[go: up one dir, main page]

WO2024233642A1 - Heterocyclic compounds and uses thereof - Google Patents

Heterocyclic compounds and uses thereof Download PDF

Info

Publication number
WO2024233642A1
WO2024233642A1 PCT/US2024/028330 US2024028330W WO2024233642A1 WO 2024233642 A1 WO2024233642 A1 WO 2024233642A1 US 2024028330 W US2024028330 W US 2024028330W WO 2024233642 A1 WO2024233642 A1 WO 2024233642A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
subject
pharmaceutically acceptable
solvate
alkyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
PCT/US2024/028330
Other languages
French (fr)
Inventor
Sanjay Shivayogi Magavi
Daniel J. Parks
Bradley Dean Tait
Jinhyung CHO
Adrianne KOLPAK
Patricia R. SHAW
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Progentos Therapeutics Inc
Original Assignee
Progentos Therapeutics 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 Progentos Therapeutics Inc filed Critical Progentos Therapeutics Inc
Priority to AU2024269869A priority Critical patent/AU2024269869A1/en
Publication of WO2024233642A1 publication Critical patent/WO2024233642A1/en
Priority to IL323606A priority patent/IL323606A/en
Anticipated expiration legal-status Critical
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia

Definitions

  • the present disclosure relates to compounds that can promote remyelination. Specifically, the disclosure relates to PDGFR ⁇ inhibitors and the use of such inhibitors to treat diseases associated with impaired myelination (e.g., multiple sclerosis).
  • BACKGROUND OF THE DISCLOSURE [0004] Proper myelination by oligodendrocytes is essential for central nervous system (CNS) development and function. Gacem et al., Life 11(4): 327 (Apr. 2021). Myelin maintains axon potential conduction velocity of neural signals and provides metabolic support to axons, supporting their survival.
  • oligodendrocyte progenitor cells undergo morphological and molecular changes as they differentiate into oligodendrocytes that can myelinate axons. Accordingly, any abnormal developmental processes or pathogenic immune activation and failure of oligodendrocytes to myelinate axons or loss of myelin can lead to neurodegenerative diseases such as multiple sclerosis.
  • Multiple sclerosis is generally characterized by inflammation and demyelination of neuronal axons. Patients with early MS often suffer from isolated immune attacks with effective remyelination and recovery between attacks.
  • a compound of Formula I [0009] or a pharmaceutically acceptable salt or solvate thereof, wherein: indicates a single bond or a double bond such that all valences are satisfied; X 1 , X 2 , X 3 , and X 4 are selected from N and CR a , with the proviso that not more than two of X 1 , X 2 , X 3 , and X 4 are N; one of Y 1 and Y 2 is N and the other of Y 1 and Y 2 is C; each R a is independently selected from H, halo, C 1 -C 4 alkyl, and C 1 -C 4 alkoxy; R 1 is selected from C 1 -C 4 alkyl, C 3 -C 8 cycloalkyl, 3-8 membered heterocyclyl,
  • Y 1 is N and Y 2 is C. [0011] In some aspects, Y 1 is C and Y 2 is N. [0012] In some aspects, X 1 is N, X 2 is CR a , X 3 is CR a , and X 4 is CR a . [0013] In some aspects, X 1 is CR a , X 2 is N, X 3 is CR a , and X 4 is CR a . [0014] In some aspects, X 1 is CR a , X 2 is CR a , X 3 is N, and X 4 is CR a .
  • X 1 is CR a
  • X 2 is CR a
  • X 3 is CR a
  • X 4 is N.
  • X 1 is CR a
  • X 2 is CR a
  • X 3 is CR a
  • X 4 is CR a
  • R 1 is 5- or 6-membered heteroaryl optionally substituted with C 1 - C 4 alkyl or oxo.
  • R 2 is substituted with one R 3 .
  • R 1 is an optionally substituted pyrazolyl.
  • R 1 is selected from: , , nd [0021]
  • R 10 is selected from H, C 1 -C 4 alkyl, C 1 -C 4 alkoxy, aminoC 1 -C 4 alkyl, hydroxyC 1 - C 4 alkyl, and C 1 -C 4 alkylsulfonyl, wherein the C 1 -C 4 alkyl, C 1 -C 4 alkoxy, aminoC 1 -C 4 alkyl, hydroxyC 1 -C 4 alkyl, and C 1 -C 4 alkylsulfonyl can be optionally substituted by one or more substituents selected from hydroxyl, C 1 -C 4 alkoxy, NR 10a R 10b , halo, and deuterium, wherein R 10a and R 10b are selected from hydrogen and C 1 -C 4 alkyl, or wherein R 10a and R 10b taken together with the nitrogen atom to which they are attached form a 4- to 8-membere
  • R 1 is .
  • R 10 is CH 3 .
  • R 2 is heterocyclyl.
  • R 2 is selected from: , , , , ,
  • R 2 is selected from:
  • the compound, or a pharmaceutically acceptable salt or solvate thereof has Formula Ia:
  • a and b are each independently selected from 1, 2, and 3; and [0032] Q is selected from -CH- and -N-, with the proviso that if Q is -N-, a and b are not 1.
  • the compound, or a pharmaceutically acceptable salt or solvate thereof ihas Formula II: .
  • the compound, or a pharmaceutically acceptable salt or solvate thereof has Formula IIa:
  • R 3 is heteroaryl optionally substituted with one, two, three, four, or five R 30 .
  • R 3 is selected from: , , , , , , , , [0037] wherein A 1 is selected from O, S, and NR 37 , R 36 is selected from hydrogen, optionally substituted C 1 -C 6 alkyl, and optionally substituted C 1 -C 6 alkylaryl, and R 37 is selected from hydrogen and C 1 -C 6 alkyl.
  • R 3 is selected from:
  • R 30 is: .
  • R 300 is selected from:
  • R 300 is: . [0042] In some aspects, R 301 is H and R 302 is -OH or CH 3 . [0043] In some aspects, R 3 is selected from: nd . [0044] In some aspects, R 3 is selected from: , , . [0045] In some aspects, the compound, or a pharmaceutically acceptable salt or solvate thereof, has Formula IV: wherein L 1 is NH or O. [0046] In some aspects, the compound, or a pharmaceutically acceptable salt or solvate thereof, has Formula V: . [0047] In some aspects, R 31 is selected from: , , , , , ,
  • R 31a is selected from H, D, alkylamino, optionally substituted C 1 -C 4 alkyl, C 1 -C 4 alkoxy, amino, and C 1 -C 4 haloalkyl
  • each R 31b is independently selected from H, D, halo, hydroxy, amino, cyano, alkylamino, optionally substituted C 1 -C 4 alkyl, C 1 -C 4 haloalkyl, optionally substituted C 1 - C 4 alkoxy, and C 3 -C 6 cycloalkyl
  • q is 1, 2, or 3.
  • the compound of Formula I is selected from any one of the compounds of Table 1.
  • the compound of Formula I can exhibit one or more of the following properties: (i) promote the differentiation of an OPC into an oligodendrocyte, (ii) promote the expression of a protein associated with oligodendrocyte differentiation and/or myelination (e.g., G-protein coupled receptor 17 (GPR17), myelin basic protein (MBP), ASPA, GST-pi, CC1, myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte- specific protein/claudin-11, CNPase, proteolipid protein 1 (PLP1), or a combination thereof), (iii) promote the myelination of an axon, (iv) promote the remyelination of a demyelinated axon, (v) inhibit PDGFR
  • GPR17 G-protein coupled receptor 17
  • MBP myelin basic protein
  • ASPA GST-
  • the compound of Formula I can inhibit PDGFR ⁇ kinase activity.
  • the compound of Formula I can inhibit PDGFR ⁇ kinase activity with an IC 50 of less than 500 nM, less than 400 nM, less than 300 nM, less than 200 nM, less than 100 nM, less than 75 nM, less than 50 nM, less than 40 nM, less than 30 nM, less than 20 nM, less than 10 nM, less than 5 nM, less than 2.5 nM, less than 1 nM, less than 0.5 nM, or less than 0.2 nM.
  • the IC 50 of the PDGFR ⁇ inhibitor of Formula I is determined using an enzymatic PDGFR ⁇ kinase assay (e.g., Promega kinase assay described in Example 131).
  • the enzymatic PDGFR ⁇ kinase assay comprises 20 ng of purified PDGFR ⁇ protein, 150 ⁇ M of ATP and 1 ⁇ g of substrate, Poly (Glu4Tyr1) in a volume of 15 ⁇ l.
  • the compound of Formula I can inhibit PDGFR ⁇ kinase activity with an IC 50 of from about 500 nM to about 0.001 nM, from about 400 nM to about 0.001 nM, from about 300 nM to about 0.001 nM, from about 200 nM to about 0.001 nM, from about 100 nM to about 0.001 nM, from about 75 nM to about 0.001 nM, from about 50 nM to about 0.001 nM, from about 40 nM to about 0.001 nM, from about 30 nM to about 0.001 nM, from about 20 nM to about 0.001 nM, from about 10 nM to about 0.001 nM, from about 5 nM to about 0.001 nM, from about 2.5 nM to about 0.001 nM, from about 1 nM to about 0.001 nM, from about 0.5 nM to about 0.001 n
  • the disclosure provides a compound that can inhibit a PDGFR ⁇ activity of a cell and can further exhibit one or more of the following properties: (i) promote the differentiation of an OPC into an oligodendrocyte, (ii) promote the expression of a protein associated with oligodendrocyte differentiation and/or myelination (e.g., G-protein coupled receptor 17 (GPR17), myelin basic protein (MBP), ASPA, GST-pi, CC1, myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, proteolipid protein 1 (PLP1), or a combination thereof), (iii) promote the myelination of an axon, (iv) promote the remyelination of a demyelinated axon, (v) inhibit PDGFR ⁇ kinase activity, (vii) achieve a brain to plasma ratio of greater than
  • the compound can inhibit PDGFR ⁇ kinase activity with an IC 50 of less than 500 nM, less than 400 nM, less than 300 nM, less than 200 nM, less than 100 nM, less than 75 nM, less than less than 50 nM, less than 40 nM, less than 30 nM, less than 20 nM, less than 10 nM, less than 5 nM, or less than 2 nM.
  • the IC 50 of the PDGFR ⁇ inhibitor is determined using an enzymatic PDGFR ⁇ kinase assay (e.g., PromegaTM kinase assay described in Example 131).
  • the enzymatic PDGFR ⁇ kinase assay comprises 20 ng of purified PDGFR ⁇ protein, 150 ⁇ M of ATP and 1 ⁇ g of substrate, Poly (Glu4Tyr1) in a volume of 15 ⁇ l.
  • the disclosure also provides a pharmaceutical composition comprising a compound disclosed herein, or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable excipient.
  • the disclosure also provides a kit comprising a compound disclosed herein, or a pharmaceutically acceptable salt or solvate thereof, or a composition disclosed herein, and instructions for use.
  • the disclosure also provides a method of producing a PDGFR ⁇ inhibitor comprising synthesizing a compound disclosed herein.
  • the disclosure also provides a compound disclosed herein, or a pharmaceutical composition disclosed herein, for use as a medicament.
  • the disclosure also provides a compound disclosed herein, or a pharmaceutical composition disclosed herein, for use in therapy.
  • the disclosure also provides a method of treating a demyelinating disease in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound disclosed herein or a pharmaceutical composition disclosed herein.
  • the disclosure also provides a method of improving a subject’s performance in a test for assessing one or more symptoms associated with a demyelinating disease, comprising administering to the subject a therapeutically effective amount of a compound disclosed herein or a pharmaceutical composition disclosed herein, wherein after the administration, the subject’s performance in the test is improved as compared to a reference subject (e.g., the subject prior to the administration).
  • the test is selected from one or more of a visual evoked potential (VEP) test, a multifocal visual evoked potential (mfVEP) test, a low contrast visual acuity (LC-VA) test, a magnetic resonance imaging (MRI) (e.g., magnetization transfer ratio (MTR), myelin water fraction (MWF), quantitative susceptibility mapping (QSM), and T2 imaging), an electromyography (EMG), a nerve conduction velocity (NCV) test, an Extended Disability Status Scale (EDSS), a timed walk test (e.g., timed 25-foot walk), a Nine-Hole Peg Test (9HPT), an ocular coherence tomograph (OCT), a quality of life measure test (e.g., Multiple Sclerosis Quality of Life-54 and Vision-Related Quality of Life), cognitive assessment (e.g., Symbol-Digit Modalities Test or Montreal Cognitive Assessment), or combinations thereof.
  • MRI magnetic resonance imaging
  • MMR magnetization transfer ratio
  • the demyelinating disease comprises one or more of an acute disseminated encephalomyelitis (ADEM), acute hemorrhagic leukoencephalitis, acute transverse myelitis, adrenoleukodystrophy, adrenomyeloneuropathy, Alexander disease, Alzheimer’s disease, aminoacidurias, amyotrophic lateral sclerosis, anti-MAG peripheral neuropathy, anti-MOG associated spectrum, Balo concentric sclerosis, brain injury, CAMFAK syndrome, Canavan disease, carbon monoxide toxicity, central pontine myelinolysis, cerebral hypoxia, cerebral ischemia, Charcot–Marie–Tooth disease, chronic inflammatory demyelinating polyneuropathy, chronic traumatic encephalopathy, clinically isolated syndrome (CIS), congenital cataract, copper deficiency associated condition, delayed post-hypoxic leukoencephalopathy, diffuse cerebral sclerosis of Schilder, diffuse myelinoclastic sclerosis
  • ADAM acute diss
  • the demyelinating disease is characterized by demyelination of one or more cells within the CNS of the subject.
  • the demyelinating disease is multiple sclerosis.
  • the multiple sclerosis comprises a clinically isolated syndrome (“CIS”), relapsing-remitting MS (“RRMS”), secondary progressive MS (“SPMS”), primary progressive MS (“PPMS”), optic neuritis or transverse myelitis.
  • the demyelinating disease is an optic neuritis.
  • treating the demyelinating disease comprises reducing one or more symptoms associated with the demyelinating disease.
  • the one or more symptoms comprise one or more of fatigue, dizziness, malaise, elevated fever and high body temperature, extreme sensitivity to cold in the hands and feet, weakness and stiffness in muscles and joints, weight changes, digestive or gastrointestinal problems, low blood pressure, high blood pressure, irritability, anxiety, depression, impaired vision (e.g., blurred vision, double vision, reduction in low contrast visual acuity (LC-VA)), ataxia, clonus, spasms, dysarthria, weakness, clumsiness, hand paralysis, hemiparesis, genital anesthesia, sexual dysfunction, incoordination, paresthesias, ocular paralysis, impaired muscle coordination, loss of sensation, tingling, numbness, pain, neurological symptoms, impaired cognition, unsteady gait, balance problems, dizziness, spastic paraparesis, incontinence, hearing problems, speech problems, loss of olfaction, and agusia.
  • impaired vision e.g., blurred vision,
  • the disclosure also provides a method of promoting the myelination of an axon in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound disclosed herein or a pharmaceutical composition disclosed herein.
  • promoting the myelination of an axon results in an increase in the expression of one or more of the following markers within the subject: myelin basic protein (MBP) Myelin Oligodendrocyte Glycoprotein (MOG), Oligodendrocyte Specific Protein/Claudin-11, CNPase, proteolipid protein 1 (PLP1), or any combination thereof.
  • MBP myelin basic protein
  • MOG Myelin Oligodendrocyte Glycoprotein
  • PGP1 proteolipid protein 1
  • the myelination of an axon can be determined by visualizing and/or quantifying the expression of one or more of the following markers: myelin basic protein (MBP) Myelin Oligodendrocyte Glycoprotein (MOG), Oligodendrocyte Specific Protein/Claudin-11, CNPase, proteolipid protein 1 (PLP1), or any combination thereof.
  • MBP myelin basic protein
  • MOG Myelin Oligodendrocyte Glycoprotein
  • PGP1 proteolipid protein 1
  • the disclosure also provides method of promoting the remyelination of a demyelinated axon in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound disclosed herein or a pharmaceutical composition disclosed herein.
  • promoting the remyelination of a demyelinated axon results in an increase in the expression of one or more of the following markers within the subject: myelin basic protein (MBP) Myelin Oligodendrocyte Glycoprotein (MOG), Oligodendrocyte Specific Protein/Claudin-11, CNPase, proteolipid protein 1 (PLP1), or any combination thereof.
  • MBP myelin basic protein
  • MOG Myelin Oligodendrocyte Glycoprotein
  • PGP1 proteolipid protein 1
  • the remyelination of a demyelinated axon can be determined by visualizing and/or quantifying the expression of one or more of the following markers: myelin basic protein (MBP), Myelin Oligodendrocyte Glycoprotein (MOG), Oligodendrocyte Specific Protein/Claudin-11, CNPase, proteolipid protein 1 (PLP1), or any combination thereof.
  • MBP myelin basic protein
  • MOG Myelin Oligodendrocyte Glycoprotein
  • PGP1 proteolipid protein 1
  • the disclosure also provides a method of reducing the demyelination of a myelinated neuronal axon in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound disclosed herein or a pharmaceutical composition disclosed herein.
  • reducing the demyelination of a myelinated neuronal axon results in an increase in the expression of one or more of the following markers: myelin basic protein (MBP) Myelin Oligodendrocyte Glycoprotein (MOG), Oligodendrocyte Specific Protein/Claudin-11, CNPase, proteolipid protein 1 (PLP1), or any combination thereof.
  • MBP myelin basic protein
  • MOG Myelin Oligodendrocyte Glycoprotein
  • PGP1 proteolipid protein 1
  • the reduction in the demyelination of a myelinated neuronal axon can be determined by visualizing and/or quantifying the expression of one or more of the following markers: myelin basic protein (MBP) Myelin Oligodendrocyte Glycoprotein (MOG), Oligodendrocyte Specific Protein/Claudin-11, CNPase, proteolipid protein 1 (PLP1), or any combination thereof.
  • MBP myelin basic protein
  • MOG Myelin Oligodendrocyte Glycoprotein
  • PGP1 proteolipid protein 1
  • the present disclosure also provides a method of activating an oligodendrocyte progenitor cell (OPC) within the central nervous system (CNS) of a subject in need thereof, the method comprising administering to the subject an effective amount of a compound disclosed herein or a pharmaceutical composition disclosed herein.
  • OPC oligodendrocyte progenitor cell
  • CNS central nervous system
  • the method comprising administering to the subject an effective amount of a compound disclosed herein or a pharmaceutical composition disclosed herein.
  • the subject has, or is at risk of developing a demyelinating disease, for example a disease disclosed herein.
  • the method is a method of treating or preventing a demyelinating disease, for example a disease disclosed herein.
  • the compound or the pharmaceutical composition is administered to the subject once.
  • the compound or the pharmaceutical composition is administered to the subject more than once using intermittent dosing.
  • the intermittent dosing comprises administering the compound or pharmaceutical composition to the subject every other day, every three days, every four days, every five days, every six days, once a week, every eight days, every nine days, every 10 days, every 11 days, every 12 days, every 13 days, once every two weeks, once every three weeks, once a month, once every two months, once every three months, once every four months, once every five months, once every six months, or once every twelve months.
  • the intermittent dosing comprises administering to the subject a first dose and a second dose of the compound or pharmaceutical composition, wherein the second dose is administered at least one day, at least two days, at least three days, at least four days, at least five days, at least six days, at least seven days, at least eight days, at least nine days, at least 10 days, at least 11 days, at least 12 days, at least 13 days, at least two weeks, at least three weeks, at least one month, at least two months, at least three months, at least four months, at least five months, at least six months, or at least 12 months after administering the first dose.
  • the second dose is administered to the subject one day, two days, three days, four days, five days, six days, seven days, eight days, nine days, 10 days, 11 days, 12 days, 13 days, two weeks, three weeks, one month, two months, three months, four months, five months, six months, or 12 months after administering the first dose.
  • the compound or the pharmaceutical composition can achieve a brain to plasma ratio of greater than 0.1, greater than 0.2, greater than 0.3, greater than 0.4, greater than 0.5, greater than 0.6, greater than 0.7, greater than 0.8, greater than 0.9, greater than 1.0, greater than 1.1, greater than 1.2, greater than 1.3, greater than 1.4, greater than 1.5, greater than 1.6, greater than 1.7, greater than 1.8, greater than 1.9, or greater than 2.0.
  • the brain to plasma ratio may be assessed in preclinical species using bioanalytical gas or liquid chromatography and mass spectrometry methods.
  • the method further comprises administering to the subject an additional therapeutic agent.
  • the additional therapeutic agent comprises a standard care of treatment.
  • the additional therapeutic agent comprises an immunomodulatory agent.
  • the additional therapeutic agent is selected from interferon beta- 1b, interferon beta-1a, peginterferon beta-1a, alemtuzumab, natalizumab, ocrelizumab, ofatumumab, ublituximab-xiiy, glatiramer acetate, teriflunomide, dimethyl fumarate, monomethyl fumarate, diroximel fumarate, fingolimod hydrochloride, siponimod fumaric acid, ozanimod hydrochloride, ponesimod, cladribine, mitoxantrone, a BTK inhibitor (e.g., masitinib, evobrutinib, or tolebrutinib), a statin (e.g., simvastat
  • the additional therapeutic agent is administered to the subject prior to, concurrently, or after the administration of the compound or the pharmaceutical composition.
  • the disclosure also provides a method of inducing the differentiation of an oligodendrocyte progenitor cell (OPC) into an oligodendrocyte, the method comprising contacting the OPC with an effective amount of a compound disclosed herein or a pharmaceutical composition disclosed herein.
  • OPC oligodendrocyte progenitor cell
  • inducing the differentiation of the OPC into an oligodendrocyte results in an increase in the expression of the following markers in the subject: GPR17, MBP, ASPA, GST-pi, CC1, myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, proteolipid protein 1 (PLP1), or a combination thereof.
  • GPR17 GPR17
  • MBP ASPA
  • GST-pi GST-pi
  • CC1 myelin oligodendrocyte glycoprotein
  • oligodendrocyte-specific protein/claudin-11 oligodendrocyte-specific protein/claudin-11
  • CNPase proteolipid protein 1
  • PGP1 proteolipid protein 1
  • the differentiation of the OPC into an oligodendrocyte is measured by determining the expression of GPR17, MBP, ASPA, GST-pi, CC1, myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, proteolipid protein 1 (PLP1), or a combination thereof.
  • the disclosure also provides a method of inhibiting PDGFR ⁇ activity in a cell, the method comprising contacting the cell with an effective amount of a compound disclosed herein or a pharmaceutical composition disclosed herein.
  • the inhibition of the PDGFR ⁇ activity is measured by one or more of the following: an in vitro OPC differentiation assay (e.g., as described in Example 132), a cuprizone model for demyelination, an in vivo OPC differentiation assay (e.g., as described in Example 134), an enzymatic PDGFR ⁇ kinase assay (e.g., as described in Example 131), or any combination thereof.
  • the contacting occurs ex vivo or in vivo.
  • the method is a method of treating by a therapy.
  • the disclosure also provides a method of treating a PDGF-associated tumor in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound disclosed herein or a pharmaceutical composition disclosed herein, wherein after the administration, PDGFR ⁇ activity is reduced in the subject.
  • the PDGF-associated tumor comprises an oligodendroglioma.
  • the method is a method of treatment by therapy.
  • the present disclosure describes compounds that can induce the differentiation of OPCs into cells that have features of mature oligodendrocytes, including morphological characteristics and the protein expression patterns associated with myelination, and can also affect remyelination. Accordingly, as described herein, the compounds of the present disclosure can be useful in treating various diseases, such as those associated with demyelination. Additional aspects of the present disclosure are provided throughout the present application. [0110] Before the present disclosure is described in greater detail, it is to be understood that this disclosure is not limited to the particular compositions or process steps described, which can, of course, vary.
  • 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).
  • 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).
  • ranges recited are understood to be shorthand for all of the values within the range, inclusive of the recited endpoints.
  • a range of 1 to 10 is understood to include any number, combination of numbers, or sub-range from the group consisting of 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10.
  • halo as used herein by itself or as part of another group refers to Cl, F, Br, or I.
  • nitro as used herein by itself or as part of another group refers to NO 2 .
  • cyano as used herein by itself or as part of another group refers to CN.
  • hydroxy as herein used by itself or as part of another group refers to OH.
  • alkyl refers to a straight- or branched-chain aliphatic hydrocarbon containing one to twelve carbon atoms, i.e., a C 1 -C 12 alkyl, or the number of carbon atoms designated, e.g., a C 1 alkyl such as methyl, a C 2 alkyl such as ethyl, etc.
  • the alkyl is a C 1 -C 10 alkyl.
  • the alkyl is a C 1 -C 6 alkyl.
  • the alkyl is a C 1 -C 4 alkyl.
  • the alkyl is a C 1 -C3alkyl, i.e., methyl, ethyl, propyl, or isopropyl.
  • Non limiting exemplary C 1 - C 12 alkyl groups include methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, tert-butyl, iso- butyl, 3-pentyl, hexyl, heptyl, octyl, nonyl, and decyl.
  • one or more of the hydrogen atoms of the alkyl group are replaced by deuterium atoms, i.e., the alkyl group is isotopically-labeled with deuterium.
  • a non-limiting exemplary deuterated alkyl group is - CD3.
  • the term “haloalkyl” as used herein by itself or as part of another group refers to an alkyl group wherein one or more hydrogen atoms of the alkyl group are replaced by halo atoms. In some aspects, the haloalkyl group is a -CF 3 group.
  • alkoxy as used herein by itself or as part of another group refers to an alkyl group attached to a terminal oxygen atom.
  • the alkyl is a C 1 -C 8 alkyl and resulting alkoxy is thus referred to as a “C 1 -C 8 alkoxy.”
  • the alkyl is a C 1 -C 4 alkyl group.
  • Non-limiting exemplary alkoxy groups include methoxy, ethoxy, and tert-butoxy.
  • alkyoxyalkyl as used herein by itself or as part of another group refers to an alkyl group substituted with an alkoxyl group.
  • the alkyl is a C 1 -C 4 alkyl and the alkoxy is a C 1 -C 4 alkoxyl and the resulting alkoxyalkyl is thus referred to as a “C 1 -C 4 alkoxyC 1 -C 4 alkyl”.
  • amino refers to -NH2, which may be optionally substituted with one or two alkyl, two alkyl linked to form a ring, haloalkyl, (hydroxy)alkyl, (alkoxy)alkyl, (amino)alkyl, heteroalkyl, optionally substituted cycloalkyl, optionally substituted heterocyclo, optionally substituted aryl, optionally substituted heteroaryl, (aryl)alkyl, (cycloalkyl)alkyl, (heterocyclo)alkyl, or (heteroaryl)alkyl.
  • heterocyclycylamino refers to an amino group substituted with a heterocyclyl group.
  • aminoalkyl refers to an alkyl group substituted with an amino group. In some aspects, the alkyl is a C 1 -C 4 alkyl and the resulting aminoalkyl is thus referred to as an “aminoC 1 -C 4 alkyl”.
  • hydroxyalkyl refers to an alkyl group substituted with a hydroxy group.
  • the alkyl is a C 1 - C 4 alkyl and the resulting hydroxyalkyl is thus referred to as a “hydroxyC 1 -C 4 alkyl”.
  • aminoalkylamino as used herein by itself or as part of another group refers to an amino group substituted with an aminoalkyl group. In some aspects, the aminoalkylamino group is -NHCH2CH2NH2.
  • oxo as used herein refers to an oxygen atom that is connected to a carbon atom by a double bond, i.e. to form a keto group.
  • cycloalkyl refers to saturated and partially unsaturated, e.g., containing one or two double bonds, monocyclic, bicyclic, or tricyclic aliphatic hydrocarbons containing three to twelve carbon atoms, i.e., a C 3 -C 12 cycloalkyl, or the number of carbons designated, e.g., a C 3 cycloalkyl such a cyclopropyl, a C 4 cycloalkyl such as cyclobutyl, etc.
  • the cycloalkyl is bicyclic, i.e., it has two rings.
  • the cycloalkyl is monocyclic, i.e., it has one ring. In some aspects, the cycloalkyl is a C 3 -C 8 cycloalkyl. In some aspects, the cycloalkyl is a C 3-6 cycloalkyl, i.e., cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl. In some aspects, the cycloalkyl is a C 5 cycloalkyl, i.e., cyclopentyl or cyclopentenyl.
  • the cycloalkyl is a C 6 cycloalkyl, i.e., cyclohexyl or cyclohexenyl.
  • a cycloalkyl group containing one or two double bonds may also be referred to as a “cycloalkenyl” group.
  • the term “heterocyclyl” as used herein by itself or as part of another group refers to saturated and partially unsaturated, e.g., containing one or two double bonds, monocyclic, bicyclic, or tricyclic groups containing three to fourteen ring members, i.e., a 3- to 14-membered heterocyclyl, comprising one, two, three, or four heteroatoms.
  • heterocyclylaryl as used herein by itself or as part of another group refers to an aryl group that is substituted with a heterocyclyl group.
  • arylC 1 -C 4 alkyl refers to a C 1 -C 4 alkyl group substituted with an aryl group.
  • heteroarylC 1 -C 4 alkyl refers to a C 1 -C 4 alkyl group substituted with a heteroaryl group.
  • heterocyclylC 1 -C 4 alkyl refers to a C 1 -C 4 alkyl group substituted with a heterocyclyl group.
  • cycloalkylC 1 -C 4 alkyl refers to a C 1 -C 4 alkyl group substituted with a cycloalkyl group.
  • alkylsulfonyl refers to a sulfonyl group, i.e., -SO 2 -, substituted by an alkyl group.
  • a non-limiting exemplary alkylsulfonyl group is -SO 2 CH 3 .
  • aryl refers to an aromatic ring system having six to fourteen carbon atoms, i.e., C 6 -C 14 aryl.
  • Non-limiting exemplary aryl groups include phenyl (abbreviated as “Ph”), naphthyl, phenanthryl, anthracyl, indenyl, azulenyl, biphenyl, biphenylenyl, and fluorenyl groups.
  • the aryl group is phenyl.
  • heteroaryl refers to monocyclic and bicyclic aromatic ring systems having five to fourteen ring members, i.e., a 5- to 14-membered heteroaryl, comprising one, two, three, or four heteroatoms.
  • Each heteroatom is independently oxygen, sulfur, or nitrogen.
  • the heteroaryl has three heteroatoms.
  • the heteroaryl has two heteroatoms.
  • the heteroaryl has one heteroatom.
  • the heteroaryl is a 5- to 10- membered heteroaryl.
  • the heteroaryl has 5 ring atoms, e.g., thienyl, a 5- membered heteroaryl having four carbon atoms and one sulfur atom. In some aspects, the heteroaryl has 6 ring atoms, e.g., pyridyl, a 6-membered heteroaryl having five carbon atoms and one nitrogen atom.
  • Non-limiting exemplary heteroaryl groups include thienyl, benzo[b]thienyl, naphtho[2,3-b]thienyl, thianthrenyl, furyl, benzofuryl, pyranyl, isobenzofuranyl, benzoxazolyl, chromenyl, xanthenyl, 2H-pyrrolyl, pyrrolyl, imidazolyl, pyrazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, isoindolyl, 3H-indolyl, indolyl, indazolyl, purinyl, isoquinolyl, quinolyl, phthalazinyl, naphthyridinyl, cinnolinyl, quinazolinyl, pteridinyl, 4aH-carbazolyl, carbazolyl, ⁇ -carbolinyl,
  • heteroaryl also includes N-oxides.
  • a non-limiting exemplary N-oxide is pyridyl N-oxide.
  • aminoheterocyclyl as used herein by itself or as part of another group refers to a heterocyclyl group substituted with an optionally substituted amino group.
  • Non- limiting exemplary aminoheterocyclyl groups include: .
  • the chemical terms used herein may be combined to describe larger substituents. For a monovalent substituent, the substituent that is recited last in the combined term is the substituent that contains the attachment point.
  • an “arylC 1 -C 4 alkyl” group contains an attachment point on the alkyl group
  • an “C 1 -C 4 alkylaryl” group contains an attachment point on the aryl group.
  • the present disclosure encompasses any of the disclosed compounds being isotopically-labelled (i.e., radiolabeled) by having one or more atoms replaced by an atom having a different atomic mass or mass number.
  • isotopes that can be incorporated into the disclosed compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine and chlorine, such as 1 H, 2 H (or deuterium (D)), 3 H, 11 C, 1 2 C, 13 C, 14 C, 14 N 15 N, 18 O, 17 O, 16 O 31 P, 32 P, 32 S, 33 S, 34 S, 35 S, 36 S, 18 F, 19 F, 35 Cl, 37 Cl, and 3 6 Cl, respectively, e.g., 3 H, 11 C, and 14 C.
  • a composition wherein substantially all of the atoms at a position within the disclosed compound are replaced by an atom having a different atomic mass or mass number.
  • compositions wherein a portion of the atoms at a position within the disclosed compound are replaced, i.e., the disclosed compound is enriched at a position with an atom having a different atomic mass or mass number.
  • Isotopically-labelled disclosed compounds can be prepared by methods known in the art.
  • the present disclosure also encompasses any of the disclosed compounds wherein a quaternary carbon atom is replaced with a silicon atom.
  • the compounds disclosed herein contain one or more asymmetric carbon atoms and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms.
  • the present disclosure encompasses the use of all such possible forms, as well as their racemic and resolved forms and mixtures thereof.
  • the individual enantiomers can be separated according to methods known in the art in view of the present disclosure.
  • the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that they include both E and Z geometric isomers. All tautomers are also encompassed by the present disclosure.
  • the term “stereoisomers” is a general term for all isomers of individual molecules that differ only in the orientation of their atoms in space. It includes enantiomers and isomers of compounds with more than one chiral center that are not mirror images of one another (diastereomers).
  • chiral center or “asymmetric carbon atom” refers to a carbon atom to which four different groups are attached.
  • enantiomer and enantiomeric refer to a molecule that cannot be superimposed on its mirror image and hence is optically active wherein the enantiomer rotates the plane of polarized light in one direction and its mirror image compound rotates the plane of polarized light in the opposite direction.
  • racemic refers to a mixture of equal parts of enantiomers and which mixture is optically inactive. In some aspects, the compounds disclosed are racemic.
  • absolute configuration refers to the spatial arrangement of the atoms of a chiral molecular entity (or group) and its stereochemical description, e.g., R or S.
  • stereochemical terms and conventions used in the specification are meant to be consistent with those described in Pure & Appl. Chem 68:2193 (1996), unless otherwise indicated.
  • enantiomeric excess or “ee” refers to a measure for how much of one enantiomer is present compared to the other.
  • the percent enantiomeric excess is defined as ([ ⁇ ]obs/[ ⁇ ]max)*100, where [ ⁇ ]obs is the optical rotation of the mixture of enantiomers and [ ⁇ ]max is the optical rotation of the pure enantiomer. Determination of enantiomeric excess is possible using a variety of analytical techniques, including NMR spectroscopy, chiral column chromatography or optical polarimetry.
  • the terms “administration,” “administering,” and grammatical variants thereof, refer to introducing a composition, such as a PDGFR ⁇ inhibitor of the present disclosure, into a subject via a pharmaceutically acceptable route. Any suitable route of administration can be used in administering the PDGFR ⁇ inhibitors described herein to a subject. Non- limiting examples of such routes of administration are provided elsewhere in the present disclosure.
  • the term “central nervous system” or “CNS” refers to a complex of nerve tissues that control the various activities of the body (e.g., voluntary and involuntary movements) and the mind (e.g., thoughts, perceptions, and emotions).
  • the CNS generally consists of the brain and the spinal cord.
  • the term “demyelinating disease” refers to any disorder of the nervous system in which there is reduced myelination, including disorders in which insufficient or dysfunctional myelin (e.g., hypomyelination) is generated during development or disorders in which the myelin sheath of neurons is damaged.
  • myelin and Myelin sheath refer to the specialized membrane formed by oligodendrocytes that insulates the axons of neurons. The insulation provided by the myelin sheath helps to increase the rate of transmission of nerve signals along the axon, thereby promoting timely and energetically efficient neuronal signaling.
  • Myelin also provides metabolic support of the axons, maintaining their health and survival.
  • the neuronal axon of a subject suffering from or at risk of developing a demyelinating disease is completely demyelinated.
  • the neuronal axon of a subject suffering from or at risk of developing a demyelinating disease is partially demyelinated (e.g., at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, or at least 95% compared to a reference neuronal axon that is fully myelinated).
  • the term “demyelination” comprises any of the following: complete hypomyelination, complete demyelination, partial hypomyelination, partial demyelination, and combinations thereof.
  • the demyelinating diseases described herein are associated with impaired conduction of nerve signals or axonal or neuronal survival, which can, in turn, cause deficiencies in sensation, movement, cognition, or other functions depending on which neurons are affected.
  • demyelinating diseases are provided elsewhere in the present disclosure.
  • under myelination refers to a cell, tissue, or subject that lacks normal levels of myelination (e.g., level of myelination observed in a corresponding cell, tissue, or subject who does not have a demyelinating disease), regardless of etiology.
  • hypomyelination refers to a deficiency in myelin for any reason (e.g., body is unable to produce myelin at normal levels). Unless indicated otherwise, hypomyelination includes demyelination (related to myelin destruction) and dysmyelination (related to abnormal myelin deposition).
  • hypomyelination includes diseases in which insufficient myelin is generated during development as well as diseases associated with demyelination and/or dysmyelination.
  • a reference e.g., corresponding tissue, e.g., brain, of a subject who does not suffer from a demyelinating disease
  • hypomyelination of all i.e., “complete hypomyelination”
  • partial hypomyelination for example the brain (e.g., hypomyelination in white matter and/or gray matter), of at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, or at least 95%.
  • neuron includes electrically excitable cells that process and transmit information through electrical and chemical signals.
  • Neurons are major components of the brain and spinal cord of the CNS, and of the ganglia of the peripheral nervous system (PNS) and can connect to each other to form neural networks.
  • a typical neuron is composed of a cell body (soma), dendrites, and an axon.
  • the “soma” (the cell body) of a neuron contains the nucleus.
  • the “dendrites” of a neuron are cellular extensions generally having many branches, where the majority of input to the neuron occurs.
  • oligodendrocyte progenitor cells also known in the art as “oligodendrocyte precursor cells,” “polydendrocytes,” “NG2 cells,” and “O-2A cells” refer to a subtype of glial cells in the central nervous system.
  • oligodendrocytes also known as “oligodendroglia” which are responsible for generating the myelin sheath that wraps around axons, providing insulation, aiding electrical conduction, and providing metabolic support. Nerve impulses can travel up to 200 times faster along a myelinated axon compared to an unmyelinated axon.
  • OPCs and immature oligodendrocytes are generally positive for the following markers: A2B5, neuron-glial antigen 2 (NG2), and PDGFR ⁇ . Other suitable markers that can be used are known in the art.
  • the term “remyelination” refers to generation of new myelin sheaths around demyelinated (e.g., including hypomyelinated) axons.
  • the remyelination process involves the differentiation of OPCs into oligodendrocytes that generate functional myelin sheaths around demyelinated axons. Remyelination of the axons can restore action potential conduction properties to axons, and thereby, promote and/or improve neurological function. Further, remyelination can provide metabolic support to axons, preventing their damage or loss.
  • “remyelination” refers to any aspect of a process that can result in remyelination.
  • “remyelination” comprises the migration or colonization of OPCs to sites of demyelinated axons.
  • “remyelination” comprises the differentiation of OPCs into oligodendrocytes.
  • “remyelination” comprises the generation of myelin sheaths by oligodendrocytes around demyelinated axons.
  • the term “remyelination” comprises any combination of the following: (i) migration or colonization of OPCs to sites of demyelinated axons; (ii) differentiation of OPCs into oligodendrocytes; and (iii) generation of myelin sheaths by oligodendrocytes around demyelinated (e.g., including hypomyelinated) axons.
  • the term “restore” (and derivatives thereof) comprises both complete restoration and partial restoration.
  • remyelinating a demyelinated axon restores conduction properties to the axon, such that the conduction properties are the same as those of the axon prior to the demyelination (i.e., complete restoration).
  • remyelinating a demyelinated axon restores conduction properties to the axon, where the conduction properties are improved but not the same as those of the axon prior to the demyelination (i.e., partial restoration).
  • remyelination provides metabolic support to axons, preventing their damage or loss.
  • remyelination provides partial metabolic support to axons, delaying their damage or loss.
  • the term “promoting” refers to the ability of an agent (e.g., PDGFR ⁇ inhibitor described herein) to induce or increase a particular result (e.g., remyelination of a demyelinated axon). In some aspects, the term comprises both inducing and increasing a particular result.
  • the term “subject” refers to any animal subject including a human, a laboratory animal (e.g., a non-human primate, rat, and mouse), livestock (e.g., cow, sheep, goat, pig, turkey, and chicken), and household pets (e.g., dog, cat, and rodent).
  • a laboratory animal e.g., a non-human primate, rat, and mouse
  • livestock e.g., cow, sheep, goat, pig, turkey, and chicken
  • household pets e.g., dog, cat, and rodent.
  • treat refers to any type of intervention or process performed on, or administering an active agent (e.g., PDGFR ⁇ inhibitor described herein) to, the subject with the objective of reversing, alleviating, ameliorating, inhibiting, or slowing down; or preventing the progression, development, severity or recurrence of a symptom, complication, condition or one or more biochemical indicia associated with a disease, or enhancing overall survival.
  • an active agent e.g., PDGFR ⁇ inhibitor described herein
  • treatment can be of a subject having a disease (e.g., exhibiting one or more symptoms associated with the disease).
  • treatment can be of a subject with some degree of demyelination but not yet exhibiting any symptoms associated with the disease.
  • administering a PDGFR ⁇ inhibitor of the present disclosure can help delay or prevent the onset of symptoms associated with the disease.
  • effective dose or “effective amount” is defined as an amount sufficient to achieve or at least partially achieve a desired effect (e.g., induce remyelination of a demyelinated neuronal axon).
  • a “therapeutically effective amount” or “therapeutically effective dosage” of a therapeutic agent is any amount of the therapeutic agent that, when used alone or in combination with another therapeutic agent, promotes disease regression as evidenced by a decrease in severity of disease symptoms, an increase in frequency and duration of disease symptom-free periods, a prevention of impairment or disability due to the disease affliction, or a reduction in disease progression.
  • a therapeutically effective amount or dosage of a therapeutic agent includes a “prophylactically effective amount” or a “prophylactically effective dosage,” which is any amount of the therapeutic agent that, when administered alone or in combination with another therapeutic agent to a subject at risk of developing a disease or of suffering a recurrence of disease, inhibits the development or recurrence of the disease.
  • a therapeutic agent to promote disease reversal or inhibit the development, progression or recurrence of the disease can be evaluated using a variety of methods known to the skilled practitioner, such as in human subjects during clinical trials, in animal model systems predictive of efficacy in humans, or by assaying the activity of the agent in in vitro assays.
  • the term “dosing interval” refers to the amount of time that elapses between multiple (e.g., 2 or more) doses of a PDGFR ⁇ inhibitor described herein.
  • a PDGFR ⁇ inhibitor provided herein exerts its therapeutic effect by inducing the differentiation of oligodendrocyte progenitor cells (OPCs) into oligodendrocytes.
  • OPCs oligodendrocyte progenitor cells
  • a dosing interval that is suitable for the present disclosure is the amount of time required for a subject’s OPC population to have sufficiently recovered after the administration of an initial dose of a PDGFR ⁇ inhibitor provided herein, or a period longer than that, such that the administration of a second (or additional dose) of the PDGFR ⁇ inhibitor would have a therapeutic effect (e.g., such as those described herein, e.g., increases the number of differentiated oligodendrocytes) in the subject.
  • a PDGFR ⁇ inhibitor described herein is administered to a subject at a dosing interval, wherein the dosing interval is the amount of time required, after the administration of an initial dose of the PDGFR ⁇ inhibitor, for the subject’s OPC population (e.g., in size) to be at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 100% to that of a reference OPC population (e.g., the subject’s OPC population prior to the initial administration of the PDGFR ⁇ inhibitor).
  • OPC population e.g., in size
  • an OPC population may be measured by quantifying the density of NG2+ (neuron-glia antigen 2/chondroitin sulfate proteoglycan-4) or PDGFRa+ cells in mice, rats, pigs, dogs, non-human primates or other species using immunohistochemistry or fluorescence-activated cell sorting. Repopulation may be calculated as the OPC cell density at a given time after dosing relative to the OPC population in an untreated reference animal.
  • a suitable dosing interval for the present disclosure comprises the amount of time required, after an initial dose of the PDGFR ⁇ inhibitor, for the plasma level of the PDGFR ⁇ inhibitor to reach less than 50%, less than 40%, less than 30%, less than 20%, less than 10%, or less than 5%, as compared to a reference (e.g., plasma level of the PDGFR ⁇ inhibitor in a corresponding subject immediately after, e.g., about 4 hours after, the administration of the initial dose of the PDGFR ⁇ inhibitor), a suitable dosing interval is the time between administering the first dose of the PDGFR ⁇ inhibitor and the subject’s plasma level of the PDGFR ⁇ inhibitor reaching a reduced level as compared to the reference.
  • a reference e.g., plasma level of the PDGFR ⁇ inhibitor in a corresponding subject immediately after, e.g., about 4 hours after, the administration of the initial dose of the PDGFR ⁇ inhibitor
  • the additional dose of the PDGFR ⁇ inhibitor is administered to the subject when the subject’s plasma level has decreased by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or about 100% as compared to the reference.
  • the brain to plasma ratio may be assessed in preclinical species using bioanalytical gas or liquid chromatography and mass spectrometry methods.
  • binds As used herein, the terms “binds,” “recognizes,” “targets” are analogous terms and refer to molecules (e.g., PDGFR ⁇ inhibitors described herein) that can bind and/or target a particular region of a protein (e.g., PDGFR ⁇ ), as such binding and/or targeting is understood by one skilled in the art.
  • a PDGFR ⁇ inhibitor is a small molecule that can target and inhibit the kinase portion of PDGFR ⁇ (e.g., by binding to the ATP binding site, by allosterically interfering with PDGFR ⁇ kinase activity, or both), and thereby, inhibit its activity.
  • the above terms i.e., binds, recognizes, and targets
  • PDGFR ⁇ any binding or targeting of PDGFR ⁇ , such that its activity is reduced and/or inhibited (e.g., binding of a small molecule to an ATP binding site of PDGFR ⁇ , binding of a small molecule to a substrate binding site of PDGFR ⁇ , allosterically interfering with PDGFR ⁇ kinase activity, or a combination thereof).
  • “Potency” is an expression of the activity of a therapeutic agent in terms of the amount or concentration of the therapeutic agent that achieves a desired effect. Accordingly, functional assays, such as those described herein, can be used to identify compounds useful for the present disclosure.
  • Molecules that “compete with another protein or compound for binding to a target” refers to molecules that inhibit (partially or completely) the binding of the other protein (e.g., naturally existing PDGFR ⁇ ligand) to the target (e.g., PDGFR ⁇ ). Whether two compounds compete with each other for binding to a target, i.e., whether and to what extent a PDGFR ⁇ inhibitor described herein inhibits the binding of the naturally existing ligand to a PDGFR ⁇ , can be determined using known competition experiments.
  • a PDGFR ⁇ inhibitor described herein competes with, and inhibits the binding of the naturally existing ligand to PDGFR ⁇ by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or 100%.
  • Competition assays can be conducted as described herein or, for example, in Ed Harlow and David Lane, Cold Spring Harb. Protoc.; 2006; doi: 10.1101/pdb.prot4277 or in Chapter 11 of “Using Antibodies” by Ed Harlow and David Lane, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, USA 1999.
  • Additional non-limiting examples of other competitive binding assays include: solid phase direct or indirect radioimmunoassay (RIA), solid phase direct or indirect enzyme immunoassay (EIA), sandwich competition assay (see Stahli et al., Methods in Enzymology 9:242 (1983)); solid phase direct biotin- avidin EIA (see Kirkland et al., J. Immunol.137:3614 (1986)); solid phase direct labeled assay, solid phase direct labeled sandwich assay (see Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Press, Second Edition (2014)); solid phase direct label RIA using 1-125 label (see Morel et al., Mol.
  • RIA solid phase direct or indirect radioimmunoassay
  • EIA enzyme immunoassay
  • sandwich competition assay see Stahli et al., Methods in Enzymology 9:242 (1983)
  • solid phase direct biotin- avidin EIA see Kirkland et al.
  • the antagonistic activity of a PDGFR ⁇ inhibitor described herein does not depend on interfering with the binding of another protein or compound to PDGFR ⁇ .
  • Another non-limiting example includes small molecules that can bind to PDGFR ⁇ and inhibit (partially or completely) binding of ATP or substrate, or otherwise inhibit PDGFR ⁇ activity.
  • a PDGFR ⁇ inhibitor described herein can inhibit or reduce PDGFR ⁇ activity via allosteric inhibition. In some aspects, a PDGFR ⁇ inhibitor can reduce protein levels of PDGFR ⁇ by reducing synthesis or enhancing degradation. While exemplary mechanisms of actions are provided, it will be apparent from the present disclosure that the PDGFR ⁇ inhibitors described herein are not intended to be limited to such mechanisms. [0173] As used herein, the terms “ug” and “uM” are used interchangeably with “ ⁇ g” and “ ⁇ ,” respectively. [0174] Various aspects described herein are described in further detail in the following subsections. The various aspects and embodiments of the present disclosure can optionally be combined.
  • any of the disclosed compounds can be used in any of the disclosed methods and uses.
  • PDGFR ⁇ Inhibitors Disclosed herein are compounds that can inhibit PDGFR ⁇ activity and/or elicit one or more activities associated with inhibition of the receptor. PDGFR ⁇ can bind with high affinity to several PDGF isoforms (i.e., PDGF-A, PDGF-B, PDGF-C, and PDGF-D). As used herein, references to inhibition of PDGFR ⁇ binding to its ligand refer to inhibition of binding of one or more of the PDGF isoforms that bind to PDGFR ⁇ .
  • the binding of all isoforms of PDGF that bind to PDGFR ⁇ is inhibited.
  • the binding of the platelet-derived growth factor (PDGF) isoforms to PDGFR ⁇ affects various cell signaling pathways, such as those involved in cell proliferation and differentiation. Accordingly, PDGFR ⁇ activity is critical for both the proper development and long-term maintenance of certain tissues and organs. For example, animals globally lacking a functional PDGFRA gene die at birth or soon thereafter due to various developmental defects (e.g., cardiac malformations). Bax et al., Dev. Dyn. 239(8): 2307-2317 (Aug. 2010).
  • PDGFR ⁇ knockout in OPCs during animal development results in severe hypomyelination and the animals die soon after birth.
  • PDGFRA the gene encoding PDGFR ⁇ (i.e., PDGFRA) is located on chromosome 4 (for example, nucleotides 54,229,127-54,298,245 of GenBank Accession Number NC_000004.12; plus strand orientation).
  • PDGFR ⁇ includes any variants or isoforms of PDGFR ⁇ that are naturally expressed by cells.
  • inhibiting or reducing PDGFR ⁇ activity for example, with one or more of the compounds described herein, can have certain biological effects that can be useful for treating demyelinating diseases, such as those described herein.
  • inhibiting or reducing PDGFR ⁇ activity with the PDGFR ⁇ inhibitors described herein can promote oligodendrocyte differentiation and induce myelin formation.
  • the PDGFR ⁇ inhibitors of the present disclosure are capable of (and in some embodiments used for) promoting remyelination of demyelinated neuronal axons, which can be useful in the treatment of various demyelinating diseases (e.g., multiple sclerosis).
  • demyelinating diseases e.g., multiple sclerosis
  • the terms “PDGFR ⁇ inhibitors” and “PDGFR ⁇ antagonists” are used interchangeably and refer to any compound that is capable of (and in some embodiments used for) reducing and/or inhibiting PDGFR ⁇ activity (e.g., by any of the exemplary mechanisms or methods described herein).
  • a PDGFR ⁇ inhibitor provided herein can inhibit a tyrosine kinase activity of a PDGFR ⁇ .
  • a PDGFR ⁇ inhibitor provided herein can inhibit any other activity of a PDGFR ⁇ .
  • the term “capable of” is used herein to describe certain features of a PDGFR ⁇ inhibitor, the term means that the PDGFR ⁇ inhibitor can (i.e., has the ability to) exhibit such features, e.g., under appropriate conditions. Unless indicated otherwise, the term does not mean that the PDGFR ⁇ inhibitor always exhibits such features, e.g., when administered to a subject with impaired oligodendrocyte progenitor cell population.
  • the term “capable of” and “can” are used interchangeably in the present application.
  • a PDGFR ⁇ inhibitor inhibits the kinase activity of PDGFR ⁇ .
  • the term “inhibit” comprises both complete inhibition and partial inhibition (e.g., reduced kinase activity). Accordingly, in some aspects, when an OPC (expressing PDGFR ⁇ ) interacts with a PDGFR ⁇ inhibitor described herein, the PDGFR ⁇ kinase activity is reduced by at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90% or more (for example, at least 50% or more), compared to PDGFR ⁇ kinase activity in a corresponding OPC that was not contacted with the PDGFR ⁇ inhibitor described herein (e.g., contacted with a vehicle control).
  • the kinase activity is completely inhibited.
  • the inhibition (partial or complete) of PDGFR ⁇ kinase activity in an OPC can promote the differentiation of the OPC into a myelinating oligodendrocyte. Inhibition of PDGFR ⁇ kinase activity can be assayed using methods known in the art. In some aspects, the inhibition of PDGFR ⁇ kinase activity can be determined using the in vitro differentiation assay described in Example 132 of the present application.
  • the inhibition of PDGFR ⁇ kinase activity can be assessed using a cuprizone model for demyelination, such as that described in Torkildsen et al., Acta Neurol Scand Suppl 188:72-6 (2008).
  • the inhibition of PDGFR ⁇ kinase activity is determined using an enzymatic PDGFR ⁇ kinase assay, such as that described in Example 131 of the present disclosure.
  • the inhibition of PDGFR ⁇ kinase activity can be determined using an in vivo OPC differentiation assay, such as that described in Example 134.
  • a PDGFR ⁇ inhibitor useful for the present disclosure comprises one or more of the following properties: (1) capable of (and in some embodiments used for) promoting (e.g., inducing and/or enhancing) the differentiation of an OPC into an oligodendrocyte (in vitro, in vivo, or both); (2) capable of (and in some embodiments used for) promoting (e.g., inducing and/or enhancing) the expression of a protein associated with oligodendrocyte differentiation and/or myelination (e.g., G-protein coupled receptor 17 (GPR17), myelin basic protein (MBP), ASPA, GST-pi, CC1, myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, proteolipid protein 1 (PLP1), or a combination thereof); (3) capable of (and in some embodiments used for) promoting (e.g., GPR17), myelin basic protein
  • a PDGFR ⁇ inhibitor described herein can comprise one or more additional features that are not present in other inhibitors known in the art. As is apparent from the present disclosure, such additional features can be useful in various clinical settings, e.g., to treat a demyelinating disease, such as those described herein.
  • some exemplary PDGFR ⁇ inhibitors of the present disclosure are capable of have greater therapeutic effect than inhibitors known in the art, e.g., when an inhibitor known in the art and an exemplary inhibitor provided herein are assayed against a vehicle control.
  • a PDGFR ⁇ inhibitor can have greater potency compared to inhibitors known in the art.
  • a PDGFR ⁇ inhibitor can better penetrate the CNS (e.g., can more effectively pass across the blood-brain barrier).
  • PDGFR ⁇ inhibitors described herein are more effective at promoting OPC differentiation as compared to inhibitors known in the art.
  • a PDGFR ⁇ inhibitor of the present disclosure can achieve greater than 2-fold increase in GPR17 expression compared to a vehicle control at a dose of less than 50 mg/kg, as measured using an in vivo GPR17 assay (such as that described in Example 134).
  • a PDGFR ⁇ inhibitor of the present disclosure can achieve greater than 2-fold increase in GPR17 expression compared to the vehicle control at a dose of less than 40 mg/kg, less than 30 mg/kg, less than 20 mg/kg, less than 10 mg/kg, less than 5 mg/kg, less than 4 mg/kg, less than 3 mg/kg, less than 2 mg/kg, or less than 1 mg/kg, as measured using an in vivo GPR17 assay (such as that described in Example 134).
  • GPR17 expression is a suitable marker for OPC differentiation.
  • any of the other proteins associated with oligodendrocyte differentiation and/or myelination can be used instead of GPR17 or in combination with GPR17.
  • Non- limiting examples of such proteins include: MBP, ASPA, GST-pi, CC1, MOG, oligodendrocyte-specific protein/claudin-11, CNPase, proteolipid protein 1 (PLP1), or a combination thereof.
  • the PDGFR ⁇ inhibitor is a compound of Formula I: , [0183] or a pharmaceutically acceptable salt or solvate thereof, wherein: indicates a single bond or a double bond such that all valences are satisfied; X 1 , X 2 , X 3 , and X 4 are selected from N and CR a , with the proviso that not more than two of X 1 , X 2 , X 3 , and X 4 are N; one of Y 1 and Y 2 is N and the other of Y 1 and Y 2 is C; each R a is independently selected from H, halo, C 1 -C 4 alkyl, and C 1 -C 4 alkoxy; R 1 is selected from C 1 -C 4 alkyl, C 3 -C 8 cycloalkyl, 3-8 membered heterocyclyl, heteroaryl, aryl, and C 1 -C 8 alkoxy, all of which can be optionally substituted with one
  • Y 1 is N and Y 2 is C. [0185] In some aspects, Y 1 is C and Y 2 is N. [0186] In some aspects, X 1 is N, X 2 is CR a , X 3 is CR a , and X 4 is CR a . [0187] In some aspects, X 1 is CR a , X 2 is N, X 3 is CR a , and X 4 is CR a . [0188] In some aspects, X 1 is CR a , X 2 is CR a , X 3 is N, and X 4 is CR a .
  • X 1 is CR a
  • X 2 is CR a
  • X 3 is CR a
  • X 4 is N.
  • X 1 is CR a
  • X 2 is CR a
  • X 3 is CR a
  • X 4 is CR a
  • R 1 is 5- or 6-membered heteroaryl.
  • R 1 is an optionally substituted pyrazolyl.
  • R 1 is selected from: , , an d ; and R 10 is selected from H, C 1 -C 4 alkyl, C 1 -C 4 alkoxy, aminoC 1 -C 4 alkyl, hydroxyC 1 - C 4 alkyl, and C 1 -C 4 alkylsulfonyl, wherein the C 1 -C 4 alkyl, C 1 -C 4 alkoxy, aminoC 1 -C 4 alkyl, hydroxyC 1 -C 4 alkyl, and C 1 -C 4 alkylsulfonyl can be optionally substituted by one or more substituents selected from hydroxyl, C 1 -C 4 alkoxy, NR 10a R 10b , halo, and deuterium, wherein R 10a and R 10b are selected from hydrogen and C 1 -C 4 alkyl, or wherein R 10a and R 10b taken together with the nitrogen atom to which they are attached form a 4- to 8-membere
  • R 1 is .
  • R 10 is CH 3 .
  • R 2 is selected from cycloalkyl, cycloalkenyl, alkyl, oxoalkylamino, aminoalkylamino, amino, heterocyclyl, heteroaryl, aminoheterocyclyl, heterocyclylamino, and aminoalkylamino, all of which can be optionally substituted with six or more substituents selected from D, halo, hydroxy, oxo, and C 1 -C 4 alkyl.
  • R 2 is heterocyclyl optionally substituted with C 1 -C 4 alkyl or oxo.
  • R 2 is substituted with one R 3 .
  • R 2 is selected from: , , , , , , ,
  • nd indicates a single bond or a double bond such that all valences are satisfied; m is selected from 0, 1, 2, 3, 4, 5, and 6; and Z 1 , Z 2 , and Z 3 are selected from N and CR a .
  • R 2 is selected from:
  • R 2 is selected from the group consisting of:
  • R 2 is: .
  • R 3 is selected from aryl, heteroaryl, -C(O)R 31 , -C(O)OR 31 , - C(O)NR 31 R 32 , -S(O)2NR 31 R 32 , cycloalkyl, and alkyl, all of which can be optionally substituted with one, two, three, four, or five R 30 .
  • R 3 is selected from heteroaryl optionally substituted with one, two three, four, or five R 30 and -C(O)OR 31 .
  • the PDGFR ⁇ inhibitor is a compound of Formula Ia: , [0206] or a pharmaceutically acceptable salt or solvate thereof, wherein a and b are each independently selected from 1, 2, and 3; and Q is selected from -CH- and -N-, with the proviso that if Q is –N-, a and b are not 1.
  • the PDGFR ⁇ inhibitor is a compound of Formula II: , or a pharmaceutically acceptable salt or solvate thereof.
  • the PDGFR ⁇ inhibitor is a compound of Formula IIa:
  • R 3 is selected from: , , , , , , ,
  • R 3 is selected from:
  • R 3 is selected from: . [0212] In some aspects, R 3 is heteroaryl. [0213] In some aspects, R 3 is: . [0214] In some aspects, R 30 is: . [0215] In some aspects, R 300 is selected from: , , , , [0216] In some aspects, R 300 is: . [0217] In some aspects, R 301 is selected from H and CH 3 . [0218] In some aspects, R 301 is H. [0219] In some aspects, R 302 is H. [0220] In some aspects, R 301 and R 302 are H. [0221] In some aspects, R 30 is benzyl.
  • R 3 is selected from: d .
  • R 3 is selected from: , , .
  • R 32 is H.
  • the PDGFR ⁇ inhibitor is a compound of Formula IV:
  • the PDGFR ⁇ inhibitor is a compound of Formula V: , or a pharmaceutically acceptable salt or solvate thereof.
  • R 31 is selected from:
  • R 31a is selected from H, D, alkylamino, optionally substituted C 1 -C 4 alkyl, C 1 - C 4 alkoxy, amino, and C 1 -C 4 haloalkyl; each R 31b is independently selected from H, D, halo, hydroxy, amino, cyano, alkylamino, optionally substituted C 1 -C 4 alkyl, C 1 -C 4 haloalkyl, optionally substituted C 1 -C 4 alkoxy, and C 3 - C 6 cycloalkyl, and q is 1, 2, or 3.
  • Table 1 (below) provides the structure of exemplary compounds that are useful for the present disclosure (e.g., can target and inhibit kinase activity associated with PDGFR ⁇ expressed on OPCs). Table 1. Exemplary Compounds With PDGFR ⁇ Inhibitory Activity [0229] In some aspects, the exemplary compounds of the present invention do not include compounds 156, 250 and 275. In some aspects, the exemplary compounds of the present invention do not include the compounds described in Table 1 on pp. 117-170 of International Appl. No. PCT/US2022/079480, which is herein incorporated by reference in its entirety. [0230] In some aspects, the PDGFRa inhibitor is a compound described in Table 1 on pp. 117-170 of International Appl. No.
  • compositions comprising a PDGFR ⁇ inhibitor of the present disclosure having the desired degree of purity, and a pharmaceutically acceptable carrier or excipient, in a form suitable for administration to a subject.
  • the PDGFR ⁇ inhibitor has a purity of about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or greater than about 99%.
  • compositions can be determined in part by the particular composition being administered, as well as by the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of pharmaceutical compositions (See, e.g., Remington, 23 rd Edition, The Science and Practice of Pharmacy, editor: A. Adejare, 2020, Adademic Press.). The pharmaceutical compositions are generally formulated sterile and in full compliance with all Good Manufacturing Practice (GMP) regulations of the U.S. Food and Drug Administration. [0233]
  • a pharmaceutical composition comprises a PDGFR ⁇ inhibitor described herein, and a pharmaceutically acceptable carrier.
  • Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine,
  • a pharmaceutical composition disclosed herein comprises one or more additional components selected from: a bulking agent, stabilizing agent, surfactant, buffering agent, or combinations thereof.
  • Buffering agents useful for the current disclosure can be a weak acid or base used to maintain the acidity (pH) of a solution near a chosen value after the addition of another acid or base. Suitable buffering agents can maximize the stability of the pharmaceutical compositions by maintaining pH control of the composition. Suitable buffering agents can also ensure physiological compatibility or optimize solubility. Rheology, viscosity and other properties can also be dependent on the pH of the composition.
  • Common buffering agents include, but are not limited to, a Tris buffer, a Tris-Cl buffer, a histidine buffer, a TAE buffer, a HEPES buffer, a TBE buffer, a sodium phosphate buffer, a MES buffer, an ammonium sulfate buffer, a potassium phosphate buffer, a potassium thiocyanate buffer, a succinate buffer, a tartrate buffer, a DIPSO buffer, a HEPPSO buffer, a POPSO buffer, a PIPES buffer, a PBS buffer, a MOPS buffer, an acetate buffer, a phosphate buffer, a cacodylate buffer, a glycine buffer, a sulfate buffer, an imidazole buffer, a guanidine hydrochloride buffer, a phosphate-citrate buffer, a borate buffer, a malonate buffer, a 3- picoline buffer, a 2-picoline buffer, a 4-picoline buffer,
  • a pharmaceutical composition disclosed herein further comprises a bulking agent.
  • Bulking agents can be added to a pharmaceutical product in order to add volume and mass to the product, thereby facilitating precise metering and handling thereof.
  • Bulking agents that can be used with the present disclosure include, but are not limited to, sodium chloride (NaCl), mannitol, glycine, alanine, or combinations thereof.
  • a pharmaceutical composition disclosed herein can also comprise a stabilizing agent.
  • stabilizing agents that can be used with the present disclosure include: sucrose, trehalose, raffinose, arginine, or combinations thereof.
  • a pharmaceutical composition disclosed herein comprises a surfactant.
  • the surfactant can be selected from the following: alkyl ethoxylate, nonylphenol ethoxylate, amine ethoxylate, polyethylene oxide, polypropylene oxide, fatty alcohols such as cetyl alcohol or oleyl alcohol, cocamide MEA, cocamide DEA, polysorbates, dodecyl dimethylamine oxide, or combinations thereof.
  • the surfactant is polysorbate 20 or polysorbate 80.
  • a pharmaceutical composition disclosed herein e.g., comprising a PDGFR ⁇ inhibitor
  • further comprises an amino acid e.g., comprising a PDGFR ⁇ inhibitor
  • the amino acid is selected from arginine, glutamate, glycine, histidine, or combinations thereof.
  • the composition further comprises a sugar alcohol.
  • sugar alcohol includes: sorbitol, xylitol, maltitol, mannitol, or combinations thereof.
  • routes of administration include intramuscularly, cutaneously, subcutaneously, ophthalmic, intravenously, intraperitoneally, intradermally, intraorbitally, intracerebrally, intracranially, intraspinally, intraventricularly, intrathecally, intracapsularly, orally, pulmonarily, intranasally, intra-arterially, intralymphatically, periocularly, topically, transdermally, rectally, vaginally, or intratumorally, or via intratympanic injection.
  • Parenteral administration characterized by, e.g., cutaneous, subcutaneous, intramuscular, or intravenous injection, is also contemplated herein.
  • injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions.
  • the injectables, solutions and emulsions also contain one or more excipients. Suitable excipients are, for example, water, saline, dextrose, glycerol, or ethanol.
  • compositions to be administered can also contain minor amounts of non-toxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents, stabilizers, solubility enhancers, and other such agents, such as for example, sodium acetate, sorbitan monolaurate, triethanolamine oleate, and cyclodextrins.
  • auxiliary substances such as wetting or emulsifying agents, pH buffering agents, stabilizers, solubility enhancers, and other such agents, such as for example, sodium acetate, sorbitan monolaurate, triethanolamine oleate, and cyclodextrins.
  • Pharmaceutically acceptable carriers used in parenteral preparations include aqueous vehicles, nonaqueous vehicles, antimicrobial agents, isotonic agents, buffers, antioxidants, local anesthetics, suspending and dispersing agents, emulsifying agents, sequestering or chelating agents, and other pharmaceutically acceptable substances.
  • aqueous vehicles examples include sodium chloride injection, Ringer’s injection, isotonic dextrose injection, sterile water injection, dextrose, and lactated Ringer’s injection.
  • Nonaqueous parenteral vehicles include fixed oils of vegetable origin, cottonseed oil, corn oil, sesame oil and peanut oil.
  • Antimicrobial agents in bacteriostatic or fungistatic concentrations can be added to parenteral preparations packaged in multiple-dose containers which include, for example, phenols or cresols, mercurials, benzyl alcohol, chlorobutanol, methyl and propyl p-hydroxybenzoic acid esters, thimerosal, benzalkonium chloride, and benzethonium chloride.
  • Isotonic agents include sodium chloride, and dextrose. Buffers include phosphate and citrate. Antioxidants include sodium bisulfate. Local anesthetics include procaine hydrochloride. Suspending and dispersing agents include sodium carboxymethylcelluose, hydroxypropyl methylcellulose, and polyvinylpyrrolidone. Emulsifying agents include Polysorbate 80 (TWEEN ® 80). Sequestering or chelating agent of metal ions includes EDTA. Pharmaceutical carriers also include ethyl alcohol, polyethylene glycol, and propylene glycol for water miscible vehicles; and sodium hydroxide, hydrochloric acid, citric acid, or lactic acid for pH adjustment.
  • Preparations for parenteral administration include sterile solutions ready for injection, sterile dry soluble products, such as lyophilized powders, ready to be combined with a solvent just prior to use, including hypodermic tablets, sterile suspensions ready for injection, sterile dry insoluble products ready to be combined with a vehicle just prior to use, and sterile emulsions.
  • the solutions can be either aqueous or nonaqueous.
  • suitable carriers include physiological saline or phosphate buffered saline (PBS), and solutions containing thickening and solubilizing agents, such as glucose, polyethylene glycol, and polypropylene glycol, and mixtures thereof.
  • Topical mixtures comprising a compound described herein are prepared as described for the local and systemic administration.
  • the resulting mixture can be a solution, suspension, emulsions, or the like and can be formulated as creams, gels, ointments, emulsions, solutions, elixirs, lotions, suspensions, tinctures, pastes, foams, aerosols, irrigations, sprays, suppositories, bandages, dermal patches, or any other formulations suitable for topical administration.
  • a therapeutic agent described herein e.g., PDGFR ⁇ inhibitor
  • formulations for administration to the respiratory tract can be in the form of an aerosol or solution for a nebulizer, or as a microfine powder for insufflations, alone or in combination with an inert carrier such as lactose.
  • the particles of the formulation can have diameters of less than about 50 microns, e.g., less than about 10 microns.
  • the particle diameters can have diameters of less than about 50 microns, e.g., less than about 10 microns, as measured by dynamic light scattering (DLS) or static image analysis using microscopy.
  • a therapeutic agent disclosed herein e.g., PDGFR ⁇ inhibitor
  • PDGFR ⁇ inhibitor can be formulated for local or topical application, such as for topical application to the skin and mucous membranes, such as in the eye, in the form of gels, creams, and lotions and for application to the eye or for intracisternal or intraspinal application. Topical administration is contemplated for transdermal delivery and also for administration to the eyes or mucosa, or for inhalation therapies.
  • Transdermal patches including iontophoretic and electrophoretic devices, are well known to those of skill in the art, and can be used to administer a therapeutic agent (e.g., those disclosed herein).
  • a therapeutic agent e.g., those disclosed herein.
  • such patches are disclosed in U.S. Patent Nos. 6,267,983; 6,261,595; 6,256,533; 6,167,301; 6,024,975; 6,010,715; 5,985,317; 5,983,134; 5,948,433; and 5,860,957.
  • a pharmaceutical composition comprising a therapeutic agent described herein (e.g., PDGFR ⁇ inhibitor) is a lyophilized powder, which can be reconstituted for administration as solutions, emulsions, and other mixtures. It can also be reconstituted and formulated as a solid or a gel.
  • the lyophilized powder is prepared by dissolving an compound as described herein, or a pharmaceutically acceptable derivative thereof, in a suitable solvent.
  • the lyophilized powder is sterile.
  • the solvent can contain an excipient, which improves the stability or other pharmacological component of the powder or reconstituted solution that is prepared from the powder.
  • Excipients that can be used include, but are not limited to, dextrose, sorbitol, fructose, corn syrup, xylitol, glycerin, glucose, sucrose, or other suitable agent.
  • the solvent can also contain a buffer, such as citrate, sodium or potassium phosphate or other such buffer known to those of skill in the art at, in some aspects, about neutral pH.
  • a buffer such as citrate, sodium or potassium phosphate or other such buffer known to those of skill in the art at, in some aspects, about neutral pH.
  • Subsequent sterile filtration of the solution followed by lyophilization under standard conditions known to those of skill in the art provides the desired formulation.
  • the resulting solution can be apportioned into vials for lyophilization. Each vial can contain a single dosage or multiple dosages of the compound.
  • the lyophilized powder can be stored under appropriate conditions, such as at about 4°C to room temperature.
  • Reconstitution of a lyophilized powder with water for injection provides a formulation for use in parenteral administration.
  • the lyophilized powder is added to sterile water or other suitable carrier.
  • the precise amount depends upon the selected compound. Such an amount can be empirically determined.
  • Pharmaceutical compositions provided herein e.g., comprising a PDGFR ⁇ inhibitor
  • compositions to be used for in vivo administration can be sterile. This can be accomplished, for example, by filtration through, e.g., a sterile filtration membrane. IV.
  • kits comprising one or more PDGFR ⁇ inhibitors described herein.
  • a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions described herein, such as one or more PDGFR ⁇ inhibitors provided herein, and optionally, an instruction for use.
  • the kits contain a pharmaceutical composition described herein (e.g., a PDGFR ⁇ inhibitor) and any prophylactic or therapeutic agent, such as those described herein. V.
  • PDGFR ⁇ inhibitors of the present disclosure are useful in reducing or inhibiting PDGFR ⁇ activity, which, in some aspects, can promote the myelination of an axon and/or remyelination of a demyelinated neuronal axon.
  • the present disclosure relates to methods of inhibiting or reducing PDGFR ⁇ activity in a cell or tissue, e.g., of a subject in need thereof, comprising contacting the cell or tissue with a PDGFR ⁇ inhibitor described herein.
  • PDGFR ⁇ kinase activity in the cell or tissue is inhibited by at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or 100% compared to a reference (e.g., PDGFR ⁇ activity in the cell prior to the contacting and/or PDGFR ⁇ activity in a corresponding cell that was not contacted with the PDGFR ⁇ inhibitor).
  • the PDGFR ⁇ activity status of a cell can be determined using any suitable method known in the art.
  • PDGFR ⁇ activity can be determined by measuring the corresponding gene expression level, e.g., by qRT- PCR.
  • PDGFR ⁇ activity can be determined by measuring the PDGFR ⁇ protein level, e.g., by immunoblotting. In some aspects, the PDGFR ⁇ activity can be determined by measuring receptor phosphorylation, e.g., by western blotting.
  • An exemplary method of assaying such an activity a purified protein enzyme assay, is provided in Example 131 (also referred to herein as the "Promega Assay").
  • the enzymatic PDGFR ⁇ kinase assay comprises 20 ng of purified PDGFR ⁇ protein, 150 ⁇ M of ATP and 1 ⁇ g of substrate, Poly (Glu4Tyr1) in a volume of 15 ⁇ l. The potency of a compound can be determined using such assays.
  • compounds most useful in the methods of the present disclosure have an IC 50 below 10,000 nM (e.g., less than 9,000 nM, less than 8,000 nM, less than 7,000 nM, less than 6,000 nM, less than 5,000 nM, less than 4,000 nM, less than 3,000 nM, less than 2,000 nM, less than 1,000 nM, less than 900 nM, less than 800 nM, less than 700 nM, less than 600 nM, less than 500 nM, less than 400 nM, less than 300 nM, less than 200 nM, less than 100 nM, less than 50 nM, less than 25 nM, less than 10 nM, less than 5 nM, less than 2.5 nM, less than 1 nM, less than 0.5 nM, or less than 0.2 nM, for instance, less than 500 nM) in such an assay.
  • 10,000 nM e.g., less than 9,000 n
  • the cell is an oligodendrocyte progenitor cell (OPC).
  • OPC oligodendrocyte progenitor cell
  • inhibiting or reducing PDGFR ⁇ activity can promote the myelination of a neuron (e.g., an axon of a neuron,), e.g., by promoting the differentiation of an OPC into an oligodendrocyte that, in turn, myelinates the axon.
  • inhibiting or reducing PDGFR ⁇ activity can also promote the remyelination of a demyelinated neuronal axon.
  • remyelination can repair damage to demyelinated (e.g., hypomyelinated) axons, and thereby, aid in recovering axonal signalling or reducing axonal injury or loss.
  • demyelinated e.g., hypomyelinated
  • a method of promoting the myelination of an axon comprising contacting an OPC with an effective amount of a PDGFR ⁇ inhibitor described herein, wherein the contacting results in the OPC to differentiate into an oligodendrocyte, and wherein the oligodendrocyte is capable of (and in some embodiments used for) promoting the myelination of the axon.
  • the number of neurons with axons that are myelinated is increased by at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, at least 10-fold, at least 15-fold, at least 20- fold, at least 25-fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, or at least 50-fold compared to a reference (e.g., the number of neurons with myelinated axons in the subject prior to the administration and/or the number of neurons with myelinated axons in a corresponding subject that did not receive the administration of the PDGFR ⁇ inhibitor).
  • a reference e.g., the number of neurons with myelinated axons in the subject prior to the administration and/or the number of neurons with myelinated axons in a corresponding subject that did not receive the administration of the PDGFR ⁇ inhibitor.
  • the increase in the number of neurons with axons that are myelinated can be determined using any suitable methods known in the art or described herein.
  • the increase in the number of neurons with axons that are myelinated can be determined by visualizing and/or quantifying the expression of a marker associated with myelinated neurons.
  • the marker associated with myelinated neurons comprises myelin basic protein (MBP), myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, proteolipid protein 1 (PLP1), or a combination thereof.
  • a method of promoting remyelination of an axon of a neuron comprising contacting an OPC with an effective amount of any of the PDGFR ⁇ inhibitors described herein, wherein the contacting results in the OPC to differentiate into an oligodendrocyte, and wherein the oligodendrocyte is capable of (and in some embodiments used for) promoting the remyelination of the demyelinated neuronal axon.
  • the number of demyelinated axons that are myelinated is increased by at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, at least 10-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, or at least 50-fold compared to a reference (e.g., the number of demyelinated neuronal axons in the subject prior to the administration and/or the amount of demyelinated neuronal axons in a corresponding subject that did not receive the administration of the PDGFR ⁇ inhibitor).
  • a reference e.g., the number of demyelinated neuronal axons in the subject prior to the administration and/or the amount of demyelinated neuronal axons in a corresponding subject that did not receive
  • the increase in the number of demyelinated axons that are myelinated can be determined using any suitable methods known in the art or described herein.
  • the increase in the number of demyelinated axons that are myelinated can be determined by visualizing and/or quantifying the expression of a marker associated with myelinated neurons.
  • the marker associated with myelinated neurons comprises myelin basic protein (MBP), myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, proteolipid protein 1 (PLP1), or a combination thereof.
  • a method of increasing remyelination in a subject comprising contacting an OPC with an effective amount of any of the PDGFR ⁇ inhibitors described herein, wherein the contacting results in the OPC to differentiate into an oligodendrocyte, and wherein the oligodendrocyte is capable of (and in some embodiments used for) increasing remyelination in the subject.
  • the remyelination is increased by at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, at least 10-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, or at least 50-fold compared to a reference (e.g., the number of demyelinated neuronal axons in the subject prior to the administration and/or the amount of demyelinated neuronal axons in a corresponding subject that did not receive the administration of the PDGFR ⁇ inhibitor).
  • a reference e.g., the number of demyelinated neuronal axons in the subject prior to the administration and/or the amount of demyelinated neuronal axons in a corresponding subject that did not receive the administration of the PDGFR ⁇ inhibitor.
  • the increase in the number of demyelinated axons that are myelinated can be determined using any suitable methods known in the art or described herein.
  • the increase in the number of demyelinated axons that are myelinated can be determined by visualizing and/or quantifying the expression of a marker associated with myelinated neurons.
  • the marker associated with myelinated neurons comprises myelin basic protein (MBP), myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, proteolipid protein 1 (PLP1), or a combination thereof.
  • a method of reducing the demyelination of a myelinated axon comprising contacting an OPC with an effective amount of any of the PDGFR ⁇ inhibitors described herein, wherein the contacting results in the OPC to differentiate into an oligodendrocyte, and wherein the oligodendrocyte is capable of (and in some embodiments used for) reducing the demyelination of the myelinated axon.
  • demyelination of a myelinated axon in the subject is reduced by at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, at least 10-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, or at least 50-fold compared to a reference (e.g., demyelination of a myelinated axon in a corresponding subject that did not receive an administration of the PDGFR ⁇ inhibitor).
  • a reference e.g., demyelination of a myelinated axon in a corresponding subject that did not receive an administration of the PDGFR ⁇ inhibitor.
  • the decrease in the number of neurons with axons that are demyelinated can be determined using any suitable methods known in the art or described herein.
  • the decrease in the number of neurons with axons that are demyelinated can be determined by visualizing and/or quantifying the expression of a marker associated with myelinated neurons.
  • the marker associated with myelinated neurons comprises myelin basic protein (MBP), myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, proteolipid protein 1 (PLP1), or a combination thereof.
  • a method of decreasing the rate of demyelination comprising contacting an OPC with an effective amount of any of the PDGFR ⁇ inhibitors described herein, wherein the contacting results in the OPC to differentiate into an oligodendrocyte, and wherein the oligodendrocyte is capable of (and in some embodiments used for) reducing the rate of demyelination in the subject.
  • the rate of demyelination in the subject is decreasing by at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, at least 10-fold, at least 15-fold, at least 20-fold, at least 25- fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, or at least 50-fold compared to a reference (e.g., the rate of demyelination in a corresponding subject that did not receive an administration of the PDGFR ⁇ inhibitor).
  • a reference e.g., the rate of demyelination in a corresponding subject that did not receive an administration of the PDGFR ⁇ inhibitor.
  • the decrease in the number of neurons with axons that are demyelinated can be determined using any suitable methods known in the art or described herein.
  • the decrease in the number of neurons with axons that are demyelinated can be determined by visualizing and/or quantifying the expression of a marker associated with myelinated neurons.
  • the marker associated with myelinated neurons comprises myelin basic protein (MBP), myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, proteolipid protein 1 (PLP1), or a combination thereof.
  • the contacting can occur ex vivo (e.g., OPCs of a subject can be isolated and contacted with a PDGFR ⁇ inhibitor in vitro to assess the therapeutic efficacy of the inhibitor).
  • the contacting occurs in vivo (e.g., in a subject in need thereof).
  • the methods can further comprise administering to the subject an effective amount of any of the PDGFR ⁇ inhibitors described herein.
  • a PDGFR ⁇ inhibitor described herein is capable of (and in some embodiments used for) migrating into the CNS of the subject (e.g., by crossing the blood-brain barrier) and interacting with the nervous system cells present within the CNS.
  • PDGFR ⁇ inhibitors of the present disclosure are capable of (and in some embodiments used for) targeting PDGFR ⁇ expressed on an OPC, and thereby inhibit or reduce the PDGFR ⁇ activity of the OPC. In some aspects, this results in the activation of the OPC and the subsequent differentiation of the OPC into an oligodendrocyte.
  • a method of activating an OPC within the CNS of a subject in need thereof comprising administering to the subject any of the PDGFR ⁇ inhibitors described herein.
  • the myelin sheath which surrounds the neuronal axons can affect various functions of neurons.
  • PDGFR ⁇ inhibitors described herein can increase the rate of transmission of electrical impulses (i.e., nerve signals or action potentials) along the axon of a neuron.
  • the present disclosure provides a method of increasing electrical impulse transmission along the axon of a neuron, comprising contacting an OPC with an effective amount of any of the PDGFR ⁇ inhibitors described herein, wherein the contacting results in the OPC to differentiate into an oligodendrocyte, and wherein the oligodendrocyte is capable of (and in some embodiments used for) myelinating the axon of the neuron, and thereby increase the electrical impulse transmission.
  • the electrical impulse transmission along the axon of the neuron is increased by at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, at least 10-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, or at least 50-fold compared to a reference (e.g., electrical impulse transmission across the axon prior to or in the absence of contact with the PDGFR ⁇ inhibitors).
  • a reference e.g., electrical impulse transmission across the axon prior to or in the absence of contact with the PDGFR ⁇ inhibitors.
  • Action potential conduction velocities can be measured as compound action potentials across the corpus callosum in vitro or by auditory or visual evoked potential recording in vivo.
  • remyelination may overcome conduction block, allowing axonal signals that would be prematurely terminated to be transmitted. Remyelination can enhance axonal health and prevent axonal degradation in a diseased state.
  • Non-limiting examples of methods useful for measuring such properties of a neuronal axon are described in, e.g., Li et al., PLoS One 11(11): e0165637 (Nov.2016); Maheras et al., Sci Rep 8(1): 3798 (Feb.
  • the PDGFR ⁇ inhibitors described herein can be useful in the treatment of a demyelinating disease, e.g., by inducing the differentiation of an OPC into an oligodendrocyte, thereby promoting the remyelination of neuronal axons. Accordingly, in some aspects, the present disclosure is directed to a method of treating a demyelinating disease in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a PDGFR ⁇ inhibitor described herein.
  • references to a method of treating a demyelinating disease as described herein also refer to the agents and compositions as described herein for use in treating a demyelinating disease.
  • the PDGFR ⁇ inhibitors provided herein can be used to treat a wide range of demyelinating diseases (including those that are associated with demyelination and/or hypomyelination).
  • the demyelinating disease that can be treated with the present disclosure comprise those that are characterized by demyelination of one or more cells within the CNS.
  • demyelinating disease that can be treated with the present disclosure include one or more of: acute disseminated encephalomyelitis (ADEM), acute hemorrhagic leukoencephalitis, acute optic neuritis, acute transverse myelitis, adrenoleukodystrophy, adrenomyeloneuropathy, Alexander disease, Alzheimer's disease, aminoacidurias, amyotrophic lateral sclerosis, anti-MAG peripheral neuropathy, anti-MOG associated spectrum, Balo concentric sclerosis, brain injury, CAMFAK Syndrome, Canavan disease, carbon monoxide toxicity, central pontine myelinolysis, cerebral hypoxia, cerebral ischemia, Charcot–Marie–Tooth disease, chronic inflammatory demyelinating polyneuropathy, chronic relapsing inflammatory optic neuritis (CRION), chronic traumatic encephalopathy, clinically isolated syndrome (CIS), congenital cataract, copper deficiency associated condition, delayed post-hypoxic leuk
  • ADAM
  • a disease that can be treated with the present disclosure comprises a tumor associated with abnormal (e.g., increased) PDGF or PDGFR ⁇ activity.
  • PDGF-associated tumor Such a tumor is referred to herein as "PDGF- associated tumor.”
  • a method of treating a PDGF-associated tumor in a subject in need thereof comprising administering to the subject any of the PDGFR ⁇ inhibitor described herein.
  • a non-limiting example of a PDGF- associated tumor comprises oligodendroglioma.
  • the demyelinating disease that can be treated with a PDGFR ⁇ inhibitor described herein comprises multiple sclerosis.
  • MS multiple sclerosis
  • PPMS primary progressive multiple sclerosis
  • RRMS relapsing-remitting multiple sclerosis
  • SPMS secondary progressive multiple sclerosis
  • the PDGFR ⁇ inhibitors of the present disclosure can be used to treat all types of MS.
  • a PDGFR ⁇ inhibitor of the present disclosure can be used to treat clinically isolated syndrome (CIS).
  • a PDGFR ⁇ inhibitor of the present disclosure can be used to treat radiologically isolated syndrome (RIS).
  • a method of treating a relapsing form of multiple sclerosis in a subject in need thereof comprising administering to the subject a therapeutically effective amount of any of the PDGFR ⁇ inhibitors provided herein.
  • the demyelinating disease that can be treated with the present disclosure is an optic neuritis.
  • a PDGFR ⁇ inhibitor is used to ameliorate optic neuritis, e.g., optic neuritis that is due to multiple sclerosis.
  • PDGFR ⁇ inhibitors of the present disclosure can help restore nervous system cell function and thereby, reduce and/or alleviate one or more symptoms associated with a demyelinating disease. Accordingly, in some aspects, treating a demyelinating disease comprises reducing one or more symptoms associated with the demyelinating disease.
  • Non-limiting examples of such symptoms include one or more of: fatigue, dizziness, malaise, elevated fever and high body temperature, extreme sensitivity to cold in the hands and feet, weakness and stiffness in muscles and joints, weight changes, digestive or gastrointestinal problems, low blood pressure, high blood pressure, irritability, anxiety, depression, blurred vision, double vision, ataxia, clonus, spasms, dysarthria, weakness, clumsiness, hand paralysis, hemiparesis, genital anesthesia, sexual dysfunction, incoordination, paresthesias, ocular paralysis, impaired muscle coordination, loss of sensation, tingling, numbness, pain, impaired vision, neurological symptoms, unsteady gait, balance problems, dizziness, spastic paraparesis, incontinence, hearing problems, speech problems, loss of olfaction, and agusia.
  • PDGFR ⁇ inhibitors described herein can also be used to prevent or delay the onset of one or more symptoms associated with a demyelinating disease.
  • demyelinating diseases particularly during the early stages, a subject may not exhibit any apparent symptoms of the disease.
  • the damage to the myelin sheaths may not be as severe, such that the oligodendrocytes present within the CNS of the subject can adequately repair any damages.
  • the damage to the myelin sheaths can become more severe, resulting in the manifestation of one or more symptoms of the disease.
  • the manifestation of the one or more symptoms of the disease can be prevented or delayed.
  • the manifestation of the one or more symptoms of the disease is delayed by at least 1.1-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, at least 10-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, or at least 50-fold.
  • the subject treated with a PDGFR ⁇ inhibitor described herein exhibits less number of symptoms and/or the symptoms are reduced in severity.
  • Whether a subject exhibits one or more symptoms associated with a demyelinating disease can be assessed using any suitable approaches known in the art.
  • whether a subject exhibits the one or more symptoms associated with a demyelinating disease can be determined by assessing the subject's performance in one or more of the following tests: a visual evoked potential (VEP) test, a multifocal visual evoked potential (mfVEP) test, a low contrast visual acuity (LCVA) test, a magnetic resonance imaging (MRI) (e.g., magnetization transfer ratio (MTR), myelin water fraction (MWF), quantitative susceptibility mapping (QSM), and T2 imaging), an electromyography (EMG), a nerve conduction velocity (NCV) test, an Extended Disability Status Scale (EDSS), a timed walk test (e.g., timed 25-foot walk), a Nine-Hole Peg Test (9HPT), an ocular coherence tomograph (OCT), a quality of life measure test (e.g., Multiple Sclerosis Quality of Life-54 and Vision-Related Quality of Life), cognitive assessment (e.g., Symbol
  • whether a subject exhibits one or more symptoms associated with a demyelinating disease is determined by assessing the subject's performance in a LC-VA test. In some aspects, whether a subject exhibits one or more symptoms associated with a demyelinating disease is determined by assessing the subject's performance in a cognitive assessment. [0274] In some aspects, an improved performance as compared to a reference performance (e.g., a corresponding subject's performance in the test where the reference subject was not treated with a PDGFR ⁇ inhibitor described herein and/or the subject's performance prior to the treatment with a PDGFR ⁇ inhibitor) indicates that the one or more symptoms associated with a demyelinating disease is reduced or alleviated in the subject.
  • a reference performance e.g., a corresponding subject's performance in the test where the reference subject was not treated with a PDGFR ⁇ inhibitor described herein and/or the subject's performance prior to the treatment with a PDGFR ⁇ inhibitor
  • a comparable or reduced performance as compared to a reference performance indicates that the one or more symptoms associated with a demyelinating disease is maintained or has worsened in the subject.
  • some aspects of the present disclosure are directed to methods of treating a demyelinating disease in a subject in need thereof, comprising assessing the subject's performance in a test for assessing one or more symptoms associated with a demyelinating disease, wherein the subject received a treatment comprising a PDGFR ⁇ inhibitor described herein prior to the assessment.
  • the treatment comprising a PDGFR ⁇ inhibitor can be maintained in the subject (e.g., the subject receives one or more additional administration of the PDGFR ⁇ inhibitor at the same dose and/or dosing interval).
  • the treatment comprising a PDGFR ⁇ inhibitor is reduced (e.g., the subject receives one or more additional administration of the PDGFR ⁇ inhibitor but at a lower dose and/or longer dosing interval) or stopped.
  • the subject's performance is comparable or reduced as compared to a reference performance, (e.g., a corresponding subject's performance in the test where the subject was not treated with a PDGFR ⁇ inhibitor described herein and/or the subject's performance prior to the treatment with a PDGFR ⁇ inhibitor)
  • the treatment comprising a PDGFR ⁇ inhibitor is adjusted such that the subject receives one or more administrations of the PDGFR ⁇ inhibitor at a higher dose and/or shorter dosing interval.
  • Some aspects of the present disclosure are directed to methods of improving a subject's performance in a test for assessing one or more symptoms associated with a demyelinating disease, comprising administering to the subject any of the PDGFR ⁇ inhibitors described herein.
  • the subject's performance in the test is increased as compared to a reference performance (e.g., a corresponding subject's performance in the test where the reference subject was not treated with a PDGFR ⁇ inhibitor described herein and/or the subject's performance prior to the treatment with a PDGFR ⁇ inhibitor).
  • the test is one or more of a visual evoked potential (VEP) test, a multifocal visual evoked potential (mfVEP) test, a low contrast visual acuity (LC-VA) test, a magnetic resonance imaging (MRI) (e.g., magnetization transfer ratio (MTR), myelin water fraction (MWF), quantitative susceptibility mapping (QSM), and T2 imaging), an electromyography (EMG), a nerve conduction velocity (NCV) test, an Extended Disability Status Scale (EDSS), a timed walk test (e.g., timed 25-foot walk), a Nine-Hole Peg Test (9HPT), an ocular coherence tomograph (OCT), a quality of life measure test (e.g., Multiple Sclerosis Quality of Life- 54 and Vision-Related Quality of Life), cognitive assessment (e.g., Symbol-Digit Modalities Test or Montreal Cognitive Assessment), or combinations thereof.
  • MRI magnetic resonance imaging
  • MMR magnetization transfer ratio
  • MMF
  • the test is a LCVA test. In some aspects, the test is a cognitive assessment.
  • the introduction of a PDGFR ⁇ inhibitor into a subject can be done by any suitable route, including, but not limited to, intratumorally, orally, pulmonarily, intranasally, parenterally (intravenously, intra- arterially, intramuscularly, intraperitoneally, or subcutaneously), rectally, vaginally, intralymphatically, intrathecally, periocularly, cutaneously, intradermally, intraorbitally, intracerebrally, intracranially, intraspinally, intraventricuarly, intracapsularly, or topically.
  • Administration includes self-administration and the administration by another.
  • a suitable route of administration allows the PDGFR ⁇ inhibitors described herein to perform its intended function (e.g., inhibit PDGFR ⁇ activity and induce OPC differentiation).
  • a suitable route of administration includes that which would allow a PDGFR ⁇ inhibitor to reach the CNS or any other sites where OPCs are present. Non-limiting examples of such routes include intranasal delivery, intrathecal administration, intracranial administration, and combinations thereof.
  • a PDGFR ⁇ inhibitor described herein is administered orally.
  • a PDGFR ⁇ inhibitor of the present disclosure is administered intravenously.
  • a PDGFR ⁇ inhibitor can be administered both orally and intravenously.
  • the PDGFR ⁇ inhibitor can be administered to the subject using any suitable dosing schedule.
  • the PDGFR ⁇ inhibitor is administered to the subject once.
  • multiple doses of the PDGFR ⁇ inhibitor are administered to the subject.
  • a PDGFR ⁇ inhibitor described herein is administered to a subject according to an intermittent dosing schedule.
  • intermittent dosing schedule refers to a dosing schedule in which the therapeutic agent (e.g., PDGFR ⁇ inhibitor) is administered non-continuously (i.e., an intervening period exists between doses).
  • An intermittent dosing schedule useful for the present disclosure can encompass any discontinuous administration regimen that provides a therapeutically effective amount of a PDGFR ⁇ inhibitor to a subject in need thereof. Intermittent dosing regimens can use equivalent, lower, or higher doses of a PDGFR ⁇ inhibitor than would be used in continuous dosing regimens. Advantages of intermittent dose administration include, but are not limited to, improved safety, decreased toxicity (e.g., decreased weight loss), acceptable levels of ADME criteria, acceptable levels of undesirable effects on organ systems such as heart, pulmonary, hepatic, reproductive (for example, ovarian or testicular) or gastrointestinal, increased exposure, increased efficacy, and/or increased subject compliance.
  • the PDGFR ⁇ inhibitor is administered as a single agent and/or when administered in combination with one or more additional therapeutic agents, e.g., standard of care.
  • the dosing regimen for the PDGFR ⁇ inhibitor e.g., intermittent dosing
  • the additional therapeutic agents are independent of each other.
  • the PDGFR ⁇ inhibitor is administered using a first dosing regimen and the additional therapeutic agent is administered using a second dosing regimen, wherein the first and second dosing regimens are different.
  • the intermittent dosing schedule comprises administering the PDGFR ⁇ inhibitor to the subject every other day.
  • the PDGFR ⁇ inhibitor is administered to the subject one time per day. In some aspects, the PDGFR ⁇ inhibitor is administered to the subject two times per day. In some aspects, the PDGFR ⁇ inhibitor is administered to the subject three times per day. In some aspects, the PDGFR ⁇ inhibitor is administered to the subject four times per day. In some aspects, the PDGFR ⁇ inhibitor is administered to the subject every three days. In some aspects, the PDGFR ⁇ inhibitor is administered to the subject every four days. In some aspects, the PDGFR ⁇ inhibitor is administered to the subject every five days. In some aspects, the PDGFR ⁇ inhibitor is administered to the subject every six days. In some aspects, the PDGFR ⁇ inhibitor is administered to the subject once a week.
  • the PDGFR ⁇ inhibitor is administered to the subject once every eight days. In some aspects, the PDGFR ⁇ inhibitor is administered to the subject once every nine days. In some aspects, the PDGFR ⁇ inhibitor is administered to the subject every 10 days. In some aspects, the PDGFR ⁇ inhibitor is administered to the subject every 11 days. In some aspects, the PDGFR ⁇ inhibitor is administered to the subject every 12 days. In some aspects, the PDGFR ⁇ inhibitor is administered to the subject every 13 days. In some aspects, the PDGFR ⁇ inhibitor is administered to the subject once every two weeks. In some aspects, the PDGFR ⁇ inhibitor is administered to the subject once every three weeks. In some aspects, the PDGFR ⁇ inhibitor is administered to the subject once a month.
  • the PDGFR ⁇ inhibitor is administered to the subject once every five weeks. In some aspects, the PDGFR ⁇ inhibitor is administered to the subject once every six weeks, In some aspects, the PDGFR ⁇ inhibitor is administered to the subject once every seven weeks. In some aspects, the PDGFR ⁇ inhibitor is administered to the subject once every two months. In some aspects, the PDGFR ⁇ inhibitor is administered to the subject once every nine weeks. In some aspects, the PDGFR ⁇ inhibitor is administered to the subject once every 10 weeks. In some aspects, the PDGFR ⁇ inhibitor is administered to the subject once every 11 weeks. In some aspects, the PDGFR ⁇ inhibitor is administered to the subject once every three months. In some aspects, the PDGFR ⁇ inhibitor is administered to the subject once every four months.
  • the PDGFR ⁇ inhibitor is administered to the subject once every five months. In some aspects, the PDGFR ⁇ inhibitor is administered to the subject once every six months. In some aspects, the PDGFR ⁇ inhibitor is administered to the subject once every twelve months. [0280] In some aspects, the intermittent dosing schedule comprises administering to the subject a first dose and a second dose of a PDGFR ⁇ inhibitor, wherein the second dose is administered at least one day after administering the first dose. In some aspects, the second dose is administered at least two days after administering the first dose. In some aspects, the second dose is administered at least three days after administering the first dose. In some aspects, the second dose is administered at least four days after administering the first dose.
  • the second dose is administered at least five days after administering the first dose. In some aspects, the second dose is administered at least six days after administering the first dose. In some aspects, the second dose is administered at least seven days after administering the first dose. In some aspects, the second dose is administered at least eight days after administering the first dose. In some aspects, the second dose is administered at least nine days after administering the first dose. In some aspects, the second dose is administered at least 10 days after administering the first dose. In some aspects, the second dose is administered at least 11 days after administering the first dose. In some aspects, the second dose is administered at least 12 days after administering the first dose. In some aspects, the second dose is administered at least 13 days after administering the first dose.
  • the second dose is administered at least two weeks after administering the first dose. In some aspects, the second dose is administered at least three weeks after administering the first dose. In some aspects, the second dose is administered at least one month after administering the first dose. In some aspects, the second dose is administered at least two months after administering the first dose. In some aspects, the second dose is administered at least three months after administering the first dose. In some aspects, the second dose is administered at least four months after administering the first dose. In some aspects, the second dose is administered at least five months after administering the first dose. In some aspects, the second dose is administered at least six months after administering the first dose. In some aspects, the second dose is administered at least 12 months after administering the first dose.
  • the intermittent dosing schedule comprises administering to the subject a first set of doses and a second set of doses of a PDGFR ⁇ inhibitor, wherein the second set of doses is administered at least one day after administering the first set of doses.
  • the first set of doses can be, e.g., one, two, three, or four doses per day, administered over a period of, e.g., one, two, three, or four days.
  • the second set of doses can be, e.g., one, two, three, or four doses per day, administered over a period of, e.g., one, two, three, or four days.
  • the second set of doses is administered at least two days after administering the first set of doses. In some aspects, the second set of doses is administered at least three days after administering the first set of doses. In some aspects, the second set of doses is administered at least four days after administering the first set of doses. In some aspects, the second set of doses is administered at least five days after administering the first set of doses. In some aspects, the second set of doses is administered at least six days after administering the first set of doses. In some aspects, the second set of doses is administered at least seven days after administering the first set of doses. In some aspects, the second set of doses is administered at least eight days after administering the first set of doses.
  • the second set of doses is administered at least nine days after administering the first set of doses. In some aspects, the second set of doses is administered at least 10 days after administering the first set of doses. In some aspects, the second set of doses is administered at least 11 days after administering the first set of doses. In some aspects, the second set of doses is administered at least 12 days after administering the first set of doses. In some aspects, the second set of doses is administered at least 13 days after administering the first set of doses. In some aspects, the second set of doses is administered at least two weeks after administering the first set of doses. In some aspects, the second set of doses is administered at least three weeks after administering the first set of doses.
  • the second set of doses is administered at least one month after administering the first set of doses. In some aspects, the second set of doses is administered at least two months after administering the first set of doses. In some aspects, the second set of doses is administered at least three months after administering the first set of doses. In some aspects, the second set of doses is administered at least four months after administering the first set of doses. In some aspects, the second set of doses is administered at least five months after administering the first set of doses. In some aspects, the second set of doses is administered at least six months after administering the first set of doses. In some aspects, the second set of doses is administered at least 12 months after administering the first set of doses.
  • the use of an intermittent dosing schedule allows the OPC population of the treated subject (i.e., subject who previously received an administration of the PDGFR ⁇ inhibitor) to sufficiently recover prior to the next dose of the PDGFR ⁇ inhibitor.
  • OPCs are capable of self-renewal and therefore, under a steady-state condition (i.e., no inhibition of PDGFR ⁇ activity), the OPC population of a subject is continuously replenished and maintained.
  • a PDGFR ⁇ inhibitor described herein when a PDGFR ⁇ inhibitor described herein is administered to a subject, the PDGFR ⁇ inhibitor induces the differentiation of the OPCs within the subject into oligodendrocytes.
  • an intermittent dosing schedule useful for the present disclosure comprises administering a first dose of a PDGFR ⁇ inhibitor and a second dose of a PDGFR ⁇ inhibitor, wherein the second dose of the PDGFR ⁇ inhibitor is administered to the subject after the OPC population of the subject has sufficiently recovered from the effects of the first dose of the PDGFR ⁇ inhibitor.
  • an OPC population has sufficiently recovered is comparable to that of a reference subject, wherein the reference subject comprises (i) the subject prior to the initial administration of the PDGFR ⁇ inhibitor, (ii) a corresponding, normal healthy subject (i.e., does not suffer from a demyelinating disease) who has not received an administration of the PDGFR ⁇ inhibitor, or (iii) both (i) and (ii).
  • the number of OPCs within an OPC population that has sufficiently recovered is at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or 100% of the number of OPCs within the OPC population of the reference subject.
  • a method provided herein comprises administering a first dose and a second dose of a PDGFR ⁇ inhibitor to a subject (e.g., to treat a demyelinating disease)
  • a first dose of the PDGFR ⁇ inhibitor is administered to a subject and then the subject's OPC population is assessed, wherein if the subject's OPC population has sufficiently recovered as compared to a reference subject (e.g., as described above), a second dose of the PDGFR ⁇ inhibitor is administered to the subject.
  • a second dose of the PDGFR ⁇ inhibitor is administered to the subject if the size of the subject's OPC population (e.g., number of OPCs within the OPC population) is at least at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or 100% of the size of the reference subject's OPC population.
  • the additional dose is administered to the subject at the same dosing interval as that used for the first and second doses of the PDGFR ⁇ inhibitor.
  • the method comprises assessing the subject's OPC population again after the second administration, wherein if the subject's OPC population has sufficiently recovered as compared to a reference subject (e.g., as described above), the additional dose of the PDGFR ⁇ inhibitor is administered to the subject.
  • the additional dose of the PDGFR ⁇ inhibitor is administered to the subject if the size of the subject's OPC population (e.g., number of OPCs within the OPC population) is at least at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or 100% of the size of the reference subject's OPC population.
  • the amount of time required for the subject's OPC population to have sufficiently recovered after an initial administration of a PDGFR ⁇ inhibitor is the dosing interval of the PDGFR ⁇ inhibitor.
  • Whether a subject's OPC population has sufficiently recovered can be determined using any suitable method known in the art. For example, in some aspects, a cuprizone animal model can be used.
  • a dosing interval suitable for a PDGFR ⁇ inhibitor described herein can be determined by administering a first dose and second dose of the PDGFR ⁇ inhibitor to the cuprizone animal model, and then assessing the number of newly generated oligodendrocytes present within the brain of the animals (e.g., by quantifying the number of GPR17+ cells within the brain).
  • a suitable dosing interval is the time between the administration of the first dose and the second dose of the PDGFR ⁇ inhibitor or any time longer than that.
  • a suitable dosing interval is longer than the time between the administration of the first dose and the second dose of the PDGFR ⁇ inhibitor.
  • a suitable dosing interval for a PDGFR ⁇ inhibitor is directly related to the plasma level of the PDGFR ⁇ inhibitor.
  • a method of determining a suitable dosing interval for a PDGFR ⁇ inhibitor comprises administering a first dose of the PDGFR ⁇ inhibitor to a subject and determining the plasma level of the PDGFR ⁇ inhibitor in the subject.
  • a suitable dosing interval is the time between administering the first dose of the PDGFR ⁇ inhibitor and the subject's plasma level of the PDGFR ⁇ inhibitor being comparable to that of the reference.
  • a suitable dosing interval is the time between administering the first dose of the PDGFR ⁇ inhibitor and the subject's plasma level of the PDGFR ⁇ inhibitor reaching a reduced level as compared to the reference.
  • the additional dose of the PDGFR ⁇ inhibitor is administered to the subject when the subject's plasma level has decreased by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or about 100% as compared to the reference.
  • the brain to plasma ratio may be assessed in preclinical species using bioanalytical gas or liquid chromatography and mass spectrometry methods.
  • the plasma level of the PDGFR ⁇ inhibitor is related to the half-life of the PDGFR ⁇ inhibitor.
  • an intermittent dosing schedule useful for the present disclosure comprises administering two or more doses of a PDGFR ⁇ inhibitor at a dosing interval that is longer than the half-life of the PDGFR ⁇ inhibitor.
  • the dosing interval is at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 100% longer than the half-life of the PDGFR ⁇ inhibitor.
  • the dosing interval is at least 1.5-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, at least 10-fold, at least 15-fold, at least 20-fold, at least about 30-fold, at least 40-fold, or at least 50-fold longer than the half-life of the PDGFR ⁇ inhibitor.
  • the dosing interval is at least one day, at least two days, at least three days, at least four days, at least five days, at least six days, at least one week, at least two weeks, at least three weeks, at least one month, at least two months, or at least three months longer.
  • the methods described herein e.g., treating a demyelinating disease
  • the PDGFR ⁇ inhibitors described herein are used in conjunction with an immunomodulatory drug.
  • the PDGFR ⁇ inhibitors are used alone in the methods provided herein.
  • immunomodulatory drug refers to a therapeutic agent that works by modulating (e.g., increasing and/or decreasing) one or more aspects of an immune response.
  • an immunomodulatory drug useful for the present disclosure is capable of reducing or alleviating the proinflammatory nature of a demyelinating disease (e.g., multiple sclerosis).
  • the immunomodulatory drug could block the production of proinflammatory mediators, promote the production of anti- inflammatory cytokines (e.g., IL-10 or TGF- ⁇ ), promote the production of regulatory T cells (Tregs), influence B cells, prevent the entry of immune cells into the brain, or any combination thereof.
  • Non-limiting examples of immunomodulatory drugs that are useful for the present disclosure include: interferon beta-1b (BETASERON ® , EXTAVIA ® ), interferon beta-1a (AVONEX ® , REBIF ® ), peginterferon beta-1a (PLEGRIDY ® ), alemtuzumab (LEMTRADA ® ), natalizumab (TYSABRI ® ), ocrelizumab (OCREVUS ® ), ofatumumab (KESIMPTA ® ), glatiramer acetate (COPAXONE ® , GLATOPA ® ), teriflunomide (AUBAGIO ® ), dimethyl fumarate (TECFIDERA ® ), monomethyl fumarate (BAFIERTAM ® ), diroximel fumarate (Vumerity ® ), and combinations thereof.
  • interferon beta-1b BETASERON ® , EXTAVIA ®
  • an additional therapeutic agent e.g., immunomodulatory drug
  • the additional therapeutic agent is administered to the subject prior to the administration of a PDGFR ⁇ inhibitor.
  • any of the methods provided herein e.g., method of treating a demyelinating disease
  • the additional therapeutic agent is administered after the administration of a PDGFR ⁇ inhibitor.
  • the additional therapeutic agent is administered concurrently with the administration of a PDGFR ⁇ inhibitor.
  • a PDGFR ⁇ inhibitor described herein can be administered to a subject in combination with an agent that assists the delivery of the inhibitor to the CNS (e.g., allows the inhibitor to cross the blood-brain barrier).
  • a PDGFR ⁇ inhibitor described herein is administered to a subject in combination with a peptide blood-brain barrier (BBB) shuttle, wherein the peptide BBB shuttle enhances the ability of the PDGFR ⁇ inhibitor to cross the blood-brain barrier and reach the CNS.
  • BBB peptide blood-brain barrier
  • R 1 is selected from: , , nd [0328]
  • R 10 is selected from H, C 1 -C 4 alkyl, C 1 -C 4 alkoxy, aminoC 1 -C 4 alkyl, hydroxyC 1 - C 4 alkyl, and C 1 -C 4 alkylsulfonyl, wherein the C 1 -C 4 alkyl, C 1 -C 4 alkoxy, aminoC 1 -C 4 alkyl, hydroxyC 1 -C 4 alkyl, and C 1 -C 4 alkylsulfonyl can be optionally substituted by one or more substituents selected from hydroxyl, C 1 -C 4 alkoxy, NR 10
  • Clause 12 The compound of clause 11, or a pharmaceutically acceptable salt or solvate thereof, wherein R 1 is .
  • Clause 13 The compound of clause 11 or 12, or a pharmaceutically acceptable salt or solvate thereof, wherein R 10 is CH 3 .
  • Clause 14 The compound of any one of clauses 1 to 13, or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is heterocyclyl optionally substituted with C 1 - C 4 alkyl or oxo.
  • Clause 15. The compound of any one of clauses 1 to 14, or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is substituted with one R 3 .
  • Clause 16 The compound of any one of clauses 1 to 15, or a pharmaceutically acceptable salt or solvate thereof, wherein R 2 is selected from: , ,
  • Clause 18 The compound of any one of clauses 1 to 17, or a pharmaceutically acceptable salt or solvate thereof, having Formula Ia:
  • Clause 21 The compound of any one of clauses 1 to 20, or a pharmaceutically acceptable salt or solvate thereof, wherein R 3 is heteroaryl optionally substituted with one, two, three, four, or five R 30 .
  • Clause 22 The compound of any one of clauses 1 to 21, or a pharmaceutically acceptable salt or solvate thereof, wherein R 3 is selected from: , , , , , , , , [0345] wherein A 1 is selected from O, S, and N and R 36 is selected from hydrogen, optionally substituted C 1 -C 6 alkyl, and optionally substituted C 1 -C 6 alkylaryl.
  • Clause 28 The compound of any of clauses 1 to 22, or a pharmaceutically acceptable salt or solvate thereof, wherein R 3 is selected from: nd .
  • Clause 29 The compound of clause 28, or a pharmaceutically acceptable salt or solvate thereof, wherein R 3 is selected from: , , .
  • Clause 30 The compound of clause 29, or a pharmaceutically acceptable salt or solvate thereof, wherein R 32 is H.
  • Clause 31 The compound of any one of clauses 28 to 30, or a pharmaceutically acceptable salt or solvate thereof, wherein R 31 is selected from:
  • R 31a is selected from H, D, alkylamino, C 1 -C 4 alkyl, and -CF 3
  • R 31b is selected from H, D, halo, hydroxy, amino, alkylamino, C 1 -C 4 alkyl, -CF 3 , and -OCF 3 .
  • Clause 32 The compound of clause 1, or a pharmaceutically acceptable salt or solvate thereof, selected from any one of compounds 156, 250, 275 and 326-406 of Table 1.
  • Clause 33 Clause 33.
  • the compound of any one of clauses 1 to 32 which can exhibit one or more of the following properties: (i) promote the differentiation of an OPC into an oligodendrocyte, (ii) promote the expression of a protein associated with oligodendrocyte differentiation and/or myelination (e.g., G-protein coupled receptor 17 (GPR17), myelin basic protein (MBP), ASPA, GST-pi, CC1, myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, or a combination thereof), (iii) promote the myelination of an axon, (iv) promote the remyelination of a demyelinated axon, (v) inhibit PDGFR ⁇ kinase activity, (vi) achieve a brain to plasma ratio of greater than 0.1 when systemically administered to a subject, and (vii) any combination thereof.
  • Clause 34 The compound of clause 33, which can inhibit PDGFR ⁇ kinase activity.
  • Clause 35 The compound of clause 34, which can inhibit PDGFR ⁇ kinase activity with an IC 50 of less than 500 nM, less than 400 nM, less than 300 nM, less than 200 nM, less than 100 nM, less than 75 nM, less than less than 50 nM, less than 40 nM, less than 30 nM, less than 20 nM, less than 10 nM, or less than 5 nM.
  • Clause 36 Clause 36.
  • Clause 37 The compound of clause 36, wherein the enzymatic PDGFR ⁇ kinase assay comprises 20 ng of purified PDGFR ⁇ protein, 150 ⁇ M of ATP and 1 ⁇ g of substrate, Poly (Glu4Tyr1) in a volume of 15 ⁇ l.
  • Clause 38 A pharmaceutical composition comprising the compound of any one of clauses 1 to 37, or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable excipient.
  • Clause 39 A kit comprising the compound of any one of clauses 1 to 37, or a pharmaceutically acceptable salt or solvate thereof, or the composition of clause 38, and instructions for use.
  • Clause 40 A compound of any one of clauses 1 to 37, or the pharmaceutical composition of clause 38, for use in therapy.
  • Clause 41 A method of treating a demyelinating disease in a subject in need thereof comprising administering to the subject a therapeutically effective amount of the compound of any one of clauses 1 to 37 or the pharmaceutical composition of clause 38.
  • Clause 42 A method of treating a demyelinating disease in a subject in need thereof comprising administering to the subject a therapeutically effective amount of the compound of any one of clauses 1 to 37 or the pharmaceutical composition of clause 38.
  • a method of improving a subject's performance in a test for assessing one or more symptoms associated with a demyelinating disease comprising administering to the subject a therapeutically effective amount of the compound of any one of clauses 1 to 37 or the pharmaceutical composition of clause 38, wherein after the administration, the subject's performance in the test is improved as compared to a reference subject (e.g., the subject prior to the administration).
  • test is selected from a visual evoked potential (VEP) test, a multifocal visual evoked potential (mfVEP) test, a low contrast visual acuity (LC-VA) test, a magnetic resonance imaging (MRI) (e.g., magnetic transfer resonance, myelin water fraction (MWF), and quantitative susceptibility mapping (QSM)), an electromyography (EMG), a nerve conduction velocity (NCV) test, an Extended Disability Status Scale (EDSS), a timed walk test (e.g., timed 25-foot walk), a Nine-Hole Peg Test (9HPT), an ocular coherence tomograph (OCT), a quality of life measure test (e.g., Multiple Sclerosis Quality of Life-54 and Vision-Related Quality of Life), cognitive assessment (e.g., Montreal Cognitive Assessment), or combinations thereof.
  • VEP visual evoked potential
  • mfVEP multifocal visual evoked potential
  • LC-VA low contrast visual acuity
  • the demyelinating disease comprises an acute disseminated encephalomyelitis (ADEM), acute hemorrhagic leukoencephalitis, acute transverse myelitis, adrenoleukodystrophy, adrenomyeloneuropathy, Alexander disease, Alzheimer's disease, aminoacidurias, amyotrophic lateral sclerosis, anti-MAG peripheral neuropathy, anti-MOG associated spectrum, Balo concentric sclerosis, brain injury, CAMFAK syndrome, Canavan disease, carbon monoxide toxicity, central pontine myelinolysis, cerebral hypoxia, cerebral ischemia, Charcot–Marie–Tooth disease, chronic inflammatory demyelinating polyneuropathy, chronic traumatic encephalopathy, clinically isolated syndrome (CIS), congenital cataract, copper deficiency associated condition, delayed post-hypoxic leukoencephalopathy, diffuse cerebral sclerosis of Schilder, diffuse
  • ADAM acute disseminated encephalomyelitis
  • Alexander disease
  • Clause 45 The method of any one of clauses 41 to 44, wherein the demyelinating disease is characterized by demyelination of one or more cells within the CNS of the subject.
  • Clause 46 The method of any one of clauses 41 to 45, wherein the demyelinating disease is multiple sclerosis.
  • Clause 47 The method of clause 46, wherein the multiple sclerosis comprises a clinically isolated syndrome ("CIS"), relapsing-remitting MS ("RRMS”), secondary progressive MS (“SPMS”), primary progressive MS (“PPMS”), optic neuritis or transverse myelitis.
  • CIS clinically isolated syndrome
  • RRMS relapsing-remitting MS
  • SPMS secondary progressive MS
  • PPMS primary progressive MS
  • optic neuritis or transverse myelitis optic neuritis or transverse myelitis.
  • the one or more symptoms comprise fatigue, dizziness, malaise, elevated fever and high body temperature, extreme sensitivity to cold in the hands and feet, weakness and stiffness in muscles and joints, weight changes, digestive or gastrointestinal problems, low blood pressure, high blood pressure, irritability, anxiety, depression, impaired vision (e.g., blurred vision, double vision, reduction in low contrast visual acuity (LC-VA)), ataxia, clonus, spasms, dysarthria, weakness, clumsiness, hand paralysis, hemiparesis, genital anesthesia, sexual dysfunction, incoordination, paresthesias, ocular paralysis, impaired muscle coordination, loss of sensation, tingling, numbness, pain, neurological symptoms, impaired cognition, unsteady gait, balance problems, dizziness, spastic paraparesis, incontinence, hearing problems, speech problems, loss of olfaction, agusia, or combinations thereof.
  • impaired vision e.g., blurred vision, double
  • Clause 51 A method of promoting the myelination of an axon in a subject in need thereof, the method comprising administering to the subject an effective amount of the compound of any one of clauses 1 to 37 or the pharmaceutical composition of clause 38.
  • Clause 52 The method of clause 51, wherein promoting the myelination of an axon results in an increase in the expression of one or more of the following markers within the subject: myelin basic protein (MBP) Myelin Oligodendrocyte Glycoprotein (MOG), Oligodendrocyte Specific Protein/Claudin-11, CNPase, or any combination thereof.
  • MBP myelin basic protein
  • MOG Oligodendrocyte Glycoprotein
  • CNPase Oligodendrocyte Specific Protein/Claudin-11
  • CNPase CNPase
  • Clause 61 A method according to any one of clauses 51 to 60, wherein the subject has, or is at risk of developing a demyelinating disease, for example a disease according to any one of clauses 44 to 50.
  • Clause 62 A method according to any one of clauses 51 to 60, wherein the method is a method of treating or preventing a demyelinating disease, for example a disease according to any one of clauses 44 to 50.
  • Clause 63 The method of any one of clauses 41 to 62, wherein the compound or the pharmaceutical composition is administered to the subject once.
  • Clause 65 The method of clause 64, wherein the intermittent dosing comprises administering the compound or the pharmaceutical composition to the subject every other day, every three days, every four days, every five days, every six days, once a week, every eight days, every nine days, every 10 days, every 11 days, every 12 days, every 13 days, once every two weeks, once every three weeks, once a month, once every two months, once every three months, once every four months, once every five months, once every six months, or once every twelve months.
  • Clause 66 Clause 66.
  • the intermittent dosing comprises administering to the subject a first dose and a second dose of the compound or the pharmaceutical composition, wherein the second dose is administered at least one day, at least two days, at least three days, at least four days, at least five days, at least six days, at least seven days, at least eight days, at least nine days, at least 10 days, at least 11 days, at least 12 days, at least 13 days, at least two weeks, at least three weeks, at least one month, at least two months, at least three months, at least four months, at least five months, at least six months, or at least 12 months after administering the first dose.
  • Clause 67 Clause 67.
  • Clause 71 The method of clause 69 or 70, wherein the additional therapeutic agent comprises an immunomodulatory agent.
  • Clause 72 The method of clause 71, wherein the additional therapeutic agent is selected from interferon beta-1b, interferon beta-1a, peginterferon beta-1a, alemtuzumab, natalizumab, ocrelizumab, ofatumumab, glatiramer acetate, teriflunomide, dimethyl fumarate, monomethyl fumarate, diroximel fumarate, fingolimod hydrochloride, siponimod fumaric acid, ozanimod hydrochloride, BTK inhibitor, or a pharmaceutically acceptable salt thereof [0398] Clause 73.
  • Clause 74 A method of inducing the differentiation of an oligodendrocyte progenitor cell (OPC) into an oligodendrocyte, the method comprising contacting the OPC with an effective amount of the compound of any one of clauses 1 to 37 or the pharmaceutical composition of clause 38.
  • OPC oligodendrocyte progenitor cell
  • Clause 81 A method of treating a relapsing form of multiple sclerosis in a subject in need thereof, the method comprising administering to the subject an effective amount of the compound of any one of clauses 1 to 37 or the pharmaceutical composition of clause 38.
  • Clause 82 A method of treating a relapsing form of multiple sclerosis in a subject in need thereof, the method comprising administering to the subject an effective amount of the compound of any one of clauses 1 to 37 or the pharmaceutical composition of clause 38.
  • Clause 85 The compound of any one of clauses 1 to 37 or the pharmaceutical composition of clause 38, for use in a method of one or more of the following: (i) promote the differentiation of an OPC into an oligodendrocyte, (ii) promote the expression of a protein associated with oligodendrocyte differentiation and/or myelination (e.g., G-protein coupled receptor 17, myelin basic protein (MBP), ASPA, GST-pi, CC1, myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, or a combination thereof), (iii) promote the myelination of an axon, (iv) promote the remyelination of a demyelinated axon, (v) inhibit PDGFR ⁇ kinase activity, (vi) achieve a brain to plasma ratio of greater than 0.1 when systemically administered to a subject, and (vii) promote a
  • Clause 86 A method of treating a PDGF-associated tumor in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound of any one of clauses 1 to 37 or the pharmaceutical composition of clause 38, wherein after the administration, PDGFR ⁇ activity is reduced in the subject.
  • Clause 87 The method of clause 86, wherein the PDGF-associated tumor comprises an oligodendroglioma.
  • Clause 88. The method of clause 86 or 87, wherein the method is a method of treatment by therapy.
  • the reaction was sparged with argon for 10 min, then heated to 75 °C for 6 h under an argon. The reaction was cooled to rt and stirred overnight. The mixture was concentrated under reduced pressure then water (30 mL) was added. The aqueous mixture was extracted with 10% EtOH in EtOAc (4 x 50 mL) and 50% EtOAc in THF (3 x 50 mL). Brine (30 mL) was added to the aqueous layer, then extracted with 10% EtOH in EtOAc (3 x 50 mL). The organic layers were combined, dried over anhydrous Na 2 SO 4 , and filtered.
  • the reaction was stirred at 120 °C for 27 h, then cooled to rt, and concentrated under reduced pressure. The residue was partitioned between sat. aqueous NaHCO 3 (100 mL) and 10% MeOH in DCM (100 mL). The aqueous layer was washed with 10% MeOH in DCM (4 x 100 mL). The organic layers were combined, dried with anhydrous Na 2 SO 4 , and filtered.
  • the aqueous layer was washed with DCM (2 x 300 mL).
  • the aqueous layer containing an emulsion was filtered through a pad of Celite.
  • the filter pad was washed with DCM which was then used to wash the aqueous layer (2 x 300 mL).
  • the filter pad was then washed with 10% MeOH in DCM that was in turn used to wash the aqueous layer (4 x 300 mL).
  • the aqueous layer was washed again with 10% MeOH in DCM (4 x 100 mL) or until no product was evident in the aqueous layer by HPLC.
  • the organic layers were combined, dried with anhydrous Na 2 SO 4 , and filtered.
  • Step 2.2-Chloro-5-(pyridin-2-ylmethyl)pyrimidine (Compound S80) [0453] 5-Bromo-2-chloropyrimidine (110 mg, 0.590 mmol), Xantphos (22 mg, 0.04 mmol), Pd2(dba)3 (10 mg, 0.01 mmol), potassium pyridin-2-ylacetate (125 mg, 0.713 mmol, as prepared in the previous step), and mesitylene (1.2 mL) were added to a pressure tube that was pre-flushed with argon. It was sparged with more argon, sealed, and heated at 150°C for 24 h.
  • Step 2.1-(2-methylquinolin-6-yl)ethan-1-one To a stirred solution of N-methoxy-N,2-dimethylquinoline-6-carboxamide (1.5 g, 6.5 mmol, as prepared in the previous step) in THF (20 mL) cooled to -78°C was added 3M MeMgBr in THF (5 mL, 19.5 mmol) dropwise under nitrogen then the mixture was stirred at -78°C for 1 h under nitrogen. The reaction was warmed to 0°C, quenched with saturated aqueous NH 4 Cl solution, and extracted with DCM (3x300 mL). The organic extracts were combined, dried over anhydrous Na 2 SO 4 , and filtered.
  • Step 3.1-(2-methylquinolin-6-yl)ethan-1-ol (Compound S117) [0496] To a mixture of 1-(2-methylquinolin-6-yl)ethan-1-one (500 mg, 2.7 mmol, as prepared in the previous step) in MeOH (10 mL) cooled to 0°C was added NaBH4 (204 mg, 5.4 mmol) then the mixture was stirred for 1 h. The reaction was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with PE/EtOAc (1:1) to afford 320 mg (63%) of 1-(2-methylquinolin-6-yl)ethan-1-ol (Compound S117) as colorless oil.
  • Step 2.1-(2-chloroquinolin-6-yl)ethan-1-ol (Compound S124) [0504] To a solution of 1-(2-chloroquinolin-6-yl)ethan-1-one (190 mg, 0.9 mmol, as prepared in the previous step) in MeOH (10 mL) cooled to 0°C was added NaBH4 (41.9 mg, 1.1 mmol) in portions then the mixture was stirred at 0°C for 1 h.
  • Example 1 Synthesis of Exemplary Compound 156 3-(4-(1-benzyl-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine (Compound 156) Step 1.
  • Example 2 Synthesis of Exemplary Compound 275 1-(5-benzylpyrimidin-2-yl)-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)-1,4- diazepan-2-one (Compound 275).
  • Step 3 Preparation of ethyl N-(3-aminopropyl)-N-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)glycinate hydrochloride salt [0516] To a solution of ethyl N-(3-((tert-butoxycarbonyl)amino)propyl)-N-(6-(1-methyl- 1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)glycinate (300 mg, 0.68 mmol, as prepared in the previous step) in DCM (10 mL) was added 4M HCl in dioxane (3 mL) then the mixture was stirred at rt under nitrogen for 2 h.
  • Example 10 Synthesis of Exemplary Compound 333 3-(4-(isobutylsulfonyl)cyclohexyl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 333) Step 1.
  • Step 3 Preparation of 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)cyclohexyl methanesulfonate [0535] To a solution of 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)cyclohexan-1-ol (615 mg, 2.08 mmol, as prepared in the previous step) and TEA (420 mg, 4.15 mmol) in DCM (10 mL) was added Ms2O (542 mg, 3.11 mmol) at 0°C then the reaction was stirred at rt for 2 h.
  • Example 11 Synthesis of Exemplary Compound 334 tert-butyl 6-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)-3,6- diazabicyclo[3.2.1]octane-3-carboxylate (Compound 334) [0539] To a stirred solution of 3-bromo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridine (Compound S9) (1.20 g, 4.33 mmol) and tert-butyl 3,6- diazabicyclo[3.2.1]octane-3-carboxylate (1.84 g, 8.66 mmol) in tBuOH (20 mL) and dioxane (10 mL) were added KOtBu (728.8
  • Example 12 Synthesis of Exemplary Compound 335 Mixture of 3-(4-(5-benzylpyrimidin-2-yl)-2,3,6,7-tetrahydro-1H-azepin-1-yl)-6-(1-methyl- 1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine and 3-(5-(5-benzylpyrimidin-2-yl)-2,3,4,7- tetrahydro-1H-azepin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 335) Step 1.
  • Example 17 Synthesis of Exemplary Compound 340 3-(3-(5-benzylpyrimidin-2-yl)-3,6-diazabicyclo[3.2.1]octan-6-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine (Compound 340) Step 1.
  • Example 18 Synthesis of Exemplary Compound 341 4-(5-benzylpyrimidin-2-yl)-1-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)-1,4- diazepan-5-one (Compound 341) Step 1.
  • Example 20 Synthesis of Exemplary Compound 343 (R)-1-(5-methylpyridin-2-yl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate (Compound 343) Step 1.
  • the mixture was diluted with sat. aqueous NaHCO 3 (50 mL) and EtOAc (50 mL). The layers were separated and the EtOAc layer was washed with water (3 x 20 mL) and sat. aqueous NaHCO 3 (20 mL), dried over anhydrous Na 2 SO 4 , and filtered.
  • Example 21 Synthesis of Exemplary Compound 345 tert-butyl 4-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-a]pyridin-3-yl)piperazine-1- carboxylate (Compound 345) [0560] To a glass vial containing 3-bromo-7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2- a]pyridine (Compound S7) (0.250 g, 0.90 mmol) and tert-butyl-1-piperazinecarboxylate (0.672 g, 3.61 mmol) was added NaOtBu (0.152 g, 1.35 mmol) and tBuXPhos Pd G1 (0.062 g, 0.09 mmol) and the headspace was flushed with nitrogen.
  • Compound 345 tert-butyl 4-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-a]pyridin-3-y
  • Example 22 Synthesis of Exemplary Compound 344 (R)-1-(4-chlorophenyl)ethyl 4-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-a]pyridin-3- yl)piperazine-1-carboxylate (Compound 344) Step 1.
  • Example 23 Synthesis of Exemplary Compound 346 tert-butyl 4-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-a]pyridin-3-yl)-1,4-diazepane-1- carboxylate (Compound 346) Step 1.
  • Example 24 Synthesis of Exemplary Compound 347 (R)-1-phenylethyl 4-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-a]pyridin-3-yl)-1,4- diazepane-1-carboxylate (Compound 347) Step 1.
  • Example 25 Synthesis of Exemplary Compound 348 3-(3-(5-benzylpyrimidin-2-yl)pyrrolidin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- b]pyridazine (Compound 348) [0567] To a solution of 5-benzyl-2-pyrrolidin-3-ylpyrimidine (Compound S100) (344.2 mg, 1.44 mmol) in dioxane (8 mL) was added 3-bromo-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-b]pyridazine (Compound S8) (200.0 mg, 0.72 mmol), C s2 CO 3 (468.6 mg, 1.44 mmol), and tBuXPhos Pd G1 (74.1 mg, 0.11 mmol) at rt under nitrogen then the reaction was stirred at 110°C under nitrogen for 10 h.
  • Compound S100
  • Example 26 Synthesis of Exemplary Compound 349 3-(3-(1-benzyl-1H-1,2,4-triazol-3-yl)pyrrolidin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-b]pyridazine (Compound 349) [0568] To a solution of 1-benzyl-3-(pyrrolidin-3-yl)-1H-1,2,4-triazole (Compound S101) (492.5 mg, 2.16 mmol) in dioxane (10 mL) was added 3-bromo-6-(1-methyl-1H-pyrazol- 4-yl)pyrazolo[1,5-b]pyridazine (Compound S8) (300.0 mg, 1.08 mmol), C s2 CO 3 (1.05 g, 3.24 mmol), and tBuXPhos Pd G1 (111.1 mg, 0.16 mmol) at rt under nitrogen then the reaction was stirred at
  • Example 27 Synthesis of Exemplary Compound 350 3-(3-(5-benzylpyrimidin-2-yl)pyrrolidin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrazine (Compound 350) Step 1.
  • Example 29 Synthesis of Exemplary Compound 352 3-(4-(5-benzylpyrimidin-2-yl)-1,4-diazepan-1-yl)-7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2- a]pyridine (Compound 352) Step 1.
  • Example 30 Synthesis of Exemplary Compound 353 N-((5-chloropyridin-2-yl)methyl)-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxamide (Compound 353) [0575] To a solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-piperazin-1-ylpyrazolo[1,5- a]pyridine (Compound 12) (300 mg, 1.06 mmol), TEA (323 mg, 3.19 mmol) in DCM (15 mL) was added triphosgene (94.6 mg, 0.32 mmol) at 0°C.
  • Example 33 Synthesis of Exemplary Compound 356 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)-N-((6-methylpyridin-3- yl)methyl)piperazine-1-carboxamide (Compound 356) [0578] To a solution of (6-methylpyridin-3-yl)methanamine (400 mg, 3.27 mmol) and TEA (994 mg, 9.82 mmol) in DCM (5 mL) cooled to 0°C was added triphosgene (291.5 mg, 0.98 mmol) then the reaction was stirred at 0°C under nitrogen for 2 h.
  • Example 35 Synthesis of Exemplary Compound 358 3-(3-(1-benzyl-1H-1,2,4-triazol-3-yl)pyrrolidin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine (Compound 358) [0580] To a vial pre-sparged with argon 20 minutes containing 3-iodo-6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridine (234 mg, 0.72 mmol, as prepared for Compound S57, Step 1), 1-benzyl-3-pyrrolidin-3-yl-1H-1,2,4-triazole (Compound S101) (110 mg, 0.48 mmol), K 3 PO 4 (307 mg, 1.44 mmol), and CuI (37 mg, 0.193 mmol) was added 1-butanol (2.8 mL) and 1, 2-ethanediol (0.6 mL
  • Example 36 Synthesis of Exemplary Compound 359 (R)-1-(6-methylpyridin-3-yl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate (Compound 359) Step 1.
  • Example 39 Synthesis of Exemplary Compound 362 (R)-N-(4-chlorobenzyl)-2-methyl-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxamide (Compound 362) Step 1.
  • Example 45 Synthesis of Exemplary Compound 368 (R)-3-(4-(1-benzyl-1H-1,2,4-triazol-3-yl)-3-methylpiperazin-1-yl)-6-(1-methyl-1H-pyrazol- 4-yl)pyrazolo[1,5-a]pyridine (Compound 368) [0600] To a solution of (R)-6-(1-methyl-1H-pyrazol-4-yl)-3-(3-methylpiperazin-1- yl)pyrazolo[1,5-a]pyridine hydrochloride salt (300 mg, 0.90 mmol, as prepared in Example 39, Step 1) and 1-benzyl-3-chloro-1H-1,2,4-triazole (174 mg, 0.90 mmol, as prepared in Example 1, Step 1) in dioxane (10 mL) were
  • Example 51 Synthesis of Exemplary Compound 374 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(5-((5-methylpyridin-2-yl)methyl)pyrimidin-2- yl)piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 374) Step 1. Preparation of potassium 2-(5-methylpyridin-2-yl)acetate [0608] To a solution of methyl 2-(5-methylpyridin-2-yl)acetate (2.00 g, 12.11 mmol) dissolved in EtOH (20 mL) was added H 2 O (0.23 g, 12.71 mmol). The resulting mixture was stirred for 10 minutes at 60°C under nitrogen.
  • Example 52 Synthesis of Exemplary Compound 375 3-(4-(1-benzyl-1H-pyrazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridine (Compound 375) Step 1.
  • Example 53 Synthesis of Exemplary Compound 376 3-(4-(1-(4-fluorobenzyl)-1H-1,2,4-triazol-5-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine (Compound 376) Step 1.
  • Example 54 Synthesis of Exemplary Compound 377 3-(4-(1-benzyl-1H-pyrazol-5-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridine (Compound 377) Step 1.
  • Example 56 Synthesis of Exemplary Compound 379 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(pyridin-2-ylmethyl)-1H-1,2,4-triazol-3-yl)piperazin- 1-yl)pyrazolo[1,5-a]pyridine (Compound 379) Step 1.
  • Example 58 Synthesis of Exemplary Compound 381 3-(4-(1-(4-fluorobenzyl)-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyrimidine (Compound 381) [0624] To a solution of 3-chloro-1-(4-fluorobenzyl)-1H-1,2,4-triazole (194.1 mg, 0.91 mmol, as prepared in Example 49, Step 1) and 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin- 1-yl)pyrazolo[1,5-a]pyrimidine (Compound S98) (200 mg, 0.706 mmol) in dioxane (20 mL) under nitrogen were added C s2 CO 3 (460.5 mg
  • Example 60 Synthesis of Exemplary Compound 383 3-(3-(1-benzyl-1H-1,2,4-triazol-3-yl)pyrrolidin-1-yl)-7-(1-methyl-1H-pyrazol-4- yl)imidazo[1,2-b]pyridazine (Compound 383) Step 1.
  • Example 61 Synthesis of Exemplary Compound 384 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(pyridin-3-ylmethyl)-1H-1,2,4-triazol-3-yl)piperazin- 1-yl)pyrazolo[1,5-a]pyridine (Compound 384) Step 1.
  • Example 63 Synthesis of Exemplary Compound 386 3-(4-(5-benzylpyrimidin-2-yl)-1,4-diazepan-1-yl)-7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2- b]pyridazine (Compound 386) Step 1.
  • the mixture was filtered through a pad of Celite, and the layers separated.
  • the filter pad was washed with DCM and this filtrate was used to extract the aqueous layer.
  • the filter pad was washed with DCM and the filtrate was used to extract the aqueous layer an additional three times until the aqueous layer showed no desired product by HPLC.
  • the filter pad was washed with DCM until HPLC showed no additional product eluted from the Celite pad. All the DCM extracts were combined, dried over anhydrous Na 2 SO 4 , and filtered.
  • Example 64 Synthesis of Exemplary Compound 387 N-(4-chlorobenzyl)-4-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazin-3- yl)piperazine-1-carboxamide (Compound 387) Step 1.
  • the reaction was treated with tBuXPhos Pd G1 (2.43 g, 3.54 mmol) and the mixture sparged again for another 10 minutes. The reaction was sparged an additional 2 minutes while being agitated in an ultrasound bath. The reaction was then warmed at 55 °C under argon for 3 h. The reaction was concentrated under reduced pressure and the residue was diluted with DCM (300 mL). The mixture was washed with 10% citric acid (4 x 100 mL). The combined citric acid washes were extracted with DCM (100 mL). The combined DCM extracts were washed with water (3 x 100 mL). The combined water washes were extracted with DCM (100 mL). The combined DCM extracts were washed with sat.
  • the reaction was warmed to rt and stirred for 30 minutes, then additional ACN (3 mL) was added. After stirring 1 h, the reaction was concentrated under reduced pressure. The residue was partitioned between water (10 mL), DCM (10 mL), and 10% MeOH in DCM (5 mL). The water layer was washed with 10% MeOH in DCM (3 x 5 mL). The combined organic extracts were washed with sat. aqueous NaHCO 3 (5 mL), and brine (5 mL), dried over anhydrous Na 2 SO 4 , and filtered.
  • Example 65 Synthesis of Exemplary Compound 388 2-benzyl-5-(4-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazin-3-yl)piperazin-1-yl)- 1,3,4-thiadiazole (Compound 388) [0639] To a mixture of 2-benzyl-5-bromo-1,3,4-thiadiazole (243 mg, 0.95 mmol, as prepared in Example 55, Step 2) and 7-(1-methyl-1H-pyrazol-4-yl)-3-piperazin-1- ylimadazo[1,2-b]pyridazine trihydrochloride salt (404 mg, 1.03 mmol, as prepared in Example 64, Step 2) in DMAc (6.8 mL) was added DIEA (1.60 mL
  • Example 66 Synthesis of Exemplary Compound 389 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-((6-methylpyridin-3-yl)methyl)-1H-1,2,4-triazol-3- yl)piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 389) Step 1.
  • Example 69 Synthesis of Exemplary Compound 392 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(1-phenylethyl)-1H-1,2,4-triazol-3-yl)piperazin-1- yl)pyrazolo[1,5-a]pyridine (Compound 392) Step 1.
  • Example 71 Synthesis of Exemplary Compound 394 3-(3-(5-benzylpyrimidin-2-yl)pyrrolidin-1-yl)-7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2- b]pyridazine (Compound 394) [0649] A mixture of 3-bromo-7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazine (Compound S3) (0.186 g) and 5-benzyl-2-pyrrolidin-3-ylpyrimidine (Compound S100) (0.400 g, 1.67 mmol) in tBuOH (1.85 mL) and dioxane (0.939 mL) was sparged with argon for 15 minutes then tBuXPhos Pd G1
  • the reaction was warmed to 60°C and stirred for 1 h.
  • the reaction was cooled to rt andL concentrated under reduced pressure.
  • the residue was partitioned between EtOAc (40 mL) and water (10 mL).
  • the aqueous layer was washed with EtOAc (3 x 40 mL).
  • the combined organic extracts were dried over anhydrous Na 2 SO 4 , and filtered.
  • the filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with 0-100% (10% MeOH in DCM) / DCM.
  • the pure fractions were combined and concentrated under reduced pressure then the residue was dissolved in hexane and concentrated under reduced pressure. This was repeated until a yellow solid formed.
  • Example 73 Synthesis of Exemplary Compound 396 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(pyridin-4-ylmethyl)-1H-1,2,4-triazol-3-yl)piperazin- 1-yl)pyrazolo[1,5-a]pyridine (Compound 396) Step 1.4-((3-chloro-1H-1,2,4-triazol-1-yl)methyl)pyridine [0651] To a solution of 3-chloro-1H-1,2,4-triazole (2.00 g, 19.32 mmol) and 4- (bromomethyl)pyridine (3.99 g, 23.19 mmol) in DMF (40 mL) was added K2CO 3 (5.34 g, 38.64 mmol) then the reaction was warmed to 80°C and stirred overnight
  • Example 78 Synthesis of Exemplary Compound 401 2-benzyl-5-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrazin-3-yl)piperazin-1-yl)- 1,3,4-thiadiazole (Compound 401) [0657] To a solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)pyrazolo[1,5- a]pyrazine (Compound S103) (60 mg, 0.21 mmol) in DMSO (2 mL) under nitrogen was added 2-benzyl-5-bromo-1,3,4-thiadiazole (64.8 mg, 0.25 mmol, as prepared in Example 55, Step 2) and DIEA (82.1 mg, 0.64 mmol) then the reaction was heated to 110°C and stirred 16 h under nitrogen.
  • 2-benzyl-5-bromo-1,3,4-thiadiazole 64.8 mg, 0.25
  • Example 79 Synthesis of Exemplary Compound 402 benzo[d]thiazol-6-ylmethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate (Compound 402) [0658] To a solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-piperazin-1-ylpyrazolo[1,5- a]pyridine (Compound 12) (150 mg, 0.53 mmol) and 1,3-benzothiazol-6-ylmethanol (263 mg, 1.59 mmol) in pyridine (2 mL) cooled to 0°C was added triphosgene (473 mg, 1.59 mmol) then the reaction was stirred at 0°C for 1 h.
  • triphosgene 473 mg, 1.59 mmol
  • Example 81 Synthesis of Exemplary Compound 404 3-(4-(1-benzyl-1H-1,2,4-triazol-3-yl)piperazin-1-yl-2,2,3,3,5,5,6,6-d8)-6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 404) Step 1.
  • tBuXPhos Pd G1 (0.480 g, 0.70 mmol) was added and the mixture was sparged for 5 minutes with argon, then the reaction was heated to 55°C and stirred for 2 h under argon. The mixture was cooled to rt, diluted with EtOAc, washed with water and brine, dried over anhydrous Na 2 SO 4 , and filtered.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Psychiatry (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Hospice & Palliative Care (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present disclosure provides compounds that can specifically target a PDGFRα, and thereby, reduce and/or inhibit the activation of the receptor ("PDGFRα inhibitor"). The present disclosure also provides methods of treating a demyelinating disease using the disclosed PDGFRα inhibitors.

