Tetracyclic heterocycle compounds and methods of use thereof for the treatment of hepatitis C

Information

  • Patent Grant
  • 9243002
  • Patent Number
    9,243,002
  • Date Filed
    Monday, February 3, 2014
    10 years ago
  • Date Issued
    Tuesday, January 26, 2016
    8 years ago
Abstract
The present invention relates to compounds of formula I that are useful as hepatitis C virus (HCV) NS5B polymerase inhibitors, the synthesis of such compounds, and the use of such compounds for inhibiting HCV NS5B polymerase activity, for treating or preventing HCV infections and for inhibiting HCV viral replication and/or viral production in a cell-based system.
Description
FIELD OF THE INVENTION

The present disclosure relates to antiviral compounds that are useful as inhibitors of the hepatitis C virus (HCV) NS5B (non-structural protein 5B) polymerase, compositions comprising such compounds, the use of such compounds for treating HCV infection and/or reducing the likelihood or severity of symptoms of HCV infection, methods for inhibiting the function of the NS5B polymerase, and methods for inhibiting HCV viral replication and/or viral production.


CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a §371 National Stage Application of PCT/US2014/014361, international filing date of Feb. 3, 2014, which claims the benefit of International Application No. PCT/CN2013/000128, filed Feb. 7, 2013, now expired.


BACKGROUND OF THE INVENTION

Hepatitis C virus (HCV) infection is a major health problem that leads to chronic liver disease, such as cirrhosis and hepatocellular carcinoma, in a substantial number of infected individuals. Current treatments for HCV infection include immunotherapy with recombinant interferon-α alone or in combination with the nucleoside analog ribavirin.


Several virally-encoded enzymes are putative targets for therapeutic intervention, including a metalloprotease (NS2-3), a serine protease (NS3, amino acid residues 1-180), a helicase (NS3, full length), an NS3 protease cofactor (NS4A), a membrane protein (NS4B), a zinc metalloprotein (NS5A) and an RNA-dependent RNA polymerase (NS5B).


One identified target for therapeutic intervention is HCV NS5B polymerase. Sven-Erik Behrens et al., Identification and properties of the RNA-dependent RNA polymerase of heptatitis C virus, 15(1) EMBO J. 12-22 (1996). Antagonists of NS5B activity are inhibitors of HCV replication. Steven S. Carroll et al., Inhibition of Hepatitis C Virus RNA Replication by 2′-Modified Nucleoside Analogs, 278 (14) J. BIOL. CHEM. 11979-84 (2003).


There is a clear and long-felt need to develop effective therapeutics for treatment of HCV infection. Specifically, there is a need to develop compounds that selectively inhibit HCV viral replication and that would be useful for treating HCV-infected patients.


SUMMARY OF THE INVENTION

The present disclosure relates to novel compounds of formula I and/or pharmaceutically acceptable salts thereof. These compounds are useful, either as compounds or their pharmaceutically acceptable salts (when appropriate), in the inhibition of HCV (hepatitis C virus) NS5B (non-structural 5B) polymerase, the prevention or treatment of one or more of the symptoms of HCV infection, the inhibition of HCV viral replication and/or HCV viral production, and/or as pharmaceutical composition ingredients. As pharmaceutical composition ingredients, these compounds and their salts may be the primary active therapeutic agent, and, when appropriate, may be combined with other therapeutic agents including but not limited to other HCV antivirals, anti-infectives, immunomodulators, antibiotics or vaccines, as well as the present Standard of Care treatment options for HCV.


In one aspect, the present invention relates to a compound of formula I:




embedded image



or a pharmaceutically acceptable salt thereof, wherein:


A is a C3-C6 cycloalkyl or an aromatic ring system selected from:

    • (i) 5-6 membered monocyclic ring with 0, 1, 2 or 3 heteroatom ring atoms selected from N, S or O, optionally substituted with 1 or 2 substituents independently selected from C1-C6 alkyl, C1-C6 alkoxy, C1-C6 haloalkyl, cyano, oxo, halo, —O—C1-C6 haloalkyl, C1-C6 hydroxyalkyl, C3-C6cycloalkyl, phenyl, pyrrolidinyl, and —O—C3-C6cycloalkyl; and
    • (ii) 8-10 membered bicyclic rings with 2 or 3 heteroatom ring atoms selected from N and O;




embedded image



is




embedded image


Z is N or CRa;


Z1, Z2 and Z3 are independently selected from CH and N, wherein 1 or 2 of Z1, Z2 and Z3 are N;


p is 0, 1, or 2;


q is 0 or 1;


R2 is hydrogen, C1-C6 alkyl or —O—C1-C6 alkyl;


R3 is hydrogen or C1-C6 alkyl;


R4 is C1-C6 alkyl or —O—C1-C6 alkyl;


R5 is hydrogen, C1-C6 alkyl, —OH, halo, or a 4- to 7-membered heterocycloalkyl substituted with halo;


R9 is hydrogen; or


R5 and R9 together with the C to which they are attached form a C3-C6cycloalkyl,


R6 is hydrogen, —OH, C1-C6 alkyl, C1-C6 haloalkyl, C1-C6 hydroxyalkyl, —CH2SO2CH3, —C1-C6-(4- to 6-membered monocyclic heterocycloalkyl);


wherein the 4- to 6-membered monocyclic heterocycloalkyl is optionally substituted with one or two F substituents;


R7 is hydrogen or C1-C6 alkyl;


or R6 and R7 together with the C to which they are attached form an oxo; R8 is hydrogen, halo or —ORb;


R10 is hydrogen or C1-C6 alkyl;


Ra is hydrogen, halo, or cyano;


each Rb is independently H or C1-C6 alkyl;


wherein when q is 0, p is 0 or 1;

    • when q is 1 and p is 2, custom character is a single bond; and
    • when custom character is a double bond, R7 and R9 are absent.


The present invention also includes pharmaceutical compositions containing a compound of the present invention and methods of preparing such pharmaceutical compositions. The present invention further includes methods of treating or reducing the likelihood or severity of HCV infection, methods for inhibiting the activity of the NS5B polymerase, and methods for inhibiting HCV viral replication and/or viral production.


Other embodiments, aspects and features of the present invention are either further described in or will be apparent from the ensuing description, examples and appended claims.







DETAILED DESCRIPTION OF THE INVENTION

The present invention includes compounds of formula I above, and pharmaceutically acceptable salts thereof. The compounds of formula I are HCV NS5B polymerase inhibitors.


For purposes of illustration, the portion of formula I represented by




embedded image



can be




embedded image


In a first embodiment of the invention, R2 and R4 are independently C1-C6 alkyl. In this embodiment, all other groups are as provided in the general formula above.


In a second embodiment of the invention, R2, R3 and R4 are methyl. In this embodiment, all other groups are as provided in the general formula above and/or in the first embodiment.


In a third embodiment of the invention, each halo is F. In this embodiment, all other groups are as provided in the general formula above and/or in the first or second embodiments.


In a fourth embodiment of the invention, the compound of the invention has the formula:




embedded image



or a pharmaceutically acceptable salt thereof. In this embodiment, all other groups are as provided in the general formula above and/or in the first through third embodiments.


In a fifth embodiment of the invention,


R5 is hydrogen or methyl;


R6 is hydrogen, methyl, —CH2OH, —CH2CH2OH, —CH2SO2CH3,




embedded image



and


R7 is hydrogen or methyl.


In this embodiment, all other groups are as provided in the general formula above and/or in the first through fourth embodiments.


In a sixth embodiment of the invention, A is C3-C6 cycloalkyl, or a 5-6 membered aromatic monocyclic ring with 0, 1, 2 or 3 heteroatom ring atoms selected from N and S, optionally substituted with 1 or 2 substituents independently selected from C1-C6 alkyl, C1-C6 alkoxy, C1-C6 haloalkyl, oxo, halo, and —O—C1-C6 haloalkyl. In an aspect of this seventh embodiment, A is cyclopropyl,




embedded image



wherein R1a, R1b, R1c and R1d are independently selected from hydrogen, F, methyl, ethyl, hydroxymethyl, isopropyl, methoxy, ethoxy, —OCHF2, —OCH2CF3, cyclopropyl, phenyl, pyrrolidinyl, —O-cyclopropyl, —CHF2, cyano, —CF3 and —CH2CF3. In another aspect of this embodiment, A is




embedded image



In this embodiment, all other groups are as provided in the general formula above and/or in the first through fifth embodiments.


In another embodiment of the invention, the compound of the invention is selected from the exemplary species depicted in Examples 1 through 99 shown below, and pharmaceutically acceptable salts thereof


In another embodiment of the invention, the compound of the invention is selected from the exemplary species depicted in Examples 1, 15, 30, 34, 38, 42, 45, 46, 54, 55, 56, 60, 63, 64, 65, 82, 85, 88, 90 and 94 shown below, and pharmaceutically acceptable salts thereof


Other embodiments of the present invention include the following:


(a) A pharmaceutical composition comprising an effective amount of a compound of formula I and a pharmaceutically acceptable carrier.


(b) The pharmaceutical composition of (a), further comprising a second therapeutic agent selected from the group consisting of HCV antiviral agents, immunomodulators, and anti-infective agents.


(c) The pharmaceutical composition of (b), wherein the HCV antiviral agent is an antiviral selected from the group consisting of direct inhibitors of HCV, including but not limited to NS3 and NS3/4A protease inhibitors, NS5A inhibitors and HCV NS5B polymerase inhibitors.


(d) A pharmaceutical combination that is (i) a compound of formula I and (ii) a second therapeutic agent selected from the group consisting of HCV antiviral agents, immunomodulators, and anti-infective agents; wherein the compound of formula I and the second therapeutic agent are each employed in an amount that renders the combination effective for inhibiting HCV NS5B activity, or for inhibiting HCV viral replication, or for treating HCV infection and/or reducing the likelihood or severity of symptoms of HCV infection.


(e) The combination of (d), wherein the HCV antiviral agents are one or more antiviral agents selected from the group consisting of direct inhibitors of HCV, including but not limited to NS3 and NS3/4A protease inhibitors, NS5A inhibitors and HCV NS5B polymerase inhibitors.


(f) A use of a compound of formula I in the preparation of a medicament for inhibiting HCV NS5B activity in a subject in need thereof


(g) A use of a compound of formula I in the preparation of a medicament for preventing and/or treating infection by HCV in a subject in need thereof


(h) A method of treating HCV infection and/or reducing the likelihood or severity of symptoms of HCV infection in a subject in need thereof, which comprises administering to the subject an effective amount of a compound of formula I.


(i) The method of (h), wherein the compound of formula I is administered in combination with an effective amount of at least one second therapeutic agent selected from the group consisting of HCV antiviral agents, immunomodulators, and anti-infective agents.


(j) The method of (i), wherein the HCV antiviral agent is an antiviral selected from the group consisting of direct inhibitors of HCV, including but not limited to NS3 and NS3/4A protease inhibitors, NS5A inhibitors and HCV NS5B polymerase inhibitors.


(k) A method of inhibiting HCV viral replication and/or HCV viral production in a cell-based system, which comprises administering to the subject an effective amount of a compound of formula I in combination with an effective amount of at least one second therapeutic agent selected from the group consisting of HCV antiviral agents, immunomodulators, and anti-infective agents.


(l) The method of (k), wherein the HCV antiviral agent is an antiviral selected from the group consisting of direct inhibitors of HCV, including but not limited to NS3 and NS3/4A protease inhibitors, NS5A inhibitors and HCV NS5B polymerase inhibitors.


(m) A method of inhibiting HCV NS5B activity in a subject in need thereof, which comprises administering to the subject the pharmaceutical composition of (a), (b), or (c) or the combination of (d) or (e).


(n) A method of treating HCV infection and/or reducing the likelihood or severity of symptoms of HCV infection in a subject in need thereof, which comprises administering to the subject the pharmaceutical composition of (a), (b), or (c) or the combination of (d) or (e).


In the embodiments of the compounds and salts provided above, it is to be understood that each embodiment may be combined with one or more other embodiments, to the extent that such a combination provides a stable compound or salt and is consistent with the description of the embodiments. It is further to be understood that the embodiments of compositions and methods provided as (a) through (n) above are understood to include all embodiments of the compounds and/or salts, including such embodiments as result from combinations of embodiments.


Additional embodiments of the invention include the pharmaceutical compositions, combinations, uses and methods set forth in (a) through (n) above, wherein the compound of the present invention employed therein is a compound of one of the embodiments, aspects, classes, sub-classes, or features of the compounds described above. In all of these embodiments, the compound may optionally be used in the form of a pharmaceutically acceptable salt or hydrate as appropriate.


The present invention also includes a compound of the present invention for use (i) in, (ii) as a medicament for, or (iii) in the preparation of a medicament for: (a) inhibiting HCV NS5B activity, or (b) inhibiting HCV viral replication, or (c) treating HCV infection and/or reducing the likelihood or severity of symptoms of HCV infection, or (d) use in medicine. In these uses, the compounds of the present invention can optionally be employed in combination with one or more second therapeutic agents selected from HCV antiviral agents, anti-infective agents, and immunomodulators.


Chemical names, common names, and chemical structures may be used interchangeably to describe the same structure. If a chemical compound is referred to using both a chemical structure and a chemical name and an ambiguity exists between the structure and the name, the structure is understood to predominate.


