COMPOUNDS TARGETING PRMT5

Provided herein are compounds of Formula (I), or pharmaceutically acceptable salts thereof, pharmaceutical compositions that include a compound described herein (including pharmaceutically acceptable salts of a compound described herein) and methods of synthesizing the same. Also provided herein are methods of treating diseases and/or conditions with a compound of Formula (I), or a pharmaceutically acceptable salt thereof.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
INCORPORATION BY REFERENCE TO ANY PRIORITY APPLICATIONS

Any and all applications for which a foreign or domestic priority claim is identified, for example, in the Application Data Sheet or Request as filed with the present application, are hereby incorporated by reference under 37 CFR 1.57, and Rules 4.18 and 20.6, including U.S. application Ser. No. 16/836,132, filed Mar. 31, 2020, and U.S. Provisional Application Nos. 62/828,282, filed Apr. 2, 2019, 62/877,411, filed Jul. 23, 2019 and 62/946,649, filed Dec. 11, 2019. The present application is a continuation of U.S. application Ser. No. 16/836,132, filed Mar. 31, 2020, which claims priority to U.S. Provisional Application Nos. 62/828,282, filed Apr. 2, 2019, 62/877,411, filed Jul. 23, 2019 and 62/946,649, filed Dec. 11, 2019.

BACKGROUND Field

The present application relates to the fields of chemistry, biochemistry and medicine. Disclosed herein are compounds of Formula (I), or pharmaceutically acceptable salt thereof, pharmaceutical compositions that include a compound described herein (including pharmaceutically acceptable salts of a compound described herein) and methods of synthesizing the same. Also disclosed herein are methods of treating diseases and/or conditions with a compound of Formula (I), or a pharmaceutically acceptable salt thereof.

Description

In mammals, there are nine enzymes in the seven-β-strand family of protein arginine methyltransferases (PRMTs), designated PRMT1-9. These PRMTs are further divided into three types based on the different methylarginine derivatives they produce: Type I PRMTs (PRMT1-4, 6, and 8) catalyze the production of monomethylarginine (MMA) and asymmetric dimethylarginine (ADMA); Type II PRMTs (PRMTS and 9) catalyze MMA and symmetric dimethylarginine (SDMA) production; and Type III enzymes (PRMT7) catalyze only the production of MMA residues.

SUMMARY

Some embodiments disclosed herein relate to a compound of Formula (I), or a pharmaceutically acceptable salt thereof.

Some embodiments disclosed herein relate to a pharmaceutical composition that can contain an effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof.

These are other embodiments are described in greater detail below.

DETAILED DESCRIPTION

PRMT5 is a Type II protein arginine methyltransferase that catalyzes SDMA modification in histones and non-histone substrates, including three subunits of the Survival of Motor Neuron (SMN) complex (SmB, SmD1 and SmD3). These proteins are essential components of the spliceosome machinery (See Friesen et al., Molecular and Cellular Biology (2001) 21(24):8289-8300; Matera et al., Nature Reviews Molecular Cell Biology (2014) 14:108-121; and Meister et al., Current Biology (2001) 11(24):1990-1994), and PRMT5 depletion triggers aberrant splicing in the adult hematopoietic compartment (Bezzi et al., Genes & Development (2013) 27:1903-1916; Koh et al., Nature (2015) 523:96-100; and Liu et al., J. Clin. Invest. (2015) 125(9):3532-3544).

PRMT5 is overexpressed in a variety of human cancers, including several hematological malignancies such as lymphoma and leukemia (Yang et al., Nature Reviews Cancer (2013) 13:37-50 and Chung et al., J. Biol. Chem. (2013) 288(49):35534-35547), as well as liver cancer (Jiang et al., Cancer Medicine (2018) 7(3):869-882), lung cancer (Wei et al., Cancer Science (2012) 103(9): 1640-1650), breast cancer (Powers et al., Cancer Research (2011) 71(16):5579-5587), and colorectal cancer (Cho et al., The EMBO Journal (2012) 31:1785-1797). Enhanced PRMT5 expression correlates with reduced overall survival and higher recurrence rates for patients with hepatocellular carcinoma (HCC) (Jiang et al., Cancer Medicine (2018) 7(3):869-882). Knocking down PRMT5 expression with shRNA can prevent cell proliferation and colony formation in Huh-7 and SK-Hep 1 HCC cells. In a mouse xenograph model for HCC, this approach can result in tumor regression.

Inhibition of PRMT5 has been shown to result in anti-tumor activity in lymphomas (Chan-Penebre et al., Nature Chemical Biology (2015) 11:432-437), MLL-rearranged acute leukemia models (Kaushik et al., Leukemia (2018) 32:499-509), and several other types of leukemia in vitro (Tarighat et al., Leukemia (2016) 30:789-799). In addition, cells lacking MTAP, a critical enzyme in the methionine salvage pathway that is deleted in approximately 15% of all human cancers, can be more sensitive to PRMT5 depletion than MTAP wild type cells (Kryukov et al., Science (2016) 351(6278):1214-1218; Marjon et al., Cell Report (2016) 15(3):574-587; and Mavrakis et al., Science (2016) 351(6278):1208-1213). Small molecule inhibitors of PRMTS have shown preferential impairment of cell viability for MTAP-null cancer cell lines compared with isogenic MTAP-expressing counterparts, making PRMT5 a potential vulnerability across multiple cancer lineages.

Definitions

Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of ordinary skill in the art. All patents, applications, published applications and other publications referenced herein are incorporated by reference in their entirety unless stated otherwise. In the event that there are a plurality of definitions for a term herein, those in this section prevail unless stated otherwise.

Whenever a group is described as being “optionally substituted” that group may be unsubstituted or substituted with one or more of the indicated substituents. Likewise, when a group is described as being “unsubstituted or substituted” if substituted, the substituent(s) may be selected from one or more of the indicated substituents. If no substituents are indicated, it is meant that the indicated “optionally substituted” or “substituted” group may be substituted with one or more group(s) individually and independently selected from deuterium, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl(alkyl), (heterocyclyl)alkyl, hydroxy, alkoxy, acyl, cyano, halogen, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, O-carboxy, isocyanato, thiocyanato, nitro, azido, silyl, sulfenyl, sulfinyl, sulfonyl, haloalkyl, haloalkoxy, trihalomethanesulfonyl, trihalomethanesulfonamido, an amino, a mono-substituted amino group and a di-substituted amino group.

As used herein, “Ca to Cb” in which “a” and “b” are integers refer to the number of carbon atoms in an alkyl, alkenyl or alkynyl group, or the number of carbon atoms in the ring of a cycloalkyl, cycloalkenyl, aryl, heteroaryl or heterocyclyl group. That is, the alkyl, alkenyl, alkynyl, ring of the cycloalkyl, ring of the cycloalkenyl, ring of the aryl, ring of the heteroaryl or ring of the heterocyclyl can contain from “a” to “b”, inclusive, carbon atoms. Thus, for example, a “C1 to C4 alkyl” group refers to all alkyl groups having from 1 to 4 carbons, that is, CH3—, CH3CH2—, CH3CH2CH2—, (CH3)2CH—, CH3CH2CH2CH2—, CH3CH2CH(CH3)— and (CH3)3C—. If no “a” and “b” are designated with regard to an alkyl, alkenyl, alkynyl, cycloalkyl cycloalkenyl, aryl, heteroaryl or heterocyclyl group, the broadest range described in these definitions is to be assumed.

As used herein, “alkyl” refers to a straight or branched hydrocarbon chain that comprises a fully saturated (no double or triple bonds) hydrocarbon group. The alkyl group may have 1 to 20 carbon atoms (whenever it appears herein, a numerical range such as “1 to 20” refers to each integer in the given range; e.g., “1 to 20 carbon atoms” means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 20 carbon atoms, although the present definition also covers the occurrence of the term “alkyl” where no numerical range is designated). The alkyl group may also be a medium size alkyl having 1 to 10 carbon atoms. The alkyl group could also be a lower alkyl having 1 to 6 carbon atoms. The alkyl group of the compounds may be designated as “C1-C4 alkyl” or similar designations. By way of example only, “C1-C4 alkyl” indicates that there are one to four carbon atoms in the alkyl chain, i.e., the alkyl chain is selected from methyl, ethyl, propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl and t-butyl. Typical alkyl groups include, but are in no way limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl and hexyl. The alkyl group may be substituted or unsubstituted.

As used herein, “alkenyl” refers to an alkyl group that contains in the straight or branched hydrocarbon chain one or more double bonds. The length of an alkenyl can vary. For example, the alkenyl can be a C2-4 alkenyl, C2-6 alkenyl or C2-8 alkenyl. Examples of alkenyl groups include allenyl, vinylmethyl and ethenyl. An alkenyl group may be unsubstituted or substituted.

As used herein, “alkynyl” refers to an alkyl group that contains in the straight or branched hydrocarbon chain one or more triple bonds. The length of an alkynyl can vary. For example, the alkynyl can be a C2-4 alkynyl, C2-6 alkynyl or C2-8 alkynyl. Examples of alkynyls include ethynyl and propynyl. An alkynyl group may be unsubstituted or substituted.

As used herein, “cycloalkyl” refers to a completely saturated (no double or triple bonds) mono- or multi-cyclic hydrocarbon ring system. When composed of two or more rings, the rings may be joined together in a fused fashion. Cycloalkyl groups can contain 3 to 10 atoms in the ring(s). 3 to 8 atoms in the ring(s) or 3 to 6 atoms in the ring(s). A cycloalkyl group may be unsubstituted or substituted. Typical cycloalkyl groups include, but are in no way limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl.

As used herein, “cycloalkenyl” refers to a mono- or multi-cyclic hydrocarbon ring system that contains one or more double bonds in at least one ring; although, if there is more than one, the double bonds cannot form a fully delocalized pi-electron system throughout all the rings (otherwise the group would be “aryl,” as defined herein). When composed of two or more rings, the rings may be connected together in a fused fashion. A cycloalkenyl can contain 3 to 10 atoms in the ring(s) or 3 to 8 atoms in the ring(s). A cycloalkenyl group may be unsubstituted or substituted.

As used herein, “aryl” refers to a carbocyclic (all carbon) monocyclic or multicyclic aromatic ring system (including fused ring systems where two carbocyclic rings share a chemical bond) that has a fully delocalized pi-electron system throughout all the rings. The number of carbon atoms in an aryl group can vary. For example, the aryl group can be a C6-C14 aryl group, a C6-C10 aryl group, or a C6 aryl group. Examples of aryl groups include, but are not limited to, benzene, naphthalene and azulene. An aryl group may be substituted or unsubstituted.

As used herein, “heteroaryl” refers to a monocyclic, bicyclic and tricyclic aromatic ring system (a ring system with fully delocalized pi-electron system) that contain(s) one or more heteroatoms (for example, 1 to 5 heteroatoms), that is, an element other than carbon, including but not limited to, nitrogen, oxygen and sulfur. The number of atoms in the ring(s) of a heteroaryl group can vary. For example, the heteroaryl group can contain 4 to 14 atoms in the ring(s), 5 to 10 atoms in the ring(s) or 5 to 6 atoms in the ring(s). Furthermore, the term “heteroaryl” includes fused ring systems where two rings, such as at least one aryl ring and at least one heteroaryl ring, or at least two heteroaryl rings, share at least one chemical bond. Examples of heteroaryl rings include, but are not limited to, furan, furazan, thiophene, benzothiophene, phthalazine, pyrrole, oxazole, benzoxazole, 1,2,3-oxadiazole, 1,2,4-oxadiazole, thiazole, 1,2,3-thiadiazole, 1,2,4-thiadiazole, benzothiazole, imidazole, benzimidazole, indole, indazole, pyrazole, benzopyrazole, isoxazole, benzoisoxazole, isothiazole, triazole, benzotriazole, thiadiazole, tetrazole, pyridine, pyridazine, pyrimidine, pyrazine, purine, pteridine, quinoline, isoquinoline, quinazoline, quinoxaline, cinnoline and triazine. A heteroaryl group may be substituted or unsubstituted.

As used herein, “heterocyclyl” refers to a monocyclic, bicyclic and tricyclic ring system wherein carbon atoms together with from 1 to 5 heteroatoms constitute said ring system. A heterocycle may optionally contain one or more unsaturated bonds situated in such a way, however, that a fully delocalized pi-electron system does not occur throughout all the rings. The number of atoms in the ring(s) of a heterocyclyl group can vary. For example, the heterocyclyl group can contain 4 to 14 atoms in the ring(s), 5 to 10 atoms in the ring(s) or 5 to 6 atoms in the ring(s). The heteroatom(s) is an element other than carbon including, but not limited to, oxygen, sulfur and nitrogen. A heterocycle may further contain one or more carbonyl or thiocarbonyl functionalities, so as to make the definition include oxo-systems and thio-systems such as lactams, lactones, cyclic imides, cyclic thioimides and cyclic carbamates. When composed of two or more rings, the rings may be joined together in a fused fashion. Additionally, any nitrogens in a heterocyclyl may be quaternized. Heterocyclyl groups may be unsubstituted or substituted. Examples of such “heterocyclyl groups include but are not limited to, 1,3-dioxin, 1,3-dioxane, 1,4-dioxane, 1,2-dioxolane, 1,3-dioxolane, 1,4-dioxolane, 1,3-oxathiane, 1,4-oxathiin, 1,3-oxathiolane, 1,3-dithiole, 1,3-dithiolane, 1,4-oxathiane, tetrahydro-1,4-thiazine, 2H-1,2-oxazine, maleimide, succinimide, barbituric acid, thiobarbituric acid, dioxopiperazine, hydantoin, dihydrouracil, trioxane, hexahydro-1,3,5-triazine, imidazoline, imidazolidine, isoxazoline, isoxazolidine, oxazoline, oxazolidine, oxazolidinone, thiazoline, thiazolidine, morpholine, oxirane, piperidine N-Oxide, piperidine, piperazine, pyrrolidine, pyrrolidone, pyrrolidione, 4-piperidone, pyrazoline, pyrazolidine, 2-oxopyrrolidine, tetrahydropyran, 4H-pyran, tetrahydrothiopyran, thiomorpholine, thiomorpholine sulfoxide, thiomorpholine sulfone and their benzo-fused analogs (e.g., benzimidazolidinone, tetrahydroquinoline and 3,4-methylenedioxyphenyl).

As used herein, “aryl(alkyl)” refer to an aryl group connected, as a substituent, via a lower alkylene group. The lower alkylene and aryl group of an aryl(alkyl) may be substituted or unsubstituted. Examples include but are not limited to benzyl, 2-phenyl(alkyl), 3-phenyl(alkyl) and naphthyl(alkyl).

As used herein, “heteroaryl(alkyl)” refer to a heteroaryl group connected, as a substituent, via a lower alkylene group. The lower alkylene and heteroaryl group of heteroaryl(alkyl) may be substituted or unsubstituted. Examples include but are not limited to 2-thienyl(alkyl), 3-thienyl(alkyl), furyl(alkyl), thienyl(alkyl), pyrrolyl(alkyl), pyridyl(alkyl), isoxazolyl(alkyl), imidazolyl(alkyl) and their benzo-fused analogs.

A “(heterocyclyl)alkyl” refer to a heterocyclic group connected, as a substituent, via a lower alkylene group. The lower alkylene and heterocyclyl of a heterocyclyl(alkyl) may be substituted or unsubstituted. Examples include but are not limited tetrahydro-2H-pyran-4-yl(methyl), piperidin-4-yl(ethyl), piperidin-4-yl(propyl), tetrahydro-2H-thiopyran-4-yl(methyl) and 1,3-thiazinan-4-yl(methyl).

“Lower alkylene groups” are straight-chained —CH2— tethering groups, forming bonds to connect molecular fragments via their terminal carbon atoms. Examples include but are not limited to methylene (—CH2—), ethylene (—CH2CH2—), propylene (—CH2CH2CH2—) and butylene (—CH2CH2CH2CH2—). A lower alkylene group can be substituted by replacing one or more hydrogen of the lower alkylene group with a substituent(s) listed under the definition of “substituted.”

As used herein, “alkoxy” refers to the formula OR wherein R is an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl) is defined herein. A non-limiting list of alkoxys are methoxy, ethoxy, n-propoxy, 1-methylethoxy (isopropoxy), n-butoxy, iso-butoxy, sec-butoxy, tert-butoxy, phenoxy and benzoxy. An alkoxy may be substituted or unsubstituted.

As used herein, “acyl” refers to a hydrogen an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl) connected, as substituents, via a carbonyl group. Examples include formyl, acetyl, propanoyl, benzoyl and acryl. An acyl may be substituted or unsubstituted.

As used herein, “haloalkyl” refers to an alkyl group in which one or more of the hydrogen atoms are replaced by a halogen (e.g., mono-haloalkyl, di-haloalkyl and tri-haloalkyl). Such groups include but are not limited to, chloromethyl, fluoromethyl, difluoromethyl, trifluoromethyl, 1-chloro-2-fluoromethyl and 2-fluoroisobutyl. A haloalkyl may be substituted or unsubstituted.

As used herein, “haloalkoxy” refers to a O-alkyl group in which one or more of the hydrogen atoms are replaced by a halogen (e.g., mono-haloalkoxy, di-haloalkoxy and tri-haloalkoxy). Such groups include but are not limited to, chloromethoxy, fluoromethoxy, difluoromethoxy, trifluoromethoxy, 1-chloro-2-fluoromethoxy and 2-fluoroisobutoxy. A haloalkoxy may be substituted or unsubstituted.

A “sulfenyl” group refers to an “—SR” group in which R can be hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). A sulfenyl may be substituted or unsubstituted.

A “sulfinyl” group refers to an “—S(═O)—R” group in which R can be the same as defined with respect to sulfenyl. A sulfinyl may be substituted or unsubstituted.

A “sulfonyl” group refers to an “SO2R” group in which R can be the same as defined with respect to sulfenyl. A sulfonyl may be substituted or unsubstituted.

An “O-carboxy” group refers to a “RC(═O)O—” group in which R can be hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl), as defined herein. An O-carboxy may be substituted or unsubstituted.

The terms “ester” and “C-carboxy” refer to a “—C(═O)OR” group in which R can be the same as defined with respect to O-carboxy. An ester and C-carboxy may be substituted or unsubstituted.

A “thiocarbonyl” group refers to a “—C(═S)R” group in which R can be the same as defined with respect to O-carboxy. A thiocarbonyl may be substituted or unsubstituted.

A “trihalomethanesulfonyl” group refers to an “X3CSO2—” group wherein each X is a halogen.

A “trihalomethanesulfonamido” group refers to an “X3CS(O)2N(RA)—” group wherein each X is a halogen, and RA is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl).

The term “amino” as used herein refers to a —NH2 group.

As used herein, the term “hydroxy” refers to a —OH group.

A “cyano” group refers to a “—CN” group.

The term “azido” as used herein refers to a —N3 group.

An “isocyanato” group refers to a “—NCO” group.

A “thiocyanato” group refers to a “—CNS” group.

An “isothiocyanato” group refers to an “—NCS” group.

A “mercapto” group refers to an “—SH” group.

A “carbonyl” group refers to a C═O group.

An “S-sulfonamido” group refers to a “—SO2N(RARB)” group in which RA and RB can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). An S-sulfonamido may be substituted or unsubstituted.

An “N-sulfonamido” group refers to a “RSO2N(RA)—” group in which R and RA can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). An N-sulfonamido may be substituted or unsubstituted.

An “O-carbamyl” group refers to a “—OC(═O)N(RARB)” group in which RA and RB can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). An O-carbamyl may be substituted or unsubstituted.

An “N-carbamyl” group refers to an “ROC(═O)N(RA)—” group in which R and RA can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). An N-carbamyl may be substituted or unsubstituted.

An “O-thiocarbamyl” group refers to a “—OC(═S)—N(RARB)” group in which RA and RB can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). An O-thiocarbamyl may be substituted or unsubstituted.

An “N-thiocarbamyl” group refers to an “ROC(═S)N(RA)—” group in which R and RA can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). An N-thiocarbamyl may be substituted or unsubstituted.

A “C-amido” group refers to a “—C(═O)N(RARB)” group in which RA and RB can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). A C-amido may be substituted or unsubstituted.

An “N-amido” group refers to a “RC(═O)N(RA)—” group in which R and RA can be independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). An N-amido may be substituted or unsubstituted.

The term “halogen atom” or “halogen” as used herein, means any one of the radio-stable atoms of column 7 of the Periodic Table of the Elements, such as, fluorine, chlorine, bromine and iodine.

Where the numbers of substituents is not specified (e.g. haloalkyl), there may be one or more substituents present. For example “haloalkyl” may include one or more of the same or different halogens. As another example, “C1-C3 alkoxyphenyl” may include one or more of the same or different alkoxy groups containing one, two or three atoms.

As used herein, the abbreviations for any protective groups, amino acids and other compounds, are, unless indicated otherwise, in accord with their common usage, recognized abbreviations, or the IUPAC-IUB Commission on Biochemical Nomenclature (See, Biochem. 11:942-944 (1972)).

The term “pharmaceutically acceptable salt” refers to a salt of a compound that does not cause significant irritation to an organism to which it is administered and does not abrogate the biological activity and properties of the compound. In some embodiments, the salt is an acid addition salt of the compound. Pharmaceutical salts can be obtained by reacting a compound with inorganic acids such as hydrohalic acid (e.g., hydrochloric acid or hydrobromic acid), sulfuric acid, nitric acid and phosphoric acid. Pharmaceutical salts can also be obtained by reacting a compound with an organic acid such as aliphatic or aromatic carboxylic or sulfonic acids, for example formic, acetic, succinic, lactic, malic, tartaric, citric, ascorbic, nicotinic, methanesulfonic, ethanesulfonic, p-toluenesulfonic, salicylic or naphthalenesulfonic acid. Pharmaceutical salts can also be obtained by reacting a compound with a base to form a salt such as an ammonium salt, an alkali metal salt, such as a sodium or a potassium salt, an alkaline earth metal salt, such as a calcium or a magnesium salt, a salt of organic bases such as dicyclohexylamine, N-methyl-D-glucamine, tris(hydroxymethyl)methylamine, C1-C7 alkylamine, cyclohexylamine, triethanolamine, ethylenediamine, and salts with amino acids such as arginine and lysine.

Terms and phrases used in this application, and variations thereof, especially in the appended claims, unless otherwise expressly stated, should be construed as open ended as opposed to limiting. As examples of the foregoing, the term ‘including’ should be read to mean ‘including, without limitation,’ ‘including but not limited to,’ or the like; the term ‘comprising’ as used herein is synonymous with ‘including,’ ‘containing,’ or ‘characterized by,’ and is inclusive or open-ended and does not exclude additional, unrecited elements or method steps; the term ‘having’ should be interpreted as ‘having at least;’ the term ‘includes’ should be interpreted as ‘includes but is not limited to;’ the term ‘example’ is used to provide exemplary instances of the item in discussion, not an exhaustive or limiting list thereof. In addition, the term “comprising” is to be interpreted synonymously with the phrases “having at least” or “including at least”. When used in the context of a compound or composition, the term “comprising” means that the compound or composition includes at least the recited features or components, but may also include additional features or components.

With respect to the use of substantially any plural and/or singular terms herein, those having skill in the art can translate from the plural to the singular and/or from the singular to the plural as is appropriate to the context and/or application. The various singular/plural permutations may be expressly set forth herein for sake of clarity. The indefinite article “a” or “an” does not exclude a plurality.

It is understood that, in any compound described herein having one or more chiral centers, if an absolute stereochemistry is not expressly indicated, then each center may independently be of (R)-configuration or (S)-configuration or a mixture thereof. Thus, the compounds provided herein may be enantiomerically pure, enantiomerically enriched, racemic mixture, diastereomerically pure, diastereomerically enriched, or a stereoisomeric mixture. In addition it is understood that, in any compound described herein having one or more double bond(s) generating geometrical isomers that can be defined as E or Z, each double bond may independently be E or Z a mixture thereof. Likewise, it is understood that, in any compound described, all tautomeric forms are also intended to be included.

It is to be understood that where compounds disclosed herein have unfilled valencies, then the valencies are to be filled with hydrogens or isotopes thereof, e.g., hydrogen-1 (protium) and hydrogen-2 (deuterium).

It is understood that the compounds described herein can be labeled isotopically. Substitution with isotopes such as deuterium may afford certain therapeutic advantages resulting from greater metabolic stability, such as, for example, increased in vivo half-life or reduced dosage requirements. Each chemical element as represented in a compound structure may include any isotope of said element. For example, in a compound structure a hydrogen atom may be explicitly disclosed or understood to be present in the compound. At any position of the compound that a hydrogen atom may be present, the hydrogen atom can be any isotope of hydrogen, including but not limited to hydrogen-1 (protium) and hydrogen-2 (deuterium). Thus, reference herein to a compound encompasses all potential isotopic forms unless the context clearly dictates otherwise.

Where a range of values is provided, it is understood that the upper and lower limit, and each intervening value between the upper and lower limit of the range is encompassed within the embodiments.

Compounds

Some embodiments disclosed herein relate to a compound of Formula (I), or a pharmaceutically acceptable salt thereof:

wherein: B1 can be an optionally substituted

an optionally substituted

an optionally substituted

or an optionally substituted

wherein X1 can be N (nitrogen) or CRC1; X2 can be N (nitrogen) or CRC2; X3 can be N (nitrogen) or CRC3; X4 can be N (nitrogen) or CRC4; X5 can be N (nitrogen) or CRC5; and RC1, RC2, RC3, RC4 and RC5 can be independently hydrogen or halogen; R1B, R1C, R1D and R1E can be independently hydrogen, halogen, hydroxy, an unsubstituted C1-4 alkyl, an unsubstituted C2-4 alkenyl, an unsubstituted C3-C6 cycloalkyl, an unsubstituted C1-4 alkoxy or NRA1RA2; and RA1 and RA2 can be independently selected from hydrogen, hydroxy, an unsubstituted C1-4 alkyl, an unsubstituted C1-4 alkoxy and —C(═O)RC6, wherein RC6 can be hydrogen, an unsubstituted C1-4 alkyl or an unsubstituted C3-4 monocyclic cycloalkyl; R1 can be hydrogen or an unsubstituted C1-4 alkyl; R2A can be hydrogen or an unsubstituted C1-4 alkyl; R2B can be halogen, OH, —O—C(═O)—C1-4 alkyl or —O—C(═O)—CH(R1′)—NH2, wherein R1′ can be H, —CH3, —CH(CH3)2, —CH2—CH(CH3)2 or —CH(CH3)—CH(CH3)2; R3A can be hydrogen, an unsubstituted or a substituted C1-4 alkyl, an unsubstituted or a substituted C2-4 alkenyl or an unsubstituted or a substituted C2-4 alkynyl, wherein when the C1-4 alkyl, the C2-4 alkenyl and the C2-4 alkynyl are substituted, each can be independently substituted with 1 or more fluoros; R3B can be halogen, OH, —O—C(═O)—C1-4 alkyl or —O—C(═O)—CH(R1″)—NH2, wherein R1″ is H, —CH3, —CH(CH3)2, —CH2—CH(CH3)2 or —CH(CH3)—CH(CH3)2; R4A can be —(CRD1RE1)(CRD2RE2)n-RF1, —(CRG1RH1)—O—RJ1, —O—(CRK1RL1)—RM1 or —(CRN1RO1)p-RP1; wherein RD1, RE1, RD2 and RE2 can be independently selected from hydrogen, halogen, hydroxy and an unsubstituted C1-3 alkyl; n can be 0 or 1; and RF1 can be an unsubstituted or a substituted aryl, an unsubstituted or a substituted heteroaryl or an unsubstituted or a substituted heterocyclyl; or RD1 and RE1 can be taken together with the carbon to which RD1 and RE1 are attached to form an unsubstituted cyclopropyl ring; and RD2 and RE2 can be independently selected from hydrogen, halogen, hydroxy and an unsubstituted C1-3 alkyl; n can be 1; and RF1 can be an unsubstituted or a substituted aryl, an unsubstituted or a substituted heteroaryl or an unsubstituted or a substituted heterocyclyl; or RD1 and RE2 can be independently selected from hydrogen, halogen, hydroxy and an unsubstituted C1-3 alkyl; RE1 and RD2 can be taken together with the carbon to which RE1 and RD2 are attached to form an unsubstituted cyclopropyl ring; n can be 1; and RF1 can be an unsubstituted or a substituted aryl, an unsubstituted or a substituted heteroaryl or an unsubstituted or a substituted heterocyclyl; or RD1 and RE2 can be independently selected from hydrogen, halogen, hydroxy and an unsubstituted C1-3 alkyl; RE1 and RD2 together form a double bond; n can be 1; and RF1 can be an unsubstituted or a substituted aryl, an unsubstituted or a substituted heteroaryl or an unsubstituted or a substituted heterocyclyl; RG1, RH1, RK1, RL1, RN1 and RO1 can be independently selected from hydrogen, halogen, hydroxy and an unsubstituted C1-3 alkyl; RJ1 and RM1 can be independently an unsubstituted or a substituted aryl, an unsubstituted or a substituted heteroaryl or an unsubstituted or a substituted heterocyclyl; RP1 can be an unsubstituted or a substituted heteroaryl; and p can be 3 or 4; R4B can be halogen, cyano, azido, —C(═O)NH2, an unsubstituted or a substituted C1-4 alkyl, an unsubstituted or a substituted C2-4 alkenyl, an unsubstituted or a substituted C2-4 alkynyl or an unsubstituted or a substituted C3-C4 cycloalkyl, wherein when the C1-4 alkyl is substituted, the C1-4 alkyl can be substituted with 1 or more substituents independently selected from halogen, OH and cyano, and wherein when the C2-4 alkenyl is substituted, the C2-4 alkenyl can be substituted independently with 1 or more halogens; Z1 can be CR5AR5B, O (oxygen), S (sulfur) or N(an unsubstituted C1-4 alkyl); R5A and R5B can be independently hydrogen, halogen, cyano or an unsubstituted or a substituted C1-4 alkyl, wherein when the C1-4 alkyl is substituted, the C1-4 alkyl can be substituted with 1 or more substituents independently selected from fluoro and hydroxy; or R5A and R5B together with the carbon R5A and R5B are attached can form a double bond optionally substituted with one or two halogen, R5A and R5B together with the carbon R5A and R5B are attached can form an unsubstituted cyclopropyl or R5A and R5B together with the carbon R5A and R5B are attached can form an unsubstituted or a substituted oxetane, wherein when the oxetane is substituted, the oxetane can be substituted independently with 1 or 2 halogens; or R2A and R2B together with the carbon R2A and R2B are attached can form a 3, 4 or 5 membered monocyclic cycloalkyl or a 3, 4 or 5 membered monocyclic heterocyclyl; or R3A and R3B together with the carbon R3A and R3B are attached can form a 3, 4 or 5 membered monocyclic cycloalkyl or a 3, 4 or 5 membered monocyclic heterocyclyl; or R4B and R3B together with the carbon R4B and R3B are attached can form an unsubstituted oxetane; or R4B and R5B together with the carbon R4B and R5B are attached can form an unsubstituted cyclopropyl; or R1 and R5B together with the carbon R1 and R5B are attached can form an unsubstituted cyclopropyl; or when Z1 is O, then R2B and R4B can be connected via —(CH2)y-O—, wherein y can be 1 or 2,

wherein NRE3 can be RE3 can be hydrogen or an unsubstituted C1-7 alkyl or

The 5-membered ring of Formula (I) can be a carbocyclyl or a heterocyclyl. In some embodiments, the 5-membered ring of Formula (I) can be a carbocyclyl when Z1 is CR5AR5B. Various substituents can be present at R5A and R5B. In some embodiments, R5A and R5B can be each hydrogen such that Z1 is CH2. In some embodiments, at least one of R5A and R5B can be halogen, for example F. In some embodiments, R5A and R5B can be each halogen. When R5A and R5B are each halogen, the halogens can be the same or different. An example of R5A and R5B each being halogen is CF2. In some embodiments, at least one of R5A and R5B can be cyano. In some embodiments, at least one of R5A and R5B can be an unsubstituted C1-4 alkyl. Examples of unsubstituted C1-4 alkyls include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl. In some embodiments, one of R5A and R5B can be an unsubstituted C1-4 alkyl (such as those described herein); and the other R5A and R5B can be hydrogen. In some embodiments, at least one of R5A and R5B can be a substituted C1-4 alkyl (such as those C1-4 alkyls described herein) substituted with 1 or more substituents independently selected from fluoro and hydroxy. Those skilled in the art understand that when Z1 is CR5AR5B, the carbon to which R5A and R5B are attached can be a stereocenter. In some embodiments, the carbon to which R5A and R5B are attached can be in the R-configuration

In other embodiments, the carbon to which R5A and R5B are attached can be in the S-configuration

In some embodiments, Z1 can be CR5AR5B, wherein R5A and R5B together with the carbon R5A and R5B are attached form a double bond optionally substituted with one or two halogen. For example, Z1 can be C═CH2, C═CCl2 or C═CF2. In other embodiments, when Z1 is CR5AR5B, R5A and R5B together with the carbon R5A and R5B are attached form an unsubstituted cyclopropyl. In still other embodiments, when Z1 is CR5AR5B, R5A and R5B together with the carbon R5A and R5B are attached form an unsubstituted or a substituted oxetane, wherein when the oxetane is substituted, the oxetane is substituted independently with 1 or 2 halogens (for example, fluoro or chloro). When R5A and R5B together with the carbon R5A and R5B are attached form an unsubstituted cyclopropyl or an unsubstituted or a substituted oxetane, the 5-membered ring of Formula (I) and the unsubstituted cyclopropyl or an unsubstituted or a substituted oxetane are connected in a spiro-manner.

As described herein, the 5-membered ring of Formula (I) can be a heterocyclyl. In some embodiments, Z1 can be S (sulfur). In other embodiments, Z1 can be N (an unsubstituted C1-4 alkyl). Exemplary C1-4 alkyls are described herein, for example, methyl, ethyl, n-propyl, isopropyl, n-butyl, iso-butyl and tert-butyl.

The 2′-position of the 5-membered ring of Formula (I) can have present various substituents. The positions of the 5-membered ring as referred to herein are as follows:

In some embodiments, R2A can be hydrogen. In other embodiments, R2A can be an unsubstituted C1-4 alkyl. Suitable examples of C1-4 alkyls are provided herein and include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl. In some embodiments, R2B can be OH. In other embodiments, R2B can be —O—C(═O)—C1-4 alkyl, such as —O—C(═O)—CH3, —O—C(—O)—CH2CH3, —O—C(═O)—CH2CH2CH3, —O—C(═O)—CH2CH2CH2CH3, —O—C(═O)—CH(CH3)2 and —O—C(═O)—C(CH3)3. In still other embodiments, R2B can be an alpha-amino acid linked via its carboxy group. Alpha-amino acids are known to those skilled in the art, and include, but are not limited to, alanine, asparagine, aspartate, cysteine, glutamate, glutamine, glycine, proline, serine, tyrosine, arginine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan and valine. In yet still other embodiments, R2B can be —O—C(═O)—CH(R1″)—NH2, wherein R1″ can be H, —CH3, —CH(CH3)2, —CH2—CH(CH3)2 or —CH(CH3)—CH(CH3)2. In some embodiments, R2A can be halogen. Examples of halogens include F, Cl, Br and I. In yet still other embodiments, R2A and R2B together with the carbon R2A and R2B are attached form a 3, 4 or 5 membered monocyclic cycloalkyl or a 3, 4 or 5 membered monocyclic heterocyclyl. The 3, 4 or 5 membered monocyclic heterocyclyl formed from R2A and R2B being taken together with the carbon to which R2A and R2B are attached include, but are not limited to, oxetane and thietane. As described herein, R2B can be —O—C(═O)—C1-4 alkyl, an alpha-amino acid linked via its carboxy group or —O—C(═O)—CH(R1″)—NH2, and those skilled in the art understand that when R2B is one of the aforementioned substituents, that compound of Formula (I) can be considered a prodrug of the corresponding a compound of Formula (I) where R2B is OH. In some embodiments, In other embodiments, R2B can be halogen, —O—C(═O)—C1-4 alkyl or —O—C(═O)—CH(R1′)—NH2 and/or R3B can be halogen, —O—C(═O)—C1-4 alkyl or —O—C(═O)—CH(R1″)—NH2.

A variety of substituents can also be present at the 3′-position of the 5-membered ring of Formula (I). In some embodiments, R3A can be hydrogen. In other embodiments, R3A can be an unsubstituted C1-4 alkyl, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl. In still other embodiments, R3A can be a substituted C1-4 alkyl (such as those described herein) substituted with 1 or more fluoros. In some embodiments, R3A can be an unsubstituted C2-4 alkenyl. In other embodiments, R3A can be a substituted C2-4 alkenyl substituted with 1 or more fluoros. In still other embodiments, R3A can be an unsubstituted C2-4 alkynyl. In yet still other embodiments, R3A can be a substituted C2-4 alkynyl substituted with 1 or more fluoros.

Further groups can be present at the 3′-position of the 5-membered ring of Formula (I). In some embodiments, R3B can be OH. In other embodiments, R3B can be —O—C(═O)—C1-4 alkyl. Exemplary C1-4 alkyls are described herein. In still other embodiments, R3B can be an alpha-amino acid linked via its carboxy group. Several alpha-amino acids are known to those skilled in the art, and described herein. In yet still other embodiments, R3B can be —O—C(═O)—CH(R1″)—NH2, wherein R1′ can be H, —CH3, —CH(CH3)2, —CH2—CH(CH3)2 or —CH(CH3)—CH(CH3)2. In some embodiments, R3B can be halogen. For example, R3B can be fluoro. In some embodiments, R3A and R3B together with the carbon R3A and R3B are attached form a 3, 4 or 5 membered monocyclic cycloalkyl or a 3, 4 or 5 membered monocyclic heterocyclyl. When R3B is —O—C(═O)—C1-4 alkyl, an alpha-amino acid linked via its carboxy group or —O—C(═O)—CH(R1″)—NH2, the compound of Formula (I) can be considered a prodrug of the corresponding a compound of Formula (I) where R3B is OH. For example, when R2B and R3B are each —O—C(═O)—C1-4 alkyl, that compound of Formula (I) can be considered a prodrug a compound of Formula (I) where R2B and R3B are each OH. An example of this type of prodrug is Compound 26, wherein Compound 26 being considered a prodrug of Compound 12. The structure of Compounds 12 and 26 are provided herein.

As with the other positions on the 5-membered ring, the substituents present at the 4′-position can vary. In some embodiments, R4A can be —(CRD1RE1)(CRD2RE2)n-RF1. Further, the substituents for RD1, RE1, RD2 and RE2 can also vary. In some embodiments, n can be 0. In other embodiments, n can be 1. In some embodiments, RD1, RE1, RD2 and RE2 can be each hydrogen, such that —(CRD1RE1)(CRD2RE2)n-RF1 can be —CH2—RF1 or —CH2CH2—RF1. In some embodiments, at least one of RD1 and RE1 can be hydrogen; and the other of RD1 and RE1 can be a non-hydrogen moiety as described herein. For example, one of RD1 and RE1 can be hydrogen; and the other of RD1 and RE1 can be halogen, or one of RD1 and RE1 can be hydrogen; the other of RD1 and RE1 can be hydroxy; and one of RD1 and RE1 can be hydrogen; the other of RD1 and RE1 can be an unsubstituted C1-3 alkyl. In other embodiments, RD1 and RE1 can be each halogen, for example, fluoro. In some embodiments, at least one of RD2 and RE2 can be hydrogen; and the other of RD2 and RE2 can be a non-hydrogen moiety as described herein. For example, one of RD2 and RE2 can be hydrogen; and the other of RD2 and RE2 can be halogen, or one of RD2 and RE2 can be hydrogen; the other of RD2 and RE2 can be hydroxy; and one of RD2 and RE2 can be hydrogen; the other of RD2 and RE2 can be an unsubstituted C1-3 alkyl. In other embodiments, RD2 and RE2 can be each halogen, for example, fluoro.

As described herein the substituents, RD1, RE1, RD2 and RE2 can vary. In some embodiments, R4A can be —(CRD1RE1)(CRD2RE2)n-RF1, wherein two of RD1, RE1, RD2 and RE2 can be taken together to form an unsubstituted cyclopropyl or a double bond. Examples of when two of RD1, RE1, RD2 and RE2 can be taken together to form an unsubstituted cyclopropyl include the embodiments described in this paragraph. In some embodiments, RD1 and RE1 can be taken together with the carbon to which RD1 and RE1 are attached to form an unsubstituted cyclopropyl ring; and RD2 and RE2 can be independently selected from hydrogen, halogen, hydroxy and an unsubstituted C1-3 alkyl; and RF1 can be an unsubstituted or a substituted aryl, an unsubstituted or a substituted heteroaryl or an unsubstituted or a substituted heterocyclyl. In other embodiments, RD1 and RE2 can be independently selected from hydrogen, halogen, hydroxy and an unsubstituted C1-3 alkyl; RE1 and RD2 can be taken together with the carbon to which RE1 and RD2 are attached to form an unsubstituted cyclopropyl ring; and RF1 can be an unsubstituted or a substituted aryl, an unsubstituted or a substituted heteroaryl or an unsubstituted or a substituted heterocyclyl.

In some embodiments, RD1 and RE2 can be independently selected from hydrogen, halogen, hydroxy and an unsubstituted C1-3 alkyl; RE1 and RD2 together form a double bond; and RF1 can be an unsubstituted or a substituted aryl, an unsubstituted or a substituted heteroaryl or an unsubstituted or a substituted heterocyclyl. Examples of R4A include, but are not limited to, —CH2—RF1, —CH2CH2—RF1, —CF2—RF1, —CH(OH)—RF1,

As described herein, RF1 can be various ring structures. In some embodiments, RF1 can be an unsubstituted aryl. In other embodiments, RF1 can be a substituted aryl. When the aryl is monocyclic, RF1 can be an unsubstituted or a substituted phenyl. Multicyclic aryl groups can also be present at RF1, such as naphthyl and anthracenyl. In some embodiments, RF1 can be an unsubstituted heteroaryl. In other embodiments, RF1 can be a substituted heteroaryl. The heteroaryl for RF1 can be also monocyclic (such as a 5- or 6-membered monocyclic) or multicyclic (for example, bicyclic). In some embodiments, RF1 can be 9- or 10-membered bicyclic heteroaryl. Examples of suitable heteroaryls for RF1 include quinolinyl and imidazo[1,2-a]pyridinyl. In still other embodiments, RF1 can be an unsubstituted heterocyclyl. In yet still other embodiments, RF1 can be a substituted heterocyclyl. The heterocyclyls for RF1 can be monocyclic or multicyclic. For example, RF1 can be a bicyclic heterocyclyl, such as a 9- or 10-membered bicyclic heterocyclyl. Exemplary further RF1 groups include quinazoline, quinazolin-4-one, quinoxaline, isoquinoline, cinnoline, naphthyridine, benzimidazole and benzothiazole.

In other embodiments, R4A can be —(CRG1RH1)—O—RJ1. As described herein, RG1 and RH1 can be independently hydrogen, halogen or hydroxy. In some embodiments, RG1 and RH1 can be each hydrogen, such that R4A can be —CH2—O—RJ1. In some embodiments, at least one of RG1 and RH1 can be halogen, such as fluoro; and the other of RG1 and RH1 can be hydrogen. In other embodiments, RG1 and RH1 can be each halogen. As example of when RG1 and RH1 are each halogen is —CF2—O—RJ1. In some embodiments, at least one of RG1 and RH1 can be hydroxy. In some embodiments, at least one of RG1 and RH1 can be hydrogen. When at least one of RG1 and RH1 can be hydrogen, —(CRG1RH1)—O—RJ1 can be —CH(CH3)—O—RJ1.

As with RF1, RJ1 can be various cyclic moieties. In some embodiments, RJ1 can be an unsubstituted aryl, such as an unsubstituted phenyl or an unsubstituted naphthyl. In other embodiments, RJ1 can be a substituted aryl, for example, a substituted phenyl or a substituted naphthyl. In some embodiments, RJ1 is an unsubstituted heteroaryl. In other embodiments, RJ1 is a substituted heteroaryl. In still other embodiments, RJ1 is an unsubstituted heterocyclyl. In yet still other embodiments, RJ1 is a substituted heterocyclyl. The heteroaryl and heterocyclyl for RJ1 can be monocyclic or bicyclic, for example, RJ1 can be a 5-membered monocyclic heteroaryl, 6-membered monocyclic heteroaryl, 9-membered bicyclic heteroaryl, 10-membered bicyclic heteroaryl, 5-membered monocyclic heterocyclyl, 6-membered monocyclic heterocyclyl, 9-membered bicyclic heterocyclyl or 10-membered bicyclic heterocyclyl. Examples of cyclic moieties that can be RJ1 include, but are not limited to, quinolinyl, imidazo[1,2-a]pyridinyl, quinazoline, quinazolin-4-one, quinoxaline, isoquinoline, cinnoline, naphthyridine, benzimidazole and benzothiazole.

In still other embodiments, R4A can be —O—(CRK1RL1)—RM1. In some embodiments, RK1 and RL1 can be each hydrogen, such that R4A can be —O—CH2—RM1. In some embodiments, at least one of RK1 and RL1 can be halogen, such as fluoro; and the other of RK1 and RL1 can be hydrogen. In other embodiments, RK1 and RH1 can be each halogen, for example, —O—CF2—RM1. In some embodiments, at least one of RK1 and RL1 can be hydroxy. In some embodiments, at least one of RK1 and RJ1 can be hydrogen. When at least one of RK1 and RL1 can be hydrogen, —O—(CRK1RL1)—RM1 can be —O—CH(CH3)—RM1.

In some embodiments, RM1 can be an unsubstituted aryl, such as an unsubstituted phenyl or an unsubstituted naphthyl. In other embodiments, RM1 can be a substituted aryl, for example, a substituted phenyl or a substituted naphthyl. In some embodiments, RM1 is an unsubstituted heteroaryl. In other embodiments, RM1 is a substituted heteroaryl. The heteroaryl can be a monocyclic heteroaryl (such as a 5- or 6-membered monocyclic heteroaryl) or a bicyclic heteroaryl (such as a 9- or 10-membered bicyclic heteroaryl). In still other embodiments, RM1 is an unsubstituted heterocyclyl. In yet still other embodiments, RM1 is a substituted heterocyclyl. As with the heteroaryl, the heterocyclyl can be a monocyclic heterocyclyl (such as a 5- or 6-membered monocyclic heterocyclyl) or a bicyclic heterocyclyl (such as a 9- or 10-membered bicyclic heterocyclyl). Examples of RM1 group include, but are not limited to, quinolinyl, imidazo[1,2-a]pyridinyl, quinazoline, quinazolin-4-one, quinoxaline, isoquinoline, cinnoline, naphthyridine, benzimidazole and benzothiazole.

In some embodiments, R4A can be —(CRN1RO1)p-RP1. As described herein, in some embodiments, p can be 3. In other embodiments, p can be 4. In some embodiments, each RN1 and each RO1 can be hydrogen. In some embodiments, at least one RN1 and/or at least one RO1 can be halogen, such as fluoro; and the remaining RN1's and RO1's can be hydrogen. In other embodiments, at least one RN1 and/or at least one RO1 can be hydroxy; and the remaining RN1's and RO1's can be hydrogen. In still other embodiments, at least one RN1 and/or at least one RO1 can be an unsubstituted C1-3 alkyl; and the remaining RN1's and RO1's can be hydrogen. As provided herein, RP1 can be an unsubstituted or a substituted heteroaryl. In some embodiments, RP1 can be an unsubstituted heteroaryl. In other embodiments, RP1 can be an unsubstituted heteroaryl. The heteroaryl for RP1 can be a monocyclic of a bicyclic heteroaryl. In some embodiments, RP1 can be an unsubstituted monocyclic heteroaryl, such as a nitrogen-containing an unsubstituted monocyclic heteroaryl. In other embodiments, RP1 can be a substituted monocyclic heteroaryl, for example, a nitrogen-containing a substituted monocyclic heteroaryl.

When substituted, RF1, RJ1, RM1 and RP1 can be substituted 1, 2, 3 or more than 3 times with a variety of groups. When more than one group is present, one or more of the groups can be the same. The groups on RF1, RJ1, RM1 and RP1, when substituted, can be different from each other. Examples of groups that can be present on a substituted RF1, RJ1 and/or RM1 include, but are not limited to, halogen (for example, F, Cl and Br), cyano, an unsubstituted C1-4 alkyl, an unsubstituted C1-4 haloalkyl (such as CH2F, CHF2, CF3, CH2Cl, CHCl2 and Cl3), an unsubstituted monocyclic C3-6 cycloalkyl, an optionally substituted C-carboxy, an optionally substituted N-amido, amino, a mono-substituted amine, a di-substituted amine, —NH—C(═O)-unsubstituted C1-8 alkyl, —NH—C(═O)—O-unsubstituted C1-8 alkyl, —NH—C(═O)-unsubstituted C3-6 cycloalkyl and —NH—C(═O)—O-unsubstituted C3-6 cycloalkyl. Further examples that can be present on a substituted RF1, RJ1, RM1 and/or RP1 include, but are not limited to, an unsubstituted C1-4 alkoxy, an unsubstituted or a substituted phenyl and an unsubstituted or a substituted monocyclic heteroaryl (such as an unsubstituted or a substituted 5- or 6-membered heteroaryl). Prodrugs of compounds of Formula (I) can be obtained by substituting RF1, RJ1, RM1 and/or RP1 with an appropriate group. As an example, when RF1, RJ1, RM1 and/or RP1 is substituted with —NH—C(═O)-unsubstituted C1-8 alkyl, —NH—C(═O)—O-unsubstituted C1-8 alkyl, —NH—C(═O)-unsubstituted C3-6 cycloalkyl and —NH—C(═O)—O-unsubstituted C3-6 cycloalkyl, that compound of Formula (I) can be considered a prodrug of a compound of Formula (I) where an NH2 group replaces the —NH—C(═O)-unsubstituted C1-8 alkyl, —NH—C(═O)—O-unsubstituted C1-8 alkyl, —NH—C(═O)-unsubstituted C3-6 cycloalkyl or NH—C(═O)—O-unsubstituted C3-6 cycloalkyl group. A specific example is Compound 20 as described herein can be considered a prodrug of Compound 12. The specific structure of each of Compound 12 and Compound 20 are provided herein.

Exemplary RF1, RJ1 and RM1 groups include, but are not limited to, the following:

A non-limiting list of RP1 groups include the following:

Various other groups can be present at the 4′-position of the 5-membered ring of Formula (I). In some embodiments, R4B can be hydrogen. In other embodiments, R4B can be halogen, such as F. In still other embodiments, R4B can be cyano. In yet still other embodiments, R4B can be azido. In some embodiments, R4B can be —C(═O)NH2. In other embodiments, R4B can be an unsubstituted C1-4 alkyl, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl and tert-butyl. In still other embodiments, R4B can be a substituted C1-4 alkyl substituted with 1 or more substituents independently selected from the halogen (such as F and/or Cl), OH, OCH3 and cyano. Examples of substituted C1-4 alkyl for R4B include —CH2F, —CHF2, —CF3, —CH2Cl, —CHCl2, —CCl3, —CH2OH and —CH2CN. In yet still other embodiments, R4B can be an unsubstituted C2-4 alkenyl. In some embodiments, R4B can be a substituted C2-4 alkenyl substituted independently with 1 or more halogens, for example, fluoro and/or chloro. In other embodiments, R4B can be an unsubstituted C2-4 alkynyl. In still other embodiments, R4B can be a substituted C2-4 alkynyl. Examples of C2-4 alkenyl and C2-4 alkynyl include, but are not limited to, ethenyl, propenyl (straight-chained and branched), butenyl (straight-chained and branched), ethynyl, propynyl (straight-chained and branched) and butynyl (straight-chained and branched). In some embodiments, R4B can be an unsubstituted C3-C4 cycloalkyl. In other embodiments, R4B can be a substituted C3-C4 cycloalkyl. For example, R4B can be an unsubstituted or a substituted cyclopropyl or an unsubstituted or a substituted cyclobutyl. Alternatively, the 4′-position can be substituted by taking R4B and R3B together with the carbon R4B and R3B are attached form an unsubstituted oxetane. In some embodiments, R4B can be halogen, cyano, azido, —C(═O)NH2, a substituted C1-4 alkyl substituted with OH, OCH3 or cyano, an unsubstituted or a substituted C3-4 alkenyl, an unsubstituted or a substituted C2-4 alkynyl or an unsubstituted or a substituted C3-C4 cycloalkyl.

As provided herein, the 2′-position and the 4′-position can be connected via various moieties. In some embodiments, the 2′-position and the 4′-position can be connected via a —(CH2)y-O-moiety, wherein y can be 1 or 2. In some embodiments, R2B and R4B can be connected via —(CH2)—O-. In other embodiments, R2B and R4B are connected via —CH2CH2—O—. In some embodiments, the 2′-position and the 4′-position can be connected via

In other embodiments, the 2′-position and the 4′-position can be connected via

In still other embodiments, the 2′-position and the 4′-position can be connected via

wherein RE3 can be hydrogen or an unsubstituted C1-7 alkyl, for example,

can be

In yet still other embodiments, the 2′-position and the 4′-position can be connected via

In any embodiments of this paragraph, Z1 can be O (oxygen).

The base, B1, can be an optionally substituted, N-linked, 9-membered heteroaryl, such as those described herein. In some embodiments, B1 can be an optionally substituted

such as an optionally substituted

and an optionally substituted

In some embodiments, X1 can be N (nitrogen). In other embodiments, X1 can be CRC1. In some embodiments, X2 can be N (nitrogen). In other embodiments, X2 can be CRC2. In some embodiments, X3 can be N (nitrogen). In other embodiments, X3 can be CRC3. In some embodiments, X4 can be N (nitrogen). In other embodiments, X4 can be CRC4. In some embodiments, RC1, RC2, RC3 and/or RC4 can be hydrogen. In some embodiments, RC1, RC2, RC3 and/or RC4 can be halogen. In some embodiments, RC2, RC3 and/or RC4 can be an unsubstituted C1-4 alkyl. In other embodiments, B1 can be

for example, B1 can be

wherein RC2 can be halogen (such as F, Cl or Br). In still other embodiments, B1 can be an optionally substituted

such as

In some embodiments, R1B can be hydrogen, such that B1 can be an optionally substituted

an optionally substituted

and an optionally substituted

In other embodiments, R1B can be hydroxy or an unsubstituted C1-4 alkoxy. In still other embodiments, R1B can be an unsubstituted C1-4 alkyl, for example an unsubstituted C1-4 alkyl described herein, or an unsubstituted C2-4 alkenyl. In yet still other embodiments, R1B can be an unsubstituted C3-C6 cycloalkyl. In some embodiments, R1B can be NRA1RA2, such that B1 can be an optionally substituted

an optionally substituted

an optionally substituted

an optionally substituted

and an optionally substituted

In some embodiments of this paragraph, RC1 can be an unsubstituted C1-4 alkyl. In other embodiments of this paragraph, RC1 can be hydrogen. In still other embodiments of this paragraph, RC1 can be halogen, for example, F, Cl or Br. In some embodiments, B1 can be an unsubstituted

In other embodiments, B1 can be a substituted

For example,

can be substituted with one or more substituents selected from halogen and an unsubstituted C1-4 alkyl.

In other embodiments, B1 can be an optionally substituted

In still other embodiments, B1 can be an optionally substituted

such as an optionally substituted

In some embodiments, RC4 can be hydrogen. In other embodiments, RC4 can be halogen. In still other embodiments, RC4 can be an unsubstituted C1-4 alkyl. In yet still other embodiments, B1 can be an optionally substituted

In some embodiments, when B1 is

then X5 can be N (nitrogen). In other embodiments, when B1 is

then X5 can be CRC5. In some embodiments, RC5 can be hydrogen. In other embodiments, RC5 can be halogen. In still other embodiments, RC5 can be an unsubstituted C1-4 alkyl.