Description

HETEROCYCLIC COMPOUNDS AND USES THEREOF CROSS-REFERENCE TO RELATED APPLICATION [0001] This PCT application claims the priority benefit of U.S. Provisional Application No.63/501,005, filed May 9, 2023, which is incorporated herein by reference in its entirety. REFERENCE TO SEQUENCE LISTING SUBMITTED ELECTRONICALLY VIA EFS-WEB [0002] The content of the electronically submitted sequence listing (Name: 5411_003PC01_Seqlisting_ST26; Size: 29,305 bytes; and Date of Creation: May 8, 2024) submitted in this application is incorporated herein by reference in its entirety. FIELD OF THE DISCLOSURE [0003] The present disclosure relates to compounds that can promote remyelination. Specifically, the disclosure relates to PDGFRα inhibitors and the use of such inhibitors to treat diseases associated with impaired myelination (e.g., multiple sclerosis). BACKGROUND OF THE DISCLOSURE [0004] Proper myelination by oligodendrocytes is essential for central nervous system (CNS) development and function. Gacem et al., Life 11(4): 327 (Apr. 2021). Myelin maintains axon potential conduction velocity of neural signals and provides metabolic support to axons, supporting their survival. During normal development, oligodendrocyte progenitor cells (OPCs) undergo morphological and molecular changes as they differentiate into oligodendrocytes that can myelinate axons. Accordingly, any abnormal developmental processes or pathogenic immune activation and failure of oligodendrocytes to myelinate axons or loss of myelin can lead to neurodegenerative diseases such as multiple sclerosis. [0005] Multiple sclerosis (MS) is generally characterized by inflammation and demyelination of neuronal axons. Patients with early MS often suffer from isolated immune attacks with effective remyelination and recovery between attacks. However, as the disease progresses, MS patients have a reduced ability to effectively remyelinate resulting in permanent neurological disability. Current treatments for MS and other neurodegenerative diseases largely focus on reducing the immune attack on myelin but do not restore myelin on the damaged neuronal axons. [0006] Therefore, there remains a need for a new and more effective treatment of such demyelinating diseases, particularly treatments that can induce the remyelination of demyelinated, vulnerable neuronal axons. BRIEF SUMMARY OF THE DISCLOSURE [0007] The present disclosure describes the discovery that compounds that inhibit PDGFRα can promote remyelination. Compounds and methods of the present disclosure are therefore useful for treating diseases associated with demyelination (e.g., hypomyelination). [0008] Provided herein is a compound of Formula I:
Figure imgf000003_0001
, [0009] or a pharmaceutically acceptable salt or solvate thereof, wherein: indicates a single bond or a double bond such that all valences are satisfied; X1, X2, X3, and X4 are selected from N and CRa, with the proviso that not more than two of X1, X2, X3, and X4 are N; one of Y1 and Y2 is N and the other of Y1 and Y2 is C; each Ra is independently selected from H, halo, C1-C4alkyl, and C1-C4alkoxy; R1 is selected from C1-C4alkyl, C3-C8cycloalkyl, 3-8 membered heterocyclyl, heteroaryl, aryl, and C1-C8alkoxy, all of which can be optionally substituted with one, two, three, four, five, or six substituents selected from halo, hydroxy, oxo, C1-C4alkyl, aminoC1- C4alkyl, hydroxyC1-C4alkyl, C1-C4alkoxy, C1-C4alkoxyC1-C4alkyl, 3-8 membered heterocyclyl, and 3-8 membered heterocyclylC1-C4alkyl, with the proviso that the number of substituents does not exceed the number of substitutable positions; R2 is selected from cycloalkyl, cycloalkenyl, alkyl, oxoalkylamino, aminoalkylamino, amino, heterocyclyl, heteroaryl, aminoheterocyclyl, heterocyclylamino, and aminoalkylamino, all of which can be optionally substituted with one or more substituents selected from D, halo, hydroxy, oxo, and C1-C4alkyl; R2 is substituted by one, two, or three R3; R3 is selected from aryl, heteroaryl, -C(O)R31, -C(O)OR31, -C(O)NR31R32, -S(O)2NR31R32, -S(O)(NR33)R31, -S(O)2R31, -S(O)(NR33)NR31R32, -C(S)NR31R32, C3-C8cycloalkyl, 3-8 membered heterocyclyl, and C1-C4alkyl, all of which can be optionally substituted with one, two, three, four, or five R30; each R30 is independently selected from D, halo, aryl, -OR300, -NR300R303, -S(O)rR300, -C(O)R300, -C(=CR34R35)R300, and
Figure imgf000004_0001
; r is selected from 0, 1, and 2; each R300 is independently selected from C1-C6alkyl, C3-C7cycloalkyl, aryl, heteroaryl, 3-8 membered heterocyclyl, and 3-8 membered heterocyclylaryl, all of which can be optionally substituted with one, two, three, four, or five substituents selected from D, halo, hydroxy, amino, alkylamino, cyano, C1-C4alkyl, C1-C4haloalkyl, and C1-C4alkoxy; each R301 is independently selected from H, halo, and C1-C4alkyl; each R302 is independently selected from H, F, hydroxyl, amino, alkylamino, oxo, and C1-C4alkyoxy; each R303 is independently selected from H and C1-C4alkyl; n, o, and p are each independently selected from 0, 1, 2, 3, and 4; each R31 is independently selected from C1-C8alkyl, arylC1-C4alkyl, heteroarylC1- C4alkyl, heterocyclyl, heterocyclylC1-C4alkyl, cycloalkyl, and cycloalkylC1-C4alkyl, all of which can be optionally substituted with one, two, three, four, or five substituents selected from D, halo, cyano, hydroxy, amino, -OCF3, C1-C4alkyl, C1-C4haloalkyl, C1-C4alkoxy, hydroxyC1-C4alkyl, -S(O)2NR304R305, -C(O)OR304R305, -C(O)NR304R305, and -NR304C(O)R305; each R304 and R305 is independently selected from H and C1-C4alkyl; each R32 is independently selected from H and C1-C4alkyl; or R31 and R32 together with the atom to which they are connected to form a 5-8 membered heterocycyl, optionally substituted with one, two, three, four, or five substituents selected from D, halo, cyano, C1-C4alkyl, C1-C4haloalkyl, and -C(O)NR34R35; and each R34 and R35 is independently selected from H, C1-C4alkyl, and C1-C4haloalkyl; and each R33 is independently selected from H, C1-C4alkyl, C1-C4haloalkyl, and –C(O)R34; or R31 and R33 together with the atoms to which they are connected form a 4-8 membered heterocycyl. [0010] In some aspects, Y1 is N and Y2 is C. [0011] In some aspects, Y1 is C and Y2 is N. [0012] In some aspects, X1 is N, X2 is CRa, X3 is CRa, and X4 is CRa. [0013] In some aspects, X1 is CRa, X2 is N, X3 is CRa, and X4 is CRa. [0014] In some aspects, X1 is CRa, X2 is CRa, X3 is N, and X4 is CRa. [0015] In some aspects, X1 is CRa, X2 is CRa, X3 is CRa, and X4 is N. [0016] In some aspects, X1 is CRa, X2 is CRa, X3 is CRa, and X4 is CRa. [0017] In some aspects, R1 is 5- or 6-membered heteroaryl optionally substituted with C1- C4 alkyl or oxo. [0018] In some aspects, R2 is substituted with one R3. [0019] In some aspects, R1 is an optionally substituted pyrazolyl. [0020] In some aspects, R1 is selected from:
Figure imgf000005_0001
, , nd [0021] R10 is selected from H, C1-C4alkyl, C1-C4alkoxy, aminoC1-C4alkyl, hydroxyC1- C4alkyl, and C1-C4alkylsulfonyl, wherein the C1-C4alkyl, C1-C4alkoxy, aminoC1-C4alkyl, hydroxyC1-C4alkyl, and C1-C4alkylsulfonyl can be optionally substituted by one or more substituents selected from hydroxyl, C1-C4alkoxy, NR10aR10b, halo, and deuterium, wherein R10a and R10b are selected from hydrogen and C1-C4alkyl, or wherein R10a and R10b taken together with the nitrogen atom to which they are attached form a 4- to 8-membered ring. [0022] In some aspects, R1 is
Figure imgf000006_0001
. [0023] In some aspects, R10 is CH3. [0024] In some aspects, R2 is heterocyclyl. [0025] In some aspects, R2 is selected from: ,
Figure imgf000006_0002
, , , ,
, ,
Figure imgf000007_0001
, , , ,
, and
Figure imgf000008_0002
; [0026]
Figure imgf000008_0001
indicates a single bond or a double bond such that all valences are satisfied; [0027] m is selected from 0, 1, 2, 3, 4, 5, and 6; and [0028] Z1, Z2, and Z3 are selected from N and CRa. [0029] In some aspects, R2 is selected from:
,
Figure imgf000009_0001
, , , . [0030] In some aspects, the compound, or a pharmaceutically acceptable salt or solvate thereof, has Formula Ia:
Figure imgf000010_0001
[0031] wherein a and b are each independently selected from 1, 2, and 3; and [0032] Q is selected from -CH- and -N-, with the proviso that if Q is -N-, a and b are not 1. [0033] In some aspects, the compound, or a pharmaceutically acceptable salt or solvate thereof, ihas Formula II:
Figure imgf000010_0002
. [0034] In some aspects, the compound, or a pharmaceutically acceptable salt or solvate thereof, has Formula IIa:
Figure imgf000011_0001
. [0035] In some aspects, R3 is heteroaryl optionally substituted with one, two, three, four, or five R30. [0036] In some aspects, R3 is selected from: ,
Figure imgf000011_0002
, , , , , , ,
Figure imgf000012_0001
, [0037] wherein A1 is selected from O, S, and NR37, R36 is selected from hydrogen, optionally substituted C1-C6alkyl, and optionally substituted C1-C6alkylaryl, and R37 is selected from hydrogen and C1-C6alkyl. [0038] In some aspects, R3 is selected from:
Figure imgf000013_0001
Figure imgf000014_0001
, , [0039] In some aspects, R30 is:
Figure imgf000014_0002
. [0040] In some aspects, R300 is selected from:
Figure imgf000015_0001
[0041] In some aspects, R300 is:
Figure imgf000015_0002
. [0042] In some aspects, R301 is H and R302 is -OH or CH3. [0043] In some aspects, R3 is selected from: nd
Figure imgf000016_0001
. [0044] In some aspects, R3 is selected from:
Figure imgf000016_0002
, , . [0045] In some aspects, the compound, or a pharmaceutically acceptable salt or solvate thereof, has Formula IV:
Figure imgf000016_0003
wherein L1 is NH or O. [0046] In some aspects, the compound, or a pharmaceutically acceptable salt or solvate thereof, has Formula V:
Figure imgf000017_0001
. [0047] In some aspects, R31 is selected from: ,
Figure imgf000017_0002
, , ,
,
Figure imgf000018_0001
, , ,
Figure imgf000019_0001
,
Figure imgf000020_0001
, [0048] wherein R31a is selected from H, D, alkylamino, optionally substituted C1-C4alkyl, C1-C4alkoxy, amino, and C1-C4haloalkyl, [0049] each R31b is independently selected from H, D, halo, hydroxy, amino, cyano, alkylamino, optionally substituted C1-C4alkyl, C1-C4haloalkyl, optionally substituted C1- C4alkoxy, and C3-C6cycloalkyl, and q is 1, 2, or 3. [0050] In some aspects, the compound of Formula I, or a pharmaceutically acceptable salt or solvate thereof, is selected from any one of the compounds of Table 1. [0051] In some aspects, the compound of Formula I can exhibit one or more of the following properties: (i) promote the differentiation of an OPC into an oligodendrocyte, (ii) promote the expression of a protein associated with oligodendrocyte differentiation and/or myelination (e.g., G-protein coupled receptor 17 (GPR17), myelin basic protein (MBP), ASPA, GST-pi, CC1, myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte- specific protein/claudin-11, CNPase, proteolipid protein 1 (PLP1), or a combination thereof), (iii) promote the myelination of an axon, (iv) promote the remyelination of a demyelinated axon, (v) inhibit PDGFRα kinase activity, (vi) achieve a brain to plasma ratio of greater than 0.1 when systemically administered to a subject, and (vii) any combination thereof. [0052] In some aspects, the compound of Formula I can inhibit PDGFRα kinase activity. [0053] In some aspects, the compound of Formula I can inhibit PDGFRα kinase activity with an IC50 of less than 500 nM, less than 400 nM, less than 300 nM, less than 200 nM, less than 100 nM, less than 75 nM, less than 50 nM, less than 40 nM, less than 30 nM, less than 20 nM, less than 10 nM, less than 5 nM, less than 2.5 nM, less than 1 nM, less than 0.5 nM, or less than 0.2 nM. [0054] In some aspects, the IC50 of the PDGFRα inhibitor of Formula I is determined using an enzymatic PDGFRα kinase assay (e.g., Promega kinase assay described in Example 131). [0055] In some aspects, the enzymatic PDGFRα kinase assay comprises 20 ng of purified PDGFRα protein, 150 µM of ATP and 1 µg of substrate, Poly (Glu4Tyr1) in a volume of 15 µl. [0056] In some aspects, the compound of Formula I can inhibit PDGFRα kinase activity with an IC50 of from about 500 nM to about 0.001 nM, from about 400 nM to about 0.001 nM, from about 300 nM to about 0.001 nM, from about 200 nM to about 0.001 nM, from about 100 nM to about 0.001 nM, from about 75 nM to about 0.001 nM, from about 50 nM to about 0.001 nM, from about 40 nM to about 0.001 nM, from about 30 nM to about 0.001 nM, from about 20 nM to about 0.001 nM, from about 10 nM to about 0.001 nM, from about 5 nM to about 0.001 nM, from about 2.5 nM to about 0.001 nM, from about 1 nM to about 0.001 nM, from about 0.5 nM to about 0.001 nM, or from about 0.2 nM to about 0.001 nM. [0057] In some aspects, the disclosure provides a compound that can inhibit a PDGFRα activity of a cell and can further exhibit one or more of the following properties: (i) promote the differentiation of an OPC into an oligodendrocyte, (ii) promote the expression of a protein associated with oligodendrocyte differentiation and/or myelination (e.g., G-protein coupled receptor 17 (GPR17), myelin basic protein (MBP), ASPA, GST-pi, CC1, myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, proteolipid protein 1 (PLP1), or a combination thereof), (iii) promote the myelination of an axon, (iv) promote the remyelination of a demyelinated axon, (v) inhibit PDGFRα kinase activity, (vii) achieve a brain to plasma ratio of greater than 0.1 when systemically administered to a subject, or (viii) any combination thereof. [0058] In some aspects, the compound can inhibit PDGFRα kinase activity with an IC50 of less than 500 nM, less than 400 nM, less than 300 nM, less than 200 nM, less than 100 nM, less than 75 nM, less than less than 50 nM, less than 40 nM, less than 30 nM, less than 20 nM, less than 10 nM, less than 5 nM, or less than 2 nM. [0059] In some aspects, the IC50 of the PDGFRα inhibitor is determined using an enzymatic PDGFRα kinase assay (e.g., Promega™ kinase assay described in Example 131). [0060] In some aspects, the enzymatic PDGFRα kinase assay comprises 20 ng of purified PDGFRα protein, 150 µM of ATP and 1 µg of substrate, Poly (Glu4Tyr1) in a volume of 15 µl. [0061] The disclosure also provides a pharmaceutical composition comprising a compound disclosed herein, or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable excipient. [0062] The disclosure also provides a kit comprising a compound disclosed herein, or a pharmaceutically acceptable salt or solvate thereof, or a composition disclosed herein, and instructions for use. [0063] The disclosure also provides a method of producing a PDGFRα inhibitor comprising synthesizing a compound disclosed herein. [0064] The disclosure also provides a compound disclosed herein, or a pharmaceutical composition disclosed herein, for use as a medicament. [0065] The disclosure also provides a compound disclosed herein, or a pharmaceutical composition disclosed herein, for use in therapy. [0066] The disclosure also provides a method of treating a demyelinating disease in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound disclosed herein or a pharmaceutical composition disclosed herein. [0067] The disclosure also provides a method of improving a subject’s performance in a test for assessing one or more symptoms associated with a demyelinating disease, comprising administering to the subject a therapeutically effective amount of a compound disclosed herein or a pharmaceutical composition disclosed herein, wherein after the administration, the subject’s performance in the test is improved as compared to a reference subject (e.g., the subject prior to the administration). [0068] In some aspects, the test is selected from one or more of a visual evoked potential (VEP) test, a multifocal visual evoked potential (mfVEP) test, a low contrast visual acuity (LC-VA) test, a magnetic resonance imaging (MRI) (e.g., magnetization transfer ratio (MTR), myelin water fraction (MWF), quantitative susceptibility mapping (QSM), and T2 imaging), an electromyography (EMG), a nerve conduction velocity (NCV) test, an Extended Disability Status Scale (EDSS), a timed walk test (e.g., timed 25-foot walk), a Nine-Hole Peg Test (9HPT), an ocular coherence tomograph (OCT), a quality of life measure test (e.g., Multiple Sclerosis Quality of Life-54 and Vision-Related Quality of Life), cognitive assessment (e.g., Symbol-Digit Modalities Test or Montreal Cognitive Assessment), or combinations thereof. [0069] In some aspects, the demyelinating disease comprises one or more of an acute disseminated encephalomyelitis (ADEM), acute hemorrhagic leukoencephalitis, acute transverse myelitis, adrenoleukodystrophy, adrenomyeloneuropathy, Alexander disease, Alzheimer’s disease, aminoacidurias, amyotrophic lateral sclerosis, anti-MAG peripheral neuropathy, anti-MOG associated spectrum, Balo concentric sclerosis, brain injury, CAMFAK syndrome, Canavan disease, carbon monoxide toxicity, central pontine myelinolysis, cerebral hypoxia, cerebral ischemia, Charcot–Marie–Tooth disease, chronic inflammatory demyelinating polyneuropathy, chronic traumatic encephalopathy, clinically isolated syndrome (CIS), congenital cataract, copper deficiency associated condition, delayed post-hypoxic leukoencephalopathy, diffuse cerebral sclerosis of Schilder, diffuse myelinoclastic sclerosis, extrapontine myelinolysis Gaucher disease, Guillain–Barré syndrome, hereditary neuropathy, hereditary neuropathy with liability to pressure palsy, HTLV-1–associated myelopathy, Hurler syndrome, hypomyelination, hypoxic brain injury, Krabbe disease, Leber hereditary optic atrophy and related mitochondrial disorders, leukodystrophic disorders, Marchiafava-Bignami disease, metachromatic leukodystrophy, multiple sclerosis (e.g., primary progressive multiple sclerosis (PPMS), relapsing-remitting multiple sclerosis (RRMS), secondary progressive multiple sclerosis (SPMS), progressive relapsing multiple sclerosis, Marburg multiple sclerosis, tumefactive multiple sclerosis, and optic-spinal multiple sclerosis), multiple system atrophy, myelinoclastic disorders, myelopathy, nerve injury, neuromyelitis optica (NMO), Niemann-Pick disease, optic neuropathy, optic neuritis (e.g., acute optic neuritis and chronic relapsing inflammatory optic neuritis (CRION)), osmotic demyelination syndrome, Parkinson’s disease, Pelizaeus‐ Merzbacher disease, peripheral neuropathy, phenylketonuria, progressive inflammatory neuropathy, progressive multifocal leukoencephalopathy, progressive subcortical ischemic demyelination, reperfusion injury, Schilder disease, solitary sclerosis, spinal cord injury, subacute sclerosing panencephalitis, Tabes dorsalis, Tay-Sachs disease, transverse myelitis, traumatic brain injury, tropical spastic paraparesis, vitamin B12 deficiency, and cerebral palsy. [0070] In some aspects, the demyelinating disease is characterized by demyelination of one or more cells within the CNS of the subject. [0071] In some aspects, the demyelinating disease is multiple sclerosis. [0072] In some aspects, the multiple sclerosis comprises a clinically isolated syndrome (“CIS”), relapsing-remitting MS (“RRMS”), secondary progressive MS (“SPMS”), primary progressive MS (“PPMS”), optic neuritis or transverse myelitis. [0073] In some aspects, the demyelinating disease is an optic neuritis. [0074] In some aspects, treating the demyelinating disease comprises reducing one or more symptoms associated with the demyelinating disease. [0075] In some aspects, the one or more symptoms comprise one or more of fatigue, dizziness, malaise, elevated fever and high body temperature, extreme sensitivity to cold in the hands and feet, weakness and stiffness in muscles and joints, weight changes, digestive or gastrointestinal problems, low blood pressure, high blood pressure, irritability, anxiety, depression, impaired vision (e.g., blurred vision, double vision, reduction in low contrast visual acuity (LC-VA)), ataxia, clonus, spasms, dysarthria, weakness, clumsiness, hand paralysis, hemiparesis, genital anesthesia, sexual dysfunction, incoordination, paresthesias, ocular paralysis, impaired muscle coordination, loss of sensation, tingling, numbness, pain, neurological symptoms, impaired cognition, unsteady gait, balance problems, dizziness, spastic paraparesis, incontinence, hearing problems, speech problems, loss of olfaction, and agusia. [0076] The disclosure also provides a method of promoting the myelination of an axon in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound disclosed herein or a pharmaceutical composition disclosed herein. [0077] In some aspects, promoting the myelination of an axon results in an increase in the expression of one or more of the following markers within the subject: myelin basic protein (MBP) Myelin Oligodendrocyte Glycoprotein (MOG), Oligodendrocyte Specific Protein/Claudin-11, CNPase, proteolipid protein 1 (PLP1), or any combination thereof. [0078] In some aspects, the myelination of an axon can be determined by visualizing and/or quantifying the expression of one or more of the following markers: myelin basic protein (MBP) Myelin Oligodendrocyte Glycoprotein (MOG), Oligodendrocyte Specific Protein/Claudin-11, CNPase, proteolipid protein 1 (PLP1), or any combination thereof. [0079] The disclosure also provides method of promoting the remyelination of a demyelinated axon in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound disclosed herein or a pharmaceutical composition disclosed herein. [0080] In some aspects, promoting the remyelination of a demyelinated axon results in an increase in the expression of one or more of the following markers within the subject: myelin basic protein (MBP) Myelin Oligodendrocyte Glycoprotein (MOG), Oligodendrocyte Specific Protein/Claudin-11, CNPase, proteolipid protein 1 (PLP1), or any combination thereof. [0081] In some aspects, the remyelination of a demyelinated axon can be determined by visualizing and/or quantifying the expression of one or more of the following markers: myelin basic protein (MBP), Myelin Oligodendrocyte Glycoprotein (MOG), Oligodendrocyte Specific Protein/Claudin-11, CNPase, proteolipid protein 1 (PLP1), or any combination thereof. [0082] The disclosure also provides a method of reducing the demyelination of a myelinated neuronal axon in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound disclosed herein or a pharmaceutical composition disclosed herein. [0083] In some aspects, reducing the demyelination of a myelinated neuronal axon results in an increase in the expression of one or more of the following markers: myelin basic protein (MBP) Myelin Oligodendrocyte Glycoprotein (MOG), Oligodendrocyte Specific Protein/Claudin-11, CNPase, proteolipid protein 1 (PLP1), or any combination thereof. [0084] In some aspects, the reduction in the demyelination of a myelinated neuronal axon can be determined by visualizing and/or quantifying the expression of one or more of the following markers: myelin basic protein (MBP) Myelin Oligodendrocyte Glycoprotein (MOG), Oligodendrocyte Specific Protein/Claudin-11, CNPase, proteolipid protein 1 (PLP1), or any combination thereof. [0085] The present disclosure also provides a method of activating an oligodendrocyte progenitor cell (OPC) within the central nervous system (CNS) of a subject in need thereof, the method comprising administering to the subject an effective amount of a compound disclosed herein or a pharmaceutical composition disclosed herein. [0086] In some aspects, the subject has, or is at risk of developing a demyelinating disease, for example a disease disclosed herein. [0087] In some aspects, the method is a method of treating or preventing a demyelinating disease, for example a disease disclosed herein. [0088] In some aspects, the compound or the pharmaceutical composition is administered to the subject once. [0089] In some aspects, the compound or the pharmaceutical composition is administered to the subject more than once using intermittent dosing. [0090] In some aspects, the intermittent dosing comprises administering the compound or pharmaceutical composition to the subject every other day, every three days, every four days, every five days, every six days, once a week, every eight days, every nine days, every 10 days, every 11 days, every 12 days, every 13 days, once every two weeks, once every three weeks, once a month, once every two months, once every three months, once every four months, once every five months, once every six months, or once every twelve months. [0091] In some aspects, the intermittent dosing comprises administering to the subject a first dose and a second dose of the compound or pharmaceutical composition, wherein the second dose is administered at least one day, at least two days, at least three days, at least four days, at least five days, at least six days, at least seven days, at least eight days, at least nine days, at least 10 days, at least 11 days, at least 12 days, at least 13 days, at least two weeks, at least three weeks, at least one month, at least two months, at least three months, at least four months, at least five months, at least six months, or at least 12 months after administering the first dose. [0092] In some aspects, the second dose is administered to the subject one day, two days, three days, four days, five days, six days, seven days, eight days, nine days, 10 days, 11 days, 12 days, 13 days, two weeks, three weeks, one month, two months, three months, four months, five months, six months, or 12 months after administering the first dose. [0093] In some aspects, after the administration, the compound or the pharmaceutical composition can achieve a brain to plasma ratio of greater than 0.1, greater than 0.2, greater than 0.3, greater than 0.4, greater than 0.5, greater than 0.6, greater than 0.7, greater than 0.8, greater than 0.9, greater than 1.0, greater than 1.1, greater than 1.2, greater than 1.3, greater than 1.4, greater than 1.5, greater than 1.6, greater than 1.7, greater than 1.8, greater than 1.9, or greater than 2.0. In some aspects, the brain to plasma ratio may be assessed in preclinical species using bioanalytical gas or liquid chromatography and mass spectrometry methods. [0094] In some aspects, the method further comprises administering to the subject an additional therapeutic agent. [0095] In some aspects, the additional therapeutic agent comprises a standard care of treatment. [0096] In some aspects, the additional therapeutic agent comprises an immunomodulatory agent. [0097] In some aspects, the additional therapeutic agent is selected from interferon beta- 1b, interferon beta-1a, peginterferon beta-1a, alemtuzumab, natalizumab, ocrelizumab, ofatumumab, ublituximab-xiiy, glatiramer acetate, teriflunomide, dimethyl fumarate, monomethyl fumarate, diroximel fumarate, fingolimod hydrochloride, siponimod fumaric acid, ozanimod hydrochloride, ponesimod, cladribine, mitoxantrone, a BTK inhibitor (e.g., masitinib, evobrutinib, or tolebrutinib), a statin (e.g., simvastatin), or a pharmaceutically acceptable salt thereof. [0098] In some aspects, the additional therapeutic agent is administered to the subject prior to, concurrently, or after the administration of the compound or the pharmaceutical composition. [0099] The disclosure also provides a method of inducing the differentiation of an oligodendrocyte progenitor cell (OPC) into an oligodendrocyte, the method comprising contacting the OPC with an effective amount of a compound disclosed herein or a pharmaceutical composition disclosed herein. [0100] In some aspects, inducing the differentiation of the OPC into an oligodendrocyte results in an increase in the expression of the following markers in the subject: GPR17, MBP, ASPA, GST-pi, CC1, myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, proteolipid protein 1 (PLP1), or a combination thereof. [0101] In some aspects, the differentiation of the OPC into an oligodendrocyte is measured by determining the expression of GPR17, MBP, ASPA, GST-pi, CC1, myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, proteolipid protein 1 (PLP1), or a combination thereof. [0102] The disclosure also provides a method of inhibiting PDGFRα activity in a cell, the method comprising contacting the cell with an effective amount of a compound disclosed herein or a pharmaceutical composition disclosed herein. [0103] In some aspects, the inhibition of the PDGFRα activity is measured by one or more of the following: an in vitro OPC differentiation assay (e.g., as described in Example 132), a cuprizone model for demyelination, an in vivo OPC differentiation assay (e.g., as described in Example 134), an enzymatic PDGFRα kinase assay (e.g., as described in Example 131), or any combination thereof. [0104] In some aspects, the contacting occurs ex vivo or in vivo. [0105] In some aspects, the method is a method of treating by a therapy. [0106] The disclosure also provides a method of treating a PDGF-associated tumor in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound disclosed herein or a pharmaceutical composition disclosed herein, wherein after the administration, PDGFRα activity is reduced in the subject. [0107] In some aspects, the PDGF-associated tumor comprises an oligodendroglioma. [0108] In some aspects, the method is a method of treatment by therapy. DETAILED DESCRIPTION OF THE DISCLOSURE [0109] The present disclosure describes compounds that can induce the differentiation of OPCs into cells that have features of mature oligodendrocytes, including morphological characteristics and the protein expression patterns associated with myelination, and can also affect remyelination. Accordingly, as described herein, the compounds of the present disclosure can be useful in treating various diseases, such as those associated with demyelination. Additional aspects of the present disclosure are provided throughout the present application. [0110] Before the present disclosure is described in greater detail, it is to be understood that this disclosure is not limited to the particular compositions or process steps described, which can, of course, vary. As will be apparent to those of skill in the art upon reading this disclosure, each of the individual aspects described and illustrated herein has discrete components and features, which can be readily separated from, or combined with, the features of any of the other several aspects without departing from the scope or spirit of the present disclosure. Any recited method can be carried out in the order of events recited or in any other order which is logically possible. [0111] The headings provided herein are not limitations of the various aspects of the disclosure, which can be defined by reference to the specification as a whole. It is also to be understood that the terminology used herein is for the purpose of describing particular aspects only, and is not intended to be limiting, since the scope of the present disclosure will be limited only by the appended claims. I. Terms [0112] In order that the present disclosure can be more readily understood, certain terms are first defined. As used in this application, except as otherwise expressly provided herein, each of the following terms shall have the meaning set forth below. Additional definitions are set forth throughout the application. [0113] The term “a” or “an” entity refers to one or more of that entity; for example, “a nucleotide sequence,” is understood to represent one or more nucleotide sequences. As such, the terms “a” (or “an”), “one or more,” and “at least one” can be used interchangeably herein. [0114] Furthermore, “and/or” where used herein is to be taken as specific disclosure of each of the two specified features or components with or without the other. Thus, 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). [0115] It is understood that wherever aspects are described herein with the language “comprising,” otherwise analogous aspects described in terms of “consisting of” and/or “consisting essentially of” are also provided. [0116] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure is related. For example, the Concise Dictionary of Biomedicine and Molecular Biology, Juo, Pei-Show, 2nd ed., 2002, CRC Press; The Dictionary of Cell and Molecular Biology, 5th ed., 2013, Academic Press; and the Oxford Dictionary of Biochemistry and Molecular Biology, 2nd ed., 2008, Oxford University Press, provide one of skill with a general dictionary of many of the terms used in this disclosure. [0117] Units, prefixes, and symbols are denoted in their Système International de Unites (SI) accepted form. Numeric ranges are inclusive of the numbers defining the range. Where a range of values is recited, it is to be understood that each intervening integer value, and each fraction thereof, between the recited upper and lower limits of that range is also specifically disclosed, along with each subrange between such values. The upper and lower limits of any range can independently be included in or excluded from the range, and each range where either, neither or both limits are included is also encompassed within the disclosure. Thus, ranges recited herein are understood to be shorthand for all of the values within the range, inclusive of the recited endpoints. For example, a range of 1 to 10 is understood to include any number, combination of numbers, or sub-range from the group consisting of 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10. [0118] Where a value is explicitly recited (e.g., 10), it is to be understood that values that are about the same quantity or amount as the recited value (e.g., ± 10%) are also within the scope of the disclosure. Where a combination is disclosed, each subcombination of the elements of that combination is also specifically disclosed and is within the scope of the disclosure. Conversely, where different elements or groups of elements are individually disclosed, combinations thereof are also disclosed. Where any element of a disclosure is disclosed as having a plurality of alternatives, examples of that disclosure in which each alternative is excluded singly or in any combination with the other alternatives are also hereby disclosed; more than one element of a disclosure can have such exclusions, and all combinations of elements having such exclusions are hereby disclosed. [0119] The term “halo” as used herein by itself or as part of another group refers to Cl, F, Br, or I. [0120] The term “nitro” as used herein by itself or as part of another group refers to NO2. [0121] The term “cyano” as used herein by itself or as part of another group refers to CN. [0122] The term “hydroxy” as herein used by itself or as part of another group refers to OH. [0123] The term “alkyl” as used herein by itself or as part of another group refers to a straight- or branched-chain aliphatic hydrocarbon containing one to twelve carbon atoms, i.e., a C1-C12alkyl, or the number of carbon atoms designated, e.g., a C1alkyl such as methyl, a C2alkyl such as ethyl, etc. In some aspects, the alkyl is a C1-C10alkyl. In some aspects, the alkyl is a C1-C6alkyl. In some aspects, the alkyl is a C1-C4alkyl. In some aspects, the alkyl is a C1-C3alkyl, i.e., methyl, ethyl, propyl, or isopropyl. Non limiting exemplary C1- C12 alkyl groups include methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, tert-butyl, iso- butyl, 3-pentyl, hexyl, heptyl, octyl, nonyl, and decyl. In some aspects, one or more of the hydrogen atoms of the alkyl group are replaced by deuterium atoms, i.e., the alkyl group is isotopically-labeled with deuterium. A non-limiting exemplary deuterated alkyl group is - CD3. [0124] The term “haloalkyl” as used herein by itself or as part of another group refers to an alkyl group wherein one or more hydrogen atoms of the alkyl group are replaced by halo atoms. In some aspects, the haloalkyl group is a -CF3 group. [0125] The term “alkoxy” as used herein by itself or as part of another group refers to an alkyl group attached to a terminal oxygen atom. In some aspects, the alkyl is a C1-C8alkyl and resulting alkoxy is thus referred to as a “C1-C8alkoxy.” In some aspects, the alkyl is a C1-C4alkyl group. Non-limiting exemplary alkoxy groups include methoxy, ethoxy, and tert-butoxy. [0126] The term “alkyoxyalkyl” as used herein by itself or as part of another group refers to an alkyl group substituted with an alkoxyl group. In some aspects, the alkyl is a C1-C4 alkyl and the alkoxy is a C1-C4 alkoxyl and the resulting alkoxyalkyl is thus referred to as a “C1-C4alkoxyC1-C4alkyl”. [0127] The term “amino” as used herein by itself or as part of another group refers to -NH2, which may be optionally substituted with one or two alkyl, two alkyl linked to form a ring, haloalkyl, (hydroxy)alkyl, (alkoxy)alkyl, (amino)alkyl, heteroalkyl, optionally substituted cycloalkyl, optionally substituted heterocyclo, optionally substituted aryl, optionally substituted heteroaryl, (aryl)alkyl, (cycloalkyl)alkyl, (heterocyclo)alkyl, or (heteroaryl)alkyl. [0128] The term “heterocycylamino” as used herein by itself or as part of another group refers to an amino group substituted with a heterocyclyl group. [0129] The term “aminoalkyl” as used herein by itself or as part of another group refers to an alkyl group substituted with an amino group. In some aspects, the alkyl is a C1-C4alkyl and the resulting aminoalkyl is thus referred to as an “aminoC1-C4alkyl”. [0130] The term “hydroxyalkyl” as used herein by itself or as part of another group refers to an alkyl group substituted with a hydroxy group. In some aspects, the alkyl is a C1- C4alkyl and the resulting hydroxyalkyl is thus referred to as a “hydroxyC1-C4alkyl”. [0131] The term “aminoalkylamino” as used herein by itself or as part of another group refers to an amino group substituted with an aminoalkyl group. In some aspects, the aminoalkylamino group is -NHCH2CH2NH2. [0132] The term “oxo” as used herein refers to an oxygen atom that is connected to a carbon atom by a double bond, i.e. to form a keto group. [0133] The term “cycloalkyl” as used herein by itself or as part of another group refers to saturated and partially unsaturated, e.g., containing one or two double bonds, monocyclic, bicyclic, or tricyclic aliphatic hydrocarbons containing three to twelve carbon atoms, i.e., a C3-C12cycloalkyl, or the number of carbons designated, e.g., a C3cycloalkyl such a cyclopropyl, a C4cycloalkyl such as cyclobutyl, etc. In some aspects, the cycloalkyl is bicyclic, i.e., it has two rings. In some aspects, the cycloalkyl is monocyclic, i.e., it has one ring. In some aspects, the cycloalkyl is a C3-C8cycloalkyl. In some aspects, the cycloalkyl is a C3-6cycloalkyl, i.e., cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl. In some aspects, the cycloalkyl is a C5cycloalkyl, i.e., cyclopentyl or cyclopentenyl. In some aspects, the cycloalkyl is a C6cycloalkyl, i.e., cyclohexyl or cyclohexenyl. A cycloalkyl group containing one or two double bonds may also be referred to as a “cycloalkenyl” group. [0134] The term “heterocyclyl” as used herein by itself or as part of another group refers to saturated and partially unsaturated, e.g., containing one or two double bonds, monocyclic, bicyclic, or tricyclic groups containing three to fourteen ring members, i.e., a 3- to 14-membered heterocyclyl, comprising one, two, three, or four heteroatoms. Each heteroatom is independently oxygen, sulfur, or nitrogen. The term heterocyclyl includes groups wherein one or more -CH2- groups is replaced with one or more -C(=O)- groups, including cyclic ureido groups such as imidazolidinyl-2-one, cyclic amide groups such as piperidin-2-one or piperazin-2-one, and cyclic carbamate groups such as oxazolidinyl-2- one. [0135] The term “heterocyclylaryl” as used herein by itself or as part of another group refers to an aryl group that is substituted with a heterocyclyl group. [0136] The term “arylC1-C4alkyl” as used herein by itself or as part of another group refers to a C1-C4alkyl group substituted with an aryl group. [0137] The term “heteroarylC1-C4alkyl” as used herein by itself or as part of another group refers to a C1-C4alkyl group substituted with a heteroaryl group. [0138] The term “heterocyclylC1-C4alkyl” as used herein by itself or as part of another group refers to a C1-C4alkyl group substituted with a heterocyclyl group. [0139] The term “cycloalkylC1-C4alkyl” as used herein by itself or as part of another group refers to a C1-C4alkyl group substituted with a cycloalkyl group. [0140] The term “alkylsulfonyl” as used herein by itself or as part of another group refers to a sulfonyl group, i.e., -SO2-, substituted by an alkyl group. A non-limiting exemplary alkylsulfonyl group is -SO2CH3. [0141] The term “aryl” as used herein by itself or as part of another group refers to an aromatic ring system having six to fourteen carbon atoms, i.e., C6-C14 aryl. Non-limiting exemplary aryl groups include phenyl (abbreviated as “Ph”), naphthyl, phenanthryl, anthracyl, indenyl, azulenyl, biphenyl, biphenylenyl, and fluorenyl groups. In some aspects, the aryl group is phenyl. [0142] The term “heteroaryl” as used herein by itself or as part of another group refers to monocyclic and bicyclic aromatic ring systems having five to fourteen ring members, i.e., a 5- to 14-membered heteroaryl, comprising one, two, three, or four heteroatoms. Each heteroatom is independently oxygen, sulfur, or nitrogen. In some aspects, the heteroaryl has three heteroatoms. In some aspects, the heteroaryl has two heteroatoms. In some aspects, the heteroaryl has one heteroatom. In some aspects, the heteroaryl is a 5- to 10- membered heteroaryl. In some aspects, the heteroaryl has 5 ring atoms, e.g., thienyl, a 5- membered heteroaryl having four carbon atoms and one sulfur atom. In some aspects, the heteroaryl has 6 ring atoms, e.g., pyridyl, a 6-membered heteroaryl having five carbon atoms and one nitrogen atom. Non-limiting exemplary heteroaryl groups include thienyl, benzo[b]thienyl, naphtho[2,3-b]thienyl, thianthrenyl, furyl, benzofuryl, pyranyl, isobenzofuranyl, benzoxazolyl, chromenyl, xanthenyl, 2H-pyrrolyl, pyrrolyl, imidazolyl, pyrazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, isoindolyl, 3H-indolyl, indolyl, indazolyl, purinyl, isoquinolyl, quinolyl, phthalazinyl, naphthyridinyl, cinnolinyl, quinazolinyl, pteridinyl, 4aH-carbazolyl, carbazolyl, β-carbolinyl, phenanthridinyl, acridinyl, pyrimidinyl, phenanthrolinyl, phenazinyl, thiazolyl, isothiazolyl, phenothiazolyl, isoxazolyl, furazanyl, and phenoxazinyl. The term heteroaryl also includes N-oxides. A non-limiting exemplary N-oxide is pyridyl N-oxide. [0143] The term “aminoheterocyclyl” as used herein by itself or as part of another group refers to a heterocyclyl group substituted with an optionally substituted amino group. Non- limiting exemplary aminoheterocyclyl groups include:
Figure imgf000034_0001
. [0144] The chemical terms used herein may be combined to describe larger substituents. For a monovalent substituent, the substituent that is recited last in the combined term is the substituent that contains the attachment point. For example, an “arylC1-C4alkyl” group contains an attachment point on the alkyl group, whereas an “C1-C4alkylaryl” group contains an attachment point on the aryl group. [0145] The present disclosure encompasses any of the disclosed compounds being isotopically-labelled (i.e., radiolabeled) by having one or more atoms replaced by an atom having a different atomic mass or mass number. Examples of isotopes that can be incorporated into the disclosed compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine and chlorine, such as 1H, 2H (or deuterium (D)), 3H, 11C, 12C, 13C, 14C, 14N 15N, 18O, 17O, 16O 31P, 32P, 32S, 33S, 34S, 35S, 36S, 18F, 19F, 35Cl, 37Cl, and 36Cl, respectively, e.g., 3H, 11C, and 14C. In some aspects, provided is a composition wherein substantially all of the atoms at a position within the disclosed compound are replaced by an atom having a different atomic mass or mass number. In some aspects, provided is a composition wherein a portion of the atoms at a position within the disclosed compound are replaced, i.e., the disclosed compound is enriched at a position with an atom having a different atomic mass or mass number. Isotopically-labelled disclosed compounds can be prepared by methods known in the art. The present disclosure also encompasses any of the disclosed compounds wherein a quaternary carbon atom is replaced with a silicon atom. [0146] As noted above, the compounds disclosed herein contain one or more asymmetric carbon atoms and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms. The present disclosure encompasses the use of all such possible forms, as well as their racemic and resolved forms and mixtures thereof. The individual enantiomers can be separated according to methods known in the art in view of the present disclosure. When the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that they include both E and Z geometric isomers. All tautomers are also encompassed by the present disclosure. [0147] As used herein, the term “stereoisomers” is a general term for all isomers of individual molecules that differ only in the orientation of their atoms in space. It includes enantiomers and isomers of compounds with more than one chiral center that are not mirror images of one another (diastereomers). [0148] The term “chiral center” or “asymmetric carbon atom” refers to a carbon atom to which four different groups are attached. [0149] The terms “enantiomer” and “enantiomeric” refer to a molecule that cannot be superimposed on its mirror image and hence is optically active wherein the enantiomer rotates the plane of polarized light in one direction and its mirror image compound rotates the plane of polarized light in the opposite direction. [0150] The term “racemic” refers to a mixture of equal parts of enantiomers and which mixture is optically inactive. In some aspects, the compounds disclosed are racemic. [0151] The term “absolute configuration” refers to the spatial arrangement of the atoms of a chiral molecular entity (or group) and its stereochemical description, e.g., R or S. [0152] The stereochemical terms and conventions used in the specification are meant to be consistent with those described in Pure & Appl. Chem 68:2193 (1996), unless otherwise indicated. [0153] The term “enantiomeric excess” or “ee” refers to a measure for how much of one enantiomer is present compared to the other. For a mixture of R and S enantiomers, the percent enantiomeric excess is defined as │R – S│*100, where R and S are the respective mole or weight fractions of enantiomers in a mixture such that R + S = 1. With knowledge of the optical rotation of a chiral substance, the percent enantiomeric excess is defined as ([α]obs/[α]max)*100, where [α]obs is the optical rotation of the mixture of enantiomers and [α]max is the optical rotation of the pure enantiomer. Determination of enantiomeric excess is possible using a variety of analytical techniques, including NMR spectroscopy, chiral column chromatography or optical polarimetry. [0154] The terms “administration,” “administering,” and grammatical variants thereof, refer to introducing a composition, such as a PDGFRα inhibitor of the present disclosure, into a subject via a pharmaceutically acceptable route. Any suitable route of administration can be used in administering the PDGFRα inhibitors described herein to a subject. Non- limiting examples of such routes of administration are provided elsewhere in the present disclosure. [0155] The term “central nervous system” or “CNS” refers to a complex of nerve tissues that control the various activities of the body (e.g., voluntary and involuntary movements) and the mind (e.g., thoughts, perceptions, and emotions). The CNS generally consists of the brain and the spinal cord. [0156] As used herein, the term “demyelinating disease” refers to any disorder of the nervous system in which there is reduced myelination, including disorders in which insufficient or dysfunctional myelin (e.g., hypomyelination) is generated during development or disorders in which the myelin sheath of neurons is damaged. “Myelin” and “Myelin sheath” refer to the specialized membrane formed by oligodendrocytes that insulates the axons of neurons. The insulation provided by the myelin sheath helps to increase the rate of transmission of nerve signals along the axon, thereby promoting timely and energetically efficient neuronal signaling. Myelin also provides metabolic support of the axons, maintaining their health and survival. In some aspects, the neuronal axon of a subject suffering from or at risk of developing a demyelinating disease is completely demyelinated. In some aspects, the neuronal axon of a subject suffering from or at risk of developing a demyelinating disease is partially demyelinated (e.g., at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, or at least 95% compared to a reference neuronal axon that is fully myelinated). Accordingly, unless indicated otherwise, the term “demyelination” comprises any of the following: complete hypomyelination, complete demyelination, partial hypomyelination, partial demyelination, and combinations thereof. In some aspects, because of the partial and/or complete demyelination, the demyelinating diseases described herein are associated with impaired conduction of nerve signals or axonal or neuronal survival, which can, in turn, cause deficiencies in sensation, movement, cognition, or other functions depending on which neurons are affected. Non-limiting examples of demyelinating diseases are provided elsewhere in the present disclosure. In general, the term “under myelination” refers to a cell, tissue, or subject that lacks normal levels of myelination (e.g., level of myelination observed in a corresponding cell, tissue, or subject who does not have a demyelinating disease), regardless of etiology. [0157] As used herein, the term “hypomyelination” refers to a deficiency in myelin for any reason (e.g., body is unable to produce myelin at normal levels). Unless indicated otherwise, hypomyelination includes demyelination (related to myelin destruction) and dysmyelination (related to abnormal myelin deposition). Accordingly, hypomyelination includes diseases in which insufficient myelin is generated during development as well as diseases associated with demyelination and/or dysmyelination. In some aspects, compared to a reference (e.g., corresponding tissue, e.g., brain, of a subject who does not suffer from a demyelinating disease), there is hypomyelination of all (i.e., “complete hypomyelination”) or a portion of the nervous system (i.e., “partial hypomyelination”), for example the brain (e.g., hypomyelination in white matter and/or gray matter), of at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, or at least 95%. [0158] As used herein, the term “neuron” includes electrically excitable cells that process and transmit information through electrical and chemical signals. Neurons are major components of the brain and spinal cord of the CNS, and of the ganglia of the peripheral nervous system (PNS) and can connect to each other to form neural networks. A typical neuron is composed of a cell body (soma), dendrites, and an axon. The “soma” (the cell body) of a neuron contains the nucleus. The “dendrites” of a neuron are cellular extensions generally having many branches, where the majority of input to the neuron occurs. The “axon” (also referred to herein as “neuronal axon”) extends from the soma and carries nerve signals away from the soma and certain types of information back to the soma. [0159] As used herein, the term “oligodendrocyte progenitor cells” or “OPCs” (also known in the art as “oligodendrocyte precursor cells,” “polydendrocytes,” “NG2 cells,” and “O-2A cells”) refer to a subtype of glial cells in the central nervous system. They are precursors to “oligodendrocytes,” (also known as “oligodendroglia”) which are responsible for generating the myelin sheath that wraps around axons, providing insulation, aiding electrical conduction, and providing metabolic support. Nerve impulses can travel up to 200 times faster along a myelinated axon compared to an unmyelinated axon. OPCs and immature oligodendrocytes are generally positive for the following markers: A2B5, neuron-glial antigen 2 (NG2), and PDGFRα. Other suitable markers that can be used are known in the art. [0160] As used herein, the term “remyelination” (or derivatives thereof) refers to generation of new myelin sheaths around demyelinated (e.g., including hypomyelinated) axons. The remyelination process involves the differentiation of OPCs into oligodendrocytes that generate functional myelin sheaths around demyelinated axons. Remyelination of the axons can restore action potential conduction properties to axons, and thereby, promote and/or improve neurological function. Further, remyelination can provide metabolic support to axons, preventing their damage or loss. In the context of the present application, unless otherwise specified, “remyelination” refers to any aspect of a process that can result in remyelination. For example, in some aspects, “remyelination” comprises the migration or colonization of OPCs to sites of demyelinated axons. In some aspects, “remyelination” comprises the differentiation of OPCs into oligodendrocytes. In some aspects, “remyelination” comprises the generation of myelin sheaths by oligodendrocytes around demyelinated axons. In some aspects, the term “remyelination” comprises any combination of the following: (i) migration or colonization of OPCs to sites of demyelinated axons; (ii) differentiation of OPCs into oligodendrocytes; and (iii) generation of myelin sheaths by oligodendrocytes around demyelinated (e.g., including hypomyelinated) axons. [0161] As used herein, the term “restore” (and derivatives thereof) comprises both complete restoration and partial restoration. For example, in some aspects, remyelinating a demyelinated axon restores conduction properties to the axon, such that the conduction properties are the same as those of the axon prior to the demyelination (i.e., complete restoration). In some aspects, remyelinating a demyelinated axon restores conduction properties to the axon, where the conduction properties are improved but not the same as those of the axon prior to the demyelination (i.e., partial restoration). In some aspects, remyelination provides metabolic support to axons, preventing their damage or loss. In some aspects, remyelination provides partial metabolic support to axons, delaying their damage or loss. [0162] As used herein, the term “promoting” refers to the ability of an agent (e.g., PDGFRα inhibitor described herein) to induce or increase a particular result (e.g., remyelination of a demyelinated axon). In some aspects, the term comprises both inducing and increasing a particular result. [0163] As used herein, the term “subject” refers to any animal subject including a human, a laboratory animal (e.g., a non-human primate, rat, and mouse), livestock (e.g., cow, sheep, goat, pig, turkey, and chicken), and household pets (e.g., dog, cat, and rodent). [0164] The terms “treat,” “treating,” and “treatment,” as used herein, refer to any type of intervention or process performed on, or administering an active agent (e.g., PDGFRα inhibitor described herein) to, the subject with the objective of reversing, alleviating, ameliorating, inhibiting, or slowing down; or preventing the progression, development, severity or recurrence of a symptom, complication, condition or one or more biochemical indicia associated with a disease, or enhancing overall survival. As described herein, in some aspects, treatment can be of a subject having a disease (e.g., exhibiting one or more symptoms associated with the disease). In some aspects, treatment can be of a subject with some degree of demyelination but not yet exhibiting any symptoms associated with the disease. In the context of such subjects, administering a PDGFRα inhibitor of the present disclosure can help delay or prevent the onset of symptoms associated with the disease. [0165] The term “effective dose” or “effective amount” is defined as an amount sufficient to achieve or at least partially achieve a desired effect (e.g., induce remyelination of a demyelinated neuronal axon). A “therapeutically effective amount” or “therapeutically effective dosage” of a therapeutic agent (e.g., PDGFRα inhibitor) is any amount of the therapeutic agent that, when used alone or in combination with another therapeutic agent, promotes disease regression as evidenced by a decrease in severity of disease symptoms, an increase in frequency and duration of disease symptom-free periods, a prevention of impairment or disability due to the disease affliction, or a reduction in disease progression. A therapeutically effective amount or dosage of a therapeutic agent includes a “prophylactically effective amount” or a “prophylactically effective dosage,” which is any amount of the therapeutic agent that, when administered alone or in combination with another therapeutic agent to a subject at risk of developing a disease or of suffering a recurrence of disease, inhibits the development or recurrence of the disease. The ability of a therapeutic agent to promote disease reversal or inhibit the development, progression or recurrence of the disease can be evaluated using a variety of methods known to the skilled practitioner, such as in human subjects during clinical trials, in animal model systems predictive of efficacy in humans, or by assaying the activity of the agent in in vitro assays. [0166] As used herein, the term “dosing interval” refers to the amount of time that elapses between multiple (e.g., 2 or more) doses of a PDGFRα inhibitor described herein. Not to be bound by any one theory, in some aspects, a PDGFRα inhibitor provided herein exerts its therapeutic effect by inducing the differentiation of oligodendrocyte progenitor cells (OPCs) into oligodendrocytes. Accordingly, in some aspects, for a PDGFRα inhibitor to have a therapeutic effect in a subject (e.g., by inducing OPC differentiation), it is necessary that the subject has a pool of OPCs which a PDGFRα inhibitor described herein can act upon. As demonstrated herein, administering a PDGFRα inhibitor too frequently to a subject can be counterproductive in treating a demyelinating disease because the OPC population would not have sufficient time to repopulate the brain. Accordingly, in some aspects, a dosing interval that is suitable for the present disclosure is the amount of time required for a subject’s OPC population to have sufficiently recovered after the administration of an initial dose of a PDGFRα inhibitor provided herein, or a period longer than that, such that the administration of a second (or additional dose) of the PDGFRα inhibitor would have a therapeutic effect (e.g., such as those described herein, e.g., increases the number of differentiated oligodendrocytes) in the subject. As is apparent from the present disclosure, in some aspects, a PDGFRα inhibitor described herein is administered to a subject at a dosing interval, wherein the dosing interval is the amount of time required, after the administration of an initial dose of the PDGFRα inhibitor, for the subject’s OPC population (e.g., in size) to be at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 100% to that of a reference OPC population (e.g., the subject’s OPC population prior to the initial administration of the PDGFRα inhibitor). In some aspects, an OPC population may be measured by quantifying the density of NG2+ (neuron-glia antigen 2/chondroitin sulfate proteoglycan-4) or PDGFRa+ cells in mice, rats, pigs, dogs, non-human primates or other species using immunohistochemistry or fluorescence-activated cell sorting. Repopulation may be calculated as the OPC cell density at a given time after dosing relative to the OPC population in an untreated reference animal. [0167] In some aspects, a suitable dosing interval for the present disclosure comprises the amount of time required, after an initial dose of the PDGFRα inhibitor, for the plasma level of the PDGFRα inhibitor to reach less than 50%, less than 40%, less than 30%, less than 20%, less than 10%, or less than 5%, as compared to a reference (e.g., plasma level of the PDGFRα inhibitor in a corresponding subject immediately after, e.g., about 4 hours after, the administration of the initial dose of the PDGFRα inhibitor), a suitable dosing interval is the time between administering the first dose of the PDGFRα inhibitor and the subject’s plasma level of the PDGFRα inhibitor reaching a reduced level as compared to the reference. In some aspects, the additional dose of the PDGFRα inhibitor is administered to the subject when the subject’s plasma level has decreased by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or about 100% as compared to the reference. In some aspects, the brain to plasma ratio may be assessed in preclinical species using bioanalytical gas or liquid chromatography and mass spectrometry methods. [0168] As used herein, the terms “binds,” “recognizes,” “targets” are analogous terms and refer to molecules (e.g., PDGFRα inhibitors described herein) that can bind and/or target a particular region of a protein (e.g., PDGFRα), as such binding and/or targeting is understood by one skilled in the art. For instance, as described herein, in some aspects, a PDGFRα inhibitor is a small molecule that can target and inhibit the kinase portion of PDGFRα (e.g., by binding to the ATP binding site, by allosterically interfering with PDGFRα kinase activity, or both), and thereby, inhibit its activity. Unless indicated otherwise, the above terms (i.e., binds, recognizes, and targets) are used interchangeably and comprises any binding or targeting of PDGFRα, such that its activity is reduced and/or inhibited (e.g., binding of a small molecule to an ATP binding site of PDGFRα, binding of a small molecule to a substrate binding site of PDGFRα, allosterically interfering with PDGFRα kinase activity, or a combination thereof). [0169] “Potency” is an expression of the activity of a therapeutic agent in terms of the amount or concentration of the therapeutic agent that achieves a desired effect. Accordingly, functional assays, such as those described herein, can be used to identify compounds useful for the present disclosure. [0170] Molecules (e.g., PDGFRα inhibitors, such as those described herein) that “compete with another protein or compound for binding to a target” refers to molecules that inhibit (partially or completely) the binding of the other protein (e.g., naturally existing PDGFRα ligand) to the target (e.g., PDGFRα). Whether two compounds compete with each other for binding to a target, i.e., whether and to what extent a PDGFRα inhibitor described herein inhibits the binding of the naturally existing ligand to a PDGFRα, can be determined using known competition experiments. In some aspects, a PDGFRα inhibitor described herein competes with, and inhibits the binding of the naturally existing ligand to PDGFRα by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or 100%. Competition assays can be conducted as described herein or, for example, in Ed Harlow and David Lane, Cold Spring Harb. Protoc.; 2006; doi: 10.1101/pdb.prot4277 or in Chapter 11 of “Using Antibodies” by Ed Harlow and David Lane, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, USA 1999. [0171] Additional non-limiting examples of other competitive binding assays that can be used with the present disclosure include: solid phase direct or indirect radioimmunoassay (RIA), solid phase direct or indirect enzyme immunoassay (EIA), sandwich competition assay (see Stahli et al., Methods in Enzymology 9:242 (1983)); solid phase direct biotin- avidin EIA (see Kirkland et al., J. Immunol.137:3614 (1986)); solid phase direct labeled assay, solid phase direct labeled sandwich assay (see Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Press, Second Edition (2014)); solid phase direct label RIA using 1-125 label (see Morel et al., Mol. Immunol.25(1):7 (1988)); solid phase direct biotin-avidin EIA (Cheung et al., Virology 176:546 (1990)); and direct labeled RIA. (Moldenhauer et al., Scand. J. Immunol.32:77 (1990)). [0172] As further described elsewhere in the present disclosure, in some aspects, the antagonistic activity of a PDGFRα inhibitor described herein does not depend on interfering with the binding of another protein or compound to PDGFRα. Another non-limiting example includes small molecules that can bind to PDGFRα and inhibit (partially or completely) binding of ATP or substrate, or otherwise inhibit PDGFRα activity. In some aspects, a PDGFRα inhibitor described herein can inhibit or reduce PDGFRα activity via allosteric inhibition. In some aspects, a PDGFRα inhibitor can reduce protein levels of PDGFRα by reducing synthesis or enhancing degradation. While exemplary mechanisms of actions are provided, it will be apparent from the present disclosure that the PDGFRα inhibitors described herein are not intended to be limited to such mechanisms. [0173] As used herein, the terms “ug” and “uM” are used interchangeably with “μg” and “μΜ,” respectively. [0174] Various aspects described herein are described in further detail in the following subsections. The various aspects and embodiments of the present disclosure can optionally be combined. For instance, in some embodiments any of the disclosed compounds can be used in any of the disclosed methods and uses. II. PDGFRα Inhibitors [0175] Disclosed herein are compounds that can inhibit PDGFRα activity and/or elicit one or more activities associated with inhibition of the receptor. PDGFRα can bind with high affinity to several PDGF isoforms (i.e., PDGF-A, PDGF-B, PDGF-C, and PDGF-D). As used herein, references to inhibition of PDGFRα binding to its ligand refer to inhibition of binding of one or more of the PDGF isoforms that bind to PDGFRα. In some embodiments, the binding of all isoforms of PDGF that bind to PDGFRα is inhibited. The binding of the platelet-derived growth factor (PDGF) isoforms to PDGFRα affects various cell signaling pathways, such as those involved in cell proliferation and differentiation. Accordingly, PDGFRα activity is critical for both the proper development and long-term maintenance of certain tissues and organs. For example, animals globally lacking a functional PDGFRA gene die at birth or soon thereafter due to various developmental defects (e.g., cardiac malformations). Bax et al., Dev. Dyn. 239(8): 2307-2317 (Aug. 2010). Conditional PDGFRα knockout in OPCs during animal development results in severe hypomyelination and the animals die soon after birth. Hamashima et al., Neuroscience 436: 11-26 (Jun.2020) [0176] In humans, the gene encoding PDGFRα (i.e., PDGFRA) is located on chromosome 4 (for example, nucleotides 54,229,127-54,298,245 of GenBank Accession Number NC_000004.12; plus strand orientation). In addition to those provided above, other synonyms of PDGFRα are known and include: “PDGFRa,” “PDGFR2,” “alpha-type platelet-derived growth factor receptor,” “platelet-derived growth factor receptor A,” “platelet-derived growth factor receptor alpha,” “platelet-derived growth factor receptor 2,” “CD140a,” and “CD140 antigen-like family member A.” As used herein, the term “PDGFRα” includes any variants or isoforms of PDGFRα that are naturally expressed by cells. [0177] Despite its importance in various biological processes, the present disclosure describes the discovery that inhibiting or reducing PDGFRα activity, for example, with one or more of the compounds described herein, can have certain biological effects that can be useful for treating demyelinating diseases, such as those described herein. As demonstrated herein, in some aspects, inhibiting or reducing PDGFRα activity with the PDGFRα inhibitors described herein can promote oligodendrocyte differentiation and induce myelin formation. In some aspects, the PDGFRα inhibitors of the present disclosure are capable of (and in some embodiments used for) promoting remyelination of demyelinated neuronal axons, which can be useful in the treatment of various demyelinating diseases (e.g., multiple sclerosis). As used herein, the terms “PDGFRα inhibitors” and “PDGFRα antagonists” are used interchangeably and refer to any compound that is capable of (and in some embodiments used for) reducing and/or inhibiting PDGFRα activity (e.g., by any of the exemplary mechanisms or methods described herein). In some aspects, a PDGFRα inhibitor provided herein can inhibit a tyrosine kinase activity of a PDGFRα. In some aspects, a PDGFRα inhibitor provided herein can inhibit any other activity of a PDGFRα. Where the term “capable of” is used herein to describe certain features of a PDGFRα inhibitor, the term means that the PDGFRα inhibitor can (i.e., has the ability to) exhibit such features, e.g., under appropriate conditions. Unless indicated otherwise, the term does not mean that the PDGFRα inhibitor always exhibits such features, e.g., when administered to a subject with impaired oligodendrocyte progenitor cell population. The term “capable of” and “can” are used interchangeably in the present application. [0178] In some aspects, a PDGFRα inhibitor inhibits the kinase activity of PDGFRα. As used herein, unless indicated otherwise, the term “inhibit” (and derivatives thereof) comprises both complete inhibition and partial inhibition (e.g., reduced kinase activity). Accordingly, in some aspects, when an OPC (expressing PDGFRα) interacts with a PDGFRα inhibitor described herein, the PDGFRα kinase activity is reduced by at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90% or more (for example, at least 50% or more), compared to PDGFRα kinase activity in a corresponding OPC that was not contacted with the PDGFRα inhibitor described herein (e.g., contacted with a vehicle control). In some aspects, the kinase activity is completely inhibited. As is apparent from the present disclosure, the inhibition (partial or complete) of PDGFRα kinase activity in an OPC can promote the differentiation of the OPC into a myelinating oligodendrocyte. Inhibition of PDGFRα kinase activity can be assayed using methods known in the art. In some aspects, the inhibition of PDGFRα kinase activity can be determined using the in vitro differentiation assay described in Example 132 of the present application. In some aspects, the inhibition of PDGFRα kinase activity can be assessed using a cuprizone model for demyelination, such as that described in Torkildsen et al., Acta Neurol Scand Suppl 188:72-6 (2008). In some aspects, the inhibition of PDGFRα kinase activity is determined using an enzymatic PDGFRα kinase assay, such as that described in Example 131 of the present disclosure. In some aspects, the inhibition of PDGFRα kinase activity can be determined using an in vivo OPC differentiation assay, such as that described in Example 134. [0179] As described herein, in some aspects, a PDGFRα inhibitor useful for the present disclosure comprises one or more of the following properties: (1) capable of (and in some embodiments used for) promoting (e.g., inducing and/or enhancing) the differentiation of an OPC into an oligodendrocyte (in vitro, in vivo, or both); (2) capable of (and in some embodiments used for) promoting (e.g., inducing and/or enhancing) the expression of a protein associated with oligodendrocyte differentiation and/or myelination (e.g., G-protein coupled receptor 17 (GPR17), myelin basic protein (MBP), ASPA, GST-pi, CC1, myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, proteolipid protein 1 (PLP1), or a combination thereof); (3) capable of (and in some embodiments used for) promoting (e.g., inducing and/or enhancing) the myelination of a neuronal axon; (4) capable of (and in some embodiments used for) promoting (e.g., inducing and/or enhancing) the remyelination of a demyelinated neuronal axon; (5) capable of (and in some embodiments used for) inhibiting a PDGFRα kinase activity (e.g., IC50 of less than 10,000 nM, as measured using a Promega kinase assay; (e.g., described in Example 131); (6) capable of (and in some embodiments used for) penetrating the brain (e.g., following systemic administration) at a sufficient level to support activity in the CNS; and (7) any combination thereof. Non-limiting examples of additional properties are provided elsewhere in the present disclosure. [0180] In addition to the properties described above, a PDGFRα inhibitor described herein can comprise one or more additional features that are not present in other inhibitors known in the art. As is apparent from the present disclosure, such additional features can be useful in various clinical settings, e.g., to treat a demyelinating disease, such as those described herein. [0181] As described elsewhere in the present disclosure, some exemplary PDGFRα inhibitors of the present disclosure are capable of have greater therapeutic effect than inhibitors known in the art, e.g., when an inhibitor known in the art and an exemplary inhibitor provided herein are assayed against a vehicle control. For example, in some aspects, a PDGFRα inhibitor can have greater potency compared to inhibitors known in the art. In some aspects, a PDGFRα inhibitor can better penetrate the CNS (e.g., can more effectively pass across the blood-brain barrier). Accordingly, in some aspects, PDGFRα inhibitors described herein are more effective at promoting OPC differentiation as compared to inhibitors known in the art. For example, in some aspects, a PDGFRα inhibitor of the present disclosure can achieve greater than 2-fold increase in GPR17 expression compared to a vehicle control at a dose of less than 50 mg/kg, as measured using an in vivo GPR17 assay (such as that described in Example 134). In some aspects, a PDGFRα inhibitor of the present disclosure can achieve greater than 2-fold increase in GPR17 expression compared to the vehicle control at a dose of less than 40 mg/kg, less than 30 mg/kg, less than 20 mg/kg, less than 10 mg/kg, less than 5 mg/kg, less than 4 mg/kg, less than 3 mg/kg, less than 2 mg/kg, or less than 1 mg/kg, as measured using an in vivo GPR17 assay (such as that described in Example 134). As described elsewhere in the present disclosure, GPR17 expression is a suitable marker for OPC differentiation. Additionally, in some aspects, any of the other proteins associated with oligodendrocyte differentiation and/or myelination can be used instead of GPR17 or in combination with GPR17. Non- limiting examples of such proteins include: MBP, ASPA, GST-pi, CC1, MOG, oligodendrocyte-specific protein/claudin-11, CNPase, proteolipid protein 1 (PLP1), or a combination thereof. [0182] In some aspects, the PDGFRα inhibitor is a compound of Formula I:
Figure imgf000047_0001
, [0183] or a pharmaceutically acceptable salt or solvate thereof, wherein: indicates a single bond or a double bond such that all valences are satisfied; X1, X2, X3, and X4 are selected from N and CRa, with the proviso that not more than two of X1, X2, X3, and X4 are N; one of Y1 and Y2 is N and the other of Y1 and Y2 is C; each Ra is independently selected from H, halo, C1-C4alkyl, and C1-C4alkoxy; R1 is selected from C1-C4alkyl, C3-C8cycloalkyl, 3-8 membered heterocyclyl, heteroaryl, aryl, and C1-C8alkoxy, all of which can be optionally substituted with one, two, three, four, five, or six substituents selected from halo, hydroxy, oxo, C1-C4alkyl, aminoC1- C4alkyl, hydroxyC1-C4alkyl, C1-C4alkoxy, C1-C4alkoxyC1-C4alkyl, 3-8 membered heterocyclyl, and 3-8 membered heterocyclylC1-C4alkyl, with the proviso that the number of substituents does not exceed the number of substitutable positions; R2 is selected from cycloalkyl, cycloalkenyl, alkyl, aminoalkylamino, amino, heterocyclyl, heteroaryl, aminoheterocyclyl, heterocyclylamino, and aminoalkylamino, all of which can be optionally substituted with one or more substituents selected from D, halo, oxo, and C1-C4alkyl; R2 is substituted with one, two, or three R3; R3 is selected from aryl, heteroaryl, -C(O)R31, -C(O)OR31, -C(O)NR31R32, -S(O)2NR31R32, -S(O)2R31, -S(O)(NR33)R31, -S(O)(NR33)NR31R32, -C(S)NR31R32, C3- C8cycloalkyl, 3-8 membered heterocyclyl, and C1-C4alkyl, all of which can be optionally substituted with one, two, three, four, or five R30; each R30 is independently selected from D, halo, aryl, -OR300, -NR300R301, -S(O)rR300, -C(O)R300, -C(=CR34R35)R300, and ; r is selected from 0, 1, and 2;
Figure imgf000048_0001
each R300 is independently selected from C1-C6alkyl, C3-C7cycloalkyl, aryl, heteroaryl, 3-8 membered heterocyclyl, and 3-8 membered heterocyclylaryl, all of which can be optionally substituted with one, two, three, four, or five substituents selected from D, halo, hydroxy, amino, alkylamino, cyano, C1-C4alkyl, C1-C4haloalkyl, and C1-C4alkoxy; each R301 is independently selected from H, halo, and C1-C4alkyl; each R302 is independently selected from H, F, hydroxy, amino, alkylamino, oxo, and C1-C4alkoxy; each R303 is independently selected from H and C1-C4alkyl; n, o, and p are each independently selected from 0, 1, 2, 3, and 4; each R31 is independently selected from C1-C8alkyl, arylC1-C4alkyl, heteroarylC1- C4alkyl, heterocyclyl, heterocyclylC1-C4alkyl, cycloalkyl, and cycloalkylC1-C4alkyl, all of which can be optionally substituted with one, two, three, four, or five substituents selected from D, halo, cyano, hydroxy, amino, -OCF3, C1-C4alkyl, C1-C4haloalkyl, C1-C4alkoxy, hydroxyC1-C4alkyl, -S(O)2NR304R305, -C(O)OR304R305, -C(O)NR304R305, and -NR304C(O)R305; each R304 and R305 is independently selected from H and C1-C4 alkyl; each R32 is independently selected from H and C1-C4alkyl; or R31 and R32 together with the atom to which they are connected form a 5-8 membered heterocycyl, optionally substituted with one, two, three, four, or five substituents selected from D, halo, cyano, C1-C4alkyl, and C1-C4haloalkyl, and -C(O)NR34R35; each R34 and R35 is independently selected from H, C1-C4alkyl, and C1-C4haloalkyl; and each R33 is independently selected from H, C1-C4alkyl, C1-C4haloalkyl, and –C(O)R34; or R31 and R33 together with the atoms to which they are connected form a 4-8 membered heterocycyl. [0184] In some aspects, Y1 is N and Y2 is C. [0185] In some aspects, Y1 is C and Y2 is N. [0186] In some aspects, X1 is N, X2 is CRa, X3 is CRa, and X4 is CRa. [0187] In some aspects, X1 is CRa, X2 is N, X3 is CRa, and X4 is CRa. [0188] In some aspects, X1 is CRa, X2 is CRa, X3 is N, and X4 is CRa. [0189] In some aspects, X1 is CRa, X2 is CRa, X3 is CRa, and X4 is N. [0190] In some aspects, X1 is CRa, X2 is CRa, X3 is CRa, and X4 is CRa. [0191] In some aspects, R1 is 5- or 6-membered heteroaryl. [0192] In some aspects, R1 is an optionally substituted pyrazolyl. [0193] In some aspects, R1 is selected from:
Figure imgf000049_0002
, , an
Figure imgf000049_0003
d ; and R10 is selected from H, C1-C4alkyl, C1-C4alkoxy, aminoC1-C4alkyl, hydroxyC1- C4alkyl, and C1-C4alkylsulfonyl, wherein the C1-C4alkyl, C1-C4alkoxy, aminoC1-C4alkyl, hydroxyC1-C4alkyl, and C1-C4alkylsulfonyl can be optionally substituted by one or more substituents selected from hydroxyl, C1-C4alkoxy, NR10aR10b, halo, and deuterium, wherein R10a and R10b are selected from hydrogen and C1-C4alkyl, or wherein R10a and R10b taken together with the nitrogen atom to which they are attached form a 4- to 8-membered ring. [0194] In some aspects, R1 is
Figure imgf000049_0001
. [0195] In some aspects, R10 is CH3. [0196] In some aspects, R2 is selected from cycloalkyl, cycloalkenyl, alkyl, oxoalkylamino, aminoalkylamino, amino, heterocyclyl, heteroaryl, aminoheterocyclyl, heterocyclylamino, and aminoalkylamino, all of which can be optionally substituted with six or more substituents selected from D, halo, hydroxy, oxo, and C1-C4alkyl. [0197] In some aspects, R2 is heterocyclyl optionally substituted with C1-C4alkyl or oxo. [0198] In some aspects, R2 is substituted with one R3. [0199] In some aspects, R2 is selected from: ,
Figure imgf000050_0001
, , , ,
, ,
Figure imgf000051_0001
, , , ,
nd
Figure imgf000052_0001
indicates a single bond or a double bond such that all valences are satisfied; m is selected from 0, 1, 2, 3, 4, 5, and 6; and Z1, Z2, and Z3 are selected from N and CRa. [0200] In some aspects, R2 is selected from:
Figure imgf000053_0001
, , , , . [0201] In some aspects, R2 is selected from the group consisting of:
Figure imgf000054_0001
. [0202] In some aspects, R2 is:
Figure imgf000054_0002
. [0203] In some aspects, R3 is selected from aryl, heteroaryl, -C(O)R31, -C(O)OR31, - C(O)NR31R32, -S(O)2NR31R32, cycloalkyl, and alkyl, all of which can be optionally substituted with one, two, three, four, or five R30. [0204] In some aspects, R3 is selected from heteroaryl optionally substituted with one, two three, four, or five R30 and -C(O)OR31. [0205] In some aspects, the PDGFRα inhibitor is a compound of Formula Ia:
Figure imgf000055_0001
, [0206] or a pharmaceutically acceptable salt or solvate thereof, wherein a and b are each independently selected from 1, 2, and 3; and Q is selected from -CH- and -N-, with the proviso that if Q is –N-, a and b are not 1. [0207] In some aspects, the PDGFRα inhibitor is a compound of Formula II:
Figure imgf000055_0002
, or a pharmaceutically acceptable salt or solvate thereof. [0208] In some aspects, the PDGFRα inhibitor is a compound of Formula IIa:
Figure imgf000056_0001
, or a pharmaceutically acceptable salt or solvate thereof. [0209] In some aspects, R3 is selected from: ,
Figure imgf000056_0002
, , , , ,
, ,
Figure imgf000057_0001
, , wherein A1 is selected from O, S, and NR37, R36 is selected from hydrogen, optionally substituted C1-C6alkyl, and optionally substituted C1-C6alkylaryl, and R37 is selected from hydrogen and C1-C6alkyl. [0210] In some aspects, R3 is selected from:
,
Figure imgf000058_0001
,
Figure imgf000059_0001
, [0211] In some aspects, R3 is selected from:
Figure imgf000059_0002
. [0212] In some aspects, R3 is heteroaryl. [0213] In some aspects, R3 is:
Figure imgf000059_0003
. [0214] In some aspects, R30 is:
Figure imgf000060_0001
. [0215] In some aspects, R300 is selected from:
Figure imgf000060_0002
, , , , , [0216] In some aspects, R300 is:
Figure imgf000060_0003
. [0217] In some aspects, R301 is selected from H and CH3. [0218] In some aspects, R301 is H. [0219] In some aspects, R302 is H. [0220] In some aspects, R301 and R302 are H. [0221] In some aspects, R30 is benzyl. [0222] In some aspects, R3 is selected from: d
Figure imgf000061_0001
. [0223] In some aspects, R3 is selected from:
Figure imgf000061_0002
, , . [0224] In some aspects, R32 is H. [0225] In some aspects, the PDGFRα inhibitor is a compound of Formula IV:
Figure imgf000062_0001
, or a pharmaceutically acceptable salt or solvate thereof, wherein L1 is a NH or O. [0226] In some aspects, the PDGFRα inhibitor is a compound of Formula V:
Figure imgf000062_0002
, or a pharmaceutically acceptable salt or solvate thereof. [0227] In some aspects, R31 is selected from:
Figure imgf000063_0001
, , , ,
Figure imgf000064_0001
,
Figure imgf000065_0001
, wherein R31a is selected from H, D, alkylamino, optionally substituted C1-C4alkyl, C1- C4alkoxy, amino, and C1-C4haloalkyl; each R31b is independently selected from H, D, halo, hydroxy, amino, cyano, alkylamino, optionally substituted C1-C4alkyl, C1-C4haloalkyl, optionally substituted C1-C4alkoxy, and C3- C6cycloalkyl, and q is 1, 2, or 3. [0228] Table 1 (below) provides the structure of exemplary compounds that are useful for the present disclosure (e.g., can target and inhibit kinase activity associated with PDGFRα expressed on OPCs). Table 1. Exemplary Compounds With PDGFRα Inhibitory Activity
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
Figure imgf000087_0001
Figure imgf000088_0001
Figure imgf000089_0001
[0229] In some aspects, the exemplary compounds of the present invention do not include compounds 156, 250 and 275. In some aspects, the exemplary compounds of the present invention do not include the compounds described in Table 1 on pp. 117-170 of International Appl. No. PCT/US2022/079480, which is herein incorporated by reference in its entirety. [0230] In some aspects, the PDGFRa inhibitor is a compound described in Table 1 on pp. 117-170 of International Appl. No. PCT/US2022/079480. [0231] In some aspects, the PDGFRa inhibitor is not a compound described in Table 1 on pp.117-170 of International Appl. No. PCT/US2022/079480. III. Pharmaceutical Compositions [0232] Provided herein are compositions comprising a PDGFRα inhibitor of the present disclosure having the desired degree of purity, and a pharmaceutically acceptable carrier or excipient, in a form suitable for administration to a subject. In some aspects, the PDGFRα inhibitor has a purity of about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or greater than about 99%. Pharmaceutically acceptable excipients or carriers can be determined in part by the particular composition being administered, as well as by the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of pharmaceutical compositions (See, e.g., Remington, 23rd Edition, The Science and Practice of Pharmacy, editor: A. Adejare, 2020, Adademic Press.). The pharmaceutical compositions are generally formulated sterile and in full compliance with all Good Manufacturing Practice (GMP) regulations of the U.S. Food and Drug Administration. [0233] In some aspects, a pharmaceutical composition comprises a PDGFRα inhibitor described herein, and a pharmaceutically acceptable carrier. Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; 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 (e.g., Zn-protein complexes); and/or non-ionic surfactants such as TWEEN®, PLURONICS®, or polyethylene glycol (PEG). [0234] In some aspects, a pharmaceutical composition disclosed herein comprises one or more additional components selected from: a bulking agent, stabilizing agent, surfactant, buffering agent, or combinations thereof. [0235] Buffering agents useful for the current disclosure can be a weak acid or base used to maintain the acidity (pH) of a solution near a chosen value after the addition of another acid or base. Suitable buffering agents can maximize the stability of the pharmaceutical compositions by maintaining pH control of the composition. Suitable buffering agents can also ensure physiological compatibility or optimize solubility. Rheology, viscosity and other properties can also be dependent on the pH of the composition. Common buffering agents include, but are not limited to, a Tris buffer, a Tris-Cl buffer, a histidine buffer, a TAE buffer, a HEPES buffer, a TBE buffer, a sodium phosphate buffer, a MES buffer, an ammonium sulfate buffer, a potassium phosphate buffer, a potassium thiocyanate buffer, a succinate buffer, a tartrate buffer, a DIPSO buffer, a HEPPSO buffer, a POPSO buffer, a PIPES buffer, a PBS buffer, a MOPS buffer, an acetate buffer, a phosphate buffer, a cacodylate buffer, a glycine buffer, a sulfate buffer, an imidazole buffer, a guanidine hydrochloride buffer, a phosphate-citrate buffer, a borate buffer, a malonate buffer, a 3- picoline buffer, a 2-picoline buffer, a 4-picoline buffer, a 3,5-lutidine buffer, a 3,4-lutidine buffer, a 2,4-lutidine buffer, a Aces, a diethylmalonate buffer, a N-methylimidazole buffer, a 1,2-dimethylimidazole buffer, a TAPS buffer, a bis- Tris buffer, a L-arginine buffer, a lactate buffer, a glycolate buffer, or combinations thereof. [0236] In some aspects, a pharmaceutical composition disclosed herein further comprises a bulking agent. Bulking agents can be added to a pharmaceutical product in order to add volume and mass to the product, thereby facilitating precise metering and handling thereof. Bulking agents that can be used with the present disclosure include, but are not limited to, sodium chloride (NaCl), mannitol, glycine, alanine, or combinations thereof. [0237] In some aspects, a pharmaceutical composition disclosed herein can also comprise a stabilizing agent. Non-limiting examples of stabilizing agents that can be used with the present disclosure include: sucrose, trehalose, raffinose, arginine, or combinations thereof. [0238] In some aspects, a pharmaceutical composition disclosed herein comprises a surfactant. In some aspects, the surfactant can be selected from the following: alkyl ethoxylate, nonylphenol ethoxylate, amine ethoxylate, polyethylene oxide, polypropylene oxide, fatty alcohols such as cetyl alcohol or oleyl alcohol, cocamide MEA, cocamide DEA, polysorbates, dodecyl dimethylamine oxide, or combinations thereof. In some aspects, the surfactant is polysorbate 20 or polysorbate 80. [0239] In some aspects, a pharmaceutical composition disclosed herein (e.g., comprising a PDGFRα inhibitor) further comprises an amino acid. In some aspects, the amino acid is selected from arginine, glutamate, glycine, histidine, or combinations thereof. In some aspects, the composition further comprises a sugar alcohol. Non-limiting examples of sugar alcohol includes: sorbitol, xylitol, maltitol, mannitol, or combinations thereof. [0240] A pharmaceutical composition disclosed herein (e.g., comprising a PDGFRα inhibitor) can be formulated for any route of administration to a subject. Specific examples of routes of administration include intramuscularly, cutaneously, subcutaneously, ophthalmic, intravenously, intraperitoneally, intradermally, intraorbitally, intracerebrally, intracranially, intraspinally, intraventricularly, intrathecally, intracapsularly, orally, pulmonarily, intranasally, intra-arterially, intralymphatically, periocularly, topically, transdermally, rectally, vaginally, or intratumorally, or via intratympanic injection. Parenteral administration, characterized by, e.g., cutaneous, subcutaneous, intramuscular, or intravenous injection, is also contemplated herein. [0241] Injectables can be prepared in conventional forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions. The injectables, solutions and emulsions also contain one or more excipients. Suitable excipients are, for example, water, saline, dextrose, glycerol, or ethanol. In addition, if desired, the pharmaceutical compositions to be administered can also contain minor amounts of non-toxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents, stabilizers, solubility enhancers, and other such agents, such as for example, sodium acetate, sorbitan monolaurate, triethanolamine oleate, and cyclodextrins. [0242] Pharmaceutically acceptable carriers used in parenteral preparations include aqueous vehicles, nonaqueous vehicles, antimicrobial agents, isotonic agents, buffers, antioxidants, local anesthetics, suspending and dispersing agents, emulsifying agents, sequestering or chelating agents, and other pharmaceutically acceptable substances. Examples of aqueous vehicles include sodium chloride injection, Ringer’s injection, isotonic dextrose injection, sterile water injection, dextrose, and lactated Ringer’s injection. Nonaqueous parenteral vehicles include fixed oils of vegetable origin, cottonseed oil, corn oil, sesame oil and peanut oil. Antimicrobial agents in bacteriostatic or fungistatic concentrations can be added to parenteral preparations packaged in multiple-dose containers which include, for example, phenols or cresols, mercurials, benzyl alcohol, chlorobutanol, methyl and propyl p-hydroxybenzoic acid esters, thimerosal, benzalkonium chloride, and benzethonium chloride. Isotonic agents include sodium chloride, and dextrose. Buffers include phosphate and citrate. Antioxidants include sodium bisulfate. Local anesthetics include procaine hydrochloride. Suspending and dispersing agents include sodium carboxymethylcelluose, hydroxypropyl methylcellulose, and polyvinylpyrrolidone. Emulsifying agents include Polysorbate 80 (TWEEN® 80). Sequestering or chelating agent of metal ions includes EDTA. Pharmaceutical carriers also include ethyl alcohol, polyethylene glycol, and propylene glycol for water miscible vehicles; and sodium hydroxide, hydrochloric acid, citric acid, or lactic acid for pH adjustment. [0243] Preparations for parenteral administration include sterile solutions ready for injection, sterile dry soluble products, such as lyophilized powders, ready to be combined with a solvent just prior to use, including hypodermic tablets, sterile suspensions ready for injection, sterile dry insoluble products ready to be combined with a vehicle just prior to use, and sterile emulsions. The solutions can be either aqueous or nonaqueous. [0244] If administered intravenously, suitable carriers include physiological saline or phosphate buffered saline (PBS), and solutions containing thickening and solubilizing agents, such as glucose, polyethylene glycol, and polypropylene glycol, and mixtures thereof. [0245] Topical mixtures comprising a compound described herein are prepared as described for the local and systemic administration. The resulting mixture can be a solution, suspension, emulsions, or the like and can be formulated as creams, gels, ointments, emulsions, solutions, elixirs, lotions, suspensions, tinctures, pastes, foams, aerosols, irrigations, sprays, suppositories, bandages, dermal patches, or any other formulations suitable for topical administration. [0246] A therapeutic agent described herein (e.g., PDGFRα inhibitor) can be formulated as an aerosol for topical application, such as by inhalation (see, e.g., U.S. Patent Nos. 4,044,126; 4,414,209; and 4,364,923, which describe aerosols for delivery of a steroid useful for treatment of inflammatory diseases, particularly asthma). These formulations for administration to the respiratory tract can be in the form of an aerosol or solution for a nebulizer, or as a microfine powder for insufflations, alone or in combination with an inert carrier such as lactose. In such a case, the particles of the formulation can have diameters of less than about 50 microns, e.g., less than about 10 microns. In some aspects, the particle diameters can have diameters of less than about 50 microns, e.g., less than about 10 microns, as measured by dynamic light scattering (DLS) or static image analysis using microscopy. [0247] A therapeutic agent disclosed herein (e.g., PDGFRα inhibitor) can be formulated for local or topical application, such as for topical application to the skin and mucous membranes, such as in the eye, in the form of gels, creams, and lotions and for application to the eye or for intracisternal or intraspinal application. Topical administration is contemplated for transdermal delivery and also for administration to the eyes or mucosa, or for inhalation therapies. [0248] Transdermal patches, including iontophoretic and electrophoretic devices, are well known to those of skill in the art, and can be used to administer a therapeutic agent (e.g., those disclosed herein). For example, such patches are disclosed in U.S. Patent Nos. 6,267,983; 6,261,595; 6,256,533; 6,167,301; 6,024,975; 6,010,715; 5,985,317; 5,983,134; 5,948,433; and 5,860,957. [0249] In some aspects, a pharmaceutical composition comprising a therapeutic agent described herein (e.g., PDGFRα inhibitor) is a lyophilized powder, which can be reconstituted for administration as solutions, emulsions, and other mixtures. It can also be reconstituted and formulated as a solid or a gel. The lyophilized powder is prepared by dissolving an compound as described herein, or a pharmaceutically acceptable derivative thereof, in a suitable solvent. In some aspects, the lyophilized powder is sterile. The solvent can contain an excipient, which improves the stability or other pharmacological component of the powder or reconstituted solution that is prepared from the powder. Excipients that can be used include, but are not limited to, dextrose, sorbitol, fructose, corn syrup, xylitol, glycerin, glucose, sucrose, or other suitable agent. The solvent can also contain a buffer, such as citrate, sodium or potassium phosphate or other such buffer known to those of skill in the art at, in some aspects, about neutral pH. Subsequent sterile filtration of the solution followed by lyophilization under standard conditions known to those of skill in the art provides the desired formulation. In some aspects, the resulting solution can be apportioned into vials for lyophilization. Each vial can contain a single dosage or multiple dosages of the compound. The lyophilized powder can be stored under appropriate conditions, such as at about 4°C to room temperature. [0250] Reconstitution of a lyophilized powder with water for injection provides a formulation for use in parenteral administration. For reconstitution, the lyophilized powder is added to sterile water or other suitable carrier. The precise amount depends upon the selected compound. Such an amount can be empirically determined. [0251] Pharmaceutical compositions provided herein (e.g., comprising a PDGFRα inhibitor) can also be formulated to be targeted to a particular tissue, receptor, or other area of the body of the subject to be treated. Many such targeting methods are known to those of skill in the art. All such targeting methods are contemplated herein for use in the instant compositions. For non-limiting examples of targeting methods, see, e.g., U.S. Patent Nos. 6,316,652; 6,274,552; 6,271,359; 6,253,872; 6,139,865; 6,131,570; 6,120,751; 6,071,495; 6,060,082; 6,048,736; 6,039,975; 6,004,534; 5,985,307; 5,972,366; 5,900,252; 5,840,674; 5,759,542; and 5,709,874. [0252] The pharmaceutical compositions to be used for in vivo administration can be sterile. This can be accomplished, for example, by filtration through, e.g., a sterile filtration membrane. IV. Kits [0253] Also provided herein are kits comprising one or more PDGFRα inhibitors described herein. In some aspects, provided herein is a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions described herein, such as one or more PDGFRα inhibitors provided herein, and optionally, an instruction for use. In some aspects, the kits contain a pharmaceutical composition described herein (e.g., a PDGFRα inhibitor) and any prophylactic or therapeutic agent, such as those described herein. V. Methods of the Disclosure [0254] As demonstrated herein, PDGFRα inhibitors of the present disclosure are useful in reducing or inhibiting PDGFRα activity, which, in some aspects, can promote the myelination of an axon and/or remyelination of a demyelinated neuronal axon. [0255] In some aspects, the present disclosure relates to methods of inhibiting or reducing PDGFRα activity in a cell or tissue, e.g., of a subject in need thereof, comprising contacting the cell or tissue with a PDGFRα inhibitor described herein. In some aspects, after the contacting, PDGFRα kinase activity in the cell or tissue is inhibited by at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or 100% compared to a reference (e.g., PDGFRα activity in the cell prior to the contacting and/or PDGFRα activity in a corresponding cell that was not contacted with the PDGFRα inhibitor). The PDGFRα activity status of a cell can be determined using any suitable method known in the art. In some aspects, PDGFRα activity can be determined by measuring the corresponding gene expression level, e.g., by qRT- PCR. In some aspects, PDGFRα activity can be determined by measuring the PDGFRα protein level, e.g., by immunoblotting. In some aspects, the PDGFRα activity can be determined by measuring receptor phosphorylation, e.g., by western blotting. An exemplary method of assaying such an activity, a purified protein enzyme assay, is provided in Example 131 (also referred to herein as the "Promega Assay"). In some aspects, the enzymatic PDGFRα kinase assay comprises 20 ng of purified PDGFRα protein, 150 µM of ATP and 1 µg of substrate, Poly (Glu4Tyr1) in a volume of 15 µl. The potency of a compound can be determined using such assays. In general, compounds most useful in the methods of the present disclosure have an IC50 below 10,000 nM (e.g., less than 9,000 nM, less than 8,000 nM, less than 7,000 nM, less than 6,000 nM, less than 5,000 nM, less than 4,000 nM, less than 3,000 nM, less than 2,000 nM, less than 1,000 nM, less than 900 nM, less than 800 nM, less than 700 nM, less than 600 nM, less than 500 nM, less than 400 nM, less than 300 nM, less than 200 nM, less than 100 nM, less than 50 nM, less than 25 nM, less than 10 nM, less than 5 nM, less than 2.5 nM, less than 1 nM, less than 0.5 nM, or less than 0.2 nM, for instance, less than 500 nM) in such an assay. As described herein, in some aspects, the cell is an oligodendrocyte progenitor cell (OPC). [0256] As both described and demonstrated herein, inhibiting or reducing PDGFRα activity can promote the myelination of a neuron (e.g., an axon of a neuron,), e.g., by promoting the differentiation of an OPC into an oligodendrocyte that, in turn, myelinates the axon. Similarly, inhibiting or reducing PDGFRα activity can also promote the remyelination of a demyelinated neuronal axon. And, as will be apparent to those skilled in the art, in some aspects, remyelination can repair damage to demyelinated (e.g., hypomyelinated) axons, and thereby, aid in recovering axonal signalling or reducing axonal injury or loss. [0257] Accordingly, in some aspects, provided herein is a method of promoting the myelination of an axon, e.g., in a subject in need thereof (e.g., subject suffering from or at risk of developing a demyelinating disease), the method comprising contacting an OPC with an effective amount of a PDGFRα inhibitor described herein, wherein the contacting results in the OPC to differentiate into an oligodendrocyte, and wherein the oligodendrocyte is capable of (and in some embodiments used for) promoting the myelination of the axon. In some aspects, after the contacting, the number of neurons with axons that are myelinated is increased by at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, at least 10-fold, at least 15-fold, at least 20- fold, at least 25-fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, or at least 50-fold compared to a reference (e.g., the number of neurons with myelinated axons in the subject prior to the administration and/or the number of neurons with myelinated axons in a corresponding subject that did not receive the administration of the PDGFRα inhibitor). The increase in the number of neurons with axons that are myelinated