As used herein, the term “administration” and variants thereof (e.g., “administering” a compound) in reference to a compound of the invention mean providing the compound to the individual in need of treatment. When a compound of the invention is provided in combination with one or more other active agents (e.g., antiviral agents useful for treating HCV infection), “administration” and its variants are each understood to include concurrent and sequential provision of the compound or salt and other agents.


The term “alkoxy” refers to an “alkyl-O-” group. Alkoxy groups may be substituted as indicated.


The term “alkyl” refers to an aliphatic hydrocarbon group having one of its hydrogen atoms replaced with a bond. An alkyl group may be straight or branched and contain from about 1 to about 20 carbon atoms. In one embodiment, an alkyl group contains from about 1 to about 12 carbon atoms. In different embodiments, an alkyl group contains from 1 to 6 carbon atoms (C1-C6 alkyl) or from about 1 to about 3 carbon atoms (C1-C3 alkyl). Non-limiting examples of alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, neopentyl, isopentyl, n-hexyl, isohexyl and neohexyl. An alkyl group may be unsubstituted or substituted by one or more substituents which may be the same or different, each substituent being independently selected from the group consisting of halo, alkenyl, alkynyl, aryl, cycloalkyl, cyano, hydroxy, —O-alkyl, —O-aryl, -alkylene-O-alkyl, alkylthio, —NH2, —NH(alkyl), —N(alkyl)2, —NH(cycloalkyl), —O—C(O)-alkyl, —O—C(O)-aryl, —O—C(O)-cycloalkyl, —C(O)OH and —C(O)O-alkyl. In one embodiment, an alkyl group is linear. In another embodiment, an alkyl group is branched. Unless otherwise indicated, an alkyl group is unsubstituted.


The term “aryl” (or “aryl ring system”) refers to aromatic mono- and poly-carbocyclic ring systems wherein the individual carbocyclic rings in the polyring systems are fused or attached to each other via a single bond. As used herein, the term aryl includes aromatic mono- and poly-carbocyclic ring systems that include from 0 to 4 heteroatoms (non-carbon atoms) that are independently chosen from N, O and S. Suitable aryl groups include phenyl, naphthyl, biphenylenyl, pyridinyl, pyrimidinyl and pyrrolyl, as well as those discussed below. Aryl groups may be substituted as indicated. Aryl ring systems may include, where appropriate, an indication of the variable to which a particular ring atom is attached. Unless otherwise indicated, substituents to the aryl ring systems can be attached to any ring atom, provided that such attachment results in formation of a stable ring system.


The term “carbocycle” (and variations thereof such as “carbocyclic”) as used herein, unless otherwise indicated, refers to (i) a C5 to C7 monocyclic, saturated or unsaturated ring, or (ii) a C8 to C10 bicyclic saturated or unsaturated ring system. Each ring in (ii) is either independent of, or fused to, the other ring, and each ring is saturated or unsaturated. Carbocycle groups may be substituted as indicated. When the carbocycles contain one or more heteroatoms independently chosen from N, O and S, the carbocycles may also be referred to as “heterocycles,” as defined below. The carbocycle may be attached to the rest of the molecule at any carbon or nitrogen atom that results in a stable compound. The fused bicyclic carbocycles are a subset of the carbocycles; i.e., the term “fused bicyclic carbocycle” generally refers to a C8 to C10 bicyclic ring system in which each ring is saturated or unsaturated and two adjacent carbon atoms are shared by each of the rings in the ring system. A fused bicyclic carbocycle in which both rings are saturated is a saturated bicyclic ring system. Saturated carbocyclic rings are also referred to as cycloalkyl rings, e.g., cyclopropyl, cyclobutyl, etc. A fused bicyclic carbocycle in which one or both rings are unsaturated is an unsaturated bicyclic ring system. Carbocycle ring systems may include, where appropriate, an indication of the variable to which a particular ring atom is attached. Unless otherwise indicated, substituents to the ring systems can be attached to any ring atom, provided that such attachment results in formation of a stable ring system.


The term “composition” is intended to encompass a product comprising the specified ingredients, as well as any product which results, directly or indirectly, from combining the specified ingredients.


The term “compound” is intended to encompass chemical agents described by generic formula I in all forms, including hydrates and solvates of such chemical agents.


The term “cycloalkyl,” as used herein, refers to a non-aromatic mono- or multicyclic ring system comprising from about 3 to about 10 ring carbon atoms. In one embodiment, a cycloalkyl contains from about 5 to about 10 ring carbon atoms. In another embodiment, a cycloalkyl contains from about 3 to about 7 ring atoms. In another embodiment, a cycloalkyl contains from about 5 to about 7 ring atoms. In another embodiment, a cycloalkyl contains from about 5 to about 6 ring atoms. The term “cycloalkyl” also encompasses a cycloalkyl group, as defined above, which is fused to an aryl (e.g., benzene) or heteroaryl ring. Non-limiting examples of monocyclic cycloalkyls include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl. Non-limiting examples of multicyclic cycloalkyls include 1-decalinyl, norbornyl and adamantyl. A cycloalkyl group can be optionally substituted with one or more “ring system substituents” which may be the same or different, and are as defined herein below. In one embodiment, a cycloalkyl group is unsubstituted. The term “3 to 7-membered cycloalkyl” refers to a cycloalkyl group having from 3 to 7 ring carbon atoms. Unless otherwise indicated, a cycloalkyl group is unsubstituted. A ring carbon atom of a cycloalkyl group may be functionalized as a carbonyl group. An illustrative example of such a cycloalkyl group (also referred to herein as a “cycloalkanoyl” group) includes, but is not limited to, cyclobutanoyl:




embedded image


The term “effective amount” as used herein means that amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue, system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician. In one embodiment, the effective amount is a “therapeutically effective amount” for the alleviation of one or more symptoms of the disease or condition being treated. In another embodiment, the effective amount is a “prophylactically effective amount” for reduction of the severity or likelihood of one or more symptoms of the disease or condition. In another embodiment, the effective amount is a “therapeutically effective amount” for inhibition of HCV viral replication and/or HCV viral production. The term also includes herein the amount of active compound sufficient to inhibit HCV NS5B activity and thereby elicit the response being sought (i.e., an “inhibition effective amount”). When the active compound (i.e., active ingredient) is administered as the salt, references to the amount of active ingredient are to the free acid or free base form of the compound.


The term “haloalkyl,” as used herein, refers to an alkyl group as defined above, wherein one or more of the alkyl group's hydrogen atoms has been replaced with a halogen. In one embodiment, a haloalkyl group has from 1 to 6 carbon atoms. In another embodiment, a haloalkyl group is substituted with from 1 to 3 F atoms. Non-limiting examples of haloalkyl groups include —CH2F, —CHF2, —CF3, —CH2Cl and —CCl3. The term “C1-C6 haloalkyl” refers to a haloalkyl group having from 1 to 6 carbon atoms.


The term “halogen” (or “halo”) refers to atoms of fluorine, chlorine, bromine and iodine (alternatively referred to as fluoro, chloro, bromo, and iodo).


The term “heteroaryl,” as used herein, refers to an aromatic monocyclic or multicyclic ring system comprising about 5 to about 14 ring atoms, wherein from 1 to 4 of the ring atoms is independently O, N or S and the remaining ring atoms are carbon atoms. In one embodiment, a heteroaryl group has 5 to 10 ring atoms. In another embodiment, a heteroaryl group is monocyclic and has 5 or 6 ring atoms. In another embodiment, a heteroaryl group is bicyclic and has 9 or 10 ring atoms. A heteroaryl group can be optionally substituted by one or more “ring system substituents” which may be the same or different, and are as defined herein below. A heteroaryl group is joined via a ring carbon atom, and any nitrogen atom of a heteroaryl can be optionally oxidized to the corresponding N-oxide. The term “heteroaryl” also encompasses a heteroaryl group, as defined above, which is fused to a benzene ring. The term “heteroaryl” also encompasses any fused polycyclic ring system containing at least one ring heteroatom selected from N, O and S, wherein at least one ring of the fused polycyclic ring system is aromatic. For example, the term “9 to 10-membered bicyclic heteroaryl” encompasses a non-aromatic 5 membered heterocyclic ring that is fused to a benzene or pyridyl ring. Non-limiting examples of heteroaryls include pyridyl, pyrazinyl, furanyl, thienyl, pyrimidinyl, pyridone (including N-substituted pyridones), isoxazolyl, isothiazolyl, oxazolyl, oxadiazolyl, thiazolyl, pyrazolyl, furazanyl, pyrrolyl, triazolyl, 1,2,4-thiadiazolyl, pyrazinyl, pyridazinyl, quinoxalinyl, phthalazinyl, oxindolyl, imidazo[1,2-a]pyridinyl, imidazo[2,1-b]thiazolyl, benzofurazanyl, indolyl, azaindolyl, benzimidazolyl, benzothienyl, quinolinyl, imidazolyl, benzimidazolyl, thienopyridyl, quinazolinyl, thienopyrimidyl, pyrrolopyridyl, imidazopyridyl, isoquinolinyl, benzoazaindolyl, 1,2,4-triazinyl, benzothiazolyl and the like, and all isomeric forms thereof. The term “heteroaryl” also refers to partially saturated heteroaryl moieties such as, for example, tetrahydroisoquinolyl, tetrahydroquinolyl and the like. In one embodiment, a heteroaryl group is a 5-membered heteroaryl. In another embodiment, a heteroaryl group is a 6-membered heteroaryl. In another embodiment, a heteroaryl group comprises a 5- to 6-membered heteroaryl group fused to a benzene ring. Unless otherwise indicated, a heteroaryl group is unsubstituted.


As used herein, the term “heterocycle” (and variations thereof such as “heterocyclic” or “heterocyclyl”) broadly refers to (i) a stable 5- to 7-membered, saturated or unsaturated monocyclic ring, or (ii) a stable 8- to 10-membered bicyclic ring system, wherein each ring in (ii) is independent of, or fused to, the other ring or rings and each ring is saturated or unsaturated, and the monocyclic ring or bicyclic ring system contains one or more heteroatoms (e.g., from 1 to 6 heteroatoms, or from 1 to 4 heteroatoms) independently selected from N, O and S and a balance of carbon atoms (the monocyclic ring typically contains at least one carbon atom and the bicyclic ring systems typically contain at least two carbon atoms); and wherein any one or more of the nitrogen and sulfur heteroatoms is optionally oxidized, and any one or more of the nitrogen heteroatoms is optionally quaternized. Unless otherwise specified, the heterocyclic ring may be attached at any heteroatom or carbon atom, provided that attachment results in the creation of a stable structure. Heterocycle groups may be substituted as indicated, and unless otherwise specified, the substituents may be attached to any atom in the ring, whether a heteroatom or a carbon atom, provided that a stable chemical structure results. Representative examples include piperidinyl, piperazinyl, azepanyl, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, oxazolidinyl, isoxazolidinyl, morpholinyl, thiomorpholinyl, thiazolidinyl, isothiazolidinyl, and tetrahydrofuryl (or tetrahydrofuranyl).


The term “heterocycloalkyl,” as used herein, refers to a non-aromatic saturated monocyclic or multicyclic ring system comprising 3 to about 11 ring atoms, wherein from 1 to 4 of the ring atoms are independently O, S, or N, and the remainder of the ring atoms are carbon atoms. A heterocycloalkyl group can be joined via a ring carbon or ring nitrogen atom. In one embodiment, a heterocycloalkyl group is monocyclic and has from about 3 to about 7 ring atoms. In another embodiment, a heterocycloalkyl group is monocyclic has from about 4 to about 7 ring atoms. In another embodiment, a heterocycloalkyl group is monocyclic has from about 5 to about 7 ring atoms. In another embodiment, a heterocycloalkyl group is bicyclic and has from about 7 to about 11 ring atoms. In still another embodiment, a heterocycloalkyl group is monocyclic and has 5 or 6 ring atoms. In one embodiment, a heterocycloalkyl group is monocyclic. In another embodiment, a heterocycloalkyl group is bicyclic. There are no adjacent oxygen and/or sulfur atoms present in the ring system. Any —NH group in a heterocycloalkyl ring may exist protected such as, for example, as an —N(BOC), —N(Cbz), —N(Tos) group and the like; such protected heterocycloalkyl groups are considered part of this invention. The term “heterocycloalkyl” also encompasses a heterocycloalkyl group, as defined above, which is fused to an aryl (e.g., benzene) or heteroaryl ring. A heterocycloalkyl group can be optionally substituted by one or more “ring system substituents” which may be the same or different, and are as defined herein below. The nitrogen or sulfur atom of the heterocycloalkyl can be optionally oxidized to the corresponding N-oxide, S-oxide or S,S-dioxide. Non-limiting examples of monocyclic heterocycloalkyl rings include oxetanyl, piperidyl, pyrrolidinyl, piperazinyl, morpholinyl, thiomorpholinyl, thiazolidinyl, 1,4-dioxanyl, tetrahydrofuranyl, tetrahydrothiophenyl, delta-lactam, delta-lactone and the like, and all isomers thereof.


