MH+). 1HNMR (300 MHz, CD3OD) δ 7.75 (s, 1H), 7.72 (d, J = 6.9 Hz, 2H), 7.52-7.40 (m, 3H), 6.43(s, 1H), 4.36-4.18 (m, 4H), 3.91-3.86 (m, 4H), 3.39 (s, 3H). [0389] EXAMPLE 19: Compound 66 using General Synthetic Route 19:
Figure imgf000134_0002
1.1) Synthesis of 2-chloro-N-(cyclopropylmethyl)-N-methyl-4-morpholinofuro[3,2-d] pyrimidine-6-carboxamide
Figure imgf000135_0001
[0390] A solution of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid (270 mg, 0.95 mmol), 1-cyclopropyl-N-methylmethanamine (80 mg, 0.95 mmol), DMAP (292 mg, 2.4 mmol) and EDCl (460 mg, 2.4 mmol) in DCM (20 mL) was stirred at room temperature overnight. The completion of the reaction was monitored by TLC. The reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 2% MeOH/DCM to provide 2-chloro-N-(cyclopropylmethyl)-N-methyl- 4-morpholinofuro[3,2-d] pyrimidine-6-carboxamide (272mg, 0.78 mmol) as a white solid. LC- MS (ESI+): m/z 351/353 (MH+).1HNMR (300 MHz, CDCl3) δ 7.09 (s, 1H), 4.09-4.04 (m, 4H), 3.88-3.82 (m, 4H), 3.48-3.37 (m, 2H), 3.29 (s, 1.5H), 3.20 (s, 1.5H), 1.11-0.98 (m, 1H), 0.65- 0.60 (m, 2H), 0.34-0.21 (m, 2H). 1.2) Synthesis of Compound 66, N-(cyclopropylmethyl)-N-methyl-4-morpholino-2-(4-phenyl - 1H-pyrazol-1-yl)furo[3,2-d]pyrimidine-6-carboxamide
Figure imgf000135_0002
[0391] A suspension of 2-chloro-N-(cyclopropylmethyl)-N-methyl-4-morpholinofuro[3,2- d]pyrimidine- 6-carboxamide (100 mg, 0.29 mmol), 4-phenyl-1H-pyrazole (41 mg, 0.29 mmol), CuI (11 mg, 0.06 mmol) and Cs2CO3 (186 mg, 0.57 mmol) in DMF (10 mL) was stirred at 100 °C overnight. The completion of the reaction was monitored by TLC. The reaction mixture was diluted with water (20 mL) and extracted with DCM/MeOH (15/1, 3 x 30 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The residue was purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 2% MeOH/DCM to provide N-(cyclopropylmethyl)-N-methyl-4- morpholino-2- (4-phenyl-1H-pyrazol-1-yl)furo[3,2-d]pyrimidine-6-carboxamide (Compound 66, 27 mg, 0.059 mmol) as a white solid. LC-MS (ESI+): m/z 459 (MH+).1HNMR (300 MHz, DMSO-d6) δ 9.05 (s, 1H), 8.35 (s, 1H), 7.80 (d, J = 7.2 Hz, 2H), 7.44-7.39 (m, 3H), 7.30-7.28 (m, 1H), 4.12-4.06 (m, 4H), 3.85-3.78 (m, 4H), 3.39-3.30 (m, 2H), 3.30 (s, 1.5H), 3.10 (s, 1.5H), 1.10-1.01 (m, 1H), 0.53-0.51 (m, 2H), 0.34-0.15 (m, 2H). [0392] EXAMPLE 20: Compound 69 using General Synthetic Route 20:
Figure imgf000136_0002
1.1) Synthesis of 4-cyclopropyl-3-oxobutanenitrile
Figure imgf000136_0001
[0393] To a solution of acetonitrile (1.08 g, 26.3 mmol) and methyl 2-cyclopropylacetate (2.0 g, 17.5 mmol) in anhydrous THF (40 mL) at 0 °C under N2 was added NaHDMS (13.2 mL, 26.3 mmol) dropwise. After addition, the solution was stirred at room temperature for 2 h. The completion of the reaction was monitored by TLC. The reaction was quenched with NH4Cl aqueous solution (20 mL) and extracted with EtOAc (3 x 30 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 33% EtOAc/PE to provide 4-cyclopropyl-3-oxobutanenitrile (1.6 g, 13.1 mmol) as yellow oil. 1HNMR (300 MHz, CDCl3) δ 3.34 (s, 2H), 2.29 (d, J = 6.9 Hz, 2H), 1.89- 1.84 (m, 1H), 0.85-0.79 (m, 2H), 0.06-0.01 (m, 2H). 1.2) Synthesis of 3-(cyclopropylmethyl)-1H-pyrazol-5-amine
Figure imgf000137_0001
[0394] To a solution of 4-cyclopropyl-3-oxobutanenitrile (1.15 g, 7.2 mmol) in EtOH (40 mL) was added NH2NH2.H2O (0.98 g, 19.5 mmol). The reaction mixture was heated to reflux overnight. The completion of the reaction was monitored by TLC. The reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 3% MeOH/DCM to provide 3-(cyclopropylmethyl)-1H-pyrazol-5-amine (1.64 g, 0.012 mmol) as a yellow oil. LC-MS (ESI+): m/z 138 (MH+).1HNMR (300 MHz, CDCl3) δ 5.51 (m, 1H), 4.41 (brs, 2H), 2.47 (d, J = 6.9 Hz, 2H), 1.02-0.90 (m, 1H), 0.59-0.56 (m, 2H), 0.22-0.17 (m, 2H). 1.3) Synthesis of 5-amino-3-(cyclopropylmethyl)-N,N-dimethyl-1H-pyrazole-1-sulfonamide
Figure imgf000137_0002
[0395] To a solution of 3-(cyclopropylmethyl)-1H-pyrazol-5-amine (1.0 g, 7.25 mmol) in THF (30 mL) at 0 °C was added NaH (521 mg, 8.7 mmol). After stirred at 0 °C for 1 h, to the solution was added dimethylsulfamoyl chloride (1.14 g, 7.97 mmol). The completion of the reaction was monitored by TLC. The reaction mixture was quenched with water and extracted with EtOAc (3 x 50 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 33% EtOAc/PE to provide 5-amino-3-(cyclopropylmethyl)-N,N-dimethyl-1H-pyrazole-1-sulfonamide (540 mg, 2.2 mmol). LC-MS: m/z 245 (MH+).1HNMR (300 MHz, CDCl3) δ 5.79 (s, 1H), 3.83 (brs, 2H), 2.94 (s, 6H), 2.71 (d, J = 6.9 Hz, 2H), 1.08-1.01 (m, 1H), 0.60-0.54 (m, 2H), 0.23-0.19 (m, 2H). 1.4) Synthesis of 3-(cyclopropylmethyl)-N,N-dimethyl-5-((4-morpholino-6-(pyridin-4-yl) furo[3,2-d]pyrimidin-2-yl)amino)-1H-pyrazole-1-sulfonamide [0396] A suspension of 2-brom
Figure imgf000137_0003
o-4-morpholino-6-(pyridin-4-yl)furo[3,2-d]pyrimidine (200 mg, 0.56 mmol), 5-amino-3-(cyclopropylmethyl)-N,N-dimethyl-1H-pyrazole-1-sulfonamide (162 mg, 0.67 mmol), Cs2CO3 (365 mg, 1.12 mmol), Pd(OAc)2 (12 mg, 0.056 mmol) and Xantphos (32 mg, 0.056 mmol) in DMF/1,4-dioxane (7/1, 8 mL) was heated to 85 °C for 40 min under microwave condition. The reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide 3-(cyclopropylmethyl)-N,N-dimethyl-5-((4-morpholino-6-(pyridin-4- yl)furo[3,2-d] pyrimidin-2-yl)amino)-1H-pyrazole-1-sulfonamide (47 mg, 0.09 mmol) as a white solid. LC-MS (ESI+): m/z 525 (MH+). 1HNMR (300 MHz, CDCl3) δ 8.74 (d, J = 5.7 Hz, 1H), 8.69 (s, 1H), 7.64 (d, J = 6.0 Hz, 1H), 7.18 (s, 1H), 6.85 (s, 1H), 4.12-4.07 (m, 4H), 3.92-3.88 (m, 4H), 2.99 (s, 6H), 2.54 (d, J = 7.2 Hz, 2H), 1.11-1.02 (m, 1H), 0.57-0.53 (m, 2H), 0.28-0.23 (m, 2H). 1.5) Synthesis of Compound 69, N-(3-(cyclopropylmethyl)-1H-pyrazol-5-yl)-4-morpholino- 6- (pyridin-4-yl)furo[3,2-d]pyrimidin-2-amine hydrochloride
Figure imgf000138_0001
[0397] To a solution of 3-(cyclopropylmethyl)-N,N-dimethyl-5-((4-morpholino-6-(pyridin-4- yl) furo[3,2-d]pyrimidin-2-yl)amino)-1H-pyrazole-1-sulfonamide (47 mg, 0.09 mmol) in DCM (4 mL) was added HCl/Et2O (2 mL). The reaction mixture was stirred at room temperature for 2 h. After concentration and slurry in MeOH/Et2O (1/20, 2 mL), N-(3-(cyclopropylmethyl)-1H- pyrazol-5-yl)- 4-morpholino-6-(pyridin-4-yl)furo[3,2-d] pyrimidin-2-amine hydrochloride (Compound 69, 32.5 mg, 0.072 mmol) was obtained as a yellow solid. LC-MS (ESI+): m/z 418 (MH+).1HNMR (300 MHz, CD3OD) δ 8.98 (d, J = 6.9 Hz, 2H), 8.58 (d, J = 6.9 Hz, 2H), 8.06 (s, 1H), 6.00 (s, 1H), 4.37-4.27 (m, 4H), 3.94-3.90 (m, 4H), 2.61 (d, J = 6.9 Hz, 2H), 1.09-1.02 (m, 1H), 0.62-0.54 (m, 2H), 0.29-0.24 (m, 2H). [0398] EXAMPLE 21: Compound 78 using General Synthetic Route 21:
Figure imgf000138_0002
1.1) Synthesis of 5-amino-3-cyclopropyl-N,N-dimethyl-1H-pyrazole-1-sulfonamide H N
Figure imgf000139_0001
[0399] To a solution of 3-cyclopropyl-1H-pyrazol-5-amine (500 mg, 4.06 mmol) in THF (30 mL) at 0 °C was added NaH (243 mg, 6.1 mmol). After stirred at 0 °C for 1 h, to the solution was added dimethylsulfamoyl chloride (755 mg, 5.28 mmol). The completion of the reaction was monitored by TLC. The reaction mixture was quenched with water and extracted with EtOAc (3 x 50 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 25% EtOAc/PE to provide 5- amino-3-cyclopropyl-N,N-dimethyl-1H-pyrazole-1-sulfonamide (372 mg, 1.6 mmol). LC-MS: m/z 231 (MH+).1HNMR (300 MHz, CDCl3) δ 5.05 (s, 1H), 4.71 (brs, 2H), 2.96 (s, 6H), 1.84- 1.80 (m, 1H), 0.93-0.86 (m, 2H), 0.72-0.67 (m, 2H). 1.2) Synthesis of 2-chloro-N-methyl-4-morpholinofuro[3,2-d]pyrimidine-6-carboxamide [0400] A solution of 2-chloro-4-m
Figure imgf000139_0002
orpholinofuro[3,2-d]pyrimidine-6-carboxylic acid (400 mg, 1.4 mmol), methanamine hydrochloride (113 mg, 1.70 mmol), EDCl (678 mg, 3.5 mmol) and DMAP (431 mg, 3.5 mmol) in DCM (3 mL) was stirred at room temperature overnight. The completion of the reaction was monitored by TLC. The reaction mixture was quenched with water (10 ml) and extracted with DCM/MeOH (15/1, 3 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The residue was purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide 2-chloro-N-methyl-4-morpholinofuro[3,2- d]pyrimidine-6-carboxamide (230 mg, 0.78 mmol). LC-MS (ESI+): m/z 297/299 (MH+). 1HNMR (300 MHz, CDCl3) δ 7.35 (s, 1H), 6.34 (brs, 1H), 4.08-4.03 (m, 4H), 3.88-3.83 (m, 4H), 3.07 (d, J = 5.1 Hz, 3H). 1.3) Synthesis of 2-bromo-N-methyl-4-morpholinofuro[3,2-d]pyrimidine-6-carboxamide
Figure imgf000140_0002
[0401] A solution of 2-chloro-N-methyl-4-morpholinofuro[3,2-d]pyrimidine-6-carboxamide (230 mg, 0.78 mmol) in HBr/AcOH (33 wt.% in Acetic acid, 3 mL) was heated to reflux for 3.5 h. The completion of the reaction was monitored by LC-MS. The reaction mixture was quenched with a saturated NaHCO3 aqueous solution and the pH was adjusted to 8. The aqueous solution was extracted with DCM/MeOH (15/1, 3 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure to provide 245 mg of crude 2-bromo-N-methyl-4-morpholinofuro[3,2-d]pyrimidine-6-carboxamide as a brown solid. The crude product was used directly for the next step without further purification. LC-MS (ESI+): m/z 341/343 (MH+). 1.4) Synthesis of 2-((3-cyclopropyl-1-(N,N-dimethylsulfamoyl)-1H-pyrazol-5-yl)amino) -N- methyl-4-morpholinofuro[3,2-d]pyrimidine-6-carboxamide
Figure imgf000140_0001
[0402] A solution of 2-bromo-N-methyl-4-morpholinofuro[3,2-d]pyrimidine-6-carboxamide (50 mg x3, 0.15 mmol), 5-amino-3-cyclopropyl-N,N-dimethyl-1H-pyrazole-1-sulfonamide (40 mg, 0.18 mmol), Cs2CO3 (110 mg, 0.35 mmol), Pd(OAc)2 (3.5 mg, 0.015 mmol) and Xantphos (8.5 mg, 0.015 mmol) in DMF/1,4-dioxane (7/1, 4 mL) was heated to 80 °C for 20 min under microwave condition. The reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide 2-((3-cyclopropyl-1-(N,N-dimethylsulfamoyl)-1H-pyrazol-5-yl)amino)-N-methyl-4- morpholinofuro[3,2-d]pyrimidine-6-carboxamide (62 mg, 0.13 mmol) as a white solid. LC-MS (ESI+): m/z 491 (MH+).1HNMR (300 MHz, CDCl3) δ8.67 (s, 1H), 7.32 (s, 1H), 6.55 (s, 1H), 6.33 (d, J = 5.1 Hz, 1H), 4.03-3.95 (m, 4H), 3.88-3.84 (m, 4H), 3.05 (d, J = 5.1 Hz, 3H), 2.89 (s, 6H), 1.95-1.92 (m, 1H), 0.99-0.93 (m, 2H), 0.87-0.84 (m, 2H). 1.5) Synthesis of Compound 78, 2-((3-cyclopropyl-1H-pyrazol-5-yl)amino)-N-methyl-4- morpholinofuro[3,2-d]pyrimidine-6-carboxamide hydrochloride
Figure imgf000141_0001
[0403] To a solution of 2-((3-cyclopropyl-1-(N,N-dimethylsulfamoyl)-1H-pyrazol-5- yl)amino)-N-methyl- 4-morpholinofuro[3,2-d]pyrimidine-6-carboxamide (62 mg, 0.13 mmol) in DCM (4 mL) was added HCl/Et2O (3 mL). The reaction mixture was stirred at room temperature for 2 h. After concentration and slurry in MeOH/Et2O (1/20, 2 mL), 2-((3- cyclopropyl-1H-pyrazol-5-yl)amino)-N-methyl- 4-morpholinofuro[3,2-d]pyrimidine-6- carboxamide hydrochloride (Compound 78, 32.8 mg, 0.078 mmol) was obtained as a white solid. LC-MS (ESI+): m/z 384 (MH+). 1HNMR (300 MHz, CD3OD) δ 7.51 (s, 1H), 5.73 (s, 1H), 4.30-4.15 (m, 4H), 3.92-3.86 (m, 4H), 2.96 (s, 3H), 1.99-1.91 (m, 1H), 1.19-1.15 (m, 2H), 0.89- 0.67 (m, 2H). [0404] The compounds in Table 2 were prepared using methods analogous to those described in the referenced General Synthetic Routes. Table 2
Figure imgf000141_0002
Figure imgf000142_0001
Figure imgf000143_0001
Figure imgf000144_0001
Figure imgf000145_0001
Figure imgf000146_0001
Figure imgf000147_0001
Figure imgf000148_0001
Figure imgf000149_0001
Figure imgf000150_0001
Figure imgf000151_0001
Figure imgf000152_0001
Figure imgf000153_0001
Figure imgf000154_0001
Figure imgf000155_0001
Figure imgf000156_0001
Figure imgf000157_0001
[0405] EXAMPLE 22: Compound 89 using General Synthetic Route 22:
Figure imgf000158_0002
1.1) Synthesis of (Z)-methyl 2-((2-cyano-1-(pyridin-4-yl)vinyl)oxy)acetate
Figure imgf000158_0003
[0406] To a solution of triphenylphosphine (3.5 g, 13.4 mmol) in dry tetrahydrofuran (THF) was added Diethyl azodicarboxylate (DEAD) (2.3 g, 13.4 mmol), 3-oxo-3-(pyridin-4- yl)propanenitrile (1.5 g, 10.3 mmol) and methyl 2-hydroxyacetate (1.2 g, 13.4 mmol) under N2. The reaction was stirred at room temperature overnight. Upon the completion of the reaction as monitored by thin layer chromatography (TLC), the reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 2% MeOH/DCM to provide 4.4 g of impure (Z)-methyl2-((2-cyano-1-(pyridin-4- yl)vinyl)oxy)acetate containing triphenylphosphine oxide as a yellow solid. LC-MS (ESI+): m/z 219 (MH+). 1.2) Synthesis of methyl 3-amino-5-(pyridin-4-yl)furan-2-carboxylate NH
Figure imgf000158_0001
[0407] To a solution of impure (Z)-methyl2-((2-cyano-1-(pyridin-4-yl)vinyl)oxy)acetate (4.6 g, 21.1 mmol) in dry THF at 0°C was added NaH (1.5 g, 31.6 mmol). The reaction was warmed to room temperature and stirred at room temperature for 2 h. Upon the completion of the reaction as monitored by TLC, the reaction was quenched with a saturated NH4Cl solution and the pH was adjusted to 3 using 2 N HCl aqueous solutions. The aqueous solution was washed with ethyl acetate (3 x 50 mL) to remove some impurities. To the aqueous solution was added a saturated Na2CO3 solution to adjust the pH to 11 and extracted with DCM/MeOH (10/1, 3 x 60 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated to provide 1.35 g of crude methyl 3-amino-5-(pyridin-4-yl)furan-2- carboxylate as an oil. The crude product was used directly for the next step without further purification. LC-MS (ESI+): m/z 219 (MH+).1HNMR (300 MHz, CDCl3) δ 8.66 (dd, J = 4.5, 1.5 Hz, 2H), 7.58 (dd, J = 4.8, 1.2 Hz, 2H), 6.58 (s, 1H), 4.67 (brs, 2H), 3.93 (s, 3H). 1.3) Synthesis of methyl 5-(pyridin-4-yl)-3-ureidofuran-2-carboxylate
Figure imgf000159_0001
[0408] To a solution of methyl 3-amino-5-(pyridin-4-yl)furan-2-carboxylate (1.94 g, 8.9 mmol) in DCM (40 mL) at -78 °C under N2 was added sulfurisocyanatidic chloride (3.79 g, 26.7 mmol) dropwise. After addition, the reaction was warmed to room temperature and stirred at room temperature for 1 h. The organic solvent was removed by evaporation in vacuo. To the resulting residue was added 6 N HCl (10 mL) aqueous solutions. The mixture was heated to reflux for 30 min. Upon the completion of the reaction as monitored by TLC, the reaction was cooled to room temperature and the pH was adjusted to 9 using a saturated NaHCO3 solution. A large amount of solid was precipitated. After filtration, the filter cake was washed with water and dried to provide 2.7 g of crude methyl5-(pyridin-4-yl)-3-ureidofuran-2-carboxylate as a yellow solid. LC-MS (ESI+): m/z 262 (MH+).1HNMR (300 MHz, CD3OD) δ 8.61 (dd, J = 4.8, 1.5 Hz, 2H), 7.89 (s, 1H), 7.78 (dd, J = 5.1, 1.5 Hz, 2H), 3.95 (s, 3H). 1.4) Synthesis of 6-(pyridin-4-yl)furo[3,2-d]pyrimidine-2,4-diol [0409] To a solution of crude
Figure imgf000159_0002
methyl 5-(pyridin-4-yl)-3-ureidofuran-2-carboxylate (2.7 g, 10.3 mmol) in MeOH (40 mL) was added 1.5 N NaOH (15 mL). The reaction was heated to reflux for 1.5 h. Upon the completion of the reaction as monitored by TLC, the solvent MeOH was removed by evaporation in vacuo. To the resulting residue were added 6 N HCl solutions until the pH to 2. A large amount of solid was precipitated. After filtration, the filter cake was washed with water and dried to provide crude 2.1 g of 6-(pyridin-4-yl)furo[3,2-d]pyrimidine-2,4-diol as a yellow solid. LC-MS (ESI+): m/z 230 (MH+).1HNMR (300 MHz, DMSO-d6) δ 11.61 (s, 1H), 11.37 (s, 1H), 8.89 (d, J = 6.6 Hz, 2H), 8.24 (d, J = 6.3 Hz, 2H), 7.63 (s, 1H). 1.5) Synthesis of 2,4-dichloro-6-(pyridin-4-yl)furo[3,2-d]pyrimidine
Figure imgf000160_0001
[0410] To a solution of 6-(pyridin-4-yl)furo[3,2-d]pyrimidine-2,4-diol (1.5 g, 6.54 mmol) in phenylphosphonic dichloride (30 mL) was added DIPEA (8.43 g, 65.4 mmol). The reaction mixture was heated to 120 °C overnight. Upon the completion of the reaction as monitored by TLC, the reaction was cooled to room temperature, a saturated NaHCO3 solution was added to adjust the pH to 8. The aqueous solution was extracted with EtOAc (3 x 50 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated. The resulting residue was purified by silica gel column chromatography with a gradient elution of 50% EtOAc/PE to 75% EtOAc/PE to provide 2,4-dichloro-6-(pyridin-4-yl) furo[3,2-d]pyrimidine (1.6 g, 6.9 mmol) as a yellow solid. LC-MS (ESI+): m/z 266/268 (MH+).1HNMR (300 MHz, CDCl3) δ 8.85 (dd, J = 4.8, 1.5 Hz, 2H), 7.82 (dd, J = 4.5, 1.5 Hz, 2H), 7.38 (s, 1H). 1.6) Synthesis of 2-chloro-4-morpholino-6-(pyridin-4-yl)furo[3,2-d]pyrimidine
Figure imgf000160_0002
[0411] To a solution of 2,4-dichloro-6-(pyridin-4-yl)furo[3,2-d]pyrimidine (1.6 g, 6.9 mmol) in DCM/EtOH (1/3, 120 mL) was added morpholine (0.91 g, 10.5 mmol) and K2CO3 (1.91 g, 14 mmol). The reaction was stirred at room temperature for 2 h. Upon the completion of the reaction as monitored by TLC, the reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 2% MeOH/DCM to 3% MeOH/DCM to provide 2-chloro-4-morpholino-6-(pyridin-4-yl)furo[3,2-d]pyrimidine (770 mg, 2.43 mmol) as a yellow solid. LC-MS (ESI+): m/z 317/319 (MH+).1HNMR (300 MHz, DMSO- d6) δ 8.76 (d, J = 6.0 Hz, 2H), 7.97 (d, J = 6.0 Hz, 2H), 7.82 (s, 1H), 4.06-3.97 (m, 4H), 3.82- 3.75 (m, 4H). 1.7) Synthesis of 4-morpholino-6-(pyridin-4-yl)-2-(3-(m-tolyl)-1H-pyrazol-1-yl)furo [3,2- d]pyrimidine
Figure imgf000161_0001
[0412] To a solution of 2-chloro-4-morpholino-6-(pyridin-4-yl)furo[3,2-d]pyrimidine (100 mg, 0.32 mmol) in DMF (10 mL) was added 3-(m-tolyl)-1H-pyrazole (55 mg, 0.35 mmol), Cs2CO3 (210 mg, 0.64 mmol) and CuI (12 mg, 0.064 mmol). The reaction mixture was stirred at 110 °C overnight. Upon the completion of the reaction as monitored by TLC, the reaction was cooled to room temperature, quenched with water (10 mL) and extracted with EtOAc (3 x 10 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 4% MeOH/DCM to provide 4-morpholino-6- (pyridin-4-yl)-2-(3-(m-tolyl)-1H-pyrazol-1-yl)furo [3,2-d]pyrimidine (28 mg, 0.064 mmol) as a white solid. LC-MS (ESI+): m/z 439 (MH+).1HNMR (300 MHz, DMSO-d6) δ 8.78 (brs, 2H), 8.72 (s, 1H), 8.05-8.01 (m, 2H), 7.91 (s, 1H), 7.81 (s, 1H), 7.55 (d, J = 7.5 Hz, 1H), 7.36 (t, J = 7.5 Hz, 1H), 7.20 (d, J = 7.2 Hz, 1H), 7.03 (s, 1H), 4.18-4.12 (m, 4H), 3.90-3.84 (m, 4H), 2.40 (s, 3H).