In some embodiments, B1 can be an unsubstituted or a substituted

an unsubstituted or a substituted

or an unsubstituted or a substituted

In other embodiments, B1 can be an unsubstituted or a substituted

wherein X2 can be CRC2. In still other embodiments, B1 can be an unsubstituted or a substituted

wherein X3 can be N. In yet still other embodiments, B1 can be an unsubstituted or a substituted

wherein X1 can be CRC1; and RC1 can be hydroxy, an unsubstituted C2-4 alkenyl, an unsubstituted C1-4 alkoxy or NRA1RA2.

As described herein, R1C, R1D and/or R1E can be hydrogen, hydroxy, an unsubstituted C1-4 alkyl or NRA1RA2. In some embodiments, R1C can be hydrogen. In other embodiments, R1C can be hydroxy. In still other embodiments, R1C can be an unsubstituted C1-4 alkyl. In yet still other embodiments, R1C can be an unsubstituted C2-4 alkenyl. In some embodiments, R1C can be an unsubstituted C1-4 alkoxy. In other embodiments, R1C can be an unsubstituted C3-C6 cycloalkyl. In still other embodiments, R1C can be NRA1RA2. In some embodiments, R1D can be hydrogen. In other embodiments, R1D can be hydroxy. In still other embodiments, R1D can be an unsubstituted C1-4 alkyl. In yet still other embodiments, R1D can be an unsubstituted C2-4 alkenyl. In some embodiments, R1D can be an unsubstituted C1-4 alkoxy. In other embodiments, R1D can be an unsubstituted C3-C6 cycloalkyl. In still other embodiments, R1D can be NRA1RA2. In some embodiments, R1E can be hydrogen. In other embodiments, R1E can be hydroxy. In still other embodiments, R1E can be an unsubstituted C1-4 alkyl. In yet still other embodiments, R1E can be an unsubstituted C2-4 alkenyl. In some embodiments, R1E can be an unsubstituted C1-4 alkoxy. In other embodiments, R1E can be an unsubstituted C3-C6 cycloalkyl. In still other embodiments, R1E can be NRA1RA2.

When R1B, R1C, R1D and/or R1E are NRA1RA2, RA1 and RA2 can be independently selected from hydrogen, hydroxy, an unsubstituted C1-4 alkyl, an unsubstituted C1-4 alkoxy and —C(═O)RC6, wherein RC6 can be hydrogen, an unsubstituted C1-4 alkyl or an unsubstituted C3-4 monocyclic cycloalkyl. In some embodiments, when R1B, R1C, R1D and/or R1E is NRA1RA2 RA1 and RA2 can be each hydrogen. For example, B1 can be an optionally substituted

an optionally substituted

an optionally substituted

an optionally substituted

or an optionally substituted

In other embodiments, when R1B, R1C, R1D and/or R1E is NRA1RA2, one of RA1 and RA2 can be hydrogen, and the other of RA1 and RA2 can be hydroxy. In still other embodiments, when R1B, R1C, R1D and/or R1E is NRA1RA2, one of RA1 and RA2 can be hydrogen, and the other of RA1 and RA2 can be an unsubstituted C1-4 alkyl (for example, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl or tert-butyl). In yet still other embodiments, when R1B, R1C, R1D and/or R1E is NRA1RA2, one of RA1 and RA2 can be hydrogen, and the other of RA1 and RA2 can be an unsubstituted C1-4 alkoxy. In some embodiments, when R1B, R1C, R1D and/or R1E is NRA1RA2, one of RA1 and RA2 can be hydrogen, and the other of RA1 and RA2 can be —C(═O)RC6, wherein RC6 can be hydrogen, an unsubstituted C1-4 alkyl or an unsubstituted C3-4 monocyclic cycloalkyl. In some embodiments, the B1 groups described herein can be unsubstituted. In some embodiments, the B1 groups described herein can be substituted, for example, substituted one or more times with a variable selected from halogen and an unsubstituted C1-4 alkyl.

Provided herein are a variety of B1 groups, including the following:

Prodrugs of compounds of Formula (I) can be obtained by substituting B1 with an appropriate group. For example, when R1B, R1C, R1D and/or R1E is —NH—C(═O)RC6, a compound of Formula (I) with the aforementioned group at R1B, R1C, R1D and/or R1E can be a considered a prodrug of a compound of Formula (I) where R1B, R1C, R1D and/or R1E is NH2.

The 1′-position of the 5-membered ring of Formula (I) can be unsubstituted or substituted. In some embodiments, R1 can be hydrogen. In other embodiments, R1 can be an unsubstituted C1-4 alkyl, such as those described herein.

As provided herein, compounds of Formula (I), or a pharmaceutically acceptable salt thereof, can have various substituents attached to the 5-membered ring of Formula (I). For example, in some embodiments, R1, R2A and R3A can be each hydrogen; R2B and R3B can be each OH; Z1 can be CH2; R4B can be an unsubstituted C1-4 alkyl; B1 can be a substituted or an unsubstituted

wherein X1 can be N or CRC1; X2 can be N or CRC2; X3 can be N or CRC3; RC1, RC2 and RC3 can be independently hydrogen, halogen or an unsubstituted C1-4 alkyl; and R1B can be hydrogen or NH2; and R4A —(CRD1RE1)(CRD2RE2)n-RF1, wherein RD1, RE1, RD2 and RE2 can be independently selected from hydrogen, halogen, hydroxy and an unsubstituted C1-3 alkyl; n can be 1; and RF1 can be an unsubstituted or a substituted heteroaryl. In other embodiments, R1, R2A and R3A can be each hydrogen; R2B and R3B can be each OH; Z1 can be CH2; R4B can be an unsubstituted C1-4 alkyl; B1 can be a substituted or an unsubstituted

a substituted or an unsubstituted

or a substituted or an unsubstituted

wherein X4 can be N or CRC4; X5 can be N or CRC5; RC4 and RC5 can be independently hydrogen, halogen or an unsubstituted C1-4 alkyl; and R1C, R1D and R1E can be independently hydrogen or NH2, and R4A —(CRD1RE1)(CRD2RE2)n-RF1, wherein RD1, RE1, RD2 and RE2 can be independently selected from hydrogen, halogen, hydroxy and an unsubstituted C1-3 alkyl; n can be 1; and RF1 can be an unsubstituted or a substituted heteroaryl.

In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, can be any one of the following formulae:

or a pharmaceutically acceptable salt of any of the foregoing. In some embodiments of this paragraph, R4B can be halogen, such as F. In other embodiments of this paragraph, R4B can bean unsubstituted C1-4 alkyl, such as those described herein and including methyl. In still other embodiments of this paragraph, R4B can be hydrogen. In some embodiments of this paragraph, R4B and R5B can be together with the carbon R4B and R5B are attached form an unsubstituted cyclopropyl. In some embodiments of this paragraph, B1 can be

In other embodiments of this paragraph, B1 can be

In still other embodiments of this paragraph, B1 can be

In yet still other embodiments of this paragraph, B1 can be

In some embodiments or this paragraph, B1 can be

such as

wherein RC5 can be halogen or an unsubstituted C1-4 alkyl, or

In some embodiments of this paragraph, B1B can be

In some embodiments of this paragraph, B1 can be an unsubstituted

In other embodiments of this paragraph, B1 can be a substituted

such as those described herein. In some embodiments of this paragraph, B1 can be an unsubstituted

In some embodiments of this paragraph, R4A can be —(CRD1RE1)(CRD2RE2)n—RF1, for example, —CH2—RF1, —CF2—RF1 and —CH(OH)—RF1. In some embodiments of this paragraph, R4A can be —(CRG1RH1)—O—RJ1, such as —CH2—O—RJ1. In some embodiments of this paragraph, R4A can be —O—(CRK1RL1)—RM1, such as —O—CH2—RM1. In some embodiments of this paragraph, R4A can be —(CRN1RO1)p-RP1. In some embodiments of this paragraph, R1 can be hydrogen. In some embodiments of this paragraph, R2A can be hydrogen. In some embodiments of this paragraph, R3A can be hydrogen. In other embodiments of this paragraph, R3A can be an unsubstituted C1-4 alkyl. In some embodiments of this paragraph, RF1, RJ1 and/or RM1 can be an unsubstituted or a substituted heteroaryl. In some embodiments of this paragraph, RF1, RJ1 and/or RM1 can be a substituted heteroaryl. In some embodiments of this paragraph, RF1, RJ1 and/or RM1 can be an unsubstituted or a substituted heterocyclyl. In some embodiments of this paragraph, RF1, RJ1 and/or RM1 can be a substituted heterocyclyl. In some embodiments of this paragraph, RF1, RJ1 and/or RM1 can be selected from

In some embodiments, a compound of Formula (I) can have one of the following structures:

In some embodiments of this paragraph, B1 can be

such as

wherein RC5 can be halogen or an unsubstituted C1-4 alkyl, or

In some embodiments of this paragraph, B1 can be

for example,

In some embodiments of this paragraph, B1 can be

including

In some embodiments of this paragraph, B1 can be

In some embodiments of this paragraph, B1 can be an unsubstituted or a substituted

wherein X2 can be CRC2; an unsubstituted or a substituted

wherein X3 can be N; or an unsubstituted or a substituted

wherein X1 can be CRC1, and RC1 can be hydroxy, an unsubstituted C2-4 alkenyl, an unsubstituted C1-4 alkoxy or NRA1RA2.

Examples of compounds of Formula (I), or a pharmaceutically acceptable salt thereof, include the following:

or a pharmaceutically acceptable salt of any of the foregoing.

In some embodiments, RF1 cannot be an optionally substituted imidazo[1,2-a]pyridine, an optionally substituted 1H-benzo[d]imidazole, an optionally substituted benzo[d]thiazole, an optionally substituted 1H-pyrrolo[3,2-b]pyridine, an optionally substituted thieno[3,2-b]pyridine, an optionally substituted furo[3,2-b]pyridine, an optionally substituted 1H-pyrrolo[2,3-b]pyridine, an optionally substituted 1H-pyrazole, an optionally substituted pyrimidine, an optionally substituted 1,8a-dihydroimidazo[1,2-a]pyridin-2(3H)-one, an optionally substituted 3,4-dihydro-2H-pyrido[3,2-b][1,4]oxazine, an optionally substituted 2,3-dihydro-1H-pyrrolo[2,3-b]pyridine, an optionally substituted 1H-imidazole and/or an optionally substituted 1H-pyrrolo[2,3-c]pyridine. In some embodiments, when R1, R2A, R3A, R4B, R5A and R5B are each H; R2B and R3B are each OH; and R4A is —CH2—RF1 or —(CH2)2—RF1, then RF1 cannot be an optionally substituted imidazo[1,2-a]pyridine, an optionally substituted 1H-benzo[d]imidazole, an optionally substituted benzo[d]thiazole, an optionally substituted 1H-pyrrolo[3,2-b]pyridine, an optionally substituted thieno[3,2-b]pyridine, an optionally substituted furo[3,2-b]pyridine, an optionally substituted 1H-pyrrolo[2,3-b]pyridine, an optionally substituted 1H-pyrazole, an optionally substituted pyrimidine, an optionally substituted 1,8a-dihydroimidazo[1,2-a]pyridin-2(3H)-one, an optionally substituted 3,4-dihydro-2H-pyrido[3,2-b][1,4]oxazine, an optionally substituted 2,3-dihydro-1H-pyrrolo[2,3-b]pyridine, an optionally substituted 1H-imidazole and/or an optionally substituted 1H-pyrrolo[2,3-c]pyridine. In some embodiments, when R1, R2A, R3A, R4B, R5A and R5B are each H; R2B and R3B are each OH; and R4A is —CH2—O—RJ1, then RJ1 cannot be an optionally substituted imidazo[1,2-a]pyridine, an optionally substituted 1H-benzo[d]imidazole, an optionally substituted benzo[d]thiazole, an optionally substituted 1H-pyrrolo[3,2-b]pyridine, an optionally substituted thieno[3,2-b]pyridine, an optionally substituted furo[3,2-b]pyridine, an optionally substituted 1H-pyrrolo[2,3-b]pyridine, an optionally substituted 1H-pyrazole, an optionally substituted pyrimidine, an optionally substituted 1,8a-dihydroimidazo[1,2-a]pyridin-2(3H)-one, an optionally substituted 3,4-dihydro-2H-pyrido[3,2-b][1,4]oxazine, an optionally substituted 2,3-dihydro-1H-pyrrolo[2,3-b]pyridine, an optionally substituted 1H-imidazole and/or an optionally substituted 1H-pyrrolo[2,3-c]pyridine. In some embodiments, when B1 is

(such as

then RF1 cannot be an optionally substituted imidazo[1,2-a]pyridine, an optionally substituted 1H-benzo[d]imidazole, an optionally substituted benzo[d]thiazole, an optionally substituted 1H-pyrrolo[3,2-b]pyridine, an optionally substituted thieno[3,2-b]pyridine, an optionally substituted furo[3,2-b]pyridine, an optionally substituted 1H-pyrrolo[2,3-b]pyridine, an optionally substituted 1H-pyrazole, an optionally substituted pyrimidine, an optionally substituted 1,8a-dihydroimidazo[1,2-a]pyridin-2(3H)-one, an optionally substituted 3,4-dihydro-2H-pyrido[3,2-b][1,4]oxazine, an optionally substituted 2,3-dihydro-1H-pyrrolo[2,3-b]pyridine, an optionally substituted 1H-imidazole and/or an optionally substituted 1H-pyrrolo[2,3-c]pyridine. In some embodiments, when R1, R2A, R3A, R4B, R5A and R5B are each H; R2B and R3B are each OH; R4A is —CH2—RF1, —(CH2)2—RF1 or —CH2—O—RJ1; and B1 is

(such as

then RF1 and/or RJ1 cannot be an optionally substituted imidazo[1,2-a]pyridine, an optionally substituted 1H-benzo[d]imidazole, an optionally substituted benzo[d]thiazole, an optionally substituted 1H-pyrrolo[3,2-b]pyridine, an optionally substituted thieno[3,2-b]pyridine, an optionally substituted furo[3,2-b]pyridine, an optionally substituted 1H-pyrrolo[2,3-b]pyridine, an optionally substituted 1H-pyrazole, an optionally substituted pyrimidine, an optionally substituted 1,8a-dihydroimidazo[1,2-a]pyridin-2(3H)-one, an optionally substituted 3,4-dihydro-2H-pyrido[3,2-b][1,4]oxazine, an optionally substituted 2,3-dihydro-1H-pyrrolo[2,3-b]pyridine, an optionally substituted 1H-imidazole and/or an optionally substituted 1H-pyrrolo[2,3-c]pyridine.

In some embodiments, RF1 cannot be an optionally substituted quinoline. In some embodiments, RF1 cannot be an optionally substituted quinazoline. In some embodiments, RF1 cannot be an optionally substituted quinoxaline. In some embodiments, when R1, R2A, R3A, R4B, R5A and R5B are each H; R2B and R3B are each OH; and R4A is —(CH2)2—RF1, then RF1 cannot be an optionally substituted quinolone, an optionally substituted quinazoline and/or an optionally substituted quinoxaline. In some embodiments, when B1 is

(such as

then RF1 cannot be an optionally substituted quinolone, an optionally substituted quinazoline and/or an optionally substituted quinoxaline. In some embodiments, when R1, R2A, R3A, R4B, R5A and R5B are each H; R2B and R3B are each OH; R4A is —(CH2)2—RF1; and B1 is

(such as

then RF1 cannot be an optionally substituted quinoline, an optionally substituted quinazoline and/or an optionally substituted quinoxaline. In some embodiments, when R1, R2A, R3A, R4B, R5A and R5B are each H; R2B and R3B are each OH; R4A is —(CH2)2—RF1; and B1 is

(such as

then RF1 cannot be an optionally substituted quinolone, an optionally substituted quinazoline and/or an optionally substituted quinoxaline. In some embodiments, when R1, R2A, R3A, R4B, R5A and R5B are each H; R2B and R3B are each OH; R4A is —O—(CH2)—RM1; and B1 is

(such as

then RM1 cannot be an optionally substituted quinolone, an optionally substituted quinazoline and/or an optionally substituted quinoxaline. In some embodiments, when R1, R2A, R3A, R4B, R5A and R5B are each H; R2B and R3B are each OH; R4A is —CH2—RF1; and B1 is

(such as

then RF1 cannot be an optionally substituted quinolone, an optionally substituted quinazoline and/or an optionally substituted quinoxaline. In some embodiments, when R1, R2A, R3A, R4B, R5A and R5B are each H; R2B and R3B are each OH; R4A is —CH2—RF1; and B1 is

(such as

then RF1 cannot be an optionally substituted naphthalene. In some embodiments, when R1, R2A, R3A, R4B, R5A and R5B are each H; R2B and R3B are each OH; R4A is —CH2—O—RJ1; and B1 is

(such as

then RJ1 cannot be an optionally substituted quinolone, an optionally substituted quinazoline and/or an optionally substituted quinoxaline.

In some embodiments, RF1 cannot be an optionally substituted phenyl, an optionally substituted thiophene, an optionally substituted pyridine and/or an optionally substituted 1,2,3,4-tetrahydroisoquinoline. In some embodiments, when R1, R2A, R3A, R4B, R5A and R5B are each H; R2B and R3B are each OH; and R4A is —CH2—RF1, —CH(OH)—RF1, —CH(F)—RF1 or —CH(OH)—CH2—RF1, then RF1 cannot be an optionally substituted phenyl, an optionally substituted thiophene, an optionally substituted pyridine and/or an optionally substituted 1,2,3,4-tetrahydroisoquinoline. In some embodiments, when R1, R2A, R3A, R4B, R5A and R5B are each H; R2B and R3B are each OH; R4A is —CH2—RF1, —CH(OH)—RF1, —CH(F)—RF1 or —CH(OH)—CH2—RF1; and B1 is

(such as

(such as

(for example,

then RF1 cannot be an optionally substituted phenyl, an optionally substituted thiophene, an optionally substituted pyridine and/or an optionally substituted 1,2,3,4-tetrahydroisoquinoline. In some embodiments, R4A cannot be —CH(OH)—RF1.

In some embodiments, B1 cannot be an optionally substituted

In some embodiments, B1 cannot be an optionally substituted

In some embodiments, when R1, R2A, R3A, R4B, R5A and R5B are each H; R2B and R3B are each OH; R4A is —CH(OH)—RF1; RF1 is an optionally substituted phenyl; then B1 cannot be an optionally substituted

(such as

or an optionally substituted

In some embodiments, when R1, R2A, R3A, R4B, R5A and R5B are each H; R2B and R3B are each OH; R4A is —(CH2)2—RF1 or —CH2—O—RJ1; RF1 and/or RJ1 is an optionally substituted quinoline; then B1 cannot be an optionally substituted

(such as

or an optionally substituted

In some embodiments, B1 cannot be

In some embodiments, B1 cannot be one or more of the following:

In some embodiments, B1 cannot be one or more of the following:

In some embodiments, B1 cannot be one or more of the following:

In some embodiments, when R1, R2A, R3A, R4B, R5A and R5B are each H; R2B and R3B are each OH; R4A is —CH(OH)—RF1, —(CH2)1-2—RF1, —CH(F)—RF1, —CH(OH)—CH2—RF1, —CH2—O—RJ1 or —O—CH2—RM1; then RF1, RJ1 and/or RM1 cannot be an optionally substituted phenyl, an optionally substituted naphthalene, an optionally substituted pyridine, an optionally substituted 1,2,3,4-tetrahydroisoquinoline, an optionally substituted quinoline, an optionally substituted quinazoline, an optionally substituted quinoxaline and/or an optionally substituted imidazo[1,2-a]pyridine. In some embodiments, RF1, RJ1 and/or RM1 cannot be an optionally substituted phenyl, an optionally substituted naphthalene, an optionally substituted pyridine, an optionally substituted 1,2,3,4-tetrahydroisoquinoline, an optionally substituted quinoline, an optionally substituted quinazoline, an optionally substituted quinoxaline, an optionally substituted imidazo[1,2-a]pyridine, an optionally substituted 1H-benzo[d]imidazole, an optionally substituted benzo[d]thiazole, an optionally substituted 1H-pyrrolo[3,2-b]pyridine, an optionally substituted thieno[3,2-b]pyridine, an optionally substituted furo[3,2-b]pyridine, an optionally substituted 1H-pyrrolo[2,3-b]pyridine, an optionally substituted 1H-pyrazole, an optionally substituted pyrimidine, an optionally substituted 1,8a-dihydroimidazo[1,2-a]pyridin-2(3H)-one, an optionally substituted 3,4-dihydro-2H-pyrido[3,2-b][1,4]oxazine, an optionally substituted 2,3-dihydro-1H-pyrrolo[2,3-b]pyridine, an optionally substituted 1H-imidazole and/or an optionally substituted 1H-pyrrolo[2,3-c]pyridine. In some embodiments, RF1 cannot be

In some embodiments, RJ1 cannot be

In some embodiments, RJ1 cannot be

In some embodiments, RJ1 cannot be

In some embodiments, when Z1 is O, R4B is an unsubstituted C1-4 alkyl (such as methyl), then RJ1 cannot be

In some embodiments, when Z1 is CH2, R4B is an unsubstituted C1-4 alkyl (such as methyl), then RF1 cannot be

In some embodiments, when Z1 is S, R4B is an unsubstituted C1-4 alkyl (such as methyl), then RF1 cannot be

In some embodiments, when R1, R2A, R3A and R5A are each H; R4B and R5B together with the carbon R4B and R5B are attached form an unsubstituted cyclopropyl; R2B and R3B are each OH; and R4A is —CH2CH2—RF1, —CH2CH(CH3)—RF1, —CH(CH3)CH2—RF1 or —CH2O—RJ1; and B1 is

then RF1 cannot be

In some embodiments, when R1, R2A, R3A and R5A are each H; and R2B and R3B are each OH; then R4B and R5B together with the carbon R4B and R5B are attached form an unsubstituted cyclopropyl. In some embodiments, when R1, R2A, R3A and R5A are each H; R4B and R5B together with the carbon R4B and R5B are attached form an unsubstituted cyclopropyl; R2B and R3B are each OH; and R4A is —CH2CH2—RF1, —CH2CH(CH3)—RF1, —CH(CH3)CH2—RF1 or —CH2O—RJ1; and B1 is

then RF1 cannot be an optionally substituted heteroaryl. In some embodiments, R4B and R5B cannot be together with the carbon R4B and R5B are attached form an unsubstituted cyclopropyl.

In some embodiments, R4A cannot be —(CRD1RE1)(CRD1RE1)n—RF1. In some embodiments, R4A cannot be —CH2—RF1. In some embodiments, R4A cannot be —(CH2)2—RF1. In some embodiments, R4A cannot be —CH(OH)—RF1. In other embodiments, R4A cannot be (CRG1RH1)—O—RJ1. In some embodiments, R4A cannot be —CH2—O—RJ1. In still other embodiments, R4A cannot be —O—(CRK1RL1)—RM1. In some embodiments, R4A cannot be —O—CH2—RM1. In some embodiments, R4A cannot be —(CRN1RO1)p-RP1. In some embodiments, R1, R2A, R3A, R4B, R5A and R5B cannot be each hydrogen.

In some embodiments, RF1 cannot be an optionally substituted bicyclic heteroaryl. In other embodiments, RF1 cannot be an optionally substituted bicyclic heterocyclyl. In still other embodiments, RF1 cannot be an optionally substituted phenyl. In some embodiments, RJ1 cannot be an optionally substituted bicyclic heteroaryl. In other embodiments, RJ1 cannot be an optionally substituted bicyclic heterocyclyl. In still other embodiments, RJ1 cannot be an optionally substituted phenyl. In some embodiments, RM1 cannot be an optionally substituted bicyclic heteroaryl. In other embodiments, RM1 cannot be an optionally substituted bicyclic heterocyclyl. In still other embodiments, RM1 cannot be an optionally substituted phenyl. In some embodiments, RF1, RJ1 and/or RM1 cannot be an optionally substituted

an optionally substituted

an optionally substituted

an optionally substituted

an optionally substituted

an optionally substituted

an optionally substituted phenyl, an optionally substituted pyridinyl (such as an optionally substituted

and/or an optionally substituted

an optionally substituted

an optionally substituted

and/or an optionally substituted

In some embodiments, R4B can be an unsubstituted C1-4 alkyl (such as methyl); and B1 can be an unsubstituted or a substituted

an unsubstituted or a substituted

or an unsubstituted or a substituted

In some embodiments, R4B can be an unsubstituted C1-4 alkyl (such as methyl); and B1 can be an unsubstituted or a substituted

In some embodiments, R4B can be an unsubstituted C1-4 alkyl (such as methyl); and B1 can be an unsubstituted or a substituted

wherein X2 can be CRC2. In some embodiments, R4B can be an unsubstituted C1-4 alkyl (such as methyl); and B1 can be an unsubstituted or a substituted

wherein X3 can be N, or an unsubstituted or a substituted

wherein X1 can be CRC1; and RC1 can be hydroxy, an unsubstituted C2-4 alkenyl, an unsubstituted C1-4 alkoxy or NRA1RA2.

In some embodiments, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, cannot be a compound as provided in WO 2018/065354, WO 2018/154104, WO 2018/152548, WO 2018/160824, WO 2017/212385, WO 2017/032840, WO 2019/116302, WO 2020/033282, WO 2020/033285 and/or WO 2020/033288.

Synthesis

Compounds of Formula (I) along with those described herein may be prepared in various ways. General synthetic routes for preparing compounds of Formula (I) are shown and described herein along with some examples of starting materials used to synthesize compounds described herein. The routes shown and described herein are illustrative only and are not intended, nor are they to be construed, to limit the scope of the claims in any manner whatsoever. Those skilled in the art will be able to recognize modifications of the disclosed syntheses and to devise alternate routes based on the disclosures herein; all such modifications and alternate routes are within the scope of the claims.

Deprotection of the acetonide of General Formula II can be performed in the presence of a suitable acid, for example HCl in MeOH, at a suitable temperature (such as room temperature), resulting in the formation of the compound of General Formula III. Optionally, compound of General Formula III can be converted to its corresponding ester of General Formula IV, by reaction with a suitable C1-4 alkyl acid anhydride or C1-4 alkyl acid chloride.

As depicted in Scheme 2, introduction of an aryl group (Ar) or in extension, if applicable, RF1, on the vinyl side chain on a compound of General Formula V, is performed by a reaction of the double bond with 9-BBN under an inert atmosphere, for example in a suitable solvent under appropriate conditions. An example of a suitable solvent and temperature is THF, at 50° C. The carbon-carbon bond can then be formed with a suitable Ar—Br or Ar—I (or in extension, if applicable RF1—Br or RF1—I) using a suitable catalyst (such as Pd(dppf)Cl2) in the presence of a base (for example, K3PO4), resulting in the formation of a compound of General Formula VI. In the context of the generic synthesis scheme, functional group conversions can be performed on the compound of General Formula VI, for example conversion of R4B from —CH2—OH via —C(═O)—H to —CN (as described in example 40), —(C═O)NH2 (as described in example 40), vinyl (as described in example 46) or alkyne (as described in example 47).

As depicted in scheme 3, Mitsunobu reaction of a compound of General Formula VII with ArOH (or in extension, if applicable, RJ1OH), results in the formation of a compound of General Formula VIII (where in extension ArO—, can be RJ1O—), for example, by using PPh3 and DIAD in a solvent like THF, or cyanomethylenetributylphosphorane (CMBP) in a suitable solvent (for example, toluene).

A compound of General Formula VII can be oxidized to the corresponding aldehyde of General Formula IX, followed by addition of an organometallic reagent like ArylMg(halide), or in extension, if applicable, RF1—Mg(halide), resulting in the formation of a compound of General Formula X (wherein, in extension Ar— can be RF1—). Alternatively, a compound of General Formula IX can be formed by oxidation of the vinyl functionality of compound of General Formula V, for example, by dihydroxylation with OsO4, followed by oxidation with NaIO4. A compound of General Formula V can be formed by a Wittig reaction of the aldehyde of General Formula IX.

As known to those skilled in the art, the compounds of Scheme 1 to 3 can be suitably protected when required. Compounds of General Formulae VII and V can be commercially available or can be obtained by methods known to those skilled in the art. An example of a compound of General Formula VII is

An example of a compound of General Formula V is

Another example of General Formulae VII and V are

respectively, for which the synthesis is described in example 31.

Introduction of a nucleobase described herein (denoted as B1) can be performed as exemplified in Scheme 5a and 5b, either using Mitsunobu-like conditions, for example using DIAD and PPh3, in THF at room temperature, and then converting the compound of General Formula XIa or XIb to a compound of General Formula XIIa or XIIb, respectively. Alternatively, General Formula XIa or XIb can be converted to a triflate of General Formula XIIIa or XIIIb, respectively. Following substitution, a compound of General Formula XIIa or XIIb, respectively, can be obtained. An example of a compound of General Formula XIB is:

An example of a compound of General Formulae XIa and XIIa, within the context of the generic synthesis scheme, are

respectively, prepared as described in example 36. Trt or Trityl is a protecting group that can be removed in the course of the synthetic route. Another example of compounds of General Formulae XIa and XIIa are

respectively, prepared as described in example 53.

As shown in Scheme 6, when B1 is connect to the rest of the scaffold via a nitrogen, amines of General Formulae XIV and XVI (PG represents a protection group) can be converted to compounds of General Formulae XV and XVII, respectively, using methods know to those skilled in the art. Amines of General Formulae XIV and XVI can be obtained utilizing methods known to those skilled in the art. An example of a compound of General Formula XVI is

Where desired, functional group transformations can be performed on the compounds of general formulae depicted in Scheme 1 to 6, containing a B1 group. For example, conversion of the R1B, substituent from chloro to NH2, via displacement with ammonia, such as described in example 1 for conversion of 8A to 9A. Or for example, conversion of the R1B, substituent from chloro to NH2, by palladium catalyzed coupling, such as with diphenylmethanimine, followed by removal of the protecting group as exemplified in example 17. Another example can be the conversion of R113 from chloride to methyl, as exemplified in example 28.

As exemplified in Scheme 7, a compound of the General Formula XVIII can be converted to a compound of General Formula XIX, using methods similar to those described for the conversion of a compound of General Formula V to a compound of General Formula VI. Oxidation of the alcohol to the ketone, for example, using IBX (2-Iodoxybenzoic acid) in acetonitrile at a temperature of 60° C., can provide a compound of General Formula XX. Functional group modification on R5B/R5A, or introduction of R5B/R5A can be performed utilizing a compound of General Formula XX. For example if R5B and R5A are each hydrogen, introduction of an exocyclic vinyl can be performed by using an Eschenmoser's salt, followed by amine methylation under the influence of MeI and subsequent elimination. The formed ketone of General Formula XX can be, after functional group modification of R5B and/or R5A, reduced back to the alcohol of General Formula XIX. An example of such a ketone of General Formula XX, formed by functional group modification at the stage of a compound of General Formula XX, is:

As described in Scheme 8, addition of an alkyl group as described herein to the 4′-position of the 5-membered ring of a compound of Formula (I) can be accomplished with an enone (3aR,6aR)-2,2-dimethyl-3a,6a-dihydrocyclopenta[d][1,3]dioxol-4-one. For example, using a cupper reagent, made from alkyl lithium in the presence of CuI in THF at 0° C., followed by addition to the enone at −78° C., can result in the formation of an intermediate of General Formula Int-II. Oxidation of the intermediate of General Formula Int-II to General Formula Int-III can be performed by forming the TES-enol, followed by oxidation in the presence of Pd(OAc)2 and oxygen in DMSO at a suitable temperature (such as 60° C.). Stereoselective addition of a vinyl group to the enone of General Formula Int-III, can be performed, for example, by treating a mixture of LiCl and CuI in THF with a mixture of TMSCl and General Formula Int-III, followed by addition of vinylmagnesium bromide at 0° C. This can be followed by the deprotection of any formed silyl enolate. Treatment with an acid, like HCl, in acetone/MeOH at a suitable temperature (for example, room temperature) can provide a compound of General Formula Int-IV. The ketone can be reduced to the alcohol of General Formula Int-IV, for example, by treatment with NaBH4 in MeOH at 0° C.

Scheme 9 describes a generic synthesis of the compounds which has B1 connected to the five-membered ring via a carbon-carbon bond. A compound of General Formula XXII can be formed by addition of an organometallic reagent to the ketone of General Formula XXI. An example of such organometallic reagent can be generated from reacting

with i-PrMgCl.LiCl, as described in example 51. Another example of such organometallic reagent, can be prepared by reaction of

with i-PrMgCl.LiCl, as described in example 43. Further transformations involve introduction of —Ar from General Formula XXII to General Formula XXIII, similar as described for the conversion of a compound of General Formula V to a compound of General Formula VI. Elimination of the —OH of General Formula XXIII to the alkene of General Formula XXIV, can be performed under acidic conditions, or, for example, by treating with DAST. The acetonide protecting group can be removed under acidic conditions, for example, by the treatment with aqueous HCl. Reduction of the double bond in General Formula XXIV, can be accomplished by hydrogenation using a heterogeneous catalyst like PtO2 in a suitable solvent (such as THF) under a hydrogen atmosphere. Alternatively, depending on the substituents on General Formula XXIV, Crabtree's catalyst can be used for the hydrogenation, for example, in MeOH under hydrogen atmosphere. In case diastereoisomers are obtained after the reduction, the desired isomer of General Formula XXV can be isolated out.

During the synthesis of compounds of Formula (I), such as those shown in Schemes 1-9, one or more moieties can be protected with one or more suitable protecting groups. Those skilled in the art know and can select the suitable protecting group(s) and the conditions to add and remove the suitable protecting group(s). The protecting group(s) may be chosen in such a way, that they are stable to certain reaction conditions and readily removed at a convenient stage using methodology known from the art.

Pharmaceutical Compositions

Some embodiments described herein relate to a pharmaceutical composition, that can include an effective amount of a compound described herein (e.g., a compound, or a pharmaceutically acceptable salt thereof, as described herein) and a pharmaceutically acceptable carrier, excipient or combination thereof. A pharmaceutical composition described herein is suitable for human and/or veterinary applications.

As used herein, a “carrier” refers to a compound that facilitates the incorporation of a compound into cells or tissues. For example, without limitation, dimethyl sulfoxide (DMSO) is a commonly utilized carrier that facilitates the uptake of many organic compounds into cells or tissues of a subject.

As used herein, a “diluent” refers to an ingredient in a pharmaceutical composition that lacks pharmacological activity but may be pharmaceutically necessary or desirable. For example, a diluent may be used to increase the bulk of a potent drug whose mass is too small for manufacture and/or administration. It may also be a liquid for the dissolution of a drug to be administered by injection, ingestion or inhalation. A common form of diluent in the art is a buffered aqueous solution such as, without limitation, phosphate buffered saline that mimics the composition of human blood.

As used herein, an “excipient” refers to an inert substance that is added to a pharmaceutical composition to provide, without limitation, bulk, consistency, stability, binding ability, lubrication, disintegrating ability etc., to the composition. A “diluent” is a type of excipient.

Pharmaceutical compositions may be formulated in a variety forms, such as tablets, capsules or solutions for oral administration; suppositories for rectal or vaginal administration; sterile solutions or suspensions for injectable administration. 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.

Proper formulation is dependent upon the route of administration chosen. Techniques for formulation and administration of the compounds described herein are known to those skilled in the art. Multiple techniques of administering a compound exist in the art including, but not limited to, oral, rectal, topical, aerosol, injection and parenteral delivery, including intramuscular, subcutaneous, intravenous, intramedullary injections, intrathecal, direct intraventricular, intraperitoneal, intranasal and intraocular injections. Pharmaceutical compositions will generally be tailored to the specific intended route of administration.

One may also administer the compound in a local rather than systemic manner, for example, via injection of the compound directly into the infected area, often in a depot or sustained release formulation. Furthermore, one may administer the compound in a targeted drug delivery system, for example, in a liposome coated with a tissue-specific antibody. The liposomes may be targeted to and taken up selectively by the organ.

The pharmaceutical compositions disclosed herein may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or tableting processes. As described herein, compounds used in a pharmaceutical composition may be provided as salts with pharmaceutically compatible counterions.

Methods of Use

Some embodiments described herein relate to a method of treating a cancer that can include administering to a subject identified as suffering from a cancer an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition that includes an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof. Other embodiments described herein relate to using a compound described herein, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating a cancer. Still other embodiments described herein relate to the use of a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition that includes a compound described herein, or a pharmaceutically acceptable salt thereof, for treating a cancer. Examples of suitable cancers include lymphomas, leukemias, liver cancers, lung cancers, breast cancers and/or colorectal cancers.

Some embodiments described herein relate to a method of treating a liver cancer (for example, hepatocellular carcinoma (HCC)) that can include administering to a subject identified as suffering from the liver cancer an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition that includes an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof. Other embodiments described herein relate to using a compound described herein, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating a liver cancer (such as HCC). Still other embodiments described herein relate to the use of a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition that includes a compound described herein, or a pharmaceutically acceptable salt thereof, for treating a liver cancer (for example, HCC).

Some embodiments described herein relate to a method for inhibiting replication of a cancer cell that can include contacting the cancer cell or administering to a subject identified as suffering from HCC with an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition that includes of a compound described herein, or a pharmaceutically acceptable salt thereof. Other embodiments described herein relate to the use of an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition that includes of a compound described herein, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for inhibiting replication of a cancer cell. Still other embodiments described herein relate to an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition that includes of a compound described herein, or a pharmaceutically acceptable salt thereof, for inhibiting replication of a cancer cell.

Some embodiments described herein relate to a method for inhibiting cell proliferation, such as inhibiting cell proliferation of cancer cells, that can include administering to a subject identified as suffering from a disease wherein inhibiting cell proliferation is desirable with an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition that includes of a compound described herein, or a pharmaceutically acceptable salt thereof. Other embodiments described herein relate to the use of an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition that includes of a compound described herein, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for inhibiting cell proliferation, such as inhibiting cell proliferation of cancer cells. Still other embodiments described herein relate to an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition that includes of a compound described herein, or a pharmaceutically acceptable salt thereof, for inhibiting cell proliferation, such as inhibiting cell proliferation of cancer cells.

Some embodiments described herein relate to a method of modulating a PRMT5 enzyme that can include contacting a cell (for example, a cancer cell described herein) with an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition that includes an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof. Other embodiments described herein relate to using a compound described herein, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for modulating a PRMT5 enzyme. Still other embodiments described herein relate to the use of a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition that includes a compound described herein, or a pharmaceutically acceptable salt thereof, for modulating a PRMT5 enzyme.

Some embodiments described herein relate to a method of inhibiting the activity of a PRMT5 enzyme that can include contacting a cell (for example, a cancer cell described herein) with an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition that includes an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof. Other embodiments described herein relate to using a compound described herein, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for inhibiting the activity of a PRMT5 enzyme. Still other embodiments described herein relate to the use of a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition that includes a compound described herein, or a pharmaceutically acceptable salt thereof, for inhibiting the activity of a PRMT5 enzyme.

Some embodiments described herein relate to a method of inducing apoptosis of a cell (for example, a cancer cell described herein) that can include contacting the cell with an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition that includes an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof. Other embodiments described herein relate to using a compound described herein, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for inducing apoptosis of a cell, such as a cancer cell described herein. Still other embodiments described herein relate to the use of a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition that includes a compound described herein, or a pharmaceutically acceptable salt thereof, for inducing apoptosis of a cell, such as a cancer cell described herein.

Some embodiments described herein relate to a method of decreasing the viability of a cell (for example, a cancer cell described herein) that can include contacting the cell with an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition that includes an effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof. Other embodiments described herein relate to using a compound described herein, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for decreasing the viability of a cell, such as a cancer cell described herein. Still other embodiments described herein relate to the use of a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition that includes a compound described herein, or a pharmaceutically acceptable salt thereof, for decreasing the viability of a cell, such as a cancer cell described herein. Exemplary cancer cells include lymphoma cells, leukemia cells, liver cancer cells, lung cancer cells, breast cancer cells and/or colorectal cancer cells. In some embodiments, the cancer cell can be a liver cancer cell.

For treatment of liver cancer, a high liver to plasma ratio can be useful. Accordingly, compounds that with a high liver to plasma ratio are of interest. In some embodiments, a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) can have a liver to plasma ratio of >5. In some embodiments, a compound described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt thereof) can have a liver to plasma ratio of >10.

Compounds disclosed herein can be evaluated for efficacy and toxicity using known methods. A non-limiting list of potential advantages of compounds described herein (such as a compound of Formula (I), and pharmaceutically acceptable salts thereof) include improved stability, increased safety profile, increased efficacy, increased binding to the target, increased specificity for the target (for example, a cancer cell).

As used herein, the terms “treat,” “treating,” “treatment,” “therapeutic,” and “therapy” do not necessarily mean total cure or abolition of the disease or condition. Any alleviation of any undesired signs or symptoms of a disease or condition, to any extent can be considered treatment and/or therapy. Furthermore, treatment may include acts that may worsen the subject's overall feeling of well-being or appearance.

As used herein, a “subject” refers to an animal that is the object of treatment, observation or experiment. “Animal” includes cold- and warm-blooded vertebrates and invertebrates such as fish, shellfish, reptiles and, in particular, mammals. “Mammal” includes, without limitation, mice, rats, rabbits, guinea pigs, dogs, cats, sheep, goats, cows, horses, primates, such as monkeys, chimpanzees, and apes, and, in particular, humans. In some embodiments, the subject is human.

The term “effective amount” is used to indicate an amount of an active compound, or pharmaceutical agent, that elicits the biological or medicinal response indicated. For example, an effective amount of compound can be the amount needed to alleviate or ameliorate symptoms of disease or prolong the survival of the subject being treated This response may occur in a tissue, system, animal or human and includes alleviation of the signs or symptoms of the disease being treated. Determination of an effective amount is well within the capability of those skilled in the art, in view of the disclosure provided herein. The effective amount of the compounds disclosed herein required as a dose will depend on the route of administration, the type of animal, including human, being treated, and the physical characteristics of the specific animal under consideration. The dose can be tailored to achieve a desired effect, but will depend on such factors as weight, diet, concurrent medication and other factors which those skilled in the medical arts will recognize.

The dosage may range broadly, depending upon the desired effects and the therapeutic indication. Alternatively dosages may be based and calculated upon the surface area of the patient, as understood by those of skill in the art. Although the exact dosage will be determined on a drug-by-drug basis, in most cases, some generalizations regarding the dosage can be made. The daily dosage regimen for an adult human patient may be, for example, an oral dose of between 0.01 mg and 3000 mg of each active ingredient, preferably between 1 mg and 700 mg, e.g. 5 to 200 mg. The dosage may be a single one or a series of two or more given in the course of one or more days, as is needed by the subject.

In instances where human dosages for compounds have been established for at least some condition, those same dosages may be used, or dosages that are between about 0.1% and 500%, more preferably between about 25% and 250% of the established human dosage. Where no human dosage is established, as will be the case for newly-discovered pharmaceutical compositions, a suitable human dosage can be inferred from ED50 or ID50 values, or other appropriate values derived from in vitro or in vivo studies, as qualified by toxicity studies and efficacy studies in animals.

In cases of administration of a pharmaceutically acceptable salt, dosages may be calculated as the free base. As will be understood by those of skill in the art, in certain situations it may be necessary to administer the compounds disclosed herein in amounts that exceed, or even far exceed, the above-stated, preferred dosage range in order to effectively and aggressively treat particularly aggressive diseases or infections.

Dosage amount and interval may be adjusted individually to provide plasma levels of the active moiety which are sufficient to maintain the modulating effects, or minimal effective concentration (MEC). The MEC will vary for each compound but can be estimated from in vitro data. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. However, HPLC assays or bioassays can be used to determine plasma concentrations. Dosage intervals can also be determined using MEC value. Compositions should be administered using a regimen which maintains plasma levels above the MEC for 10-90% of the time, preferably between 30-90% and most preferably between 50-90%. In cases of local administration or selective uptake, the effective local concentration of the drug may not be related to plasma concentration.

It should be noted that the attending physician would know how to and when to terminate, interrupt, or adjust administration due to toxicity or organ dysfunctions. Conversely, the attending physician would also know to adjust treatment to higher levels if the clinical response were not adequate (precluding toxicity). The magnitude of an administrated dose in the management of the disorder of interest will vary with the severity of the condition to be treated and to the route of administration. The severity of the condition may, for example, be evaluated, in part, by standard prognostic evaluation methods. Further, the dose and perhaps dose frequency, will also vary according to the age, body weight, and response of the individual patient. A program comparable to that discussed above may be used in veterinary medicine.

Compounds disclosed herein can be evaluated for efficacy and toxicity using known methods. For example, the toxicology of a particular compound, or of a subset of the compounds, sharing certain chemical moieties, may be established by determining in vitro toxicity towards a cell line, such as a mammalian, including a human cell line. The results of such studies are often predictive of toxicity in animals, such as mammals, or more specifically, humans. Alternatively, the toxicity of particular compounds in an animal model, such as mice, rats, rabbits, or monkeys, may be determined using known methods. The efficacy of a particular compound may be established using several recognized methods, such as in vitro methods, animal models, or human clinical trials. When selecting a model to determine efficacy, the skilled artisan can be guided by the state of the art to choose an appropriate model, dose, route of administration and/or regime.

Combination Therapies

In some embodiments, a compound described herein, or a pharmaceutically acceptable salt thereof, can be used in combination with one or more additional agent(s) for treating and/or inhibiting replication HCC. Additional agents include, but are not limited to, a kinase inhibitor (such as Sorafenib, Lenvatinib and Apatinib), a checkpoint inhibitor/modulator (such as a PD1/PDL1 inhibitor, an anti-PD1 antibody, for example, Nivolumab, Keytruda® and cemiplimab, an anti-PDL1 antibody, such as atezolizumab, avelumab and durvalumab, and an anti-CTLA4 antibody, such as Tremelimumab and Ipilimumab) and an anti-VEGF antibody (such as Bevacizumab).

In some embodiments, a compound described herein, or a pharmaceutically acceptable salt thereof, can be administered with one or more additional agent(s) together in a single pharmaceutical composition. In some embodiments, a compound described herein, or a pharmaceutically acceptable salt thereof, can be administered with one or more additional agent(s) as two or more separate pharmaceutical compositions. Further, the order of administration of a compound described herein, or a pharmaceutically acceptable salt thereof, with one or more additional agent(s) can vary.

EXAMPLES Table of Abbreviations:

The following abbreviations may appear in the present disclosure:

Abbreviation Name Ac Acetate ACN Acetonitrile anhyd. Anhydrous aq. Aqueous BPO Benzoylperoxide Bu Butyl CAN Ceric ammonium nitrate conc. Concentrated DCM Dichloromethane DIPEA N,N-Diisopropylethylamine DMA N,N-dimethylacetamide DMF N,N-Dimethylformamide DMSO Dimethyl sulfoxide DPPA Diphenylphosphoryl azide Dppf 1,1′-Bis(diphenylphosphino)ferrocene EA = EtOAc Ethyl acetate ECF Ethyl chloroformate Et Ethyl FA Formic acid G Gram(s) H Hour(s) IBX 2-Iodoxybenzoic acid Me Methyl MeOH Methanol Min Minute(s) NBS N-Bromosuccinimide NIS N-Iodosuccinimide PE Petroleum ether rt Room temperature sat. Saturated TBAF Tetra-n-butylammonium fluoride TBSCL t-Butyldimethylsilyl chloride TFA Trifluoroacetic acid THF Tetrahydrofuran

Additional embodiments are disclosed in further detail in the following examples, which are not in any way intended to limit the scope of the claims.

Example 1 Compound 1

MeLi (1.6 M in Et2O, 57.50 mL, 2.3 eq.) was added dropwise to a mixture of CuI (9.14 g, 48.00 mmol, 1.2 eq.) in THF (10 mL) at 0° C. The mixture was stirred at 0° C. for 10 min, then a solution of (3aR,6aR)-2,2-dimethyl-3a,6a-dihydrocyclopenta[d][1,3]dioxol-4-one (1A) (6.17 g, 40 mmol, 1 eq.) in THF (10 mL) was added dropwise at −78° C. The mixture was stirred at −78° C. for 20 min. The reaction progress was monitored by TLC (PE:EA=5:1). Upon completion, the reaction was quenched by NH4Cl (sat. aq., 50 mL) and extracted with EA (2×50 mL). The combined organic layers were washed with brine (20 mL), dried over Na2SO4, and concentrated to give a residue. The residue was purified by silica gel chromatography (Petroleum ether:Ethyl acetate (PE:EA)=10:1) to afford 3aR,6S,6aR)-2,2,6-trimethyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-4-one (2A) (5.2 g, 30.55 mmol, 76% yield) as a colorless oil.

LiHMDS (1.0 M in THF, 49.35 mL, 1.5 eq.) was added dropwise to a mixture of 2A (5.6 g, 32.90 mmol, 1 eq.) and chlorotriethylsilane (9.92 g, 65.80 mmol, 11.19 mL, 2 eq.) in THF (60 mL) at −78° C. The mixture was stirred at −78° C. for 10 min. The reaction progress was monitored by TLC (PE:EA=10:1). Upon completion, the mixture was quenched by NH4Cl (sat. aq., 80 mL) and brine (50 mL), and extracted with MTBE (2×100 mL). The combined organic layers were washed with brine (80 mL), dried over Na2SO4, and concentrated to give a residue. The residue was purified by silica gel chromatography (PE:EA=50: 1) to afford [(3aR,6aR)-2,2,6-trimethyl-6,6a-dihydro-3aH-cyclopenta[d][1,3]dioxol-4-yl]oxy-triethyl-silane (3A) (7.9 g, 27.77 mmol, 84% yield) as a colorless oil.

A mixture of 3A (7.9 g, 27.77 mmol, 1 eq.) and Pd(OAc)2 (1.87 g, 8.33 mmol, 0.3 eq.) in DMSO (80 mL) was stirred at 60° C. under O2 atmosphere for 6 h. The reaction progress was monitored by TLC (PE:EA=10:1). Upon completion, the mixture was diluted with water (300 mL) and extracted with MBTE (2×100 mL). The combined organic layers were combined and washed with water (100 mL) and brine (80 mL), dried over anhydrous Na2SO4, and concentrated to give a residue. The residue was purified by silica gel chromatography (PE:EA=5:1) to afford (3aR, 6aR)-2,2,6-trimethyl-3a,6a-dihydrocyclopenta[d][1,3]dioxol-4-one (4A) (3.7 g, 22.00 mmol, 79% yield) as a colorless oil.

A mixture of LiCl (8.48 mg, 200.00 μmol, 4.10 μL, 0.2 eq.), CuI (19.04 mg, 100.00 μmol, 0.1 eq.) in THF (2 mL) was stirred at 0° C. for 10 min. Then a mixture of TMSCl (130.37 mg, 1.20 mmol, 152.30 μL, 1.2 eq.) and 4A (168.19 mg, 1 mmol, 1 eq.) in THF (1 mL) was added dropwise at 0° C., and string was continued at 0° C. for 20 min. Vinylmagnesium bromide (1 M in THF, 1.60 mL, 1.6 eq.) was added dropwise at 0° C., and the mixture was stirred at 0° C. for 30 min. The reaction progress was monitored by TLC (PE:EA=5:1). Upon completion, the mixture was quenched by NH4Cl (sat. aq., 10 mL) and extracted with EA (2×20 mL). The combined organic layers were washed with brine (20 mL), dried over anhydrous Na2SO4, and concentrated to give a mixture of 5A and silyl enolate. The mixture was dissolved in acetone (2 mL) and MeOH (2 mL), conc. HCl (0.05 mL) was added, and the mixture was stirred at room temperature (rt) for 10 min. TEA (1 mL) was added to the mixture to quench the reaction. The solvent was removed under reduced pressure to give a residue. The residue was purified by silica gel chromatography (PE/EA=10:1) to afford (3aR,4R,6aR)-2,2,4-trimethyl-4-vinyl-5,6a-dihydro-3aH-cyclopenta[d][1,3]dioxol-6-one (5A) (97 mg, 494.29 μmol, 49% yield) as a colorless oil.

NaBH4 (35.5 mg, 937.62 μmol, 2 eq.) was added to a mixture of 5A (92 mg, 468.81 μmol, 1 eq.) and THF (1 mL) at 0° C. The mixture was stirred at 0° C. for 0.5 h. The reaction progress was monitored by TLC (PE:EA=10:1). Upon completion, the mixture was quenched by acetone (0.5 mL) and then concentrated to give a residue. The residue was diluted with aqueous potassium sodium tartrate (sat. aq., 20 mL) and then extracted with EA (2×20 mL). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na2SO4, and concentrated to give a residue. The residue was purified by silica gel chromatography (PE:EA=20:1) to afford (3aR,4R,6S,6aS)-2,2,4-trimethyl-4-vinyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-6-ol (6A) (67 mg, 337.54 μmol, 72% yield) as a colorless oil.

Trifluoromethanesulfonic anhydride (423.21 mg, 1.50 mmol, 247.49 μL, 1.5 eq.) was added to a mixture of 6A (198.26 mg, 1 mmol, 1 eq.) and pyridine (316.40 mg, 4.00 mmol, 322.86 μL, 4 eq.) in DCM (5 mL) at 0° C. The mixture was stirred at 0° C. for 1 h. The reaction progress was monitored by TLC (PE:EA=10:1). The mixture was quenched by ice-water (10 mL) and extracted with DCM (2×15 mL). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na2SO4, and concentrated to give crude (7A) (330 mg) as a yellow oil, which was used for next step without further purification.

A solution of (4-chloropyrrolo[2,3-d]pyrimidin-7-yl)potassium (7-a) (191.47 mg, 999.03 μmol, 1 eq.) in DMF (1 mL) was added dropwise to a solution of crude 7A (330 mg) in DMF (3 mL) at 0° C. The mixture was stirred at rt for 36 h. The reaction progress was monitored by TLC (PE:EA=5:1). The mixture was diluted with water (20 mL) and extracted with EA (2×20 mL). The combined organic layers were washed with water (20 mL) and brine (30 mL), dried over anhydrous Na2SO4, and concentrated to give a residue. The residue was purified by silica gel chromatography (PE:EA=10:1) to afford 7-[(3aR,4R,6R,6aS)-2,2,4-trimethyl-4-vinyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-6-yl]-4-chloro-pyrrolo[2,3-d]pyrimidine (8A) (134 mg, 397.41 μmol, 40% yield) as a colorless gum.

A mixture of 8A (700 mg, 2.10 mmol, 1 eq.) and NH3.H2O (28.00 g, 199.74 mmol, 30.77 mL, 95.25 eq.) in dioxane (16 mL) was stirred at 100° C. for 60 h. The reaction progress was monitored by LCMS. Upon completion, the mixture was concentrated under reduced pressure to give a residue. The residue was purified by silica gel chromatography (PE:EA=6:1, 200 mL, DCM:MeOH=20:1, 200 mL) to give 7-[(3aR,4R,6R,6aS)-2,2,4-trimethyl-4-vinyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-6-yl]pyrrolo [2,3-d]pyrimidin-4-amine (9A) (490 mg, 1.56 mmol, 74% yield) as a yellow foam.