A ring carbon atom of a heterocycloalkyl group may be functionalized as a carbonyl group. Illustrative example of such heterocycloalkyl groups, include:




embedded image


In one embodiment, a heterocycloalkyl group is a 5-membered monocyclic heterocycloalkyl. In another embodiment, a heterocycloalkyl group is a 6-membered monocyclic heterocycloalkyl. The term “3 to 7-membered monocyclic cycloalkyl” refers to a monocyclic heterocycloalkyl group having from 3 to 7 ring atoms. The term “4 to 7-membered monocyclic cycloalkyl” refers to a monocyclic heterocycloalkyl group having from 4 to 7 ring atoms. The term “5 to 7-membered monocyclic cycloalkyl” refers to a monocyclic heterocycloalkyl group having from 5 to 7 ring atoms. The term “7 to 11-membered bicyclic heterocycloalkyl” refers to a bicyclic heterocycloalkyl group having from 7 to 11 ring atoms. Unless otherwise indicated, an heterocycloalkyl group is unsubstituted.


The term “hydroxyalkyl,” as used herein, refers to an alkyl group as defined above, wherein one or more of the alkyl group's hydrogen atoms has been replaced with an —OH group. In one embodiment, a hydroxyalkyl group has from 1 to 6 carbon atoms. Non-limiting examples of hydroxyalkyl groups include —CH2OH, —CH2CH2OH, —CH2CH2CH2OH and —CH2CH(OH)CH3. The term “C1-C6 hydroxyalkyl” refers to a hydroxyalkyl group having from 1 to 6 carbon atoms.


As used herein, the term “oxo” or “═O” forms a carbonyl moiety with the carbon atom to which it is attached.


By “pharmaceutically acceptable” is meant that the ingredients of the pharmaceutical composition must be compatible with each other and not deleterious to the recipient thereof.


The term “preventing,” as used herein with respect to an HCV viral infection or HCV-virus related disorder, refers to reducing the likelihood of HCV infection.


The term “ring system substituent,” as used herein, refers to a substituent group attached to an aromatic or non-aromatic ring system which, for example, replaces an available hydrogen on the ring system. Ring system substituents may be the same or different, each being independently selected from the group consisting of alkyl, alkenyl, alkynyl, aryl, heteroaryl, -alkylene-aryl, -arylene-alkyl, -alkylene-heteroaryl, -alkenylene-heteroaryl, -alkynylene-heteroaryl, —OH, hydroxyalkyl, haloalkyl, —O-alkyl, —O-haloalkyl, -alkylene-O-alkyl, —O-aryl, —O— alkylene-aryl, acyl, —C(O)-aryl, halo, —NO2, —CN, —SF5, —C(O)OH, —C(O)O-alkyl, —C(O)O-aryl, —C(O)O-alkylene-aryl, —S(O)-alkyl, —S(O)2-alkyl, —S(O)-aryl, —S(O)2-aryl, —S(O)-heteroaryl, —S(O)2-heteroaryl, —S-alkyl, —S-aryl, —S-heteroaryl, —S-alkylene-aryl, —S-alkylene-heteroaryl, —S(O)2-alkylene-aryl, —S(O)2-alkylene-heteroaryl, cycloalkyl, heterocycloalkyl, —O—C(O)-alkyl, —O—C(O)-aryl, —O—C(O)-cycloalkyl, —C(═N—CN)—NH2, —C(═NH)—NH2, —C(═NH)—NH(alkyl), —N(Y1)(Y2), -alkylene-N(Y1)(Y2), —C(O)N(Y1)(Y2) and —S(O)2N(Y1)(Y2), wherein Y1 and Y2 can be the same or different and are independently selected from the group consisting of hydrogen, alkyl, aryl, cycloalkyl, and -alkylene-aryl. “Ring system substituent” may also mean a single moiety which simultaneously replaces two available hydrogens on two adjacent carbon atoms (one H on each carbon) on a ring system. Examples of such moiety are methylenedioxy, ethylenedioxy, —C(CH3)2— and the like which form moieties such as, for example:




embedded image


The term “subject” (alternatively referred to herein as “patient”), as used herein, refers to an animal, preferably a mammal, most preferably a human.


The term “substituted” means that one or more hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valency under the existing circumstances is not exceeded, and that the substitution results in a stable compound. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds. By “stable compound” or “stable structure” is meant a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture.


In the compounds of formula I, the atoms may exhibit their natural isotopic abundances, or one or more of the atoms may be artificially enriched in a particular isotope having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number predominantly found in nature. The present invention is meant to include all suitable isotopic variations of the compounds of formula I. For example, different isotopic forms of hydrogen (H) include protium (1H) and deuterium (2H or D). Protium is the predominant hydrogen isotope found in nature. Enriching for deuterium may afford certain therapeutic advantages, such as increasing in vivo half-life or reducing dosage requirements, or may provide a compound useful as a standard for characterization of biological samples. Isotopically-enriched compounds within formula I can be prepared without undue experimentation by conventional techniques well known to those skilled in the art or by processes analogous to those described in the Schemes and Examples herein using appropriate isotopically-enriched reagents and/or intermediates.


Unless expressly stated to the contrary, all ranges cited herein are inclusive. For example, a heteroaryl ring described as containing from “0 to 3 heteroatoms” means the ring can contain 0, 1, 2, or 3 heteroatoms. It is also to be understood that any range cited herein includes within its scope all of the sub-ranges within that range. The oxidized forms of the heteroatoms N and S are also included within the scope of the present invention.


When any variable (for example, R1 or R3) occurs more than one time in any constituent or in formula I or in any other formula depicting and describing compounds of the invention, its definition on each occurrence is independent of its definition at every other occurrence. Also, combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.


Unless expressly stated to the contrary, substitution by a named substituent is permitted on any atom provided such substitution is chemically allowed and results in a stable compound. A “stable” compound is a compound that can be prepared and isolated and whose structure and properties remain or can be caused to remain essentially unchanged for a period of time sufficient to allow use of the compound for the purposes described herein (e.g., therapeutic or prophylactic administration to a subject).


As a result of the selection of substituents and substituent patterns, certain of the compounds of the present invention can have asymmetric centers and can occur as mixtures of stereoisomers, or as individual diastereomers, or enantiomers. All isomeric forms of these compounds, whether isolated or in mixtures, are within the scope of the present invention.


As would be recognized by one of ordinary skill in the art, certain of the compounds of the present invention can exist as tautomers. For the purposes of the present invention a reference to a compound of formula I is a reference to the compound per se, or to any one of its tautomers per se, or to mixtures of two or more tautomers.


The compounds of the present inventions are useful in the inhibition of HCV replication (e.g., HCV NS5B activity), the treatment of HCV infection and/or reduction of the likelihood or severity of symptoms of HCV infection. For example, the compounds of this invention are useful in treating infection by HCV after suspected past exposure to HCV by such means as blood transfusion, exchange of body fluids, bites, accidental needle stick, or exposure to patient blood during surgery.


The compounds of this invention are useful in the preparation and execution of screening assays for antiviral compounds. For example, the compounds of this invention are useful for identifying resistant HCV replicon cell lines harboring mutations within NS5B, which are excellent screening tools for more powerful antiviral compounds. Furthermore, the compounds of this invention are useful in establishing or determining the binding site of other antivirals to the HCV replicase.


The compounds of the present invention may be administered in the form of pharmaceutically acceptable salts. The term “pharmaceutically acceptable salt” refers to a salt that possesses the effectiveness of the parent compound and that is not biologically or otherwise undesirable (e.g., is neither toxic nor otherwise deleterious to the recipient thereof). Suitable salts include acid addition salts that may, for example, be formed by mixing a solution of the compound of the present invention with a solution of a pharmaceutically acceptable acid such as hydrochloric acid, sulfuric acid, acetic acid, trifluoroacetic acid, or benzoic acid. Many of the compounds of the invention carry an acidic moiety, in which case suitable pharmaceutically acceptable salts thereof can include alkali metal salts (e.g., sodium or potassium salts), alkaline earth metal salts (e.g., calcium or magnesium salts), and salts formed with suitable organic ligands such as quaternary ammonium salts. Also, in the case of an acid (—COOH) or alcohol group being present, pharmaceutically acceptable esters can be employed to modify the solubility or hydrolysis characteristics of the compound.


Exemplary acid addition salts include acetates, ascorbates, benzoates, benzenesulfonates, bisulfates, borates, butyrates, citrates, camphorates, camphorsulfonates, fumarates, hydrochlorides, hydrobromides, hydroiodides, lactates, maleates, methanesulfonates (“mesylates”), naphthalenesulfonates, nitrates, oxalates, phosphates, propionates, salicylates, succinates, sulfates, tartarates, thiocyanates, toluenesulfonates (also known as tosylates) and the like. Additionally, acids which are generally considered suitable for the formation of pharmaceutically useful salts from basic pharmaceutical compounds are discussed, for example, by P. Stahl et al, Camille G. (eds.) Handbook of Pharmaceutical Salts. Properties, Selection and Use. (2002) Zurich: Wiley-VCH; S. Berge et al, Journal of Pharmaceutical Sciences (1977) 66(1) 1-19; P. Gould, International J. of Pharmaceutics (1986) 33 201-217; Anderson et al, The Practice of Medicinal Chemistry (1996), Academic Press, New York; and in The Orange Book (Food & Drug Administration, Washington, D.C. on their website). These disclosures are incorporated herein by reference thereto.


Exemplary basic salts include ammonium salts, alkali metal salts such as sodium, lithium, and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, salts with organic bases (for example, organic amines) such as dicyclohexylamine, t-butyl amine, choline, and salts with amino acids such as arginine, lysine and the like. Basic nitrogen-containing groups may be quarternized with agents such as lower alkyl halides (e.g., methyl, ethyl, and butyl chlorides, bromides and iodides), dialkyl sulfates (e.g., dimethyl, diethyl, and dibutyl sulfates), long chain halides (e.g., decyl, lauryl, and stearyl chlorides, bromides and iodides), aralkyl halides (e.g., benzyl and phenethyl bromides), and others.


For the purposes of inhibiting HCV NS5B polymerase, treating HCV infection and/or reducing the likelihood or severity of symptoms of HCV infection and inhibiting HCV viral replication and/or HCV viral production, the compounds of the present invention, optionally in the form of a salt, can be administered by any means that produces contact of the active agent with the agent's site of action. They can be administered by one or more conventional means available for use in conjunction with pharmaceuticals, either as individual therapeutic agents or in a combination of therapeutic agents. They can be administered alone, but typically are administered with a pharmaceutical carrier selected on the basis of the chosen route of administration and standard pharmaceutical practice. The compounds of the invention can, for example, be administered by one or more of the following: orally, parenterally (including subcutaneous injections, intravenous, intramuscular, intrasternal injection or infusion techniques), by inhalation (such as in a spray form), or rectally, in the form of a unit dosage of a pharmaceutical composition containing an effective amount of the compound and conventional non-toxic pharmaceutically-acceptable carriers, adjuvants and vehicles. Liquid preparations suitable for oral administration (e.g., suspensions, syrups, elixirs and the like) can be prepared according to techniques known in the art and can employ any of the usual media such as water, glycols, oils, alcohols and the like. Solid preparations suitable for oral administration (e.g., powders, pills, capsules and tablets) can be prepared according to techniques known in the art and can employ such solid excipients as starches, sugars, kaolin, lubricants, binders, disintegrating agents and the like. Parenteral compositions can be prepared according to techniques known in the art and typically employ sterile water as a carrier and optionally other ingredients, such as solubility aids. Injectable solutions can be prepared according to methods known in the art wherein the carrier comprises a saline solution, a glucose solution or a solution containing a mixture of saline and glucose. Further description of methods suitable for use in preparing pharmaceutical compositions of the present invention and of ingredients suitable for use in said compositions is provided in Remington's Pharmaceutical Sciences, 18th edition (ed. A. R. Gennaro, Mack Publishing Co., 1990).


The compounds of this invention can be administered orally in a dosage range of 0.001 to 1000 mg/kg of mammal (e.g., human) body weight per day in a single dose or in divided doses. One dosage range is 0.01 to 500 mg/kg body weight per day orally in a single dose or in divided doses. Another dosage range is 0.1 to 100 mg/kg body weight per day orally in single or divided doses. For oral administration, the compositions can be provided in the form of tablets or capsules containing 1.0 to 500 mg of the active ingredient, particularly 1, 5, 10, 15, 20, 25, 50, 75, 100, 150, 200, 250, 300, 400, and 500 mg of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated. The specific dose level and frequency of dosage for any particular patient may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, HCV viral genotype, viral resistance, and the host undergoing therapy.