[0413] EXAMPLE 23: Compound 90 using General Synthetic Route 23:
Figure imgf000162_0001
1.1) Synthesis of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine
Figure imgf000162_0002
[0414] To a solution of 2,4-dichlorofuro[3,2-d]pyrimidine (6.46 g, 34.2 mmol mmol) in 1,4- dioxane (100 mL) was added morpholine (5.95 g, 68.4 mmol). The reaction was stirred at room temperature for 30 min. Upon the completion of the reaction as monitored by TLC, the reaction mixture was concentrated directly and the resulting residue was purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 20% EtOAc/PE to provide 2- chloro-4-morpholinofuro[3,2-d]pyrimidine (7.5 g, 40.1 mmol) as a white solid. LC-MS (ESI+): m/z 240/242 (MH+).1HNMR (300 MHz, CDCl3) δ 7.74 (d, J = 1.8 Hz, 1H), 6.79 (d, J = 2.1 Hz, 1H), 4.05-4.02 (m, 4H), 3.85-3.82 (m, 4H). 1.2) Synthesis of 2-chloro-6-iodo-4-morpholinofuro[3,2-d]pyrimidine
Figure imgf000163_0001
[0415] To a solution of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine (2.0 g, 0.83 mmol) in anhydrous THF (30 mL) at -78 °C under N2 was added LDA (1.33 mL, 2M, 2.66 mmol). After stirred at -78 °C for 1 h, to the solution was added a solution of NIS (2.25 g, 1.0 mmol) in anhydrous THF (10 mL). Upon the completion of the reaction as monitored by TLC, the reaction was quenched with water (50 mL) and extracted with DCM (3 x 50 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 5% EtOAc/PE to 10% EtOAc/PE to provide 2-chloro-6-iodo-4- morpholinofuro[3,2-d]pyrimidine (1.6 g, 4.4 mmol) as yellow solid. LC-MS (ESI+): m/z 366/368 (MH+).1HNMR (300 MHz, CDCl3) δ 6.97 (s, 1H), 4.01-3.98 (m, 4H), 3.85-3.82 (m, 4H). 1.3) Synthesis of 2-chloro-4-morpholino-6-(pyridin-3-yl)furo[3,2-d]pyrimidine
Figure imgf000163_0002
[0416] To a solution of 2-chloro-6-iodo-4-morpholinofuro[3,2-d]pyrimidine (500 mg, 1.37 mmol) in 1,4-dioxane/H2O (2/1, 30 mL) was added pyridin-3-ylboronic acid (168 mg, 1.37 mmol), K2CO3 (567 mg, 4.1 mmol) and Pd(PPh3)4 (158 mg, 0.13 mmol). The reaction was stirred at 90 °C for 5 h. Upon the completion of the reaction as monitored by TLC, the reaction solution was concentrated directly and the resulting residue was purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 3% MeOH/DCM to provide 2- chloro-4-morpholino-6-(pyridin-3-yl)furo[3,2-d] pyrimidine (410 mg, 1.29 mmol) as a light yellow solid. LC-MS (ESI+): m/z 317/319 (MH+).1HNMR (300 MHz, CDCl3) δ 9.07 (d, J = 1.5 Hz, 1H), 8.69-8.68 (m, 1H), 8.06 (d, J = 8.1 Hz, 1H), 7.46-7.42 (m, 1H), 7.08 (s, 1H), 4.11-4.08 (m, 4H), 3.90-3.87 (m, 4H). 1.4) Synthesis of 4-morpholino-6-(pyridin-3-yl)-2-(3-(m-tolyl)-1H-pyrazol-1-yl) furo[3,2- d]pyrimidine
Figure imgf000164_0001
[0417] To a solution of 2-chloro-4-morpholino-6-(pyridin-3-yl)furo[3,2-d]pyrimidine (100 mg, 0.32 mmol) in DMF (10 mL) was added 3-(m-tolyl)-1H-pyrazole (60 mg, 0.38 mmol) and NaH (25 mg, 0.63 mmol). The reaction was stirred at 90 °C overnight. Upon the completion of the reaction as monitored by TLC, the reaction was quenched with water (50 mL) and extracted with EtOAc (3 x 30 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 3% MeOH/DCM to provide 4-morpholino-6-(pyridin-3-yl)-2-(3-(m-tolyl) -1H-pyrazol-1-yl)furo[3,2-d]pyrimidine (80 mg, 0.18 mmol) as an off-white solid. LC-MS (ESI+): m/z 439 (MH+).1HNMR (300 MHz, DMSO-d6) δ 9.30 (s, 1H), 8.71-8.69 (m, 2H), 8.43 (d, J = 7.5 Hz, 1H), 7.81-7.74 (m, 3H), 7.63- 7.60 (m, 1H), 7.35 (t, J = 7.5 Hz, 1H), 7.20 (d, J = 7.8 Hz, 1H), 7.02 (d, J = 2.7 Hz, 1H), 4.15- 4.11 (m, 4H), 3.90-3.82 (m, 4H), 2.41 (s, 3H). [0418] EXAMPLE 24: Compound 91 using General Synthetic Route 24:
Figure imgf000164_0002
1.1) Synthesis of 2-chloro-4-morpholino-6-(pyridin-2-yl)furo[3,2-d]pyrimidine
Figure imgf000164_0003
[0419] A solution of 2-chloro-6-iodo-4-morpholinofuro[3,2-d]pyrimidine (1 g, 2.7 mmol), 2- (tributylstannyl)pyridine (1.2 g, 3.3 mmol) and Pd(PPh3)4 (155 mg, 0.14 mmol) in toluene (5 mL) was heated to 90 °C overnight. Upon the completion of the reaction as monitored by TLC, the reaction was cooled to room temperature, diluted with water and extracted with DCM/MeOH (15/1, 3 x 50 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 5% MeOH/DCM to provide 2-chloro-4-morpholino-6-(pyridin-2-yl)furo[3,2-d] pyrimidine (352 mg, 1.11 mmol) as a yellow solid. LC-MS (ESI+): m/z 317/319 (MH+).1HNMR (300 MHz, DMSO- d6) δ 8.74-8.70 (m, 1H), 8.10 (d, J = 8.1 Hz, 1H), 8.00 (d, J = 7.5 Hz, 1H), 7.63-7.49 (m, 2H), 4.05-3.97 (m, 4H), 3.82-3.76 (m, 4H). 1.2) Synthesis of 4-morpholino-6-(pyridin-2-yl)-2-(3-(m-tolyl)-1H-pyrazol-1-yl) furo[3,2- d]pyrimidine
Figure imgf000165_0001
[0420] To a solution of 2-chloro-4-morpholino-6-(pyridin-2-yl)furo[3,2-d]pyrimidine (80 mg, 0.25 mmol) in DMF (10 mL) was added 3-(m-tolyl)-1H-pyrazole (48 mg, 0.30 mmol), Cs2CO3 (165 mg, 0.51 mmol) and Cu2O (3.6 mg, 0.025 mmol). The reaction was stirred at 110 °C overnight. Upon the completion of the reaction as monitored by TLC, the reaction was cooled to room temperature, quenched with water (10 mL) and extracted with DCM/MeOH (15/1, 3 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by preparative TLC to provide 4- morpholino-6-(pyridin-2-yl)-2-(3-(m-tolyl)-1H- pyrazol-1-yl)furo[3,2-d]pyrimidine (39 mg, 0.089 mmol) as a white solid. LC-MS (ESI+): m/z 439 (MH+).1HNMR (300 MHz, DMSO-d6) δ 8.76-8.71 (m, 2H), 8.12 (d, J = 7.5 Hz, 1H), 8.01 (t, J = 7.5 Hz, 1H), 7.80 (s, 1H), 7.75 (d, J = 7.2 Hz, 1H), 7.68 (s, 1H), 7.62-7.49 (m, 1H), 7.36 (t, J = 7.5 Hz, 1H), 7.20 (d, J = 7.2 Hz, 1H), 7.02 (s, 1H), 4.18-4.12 (m, 4H), 3.90-3.84 (m, 4H), 2.40 (s, 3H). [0421] EXAMPLE 25: Compound 92 using General Synthetic Route 25:
Figure imgf000166_0001
1.1) Synthesis of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid
Figure imgf000166_0002
[0422] To a solution of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine (2.4 g, 10 mmol) in anhydrous THF (40 mL) at -78 °C under N2 was added n-BuLi ( 5.2 mL, 2.5 M, 13 mmol) dropwise. The reaction mixture was stirred at that temperature for 1 h. To the solution was added excessive amount of dry ice in one portion. The resulting reaction mixture was stirred at that temperature for 3 h. Upon the completion of the reaction as monitored by TLC, the reaction was quenched with water and the pH was adjusted to 5 using 1 N HCl aqueous solution. The aqueous solution was extracted with DCM (3 x 80 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was slurry in Et2O to provide 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid ( 2.92 g, 10.3 mmol) as a yellow solid. LC-MS (ESI+): m/z 284/286 (MH+).1HNMR (300 MHz, CDCl3) δ 7.58 (s, 1H), 4.04-3.95 (m, 4H), 3.78-3.76 (m, 4H). 1.2) Synthesis of 2-chloro-N-methyl-4-morpholinofuro[3,2-d]pyrimidine-6-carboxamide
Figure imgf000166_0003
[0423] To a solution of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid (100 mg, 0.36 mmol) in DCM (10 mL) was added methanamine hydrochloride (13.5 mg, 0.2 mmol), EDCl (86 mg, 0.45 mmol) and DMAP (55 mg, 0.45 mmol). The reaction was stirred at room temperature overnight. Upon the completion of the reaction as monitored by TLC, the reaction was quenched with water (10 mL) and extracted with DCM (3 x 10 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 2% MeOH/DCM to provide 2-chloro-N-methyl-4-morpholinofuro[3,2- d]pyrimidine-6- carboxamide (67 mg, 0.22 mmol) as white solid. LC-MS (ESI+): m/z 297/299 (MH+).1HNMR (300 MHz, CDCl3) δ 7.61 (s, 1H), 6.35 (brs, 1H), 4.06-4.03 (m, 4H), 3.88-3.85 (m, 4H), 3.06 (d, J = 5.1 Hz, 3H). 1.3) Synthesis of N-methyl-4-morpholino-2-(3-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d] pyrimidine- 6-carboxamide
Figure imgf000167_0001
[0424] To a solution of 2-chloro-N-methyl-4-morpholinofuro[3,2-d]pyrimidine-6- carboxamide (60 mg, 0.19 mmol) in DMF (4 mL) was added 3-(m-tolyl)-1H-pyrazole (35 mg, 0.23 mmol), Cs2CO3 (123 mg, 0.38 mmol) and CuI (8 mg, 0.038 mmol). The reaction was stirred at 110 °C overnight. Upon the completion of the reaction as monitored by TLC, the reaction was then cooled to room temperature, quenched with water (10 mL) and extracted with EtOAc (3 x 10 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by preparative TLC to provide N-methyl-4-morpholino-2-(3-(m-tolyl)-1H-pyrazol-1-yl) furo[3,2-d]pyrimidine-6- carboxamide (15 mg, 0.036 mmol) as a white solid. LC-MS (ESI+): m/z 419 (MH+).1HNMR (300 MHz, DMSO-d6) δ 8.85-8.82 (m, 1H), 8.70 (d, J = 2.4 Hz, 1H), 7.79 (s, 1H), 7.74 (d, J = 8.1 Hz, 1H), 7.54 (s, 1H), 7.36 (t, J = 7.8 Hz, 1H), 7.20 (d, J = 7.2 Hz, 1H), 7.02 (d, J = 2.4 Hz, 1H), 4.12-4.08 (m, 4H), 3.86-3.82 (m, 4H), 2.86 (d, J = 4.5 Hz, 3H), 2.39 (s, 3H).
[0425] EXAMPLE 26: Compound 98 using General Synthetic Route 26:
Figure imgf000168_0001
1.1) Synthesis of 4-morpholino-2-(3-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d]pyrimidine [0426] To a solution of 2-
Figure imgf000168_0002
chloro-4-morpholinofuro[3,2-d]pyrimidine (300 mg, 1.25 mmol) in DMF (4 mL) was added 3-(m-tolyl)-1H-pyrazole (237 mg, 0.35 mmol), Cs2CO3 (815 mg, 2.5 mmol) and CuI (24 mg, 0.125 mmol). The reaction was stirred at 110 °C overnight. Upon the completion of the reaction as monitored by TLC, the reaction was then cooled to room temperature, quenched with water (10 mL) and extracted with EtOAc (3 x 10 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 30% EtOAc/PE to provide 4-morpholino-2-(3- (m-tolyl)- 1H-pyrazol-1-yl)furo[3,2-d]pyrimidine (269 mg, 0.75 mmol) as a white solid. LC-MS (ESI+): m/z 362 (MH+).1HNMR (300 MHz, DMSO-d6) δ 8.75 (d, J = 2.7 Hz, 1H), 8.38 (d, J = 1.8 Hz, 1H), 7.82 (s, 1H), 7.77 (d, J = 7.8 Hz, 1H), 7.36 (t, J = 7.8 Hz, 1H), 7.21 (d, J = 7.8 Hz, 1H), 7.13 (d, J = 1.8 Hz, 1H), 7.07 (d, J = 2.7 Hz, 1H), 4.12-4.06 (m, 4H), 3.92-3.86 (m, 4H), 2.40 (s, 3H). 1.2) Synthesis of 4-morpholino-2-(3-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d]pyrimidine -6- carboxylic acid
Figure imgf000169_0001
[0427] To a solution of 4-morpholino-2-(3-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d]pyrimidine (493 mg, 1.36 mmol) in anhydrous THF (20 mL) at -78 °C under N2 was added n-BuLi ( 0.8 mL, 2.5 M, 2.0 mmol) dropwise. The reaction mixture was stirred at that temperature for 1 h. To the solution was added excessive amount of dry ice in one portion. The resulting reaction mixture was stirred at that temperature for 3 h. Upon the completion of the reaction as monitored by TLC, the reaction was quenched with water and the pH was adjusted to 5 using 1 N HCl aqueous solution. The aqueous solution was extracted with DCM/MeOH (15/1, 2 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was slurry in Et2O to provide 4-morpholino-2-(3-(m- tolyl)-1H-pyrazol-1-yl)furo [3,2-d]pyrimidine-6-carboxylic acid (361 mg, 0.89 mmol) as a yellow solid. LC-MS (ESI+): m/z 406 (MH+).1HNMR (300 MHz, DMSO-d6) δ 14.18 (s,1H), 8.72 (d, J = 2.7 Hz, 1H), 7.79-7.71 (m, 3H), 7.35 (t, J = 7.5 Hz, 1H), 7.20 (d, J = 7.2 Hz, 1H), 7.03 (d, J = 2.4 Hz, 1H), 4.11-4.05 (m, 4H), 3.88-3.82 (m, 4H), 2.39 (s, 3H). 1.3) Synthesis of N-(methylsulfonyl)-4-morpholino-2-(3-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2- d]pyrimidine-6-carboxamide
Figure imgf000169_0002
[0428] To a solution of 4-morpholino-2-(3-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d]pyrimidine - 6-carboxylic acid (60 mg, 0.15 mmol) in DCM (10 mL) was added methanesulfonamide (28 mg, 0.30 mmol), 2-chloro-1-methylpyridinium iodide (45 mg, 0.18 mmol) and DMAP (1 mg, 0.007 mmol). The reaction was stirred at room temperature overnight. Upon the completion of the reaction as monitored by TLC, the reaction was quenched with water (10 mL) and extracted with DCM (3 x 10 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by preparative TLC to provide N-(methylsulfonyl)-4-morpholino -2-(3-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2- d]pyrimidine-6-carboxamide (14.2 mg, 0.03 mmol) as a yellow solid. LC-MS (ESI+): m/z 483 (MH+).1HNMR (300 MHz, CD3OD) δ 8.61 (s, 1H), 7.79-7.69 (m, 2H), 7.61-7.59 (m, 1H), 7.50- 7.18 (m, 2H), 6.84-6.80 (m, 1H), 4.25-4.14 (m, 4H), 3.91-3.85 (m, 4H), 3.16 (s, 3H), 2.37 (s, 3H). [0429] EXAMPLE 27: Compound 127 using General Synthetic Route 27:
Figure imgf000170_0001
1.1) Synthesis of 2-chloro-7-iodo-4-morpholinofuro[3,2-d]pyrimidine
Figure imgf000170_0002
[0430] To a solution of 2-chloro-6-iodo-4-morpholinofuro[3,2-d]pyrimidine (5.0 g, 13.7 mmol) in anhydrous THF (30 mL) at -78 °C under N2 was added LDA (14 mL, 2 M, 27.4 mmol). The reaction mixture was stirred at -78 °C for 1 h. Upon the completion of the reaction as monitored by TLC, the reaction was quenched with H2O (100 mL) and extracted with DCM (3 x 200 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduce pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 30% EtOAc/PE to provide 2- chloro-7-iodo-4-morpholinofuro[3,2-d]pyrimidine (2.5 g, 6.85 mmol) as a white solid. LC-MS (ESI+): m/z 366/368 (MH+).1HNMR (300 MHz, CDCl3) δ 7.77 (s, 1H), 4.04-3.97 (m, 4H), 3.85- 3.81 (m, 4H). 1.2) Synthesis of 2-chloro-7-methyl-4-morpholinofuro[3,2-d]pyrimidine
Figure imgf000171_0001
[0431] To a solution of 2-chloro-7-iodo-4-morpholinofuro[3,2-d]pyrimidine (50 mg, 0.14 mmol) in DME/H2O (2/1, 3 mL) was added methylboronic acid (25 mg, 0.42 mmol), K3PO4 (44 mg, 0.21 mmol) and Pd(PPh3)4 (15 mg, 0.014 mmol). The reaction was stirred at 120 °C for 30 min under Microwave condition. Upon the completion of the reaction as monitored by TLC, the reaction was cooled to room temperature, quenched with water (30 mL) and extracted with EtOAc (2 x 10 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 30% EtOAc/PE to provide 2- chloro-7-methyl-4-morpholinofuro[3,2-d]pyrimidine (35 mg, 0.14 mmol) as a white solid. LC- MS (ESI+): m/z 254/256 (MH+).1HNMR (300 MHz, CDCl3) δ 7.53 (s, 1H), 4.04-3.98 (m, 4H), 3.86-3.82 (m, 4H), 2.22 (s, 3H). 1.3) Synthesis of 2-chloro-7-methyl-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid
Figure imgf000171_0002
[0432] To a solution of 2-chloro-6-iodo-7-methyl-4-morpholinofuro[3,2-d]pyrimidine (90 mg, 0.24 mmol) in anhydrous THF (100 mL) at -78 °C under N2 was added n-BuLi (0.15 mL, 2.5 M, 0.36 mmol) dropwise. The reaction mixture was stirred at that temperature for 1 h. To the solution was added excessive amount of dry ice in one portion. The resulting reaction mixture was stirred at that temperature for 1 h. Upon the completion of the reaction as monitored by TLC, the reaction was quenched with water and the pH was adjusted to 5 using 1 N HCl aqueous solution. The aqueous solution was extracted with DCM (2 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was slurry in Et2O to provide 2-chloro-7-methyl-4- morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid (60 mg, 0.20 mmol) as a yellow solid. LC- MS (ESI+): m/z 298/300 (MH+). 1.4) Synthesis of 2-chloro-N-cyclopropyl-7-methyl-4-morpholinofuro[3,2-d]pyrimidine-6- carboxamide
Figure imgf000172_0001
[0433] To a solution of 2-chloro-7-methyl-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid (60 mg, 0.20 mmol) in DCM (40 mL) was added oxalyl dichloride (52 mg, 0.4 mmol) and DMF (10 mg, 0.13 mmol). The reaction was stirred at room temperature for 2 h. Upon the completion of the reaction as monitored by TLC, the reaction solution was concentrated directly and the resulting residue was dissolved in DCM (20 mL). To the solution was added cyclopropanamine (23 mg, 0.4 mmol), followed by Et3N (40 mg, 0.4 mmol) dropwise. The completion of the reaction was monitored by TLC. The reaction was quenched with water (20 mL) and extracted with EtOAc (2 x 10 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 20% EtOAc/PE to 50% EtOAc/PE to provide 2-chloro-N-cyclopropyl-7- methyl-4-morpholinofuro[3,2-d]pyrimidine-6- carboxamide (54 mg, 0.16mmol) as a white solid. LC-MS (ESI+): m/z 337/339 (MH+).1HNMR (300 MHz, CDCl3) δ 6.37 (s, 1H), 4.03-3.99 (m, 4H), 3.87-3.84 (m, 4H), 2.88-2.83 (m, 1H), 2.55 (s, 3H), 0.98-0.92 (m, 2H), 0.71-0.65 (m, 2H). 1.5) Synthesis of N-cyclopropyl-7-methyl-4-morpholino-2-(4-(m-tolyl)-1H-pyrazol-1- yl)furo[3,2-d]pyrimidine-6-carboxamide
Figure imgf000172_0002
[0434] To a solution of 2-chloro-N-cyclopropyl-7-methyl-4-morpholinofuro[3,2- d]pyrimidine-6- carboxamide (54 mg, 0.16mmol) in DMF (3.0 mL) was added 4-(m-tolyl)-1H- pyrazole (50 mg, 0.32 mmol), Cs2CO3 (105 mg, 0.32 mmol) and Cu2O (2.2 mg, 0.016 mmol). The reaction was stirred at 110 °C overnight. Upon the completion of the reaction as monitored by TLC, the reaction was quenched with water (10 mL) and a large amount of solid was precipitated. After filtration, the filtrate was concentrated directly and purified by flash silica gel column chromatography with a gradient elution of 20% EtOAc/PE to 50% EtOAc/PE to provide N-cyclopropyl-7-methyl-4-morpholino-2-(4-(m-tolyl)-1H -pyrazol-1-yl)furo[3,2-d]pyrimidine-6-carboxamide (25 mg, 0.054 mmol) as white solid. LC- MS (ESI+): m/z 459 (MH+).1HNMR (300 MHz, CDCl3) δ 8.74 (s, 1H), 8.08 (s, 1H), 7.43-7.40 (m, 2H), 7.30-7.27 (m, 1H), 7.10 (d, J = 6.9 Hz, 1H), 6.45 (s, 1H), 4.13-4.08 (m, 4H), 3.92-3.89 (m, 4H), 2.95-2.86 (m, 1H), 2.65 (s, 3H), 2.55 (s, 3H), 0.97-0.93 (m, 2H), 0.77-0.70 (m, 2H). [0435] EXAMPLE 28: Compound 122 using General Synthetic Route 28:
Figure imgf000173_0001
1.1) Synthesis of 2-chloro-6-iodo-7-met
Figure imgf000173_0002
hyl-4-morpholinofuro[3,2-d]pyrimidine
Figure imgf000173_0003
[0436] To a solution of 2-chloro-7-methyl-4-morpholinofuro[3,2-d]pyrimidine (350 mg, 1.38 mmol) in anhydrous THF (30 mL) at -78 °C under N2 was added LDA (1.4 mL, 2 M, 2.76 mmol). After stirred at -78 °C for 1 h, to the solution was added a solution of NIS (374 mg, 1.66 mmol) in anhydrous THF (5 mL). Upon the completion of the reaction as monitored by TLC, the reaction was quenched with water (50 mL) and extracted with EtOAc (3 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 30% EtOAc/PE to provide 2-chloro-6-iodo-7-methyl-4- morpholinofuro[3,2-d]pyrimidine (280 mg, 2.55 mmol) as yellow solid. LC-MS (ESI+): m/z 380/382 (MH+). 1.2) Synthesis of 2-chloro-7-methyl-4-morpholino-6-(pyridin-2-yl)furo[3,2-d]pyrimidine