To a solution of 9A (314.38 mg, 1 mmol, 1 eq.) in THF (5 mL) was added 9-BBN dimer (532.44 mg, 2.20 mmol, 2.2 eq.). The mixture was stirred at 50° C. for 2 h and then cooled to 20° C. K3PO4 (1.06 g, 5.00 mmol, 5 eq.). To the mixture was added H2O (0.5 mL), and the mixture was stirred at rt for 0.5 h. 3-bromo-7-iodo-quinolin-2-amine (Qa) (488.55 mg, 1.40 mmol, 1.4 eq.) and Pd(dppf)Cl2 (73.17 mg, 100.00 μmol, 0.1 eq.) were added to the mixture. The mixture was stirred at 60° C. for 12 h. The reaction progress was monitored by LCMS. Upon completion, the mixture was diluted with brine (20 mL) and extracted with EA (3×30 mL). The combined organic layers were dried over anhydrous Na2SO4 and concentrated to give a residue. The residue was purified by silica gel chromatography (EA=100%, 200 mL; DCM:MeOH=20:1, 500 mL) to afford the crude product, which was further purified by prep-HPLC (column: Phenomenex Gemini-NX 150*30 mm*5 um; mobile phase: [water (0.04% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 42%-72%, 8 min) to afford 7-[(E)-2-[(3aR,4R,6R,6aS)-6-(4-aminopyrrolo[2,3-d]pyrimidin-7-yl)-2,2,4-trimethyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-4-yl]vinyl]-3-bromo-quinolin-2-amine (10A) (350 mg, 653.67 μmol, 65% yield) as a yellow foam.

A mixture of 10A (150 mg, 280.15 μmol, 1 eq.) in MeOH (15 mL) and HCl (4 M, 4 mL, 57.11 eq.) was stirred at rt for 12 h. The reaction progress was monitored by LCMS. Upon completion, the mixture was concentrated under reduced pressure to afford a residue. The residue was suspended in MeOH (4 mL) and neutralized by NH4OH (aq., 25%) to reach pH 8.0. The solid was dissolved first and then precipitated again. The resulting suspension was filtered and the collected solid was washed with water to afford (1S,2R,3R,5R)-3-[(E)-2-(2-amino-3-bromo-7-quinolyl)vinyl]-5-(4-aminopyrrolo[2,3-d]pyrimidin-7-yl)-3-methyl-cyclopentane-1,2-diol (1) (130 mg, 262.43 μmol, 94% yield) as an off-white solid. LCMS: (ESI): m/z calcd. for C23H26BrN6O2 497.13 [M+H]+, found 497.1. 1H NMR (400 MHz, DMSO-d6) δ: 8.32 (s, 1H), 8.03 (s, 1H), 7.58 (d, J=8.2 Hz, 1H), 7.33 (s, 1H), 7.27 (d, J=3.5 Hz, 1H), 7.12 (br d, J=8.4 Hz, 1H), 6.91 (br s, 2H), 6.56 (br s, 2H), 6.53 (d, J=3.3 Hz, 1H), 4.94-4.83 (m, 2H), 4.60 (d, J=5.5 Hz, 1H), 4.39 (q, J=6.4 Hz, 1H), 3.77 (t, J=5.7 Hz, 1H), 2.83-2.59 (m, 2H), 1.96-1.66 (m, 4H), 1.10 (s, 3H).

Example 2 Compound 2

To a solution of 1B (50 g, 333.04 mmol, 1 eq.) in acetone (500 mL) was added 2,2-dimethoxypropane (36.42 g, 349.70 mmol, 42.85 mL, 1.05 eq.) and TsOH.H2O (633.51 mg, 3.33 mmol, 0.01 eq.) in one portion at 25° C. under N2. The mixture was stirred at 25° C. for 12 h. The mixture was neutralized by solid NaHCO3 to reach PH 8.0 and then filtrated. The filtrate was concentrated to give a residue as brown oil. The residue was purified by column chromatography (SiO2, PE:EA=5:1 to 1:1) to give 2B (55 g, 274.72 mmol, 82.49% yield, 95% purity) as a brown oil.

To a solution of Ph3PMeBr (88.32 g, 247.25 mmol, 2.85 eq.) in THF (800 mL) was added t-BuOK (32.79 g, 277.61 mmol, 95% purity, 3.2 eq.) at 0° C. The mixture was stirred at 0° C. for 0.5 h and then at 20° C. for 1 h. A solution of 2B in THF (200 mL) was added dropwise to the mixture at 0° C. during a period of 0.5 h, and the mixture was stirred at 20° C. for an additional 12 h. Upon completion, the mixture was diluted with H2O (400 mL) and EA (500 mL). The mixture was extracted with EA (2×200 mL). The combined organic phase was washed with brine (100 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuum to give a residue. The residue was purified by column chromatography (SiO2, PE:EA=5:1 to 1:1) to give 3B (25 g, crude) as a brown oil.

To a mixture of 3B in DCM (30 mL) was added a solution of NaIO4 (3.41 g, 15.94 mmol, 883.20 μL, 1 eq.) in H2O (20 mL) at 25° C., and the mixture was stirred at 25° C. for 1 h. Upon completion, the mixture was diluted with DCM (60 mL) and water (50 mL). The aqueous phase was extracted with DCM (2×20 mL). The combined organic phase was washed with brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by column chromatography (SiO2, PE:EA=15:1 to 5:1) to give 4B (2 g, 12.81 mmol, 80.34% yield) as a brown oil.

To a mixture of 4B (1.2 g, 7.68 mmol, 1 eq.) in anhydrous DCM (8 mL) was added SnCl2 (174.83 mg, 922.02 μmol, 23.92 μL, 0.12 eq.) and a solution of ethyl 2-azidoacetate (1.09 g, 8.45 mmol, 1.19 mL, 1.1 eq.) in DCM (5 mL) at 0° C. The reaction was stirred at 25° C. for 1 h and then filtrated through a pad of Celite. The filtrate was concentrated to give a residue as a brown oil. The residue was purified by column chromatography (SiO2, PE:EA=50:1 to 15:1) to give 5B (1.2 g, 4.46 mmol, 58.02% yield, 90% purity) as a brown oil.

To a solution of 5B (1.2 g, 4.95 mmol, 1 eq.) and N-diazo-4-methyl-benzenesulfonamide (976.84 mg, 4.95 mmol, 1 eq.) in CH3CN (10 mL) was added TEA (1.00 g, 9.91 mmol, 1.38 mL, 2 eq.) dropwise at 0° C. The reaction was stirred at 0° C. for 0.5 h and then at 25° C. for another 1.5 h. The mixture was diluted with EA (20 mL) and water (10 mL). The organic layer was separated and concentrated under reduce pressure to give a residue as a brown oil. The residue was purified by column chromatography (SiO2, PE:EA=50:1 to 20:1) to give 6B (1 g, 3.35 mmol, 67.73% yield, 90% purity) as a brown oil.

To a solution of 6B (400.00 mg, 1.49 mmol, 1 eq.) in toluene (4 mL) was added CuI (14.20 mg, 74.55 μmol, 0.05 eq.) at 25° C. The reaction was stirred at 110° C. for 12 h and then diluted with EA (60 mL) and water (40 mL). The aqueous phase was extracted with EA (2×20 mL). The combined organic phase was washed with brine (15 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by column chromatography (SiO2, PE:EA=60:1 to 10:1) to give 7B (200 mg, 832.46 μmol, 27.92% yield) as a brown oil.

To a solution of 7B (0.22 g, 915.71 μmol, 1 eq.) in MeOH (6 mL) was added NaBH4 (34.64 mg, 915.71 μmol, 1 eq.) at 25° C. The reaction was stirred at 25° C. for 1 h. Acetone (1 mL) was added, and the mixture was stirred for 5 min. The reaction was concentrated in vacuum to give a residue. The residue was purified by column chromatography (SiO2, PE:EA=8:1 to 2:1) to give 8B (200 mg, 825.53 μmol, 90.15% yield) as a brown oil.

To a solution of 8B (0.2 g, 832.46 μmol, 1 eq.) in acetone (8 mL) was added TsOH.H2O (79.17 mg, 416.23 μmol, 0.5 eq.) at 25° C. The reaction was stirred at 80° C. for 8 h. TEA (0.5 mL) was added to the mixture. The mixture was diluted with EA (60 mL) and water (40 mL). The aqueous phase was extracted with EA (2×20 mL). The combined organic phase was washed with brine (15 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by column chromatography (SiO2, PE:EA=6:1 to 2:1) to give 9B (160 mg, crude) as a brown oil.

To a solution of triphenyl phosphine (2.60 g, 9.91 mmol, 2 eq.) and 4-chloro-7H-pyrrolo[2,3-d]pyrimidine (1.52 g, 9.91 mmol, 2 eq.) in THF (20 mL) was added diisopropylazodicarboxylate (2.00 g, 9.91 mmol, 1.93 mL, 2 eq.) at 20° C. The mixture was stirred at 20° C. for 0.5 h. The mixture was added to a solution of 9B (1.2 g, 4.95 mmol, 1 eq.) in THF (20 mL) dropwise at 20° C. The reaction was stirred at 20° C. for 2 h, and then concentrated in vacuum to give a residue. The residue was purified by column chromatography (SiO2, PE:EA=8:1 to 2:1) to give 10A (1.5 g, crude), which was then purified by prep-HPLC (Neutral condition) (column: Xtimate C18 150*25 mm*5 um; mobile phase: [water (10 mM NH4HCO3)-ACN]; B%: 63%-63%, 10 min) to give 10A (0.8 g, 2.08 mmol, 41.89% yield, 98% purity) as a brown solid.

To a solution of 10A (0.93 g, 2.46 mmol, 1 eq.) in DCM (20 mL) was added DIBAL-H (1 M in toluene, 6.15 mL, 2.5 eq.) at −78° C. The reaction was stirred at −78° C. for 1.5 h. The reaction was treated sequentially with H2O (1 mL) and NaOH (aq. 2M, 1mL) and stirred for 5 min. Additional H2O (1 mL) was added, and the mixture was stirred for another 5 min. The mixture was filtrated through a pad of Celite and concentrated in vacuum to give 11A (830 mg, crude) as a brown oil, which was used for the next step without further purification.

To a solution of 11B (100 mg, 297.81 μmol, 1 eq.) in CH3CN (3 mL) was added IBX (125.09 mg, 446.71 μmol, 1.5 eq.) at 25° C. The reaction was stirred at 80° C. for 1.5 h and then concentrated in vacuum to give 12B (100 mg, crude) as a brown oil. The residue was used for the next step without further purification.

To a solution of Ph3PMeBr (267.57 mg, 749.02 μmol, 2.5 eq.) in THF (2 mL) was added t-BuOK (84.05 mg, 749.02 μmol, 2.5 eq.) at 0° C. The reaction was stirred at 0° C. for 10 min, and then at 20° C. for 20 min. A solution of 12B (100 mg, 299.61 μmol, 1 eq.) in THF (1.5 mL) was added dropwise at 0° C., and the reaction was stirred at 20° C. for another 0.5 h. Upon completion, the reaction was diluted with H2O (20 mL) and EA (50 mL). The aqueous phase was extracted with EA (60 mL). The combined organic phase was washed with brine (10 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by column chromatography (SiO2, PE:EA=12:1 to 5:1) to give 13B (90 mg, 271.25 μmol, 90.54% yield, 100% purity) as a brown oil.

A mixture of 13B (220 mg, 663.06 μmol, 1 eq.) and NH3H2O (24.38 g, 194.75 mmol, 26.79 mL, 28% purity, 293.71 eq.) in dioxane (10 mL) was stirred at 100° C. for 72 h in a steel sealed tube. The mixture was concentrated in vacuum to give a residue as a brown oil. The residue was purified by column chromatography (SiO2, DCM:MeOH=200:1 to 100:2) to give 13B (350 mg, 1.09 mmol, 81.96% yield, 97% purity, from parallel 2 batches) as a brown oil.

To a solution of 14B (350 mg, 1.12 mmol, 1 eq.) in THF (12 mL) was added 9-BBN (solid, dimer, 542.35 mg, 2.24 mmol, 2 eq.) at 20° C. The mixture was degassed and then stirred at 50° C. for 2 h. The reaction was cooled to 20° C. and treated with a solution of K3PO4 (1.19 g, 5.60 mmol, 5 eq.) in H2O (1.2 mL). Stirring was continued for another 30 min. To the mixture were added 3-bromo-7-iodo-quinolin-2-amine (586.51 mg, 1.68 mmol, 1.5 eq.) and Pd(dppf)Cl2 (81.99 mg, 112.05 μmol, 0.1 eq.). The mixture was degassed, stirred at 60° C. for another 15 h, and diluted with EA (50 mL) and water (30 mL). The aqueous phase was extracted with EA (50 mL). The combined organic phase was washed with brine (30 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuum to give a residue as a brown oil. The residue was purified by column chromatography (SiO2, DCM:MeOH=200:1 to 20:1) to give 15B (360 mg, 611.84 μmol, 54.61% yield, 91% purity) as a brown solid. The product was then purified by reversed-phase HPLC (Neutral condition) (column: Xtimate C18 150*25 mm*5 um; mobile phase: [water (10 mM NH4HCO3)-ACN]; B%: 46%-76%, 11 min) to give 15B (350 mg, 647.14 μmol, 80.58% yield, 99% purity) as a white solid.

To a solution of 15B (350 mg, 653.67 μmol, 1 eq.) in THF (10 mL) was added HCl (aq., 4 M, 5 mL, 30.60 eq.) at 20° C. The reaction was stirred at 20° C. for 5 h and then concentrated in vacuum to give crude product (315 mg, HCl salt) as a brown oil. The crude product (280 mg) was purified by reversed-phase HPLC (column: Agela DuraShell 150 mm_25 mm_5 um; mobile phase: [water (0.05% HCl)-ACN]; B%: 0%-30%, 8 min) to give 2 (160 mg, 322.99 μmol, 61.35% yield) as a white solid. MS: (ESI): m/z calcd. for C23H24BrN6O2 495.11 [M+H]+, found 495.3. 1H NMR (400 MHz, CD3OD) δ: 8.79 (s, 1H), 8.27 (s, 1H), 7.81 (d, J=8.3 Hz, 1H), 7.65 (s, 1H), 7.51 (dd, J=1.4, 8.2Hz, 1H), 7.38 (d, J=3.8 Hz, 1H),6.94 (d, J=3.8 Hz, 1H), 5.05 (s, 1H),4.67 (d, J=6.8 Hz, 1H), 3.96 (d, J=6.8 Hz, 1H), 3.20-2.96 (m, 2H), 2.41-2.26 (m, 1H), 1.88 (ddd, J=6.3, 11.0, 13.9 Hz, 1H), 1.46-1.33 (m, 2H), 0.74-0.55 (m, 1H).

Example 2 Compounds 3 and 4

TMEDA (5.65 g, 48.65 mmol, 7.34 mL, 1.5 eq.) was added to a mixture of CuI (339.73 mg, 1.78 mmol, 0.055 eq.) in THF (125 mL) at 0° C. The mixture was stirred at 0° C. for 5 min, and then cooled to −78° C. A solution of vinylmagnesium bromide (1 M in THF, 48.65 mL, 1.5 eq.) was added, and the mixture was stirred at −78° C. for 20 min. TMSCl (4.23 g, 38.92 mmol, 4.94 mL, 1.2 eq.) was added, followed by a solution of 1C (5 g, 32.43 mmol, 1 eq.) in THF (35 mL). The mixture was stirred at −78° C. for 3 h. The reaction progress was monitored by TLC (PE:EA=5:1). Upon completion, the reaction was quenched by NH4Cl (sat., aq., 50 mL), and the mixture was extracted with EA (2×100 mL). The combined organic layers were washed with brine (2×100 mL), dried over Na2SO4, filtered and concentrated to give a residue. The residue was purified by column chromatography (SiO2, PE:EA=20:1 to 10:1) to afford 2C (3.8 g, 18.77 mmol, 57% yield) as a yellow oil.

NaBH4 (151.3 mg, 4.00 mmol, 2 eq.) was added to a mixture of 2C (364.4 mg, 2 mmol, 1 eq.) in MeOH (30 mL) at 0° C. The mixture was stirred at 0° C. for 0.5 h. The reaction progress was monitored by TLC (PE:EA=5:1). Upon completion, the mixture was quenched by acetone (5 mL) and then concentrated under reduced pressure to afford a residue. The residue was diluted with brine (50 mL) and then extracted with EA (2×50 mL). The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated to afford a crude product. The crude product was purified by silica gel chromatography (PE:EA=20:1) to afford 3C (260.0 mg, 1.41 mmol, 71% yield) as a colorless oil.

9-BBN dimer (181.5 mg, 750.00 μmol, 1.5 eq.) was added to a mixture of 3C (92.1 mg, 0.5 mmol, 1 eq.) in THF (5 mL). The mixture was stirred at 50° C. under Ar atmosphere for 1 h. The mixture was cooled to rt, and then a solution of K3PO4 (530.7 mg, 2.50 mmol, 5 eq.) in H2O (0.5 mL) was added. After stirring at rt for 0.5 h, 3-bromo-2-chloro-7-iodo-quinoline (Q2, 221.0 mg, 600.00 μmol, 1.2 eq.) and Pd(dppf)Cl2 (36.6 mg, 50.00 μmol, 0.1 eq.) were added. The flask was degassed for several times and then stirred at 50° C. under Ar atmosphere for 11.5 h. The reaction progress was monitored by TLC (PE:EA=5:1). Upon completion, the mixture was diluted with brine (10 mL) and extracted with EA (2×20 mL). The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated to afford a residue. The residue was purified by silica gel chromatography (PE:EA=3:1) to afford 4C (146 mg, 342.14 μmol 68% yield) as a yellow gum.

To a solution of 4C (0.2 g, 468.68 μmol 1 eq.) in ACN (4 mL) was added IBX (196.86 mg, 703.02 μmol 1.5 eq.) at 20° C., and the reaction was stirred at 60° C. for 2 h. The mixture was then cooled to 20° C. and filtered. The collected solid was washed with acetonitrile. The filtrate was concentrated under reduced pressure to afford a residue. The residue was purified by column chromatography (SiO2, PE:EA=10:1 to 2:1) to give 5C (0.17 g, 400.27 μmol, 85.40% yield) as a white solid.

To a solution of 5C (200 mg, 470.90 μmol, 1 eq.) in THF (4 mL) was added LDA (2 M in THF, 0.3 mL, 1.27 eq.) at −78° C. The reaction was stirred at −78° C. for 0.5 h, and then dimethyl(methylene)ammonium iodide (5a) (348.48 mg, 1.88 mmol, 4 eq.) was added. The mixture was stirred at −78° C. for 0.5 h and then warmed to rt and stirred for 11.5 h. Then CH3I (0.89 g, 6.27 mmol, 390.35 μL, 13.32 eq.) was added at rt, and the mixture was stirred for another 3 h. The reaction progress was monitored by TLC (PE:EA=3:1). Upon completion, the mixture was quenched by 10% NaHCO3 (aq., 5 mL) and stirred for 0.5 h. The mixture was extracted with EA (2×50 mL). The combined organic layers were washed with 10% NaHCO3 (aq., 20 mL), brine (20 mL), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by vacuum silica gel column chromatography (SiO2, PE:EA=15:1 to 5:1) to give 6C (120 mg, 219.82 μmol, 46.68% yield, 80% purity) as a white solid.

To a solution of 6C (854 mg, 1.96 mmol, 1 eq.) in MeOH (10 mL) and THF (10 mL) was added CeCl3.7 H2O (801.43 mg, 2.15 mmol, 204.45 μL, 1.1 eq.) at −78° C. The mixture was stirred at the same temperature for 10 min, then NaBH4 (81.38 mg, 2.15 mmol, 1.1 eq.) was added. The mixture was stirred at −78° C. and stirred for another 10 min. The mixture was warmed to 0° C. and stirred at 0° C. for 10 min. The reaction was quenched with NH4Cl (sat., aq., 5 mL), and the mixture was extracted with EA (2×50 mL). The combined organic layers were washed with brine (20 mL), dried with anhydrous Na2SO4, filtered and concentrated in vacuum to give a residue. The residue was purified by column chromatography (SiO2, PE:EA=8:1 to 3:1) to give 7C (654 mg, 894.38 μmol, 45.7% yield, 60% purity) as a brown solid.

To a solution of triphenyl phosphine (896.73 mg, 3.42 mmol, 2.5 eq.), 4-chloro-7H-pyrrolo[2,3-d]pyrimidine (315.02 mg, 2.05 mmol, 1.5 eq.), and 7C (600 mg, 1.37 mmol, 1 eq.) in THF (12 mL) was added DIAD (636.02 mg, 3.15 mmol, 611.56 μL, 2.3 eq.) at 20° C. After stirring at rt for 5 h, the mixture was concentrated in vacuum to give a residue. The residue was purified by column chromatography (SiO2, PE:EA=20:1 to 8:1) to give 8C (600 mg, 1.02 mmol, 74.8% yield, 98% purity) as a brown solid.

8C (500 mg, 870.63 μmol, 1 eq.) was dissolved in NH3.H2O (10 mL) and dioxane (10 mL) at 20° C. in an autoclave. The mixture was stirred at 140° C. for 48 h and then concentrated to give a brown residue. The residue was purified by column chromatography (SiO2, DCM:MeOH=40:1 to 10:1) to give 9C (70 mg, 14.49% yield) and 9C-a (280 mg, 449.73 μmol, 51.6% yield, 86% purity).

To a solution of 9C (60 mg, 108.13 μmol, 1 eq.) in THF (3 mL) was added HCl (4 M, 1.50 mL eq.) at 20° C. The mixture was stirred at 20° C. for 4 h and then concentrated. The residue was neutralized by NH3.H2O to reach pH 8 and then purified by prep-HPLC (basic condition, column: Phenomenex Gemini-NX 150*30 mm*5 um; mobile phase: [water (0.04% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 31%-71%, 8 min) to give 3 (35 mg, 67.72 μmol, 62.6% yield, 99.6% purity) as a white solid. LCMS: (ESI): m/z calcd. for C23H22BrClN5O2, 516.1 [M+H]+, found 516.1. 1H NMR (400 MHz, DMSO) δ: 8.92 (s, 1H), 8.03 (s, 1H), 7.98 (d, J=8.3 Hz, 1H), 7.88 (s, 1H), 7.68 (dd, J=1.5, 8.5 Hz, 1H), 7.12 (d, J=3.5 Hz, 1H), 6.95 (br s, 2H), 6.58 (d, J=3.5 Hz, 1H), 5.44 (br d, J=9.3 Hz, 1H), 5.03 (d, J=6.8 Hz, 1H), 4.98 (br s, 1H), 4.91 (d, J=3.5 Hz, 1H), 4.44-4.34 (m, 1H), 4.31 (br s, 1H), 3.96 (br s, 1H), 3.31-3.24 (m, 1H), 3.07-2.85 (m, 2H), 2.07-1.77 (m, 2H).

To a solution of 9C-a (80 mg, 149.41 μmol, 1 eq.) in THF (4.5 mL) was added HCl (4 M, 2.25 mL, 55.49 eq.) at 20° C. The mixture was stirred at 20° C. for 4 h and then concentrated. The residue was neutralized by NH3.H2O to reach pH ˜8 and then purified by prep-HPLC (basic condition, column: Phenomenex Gemini-NX 150*30 mm*5 um; mobile phase: [water (0.04% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 25%-55%, 8 min) to give 4 (45 mg, 89.48 μmol, 60% yield, 98.5% purity) as white solid. LCMS: (ESI): m/z calcd. for C23H24BrN6O2, 495.1, 497.1 [M+H]+, found 495.2,497.2. 41 NMR (400 MHz, CD3OD) δ: 8.28 (s, 1H), 8.06 (s, 1H), 7.59 (d, J=8.3 Hz, 1H), 7.45 (s, 1H), 7.24 (d, J=8.0 Hz, 1H), 7.09 (d, J=3.5 Hz, 1H), 6.64 (d, J=3.8 Hz, 1H), 5.57 (br d, J=9.3 Hz, 1H), 5.10 (br s, 1H), 4.54 (s, 1H), 4.44 (dd, J=4.9, 9.2 Hz, 1H), 4.08 (br d, J=2.8 Hz, 1H), 3.07-2.85 (m, 2H), 2.69 (br s, 1H), 2.15-1.84 (m, 2H).

Example 3 Compounds 5 and 6

TMEDA (5.65 g, 48.65 mmol, 7.34 mL, 1.5 eq.) was added to a mixture of CuI (339.73 mg, 1.78 mmol, 0.055 eq.) in THF (125 mL) at 0° C. The mixture was stirred at 0° C. for 5 min and then cooled to −78° C. A solution of vinylmagnesium bromide (1 M in THF, 48.65 mL, 1.5 eq.) was added, and the mixture was stirred at −78° C. for 20 min. TMSCl (4.23 g, 38.92 mmol, 4.94 mL, 1.2 eq.) was added, followed by a solution of 1D (5 g, 32.43 mmol, 1 eq.) in THF (35 mL). The mixture was stirred at −78° C. for 3 h. The reaction progress was monitored by TLC (PE:EA=5:1). Upon completion, the reaction was quenched by NH4Cl (sat., aq., 50 mL), and the mixture was extracted with EA (2×100 mL). The combined organic layers were washed with brine (2×100 mL), dried over Na2SO4, filtered and concentrated to give a residue. The residue was purified by column chromatography (SiO2, PE:EA=20:1 to 10:1) to afford 2D (3.8 g, 18.77 mmol, 57% yield) as a yellow oil.

NaBH4 (151.3 mg, 4.00 mmol, 2 eq.) was added to a mixture of 2D (364.4 mg, 2 mmol, 1 eq.) in MeOH (30 mL) at 0° C. The mixture was stirred at 0° C. for 0.5 h. The reaction progress was monitored by TLC (PE:EA=5:1). Upon completion, the mixture was quenched by acetone (5 mL) and then concentrated under reduced pressure to afford a residue. The residue was diluted with brine (50 mL) and then extracted with EA (2×50 mL). The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated to afford a crude product. The crude product was purified by silica gel chromatography (PE:EA=20:1) to afford 3D (260.0 mg, 1.41 mmol, 71% yield) as a colorless oil.

9-BBN dimer (181.5 mg, 750.00 μmol, 1.5 eq.) was added to a mixture of 3D (92.1 mg, 0.5 mmol, 1 eq.) in THF (5 mL). The mixture was stirred at 50° C. under Ar for 1 h. The mixture was cooled to rt, and then a solution of K3PO4 (530.7 mg, 2.50 mmol, 5 eq.) in H2O (0.5 mL) was added. After stirring at rt for 0.5 h, Q2 (221.0 mg, 600.00 μmol, 1.2 eq.) and Pd(dppf)Cl2 (36.6 mg, 50.00 μmol, 0.1 eq.) were added. The flask was degassed for several times and then stirred at 50° C. under Ar for 11.5 h. The reaction progress was monitored by TLC (PE:EA=5:1). Upon completion, the mixture was diluted with brine (10 mL) and extracted with EA (2×20 mL). The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated to afford a residue. The residue was purified by silica gel chromatography (PE:EA=3:1) to afford 4D (146 mg, 342.14 μmol, 68% yield) as a yellow gum.

To a solution of 4D (0.2 g, 468.68 μmol, 1 eq.) in ACN (4 mL) was added IBX (196.86 mg, 703.02 μmol, 1.5 eq.) at 20° C., and the mixture was stirred at 60° C. for 2 h. The mixture was then cooled to 20° C. and filtered. The collected solid was washed with ACN. The filtrate was concentrated under reduced pressure to afford a residue. The residue was purified by column chromatography (SiO2, PE:EA=10:1 to 2:1) to give 5D (0.17 g, 400.27 μmol, 85.40% yield) as a white solid.

To a solution of 5D (200 mg, 470.90 μmol, 1 eq.) in THF (4 mL) was added LDA (2 M in THF, 0.3 mL, 1.27 eq.) at −78° C. The mixture was stirred at −78° C. for 0.5 h, and then dimethyl(methylene)ammonium iodide (5a) (348.48 mg, 1.88 mmol, 4 eq.) was added. The mixture was stirred at −78° C. for 0.5 h. The mixture was then warmed to rt and stirred for 11.5 h. CH3I (0.89 g, 6.27 mmol, 390.35 μL, 13.32 eq.) was added at rt, and the mixture was stirred for another 3 h. The reaction progress was monitored by TLC (PE:EA=3:1). Upon completion, the reaction was quenched by 10% NaHCO3 (aq., 5 mL) and stirred for 0.5 h. The mixture was extracted with EA (2×50 mL). The combined organic layers were washed with 10% NaHCO3 (aq., 20 mL) and brine (20 mL), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by vacuum silica gel column chromatography (SiO2, PE:EA=15:1 to 5:1) to give 6D (120 mg, 219.82 μmol, 46.7% yield, 80% purity) as a white solid.

To a solution of 6D (854 mg, 1.96 mmol, 1 eq.) in MeOH (10 mL) and THF (10 mL) was added CeCl3.7H2O (801.43 mg, 2.15 mmol, 204.45 μL, 1.1 eq.) at −78° C. The mixture was stirred at the same temperature for 10 min. NaBH4 (81.38 mg, 2.15 mmol, 1.1 eq.) was added at −78° C., and the mixture was stirred for 10 min. The mixture was warmed to 0° C. and then stirred at 0° C. for 10 min. The reaction was quenched with NH4Cl (sat., aq., 5 mL), and the mixture was extracted with EA (2×50 mL). The combined organic layers were washed with brine (20 mL), dried with anhydrous Na2SO4, filtered and concentrated in vacuum to give a residue. The residue was purified by column chromatography (SiO2, PE:EA=8:1 to 3:1) to give 7D (654 mg, 894.38 μmol, 45.7% yield, 60% purity) as a brown solid.

To a solution of triphenyl phosphine (896.73 mg, 3.42 mmol, 2.5 eq.), 4-chloro-7H-pyrrolo[2,3-d]pyrimidine (315.02 mg, 2.05 mmol, 1.5 eq.), and 7D (600 mg, 1.37 mmol, 1 eq.) in THF (12 mL) was added DIAD (636.02 mg, 3.15 mmol, 611.56 μL, 2.3 eq.) at 20° C. After stirring at rt for 5 h, the mixture was concentrated in vacuum to give a residue. The residue was purified by column chromatography (SiO2, PE:EA=20:1 to 8:1) to give 8D (600 mg, 1.02 mmol, 74.8% yield, 98% purity) as a brown solid.

8D (500 mg, 870.63 μmol, 1 eq.) was dissolved in NH3H2O (10 mL) and dioxane (10 mL) at 20° C. in an autoclave. The mixture was stirred at 140° C. for 48 h and then concentrated to give a brown residue. The residue was purified by column chromatography (SiO2, DCM:MeOH=40:1 to 10:1) to give 9D (70 mg, 14.5% yield) and 9D-a (280 mg, 449.73 μmol, 51.6% yield, 86% purity).

To a solution of 9D-a (45 mg, 84.04 μmol) in EtOH (4 mL) was added PtO2 (3.82 mg, 16.81 μmol, 0.2 eq.) at rt. The mixture was degassed under vacuum and purged with H2 (15 psi) several times. The mixture was stirred under H2 (15psi) at rt for 6 h and then filtered over a pad of Celite. The filtrate was concentrated to give a residue. The residue was purified by column chromatography (SiO2, DCM:MeOH=20:1 to 7:1) to give a mixture of 10D-a and 10D-b (35 mg) as a white solid. The ratio of isomers was approximately 2:1 based on SFC analysis. The mixture was purified by SFC separation (column: DAICEL CHIRALCELOD (250 mm*30 mm, 10 um); mobile phase: [0.1% NH3H2O ETOH]; B%: 45%) to give 10D-a (retention time: 3.471 min) (20 mg, 43.62 μmol, 57.1% yield) as a white solid and 10D-b (retention time: 2.779 min) (14 mg, 30.53 μmol, 40.0% yield) as a white solid. 10D-a: 1H NMR (400 MHz, CD3OD) δ: 8.04 (s, 1H), 7.87 (d, J=9.0 Hz, 1H), 7.55 (d, J=8.1 Hz, 1H), 7.41 (s, 1H), 7.27-7.13 (m, 2H), 6.75 (d, J=8.8 Hz, 1H), 6.62 (d, J=3.4 Hz, 1H), 4.94-4.89 (m, 1H), 4.61-4.46 (m, 2H),3.14-2.92 (m, 1H), 2.83 (br dd, J=8.8, 14.2 Hz, 1H), 2.26 (dt, J=6.6,11.4 Hz, 1H), 2.09-1.97 (m, 2H),1.54 (s, 3H), 1.57-1.51 (m, 1H), 1.30 (s, 3H), 0.81 (br d, J=6.6 Hz, 3H). LCMS: (ESI): m/z calcd. for C26H31N6O2, 459.2 [M+H]+, found 459.2. 10D-b: 1H NMR (400 MHz, CD3OD) δ: 8.04 (s, 1H), 7.86 (d, J=8.8 Hz, 1H), 7.54 (d, J=8.3 Hz, 1H), 7.37 (s, 2H), 7.14 (dd, J=1.5, 8.1 Hz, 1H), 6.74 (d, J=8.8 Hz, 1H), 6.60 (d, J=3.7 Hz, 1H), 5.38 (t, J=7.5 Hz, 1H), 4.99 (t, J=6.7 Hz, 1H), 4.59 (t, J=7.1 Hz, 1H), 2.90-2.66 (m, 3H), 2.39-2.22 (m, 1H), 1.94-1.80 (m, 2H), 1.49 (s, 3H), 1.35 (s, 3H), 0.65 (d, J=7.6 Hz, 3H). LCMS: (ESI): m/z calcd. for C26H31N6O2, 459.2 [M+H]+, found 459.2.

To a solution of 10D-a (20 mg, 43.62 μmol, 1 eq.) in THF (3 mL) was added HCl (4 M, 1.5 mL in H2O) at rt. The mixture was stirred at rt for 12 h. The mixture was then concentrated in vacuum to give a residue. The mixture was neutralized with NH3H2O to pH 9. The residue was purified by prep-HPLC (basic condition; column: Phenomenex Gemini-NX 150*30 mm*5 um; mobile phase: [water (0.04% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 20%-50%, 8 min) to give 5 (12 mg, 28.53 μmol, 65.4% yield, 99.5% purity) as a white solid. 1H NMR (400 MHz, CD3OD) δ: 8.04 (s, 1H), 7.88 (d, J=8.8 Hz, 1H), 7.56 (d, J=8.0 Hz, 1H), 7.40 (s, 1H), 7.27-7.13 (m, 2H), 6.75 (d, J=9.0 Hz, 1H), 6.61 (d, J=3.5 Hz, 1H), 4.58-4.45 (m, 1H), 4.36 (dd, J=6.3, 9.0 Hz, 1H), 3.97 (dd, J=3.9, 6.1 Hz, 1H), 3.04-2.77 (m, 2H), 2.06-1.77 (m, 3H), 1.71-1.60 (m, 1H), 0.93 (d, J=6.5 Hz, 3H). LCMS: (ESI): m/z calcd. for C23H27N6O2, 419.5 [M+H]+, found 419.2.

To a solution of 10D-b (14 mg, 30.53 μmol, 1 eq.) in THF (3 mL) was added HCl (4 M, 1.5 mL in water) at 20° C. The mixture was stirred at 20° C. for 12 h. The mixture was concentrated in vacuum to give a residue. The residue was neutralized with NH3.H2O to pH 9. The residue was purified by prep-HPLC (basic condition; column: Phenomenex Gemini-NX 150*30 mm*5 um; mobile phase: [water (0.04% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 20%-50%, 8 min) to give 6 (7 mg, 16.54 μmol, 54.17% yield, 98.88% purity) as a white solid. 1H NMR (400 MHz, CD3OD) δ: 8.06 (s, 1H),7.87 (d, J=8.8 Hz, 1H), 7.56 (d, J=7.8 Hz, 1H), 7.38 (s, 1H), 7.32 (d, J=3.8 Hz, 1H), 7.20-7.09 (m, 1H),6.75 (d, J=8.8 Hz, 1H), 6.60 (d, J=3.8 Hz, 1H), 4.86 (br s, 1H), 4.76-4.68 (m, 1H), 3.95 (t, J=7.3 Hz, 1H), 2.84 (t, J=8.2 Hz, 2H), 2.71-2.57 (m, 1H), 2.24-2.09 (m, 1H),2.08-1.78 (m, 2H), 0.58 (d, J=7.5Hz, 3H). LCMS: (ESI): m/z calcd. for C23H27N6O2, 419.5 [M+H]+, found 419.2.

Example 4 Compounds 7 and 8

Chloro(isopropyl)magnesium (2 M, 449.41 μL, 2 eq.) was added dropwise to a solution of 4-bromo-1,2-dichloro-benzene (203.04 mg, 898.83 μmol, 2 eq.) in THF (1 mL) at −15 C. The mixture was stirred at −15° C. for 10 min, and then warmed to 0° C. The mixture was stirred at 0° C. for 1 h. A solution of 12B (150 mg, 449.41 μmol, 1 eq.) in THF (1.5 mL) was added at −20° C., and the mixture was stirred at 0° C. for 20 min. The reaction progress was monitored by TLC (PE:EA=2:1). Upon completion, the reaction was quenched with NH4Cl (sat. aq., 2 mL) and extracted with EA (2×3 mL). The combined organic layers were washed with brine (2×3 mL), dried over Na2SO4, and concentrated to give a residue. The residue was purified by column chromatography (SiO2, PE:EA=20:1 to 6:1) to afford two isomers, (R or S)-[(15S,16R,17S,18R,21R)-16-(4-chloropyrrolo[2,3-d]pyrimidin-7-yl)-20,20-dimethyl-26,27-dioxatricyclononan-21-yl]-(3,4-dichlorophenyl)methanol (2E-a) (60 mg, 92.35 μmol, 20% yield) as a white solid, and (S or R)-[(15S,16R,17S,18R,21R)-16-(4-chloropyrrolo[2,3-d]pyrimidin-7-yl)-20,20-dimethyl-26,27-dioxatricyclononan-21-yl]-(3,4-dichlorophenyl)methanol (2E-b) (70 mg, 110.66 μmol, 24% yield) as a white solid.

Compound 2E-a: LCMS: (ESI): m/z calcd. for C22H21Cl3N3O3 482.05 [M+H]+, found 481.7. 1H NMR (400 MHz, CDCl3) δ: 8.70 (s, 1H),7.70 (s, 1H), 7.48-7.35 (m, 2H),7.30 (d, J=3.5 Hz, 1H), 6.66 (d, J=3.3 Hz, 1H), 5.84 (s, 1H), 5.49 (d, J=7.3 Hz, 1H), 4.74 (s, 1H), 4.68 (d, J=7.5 Hz, 1H), 4.44 (s, 1H), 1.94 (br dd, J=4.7, 9.2 Hz, 1H), 1.46 (s, 3H),1.32-1.28 (m, 2H), 1.15 (s, 3H). Compound 2E-b: LCMS: (ESI): m/z calcd. for C22H21Cl3N3O3 482.05 [M+H]+, found 481.9. 1H NMR (400 MHz, CDCl3) δ: 8.72 (s, 1H),7.39-7.32 (m, 2H), 7.26 (br s, 1H),7.09 (dd, J=1.5, 8.2 Hz, 1H), 6.66(d, J=3.5 Hz, 1H), 5.68 (d, J=7.3 Hz, 1H), 5.59 (d, J=6.2 Hz, 1H),5.35 (d, J=6.2 Hz, 1H), 4.81 (s, 1H),4.67 (d, J=7.5 Hz, 1H), 1.66 (br dd, J=4.4, 9.3 Hz, 2H), 1.59-1.57 (m,6H), 1.10 (s, 1H), 0.56 (br dd, J=6.5, 8.5 Hz, 1H).

NH3.H2O (728.00 mg, 5.82 mmol, 0.8 mL, 28% purity, 46.61 eq.) was added to a solution of (2E-a) (60 mg, 124.80 μmol, 1 eq.) in dioxane (1.2 mL). The mixture was stirred at 100° C. for 16 h. The reaction progress was monitored by TLC (PE:EA=1:1). Upon completion, the mixture was extracted with EA (3×5 mL). The combined organic layers were washed with brine (3×5 mL), dried over Na2SO4, and concentrated to give a residue that afforded (R or S)-[(15S,16R,17S,18R,21R)-16-(4-aminopyrrolo[2,3-d]pyrimidin-7-yl)-20,20-dimethyl-27,28-dioxatricyclononan-21-yl]-(3,4-dichlorophenyl)methanol (3E-a) (68 mg, crude) as a pale yellow solid. LCMS: (ESI): m/z calcd. for C22H23Cl2N4O3 461.11 [M+H]+, found 461.0.

(S or R)-[(15S,16R,17S,18R,21R)-16-(4-aminopyrrolo[2,3-d]pyrimidin-7-yl)-20,20-dimethyl-27,28-dioxatricyclononan-21-yl]-(3,4-dichlorophenyl)methanol (3E-b) (67 mg, crude) was obtained as a pale yellow solid by the same procedure from (S or R)-[(15S,16R,17S,18R,21R)-16-(4-chloropyrrolo[2,3-d]pyrimidin-7-yl)-20,20-dimethyl-26,27-dioxatricyclononan-21-yl]-(3,4-dichlorophenyl)methanol (2E-b) (70 mg, 145.60 μmol, 1 eq.). LCMS: (ESI): m/z calcd. for C22H23Cl2N4O3 461.11 [M+H]+, found 461.1.

HCl (4 M, aq., 0.75 mL, 20.35 eq.) was added to a solution of (3E-a) (68 mg, 147.40 μmol, 1 eq.) in THF (1.5 mL). The mixture was stirred at 25° C. for 16 h. The reaction progress was monitored by TLC (DCM:MeOH=10:1). Upon completion, the reaction mixture was quenched by NH4OH (25% wt, 1 mL) and then concentrated to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Gemini-NX 150*30 mm*5 um; mobile phase: [water (0.04% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 24%-54%, 8 min) and SFC (column: DAICEL CHIRALPAK AD(250 mm*30 mm, 10 um); mobile phase: [0.1% NH3H2O ETOH]; B%: 45%) to afford (1R,2R,3S,4R,5S)-4-(4-aminopyrrolo[2,3-d]pyrimidin-7-yl)-1-[(R or S)-(3,4-dichlorophenyl)-hydroxy-methyl]bicyclo[3.1.0]hexane-2,3-diol (7) (15 mg, 35.29 μmol, 99% purity) as a white solid. LCMS: (ESI): m/z calcd. for C19H19Cl2N4O3 421.08 [M+H]+, found 421.0. 1H NMR (400 MHz, MeOD) δ: 8.09 (s, 1H),7.69 (s, 1H), 7.46 (q, J=8.4 Hz, 2H), 7.24 (d, J=3.3 Hz, 1H), 6.57 (d, J=3.3 Hz, 1H), 4.83 (br s, 1H), 4.74(br d, J=6.5 Hz, 1H), 4.58 (s, 1H),3.88 (br d, J=6.5 Hz, 1H), 1.62 (brd, J=9.3 Hz, 1H), 1.48 (br s, 1H),1.16-1.08 (m, 1H).

(1R, 2R, 3S, 4R, 5S)-4-(4-aminopyrrolo[2,3-d]pyrimidin-7-yl)-1-[(S or R)-(3,4-dichlorophenyl)-hydroxy-methyl]bicyclo[3.1.0]hexane-2,3-diol (8) (19 mg, 44.65 μmol, 99% purity) was obtained as a white solid by the same procedure and purified by prep-HPLC (column: Phenomenex Gemini-NX 150*30 mm*5 um; mobile phase: [water (0.04% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 24%-54%, 8 min) and SFC (column: DAICEL CHIRALPAK AD(250 mm*30 mm, 10 um); mobile phase: [0.1% NH3H2O ETOH]; B%: 45%) from (S or R)-[(15S,16R,17S,18R,21R)-16-(4-aminopyrrolo[2,3-d]pyrimidin-7-yl)-20,20-dimethyl-27,28-dioxatricyclononan-21-yl]-(3,4-dichlorophenyl)methanol (3E-b) (67 mg, crude). Compound 8: LCMS: (ESI): m/z calcd. for C19H19Cl2N4O3 421.08 [M+H]+, found 421.1. 1H NMR (400 MHz, CD3OD) (signals under solvent peak not listed) δ: 8.08 (br s,1H), 7.54-7.45 (m, 2H), 7.42 (br s,1H), 7.31 (br s, 1H), 6.58 (br s, 1H),5.17 (br s, 1H), 3.78 (br s, 1H), 1.52-1.39 (m, 2H), 0.38 (br s, 1H).

Example 5 Compound 9

To a solution of (1R,2S,3R,5R)-3-amino-5-(hydroxymethyl)cyclopentane-1,2-diol (1F) (3.67 g, 20.0 mmol, 1 eq., HCl salt) and 2-(4,6-dichloropyrimidin-5-yl)acetaldehyde (1F-a) (3.82 g, 20.0 mmol, 1 eq.) in EtOH (80 mL) was added TEA (6.07 g, 60 mmol, 8.4 mL, 3 eq.). The mixture was refluxed for 24 h. The reaction progress was monitored by LCMS. Upon completion, the mixture was concentrated under vacuum to give a residue. The residue was dissolved in sat. NaHCO3 solution (50 mL), and then extracted with EtOAc (9×50 mL). The combined organic layer were washed with brine (300 mL), dried over Na2SO4, and then filtered. The filtrate was concentrated under reduced pressure to afford the crude 2F (5.8 g) as a yellow gum, which was used for next step without further purification.

To a solution of 2F (5.8 g, crude) and 2,2-dimethoxypropane (4.26 g, 40.8 mmol, 5.0 mL, 2 eq.) in acetone (60 mL) was added 4-methylbenzenesulfonic acid hydrate (388.9 mg, 2.0 mmol, 0.1 eq.). The mixture was stirred at rt for 2 h and then refluxed for 24 h. The reaction progress was monitored by LCMS. Upon completion, the reaction was quenched by Et3N (0.7 mL), and then concentrated under reduced pressure to give a residue. The residue was treated with sat. NaHCO3 solution (20 mL) and brine (20 mL). The resulting mixture was extracted with EtOAc (3×40 mL). The combined organic layers were washed with brine (100 mL), dried over Na2SO4, and then filtered. The filtrate was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (PE:EA=5:1 to 1:1) to afford [(3aS,4R,6R,6aR)-4-(4-chloropyrrolo[2,3-d]pyrimidin-7-yl)-2,2-dimethyl-4,5,6,6a-tetrahydro-3aH-cyclopenta[d][1,3]dioxol-6-yl]methanol (3F) (4.25 g, 65% yield, 2 steps from 1F) as a yellow solid. LCMS: (ESI): m/z calcd. for C15H19ClN3O3 324.11 [M+H]+, found 324.2.

To a solution of PPh3 (6.72 g, 25.6 mmol, 2 eq.) and imidazole (1.83 g, 26.9 mmol, 2.1 eq.) in THF (60 mL) was added I2 (6.51 g, 25.6 mmol, 2 eq.). The mixture was stirred at rt for 15 min under N2, and then a solution of 3F (4.15 g, 12.8 mmol, 1 eq.) in THF (40 mL) was added. The mixture was stirred at rt for 1.5 h under N2. The reaction progress was monitored by TLC (PE:E=5:1). Upon completion, the reaction was quenched with sat. Na2S2O3 solution (100 mL), and then extracted with DCM (3×100 mL). The combined organic layers were washed with brine (300 mL), dried over Na2SO4, and then filtered. The filtrate was concentrated in vacuum to give a residue. The residue was purified by column chromatography (PE:EA=20:1 to 5:1) to afford 7-[(3aS,4R,6S,6aR)-6-(iodomethyl)-2,2-dimethyl-4,5,6,6a-tetrahydro-3aH-cyclopenta[d][1,3]dioxol-4-yl]-4-chloro-pyrrolo[2,3-d]pyrimidine (4F) (4.16 g, 70% yield) as a yellow solid. LCMS: (ESI): m/z calcd. for C15H18ClIN3O2 434.01 [M+H]+, found 434.0.

tBuOK (697.0 mg, 6.2 mmol, 1 eq.) in THF (8 mL) was added dropwise to a mixture of 4F (2.69 g, 6.2 mmol, 1 eq.) and THF (30 mL) at 0° C. The mixture was stirred at 0° C. for 0.5 h. The reaction progress was monitored by TLC (PE:EA=5:1). Upon completion, the reaction was quenched with sat. aq. NH4Cl (50 mL). The mixture was extracted with EA (2×50 mL). The separated organic layers were combined and washed with brine (30 mL), dried over anhydrous Na2SO4 and concentrated to afford a residue. The residue was purified by silica gel chromatography (EA:PE=8:1) to afford 7-[(3aR,6R,6aS)-2,2-dimethyl-4-methylene-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-6-yl]-4-chloro-pyrrolo[2,3-d]pyrimidine (5F) (1.9 g, 94% yield) as colorless foam. LCMS: (ESI): m/z calcd. for C15H17ClN3O2 306.10 [M+H]+, found 306.1.

K2OsO4.2H2O (75.32 mg, 204.41 μmol, 0.025 eq.) was added to a mixture of 5F (2.5 g, 8.2 mmol, 1 eq.) and NMO (1.9 g, 16.4 mmol, 2 eq.) in a mixed solvent of acetone (40 mL) and H2O (8 mL). The mixture was stirred at rt for 20 h. The reaction progress was monitored by TLC (PE:EA=5:1). Upon completion, the mixture was concentrated under reduced pressure to afford a residue. The residue was purified by silica gel chromatography (PE:EA:EtOH=30:10:1) to afford (3aS,4R,6R,6aS)-6-(4-chloropyrrolo[2,3-d]pyrimidin-7-yl)-4-(hydroxymethyl)-2,2-dimethyl-3a,5,6,6a tetrahydrocyclopenta[d][1,3]dioxol-4-ol (6F) (2.51 g, 90% yield) as a light yellow foam. LCMS: (ESI): m/z calcd. for C15H19ClN3O4 340.11 [M+H]+, found 340.0.

Benzoyl chloride (297.87 mg, 2.12 mmol, 246.17 μL, 1.2 eq.) was added dropwise to a mixture of 6F (600 mg, 1.77 mmol, 1 eq.), DMAP (21.6 mg, 176.6 μmol, 0.1 eq.), and Et3N (536.1 mg, 5.3 mmol, 737.4 μL, 3 eq.) in DCM (10 mL) at rt. The mixture was stirred at rt for 3 h. The reaction progress was monitored by LCMS. Upon completion, the mixture was concentrated under reduced pressure to afford a residue. The residue was purified by silica gel chromatography (EA:PE=3:1) to afford [(3aS,4R,6R,6aS)-6-(4-chloropyrrolo[2,3-d]pyrimidin-7-yl)-4-hydroxy-2,2-dimethyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-4-yl]methyl benzoate (7F) (743 mg, 94% yield) as a white foam. LCMS: (ESI): m/z calcd. for C22H23ClN3O5 444.13 [M+H]+, found 444.1.

DAST (435.76 mg, 2.70 mmol, 357.18 μL, 2 eq.) was added to a solution of 7F (600 mg, 1.35 mmol, 1 eq.) in DCM (10 mL) at 0° C. The mixture was stirred at 0° C. for 1 h. The reaction progress was monitored by LCMS. Upon completion, the reaction was quenched with sat. NaHCO3 (aq., 10 mL). The mixture was extracted with DCM (2×20 mL). The separated organic layers were combined and washed with brine (10 mL), dried over anhydrous Na2SO4 and concentrated to afford a residue. The residue was purified by chromatography on silica gel (PE:EA=8:1) to afford a mixture of 8F and 8F-a (200 mg, 8:8a=1:2) as a white foam. 8F: LCMS (ESI): m/z calcd. for C22H22ClFN3O4 446.13 [M+H]+, found 446.2; and 8F-a: LCMS (ESI): m/z calcd. for C22H21ClN3O4 426.12 [M+H]+, found 426.2.

A mixture of 8F:8a-F (=1:2, 50.00 mg) and NH3.H2O (1.7 mL, 25% wt) in dioxane (10 mL) was heated at 100° C. for 16 h. The mixture was concentrated under reduced pressure to afford a residue. The residue was purified by silica gel chromatography (DCM:MeOH=20:1) to afford the crude 9F as colorless gum (crude, 32 mg). LCMS: (ESI): m/z calcd. for C15H20FN4O3 323.15 [M+H]+, found 323.2.

A mixture of 9F (crude 90 mg), DMF-DMA (133.1 mg, 1.1 mmol, 148.4 μL, 4 eq.) and THF (2 mL) was stirred at 60° C. for 48 h. The reaction progress was monitored by LCMS. Upon completion, the mixture was concentrated to afford crude 10F (crude, 90 mg) as a yellow gum, which was used for next step without further purification.

p-TsCl (159.12 mg, 834.63 μmol, 3 eq.) was added to a mixture of 10F (105 mg, 278.2 μmol, 1 eq.), TEA (140.8 mg, 1.4 mmol, 194 μL, 5 eq.), DMAP (3.4 mg, 27.8 μmol, 0.1 eq.) and DCM (1 mL). The mixture was stirred at 15° C. for 2 h. The reaction progress was monitored by TLC (DCM:MeOH=10:1). Upon completion, the mixture was concentrated to afford a residue. The residue was purified by silica gel chromatography (DCM:MeOH=30:1) to afford [(3aS,4S,6R,6aS)-6-[4-[(E)-dimethylaminomethyleneamino]pyrrolo[2,3-d]pyrimidin-7-yl]-4-fluoro-2,2-dimethyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-4-yl]methyl 4-methylbenzenesulfonate (11F) (58 mg) as a yellow foam. LCMS: (ESI): m/z calcd. for C25H31FN5O5S 532.20 [M+H]+, found 532.1.

A mixture of 11F (58 mg, 109.10 μmol, 1 eq.), 2-amino-3-bromo-quinolin-7-ol (Q3) (39.1 mg, 163.7 μmol, 1.5 eq.) and Cs2CO3 (106.7 mg, 327.3 μmol, 3 eq.) in DMF (1 mL) was stirred at 70° C. for 16 h. The reaction progress was monitored by LCMS. Upon completion, the mixture was diluted with water (10 mL). The resulting mixture was extracted with EA (2×20 m). The separated organic layers were combined and washed with brine (10 mL), dried over anhydrous Na2SO4, and concentrated to afford a residue. The residue was purified by silica gel chromatography (DCM:MeOH:Et3N=30:1:0.3) to afford the crude product (30 mg). The crude product was further purified by chiral-SFC (column: DAICEL CHIRALCEL OD-H(250 mm*30 mm, 5 um); mobile phase: [0.1% NH3H2O ETOH]; B%: 45%) to afford 7-[[(3aS,4S,6R,6aS)-6-(4-aminopyrrolo[2,3-d]pyrimidin-7-yl)-4-fluoro-2,2-dimethyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-4-yl]methoxy]-3-bromo-quinolin-2-amine (12F) (18 mg, 26% yield) as a white solid. LCMS: (ESI): m/z calcd. for C24H25BrFN6O3 543.12 [M+H]+, found 543.2.

HCl (4 M, 1 mL, 120.75 eq.) was added to the mixture of 12F (18 mg, 33.1 μmol, 1 eq.) and THF (2 mL) at rt. The mixture was stirred at rt, and the reaction progress was monitored by LCMS. Upon completion, the mixture was concentrated under reduced pressure to afford a residue. NH3H2O (0.1 mL) was added to neutralize the residue. The resultant mixture was dissolved in MeCN:H2O=1:1 (3 mL) and then purified by prep-HPLC (column: Phenomenex Gemini-NX 150*30 mm*5 um; mobile phase: [water (0.04% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 27%-57%, 8 min) to afford (1S,2S,3S,5R)-3-[(2-amino-3-bromo-7-quinolyl)oxymethyl]-5-(4-aminopyrrolo[2,3-d]pyrimidin-7-yl)-3-fluoro-cyclopentane-1,2-diol (9) (9 mg, 54% yield) as white solid. LCMS: (ESI): m/z calcd. for C21H21BrFN6O3 503.08 [M+H]+, found 503.1. 1H NMR (400 MHz, CD3OD) δ: 8.22 (s, 1H), 8.04 (s, 1H), 7.56 (d, J=9.0 Hz, 1H), 7.24 (d, J=3.7 Hz, 1H), 7.07 (d, J=2.2 Hz, 1H), 7.00 (dd, J=2.4, 8.8 Hz, 1H), 6.59 (d, J=3.7 Hz, 1H), 5.19-5.06 (m, 1H), 4.60-4.56 (m, 1H), 4.53-4.35 (m, 3H), 2.76-2.45 (m, 2H).