can be determined using any suitable methods known in the art or described herein. In some aspects, the increase in the number of neurons with axons that are myelinated can be determined by visualizing and/or quantifying the expression of a marker associated with myelinated neurons. For example, in some aspects, the marker associated with myelinated neurons comprises myelin basic protein (MBP), myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, proteolipid protein 1 (PLP1), or a combination thereof. [0258] In some aspects, also provided herein is a method of promoting remyelination of an axon of a neuron (e.g., demyelinated neuronal axon in a subject in need thereof), comprising contacting an OPC with an effective amount of any of the PDGFRα inhibitors described herein, wherein the contacting results in the OPC to differentiate into an oligodendrocyte, and wherein the oligodendrocyte is capable of (and in some embodiments used for) promoting the remyelination of the demyelinated neuronal axon. In some aspects, after the contacting, the number of demyelinated axons that are myelinated is increased by at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, at least 10-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, or at least 50-fold compared to a reference (e.g., the number of demyelinated neuronal axons in the subject prior to the administration and/or the amount of demyelinated neuronal axons in a corresponding subject that did not receive the administration of the PDGFRα inhibitor). The increase in the number of demyelinated axons that are myelinated can be determined using any suitable methods known in the art or described herein. In some aspects, the increase in the number of demyelinated axons that are myelinated can be determined by visualizing and/or quantifying the expression of a marker associated with myelinated neurons. For example, in some aspects, the marker associated with myelinated neurons comprises myelin basic protein (MBP), myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, proteolipid protein 1 (PLP1), or a combination thereof. [0259] In some aspects, also provided herein is a method of increasing remyelination in a subject (e.g., increasing remyelination of a demyelinated neuronal axon in a subject in need thereof), comprising contacting an OPC with an effective amount of any of the PDGFRα inhibitors described herein, wherein the contacting results in the OPC to differentiate into an oligodendrocyte, and wherein the oligodendrocyte is capable of (and in some embodiments used for) increasing remyelination in the subject. In some aspects, after the contacting, the remyelination is increased by at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, at least 10-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, or at least 50-fold compared to a reference (e.g., the number of demyelinated neuronal axons in the subject prior to the administration and/or the amount of demyelinated neuronal axons in a corresponding subject that did not receive the administration of the PDGFRα inhibitor). The increase in the number of demyelinated axons that are myelinated can be determined using any suitable methods known in the art or described herein. In some aspects, the increase in the number of demyelinated axons that are myelinated can be determined by visualizing and/or quantifying the expression of a marker associated with myelinated neurons. For example, in some aspects, the marker associated with myelinated neurons comprises myelin basic protein (MBP), myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, proteolipid protein 1 (PLP1), or a combination thereof. [0260] In some aspects, provided herein is a method of reducing the demyelination of a myelinated axon (e.g., reducing the rate at which the myelinated axons become demyelinated) in a subject in need thereof, comprising contacting an OPC with an effective amount of any of the PDGFRα inhibitors described herein, wherein the contacting results in the OPC to differentiate into an oligodendrocyte, and wherein the oligodendrocyte is capable of (and in some embodiments used for) reducing the demyelination of the myelinated axon. In some aspects, after the contacting, demyelination of a myelinated axon in the subject is reduced by at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, at least 10-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, or at least 50-fold compared to a reference (e.g., demyelination of a myelinated axon in a corresponding subject that did not receive an administration of the PDGFRα inhibitor). The decrease in the number of neurons with axons that are demyelinated can be determined using any suitable methods known in the art or described herein. In some aspects, the decrease in the number of neurons with axons that are demyelinated can be determined by visualizing and/or quantifying the expression of a marker associated with myelinated neurons. For example, in some aspects, the marker associated with myelinated neurons comprises myelin basic protein (MBP), myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, proteolipid protein 1 (PLP1), or a combination thereof. [0261] In some aspects, provided herein is a method of decreasing the rate of demyelination (e.g., decreasing the rate at which myelinated axons in a subject become demyelinated) in a subject in need thereof, comprising contacting an OPC with an effective amount of any of the PDGFRα inhibitors described herein, wherein the contacting results in the OPC to differentiate into an oligodendrocyte, and wherein the oligodendrocyte is capable of (and in some embodiments used for) reducing the rate of demyelination in the subject. In some aspects, after the contacting, the rate of demyelination in the subject is decreasing by at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, at least 10-fold, at least 15-fold, at least 20-fold, at least 25- fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, or at least 50-fold compared to a reference (e.g., the rate of demyelination in a corresponding subject that did not receive an administration of the PDGFRα inhibitor). The decrease in the number of neurons with axons that are demyelinated can be determined using any suitable methods known in the art or described herein. In some aspects, the decrease in the number of neurons with axons that are demyelinated can be determined by visualizing and/or quantifying the expression of a marker associated with myelinated neurons. For example, in some aspects, the marker associated with myelinated neurons comprises myelin basic protein (MBP), myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, proteolipid protein 1 (PLP1), or a combination thereof. [0262] In any of the methods provided herein, in some aspects, the contacting can occur ex vivo (e.g., OPCs of a subject can be isolated and contacted with a PDGFRα inhibitor in vitro to assess the therapeutic efficacy of the inhibitor). In some aspects, the contacting occurs in vivo (e.g., in a subject in need thereof). Where the contacting occurs in vivo, the methods can further comprise administering to the subject an effective amount of any of the PDGFRα inhibitors described herein. [0263] Not to be bound by any one theory, in some aspects, after administration to a subject, a PDGFRα inhibitor described herein is capable of (and in some embodiments used for) migrating into the CNS of the subject (e.g., by crossing the blood-brain barrier) and interacting with the nervous system cells present within the CNS. For instance, as described and demonstrated herein, PDGFRα inhibitors of the present disclosure are capable of (and in some embodiments used for) targeting PDGFRα expressed on an OPC, and thereby inhibit or reduce the PDGFRα activity of the OPC. In some aspects, this results in the activation of the OPC and the subsequent differentiation of the OPC into an oligodendrocyte. Accordingly, in some aspects, provided herein is a method of activating an OPC within the CNS of a subject in need thereof, the method comprising administering to the subject any of the PDGFRα inhibitors described herein. [0264] As described herein, the myelin sheath which surrounds the neuronal axons can affect various functions of neurons. For example, in some aspects, by increasing the myelination of neuronal axons, PDGFRα inhibitors described herein can increase the rate of transmission of electrical impulses (i.e., nerve signals or action potentials) along the axon of a neuron. Accordingly, in some aspects, the present disclosure provides a method of increasing electrical impulse transmission along the axon of a neuron, comprising contacting an OPC with an effective amount of any of the PDGFRα inhibitors described herein, wherein the contacting results in the OPC to differentiate into an oligodendrocyte, and wherein the oligodendrocyte is capable of (and in some embodiments used for) myelinating the axon of the neuron, and thereby increase the electrical impulse transmission. In some aspects, after contacting the OPC, the electrical impulse transmission along the axon of the neuron is increased by at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, at least 10-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, or at least 50-fold compared to a reference (e.g., electrical impulse transmission across the axon prior to or in the absence of contact with the PDGFRα inhibitors). Action potential conduction velocities can be measured as compound action potentials across the corpus callosum in vitro or by auditory or visual evoked potential recording in vivo. In some aspects, remyelination may overcome conduction block, allowing axonal signals that would be prematurely terminated to be transmitted. Remyelination can enhance axonal health and prevent axonal degradation in a diseased state. Non-limiting examples of methods useful for measuring such properties of a neuronal axon are described in, e.g., Li et al., PLoS One 11(11): e0165637 (Nov.2016); Maheras et al., Sci Rep 8(1): 3798 (Feb. 2018); and Alqudah et al., Audiol Neurootol 23(1): 20-31 (2018). [0265] As is apparent from the present disclosure, the PDGFRα inhibitors described herein can be useful in the treatment of a demyelinating disease, e.g., by inducing the differentiation of an OPC into an oligodendrocyte, thereby promoting the remyelination of neuronal axons. Accordingly, in some aspects, the present disclosure is directed to a method of treating a demyelinating disease in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a PDGFRα inhibitor described herein. References to a method of treating a demyelinating disease as described herein also refer to the agents and compositions as described herein for use in treating a demyelinating disease. [0266] The PDGFRα inhibitors provided herein can be used to treat a wide range of demyelinating diseases (including those that are associated with demyelination and/or hypomyelination). In some aspects, the demyelinating disease that can be treated with the present disclosure comprise those that are characterized by demyelination of one or more cells within the CNS. Non-limiting examples of demyelinating disease that can be treated with the present disclosure include one or more of: acute disseminated encephalomyelitis (ADEM), acute hemorrhagic leukoencephalitis, acute optic neuritis, acute transverse myelitis, adrenoleukodystrophy, adrenomyeloneuropathy, Alexander disease, Alzheimer's disease, aminoacidurias, amyotrophic lateral sclerosis, anti-MAG peripheral neuropathy, anti-MOG associated spectrum, Balo concentric sclerosis, brain injury, CAMFAK Syndrome, Canavan disease, carbon monoxide toxicity, central pontine myelinolysis, cerebral hypoxia, cerebral ischemia, Charcot–Marie–Tooth disease, chronic inflammatory demyelinating polyneuropathy, chronic relapsing inflammatory optic neuritis (CRION), chronic traumatic encephalopathy, clinically isolated syndrome (CIS), congenital cataract, copper deficiency associated condition, delayed post-hypoxic leukoencephalopathy, diffuse cerebral sclerosis of Schilder, diffuse myelinoclastic sclerosis, extrapontine myelinolysis Gaucher disease, Guillain–Barré syndrome, hereditary neuropathy, hereditary neuropathy with liability to pressure palsy, HTLV-1–associated myelopathy, Hurler syndrome, hypomyelination, hypoxic brain injury, Krabbe disease, Leber hereditary optic atrophy and related mitochondrial disorders, leukodystrophic disorders, Marburg multiple sclerosis, Marchiafava-Bignami disease, metachromatic leukodystrophy, multiple sclerosis, multiple system atrophy, myelinoclastic disorders, myelopathy, nerve injury, neuromyelitis optica (NMO), Niemann-Pick disease, optic neuropathy, optic-spinal multiple sclerosis, osmotic demyelination syndrome, Parkinson's disease, Pelizaeus‐Merzbacher disease, periventricular leukomalacia, peripheral neuropathy, phenylketonuria, primary progressive multiple sclerosis (PPMS), progressive inflammatory neuropathy, progressive multifocal leukoencephalopathy, progressive subcortical ischemic demyelination, progressive-onset multiple sclerosis, relapsing-onset multiple sclerosis, relapsing-remitting multiple sclerosis (RRMS), reperfusion injury, Schilder disease, secondary progressive multiple sclerosis (SPMS), solitary sclerosis, spinal cord injury, subacute sclerosing panencephalitis, Tabes dorsalis, Tay-Sachs disease, transverse myelitis, traumatic brain injury, tropical spastic paraparesis, tumefactive multiple sclerosis, vitamin B12 deficiency, and cerebral palsy. In some aspects, a disease that can be treated with the present disclosure comprises a tumor associated with abnormal (e.g., increased) PDGF or PDGFRα activity. Such a tumor is referred to herein as "PDGF- associated tumor." Accordingly, in some aspects, provided herein is a method of treating a PDGF-associated tumor in a subject in need thereof, comprising administering to the subject any of the PDGFRα inhibitor described herein. A non-limiting example of a PDGF- associated tumor comprises oligodendroglioma. [0267] In some aspects, the demyelinating disease that can be treated with a PDGFRα inhibitor described herein comprises multiple sclerosis. As used herein, the term "multiple sclerosis" (MS) refers to a chronic and often disabling disease of the central nervous system characterized by the progressive destruction of the myelin sheath. Multiple sclerosis is generally diagnosed as one of four internationally recognized categories or stages of MS: (1) primary progressive multiple sclerosis (PPMS), (2) relapsing-remitting multiple sclerosis (RRMS), (3) secondary progressive multiple sclerosis (SPMS), and (4) progressive relapsing multiple sclerosis. Standards for diagnosis are known to those of skill in the art and exemplary diagnostic criteria are described in, e.g., "Merck Manual, Professional Version" (www.merckmanuals.com/professional/neurologic- disorders/demyelinating-disorders/multiple-sclerosis-ms). Unless indicated otherwise, the term "multiple sclerosis" encompasses all of the different categories of MS. Accordingly, in some aspects, the PDGFRα inhibitors of the present disclosure can be used to treat all types of MS. [0268] In some aspects, a PDGFRα inhibitor of the present disclosure can be used to treat clinically isolated syndrome (CIS). In some aspects, a PDGFRα inhibitor of the present disclosure can be used to treat radiologically isolated syndrome (RIS). [0269] In some aspects, provided herein is a method of treating a relapsing form of multiple sclerosis in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of any of the PDGFRα inhibitors provided herein. [0270] In some aspects, the demyelinating disease that can be treated with the present disclosure is an optic neuritis. Accordingly, in some aspects, a PDGFRα inhibitor is used to ameliorate optic neuritis, e.g., optic neuritis that is due to multiple sclerosis. [0271] As is apparent from the present disclosure, by promoting the myelination of an axon and/or the remyelination of a demyelinated axon, PDGFRα inhibitors of the present disclosure can help restore nervous system cell function and thereby, reduce and/or alleviate one or more symptoms associated with a demyelinating disease. Accordingly, in some aspects, treating a demyelinating disease comprises reducing one or more symptoms associated with the demyelinating disease. Non-limiting examples of such symptoms include one or more of: fatigue, dizziness, malaise, elevated fever and high body temperature, extreme sensitivity to cold in the hands and feet, weakness and stiffness in muscles and joints, weight changes, digestive or gastrointestinal problems, low blood pressure, high blood pressure, irritability, anxiety, depression, blurred vision, double vision, ataxia, clonus, spasms, dysarthria, weakness, clumsiness, hand paralysis, hemiparesis, genital anesthesia, sexual dysfunction, incoordination, paresthesias, ocular paralysis, impaired muscle coordination, loss of sensation, tingling, numbness, pain, impaired vision, neurological symptoms, unsteady gait, balance problems, dizziness, spastic paraparesis, incontinence, hearing problems, speech problems, loss of olfaction, and agusia. [0272] Moreover, PDGFRα inhibitors described herein can also be used to prevent or delay the onset of one or more symptoms associated with a demyelinating disease. With many demyelinating diseases, particularly during the early stages, a subject may not exhibit any apparent symptoms of the disease. For instance, in some aspects, the damage to the myelin sheaths may not be as severe, such that the oligodendrocytes present within the CNS of the subject can adequately repair any damages. However, as is the case with most demyelinating diseases (e.g., multiple sclerosis), the damage to the myelin sheaths can become more severe, resulting in the manifestation of one or more symptoms of the disease. In some aspects, by administering a PDGFRα inhibitor described herein to the subject when the damage to the myelin sheath is still minor, the manifestation of the one or more symptoms of the disease can be prevented or delayed. In some aspects, compared to a reference subject (e.g., corresponding subject that did not receive an administration of the PDGFRα inhibitor), the manifestation of the one or more symptoms of the disease is delayed by at least 1.1-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, at least 10-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 35-fold, at least 40-fold, at least 45-fold, or at least 50-fold. In some aspects, compared to the reference subject, the subject treated with a PDGFRα inhibitor described herein exhibits less number of symptoms and/or the symptoms are reduced in severity. [0273] Whether a subject exhibits one or more symptoms associated with a demyelinating disease can be assessed using any suitable approaches known in the art. In some aspects, whether a subject exhibits the one or more symptoms associated with a demyelinating disease can be determined by assessing the subject's performance in one or more of the following tests: a visual evoked potential (VEP) test, a multifocal visual evoked potential (mfVEP) test, a low contrast visual acuity (LCVA) test, a magnetic resonance imaging (MRI) (e.g., magnetization transfer ratio (MTR), myelin water fraction (MWF), quantitative susceptibility mapping (QSM), and T2 imaging), an electromyography (EMG), a nerve conduction velocity (NCV) test, an Extended Disability Status Scale (EDSS), a timed walk test (e.g., timed 25-foot walk), a Nine-Hole Peg Test (9HPT), an ocular coherence tomograph (OCT), a quality of life measure test (e.g., Multiple Sclerosis Quality of Life-54 and Vision-Related Quality of Life), cognitive assessment (e.g., Symbol-Digit Modalities Test or Montreal Cognitive Assessment), or combinations thereof. In some aspects, whether a subject exhibits one or more symptoms associated with a demyelinating disease is determined by assessing the subject's performance in a LC-VA test. In some aspects, whether a subject exhibits one or more symptoms associated with a demyelinating disease is determined by assessing the subject's performance in a cognitive assessment. [0274] In some aspects, an improved performance as compared to a reference performance (e.g., a corresponding subject's performance in the test where the reference subject was not treated with a PDGFRα inhibitor described herein and/or the subject's performance prior to the treatment with a PDGFRα inhibitor) indicates that the one or more symptoms associated with a demyelinating disease is reduced or alleviated in the subject. In some aspects, a comparable or reduced performance as compared to a reference performance (e.g., a corresponding subject's performance in the test where the subject was not treated with a PDGFRα inhibitor described herein and/or the subject's performance prior to the treatment with a PDGFRα inhibitor) indicates that the one or more symptoms associated with a demyelinating disease is maintained or has worsened in the subject. [0275] Accordingly, some aspects of the present disclosure are directed to methods of treating a demyelinating disease in a subject in need thereof, comprising assessing the subject's performance in a test for assessing one or more symptoms associated with a demyelinating disease, wherein the subject received a treatment comprising a PDGFRα inhibitor described herein prior to the assessment. In some aspects, if the subject's performance in the test is improved as compared to a reference performance (e.g., a corresponding subject's performance in the test where the reference subject was not treated with a PDGFRα inhibitor described herein and/or the subject's performance prior to the treatment with a PDGFRα inhibitor), the treatment comprising a PDGFRα inhibitor can be maintained in the subject (e.g., the subject receives one or more additional administration of the PDGFRα inhibitor at the same dose and/or dosing interval). In some aspects, where the subject exhibits improved performance, the treatment comprising a PDGFRα inhibitor is reduced (e.g., the subject receives one or more additional administration of the PDGFRα inhibitor but at a lower dose and/or longer dosing interval) or stopped. In some aspects, if the subject's performance is comparable or reduced as compared to a reference performance, (e.g., a corresponding subject's performance in the test where the subject was not treated with a PDGFRα inhibitor described herein and/or the subject's performance prior to the treatment with a PDGFRα inhibitor), the treatment comprising a PDGFRα inhibitor is adjusted such that the subject receives one or more administrations of the PDGFRα inhibitor at a higher dose and/or shorter dosing interval. [0276] Some aspects of the present disclosure are directed to methods of improving a subject's performance in a test for assessing one or more symptoms associated with a demyelinating disease, comprising administering to the subject any of the PDGFRα inhibitors described herein. In some aspects, after the administration, the subject's performance in the test is increased as compared to a reference performance (e.g., a corresponding subject's performance in the test where the reference subject was not treated with a PDGFRα inhibitor described herein and/or the subject's performance prior to the treatment with a PDGFRα inhibitor). In some aspects, the test is one or more of a visual evoked potential (VEP) test, a multifocal visual evoked potential (mfVEP) test, a low contrast visual acuity (LC-VA) test, a magnetic resonance imaging (MRI) (e.g., magnetization transfer ratio (MTR), myelin water fraction (MWF), quantitative susceptibility mapping (QSM), and T2 imaging), an electromyography (EMG), a nerve conduction velocity (NCV) test, an Extended Disability Status Scale (EDSS), a timed walk test (e.g., timed 25-foot walk), a Nine-Hole Peg Test (9HPT), an ocular coherence tomograph (OCT), a quality of life measure test (e.g., Multiple Sclerosis Quality of Life- 54 and Vision-Related Quality of Life), cognitive assessment (e.g., Symbol-Digit Modalities Test or Montreal Cognitive Assessment), or combinations thereof. In some aspects, the test is a LCVA test. In some aspects, the test is a cognitive assessment. [0277] As is apparent from the present disclosure, the introduction of a PDGFRα inhibitor into a subject can be done by any suitable route, including, but not limited to, intratumorally, orally, pulmonarily, intranasally, parenterally (intravenously, intra- arterially, intramuscularly, intraperitoneally, or subcutaneously), rectally, vaginally, intralymphatically, intrathecally, periocularly, cutaneously, intradermally, intraorbitally, intracerebrally, intracranially, intraspinally, intraventricuarly, intracapsularly, or topically. Administration includes self-administration and the administration by another. A suitable route of administration allows the PDGFRα inhibitors described herein to perform its intended function (e.g., inhibit PDGFRα activity and induce OPC differentiation). In some aspects, a suitable route of administration includes that which would allow a PDGFRα inhibitor to reach the CNS or any other sites where OPCs are present. Non-limiting examples of such routes include intranasal delivery, intrathecal administration, intracranial administration, and combinations thereof. In some aspects, a PDGFRα inhibitor described herein is administered orally. In some aspects, a PDGFRα inhibitor of the present disclosure is administered intravenously. In some aspects, a PDGFRα inhibitor can be administered both orally and intravenously. [0278] In any of the methods provided herein where a PDGFRα inhibitor is administered to a subject, the PDGFRα inhibitor can be administered to the subject using any suitable dosing schedule. In some aspects, the PDGFRα inhibitor is administered to the subject once. In some aspects, multiple doses of the PDGFRα inhibitor are administered to the subject. In some aspects, a PDGFRα inhibitor described herein is administered to a subject according to an intermittent dosing schedule. As used herein, the term "intermittent dosing schedule" (and any variants thereof) refers to a dosing schedule in which the therapeutic agent (e.g., PDGFRα inhibitor) is administered non-continuously (i.e., an intervening period exists between doses). An intermittent dosing schedule useful for the present disclosure can encompass any discontinuous administration regimen that provides a therapeutically effective amount of a PDGFRα inhibitor to a subject in need thereof. Intermittent dosing regimens can use equivalent, lower, or higher doses of a PDGFRα inhibitor than would be used in continuous dosing regimens. Advantages of intermittent dose administration include, but are not limited to, improved safety, decreased toxicity (e.g., decreased weight loss), acceptable levels of ADME criteria, acceptable levels of undesirable effects on organ systems such as heart, pulmonary, hepatic, reproductive (for example, ovarian or testicular) or gastrointestinal, increased exposure, increased efficacy, and/or increased subject compliance. These advantages can be realized when the PDGFRα inhibitor is administered as a single agent and/or when administered in combination with one or more additional therapeutic agents, e.g., standard of care. When administered in combination with one or more additional therapeutic agents, in some aspects, the dosing regimen for the PDGFRα inhibitor (e.g., intermittent dosing) and the additional therapeutic agents are independent of each other. For example, in some aspects, the PDGFRα inhibitor is administered using a first dosing regimen and the additional therapeutic agent is administered using a second dosing regimen, wherein the first and second dosing regimens are different. [0279] In some aspects, the intermittent dosing schedule comprises administering the PDGFRα inhibitor to the subject every other day. In some aspects, the PDGFRα inhibitor is administered to the subject one time per day. In some aspects, the PDGFRα inhibitor is administered to the subject two times per day. In some aspects, the PDGFRα inhibitor is administered to the subject three times per day. In some aspects, the PDGFRα inhibitor is administered to the subject four times per day. In some aspects, the PDGFRα inhibitor is administered to the subject every three days. In some aspects, the PDGFRα inhibitor is administered to the subject every four days. In some aspects, the PDGFRα inhibitor is administered to the subject every five days. In some aspects, the PDGFRα inhibitor is administered to the subject every six days. In some aspects, the PDGFRα inhibitor is administered to the subject once a week. In some aspects, the PDGFRα inhibitor is administered to the subject once every eight days. In some aspects, the PDGFRα inhibitor is administered to the subject once every nine days. In some aspects, the PDGFRα inhibitor is administered to the subject every 10 days. In some aspects, the PDGFRα inhibitor is administered to the subject every 11 days. In some aspects, the PDGFRα inhibitor is administered to the subject every 12 days. In some aspects, the PDGFRα inhibitor is administered to the subject every 13 days. In some aspects, the PDGFRα inhibitor is administered to the subject once every two weeks. In some aspects, the PDGFRα inhibitor is administered to the subject once every three weeks. In some aspects, the PDGFRα inhibitor is administered to the subject once a month. In some aspects, the PDGFRα inhibitor is administered to the subject once every five weeks. In some aspects, the PDGFRα inhibitor is administered to the subject once every six weeks, In some aspects, the PDGFRα inhibitor is administered to the subject once every seven weeks. In some aspects, the PDGFRα inhibitor is administered to the subject once every two months. In some aspects, the PDGFRα inhibitor is administered to the subject once every nine weeks. In some aspects, the PDGFRα inhibitor is administered to the subject once every 10 weeks. In some aspects, the PDGFRα inhibitor is administered to the subject once every 11 weeks. In some aspects, the PDGFRα inhibitor is administered to the subject once every three months. In some aspects, the PDGFRα inhibitor is administered to the subject once every four months. In some aspects, the PDGFRα inhibitor is administered to the subject once every five months. In some aspects, the PDGFRα inhibitor is administered to the subject once every six months. In some aspects, the PDGFRα inhibitor is administered to the subject once every twelve months. [0280] In some aspects, the intermittent dosing schedule comprises administering to the subject a first dose and a second dose of a PDGFRα inhibitor, wherein the second dose is administered at least one day after administering the first dose. In some aspects, the second dose is administered at least two days after administering the first dose. In some aspects, the second dose is administered at least three days after administering the first dose. In some aspects, the second dose is administered at least four days after administering the first dose. In some aspects, the second dose is administered at least five days after administering the first dose. In some aspects, the second dose is administered at least six days after administering the first dose. In some aspects, the second dose is administered at least seven days after administering the first dose. In some aspects, the second dose is administered at least eight days after administering the first dose. In some aspects, the second dose is administered at least nine days after administering the first dose. In some aspects, the second dose is administered at least 10 days after administering the first dose. In some aspects, the second dose is administered at least 11 days after administering the first dose. In some aspects, the second dose is administered at least 12 days after administering the first dose. In some aspects, the second dose is administered at least 13 days after administering the first dose. In some aspects, the second dose is administered at least two weeks after administering the first dose. In some aspects, the second dose is administered at least three weeks after administering the first dose. In some aspects, the second dose is administered at least one month after administering the first dose. In some aspects, the second dose is administered at least two months after administering the first dose. In some aspects, the second dose is administered at least three months after administering the first dose. In some aspects, the second dose is administered at least four months after administering the first dose. In some aspects, the second dose is administered at least five months after administering the first dose. In some aspects, the second dose is administered at least six months after administering the first dose. In some aspects, the second dose is administered at least 12 months after administering the first dose. [0281] In some aspects, the intermittent dosing schedule comprises administering to the subject a first set of doses and a second set of doses of a PDGFRα inhibitor, wherein the second set of doses is administered at least one day after administering the first set of doses. In some aspects, the first set of doses can be, e.g., one, two, three, or four doses per day, administered over a period of, e.g., one, two, three, or four days. In some aspects, the second set of doses can be, e.g., one, two, three, or four doses per day, administered over a period of, e.g., one, two, three, or four days. [0282] In some aspects, the second set of doses is administered at least two days after administering the first set of doses. In some aspects, the second set of doses is administered at least three days after administering the first set of doses. In some aspects, the second set of doses is administered at least four days after administering the first set of doses. In some aspects, the second set of doses is administered at least five days after administering the first set of doses. In some aspects, the second set of doses is administered at least six days after administering the first set of doses. In some aspects, the second set of doses is administered at least seven days after administering the first set of doses. In some aspects, the second set of doses is administered at least eight days after administering the first set of doses. In some aspects, the second set of doses is administered at least nine days after administering the first set of doses. In some aspects, the second set of doses is administered at least 10 days after administering the first set of doses. In some aspects, the second set of doses is administered at least 11 days after administering the first set of doses. In some aspects, the second set of doses is administered at least 12 days after administering the first set of doses. In some aspects, the second set of doses is administered at least 13 days after administering the first set of doses. In some aspects, the second set of doses is administered at least two weeks after administering the first set of doses. In some aspects, the second set of doses is administered at least three weeks after administering the first set of doses. In some aspects, the second set of doses is administered at least one month after administering the first set of doses. In some aspects, the second set of doses is administered at least two months after administering the first set of doses. In some aspects, the second set of doses is administered at least three months after administering the first set of doses. In some aspects, the second set of doses is administered at least four months after administering the first set of doses. In some aspects, the second set of doses is administered at least five months after administering the first set of doses. In some aspects, the second set of doses is administered at least six months after administering the first set of doses. In some aspects, the second set of doses is administered at least 12 months after administering the first set of doses. [0283] Not to be bound by any one theory, in some aspects, the use of an intermittent dosing schedule allows the OPC population of the treated subject (i.e., subject who previously received an administration of the PDGFRα inhibitor) to sufficiently recover prior to the next dose of the PDGFRα inhibitor. As described herein, OPCs are capable of self-renewal and therefore, under a steady-state condition (i.e., no inhibition of PDGFRα activity), the OPC population of a subject is continuously replenished and maintained. As described and demonstrated herein, when a PDGFRα inhibitor described herein is administered to a subject, the PDGFRα inhibitor induces the differentiation of the OPCs within the subject into oligodendrocytes. In some aspects, this can result in a decrease of the OPC population within the subject, and therefore, an immediate or rapid subsequent administration of a PDGFRα inhibitor would have diminished therapeutic activity (e.g., reduced further generation of oligodendrocytes). Accordingly, in some aspects, an intermittent dosing schedule useful for the present disclosure comprises administering a first dose of a PDGFRα inhibitor and a second dose of a PDGFRα inhibitor, wherein the second dose of the PDGFRα inhibitor is administered to the subject after the OPC population of the subject has sufficiently recovered from the effects of the first dose of the PDGFRα inhibitor. When referring to an OPC population (e.g., of a subject treated with a PDGFRα inhibitor), the expression "sufficiently recovered" means that the number of OPCs within the subject has increased, such that an additional administration of the PDGFRα inhibitor is capable of resulting in additional generation of oligodendrocytes. In some aspects, an OPC population has sufficiently recovered is comparable to that of a reference subject, wherein the reference subject comprises (i) the subject prior to the initial administration of the PDGFRα inhibitor, (ii) a corresponding, normal healthy subject (i.e., does not suffer from a demyelinating disease) who has not received an administration of the PDGFRα inhibitor, or (iii) both (i) and (ii). In some aspects, the number of OPCs within an OPC population that has sufficiently recovered is at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or 100% of the number of OPCs within the OPC population of the reference subject. [0284] Accordingly, where a method provided herein comprises administering a first dose and a second dose of a PDGFRα inhibitor to a subject (e.g., to treat a demyelinating disease), in some aspects, a first dose of the PDGFRα inhibitor is administered to a subject and then the subject's OPC population is assessed, wherein if the subject's OPC population has sufficiently recovered as compared to a reference subject (e.g., as described above), a second dose of the PDGFRα inhibitor is administered to the subject. In some aspects, a second dose of the PDGFRα inhibitor is administered to the subject if the size of the subject's OPC population (e.g., number of OPCs within the OPC population) is at least at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or 100% of the size of the reference subject's OPC population. In some aspects, where an additional dose of the PDGFRα inhibitor is administered to the subject, the additional dose is administered to the subject at the same dosing interval as that used for the first and second doses of the PDGFRα inhibitor. In some aspects, where an additional dose of the PDGFRα inhibitor is administered to the subject, the method comprises assessing the subject's OPC population again after the second administration, wherein if the subject's OPC population has sufficiently recovered as compared to a reference subject (e.g., as described above), the additional dose of the PDGFRα inhibitor is administered to the subject. In some aspects, the additional dose of the PDGFRα inhibitor is administered to the subject if the size of the subject's OPC population (e.g., number of OPCs within the OPC population) is at least at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or 100% of the size of the reference subject's OPC population. As further described elsewhere in the present disclosure, in some aspects, the amount of time required for the subject's OPC population to have sufficiently recovered after an initial administration of a PDGFRα inhibitor is the dosing interval of the PDGFRα inhibitor. [0285] Whether a subject's OPC population has sufficiently recovered (and thus, determine a dosing interval for a PDGFRα inhibitor) can be determined using any suitable method known in the art. For example, in some aspects, a cuprizone animal model can be used. As demonstrated herein, in some aspects, a dosing interval suitable for a PDGFRα inhibitor described herein can be determined by administering a first dose and second dose of the PDGFRα inhibitor to the cuprizone animal model, and then assessing the number of newly generated oligodendrocytes present within the brain of the animals (e.g., by quantifying the number of GPR17+ cells within the brain). If the number of newly generated oligodendrocytes within the brain is the same or increased compared to a reference (e.g., the number of newly generated oligodendrocytes present within the brain of corresponding animal that received a single administration of the PDGFRα inhibitor), then a suitable dosing interval is the time between the administration of the first dose and the second dose of the PDGFRα inhibitor or any time longer than that. If the number of newly generated oligodendrocytes within the brain is reduced compared to a reference (e.g., the number of newly generated oligodendrocytes present within the brain of corresponding animal that received a single administration of the PDGFRα inhibitor), then a suitable dosing interval is longer than the time between the administration of the first dose and the second dose of the PDGFRα inhibitor. [0286] In some aspects, a suitable dosing interval for a PDGFRα inhibitor is directly related to the plasma level of the PDGFRα inhibitor. Accordingly, in some aspects, a method of determining a suitable dosing interval for a PDGFRα inhibitor comprises administering a first dose of the PDGFRα inhibitor to a subject and determining the plasma level of the PDGFRα inhibitor in the subject. In some aspects, where the plasma level of the PDGFRα inhibitor is comparable to that of a reference (e.g., a subject who has not previously received a dose of the PDGFRα inhibitor and/or a subject who has previously received a dose of the PDGFRα inhibitor but the subject's OPC population has sufficiently recovered as described herein), a suitable dosing interval is the time between administering the first dose of the PDGFRα inhibitor and the subject's plasma level of the PDGFRα inhibitor being comparable to that of the reference. In some aspects, where the plasma level of the PDGFRα inhibitor is reduced as compared to a reference (e.g., plasma level of the PDGFRα inhibitor in the subject immediately after, e.g., about 4 hours after, the administration of the initial dose of the PDGFRα inhibitor), a suitable dosing interval is the time between administering the first dose of the PDGFRα inhibitor and the subject's plasma level of the PDGFRα inhibitor reaching a reduced level as compared to the reference. In some aspects, the additional dose of the PDGFRα inhibitor is administered to the subject when the subject's plasma level has decreased by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or about 100% as compared to the reference. In some aspects, the brain to plasma ratio may be assessed in preclinical species using bioanalytical gas or liquid chromatography and mass spectrometry methods. [0287] In some aspects, the plasma level of the PDGFRα inhibitor is related to the half-life of the PDGFRα inhibitor. In some aspects, an intermittent dosing schedule useful for the present disclosure comprises administering two or more doses of a PDGFRα inhibitor at a dosing interval that is longer than the half-life of the PDGFRα inhibitor. In some aspects, the dosing interval is at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 100% longer than the half-life of the PDGFRα inhibitor. In some aspects, the dosing interval is at least 1.5-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, at least 10-fold, at least 15-fold, at least 20-fold, at least about 30-fold, at least 40-fold, or at least 50-fold longer than the half-life of the PDGFRα inhibitor. In some aspects, compared to the half-life of the PDGFRα inhibitor, the dosing interval is at least one day, at least two days, at least three days, at least four days, at least five days, at least six days, at least one week, at least two weeks, at least three weeks, at least one month, at least two months, or at least three months longer. [0288] In some aspects, the methods described herein (e.g., treating a demyelinating disease) can comprise administering an additional therapeutic agent to the subject. For examples, in some aspects, the PDGFRα inhibitors described herein are used in conjunction with an immunomodulatory drug. In some aspects, the PDGFRα inhibitors are used alone in the methods provided herein. [0289] As described herein, "immunomodulatory drug" refers to a therapeutic agent that works by modulating (e.g., increasing and/or decreasing) one or more aspects of an immune response. In some aspects, an immunomodulatory drug useful for the present disclosure is capable of reducing or alleviating the proinflammatory nature of a demyelinating disease (e.g., multiple sclerosis). For instance, in some aspects, the immunomodulatory drug could block the production of proinflammatory mediators, promote the production of anti- inflammatory cytokines (e.g., IL-10 or TGF-β), promote the production of regulatory T cells (Tregs), influence B cells, prevent the entry of immune cells into the brain, or any combination thereof. Non-limiting examples of immunomodulatory drugs that are useful for the present disclosure include: interferon beta-1b (BETASERON®, EXTAVIA®), interferon beta-1a (AVONEX®, REBIF®), peginterferon beta-1a (PLEGRIDY®), alemtuzumab (LEMTRADA®), natalizumab (TYSABRI®), ocrelizumab (OCREVUS®), ofatumumab (KESIMPTA®), glatiramer acetate (COPAXONE®, GLATOPA®), teriflunomide (AUBAGIO®), dimethyl fumarate (TECFIDERA®), monomethyl fumarate (BAFIERTAM®), diroximel fumarate (Vumerity®), and combinations thereof. [0290] Where an additional therapeutic agent (e.g., immunomodulatory drug) is administered to a subject, in some aspects, the additional therapeutic agent is administered to the subject prior to the administration of a PDGFRα inhibitor. Accordingly, in some aspects, any of the methods provided herein (e.g., method of treating a demyelinating disease) comprise administering to the subject a PDGFRα inhibitor, wherein the subject had previously received an additional therapeutic agent (e.g., immunomodulatory drug). In some aspects, the additional therapeutic agent is administered after the administration of a PDGFRα inhibitor. In some aspects, the additional therapeutic agent is administered concurrently with the administration of a PDGFRα inhibitor. [0291] In some aspects, to improve delivery to the CNS, a PDGFRα inhibitor described herein can be administered to a subject in combination with an agent that assists the delivery of the inhibitor to the CNS (e.g., allows the inhibitor to cross the blood-brain barrier). In some aspects, a PDGFRα inhibitor described herein is administered to a subject in combination with a peptide blood-brain barrier (BBB) shuttle, wherein the peptide BBB shuttle enhances the ability of the PDGFRα inhibitor to cross the blood-brain barrier and reach the CNS. Non-limiting examples of such peptide BBB shuttles are provided in Table 2 (below). See, e.g., Oller-Salvia et al., Chem Soc Rev 45: 4690-4707 (2016), and Jafari et al., Expert Opinion on Drug Delivery 16:583-605 (2019). TABLE 2: Peptide BBB Shuttle
Figure imgf000115_0001
Figure imgf000116_0002
Nomenclature for cyclic peptides (&) is adapted to the 3-letter amino acid code from the one described by Spengler et al. Pept. Res., 2005, 65, 550–555 [Dap] stands for diaminopropionic acid. VI. Enumerated Clauses [0292] The present disclosure also provides the following enumerated clauses: [0293] Clause 1. A compound of Formula I:
Figure imgf000116_0001
, [0294] or a pharmaceutically acceptable salt or solvate thereof, wherein: [0295] indicates a single bond or a double bond such that all valences are satisfied; [0296] X1, X2, X3, and X4 are selected from N and CRa, with the proviso that not more than two of X1, X2, X3, and X4 are N; [0297] one of Y1 and Y2 is N and the other of Y1 and Y2 is C; [0298] each Ra is independently selected from H, halo, C1-C4alkyl, and C1-C4alkoxy; [0299] R1 is selected from C1-C4alkyl, C3-C8cycloalkyl, 3-8 membered heterocyclyl, heteroaryl, aryl, and C1-C8alkoxy, all of which can be optionally substituted with one, two, three, four, five, or six substituents selected from halo, hydroxy, oxo, C1-C4alkyl, aminoC1- C4alkyl, hydroxyC1-C4alkyl, C1-C4alkoxy, C1-C4alkoxyC1-C4alkyl, 3-8 membered heterocyclyl, and 3-8 membered heterocyclylC1-C4alkyl, with the proviso that the number of substituents does not exceed the number of substitutable positions; [0300] R2 is selected from cycloalkyl, cycloalkenyl, alkyl, oxoalkylamino, aminoalkylamino, amino, heterocyclyl, heteroaryl, aminoheterocyclyl, heterocyclylamino, and aminoalkylamino, all of which can be optionally substituted with one or more substituents selected from D, halo, hydroxy, oxo, and C1-C4alkyl; [0301] R2 is substituted with one, two, or three R3; [0302] R3 is selected from aryl, heteroaryl, -C(O)R31, -C(O)OR31, -C(O)NR31R32, - S(O)2NR31R32, -S(O)(NR33)R31, -S(O)2R31, -S(O)(NR33)NR31R32, -C(S)NR31R32, C3- C8cycloalkyl, 3-8 membered heterocyclyl, and C1-C4alkyl, all of which can be optionally substituted with one, two, three, four, or five R30; [0303] each R30 is independently selected from D, halo, aryl, -OR300, -NR300R303, -S(O)rR300, -C(O)R300, -C(=CR34R35)R300, and
Figure imgf000117_0001
; [0304] r is selected from 0, 1, and 2; [0305] each R300 is independently selected from C1-C6alkyl, C3-C7cycloalkyl, aryl, heteroaryl, 3-8 membered heterocyclyl, and 3-8 membered heterocyclylaryl, all of which can be optionally substituted with one, two, three, four, or five substituents selected from D, halo, hydroxy, amino, alkylamino, cyano, C1-C4alkyl, C1-C4haloalkyl, and C1-C4alkoxy; [0306] each R301 is independently selected from H, halo, and C1-C4alkyl; [0307] each R302 is independently selected from H, F, hydroxyl, amino, alkylamino, oxo, and C1-C4alkyoxy; [0308] each R303 is independently selected from H and C1-C4alkyl; [0309] n, o, and p are each independently selected from 0, 1, 2, 3, and 4; [0310] each R31 is independently selected from C1-C8alkyl, arylC1-C4alkyl, heteroarylC1- C4alkyl, heterocyclyl, heterocyclylC1-C4alkyl, cycloalkyl, and cycloalkylC1-C4alkyl, all of which can be optionally substituted with one, two, three, four, or five substituents selected from D, halo, cyano, hydroxy, amino, -OCF3, C1-C4alkyl, C1-C4haloalkyl, C1-C4alkoxy, hydroxyC1-C4alkyl, -S(O)2NR304R305, -C(O)OR304R305, -C(O)NR304R305, and - NR304C(O)R305; [0311] each R304 and R305 is independently selected from H and C1-C4alkyl; [0312] each R32 is independently selected from H and C1-C4alkyl; or [0313] R31 and R32 together with the atom to which they are connected to form a 5-8 membered heterocycyl, optionally substituted with one, two, three, four, or five substituents selected from D, halo, cyano, C1-C4alkyl, C1-C4haloalkyl, and -C(O)NR34R35; and [0314] each R34 and R35 is independently selected from H, C1-C4alkyl, and C1-C4haloalkyl; and [0315] each R33 is independently selected from H, C1-C4alkyl, C1-C4haloalkyl, and – C(O)R34; or [0316] R31 and R33 together with the atoms to which they are connected form a 4-8 membered heterocycyl. [0317] Clause 2. The compound of clause 1, or a pharmaceutically acceptable salt or solvate thereof, wherein Y1 is N and Y2 is C. [0318] Clause 3. The compound of clause 1, or a pharmaceutically acceptable salt or solvate thereof, wherein Y1 is C and Y2 is N. [0319] Clause 4. The compound of any one of clauses 1 to 3, or a pharmaceutically acceptable salt or solvate thereof, wherein X1 is N, X2 is CRa, X3 is CRa, and X4 is CRa. [0320] Clause 5. The compound of any one of clauses 1 to 3, or a pharmaceutically acceptable salt or solvate thereof, wherein X1 is CRa, X2 is N, X3 is CRa, and X4 is CRa. [0321] Clause 6. The compound of any one of clauses 1 to 3, or a pharmaceutically acceptable salt or solvate thereof, wherein X1 is CRa, X2 is CRa, X3 is N, and X4 is CRa. [0322] Clause 7. The compound of any one of clauses 1 to 3, or a pharmaceutically acceptable salt or solvate thereof, wherein X1 is CRa, X2 is CRa, X3 is CRa, and X4 is N. [0323] Clause 8. The compound of any one of clauses 1 to 3, or a pharmaceutically acceptable salt or solvate thereof, wherein X1 is CRa, X2 is CRa, X3 is CRa, and X4 is CRa. [0324] Clause 9. The compound of any one of clauses 1 to 8, or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is 5- or 6-membered heteroaryl. [0325] Clause 10. The compound of any one of clauses 1 to 9, or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is an optionally substituted pyrazolyl. [0326] Clause 11. The compound of any one of clauses 1 to 10, or a pharmaceutically acceptable salt or solvate thereof, wherein: [0327] R1 is selected from:
Figure imgf000119_0001
, , nd [0328] R10 is selected from H, C1-C4alkyl, C1-C4alkoxy, aminoC1-C4alkyl, hydroxyC1- C4alkyl, and C1-C4alkylsulfonyl, wherein the C1-C4alkyl, C1-C4alkoxy, aminoC1-C4alkyl, hydroxyC1-C4alkyl, and C1-C4alkylsulfonyl can be optionally substituted by one or more substituents selected from hydroxyl, C1-C4alkoxy, NR10aR10b, halo, and deuterium, wherein R10a and R10b are selected from hydrogen and C1-C4alkyl, or wherein R10a and R10b taken together with the nitrogen atom to which they are attached form a 4- to 8-membered ring. [0329] Clause 12. The compound of clause 11, or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is
Figure imgf000119_0002
. [0330] Clause 13. The compound of clause 11 or 12, or a pharmaceutically acceptable salt or solvate thereof, wherein R10 is CH3. [0331] Clause 14. The compound of any one of clauses 1 to 13, or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is heterocyclyl optionally substituted with C1- C4alkyl or oxo. [0332] Clause 15. The compound of any one of clauses 1 to 14, or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is substituted with one R3. [0333] Clause 16. The compound of any one of clauses 1 to 15, or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is selected from: ,
Figure imgf000120_0001
,
Figure imgf000121_0001
, , , , [0334] indicates a single bond or a double bond such that all valences are satisfied; [0335] m is selected from 0, 1, 2, 3, 4, 5, and 6; and [0336] Z1, Z2, and Z3 are selected from N and CRa. [0337] Clause 17. The compound of clause 16, or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is selected from:
Figure imgf000122_0001
, , , , . [0338] Clause 18. The compound of any one of clauses 1 to 17, or a pharmaceutically acceptable salt or solvate thereof, having Formula Ia:
Figure imgf000123_0001
[0339] wherein a and b are each independently selected from 1, 2, and 3; and [0340] Q is selected from -CH- and -N-, with the proviso that if Q is -N-, a and b are not 1. [0341] Clause 19. The compound of any one of clauses 1 to 18, or a pharmaceutically acceptable salt or solvate thereof, having Formula II:
Figure imgf000123_0002
. [0342] Clause 20. The compound of any one of clauses 1 to 19, or a pharmaceutically acceptable salt or solvate thereof, having Formula IIa:
Figure imgf000124_0001
. [0343] Clause 21. The compound of any one of clauses 1 to 20, or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is heteroaryl optionally substituted with one, two, three, four, or five R30. [0344] Clause 22. The compound of any one of clauses 1 to 21, or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is selected from: ,
Figure imgf000124_0002
, , , , , , ,
Figure imgf000125_0001
, [0345] wherein A1 is selected from O, S, and N and R36 is selected from hydrogen, optionally substituted C1-C6alkyl, and optionally substituted C1-C6alkylaryl. [0346] Clause 23. The compound of any one of clauses 1 to 22, or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is selected from: ,
Figure imgf000126_0001
[0347] Clause 24. The compound of any of clauses 1 to 23, or a pharmaceutically acceptable salt or solvate thereof, wherein R30 is:
Figure imgf000127_0001
. [0348] Clause 25. The compound of clause 24, or a pharmaceutically acceptable salt or solvate thereof, wherein R300 is selected from: , ,
Figure imgf000127_0002
, , , , , . [0349] Clause 26. The compound of clause 25, or a pharmaceutically acceptable salt or solvate thereof, wherein R300 is:
Figure imgf000127_0003
. [0350] Clause 27. The compound of any one of clauses 1 to 26, or a pharmaceutically acceptable salt or solvate thereof, wherein R301 is H and R302 is H or CH3. [0351] Clause 28. The compound of any of clauses 1 to 22, or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is selected from: nd
Figure imgf000128_0001
. [0352] Clause 29. The compound of clause 28, or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is selected from:
Figure imgf000128_0002
, , . [0353] Clause 30. The compound of clause 29, or a pharmaceutically acceptable salt or solvate thereof, wherein R32 is H. [0354] Clause 31. The compound of any one of clauses 28 to 30, or a pharmaceutically acceptable salt or solvate thereof, wherein R31 is selected from:
Figure imgf000129_0001
, , , ,
Figure imgf000130_0001
, , , , [0355] wherein R31a is selected from H, D, alkylamino, C1-C4alkyl, and -CF3, and [0356] R31b is selected from H, D, halo, hydroxy, amino, alkylamino, C1-C4alkyl, -CF3, and -OCF3. [0357] Clause 32. The compound of clause 1, or a pharmaceutically acceptable salt or solvate thereof, selected from any one of compounds 156, 250, 275 and 326-406 of Table 1. [0358] Clause 33. The compound of any one of clauses 1 to 32, which can exhibit one or more of the following properties: (i) promote the differentiation of an OPC into an oligodendrocyte, (ii) promote the expression of a protein associated with oligodendrocyte differentiation and/or myelination (e.g., G-protein coupled receptor 17 (GPR17), myelin basic protein (MBP), ASPA, GST-pi, CC1, myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, or a combination thereof), (iii) promote the myelination of an axon, (iv) promote the remyelination of a demyelinated axon, (v) inhibit PDGFRα kinase activity, (vi) achieve a brain to plasma ratio of greater than 0.1 when systemically administered to a subject, and (vii) any combination thereof. [0359] Clause 34. The compound of clause 33, which can inhibit PDGFRα kinase activity. [0360] Clause 35. The compound of clause 34, which can inhibit PDGFRα kinase activity with an IC50 of less than 500 nM, less than 400 nM, less than 300 nM, less than 200 nM, less than 100 nM, less than 75 nM, less than less than 50 nM, less than 40 nM, less than 30 nM, less than 20 nM, less than 10 nM, or less than 5 nM. [0361] Clause 36. The compound of clause 35, wherein the IC50 of the PDGFRα inhibitor is determined using an enzymatic PDGFRα kinase assay (e.g., Promega kinase assay described in Example 131). [0362] Clause 37. The compound of clause 36, wherein the enzymatic PDGFRα kinase assay comprises 20 ng of purified PDGFRα protein, 150 µM of ATP and 1 µg of substrate, Poly (Glu4Tyr1) in a volume of 15 µl. [0363] Clause 38. A pharmaceutical composition comprising the compound of any one of clauses 1 to 37, or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable excipient. [0364] Clause 39. A kit comprising the compound of any one of clauses 1 to 37, or a pharmaceutically acceptable salt or solvate thereof, or the composition of clause 38, and instructions for use. [0365] Clause 40. A compound of any one of clauses 1 to 37, or the pharmaceutical composition of clause 38, for use in therapy. [0366] Clause 41. A method of treating a demyelinating disease in a subject in need thereof comprising administering to the subject a therapeutically effective amount of the compound of any one of clauses 1 to 37 or the pharmaceutical composition of clause 38. [0367] Clause 42. A method of improving a subject's performance in a test for assessing one or more symptoms associated with a demyelinating disease, comprising administering to the subject a therapeutically effective amount of the compound of any one of clauses 1 to 37 or the pharmaceutical composition of clause 38, wherein after the administration, the subject's performance in the test is improved as compared to a reference subject (e.g., the subject prior to the administration). [0368] Clause 43. The method of clause 42, wherein the test is selected from a visual evoked potential (VEP) test, a multifocal visual evoked potential (mfVEP) test, a low contrast visual acuity (LC-VA) test, a magnetic resonance imaging (MRI) (e.g., magnetic transfer resonance, myelin water fraction (MWF), and quantitative susceptibility mapping (QSM)), an electromyography (EMG), a nerve conduction velocity (NCV) test, an Extended Disability Status Scale (EDSS), a timed walk test (e.g., timed 25-foot walk), a Nine-Hole Peg Test (9HPT), an ocular coherence tomograph (OCT), a quality of life measure test (e.g., Multiple Sclerosis Quality of Life-54 and Vision-Related Quality of Life), cognitive assessment (e.g., Montreal Cognitive Assessment), or combinations thereof. [0369] Clause 44. The method of any one of clauses 41 to 43, wherein the demyelinating disease comprises an acute disseminated encephalomyelitis (ADEM), acute hemorrhagic leukoencephalitis, acute transverse myelitis, adrenoleukodystrophy, adrenomyeloneuropathy, Alexander disease, Alzheimer's disease, aminoacidurias, amyotrophic lateral sclerosis, anti-MAG peripheral neuropathy, anti-MOG associated spectrum, Balo concentric sclerosis, brain injury, CAMFAK syndrome, Canavan disease, carbon monoxide toxicity, central pontine myelinolysis, cerebral hypoxia, cerebral ischemia, Charcot–Marie–Tooth disease, chronic inflammatory demyelinating polyneuropathy, chronic traumatic encephalopathy, clinically isolated syndrome (CIS), congenital cataract, copper deficiency associated condition, delayed post-hypoxic leukoencephalopathy, diffuse cerebral sclerosis of Schilder, diffuse myelinoclastic sclerosis, extrapontine myelinolysis Gaucher disease, Guillain–Barré syndrome, hereditary neuropathy, hereditary neuropathy with liability to pressure palsy, HTLV-1–associated myelopathy, Hurler syndrome, hypomyelination, hypoxic brain injury, Krabbe disease, Leber hereditary optic atrophy and related mitochondrial disorders, leukodystrophic disorders, Marchiafava-Bignami disease, metachromatic leukodystrophy, multiple sclerosis (e.g., primary progressive multiple sclerosis (PPMS), relapsing-remitting multiple sclerosis (RRMS), secondary progressive multiple sclerosis (SPMS), progressive relapsing multiple sclerosis, Marburg multiple sclerosis, tumefactive multiple sclerosis, and optic- spinal multiple sclerosis), multiple system atrophy, myelinoclastic disorders, myelopathy, nerve injury, neuromyelitis optica (NMO), Niemann-Pick disease, optic neuropathy, optic neuritis (e.g., acute optic neuritis and chronic relapsing inflammatory optic neuritis (CRION)), osmotic demyelination syndrome, Parkinson's disease, Pelizaeus‐Merzbacher disease, peripheral neuropathy, phenylketonuria, progressive inflammatory neuropathy, progressive multifocal leukoencephalopathy, progressive subcortical ischemic demyelination, reperfusion injury, Schilder disease, solitary sclerosis, spinal cord injury, subacute sclerosing panencephalitis, Tabes dorsalis, Tay-Sachs disease, transverse myelitis, traumatic brain injury, tropical spastic paraparesis, vitamin B12 deficiency, cerebral palsy, or a combination thereof. [0370] Clause 45. The method of any one of clauses 41 to 44, wherein the demyelinating disease is characterized by demyelination of one or more cells within the CNS of the subject. [0371] Clause 46. The method of any one of clauses 41 to 45, wherein the demyelinating disease is multiple sclerosis. [0372] Clause 47. The method of clause 46, wherein the multiple sclerosis comprises a clinically isolated syndrome ("CIS"), relapsing-remitting MS ("RRMS"), secondary progressive MS ("SPMS"), primary progressive MS ("PPMS"), optic neuritis or transverse myelitis. [0373] Clause 48. The method of any one of clauses 41 to 45, wherein the demyelinating disease is an optic neuritis. [0374] Clause 49. The method of any one of clauses 41 to 48, wherein treating the demyelinating disease comprises reducing one or more symptoms associated with the demyelinating disease. [0375] Clause 50. The method of clause 49, wherein the one or more symptoms comprise fatigue, dizziness, malaise, elevated fever and high body temperature, extreme sensitivity to cold in the hands and feet, weakness and stiffness in muscles and joints, weight changes, digestive or gastrointestinal problems, low blood pressure, high blood pressure, irritability, anxiety, depression, impaired vision (e.g., blurred vision, double vision, reduction in low contrast visual acuity (LC-VA)), ataxia, clonus, spasms, dysarthria, weakness, clumsiness, hand paralysis, hemiparesis, genital anesthesia, sexual dysfunction, incoordination, paresthesias, ocular paralysis, impaired muscle coordination, loss of sensation, tingling, numbness, pain, neurological symptoms, impaired cognition, unsteady gait, balance problems, dizziness, spastic paraparesis, incontinence, hearing problems, speech problems, loss of olfaction, agusia, or combinations thereof. [0376] Clause 51. A method of promoting the myelination of an axon in a subject in need thereof, the method comprising administering to the subject an effective amount of the compound of any one of clauses 1 to 37 or the pharmaceutical composition of clause 38. [0377] Clause 52. The method of clause 51, wherein promoting the myelination of an axon results in an increase in the expression of one or more of the following markers within the subject: myelin basic protein (MBP) Myelin Oligodendrocyte Glycoprotein (MOG), Oligodendrocyte Specific Protein/Claudin-11, CNPase, or any combination thereof. [0378] Clause 53. The method of clause 51 or 52, wherein the myelination of an axon can be determined by visualizing and/or quantifying the expression of one or more of the following markers: myelin basic protein (MBP) Myelin Oligodendrocyte Glycoprotein (MOG), Oligodendrocyte Specific Protein/Claudin-11, CNPase, or any combination thereof. [0379] Clause 54. A method of promoting the remyelination of a demyelinated axon in a subject in need thereof, the method comprising administering to the subject an effective amount of the compound of any one of clauses 1 to 37 or the pharmaceutical composition of clause 38. [0380] Clause 55. The method of clause 54, wherein promoting the remyelination of a demyelinated axon results in an increase in the expression of one or more of the following markers within the subject: myelin basic protein (MBP) Myelin Oligodendrocyte Glycoprotein (MOG), Oligodendrocyte Specific Protein/Claudin-11, CNPase, or any combination thereof [0381] Clause 56. The method of clause 54 or 55, wherein the remyelination of a demyelinated axon can be determined by visualizing and/or quantifying the expression of one or more of the following markers: myelin basic protein (MBP) Myelin Oligodendrocyte Glycoprotein (MOG), Oligodendrocyte Specific Protein/Claudin-11, CNPase, or any combination thereof. [0382] Clause 57. A method of reducing the demyelination of a myelinated neuronal axon in a subject in need thereof, the method comprising administering to the subject an effective amount of the compound of any one of clauses 1 to 37 or the pharmaceutical composition of clause 38. [0383] Clause 58. The method of clause 57, wherein reducing the demyelination of a myelinated neuronal axon results in an increase in the expression of one or more of the following markers: myelin basic protein (MBP) Myelin Oligodendrocyte Glycoprotein (MOG), Oligodendrocyte Specific Protein/Claudin-11, CNPase, or any combination thereof. [0384] Clause 59. The method of clause 57 or 58, wherein the reduction in the demyelination of a myelinated neuronal axon can be determined by visualizing and/or quantifying the expression of one or more of the following markers: myelin basic protein (MBP) Myelin Oligodendrocyte Glycoprotein (MOG), Oligodendrocyte Specific Protein/Claudin-11, CNPase, or any combination thereof. [0385] Clause 60. A method of activating an oligodendrocyte progenitor cell (OPC) within the central nervous system (CNS) of a subject in need thereof, the method comprising administering to the subject an effective amount of the compound of any one of clauses 1 to 37 or the pharmaceutical composition of clause 38. [0386] Clause 61. A method according to any one of clauses 51 to 60, wherein the subject has, or is at risk of developing a demyelinating disease, for example a disease according to any one of clauses 44 to 50. [0387] Clause 62. A method according to any one of clauses 51 to 60, wherein the method is a method of treating or preventing a demyelinating disease, for example a disease according to any one of clauses 44 to 50. [0388] Clause 63. The method of any one of clauses 41 to 62, wherein the compound or the pharmaceutical composition is administered to the subject once. [0389] Clause 64. The method of any one of clauses 41 to 62, wherein the compound or the pharmaceutical composition is administered to the subject more than once using intermittent dosing. [0390] Clause 65. The method of clause 64, wherein the intermittent dosing comprises administering the compound or the pharmaceutical composition to the subject every other day, every three days, every four days, every five days, every six days, once a week, every eight days, every nine days, every 10 days, every 11 days, every 12 days, every 13 days, once every two weeks, once every three weeks, once a month, once every two months, once every three months, once every four months, once every five months, once every six months, or once every twelve months. [0391] Clause 66. The method of clause 64 or 65, wherein the intermittent dosing comprises administering to the subject a first dose and a second dose of the compound or the pharmaceutical composition, wherein the second dose is administered at least one day, at least two days, at least three days, at least four days, at least five days, at least six days, at least seven days, at least eight days, at least nine days, at least 10 days, at least 11 days, at least 12 days, at least 13 days, at least two weeks, at least three weeks, at least one month, at least two months, at least three months, at least four months, at least five months, at least six months, or at least 12 months after administering the first dose. [0392] Clause 67. The method of clause 66, wherein the second dose is administered to the subject one day, two days, three days, four days, five days, six days, seven days, eight days, nine days, 10 days, 11 days, 12 days, 13 days, two weeks, three weeks, one month, two months, three months, four months, five months, six months, or 12 months after administering the first dose. [0393] Clause 68. The method of any one of clauses 41 to 67, wherein after the administration, the compound or the pharmaceutical composition can achieve a brain to plasma ratio of greater than 0.1, greater than 0.2, greater than 0.3, greater than 0.4, greater than 0.5, greater than 0.6, greater than 0.7, greater than 0.8, greater than 0.9, greater than 1.0, greater than 1.1, greater than 1.2, greater than 1.3, greater than 1.4, greater than 1.5, greater than 1.6, greater than 1.7, greater than 1.8, greater than 1.9, or greater than 2.0. [0394] Clause 69. The method of any one of clauses 41 to 68, further comprising administering to the subject an additional therapeutic agent. [0395] Clause 70. The method of clause 69, wherein the additional therapeutic agent comprises a standard care of treatment. [0396] Clause 71. The method of clause 69 or 70, wherein the additional therapeutic agent comprises an immunomodulatory agent. [0397] Clause 72. The method of clause 71, wherein the additional therapeutic agent is selected from interferon beta-1b, interferon beta-1a, peginterferon beta-1a, alemtuzumab, natalizumab, ocrelizumab, ofatumumab, glatiramer acetate, teriflunomide, dimethyl fumarate, monomethyl fumarate, diroximel fumarate, fingolimod hydrochloride, siponimod fumaric acid, ozanimod hydrochloride, BTK inhibitor, or a pharmaceutically acceptable salt thereof [0398] Clause 73. The method of any one of clauses 69 to 72, wherein the additional therapeutic agent is administered to the subject prior to, concurrently, or after the administration of the compound or the pharmaceutical composition. [0399] Clause 74. A method of inducing the differentiation of an oligodendrocyte progenitor cell (OPC) into an oligodendrocyte, the method comprising contacting the OPC with an effective amount of the compound of any one of clauses 1 to 37 or the pharmaceutical composition of clause 38. [0400] Clause 75. The method of clause 74, wherein inducing the differentiation of the OPC into an oligodendrocyte results in an increase in the expression of the following markers in the subject: GPR17, MBP, ASPA, GST-pi, CC1, myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, or a combination thereof. [0401] Clause 76. The method of clause 74 or 75, wherein the differentiation of the OPC into an oligodendrocyte is measured by determining the expression of GPR17, MBP, ASPA, GST-pi, CC1, myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte- specific protein/claudin-11, CNPase, or a combination thereof. [0402] Clause 77. A method of inhibiting PDGFRα activity in a cell, the method comprising contacting the cell with an effective amount of the compound of any one of clauses 1 to 37 or the pharmaceutical composition of clause 38. [0403] Clause 78. The method of clause 77, wherein the inhibition of the PDGFRα activity is measured by one or more of the following: an in vitro OPC differentiation assay (e.g., as described in Example 132), a cuprizone model for demyelination (e.g., as described in Example 134), an in vivo OPC differentiation assay (e.g., as described in Example 134), an enzymatic PDGFRα kinase assay (e.g., as described in Example 131), or any combination thereof. [0404] Clause 79. The method of any one of clauses 74 to 78, wherein the contacting occurs ex vivo or in vivo. [0405] Clause 80. The method of any one of clauses 74 to 79, wherein the method is a method of treating by a therapy. [0406] Clause 81. A method of treating a relapsing form of multiple sclerosis in a subject in need thereof, the method comprising administering to the subject an effective amount of the compound of any one of clauses 1 to 37 or the pharmaceutical composition of clause 38. [0407] Clause 82. The method of clause 81, wherein inducing the differentiation of the OPC into an oligodendrocyte results in an increase in the expression of the following marker in the subject: GPR17, MBP, ASPA, GST-pi, CC1, myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, or a combination thereof. [0408] Clause 83. The method of clause 81 or 82, wherein the differentiation of the OPC into an oligodendrocyte is measured by determining the expression of GPR17, MBP, ASPA, GST-pi, CC1, myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte- specific protein/claudin-11, CNPase, or a combination thereof. [0409] Clause 84. The method of any one of clauses 81 to 83, wherein the relapsing form of multiple sclerosis comprises a clinically isolated syndrome ("CIS"), relapsing- remitting MS ("RRMS"), secondary progressive MS ("SPMS"), primary progressive MS ("PPMS"), or transverse myelitis. [0410] Clause 85. The compound of any one of clauses 1 to 37 or the pharmaceutical composition of clause 38, for use in a method of one or more of the following: (i) promote the differentiation of an OPC into an oligodendrocyte, (ii) promote the expression of a protein associated with oligodendrocyte differentiation and/or myelination (e.g., G-protein coupled receptor 17, myelin basic protein (MBP), ASPA, GST-pi, CC1, myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, or a combination thereof), (iii) promote the myelination of an axon, (iv) promote the remyelination of a demyelinated axon, (v) inhibit PDGFRα kinase activity, (vi) achieve a brain to plasma ratio of greater than 0.1 when systemically administered to a subject, and (vii) any combination thereof. [0411] Clause 86. A method of treating a PDGF-associated tumor in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound of any one of clauses 1 to 37 or the pharmaceutical composition of clause 38, wherein after the administration, PDGFRα activity is reduced in the subject. [0412] Clause 87. The method of clause 86, wherein the PDGF-associated tumor comprises an oligodendroglioma. [0413] Clause 88. The method of clause 86 or 87, wherein the method is a method of treatment by therapy. [0414] The specification is most thoroughly understood in light of the teachings of the references cited within the specification. The embodiments within the specification provide an illustration of embodiments and should not be construed to limit the scope. The skilled artisan readily recognizes that many other embodiments are encompassed. All publications and patents cited in this disclosure are incorporated by reference in their entirety. To the extent the material incorporated by reference contradicts or is inconsistent with this specification, the specification will supersede any such material. The citation of any references herein is not an admission that such references are prior art. [0415] Unless otherwise indicated, all numbers expressing quantities of ingredients, reaction conditions, and so forth used in the specification, including claims, are to be understood as being modified in all instances by the term "about." Accordingly, unless otherwise indicated to the contrary, the numerical parameters are approximations and can vary depending upon the desired properties sought to be obtained. At the very least, and not as an attempt to limit the application of the doctrine of equivalents to the scope of the claims, each numerical parameter should be construed in light of the number of significant digits and ordinary rounding approaches. [0416] The following examples are offered by way of illustration and not by way of limitation. The contents of all references cited throughout this application are expressly incorporated herein by reference. EXAMPLES [0417] As further detailed below, the present disclosure demonstrates that compounds that inhibit PDGFRα kinase activity in OPCs can induce oligodendrocyte differentiation (e.g., in vitro and in vivo), and that the effect is likely specific to the inhibition of PDGFRα kinase activity. Furthermore, the present disclosure demonstrates that such compounds can induce remyelination in an animal model of demyelination. [0418] Table 3 below defines the different abbreviations used in the below examples and else wherein the present disclosure. Table 3. Abbreviations
Figure imgf000140_0001
Figure imgf000141_0001
e
Figure imgf000142_0001
[0419] In general, for compounds having stereoisomers that were separated, e.g., using chiral chromatography, the absolute stereochemistry of each compound was arbitrarily assigned. Preparation of Compounds for Synthesis of Exemplary PDGFRα Inhibitors [0420] To synthesize the exemplary compounds disclosed herein, intermediate compounds were first prepared as described below. Synthesis of 3-bromo-7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazine (Compound S3)
Figure imgf000143_0001
Step 1. Preparation of 5-(1-methyl-1H-pyrazol-4-yl)pyridazin-3-ol
Figure imgf000143_0002
[0421] To a solution of 5-chloropyridazin-3-ol (1.00 g, 7.66 mmol) and 1-methylpyrazole- 4-boronic acid pinacol ester (1.59 g, 7.66 mmol) in dioxane (18 mL) was added 2M Na2CO3 (15.3 mL, 30.6 mmol). The biphasic mixture was sparged for 10 min with argon then Pd(dppf)Cl2 (0.28 g, 0.38 mmol) was added. The reaction was sparged with argon for 10 min, then heated to 75 °C for 6 h under an argon. The reaction was cooled to rt and stirred overnight. The mixture was concentrated under reduced pressure then water (30 mL) was added. The aqueous mixture was extracted with 10% EtOH in EtOAc (4 x 50 mL) and 50% EtOAc in THF (3 x 50 mL). Brine (30 mL) was added to the aqueous layer, then extracted with 10% EtOH in EtOAc (3 x 50 mL). The organic layers were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with 0-10% MeOH in DCM to afford 0.93 g (69%) of 5-(1-methyl-1H-pyrazol-4-yl)pyridazin-3-ol as a pale yellow solid. MS (ESI) m/z [M+H]+ calcd. for C8H9N4O, 171.1; found, 171.0. Step 2. Preparation of 3-chloro-5-(1-methyl-1H-pyrazol-4-yl)pyridazine
Figure imgf000144_0001
[0422] 5-(1-methyl-1H-pyrazol-4-yl)pyridazin-3-ol (6.0 g, 30 mmol, as prepared in the previous step) was treated with POCl3 (75 mL, 0.80 mol). The suspension was warmed at 80 °C for 1.5 h. The reaction was cooled to rt and the excess POCl3 was removed in vacuo. Toluene was added to the residue then the solvent was removed under reduced pressure. This was repeated twice more. The residue was slowly treated with ice water (350 mL) then solid NaHCO3 was slowly added to adjust the pH to 8. The reaction was stirred for 30 min, then extracted with 10% MeOH in DCM (2 x 200 mL). The aqueous layer was diluted with water (100 mL) and extracted with 10% MeOH in DCM (2 x 200 mL) and 10% trifluoroethanol in DCM (3 x 200 mL). The organic layers were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by silica gel chromatography eluting with 0-100% of 5% MeOH in DCM solution/DCM to afford 5.14 g (87%) of 3-chloro-5-(1-methyl-1H-pyrazol-4- yl)pyridazine as a white solid. MS (ESI) m/z [M+H]+ calcd. for C8H8ClN4, 195.0; found, 195.0. Step 3. Preparation of N-(2,2-dimethoxyethyl)-5-(1-methyl-1H-pyrazol-4-yl)pyridazin-3- amine
Figure imgf000144_0002
[0423] A suspension of 3-chloro-5-(1-methyl-1H-pyrazol-4-yl)pyridazine (5.10 g, 26.2 mmol, as prepared in the previous step) and aminoacetaldehyde dimethyl acetal (29 mL, 0.27 mol) was warmed in an oil bath at 120 °C for 20 h, then additional aminoacetaldehyde dimethyl acetal (15 mL, 0.14 mol) was added. The reaction was stirred at 120 °C for 27 h, then cooled to rt, and concentrated under reduced pressure. The residue was partitioned between sat. aqueous NaHCO3 (100 mL) and 10% MeOH in DCM (100 mL). The aqueous layer was washed with 10% MeOH in DCM (4 x 100 mL). The organic layers were combined, dried with anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with 0-100% of 10% MeOH in DCM solution/ DCM to afford 6.56 g (95%) of N-(2,2- dimethoxyethyl)-5-(1-methyl-1H-pyrazol-4-yl)pyridazin-3-amine as a tan solid. MS (ESI) m/z [M+H]+ calcd. for C12H18N5O2, 264.2; found, 264.0. Step 4.7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazine
Figure imgf000145_0001
[0424] Conc. H2SO4 (52 mL) was cautiously added to N-(2,2-dimethoxyethyl)-5-(1- methyl-1H-pyrazol-4-yl)pyridazin-3-amine (15.7 g, 59.6 mmol, as prepared in the previous step) while stirring. After 15 minutes, the reaction was cooled in an ice bath and 1M NaOH (100 mL) was carefully added to the reaction. 50% NaOH (approx. 210 mL) was slowly added to the cold reaction until basic (pH 12). The pH was adjusted to pH 8 with slow addition of 1M H2SO4. The reaction was partitioned between DCM (400 mL) and water (1200 mL). The aqueous layer was washed with DCM (2 x 300 mL). The aqueous layer containing an emulsion was filtered through a pad of Celite. The filter pad was washed with DCM which was then used to wash the aqueous layer (2 x 300 mL). The filter pad was then washed with 10% MeOH in DCM that was in turn used to wash the aqueous layer (4 x 300 mL). The aqueous layer was washed again with 10% MeOH in DCM (4 x 100 mL) or until no product was evident in the aqueous layer by HPLC. The organic layers were combined, dried with anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure to afford 10.5 g (89%) of 7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazine as a tan solid. MS (ESI) m/z [M+H]+ calcd. for C10H10N5, 200.1; found, 200.0. Step 5. Preparation of 3-bromo-7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazine (Compound S3)
Figure imgf000145_0002
[0425] A solution of 7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazine (2.98 g, 15.0 mmol, as prepared in the previous step) in DMF (85.0 mL) was cooled in an ice bath then NBS (2.93 g, 16.4 mmol) was added. The reaction was stirred at 0°C for 2.5 h, then was poured into a solution of sat. aqueous Na2S2O3 (28.6 mL, 112 mmol) and sat. aqueous NaHCO3 (66.1 mL, 68.3 mmol). The reaction was diluted with water (120 mL). The ice bath was removed, and the reaction allowed to warm to rt. After stirring 1 h, the reaction was further diluted with water (270 mL). The solids were filtered off, washed with water (2 x 45 mL) and dried under a nitrogen press to afford 3.80 g (91%) of 3-bromo-7-(1- methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazine (Compound S3) as a yellow solid. (400 MHz, DMSO-d6) δ (ppm) 9.00 (d, J=1.96 Hz, 1H) 8.45 (s, 1H) 8.33 (d, J=2.08 Hz, 1H) 8.17 (s, 1H) 7.85 (s, 1H) 3.90 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C10H9BrN5, 278.0; found, 277.9; HPLC purity: 210 nm: 97.9%; 254 nm: 97.5%. Synthesis of O-(mesitylsulfonyl)hydroxylamine (Compound S4)
Figure imgf000146_0001
[0426] To a solution of ethyl N-hydroxyacetimidate (40.0 g, 329.2 mmol) and 2,4,6- trimethylbenzenesulfonyl chloride (40.0 g, 182.8 mmol) in DMF (300 mL) was added TEA (27.7 g, 274.3 mmol) dropwise at rt then the mixture was stirred at rt for 2 h. The reaction was poured into ice water (900 mL) then the mixture was filtered, and the filter cake was washed with H2O (3 x 200 mL) and dried under reduced pressure. The solids were dissolved in dioxane (26 mL), then solution was cooled to 0°C, then HClO4 (16 mL) was added dropwise at 0°C. After completion of addition, the mixture was stirred at 0°C for 30 min, then poured into ice water (400 mL). The mixture was filtered, then the filter cake was washed with H2O (3 x 200 mL). The solids were dissolved in DCM (200 mL) to afford a DCM solution of O-(mesitylsulfonyl)hydroxylamine (Compound S4). Synthesis of 3-bromo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrazine (Compound S5)
Figure imgf000146_0002
Step 1. Synthesis of 1-amino-3-bromopyrazin-1-ium 2,4,6-trimethylbenzenesulfonate
Figure imgf000147_0001
[0427] To a solution of 2-bromopyrazine (22.0 g, 138.3 mmol) in DCM (154 mL) was added a solution of O-(mesitylsulfonyl)hydroxylamine (Compound S4) (38.6 g, 179.8 mmol) in DCM (308 mL) dropwise at rt, then the mixture was stirred at rt overnight. The reaction was concentrated under reduced pressure then the residue was triturated with MTBE (100 mL) and dried under reduced pressure to afford 34.0 g (66%) of 1-amino-3- bromopyrazin-1-ium 2,4,6-trimethylbenzenesulfonate as a black solid. Step 2. Synthesis of ethyl 6-bromopyrazolo[1,5-a]pyrazine-3-carboxylate
Figure imgf000147_0002
[0428] To a solution of K2CO3 (12.4 g, 97.4 mmol) and ethyl propiolate (27.0 g, 108.2 mmol) in DMF (270 mL) was added 1-amino-3-bromopyrazin-1-ium 2,4,6- trimethylbenzenesulfonate (27.0 g, 72.1 mmol, as prepared in the previous step) in portions at rt, then the reaction mixture was stirred at rt overnight. The mixture was diluted with H2O, the mixture was extracted with MTBE (3 x 200 mL). The combined organic layers were washed with brine, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel chromatography to afford 2.0 g (10%) of ethyl 6-bromopyrazolo[1,5-a]pyrazine-3- carboxylate as a yellow solid.1H NMR (400 MHz, CDCl3) δ (ppm) 9.41 (s, 1H), 8.66 (d, J=0.8Hz, 1H), 8.46 (s, 1H), 4.45 (q, J=7.2 Hz, 2H), 1.45 (t, J=7.2Hz, 3H). Step 3. Synthesis of ethyl 6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrazine-3- carboxylate
Figure imgf000147_0003
[0429] To a mixture of ethyl 6-bromopyrazolo[1,5-a]pyrazine-3-carboxylate (5.0 g, 18.5 mmol, as prepared in the previous step) in dioxane (80 mL) and water (20 mL) was added 1-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (5.7g, 27.7 mmol), Na2CO3 (5.7 g, 55.4 mmol), and Pd(dppf)Cl2 (1.0 g) under nitrogen. The mixture was heated to reflux and stirred under nitrogen for 2.5 h. The mixture was cooled to rt and filtered. The filtrate was poured into cold water, extracted with DCM/MeOH (10/1) ten times, the organic phase was washed with brine, dried over Na2SO4, and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel chromatography to afford 3.5 g (70%) of ethyl 6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrazine-3-carboxylate as a yellow solid.1H NMR (400 MHz, CDCl3) δ (ppm) 9.56 (d, J=1.6 Hz, 1H), 8.59 (d, J=1.2 Hz, 1H), 8.45 (s, 1H), 7.96 (d, J=7.2 Hz, 2H), 4.45 (q, J=7.2Hz, 2H), 4.01 (s, 3H), 1.46 (t, J=7.2 Hz, 3H). Step 4. Synthesis of 6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrazine
Figure imgf000148_0001
[0430] To a mixture of ethyl 6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrazine-3- carboxylate (3.5 g, 12.9 mmol, as prepared in the previous step) in 40% aqueous H2SO4 (35 mL) was stirred at 100°C overnight, then the mixture was poured into water, and the pH was adjusted to 8 with aqueous NaOH. The precipitate was isolated by filtration, the filter cake was washed with water, and dried to afford 1.7 g (66%) of 6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyrazine as yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.19 (d, J=1.6 Hz, 1H), 9.14 (d, J=0.8 Hz, 1H), 8.26 (s, 1H), 8.12 (d, J=2.4 Hz, 1H), 8.05 (s, 1H), 6.956 (dd, J=0.8, 2.4Hz, 1H), 3.89 (s, 3H). Step 5. Synthesis of 3-bromo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrazine (Compound S5)
Figure imgf000148_0002
[0431] To a mixture of 6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrazine (1.0 g, 5.0 mmol, as prepared in the previous step) in DMF (30 mL) was added NBS (1.76 g, 5.0 mmol) in portions. The mixture was stirred at rt for 1 h, poured into water, and extracted with EtOAc three times. The combined organic phase was washed with water and brine, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure and the residue was purified by silica gel chromatography eluting with DCM/MeOH from 1:0 to 100:1 to afford 600 mg (43%) of 3-bromo-6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyrazine (Compound S5) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.19 (d, J=1.2 Hz, 1H), 9.09 (d, J=1.2 Hz, 1H), 8.28 (s, 2H), 8.07 (s, 1H), 3.90 (s, 3H). Synthesis of 3-bromo-7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-a]pyridine (Compound S7)
Figure imgf000149_0001
Step 1. Synthesis of 7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-a]pyridine
Figure imgf000149_0002
[0432] To a mixture of 7-bromoimidazo[1,2-a]pyridine (4.5 g, 22.8 mmol) in dioxane (100 mL) and H2O (20 mL) was added 1-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)-1H-pyrazole (7.1 g, 34.3 mmol), Na2CO3 (5.3 g, 50.3 mmol), and Pd(dppf)Cl2 (0.9 g) under nitrogen. The mixture was heated to reflux and stirred under nitrogen for 6 h. The mixture was cooled to rt and filtered. The filtrate was poured into cold water, extracted with DCM/MeOH (10/1) ten times, then the combined organic phase was washed with brine, dried over Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography to afford 3.4 g (75%) of 7-(1-methyl- 1H-pyrazol-4-yl)imidazo[1,2-a]pyridine as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.50 (d, J=7.2Hz, 1H), 8.29 (s, 1H), 8.03 (s, 1H), 7.86 (s, 1H), 7.76 (s, 1H), 7.52 (s,1H), 7.14 - 7.16 (m, 1H), 3.88 (s, 3H). Step 2. Synthesis of 3-bromo-7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-a]pyridine (Compound S7)
Figure imgf000150_0001
[0433] To a mixture of 7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-a]pyridine (4.1 g, 20.7 mmol, as prepared in the previous step) in ACN (120 mL) was added NBS (3.7 g, 20.7 mmol) in portions. The mixture was stirred at rt for 2 h, poured into water and extracted with DCM/MeOH (10/1) ten times. The combined organic phase was washed with brine, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography to afford 2.2 g (39%) of 3-bromo-7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-a]pyridine (Compound S7) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.36 (s, 1H), 8.29 (d, J=7.2Hz, 1H), 8.08 (s, 1H), 7.85 (s, 1H), 7.67 (s, 1H), 7.32 - 7.35 (m, 1H), 3.88 (s, 3H). Synthesis of 3-bromo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-b]pyridazine (Compound S8)
Figure imgf000150_0002
Step 1. Synthesis of 1-amino-3-bromopyridazin-1-ium 2,4,6-trimethylbenzenesulfonate
Figure imgf000150_0003
[0434] To a solution of 3-bromopyridazine (20.0 g, 125.7 mmol) in DCM (300 mL) was added the solution of O-(mesitylsulfonyl)hydroxylamine (Compound S4) (35.1 g, 125.7 mmol) in DCM (300 mL) dropwise at rt then the reaction mixture was stirred overnight at rt. The mixture was concentrated under reduced pressure then the residue was triturated with MTBE (80 mL) and filtered to afford 24.0 g (51%) of 1-amino-3-bromopyridazin-1- ium 2,4,6-trimethylbenzenesulfonate as black solid. Step 2. Synthesis of ethyl 6-bromopyrazolo[1,5-b]pyridazine-3-carboxylate
Figure imgf000151_0001
[0435] To a solution of K2CO3 (12.4 g, 89.8 mmol) and ethyl propiolate (7.5 g, 77.0 mmol) in DMF (230 mL) was added 1-amino-3-bromopyridazin-1-ium 2,4,6- trimethylbenzenesulfonate (24.0 g, 64.1 mmol, as prepared in the previous step) in portions at rt then the reaction mixture was stirred overnight at rt. The mixture was diluted with H2O and extracted with MTBE (3x200 mL). The combined organic layers were washed with brine, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by chromatography to afford 2.2 g (13%) of ethyl 6-bromopyrazolo[1,5-b]pyridazine-3-carboxylate as a yellow solid. 1H NMR (400 MHz, CDCl3) δ (ppm) 8.40-8.44 (m, 2H), 7.40 (d, J=9.6 Hz, 1H), 4.43 (q, J=7.2 Hz, 2H), 1.44 (t, J=7.2 Hz, 3H). Step 3. Synthesis of ethyl 6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-b]pyridazine-3- carboxylate
Figure imgf000151_0002
[0436] To a mixture of ethyl 6-bromopyrazolo[1,5-b]pyridazine-3-carboxylate (9.0 g, 33.3 mmol, as prepared in the previous step) in dioxane (144 mL) and water (36 mL) was added 1-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (10.4 g, 49.9 mmol), Na2CO3 (10.6g, 99.9 mmol), and Pd(dppf)Cl2 (1.0 g) under nitrogen. The mixture was heated to reflux and stirred under nitrogen for 2 h. The mixture was cooled to rt and filtered. The filtrate was poured into cold water and extracted with DCM/MeOH (10/1) ten times. The combined organic phase was washed with brine, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography to afford 9 g (99%) of ethyl 6-(1-methyl-1H-pyrazol- 4-yl)pyrazolo[1,5-b]pyridazine-3-carboxylate as a yellow solid. 1H NMR (400 MHz, CDCl3) δ (ppm) 8.48 (d, J=9.2 Hz, 1H), 8.42(s, 1H), 8.06 (d, J=4.4 Hz, 1H), 7.45 (d, J=9.2Hz, 1H), 7.86 (s, 1H), 4.43 (q, J=7.2 Hz, 2H), 4.03 (s, 3H), 1.45 (t, J=7.2 Hz, 3H). Step 4. Synthesis of 6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-b]pyridazine
Figure imgf000152_0001
[0437] To a mixture of ethyl 6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-b]pyridazine-3- carboxylate (4.5 g, 16.6 mmol, as prepared in the previous step) in 40% aqueous H2SO4 (45 mL) was stirred at 100°C overnight, then the mixture was poured into water and extracted with DCM/MeOH (10/1) ten times. The combined organic phase was washed with brine, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was washed with MTBE to afford 2.5 g (76%) of 6-(1- methyl-1H-pyrazol-4-yl)pyrazolo[1,5-b]pyridazine as a yellow solid.1H NMR (400 MHz, CDCl3) δ (ppm) 8.04 (d, J=7.2 Hz, 2H), 7.98 (d, J=2.4 Hz, 1H), 7.93 (d, J=9.6 Hz, 1H), 7.21 (d, J=9.2 Hz, 1H), 6.61 (d, J=2.4 Hz, 1H), 4.00 (s, 3H). Step 5. Synthesis of 3-bromo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-b]pyridazine (Compound S8)
Figure imgf000152_0002
[0438] To a mixture of 6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-b]pyridazine (2.5 g, 12.5 mmol, as prepared in the previous step) in ACN (50 mL) was added NBS (2.2 g, 12.5 mmol) in portions. The mixture was stirred at rt for 1 h, then poured into water and extracted with DCM/MeOH (10/1) ten times. The combined organic phase was washed with brine, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure to afford 2.5 g (71%) of 3-bromo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- b]pyridazine (Compound S8) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.51 (s, 1H), 8.14-8.19 (m, 3H), 7.65 (d, J=9.6Hz, 1H), 3.93 (s, 3H). Synthesis of 3-bromo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound S9)
Figure imgf000153_0001
Step 1. Synthesis of 6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine
Figure imgf000153_0002
[0439] To a mixture of 6-bromopyrazolo[1,5-a]pyridine (4.5 g, 22.8 mmol) in dioxane (90 mL) and water (18 mL) was added 1-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)-1H-pyrazole (7.1 g, 34.2 mmol), Na2CO3 (7.2g, 68.8 mmol), and Pd(dppf)Cl2 (0.45 g) under nitrogen. The mixture was heated to reflux and stirred under nitrogen for 3 h. The mixture was cooled to rt and filtered. The filtrate was poured into cold water and extracted with DCM/MeOH (10/1) ten times. The combined organic phase was washed with brine, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography to afford 3.5 g (78%) of 6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine as a yellow solid. 1H NMR (400 MHz, CDCl3) δ (ppm) 8.61 (s, 1H), 7.95(d, J=1.6 Hz, 1H), 7.77 (s, 1H), 7.63 (s, 1H), 7.57 (d, J=9.2 Hz, 1H), 7.23-7.26 (m,1H), 6.52 (d, J=1.6 Hz, 1H), 3.99 (s, 3H). Step 2. Synthesis of 3-bromo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound S9)
Figure imgf000153_0003
[0440] To a mixture of 6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (4.0 g, 20.2 mmol, as prepared in the previous step) in ACN (80 mL) was added NBS (4.3 g, 24.2 mmol) in portions. The mixture was stirred at rt for 1 h, then poured into water and extracted with DCM/MeOH (10/1) ten times. The combined organic phase was washed with brine, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography to afford 2.5 g (45%) of 3- bromo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound S9) as a yellow solid.1H NMR (400 MHz, CDCl3) δ (ppm) 8.55 (s, 1H), 7.92 (s, 1H), 7.77 (s, 1H), 7.65 (s, 1H), 7.53 (d, J=8.8 Hz, 1H), 7.53 (dd, J=0.8, 9.2 Hz, 1H), 4.00 (s, 3H). Synthesis of 3-bromo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidine (Compound S10)
Figure imgf000154_0001
Step 1. Synthesis of 6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidine
Figure imgf000154_0002
[0441] To a mixture of 6-bromopyrazolo[1,5-a]pyrimidine (4.5 g, 22.7 mmol) in dioxane (90 mL) and water (18 mL) was added 1-methyl-4-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)-1H-pyrazole (7.0 g, 34.1 mmol), Na2CO3 (7.2g, 68.1 mmol), and Pd(dppf)Cl2 (0.45 g) under nitrogen. The mixture was heated to reflux and stirred under nitrogen for 3 h. The mixture was cooled to at rt and filtered. The filtrate was poured into cold water and extracted with DCM/MeOH (10/1) ten times. The combined organic phase was washed with brine, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography to afford 3.5 g (78%) of 6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrimidine as a yellow solid. 1H NMR (400 MHz, CDCl3) δ (ppm) 8.75-8.76 (m, 1H), 8.64 (d, J=2 Hz, 1H), 8.12 (d, J=2.4 Hz, 1H), 7.80 (s, 1H), 7.70 (s, 1H), 6.72-6.73 (m,1H), 4.02 (s, 3H). Step 2. Synthesis of 3-bromo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidine (Compound S10)
Figure imgf000155_0001
[0442] To a mixture of 6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidine (7.0 g, 35.1 mmol, as prepared in the previous step) in ACN (140 mL) was added NBS (7.5 g, 42.1 mmol) in portions. The mixture was stirred at rt for 1 h, poured into water and extracted with DCM/MeOH (10/1) ten times. The combined organic phase was washed with brine, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography to afford 5 g (51%) of 3-bromo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidine (Compound S10) as a yellow solid. 1H NMR (400 MHz, CDCl3) δ (ppm) 8.72-8.69 (m, 2H), 8.10 (s, 1H), 7.79 (s, 1H), 7.72 (s, 1H), 4.02 (s, 3H). Synthesis of tert-butyl 4-[6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl]piperazine-1-carboxylate (Compound 11)
Figure imgf000155_0002
[0443] To a round bottom flask containing 3-bromo-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine (Compound S9) (4.00 g, 14.4 mmol) and tert-butyl-1- piperazinecarboxylate (3.23 g, 17.3 mmol) was added tBuOH (202 mL) and dioxane (124 mL). The solution was sparged with nitrogen for 20 min at 25°C. To the solution was added tBuXPhos Pd G1 (248 mg, 5.41 mmol) and NaOtBu (520 mg, 0.54 mmol) and the reaction mixture was sparged with nitrogen for an additional 10 min at 25°C. The flask was heated in a 55°C bath for 1.5 h, cooled to rt and diluted with EtOAc. The mixture was washed with H2O and brine, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with 10-90% EtOAc/DCM. The solids obtained were dried under reduced pressure to afford 1.62 g (29%) of tert-butyl 4-[6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl]piperazine-1-carboxylate (Compound 11) as a yellow solid.1H NMR (400 MHz, CDCl3) δ (ppm) 8.46 (s, 1H), 7.74 (s, 1H), 7.70 (s, 1H), 7.63 (s, 1H), 7.50 (d, J = 7.3 Hz, 1H), 7.14 (d, J = 1.47 Hz, 1H), 4.03-4.01 (m, 4H), 3.98 (s, 3H), 3.64 (m, 4H), 3.02 (m, 4H), 1.50 (s, 9H); MS (ESI) m/z [M+H]+ calcd. for C20H27N6O2, 383.2; found, 383.3; HPLC purity: 210 nm: 99.4%; 254 nm: 99.2%. Synthesis of 6-(1-methyl-1H-pyrazol-4-yl)-3-piperazin-1-ylpyrazolo[1,5-a]pyridine (Compound 12)
Figure imgf000156_0001