As noted above, the present invention also relates to a method of inhibiting HCV NS5B activity, inhibiting HCV viral replication and/or HCV viral production, treating HCV infection and/or reducing the likelihood or severity of symptoms of HCV infection with a compound of the present invention in combination with one or more therapeutic agents and a pharmaceutical composition comprising a compound of the present invention and one or more therapeutic agents selected from the group consisting of a HCV antiviral agent, an immunomodulator, and an anti-infective agent. Such therapeutic agents active against HCV include, but are not limited to, ribavirin, levovirin, viramidine, thymosin alpha-1, R7025 (an enhanced interferon (Roche)), interferon-β, interferon-α, pegylated interferon-α (peginterferon-α), a combination of interferon-α and ribavirin, a combination of peginterferon-α and ribavirin, a combination of interferon-α and levovirin, and a combination of peginterferon-α and levovirin. Interferon-α includes, but is not limited to, recombinant interferon-α2a (such as ROFERON interferon available from Hoffmann-LaRoche, Nutley, N.J.), pegylated interferon-α2a (PEGASYS), interferon-α2b (such as INTRON-A interferon available from Schering Corp., Kenilworth, N.J.), pegylated interferon-α2b (PEGINTRON), a recombinant consensus interferon (such as interferon alphacon-1), albuferon (interferon-α bound to human serum albumin (Human Genome Sciences)), and a purified interferon-α product. Amgen's recombinant consensus interferon has the brand name INFERGEN. Levovirin is the L-enantiomer of ribavirin which has shown immunomodulatory activity similar to ribavirin. Viramidine represents an analog of ribavirin disclosed in International Patent Application Publication WO 01/60379. In accordance with the method of the present invention, the individual components of the combination can be administered separately at different times during the course of therapy or concurrently in divided or single combination forms.


For the treatment of HCV infection, the compounds of the invention may also be administered in combination with the antiviral agent NS5B polymerase inhibitor R7128 (Roche). The compounds of the present invention also may be combined for the treatment of HCV infection with antiviral 2′-C-branched ribonucleosides disclosed in Rogers E. Harry-O'Kuru et al., A Short, Flexible Route toward 2′-C-Branched Ribonucleosides, 62 J. ORG. CHEM. 1754-59 (1997); Michael S. Wolfe & Rogers E. Harry-O'Kuru, A Concise Synthesis of 2′-C-Methylribonucleosides, 36 (42) TETRAHEDRON LETTERS 7611-14 (1995); U.S. Pat. No. 3,480,613; and International Patent Application Publications WO 01/90121, WO 01/92282, WO 02/32920, WO 04/002999, WO 04/003000 and WO 04/002422; the entire contents of each of which are incorporated by reference. Such 2′-C-branched ribonucleosides include, but are not limited to, 2′-C-methyl-cytidine, 2′-C-methyl-uridine, 2′-C-methyl-adenosine, 2′-C-methyl-guanosine, and 9-(2-C-methyl-β-D-ribofuranosyl)-2,6-diaminopurine, and the corresponding amino acid ester of the ribose C-2′, C-3′, and C-5′ hydroxyls and the corresponding optionally substituted cyclic 1,3-propanediol esters of the 5′-phosphate derivatives.


For the treatment of HCV infection, the compounds of the present invention may also be administered in combination with an agent that is an inhibitor of HCV NS3 serine protease. HCV NS3 serine protease is an essential viral enzyme and has been described to be an excellent target for inhibition of HCV replication. Exemplary substrate and non-substrate based inhibitors of HCV NS3 protease inhibitors are disclosed in International Patent Application Publications WO 98/22496, WO 98/46630, WO 99/07733, WO 99/07734, WO 99/38888, WO 99/50230, WO 99/64442, WO 00/09543, WO 00/59929, WO 02/48116, WO 02/48172, WO 2008/057208 and WO 2008/057209, in British Patent No. GB 2 337 262, and in U.S. Pat. Nos. 6,323,180 and 7,470,664.


The compounds of the present invention may also be combined for the treatment of HCV infection with nucleosides having anti-HCV properties, such as those disclosed in International Patent Application Publications WO 02/51425, WO 01/79246, WO 02/32920, WO 02/48165 and WO 2005/003147 (including R1656, (2′R)-2′-deoxy-2′-fluoro-2′-C-methylcytidine, shown as compounds 3-6 on page 77); WO 01/68663; WO 99/43691; WO 02/18404 and WO 2006/021341, and U.S. Patent Application Publication US 2005/0038240, including 4′-azido nucleosides such as R1626,4′-azidocytidine; U.S. Patent Application Publications US 2002/0019363, US 2003/0236216, US 2004/0006007, US 2004/0063658 and US 2004/0110717; U.S. Pat. Nos. 7,105,499, 7,125,855, 7,202,224; and International Patent Application Publications WO 02/100415, WO 03/026589, WO 03/026675, WO 03/093290, WO 04/011478, WO 04/013300 and WO 04/028481; the content of each is incorporated herein by reference in its entirety.


For the treatment of HCV infection, the compounds of the present invention may also be administered in combination with an agent that is an inhibitor of HCV NS5B polymerase. Such HCV NS5B polymerase inhibitors that may be used as combination therapy include, but are not limited to, those disclosed in International Patent Application Publications WO 02/057287, WO 02/057425, WO 03/068244, WO 2004/000858, WO 04/003138 and WO 2004/007512; U.S. Pat. Nos. 6,777,392, 7,105,499, 7,125,855, 7,202,224 and U.S. Patent Application Publications US 2004/0067901 and US 2004/0110717; the content of each is incorporated herein by reference in its entirety. Other such HCV polymerase inhibitors include, but are not limited to, valopicitabine (NM-283; Idenix) and 2′-F-2′-beta-methylcytidine (see also WO 2005/003147).


In one embodiment, additional nucleoside HCV NS5B polymerase inhibitors that are used in combination with the present HCV NS5B inhibitors are selected from the following compounds: 4-amino-7-(2-C-methyl-β-D-arabinofuranosyl)-7H-pyrrolo[2,3-d]pyrimidine; 4-amino-7-(2-C-methyl-β-D-ribofuranosyl)-7H-pyrrolo[2,3-d]pyrimidine; 4-methylamino-7-(2-C-methyl-β-D-ribofuranosyl)-7H-pyrrolo[2,3-d]pyrimidine; 4-dimethylamino-7-(2-C-methyl-β-D-ribofuranosyl)-7H-pyrrolo[2,3-d]pyrimidine; 4-cyclopropylamino-7-(2-C-methyl-β-D-ribofuranosyl)-7H-pyrrolo[2,3-d]pyrimidine; 4-amino-7-(2-C-vinyl-β-D-ribofuranosyl)-7H-pyrrolo[2,3-d]pyrimidine; 4-amino-7-(2-C-hydroxymethyl-β-D-ribofuranosyl)-7H-pyrrolo[2,3-d]pyrimidine; 4-amino-7-(2-C-fluoromethyl-β-D-ribofuranosyl)-7H-pyrrolo[2,3-d]pyrimidine; 4-amino-5-methyl-7-(2-C-methyl-β-D-ribofuranosyl)-7H-pyrrolo[2,3-d]pyrimidine; 4-amino-7-(2-C-methyl-β-D-ribofuranosyl)-7H-pyrrolo[2,3-d]pyrimidine-5-carboxylic acid; 4-amino-5-bromo-7-(2-C-methyl-β-D-ribofuranosyl)-7H-pyrrolo[2,3-d]pyrimidine; 4-amino-5-chloro-7-(2-C-methyl-β-D-ribofuranosyl)-7H-pyrrolo[2,3-d]pyrimidine; 4-amino-5-fluoro-7-(2-C-methyl-β-D-ribofuranosyl)-7H-pyrrolo[2,3-d]pyrimidine; 2,4-diamino-7-(2-C-methyl-β-D-ribofuranosyl)-7H-pyrrolo[2,3-d]pyrimidine; 2-amino-7-(2-C-methyl-β-D-ribofuranosyl)-7H-pyrrolo[2,3-d]pyrimidine; 2-amino-4-cyclopropylamino-7-(2-C-methyl-β-D-ribofuranosyl)-7H-pyrrolo[2,3-d]pyrimidine; 2-amino-7-(2-C-methyl-β-D-ribofuranosyl)-7H-pyrrolo[2,3-d]pyrimidin-4 (3H)-one; 4-amino-7-(2-C-ethyl-β-D-ribofuranosyl)-7H-pyrrolo[2,3-d]pyrimidine; 4-amino-7-(2-β-O-dimethyl-β-D-ribofuranosyl)-7H-pyrrolo[2,3-d]pyrimidine; 7-(2-C-methyl-β-D-ribofuranosyl)-7H-pyrrolo[2,3-d]pyrimidin-4 (3H)-one; 2-amino-5-methyl-7-(2-C, 2-O-dimethyl-β-D-ribofuranosyl)-7H-pyrrolo[2,3-d]pyrimidin-4 (3H)-one; 4-amino-7-(3-deoxy-2-C-methyl-β-D-ribofuranosyl)-7H-pyrrolo[2,3-d]pyrimidine; 4-amino-7-(3-deoxy-2-C-methyl-β-D-arabinofuranosyl)-7H-pyrrolo[2,3-d]pyrimidine; 4-amino-2-fluoro-7-(2-C-methyl-β-D-ribofuranosyl)-7H-pyrrolo[2,3-d]pyrimidine; 4-amino-7-(3-C-methyl-(3-D-ribofuranosyl)-7H-pyrrolo[2,3-d]pyrimidine; 4-amino-7-(3-C-methyl-β-D-xylofuranosyl)-7H-pyrrolo[2,3-d]pyrimidine; 4-amino-7-(2,4-di-C-methyl-β-D-ribofuranosyl)-7H-pyrrolo[2,3-d]pyrimidine; 4-amino-7-(3-deoxy-3-fluoro-2-C-methyl-β-D-ribofuranosyl)-7H-pyrrolo[2,3-d]pyrimidine; and the corresponding 5′-triphosphates; or a pharmaceutically acceptable salt thereof


The compounds of the present invention may also be combined for the treatment of HCV infection with non-nucleoside inhibitors of HCV polymerase such as those disclosed in U.S. Patent Application Publications US 2006/0100262 and US 2009/0048239; International Patent Application Publications WO 01/77091, WO 01/47883, WO 02/04425, WO 02/06246, WO 02/20497, WO 2005/016927 (in particular JTK003), WO 2004/041201, WO 2006/066079, WO 2006/066080, WO 2008/075103, WO 2009/010783 and WO 2009/010785; the content of each is incorporated herein by reference in its entirety.


In one embodiment, additional non-nucleoside HCV NS5B polymerase inhibitors that are used in combination with the present HCV NS5B inhibitors are selected from the following compounds: 14-cyclohexyl-6-[2-(dimethylamino)ethyl]-7-oxo-5,6,7,8-tetrahydroindolo[2,1-a][2,5]benzodiazocine-11-carboxylic acid; 14-cyclohexyl-6-(2-morpholin-4-ylethyl)-5,6,7,8-tetrahydroindolo[2,1-a][2,5]benzodiazocine-11-carboxylic acid; 14-cyclohexyl-6-[2-(dimethylamino)ethyl]-3-methoxy-5,6,7,8-tetrahydroindolo[2,1-a][2,5]benzodiazocine-11-carboxylic acid; 14-cyclohexyl-3-methoxy-6-methyl-5,6,7,8-tetrahydroindolo[2,1-a][2,5]benzodiazocine-11-carboxylic acid; methyl ({[(14-cyclohexyl-3-methoxy-6-methyl-5,6,7,8-tetrahydroindolo[2,1-a][2,5]benzodiazocin-11-yl)carbonyl]amino}sulfonyl)acetate; ({[(14-cyclohexyl-3-methoxy-6-methyl-5,6,7,8-tetrahydroindolo[2,1-a][2,5]benzodiazocin-11-yl)carbonyl]amino}sulfonyl)acetic acid; 14-cyclohexyl-N-[(dimethylamino)sulfonyl]-3-methoxy-6-methyl-5,6,7,8-tetrahydroindolo[2,1-a][2,5]benzodiazocine-11-carboxamide; 3-chloro-14-cyclohexyl-6-[2-(dimethylamino)ethyl]-7-oxo-5,6,7,8-tetrahydroindolo[2,1-a][2,5]benzodiazocine 11-carboxylic acid; N′-(11-carboxy-14-cyclohexyl-7,8-dihydro-6H-indolo[1,2-e][1,5]benzoxazocin-7-yl)-N,N-dimethylethane-1,2-diaminium bis(trifluoroacetate); 14-cyclohexyl-7,8-dihydro-6H-indolo[1,2-e][1,5]benzoxazocine-11-carboxylic acid; 14-cyclohexyl-6-methyl-7-oxo-5,6,7,8-tetrahydroindolo[2,1-a][2,5]benzodiazocine-11-carboxylic acid; 14-cyclohexyl-3-methoxy-6-methyl-7-oxo-5,6,7,8-tetrahydroindolo[2,1-a][2,5]benzodiazocine-11-carboxylic acid; 14-cyclohexyl-6-[2-(dimethylamino)ethyl]-3-methoxy-7-oxo-5,6,7,8-tetrahydroindolo[2,1-a][2,5]benzodiazocine-11-carboxylic acid; 14-cyclohexyl-6-[3-(dimethylamino)propyl]-7-oxo-5,6,7,8-tetrahydroindolo[2,1-a][2,5]benzodiazocine-11-carboxylic acid; 14-cyclohexyl-7-oxo-6-(2-piperidin-1-ylethyl)-5,6,7,8-tetrahydroindolo[2,1-a][2,5]benzodiazocine-11-carboxylic acid; 14-cyclohexyl-6-(2-morpholin-4-ylethyl)-7-oxo-5,6,7,8-tetrahydroindolo[2,1-a][2,5]benzodiazocine-11-carboxylic acid; 14-cyclohexyl-6-[2-(diethylamino)ethyl]-7-oxo-5,6,7,8-tetrahydroindolo[2,1-a][2,5]benzodiazocine-11-carboxylic acid; 14-cyclohexyl-6-(1-methylpiperidin-4-yl)-7-oxo-5,6,7,8-tetrahydroindolo[2,1-a][2,5]benzodiazocine-11-carboxylic acid; 14-cyclohexyl-N-[(dimethylamino)sulfonyl]-7-oxo-6-(2-piperidin-1-ylethyl)-5,6,7,8-tetrahydroindolo[2,1-a][2,5]benzo diazocine-11-carboxamide; 14-cyclohexyl-6-[2-(dimethylamino)ethyl]-N-[(dimethylamino)sulfonyl]-7-oxo-5,6,7,8-tetrahydroindolo[2,1-a][2,5]benzodiazocine-11-carboxamide; 14-cyclopentyl-6-[2-(dimethylamino)ethyl]-7-oxo-5,6,7,8-tetrahydroindolo[2,1-a][2,5]benzodiazocine-11-carboxylic acid; 14-cyclohexyl-5,6,7,8-tetrahydroindolo[2,1-a][2,5]benzodiazocine-11-carboxylic acid; 6-allyl-14-cyclohexyl-3-methoxy-5,6,7,8-tetrahydroindolo[2,1-a][2,5]benzodiazocine-11-carboxylic acid; 14-cyclopentyl-6-[2-(dimethylamino)ethyl]-5,6,7,8-tetrahydroindolo[2,1-a][2,5]benzodiazocine-11-carboxylic acid; 14-cyclohexyl-6-[2-(dimethylamino)ethyl]-5,6,7,8-tetrahydroindolo[2,1-a][2,5]benzodiazocine-11-carboxylic acid; 13-cyclohexyl-5-methyl-4,5,6,7-tetrahydrofuro[3′,2′:6,7][1,4]diazocino[1,8-a]indole-10-carboxylic acid; 15-cyclohexyl-6-[2-(dimethylamino)ethyl]-7-oxo-6,7,8,9-tetrahydro-5H-indolo[2,1-a][2,6]benzodiazonine-12-carboxylic acid; 15-cyclohexyl-8-oxo-6,7,8,9-tetrahydro-5H-indolo[2,1-a][2,5]benzodiazonine-12-carboxylic acid; 13-cyclohexyl-6-oxo-6,7-dihydro-5H-indolo[1,2-d][1,4]benzodiazepine-10-carboxylic acid; and pharmaceutically acceptable salts thereof