Example 6 Compound 10

A mixture of 1 (20.1 mg, 40.4 μmol, 1 eq.), 2,4,6-trimethyl-1,3,5,2,4,6-trioxatriborinane (15.2 mg, 121.1 μmol, 16.9 μL, 3 eq.), K3PO4 (25.7 mg, 121.1 μmol, 3 eq.) and Pd(dppf)Cl2 (2.9 mg, 4.0 μmol, 0.1 eq.) in a mixed solvent of dioxane (1 mL) and water (0.2 mL) was stirred at 90° C. under Ar for 16 h. The reaction progress was monitored by LCMS. Upon completion, the mixture was filtered through a filter element. The filtrate was purified by acid pre-HPLC (column: Venusil ASB Phenyl 150*30 mm*5 um; mobile phase: [water (0.05% HCl)-ACN]; B%: 15%-45%,10 min) and then by basic pre-HPLC (column: Phenomenex Gemini-NX 150*30 mm*5 um; mobile phase: [water (0.04% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 25%-55%, 8 min) to afford (1S,2R,3S,5R)-3-[2-(2-amino-3-methyl-7-quinolyl)ethyl]-5-(4-aminopyrrolo[2,3-d]pyrimidin-7-yl)-3-methyl-cyclopentane-1,2-diol (10) (5.4 mg, 31% yield) as a white solid. LCMS: (ESI): m/z calcd. for C24H29N6O2 433.23 [M+H]+, found 433.3. 1H NMR (400 MHz, CD3OD) δ: 8.08 (s, 1H), 7.74 (s, 1H), 7.54 (d, J=8.1 Hz, 1H), 7.38 (s, 1H), 7.24 (d, J=3.7 Hz, 1H), 7.14 (dd, J=1.3, 8.2 Hz, 1H), 6.60 (d, J=3.7 Hz, 1H), 5.07-4.98 (m, 1H), 4.52 (t, J=7.0 Hz, 1H), 3.94 (d, J=6.4 Hz, 1H), 2.92-2.71 (m, 2H), 2.29 (s, 3H), 2.12-2.04 (m, 1H), 1.99-1.80 (m, 3H), 1.25 (s, 3H).

Example 7 Compound 11

Tert-butoxycarbonyl tert-butyl carbonate (30.00 g, 137.46 mmol, 31.58 mL, 1.5 eq.) was added dropwise to a mixture of (1S,4R)-3-azabicyclo[2.2.1]hept-5-en-2-one (1H) (10 g, 91.64 mmol, 1 eq.), DMAP (1.12 g, 9.16 mmol, 0.1 eq.) and TEA (18.55 g, 183.27 mmol, 25.51 mL, 2 eq.) in DCM (150 mL) at 0° C. The mixture was stirred at 25° C. for 12 h. The reaction progress was monitored by TLC (PE:EA=3:1). Upon completion, the reaction was quenched by NaHCO3 (sat. aq., 20 mL) and extracted with DCM (2×20 mL). The combined organic layers were washed with brine (2×20 mL), dried over MgSO4 and concentrated to give a residue. The residue was purified by column chromatography (SiO2, PE:EA=15:1 to 5:1) to afford tert-butyl (1S, 4R)-2-oxo-3-azabicyclo [2.2.1] hept-5-ene-3-carboxylate (2H) (16.5 g, 78.86 mmol, 86% yield) as a white solid. LCMS: (ER): m/z calcd. for C11H15NNaO3 232.09 [M+Na]+, found 232.0.

m-CPBA (19.41 g, 95.58 mmol, 85% purity, 4 eq.) was added to a solution of 2H (5 g, 23.90 mmol, 1 eq.) in DCE (150 mL). The mixture was stirred at 25° C. for 96 h. The mixture was stirred at 25° C. for 12 h. The reaction progress was monitored by TLC (PE:EA=3:1). Upon completion, the reaction was quenched by Na2S2O3 (sat. aq., 10 mL) and extracted with DCM (2×30 mL). The combined organic layers were washed with brine (2×30 mL), dried over MgSO4 and concentrated to give a residue. The residue was purified by column chromatography (SiO2, PE:EA=30:1 to 5:1) to afford tert-butyl (4S, 5R, 6R, 7S)-8-oxo-14-oxa-11-azatricyclooctane-11-carboxylate (3H) (4.6 g, 14.30 mmol, 60% yield) as a white solid. LCMS: (ER): m/z calcd. for C11H15NNaO4 248.09 [M+Na]+, found 247.9.

NaBH4 (2.65 g, 69.93 mmol, 5 eq.) was added in portions to a solution of 3H (4.5 g, 13.99 mmol, 1 eq.) in MeOH (120 mL) at 0° C. The mixture was stirred at 0° C. for 1 h. The reaction progress was monitored by TLC (PE:EA=1:1). Upon completion, the mixture was neutralized by a solution of 10% acetic acid in MeOH, and then extracted with DCM (3×50 mL). The combined organic layers were washed with brine (2×50 mL), dried over Na2SO4, and concentrated to give a residue. The residue was purified by column chromatography (SiO2, PE:EA=15:1 to 3:1) to afford tert-butyl N-[(1S, 2R, 4R, 5R)-4-(hydroxymethyl)-6-oxabicyclo [3.1.0]hexan-2-yl]carbamate (4H) (2.35 g, 10.25 mmol, 71% yield) as a white solid. 1H NMR (400 MHz, CDCl3) δ: 5.79 (br d, J=7.5 Hz, 1H), 4.29 (br s, 1H), 3.93 (br d, J=10.3 Hz, 1H), 3.69 (td, J=3.5, 10.1 Hz, 1H), 3.44 (s, 2H), 2.42 (br d, J=9.0 Hz, 1H), 2.19-2.06 (m, 1H), 1.78 (br s, 1H), 1.46(s, 9H), 1.43 (br s, 1H).

To a solution of NaOH (1 M, 88 mL, 9.17 eq.) in H2O was added 4H (2.2 g, 9.60 mmol, 1 eq.). The mixture was stirred at 75° C. for 2 h. The mixture was stirred at 0° C. for 1 h. The reaction progress was monitored by TLC (DCM:MeOH=20:1). Upon completion, the reaction mixture was quenched by H-form ion-exchange resin (pH to ˜8) and then filtered. The filtrate was concentrated under reduced pressure to give a residue to afford (1R,2R,3R,5R)-3-amino-5-(hydroxymethyl)cyclopentane-1,2-diol (5H) (2.13 g, crude) as a yellow solid.

1-fluoro-2,4-dinitro-benzene (5H-a) (1.61 g, 8.66 mmol, 1.09 mL, 1 eq.) and Na2CO3 (917.36 mg, 8.66 mmol, 1 eq.) were added to a solution of 5H (1.93 g, 8.66 mmol, 1 eq., crude) in DMF (20 mL). The mixture was stirred at 25° C. for 2 h. The reaction progress was monitored by TLC (DCM:MeOH=20:1). Upon completion, the mixture was extracted with DCM (2×30 mL). The combined organic layers were washed with brine (2×30 mL), dried over Na2SO4, and concentrated to give a residue. The residue was purified by column chromatography (SiO2, DCM:MeOH=100:1 to 20:1) to afford (1R,2R,3R,5R)-3-(2,4-dinitroanilino)-5-(hydroxymethyl)cyclopentane-1,2-diol (6H) (1.3 g, 4.07 mmol, 47% yield) as a yellow solid. LCMS: (ESI): m/z calcd. for C12H16N3O7 314.09 [M+H]+, found 313.9.

Chloro-[chloro(diisopropyl)silyl]oxy-diisopropyl-silane (6H-a) (1.44 g, 4.56 mmol, 1.46 mL, 1.1 eq.) was added dropwise to a solution of 6H (1.3 g, 4.15 mmol, 1 eq.) in pyridine (15 mL). The mixture was stirred at 25° C. for 12 h. The reaction progress was monitored by TLC (PE:EA=3:1). Upon completion, the mixture was concentrated to give a residue. The residue was purified by column chromatography (SiO2, PE:EA=20:1 to 5:1) to afford (6aR,8R,9R,9aR)-8-(2,4-dinitroanilino)-2,2,4,4-tetraisopropyl-6,6a,7,8,9,9a-hexahydrocyclopenta[f][1,3,5,2,4]trioxadisilocin-9-ol (7H) (1.62 g, 2.91 mmol, 70% yield) as a yellow solid. LCMS: (ESI): m/z calcd. for C24H42N3O8Si2 556.24 [M+H]+, found 556.1.

DAST (555.89 mg, 3.10 mmol, 455.65 μL, 90% purity, 1.5 eq.) was added to a solution of 7H (1.15 g, 2.07 mmol, 1 eq.) in DCM (25 mL) at 0° C. The mixture was stirred at 0° C. for 1 h. The reaction progress was monitored by TLC (PE:EA=3:1). Upon completion, the mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, PE:EA=40:1 to 15:1) to afford (6aR,8R,9S,9aR)-N-(2,4-dinitrophenyl)-9-fluoro-2,2,4,4-tetraisopropyl-6,6a,7,8,9,9a-hexahydrocyclopenta[f][1,3,5,2,4]trioxadisilocin-8-amine (8H) (630 mg, 508.28 μmol, 24% yield) as a yellow solid. LCMS: (ESI): m/z calcd. for C24H41FN3O7Si2 558.24 [M+H]+, found 558.3.

NH4F (470.63 mg, 12.71 mmol, 25 eq.) was added to a solution of 8H (630 mg, 508.28 μmol, 1 eq.) in MeOH (23 mL). The mixture was stirred at 25° C. for 12 h. The reaction progress was monitored by TLC (DCM:MeOH=20:1). Upon completion, the mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, DCM:MeOH=200:1 to 100:1) to afford (1R, 2S, 3R, 5R)-3-(2, 4-dinitroanilino)-2-fluoro-5-(hydroxymethyl) cyclopentanol (9H) (135 mg, 398.25 μmol, 78% yield) as a yellow solid. LCMS: (ESI): m/z calcd. for C12H15FN3O6 316.09 [M+H]+, found 315.9.

Amberlite IRA 400(OH—) resin (2.2 g) was added to a solution of 9H (135 mg, 428.23 μmol, 1 eq.) in acetone (3.8 mL) and H2O (1.9 mL). The mixture was stirred at 25° C. for 12 h. The reaction progress was monitored by TLC (DCM:MeOH=10:1). Upon completion, the mixture was filtered and washed with water (2 mL). HCl (1 N, 0.8 mL) was added, and the solution was washed with EA (3×5 mL). The mixture was concentrated under reduced pressure to give (1R, 2S, 3R, 5R)-3-amino-2-fluoro-5 (hydroxymethyl) cyclopentanol (10H) (97 mg, crude, HCl salt) as a pale yellow solid. 1H NMR (400 MHz, DMSO-d6) δ: 8.59 (br s, 3H), 4.91-4.68 (m, 1H), 3.61-3.49 (m, 1H), 3.49-3.30 (m, 2H), 2.15 (td, J=8.4, 13.2 Hz, 1H), 1.95 (br d, J=5.3 Hz, 1H), 1.33 (td, J=9.3, 13.2 Hz, 1H).

Et3N (181.53 mg, 1.79 mmol, 249.70 μL, 3 eq.) and 2-(4, 6-dichloropyrimidin-5-yl) acetaldehyde (10H-a) (114.22 mg, 597.98 μmol, 1 eq.) was added to a solution of 10H (185 mg, 597.98 μmol, 1 eq., HCl) in EtOH (3 mL). The mixture was stirred at 80° C. for 2 h. The reaction progress was monitored by TLC (DCM:MeOH=10:1). Upon completion, the mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, DCM:MeOH=200:1 to 50:1) to afford (1R, 2S, 3R, 5R)-3-(4-chloropyrrolo [2, 3-d] pyrimidin-7-yl)-2-fluoro-5-(hydroxymethyl) cyclopentanol (11H) (144 mg, 504.02 μmol, 84% yield) as a yellow oil. LCMS: (ESI): m/z calcd. for C12H14ClFN3O2 286.07 [M+H]+, found 285.9.

4-methylbenzenesulfonyl chloride (80.08 mg, 420.02 μmol, 1.2 eq.) was added to a solution of 11H (100 mg, 350.02 μmol, 1 eq.) in pyridine (0.5 mL) at 0° C. The mixture was stirred at 25° C. for 2 h. The reaction progress was monitored by TLC (DCM:MeOH=20:1). The mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, PE:EA=20:1 to 1:2) to afford [(1R,2R,3S,4R)-4-(4-chloropyrrolo[2,3-d]pyrimidin-7-yl)-3-fluoro-2-hydroxy-cyclopentyl]methyl 4-methylbenzenesulfonate (12H) (57 mg, 129.58 μmol, 37% yield) as a yellow solid. LCMS: (ESI): m/z calcd. for C19H20ClFN3O4S 440.08 [M+H]+, found 440.0.

Cs2CO3 (148.88 mg, 456.94 μmol, 3 eq.) was added to a mixture of 12H (67 mg, 152.31 μmol, 1 eq.) and 2-amino-3-bromo-quinolin-7-ol (36.41 mg, 152.31 μmol, 1 eq.) (Q3) in DMF (0.5 mL). The mixture was stirred at 25° C. for 15 h. The reaction progress was monitored by TLC (DCM:MeOH=20:1). Upon completion, the mixture was diluted with water (5 mL) and extracted with EA (3×3 mL). The combined organic layers were washed with brine (2×5 mL), dried over Na2SO4, and concentrated to give a residue. The residue was purified by prep-TLC (SiO2, DCM:MeOH=20:1) to afford (1R, 2S, 3R, 5R)-5-[(2-amino-3-bromo-7-quinolyl) oxymethyl]-3-(4-chloropyrrolo [2,3-d] pyrimidin-7-yl)-2-fluoro-cyclopentanol (13H) (41 mg, 79.94 μmol, 52% yield) as a white solid. LCMS: (ESI): m/z calcd. for C21H19BrClFN5O2 508.03 [M+H]+, found 508.0.

NH3.H2O (4.55 g, 36.35 mmol, 5 mL, 28% purity, 526.34 eq.) was added to a solution of 13H (35 mg, 69.07 μmol, 1 eq.) in dioxane (5 mL). The mixture was stirred at 100° C. for 48 h. The reaction progress was monitored by TLC (DCM:MeOH=10:1). Upon completion, the mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Gemini-NX 150*30 mm*5 um; mobile phase: [water (0.04% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 26%-56%, 8 min) to afford (1R,2S,3R,5R)-5-[(2-amino-3-bromo-7-quinolyl)oxymethyl]-3-(4-aminopyrrolo[2,3-d]pyrimidin-7-yl)-2-fluoro-cyclopentanol (11) (12 mg, 24.25 μmol, 35% yield) as a white solid. LCMS: (ESI): m/z calcd. for C21H21BrFN6O2 487.08 [M+H]+, found 487.1. 1H NMR (400 MHz, CD3OD) δ: 8.21 (s, 1H),8.10 (s, 1H), 7.55 (d, J=9.0 Hz, 1H),7.31 (d, J=3.5 Hz, 1H), 7.04 (d, J=2.3 Hz, 1H), 7.00 (d, J=8.8 Hz, 1H), 6.66 (d, J=3.5 Hz, 1H), 5.37-5.11 (m, 2H), 4.40 (td, J=5.6, 13.2 Hz, 1H), 4.28 (br dd, J=4.6, 8.4 Hz, 2H), 2.62 (br d, J=7.8 Hz, 2H), 2.08-1.96 (m, 1H).

Example 8 Compound 12

To a solution of 7-[(3aR,4R,6R,6aS)-2,2,4-trimethyl-4-vinyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-6-yl]pyrrolo[2,3-d]pyrimidin-4-amine (9A) (180 mg, 572.6 μmol, 1 eq.) in THF (6 mL) was added 9-BBN dimer (304.9 mg, 1.3 mmol, 2.2 eq.). The mixture was stirred at 50° C. for 2 h and cooled to 20° C. K3PO4 (607.7 mg, 2.9 mmol, 5 eq.) and H2O (0.6 mL) were added. The mixture was stirred at 20° C. for 0.5 h. Then 7-bromoquinolin-2-amine (153.3 mg, 687.1 μmol, 1.2 eq.) and Pd(dppf)Cl2 (41.9 mg, 57.3 μmol, 0.1 eq.) were added. The mixture was stirred at 70° C. for 12 h. The reaction progress was monitored by LCMS. Upon completion, the mixture was diluted with brine (10 mL). The mixture was extracted with EA (3×20 mL). The organic layers were combined, dried over anhydrous Na2SO4, and concentrated to afford a residue. The residue was purified by reversed-phase HPLC (0.1% NH3H2O) to afford 7-[2-[(3aR,4S,6R,6aS)-6-(4-aminopyrrolo[2,3-d]pyrimidin-7-yl)-2,2,4-trimethyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-4-yl]ethyl]quinolin-2-amine (2J) (175 mg, 64.5% yield) as white solid. LCMS: (ESI): m/z calcd. for C26H31N6O2 459.25 [M+H]+, found 459.4.

A mixture of 2J (230 mg, 501.58 μmol, 1 eq.), HCl (4 M, 3 mL) and THF (6 mL) was stirred at rt for 6 h. The reaction progress was monitored by LCMS. Upon completion, the mixture was concentrated under reduced pressure to afford a solid. The solid was washed with MeCN:H2O (10:1, 6 mL) to afford (1S,2R,3S,5R)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-3-(2-(2-aminoquinolin-7-yl)ethyl)-3-methylcyclopentane-1,2-diol (12) as a hydrochloride salt (white solid, 180 mg, 72% yield). LCMS: (ESI): m/z calcd. for C23H27N6O2 419.22 [M+H]+, found 419.3. 1H NMR (CD3OD) δ: 8.31 (d, J=9.3Hz, 1H), 8.25 (s, 1H), 7.82 (d, J=8.2 Hz, 1H), 7.58 (d, J=3.7 Hz, 1H), 7.53 (s, 1H), 7.45 (dd, J=1.3, 8.2 Hz, 1H), 7.01 (d, J=9.5 Hz, 1H), 6.92 (d, J=3.5 Hz, 1H), 5.19-5.07 (m, 1H), 4.61-4.49 (m, 1H), 3.96 (d, J=6.4 Hz, 1H), 3.02-2.78 (m, 2H), 2.17-1.80 (m, 4H), 1.25 (s, 3H).

Example 9 Compound 13

NH3.H2O (20 mL, 28% wt) was added to a solution of 4-chloro-7-[(11S,12R,13S,14R,16S)-15,15-dimethyl-16-vinyl-20,21-dioxatricyclononan-12-yl]pyrrolo[2,3-d]pyrimidine (13B) (788 mg, 2.37 mmol, 1 eq.) in dioxane (20 mL). The mixture was stirred at 100° C. for 48 h. The reaction progress was monitored by TLC (DCM:MeOH=20:1). Upon completion, the mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 12 g Sepa Flash® Silica Flash Column, Eluent of 0˜8% DCM/MeOH @ 30 mL/min) to afford 7-[(11S,12R,13S,14R,16S)-15,15-dimethyl-16-vinyl-21,22-dioxatricyclononan-12-yl]pyrrolo[2,3-d]pyrimidin-4-amine (14B) (773 mg, 2.03 mmol, 85% yield) as a white solid. LCMS: (ESI): m/z calcd. for C17H21N4O2 313.16 [M+H]+, found 312.9.

9-BBN dimer (1.05 g, 4.32 mmol, 2.7 eq.) was added to a solution of 14B (500 mg, 1.60 mmol, 1 eq.) in THF (10 mL), and the mixture was stirred at 50° C. for 3.5 h under Ar atmosphere. The mixture was cooled to rt, and then K3PO4 (1.70 g, 8.00 mmol, 5 eq.) in H2O (1 mL) were added. The resulting mixture was stirred at 25° C. for 0.5 h, and then 7-bromoquinolin-2-amine (Q5) (357.06 mg, 1.60 mmol, 1 eq.) and Pd(dppf)Cl2 (117.12 mg, 160.07 μmol, 0.1 eq.) were added. The mixture was purged with Ar (3×), and then stirred at 72° C. for 12 h. The reaction progress was monitored by TLC (DCM:MeOH=10:1). Upon completion, the mixture was extracted with EA (3×10 mL). The combined organic layers were washed with brine (3×20 mL), dried over Na2SO4 and concentrated to give a residue. The residue was purified by prep-HPLC Combi-flash reversed-phase C-18 column chromatography (10%˜60% CH3CN/water (1 mL/3 L NH3.H2O in water)@ 75 mL/min) to afford 7-[2-[(20S,21R,22S,23R,25R)-21-(4-aminopyrrolo[2,3-d]pyrimidin-7-yl)-24,24-dimethyl-32,33-dioxatricyclononan-25-yl]ethyl]quinolin-2-amine (2K) (446 mg, 976.92 μmol, 19% yield) as a yellow solid. LCMS: (ESI): m/z calcd. for C26H29N6O2 457.23 [M+H]+, found 457.1.

To a solution of 2K (446 mg, 976.92 μmol, 1 eq.) in THF (10 mL) was added HCl (4 M, 5 mL). The mixture was stirred at 25° C. for 12 h. The reaction progress was monitored by TLC (DCM:MeOH=5:1). Upon completion, the mixture was filtered and concentrated under reduced pressure to give a residue (400 mg, crude, HCl salt) as a brown solid. This residue (200 mg) was purified by prep-HPLC (column: Phenomenex Gemini-NX 150*30 mm*5 um; mobile phase: [water (0.04% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 20%-50%, 8 min) to afford (1R,2R,3S,4R,5S)-4-(4-aminopyrrolo[2,3-d]pyrimidin-7-yl)-1-[2-(2-amino-7-quinolyl)ethyl]bicyclo[3.1.0]hexane-2,3-diol (13) (115 mg, 274.39 μmol, 58% yield) as a white solid. LCMS: (ESI): m/z calcd. for C23H25N6O2 417.2 [M+H]+, found 417.2. 1H NMR (400 MHz, CD3OD) δ: 8.09 (s, 1H), 7.90 (d, J=8.8 Hz, 1H), 7.57 (d, J=8.3 Hz, 1H), 7.41 (s, 1H), 7.18(dd, J=1.4, 8.2 Hz, 1H), 7.01 (d, J=3.8 Hz, 1H), 6.77 (d, J=9.0 Hz, 1H), 6.57 (d, J=3.8 Hz, 1H), 4.95 (s,1H), 4.60 (br d, J=6.5 Hz, 1H), 3.86 (d, J=6.8 Hz, 1H), 3.12-2.87 (m,2H), 2.30-2.18 (m, 1H), 2.05-1.93(m, 1H), 1.47 (t, J=4.3 Hz, 1H), 1.39 (dd, J=3.3, 8.5 Hz, 1H), 0.68 (dd, J=5.3, 7.0 Hz, 1H).

Example 10 Compound 14

To a solution of 1 (40 mg, 80.4 μmol, 1 eq.) in dioxane (1 mL) and H2O (0.2 mL) were added K2CO3 (27.8 mg, 201.05 μmol, 2.5 eq.), cyclopropyl boronic acid (17.3 mg, 201.1 μmol, 2.5 eq.), and Pd(dppf)Cl2 (5.9 mg, 8.0 μmol, 0.1 eq.). The mixture was stirred at 100° C. for 16 h under Ar atmosphere. The reaction was monitored by LC-MS. Upon completion, the mixture was filtrated. The filter cake was washed with dioxane/water (5:1) (4 mL). The residue was purified by prep-HPLC (HCl condition; water (0.05% HCl)-ACN, column: Phenomenex Gemini-NX 150*30 mm*5 um, begin: 10, end: 30, Gradient Time (min): 7 min, 100% B Hold Time (min): 0, FlowRate (ml/min): 35) to give (1S,2R,3S,5R)-3-[2-(2-amino-3-cyclopropyl-7-quinolyl)ethyl]-5-(4-aminopyrrolo[2,3-d]pyrimidin-7-yl)-3-methylcyclopentane-1,2-diol (14) (24 mg, 45.05 μmol, 56.0% yield, 99.8% purity, 2 HCl) as a white solid. LCMS: (ESI): m/z calcd. for C26H31N6O2 459.24 [M+H]+, found 459.2. 1HNMR (400 MHz, CD3OD) δ: 8.25 (s, 1 H), 8.12 (s, 1 H), 7.79 (d, J=8.3 Hz, 1 H), 7.56 (d, J=3.4 Hz, 1 H), 7.53 (s, 1 H), 7.40-7.44 (m, 1 H), 6.90-6.93 (m, 1 H), 5.08-5.16 (m, 1 H), 4.52-4.58 (m, 1 H), 3.96 (d, J=6.4 Hz, 1 H), 2.81-3.00 (m, 2 H), 1.97-2.14 (m, 2 H), 1.81-1.96 (m, 3 H), 1.25 (s, 3 H), 1.11-1.17 (m, 2 H), 0.80-0.85 (m, 2 H).

Example 11 Compound 15

To a solution of [(10S,11R,12S,13R,15R)-11-(4-chloropyrrolo[2,3-d]pyrimidin-7-yl)-14,14-dimethyl-20,21-dioxatricyclononan-15-yl]methanol (11B) (500 mg, 1.49 mmol, 1 eq.) in DCM (10 mL) were added Et3N (1.51 g, 14.90 mmol, 2.1 mL, 10 eq.), DMAP (36.4 mg, 298.0 μmol, 0.2 eq.), and 4-methylbenzenesulfonyl chloride (852.2 mg, 4.47 mmol, 3 eq.). The mixture was stirred at 25° C. for 16 h. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 12 g SepaFlash® Silica Flash Column, Eluent of 0˜32% EA:PE gradient @ 36 mL/min) to give 2L (420 mg, 831.5 μmol, 55.8% yield) as a white solid. LCMS: (ESI): m/z calcd. for C23H25ClN3O5S 490.11 [M+H]+, found 490.1.

To a mixture of 2L (345 mg, 704.12 μmol, 1 eq.) and 2-amino-3-bromo-quinolin-7-ol (Q3) (168.33 mg, 704.12 μmol, 1.0 eq.) in DMF (3 mL) was added Cs2CO3 (688.25 mg, 2.11 mmol, 3.0 eq.). The mixture was stirred at 20° C. for 16 h. The reaction progress was monitored by TLC. The mixture was then diluted with H2O (30 mL) and extracted with EtOAc (3×10 mL). The combined organic layers were washed with brine (2×20 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 12 g SepaFlash® Silica Flash Column, Eluent of 0˜80% EA:PE gradient @ 30 mL/min). 3-bromo-7-[[(19S,20R,21S,22R,24R)-20-(4-chloropyrrolo[2,3-d]pyrimidin-7-yl)-23,23-dimethyl-30,31-dioxatricyclononan-24-yl]methoxy]quinolin-2-amine (3L) (280 mg, 482.7 μmol, 68% yield, 96% purity) was obtained as a light yellow solid. LCMS: (ESI): m/z calcd. for C25H25BrClN5O3 558.07 [M+H]+, found 557.9.

A mixture of 3L (280 mg, 502.84 μmol, 1 eq.) and NH3·H2O (5 mL, 25% wt) in dioxane (5 mL) was heated in a sealed tube at 100° C. for 48 h. The reaction progress was monitored by LCMS. The mixture was then extracted with DCM (3×10 mL). The combined organic phase was washed with brine (20 mL), dried over Na2SO4, filtered and concentrated to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Gemini-NX 150*30 mm*5 μm; mobile phase: [water (0.04% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 42%-72%, 8 min). 7-[[(19S,20R,21S,22R,24R)-20-(4-aminopyrrolo[2,3-d]pyrimidin-7-yl)-23,23-dimethyl-31,32-dioxatricyclononan-24-yl]methoxy]-3-bromo-quinolin-2-amine (4L) (140 mg, 247.48 μmol, 49.22% yield, 95% purity) was obtained as a light yellow solid. LCMS: (ESI): m/z calcd. for C25H26BrN6O3 539.12 [M+H]+, found 539.2.

To a mixture of 4L (140 mg, 260.5 μmol, 1.0 eq.) in THF (6 mL) was added HCl (4 M, 2.6 mL). The mixture was then stirred at 20° C. for 16 h. The reaction progress was monitored by LCMS. Upon completion the solution was adjusted to pH=8 with NH3·H2O and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Gemini-NX 150*30 mm*5 μm; mobile phase: [water (0.04% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 25%-55%, 8 min). (1R,2R,3S,4R,5S)-1-[(2-amino-3-bromo-7-quinolyl)oxymethyl]-4-(4-aminopyrrolo[2,3-d]pyrimidin-7-yl)bicyclo[3.1.0]hexane-2,3-diol (15) (74 mg, 146.2 μmol, 56% yield) was obtained as a white solid. LCMS: (ESI): m/z calcd. for C22H22BrN6O3 497.09 [M+H]+, found 497.1. 1H NMR (400 MHz, CD3OD) δ: 8.22 (s, 1H), 8.11 (s, 1H), 7.71 (d, J=3.7 Hz, 1H), 7.59 (d, J=8.8 Hz, 1H), 7.08-7.01 (m, 2H), 6.66 (d, J=3.4 Hz, 1H), 5.10 (s, 1H), 4.84-4.79 (m, 1H), 4.61 (s, 1H), 3.87 (d, J=10.3 Hz, 1H), 3.78 (d, J=6.8 Hz, 1H), 1.77-1.71 (m, 2H), 0.92 (br s, 1H).

Example 12 Compound 16

A mixture of 4-benzyloxypyridin-2-amine (11B) (100 mg, 499.4 μmol, 1 eq.) and Boc2O (119.9 mg, 549.4 μmol, 1.1 eq.) in t-BuOH (1.5 mL) was stirred at 50° C. for 1 h. The reaction progress was monitored by LCMS. Upon completion, the mixture was cooled to rt and diluted with EtOH (5 mL). The precipitate was filtered and dried under high vacuum to give crude 2L (116 mg, 363.1 μmol, 72.7% yield) as a white solid. LCMS: (ESI): m/z calcd. for C17H21N2O3 301.15 [M+H]+, found 301.0.

To a solution of 2L (116 mg, 386.2 μmol, 1 eq.) in DMF (2 mL) was added NaH (23.2 mg, 579.3 μmol, 60% purity, 1.5 eq.) in portions at 17° C. under a N2 atmosphere. Upon completion of the addition, the mixture was stirred for 10 min. 3-bromoprop-1-yne (68.9 mg, 579.3 μmol, 49.9 μL, 1.5 eq.) was then added, and the mixture was stirred at 17° C. for 1 h. The reaction progress was monitored by LCMS. Upon completion, the reaction was quenched by addition H2O (3 mL) and extracted with EA (2×3 mL). The combined organic layers were washed with H2O (2×3 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 12 g SepaFlash® Silica Flash Column, Eluent of 0˜22% EA:PE gradient @ 36 mL/min) to give 3M (110 mg, 312.1 μmol, 80.8% yield) as a light yellow oil. LCMS: (ESI): m/z calcd. for C20H23N2O3 339.16 [M+H]+, found 339.0.

To a solution of 3M (110 mg, 325.1 μmol, 1 eq.) in THF (3 mL) was added t-BuOK (43.8 mg, 390.1 μmol, 1.2 eq.). The mixture was stirred at 15° C. for 30 min. The reaction progress was monitored by LCMS. Upon completion, the mixture was diluted with H2O (3 mL) and extracted with EA (2×3 mL). The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 4 g SepaFlash® Silica Flash Column, Eluent of 0-5% Methanol/Dichloromethane @ 36 mL/min) to give 4M (39 mg, 148.9 μmol, 45.8% yield) as a yellow solid. LCMS: (ESI): m/z calcd. for C15H15N2O 239.11 [M+H]+, found 238.9.

To a solution of 4M (2.3 g, 9.65 mmol, 1 eq.) in MeOH (60 mL) was added Pd/C (10%, 1 g) under N2 atmosphere. The mixture was stirred under H2 (15 Psi) at 15° C. for 1 h. The reaction progress was monitored by LCMS. Upon completion, the mixture was filtered and concentrated under reduced pressure to give a crude Q4 (1.48 g, crude, 85% purity) as a black solid. LCMS: (ESI): m/z calcd. for C8H9N2O 149.06 [M+H]+, found 149.2.

To a solution of [(10S,11R,12S,13R,15R)-11-(4-chloropyrrolo[2,3-d]pyrimidin-7-yl)-14,14-dimethyl-20,21-dioxatricyclononan-15-yl]methanol (11B) (500 mg, 1.49 mmol, 1 eq.) in DCM (10 mL) was added Et3N (1.51 g, 14.90 mmol, 2.1 mL, 10 eq.), DMAP (36.4 mg, 298.0 μmol, 0.2 eq.) and 4-methylbenzenesulfonyl chloride (852.2 mg, 4.47 mmol, 3 eq.). The mixture was stirred at 25° C. for 16 h. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 12 g SepaFlash® Silica Flash Column, Eluent of 0˜32% EA:PE gradient @ 36 mL/min) to give 2L (420 mg, 831.5 μmol, 55.8% yield) as a white solid. LCMS: (ESI): m/z calcd. for C23H25ClN3O5S 490.11 [M+H]+, found 490.1.

To a mixture of 2L (500 mg, 1.02 mmol, 1 eq.) and Q4 (181.4 mg, 1.22 mmol, 1.2 eq.) in DMF (5 mL) was added Cs2CO3 (997.5 mg, 3.06 mmol, 3 eq.). The mixture was stirred at 20° C. for 16 h. The reaction progress was monitored by LCMS. Upon completion, the mixture was diluted with H2O (30 mL) and extracted with EA (3×10 mL). The combined organic layers were washed with brine (2×20 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 12 g SepaFlash® Silica Flash Column, Eluent of 0˜5% MeOH/DCM gradient @ 30 mL/min) to give 7M (350 mg, 721.14 μmol, 70.67% yield, 96% purity) as a slightly yellow solid. LCMS: (ESI): m/z calcd. for C24H25ClN5O3 466.16 [M+H]+, found 466.3.

To a solution of NH3.H2O (10 mL, 25% wt) and dioxane (10 mL) was added 7M (350 mg, 751.2 μmol, 1 eq.). The mixture was heated in a sealed tube at 100° C. and stirred for 40 h. The mixture was extracted with EA (2×10 mL). The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (basic condition: column: Phenomenex Gemini-NX 150*30 mm*5 μm; mobile phase: [water (0.04% NH3.H2O+10 mM NH4HCO3)-ACN]; B%: 28%-58%, 8 min) to give 8M (226 mg, 491 μmol, 65.4% yield) as a light yellow solid. LCMS: (ESI): m/z calcd. for C24H27N6O3 447.21 [M+H]+, found 447.3.

To a solution of 8M (226 mg, 506.16 μmol, 1 eq.) in THF (6 mL) was added HCl (4 M, 3 mL, 23.71 eq.). The mixture was stirred at 25° C. for 12 h. The reaction progress was monitored by LCMS. Upon completion, the mixture was concentrated under reduced pressure to give a residue. The residue was diluted with H2O (10 mL) and lyophilized to give 16 (2 HCl, 245 mg, 505.5 μmol, 99.9% yield) as a white solid. LCMS: (ESI): m/z calcd. for C21H23N6O3 407.18 [M+H]+, found 407.2. 1H NMR (400 MHz, CD3OD) δ: 8.55 (d, J=7.5 Hz, 1H), 8.30 (s, 1H), 7.76 (d, J=3.8 Hz, 1H), 7.63 (d, J=1.0 Hz, 1H), 7.37-7.25 (m, 2H), 7.02 (d, J=3.8 Hz, 1H), 5.24 (s, 1H), 4.85-4.80 (m, 2H), 4.16 (d, J=10.3 Hz, 1H), 3.93 (d, J=6.5 Hz, 1H), 2.59 (d, J=1.0 Hz, 3H), 1.84-1.72 (m, 2H), 1.06-0.96 (m, 1H).

Example 13 Compound 17

A mixture of Rac-[(10S,11R,12R,13S)-15-(4-aminopyrrolo[2,3-d]pyrimidin-7-yl)-14,14-dimethyl-21,22-dioxatricyclononan-11-yl]methanol (25 mg, 79.03 μmol, 1 eq.) in THF (2 mL) was added 1,1-dimethoxy-N,N-dimethyl-methanamine (188.4 mg, 1.58 mmol, 210.0 μL, 20 eq.), and the mixture was then stirred at 60° C. for 24 h. The reaction progress was monitored by LCMS. Upon completion, the mixture was concentrated to afford a residue. The residue was purified by pre-TLC (DCM:MeOH=10:1) to afford Compound rac-N′-[7-[(13S,14R,15R,16S)-14-(hydroxymethyl)-17,17-dimethyl-25,26-dioxatricyclononan-18-yl]pyrrolo[2,3 d]pyrimidin-11-yl]-N,N-dimethyl-formamidine (2N) (26 mg, 70.0 μmol, 88.6% yield) as a brown gum. LCMS: (ESI): m/z calcd, for C19H26N5O3 372.20 [M+H]+, found 372.1.

4-methylbenzenesulfonyl chloride (40.1 mg, 210.0 μmol, 3 eq.) was added to the mixture of 2N (26 mg, 70.0 μmol, 1 eq.), TEA (35.4 mg, 350.0 μmol, 49 μL, 5 eq.) and DMAP (1.7 mg, 14.0 μmol, 0.2 eq.) in DCM (1 mL). After being stirred at 25° C. for 2 h, the mixture was concentrated and purified by pre-TLC (EA) to afford rac-[(20S,21R,22R,23S)-25-[4-[(E)-dimethylaminomethyleneamino]pyrrolo[2,3-d]pyrimidin-7-yl]-24,24-dimethyl-33,34-dioxatricyclononan-21-yl]methyl 15-methylbenzenesulfonate (3N) (15 mg, 28.5 μmol, 40.8% yield) as a brown gum. LCMS: (ESI): m/z calcd, for C26H32N5O5S 526.21 [M+H]+, found 526.1.

To a solution of 3N (15 mg, 28.54 μmol, 1 eq.) and 2-amino-3-bromo-quinolin-7-ol (6.8 mg, 28.5 μmol, 1 eq.) in DMF (1 mL) was added Cs2CO3 (27.9 mg, 85.6 μmol, 3 eq.). The mixture was stirred at 20° C. for 12 h. The reaction progress was monitored by LCMS. Upon completion, the mixture was concentrated to afford a residue. The residue was purified by pre-TLC (EA, 100%) to afford rac-N′-[7-[(22S,23R,24R,25S)-23-[(2-amino-3-bromo-7-quinolyl)oxymethyl]-26,26-dimethyl-35,36-dioxatricyclononan-27-yl]pyrrolo[2,3-d]pyrimidin-20-yl]-N,N-dimethyl-formamidine (4N) (7 mg, 8.1 μmol, 28.5% yield, 68.8% purity) was obtained as a white solid. LCMS: (ESI): m/z calcd, for C28H31BrN7O3 594.16 [M+H]+, found 594.2.

To a solution of 4N (7 mg, 8.1 μmol, 68.8% purity, 1 eq.) in THF (0.6 mL) was added HCl (4 M, 0.3 mL). The mixture was stirred at 20° C. for 12 h. The reaction progress was monitored by LCMS. Upon completion, the mixture was concentrated under reduced pressure to give a residue. The residue was neutralized by NH4OH (0.1 mL), and then purified by prep-HPLC (Column: Boston Prime C18 150*30 mm*5 um, Condition: water(0.04% NH3H2O+10 mM NH4HCO3)-ACN, begin B: 30%, end B: 60%, Gradient Time(min): 8, 100% B Hold Time(min): 2, FlowRate (ml/min): 25) to give rac-(2S,3R,4R,5S)-4-[(2-amino-3-bromo-7-quinolyl)oxymethyl]-1-(4-aminopyrrolo[2,3-d]pyrimidin-7-yl)bicyclo[3.1.0]hexane-2,3-diol (17) (2.6 mg, 5.1 μmol, 43.4% yield, 98.08% purity) as a white solid. LCMS: (ESI): m/z calcd, for C22H22BrN6O3 499.09 [M+H]+, found 499.1. 1H NMR (400 MHz, CD3OD) δ: ppm 8.21 (s, 1 H), 8.03 (s, 1 H), 7.56 (d, J=8.8 Hz, 1 H), 7.15 (d, J=3.7 Hz, 1 H), 7.11 (d, J=2.2 Hz, 1 H), 6.98 (dd, J=8.8, 2.4 Hz, 1 H), 6.51 (d, J=3.7 Hz, 1 H), 4.78 (br d, J=6.4 Hz, 1 H), 4.43-4.52 (m, 2 H), 4.11 (d, J=6.4 Hz, 1 H), 2.60 (t, J=6.1 Hz, 1 H), 1.94-1.98 (m, 2 H), 1.22-1.29 (m, 1 H).

Example 14 Compound 18

To a solution of 7-[(3aR,4R,6R,6aS)-2,2,4-trimethyl-4-vinyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-6-yl]pyrrolo[2,3-d]pyrimidin-4-amine (9A) (200 mg, 636.17 μmol, 1 eq.) in THF (10 mL) was added 9-BBN dimer (338.7 mg, 1.40 mmol, 2.2 eq.) at 20° C. The reaction was stirred at 50° C. for 1 h. The mixture was cooled to 20° C., and a solution of K3PO4 (675.2 mg, 3.18 mmol, 5 eq.) in H2O (2 mL) was added. The mixture stirred for 30 min, and then 7-bromo-3-chloroquinolin-2-amine (Q8) (213 mg, 827.0 μmol, 1.3 eq.) and Pd(dppf)Cl2 (46.6 mg, 63.6 μmol, 0.1 eq.) were added at 20° C. The mixture was stirred at 70° C. for 15 h. The reaction progress was monitored by LCMS. Upon completion the mixture was filtered, diluted with brine (10 mL) and extracted with DCM (2×10 mL). The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, DCM:MeOH=50:1 to 20:1) to give 2O (280 mg, 482.8 μmol, 75.9% yield, 85% purity) as a white solid. LCMS: (ESI): m/z calcd. for C26H30ClN6O2 493.20 [M+H]+, found 493.3.

To a solution of 20 (280 mg, 567.95 μmol, 1 eq.) in THF (5 mL) was added HCl (4 M, 2.5 mL, 17.61 eq.). The mixture was stirred at 20° C. for 12 h. The mixture was concentrated under reduced pressure to give a residue. The crude product was triturated with CH3CN:H2O=10:1 (2×11 mL) at 25° C. for 30 min to give 18 (2 HCl, 218 mg, 406.3 μmol, 71.5% yield, 98% purity, 2 HCl) as a white solid. LCMS: (ESI): m/z calcd. for C23H26ClN6O2 453.17 [M+H]+, found 453.3. 1H NMR (400 MHz, CD3OD) δ: 8.59 (s, 1H), 8.25 (s, 1H), 7.83 (d, J=8.3 Hz, 1H), 7.63-7.55 (m, 2H), 7.49 (d, J=8.3 Hz, 1H), 6.92 (d, J=3.7 Hz, 1H), 5.19-5.05 (m, 1H), 4.61-4.50 (m, 1H), 3.96 (d, J=6.1 Hz, 1H), 3.02-2.82 (m, 2H), 2.15-2.07 (m, 1H), 2.06-1.98 (m, 1H), 1.97-1.83 (m, 2H), 1.25 (s, 3H).

Example 15 Compound 19

9-BBN dimer (193.7 mg, 800.3 μmol, 2.5 eq.) was added to a solution of 7-[(11S,12R,13S,14R,16S)-15,15-dimethyl-16-vinyl-21,22-dioxatricyclononan-12-yl]pyrrolo[2,3-d]pyrimidin-4-amine (14B) (100 mg, 320.1 μmol, 1 eq.) in THF (4 mL). The mixture was stirred at 50° C. for 2 h under N2, and then cooled to 25° C. A solution of K3PO4 (339.8 mg, 1.60 mmol, 5 eq.) in H2O (0.4 mL) were added. The mixture was stirred at rt for 0.5 h. 7-bromo-3-chloro-quinolin-2-amine (123.7 mg, 480.2 μmol, 1.5 eq.) and Pd(dppf)Cl2 (23.4 mg, 32.0 μmol, 0.1 eq.) were added. The mixture was stirred at 70° C. for 12 h under N2. The reaction progress was monitored by TLC (DCM:MeOH=10:1). Upon completion, the mixture was diluted with water (10 mL) and extracted with EA (2×10 mL). The combined organic layers were washed with brine (2×10 mL), dried over Na2SO4 and concentrated to give a crude product (600 mg). An additional reaction (100 mg of 14B) was performed using the above procedure, and 700 mg of crude product was obtained.

The crude products from two batches were combined and purified by column chromatography (SiO2, DCM:MeOH=50:1 to 30:1) to afford 7-[2-[(20S,21R,22S,23R,25R)-21-(4-aminopyrrolo[2,3-d]pyrimidin-7-yl)-24,24-dimethyl-32,33-dioxatricyclononan-25-yl]ethyl]-3-chloro-quinolin-2-amine (2P) (240 mg, 87% purity, 427.2 μmol, 66% yield, average of 2 batches) as a yellow solid. LCMS: (ESI): m/z calcd. for C26H28ClN6O2 491.19 [M+H]+, found 491.2

To a solution of 2P (240 mg, 87% purity, 427.2 μmol, 1 eq.) in THF (5 mL) was added HCl (4 M, 2.5 mL). The mixture was stirred at 25° C. for 12 h. The reaction progress was monitored by TLC (DCM:MeOH=5:1). Upon completion, the mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Gemini-NX 150*30 mm*5 um; mobile phase: [water (0.04% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 26%-56%, 8 min) to afford (1R,2R,3 S,4R,5 S)-1-[2-(2-amino-3-chloro-7-quinolyl)ethyl]-4-(4-aminopyrrolo[2,3-d]pyrimidin-7-yl)bicyclo[3.1.0]hexane-2,3-diol (19) (110 mg, 241.8 μmol, 57% yield) as a white solid. LCMS: (ESI): m/z calcd. for C23H24ClN6O2 451.16 [M+H]+, found 451.2. 1H NMR (400 MHz, CD3OD) δ: 8.08 (d, J=11.5 Hz, 2H), 7.56 (d, J=8.3 Hz, 1H), 7.45 (s, 1H), 7.21 (dd, J=1.3,8.3 Hz, 1H), 7.02 (d, J=3.5 Hz, 1H),6.57 (d, J=3.8 Hz, 1H), 4.94 (s, 1H),4.59 (s, 1H), 3.86 (d, J=6.5 Hz, 1H),3.10-2.90 (m, 2H), 2.29-2.19 (m,1H), 2.02-1.93 (m, 1H), 1.48-1.35 (m, 2H), 0.68 (br dd, J=5.4, 7.2 Hz, 1H).

Example 16 Compounds 20 and 20-A

To a solution of 7-bromoquinolin-2-amine (Q5) (4.2 g, 16.03 mmol) in DCM (50 mL) were added N-methylimidazole (7.90 g, 96.20 mmol, 7.67 mL) and pentyl carbonochloridate (7.24 g, 48.10 mmol) at 0° C. The mixture was stirred at 20° C. for 12 h. The mixture was partitioned between DCM (30 mL) and brine (30 mL). The organic phase was separated, and the aqueous phase extracted with DCM (3×30 mL). The organic layers were combined and dried over Na2SO4. The solids were removed by filtration, and the filtrate was concentrated under reduced pressure. The crude was purified by column chromatography (SiO2, PE:EA=0:1 to 20:1) to afford pentyl N-(7-bromo-2-quinolyl) carbamate (Q9) (3.2 g, 9.49 mmol, 59% yield, 100% purity) was obtained as a white solid. LCMS: (ESI): m/z calcd. for C15H18BrN2O2 337.05 [M+H]+, found 337.1.

A mixture of 7-[(3aR,4R,6R,6aS)-2,2,4-trimethyl-4-vinyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-6-yl]pyrrolo[2,3-d]pyrimidin-4-amine (9A) (1 g, 3.18 mmol) and 9-BBN dimer (1.92 g, 7.95 mmol) in THF (30 mL) was stirred at 50° C. for 1.5 h under Ar and then cooled to 20° C. A solution of K3PO4 (3.38 g, 15.90 mmol) in H2O (8 mL) was added. The mixture was stirred at 20° C. for 0.5 h. Pentyl N-(7-bromo-2-quinolyl)carbamate (1.29 g, 3.82 mmol) (Q9) and Pd(dppf)Cl2 (232.75 mg, 318.09 μmol) were added. The mixture was stirred at 60° C. for 12 h under Ar. The mixture was partitioned between EA (30 mL) and water (30 mL). The organic phase was separated, and the aqueous phase washed with EA (3×30 mL). The organic layers were combined, washed with brine (30 mL) and dried over Na2SO4. The solids were removed by filtration, and the filtrate was concentrated under reduced pressure. The crude was purified by column chromatography (SiO2, PE:EA=1:1 then DCM:MeOH=20:1) to afford pentyl (7-(2-((3aR,4S,6R,6aS)-6-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-2,2,4-trimethyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)ethyl)quinolin-2-yl)carbamate (2Z) (2.01 g, 3.14 mmol, 98% yield, 89% purity) as a yellow solid. LCMS: (ESI): m/z calcd. for C32H41N6O4 573.31 [M+H]+, found 573.5.

To a solution of pentyl N-[7-[2-[(3aR,4S,6R,6aS)-6-(4-aminopyrrolo[2,3-d]pyrimidin-7-yl)-2,2,4-trimethyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-4-yl]ethyl]-2-quinolyl]carbamate (2Z) (2.01 g, 3.14 mmol, 89% purity) in THF (10 mL) was added HCl (4 M, 8.96 mL). The mixture was stirred at 20° C. for 1 h. The mixture was concentrated under reduced pressure to afford a residue. The residue was purified by prep-HPLC (HCl condition, column: YMC-Triart Prep C18 150×40 mm×7 um; mobile phase: [water (0.225%HCl)-ACN]; B%: 22%-52%,7.7min) to afford pentyl N-[7-[2-[(1S,2R,3S,4R)-4-(4-aminopyrrolo[2,3-d]pyrimidin-7-yl)-2,3-dihydroxy-1-methyl-cyclopentyl]ethyl]-2-quinolyl]carbamate (20) (1.2 g, 1.98 mmol, 63% yield, 99% purity, 2 HCl) as an off-white solid. LCMS: (ESI): m/z calcd. for C29H37N6O4 533.28 [M+H]+, found 533.5. 1H NMR (400 MHz, CD3OD) δ: 8.78 (d, J=9.0 Hz, 1 H), 8.25 (s, 1 H), 8.06 (d, J=8.4 Hz, 1 H), 8.02 (s, 1 H), 7.71 (d, J=8.2 Hz, 1 H), 7.59 (d, J=3.5 Hz, 1 H), 7.42 (d, J=9.3 Hz, 1 H), 6.93 (d, J=3.5 Hz, 1 H), 5.08-5.20 (m, 1 H), 4.52-4.59 (m, 1 H), 4.41 (t, J=6.6 Hz, 2 H), 3.98 (d, J=6.2 Hz, 1 H), 2.89-3.10 (m, 2 H), 1.87-2.17 (m, 4 H), 1.76-1.86 (m, 2 H), 1.37-1.52 (m, 4 H), 1.27 (s, 3 H), 0.92-1.03 (m, 3 H).

Pentyl carbonochloridate (202.54 mg, 1.34 mmol, 3 eq.) was added dropwise to a solution of 6-bromoquinolin-2-amine (Q5-A) (100 mg, 448.3 μmol, 1 eq.) and 1-methylimidazole (220.8 mg, 2.69 mmol, 214 μL) in DCM (2.5 mL) at 0° C. The mixture was stirred at 25° C. for 12 h. The reaction progress was monitored by TLC (PE:EA=1:1). Upon completion, the mixture was extracted with EA (2×5 mL). The combined organic layers were washed with brine (2×5 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 4 g SepaFlash® Silica Flash Column, Eluent of 0˜10% EA:PE gradient @ 18 mL/min) to afford pentyl N-(6-bromo-2-quinolyl) carbamate (Q9-A) (130 mg, 380.9 μmol, 84% yield) as a white solid. LCMS: (ESI): m/z calcd. for C15H18BrN2O2 339.05 [M+H]+, found 338.8.

9-BBN dimer (192.45 mg, 795.21 μmol, 2.5 eq.) was added to a solution of 9A (100 mg, 318.09 μmol, 1 eq.) in THF (4 mL), and the mixture was stirred at 50° C. for 2 h under Ar. The mixture was cooled to rt, and then a solution of K3PO4 (337.59 mg, 1.59 mmol, 5 eq.) in H2O (0.4 mL) were added. The mixture was stirred for 0.5 h, and then Q9-A (128.71 mg, 381.70 μmol, 1.2 eq.) and Pd(dppf)Cl2 (23.27 mg, 31.81 μmol, 0.1 eq.) were added. The mixture was stirred at 70° C. for 12 h. The reaction progress was monitored by TLC (DCM:MeOH=10:1). Upon completion, the reaction mixture was extracted with EA (2×5 mL). The combined organic layers were washed with brine (2×5 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 12 g SepaFlash® Silica Flash Column, Eluent of 0-5% DCM/MeOH @ 30 mL/min) to afford pentyl N-[7-[2-[(3aR,4S,6R,6aS)-6-(4-aminopyrrolo[2,3-d]pyrimidin-7-yl)-2,2,4-trimethyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-4-yl]ethyl]-2-quinolyl]carbamate (2Z-A) (110 mg, 79% purity, 151.7 μmol, 47% yield) as a yellow oil. LCMS: (ESI): m/z calcd. for C32H41N6O2 573.31 [M+H]+, found 573.3.

To a solution of 2Z-A (110 mg, 79% purity, 151.7 μmol, 1 eq.) in THF (4 mL) was added HCl (4 M, 2 mL). The mixture was stirred at 25° C. for 12 h. The reaction progress was monitored by TLC (DCM:MeOH=5:1). Upon completion, the mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Gemini-NX 150*30 mm*5 um; mobile phase: [water (0.05%HCl)-ACN]; B%: 20%-42%, 5 min) to afford pentyl N-[7-[2-[(1S,2R,3S,4R)-4-(4-aminopyrrolo[2,3-d]pyrimidin-7-yl)-2,3-dihydroxy-1-methyl-cyclopentyl]ethyl]-2-quinolyl]carbamate (20-A) (68 mg, 111.6 μmol, 73% yield, 99.4% purity, 2 HCl salt) as a white solid. LCMS: (ESI): m/z calcd. for C29H37N6O4 533.28 [M+H]+, found 533.3. 1H NMR (400 MHz, CD3OD) δ: 8.81 (d, J=9.0Hz, 1H), 8.24 (s, 1H), 8.12 (d, J=8.6Hz, 1H), 8.03-7.94 (m, 2H), 7.59 (d, J=3.7 Hz, 1H), 7.43 (d, J=9.3Hz, 1H), 6.93 (d, J=3.7 Hz, 1H), 5.14 (td, J=8.3, 10.7 Hz, 1H), 4.58-4.52 (m, 1H), 4.42 (t, J=6.6 Hz, 2H), 3.98 (d, J=6.4 Hz, 1H), 3.05-2.87 (m, 2H), 2.15-1.98 (m, 2H), 1.92 (dt, J=5.4, 11.5 Hz, 2H), 1.86-1.77 (m, 2H), 1.52-1.38 (m, 4H), 1.26 (s, 3H), 1.00-0.94 (m, 3H).