[0444] To a round bottom flask containing tert-butyl 4-[6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl]piperazine-1-carboxylate (Compound 11) (0.340 g, 0.89 mmol) was added DCM (6.80 mL) then the solution was cooled on an ice bath and TFA (1.70 mL, 22.1 mmol) was added slowly. After 1 h the solvent was removed under reduced pressure, then MTBE was added to the residue and concentrated under reduced pressure to remove excess TFA. The residue was dissolved in DCM and swirled with sat. aqueous NaHCO3, diluted with H2O, and diluted with MTBE. The aqueous layer was washed with MTBE then extracted with DCM. The DCM layer was dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure to afford 105 mg (42%) of 6- (1-methyl-1H-pyrazol-4-yl)-3-piperazin-1-ylpyrazolo[1,5-a]pyridine (Compound 12) as a yellow solid.1H NMR (400 MHz, CDCl3) δ (ppm) 8.45 (s, 1H), 7.74 (s, 1H), 7.70 (s, 1H), 7.60 (s, 1H), 7.52 (d, J = 9.17 Hz, 1H), 7.11 (d, J = 9.05 Hz, 1H), 3.97 (s, 3H), 3.08 (bs, 5H), 3.06 (bs, 4H); MS (ESI+, m/z): Calcd. for C15H19N6: 283.2. Found: 283.1; HPLC purity: 210 nm: 100%; 254 nm: 100%. Synthesis of 6-(1-methyl-1H-pyrazol-4-yl)-3-piperazin-1-ylpyrazolo[1,5-a]pyridine dihydrochloride salt (Compound S19)
Figure imgf000156_0002
[0445] To a round bottom flask containing tert-butyl 4-[6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl]piperazine-1-carboxylate (Compound 11) (0.250 g, 0.65 mmol) was added MeOH (5.00 mL) and 4M HCl in dioxane (2 mL). The reaction was stirred at 27°C for 24 h, then stirred another 5 h at 24°C. The mixture was filtered through a frit, and the solid was rinsed with MeOH (5 mL). The solid was dried under reduced pressure to afford 227 mg (98%) of 6-(1-methyl-1H-pyrazol-4-yl)-3-piperazin-1- ylpyrazolo[1,5-a]pyridine dihydrochloride salt (Compound S19) as a light gray solid. MS (ESI) m/z [M+H]+ calcd. for C15H21Cl2N6, 283.2; found, 283.2. Synthesis of di-tert-butyl 4,4'-(1,2-bis(1H-benzo[d][1,2,3]triazol-1-yl)ethane-1,2- diyl)bis(piperazine-1-carboxylate) (Compound S47)
Figure imgf000157_0001
[0446] A solution of 1H-1,2,3-benzotriazole (19.2 g, 161.1 mmol) and tert-butyl piperazine-1-carboxylate (30.0 g, 161.1 mmol) in EtOH (300 mL) was stirred under nitrogen at 25℃ for 20 min. Then glyoxal (40 wt.% in H2O, 4.7 g, 80.5 mmol) was added to the mixture at 25℃. The reaction mixture was stirred at 30℃ for 16 h. The reaction mixture was concentrated under reduced pressure then PE was added. The mixture was stirred for 30 min and filtered, the filter cake was dried to afford 42.0 g (32%) of di-tert- butyl 4,4'-(1,2-bis(1H-benzo[d][1,2,3]triazol-1-yl)ethane-1,2-diyl)bis(piperazine-1- carboxylate (Compound S47) as a white solid. [0447] Using the procedures described for Compound S47 and reagents, starting materials, and conditions known to those skilled in the art, the following compounds representative of the present disclosure were prepared:
Figure imgf000157_0003
Synthesis of 6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-amine (Compound S56)
Figure imgf000157_0002
Step 1. Synthesis of 6-(1-methyl-1H-pyrazol-4-yl)-3-nitropyrazolo[1,5-a]pyridine
Figure imgf000158_0001
[0448] To a solution of 6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (4.0 g, 5.05 mmol, as prepared in Compound S9, Step 1) dissolved in conc. H2SO4 (10 mL) was added KNO3 (2.24 g, 5.55 mmol). The reaction mixture was stirred for 1 h at rt under nitrogen. The mixture was cooled to 0°C and diluted with water (300 mL). The mixture was neutralized to pH 8 with sat. aqueous Na2CO3. The precipitated solids were collected by filtration and washed with water (3x300 mL). The filter cake was dried under reduced pressure to afford 3.5 g (71%) of 6-(1-methyl-1H-pyrazol-4-yl)-3-nitropyrazolo[1,5- a]pyridine as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.34 (s, 1H), 8.90 (s, 1H), 8.38 (s, 1H), 8.24 (d, J = 9.1 Hz, 1H), 8.15 (d, J = 9.3 Hz, 1H), 8.11 (s, 1H), 3.90 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C11H9N5O2, 244.1; found, 244.1. Step 2. Synthesis of 6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-amine (Compound S56)
Figure imgf000158_0002
[0449] To a solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-nitropyrazolo[1,5-a]pyridine (3.0 g, 12.33 mmol, as prepared in the previous step) dissolved in aqueous HBr (50 mL) was added SnCl2·2H2O (8.42 g, 37.0 mmol). The resulting solution was stirred for 4 h at 90°C under nitrogen. The mixture was cooled to rt, the pH was adjusted to pH 9 with aqueous NaOH, and extracted with EtOAc (3 x 500 mL). The combined organic extracts were washed with brine (3x100 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure to afford 2.5 g (90%) of 6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-amine (Compound S56) as a yellow-brown solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.63 (s, 1H), 8.15 (s, 1H), 7.92 (s, 1H), 7.59 (d, J = 9.2 Hz, 1H), 7.44 (s, 1H), 7.11 (d, J = 9.2 Hz, 1H), 4.33 (s, 2H), 3.86 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C11H11N5, 214.1; found, 214.1. Synthesis of 6-(1-methyl-1H-pyrazol-4-yl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)pyrazolo[1,5-a]pyridine (Compound S57)
Figure imgf000159_0001
Step 1. Preparation of 3-iodo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine
Figure imgf000159_0002
[0450] To a solution of 6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (7.0 g, 35.3 mmol, as prepared in Compound S9, Step 1) in DMF (70 mL) was added NIS (9.53 g, 42.4 mmol). The resulting mixture was stirred for 1 h at rt. The reaction was poured into a stirred mixture of 2.8M aqueous Na2S2O3 (64 mL) and 1.14M aqueous NaHCO3 (144 mL). The mixture was stirred vigorously for 1 h resulting in an off-white precipitate. The solids were collected by filtration, washed with H2O (2 x 10 mL), and dried under high vacuum to afford 10 g (79%) of 3-iodo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine as a white solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.06 (s, 1H), 8.28 (s, 1H), 8.08 (s, 1H), 8.02 (d, J = 0.8 Hz, 1H), 7.60 (d, J = 9.2 Hz, 1H), 7.52 (d, J = 9.2 Hz, 1H), 3.88 (s, 3H).; MS (ESI) m/z [M+H]+ calcd. for C11H9IN4, 325.0; found, 325.1. Step 2. Preparation of 6-(1-methyl-1H-pyrazol-4-yl)-3-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyrazolo[1,5-a]pyridine (Compound S57)
Figure imgf000159_0003
[0451] To a solution of 3-iodo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (9.0 g, 27.8 mmol, as prepared in the previous step) in THF (180 mL) was added 2-isopropoxy- 4,4,5,5-tetramethyl-1,3,2-dioxaborolane (17.1 g, 91.7 mmol) then cooled to 0°C under nitrogen and 1.3M iPrMgCl·LiCl in THF (31.9 mL, 41.7 mmol) was added dropwise. The reaction was stirred for 3 h at 0°C under nitrogen, diluted with DCM (160 mL), and filtered through a pad of silica gel. The pad was washed with EtOAc (100 mL) and the combined filtrates were concentrated under reduced pressure to afford 5.0 g (42%) of 6-(1-methyl- 1H-pyrazol-4-yl)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazolo[1,5-a]pyridine (Compound S57) as a grey white solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.08 (s, 1H), 8.26 (s, 1H), 8.12 (s, 1H), 8.01 (d, J = 0.8 Hz, 1H), 7.82 (d, J = 9.1 Hz, 1H), 7.65 (d, J = 9.1 Hz, 1H), 3.89 (s, 3H), 1.32 (s, 12H).; MS (ESI) m/z [M+H]+ calcd. for C17H21BN4O2, 325.2; found, 325.2. Synthesis of 2-chloro-5-(pyridin-2-ylmethyl)pyrimidine (Compound S80, Steps 1-2)
Figure imgf000160_0001
Step 1. Preparation of potassium pyridine-2-ylacetate
Figure imgf000160_0002
[0452] In dried glassware, a solution of methyl 2-pyridylacetate (0.500 g, 3.31 mmol), absolute ethanol (6.6 mL), and water (0.63 mL, 3.47 mmol) was heated at 60°C. A solution of potassium tert-butoxide (0.390 g, 3.47 mmol) was prepared in absolute ethanol (3.3 mL) in dried glassware, and then added dropwise to the initial solution at 60°C over 30 min. After complete conversion, the solvent was removed under vacuum and the residue was stirred in anhydrous Et2O (6.6 mL) for 30 min. The solids were filtered through a frit and washed in quick sequence with ethanol/ether (1:1, 0.6 mL x2) and Et2O (3 mL x3). The white solid was transferred to a dried flask and placed under vacuum in a 30°C bath for 2 h to afford 468 mg (81%) of potassium pyridine-2-ylacetate as a white solid.1H NMR (400 MHz, DMSO-d6) d (ppm) 8.33 (d, J = 4.3 Hz, 1H) 7.57 (td, J = 7.6, 1.8, 1H) 7.27 (d, J = 7.8 Hz, 1H) 7.06 (dd, J = 6.6, 5.4 Hz 1H) 3.32 (s, 2H); 13C NMR (100 MHz, DMSO-d6) d (ppm) 49.5, 119.9, 123.8, 135.1, 147.8, 160.5, 171.5; MS (ESI) m/z [M+H]+ calcd. for C7H8NO2: 138.1; found, 138.0. Step 2.2-Chloro-5-(pyridin-2-ylmethyl)pyrimidine (Compound S80)
Figure imgf000161_0001
[0453] 5-Bromo-2-chloropyrimidine (110 mg, 0.590 mmol), Xantphos (22 mg, 0.04 mmol), Pd2(dba)3 (10 mg, 0.01 mmol), potassium pyridin-2-ylacetate (125 mg, 0.713 mmol, as prepared in the previous step), and mesitylene (1.2 mL) were added to a pressure tube that was pre-flushed with argon. It was sparged with more argon, sealed, and heated at 150°C for 24 h. The mixture was cooled to rt and purified by silica gel flash chromatography (acetone with 0.1% Et3N/DCM) to afford 72 mg (59%) of 2-chloro-5- (pyridin-2-ylmethyl)pyrimidine (Compound S80) as an oil.1H NMR (400 MHz, CDCl3) d (ppm) 8.59 (s, 2H) 8.57 (m, 1H) 7.68 (td, J=7.6, 1.8 Hz 1H) 7.21 (m, 2H) 4.13 (s, 2H); MS (ESI) m/z [M+H]+ calcd. for C10H9ClN3: 206.5; found, 206.0; HPLC purity: 210 nm: 93.3%; 254 nm: 100.0%. Synthesis of 7-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)imidazo[1,2-b]pyridazine hydrochloride salt (Compound S83)
Figure imgf000161_0002
Step 1. Synthesis of tert-butyl 4-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazin-3- yl)piperazine-1-carboxylate
Figure imgf000161_0003
[0454] To a 20 mL vial containing 3-bromo-7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2- b]pyridazine (Compound S3) (300.4 mg, 1.080 mmol) and tert-butyl 1- piperazinecarboxylate (1.00 g, 5.40 mmol) was added tBuOH (3.0 mL) and dioxane (1.5 mL) then the solution was sparged with argon for 10 min. KOtBu (182 mg, 1.62 mmol) was added and the mixture was sparged again for 10 min with argon. tBuXPhos Pd G1 (111 mg, 0.162 mmol) was added, and the mixture was sparged for 10 min with argon. The mixture was stirred at 55°C under an argon for 1 h. The reaction was cooled to rt, diluted with DCM (40 mL), and washed with 10% citric acid solution (3 x 10 mL), H2O (3 x 10 mL), and sat. aqueous NaHCO3 (2 x 10 mL). The organic layer was dried over anhydrous Na2SO4 and filtered, then the filtrate was concentrated under reduced pressure. The residue was purified by silica gel flash chromatography eluted with 0-100% (10% MeOH in DCM)/DCM to afford 280.8 mg (68%) of tert-butyl 4-(7-(1-methyl-1H-pyrazol-4- yl)imidazo[1,2-b]pyridazin-3-yl)piperazine-1-carboxylate as a yellow solid.1H NMR (400 MHz, CDCl3) δ (ppm) 8.51 (d, J = 2.0 Hz, 1H), 7.91 (d, J = 2.0 Hz, 1H), 7.84 (s, 1H), 7.75 (s, 1H), 7.25 (s, 1H), 4.00 (s, 3H), 3.75 - 3.64 (m, 4H), 3.29 - 3.20 (m, 4H), 1.51 (s, 9H); MS (ESI) m/z [M+H]+ calcd. for C19H25N7O2: 384.2, Found: 384.2; HPLC purity: 210 nm: 97.8%; 254 nm: 100.0%. Step 2. Synthesis of 7-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)imidazo[1,2- b]pyridazine hydrochloride salt (Compound S83)
Figure imgf000162_0001
[0455] A solution of tert-butyl 4-[7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazin- 3-yl]piperazine-1-carboxylate (274.5 mg, 0.7159 mmol, as prepared in the previous step) in DCM (30 mL) was cooled to 0°C then a solution of 4M HCl in dioxane (1.8 mL, 7.3 mmol) was added dropwise. After completion of addition, the mixture was warmed to rt and stirred for 7 h. The reaction was concentrated under reduced pressure, then the residue was triturated with ether and filtered. The solid was dried under reduced pressure to afford 214.6 mg (94%) of 7-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)imidazo[1,2- b]pyridazine hydrochloride salt (Compound S83) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.69 (br s, 2H), 9.34 (d, J = 2.0 Hz, 1H), 8.69 (s, 1H), 8.44 (d, J = 2.0 Hz, 1H), 8.34 (s, 1H), 7.98 (s, 1H), 3.94 (s, 3H), 3.54 - 3.49 (m, 4H), 3.37 - 3.25 (m, 4H); MS (ESI) m/z [M+H]+ calcd. for C14H17N7: 284.2, Found: 284.2; HPLC purity: 210 nm: 95.9%; 254 nm: 100.0%. Synthesis of 5-benzyl-2-chloropyrimidine (Compound S88)
Figure imgf000162_0002
[0456] To a mixture of (2-chloropyrimidin-5-yl)boronic acid (20 g, 0.126 mol) and Na2CO3 (40.43 g, 0.381 mol) in dioxane (200 mL) and H2O (80mL) stirred under nitrogen at 25°C was added (Ph3P2PdCl2 (4.43 g, 0.0063 mol);benzyl bromide (23.76 g, 0.138 mol) was added after 20 min The reaction mixture was stirred at 100°C for 1 h. The mixture was diluted with water and extracted with EtOAc (3x50mL). The combined organic layers were concentrated under vacuum to give crude product, which was purified by silica gel column eluting with DCM/MeOH (30:1~15:1) to afford 18.18 g (69%) of 5-benzyl-2- chloropyrimidine (Compound S88) as a white solid.1H NMR (400 MHz, CDCl3) δ (ppm) 8.48 (s, 2H), 7.34~7.37 (m, 2H), 7.27~7.31 (m, 1H), 7.19 (d, J=7.2Hz, 2H), 3.98 (s,2H). Synthesis of (R)-1-(4-chlorophenyl)ethyl (2,5-dioxopyrrolidin-1-yl) carbonate (Compound S95)
Figure imgf000163_0001
[0457] To a solution of (1R)-1-(4-chlorophenyl)ethanol (20.00 g, 127.7 mmol) in ACN (240 mL was added di(N-succinimidyl) carbonate (49.07 g, 191.6 mmol) then the mixture was cooled to 0°C. TEA (53 mL, 380 mmol) was added dropwise over 5 min then stirred at 0°C for 30 min. The solution was warmed to rt and stirred overnight, then the mixture was concentrated under reduced pressure. The residue was diluted with EtOAc (250 mL) and washed with sat. aqueous NaHCO3. Additional EtOAc (100 mL) was added, then the organic layer was washed with water and brine, then dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure. To the residue was added MTBE (150 mL) and the mixture was heated to 45°C. Hexanes (100 mL) was added hot, then the mixture was allowed to cool with efficient stirring. The solids were isolated by filtration, washed with hexanes (100 mL), and dried under reduced pressure to afford 31.85 g (84%) of (R)-1-(4-chlorophenyl)ethyl (2,5-dioxopyrrolidin-1-yl) carbonate (Compound S95) as a light tan solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 7.49 (s, 4H), 5.89 (q, J = 6.8 Hz, 1H), 2.80 (s, 4H), 1.62 (d, J = 6.8 Hz, 3H). Synthesis of 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)pyrazolo[1,5-a]pyrimidine (Compound S98)
Figure imgf000164_0001
Step 1. Preparation of tert-butyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrimidin-3-yl)piperazine-1-carboxylate
Figure imgf000164_0002
[0458] To a vial was added 3-bromo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrimidine (Compound S10) (0.150 g, 0.539 mmol) and tert-butyl 1- piperazinecarboxylate (0.120 g, 0.647 mmol) followed by tBuOH (1.5 mL) and dioxane (3 mL) then the mixture was sparged with argon for 5 min. NaOtBu (0.0778 g, 0.809 mmol) was added then the mixture was sparged for 5 min with argon. tBuXPhos Pd G1 (37.0 mg, 0.0539 mmol) was added then the mixture was sparged with argon for 5 min. The reaction was heated to 80°C for 2.5 h then additional tBuXPhos Pd G1 (15 mg) was added as a slurry in dioxane (0.5 mL) and the mixture was stirred at 80°C overnight. The reaction was cooled to rt, quenched with sat. aqueous NaHCO3, and extracted with DCM (3x). The combined organic extracts were washed with water, then dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography to afford 95 mg (46%) of tert-butyl 4-(6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin-3-yl)piperazine-1-carboxylate as a yellow-orange solid.1H NMR (400 MHz, CDCl3) δ (ppm) 8.58 (d, J = 2.0 Hz, 1H), 8.45 (d, J = 2.4 Hz, 1H), 7.78 (s, 1H), 7.77 (s, 1H), 7.68 (s, 1H), 4.01 (s, 3H), 3.71 – 3.65 (m, 4H), 3.30 – 3.24 (m, 4H), 1.51 (s, 9H). MS (ESI) m/z [M+H]+ calcd. for C19H25N7O2, 384.2; found, 384.3. Step 2. Preparation of 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)pyrazolo[1,5- a]pyrimidine (Compound S98)
Figure imgf000165_0001
[0459] To a solution of tert-butyl 4-[6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrimidin-3-yl]piperazine-1-carboxylate (0.09 g, 0.2 mmol, as prepared in the previous step) in DCM (2 mL) was added TFA (0.2 mL) and the reaction was stirred at rt overnight. The mixture was diluted with DCM (20 mL) and washed with sat. aqueous NaHCO3 (25 mL). The aqueous layer was extracted with DCM (2 x 20 mL) and the combined organic extracts were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure to afford 65 mg (93%) of 6-(1-methyl-1H-pyrazol-4-yl)-3- (piperazin-1-yl)pyrazolo[1,5-a]pyrimidine (Compound S98) as an orange solid. 1H NMR (400 MHz, CDCl3) δ (ppm) 8.58 (d, J = 2.0 Hz, 1H), 8.45 (d, J = 2.4 Hz, 1H), 7.79 (s, 1H), 7.77 (s, 1H), 7.68 (s, 1H), 4.01 (s, 3H), 3.37 – 3.31 (m, 4H), 3.20 – 3.14 (m, 4H). MS (ESI) m/z [M+H]+ calcd. for C14H17N7, 284.2; found, 284.1. Synthesis of 5-benzyl-2-pyrrolidin-3-ylpyrimidine (Compound S100)
Figure imgf000165_0002
Step 1. Preparation of tert-butyl 3-(5-benzylpyrimidin-2-yl)-2,5-dihydro-1H-pyrrole-1- carboxylate
Figure imgf000165_0003
[0460] A mixture of tert-butyl 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2,5- dihydro-1H-pyrrole-1-carboxylate (500.0 mg, 1.69 mmol), 5-benzyl-2-chloropyrimidine (Compound S88) (289 mg, 1.41 mmol) and K3PO4 (599 mg, 2.82 mmol) in dioxane (14.3 mL) and H2O (5.1 mL) was sparged with argon for 20 min. (tBu3P)2Pd (108 mg, 0.21 mmol) was added then the mixture was heated to 80°C for 4 h. The reaction was cooled to rt, diluted with H2O (40 mL), and extracted with EtOAc (2 x 25 mL). The combined organic extracts were washed with H2O (25 mL), dried over anhydrous MgSO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with 20-30% EtOAc/hexanes to afford 420 mg (88%) of tert-butyl 3-(5-benzylpyrimidin-2-yl)-2,5-dihydro-1H-pyrrole-1-carboxylate as a white solid. 1H NMR (400 MHz, CDCl3) δ (ppm) 8.56 (s, 2H), 7.31 - 7.39 (m, 2H), 7.24 - 7.31 (m, 1H), 7.20 (d, J=6.97 Hz, 2H), 6.83 - 6.95 (m, 1H), 4.53 - 4.69 (m, 2H), 4.33 - 4.49 (m, 2H), 3.89 - 4.03 (m, 2 H), 1.53 (s, 9H); MS (ESI) m/z [M+H]+ calcd. for C20H24N3O2: 338.2; found, 338.3; HPLC purity 210 nm: 98.3%; 254 nm: 100.0%. Step 2. Preparation of tert-butyl 3-(5-benzylpyrimidin-2-yl)pyrrolidine-1-carboxylate
Figure imgf000166_0001
[0461] To a solution of tert-butyl 3-(5-benzylpyrimidin-2-yl)-2,5-dihydro-1H-pyrrole-1- carboxylate (437 mg, 1.30 mmol, as prepared in the previous step) in EtOAc (20 mL) was added 10% Pd/C (100 mg, 50% H2O). The reaction flask was evacuated and filled with hydrogen gas three times and allowed to stir under an atmosphere of hydrogen. After 19 h, the reaction mixture was filtered through a pad of celite, and the pad was washed with an additional 40 mL of EtOAc. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with 25-40% EtOAc/hexanes to afford 377 mg (86%) of tert-butyl 3-(5-benzylpyrimidin-2-yl)pyrrolidine-1-carboxylate as a colorless, viscous oil.1H NMR (400 MHz, CDCl3) δ (ppm) 8.52 (s, 2H), 7.30 - 7.39 (m, 2H), 7.22 - 7.30 (m, 1H), 7.19 (d, J=7.09 Hz, 2H), 3.95 (s, 2H), 3.83 (m, 1H), 3.53 - 3.73 (m, 3H), 3.43 (m, 1H), 2.18 - 2.43 (m, 2H), 1.47 (s, 9H); MS (ESI) m/z [M+H]+ calcd. for C20H25N3O2: 340.2; found, 340.3; HPLC purity: 210 nm: 100.0%; 254 nm 100.0%. Step 3. Preparation of 5-benzyl-2-pyrrolidin-3-ylpyrimidine (Compound S100)
Figure imgf000166_0002
[0462] A solution of tert-butyl 3-(5-benzylpyrimidin-2-yl)pyrrolidine-1-carboxylate (377 mg, 1.11 mmol, as prepared in the previous step) in DCM (9.0 mL) was cooled to 0°C, then TFA (1.0 mL) was added dropwise. The mixture was stirred for 30 min at 0°C then warmed to rt for 3 h. The reaction was concentrated under reduced pressure then the residue was dissolved in DCM (10 mL) and concentrated again. The residue was dissolved in DCM (20 mL) and washed with 20 mL sat. aqueous NaHCO3. The organic phase was dried over anhydrous Na2SO4 and filtered, then the filtrate was concentrated under reduced pressure to afford 211 mg (79%) of 5-benzyl-2-pyrrolidin-3-ylpyrimidine (Compound S100) as a light pink viscous oil.1H NMR (400 MHz, CD3OD) δ (ppm) 8.58 (s, 2H), 7.27 - 7.36 (m, 2H), 7.19 - 7.27 (m, 3H), 3.99 (s, 2H), 3.56 - 3.69 (m, 1H), 3.32 - 3.38 (m, 1H), 3.23 - 3.30 (m, 1H), 3.13 - 3.23 (m, 1H), 3.08 (dt, J=11.19, 7.37 Hz, 1H), 2.24 - 2.38 (m, 1H), 2.07 - 2.24 (m, 1H); MS (ESI) m/z [M+H]+ calcd. for C15H17N3: 240.2; found, 240.1; HPLC purity: 210 nm: 100.0%; 254 nm: 100.0%. Synthesis of 1-benzyl-3-(pyrrolidin-3-yl)-1H-1,2,4-triazole (Compound S101)
Figure imgf000167_0001
Step 1. Preparation of 1-benzyl-3-bromo-1H-1,2,4-triazole
Figure imgf000167_0002
[0463] A mixture of 3-bromo-1H-1,2,4-triazole (5.0 g, 33.8 mmol), benzyl bromide (5.8 g, 33.8 mmol) and NaOMe (1.8 g, 33.8 mmol) in DMF (50 mL) was stirred under nitrogen at 35°C for 6 h, then diluted with water and extracted with EtOAc (5 x 100 mL). The combined organic extracts were concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with 1:50 EtOAc/PE to 1:20 EtOAc/PE to afford 2.5 g (31%) of 1-benzyl-3-bromo-1H-1,2,4-triazole as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.72 (s, 1H), 7.30~7.40 (m, 5H), 5.40 (s, 2H). Step 2. Preparation of tert-butyl 3-(1-benzyl-1H-1,2,4-triazol-3-yl)-2,5-dihydro-1H- pyrrole-1-carboxylate
Figure imgf000167_0003
[0464] To a solution of 1-benzyl-3-bromo-1H-1,2,4-triazole (3.0 g, 12.6 mmol, as prepared in the previous step) in a mixture of dioxane (10 mL) and H2O (30 mL) was added tert- butyl 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2,5-dihydro-1H-pyrrole-1- carboxylate (4.5 g, 15.1 mmol), K3PO4 (5.4 g, 25.2 mmol), and Pd(PPh3)2Cl2 (0.4 g, 0.6 mmol) under nitrogen. The reaction was stirred at 80°C for 4 h under nitrogen, then poured into water (100 mL) and extracted with EtOAc (3 x 50 mL). The combined organic extracts were washed with brine, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with 1:10 EtOAc/PE to 1:1 EtOAc/PE to afford 3.3 g (80%) of tert-butyl 3-(1-benzyl-1H-1,2,4-triazol-3-yl)-2,5-dihydro-1H-pyrrole-1-carboxylate as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.67 (d, J=4.4Hz, 1H), 7.26~7.35 (m, 5H), 6.42 (d, J=12.8Hz, 1H), 5.41 (s, 1H), 4.32 (s, 2H), 4.18 (s, 2H), 1.41~1.43 (m, 12H). Step 3. Preparation of tert-butyl 3-(1-benzyl-1H-1,2,4-triazol-3-yl)pyrrolidine-1- carboxylate
Figure imgf000168_0001
[0465] A mixture of tert-butyl 3-(1-benzyl-1H-1,2,4-triazol-3-yl)-2,5-dihydro-1H-pyrrole- 1-carboxylate (3.0 g, 9.2 mmol, as prepared in the previous step) and 10% Pd/C (50% water, 1.6 g) in EtOAc (30 mL) was stirred at 25°C for 5 h under an atmosphere of hydrogen (20 psi). The mixture was filtered and the filtrate was concentrated to afford 3.0 g (99%) of tert-butyl 3-(1-benzyl-1H-1,2,4-triazol-3-yl)pyrrolidine-1-carboxylate as a yellow solid. 1HNMR (400 MHz, CDCl3) δ (ppm) 7.94 (s, 1H), 7.37 (d, J=7.2Hz, 3H), 7.25-7.29 (m, 2H), 5.28 (d, J=7.2Hz, 2H), 3.79 (s, 1H), 3.42~3.52 (m, 3H), 3.26~3.28 (m, 1H), 2.20~2.23 (m, 2H), 1.37~1.45 (m, 9H). Step 4. Preparation of 1-benzyl-3-(pyrrolidin-3-yl)-1H-1,2,4-triazole (Compound S101)
Figure imgf000168_0002
[0466] To a solution of tert-butyl 3-(1-benzyl-1H-1,2,4-triazol-3-yl)pyrrolidine-1- carboxylate (2.9 g, 8.8 mmol, as prepared in the previous step) in DCM (30 mL) was added TFA (10 mL) at 0°C. The reaction mixture was stirred at 25°C for 3 h under nitrogen. The pH of the mixture was adjusted to pH 9 with sat. aqueous NaHCO3, then extracted with EtOAc (3 x 50 mL). The combined organic extracts were washed with brine, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure to afford 1.1 g (55%) of 1-benzyl-3-(pyrrolidin-3-yl)-1H-1,2,4-triazole (Compound S101) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.53 (s, 1H), 7.31~7.34 (m, 3H), 7.26~7.30 (m, 2H), 5.33 (s, 2H), 3.21~3.38 (m, 2H), 2.86~2.96 (m, 3H), 1.87 ~2.05 (m, 2H). Synthesis of 3-(4-(1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine (Compound S102)
Figure imgf000169_0001
Step 1. Preparation of a mixture of 3-bromo-1-(tetrahydro-2H-pyran-2-yl)-1H-1,2,4- triazole and 3-bromo-4-(tetrahydro-2H-pyran-2-yl)-4H-1,2,4-triazole and 5-bromo-1- (tetrahydro-2H-pyran-2-yl)-1H-1,2,4-triazole
Figure imgf000169_0002
[0467] To a solution of 3-bromo-1H-1,2,4-triazole (500 mg, 3.37 mmol) and 3,4-dihydro- 2H-pyran (426.3 mg, 5.06 mmol) in toluene (20 mL) was added TsOH·H2O (64.2 mg, 0.33 mmol) then the mixture was heated to 105°C and stirred overnight under nitrogen. The reaction was cooled to rt then concentrated under reduced pressure. The residue was dissolved in water (100 mL) and extracted with DCM (3 x 100 mL). The combined organic extracts were dried over anhydrous Na2SO4 and filtered, then the filtrate was concentrated under reduced pressure to afford 700 mg (89%) of a mixture of 3-bromo-1-(tetrahydro-2H- pyran-2-yl)-1H-1,2,4-triazole and 3-bromo-4-(tetrahydro-2H-pyran-2-yl)-4H-1,2,4- triazole and 5-bromo-1-(tetrahydro-2H-pyran-2-yl)-1H-1,2,4-triazole as a black oil. MS (ESI) m/z [M+H]+ calcd. for C7H10BrN3O, 232.0; found, 232.0. LCMS purity: 254 nm:85%. Step 2. Preparation of a mixture of 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(tetrahydro-2H- pyran-2-yl)-1H-1,2,4-triazol-3-yl)piperazin-1-yl)pyrazolo[1,5-a]pyridine, 6-(1-methyl- 1H-pyrazol-4-yl)-3-(4-(4-(tetrahydro-2H-pyran-2-yl)-4H-1,2,4-triazol-3-yl)piperazin-1- yl)pyrazolo[1,5-a]pyridine, and 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(tetrahydro-2H- pyran-2-yl)-1H-1,2,4-triazol-5-yl)piperazin-1-yl)pyrazolo[1,5-a]pyridine
Figure imgf000170_0001
[0468] To a solution of the mixture of 3-bromo-1-(tetrahydro-2H-pyran-2-yl)-1H-1,2,4- triazole and 3-bromo-4-(tetrahydro-2H-pyran-2-yl)-4H-1,2,4-triazole and 5-bromo-1- (tetrahydro-2H-pyran-2-yl)-1H-1,2,4-triazole (600 mg, 2.58 mmol, as prepared in the previous step) and 6-(1-methyl-1H-pyrazol-4-yl)-3-piperazin-1-ylpyrazolo[1,5-a]pyridine (Compound 12) (364.9 mg, 1.29 mmol) in dioxane (20 mL) were added Cs2CO3 (1.68 g, 5.17 mmol) and Pd-PEPPSI-IPentCl 2-methylpyridine (326.1 mg, 0.38 mmol), then the reaction was heated to 100°C and stirred overnight under nitrogen. The mixture was cooled to rt then concentrated under reduced pressure. The residue was dissolved in water (100 mL) and extracted with DCM (3 x 100 mL). The combined organic extracts were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by reversed-phase flash chromatography with a C18 silica gel column eluting with 10% to 50% ACN / water (10mM NH4HCO3 to afford 450 mg (40%) of a mixture of 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(tetrahydro-2H-pyran-2-yl)-1H- 1,2,4-triazol-3-yl)piperazin-1-yl)pyrazolo[1,5-a]pyridine, 6-(1-methyl-1H-pyrazol-4-yl)- 3-(4-(4-(tetrahydro-2H-pyran-2-yl)-4H-1,2,4-triazol-3-yl)piperazin-1-yl)pyrazolo[1,5- a]pyridine, and 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(tetrahydro-2H-pyran-2-yl)-1H- 1,2,4-triazol-5-yl)piperazin-1-yl)pyrazolo[1,5-a]pyridine as a yellow oil. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 – 8.76 (m, 1H), 8.36 (s, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.78 (s, 1H), 7.67 (dd, J = 9.3, 0.9 Hz, 1H), 7.32 (dd, J = 9.2, 1.5 Hz, 1H), 5.33 (dd, J = 9.9, 2.3 Hz, 1H), 3.97 – 3.90 (m, 1H), 3.87 (s, 3H), 3.67 – 3.57 (m, 1H), 3.49-3.47 (m, 4H), 3.07 (t, J = 5.0 Hz, 4H), 2.05 – 1.90 (m, 3H), 1.67 – 1.52 (m, 3H). MS (ESI) m/z [M+H]+ calcd. for C22H27N9O, 434.2; found, 434.2.LCMS purity:254 nm:90%. Step 3. 3-(4-(1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine (Compound S102)
Figure imgf000171_0001
[0469] To a solution of a mixture of 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(tetrahydro-2H- pyran-2-yl)-1H-1,2,4-triazol-3-yl)piperazin-1-yl)pyrazolo[1,5-a]pyridine, 6-(1-methyl- 1H-pyrazol-4-yl)-3-(4-(4-(tetrahydro-2H-pyran-2-yl)-4H-1,2,4-triazol-3-yl)piperazin-1- yl)pyrazolo[1,5-a]pyridine, and 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(tetrahydro-2H- pyran-2-yl)-1H-1,2,4-triazol-5-yl)piperazin-1-yl)pyrazolo[1,5-a]pyridine (400 mg, 0.92 mmol, as prepared in the previous step) in DCM (20 mL) was added 4M HCl in dioxane (336.4 mg, 9.23 mmol) then the reaction was stirred at rt for 2 h under nitrogen. The mixture was concentrated under reduced pressure then the pH of the residue was adjusted to pH 7 with aqueous NH3. The residue was purified by reversed-phase flash chromatography with a C18 silica gel column eluting with10% to 50% ACN / water (10mM NH4HCO3) to afford 270 mg (84%) of 3-(4-(1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine (Compound S102) as a yellow oil.1H NMR (400 MHz, DMSO- d6) δ (ppm) 13.13-12.70 (m, 1H), 8.82 (s, 1H), 8.57-8.20 (m, 2H), 7.97 (s, 1H), 7.78 (s, 1H), 7.68 (d, J = 9.2 Hz, 1H), 7.34-7.30 (m, 1H), 3.87 (s, 3H), 3.50 (t, J = 4.8 Hz, 4H), 3.08 (t, J = 4.8 Hz, 4H). MS (ESI) m/z [M+H]+ calcd. for C17H19N9, 350.2; found, 350.2.LCMS purity:254 nm:95%. Synthesis of 1-[6-(1-methylpyrazol-4-yl)pyrazolo[1,5-a]pyrazin-3-yl]piperazine (Compound S103)
Figure imgf000171_0002
[0470] To a solution of 3-bromo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrazine (Compound S5) (900 mg, 3.25 mmol) in dioxane (20 mL) was added piperazine (1.39 g, 16.18 mmol), Pd-PEPPSI-IPentCl 2-methylpyridine (272.2 mg, 0.32 mmol), and Cs2CO3 (2.11 g, 6.47 mmol) then the mixture was heated to 100°C and stirred for 16 h under nitrogen. The mixture was cooled to rt, diluted with water (50 mL), and extracted with EtOAc (3 x 50 mL). The combined organic extracts were washed with brine (3 x 10 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by reversed-phase flash chromatography with a C18 silica gel column eluting with 10% to 100% ACN / water (0.1% FA) to afford 500 mg (42%) of 1-[6-(1-methylpyrazol-4-yl)pyrazolo[1,5-a]pyrazin-3-yl]piperazine as a light yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.17 (s, 1H), 8.89 (s, 1H), 8.28 (d, J = 5.6 Hz, 1H), 8.21 (s, 1H), 8.00 (s, 1H), 7.81 (s, 1H), 3.88 (s, 3H), 3.21 – 3.11 (m, 4H), 3.08 – 2.94 (m, 4H); MS (ESI) m/z [M+H]+ calcd. for C14H17N7, 284.1; found, 284.1; LCMS purity: 254 nm: 98.6%. Synthesis of 1-(2-methylbenzo[d]thiazol-6-yl)ethan-1-one (Compound S104)
Figure imgf000172_0001
[0471] To a solution of 6-bromo-2-methylbenzo[d]thiazole (5 g, 21.92 mmol) and tributyl(1-ethoxyvinyl)stannane (11.87 g, 32.88 mmol) in dioxane (500 mL) was added Pd(PPh3)4 (2.53 g, 2.19 mmol) then the mixture was heated to 90°C and stirred for 16 h under nitrogen. The reaction was cooled to rt and concentrated under reduced pressure. Water (150 mL) was added to the residue then the mixture was extracted with EtOAc (3x300 mL). The organic extracts were combined, washed with brine (50 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with PE/EtOAc (1:1) to afford 3.5 g (83%) of 1-(2-methylbenzo[d]thiazol-6-yl)ethan-1-one (Compound S104) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.72 (d, J = 1.8 Hz, 1H), 8.06 – 7.95 (m, 2H), 2.85 (s, 3H), 2.65 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C10H9NOS, 192.0; found, 192.1. Synthesis of (2-methylbenzo[d]thiazol-6-yl)methanol (Compound S105)
Figure imgf000172_0002
[0472] To a stirred solution of 2-methylbenzo[d]thiazole-6-carboxylic acid (1 g, 5.17 mmol) in THF (20 mL) cooled to 0°C was added a 2M solution of LiAlH4 in THF (5.2 mL) then the mixture was warmed to rt and stirred for 3 h. The reaction was cooled to 0°C and quenched by the addition of saturated aqueous NaHCO3 solution. The mixture was filtered, and the filter cake was washed with DCM (3x30 mL). The filtrate was extracted with DCM/MeOH (10:1) (3x80 mL), then the organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel column chromatography, eluted with PE/EtOAc (1:1) to afford 400 mg (43%) of (2-methylbenzo[d]thiazol-6-yl)methanol (Compound S105) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 7.93 (d, J = 1.5 Hz, 1H), 7.84 (d, J = 8.3 Hz, 1H), 7.41 (dd, J = 8.3, 1.5 Hz, 1H), 5.31 (t, J = 5.7 Hz, 1H), 4.61 (d, J = 5.7 Hz, 2H), 2.78 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C9H9NOS, 180.0; found, 180.1. Synthesis of 2-(hydroxymethyl)-5-methylbenzonitrile (Compound S106)
Figure imgf000173_0001
[0473] To a stirred solution of methyl 2-cyano-4-methylbenzoate (500 mg, 2.85 mmol) and NaBH4 (323.91 mg, 8.56 mmol) in MeOH (10 mL) cooled to 0°C was added LiCl (241.97 mg, 5.70 mmol) in portions then the mixture was warmed to rt and stirred for 1 h. The reaction was diluted with water (30 mL) and extracted with EtOAc (3x50 mL). The organic extracts were combined, washed with brine (2x2 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure to afford 400 mg (38%) of 2- (hydroxymethyl)-5-methylbenzonitrile (Compound S106) as a yellow solid. MS (ESI) m/z [M+H]+ calcd. for C9H9NO, 148.2; found, 148.0. Synthesis of 2-(hydroxymethyl)-5-(trifluoromethyl)benzonitrile (Compound S107)
Figure imgf000173_0002
Step 1. Preparation of methyl 2-cyano-4-(trifluoromethyl)benzoate
Figure imgf000173_0003
[0474] To a solution of 2-bromo-5-(trifluoromethyl)benzonitrile (5 g, 19.99 mmol) in MeOH (30 mL) was added Pd(dppf)Cl2 (1.46 g, 2 mmol) in a pressure vessel then the mixture was pressurized to 15 atm with carbon monoxide at 80°C overnight. The reaction was cooled to rt and filtered, then the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography eluting with PE/EtOAc (1:1) to afford 3 g (65%) of methyl 2-cyano-4-(trifluoromethyl)benzoate as a brown solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.55 – 8.50 (m, 1H), 8.30 (d, J = 8.2 Hz, 1H), 8.24 (dd, J = 8.3, 1.9 Hz, 1H), 3.96 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C10H6F3NO2, 229.0; found, 229.2. Step 2. Preparation of 2-(hydroxymethyl)-5-(trifluoromethyl)benzonitrile (Compound S107)
Figure imgf000174_0001
[0475] To a stirred solution of methyl 2-cyano-4-(trifluoromethyl)benzoate (2.9 g, 12.65 mmol, as prepared in the previous step) in MeOH (29 mL) cooled to 0°C was added NaBH4 (0.96 g, 25.3 mmol) in portions then the mixture was warmed to rt and stirred for 2 h. The reaction was concentrated under reduced pressure then the residue was dissolved in water (300 mL) and extracted with EtOAc (2x200 mL). The organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure to afford 1.5 g (59%) of 2-(hydroxymethyl)-5-(trifluoromethyl)benzonitrile (Compound S107) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.05 – 7.94 (m, 2H), 7.82 (d, J = 3.8 Hz, 1H), 5.43 (d, J = 8.7 Hz, 2H), 4.55 – 4.53 (m, 1H); MS (ESI) m/z [M+H]+ calcd. for C9H6F3NO, 201.0; found, 201.1. Synthesis of 1-(4-(fluoromethyl)phenyl)ethan-1-ol (Compound S108)
Figure imgf000174_0002
Step 1. Preparation of 1-(4-(fluoromethyl)phenyl)ethan-1-one
Figure imgf000174_0003
[0476] To a stirred solution of 1-(p-tolyl)ethan-1-one (3 g, 22.3 mmol) and K2S2O8 (18.13 g, 67 mmol) in ACN (30 mL) and H2O (30 mL) was added F-TEDA (23.7 g, 67 mmol) in portions at rt then the mixture was heated to 80°C and stirred overnight. The reaction was cooled to rt and extracted with EtOAc (3x200 mL). The organic extracts were combined, washed with brine (3x100 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with PE/EtOAc (1:1) to afford 2 g (59%) of 1-(4- (fluoromethyl)phenyl)ethan-1-one as colorless liquid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.00 (dd, J = 8.3, 1.3 Hz, 2H), 7.62 – 7.49 (m, 2H), 5.53 (d, J = 48 Hz, 2H), 2.60 (d, J = 0.8 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C9H9FO, 153.0; found, 153.0. Step 2. Preparation of 1-(4-(fluoromethyl)phenyl)ethan-1-ol (Compound S108)
Figure imgf000175_0001
[0477] To a solution of 1-(4-(fluoromethyl)phenyl)ethan-1-one (800 mg, 5.2 mmol, as prepared in the previous step) in MeOH (10 mL) cooled to 0°C was added NaBH4 (238 mg, 6.3 mmol) then the mixture was stirred for 1 h. The reaction was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with PE/EtOAc (1:1) to afford 600 mg (74%) of 1-(4-(fluoromethyl)phenyl)ethan-1-ol (Compound S108) as colorless liquid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 7.43 – 7.29 (m, 4H), 5.38 (d, J = 48 Hz, 2H), 5.20 (d, J = 4.2 Hz, 1H), 4.87 – 4.68 (m, 1H), 1.33 (d, J = 6.4 Hz, 3H). Synthesis of 1-(4-(difluoromethyl)phenyl)ethan-1-ol (Compound S109)
Figure imgf000175_0002
[0478] To a solution of 1-(4-(difluoromethyl)phenyl)ethan-1-one (600 mg, 3.5 mmol) in MeOH (8 mL) cooled to 0°C was added NaBH4 (267 mg, 7 mmol) then the mixture was stirred for 2 h. The reaction was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with PE/EtOAc (1:1) to afford 550 mg (91%) of 1-(4-(difluoromethyl)phenyl)ethan-1-ol (Compound S109) as a colorless oil. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 7.76 – 7.41 (m, 4H), 7.01 (t, J = 56.0 Hz, 1H), 5.29 (d, J = 4.3 Hz, 1H), 4.95 – 4.61 (m, 1H), 1.34 (d, J = 6.5 Hz, 3H). Synthesis of 2-((tert-butyldimethylsilyl)oxy)-1-(p-tolyl)ethan-1-ol (Compound S110) and 2-((tert-butyldimethylsilyl)oxy)-2-(p-tolyl)ethan-1-ol (Compound S111)
Figure imgf000176_0003
Step 1. Preparation of 2-hydroxy-1-(p-tolyl)ethan-1-one
Figure imgf000176_0001
[0479] To a solution of 2-bromo-1-(4-methylphenyl)ethanone (3 g, 14.08 mmol) in MeOH (20 mL) was added NaOAc (9.24 g, 112.64 mmol) then the mixture was heated to 80°C and stirred for 12 h under nitrogen. The reaction was cooled to rt and concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with PE/EtOAc (5:1) to afford 1.47 g (70%) of 2-hydroxy-1-(4-methylphenyl)ethanone as a white solid.1H NMR (400 MHz, CDCl3) δ (ppm) 7.82 (d, J = 8.2 Hz, 2H), 7.30 (d, J = 8.0 Hz, 2H), 4.85 (s, 2H), 3.50 (d, J = 7.4 Hz, 1H), 2.43 (s, 3H). Step 2. Preparation of 2-((tert-butyldimethylsilyl)oxy)-1-(p-tolyl)ethan-1-one
Figure imgf000176_0002
[0480] To a solution of 2-hydroxy-1-(4-methylphenyl)ethanone (1.27 g, 8.45 mmol, as prepared in the previous step) in THF (20 mL) was added TBSCl (1.91 g, 12.68 mmol) and imidazole (0.87 g, 12.68 mmol) then the mixture was stirred at rt for 2 h. The reaction was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with PE/EtOAc (5:1) to afford 2.17 g (97%) of 2-((tert- butyldimethylsilyl)oxy)-1-(p-tolyl)ethan-1-one as a colorless liquid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 7.83 (d, J = 8.2 Hz, 2H), 7.32 (d, J = 8.0 Hz, 2H), 4.99 (s, 2H), 2.37 (s, 3H), 0.90 (s, 9H), 0.07 (s, 6H). Step 3. Preparation of 2-((tert-butyldimethylsilyl)oxy)-1-(p-tolyl)ethan-1-ol (Compound S110) and 2-((tert-butyldimethylsilyl)oxy)-2-(p-tolyl)ethan-1-ol (Compound S111)
Figure imgf000177_0003
[0481] To a stirred solution of 2-((tert-butyldimethylsilyl)oxy)-1-(p-tolyl)ethan-1-one (1 g, 3.78 mmol, as prepared in the previous step) in MeOH (10 mL) cooled to 0°C was added NaBH4 (0.22 g, 5.673 mmol) in portions then the mixture was warmed to rt and stirred at rt for 2 h under nitrogen. The reaction was concentrated under reduced pressure then the residue was purified by reversed-phase flash chromatography using a Prep Phenyl column eluting with 35-65% MeOH/water (10mM NH4HCO3) to afford 250 mg (25%) of 2-((tert- butyldimethylsilyl)oxy)-1-(p-tolyl)ethan-1-ol (Compound S110) as colorless liquid and 200 mg (20%) of 2-((tert-butyldimethylsilyl)oxy)-2-(p-tolyl)ethan-1-ol (Compound S111) as a colorless oil. [0482] 2-((tert-butyldimethylsilyl)oxy)-1-(p-tolyl)ethan-1-ol (Compound S110).1H NMR (400 MHz, DMSO-d6) δ (ppm) 7.21 (d, J = 8.0 Hz, 2H), 7.10 (d, J = 7.9 Hz, 2H), 5.16 (d, J = 4.3 Hz, 1H), 4.49 (q, J = 5.7 Hz, 1H), 3.65 – 3.58 (m, 1H), 3.55 – 3.48 (m, 1H), 2.27 (s, 3H), 0.81 (s, 9H), -0.04 (s, 3H), -0.07 (s, 3H). [0483] 2-((tert-butyldimethylsilyl)oxy)-2-(p-tolyl)ethan-1-ol (Compound S111).1H NMR (400 MHz, DMSO-d6) δ (ppm) 7.20 (d, J = 8.0 Hz, 2H), 7.12 (d, J = 7.9 Hz, 2H), 4.70 (t, J = 5.7 Hz, 1H), 4.66 – 4.61 (m, 1H), 3.44 – 3.31 (m, 2H), 2.28 (s, 3H), 0.85 (s, 9H), 0.05 (s, 3H), -0.05 (s, 3H). Synthesis of 1-(1,5-dimethyl-1H-pyrazol-3-yl)ethan-1-ol (Compound S112)
Figure imgf000177_0001
Step 1. Preparation of 1-(1,5-dimethyl-1H-pyrazol-3-yl)ethan-1-one
Figure imgf000177_0002
[0484] To a solution of 1-(5-methyl-1H-pyrazol-3-yl)ethan-1-one (1 g, 8.05 mmol) in DCM (20 mL) was added MeI (1.72 g, 12.08 mmol) then the mixture was stirred at rt for 2 h. The reaction was filtered, and the filter cake was washed with DCM (2x20 mL), then the filtrate was concentrated under reduced pressure. The residue was purified by reversed- phase flash chromatography using a C18 silica gel column eluting with 20-30% ACN/water (10mM NH4HCO3) to afford 240 mg (22%) of 1-(1,5-dimethyl-1H-pyrazol-3-yl)ethan-1- one as a white solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 6.47 (s, 1H), 3.81 (s, 3H), 2.41 (s, 3H), 2.27 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C7H10N2O, 139.1; found, 139.2. Step 2. Preparation of 1-(1,5-dimethyl-1H-pyrazol-3-yl)ethan-1-ol (Compound S112)
Figure imgf000178_0002
[0485] To a stirred mixture of 1-(1,5-dimethyl-1H-pyrazol-3-yl)ethan-1-one (220 mg, 1.59 mmol, as prepared in the previous step) in MeOH (5 mL) cooled to 0°C was added NaBH4 (90.3 mg, 2.38 mmol) in portions then the mixture was warmed to rt and stirred for 2 h. The reaction was cooled to 0°C, quenched with water, and extracted with EtOAc (3x20 mL). The organic extracts were combined, washed with water (3x20 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by reversed-phase flash chromatography using a C18 silica gel column eluting with 5-10% ACN/water (10mM NH4HCO3) to afford 140 mg (63%) of 1- (1,5-dimethyl-1H-pyrazol-3-yl)ethan-1-ol (Compound S112) as yellow liquid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 5.92 (s, 1H), 4.83 (d, J = 4.7 Hz, 1H), 4.63 – 4.52 (m, 1H), 3.63 (s, 3H), 2.19 (s, 3H), 1.29 (d, J = 6.5 Hz, 3H). Synthesis of 3-(4-cyclopropylphenyl)propanoic acid (Compound S113)
Figure imgf000178_0001
[0486] To a stirred mixture of 3-(4-bromophenyl)propanoic acid (1 g, 4.36 mmol) and cyclopropylboronic acid (449.9 mg, 5.23 mmol) in dioxane (10 mL) were added Pd(dppf)Cl2 (319.4 mg, 0.43 mmol) and Cs2CO3 (2.8 g, 8.73 mmol) in portions at rt under nitrogen then the mixture was heated to 100°C and stirred for 2 h. The reaction was cooled to rt, diluted with water (200 mL), and extracted with DCM (2x150 mL). The organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by reversed-phase flash chromatography using a C18 silica gel column eluting with 5-100% ACN/water (10mM NH4HCO3) to afford 500 mg (60%) of 3-(4-cyclopropylphenyl)propanoic acid (Compound S113) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 7.08 (d, J = 7.9 Hz, 2H), 6.97 (d, J = 7.9 Hz, 2H), 2.75 (t, J = 7.7 Hz, 2H), 2.44 (t, J = 7.7 Hz, 2H), 1.92 – 1.78 (m, 1H), 0.96 – 0.82 (m, 2H), 0.67 – 0.55 (m, 2H). Synthesis of 1-(4-(2,2,2-trifluoroethyl)phenyl)ethan-1-ol (Compound S114)
Figure imgf000179_0001
Step 1. Preparation of 1-(4-(2,2,2-trifluoroethyl)phenyl)ethan-1-one
Figure imgf000179_0002
[0487] To a stirred mixture of 4-acetylphenylboronic acid (1 g, 6.1 mmol) and 1,1,1- trifluoro-2-iodoethane (5.1 g, 24.3 mmol) in dioxane (20 mL) were added Cs2CO3 (5.96 g, 18.3 mmol), Pd2(dba)3 (1.1 g, 1.2 mmol), and XantPhos (705 mg, 1.2 mmol) in portions at rt under nitrogen then the mixture was heated to 100°C and stirred overnight under nitrogen. The reaction was cooled to rt and concentrated under reduced pressure. The residue was purified by silica gel column chromatography eluting with PE/EtOAc (5:1) to afford 800 mg (65%) of 1-(4-(2,2,2-trifluoroethyl)phenyl)ethan-1-one as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.02 – 7.95 (m, 2H), 7.52 (d, J = 8.0 Hz, 2H), 3.77 (q, J = 11.5 Hz, 2H), 2.59 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C10H9F3O, 203.1; found, 203.1. Step 2. Preparation of 1-(4-(2,2,2-trifluoroethyl)phenyl)ethan-1-ol (Compound S114)
Figure imgf000179_0003
[0488] To a stirred solution of 1-(4-(2,2,2-trifluoroethyl)phenyl)ethan-1-one (400 mg, 2 mmol) in MeOH (10 mL) cooled to 0°C was added NaBH4 (89.8 mg, 2.4 mmol) in portions then the mixture was warmed to rt and stirred for 1 h. The reaction was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with PE/EtOAc (1:1) to afford 150 mg (37%) of 1-(4-(2,2,2-trifluoroethyl)phenyl)ethan-1- ol (Compound S114) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 7.38 – 7.32 (m, 2H), 7.32 – 7.23 (m, 2H), 5.16 (d, J = 4.2 Hz, 1H), 4.85 – 4.53 (m, 1H), 3.61 (q, J = 11.6 Hz, 2H), 1.32 (d, J = 6.4 Hz, 3H). Synthesis of 1-(4-(1,1-difluoroethyl)phenyl)ethan-1-ol (Compound S115)
Figure imgf000180_0001
Step 1. Preparation of 1-bromo-4-(1,1-difluoroethyl)benzene
Figure imgf000180_0002
[0489] To a solution of 1-(4-bromophenyl)ethan-1-one (10 g, 50.23 mmol) in DCM (60 mL) was added DAST (64.7 g, 401.9 mmol) dropwise at rt then the mixture was heated to 50°C and stirred overnight. The reaction was cooled to 0°C, quenched with water, and extracted with DCM (2x100 mL). The organic extracts were combined, washed with water (2x100 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with DCM/PE (50:1) to afford 3.4 g (31%) of 1-bromo-4-(1,1- difluoroethyl)benzene as colorless liquid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 7.70 (d, J = 8.7 Hz, 2H), 7.53 (d, J = 8.7 Hz, 2H), 1.97 (t, J = 18.9 Hz, 3H). Step 2. Preparation of 1-(4-(1,1-difluoroethyl)phenyl)ethan-1-ol (Compound S115)
Figure imgf000180_0003
[0490] To a stirred solution of 1-bromo-4-(1,1-difluoroethyl)benzene (500 mg, 2.26 mmol, as prepared in the previous step) in THF (10 mL) cooled to -78°C was added 2.5M nBuLi in hexane (1.4 mL, 3.39 mmol) dropwise under nitrogen then the mixture was stirred at - 78°C for 30 minutes under nitrogen. A solution of 5M acetaldehyde in THF (0.6 mL, 2.71 mmol) was added dropwise over 5 minutes at -78°C then the reaction was warmed to rt and stirred for 1 h. The reaction was cooled to 0°C, quenched with saturated aqueous NH4Cl solution, and extracted with EtOAc (2x30 mL). The organic extracts were combined, washed with water (2x30 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by reversed-phase flash chromatography using a C18 silica gel column eluting with 30-40% ACN/water (10mM NH4HCO3) to afford 180 mg (43%) of 1-(4-(1,1-difluoroethyl)phenyl)ethan-1-ol (Compound S115) as yellow liquid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 7.50 (d, J = 8.3 Hz, 2H), 7.44 (d, J = 8.2 Hz, 2H), 5.25 (d, J = 4.3 Hz, 1H), 4.81 – 4.71 (m, 1H), 1.95 (t, J = 18.8 Hz, 3H), 1.32 (d, J = 6.5 Hz, 3H). Synthesis of 2-(4-(trifluoromethyl)phenyl)propanoic acid (Compound S116)
Figure imgf000181_0001
Step 1. Preparation of methyl 2-(4-(trifluoromethyl)phenyl)acetate
Figure imgf000181_0002
[0491] To a stirred solution of 2-(4-(trifluoromethyl)phenyl)acetic acid (1.2 g, 5.878 mmol) in MeOH (20 mL) was added conc. H2SO4 (1.15 g, 11.756 mmol) dropwise at rt then the mixture was heated to 80°C and stirred overnight. The reaction was cooled to rt, diluted with water (50 mL), and the pH was adjusted to pH 7 with saturated aqueous NaHCO3 solution. The resulting mixture was extracted with Et2O (3x100mL) then the organic extracts were combined, washed with brine (3x10 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with PE/EtOAc (1:1) to afford 800 mg (62%) of methyl 2-(4-(trifluoromethyl)phenyl)acetate as a yellow oil. The product was used without further purification. Step 2. Preparation of methyl 2-(4-(trifluoromethyl)phenyl)propanoate
Figure imgf000181_0003
[0492] To a stirred solution of methyl 2-(4-(trifluoromethyl)phenyl)acetate (500 mg, 2.292 mmol, as prepared in the previous step) in THF (20 mL) cooled to -78°C was added NaH (60% wt., 183.2 mg, 4.584 mmol) in portions under nitrogen then the mixture was stirred at -78°C for 30 minutes under nitrogen. To the stirred reaction at -78°C was added MeI (650.5 mg, 4.584 mmol) in portions then the mixture was warmed to rt and stirred overnight. The reaction was poured into a water /ice mixture, then extracted with EtOAc (3x100 mL). The organic extracts were combined, washed with brine (3x50 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with PE/EtOAc (1:1) to afford 130 mg (24%) of methyl 2-(4-(trifluoromethyl)phenyl)propanoate as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 7.70 (d, J = 8.0 Hz, 2H), 7.55 (d, J = 8.0 Hz, 2H), 3.97 (q, J = 7.2 Hz, 1H), 3.61 (s, 3H), 1.43 (d, J = 7.2 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C11H11F3O, 231.0; found, 231.2. Step 3. Preparation of 2-(4-(trifluoromethyl)phenyl)propanoic acid (Compound S116)
Figure imgf000182_0001
[0493] To a stirred solution of methyl 2-(4-(trifluoromethyl)phenyl)propanoate (110 mg, 0.474 mmol, as prepared in the previous step) in MeOH (10 mL) was added LiOH (22.6 mg, 0.948 mmol) in portions at rt then the mixture was heated to 50°C and stirred overnight. The reaction was cooled to rt, diluted with water (20 mL), and the pH was adjusted to pH 4 with 1M HCl solution. The mixture was extracted with EtOAc (3x50 mL) then the organic extracts were combined, washed with brine (3x10 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with PE/EtOAc (1:1) to afford 150 mg (145%) of 2-(4-(trifluoromethyl)phenyl)propanoic acid (Compound S116) as a yellow green solid. The solid was used without further purification. MS (ESI) m/z [M-H]- calcd. for C10H9F3O2, 217.2; found, 217.0. Synthesis of 1-(2-methylquinolin-6-yl)ethan-1-ol (Compound S117)
Figure imgf000182_0002
Step 1. Preparation of N-methoxy-N,2-dimethylquinoline-6-carboxamide
Figure imgf000183_0001
[0494] To a stirred mixture of 2-methylquinoline-6-carboxylic acid (2 g, 10.7 mmol) and N,O-dimethylhydroxylamine hydrochloride (1.5 g, 16.11 mmol) in DCM (30 mL) were added HATU (6.1 g, 16 mmol) and DIEA (5.5 g, 42.71 mmol) then the mixture was stirred at rt for 3 h. The reaction was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with PE/EtOAc (1:1) to afford 1.6 g (65%) of N-methoxy-N,2-dimethylquinoline-6-carboxamide as a white solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.35 (d, J = 8.4 Hz, 1H), 8.22 (d, J = 1.8 Hz, 1H), 8.00 – 7.93 (m, 1H), 7.88 (dd, J = 8.7, 1.9 Hz, 1H), 7.48 (d, J = 8.4 Hz, 1H), 3.57 (s, 3H), 3.32 (s, 3H), 2.68 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C13H14N2O2, 231.1; found, 231.1. Step 2.1-(2-methylquinolin-6-yl)ethan-1-one
Figure imgf000183_0002
[0495] To a stirred solution of N-methoxy-N,2-dimethylquinoline-6-carboxamide (1.5 g, 6.5 mmol, as prepared in the previous step) in THF (20 mL) cooled to -78°C was added 3M MeMgBr in THF (5 mL, 19.5 mmol) dropwise under nitrogen then the mixture was stirred at -78°C for 1 h under nitrogen. The reaction was warmed to 0°C, quenched with saturated aqueous NH4Cl solution, and extracted with DCM (3x300 mL). The organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with PE/EtOAc (1:1) to afford 800 mg (66%) of 1-(2- methylquinolin-6-yl)ethan-1-one as a white solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.67 (d, J = 2.1 Hz, 1H), 8.49 – 8.39 (m, 1H), 8.17 (dd, J = 8.8, 2.0 Hz, 1H), 7.99 (d, J = 8.8 Hz, 1H), 7.53 (d, J = 8.4 Hz, 1H), 2.71 (s, 3H), 2.70 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C12H11NO, 186.1; found, 186.1. Step 3.1-(2-methylquinolin-6-yl)ethan-1-ol (Compound S117)
Figure imgf000184_0001
[0496] To a mixture of 1-(2-methylquinolin-6-yl)ethan-1-one (500 mg, 2.7 mmol, as prepared in the previous step) in MeOH (10 mL) cooled to 0°C was added NaBH4 (204 mg, 5.4 mmol) then the mixture was stirred for 1 h. The reaction was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with PE/EtOAc (1:1) to afford 320 mg (63%) of 1-(2-methylquinolin-6-yl)ethan-1-ol (Compound S117) as colorless oil.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.21 (dd, J = 8.5, 0.8 Hz, 1H), 7.88 (d, J = 8.7 Hz, 1H), 7.84 (d, J = 2.0 Hz, 1H), 7.71 (dd, J = 8.7, 2.0 Hz, 1H), 7.38 (d, J = 8.4 Hz, 1H), 5.37 (d, J = 4.2 Hz, 1H), 5.07 – 4.86 (m, 1H), 2.64 (s, 3H), 1.42 (d, J = 6.4 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C12H13NO, 188.1; found, 188.1. Synthesis of 1-(2-methylquinolin-7-yl)ethan-1-ol (Compound S118)
Figure imgf000184_0002
[0497] To a solution of 7-bromo-2-methylquinoline (500 mg, 2.25 mmol) in THF (10 mL) cooled to -78°C was added 2.5M nBuLi in hexane (1.4 mL, 3.37 mmol) dropwise over 5 minutes under nitrogen, followed by the dropwise addition of 5M acetaldehyde in THF solution (0.54 mL, 2.70 mmol) at -78°C then the mixture was warmed to rt and stirred for 1 h. The reaction was cooled to 0°C, quenched with saturated aqueous NH4Cl solution, and extracted with DCM (3x20 mL). The organic extracts were combined, washed with water (3x20 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by reversed-phase flash chromatography using a C18 silica gel column eluting with 40-50% ACN/water (10mM NH4HCO3) to afford 210 mg (50%) of 1-(2-methylquinolin-7-yl)ethanol (Compound S118) as a yellow solid. MS (ESI) m/z [M+H]+ calcd. for C12H13NO, 188.1; found, 188.1. Synthesis of 1-(4-(oxetan-3-yl)phenyl)ethan-1-ol (Compound S119)
Figure imgf000185_0001
[0498] To a stirred solution of 3-(4-bromophenyl)oxetane (500 mg, 2.34 mmol) in THF (6 mL) cooled to -78°C was added 2.5M nBuLi in hexane (1.4 mL, 3.52 mmol) dropwise under nitrogen then the mixture was stirred for 30 minutes. To the stirred reaction was added 5M acetaldehyde in THF solution (0.56 mL, 2.81 mmol) dropwise at -78°C then the mixture was warmed to rt and stirred for 2 h. The reaction was quenched with water (100 mL) and extracted with EtOAc (2x150 mL). The organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by reversed-phase flash chromatography using a C18 silica gel column eluting with 5-100% ACN/water (10mM NH4HCO3) to afford 200 mg (48%) of 1-(4-(oxetan-3-yl)phenyl)ethan-1-ol (Compound S119) as a yellow solid. MS (ESI) m/z [M+H]+ calcd. for C11H14O2, 178.1; found, 178.2. Synthesis of 1-(4-isopropylphenyl)ethan-1-ol (Compound S120)
Figure imgf000185_0002
[0499] To a stirred solution of 1-(4-isopropylphenyl)ethan-1-one (500 mg, 3.1 mmol) in MeOH (10 mL) cooled to 0°C was added NaBH4 (140 mg, 3.7 mmol) in portions then the mixture was warmed to rt and stirred for 2 h. The reaction was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with PE/EtOAc (1:1) to afford 500 mg (99%) of 1-(4-isopropylphenyl)ethan-1-ol (Compound S120) as colorless oil.1H NMR (400 MHz, DMSO-d6) δ (ppm) 7.28 – 7.22 (m, 2H), 7.18 – 7.16 (m, 2H), 5.04 (d, J = 4.2 Hz, 1H), 4.73 – 4.62 (m, 1H), 2.92 – 2.79 (m, 1H), 1.30 (d, J = 6.5 Hz, 3H), 1.19 (d, J = 6.9 Hz, 6H). Synthesis of 1-(quinolin-7-yl)ethan-1-ol (Compound S121)
Figure imgf000185_0003
[0500] To a stirred solution of 1-(quinolin-7-yl)ethan-1-one (400 mg, 2.33 mmol) in MeOH (10 mL) cooled to 0°C was added NaBH4 (132.5 mg, 3.50 mmol) in portions then the mixture was warmed to rt and stirred for 2 h. The reaction was quenched with water (50 mL) and extracted with DCM (2x150 mL). The organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure to afford 350 mg (86%) of 1-(quinolin-7-yl)ethan-1-ol (Compound S121) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.89 (dd, J = 4.2, 1.8 Hz, 1H), 8.33 (dd, J = 8.6, 2.2 Hz, 1H), 8.00 – 7.90 (m, 2H), 7.62 (dd, J = 8.5, 1.7 Hz, 1H), 7.49 (dd, J = 8.3, 4.2 Hz, 1H), 5.45 – 5.38 (m, 1H), 5.01 – 4.91 (m, 1H), 1.44 (d, J = 6.4 Hz, 3H). Synthesis of 1-(4-(oxetan-2-yl)phenyl)ethan-1-ol (Compound S122)
Figure imgf000186_0001
[0501] To a stirred solution of 2-(4-bromophenyl)oxetane (500 mg, 2.34 mmol) in THF (10 mL) cooled to -78°C was added 2.5M nBuLi in hexane (1.4 mL, 3.52 mmol) dropwise under nitrogen followed by the addition of 5M acetaldehyde in THF solution (0.56 mL, 2.81 mmol) dropwise at -78°C then the mixture was warmed to rt and stirred for 1 h. The reaction was cooled to 0°C, quenched with saturated aqueous NH4Cl solution, and extracted with DCM (3x20 mL). The organic extracts were combined, washed with water (3x20 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by reversed-phase flash chromatography using a C18 silica gel column eluting with 15-20% ACN/water (10mM NH4HCO3) to afford 170 mg (41%) of 1-(4-(oxetan-2-yl)phenyl)ethan-1-ol (Compound S122) as a yellow liquid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 7.40 – 7.33 (m, 4H), 5.69 (t, J = 7.5 Hz, 1H), 5.13 (d, J = 4.2 Hz, 1H), 4.77 – 4.62 (m, 2H), 4.56 – 4.48 (m, 1H), 3.00 – 2.90 (m, 1H), 2.60 – 2.52 (m, 1H), 1.31 (d, J = 6.4 Hz, 3H). Synthesis of 1-(quinolin-6-yl)ethan-1-ol (Compound S123)
Figure imgf000186_0002
[0502] To a stirred solution of 1-(quinolin-7-yl)ethan-1-one (300 mg, 1.75 mmol) in MeOH (5 mL) cooled to 0°C was added NaBH4 (132.5 mg, 3.50 mmol) in portions then the mixture was warmed to rt and stirred for 2 h. The reaction was quenched with water (20 mL) and extracted with EtOAc (3x20 mL). The organic extracts were combined, washed with brine (2x3 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure to afford 300 mg (99%) of 1-(quinolin-7-yl)ethan-1-ol (Compound S123) as a colorless oil.1H NMR (400 MHz, CDCl3) δ (ppm) 8.89 (dd, J = 4.2, 1.6 Hz, 1H), 8.17 (d, J = 8.2 Hz, 1H), 8.11 (d, J = 8.7 Hz, 1H), 7.82 (d, J = 1.5 Hz, 1H), 7.74 (dd, J = 8.7, 1.9 Hz, 1H), 7.41 (dd, J = 8.3, 4.3 Hz, 1H), 5.12 (q, J = 6.5 Hz, 1H), 3.49 (s, 1H), 1.60 (d, J = 6.5 Hz, 3H). Synthesis of 1-(2-chloroquinolin-6-yl)ethan-1-ol (Compound S124)
Figure imgf000187_0001
Step 1. Preparation of 1-(2-chloroquinolin-6-yl)ethan-1-one
Figure imgf000187_0002
[0503] To a stirred solution of 6-bromo-2-chloroquinoline (1 g, 4.1 mmol) and tributyl(1- ethoxyethenyl)stannane (1.8 g, 4.9 mmol) in dioxane (15 mL) was added Pd(dppf)Cl2 (0.6 g, 0.8 mmol) in portions under nitrogen then the mixture was heated to 100°C and stirred overnight. The reaction was cooled to rt and 0.1M HCl solution (15 mL, 2 mmol) was added dropwise over 5 minutes. The reaction was stirred at rt for 1 h then extracted with DCM (3x300 mL). The organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with DCM/MeOH (10:1) to afford 200 mg (24%) of 1-(2-chloroquinolin-6-yl)ethan-1-one as a white solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.80 (d, J = 2.0 Hz, 1H), 8.65 (d, J = 8.6 Hz, 1H), 8.28 (dd, J = 8.8, 2.0 Hz, 1H), 8.05 (d, J = 8.9 Hz, 1H), 7.73 (d, J = 8.6 Hz, 1H), 2.73 (s, 3H). Step 2.1-(2-chloroquinolin-6-yl)ethan-1-ol (Compound S124)
Figure imgf000188_0001
[0504] To a solution of 1-(2-chloroquinolin-6-yl)ethan-1-one (190 mg, 0.9 mmol, as prepared in the previous step) in MeOH (10 mL) cooled to 0°C was added NaBH4 (41.9 mg, 1.1 mmol) in portions then the mixture was stirred at 0°C for 1 h. The reaction concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with PE/EtOAc (1:1) to afford 170 mg (89%) of 1-(2- chloroquinolin-6-yl)ethan-1-ol (Compound S124) as an off-white solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.45 (dd, J = 8.7, 0.8 Hz, 1H), 7.98 (d, J = 1.9 Hz, 1H), 7.92 (d, J = 8.7 Hz, 1H), 7.83 (dd, J = 8.8, 2.0 Hz, 1H), 7.57 (d, J = 8.6 Hz, 1H), 5.43 (d, J = 4.2 Hz, 1H), 4.98 – 4.88 (m, 1H), 1.42 (d, J = 6.5 Hz, 3H). Synthesis of 2-((tert-butyldimethylsilyl)oxy)-1-(4-fluorophenyl)ethan-1-ol (Compound S125) and 2-((tert-butyldimethylsilyl)oxy)-2-(4-fluorophenyl)ethan-1-ol (Compound S126)
Figure imgf000188_0003
Step 1. Preparation of 2-((tert-butyldimethylsilyl)oxy)-1-(4-fluorophenyl)ethan-1-one
Figure imgf000188_0002
[0505] To a stirred mixture of 1-(4-fluorophenyl)-2-hydroxyethan-1-one (1 g, 6.49 mmol) and imidazole (0.88 g, 12.9 mmol) in THF (20 mL) cooled to 0°C was added TBSCl (1.6 g, 9.73 mmol) in portions then the mixture was warmed to rt and stirred overnight. The reaction was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with PE/EtOAc (1:1) to afford 600 mg (34%) of 2- ((tert-butyldimethylsilyl)oxy)-1-(4-fluorophenyl)ethan-1-one as colorless oil. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.06 – 7.98 (m, 2H), 7.40 – 7.32 (m, 2H), 5.03 (s, 2H), 0.90 (s, 9H), 0.08 (s, 6H). Step 2. Preparation of 2-((tert-butyldimethylsilyl)oxy)-1-(4-fluorophenyl)ethan-1-ol (Compound S125) and 2-((tert-butyldimethylsilyl)oxy)-2-(4-fluorophenyl)ethan-1-ol (Compound S126)
Figure imgf000189_0003
[0506] To a stirred solution of 2-((tert-butyldimethylsilyl)oxy)-1-(4-fluorophenyl)ethan-1- one (1 g, 3.72 mmol, as prepared in the previous step) in MeOH (20 mL) cooled to 0°C was added NaBH4 (84.5 mg, 2.23 mmol) in portions then the mixture was warmed to rt and stirred overnight. The reaction was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with PE/EtOAc (1:1) to afford 700 mg (69%) of 2-((tert-butyldimethylsilyl)oxy)-1-(4-fluorophenyl)ethan-1-ol (Compound S125) as a yellow solid and 350 mg (35%) of 2-((tert-butyldimethylsilyl)oxy)- 2-(4-fluorophenyl)ethan-1-ol (Compound S126) as a yellow solid. [0507] 2-((tert-butyldimethylsilyl)oxy)-1-(4-fluorophenyl)ethan-1-ol (Compound S125). 1H NMR (400 MHz, DMSO-d6) δ (ppm) 7.42 – 7.32 (m, 2H), 7.18 – 7.08 (m, 2H), 5.33 (d, J = 4.4 Hz, 1H), 4.60 – 4.51 (m, 1H), 3.66 (dd, J = 10.1, 6.0 Hz, 1H), 3.53 (dd, J = 10.1, 5.9 Hz, 1H), 0.81 (s, 9H), -0.05 (s, 3H), -0.08 (s, 3H). [0508] 2-((tert-butyldimethylsilyl)oxy)-2-(4-fluorophenyl)ethan-1-ol (Compound S126). 1H NMR (400 MHz, DMSO-d6) δ (ppm) 7.40 – 7.30 (m, 2H), 7.18 – 7.09 (m, 2H), 4.77 (t, J = 5.7 Hz, 1H), 4.68 (dd, J = 6.8, 5.2 Hz, 1H), 3.47 – 3.38 (m, 2H), 0.84 (s, 9H), 0.05 (s, 6H). Synthesis of 2-(4-(1-hydroxyethyl)phenyl)propan-2-ol (Compound S127)
Figure imgf000189_0001
Step 1. Preparation of 1-(4-(2-hydroxypropan-2-yl)phenyl)ethan-1-one
Figure imgf000189_0002
[0509] To a stirred mixture of N,O-dimethylhydroxylamine hydrochloride (5.47 g, 56.12 mmol) in THF (30 mL) cooled to 0°C was added 1.5M DIBAL-H in toluene (38 mL, 56.12 mmol) dropwise under nitrogen then the mixture was warmed to rt and stirred for 2 h. The reaction was cooled to 0°C, methyl 4-acetylbenzoate (5 g, 28.06 mmol) was added, and the mixture was stirred at 0 °C for 1 h. To the stirred mixture at 0°C was added 1M MeMgBr in THF (33.7 mL, 33.672 mmol) dropwise over 30 minutes then the reaction was stirred at 0°C for 2 h. The reaction was cooled to -78°C and 3M MeLi in Et2O (11.2 mL, 33.672 mmol) was added dropwise over 30 minutes then the mixture was stirred at -78°C for 1 h. The reaction was poured into a water/ice mixture and extracted with EtOAc (3x100 mL). The organic extracts were combined, washed with brine (3x10 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with PE/EtOAc (1:1) to afford 1 g (20%) of 1-(4-(2-hydroxypropan-2-yl)phenyl)ethan-1-one as yellow oil.1H NMR (400 MHz, DMSO-d6) δ (ppm) 7.92 – 7.90 (m, 2H), 7.59 – 7.57 (m, 2H), 2.58 (s, 3H), 1.59 (s, 6H). Step 2. Preparation of 2-(4-(1-hydroxyethyl)phenyl)propan-2-ol (Compound S127)
Figure imgf000190_0001
[0510] To a stirred solution of 1-(4-(2-hydroxypropan-2-yl)phenyl)ethan-1-one (1.8 g, 10.09 mmol, as prepared in the previous step) in MeOH (10 mL) cooled to 0°C was added NaBH4 (460 mg, 12.11 mmol) in portions under nitrogen then the mixture was warmed to rt and stirred overnight. The reaction was concentrated under reduced pressure to afford 300 mg of 2-(4-(1-hydroxyethyl)phenyl)propan-2-ol (Compound S127) was used directly without further purification. Example 1: Synthesis of Exemplary Compound 156 3-(4-(1-benzyl-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine (Compound 156)
Figure imgf000191_0001
Step 1. Preparation of 1-benzyl-3-chloro-1H-1,2,4-triazole
Figure imgf000191_0002
[0511] To a solution of 3-chloro-1H-1,2,4-triazole (5 g, 48.3 mmol) and benzyl bromide (9.09 g, 53.14 mmol) in DMF (200 mL) was added K2CO3 (13.35 g, 96.61 mmol) then the reaction was stirred at 80°C under nitrogen for 16 h. The mixture was cooled to rt, then concentrated under reduced pressure. The residue was filtered, then the filter cake was washed with DCM (3 x 100 mL). The filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with a C18 silica gel column eluting with 10% to 100% ACN / water (10mM NH4HCO3) to afford 3.9 g (42%) of 1-benzyl-3-chloro-1H-1,2,4-triazole as a white solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.76 (s, 1H), 7.46 – 7.26 (m, 5H), 5.40 (s, 2H).; MS (ESI) m/z [M+H]+ calcd. for C9H8ClN3, 194.1; found, 194.2. Step 2. 3-(4-(1-benzyl-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine (Compound 156)
Figure imgf000191_0003
[0512] To a solution of 1-benzyl-3-chloro-1H-1,2,4-triazole (5 g, 25.82 mmol, as prepared in the previous step) and 6-(1-methyl-1H-pyrazol-4-yl)-3-piperazin-1-ylpyrazolo[1,5- a]pyridine (Compound 12) (7.29 g, 25.82 mmol) in dioxane (200 mL) were added Cs2CO3 (16.83 g, 51.64 mmol) and Pd-PEPPSI-IPentCl 2-methylpyridine (2.17 g, 2.58 mmol). The reaction was stirred at 90°C under nitrogen for 16 h then the resulting mixture was concentrated under reduced pressure. The residue was diluted with water and extracted with DCM (6 x 100 mL). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with MeOH/DCM (1:15) then recrystallized from ACN/THF (5:1) to afford 5.14 g (45%) of 3-(4-(1-benzyl-1H- 1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 156) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.32 (s, 1H), 8.23 (s, 1H), 7.97 (s, 1H), 7.77 (s, 1H), 7.66 (d, J = 9.2 Hz, 1H), 7.43 – 7.25 (m, 6H), 5.24 (s, 2H), 3.87 (s, 3H), 3.46 (t, J = 4.8 Hz, 4H), 3.06 (t, J = 4.8 Hz, 4H); MS (ESI) m/z [M+H]+ calcd. for C24H25N9, 440.2; found, 440.05; LCMS purity: 254 nm: 99.7%. Example 2: Synthesis of Exemplary Compound 275 1-(5-benzylpyrimidin-2-yl)-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)-1,4- diazepan-2-one (Compound 275).
Figure imgf000192_0001
[0513] 6-(1-Methyl-1H-pyrazol-4-yl)-3-piperazin-1-ylpyrazolo[1,5-a]pyridine hydrochloride salt (Compound S19) (5.25 g, 16.5 mmol) was added to a solution of 1- chloro-4-(isocyanatomethyl)benzene (2.76 g, 16.5 mmol) in ACN (110 mL) and DIEA (14.0 mL, 80.4 mmol). The reaction was stirred at rt for 1 h, then the mixture was concentrated under reduced pressure. The residue was purified by silica gel chromatography eluting with acetone (0.1% TEA additive)/EtOAc to afford 5.90 g (80%) of N-(4-chlorobenzyl)-4-[6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl]piperazine-1-carboxamide (Compound 275) as a solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.81 (s, 1H), 8.22 (s, 1H) 7.96 (s, 1H) 7.77 (s, 1H), 7.66 (d, J = 8.7 Hz, 1H), 7.37 (m, 2H), 7.31 (m, 3H) 7.21 (t, J = 5.9 Hz, 1H), 4.25 (d, J = 5.7 Hz, 2H), 3.87 (s, 3H), 3.52 (m, 4H), 2.95 (m, 4H); MS (ESI) m/z [M+Na]+ calcd. for C23H24ClN7NaO: 472.2. Found: 472.2; HPLC purity: 210 nm: 98.7%; 254 nm: 98.3%. Example 3: Synthesis of Exemplary Compound 326 1-(5-benzylpyrimidin-2-yl)-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)-1,4- diazepan-2-one (Compound 326)
Figure imgf000193_0001
Step 1. Preparation of tert-butyl (3-((6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin- 3-yl)amino)propyl)carbamate
Figure imgf000193_0002
[0514] To a solution of 6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-amine (Compound S56) (1.0 g, 4.69 mmol) in DMF (30 mL) was added tert-butyl (3- bromopropyl)carbamate (1.34 g, 5.63 mmol) and K2CO3 (1.94 g, 14.07 mmol). The reaction was stirred at 90°C under nitrogen for 16 h, cooled to rt, and diluted with water (50 mL). The mixture was extracted with EtOAc (3 x 200 mL), then the combined organic extracts were washed with brine (3x100 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep- TLC (MeOH / DCM 1:20) to afford 500 mg (19%) of tert-butyl (3-((6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)amino)propyl)carbamate as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.67 (s, 1H), 8.17 (s, 1H), 7.93 (s, 1H), 7.60 (dd, J = 9.2, 1.2 Hz, 1H), 7.49 (s, 1H), 7.14 (dd, J = 9.2, 1.6 Hz, 1H), 6.88 (t, J = 5.6 Hz, 1H), 4.70 (t, J = 6.4 Hz, 1H), 3.86 (s, 3H), 3.09 – 2.98 (m, 4H), 1.73 – 1.62 (m, 2H), 1.38 (s, 9H); MS (ESI) m/z [M+H]+ calcd. for C19H26N6O2, 371.2; found, 371.2. Step 2. Preparation of ethyl N-(3-((tert-butoxycarbonyl)amino)propyl)-N-(6-(1-methyl- 1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)glycinate
Figure imgf000193_0003
[0515] To a solution of tert-butyl (3-((6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)amino)propyl)carbamate (500 mg, 1.35 mmol, as prepared in the previous step) and ethyl 2-iodoacetate (346.6 mg, 1.62 mmol) in DMF (10 mL) was added K2CO3 (373.1 mg, 2.70 mmol) then the reaction was stirred at rt under nitrogen for 16 h. The mixture was concentrated under reduced pressure then the residue was dissolved in water (30 mL). The mixture was extracted with EtOAc (3 x 50 mL), then the combined organic extracts were washed with brine (10 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by reversed- phase flash chromatography with a C18 silica gel column eluting with 10% to 100% ACN / water (0.1% FA) to afford 300 mg (49%) of ethyl N-(3-((tert- butoxycarbonyl)amino)propyl)-N-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin- 3-yl)glycinate as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.77 (s, 1H), 8.20 (s, 1H), 7.95 (s, 1H), 7.76 (s, 1H), 7.57 (d, J = 9.3 Hz, 1H), 7.29 (dd, J = 9.3 ,1.2 Hz, 1H), 6.79 (t, J = 5.3 Hz, 1H), 4.04 (q, J = 7.1 Hz, 2H), 3.93 - 3.82 (m, 5H), 3.22 - 3.13 (m, 2H), 2.96 (q, J = 6.3 Hz, 2H), 1.52 (q, J = 6.3 Hz, 2H), 1.35 (s, 9H), 1.14 (t, J = 7.1 Hz, 3H) ;MS (ESI) m/z [M+H]+ calcd. for C23H32N6O4, 457.2; found, 457.3. Step 3. Preparation of ethyl N-(3-aminopropyl)-N-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)glycinate hydrochloride salt
Figure imgf000194_0001
[0516] To a solution of ethyl N-(3-((tert-butoxycarbonyl)amino)propyl)-N-(6-(1-methyl- 1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)glycinate (300 mg, 0.68 mmol, as prepared in the previous step) in DCM (10 mL) was added 4M HCl in dioxane (3 mL) then the mixture was stirred at rt under nitrogen for 2 h. The reaction was concentrated under reduced pressure to afford 200 mg (77%) of ethyl N-(3-aminopropyl)-N-(6-(1-methyl-1H-pyrazol- 4-yl)pyrazolo[1,5-a]pyridin-3-yl)glycinate hydrochloride salt as a white solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.77 (s, 1H), 8.19 (s, 1H), 7.95 (s, 1H), 7.77 (s, 1H), 7.63 - 7.54 (m, 1H), 7.29 (d, J = 9.3 Hz, 1H), 4.04 (q, J = 7.1 Hz, 2H), 3.94 - 3.88 (m, 2H), 3.86 (s, 3H), 3.26 - 3.18 (m, 2H), 2.98 (q, J = 5.8 Hz, 1H), 2.58 (t, J = 6.7 Hz, 1H), 1.58 - 1.43 (m, 2H), 1.14 (t, J = 7.1 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C19H25N2O2, 356.2 found, 356.2. Step 4. Preparation of 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)-1,4- diazepan-2-one
Figure imgf000195_0001
[0517] To a solution of ethyl N-(3-aminopropyl)-N-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)glycinate hydrochloride (200 mg, 0.55 mmol, as prepared in the previous step) in MeOH (5 mL ) was added 1,5,7-triazabicyclo[4.4.0]dec-5-ene (152.6 mg, 1.10 mmol) then the mixture was stirred at 90°C under nitrogen for 16 h. The reaction was concentrated under reduced pressure then the residue was dissolved in water (30 mL). The mixture was extracted with EtOAc (3 x 50 mL), then the combined organic extracts were washed with brine (10 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-TLC (MeOH / DCM 1:3) to afford 160 mg (84%) of 4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)-1,4-diazepan-2-one as a white solid. 1H NMR (400 MHz, CD3OD) δ (ppm) 8.56 (s, 1H), 8.03 (s, 1H), 7.87 (s, 1H), 7.80 (s, 1H), 7.66 (d, J = 9.3 Hz, 1H), 7.34 (dd, J = 9.3, 1.4 Hz, 1H), 3.95 (d, J = 5.5 Hz, 1H), 3.59 - 3.52 (m, 2H), 3.45 - 3.39 (m, 2H), 1.94 - 1.85 (m, 2H); MS (ESI) m/z [M+H]+ calcd. for C16H18N6O, 311.2; found, 311.3. Step 5. 1-(5-benzylpyrimidin-2-yl)-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)-1,4-diazepan-2-one (Compound 326)
Figure imgf000195_0002
[0518] To a solution of 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)-1,4- diazepan-2-one (150.0 mg, 0.48 mmol, as prepared in the previous step) and 5-benzyl-2- chloropyrimidine (Compound S88) (118.7 mg, 0.58 mmol) in dioxane (10 mL) were added Pd2(dba)3 (88.5 mg, 0.01 mmol), XantPhos (55.9 mg, 0.01 mmol), and K3PO4 (205.2 mg, 0.97 mmol), then the mixture was stirred at 90°C under nitrogen for 16 h. The reaction was concentrated under reduced pressure then the residue was purified by Prep-TLC (MeOH / DCM 1:10) and by Prep-HPLC to afford 42.3 mg (18%) of 1-(5-benzylpyrimidin-2-yl)-4- (6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)-1,4-diazepan-2-one (Compound 326) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.87(s, 1H), 8.70 (m, 3H), 8.21 (s, 1H), 7.90 (s, 1H), 7.65 (dd, J = 9.2, 1.0 Hz, 1H), 7.35 - 7.27 (m, 5H), 7.26 - 7.18 (m, 1H), 4.15 (d, J = 7.2 Hz, 4H), 4.00-3.80 (m, 5H), 3.52 (t, J = 5.4 Hz, 2H), 1.94 - 1.81 (m, 2H); MS (ESI) m/z [M+H]+ calcd. for C27H26N8O,479.2; found,479.1; LCMS purity: 254 nm: 99.0% Example 4: Synthesis of Exemplary Compound 327 (R)-1-phenylethyl (R)-2-methyl-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate (Compound 327)
Figure imgf000196_0001
Step 1. Preparation of tert-butyl (R)-2-methyl-4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate
Figure imgf000196_0002
[0519] To a solution of 3-bromo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound S9) (2.0 g, 7.22 mmol) dissolved in dioxane (30 mL) and tBuOH (30 mL) was added tert-butyl (R)-2-methylpiperazine-1-carboxylate (7.21 g, 36.08 mmol), tBuXPhos Pd G1 (0.99 g, 1.44 mmol), and KOtBu (1.62 g, 14.43 mmol), then the reaction was stirred at 90°C under nitrogen for 16 h. The reaction was cooled rt then diluted with water (200 mL). The mixture was extracted with EtOAc (3 x 200 mL) then the combined organic extracts were washed with brine (3 x 200 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with EtOAc / PE (3:2) to afford 1.2 g (42%) of tert-butyl (R)-2- methyl-4-[6-(1-methylpyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl]piperazine-1-carboxylate as a light blue solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.22 (s, 1H), 7.96 (s, 1H), 7.75 (s, 1H), 7.57 (d, J = 9.2 Hz, 1H), 7.33 (dd, J = 9.2, 1.6 Hz, 1H), 4.22 (dd, J = 7.6, 4.0 Hz, 1H), 3.87 (s, 3H), 3.84 – 3.76 (m, 1H), 3.22 – 3.19 (m, 1H), 3.14 – 3.07 (m, 1H), 2.77 (dd, J = 11.6, 3.6 Hz, 1H), 2.68 – 2.58 (m, 1H), 2.08 – 1.93 (m, 1H), 1.43 (s, 9H), 1.34 (d, J = 6.8 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C21H28N6O2, 397.2; found, 397.2. Step 2. Preparation of (R)-6-(1-methyl-1H-pyrazol-4-yl)-3-(3-methylpiperazin-1- yl)pyrazolo[1,5-a]pyridine
Figure imgf000197_0001
[0520] To a solution of tert-butyl (R)-2-methyl-4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (800 mg, 2.02 mmol, as prepared in the previous step) in DCM (10 mL) cooled to 0°C was added 4M HCl in dioxane (3 mL) then the reaction was stirred at 0°C under nitrogen for 1 h. The mixture was neutralized to pH 8 with sat. aqueous Na2CO3 then extracted with MeOH / DCM (1:10) (3 x 100 mL). The combined organic extracts were washed with brine (3 x 100 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure to afford 500 mg (83%) of (R)-6-(1-methyl-1H-pyrazol-4-yl)-3-(3-methylpiperazin-1-yl)pyrazolo[1,5- a]pyridine as a light yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.79 (s, 1H), 8.21 (s, 1H), 7.96 (d, J = 0.8 Hz, 1H), 7.70 (s, 1H), 7.60 (dd, J = 9.2, 1.0 Hz, 1H), 7.27 (dd, J = 9.2, 1.2 Hz, 1H), 3.86 (s, 3H), 3.21 – 3.11 (m, 2H), 2.91 (dd, J = 8.4, 2.8 Hz, 3H), 2.67 – 2.54 (m, 1H), 2.28 (d, J = 10.4 Hz, 1H), 1.23 (s, 1H), 1.00 (d, J = 6.4 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C16H20N6, 297.2; found, 297.2. Step 3. Preparation of (R)-1-phenylethyl (R)-2-methyl-4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 327)
Figure imgf000197_0002
[0521] To a solution of (R)-6-(1-methyl-1H-pyrazol-4-yl)-3-(3-methylpiperazin-1- yl)pyrazolo[1,5-a]pyridine (400 mg, 1.35 mmol, as prepared in the previous step) and (R)- 1-phenylethan-1-ol (494.6 mg, 4.05 mmol) in pyridine (15 mL) cooled to 0°C was added triphosgene (400.5 mg, 1.35 mmol) in portions over 1 min. The reaction was warmed to rt and stirred for 1 h, then diluted with water (100 mL) and extracted with EtOAc (6 x 100 mL). The combined organic extracts were washed with brine (6 x 100 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 158 mg (26%) of (R)-1-phenylethyl (R)- 2-methyl-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1- carboxylate (Compound 327) as an off-white solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.84 (s, 1H), 8.23 (s, 1H), 7.98 (d, J = 0.8 Hz, 1H), 7.76 (s, 1H), 7.59 (dd, J = 9.2, 1.0 Hz, 1H), 7.43 – 7.31 (m, 5H), 7.31 – 7.26 (m, 1H), 5.76 (q, J = 6.4 Hz, 1H), 4.33 (s, 1H), 3.92 (d, J = 13.2 Hz, 1H), 3.88 (s, 3H), 3.38 – 3.28 (m, 1H), 3.26 (d, J = 12.4 Hz, 1H), 3.18 – 3.09 (m, 1H), 2.81 (dd, J = 11.4, 4.0 Hz, 1H), 2.73 – 2.63 (m, 1H), 1.49 (d, J = 6.8 Hz, 3H), 1.38 (d, J = 6.0 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C25H28N6O2, 445.2; found, 445.1; LCMS purity: 254 nm: 98.5%; chiral purity: 100%. Example 5: Synthesis of Exemplary Compound 329 (R)-1-phenylethyl (S)-2-methyl-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate (Compound 329)
Figure imgf000198_0001
Step 1. Preparation of tert-butyl (S)-2-methyl-4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate
Figure imgf000198_0002
[0522] To a solution of 3-bromo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound S9) (2 g, 7.21 mmol) and tert-butyl (S)-2-methylpiperazine-1-carboxylate (7.23 g, 36.08 mmol) in dioxane (50 mL) and tBuOH (50 mL) were added KOtBu (1.62 g, 14.43 mmol) and tBuXPhos Pd G1 (0.99 g, 1.44 mmol) then the mixture was stirred at 90°C under nitrogen overnight. The reaction was cooled to rt and concentrated under reduced pressure. The mixture was diluted with water (100 mL) and extracted with DCM (3 x 100 mL). The combined organic extracts were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by reversed-phase flash chromatography with a C18 silica gel column eluting with 10% to 50% ACN / water (0.1% FA) to afford 370 mg (13%) of tert-butyl (S)-2-methyl-4-(6-(1- methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.84 (t, J = 1.3 Hz, 1H), 8.23 (s, 1H), 7.97 (d, J = 0.8 Hz, 1H), 7.76 (s, 1H), 7.59 – 7.57 (m, 1H), 7.35 – 7.33 (m, 1H), 4.22 (s, 1H), 3.87 (s, 3H), 3.85 – 3.77 (m, 1H), 3.25 – 3.20 (m, 2H),3.13 – 3.09 (m, 1H), 2.79 – 2.75 (m, 1H), 2.69 – 2.58 (m, 1H), 1.43 (s, 9H), 1.34 (d, J = 6.7 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C21H28N6O2, 397.2; found, 397.2. Step 2. Preparation of (S)-6-(1-methyl-1H-pyrazol-4-yl)-3-(3-methylpiperazin-1- yl)pyrazolo[1,5-a]pyridine hydrochloride salt
Figure imgf000199_0001
[0523] To a solution of (S)-2-methyl-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazine-1-carboxylate (360 mg, 0.90 mmol, as prepared in the previous step) in dioxane (10 mL) was added 4M HCl in dioxane (2.3 mL, 9.0 mmol) then the mixture was stirred at rt for 30 minutes. The reaction was concentrated under reduced pressure to afford 300 mg (78%) of (S)-6-(1-methyl-1H-pyrazol-4-yl)-3-(3- methylpiperazin-1-yl)pyrazolo[1,5-a]pyridine hydrochloride salt as a white solid. MS (ESI) m/z [M+H]+ calcd. for C16H20N6, 297.1; found, 297.1. Step 3. (R)-1-phenylethyl (S)-2-methyl-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazine-1-carboxylate (Compound 329)
Figure imgf000199_0002
[0524] To a solution of (S)-6-(1-methyl-1H-pyrazol-4-yl)-3-(3-methylpiperazin-1- yl)pyrazolo[1,5-a]pyridine hydrochloride salt (200 mg, 0.67 mmol, as prepared in the previous step) and (R)-1-phenylethan-1-ol (247.3 mg, 2.02 mmol) in pyridine (15 mL) was added triphosgene (200.2 mg, 0.67 mmol), then the mixture was stirred at rt under nitrogen overnight. The reaction was concentrated under reduced pressure then the residue was dissolved in water (50 mL) and the mixture was extracted with DCM (3 x 50 mL). The combined organic extracts were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 57.2 mg (19%) of (R)-1-phenylethyl (S)-2-methyl-4-[6-(1-methylpyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl]piperazine-1-carboxylate (Compound 329) as an off-white solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.84 (s,1H), 8.23 (s, 1H), 7.97 (s,1H), 7.76 (s, 1H), 7.59 (d, J = 9.2 Hz, 1H), 7.43 – 7.26 (m, 6H), 5.75 (q, J = 6.6 Hz, 1H), 4.34 (s, 1H), 3.92 (d, J = 12.9 Hz, 1H), 3.87 (s, 3H), 3.28 – 3.11 (m, 3H), 2.80 (d, J = 11.3 Hz, 1H), 2.72 – 2.62 (m, 1H), 1.49 (d, J = 6.6 Hz, 3H), 1.37 (d, J = 6.7 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C25H28N6O2, 445.1; found, 445.1.LCMS purity:254 nm: 99.9%; chiral purity: 99.6%. Example 6: Synthesis of Exemplary Compound 330 (R)-1-phenylethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrazin-3-yl)-1,4- diazepane-1-carboxylate (Compound 330)
Figure imgf000200_0001
Step 1. Preparation of tert-butyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrazin- 3-yl)-1,4-diazepane-1-carboxylate
Figure imgf000200_0002
[0525] To a solution of 3-bromo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrazine (Compound S5) (500 mg, 1.80 mmol) and tert-butyl 1,4-diazepane-1-carboxylate (1.80 g, 9.00 mmol) in tBuOH (6 mL) and dioxane (3 mL) was added KOtBu (302 mg, 2.70 mmol) and tBuXPhos Pd G1 (185 mg, 0.27 mmol) under nitrogen then the reaction was stirred at 60°C under nitrogen for 16 h. The mixture was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with EtOAc / PE (2:1) and reversed-phase flash chromatography with a C18 silica gel column eluting with 10% to 70% ACN / water (10mM NH4HCO3) to afford 150 mg (19%) of tert-butyl 4-(6-(1-methyl- 1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrazin-3-yl)-1,4-diazepane-1-carboxylate as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.06 (d, J = 4.4 Hz, 1H), 8.74 (d, J = 5.6 Hz, 1H), 8.16 (s, 1H), 7.97 (s, 1H), 7.65 (d, J = 9.2 Hz, 1H), 3.87 (s, 3H), 3.71 – 3.63 (m, 1H), 3.60 – 3.48 (m, 5H), 3.35 – 3.24 (m, 2H), 1.90 – 1.81 (m, 2H), 1.37 – 1.01 (m, 9H); MS (ESI) m/z [M+H]+ calcd. for C20H27N7O2, 398.2; found, 398.3. Step 2. Preparation of 3-(1,4-diazepan-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrazine hydrochloride salt
Figure imgf000201_0001
[0526] To a solution of tert-butyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin- 3-yl)-1,4-diazepane-1-carboxylate (150 mg, 0.38 mmol, as prepared in the previous step) in dioxane (5 mL) was added 4M HCl in dioxane (5 mL) then the reaction was stirred at rt for 1 h. The mixture was concentrated under reduced pressure to afford 170 mg (135%) of 3-(1,4-diazepan-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrazine hydrochloride salt as a red solid. MS (ESI) m/z [M+H]+ calcd. for C15H19N7, 298.2; found, 298.3. Step 3. Preparation of (R)-1-phenylethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrazin-3-yl)-1,4-diazepane-1-carboxylate (Compound 330).
Figure imgf000202_0001
[0527] To a solution of 3-(1,4-diazepan-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrazine hydrochloride salt (75 mg, 0.25 mmol, as prepared in the previous step), (R)-1- phenylethan-1-ol (37.0 mg, 0.30 mmol), and pyridine (79.8 mg, 1.01 mmol) in DCM (2 mL) cooled to 0°C was added triphosgene (22.5 mg, 0.08 mmol) in small portions then the reaction was stirred at 0°C for 1h. MeOH (2 mL) was added then the mixture was concentrated under reduced pressure. The residue was purified by Prep-HPLC to afford 16.3 mg (14%) of (R)-1-phenylethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrazin-3-yl)-1,4-diazepane-1-carboxylate (Compound 330) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.11 – 9.02 (m, 1H), 8.79 – 8.71 (m, 1H), 8.17 (s, 1H), 7.97 (s, 1H), 7.66 (d, J = 3.0 Hz, 1H), 7.34 – 7.15 (m, 4H), 7.15 – 7.06 (m, 1H), 5.66 – 5.47 (m, 1H), 3.88 (s, 3H), 3.83 – 3.69 (m, 1H), 3.69 – 3.34 (m, 7H), 2.03 – 1.70 (m, 2H), 1.41 – 1.15 (m, 3H); MS (ESI) m/z [M+H]+ calcd. for C24H27N7O2, 446.2; found, 446.1; LCMS purity: 254 nm: 99.0%. Example 7: Synthesis of Exemplary Compound 328 (R)-1-phenylethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)-1,4- diazepane-1-carboxylate (Compound 328)
Figure imgf000202_0002
Step 1. Preparation of tert-butyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)-1,4-diazepane-1-carboxylate
Figure imgf000203_0001
[0528] To a solution of 3-bromo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound S9) (500 mg, 1.80 mmol) and tert-butyl 1,4-diazepane-1-carboxylate (1.81 g, 9.02 mmol) in tBuOH (6 mL) and dioxane (3 mL) were added KOtBu (304 mg, 2.71 mmol) and tBuXPhos Pd G1 (186 mg, 0.27 mmol) under nitrogen, then the reaction was stirred at 60℃ under nitrogen for 16 h. The mixture was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with EtOAc / PE (1:1) and reversed-phase flash chromatography with a C18 silica gel column eluting with 10% to 70% ACN / water (10mM NH4HCO3) to afford 255 mg (32%) of tert-butyl 4-(6-(1-methyl- 1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)-1,4-diazepane-1-carboxylate as a yellow oil. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.73 (s, 1H), 8.19 (s, 1H), 7.94 (s, 1H), 7.65 (d, J = 9.2 Hz, 1H), 7.57 (d, J = 9.4 Hz, 1H), 7.24 – 7.14 (m, 1H), 3.86 (s, 3H), 3.55 – 3.49 (m, 2H), 3.46 – 3.39 (m, 1H), 3.39 – 3.32 (m, 4H), 3.30 (s, 1H), 1.91 – 1.79 (m, 2H), 1.46 – 1.13 (m, 9H); MS (ESI) m/z [M+H]+ calcd. for C21H28N6O2, 397.2; found, 397.3. Step 2. Preparation of 3-(1,4-diazepan-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridine hydrochloride salt
Figure imgf000203_0002
[0529] To a solution of tert-butyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin- 3-yl)-1,4-diazepane-1-carboxylate (245 mg, 0.62 mmol, as prepared in the previous step) in dioxane (6 mL) was added 4M HCl in dioxane (12 mL) then the reaction was stirred at rt for 1 h. The mixture was concentrated under reduced pressure to afford 290 mg (87%) of 3-(1,4-diazepan-1-yl)-6-(1-methyl-1H-pyrazol-4-yl) pyrazolo[1,5-a]pyridine hydrochloride salt as a light yellow solid. MS (ESI) m/z [M+H]+ calcd. for C16H20N6, 297.2; found, 297.3. Step 3. Preparation of (R)-1-phenylethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)-1,4-diazepane-1-carboxylate (Compound 328)
Figure imgf000204_0001
[0530] To a solution of 3-(1,4-diazepan-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridine hydrochloride salt (50 mg, 0.17 mmol, as prepared in the previous step), (R)-1- phenylethan-1-ol (25 mg, 0.20 mmol) and pyridine (20 mg, 0.25 mmol) in DCM (2 mL) cooled to 0°C was added triphosgene (15 mg, 0.05 mmol) in small portions then the reaction was stirred at 0°C for 1 h. The mixture was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 54 mg (72%) of (R)-1-phenylethyl 4-(6-(1- methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)-1,4-diazepane-1-carboxylate (Compound 328) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.76 – 8.71 (m, 1H), 8.20 (s, 1H), 7.95 (s, 1H), 7.66 (d, J = 1.2 Hz, 1H), 7.60 – 7.53 (m, 1H), 7.34 (d, J = 4.3 Hz, 2H), 7.31 – 7.18 (m, 4H), 5.75 – 5.59 (m, 1H), 3.87 (s, 3H), 3.75 – 3.49 (m, 3H), 3.49 – 3.41 (m, 2H), 3.41 – 3.33 (m, 2H), 3.31 – 3.22 (m, 1H), 2.01 – 1.73 (m, 2H), 1.50 – 1.27 (m, 3H); MS (ESI) m/z [M+H]+ calcd. for C25H28N6O2, 445.2; found, 445.1; LCMS purity: 254 nm: 99.4% Example 8: Synthesis of Exemplary Compound 331 3-(4-(5-benzylpyrimidin-2-yl)-1,4-diazepan-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrazine (Compound 331)
Figure imgf000204_0002
[0531] To a solution of 3-(1,4-diazepan-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrazine hydrochloride salt (50.0 mg, 0.15 mmol, as prepared in Example 6, Step 2) and DIEA (58.1 mg, 0.45 mmol) in IPA (2 mL) was added 5-benzyl-2-chloropyrimidine (Compound S88) (30.7 mg, 0.15 mmol) then the reaction was stirred at 100°C for 16 h. The mixture was cooled to rt and concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 26.1 mg (37%) of 3-(4-(5-benzylpyrimidin-2-yl)-1,4- diazepan-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrazine (Compound 331) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.04 (d, J = 1.5 Hz, 1H), 8.70 (d, J = 1.4 Hz, 1H), 8.19 (s, 2H), 8.16 (s, 1H), 7.96 (d, J = 0.8 Hz, 1H), 7.63 (s, 1H), 7.32 – 7.23 (m, 2H), 7.22 – 7.14 (m, 3H), 3.96 (t, J = 5.4 Hz, 2H), 3.87 (s, 3H), 3.73 (s, 2H), 3.67 (t, J = 6.0 Hz, 2H), 3.65 – 3.60 (m, 2H), 3.51 – 3.46 (m, 2H), 2.01 – 1.91 (m, 2H); MS (ESI) m/z [M+H]+ calcd. for C26H27N9, 466.2; found, 466.1; LCMS purity: 254 nm: 99.1% Example 9: Synthesis of Exemplary Compound 332 tert-butyl 8-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)-3,8- diazabicyclo[3.2.1]octane-3-carboxylate (Compound 332)
Figure imgf000205_0001
[0532] To a round bottom flask containing 3-bromo-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine (Compound S9) (200 mg, 0.72 mmol) and tert-butyl-3,8- diazabicyclo[3.2.1]octane-3-carboxylate (613 mg, 2.89 mmol) was added tBuOH (2 mL) and dioxane (1 mL). The solution was sparged with nitrogen for 20 min at rt then tBuXPhos Pd G1 (50 mg, 0.07 mmol) and KOtBu (121 mg, 1.08 mmol) were added and the reaction mixture was sparged with nitrogen for 5 min at rt. The reaction was heated to 60°C for 4 h, cooled to rt, and concentrated under reduced pressure. The residue was purified by silica gel chromatography eluting with 10-60% acetone / DCM to afford 166 mg (56%) of tert- butyl 8-[6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl]piperazine-3,8- diazabicyclo[3.2.1]octane-3-carboxylate (Compound 332).1H NMR (400 MHz, CDCl3) δ (ppm) 8.42 (s, 1H), 7.74 (s, 1H) 7.61 (s, 1H) 7.57 (s, 1H) 7.51 (d, J = 9.3 Hz, 1H) 7.27 (s, 1H) 7.10 (dd, J = 9.2, 1.5 Hz, 1H) 3.98 (s, 3H) 3.75-3.91 (m, 4H) 3.25-3.36 (m, 2H) 2.04 (d, J = 4 Hz, 2H) 1.81 (dd, J = 13, 7.4 Hz, 2H) 1.49 (s, 9H); MS (ESI) m/z [M+H]+ calcd. for C22H29N6O2, 409.2; found, 409.2; HPLC purity: 210 nm: 100%; 254 nm: 100%. Example 10: Synthesis of Exemplary Compound 333 3-(4-(isobutylsulfonyl)cyclohexyl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 333)
Figure imgf000206_0001
Step 1. Preparation of 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)cyclohex-3-en-1-ol
Figure imgf000206_0002
[0533] To a solution of 3-bromo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound S9) (1.20 g, 4.33 mmol) and 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)cyclohex-3-en-1-ol (0.97 g, 4.33 mmol) in dioxane (10 mL) and H2O (1 mL) was added XPhos Pd G3 (0.37 g, 0.43 mmol), XPhos (0.21 g, 0.43 mmol), and K3PO4 (1.84 g, 8.66 mmol) under nitrogen, then the reaction was stirred at 90°C under nitrogen for 16 h. The mixture was cooled to rt, then concentrated under reduced pressure. The residue was purified by silica gel chromatography eluting with EtOAc / PE (3:1) to afford 1.79 g (98%) of 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)cyclohex-3-en-1-ol as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.93 (s, 1H), 8.25 – 8.23 (m, 1H), 8.04 – 7.97 (m, 2H), 7.85 – 7.77 (m, 1H), 7.46 (dd, J = 9.3, 1.7 Hz, 1H), 6.02 – 5.92 (m, 1H), 4.67 (d, J = 4.1 Hz, 1H), 3.87 (s, 3H), 3.85 – 3.76 (m, 1H), 2.64 – 2.54 (m, 1H), 2.47 – 2.38 (m, 1H), 2.14 – 1.84 (m, 3H), 1.67 – 1.56 (m, 1H); MS (ESI) m/z [M+H]+ calcd. for C17H18N4O, 295.1; found, 295.2. Step 2. Preparation of 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)cyclohexan-1-ol
Figure imgf000206_0003
[0534] To a solution of 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)cyclohex-3-en-1-ol (1.5 g, 5.10 mmol, as prepared in the previous step) in MeOH (20 mL) was added Pd/C (0.54 g, 5.10 mmol) then the reaction was stirred at rt under an atmosphere of hydrogen for 40 h. The mixture was filtered, and the filter cake was washed with MeOH (2 x 5 mL). The filtrate was concentrated under reduced pressure to afford 760 mg (45%) of 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)cyclohexan-1- ol as a grey solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.92 – 8.83 (m, 1H), 8.22 (s, 1H), 7.97 (d, J = 0.8 Hz, 1H), 7.80 (d, J = 5.1 Hz, 1H), 7.73 – 7.61 (m, 1H), 7.42 – 7.33 (m, 1H), 4.38 (d, J = 3.3 Hz, 1H), 3.89 (s, 1H), 3.87 (d, J = 1.0 Hz, 3H), 2.91 – 2.62 (m, 1H), 2.01 – 1.44 (m, 8H); MS (ESI) m/z [M+H]+ calcd. for C17H20N4O, 297.2; found, 297.3. Step 3. Preparation of 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)cyclohexyl methanesulfonate
Figure imgf000207_0001
[0535] To a solution of 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)cyclohexan-1-ol (615 mg, 2.08 mmol, as prepared in the previous step) and TEA (420 mg, 4.15 mmol) in DCM (10 mL) was added Ms2O (542 mg, 3.11 mmol) at 0°C then the reaction was stirred at rt for 2 h. The mixture was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with MeOH / DCM (1:20) to afford 580 mg (678%) of 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)cyclohexyl methanesulfonate as a green solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.90 (s, 1H), 8.23 (s, 1H), 7.98 (s, 1H), 7.87 – 7.79 (m, 1H), 7.78 – 7.66 (m, 1H), 7.45 – 7.36 (m, 1H), 5.00 (s, 1H), 3.87 (s, 3H), 3.21 (s, 3H), 2.99 – 2.79 (m, 1H), 2.17 (s, 1H), 2.10 – 1.94 (m, 1H), 1.90 – 1.60 (m, 6H); MS (ESI) m/z [M+H]+ calcd. for C18H22N4O3S, 375.1; found, 375.2. Step 4. Preparation of S-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)cyclohexyl) ethanethioate
Figure imgf000207_0002
[0536] To a solution of 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)cyclohexyl methanesulfonate (500 mg, 1.34 mmol, as prepared in the previous step) in DMF (10 mL) was added potassium thioacetate (610 mg, 5.34 mmol) then the reaction was stirred at 90°C for 16 h. The mixture was cooled to rt, water (100 mL) was added, then the mixture was extracted with EtOAc (3 x 100 mL). The combined organic extracts were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-TLC (EtOAc) to afford 200 mg (42%) of S-(4-(6-(1- methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)cyclohexyl) ethanethioate as a yellow oil. MS (ESI) m/z [M+H]+ calcd. for C16H19N3, 254.2; found, 254.3. Step 5. Preparation of 3-(4-(isobutylthio)cyclohexyl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine
Figure imgf000208_0001
[0537] To a solution of S-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)cyclohexyl) ethanethioate (200 mg, 0.11 mmol, as prepared in the previous step) in DMSO (3 mL) and H2O (1 mL) was added KOH (13.3 mg, 0.24 mmol) then the reaction was stirred at rt for 1 h. To the mixture was added 1-bromo-2-methylpropane (15.5 mg, 0.11 mmol) and the reaction was stirred at rt for 1 h. H2O (20 mL) was added then the mixture was extracted with EtOAc (3 x 40 mL). The combined organic extracts were dried over anhydrous Na2SO4 and filtered then the filtrate was concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with a C18 silica gel column eluting with 10% to 90% ACN / water (10mM NH4HCO3) to afford 20 mg (10%) of 3-(4-(isobutylthio)cyclohexyl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridine as a colorless oil.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.91 – 8.85 (m, 1H), 8.22 (s, 1H), 7.96 (s, 1H), 7.83 – 7.77 (m, 1H), 7.74 – 7.63 (m, 1H), 7.43 – 7.34 (m, 1H), 3.87 (s, 3H), 3.20 – 3.10 (m, 1H), 2.46 – 2.39 (m, 2H), 2.11 – 2.03 (m, 1H), 1.99 – 1.81 (m, 4H), 1.79 – 1.68 (m, 2H), 1.64 – 1.37 (m, 3H), 1.03 – 0.89 (m, 6H); MS (ESI) m/z [M+H]+ calcd. for C21H28N4S, 369.2; found, 369.3. Step 6. Preparation of 3-(4-(isobutylsulfonyl)cyclohexyl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a] pyridine (Compound 333)
Figure imgf000209_0001
[0538] To a solution of 3-(4-(isobutylthio)cyclohexyl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine (15 mg, 0.04 mmol, as prepared in the previous step) in DCM (2 mL) cooled to 0°C was added mCPBA (35 mg, 0.21 mmol) then the reaction was stirred at 0°C for 1 h. The mixture was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 3.5 mg (21%) of 3-(4-(isobutylsulfonyl)cyclohexyl)-6-(1- methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a] pyridine (Compound 333) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.97 – 8.83 (m, 1H), 8.23 (s, 1H), 7.98 (d, J = 0.8 Hz, 1H), 7.88 – 7.78 (m, 1H), 7.79 – 7.66 (m, 1H), 7.47 – 7.36 (m, 1H), 3.87 (s, 3H), 3.29 – 3.10 (m, 2H), 3.05 – 2.97 (m, 2H), 2.31 – 2.10 (m, 3H), 2.09 – 1.87 (m, 3H), 1.86 – 1.76 (m, 1H), 1.71 – 1.57 (m, 2H), 1.10 – 1.03 (m, 6H); MS (ESI) m/z [M+H]+ calcd. for C21H28N4O2S, 401.2; found,401.2; LCMS purity: 254 nm: 99.9%. Example 11: Synthesis of Exemplary Compound 334 tert-butyl 6-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)-3,6- diazabicyclo[3.2.1]octane-3-carboxylate (Compound 334)
Figure imgf000209_0002
[0539] To a stirred solution of 3-bromo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridine (Compound S9) (1.20 g, 4.33 mmol) and tert-butyl 3,6- diazabicyclo[3.2.1]octane-3-carboxylate (1.84 g, 8.66 mmol) in tBuOH (20 mL) and dioxane (10 mL) were added KOtBu (728.8 mg, 6.50 mmol) and tBuXPhos Pd G1 (594.7 mg, 0.87 mmol) at rt then the reaction was stirred at 50°C under nitrogen for 48 h. The mixture was cooled to rt, concentrated under reduced pressure, and the residue was dissolved in water (100 mL). The resulting mixture was extracted with DCM (3 x 80 mL) then the combined organic extracts were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with EtOAc / PE (10:1) and by Prep-HPLC to afford 20.4 mg (1%) of tert-butyl 6-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)-3,6- diazabicyclo[3.2.1]octane-3-carboxylate (Compound 334) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.76 – 8.55 (m, 1H), 8.17 (s, 1H), 7.93 (s, 1H), 7.64 (d, J = 9.3 Hz, 1H), 7.51 – 7.43 (m, 1H), 7.10 – 7.01 (m, 1H), 4.23 – 3.70 (m, 6H), 3.64 – 3.48 (m, 1H), 3.23 – 3.00 (m, 1H), 2.91 – 2.63 (m, 2H), 2.49 – 2.43 (m, 1H), 2.13 – 1.92 (m, 1H), 1.87 – 1.73 (m, 1H), 1.30 (s, 3H), 0.92 (s, 6H); MS (ESI) m/z [M+H]+ calcd. for C22H28N6O2, 409.2; found, 409.15; LCMS purity: 254 nm: 98.9%. Example 12: Synthesis of Exemplary Compound 335 Mixture of 3-(4-(5-benzylpyrimidin-2-yl)-2,3,6,7-tetrahydro-1H-azepin-1-yl)-6-(1-methyl- 1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine and 3-(5-(5-benzylpyrimidin-2-yl)-2,3,4,7- tetrahydro-1H-azepin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 335)
Figure imgf000210_0001
Step 1. Preparation of tert-butyl 4-(5-benzylpyrimidin-2-yl)-2,3,6,7-tetrahydro-1H- azepine-1-carboxylate and tert-butyl 5-(5-benzylpyrimidin-2-yl)-2,3,4,7-tetrahydro-1H- azepine-1-carboxylate
Figure imgf000210_0002
[0540] To a solution of 5-benzyl-2-chloropyrimidine (Compound S88) (500 mg, 2.44 mmol) in dioxane (25 mL) and H2O (5 mL) was added a mixture of tert-butyl 5-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)-2,3,4,7-tetrahydro-1H-azepine-1-carboxylate and tert-butyl 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2,3,6,7-tetrahydro-1H-azepine- 1-carboxylate (789.7 mg, 2.44 mmol), Pd(dppf)Cl2 (268.1 mg, 0.37 mmol), and Cs2CO3 (1.59 g, 4.89 mmol) then the reaction was stirred at 90°C under nitrogen for 16 h. The mixture was cooled to rt and extracted with EtOAc (3 x 100 mL). The combined organic extracts were washed with brine (3x100 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with EtOAc/ PE (1:2) to afford 700 mg (78%) of a mixture of tert-butyl 4-(5-benzylpyrimidin-2-yl)-2,3,6,7-tetrahydro-1H-azepine-1-carboxylate and tert-butyl 5-(5-benzylpyrimidin-2-yl)-2,3,4,7-tetrahydro-1H-azepine-1-carboxylate as a yellow oil.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.67 (s, 2H), 7.35 – 7.24 (m, 4H), 7.24 – 7.16 (m, 2H), 4.12 – 4.04 (m, 2H), 3.96 (s, 2H), 3.56 – 3.48 (m, 2H), 2.89 – 2.81 (m, 2H), 1.79 (s, 2H), 1.38 (s, 5H), 1.36 (s, 4H); MS (ESI) m/z [M+H]+ calcd. for C22H27N3O2, 366.2; found, 366.2. Step 2. Preparation of 4-(5-benzylpyrimidin-2-yl)-2,3,6,7-tetrahydro-1H-azepine and 5-(5- benzylpyrimidin-2-yl)-2,3,4,7-tetrahydro-1H-azepine
Figure imgf000211_0001
[0541] To a solution of a mixture of tert-butyl 4-(5-benzylpyrimidin-2-yl)-2,3,6,7- tetrahydro-1H-azepine-1-carboxylate and tert-butyl 5-(5-benzylpyrimidin-2-yl)-2,3,4,7- tetrahydro-1H-azepine-1-carboxylate (700 mg, 1.92 mmol, as prepared in the previous step) in DCM (15 mL) cooled to 0°C was added TFA (5 mL) then the reaction was stirred at 0°C under nitrogen for 1 h. The mixture was neutralized to pH 8 with sat. aqueous Na2CO3 then extracted with MeOH / DCM (1:10) (3 x 100 mL). The combined organic extracts were washed with brine (3x100 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure to afford 460 mg (90%) of a mixture of 4-(5-benzylpyrimidin-2-yl)-2,3,6,7-tetrahydro-1H-azepine and 5-(5-benzylpyrimidin-2- yl)-2,3,4,7-tetrahydro-1H-azepine as a yellow oil.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.69 – 8.62 (m, 2H), 7.37 – 7.18 (m, 6H), 4.12 – 4.06 (m, 1H), 3.96 (s, 2H), 3.59 – 3.52 (m, 1H), 3.46 – 3.41 (m, 1H), 2.99 – 2.86 (m, 3H), 1.81 – 1.76 (m, 1H), 1.72 – 1.61 (m, 1H); MS (ESI) m/z [M+H]+ calcd. for C17H19N3, 266.2; found, 266.2. Step 3. Preparation of a mixture of 3-(4-(5-benzylpyrimidin-2-yl)-2,3,6,7-tetrahydro-1H- azepin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine and 3-(5-(5- benzylpyrimidin-2-yl)-2,3,4,7-tetrahydro-1H-azepin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine (Compound 335)
Figure imgf000212_0001
[0542] To a solution of a mixture of 4-(5-benzylpyrimidin-2-yl)-2,3,6,7-tetrahydro-1H- azepine and 5-(5-benzylpyrimidin-2-yl)-2,3,4,7-tetrahydro-1H-azepine (200 mg, 0.75 mmol, as prepared in the previous step) in dioxane (8 mL) was added 3-iodo-6-(1-methyl- 1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (293.2 mg, 0.91 mmol, as prepared for Compound S57, Step 1), Pd-PEPPSI-IPentCl 2-methylpyridine (95.1 mg, 0.11 mmol) and Cs2CO3 (491.1 mg, 1.51 mmol), then the reaction was stirred at 100°C under nitrogen for 16 h. The mixture was cooled to rt, diluted with water (30 mL), and extracted with EtOAc (3 x 30 mL). The combined organic extracts were washed with brine (3 x 30 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 13 mg (3.65%) of a mixture of 3-(4- (5-benzylpyrimidin-2-yl)-2,3,6,7-tetrahydro-1H-azepin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine and 3-(5-(5-benzylpyrimidin-2-yl)-2,3,4,7-tetrahydro-1H- azepin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 335) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.86 – 8.82 (m, 1H), 8.62 – 8.57 (m, 2H), 8.22 – 8.18 (m, 1H), 7.97 – 7.93 (m, 1H), 7.80 – 7.76 (s, 1H), 7.76 – 7.62 (m, 1H), 7.57 – 7.50 (m, 1H), 7.43 – 7.33 (m, 1H), 7.33 – 7.14 (m, 5H), 5.16 – 5.09 (m, 1H), 3.93 – 3.80 (m, 5H), 3.44 – 3.34 (m, 1H), 3.12 – 3.05 (m, 1H), 3.03 – 2.94 (m, 1H), 2.63 – 2.53 (m, 2H), 2.49 – 2.38 (m, 2H); MS (ESI) m/z [M+H]+ calcd. for C28H27N7, 462.2; found, 462.1; HPLC purity: 254 nm: 97.7% Example 13: Synthesis of Exemplary Compound 336 (5-methylfuran-3-yl)methyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate (Compound 336)
Figure imgf000213_0001