In another embodiment, the present HCV NS5B polymerase inhibitors are used in combination with non-nucleoside HCV NS5A inhibitors and pharmaceutically acceptable salts thereof


The HCV NS5B inhibitory activity of the present compounds may be tested using assays known in the art. The HCV NS5B polymerase inhibitors described herein have activities in a genotype 1b replicon assay as described in the Examples. The assay is performed by incubating a replicon harboring cell-line in the presence of inhibitor for a set period of time and measuring the effect of the inhibitor on HCV replicon replication either directly by quantifying replicon RNA level, or indirectly by measuring enzymatic activity of a co-encoded reporter enzyme such as luciferase or β-lactamase. By performing a series of such measurements at different inhibitor concentrations, the effective inhibitory concentration of the inhibitor (EC50 or EC90) is determined See Jan M. Vrolijk et al., A replicons-based bioassay for the measurement of interferons in patients with chronic hepatitis C, 110 J. VIROLOGICAL METHODS 201 (2003). Such assays may also be run in an automated format for high through-put screening. See Paul Zuck et al., A cell-based β-lactamase reporter gene assay for the identification of inhibitors of hepatitis C virus replication, 334 ANALYTICAL BIOCHEMISTRY 344 (2004).


The present invention also includes processes for making compounds of formula I. The Compounds of Formula (I) may be prepared from known or readily prepared starting materials, following methods known to one skilled in the art of organic synthesis. Methods useful for making the Compounds of Formula (I) are set forth in the Examples below and generalized in Schemes 1-6 below. Alternative synthetic pathways and analogous structures will be apparent to those skilled in the art of organic synthesis. All stereoisomers and tautomeric forms of the compounds are contemplated.


Some commercially available starting materials and intermediates used for the synthesis of the Compounds of Formula (I) are available. These starting materials and intermediates are available from commercial suppliers such as Sigma-Aldrich (St. Louis, Mo.) and Acros Organics Co. (Fair Lawn, N.J.). Such starting materials and intermediates compounds are used as received.


Scheme 1 shows methods useful for making the intermediates of formula A and B, which can be converted to intermediate C.




embedded image


embedded image


Scheme 2 shows a method useful for converting intermediate of formula A or B to intermediate C, which can be converted to Compounds of Formula (I).




embedded image


embedded image


Scheme 3 shows methods useful for making the intermediates of formula D, which can be converted to compounds of Formula (I).




embedded image


Scheme 4 shows a method useful for converting intermediate of formula D to compounds of formula (Ia).




embedded image


Scheme 5 shows an alternative route to compounds of formula (Ia), when R6 is H.




embedded image


Scheme 6 shows an alternative route to compounds of formula (Ia), when R6 is H, n is 0, 1, 2 or 3.




embedded image


LIST OF ABBREVIATIONS



  • AIBN 2,2′-azoisobutyronitrile

  • a.q., aq Aqueous

  • Al2O3 Aluminum oxide

  • Ar2 Argon

  • ATM Atmosphere

  • BH3-DMS Borane methyl sulfide

  • Boc t-butyloxycarbonyl

  • Boc2O Di-tert-butyl-dicarbonate

  • n-Bu n-butyl

  • n-BuLi n-butyllithium

  • Bu3SnH Tributylin hydride

  • Bu4NCl Tetrabutylammonium chloride

  • conc. concentrated

  • CDCl3 Trichloro(2H)methane or deuterio-trichloromethane

  • CHCl3 Chloroform

  • CH3SNa Sodium thiomethoxide

  • CO Carbon monoxide

  • Cs2CO3 Cesium carbonate

  • DAST Diethylaminosulfur trifluoride

  • DCM, CH2Cl2 Dichloromethane

  • DEAD Diethyl azodicarboxylate

  • DIAD Diisopropyl azodicarboxylate

  • DIBAL-H Diisobutylaluminium hydride

  • DIPEA, DIEA N,N-Diisopropylethylamine

  • DMAc Dimethylacetamide

  • DMAP 4-dimethylaminopyridine

  • DMF Dimethylformamide

  • DMSO Dimethylsulfoxide

  • EDCI N-(3-Dimethylaminopropyl)-N′-ethylcarbodiimide (also EDC)

  • Et Ethyl

  • Et3N Triethylamine

  • EtOAc, EA Ethyl acetate

  • EtOH Ethanol

  • Fe Iron

  • HATU (Dimethylamino)-N,N-dimethyl(3H-[1,2,3]triazolo[4,5-b]pyridin-3-yloxy)methaniminium hexafluorophosphate

  • HBF4 Fluoroboric acid

  • HCl Hydrochloric acid

  • H2 Hydrogen gas or atmosphere

  • HNO3 Nitric acid

  • H2O Water

  • H2O2 Hydrogen peroxide

  • H2SO4 Sulfuric acid

  • HOBT 1-Hydroxy benzotriazole


  • 1H-NMR Proton Nuclear Magnetic Resonance

  • HPLC High Performance Liquid Chromatography

  • IPA Isopropyl alcohol

  • KCN Potassium cyanide

  • K2CO3 Potassium carbonate

  • K3CO3 Potassium bicarbonate

  • KOAc Potassium acetate

  • KOH Potassium hydroxide

  • K3PO4 Potassium Phosphate

  • LDA Lithium diisopropylamide

  • LiCl Lithium chloride

  • LiOH Lithium hydroxide

  • MeNH2, CH3NH2 Methylamine

  • MeCN, CH3CN Acetonitrile

  • Met, CH3I Methyl iodide

  • MeOD Methan(2H)ol

  • MeOH, CH3OH Methanol

  • MS Mass spectroscopy

  • Ms Methanesulfonyl (or mesyl) group

  • MsCl Methanesulfonyl chloride

  • N2 Nitrogen gas or atmosphere

  • NaBH4 Sodium borohydride

  • NaBO3 Sodium perborate

  • NaCN Sodium cyanide

  • Na2CO3 Sodium carbonate

  • NaHCO3 Sodium bicarbonate

  • NaIO4 Sodium periodate

  • Na(OAc)3BH Sodium triactetoxy borohydride

  • Na2SO4 Sodium sulfate (anhydrous)

  • Na2S2O3 Sodium thiosulfate

  • NaH Sodium hydride

  • NCS N-chlorosuccinimide

  • NH4Cl Ammonium chloride

  • NH2NH2.H2SO4 Hydrazine sulfate

  • NIS N-iodosuccinimide

  • NMO N-methylmorpholine-N-oxide

  • OPPh3 Triphenyl phosphine oxide

  • OsO4 Osmium tetroxide

  • Pd Palladium

  • Pd/C Palladium on carbon

  • PdCl2 Palladium(II) chloride

  • Pd2(dba)3 Tris(dibenzylideneacetone)dipalladium(0)

  • Pd(dppf)Cl2 1,1′-bis(diphenylphosphino)ferrocene-palladium(II)dichloride

  • Pd(dtbpf)Cl2, DTBPF 1,1′-bis(di-tert-butylphosphino)ferrocene-dichloropalladium(II)

  • PdCl2 Palladium(II)chloride

  • Pd(OAc)2 Palladium(II)acetate

  • Pd(PPh3)2Cl2 1,1′-bis(tetrakis(triphenylphosphine))palladium(II)dichloride

  • PE Petroleum ether

  • Ph Phenyl

  • PPh3 Triphenylphosphine

  • POCl3 Phosphoryl chloride

  • Py Pyridine

  • RT Room temperature, approximately 25° C.

  • sat saturated

  • SFC Supercritical fluid chromatography

  • TBAc Tert-butyl acetate

  • TEA Triethylamine

  • TFA Trifluoroacetic acid

  • THF Tetrahydrofuran

  • TLC Thin layer chromatography

  • TMSCl Trimethylsilyl chloride



EXAMPLES
Example 1
5-(11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-N-methyl-6-(N-methylmethylsulfonamido)-2-(4-(trifluoromethyl)phenyl)benzofuran-3-carboxamide



embedded image


Step 1—Synthesis of ethyl 5-bromobenzofuran-3-carboxylate



embedded image


HBF4.Et2O (16.2 g, 99.5 mmol) was added to a solution of 5-bromo-2-hydroxybenzaldehyde (200 g, 995 mmol) in CH2Cl2 (500 mL), and then a solution of ethyl diazoacetate (180 g, 1.42 mol) in CH2Cl2 (500 mL) was introduced as evolution of N2 gas while the reaction was not allowed over 38° C. Once gas evolution ceased, the reaction mixture was concentrated by rotary evaporator and conc. H2SO4 (129 g, 1.29 mol, 98%) was added to the mixture while stirring. After 20 minutes, the acidic mixture was neutralized with Na2CO3 (a.q). After the mixture was stored and crystallized overnight, ethyl 5-bromobenzofuran-3-carboxylate (100 g, yield: 75%) was obtained by filtration. 1H-NMR (CDCl3, 400 MHz) δ 8.25 (s, 1H), 8.15˜8.21 (m, 1H), 7.44˜7.50 (m, 1H), 7.37˜7.42 (m, 1H), 4.41 (q, J=7.2 Hz, 2H), 1.43 (t, J=7.2 Hz, 3H). MS (M+H)+: 269/271.


Step 2—Synthesis of ethyl 5-bromo-6-nitrobenzofuran-3-carboxylate



embedded image


To a solution of ethyl 5-bromobenzofuran-3-carboxylate (95 g, 353 mmol) in CHCl3 (1000 mL), fuming HNO3 (192 mL, 95%) was added dropwise at −20° C. over 90 min and stirred at 0° C. for 1 hour. The reaction mixture was added to ice water and extracted with CH2Cl2. The organic layer was washed with NaHCO3 and brine. The solvent was removed by distillation to provide the crude product of ethyl 5-bromo-6-nitrobenzofuran-3-carboxylate (95 g, yield: 85%). It was used for the next step without further purification.


Step 3—Synthesis of ethyl 6-amino-5-bromobenzofuran-3-carboxylate



embedded image


A mixture of crude ethyl 5-bromo-6-nitrobenzofuran-3-carboxylate (95 g, 302 mmol), iron filings (50.67 g, 907 mmol) and NH4Cl (97 g, 1.82 mol) in MeOH-THF—H2O (2:2:1, 1000 mL) were stirred at refluxing for 3 hours. After filtered and concentrated in vacuum, the residue was purified by column chromatography (eluted with PE:EA from 20:1 to 10:1) to furnish the pure product of ethyl 6-amino-5-bromobenzofuran-3-carboxylate (58.0 g, yield: 68%). 1H-NMR (DMSO-d6, 400 MHz) δ 8.46 (s, 1H), 7.85 (s, 1H), 7.03 (s, 1H), 5.55 (br s, 2H), 4.31 (q, J=7.2 Hz, 2H), 1.32 (t, J=7.2 Hz, 3H). MS (M+H)+: 284/286.