Figure imgf000174_0002
[0437] A solution of 2-chloro-6-iodo-7-methyl-4-morpholinofuro[3,2-d]pyrimidine (200 mg, 0.53 mmol), 2-(tributylstannyl)pyridine (389 mg, 1.06 mmol) and Pd(PPh3)4 (61 mg, 0.053 mmol) in toluene (5 mL) was heated to 90 °C overnight. Upon the completion of the reaction as monitored by TLC, the reaction mixture was diluted with water and extracted with DCM/MeOH (15/1, 3 x 50 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 30% EtOAc/PE to 50% EtOAc/PE to provide 2- chloro-7-methyl-4-morpholino-6-(pyridin-2-yl) furo[3,2-d]pyrimidine (70 mg, 0.21 mmol) as a white solid. LC-MS (ESI+): m/z 331/333 (MH+). 1.3) Synthesis of 7-methyl-4-morpholino-6-(pyridin-2-yl)-2-(3-(m-tolyl)-1H- pyrazol-1- yl)furo[3,2-d]pyrimidine
Figure imgf000174_0001
[0438] To a solution of 2-chloro-7-methyl-4-morpholino-6-(pyridin-2-yl)furo[3,2- d]pyrimidine (80 mg, 0.24 mmol) in CH3CN (10 mL) was added 3-(m-tolyl)-1H-pyrazole (77 mg, 0.32 mmol) and Cs2CO3 (158 mg, 0.48 mmol). The reaction was stirred at 160 °C in a sealed tube overnight. The reaction mixture was then cooled to room temperature, diluted with water (10 mL) and extracted with DCM/MeOH (15/1, 3 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 30% EtOAc/PE to 50% EtOAc/PE to provide 7-methyl-4-morpholino-6- (pyridin-2-yl)-2-(3-(m- tolyl)-1H-pyrazol-1-yl)furo [3,2-d]pyrimidine (9.2 mg, 0.02 mmol) as white solid. LC-MS (ESI+): m/z 453 (MH+).1HNMR (300 MHz, CDCl3) δ 8.75 (d, J = 4.2 Hz, 1H), 8.61 (d, J = 2.1 Hz, 1H), 7.90 (s, 1H), 7.85-7.77 (m, 3H), 7.43-7.27 (m, 2H), 7.16 (d, J = 7.5 Hz, 1H), 6.78 (s, 1H), 4.23-4.17 (m, 4H), 3.96-3.87 (m, 4H), 2.77 (s, 3H), 2.43 (s, 3H). [0439] EXAMPLE 29: Compound 112 using General Synthetic Route 29:
Figure imgf000175_0001
1.1) Synthesis of 2-chloro-4-morpholino-7-(trifluoromethyl)furo[3,2-d]pyrimidine F C
Figure imgf000175_0002
[0440] A solid mixture of 2-chloro-7-iodo-4-morpholinofuro[3,2-d]pyrimidine (300 mg, 0.82 mmol), KF (144 mg, 2.47 mmol), CuI (30 mg, 0.16 mmol) and 1,10-phenanthroline (30 mg, 0.16 mmol) in three neck flask was heated to 100 °C under reduced pressure using oil pump for 1 h. After being cooled to room temperature, to the mixture was added anhydrous DMSO (6.0 mL) to form a brown solution. To the solution was added B(OMe)3 (252 mg, 2.47 mmol) and TMSCF3 (348 mg, 2.47 mmol) dropwise. The reaction mixture was then heated to 55 °C. After stirred at that temperature for 1 h, an additional TMSCF3 (348 mg, 2.47 mmol) was added dropwise. Upon the completion of the reaction as monitored by TLC, the reaction was quenched with water (10 mL) and extracted with DCM (3 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 30% EtOAc/PE to provide 2-chloro-4-morpholino -7-(trifluoromethyl)furo[3,2- d]pyrimidine (98 mg, 0.32 mmol) as a yellow solid. LC-MS (ESI+): m/z 308/310 (MH+). 1.2) Synthesis of 2-chloro-6-iodo-4-morpholino-7-(trifluoromethyl)furo[3,2-d]pyrimidine
Figure imgf000176_0001
[0441] To a solution of 2-chloro-4-morpholino-7-(trifluoromethyl)furo[3,2-d]pyrimidine (160 mg, 0.52 mmol) in anhydrous THF (10 mL) at -78 °C under N2 was added LDA (0.39 mL, 2 M, 0.78 mmol). After stirred at -78 °C for 1 h, to the solution was added a solution of NIS (141 mg, 0.63 mmol) in THF (10 mL). Upon the completion of the reaction as monitored by TLC, the reaction was quenched with water (25 mL) and extracted with DCM (3 x 15 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 30% EtOAc/PE to provide 2-chloro-6-iodo-4-morpholino- 7-(trifluoromethyl)furo[3,2-d] pyrimidine (148 mg, 0.34 mmol) as a white solid. LC-MS (ESI+): m/z 434/436 (MH+). 1.3) Synthesis of 2-chloro-4-morpholino-6-(pyridin-2-yl)-7-(trifluoromethyl)furo[3,2- d]pyrimidine [0442] A solution of 2-chloro
Figure imgf000176_0002
-6-iodo-4-morpholino-7-(trifluoromethyl)furo[3,2-d]pyrimidine (100 mg, 0.23 mmol), 2-(tributylstannyl)pyridine (170 mg, 0.46 mmol) and Pd(PPh3)4 (13 mg, 0.023 mmol) in toluene (5 mL) was heated to 90 °C overnight. Upon the completion of the reaction as monitored by TLC, the reaction mixture was diluted with water and extracted with EtOAc (3 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 20% EtOAc/PE to 40% EtOAc/PE to provide 2- chloro-4-morpholino-6-(pyridin-2-yl)-7- (trifluoromethyl)furo[3,2-d]pyrimidine (55 mg, 0.14 mmol) as a yellow solid. LC-MS (ESI+): m/z 385/387 (MH+). 1.4) Synthesis of 4-morpholino-6-(pyridin-2-yl)-2-(3-(m-tolyl)-1H-pyrazol-1-yl)-7- (trifluoromethyl)furo[3,2-d]pyrimidine
Figure imgf000177_0001
[0443] To a solution of 2-chloro-4-morpholino-6-(pyridin-2-yl)-7-(trifluoromethyl) furo[3,2- d]pyrimidine (55 mg, 0.14 mmol) in DMF (6 mL) was added 3-(m-tolyl)- 1H-pyrazole (45 mg, 0.29 mmol), Cs2CO3 (92 mg, 0.29 mmol) and Cu2O (2 mg, 0.014 mmol). The reaction was stirred at 110 °C overnight. The reaction mixture was then diluted with water (10 mL) and extracted with EtOAc (3 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 20% EtOAc/PE to 50% EtOAc/PE to provide 4-morpholino-6- (pyridin-2-yl)-2-(3-(m-tolyl)-1H-pyrazol-1-yl)-7- (trifluoromethyl)furo[3,2-d]pyrimidine (9.3 mg, 0.018 mmol) as a yellow solid. LC-MS (ESI+): m/z 507 (MH+).1HNMR (300 MHz, CDCl3) δ 9.04 (d, J = 4.2 Hz, 1H), 8.66 (d, J = 2.1 Hz, 1H), 8.28-8.25 (m, 1H), 8.13-8.08 (m, 1H), 7.86-7.78 (m, 3H), 7.36-7.31 (m, 1H), 7.19 (d, J = 7.2 Hz, 1H), 6.82 (s,1H), 4.40-4.31 (m, 4H), 4.02-3.92 (m, 4H), 2.43 (s, 3H). [0444] EXAMPLE 30: Compound 134 using General Synthetic Route 30:
Figure imgf000177_0002
1.1) Synthesis of 2,4-dichlorofuro[3,2-d]pyrimidine-6-carboxylic acid
Figure imgf000178_0002
[0445] To a solution of 2,4-dichlorofuro[3,2-d]pyrimidine (1 g, 5.29 mmol) in anhydrous THF (100 mL) at -78 °C under N2 was added LDA ( 5.3 mL, 2 M, 10.6 mmol) dropwise. The reaction mixture was stirred at that temperature for 1 h. To the solution was added excessive amount of dry ice in one portion. The resulting reaction mixture was stirred at that temperature for 1 h. Upon the completion of the reaction as monitored by TLC, the reaction was quenched with water and the pH was adjusted to 5 using 1 N HCl aqueous solution. The aqueous solution was extracted with DCM (2 x 40 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was slurried in Et2O to provide 2,4-dichlorofuro[3,2-d] pyrimidine-6-carboxylic acid (650 mg, 2.8 mmol) as a yellow solid. LC-MS (ESI+): m/z 233/235 (MH+). 1.2) Synthesis of 2,4-dichloro-N-ethylfuro[3,2-d]pyrimidine-6-carboxamide
Figure imgf000178_0001
[0446] To a solution of 2,4-dichlorofuro[3,2-d]pyrimidine-6-carboxylic acid (519 mg, 2.23 mmol) in DCM (40 mL) was added oxalyl dichloride (566 mg, 4.45 mmol) and DMF (20 mg, 0.27 mmol). The reaction was stirred at room temperature for 2 h. The completion of the reaction was monitored by TLC. The solution was concentrated directly without work-up. The resulting residue was dissolved in DCM (20 mL). To the solution was added ethanamine hydrochloride (218 mg, 2.67 mmol) and followed by Et3N (450 mg, 4.45 mmol) dropwise. Upon the completion of the reaction as monitored by TLC, the reaction solution was concentrated directly and purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 30% EtOAc/PE to provide 2,4-dichloro-N- ethylfuro[3,2-d]pyrimidine-6- carboxamide (160 mg, 0.62 mmol) as a yellow solid. LC-MS (ESI+): m/z 260/262 (MH+). 1HNMR (300 MHz, CDCl3) δ 7.59 (s, 1H), 6.76 (brs, 1H), 3.61-3.54 (m, 2H), 1.31 (t, J = 7.5 Hz, 3H). 1.3) Synthesis of 2-chloro-N-ethyl-4-(2,2,6,6-tetrafluoromorpholino)furo[3,2-d] pyrimidine-6- carboxamide
Figure imgf000179_0001
[0447] To a solution of 2,4-dichloro-N-ethylfuro[3,2-d]pyrimidine-6-carboxamide (200 mg, 0.77 mmol) in 1,4-dioxane/H2O (2/1, 10 mL) under N2 was added 2,2,6,6-tetrafluoromorpholine (110 mg, 0.69 mmol), Cs2CO3 (276 mg, 0.85 mmol), Pd(OAc)2 (17 mg, 0.077 mmol) and Xantphos (45 mg, 0.077 mmol). The reaction mixture was stirred at 80 °C for 1 h. Upon the completion of the reaction as monitored by TLC, the reaction was quenched with water (30 mL) and extracted with EtOAc (2 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 30% EtOAc/PE to provide 2-chloro-N-ethyl-4-(2,2,6,6-tetrafluoromorpholino)furo[3,2-d]pyrimidine-6- carboxamide (83 mg, 0.22 mmol) as a brown solid. LC-MS (ESI+): m/z 383/385 (MH+).1HNMR (300 MHz, CDCl3) δ 7.61 (s, 1H), 6.36 (brs, 1H), 4.53-4.48 (m, 4H), 3.61-3.52 (m, 2H), 1.31 (t, J = 7.5 Hz, 3H). 1.4) Synthesis of N-ethyl-4-(2,2,6,6-tetrafluoromorpholino)-2-(4-(m-tolyl)-1H-pyrazol-1- yl)furo[3,2-d]pyrimidine-6-carboxamide
Figure imgf000179_0002
[0448] To a solution of 2-chloro-6-(pyridin-3-yl)-4-(2,2,6,6-tetrafluoromorpholino)furo [3,2- d]pyrimidine (59 mg, 0.15 mmol) in DMF (1 mL) was added 4-(m-tolyl)-1H- pyrazole (29 mg, 0.19 mmol), Cs2CO3 (102 mg, 0.31 mmol) and Cu2O (2 mg, 0.015 mmol). The reaction was stirred at 110 °C for 1 h. The solution was then cooled to room temperature and concentrated directly. The resulting residue was purified by preparative TLC to provide N-ethyl-4-(2,2,6,6- tetrafluoromorpholino)-2-(4-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d]pyrimidine-6-carboxamide (11 mg, 0.022 mmol) as a brown solid. LC-MS (ESI+): m/z 505 (MH+).1HNMR (300 MHz, CDCl3) δ 8.69 (s, 1H), 8.10 (s, 1H), 7.51 (s, 1H), 7.44-7.41 (m, 2H), 7.34-7.29 (m, 1H), 7.13 (d, J = 7.5 Hz, 1H), 6.40-6.38 (m, 1H), 4.63-4.58 (m, 4H), 3.63-3.54 (m, 2H), 2.42 (s, 3H), 1.32 (t, J = 7.2 Hz, 3H). [0449] EXAMPLE 31: Compound 133 using General Synthetic Route 31:
Figure imgf000180_0002
1.1) Synthesis of 2,4-dichloro-6-iodofuro[3,2-d]pyrimidine
Figure imgf000180_0001
[0450] To a solution of 2,4-dichlorofuro[3,2-d]pyrimidine (1.0 g, 5.32 mmol) in anhydrous THF (30 mL) at -78 °C under N2 was added n-BuLi (5.33 mL, 2.5M, 13.3 mmol). After stirred at -78 °C for 1 h, to the solution was added a solution of NIS (1.44 g, 6.38 mmol) in anhydrous THF (10 mL). Upon the completion of the reaction as monitored by TLC, the reaction was quenched with water (50 mL) and extracted with DCM (3 x 50 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 5% EtOAc/PE to 10% EtOAc/PE to provide 2,4-dichloro-6-iodofuro[3,2-d]pyrimidine (800 mg, 2.55 mmol) as yellow solid. LC-MS (ESI+): m/z 315/317 (MH+).1HNMR (300 MHz, CDCl3) δ 7.21 (s, 1H). 1.2) Synthesis of 2,4-dichloro-6-(pyridin-3-yl)furo[3,2-d]pyrimidine
Figure imgf000180_0003
[0451] To a solution of 2,4-dichloro-6-iodofuro[3,2-d]pyrimidine (500 mg, 1.59 mmol) in 1,4- dioxane/H2O (2/1, 30 mL) was added pyridin-3-ylboronic acid (156 mg, 1.27 mmol), K2CO3 (567 mg, 4.1 mmol) and PdCl2(dppf) (117 mg, 0.16 mmol). The reaction was stirred at 100 °C for 1 h. Upon the completion of the reaction as monitored by TLC, the reaction was cooled to room temperature, quenched with water (30 mL) and extracted with EtOAc (4 x 10 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 30% EtOAc/PE to provide 2,4-dichloro-6-(pyridin-3- yl)furo[3,2-d]pyrimidine (295 mg, 1.11 mmol) as a brown solid. LC-MS (ESI+): m/z 266/268 (MH+).1HNMR (300 MHz, CDCl3) δ 9.21 (s, 1H), 8.78 (d, J = 3.3 Hz, 1H), 8.27 (d, J = 8.1 Hz, 1H), 7.74-7.71 (m, 1H), 7.21 (s, 1H). 1.3) Synthesis of 2-chloro-6-(pyridin-3-yl)-4-(2,2,6,6-tetrafluoromorpholino)furo [3,2- d]pyrimidine
Figure imgf000181_0001
[0452] To a solution of 2,4-dichloro-6-(pyridin-3-yl)furo[3,2-d]pyrimidine (117 mg, 0.43 mmol) in 1,4-dioxane/H2O (2/1, 10 mL) under N2 was added 2,2,6,6-tetrafluoromorpholine (63 mg, 0.39 mmol), Cs2CO3 (154 mg, 0.47 mmol), Pd(OAc)2 (9 mg, 0.04 mmol) and Xantphos (27 mg, 0.04 mmol). The reaction was stirred at 80 °C for 1 h. Upon the completion of the reaction as monitored by TLC, the reaction was cooled to room temperature, quenched with water (30 mL) and extracted with DCM (4 x 10 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 30% EtOAc/PE to provide 2-chloro-6-(pyridin-3-yl)- 4-(2,2,6,6-tetrafluoromorpholino)furo[3,2- d]pyrimidine (97 mg, 0.25 mmol) as a brown solid. LC-MS (ESI+): m/z 389/391 (MH+). 1HNMR (300 MHz, CDCl3) δ 9.11 (s, 1H), 8.74 (d, J = 3.9 Hz, 1H), 8.09 (d, J = 7.2 Hz, 1H), 7.51-7.47 (m, 1H), 7.16 (s, 1H), 4.58-4.53 (m, 4H). 1.4) Synthesis of 6-(pyridin-3-yl)-4-(2,2,6,6-tetrafluoromorpholino)-2-(4-(m-tolyl)-1H -pyrazol- 1-yl)furo[3,2-d]pyrimidine
Figure imgf000182_0001
[0453] To a solution of 2-chloro-6-(pyridin-3-yl)-4-(2,2,6,6-tetrafluoromorpholino) furo[3,2- d]pyrimidine (87 mg, 0.22 mmol) in DMF (1 mL) was added 4-(m-tolyl)- 1H-pyrazole (43 mg, 0.27 mmol), Cs2CO3 (147 mg, 0.45 mmol) and Cu2O (4 mg, 0.02 mmol). The reaction was stirred at 110 °C for 5 h. Upon the completion of the reaction as monitored by TLC, the reaction was cooled to room temperature, quenched with water (10 mL) and extracted with DCM/MeOH (15/1, 3 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by preparative TLC to provide 6-(pyridin-3-yl)-4-(2,2,6,6- tetrafluoromorpholino)-2-(4-(m-tolyl)-1H-pyrazol-1- yl)furo[3,2-d]pyrimidine (7.5 mg, 0.014 mmol) as a white solid. LC-MS (ESI+): m/z 511 (MH+).1HNMR (300 MHz, CDCl3) δ 9.14 (s, 1H), 8.74-8.71 (m, 2H), 8.13-8.11 (m, 2H), 7.52- 7.42 (m, 3H), 7.34-7.32 (m, 2H), 7.13 (d, J = 7.5 Hz, 1H), 4.67-4.62 (m, 4H), 2.42 (s, 3H). [0454] Compounds 89 to 142 in Table 3 are made according to the procedures above. Table 3
Figure imgf000182_0002
Figure imgf000183_0001
Figure imgf000184_0001
Figure imgf000185_0001
Figure imgf000186_0001
Figure imgf000187_0001
Figure imgf000188_0001
Figure imgf000189_0001
Figure imgf000190_0001
Figure imgf000191_0001
Figure imgf000192_0001
Figure imgf000193_0001
Figure imgf000194_0001
Figure imgf000195_0001
Figure imgf000196_0001
[0455] EXAMPLE 32: Compound 143 using General synthetic route 32: N N N Cl Sn N Cl I O N N N O N N Pd(PPh3)4 /CuCl/LiCl/THF N O O 1 2 N HN N N N N N O N Cu2O/Cs2CO3/DMF N O Compound 143 1) Synthesis of 2-chloro-4-morpholino-6-(pyrimidin-4-yl)furo[3,2-d]pyrimidine N Cl N N O N N O [0456] A suspension of 2-chloro-6-iodo-4-morpholinofuro[3,2-d]pyrimidine (80 mg, 0.22 mmol), 4-(tributylstannyl)pyrimidine (128 mg, 0.34 mmol), LiCl (1 mg, 0.022 mmol) and Pd(PPh3)4 (25 mg, 0.022 mmol) in DMF (5 mL) under N2 was heated to 90 °C for 3 h. The completion of the reaction was monitored by TLC. The reaction mixture was concentrated directly. The resulting residue was purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 2% MeOH/DCM to provide 2-chloro-4-morpholino-6-(pyrimidin- 4-yl)furo[3,2-d] pyrimidine (87 mg, 0.27 mmol) as a yellow solid. LC-MS (ESI+): m/z 318/320 (MH+). 2) Synthesis of 4-morpholino-6-(pyrimidin-4-yl)-2-(4-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2- d]pyrimidine
Figure imgf000197_0001
[0457] To a solution of 2-chloro-4-morpholino-6-(pyrimidin-4-yl)furo[3,2-d]pyrimidine (73 mg, 0.23 mmol) in DMF (10 mL) was added 4-(m-tolyl)-1H-pyrazole (44 mg, 0.28 mmol), Cs2CO3 (151 mg, 0.46 mmol) and Cu2O (4 mg, 0.023 mmol). The reaction mixture was stirred at 110 °C overnight. The completion of the reaction was monitored by TLC. The reaction mixture was concentrated directly. The resulting residue was purified by silica gel column chromatography with a gradient elution of 2% MeOH/DCM to 10% MeOH/DCM to provide 4- morpholino-6-(pyrimidin-4-yl)-2-(4-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d]pyrimidine (Compound 143, 28 mg, 0.064 mmol) as a white solid. LC-MS (ESI+): m/z 440 (MH+).1HNMR (300 MHz, DMSO-d6) δ 9.34 (s, 1H), 9.05 (d, J = 4.2 Hz, 2H), 8.31-8.14 (m, 2H), 7.87-7.82 (m, 1H), 7.62-7.57 (m, 2H), 7.32-7.27 (m, 1H), 7.10-7.05 (m, 1H), 4.27-4.14 (m, 4H), 3.95-3.85 (m, 4H), 2.36 (s, 3H).
EXAMPLE 33: Compound 156 using General synthetic route 33:
Figure imgf000198_0002
1) Synthesis of 2-chloro-4-morpholino-6-(pyridin-3-yl)furo[3,2-d]pyrimidine [0458] To a solution of 2-chloro
Figure imgf000198_0003
-6-iodo-4-morpholinofuro[3,2-d]pyrimidine (500 mg, 1.37 mmol) in 1,4-dioxane/H2O (2/1, 30 mL) was added pyridin-3-ylboronic acid (185 mg, 1.51 mmol), K2CO3 (378 mg, 2.74 mmol) and PdCl2(PPh3)2 (48 mg, 0.068 mmol) under N2. The reaction mixture was stirred at 90 °C for 5 h. The completion of the reaction was monitored by TLC. The solution was concentrated directly and the resulting residue was purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 3% MeOH/DCM to provide 2-chloro-4-morpholino-6-(pyridin-3-yl)furo[3,2-d]pyrimidine (410 mg, 1.29 mmol) as a light yellow solid. LC-MS (ESI+): m/z 317/319 (MH+).1HNMR (300 MHz, CDCl3) δ 9.07 (s, 1H), 8.69 (d, J = 6.9 Hz, 1H), 8.05 (d, J = 8.1 Hz, 1H), 7.71-7.40 (m, 1H), 7.08 (s, 1H), 4.15- 4.08 (m, 4H), 3.92-3.86 (m, 4H). 2) Synthesis of 4-morpholino-2-(3-phenyl-1H-pyrazol-1-yl)-6-(pyridin-3-yl)furo[3,2- d]pyrimidine
Figure imgf000198_0001
[0459] To a solution of 2-chloro-4-morpholino-6-(pyridin-3-yl)furo[3,2-d]pyrimidine (120 mg, 0.38 mmol) in DMF (10 mL) was added 3-phenyl-1H-pyrazole (60 mg, 0.42 mmol), Cs2CO3 (248 mg, 0.76 mmol) and Cu2O (6 mg, 0.038 mmol). The reaction was stirred at 110 °C overnight. The completion was monitored by TLC. The reaction mixture was quenched with water (50 mL). The aqueous solution was extracted with DCM (3 x 30 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduce pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 3% MeOH/DCM to provide 4-morpholino-2-(3-phenyl- 1H-pyrazol-1-yl)-6- (pyridin-3-yl)furo[3,2-d]pyrimidine (114 mg, 0.27 mmol) as an off-white solid. LC-MS (ESI+): m/z 425 (MH+). 1HNMR (300 MHz, CDCl3) δ 9.11 (s, 1H), 8.69 (d, J = 3.9 Hz, 1H), 8.57 (d, J = 2.7 Hz, 1H), 8.09 (d, J = 8.1 Hz, 1H), 8.01 (d, J = 6.9 Hz, 2H), 7.45- 7.32 (m, 4H), 7.30-7.26 (m, 1H), 6.79 (d, J = 2.7 Hz, 1H), 4.20-4.14 (m, 4H), 3.97-3.92 (m, 4H). EXAMPLE 34: Compounds 145 and 146 using General synthetic route 34: H I
Figure imgf000199_0001
1) Synthesis of tert-butyl 5-(2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)-3-methyl-1H- pyrazole-1-carboxylate N
Figure imgf000199_0002