[0543] To a solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)pyrazolo[1,5- a]pyridine (Compound 12) (200 mg, 0.71 mmol) and (5-methylfuran-3-yl)methanol (95 mg, 0.85 mmol) in pyridine (5 mL) was added triphosgene (63.1 mg, 0.21 mmol) then the reaction was stirred at rt under nitrogen for 6 h. The mixture was concentrated under reduced pressure then the residue was dissolved in water (30 mL) and extracted with EtOAc (3 x 50 mL). The combined organic extracts were washed with brine (10 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 80.8 mg (27%) of (5-methylfuran-3- yl)methyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1- carboxylate (Compound 336) as an off-white solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (s, 1H), 8.23 (s, 1H), 7.98 (s, 1H), 7.77 (s, 1H), 7.65 (d, J = 9.2 Hz, 1H), 7.56 (s, 1H), 7.33 (dd, J = 9.2, 1.5 Hz, 1H), 6.15 (s, 1H), 4.90 (s, 2H), 3.87 (s, 3H), 3.57 (t, J = 5.0 Hz, 4H), 2.96 (t, J = 4.9 Hz, 4H), 2.25 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C22H24N6O3, 421.2; found, 421.1; LCMS purity: 254 nm: 99.7% Example 14: Synthesis of Exemplary Compound 337 (R)-1-phenylethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin-3-yl)-1,4- diazepane-1-carboxylate (Compound 337)
Figure imgf000213_0002
Step 1. Preparation of 3-(1,4-diazepan-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrimidine formic acid salt
Figure imgf000214_0001
[0544] To a solution of 3-bromo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidine (Compound S10) (2.00 g, 7.19 mmol) and 1,4-diazepane (3.60 g, 35.96 mmol) in DMF (120 mL) were added Pd-PEPPSI-IPentCl 2-methylpyridine (1.21 g, 1.44 mmol) and Cs2CO3 (4.69 g, 14.38 mmol) then the reaction was stirred at 90°C under nitrogen for 16 h. The mixture was cooled to rt then concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with a C18 silica gel column eluting with 10% to 100% ACN / water (0.1% FA) to afford 550 mg (22%) of 3-(1,4-diazepan-1-yl)-6- (1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidine formic acid salt as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.08 (d, J = 2.1 Hz, 1H), 8.52 (d, J = 2.1 Hz, 1H), 8.33-8.24 (m, 2H), 8.01 (s, 1H), 7.81 (s, 1H), 3.88 (s, 3H), 3.73 (t, J = 5.1 Hz, 2H), 3.60 (t, J = 6.0 Hz, 2H), 3.15 - 3.12 (m, 2H), 3.04 -2.98 (m, 2H), 2.04 - 1.93 (m, 2H); MS (ESI) m/z [M+H]+ calcd. for C15H19N, 298.2 found, 298.1. Step 2. (R)-1-phenylethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin-3- yl)-1,4-diazepane-1-carboxylate (Compound 337)
Figure imgf000214_0002
[0545] To a solution of 3-(1,4-diazepan-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrimidine formic acid salt (200 mg, 0.61 mmol, as prepared in the previous step) and (R)-1-phenylethanol (89 mg, 0.73 mmol) in pyridine (5 mL) was added triphosgene (54.1 mg, 0.18 mmol) then the reaction was stirred at rt under nitrogen for 6 h. The mixture was concentrated under reduced pressure then the residue was dissolved in water (30 mL) and extracted with EtOAc (3 x 50 mL). The combined organic extracts were washed with brine (10 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 47.7 mg (18%) of (R)-1-phenylethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin-3-yl)-1,4- diazepane-1-carboxylate (Compound 337) as a red solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.06 (dd, J = 6.1, 2.1 Hz, 1H), 8.51 (dd, J = 6.5, 2.2 Hz, 1H), 8.25 (s, 1H), 8.01 (d, J = 0.8 Hz, 1H), 7.79 (d, J = 1.7 Hz, 1H), 7.36 – 7.09 (m, 5H), 5.70 - 5.48 (m, 1H), 3.89 (s, 3H), 3.85 - 3.48 (m, 6H), 3.59-3.34 (m, 2H), 2.03 - 1.68 (m, 2H), 1.44 - 1.25 (m, 3H); MS (ESI) m/z [M+H]+ calcd. for C24H27N7O2, 446.2,found, 446.2; LCMS purity: 254 nm: 99.4% Example 15: Synthesis of Exemplary Compound 338 3-(4-(5-benzylpyrimidin-2-yl)-1,4-diazepan-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrimidine (Compound 338)
Figure imgf000215_0001
[0546] To a solution of 3-(1,4-diazepan-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrimidine formate (200 mg, 0.58 mmol, as prepared in Example 12, Step 1) and 5- benzyl-2-chloropyrimidine (Compound S88) (143 mg, 0.70 mmol) in IPA (3 mL) was added DIEA (225.8 mg, 1.72 mmol) then the reaction was stirred at 90°C under nitrogen for 16 h. The mixture was cooled to rt and concentrated under reduced pressure, then the residue was purified by Prep-HPLC to afford 15.6 mg (6%) of 3-(4-(5-benzylpyrimidin-2- yl)-1,4-diazepan-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidine (Compound 338) as a red solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.01 (s, 1H), 8.50 (s, 1H), 8.27-8.18 (m, 3H), 8.00 (s, 1H), 7.77 (s, 1H), 7.32-7.24 (m, 2H), 7.23-7.14 (m, 3H), 3.96 - 3.86 (m, 5H), 3.78 – 3.62 (m, 6H), 3.53 (t, J = 5.9 Hz, 2H), 1.97 (t, J = 5.9 Hz, 2H); MS (ESI) m/z [M+H]+ calcd. for C26H27N9, 466.2; found, 466.2; LCMS purity: 254 nm: 99.2% Example 16: Synthesis of Exemplary Compound 339 tert-butyl 3-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)-3,8- diazabicyclo[3.2.1]octane-8-carboxylate (Compound 339)
Figure imgf000216_0001
[0547] To a glass vial containing 3-iodo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridine (220 mg, 0.68 mmol, as prepared for Compound S57, Step 1), tert-butyl-3,8- diazabicyclo[3.2.1]octane-8 carboxylate (433 mg, 2.04 mmol), CuI (52 mg, 0.27 mmol), and K3PO4 (288 mg, 1.36 mmol) under nitrogen was added 1-butanol (4 mL) and 1,2- ethanediol (0.66 mL), then the reaction was heated to 100°C for 44 h. The mixture was cooled to rt, diluted with DCM and washed with water and brine. The organic layer was dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with acetone / DCM to afford 154 mg (56%) of tert-butyl 3-[6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl]-3,8-diazabicyclo[3.2.1]octane-8-carboxylate (Compound 339). 1H NMR (400 MHz, CDCl3) δ (ppm) 8.45 (br. s., 1H) 7.74 (s, 1H) 7.70 (s, 1H) 7.67 (br. s., 1H) 7.61 (s, 1H) 7.47 (d, J = 8.7 Hz, 1H) 7.11 (d, J = 8.6 Hz, 1H) 4.34 (br. s., 4H) 3.97 (s, 3H) 3.09 (br. s., 4H) 2.09 (m, 2H) 1.99 (m, 2H) 1.51 (s, 9H); MS (ESI) m/z [M+H]+ calcd. for C22H29N6O2, 409.2; found, 409.2; HPLC purity: 210 nm: 98.8%; 254 nm: 97.6%. Example 17: Synthesis of Exemplary Compound 340 3-(3-(5-benzylpyrimidin-2-yl)-3,6-diazabicyclo[3.2.1]octan-6-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine (Compound 340)
Figure imgf000216_0002
Step 1. Preparation of 3-(3,6-diazabicyclo[3.2.1]octan-6-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine hydrochloride salt
Figure imgf000217_0001
[0548] To a solution of tert-butyl 6-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin- 3-yl)-3,6-diazabicyclo[3.2.1]octane-3-carboxylate (Compound 334) (40 mg, 0.10 mmol) in DCM (0.5 mL) cooled to 0°C was added 4M HCl in dioxane (2.6 mL) then the reaction was warmed to rt and stirred for 1 h. The mixture was concentrated under reduced pressure to afford 30 mg (89%) of 3-(3,6-diazabicyclo[3.2.1]octan-6-yl)-6-(1-methyl-1H-pyrazol- 4-yl)pyrazolo[1,5-a]pyridine hydrochloride salt as a yellow solid. MS (ESI) m/z [M+H]+ calcd. for C9H8N2O2, 309.2; found, 309.3. Step 2. Preparation of 3-(3-(5-benzylpyrimidin-2-yl)-3,6-diazabicyclo[3.2.1]octan-6-yl)-6- (1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 340)
Figure imgf000217_0002
[0549] To a solution of 3-(3,6-diazabicyclo[3.2.1]octan-6-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine hydrochloride salt (28 mg, 0.08 mmol, as prepared in the previous step) and 5-benzyl-2-chloropyrimidine (Compound S88) (16.6 mg, 0.08 mmol) in IPA (2 mL) was added DIEA (31.5 mg, 0.24 mmol) then the reaction was stirred at 100°C for 24 h. The resulting mixture was cooled to rt and concentrated under reduced pressure. The residue was purified by Prep-HPLC to afford 16.7 mg (43%) of 3-(3-(5- benzylpyrimidin-2-yl)-3,6-diazabicyclo[3.2.1]octan-6-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine (Compound 340) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.62 (s, 1H), 8.17 (s, 1H), 8.14 (s, 2H), 7.92 (d, J = 0.4 Hz, 1H), 7.66 (d, J = 9.4 Hz, 1H), 7.46 (s, 1H), 7.30 – 7.20 (m, 2H), 7.19 – 7.12 (m, 3H), 7.06 (dd, J = 9.3, 1.6 Hz, 1H), 4.40 (d, J = 12.6 Hz, 1H), 4.29 (d, J = 12.4 Hz, 1H), 4.19 – 4.11 (m, 1H), 3.87 (s, 3H), 3.70 (s, 2H), 3.63 – 3.56 (m, 1H), 3.17 (d, J = 8.5 Hz, 1H), 3.08 (d, J = 12.0 Hz, 1H), 2.85 (d, J = 12.5 Hz, 1H), 2.72 – 2.60 (m, 1H), 2.13 – 2.02 (m, 1H), 1.88 (d, J = 10.7 Hz, 1H); MS (ESI) m/z [M+H]+ calcd. for C28H28N8, 477.2; found, 477.50; HPLC purity: 254 nm: 95.5%. Example 18: Synthesis of Exemplary Compound 341 4-(5-benzylpyrimidin-2-yl)-1-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)-1,4- diazepan-5-one (Compound 341)
Figure imgf000218_0001
Step 1. Preparation of ethyl 3-((6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)amino)propanoate
Figure imgf000218_0002
[0550] To a solution of 6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-amine (Compound S56) (800 mg, 3.75 mmol) and ethyl 3-iodopropanoate (1.28 g, 5.62 mmol) in DMF (50 mL) was added K2CO3 (1.04 g, 7.50 mmol) then the reaction was stirred at 90°C under nitrogen overnight. The mixture was cooled to rt then purified by reversed-phase flash chromatography with a C18 silica gel column eluting with 10% to 60% ACN / water (0.1%FA) to afford 370 mg (31%) of ethyl 3-((6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)amino)propanoate as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.68 (s,1H), 8.17 (s,1H), 7.93 (s,1H), 7.62 – 7.52 (m, 2H), 7.16 (d, J = 9.2Hz, 1H), 4.75 (t, J = 6.6 Hz, 1H), 4.06 (q, J = 7.1 Hz, 2H), 3.86 (s, 3H), 3.30 (t, J = 6.6 Hz, 2H), 2.56 (t, J = 6.7 Hz, 2H), 1.18 (t, J = 7.1 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C16H19N5O2, 314.1; found, 314.1. Step 2. Preparation of a mixture of ethyl 3-((2-((tert-butoxycarbonyl)amino)ethyl)(6-(1- methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)amino)propanoate and methyl 3-((2- ((tert-butoxycarbonyl)amino)ethyl)(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin- 3-yl)amino)propanoate
Figure imgf000218_0003
[0551] To a solution of ethyl 3-{[6-(1-methylpyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl]amino}propanoate (320 mg, 1.02 mmol, as prepared in the previous step) and tert-butyl N-(2-oxoethyl)carbamate (812.7 mg, 5.10 mmol) in MeOH (10 mL) was added NaHCO3 (257.3 mg, 3.06 mmol) then the reaction was stirred at 60°C under nitrogen for 72 h. The mixture was cooled to rt and concentrated under reduced pressure. The residue was dissolved in THF (10mL), the solution was cooled to 0°C then borane THF (877.6 mg, 10.21 mmol) was added dropwise over 1 minute. The resulting mixture was warmed to rt and stirred overnight. The reaction was concentrated under reduced pressure, then the residue was dissolved in water (50 mL) and extracted with DCM (3 x 50mL). The combined organic extracts were dried over anhydrous Na2SO4and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 40 mg (9%) of ethyl 3-((2-((tert-butoxycarbonyl)amino)ethyl)(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)amino)propanoate as a white solid and 110 mg (24%) of methyl 3-((2-((tert-butoxycarbonyl)amino)ethyl)(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)amino)propanoate as a white solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.81 (t, J = 1.2 Hz, 1H), 8.21 (s, 1H), 7.96 (d, J = 0.8 Hz, 1H), 7.84 (s, 1H), 7.55 (d, J = 9.3 Hz, 1H), 7.34 – 7.31 (m, 1H), 6.71 – 6.70 (m, 1H), 3.87 (s, 3H), 3.54 (s, 3H), 3.17 (d, J = 5.3 Hz, 2H), 3.08 – 2.87 (m, 4H), 2.40 (t, J = 6.8 Hz, 2H), 1.36 (s, 9H); MS (ESI) m/z [M+H]+ calcd. for C23H32N6O4, 457.2; found, 457.2 and MS (ESI) m/z [M+H]+ calcd. for C22H30N6O4, 443.2; found, 443.2 Step 3. Preparation of a mixture of ethyl 3-((2-aminoethyl)(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)amino)propanoate hydrochloride salt and methyl 3-((2- aminoethyl)(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)amino)propanoate hydrochloride salt
Figure imgf000219_0001
[0552] To a solution of a mixture of ethyl 3-((2-((tert-butoxycarbonyl)amino)ethyl)(6-(1- methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)amino)propanoate and methyl 3-((2- ((tert-butoxycarbonyl)amino)ethyl)(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin- 3-yl)amino)propanoate (150 mg, 0.33, as prepared in the previous step) in DCM (5 mL) was added 4M HCl in dioxane (119.7 mg, 3.30 mmol) then the reaction was stirred at rt under nitrogen for 1 h. The mixture was concentrated under reduced pressure to afford 150 mg of a mixture of ethyl 3-((2-aminoethyl)(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)amino)propanoate hydrochloride salt and methyl 3-((2-aminoethyl)(6-(1- methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)amino)propanoate hydrochloride salt as a white solid. MS (ESI) m/z [M+H]+ calcd. for C18H24N6O2, 357.2; found, 357.2 and MS (ESI) m/z [M+H]+ calcd. for C17H22N6O2, 343.2; found, 343.2. Step 4. Preparation of 1-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)-1,4- diazepan-5-one
Figure imgf000220_0001
[0553] To a solution of the mixture of ethyl 3-((2-aminoethyl)(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)amino)propanoate hydrochloride salt and methyl 3-((2- aminoethyl)(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)amino)propanoate hydrochloride salt (150 mg, 0.43 mmol, as prepared in the previous step) in MeOH (5 mL) was added 1,5,7-triazabicyclo[4.4.0]dec-5-ene (121.9 mg, 0.87 mmol) then the recation was stirred at 80°C under nitrogen overnight. The mixture was cooled to rt, then concentrated under reduced pressure. The residue was dissolved in water (20 mL) then extracted with DCM (3 x 20 mL). The combined organic extracts were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-TLC (MeOH / DCM 1:1) to afford 100 mg (74%) of 1-(6- (1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)-1,4-diazepan-5-one as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.81 (s, 1H), 8.22 (s, 1H), 7.96 (s, 1H), 7.79 (s, 1H), 7.68 – 7.65 (m, 1H), 7.59 (dd, J = 9.2, 1.0 Hz, 1H), 7.32 (dd, J = 9.3, 1.5 Hz, 1H), 5.76 (s, 1H), 3.86 (s, 3H), 3.18 – 3.09 (m, 4H), 2.64 – 2.57 (m, 2H), 1.23 (s, 1H) ;MS (ESI) m/z [M+H]+ calcd. for C16H18N6O, 311.1; found, 311.1. Step 5. 4-(5-benzylpyrimidin-2-yl)-1-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)-1,4-diazepan-5-one (Compound 341)
Figure imgf000221_0001
[0554] To a solution of 1-[6-(1-methylpyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl]-1,4- diazepan-5-one (100 mg, 0.32 mmol, as prepared in the previous step) and 5-benzyl-2- chloropyrimidine (Compound S88) (79.1 mg, 0.38 mmol) in dioxane (10 mL) were added Pd2(dba)3 (44.2 mg, 0.05 mmol), XantPhos (27.9 mg, 0.05 mmol), and K3PO4 (136.7 mg, 0.64 mmol) then the reaction was stirred at 90°C under nitrogen overnight. The mixture was cooled to rt and concentrated under reduced pressure. The residue was dissolved in water (20 mL) then extracted with EtOAc (3 x 20 mL). The combined organic extracts were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 52.7 mg (34%) of 4-(5- benzylpyrimidin-2-yl)-1-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)-1,4- diazepan-5-one (Compound 341) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (s, 1H), 8.72 (s, 2H), 8.23 (s, 1H), 7.97 (s, 1H), 7.84 (s, 1H), 7.63 (d, J = 9.3 Hz, 1H), 7.42 – 7.28 (m, 5H), 7.25 – 7.21 (m, 1H), 4.18 – 4.16 (m, 2H), 4.00 (s, 2H), 3.87 (s, 3H), 3.29 – 3.25 (m, 4H), 3.00 – 2.92 (m, 2H); MS (ESI) m/z [M+H]+ calcd. for C27H28N8O, 479.2; found, 479.2; LCMS purity: 254 nm: 99.7%. Example 19: Synthesis of Exemplary Compound 342 (R)-1-(4-(trifluoromethoxy)phenyl)ethyl 4-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2- b]pyridazin-3-yl)piperazine-1-carboxylate (Compound 342)
Figure imgf000221_0002
Step 1. Preparation of 4-nitrophenyl (1R)-1-[4-(trifluoromethoxy)phenyl]ethyl carbonate
Figure imgf000222_0001
[0555] To a solution of (R)-1-[4-(trifluoromethoxy)phenyl]ethanol (300 mg, 1.46 mmol) in DCM (15 mL) cooled to 0 °C was added pyridine (0.50 mL, 6.2 mmol) then the mixture was stirred for 10 minutes, then 4-nitrophenyl chloroformate (1.21 g, 2.94 mmol) was added in one portion. The reaction was warmed to rt and stirred overnight. The mixture was filtered, then the filtrate was concentrated under reduced pressure. The residue was purified by silica gel chromatography eluting with 0-80% DCM / hexanes to afford 0.54 g (94%) of 4-nitrophenyl (1R)-1-[4-(trifluoromethoxy)phenyl]ethyl carbonate as a white solid. 1H NMR (400 MHz, CDCl3) δ (ppm) 8.27 (d, J=9.17 Hz, 2H) 7.47 (d, J=8.56 Hz, 2H) 7.37 (d, J=9.17 Hz, 2H) 7.20 - 7.30 (m, 2H) 5.85 (q, J=6.60 Hz, 1H) 1.71 (d, J=6.72 Hz, 3H). Step 2. Preparation of (R)-1-[4-(trifluoromethoxy)phenyl]ethyl 4-[7-(1-methyl-1H- pyrazol-4-yl)imidazo[1,2-b]pyridazin-3-yl]piperazine-1-carboxylate (Compound 342)
Figure imgf000222_0002
[0556] To a solution of 4-nitrophenyl (R)-1-[4-(trifluoromethoxy)phenyl]ethyl carbonate (144 mg, 0.39 mmol, as prepared in the previous step) in ACN (3.1 mL) was added 7-(1- methyl-1H-pyrazol-4-yl)-3-piperazin-1-ylimidazo[1,2-b]pyridazine trihydrochloride salt (84 mg, 0.21 mmol, as prepared in Example 64, Step 2) followed by DIEA (1.1 mL, 6.4 mmol) then the reaction was stirred at rt under nitrogen for 22 h. The mixture was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with 0-100% (10% IPA in DCM)/ DCM, then the combined fractions were concentrated under reduced pressure. The residue was suspended in 25% ether in hexanes then concentrated under reduced pressure. This was repeated four times to afford 78 mg (70%) of (R)-1-[4-(trifluoromethoxy)phenyl]ethyl 4-[7-(1-methyl-1H- pyrazol-4-yl)imidazo[1,2-b]pyridazin-3-yl]piperazine-1-carboxylate (Compound 342) as a yellow solid.1H NMR (400 MHz, CD3OD) δ (ppm) 8.74 (d, J=2.08 Hz, 1H) 8.21 (s, 1H) 8.02 (s, 1H) 7.97 (d, J=1.96 Hz, 1H) 7.49 (d, J=8.68 Hz, 2H) 7.28 (d, J=7.95 Hz, 3H) 5.83 (q, J=6.56 Hz, 1H) 3.97 (s, 3H) 3.60 - 3.90 (m, 4H) 3.27 (br. s., 4H) 1.57 (d, J=6.60 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C24H25F3N7O3, 516.2; found, 516.2; HPLC purity: 210 nm: 97.0%; 254 nm: 97.0%. Example 20: Synthesis of Exemplary Compound 343 (R)-1-(5-methylpyridin-2-yl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate (Compound 343)
Figure imgf000223_0001
Step 1. Preparation of 4-nitrophenyl 4-[6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl]piperazine- 1-carboxylate
Figure imgf000223_0002
[0557] Pyridine (0.229 mL, 2.83 mmol) was added to a solution of 6-(1-methyl-1H- pyrazol-4-yl)-3-piperazin-1-ylpyrazolo[1,5-a]pyridine (Compound 12) (0.400 g, 1.42 mmol) and 4-nitrophenyl chloroformate (0.314 g, 1.56 mmol) in ACN (10 mL) at rt. The solvent was removed under reduced pressure, then MTBE (50 mL) was added to the residue and the mixture was stirred at rt overnight. The solid was collected by filtration and air dried to give 600 mg (100%) of 4-nitrophenyl 4-[6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl]piperazine-1-carboxylate as a yellow solid. 1H NMR (400 MHz, CDCl3) δ (ppm) 8.50 (s, 1H), 8.23 - 8.37 (m, 2H), 7.72 - 7.82 (m, 2H), 7.64 (s, 1H), 7.48 - 7.58 (m, 1H), 7.33 - 7.41 (m, 2H), 7.20 (dd, J=9.17, 1.47 Hz, 1H), 4.00 (s, 3H) 3.91 (br. s., 2H), 3.82 (br. s., 2H), 3.08 - 3.21 (m, 4H); MS (ESI) m/z [M+H]+ calcd. for C22H22N7O4: 448.4; found: 448.1; HPLC purity: 210 nm: 90.4%; 254 nm: 89.3% Step 2. Preparation of (R)-1-(5-methylpyridin-2-yl)ethanol
Figure imgf000223_0003
[0558] To a solution of (R,R)-Ts-DENEB (0.0480 g, 0.0740 mmol) in toluene (5 mL) was added 1-(5-methylpyridin-2-yl)ethanone (1.00 g, 7.40 mmol) under nitrogen at rt. TEA (2.58 mL, 18.5 mmol) was added followed by formic acid (0.698 mL, 18.5 mmol) then the reaction was stirred at rt overnight. The mixture was partitioned between sat. aqueous NaHCO3 (20 mL) and EtOAc (20 mL) then the aqueous phase was extracted with EtOAc (2 x 20 mL). The combined organic extracts were washed with sat. aqueous NaHCO3 (20 mL), then the combined aqueous layers were extracted with EtOAc (20 mL). The combined organic extracts were washed with brine (20 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with 0-80% acetone / DCM to afford 700 mg (69%) of (R)-1-(5-methylpyridin-2-yl)ethanol as a light brown oil.1H NMR (300 MHz, CDCl3) δ (ppm) 8.36 (s, 1H), 7.50 (ddt, J=7.96, 1.40, 0.63, 0.63 Hz, 1H), 7.18 (d, J=7.92 Hz, 1H), 4.86 (q, J=6.52 Hz, 1H), 4.29 (br. s., 1H), 2.34 (s, 3H), 1.49 (d, J=6.61 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C8H12NO: 138.2; found: 138.0. Step 3. Preparation of (R)-1-(5-methylpyridin-2-yl)ethyl 4-[6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1, 5-a]pyridin-3-yl]piperazine-1-carboxylate (Compound 343)
Figure imgf000224_0001
[0559] To a solution of (R)-1-(5-methylpyridin-2-yl)ethanol (0.100 g, 0.729 mmol, as prepared in the previous step) in DMF (6 mL) cooled to 0°C under nitrogen was added NaH (267.3 mg, 0.67 mmol) then the reaction was stirred at 0°C for 20 minutes.4-Nitrophenyl 4-[6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl]piperazine-1-carboxylate (0.309 g, 0.607 mmol, , as prepared in Step 1) was added then the reaction was stirred at 0°C for 30 minutes, then warmed to rt and stirred overnight. The mixture was diluted with sat. aqueous NaHCO3 (50 mL) and EtOAc (50 mL). The layers were separated and the EtOAc layer was washed with water (3 x 20 mL) and sat. aqueous NaHCO3 (20 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with 5% MeOH / DCM to afford 40 mg (15%) of (R)-1-(5-methylpyridin-2-yl)ethyl 4-[6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl]piperazine-1-carboxylate (Compound 343) as an orange solid.1H NMR (300 MHz, CDCl3) δ (ppm) 8.37 - 8.56 (m, 2H), 7.71 (s, 1H), 7.75 (s, 1H), 7.61 (s, 1H), 7.50 (d, J=9.22 Hz, 2H), 7.20 - 7.34 (m, 1H), 7.15 (dd, J=9.17, 1.26 Hz, 1H), 5.87 (q, J=6.67 Hz, 1H), 3.98 (s, 3H), 3.74 (br. s., 4H), 2.96 - 3.14 (m, 4H), 2.34 (s, 3H), 1.63 (d, J=6.61 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C24H28N7O2: 446.5; found: 446.2; HPLC purity: 210 nm: 100.0%; 254 nm: 100.0%. Example 21: Synthesis of Exemplary Compound 345 tert-butyl 4-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-a]pyridin-3-yl)piperazine-1- carboxylate (Compound 345)
Figure imgf000225_0001
[0560] To a glass vial containing 3-bromo-7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2- a]pyridine (Compound S7) (0.250 g, 0.90 mmol) and tert-butyl-1-piperazinecarboxylate (0.672 g, 3.61 mmol) was added NaOtBu (0.152 g, 1.35 mmol) and tBuXPhos Pd G1 (0.062 g, 0.09 mmol) and the headspace was flushed with nitrogen. To another glass vial was added tBuOH (2 mL) and dioxane (2 mL) and the mixture was sparged with nitrogen for 15 minutes at rt. The solvent mixture was added to the vial containing the solids then the reaction was heated to 50°C for 2 h, cooled to rt, and concentrated under reduced pressure. The residue was treated with ACN then concentrated under reduced pressure. The residue was purified by silica gel chromatography eluting with 20% MeOH (0.1%TEA) / DCM to afford 312 mg (90%) of tert-butyl 4-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-a]pyridin- 3-yl)piperazine-1-carboxylate (Compound 345) as an amber oil. 1H NMR (400 MHz, CDCl3) δ (ppm) 7.97 (d, J= 7.1 Hz, 1H), 7.82 (s, 1H), 7.73 (s, 1H), 7.64 (s, 1H), 7.26 (s, 1H), 3.98 (s, 3H), 3.98 (s, 3H), 3.65 (m, 4H), 3.01 (m, 4H), 1.50 (s, 9H); MS (ESI) m/z [M+H]+ calcd. for C20H27N6O2, 383.2; found, 383.3; HPLC purity: 210 nm: 86.8%; 254 nm: 87.4%. Example 22: Synthesis of Exemplary Compound 344 (R)-1-(4-chlorophenyl)ethyl 4-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-a]pyridin-3- yl)piperazine-1-carboxylate (Compound 344)
Figure imgf000226_0001
Step 1. Preparation of 7-(1-methyl-1H-pyrazol-4-yl)-3-piperazin-1-ylimidazo[1,2- a]pyridine trihydrochloride salt
Figure imgf000226_0002
[0561] To solution of tert-butyl 4-[7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-a]pyridin-3- yl]piperazine-1-carboxylate (Compound 345) (0.300 g, 0.78 mmol) in EtOAc (5 mL) was heated to 50 °C then conc. HCl (0.32 mL, 3.92 mmol) was slowly added. After 1 h MTBE (9 mL) was added with vigorous stirring at 50 °C for 30 minutes, then the mixture was cooled to rt, filtered, and the filter cake was washed with MTBE. The solid was dried in a vacuum oven to afford 296 mg (97%) of 7-(1-methyl-1H-pyrazol-4-yl)-3-piperazin-1- ylimidazo[1,2-a]pyridine trihydrochloride salt as a tan solid.1H NMR (400 MHz, DMSO- d6) δ (ppm) 8.73 (s, 1H), 8.64 (s, 1H), 8.25 (s, 1H), 8.03 (s, 1H), 7.96 (s, 1H), 7.73 (dd, J = 7.2, 1.5 Hz, 1H), 3.92(s, 3H), 3.31 (bs, 8H); MS (ESI+, m/z): Calcd. for C15H19N6: 283.2. Found: 283.1; HPLC purity: 210 nm: 66%; 254 nm: 74%. Step 2. Preparation of (R)-1-(4-chlorophenyl)ethyl 4-[7-(1-methyl-1H-pyrazol-4- yl)imidazo[1,2-a]pyridin-3-yl]piperazine-1-carboxylate (Compound 344)
Figure imgf000226_0003
[0562] To a solution of (R)-1-(4-chlorophenyl)ethyl (2,5-dioxopyrrolidin-1-yl) carbonate (Compound S95) (297 mg, 1.00 mmol) in ACN (9 mL) was added 7-(1-methyl-1H-pyrazol- 4-yl)-3-piperazin-1-ylimidazo[1,2-a]pyridine trihydrochloride salt (290 mg, 0.740 mmol, as prepared in the previous step) and DIEA (3 mL, 18.0 mmol), then the reaction was stirred for 90 minutes. The mixture was concentrated under reduced pressure then the residue was partitioned between water (adjusted to pH 9) and EtOAc. The organic layer was washed with brine, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with MeOH (0.1% TEA) / DCM to afford 185 mg (54%) of (R)-1-(4-chlorophenyl)ethyl 4-[7- (1-methyl-1H-pyrazol-4-yl)imidazo[1,2-a]pyridin-3-yl]piperazine-1-carboxylate (Compound 344) as a foam solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.30 (s, 1H) 8.16 (d, J=7 Hz, 1H) 8.02 (s, 1H) 7.70 (s, 1H) 7.44 (m, 4H), 7.24 (s, 1H) 7.15 (dd, J=7.2, 1.5 Hz, 1H) 5.75 (q, J=6.5, 1H) 3.88 (s, 3H) 3.64 (m, 4H) 2.97 (br. s., 4H) 1.49 (d, J=6.6, 3H); MS (ESI) m/z [M+H]+ calcd. for C24H26ClN6O2: 465.2, Found: 465.2; HPLC purity: 210 nm: 100.0%; 254 nm: 100.0%. Example 23: Synthesis of Exemplary Compound 346 tert-butyl 4-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-a]pyridin-3-yl)-1,4-diazepane-1- carboxylate (Compound 346)
Figure imgf000227_0001
Step 1. Preparation of 4-(1-methyl-1H-pyrazol-4-yl)pyridin-2-amine
Figure imgf000227_0002
[0563] To a solution of 4-bromopyridin-2-amine (10.0 g, 57.8 mmol) in dioxane (175 mL) and H2O (35 mL) was added 1-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)- 1H-pyrazole (15.6 g, 75.1 mmol), Na2CO3 (12.2 g, 115.6 mmol), and Pd(dppf)Cl2 (1.2 g, 1.7 mmol) at rt under nitrogen then the reaction was stirred at 100°C under nitrogen for 4 h. The mixture was cooled to rt, filtered through a Celite pad, and the filtrate was poured into H2O (300 mL) and extracted with EtOAc (4 x 150 mL). The combined organic extracts were washed with brine (100 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography to afford 9.8 g (95%) of 4-(1-methyl-1H-pyrazol-4-yl)pyridin-2-amine as a yellow solid. 1HNMR (400 MHz, DMSO-d6) δ (ppm) 8.14 (s, 1H), 7.84 (d, J= 5.2Hz, 2H), 7.82 (s, 1H), 6.67 (d, J=5.2Hz, 1H), 6.55 (s, 1H), 5.80 (s, 2H), 3.86 (s, 3H). Step 2. Preparation of tert-butyl 4-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-a]pyridin-3- yl)-1,4-diazepane-1-carboxylate (Compound 346)
Figure imgf000228_0001
[0564] To a solution of 4-(1-methyl-1H-pyrazol-4-yl)pyridin-2-amine (1.0 g, 5.7 mmol, as prepared in the previous step), di-tert-butyl 4,4'-(1,2-bis(1H-benzo[d][1,2,3]triazol-1- yl)ethane-1,2-diyl)bis(1,4-diazepane-1-carboxylate) (Compound S48) (5.6 g, 8.5 mmol) in DCM (10 mL) was added ZnBr2 (3.8 g, 17.1 mmol) then the reaction was stirred at reflux under nitrogen for 5 h. The reaction mixture was cooled to rt, poured into 1N NaOH (40 mL), and extracted with EtOAc (3 x 50 mL). The combined organic extracts were washed with brine (30 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography and prep-HPLC to afford 120 mg (5%) of tert-butyl 4-(7-(1-methyl-1H- pyrazol-4-yl)imidazo[1,2-a]pyridin-3-yl)-1,4-diazepane-1-carboxylate (Compound 346) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.26 (s, 1H), 8.20~8.22 (m, 1H), 7.86 (s, 1H), 7.82~7.84 (m, 1H), 7.23~7.33 (d, J=6.8 Hz, 1H), 7.28 (s, 1H), 3.89 (s, 3H), 3.52~3.54 (m, 4H), 3.14~3.18 (m, 4H), 1.82~1.83 (m, 2H), 1.41 (d, J=11.6 Hz, 9H). Example 24: Synthesis of Exemplary Compound 347 (R)-1-phenylethyl 4-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-a]pyridin-3-yl)-1,4- diazepane-1-carboxylate (Compound 347)
Figure imgf000228_0002
Step 1. Preparation of (R)-4-nitrophenyl (1-phenylethyl) carbonate
Figure imgf000229_0001
[0565] To a solution of 4-nitrophenyl chloroformate (4.0 g, 19.8 mmol) and (R)-1- phenylethanol (1.21 g, 9.9 mmol) in DCM (50 mL) was added pyridine (1.57 g, 19.8 mmol) at rt then the reaction was stirred at rt for 3h. The mixture was concentrated under reduced pressure then the residue was purified by silica gel chromatography to afford 3.7 g (65%) of (R)-4-nitrophenyl (1-phenylethyl) carbonate as yellow oil.1HNMR (400 MHz, CDCl3) δ (ppm) 8.21-8.28 (m, 2H), 7.25-7.41 (m, 7H), 5.82-5.87 (m, 1H), 1.70 (d, J= 6.4 Hz, 3H). Step 2. Preparation of (R)-1-phenylethyl 4-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2- a]pyridin-3-yl)-1,4-diazepane-1-carboxylate (Compound 347)
Figure imgf000229_0002
[0566] To a solution of 3-(1,4-diazepan-1-yl)-7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2- a]pyridine hydrochloride salt (1.69 g, 5.1 mmol, as prepared in Example 29, Step 1) in ACN (15 mL) was added (R)-4-nitrophenyl (1-phenylethyl) carbonate (1.47 g, 5.1 mmol, as prepared in the previous step) and DIEA (1.98 g, 15.3 mmol) then the reaction was stirred at 80°C under nitrogen for 4 h. The mixture was cooled to rt, then poured into water (100 mL) and extracted with EtOAc (4 x 30 mL). The combined organic extracts were washed with brine, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by prep-HPLC to afford 169 mg (8%) of (R)-1-phenylethyl 4-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-a]pyridin-3-yl)-1,4- diazepane-1-carboxylate (Compound 347) as a yellow solid.1H NMR (400 MHz, DMSO- d6) δ (ppm) 8.28 (s, 1H), 8.02 (s, 1H), 8.00 (s, 1H), 7.71 (s, 1H), 7.15~7.35 (m, 7H), 5.75~5.78 (m, 1H), 3.88 (s, 3H), 3.66~3.68 (m, 2H), 3.56~3.61 (m, 2H), 3.12~3.24 (m, 4H), 1.86 (s, 2H), 1.45~1.49 (m, 3H). Example 25: Synthesis of Exemplary Compound 348 3-(3-(5-benzylpyrimidin-2-yl)pyrrolidin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- b]pyridazine (Compound 348)
Figure imgf000230_0001
[0567] To a solution of 5-benzyl-2-pyrrolidin-3-ylpyrimidine (Compound S100) (344.2 mg, 1.44 mmol) in dioxane (8 mL) was added 3-bromo-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-b]pyridazine (Compound S8) (200.0 mg, 0.72 mmol), Cs2CO3 (468.6 mg, 1.44 mmol), and tBuXPhos Pd G1 (74.1 mg, 0.11 mmol) at rt under nitrogen then the reaction was stirred at 110°C under nitrogen for 10 h. The mixture was cooled to rt, then concentrated under reduced pressure. The residue purified by prep-HPLC to afford 7.0 mg (2%) of 3-(3-(5-benzylpyrimidin-2-yl)pyrrolidin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-b]pyridazine (Compound 348) as a yellow solid. 1HNMR (400 MHz, CDCl3) δ (ppm) 8.55 (s, 2H), 7.94-7.98 (m, 3H), 7.45 (s, 2H), 7.31-7.35 (m, 1H), 7.27-7.28 (m, 1H), 7.19 (d, J = 7.6 Hz, 2H), 6.83 (d, J = 9.6 Hz, 1H), 3.97 (s, 3H), 3.82-3.84 (m, 2H), 3.76-3.80 (m, 3H), 3.53-3.59 (m, 2H), 2.48-2.52 (m, 2H). Example 26: Synthesis of Exemplary Compound 349 3-(3-(1-benzyl-1H-1,2,4-triazol-3-yl)pyrrolidin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-b]pyridazine (Compound 349)
Figure imgf000230_0002
[0568] To a solution of 1-benzyl-3-(pyrrolidin-3-yl)-1H-1,2,4-triazole (Compound S101) (492.5 mg, 2.16 mmol) in dioxane (10 mL) was added 3-bromo-6-(1-methyl-1H-pyrazol- 4-yl)pyrazolo[1,5-b]pyridazine (Compound S8) (300.0 mg, 1.08 mmol), Cs2CO3 (1.05 g, 3.24 mmol), and tBuXPhos Pd G1 (111.1 mg, 0.16 mmol) at rt under nitrogen then the reaction was stirred at 110°C under nitrogen for 16 h. The reaction mixture was cooled to rt, then concentrated under reduced pressure. The residue was purified by prep-HPLC to afford 4.0 mg (1%) of 3-(3-(1-benzyl-1H-1,2,4-triazol-3-yl)pyrrolidin-1-yl)-6-(1-methyl- 1H-pyrazol-4-yl)pyrazolo[1,5-b]pyridazine (Compound 349) as a yellow solid. 1H NMR (400 MHz, CDCl3) δ (ppm) 9.24 (d, J = 9.2 Hz, 1H), 8.43 (s, 1H), 8.38 (s, 1H), 8.05 (d, J = 10.0 Hz, 2H), 7.40-7.45 (m, 2H), 7.33-7.36 (m, 3H), 7.21-7.22 (m, 1H), 5.38 (s, 2H), 4.35-4.42 (m, 3H), 4.10-4.23 (m, 2H), 4.01 (s, 3H), 2.95-3.00 (m, 1H), 2.64-2.70 (m, 1H). Example 27: Synthesis of Exemplary Compound 350 3-(3-(5-benzylpyrimidin-2-yl)pyrrolidin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrazine (Compound 350)
Figure imgf000231_0001
Step 1. Preparation of 5-benzyl-2-(pyrrolidin-3-yl)pyrimidine trihydrochloride salt
Figure imgf000231_0002
[0569] To a solution of tert-butyl 3-(5-benzylpyrimidin-2-yl)pyrrolidine-1-carboxylate (3.00 g, 8.84 mmol, as prepared for Compound S100, Step 2) in dioxane (20 mL) was added 4M HCl in dioxane (10 mL) at rt then the reaction was stirred at rt for 2 h. The precipitated solids were collected by filtration, washed with dioxane (2 x 3 mL), and dried to afford 2.4 g (113%) of 5-benzyl-2-(pyrrolidin-3-yl)pyrimidine trihydrochloride salt as a dark green oil: MS (ESI) m/z [M+H]+ calcd. for C15H17N3, 240.1; found, 240.1. Step 2. Preparation of 3-(3-(5-benzylpyrimidin-2-yl)pyrrolidin-1-yl)-6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyrazine (Compound 350)
Figure imgf000231_0003
[0570] To a solution of 3-bromo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrazine (Compound S5) (200 mg, 0.719 mmol) and 5-benzyl-2-(pyrrolidin-3-yl)pyrimidine trihydrochloride salt (502 mg, 1.44 mmol, as prepared in the previous step) in tBuOH (12 mL) and dioxane (4 mL) was added KOtBu (807 mg, 7.19 mmol) and tBuXPhos Pd G1 (99 mg, 0.144 mmol) under nitrogen then the reaction was stirred at 90°C under nitrogen for 40 h. The mixture was cooled to rt, then concentrated under reduced pressure. The residue was purified by silica gel chromatography eluting with EtOAc / PE (1:1) and Prep-HPLC to afford 3.6 mg (1%) of 3-(3-(5-benzylpyrimidin-2-yl)pyrrolidin-1-yl)-6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyrazine (Compound 350) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.08 (d, J = 1.4 Hz, 1H), 8.73 (d, J = 1.4 Hz, 1H), 8.70 (s, 2H), 8.16 (s, 1H), 7.99 – 7.94 (m, 1H), 7.55 (s, 1H), 7.36 – 7.25 (m, 4H), 7.25 – 7.17 (m, 1H), 3.97 (s, 2H), 3.87 (s, 3H), 3.85 – 3.77 (m, 2H), 3.71 – 3.65 (m, 1H), 3.61 – 3.53 (m, 2H), 2.49 – 2.29 (m, 2H); MS (ESI) m/z [M+H]+ calcd. for C25H24N8, 437.2; found, 437.2; LCMS purity: 254 nm: 99.5% Example 28: Synthesis of Exemplary Compound 351 (R)-1-phenylethyl 4-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-a]pyridin-3-yl)piperazine-1- carboxylate (Compound 351)
Figure imgf000232_0001
Step 1. Preparation of 7-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)imidazo[1,2- a]pyridine hydrochloride salt
Figure imgf000232_0002
[0571] To a solution of tert-butyl 4-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-a]pyridin- 3-yl)piperazine-1-carboxylate (Compound 345) (8.2 g, 21.4 mmol) in DCM (80 mL) cooled to 0°C was added 4M HCl in dioxane (22 mL) then the reaction was warmed to rt and stirred for 2 h. The mixture was filtered and the filter cake was dried to afford 6.2 g (91%) of 7-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)imidazo[1,2-a]pyridine hydrochloride salt as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.74 (d, J = 6.8 Hz, 1H), 8.65 (s, 1H), 8.26 (s, 1H), 7.91 (s, 1H), 7.72~7.74 (m, 1H), 7.43~7.45 (m, 1H), 3.92 (s, 3H), 3.56 (s, 2H), 3.32~3.34 (m, 6H). Step 2. Preparation of (R)-1-phenylethyl 4-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2- a]pyridin-3-yl)piperazine-1-carboxylate (Compound 351)
Figure imgf000233_0001
[0572] To a solution of 7-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)imidazo[1,2- a]pyridine hydrochloride salt (3.0 g, 9.4 mmol, as prepared in the previous step) in ACN (30 mL) was added (R)-4-nitrophenyl (1-phenylethyl) carbonate (2.7 g, 9.4 mmol, as prepared in Example 23, Step 1) and DIEA (3.6 g, 28.2 mmol) then the reaction was stirred at 80°C under nitrogen for 4 h. The reaction mixture was cooled to rt, poured into water (80 mL), and extracted with EtOAc (4 x 30 mL). The combined organic extracts were washed with brine, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by prep-HPLC to afford 65 mg (2%) of (R)-1-phenylethyl 4-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-a]pyridin- 3-yl)piperazine-1-carboxylate (Compound 351) as a yellow solid. 1H NMR (400 MHz, CDCl3) δ (ppm) 7.93~7.95 (m, 1H), 7.80 (s, 1H), 7.68 (s, 1H), 7.59 (s, 1H), 7.36~7.37 (m, 4H), 7.30~7.32 (m, 1H), 7.24~7.25 (m, 1H), 6.93~6.95 (m, 1H), 5.86~5.89 (m, 1H), 3.97 (s, 3H), 3.71 (s, 4H), 3.02 (s, 4H), 1.57~1.59 (d, J=6.4Hz, 3H). Example 29: Synthesis of Exemplary Compound 352 3-(4-(5-benzylpyrimidin-2-yl)-1,4-diazepan-1-yl)-7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2- a]pyridine (Compound 352)
Figure imgf000233_0002
Step 1. Preparation of 3-(1,4-diazepan-1-yl)-7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2- a]pyridine hydrochloride salt
Figure imgf000233_0003
[0573] To a solution of tert-butyl 4-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-a]pyridin- 3-yl)-1,4-diazepane-1-carboxylate (Compound 346) (5.5 g, 13.9 mmol) in DCM (55 mL) cooled to 0°C was added 4M HCl in dioxane (18 mL) then the reaction was warmed to rt and stirred for 2 h. The mixture was filtered and the filter cake was dried to afford 5.9 g (128%) of 3-(1,4-diazepan-1-yl)-7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-a]pyridine hydrochloride salt as a yellow solid.1HNMR (400 MHz, DMSO-d6) δ (ppm) 8.74 (d, J = 7.2 Hz, 1H), 8.62 (s, 1H), 7.95 (s, 1H), 7.71 (d, J = 7.2 Hz, 1H), 7.65~7.68 (m, 1H), 7.43~7.45 (m, 1H), 3.92 (m, 3H), 3.46~3.53 (m, 4H), 3.24~3.32 (m, 3H), 2.13 (s, 3H). Step 2. Preparation of 3-(4-(5-benzylpyrimidin-2-yl)-1,4-diazepan-1-yl)-7-(1-methyl-1H- pyrazol-4-yl)imidazo[1,2-a]pyridine (Compound 352)
Figure imgf000234_0001
[0574] To a solution of 3-(1,4-diazepan-1-yl)-7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2- a]pyridine hydrochloride salt (0.2 g, 0.6 mmol, as prepared in the previous step) in dioxane (10 mL) was added Cs2CO3 (0.59 g, 1.8 mmol), 5-benzyl-2-chloropyrimidine (Compound S88) (0.12 g, 0.6 mmol), and tBuXPhos Pd G1 (0.12 g, 0.18 mmol) then the reaction was stirred at 90°C under nitrogen for 6 h. The reaction mixture was cooled to rt, poured into water (40 mL), and extracted with EtOAc (3 x 50 mL). The combined organic extracts were washed with brine, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by prep-HPLC to afford 30 mg (11%) of 3-(4-(5-benzylpyrimidin-2-yl)-1,4-diazepan-1-yl)-7-(1-methyl-1H- pyrazol-4-yl)imidazo[1,2-a]pyridine (Compound 352) as a yellow solid. 1HNMR (400 MHz, CDCl3) δ (ppm) 8.20 (s, 2H), 7.79~7.82 (m, 1H), 7.71 (s, 1H), 7.65 (s, 1H), 7.34 (s, 1H), 7.28~7.32 (m, 2H), 7.15~7.24 (m, 4H), 6.85~6.87 (m, 1H), 3.99~4.02 (m, 4H), 3.96 (s, 3H), 3.81 (s, 2H), 3.55~3.62 (m, 2H), 3.18~3.20 (m, 2H), 2.04~2.07 (m, 2H). Example 30: Synthesis of Exemplary Compound 353 N-((5-chloropyridin-2-yl)methyl)-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxamide (Compound 353)
Figure imgf000235_0001
[0575] To a solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-piperazin-1-ylpyrazolo[1,5- a]pyridine (Compound 12) (300 mg, 1.06 mmol), TEA (323 mg, 3.19 mmol) in DCM (15 mL) was added triphosgene (94.6 mg, 0.32 mmol) at 0°C. The resulting mixture was stirred for 0.5 h at 0°C. Then resulting mixture was added 1-(5-chloropyridin-2-yl)methanamine (303 mg, 2.13 mmol) at 0°C. The resulting mixture was stirred for 16h at rt. Desired product could be detected. The resulting mixture was concentrated under reduced pressure. The residue was purified by silica gel chromatography, eluted with DCM / MeOH (10:1).The crude product (600 mg) was purified by Prep-HPLC with the following conditions (Column: Xselect CSH Prep Fluoro-Phenyl Column, 19*250 mm, 5μm; Mobile Phase A: Water (0.1% FA), Mobile Phase B: ACN; Flow rate: 25 mL/min mL/min; Gradient: 33% B to 43% B in 12min; Wave Length: 254nm/220nm nm; RT1(min): 8.65) to afford N-((5- chloropyridin-2-yl)methyl)-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxamide (279.7 mg, 57.97%) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.87 (s, 1H), 8.55 (d, J = 2.5 Hz, 1H), 8.25 (s, 1H), 7.99 (s, 1H), 7.94 – 7.85 (m, 2H), 7.77 (d, J = 9.2 Hz, 1H), 7.42 – 7.32 (m, 3H), 4.35 (s, 2H), 3.88 (s, 3H), 3.64 – 3.57 (m, 4H), 3.09 (s, 4H); MS (ESI) m/z [M+H]+ calcd. for C22H23ClN8O, 451.2; found, 451.2; LCMS purity: 254 nm: 99.3% Example 31: Synthesis of Exemplary Compound 354 N-((6-chloropyridin-3-yl)methyl)-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxamide (Compound 354)
Figure imgf000235_0002
[0576] To a solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)pyrazolo[1,5- a]pyridine (Compound 12) (400 mg, 1.42 mmol) in DCM (5 mL) cooled to 0°C was added triphosgene (126.1 mg, 0.43 mmol) then the reaction was stirred at 0°C under nitrogen for 16 h. The mixture was added dropwise to a DCM (5 mL) solution of (6-chloropyridin-3- yl)methanamine (148.8 mg, 1.10 mmol) and TEA (176.1 mg, 1.74 mmol) cooled to 0°C, then stirred at 0°C under nitrogen for 2 h. The mixture was concentrated under reduced pressure then the residue was dissolved in water (30 mL) and extracted with DCM (3 x 50 mL). The combined organic extracts were washed with brine (10 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 131.6 mg (33%) of N-((6-chloropyridin-3-yl)methyl)- 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxamide (Compound 354) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (s, 1H), 8.37 – 8.21 (m, 2H), 7.98 (s, 1H), 7.79 – 7.23 (m, 2H), 7.70 – 7.63 (m, 1H), 7.48 (d, J = 8.2 Hz, 1H), 7.36 – 7.24 (m, 2H), 4.27 (d, J = 5.6 Hz, 2H), 3.87 (s, 3H), 3.52 (t, J = 4.9 Hz, 4H), 2.95 (t, J = 4.9 Hz, 4H); MS (ESI) m/z [M+H]+ calcd. for C22H23ClN8O, 451.2; found, 451.1; LCMS purity: 254 nm: 99.7% Example 32: Synthesis of Exemplary Compound 355 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)-N-((5-methylpyridin-2- yl)methyl)piperazine-1-carboxamide (Compound 355)
Figure imgf000236_0001
[0577] To a solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)pyrazolo[1,5- a]pyridine (Compound 12) (500 mg, 1.77 mmol) in DCM (20 mL) cooled to 0°C was added TEA (537.6 mg, 5.31 mmol) and triphosgene (173.4 mg, 0.58 mmol) at 0°C then the reaction was warmed to rt and stirred under nitrogen for 30 minutes. To the reaction was added (5-methylpyridin-2-yl)methanamine (432.7 mg, 3.54 mmol) dropwise over 1 minute then the reaction was stirred at rt for 16 h. The mixture was diluted with water (100 mL) and extracted with DCM (3 x 50 mL). The combined organic extracts were washed with brine (3x50 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 250.1 mg (32%) of 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)-N-((5- methylpyridin-2-yl)methyl)piperazine-1-carboxamide (Compound 355) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (s, 1H), 8.32 (d, J = 2.2 Hz, 1H), 8.23 (s, 1H), 7.98 (s, 1H), 7.78 (s, 1H), 7.68 (dd, J = 9.2, 0.8 Hz, 1H), 7.57 (dd, J = 8.0, 2.4 Hz, 1H), 7.33 (dd, J = 9.2, 1.5 Hz, 1H), 7.24 (t, J = 6.0 Hz, 1H), 7.19 (d, J = 8.0 Hz, 1H), 4.32 (d, J = 5.6 Hz, 2H), 3.88 (s, 3H), 3.55 (d, J = 9.9 Hz, 4H), 2.97 (t, J = 4.8 Hz, 4H), 2.27 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C23H26N8O, 431.2; found, 431.2; LCMS purity: 254 nm: 99.3%. Example 33: Synthesis of Exemplary Compound 356 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)-N-((6-methylpyridin-3- yl)methyl)piperazine-1-carboxamide (Compound 356)
Figure imgf000237_0001
[0578] To a solution of (6-methylpyridin-3-yl)methanamine (400 mg, 3.27 mmol) and TEA (994 mg, 9.82 mmol) in DCM (5 mL) cooled to 0°C was added triphosgene (291.5 mg, 0.98 mmol) then the reaction was stirred at 0°C under nitrogen for 2 h. The mixture was added to a solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)pyrazolo[1,5- a]pyridine (Compound 12) (686 mg, 2.43 mmol) and TEA (409.8 mg, 4.05 mmol) in DCM (5 mL) cooled to 0°C, then the reaction was stirred at 0°C under nitrogen for 2 h. The mixture was concentrated under reduced pressure, then water (30 mL) was added and the mixture was extracted with EtOAc (3 x 30 mL). Then combined organic extracts were washed with brine (10 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 100 mg (11%) of 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)-N-((6- methylpyridin-3-yl)methyl)piperazine-1-carboxamide (Compound 356) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.35 (d, J = 2.3 Hz, 1H), 8.23 (s, 1H), 7.97 (s, 1H), 7.77 (s, 1H), 7.66 (d, J = 9.2 Hz, 1H), 7.56 (dd, J = 8.0, 2.3 Hz, 1H), 7.32 (dd, J = 9.3, 1.5 Hz, 1H), 7.23-7.16 (m, 2H), 4.23 (d, J = 5.6 Hz, 2H), 3.87 (s, 3H), 3.51 (t, J = 4.8 Hz, 4H), 2.94 (t, J = 4.8 Hz, 4H), 2.43 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C23H26N8O, 430.2; found, 431.1; LCMS purity: 254 nm: 99.5% Example 34: Synthesis of Exemplary Compound 357 3-(4-(1-benzyl-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyrimidine (Compound 357)
Figure imgf000238_0001
[0579] To a solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)pyrazolo[1,5- a]pyrimidine (Compound S98) (250 mg, 0.88 mmol) and 1-benzyl-3-chloro-1H-1,2,4- triazole (256.2 mg, 1.32 mmol, as prepared in Example 1, Step 1) in dioxane (20 mL) were added Pd-PEPPSI-IPentCl 2-methylpyridine (111.3 mg, 0.13 mmol) and Cs2CO3 (574.9 mg, 1.76 mmol) then the reaction was stirred at 100°C under nitrogen overnight. The mixture was cooled to rt and concentrated under reduced pressure. The residue was dissolved in water (50 mL), then the mixture was extracted with DCM (3 x 50 mL). The combined organic extracts were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 74.3 mg (19%) of 3-(4-(1-benzyl-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyrimidine (Compound 357) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.17 (d, J = 2.2 Hz, 1H), 8.63 (d, J = 2.1 Hz, 1H), 8.31 – 8.29 (m, 2H), 8.03 (s, 1H), 7.90 (s, 1H), 7.44 – 7.20 (m, 5H), 5.23 (s, 2H), 3.89 (s, 3H), 3.46 – 3.43 (m, 4H).3.32 – 3.29 (m, 4H); MS (ESI) m/z [M+H]+ calcd. for C23H24N10, 441.2; found, 441.15.LCMS purity:254 nm:99.1%. Example 35: Synthesis of Exemplary Compound 358 3-(3-(1-benzyl-1H-1,2,4-triazol-3-yl)pyrrolidin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine (Compound 358)
Figure imgf000238_0002
[0580] To a vial pre-sparged with argon 20 minutes containing 3-iodo-6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridine (234 mg, 0.72 mmol, as prepared for Compound S57, Step 1), 1-benzyl-3-pyrrolidin-3-yl-1H-1,2,4-triazole (Compound S101) (110 mg, 0.48 mmol), K3PO4 (307 mg, 1.44 mmol), and CuI (37 mg, 0.193 mmol) was added 1-butanol (2.8 mL) and 1, 2-ethanediol (0.6 mL) then the reaction was heated to 100°C for 48 h. The mixture was cooled to rt, diluted with 10% IPA / DCM, and washed with (7:3) sat. aqueous NH4Cl / conc. NH4OH (3 times) and brine, then dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with 0-6% MeOH / DCM. The pure fractions were concentrated under reduced pressure then the residue was stirred with a mixture of (6:1:2) MTBE/MeOH/heptane. The solvent was removed under reduced pressure then the solid was triturated with MTBE (1 mL) overnight. The solid was collected by filtration and dried under reduced pressure to afford 29 mg (16%) of 3-[3-(1-benzyl-1H-1,2,4-triazol-3- yl)pyrrolidin-1-yl]-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 358) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.69 (s, 1H), 8.55 (s, 1H), 8.18 (s, 1H), 7.93 (s, 1H), 7.68 (d, J = 9.3 Hz, 1H), 7.53 (s, 1H), 7.40-7.25 (m, 5H), 7.11 (m, 1H), 5.36 (s, 2H), 3.86 (s, 3H), 3.70-3.54 (m, 2H), 3.48-3.32 (m, 3H), 2.40-2.17 (m, 2H); MS (ESI) m/z [M+H]+ calcd. for C24H25N8, 425.2; found, 425.2; HPLC purity: 210 nm: 95.8%; 254 nm: 96.2%. Example 36: Synthesis of Exemplary Compound 359 (R)-1-(6-methylpyridin-3-yl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate (Compound 359)
Figure imgf000239_0001
Step 1. Preparation of (1R)-1-(6-methylpyridin-3-yl)ethanol
Figure imgf000239_0002
[0581] To a solution of (R,R)-Ts-DENEB (0.263 g, 0.405 mmol) in toluene (5 mL) under nitrogen was added 3-acetyl-6-methylpyridine (5.48 g, 40.5 mmol) at rt then TEA (14.1 mL, 101 mmol) and formic acid (3.82 mL, 101 mmol) were added. The reaction was stirred at rt overnight then the mixture was partitioned between sat. aqueous NaHCO3 (100 mL) and EtOAc (100 mL). The layers were separated, and the aqueous phase was extracted with EtOAc (2 x 100 mL). The combined organic extracts were washed with sat. aqueous NaHCO3 (50 mL). The combined aqueous layers were extracted with EtOAc (100 mL). The organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with 30-100% acetone / DCM to afford 4 g (72%) of (R)-1-(6- methylpyridin-3-yl)ethanol as a dark solid. 1H NMR (300 MHz, CDCl3) δ (ppm) 8.30 - 8.46 (m, 1H), 7.62 (dd, J=7.96, 2.10 Hz, 1H), 7.12 (d, J=8.01 Hz, 1H), 4.90 (q, J=6.33 Hz, 1H), 3.12 (s, 1H), 2.51 (s, 3H), 1.49 (d, J=6.43 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C8H12NO: 138.2; found: 137.9. Step 2. Preparation of (R)-1-(6-methylpyridin-3-yl)ethyl 4-[6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1, 5-a]pyridin-3-yl]piperazine-1-carboxylate (Compound 359)
Figure imgf000240_0001
[0582] To a solution of (R)-1-(6-methylpyridin-3-yl)ethanol (1.30 g, 9.44 mmol, as prepared in the previous step) in NMP (20 mL) cooled to 0°C was added 1M LiHMDS in THF (8.64 mL, 8.64 mmol) under nitrogen over 10 minutes, then the reaction was stirred at 0°C for 20 minutes. 4-nitrophenyl 4-[6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl]piperazine-1-carboxylate (4.00 g, 7.87 mmol, as prepared in Example 18, Step 1) was added portion wise as a solid, then the mixture was stirred at 0°C for 15 minutes. The reaction was warmed to rt and stirred overnight. The reaction mixture was diluted with EtOAc (100 mL) then washed with sat. aqueous NaHCO3 (3 x 100 mL) and water (4 x 100 mL). The combined aqueous washes were extracted with EtOAc (50 mL). The combined organic extracts were washed with brine (50 mL), dried over Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with 5% MeOH / DCM followed by silica gel chromatography eluting with 20-80% acetone / DCM to afford 2.3g (54%) of (R)-1-(6- methylpyridin-3-yl)ethyl 4-[6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl]piperazine-1-carboxylate (Compound 359) as a yellow foam. 1H NMR (300 MHz, CDCl3) δ (ppm) 8.54 (d, J=2.05 Hz, 1H), 8.46 (s, 1H), 7.74 (s, 1H), 7.70 (s, 1H), 7.53 - 7.65 (m, 2H), 7.49 (d, J=9.22 Hz, 1H), 7.07 - 7.21 (m, 2H), 5.86 (q, J=6.61 Hz, 1H), 3.89 - 4.09 (m, 3H), 3.70 (br. s., 4H), 2.93 - 3.13 (m, 4H), 2.56 (s, 3H), 1.60 (d, J=6.61 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C24H28N7O2: 446.5; found: 446.0; HPLC purity: 210 nm: 100.0%; 254 nm: 100.0% Example 37: Synthesis of Exemplary Compound 360 5-benzyl-2-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1- yl)thiazole (Compound 360)
Figure imgf000241_0001
Step 1. Preparation of 5-benzyl-2-chlorothiazole
Figure imgf000241_0002
[0583] To a solution of CuCl2 (848 mg, 6.31 mmol) in ACN (20 mL) was added tert-butyl nitrite (867.2 mg, 8.41 mmol) at rt. The reaction was stirred for 30 minutes then 5-benzyl- 1,3-thiazol-2-amine (1 g, 5.26 mmol) was added. The mixture was stirred at rt for 1 h then 1M HCl was added and the reaction was stirred at rt for 1 h, then concentrated under reduced pressure. Then residue was purified by silica gel chromatography eluting with EtOAc / PE (1:5) to afford 800 mg (73%) of 5-benzyl-2-chlorothiazole as a light yellow solid.1H NMR (400 MHz, CDCl3) δ 7.39 – 7.33 (m, 2H), 7.32 – 7.29 (m, 2H), 7.24 (dd, J = 6.8, 1.6 Hz, 2H), 4.11 (s, 2H); MS (ESI) m/z [M+H]+ calcd. for C10H8ClNS, 210.0; found, 210.1. Step 2. Preparation of 5-benzyl-2-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazin-1-yl)thiazole (Compound 360)
Figure imgf000241_0003
[0584] To a solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)pyrazolo[1,5- a]pyridine (Compound 12) (200 mg, 0.71 mmol) and 5-benzyl-2-chlorothiazole (222.8 mg, 1.06 mmol, as prepared in the previous step) in ACN (15 mL) was added Cs2CO3 (577 mg, 1.77 mmol) then the reaction was stirred at 90°C for 48 h. The mixture was cooled to rt, then concentrated under reduced pressure. The residue was diluted with water (100 mL) and extracted with EtOAc (2 x 200 mL). The combined organic extracts were washed with brine (2x100 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 175.3 mg (54%) of 5-benzyl-2-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazin-1-yl)thiazole (Compound 360) as a light yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (s, 1H), 8.23 (s, 1H), 7.98 (s, 1H), 7.79 (s, 1H), 7.67 (dd, J = 9.2, 1.0 Hz, 1H), 7.38 – 7.18 (m, 6H), 7.00 (s, 1H), 4.00 (s, 2H), 3.87 (s, 3H), 3.52 (t, J = 4.4 Hz, 4H), 3.13 – 3.03 (m, 4H); MS (ESI) m/z [M+H]+ calcd. for C25H25N7S, 456.1; found, 456.1; LCMS purity: 254 nm: 99.1% Example 38: Synthesis of Exemplary Compound 361 5-benzyl-3-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)- 1,2,4-oxadiazole (Compound 361)
Figure imgf000242_0001
Step 1. Preparation of 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carbonitrile
Figure imgf000242_0002
[0585] To a solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)pyrazolo[1,5- a]pyridine (Compound 12) (500 mg, 1.77 mmol) and TEA (268.8 mg, 2.66 mmol) in DCM (6 mL) cooled to 0°C was added BrCN (187.6 mg, 1.77 mmol) then the reaction was stirred at 0°C for 3 h. The mixture was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with MeOH / DCM (1:30) to afford 410 mg (75%) of 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1- carbonitrile as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.84 (s, 1H), 8.24 (s, 1H), 7.98 (d, J = 0.6 Hz, 1H), 7.81 (s, 1H), 7.66 (dd, J = 9.2, 0.7 Hz, 1H), 7.35 (dd, J = 9.2, 1.5 Hz, 1H), 3.87 (s, 3H), 3.41 (t, J = 4.9 Hz, 4H), 3.06 (t, J = 4.9 Hz, 4H); MS (ESI) m/z [M+H]+ calcd. for C16H17N7, 308.1; found, 308.2. Step 2. Preparation of N'-hydroxy-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazine-1-carboximidamide
Figure imgf000243_0001
[0586] To a solution of 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carbonitrile (400 mg, 1.30 mmol, as prepared in the previous step) and TEA (263.4 mg, 2.60 mmol) in EtOH (6 mL) cooled to 0°C was added NH2OH·HCl (90.4 mg, 1.30 mmol) then the reaction was warmed to rt and stirred for 5 h. The mixture was filtered, the filter cake was washed with EtOH (3 x 20 mL),and dried under reduced pressure to afford 400 mg (90%) of N'-hydroxy-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazine-1-carboximidamide as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 10.73 (s, 1H), 10.02 (s, 1H), 8.85 (s, 1H), 8.25 (s, 1H), 8.10 (s, 2H), 7.99 (s, 1H), 7.80 (s, 1H), 7.71 (d, J = 9.2 Hz, 1H), 7.36 (dd, J = 9.3, 1.4 Hz, 1H), 3.88 (s, 3H), 3.59 (t, J = 4.6Hz, 4H), 3.06 (t, J = 4.6 Hz, 4H); MS (ESI) m/z [M+H]+ calcd. for C16H20N8O, 341.1; found, 341.2. Step 3. Preparation of 5-benzyl-3-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazin-1-yl)-1,2,4-oxadiazole (Compound 361)
Figure imgf000243_0002
[0587] To a solution of 2-phenylacetic acid (160 mg, 1.17 mmol) in THF (6 mL) was added CDI (191 mg, 1.17 mmol) then the reaction was stirred for 1 h at 55°C. The mixture was cooled to rt and N'-hydroxy-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboximidamide (400 mg, 1.17 mmol, as prepared in the previous step) was added. The reaction was heated to 55°C and stirred for 1 h then the mixture was heated to 150°C for 20 minutes. The reaction was cooled to rt and concentrated under reduced pressure. The residue was purified by silica gel chromatography eluting with MeOH / DCM (1:15) then the pure fractions were concentrated under reduced pressure. The residue was triturated with MeOH to afford 100.5 mg (19%) of 5-benzyl-3-(4-(6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)-1,2,4-oxadiazole (Compound 361) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (s, 1H), 8.23 (s, 1H), 7.98 (s, 1H), 7.78 (s, 1H), 7.67 (d, J = 9.3 Hz, 1H), 7.44 – 7.18 (m, 6H), 4.23 (s, 2H), 3.87 (s, 3H), 3.52 (t, J = 4.9 Hz, 4H), 3.07 (t, J = 4.9 Hz, 4H); MS (ESI) m/z [M+H]+ calcd. for C24H24N8O, 441.1; found, 441.20; LCMS purity: 254 nm: 99.5%. Example 39: Synthesis of Exemplary Compound 362 (R)-N-(4-chlorobenzyl)-2-methyl-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxamide (Compound 362)
Figure imgf000244_0001
Step 1. Preparation of (R)-6-(1-methyl-1H-pyrazol-4-yl)-3-(3-methylpiperazin-1- yl)pyrazolo[1,5-a]pyridine hydrochloride salt
Figure imgf000244_0002
[0588] To a solution of tert-butyl (R)-2-methyl-4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (600 mg, 1.51 mmol, as prepared in Example 4, Step 1) in DCM (20 mL) cooled to 0°C was added 4M HCl in dioxane (10 mL) dropwise then the reaction was warmed to rt and stirred for 5 h. The mixture was concentrated under reduced pressure to afford 690 mg (137%) (R)-6-(1-methyl-1H- pyrazol-4-yl)-3-(3-methylpiperazin-1-yl)pyrazolo[1,5-a]pyridine hydrochloride salt as a brown solid: MS (ESI) m/z [M+H]+ calcd. for C16H20N6, 297.2; found, 297.3. Step 2. Preparation of (R)-N-(4-chlorobenzyl)-2-methyl-4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxamide (Compound 362)
Figure imgf000245_0001
[0589] To a solution of (R)-6-(1-methyl-1H-pyrazol-4-yl)-3-(3-methylpiperazin-1- yl)pyrazolo[1,5-a]pyridine hydrochloride salt (300 mg, 0.68 mmol, as prepared in the previous step) and TEA (410 mg, 4.05 mmol) in DCM (5 mL) cooled to 0°C was added triphosgene (60.1 mg, 0.20 mmol) then the reaction was stirred for 30 minutes. To the mixture at 0°C was added 1-(4-chlorophenyl)methanamine (191 mg, 1.35 mmol), then the reaction was warmed to rt and stirred for 2 h. The mixture was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with EtOAc / PE (5:1) followed by Prep-HPLC to afford 150.5 mg (48%) of (R)-N-(4-chlorobenzyl)-2- methyl-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1- carboxamide (Compound 362) as a light green solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.76 (s, 1H), 7.59 (d, J = 9.2 Hz, 1H), 7.42 – 7.35 (m, 2H), 7.35 – 7.25 (m, 3H), 7.16 (t, J = 5.9 Hz, 1H), 4.33 – 4.17 (m, 3H), 3.93 – 3.78 (m, 4H), 3.25 – 3.09 (m, 3H), 2.82 – 2.74 (m, 1H), 2.70 – 2.59 (m, 1H), 1.33 (d, J = 6.6 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C24H26ClN7O, 464.2; found, 464.2; LCMS purity: 254 nm: 99.4%. Example 40: Synthesis of Exemplary Compound 363 (S)-N-(4-chlorobenzyl)-2-methyl-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxamide (Compound 363)
Figure imgf000245_0002
Step 1. Preparation of (S)-6-(1-methyl-1H-pyrazol-4-yl)-3-(3-methylpiperazin-1- yl)pyrazolo[1,5-a]pyridine hydrochloride salt
Figure imgf000246_0001
[0590] To a solution of (S)-2-methyl-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazine-1-carboxylate (600 mg, 1.51 mmol, as prepared in Example 5, Step 1) in DCM (20 mL) was added 4M HCl in dioxane (551.7 mg, 15.13 mmol) then the reaction was stirred at rt under nitrogen overnight. The mixture was concentrated under reduced pressure to afford 500 mg (89%) of (S)-6-(1-methyl-1H-pyrazol-4-yl)-3-(3- methylpiperazin-1-yl)pyrazolo[1,5-a]pyridine hydrochloride salt as a grey solid. MS (ESI) m/z [M+H]+ calcd. for C16H20N6, 297.1; found, 297.1. Step 2. (S)-N-(4-chlorobenzyl)-2-methyl-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazine-1-carboxamide (Compound 363)
Figure imgf000246_0002
[0591] To a solution of 1-(4-chlorophenyl)methanamine (500 mg, 3.53 mmol) in DCM (20 mL) was added triphosgene (1 mg, 3.53 mmol) then the reaction was stirred at rt under nitrogen for 4 h. The solution was added dropwise to a mixture of (S)-6-(1-methyl-1H- pyrazol-4-yl)-3-(3-methylpiperazin-1-yl)pyrazolo[1,5-a]pyridine hydrochloride salt (265.2 mg, 0.89 mmol, as prepared in the previous step) and TEA (1.63 g, 16.11 mmol) in DCM (20 mL), then the reaction was stirred under nitrogen overnight. The mixture was concentrated under reduced pressure then the residue was dissolved in water (50 mL). The mixture was extracted with DCM (3 x 50 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure, then the residue was purified by Prep- HPLC to afford 80.9 mg (10%) of (S)-N-[(4-chlorophenyl)methyl]-2-methyl-4-[6-(1- methylpyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl]piperazine-1-carboxamide (Compound 363) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.76 (s, 1H), 7.59 (d, J = 9.2 Hz, 1H), 7.40 – 7.29 (m, 4H), 7.16 (t, J = 5.9 Hz, 1H), 4.29-4.23 (m, 3H), 3.87-3.83 (m, 4H), 3.28 – 3.11 (m, 3H), 2.80 – 2.76 (m, 1H), 2.64 – 2.54 (m,1H), 1.33 (d, J = 6.6 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C24H26ClN7O, 464.2; found, 464.2.LCMS purity:254 nm:99.5%; HPLC purity: 98.2%. Example 41: Synthesis of Exemplary Compound 364 (R)-1-(5-methylpyridin-2-yl)ethyl 4-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazin- 3-yl)piperazine-1-carboxylate (Compound 364)
Figure imgf000247_0001
Step 1. Preparation of 4-nitrophenyl 4-[7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2- b]pyridazin-3- yl]piperazine-1-carboxylate
Figure imgf000247_0002
[0592] To a suspension of 7-(1-methyl-1H-pyrazol-4-yl)-3-piperazin-1-ylimidazo[1,2- b]pyridazine hydrochloride salt (Compound S84) (0.201 g, 0.564 mmol) in ACN (4 mL) was added TEA (0.314 mL, 2.26 mmol) then the mixture was stirred for 5 minutes at rt.4- Nitrophenyl chloroformate (0.125 g, 0.620 mmol) was added and the mixture was stirred at rt overnight. The reaction was concentrated under reduced pressure then the residue was partitioned between 1N NaOH (10 mL) and DCM (10 mL). The layers were separated, and the DCM layer was extracted with 1N NaOH (2 x 10 mL). The combined aqueous layers were extracted with DCM (10 mL) then the combined DCM layers were dried over Na2SO4 and filtered. The filtrate was concentrated under reduced pressure to afford 190 mg (75%) of 4-nitrophenyl 4-[7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazin-3- yl]piperazine-1-carboxylate as a yellow solid. 1H NMR (400 MHz, CDCl3) δ (ppm) 8.52 (d, J=2.08 Hz, 1H), 8.23 - 8.36 (m, 2H), 7.82 - 7.92 (m, 2H), 7.75 (s, 1H), 7.34 - 7.41 (m, 2H), 7.32 (s, 1H), 4.02 (s, 3H), 3.98 (br. s., 2H), 3.89 (br. s., 2H), 3.35 - 3.46 (m, 4H); MS (ESI) m/z [M+H]+ calcd. For C21H21N8O4: 449.4; found: 449.1; HPLC purity: 210 nm: 100.0%; 254 nm: 100.0% Step 2. Preparation of (1R)-1-(5-methylpyridin-2-yl)ethyl 4-[7-(1-methyl-1H-pyrazol-4- yl)imidazo[1, 2-b]pyridazin-3-yl]piperazine-1-carboxylate (Compound 364)
Figure imgf000248_0001
[0593] To a solution of (R)-1-(5-methylpyridin-2-yl)ethanol (0.0697 g, 0.508 mmol, as prepared in Example 20, Step 2) in DMF (4 mL) cooled to 0°C was added 60% NaH in mineral oil (0.0203 g, 0.508 mmol) under nitrogen, then the mixture was stirred at 0°C for 20 minutes. 4-Nitrophenyl 4-[7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazin-3- yl]piperazine-1-carboxylate (0.190 g, 0.424 mmol, as prepared in the previous step) was added as a solid and the reaction was stirred at 0°C for 30 minutes, then warmed to rt and stirred overnight. The mixture was diluted with sat. aqueous NaHCO3 (20 mL) and EtOAc (20 mL). The layers were separated and the EtOAc layer was extracted with water (3 x 20 mL) and sat. aqueous NaHCO3 (20 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with 5% MeOH / DCM to afford 74 mg (39%) of (R)-1-(5- methylpyridin-2-yl)ethyl 4-[7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazin-3- yl]piperazine-1-carboxylate (Compound 364) as a yellow foam. 1H NMR (300 MHz, DMSO-d6) δ (ppm) 8.75 - 8.90 (m, 1H), 8.32 - 8.43 (m, 2H), 8.13 - 8.20 (m, 1H), 8.10 (s, 1H), 7.62 (d, J=6.98 Hz, 1H), 7.31 (d, J=6.78 Hz, 1H), 7.29 (s, 1H), 5.71 (q, J=6.61 Hz, 1H), 3.89 (s, 3H), 3.64 (br. s., 4H), 3.19 (br. s., 4H), 2.29 (s, 3H), 1.50 (d, J=6.61 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C23H27N8O2: 447.5; found: 447.2; HPLC purity: 210 nm: 100.0%; 254 nm: 100.0% Example 42: Synthesis of Exemplary Compound 365 (R)-1-(6-methylpyridin-3-yl)ethyl 4-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazin- 3-yl)piperazine-1-carboxylate (Compound 365)
Figure imgf000248_0002
Step 1. Preparation of (R)-1-(6-methylpyridin-3-yl)ethyl (4-nitrophenyl) carbonate
Figure imgf000249_0001
[0594] To a solution of (R)-1-(6-methylpyridin-3-yl)ethanol (600 mg, 4.37 mol, as prepared in Example 36, Step 1) in ACN (6 mL) was added pyridine (0.708 mL, 0.00875 mol) then p-nitrophenyl chloroformate (0.970 g, 0.00481 mol) was added and the reaction was stirred at rt overnight. The mixture was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with 0-50% EtOAc / DCM to afford 900 mg (68%) of (R)-1-(6-methylpyridin-3-yl)ethyl 4-nitrophenyl carbonate as a clear oil. 1H NMR (300 MHz, CDCl3) δ (ppm) 8.59 (d, J=1.86 Hz, 1H), 8.21 - 8.34 (m, 2H), 7.67 (dd, J=7.96, 2.37 Hz, 1H), 7.31 - 7.44 (m, 2H), 7.21 (d, J=8.01 Hz, 1H), 5.86 (q, J=6.61 Hz, 1H), 2.50 - 2.68 (m, 3H), 1.73 (d, J=6.71 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. For C15H15N2O5: 303.3; found: 303.3; HPLC purity: 210 nm: 94.3%; 254 nm: 96.1% Step 2. Preparation of (1R)-1-(6-methylpyridin-3-yl)ethyl 4-[7-(1-methyl-1H-pyrazol-4- yl)imidazo[1, 2-b]pyridazin-3-yl]piperazine-1-carboxylate (Compound 365)
Figure imgf000249_0002
[0595] To a mixture of 7-(1-methyl-1H-pyrazol-4-yl)-3-piperazin-1-ylimidazo[1,2- b]pyridazine hydrochloride salt (Compound S84) (0.150 g, 0.421 mmol) in ACN (2 mL) was added TEA (0.196 mL, 1.40 mmol). The mixture was stirred at rt for 5 minutes then (1R)-1-(6-methylpyridin-3-yl)ethyl 4-nitrophenyl carbonate (0.106 g, 0.351 mmol, as prepared in the previous step) was added and the reaction was stirred at rt for 2 h. The mixture was concentrated under reduced pressure then the residue was dissolved in DCM (20 mL). The mixture was washed with 1N NaOH (2 x 10 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with 5% MeOH / DCM and further purified by silica gel chromatography eluting with acetone to afford 70 mg (45%) of (R)- 1-(6-methylpyridin-3-yl)ethyl 4-[7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazin- 3-yl]piperazine-1-carboxylate (Compound 365) as a yellow glass. 1H NMR (400 MHz, CDCl3) δ (ppm) 8.55 (d, J=2.08 Hz, 1H), 8.49 (d, J=1.96 Hz, 1H), 7.80 - 7.90 (m, 2H), 7.73 (s, 1H), 7.59 (dd, J=7.95, 2.32 Hz, 1H), 7.23 - 7.30 (m, 1H), 7.17 (d, J=8.07 Hz, 1H), 5.88 (q, J=6.60 Hz, 1H), 4.01 (s, 3H), 3.78 (br. s., 4H), 3.27 (br. s., 4H), 2.58 (s, 3H), 1.62 (d, J=6.72 Hz,3 H); MS (ESI) m/z [M+H]+ calcd. For C23H27N8O2: 447.5; found: 447.2; HPLC purity: 210 nm: 100.0%; 254 nm: 100.0%. Example 43: Synthesis of Exemplary Compound 366 4-benzyl-2-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1- yl)thiazole (Compound 366)
Figure imgf000250_0001
Step 1. Preparation of 4-benzyl-2-chlorothiazole
Figure imgf000250_0002
[0596] To a solution of 4-benzyl-1,3-thiazol-2-amine (1 g, 5.25 mmol) and tert-butyl nitrite (1.08 g, 10.51 mmol) in ACN (20 mL) was added CuCl2 (1.06 g, 7.88 mmol) then the reaction was stirredat rt under nitrogen for 4 h. The mixture was concentrated under reduced pressure then the residue was dissolved in water (100 mL). The mixture was extracted with DCM (3 x 100 mL), then the combined organic extracts were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with EtOAc / PE (1:10) to afford 250 mg (23%) of 4-benzyl-2-chlorothiazole as a yellow oil. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 7.36 – 7.18 (m, 6H), 4.01 (s, 2H); MS (ESI) m/z [M+H]+ calcd. for C10H8ClNS, 210.0; found, 210.0. Step 2. 4-benzyl-2-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazin-1-yl)thiazole (Compound 366)
Figure imgf000250_0003
[0597] To a solution of 4-benzyl-2-chloro-1,3-thiazole (222.8 mg, 1.06 mmol, as prepared in the previous step) and 6-(1-methyl-1H-pyrazol-4-yl)-3-piperazin-1-ylpyrazolo[1,5- a]pyridine (Compound 12) (200 mg, 0.70 mmol) in dioxane (20 mL) were added Cs2CO3 (461.5 mg, 1.41 mmol) and Pd-PEPPSI-IPentCl 2-methylpyridine (59.5 mg, 0.07 mmol) under nitrogen then the reaction was heated to 100°C and stirred overnight. The mixture was cooled to rt then concentrated under reduced pressure. The residue was dissolved in water (50 mL) then the mixture was extracted with DCM (3 x 50 mL). The combined organic extracts were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 126.7 mg (39%) of 4-benzyl-2-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazin-1-yl)thiazole as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.84 (s, 1H), 8.24 (s, 1H), 7.98 (s, 1H), 7.80 (s, 1H), 7.69 – 7.67 (m, 1H), 7.38 – 7.32 (m, 1H), 7.32 – 7.31 (m, 1H), 7.30 – 7.26 (m, 3H), 7.24 – 7.18 (m, 1H), 6.41 (s, 1H), 3.88 – 3.84 (m, 5H), 3.56 (t, J = 5.1 Hz, 4H), 3.10 (t, J = 5.1 Hz, 4H). MS (ESI) m/z [M+H]+ calcd. for C25H25N7S, 456.2; found, 455.95.LCMS purity:254 nm:99.7%. Example 44: Synthesis of Exemplary Compound 367 3-benzyl-5-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)- 1,2,4-oxadiazole (Compound 367)
Figure imgf000251_0001
Step 1. Preparation of 3-benzyl-1,2,4-oxadiazol-5(4H)-one
Figure imgf000251_0002
[0598] To a solution of N'-hydroxy-2-phenylethanimidamide (5.00 g, 33.3 mmol) in dioxane (50 mL) was added CDI (8.10 g, 49.9 mmol) and DBU (5.48 mL, 36.6 mmol). The solution was warmed to 105 °C and stirred for 3 h. The reaction was cooled to rt then partitioned between EtOAc and water. The pH of the aqueous layer was adjusted to pH 2 with 1M HCl, then the aqueous layer was extracted with EtOAc (3 times). The combined organic extracts were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with 0-100% (10% MeOH in DCM) / DCM to afford 4.18 g (71%) of 3-benzyl-1,2,4-oxadiazol-5(4H)-one as a white solid. MS (ESI) m/z [M+H]+ calcd. for C9H9N2O2, 177.1; found, 176.9. Step 2. Preparation of 3-[4-(3-benzyl-1,2,4-oxadiazol-5-yl)piperazin-1-yl]-6-(1-methyl- 1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 367)
Figure imgf000252_0001
[0599] To a solution of 3-benzyl-1,2,4-oxadiazol-5(4H)-one (0.15 g, 0.85 mmol, as prepared in the previous step) and PyBroP (0.40 g, 0.85 mmol) in dioxane (5 mL) was added DIEA (0.43 mL, 2.4 mmol). The reaction was heated to 50 °C for 2 h, cooled rt, then 6-(1-methyl-1H-pyrazol-4-yl)-3-piperazin-1-ylpyrazolo[1,5-a]pyridine (Compound 12) (0.12 g, 0.43 mmol) was added. The reaction was stirred at rt overnight then concentrated under reduced pressure. The residue was partitioned between DCM (20 mL) and water (10 mL). The DCM layer was washed with brine, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with 0-100% (10% MeOH in DCM) / DCM. The pure fractions were concentrated under reduced pressure then the residue was dissolved in DCM and extracted with water (x3) and aqueous LiCl (x3), then dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure. The residue was stirred with hexane and concentrated to dryness (x3) to afford 52 mg (28%) of 3-[4-(3-benzyl-1,2,4- oxadiazol-5-yl)piperazin-1-yl]-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 367) as a grey solid.1H NMR (400 MHz, CD3OD) δ (ppm) 8.60 (s, 1H) 8.03 (s, 1H) 7.87 (s, 1H) 7.80 (s, 1H) 7.66 (d, J=9.29 Hz, 1H) 7.37 (dd, J=9.23, 1.41 Hz, 1H) 7.31 (d, J=4.28 Hz, 4H) 7.21 - 7.27 (m, 1H) 3.94 (s, 3H) 3.87 (s, 2H) 3.77 - 3.83 (m, 4H) 3.12 - 3.19 (m, 4H); MS (ESI) m/z [M+H]+ calcd. for C24H25N8O, 441.2; found, 441.2; HPLC purity: 210 nm: 100.0%; 254 nm: 100.0%. Example 45: Synthesis of Exemplary Compound 368 (R)-3-(4-(1-benzyl-1H-1,2,4-triazol-3-yl)-3-methylpiperazin-1-yl)-6-(1-methyl-1H-pyrazol- 4-yl)pyrazolo[1,5-a]pyridine (Compound 368)
Figure imgf000253_0001
[0600] To a solution of (R)-6-(1-methyl-1H-pyrazol-4-yl)-3-(3-methylpiperazin-1- yl)pyrazolo[1,5-a]pyridine hydrochloride salt (300 mg, 0.90 mmol, as prepared in Example 39, Step 1) and 1-benzyl-3-chloro-1H-1,2,4-triazole (174 mg, 0.90 mmol, as prepared in Example 1, Step 1) in dioxane (10 mL) were added Cs2CO3 (1.17 g, 3.61 mmol) and Pd- PEPPSI-IPentCl 2-methylpyridine (76 mg, 0.09 mmol) then the reaction was heated to 90°C and stirred for 16 h under nitrogen. The mixture was cooled to rt and filtered, then the filter cake was washed with EtOAc (2 x 5 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel chromatography eluting with EtOAc and further purified by Prep-HPLC to afford 194.3 mg (47%) of (R)-3-(4-(1-benzyl-1H-1,2,4- triazol-3-yl)-3-methylpiperazin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridine (Compound 368) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.84 – 8.80 (m, 1H), 8.30 – 8.20 (m, 2H), 7.96 (s, 1H), 7.75 (s, 1H), 7.58 (d, J = 9.2 Hz, 1H), 7.41 – 7.23 (m, 6H), 5.23 (s, 2H), 4.26 – 4.12 (m, 1H), 3.87 (s, 3H), 3.74 – 3.64 (m, 1H), 3.31 – 3.22 (m, 2H), 3.18 – 3.10 (m, 1H), 2.92 (dd, J = 11.3, 3.6 Hz, 1H), 2.85 – 2.75 (m, 1H), 1.29 (d, J = 6.5 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C25H27N9, 454.2; found, 454.2; LCMS purity: 254 nm: 99.8% Example 46: Synthesis of Exemplary Compound 369 (S)-3-(4-(1-benzyl-1H-1,2,4-triazol-3-yl)-3-methylpiperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine (Compound 369)
Figure imgf000253_0002
[0601] To a solution of (S)-6-(1-methyl-1H-pyrazol-4-yl)-3-(3-methylpiperazin-1- yl)pyrazolo[1,5-a]pyridine hydrochloride salt (300 mg, 1.01 mmol, as prepared in Example 40, Step 1) and 1-benzyl-3-chloro-1H-1,2,4-triazole (215.6 mg, 1.11 mmol, as prepared in Example 1, Step 1) in dioxane (20 mL) were added Cs2CO3 (1.32 g, 4.04 mmol) and Pd- PEPPSI-IPentCl 2-methylpyridine (85.1 mg, 0.10 mmol) then the reaction was heated to 90°C and stirred overnight under nitrogen. The mixture was cooled to rt then concentrated under reduced pressure. The residue was dissolved in water (50 mL). The resulting mixture was extracted with DCM (3 x 50 mL) then the combined organic extracts were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 145.9 mg (32%) of (S)-3-(4-(1-benzyl- 1H-1,2,4-triazol-3-yl)-3-methylpiperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine (Compound 369) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.29 (s, 1H), 8.22 (s, 1H), 7.96 (s, 1H), 7.75 (s, 1H), 7.60 – 7.57 (m,1H), 7.45 – 7.18 (m, 6H), 5.23 (s, 2H), 4.27 – 4.13 (m, 1H), 3.87 (s, 3H), 3.74 – 3.62 (m, 1H), 3.31 – 3.21 (m, 2H), 3.16 – 3.13 (m, 1H), 2.93 – 2.91 (m, 1H), 2.80 – 2.79 (m, 1H), 1.29 (d, J = 6.5 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C25H27N9, 454.2; found, 453.90.LCMS purity:254 nm:99.9%; ANAL_SFC purity: 97.9%. Example 47: Synthesis of Exemplary Compound 370 (R)-1-(4-(trifluoromethyl)phenyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrimidin-3-yl)piperazine-1-carboxylate (Compound 370)
Figure imgf000254_0001
[0602] To a solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)pyrazolo[1,5- a]pyrimidine (Compound S98) (150 mg, 0.53 mmol) and ((R)-1-(4- (trifluoromethyl)phenyl)ethan-1-ol (120.8 mg, 0.63 mmol) in pyridine (5 mL) was added triphosgene (63.5 mg, 0.21 mmol) then the reaction was stirred at rt under nitrogen for 16 h. The mixture was concentrated under reduced pressure. The residue was treated with water (30 mL), then the mixture was extracted with EtOAc (3 x 50 mL). The combined organic extracts were washed with brine (10 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 54.0 mg (20%) of (R)-1-(4-(trifluoromethyl)phenyl)ethyl 4-(6-(1- methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin-3-yl)piperazine-1-carboxylate (Compound 370) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.19 (d, J = 2.1 Hz, 1H), 8.65 (d, J = 2.1 Hz, 1H), 8.30 (s, 1H), 8.04 (s, 1H), 7.91 (s, 1H), 7.74 (d, J = 8.2 Hz, 2H), 7.61 (d, J = 8.1 Hz, 2H), 5.82 (q, J = 6.5 Hz, 1H), 3.89 (s, 3H), 3.80 - 3.45 (m, 4H), 3.33 (s, 4H), 1.51 (d, J = 6.6 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C24H24F3N7O2, 500.2; found, 500.0; LCMS purity: 254 nm: 99.7% Example 48: Synthesis of Exemplary Compound 371 (R)-1-(4-chlorophenyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin-3- yl)piperazine-1-carboxylate (Compound 371)
Figure imgf000255_0001
[0603] To a solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)pyrazolo[1,5- a]pyrimidine (Compound S98) (250 mg, 0.88 mmol) and (R)-1-(4-chlorophenyl)ethanol (207.3 mg, 1.32 mmol) in pyridine (5 mL) cooled to 0°C was added triphosgene (105.8 mg, 0.35 mmol) then the reaction was stirred at 0°C under nitrogen for 16 h. The mixture was concentrated under reduced pressure then the residue was treated with water (30 mL). The mixture was extracted with EtOAc (3 x 50 mL) then the combined organic extracts were washed with brine (10 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 101.1 mg (24%) of (R)-1-(4-chlorophenyl)ethyl 4-(6-(1-methyl-4,5-dihydro-1H-pyrazol- 4-yl)pyrazolo[1,5-a]pyrimidin-3-yl)piperazine-1-carboxylate (Compound 371) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.19 (d, J = 2.1 Hz, 1H), 8.64 (d, J = 2.1 Hz, 1H), 8.30 (s, 1H), 8.03 (d, J = 0.9 Hz, 1H), 7.90 (s, 1H), 7.47 – 7.37 (m, 4H), 5.73 (q, J = 6.5 Hz, 1H), 3.89 (s, 3H), 3.75 – 3.45 (m, 4H), 3.22 (s, 3H), 1.47 (d, J = 6.6 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C23H24ClN7O2, 466.2; found, 466.2; LCMS purity: 254 nm: 99.3% Example 49: Synthesis of Exemplary Compound 372 3-(4-(1-(4-fluorobenzyl)-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine (Compound 372)
Figure imgf000256_0001
Step 1. Preparation of 3-chloro-1-(4-fluorobenzyl)-1H-1,2,4-triazole
Figure imgf000256_0002
[0604] To a solution of 3-chloro-1H-1,2,4-triazole (3.00 g, 28.9 mmol) dissolved in DMF (60 mL) was added 1-(bromomethyl)-4-fluorobenzene (6.57 g, 34.8 mmol) and K2CO3 (8.01 g, 58.0 mmol) then the reaction was heated to 80°C and stirred for 16 h. The mixture was cooled to rt and filtered, then the filter cake was washed with DMF (2 x 1 mL). The filtrate was concentrated under reduced pressure then the residue was purified by Prep- HPLC to afford 4.5 g (58.7%) of 3-chloro-1-(4-fluorobenzyl)-1H-1,2,4-triazole as a white solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.73 (s, 1H) 7.42 – 7.37 (m, 2H) 7.25 – 7.19 (m, 2H) 5.38 (s, 2H); MS (ESI) m/z [M+H]+ calcd. for C9H7ClFN3, 212.0; found, 212.0. Step 2. 3-(4-(1-(4-fluorobenzyl)-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 372)
Figure imgf000256_0003
[0605] To a solution of 3-chloro-1-(4-fluorobenzyl)-1H-1,2,4-triazole (300 mg, 1.42 mmol, as prepared in the previous step) dissolved in dioxane (8 mL) was added 6-(1- methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 12) (400 mg, 1.42 mmol), Cs2CO3 (924 mg, 2.84 mmol), and Pd-PEPPSI-IPentCl 2-methylpyridine (119 mg, 0.142 mmol), then the reaction under nitrogen was heated to 100°C and stirred for 16 h at 100°C. The mixture was cooled to rt and concentrated under reduced pressure. The residue was purified by Prep-Achiral-SFC to afford 161.6 mg (25%) of 3-(4-(1-(4- fluorobenzyl)-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine (Compound 372) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (t, J = 1.3 Hz, 1H) 8.31 (s, 1H) 8.23 (s, 1H) 7.97 (s, 1H) 7.77 (s, 1H) 7.66 (d, J = 9.3 Hz, 1H) 7.39 – 7.28 (m, 3H) 7.25 – 7.16 (m, 2H) 5.23 (s, 2H) 3.87 (s, 3H) 3.45 (t, J = 4.9 Hz, 4H) 3.06 (t, J = 5.0 Hz, 4H); MS (ESI) m/z [M+H]+ calcd. for C24H24FN9, 458.2; found, 458.5; LCMS purity: 254 nm: 99.7% Example 50: Synthesis of Exemplary Compound 373 3-(4-(2-benzyl-2H-tetrazol-5-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridine (Compound 373)
Figure imgf000257_0001
Step 1. Preparation of 2-benzyl-5-bromo-2H-tetrazole
Figure imgf000257_0002
[0606] To a solution of 5-bromo-2H-1,2,3,4-tetrazole (100 mg, 0.67 mmol) and K2CO3 (92.8 mg, 0.67 mmol) in ACN (10 mL) was added benzyl bromide (114.8 mg, 0.67 mmol) at rt then the reaction was heated to 80°C and stirred for 3 h. The mixture was cooled to rt, filtered, and the filtrate was concentrated under reduced pressure. The residue was purified by Prep-TLC (EtOAc / PE 1:5) to afford 45 mg (28%) of 2-benzyl-5-bromo-2H-tetrazole as a colorless oil. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 7.48 – 7.32 (m, 5H), 5.98 (s, 2H); MS (ESI) m/z [M+H] + calcd. for C8H7BrN4, 239.0; found, 239.1. Step 2. Preparation of 3-(4-(2-benzyl-2H-tetrazol-5-yl)piperazin-1-yl)-6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 373)
Figure imgf000257_0003
[0607] To a solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-piperazin-1-ylpyrazolo[1,5- a]pyridine (Compound 12) (413.3 mg, 1.46 mmol), Cs2CO3 (954 mg, 2.93 mmol), and 2- benzyl-5-bromo-2H-tetrazole (350 mg, 1.46 mmol, as prepared in the previous step) in dioxane (15 mL) was added Pd-PEPPSI-IPentCl 2-methylpyridine (123.1 mg, 0.15 mmol) under nitrogen, then the reaction was heated to 90°C and stirred for 16 h. The mixture was cooled to rt, concentrated under reduced pressure, and the residue was purified by Prep- TLC (MeOH / DCM 1:10), then the crude product was re-crystallized from THF/ACN (1:5) (5 mL) to afford 164.5 mg (26%) of 3-(4-(2-benzyl-2H-tetrazol-5-yl)piperazin-1-yl)-6-(1- methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 373) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (t, J = 1.3 Hz, 1H), 8.23 (s, 1H), 7.98 (d, J = 0.8 Hz, 1H), 7.79 (s, 1H), 7.67 (dd, J = 9.2, 0.9 Hz, 1H), 7.46 – 7.30 (m, 6H), 5.75 (s, 2H), 3.87 (s, 3H), 3.56 (t, J = 5.0 Hz, 4H), 3.14 – 3.18 (m, 4H); MS (ESI) m/z [M+H]+ calcd. for C23H24N10, 441.2; found, 441.3; LCMS purity: 254 nm: 98.6%. Example 51: Synthesis of Exemplary Compound 374 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(5-((5-methylpyridin-2-yl)methyl)pyrimidin-2- yl)piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 374)
Figure imgf000258_0001
Step 1. Preparation of potassium 2-(5-methylpyridin-2-yl)acetate
Figure imgf000258_0002
[0608] To a solution of methyl 2-(5-methylpyridin-2-yl)acetate (2.00 g, 12.11 mmol) dissolved in EtOH (20 mL) was added H2O (0.23 g, 12.71 mmol). The resulting mixture was stirred for 10 minutes at 60°C under nitrogen. A solution of KOtBu (1.43 g, 12.71 mmol) was prepared in absolute EtOH (10 mL) in dried glassware, and then added dropwise over 10 minutes to the initial solution at 60°C. After complete conversion as monitored by HPLC, the mixture was concentrated under reduced pressure and the residue was stirred in anhydrous MTBE (10 mL) for 30 minutes. The solids were isolated by filtration and washed with EtOH/MTBE (1:1) (2 x 10 mL) and MTBE (3 x10 mL). The solids were dried under reduced pressure to afford 1.85 g (77%) of potassium 2-(5-methylpyridin-2-yl)acetate as a white solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.17 (s, 1H), 7.39 (d, J = 8.0 Hz, 1H), 7.17 (d, J = 8.0 Hz, 1H), 3.29 (s, 2H), 2.21 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C8H8KNO2, 190.0; found, 152.1. Step 2. Preparation of 2-chloro-5-((5-methylpyridin-2-yl)methyl)pyrimidine
Figure imgf000259_0001
[0609] To a solution of potassium 2-(5-methylpyridin-2-yl)acetate (1.3 g, 6.87 mmol, as prepared in the previous step) dissolved in mesitylene (50 mL) was added 5-bromo-2- chloropyrimidine (1.06 g, 5.50 mmol), Pd2(dba)3 (0.09 g, 0.10 mmol), and Xantphos (0.24 g, 0.41 mmol) under nitrogen, then the reaction was heated to 135°C and stirred for 24 h. The mixture was cooled to rt, diluted with water (50 mL), and extracted with EtOAc (3 x 50 mL). The combined organic extracts were washed with brine (3 x 30 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with EtOAc / PE (1:3) to afford 1.0 g (53%) of 2-chloro-5-((5-methylpyridin-2-yl)methyl)pyrimidine as an orange solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.70 (s, 2H), 8.30 (d, J = 2.4 Hz, 1H), 7.54 (dt, J = 7.9, 1.5 Hz, 1H), 7.27 (d, J = 7.9 Hz, 1H), 4.10 (s, 2H), 2.23 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C11H10ClN3, 220.1; found, 220.1. Step 3. Preparation of 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(5-((5-methylpyridin-2- yl)methyl)pyrimidin-2-yl)piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 374)
Figure imgf000259_0002
[0610] To a solution of 2-chloro-5-((5-methylpyridin-2-yl)methyl)pyrimidine (400 mg, 1.82 mmol, as prepared in the previous step) dissolved in NMP (10 mL) was added 6-(1- methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 12) (411.3 mg, 1.46 mmol) and K2CO3 (755 mg, 5.46 mmol) under nitrogen, then the reaction was heated to 110°C and stirred for 16 h. The mixture was cooled to rt, diluted with water (50 mL), and extracted with DCM (3 x 50 mL). The combined organic extracts were washed with brine (3 x 50 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 120.6 mg (14%) of 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(5-((5-methylpyridin-2- yl)methyl)pyrimidin-2-yl)piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 374) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.34 – 8.29 (m, 3H), 8.23 (s, 1H), 7.97 (d, J = 0.9 Hz, 1H), 7.77 (s, 1H), 7.68 (dd, J = 9.3, 1.0 Hz, 1H), 7.53 (dd, J = 8.1, 2.4 Hz, 1H), 7.32 (dd, J = 9.2, 1.6 Hz, 1H), 7.20 (d, J = 8.0 Hz, 1H), 3.92 – 3.84 (m, 9H), 3.03 (t, J = 5.0 Hz, 4H), 2.25 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C26H27N9, 466.2; found, 466.2; LCMS purity: 254 nm: 99.0%. Example 52: Synthesis of Exemplary Compound 375 3-(4-(1-benzyl-1H-pyrazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridine (Compound 375)
Figure imgf000260_0001
Step 1. Preparation of 1-benzyl-3-bromo-1H-pyrazole
Figure imgf000260_0002
[0611] To a solution of 3-bromo-1H-pyrazole (3 g, 20.41 mmol) and K2CO3 (4.23 g, 30.62 mmol) in ACN (50 mL) cooled to 0°C was added benzyl bromide (4.19 g, 24.49 mmol), then the reaction was heated to 80°C and stirred for 5 h. The mixture was cooled to rt, the precipitated solids were collected by filtration and washed with DCM (3 x 60 mL). The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with EtOAc / PE (1:5) to afford 3.2 g (66%) of 1-benzyl-3-bromo- 1H-pyrazole as a white solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 7.88 (d, J = 2.3 Hz, 1H), 7.41 – 7.19 (m, 5H), 6.40 (d, J = 2.3 Hz, 1H), 5.30 (s, 2H); MS (ESI) m/z [M+H]+ calcd. for C10H9BrN2, 237.0; found, 237.1. Step 2. Preparation of 3-(4-(1-benzyl-1H-pyrazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 375)
Figure imgf000261_0001
[0612] To a solution of 1-benzyl-3-bromo-1H-pyrazole (1 g, 4.22 mmol, as prepared in the previous step) and 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)pyrazolo[1,5- a]pyridine (Compound 12) (1.19 g, 4.22 mmol) in dioxane (30 mL) under nitrogen were added Pd-PEPPSI-IPentCl 2-methylpyridine (354.77 mg, 0.42 mmol) and K3PO4 (1.79 g, 8.44 mmol) then the reaction was heated to 100°C and stirred for 16 h. The mixture was cooled to rt, concentrated under reduced pressure, and the residue was dissolved in water. The aqueous mixture was extracted with DCM (5 x 80 mL), then the combined organic extracts were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with MeOH / DCM (1:15) and Prep-HPLC to afford 158.2 mg (8%) of 3-(4-(1-benzyl-1H- pyrazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 375) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.77 (s, 1H), 7.67 – 7.57 (m, 2H), 7.38 – 7.16 (m, 6H), 5.80 (d, J = 2.3 Hz, 1H), 5.15 (s, 2H), 3.87 (s, 3H), 3.26 (t, J = 4.8 Hz, 4H), 3.08 (t, J = 4.8 Hz, 4H); MS (ESI) m/z [M+H]+ calcd. for C25H26N8, 439.2; found, 439.15; LCMS purity: 254 nm: 99.3%. Example 53: Synthesis of Exemplary Compound 376 3-(4-(1-(4-fluorobenzyl)-1H-1,2,4-triazol-5-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine (Compound 376)
Figure imgf000261_0002
Step 1. Preparation of 5-chloro-1-(4-fluorobenzyl)-1H-1,2,4-triazole
Figure imgf000262_0001
[0613] To a solution of 3-chloro-1H-1,2,4-triazole (3 g, 28.9 mmol) dissolved in DMF (60 mL) was added 1-(bromomethyl)-4-fluorobenzene (6.57 g, 34.8 mmol) and K2CO3 (8.01 g, 58.0 mmol) then the reaction was heated to 80°C and stirred for 16 h. The mixture was cooled to rt, filtered, and the filter cake was washed with DMF (2 x 5 mL). The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 180 mg (0.3%) of 5-chloro-1-(4-fluorobenzyl)-1H-1,2,4-triazole as a white solid. MS (ESI) m/z [M+H]+ calcd. for C9H7ClFN3, 212.0; found, 212.0; LCMS purity: 254 nm: 70%. Step 2. Preparation of 3-(4-(1-(4-fluorobenzyl)-1H-1,2,4-triazol-5-yl)piperazin-1-yl)-6-(1- methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 376)
Figure imgf000262_0002
[0614] To a solution of 5-chloro-1-(4-fluorobenzyl)-1H-1,2,4-triazole (20 mg, 0.095 mmol, as prepared in the previous step) in dioxane (2 mL) under nitrogen was added 6-(1- methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 12) (26.7 mg, 0.095 mmol), Cs2CO3 (61.6 mg, 0.19 mmol), and Pd-PEPPSI-IPentCl 2-methylpyridine (7.95 mg, 0.01 mmol), then the reaction was heated to 100°C and stirred for 16 h under nitrogen. The mixture was cooled to rt and concentrated under reduced pressure. The residue was purified by Prep-Achiral-SFC to afford 7.2 mg (17%) of 3-(4-(1-(4- fluorobenzyl)-1H-1,2,4-triazol-5-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine (Compound 376) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (t, J = 1.2 Hz, 1H) 8.23 (s, 1H) 7.98 (d, J = 0.9 Hz, 1H) 7.77 (d, J = 7.3 Hz, 2H) 7.68 (dd, J = 9.3, 1.0 Hz, 1H) 7.36 – 7.25 (m, 3H) 7.24 – 7.17 (m, 2H) 5.27 (s, 2H) 3.87 (s, 3H) 3.27 – 3.22 (m, 4H) 3.15 – 3.09 (m, 4H); MS (ESI) m/z [M+H]+ calcd. for C24H24FN9, 458.2; found, 458.2; LCMS purity: 254 nm: 99.5%. Example 54: Synthesis of Exemplary Compound 377 3-(4-(1-benzyl-1H-pyrazol-5-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridine (Compound 377)
Figure imgf000263_0001
Step 1. Preparation of 1-benzyl-5-bromo-1H-pyrazole
Figure imgf000263_0003
[0615] To a stirred solution of 3-bromo-1H-pyrazole (3 g, 20.41 mmol) and K2CO3 (4.23 g, 30.62 mmol) in ACN (60 mL) cooled to 0°C was added benzyl bromide (4.19 g, 24.49 mmol) then the reaction was heated to 80°C and stirred for 16 h. The mixture was cooled to rt then the precipitated solids were collected by filtration and washed with DCM (3 x 60 mL). The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with EtOAc / PE (1:5) to afford 800 mg (17%) of 1- benzyl-5-bromo-1H-pyrazole as a white solid. MS: (ESI) m/z [M+H]+ calcd. for C10H9BrN2, 237.0; found, 237.1. Step 2. Preparation of 3-(4-(1-benzyl-1H-pyrazol-5-yl)piperazin-1-yl)-6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 377)
Figure imgf000263_0002
[0616] To a solution of 1-benzyl-5-bromo-1H-pyrazole (300 mg, 1.26 mmol, as prepared in the previous step) and 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)pyrazolo[1,5- a]pyridine (Compound 12) (357.3 mg, 1.26 mmol) in dioxane (5 mL) under nitrogen were added Pd-PEPPSI-IPentCl 2-methylpyridine (106.4 mg, 0.13 mmol) and K3PO4 (537.2 mg, 2.53 mmol), then the reaction was heated to 100°C and stirred for 16 h under nitrogen. The mixture was cooled to rt, concentrated under reduced pressure, and the residue was dissolved in water. The aqueous mixture was extracted with DCM (3 x 80 mL) then the combined organic extracts were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure, then the residue was purified by Prep-TLC (MeOH / DCM 1:15) and Prep-HPLC to afford 27.1 mg (5%) of 3-(4-(1-benzyl-1H-pyrazol-5- yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 377) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (s, 1H), 8.23 (s, 1H), 7.98 (d, J = 0.6 Hz, 1H), 7.79 (s, 1H), 7.65 (dd, J = 0.6 Hz, 9.2 Hz 1H), 7.42 (d, J = 1.9 Hz, 1H), 7.38 – 7.15 (m, 6H), 6.07 (d, J = 1.9 Hz, 1H), 5.25 (s, 2H), 3.87 (s, 3H), 3.12 (t, J = 4.6 Hz, 4H), 2.98 (t, J = 4.6 Hz, 4H); MS (ESI) m/z [M+H]+ calcd. for C25H26N8, 439.2; found, 439.15; LCMS purity: 254 nm: 99.6%. Example 55: Synthesis of Exemplary Compound 378 2-benzyl-5-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin-3-yl)piperazin-1-yl)- 1,3,4-thiadiazole (Compound 378)
Figure imgf000264_0001
Step 1. Preparation of 5-benzyl-1,3,4-thiadiazol-2-amine
Figure imgf000264_0002
[0617] To a solution of 2-phenylacetic acid (1.3 g, 9.55 mmol) in POCl3 (5.86 g, 38.19 mmol) was added hydrazinecarbothioamide (1.31 g, 14.32 mmol) then the reaction was heated to 80°C and stirred for 4 h under nitrogen. The mixture was cooled in an ice bath, neutralized to pH 8 with 50% NaOH solution, and filtered. The filter cake was washed with EtOH (3 x 20 mL) then the filtrate was concentrated under reduced pressure. The residue was purified by silica gel chromatography eluting with EtOAc / PE (2:1) to afford 1.1 g (60%) of 5-benzyl-1,3,4-thiadiazol-2-amine as a white solid.1H NMR (400 MHz, DMSO- d6) δ (ppm) 7.37 – 7.20 (m, 5H), 7.03 (s, 2H), 4.14 (s, 2H); MS (ESI) m/z [M+H]+ calcd. for C9H9N3S, 192.1; found, 192.1. Step 2. Preparation of 2-benzyl-5-bromo-1,3,4-thiadiazole
Figure imgf000265_0001
[0618] To a solution of 5-benzyl-1,3,4-thiadiazol-2-amine (500 mg, 2.61 mmol, as prepared in the previous step) in ACN (20 mL) was added CuBr2 (700.7 mg, 3.137 mmol) and tert-butyl nitrite (404.4 mg, 3.92 mmol) then the reaction was stirred at rt for 16 h under nitrogen. The reaction was cooled to 0°C then quenched by the addition of sat. aqueous NH4Cl (30 mL). The resulting mixture was extracted with EtOAc (3 x 30 mL). The combined organic extracts were washed with brine (3 x 10 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with EtOAc / PE (1:2) to afford 400 mg (59%) of 2-benzyl-5-bromo-1,3,4-thiadiazole as a light-yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 7.40 – 7.24 (m, 5H), 4.49 (s, 2H); MS (ESI) m/z [M+H]+ calcd. for C9H7BrN2S, 255.0; found, 255.0. Step 3. Preparation of 2-benzyl-5-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrimidin-3-yl)piperazin-1-yl)-1,3,4-thiadiazole (Compound 378)
Figure imgf000265_0002
[0619] To a solution of 2-benzyl-5-bromo-1,3,4-thiadiazole (200 mg, 0.78 mmol, as prepared in the previous step) in DMSO (5 mL) was added 6-(1-methyl-1H-pyrazol-4-yl)- 3-(piperazin-1-yl)pyrazolo[1,5-a]pyrimidine (Compound S98) (222.1 mg, 0.78 mmol) and DIEA (202.6 mg, 1.57 mmol), then the reaction was heated to 110°C and stirred for 16 h under nitrogen. The mixture was cooled to rt, diluted with water (30 mL), and extracted with EtOAc (3 x 30 mL). The combined organic extracts were washed with brine (3x30 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 27.5 mg (7%) of 2- benzyl-5-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin-3-yl)piperazin-1-yl)- 1,3,4-thiadiazole (Compound 378) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.20 (d, J = 2.1 Hz, 1H), 8.65 (d, J = 2.1 Hz, 1H), 8.30 (s, 1H), 8.04 (d, J = 0.8 Hz, 1H), 7.93 (s, 1H), 7.40 – 7.23 (m, 5H), 4.25 (s, 2H), 3.89 (s, 3H), 3.59 (dd, J = 6.6, 3.8 Hz, 4H), 3.39 – 3.34 (m, 4H); MS (ESI) m/z [M+H]+ calcd. for C23H23N9S, 458.1; found, 458.1; LCMS purity: 254 nm: 98.9%. Example 56: Synthesis of Exemplary Compound 379 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(pyridin-2-ylmethyl)-1H-1,2,4-triazol-3-yl)piperazin- 1-yl)pyrazolo[1,5-a]pyridine (Compound 379)
Figure imgf000266_0001
Step 1. Preparation of 2-((3-chloro-1H-1,2,4-triazol-1-yl)methyl)pyridine
Figure imgf000266_0002
[0620] To a solution of 2-(bromomethyl)pyridine hydrobromide (8.8 g, 34.8 mmol) in DMF (30 mL) was added 3-chloro-1H-1,2,4-triazole (3.00 g, 29.0 mmol) and K2CO3 (8.01 g, 58.0 mmol) then the reaction was heated to 80°C and stirred for 16 h under nitrogen. The mixture was cooled to rt and concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with a C18 silica gel column eluting with 10-50% ACN / water (10mM NH4HCO3) to afford 5.4 g (48%) of 2-((3-chloro-1H-1,2,4-triazol-1- yl)methyl)pyridine as a brown liquid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.76 (s, 1H), 8.56 – 8.45 (m, 1H), 7.86 – 7.78 (m, 1H), 7.38 – 7.27 (m, 2H), 5.55 – 5.50 (m, 2H); MS (ESI) m/z [M+H]+ calcd. for C8H7ClN4, 195.0; found, 195.0. Step 2. Preparation of 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(pyridin-2-ylmethyl)-1H- 1,2,4-triazol-3-yl)piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 379)
Figure imgf000266_0003
[0621] To a solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-piperazin-1-ylpyrazolo[1,5- a]pyridine (Compound 12) (300 mg, 1.06 mmol) and 2-((3-chloro-1H-1,2,4-triazol-1- yl)methyl)pyridine (248 mg, 1.28 mmol, as prepared in the previous step) in dioxane (10 mL) under nitrogen was added Cs2CO3 (692 mg, 2.13 mmol) and Pd-PEPPSI-IPentCl 2- methylpyridine (90 mg, 0.11 mmol) then the reaction was heated to 90°C and stirred for 16 h under nitrogen. The mixture was cooled to rt, filtered, and the filter cake was washed with EtOAc (2 x 5 mL). The filtrate was concentrated under reduced pressure. The residue was purified by silica gel chromatography eluting with MeOH / DCM (1:25) and Prep-Achiral- SFC to afford 120.9 mg (26%) of 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(pyridin-2- ylmethyl)-1H-1,2,4-triazol-3-yl)piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 379) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.81 (s, 1H), 8.57 – 8.51 (m, 1H), 8.33 (s, 1H), 8.22 (s, 1H), 7.96 (s, 1H), 7.84 – 7.77 (m, 1H), 7.76 (s, 1H), 7.68 – 7.61 (m, 1H), 7.37 – 7.27 (m, 2H), 7.25 – 7.18 (m, 1H), 5.33 (s, 2H), 3.86 (s, 3H), 3.48 – 3.41 (m, 4H), 3.09 – 3.02 (m, 4H); MS (ESI) m/z [M+H]+ calcd. for C23H24N10, 441.2; found, 441.1; LCMS purity: 254 nm: 99.0% Example 57: Synthesis of Exemplary Compound 380 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-((5-methylpyridin-2-yl)methyl)-1H-1,2,4-triazol-3- yl)piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 380)
Figure imgf000267_0001
Step 1. Preparation of 2-(bromomethyl)-5-methylpyridine
Figure imgf000267_0002
[0622] To a solution of (5-methylpyridin-2-yl)methanol (300 mg, 2.43 mmol) in DCM (6 mL) was added POBr3 (698.3 mg, 2.43 mmol) then the reaction was stirred at rt for 30 minutes under nitrogen. Step 2. 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-((5-methylpyridin-2-yl)methyl)-1H-1,2,4- triazol-3-yl)piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 380).
Figure imgf000268_0001
[0623] To a solution of 3-(4-(1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound S102) (120 mg, 0.34 mmol) in DMF (10 mL) were added K2CO3 (189.8 mg, 1.37 mmol) and 2-(bromomethyl)-5-methylpyridine (127.8 mg, 0.68 mmol, as prepared in the previous step) then the reaction was heated to 90°C and stirred overnight under nitrogen. The mixture was cooled to rt and concentrated under reduced pressure. The residue was dissolved in water (50 mL) and the aqueous mixture was extracted with DCM (3 x 50 mL). The combined organic extracts were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 15.1 mg (10%) of 6-(1-methyl-1H- pyrazol-4-yl)-3-(4-(1-((5-methylpyridin-2-yl)methyl)-1H-1,2,4-triazol-3-yl)piperazin-1- yl)pyrazolo[1,5-a]pyridine (Compound 380) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.81 (s, 1H), 8.37 (s, 1H), 8.31 (s, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.76 (s, 1H), 7.66-7.60 (m,2H), 7.32-7.30 (m,1H), 7.15-7.13 (m,1H), 5.27 (s, 2H), 3.86 (s, 3H), 3.44 (t, J = 5.0 Hz, 4H), 3.05 (t, J = 5.0 Hz, 4H), 2.28 (s, 3H). MS (ESI) m/z [M+H]+ calcd. for C24H26N10, 455.2; found, 455.15.LCMS purity:254 nm:99.2%. Example 58: Synthesis of Exemplary Compound 381 3-(4-(1-(4-fluorobenzyl)-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyrimidine (Compound 381)
Figure imgf000268_0002
[0624] To a solution of 3-chloro-1-(4-fluorobenzyl)-1H-1,2,4-triazole (194.1 mg, 0.91 mmol, as prepared in Example 49, Step 1) and 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin- 1-yl)pyrazolo[1,5-a]pyrimidine (Compound S98) (200 mg, 0.706 mmol) in dioxane (20 mL) under nitrogen were added Cs2CO3 (460.5 mg, 1.41 mmol) and Pd-PEPPSI-IPentCl 2- methylpyridine (59.3 mg, 0.07 mmol) then the reaction was heated to 100°C and stirred overnight under nitrogen. The mixture was cooled to rt and concentrated under reduced pressure. The residue was dissolved in water (50 mL) then the aqueous mixture was extracted with DCM (3 x 50 mL). The combined organic extracts were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 12.9 mg (4%) of 3-(4-(1-(4-fluorobenzyl)- 1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrimidine (Compound 381) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.18 (d, J = 2.1 Hz, 1H), 8.63 (d, J = 2.1 Hz, 1H), 8.31-8.28 (m, 2H), 8.03 (s, 1H), 7.90 (s, 1H), 7.44 – 7.26 (m, 2H), 7.26 – 7.12 (m, 2H), 5.22 (s, 2H), 3.89 (s, 3H), 3.47-3.42 (m, 4H), 3.32-3.29 (m, 4H); MS (ESI) m/z [M+H]+ calcd. for C23H23FN10, 459.2; found, 459.20. LCMS purity:254 nm:99.6%. Example 59: Synthesis of Exemplary Compound 382 3-(4-(1-benzyl-5-methyl-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine (Compound 382)
Figure imgf000269_0001
[0625] To a stirred solution of 1-benzyl-3-chloro-5-methyl-1H-1,2,4-triazole (150 mg, 0.72 mmol) and 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 12) (204 mg, 0.72 mmol) in dioxane (5 mL) under nitrogen were added Pd- PEPPSI-IPentCl 2-methylpyridine (60.8 mg, 0.072 mmol) and Cs2CO3 (470.7 mg, 1.44 mmol) then the reaction was heated to 90°C and stirred for 16 h under nitrogen. The mixture was cooled to rt, diluted with water, and extracted with DCM (3x60 mL). The combined organic extracts were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with MeOH / DCM (1:15) and Prep-HPLC to afford 166.7 mg (51%) of 3-(4-(1-benzyl-5-methyl-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 382) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.22 (s, 1H), 7.97 (d, J = 0.5 Hz, 1H), 7.76 (s, 1H), 7.65 (dd, J = 0.5Hz, 9.2 Hz, 1H), 7.45 – 7.15 (m, 6H), 5.19 (s, 2H), 3.87 (s, 3H), 3.43 (t, J = 4.8 Hz, 4H), 3.05 (t, J = 4.8 Hz, 4H), 2.32 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C25H27N9, 454.2; found, 454.10; LCMS purity: 254 nm: 99.7%. Example 60: Synthesis of Exemplary Compound 383 3-(3-(1-benzyl-1H-1,2,4-triazol-3-yl)pyrrolidin-1-yl)-7-(1-methyl-1H-pyrazol-4- yl)imidazo[1,2-b]pyridazine (Compound 383)
Figure imgf000270_0001
Step 1. Preparation of 3-iodo-7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazine
Figure imgf000270_0002
[0626] To a round bottom flask containing 7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2- b]pyridazine (1.00 g, 5.02 mmol, as prepared for Compound S3, Step 4) was added DMF (10 mL) then the solution was cooled to 0°C and NIS (1.36 g, 6.02 mmol) was added at a rate to keep the temperature <10°C. The reaction was warmed to rt and stirred for 75 minutes then the mixture was added to a stirred solution of Na2S2O3 (1.98 g, 12.5 mmol) and NaHCO3 (1.05 g, 12.5 mmol) in water (10 mL). The reaction flask was rinsed with THF (2 x 1.5 mL) and the slurry was stirred vigorously for 1 h. The mixture was filtered and the solids were washed with water (3 x 5.5 mL) and dried to afford 1.54 g (94%) of 3- iodo-7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazine as a white solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.97 (d, J = 2.1 Hz, 1H), 8.43 (s, 1H), 8.27 (d, J = 2.1 Hz, 1H), 8.16 (s, 1H), 7.83 (s, 1H), 3.90 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C10H9IN5, 326.1; found, 325.8; HPLC purity: 210 nm: 100.0%; 254 nm: 100.0%. Step 2. Preparation of 3-(3-(1-benzyl-1H-1,2,4-triazol-3-yl)pyrrolidin-1-yl)-7-(1-methyl- 1H-pyrazol-4-yl)imidazo[1,2-b]pyridazine (Compound 383)