Example 17 Compounds 21 and 21-A

t-BuOK (951 mg, 8.48 mmol) was added to a mixture of 4-chloro-7-fluoro-1H-pyrrolo[3,2-c]pyridine (1Q-a) (1.55 g, 9.08 mmol) and DMF (15 mL). The mixture was stirred at rt for 30 min. Then [(3aR,4R,6S,6aR)-2,2,4-trimethyl-4-vinyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-6-yl] trifluoromethanesulfonate (7A) (2.0 g, 6.05 mmol) was added. The mixture was stirred at rt for 19 h. The mixture was diluted with water (20 mL) and then extracted with EA (2×20 mL). The organic layers were combined, washed with brine (10 mL) and dried over anhydrous Na2SO4. The solids were removed by filtration, and the filtrate was concentrated to afford a residue. The residue was purified by reversed-phase HPLC (A: 0.04% NH3.H2O, B: CH3OH) to afford 4-chloro-7-fluoro-1-((3aS,4R,6R,6aR)-2,2,6-trimethyl-6-vinyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)-1H-pyrrolo[3,2-c]pyridine (2Q) (710 mg, 33% yield) as a yellow gum. LCMS: (ESI): m/z calcd. for C18H21ClFN2O2 351.13 [M+H]+, found 351.1.

A mixture of 2Q (70.2 mg, 0.2 mmol), diphenylmethanimine (54.4 mg, 0.300 mmol, 0.050 mL), BINAP (24.9 mg, 40.0 μmol), Pd2(dba)3 (18.3 mg, 0.020 mmol) and t-BuONa (38.4 mg, 0.400 mmol) in toluene (2 mL) was degassed under vacuum and purged with Ar. The mixture was stirred at 110° C. for 18 h. The reaction was quenched with NH4Cl (sat., aq., 1 mL). The mixture was diluted with EA (20 mL) and washed with brine (10 mL). The organic layer was dried over anhydrous Na2SO4 and concentrated to afford a crude imine intermediate. The crude intermediate was dissolved in CH3OH (3 mL). Hydroxylamine (64 mg, 50% solution in water) was added at rt. The mixture was stirred at rt for 1 h. The mixture was concentrated under reduced pressure. The residue was purified by silica gel chromatography (PA:EA=2:1, 200 mL; then DCM:MeOH=20:1) to afford 1-[(3aR,4R,6R,6aS)-2,2,4-trimethyl-4-vinyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-6-yl]-7-fluoro-pyrrolo[3,2-c]pyridin-4-amine (3Q) (40 mg, 60% yield over 2 steps) as a yellow gum. LCMS: (ESI): m/z calcd. for C18H23FN3O2 332.18 [M+H]+, found 332.1.

9-BBN dimer (255.61 mg, 1.06 mmol) was added to a solution of 3Q (140 mg, 422.47 μmol) in THF (4 mL). The mixture was stirred at 50° C. for 2 h under Ar. The mixture was cooled to 25° C., and then a solution of K3PO4 (448.38 mg, 2.11 mmol) in H2O (0.4 mL) were added. The mixture was stirred for 0.5 h. 7-bromoquinolin-2-amine (Q5) (122.51 mg, 549.21 μmol) and Pd (dppf)Cl2 (30.91 mg, 42.25 μmol) were added. The mixture was stirred at 70° C. for 12 h under Ar. The mixture was filtered and concentrated under reduced pressure. The residue was purified by reverse-phase column (C18: 0%˜70% water (0.5 mL NH3.H2O in 1L H2O)/CH3CN at 40 mL/min) to afford 7-[2-[(3aR,4S,6R,6aS)-6-(4-amino-7-fluoro-pyrrolo[3,2-c]pyridin-1-yl)-2,2,4-trimethyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-4-yl]ethyl]quinolin-2-amine (4P) (110 mg, 0.212 mmol, 50% yield, 92% purity) as a yellow solid. LCMS: (ESI): m/z calcd. for C27H31FN5O2 476.24 [M+H]+, found 476.2.

HCl (4 M, 2 mL) was added to a solution of 4P (110 mg, 212.80 μmol) in THF (4 mL). The mixture was stirred at 25° C. for 12 h. The mixture was filtered and then concentrated under reduced pressure. The residue was purified by prep-HPLC (column: Phenomenex Gemini-NX 150×30 mm×5 um; mobile phase: [water (0.05%HCl)-ACN]; B%: 5%-35%,7 min) to afford (1S,2R,3S,5R)-5-(4-amino-7-fluoro-pyrrolo[3,2-c]pyridin-1-yl)-3-[2-(2-amino-7-quinolyl)ethyl]-3-methyl-cyclopentane-1,2-diol (21) as a hydrochloride salt (white solid, 62 mg, 120.46 μmol, 56% yield, 98.78% purity). LCMS: (ESI): m/z calcd. for C24H27FN5O2. 436.21 [M+H]+, found 436.2. 1H NMR (400 MHz, CDCl3) δ: 8.31 (d, J=9.3Hz, 1H), 7.82 (d, J=8.2 Hz, 1H),7.73 (d, J=3.5 Hz, 1H), 7.60 (d, J=6.4 Hz, 1H), 7.53 (s, 1H), 7.45(dd, J=1.3, 8.1 Hz, 1H), 7.13 (dd, J=2.0, 3.2 Hz, 1H), 7.01 (d, J=9.3Hz, 1H), 5.19-5.11 (m, 1H), 4.45-4.40 (m, 1H), 3.93 (d, J=6.4 Hz, 1H), 2.98-2.82 (m, 2H), 2.17 (dd, J=8.2, 12.9 Hz, 1H), 1.95-1.80 (m,3H), 1.24 (s, 3H).

9-BBN dimer (182.6 mg, 754.4 μmol, 2.5 eq.) was added to a solution of 3Q (100 mg, 301.8 μmol, 1 eq.) in THF (4 mL), and the mixture was stirred at 50° C. for 2 h under Ar. The mixture was cooled to 25° C., and then a solution of K3PO4 (320.3 mg, 1.51 mmol, 5 eq.) in H2O (0.4 mL) were added. The mixture was stirred for 0.5 h. 7-bromoquinolin-2-amine (Q5-A) (87.5 mg, 392.3 μmol, 1.3 eq.) and Pd (dppf)Cl2 (22.1 mg, 30.2 μmol, 0.1 eq.) were added. The mixture was stirred at 70° C. for 12 h under Ar. The reaction progress was monitored by TLC (DCM:MeOH=10:1). Upon completion, the mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by reversed-phase HPLC (40 g C-18 column: chromatography (10%˜70% water(0.5 mL NH3.H2O in 1L H2O)/CH3CN @ 40 mL/min) to afford 7-[2-[(3aR,4S,6R,6aS)-6-(4-amino-7-fluoro-pyrrolo[3,2-c]pyridin-1-yl)-2,2,4-trimethyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-4-yl]ethyl]quinolin-2-amine (4P-A) (100 mg, 92.8% purity, 193.5 μmol, 64% yield) as a yellow solid. LCMS: (ESI): m/z calcd. for C27H31FN5O2 476.24 [M+H]+, found 476.3.

HCl (4 M, 2 mL) was added to a solution of 4P-A (100 mg, 92.8% purity, 193.5 μmol, 1 eq.) in THF (4 mL). The mixture was stirred at 25° C. for 12 h. The reaction progress was monitored by TLC (DCM:MeOH=10:1). Upon completion, the mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Gemini-NX 150*30 mm*5 um; mobile phase: [water (0.05%HCl)-ACN]; B%: 0%-30%, 7 min) to afford (1S,2R,3S,5R)-5-(4-amino-7-fluoro-pyrrolo[3,2-c]pyridin-1-yl)-3-[2-(2-amino-7-quinolyl)ethyl]-3-methyl-cyclopentane-1,2-diol (21-A) as a hydrochloride salt (white solid, 62 mg, 0.122 mmol, 63% yield, 100% purity). LCMS: (ESI): m/z calcd. for C24H27FN5O2 436.21 [M+H]+, found 436.3. 1H NMR (400 MHz, CDCl3) δ: 8.31 (d, J=9.3Hz, 1H), 7.76 (s, 1H), 7.75-7.69 (m, 2H), 7.62-7.58 (m, 2H), 7.14-7.12 (m, 1H), 7.06 (d, J=9.3 Hz, 1H), 5.19-5.11 (m, 1H), 4.45-4.40(m, 1H), 3.93 (d, J=6.3 Hz, 1H), 2.94-2.78 (m, 2H), 2.16 (dd, J=8.3,13.1 Hz, 1H), 1.93-1.78 (m, 3H), 1.23 (s, 3H).

Example 18 Compound 22

A mixture of 7-amino-1H-1,8-naphthyridin-2-one (1R) (1 g, 6.20 mmol, 1 eq.) and POBr3 (5.34 g, 18.61 mmol, 1.89 mL, 3 eq.) in MeCN (10 mL) was refluxed for 3 h under Ar atmosphere. The reaction progress was monitored by LCMS. Upon completion, the mixture was cooled to rt, and the reaction quenched by ice-water (20 mL). The mixture was neutralized by NH4OH to pH>8. The precipitated solid was filtered and washed with water. The filter cake was triturated with MeOH (50 mL) at rt for 30 min. The insoluble solid was filtered off. The filtrate was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, DCM:MeOH=50:1 to 20:1). 7-bromo-1,8-naphthyridin-2-amine (Q11) (90 mg, 5.6% yield, 89% purity) was obtained as a yellow solid. LCMS: (ESI): m/z calcd. for C8H7BrN3 225.97 [M+H]+, found 226.1.

7-[(12R,13 S,14R,16S)-15,15-dimethyl-16-vinyl-21,22-dioxatricyclononan-12-yl]pyrrolo[2,3-d]pyrimidin-4-amine (14B) (100 mg, 320.1 μmol, 1 eq.) in THF (5 mL) was added 9-BBN dimer (170.5 mg, 704.3 μmol, 2.2 eq.) at 20° C. The mixture was stirred at 50° C. for 60 min, and then cooled to 20° C. K3PO4 (339.8 mg, 1.60 mmol, 5 eq.) in H2O (1 mL) was added, and the mixture and stirred for 30 min. Q11 (86.1 mg, 384.17 μmol, 1.2 eq.) and Pd(dppf)Cl2 (23.4 mg, 32.01 μmol, 0.1 eq.) were added. The reaction was degassed for 3 times and then stirred at 70° C. for 15 h. The reaction progress was monitored by LCMS. Upon completion, the mixture was diluted with brine (10 mL) and extracted with EA (3×20 mL). The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by reversed-phase HPLC (0.05% NH3H2O condition) to give 7-[2-[(20R,21S,22R,24R)-20-(4-aminopyrrolo[2,3-d]pyrimidin-7-yl)-23,23-dimethyl-32,33-dioxatricyclononan-24-yl]ethyl]-1,8-naphthyridin-2-amine (3R) (81 mg, 173.4 μmol, 54.2% yield, 97.9% purity) as a yellow solid. LCMS: (ESI): m/z calcd. for C25H28N7O2 458.22 [M+H]+, found 458.2.

To a solution of 3R (81 mg, 173.4 μmol, 97.9% purity, 1 eq.) in THF (6 mL) was added HCl (4 M, 3 mL). The mixture was stirred at 20° C. for 4 h. The reaction progress was monitored by LCMS. Upon completion, the mixture was concentrated under reduced pressure to afford a residue. The residue was neutralized by NH4OH to pH>8 and then purified by prep-HPLC (column: YMC Triart C18 150*25 mm*5 um; mobile phase: [water (10 mM NH4HCO3)-ACN]; B%: 21%-51%) to afford (1R,2R,3S,4R)-1-[2-(7-amino-1,8-naphthyridin-2-yl)ethyl]-4-(4-aminopyrrolo[2,3-d]pyrimidin-7-yl)bicyclo[3.1.0]hexane-2,3-diol (22) (37 mg, 88.5 μmol, 51% yield, 99.87% purity) as a white solid. LCMS: (ESI): m/z calcd. for C22H24N7O2 418.19 [M+H]+, found 418.2. 1H NMR (400 MHz, CD3OD) δ: 8.08 (s, 1 H), 8.00 (d, J=7.8 Hz, 1 H), 7.92 (d, J=8.8 Hz, 1 H), 7.22 (d, J=7.8 Hz, 1 H), 7.06 (d, J=3.4 Hz, 1 H), 6.85 (d, J=8.8 Hz, 1 H), 6.58 (d, J=3.7 Hz, 1 H), 4.94 (s, 1 H), 4.63 (br d, J=6.8 Hz, 1 H), 3.87 (d, J=6.6 Hz, 1 H), 3.06-3.23 (m, 2 H), 2.40 (ddd, J=14.2, 9.2, 5.4 Hz, 1 H), 2.04 (ddd, J=13.9, 9.7, 7.0 Hz, 1 H), 1.34-1.45 (m, 2 H), 0.58 (br dd, J=7.0, 5.3 Hz, 1 H).

Example 19 Compound 23

Piperidine (42.6 mg, 0.5 mmol, 50 μL, 0.1 eq.) was added to a mixture of 2-amino-4-bromo-benzaldehyde (1.0 g, 5.00 mmol, 1 eq.) and malononitrile (396.3 mg, 6.0 mmol, 1.2 eq.) in EtOH (20 mL). The mixture was refluxed for 4 h, and the product precipitated as a yellow solid. The reaction progress was monitored by LCMS. Upon completion, the solid was filtered. The collected solid was washed successively with EtOH (5 mL), MeOH (10 mL) and MTBE (10 mL). The mixture was dried under high vacuum to afford 2-amino-7-bromo-quinoline-3-carbonitrile (2S) (1.2 g, 4.84 mmol, 96.8% yield) as a yellow solid, which was used in the next step without further purification. LCMS: (ESI): m/z calcd. for C10H7BrN3 249.98 [M+H]+, found 250.1.

A solution of DIBAL-H (1 M in toluene, 3.30 mL, 3.3 eq.) was added dropwise to a mixture of 2S (248.1 mg, 1.0 mmol, 1 eq.) in DCM (10 mL) at −78° C. under N2. After completion of the addition, the mixture was stirred at −78° C. for 4 h. The flask was then transformed to an ice-water bath, and the mixture was stirred at 0° C. for 30 min. The reaction was quenched by 2 M HCl to pH=3. The mixture was extracted with DCM:MeOH (10:1, 6×30 mL). The separated organic layers were combined, washed with brine (10 mL), dried over anhydrous Na2SO4, and concentrated to afford crude 2-amino-7-bromo-quinoline-3-carbaldehyde (3S) (146 mg, 44% purity) as a yellow solid, which was used in the next step without further purification.

To a solution of 3S (146 mg) in DCM (6 mL) was added DAST (562.4 mg, 3.49 mmol, 461 μL, 6 eq.) at 0° C. The mixture was stirred at rt for 16 h. The reaction progress was monitored by LCMS. Upon completion, the reaction was quenched by sat. aq. solution of NaHCO3 (10 mL). The mixture was diluted with water (10 mL) and then extracted with EA (2×20 mL). The separated organic layers were combined, washed with brine (10 mL), dried over anhydrous Na2SO4, and concentrated to afford a brown residue. The residue was purified by reversed pre-HPLC (0.05% NH3H2O condition) to afford 7-bromo-3-(difluoromethyl)quinolin-2-amine (Q7) (34 mg, 12% yield over two steps) as a yellow solid. LCMS: (ESI): m/z calcd. for C10H8BrF2N2 274.98 [M+H]+, found 274.9.

A mixture of 7-[(3aR,4R,6R,6aS)-2,2,4-trimethyl-4-vinyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-6-yl]pyrrolo[2,3-d]pyrimidin-4-amine (9A) (150 mg, 477.1 μmol, 1 eq.) and 9-BBN dimer (254.0 mg, 1.05 mmol, 2.2 eq.) in dry THF (5 mL) was stirred at 50° C. for 2 h under Ar. The mixture was cooled to rt. A solution of K3PO4 (506.40 mg, 2.39 mmol, 5 eq.) in H2O (0.5 mL) was added. The mixture was stirred at rt for 0.5 h. 7-bromo-3-(difluoromethyl)quinolin-2-amine (Q7) (169.4 mg, 620 μmol, 1.3 eq.) and Pd(dppf)Cl2 (34.9 mg, 47.7 μmol, 0.1 eq.) were added. The mixture was stirred at 70° C. for 12 h under Ar. The reaction progress was monitored by LCMS. Upon completion, the mixture was diluted with brine (15 mL) and extracted with EA (4×20 mL). The separated organic layers were combined, dried over anhydrous Na2SO4, and concentrated to afford a residue. The residue was purified by silica gel chromatography (PE:EA=1:0 to 1:1, and then DCM:MeOH=10:0 to 10:1) to afford 5S (190 mg, 367.6 μmol, 77% yield, 98.4% purity) as light yellow solid. LCMS: (ESI): m/z calcd. for C27H31F2N6O2 509.25 [M+H]+, found 509.4.

A mixture of 5S (185 mg, 363.8 μmol, 1 eq.), HCl (4 M, 2 mL), and THF (4 mL) was stirred at 20° C. for 16 h. The mixture was concentrated under high vacuum to afford a residue. The residue was dissolved in MeCN:H2O (1:1, 1 mL). MeCN (4 mL) was added dropwise to the mixture. The precipitated light yellow solid was collected by filtration and washed with MeCN:H2O (10:1, 3 mL) to afford Compound 23 as a hydrochloride salt (light yellow solid, 126 mg, 63.6% yield, 99.3% purity). LCMS: (ESI): m/z calcd. for C24H27F2N6O2 469.22 [M+H]+, found 469.2. 1H NMR (400 MHz, CD3OD) δ: 8.66 (s, 1H), 8.25 (s, 1H), 7.94 (d, J=8.3 Hz, 1H), 7.61 (s, 1H), 7.57 (d, J=3.5 Hz, 1H), 7.51 (d, J=8.3 Hz, 1H), 7.21-6.89 (m, 2H), 5.17-5.06 (m, 1H), 4.59-4.52 (m, 1H), 3.96 (d, J=6.3 Hz, 1H), 3.05-2.82 (m, 2H), 2.14-1.98 (m, 2H), 1.98-1.83 (m, 2H), 1.25 (s, 3H). 19F NMR (376 MHz, CD3OD) δ: 120.5.

Example 20 Compound 24

A mixture of 7-[(3aR,4R,6R,6aS)-2,2,4-trimethyl-4-vinyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-6-yl]pyrrolo[2,3-d]pyrimidin-4-amine (9A) (150 mg, 477.1 μmol, 1 eq.) and 9-BBN dimer (288.7 mg, 1.19 mmol, 2.5 eq.) in THF (5 mL) was stirred at 50° C. for 2 h under Ar. The mixture was then cooled to rt. A solution of K3PO4 (506.4 mg, 2.39 mmol, 5 eq.) in H2O (1 mL) was added. The mixture was stirred at rt for 30 min. 7-bromo-1, 8-naphthyridin-2-amine (Q11) (139.0 mg, 620.3 μmol, 1.3 eq.) and Pd(dppf)Cl2 (34.9 mg, 47.7 μmol, 0.1 eq.) were added. The mixture was stirred at 70° C. for 16 h under Ar. The reaction progress was monitored by LCMS. Upon completion, the mixture was diluted with H2O (10 mL) and extracted with EA (3×20 mL). The combined organic layers were washed with brine (10 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (0.05% NH3.H2O condition) to afford 2T (122 mg, 259.3 μmol, 54.4% yield, 97.7% purity) as a yellow gum. LCMS: (ESI): m/z calcd. for C25H30N7O2 460.24 [M+H]+, found 460.3.

To a mixture of 2T (122 mg, 259.3 μmol, 54.4% yield, 97.7% purity) in THF (6 mL) was added HCl (4 M, 3 mL). The mixture was stirred at rt for 12 h. The reaction progress was monitored by LCMS. Upon completion, the mixture was concentrated under reduced pressure to afford a residue. The residue was triturated with MeCN:water (10:1, 10 mL) at 20° C. for 30 min. The solid was collected by filtration and then suspended in MeCN:water (1:1, 1 mL). MeCN:water (10:1, 5 mL) was added dropwise. The solid was filtered and washed with MeCN:water (10:1, 5 mL) to afford Compound 24 as a hydrochloride salt (white solid, 83 mg, 167 μmol, 64.4% yield, and 99.1% purity). LCMS: (ESI): m/z calcd. for C22H26N7O2 420.21 [M+H]+, found 420.3. 1H NMR (400 MHz, CD3OD) δ: 8.37 (d, J=8.1 Hz, 1 H), 8.21-8.27 (m, 2 H), 7.56 (d, J=3.7 Hz, 1 H), 7.50 (d, J=8.1 Hz, 1 H), 7.07 (d, J=9.0 Hz, 1 H), 6.91 (d, J=3.7 Hz, 1 H), 5.06-5.19 (m, 1 H), 4.54 (t, J=7.0 Hz, 1 H), 3.97 (d, J=6.4 Hz, 1 H), 2.97-3.20 (m, 2 H), 1.93-2.15 (m, 4 H), 1.24 (s, 3 H).

Example 21 Compound 25-A

CbzCl (1.84 g, 10.76 mmol, 1.53 mL) was added dropwise to a solution of 1 (600 mg, 2.69 mmol) and 1-methylimidazole (1.77 g, 21.52 mmol, 1.72 mL) in DCM (15 mL) at 0° C. The mixture was stirred at 25° C. for 12 h. The reaction was quenched with NaHCO3 (sat., aq., 10 mL) and extracted with EA (2×10 mL). The combined organic layers were washed with brine (10 mL×2), dried over Na2SO4. The solids were removed by filtration, and the filtrate was concentrated under reduced pressure. The residue was purified by silica column chromatography (PE:EA=20:1 to 10:1) to afford benzyl N-(6-bromo-2-quinolyl)carbamate (Q10-A) (780 mg, 2.14 mmol, 76% yield) as a white solid. LCMS: (ESI): m/z calcd. for C17H14BrN2O2 357.02 [M+H]+, found 357.0.

A mixture of 7-[(11S,12R,135,14R,16S)-15,15-dimethyl-16-vinyl-21,22-dioxatricyclononan-12-yl]pyrrolo[2,3-d]pyrimidin-4-amine (14B) (200 mg, 0.640 mmol) and 9-BBN dimer (387.4 mg, 1.60 mmol) in THF (8 mL) was stirred at 50° C. for 2 h under Ar and then cooled to rt. A solution of K3PO4 (679.54 mg, 3.20 mmol) in H2O (0.8 mL) was added. The mixture was stirred at rt for 0.5 h. Compound Q10-A (297.3 mg, 0.832 mol) and Pd(dppf)Cl2 (46.9 mg, 0.064 mmol) were added. The mixture was stirred at 70° C. for 12 h under Ar. The mixture was filtered and concentrated under reduced pressure. The residue was purified by silica column chromatography (PE:EA=3:1 to 1:1 and DCM:MeOH=100:1 to 20:1) to afford benzyl (6-(2-((3aR,3bR,4aS,5R,5aS)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-2,2-dimethyltetrahydrocyclopropa[3,4]cyclopenta[1,2-d][1,3]dioxol-3b(3aH)-yl)ethyl)quinolin-2-yl)carbamate (2U-A) (273 mg, 0.374 mmol, 58%) as a yellow solid. LCMS: (ESI): m/z calcd. for C34H35N6O4 591.26 [M+H]+, found 591.2.

CbzCl (369.68 mg, 2.17 mmol, 0.308 mL) was added dropwise to a solution of 2U-A (320 mg, 0.542 mmol) and 1-methylimidazole (355.84 mg, 4.33 mmol, 0.345 mL) in DCM (8 mL) at 0° C. The mixture was stirred at rt for 12 h, and then quenched by addition of NaHCO3 (sat., aq., 5 mL). The mixture was extracted with DCM (3×5 mL). The combined organic layers were washed with brine, dried over Na2SO4. The solids were removed by filtration, and the filtrate was concentrated under reduced pressure. The residue was purified by silica column chromatography (PE:EA=1:1 to DCM:MeOH=50:1 to 20:1) to afford benzyl (7-((3aR,3bR,4aS,5R,5aS)-3b-(2-(2-(((benzyloxy)carbonyl)amino)quinolin-6-yl)ethyl)-2,2-dimethylhexahydrocyclopropa[3,4]cyclopenta[1,2-d][1,3]dioxol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl)carbamate (3U-A) (158 mg, 0.202 mmol, 37% yield) as a white oil. LCMS: (ESI): m/z calcd. for C42H41N6O6 725.3 [M+H]+, found 725.4.

To a solution of 3U-A (158 mg, 0.202 mmol) in THF (2 mL) was added HCl (4 M, 0.929 mL). The mixture was stirred at 25° C. for 3 h. The reaction was quenched by NH3.H2O (1 mL) and extracted with EA (2×5 mL). The combined organic layers were washed with brine (2×5 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give benzyl (7-((1S,2R,3S,4R,5R)-5-(2-(2-(((benzyloxy)carbonyl)amino)quinolin-6-yl)ethyl)-3,4-dihydroxybicyclo[3.1.0]hexan-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl)carbamate (4U-A) (143 mg, crude) as a colorless solid. LCMS: (ESI): m/z calcd. for C39H37N6O6 685.27 [M+H]+, found 685.3.

Isobutyric anhydride (160.8 mg, 1.02 mmol, 169 μL) was added to a solution of 4U-A (232 mg, 0.339 mmol), TEA (205.7 mg, 2.03 mmol, 0.283 mL) and DMAP (4.1 mg, 0.034 mmol) in DMF (5 mL). The mixture was stirred at 60° C. for 3 h, and then quenched with NaHCO3 (sat., aq., 5 mL) and extracted with EA (3×5 mL). The combined organic layers were washed with brine (2×5 mL), dried over Na2SO4. The solids were removed by filtration, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel chromatography (0˜50% EA:PE gradient) to afford (1R,2R,3S,4R,5S)-4-(4-(((benzyloxy)carbonyl)amino)-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-1-(2-(2-(((benzyloxy)carbonyl)amino)quinolin-6-yl)ethyl)bicyclo[3.1.0]hexane-2,3-diyl bis(2-methylpropanoate) (5U-A) (157 mg, 0.169 mmol, 49%) as a white solid. LCMS: (ESI): m/z calcd. for C47H49N6O8 825.35 [M+H]+, found 825.4.

To a solution of 5U-A (187 mg, 0.226 mmol) in EtOH (5 mL) and THF (2 mL) was added Pd/C (100 mg, 10% wt). The mixture was degassed under reduced pressure, purged with H2 and then stirred under H2 atmosphere (15 psi) at 25° C. for 20 h. The mixture was filtered through a Celite pad to remove the Pd/C. The filtrate was concentrated under reduced pressure to afford the residue. The residue was purified by prep-HPLC (column: YMC Triart C18 150×25 mm×5 μm; mobile phase: [water (10 mM NH4HCO3)-ACN]; B%: 42%-72%, 9.5 min) to afford (1R,2R,3S,4R,5S)-4-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-1-(2-(2-aminoquinolin-6-yl)ethyl)bicyclo[3.1.0]hexane-2,3-diyl bis(2-methylpropanoate) (25-A) (82 mg, 0.147 mmol, 65%) as a white solid. LCMS: (ESI): m/z calcd. for C31H37N6O4 557.28 [M+H]+, found 557.3. 1H NMR (400 MHz, CD3OD) δ: 8.08 (s, 1H), 7.86 (d, J=8.8 Hz, 1H), 7.48-7.41 (m, 3H), 7.01 (d, J=3.8 Hz, 1H), 6.80 (d, J=8.8 Hz, 1H), 6.60 (d, J=3.8 Hz, 1H), 5.88 (d, J=6.3 Hz, 1H), 5.23 (br d, J=7.3 Hz, 1H), 4.98 (d, J=1.8 Hz, 1H), 2.95-2.83 (m, 2H), 2.58 (quin, J=7.0, 11.3 Hz, 2H), 2.23-2.15 (m, 1H), 2.01-1.93 (m, 1H), 1.50 (dd, J=3.9, 8.4 Hz, 1H), 1.37-1.33 (m, 1H), 1.22 (d, J=7.0 Hz, 3H), 1.19-1.13 (m, 9H), 0.94-0.89 (m, 1H).

Example 22 Compounds 26 and 26-A

A mixture of 7-[(3aR,4R,6R,6aS)-2,2,4-trimethyl-4-vinyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-6-yl]pyrrolo[2,3-d]pyrimidin-4-amine (9A) (400 mg, 1.27 mmol) and 9-BBN dimer (769.82 mg, 3.18 mmol) in THF (8 mL) was stirred at 50° C. for 2 h under Ar and then cooled to rt. A solution of K3PO4 (1.35 g, 6.36 mmol) in H2O (0.8 mL) was added. The mixture was stirred for 0.5 h. Benzyl N-(7-bromo-2-quinolyl)carbamate (Q10) (545.38 mg, 1.53 mmol) and Pd(dppf)Cl2 (93.10 mg, 0.127 mmol) were added. The mixture was stirred at 70° C. for 12 h under Ar. The mixture was extracted with EA (2×10 mL). The combined organic layers were washed with brine (2×10 mL) and dried over Na2SO4. The solids were removed by filtration, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel chromatography (20%-50% EA/PE gradient to 0%˜12% DCM/CH3OH) to afford benzyl (7-(2-((3aR,4S,6R,6aS)-6-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-2,2,4-trimethyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)ethyl)quinolin-2-yl)carbamate (2V) (584 mg, 0.871 mmol, 68% yield) as a yellow solid. LCMS: (ESI): m/z calcd. for C34H37N6O4 593.28 [M+H]+, found 593.4.

Intermediate 3V was prepared similarly as described for 3U-A starting from 2V with the change that the reaction was stirred for 16 h to afford benzyl (7-((3aS,4R,6S,6aR)-6-(2-(2-(((benzyloxy)carbonyl)amino)quinolin-7-yl)ethyl)-2,2,6-trimethyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl)carbamate (3V) (360 mg, 0.492 mmol, 56%) as a white solid. LCMS: (ESI): m/z calcd. for C42H43N6O6 727.32 [M+H]+, found 727.4.

To a solution of 3V (360 mg, 492.34 μmol) in THF (4 mL) was added HCl (4 M, 2 mL). The mixture was stirred at 25° C. for 12 h. The reaction was quenched by NH3.H2O (1 mL) and extracted with EA (2×5 mL). The combined organic layers were washed with brine (5 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give benzyl (7-((1R,2S,3R,4S)-4-(2-(2-(((benzyloxy)carbonyl)amino)quinolin-7-yl)ethyl)-2,3-dihydroxy-4-methylcyclopentyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl)carbamate (4V) (328 mg, crude) as a white solid. LCMS: (ESI): m/z calcd. for C39H39N6O6 687.29 [M+H]+, found 687.3.

To a mixture of 4V (328 mg, 0.478 mmol), TEA (289.98 mg, 2.87 mmol, 0.399 mL), and DMAP (5.83 mg, 0.048 mmol) in DMF (5 mL) was added 2-methylpropanoyl 2-methylpropanoate (4V-a) (226.67 mg, 1.43 mmol, 0.238 mL). The mixture was stirred at 60° C. for 3 h. The reaction was quenched by NaHCO3 (sat., aq., 5 mL) and extracted with EA (2×5 mL). The combined organic layers were washed with brine (5 mL) and dried over Na2SO4. The solids were removed by filtration, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel chromatography (0˜50% EA/PE gradient) to afford (1S,2R,3S,5R)-5-(4-(((benzyloxy)carbonyl)amino)-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-3-(2-(2-(((benzyloxy)carbonyl)amino)quinolin-7-yl)ethyl)-3-methylcyclopentane-1,2-diyl bis(2-methylpropanoate) (5V) (295 mg, 0.320 mmol, 67% yield) as a white solid. LCMS: (ESI): m/z calcd. for C47H51N6O8 827.37 [M+H]+, found 827.4. 1H NMR (400 MHz, CDCl3) δ: 8.50 (s, 1H), 8.20-8.10 (m, 2H),7.81 (br s, 2H), 7.70 (d, J=7.8 Hz, 1H),7.62 (s, 1H), 7.44-7.34 (m, 10H), 7.30(br dd, J=1.5, 8.3 Hz, 1H), 7.14 (d, J=3.7Hz, 1H), 7.05 (d, J=3.7 Hz, 1H), 5.77(dd, J=6.0, 7.8 Hz, 1H), 5.40-5.31 (m,2H), 5.26 (s, 4H), 2.93-2.78 (m, 2H),2.65 (quin, J=7.0 Hz, 1H), 2.41 (td, J=7.0, 14.0 Hz, 1H), 2.29-2.20 (m, 1H),2.18-2.11 (m, 1H), 1.99 (br t, J=8.6 Hz, 2H), 1.29-1.25 (m, 9H), 1.04 (d, J=7.0Hz, 3H), 0.99 (d, J=7.0 Hz, 3H).

Cbz deprotection was prepared similarly as described for 5U-A, starting from 5V, to afford (1S,2R,3S,5R)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-3-(2-(2-aminoquinolin-7-yl)ethyl)-3-methylcyclopentane-1,2-diyl bis(2-methylpropanoate) (26) (105 mg, 187.46 μmol, 58%) as a white solid. LCMS: (ESI): m/z calcd. for C31H39N6O4 559.30 [M+H]+, found 559.5. 1H NMR (400 MHz, CD3OD) δ: 8.08 (s, 1H), 7.88 (d, J=9.0 Hz, 1H),7.56 (d, J=8.0 Hz, 1H), 7.39 (s, 1H),7.26 (d, J=3.5 Hz, 1H), 7.15 (dd, J=1.4,8.2 Hz, 1H), 6.76 (d, J=8.8 Hz, 1H), 6.60(d, J=3.8 Hz, 1H), 5.69 (dd, J=6.4, 7.7Hz, 1H), 5.36 (d, J=6.5 Hz, 1H), 5.34-5.28 (m, 1H), 2.89-2.76 (m, 2H), 2.71-2.61 (m, 1H), 2.46-2.37 (m, 1H), 2.19(d, J=9.3 Hz, 2H), 1.97 (t, J=8.5 Hz, 2H), 1.29 (s, 3H), 1.23 (dd, J=7.0, 8.8Hz, 6H), 1.02 (d, J=6.8 Hz, 3H), 0.97 (d, J=6.8 Hz, 3H).

A mixture of 7-[(3aR,4R,6R,6aS)-2,2,4-trimethyl-4-vinyl-3a,5,6,6a-tetrahydrocyclopenta[d] [1,3 ] dioxol-6-yl]pyrrolo[2,3-d]pyrimidin-4-amine (9A) (300 mg, 954.3 μmol, 1 eq.) and 9-BBN dimer (577.4 mg, 2.39 mmol, 2.5 eq.) in THF (8 mL) was stirred at 50° C. for 2 h under Ar and then cooled to rt. A solution of K3PO4 (1.01 g, 4.77 mmol, 5 eq.) in H2O (0.8 mL) was added. The mixture was stirred for 0.5 h. Benzyl N-(6-bromo-2-quinolyl)carbamate (Q10-a) (409.0 mg, 1.15 mmol, 1.2 eq.) and Pd(dppf)Cl2 (69.8 mg, 95.4 μmol, 0.1 eq.) were added. The mixture was stirred at 70° C. for 12 h under Ar. The reaction progress was monitored by TLC (DCM:MeOH=20:1). Upon completion, the mixture was extracted with EA (2×5 mL). The combined organic layers were washed with brine (2×5 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 20 g SepaFlash® Silica Flash Column, Eluent of 10° A-50% EA:PE gradient to 0%-10% DCM/MeOH @ 35 mL/min) to afford 2V-A (312 mg, 431.7 μmol, 45% yield) as a yellow solid. LCMS: (ESI): m/z calcd. for C34H37N6O4 593.28 [M+H]+, found 593.3.

To a solution of 2V-A (312 mg, 431.7 μmol, 1 eq.) in DCM (8 mL) were added 1-methylimidazole (283.5 mg, 3.45 mmol, 275, 8 eq.) and CbzCl (294.6 mg, 1.73 mmol, 245 μL, 4 eq.) at 0° C. The mixture was stirred at 25° C. for 16 h. The reaction was quenched by addition of sat. NaHCO3 solution (5 mL) and then extracted with DCM (3×5 mL). The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 20 g SepaFlash® Silica Flash Column, Eluent of 10%˜50% EA:PE gradient to 0%˜15% DCM/MeOH @ 35 mL/min) to afford 3V-A (178 mg, 233.9 μmol, 54% yield) as a white solid. LCMS: (ESI): m/z calcd. for C42H43N6O6 727.32 [M+H]+, found 727.3.

To a solution of 3V-A (178 mg, 233.88 μmol, 1 eq.) in THF (2 mL) was added HCl (4 M, 1 mL). The mixture was stirred at 25° C. for 12 h. The reaction progress was monitored by TLC (DCM:MeOH=20:1). Upon completion, the reaction was quenched by NH3.H2O solution (1 mL) and extracted with EA (2×5 mL). The combined organic layers were washed with brine (2×5 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give 4V-A (161 mg, crude) as a white solid. LCMS: (ESI): m/z calcd. for C39H39N6O6 687.29 [M+H]+, found 687.3.

To a mixture of 4V-A (200 mg, 291.2 μmol, 1 eq.), TEA (176.8 mg, 1.75 mmol, 243 μL, 6 eq.) and DMAP (3.6 mg, 29.1 μmol, 0.1 eq.) in DMF (5 mL) was added 2-methylpropanoyl 2-methylpropanoate (4V-a) (138.2 mg, 873.7 μmol, 145 μL, 3 eq.). The mixture was stirred at 60° C. for 3 h. The reaction progress was monitored by TLC (PE:EA=1:1). Upon completion, the reaction was quenched by sat.NaHCO3 solution (5 mL) and extracted with EA (2×5 mL). The combined organic layers were washed with brine (2×5 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 12 g SepaFlash® Silica Flash Column, Eluent of 0˜50% EA:PE gradient @ 30 mL/min) to afford 5V-A (196 mg, 208.6 μmol, 71% yield) as a white solid. LCMS: (ESI): m/z calcd. for C47H51N6O8 827.37 [M+H]+, found 827.6.

To a solution of 5V-A (213 mg, 257.6 μmol, 1 eq.) in EtOH (5 mL) and THF (2 mL) was added Pd/C (100 mg, 10% wt). The mixture was degassed under reduced pressure and purged with H2 (3×). The mixture was then stirred under H2 atmosphere (15 psi) at 25° C. for 20 h. The reaction progress was monitored by TLC (DCM:MeOH=10:1). Upon completion, the mixture was filtered through a Celite pad to remove the Pd/C. The filtrate was concentrated under reduced pressure to afford a residue. The residue was purified by prep-HPLC (column: Welch Xtimate C18 150*25 mm*5 um; mobile phase: [water (10 mM NH4HCO3)-ACN]; B%: 45%-75%, 8.5 min) to afford Compound 26-A (73 mg, 130.5 μmol, 50.7% yield, 99.9% purity) as a white solid. LCMS: (ESI): m/z calcd. for C31H39N6O4 558.30 [M+H]+, found 558.3. 1H NMR (400 MHz, CD3OD) δ: 8.07 (s, 1H), 7.89 (d, J=8.8 Hz, 1H), 7.51-7.43 (m, 3H), 7.25 (d, J=3.8 Hz, 1H), 6.80 (d, J=8.8 Hz, 1H), 6.60 (d, J=3.8 Hz, 1H), 5.70 (dd, J=6.4, 7.9 Hz, 1H), 5.38-5.28 (m, 2H), 2.88-2.74 (m, 2H), 2.67 (spt, J=7.0 Hz, 1H), 2.48-2.36 (m,1H), 2.25-2.13 (m, 2H), 1.96 (dd, J=7.8, 9.3 Hz, 2H), 1.29 (s, 3H), 1.23 (dd, J=7.0, 8.3 Hz, 6H), 1.02 (d, J=6.8 Hz, 3H), 0.97 (d, J=7.0 Hz, 3H).

Example 23 Compound 27

To a mixture of 7-[(3aR,4R,6R,6aS)-2,2,4-trimethyl-4-vinyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-6-yl]pyrrolo[2,3-d]pyrimidin-4-amine (9A) (150 mg, 477.1 μmol, 1 eq.) in THF (5 mL) was added 9-BBN dimer (254.0 mg, 1.05 mmol, 2.2 eq.) at 20° C. The mixture was stirred at 50° C. for 1.5 h under Ar, and then cooled to 20° C. A solution of K3PO4 (506.4 mg, 2.39 mmol, 5 eq.) in H2O (1 mL) was added, and the mixture was stirred for 30 min. 2-amino-7-bromo-quinoline-3-carbonitrile (142.0 mg, 572.6 μmol, 1.2 eq.) and Pd(dppf)Cl2 (34.9 mg, 47.7 μmol, 0.1 eq.) were added to the mixture at 20° C., and the mixture was degassed for several times. The mixture was then stirred at 70° C. for 16 h under Ar. The reaction progress was monitored by LCMS. Upon completion, the mixture was partitioned between EA (20 mL) and brine (10 mL). The organic phase was separated, and the aqueous phase was extracted with EA (3×20 mL). The organics were combined and dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, PE:EA:EtOH=20:20:1 to 30:70:3) to provide 2W (105 mg, 209.1 μmol, 43.8% yield, 96.3% purity) as a yellow solid. LCMS: (ESI): m/z calcd. for C27H30N7O2 484.25 [M+H]+, found 484.2.

To a solution of 2W (105 mg, 209.1 μmol, 43.8% yield, 96.3% purity, 1 eq.) in THF (6 mL) was added HCl (4 M, 3 mL), and the mixture was stirred at 20° C. for 12 h. The mixture was concentrated under reduced pressure give a residue. The residue was purified by prep-HPLC (HCl condition, column: Phenomenex Gemini-NX 150*30 mm*5 um; mobile phase: [water (0.05% HCl)-ACN]; B%: 3%-30%) followed by prep-HPLC (basic condition, Column: YMC Triart C18 150*25 mm*5 um, Condition: water (10 mM NH4HCO3)-ACN, B%: 24%-54%) to afford Compound 27 (54 mg, 120.8 μmol, 57.7% yield, 99.2% purity) as a white solid. LCMS: (ESI): m/z calcd. for C24H26N7O2 444.21 [M+H]+, found 444.3. 1H NMR (400 MHz, CD3OD) δ: 8.46 (s, 1 H), 8.08 (s, 1 H), 7.65 (d, J=8.3 Hz, 1 H), 7.43 (s, 1 H), 7.18-7.28 (m, 2 H), 6.60 (d, J=3.4 Hz, 1 H), 4.95-5.06 (m, 1 H), 4.53 (t, J=6.8 Hz, 1 H), 3.93 (d, J=6.4 Hz, 1 H), 2.72-2.95 (m, 2 H), 2.03-2.13 (m, 1 H), 1.81-2.00 (m, 3 H), 1.24 (s, 3 H).

Example 24 Compound 28

To a solution of 8A (1 g, 3.00 mmol, 1 eq.) in THF (10 mL) was added HCl (4 M, 5 mL, 6.68 eq.). The mixture was stirred at 20° C. for 16 h. The reaction progress was monitored by LCMS. Upon completion, the mixture was concentrated under reduced pressure to give a residue and then adjusted to pH=7 with NaHCO3 solution before extraction with DCM (2×10 mL). The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure to give (1S,2R,3R,5R)-5-(4-chloropyrrolo[2,3-d]pyrimidin-7-yl)-3-methyl-3-vinyl-cyclopentane-1,2-diol (2X ) (790 mg, 2.66 mmol, 88.9% yield, 99% purity) as a white solid. LCMS: (ESI): m/z calcd. for C14H17ClN3O2 294.09 [M+H]+, found 293.9.

To a solution of 2X (790 mg, 2.69 mmol, 1 eq.) in DCM (8 mL) were added 2,4,6-trimethylpyridine (651.8 mg, 5.38 mmol, 711 μL, 2 eq.), AgNO3 (685.3 mg, 4.03 mmol, 1.5 eq.) and TBSCl (445.9 mg, 2.96 mmol, 362 μL, 1.1 eq.). The mixture was stirred at 20° C. for 5 h, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 20 g SepaFlash® Silica Flash Column, Eluent of 0˜17% EA:PE ether gradient @ 35 mL/min) to give a mixture of 3X-a and 3X-b (690 mg, 3a:3b=2.5:1).

To a solution of 3X-a and 3X-b (850 mg, 2.08 mmol, 1 eq.) in DCM (10 mL) was added PCC (898.1 mg, 4.17 mmol, 2 eq.) and 4A molecular sieves (800 mg). The mixture was stirred at 20° C. for 2 h. The reaction progress was monitored by LCMS. Upon completion, the mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 12 g SepaFlash® Silica Flash Column, Eluent of 0˜25% EA:PE gradient @ 30 mL/min), and then by prep-HPLC (neutral condition: column: YMC Triart C18 150*25 mm*5 um; mobile phase: [water (10 mM NH4HCO3)-ACN]; B%: 77%-100%, 9.5 min) to give (2R,4R,5S)-5-[tert-butyl(dimethyl)silyl]oxy-4-(4-chloropyrrolo[2,3-d]pyrimidin-7-yl)-2-methyl-2-vinylcyclopentanone (4X) (540 mg, 1.32 mmol, 63.2% yield, 99% purity) as a white solid. LCMS: (ESI): m/z calcd. for C20H29ClN3O2Si 406.16 [M+H]+, found 406.3.

To a solution of 4X (540 mg, 1.33 mmol, 1 eq.) in THF (6 mL) was added bromo(methyl)magnesium (3 M, 1.33 mL, 3 eq.) at −78° C. The mixture was stirred at 0° C. for 6 h. The reaction progress was monitored by LCMS. Upon completion, the reaction was quenched by NH4Cl solution (5 mL), diluted with water (5 mL) and extracted with DCM (2×5 mL). The combined organic layers were washed with brine (2×5 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give (1R,2R,4R,5S)-5-[tert-butyl(dimethyl)silyl]oxy-4-(4-chloropyrrolo[2,3-d]pyrimidin-7-yl)-1,2-dimethyl-2-vinylcyclopentanol (5X) (480 mg, crude) as a colorless oil.

To a solution of 5X (480 mg, crude, 1 eq.) in THF (10 mL) was added HCl (4 M, 5 mL). The mixture was stirred at 20° C. for 36 h. The reaction progress was monitored by LCMS. Upon completion, the mixture was concentrated under reduced pressure to give a residue. The residue was then adjusted to pH=7 with NaHCO3 solution and extracted with DCM (2×10 mL). The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure to give (1R,2S,3R,5R)-3-(4-chloropyrrolo[2,3-d]pyrimidin-7-yl)-1,5-dimethyl-5-vinyl-cyclopentane-1,2-diol (6X) (380 mg, crude) as a purple solid.

To a solution of 6X (380 mg, crude, 1 eq.) and 2,2-dimethoxypropane (385.76 mg, 3.70 mmol, 454 μL, 3 eq.) in acetone (10 mL) was added TsOH.H2O (7.0 mg, 37.0 μmol, 0.03 eq.) at 0° C. The mixture was stirred at 25° C. for 2 h. The reaction progress was monitored by LCMS. Upon completion, the reaction was quenched by the addition NaHCO3 solution (10 mL) and extracted with DCM (2×10 mL). The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 12 g SepaFlash® Silica Flash Column, Eluent of 0˜8% EA:PE gradient @ 30 mL/min) to give 7X (280 mg, 780.8 μmol, 63% yield, 97% purity) as a colorless oil. LCMS: (ESI): m/z calcd. for C18H23ClN3O2 348.14 [M+H]+, found 348.1.

To a mixture of NH3H2O (4.55 g, 32.46 mmol, 5 mL, 25%, 45.16 eq.) and dioxane (5 mL) was added 7X (250 mg, 718.7 μmol, 1 eq.). The mixture was heated in a sealed tube at 100° C. and stirred for 48 h. The reaction progress was monitored by LCMS. Upon completion, the mixture was extracted with DCM (2×5 mL). The combined organic layers were washed with brine (2×5 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 12 g SepaFlash® Silica Flash Column, Eluent of 0˜75% EA/PE @ 30 mL/min) to give 7-[(3aR,4R,6R,6aS)-2,2,3 a,4-tetramethyl-4-vinyl-6,6a-dihydro-5H-cyclopenta[d][1,3]dioxol-6-yl]pyrrolo[2,3-d]pyrimidin-4-amine (8X) (158 mg, 476.3 μmol, 66.3% yield, 99% purity) as a colorless oil. LCMS: (ESI): m/z calcd. for C18H25N4O2 329.19 [M+H]+, found 329.3.

To a solution of 8X (158 mg, 481.1 μmol, 1 eq.) in THF (10 mL) was added 9-BBN dimer (465.8 mg, 1.92 mmol, 4 eq.) at 25° C. The mixture was stirred at 50° C. for 1 h. The mixture was then cooled to 25° C. and a solution of K3PO4 (1.02 g, 4.81 mmol, 10 eq.) in H2O (4 mL) was added. The mixture was stirred for 30 min, and then 7-bromo-3-chloro-quinolin-2-amine (161.1 mg, 625.4 μmol, 1.3 eq.) and Pd(dppf)Cl2 (35.2 mg, 48.1 μmol, 0.1 eq.) were added at 25° C. The mixture was stirred at 70° C. for 15 h. The reaction progress was monitored by LCMS. Upon completion, the reaction mixture was filtered. The filtrate was diluted with brine (10 mL) and extracted with DCM (2×10 mL). The combined organic layers dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, PE:EA=100:1 to 0:1, DCM:MeOH=50:1 to 10:1) to give 7-[2-[(3aR,4S,6R,6aS)-6-(4-aminopyrrolo[2,3-d]pyrimidin-7-yl)-2,2,3a,4-tetramethyl-6,6a-dihydro-5Hcyclopenta[d][1,3]dioxol-4-yl]ethyl]-3-chloro-quinolin-2-amine (9X) (180 mg, 284.0 μmol, 59.0% yield, 80% purity) as a yellow solid. LCMS: (ESI): m/z calcd. for C27H32ClN6O2 507.22 [M+H]+, found 507.2.

To a solution of 9X (180 mg, 284.01 μmol, 80% purity, 1 eq.) in THF (5 mL) was added HCl (4 M, 2.00 mL). The mixture was stirred at 20° C. for 12 h. The reaction progress was monitored by LCMS. Upon completion, the mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (basic condition: column: Phenomenex Gemini-NX C18 75*30 mm*3 um; mobile phase: [water (0.04% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 30%-60%, 7 min) to give Compound 28 (63 mg, 132.2 μmol, 46.6% yield, 98% purity) as a white solid. LCMS: (ESI): m/z calcd. for C24H28ClN6O2 467.19 [M+H]+, found 467.2. 1H NMR (400 MHz, CD3OD) δ: 8.07 (d, J=2.4 Hz, 2H), 7.58 (d, J=8.3 Hz, 1H), 7.45 (s, 1H), 7.20 (dd, J=1.2, 8.3 Hz, 1H), 7.17 (d, J=3.7 Hz, 1H), 6.59 (d, J=3.4 Hz, 1H), 4.96 (q, J=8.9 Hz, 1H), 4.48 (d, J=9.0 Hz, 1H), 2.87-2.71 (m, 2H), 2.23-2.13 (m, 2H), 2.00 (dt, J=4.9, 12.2 Hz, 1H), 1.79 (dt, J=5.6, 12.3 Hz, 1H), 1.23 (s, 3H), 1.20 (s, 3H).

Example 25 Compound 29

To a solution of phenylurea (1Y) (1 g, 7.3 mmol, 1 eq.) in pyridine (5 mL) was added dropwise toluenesulfonyl chloride (4.90 g, 25.7 mmol, 3.71 mL, 3.5 eq.) at 20° C. After addition, the mixture was stirred at 20° C. for 15 min. The reaction progress was monitored by TLC (PE:EA=1:1). Upon completion, the reaction was quenched by addition of ice-cooled water (30 mL) at 20° C. The precipitate formed during stirring was filtered and washed with water. The solid was collected by filtration to give a crude product. The crude product was triturated with EtOH (5 mL) at 20° C. for 30 min and then filtered to give 2Y (800 mg, 2.9 mmol, 40% yield) as a white solid. 1H NMR (400 MHz, CDCl3) δ: 7.64 (d, J=8.3 Hz, 2 H), 7.33-7.45 (m, 5 H), 7.20 (d, J=7.5 Hz, 2 H), 2.48 (s, 3 H).

To a solution of 2-amino-4-iodo-benzoic acid (3Y) (1 g, 3.8 mmol, 1 eq.), methanamine hydrochloride (308 mg, 4.6 mmol, 1.20 eq.) and DIEA (1.47 g, 11.4 mmol, 1.99 mL, 3 eq.) in DCM (12 mL) were added EDCI (729 mg, 3.8 mmol, 1 eq.) and HOBt (616 mg, 4.7 mmol, 1.2 eq.). The mixture was stirred at 20° C. for 3.5 h. The reaction progress was monitored by LCMS. Upon completion, the reaction was quenched by addition of water (15 mL). The mixture was then partitioned between EA (20 mL) and a sat. Na2CO3 solution (30 mL). The organic phase was separated, and the aqueous phase washed with EA (3×20 mL). The combined organic layers were combined, dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 20 g SepaFlash® Silica Flash Column, Eluent of 0˜50% EA:PE gradient @ 30 mL/min) to give 2-amino-4-iodo-N-methyl-benzamide (4Y) (1 g, 3.6 mmol, 95% yield, 100% purity) as a white solid. LCMS: (ESI): m/z calcd. for C8H10IN2O 276.98 [M+H]+, found 276.9.

To a solution of 4Y (300 mg, 1.1 mmol, 1 eq.) in dioxane (8 mL) were added N-cyano-4-methyl-N-phenyl-benzenesulfonamide (296 mg, 1.1 mmol, 1 eq.) and LiHMDS (1 M, 3.3 mL, 3 eq.) at 20° C. The mixture was stirred at 100° C. for 1 h. The reaction progress was monitored by LCMS. Upon completion, the reaction was quenched by water (10 mL). The aqueous phase washed with EA (3×20 mL). The organic layers were combined, washed with brine (20 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 20 g SepaFlash® Silica Flash Column, Eluent of 0˜100% EA:PE gradient @ 35 mL/min). Q15 (225 mg, 729 μmol, 67% yield, 98% purity) was obtained as a white solid. LCMS: (ESI): m/z calcd. for C9H9IN3O 301.97 [M+H]+, found 301.9.

A mixture of 7-[(3aR,4R,6R,6aS)-2,2,4-trimethyl-4-vinyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-6-yl]pyrrolo[2,3-d]pyrimidin-4-amine (9A) (150 mg, 477 μmol, 1 eq.) and 9-BBN dimer (289 mg, 1.2 mmol, 2.5 eq.) in THF (5 mL) was stirred at 50° C. for 2 h under Ar. The mixture was cooled to 20° C. and then a solution of K3PO4 (506 mg, 2.4 mmol, 5 eq.) in H2O (1 mL) was added. The mixture was then stirred for 30 min. Compound Q15 (172 mg, 573 μmol, 98% purity, 1.2 eq.) and Pd(dppf)Cl2 (35 mg, 48 μmol, 0.1 eq.) were added. The mixture was stirred at 70° C. for 16 h under Ar. The reaction progress was monitored by LCMS. Upon completion, the mixture was partitioned between EA (20 mL) and brine (10 mL). The organic phase was separated, and the aqueous phase was extracted with EA (3×20 mL). The separated organic layers were combined, dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, DCM: MeOH=20:1 to 100:7). Compound 5Y (145 mg, 277 μmol, 58% yield, 93% purity) was obtained as a yellow gum. LCMS: (ESI): m/z calcd. for C26H32N7O3 490.25 [M+H]+, found 490.2.