[0460] A suspension of 2-chloro-6-iodo-4-morpholinofuro[3,2-d]pyrimidine (5.72 g, 15.67 mmol), (1-(tert-butoxycarbonyl)-3-methyl-1H-pyrazol-5-yl)boronic acid (3.90 g, 17.24 mmol), Pd(PPh)2Cl2 (2.20 g, 3.13 mmol) and CsF (7.15 g, 47.01 mmol) in 1,4-dioxane/H2O (4/1, 330 mL) under N2 was heated to 80 °C for 1 h. The completion of the reaction was monitored by TLC. The reaction mixture was concentrated directly and the resulting residue was purified by silica gel column chromatography with a gradient elution of 25% EtOAc/Hex to EtOAc to provide tert-butyl 5-(2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)-3-methyl-1H-pyrazole-1- carboxylate (10.32 g, 24.57 mmol) as a light yellow solid. LC-MS (ESI+): m/z 420/422 (MH+). 1HNMR (300 MHz, DMSO-d6) δ 7.31 (s, 1H), 6.94 (s, 1H), 4.01-3.90 (m, 4H), 3.78-3.70 (m, 4H), 2.21 (s, 3H), 1.44 (s, 9H). 2) Synthesis of 2-chloro-6-(3-methyl-1H-pyrazol-5-yl)-4-morpholinofuro[3,2-d]pyrimidine
Figure imgf000200_0001
[0461] To a solution of tert-butyl 5-(2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)- 3- methyl-1H-pyrazole-1-carboxylate (10.32 g, 24.63 mmol) in DCM (300 mL) was added TFA (30 mL). The mixture was stirred at rt for 2 h. The completion of the reaction was monitored by TLC. The reaction mixture was quenched with saturated NaHCO3 solution until the pH = 8. A large amount of solid was precipitated. After filtration, the filter cake was washed with ether twice to provide crude 4-(5-(benzyloxy)-2-(5-methyl-1H-pyrazol-3-yl)pyrazolo[1,5-a] pyrimidin-7-yl)morpholine (7.96 g, 24.95 mmol) as a light yellow solid. The crude product was used directly for the next step without further purification. LC-MS (ESI+): m/z 320/322 (MH+). 3) Synthesis of 2-(5-(2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)-3-methyl-1H- pyrazol-1-yl)-N,N-dimethylethan-1-amine (to Compound 146) and 2-(3-(2-chloro-4- morpholinofuro[3,2-d]pyrimidin-6-yl)-5-methyl-1H-pyrazol-1-yl)-N,N-dimethylethan-1- amine (to Compound 145)
Figure imgf000200_0002
[0462] To a solution of crude 4-(5-(benzyloxy)-2-(5-methyl-1H-pyrazol-3-yl)pyrazolo [1,5- a]pyrimidin-7-yl)morpholine (170 mg, 0.53 mmol) in DMF was added K2CO3 (220 mg, 1.60 mmol) and 2-chloro-N,N-dimethylethanamine hydrochloride (115 mg, 0.80 mmol). The mixture was stirred at 50 °C for 2 h. The completion of the reaction was monitored by TLC. The reaction mixture was quenched with water and extracted with EtOAc (3 x 10 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduce pressure. The residue was purified by preparative TLC with an elution of 10% MeOH/DCM to provide 2-(3- (2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)-5-methyl-1H- pyrazol-1-yl)-N,N- dimethylethanamine (lower spot, 90 mg, 0.23 mmol) and 2-(5-(2-chloro-4-morpholinofuro[3,2- d]pyrimidin-6-yl)-3-methyl-1H-pyrazol-1-yl)-N,N-dimethylethanamine (upper spot, 60 mg, 0.15 mmol). [0463] 2-(3-(2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)-5-methyl-1H-pyrazol-1-yl)- N,N-dimethylethanamine (lower spot): LC-MS (ESI+): m/z 391/393 (MH+) 1HNMR (300 MHz, DMSO-d6) δ 7.06 (s, 1H), 6.65 (s, 1H), 4.23 (t, J = 6.3 Hz, 2H), 3.96-3.88 (m, 4H), 3.82-3.75 (m, 4H), 2.71 (t, J = 6.0 Hz, 2H), 2.35 (s, 3H), 2.24 (s, 6H). 2-(5-(2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)-3-methyl-1H-pyrazol-1-yl)-N,N- dimethylethanamine (upper spot): LC-MS (ESI+): m/z 391/393 (MH+) 1HNMR (300 MHz, DMSO-d6) δ 7.32 (s, 1H), 6.73 (s, 1H), 4.43 (t, J = 6.9 Hz, 2H), 3.97-3.88 (m, 4H), 3.82-3.73 (m, 4H), 2.68 (t, J = 6.9 Hz, 2H), 2.21 (s, 3H), 2.18 (s, 6H). 4) Synthesis of N,N-dimethyl-2-(5-methyl-3-(4-morpholino-2-(4-(m-tolyl)-1H-pyrazol-1- yl)furo[3,2-d]pyrimidin-6-yl)-1H-pyrazol-1-yl)ethanamine (Compound 146)
Figure imgf000201_0001
[0464] A suspension of 2-(3-(2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)-5-methyl-1H- pyrazol-1-yl)-N,N-dimethylethanamine (50 mg, 0.13 mmol), 4-(m-tolyl)-1H-pyrazole (24 mg, 0.15 mmol), Cs2CO3 (83 mg, 0.26 mmol) and Cu2O (1.8 mg, 0.01 mmol) in DMF (5 mL) was heated to 110 °C overnight. The completion of the reaction was monitored by TLC. The reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 5% MeOH/DCM to 10% MeOH/DCM to provide N,N-dimethyl-2-(5- methyl-3-(4-morpholino-2-(4-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d]pyrimidin-6-yl)-1H-pyrazol- 1-yl)ethanamine (20 mg, 0.04 mmol) as a white solid. LC-MS (ESI+): m/z 513 (MH+).1HNMR (300 MHz, CDCl3) δ 8.72 (s, 1H), 8.07 (s, 1H), 7.61-7.40 (m, 2H), 7.35-7.27 (m, 1H), 7.11-7.06 (m, 2H), 6.44 (s, 1H), 4.40-4.37 (m, 2H), 4.16-4.10 (m, 4H), 3.93-3.88 (m, 4H), 3.10-2.98 (m, 2H), 2.45 (m, 3H), 2.41 (s, 9H). 5) Synthesis of N,N-dimethyl-2-[3-methyl-5-[4-morpholino-2-[4-(m-tolyl)pyrazol-1- yl]furo[3,2-d]pyrimidin-6-yl]pyrazol-1-yl]ethanamine (Compound 145)
Figure imgf000202_0001
[0465] Compound 145 was prepared by the same method used for Compound 146. [0466] EXAMPLE 35: Compound 148 using General synthetic route 35:
Figure imgf000202_0002
1) Synthesis of 2,4-dichloro-6-(pyridin-2-yl)furo[3,2-d]pyrimidine
Figure imgf000202_0003
[0467] To a solution of 2,4-dichloro-6-iodofuro[3,2-d]pyrimidine (2.3 g, 7.3 mmol) in DMF (60 mL) under N2 was added 2-(tributylstannyl)pyridine (2.7 g, 7.3 mmol), CuI (416 mg, 2.2 mmol) and PdCl2(dppf) (534 mg, 0.73 mmol). The reaction was stirred at 100 °C for 3 h. The completion of the reaction was monitored by TLC. The reaction mixture was quenched with water (30 mL) and extracted with DCM (4 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduce pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 30% EtOAc/PE to provide 2,4-dichloro-6-(pyridin-2-yl)furo[3,2-d]pyrimidine (1.2 g, 4.53 mmol) as a yellow solid. LC-MS (ESI+): m/z 266/268 (MH+). 1HNMR (300 MHz, CDCl3) δ 8.78 (d, J = 4.2 Hz, 1H), 8.09 (d, J = 7.8 Hz, 1H), 7.92 (t, J = 7.8 Hz, 1H), 7.56 (s, 1H), 7.47- 7.41 (m, 1H). 2) Synthesis of 2-chloro-4-methoxy-6-(pyridin-2-yl)furo[3,2-d]pyrimidine
Figure imgf000203_0001
[0468] To a solution of 2,4-dichloro-6-(pyridin-2-yl)furo[3,2-d]pyrimidine (500 mg, 1.89 mmol) in DMF/MeOH (1:1, 30 mL) at 0 °C was added sodium methanolate (204 mg, 3.78 mmol). The reaction was stirred at 0 °C for 3 h. The completion of the reaction was monitored by TLC. The reaction mixture was quenched with water (30 mL) and a large amount of solid was precipitated. After filtration, the filter cake was washed with Et2O to provide 2-chloro-4- methoxy-6-(pyridin-2-yl)furo[3,2-d]pyrimidine (450 mg, 1.72 mmol) as a brown solid. LC-MS (ESI+): m/z 262/264 (MH+).1HNMR (300 MHz, CDCl3) 1HNMR (300 MHz, CDCl3) δ 8.78 (d, J = 4.2 Hz, 1H), 8.09 (d, J = 7.8 Hz, 1H), 7.85 (t, J = 7.8 Hz, 1H), 7.53 (s, 1H), 7.39-7.35 (m, 1H), 4.23 (s, 3H). 3) Synthesis of 2-bromo-6-(pyridin-2-yl)furo[3,2-d]pyrimidin-4-ol [0469] A solution of 2-chloro-4
Figure imgf000203_0002
-methoxy-6-(pyridin-2-yl)furo[3,2-d]pyrimidine (200 mg, 0.76 mmol) in HBr/AcOH (33 wt.% in Acetic acid, 10 mL) was heated to refluxed for 2 h. The completion of the reaction was monitored by LC-MS. The reaction mixture was quenched with water (30 mL) and a large amount of solid was precipitated. After filtration, the filter cake was washed with Et2O to provide 292 mg of crude 2-bromo-6-(pyridin-2-yl)furo[3,2-d]pyrimidin-4- ol as a yellow solid. The crude product was used directly for the next step without further purification. LC-MS (ESI+): m/z 292/294 (MH+). 4) Synthesis of 6-(pyridin-2-yl)-2-(3-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d]pyrimidin-4-ol
Figure imgf000204_0003
[0470] A suspension of crude 2-bromo-6-(pyridin-2-yl)furo[3,2-d]pyrimidin-4-ol (200 mg, 0.68 mmol), 3-(m-tolyl)-1H-pyrazole (108 mg, 0.68 mmol), Cs2CO3 (447 mg, 1.37 mmol) and Cu2O (10 mg, 0.068 mmol) in DMF (10 mL) was heated to 110 °C overnight. The completion of the reaction was monitored by TLC. The reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 5% MeOH/DCM to 10% MeOH/DCM to provide 6-(pyridin-2-yl)-2-(3-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d]pyrimidin- 4-ol (100 mg, 0.27 mmol) as a green solid. LC-MS (ESI+): m/z 370 (MH+). 5) Synthesis of 4-chloro-6-(pyridin-2-yl)-2-(3-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2- d]pyrimidine
Figure imgf000204_0001
[0471] A solution of 6-(pyridin-2-yl)-2-(3-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d]pyrimidin-4- ol (114 mg, 0.31 mmol) in phenylphosphonic dichloride (5 mL) was heated to 120 °C for 2 h. The completion of the reaction was monitored by TLC. The reaction mixture was quenched with water (20 mL) and a large amount of yellow solid was precipitated. After filtration, the filter cake was washed with Et2O to provide 80 mg of crude 4-chloro-6-(pyridin-2-yl)-2-(3-(m-tolyl)- 1H-pyrazol-1-yl)furo[3,2-d]pyrimidine as a yellow solid. The crude product was used directly for the next step without further purification. LC-MS (ESI+): m/z 388/390 (MH+). 6) Synthesis of 4-(6-(pyridin-2-yl)-2-(4-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d]pyrimidin-4- yl)morpholin-3-one N
Figure imgf000204_0002
[0472] A suspension of 4-chloro-6-(pyridin-2-yl)-2-(3-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2- d]pyrimidine (40 mg, 0.1 mmol), morpholin-3-one (12.5 mg, 0.12 mmol), Pd(PPh)2Cl2 (7.2 mg, 0.01 mmol), Cs2CO3 (78 mg, 0.2 mmol) and Xantphos (12 mg, 0.02 mmol) in 1,4-dioxane (10 mL) under N2 was heated to 90 °C for 1 h. The completion of the reaction was monitored by TLC. The reaction mixture was quenched with water and extracted with EtOAc (3 x 10 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by preparative TLC with a elution of 10% MeOH/DCM to provide 4-(6-(pyridin-2-yl)-2-(4-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d] pyrimidin-4-yl)morpholin-3-one (5 mg, 0.011mmol) as a white solid. LC-MS (ESI+): m/z 453 (MH+).1HNMR (300 MHz, CDCl3) δ 8.75 (d, J = 3.9 Hz, 1H), 8.62 (s, 1H), 8.02 (d, J = 7.8 Hz, 1H), 7.96-7.89 (m, 2H), 7.76 (d, J = 7.5 Hz, 1H), 7.67 (s, 1H), 7.48-7.31 (m, 2H), 7.20-7.15 (m, 1H), 6.82 (s, 1H), 4.53 (s, 2H), 4.32-4.24 (m, 2H), 4.19-4.10 (m, 2H), 2.43 (s, 3H). [0473] EXAMPLE 36: Compound 150 using General synthetic route 36:
Figure imgf000205_0001
1) Synthesis of (2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)methanol
Figure imgf000205_0002
[0474] To a solution of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid (1 g, 3.53 mmol) in THF (10 mL) at 0 °C was added BH3/THF (1 mol/L, 14 mL) dropwise. The reaction mixture was stirred at rt overnight. The completion of the reaction was monitored by TLC. The reaction was quenched with 1N HCl. The mixture was heated under reflux for 2h. The reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 3% MeOH/DCM to provide (2-chloro-4- morpholinofuro[3,2-d]pyrimidin-6-yl)methanol (165 mg, 0.61 mmol) as a white solid. LC-MS (ESI+): m/z 270 (MH+). 2) Synthesis of (4-morpholino-2-(4-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d]pyrimidin-6- yl)methanol
Figure imgf000206_0001
[0475] To a solution of (2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)methanol (250 mg, 0.93 mmol) in 1,4-dioxane (20 mL) was added 4-(m-tolyl)-1H-pyrazole (176 mg, 0.42 mmol), Pd2(dba)3 (85 mg, 0.093 mmol), t-Buxphos and K3PO4 (900 mg, 3.72 mmol). The reaction mixture was stirred at 90 °C overnight. The completion of the reaction was monitored by TLC. The reaction mixture was quenched with water (50 mL). The aqueous solution was extracted with DCM (3 x 30 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduce pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 3% MeOH/DCM to 8% MeOH/DCM to provide (4-morpholino-2-(4-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d]pyrimidin-6-yl)methanol (120 mg, 0.31 mmol) as an off-white solid. LC-MS (ESI+): m/z 392 (MH+). 3) Synthesis of 4-morpholino-2-(4-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d]pyrimidine-6- carbaldehyde N
Figure imgf000206_0002
[0476] To a solution of (4-morpholino-2-(4-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d]pyrimidin- 6-yl) methanol (60 mg, 0.15 mmol) in DCM at rt (10 mL) was added Dess-Martin periodinane (DMP) (120 mg, 0.28 mmol). The reaction was stirred at rt for 2 h. The completion of the reaction was monitored by TLC. The reaction was quenched with saturated NaHCO3 solution. The aqueous solution was extracted with MeOH/DCM (1/10, 3 x 10 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduce pressure. The residue was purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 3% MeOH/DCM to provide 4-morpholino-2-(4-(m-tolyl)-1H-pyrazol-1- yl)furo[3,2-d]pyrimidine-6- carbaldehyde (32 mg, 0.08 mmol) as a light yellow solid. LC-MS (ESI+): m/z 390 (MH+).1HNMR (300 MHz, CDCl3) δ 9.94 (s, 1H), 8.69 (s, 1H), 8.09 (s, 1H), 7.61 (s, 1H), 7.48-7.40 (m, 2H), 7.35-7.26 (m, 2H), 7.11 (d, J = 7.8 Hz, 1H), 4.25-4.15 (m, 4H), 3.95-3.85 (m, 4H), 2.41 (s, 3H). 4) Synthesis of 4-morpholino-2-(4-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d]pyrimidine-6- carboxylic acid
Figure imgf000207_0001
[0477] To a solution of 4-morpholino-2-(4-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d]pyrimidine- 6-carbaldehyde (32 mg, 0.08 mmol) in i-PrOH/H2O (4:1, 5 mL) at 0 °C was added NaH2PO4.H2O (64.2 mg, 0.41 mmol) and NaClO2 (37 mg, 0.41 mmol) in portions. The reaction was stirred at rt for 1 h. The completion of the reaction was monitored by TLC. The reaction was quenched with a 1N HCl solution. The aqueous solution was extracted with MeOH/DCM (1/10, 3 x 10 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated to provide crude 4-morpholino-2-(4-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d] pyrimidine-6-carboxylic acid (38 mg, 0.09 mmol) as a white solid. LC-MS (ESI+): m/z 406 (MH+) 5) Synthesis of N-(2-(methylsulfonyl)ethyl)-4-morpholino-2-(4-(m-tolyl)-1H-pyrazol-1- yl)furo[3,2-d]pyrimidine-6-carboxamide N
Figure imgf000207_0002
[0478] A solution of 4-morpholino-2-(4-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d]pyrimidine-6- carboxylic acid (38 mg, 0.09 mmol), 2-(methylsulfonyl)ethanamine (16 mg, 0.10 mmol), EDCl (37.8 mg, 0.20 mmol) and HOBT (26 mg, 0.20 mmol) in DCM was stirred at rt for 2 h. The completion of the reaction was monitored by TLC. The reaction mixture was concentrated directly and purified by silica gel column chromatography with a gradient elution of 2% MeOH/DCM to 5% MeOH/DCM to provide N-(2-(methylsulfonyl)ethyl)-4-morpholino-2-(4- (m-tolyl)-1H-pyrazol-1-yl) furo[3,2-d]pyrimidine-6-carboxamide (14 mg, 0.027 mmol) as a white solid. LC-MS (ESI+): m/z 511 (MH+).1HNMR (300 MHz, CDCl3) δ 8.70 (s, 1H), 8.07 (s, 1H), 7.67-7.62 (m, 1H), 7.53 (s, 1H), 7.48-7.40 (m, 2H), 7.35-7.26 (m, 1H), 7.12-7.09 (m, 1H), 4.15-4.05 (m, 6H), 3.92-3.85 (m, 4H), 3.34-3.07 (m, 2H), 3.04 (s, 3H), 2.42 (s, 3H). [0479] EXAMPLE 37: Compound 192 using General synthetic route 37:
Figure imgf000208_0001
1)Synthesis of tert-butyl 4-(3-chlorophenyl)-1H-pyrazole-1-carboxylate
Figure imgf000208_0002
[0480] To a solution of tert-butyl 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole -1-carboxylate (1 g, 4.20 mmol) in 1,4-dioxane/H2O (10/1, 20 mL) was added 1-chloro-3- iodobenzene (1.24 g, 1.51 mmol), CsF (958 mg, 6.30 mmol) and PdCl2(PPh3)2 (295 mg, 0.42 mmol) under N2. The reaction was stirred at 80 °C for 2 h. The completion of the reaction was monitored by TLC. The reaction was quenched with water. The aqueous solution was extracted with DCM (3 x 80 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduce pressure. The residue was purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 30% EtOAc/PE to provide tert- butyl 4-(3-chlorophenyl)-1H-pyrazole-1-carboxylate (650 mg, 2.34 mmol) as a light yellow oil. LC-MS (ESI+): m/z 279/281 (MH+). 2) Synthesis of 4-(3-chlorophenyl)-1H-pyrazole
Figure imgf000209_0001
[0481] To a solution of tert-butyl 4-(3-chlorophenyl)-1H-pyrazole-1-carboxylate (650 mg, 2.34 mmol) in DCM (10 mL) was added TFA (2 mL). The mixture was stirred at rt for 2 h. The completion of the reaction was monitored by TLC. The reaction mixture was quenched with saturated NaHCO3 solution until the pH = 8. The aqueous solution was extracted with DCM (3 x 80 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduce pressure. The residue was purified by silica gel column chromatography with a gradient elution of 20% EtOAc/PE to 50% EtOAc/PE to provide 4-(3- chlorophenyl)-1H-pyrazole (130 mg, 0.73 mmol) as a light yellow solid. LC-MS (ESI+): m/z 179/181 (MH+).1HNMR (300 MHz, CDCl3) δ 7.87 (brs, 2H), 7.46 (s, 1H), 7.39 (d, J = 7.8 Hz, 1H), 7.30-7.28 (m, 1H), 7.23-7.20 (m, 1H). 3) Synthesis of 2-(4-(3-chlorophenyl)-1H-pyrazol-1-yl)-6-(1-methyl-1H-pyrazol-3-yl)-4- morpholinofuro[3,2-d]pyrimidine
Figure imgf000209_0002
[0482] To a solution of 2-chloro-6-(1-methyl-1H-pyrazol-3-yl)-4-morpholinofuro[3,2- d]pyrimidine (60 mg, 0.19 mmol) in DMF (10 mL) was added 4-(3-chlorophenyl)-1H-pyrazole (40 mg, 0.19 mmol), Cs2CO3 (221 mg, 0.68 mmol) and Cu2O (5 mg, 0.04 mmol). The reaction was stirred at 110 °C overnight. The completion of the reaction was monitored by TLC. The reaction mixture was quenched with water (50 mL). The aqueous solution was extracted with DCM (3 x 30 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduce pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 3% MeOH/DCM to provide 2- (4-(3-chlorophenyl)-1H-pyrazol-1-yl)-6-(1-methyl-1H-pyrazol-3-yl)-4-morpholinofuro[3,2- d]pyrimidine (22 mg, 0.048 mmol) as an off-white solid. LC-MS (ESI+): m/z 462/464 (MH+). 1HNMR (300 MHz, CDCl3) δ 8.75 (s, 1H), 8.06 (s, 1H), 7.62 (s, 1H), 7.50-7.40 (m, 2H), 7.26- 7.20 (m, 2H), 7.11 (s, 1H), 6.66 (d, J = 2.4 Hz, 1H), 4.21-4.15 (m, 4H), 4.02 (s, 3H), 3.95-3.89 (m, 4H). [0483] EXAMPLE 38: Compound 157 using General synthetic route 38:
Figure imgf000210_0001
1)Synthesis of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid
Figure imgf000210_0002
[0484] To a solution of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine (2.4 g, 10 mmol) in anhydrous THF (40 mL) at -78 °C under N2 was added n-BuLi (5.2 mL, 2.5 M, 13 mmol) dropwise. The reaction mixture was stirred at that temperature for 1 h. To the above solution was added dry ice (4.4 g, 100 mmol) in one potion. The resulting reaction mixture was stirred at that temperature for 3 h. The completion of the reaction was monitored by TLC. The reaction was quenched with water and the pH was adjusted to 5 using 1 N HCl solution. The aqueous solution was extracted with DCM (3 x 80 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduce pressure. The resulting residue was slurry in Et2O to provide 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid ( 2.92 g, 10.3 mmol) as a yellow solid. LC-MS (ESI+): m/z 284/286 (MH+).1HNMR (300 MHz, CDCl3) δ 7.58 (s, 1H), 4.04-3.95 (m, 4H), 3.78-3.76 (m, 4H). 2) Synthesis of 2-chloro-N-(1-methylpiperidin-4-yl)-4-morpholinofuro[3,2-d]pyrimidine-6- carboxamide