Figure imgf000271_0001
[0627] To a round bottom flask containing 3-iodo-7-(1-methyl-1H-pyrazol-4- yl)imidazo[1,2-b]pyridazine (1.45 g, 4.46 mmol, as prepared in the previous step), 1- benzyl-3-pyrrolidin-3-yl-1H-1,2,4-triazole (Compound S101) (750 mg, 3.30 mmol), K3PO4 (2.09 g, 9.86 mmol), and CuI (256 mg, 1.35 mmol) (pre-sparged with argon for 20 minutes) was added 1-butanol (19 mL) and 1,2-ethanediol (3.8 mL) under nitrogen, then the reaction was heated to 100°C and stirred for 60 h under nitrogen. The mixture was cooled to rt and diluted with MeOH (100 mL). After stirring rapidly for 20 minutes, the mixture was filtered through a bed of Celite, and the filtrate was concentrated under reduced pressure. The residue was dissolved in 10% IPA/DCM, washed with an equal volume of (7:3) sat. aqueous NH4Cl/conc. NH4OH (3 times), brine, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with of 0-10% MeOH / DCM. The pure fractions were concentrated under reduced pressure then the residue was dissolved in a warm mixture of acetone / MTBE. After cooling to rt, the solid was collected by filtration and dried under reduced pressure to afford 463 mg (33%) of 3-[3-(1-benzyl-1H-1,2,4-triazol-3- yl)pyrrolidin-1-yl]-7-(1-methyl-1H-pyrazol-4-yl)imadazo[1,2-b]pyridazine (Compound 383) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.72 (m, 1H), 8.56 (s, 1H), 8.33 (s, 1H), 8.07 (s, 2H), 7.40-7.25 (m, 5H), 7.13 (s, 1H), 5.36 (s, 2H), 3.88 (s, 3H), 3.80 (m, 1H), 3.66-3.51 (m, 3H), 3.50-3.43 (m,1H), 2.42-2.31 (m, 1H); 2.30-2.19 (m, 1H); MS (ESI) m/z [M+H]+ calcd. for C24H24N8, 426.2; found, 426.1; HPLC purity: 210 nm: 98.5%; 254 nm: 98.0%. Example 61: Synthesis of Exemplary Compound 384 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(pyridin-3-ylmethyl)-1H-1,2,4-triazol-3-yl)piperazin- 1-yl)pyrazolo[1,5-a]pyridine (Compound 384)
Figure imgf000271_0002
Step 1. Preparation of 3-((3-chloro-1H-1,2,4-triazol-1-yl)methyl)pyridine
Figure imgf000272_0001
[0628] To a solution of 3-chloro-1H-1,2,4-triazole (3 g, 28.98 mmol) and 3- (bromomethyl)pyridine hydrobromide (8.8 g, 34.78 mmol) in DMF (40 mL) was added K2CO3 (8.01 g, 57.97 mmol) under nitrogen then the reaction was heated to 80°C and stirred overnight. The mixture was cooled to rt, diluted with water (110 mL), and extracted with EtOAc (2 x 100 mL). The combined organic extracts were washed with brine (2 x 80 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 1.70 g (30%) of 3-((3- chloro-1H-1,2,4-triazol-1-yl)methyl)pyridine as a white solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.76 (s, 1H), 8.59 (d, J = 2.0 Hz, 1H), 8.55 (dd, J = 4.8, 1.6 Hz, 1H), 7.74 (dt, J = 8.0, 2.0 Hz, 1H), 7.42 (dd, J = 7.8, 4.8 Hz, 1H), 5.45 (s, 2H); MS (ESI) m/z [M+H]+ calcd. for C8H7ClN4, 195.0; found, 195.1. Step 2. Preparation of 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(pyridin-3-ylmethyl)-1H- 1,2,4-triazol-3-yl) piperazin-1-yl)pyrazolo[1,5-a]pyridine(Compound 384)
Figure imgf000272_0002
[0629] To a solution of 3-((3-chloro-1H-1,2,4-triazol-1-yl)methyl)pyridine (200 mg, 1.03 mmol, as prepared in the previous step), 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1- yl)pyrazolo[1,5-a]pyridine (Compound 12) (290.2 mg, 0.77 mmol), and Cs2CO3 (837.1 mg, 2.57 mmol) in dioxane (10 mL) under nitrogen was added Pd-PEPPSI-IPentCl 2- methylpyridine (86.4 mg, 0.10 mmol) then the reaction was heated to 100°C and stirred overnight under nitrogen. The mixture was cooled to rt and concentrated under reduced pressure. The residue was diluted with water (60 mL) and extracted with EtOAc (2 x 50 mL). The combined organic extracts were washed with brine (2 x 40 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by reversed-phase flash chromatography with a C18 silica gel column eluting with 10-50% ACN / water (10mM NH4HCO3) to afford 155.1 mg (34%) of 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(pyridin-3-ylmethyl)-1H-1,2,4-triazol-3-yl) piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 384) as a light yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.81 (s, 1H), 8.58 – 8.50 (m, 2H), 8.35 (s, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.76 (s, 1H), 7.71 – 7.69 (m, 1H), 7.65 (d, J = 9.2 Hz, 1H), 7.40 (dd, J = 7.6, 4.8 Hz, 1H), 7.31 (dd, J = 9.2, 1.5 Hz, 1H), 5.29 (s, 2H), 3.86 (s, 3H), 3.50 – 3.40 (m, 4H), 3.06 – 3.04 (m, 4H); MS (ESI) m/z [M+H]+ calcd. for C23H24N10, 441.2; found, 441.2; LCMS purity: 254 nm: 98.9% Example 62: Synthesis of Exemplary Compound 385 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(4-methylbenzyl)-1H-1,2,4-triazol-3-yl)piperazin-1- yl)pyrazolo[1,5-a]pyridine (Compound 385)
Figure imgf000273_0001
Step 1. Preparation of 3-chloro-1-(4-methylbenzyl)-1H-1,2,4-triazole
Figure imgf000273_0002
[0630] To a solution of 3-chloro-1H-1,2,4-triazole (1 g, 9.66 mmol) and 1-(bromomethyl)- 4-methylbenzene (2.15 g, 11.59 mmol) in DMF (20 mL) were added K2CO3 (2.67 g, 19.32 mmol) then the reaction was heated to 80°C and stirred for 16 h under nitrogen. The mixture was cooled to rt and concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with a C18 silica gel column eluting with 10-100% ACN / water (10mM NH4HCO3) to afford 1.5 g (75%) of 3-chloro-1-[(4- methylphenyl)methyl]-1,2,4-triazole as a white solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.72 (s, 1H), 7.29-7.14 (m, 4H), 5.34 (s, 2H), 2.29 (s, 3H). MS (ESI) m/z [M+H]+ calcd. for C10H10ClN3, 208.0; found, 208. Step 2. Preparation of 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(4-methylbenzyl)-1H-1,2,4- triazol-3-yl)piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 385)
Figure imgf000273_0003
[0631] To a solution of 3-chloro-1-[(4-methylphenyl)methyl]-1,2,4-triazole (264.7 mg, 1.27 mmol, as prepared in the previous step) and 6-(1-methyl-1H-pyrazol-4-yl)-3- piperazin-1-ylpyrazolo[1,5-a]pyridine (Compound 12) (300 mg, 1.06 mmol) in dioxane (20 mL) under nitrogen were added Cs2CO3 (692.3 mg, 2.12 mmol) and Pd-PEPPSI-IPentCl 2- methylpyridine (89.3 mg, 0.10 mmol) then the reaction was heated to 100°C and stirred for 16 h under nitrogen. The mixture was cooled to rt and concentrated under reduced pressure. The residue was purified by Prep-TLC (MeOH / DCM 1:10) and Prep-HPLC to afford 122.7 mg (25%) of 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(4-methylbenzyl)-1H-1,2,4- triazol-3-yl)piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 385) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.81 (d, J = 1.6 Hz, 1H), 8.28 (s, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.76 (s, 1H), 7.65 (d, J = 9.2 Hz, 1H), 7.31 (d, J = 9.2, 1H), 7.17 (s, 4H), 5.17 (s, 2H), 3.87 (s, 3H), 3.44 (t, J = 4.9 Hz, 4H), 3.05 (t, J = 4.9 Hz, 4H), 2.28 (s, 3H). MS (ESI) m/z [M+H]+ calcd. for C25H27N9, 454.1; found, 454.10.LCMS purity:254 nm:99.9%. Example 63: Synthesis of Exemplary Compound 386 3-(4-(5-benzylpyrimidin-2-yl)-1,4-diazepan-1-yl)-7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2- b]pyridazine (Compound 386)
Figure imgf000274_0001
Step 1. Preparation of tert-butyl 4-[7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazin- 3-yl]-5-oxo-1,4-diazepane-1-carboxylate
Figure imgf000274_0002
[0632] A suspension of 3-bromo-7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazine (Compound S3) (3.91 g, 13.3 mmol) and tert-butyl 5-oxo-1,4-diazepane-1-carboxylate (3.13 g, 14.6 mmol) in dioxane (72 mL) under nitrogen was treated with N,N'-dimethyl- 1,2-ethanediamine (0.57 mL, 5.3 mmol), CuI (0.507 g, 2.66 mL), and K3PO4 (8.47 g, 39.9 mmol) then the reaction was heated to 100°C and stirred overnight under nitrogen. The mixture was cooled to rt and filtered through a pad of Celite. The filter pad was washed with DCM and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel chromatography eluting with 0-100% of (10% MeOH in DCM) / DCM to afford 2.18 g (40%) of tert-butyl 4-[7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2- b]pyridazin-3-yl]-5-oxo-1,4-diazepane-1-carboxylate as a tan solid. MS (ESI) m/z [M+H]+ calcd. for C20H26N7O3, 412.2; found, 411.7 Step 2. Preparation of tert-butyl 4-[7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazin- 3-yl]-5-thioxo-1,4-diazepane-1-carboxylate
Figure imgf000275_0001
[0633] A suspension of tert-butyl 4-[7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2- b]pyridazin-3-yl]-5-oxo-1,4-diazepane-1-carboxylate (1.95 g, 4.74 mmol, as prepared in the previous step) in THF (75 mL) was warmed to 55 °C then 2,4-bis(4-methoxyphenyl)- 2,4-dithioxo-1,3,2,4-dithiadiphosphetane (1.3 g, 3.3 mmol) was added. The reaction warmed to 60 °C and stirred for 2 h, then additional 2,4-bis(4-methoxyphenyl)-2,4- dithioxo-1,3,2,4-dithiadiphosphetane (1.34 g, 3.32 mmol) was added and the reaction was stirred at 60 °C for 4.5 h. Additional 2,4-bis(4-methoxyphenyl)-2,4-dithioxo-1,3,2,4- dithiadiphosphetane (1.50 g, 3.71 mmol) was added and the reaction was stirred at 60°C for 1.5 h at 60 °C. The mixture was cooled to rt and stirred overnight then rewarmed to 60 °C, 2,4-bis(4-methoxyphenyl)-2,4-dithioxo-1,3,2,4-dithiadiphosphetane (1.4 g, 3.5 mmol) was added, and the mixture was stirred for 2.5 h. The reaction was cooled to rt, concentrated under reduced pressure, and the residue was dissolved in DCM (100 mL). The mixture was washed with a mixture of sat. aqueous NaHCO3 (50 mL) and water (100 mL). DCM (100 mL) was added to the aqueous layer resulting in an emulsion. The mixture was filtered through a pad of Celite, and the layers separated. The filter pad was washed with DCM and this filtrate was used to extract the aqueous layer. The filter pad was washed with DCM and the filtrate was used to extract the aqueous layer an additional three times until the aqueous layer showed no desired product by HPLC. The filter pad was washed with DCM until HPLC showed no additional product eluted from the Celite pad. All the DCM extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with 20% DCM in acetone to afford 1.83g (90%) of tert-butyl 4- [7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazin-3-yl]-5-thioxo-1,4-diazepane-1- carboxylate as a yellow solid. MS (ESI) m/z [M+H]+ calcd. for C20H26N7O2S, 428.2; found, 428.1 Step 3. Preparation of tert-butyl 4-[7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazin- 3-yl]-1,4-diazepane-1-carboxylate
Figure imgf000276_0001
[0634] To a mixture of tert-butyl 4-[7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2- b]pyridazin-3-yl]-5-thioxo-1,4-diazepane-1-carboxylate (107 mg, 0.25 mmol, as prepared in the previous step) in MeOH (6 mL) was added Raney nickel (1.5 g, 26 mmol) as a suspension in MeOH (5 mL) then the reaction was stirred vigorously at rt overnight. The mixture was filtered through a pad of Celite then the filter pad was washed well with THF. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with 20% DCM in acetone to afford 65 mg (66%) of tert-butyl 4-[7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazin-3-yl]-1,4-diazepane-1- carboxylate as an orange foam. MS (ESI) m/z [M+H]+ calcd. for C20H28N7O2, 398.2; found, 398.0 Step 4. Preparation of 3-[4-(5-benzylpyrimidin-2yl)-1,4-diazepan-1-yl]-7-(1-methyl-1H- pyrazol-4-yl)imidazo[1,2-b]pyridazine (Compound 386)
Figure imgf000276_0002
[0635] To a solution of tert-butyl 4-[7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2- b]pyridazin-3-yl]-1,4-diazepane-1-carboxylate (124 mg, 0.31 mmol, as prepared in the previous step) in DCM (6.3 mL) cooled to 0 °C was added TFA (1.3 mL, 17 mmol) dropwise then the reaction was stirred for 30 minutes. The mixture was concentrated under reduced pressure then the residue was stirred with toluene and concentrated under reduced pressure. This was repeated two more times. The residue was then stirred with DCM and concentrated under reduced pressure. This was repeated two more times and the residue placed under high vacuum. The residue was dissolved in NMP (2.3 mL) then 5-benzyl-2- chloropyrimidine (67 mg, 0.33 mmol) and K2CO3 (216 mg, 1.57 mmol) were added. The mixture was heated to 110 °C for 3.5 h, cooled to rt, and stirred overnight. The reaction was rewarmed to 110 °C and stirred for another 4.5 h. The reaction was cooled to rt, diluted with EtOAc (30 mL), and washed with water (2 x 10 mL). The aqueous layer was extracted with EtOAc (2 x 10 mL). The combined organic extracts were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with 0-100% of (10% MeOH in DCM) / DCM. The pure fractions were combined and concentrated under reduced pressure then the residue was stirred with hexane and concentrated (x3), then dried under reduced pressure overnight. The solid residue was triturated with 25% ether in hexane, filtered, washed with hexane and dried at 50 °C overnight under reduced pressure to afford 77 mg (53%) of 3-[4-(5- benzylpyrimidin-2yl)-1,4-diazepan-1-yl]-7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2- b]pyridazine (Compound 386) as a yellow solid. 1H NMR (400 MHz, CD3OD) δ (ppm) 8.66 (d, J=2.08 Hz, 1H) 8.12 - 8.19 (m, 3H) 7.98 (s, 1H) 7.87 (d, J=2.20 Hz, 1H) 7.24 - 7.32 (m, 2H) 7.15 - 7.22 (m, 3H) 7.11 (s, 1H) 3.98 - 4.04 (m, 2H) 3.96 (s, 3H) 3.91 (t, J=6.05 Hz, 2H) 3.79 (s, 2H) 3.62 - 3.69 (m, 2H) 3.44 - 3.53 (m, 2H) 2.06 (quin, J=5.87 Hz, 2H); MS (ESI) m/z [M+H]+ calcd. for C26H28N9, 466.3; found, 466.1; HPLC purity: 210 nm: 99.6%; 254 nm: 99.6%. Example 64: Synthesis of Exemplary Compound 387 N-(4-chlorobenzyl)-4-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazin-3- yl)piperazine-1-carboxamide (Compound 387)
Figure imgf000277_0001
Step 1. Preparation of tert-butyl 4-[7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazin- 3-yl]piperazine-1-carboxylate
Figure imgf000278_0001
[0636] A suspension of 3-bromo-7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazine (Compound S3) (6.56 g, 23.6 mmol), tert-butyl 1-piperazinecarboxylate (22.0 g, 118 mmol) and KOtBu (3.97 g, 35.4 mmol) in tBuOH (66 mL) and dioxane (33 mL) was sparged with argon for 10 minutes. The reaction was treated with tBuXPhos Pd G1 (2.43 g, 3.54 mmol) and the mixture sparged again for another 10 minutes. The reaction was sparged an additional 2 minutes while being agitated in an ultrasound bath. The reaction was then warmed at 55 °C under argon for 3 h. The reaction was concentrated under reduced pressure and the residue was diluted with DCM (300 mL). The mixture was washed with 10% citric acid (4 x 100 mL). The combined citric acid washes were extracted with DCM (100 mL). The combined DCM extracts were washed with water (3 x 100 mL). The combined water washes were extracted with DCM (100 mL). The combined DCM extracts were washed with sat. aqueous NaHCO3 (100 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with 0-100% of (10% MeOH in DCM) / DCM to afford 6.62 g (73%) of tert-butyl 4-[7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazin-3- yl]piperazine-1-carboxylate as a yellow green solid. MS (ESI) m/z [M+H]+ calcd. for C19H26N7O2, 384.2; found, 394.0. Step 2. Preparation of 7-(1-methyl-1H-pyrazol-4-yl)-3-piperazin-1-ylimidazo[1,2- b]pyridazine trihydrochloride salt
Figure imgf000278_0002
[0637] To a suspension of tert-butyl 4-[7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2- b]pyridazin-3-yl]piperazine-1-carboxylate (4.6 g, 12 mmol, as prepared in the previous step) in EtOAc (69 mL) heated to 40 °C was added conc. HCl (5.1 mL, 60 mmol) slowly over 10 minutes while under nitrogen. The slurry was stirred at 45 °C for 3 h, cooled to rt, and diluted with MTBE (180 mL). After stirring 10 minutes, the mixture was filtered, and the filter pad washed with MTBE (4 x 40 mL). The solids were air dried to afford 4.50 g (96%) of 7-(1-methyl-1H-pyrazol-4-yl)-3-piperazin-1-ylimidazo[1,2-b]pyridazine trihydrochloride salt as a yellow-green solid. MS (ESI) m/z [M+H]+ calcd. for C14H18N7, 284.2; found, 284.1. Step 3. Preparation of N-(4-chlorobenzyl)-4-[6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl]piperazine-1-carboxamide (Compound 387)
Figure imgf000279_0001
[0638] A mixture of 7-(1-methyl-1H-pyrazol-4-yl)-3-piperazin-1-ylimidazo[1,2- b]pyridazine trihydrochloride salt (100 mg, 25 mmol, as prepared in the previous step) in ACN (3 mL) was cooled to 0°C then DIEA (0.22 mL, 1.3 mmol) and 1-chloro-4- (isocyanatomethyl)benzene (34 μL, 0.26 mmol) were added. The reaction was warmed to rt and stirred for 30 minutes, then additional ACN (3 mL) was added. After stirring 1 h, the reaction was concentrated under reduced pressure. The residue was partitioned between water (10 mL), DCM (10 mL), and 10% MeOH in DCM (5 mL). The water layer was washed with 10% MeOH in DCM (3 x 5 mL). The combined organic extracts were washed with sat. aqueous NaHCO3 (5 mL), and brine (5 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with 0-100% (10% MeOH in DCM) / DCM to afford 73 mg (64%) of N-(4-chlorobenzyl)-4-[6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl]piperazine-1-carboxamide (Compound 387) as a yellow solid; 1H NMR (400 MHz, CD3OD) d (ppm) 8.74 (d, J=1.96 Hz, 1H) 8.20 (s, 1H) 8.02 (s, 1H) 7.96 (d, J=2.08 Hz, 1H) 7.23 - 7.35 (m, 5H) 4.36 (s, 2H) 3.96 (s, 3H) 3.62 - 3.71 (m, 4H) 3.23 - 3.30 (m, 4H); MS (ESI) m/z [M+H]+ calcd. for C22H24ClN8O, 451.2; found, 451.0; HPLC purity: 210 nm: 99.5%; 254 nm: 99.4%. Example 65: Synthesis of Exemplary Compound 388 2-benzyl-5-(4-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazin-3-yl)piperazin-1-yl)- 1,3,4-thiadiazole (Compound 388)
Figure imgf000280_0001
[0639] To a mixture of 2-benzyl-5-bromo-1,3,4-thiadiazole (243 mg, 0.95 mmol, as prepared in Example 55, Step 2) and 7-(1-methyl-1H-pyrazol-4-yl)-3-piperazin-1- ylimadazo[1,2-b]pyridazine trihydrochloride salt (404 mg, 1.03 mmol, as prepared in Example 64, Step 2) in DMAc (6.8 mL) was added DIEA (1.60 mL, 9.18 mmol) under a nitrogen then the reaction was heated to 100°C and stirred for 25 h. The mixture was cooled to rt, diluted with EtOAc, was washed with water (3 times) and brine, then dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with 0-15% MeOH/DCM to afford 114 mg (26%) of 3-[4-(5-benzyl-1,3,4-thiadiazol-2-yl)piperazin-1-yl]-7-(1-methyl- 1H-pyrazol-4-yl)imidazo[1,2-b]pyridazine (Compound 388) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (d, J= 2.1 Hz, 1H), 8.36 (s, 1H), 8.17 (d, J= 2.1 Hz, 1H), 8.10 (s, 1H), 7.40-7.21 (m, 6H), 4.25 (s, 2H), 3.89 (s, 3H), 3.62 (m, 4H), 3.31 (m, 5H); MS (ESI) m/z [M+H]+ calcd. for C23H24N9S, 458.2; found, 458.1; HPLC purity: 210 nm: 97.2%; 254 nm: 98.1%. Example 66: Synthesis of Exemplary Compound 389 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-((6-methylpyridin-3-yl)methyl)-1H-1,2,4-triazol-3- yl)piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 389)
Figure imgf000280_0002
Step 1. Preparation of 5-((3-chloro-1H-1,2,4-triazol-1-yl)methyl)-2-methylpyridine
Figure imgf000281_0001
[0640] To a solution of 3-chloro-1H-1,2,4-triazole (1.00 g, 9.66 mmol) and 5- (bromomethyl)-2-methylpyridine hydrobromide (3.09 g, 11.59 mmol) in DMF (100 mL) was added K2CO3 (2.67 g, 19.32 mmol) then the reaction was heated to 90°C and stirred for 16 h under nitrogen. The mixture was cooled to rt and concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with a C18 silica gel column eluting with 10-100% ACN / water (10 mM NH4HCO3) to afford 240 mg (12%) of 5-((3-chloro-1H-1,2,4-triazol-1-yl)methyl)-2-methylpyridine as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.74 (s, 1H), 8.46 (s, 1H), 7.65-7.62 (m, 1H), 7.28- 7.25 (m, 1H), 5.40 (s, 2H), 2.46 (s, 3H). MS (ESI) m/z [M+H]+ calcd. for C9H9ClN4, 209.0; found, 209.0. Step 2. 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-((6-methylpyridin-3-yl)methyl)-1H-1,2,4- triazol-3-yl)piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 389)
Figure imgf000281_0002
[0641] To a solution of 5-((3-chloro-1H-1,2,4-triazol-1-yl)methyl)-2-methylpyridine (221.6 mg, 1.06 mmol, as prepared in the previous step) and 6-(1-methyl-1H-pyrazol-4- yl)-3-(piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 12) (300 mg, 1.06 mmol) in dioxane (20 mL) under nitrogen were added Cs2CO3 (692.3 mg, 2.12 mmol) and Pd- PEPPSI-IPentCl 2-methylpyridine (89.3 mg, 0.10 mmol) then the reaction was heated to 100°C and stirred overnight under nitrogen. The mixture was cooled to rt and concentrated under reduced pressure. The residue was dissolved in water (100 mL) then the aqueous mixture was extracted with EtOAc (3 x 100 mL). The combined organic extracts were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 122.7 mg (25%) of 6-(1-methyl-1H- pyrazol-4-yl)-3-(4-(1-((6-methylpyridin-3-yl)methyl)-1H-1,2,4-triazol-3-yl)piperazin-1- yl)pyrazolo[1,5-a]pyridine (Compound 389) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.81 (s, 1H), 8.42 (d, J = 2.3 Hz, 1H), 8.32 (s, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.76 (s, 1H), 7.66-7.63 (m, 1H), 7.60-7.58 (m, 1H), 7.32-7.30 (m,1H), 7.26-7.24 (m, 1H), 5.23 (s, 2H), 3.86 (s, 3H), 3.44 (t, J = 5.0 Hz, 4H), 3.05 (t, J = 5.0 Hz, 4H), 2.45 (s, 3H). MS (ESI) m/z [M+H]+ calcd. for C24H26N10, 455.2; found, 455.25.LCMS purity:254 nm: 99.8%. Example 67: Synthesis of Exemplary Compound 390 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(3-methylbenzyl)-1H-1,2,4-triazol-3-yl)piperazin-1- yl)pyrazolo[1,5-a]pyridine (Compound 390)
Figure imgf000282_0001
Step 1. Preparation of 3-chloro-1-(3-methylbenzyl)-1H-1,2,4-triazole
Figure imgf000282_0002
[0642] To a solution of 3-chloro-1H-1,2,4-triazole (2 g, 19.32 mmol) and 1- (bromomethyl)-3-methylbenzene (4.29 g, 23.19 mmol) in DMF (20 mL) was added K2CO3 (5.34 g, 38.64 mmol) in portions at rt then the reaction was heated to 80°C and stirred overnight. The mixture was cooled to rt and purified by Prep-AChiral-HPLC to afford 2.75 g (69%) of 3-chloro-1-(3-methylbenzyl)-1H-1,2,4-triazole as a colorless oil:1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.73 (s, 1H), 7.23 – 7.24 (m, 1H), 7.15 – 7.09 (m, 3H), 5.34 (s, 2H), 2.29 (s, 3H).MS (ESI) m/z [M+H]+ calcd. for C10H10ClN, 208.0; found, 208.1. Step 2. Preparation of 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(3-methylbenzyl)-1H-1,2,4- triazol-3-yl)piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 390)
Figure imgf000282_0003
[0643] To a solution of 3-chloro-1-(3-methylbenzyl)-1H-1,2,4-triazole (200 mg, 0.96 mmol, as prepared in the previous step), 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1- yl)pyrazolo[1,5-a]pyridine (Compound 12) (271.9 mg, 0.96 mmol) and Cs2CO3 (784.5 mg, 2.41 mmol) in dioxane (10 mL) under nitrogen was added Pd-PEPPSI-IPentCl 2- methylpyridine (81 mg, 0.10 mmol) then the reaction was heated to 100°C and stirred overnight under nitrogen. The mixture was cooled to rt and concentrated under reduced pressure. The residue was diluted with water (60 mL) then the aqueous mixture was extracted with EtOAc (2 x 50 mL). The combined organic extracts were washed with brine (2 x 40 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 158.3 mg (36%) of 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(3-methylbenzyl)-1H-1,2,4-triazol-3-yl) piperazin- 1-yl)pyrazolo[1,5-a]pyridine (Compound 390) as a light yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.30 (s, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.77 (s, 1H), 7.65 (d, J = 9.2 Hz, 1H), 7.31 (dd, J = 9.3, 1.4 Hz, 1H), 7.25 (t, J = 7.5 Hz, 1H), 7.10 (dd, J = 14.2, 8.0 Hz, 3H), 5.19 (s, 2H), 3.87 (s, 3H), 3.51 – 3.39 (m, 4H), 3.12 – 2.99 (m, 4H), 2.30 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C25H27N9, 454.3; found, 454.3; LCMS purity: 254 nm: 99.9% Example 68: Synthesis of Exemplary Compound 391 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(4-(trifluoromethyl)benzyl)-1H-1,2,4-triazol-3- yl)piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 391)
Figure imgf000283_0001
Step 1. Preparation of 3-bromo-1-(4-(trifluoromethyl)benzyl)-1H-1,2,4-triazole
Figure imgf000283_0002
[0644] To a solution of 3-bromo-1H-1,2,4-triazole (3 g, 20.27 mmol) in DMF (30 mL) were added 1-(bromomethyl)-4-(trifluoromethyl)benzene (5.82 g, 24.33 mmol) and K2CO3 (5.60 g, 40.55 mmol) then the reaction was warmed to 80°C and stirred for 16 h. The mixture was cooled to rt, and filtered, then the filter cake was washed with DMF (2 x 1 mL). The filtrate was concentrated under reduced pressure then the residue was purified by reversed-phase flash chromatography with a C18 silica gel column eluting with 10-80% ACN / water to afford 4 g (58%) of 3-bromo-1-(4-(trifluoromethyl)benzyl)-1H-1,2,4- triazole as a white solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.75 (s, 1H), 7.76 (d, J = 8.1 Hz, 2H), 7.51 (d, J = 8.0 Hz, 2H), 5.53 (s, 2H). MS (ESI) m/z [M+H]+ calcd. for C10H7BrF3N3, 306.0; found, 306.0. Step 2. Preparation of 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(4-(trifluoromethyl)benzyl)- 1H-1,2,4-triazol-3-yl)piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 391)
Figure imgf000284_0001
[0645] To a solution of 3-bromo-1-(4-(trifluoromethyl)benzyl)-1H-1,2,4-triazole (277.9 mg, 1.06 mmol, as prepared in the previous step) and 6-(1-methyl-1H-pyrazol-4-yl)-3- piperazin-1-ylpyrazolo[1,5-a]pyridine (Compound 12) (200 mg, 0.71 mmol) in dioxane (10 mL) under nitrogen were added Cs2CO3 (461.5 mg, 1.41 mmol) and Pd-PEPPSI-IPentCl 2- methylpyridine (59.5 mg, 0.07 mmol) then the reaction was warmed to 100°C and stirred overnight under nitrogen. The mixture was cooled to rt and concentrated under reduced pressure. The residue was dissolved in water (50 mL) then the aqueous mixture was extracted with EtOAc (3 x 50 mL). The combined organic extracts were dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 105.3 mg (29%) of 6-(1-methyl-1H- pyrazol-4-yl)-3-(4-(1-(4-(trifluoromethyl)benzyl)-1H-1,2,4-triazol-3-yl)piperazin-1- yl)pyrazolo[1,5-a]pyridine (Compound 391) as a white solid.1H NMR (400 MHz, DMSO- d6) δ (ppm) 8.82 (s, 1H), 8.37 (s, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.78 – 7.71 (m, 3H), 7.65 (d, J = 9.2 Hz, 1H), 7.47 (d, J = 8.0 Hz, 2H), 7.33-7.29 (m, 1H), 5.36 (s, 2H), 3.86 (s, 3H), 3.45 (t, J = 4.8 Hz, 4H), 3.06 (t, J = 4.9 Hz, 4H). MS (ESI) m/z [M+H]+ calcd. for C25H24F3N9, 508.2; found, 508.25.LCMS purity:254 nm:99.9%. Example 69: Synthesis of Exemplary Compound 392 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(1-phenylethyl)-1H-1,2,4-triazol-3-yl)piperazin-1- yl)pyrazolo[1,5-a]pyridine (Compound 392)
Figure imgf000285_0001
Step 1. Preparation of 3-bromo-1-(1-phenylethyl)-1H-1,2,4-triazole
Figure imgf000285_0002
[0646] To a solution of 3-bromo-1H-1,2,4-triazole (3 g, 20.3 mmol) in DMF (30 mL) was added (1-bromoethyl)benzene (4.5 g, 24.3 mmol) and K2CO3 (5.6 g, 40.5 mmol) then the reaction was warmed to 80°C and stirred for 16 h. The mixture was cooled to rt and concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography with a C18 silica gel column eluting with 10-50% ACN / water (10mM NH4HCO3) and Prep-HPLC to afford 2.9 g (51%) of 3-bromo-1-(1-phenylethyl)-1,2,4- triazole as a light yellow oil.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.75 (s, 1H), 7.41 – 7.27 (m, 5H), 5.79 – 5.69 (m, 1H), 1.81 (d, J = 7.1 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C10H10BrN3, 252.0; found, 252.1. Step 2. Preparation of 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(1-phenylethyl)-1H-1,2,4- triazol-3-yl)piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 392)
Figure imgf000285_0003
[0647] To a solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-piperazin-1-ylpyrazolo[1,5- a]pyridine (Compound 12) (200 mg, 0.71 mmol) and 3-bromo-1-(1-phenylethyl)-1H-1,2,4- triazole (214 mg, 0.85 mmol, as prepared in the previous step) in dioxane (5 mL) under nitrogen was added Cs2CO3 (462 mg, 1.42 mmol) and Pd-PEPPSI-IPentCl 2-methylpyridine (60 mg, 0.07 mmol) then the reaction was warmed to 90°C and stirred for 16 h under nitrogen. The mixture was cooled to rt and filtered, then the filter cake was washed with EtOAc (2x5 mL). The filtrate was concentrated under reduced pressurethen the residue was purified by silica gel chromatography eluting with MeOH / DCM (1:25) and Prep-HPLC to afford 111.5 mg (35%) of 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(1-phenylethyl)-1H- 1,2,4-triazol-3-yl)piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 392) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.81 (s, 1H), 8.34 (s, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.76 (s, 1H), 7.65 (d, J = 6.2 Hz, 1H), 7.41 – 7.24 (m, 6H), 5.51 (q, J = 7.0 Hz, 1H), 3.87 (s, 3H), 3.48 – 3.41 (m, 4H), 3.09 – 3.02 (m, 4H), 1.77 (d, J = 7.1 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C25H27N9, 454.2; found, 454.2; LCMS purity: 254 nm: 99.7% Example 70: Synthesis of Exemplary Compound 393 (R)-1-(4-chlorophenyl)ethyl 4-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazin-3-yl)- 1,4-diazepane-1-carboxylate (Compound 393)
Figure imgf000286_0001
[0648] To a solution of tert-butyl 4-[7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2- b]pyridazin-3-yl]-1,4-diazepane-1-carboxylate (182 mg, 0.31 mmol, as prepared in Example 63, Step 3) in DCM (9.1 mL) cooled to 0 °C was added TFA (2 mL, 25 mmol) dropwise, then the reaction was stirred for 30 minutes. The mixture was concentrated under reduced pressure then the residue was stirred with toluene and concentrated under reduced pressure. This was repeated three more times, then the residue was suspended in EtOAc (3 mL) and cooled to 0 °C. TEA (0.277 mL, 1.99 mmol) was added and the mixture was stirred at 0°C for 5 minutes, then (R)-1-(4-chlorophenyl)ethyl (2,5-dioxopyrrolidin-1-yl) carbonate (Compound S95) (118 mg, 0.40 mmol) was added and the reaction was stirred at 0°C for 2 h. The mixture was filtered through a pad of Celite and the filter pad was washed with EtOAc (6 x 5mL). The combined filtrates were washed with sat. aqueous NaHCO3 (2 x 8 mL) and water (8 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with 0-100% (10% MeOH in DCM) / DCM. The pure fractions were combined and concentrated under reduced pressure then the residue was stirred with hexane and concentrated several times then dried under vacuum at 40 °C to afford 158 mg (91%) of (1R)-1-(4-chlorophenyl)ethyl 4-[6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl]piperazine-1-carboxylate (Compound 393) as a yellow solid.1H NMR (400 MHz, METHANOL-d4) d ppm 8.66 (dd, J=10.64, 1.96 Hz, 1H) 8.17 (s, 1H) 8.00 (s, 1H) 7.88 (dd, J=9.41, 2.08 Hz, 1H) 7.28 - 7.38 (m, 2H) 7.13 - 7.23 (m, 3H) 5.61 - 5.78 (m, 1H) 3.96 (d, J=1.47 Hz, 3H) 3.43 - 3.85 (m, 8H) 1.89 - 2.12 (m, 2H) 1.40 - 1.57 (m, 3H); MS (ESI) m/z [M+H]+ calcd. for C24H27ClN7O2, 480.2; found, 480.1; HPLC purity: 210 nm: 100.0%; 254 nm: 100.0%. Example 71: Synthesis of Exemplary Compound 394 3-(3-(5-benzylpyrimidin-2-yl)pyrrolidin-1-yl)-7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2- b]pyridazine (Compound 394)
Figure imgf000287_0001
[0649] A mixture of 3-bromo-7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazine (Compound S3) (0.186 g) and 5-benzyl-2-pyrrolidin-3-ylpyrimidine (Compound S100) (0.400 g, 1.67 mmol) in tBuOH (1.85 mL) and dioxane (0.939 mL) was sparged with argon for 15 minutes then tBuXPhos Pd G1 (0.0459 g, 0.0668 mmol) and KOtBu (0.112 g, 1.00 mmol) were added and the mixture was sparged again for 10 minutes with argon. The reaction was warmed to 30°C and stirred for 12 h. The mixture was concentrated under reduced pressure and the residue was taken up in EtOAc (50 mL). The mixture was washed with water (3 x 50 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with 5% MeOH / DCM to afford 39 mg (13%) of 3-[3-(5- benzylpyrimidin-2-yl)pyrrolidin-1-yl]-7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2- b]pyridazine (Compound 394) as an orange solid. 1H NMR (400 MHz, CDCl3) δ (ppm) 8.57 (s, 2H), 8.42 (d, J=2.20 Hz, 1H), 7.82 (s, 1H), 7.79 (d, J=2.20 Hz, 1H), 7.71 (s, 1H), 7.32 - 7.39 (m, 2H), 7.26 - 7.30 (m, 1H), 7.17 - 7.24 (m, 3H), 4.02 - 4.07 (m, 1H), 4.00 (s, 3H), 3.91 - 3.99 (m, 3H), 3.72 - 3.84 (m, 2H), 3.61 (td, J=8.68, 5.50 Hz, 1H), 2.53 - 2.64 (m, 1H), 2.44 - 2.53 (m, 1H); MS (ESI) m/z [M+H]+ calcd. For C25H25N8: 437.5; found: 437.1; HPLC purity: 210 nm: 100.0%; 254 nm: 100.0% Example 72: Synthesis of Exemplary Compound 395 3-(4-(1-benzyl-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-7-(1-methyl-1H-pyrazol-4- yl)imidazo[1,2-b]pyridazine (Compound 395)
Figure imgf000288_0001
[0650] A mixture of tBuOH (0.73 mL), dioxane (0.12 mL), and water (5 μL) sparged with a slow stream of argon for 20 minutes was added to a mixture of 1-benzyl-3-bromo-1H- 1,2,4-triazole (85 mg, 0.36 mmol, as prepared for Compound S101, Step 1), 7-(1-methyl- 1H-pyrazol-4-yl)-3-pyridazine trihydrochloride (155 mg, 0.39 mmol, as prepared in Example 64, Step 2), KOtBu (181 mg, 1.61 mmol), and tBuXPhos Pd G1 (12 mg, 0.02 mmol) under argon, then the reaction was evacuated and back filled with argon three times. The reaction was warmed to 60°C and stirred for 1 h. The reaction was cooled to rt andL concentrated under reduced pressure. The residue was partitioned between EtOAc (40 mL) and water (10 mL). The aqueous layer was washed with EtOAc (3 x 40 mL). The combined organic extracts were dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with 0-100% (10% MeOH in DCM) / DCM. The pure fractions were combined and concentrated under reduced pressure then the residue was dissolved in hexane and concentrated under reduced pressure. This was repeated until a yellow solid formed. The residue was dried under reduced pressure at 50 °C to afford 107 mg (68%) of 3-[4-(1-benzyl-1H-1,2,4-triazol-3-yl)piperazin-1-yl]-7-(1-methyl-1H-pyrazol-4- yl)imidazo[1,2-b]pyridazine (Compound 395) as a yellow solid; 1H NMR (400 MHz, CD3OD) d (ppm) 8.73 (d, J=2.08 Hz, 1H) 8.20 (d, J=1.59 Hz, 2H) 8.02 (s, 1H) 7.96 (d, J=2.08 Hz, 1H) 7.24 - 7.41 (m, 6H) 5.25 (s, 2H) 3.96 (s, 3H) 3.56 - 3.64 (m, 4H) 3.33 - 3.41 (m, 4H); MS (ESI) m/z [M+H]+ calcd. for C23H25N10, 441.2; found, 441.2; HPLC purity: 210 nm: 100.0%; 254 nm: 100.0%. Example 73: Synthesis of Exemplary Compound 396 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(pyridin-4-ylmethyl)-1H-1,2,4-triazol-3-yl)piperazin- 1-yl)pyrazolo[1,5-a]pyridine (Compound 396)
Figure imgf000289_0001
Step 1.4-((3-chloro-1H-1,2,4-triazol-1-yl)methyl)pyridine
Figure imgf000289_0002
[0651] To a solution of 3-chloro-1H-1,2,4-triazole (2.00 g, 19.32 mmol) and 4- (bromomethyl)pyridine (3.99 g, 23.19 mmol) in DMF (40 mL) was added K2CO3 (5.34 g, 38.64 mmol) then the reaction was warmed to 80°C and stirred overnight. The mixture was cooled to rt and purified by reversed-phase flash chromatography with a C18 silica gel column eluting with 10-50% ACN / water (10mM NH4HCO3) to afford 600 mg (16%) of 4-((3-chloro-1H-1,2,4-triazol-1-yl)methyl)pyridine as a black oil. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.78 (s, 1H), 8.57 (d, J = 4.9 Hz, 2H), 7.24 (d, J = 4.7 Hz, 2H), 5.48 (s, 2H) ; MS (ESI) m/z [M+H] + calcd. for C8H7ClN4, 195; found, 195. Step 2. 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(pyridin-4-ylmethyl)-1H-1,2,4-triazol-3- yl)piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 396)
Figure imgf000289_0003
[0652] To a solution of 4-((3-chloro-1H-1,2,4-triazol-1-yl)methyl)pyridine (520 mg, 2.67 mmol, as prepared in the previous step) and 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1- yl)pyrazolo[1,5-a]pyridine (Compound 12) (0.75 g, 2.67 mmol) in dioxane (20 mL) under nitrogen were added Pd-PEPPSI-IPentCl 2-methylpyridine (0.22 g, 0.27 mmol) and Cs2CO3 (1.74 g, 5.34 mmol) then the reaction was warmed to 100°C and stirred overnight under nitrogen. The mixture was cooled to rt and purified by Prep-HPLC to afford 116.7 mg, 10%) of 6-(1-methyl-1H-pyrazol-4-yl)-3-(4-(1-(pyridin-4-ylmethyl)-1H-1,2,4-triazol-3- yl)piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 396) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (t, J = 1.2 Hz, 1H), 8.57 – 8.53 (m, 2H), 8.36 (s, 1H), 8.22 (s, 1H), 7.97 (d, J = 0.8 Hz, 1H), 7.76 (s, 1H), 7.65 (dd, J = 9.3, 1.0 Hz, 1H), 7.31 (dd, J = 9.3, 1.6 Hz, 1H), 7.25 – 7.16 (m, 2H), 5.31 (s, 2H), 3.86 (s, 3H), 3.46 (t, J = 5.0 Hz, 4H), 3.06 (t, J = 5.0 Hz, 4H); MS (ESI) m/z [M+H]+ calcd. for C23H24N10, 441.2; found, 441.2; LCMS purity: 254 nm: 99.7% Example 74: Synthesis of Exemplary Compound 397 (R)-1-(4-(trifluoromethyl)phenyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrazin-3-yl)piperazine-1-carboxylate (Compound 397)
Figure imgf000290_0001
[0653] To a solution of 1-[6-(1-methylpyrazol-4-yl)pyrazolo[1,5-a]pyrazin-3- yl]piperazine (Compound S103) (120 mg, 0.42 mmol) and (1R)-1-[4- (trifluoromethyl)phenyl]ethanol (242 mg, 1.27 mmol) in pyridine (2 mL) cooled to 0°C was added triphosgene (377 mg, 1.27 mmol) then the reaction was stirred at 0°C for 2 h. The mixture was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with MeOH / DCM (1:20) and Prep-HPLC to afford 73.6 mg (35%) of (R)-1-(4-(trifluoromethyl)phenyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyrazin-3-yl)piperazine-1-carboxylate (Compound 397) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.19 (d, J = 1.4 Hz, 1H), 8.91 (d, J = 1.4 Hz, 1H), 8.22 (s, 1H), 8.01 (s, 1H), 7.83 (s, 1H), 7.74 (d, J = 8.2 Hz, 2H), 7.61 (d, J = 8.1 Hz, 2H), 5.82 (q, J = 6.5 Hz, 1H), 3.88 (s, 3H), 3.80 – 3.48 (m, 4H), 3.14 (s, 4H), 1.51 (d, J = 6.5 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C24H24F3N7O2, 500.2; found, 500.2; LCMS purity: 254 nm: 99.8% Example 75: Synthesis of Exemplary Compound 398 (R)-1-(4-chlorophenyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrazin-3- yl)piperazine-1-carboxylate (Compound 398)
Figure imgf000291_0001
[0654] To a solution of 1-[6-(1-methylpyrazol-4-yl)pyrazolo[1,5-a]pyrazin-3- yl]piperazine (Compound S103) (120 mg, 0.42 mmol) and (R)-1-(4-chlorophenyl)ethanol (199 mg, 1.27 mmol) in pyridine (2 mL) cooled to 0°C was added triphosgene (377 mg, 1.27 mmol) then the reaction was stirred at 0°C for 2 h. The mixture was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with MeOH / DCM (1:20) and Prep-HPLC to afford 65.6 mg (32%) of (R)-1-(4- chlorophenyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrazin-3- yl)piperazine-1-carboxylate (Compound 398) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.19 (s, 1H), 8.91 (s, 1H), 8.22 (s, 1H), 8.01 (s, 1H), 7.83 (s, 1H), 7.47 – 7.37 (m, 4H), 5.74 (q, J = 6.5 Hz, 1H), 3.88 (s, 3H), 3.74 – 3.50 (m,4H), 3.12 (s, 4H), 1.48 (d, J = 6.6 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C23H24ClN7O2, 466.2; found, 466.1; LCMS purity: 254 nm: 97.7% Example 76: Synthesis of Exemplary Compound 399 (2-methylbenzo[d]thiazol-5-yl)methyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazine-1-carboxylate (Compound 399)
Figure imgf000291_0002
[0655] To a stirred solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1- yl)pyrazolo[1,5-a]pyridine (Compound 12) (200 mg, 0.71 mmol) and (2- methylbenzo[d]thiazol-5-yl)methanol (190.4 mg, 1.06 mmol) in pyridine (10 mL) was added triphosgene (84.1 mg, 0.28 mmol) then the reaction was stirred at rt for 16 h. The mixture was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with MeOH / DCM (1:40) and Prep-HPLC to afford 132.7 mg (38%) of (2-methylbenzo[d]thiazol-5-yl)methyl 4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 399) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (t, J = 1.2 Hz, 1H), 8.23 (s, 1H), 8.04 (d, J = 8.2 Hz, 1H), 7.98 – 7.95 (m, 1H), 7.93 – 7.90 (m, 1H), 7.76 (s, 1H), 7.67 – 7.61 (m, 1H), 7.42 (dd, J = 8.2, 1.5 Hz, 1H), 7.32 (dd, J = 9.2, 1.5 Hz, 1H), 5.26 (s, 2H), 3.87 (s, 3H), 3.62 (s, 4H), 3.03 – 2.91 (m, 4H), 2.81 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C25H25N7O2S, 488.2; found, 488.2; purity: 254 nm: 99.9% Example 77: Synthesis of Exemplary Compound 400 3-(4-(1-benzyl-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyrazine (Compound 400)
Figure imgf000292_0001
[0656] To a solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)pyrazolo[1,5- a]pyrazine (Compound S103) (120 mg, 0.42 mmol) in dioxane (5 mL) under nitrogen was added 1-benzyl-3-bromo-1H-1,2,4-triazole (121 mg, 0.51 mmol, as prepared for Compound S101, Step 1), Pd-PEPPSI-IPentCl 2-methylpyridine (35.6 mg, 0.04 mmol), and Cs2CO3 (414 mg, 1.27 mmol) then the reaction was heated to 100°C and stirred for 16 h under nitrogen. The mixture was cooled to rt, diluted with water (20 mL), and extracted with EtOAc (3 x 20 mL). The combined organic extracts were washed with brine (3 x 10 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 101 mg (52%) of 3-(4-(1-benzyl-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyrazine (Compound 400) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.20 (d, J = 1.2 Hz, 1H), 8.90 (d, J = 1.2 Hz, 1H), 8.33 (s, 1H), 8.22 (s, 1H), 8.01 (s, 1H), 7.83 (s, 1H), 7.42 – 7.25 (m, 5H), 5.24 (s, 2H), 3.89 (s, 3H), 3.51-3.44 (m, 3.8 Hz, 4H), 3.24-3.18 (m, 4H); MS (ESI) m/z [M+H]+ calcd. for C23H24N10, 441.2; found, 441.2; LCMS purity: 254 nm: 97.7%. Example 78: Synthesis of Exemplary Compound 401 2-benzyl-5-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrazin-3-yl)piperazin-1-yl)- 1,3,4-thiadiazole (Compound 401)
Figure imgf000293_0001
[0657] To a solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)pyrazolo[1,5- a]pyrazine (Compound S103) (60 mg, 0.21 mmol) in DMSO (2 mL) under nitrogen was added 2-benzyl-5-bromo-1,3,4-thiadiazole (64.8 mg, 0.25 mmol, as prepared in Example 55, Step 2) and DIEA (82.1 mg, 0.64 mmol) then the reaction was heated to 110°C and stirred 16 h under nitrogen. The mixture was cooled to rt, diluted with water (20 mL), and extracted with EtOAc (3 x 20 mL). The combined organic extracts were washed with brine (3 x 10 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 35.4 mg (36%) of 2-benzyl-5-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrazin-3-yl)piperazin-1-yl)- 1,3,4-thiadiazole (Compound 401) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.21 (d, J = 1.2 Hz, 1H), 8.92 (d, J = 1.2 Hz, 1H), 8.23 (s, 1H), 8.02 (s, 1H), 7.86 (s, 1H), 7.40 – 7.23 (m, 5H), 4.26 (s, 2H), 3.89 (s, 3H), 3.65 – 3.57 (m, 4H), 3.28 – 3.22 (m, 4H); MS (ESI) m/z [M+H]+ calcd. for C23H23N9S, 458.2; found, 458.2; LCMS purity: 254 nm: 98.4%. Example 79: Synthesis of Exemplary Compound 402 benzo[d]thiazol-6-ylmethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate (Compound 402)
Figure imgf000293_0002
[0658] To a solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-piperazin-1-ylpyrazolo[1,5- a]pyridine (Compound 12) (150 mg, 0.53 mmol) and 1,3-benzothiazol-6-ylmethanol (263 mg, 1.59 mmol) in pyridine (2 mL) cooled to 0°C was added triphosgene (473 mg, 1.59 mmol) then the reaction was stirred at 0°C for 1 h. The mixture was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with MeOH / DCM (1:20) and Prep-HPLC to afford 114.1 mg (45%) of benzo[d]thiazol-6- ylmethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1- carboxylate (Compound 402) as a light yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.41 (s, 1H), 8.82 (s, 1H), 8.24 – 8.18 (m, 2H), 8.10 (d, J = 8.3 Hz, 1H), 7.97 (s, 1H), 7.76 (s, 1H), 7.64 (d, J = 9.2 Hz, 1H), 7.57 (dd, J = 8.4, 1.7 Hz, 1H), 7.32 (dd, J = 9.2, 1.5 Hz, 1H), 5.27 (s, 2H), 3.86 (s, 3H), 3.61 (s, 4H), 2.98 (t, J = 5.0 Hz, 4H); MS (ESI) m/z [M+H]+ calcd. for C24H23N7O2S, 474.2; found, 474.2; LCMS purity: 254 nm: 99.6% Example 80: Synthesis of Exemplary Compound 403 3-benzyl-5-(4-(7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazin-3-yl)piperazin-1-yl)- 1,2,4-oxadiazole (Compound 403)
Figure imgf000294_0001
[0659] To a solution of 3-benzyl-1,2,4-oxadiazol-5(4H)-one (0.18 g, 1.02 mmol, as prepared in Example 44, Step 1) and PyBroP (0.48 g, 1.02 mmol) in dioxane (6 mL) was added DIEA (0.53 mL, 3.06 mmol) then the reaction was warmed to 50 °C and stirred for 2 h. 7-(1-methyl-1H-pyrazol-4-yl)-3-piperazin-1-ylimidazo[1,2-b]pyridazine trihydrochloride salt (0.20 g, 0.51 mmol, as prepared in Example 64, Step 2) was added and the reaction was stirred at 50°C overnight. The reaction was cooled to rt and concentrated under reduced pressure. The residue was partitioned between DCM (20 mL) and water (10 mL). The aqueous layer was extracted with DCM (2 x 20 mL). The combined organic extracts were washed with brine, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with 0-100% (10% MeOH in DCM) / DCM. The pure fractions were combined and concentrated under reduced pressure. The residue was triturated with hexane. The resulting solid was further purified by silica gel chromatography eluting with 5% MeOH in EtOAc to afford 29 mg (13%) of 3-[4-(3-benzyl-1,2,4-oxadiazol-5- yl)piperazin-1-yl]-7-(1-methyl-1H-pyrazol-4-yl)imidazo[1,2-b]pyridazine (Compound 403) as a yellow solid.1H NMR (400 MHz, CD3OD) d (ppm) 8.75 (d, J=2.08 Hz, 1H) 8.21 (s, 1H) 8.03 (s, 1H) 7.97 (d, J=2.20 Hz, 1H) 7.20 - 7.35 (m, 6H) 3.97 (s, 3H) 3.80 - 3.90 (m, 6H) 3.35 - 3.41 (m, 4H); MS (ESI) m/z [M+H]+ calcd. for C23H24N9O, 442.2; found, 442.1; HPLC purity: 210 nm: 95.9%; 254 nm: 95.6%. Example 81: Synthesis of Exemplary Compound 404 3-(4-(1-benzyl-1H-1,2,4-triazol-3-yl)piperazin-1-yl-2,2,3,3,5,5,6,6-d8)-6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 404)
Figure imgf000295_0001
Step 1. Preparation of tert-butyl 4-[6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl]piperazine-1-carboxylate-(d9, d10)
Figure imgf000295_0002
[0660] To a mixture of 3-bromo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound S9) (1.290 g, 4.66 mmol) and tert-butyl piperazine-1-carboxylate-d8 (0.950 g, 4.9 mmol) in tBuOH-d10 (13.1 mL) and dioxane (6.6 mL) was sparged with argon for 10 minutes was added KOtBu (784 mg, 6.98 mmol) and the mixture was sparged with argon for 5 minutes. tBuXPhos Pd G1 (0.480 g, 0.70 mmol) was added and the mixture was sparged for 5 minutes with argon, then the reaction was heated to 55°C and stirred for 2 h under argon. The mixture was cooled to rt, diluted with EtOAc, washed with water and brine, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with a gradient of MeOH in DCM to afford 1.058 g (58%) of tert-butyl 4-[6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl]piperazine-1-carboxylate-(d9 and d10 mixture). 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 0.15H), 8.21 (br. s., 0.16H), 7.97 (s, 1H), 7.77 (s, 0.89H), 7.65 (d, J = 9.29 Hz, 1H), 7.32 (d, J = 9.29 Hz, 1H), 3.87 (s, 3H), 1.43 (s, 9H). MS (ESI) m/z [M+H]+ calcd. for C20H18D9N6O2, 392.27; found, 392.1 (42% of base peak); MS (ESI) m/z [M+H]+ calcd. for C20H17D10N6O2, 393.27; found, 393.1 (base peak); HPLC purity: 210 nm: 100.0%; 254 nm: 100.0%. Step 2. Preparation of 6-(1-methyl-1H-pyrazol-4-yl)-3-piperazin-1-ylpyrazolo[1,5- a]pyridine-(d9, d10)
Figure imgf000296_0001
[0661] To a mixture of tert-butyl 4-[6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin- 3-yl]piperazine-1-carboxylate-(d9,d10 ) (0.950 g, 2.43 mmol, as prepared in the previous step) in EtOAc (20 mL) warmed to 35°C was slowly added conc. HCl (1.01 mL, 12.2 mmol) then the mixture was stirred at 35°C for 1 h. The mixture was cooled to rt, diluted with EtOAc (40 mL) and stirred for 10 minutes. The solids were collected by filtration and washed with MTBE (2 X 20 mL). The solids were partially dissolved in a solution of 2N NaOH (6 mL) and water (10 mL) and extracted several times with DCM. The combined organic extracts were washed with water, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure to afford 660 mg (93%) of 4-[6-(1-methyl- 1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl]piperazine-1-carboxylate-(d9, d10) as a yellow solid. 1H NMR (400 MHz, CDCl3) δ (ppm) 8.47 (s, 0.14H), 7.75 (s, 1H), 7.72 (s, 0.83H), 7.62 (s, 0.16H), 7.53 (d, J = 9.17 Hz, 1H), 7.28 (s, 1H), 7.13 (d, J = 9.17 Hz, 1H), 3.99 (s, 3H); HPLC purity: 210 nm: 100.0%; 254 nm: 100.0%. Step 3. Preparation of 3-[4-(1-benzyl-1H-1,2,4-triazol-3-yl)piperazin-1-yl]-6-(1-methyl- 1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine-d8 (Compound 404)
Figure imgf000296_0002
[0662] A mixture of 6-(1-methyl-1H-pyrazol-4-yl)-3-piperazin-1-ylpyrazolo[1,5- a]pyridine-(d9,d10) (0.650 g, 2.24 mmol, as prepared in the previous step) and 1-benzyl-3- bromo-1H-1,2,4-triazole (0.640 g, 2.69 mmol, as prepared for Compound S101, Step 1) in tBuOH (9.8 mL), dioxane (1.5 mL), THF (3.0 mL), and water (0.032 mL, 1.79 mmol) was sparged with argon for 5 minutes then KOtBu (0.377 g, 3.36 mmol) and tBuXPhos Pd G1 (0.154 g, 0.22 mmol) were added. The mixture was sparged with argon for 5 minutes then the reaction was heated to 50°C under argon and stirred for 2 h. Additional KOtBu (0.126 g, 1.12 mmol), tBuXPhos Pd G1 (0.154 g, 0.22 mmol) and water (0.032 mL, 1.79 mmol) were added, and the reaction was stirred under argon at 55°C overnight. The mixture was cooled to rt, diluted with EtOAc, and washed with water and brine. The organic layer was dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was suspended in a mixture of tBuOH (9.8 mL), dioxane (1.5 mL), THF (3.0 mL), and water (0.032 mL, 1.79 mmol) and sparged with argon for 10 minutes. To the mixture was added KOtBu (0.377 g, 3.36 mmol) and tBuXPhos Pd G1 (0.154 g, 0.22 mmol) then the mixture was sparged with argon for 5 minutes, heated to 55°C, and stirred overnight. The mixture was cooled to rt, diluted with EtOAc, washed with water and brine, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel chromatography eluting with a gradient of MeOH in DCM to afford 680 mg (66%) of 3-[4-(1-benzyl-1H-1,2,4- triazol-3-yl)piperazin-1-yl]-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine-d8 (Compound 404) as a yellow solid.1H NMR (400 MHz, CDCl3) δ (ppm) 8.48 (s, 1H), 7.76 (s, 1H), 7.74 (d, J = 4.28 Hz, 2H), 7.63 (s, 1H), 7.55 (d, J = 9.29 Hz, 1H), 7.35 - 7.44 (m, 3H), 7.29 - 7.33 (m, 2H), 7.15 (dd, J = 1.35, 9.17 Hz, 1H), 5.21 (s, 2H), 3.99 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C24H18D8N9, 448.3; found, 448.2; HPLC purity: 210 nm: 100.0%; 254 nm: 100.0%. Example 82: Synthesis of Exemplary Compound 405 benzo[d]thiazol-5-ylmethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate (Compound 405)
Figure imgf000297_0001
[0663] To a solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)pyrazolo[1,5- a]pyridine (Compound 12) (250 mg, 0.89 mmol) and benzo[d]thiazol-5-ylmethanol (175.5 mg, 1.01 mmol) in pyridine (10 mL) were added triphosgene (106.2 mg, 0.36 mmol) then the reaction was stirred at rt for 16 h under nitrogen. The mixture was concentrated under reduced pressure then the residue was purified by Prep-TLC (MeOH / DCM 1:10) and Prep-HPLC to afford 161.5 mg (38%) of benzo[d]thiazol-5-ylmethyl 4-(6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 405) as a green solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.43 (s, 1H), 8.81 (t, J = 1.3 Hz, 1H), 8.23 – 8.16 (m, 2H), 8.11 (d, J = 1.5 Hz, 1H), 7.97 (s, 1H), 7.77 (s, 1H), 7.65 (dd, J = 9.2, 1.0 Hz, 1H), 7.54 (dd, J = 8.3, 1.6 Hz, 1H), 7.33 (dd, J = 9.2, 1.6 Hz, 1H), 5.30 (s, 2H), 3.87 (s, 3H), 3.62 (s, 4H), 2.98 (t, J = 5.0 Hz, 4H); MS (ESI) m/z [M+H]+ calcd. for C24H23N7O2S, 474.2; found, 474.1;LCMS purity: 254 nm: 99.7% Example 83: Synthesis of Exemplary Compound 406 N-(benzo[d]thiazol-6-ylmethyl)-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxamide (Compound 406)
Figure imgf000298_0001
[0664] To a solution of 2-(benzo[d]thiazol-6-yl)acetic acid (200 mg, 1.04 mmol) and DPPA (341.8 mg, 1.24 mmol) in THF (10 mL) was added TEA (314.2 mg, 3.11 mmol) dropwise at rt. The resulting mixture was stirred at rt for 5 h then concentrated under reduced pressure. The residue was dissolved in toluene (20 mL) then 6-(1-methyl-1H- pyrazol-4-yl)-3-(piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 12) (155.3 mg, 0.55 mmol) was added followed by dropwise addition of TEA (166.9 mg, 1.65 mmol) at rt. The reaction was warmed to 110°C and stirred overnight. The mixture was cooled to rt and concentrated under reduced pressure. The residue was purified by silica gel chromatography eluting with MeOH / DCM (1:40) and Prep-HPLC to afford 142.7 mg (55%) of N-(benzo[d]thiazol-6-ylmethyl)-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazine-1-carboxamide (Compound 406) as a light yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.34 (s, 1H), 8.82 (s, 1H), 8.23 (s, 1H), 8.06 – 8.01 (m, 2H), 7.97 (s, 1H), 7.77 (s, 1H), 7.67 (d, J = 9.2 Hz, 1H), 7.48 (dd, J = 8.4, 1.6 Hz, 1H), 7.35 – 7.28 (m, 2H), 4.42 (d, J = 5.6 Hz, 2H), 3.87 (s, 3H), 3.58 – 3.51 (m, 4H), 3.00 – 2.93 (m, 4H); MS (ESI) m/z [M+H]+ calcd. for C24H24N8OS, 473.2; found, 473.2; LCMS purity: 254 nm: 99.9% Example 84: Synthesis of Exemplary Compound 250 6-(1-methyl-1H-pyrazol-4-yl)-3-{4-[5-(pyridine-2-ylmethyl)pyrimidin-2-yl]piperazin-1- yl}pyrazolo[1,5-a]pyridine (Compound 250)
Figure imgf000299_0001
[0665] A mixture of 6-(1-methyl-1H-pyrazol-4-yl)-3-piperazin-1-pyrazolo[1,5-a]pyridine hydrochloride (Compound S19) (92 mg, 0.29 mmol), 2-chloro-5-(pyridine-2- ylmethyl)pyrimidine (Compound S80) (70 mg, 0.34 mmol), K2CO3 (123 mg, 0.892 mmol), and NMP (3.7 mL) was sparged for 1 min with argon. The reaction was heated at 110°C for 2 h then cooled to rt and partitioned between water and DCM. The organic layer was washed with brine, dried over anhydrous Na2SO4, and filtered. The Filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (IPA/DCM) then further purified by C18 reverse-phase silica gel flash chromatography (ACN/water) to yield 30 mg (23%) of 6-(1-methyl-1H-pyrazol-4-yl)-3- {4-[5-(pyridine-2-ylmethyl)pyrimidin-2-yl]piperazin-1-yl}pyrazolo[1,5-a]pyridine (Compound 250) as a solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.81 (s, 1H), 8.48 (m, 1H), 8.34 (s, 2H), 8.22 (s, 1H), 7.97 (s, 1H), 7.77 (s, 1H), 7.71 (m, 2H), 7.32 (m, 2H), 7.22 (dd, J=6.9, 5.3 Hz, 1H), 3.94 (s, 2H), 3.89 (m, 4H), 3.87 (s, 3H), 3.04 (m, 4H). MS (ESI) m/z [M+H]+ calcd. for C25H26N9: 452.2; found, 452.3; HPLC purity: 210 nm: 100.0%; 254 nm: 100.0%. Example 85: Synthesis of Exemplary Compound 407 3-(4-(1-(4-fluorobenzyl)-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyrimidine (Compound 407)
Figure imgf000299_0002
[0666] To a solution of 3-chloro-1-[(4-fluorophenyl)methyl]-1,2,4-triazole (194.1 mg, 0.91 mmol, as prepared in Example 49, Step 1) and 1-[6-(1-methylpyrazol-4-yl)pyrazolo[1,5- a]pyrimidin-3-yl]piperazine (Compound S98) (200 mg, 0.706 mmol) in dioxane (20 mL) were added Cs2CO3 (460.5 mg, 1.41 mmol) and Pd-PEPPSI-IPentCl 2-methylpyridine (59.3 mg, 0.07 mmol) then the mixture was stirred overnight at 100°C under nitrogen. The reaction was cooled to rt and concentrated under reduced pressure. The residue was dissolved in water (50 mL) then extracted with DCM (3 x 50 mL). The combined organic extracts were dried over anhydrous Na2SO4 and filtered, then the filtrate was concentrated under reduced pressure. The residue was purified by Prep-HPLC to afford 12.9 mg (4%) of 3-(4-(1-(4-fluorobenzyl)-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyrimidine (Compound 407) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.18 (d, J = 2.1 Hz, 1H), 8.63 (d, J = 2.1 Hz, 1H), 8.31-8.28 (m, 2H), 8.03 (s, 1H), 7.90 (s, 1H), 7.44 – 7.26 (m, 2H), 7.26 – 7.12 (m, 2H), 5.22 (s, 2H), 3.89 (s, 3H), 3.47-3.42 (m, 4H), 3.32-3.29 (m, 4H); MS (ESI) m/z [M+H]+ calcd. for C23H23FN10, 459.2; found, 459.20; HPLC purity: 254 nm:99.6%. [0667] Using the procedures described in Example 85 and reagents, starting materials, and conditions known to those skilled in the art, the following compounds representative of the present disclosure were prepared:
Figure imgf000300_0001
Figure imgf000301_0002
Example 86: Synthesis of Exemplary Compound 408 (2-methylbenzo[d]thiazol-6-yl)methyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazine-1-carboxylate (Compound 408)
Figure imgf000301_0001
[0668] To a stirred solution of (2-methylbenzo[d]thiazol-6-yl)methanol (Compound S105) (229 mg, 1.28 mmol) and 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)pyrazolo[1,5- a]pyridine (Compound 12) (180.4 mg, 0.64 mmol) in pyridine (5 mL) cooled to 0°C was added BTC (379.1 mg, 1.28 mmol) then the mixture was warmed to rt and stirred for 16 h. The reaction was concentrated under reduced pressure then the residue was purified by Prep-TLC (PE/EtOAc 1:100) and then further purified by Prep-HPLC to afford 123.9 mg (20%) of (2-methylbenzo[d]thiazol-6-yl)methyl 4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 408) as an off-white solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.22 (s, 1H), 8.06 (d, J = 1 Hz, 1H), 7.97 (s, 1H), 7.91 (d, J = 8.4 Hz, 1H), 7.76 (s, 1H), 7.64 (d, J = 9.2 Hz, 1H), 7.49 (dd, J = 8.4, 1.6 Hz, 1H), 7.32 (dd, J = 9.3, 1.4 Hz, 1H), 5.23 (s, 2H), 3.86 (s, 3H), 3.70 – 3.51 (m, 4H), 2.97 (t, J = 4.7 Hz, 4H), 2.80 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C25H25N7O2S, 488.1; found, 488.2; HPLC purity: 254 nm: 99.3%. [0669] Using the procedures described in Example 86 and reagents, starting materials, and conditions known to those skilled in the art, the following compounds representative of the present disclosure were prepared:
Figure imgf000302_0001
Figure imgf000303_0001
Figure imgf000304_0001
Example 87: Synthesis of Exemplary Compound 409 3-(4-(5-benzyl-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine (Compound 409)
Figure imgf000305_0001
Step 1. Preparation of 3-(4-(5-bromo-1-(tetrahydro-2H-pyran-2-yl)-1H-1,2,4-triazol-3- yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine
Figure imgf000305_0002
[0670] To a stirred solution of 3,5-dibromo-1-(tetrahydro-2H-pyran-2-yl)-1H-1,2,4- triazole (700 mg, 2.25 mmol) and 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1- yl)pyrazolo[1,5-a]pyridine (Compound 12) (572 mg, 2.03 mmol) in IPA (5 mL) was added DIEA (581.9 mg, 4.50 mmol) at rt then the mixture was heated to 100°C and stirred for 16 h. The reaction was cooled to rt and concentrated under reduced pressure. The residue was purified by silica gel column chromatography eluting with PE/EtOAc (1:100) to afford 600 mg (52%) of 3-(4-(5-bromo-1-(tetrahydro-2H-pyran-2-yl)-1H-1,2,4-triazol-3-yl)piperazin- 1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.23 (s, 1H), 7.97 (s, 1H), 7.80 (s, 1H), 7.69 (d, J = 9.2 Hz, 1H), 7.33 (dd, J = 9.2, 1.3 Hz, 1H), 5.28 (dd, J = 9.7, 2.3 Hz, 1H), 4.00 – 3.93 (m, 1H), 3.87 (s, 3H), 3.71 – 3.57 (m, 1H), 3.53 – 3.45 (m, 2H), 3.39 – 3.33 (m, 2H), 3.25 – 3.17 (m, 2H), 3.16 – 3.06 (m, 2H), 2.24 – 2.06 (m, 1H), 1.98 – 1.89 (m, 1H), 1.87 – 1.77 (m, 1H), 1.75 – 1.61 (m, 1H), 1.58 – 1.48 (m, 2H); MS (ESI) m/z [M+H]+ calcd. for C22H26BrN9O, 512.0; found, 512.1. Step 2. Preparation of 3-(4-(5-benzyl-1-(tetrahydro-2H-pyran-2-yl)-1H-1,2,4-triazol-3- yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine
Figure imgf000306_0001
[0671] To a stirred solution of 3-(4-(5-bromo-1-(tetrahydro-2H-pyran-2-yl)-1H-1,2,4- triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (300 mg, 0.58 mmol, as prepared in the previous step) and potassium benzyltrifluoroborate (231.9 mg, 1.17 mmol) in toluene (5 mL) and H2O (1 mL) were added Pd-PEPPSI-IPentCl 2-methylpyridine (49.25 mg, 0.06 mmol) and K3PO4 (497.1 mg, 2.34 mmol) at rt then the mixture was heated to 80°C and stirred for 16 h under nitrogen. The reaction was cooled to rt and concentrated under reduced pressure. The residue was purified by Prep-TLC (DCM/MeOH 20:1) to afford 200 mg (32%) of 3-(4-(5-benzyl-1-(tetrahydro-2H-pyran-2- yl)-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridine as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.78 (s, 1H), 7.67 (d, J = 9.2 Hz, 1H), 7.39 – 7.12 (m, 6H), 5.21 (dd, J = 9.9, 2.0 Hz, 1H), 4.01 – 3.92 (m, 1H), 3.90 – 3.82 (m, 5H), 3.67 – 3.55 (m, 1H), 3.50 – 3.39 (m, 2H), 3.30 – 3.06 (m, 6H), 2.30 – 2.13 (m, 1H), 2.03 – 1.88 (m, 1H), 1.83 – 1.74 (m, 1H), 1.72 – 1.60 (m, 1H), 1.59 – 1.46 (m, 2H); MS (ESI) m/z [M+H]+ calcd. for C29H33N9O, 524.0; found, 524.1. Step 3. Preparation of 3-(4-(5-benzyl-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl- 1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 409)
Figure imgf000306_0002
[0672] To a solution of 4M HCl in dioxane (5 mL) was added 3-(4-(5-benzyl-1- (tetrahydro-2H-pyran-2-yl)-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridine (190 mg, 0.36 mmol, as prepared in the previous step) at rt then the mixture was stirred for 3 h at rt. The reaction was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 53.4 mg (33%) of 3-(4-(5- benzyl-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridine (Compound 409) as a light yellow solid.1H NMR (400 MHz, DMSO-d6) δ 13.01 – 12.20 (m, 1H), 8.82 (s, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.77 (s, 1H), 7.66 (d, J = 9.2 Hz, 1H), 7.40 – 7.12 (m, 6H), 4.01 – 3.74 (m, 5H), 3.55 – 3.39 (m, 4H), 3.06 (t, J = 4.7 Hz, 4H); MS (ESI) m/z [M+H]+ calcd. for C24H25N9, 440.0; found, 440.2; HPLC purity: 254 nm: 99.7%. Example 88: Synthesis of Exemplary Compound 410 4-(1-benzyl-1H-1,2,4-triazol-3-yl)-1-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)-1,4-diazepan-5-one (Compound 410)
Figure imgf000307_0001
[0673] To a solution of 1-[6-(1-methylpyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl]-1,4- diazepan-5-one (300 mg, 0.967 mmol, as prepared in Example 18, Step 4) and 1-benzyl-3- bromo-1H-1,2,4-triazole (276.1 mg, 1.160 mmol, as prepared in Compound S101, Step 1) in dioxane (20 mL) were added Cs2CO3 (787.3 mg, 2.417 mmol), EPhos Pd G4 (133.1 mg, 0.145 mmol), and EPhos (77.5 mg, 0.145 mmol) then the mixture was heated to 100°C and stirred overnight under nitrogen. The reaction was cooled to rt and concentrated under reduced pressure. The crude product was purified by Prep-HPLC to afford 48.1 mg (11%) of 4-(1-benzyl-1H-1,2,4-triazol-3-yl)-1-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)-1,4-diazepan-5-one (Compound 410) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (s, 1H), 8.60 (s, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.84 (s, 1H), 7.62 (d, J = 9.2 Hz, 1H), 7.42 – 7.28 (m, 6H), 5.37 (s, 2H), 4.01 – 3.95 (m, 2H), 3.86 (s, 3H), 3.31 – 3.27 (m, 2H), 3.27 – 3.23 (m, 2H), 2.94 – 2.88 (m, 2H); MS (ESI) m/z [M+H]+ calcd. for C25H25N9O, 468.2; found, 468.2; HPLC purity: 254 nm: 99.6%. Example 89: Synthesis of Exemplary Compound 411 4-(5-benzylpyrimidin-2-yl)-1-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin-3-yl)- 1,4-diazepan-5-one (Compound 411)
Figure imgf000308_0001
Step 1. Preparation of 6-(1-methyl-1H-pyrazol-4-yl)-3-nitropyrazolo[1,5-a]pyrimidine
Figure imgf000308_0002
[0674] To a stirred solution of 6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidine (10 g, 50.2 mmol, as prepared in Compound S10, Step 1) in H2SO4 (60 mL) cooled to 0°C was added KNO3 (5.58 g, 55.22 mmol) then the mixture was warmed to rt and stirred for 16 h. The reaction was diluted with water then the solids were isolated by filtration and the filter cake was washed with water (3x60 mL). The filter cake was triturated with saturated aqueous NaHCO3 solution then the solids were isolated by filtration and the filter cake was washed with water (3x60 mL). The solids were triturated with water (200 mL), filtered, and dried to afford 8.8 g (72%) of 6-(1-methyl-1H-pyrazol-4-yl)-3-nitropyrazolo[1,5- a]pyrimidine as a red solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.70 (d, J = 2.2 Hz, 1H), 9.34 (d, J = 2.2 Hz, 1H), 9.04 (s, 1H), 8.45 (s, 1H), 8.16 (d, J = 0.5 Hz, 1H), 3.92 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C10H8N6O2, 245.1; found, 245.2. Step 2. Preparation of 6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin-3-amine
Figure imgf000308_0003
[0675] To a stirred solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-nitropyrazolo[1,5- a]pyrimidine (8.8 g, 36.03 mmol, as prepared in the previous step) in conc. HBr (100 mL) cooled to 0°C was added SnCl2.2H2O (24.61 g, 108.1 mmol) then the mixture was heated to 90°C and stirred for 16 h. The reaction was cooled to rt and diluted with water (200 mL), then the pH of the solution was adjusted to pH 8 with 6N NaOH. The mixture was extracted with DCM (3x80 mL) then the organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure to afford 5.6 g (72%) of 6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin-3-amine as a red solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.03 (d, J = 2.1 Hz, 1H), 8.50 (d, J = 2.1 Hz, 1H), 8.27 (s, 1H), 8.00 (d, J = 0.7 Hz, 1H), 7.66 (s, 1H), 4.30 (s, 2H), 3.88 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C10H10N6, 215.1; found, 215.2. Step 3. Preparation of ethyl 3-((6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin-3- yl)amino)propanoate
Figure imgf000309_0001
[0676] To a stirred solution of 6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin-3- amine (8 g, 37.34 mmol, as prepared in the previous step) and ethyl 3-iodopropanoate (12.77 g, 56.01 mmol) in DMF (150 mL) was added Al2O3 (7.61 g, 74.69 mmol) at rt then the mixture was heated to 55°C and stirred for 1 h. The reaction was cooled to rt, diluted with water, and the pH was adjusted to pH 8 with saturated aqueous NaHCO3 solution. The resulting mixture was extracted with DCM (5x80 mL) then the organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by prep HPLC to afford 3.3 g (28%) of ethyl 3-((6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin-3-yl)amino)propanoate as a red solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.07 (d, J = 2.1 Hz, 1H), 8.53 (d, J = 2.1 Hz, 1H), 8.27 (s, 1H), 8.01 (s, 1H), 7.75 (s, 1H), 4.74 (t, J = 6.7 Hz, 1H), 4.06 (q, J = 7.1 Hz, 2H), 3.88 (s, 3H), 3.41 (q, J = 6.8 Hz, 2H), 2.59 (t, J = 6.8 Hz, 2H), 1.17 (t, J = 7.1 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C15H18N6O2, 315.1; found, 315.2. Step 4. Preparation of a mixture of ethyl 3-((2-((tert-butoxycarbonyl)amino)ethyl)(6-(1- methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin-3-yl)amino)propanoate and methyl 3- ((2-((tert-butoxycarbonyl)amino)ethyl)(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrimidin-3-yl)amino)propanoate
Figure imgf000309_0002
[0677] To a stirred solution of ethyl 3-((6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrimidin-3-yl)amino)propanoate (500 mg, 1.59 mmol, as prepared in the previous step) and tert-butyl (2-oxoethyl)carbamate (3.29 g, 20.68 mmol) in MeOH (15 mL) was added NaHCO3 (267.24 mg, 3.18 mmol) at rt then the mixture was heated to 60°C and stirred for 16 h. The reaction was cooled to 0°C, then NaBH3CN (149.93 mg, 2.39 mmol) was added in portions to the stirred mixture. The reaction was stirred for 10 min at 0°C then AcOH (9.55 mg, 0.16 mmol) was added. The resulting mixture was warmed to rt, stirred for 16 h, then concentrated under reduced pressure. The residue was purified by silica gel column chromatography eluting with PE/EtOAc (1:100) to afford 428 mg (59%) of a mixture of ethyl 3-((2-((tert-butoxycarbonyl)amino)ethyl)(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyrimidin-3-yl)amino)propanoate and methyl 3-((2-((tert- butoxycarbonyl)amino)ethyl)(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin-3- yl)amino)propanoate as a red solid. MS (ESI) m/z [M+H]+ calcd. for C22H31N7O4, 458.2; found, 458.5; C21H29N7O4, 444.2; found, 444.5. Step 5. Preparation of a mixture of ethyl 3-((2-aminoethyl)(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyrimidin-3-yl)amino)propanoate hydrochloride and methyl 3-((2- aminoethyl)(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin-3- yl)amino)propanoate hydrochloride
Figure imgf000310_0001
[0678] To a stirred solution of a mixture of ethyl 3-((2-((tert- butoxycarbonyl)amino)ethyl)(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin-3- yl)amino)propanoate and methyl 3-((2-((tert-butoxycarbonyl)amino)ethyl)(6-(1-methyl- 1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin-3-yl)amino)propanoate (1.5 g, 3.278 mmol, as prepared in the previous step) in MeOH (10 mL) cooled to 0°C was added a 4M HCl solution in dioxane (15 mL) at 0°C then the mixture was warmed to rt and stirred for 1 h, then concentrated under reduced pressure to afford 1.2 g of a crude mixture of ethyl 3-((2- aminoethyl)(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin-3- yl)amino)propanoate hydrochloride and methyl 3-((2-aminoethyl)(6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin-3-yl)amino)propanoate hydrochloride as a white solid. The crude product was used in the next step directly without further purification. MS (ESI) m/z [M+H]+ calcd. for C17H23N7O2, 358.2; found, 358.4; C16H21N7O2, 344.2; found, 344.4. Step 6. Preparation of 1-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin-3-yl)- 1,4-diazepan-5-one
Figure imgf000311_0001
[0679] To a stirred solution of a mixture of ethyl 3-((2-aminoethyl)(6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin-3-yl)amino)propanoate hydrochloride and methyl 3-((2-aminoethyl)(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin-3- yl)amino)propanoate hydrochloride (1.2 g, 3.36 mmol, as prepared in the previous step) in MeOH (15 mL) was added TBD (934.72 mg, 6.71 mmol) at rt then the mixture was heated to 80°C and stirred for 16 h. The reaction was cooled to rt, concentrated under reduced pressure, and the residue was dissolved in water. The resulting mixture was extracted with DCM (3x60 mL) then the organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-TLC (DCM/MeOH 15:1) to afford 400 mg (38%) of 1-(6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin-3-yl)-1,4-diazepan-5-one as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.16 (d, J = 2.1 Hz, 1H), 8.61 (d, J = 2.1 Hz, 1H), 8.28 (s, 1H), 8.02 (s, 1H), 7.89 (s, 1H), 7.69 – 7.58 (m, 1H), 3.89 (s, 3H), 3.55 – 3.49 (m, 2H), 3.49 – 3.44 (m, 2H), 3.29 – 3.23 (m, 2H), 2.61 – 2.55 (m, 2H); MS (ESI) m/z [M+H]+ calcd. for C15H17N7O, 312.1; found, 312.3. Step 7. Preparation of 4-(5-benzylpyrimidin-2-yl)-1-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyrimidin-3-yl)-1,4-diazepan-5-one (Compound 411)
Figure imgf000311_0002
[0680] To a stirred solution of 1-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin- 3-yl)-1,4-diazepan-5-one (160 mg, 0.51 mmol, as prepared in the previous step) and 5- benzyl-2-chloropyrimidine (Compound S88) (115.69 mg, 0.56 mmol) in dioxane (5 mL) were added Pd2(dba)3 (70.59 mg, 0.08 mmol), XantPhos (44.60 mg, 0.08 mmol) and K3PO4 (218.16 mg, 1.03 mmol) at rt then the mixture was heated to 100°C and stirred for 16 h under nitrogen. The reaction was cooled to rt and concentrated under reduced pressure. The residue was dissolved in water (20 mL) and extracted with DCM (3x50 mL). The organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-TLC (DCM/MeOH 15:1) and Prep-HPLC to afford 112 mg (45%) of 4-(5-benzylpyrimidin-2- yl)-1-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin-3-yl)-1,4-diazepan-5-one (Compound 411) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.18 (d, J = 2.1 Hz, 1H), 8.70 (s, 2H), 8.64 (d, J = 2.1 Hz, 1H), 8.29 (s, 1H), 8.03 (s, 1H), 7.94 (s, 1H), 7.37 – 7.19 (m, 5H), 4.20 – 4.09 (m, 2H), 3.99 (s, 2H), 3.89 (s, 3H), 3.68 – 3.58 (m, 4H), 2.98 – 2.86 (m, 2H); MS (ESI) m/z [M+H]+ calcd. for C26H25N9O, 480.2; found, 480.2; HPLC purity: 254 nm: 99.9%. Example 90: Synthesis of Exemplary Compound 412 4-(1-benzyl-1H-1,2,4-triazol-3-yl)-1-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrimidin-3-yl)-1,4-diazepan-5-one (Compound 412)
Figure imgf000312_0001
[0681] To a stirred solution of 1-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin- 3-yl)-1,4-diazepan-5-one (200 mg, 0.64 mmol, as prepared in Example 89, Step 6) and 1- benzyl-3-bromo-1H-1,2,4-triazole (305.88 mg, 1.28 mmol, as prepared in Compound S101, Step 1) in dioxane (6 mL) were added EPhos Pd G4 (100.31 mg, 0.11 mmol), EPhos (58.4 mg, 0.11 mmol) and Cs2CO3 (523.24 mg, 1.60 mmol) at rt then the mixture was heated to 100°C and stirred for 16 h under nitrogen. The reaction was cooled to rt, then filtered, and the filter cake was washed with DCM (3x30 mL). The filtrate was concentrated under reduced pressure then the residue was purified by Prep-TLC (DCM/MeOH 15:1) and Prep-HPLC to afford 28.9 mg (10%) of 4-(1-benzyl-1H-1,2,4-triazol-3-yl)-1-(6-(1-methyl- 1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidin-3-yl)-1,4-diazepan-5-one (Compound 412) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.18 (d, J = 2.1 Hz, 1H), 8.64 (d, J = 2.1 Hz, 1H), 8.59 (s, 1H), 8.29 (s, 1H), 8.03 (s, 1H), 7.93 (s, 1H), 7.44 – 7.25 (m, 5H), 5.37 (s, 2H), 4.00 – 3.92 (m, 2H), 3.89 (s, 3H), 3.64 – 3.55 (m, 4H), 2.92 – 2.82 (m, 2H); MS (ESI) m/z [M+H]+ calcd. for C24H24N10O, 469.2; found, 469.2; HPLC purity: 254 nm: 99.5%. Example 91: Synthesis of Exemplary Compound 413 and Compound 414 3-(4-(5-benzyl-1-methyl-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine (Compound 413) and 3-(4-(3-benzyl-1-methyl-1H-1,2,4-triazol-5- yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 414)
Figure imgf000313_0001
Step 1. Preparation of a mixture of 5-benzyl-3-bromo-1-methyl-1H-1,2,4-triazole and 3- benzyl-5-bromo-1-methyl-1H-1,2,4-triazole
Figure imgf000313_0002
[0682] To a solution of 3,5-dibromo-1-methyl-1H-1,2,4-triazole (2 g, 8.30 mmol) and potassium benzyltrifluoroborate (1.32 g, 6.64 mmol) in a mixture of toluene (15 mL) and H2O (3 mL) was added K3PO4 (3.52 g, 16.61 mmol) and Pd-PEPPSI-IPentCl 2- methylpyridine (0.35 g, 0.42 mmol) then the mixture was heated to 80°C and stirred for 16 h under nitrogen. The reaction was cooled to rt, the organic layer was separated, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 240 mg (6%) of a mixture of 5-benzyl-3- bromo-1-methyl-1H-1,2,4-triazole and 3-benzyl-5-bromo-1-methyl-1H-1,2,4-triazole as a colorless oil. MS (ESI) m/z [M+H]+ calcd. for C10H10BrN3, 252.0; found, 252.1. Step 2. Preparation of 3-(4-(5-benzyl-1-methyl-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6- (1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 413) and 3-(4-(3-benzyl- 1-methyl-1H-1,2,4-triazol-5-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine (Compound 414)
Figure imgf000314_0001
[0683] To a solution of a mixture of 5-benzyl-3-bromo-1-methyl-1H-1,2,4-triazole and 3- benzyl-5-bromo-1-methyl-1H-1,2,4-triazole (240 mg, 0.95 mmol, as prepared in the previous step) and 1-[6-(1-methylpyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl]piperazine (Compound 12) (269 mg, 0.95 mmol) in dioxane (5 mL) were added Cs2CO3 (620 mg, 1.90 mmol) and Pd-PEPPSI-IPentCl methylpyridine (80 mg, 0.01 mmol) then the mixture was heated to 90°C and stirred for 6 h under nitrogen. The reaction was cooled to rt, then concentrated under reduced pressure. The residue was purified by silica gel column chromatography eluting with DCM/MeOH (15:1) then by Prep-HPLC to afford 90.8 mg (21%) of 3-(4-(5-benzyl-1-methyl-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 413) as a green solid and 63 mg (15%) of 3-(4-(3-benzyl-1-methyl-1H-1,2,4-triazol-5-yl)piperazin-1-yl)-6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 414) as a light green solid. [0684] 3-(4-(5-benzyl-1-methyl-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 413).1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.81 (s, 1H), 8.22 (s, 1H), 7.96 (s, 1H), 7.77 (s, 1H), 7.65 (d, J = 9.2 Hz, 1H), 7.37 – 7.27 (m, 3H), 7.31 – 7.19 (m, 3H), 4.07 (s, 2H), 3.87 (s, 3H), 3.62 (s, 3H), 3.47 – 3.40 (m, 4H), 3.06 (t, J = 5.0 Hz, 4H); MS (ESI) m/z [M+H]+ calcd. for C25H27N9, 454.2; found, 454.2; HPLC purity: 254 nm: 99.3%: [0685] 3-(4-(3-benzyl-1-methyl-1H-1,2,4-triazol-5-yl)piperazin-1-yl)-6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 414).1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.79 (s, 1H), 7.66 (d, J = 9.3 Hz, 1H), 7.32 (dd, J = 9.3, 1.5 Hz, 1H), 7.30 – 7.25 (m, 4H), 7.25 – 7.15 (m, 1H), 3.87 (s, 3H), 3.83 (s, 2H), 3.63 (s, 3H), 3.31 – 3.24 (m, 4H), 3.17 – 3.10 (m, 4H); MS (ESI) m/z [M+H]+ calcd. for C25H27N9, 454.2; found, 454.2; HPLC purity: 254 nm: 99.9%. Example 92: Synthesis of Exemplary Compound 415 3-(4-(1-(4-chlorobenzyl)-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine (Compound 415)
Figure imgf000315_0001
[0686] To a solution of 3-(4-(1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound S102) (100 mg, 0.28 mmol) and 1- (bromomethyl)-4-chlorobenzene (88.2 mg, 0.42 mmol) in DMF (10 mL) was added K2CO3 (158.2 mg, 1.14 mmol) then the mixture was stirred overnight at rt under nitrogen. The reaction was concentrated under reduced pressure then the residue was purified by Prep- HPLC to afford 52.9 mg (39%) of 3-(4-(1-(4-chlorobenzyl)-1H-1,2,4-triazol-3- yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 415) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.32 (s, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.76 (s, 1H), 7.65 (d, J = 8.4 Hz, 1H), 7.50 – 7.38 (m, 2H), 7.36 – 7.26 (m, 3H), 5.23 (s, 2H), 3.86 (s, 3H), 3.52 – 3.40 (m, 4H), 3.06 (t, J = 4.9 Hz, 4H); MS (ESI) m/z [M+H]+ calcd. for C24H24ClN9, 474.2; found, 474.2; HPLC purity:254 nm:99.4%. Example 93: Synthesis of Exemplary Compound 419 3-(4-(1-(4-fluorobenzyl)-5-methyl-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 419)
Figure imgf000315_0002
Step 1. Preparation of a mixture of 5-bromo-1-(4-fluorobenzyl)-3-methyl-1H-1,2,4-triazole formic acid salt and 3-bromo-1-(4-fluorobenzyl)-5-methyl-1H-1,2,4-triazole formic acid salt
Figure imgf000316_0001
[0687] To a solution of 3-bromo-5-methyl-1H-1,2,4-triazole (2 g, 12.34 mmol) and 1- (bromomethyl)-4-fluorobenzene (2.8 g, 14.81 mmol) in DMF (20 mL) was added K2CO3 (3.41 g, 24.69 mmol) then the mixture was stirred at for 16 h under nitrogen. The reaction was concentrated under reduced pressure then the residue was purified by reversed-phase column chromatography using a C18 silica gel column eluting with a 10-100% gradient of ACN/water (0.1% FA) to afford 2.1 g (51%) of a mixture of 5-bromo-1-(4-fluorobenzyl)- 3-methyl-1H-1,2,4-triazole formic acid salt and 3-bromo-1-(4-fluorobenzyl)-5-methyl-1H- 1,2,4-triazole formic acid salt as a white solid. MS (ESI) m/z [M+H]+ calcd. for C10H10BrFN3, 270.0; found, 270.1. Step 2. 3-(4-(1-(4-fluorobenzyl)-5-methyl-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1- methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 419)
Figure imgf000316_0002
[0688] To a solution of the mixture of 5-bromo-1-(4-fluorobenzyl)-3-methyl-1H-1,2,4- triazole formic acid salt and 3-bromo-1-(4-fluorobenzyl)-5-methyl-1H-1,2,4-triazole formic acid salt (351.1 mg, 1.11 mmol, as prepared in the previous step) and 1-[6-(1- methylpyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl]piperazine (Compound 12) (531.6 mg, 1.88 mmol) in dioxane (30.00 mL) were added Cs2CO3 (1.02 g, 3.13 mmol) and Pd- PEPPSI-IPentCl 2-methylpyridine (132 mg, 0.16 mmol) then the mixture was heated to 90°C and stirred for 16 h under nitrogen. The reaction was cooled to rt and concentrated under reduced pressure. The residue was purified by Prep-TLC (DCM/MeOH 10:1) and by Prep-HPLC to afford 125 mg (17%) of 3-(4-(1-(4-fluorobenzyl)-5-methyl-1H-1,2,4- triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 419) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (t, J = 1.3 Hz, 1H), 8.22 (s, 1H), 7.97 (d, J = 0.9 Hz, 1H), 7.77 (s, 1H), 7.68 –7.61 (m, 1H), 7.35 – 7.24 (m, 3H), 7.24 – 7.14 (m, 2H), 5.18 (s, 2H), 3.87 (s, 3H), 3.43 (t, J = 5.0 Hz, 4H), 3.05 (t, J = 5.0 Hz, 4H), 2.33 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C25H26FN9, 472.2; found, 472.1; HPLC purity: 254 nm: 99.8%. [0689] Using the procedures described in Example 93 and reagents, starting materials, and conditions known to those skilled in the art, the following compounds representative of the present disclosure were prepared:
Figure imgf000317_0001
Example 94: Synthesis of Exemplary Compound 422 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)-N-((2-methylbenzo[d]thiazol-5- yl)methyl)piperazine-1-carboxamide (Compound 422)
Figure imgf000318_0001
Step 1. Preparation of 2-(2-methylbenzo[d]thiazol-5-yl)-1-morpholinoethane-1-thione
Figure imgf000318_0002
[0690] To a solution of 1-(2-methylbenzo[d]thiazol-5-yl)ethan-1-one (1.0 g, 5.23 mmol) in ACN (20 mL) was added sulfur (251.5 mg, 7.84 mmol), morpholine (683.3 mg, 7.84 mmol) and Ti(OiPr)4 (2.97 g, 10.46 mmol) then the mixture was heated to 90°C and stirred for 16 h under nitrogen. The reaction was cooled to rt and filtered, then the filter cake was washed with ACN (3x50 mL). The filtrate was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with PE/EtOAc (2:1) to afford 600 mg (37%) of 2-(2-methylbenzo[d]thiazol-5-yl)-1-morpholinoethane-1-thione as a light yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 7.97 (d, J = 8.2 Hz, 1H), 7.86 (d, J = 1.7 Hz, 1H), 7.39 (dd, J = 8.3, 1.7 Hz, 1H), 4.43 (s, 2H), 4.27 – 4.18 (m, 2H), 3.78 – 3.71 (m, 2H), 3.64 (t, J = 4.8 Hz, 2H), 3.42 (t, J = 4.8 Hz, 2H), 2.79 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C14H16N2OS2, 293.1; found, 293.1. Step 2. Preparation of 2-(2-methylbenzo[d]thiazol-5-yl)acetic acid
Figure imgf000318_0003
[0691] To a solution of 2-(2-methylbenzo[d]thiazol-5-yl)-1-morpholinoethane-1-thione (500 mg, 1.71 mmol, as prepared in the previous step) in EtOH (12 mL) and H2O (9 mL) was added KOH (575.6 mg, 10.26 mmol) then the mixture was stirred at rt for 8 h under nitrogen. The pH of the reaction was adjusted to pH 5 with conc. HCl then the mixture was extracted with EtOAc (3x30 mL). The organic extracts were combined, washed with brine (3x10 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with DCM/MeOH (20:1) to afford 400 mg (90%) of 2-(2-methylbenzo[d]thiazol-5- yl)acetic acid as a light yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 12.22 (s, 1H), 7.93 (d, J = 8.2 Hz, 1H), 7.79 (d, J = 1.6 Hz, 1H), 7.29 (dd, J = 8.2, 1.7 Hz, 1H), 3.70 (s, 2H), 2.78 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C10H9NO2S, 208.0 found, 208.0. Step 3. Preparation of 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)-N-((2- methylbenzo[d]thiazol-5-yl)methyl)piperazine-1-carboxamide (Compound 422)
Figure imgf000319_0001
[0692] To a solution of 2-(2-methylbenzo[d]thiazol-5-yl)acetic acid (200 mg, 0.97 mmol, as prepared in the previous step) in ACN (10 mL) cooled to 0°C was added DPPA (398.4 mg, 1.45 mmol) and TEA (293 mg, 2.90 mmol) then the mixture was warmed to rt and stirred for 2 h under nitrogen. The reaction was concentrated under reduced pressure to afford crude 2-(2-methylbenzo[d]thiazol-5-yl)acetyl azide, which was used directly without purification. MS (ESI) m/z [M+H]+ calcd. for C10H8N4OS, 233.0 found, 233.0. [0693] To a solution of the crude 2-(2-methylbenzo[d]thiazol-5-yl)acetyl azide (200 mg, 0.86 mmol) in toluene (5 mL, 46.99 mmol) were added 6-(1-methyl-1H-pyrazol-4-yl)-3- (piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 12) (243.1 mg, 0.86 mmol) and TEA (261.4 mg, 2.58 mmol) then the mixture was heated to 110°C and stirred for 16 h under nitrogen. The reaction was cooled to rt, water (30 mL) was added and the mixture was extracted with EtOAc (3x30 mL). The organic extracts were combined, washed with brine (3x10 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 20.3 mg (5%) of 4- (6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)-N-((2-methylbenzo[d]thiazol- 5-yl)methyl)piperazine-1-carboxamide (Compound 422) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.23 (s, 1H), 8.00 – 7.92 (m, 2H), 7.82 – 7.76 (m, 2H), 7.67 (d, J = 9.2 Hz, 1H), 7.37 – 7.26 (m, 3H), 4.39 (d, J = 6.0 Hz, 2H), 3.88 (s, 3H), 3.54 (t, J = 4.9 Hz, 4H), 2.96 (t, J = 4.9 Hz, 4H), 2.79 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C25H26N8OS, 487.2; found, 487.2; HPLC purity: 254 nm: 99.2%. [0694] Using the procedures described in Example 94 and reagents, starting materials, and conditions known to those skilled in the art, the following compounds representative of the present disclosure were prepared:
Figure imgf000320_0002
Example 95: Synthesis of Exemplary Compound 423 3-(3-(5-benzylpyrimidin-2-yl)pyrrolidin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrimidine (Compound 423)
Figure imgf000320_0001
[0695] To a solution of 5-benzyl-2-(pyrrolidin-3-yl)pyrimidine (Compound S100) (800 mg, 3.34 mmol) and 3-bromo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrimidine (Compound S10) (743.7 mg, 2.67 mmol) in dioxane (20 mL) were added KOTMS (857.6 mg, 6.68 mmol), tBuXPhos Pd G3 (797.6 mg, 1.00 mmol), and tBuXPhos (425.8 mg, 1.00 mmol) then the mixture was heated to 90°C and stirred overnight under nitrogen. The reaction was cooled to rt and concentrated under reduced pressure. Water (100 mL) was added to the residue then the mixture was extracted with DCM (3x100 mL). The organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with PE/EtOAc (1:1) and Prep-HPLC to afford 107.1 mg (7%) of 3-(3-(5-benzylpyrimidin-2-yl)pyrrolidin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrimidine (Compound 423) as an orange solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.06 (d, J = 2.1 Hz, 1H), 8.68 (s, 2H), 8.49 (d, J = 2.1 Hz, 1H), 8.26 (s, 1H), 8.00 (s, 1H), 7.71 (s, 1H), 7.35 – 7.26 (m, 4H), 7.26 – 7.17 (m, 1H), 3.97 (s, 2H), 3.93-3.85 (m, 4H), 3.82-3.72 (m, 1H), 3.66-3.60 (m, 1H), 3.58-3.48 (m, 2H), 2.40-2.30 (m, 2H); MS (ESI) m/z [M+H]+ calcd. for C25H24N8, 437.2; found, 437.1; HPLC purity: 254 nm: 96.3%. Example 96: Synthesis of Exemplary Compound 424 N-(benzo[d]thiazol-5-ylmethyl)-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxamide (Compound 424)
Figure imgf000321_0001
Step 1. Preparation of methyl 2-(3-thioureidophenyl)acetate
Figure imgf000321_0002
[0696] To a solution of methyl 2-(3-aminophenyl)acetate (10 g, 60.5 mmol) in chloroform (150 mL) was added NaSCN (7.36 g, 90.8 mmol) and TFA (17.3 g, 151 mmol) then the mixture was heated to 80°C and stirred for 2 h. The reaction was cooled to rt and water (200 mL) was added then the mixture was extracted with EtOAc (3x200 mL). The organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with PE/EtOAc (1:1) to afford 3 g (19%) of methyl 2-(3- thioureidophenyl)acetate as a light yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.72 (s, 1H), 7.35 (d, J = 8.0 Hz, 1H), 7.30 – 7.18 (m, 2H), 7.01 (d, J = 7.6 Hz, 1H), 6.95 – 6.86 (m, 1H), 3.68 – 3.59 (m, 5H); MS (ESI) m/z [M+H]+ calcd. for C10H12N2O2S, 225.1; found, 225.1. Step 2. Preparation of a mixture of methyl 2-(2-aminobenzo[d]thiazol-5-yl) acetate and methyl 2-(2-aminobenzo[d]thiazol-7-yl) acetate
Figure imgf000321_0003
[0697] To a solution of methyl 2-(3-thioureidophenyl)acetate (1.0 g, 4.46 mmol, as prepared in the previous step) in AcOH (10 mL) was added Br2 (0.86 g, 5.40 mmol) then the mixture was heated to 80°C and stirred for 2 h. The reaction was cooled to rt and concentrated under reduced pressure. To the residue was added an aqueous solution of Na2S2O3 and an aqueous solution of Na2CO3, then the mixture was extracted with EtOAc (3x200 mL). The organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with PE/EtOAc (3:7) to afford 850 mg (34%) of a mixture of methyl 2-(2-aminobenzo[d]thiazol-5-yl) acetate and methyl 2-(2- aminobenzo[d]thiazol-7-yl) acetate as a brown oil. MS (ESI) m/z [M+H]+ calcd. for C10H10N2O2S, 223.0; found, 223.0. Step 3. Preparation of a mixture of methyl 2-(benzo[d]thiazol-5-yl) acetate and methyl 2- (benzo[d]thiazol-7-yl) acetate
Figure imgf000322_0001
[0698] To a solution of a mixture of methyl 2-(2-aminobenzo[d]thiazol-5-yl) acetate and methyl 2-(2-aminobenzo[d]thiazol-7-yl) acetate (800 mg, 3.60 mmol, as prepared in the previous step) in THF (10 mL) was added isoamyl nitrite (843 mg, 7.20 mmol) then the mixture was heated to 70°C and stirred for 2 h. The reaction was cooled to rt and concentrated under reduced pressure. The residue was purified by silica gel column chromatography eluting with PE/EtOAc (5:1) to afford 490 mg (16%) of a mixture of methyl 2-(benzo[d]thiazol-5-yl) acetate and methyl 2-(benzo[d]thiazol-7-yl) acetate as a yellow oil. MS (ESI) m/z [M+H]+ calcd. for C10H9NO2S, 208.0; found, 208.0. Step 4. Preparation of 2-(benzo[d]thiazol-5-yl) acetic acid
Figure imgf000322_0002
[0699] To a solution of a mixture of methyl 2-(benzo[d]thiazol-5-yl) acetate and methyl 2- (benzo[d]thiazol-7-yl) acetate (450 mg, 2.17 mmol, as prepared in the previous step) in MeOH (6 mL) and H2O (2 mL) was added NaOH (174 mg, 4.34 mmol) then the mixture was stirred at rt for 2 h. The reaction was concentrated under reduced pressure then water (50mL) was added, and the mixture was extracted with DCM (3x80 mL). The organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-ACHIRAL-SFC to afford 120 mg (26%) of 2-(benzo[d]thiazol-5-yl) acetic acid as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.35 (s, 1H), 8.04 (d, J = 8.2 Hz, 1H), 7.95 (s, 1H), 7.37 (dd, J = 8.3, 1.6 Hz, 1H), 3.63 (s, 2H); MS (ESI) m/z [M+H]+ calcd. for C9H7NO2S, 194.0; found, 194.0. Step 5. Preparation of N-(benzo[d]thiazol-5-ylmethyl)-4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxamide (Compound 424)
Figure imgf000323_0001
[0700] To a solution of 2-(benzo[d]thiazol-5-yl) acetic acid (90 mg, 0.47 mmol, as prepared in the previous step) and TEA (141 mg, 1.40 mmol) in THF (4 mL) cooled to 0°C was added DPPA (256 mg, 0.93 mmol) then the mixture was warmed to rt and stirred for 2 h. The reaction was concentrated under reduced pressure to afford crude 2-(benzo[d]thiazol- 5-yl)acetyl azide, which was used directly without purification. [0701] To a solution of crude 2-(benzo[d]thiazol-5-yl) acetyl azide (100 mg, 0.46 mmol) and TEA (93 mg, 0.92 mmol) in toluene (4 mL) was added 6-(1-methyl-1H-pyrazol-4-yl)- 3-(piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 12) (129 mg, 0.46 mmol) then the mixture was heated to 100°C and stirred for 2 h. The reaction was cooled to rt and concentrated under reduced pressure. The residue was purified by silica gel column chromatography eluting with DCM/MeOH (20:1) and Prep-HPLC to afford 25.9 mg (12%) of N-(benzo[d]thiazol-5-ylmethyl)-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazine-1-carboxamide (Compound 424) as a white solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.38 (s, 1H), 8.82 (s, 1H), 8.22 (s, 1H), 8.10 (d, J = 8.3 Hz, 1H), 7.97 (s, 2H), 7.77 (s, 1H), 7.67 (d, J = 9.3 Hz, 1H), 7.43 (dd, J = 8.3, 1.6 Hz, 1H), 7.35 – 7.28 (m, 2H), 4.43 (d, J = 5.7 Hz, 2H), 3.87 (s, 3H), 3.55 (t, J = 4.9 Hz, 4H), 2.97 (t, J = 4.9 Hz, 4H); MS (ESI) m/z [M+H]+ calcd. for C24H24N8OS, 473.2; found, 473.1; HPLC purity: 254 nm: 99.7%. Example 97: Synthesis of Exemplary Compound 425 (1-benzyl-3-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)- 1H-1,2,4-triazol-5-yl)methanol (Compound 425)
Figure imgf000324_0001
Step 1. Preparation of (1-benzyl-3-bromo-1H-1,2,4-triazol-5-yl)methanol
Figure imgf000324_0002
[0702] To a stirred solution of 3-bromo-1H-1,2,4-triazole-5-carboxylic acid (10 g, 52.09 mmol) in THF (150 mL) cooled to 0°C was added BH3·THF (208.4 mL, 208.36 mmol) then the mixture was warmed to rt and stirred for 30 minutes. The reaction was heated to 60°C and stirred overnight. The reaction was cooled to 0°C and quenched with MeOH. The resulting mixture was concentrated under reduced pressure then the residue was passed through a silica gel column eluting with DCM/MeOH (40:1) to afford 5.0 g (54%) of crude (3-bromo-1H-1,2,4-triazol-5-yl)methanol as a light yellow oil. [0703] To a stirred solution of 3-bromo-1H-1,2,4-triazol-5-yl)methanol (5.0 g, 28.1 mmol) and benzyl bromide (9.23 g, 53.94 mmol) in DMF (150 mL) was added K2CO3 (12.42 g, 89.89 mmol) then the mixture was heated to 80°C and stirred for 2 h. The reaction was cooled to rt and filtered, then the filter cake was washed with EtOAc (2x100 mL). The filtrate was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with DCM/MeOH (40/1) and reversed-phase flash chromatography using a fluoro-phenyl column eluting with 16-28% ACN/water (0.05% TFA), to afford 195 mg (3%) of (1-benzyl-3-bromo-1H-1,2,4-triazol-5-yl)methanol as a light yellow oil.1H NMR (400 MHz, DMSO-d6) δ (ppm) 7.44 – 7.25 (m, 5H), 5.86 (t, J = 5.9 Hz, 1H), 5.41 (s, 2H), 4.64 (d, J = 5.8 Hz, 2H); MS (ESI) m/z [M+H]+ calcd. for C10H10BrN3O, 268.0; found,268.1. Step 2. (1-benzyl-3-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazin-1-yl)-1H-1,2,4-triazol-5-yl)methanol (Compound 425)
Figure imgf000325_0001
[0704] To a solution of (1-benzyl-3-bromo-1H-1,2,4-triazol-5-yl)methanol (160 mg, 0.60 mmol, as prepared in the previous step) and 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1- yl)pyrazolo[1,5-a]pyridine (Compound 12) (202.2 mg, 0.72 mmol) in dioxane (20 mL) were added Pd-PEPPSI-IPentCl 2-methylpyridine (100.4 mg, 0.12 mmol) and Cs2CO3 (388.9 mg, 1.20 mmol) then the mixture was heated to 90°C and stirred for 16 h under nitrogen. The reaction was cooled to rt and concentrated under reduced pressure. The residue was purified by Prep-HPLC to afford 44.4 mg (16%) of (1-benzyl-3-(4-(6-(1- methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)-1H-1,2,4-triazol-5- yl)methanol (Compound 425) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.76 (s, 1H), 7.65 (d, J = 9.2 Hz, 1H), 7.42 – 7.23 (m, 6H), 5.66 (s, 1H), 5.27 (s, 2H), 4.54 (s, 2H), 3.87 (s, 3H), 3.44 (t, J = 5.0 Hz, 4H), 3.05 (t, J = 5.0 Hz, 4H); MS (ESI) m/z [M+H]+ calcd. for C25H27N9O, 470.2; found,470.1; HPLC purity: 254 nm: 99.0%. Example 98: Synthesis of Exemplary Compound 426 3-(4-(1-benzyl-5-(methoxymethyl)-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 426)
Figure imgf000325_0002
Step 1. Preparation of 1-benzyl-3-bromo-5-(methoxymethyl)-1H-1,2,4-triazole
Figure imgf000326_0001
[0705] To a stirred mixture of 3-bromo-5-(methoxymethyl)-1H-1,2,4-triazole (2 g, 10.41 mmol) and benzyl bromide (2.14 g, 12.49 mmol) in DMF (10 mL) was added K2CO3 (2.88 g, 20.83 mmol) in portions at rt then the mixture was heated to 100°C and stirred for 3 h under nitrogen. The reaction was cooled to rt and filtered, then the filter cake was washed with EtOAc (2x10 mL). The filtrate was concentrated under reduced pressure and the residue was purified by Prep-HPLC to afford 580 mg (20%) of 1-benzyl-3-bromo-5- (methoxymethyl)-1H-1,2,4-triazole as a white solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 7.43 – 7.29 (m, 3H), 7.33 – 7.25 (m, 2H), 5.41 (s, 2H), 4.64 (s, 2H), 3.30 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C26H29N9O, 282.1; found, 281.9. Step 2. Preparation of 3-(4-(1-benzyl-5-(methoxymethyl)-1H-1,2,4-triazol-3-yl)piperazin- 1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 426)
Figure imgf000326_0002
[0706] To a solution of 1-benzyl-3-bromo-5-(methoxymethyl)-1,2,4-triazole (200 mg, 0.70 mmol, as prepared in the previous step) and 1-[6-(1-methylpyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl]piperazine (Compound 12) (200.1 mg, 0.70 mmol) in dioxane (3 mL) were added Pd-PEPPSI-IPentCl 2-methylpyridine (59.6 mg, 0.07 mmol) and Cs2CO3 (461.9 mg, 1.41 mmol) then the mixture was heated to 100°C and stirred for 2 h under nitrogen. The reaction was cooled to rt, water (100 mL) was added, and the mixture was extracted with EtOAc (2x50 mL). The organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 131.7 mg (38%) of 3-(4-(1-benzyl-5-(methoxymethyl)-1H-1,2,4- triazol-3-yl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound 426) as a yellow green solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (t, J = 1.2 Hz, 1H), 8.23 (s, 1H), 7.98 (s, 1H), 7.77 (s, 1H), 7.66 (d, J = 9.3 Hz, 1H), 7.41 – 7.24 (m, 6H), 5.25 (s, 2H), 4.52 (s, 2H), 3.88 (s, 3H), 3.46 (t, J = 4.9 Hz, 4H), 3.29 (s, 3H), 3.06 (t, J = 5.0 Hz, 4H); MS (ESI) m/z [M+H]+ calcd. for C26H29N9O, 484.3; found, 484.2; HPLC purity: 254 nm: 99.8%. Example 99: Synthesis of Exemplary Compound 427 3-(4-(1-benzyl-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-4-methoxy-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyrazine (Compound 427)
Figure imgf000327_0001
Step 1. Preparation of diethyl 1-(2-(1-methyl-1H-pyrazol-4-yl)-2-oxoethyl)-1H-pyrazole- 3,5-dicarboxylate
Figure imgf000327_0002
[0707] To a stirred solution of diethyl 1H-pyrazole-3,5-dicarboxylate (18.9 g, 89.06 mmol) and 2-bromo-1-(1-methyl-1H-pyrazol-4-yl)ethan-1-one (18.08 g, 89.06 mmol) in ACN (250 mL) was added Cs2CO3 (58.04 g, 178.13 mmol) at rt then the mixture was stirred for 4 h. The reaction was filtered, and the filter cake was washed with ACN (100 mL). The filtrate was concentrated under reduced pressure then the residue was dissolved in EtOAc (80 mL), then heptane (200 mL) was added. The mixture was stirred at rt for 2 h, then the suspension was filtered, and the filter cake was washed with heptane. The solid was dried under reduced pressure to afford 25 g (84%) of diethyl 1-(2-(1-methyl-1H-pyrazol-4-yl)-2- oxoethyl)-1H-pyrazole-3,5-dicarboxylate as a yellow solid. 1H NMR (400 MHz, DMSO- d6) δ (ppm) 8.55 (s, 1H), 8.06 (s, 1H), 7.33 (s, 1H), 5.93 (s, 2H), 4.31 (q, J = 7.1 Hz, 2H), 4.22 (q, J = 7.1 Hz, 2H), 3.93 (s, 3H), 1.30 (t, J = 7.1 Hz, 3H), 1.21 (t, J = 7.1 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C15H18N4O5, 335.1; found, 335.2. Step 2. Preparation of 4-hydroxy-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrazine-2- carboxylic acid
Figure imgf000328_0001
[0708] To a stirred solution of diethyl 1-(2-(1-methyl-1H-pyrazol-4-yl)-2-oxoethyl)-1H- pyrazole-3,5-dicarboxylate (50 g, 149.55 mmol, as prepared in the previous step) in HOAc (1 L) was added NH4OAc (345.84 g, 4.48 mol) then the mixture was heated to 120°C and stirred for 48 h. The reaction was cooled to rt, water (6 L) was added, and the mixture was stirred for 2 h. The suspension was filtered, and the filter cake was washed with water. The solid was added to 6M HCl (350 mL) then heated to 100°C and stirred for 16 h. The reaction was cooled to rt and water (350 mL) was added. The mixture was stirred for 1 h, then filtered and the filter cake was washed with water and dried under reduced pressure to afford (24 g (76%) of 4-hydroxy-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrazine-2- carboxylic acid as a white solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 13.17 (s, 1H), 11.64 (s, 1H), 8.30 (s, 1H), 8.14 (s, 1H), 8.05 (s, 1H), 7.32 (d, J = 0.5 Hz, 1H), 3.88 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C11H9N5O3, 260.1; found, 260.2. Step 3. Preparation of 6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrazin-4-ol
Figure imgf000328_0002
[0709] To a stirred solution of 4-hydroxy-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrazine-2-carboxylic acid (2 g, 7.71 mmol, as prepared in the previous step) in NMP (20 mL) were added 1,10-phenanthroline (695.2 mg, 3.86 mmol) and Cu(OAc)2 (700.7 mg, 3.86 mmol) then the mixture was heated to 165°C and stirred for 48 h. The reaction was cooled to rt, 3M HCl (40 mL) was added, and the mixture was stirred for 3 h. The suspension was filtered, the filter cake was washed with water, and dried under reduced pressure to afford 1.53 g (92%) of 6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrazin-4- ol as an off-white solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 11.44 (s, 1H), 8.28 (s, 1H), 8.09 (s, 1H), 8.03 (s, 1H), 7.88 (d, J = 2.2 Hz, 1H), 6.99 (dd, J = 2.2, 0.8 Hz, 1H), 3.87 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C10H9N5O, 216.1; found, 216.2. Step 4. Preparation of 4-chloro-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrazine dihydrochloride
Figure imgf000329_0001
[0710] A solution of 6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrazin-4-ol (5.5 g, 25.56 mmol, as prepared in the previous step) in POCl3 (62.69 g, 408.90 mmol) was heated to 80°C and stirred for 7 h. The reaction was cooled to 50°C then ACN (45 mL) was added. The mixture was then cooled to rt and filtered. The filter cake was washed with ACN (25 mL) then dried under reduced pressure. To the filtrate was added MTBE (90 mL) and the mixture was stirred at rt for 16 h. The suspension was filtered, the filter cake was washed with ACN (25 mL) and dried under reduced pressure. The solids were combined to afford 5 g (64%) of 4-chloro-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrazine dihydrochloride as an off-white solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 10.08 – 9.39 (m, 2H), 9.21 (s, 1H), 8.27 (s, 1H), 8.20 (d, J = 2.4 Hz, 1H), 8.03 (s, 1H), 7.01 (dd, J = 2.5, 0.9 Hz, 1H), 3.88 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C10H8ClN5, 234.1; found, 234.1. Step 5. Preparation of 4-methoxy-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrazine
Figure imgf000329_0002
[0711] To a stirred solution of 4-chloro-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrazine (900 mg, 3.85 mmol, as prepared in the previous step) in MeOH (10 mL) was added NaOMe (416.2 mg, 7.70 mmol) at rt then the mixture was stirred for 1 h. The reaction was purified by silica gel column chromatography eluting with PE/EtOAc (1:1) to afford 560 mg (63%) of 4-methoxy-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrazine as a white solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.75 (s, 1H), 8.21 (s, 1H), 8.11 – 7.94 (m, 2H), 6.84 (d, J = 2.1 Hz, 1H), 4.11 (s, 3H), 3.89 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C11H11N5O, 230.1; found, 230.2. Step 6. Preparation of 3-bromo-4-methoxy-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrazine
Figure imgf000330_0001
[0712] To a stirred solution of 4-methoxy-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrazine (240 mg, 1.05 mmol, as prepared in the previous step) in ACN (9 mL) cooled to 0°C was added NBS (149.5 mg, 0.84 mmol) then the mixture was stirred for 1 h. The reaction was diluted with water and extracted with DCM (3x30 mL). The organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-TLC (DCM/MeOH 60:1) to afford 260 mg (81%) of 3-bromo-4-methoxy-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrazine as a white solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.77 (s, 1H), 8.22 (s, 1H), 8.13 (s, 1H), 8.02 (s, 1H), 4.12 (s, 3H), 3.88 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C11H10BrN5O, 308.1; found, 308.2. Step 7. Preparation of 4-methoxy-6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1- yl)pyrazolo[1,5-a]pyrazine formic acid salt
Figure imgf000330_0002
[0713] To a stirred solution of 3-bromo-4-methoxy-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyrazine (260 mg, 0.84 mmol, as prepared in the previous step) and piperazine (363.4 mg, 4.22 mmol) in DMF (5 mL) were added Pd-PEPPSI-IPentCl 2- methylpyridine (71 mg, 0.08 mmol) and Cs2CO3 (549.8 mg, 1.69 mmol) then the mixture was heated to 90°C and stirred for 16 h under nitrogen. The reaction was cooled to rt and filtered, then the filter cake was washed with MeOH (3x20 mL). The filtrate was concentrated under reduced pressure and the residue was purified by reversed-phase flash chromatography using a C18 silica gel column eluting with 5-30% ACN/water (0.1% FA) to afford 110 mg (36%) of 4-methoxy-6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1- yl)pyrazolo[1,5-a]pyrazine formic acid salt as a yellow solid.1H NMR (400 MHz, DMSO- d6) δ (ppm) 8.54 (s, 1H), 8.30 (s, 1H), 8.18 (s, 1H), 7.98 (s, 1H), 7.74 (s, 1H), 4.08 (s, 3H), 3.87 (s, 3H), 3.07 (d, J = 18.5 Hz, 8H); MS (ESI) m/z [M+H]+ calcd. for C16H21N7O3, 314.1; found, 314.2. Step 8. Preparation of 3-(4-(1-benzyl-1H-1,2,4-triazol-3-yl)piperazin-1-yl)-4-methoxy-6- (1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyrazine (Compound 427)