Step 4—Synthesis of 6-amino-5-bromobenzofuran-3-carboxylic acid



embedded image


To a solution of ethyl 6-amino-5-bromobenzofuran-3-carboxylate (58 g, 204 mmol) in 1,4-dioxane and H2O (850 mL and 150 mL) was add LiOH.H2O (42.8 g, 1.02 mol). The reaction mixture was refluxed for 2 hours, and then 400 mL H2O was added to the reaction mixture. After acidifying to pH 4˜5 with HCl, the resulting solid was filtered to give the product of 6-amino-5-bromobenzofuran-3-carboxylic acid (51 g, yield: 97%). 1H-NMR (DMSO-d6, 400 MHz) δ 8.36 (s, 1H), 7.87 (s, 1H), 7.02 (s, 1H), 5.51 (br s, 2H). MS (M+H)+: 256/258.


Step 5—Synthesis of 6-amino-5-bromo-N-methylbenzofuran-3-carboxamide



embedded image


To a solution of 6-amino-5-bromobenzofuran-3-carboxylic acid (51 g, 199 mmol) in dry DMF (500 mL) was added EDCI (53.1 g, 298.77 mmol) and HOBT (40.4 g, 299 mmol). The reaction mixture was stirred at room temperature for 2 h, and then Et3N (60.5 g, 598 mmol) and MeNH2.HCl (40.3 g, 598 mmol) were added to the reaction mixture. After stirring for another 2 hours, the reaction mixture was concentrated in vacuum and then 300 mL Na2CO3 (sat., a.q.) was added to the mixture. The resulting solid was filtered to give the crude product, which was purified by column chromatography (DCM:MeOH=30:1) to give the product 6-amino-5-bromo-N-methylbenzofuran-3-carboxamide (38 g, yield: 71%). 1H-NMR (DMSO-d6, 400 MHz) δ 8.19˜8.21 (m, 2H), 7.98 (s, 1H), 6.97 (s, 1H), 5.46 (br s, 2H), 2.75 (d, J=4.4 Hz, 3H). MS (M+H)+: 269/271.


Step 6—Synthesis of (6-amino-5-bromo-3-(methylcarbamoyl)benzofuran-2-yl)boronic acid



embedded image


To a solution of LDA in THF (62.5 mmol, 70 mL, freshly prepared), 6-amino-5-bromo-N-methylbenzofuran-3-carboxamide (4 g, 14.86 mmol) in THF (60 mL) was added dropwise at −78° C. under N2. After the mixture was stirred for 1 hour, trimethyl borate (6.18 g, 59.5 mmol) was added dropwise at −78° C. After the mixture was stirred for 1 hour, NH4Cl (a.q.) was added, and the mixture was extracted with EtOAc (100 mL×3), dried over Na2SO4, filtrated and concentrated to give (6-amino-5-bromo-3-(methylcarbamoyl)benzofuran-2-yl)boronic acid (3.3 g, yield: 70%). 1H-NMR (DMSO-d6, 400 MHz) δ 9.56 (s, 2H), 8.46 (d, J=4.8 Hz, 1H), 8.00 (s, 1H), 6.95 (s, 1H), 5.20˜5.82 (br s, 2H), 2.87 (d, J=4.8 Hz, 3H). MS (M+H)+: 313/315.


Step 7—Synthesis of 6-amino-5-bromo-2-iodo-N-methylbenzofuran-3-carboxamide



embedded image


To a solution of (6-amino-5-bromo-3-(methylcarbamoyl)benzofuran-2-yl)boronic acid (2 g, 6.4 mmol) in MeCN (20 mL) was added NIS (1.44 g, 6.4 mmol) at 0° C., and then the mixture was stirred at 25° C. overnight. After concentrated in vacuum, the residue was purified by column chromatography (DCM:EtOAc=10:1) to give to give pure 6-amino-5-bromo-2-iodo-N-methylbenzofuran-3-carboxamide (2 g, yield: 80%). 1H-NMR (CDCl3, 400 MHz) δ 7.84 (s, 1H), 6.80 (s, 1H), 6.45 (s, 1H), 2.92 (s, 3H). MS (M+H)+: 395/397.


Step 8—Synthesis of 5-bromo-2-iodo-N-methyl-6-(methylsulfonamido)benzofuran-3-carboxamide



embedded image


To a solution of 6-amino-5-bromo-2-iodo-N-methylbenzofuran-3-carboxamide (1 g, 2.53 mmol) in pyridine, MsCl (580 mg, 5.06 mmol) was added dropwise at 0° C. The mixture was allowed to warm to room temperature and stirred for 1.5 h. After the solvent was removed in vacuo, the reaction mixture was adjusted to pH=5-6 with 1 N HCl aq. After filtration, the solid was dissolved in THF:H2O=5:1 (15 mL) and then LiOH.H2O (800 mg, 20 mmol) was added. The mixture was stirred for 30 minutes at room temperature. After the solvent was removed in vacuo, the reaction mixture was adjusted to pH=5-6 with 1 N HCl aq. Finally the precipitate was collected to give 5-bromo-2-iodo-N-methyl-6-(methylsulfonamido)benzofuran-3-carboxamide (800 mg, 90% purity by HPLC, yield: 60%), which was used for the next step without further purification. 1H-NMR (CDCl3, 400 MHz) δ 8.37 (s, 1H), 7.84 (s, 1H), 6.86 (s, 1H), 6.29 (s, 1H), 3.07 (d, J=4.8 Hz, 3H), 2.99 (s, 3H). MS (M+H)+: 473/475.


Step 9—Synthesis of 5-bromo-2-iodo-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a suspension of 5-bromo-2-iodo-N-methyl-6-(methylsulfonamido)benzofuran-3-carboxamide (177 mg, 90% purity, 0.34 mmol) and K2CO3 (140 mg, 1.01 mmol) in DMF (3 mL) was added dropwise CH3I (79 mg, 0.68 mol) at 0° C. under N2, and then the mixture was stirred at 80° C. for 1 hour. After concentrated in vacuum, the residue was suspended in H2O and extracted with DCM. The residue was purified by column chromatography (eluted with DCM: EtOAc from 10:1 to 2:1) to give 5-bromo-2-iodo-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (150 mg, yield: 90%). 1H-NMR (CDCl3, 400 MHz) δ 8.38 (s, 1H), 7.66 (s, 1H), 6.27 (s, 1H), 3.32 (s, 3H), 3.08 (d, J=4.8 Hz, 3H), 2.89 (s, 3H). MS (M+H)+: 487/489.


Step 10—Synthesis of 5-bromo-N-methyl-6-(N-methylmethylsulfonamido)-2-(6-(trifluoromethyl)pyridin-3-yl)benzofuran-3-carboxamide



embedded image


A mixture of 5-bromo-2-iodo-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (1.5 g, 3.1 mmol), (6-(trifluoromethyl)pyridin-3-yl)boronic acid (705 mg, 3.7 mmol), Pd(dppf)Cl2 (245 mg, 0.3 mmol) and Na2CO3 (985 mg, 9.3 mmol) in dioxane/H2O (20 mL/4 mL) was stirred at 80° C. for 6 h. Water was added and the mixture was extracted with EtOAc. The organic phase was dried over Na2SO4 and concentrated in vacuum. The residue was purified by column chromatography (PE:EA=1:1) to give the product of 5-bromo-N-methyl-6-(N-methylmethylsulfonamido)-2-(6-(trifluoromethyl)pyridin-3-yl)benzofuran-3-carboxamide (1.4 g, yield: 90%). 1H-NMR (DMSO-d6, 400 MHz) δ 9.24 (s, 1H), 8.65 (d, J=4.4 Hz, 1H), 8.56 (d, J=8.0 Hz, 1H), 8.17 (s, 1H), 8.12 (d, J=8.0 Hz, 1H), 8.08 (s, 1H), 3.23 (s, 3H), 3.22 (s, 3H), 2.85 (d, J=4.4 Hz, 3H). MS (M+H)+: 506/508.


Step 11—Synthesis of N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2-(6-(trifluoromethyl)pyridin-3-yl)benzofuran-3-carboxamide



embedded image


A mixture of 5-bromo-N-methyl-6-(N-methylmethylsulfonamido)-2-(6-(trifluoromethyl)pyridin-3-yl)benzofuran-3-carboxamide (1.4 g, 2.7 mmol), bis(pinacolato)diboron (3.5 g, 13.8 mmol), Pd(dppf)Cl2 (226 mg, 0.27 mmol) and KOAc (814 mg, 8.3 mmol) in dioxane/H2O (10 mL/1 mL) was stirred at 120° C. for 5 h. Water was added and the mixture was extracted with EtOAc. The organic phase was dried over Na2SO4 and concentrated in vacuum. The residue was purified by column chromatography (PE:EA=1:1) to give the product of N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2-(6-(trifluoromethyl)pyridin-3-yl)benzofuran-3-carboxamide (1.3 g, yield: 87%). 1H-NMR (MeOD, 400 MHz) δ 9.22 (s, 1H), 8.54 (d, J=8.4 Hz, 1H), 8.11 (s, 1H), 7.97 (d, J=8.4 Hz, 1H), 7.79 (s, 1H), 3.38 (s, 3H), 3.01 (s, 3H), 3.00 (s, 3H), 1.39 (s, 12H). MS (M+H)+: 554.


Step 12—Synthesis of tert-butyl 4-fluoro-1H-indole-1-carboxylate



embedded image


To a solution of 4-fluoro-1H-indole (150 g, 1.11 mol) and DMAP (4.5 g, 3% Wt) in THF (2.5 L) was added (Boc)2O (255 g, 1.16 mol) dropwise. The mixture was stirred at room temperature overnight. The organic solvent was removed in vacuum, and the residue was purified by column chromatography (PE) to give tert-butyl 4-fluoro-1H-indole-1-carboxylate (250 g, yield: 96%). 1H-NMR (CDCl3, 400 MHz) δ 7.92 (d, J=8.4 Hz, 1H), 7.55 (d, J=3.6 Hz, 1H), 7.23 (m, 1H), 6.90 (m, 1H), 6.66 (d, J=3.6 Hz, 1H), 1.67 (s, 9H). MS (M+H)+: 236.


Step 13—Synthesis of (1-(tert-butoxycarbonyl)-4-fluoro-1H-indol-2-yl)boronic acid



embedded image


To a solution of diisopropylamine (175 mL, 1.25 mol) in THF (800 mL) at 0° C. was added n-BuLi (500 mL, 1.25 mol, 2.5 M in hexane) dropwise. The mixture was stirred at 0° C. for 40 min. Then the mixture was cooled to −78° C. Tert-butyl 4-fluoro-1H-indole-1-carboxylate (118 g, 0.50 mol) in THF (300 mL) was added dropwise slowly, followed by triisopropyl borate (231 mL, 1.00 mol). The mixture was stirred at −78° C. for another 40 min. The reaction was monitored by HPLC. When the reaction was completed, the reaction was quenched with NH4Cl (sat. 500 mL). Then the mixture was adjusted to pH=6 with 1 N HCl. Extracted with EtOAc (2000 mL) and the combined organic layers were washed with brine (500 mL), dried over Na2SO4, filtered and concentrated. The obtained solid was recrystallized with EtOAc and PE to give (1-(tert-butoxycarbonyl)-4-fluoro-1H-indol-2-yl)boronic acid (93 g, yield: 64%, store in fridge). 1H-NMR (CDCl3, 400 MHz) δ 7.77 (d, J=8.4 Hz, 1H), 7.57 (s, 1H), 7.44 (s, 2H), 7.24 (m, 1H), 6.90 (m, 1H), 1.66 (s, 9H). MS (M+H)+: 280.


Step 14—Synthesis of 6-chloro-2-iodopyridin-3-ol



embedded image


6-chloropyridin-3-ol (5.0 g, 38.60 mmol) was dissolved in water (50 mL) and placed under an N2 atmosphere. Na2CO3 (8.2 g, 77.37 mmol) was added followed by iodine (9.8 g, 38.81 mmol). The reaction mixture was stirred at room temperature for 2 h. The mixture was poured into 1 M Na2S2O3 and extracted with EtOAc. The combined organic phases were washed with brine, dried over Na2SO4 and concentrated to give the product of 6-chloro-2-iodopyridin-3-ol (7.0 g, yield: 70.9%). 1H-NMR (CDCl3, 400 MHz) δ 7.17 (d, J=8.4 Hz, 1H), 7.06 (d, J=8.4 Hz, 1H). MS (M+H)+: 256/258.


Step 15—Synthesis of 6-chloro-2-(4-fluoro-1H-indol-2-yl)pyridin-3-ol



embedded image


To a solution of (1-(tert-butoxycarbonyl)-4-fluoro-1H-indol-2-yl)boronic acid (126 g, 0.45 mol) and 6-chloro-2-iodopyridin-3-ol (96 g, 0.37 mmol) in 1,4-dioxane (1.8 L) and water (0.2 L) were added Pd(PPh3)2Cl2 (13.2 g, 18.6 mmol) and NaHCO3 (94.8 g, 1.13 mol) under nitrogen atmosphere, and the mixture was heated at 90° C. under N2 for 16 h. The reaction mixture was cooled to room temperature, diluted with EtOAc (900 mL), filtered and concentrated. The residue was diluted with H2O (400 mL) and EtOAc (800 mL), and the layer was separated, the aqueous layer was extracted with EtOAc (3*400 mL). The combined organic layers were washed with brine (800 mL), dried over Na2SO4, filtered and concentrated. The residue was purified by column chromatography (PE:EtOAc=20:1˜3:1) to give 6-chloro-2-(4-fluoro-1H-indol-2-yl)pyridin-3-ol (70 g, yield: 70.1%). 1H-NMR (MeOD, 400 MHz) δ 7.36 (s, 1H), 7.23˜7.27 (m, 2H), 7.03˜7.11 (m, 2H), 6.63˜6.68 (m, 1H). MS (M+H)+: 263 (M+H).