To a solution of 5Y (145 mg, 277 μmol, 93% purity, 1 eq.) in THF (6 mL) was added HCl (4 M, 3 mL). The mixture was stirred at 20° C. for 12 h. The reaction progress was monitored by LCMS. Upon completion, the mixture was concentrated under reduced pressure to remove solvent to give a residue. The residue was purified by prep-HPLC (HCl condition: Column: Venusil ASB Phenyl 150*30 mm*5 um, Condition: water (0.05%HCl)-ACN, B%: 10%-40%) to give Compound 29 as a hydrochloride salt (white solid, 110 mg, 207 μmol, 75% yield, 98% purity). LCMS: (ESI): m/z calcd. for C23H28N7O3 450.22 [M+H]+, found 450.2. 1H NMR (400 MHz, CD3OD) δ: 8.25 (s, 1 H), 8.09 (d, J=8.1 Hz, 1 H), 7.57 (d, J=3.7 Hz, 1 H), 7.37 (d, J=8.1 Hz, 1 H), 7.33 (s, 1 H), 6.92 (d, J=3.4 Hz, 1 H), 5.06-5.16 (m, 1 H), 4.55 (dd, J=7.5, 6.5 Hz, 1 H), 3.95 (d, J=6.1 Hz, 1 H), 3.55 (s, 3 H), 2.75-2.97 (m, 2 H), 1.96-2.13 (m, 2 H), 1.79-1.95 (m, 2 H), 1.23 (s, 3 H).

Example 26 Compound 30

NBS (8.03 g, 45.12 mmol) and BPO (1.30 g, 3.76 mmol, 70% purity) were added to a solution of 5-bromo-1-fluoro-2-methyl-3-nitrobenzene (8.8 g, 37.60 mmol) in CCl4 (130 mL) at 80° C. The mixture was stirred at 80° C. for 12 h. The mixture was extracted with EA (3×100 mL). The combined organic layers were washed with brine (2×100 mL) and dried over Na2SO4. The solids were removed by filtration, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel chromatography (0˜2% PE/EA gradient) to afford 5-bromo-2-(bromomethyl)-1-fluoro-3-nitro-benzene (12.4 g, 31.70 mmol, 84% yield) as a colorless oil. IH NMR (400 MHz, CDCl3) δ 8.02 (s, 1H), 7.58 (dd, J=1.7, 8.6 Hz, 1H), 4.76 (d, J=1.5 Hz, 2H).

4-methyl-4-oxido-morpholin-4-ium (8.91 g, 76.08 mmol, 8.0 mL) was added to a mixture of 5-bromo-2-(bromomethyl)-1-fluoro-3-nitro-benzene (12.4 g, 31.7 mmol) and 4Å molecular sieves (20 g) in MeCN (130 mL). The mixture was stirred at 25° C. for 3 h. The mixture was extracted with EA (3×50 mL). The combined organic layers were washed with water, 1M HCl and brine (2×50 mL) and dried over Na2SO4. The solids were removed by filtration, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel chromatography (0˜7% PE/EA gradient) to afford 4-bromo-2-fluoro-6-nitro-benzaldehyde (6.49 g, 26.17 mmol, 82%) as a white solid. 1H NMR (400 MHz, CDCl3) δ 10.24 (s, 1H), 8.02-7.98 (m, 1H), 7.68 (dd, J=1.8, 8.6 Hz, 1H).

Fe powder (14.61 g, 261.69 mmol) was added to a solution of 4-bromo-2-fluoro-6-nitro-benzaldehyde (6.49 g, 26.17 mmol) in EtOH (30 mL) and AcOH (30 mL) at 0° C. The mixture was stirred at 25° C. for 3 h, then diluted with EA (100 mL). The reaction was neutralized with NaHCO3 (sat., aq., 300 mL). The mixture was filtered through a Celite pad. The separated organic layer was washed with brine (3×100 mL) and dried over Na2SO4. The solids were removed by filtration, and the filtrate was concentrated under reduced pressure to afford 2-amino-4-bromo-6-fluoro-benzaldehyde (5.67 g, crude) as a light green solid. LCMS: (ESI): m/z calcd. for C7H6BrFNO 217.95 [M+H]+, found 217.8.

To a solution of 2-amino-4-bromo-6-fluoro-benzaldehyde (3 g, 13.76 mmol) and 2,2,2-trichloroacetonitrile (2.19 g, 15.14 mmol, 1.52 mL) in THF (40 mL) was added Fe (7.68 g, 137.60 mmol). The mixture was diluted with EA (20 mL) and filtered to give a filtrate. The mixture was then extracted with EA (3×30 mL). The combined organic layers were washed with brine (3×50 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (0˜12% EA/PE gradient) to give a solid, that was suspended in PE:EA (10:1, 20 mL) and stirred at 25° C. for 1 h. The solid was collected by filtration and dried under reduced pressure to afford 7-bromo-3-chloro-5-fluoro-quinolin-2-amine (Q16) (1.9 g, 6.90 mmol, 45% yield) as a yellow solid. LCMS: (ESI): m/z calcd. for C9H6BrClFN2, 276.93 [M+H]+, found 276.7.

A mixture of 7-[(3aR,4R,6R,6aS)-2,2,4-trimethyl-4-vinyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-6-yl]pyrrolo[2,3-d]pyrimidin-4-amine (9A) (150 mg, 0.477 mmol) and 9-BBN dimer (288.68 mg, 1.19 mmol) in THF (5 mL) was stirred at 50° C. for 2 h under Ar and then cooled to rt. A solution of K3PO4 (506.39 mg, 2.39 mmol) in H2O (0.5 mL) was added. The mixture was stirred for 0.5 h. Compound Q16 (197.18 mg, 715.69 μmol) and Pd(dppf)Cl2 (34.91 mg, 47.71 μmol) were added. The mixture was stirred at 70° C. for 12 h under Ar. The mixture was diluted with EA (2 mL) and extracted with EA (2×5 mL). The combined organic layers were washed with brine (3×5 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica column chromatography (PE:EA=50:1 to 1:2 to DCM:MeOH=100:1 to 20:1) to afford 7-(2-((3aR,4S,6R,6aS)-6-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-2,2,4-trimethyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)ethyl)-3-chloro-5-fluoroquinolin-2-amine (193 mg, 339.9 μmol, 71%) as a yellow solid. LCMS: (ESI): m/z calcd. for C26H29ClFN6O2 511.19 [M+H]+, found 511.3.

To a solution of 7-(2-((3aR,4S,6R,6aS)-6-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-2,2,4-trimethyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)ethyl)-3-chloro-5-fluoroquinolin-2-amine (193 mg, 0.340 mmol) in THF (4 mL) was added HCl (4 M, 2 mL). The mixture was stirred at 25° C. for 12 h. The mixture was filtered and concentrated under reduced pressure. The solid was added to MeCN:H2O (10:1, 3×10 mL) and stirred at 60° C. for 1 h. The mixture was filtered, and the collected solid was dried under reduced pressure to afford (1S,2R,3S,5R)-3-(2-(2-amino-3-chloro-5-fluoroquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-3-methylcyclopentane-1,2-diol (30) as a hydrochloride salt (white solid, 140 mg, 0.253 mmol, 74%). LCMS: (ESI): m/z calcd. for C23H25ClFN6O2 471.16 [M+H]+, found 471.2. 1H NMR (400 MHz, CD3OD) δ: 8.68 (s, 1H),8.25 (s, 1H), 7.59 (d, J=3.7 Hz, 1H),7.43 (s, 1H), 7.28 (d, J=10.9 Hz, 1H), 6.92 (d, J=3.5 Hz, 1H), 5.16-5.07 (m, 1H), 4.55 (dd, J=6.4, 7.5Hz, 1H), 3.95 (d, J=6.4 Hz, 1H),3.02-2.80 (m, 2H), 2.13-1.97 (m,2H), 1.96-1.82 (m, 2H), 1.24 (s,3H).

Example 27 Compound 31

To a solution of 6-bromoindoline-2,3-dione (5 g, 22.1 mmol) in EtOH (30 mL) was added diazomethyl(trimethyl)silane (5.1 g, 44.2 mmol) then followed by TEA (4.5 g, 44.2 mmol, 6.2 mL). The mixture was stirred at 20° C. for 18 h. The mixture was filtered, and the filter cake was washed with EtOH (30 mL) to afford 7-bromo-3-methoxy-1H-quinolin-2-one (3.1 g, 11.5 mmol, 52% yield, 94% purity) as a white solid. LCMS: (ESI): m/z calcd. for C10H9BrNO2 253.97 [M+H]+, found 253.9.

To a solution of 7-bromo-3-methoxy-1H-quinolin-2-one (1 g, 3.9 mmol) in toluene (20 mL) were added SOCl2 (37.5 g, 314.9 mmol, 22.8 mL) and DMF (28.8 mg, 0.394 mmol, 0.030 mL) at 20° C. under N2. The mixture was stirred at 80° C. for 2 h. The mixture was concentrated under reduced pressure to afford 7-bromo-2-chloro-3-methoxy-quinoline (1.05 g, 3.8 mmol, 96%) as a yellow solid, which was used for next step without further purification. LCMS: (ESI): m/z calcd. for C10H8BrClNO 273.94 [M+H]+, found 273.8.

A solution of 7-bromo-2-chloro-3-methoxy-quinoline (1.05 g, 3.8 mmol) in dioxane (20 mL) and NH3.H2O (20 mL, 25% wt) was stirred at 110° C. for 18 h in a 100 mL of sealed tube. The mixture was partitioned between brine (30 mL) and EA (30 mL). The organic phase was separated, washed with EA mL (3×30 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (EA/PE gradient). 7-bromo-3-methoxy-quinolin-2-amine (590 mg, 2.1 mmol, 54% yield, 89% purity) was obtained as a yellow solid. LCMS: (ESI): m/z calcd. for C10H10BrN2O 252.99 [M+H]+, found 253.0.

To a solution of 7-bromo-3-methoxy-quinolin-2-amine (590 mg, 2.1 mmol, 89% purity) in DCM (5 mL) was added BBr3 (1.57 g, 6.3 mmol, 0.602 mL). The mixture was stirred at 20° C. for 2 h. The mixture was diluted with DCM (30 mL) and then quenched by addition of MeOH at 0° C. The mixture was concentrated under reduced pressure to give 2-amino-7-bromo-quinolin-3-ol hydrobromide (785 mg, crude) as a brown solid, which was used for the next step without further purification. LCMS: (ESI): m/z calcd. for C9H8BrN2O 238.97 [M+H]+, found 240.9.

To a solution of 2-amino-7-bromo-quinolin-3-ol hydrobromide (685 mg, 2.1 mmol) in THF (34 mL) were added TEA (630.2 mg, 6.2 mmol, 0.867 mL) and 2-chloroacetyl chloride (304.8 mg, 2.7 mmol, 0.215 mL) at 0° C. The mixture was stirred at 20° C. for 2 h, then K2CO3 (573.8 mg, 4.2 mmol) was added. The mixture was stirred at 50° C. for 1 h. The reaction was quenched by addition of H2O (20 mL) at 20° C., and then extracted with EA (3×40 mL). The combined organic layers were washed with NaCl (aq., 30 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The crude product was triturated with EA (50 mL) at 20° C. for 30 min. The solid was filtered, and the filter cake was dried under reduced pressure to afford 7-bromo-2H-[1,4]oxazino[3,2-b]quinolin-3(4H)-one (Q14) (426 mg, 1.5 mmol, 72%) as a yellow solid. LCMS: (ESI): m/z calcd. for C11H8BrN2O2 278.97 [M+H]+, found 280.9.

A mixture of 7-[(3aR,4R,6R,6aS)-2,2,4-trimethyl-4-vinyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-6-yl]pyrrolo[2,3-d]pyrimidin-4-amine (9A) (150 mg, 472 μmol) and 9-BBN dimer (285.8 mg, 1.2 mmol) in THF (5 mL) was stirred at 50° C. for 1.5 h under Ar and then cooled to 20° C. K3PO4 (501.3 mg, 2.4 mmol) in water (1 mL) was added. The mixture was stirred at 20° C. for 0.5 h. Compound Q14 (173.3 mg, 614 μmol) and Pd(dppf)Cl2 (34.6 mg, 0.047 mmol) were added. The mixture was stirred at 70° C. for 12 h under Ar. The mixture was partitioned between EA (20 mL) and water (20 mL). The organic phase was separated, and the aqueous phase was extracted with EA (3×30 mL). The organic phases were combined and washed with brine (30 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica column chromatography (DCM/MeOH=100/1 to 100/3) to afford 7-(2-((3aR,4S,6R,6aS)-6-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-2,2,4-trimethyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)ethyl)-2H-[1,4]oxazino[3,2-b]quinolin-3(4H)-one (144 mg, 277 μmol, 58%) as a yellow solid. LCMS: (ESI): m/z calcd. for C28H31N6O4 515.23 [M+H]+, found 515.2.

To a solution of 7-(2-((3aR,4S,6R,6aS)-6-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-2,2,4-trimethyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)ethyl)-2H-[1,4]oxazino[3,2-b]quinolin-3(4H)-one (144 mg, 0.277 mmol) in THF (6 mL) was added HCl (4 M, 3 mL). The mixture was stirred at 20° C. for 12 h. The mixture was concentrated under reduced pressure. The residue was triturated with water/MeCN (10/1) at 20° C. for 1 h and then filtered to afford 7-(2-((1S,2R,3S,4R)-4-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-2,3-dihydroxy-1-methylcyclopentyl)ethyl)-2H-[1,4]oxazino[3,2-b]quinolin-3(4H)-one as a hydrochloride salt (31) (white solid, 117 mg, 0.209 mmol, 76%). LCMS: (ESI): m/z calcd. for C25H27N6O4 475.20 [M+H]+, found 475.2. 1H NMR (400 MHz, CD3OD) δ: 8.25 (s, 1 H), 7.66-7.76 (m, 3 H), 7.55 (d, J=3.4 Hz, 1 H), 7.39 (br d, J=8.1 Hz, 1 H), 6.91 (d, J=3.7 Hz, 1 H), 5.11 (q, J=8.8 Hz, 1 H), 4.76 (s, 2 H), 4.58 (t, J=7.0 Hz, 1 H), 3.95 (d, J=5.9 Hz, 1 H), 2.75-2.97 (m, 2 H), 1.99-2.13 (m, 2 H), 1.91 (dt, J=11.2, 5.5 Hz, 2 H), 1.25 (s, 3 H).

Example 28 Compound 32

To a solution of 4-chloro-7-((3aS,4R,6R,6aR)-2,2,6-trimethyl-6-vinyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)-7H-pyrrolo[2,3-d]pyrimidine (300 mg, 0.899 mmol) in dioxane (5 mL) and H2O (1 mL) were added 2,4,6-trimethyl-1,3,5,2,4,6-trioxatriborinane (676.92 mg, 2.70 mmol, 0.754 mL, 50% wt), Pd(dppf)Cl2 (65.76 mg, 0.090 mmol), and K3PO4 (953.83 mg, 4.49 mmol). The mixture was stirred at 90° C. for 12 h. The residue was diluted with water (5 mL) and extracted with EA (2×10 mL). The combined organic layers were washed with brine (10 mL), dried over Na2SO4. The solids were removed by filtration, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel chromatography (0˜25% EA/PE gradient) to afford 4-methyl-7-((3aS,4R,6R,6aR)-2,2,6-trimethyl-6-vinyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)-7H-pyrrolo[2,3-d]pyrimidine (189 mg, 597.05 μmol, 66%) as colorless oil. LCMS: (ESI): m/z calcd. for C18H24N3O2 314.18 [M+H]+, found 314.1.

A mixture of 4-methyl-7-((3aS,4R,6R,6aR)-2,2,6-trimethyl-6-vinyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)-7H-pyrrolo[2,3-d]pyrimidine (189 mg, 0.597 mmol) and 9-BBN dimer (361.24 mg, 1.49 mmol) in THF (5 mL) was stirred at 50° C. for 2 h under N2. The mixture was cooled to rt, and then a solution of K3PO4 (633.66 mg, 2.99 mmol) in H2O (0.5 mL) was added. After stirring at rt for 0.5 h, 7-bromo-3-chloro-5-fluoro-quinolin-2-amine (246.73 mg, 0.896 mmol) and Pd(dppf)Cl2 (43.69 mg, 0.060 mmol) were added. The mixture was stirred at 70° C. under N2 for 12 h. The mixture was diluted with water (10 mL) and then extracted with EA (2×10 mL). The combined organic layers were washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica column chromatography (PE:EA=3:1 to 1:1, then DCM:MeOH=100:1 to 20:1) followed by prep-HPLC purification (column: Phenomenex Gemini-NX 150×30 mm×5 μm; mobile phase: [water (0.04% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 53%-83%,8 min) to afford 3-chloro-5-fluoro-7-(2-((3aR,4S,6R,6aS)-2,2,4-trimethyl-6-(4-methyl-7H-pyrrolo[2,3-d] pyrimidin-7-yl)tetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)ethyl)quinolin-2-amine (152 mg, 291.78 μmol, 49%) as an off-white solid. LCMS: (ESI): m/z calcd. for C27H30ClFN5O2 510.20 [M+H]+, found 510.3.

To a solution of 3-chloro-5-fluoro-7-(2-((3aR,4S,6R,6aS)-2,2,4-trimethyl-6-(4-methyl-7H-pyrrolo[2,3-d]pyrimidin-7-yl)tetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)ethyl)quinolin-2-amine (152 mg, 291.78 μmol) in THF (4 mL) was added HCl (4 M, 2 mL). The mixture was stirred at 25° C. for 7 h. The mixture was concentrated under reduced pressure. The residue was washed with MeCN:H2O (10:1, 10 mL) to afford (1S,2R,3S,5R)-3-(2-(2-amino-3-chloro-5-fluoroquinolin-7-yl)ethyl)-3-methyl-5-(4-methyl-7H-pyrrolo[2,3-d]pyrimidin-7-yl)cyclopentane-1,2-diol (32) as a hydrochloride salt (white solid, 113 mg, 206.93 μmol, 71%). LCMS: (ESI): m/z calcd. for C24H26ClFN5O2 470.17 [M+H]+, found 470.3. 1H NMR (400 MHz, CD3OD) δ: 8.97 (s, 1H), 8.60 (s, 1H), 8.06 (d, J=4.0 Hz, 1H), 7.43 (s, 1H), 7.24 (d, J=10.3 Hz, 1H), 7.17 (d, J=3.8 Hz, 1H), 5.32-5.24 (m, 1H), 4.62 (dd, J=6.3, 7.8 Hz, 1H), 3.98 (d, J=6.3 Hz, 1H), 2.98 (s, 3H), 2.97-2.82 (m, 2H), 2.16-2.04 (m, 2H), 1.98-1.86 (m, 2H), 1.26 (s, 3H).

Example 29 Compound 33

To a solution of 7-bromo-3-chloroquinolin-2-amine (Q8) (150 mg, 0.583 mmol) in DCM (3 mL) were added N-methylimidazole (286.95 mg, 3.49 mmol, 0.279 mL) and pentyl carbonochloridate (263.2 mg, 1.75 mmol) at 0° C. The mixture was stirred at rt for 12 h. The mixture was partitioned between EA (5 mL) and water (5 mL). The organic phase was separated, the aqueous phase extracted with EA (5 mL). The organic layers were combined, washed with brine (5 mL) and dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica gel chromatography (5%˜9% PE/EA gradient) to afford pentyl (7-bromo-3-chloroquinolin-2-yl)carbamate (Q18) (75 mg, 0.200 mmol, 34% yield, 99.1% purity) as a pale yellow solid. LCMS: (ESI): m/z calcd. for C15H17BrClN2O2 373.01 [M+H]+, found 372.9.

A mixture of 7-((3aS,4R,6R,6aR)-2,2,6-trimethyl-6-vinyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine (9A) (170 mg, 540.74 μmol) and 9-BBN dimer (327.18 mg, 1.35 mmol) in THF (5 mL) was stirred at 50° C. for 2 h under N2 and then cooled to 20° C. A solution of K3PO4 (573.9 mg, 2.70 mmol) in water (0.5 mL) was added. The mixture was stirred at 20° C. for 0.5 h. Compound Q18 (241.2 mg, 0.649 mmol) and Pd(dppf)Cl2 (39.57 mg, 54.07 μmol) were added. The mixture was stirred at 60° C. for 2 h under Ar. The mixture was partitioned between EA (5 mL) and water (5 mL). The organic phase was separated, and the aqueous phase washed with EA (5 mL). The organic layers were combined and washed with brine (5 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, PE:EA=2:1 then DCM:MeOH=100:1 to 30:1) to afford pentyl (7-(2-((3aR,4S,6R,6aS)-6-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-2,2,4-trimethyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)ethyl)-3-chloroquinolin-2-yl)carbamate (235 mg, 0.327 mmol, 60%, 84% purity) as a yellow solid. LCMS: (ESI): m/z calcd. for C32H40ClN6O4 607.27 [M+H]+, found 607.5.

To a solution of pentyl (7-(2-((3aR,4S,6R,6aS)-6-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-2,2,4-trimethyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)ethyl)-3-chloroquinolin-2-yl)carbamate (185 mg, 257.17 μmol) in THF (4 mL) was added HCl (4 M, 2 mL). The mixture was stirred at rt for 12 h. The mixture was concentrated under reduced pressure. The residue was purified by prep-HPLC (Phenomenex Gemini-NX 80×40 mm×3 μm; mobile phase: [water (0.05% NH3 in H2O+10 mM NH4HCO3)-ACN]; B%: 39%-69%,8 min) to afford pentyl (7-(2-((1S,2R,3S,4R)-4-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-2,3-dihydroxy-1-methylcyclopentyl)ethyl)-3-chloroquinolin-2-yl)carbamate (33) (76 mg, 0.133 mmol, 52%) as a white solid. LCMS: (ESI): m/z calcd. for C29H36ClN6O4 567.24 [M+H]+, found 567.2. 1H NMR (400 MHz, CD3OD) δ: 8.38 (s, 1H), 8.07 (s, 1H), 7.82 (s, 1H), 7.78 (d, J=8.3 Hz, 1H), 7.49 (d, J=7.0 Hz, 1H), 7.24 (d, J=3.8 Hz, 1H), 6.60 (d, J=3.5 Hz, 1H), 5.07-4.97 (m, 1H), 4.53 (t, J=6.9 Hz, 1H), 4.23 (t, J=6.7 Hz, 2H), 3.95 (d, J=6.3 Hz, 1H), 3.00-2.80 (m, 2H), 2.13-2.05 (m, 1H), 2.00-1.84 (m, 3H), 1.74 (quin, J=6.9 Hz, 2H), 1.48-1.34 (m, 4H), 1.26 (s, 3H), 0.98-0.91 (m, 3H).

Example 30 Compound 34

A mixture of 1-[(3aR,4R,6R,6aS)-2,2,4-trimethyl-4-vinyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-6-yl]-7-fluoro-pyrrolo[3,2-c]pyridin-4-amine (3Q) (140 mg, 0.422 mmol) and 9-BBN dimer (255.6 mg, 1.06 mmol) in THF (5 mL) was stirred at 50° C. for 1.5 h under Ar and then cooled to 20° C. A solution of K3PO4 (448.4 mg, 2.11 mmol) in H2O (1 mL) was added. The mixture was stirred at 20° C. for 0.5 h. 7-bromo-3-chloro-5-fluoro-quinolin-2-amine (168.1 mg, 0.549 mmol) and Pd(dppf)Cl2 (30.9 mg, 0.042 mmol) were added. The mixture was stirred at 70° C. for 12 h under Ar. The mixture was partitioned between EA (10 mL) and water (10 mL). The organic phase was separated, the aqueous phase washed with EA (3×10 mL). The organic layers were combined and washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica column chromatography (PE:EA=1:1 then DCM:MeOH=20:1) to afford 7-[2-[(3aR,4S,6R,6aS)-6-(4-amino-7-fluoro-pyrrolo[3,2-c]pyridin-1-yl)-2,2,4-trimethyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-4-yl]ethyl]-3-chloro-5-fluoro-quinolin-2-amine (174 mg, 264.68 μmol, 63% yield, 80% purity) as a yellow solid. LCMS: (ESI): m/z calcd. for C27H29ClF2N5O2 528.19 [M+H]+, found 528.2.

To a solution of 7-[2-[(3aR,4S,6R,6aS)-6-(4-amino-7-fluoro-pyrrolo[3,2-c]pyridin-1-yl)-2,2,4-trimethyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-4-yl]ethyl]-3-chloro-5-fluoro-quinolin-2-amine (174 mg, 0.265 mmol, 80% purity) in THF (4 mL) was added HCl (4 M, 2 mL). The mixture was stirred at 20° C. for 2 h. The mixture was concentrated under reduced pressure to remove the solvent. The residue was purified by prep-HPLC (HCl conditions, column: Venusil ASB Phenyl 150×30 mm×5 μm; mobile phase: [water (0.05%HCl)-ACN]; B%: 17%-47%, 9 min) to afford (1S,2R,3S,5R)-3-[2-(2-amino-3-chloro-5-fluoro-7-quinolyl)ethyl]-5-(4-amino-7-fluoro-pyrrolo[3,2-c]pyridin-1-yl)-3-methyl-cyclopentane-1,2-diol (34) as a hydrochloride salt (off-white solid, 88 mg, 0.157 mmol, 59%). LCMS: (ESI): m/z calcd. for C24H25ClF2N5O2 488.16 [M+H]+, found 488.4. 1H NMR (400 MHz, CD3OD) δ: 8.68 (s, 1 H), 7.74 (d, J=3.5 Hz, 1 H), 7.60 (d, J=6.4 Hz, 1 H), 7.44 (s, 1 H), 7.29 (d, J=9.7 Hz, 1 H), 7.13 (dd, J=3.1, 2.0 Hz, 1 H), 5.09-5.20 (m, 1 H), 4.40-4.46 (m, 1 H), 3.92 (d, J=6.4 Hz, 1 H), 2.82-2.99 (m, 2 H), 2.16 (dd, J=12.9, 8.3 Hz, 1 H), 1.79-1.95 (m, 3 H), 1.23 (s, 3 H).

Example 31 Compound 35

DAST (6.54 g, 40.55 mmol, 5.4 mL) was added dropwise to a solution of [(3aS,4R,6R,6aS)-6-(4-chloropyrrolo [2,3-d]pyrimidin-7-yl)-4-hydroxy-2,2-dimethyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-4-yl]methyl benzoate (4.5 g, 10.14 mmol) in DCM (50 mL) at 0° C. The mixture was stirred at 0° C. for 1 h, and then the reaction was quenched by NaHCO3 (sat., aq., 100 mL). The mixture was extracted with DCM (2×50 mL). The separated organic layers were combined and washed with brine (20 mL) and dried over anhydrous Na2SO4. The solids were removed by filtration, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column (PE/EA=8/1) to afford ((3aS,4S,6R,6aS)-6-(4-chloro-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-4-fluoro-2,2-dimethyltetrahydro-4H-cyclopenta [d][1,3]dioxol-4-yl)methyl benzoate (880 mg, 60% pure) as a white foam. LCMS: (ESI): m/z calcd. for C22H22ClFN3O4 446.12 [M+H]+, found 446.2.

K2CO3 (53.5 mg, 386.8 μmol) was added to a solution of ((3aS,4S,6R,6aS)-6-(4-chloro-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-4-fluoro-2,2-dimethyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)methyl benzoate (880 mg, 60% pure) in MeOH (15 mL) at 25° C. The mixture was stirred at 25° C. for 1 h, and then the reaction was quenched with AcOH (60 mg) and diluted with brine (10 mL). The mixture was extracted with EA (2×50 mL). The separated organic layers were combined and dried over anhydrous Na2SO4. The solids were removed by filtration, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel chromatography (PE:EA:EtOH=30:10:1) to afford ((3aS,4S,6R,6aS)-6-(4-chloro-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-4-fluoro-2,2-dimethyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)methanol (570 mg, 60% pure) as a white foam. LCMS: (ESI): m/z calcd. for C15H18ClFN3O3 342.09 [M+H]+, found 342.1.

To a solution of ((3aS,4S,6R,6aS)-6-(4-chloro-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-4-fluoro-2,2-dimethyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)methanol (200 mg, 60% pure) in MeCN (5 mL) was added IBX (186.8 mg, 667.1 μmol). The mixture was stirred at 70° C. for 6 h. The mixture was cooled to rt and filtered. The filtrate was concentrated to afford (3aS,4R,6R,6aS)-6-(4-chloro-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-4-fluoro-2,2-dimethyltetrahydro-4H-cyclopenta[d][1,3]dioxole-4-carbaldehyde (210 mg, 46% pure) as a white foam, which was used in the next step without further purification. LCMS: (ESI): m/z calcd. for C15H18ClFN3O4 358.09 [M+H3O]+, found 358.2.

A solution of tBuOK (123.1 mg, 1.10 mmol) in THF (2 mL) was added dropwise to a mixture of methyl(triphenyl)phosphonium bromide (420.0 mg, 1.18 mmol) in toluene (6 mL) at 25° C. The mixture was stirred at 25° C. for 1 h. (3aS,4R,6R,6aS)-6-(4-chloro-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-4-fluoro-2,2-dimethyltetrahydro-4H-cyclopenta[d][1,3]dioxole-4-carbaldehyde (170 mg, 46% pure) in toluene (2 mL) was added dropwise at 0° C. After addition, the mixture was stirred at 25° C. for 16 h. NH4Cl (aq., 5 mL) was added to quench the reaction. The mixture was extracted with EA (2×20 mL). The separated organic layers were combined, washed with brine (30 mL) and dried over anhydrous Na2SO4. The solids were removed by filtration, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel chromatography (PE:EA=100:0 to 85:15) to afford 4-chloro-7-((3aS,4R,6S,6aS)-6-fluoro-2,2-dimethyl-6-vinyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)-7H-pyrrolo[2,3-d]pyrimidine (51 mg, 0.147 mmol) as a colorless gum. LCMS: (ESI): m/z calcd. for C16H18ClFN3O2 338.10 [M+H]+, found 338.1.

A mixture of 4-chloro-7-((3aS,4R,6S,6aS)-6-fluoro-2,2-dimethyl-6-vinyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)-7H-pyrrolo[2,3-d]pyrimidine (100 mg, 0.296 mmol) and NH4OH (6.7 mL, aq., 25% wt) in dioxane (10 mL) was sealed in a tube and stirred at 100° C. for 36 h. The mixture was concentrated under reduced pressure, diluted with brine (10 mL) and then extracted with EA (2×20 mL). The separated organic layers were combined and dried over anhydrous Na2SO4. The solids were removed by filtration, and the filtrate was concentrated. The residue was purified by silica gel chromatography (PE:EA=3:1, 160 mL; then DCM:MeOH=20:1, 300 mL) to afford 7-((3aS,4R,6S,6aS)-6-fluoro-2,2-dimethyl-6-vinyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine (68 mg, 0.207 mmol, 70%) as a white solid. LCMS: (ESI): m/z calcd. for C16H20FN4O2 319.15 [M+H]+, found 319.3.

To a mixture of 7-((3aS,4R,6S,6aS)-6-fluoro-2,2-dimethyl-6-vinyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine (62 mg, 195 μmol) in THF (3 mL) was added 9-BBN dimer (122.7 mg, 0.507 mmol). The mixture was stirred at 50° C. for 1 h and cooled to 20° C. A solution of K3PO4 (206.7 mg, 0.974 mmol) in H2O (0.3 mL) was added. The mixture was stirred at 20° C. for 0.5 h, and then 7-bromoquinolin-2-amine (Q5) (60.8 mg, 273 μmol) and Pd(dppf)Cl2 (14.2 mg, 0.019 mmol) were added. The mixture was stirred at 65° C. for 19 h. The mixture was diluted with brine (20 mL) and extracted with EA (6×30 mL). The separated organic layers were combined and dried over anhydrous Na2SO4. The solids were removed by filtration, and the filtrate was concentrated. The residue was purified by prep-HPLC (column: C18; mobile phase: [water (0.05% NH3H2O)-ACN]; B%: 5%-60%,15 min) to afford 7-(2-((3aS,4R,6R,6aS)-6-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-4-fluoro-2,2-dimethyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)ethyl)quinolin-2-amine (35 mg, 0.053 mmol, 27% yield, 70% purity) as white solid. LCMS: (ESI): m/z calcd. for C25H28FN6O2 463.22 [M+H]+, found 463.4.

To a solution of 7-(2-((3aS,4R,6R,6aS)-6-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-4-fluoro-2,2-dimethyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)ethyl)quinolin-2-amine (32.2 mg, 69.5% purity, 48.39 μmol) in THF (4.6 mL) was added HCl (4 M, 2.30 mL) at 25° C. The mixture was stirred at 25° C. for 16 h. The mixture was concentrated under reduced pressure. The residue was purified by pre-HPLC (column: Phenomenex Gemini-NX 150×30 mm×5 μm; mobile phase: [water (0.05%HCl)-ACN]; B%: 3%-30%, 7 min) to afford (1S,2S,3R,5R)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-3-(2-(2-aminoquinolin-7-yl)ethyl)-3-fluorocyclopentane-1,2-diol (35) as a hydrochloride salt (light yellow solid, 14 mg, 0.028 mmol, 58%). LCMS: (ESI): m/z calcd. for C22H24FN6O2 423.19 [M+H]+, found 423.2. 1H NMR (400 MHz, CD3OD) δ: 8.31 (d, J=9.3 Hz, 1H), 8.26 (s, 1H), 7.83 (d, J=8.2 Hz, 1H), 7.58 (d, J=3.7 Hz, 1H), 7.55 (s, 1H), 7.47 (dd, J=1.4, 8.2 Hz, 1H), 7.01 (d, J=9.3 Hz, 1H), 6.93 (d, J=3.7 Hz, 1H), 5.20 (ddd, J=6.1, 8.6, 11.0 Hz, 1H), 4.47 (t, J=6.6 Hz, 1H), 4.29-4.17 (m, 1H), 3.17-2.95 (m, 2H), 2.67-2.52 (m, 1H), 2.46-2.10 (m, 3H). 19 F NMR (376 MHz, CDCl3) δ: 169.91.

Example 32 Compound 36

A mixture of 7-((3aS,4R,6S,6aS)-6-fluoro-2,2-dimethyl-6-vinyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine (100 mg, 0.307 mmol) and 9-BBN dimer (185.8 mg, 767.88 μmol) in THF (5 mL) was stirred at 50° C. for 1.5 h under Ar and then cooled to 20° C. A solution of K3PO4 (326.0 mg, 1.54 mmol) in H2O (1 mL) was added. The mixture was stirred at 20° C. for 0.5 h. 7-bromo-3-chloro-5-fluoro-quinolin-2-amine (Q16) (101.6 mg, 0.369 mmol) and Pd(dppf)Cl2 (22.5 mg, 30.72 μmol) were added. The mixture was stirred at 60° C. for 12 h under Ar. The mixture was partitioned between EA (10 mL) and water (10 mL). The organic phase was separated, and the aqueous phase washed with EA (3×10 mL). The organic layers were combined, washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica column chromatography (PE:EA=1:1 then DCM:MeOH=20:1) to afford 7-(2-((3aS,4R, 6R,6aS)-6-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-4-fluoro-2,2-dimethyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)ethyl)-3-chloro-5-fluoroquinolin-2-amine (134 mg, 0.234 mmol, 76%, 90% purity) as yellow gum. LCMS: (ESI): m/z calcd. for C25H26ClF2N6O2 515.17 [M+H]+, found 515.3.

To a solution of 7-(2-((3aS,4R,6R,6aS)-6-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-4-fluoro-2,2-dimethyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)ethyl)-3-chloro-5-fluoroquinolin-2-amine (134 mg, 90% purity, 0.235 mmol) in THF (4 mL) was added HCl (4 M, 2 mL). The mixture was stirred at 25° C. for 12 h. The mixture was concentrated under reduced pressure. The residue was purified by prep-HPLC (Venusil ASB Phenyl 150×30 mm×5 μm; mobile phase: [water (0.05%HCl)-ACN]; B%: 10%-40%, 9 min) to afford (1S,2S,3R,5R)-3-(2-(2-amino-3-chloro-5-fluoroquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-3-fluorocyclopentane-1,2-diol (36) as a hydrochloride salt (off-white solid, 75 mg, 0.135 mmol, 62%). LCMS: (ESI): m/z calcd. for C22H22ClF2N6O2 475.14 [M+H]+, found 475.1. 1H NMR (400 MHz, CD3OD) δ: 8.69 (s, 1 H), 8.27 (s, 1 H), 7.59 (d, J=3.5 Hz, 1 H), 7.46 (s, 1 H), 7.31 (d, J=9.7 Hz, 1 H), 6.93 (d, J=3.7 Hz, 1 H), 5.19 (ddd, J=10.9, 8.4, 6.1 Hz, 1 H), 4.47 (t, J=6.5 Hz, 1 H), 4.18-4.29 (m, 1 H), 2.97-3.16 (m, 2 H), 2.59 (td, J=14.9, 8.6 Hz, 1 H), 2.09-2.46 (m, 3 H). 19F NMR (376 MHz, CD3OD) δ: −121.36 (br d, J=10.3 Hz, 1 F), −172.93-167.03 (m, 1 F).

Example 33 Compound 37

To a solution of 2-amino-4-bromo-6-fluoro-benzaldehyde (1 g, 4.59 mmol) and acetonitrile (376.58 mg, 9.17 mmol, 0.483 mL) in DMSO (20 mL) was added t-BuOK (1.03 g, 9.17 mmol) at 0° C. The mixture was stirred at rt for 15 min. The mixture was partitioned between EA (30 mL) and water (30 mL). The organic phase was separated, and the aqueous phase extracted with EA (30 mL). The organic layers were combined, washed with brine (50 mL) and dried over Na2SO4. The solids were removed by filtration, and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel chromatography (0%-35% PE/EA gradient) to afford 7-bromo-5-fluoroquinolin-2-amine (Q20) (715 mg, 2.97 mmol, 64%) as a yellow solid. LCMS: (ESI): m/z calcd. for C9H7BrFN2 242.97 [M+H]+, found 242.8.

A mixture of 7-((3aS,4R,6R,6aR)-2,2,6-trimethyl-6-vinyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine (9A) (100 mg, 0.318 mmol) and 9-BBN dimer (192.45 mg, 0.795 mmol) in THF (3 mL) was stirred at 50° C. for 2 h under N2 and then cooled to 20° C. A solution of K3PO4 (337.59 mg, 1.59 mmol) in water (0.4 mL) was added. The mixture was stirred at 20° C. for 0.5 h. Compound Q20 (92.01 mg, 0.382 mmol) and Pd(dppf)Cl2 (23.27 mg, 0.032 mmol) were added. The mixture was stirred at 60° C. for 12 h under Ar. The mixture was partitioned between EA (10 mL) and water (5 mL). The organic phase was separated, and the aqueous phase washed with EA (10 mL). The organic layers were combined, washed with brine (5 mL), dried over Na2SO4, filtered and concentrated under reduced pressure. The residue was purified by silica column chromatography (PE:EA=2:1 to 1:1 then DCM:MeOH=50:1 to 10:1) to afford 7-(2-((3aR,4S,6R,6aS)-6-(4-amino-7H-pyrrolo [2,3-d] pyrimidin-7-yl)-2,2,4-trimethyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)ethyl)-5-fluoroquinolin-2-amine (149 mg, 0.300 mmol, 94% yield, 96% purity) as a white solid. LCMS: (ESI): m/z calcd. for C26H30FN6O2 477.23 [M+H]+, found 477.2.

To a solution of 7-(2-((3aR,4S,6R,6aS)-6-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-2,2,4-trimethyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)ethyl)-5-fluoroquinolin-2-amine (149 mg, 0.300 mmol) in THF (4 mL) was added HCl (4 M, 2 mL). The mixture was stirred at rt for 12 h. The mixture was concentrated under reduced pressure. The residue was triturated with CH3CN:H2O (10:1, 20 mL) at rt for 30 min. The crude product was purified by prep-HPLC (column: Phenomenex Gemini-NX 80×40 mm×3 μm; mobile phase: [water (0.05% NH3in H2O+10 mM NH4HCO3)-ACN]; B%: 20%-50%, 8 min) to afford (1S,2R,3S,5R)-3-(2-(2-amino-5-fluoroquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-3-methylcyclopentane-1,2-diol (37) (73 mg, 0.167 mmol, 56%) as a white solid. LCMS: (ESI): m/z calcd. for C23H26FN6O2 437.20 [M+H]+, found 437.2. 1H NMR (400 MHz, CD3OD-d4) δ: 8.08 (s, 1H), 8.06 (d, J=9.7 Hz, 1H), 7.24 (s, 1H), 7.19 (s, 1H), 6.87 (d, J=11.0 Hz, 1H), 6.81 (d, J=9.1 Hz, 1H), 6.60 (d, J=3.6 Hz, 1H), 5.05-4.97 (m, 1H), 4.53 (t, J=6.9 Hz, 1H), 3.93 (d, J=6.4 Hz, 1H), 2.88-2.70 (m, 2H), 2.11-2.03(m, 1H), 1.96 (d, J=10.7 Hz, 1H),1.93-1.80 (m, 2H), 1.23 (s, 3H). 19F NMR (376 MHz, CD3OD-d4) δ: −126.40 (1 F).

Example 34 Compound 38

To a solution of 4-chloro-1H-pyrrolo[3,2-c]pyridine (554.30 mg, 3.63 mmol) in DMF (15 mL) was added t-BuOK (380.47 mg, 3.39 mmol). The mixture was stirred at 25° C. for 0.5 h followed by the addition of (3aR,4S,6R,6aR)-2,2,6-trimethyl-6-vinyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl trifluoromethanesulfonate (0.8 g, 2.42 mmol). The mixture was stirred at 25° C. for 12 h. The reaction progress was monitored by TLC (PE:EA=10:1). Upon completion, the mixture was diluted with water (30 mL) and then extracted with EA (2×30 mL). The separated organic layers were combined, washed with brine (60 mL), dried over anhydrous Na2SO4, and concentrated to afford a residue. The residue was purified by flash silica gel chromatography (eluent of 0˜4% Methanol/DCM gradient) to afford 4-chloro-1-((3aS,4R,6R,6aR)-2,2,6-trimethyl-6-vinyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)-1H-pyrrolo[3,2-c]pyridine (0.29 g, 0.871 mmol, 36%) as white solid. LCMS: (ESI): RT=3.328 min, m/z calcd. for C18H22N2O2Cl 333.13, [M+H]+, found 333.1.

To a solution of 4-chloro-1-((3aS,4R,6R,6aR)-2,2,6-trimethyl-6-vinyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)-1H-pyrrolo[3,2-c]pyridine (0.29 g, 0.871 mmol) and diphenylmethanimine (236.87 mg, 1.31 mmol, 0.219 mL) in toluene (8 mL) were added BINAP (108.51 mg, 0.174 mmol), Pd2(dba)3 (79.79 mg, 0.087 mmol) and t-BuONa (167.48 mg, 1.74 mmol). The mixture was stirred at 110° C. for 18 h under N2. The reaction was quenched with NH4Cl (sat., aq., 10 mL). The mixture was extracted with EA (3×10 mL). The separated organic layers were combined, washed with brine (30 mL), dried over anhydrous Na2SO4, filtered and concentrated to afford a crude imine intermediate. The crude intermediate was dissolved in MeOH (10 mL). Hydroxylamine (276.87 mg, 4.19 mmol, 50% wt in water) was added at 20° C. The mixture was stirred at 20° C. for 1 h. Upon completion of the reaction, the mixture was concentrated to dryness. The residue was purified by flash silica gel chromatography (eluent of 0˜5% Methanol/DCM gradient) to afford 1-((3aS,4R,6R,6aR)-2,2,6-trimethyl-6-vinyltetrahydro-4H-cyclopenta[d][1,3 ]dioxol-4-yl)-1H-pyrrolo [3,2-c]pyridin-4-amine (0.19 g, 69% yield over 2 steps) as a yellow semi-solid. LCMS: (ESI): RT=2.062 min, m/z calcd. for C18H24O2N3 314.18, [M+H]+, found 314.1.

To a solution of 1-[(3aR,4R,6R,6aS)-2,2,4-trimethyl-4-vinyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-6-yl]pyrrolo[3,2-c]pyridin-4-amine (100 mg, 0.319 mmol) in THF (5 mL) was added 9-BBN dimer (115.84 mg, 0.479 mmol). The mixture was stirred at 50° C. for 1 h and then cooled to 30° C. A solution of K3PO4 (338.66 mg, 1.60 mmol) in H2O (0.5 mL) was added. The mixture was stirred at 30° C. for 0.5 h, followed by addition of 7-bromoquinolin-2-amine (Q5) (85.41 mg, 0.383 mmol) and Pd(dppf)Cl2 (23.35 mg, 0.032 mmol). The mixture was degassed (3×) and stirred at 60° C. for 10.5 h. Upon completion, the reaction was quenched with brine (20 mL), and then extracted with EA (3×20 mL). The combined organic layers were washed with brine (60 mL) and dried over anhydrous Na2SO4. The solids were removed by filtration, and the filtrate was concentrated under reduced pressure. The residue was purified by prep-HPLC (column: Phenomenex Gemini-NX 80×40 mm×3 um; mobile phase: [water(0.05% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 17%-47%, 8 min) to afford 7-(2-((3aR,4S,6R,6aS)-6-(4-amino-1H-pyrrolo[3,2-c]pyridin-1-yl)-2,2,4-trimethyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)ethyl)quinolin-2-amine (35 mg, 0.0744 mmol, 23%) as a brown semi-solid. LCMS: (ESI): RT=4.145 min, m/z calcd. for C27H32O2N5 458.3, [M+H]+, found 458.4.

To a solution of 7-[2-[(3aR,4S,6R,6aS)-6-(4-aminopyrrolo[3,2-c]pyridin-1-yl)-2,2,4-trimethyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-4-yl]ethyl]quinolin-2-amine (32 mg, 0.070 mmol) in THF (2 mL) was added HCl (4 M, 1 mL). The mixture was stirred at 20° C. for 4 h. Upon completion, the mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Gemini-NX 80×40 mm×3 um; mobile phase: [water (0.05% NH3H2O+10 mM NH4HCO3)-ACN]; 13%: 19%-49%, 8 min) to afford (1S,2R,3S,5R)-5-(4-amino-1H-pyrrolo[3,2-c]pyridin-1-yl)-3-(2-(2-aminoquinolin-7-yl)ethyl)-3-methylcyclopentane-1,2-diol (38) (14 mg, 0.0326 mmol, 45%) as a white solid. LCMS: (ESI): RT=2.561 min, m/z calcd. for C24H28O2N5 418.22, [M+H]+, found 418.3. 1H NMR (400 MHz, CD3OD) δ: 7.88 (d, J=8.8 Hz, 1H), 7.59-7.50 (m, 2H), 7.38 (s, 1H), 7.35 (d, J=3.5 Hz, 1H), 7.14 (dd, J=1.5, 8.2 Hz, 1H), 6.95 (d, J=6.6 Hz, 1H), 6.78-6.72 (m, 2H), 4.82-4.76 (m, 1H), 4.39-4.33 (m, 1H), 3.90 (d, J=6.2 Hz, 1H), 2.91-2.71 (m, 2H), 2.12 (dd, J=8.7, 13.1 Hz, 1H), 1.97-1.77 (m, 3H), 1.25 (s, 3H).

Example 35 Compound 39

(1S,2R,3S,5R)-5-(4-amino-1H-pyrrolo[3,2-c]pyridin-1-yl)-3-(2-(2-amino-3-chloro-5-fluoroquinolin-7-yl)ethyl)-3-methylcyclopentane-1,2-diol was prepared similarly as described for (1S,2R,3S,5R)-5-(4-amino-1H-pyrrolo[3,2-c]pyridin-1-yl)-3-(2-(2-aminoquinolin-7-yl)ethyl)-3-methylcyclopentane-1,2-diol starting from (3aR,4S,6R,6aR)-2,2,6-trimethyl-6-vinyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl trifluoromethanesulfonate in a reaction with 7-bromo-3-chloro-5-fluoro-quinolin-2-amine (Q16). LCMS: (ESI): RT=2.905 min, m/z calcd. for C24H26O2N5ClF 470.17, [M+H]+, found 470.2. 1H NMR (400 MHz, CD3OD) δ=8.18 (s, 1H), 7.54 (d, J=6.2 Hz, 1H), 7.29 (d, J=3.3 Hz, 1H), 7.23 (s, 1H), 6.92 (dd, J=1.0, 10.9 Hz, 1H), 6.88 (d, J=6.3 Hz, 1H), 6.67 (d, J=3.2 Hz, 1H), 4.82-4.73 (m, 1H), 4.35 (t, J=6.9 Hz, 1H), 3.90 (d, J=6.3 Hz, 1H), 2.90-2.70 (m, 2H), 2.10 (dd, J=8.6, 13.1 Hz, 1H), 1.95-1.75 (m, 3H), 1.23 (s, 3H). 19F NMR (376 MHz, CD3OD) δ: −125.76 (s, 1F).

Example 36 Compound 40

To a solution of (3aS,4S,6R,6aR)-2,2-dimethyl-6-vinyltetrahydro-3aH-cyclopenta[d][1,3]dioxol-4-ol (7.4 g, 40.17 mmol, 1 eq.) in DMF (40 mL) was added NaH (3.21 g, 80.33 mmol, 60% purity, 2.0 eq.) at 0° C. After stirring at 0° C. for 0.5 h, PMB-Cl (11.32 g, 72.30 mmol, 9.85 mL, 1.8 eq.) was added, and the mixture was stirred at 25° C. for 2 h. The reaction progress was monitored by TLC (PE:EtOAc=5:1). Upon completion, the reaction was quenched by addition of NH4Cl (40 mL), and then extracted with EtOAc (3×100 mL). The combined organic layers was washed with brine 300 mL (3×100 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 120 g SepaFlash® Silica Flash Column, Eluent of 0˜10% EA/PE gradient @ 45 mL/min) to afford (3aS,4S,6R,6aR)-4-((4-methoxybenzyl)oxy)-2,2-dimethyl-6-vinyltetrahydro-3aH-cyclopenta[d][1,3]dioxole (7.6 g, 24.97 mmol) as a colorless oil.

To a solution of (3aS,4S,6R,6aR)-4-((4-methoxybenzyl)oxy)-2,2-dimethyl-6-vinyltetrahydro-3aH-cyclopenta[d][1,3]dioxole (22.26 g, 73.13 mmol, 1 eq.) in a mixed solvent of THF (150 mL) and H2O (150 mL) were added K2OsO4 (2.09 g, 10.97 mmol, 0.15 eq.) and NMO (17.13 g, 146.26 mmol, 2.0 eq.). The mixture was stirred at 25° C. for 18 h. Upon completion, the reaction was quenched by the addition of sat. aq. Na2S2O3 (100 mL), and then extracted with EtOAc (3×200 mL). The combined organic layers was washed with brine (300 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, DCM:MeOH=30:1 to 10:1) to afford 1-((3aR,4R,6S,6aS)-6-((4-methoxybenzyl)oxy)-2,2-dimethyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)ethane-1,2-diol (24.3 g, 71.81 mmol, 98% yield) as a yellow oil. LCMS: (ESI): m/z calcd. for C18H26O6Na, 361.17 [M+Na]+, found 361.1

To a mixture of 1-((3aR,4R,6S,6aS)-6-((4-methoxybenzyl)oxy)-2,2-dimethyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)ethane-1,2-diol (24.3 g, 71.81 mmol, 1 eq.) in THF (50 mL) and H2O (50 mL) was added NaIO4 (15.36 g, 71.81 mmol, 3.98 mL, 1.0 eq.). The mixture was stirred at 25° C. for 1 h. Upon completion, the mixture was diluted by the addition water (50 mL), and then extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (100 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was concentrated under reduced pressure to give (17.53 g, 57.22 mmol, crude) as a colorless oil. LCMS: (ESI): m/z calcd. for C17H24O6Na, 347.2 [M+H2O+Na]+, found 347.0.

To a solution of (3aR,4S,6S,6aS)-6-((4-methoxybenzyl)oxy)-2,2-dimethyltetrahydro-3aH-cyclopenta[d][1,3]dioxole-4-carbaldehyde (17.53 g, 57.22 mmol, 1 eq.) in dioxane (20 mL) were added KOH (2 M, 57.22 mL, 2 eq.) and HCHO (37.15 g, 457.77 mmol, 34.08 mL, 37% aq solution, 8 eq.). The mixture was stirred at 25° C. for 2 h. The reaction was quenched with H2O (30 mL), and then extracted with EtOAc (3×100 mL). The combined organic layers was washed with brine (100 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give crude (3aR,6S,6aS)-4-(hydroxymethyl)-6-((4-methoxybenzyl)oxy)-2,2-dimethyltetrahydro-4H-cyclopenta[d][1,3]dioxole-4-carbaldehyde.

To a solution of crude (3aR,6S,6aS)-4-(hydroxymethyl)-6-((4-methoxybenzyl)oxy)-2,2-dimethyltetrahydro-4H-cyclopenta[d][1,3]dioxole-4-carbaldehyde in MeOH (20 mL) was added NaBH4 (6.49 g, 171.66 mmol, 3 eq.) at 0° C., and the mixture was stirred at 0° C. for 1 h. The reaction progress was monitored by TLC (PE:EtOAc=0:1). Upon completion, the reaction was quenched by sat. NH4Cl solution (30 mL), and then extracted with EtOAc (3×100 mL). The combined organic layers was washed with brine (100 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 120 g SepaFlash® Silica Flash Column, Eluent of 0˜79% EA/PE ether gradient @ 45 mL/min) to afford ((3aR,6S,6aS)-6-((4-methoxybenzyl)oxy)-2,2-dimethyltetrahydro-3aH-cyclopenta[d][1,3]dioxole-4,4-diyl)dimethanol (16 g, 47.28 mmol, 83% yield) as a colorless oil. LCMS: (ESI): m/z calcd. for C18H26O6Na, 361.17 [M+Na]+, found 361.0.

To a mixture of ((3aR,6S,6aS)-6-((4-methoxybenzyl)oxy)-2,2-dimethyltetrahydro-3aH-cyclopenta[d][1,3]dioxole-4,4-diyl)dimethanol (16 g, 47.28 mmol, 1 eq.) in DCM (100 mL) were added TEA (47.84 g, 472.82 mmol, 65.81 mL, 10 eq.) and TrtCl (15.82 g, 56.74 mmol, 1.2 eq.) at 0° C. The mixture was stirred at 25° C. for 12 h. The reaction progress was monitored by TLC (PE:EtOAc=0:1 and then toluene:EA=5:1). Upon completion, the reaction was quenched by the addition of HCl (4 M, 15 mL), and then extracted with EtOAc (3×200 mL). The combined organic layers were washed with brine (200 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel flash chromatography (ISCO®; 220 g SepaFlash® Silica Flash Column, Eluent of 0-20% EA/PE gradient @ 65 mL/min), followed by re-purification by silica gel flash chromatography (ISCO®; 120 g SepaFlash® Silica Flash Column, Eluent of 0-7% EA/toluene ether gradient @ 45 mL/min) to afford ((3aR,4R,6S,6aS)-6-((4-methoxybenzyl)oxy)-2,2-dimethyl-4-((trityloxy)methyl)tetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)methanol (12.59 g, 21.68 mmol, 46% yield) and ((3aR,4S,6S,6aS)-6-((4-methoxybenzyl)oxy)-2,2-dimethyl-4-((trityloxy)methyl)tetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)methanol (2.65 g, 4.56 mmol, 10% yield). ((3aR,4R,6S,6aS)-6-((4-methoxybenzyl)oxy)-2,2-dimethyl-4-((trityloxy)methyl)tetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)methanol: LCMS: (ESI): m/z calcd. for C37H40O6Na, 603.28 [M+Na]+, found 603.3.