Figure imgf000331_0001
[0714] To a stirred solution of 4-methoxy-6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1- yl)pyrazolo[1,5-a]pyrazine formic acid salt (100 mg, 0.28 mmol, as prepared in the previous step) and 1-benzyl-3-bromo-1H-1,2,4-triazole (66.3 mg, 0.28 mmol, as prepared in Compound S101, Step 1) in dioxane (4 mL) were added Pd-PEPPSI-IPentCl 2- methylpyridine (23.4 mg, 0.03 mmol) and Cs2CO3 (181.3 mg, 0.56 mmol) then the mixture was heated to 90°C and stirred for 16 h under nitrogen. The reaction was cooled to rt and filtered, then the filter cake was washed with DCM (3x30 mL). The filtrate was concentrated under reduced pressure then the residue was purified by Prep-TLC (DCM/MeOH 15:1) and Prep-HPLC to afford 47 mg (36%) of 3-(4-(1-benzyl-1H-1,2,4- triazol-3-yl)piperazin-1-yl)-4-methoxy-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyrazine (Compound 427) as a white solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.53 (s, 1H), 8.32 (s, 1H), 8.18 (s, 1H), 7.98 (s, 1H), 7.73 (s, 1H), 7.42 – 7.23 (m, 5H), 5.23 (s, 2H), 4.08 (s, 3H), 3.87 (s, 3H), 3.43 (t, J = 4.7 Hz, 4H), 3.11 (t, J = 4.7 Hz, 4H); MS (ESI) m/z [M+H]+ calcd. for C24H26N10O, 471.2; found, 471.3; HPLC purity: 254 nm: 99.4%.
Example 100: Synthesis of Exemplary Compound 428 1-(benzo[d]thiazol-5-yl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate (Compound 428)
Figure imgf000332_0001
Step 1.1-(benzo[d]thiazol-5-yl)ethan-1-ol
Figure imgf000332_0002
[0715] To a solution of 1-(1,3-benzothiazol-5-yl)ethanone (500 mg, 2.82 mmol) in MeOH (10 mL) was added NaBH4 (213.4 mg, 5.64 mmol) at rt then the mixture was stirred for 1 h under nitrogen. The reaction was concentrated under reduced pressure, then the residue was dissolved in water (50 mL) and extracted with DCM (3x50 mL). The organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure to afford 400 mg (79%) of 1-(benzo[d]thiazol-5-yl)ethan-1-ol as a brown solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.37 (s, 1H), 8.16 – 7.97 (m, 2H), 7.48 (dd, J = 8.3, 1.6 Hz, 1H), 5.31 (d, J = 4.3 Hz, 1H), 4.97 – 4.82 (m, 1H), 1.39 (d, J = 6.4 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C9H9NOS, 180.0; found, 180.0; HPLC purity: 254 nm:95%. Step 2. Preparation of 1-(benzo[d]thiazol-5-yl)ethyl 4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 428)
Figure imgf000332_0003
[0716] To a solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)pyrazolo[1,5- a]pyridine (Compound 12) (300 mg, 1.06 mmol) in pyridine (15 mL)were added BTC (1.58 g, 5.31 mmol) and 1-(1,3-benzothiazol-5-yl)ethanol (209.4 mg, 1.16 mmol, as prepared in the previous step) at rt then the mixture was stirred overnight under nitrogen. The reaction was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with DCM/MeOH (10:1) and Prep-HPLC to afford 130.7 mg (25%) of 1-(benzo[d]thiazol-5-yl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazine-1-carboxylate (Compound 428) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.42 (s, 1H), 8.82 (s, 1H), 8.22 (s, 1H), 8.17 (d, J = 8.3 Hz, 1H), 8.08 (d, J = 1.6 Hz, 1H), 7.97 (s, 1H), 7.76 (s, 1H), 7.68-7.63 (m, 1H), 7.55-7.50 (m, 1H), 7.35-7.30 (m, 1H), 5.91 (q, J = 6.5 Hz, 1H), 3.86 (s, 3H), 3.75-3.50 (m, 4H), 2.97 (br s, 4H), 1.57 (d, J = 6.5 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C25H25N7O2S, 488.2; found, 488.1; HPLC purity: 254 nm: 98.4%. [0717] Using the procedures described in Example 100 and reagents, starting materials, and conditions known to those skilled in the art, the following compounds representative of the present disclosure were prepared:
Figure imgf000333_0002
Example 101: Synthesis of Exemplary Compound 431 2-(4-chlorophenoxy)-1-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazin-1-yl)ethan-1-one (Compound 431)
Figure imgf000333_0001
[0718] To a solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)pyrazolo[1,5- a]pyridine (Compound 12) (150 mg, 0.53 mmol) and 2-(4-chlorophenoxy)acetyl chloride (130.7 mg, 0.64 mmol) in DCM (10 mL) cooled to 0°C was added TEA (107.5 mg, 1.06 mmol) then the mixture was stirred for 2 h under nitrogen. The reaction was warmed to rt and concentrated under reduced pressure then the residue was purified by Prep-TLC (DCM/MeOH 10:1) and Prep-HPLC to afford 151.1 mg (63%) of 2-(4-chlorophenoxy)-1- (4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)ethan-1-one (Compound 431) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (t, J = 1.3 Hz, 1H), 8.23 (s, 1H), 7.98 (s, 1H), 7.77 (s, 1H), 7.68 (dd, J = 9.2, 0.8 Hz, 1H), 7.38 – 7.28 (m, 3H), 7.02 – 6.93 (m, 2H), 4.91 (s, 2H), 3.87 (s, 3H), 3.68 – 3.61 (m, 4H), 3.08 – 2.92 (m, 4H); MS (ESI) m/z [M+H]+ calcd. for C23H23ClN6O2, 451.2; found, 451.0; HPLC purity: 254 nm: 99.6%. Example 102: Synthesis of Exemplary Compound 432 (R)-2-amino-1-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)- 3-phenylpropan-1-one (Compound 432)
Figure imgf000334_0001
Step 1. Preparation of tert-butyl (R)-(1-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazin-1-yl)-1-oxo-3-phenylpropan-2-yl)carbamate
Figure imgf000334_0002
[0719] To a solution of (tert-butoxycarbonyl)-D-phenylalanine (281.9 mg, 1.06 mmol) in DCM (10 mL) cooled to 0°C was added HATU (606 mg, 1.60 mmol) and DIEA (274.7 mg, 2.13 mmol) then the mixture was stirred for 10 minutes under nitrogen. To the reaction was added 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 12) (300 mg, 1.06 mmol) at 0°C then the mixture was stirred for 2 h. The reaction was diluted with water (20 mL) and extracted with DCM (3x20 mL). The organic extracts were combined, washed with brine (3x10 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with DCM/MeOH (20:1) to afford 550 mg (78%) of tert-butyl (R)-(1-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazin-1-yl)-1-oxo-3-phenylpropan-2-yl)carbamate as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.78 (s, 1H), 8.19 (s, 1H), 7.95 (s, 1H), 7.67 (s, 1H), 7.62 (d, J = 9.2 Hz, 1H), 7.34 – 7.23 (m, 5H), 7.22 – 7.09 (m, 2H), 4.71 – 4.58 (m, 1H), 3.86 (s, 3H), 3.79 – 3.58 (m, 4H), 2.93 – 2.57 (m, 6H), 1.31 (s, 9H); MS (ESI) m/z [M+H]+ calcd. for C29H35N7O3, 530.3; found, 530.3. Step 2. Preparation of (R)-2-amino-1-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazin-1-yl)-3-phenylpropan-1-one (Compound 432)
Figure imgf000335_0001
[0720] To a solution of tert-butyl (R)-(1-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazin-1-yl)-1-oxo-3-phenylpropan-2-yl)carbamate (500 mg, 0.94 mmol, as prepared in the previous step) in DCM (5 mL) cooled to 0°C was added 4M HCl in dioxane (2.36 mL, 9.44 mmol) then the mixture was stirred for 2 h under nitrogen. The reaction was concentrated under reduced pressure then the residue was purified by Prep- HPLC to afford 123.9 mg (30%) of (R)-2-amino-1-(4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)-3-phenylpropan-1-one (Compound 432) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.23 (s, 1H), 7.97 (s, 1H), 7.67 (s, 1H), 7.61 (d, J = 9.3 Hz, 1H), 7.36 – 7.15 (m, 6H), 3.97 (t, J = 7.0 Hz, 1H), 3.87 (s, 3H), 3.74 – 3.64 (m, 1H), 3.61 – 3.48 (m, 2H), 3.47 – 3.38 (m, 1H), 2.98 – 2.82 (m, 2H), 2.80 – 2.73 (m, 2H), 2.72 – 2.65 (m, 1H), 2.49 – 3.39 (m, 1H), 1.84 (s, 2H); MS (ESI) m/z [M+H]+ calcd. for C24H27N7O, 430.2; found, 430.1; HPLC purity: 254 nm: 99.2%. [0721] Using the procedures described in Example 102 and reagents, starting materials, and conditions known to those skilled in the art, the following compounds representative of the present disclosure were prepared:
Figure imgf000336_0002
Example 103: Synthesis of Exemplary Compound 433 2-(4-fluorophenyl)-1-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin- 1-yl)ethan-1-one (Compound 433)
Figure imgf000336_0001
[0722] To a stirred solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1- yl)pyrazolo[1,5-a]pyridine (Compound 12) (200 mg, 0.71 mmol), DIEA (183.1 mg, 1.42 mmol) and 2-(4-fluorophenyl)acetic acid (131 mg, 0.85 mmol) in DCM (4 mL) cooled to 0°C was added HATU (404 mg, 1.06 mmol) then the mixture was warmed to rt and stirred for 1 h. The reaction was diluted with DCM (30 mL), washed with water (3x30 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-TLC (DCM/MeOH 40:1) and Prep-HPLC to afford 201 mg (65%) of 2-(4-fluorophenyl)-1-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazin-1-yl)ethan-1-one (Compound 433) as an off-white solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (s, 1H), 8.23 (s, 1H), 7.98 (s, 1H), 7.75 (s, 1H), 7.66 (d, J = 9.3 Hz, 1H), 7.36 – 7.26 (m, 3H), 7.19 – 7.10 (m, 2H), 3.87 (s, 3H), 3.78 (s, 2H), 3.74 – 3.61 (m, 4H), 3.02 – 2.86 (m, 4H); MS (ESI) m/z [M+H]+ calcd. for C23H23FN6O, 419.2; found, 419.2; HPLC purity: 254 nm: 95.8%. [0723] Using the procedures described in Example 103 and reagents, starting materials, and conditions known to those skilled in the art, the following compounds representative of the present disclosure were prepared:
Figure imgf000337_0001
Figure imgf000338_0001
Figure imgf000339_0001
Figure imgf000340_0001
Figure imgf000341_0001
Figure imgf000342_0001
Example 104: Synthesis of Exemplary Compound 451 (R)-1-(2-cyanophenyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate (Compound 451) and (S)-1-(2-cyanophenyl)ethyl 4-(6-(1- methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 452)
Figure imgf000343_0001
[0724] To a stirred mixture of 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1- yl)pyrazolo[1,5-a]pyridine (Compound 12) (500 mg, 1.7 mmol) and 2-(1- hydroxyethyl)benzonitrile (286 mg, 1.9 mmol) in pyridine (10 mL) cooled to 0°C was added BTC (1.05 g, 3.5 mmol) in portions then the mixture was warmed to rt and stirred for 2 h. The reaction was cooled to 0°C, quenched with water, and extracted with DCM (3x200 mL). The organic extracts were combined, then concentrated under reduced pressure. The residue was purified by Prep-HPLC to afford racemic 1-(2- cyanophenyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate as a yellow solid. [0725] Racemic 1-(2-cyanophenyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazine-1-carboxylate was separated by Chiral-Prep-HPLC to afford 100.7 mg (25%) of (R)-1-(2-cyanophenyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 451, first eluting peak) as a yellow solid and 102.9 mg (25%) of (S)-1-(2-cyanophenyl)ethyl 4-(6-(1-methyl- 1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 452, second eluting peak) as a yellow solid. [0726] (R)-1-(2-cyanophenyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazine-1-carboxylate (Compound 451, first eluting peak).1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (s, 1H), 8.23 (s, 1H), 7.98 (s, 1H), 7.82 (d, J = 7.8 Hz, 1H), 7.78 (s, 1H), 7.74 (t, J = 7.4 Hz, 1H), 7.69 – 7.63 (m, 2H), 7.62 – 7.54 (m, 1H), 7.33 (dd, J = 9.2, 1.5 Hz, 1H), 5.84 (q, J = 6.6 Hz, 1H), 3.87 (s, 3H), 3.76 – 3.66 (m, 2H), 3.56 (t, J = 5.0 Hz, 2H), 3.10 – 2.83 (m, 4H), 1.58 (d, J = 6.6 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C25H25N7O2, 456.2; found, 456.2; HPLC purity: 254 nm: 99.0%. [0727] (S)-1-(2-cyanophenyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazine-1-carboxylate (Compound 452, second eluting peak). 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (s, 1H), 8.23 (s, 1H), 7.98 (s, 1H), 7.82 (d, J = 7.8 Hz, 1H), 7.78 (s, 1H), 7.74 (t, J = 7.4 Hz, 1H), 7.69 – 7.63 (m, 2H), 7.62 – 7.54 (m, 1H), 7.33 (dd, J = 9.2, 1.5 Hz, 1H), 5.84 (q, J = 6.6 Hz, 1H), 3.87 (s, 3H), 3.76 – 3.66 (m, 2H), 3.56 (t, J = 5.0 Hz, 2H), 3.10 – 2.83 (m, 4H), 1.58 (d, J = 6.6 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C25H25N7O2, 456.2; found, 456.2; HPLC purity: 254 nm: 98.3%. Example 105: Synthesis of Exemplary Compound 455 (S)-1-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)-3- phenylbutan-1-one (Compound 455)
Figure imgf000344_0001
[0728] To a stirred mixture of 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1- yl)pyrazolo[1,5-a]pyridine (Compound 12) (300 mg, 1.06 mmol) and (S)-3-phenylbutanoic acid (209.36 mg, 1.27 mmol) in DMF (10 mL) cooled to 0°C were added HATU (606 mg, 1.59 mmol) and DIEA (205.99 mg, 1.59 mmol) in portions then the mixture was warmed to rt and stirred for 2 h. The reaction was concentrated under reduced pressure then the residue was dissolved in water (200 mL) and extracted with EtOAc (2x120 mL). The organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 253.9 mg (56%) of (S)-1-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin- 3-yl)piperazin-1-yl)-3-phenylbutan-1-one (Compound 455) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (t, J = 1.2 Hz, 1H), 8.23 (s, 1H), 7.98 (d, J = 0.8 Hz, 1H), 7.72 (s, 1H), 7.64 (dd, J = 9.3 Hz, 1.0 Hz, 1H), 7.34 (dd, J = 9.2 Hz, 1.6 Hz, 1H), 7.30 (d, J = 4.3 Hz, 4H), 7.22 – 7.16 (m, 1H), 3.88 (s, 3H), 3.70 – 3.51 (m, 4H), 3.27 – 3.16 (m, 1H), 2.97 – 2.88 (m, 2H), 2.87 – 2.78 (m, 1H), 2.79 – 2.70 (m, 1H), 2.68 (d, J = 6.6 Hz, 1H), 2.61 (dd, J = 15.1 Hz, 7.7 Hz, 1H), 1.25 (d, J = 6.9 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C25H28N6O, 429.2; found, 429.2; HPLC purity: 254 nm: 99.6%. [0729] Using the procedures described in Example 105 and reagents, starting materials, and conditions known to those skilled in the art, the following compounds representative of the present disclosure were prepared:
Figure imgf000345_0001
Figure imgf000346_0001
Figure imgf000347_0001
Figure imgf000348_0002
Example 106: Synthesis of Exemplary Compound 457 (S)-3-amino-1-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)- 3-phenylpropan-1-one (Compound 457)
Figure imgf000348_0001
Step 1. Preparation of tert-butyl (S)-(3-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazin-1-yl)-3-oxo-1-phenylpropyl)carbamate
Figure imgf000349_0001
[0730] To a stirred solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1- yl)pyrazolo[1,5-a]pyridine (Compound 12) (300 mg, 1.06 mmol) and (S)-3-((tert- butoxycarbonyl)amino)-3-phenylpropanoic acid (563.8 mg, 2.12 mmol) in DMF (10 mL) cooled to 0°C were added HATU (606 mg, 1.59 mmol) and DIEA (206 mg, 1.59 mmol) in portions then the mixture was warmed to rt and stirred for 1 h. The reaction was diluted with water and extracted with EtOAc. The organic extracts were combined, washed with brine, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by reversed-phase flash chromatography using a C18 silica gel column eluting with 10-50% ACN/water (10mM NH4HCO3) to afford 500 mg (89%) of tert-butyl (S)-(3-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazin-1-yl)-3-oxo-1-phenylpropyl)carbamate as a white solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (s, 1H), 8.23 (s, 1H), 7.98 (s, 1H), 7.70 (s, 1H), 7.65 (d, J = 9.3 Hz, 1H), 7.38 (d, J = 8.6 Hz, 1H), 7.35 – 7.32 (m, 5H), 7.27 – 7.20 (m, 1H), 4.98 (d, J = 7.5 Hz, 1H), 3.88 (s, 3H), 3.64 – 3.49 (m, 4H), 2.91 – 2.82 (m, 4H), 2.78 – 2.62 (m, 2H), 1.36 (s, 9H); MS (ESI) m/z [M+H]+ calcd. for C29H35N7O3, 530.3; found, 530.0. Step 2. Preparation of (S)-3-amino-1-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazin-1-yl)-3-phenylpropan-1-one (Compound 457)
Figure imgf000349_0002
[0731] To a stirred solution of tert-butyl (S)-(3-(4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)-3-oxo-1-phenylpropyl)carbamate (400 mg, 0.75 mmol, as prepared in the previous step) in DCM (9 mL) cooled to 0°C was added TFA (3 mL) dropwise then the mixture was warmed to rt and stirred for 2 h. The reaction was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 146.7 mg (44%) of (S)-3-amino-1-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazin-1-yl)-3-phenylpropan-1-one (Compound 457) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (t, J = 1.2 Hz, 1H), 8.22 (s, 1H), 7.97 (d, J = 0.8 Hz, 1H), 7.72 (s, 1H), 7.64 (dd, J = 9.3, 1.0 Hz, 1H), 7.43 – 7.38 (m, 2H), 7.37 – 7.31 (m, 3H), 7.26 – 7.22 (m, 1H), 4.31 (t, J = 6.6 Hz, 1H), 3.87 (s, 3H), 3.71 – 3.51 (m, 4H), 2.96 – 2.84 (m, 3H), 2.81 – 2.78 (m, 1H), 2.69 (d, J = 6.7 Hz, 2H); MS (ESI) m/z [M+H]+ calcd. for C24H27N7O, 430.2; found, 430.2; HPLC purity: 254 nm: 98.2%. [0732] Using the procedures described in Example 106 and reagents, starting materials, and conditions known to those skilled in the art, the following compounds representative of the present disclosure were prepared:
Figure imgf000350_0001
Example 107: Synthesis of Exemplary Compound 458 and Compound 459 (R)-2-methyl-1-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)- 3-phenylpropan-1-one (Compound 458) and (S)-2-methyl-1-(4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)-3-phenylpropan-1-one (Compound 459)
Figure imgf000351_0001
[0733] To a stirred mixture of 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1- yl)pyrazolo[1,5-a]pyridine (Compound 12) (600 mg, 2.1mmol) and 2-methyl-3- phenylpropanoic acid (418 mg, 2.5) in DMF (10 mL) were added HATU (1.21 g, 3.1mmol) and DIEA (549 mg, 4.2mmol) then the mixture was stirred at rt for 3 h. The reaction was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford racemic 2-methyl-1-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazin-1-yl)-3-phenylpropan-1-one as a yellow solid. [0734] The racemic 2-methyl-1-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin- 3-yl)piperazin-1-yl)-3-phenylpropan-1-one (400 mg, 0.933 mmol) was separated by Chiral-Prep-HPLC to afford 114.3 mg (28%) of (R)-2-methyl-1-(4-(6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)-3-phenylpropan-1-one (Compound 458, first eluting peak) as a yellow solid and 134.7 mg (34%) of (S)-2-methyl- 1-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)-3- phenylpropan-1-one (Compound 459, second eluting peak) as a yellow solid. [0735] (R)-2-methyl-1-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazin-1-yl)-3-phenylpropan-1-one (Compound 458, first eluting peak). 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (s, 1H), 8.23 (s, 1H), 7.98 (s, 1H), 7.67 (s, 1H), 7.60 (d, J = 9.2 Hz, 1H), 7.33 – 7.28 (m, 3H), 7.22 – 7.16 (m, 3H), 3.87 (s, 3H), 3.72 – 3.69 (m, 1H), 3.59 – 3.48 (m, 3H), 3.20 – 3.15 (m, 1H), 2.93 – 2.82 (m, 3H), 2.74 – 2.70 (m, 1H), 2.69 – 2.67 (m, 1H), 2.48 – 2.45 (1H), 1.04 (d, J = 6.4 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C25H28N6O, 429.2; found, 429.2; HPLC purity: 254 nm: 99.5% [0736] (S)-2-methyl-1-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazin-1-yl)-3-phenylpropan-1-one (Compound 459, second eluting peak).1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (s, 1H), 8.23 (s, 1H), 7.98 (s, 1H), 7.67 (s, 1H), 7.60 (d, J = 9.2 Hz, 1H), 7.33 – 7.28 (m, 3H), 7.22 – 7.16 (m, 3H), 3.87 (s, 3H), 3.72 – 3.69 (m, 1H), 3.59 – 3.48 (m, 3H), 3.20 – 3.15 (m, 1H), 2.93 – 2.82 (m, 3H), 2.74 – 2.70 (m, 1H), 2.69 – 2.67 (m, 1H), 2.48 – 2.45 (1H), 1.04 (d, J = 6.4 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C25H28N6O, 429.2; found, 429.2; HPLC purity: 254 nm: 99.0%. [0737] Using the procedures described in Example 107 and reagents, starting materials, and conditions known to those skilled in the art, the following compounds representative of the present disclosure were prepared:
Figure imgf000352_0001
Figure imgf000353_0002
Example 108: Synthesis of Exemplary Compound 460 (S)-1-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)-2- phenylpropan-1-one (Compound 460)
Figure imgf000353_0001
[0738] To a stirred solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1- yl)pyrazolo[1,5-a]pyridine (Compound 12) (200 mg, 0.70 mmol) and (S)-2- phenylpropanoic acid (212.8 mg, 1.41 mmol) in DMF (10 mL) were added TCFH (298.1 mg, 1.06 mmol) and NMI (87.2 mg, 1.06 mmol) in portions at rt then the mixture was stirred for 1 h. The reaction was diluted with water (50 mL) and extracted with EtOAc (3 x50 mL). The organic extracts were combined, washed with brine (2x3 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by reversed-phase flash chromatography using a C18 silica gel column eluting with 10-50% ACN/water (10mM NH4HCO3) to afford 155.6 mg (52%) of (S)-1-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)-2- phenylpropan-1-one (Compound 460) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.80 (t, J = 1.2 Hz, 1H), 8.21 (s, 1H), 7.96 (d, J = 0.8 Hz, 1H), 7.65 (s, 1H), 7.59 (dd, J = 9.3, 1.0 Hz, 1H), 7.36 – 7.28 (m, 5H), 7.25 – 7.23 (m, 1H), 4.16 (q, J = 6.8 Hz, 1H), 3.86 (s, 3H), 3.68 – 3.67 (m, 3H), 3.47 – 3.44 (m, 1H), 2.96 – 2.88 (m, 1H), 2.83 (dd, J = 12.3, 7.8 Hz, 2H), 2.50 – 2.45 (m, 1H), 1.31 (d, J = 6.8 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C24H26N6O, 415.2; found, 415.2; HPLC purity: 254 nm: 97.4%. Example 109: Synthesis of Exemplary Compound 468 (R)-1-(p-tolyl)ethyl (2S,6R)-2,6-dimethyl-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazine-1-carboxylate (Compound 468)
Figure imgf000354_0001
Step 1. Preparation of tert-butyl (2R,6S)-2,6-dimethyl-4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate
Figure imgf000354_0002
[0739] To a stirred mixture of 3-bromo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridine (Compound S9) (1 g, 3.60 mmol) and tert-butyl (2S,6R)-2,6- dimethylpiperazine-1-carboxylate (3.87 g, 18.04 mmol) in dioxane (5 mL) and tBuOH (10mL) were added KOtBu (0.61 g, 5.41 mmol) and tBuXPhos Pd G1 (247 mg, 0.36 mmol) then the mixture was heated to 90°C and stirred overnight under nitrogen. The reaction was cooled to rt and concentrated under reduced pressure then water (300 mL) was added to the residue and the mixture was extracted with EtOAc (2x200 mL). The organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by reversed-phase flash chromatography using a C18 silica gel column eluting with 5-100% ACN/water (10mM NH4HCO3) to afford 400mg400mg400 mg (27%) of tert-butyl (2R,6S)-2,6-dimethyl-4-(6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate as a brown solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.86 (t, J = 1.2 Hz, 1H), 8.23 (d, J = 0.8 Hz, 1H), 7.98 (d, J = 0.8 Hz, 1H), 7.77 (s, 1H), 7.54 (dd, J = 9.2, 1.0 Hz, 1H), 7.37 (dd, J = 9.2, 1.5 Hz, 1H), 4.16 – 4.05 (m, 2H), 3.88 (s, 3H), 3.15 – 3.07 (m, 2H), 2.81 (dd, J = 11.8, 4.3 Hz, 2H), 1.45 (s, 9H), 1.39 (d, J = 6.8 Hz, 6H). Step 2. Preparation of 3-((3R,5S)-3,5-dimethylpiperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine
Figure imgf000355_0001
[0740] A solution of tert-butyl (2R,6S)-2,6-dimethyl-4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (300 mg, 0.73 mmol, as prepared in the previous step) in TFA (1 mL) was stirred at rt for 1 h. The reaction was concentrated under reduced pressure then water (300 mL) was added to the residue and the mixture was extracted with DCM (2x200 mL). The organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure to afford 200 mg (88%) of 3-((3R,5S)-3,5-dimethylpiperazin-1-yl)-6-(1-methyl-1H-pyrazol- 4-yl)pyrazolo[1,5-a]pyridine as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 9.12 (d, J = 10.7 Hz, 1H), 8.85 (s, 1H), 8.25 (s, 1H), 7.99 (s, 1H), 7.84 (s, 1H), 7.74 (d, J = 9.3 Hz, 1H), 7.38 (d, J = 9.2 Hz, 1H), 3.88 (s, 3H), 3.54 (d, J = 9.7 Hz, 2H), 3.46 (d, J = 12.7 Hz, 2H), 2.70 (t, J = 11.6 Hz, 2H), 1.28 (d, J = 6.4 Hz, 6H). Step 3. (R)-1-(p-tolyl)ethyl (2R,6S)-2,6-dimethyl-4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 468)
Figure imgf000356_0001
[0741] To a stirred solution of 3-((3R,5S)-3,5-dimethylpiperazin-1-yl)-6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridine (170 mg, 0.54 mmol, as prepared in the previous step) in THF (10 mL) cooled to 0°C were added DIEA (141.5 mg, 1.09 mmol) and 2,2,2- trichloroethyl chloroformate (232.0 mg, 1.09 mmol) in portions then the mixture was stirred for 1 h. The reaction was concentrated under reduced pressure then water (200 mL) was added to the residue and the resulting mixture was extracted with EtOAc (2x120 mL). The organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was dissolved in DCE (2 mL) and (R)-1-(p-tolyl)ethan-1-ol (111.88 mg, 0.82 mmol) was added. To the stirred solution was added NaH (60% wt., 131.2 mg, 3.28 mmol) in portions at rt then the reaction was heated to 90°C and stirred overnight. The reaction was cooled to rt and concentrated under reduced pressure. Water (200 mL) was added to the residue and the resulting mixture was extracted with DCM (2x120 mL). The organic extracts were combined, dried over anhydrous Na2SO, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-TLC (PE/EtOAc 1:1) and Prep-HPLC to afford 51.3 mg (20%) of (R)-1-(p-tolyl)ethyl (2R,6S)-2,6-dimethyl-4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 468) as a light yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.85 (t, J = 1.3 Hz, 1H), 8.22 (s, 1H), 7.98 (d, J = 0.8 Hz, 1H), 7.78 (s, 1H), 7.54 (dd, J = 9.2, 1.0 Hz, 1H), 7.37 (dd, J = 9.2, 1.5 Hz, 1H), 7.31 – 7.24 (m, 2H), 7.19 (d, J = 7.9 Hz, 2H), 5.79 – 5.70 (m, 1H), 4.26 – 4.17 (m, 2H), 3.88 (s, 3H), 3.12 (d, J = 11.5 Hz, 2H), 2.84 (dd, J = 11.9, 4.4 Hz, 2H), 2.30 (s, 3H), 1.48 (d, J = 6.6 Hz, 3H), 1.41 (t, J = 6.2 Hz, 6H); MS (ESI) m/z [M+H]+ calcd. for C27H32N6O2, 472.3; found,473.3; HPLC purity: 254 nm: 99.6%. [0742] Using the procedures described in Example 109 and reagents, starting materials, and conditions known to those skilled in the art, the following compounds representative of the present disclosure were prepared:
Figure imgf000357_0001
Example 110: Synthesis of Exemplary Compound 470 (R)-1-(p-tolyl)ethyl 2,2-dimethyl-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate (Compound 470)
Figure imgf000358_0001
Step 1. Preparation of tert-butyl 2,2-dimethyl-4-[6-(1-methylpyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl]piperazine-1-carboxylate
Figure imgf000358_0002
[0743] A mixture of 3-bromo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound S9) (1 g, 3.608 mmol) and tert-butyl 2,2-dimethylpiperazine-1-carboxylate (928 mg, 4.330 mmol), Pd-PEPPSI-IHeptCl 3-chloropyridine (303.5 mg, 0.361 mmol), and NaOTMS (1214.3 mg, 10.824 mmol) in dioxane (20 mL) was heated to 100°C and stirred for 2 h under nitrogen. The reaction was cooled to rt and filtered, then the filter cake was washed with EtOAc (2x10 mL). The filtrate was concentrated under reduced pressure then the residue was purified by reversed-phase flash chromatography using a C18 silica gel column eluting with 50-60% ACN/water (10mM NH4HCO3) to afford 677 mg (42%) of tert-butyl 2,2-dimethyl-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.74 (s, 1H), 7.59 (d, J = 9.2 Hz, 1H), 7.31 (dd, J = 9.2, 1.4 Hz, 1H), 3.87 (s, 3H), 3.56 – 3.51 (m, 2H), 3.07 – 3.01 (m, 2H), 2.81 (s, 2H), 1.44 (s, 6H), 1.43 (s, 9H). Step 2. Preparation of 3-(3,3-dimethylpiperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine
Figure imgf000358_0003
[0744] To a mixture of tert-butyl 2,2-dimethyl-4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (677 mg, 1.649 mmol, as prepared in the previous step) in DCM (6 mL) was added TFA (2 mL) then the mixture was stirred at rt for 2 h. To the reaction was added saturated aqueous Na2CO3 solution to adjust the pH to 8, then the mixture was extracted with DCM (3x20mL). The organic extracts were combined, washed with water (3x20 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure to afford 490 mg (92%) of 3-(3,3- dimethylpiperazin-1-yl)-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.81 (s, 1H), 8.21 (s, 1H), 7.96 (s, 1H), 7.73 (s, 1H), 7.55 (d, J = 9.2 Hz, 1H), 7.31 (dd, J = 9.2, 1.3 Hz, 1H), 3.87 (s, 3H), 3.03 – 2.98 (m, 2H), 2.93 – 2.87 (m, 2H), 2.72 (s, 2H), 1.23 (s, 6H). Step 3. Preparation of 2,2,2-trichloroethyl 2,2-dimethyl-4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate
Figure imgf000359_0001
[0745] To a stirred solution of 3-(3,3-dimethylpiperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine (320 mg, 1.031 mmol, as prepared in the previous step) and DIEA (399.7 mg, 3.093 mmol) in THF (10 mL) was added 2,2,2-trichloroethyl chloroformate (305.7 mg, 1.443 mmol) then the mixture was stirred at rt for 2 h. The reaction was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with PE/EtOAc (1:1) to afford 426 mg (88%) of 2,2,2- trichloroethyl 2,2-dimethyl-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.23 (s, 1H), 7.97 (s, 1H), 7.77 (s, 1H), 7.62 (d, J = 9.2 Hz, 1H), 7.33 (d, J = 9.2 Hz, 1H), 4.88 (s, 2H), 3.87 (s, 3H), 3.76 – 3.68 (m, 2H), 3.12 (q, J = 4.9, 4.1 Hz, 2H), 2.90 (s, 2H), 1.30 – 1.25 (m, 6H). Step 4. Preparation of (R)-1-(p-tolyl)ethyl 2,2-dimethyl-4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 470)
Figure imgf000360_0001
[0746] To a solution of (R)-1-(p-tolyl)ethan-1-ol (196.2 mg, 1.440 mmol) in DCM (6 mL) cooled to 0°C was added NaH (60%wt., 144 mg, 3.6 mmol) in portions then the mixture was stirred for 10 minutes under nitrogen. To the stirred reaction was added 2,2,2- trichloroethyl 2,2-dimethyl-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate (350 mg, 0.720 mmol, as prepared in the previous step) at 0°C, then the reaction was warmed to 90°C and stirred for 1 h. The reaction was cooled to rt, quenched by the addition of MeOH (10 mL), and concentrated under reduced pressure. The residue was purified by reversed-phase flash chromatography using a Prep Phenyl column eluting with 35-65% MeOH/water (10mM NH4HCO3) to afford 181.6 mg (53%) of (R)-1- (p-tolyl)ethyl 2,2-dimethyl-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate (Compound 470) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.75 (s, 1H), 7.60 (d, J = 9.1 Hz, 1H), 7.32 (dd, J = 9.2, 1.4 Hz, 1H), 7.26 (d, J = 8.1 Hz, 2H), 7.18 (d, J = 8.0 Hz, 2H), 5.69 (q, J = 6.7 Hz, 1H), 3.87 (s, 3H), 3.70 – 3.55 (m, 2H), 3.09 – 3.01 (m, 2H), 2.83 (s, 2H), 2.30 (s, 3H), 1.47 – 1.44 (m, 9H); MS (ESI) m/z [M+H]+ calcd. for C27H32N6O2, 473.3; found, 473.3; HPLC purity: 254 nm: 98.9%. Example 111: Synthesis of Exemplary Compound 475 1-(4-methoxyphenyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate (Compound 475)
Figure imgf000360_0002
[0747] To a stirred solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1- yl)pyrazolo[1,5-a]pyridine (Compound 12) (300 mg, 1.06 mmol) and 2,2,2-trichloroethyl chloroformate (450.2 mg, 2.12 mmol) in THF (10 mL) was added DIEA (274.6 mg, 2.12 mmol) in portions at rt then the mixture was stirred for 2 h. The reaction was concentrated under reduced pressure then water (200 mL) was added to the residue and the mixture was extracted with EtOAc (2x150 mL). The organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was dissolved in DCE (4 mL). To the stirred solution was added NaH (60%wt., 212.4 mg, 5.31 mmol) and 1-(4-methoxyphenyl)ethan-1-ol (194 mg, 1.27 mmol) in portions at rt then the mixture was heated to 90°C and stirred for 2 h. The reaction was cooled to rt and concentrated under reduced pressure. Water (200 mL) was added to the residue then the resulting mixture was extracted with DCM (2x150 mL). The organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 224.5 mg (46%) of 1-(4-methoxyphenyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 475) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (t, J = 1.2 Hz, 1H), 8.23 (s, 1H), 7.98 (d, J = 0.8 Hz, 1H), 7.77 (s, 1H), 7.65 (dd, J = 9.3, 0.9 Hz, 1H), 7.35 – 7.27 (m, 3H), 6.97 – 6.90 (m, 2H), 5.75 – 5.65 (m, 1H), 3.88 (s, 3H), 3.76 (s, 3H), 3.67 – 3.45 (m, 4H), 2.96 (s, 4H), 1.48 (d, J = 6.6 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C25H28N6O3, 461.2; found, 461.2; HPLC purity: 254 nm: 99.3%. [0748] Using the procedures described in Example 111 and reagents, starting materials, and conditions known to those skilled in the art, the following compounds representative of the present disclosure were prepared:
Figure imgf000361_0001
Example 112: Synthesis of Exemplary Compound 477 (R)-1-(p-tolyl)ethyl (2R,6R)-2,6-dimethyl-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazine-1-carboxylate (Compound 477)
Figure imgf000362_0001
Step 1. Preparation of tert-butyl (2R,6R)-2,6-dimethyl-4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate
Figure imgf000362_0002
[0749] To a stirred solution of 3-bromo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridine (Compound S9) (1 g, 3.608 mmol) and tert-butyl (2R,6R)-2,6- dimethylpiperazine-1-carboxylate (2.32 g, 10.82 mmol) in dioxane (10 mL) and tBuOH (20 mL) were added tBuXPhos Pd G1 (371.69 mg, 0.54 mmol) and KOtBu (809.84 mg, 7.21 mmol) in portions at rt then the mixture was heated to 55°C and stirred overnight under nitrogen. The reaction was cooled to rt and diluted with water (100 mL), then extracted with EtOAc (3x100 mL). The organic extracts were combined, washed with brine (2x2 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by reversed-phase flash chromatography using a C18 silica gel column eluting with 30-60% ACN/water (10mM NH4HCO3) to afford 250 mg (17%) of tert-butyl (2R,6R)-2,6-dimethyl-4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.77 (s, 1H), 8.20 (s, 1H), 7.96 (s, 1H), 7.68 (s, 1H), 7.61 (d, J = 9.2 Hz, 1H), 7.24 (dd, J = 9.3, 1.6 Hz, 1H), 4.04 – 3.93 (m, 2H), 3.87 (s, 3H), 3.42 (dd, J = 11.7, 3.8 Hz, 2H), 3.02 (dd, J = 11.7, 4.3 Hz, 2H), 1.43 (s, 9H), 1.31 (d, J = 6.5 Hz, 6H); MS (ESI) m/z [M+H]+ calcd. for C22H30N6O2, 411.2; found, 411.2. Step 2. Preparation of 3-((3R,5R)-3,5-dimethylpiperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine
Figure imgf000363_0001
[0750] To a stirred solution of tert-butyl (2R,6R)-2,6-dimethyl-4-(6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (250 mg, 0.61 mmol, as prepared in the previous step) in DCM (3 mL) cooled to 0°C was added TFA (1 mL) dropwise then the mixture was warmed to rt and stirred for 1 h. The reaction was concentrated under reduced pressure then the residue was purified by reversed-phase flash chromatography using a C18 silica gel column eluting with 10-50% ACN/water (10mM NH4HCO3) to afford 200 mg (90%) of 3-((3R,5R)-3,5-dimethylpiperazin-1-yl)-6-(1- methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.85 (s, 1H), 8.23 (s, 1H), 7.98 (s, 1H), 7.83 (s, 1H), 7.63 (d, J = 9.2 Hz, 1H), 7.38 (dd, J = 9.2, 1.5 Hz, 1H), 3.87 (s, 3H), 3.78 – 3.70 (m, 2H), 3.21 (dd, J = 12.5, 3.3 Hz, 2H), 2.93 (dd, J = 12.5, 6.3 Hz, 2H), 1.40 (d, J = 6.6 Hz, 6H), 1.17 (t, J = 7.3 Hz, 1H); MS (ESI) m/z [M+H]+ calcd. for C17H22N6, 311.2; found, 311.3. Step 3. Preparation of (R)-4-nitrophenyl (1-(p-tolyl)ethyl) carbonate
Figure imgf000363_0002
[0751] To a stirred solution of 4-nitrophenyl chloroformate (1 g, 4.96 mmol) and (R)-1-(p- tolyl)ethan-1-ol (675.70 mg, 4.96 mmol) in DCM (20 mL) was added pyridine (784.88 mg, 9.92 mmol) dropwise at rt then the mixture was stirred for 2 h. The reaction was concentrated under reduced pressure then the residue was purified by Prep-TLC (PE/EtOAc 1:1) to afford 500 mg (80%) (R)-4-nitrophenyl (1-(p-tolyl)ethyl) carbonate as a yellow solid.1H NMR (400 MHz, CDCl3) δ (ppm) 8.25 (d, J = 8.0 Hz, 2H), 7.37 – 7.32 (m, 4H), 7.21 (d, J = 7.9 Hz, 2H), 5.82 (q, J = 6.6 Hz, 1H), 2.37 (s, 3H), 1.70 (d, J = 6.6 Hz, 3H). Step 4. Preparation of (R)-1-(p-tolyl)ethyl (2R,6R)-2,6-dimethyl-4-(6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 477)
Figure imgf000364_0001
[0752] To a stirred solution of (R)-4-nitrophenyl (1-(p-tolyl)ethyl) carbonate (200 mg, 0.66 mmol, as prepared in the previous step) and 3-((3R,5R)-3,5-dimethylpiperazin-1-yl)-6-(1- methyl-1H-pyrazol-4-yl)pyrazolo [1,5-a]pyridine (206 mg, 0.66 mmol, as prepared in step 2) in ACN (10 mL) was added DIEA (857.9 mg, 6.64 mmol) dropwise at room temperature. The resulting mixture was stirred at rt overnight. The reaction was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 20.1 mg (6%) of (R)-1-(p-tolyl)ethyl (2R,6R)-2,6-dimethyl-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazine-1-carboxylate (Compound 477) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.77 (s, 1H), 8.21 (s, 1H), 7.96 (s, 1H), 7.69 (s, 1H), 7.62 (d, J = 9.3 Hz, 1H), 7.29 – 7.22 (m, 3H), 7.18 (d, J = 7.8 Hz, 2H), 5.73 (q, J = 6.5 Hz, 1H), 4.14 – 4.02 (m, 2H), 3.87 (s, 3H), 3.45 (dd, J = 11.8, 3.7 Hz, 2H), 3.05 (dd, J = 11.8, 4.3 Hz, 2H), 2.30 (s, 3H), 1.47 (d, J = 6.5 Hz, 3H), 1.34 (d, J = 6.5 Hz, 6H); MS (ESI) m/z [M+H]+ calcd. for C27H32N6O2, 473.3; found, 473.20; HPLC purity: 254 nm: 99.4%. Example 113: Synthesis of Exemplary Compound 478 2-hydroxy-1-(p-tolyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate (Compound 478)
Figure imgf000364_0002
Step 1. Preparation of 2-((tert-butyldimethylsilyl)oxy)-1-(p-tolyl)ethyl 4-(6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate
Figure imgf000365_0001
[0753] To a stirred solution of 2-((tert-butyldimethylsilyl)oxy)-1-(p-tolyl)ethan-1-ol (Compound S110) (250 mg, 0.93 mmol) and 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin- 1-yl)pyrazolo[1,5-a]pyridine (Compound 12) (198.7 mg, 0.70 mmol) in pyridine (5 mL) cooled to 0°C was added BTC (278.4 mg, 0.94 mmol) in portions then the mixture was warmed to rt and stirred for 1 h. The reaction was cooled to 0°C and quenched with water, then the mixture was extracted with EtOAc (3x20 mL). The organic extracts were combined, washed with water (3x20 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by reversed- phase flash chromatography using a C18 silica gel column eluting with 45-55% ACN/water (10mM NH4HCO3) to afford 320 mg (59%) of 2-((tert-butyldimethylsilyl)oxy)-1-(p- tolyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1- carboxylate as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.23 (s, 1H), 7.97 (s, 1H), 7.75 (s, 1H), 7.66 (d, J = 9.7 Hz, 1H), 7.33 (dd, J = 9.2, 1.4 Hz, 1H), 7.25 (d, J = 8.1 Hz, 2H), 7.17 (d, J = 8.0 Hz, 2H), 5.69 – 5.61 (m, 1H), 3.87 (s, 3H), 3.83 – 3.75 (m, 2H), 3.69 (s, 2H), 3.51 (s, 2H), 2.97 (s, 4H), 2.29 (s, 3H), 0.84 (s, 9H), 0.01 (s, 6H); MS (ESI) m/z [M+H]+ calcd. For C31H42N6O3Si, 575.3; found, 575.3. Step 2. Preparation of 2-hydroxy-1-(p-tolyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 478)
Figure imgf000365_0002
[0754] A solution of 2-((tert-butyldimethylsilyl)oxy)-1-(p-tolyl)ethyl 4-(6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (300 mg, 0.52 mmol, as prepared in the previous step) in THF (8 mL) was added Et3N·3HF (3 mL) then the mixture was stirred at rt for 2 h. To the reaction was added saturated aqueous Na2CO3 solution to adjust the pH to 8 then the mixture was extracted with EtOAc (3x20 mL). The organic extracts were combined, washed with water (3x20 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by reversed-phase flash chromatography using a Prep Phenyl column eluting with35with35with 35-65% MeOH/water (10mM NH4HCO3) to afford 151.8 mg (63%) of 2-hydroxy-1-(p-tolyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate (Compound 478) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.77 (s, 1H), 7.65 (d, J = 9.3 Hz, 1H), 7.33 (dd, J = 9.2, 1.6 Hz, 1H), 7.22 (d, J = 8.0 Hz, 2H), 7.16 (d, J = 8.0 Hz, 2H), 5.59 – 5.54 (m, 1H), 5.01 (t, J = 6.0 Hz, 1H), 3.87 (s, 3H), 3.74 – 3.48 (m, 6H), 2.96 (s, 4H), 2.28 (s, 3H); MS (ESI) m/z [M+H]+ calcd. For C25H28N6O3, 461.2; foumd, 461.2; HPLC purity: 254 nm: 99.9%. Example 114: Synthesis of Exemplary Compound 479 (R)-O-(1-(p-tolyl)ethyl) 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carbothioate (Compound 479)
Figure imgf000366_0001
Step 1. Preparation of (1H-imidazol-1-yl)(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazin-1-yl)methanethione
Figure imgf000366_0002
[0755] To a stirred solution of 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1- yl)pyrazolo[1,5-a]pyridine (Compound 12) (200 mg, 0.7 mmol) and di(1H-imidazol-1- yl)methanethione (189.3 mg, 1.06 mmol) in DMF (10 mL) was added TEA (143.3 mg, 1.41 mmol) in portions at rt then the mixture was stirred for 2 h. The reaction was concentrated under reduced pressure then the water (200 mL) was added to the residue and the resulting mixture was extracted with EtOAc (2x150 mL). The organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-TLC (PE/EtOAc 1:1) to afford 250 mg (90%) of (1H- imidazol-1-yl)(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1- yl)methanethione as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.84 (t, J = 1.2 Hz, 1H), 8.24 (s, 1H), 8.09 (d, J = 1.1 Hz, 1H), 7.98 (s, 1H), 7.81 (s, 1H), 7.71 (dd, J = 9.3, 1.0 Hz, 1H), 7.57 (t, J = 1.4 Hz, 1H), 7.36 (dd, J = 9.3, 1.5 Hz, 1H), 7.07 (t, J = 1.2 Hz, 1H), 4.11 (br s, 4H), 3.87 (s, 3H), 3.18 (br s, 4H). Step 2. (R)-O-(1-(p-tolyl)ethyl) 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carbothioate (Compound 479)
Figure imgf000367_0001
[0756] To a stirred mixture of (1H-imidazol-1-yl)(4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)methanethione (250 mg, 0.63 mmol, as prepared in the previous step) and (R)-1-(p-tolyl)ethan-1-ol (104.1 mg, 0.76 mmol) in DMF (10 mL) was added Cs2CO3 (415 mg, 1.27 mmol) in portions at rt then the mixture was stirred overnight. The reaction was concentrated under reduced pressure then water (300 mL) was added to the residue and the resulting mixture was extracted with DCM (2x200 mL). The organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep- HPLC to afford 107.3 mg (37%) of (R)-O-(1-(p-tolyl)ethyl) 4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carbothioate (Compound 479) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.84 (t, J = 1.2 Hz, 1H), 8.24 (s, 1H), 7.98 (d, J = 0.8 Hz, 1H), 7.79 (s, 1H), 7.68 (dd, J = 9.2, 0.9 Hz, 1H), 7.35 (dd, J = 9.3, 1.5 Hz, 1H), 7.32 – 7.26 (m, 2H), 7.19 (d, J = 7.9 Hz, 2H), 6.49 – 6.40 (m, 1H), 4.20 – 4.11 (m, 2H), 4.04 – 3.91 (m, 2H), 3.87 (s, 3H), 3.05 – 2.95 (m, 4H), 2.30 (s, 3H), 1.56 (d, J = 6.6 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C25H28N6OS, 461.2; found, 461.2; HPLC purity: 254 nm: 99.7%. Example 115: Synthesis of Exemplary Compound 480 1-(4-(hydroxymethyl)phenyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate (Compound 480)
Figure imgf000368_0001
Step 1. Preparation of 1-(4-(((tert-butyldimethylsilyl)oxy)methyl)phenyl)ethan-1-one
Figure imgf000368_0002
[0757] To a stirred solution of 1-(4-(hydroxymethyl)phenyl)ethan-1-one (600 mg, 4.0 mmol) and imidazole (544 mg, 8.0 mmol) in THF (10 mL) cooled to 0°C was added TBSCl (1.2 g, 8.0 mmol) in portions then the mixture was warmed to rt and stirred for 2 h. The reaction was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with PE/EtOAc (5:1) to afford 700 mg (66%) of 1-(4- (((tert-butyldimethylsilyl)oxy)methyl)phenyl)ethan-1-one as a colorless oil.1H NMR (400 MHz, DMSO-d6) δ (ppm) 7.94 (d, J = 8.1 Hz, 2H), 7.44 (d, J = 8.1 Hz, 2H), 4.79 (s, 2H), 2.56 (s, 3H), 0.91 (s, 9H), 0.09 (s, 6H); MS (ESI) m/z [M+H]+ calcd. for C15H24O2Si, 265.1; found, 265.1. Step 2.1-(4-(((tert-butyldimethylsilyl)oxy)methyl)phenyl)ethan-1-ol
Figure imgf000368_0003
[0758] To a solution of 1-(4-(((tert-butyldimethylsilyl)oxy)methyl)phenyl)ethan-1-one (700 mg, 2.6 mmol, as prepared in the previous step) in MeOH (10 mL) cooled to 0°C was added NaBH4 (120 mg, 3.1 mmol) then the mixture was stirred for 2 h. The reaction was concentrated under reduced pressure then the residue was purified by Prep-TLC (PE/EtOAc 3:1) to afford 550 mg (78%) of 1-(4-(((tert-butyldimethylsilyl)oxy)methyl)phenyl)ethan-1- ol as colorless oil.1H NMR (400 MHz, DMSO-d6) δ (ppm) 7.31 (d, J = 8.1 Hz, 2H), 7.24 (d, J = 8.2 Hz, 2H), 5.10 (d, J = 4.2 Hz, 1H), 4.71 – 4.58 (m, 3H), 1.31 (d, J = 6.4 Hz, 3H), 0.91 (s, 9H), 0.08 (s, 6H); MS (ESI) m/z [M+H]+ calcd. for C15H26O2Si, 267.2; found, 267.2. Step 3. 1-(4-(((tert-butyldimethylsilyl)oxy)methyl)phenyl)ethyl 4-(6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate
Figure imgf000369_0001
[0759] To a stirred solution of 1-(4-(((tert-butyldimethylsilyl)oxy)methyl)phenyl)ethan-1- ol (530 mg, 2.0 mmol, as prepared in the previous step) and 4-nitrophenyl 4-[6-(1-methyl- 1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl]piperazine-1-carboxylate (0.89 g, 2.0 mmol, as prepared in Example 20, Step 1) in DMF (10 mL) was added Cs2CO3 (1.3 g, 4.0 mmol) then the mixture was stirred at rt overnight. The reaction was quenched with water (200 mL) and extracted with EtOAc (3x100 mL). The organic extracts were combined, washed with brine (3x50 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with PE/EtOAc (1:1) to afford 430 mg (37%) of 1-(4-(((tert- butyldimethylsilyl)oxy)methyl)phenyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.81 (s, 1H), 8.21 (s, 1H), 7.96 (s, 1H), 7.76 (s, 1H), 7.65 (dd, J = 9.3, 0.9 Hz, 1H), 7.39 – 7.21 (m, 5H), 5.73 (q, J = 6.5 Hz, 1H), 4.69 (s, 2H), 3.87 (s, 3H), 2.96 (s, 4H), 2.07 (s, 10H), 1.48 (d, J = 6.5 Hz, 3H), 0.90 (s, 9H); MS (ESI) m/z [M+H]+ calcd. for C31H42N6O3Si, 575.3; found, 575.3. Step 4. 1-(4-(hydroxymethyl)phenyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazine-1-carboxylate (Compound 480)
Figure imgf000369_0002
[0760] To a solution of 1-(4-(((tert-butyldimethylsilyl)oxy)methyl)phenyl)ethyl 4-(6-(1- methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (300 mg, 0.522 mmol, as prepared in the previous step) was added Et3N·3HF (2 mL) then the mixture was stirred at rt for 2 h. A saturated aqueous NaHCO3 solution was added to the reaction to adjust the pH to 8 then the mixture was extracted with DCM (3x100 mL). The organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 140.7 mg (59%) of 1-(4-(hydroxymethyl)phenyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 480) as an off-white solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (s, 1H), 8.23 (s, 1H), 7.97 (s, 1H), 7.77 (s, 1H), 7.65 (d, J = 9.2 Hz, 1H), 7.39 – 7.21 (m, 5H), 5.73 (q, J = 6.5 Hz, 1H), 5.16 (t, J = 5.7 Hz, 1H), 4.49 (d, J = 5.7 Hz, 2H), 3.87 (s, 3H), 3.69 (s, 4H), 2.97 (s, 4H), 1.49 (d, J = 6.6 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C25H28N6O3, 461.2; found, 461.2; HPLC purity: 254 nm: 99.6%. Example 116: Synthesis of Exemplary Compound 482 1-(4-(fluoromethyl)phenyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate (Compound 482)
Figure imgf000370_0001
[0761] To a stirred solution of 1-(4-(fluoromethyl)phenyl)ethan-1-ol (Compound S108) (103.3 mg, 0.67 mmol) and 4-nitrophenyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazine-1-carboxylate (300 mg, 0.67 mmol, as prepared in Example 20, Step 1) in DCE (5 mL) cooled to 0°C was added NaH (60%wt., 134 mg, 3.35 mmol) then the mixture was warmed to rt and stirred for 2 h. The reaction was poured into water (200 mL) and extracted with DCM (3x50 mL). The organic extracts were combined and concentrated under reduced pressure. The residue was purified by Prep-HPLC to afford 135 mg (44%) of 1-(4-(fluoromethyl)phenyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 482) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.84 (t, J = 1.2 Hz, 1H), 8.24 (s, 1H), 7.98 (d, J = 0.8 Hz, 1H), 7.78 (s, 1H), 7.66 (d, J = 9.3 Hz, 1H), 7.44 (s, 4H), 7.34 (dd, J = 9.3, 1.5 Hz, 1H), 5.84 – 5.69 (m, 1H), 5.41 (d, J = 48 Hz, 2H), 3.88 (s, 3H), 3.78 – 3.47 (m, 4H), 2.98 (s, 4H), 1.50 (d, J = 6.6 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C25H27FN6O2, 463.2; found, 463.2; HPLC purity: 254 nm: 99.6%. [0762] Using the procedures described in Example 116 and reagents, starting materials, and conditions known to those skilled in the art, the following compounds representative of the present disclosure were prepared:
Figure imgf000371_0002
Example 117: Synthesis of Exemplary Compound 484 1-(1,3-dimethyl-1H-pyrazol-5-yl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazine-1-carboxylate (Compound 484)
Figure imgf000371_0001
[0763] To a stirred solution of 4-nitrophenyl 4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (250 mg, 0.56 mmol, as prepared in Example 20, Step 1) and 1-(1,3-dimethyl-1H-pyrazol-5-yl)ethan-1-ol (391.6 mg, 2.80 mmol) in DMF (10 mL) was added Cs2CO3 (364 mg, 1.12 mmol) in portions then the mixture was stirred at rt overnight. The reaction was diluted with water (100 mL) and extracted with EtOAc (3x150 mL). The organic extracts were combined, washed with brine (3x5 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by reversed-phase flash chromatography using a C18 silica gel column eluting with 30-60% ACN/water (10mM NH4HCO3) to afford 151.2 mg (59%) of 1-(1,3-dimethyl-1H-pyrazol-5-yl)ethyl 4-(6-(1-methyl-1H- pyrazol-4-yl) pyrazolo [1,5-a]pyridin-3-yl) piperazine-1-carboxylate (Compound 484) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (t, J = 1.2 Hz, 1H), 8.22 (s, 1H), 7.97 (d, J = 0.8 Hz, 1H), 7.76 (s, 1H), 7.64 (dd, J = 9.3, 1.0 Hz, 1H), 7.32 (dd, J = 9.3, 1.6 Hz, 1H), 6.10 (s, 1H), 5.84 (q, J = 6.5 Hz, 1H), 3.86 (s, 3H), 3.71 (s, 3H), 3.55 (t, J = 5.1 Hz, 4H), 2.95 (s, 4H), 2.10 (s, 3H), 1.52 (d, J = 6.6 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C23H28N8O2, 449.2; found, 449.3; HPLC purity: 254 nm: 98.1%. [0764] Using the procedures described in Example 117 and reagents, starting materials, and conditions known to those skilled in the art, the following compounds representative of the present disclosure were prepared:
Figure imgf000372_0003
Example 118: Synthesis of Exemplary Compound 486 (R)-1-(p-tolyl)ethyl 2-(hydroxymethyl)-4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazine-1-carboxylate (Compound 486)
Figure imgf000372_0001
Step 1. Preparation of tert-butyl 2-(((tert-butyldimethylsilyl)oxy)methyl)piperazine-1- carboxylate
Figure imgf000372_0002
[0765] To a stirred solution of tert-butyl 2-(hydroxymethyl)piperazine-1-carboxylate (6 g, 27.74 mmol) and imidazole (3.78 g, 55.48 mmol) in THF (100 mL) cooled to 0°C was added TBSCl (6.27 g, 41.61 mmol) in portions then the mixture was warmed to rt and stirred for 2 h. The reaction was diluted with water (500 mL) and extracted with EtOAc (3x500 mL). The organic extracts were combined, washed with brine (3x10 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with PE/EtOAc (1:1) to afford 8 g (87%) of tert-butyl 2-(((tert-butyldimethylsilyl)oxy)methyl)piperazine-1- carboxylate as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 3.93 – 3.74 (m, 2H), 3.63 (d, J = 12.8 Hz, 1H), 3.58 – 3.49 (m, 1H), 2.95 (d, J = 12.0 Hz, 1H), 2.79 (d, J = 11.2 Hz, 2H), 2.53 – 2.50 (m, 1H), 2.41 (td, J = 12.0, 3.2 Hz, 1H), 1.39 (s, 9H), 0.87 (s, 9H), 0.05 (s, 6H). Step 2. Preparation of tert-butyl 2-(((tert-butyldimethylsilyl)oxy)methyl)-4-(6-(1-methyl- 1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate
Figure imgf000373_0001
[0766] To a stirred solution of tert-butyl 2-(((tert- butyldimethylsilyl)oxy)methyl)piperazine-1-carboxylate (6 g, 18.15 mmol, as prepared in the previous step) and 3-bromo-6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (Compound S9) (5.03 g, 18.15 mmol) in tBuOH (80 mL) and dioxane (40 mL) were added tBuXPhos Pd G1 (1.53 g, 1.81 mmol) and KOtBu (4.07 g, 36.30 mmol) in portions under nitrogen then the mixture was heated to 90°C and stirred for 2 h under nitrogen. The reaction was cooled to rt, diluted with water (300 mL), and extracted with EtOAc (3x300 mL). The organic extracts were combined, washed with brine (2x10 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with PE/EtOAc (1:1) to afford 3 g (31%) of tert-butyl 2-(((tert-butyldimethylsilyl)oxy)methyl)-4-(6-(1-methyl- 1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (s, 1H), 8.24 (s, 1H), 7.97 (s, 1H), 7.75 (s, 1H), 7.59 (d, J = 9.2 Hz, 1H), 7.29 (dd, J = 9.2, 1.2 Hz, 1H), 4.17 – 3.97 (m, 2H), 3.87 (s, 4H), 3.68 – 3.62 (m, 1H), 3.48 – 3.45 (m, 1H), 3.27 – 3.08 (m, 2H), 2.73 – 2.62 (m, 2H), 1.43 (s, 9H), 0.90 (s, 9H), 0.10 (s, 6H). Step 3. Preparation of 3-(3-(((tert-butyldimethylsilyl)oxy)methyl)piperazin-1-yl)-6-(1- methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine
Figure imgf000374_0001
[0767] To a stirred solution of tert-butyl 2-(((tert-butyldimethylsilyl)oxy)methyl)-4-(6-(1- methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (2 g, 3.79 mmol, as prepared in the previous step) in ACN (40 mL) cooled to 0°C was added TMSI (1.60 g, 11.39 mmol) in portions then the mixture was warmed to rt and stirred at rt overnight. The reaction was concentrated under reduced pressure then the residue was purified by reversed-phase flash chromatography using a C18 silica gel column eluting with 10-80% ACN/water (10mM NH4HCO3) to afford 320 mg (20%) of 3-(3-(((tert- butyldimethylsilyl)oxy)methyl)piperazin-1-yl)-6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridine as a brown solid. MS (ESI) m/z [M+H]+ calcd. for C22H34N6OSi, 427.3; found, 427.3. Step 4. Preparation of (R)-1-(p-tolyl)ethyl 2-(((tert-butyldimethylsilyl)oxy)methyl)-4-(6- (1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate
Figure imgf000374_0002
[0768] To a stirred solution of 3-(3-(((tert-butyldimethylsilyl)oxy)methyl)piperazin-1-yl)- 6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridine (160 mg, 0.37 mmol, as prepared in the previous step) and (R)-4-nitrophenyl (1-(p-tolyl)ethyl) carbonate (113.0 mg, 0.37 mmol, as prepared in Example 112, Step 3) in ACN (3 mL) was added DIEA (96.9 mg, 0.75 mmol) dropwise then the mixture was heated to 60°C and stirred for 48 h. The reaction was cooled to 0°C, quenched by the addition of water (20 mL), and extracted with EtOAc (3x30 mL). The organic extracts were combined, washed with brine (3x10 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by reversed-phase flash chromatography using a C18 silica gel column eluting with 50-100% ACN/water (10mM NH4HCO3) to afford 170 mg (77%) of (R)-1-(p-tolyl)ethyl 2-(((tert-butyldimethylsilyl)oxy)methyl)-4-(6-(1-methyl-1H-pyrazol- 4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate as a brown solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.84 (s, 1H), 8.25 (s, 1H), 7.99 (s, 1H), 7.77 (s, 1H), 7.60 (dd, J = 9.4, 3.4 Hz, 1H), 7.35 – 7.24 (m, 3H), 7.18 (d, J = 7.9 Hz, 2H), 5.76 – 5.68 (m, 1H), 4.19 – 4.06 (m, 2H), 3.88 (s, 3H), 3.48 (d, J = 11.7 Hz, 1H), 3.27 – 3.16 (m, 2H), 2.77 – 2.66 (m, 2H), 2.30 (s, 3H), 1.48 (d, J = 6.5 Hz, 3H), 1.25 (s, 2H), 0.88 (s, 9H), 0.01 (s, 6H). Step 5. Preparation of (R)-1-(p-tolyl)ethyl 2-(hydroxymethyl)-4-(6-(1-methyl-1H-pyrazol- 4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 486)
Figure imgf000375_0001
[0769] To a stirred solution of TBAF (124.3 mg, 0.47 mmol) in THF (2 mL) was added (R)-1-(p-tolyl)ethyl 2-(((tert-butyldimethylsilyl)oxy)methyl)-4-(6-(1-methyl-1H-pyrazol- 4-yl) pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (140 mg, 0.23 mmol, as prepared in the previous step) in portions then the mixture was stirred at rt for 1 h. The reaction was concentrated under reduced pressure then the residue was purified by Prep- HPLC to afford 68.5 mg (61%) of (R)-1-(p-tolyl)ethyl 2-(hydroxymethyl)-4-(6-(1-methyl- 1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 486) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.75 (s, 1H), 7.64 (d, J = 9.2 Hz, 1H), 7.34 (dd, J = 9.2, 1.2 Hz, 1H), 7.30 – 7.23 (m, 2H), 7.17 (d, J = 7.6 Hz, 2H), 5.76 – 5.65 (m, 1H), 5.01 (s, 1H), 4.21 – 4.04 (m, 1H), 3.98 – 3.89 (m, 2H), 3.87 (s, 3H), 3.50 (d, J = 11.6 Hz, 2H), 3.27 – 3.09 (m, 2H), 2.78 – 2.57 (m, 2H), 2.29 (s, 3H), 1.46 (d, J = 6.6 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C26H30N6O3, 475.2; found, 475.3; HPLC purity: 254 nm: 98.9%. Example 119: Synthesis of Exemplary Compound 487 1-(5-methyl-1H-pyrazol-3-yl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate (Compound 487)
Figure imgf000376_0001
Step 1. Preparation of 1-(5-methyl-1H-pyrazol-3-yl)ethan-1-one
Figure imgf000376_0002
[0770] To a solution of TEA (9.85 g, 97.29 mmol) and CH3MgI in Et2O (3M, 32 mL, 97.29 mmol) in THF (130 mL) was added a solution of ethyl 5-methyl-1H-pyrazole-3- carboxylate (5 g, 32.43 mmol) in THF (10 mL) cooled to -10°C dropwise then the mixture was warmed to rt and stirred for 2 h. The reaction was acidified to pH=4 with aqueous HCl solution then the mixture was extracted with EtOAc (3x200 mL). The organic extracts were combined, washed with water (3x200 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by reversed- phase flash chromatography using a C18 silica gel column eluting with 5-10% ACN/water (10mM NH4HCO3) to afford 1.6 g (38%) of 1-(5-methyl-1H-pyrazol-3-yl)ethan-1-one as yellow liquid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 13.13 (s, 1H), 6.42 (s, 1H), 2.43 (s, 3H), 2.26 (s, 3H). Step 2. Preparation of 1-(5-methyl-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazol-3- yl)ethan-1-one
Figure imgf000376_0003
[0771] To a mixture of 1-(5-methyl-1H-pyrazol-3-yl)ethan-1-one (1.6 g, 12.88 mmol, as prepared in the previous step), SEM-Cl (1.07 g, 64.44 mmol) in DCM (20 mL) was added Cs2CO3 (8.40 mg, 25.77 mmol) then the reaction was stirred at rt for 2 h. Water was added and the mixture was extracted with DCM (3x20 mL). The organic extracts were combined, washed with water (3x20 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by reversed-phase flash chromatography using a C18 silica gel column eluting with 60-70% ACN/water (10mM NH4HCO3) to afford 470 mg (14%) of 1-(5-methyl-1-((2-(trimethylsilyl)ethoxy)methyl)- 1H-pyrazol-3-yl)ethan-1-one as yellow liquid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 6.54 (s, 1H), 5.48 (s, 2H), 3.56 (t, J=8.0 Hz, 2H), 2.45 (s, 3H), 2.34 (s, 3H), 0.86 – 0.84 (m, 2H), -0.05 (s, 9H). Step 3. Preparation of 1-(5-methyl-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazol-3- yl)ethan-1-ol
Figure imgf000377_0001
[0772] To a solution of 1-(5-methyl-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazol-3- yl)ethan-1-one (420 mg, 1.65 mmol, as prepared in the previous step) in MeOH (10 mL) cooled to 0°C was added NaBH4 (93.6 mg, 2.47 mmol) in portions then the mixture was warmed to rt and stirred for 2 h. The reaction was cooled to 0°C, quenched with water, and extracted with EtOAc (3x30 mL). The organic extracts were combined, washed with water (3x30 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure to afford 400 mg (94%) of 1-(5-methyl-1-((2- (trimethylsilyl)ethoxy)methyl)-1H-pyrazol-3-yl)ethan-1-ol as yellow liquid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 6.02 (s, 1H), 5.28 (d, J = 2.4 Hz, 2H), 4.93 (d, J = 4.9 Hz, 1H), 4.65 – 4.57 (m, 1H), 3.49 (t, J = 8.0 Hz, 2H), 2.25 (s, 3H), 1.30 (d, J = 6.5 Hz, 3H), 0.85 – 0.75 (m, 2H), -0.06 (s, 9H). Step 4. Preparation of 1-(5-methyl-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazol-3- yl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1- carboxylate
Figure imgf000377_0002
[0773] A solution of 1-(5-methyl-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazol-3- yl)ethan-1-ol (200 mg, 0.78 mmol, as prepared in the previous step) in DMF (5 mL) cooled to 0°C was added NaH (60% wt., 93.6 mg, 2.34 mmol) in portions then the mixture was stirred for 10 minutes under nitrogen. The reaction was warmed to rt and 4-nitrophenyl 4- (6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (349.0 mg, 0.78 mmol, as prepared in Example 20, Step 1) in portions then the mixture was stirred at rt for 3 h. The reaction was cooled to 0°C, quenched with water, and extracted with EtOAc (3x30 mL). The organic extracts were combined, washed with water (3x30 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by reversed-phase flash chromatography using a C18 silica gel column eluting with 55-65% ACN/water (10mM NH4HCO3) to afford 380 mg (85%) of 1-(5-methyl-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazol-3- yl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1- carboxylate as yellow oil. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.22 (s, 1H), 7.96 (s, 1H), 7.76 (s, 1H), 7.64 (d, J = 9.3 Hz, 1H), 7.32 (dd, J = 9.3, 1.3 Hz, 1H), 6.11 (s, 1H), 5.70 (q, J = 6.5 Hz, 1H), 5.32 (s, 2H), 3.86 (s, 3H), 3.55 (s, 4H), 3.52 – 3.47 (m, 2H), 2.95 (s, 4H), 2.27 (s, 3H), 1.48 (d, J = 6.6 Hz, 3H), 0.82 – 0.76 (m, 2H), -0.07 (s, 9H). Step 5. Preparation of 1-(5-methyl-1H-pyrazol-3-yl)ethyl 4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 487)
Figure imgf000378_0001
[0774] To a solution of 1-(5-methyl-1-((2-(trimethylsilyl)ethoxy)methyl)-1H-pyrazol-3- yl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1- carboxylate (340 mg, 0.60 mmol, as prepared in the previous step) in DCM (6 mL) was added TFA (2 mL) then the mixture was stirred at rt for 2 h. The pH of the reaction was adjusted to pH 8 with saturated aqueous Na2CO3 solution then the mixture was extracted with DCM (3x20 mL). The organic extracts were combined, washed with water (3x20 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by reverse phase flash chromatography using a Prep OBD RP18 column eluting with 17-37% ACN/water (10mM NH4HCO3) to afford 146.2 mg (56%) of 1-(5-methyl-1H-pyrazol-3-yl)ethyl 4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 487) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 12.35 (s, 1H), 8.82 (s, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.76 (s, 1H), 7.64 (d, J = 9.3 Hz, 1H), 7.32 (dd, J = 9.2, 1.3 Hz, 1H), 5.99 (s, 1H), 5.76-5.71 (m, 1H), 3.86 (s, 3H), 3.55 (s, 4H), 2.95 (s, 4H), 2.19 (s, 3H), 1.48 (d, J = 6.8 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. For C22H26N8O2, 435.2; found, 435.1; HPLC purity: 254 nm: 96.8%. Example 120: Synthesis of Exemplary Compound 488 1-(1,5-dimethyl-1H-pyrazol-3-yl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazine-1-carboxylate (Compound 488)
Figure imgf000379_0001
[0775] To a solution of 1-(1,5-dimethyl-1H-pyrazol-3-yl)ethan-1-ol (Compound S112) (130 mg, 0.92 mmol) in DMF (8 mL) cooled to 0°C was added NaH (60% wt., 111.2 mg, 2.78 mmol) then the mixture was stirred for 10 minutes under nitrogen. The reaction was warmed to rt then 4-nitrophenyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate (497.9 mg, 1.11 mmol, as prepared in Example 20, Step 1) was added then the mixture was stirred at rt for 1 h. The reaction was cooled to 0°C, quenched with water, and extracted with EtOAc (3x20 mL). The organic extracts were combined, washed with water (3x20 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by reverse phase flash chromatography using a Prep OBD C18 column eluting with 21-41% ACN/water (10mM NH4HCO3) to afford 144.5 mg (35%) of 1-(1,5-dimethyl-1H-pyrazol-3-yl)ethyl 4-(6-(1- methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 488) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.76 (s, 1H), 7.64 (d, J = 9.3 Hz, 1H), 7.32 (dd, J = 9.2, 1.4 Hz, 1H), 6.02 (s, 1H), 5.68 (q, J = 6.6 Hz, 1H), 3.87 (s, 3H), 3.67 (s, 3H), 3.54 (s, 4H), 2.95 (s, 4H), 2.22 (s, 3H), 1.47 (d, J = 6.6 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. For C23H28N8O2, 449.2; found, 449.1; HPLC purity: 254 nm: 99.8%. [0776] Using the procedures described in Example 120 and reagents, starting materials, and conditions known to those skilled in the art, the following compounds representative of the present disclosure were prepared:
Figure imgf000380_0001
Figure imgf000381_0001
Figure imgf000382_0001
Figure imgf000383_0003
Example 121: Synthesis of Exemplary Compound 494 and Compound 499 3-hydroxy-1-(p-tolyl)propyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate (Compound 494) and 3-hydroxy-3-(p-tolyl)propyl 4-(6-(1- methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 499)
Figure imgf000383_0001
Step 1. Preparation of 3-((tert-butyldimethylsilyl)oxy)-1-(p-tolyl)propan-1-ol
Figure imgf000383_0002
[0777] To a stirred mixture of 1-(p-tolyl)propane-1,3-diol (500 mg, 3 mmol) and imidazole (409.5 mg, 6.01 mmol) in THF (10 mL) was added TBSCl (453.3 mg, 3 mmol) in portions at rt then the mixture was stirred for 2 h. The reaction was concentrated under reduced pressure then water 200 mL) was added to the residue and the mixture was extracted with EtOAc (2x150 mL). The organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure to afford 500 mg (59%) of 3-((tert-butyldimethylsilyl)oxy)-1-(p-tolyl)propan-1-ol as a yellow solid. The crude product was used for the next step directly without further purification. Step 2. Preparation of the mixture of 3-((tert-butyldimethylsilyl)oxy)-1-(p-tolyl)propyl 4- (6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate and 3-((tert-butyldimethylsilyl)oxy)-3-(p-tolyl)propyl 4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate
Figure imgf000384_0001
[0778] To a stirred mixture of 3-((tert-butyldimethylsilyl)oxy)-1-(p-tolyl)propan-1-ol (250 mg, 0.89 mmol, as prepared in the previous step) and 4-nitrophenyl 4-(6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (478.5 mg, 1.06 mmol, as prepared in Example 20, Step 1) in DMF (10 mL) was added NaH (60% wt., 71.2 mg, 1.78 mmol) in portions at rt then the mixture was stirred for 2 h. The reaction was quenched with water (200 mL) and extracted with DCM (2x150 mL). The organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-TLC (DCM/MeOH 10:1) to afford 500 mg (95%) of a mixture of 3-((tert-butyldimethylsilyl)oxy)-1-(p-tolyl)propyl 4-(6-(1- methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate and 3- ((tert-butyldimethylsilyl)oxy)-3-(p-tolyl)propyl 4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate as a yellow solid. MS (ESI) m/z [M+H]+ calcd. for C32H44N6O3Si, 589.3; found, 588.8. Step 3. 3-hydroxy-1-(p-tolyl)propyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazine-1-carboxylate (Compound 494) and 3-hydroxy-3-(p-tolyl)propyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 499)
Figure imgf000384_0002
[0779] To a stirred solution of the mixture of 3-((tert-butyldimethylsilyl)oxy)-1-(p- tolyl)propyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1- carboxylate and 3-((tert-butyldimethylsilyl)oxy)-3-(p-tolyl)propyl 4-(6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (250 mg, 0.42 mmol, as prepared in the previous step) in THF (2 mL) cooled to 0°C was added TEA·HF (1 mL) dropwise, then the mixture was warmed to rt and stirred for 2 h. The reaction was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 98.1 mg (49%) of 3-hydroxy-1-(p-tolyl)propyl 4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 494) as a yellow solid and 82.3 mg (41%) of 3-hydroxy-3-(p-tolyl)propyl 4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 499) as a yellow solid. [0780] 3-hydroxy-1-(p-tolyl)propyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazine-1-carboxylate (Compound 494).1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (s, 1H), 8.23 (s, 1H), 7.97 (s, 1H), 7.78 (s, 1H), 7.63 (d, J = 9.2 Hz, 1H), 7.34 (dd, J = 9.6, 1.6 Hz, 1H), 7.24 (d, J = 8.1 Hz, 2H), 7.18 (d, J = 8.0 Hz, 2H), 5.71 (dd, J = 8.3, 5.6 Hz, 1H), 4.57 (t, J = 5.0 Hz, 1H), 3.88 (s, 3H), 3.69 – 3.47 (m, 3H), 3.50 – 3.42 (m, 2H), 3.42 – 3.35 (m, 1H), 2.95 (s, 4H), 2.29 (s, 3H), 2.08 – 1.98 (m, 1H), 1.89 – 1.78 (m, 1H); MS (ESI) m/z [M+H]+ calcd. for C26H30N6O3, 475.2; found,475.2; HPLC purity: 254 nm: 99.7%. [0781] 3-hydroxy-3-(p-tolyl)propyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazine-1-carboxylate (Compound 499).1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (s, 1H), 8.24 (s, 1H), 7.97 (s, 1H), 7.78 (s, 1H), 7.66 (d, J = 9.2 Hz, 1H), 7.34 (dd, J = 9.3, 1.5 Hz, 1H), 7.24 (d, J = 8.0 Hz, 2H), 7.15 (d, J = 7.8 Hz, 2H), 5.26 (d, J = 4.5 Hz, 1H), 4.68 – 4.59 (m, 1H), 4.17 – 4.07 (m, 1H), 4.07 – 3.98 (m, 1H), 3.87 (s, 3H), 3.53 (t, J = 4.9 Hz, 4H), 2.96 (t, J = 5.0 Hz, 4H), 2.29 (s, 3H), 2.01 – 1.83 (m, 2H).; MS (ESI) m/z [M+H]+ calcd. for C26H30N6O3, 475.2; found, 475.3; HPLC purity: 254 nm: 99.5%. Example 122: Synthesis of Exemplary Compound 496 and Compound 497 (R)-2-(4-fluorophenyl)-1-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazin-1-yl)propan-1-one (Compound 496) and (S)-2-(4-fluorophenyl)-1-(4-(6-(1- methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)propan-1-one (Compound 497)
Figure imgf000386_0001
[0782] A solution of 2-(4-fluorophenyl)propanoic acid (200 mg, 1.18 mmol), HATU (678.3 mg, 1.78 mmol) and DIEA (307.4 mg, 2.37 mmol) in DMF (10 mL) was stirred at rt for 10 minutes, then 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)pyrazolo[1,5- a]pyridine (Compound 12) (402.9 mg, 1.42 mmol) was added and the mixture was stirred at rt for 4 h. The reaction was quenched with saturated aqueous NH4Cl solution then the mixture was extracted with EtOAc (3x20 mL). The organic extracts were combined, washed with water (3x20 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by reversed-phase flash chromatography using a C18 silica gel column eluting with 50-60% ACN/water (10mM NH4HCO3) to afford racemic 2-(4-fluorophenyl)-1-(4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)propan-1-one as a yellow solid. [0783] Racemic 2-(4-fluorophenyl)-1-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazin-1-yl)propan-1-one was separated by Prep-Chiral-HPLC to afford 165.5 mg (32%) of (R)-2-(4-fluorophenyl)-1-(4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)propan-1-one (Compound 496, first eluting peak) as a yellow solid and 178.2 mg (35%) of (S)-2-(4-fluorophenyl)-1-(4-(6-(1-methyl- 1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)propan-1-one (Compound 497, second eluting peak) as a yellow solid. [0784] (R)-2-(4-fluorophenyl)-1-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazin-1-yl)propan-1-one (Compound 496, first eluting peak). 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.80 (s, 1H), 8.21 (s, 1H), 7.96 (s, 1H), 7.67 (s, 1H), 7.61 (d, J = 9.6 Hz, 1H), 7.36 – 7.28 (m, 3H), 7.19 – 7.12 (m, 2H), 4.21 (q, J = 6.4 Hz, 1H), 3.86 (s, 3H), 3.73 – 3.62 (m, 3H), 3.51 – 3.42 (m, 1H), 3.00 – 2.78 (m, 3H), 2.58 – 2.52 (m, 1H), 1.30 (d, J = 6.8 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C24H25FN6O, 433.2; found, 433.2; HPLC purity: 254 nm: 99.6%. [0785] (S)-2-(4-fluorophenyl)-1-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin- 3-yl)piperazin-1-yl)propan-1-one (Compound 497, second eluting peak). 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.80 (s, 1H), 8.21 (s, 1H), 7.96 (s, 1H), 7.67 (s, 1H), 7.61 (d, J = 9.6 Hz, 1H), 7.36 – 7.28 (m, 3H), 7.19 – 7.12 (m, 2H), 4.21 (q, J = 6.9 Hz, 1H), 3.86 (s, 3H), 3.73 – 3.62 (m, 3H), 3.51 – 3.42 (m, 1H), 3.00 – 2.78 (m, 3H), 2.58 – 2.52 (m, 1H), 1.30 (d, J = 6.4 Hz, 3H).; MS (ESI) m/z [M+H]+ calcd. for C24H25FN6O, 433.2; found, 433.2; HPLC purity: 254 nm: 99.6%. [0786] Using the procedures described in Example 122 and reagents, starting materials, and conditions known to those skilled in the art, the following compounds representative of the present disclosure were prepared:
Figure imgf000387_0001
Figure imgf000388_0002
Example 123: Synthesis of Exemplary Compound 513 and Compound 514 (R)-1-(4-(difluoromethyl)phenyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazine-1-carboxylate (Compound 513) and (S)-1-(4- (difluoromethyl)phenyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate (Compound 514)
Figure imgf000388_0001
[0787] Racemic 1-(4-(difluoromethyl)phenyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 483) (300 mg, 0.62 mmol) was separated by Prep-Chiral-HPLC to afford 134.8 mg (45%) of (R)-1-(4- (difluoromethyl)phenyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate (Compound 513, first eluting peak) as a yellow solid and 145.7 mg ( 49%) of (S)-1-(4-(difluoromethyl)phenyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 514, second eluting peak) as a yellow solid. [0788] (R)-1-(4-(difluoromethyl)phenyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 513, first eluting peak). 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (s, 1H), 8.23 (s, 1H), 7.98 (s, 1H), 7.78 (s, 1H), 7.65 (d, J = 9.3 Hz, 1H), 7.58 (d, J = 8.0 Hz, 2H), 7.53 (d, J = 8.1 Hz, 2H), 7.34 (dd, J = 9.2, 1.5 Hz, 1H), 7.04 (t, J = 55.9 Hz, 1H), 5.79 (q, J = 6.5 Hz, 1H), 3.87 (s, 3H), 3.70 – 3.55 (m, 4H), 2.98 (s, 4H), 1.51 (d, J = 6.6 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C25H26F2N6O2, 481.2; found, 481.1, HPLC purity: 254 nm: 99.7%. [0789] (S)-1-(4-(difluoromethyl)phenyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 514, second eluting peak). 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (s, 1H), 8.23 (s, 1H), 7.98 (s, 1H), 7.78 (s, 1H), 7.65 (d, J = 9.3 Hz, 1H), 7.58 (d, J = 8.0 Hz, 2H), 7.53 (d, J = 8.1 Hz, 2H), 7.34 (dd, J = 9.2, 1.5 Hz, 1H), 7.04 (t, J = 55.9 Hz, 1H), 5.79 (q, J = 6.5 Hz, 1H), 3.87 (s, 3H), 3.70 – 3.55 (m, 4H), 2.98 (s, 4H), 1.51 (d, J = 6.6 Hz, 3H); MS (ESI) m/z [M+H]+ calcd. for C25H26F2N6O2, 481.2; found, 481.1, HPLC purity: 254 nm: 99.7%. Example 124: Synthesis of Exemplary Compound 518 1-(4-(tert-butyl)phenyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate (Compound 518)
Figure imgf000389_0001
Step 1. Preparation of 1-(4-(tert-butyl)phenyl)ethan-1-ol
Figure imgf000390_0001
[0790] To a stirred solution of 1-(4-(tert-butyl)phenyl)ethan-1-one (400 mg, 2.26 mmol) in MeOH (5 mL) cooled to 0°C was added NaBH4 (128.7 mg, 3.40 mmol) in portions then the mixture was warmed to rt and stirred for 2 h. The reaction was quenched with water (50 mL) and extracted with DCM (2x150 mL). The organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure to afford 330 mg (82%) of 1-(4-(tert-butyl)phenyl)ethan-1-ol as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 7.35 – 7.31 (m, 2H), 7.28 – 7.24 (m, 2H), 5.04 (dd, J = 4.1, 1.7 Hz, 1H), 4.71 – 4.64 (m, 1H), 1.31 (dd, J = 6.5, 1.5 Hz, 3H), 1.28 (s, 9H). Step 2. 1-(4-(tert-butyl)phenyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazine-1-carboxylate (Compound 518)
Figure imgf000390_0002
[0791] To a stirred mixture of 1-(4-(tert-butyl)phenyl)ethan-1-ol (300 mg, 1.68 mmol, as prepared in the previous step) and 4-nitrophenyl 4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (828.2 mg, 1.85 mmol, as prepared in Example 20, Step 1) in DMF (4 mL) was added Cs2CO3 (256.2 mg, 3.36 mmol) in portions then the mixture was heated to 100°C and stirred for 2 h. The reaction was cooled to rt and concentrated under reduced pressure. The residue was purified by Prep- HPLC to afford 95.7 mg (12%) of 1-(4-(tert-butyl)phenyl)ethyl 4-(6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 518) as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (s, 1H), 8.23 (s, 1H), 7.98 (s, 1H), 7.78 (s, 1H), 7.66 (d, J = 9.2 Hz, 1H), 7.43 – 7.37 (m, 2H), 7.35 (d, J =8.8 Hz, 1H), 7.33 – 7.27 (m, 2H), 5.72 (q, J = 6.5 Hz, 1H), 3.88 (s, 3H), 3.62 – 3.55 (m, 4H), 2.97 (s, 4H), 1.48 (d, J = 6.6 Hz, 3H), 1.28 (s, 9H); MS (ESI) m/z [M+H]+ calcd. for C28H34N6O2, 487.3; found, 487.3; HPLC purity: 254 nm: 99.5%. [0792] Using the procedures described in Example 124 and reagents, starting materials, and conditions known to those skilled in the art, the following compounds representative of the present disclosure were prepared:
Figure imgf000391_0003
Example 125: Synthesis of Exemplary Compound 527 3-(4-chlorophenyl)-3-fluoro-1-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazin-1-yl)propan-1-one (Compound 527)
Figure imgf000391_0001
Step 1. Preparation of 3-(4-chlorophenyl)-3-hydroxy-1-(4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)propan-1-one
Figure imgf000391_0002
[0793] A mixture of 3-(4-chlorophenyl)-3-hydroxypropanoic acid (1 g, 4.98 mmol), HATU (2.84 g, 7.48 mmol), and DIEA (1.29 g, 9.97 mmol) in DMF (15 mL) was stirred at rt for 10 minutes then 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)pyrazolo[1,5- a]pyridine (Compound 12) (1.41 g, 4.98 mmol) was added then the mixture was stirred for 4 h. The reaction was quenched with saturated aqueous NH4Cl solution and extracted with EtOAc (3x200 mL). The organic extracts were combined, washed with brine (3x20 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by silica gel column chromatography eluting with DCM/MeOH (10:1) to afford 1 g (43%) of 3-(4-chlorophenyl)-3-hydroxy-1-(4-(6-(1- methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)propan-1-one as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.72 (s, 1H), 7.64 (d, J = 9.2 Hz, 1H), 7.44 – 7.36 (m, 4H), 7.32 (dd, J = 9.2, 1.2 Hz, 1H), 5.46 (d, J = 4.4 Hz, 1H), 5.02 – 4.97 (m, 1H), 3.87 (s, 3H), 3.69 – 3.54 (m, 4H), 2.95 – 2.93 (m, 2H), 2.90 – 2.83 (m, 1H), 2.82 – 2.75 (m, 2H), 2.65– 2.60 (m, 1H); MS (ESI) m/z [M+H]+ calcd. for C24H25ClN6O2, 465.2; found, 465.0. Step 2. Preparation of 3-(4-chlorophenyl)-3-fluoro-1-(4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)propan-1-one (Compound 527)
Figure imgf000392_0001
[0794] To a solution of 3-(4-chlorophenyl)-3-hydroxy-1-(4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)propan-1-one (1 g, 2.15 mmol, as prepared in the previous step) in DCM (30 mL) cooled to -78°C was added a solution of DAST (571 mg, 2.58 mmol) in DCM (3 mL) dropwise under nitrogen then the mixture was stirred for 10 minutes. The reaction was quenched by the addition of a water/ice mixture (20 mL) at - 78°C then the mixture was warmed to rt and extracted with DCM (3x100 mL). The organic extracts were combined, washed with brine (3x10 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 40 mg (4%) of 3-(4-chlorophenyl)-3-fluoro-1-(4-(6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)propan-1-one (Compound 527) as a light yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.84 (s, 1H), 8.23 (s, 1H), 7.98 (s, 1H), 7.76 (s, 1H), 7.67 (d, J = 9.3 Hz, 1H), 7.51 (q, J = 8.5 Hz, 4H), 7.34 (d, J = 9.3 Hz, 1H), 6.19 – 5.82 (m, 1H), 3.88 (s, 3H), 3.74 – 3.54 (m, 4H), 3.26 (dd, J = 15.8, 8.4 Hz, 1H), 3.06 – 2.78 (m, 5H); MS (ESI) m/z [M+H]+ calcd. for C24H24ClFN6O, 467.2; found, 467.1; HPLC purity: 254 nm: 99.6%. Example 126: Synthesis of Exemplary Compound 528 3-(4-chlorophenyl)-2-fluoro-1-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazin-1-yl)propan-1-one (Compound 528)
Figure imgf000393_0001
Step 1. Preparation of 3-(4-chlorophenyl)-2-hydroxy-1-(4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)propan-1-one
Figure imgf000393_0002
[0795] A solution of 3-(4-chlorophenyl)-2-hydroxypropanoic acid (500 mg, 2.49 mmol), EDC (573.3 mg, 2.99 mmol), HOBt (404.1 mg, 2.99 mmol) and 6-(1-methyl-1H-pyrazol- 4-yl)-3-(piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 12) (844.4 mg, 2.99 mmol) in DCM (10 mL) was stirred for 16 h at rt. The reaction was quenched with saturated aqueous NH4Cl solution and extracted with DCM (3x20 mL). The organic extracts were combined, washed with water (3x20 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by reversed-phase flash chromatography using a C18 silica gel column eluting with 40-50% ACN/water (10mM NH4HCO3) to afford 390 mg (34%) of 3-(4-chlorophenyl)-2-hydroxy-1-(4-(6-(1-methyl- 1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)propan-1-one as a yellow solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.73 (s, 1H), 7.65 (d, J = 9.2 Hz, 1H), 7.37 – 7.26 (m, 5H), 5.18 (d, J = 7.9 Hz, 1H), 4.61 – 4.52 (m, 1H), 3.87 (s, 3H), 3.73 – 3.62 (m, 4H), 2.99 – 2.88 (m, 4H), 2.86 – 2.72 (m, 2H); MS (ESI) m/z [M+H]+ calcd. for C24H25ClN6O2, 465.2; found, 465.1; HPLC purity: 254 nm: 90%. Step 2. Preparation of 3-(4-chlorophenyl)-2-fluoro-1-(4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)propan-1-one (Compound 528)
Figure imgf000394_0001
[0796] To a solution of 3-(4-chlorophenyl)-2-hydroxy-1-(4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)propan-1-one (200 mg, 0.43 mmol, as prepared in the previous step) in DCM (5 mL) cooled to 0°C was added DAST (83.2 mg, 0.51 mmol) then the mixture was stirred for 1 h. The reaction was quenched with water then extracted with DCM (2x20 mL). The organic extracts were combined, washed with water (2x20 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 33.3 mg (17%) of 3-(4-chlorophenyl)-2-fluoro-1-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5- a]pyridin-3-yl)piperazin-1-yl)propan-1-one (Compound 528) as a light yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (s, 1H), 8.23 (s, 1H), 7.97 (s, 1H), 7.74 (s, 1H), 7.66 (d, J = 9.2 Hz, 1H), 7.39 (d, J = 8.8 Hz, 2H), 7.37 – 7.31 (m, 3H), 5.78 – 5.60 (m, 1H), 3.87 (s, 3H), 3.84 – 3.58 (m, 4H), 3.19 – 3.06 (m, 2H), 3.04 – 2.88 (m, 3H), 2.87 – 2.78 (m, 1H); MS (ESI) m/z [M+H]+ calcd. For C24H24ClFN6O, 467.2; foumd, 467.1; HPLC purity: 254 nm: 99.8%. Example 127: Synthesis of Exemplary Compound 529 1-(4-fluorophenyl)-2-hydroxyethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate (Compound 529)
Figure imgf000394_0002
[0797] To a stirred mixture of 2-((tert-butyldimethylsilyl)oxy)-1-(4-fluorophenyl)ethan-1- ol (Compound S125) (300 mg, 1.11 mmol) and 4-nitrophenyl 4-(6-(1-methyl-1H-pyrazol- 4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (496.4 mg, 1.11 mmol, as prepared in Example 20, Step 1) in DMF (5 mL) cooled to 0°C was added NaH (60%wt., 88.7 mg, 2.2 mmol) in portions then the mixture was warmed to rt and stirred for 2 h. The reaction was cooled to 0°C, quenched with water, and extracted with EtOAc (3x50 mL). The organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep- HPLC to afford 29.8 mg (6%) of 1-(4-fluorophenyl)-2-hydroxyethyl 4-(6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 529) as an off-white solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.84 (s, 1H), 8.23 (s, 1H), 7.98 (s, 1H), 7.78 (s, 1H), 7.66 (d, J = 9.3 Hz, 1H), 7.43 – 7.37 (m, 2H), 7.34 (dd, J = 9.3, 1.5 Hz, 1H), 7.23 – 7.15 (m, 2H), 5.61 (dd, J = 7.0, 4.4 Hz, 1H), 5.05 (t, J = 5.9 Hz, 1H), 3.88 (s, 3H), 3.77 – 3.41 (m, 6H), 2.98 (s, 4H); MS (ESI) m/z [M+H]+ calcd. for C24H25FN6O3, 465.2; found, 465.2; HPLC purity: 254 nm: 99.8%. Example 128: Synthesis of Exemplary Compound 530 2-(4-fluorophenyl)-2-hydroxyethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate (Compound 530)
Figure imgf000395_0001
[0798] To a stirred mixture of 2-((tert-butyldimethylsilyl)oxy)-2-(4-fluorophenyl)ethan-1- ol (Compound S126) (300 mg, 1.11 mmol) and 4-nitrophenyl 4-(6-(1-methyl-1H-pyrazol- 4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (990 mg, 2.22 mmol, as prepared in Example 20, Step 1) in DMF (5 mL) cooled to 0°C was added NaH (60%wt., 88.7 mg, 2.2 mmol) in portions then the mixture was warmed to rt and stirred for 2 h. The reaction was cooled to 0°C, quenched with water, and extracted with EtOAc (3x50 mL). The organic extracts were combined, dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep- HPLC to afford 91.9 mg (18%) of 2-(4-fluorophenyl)-2-hydroxyethyl 4-(6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 530) as an off-white solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.83 (s, 1H), 8.24 (s, 1H), 7.98 (s, 1H), 7.77 (s, 1H), 7.66 (d, J = 9.2 Hz, 1H), 7.43 (dd, J = 8.6, 5.7 Hz, 2H), 7.34 (dd, J = 9.2, 1.6 Hz, 1H), 7.18 (t, J = 8.9 Hz, 2H), 5.66 (d, J = 4.6 Hz, 1H), 4.82 (q, J = 5.4 Hz, 1H), 4.11 – 4.03 (m, 2H), 3.88 (s, 3H), 3.54 (s, 4H), 2.94 (s, 4H); MS (ESI) m/z [M+H]+ calcd. for C24H25FN6O3, 465.2; found, 465.3; HPLC purity: 254 nm: 99.7%. Example 129: Synthesis of Exemplary Compound 533 1-(4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)-2-(5- methylpyridin-2-yl)ethan-1-one (Compound 533)
Figure imgf000396_0001
[0799] To a solution of (5-methylpyridin-2-yl)acetic acid (200 mg, 1.32 mmol), HATU (628.84 mg, 1.65 mmol), and DIEA (342 mg, 2.64 mmol) in DMF (5 mL) was added 6-(1- methyl-1H-pyrazol-4-yl)-3-(piperazin-1-yl)pyrazolo[1,5-a]pyridine (Compound 12) (448.2 mg, 1.58 mmol) then the mixture was heated to 60°C and stirred for 3 h. The reaction was cooled to 0°C, quenched with saturated aqueous NH4Cl solution, and extracted with EtOAc (3x20 mL). The organic extracts were combined, washed with water (3x20 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by Prep-HPLC to afford 138.5 mg (25%) of 1-(4-(6- (1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazin-1-yl)-2-(5- methylpyridin-2-yl)ethan-1-one (Compound 533) as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.34 – 8.30 (m, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.74 (s, 1H), 7.65 (d, J = 9.2 Hz, 1H), 7.56 (dd, J = 8.0, 2.1 Hz, 1H), 7.32 (dd, J = 9.2, 1.4 Hz, 1H), 7.20 (d, J = 7.9 Hz, 1H), 3.88 (s, 2H), 3.86 (s, 3H), 3.72 – 3.69 (m, 2H), 3.68 – 3.64 (m, 2H), 2.95 – 2.91 (m, 4H), 2.27 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C23H25N7O, 416.2; found, 416.3; HPLC purity: 254 nm: 99.4%.
Example 130: Synthesis of Exemplary Compound 485 2-hydroxy-2-(p-tolyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3- yl)piperazine-1-carboxylate (Compound 485)
Figure imgf000397_0001
Step 1. Preparation of 2-((tert-butyldimethylsilyl)oxy)-2-(p-tolyl)ethyl 4-(6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate
Figure imgf000397_0002
[0800] To a stirred solution of 2-((tert-butyldimethylsilyl)oxy)-2-(p-tolyl)ethan-1-ol (Compound S111) (200 mg, 0.75 mmol) and 6-(1-methyl-1H-pyrazol-4-yl)-3-(piperazin- 1-yl)pyrazolo[1,5-a]pyridine (Compound 12) (159.0 mg, 0.56 mmol) in pyridine (5 mL) cooled to 0°C was added BTC (222.5 mg, 0.75 mmol) in portions then the mixture was warmed to rt and stirred for 1 h. The reaction was cooled to 0°C, quenched with water, and extracted with EtOAc (3x20 mL). The organic extracts were combined, washed with water (3x20 mL), dried over anhydrous Na2SO4, and filtered. The filtrate was concentrated under reduced pressure then the residue was purified by reversed-phase flash chromatography using a C18 silica gel column eluting with 45-55% ACN/water (10mM NH4HCO3) to afford 210 mg (49%) of 2-((tert-butyldimethylsilyl)oxy)-2-(p-tolyl)ethyl 4-(6-(1-methyl- 1H-pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate as a yellow solid. 1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.23 (s, 1H), 7.97 (s, 1H), 7.76 (s, 1H), 7.67 (d, J = 9.2 Hz, 1H), 7.33 (dd, J = 9.3, 1.4 Hz, 1H), 7.26 (d, J = 8.0 Hz, 2H), 7.17 (d, J = 8.0 Hz, 2H), 4.96 – 4.91 (m, 1H), 4.06 – 3.99 (m, 2H), 3.87 (s, 3H), 3.56 (s, 4H), 2.95 (s, 4H), 2.29 (s, 3H), 0.85 (s, 9H), 0.04 (s, 3H), -0.09 (s, 3H); MS (ESI) m/z [M+H]+ calcd. For C31H42N6O3Si, 575.3; found, 575.3. Step 2. Preparation of 2-hydroxy-2-(p-tolyl)ethyl 4-(6-(1-methyl-1H-pyrazol-4- yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 485)
Figure imgf000398_0001
[0801] A mixture of 2-((tert-butyldimethylsilyl)oxy)-2-(p-tolyl)ethyl 4-(6-(1-methyl-1H- pyrazol-4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (210 mg, 0.36 mmol, as prepared in the previous step) in 4M HCl in EtOAc (5 mL) was stirred at rt for 1 h. The reaction was quenched with saturated aqueous Na2CO3 solution adjusting the pH to 8 then the mixture was concentrated under reduced pressure. The residue was purified by Prep- HPLC to afford 52.6 mg (31%) of 2-hydroxy-2-(p-tolyl)ethyl 4-(6-(1-methyl-1H-pyrazol- 4-yl)pyrazolo[1,5-a]pyridin-3-yl)piperazine-1-carboxylate (Compound 485) as a white solid.1H NMR (400 MHz, DMSO-d6) δ (ppm) 8.82 (s, 1H), 8.22 (s, 1H), 7.97 (s, 1H), 7.75 (s, 1H), 7.66 (d, J = 9.2 Hz, 1H), 7.34 (dd, J = 9.2, 1.6 Hz, 1H), 7.27 (d, J = 8.0 Hz, 2H), 7.16 (d, J = 8.0 Hz, 2H), 5.54 (d, J = 4.4 Hz, 1H), 4.76 (q, J = 5.6 Hz, 1H), 4.05 (d, J = 5.6 Hz, 2H), 3.88 (s, 3H), 3.55 (s, 4H), 2.94 (s, 4H), 2.29 (s, 3H); MS (ESI) m/z [M+H]+ calcd. for C25H28N6O3, 461.2; found, 461.2; HPLC purity: 254 nm: 99.2%. Example 131: Functional Analysis of the Exemplary PDGFRα Inhibitors [0802] To assess the inhibitory capability of the PDGFRα inhibitors synthesized in Examples 1-130, an enzymatic PDGFRα kinase assay was used as described below. Methods: [0803] The PDGFRα enzyme assays were conducted per the manufacturer's directions (Promega PDGFRα Kinase Enzyme System and ADP Glo Assay Cat# V8011). Briefly, exemplary PDGFRα inhibitor compounds described herein were assayed in an 11-point dose response curve with a maximum concentration of 10,000 nM and 3-fold dilutions to a minimum concentration of 0.169 nM.20 ng of purified PDGFRα protein was added to the test article. Subsequently, 150 µM of ATP and 1 ug of substrate, Poly (Glu4Tyr1) was added to each reaction, and the PDGFRα kinase mediated conversion of ATP (adenosine triphosphate) to ADP (adenosine diphosphate) was allowed to continue for two hours. ADP-Glo was added to halt the kinase reaction and deplete the remaining ATP. Kinase detection reagent, containing luciferase and luciferin, was used to convert the ADP signal into luminescence. Luminescence was measured using a Molecular Devices Spectramax ID5 and compared to a standard curve to determine kinase activity. IC50s were calculated using Prism 9, Graphpad software. Results: [0804] As shown in Table 4 (below), the compounds exhibited varying potency with respect to inhibition of PDGFRα kinase activity. Of those tested, several compounds inhibited PDGFRα kinase activity with an IC50 of less than 1,000 nM. The results confirm the inhibitory activity of the novel PDGFRα inhibitors described herein. Table 4. IC50 Values for PDGFRα Kinase Inhibition of Exemplary Compounds
Figure imgf000399_0001
Figure imgf000400_0001
Figure imgf000401_0001
Figure imgf000402_0001
Figure imgf000403_0001
Figure imgf000404_0001
BLQ = below limit of quantitation, <0.17 nM Example 132: Analysis of the Effect of PDGFRα Inhibition on GPR17 Expression in vitro [0805] To assess the effects of the PDGFRα inhibitors on OPC differentiation, primary OPCs were cultured and exposed to compounds disclosed herein. The number of newly generated oligodendrocytes was assessed by monitoring GPR17 expression. GPR17 is preferentially expressed in actively differentiating oligodendrocytes, allowing them to be distinguished from precursors and more differentiated oligodendrocytes. Methods: OPC isolation and seeding: [0806] Cortices from postnatal day (P) 1-3 rats were dissected and collected into ice cold Hank's Balanced Salt Solution (HBSS). A homogenous cell suspension of the cortices was prepared using the Neural Tissue Dissociation Kit (P) (Miltenyi #130-092-628) as per manufacturer's protocol. Oligodendrocyte progenitor cells (OPCs) were isolated from the mixed cortical cell suspension by positive selection using Anti-A2B5 MicroBeads (Miltenyi #130-093-388), as per manufacturer’s protocol. Briefly, the cortical cell suspension was centrifuged and washed with buffer (0.5% BSA in PBS) and labeled with Anti-A2B5 MicroBeads (10 µL beads per 1 x 107 cells) for 15 minutes at 4°C. Following the incubation, cells were washed once with BSA buffer to remove unlabeled beads. A2B5 positive cells were eluted on the column using the AutoMACS® (Miltenyi, as per manufacturer's protocol using "Possel-s" program). Eluted cells were further washed twice with OPC growth media. Cell counts were performed using Countess III cell counter (ThermoFisher; AMQAX2000) and cells were plated. [0807] Following cell count, OPCs were seeded at a density of approximately 10,000 cells per well in poly-D-lysine coated 96 well plates (PE Cell Carrier #6005550) or approximately 2500 cells per well in poly-D-lysine coated 384 well plate (PE Cell Carrier # 6057500) in 90 µL of OPC growth medium (DMEM/F12 + B27 without vitamin A + pen/strep) supplemented with 10 ng/mL of PDGF-AA (Peprotech 100-13A) and 10 ng/mL of FGF (fibroblast growth factor; R&D Systems #233 FB 010). The plates were allowed to sit at room temperature for 15 minutes to ensure proper dispersion of cells in a well. The plates were then transferred to the incubator (37°C; 5% CO2). Compound Treatment [0808] Following 3-4 hours of incubation (to allow cells to adhere), cells (seeded in 90 µl OPC growth media) were treated with 10 µl of 10X concentration of vehicle (DMSO; 0.03%) control (Santa Cruz Biotechnology; sc-358801), or PDGFRα inhibitors disclosed herein (3 µM), resulting in a final 1X concentration of 0.003% for the vehicle or 0.3 µM for the PDGFRα inhibitors. Cells were then incubated for 72 hours at 37°C, 5% CO2 before immunostaining for GPR17 marker expression. Immunocytochemistry: [0809] Following 72 hours in culture, cells were fixed with 4% paraformaldehyde for 15 minutes and then washed three times in Dulbecco's phosphate buffered saline (DPBS). Prior to immunostaining, cells were treated with a blocking solution (10% normal goat serum containing 1% BSA and 0.1% Triton® X-100 in DPBS) for 15 minutes at room temperature. Primary antibody (anti-GPR17 at 1:2000; Origene TA811615) was applied to cells overnight at 4°C in incubation solution containing 1% normal goat serum, 1% BSA, and 0.1% Triton X-100 in DPBS. Following overnight primary incubation, cells were washed three times in DPBS, and then, secondary antibody (Goat anti-mouse Alexa Fluor 594 at 1:1000, Jackson Immunoresearch 112-545-003) was applied to the cells. The secondary antibody was applied in incubation solution for 2 hours at room temperature, protected from light. Following incubation with the secondary antibodies, cells were washed three times with DPBS and nuclei stained with Hoechst (BioRad #1351304) before imaging. Image analysis and quantification: [0810] Cells were imaged using ImageXpress® confocal system (Molecular Devices; 5150105) and analyzed using a custom protocol developed using MetaXpress Custom Module software. The number of differentiated GPR17+ cells was normalized to the total nuclei count and percent differentiated GPR17+ cells were reported. Statistical analysis was done using one-way ANOVA with multiple comparisons and post-hoc Tukey test in GraphPad Prism (GraphPad Software). Results: [0811] Treatment of the OPCs with PDGFRα inhibitors led to an increase in differentiated GPR17+ cells, a marker of oligodendrocyte differentiation. The increase is represented by fold change over vehicle for the PDGFRα inhibitors (Table 5). These results demonstrate that PDGFRα inhibition induces OPC differentiation, indicating that compounds that inhibit PDGFRα activity could be useful for inducing remyelination. Table 5. In vitro GPR17 Expression Induced by Exemplary PDGFRα Inhibitors
Figure imgf000406_0001
Figure imgf000407_0001
Figure imgf000408_0001
Figure imgf000409_0001
Example 133: Analysis of the Effect of PDGFRα Inhibition on MBP Expression in vitro [0812] While expression of GPR17 indicates differentiated or pre-myelinating oligodendrocytes, terminal differentiation of the OPCs to oligodendrocytes and formation of myelin is characterized by expression of myelin basic protein (MBP), which is an important element of myelin. [0813] To further confirm the results shown in Example 132 that PDGFRα inhibitors can lead to terminal oligodendrocyte differentiation, select PDGFRα inhibitors were tested in the in vitro OPC differentiation assay by measuring the number of MBP+ oligodendrocytes. Methods: OPC isolation and seeding: [0814] Cortices from postnatal day (P) 1-3 rats were dissected and collected into ice cold Hank's Balanced Salt Solution (HBSS). A homogenous cell suspension of the cortices was prepared using the Neural Tissue Dissociation Kit (P) (Miltenyi #130-092-628) as per manufacturer's protocol. Oligodendrocyte progenitor cells (OPCs) were isolated from the mixed cortical cell suspension by positive selection using Anti-A2B5 MicroBeads (Miltenyi #130-093-388), as per manufacturer's protocol. Briefly, the cortical cell suspension was centrifuged and washed with buffer (0.5% BSA in PBS) and labeled with Anti-A2B5 MicroBeads (10 µL beads per 1 x 107 cells) for 15 minutes at 4°C. Following the incubation, cells were washed once with BSA buffer to remove unlabeled beads. A2B5 positive cells were eluted on the column using the AutoMACS® (Miltenyi, as per manufacturer's protocol using "possel-s" program). Eluted cells were further washed twice with OPC growth media. Cell count was performed using Countess III cell counter (ThermoFisher; AMQAX2000) and cells were plated. [0815] A2B5+ OPCs were plated in poly-d-lysine (PDL) coated T75 flask at a density of ~3 million cells per flask. Cells were allowed to expand for 3 days at 37°C, 5% CO2 in OPC growth media containing 10 ng/mL of PDGF (Peprotech 100-13A) and 10 ng/mL FGF (R&D Systems #233-FB-010). Following three days of expansion (~60-70% confluency), cells were lifted using TrypLE (Thermo 12604013) for 3-5 minutes at 37°C, 5% CO2. TrypLE was normalized by adding 10mL growth media and cells were centrifuged at 1400 rpm for 10 minutes. Cell count was performed, and cells were plated into PDL coated 96 well plates (PE Cell Carrier #6005550) at approximately 5000 cells per well in 90 µL of OPC growth medium (DMEM/F12 + B27 without vitamin A + pen/strep) supplemented with 10 ng/mL of PDGF-AA (Peprotech 100-13A) and 10 ng/mL of FGF (fibroblast growth factor; R&D Systems #233 FB 010). The plates were allowed to sit at room temperature for 15 minutes to ensure proper dispersion of cells in a well. The plates were then transferred to the incubator (37°C; 5% CO2). Compound Treatment [0816] Following 3-4 hours of incubation (to allow cells to adhere), cells (seeded in 90µl OPC growth media) were treated with 10µl of 10X concentration of vehicle (DMSO; 0.03%) control (Santa Cruz Biotechnology; sc-358801), or PDGFRα inhibitors disclosed herein (3µM), resulting in a final 1X concentration of 0.3% for the vehicle or 0.3 µM for the PDGFRα inhibitors. Cells were then incubated for 72 hours at 37°C, 5% CO2 before immunostaining for MBP marker expression. Immunocytochemistry: [0817] Following 72 hours of treatment, cells were fixed with 4% paraformaldehyde for 15 minutes then washed three times in DPBS. Prior to staining, cells were blocked for 15 minutes in 10% normal goat serum containing 1% BSA and 0.1% Triton X-100 in DPBS. Primary antibody (MBP @ 1:2000; Abcam AB7349) was applied to cells overnight at 4°C in incubation solution containing 1% normal goat serum, 1% BSA, and 0.1% Triton X-100 in DPBS. After primary incubation, cells were washed three times in DPBS and secondary antibodies (Goat anti-Rat Alexa Fluor 488, Jackson Immunoresearch 112-545- 003, 1:1000) were applied in incubation solution for two hours at room temperature, protected from light. Following secondary antibodies, cells were washed three times with DPBS and nuclei stained with Hoechst (BioRad #1351304). Image analysis and quantification: [0818] Cells were imaged using ImageXpress® confocal system (Molecular Devices; 5150105) and analyzed using a custom protocol from MetaXpress Custom Module software. The number of differentiated MBP+ cells was normalized to the total nuclei count and percent differentiated MBP+ cells were reported. Statistical analysis was done using one-way ANOVA with multiple comparisons and post-hoc Tukey test in GraphPad Prism (GraphPad Software). Results: [0819] Treatment of OPCs with PDGFRα inhibitors led to increases in differentiated MBP+ cells. The increase is represented by fold change over vehicle for the PDGFRα inhibitors (Table 6). In agreement with results from Example 132, these data demonstrate that PDGFRα inhibition can drive OPC differentiation and expression of myelin specific proteins. These findings further support the therapeutic potential of PDGFRα inhibitors in remyelination. [0820] In parallel with the PDGFRα kinase inhibition data, the novel chemical compounds described herein were also able to induce OPC differentiation and myelin protein formation in vitro, as evidenced by the increased GPR17 and MBP expression. Collectively, the above results demonstrate that the novel PDGFRα inhibitors described herein could be useful in inducing remyelination as a therapeutic for the treatment of demyelinating diseases. Table 6. In vitro MBP Expression Induced by Exemplary PDGFRα Inhibitors
Figure imgf000411_0001
Figure imgf000412_0001
Figure imgf000413_0001
Example 134: PDGFRα inhibitors induce OPC differentiation in vivo [0821] The ability of compounds disclosed herein to drive OPC differentiation and myelination in vivo was tested. The ability of these compounds to drive OPC differentiation in vivo was tested using GPR17 as an indicator of newly differentiated oligodendrocytes. Materials Mice: [0822] Age matched (> 6 months old) adult female C57BL/6 mice (Strain Code 000664) were purchased from The Jackson Laboratory. Methods: Compound treatment: [0823] All experiments were conducted in accordance with animal welfare and IACUC guidelines. Compounds were delivered PO or IP either QD or BID. Mice were dosed once, transiently, or continuously through the treatment period, which ranged from 1-3 days. Table 7 (below) describes the different treatment groups. Table 7. Treatment Groups with Exemplary PDGFRα Inhibitors
Figure imgf000414_0001
Histology: [0824] Mice were euthanized by isoflurane exposure and transcardially perfused with 14 mL of PBS (Sigma Cat # P4417-50TAB) followed by 22 mL of 3.2% paraformaldehyde in PBS. The brains were removed and postfixed for a minimum of 24 hours in 20 mL of 3.2% paraformaldehyde. 3.2% paraformaldehyde was prepared from 32% paraformaldehyde (Electron microscopy Sciences Cat # 15714-S) diluted in phosphate buffered saline. [0825] 50-micron floating sections were cut using a Leica VT1200S vibratome. Every 24th section, extending through the antero-posterior extent of the brain, was stained. The selected brain sections were washed three times in PBS for 3 minutes each. The floating sections were incubated in the primary antibodies (see Table 10, below) diluted in blocking solution overnight at room temperature on a shaker. Blocking solution included: 0.3% Triton X-100 (Sigma, Cat # 234729), 0.02% sodium azide (Sigma, Cat # S2002), 8% fetal bovine serum (Sigma, Cat #F2442), 1X PBS. The next day, the sections were again washed three times in PBS for 3 minutes each and then incubated in the secondary antibodies diluted in blocking solution for 2 hours at room temperature on a shaker. After incubation, the sections were washed three times in PBS for 3 minutes each and mounted on a slide to be imaged using the Olympus Slide Scanner VS200. Table 10. Primary Antibodies
Figure imgf000415_0001
Image Analysis and quantification: [0826] The images were analyzed using a custom protocol developed using Visiopharm software to count GPR17+ cells. Cells were considered GPR17+ if they contained an Olig2 positive nucleus, and intense GPR17 staining that filled their processes. Statistical analysis was done using one-way ANOVA with multiple comparison in GraphPad Prism software. Results [0827] Treatment of the animals with PDGFRα inhibitors disclosed herein led to an increase in the number of cells with process filling GPR17+ cells, a marker for newly differentiated oligodendrocytes, throughout the entire brain. The increase is represented by fold change over vehicle treated animals for the PDGFRα inhibitors (Table 11). In agreement with the results from Example 132, many of the PDGFRα inhibitors disclosed herein that are capable of inducing OPC differentiation in vitro can also induce OPCs to differentiate in vivo. Table 11. In vivo GPR17+ cells Induced by Exemplary PDGFRα Inhibitors
Figure imgf000416_0001
Figure imgf000417_0001