Step 16—Synthesis of 6-chloro-2-(4-fluoro-1H-indol-2-yl)pyridin-3-yl trifluoromethanesulfonate



embedded image


DIPEA (1.96 g, 15.2 mmol) and DMAP (18 mg, 0.15 mmol) were added to a suspension of 6-chloro-2-(4-fluoro-1H-indol-2-yl)pyridin-3-ol (2 g, 7.6 mmol) in anhydrous


DCM (20 mL) under N2. The reaction mixture was cooled to 0° C., and then trifluoromethanesulfonic anhydride (4.28 g, 15.2 mmol) was added dropwise. After stirred at RT for 1 h, the mixture was diluted with water and extracted with CH2Cl2. The organic layers were washed with brine, dried over Na2SO4, and concentrated. The mixture was purified by column chromatography (PE:EA=200:1) to give 6-chloro-2-(4-fluoro-1H-indol-2-yl)pyridin-3-yl trifluoromethanesulfonate (2.5 g, yield: 83%). 1H-NMR (DMSO-d6, 400 MHz) δ 12.12 (br s, 1H), 8.14 (d, J=8.6 Hz, 1H), 7.64 (d, J=8.8 Hz, 1H), 7.36 (d, J=8.2 Hz, 1H), 7.24˜7.11 (m, 2H), 6.83 (dd, J=8.0, 10.4 Hz, 1H). MS (M+H)+: 395.


Step 17—Synthesis of 6-chloro-2-(4-fluoro-1H-indol-2-yl)pyridin-3-yl trifluoromethanesulfonate



embedded image


To a mixture of 6-chloro-2-(4-fluoro-1H-indol-2-yl)pyridin-3-yl trifluoromethanesulfonate (2.50 g, 6.3 mmol), tributyl(vinyl)stannane (2.41 g, 7.6 mmol) and anhydrous LiCl (805 mg, 19.0 mmol) in DMF (50 mL), Pd(PPh3)2Cl2 (222 mg, 0.3 mmol) was added under N2 protection. The reaction mixture was stirred at 60° C. for 1 h. Then it was concentrated in vacuo, the residue was purified by column chromatography (PE:EA=400:1-100:1) to give the product of 2-(6-chloro-3-vinylpyridin-2-yl)-4-fluoro-1H-indole (1.20 g, yield: 69%). 1H-NMR (CDCl3, 400 MHz) δ 9.63 (br s, 1H), 7.75 (d, J=8.4 Hz, 1H), 7.28˜7.11 (m, 4H), 7.04 (s, 1H), 6.84˜6.72 (m, 1H), 5.77 (d, J=17.2 Hz, 1H), 5.60 (d, J=11.2 Hz, 1H). MS (M+H)+: 273.


Step 18—Synthesis of 2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridine



embedded image


A mixture of 2-(6-chloro-3-vinylpyridin-2-yl)-4-fluoro-1H-indole (1.0 g, 3.7 mmol), DMSO (2.5 mL), TBAC (0.3 mL, 50% aq. solution) and KOH (10 mL, 60% a.q. solution) was stirred under N2 at 100° C. for 4 h. After cooled to RT and extracted with CH2Cl2, the organic layer was washed with water, brine, dried over Na2SO4 and concentrated on a rotary evaporator. The residue was purified by column chromatography (PE:EA=100:1˜10:1) to give the product of 2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridine (0.5 g, yield: 50%). And 150 mg of un-reacted 2-(6-chloro-3-vinylpyridin-2-yl)-4-fluoro-1H-indole was recycled during the column purification. 1H-NMR (CDCl3, 400 MHz) δ 7.50 (d, J=7.8 Hz, 1H), 7.38 (s, 1H), 7.22˜7.04 (m, 3H), 6.86˜6.71 (m, 1H), 4.27 (t, J=6.8 Hz, 2H), 3.22 (t, J=6.8 Hz, 2H). MS (M+H)+: 273.


Step 19—Synthesis of 5-(11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-N-methyl-6-(N-methylmethylsulfonamido)-2-(4-(trifluoromethyl)phenyl)benzofuran-3-carboxamide



embedded image


A mixture of N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2-(6-(trifluoromethyl)pyridin-3-yl)benzofuran-3-carboxamide (121 mg, 0.22 mmol), 2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridine (50 mg, 0.18 mmol) and K3CO3 (51 mg, 0.37 mmol) in dioxane/water (2 mL/0.4 mL) was stirred at room temperature for 10 min under N2 protection. Then Pd2(dba)3 (18 mg, 0.02 mmol) and X-Phos (17 mg, 0.04 mmol) were added to the reaction solution. The mixture was stirred at 90° C. for 2 hours under N2 protection. The mixture was then cooled to room temperature, added EA (10 mL) and then filtered through a Celite pad. The combined organic phase was washed with water and brine, dried over Na2SO4 and concentrated. The residue was purified by prep-HPLC to afford the desired product (crude: 70 mg, yield: 58%). 1H-NMR (CDCl3, 400 MHz) δ 9.29 (s, 1H), 8.63 (d, J=7.2 Hz, 1H), 7.95 (s, 1H), 7.67˜7.79 (m, 3H), 7.45 (d, J=7.6 Hz, 3H), 7.14˜7.24 (m, 3H), 6.78˜6.80 (m, 1H), 6.14˜6.15 (m, 1H), 4.33 (t, J=6.8 Hz, 1H), 3.29˜3.33 (m, 5H), 3.02 (d, J=4.8 Hz, 3H), 2.74 (s, 3H). MS (M+H)+: 663.


Examples 2-16

Examples 2-16, depicted in the table below, were prepared in accordance with the method described in Example 1.


















MS


Example
Structure
NMR
(M + H)+







 2


embedded image



1H-NMR (CDCl3, 400 MHz) δ 8.65 (s, 1H), 8.36 (d, J = 8.8 Hz, 1H), 7.89 (s, 1H), 7.67 (s, 0.25H), 7.61~7.64 (m, 2H), 7.49 (s, 0.5H), 7.40 (d, J = 8.0 Hz, 1H), 7.35 (s, 0.25H), 7.23 (s, 1H), 7.10~7.15 (m, 2H), 6.96 (d, J = 8.4 Hz, 1H), 6.61~6.72 (m, 1H), 5.95 (s, 1H), 4.28~4.31 (m, 2H), 3.30 (s, 1H), 3.21~3.29 (m, 2H), 2.99 (d, J = 4.8 Hz, 3H), 2.68 (s, 3H).

662





 3


embedded image



1H-NMR (CDCl3, 400 MHz) δ 9.22 (s, 1H), 8.55 (dd, J = 8.0, 1.6 Hz, 1H), 7.95 (s, 1H), 7.71~7.76 (m, 2H), 7.67 (d, J = 7.6 Hz, 1H), 7.45 (d, J = 7.6 Hz, 1H), 7.24 (s, 1H), 7.13~7.18 (m, 2H), 6.76~6.82 (m, 1H), 6.68 (s, 1H), 6.13 (d, J = 3.6 Hz, 1H), 4.33 (t, J = 6.8 Hz, 2H), 3.34 (s, 3H), 3.29~3.33 (m, 2H), 3.02 (d, J = 5.2 Hz, 3H), 2.73 (s, 3H).

646





 4


embedded image



1H-NMR (CDCl3, 400 MHz) δ 8.81 (d, J = 1.6 Hz, 1H), 8.35 (dd, J = 8.8, 1.6 Hz, 1H), 7.98 (s, 1H), 7.68~7.74 (m, 2H), 7.48 (d, J = 7.6 Hz, 1H), 7.31 (s, 1H), 7.15~7.22 (m, 2H), 7.01 (d, J = 8.8 Hz, 1H), 6.77~6.85 (m, 1H), 6.05 (d, J = 3.6 Hz, 1H), 4.86 (q, J = 8.8 Hz, 2H), 4.37 (t, J = 6.4 Hz, 2H), 3.38 (s, 3H), 3.34 (t, J = 6.4 Hz, 2H), 3.04 (d, J = 5.2 Hz, 3H), 2.74 (s, 3H).

694





 5


embedded image



1H-NMR (CDCl3, 400 MHz) δ 8.80 (d, J = 1.6 Hz, 1H), 8.22 (dd, J = 2.4, 8.8 Hz, 1H), 7.97 (s, 1H), 7.64~7.69 (m, 2H), 7.44 (d, J = 7.6 Hz, 1H), 7.24 (s, 1H), 7.12~7.17 (m, 2H), 6.88 (d, J = 8.8 Hz, 1H), 6.78 (dd, J = 7.2, 8.61 Hz, 1H), 6.02 (d, J = 4.4 Hz, 1H), 4.33 (t, J = 6.4 Hz, 2H), 4.28 (dt, J = 3.2, 5.6 Hz, 1H), 3.36 (s, 3H), 3.30 (t, J = 6.4 Hz, 2H), 2.98 (d, J = 5.2 Hz, 3H), 2.69 (s, 3H), 0.83 (d, J = 3.6 Hz, 2H), 0.81 (br s., 2H).

652





 6


embedded image



1H-NMR (CDCl3, 400 MHz) δ 8.94~8.97 (m, 1H), 8.14 (dd, J = 2.0, 8.0 Hz, 1H), 7.98 (s, 1H), 7.64~7.68 (m, 2H), 7.44 (d, J = 8.0 Hz, 1H), 7.25~7.29 (m, 2H), 7.12~7.18 (m, 2H), 6.74~6.81 (m, 1H), 6.01 (d, J = 4.4 Hz, 1H), 4.33 (t, J = 6.8 Hz, 2H), 3.36 (s, 3H), 3.30 (t, J = 6.4 Hz, 2H), 2.98 (d, J = 4.8 Hz, 3H), 2.69 (s, 3H), 2.06~2.13 (m, 1H), 1.11 (dd, J = 2.4, 4.4 Hz, 2H), 1.04~1.09 (m, 2H).

636





 7


embedded image



1H-NMR (CDCl3, 400 MHz) δ 9.00 (s, 1H), 8.18~8.20 (m, 1H), 7.97 (s, 1H), 7.64~7.67 (m, 2H), 7.42 (d, J = 7.6 Hz, 1H), 7.27 (d, J = 2.8 Hz, 2H), 7.13~7.19 (m, 2H), 6.76-6.80 (m, 1H), 6.20 (br s, 1H), 4.31~4.34 (m, 2H), 3.36 (s, 3H), 3.28~3.31 (m, 2H), 2.98 (d, J = 4.8 Hz, 3H), 2.69 (s, 3H), 2.61 (m., 3H).

610





 8


embedded image



1H-NMR (DMSO-d6, 400 MHz) δ 8.36 (s, 1H), 7.94 (s, 1H), 7.82 (s, 1H), 7.62 (s, 2H), 7.40 (d, J = 8.0 Hz, 1H), 7.23 (s, 2H), 7.09 (s, 2H), 6.74 (d, J = 8.0 Hz, 0.4H), 5.91 (s, 0.4H), 4.46 (s, 2H), 4.27~4.29 (m, 4H), 3.32 (s, 2H), 3.27 (d, J = 6.0 Hz, 2H), 2.95 (d, J = 4.8 Hz, 3H), 2.65 (s, 3H).

654





 9


embedded image



1H-NMR (CDCl3, 400 MHz) δ 9.05 (s, 1H), 8.23 (dd, J1 = 8.4 Hz, J2 = 2.0 Hz, 1H), 7.99 (s, 1H), 7.69 (s, 1H), 7.66 (d, J = 8.0 Hz, 1H), 7.44 (d, J = 8.0 Hz, 1H), 7.24~7.31 (m, 2H), 7.12~7.16 (m, 2H), 6.77~6.80 (m, 1H), 5.96 (br s, 1H), 4.33 (t, J = 6.4 Hz, 2H), 3.36 (s, 3H), 3.31 (t, J = 6.4 Hz, 2H), 2.99 (d, J = 4.8 Hz, 3H), 2.87~2.92 (m, 2H), 2.70 (s, 3H), 1.35 (t, J = 7.2 Hz, 3H).

624





10


embedded image



1H-NMR (CDCl3, 400 MHz) δ 8.29 (s, 1H), 7.61 (d, J = 8.0 Hz, 2H), 7.40~7.47 (m, 2H), 7.22 (s, 1H), 7.02~7.10 (m, 4H), 6.74 (d, J = 7.8 Hz, 1H), 5.43 (br s, 1H), 4.29 (t, J = 6.4 Hz, 2H), 3.37 (s, 3H), 3.23~3.28 (m, 2H), 2.77 (d, J = 4.8 Hz, 3H), 2.57 (s, 3H), 2.30 (s, 3H).