To a mixture of ((3aR,4R,6S,6aS)-6-((4-methoxybenzyl)oxy)-2,2-dimethyl-4-((trityloxy)methyl)tetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)methanol (6.89 g, 11.86 mmol, 1 eq.) in EtOAc (60 mL) was added IBX (4.98 g, 17.80 mmol, 1.5 eq.). The mixture was stirred at 60° C. for 12 h. The reaction progress was monitored by TLC (toluene:EA=3:1). Upon completion, the mixture was filtered and concentrated under reduced pressure to afford (3aR,4R,6S,6aS)-6-((4-methoxybenzyl)oxy)-2,2-dimethyl-4-((trityloxy)methyl)tetrahydro-3aH-cyclopenta[d][1,3]dioxole-4-carbaldehyde (7.18 g, crude) as a colorless oil. The crude was used for the next step without further purification.

To a mixture of methyl triphenyl phosphonium bromide (24.27 g, 67.95 mmol, 5.5 eq.) and THF (200 mL) was added t-BuOK (6.93 g, 61.78 mmol, 5 eq.) at 0° C. The mixture was stirred at 0° C. for 0.5 h and then (3aR,4R,6S,6aS)-6-((4-methoxybenzyl)oxy)-2,2-dimethyl-4-((trityloxy)methyl)tetrahydro-3aH-cyclopenta[d][1,3]dioxole-4-carbaldehyde (7.15 g, 12.36 mmol, 1 eq.) was added. The mixture was stirred at 25° C. for 1 h. The reaction progress was monitored by TLC (PE:EtOAc=5:1). Upon completion, the reaction was quenched by the addition of sat. NH4Cl solution (20 mL), and then extracted with EtOAc (3×100 mL). The combined organic layers were washed with brine (3×100 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 80 g SepaFlash® Silica Flash Column, Eluent of 0-10% EA/PE gradient @ 45 mL/min) to afford (3aR,4R,6S,6aS)-6-((4-methoxybenzyl)oxy)-2,2-dimethyl-4-((trityloxy)methyl)-4-vinyltetrahydro-3aH-cyclopenta[d][1,3]dioxole (5.54 g, 9.61 mmol, 78% yield) as a colorless oil.

To a mixture of (3aR,4R,6S,6aS)-6-((4-methoxybenzyl)oxy)-2,2-dimethyl-4-((trityloxy)methyl)-4-vinyltetrahydro-3aH-cyclopenta[d][1,3]dioxole (5.54 g, 9.61 mmol, 1 eq.) in a mixed solvent of PBS buffer (pH=7.4, 10 mL) and DCM (10 mL) was added DDQ (6.54 g, 28.82 mmol, 3 eq.). The mixture was stirred at 25° C. for 2 h. The reaction progress was monitored by TLC (PE:EtOAc=5:1). Upon completion, the reaction was quenched by addition of water (20 mL), and then extracted with EtOAc (3×100 mL). The combined organic layers was washed with brine (3×100 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 80 g SepaFlash® Silica Flash Column, Eluent of 0-25% EA/PE gradient @ 45 mL/min) to afford (3aS,4S,6R,6aR)-2,2-dimethyl-6-((trityloxy)methyl)-6-vinyltetrahydro-3aH-cyclopenta[d][1,3]dioxol-4-ol (3.97 g, 7.65 mmol, 80% yield, 88% purity) as a colorless oil.

To a solution of (3aS,4S,6R,6aR)-2,2-dimethyl-6-((trityloxy)methyl)-6-vinyltetrahydro-3aH-cyclopenta[d][1,3]dioxol-4-ol (1.95 g, 4.34 mmol, 1 eq.) in DCM (20 mL) and pyridine (1.37 g, 17.35 mmol, 1.40 mL, 4 eq.) was added dropwise Tf2O (1.84 g, 6.51mmol, 1.07 mL, 1.5 eq.) at 0° C. The mixture was stirred at 0° C. for 1 h. The reaction progress was monitored by TLC (PE:EtOAc=5:1). Upon completion, the reaction was quenched by the addition of ice water (20 mL), and then extracted with DCM (3×50 mL). The combined organic layers were washed with brine (150 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was concentrated under reduced pressure to afford (3aR,4S,6R,6aR)-2,2-dimethyl-6-((trityloxy)methyl)-6-vinyltetrahydro-3 aH-cyclopenta [d][1,3]dioxol-4-yl trifluoromethanesulfonate (2.43 g, crude) as a colorless oil.

To a solution of (3aR,4S,6R,6aR)-2,2-dimethyl-6-((trityloxy)methyl)-6-vinyltetrahydro-3aH-cyclopenta[d][1,3]dioxol-4-yl trifluoromethanesulfonate (4.2 g, 7.14 mmol, 1 eq.) in DMF (50 mL) was added the potassium salt of 4-chloro-7H-pyrrolo[2,3-d]pyrimidine (7-a) (2.05 g, 10.70 mmol, 1.5 eq.). The mixture was stirred at 25° C. for 12 h. Upon completion, the mixture was diluted with H2O (50 mL) and extracted with EtOAc (3×50 mL). The combined organic layers was washed with brine (150 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 40 g SepaFlash® Silica Flash Column, Eluent of 0˜10% EA/PE gradient @ 35 mL/min) to afford 4-chloro-7-((3aS,4R,6R,6aR)-2,2-dimethyl-6-((trityloxy)methyl)-6-vinyltetrahydro-3aH-cyclopenta[d][1,3]dioxol-4-yl)-7H-pyrrolo [2,3-d]pyrimidine (1.98 g, 3.01 mmol, 44% yield, 90% purity) as a white solid. LCMS: (ESI): m/z calcd. for C36H35ClN3O3, 592.23 [M+H]+, found 592.2.

To a solution of 4-chloro-7-((3aS,4R,6R,6aR)-2,2-dimethyl-6-((trityloxy)methyl)-6-vinyltetrahydro-3aH-cyclopenta[d][1,3]dioxol-4-yl)-7H-pyrrolo[2,3-d]pyrimidine (500 mg, 0.844 mmol, 1 eq.) in THF (10 mL) was added 9-BBN dimer (226.67 mg, 1.86 mmol, 2.2 eq.). The mixture was stirred at 50° C. for 2 h under Ar, and then cooled to 25° C. A solution of K3PO4 (896.20 mg, 4.22 mmol, 5 eq.) in H2O (1 mL) was added, and the mixture was stirred for 0.5 h. 7-bromo-3-chloro-5-fluoro-quinolin-2-amine (Q16) (302.43 mg, 1.10 mmol, 1.3 eq.) and Pd(dppf)Cl2 (61.79 mg, 84.44 μmol, 0.1 eq.) were added, and the mixture was stirred at 60° C. for 12 h. The reaction progress was monitored by TLC (DCM:MeOH=10:1). Upon completion, the reaction was quenched by addition of water (20 mL), and then extracted with EtOAc (3×30 mL). The combined organic layers were washed with brine (60 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 24 g SepaFlash® Silica Flash Column, Eluent of 0˜48% EA/PE gradient @ 35 mL/min) to afford 3-chloro-7-(2-((3aR,4S,6R,6aS)-6-(4-chloro-7H-pyrrolo [2,3-d]pyrimidin-7-yl)-2,2-dimethyl-4-((trityloxy)methyl)tetrahydro-3aH-cyclopenta[d][1,3]dioxol-4-yl)ethyl)-5-fluoroquinolin-2-amine (349 mg, 0.450 mmol, 50% yield) as a yellow solid. LCMS: (ESI): m/z calcd. for C45H41Cl2FN5O3, 788.3 [M+H]+, found 788.3.

To a solution of 3-chloro-7-(2-((3aR,4S,6R,6aS)-6-(4-chloro-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-2,2-dimethyl-4-((trityloxy)methyl)tetrahydro-3aH-cyclopenta[d][1,3]dioxol-4-yl)ethyl)-5-fluoroquinolin-2-amine (340 mg, 0.431 mmol, 1 eq.) in dioxane (4 mL) was added NH3H2O (3.64 g, 29.08 mmol, 4 mL, 28% purity, 67.46 eq.). The mixture was stirred at 110° C. for 12 h in a 30 mL sealed tube. The reaction progress was monitored by TLC (DCM:MeOH=10:1). Upon completion, the residue was diluted with NH4Cl (10 mL) and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 24 g SepaFlash® Silica Flash Column, Eluent of 0˜4% MeOH/DCM @ 35 mL/min) to afford 7-(2-((3aR,4S,6R,6aS)-6-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-2,2-dimethyl-4-((trityloxy)methyl)tetrahydro-3aH-cyclopenta[d][1,3]dioxol-4-yl)ethyl)-3-chloro-5-fluoroquinolin-2-amine (200 mg, 0.247 μmol, 57% yield, 95% purity) as a white solid. LCMS: (ESI): m/z calcd. for C45H43ClFN6O3, 769.3 [M+H]+, found 769.3.

A solution of 7-(2-((3aR,4S,6R,6aS)-6-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-2,2-dimethyl-4-((trityloxy)methyl)tetrahydro-3aH-cyclopenta[d][1,3]dioxol-4-yl)ethyl)-3-chloro-5-fluoroquinolin-2-amine (200 mg, 259.98 μmol, 1 eq.) in HCl (4M, aq., 1 mL) and THF (2 mL) was stirred at 25° C. for 12 h. The reaction progress was monitored by TLC (DCM:MeOH=10:1). Upon completion, the mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Gemini-NX 80*40 mm*3 um; mobile phase: [water (0.05% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 21%-45%, 8 min) to afford (1S,2R,3S,5R)-3-(2-(2-amino-3-chloro-5-fluoroquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-3-(hydroxymethyl)cyclopentane-1,2-diol (40) (75 mg, 154.03 μmol, 59% yield, 100% purity) as a white solid. LCMS: (ESI): m/z calcd. for C23H25ClFN6O3 487.16. [M+H]+, found 487.3. 1H NMR (400 MHz, CD3OD) δ: 8.19 (s, 1H), 8.13-8.00 (m, 1H), 7.28 (s, 1H), 7.24 (d, J=3.5 Hz, 1H), 6.97 (br d, J=10.6 Hz, 1H), 6.59 (d, J=3.5 Hz, 1H), 5.06-4.99 (m, 1H), 4.67-4.62 (m, 1H), 4.03 (d, J=5.5 Hz, 1H), 3.86 (br d, J=11.2 Hz, 1H), 3.74 (br d, J=11.2 Hz, 1H), 2.87 (dt, J=5.4, 12.6 Hz, 1H), 2.81-2.66 (m, 1H), 2.17 (br dd, J=9.2, 13.3 Hz, 1H), 2.07-1.83 (m, 3H). 19F NMR (376 MHz, CD3OD) δ: −125.85 (s, 1F).

Example 37 Compound 41

(1S,2R,3S, 5R)-3-(2-(2-amino-5,6-difluoroquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-3-methylcyclopentane-1,2-diol (41) was obtained as a hydrochloride salt and was prepared similarly as described for (1S,2R,3S,5R)-3-(2-(2-amino-5-fluoroquinolin-7-yl)ethyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-3-methylcyclopentane-1,2-diol starting from 7-((3aS,4R,6R,6aR)-2,2,6-trimethyl-6-vinyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine (9A) in a reaction with 7-bromo-5,6-difluoro-quinolin-2-amine (Q21). LCMS: (ESI): m/z calcd. for C23H25F2N6O2 455.19 [M+H]+, found 455.2. 1H NMR (400 MHz, CD3OD) δ: 8.45 (d, J=9.5 Hz, 1 H), 8.25 (s, 1 H), 7.58 (d, J=3.7 Hz, 1 H), 7.43 (br d, J=4.4 Hz, 1 H), 7.13 (d, J=9.5 Hz, 1 H), 6.92 (d, J=3.7 Hz, 1 H), 5.06-5.20 (m, 1 H), 4.55 (t, J=6.9 Hz, 1 H), 3.97 (d, J=6.4 Hz, 1 H), 2.85-3.09 (m, 2 H), 1.97-2.16 (m, 2 H), 1.81-1.96 (m, 2 H), 1.26 (s, 3 H). 19F NMR (376 MHz, CDCl3) δ: −147.26 (br d, J=19.1 Hz, 1 F), −148.94 (br d, J=19.8 Hz, 1 F).

Example 38 Compound 42

(1S,2R,3S,5R)-3-(2-(2-amino-5-fluoroquinolin-7-yl)ethyl)-5-(4-amino-7-fluoro-1H-pyrrolo[3,2-c]pyridin-1-yl)-3-methylcyclopentane-1,2-diol (42) was obtained as hydrochloride salt and was prepared similarly as described for (1S,2R,3S,5R)-3-[2-(2-amino-3-chloro-5-fluoro-7-quinolyl)ethyl]-5-(4-amino-7-fluoro-pyrrolo[3,2-c]pyridin-1-yl)-3-methyl-cyclopentane-1,2-diol hydrochloride salt starting from 1-[(3aR,4R,6R,6aS)-2,2,4-trimethyl-4-vinyl-3a,5,6,6a-tetrahydrocyclopenta[d][1,3]dioxol-6-yl]-7-fluoro-pyrrolo[3,2-c]pyridin-4-amine (3Q) in a reaction with 7-bromo-3-chloro-5-fluoro-quinolin-2-amine (Q16) to afford the desired compound (106 mg, 0.199 mmol, 95%) as an off-white solid. LCMS: (ESI): RT=4.523 min, m/z calcd. for C24H26F2O2N5 454.2, [M+H]+, found 454.1. 1H NMR (400 MHz, CD3OD) δ: 8.43 (d, J=9.5Hz, 1H), 7.74 (d, J=3.3 Hz, 1H),7.60 (d, J=6.4 Hz, 1H), 7.37 (s, 1H),7.24 (d, J=10.6 Hz, 1H), 7.13 (dd, J=2.0, 3.3 Hz, 1H), 7.06 (d, J=9.5 Hz, 1H), 5.19-5.10 (m, 1H), 4.47-4.39 (m, 1H), 3.93 (d, J=6.4 Hz, 1H), 2.99-2.79 (m, 2H), 2.16 (brdd, J=8.4, 12.8 Hz, 1H), 1.95-1.79 (m, 3H), 1.23 (s, 3H).

Example 39 Compound 43

(1S,2R,3S,5R)-3-(2-(2-amino-3-chloro-5-fluoroquinolin-7-yl)ethyl)-5-(4-amino-3-fluoro-1H-pyrrolo[3,2-c]pyridin-1-yl)-3-methylcyclopentane-1,2-diol (43) was obtained similar as described for (1S,2R,3S,5R)-5-(4-amino-1H-pyrrolo[3,2-c]pyridin-1-yl)-3-(2-(2-amino-3-chloro-5-fluoroquinolin-7-yl)ethyl)-3-methylcyclopentane-1,2-diol, using 4-chloro-3-fluoro-1H-pyrrolo[3,2-c]pyridine instead of 4-chloro-1H-pyrrolo[3,2-c]pyridine. The obtained residue was purified by prep-HPLC (column: Phenomenex Gemini-NX 80*40 mm*3 um; mobile phase: [water(0.05% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 36%-62%,8 min) to afford (1S,2R,3S,5R)-3-(2-(2-amino-3-chloro-5-fluoroquinolin-7-yl)ethyl)-5-(4-amino-3-fluoro-1H-pyrrolo[3,2-c]pyridin-1-yl)-3-methylcyclopentane-1,2-diol (65 mg, 0.132 mmol) as an off-white solid. 1H NMR (400 MHz, CD3OD) δ=8.19 (s, 1H), 7.54 (d, J=6.3 Hz, 1H), 7.24 (s, 1H), 7.17 (d, J=2.5 Hz, 1H), 6.93 (d, J=10.8 Hz, 1H), 6.82 (dd, J=2.5, 6.5 Hz, 1H), 4.73 (q, J=8.8 Hz, 1H), 4.30-4.23 (m, 1H), 3.86 (d, J=6.3 Hz, 1H), 2.89-2.71 (m, 2H), 2.12-2.02 (m, 1H), 1.93-1.73 (m, 3H), 1.22 (s, 3H). 19F NMR (376 MHz, CD3OD-d4) δ=−125.79 (s, 1F), −172.16 (s, 1F).

Example 40 Compounds 44 and 45

To a solution of 3-chloro-7-(2-((3aR,4S,6R,6aS)-6-(4-chloro-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-2,2-dimethyl-4-((trityloxy)methyl)tetrahydro-3aH-cyclopenta[d][1,3]dioxol-4-yl)ethyl)-5-fluoroquinolin-2-amine (200 mg, 0.254 mmol, 1 eq.) in DCM (25 mL) were added triethylsilane (3.64 g, 31.30 mmol, 5.00 mL, 123.4 eq.) and TFA (770.00 mg, 6.75 mmol, 0.5 mL, 26.63 eq.) in DCM (5 mL) at −20° C. The mixture was stirred at −20° C. for 5 min. Upon completion, the mixture was diluted with NH4Cl (10 mL) and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (150 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue, which was purified by flash silica gel chromatography (ISCO®; 24 g SepaFlash® Silica Flash Column, Eluent of 0˜4% MeOH/DCM @ 35 mL/min) to give ((3aR,4S,6R,6aS)-4-(2-(2-amino-3-chloro-5-fluoroquinolin-7-yl)ethyl)-6-(4-chloro-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-2,2-dimethyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)methanol (113 mg, 0.207 mmol, 82% yield) as a yellow solid. LCMS: (ESI): m/z calcd. for C26H27Cl2FN5O3, 546.1 [M+H]+, found 546.1.

To a solution of ((3aR,4S,6R,6aS)-4-(2-(2-amino-3-chloro-5-fluoroquinolin-7-yl)ethyl)-6-(4-chloro-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-2,2-dimethyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)methanol (146 mg, 0.267 mmol, 1 eq.) in DMSO (1 mL) was added IBX (149.6 mg, 0.534 mmol, 2 eq.), and the mixture was stirred at 25° C. for 12 h. The reaction progress was monitored by TLC (DCM:MeOH=10:1). Upon completion, the mixture was diluted with H2O (10 mL) and extracted with EtOAc (3×20 mL, and concentrated under reduced pressure to give (3aR,4R,6R,6aS)-4-(2-(2-amino-3-chloro-5-fluoroquinolin-7-yl)ethyl)-6-(4-chloro-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-2,2-dimethyltetrahydro-4H-cyclopenta[d][1,3]dioxole-4-carbaldehyde (150 mg, crude) as a colorless oil, which was used for next step without further purification. LCMS: (ESI): m/z calcd. for C26H25O2FN5O3 544.1. [M+H]+, found 544.1.

To a solution of (3aR,4R,6R,6aS)-4-(2-(2-amino-3-chloro-5-fluoroquinolin-7-yl)ethyl)-6-(4-chloro-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-2,2-dimethyltetrahydro-4H-cyclopenta[d][1,3]dioxole-4-carbaldehyde (100 mg, 183.69 μmol, 1 eq.) in EtOH (1.6 mL) and H2O (0.2 mL) were added NaOAc (30.14 mg, 367.37 μmol, 2 eq.) and NH2OH.HCl (25.53 mg, 367.37 μmol, 2 eq.). The mixture was stirred at 25° C. for 3 h. The reaction progress was monitored by TLC (DCM:MeOH=10:1). Upon completion, the mixture was diluted with NH4Cl (10 mL) and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to afford 4-(2-(2-amino-3-chloro-5-fluoroquinolin-7-yl)ethyl)-6-(4-chloro-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-2,2-dimethyltetrahydro-4H-cyclopenta[d][1,3]dioxole-4-carbaldehyde oxime (93 mg, crude) as a white solid, which was used for next step without further purification. LCMS: (ESI): m/z calcd. for C26H26Cl2FN6O3 559.1. [M+H]+, found 559.2.

To a solution of 4-(2-(2-amino-3-chloro-5-fluoroquinolin-7-yl)ethyl)-6-(4-chloro-7H-pyrrolo [2,3-d]pyrimidin-7-yl)-2,2-dimethyltetrahydro-4H-cyclopenta[d][1,3]dioxole-4-carbaldehyde oxime (80 mg, 0.143 mmol, 1 eq.) in CH3CN (4 mL) were added CDI (115.9 mg, 0.715 mmol, 5 eq.) and Et3N (72.4 mg, 0.715 mmol, 5 eq.). The mixture was stirred at 25° C. for 4 h. The reaction progress was monitored by TLC (DCM:MeOH=10:1). Upon completion, the residue was diluted with H2O (20 mL) and extracted with EtOAc (3×30 mL). The combined organic layers were washed with brine (90 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 12 g SepaFlash® Silica Flash Column, Eluent of 0˜8% EA:PE gradient @ 20 mL/min) to give (3aR,4S, 6R, 6aS)-4-(2-(2-amino-3-chloro-5-fluoroquinolin-7-yl)ethyl)-6-(4-chloro-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-2,2-dimethyltetrahydro-4H-cyclopenta[d][1,3]dioxole-4-carbonitrile (61.4 mg, 0.110 mmol, 77% yield,) as a white solid. LCMS: (ESI): m/z calcd. for C26H24N6O2Cl2F, 541.1 [M+H]+, found 541.0.

To a solution of (3aR,4S,6R,6aS)-4-(2-(2-amino-3-chloro-5-fluoroquinolin-7-yl)ethyl)-6-(4-chloro-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-2,2-dimethyltetrahydro-4H-cyclopenta[d][1,3]dioxole-4-carbonitrile (85 mg, 0.157 mmol, 1 eq.) in dioxane (5 mL) and NH3H2O (5 mL, 25% wt) was stirred at 110° C. for 12 h in a 30 mL sealed tube. Upon completion, the mixture was concentrated. The residue was diluted with NaHCO3 (sat. aq., 5 mL) and extracted with EtOAc (3×50 mL). The combined organic layers were washed with brine (150 mL), dried over anhydrous Na2SO4, filtered, and concentrated under reduced pressure to give a residue, which was purified by flash silica gel chromatography (ISCO®; 25 g SepaFlash® Silica Flash Column, Eluent of 0˜4.7% EA:PE gradient @ 35 mL/min) to give (3aR,4S,6R,6aS)-4-(2-(2-amino-3-chloro-5-fluoroquinolin-7-yl)ethyl)-6-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-2,2-dimethyltetrahydro-4H-cyclopenta[d][1,3]dioxole-4-carbonitrile (66 mg, 0.126 mmol, 81% yield) as a white solid. LCMS: (ESI): m/z calcd. for C26H26ClFN7O2, 522.2 [M+H]+, found 522.1.

To a solution of (3aR,4S,6R,6aS)-4-(2-(2-amino-3-chloro-5-fluoroquinolin-7-yl)ethyl)-6-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-2,2-dimethyltetrahydro-4H-cyclopenta[d][1,3]dioxole-4-carbonitrile (66 mg, 0.126 mmol, 1 eq.) in THF (2 mL) was added 4 M HCl (1 mL). The mixture was stirred at 25° C. for 15 h. Upon completion, the mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Gemini-NX 80*40 mm*3 um; mobile phase: [water (0.05% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 21%-45%,8 min) to give (1S,2R,3S,4R)-1-(2-(2-amino-3-chloro-5-fluoroquinolin-7-yl)ethyl)-4-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-2,3-dihydroxycyclopentane-1-carbonitrile (44) (32 mg, 0.066 μmol, 52% yield, 99% purity) and (1R,2R,3S,4R)-1-(2-(2-amino-3-chloro-5-fluoroquinolin-7-yl)ethyl)-4-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-2,3-dihydroxycyclopentane-1-carboxamide (45) (5 mg, 0.010 μmol, 8% yield, 98% purity).

Compound 44: LCMS: (ESI): m/z calcd. for C23H22ClFN7O2 482.1. [M+H]+, found 482.2. 1H NMR (400 MHz, CD3OD) δ: 8.20 (s, 1H), 8.09 (s, 1H), 7.30 (s, 1H), 7.21 (d, J=3.6 Hz, 1H), 6.97 (d, J=10.8 Hz, 1H), 6.58 (d, J=3.6 Hz, 1H), 5.07-4.99 (m, 1H), 4.64 (dd, J=6.3, 13.5 Hz, 1H), 4.15 (d, J=5.7 Hz, 1H), 3.10-2.91 (m, 2H), 2.72 (dd, J=9.0, 13.6 Hz, 1H), 2.37 (dd, J=10.3, 13.6 Hz, 1H), 2.32-2.23 (m, 2H). 19F NMR (376 MHz, CD3OD) δ: −125.35 (s, 1F).

Compound 45: LCMS: (ESI): m/z calcd. for C23H24ClFN7O3 500.2. [M+H]+, found 500.2. 1H NMR (400 MHz, CD3OD) δ: 8.19 (s, 1H), 8.10 (s, 1H), 7.26 (s, 1H), 7.21 (d, J=3.5 Hz, 1H), 6.93 (d, J=10.8 Hz, 1H), 6.59 (d, J=3.5 Hz, 1H), 4.96-4.92 (m, 1H), 4.72 (dd, J=4.9, 7.5 Hz, 1H), 4.19 (d, J=4.6 Hz, 1H), 3.01 (dd, J=9.9, 13.6 Hz, 1H), 2.81-2.65 (m, 2H), 2.32-2.22 (m, 2H), 2.06 (dd, J=9.3, 13.6 Hz, 1H). 19F NMR (376 MHz, CD3OD) δ: −125.75 (s, 1F).

Example 41 Compound 46

To a solution of 7-((3aS,4R,6R,6aR)-2,2,6-trimethyl-6-vinyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine (500 mg, 1.59 mmol, 1 eq.) in THF (4 mL) was added 9-BBN dimer (962.3 mg, 3.98 mmol, 2.5 eq.). The mixture was stirred at 50° C. for 2 h under N2, and then cooled to rt. A solution of K3PO4 (1.69 g, 7.95 mmol, 5 eq.) in H2O (2 mL) was added. The mixture was stirred at rt for 0.5 h, and then vinyl bromide (1 M, 7 mL, 4.40 eq.) and Pd(dppf)Cl2 (116.37 mg, 0.159 mmol, 0.1 eq.) were added. The mixture was stirred at 40° C. for 16 h under N2. The mixture was then diluted with brine (10 mL) and extracted with EA (2×20 mL). The combined organic layers were combined, dried over Na2SO4, filtered and concentrated to afford a residue, which was purified by prep-HPLC (40 g C-18 column, gradient: 0%-70% CH3CN in water (1mL NH3H2O in 2L H2O) in 15 min@ 40 mL/min to give 7-((3aS,4R,6S,6aR)-6-(but-3-en-1-yl)-2,2,6-trimethyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine (142.8 mg, 0.417 mmol, 26% yield) as a brown solid. LCMS: (ESI): RT=5.117 min, m/z calcd. for C19H27O2N4 343.2, [M+H]+, found 343.3.

To a solution of 7-((3aS,4R,6S,6aR)-6-(but-3-en-1-yl)-2,2,6-trimethyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine (142.8 mg, 0.417 mmol, 1 eq.) in THF (4 mL) was added 9-BBN dimer (252.31 mg, 1.04 mmol, 2.5 eq.). The mixture was stirred at 50° C. for 2 h under N2, and then cooled to 25° C. A solution of K3PO4 (442.59 mg, 2.09 mmol, 5 eq.) in H2O (0.4 mL) was added, and the mixture was then stirred at rt for 0.5 h. 6-bromo-3-chloro-pyridin-2-amine (103.8 mg, 0.500 mmol, 1.2 eq.) and Pd(dppf)Cl2 (30.5 mg, 0.042 mmol, 0.1 eq.) were added, and the mixture was stirred at 60° C. for 16 h. The mixture was diluted with water (5 mL) and extracted with EA (3×10 mL). The combined organic layers were washed with brine (30 mL) (3×10 mL), dried over Na2SO4, filtered and concentrated to give 7-((3aS,4R,6S,6aR)-6-(4-(6-amino-5-chloropyridin-2-yl)butyl)-2,2,6-trimethyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine (50.5 mg, 70% purity, 0.075 mmol, 18% yield). LCMS: (ESI): RT=1.757 min, m/z calcd. for C24H32ClO2N6 471.22, [M+H]+, found 471.3.

To a solution of 7-((3aS,4R,6S,6aR)-6-(4-(6-amino-5-chloropyridin-2-yl)butyl)-2,2,6-trimethyltetrahydro-4H-cyclopenta[d][1,3]dioxol-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine (50.5 mg, 70% purity, 0.075 mmol, 1 eq.) in THF (3 mL) was added HCl (4 M, 1.5 mL). The mixture was stirred at 20° C. for 12 h. Upon completion, the mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Gemini-NX 80*40 mm*3 um; mobile phase: [water (0.05% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 20%-50%,8 min) to give (1S,2R,3S,5R)-3-(4-(6-amino-5-chloropyridin-2-yl)butyl)-5-(4-amino-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-3-methylcyclopentane-1,2-diol (46) (9.0 mg, 0.021 mmol, 28%). 1H NMR (400 MHz, CD3OD) δ: 8.05 (s, 1H), 7.45 (d, J=7.8 Hz, 1H), 7.21 (d, J=3.5 Hz, 1H), 6.60 (d, J=3.5Hz, 1H), 6.51 (d, J=7.8 Hz, 1H), 5.01-4.93 (m, 1H), 4.45 (dd, J=6.3, 7.8 Hz, 1H), 3.80 (d, J=6.0 Hz, 1H), 2.64-2.57 (m, 2H), 1.98 (dd, J=8.8, 12.8 Hz, 1H), 1.77 (dd, J=10.8, 13.1 Hz, 1H), 1.68 (quin, J=7.3 Hz, 2H), 1.60-1.31 (m, 4H), 1.09 (s, 3H).

Example 42 Compound 47

To a solution of 47-1 (177.5 mg, 0.508 mmol, 1 eq.) in THF (4 mL) was added 9-BBN dimer (313.6 mg, 1.30 mmol, 2.5 eq.), and the mixture was stirred at 50° C. for 2 h under N2. The mixture was cooled to rt, and then a solution of K3PO4 (550.1 mg, 2.6 mmol, 5 eq.) in H2O (0.5 mL) was added. After stirring at rt for 0.5 h, 6-bromopyridin-2-amine (107.6 mg, 0.622 mmol, 1.2 eq.) and Pd(dppf)Cl2 (37.9 mg, 0.052 mmol, 0.1 eq.) were added, and the mixture was stirred at 60° C. for 16 h under N2. Upon completion, the mixture was diluted with brine (10 mL) and extracted with EA (2×20 mL). The combined organic layers were combined and dried over NaS2O4, filtered, and concentrated to give a residue. The residue was purified by prep-HPLC (40 g C-18 column, gradient: 0%-70% CH3CN in water (1 mL NH3H2O in 2L H2O) in 15 min@ 40 mL/min to afford 47-2 (85.4 mg, 0.196 mmol, 38% yield) as a brown solid. LCMS: (ESI): RT=1.553 min, m/z calcd. for C24H33O2N6 437.26, [M+H]+, found 437.2.

To a solution of 47-2 (85.4 mg, 0.196 mmol, 1 eq.) in THF (3 mL) was added HCl (aq., 4 M, 1.5 mL), and the mixture was stirred at 20° C. for 12 h. Upon completion, the mixture was concentrated under reduced pressure to give a residue, which was purified by prep-HPLC (column: Venusil ASB Phenyl 150*30 mm*5 um; mobile phase: [water (0.05%HCl)-ACN]; B%: 9%-39%, 9 min) to afford 47 as a hydrochloride salt (54.4 mg, 0.115 mmol, 59% yield). LCMS: (ESI): RT=3.688 min, m/z calcd. for C21H29N6O2 397.23, [M+H]+, found 397.3. 1H NMR (400 MHz, CD3OD) δ: 8.25 (s, 1H), 7.84 (dd, J=7.3, 8.8 Hz, 1H), 7.57 (d, J=3.5 Hz, 1H), 6.92 (d, J=3.5 Hz, 1H), 6.84 (d, J=8.8 Hz, 1H), 6.76 (d, J=7.3 Hz, 1H), 5.14-5.05 (m, 1H), 4.55-4.48 (m, 1H), 3.84 (d, J=6.0 Hz, 1H), 2.80 (br t, J=7.8 Hz, 2H), 2.04-1.96 (m, 1H), 1.92-1.83 (m, 1H), 1.81-1.72 (m, 2H), 1.66-1.38 (m, 4H), 1.11 (s, 3H).

Example 43 Compound 48

To a solution of 7-bromopyrrolo[2,1-f][1,2,4]triazin-4-amine (48-1) (5.5 g, 25.82 mmol, 1 eq.) in DMF (50 mL) was added NaH (2.58 g, 64.54 mmol, 60% purity, 2.5 eq.) at 0° C., and the mixture was stirred at 0° C. for 0.5 h. 2-(chloromethoxy) ethyl-trimethyl-silane (9.04 g, 54.22 mmol, 9.60 mL, 2.1 eq.) was added, and the mixture was stirred at 25° C. for 5 h. Upon completion, the reaction was quenched by sat. aq. NH4Cl (50 mL) and extracted with EA (2×50 mL). The combined organic layers were washed with brine (50 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue, which was purified by flash silica gel chromatography (ISCO®; 40 g SepaFlash® Silica Flash Column, Eluent of 0˜2% EA/PE gradient @ 40 mL/min) to afford 48-2 (7.6 g, 15.90 mmol, 61% yield) as a colorless oil. LCMS: (ESI): m/z calcd. for C18H34BrN4O2Si2 473.13 [M+H]+, found 473.2. 1H NMR (400 MHz, CDCl3) δ: 8.09 (s, 1H), 7.08 (d, J=4.8 Hz, 1H), 6.77 (d, J=4.8 Hz, 1H), 5.22 (s, 4H), 3.70-3.65 (m, 4H), 1.02-0.95 (m, 4H), 0.02-0.00 (m, 18H).

Chloro(isopropyl)magnesium lithium chloride (1.3 M, 5.88 mL in THF, 3 eq.) was added dropwise to a solution of 48-2 (3.62 g, 7.64 mmol, 3 eq.) in THF (5 mL) at −20° C. The mixture was stirred at −20° C. for 10 min, then warmed to 0° C. The mixture was stirred at 0° C. for 1 h, and a solution of 48-3 (500 mg, 2.55 mmol, 1 eq.) in THF (7 mL) was added dropwise at -20° C. The mixture was stirred at 0° C. for 1 h. Upon completion, the reaction was quenched by sat. NH4Cl solution (15 mL) and extracted with EA (2×20 mL). The combined organic layers were washed with water and brine (20 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue, which was purified by flash silica gel chromatography (ISCO®; 40 g SepaFlash® Silica Flash Column, Eluent of 0˜10% EA/PE gradient @ 30 mL/min) to give 48-4 (967 mg, 94% purity, 1.54 mmol, 60% yield) as a yellow oil. LCMS: (ESI): m/z calcd. for C29H51N4O5Si2 591.33 [M+H]+, found 591.4.

A mixture of 48-4 (470 mg, 0.795 mmol, 1 eq.) and 9-BBN dimer (481.2 mg, 1.99 mmol, 2.5 eq.) in THF (10 mL) was stirred at 50° C. for 2 h under N2. The mixture was cooled to rt, and a solution of K3PO4 (844.2 mg, 3.98 mmol, 5 eq.) in H2O (1 mL) was added. The mixture was stirred for 0.5 h. Q5 (212.9 mg, 0.954 mmol, 1.2 eq.) and Pd(dppf)Cl2 (58.20 mg, 0.080 mmol, 0.1 eq.) were added. The mixture was stirred at 70° C. under N2 for 12 h. The mixture was diluted with brine (10 mL) and extracted with EA (2×10 mL). The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, PE:EA (3:1 to 1:1) to (DCM:MeOH 30:1 to 20:1)) to afford 48-5 (508 mg, 0.580 mmol, 72% yield) as a yellow solid. LCMS: (ESI): m/z calcd. for C38H59N6O5Si2 735.4 [M+H]+, found 735.5.

To a solution of 48-5 (350 mg, 0.476 mmol, 1 eq.) in DCM (8.5 mL) was added DAST (383.8 mg, 2.38 mmol, 314 μL, 5 eq.) at 0° C. The mixture was stirred at 0° C. for 1 h. The mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, DCM:MeOH (50:1 to 20:1)) to afford 48-6 (313 mg, 0.393 mmol, 82% yield) as a white solid. LCMS: (ESI): m/z calcd. for C38H57N6O4Si2 717.39 [M+H]+, found 717.4.

To a solution of 48-6 (313 mg, 0.393 mmol, 1 eq.) in THF (8.5 mL) was added HCl (aq., 4 M, 4 mL). The mixture was stirred at 25° C. for 3.5 h. The mixture was filtered and concentrated under reduced pressure to give a residue. To a solution of the residue in a mixed a solvent of t-BuOH (7 mL) and H2O (3 mL) was added PPTS (700.71 mg, 2.79 mmol, 5 eq.). The mixture was stirred at 50° C. for 12 h, and then concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Gemini-NX 80*40 mm*3 um; mobile phase: [water (0.05% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 17%-47%, 8 min) to afford 48-7 (57 mg, 0.133 mmol, 34% yield) as a white solid. LCMS: (ESI): m/z calcd. for C23H23N6O2 417.2 [M+H]+, found 417.3.

A mixture of 48-7 (48 mg, 0.115 mmol, 1 eq.) and PtO2 (250 mg, 1.10 mmol, 9.55 eq.) in THF (10 mL) was hydrogenated under H2 (1 atm) at rt for 12 h. The mixture was filtered through a pad of Celite. The filtrate was concentrated under reduced pressure to give a residue, which was purified by prep-HPLC (column: Phenomenex Gemini-NX 80*40 mm*3 um; mobile phase: [water (0.05% NH3H2O+10 mM NH4HCO3)-ACN];13%: 24%-38%,8 min) to give 48 (10 mg, 0.0236 mmol, 20.5% yield, 98.88% purity) as a white solid. LCMS: (ESI): m/z calcd. for C23H27N6O2 419.21 [M+H]+, found 419.3. 1H NMR (400 MHz, CD3OD) δ: 7.90 (d, J=8.8Hz, 1H), 7.75 (s, 1H), 7.56 (d, J=8.3Hz, 1H), 7.35 (s, 1H), 7.12 (dd, J=1.5, 8.0 Hz, 1H), 6.84 (d, J=4.5Hz, 1H), 6.76 (d, J=8.8 Hz, 1H),6.55 (d, J=4.5 Hz, 1H), 4.46-4.41(m, 1H), 3.86 (d, J=6.0 Hz, 1H),3.78-3.70 (m, 1H), 2.88-2.69 (m,2H), 2.08-2.01 (m, 1H), 1.89-1.66(m, 3H), 1.23 (s, 3H).

Example 44 Compound 49

To a mixture of 3Q (100 mg, 0.302 mmol, 1 eq.) in THF (5 mL) was added 9-BBN dimer (160.7 mg, 0.664 mmol, 2.2 eq.). The mixture was stirred at 50° C. for 1 h and then cooled to rt. A solution of K3PO4 (320.3 mg, 1.51 mmol, 5 eq.) in H2O (0.5 mL) was added, and the mixture was stirred at rt for 0.2 h. 7-bromo-3-chloro-quinolin-2-amine Q8 (93.3 mg, 0.362 mmol, 1.2 eq.) and Pd(dppf)Cl2 (22.08 mg, 0.030 mmol, 0.1 eq.) were then added. The mixture was stirred at 60° C. for 8 h. Upon completion, the mixture was diluted with brine (10 mL) and extracted with EA (3×20 mL). The separated organic layers were combined, dried over anhydrous Na2SO4 and concentrated to give a residue. The residue was purified by prep-HPLC (40 g C-18 column: chromatography (0%-65% CH3CN/H2O (1 mL NH3H2O in 2 L H2O) @ 40 mL/min) to afford 49-2 (55 mg, 0.105 mmol, 35% yield, 97.4% purity) as a brown solid. LCMS: (ESI): m/z calcd. for C27H30ClFN5O2 510.20 [M+H]+, found 510.3.

To a solution of 49-2 (55 mg, 0.105 mmol, 1 eq.) in THF (3 mL) was added HCl (4 M, 2 mL). The mixture was stirred at rt for 6 h. Upon completion, the mixture was concentrated under reduced pressure to afford a residue, which was purified by prep-HPLC twice (column: Phenomenex Gemini-NX 80*40 mm*3 um; mobile phase: [water(0.05% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 32%-52%, 8 min) to give 49 (28 mg, 0.0593 mmol, 56% yield, 99.49% purity) as a white solid. LCMS: (ESI): m/z calcd. for C24H26ClFN5O2 470.17 [M+H]+, found 470.1. 1H NMR (400 MHz, CD3OD) δ: 8.06 (s, 1H), 7.56 (d, J=8.3 Hz, 1H), 7.42 (d, J=5.0 Hz, 1H), 7.40 (s, 1H), 7.35 (d, J=3.3 Hz, 1H), 7.17 (dd, J=1.5, 8.3 Hz, 1H), 6.73-6.68 (m, 1H), 5.11-5.00 (m, 1H), 4.46-4.38 (m, 1H), 3.91 (d, J=6.5 Hz, 1H), 2.90-2.69 (m, 2H), 2.11 (dd, J=8.3, 12.8 Hz, 1H), 1.93-1.73 (m, 3H), 1.22 (s, 3H).

Example 45 Compound 50

To a solution of 3Q (100 mg, 0.302 mmol, 1 eq.) in THF (5 mL) was added 9-BBN dimmer (160.7 mg, 0.664 mmol, 2.2 eq.). The mixture was stirred at 50° C. for 1 h and cooled to rt. A solution of K3PO4 (320.3 mg, 1.51 mmol, 5 eq.) in H2O (0.5 mL) was added, and the mixture was stirred at rt for 0.2 h. 3-bromo-7-iodo-quinolin-2-amine (Q1) (126.4 mg, 0.362 mmol, 1.2 eq.) and Pd(dppf)Cl2 (22.1 mg, 0.030 mmol, 0.1 eq.) were then added. The mixture was stirred at 50° C. for 3 h. Upon completion, the mixture was diluted with brine (10 mL) and extracted with EA (3×20 mL). The separated organic layers were combined, dried over anhydrous Na2SO4 and concentrated to give a residue, which was purified by column chromatography (100-200 mesh, SiO2, CH2Cl2/MeOH (100:1 to 10:1), eluted 1 L) to provide 50-2 (160 mg, 0.212 mmol, 73.4% purity, 70% yield) as a brown solid. LCMS: (ESI): m/z calcd. for C27H30BrFN5O2 554.15 [M+H]+, found 556.1.

To a solution of 50-2 (160 mg, 0.212 mmol, 1 eq.) in THF (4 mL) was added HCl (4 M, 2 mL). The mixture was stirred at rt for 8 h. Upon completion, the mixture was concentrated under reduced pressure to afford a residue. The residue was purified by prep-HPLC (acidic condition, column: Agela ASB 150*25 mm*5 um; mobile phase: [water(0.05%HCl)-ACN]; B%: 12%-42%, 9min), and then (basic condition, column: Phenomenex Gemini-NX 80*40 mm*3 um; mobile phase: [water(0.05% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 30%-60%, 8 min) to give 50 (25 mg, 0.0484 mmol, 23% yield, 99.66% purity) as a white solid. LCMS: (ESI): m/z calcd. for C24H26BrFN5O2 516.12 [M+H]+, found 516.1. 1H NMR (400 MHz, CD3OD) δ: 8.25 (s, 1H), 7.55 (d, J=8.3 Hz, 1H), 7.42 (d, J=5.0 Hz, 1H), 7.39 (s, 1H), 7.35 (d, J=3.3 Hz, 1H), 7.17 (dd, J=1.5, 8.3 Hz, 1H), 6.73-6.67 (m, 1H), 5.12-5.01 (m, 1H), 4.45-4.39 (m, 1H), 3.91 (d, J=6.3 Hz, 1H), 2.90-2.69 (m, 2H), 2.11 (dd, 12.5 Hz, 1H), 1.93-1.72 (m, 3H), 1.22 (s, 3H). 19F NMR (376 MHz, CD3OD) δ: −161.68 (br s, 1F).

Example 46 Compound 51

To a mixture of PPh3CH3Br (255.91 mg, 716.38 μmol, 5.2 eq.) in THF (3 mL) was added t-BuOK (77.29 mg, 688.83 μmol, 5 eq.) at 0° C., and the mixture was stirred at 25° C. for 0.5 h. Compound 51-1 (75 mg, 137.77 μmol, 1 eq.) was added, and the mixture was stirred at 25° C. for 1.5 h. Upon completion, the reaction was quenched by the addition of NH4Cl (10 mL) at 0° C. The mixture was then extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (3×20 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 20 g SepaFlash® Silica Flash Column, Eluent of 0˜1.9% MeOH/DCM @ 30 mL/min) to afford 51-2 (38 mg, 64.45 μmol, 46.78% yield, 88% purity) as a colorless oil. LCMS: (ESI): m/z calcd. for C27H27Cl2FN5O2 542.1. [M+H]+, found 542.1.

A solution of 51-2 (38 mg, 70.05 μmol, 1 eq.) in dioxane (1 mL) and NH3.H2O (4.55 g, 32.46 mmol, 5 mL, 25% aq., 206.74 eq.) was stirred in a 30 mL of sealed tube at 110° C. for 24 h. Upon completion, the reaction was quenched by the addition of NaHCO3 (sat. aq., 5 mL). The mixture was extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (3×20 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 25 g SepaFlash® Silica Flash Column, Eluent of 0˜3.1% MeOH/DCM gradient @ 30 mL/min) to afford 51-3 (26 mg, 46.73 μmol, 66.71% yield, 94% purity) as a white solid. LCMS: (ESI): m/z calcd. for C27H29ClFN6O2, 523.2 [M+H]+, found 523.1.

To a solution of 51-3 (26 mg, 49.71 μmol, 1 eq.) in THF (1 mL) was added HCl (4M, aq., 1 mL), and the mixture was stirred at 25° C. for 12 h. Upon completion, the mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Gemini-NX 80*40 mm*3 um; mobile phase: [water (0.05% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 21%-45%, 8 min) to afford 51 (15 mg, 31.06 μmol, 62.48% yield, 100% purity) as a white solid. LCMS: (ESI): m/z calcd. for C24H25ClFN6O2 483.2. [M+H]+, found 483.3. 1H NMR (400 MHz, CD3OD) δ: 8.18 (s, 1H), 8.09 (s, 1H), 7.23 (d, J=3.5 Hz, 1H), 7.21 (s, 1H), 6.89 (d, J=10.8 Hz, 1H), 6.59 (d, J=3.5 Hz, 1H), 6.13 (dd, J=11.0, 17.8 Hz, 1H), 5.42-5.22 (m, 2H), 5.03-4.92 (m, 1H), 4.60 (d, J=6.5 Hz, 1H), 4.02 (d, J=6.0 Hz, 1H), 2.81-2.61 (m, 2H), 2.57 (dd, J=9.0, 13.3 Hz, 1H), 2.11-1.92 (m, 3H). 19F NMR (376 MHz, CD3OD) δ: −125.86 (s, 1F).

Example 47 Compound 52

To a solution of 52-1 (75 mg, 137.77 μmol, 1 eq.) in CH3CN (3 mL) were added K2CO3 (38.08 mg, 275.53 μmol, 2 eq.) and 1-diazo-1-dimethoxyphosphoryl-propan-2-one (52.93 mg, 275.53 μmol, 2 eq.). The mixture was stirred at 25° C. for 12 h. Upon completion, the reaction was quenched by the addition of NH4Cl (sat. aq., 10 mL). The mixture was extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (3×20 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 20 g SepaFlash® Silica Flash Column, Eluent of 0˜2.1% MeOH/DCM @ 30 mL/min) to afford 52-2 (41 mg, 72.07 μmol, 52.32% yield, 95% purity) as a colorless oil. LCMS: (ESI): m/z calcd. for C27H25Cl2FN5O2 540.1. [M+H]+, found 540.0.

A solution of 52-2 (40 mg, 74.02 μmol, 1 eq.) in dioxane (4 mL) and NH3.H2O (3.64 g, 25.97 mmol, 4 mL, 25% purity, 206.74 eq.) was stirred in a 30 mL sealed tube at 110° C. for 24 h. Upon completion, the reaction was quenched by the addition of NaHCO3 (sat. aq., 5 mL). The mixture was extracted with EtOAc (3×20 mL). The combined organic layers were washed with brine (3×20 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 25 g SepaFlash® Silica Flash Column, Eluent of 0˜2.8% MeOH/DCM gradient @ 30 mL/min) to afford 52-3 (10 mg, 18.43 μmol, 24.90% yield, 96% purity) as a white solid. LCMS: (ESI): m/z calcd. for C27H27ClFN6O2, 521.2 [M+H]+, found 521.2.

A solution of 52-3 (10 mg, 18.50 μmol, 1 eq.) in HCl (4M aq., 1 mL) and THF (1 mL) was stirred at 25° C. for 12 h. Upon completion, the mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Gemini-NX 80*40 mm*3 um; mobile phase: [water (0.05% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 21%-45%, 8 min) to afford 52 (5 mg, 10.40 μmol, 56.19% yield, 100% purity) as a white solid. LCMS: (ESI): m/z calcd. for C24H23ClFN6O2 481.2. [M+H]+, found 481.2. 1H NMR (400 MHz, CD3OD) δ: 8.19 (s, 1H), 8.08 (s, 1H), 7.26 (s, 1H), 7.21 (d, J=3.8 Hz, 1H), 6.93 (d, J=10.0 Hz, 1H), 6.59 (d, J=3.5 Hz, 1H), 5.17-5.04 (m, 1H), 4.55 (t, J=6.8 Hz, 1H), 4.00 (d, J=6.8 Hz, 1H), 3.17-3.01 (m, 1H), 2.93 (dt, J=4.8, 12.7 Hz, 1H), 2.81 (s, 1H), 2.46 (dd, J=8.3, 12.8 Hz, 1H), 2.23-2.09 (m, 2H), 2.08-1.97 (m, 1H). 19F NMR (376 MHz, CD3OD) δ: −125.74 (s, 1F).

Example 48 Compound 53

To a solution of 53-1 (1 g, 5.04 mmol, 1 eq.) in DCM (10 mL) were added Tf2O (2.13 g, 7.57 mmol, 1.25 mL, 1.5 eq.) and pyridine (1.60 g, 20.18 mmol, 1.63 mL, 4 eq.). The mixture was stirred at 0° C. for 1 h. Upon completion, the reaction was quenched with H2O (10 mL) at 0° C., then extracted with DCM (10 mL). The aqueous phase was extracted with DCM (3×20 mL). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give 53-2 (1.8 g, crude) as a brown oil, which is used for the next step without further purification.

To a solution of 2,4-dichloro-7H-pyrrolo[2,3-d]pyrimidine (S4) (1.5 g, 7.98 mmol, 1.46 eq.) in DMF (5 mL) was added t-BuOK (832.57 mg, 7.42 mmol, 1.36 eq.). The mixture was stirred at 15° C. for 1 h. Compound 53-2 (1.8 g, 5.45 mmol, 1 eq.) was added at 0° C., and the mixture was stirred at 15° C. for 48 h under N2. Upon completion, the reaction was partitioned between DCM (20 mL) and brine (20 mL). The aqueous phase was extracted with DCM (3×30 mL). The organic layers were combined, dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 40 g SepaFlash® Silica Flash Column, Eluent of 0˜12% Ethyl acetate/Petroleum ether gradient @ 35 mL/min) to give 53-3 (0.51 g, 1.29 mmol, 25.6% yield, 93.173% purity) was obtained as colorless oil. LCMS: (ESI): m/z calcd. for C17H20Cl2N3O2 368.09 [M+H]+, found 369.9.

A solution of 53-3 (510 mg, 1.38 mmol, 1 eq.) in dioxane (10 mL) with sat. NH3H2O (9.10 g, 64.91 mmol, 10 mL, 25% purity, 46.95 eq.) was stirred at 100° C. for 48 h in a 100 mL of sealed tube. Upon completion, the reaction was partitioned between EA (30 mL) and brine (30 mL). The organic phase was separated, and the aqueous phase was extracted with EA (3×30 mL). The organic layers were combined and dried over Na2SO4, filtered and concentrated under reduced pressure to give crude 53-4 (470 mg, 1.29 mmol, 93.5% yield, 95.419% purity) as a yellow oil, which was directly used for the next step without further purification. LCMS: (ESI): m/z calcd. for C17H22ClN4O2 349.14 [M+H]+, found 349.1.

To the mixture of 53-4 (157.20 mg, 0.430 mmol, 1 eq.) in THF (4 mL) was added 9-BBN dimer (228.95 mg, 0.946 mmol, 2.2 eq.). The mixture was stirred at 50° C. for 1 h, and then cooled to 10° C. A solution of K3PO4 (456.39 mg, 2.15 mmol, 5 eq.) in H2O (0.5 mL) was added, and the mixture was stirred at 10° C. for 0.2 h. Q16 (142.16 mg, 0.516 mmol, 1.2 eq.) and Pd(dppf)Cl2 (31.46 mg, 0.043 mmol, 0.1 eq.) were added, and the mixture was stirred at 55° C. for 2 h under Ar. The mixture was partitioned between EA (20 mL) and water (10 mL). The aqueous phase was extracted with EA (3×20 mL). The organic layers were combined and washed with brine (30 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate (1:1) then DCM:MeOH (20:1)) to afford 53-5 (300 mg, 0.350 mmol, 77% yield, 63.729% purity) as a yellow solid. LCMS: (ESI): m/z calcd. for C26H28Cl2FN6O2 545.16 [M+H]+, found 545.1.

To a solution of 53-5 (300 mg, 0.350 mmol, 63.729% purity, 1 eq.) in THF (4 mL) was added HCl (4 M aq, 2 mL, 22.82 eq.). The mixture was stirred at 15° C. for 4 h, and then concentrated under reduced pressure to afford a residue. The residue was purified by prep-HPLC (column: Phenomenex Gemini-NX 80*40 mm*3 um; mobile phase: [water (0.05% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 34%-59%, 8 min) to give 53 (77 mg, 0.150 mmol, 42.88% yield, 98.64% purity) as an off-white solid. LCMS: (ESI): m/z calcd. for C23H24Cl2FN6O2 527.12 [M+Na]+, found 527.1. 1H NMR (400 MHz, CD3OD) δ: 8.19 (s, 1H), 7.24 (s, 1H), 7.18 (d, J=3.7 Hz, 1H), 6.94 (dd, J=1.1, 10.9 Hz, 1H), 6.56 (d, J=3.6 Hz, 1H), 4.97-4.91 (m, 1H), 4.58-4.48 (m, 1H), 3.93 (d, J=6.3 Hz, 1H), 2.92-2.71 (m, 2H), 2.09-1.96 (m, 2H), 1.93-1.79 (m, 2H), 1.23 (s, 3H). 19F NMR (400 MHz, CD3OD) δ: −125.87.