Claims

WHAT IS CLAIMED IS: 1. A compound of Formula I:
Figure imgf000418_0001
, or a pharmaceutically acceptable salt or solvate thereof, wherein: indicates a single bond or a double bond such that all valences are satisfied; X1, X2, X3, and X4 are selected from N and CRa, with the proviso that not more than two of X1, X2, X3, and X4 are N; one of Y1 and Y2 is N and the other of Y1 and Y2 is C; each Ra is independently selected from H, halo, C1-C4alkyl, and C1-C4alkoxy; R1 is selected from C1-C4alkyl, C3-C8cycloalkyl, 3-8 membered heterocyclyl, heteroaryl, aryl, and C1-C8alkoxy, all of which can be optionally substituted with one, two, three, four, five, or six substituents selected from halo, hydroxy, oxo, C1-C4alkyl, aminoC1-C4alkyl, hydroxyC1- C4alkyl, C1-C4alkoxy, C1-C4alkoxyC1-C4alkyl, 3-8 membered heterocyclyl, and 3-8 membered heterocyclylC1-C4alkyl, with the proviso that the number of substituents does not exceed the number of substitutable positions; R2 is selected from cycloalkyl, cycloalkenyl, alkyl, oxoalkylamino, aminoalkylamino, amino, heterocyclyl, heteroaryl, aminoheterocyclyl, heterocyclylamino, and aminoalkylamino, all of which can be optionally substituted with one or more substituents selected from D, halo, hydroxy, oxo, and C1-C4alkyl; R2 is substituted with one, two, or three R3; R3 is selected from aryl, heteroaryl, -C(O)R31, -C(O)OR31, -C(O)NR31R32, -S(O)2NR31R32, -S(O)(NR33)R31, -S(O)2R31, -S(O)(NR33)NR31R32, -C(S)NR31R32, C3-C8cycloalkyl, 3-8 membered heterocyclyl, and C1-C4alkyl, all of which can be optionally substituted with one, two, three, four, or five R30; each R30 is independently selected from D, halo, aryl, -OR300, -NR300R303, -S(O)rR300, -C(O)R300, -C(=CR34R35)R300, and
Figure imgf000419_0001
; r is selected from 0, 1, and 2; each R300 is independently selected from C1-C6alkyl, C3-C7cycloalkyl, aryl, heteroaryl, 3-8 membered heterocyclyl, and 3-8 membered heterocyclylaryl, all of which can be optionally substituted with one, two, three, four, or five substituents selected from D, halo, hydroxy, amino, alkylamino, cyano, C1-C4alkyl, C1-C4haloalkyl, and C1-C4alkoxy; each R301 is independently selected from H, halo, and C1-C4alkyl; each R302 is independently selected from H, F, hydroxyl, amino, alkylamino, oxo, and C1-C4alkyoxy; each R303 is independently selected from H and C1-C4alkyl; n, o, and p are each independently selected from 0, 1, 2, 3, and 4; each R31 is independently selected from C1-C8alkyl, arylC1-C4alkyl, heteroarylC1- C4alkyl, heterocyclyl, heterocyclylC1-C4alkyl, cycloalkyl, and cycloalkylC1-C4alkyl, all of which can be optionally substituted with one, two, three, four, or five substituents selected from D, halo, cyano, hydroxy, amino, -OCF3, C1-C4alkyl, C1-C4haloalkyl, C1-C4alkoxy, hydroxyC1-C4alkyl, -S(O)2NR304R305, -C(O)OR304R305, -C(O)NR304R305, and - NR304C(O)R305; each R304 and R305 is independently selected from H and C1-C4alkyl; each R32 is independently selected from H and C1-C4alkyl; or R31 and R32 together with the atom to which they are connected to form a 5-8 membered heterocycyl, optionally substituted with one, two, three, four, or five substituents selected from D, halo, cyano, C1-C4alkyl, C1-C4haloalkyl, and -C(O)NR34R35; and each R34 and R35 is independently selected from H, C1-C4alkyl, and C1-C4haloalkyl; and each R33 is independently selected from H, C1-C4alkyl, C1-C4haloalkyl, and – C(O)R34; or R31 and R33 together with the atoms to which they are connected form a 4-8 membered heterocycyl.
2. The compound of claim 1, or a pharmaceutically acceptable salt or solvate thereof, wherein Y1 is N and Y2 is C.
3. The compound of claim 1, or a pharmaceutically acceptable salt or solvate thereof, wherein Y1 is C and Y2 is N.
4. The compound of any one of claims 1 to 3, or a pharmaceutically acceptable salt or solvate thereof, wherein X1 is N, X2 is CRa, X3 is CRa, and X4 is CRa.
5. The compound of any one of claims 1 to 3, or a pharmaceutically acceptable salt or solvate thereof, wherein X1 is CRa, X2 is N, X3 is CRa, and X4 is CRa.
6. The compound of any one of claims 1 to 3, or a pharmaceutically acceptable salt or solvate thereof, wherein X1 is CRa, X2 is CRa, X3 is N, and X4 is CRa.
7. The compound of any one of claims 1 to 3, or a pharmaceutically acceptable salt or solvate thereof, wherein X1 is CRa, X2 is CRa, X3 is CRa, and X4 is N.
8. The compound of any one of claims 1 to 3, or a pharmaceutically acceptable salt or solvate thereof, wherein X1 is CRa, X2 is CRa, X3 is CRa, and X4 is CRa.
9. The compound of any one of claims 1 to 8, or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is 5- or 6-membered heteroaryl.
10. The compound of any one of claims 1 to 9, or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is an optionally substituted pyrazolyl.
11. The compound of any one of claims 1 to 10, or a pharmaceutically acceptable salt or solvate thereof, wherein: R1 is selected from:
Figure imgf000421_0002
, , d R10 is selected from H, C1-C4alkyl, C1-C4alkoxy, aminoC1-C4alkyl, hydroxyC1- C4alkyl, and C1-C4alkylsulfonyl, wherein the C1-C4alkyl, C1-C4alkoxy, aminoC1-C4alkyl, hydroxyC1-C4alkyl, and C1-C4alkylsulfonyl can be optionally substituted by one or more substituents selected from hydroxyl, C1-C4alkoxy, NR10aR10b, halo, and deuterium, wherein R10a and R10b are selected from hydrogen and C1-C4alkyl, or wherein R10a and R10b taken together with the nitrogen atom to which they are attached form a 4- to 8- membered ring.
12. The compound of claim 11, or a pharmaceutically acceptable salt or solvate thereof, wherein R1 is
Figure imgf000421_0001
.
13. The compound of claim 11 or 12, or a pharmaceutically acceptable salt or solvate thereof, wherein R10 is CH3.
14. The compound of any one of claims 1 to 13, or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is heterocyclyl optionally substituted with C1-C4alkyl or oxo.
15. The compound of any one of claims 1 to 14, or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is substituted with one R3.
16. The compound of any one of claims 1 to 15, or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is selected from:
,
Figure imgf000422_0001
,
,
Figure imgf000423_0001
, , , ,
Figure imgf000424_0001
indicates a single bond or a double bond such that all valences are satisfied; m is selected from 0, 1, 2, 3, 4, 5, and 6; and Z1, Z2, and Z3 are selected from N and CRa.
17. The compound of claim 16, or a pharmaceutically acceptable salt or solvate thereof, wherein R2 is selected from:
Figure imgf000425_0001
, , , , .
18. The compound of any one of claims 1 to 17, or a pharmaceutically acceptable salt or solvate thereof, having Formula Ia:
Figure imgf000426_0001
wherein a and b are each independently selected from 1, 2, and 3; and Q is selected from -CH- and -N-, with the proviso that if Q is -N-, a and b are not 1.
19. The compound of any one of claims 1 to 18, or a pharmaceutically acceptable salt or solvate thereof, having Formula II:
Figure imgf000426_0002
.
20. The compound of any one of claims 1 to 19, or a pharmaceutically acceptable salt or solvate thereof, having Formula IIa:
Figure imgf000427_0001
.
21. The compound of any one of claims 1 to 20, or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is heteroaryl optionally substituted with one, two, three, four, or five R30.
22. The compound of any one of claims 1 to 21, or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is selected from: ,
Figure imgf000427_0002
, , , , , , ,
Figure imgf000428_0001
, wherein A1 is selected from O, S, and NR37, R36 is selected from hydrogen, optionally substituted C1-C6alkyl, and optionally substituted C1-C6alkylaryl, and R37 is selected from hydrogen and C1-C6alkyl.
23. The compound of any one of claims 1 to 22, or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is selected from: , ,
Figure imgf000429_0001
,
Figure imgf000430_0001
, , .
24. The compound of any of claims 1 to 23, or a pharmaceutically acceptable salt or solvate thereof, wherein R30 is:
Figure imgf000430_0002
.
25. The compound of claim 24, or a pharmaceutically acceptable salt or solvate thereof, wherein R300 is selected from:
Figure imgf000430_0003
, , , , , , ,
Figure imgf000431_0001
, , , , , .
26. The compound of claim 25, or a pharmaceutically acceptable salt or solvate thereof, wherein R300 is:
Figure imgf000431_0002
.
27. The compound of any one of claims 1 to 26, or a pharmaceutically acceptable salt or solvate thereof, wherein R301 is H and R302 is H or CH3.
28. The compound of any of claims 1 to 22, or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is selected from:
Figure imgf000432_0002
29. The compound of claim 28, or a pharmaceutically acceptable salt or solvate thereof, wherein R3 is selected from:
Figure imgf000432_0001
, , .
30. The compound of claim 29, or a pharmaceutically acceptable salt or solvate thereof, wherein R32 is H.
31. The compound of any one of claims 28 to 30, or a pharmaceutically acceptable salt or solvate thereof, wherein R31 is selected from:
Figure imgf000433_0001
, , ,
Figure imgf000434_0001
Figure imgf000435_0001
, , , ,
Figure imgf000436_0001
, wherein R31a is selected from H, D, alkylamino, optionally substituted C1-C4alkyl, C1-C4alkoxy, amino, and C1-C4haloalkyl, each R31b is independently selected from H, D, halo, hydroxy, amino, cyano, alkylamino, optionally substituted C1-C4alkyl, C1-C4haloalkyl, optionally substituted C1- C4alkoxy, and C3-C6cycloalkyl, , and q is 1, 2, or 3.
32. The compound of claim 1, or a pharmaceutically acceptable salt or solvate thereof, selected from any one of the compounds of Table 1.
33. The compound of any one of claims 1 to 32, or a pharmaceutically acceptable salt or solvate thereof, which can exhibit one or more of the following properties: (i) promote the differentiation of an OPC into an oligodendrocyte, (ii) promote the expression of a protein associated with oligodendrocyte differentiation and/or myelination (e.g., G-protein coupled receptor 17 (GPR17), myelin basic protein (MBP), ASPA, GST-pi, CC1, myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, proteolipid protein 1 (PLP1), or a combination thereof), (iii) promote the myelination of an axon, (iv) promote the remyelination of a demyelinated axon, (v) inhibit PDGFRα kinase activity, (vi) achieve a brain to plasma ratio of greater than 0.1 when systemically administered to a subject, and (vii) any combination thereof.
34. The compound of claim 33, or a pharmaceutically acceptable salt or solvate thereof, which can inhibit PDGFRα kinase activity.
35. The compound of claim 34, or a pharmaceutically acceptable salt or solvate thereof, which can inhibit PDGFRα kinase activity with an IC50 of less than 500 nM, less than 400 nM, less than 300 nM, less than 200 nM, less than 100 nM, less than 75 nM, less than less than 50 nM, less than 40 nM, less than 30 nM, less than 20 nM, less than 10 nM, less than 5 nM, less than 2.5 nM, less than 1 nM, less than 0.5 nM, or less than 0.2 nM.
36. The compound of claim 35, or a pharmaceutically acceptable salt or solvate thereof, wherein the IC50 of the PDGFRα inhibitor is determined using an enzymatic PDGFRα kinase assay (e.g., Promega kinase assay described in Example 131).
37. The compound of claim 36, or a pharmaceutically acceptable salt or solvate thereof, wherein the enzymatic PDGFRα kinase assay comprises 20 ng of purified PDGFRα protein, 150 µM of ATP and 1 µg of substrate, Poly (Glu4Tyr1) in a volume of 15 µl.
38. A pharmaceutical composition comprising the compound of any one of claims 1 to 37, or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable excipient.
39. A kit comprising the compound of any one of claims 1 to 37, or a pharmaceutically acceptable salt or solvate thereof, or the composition of claim 38, and instructions for use.
40. A compound of any one of claims 1 to 37, or a pharmaceutically acceptable salt or solvate thereof, or the pharmaceutical composition of claim 38, for use in therapy.
41. A method of treating a demyelinating disease in a subject in need thereof comprising administering to the subject a therapeutically effective amount of the compound of any one of claims 1 to 37, or a pharmaceutically acceptable salt or solvate thereof, or the pharmaceutical composition of claim 38.
42. A method of improving a subject's performance in a test for assessing one or more symptoms associated with a demyelinating disease, comprising administering to the subject a therapeutically effective amount of the compound of any one of claims 1 to 37, or a pharmaceutically acceptable salt or solvate thereof, or the pharmaceutical composition of claim 38, wherein after the administration, the subject's performance in the test is improved as compared to a reference subject (e.g., the subject prior to the administration).
43. The method of claim 42, wherein the test is one or more of a visual evoked potential (VEP) test, a multifocal visual evoked potential (mfVEP) test, a low contrast visual acuity (LCVA) test, a magnetic resonance imaging (MRI) (e.g., magnetization transfer ratio (MTR), myelin water fraction (MWF), quantitative susceptibility mapping (QSM), and T2 imaging), an electromyography (EMG), a nerve conduction velocity (NCV) test, an Extended Disability Status Scale (EDSS), a timed walk test (e.g., timed 25-foot walk), a Nine-Hole Peg Test (9HPT), an ocular coherence tomograph (OCT), a quality of life measure test (e.g., Multiple Sclerosis Quality of Life-54 and Vision-Related Quality of Life), cognitive assessment (e.g., Symbol Digit Modalities Test or Montreal Cognitive Assessment), or combinations thereof.
44. The method of any one of claims 41 to 43, wherein the demyelinating disease comprises one or more of an acute disseminated encephalomyelitis (ADEM), acute hemorrhagic leukoencephalitis, acute transverse myelitis, adrenoleukodystrophy, adrenomyeloneuropathy, Alexander disease, Alzheimer's disease, aminoacidurias, amyotrophic lateral sclerosis, anti-MAG peripheral neuropathy, anti-MOG associated spectrum, Balo concentric sclerosis, brain injury, CAMFAK syndrome, Canavan disease, carbon monoxide toxicity, central pontine myelinolysis, cerebral hypoxia, cerebral ischemia, Charcot–Marie–Tooth disease, chronic inflammatory demyelinating polyneuropathy, chronic traumatic encephalopathy, clinically isolated syndrome (CIS), congenital cataract, copper deficiency associated condition, delayed post-hypoxic leukoencephalopathy, diffuse cerebral sclerosis of Schilder, diffuse myelinoclastic sclerosis, extrapontine myelinolysis Gaucher disease, Guillain–Barré syndrome, hereditary neuropathy, hereditary neuropathy with liability to pressure palsy, HTLV-1–associated myelopathy, Hurler syndrome, hypomyelination, hypoxic brain injury, Krabbe disease, Leber hereditary optic atrophy and related mitochondrial disorders, leukodystrophic disorders, Marchiafava-Bignami disease, metachromatic leukodystrophy, multiple sclerosis (e.g., primary progressive multiple sclerosis (PPMS), relapsing-remitting multiple sclerosis (RRMS), secondary progressive multiple sclerosis (SPMS), progressive relapsing multiple sclerosis, Marburg multiple sclerosis, tumefactive multiple sclerosis, and optic- spinal multiple sclerosis), multiple system atrophy, myelinoclastic disorders, myelopathy, nerve injury, neuromyelitis optica (NMO), Niemann-Pick disease, optic neuropathy, optic neuritis (e.g., acute optic neuritis and chronic relapsing inflammatory optic neuritis (CRION)), osmotic demyelination syndrome, Parkinson's disease, Pelizaeus‐Merzbacher disease, peripheral neuropathy, phenylketonuria, progressive inflammatory neuropathy, progressive multifocal leukoencephalopathy, progressive subcortical ischemic demyelination, reperfusion injury, Schilder disease, solitary sclerosis, spinal cord injury, subacute sclerosing panencephalitis, Tabes dorsalis, Tay-Sachs disease, transverse myelitis, traumatic brain injury, tropical spastic paraparesis, vitamin B12 deficiency, and cerebral palsy.
45. The method of any one of claims 41 to 44, wherein the demyelinating disease is characterized by demyelination of one or more cells within the CNS of the subject.
46. The method of any one of claims 41 to 45, wherein the demyelinating disease is multiple sclerosis.
47. The method of claim 46, wherein the multiple sclerosis comprises a clinically isolated syndrome ("CIS"), relapsing-remitting MS ("RRMS"), secondary progressive MS ("SPMS"), primary progressive MS ("PPMS"), optic neuritis or transverse myelitis.
48. The method of any one of claims 41 to 45, wherein the demyelinating disease is an optic neuritis.
49. The method of any one of claims 41 to 48, wherein treating the demyelinating disease comprises reducing one or more symptoms associated with the demyelinating disease.
50. The method of claim 49, wherein the one or more symptoms comprise one or more of fatigue, dizziness, malaise, elevated fever and high body temperature, extreme sensitivity to cold in the hands and feet, weakness and stiffness in muscles and joints, weight changes, digestive or gastrointestinal problems, low blood pressure, high blood pressure, irritability, anxiety, depression, impaired vision (e.g., blurred vision, double vision, reduction in low contrast visual acuity (LCVA)), ataxia, clonus, spasms, dysarthria, weakness, clumsiness, hand paralysis, hemiparesis, genital anesthesia, sexual dysfunction, incoordination, paresthesias, ocular paralysis, impaired muscle coordination, loss of sensation, tingling, numbness, pain, neurological symptoms, impaired cognition, unsteady gait, balance problems, dizziness, spastic paraparesis, incontinence, hearing problems, speech problems, loss of olfaction, and agusia.
51. A method of promoting the myelination of an axon in a subject in need thereof, the method comprising administering to the subject an effective amount of the compound of any one of claims 1 to 37, or a pharmaceutically acceptable salt or solvate thereof, or the pharmaceutical composition of claim 38.
52. The method of claim 51, wherein promoting the myelination of an axon results in an increase in the expression of one or more of the following markers within the subject: myelin basic protein (MBP) Myelin Oligodendrocyte Glycoprotein (MOG), Oligodendrocyte Specific Protein/Claudin-11, CNPase, proteolipid protein 1 (PLP1), or any combination thereof.
53. The method of claim 51 or 52, wherein the myelination of an axon can be determined by visualizing and/or quantifying the expression of one or more of the following markers: myelin basic protein (MBP) Myelin Oligodendrocyte Glycoprotein (MOG), Oligodendrocyte Specific Protein/Claudin-11, CNPase, proteolipid protein 1 (PLP1), or any combination thereof.
54. A method of promoting the remyelination of a demyelinated axon in a subject in need thereof, the method comprising administering to the subject an effective amount of the compound of any one of claims 1 to 37, or a pharmaceutically acceptable salt or solvate thereof, or the pharmaceutical composition of claim 38.
55. The method of claim 54, wherein promoting the remyelination of a demyelinated axon results in an increase in the expression of one or more of the following markers within the subject: myelin basic protein (MBP) Myelin Oligodendrocyte Glycoprotein (MOG), Oligodendrocyte Specific Protein/Claudin-11, CNPase, proteolipid protein 1 (PLP1), or any combination thereof
56. The method of claim 54 or 55, wherein the remyelination of a demyelinated axon can be determined by visualizing and/or quantifying the expression of one or more of the following markers: myelin basic protein (MBP) Myelin Oligodendrocyte Glycoprotein (MOG), Oligodendrocyte Specific Protein/Claudin-11, CNPase, proteolipid protein 1 (PLP1), or any combination thereof.
57. A method of reducing the demyelination of a myelinated neuronal axon in a subject in need thereof, the method comprising administering to the subject an effective amount of the compound of any one of claims 1 to 37, or a pharmaceutically acceptable salt or solvate thereof, or the pharmaceutical composition of claim 38.
58. The method of claim 57, wherein reducing the demyelination of a myelinated neuronal axon results in an increase in the expression of one or more of the following markers: myelin basic protein (MBP) Myelin Oligodendrocyte Glycoprotein (MOG), Oligodendrocyte Specific Protein/Claudin-11, CNPase, proteolipid protein 1 (PLP1), or any combination thereof.
59. The method of claim 57 or 58, wherein the reduction in the demyelination of a myelinated neuronal axon can be determined by visualizing and/or quantifying the expression of one or more of the following markers: myelin basic protein (MBP) Myelin Oligodendrocyte Glycoprotein (MOG), Oligodendrocyte Specific Protein/Claudin-11, CNPase, proteolipid protein 1 (PLP1), or any combination thereof.
60. A method of activating an oligodendrocyte progenitor cell (OPC) within the central nervous system (CNS) of a subject in need thereof, the method comprising administering to the subject an effective amount of the compound of any one of claims 1 to 37, or a pharmaceutically acceptable salt or solvate thereof, or the pharmaceutical composition of claim 38.
61. A method according to any one of claims 51 to 60, wherein the subject has, or is at risk of developing a demyelinating disease, for example a disease according to any one of claims 44 to 50.
62. A method according to any one of claims 51 to 60, wherein the method is a method of treating or preventing a demyelinating disease, for example a disease according to any one of claims 44 to 50.
63. The method of any one of claims 41 to 62, wherein the compound or the pharmaceutical composition is administered to the subject once.
64. The method of any one of claims 41 to 62, wherein the compound or the pharmaceutical composition is administered to the subject more than once using intermittent dosing.
65. The method of claim 64, wherein the intermittent dosing comprises administering the compound or the pharmaceutical composition to the subject every other day, every three days, every four days, every five days, every six days, once a week, every eight days, every nine days, every 10 days, every 11 days, every 12 days, every 13 days, once every two weeks, once every three weeks, once a month, once every two months, once every three months, once every four months, once every five months, once every six months, or once every twelve months.
66. The method of claim 64 or 65, wherein the intermittent dosing comprises administering to the subject a first dose and a second dose of the compound or the pharmaceutical composition, wherein the second dose is administered at least one day, at least two days, at least three days, at least four days, at least five days, at least six days, at least seven days, at least eight days, at least nine days, at least 10 days, at least 11 days, at least 12 days, at least 13 days, at least two weeks, at least three weeks, at least one month, at least two months, at least three months, at least four months, at least five months, at least six months, or at least 12 months after administering the first dose.
67. The method of claim 66, wherein the second dose is administered to the subject one day, two days, three days, four days, five days, six days, seven days, eight days, nine days, 10 days, 11 days, 12 days, 13 days, two weeks, three weeks, one month, two months, three months, four months, five months, six months, or 12 months after administering the first dose.
68. The method of any one of claims 41 to 67, wherein after the administration, the compound or the pharmaceutical composition can achieve a brain to plasma ratio of greater than 0.1, greater than 0.2, greater than 0.3, greater than 0.4, greater than 0.5, greater than 0.6, greater than 0.7, greater than 0.8, greater than 0.9, greater than 1.0, greater than 1.1, greater than 1.2, greater than 1.3, greater than 1.4, greater than 1.5, greater than 1.6, greater than 1.7, greater than 1.8, greater than 1.9, or greater than 2.0.
69. The method of any one of claims 41 to 68, further comprising administering to the subject an additional therapeutic agent.
70. The method of claim 69, wherein the additional therapeutic agent comprises a standard care of treatment.
71. The method of claim 69 or 70, wherein the additional therapeutic agent comprises an immunomodulatory agent.
72. The method of claim 71, wherein the additional therapeutic agent is selected from interferon beta-1b, interferon beta-1a, peginterferon beta-1a, alemtuzumab, natalizumab, ocrelizumab, ofatumumab, ublituximab-xiiy, glatiramer acetate, teriflunomide, dimethyl fumarate, monomethyl fumarate, diroximel fumarate, fingolimod hydrochloride, siponimod fumaric acid, ozanimod hydrochloride, ponesimod, cladribine, mitoxantrone, a BTK inhibitor, a statin, or a pharmaceutically acceptable salt thereof.
73. The method of any one of claims 69 to 72, wherein the additional therapeutic agent is administered to the subject prior to, concurrently, or after the administration of the compound or the pharmaceutical composition.
74. A method of inducing the differentiation of an oligodendrocyte progenitor cell (OPC) into an oligodendrocyte, the method comprising contacting the OPC with an effective amount of the compound of any one of claims 1 to 37, or a pharmaceutically acceptable salt or solvate thereof, or the pharmaceutical composition of claim 38.
75. The method of claim 74, wherein inducing the differentiation of the OPC into an oligodendrocyte results in an increase in the expression of the following markers in the subject: GPR17, MBP, ASPA, GST-pi, CC1, myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, proteolipid protein 1 (PLP1), or a combination thereof.
76. The method of claim 74 or 75, wherein the differentiation of the OPC into an oligodendrocyte is measured by determining the expression of GPR17, MBP, ASPA, GST- pi, CC1, myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, proteolipid protein 1 (PLP1), or a combination thereof.
77. A method of inhibiting PDGFRα activity in a cell, the method comprising contacting the cell with an effective amount of the compound of any one of claims 1 to 37, or a pharmaceutically acceptable salt or solvate thereof, or the pharmaceutical composition of claim 38.
78. The method of claim 77, wherein the inhibition of the PDGFRα activity is measured by one or more of the following: an in vitro OPC differentiation assay (e.g., as described in Example 132), a cuprizone model for demyelination, an in vivo OPC differentiation assay (e.g., as described in Example 134), an enzymatic PDGFRα kinase assay (e.g., as described in Example 131), or any combination thereof.
79. The method of any one of claims 74 to 78, wherein the contacting occurs ex vivo or in vivo.
80. The method of any one of claims 74 to 79, wherein the method is a method of treating by a therapy.
81. A method of treating a relapsing form of multiple sclerosis in a subject in need thereof, the method comprising administering to the subject an effective amount of the compound of any one of claims 1 to 37, or a pharmaceutically acceptable salt or solvate thereof, or the pharmaceutical composition of claim 38.
82. The method of claim 81, wherein inducing the differentiation of the OPC into an oligodendrocyte results in an increase in the expression of the following marker in the subject: GPR17, MBP, ASPA, GST-pi, CC1, myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, proteolipid protein 1 (PLP1), or a combination thereof.
83. The method of claim 81 or 82, wherein the differentiation of the OPC into an oligodendrocyte is measured by determining the expression of GPR17, MBP, ASPA, GST- pi, CC1, myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, proteolipid protein 1 (PLP1), or a combination thereof.
84. The method of any one of claims 81 to 83, wherein the relapsing form of multiple sclerosis comprises a clinically isolated syndrome ("CIS"), relapsing-remitting MS ("RRMS"), secondary progressive MS ("SPMS"), primary progressive MS ("PPMS"), or transverse myelitis.
85. The compound of any one of claims 1 to 37, or a pharmaceutically acceptable salt or solvate thereof, or the pharmaceutical composition of claim 38, for use in a method of one or more of the following: (i) promote the differentiation of an OPC into an oligodendrocyte, (ii) promote the expression of a protein associated with oligodendrocyte differentiation and/or myelination (e.g., G-protein coupled receptor 17, myelin basic protein (MBP), ASPA, GST-pi, CC1, myelin oligodendrocyte glycoprotein (MOG), oligodendrocyte-specific protein/claudin-11, CNPase, proteolipid protein 1 (PLP1), or a combination thereof), (iii) promote the myelination of an axon, (iv) promote the remyelination of a demyelinated axon, (v) inhibit PDGFRα kinase activity, (vi) achieve a brain to plasma ratio of greater than 0.1 when systemically administered to a subject, and (vii) any combination thereof.
86. A method of treating a PDGF-associated tumor in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of the compound of any one of claims 1 to 37, or a pharmaceutically acceptable salt or solvate thereof, or the pharmaceutical composition of claim 38, wherein after the administration, PDGFRα activity is reduced in the subject.
87. The method of claim 86, wherein the PDGF-associated tumor comprises an oligodendroglioma.
88. The method of claim 86 or 87, wherein the method is a method of treatment by therapy.
PCT/US2024/028330 2023-05-09 2024-05-08 Heterocyclic compounds and uses thereof Pending WO2024233642A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
AU2024269869A AU2024269869A1 (en) 2023-05-09 2024-05-08 Heterocyclic compounds and uses thereof
IL323606A IL323606A (en) 2023-05-09 2025-09-28 Heterocyclic compounds and uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202363501005P 2023-05-09 2023-05-09
US63/501,005 2023-05-09

Publications (1)

Publication Number Publication Date
WO2024233642A1 true WO2024233642A1 (en) 2024-11-14

Family

ID=91302209

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2024/028330 Pending WO2024233642A1 (en) 2023-05-09 2024-05-08 Heterocyclic compounds and uses thereof

Country Status (3)

Country Link
AU (1) AU2024269869A1 (en)
IL (1) IL323606A (en)
WO (1) WO2024233642A1 (en)

Citations (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4044126A (en) 1972-04-20 1977-08-23 Allen & Hanburys Limited Steroidal aerosol compositions and process for the preparation thereof
US4364923A (en) 1972-04-20 1982-12-21 Allen & Hanburs Limited Chemical compounds
US5709874A (en) 1993-04-14 1998-01-20 Emory University Device for local drug delivery and methods for using the same
US5759542A (en) 1994-08-05 1998-06-02 New England Deaconess Hospital Corporation Compositions and methods for the delivery of drugs by platelets for the treatment of cardiovascular and other diseases
US5840674A (en) 1990-11-01 1998-11-24 Oregon Health Sciences University Covalent microparticle-drug conjugates for biological targeting
US5860957A (en) 1997-02-07 1999-01-19 Sarcos, Inc. Multipathway electronically-controlled drug delivery system
US5900252A (en) 1990-04-17 1999-05-04 Eurand International S.P.A. Method for targeted and controlled release of drugs in the intestinal tract and more particularly in the colon
US5948433A (en) 1997-08-21 1999-09-07 Bertek, Inc. Transdermal patch
US5972366A (en) 1994-11-28 1999-10-26 The Unites States Of America As Represented By The Secretary Of The Army Drug releasing surgical implant or dressing material
US5983134A (en) 1995-04-23 1999-11-09 Electromagnetic Bracing Systems Inc. Electrophoretic cuff apparatus drug delivery system
US5985317A (en) 1996-09-06 1999-11-16 Theratech, Inc. Pressure sensitive adhesive matrix patches for transdermal delivery of salts of pharmaceutical agents
US5985307A (en) 1993-04-14 1999-11-16 Emory University Device and method for non-occlusive localized drug delivery
US6004534A (en) 1993-07-23 1999-12-21 Massachusetts Institute Of Technology Targeted polymerized liposomes for improved drug delivery
US6010715A (en) 1992-04-01 2000-01-04 Bertek, Inc. Transdermal patch incorporating a polymer film incorporated with an active agent
US6024975A (en) 1992-04-08 2000-02-15 Americare International Diagnostics, Inc. Method of transdermally administering high molecular weight drugs with a polymer skin enhancer
US6039975A (en) 1995-10-17 2000-03-21 Hoffman-La Roche Inc. Colon targeted delivery system
US6048736A (en) 1998-04-29 2000-04-11 Kosak; Kenneth M. Cyclodextrin polymers for carrying and releasing drugs
US6060082A (en) 1997-04-18 2000-05-09 Massachusetts Institute Of Technology Polymerized liposomes targeted to M cells and useful for oral or mucosal drug delivery
US6071495A (en) 1989-12-22 2000-06-06 Imarx Pharmaceutical Corp. Targeted gas and gaseous precursor-filled liposomes
US6120751A (en) 1997-03-21 2000-09-19 Imarx Pharmaceutical Corp. Charged lipids and uses for the same
US6131570A (en) 1998-06-30 2000-10-17 Aradigm Corporation Temperature controlling device for aerosol drug delivery
US6139865A (en) 1996-10-01 2000-10-31 Eurand America, Inc. Taste-masked microcapsule compositions and methods of manufacture
US6167301A (en) 1995-08-29 2000-12-26 Flower; Ronald J. Iontophoretic drug delivery device having high-efficiency DC-to-DC energy conversion circuit
US6256533B1 (en) 1999-06-09 2001-07-03 The Procter & Gamble Company Apparatus and method for using an intracutaneous microneedle array
US6253872B1 (en) 1996-05-29 2001-07-03 Gmundner Fertigteile Gesellschaft M.B.H & Co., Kg Track soundproofing arrangement
US6261595B1 (en) 2000-02-29 2001-07-17 Zars, Inc. Transdermal drug patch with attached pocket for controlled heating device
US6267983B1 (en) 1997-10-28 2001-07-31 Bando Chemical Industries, Ltd. Dermatological patch and process for producing thereof
US6271359B1 (en) 1999-04-14 2001-08-07 Musc Foundation For Research Development Tissue-specific and pathogen-specific toxic agents and ribozymes
US6274552B1 (en) 1993-03-18 2001-08-14 Cytimmune Sciences, Inc. Composition and method for delivery of biologically-active factors
US6316652B1 (en) 1995-06-06 2001-11-13 Kosta Steliou Drug mitochondrial targeting agents
US20200179389A1 (en) * 2017-04-27 2020-06-11 The Brigham And Women's Hospital Inc. Novel alk2 inhibitors and methods for inhibiting bmp signaling
WO2020264420A1 (en) * 2019-06-28 2020-12-30 Gb002, Inc. Heterocyclic kinase inhibitors and products and uses thereof
WO2023081923A1 (en) * 2021-11-08 2023-05-11 Frequency Therapeutics, Inc. Platelet-derived growth factor receptor (pdgfr) alpha inhibitors and uses thereof

Patent Citations (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4364923A (en) 1972-04-20 1982-12-21 Allen & Hanburs Limited Chemical compounds
US4414209A (en) 1972-04-20 1983-11-08 Allen & Hanburys Limited Micronized aerosol steroids
US4044126A (en) 1972-04-20 1977-08-23 Allen & Hanburys Limited Steroidal aerosol compositions and process for the preparation thereof
US6071495A (en) 1989-12-22 2000-06-06 Imarx Pharmaceutical Corp. Targeted gas and gaseous precursor-filled liposomes
US5900252A (en) 1990-04-17 1999-05-04 Eurand International S.P.A. Method for targeted and controlled release of drugs in the intestinal tract and more particularly in the colon
US5840674A (en) 1990-11-01 1998-11-24 Oregon Health Sciences University Covalent microparticle-drug conjugates for biological targeting
US6010715A (en) 1992-04-01 2000-01-04 Bertek, Inc. Transdermal patch incorporating a polymer film incorporated with an active agent
US6024975A (en) 1992-04-08 2000-02-15 Americare International Diagnostics, Inc. Method of transdermally administering high molecular weight drugs with a polymer skin enhancer
US6274552B1 (en) 1993-03-18 2001-08-14 Cytimmune Sciences, Inc. Composition and method for delivery of biologically-active factors
US5709874A (en) 1993-04-14 1998-01-20 Emory University Device for local drug delivery and methods for using the same
US5985307A (en) 1993-04-14 1999-11-16 Emory University Device and method for non-occlusive localized drug delivery
US6004534A (en) 1993-07-23 1999-12-21 Massachusetts Institute Of Technology Targeted polymerized liposomes for improved drug delivery
US5759542A (en) 1994-08-05 1998-06-02 New England Deaconess Hospital Corporation Compositions and methods for the delivery of drugs by platelets for the treatment of cardiovascular and other diseases
US5972366A (en) 1994-11-28 1999-10-26 The Unites States Of America As Represented By The Secretary Of The Army Drug releasing surgical implant or dressing material
US5983134A (en) 1995-04-23 1999-11-09 Electromagnetic Bracing Systems Inc. Electrophoretic cuff apparatus drug delivery system
US6316652B1 (en) 1995-06-06 2001-11-13 Kosta Steliou Drug mitochondrial targeting agents
US6167301A (en) 1995-08-29 2000-12-26 Flower; Ronald J. Iontophoretic drug delivery device having high-efficiency DC-to-DC energy conversion circuit
US6039975A (en) 1995-10-17 2000-03-21 Hoffman-La Roche Inc. Colon targeted delivery system
US6253872B1 (en) 1996-05-29 2001-07-03 Gmundner Fertigteile Gesellschaft M.B.H & Co., Kg Track soundproofing arrangement
US5985317A (en) 1996-09-06 1999-11-16 Theratech, Inc. Pressure sensitive adhesive matrix patches for transdermal delivery of salts of pharmaceutical agents
US6139865A (en) 1996-10-01 2000-10-31 Eurand America, Inc. Taste-masked microcapsule compositions and methods of manufacture
US5860957A (en) 1997-02-07 1999-01-19 Sarcos, Inc. Multipathway electronically-controlled drug delivery system
US6120751A (en) 1997-03-21 2000-09-19 Imarx Pharmaceutical Corp. Charged lipids and uses for the same
US6060082A (en) 1997-04-18 2000-05-09 Massachusetts Institute Of Technology Polymerized liposomes targeted to M cells and useful for oral or mucosal drug delivery
US5948433A (en) 1997-08-21 1999-09-07 Bertek, Inc. Transdermal patch
US6267983B1 (en) 1997-10-28 2001-07-31 Bando Chemical Industries, Ltd. Dermatological patch and process for producing thereof
US6048736A (en) 1998-04-29 2000-04-11 Kosak; Kenneth M. Cyclodextrin polymers for carrying and releasing drugs
US6131570A (en) 1998-06-30 2000-10-17 Aradigm Corporation Temperature controlling device for aerosol drug delivery
US6271359B1 (en) 1999-04-14 2001-08-07 Musc Foundation For Research Development Tissue-specific and pathogen-specific toxic agents and ribozymes
US6256533B1 (en) 1999-06-09 2001-07-03 The Procter & Gamble Company Apparatus and method for using an intracutaneous microneedle array
US6261595B1 (en) 2000-02-29 2001-07-17 Zars, Inc. Transdermal drug patch with attached pocket for controlled heating device
US20200179389A1 (en) * 2017-04-27 2020-06-11 The Brigham And Women's Hospital Inc. Novel alk2 inhibitors and methods for inhibiting bmp signaling
WO2020264420A1 (en) * 2019-06-28 2020-12-30 Gb002, Inc. Heterocyclic kinase inhibitors and products and uses thereof
WO2023081923A1 (en) * 2021-11-08 2023-05-11 Frequency Therapeutics, Inc. Platelet-derived growth factor receptor (pdgfr) alpha inhibitors and uses thereof

Non-Patent Citations (22)

* Cited by examiner, † Cited by third party
Title
"Concise Dictionary of Biomedicine and Molecular Biology", 2002, CRC PRESS
"GenBank", Database accession no. NC 000004.12
"The Dictionary of Cell and Molecular Biology", 2013, ACADEMIC PRESS
"Using Antibodies", 1999, COLD SPRING HARBOR LABORATORY PRESS
ALQUDAH ET AL., AUDIOL NEUROOTOL, vol. 23, no. 1, 2018, pages 20 - 31
BAX ET AL., DEV. DYN., vol. 239, no. 8, August 2010 (2010-08-01), pages 2307 - 2317
CHEUNG ET AL., VIROLOGY, vol. 176, 1990, pages 546
ED HARLOWDAVID LANE, COLD SPRING HARB. PROTOC., 2006
GACEM ET AL., LIFE, vol. 11, no. 4, April 2021 (2021-04-01), pages 327
HAMASHIMA ET AL., NEUROSCIENCE, vol. 436, June 2020 (2020-06-01), pages 11 - 26
HARLOWLANE: "Antibodies: A Laboratory Manual", 2014, COLD SPRING HARBOR PRESS
JAFARI ET AL., EXPERT OPINION ON DRUG DELIVERY, vol. 16, 2019, pages 583 - 605
KIRKLAND ET AL., J. IMMUNOL, vol. 137, 1986, pages 3614
LI ET AL., PLOS ONE, vol. 11, no. 11, November 2016 (2016-11-01), pages 0165637
MAHERAS ET AL., SCI REP, vol. 8, no. 1, February 2018 (2018-02-01), pages 3798
MOLDENHAUER ET AL., SCAND. J. IMMUNOL., vol. 32, 1990, pages 77
MOREL ET AL., MOL. IMMUNOL., vol. 25, no. 1, 1988, pages 7
OLLER-SALVIA ET AL., CHEM SAC REV, vol. 45, 2016, pages 4690 - 4707
REMINGTON: "The Science and Practice of Pharmacy", 2020, ADADEMIC PRESS.
SPENGLER ET AL., PEPT. RES., vol. 65, 2005, pages 550 - 555
STAHLI ET AL., METHODS IN ENZYMOLOGY, vol. 9, 1983, pages 242
TORKILDSEN ET AL., ACTA NEUROL SCAND SUPPL, vol. 188, 2008, pages 72 - 6

Also Published As

Publication number Publication date
IL323606A (en) 2025-11-01
AU2024269869A1 (en) 2025-10-16

Similar Documents

Publication Publication Date Title
ES2377849T3 (en) Amino-heterocyclic compounds
TWI687220B (en) Use of pyrazolopyrimidine derivatives for the treatment of pi3kδ related disorders
AU2017286868B2 (en) 5,7-dihydro-pyrrolo-pyridine derivatives for treating neurological and neurodegenerative diseases
US7557104B2 (en) Imidazopyrazines as protein kinase inhibitors
CA2866143C (en) Pyrazolo[1,5-a]pyrimidine-based compounds, compositions comprising them, and methods of their use
KR102249236B1 (en) Heterocyclylamines as pi3k inhibitors
CN102292340B (en) Amino-heterocyclic compounds used as pde9 inhibitors
KR20180095625A (en) Novel compounds
JP6506833B2 (en) Imidazopyridazine compounds
US20180044343A1 (en) Bicyclic imidazolo derivative
CA2948589A1 (en) 5-chloro-2-difluoromethoxyphenyl pyrazolopyrimidine compounds which are jak inhibitors
TW201619159A (en) Pyrrolo[2,3-d]pyrimidine derivatives
CN104981472A (en) Heteroaromatic compounds and their use as dopamine d1 ligands
CA2925743C (en) Novel bicyclic pyridinones as gamma-secretase modulators
JP2021518344A (en) ATR Kinase Heterocyclic Inhibitor
EA038365B1 (en) SUBSTITUTED PYRIDO[3,4-b]INDOLES FOR THE TREATMENT OF CARTILAGE DISORDERS
RU2719599C2 (en) Compounds 6,7-dihydro-5h-pyrazolo[5,1-b][1,3]oxazine-2-carboxamide
US20170007615A1 (en) Antibacterial Compounds
JP2018076285A (en) Pharmaceuticals containing bicyclic imidazolo derivatives
US20250129067A1 (en) Platelet-derived growth factor receptor (pdgfr) alpha inhibitors and uses thereof
WO2024233642A1 (en) Heterocyclic compounds and uses thereof
CN121263415A (en) Heterocyclic compounds and their use
CN118574610A (en) AKT3 modulators
CN118475580A (en) Platelet-derived growth factor receptor (PDGFR) alpha inhibitors and uses thereof

Legal Events

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

Ref document number: 24729679

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 323606

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: AU2024269869

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 825702

Country of ref document: NZ

ENP Entry into the national phase

Ref document number: 2024269869

Country of ref document: AU

Date of ref document: 20240508

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 825702

Country of ref document: NZ

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112025023390

Country of ref document: BR

WWE Wipo information: entry into national phase

Ref document number: 2024729679

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2024729679

Country of ref document: EP

Effective date: 20251209