Figure imgf000211_0001
[0485] To a solution of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine-6-carboxylic acid (400 mg, 1.42 mmol) in DCM was added oxalyl dichloride (360 mg, 2.84 mmol) and one drop of DMF. The mixture was stirred at rt for 2 h. The solution was concentrated and the resulting residue was dissolved in DCM (15 mL). To the solution was added 1-methylpiperidin-4-amine (178 mg, 1.56 mmol) and followed by DIEA (107 mg, 2.84 mmol). The completion of the reaction was monitored by TLC. The reaction mixture was quenched with water and extracted with DCM (3 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduce pressure. The residue was purified by silica gel column chromatography with a gradient elution of 5% MeOH/DCM to 10% MeOH/DCM to provide 2- chloro-N-(1-methylpiperidin-4-yl)-4-morpholinofuro[3,2-d]pyrimidine-6-carboxamide (320 mg, 0.84 mmol) as a yellow solid. LC-MS (ESI+): m/z 380/382 (MH+). 3) Synthesis of N-(1-methylpiperidin-4-yl)-4-morpholino-2-(4-phenyl-1H-pyrazol-1- yl)furo[3,2-d]pyrimidine-6-carboxamide N
Figure imgf000211_0002
[0486] To a solution of 2-chloro-N-(1-methylpiperidin-4-yl)-4-morpholinofuro[3,2- d]pyrimidine- 6-carboxamide (100 mg, 0.26 mmol) in DMF (4 mL) was added 4-phenyl-1H- pyrazole (42 mg, 0.29 mmol), Cs2CO3 (172 mg, 0.53 mmol) and Cu2O (4 mg, 0.026 mmol). The reaction was stirred at 110 °C overnight. The reaction mixture was quenched with water (10 mL). The aqueous solution was extracted with DCM (3 x 10 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated. The resulting residue was purified by preparative HPLC to provide N-(1-methylpiperidin-4-yl)-4-morpholino-2-(4-phenyl-1H- pyrazol-1-yl)furo[3,2-d]pyrimidine-6-carboxamide (32 mg, 0.065 mmol) as a white solid. LC- MS (ESI+): m/z 488 (MH+).1HNMR (300 MHz, CDCl3) δ 8.71 (s, 1H), 8.08 (s, 1H), 7.61 (d, J = 7.5 Hz, 2H), 7.44 (s, 1H), 7.38-7.26 (m, 3H), 6.33 (d, J = 7.8 Hz, 1H), 4.21-4.03 (m, 5H), 3.94-3.88 (m, 4H), 2.99-2.88 (m, 2H), 2.40 (s, 3H), 2.33-2.22 (m, 2H), 2.19-2.05 (m, 2H), 1.87- 1.75 (m, 2H). [0487] EXAMPLE 39: Compound 179 using General synthetic route 39:
Figure imgf000212_0001
1)Synthesis of 2-chloro-6-iodo-4-morpholinofuro[3,2-d]pyrimidine
Figure imgf000212_0002
[0488] To a solution of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine (2.0 g, 0.83 mmol) in THF (30 mL) at -78 °C under N2 was added LDA (1.33 mL, 2M, 2.66 mmol). After stirred at - 78 °C for 1 h, to the solution was added a solution of NIS (2.25 g, 1.0 mmol) in THF (10 mL). The completion of the reaction was monitored by TLC. The reaction mixture was quenched with water (50 mL). The aqueous solution was extracted with DCM (3 x 50 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduce pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 5% EtOAc/PE to 10% EtOAc/PE to provide 2-chloro-6-iodo-4- morpholinofuro[3,2-d]pyrimidine (1.6 g, 4.4 mmol) as a yellow solid. LC-MS (ESI+): m/z 366/368 (MH+).1HNMR (300 MHz, CDCl3) δ 6.97 (s, 1H), 4.01-3.98 (m, 4H), 3.85-3.82 (m, 4H). 2) Synthesis of 2-chloro-7-iodo-4-morpholinofuro[3,2-d]pyrimidine
Figure imgf000213_0001
[0489] To a solution of 2-chloro-6-iodo-4-morpholinofuro[3,2-d]pyrimidine (5.0 g, 13.7 mmol) in THF (30 mL) at -78 °C under N2 was added LDA (14 mL, 2M, 27.4 mmol). After addition, the reaction mixture was stirred at -78 °C for 1 h. The completion of the reaction was mornitored by TLC. The reaction mixture was quenched with H2O (100 mL). The aqueous solution was extracted with DCM (3 x 200 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduce pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 30% EtOAc/PE to provide 2-chloro-7-iodo-4-morpholinofuro[3,2-d]pyrimidine (2.5 g, 6.85 mmol) as a white solid. LC-MS (ESI+): m/z 366/368 (MH+).1HNMR (300 MHz, CDCl3) δ 7.77 (s, 1H), 4.04-3.94 (m, 4H), 3.85-3.81 (m, 4H). 3) Synthesis of 2-chloro-7-methyl-4-morpholinofuro[3,2-d]pyrimidine [0490] To a solution of 2-chloro-7-i
Figure imgf000213_0002
odo-4-morpholinofuro[3,2-d]pyrimidine (500 mg, 0.14 mmol) in DME/H2O (2/1, 30 mL) was added methylboronic acid (250 mg, 4.2 mmol), K3PO4 (440 mg, 2.1 mmol) and Pd(PPh3)4 (150 mg, 0.14 mmol). The reaction was stirred at 120 °C for overnight. The completion of the reaction was monitored by TLC. The reaction mixture was quenched with water (50 mL). The aqueous solution was extracted with EtOAc (2 x 50 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated. The resulting residue was purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 30% EtOAc/PE to provide 2-chloro-7-methyl-4-morpholinofuro[3,2- d]pyrimidine (350 mg, 1.4 mmol) as a white solid. LC-MS (ESI+): m/z 254/256 (MH+). 4) Synthesis of 2-chloro-6-iodo-7-methyl-4-morpholinofuro[3,2-d]pyrimidine
Figure imgf000214_0001
[0491] To a solution of 2-chloro-7-methyl-4-morpholinofuro[3,2-d]pyrimidine (350 mg, 1.38 mmol) in THF (30 mL) at -78 °C under N2 was added LDA (1.4 mL, 2 M, 2.76 mmol). After stirred at -78 °C for 1 h, to the solution was added a solution of NIS (374 mg, 1.66 mmol) in THF (5 mL). The completion of the reaction was monitored by TLC. The reaction mixture was quenched with water (50 mL). The aqueous solution was extracted with EtOAc (3 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduce pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 30% EtOAc/PE to provide 2-chloro-6-iodo-7-methyl-4- morpholinofuro[3,2-d]pyrimidine (280 mg, 2.55 mmol) as yellow solid. LC-MS (ESI+): m/z 380/382 (MH+). 5) Synthesis of 2-chloro-7-methyl-4-morpholino-6-(pyridin-2-yl)furo[3,2-d]pyrimidine
Figure imgf000214_0002
[0492] A solution of 2-chloro-6-iodo-7-methyl-4-morpholinofuro[3,2-d]pyrimidine (200 mg, 0.53 mmol), 2-(tributylstannyl)pyridine (389 mg, 1.06 mmol) and Pd(PPh3)4 (61 mg, 0.053 mmol) in toluene (5 mL) was heated to 90 °C overnight. The completion of the reaction was monitored by TLC. The reaction mixture was diluted with water and extracted with DCM/MeOH (15/1, 3 x 50 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduce pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 30% EtOAc/PE to 50% EtOAc/PE to provide 2-chloro-7-methyl-4-morpholino-6-(pyridin-2-yl)furo[3,2-d]pyrimidine (70 mg, 0.21 mmol) as a white solid. LC-MS (ESI+): m/z 331/333 (MH+).1HNMR (300 MHz, CDCl3) δ 8.67 (d, J = 4.8 Hz, 1H), 7.78-7.67 (m, 2H), 7.31-7.25 (m, 1H), 4.06-4.01 (m, 4H), 3.82-3.79 (m, 4H), 2.59 (s, 3H). 6) Synthesis of 7-methyl-4-morpholino-2-(3-phenyl-1H-pyrazol-1-yl)-6-(pyridin-2- yl)furo[3,2-d]pyrimidine
Figure imgf000215_0001
[0493] To a solution of 2-chloro-7-methyl-4-morpholino-6-(pyridin-2-yl)furo[3,2- d]pyrimidine (80 mg, 0.24 mmol) in DMF (10 mL) was added 3-phenyl-1H-pyrazole (42 mg, 0.29 mmol) and Cs2CO3 (160 mg, 0.49 mmol). The reaction was stirred at 100 °C overnight. The completion of the reaction was monitored by TLC. The reaction mixture was quenched with water (10 mL). The aqueous solution was extracted with EtOAc (3 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated. The resulting residue was purified by silica gel column chromatography with a gradient elution of 2% MeOH/DCM to 5% MeOH/DCM to provide 7-methyl-4-morpholino-2-(3-phenyl-1H-pyrazol-1- yl)-6-(pyridin-2-yl) furo[3,2-d]pyrimidine (28.6 mg, 0.065 mmol) as white solid. LC-MS (ESI+): m/z 439 (MH+).1HNMR (300 MHz, CDCl3) δ 8.76 (d, J = 1.5 Hz, 1H), 8.62 (d, J = 2.7 Hz, 1H), 8.02 (d, J = 7.2 Hz, 2H), 7.83-7.81 (m, 2H), 7.46-7.41 (m, 2H), 7.37-7.27 (m, 2H), 6.79 (d, J = 2.7 Hz, 1H), 4.21-4.15 (m, 4H), 3.96-3.90 (m, 4H), 2.77 (s, 3H). [0494] EXAMPLE 40: Compound 147 using General synthetic route 40:
Figure imgf000215_0002
1)Synthesis of 2-chloro-4-morpholino-7-(trifluoromethyl)furo[3,2-d]pyrimidine
Figure imgf000216_0002
[0495] To a solution of 2-chloro-7-iodo-4-morpholinofuro[3,2-d]pyrimidine (300 mg, 0.82 mmol) in anhydrous DMSO (6 mL) was added KF (144 mg, 2.47 mmol), CuI (30 mg, 0.16 mmol), 1,10-phenanthroline (30 mg, 0.16 mmol), B(OMe)3 (252 mg, 2.47 mmol) and TMSCF3 (348 mg, 2.47 mmol). The reaction mixture was stirred at 50 °C for 2 h. The reaction mixture was quenched with water (10 mL). The aqueous solution was extracted with DCM (3 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduce pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 30% EtOAc/PE to provide 2-chloro-4-morpholino-7- (trifluoromethyl)furo[3,2-d]pyrimidine (98 mg, 0.32 mmol) as a yellow solid. LC-MS (ESI+): m/z 308/310 (MH+). 2) Synthesis of 2-chloro-4-morpholino-7-(trifluoromethyl)furo[3,2-d]pyrimidine-6- carboxylic acid F
Figure imgf000216_0001
[0496] To a solution of 2-chloro-4-morpholino-7-(trifluoromethyl)furo[3,2-d]pyrimidine (490 mg, 1.60 mmol) in anhydrous THF (100 mL) at -78 °C under N2 was added n-BuLi (0.96 mL, 2.5 M, 2.4 mmol) dropwise. The reaction mixture was stirred at that temperature for 1 h. To the above solution was added dry ice (705 mg, 16 mmol) in one potion. The resulting reaction mixture was stirred at that temperature for 1 h. The completion of the reaction was monitored by TLC. The reaction was quenched with water and the pH was adjusted to 5 using 1 N HCl solution. The aqueous solution was extracted with DCM (2 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduce pressure. The resulting residue was slurry in Et2O to provide 2-chloro-4-morpholino-7- (trifluoromethyl)furo[3,2-d]pyrimidine-6-carboxylic acid (270 mg, 0.77 mmol) as a brown solid. LC-MS (ESI+): m/z 352/354 (MH+). 3) Synthesis of 2-chloro-N-cyclopropyl-4-morpholino-7-(trifluoromethyl)furo[3,2- d]pyrimidine-6-carboxamide
Figure imgf000217_0001
[0497] To a solution of 2-chloro-4-morpholino-7-(trifluoromethyl)furo[3,2-d]pyrimidine-6- carboxylic acid (270 mg, 0.77 mmol) in DCM (10 mL) was added cyclopropanamine (43 mg, 0.77 mmol), EDCl (175 mg, 0.92 mmol) and DMAP (112 mg, 0.92 mmol). The reaction was stirred at rt overnight. The completion of the reaction was monitored by TLC. The reaction was quenched with water (10 mL) and extracted with DCM (3 x 10 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduced pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 30% EtOAc/PE to provide 2-chloro-N-cyclopropyl-4-morpholino-7- (trifluoromethyl)furo[3,2-d]pyrimidine-6-carboxamide (60 mg, 0.15 mmol) as white solid. LC- MS (ESI+): m/z 391/393 (MH+). 4) Synthesis of N-cyclopropyl-4-morpholino-2-(4-(m-tolyl)-1H-pyrazol-1-yl)-7- (trifluoromethyl)furo[3,2-d]pyrimidine-6-carboxamide
Figure imgf000217_0002
[0498] To a solution of 2-chloro-N-cyclopropyl-4-morpholino-7-(trifluoromethyl)furo[3,2- d]pyrimidine -6-carboxamide (60 mg, 0.15 mmol) in 1,4-dioxane (2 mL) was added 4-(m-tolyl)- 1H-pyrazole (30 mg, 0.18 mmol), Pd2(dba)3 (15 mg, 0.015 mmol), t-Buxphos (15 mg, 0.015 mmol) and K3PO4 (50 mg, 0.63 mmol). The reaction was stirred at 80 °C under microwave for 30 min. The reaction mixture was diluted with water and extracted with DCM (3 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduce pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 2% MeOH/DCM to provide N-cyclopropyl-4- morpholino-2-(4-(m-tolyl)-1H-pyrazol-1-yl)-7-(trifluoromethyl)furo[3,2-d]pyrimidine-6- carboxamide (13 mg, 0.025 mmol) as a yellow solid. LC-MS (ESI+): m/z 513 (MH+).1HNMR (300 MHz, CDCl3) δ 8.77 (s, 1H), 8.08 (s, 1H), 7.61-7.42 (m, 2H), 7.35-7.27 (m, 1H), 7.11-7.09 (m, 1H), 6.48 (s, 1H), 4.17-4.12 (m, 4H), 3.92-3.88 (m, 4H), 2.95-2.90 (m, 1H), 2.41 (s, 3H), 0.98-0.93 (m, 2H), 0.75-0.69 (m, 2H). [0499] EXAMPLE 41: Compound 204 using General synthetic route 41:
Figure imgf000218_0001
1) Synthesis of 4-morpholino-2-(3-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d]pyrimidine
Figure imgf000218_0002
[0500] To a solution of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine (300 mg, 1.25 mmol) in DMF (4 mL) was added 3-(m-tolyl)-1H-pyrazole (237 mg, 0.35 mmol), Cs2CO3 (815 mg, 2.5 mmol) and CuI (24 mg, 0.125 mmol). The reaction mixture was stirred at 110 °C overnight. The reaction mixture was quenched with water (10 mL). The aqueous solution was extracted with EtOAc (3 x 10 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduce pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 10% EtOAc/PE to 30% EtOAc/PE to provide 4- morpholino-2-(3-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d]pyrimidine (269 mg, 0.75 mmol) as a white solid. LC-MS (ESI+): m/z 362 (MH+).1HNMR (300 MHz, DMSO-d6) δ 8.75 (d, J = 2.7 Hz, 1H), 8.38 (d, J = 1.8 Hz, 1H), 7.82 (s, 1H), 7.77 (d, J = 7.8 Hz, 1H), 7.39-7.34 (m, 1H), 7.21 (d, J = 7.8 Hz, 1H), 7.13 (d, J = 1.8 Hz, 1H), 7.07 (d, J = 2.7 Hz, 1H), 4.08-4.07 (m, 4H), 3.82- 3.81 (m, 4H), 2.35 (s, 3H). 2) Synthesis of 6-iodo-4-morpholino-2-(3-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d]pyrimidine
Figure imgf000219_0002
[0501] To a solution of 4-morpholino-2-(3-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d]pyrimidine (180 mg, 0.5 mmol) in anhydrous THF (40 mL) at -78 °C under N2 was added n-BuLi (0.24 mL, 2.5 M, 0.6 mmol) dropwise. The reaction mixture was stirred at that temperature for 1 h. To the solution was added a solution of NIS (135 mg, 0.6 mmol) in THF (3 mL) dropwise. The resulting reaction mixture was stirred at that temperature for 2 h. The completion of the reaction was monitored by TLC. The reaction was quenched with water. The aqueous solution was extracted with DCM (3 x 20 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduce pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 20% EtOAc/PE to 50% EtOAc/PE to provide 6-iodo-4-morpholino-2-(3-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d]pyrimidine (70 mg, 0.14 mmol) as a white solid. LC-MS (ESI+): m/z 488 (MH+). 3) Synthesis of 6-(3-methylisoxazol-5-yl)-4-morpholino-2-(3-(m-tolyl)-1H-pyrazol-1- yl)furo[3,2-d]pyrimidine
Figure imgf000219_0001
[0502] A suspension of 6-iodo-4-morpholino-2-(3-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2- d]pyrimidine (70 mg, 0.14 mmol), 3-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)isoxazole (36 mg, 0.172 mmol), K2CO3 (60 mg, 0.43 mmol) and Pd(dppf)Cl2 (11 mg, 0.014 mmol) in 1,4-dioxane/H2O (8/1, 10 mL) was heated to 90 °C for 2 h under N2. The completion of the reaction was monitored by TLC. The reaction was concentrated directly. The resulting residue was purified by silica gel column chromatography with a gradient elution of 2% MeOH/DCM to 6% MeOH/DCM to provide 6-(3-methylisoxazol-5-yl)-4-morpholino-2-(3-(m- tolyl)-1H- pyrazol-1-yl)furo[3,2-d]pyrimidine (22 mg, 0.05 mmol) as a light yellow solid. LC- MS (ESI+): m/z 443 (MH+).1HNMR (300 MHz, CDCl3) δ 8.54 (d, J = 2.7 Hz, 1H), 7.90 (s, 1H), 7.76 (d, J = 7.5 Hz, 1H), 7.39-7.26 (m, 2H), 7.17 (d, J = 7.8 Hz, 1H), 6.78 (d, J = 2.7 Hz, 1H), 6.58 (s, 1H), 4.17-4.13 (m, 4H), 3.95-3.91 (m, 4H), 2.42 (s, 6H). [0503] EXAMPLE 42: Compound 212 using General synthetic route 42:
Figure imgf000220_0001
1) Synthesis of (2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)boronic acid
Figure imgf000220_0002
[0504] To a solution of 2-chloro-4-morpholinofuro[3,2-d]pyrimidine (200 mg, 0.84 mmol) in THF (30 mL) at -78 °C under N2 was added n-BuLi (0.4 mL, 2.5 M, 1.00 mmol). After stirred at -78 °C for 1 h, to the solution was added a solution of triisopropyl borate (190 mg, 1.00 mmol) in THF (2 mL). The reaction mixture was stirred at rt overnight. The reaction mixture was quenched with water (0.1 mL) and a large amount of white solid was precipitated. After filtration, the filter cake was washed with 4 mL THF and dried in vacuum to provide (2-chloro- 4-morpholinofuro[3,2-d]pyrimidin-6-yl)boronic acid (150 mg, 0.53 mmol) as white solid. LC- MS (ESI+): m/z 284/286 (MH+).1HNMR (300 MHz, CD3OD) δ 6.58 (s, 1H), 4.12-4.05 (m, 4H), 3.84-3.78 (m, 4H). 2) Synthesis of 4-(2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)-N,N,2-trimethyl-1H- imidazole-1-sulfonamide
Figure imgf000221_0001
[0505] A suspension of 4-iodo-N,N,2-trimethyl-1H-imidazole-1-sulfonamide (367 mg, 1.17 mmol), (2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)boronic acid (300 mg, 1.06 mmol), K2CO3 (439 mg, 3.18 mmol) and Pd(dppf)Cl2 (78 mg, 0.106 mmol) in 1,4-dioxane/H2O (8/1, 20 mL) was heated to 90 °C for 1 h under N2. The completion of the reaction was monitored by TLC. The reaction was concentrated directly under reduce pressure. The resulting residue was purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 2% MeOH/DCM to provide 4-(2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)-N,N,2-trimethyl- 1H-imidazole-1-sulfonamide (340 mg, 0.80 mmol) as a yellow solid. LC-MS (ESI+): m/z 427/429 (MH+).1HNMR (300 MHz, CDCl3) δ 7.62 (s, 1H), 7.02 (s, 1H), 4.09-4.03 (m, 4H), 3.91-3.84 (m, 4H), 3.04 (s, 6H), 2.69 (s, 3H). 3) Synthesis of N,N,2-trimethyl-4-(4-morpholino-2-(3-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2- d]pyrimidin-6-yl)-1H-imidazole-1-sulfonamide
Figure imgf000221_0002
[0506] A suspension of 4-(2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)-N,N,2-trimethyl- 1H- imidazole-1-sulfonamide (110 mg, 0.26 mmol), 3-(m-tolyl)-1H-pyrazole (49 mg, 0.31 mmol), t-BuONa (0.52 mL, 1M, 0.52 mmol), Pd2(dba)3 (14.8 mg, 0.066 mmol) and t-BuXphos (80 mg, 0.18 mmol) in toluene (10 mL) under N2 was heated to 100 °C for 1 h. The completion of the reaction was monitored by TLC. The reaction was concentrated directly. The residue was purified by silica gel column chromatography with a gradient elution of 1% MeOH/DCM to 2% MeOH/DCM to provide to provide 50 mg of impure product. After further slurry in MeOH to provide N,N,2-trimethyl-4-(4-morpholino-2-(3-(m-tolyl)-1H-pyrazol-1-yl)furo[3,2-d] pyrimidin- 6-yl)-1H-imidazole-1-sulfonamide (35 mg, 0.064 mmol) as a white solid. LC-MS (EMS+): m/z 549 (MH+). 4) Synthesis of 6-(2-methyl-1H-imidazol-5-yl)-4-morpholino-2-(3-(m-tolyl)-1H-pyrazol-1- yl)furo[3,2-d]pyrimidine
Figure imgf000222_0001