Example 49 Compound 54

To a mixture of 53-4 (142 mg, 0.388 mmol, 1 eq.) in THF (4 mL) was added 9-BBN dimer (206.82 mg, 0.855 mmol, 2.2 eq.). The mixture was stirred at 50° C. for 1 h, and then cooled to 10° C. A solution of K3PO4 (412.26 mg, 1.94 mmol, 5 eq.) in H2O (0.5 mL) was added, and the mixture was stirred at 10° C. for 0.2 h. Q1 (162.66 mg, 0.466 mmol, 1.2 eq.) and Pd(dppf)Cl2 (28.42 mg, 0.0388 mmol, 0.1 eq.) were added, and the mixture was stirred at 55° C. for 2 h under Ar. The mixture was partitioned between EA (20 mL) and water (10 mL). The aqueous phase was extracted with EA (3×20 mL). The organic layers were combined, washed with brine (30 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate (2:1) then DCM:MeOH (20:1)) to afford 54-1 (270 mg, 0.307 mmol, 79% yield, 65% purity) as a yellow solid. LCMS: (ESI): m/z calcd. for C26H29BrClN6O2 571.11 [M+H]+, found 573.0.

To a solution of 54-1 (270 mg, 0.307 mmol, 65% purity, 1 eq.) in THF (4 mL) was added HCl (4 M aq., 2 mL, 22.82 eq.), and the mixture was stirred at 15° C. for 4 h. The mixture was concentrated under reduced pressure to afford a residue. The residue was purified by prep-HPLC (column: Phenomenex Gemini-NX 80*40 mm*3 um; mobile phase: [water (0.05% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 32%-56%, 8 min) to give 54 (70 mg, 0.132 mmol, 43% yield, 100% purity) as a white solid. LCMS: (ESI): m/z calcd. for C23H25BrClN6O2 531.08 [M+H]+, found 533.1. 1H NMR (400 MHz, CD3OD) δ: 8.26 (s, 1H), 7.55 (d, J=8.2 Hz, 1H), 7.40 (s, 1H), 7.20 (d, J=8.2 Hz, 1H), 7.17 (d, J=3.6 Hz, 1H), 6.56 (d, J=3.6 Hz, 1H), 4.98-4.91 (m, 1H), 4.52 (t, J=6.9 Hz, 1H), 3.93 (d, J=6.2 Hz, 1H), 2.93-2.70 (m, 2H), 2.11-1.93 (m, 2H), 1.92-1.79 (m, 2H), 1.23 (s, 3H).

Example 50 Compound 55

A mixture of 55-1 (613 mg, 1.04 mmol, 1 eq.) and 9-BBN dimer (627 mg, 2.59 mmol, 2.5 eq.) in THF (10 mL) was stirred at 50° C. for 2 h under Ar, and the cooled to rt. A solution of K3PO4 (1.10 g, 5.19 mmol, 5 eq.) in H2O (1 mL) was added, and the mixture was stirred for 0.5 h. Q9 (419 mg, 1.24 mmol, 1.2 eq.) and Pd(dppf)Cl2 (76 mg, 0.103 mmol, 0.1 eq.) were added, and the mixture was purged with Ar (3×) and then stirred at 70° C. for 12 h under Ar. The reaction was quenched by the addition of water (50 mL) and extracted with EA (3×100 mL). The combined organic layers were washed with brine (100 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether:Ethyl acetate=30:1 to 5:1) to afford 55-2 (550 mg, 608.80 μmol, 59% yield, 94% purity) as a white solid. LCMS: (ESI): m/z calcd. for C44H69N6O7Si2 849.47 [M+H]+, found 849.4.

To a solution of 55-2 (550 mg, 647.66 μmol, 1 eq.) in DCM (10 mL) was added DAST (1.04 g, 6.48 mmol, 855.70 μL, 10 eq.) at 0° C., and the mixture was stirred at 0° C. for 2 h. The reaction was quenched by addition of water (50 mL) and extracted with EA (3×100 mL). The combined organic layers were washed with brine (100 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether:Ethyl acetate=30:1 to 5:1) to afford 55-3 (450 mg, 498.08 μmol, 76.90% yield, 92% purity) as a white solid. LCMS: (ESI): m/z calcd. for C44H66N6O6Si2Na 853.46 [M+Na]+, found 853.4.

To a solution of 55-3 (440 mg, 529.36 μmol, 1 eq.) in THF (8 mL) was added HCl (4 M, 4 mL), and the mixture was stirred at 25° C. for 5 h. Upon completion, the mixture was concentrated under reduced pressure to give a residue. The residue was dissolved in a mixed solvent of t-BuOH (8 mL) and H2O (4 mL), and PPTS (664.15 mg, 2.64 mmol, 5 eq.) was added. The mixture was stirred at 50° C. for 3 h. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Gemini-NX 80*40 mm*3 um; mobile phase: [water (0.05% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 17%-47%,8 min) to afford 55-4 (200 mg, 373.15 μmol, 71% yield, 99% purity) as a white solid. LCMS: (ESI): m/z calcd. for C29H35N6O4 531.26 [M+H]+, found 531.4.

To a solution of 55-4 (175 mg, 329.80 μmol, 1 eq.) in THF (30 mL) was added PtO2 (875 mg, 3.85 mmol, 11.68 eq.), and the mixture was stirred under H2 (15 psi) atmosphere at 25° C. for 18 h. The mixture was filtered through a pad of Celite to remove PtO2. The filtrate was concentrated under reduced pressure to afford a residue. The residue was purified by SFC (column: Chiralcel OJ-3 100Å4.6 mm I.D., 3 um; Mobile phase: A: CO2 B: ethanol (0.05% DEA); Isocratic: 40% B; Flow rate: 2.8 mL/min; Column temp.: 35° C.; ABPR: 1500 psi; to afford 55 (70 mg, 128.79 μmol, 39% yield, 98% purity) as a white solid. LCMS: (ESI): m/z calcd. for C29H37N6O4 533.28 [M+H]+, found 533.4. 1H NMR (400 MHz, DMSO) δ: 10.33 (s, 1H), 8.24 (d, J=9.0 Hz, 1H), 7.99 (d, J=8.8 Hz, 1H), 7.81-7.75 (m, 2H), 7.53 (s, 3H),7.32 (br d, J=8.2 Hz, 1H), 6.81 (d, J=4.4 Hz, 1H), 6.51 (d, J=4.2 Hz, 1H), 4.69 (br d, J=5.7 Hz, 1H), 4.41 (br d, J=5.3 Hz, 1H),4.29 (br d, J=6.8 Hz, 1H), 4.11 (t, J=6.6 Hz, 2H), 3.72 (br t, J=5.5 Hz, 1H), 3.61 (br d, J=9.3 Hz, 1H), 2.80 (br dd, J=4.4, 12.3Hz, 1H), 2.75-2.67 (m, 1H), 1.93 (br dd, J=8.7, 12.7 Hz, 1H), 1.80-1.71 (m, 1H), 1.68-1.56 (m, 3H), 1.50 (br t, J=11.8 Hz, 1H), 1.38-1.31 (m, 4H), 1.12 (s, 3H), 0.92-0.87 (m, 3H).

Example 51 Compound 56

To a solution of 56-1 (5 g, 36.73 mmol, 1 eq.) in DMF (50 mL) was added NIS (10.74 g, 47.76 mmol, 1.3 eq.), and the mixture was stirred at 40° C. for 3.5 h. The mixture was poured into water (200 mL) and filtered to give a residue. The residue was triturated with water (50 mL×2) to give 56-2 (6.49 g, 23.78 mmol, 64% yield, 96% purity) as a brown solid. LCMS: (ESI): m/z calcd. for C5H4IN4O 262.94 [M+H]+, found 262.9.

A solution of 56-2 (6.49 g, 24.77 mmol, 1 eq.), POCl3 (83.73 g, 546.07 mmol, 50.75 mL, 22.05 eq.), and DMAP (9.08 g, 74.31 mmol, 3 eq.) was stirred at 105° C. for 2 h. The mixture was concentrated under reduced pressure to give 56-2a (15 g, crude) as a brown solid. To a solution of 56-2a (7.5 g, 26.74 mmol, 1 eq.) in DCM (80 mL) was added N-methylaniline (11.46 g, 106.97 mmol, 11.61 mL, 4 eq.) dropwise at 0° C., followed by addition of TEA (16.24 g, 160.45 mmol, 22.33 mL, 6 eq.). The mixture was stirred at 25° C. for 12 h. The mixture was diluted with H2O (50 mL) and extracted with DCM (2×50 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 120 g SepaFlash® Silica Flash Column, Eluent of 0˜20% Ethyl acetate/Petroleum ether gradient @ 50 mL/min) to give 56-3 (6.68 g, 18.83 mmol, 76% yield, 99% purity) as a white solid (Note: two parallel reactions from 56-2a were carried out, then combined for purification). LCMS: (ESI): m/z calcd. for C12H11IN5 352.0 [M+H]+, found 351.9.

To a solution of 56-3 (5.37 g, 15.29 mmol, 3 eq.) in THF (15 mL) was added i-PrMgCl.LiCl (1.3 M, 12.94 mL, 3.3 eq.) dropwise. The mixture was stirred at −20° C. for 10 min, warmed to 0° C. and then stirred at 0° C. for 1 h. A solution of 56-4 (1 g, 5.10 mmol, 1 eq.) in THF (10 mL) was added dropwise to the mixture at −20° C., and then stirred at 0° C. for 1 h 20 min. The reaction was quenched with NH4Cl solution (30 mL) and extracted with EA (2×30 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash C-18 column chromatography (40 g, 0%-60% MeCN, 0.5 mL NH3.H2O in 1 L H2O, 30 mL/min) to give 56-5 (1.01 g, 2.30 mmol, 45% yield) as a white solid. LCMS: (ESI): m/z calcd. for C23H28N5O3 422.21 [M+H]+, found 422.1.

To a solution of 56-5 (1.01 g, 2.40 mmol, 1 eq.) in THF (25 mL) was added 9-BBN dimer (1.45 g, 5.99 mmol, 2.5 eq.). The mixture was stirred at 50° C. for 2 h under N2 and then cooled to rt. A solution of K3PO4 (2.54 g, 11.98 mmol, 5 eq.) in H2O (2.5 mL) was added, and the mixture was stirred at rt for 0.5 h. 7-bromoquinolin-2-amine Q5 (694.89 mg, 3.12 mmol, 1.3 eq.) and Pd(dppf)Cl2 (175.34 mg, 0.239 mmol, 0.1 eq.) were added, and the mixture was stirred at 60° C. for 12 h. The mixture was diluted with H2O (20 mL) and extracted with EA (2×20 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether:Ethyl acetate=2:1 to 1:1 to DCM:MeOH=100:1 to 20:1) to give 56-6 (1 g, 1.71 mmol, 71% yield) as a yellow solid. LCMS: (ESI): m/z calcd. for C32H36N7O3 566.28 [M+H]+, found 566.2.

To a solution of 56-6 (0.95 g, 1.68 mmol, 1 eq.) in DCM (15 mL) was added DAST (1.35 g, 8.40 mmol, 1.11 mL, 5 eq.) at 0° C., and the mixture was stirred at 0° C. for 0.5 h. reaction was quenched by addition of NaHCO3 solution (10 mL), diluted with DCM (5 mL) and extracted with DCM (2×10 mL). The combined organic layers were washed with brine (20 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, DCM:MeOH=100:1 to 20:1) to give 56-7 (708 mg, 1.27 mmol, 75% yield, 98% purity) as a yellow solid. LCMS: (ESI): m/z calcd. for C32H34N7O2 548.27 [M+H]+, found 548.1.

To a solution of 56-7 (708 mg, 1.29 mmol, 1 eq.) in dioxane (15 mL) was added NH3H2O (13.65 g, 97.37 mmol, 15 mL, 25% purity), and the mixture was stirred at 100° C. for 24 h. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, DCM:MeOH=50:1 to 12:1) to give 56-8 (238 mg, 483.78 μmol, 37% yield, 93% purity) as a white solid. LCMS: (ESI): m/z calcd. for C25H28N7O2 458.22 [M+H]+, found 458.3.

To a solution of 56-8 (228 mg, 498.33 μmol, 1 eq.) in THF (4 mL) was added HCl (4 M aq., 2.00 mL), and the mixture was stirred at 25° C. for 12 h. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Gemini-NX 80*40 mm*3 um; mobile phase: [water (0.05% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 40%-60%, 8 min) to give 56-9 (160 mg, 375.60 μmol, 75% yield, 98% purity) as a white solid. LCMS: (ESI): m/z calcd. for C22H24N7O2 418.19 [M+H]+, found 418.1.

To a solution of 56-9 (30 mg, 71.86 μmol, 1 eq.) in a mixed solvent of THF (7.5 mL) and MeOH (1.5 mL) was added PtO2 (150 mg, 660.57 μmol, 9.19 eq.). The suspension was degassed/purged with H2 (3×) and stirred under H2 (15 Psi) atmosphere at 25° C. for 4 h. The mixture was filtered through a pad of Celite to remove PtO2. The filtrate was concentrated under reduced pressure to give a white solid (30 mg, crude product). The crude product was combined with another batch (25 mg scale) for prep-HPLC (column: Phenomenex Gemini-NX 80*40 mm*3 um; mobile phase: [water (0.05% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 12%-42%,8 min) to give 56 (impure, 10 mg, 23.39 μmol, 98.1% purity by HPLC, NMR shows an impurity) as a white solid, which was confirmed by LCMS, HPLC and 1H NMR. LCMS: (ESI): m/z calcd. for C22H26N7O2 420.2 [M+H]+, found 420.3. 1H NMR (400 MHz, CD3OD) δ: 8.28 (d, J=9.3Hz, 1H), 8.01 (d, J=9.8 Hz, 2H), 7.79 (d, J=8.3 Hz, 1H), 7.50 (s, 1H), 7.42 (d, J=8.5 Hz, 1H), 6.99 (d, J=9.3 Hz, 1H), 4.30 (dd, J=6.3, 8.0Hz, 1H), 3.86 (d, J=6.0 Hz, 1H), 3.44-3.36 (m, 1H), 2.97-2.80 (m, 2H), 2.03-1.94 (m, 1H), 1.89-1.74(m, 3H), 1.22 (s, 3H).

Impure 56 was further purified by SFC (column: DAICEL CHIRALPAK AD (250 mm*30 mm, 10 um); mobile phase: [0.1% NH3H2O ETOH]; B%: 60%-60%, min) to give 56 (2 mg, 4.57 μmol, 19% yield, 95.9% purity) as a white solid, which was confirmed by LCMS, HPLC and 1H NMR. LCMS: (ESI): m/z calcd. for C22H26N7O2 420.2 [M+H]+, found 420.3. 1H NMR (400 MHz, CD3OD) δ: 8.01 (s, s, 2H), 7.93 (d, J=9.0 Hz, 1H), 7.58 (d, J=8.2 Hz, 1H), 7.37 (s, 1H), 7.16 (dd, J=1.5, 8.1 Hz, 1H), 6.78 (d, J=8.9 Hz, 1H), 4.30 (dd, 8.2 Hz, 1H), 3.85 (d, J=6.1 Hz, 1H), 3.40 (td, J=8.4, 11.1 Hz, 1 H), 2.90-2.71 (m, 2H), 1.99 (m, 1H), 1.87-1.72 (m, 3H), 1.21 (s, 3H).

Example 52 Compound 57

To a mixture of 57-1 (500 mg, 846.17 μmol, 1 eq.) in THF (10 mL) was added 9-BBN dimer (512 mg, 2.12 mmol, 2.5 eq.). The mixture was stirred at 50° C. for 2 h under N2, and then cooled to 25° C. A solution of K3PO4 (898 mg, 4.23 mmol, 5 eq.) in H2O (1 mL) was added, and the mixture was stirred for 0.5 h. Q16 (280 mg, 1.02 mmol, 1.2 eq.) and Pd(dppf)Cl2 (62 mg, 0.084 mmol, 0.1 eq.) were added, and the mixture was purged with N2 (3×). The mixture was stirred at 60° C. for 12 h. The mixture was diluted with water (5 mL) and extracted with EA (2×5 mL). The combined organic layers were washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 40 g SepaFlash® Silica Flash Column, Eluent of 5-20% Ethyl acetate/Petroleum ether gradient @ 30 mL/min) to afford 57-2 (415 mg, 519.60 μmol, 61% yield, 98.6% purity) as a yellow solid. LCMS: (ESI): m/z calcd. for C38H57ClFN6O5Si2 787.35 [M+H]+, found 787.1.

To a solution of 57-2 (415 mg, 526.98 μmol, 1 eq.) in DCM (7 mL) was added DAST (424.72 mg, 2.63 mmol, 348.13 μL, 5 eq.) at 0° C. The mixture was stirred at 0° C. for 1 h. The mixture was diluted with water (10 mL) and extracted with DCM (2×5 mL). The combined organic layers were washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 40 g SepaFlash® Silica Flash Column, Eluent of 0˜20% Ethyl acetate/Petroleum ether gradient @ 30 mL/min) to afford 57-3 (274 mg, 341.84 μmol, 64% yield, 96% purity) as a white solid. LCMS: (ESI): m/z calcd. for C38H54ClFN6O4Si2Na 791.34 [M+Na]+, found 791.2.

To a solution of 57-3 (207 mg, 0.269 mmol, 1 eq.) in THF (4 mL) was added HCl (aq., 4 M, 2 mL). The mixture was stirred at 25° C. for 6 h. The mixture was concentrated under reduced pressure to give a residue. The residue was dissolved in t-BuOH (10 mL) and H2O (5 mL), and PPTS (352.57 mg, 1.40 mmol, 5 eq.) was added. The mixture was stirred at 50° C. for 12 h. The mixture was concentrated under reduced pressure to give a residue. The residue was purified by reverse-phase flash column (40 g C-18 column, 0%-60% MeCN in water (0.5 mL NH3.H2O in 1 L H2O)@ 35 mL/min) to afford 57-4 (85 mg, 0.171 mmol, 63% yield, 94.8% purity) as a white solid. LCMS: (ESI): m/z calcd. for C23H23ClFN6O2 469.15 [M+H]+, found 469.1.

To a solution of 57-4 (30 mg, 0.063 mmol, 1 eq.) in MeOH (12 mL) was added Crabtree' s catalyst (180.00 mg, 223.63 μmol, 3.5 eq.). The suspension was degassed and purged with H2 (3×). The mixture was stirred under H2 (15 Psi) at 25° C. for 5 h. mixture was treated with TMT (1,3,5-triazine-2,4,6-trithiol) and then kept overnight. The resulting suspension was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, DCM:MeOH=7:1) to give a syrup (10 mg), which was further purified by SFC (column: DAICEL CHIRALPAK IG (250 mm*30 mm, 10 um); mobile phase: [0.1% NH3H2O IPA]; B%: 60%-60%, min) to give impure 57 (8 mg, 16.87 μmol, 26% yield, 99.29% purity, contains some grease) as a yellow solid. It was combined with another batch (7 mg of impure product) and triturated with EA (2×0.5 mL) and isopropyl ether (0.5 mL) to afford 57 (10 mg, 21.06 μmol, 99.18% purity) as a white solid. LCMS: (ESI): m/z calcd. for C23H25ClFN6O2 471.16 [M+H]+, found 471.2. 1H NMR (400 MHz, CD3OD) δ: 8.17 (s, 1 H), 7.76 (s, 1 H), 7.21 (s, 1 H), 6.87 (dd, J=1.3, 10.9 Hz, 1 H), 6.84 (d, J=4.4 Hz, 1 H), 6.54 (d, J=4.4 Hz, 1 H), 4.44 (dd, J=6.3, 7.2 Hz, 1 H), 3.85 (d, J=6.1 Hz, 1 H), 3.78-3.67 (m, 1 H), 2.87-2.67 (m, 2 H), 2.03 (dd, J=8.9, 12.9 Hz, 1 H), 1.87-1.68 (m, 3 H), 1.21 (s, 3 H).

Example 53 Compound 58

To a solution of 58-1 (2 g, 3.44 mmol, 1 eq.) in THF (20 mL) was added NaH (826.50 mg, 20.66 mmol, 60% purity, 6 eq.) in portions at 0° C. under N2. The mixture was stirred at 0° C. for 0.5 h. MeI (8.18 g, 57.63 mmol, 3.59 mL, 16.73 eq.) was added dropwise at 0° C. The mixture was stirred at 15° C. for 12 h under N2. The reaction was quenched by dropwise addition of NH4Cl (sat., aq., 50 mL) at 0° C. and extracted with EA (3×30 mL). The combined organic layer was washed with brine (30 mL), dried over Na2SO4, filtered and concentrated in vacuum. The residue was purified by a silica gel column (PE:EA=100:0 to 5:1) to give 58-2 (1.1 g, 1.63 mmol, 47% yield, 88% purity) as a colorless oil. LCMS: (ESI): m/z calcd. for C38H42O6Na, 617.30 [M+Na]+, found 617.1.

To a solution of 58-2 (0.8 g, 1.18 mmol, 88% purity, 1 eq.) and Et3SiH (1.38 g, 11.84 mmol, 1.89 mL, 10 eq.) in DCM (20 mL) was added TFA (539.89 mg, 4.73 mmol, 350.58 μL, 4 eq.) at 0° C. The mixture was stirred at 0° C. for 0.5 h. The reaction was quenched with sat. NaHCO3 solution (30 mL) and extracted with EA (3×30 mL). The combined organic layers were washed with brine (90 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 12 g CombiFlash® Silica Flash Column, Eluent of 0˜50% Ethyl acetate/Petroleum ether gradient @ 35 mL/min) to afford 58-3 (0.31 g, 0.859 mmol, 97% purity, 74% yield) as a colorless syrup. LCMS: (ESI): m/z calcd. for C19H28O6Na 375.2, [M+Na]+, found 375.2.

To a solution of 58-3 (0.31 g, 0.859 mmol, 1 eq.) in CH3CN (60 mL) was added IBX (738.93 mg, 2.64 mmol, 3 eq.). The mixture was stirred at 60° C. for 3 h. The mixture was cooled to 10° C. and diluted with EA (100 mL). The precipitation was filtered to move insoluble matters, and the filtrate was concentrated to afford 58-4 (0.34 g, crude) as a yellow gum. LCMS: (ESI): m/z calcd. for C19H30O6N 368.2, [M+NH4]+, found 368.3.

To a solution of MePPh3Br (980.18 mg, 2.74 mmol, 5 eq.) in THF (20 mL) was added t-BuOK (1 M in THF, 2.20 mL, 4 eq.), and the mixture was stirred at 10° C. for 0.5 h. A solution of 58-4 (0.34 g, 0.549 mmol, 56.557% purity, 1 eq.) in THF (10 mL) was added. The mixture was stirred at 10° C. for 1 h. The reaction was quenched with sat. NH4Cl (30 mL). The mixture was extracted with EA (3×30 mL) and washed with brine (60 mL). The separated organic layer was dried over anhydrous Na2SO4, filtered and concentrated to afford a residue. The residue was purified by flash silica gel chromatography (ISCO®; 25 g CombiFlash® Silica Flash Column, Eluent of 0˜20% Ethyl acetate/Petroleum ether gradient @ 35 mL/min) to afford 58-5 (210 mg, 0.557 mmol, 92.4% purity, 63% yield over 2 steps) as a colorless gum. LCMS: (ESI): m/z calcd. for C20H32O5N 366.2, [M+NH4]+ found 366.3.

To a solution of 58-5 (160 mg, 0.424 mmol, 92.4% purity, 1 eq.) in a mixed solvent of DCM (6 mL) and H2O (0.6 mL) was added DDQ (192.64 mg, 0.849 mmol, 2 eq.). The mixture was stirred at 10° C. for 18 h. The reaction was quenched with sat. NaHCO3 solution (20 mL), and then extracted with EA (3×20 mL). The combined organic layers were washed with brine (60 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a crude. The residue was purified by flash silica gel chromatography (ISCO®; 25 g CombiFlash® Silica Flash Column, Eluent of 0˜25% Ethyl acetate/Petroleum ether gradient @ 35 mL/min) to afford 58-6 (90 mg, 0.394 mmol, 93% yield) as a light yellow gum. 1H NMR (400 MHz, CDCl3) δ: 5.83 (dd, J=11.0, 17.8 Hz, 1 H), 5.20 (d, J=11.0 Hz, 1 H), 5.09 (d, J=18.1 Hz, 1 H), 4.54 (dd, J=0.9, 5.4 Hz, 1 H), 4.50-4.44 (m, 1 H), 4.05 (ddd, J=4.4, 6.0, 10.4 Hz, 1 H), 3.58 (d, J=8.8 Hz, 1 H), 3.34 (s, 3 H), 3.29 (d, J=8.8 Hz, 1 H), 2.42 (d, J=10.0 Hz, 1 H), 1.99 (dd, J=6.4, 12.2 Hz, 1 H), 1.52 (s, 3 H), 1.47 (d, J=12.0 Hz, 1 H), 1.38 (s, 3H).

To a solution of 58-6 (100 mg, 0.438 mmol, 1 eq.) and pyridine (138.6 mg, 1.75 mmol, 0.141 mL, 4 eq.) in DCM (5 mL) was added Tf2O (185.3 mg, 0.657 mmol, 0.108 mL, 1.5 eq.) dropwise at 0° C. The mixture was stirred at 0° C. for 2 h. The reaction was quenched with H2O (10 mL), and then extracted with DCM (3×20 mL). The combined organic layers were washed with brine (60 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to afford 58-7 (160 mg, crude, 0.444 mmol) as a brown oil. 1H NMR (400 MHz, CDCl3) δ: 5.84 (dd, J=11.2, 17.9 Hz, 1 H), 5.27 (d, J=11.0 Hz, 1 H), 5.13 (d,=17.8 Hz, 1 H), 4.96 (td, J=6.3, 10.8 Hz, 1 H), 4.64 (t, J=5.4 Hz, 1 H), 4.53 (d, J=5.0 Hz, 1 H), 3.59 (d, J=8.8 Hz, 1 H), 3.38-3.36 (m, 1 H), 3.35 (s, 3 H), 2.21 (dd, J=6.8, 12.3 Hz, 1 H), 2.13-1.99 (m, 1 H), 1.54 (s, 3 H), 1.37 (s, 3 H).

To a solution of 7a (127.7 mg, 666 μmol, 1.5 eq.) in DMF (2 mL) was added a solution of 58-7 (160 mg, 0.444 mmol, 1 eq.) in DMF (2mL). The mixture was stirred at 10° C. for 48 h. The mixture was added H2O (10 mL), and then extracted with EA (3×10 mL). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 12 g CombiFlash® Silica Flash Column, Eluent of 0˜10% Ethyl acetate/Petroleum ether gradient @ 30 mL/min) to afford 58-8 (90 mg, 0.242 mmol, 97.9% purity, 55% yield over 2 steps) as a colorless gum. LCMS: (ESI): m/z calcd. for C18H23O3N3Cl 364.1, [M+H]+, found 364.2.

To a solution of 58-8 (46 mg, 0.124 mmol, 97.953% purity, 1 eq.) in THF (3 mL) was added 9-BBN dimer (65.94 mg, 0.272 mmol, 2.2 eq.). The mixture was stirred at 50° C. for 1.5 h, and then cooled to 10° C. A solution of K3PO4 (131.44 mg, 0.619 mmol, 5 eq.) in H2O (0.3 mL) was added, and the mixture was stirred at 10° C. for 0.5 h. Q1 (51.86 mg, 0.149 mmol, 1.2 eq.) and Pd(dppf)Cl2 (9.06 mg, 0.012 mmol, 0.1 eq.) were added. The mixture was degassed with N2 (3×) and stirred at 50° C. for 2 h. The mixture was diluted with brine (10 mL), and then extracted with EA (3×10 mL). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 12 g CombiFlash® Silica Flash Column, Eluent of 0˜50% Ethyl acetate/Petroleum ether gradient @ 30 mL/min) to afford 58-9 (36 mg, 0.058 mmol, 47% yield, 94.2% purity) as a yellow gum. LCMS: (ESI): m/z calcd. for C27H30BrClN5O3 586.1, [M+2+H]+, found 588.1.

To a solution of 58-9 (36 mg, 0.058 mmol, 94% purity, 1 eq.) in dioxane (5 mL) was added NH3 .H2O (4.55 g, 32.46 mmol, 5 mL, 25% purity, 561.78 eq.). The mixture was stirred at 100° C. for 48 h in a 30 mL sealed tube. The mixture was treated with brine (10 mL) and extracted with EA (3×20 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to afford 58-10 (34 mg, 0.050 mmol, 87% yield, 84% purity) as a brown syrup. LCMS: (ESI): m/z calcd. for C27H32O3N6Br 567.2, [M+H]+, found 567.2.

To a solution of 58-10 (34 mg, 0.050 mmol, 83.799% purity, 1 eq.) in THF (4 mL) was added HCl (4 M aq., 2 mL, 159.34 eq.). The mixture was stirred at 10° C. for 3 h. The mixture was concentrated to give the residue. The residue was purified by prep-HPLC (basic condition; column: Phenomenex Gemini-NX 80*40 mm*3 um; mobile phase: [water (0.05% NH3 H2O+10 mM NH4HCO3)-ACN]; B%: 23%-49%, 8 min) to afford 58 (17 mg, 0.032 mmol, 64% yield, 99.1% purity) as a white solid. LCMS: (ESI): m/z calcd. for C24H28O3N6Br 527.1, [M+2+H]+, found 529.2. 1H NMR (400 MHz, CD3OD) δ: 8.27 (s, 1 H), 8.08 (s, 1 H), 7.57 (d, J=8.3 Hz, 1 H), 7.43 (s, 1 H), 7.23 (d, J=3.8 Hz, 1 H), 7.20 (d, J=8.3 Hz, 1 H), 6.59 (d, J=3.5 Hz, 1 H), 5.00 (q, J=9.5 Hz, 1 H), 4.62 (dd, J=5.4, 8.9 Hz, 1 H), 3.99 (d, J=5.3 Hz, 1 H), 3.70 (d, J=9.0 Hz, 1 H), 3.51 (d, J=9.3 Hz, 1 H), 3.43 (s, 3 H),2.92-2.70 (m, 2 H), 2.17 (dd, J=9.3, 13.6 Hz, 1 H), 1.99 (br t, J=8.4 Hz, 2 H), 1.88 (dd, J=10.0, 13.6 Hz, 1 H).

Example 54 Compound 59

To a solution of 59-1 (2 g, 9.39 mmol, 1 eq.) in DMF (20 mL) was added NaH (938.7 mg, 23.47 mmol, 60% purity, 2.5 eq.) at 0° C., and the mixture was stirred at 0° C. for 0.5 h. SEM-Cl (3.29 g, 19.72 mmol, 3.49 mL, 2.1 eq.) was added at 0° C., and the mixture was stirred at 20° C. for 4 h. The reaction was quenched by the addition of H2O (60 mL) at 20° C., and then extracted with EA (2×50 mL). The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 40 g SepaFlash® Silica Flash Column, Eluent of 0˜4% Ethyl acetate/Petroleum ether gradient @ 45 mL/min) to afford 59-2 (2.64 g, 5.35 mmol, 57% yield, 96% purity) as a colorless oil. LCMS: (ESI): m/z calcd. for C18H34BrN4O2Si2 475.13 [M+2+H]+, found 475.0.

To a solution of 59-2 (3.62 g, 7.64 mmol, 3 eq.) in THF (15 mL) at −15° C. was added iPrMgBr.LiCl (1.3 M in THF, 5.88 mL, 3 eq.) dropwise. The mixture was stirred at −15° C. for 10 min, then allowed to warm to 0° C., and stirred at 0° C. for 1 h. A solution of 59-3 (500 mg, 2.55 mmol, 1 eq.) in THF (5 mL) was added dropwise at −15° C., and the mixture was stirred at 0° C. for 10 min. The reaction was quenched by the addition of NH4Cl (sat. aq., 10 mL), diluted with H2O (40 mL) and extracted with EA (2×50 mL). The combined organic layers were washed with brine (30 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 40 g SepaFlash® Silica Flash Column, Eluent of 0˜8% Ethyl acetate/Petroleum ether gradient @ 45 mL/min) to afford 59-4 (1.28 g, 2.08 mmol, 82% yield, 96% purity) as a colorless oil. LCMS: (ESI): m/z calcd. for C29H51N4O5Si2 591.33 [M+H]+, found 591.3.

A mixture of 59-4 (680 mg, 1.10 mmol, 96% purity, 1.0 eq.) and 9-BBN dimer (668.43 mg, 2.76 mmol, 2.5 eq.) in THF (15 mL) was stirred at 50° C. for 1.5 h under N2, and then cooled to 20° C. A solution of K3PO4 (1.17 g, 5.52 mmol, 5 eq.) in H2O (3 mL) was added, and the mixture was stirred for 0.5 h. Q1 (462.6 mg, 1.33 mmol, 1.2 eq.) and Pd(dppf)Cl2 (80.8 mg, 0.11 mmol, 0.1 eq.) were added. The mixture was purged with N2 (3×) and stirred at 60° C. for 2 h. The mixture was diluted with H2O (30 mL) and extracted with EA (2×50 mL). The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 40 g SepaFlash® Silica Flash Column, Eluent of 0˜31% Ethyl acetate/Petroleum ether gradient @ 45 mL/min) to afford 59-5 (663 mg, 773.8 μmol, 70% yield, 95% purity) as a colorless oil. LCMS: (ESI): m/z calcd. for C38H58BrN6O5Si2 815.31 [M+2+H]+, found 815.3.

To a solution of 59-5 (663.0 mg, 773.80 μmol, 95% purity, 1 eq.) in DCM (10 mL) was added DAST (623.64 mg, 3.87 mmol, 511.18 μL, 5 eq.) at 0° C., and the mixture was stirred at 0° C. for 1 h. The reaction was quenched by addition sat. NaHCO3 (2mL), diluted with H2O (20 mL) and extracted with EA (2×50 mL). The combined organic layers were washed with brine (10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 40 g SepaFlash® Silica Flash Column, Eluent of 0˜27% Ethyl acetate/Petroleum ether gradient @ 45 mL/min) to afford 59-6 (249 mg, 303.45 μmol, 39% yield, 97% purity) as a white solid. LCMS: (ESI): m/z calcd. for C38H56BrN6O4Si2 797.30 [M+2+H]+, found 797.3.

To a solution of 59-6 (249 mg, 303.45 μmol, 97% purity, 1 eq.) in THF (6 mL) was added HCl (4 M, 2.91 mL, 38.36 eq.) at 20° C. under N2, and the mixture was stirred at 20° C. for 5 h. The mixture was concentrated under reduced pressure to give a residue. The residue was dissolved in a mixed solvent of H2O (12 mL) and t-BuOH (12 mL). PPTS (762.57 mg, 3.03 mmol, 10 eq.) was added at 20° C. The mixture was stirred at 50° C. for 12 h, and concentrated under reduced pressure to give a residue. The residue was purified by flash column (C18 column, 0.5% NH3.H2O-ACN), and then triturated with EA (2 mL) at 20° C. for 2 h. The solid was collected by filtration and dried under reduced pressure to afford 59-7 (80 mg, 142.12 μmol, 47% yield, 88% purity) as an off-white solid. LCMS: (ESI): m/z calcd. for C23H24BrN6O2 497.11 [M+2+H]+, found 497.2.

To a solution of 59-7 (50 mg, 88.8 μmol, 88% purity, 1 eq.) in MeOH (20 mL) was added Crabtree's catalyst (600 mg, 745.44 μmol, 8.39 eq.). The mixture was degassed under vacuum and purged with H2 several times, and stirred under H2 (15 psi) at 20° C. for 24 h. TMT (1, 3, 5-triazine-2, 4, 6-trithiol) was added, and the mixture was stirred at 20° C. for 15 min. The insoluble materials were removed by filtration. The filtrate was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, DCM: MeOH=10:1) to afford the impure product (13 mg, 23.52 μmol, 26% yield, 90% purity) as a yellow solid, which was combined with other batches to give 19 mg of the impure product. The impure product (19 mg) was purified by prep-HPLC (basic condition, column: Phenomenex Gemini-NX 80*40 mm*3 um; mobile phase: [water (0.05% NH3H2O+10 mM NH4HCO3)-ACN]; B%: 30%-50%, 8 min) to give 59 (9 mg, 21.06 μmol, 98% purity) as a white solid. TLCMS: (ESI): m/z calcd. for C23H26BrN6O2 499.12 [M+2+H]+, found 499.2. 1H NMR (400 MHz, CD3OD) δ: 8.25 (s, 1 H), 7.76 (s, 1 H), 7.55 (d, J=8.3 Hz, 1 H), 7.38 (s, 1 H), 7.15 (d, J=8.3 Hz, 1 H), 6.84 (d, J=4.3 Hz, 1 H), 6.54 (d, J=4.3 Hz, 1 H), 4.44 (t, J=6.8 Hz, 1 H), 3.86 (d, J=6.3 Hz, 1 H), 3.69-3.78 (m, 1 H), 2.70-2.88 (m, 2 H), 2.00-2.09 (m, 2 H), 1.72-1.79 (m, 2 H), 1.23 (s, 3 H).

Example 55 Compound 60

Compound 60 was prepared similarly as described for 58, using 58-8, and using Q16 instead of Q1. 1H NMR (400 MHz, CD3OD) δ: 8.21 (s, 1H), 8.19-8.05 (m, 1H), 7.28 (s, 1H), 7.25 (d, J=3.5 Hz, 1H), 6.96 (d, J=11.3 Hz, 1H), 6.61 (d, J=3.5 Hz, 1H), 5.06-4.96 (m, 1H), 4.66 (dd, J=5.3, 9.0 Hz, 1H), 3.99 (d, J=5.3 Hz, 1H), 3.71 (d, J=9.5 Hz, 1H), 3.51 (d, J=9.3 Hz, 1H), 3.45 (s, 3H), 2.91-2.70 (m, 2H), 2.17 (dd, J=9.4, 13.7 Hz, 1H), 2.00 (t, J=8.5 Hz, 2H), 1.91 (dd, J=9.8, 13.6 Hz, 1H); 19F NMR (376 MHz, CD3OD) δ: −125.91 (s, 1F)

Example 56 Description of LCMS Conditions

Run Method Flow time Code Instrument Column Mobile Phase Gradient mL/min (min) 1 A Xbridge A: water(4 L) + 0%-60% (solvent B) 0.8 7 Shield RP-18, NH3 · H2O(0.8 mL) over 6 minutes and 5 um, B: acetonitrile holding at 60% for 2.1 * 50 mm 0.5 minutes 2 A Xtimate C18 A: water(4 L) + 0%-30% (solvent B) 0.8 4 2.1 * 30 mm, TFA(1.5 mL) over 3 minutes and 3um B: acetonitrile holding at 30% for (4 L) + TFA(0.75 mL) 0.5 minutes 3 B Xtimate C18 A: water(4 L) + 0%-60% (solvent B) 1.2 2 2.1 * 30 mm, TFA(1.5 mL) over 0.9 minutes and 3um B: acetonitrile holding at 60% for (4 L) + TFA(0.75 mL) 0.6 minutes 4 A Xtimate C18 A: water(4 L) + 0%-30% (solvent B) 1.2 2 2.1 * 30 mm, TFA(1.5 mL) over 0.9 minutes and 3um B: acetonitrile holding at 30% for 0.6 minutes 5 A Xbridge A: water(4 L) + 0%-30% (solvent B) 1 3 Shield RP-18, NH3 · H2O(0.8 mL) over 2 minutes and 5 um, B: acetonitrile holding at 30% for 2.1 * 50 mm 0.48 minutes 6 A Xtimate C18 A: water(4 L) + 0%-60% (solvent B) 0.8 7 2.1 * 30 mm, TFA(1.5 mL) over 6 minutes and 3um B: acetonitrile holding at 60% for (4 L) + TFA(0.75 mL) 0.5 minutes 7 A Xbridge A: water(4 L) + 10%-80% (solvent B) 0.8 7 Shield RP-18, NH3 · H2O(0.8 mL) over 6 minutes and 5 um, B: acetonitrile holding at 80% for 2.1 * 50 mm 0.5 minutes 8 A Xbridge A: water(4 L) + 10%-80% (solvent B) 1 3 Shield RP-18, NH3 · H2O(0.8 mL) over 2 minutes and 5 um, B: acetonitrile holding at 80% 2.1 * 50 mm for 0.48 minutes 9 A Xtimate C18 A: water(4 L) + 0%-30% (solvent B) 0.8 7 2.1 * 30 mm, TFA(1.5 mL) over 6 minutes and 3um B: acetonitrile holding at 30% for (4 L) + TFA(0.75 mL) 0.5 minutes 10 A Xbridge A: water(4 L) + 0%-60% (solvent B) 1 3 Shield RP-18, NH3 · H2O(0.8 mL) over 2 minutes and 5 um, B: acetonitrile and holding at 60% 2.1 * 50 mm for 0.48 minutes 11 A Xtimate C18 A: water(4 L) + 0%-60% (solvent B) 0.8 7 2.1 * 30 mm, TFA(1.5 mL) over 6 minutes and 3um B: acetonitrile holding at 60% for for 0.5 minutes 12 A Xtimate C18 A: water(4 L) + 0%-30% (solvent B) 0.8 7 2.1 * 30 mm, TFA(1.5 mL) over 6 minutes and 3um B: acetonitrile holding at 30% for 0.5 minutes A = SHIMADZU LC20-MS2020; B = Agilent LC1200-MS6110; Column Temp. = 50° C.

Rt Method Compound (min) [M + H]+ code 1 4.125 499.1 1 2 2.663 517.2 2 [M + Na]+ 3 1.177 516.1 3 4 1.299 495.2 4 5 2.546 419.2 5 6 2.498 419.2 5 7 4.160 421.0 1 8 4.209 421.1 1 9 4.051 503.1 1 10 4.114 433.3 1 11 4.364 487.1 1 12 2.604 419.3 7 13 3.681 417.2 1 14 4.448 459.2 1 15 1.548 497.1 8 16 3.260 407.2 1 17 2.022 499.1 1 18 4.048 453.3 9 19 4.187 451.2 1 20 2.040 533.5 8 20A 4.163 533.3 7 21 2.596 436.2 6 21A 2.610 436.3 6 22 1.705 418.2 10 23 2.045 469.2 10 24 3.333 420.3 1 25A 4.064 557.3 7 26 4.241 559.5 7 26-A 4.168 559.3 7 27 1.946 444.2 10 28 1.664 467.2 8 29 1.760 450.2 10 30 4.450 471.2 9 31 1.939 475.2 10 32 2.892 470.3 6 33 4.180 567.2 7 34 1.794 488.4 8 35 1.764 423.2 10 36 4.249 475.1 1 37 3.764 437.2 9 38 2.561 418.3 2 39 2.905 470.2 2 40 3.457 487.3 9 41 2.912 455.2 11 42 4.523 454.1 12 43 3.710 488.2 7 44 4.259 482.2 1 45 3.745 500.2 1 46 4.207 431.2 1 47 3.688 397.3 1 48 4.037 419.3 1 49 3.176 470.1 6 50 3.275 516.1 6 51 1.724 483.3 8 52 3.197 481.2 7 53 3.593 527.1* 6 54 3.342 533.1** 6 55 4.501 533.4 7 56 3.760 420.3 1 57 4.527 471.16 1 58 3.106 529.2 7 59 4.311 499.2** 1 60 3.299 501.2 7 *[M + Na]+ **[M + 2 + H]+

Example 57 Additional Compounds

The foregoing syntheses are exemplary and can be used as a starting point to prepare a large number of additional compounds. Examples of compounds of Formula (I), and pharmaceutically acceptable salts thereof, that can be prepared in various ways, including those synthetic schemes shown and described herein, are provided below. Those skilled in the art will be able to recognize modifications of the disclosed syntheses and to devise routes based on the disclosures herein; all such modifications and alternate routes are within the scope of the claims.

(including pharmaceutically acceptable salts of any of the foregoing).

Example A PRMT5/MEP50 Enzyme Inhibition Assay Assay 1

Compounds were tested for inhibition of methyltransferase activity in a radioisotope filter binding assay, similar to previously described in A selective inhibitor of PRMT5 with in vivo and in vitro potency in MCL models. Chan-Penebre et al., Nat Chem Biol. (2015) 11(6):432-7. In the standard PRMT5/MEP50 enzyme inhibition assay, compounds were tested in a 10-dose IC50 mode with 3- or 5-fold serial dilution, in singlet, starting at 1, 10, or 100 μM. Control compound, SAH (S-(5′-Adenosyl)-L-homocysteine), was tested in 10-dose IC50 mode with 3-fold serial dilution starting at 100 μM. Reactions were carried out at 1 μM 3H-SAM (PerkinElmer) and 5 μM histone H2A as substrates for methyl transfer. Following a 60 min incubation at 30° C., the reaction was stopped with 20% TCA. Each reaction was spotted on a filter plate (MultiScreen FB Filter plate, Millipore) and washed 5 times in PBS, after which scintillation fluid was added and signal was detected in a scintillation counter. Percent enzyme activity was calculated based on no inhibitor DMSO control as 100% activity. EC50 values were determined in GraphPad Prism 8 using the [inhibitor] vs. response—Variable slope (four parameters) function.

Assay 2

Compounds were tested for inhibition of methyltransferase activity in 384-well plate assay format using mass spectrometry technology. In this enzyme inhibition assay, compounds were tested in a 11-dose IC50 mode with 3-serial dilution, in duplicate, starting at 1, 10, or 100 μM. Reactions were carried out at 1 μM SAM and 0.1 μM histone H4 1-21 peptide as substrates for methyl transfer. Following an 18-hour incubation at rt, the reaction was stopped with 0.5% formic acid. Products of the reaction were captured while unreacted substrates were washed away, prior to MALDI mass spectrometry detection and analysis. Percent enzyme activity was calculated based on no inhibitor DMSO control as 100% activity. EC50 values were determined in GraphPad Prism 8 using the [inhibitor] vs. response—Variable slope (four parameters) function.

The IC50 values were derived from the procedure as described herein and are shown in Table 1. Compounds of Formula (I) show activity in this assay. A value of ‘A’ in the table below indicates an IC50 of <1 nM, a value of ‘B’ indicates an IC50≥1 nM to ≤100 nM, and a value of ‘C’ indicates an IC50 value of >100 nM.

TABLE 1 Assay 1 Assay 2 Compound: IC50 IC50 1 A A 2 A A 3 C C 4 A A 5 A A 6 B B 7 A B 8 B C 9 A A 10 A A 11 C B 12 A A 13 A A 14 A A 15 A 16 A 17 C 18 A 19 A 20 B 20-A C 21 A 21-A B 22 A 23 A 24 A 25-A C 26 C 26-A C 27 A 28 A 29 A 30 A 31 A 34 A 35 A 36 A 37 A 38 A 39 A 40 A 41 A 42 A 43 A 44 A 45 A 46 A 47 A 48 A 49 A 50 A 51 A 52 A 53 A 54 A 55 C 56 A 57 A 58 A 59 A

Example B Cell Proliferation Assays

HepG2 hepatoma cells (ATCC, HB-8065) were maintained in DMEM with high glucose (Lonza, 12-914F) supplemented with 10% fetal bovine serum (FBS; Biowest, S181B-500) and 2 mM L-glutamine (Biowest, X0551-100) at 37° C. and 5% CO2. A549 lung carcinoma cells (ATCC, CCL-185) were maintained in F-12K medium (ThermoFisher 21127030) supplemented with 10% FBS at 37° C. and 5% CO2. Exponentially growing HepG2 or A549 were plated in white, clear-bottom 96-well plates (Corning, 3903) at a cell density of 2000 (HepG2) or 350 (A549) cells per well in 199 μL of HepG2 medium. Next, 1μL of a 5-fold 9-point dilution series of test compound in DMSO was added to the different wells. Cells were incubated at 37° C. 5% CO2 for 7 days. Cell viability was assessed on day 7 with the CellTiter-Glo 2.0 Cell Viability assay kit (Promega, G9243) to quantify the intracellular amounts of ATP: first, 100 μL of cell culture medium was removed from each well, next 100 of CellTiter-Glo® 2.0 Reagent was added and plates were shaken for 2 minutes on an orbital shaker. After 10 minutes stabilization at room temperature, read-out was performed on a ThermoFisher VarioSkan Lux plate reader. Counts were normalized to DMSO control (0% inhibition) and the cytotoxic control (100% inhibition) and EC50 values were determined in GraphPad Prism 8 using the [Agonist] vs. response—Variable slope (four parameters) function.

The EC50 values were derived from the procedure as described herein and are shown in Table 2. As shown by the data of Table 2, compounds of Formula (I) have activity in this assay. A value of ‘A’ in the table below indicates an EC50 of <20 nM, a value of ‘B’ indicates an EC50≥20 nM to <100 nM, a value of ‘C’ indicates an EC50 value of EC50≥100 nM to <1000 nM and a value of ‘D’ indicates an EC50≥1000 nM

TABLE 2 HepG2 and A549 EC50 EC50 Compound HepG2 A549 1 A A 2 A A 3 C C 4 A A 5 C C 6 D D 7 C C 8 C D 9 B B 10 A A 11 C D 12 A A 13 A A 14 A A 15 C C 16 D D 17 D D 18 A A 19 A A 20 A B 20-A C C 21 A A 21-A D D 22 C C 23 A A 24 B B 25-A D D 26 B C 26-A D D 27 B B 28 B C 29 A B 30 A A 31 C D 32 A A 33 A A 34 A A 35 A A 36 A A 37 A A 38 A A 39 B B 40 A A 41 A A 42 A A 43 B A 44 C B 45 C C 46 C C 47 C B 48 A A 49 A A 50 A A 51 A A 52 A A 53 A A 54 A A 55 B B 56 A A

Although the foregoing has been described in some detail by way of illustrations and examples for purposes of clarity and understanding, it will be understood by those of skill in the art that numerous and various modifications can be made without departing from the spirit of the present disclosure. Therefore, it should be clearly understood that the forms disclosed herein are illustrative only and are not intended to limit the scope of the present disclosure, but rather to also cover all modification and alternatives coming with the true scope and spirit of the invention.

Claims

1. A compound of Formula (I), or a pharmaceutically acceptable salt thereof, having the structure:

wherein:
B1 is an optionally substituted
,
wherein X1 is CRC1; X2 is CRC2; X3 is CRC3; and RC1, RC2, RC3, RC4 and RC5 are independently hydrogen or halogen or an unsubstituted C1-4 alkyl;
R1B is hydrogen, halogen, hydroxy, an unsubstituted C1-4 alkyl, an unsubstituted C2-4 alkenyl, an unsubstituted C3-C6 cycloalkyl, an unsubstituted C1-4 alkoxy or NRA1RA2; and
RA1 and RA2 are each hydrogen;
R1 is hydrogen;
R2A is hydrogen;
R2B is OH or —O—C(═O)—C1-4 alkyl;
R3A is hydrogen;
R3B is OH or —O—C(═O)—C1-4 alkyl;
R4A is —(CRD1RE1)(CRD2RE2)n-RF1;
wherein RD1, RE1, RD2 and RE2 are independently selected from the group consisting of hydrogen, halogen, hydroxy and an unsubstituted C1-3 alkyl; n is 1; and RF1 is an unsubstituted or a substituted aryl, an unsubstituted or a substituted heteroaryl or an unsubstituted or a substituted heterocyclyl;
R4B is an unsubstituted C1-4 alkyl,
Z1 is CR5AR5B;
R5A and R5B are each hydrogen.

2.-18. (canceled)

19. The compound of claim 1, wherein R2B is OH.

20. The compound of claim 1, wherein R2B is —O—C(═O)—C1-4 alkyl.

21.-31. (canceled)

32. The compound of claim 1, wherein R3B is OH.

33. The compound of claim 1, wherein R3B is —O—C(═O)—C1-4 alkyl.

34.-45. (canceled)

46. The compound of claim 1, wherein RD1, RE1, RD2 and RE2 are each hydrogen.

47.-56. (canceled)

57. The compound of claim 1, wherein RF1 is an unsubstituted or a substituted heteroaryl.

58. (canceled)

59. The compound of claim 57, wherein RF1 is pyridinyl.

60. (canceled)

61. The compound of claim 59, wherein RF1 is substituted with one, two or three substituents independently selected from the group consisting of halogen, cyano, an unsubstituted C1-4 alkyl, an unsubstituted C1-4 haloalkyl, an unsubstituted monocyclic C3-6 cycloalkyl, an optionally substituted C-carboxy, an optionally substituted N-amido, amino, a mono-substituted amine, a di-substituted amine, —NH—C(═O)-unsubstituted C1-8 alkyl, —NH—C(═O)—O-unsubstituted C1-8 alkyl, —NH—C(═O)-unsubstituted C3-6 cycloalkyl and —NH—C(═O)—O-unsubstituted C3-6 cycloalkyl.

62. The compound of claim 1, wherein RF1 is selected from the group consisting of:

63.-105. (canceled)

106. The compound of claim 1, wherein R4B is CH3.

107.-134. (canceled)

135. The compound of claim 1, wherein R1B is NRA1RA2, wherein RA1 and RA2 are each hydrogen.

136.-142. (canceled)

143. The compound of claim 1, wherein B1 is

144.-147. (canceled)

148. The compound of claim 1, wherein the compound is or a pharmaceutically acceptable salt thereof.

149.-154. (canceled)

155. The compound of claim 148, wherein B1 is

156. (canceled)

157. (canceled)

158. The compound of claim 148, wherein RF1 is pyridinyl.

159. (canceled)

160. The compound of claim 158, wherein RF1 is substituted with one, two or three substituents independently selected from the group consisting of halogen, cyano, an unsubstituted C1-4 alkyl, an unsubstituted C1-4 haloalkyl, an unsubstituted monocyclic C3-6 cycloalkyl, an optionally substituted C-carboxy, an optionally substituted N-amido, amino, a mono-substituted amine, a di-substituted amine, —NH—C(═O)-unsubstituted C1-8 alkyl, —NH—C(═O)—O-unsubstituted C1-8 alkyl, —NH—C(═O)-unsubstituted C3-6 cycloalkyl and —NH—C(═O)—O-unsubstituted C3-6 cycloalkyl.

161. The compound of claim 1, wherein the compound is selected from the group consisting of: or a pharmaceutically acceptable salt of any of the foregoing.

162. A pharmaceutical composition comprising an effective amount of a compound of claim 1, or a pharmaceutically acceptable salt thereof, and excipient.

163-174. (canceled)

175. A method for treating a cancer comprising administering an effective amount of a compound of claim 1, or a pharmaceutically acceptable salt thereof, to a subject in need thereof.

176. (canceled)

177. A method for modulating PRMT5 comprising contacting the PRMT5 enzyme with an effective amount of a compound of claim 1, or a pharmaceutically acceptable salt thereof.

178. A method for inhibiting PRMT5 comprising contacting the PRMT5 enzyme Use of a compound of claim 1, or a pharmaceutically acceptable salt thereof.

179. The method of claim 175, further comprises the use of an additional agent selected from the group consisting of a kinase inhibitor, a checkpoint inhibitor/modulator and an anti-VEGF antibody.

180. (canceled)

Patent History
Publication number: 20220048922
Type: Application
Filed: Oct 28, 2021
Publication Date: Feb 17, 2022
Inventors: Koen Vandyck (Paal), Pierre Jean-Marie Bernard Raboisson (Wavre), Jerome Deval (Pacifica, CA), Leonid Beigelman (San Mateo, CA), David McGowan (Brussels), Yannick Debing (Bilzen)
Application Number: 17/452,689
Classifications
International Classification: C07D 495/04 (20060101); A61P 35/00 (20060101); C07D 513/04 (20060101); C07D 519/00 (20060101); C07D 487/04 (20060101); C07D 473/34 (20060101); A61K 31/53 (20060101); A61K 31/52 (20060101); A61K 31/519 (20060101); A61K 31/522 (20060101); C07D 473/30 (20060101); A61K 31/437 (20060101); C07D 473/26 (20060101); C07D 471/04 (20060101);