[0507] To a solution of N,N,2-trimethyl-4-(4-morpholino-2-(3-(m-tolyl)-1H-pyrazol-1-yl) furo[3,2-d]pyrimidin-6-yl)-1H-imidazole-1-sulfonamide (35 mg, 0.064 mmol) in 1,4-dioxane (4 mL) was added conc. HCl (0.3 mL). The reaction mixture was stirred at 80 °C for 2 h. The completion of the reaction was monitored by TLC. The reaction mixture was quenched with saturated NaHCO3 solution to pH = 8. A large amount of solid was precipitated. After filtration, the filter cake was slurry in Et2O to provide N,N,2-trimethyl-4-(4-morpholino-2-(3-(m-tolyl)- 1H-pyrazol-1-yl)furo[3,2-d]pyrimidin-6-yl)-1H-imidazole-1-sulfonamide (20 mg, 0.045 mmol) as a light yellow solid. LC-MS (ESI+): m/z 442 (MH+). 1HNMR (300 MHz, CDCl3) δ 8.53 (d, J = 2.7 Hz, 1H), 7.82 (s, 1H), 7.70 (d, J = 7.5 Hz, 1H), 7.43 (s, 1H), 7.31-7.26 (m, 1H), 7.14 (d, J = 7.8 Hz, 1H), 7.04 (s, 1H), 6.76 (d, J = 2.7 Hz, 1H), 4.15-4.11 (m, 4H), 3.95-3.91 (m, 4H), 2.51 (s, 3H), 2.41 (s, 3H). [0508] EXAMPLE 43: Compound 144 using General synthetic route 43: l
Figure imgf000222_0002
1) Synthesis of 2-chloro-6-(3-methyl-1H-pyrazol-5-yl)-4-morpholinofuro[3,2-d]pyrimidine [0509] A suspension of 2-chloro-6
Figure imgf000223_0001
-iodo-4-morpholinofuro[3,2-d]pyrimidine (400 mg, 1.08 mmol), (3-methyl-1H-pyrazol-5-yl)boronic acid (154 mg, 1.1 mmol), Na2CO3 (232 mg, 2.16 mmol) and Pd(PPh3)4 (12 mg, 0.01 mmol) in 1,4-dioxane/H2O (8 mL, 4:1) under N2 was heated to 50 °C for 2 h. The completion of the reaction was monitored by TLC. The reaction mixture was concentrated directly. The resulting residue was purified by silica gel column chromatography with a gradient elution of 33% EtOAc/PE to 50% EtOAc/PE to provide 2- chloro-6-(3-methyl-1H-pyrazol-5-yl)-4-morpholinofuro[3,2-d]pyrimidine (180 mg, 0.57 mmol) as a yellow solid. LC-MS (ESI+): m/z 320/322 (MH+). 2) Synthesis of 5-(2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)-N,N,3-trimethyl-1H- pyrazole-1-sulfonamide
Figure imgf000223_0002
[0510] To a solution of 2-chloro-6-(3-methyl-1H-pyrazol-5-yl)-4-morpholinofuro[3,2- d]pyrimidine (180 mg, 0.57 mmol) in THF at 0 °C was added NaH (27 mg, 0.67 mmol). The mixture was stirred at 0 °C for 0.5 h. To the above mixture at 0 °C was added Dimethylsulfamoyl chloride (104 mg, 0.73 mmol) dropwise. After addition, the reaction mixture was stirred at rt for 3.5 h. The completion of the reaction was monitored by TLC. The reaction mixture was quenched with water (20 mL). The aqueous solution was extracted with EtOAc (3 x 10 mL). The combined organic phase was dried over anhydrous Na2SO4, filtrated and concentrated under reduce pressure. The resulting residual was slurry in Et2O to provide crude 5-(2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)-N,N,3-trimethyl-1H-pyrazole-1- sulfonamide (270 mg, 0.63 mmol) as a yellow solid. LC-MS (ESI+): m/z 427/429 (MH+). 1HNMR (300 MHz, CDCl3) δ 7.06 (s, 1H), 6.49 (s, 1H), 4.13-4.04 (m, 4H), 3.87-3.81 (m, 4H), 3.10 (s, 6H), 2.58 (s, 3H). 3) Synthesis of 6-(3-methyl-1H-pyrazol-5-yl)-4-morpholino-2-(4-(m-tolyl)-1H-pyrazol-1- yl)furo[3,2-d]pyrimidine
Figure imgf000224_0001
[0511] To a solution of 5-(2-chloro-4-morpholinofuro[3,2-d]pyrimidin-6-yl)-N,N,3-trimethyl- 1H-pyrazole-1-sulfonamide (90 mg, 0.21mmol) in CH3CN (5 mL) was added 4-(m-tolyl)-1H- pyrazole (40 mg, 0.25 mmol) and Cs2CO3 (138 mg, 0.42 mmol). The reaction mixture was stirred at 160 °C in a sealed tube overnight. The completion of the reaction was monitored by TLC. The reaction mixture was concentrated directly and purified by preparative TLC with a elution of 10% MeOH/DCM to provide 6-(3-methyl-1H-pyrazol-5-yl)-4-morpholino-2-(4-(m- tolyl)-1H-pyrazol-1-yl)furo[3,2-d]pyrimidine (18 mg, 0.041 mmol) as a yellow solid. LC-MS (ESI+): m/z 442 (MH+).1HNMR (300 MHz, DMSO-d6) δ 13.52 (s, 1H), 9.00 (s, 1H), 8.22 (s, 1H), 7.61 (s, 1H), 7.57 (d, J = 7.8 Hz, 1H), 7.29 (t, J = 7.8 Hz, 1H), 7.15 (s, 1H), 7.08 (d, J = 7.8 Hz, 1H), 6.65 (s, 1H), 4.15-4.06 (m, 4H), 3.90-3.80 (m, 4H), 2.36 (s, 3H), 2.32 (s, 3H). [0512] Compounds 143 to 225 in Table 4 are made according to the procedures above. Table 4
Figure imgf000224_0002
Figure imgf000225_0001
Figure imgf000226_0001
Figure imgf000227_0001
Figure imgf000228_0001
Figure imgf000229_0001
Figure imgf000230_0001
Figure imgf000231_0001
Figure imgf000232_0001
Figure imgf000233_0001
Figure imgf000234_0001
Figure imgf000235_0001
Figure imgf000236_0001
Figure imgf000237_0001
Figure imgf000238_0001
Figure imgf000239_0001
Figure imgf000240_0001
Figure imgf000241_0001
[0513] Compounds 226-257 in Table 5 are prepared using methods analogous to those described herein. Table 5
Figure imgf000241_0002
Figure imgf000242_0001
Figure imgf000243_0001
Figure imgf000244_0001
Figure imgf000245_0001
Figure imgf000246_0001
Biological Example 1: Inhibition of PIKfyve [0514] Full length human recombinant PIKFYVE expressed in baculovirus expression system as N-terminal GST-fusion protein (265 kDa) was obtained from Carna Biosciences (Kobe, Japan). The kinase substrate was prepared by mixing and sonicating fluorescently-labeled phosphatidylinositol 3-phosphate (PI3P) with phospho-L-serine (PS) at a 1:10 ratio in 50 mM HEPES buffer pH 7.5. [0515] The kinase reactions were assembled in 384-well plates (Greiner) in a total volume of 20 mL as follows. Kinase protein was pre-diluted in an assay buffer comprising 25 mM HEPES, pH 7.5, 1 mM DTT, 2.5 mM MgCl2, and 2.5 mM MnCl2, and 0.005% Triton X-100, and dispensed into a 384-well plate (10 µL per well). Test compounds were serially pre-diluted in DMSO and added to the protein samples by acoustic dispensing (Labcyte Echo). The concentration of DMSO was equalized to 1% in all samples. All test compounds were tested at 12 concentrations. Apilimod was used as a reference compound and was tested in identical manner in each assay plate. Control samples (0%-inhibition, in the absence of inhibitor, DMSO only) and 100%-inhibition (in the absence of enzyme) were assembled in replicates of four and were used to calculate %-inhibition in the presence of compounds. The reactions were initiated by addition of 10 µL of 2x PI3P/PS substrate supplemented with ATP. The final concentration of enzyme was 2 nM, the final concentration of ATP was 10 mM, and the final concentration of PI3P/PS substrate was 1 µM (PI3P). The kinase reactions were allowed to proceed for 3 h at room temperature. Following incubation, the reactions were quenched by addition of 50 mL of termination buffer (100 mM HEPES, pH 7.5, 0.01% Triton X-100, 20 mM EDTA). Terminated plates were analyzed on a microfluidic electrophoresis instrument (Caliper LabChip® 3000, Caliper Life Sciences/Perkin Elmer). The change in the relative fluorescence intensity of the PI(3)P substrate and PI(3,5)P product peaks was measured. The activity in each test sample was determined as the product to sum ratio (PSR): P/(S+P), where P is the peak height of the product, and S is the peak height of the substrate. Percent inhibition (Pinh) was determined using the following equation: Pinh = (PSR0%inh - PSRcompound)/(PSR0%inh - PSR100%inh)*100 in which PSRcompound is the product/sum ratio in the presence of compound, PSR0%inh is the product/sum ratio in the absence of compound, and the PSR100%inh is the product/sum ratio in the absence of the enzyme. To determine the IC50 of test compounds (50%-inhibition) the %-inh cdata (Pinh versus compound concentration) were fitted by a four-parameter sigmoid dose- response model using XLfit software (IDBS).
[0516] The IC50 values for certain compounds of the disclosure are provided in Table 6 below. Table 6
Figure imgf000248_0001
Figure imgf000248_0002
Figure imgf000248_0003
Figure imgf000249_0001
Figure imgf000249_0002
Biological Example 2: Antiviral effects of PIKfyve inhibitors in a Vero-E6 SARS-CoV-2 cytopathic assay [0517] Materials and Methods [0518] An in vitro antiviral assay to test compounds acting against SARS-CoV-2 was designed and run based on cytopathic effect (CPE) in Vero (African green monkey kidney epithelial) cells essentially according to the methods of Ivens et al. (2005) Journal of Virological Methods 129, 56-63. [0519] VeroE6-EGFP cells (provided by Lab of Virology & Chemotherapy, Rega Institute, KU Leuven, Leuven, Belgium) (sometimes referred to herein as VeroE6, Vero-E6, or Vero-E6- GFP) were propagated in growth medium which was prepared by supplementing DMEM (Gibco 41965-039) with 10% v/v heat-inactivated FCS and 5 mL sodium bicarbonate 7.5% (Gibco 25080-060). Cells were cultured in T150 bottles and split 1/4 twice a week. Pen-strep was added directly to the T150 bottle at a 1/100 dilution. [0520] Assay medium was prepared by supplementing DMEM (Gibco 41965-039) with 2% v/v heat-inactivated FCS and 5 mL sodium bicarbonate 7.5% (Gibco 25080-060). [0521] Serial dilutions of compounds were prepared in 96-well plates to which cells were added (25,000 cells/well) after which plates were incubated overnight (37°C; 5% CO2). The next day virus inoculum (SARS-CoV-2 clinical isolate, Belgian strain: BetaCov/Belgium/GHB- 03021/2020) was added and plates were incubated for 4 days (37°C; 5% CO2) until a maximal cytopathic effect (CPE) could be observed in the untreated, infected (virus control) conditions. On day 4, GFP signal was determined using high-content imaging. [0522] Antiviral activity is expressed as the EC50 or concentration required to rescue 50% of the GFP signal from the virus-induced cytopathogenicy. The signal is provided as the logarithm of the surface of the well that is covered with fluorescent pixels which correlates with living cells. [0523] In parallel, and to avoid false positives, cytotoxicity was assessed by growing uninfected cells in the presence of the test compound at the concentrations tested. After a 4 day incubation, cell viability was measured using a commercial kit. [0524] Antiviral readout was performed using high-content imagers. Using a 5x objective, almost the entire well of a 384-well plate is captured at once (for an 96-well plate, the well is covered by 4 field of views). A GFP marker located in both the cell cytoplasm as well as the nucleus allowed for the calculation of the surface of the well that is (still) covered by cells (SpotTotalAreaCh2). The data are exported to .csv files and dose-response curves are processed in Dotmatics. [0525] Results and Discussion Compound 101 was tested to determine activity against SARS-CoV2. All tests were run in duplicate, with a top concentration of 10 µM (8 concentrations; dilution step 1/3). Figure 1 shows antiviral effect and cell toxicity data for Compound 101, which has an EC50 of about 0.1- 0.5 µM, CC50 > 100 µM, and Safety index (i.e., the ratio of CC50 to EC50) > 200. Compound 101 demonstrated reproducible and robust anti-viral activity in Vero-E6 cells with a therapeutically acceptable safety index. Biological Example 3: Antiviral effects of PIKfyve inhibitors in an A549-ACE2 cell assay [0526] Materials and Methods [0527] PIKfyve inhibitors were assessed for anti-viral activity against SARS-COV-2 using A549-ACE2 cells (adenocarcinomic human alveolar basal epithelial cells) transduced to express the human Angiotensin-converting enzyme 2 (ACE2), provided by Institut Pasteur, Paris, France. A549-ACE2 cells were grown in 96-well plates, and ten concentrations of each tested PIKfyve inhibitor was tested in triplicate. Concentrations of compounds tested included: 0.001 – 1uM (0, 0.001, 0.003, 0.01, 0.03, 0.1, 0.3, 1, 3, 10). The DMSO concentration in the final assay was < 1% (dilution of stock made in serial dilutions of media). [0528] Cells were pretreated for 2 hours before infection with SARS-CoV-2 virus (isolate BetaCoV/France/IDF0372/2020 C2). After virus infection, cells were incubated for 1 hour at 37°C. All cells were washed after a 48-hour incubation. [0529] Viral replication was measured by quantitative RT-PCR in the presence and absence of the tested compounds. [0530] In parallel, and to avoid false positives, cytotoxicity was assessed by growing uninfected cells in the presence of the three tested compounds at the concentrations tested. After 48 hours of incubation, cell viability was measured using a commercial kit. [0531] The test compound was provided in 10 mM solutions in 100 uL DMSO. [0532] Results [0533] Exemplary results are shown in Figure 2. The tested compound showed potent antiviral effects with minimal toxicity. Figure 2 shows results for Compound 97 where the viral titer is shown as Log10 of plaque forming units per mL (PFU/mL) on the left axis (data represented by white circles). Percent cell viability is shown on the right axis (data represented by black circles). Compound 97 has IC50 = 0.069 µM. Compounds 171 and 163 were also tested. Compound 171 has EC50 of 55.85 µM; Compound 163 has EC50 of 4.228 µM. Both compounds 163 and 171 have CC50 of >10000 nM. Biological Example 4: Reduction of virus-induced cytopathic effect (primary CPE assay) [0534] Materials and Methods [0535] Confluent or near-confluent cell culture monolayers of Vero 76 (African green monkey kidney epithelial cells, are provided by Institute for Antiviral Research, Utah State University, Utah, USA) cells and are prepared in 96-well disposable microplates the day before testing. Cells are maintained in MEM supplemented with 5% FBS. For antiviral assays, the same medium is used but with FBS reduced to 2% and supplemented with 50-µg/ml gentamicin. Compounds are dissolved in DMSO. The test compounds are prepared at four serial log10 concentrations, 0.1, 1.0, 10, and 100 µg/ml or µM. Five microwells are used per dilution: three for infected cultures and two for uninfected toxicity cultures. Controls for the experiment consisted of six microwells that are infected and not treated (virus controls) and six that are untreated and uninfected (cell controls) on every plate. A known active drug, the protease inhibitor M128533, is tested in parallel as a positive control drug using the same method as is applied for test compounds. [0536] Growth media is removed from the cells and the test compound is applied in 0.1 ml volume to wells at 2X concentration. The SARS-CoV-2, USA_WA1/2020 strain, normally at ~60 CCID50 (50% cell culture infectious dose) in 0.1 ml volume, is added to the wells designated for virus infection. Medium devoid of virus is placed in toxicity control wells and cell control wells. Plates are incubated at 37°C with 5% CO2 until marked CPE (>80% CPE for most virus strains) is observed in virus control wells. The plates are then stained with 0.011% neutral red for approximately two hours at 37°C in a 5% CO2 incubator. The neutral red medium is removed by complete aspiration, and the cells are optionally rinsed 1X with phosphate buffered solution (PBS) to remove residual dye. The PBS is completely removed, and the incorporated neutral red is eluted with 50% Sorensen’s citrate buffer/50% ethanol for at least 30 minutes. Neutral red dye penetrates into living cells, thus, the more intense the red color, the larger the number of viable cells present in the wells. The dye content in each well is quantified using a spectrophotometer at 540 nm wavelength. The dye content in each set of wells is converted to a percentage of dye present in untreated control wells using a Microsoft Excel computer-based spreadsheet and normalized based on the virus control. The 50% effective (EC50, virus-inhibitory) concentrations and 50% cytotoxic (CC50, cell-inhibitory) concentrations are then calculated by regression analysis. The quotient of CC50 divided by EC50 gives the selectivity index (SI) value. Compounds showing SI values >10 are considered active. Biological Example 5: Anti-viral Activity Against Human Alphacoronavirus Strain 229E (HCoV 229E) and Human Betacoronavirus Strain OC43 (HCoV OC43) [0537] Materials and Methods [0538] Representative compounds were tested for virustatic or virucidal properties against HCoV 229E and HCoV OC43. Stock solutions for each compound were prepared at a stock concentration of 10 mM for immediate use. [0539] EC50 (half maximal efficacious concentration) – Using appropriate concentrations of virus, this study assessed the virustatic or virucidal properties of tested Compounds. Cells were seeded into 96-well plates. The tested compounds were serially diluted (8-point, 3-fold dose titration) and added prophylactically for an hour, to cells. Cells were then infected with virus for one hour, at a single infective dose (100x median tissue culture infectious dose; TCID50). Additional media was added to the wells, with equivalent concentrations of compound, for the duration of the study. Vehicle and positive control wells were set up to control for any influence on cell viability. Cells were visually inspected daily for the appearance of any cytophathic/cytopathogenic effect (CPE). A colorimetric mammalian cell viability assay (Thiazolyl Blue Tetrazolium Bromide (MTT) assay; Sigma Catalog # M5655-1G; Lot # MKCL1832) was performed once CPE was complete. Percentage viral inhibition was calculated as follows: % Inhibition = [(A−B)/ (C−B)]×100, where: A: mean optical density of test, B: mean optical density of virus controls, C: mean optical density of cell controls. [0540] Values above 100% inhibition occur when A>C, due either to natural variation or compound effects. [0541] Values below 0% inhibition occur when A<B, due either to natural variation or compound toxicity. [0542] EC50 values were calculated using non-linear regression (log(agonist) vs. response – Variable slope (four parameters)) using log(concentration). [0543] CC50 (half maximal cytotoxic concentration) – The CC50 was determined in the manner described above for EC50. Plates were developed to show any cytotoxic effect of the compounds on cells in the absence of viral infection. The MTT assay was performed once CPE was complete. Percentage cell viability was calculated as follows: % viability = (A/B)×100, where: A: mean optical density of test, B: mean optical density of cell controls. Values above 100% inhibition occur when A>B, due either to natural variation or compound effects. [0544] CC50 values were calculated used non-linear regression as above for EC50. [0545] Experimental conditions and readouts – The virus strains/serotypes used were human alphacoronavirus 229E (HCoV 229E; ATCC® CVR-740™) and betacoronavirus 1 OC-4 (HCoV OC43; ATCC® VR-1558™). HCoV 229E was tested with human bronchial epithelial (16BHE) cells. HCoV OC43 was tested with human lung mucoepidermoid (H292) cells. Remdesivir was used as the control compound. [0546] Results [0547] Efficacy against HCoV 229E in 16BHE cells – Compound 91 showed efficacy against HCoV 229E, with percentage viral inhibition reaching 35% at approximately 0.32 µM (Figure 3A). Due to cytotoxicity observed above 0.32 µM, an EC50 value was not calculated. The CC50 was 0.21 µM. [0548] Compound 121 showed efficacy against HCoV 229E, with percentage viral inhibition reaching 33% at approximately 3.2 µM (Figure 3B). Cytotoxicity was observed between 10 µM and 3.2 µM. For Compound 121 the EC50 ≥ 3.16 µM and the CC50 was caculated to be 4.97 µM. [0549] Compound 114 was also tested and had EC50 ≥ 10 µM and CC50 of 1.02. Data not shown. [0550] Efficacy against HCoV OC43 in H292 cells – Compound 91 showed efficacy against HCoV OC43, with an EC50 of 0.08 µM (Figure 4A) and CC50 ≥ 10 µM. Compound 91 increased cell viability to levels that were higher than the uninfected cells, leading to percentages above 100%. The viability in infected cells was reduced to below 0% at 10 µM, 3.16 µM, and 1 µM. In order to calculate a more representative EC50, these data points were excluded from the calculation. [0551] Compound 114 showed efficacy against HCoV OC43, with an EC50 of 0.31 µM (Figure 4B). Compound 114 increased cell viability to levels that were higher than the uninfected cells, leading to percentages above 100%. The viability in infected cells was reduced to below 0% at 10 µM and 3.16 µM. In order to calculate a more representative EC50, these data points were excluded from the calculation. The compound had a CC50 ≥ 10 µM. [0552] Compound 121 showed efficacy against HCoV OC43, with an EC50 of 0.99 µM (Figure 4C). Compound 121 increased cell viability to levels that were higher than the uninfected cells, leading to percentages above 100%. No reduction in viability in infected cells was seen at the tested concentrations. The compound had a CC50 ≥ 10 µM. Biological Example 6: PIKfyve inhibition of SARS CoV2–induced cytopathic effect (CPE) in Vero E6 cells [0553] Materials and Methods [0554] Five PIKfyve inhibitors were assessed for anti-viral activity against SARS-CoV-2, strain USA_WA1/2020, using Vero E6 and Vero 6 cells according to the methods of Severson et al. (2007) Journal of Biomolecular Screening 33-40. [0555] Results [0556] A summary of the results is shown in Table 7. The tested compounds showed potent antiviral effects with minimal toxicity. Table 7.
Figure imgf000255_0001
Biological Example 7: Drug Drug Interaction Study with Cytochrome P450 Enzymes [0557] Preparation of stock solutions [0558] The stock solutions of test compound were prepared in dimethyl sulfoxide (DMSO) at 10 mM concentration, then diluted to 2 mM with DMSO. The final concentration of test compound was 10 μM. [0559] Preparation of positive inhibitors [0560] The concentration of positive inhibitor was listed in Table 8. For the stock solution preparation, if the positive control could not be well dissolved in the mixture of DMSO and acetonitrile (1:4) at the highest concentration, another mixture of acetonitrile and DMSO, the mixture of acetonitrile and H2O or DMSO was used to dissolve the compound. Table 8.
Figure imgf000256_0002
[0561] Preparation of substrate stock solution [0562] Preparation details of these substrates are given in Table 9. The substrate solution, which is stored at -20°C, was warmed to room temperature prior to use. Table 9.
Figure imgf000256_0001
[0563] Preparation of phosphate buffer (100 mM, pH 7.4) [0564] Solution A was prepared by adding 7.098 g of disodium hydrogen phosphate into 500 mL of pure water, followed by sonication. Solution B was prepared by adding 3.400 g of potassium dihydrogen phosphate to 250 mL of pure water, followed by sonication. Solution A was placed on a stirrer and Solution B was added slowly into Solution A until the pH reached 7.4. [0565] 10 mM NADPH solution was prepared, fresh prior to use, by dissolving nicotinamide adenine dinucleotide phosphate (NADPH) at 8.334 mg/mL in phosphate buffer. [0566] Preparation of master solution [0567] The master solution was prepared according to Table 10. The incubation was carried out in 96 deep well plates. The following volumes were dispensed into each well of the incubation plate, 179 μL of the substrate and human liver microsomes (HLM) mixture in phosphate buffer, 1 μL of the compound working solution, or vehicle (mixture of DMSO and acetonitrile (1:4)). The incubation plate was placed into the water bath and pre-warmed at 37°C for 15 minutes before the reactions were started by the addition of 20 μL of 10 mM NADPH solution in phosphate buffer. After the addition of NADPH, the incubation plate was incubated at 37°C for the corresponding time. The assay was performed in duplicate. Table 10.
Figure imgf000257_0001
[0568] The reaction was quenched by the addition of 1.5 volumes (300 μL) of cold acetonitrile containing 3% formic acid and internal standards (200 nM Labetalol, 200 nM Alprazolam and 200 nM tolbutamide). The plate was centrifuged at 3,220 g for 40 minutes, the 100 μL of the supernatant was transferred to a new plate. The supernatant was diluted with 100 μL pure water if appropriate. After mixing, the samples were analyzed using ultraperformance liquid chromatography-tandem mass spectrometry (UPLC/MS/MS). [0569] Data Analysis [0570] The automatic peak integration areas were checked for all of the samples. The Analyte Peak Area and Internal Standard Peak Area were exported into excel spreadsheet. The inhibition of each P450 enzyme in human liver microsomes was measured as the percentage decrease in the activity of marker metabolite formation compared to non-inhibited controls (= 100% activity). [0571] The percentage of remaining activity was calculated as follows: Area Ratio = Peak Area Analyte/ Peak Area Internal Standard Remaining Activity (%) = Area Ratio test compound/ Area Ratio vehicle*100% Inhibition% = 100-Remaining Activity (%) Table 11.
Figure imgf000257_0002
Biological Example 8: Permeability Study [0572] Preparation of MDCK-MDR1 Cells [0573] 50 μL and 25 mL of cell culture medium were added to each well of the Transwell® insert and reservoir, respectively. The HTS Transwell® plates were incubated at 37 °C, 5% CO2 for 1 hour before cell seeding. [0574] MDCK-MDR1 cells were diluted to 1.56х106 cells/mL with culture medium and 50 μL of cell suspension were dispensed into the filter well of the 96-well HTS Transwell plate. Cells were cultivated for 4-8 days in a cell culture incubator at 37 °C, 5% CO2, and 95% relative humidity. Cell culture medium was replaced every other day, beginning no later than 24 hours after initial plating. [0575] Preparation of Stock Solutions [0576] Stock solutions of the test compounds and of the positive controls were prepared in dimethyl sulfoxide (DMSO) at the concentration of 10 mM. Metoprolol and Digoxin were used as the control compounds. [0577] Assessment of Cell Monolayer Integrity [0578] Medium was removed from the reservoir and each Transwell insert and replaced with prewarmed fresh culture medium. Transepithelial electrical resistance (TEER) across the monolayer was measured using Millicell® Epithelial Volt-Ohm measuring system (Millipore®, USA). The Plate was returned to the incubator once the measurement was complete and the TEER value was calculated according to the following equation: [0579] TEER measurement (ohms) x Area of membrane (cm2) = TEER value (ohm•cm2) [0580] TEER value should be greater than 42 ohm•cm2, which indicates the well-qualified MDCK-MDR1 monolayer. [0581] Assay Procedures [0582] The MDCK-MDR1 plate was removed from the incubator and washed twice with pre- warmed Hanks' Balanced Salt solution (HBSS) (10 mM HEPES, pH 7.4), and then incubated at 37 °C for 30 minutes. The stock solutions of control compounds were diluted in DMSO to get 200 μM solutions and then diluted with HBSS (10 mM HEPES, pH 7.4) to get 1 μM working solutions. The test compounds were diluted in DMSO to get 200 μM solutions and then diluted with HBSS (10 mM HEPES with 4% BSA, pH 7.4) to get 1 μM working solutions. The final concentration of DMSO in the incubation system was 0.5%. [0583] To determine the rate of drug transport in the apical to basolateral direction 125 μL of the working solution was added to the Transwell insert (apical compartment), and 50 μL of sample was transferred immediately from the apical compartment to 200 μL of acetonitrile (ACN) containing internal standard (IS) (100 nM alprazolam, 200 nM Caffeine and 100 nM tolbutamide) in a new 96-well plate as the initial donor sample (A-B). After vortexing at 1000 rpm for 10 minutes, the wells in the receiver plate (basolateral compartment) were filled with 235 μL of transport buffer. [0584] To determine the rate of drug transport in the basolateral to apical direction.285 μL of the working solution were added to the receiver plate wells (basolateral compartment), and 50 μL of sample were transferred immediately from the basolateral compartment to 200 μL of acetonitrile containing IS (100 nM alprazolam, 200 nM Caffeine and 100 nM tolbutamide) in a new 96-well plate as the initial donor sample (B-A). After vortexing at 1000 rpm for 10 minutes, the Transwell insert (apical compartment) was filled with 75 μL of transport buffer, with the apical to basolateral direction and the basolateral to apical direction measured at the same time. [0585] Following incubation of the plates at 37 °C for 2 hours, 50 μL samples from donor sides (apical compartment for Ap→Bl flux, and basolateral compartment for Bl→Ap) and receiver sides (basolateral compartment for Ap→Bl flux, and apical compartment for Bl→Ap) were transferred to wells of a new 96-well plate, followed by the addition of 4 volumes of acetonitrile containing IS (100 nM alprazolam, 200 nM Caffeine and 100 nM tolbutamide). Samples were vortexed for 10 minutes and then centrifuged at 3,220 g for 40 minutes. An aliquot of 100 µL of the supernatant was mixed with an appropriate volume of ultra-pure water before LC-MS/MS analysis. [0586] To determine the Lucifer Yellow leakage after 2-hour transport period, stock solution of Lucifer yellow was prepared in DMSO and diluted with HBSS (10 mM HEPES, pH 7.4) to reach the final concentration of 100 μM.100 μL of the Lucifer yellow solution was added to each Transwell insert (apical compartment), followed by filling the wells in the receiver plate (basolateral compartment) with 300 μL of HBSS (10 mM HEPES, pH 7.4). The plates were Incubated at 37 °C for 30 mins.80 μL samples were removed directly from the apical and basolateral wells (using the basolateral access holes) and transferred to wells of new 96 wells plates. The Lucifer Yellow fluorescence (to monitor monolayer integrity) signal was measured in a fluorescence plate reader at 480 nM excitation and 530 nM emission. [0587] Data Analysis [0588] The apparent permeability coefficient (Papp), in units of centimeter per second, was calculated for the MDCK-MDR1 drug transport assays using the following equation: Papp = (VA × [drug]acceptor) / (Area × Time × [drug]initial,donor) [0589] where VA is the volume (in mL) in the acceptor well, Area is the surface area of the membrane (0.143 cm2 for Transwell-96 Well Permeable Supports), and time is the total transport time in seconds. [0590] The leakage of Lucifer Yellow, in unit of percentage (%), can be calculated using the following equation: %LY leakage = 100×[LY]acceptor/([LY]donor+[LY]acceptor) LY leakage of <1% is acceptable to indicate the well-qualified MDCK-MDR1 monolayer. [0591] Results for this Example are shown in Table 15. Biological Example 9: Hepatocyte Drug Metabolism Study [0592] Preparation of Working Solutions [0593] 10 mM stock solutions of test compound and positive controls were prepared in DMSO. In separate conical tubes, 10 mM test compound and the positive control were diluted to 100 μM by combining 198 μL of 50% acetonitrile / 50% water and 2 μL of 10 mM stock. [0594] Preparation of Hepatocytes [0595] Incubation medium (William’s E Medium supplemented with GlutaMAXTM) and hepatocyte thawing medium were placed in a 37 °C water bath, and allowed to warm for at least 15 minutes prior to use. [0596] A vial of cryopreserved hepatocytes was transferred from storage, ensuring that vials remain at cryogenic temperatures until thawing process ensued. The cells were thawed by placing the vial in a 37°C water bath and gently shaking the vials for 2 minutes. After thawing was completed, the vial was sprayed with 70% ethanol, and transferred to a biosafety cabinet. [0597] The hepatocytes were transferred into 50 mL conical tube containing thawing medium. The 50 mL conical tube was placed into a centrifuge and spun at 100 g for 10 minutes. Upon completion of spin, the thawing medium was aspirated and the hepatocytes resuspended in enough incubation medium to yield ~1.5 × 106 cells/mL. [0598] AO/PI Staining was used to count cells and determine the viable cell density, after which, the cells were diluted with incubation medium to a working cell density of 0.5 × 106 viable cells/mL. [0599] Procedure for Stability Determination [0600] 198 μL of hepatocytes were pipetted into each wells of a 96-well non-coated plate, which was then placed in the incubator to warm the hepatocytes for 10 minutes.2 μL of the 100 μM test compound or positive control were pipetted into respective wells of the 96-well non- coated plate to start the reaction, and the plate was returned to the incubator for the designed time points. [0601] The well contents were transferred in 25 μL aliquots at time points of 0, 15, 30, 60, 90 and 120 minutes, then mixed with 6 volumes (150 μL) of acetonitrile containing internal standard, IS (100 nM alprazolam, 200 nM caffeine and 100 nM tolbutamide) to terminate the reaction. After 5 minutes of vortexing, the samples were centrifuged for 45 minutes at 3,220 g. Aliquots of 100 µL of the supernatant were diluted by 100 µL ultra-pure water, and the mixture was used for liquid chromatography (LC) tandem mass spectrometry (MS) (LC/MS/MS) analysis. All incubations were performed in duplicate. [0602] Data Analysis [0603] All calculations were carried out using Microsoft Excel. Peak areas were determined from extracted ion chromatograms. The in vitro half-life (t1/2) of parent compound were determined by regression analysis of the percent parent disappearance vs. time curve. [0604] The in vitro half-life (in vitro t1/2) was determined from the slope value: in vitro t1/2 = 0.693 / k [0605] Conversion of the in vitro t1/2 (in min) into the in vitro intrinsic clearance (in vitro CLint, in µL/min/1×106 cells) was done using the following equation (mean of duplicate determinations): in vitro CLint = kV/N where V = incubation volume (0.2 mL); and N = number of hepatocytes per well (0.1 × 106 cells). [0606] The calculations of Scaled-up CLint (mL/min/kg), Predicted CLH (mL/min/kg) and EH were done using the following equation: Scaled-up CLint = (0.693/T1/2) × (1/(hepatocytes concentration (0.5 × 106 cells/mL))) × Scaling Factors (See Table 9); Predicted CLH = (QH × Scaled-up CLint × fub) / (QH + Scaled-up CLint × fub); and EH = Predicted CLH/QH where QH is the hepatic blood flow (mL/min/kg) (Table 9), fub is the fraction of unbound drug in plasma which is assumed to be 1. Table 12 shows scaling factors for intrinsic clearance prediction in human, monkey, dog, rat and mouse hepatocytes. Table 12.
Figure imgf000262_0003
[0607] The rules for data processing are shown in Table 13. Table 13.
Figure imgf000262_0002
[0608] Results for this example are shown in Table 14. Table 14.
Figure imgf000262_0001
Biological Example 10: Antiviral effects of PIKfyve inhibitors in a Vero-E6 SARS-CoV-2 cytopathic assay [0609] Materials and Methods [0610] To determine the half maximal inhibitory concentration (IC50) of the compounds against SARS-CoV-2, anti-viral screening was performed against two (2) SARS-CoV-2 strains, Wildtype (WT) and Delta (D). [0611] Adherent Vero-E6 cells were seeded in multi-well plates. Cell cultures were inoculated with a standardized amount of virus, in the absence and presence of serial compound dilutions, followed by 18-24 hours of incubation and an appropriate virus detection method (e.g., Immunostaining). Eight 3.16-fold dilutions of the compounds (lowest concentration 1 nM) were added to the assay cells for 1h before infection with each SARS-CoV-2 strain respectively. Virus signals detected in the presence of each compound concentration were used to determine the IC50. After infection, cells were fixed and immunostained for a viral antigen, and the percentage of infected cells quantified relative to an infected, untreated control using high content imaging. In parallel, the cytotoxicity of the same concentrations of each compound were tested in the absence of viral infection by MTT assay. As positive control, remdesivir was included. [0612] Results and Discussion [0613] Representative PYKfyve inhibitors were tested to determine their activity against two SARS-CoV2 strains: Wildtype (WT) and Delta (D). A summary of the results is shown in Table 15. Figures 5A, 5B and 5C show this data graphically. Table 15
Figure imgf000263_0001
[0614] The foregoing disclosure has been described in some detail by way of illustration and example, for purposes of clarity and understanding. Therefore, it is to be understood that the above description is intended to be illustrative and not restrictive. The scope of the disclosure should, therefore, be determined not with reference to the above description, but should instead be determined with reference to the following appended claims, along with the full scope of equivalents to which such claims are entitled.

Claims

What is claimed is: 1. A method of blocking alpha-coronavirus, beta-coronavirus lineage B, and beta- coronavirus lineage A entry into a host cell and preventing an infection caused by alpha- coronavirus, beta-coronavirus lineage B, and beta-coronavirus lineage A, comprising administering to a subject in need thereof a compound of (i) Formula (I)
Figure imgf000265_0001
wherein: R1a and R1b taken together with the nitrogen to which they are attached form:
Figure imgf000265_0002
wherein X and Y are independently N or CRa; wherein Ra is H or C1-4alkyl; and Rb is phenyl, monocyclic cycloalkyl, monocyclic heterocyclyl, monocyclic heterocycloalkyl, or monocyclic heteroaryl, each optionally substituted with one, two, or three Rd substituents; or R1a is H or C1-4alkyl; and R1b is a heteroaryl optionally substituted with Rc; wherein Rc is C1-4alkyl, phenyl, -C1-4alkyl-phenyl, monocyclic cycloalkyl, -C1-4alkyl-(monocyclic cycloalkyl), monocyclic heterocyclyl, monocyclic heterocycloalkyl, monocyclic heteroaryl, or -C1-4alkyl-(monocyclic heteroaryl), wherein each alkyl, phenyl, cycloalkyl, heterocyclyl, heterocycloalkyl or heteroaryl is optionally substituted with one, two, or three Rd substituents; wherein each Rd substituent is independently C1-4alkyl, C1-4alkenyl, C1-4alkynyl, -O-C1-4alkyl, halo, cyano, nitro, azido, C1-4haloalkyl, -O-C1-4-haloalkyl, -NRgRh, -NRgC(=O)Rh, -NRgC(=O)NRgRh, -NRgC(=O)ORh, =NORg, -NRgS(=O)1-2Rh, -NRgS(=O)1-2NRgRh, =NSO2Rg, -C(=O)Rg, -C(=O)ORg, -OC(=O)ORg, -OC(=O)Rg, -C(=O)NRgRh, -OC(=O)NRgRh, -ORg, -SRg, -S(=O)Rg, -S(=O)2Rg, -OS(=O)1-2Rg, -S(=O)1-2ORg, or -S(=O)1-2NRgRh; wherein Rg and Rh are each independently H or C1-4alkyl; each of R2 and R3 is independently chosen from H, C1-4alkyl, cycloalkyl, C1-4alkylcycloalkyl, heterocyclyl, heterocycloalkyl, and heteroaryl optionally substituted with one, two, or three Rj substituents; or R2 and R3 taken together with the nitrogen to which they are attached form a heterocyclyl, optionally substituted with one, two, three, or four Rj substituents, or further wherein any of the hydrogens bonded to carbon atoms are optionally replaced by deuterium; wherein each Rj substituent is independently C1-4alkyl, -OH, oxo, -NRkRl, halo, C1-4haloalkyl, -O-C1-4alkyl, or -O-C1-4-haloalkyl; where Rk and Rl are each independently H or C1-4alkyl; R4 is H, halo, -C(O)OH, C1-4alkylNRxRy, or -C(O)NRxRy, or is a cycloalkyl, heterocyclyl, heterocycloalkyl, phenyl or heteroaryl, wherein each cycloalkyl, heterocyclyl, heterocycloalkyl, phenyl or heteroaryl is optionally substituted with one, two, or three Rz substituents; wherein Rx is H or C1-4alkyl and Ry is H, C1-4alkyl, -O-C1-4alkyl, -SO2-Rr, C1-4alkyl-SO2-Rr, monocyclic cycloalkyl, -C1-4alkyl(monocyclic cycloalkyl), monocyclic heterocyclyl, or monocyclic heterocycloalkyl, each optionally substituted with one, two, or three Ro substituents; or Rx and Ry taken together with the nitrogen to which they are attached form a monocyclic heterocyclyl, optionally substituted with C1-4alkyl or -OC1-4alkyl; and each Rz substituent is independently C1-4alkyl, halo, -NRpRq, -C(O)NRpRq, -OH, or -OC1-4alkyl, wherein each alkyl is optionally substituted with -NRmRn; wherein Rm and Rn are each independently H, C1-4alkyl, C(O)C1-2alkyl, C(O)C1-2haloalkyl, C(O)C1-2alkenyl, or Rm and Rn taken together with the nitrogen to which they are attached form a monocyclic heterocycloalkyl, optionally substituted with one or two Ro substituents; wherein each Ro substituent is independently C1-4alkyl, -OH, -OC1-4alkyl, halo, cyano, methylsulfonyl, -NRpRq, or -C(O)NRpRq; wherein Rp and Rq are each independently H, C1-4alkyl, C1-4alkylNH2, C1-4alkylNH(C1- 4alkyl), or C1-4alkylN(C1-4alkyl)2; wherein each Rr is independently C1-4alkyl or NRpRq; and R5 is H, C1-4alkyl, halo, -OH, or -OC1-4alkyl; or a pharmaceutically acceptable salt or prodrug or prodrug thereof; or (ii) Formula (II) wherein
Figure imgf000267_0002
Rc1 is phenyl or pyridyl, each optionally substituted with one or two substituents selected from C1-4alkyl, -CF3, fluoro, chloro, -OCH3, and -OCF3; and R4a is C1-4alkylNRxRy or C(O)NRxRy; or is phenyl, pyrazolyl, or pyridyl, each optionally substituted with one or two Rz groups; or a pharmaceutically acceptable salt or prodrug thereof; or (iii) Formula (III): wherein
Figure imgf000267_0001
Rc1 is phenyl or pyridyl, each optionally substituted with one or two substituents selected from C1-4alkyl, -CF3, fluoro, chloro, -OCH3, and -OCF3; and R4a is C1-4alkylNRxRy or -C(O)NRxRy; or is phenyl, pyrazolyl, or pyridyl, each optionally substituted with one or two Rz groups; or a pharmaceutically acceptable salt or prodrug thereof; or (iv) Formula (IV): wherein
Figure imgf000267_0003
Rc1 is phenyl or pyridyl, each optionally substituted with one or two substituents selected from C1-4alkyl, -CF3, fluoro, chloro, -OCH3, and -OCF3; and R4a is C1-4alkylNRxRy or -C(O)NRxRy; or is phenyl, pyrazolyl, or pyridyl, each optionally substituted with one or two Rz groups; or a pharmaceutically acceptable salt or prodrug thereof. 2. The method of claim 1, wherein a. the alpha-coronavirus is HCoV 229E; b. the beta-coronavirus lineage B is SARS-CoV2; and c. the beta-coronavirus lineage A is HCoV OC43. 3. The method of claim 1, wherein the infection caused by SARS-CoV-2 is COVID-19. 4. The method of any one of claims 1 - 3, wherein R1a and R1b are taken together with the nitrogen to which they are attached to form
Figure imgf000268_0002
5. The method of any one of claims 1 - 3, wherein R1a and R1b are taken together with the nitrogen to which they are attached to form
Figure imgf000268_0001
. 6. The method of any one of claims 1 - 5, wherein X is N and Y is CRa. 7. The method of any one of claims 1 - 5, wherein X is CRa and Y is N. 8. The method of any one of claims 1 - 5, wherein X is N and Y is N. 9. The method of any one of claims 1 - 8, wherein Ra is H, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, or tert-butyl. 10. The method of any one of claims 1 - 8, wherein Ra is H or methyl. 11. The method of any one of claims 1 - 8, wherein Ra is H. 12. The method of any one of claims 1 - 11, wherein Rb is optionally substituted phenyl. 13. The method of any one of claims 1 - 11, wherein Rb is tolyl. 14. The method of any one of claims 1 - 11, wherein Rb is phenyl. 15. The method of any one of claims 1 - 11, wherein Rb is optionally substituted pyridinyl or pyrimidinyl. 16. The method of any one of claims 1 - 11, wherein Rb is optionally substituted pyridinyl. 17. The method of any one of claims 1 - 11, wherein Rb is substituted with one or two Rd substituents. 18. The method of any one of claims 1 - 11, wherein Rb is methylpryridinyl, phenyl, m-tolyl, chlorophenyl, bromophenyl, methoxyphenyl. 19. The method of any one of claims 1 - 18, wherein R1a is H or C1-4alkyl; and R1b is a 5- membered N-containing heteroaryl optionally substituted with Rc. 20. The method of any one of claims 1 - 18, wherein R1a is H.
21. The method of any one of claims 1 - 18, wherein R1a is C1-4alkyl. 22. The method of any one of claims 1 - 18, wherein R1a is methyl. 23. The method of any one of claims 1 - 22, wherein R1b is pyrrolyl, pyrazolyl, imidazolyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, oxadiazolyl, thiadiazolyl, pyrazolopyridinyl, or indazolyl, each optionally substituted with Rc. 24. The method of any one of claims 1 - 22, wherein R1b is pyrazolyl, imidazolyl, oxazolyl, oxadiazolyl or isoxazolyl, each optionally substituted with Rc. 25. The method of any one of claims 1 - 22, wherein R1b is pyrazolyl, optionally substituted with Rc. 26. The method of any one of claims 1 - 22, wherein R1b is
Figure imgf000269_0001
27. The method of any one of claims 1 - 22, wherein R 1b is
Figure imgf000269_0002
. 28. The method of any one of claims 1 - 27, wherein Rc is optionally substituted C1-4alkyl. 29. The method of any one of claims 1 - 27, wherein Rc is methyl, ethyl, isopropyl, or trifluoromethyl. 30. The method of any one of claims 1 - 27, wherein Rc is optionally substituted phenyl. 31. The method of any one of claims 1 - 27, wherein Rc is phenyl or o-, m-, p-tolyl, fluorophenyl, methoxyphenyl, or trifluoromethoxyphenyl. 32. The method of any one of claims 1 - 27, wherein Rc is phenyl. 33. The method of any one of claims 1 - 27, wherein Rc is optionally substituted monocyclic cycloalkyl. 34. The method of any one of claims 1 - 27, wherein Rc is optionally substituted cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl. 35. The method of any one of claims 1 - 27, wherein Rc is optionally substituted cyclopropyl. 36. The method of any one of claims 1 - 27, wherein Rc is optionally substituted monocyclic heterocycloalkyl. 37. The method of any one of claims 1 - 27, wherein Rc is optionally substituted cyclopropylmethyl, cyclobutylmethyl, cyclopentylmethyl, or cyclohexylmethyl.
38. The method of any one of claims 1 - 27, wherein Rc is optionally substituted monocyclic heterocyclyl. 39. The method of any one of claims 1 - 27, wherein Rc is optionally substituted pyrrolidinyl, tetrahydrofuranyl, piperidinyl, morpholinyl, or piperazinyl. 40. The method of any one of claims 1 - 27, wherein Rc is optionally substituted monocyclic heteroaryl. 41. The method of any one of claims 1 - 27, wherein Rc is optionally substituted pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, furanyl, thiophenyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, or pyridazinyl. 42. The method of any one of claims 1 - 27, wherein Rc is optionally substituted pyrazole, thiophenyl, imidazolyl, pyridinyl, or pyrimidinyl. 43. The method of any one of claims 1 - 27, wherein Rc is optionally substituted pyrazolyl. 44. The method of any one of claims 1 - 27, wherein Rc is optionally substituted pyridinyl. 45. The method of any one of claims 1 - 27, wherein Rc is methylpyridinyl. 46. The method of any one of claims 1 - 27, wherein Rc is optionally substituted -C1-4alkyl- phenyl, -C1-4alkyl-(monocyclic cycloalkyl), monocyclic heterocycloalkyl, or -C1-4alkyl- (monocyclic heteroaryl). 47. The method of any one of claims 1 - 27, wherein Rc is optionally substituted with one or two Rd substituents and each Rd substituent is independently C1-4alkyl, C1-4alkenyl, C1-4alkynyl, -O-C1-4alkyl, halo, cyano, nitro, azido, C1-4haloalkyl, -O-C1-4-haloalkyl, -NRgRh, -NRgC(=O)Rh, -NRgC(=O)NRgRh, -NRgC(=O)ORh, =NORg, -NRgS(=O)1-2Rh, -NRgS(=O)1-2NRgRh, =NSO2Rg, -C(=O)Rg, -C(=O)ORg, -OC(=O)ORg, -OC(=O)Rg, -C(=O)NRgRh, -OC(=O)NRgRh, -ORg, -SRg, -S(=O)Rg, -S(=O)2Rg, -OS(=O)1-2Rg, -S(=O)1-2ORg, or -S(=O)1-2NRgRh. 48. The method of any one of claims 1 - 47, wherein each Rd substituent is independently C1- 4alkyl, -O-C1-4alkyl, C1-4haloalkyl, or halo. 49. The method of any one of claims 1 - 47, wherein each Rd substituent is independently methyl, ethyl, isopropyl, -CF3, -OCH3, -OCF3, or fluoro. 50. The method of any one of claims 1 - 49, wherein Rg and Rh are each independently H or methyl. 51. The method of any one of claims 1 - 50, wherein each of R2 and R3 are independently selected from H, pyrrolidinyl, piperidinyl, and piperazinyl, wherein each pyrrolidinyl, piperidinyl, and piperazinyl is optionally substituted with one Rj substituent.
52. The method of any one of claims 1 - 50, wherein R2 and R3 taken together with the nitrogen to which they are attached form pyrrolidinyl, piperidinyl, piperazinyl, morpholino, or thiomorpholino, each optionally substituted with one, two, three, or four Rj substituents. 53. The method of any one of claims 1 - 50, wherein R2 and R3 taken together with the nitrogen to which they are attached form morpholino or piperazinyl, optionally substituted with one, two, three, or four Rj substituents. 54. The method of any one of claims 1 - 50, wherein R2 and R3 taken together with the nitrogen to which they are attached form 2,2,6,6-tetrafluoro-morpholino, morpholino-2-one, morpholino-3-one, piperazinyl-2-one, piperazinyl-3-one, thiomorpholino-1,1-dioxide. 55. The method of any one of claims 1 - 50, wherein each Rj substituent is independently methyl, oxo, hydroxy, NH2, -OCH3, halo, -CF3, or -OCF3. 56. The method of any one of claims 1 - 50, wherein R2 and R3 taken together with the nitrogen to which they are attached form morpholino in which 1 to 8 hydrogens are replaced with deuterium. 57. The method of any one of claims 1 - 56, wherein Rk and Rl are each independently H or methyl. 58. The method of any one of claims 1 - 57, wherein R4 is H. 59. The method of any one of claims 1 - 57, wherein R4 is chloro. 60. The method of any one of claims 1 - 57, wherein R4 is optionally substituted phenyl. 61. The method of any one of claims 1 - 57, wherein R4 is optionally substituted heteroaryl. 62. The method of any one of claims 1 - 57, wherein R4 is optionally substituted monocyclic heteroaryl. 63. The method of any one of claims 1 - 57, wherein R4 is optionally substituted pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, furanyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, or pyridazinyl. 64. The method of any one of claims 1 - 57, wherein R4 is
Figure imgf000272_0001
, , , , , , , or each optionally
Figure imgf000272_0002
substituted with 1 or 2 Rz groups.
Figure imgf000272_0003
65. The method of any one of claims 1 - 57, wherein R4 is optionally substituted pyridinyl or pyrimidinyl. 66. The method of any one of claims 1 - 57, wherein R4 is optionally substituted pyridinyl. 67. The method of any one of claims 1 - 57, wherein R4 is pyridinyl. 68. The method of any one of claims 1 - 57, wherein R4 is optionally substituted pyrazolyl. 69. The method of any one of claims 1 - 57, wherein R4 is optionally substituted with one or two Rz substituents. 70. The method of any one of claims 1 - 57, wherein R4 is pyrazolyl optionally substituted with one or two Rz substituents. 71. The method of any one of claims 1 - 57, wherein R4 is phenyl or pyridyl, each optionally substituted with one or two substituents selected from C1-4alkyl, -CF3, fluoro, chloro, -OCH3, and -OCF3. 72. The method of any one of claims 1 - 57, wherein R4 is heterocyclyl, optionally substituted with one or two Rz substituents. 73. The method of any one of claims 1 - 57, wherein R4 is pyrrolidinyl, piperidinyl, piperazinyl, morpholino, or thiomorpholino, optionally substituted with one or two Rz substituents. 74. The method of any one of claims 1 - 57, wherein R4 is heterocycloalkyl, optionally substituted with one or two Rz substituents. 75. The method of any one of claims 1 - 57, wherein R4 is pyrrolidinylmethyl, piperidinylmethyl, piperazinylmethyl, morpholinomethyl, or thiomorpholinomethyl, optionally substituted with one or two Rz substituents. 76. The method of any one of claims 1 - 57, wherein R4 is 3-methyl-1H-pyrazol-5-yl, 3- methylisothiazol-5-yl, 2-methyl-1H-imidazol-5-yl, 1-methyl-pyrazol-4-yl, 1-methylpyrazol-3-yl, 1-((1-acetamido)-eth-2-yl)-5-methyl-pyrazol-3-yl, 1-((1-chloromethylamido)-eth-2-yl)-5- methyl-pyrazol-3-yl, 1-((1-acrylamido)-eth-2-yl)-5-methyl-pyrazol-3-yl, thiazol-2-yl, pyrazol-4- yl, pyrazol-1-yl, oxazol-2-yl, 3-(1-N,N-dimethyl-eth-2-yl)-4-methyl-pyrazol-1-yl, or pyridinyl. 77. The method of any one of claims 1 - 57, wherein R4 is C1-4alkylNRxRy. 78. The method of any one of claims 1 - 57, wherein R4 is CH2NRxRy . 79. The method of any one of claims 1 - 57, wherein R4 is -C(O)NRxRy. 80. The method of any one of claims 1 - 79, wherein Rx is H. 81. The method of any one of claims 1 - 79, wherein Rx is methyl or ethyl, optionally substituted with one, two, or three Ro substituents. 82. The method of any one of claims 1 - 79, wherein Rx is methyl. 83. The method of any one of claims 1 - 82, wherein Ry is H. 84. The method of any one of claims 1 - 82, wherein Ry is C1-4alkyl, optionally substituted with one, two, or three Ro substituents. 85. The method of any one of claims 1 - 82, wherein Ry is methyl, ethyl, propyl, or isopropyl, each optionally substituted with one, two, or three Ro substituents. 86. The method of any one of claims 1 - 82, wherein Ry is H, methyl, ethyl, methyoxy, or methoxyethyl. 87. The method of any one of claims 1 - 82, wherein Ry is methyl. 88. The method of any one of claims 1 - 82, wherein Ry is -SO2-Rr or C1-4alkyl-SO2-Rr. 89. The method of any one of claims 1 - 82, wherein Ry is -SO2-Rr, C1-4alkyl-SO2-Rr; and Rr is CH3 or NH2, NHCH3, or N(CH3)2. 90. The method of any one of claims 1 - 82, wherein Ry is -SO2-methyl, C2-4alkyl-SO2- N(CH3)2. 91. The method of any one of claims 1 - 82, wherein Ry is monocyclic cycloalkyl or -C1- 2alkyl(monocyclic cycloalkyl), each optionally substituted with one, two, or three Ro substituents. 92. The method of any one of claims 1 - 82, wherein Ry is monocyclic cycloalkyl, optionally substituted with one, two, or three Ro substituents. 93. The method of any one of claims 1 - 82, wherein Ry is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl, each optionally substituted with one, two, or three Ro substituents. 94. The method of any one of claims 1 - 82, wherein Ry is cyclopropyl. 95. The method of any one of claims 1 - 82, wherein Ry is cyclopropyl, cyclobutyl, cyclopentyl, cyclopropylmethyl, 1-cyclopropylethyl, 2-cyclopropylethyl, cyclobutylmethyl, or cyclopentylmethyl.
96. The method of any one of claims 1 - 82, wherein Ry is monocyclic heterocyclyl, optionally substituted with one, two, or three Ro substituents. 97. The method of any one of claims 1 - 82, wherein Ry is optionally substituted azetidinyl, oxetanyl, pyrrolidinyl, piperidinyl, morpholinyl, piperazinyl, azepanyl, azocanyl, tetrahydrofuranyl, or tetrahydropyranyl, optionally substituted with methyl. 98. The method of any one of claims 1 - 82, wherein Ry is monocyclic heterocycloalkyl, optionally substituted with one, two, or three Ro substituents. 99. The method of any one of claims 1 - 82, wherein Ry is optionally substituted azetidinylmethyl, oxetanylmethyl, pyrrolidinylmethyl, piperidinylmethyl, morpholinylmethyl, or piperazinylmethyl, optionally substituted with methyl. 100. The method of any one of claims 1 - 79, wherein one of Rx and Ry is H and the other is - CH3. 101. The method of any one of claims 1 - 79, wherein both of Rx and Ry is H. 102. The method of any one of claims 1 - 79, wherein both of Rx and Ry is -CH3. 103. The method of any one of claims 1 - 79, wherein Rx and Ry taken together with the nitrogen to which they are attached form a monocyclic heterocyclyl, optionally substituted with C1-4alkyl. 104. The method of any one of claims 1 - 79, wherein Rx and Ry are taken together with the nitrogen to which they are attached to form azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, or thiomorpholinyl, each optionally substituted with methyl. 105. The method of any one of claims 1 - 104, wherein each Rz is independently C1-4alkyl, halo, -OH, or -OC1-4alkyl, wherein each alkyl is optionally substituted with -NRmRn. 106. The method of any one of claims 1 - 104, wherein each Rz is independently -CH3, -OH, halo, or -OCH3. 107. The method of any one of claims 1 - 104, wherein Rz is C2-4alkyl substituted with -NRmRn or OCH3. 108. The method of any one of claims 1 - 104, wherein each Rz substituent is independently -NRpRq, -C(O)NRpRq. 109. The method of any one of claims 1 - 104, wherein each Rz substituent is methyl, ethyl, isopropyl, -CF3, fluoro, chloro, -OCH3, -OCF3, methylamino, ethylamino, propylamino, butylamino, aminomethyl, aminoethyl, aminopropyl, aminobutyl, dimethylamino, dimethylaminomethyl, dimethylaminoethyl, dimethylaminopropyl, dimethylaminobutyl, - C(O)methylamino, -C(O)ethylamino, -C(O)propylamino, -C(O)butylamino, - C(O)dimethylamino, -C(O)dimethylaminomethyl, -C(O)dimethylaminoethyl, - C(O)dimethylaminopropyl, or -C(O)dimethylaminobutyl. 110. The method of any one of claims 1 - 109, wherein Rm and Rn are each independently H, C1-4alkyl, C(O)CH3, C(O)CH2Cl, or C(O)CH2CH2. 111. The method of any one of claims 1 - 109, wherein Rm and Rn are each H. 112. The method of any one of claims 1 - 109, wherein Rm and Rn are each methyl. 113. The method of any one of claims 1 - 109, wherein Rm and Rn taken together with the nitrogen to which they are attached form a monocyclic heterocyclyl, optionally substituted with one or two Ro substituents. 114. The method of any one of claims 1 - 109, wherein Rm and Rn taken together with the nitrogen to which they are attached form pyrrolidinyl, piperidinyl, piperazinyl, morpholino, thiomorpholino, or thiomorpholino-1,1-dioxide, each optionally substituted with one or two Ro substituents. 115. The method of any one of claims 1 - 109, wherein Rm and Rn taken together with the nitrogen to which they are attached form pyrrolidinyl, piperidinyl, piperazinyl, or morpholino, each optionally substituted with methyl. 116. The method of any one of claims 1 - 115, wherein each Ro substituent is C1-4alkyl, or -NRpRq. 117. The method of any one of claims 1 - 116, wherein Rp and Rq are each independently H, methyl, C1-4alkylNH2, C1-4alkylNHCH3, or C1-4alkylN(CH3)2. 118. The method of any one of claims 1 - 116, wherein Rp and Rq are each independently H or methyl. 119. The method of any one of claims 1 - 118, wherein R5 is H, methyl, ethyl, chloro, bromo, fluoro, -OH, or -OCH3. 120. The method of any one of claims 1 - 118, wherein R5 is H. 121. The method of any one of claims 1 - 3, wherein Rc1 is phenyl or pyridyl, each optionally substituted with methyl, -CF3, Cl, Br, or OCH3. 122. The method of any one of claims 1 - 3, wherein Rc1 is phenyl. 123. The method of any one of claims 1 - 3, wherein Rc1 is tolyl. 124. The method of any one of claims 1 - 3, wherein Rc1 is pyridyl optionally substituted with methyl or -CF3. 125. The method of any one of claims 1 - 3 or 121-124, wherein R4a is pyridyl, optionally substituted with one or two Rz groups. 126. The method of any one of claims 1 - 3 or 121-124, wherein R4a is pyridyl.
127. The method of any one of claims 1 - 3 or 121-124, wherein R4a is pyrazolyl optionally substituted with one or two Rz groups. 128. The method of any one of claims 1 - 3 or 121-124, wherein R4a is 3-methyl-1H-pyrazol- 5-yl, 3-methylisothiazol-5-yl, 2-methyl-1H-imidazol-5-yl, 1-methyl-pyrazol-4-yl, 1- methylpyrazol-3-yl, 1-((1-acetamido)-eth-2-yl)-5-methyl-pyrazol-3-yl, 1-((1- chloromethylamido)-eth-2-yl)-5-methyl-pyrazol-3-yl, 1-((1-acrylamido)-eth-2-yl)-5-methyl- pyrazol-3-yl, thiazol-2-yl, pyrazol-4-yl, pyrazol-1-yl, oxazol-2-yl, or 3-(1-N,N-dimethyl-eth-2- yl)-4-methyl-pyrazol-1-yl. 129. The method of any one of claims 1 - 3 or 121-124, wherein R4a is -C(O)NRxRy wherein Rx is H or C1-4alkyl and Ry is H, C1-4alkyl, -O-C1-4alkyl, -SO2-Rr, C1-4alkyl-SO2-Rr monocyclic cycloalkyl, -C1-4alkyl(monocyclic cycloalkyl), monocyclic heterocyclyl, or monocyclic heterocycloalkyl, each optionally substituted with one, two, or three Ro substituents; and Rr and Ro are as defined herein. 130. The method of any one of claims 1 - 3 or 121-124, wherein R4a is -C(O)NRxRy wherein Rx is H or methyl; and Ry is H, methyl, ethyl, butyl, isopropyl, methoxy, -SO2-methyl, C2-4alkyl-SO2-methyl, C2-4alkyl-SO2-N(CH3)2, cyclopropyl, cyclobutyl, cyclopentyl, cyclopropylmethyl, 1-cyclopropylethyl, 2-cyclopropylethyl, cyclobutylmethyl, cyclopentylmethyl, azetidinyl, oxetanyl, pyrrolidinyl, piperidinyl, morpholinyl, piperazinyl, azepanyl, azocanyl, tetrahydrofuranyl, tetrahydropyranyl, substituted azetidinylmethyl, oxetanylmethyl, pyrrolidinylmethyl, piperidinylmethyl, morpholinylmethyl, or piperazinylmethyl, each optionally substituted with one, two, or three methyl, methoxy, fluoro or amino groups. 131. The method of any one of claims 1 to 130, wherein the compound is selected from a compound of Table 1 or a pharmaceutically acceptable salt thereof. 132. The method of any one of claims 1 to 131, wherein one or more hydrogen atoms attached to carbon atoms of the compound are replaced by deuterium atoms. 133. The method of any one of claims 1 to 132, wherein the compound and/or the pharmaceutically acceptable salt is in a pharmaceutical composition. 134. The method of claim 133, wherein the pharmaceutical composition further comprises a pharmaceutically acceptable excipient.
PCT/US2022/032464 2021-06-08 2022-06-07 Methods and treatment of viral infection with substituted furo-pyrimidines WO2022261068A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163208459P 2021-06-08 2021-06-08
US63/208,459 2021-06-08

Publications (1)

Publication Number Publication Date
WO2022261068A1 true WO2022261068A1 (en) 2022-12-15

Family

ID=82595143

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2022/032464 WO2022261068A1 (en) 2021-06-08 2022-06-07 Methods and treatment of viral infection with substituted furo-pyrimidines

Country Status (2)

Country Link
TW (1) TW202313051A (en)
WO (1) WO2022261068A1 (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5846514A (en) 1994-03-25 1998-12-08 Isotechnika, Inc. Enhancement of the efficacy of nifedipine by deuteration
US6334997B1 (en) 1994-03-25 2002-01-01 Isotechnika, Inc. Method of using deuterated calcium channel blockers
WO2016118709A1 (en) * 2015-01-23 2016-07-28 Lam Therapeutics, Inc. Anti-viral compositions containing pikfyve inhibitors and use thereof
WO2020243457A1 (en) * 2019-05-29 2020-12-03 Viogen Biosciences, Llc Compounds and therapeutic uses thereof
WO2021158635A1 (en) * 2020-02-07 2021-08-12 Al Therapeutics, Inc. Anti-viral compositions and methods of use
WO2021183439A1 (en) * 2020-03-09 2021-09-16 Verge Analytics, Inc. Substituted furo[3,2-d]pyrimidines and uses thereof

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5846514A (en) 1994-03-25 1998-12-08 Isotechnika, Inc. Enhancement of the efficacy of nifedipine by deuteration
US6334997B1 (en) 1994-03-25 2002-01-01 Isotechnika, Inc. Method of using deuterated calcium channel blockers
WO2016118709A1 (en) * 2015-01-23 2016-07-28 Lam Therapeutics, Inc. Anti-viral compositions containing pikfyve inhibitors and use thereof
WO2020243457A1 (en) * 2019-05-29 2020-12-03 Viogen Biosciences, Llc Compounds and therapeutic uses thereof
WO2021158635A1 (en) * 2020-02-07 2021-08-12 Al Therapeutics, Inc. Anti-viral compositions and methods of use
WO2021183439A1 (en) * 2020-03-09 2021-09-16 Verge Analytics, Inc. Substituted furo[3,2-d]pyrimidines and uses thereof

Non-Patent Citations (23)

* Cited by examiner, † Cited by third party
Title
"Curr., Pharm. Des.", vol. 6, 2000, article "Recent Advances in the Synthesis and Applications of Radiolabeled Compounds for Drug Discovery and Development", pages: 110
"Fieser and Fieser's Reagents for Organic Synthesis", vol. 1-40, 1991, JOHN WILEY AND SONS
"Larock's Comprehensive Organic Transformations", vol. 1-5, 1989, ELSEVIER SCIENCE PUBLISHERS
"Remington's Pharmaceutical Sciences", 1985, MACK PUBLISHING COMPANY
BEZIAU ET AL., VIRUSES, vol. 12, 2020, pages 1124
BOUHADDOU ET AL., CELL, vol. 181, 2020, pages 271 - 280
EVANS, E. ANTHONY: "Synthesis of radiolabeled compounds", J. RADIOANAL. CHEM., vol. 64, no. 1-2, 1981, pages 9 - 32
GEORGE W.VARMA, RAJENDER S: "The Synthesis of Radiolabeled Compounds via Organometallic Intermediates", TETRAHEDRON, vol. 45, no. 21, 1989, pages 6601 - 21
HEALD-SARGENTGALLAGHER, VIRUSES, vol. 4, 2012, pages 557 - 580
IKONOMOV ET AL., J. BIOL. CHEM., vol. 276, no. 28, 2001, pages 26141 - 26147
IKONOMOV ET AL., J. BIOL. CHEM., vol. 277, no. 11, 2002, pages 6073 - 6079
IVENS ET AL., JOURNAL OF VIROLOGICAL METHODS, vol. 129, 2005, pages 56 - 63
JEFFERIES ET AL., EMBO REP, vol. 9, no. 2, 2008, pages 164 - 170
KANG ET AL., PNAS, vol. 117, no. 34, 2020, pages 20803 - 20813
LAU ET AL., J. VIROL., vol. 86, no. 21, 2012, pages 11906 - 11918
MAHENDRA ET AL., CUREUS, vol. 12, no. 3, 2020, pages e7423
MILLETWHITTAKER, VIRUS RES, vol. 202, 2015, pages 120 - 134
MURRAY ET AL., J. VIROLOGY, vol. 79, 2005, pages 11742 - 11751
NELSON ET AL., PLOS NEGL. TROP. DIS., vol. 11, no. 4, 2017, pages e0005540
OU ET AL., NAT. COMMUN., vol. 11, no. 1620, 2020, pages 1 - 12
RUTHERFORD ET AL., J. CELL SCI., vol. 119, no. 19, 2006, pages 3944 - 3957
SEVERSON ET AL., JOURNAL OF BIOMOLECULAR SCREENING, vol. 33-40, 2007
WOO ET AL., VIRUSES, vol. 2, no. 8, 2010, pages 1804 - 1820

Also Published As

Publication number Publication date
TW202313051A (en) 2023-04-01

Similar Documents

Publication Publication Date Title
CN109476659B (en) Novel tetrahydropyrazolopyridine compounds for the treatment of infectious diseases
EP3535262B1 (en) Novel tetrahydropyridopyrimidines for the treatment and prophylaxis of hepatitis b virus infection
EP3535261B1 (en) Novel tetrahydropyridopyrimidines for the treatment and prophylaxis of hepatitis b virus infection
CA2930414C (en) Tetrahydroquinoline compositions as bet bromodomain inhibitors
AU2014241183B2 (en) 6-(5-hydroxy-1H-pyrazol-1-yl)nicotinamide derivatives and their use as PHD inhibitors
IL261472A (en) Inhibitors of wdr5 protein-protein binding
CN113906028A (en) Fused heterocyclic derivatives
TWI770501B (en) Novel indole-2-carboxamides active against the hepatitis b virus (hbv)
WO2022253255A1 (en) Fused heterocyclic derivatives
WO2022261069A1 (en) Methods and treatment of viral infection with substituted pyrazolo-pyrimidines
EP4118087A1 (en) Substituted furo[3,2-d]pyrimidines and uses thereof
US11584737B2 (en) Heterocyclic compound
TW202106685A (en) Novel phenyl and pyridyl ureas active against the hepatitis b virus (hbv)
WO2020109224A1 (en) Further heteroaromatic compounds having activity against rsv
WO2022261068A1 (en) Methods and treatment of viral infection with substituted furo-pyrimidines
WO2022169882A1 (en) Methods and treatment of viral infection caused by sars-cov-2
WO2012010663A9 (en) 2,3,5-trisubstituted thiophene compounds and uses thereof
KR20220151655A (en) Inhibitors of human immunodeficiency virus replication

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22743630

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 22743630

Country of ref document: EP

Kind code of ref document: A1