627





11


embedded image



1H-NMR (CDCl3, 400 MHz) δ 8.69 (d, J = 4.4 Hz, 1H), 8.64 (d, J = 5.2 Hz, 1H), 8.10 (s, 1H), 7.85 (d, J = 4.8 Hz, 2H), 7.74 (s, 1H), 7.66 (d, J = 4.8 Hz, 1H), 7.52 (d, J = 8.0 Hz, 1H), 7.40 (d, J = 8.0 Hz, 1H), 7.15~7.18 (m, 1H), 7.07 (s, 1H), 6.83~6.86 (m, 1H), 4.39 (t, J = 6.4 Hz, 2H), 3.25~3.32 (m, 5H), 3.05~3.12 (m, 1H), 2.93 (s, 3H), 2.82 (d, J = 4.8 Hz, 3H), 1.26 (d, J = 6.8 Hz, 6H).

638





12


embedded image



1H-NMR (CDCl3, 400 MHz) δ 8.22 (s, 1H), 7.66~7.69 (m, 3H), 7.47 (d, J = 8.0 Hz, 1H), 7.32~7.35 (m, 1H), 7.28 (s, 1H), 7.16~7.18 (m, 3H), 6.79~6.81 (m, 1H), 5.64 (br s, 1H), 4.33~4.36 (m, 2H), 3.40 (s., 3H), 3.30~3.33 (m, 2H), 2.90 (d, J = 4.8 Hz, 3H), 2.67 (s, 3H).

647





13


embedded image



1H-NMR (CDCl3, 400 MHz) δ 8.03 (s, 1H), 7.89~8.00 (m, 1H), 7.75 (s, 1H), 7.70~7.72 (m, 1H), 7.47~7.55 (m, 3H), 7.31 (s, 1H), 7.14~7.19 (m, 2H), 6.80~6.81 (m, 1H), 6.11~6.12 (m, 1H), 4.37 (t, J = 6.8 Hz, 2H), 3.37 (s, 3H), 3.34 (t, J = 6.8 Hz, 2H), 3.02 (d, J = 4.8 Hz, 3H), 2.77 (s, 3H).

638





14


embedded image



1H-NMR (CDCl3, 400 MHz) δ 8.82 (s, 1H), 8.05 (d, J = 8.0 Hz, 1H), 7.89 (s, 1H), 7.56~7.69 (m, 2H), 7.39 (d, J = 4.8 Hz, 1H), 7.22 (s, 1H), 7.07~7.12 (m, 2H), 6.73 (t, J = 8.0 Hz, 1H), 6.17 (br s., 1H), 4.28 (t, J = 6.0 Hz, 2H), 3.29 (s, 3H), 3.25 (t, J = 6.0 Hz, 2H), 2.96 (d, J = 4.8 Hz, 3H), 2.67 (s, 3H), 2.53 (s, 3H).

628





15


embedded image



1H-NMR (CDCl3, 400 MHz) δ 8.85 (s, 1H), 8.55~8.59 (m, 1H), 7.95 (s, 1H), 7.67~7.71 (m, 2H), 7.46 (d, J = 8.0 Hz, 1H), 7.28 (s, 1H), 7.13~7.18 (m, 2H), 7.05~7.08 (m, 1H), 6.78~6.81 (m, 1H), 6.06 (br s, 1H), 4.34 (t, J = 6.4 Hz, 2H), 3.35 (s, 3H), 3.33 (d, J = 6.4 Hz, 2H), 3.02 (d, J = 4.8 Hz, 3H), 2.73 (s, 3H).

614





16


embedded image



1H-NMR (CDCl3, 400 MHz) δ 7.85 (s, 1H), 7.65 (d, J = 7.6 Hz, 1H), 7.50 (s, 1H), 7.43 (d, J = 7.6 Hz, 1H), 7.28 (s, 1H), 7.11~7.19 (m, 2H), 6.76~6.83 (m, 1H), 6.12 (d, J = 3.6 Hz, 1H), 4.33 (t, J = 6.4 Hz, 2H), 3.33 (s, 3H), 3.29 (t, J = 6.4 Hz, 2H), 3.02 (d, J = 4.8 Hz, 3H), 2.80~2.87 (m, 1H), 2.69 (s, 3H), 1.25 (d, J = 2.4 Hz, 2H), 1.15~1.21 (m, 2H).

559









Example 17



embedded image


Step 1—Synthesis of 5-bromo-2-(1-isopropyl-1H-pyrazol-5-yl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a solution of 5-bromo-2-iodo-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (50 mg, 0.12 mmol), (1-isopropyl-1H-pyrazol-5-yl)boronic acid (23 mg, 0.12 mmol) and Na2CO3 (22 mg, 0.25 mmol) in 1,4-dioxane (0.5 mL) and water (0.05 mL) was added Pd (dppf)Cl2 (10 mg) under nitrogen. The mixture was heated at 100° C. for 4 hour and filtered through the Celite pad. The filtrate was extracted with EtOAc, and then the combined organic phase was washed with brine, dried over Na2SO4 and concentrated in vacuo. The crude product was purified by the prep-TLC (PE:EA=1:1) to give 5-bromo-2-(1-isopropyl-1H-pyrazol-5-yl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (25 mg, yield: 50%). 1H-NMR (CDCl3, 400 MHz) δ 8.50 (s, 1H), 7.66 (s, 1H), 7.64 (s, 1H), 6.58 (d, J=2.0 Hz, 1H), 5.74 (d, J=3.6 Hz, 1H), 4.45˜4.52 (m, 1H), 3.28 (s, 3H), 3.04 (s, 3H), 2.82 (d, J=4.8 Hz, 3H), 1.51 (s, 6H). MS (M+H)+: 469/471.


Step 2—Synthesis of 2-(1-isopropyl-1H-pyrazol-5-yl)-N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide



embedded image


To a degassed solution of 5-bromo-2-(1-isopropyl-1H-pyrazol-5-yl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (60 mg, 0.12 mmol) and bis(pinacolato)diboron (100 mg, 0.42 mmol) in 1,4-dioxane (1.0 mL) was added Pd(dppf)Cl2 (5 mg) and KOAc (50 mg, 0.50 mmol) under N2. The mixture was heated at a pre-heated oil-bath at 110° C. for 3 h. The reaction mixture was concentrated in vacuo and it was extracted with EtOAc. The residue was washed with H2O, brine, and dried over Na2SO4. After concentration, the crude product was purified by prep-TLC (PE:EA=2:1) to give of 2-(1-isopropyl-1H-pyrazol-5-yl)-N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide (25 mg, yield: 30%). 1H-NMR (CDCl3, 400 MHz) δ 8.58 (s, 1H), 7.62 (s, 1H), 7.55 (s, 1H), 6.59 (d, J=1.6 Hz, 1H), 5.74 (s, 1H), 4.49˜4.56 (m, 1H), 3.30 (s, 3H), 2.93 (s, 3H), 2.84 (d, J=4.8 Hz, 3H), 1.44 (d, J=6.4 Hz, 6H), 1.30 (s, 12H). MS (M+H)+: 517.


Step 3—Synthesis of 5-(11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-2-(1-isopropyl-1H-pyrazol-5-yl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a solution of 2-(1-isopropyl-1H-pyrazol-5-yl)-N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide (78 mg, 0.14 mmol), 2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridine (35 mg, 0.13 mmol) and K2CO3 (27 mg, 0.19 mmol) in 1,4-dioxane (0.8 mL) and water (0.05 mL) were added X-Phos (10 mg) and Pd2(dba)3 (20 mg) under nitrogen. The mixture was heated at 120° C. for 2 hours and filtered through the Celite pad. The filtrate was extracted with EtOAc, then the combined organic phase was washed with brine, dried over Na2SO4 and concentrated in vacuo. The crude product was purified by the prep-HPLC to give the desired compound (30 mg, yield: 42%). 1H-NMR (CDCl3, 400 MHz) δ 8.41 (s, 1H), 7.72 (s, 1H), 7.69 (d, J=7.6 Hz, 1H), 7.47 (d, J=7.6 Hz, 1H), 7.28 (s, 1H), 7.17˜7.19 (m, 2H), 6.79˜6.83 (m, 1H), 6.69 (s, 1H), 5.85 (s, 1H), 4.61˜4.64 (m, 1H), 4.35˜4.38 (m, 2H), 3.44 (s, 3H), 3.32˜3.56 (m, 2H), 2.90 (d, J=5.2 Hz, 3H), 2.64 (s, 3H). MS (M+H)+: 627.


Examples 18-27

Examples 18˜27, depicted in the table below, were prepared in accordance with the method described in Example 17.


















MS


Example
Structure
NMR
(M + H)+







18


embedded image



1H-NMR (CDCl3, 400 MHz) δ 8.99 (s, 1H), 8.30 (s, 1H), 7.85 (s, 1H), 7.74 (d, J = 8.0 Hz, 2H), 7.42~7.46 (m, 3H), 7.20~7.30 (m, 1H), 7.15 (s, 1H), 7.08~7.15 (m, 2H), 6.74 (t, J = 8.4 Hz, 1H), 6.08 (s, 1H), 4.30 (t, J = 6.4 Hz, 2H), 3.30 (s, 3H), 3.26 (d, J = 6.4 Hz, 2H), 3.01 (d, J = 4.8 Hz, 3H), 2.72 (s, 3H).

661





19


embedded image



1H-NMR (CDCl3, 400 MHz) δ 8.14 (s, 1H), 7.90 (s, 1H), 7.61 (d, J = 7.6 Hz, 1H), 7.55 (s, 1H), 7.39 (d, J = 8.0 Hz, 1H), 7.22 (s, 1H), 7.08 (d, J = 6.4 Hz, 2H), 6.73 (t, J = 7.2 Hz, 1H), 5.95 (s, 1H), 4.29 (t, J = 6.4 Hz, 2H), 4.17 (t, J = 6.4 Hz, 2H), 3.32 (s, 3H), 3.26 (t, J = 6.4 Hz, 2H), 3.14 (t, J = 6.4 Hz, 2H), 2.96 (d, J = 4.2 Hz, 3H), 2.67 (t, J = 6.4 Hz, 2H), 2.61 (s, 3H).

625





20


embedded image



1H-NMR (CDCl3, 400 MHz) δ 8.08 (s, 1H), 7.85 (s, 1H), 7.64 (d, J = 7.60 Hz, 1H), 7.57 (s, 1H), 7.44 (d, J = 8.00 Hz, 1H), 7.29 (s, 1H), 7.12~7.19 (m, 2H), 6.76~6.80 (m, 1H), 6.23 (br s, 1H), 4.31~4.34 (m, 2H), 3.53~3.55 (m, 4H), 3.34 (s, 3H), 3.28~3.31 (m, 2H), 3.02 (d, J = 4.80 Hz, 3H), 2.75 (s, 3H), 2.09~2.14 (m, 4H).

660





21


embedded image



1H-NMR (CDCl3, 400 MHz) δ 8.06 (s, 1H), 8.02 (s, 1H), 7.65 (d, J = 8.0 Hz, 1H), 7.61 (s, 1H), 7.43 (d, J = 8.0 Hz, 1H), 7.24 (s, 1H), 7.09~7.19 (m, 2H), 6.73~6.81 (m, 1H), 6.01 (d, J = 3.6 Hz, 1H), 4.33 (t, J = 6.0 Hz, 2H), 4.22 (t, J = 5.6 Hz, 2H), 3.38 (s, 3H), 3.30 (t, J = 6.0 Hz, 2H), 3.06 (t, J = 5.6 Hz, 2H), 2.97 (d, J = 4.4 Hz, 3H), 2.63 (s, 3H), 2.10 (d, J = 4.8 Hz, 2H), 1.94 (d, J = 4.8 Hz, 2H).

639





22


embedded image



1H-NMR (CDCl3, 400 MHz) δ 8.08 (s, 1H), 7.85 (s, 1H), 7.64 (d, J = 7.60 Hz, 1H), 7.57 (s, 1H), 7.44 (d, J = 8.00 Hz, 1H), 7.29 (s, 1H), 7.12~7.19 (m, 2H), 6.76~6.80 (m, 1H), 6.23 (br s, 1H), 4.31~4.34 (m, 2H), 3.53~3.55 (m, 4H), 3.34 (s, 3H), 3.28~3.31 (m, 2H), 3.02 (d, J = 4.80 Hz, 3H), 2.75 (s, 3H), 2.09~2.14 (m, 4H).

671





23


embedded image



1H-NMR (CDCl3, 400 MHz) δ 8.45 (s, 1H), 8.07 (s, 1H), 7.80 (s, 1H), 7.15 (d, J = 7.2 Hz, 1H), 7.54 (s, 1H), 7.25 (d, J = 8.0 Hz, 1H), 7.25 (s, 1H), 7.10 (d, J = 6.0 Hz, 2H), 6.71~6.75 (m, 1H), 6.21 (d, J = 4.0 Hz, 1H), 4.28 (d, J = 6.8 Hz, 2H), 4.23~4.26 (m, 2H), 3.99 (s, 2H), 3.28 (s, 3H), 3.25 (s, 2H), 2.96 (d, J = 4.8 Hz, 3H), 2.70 (s, 3H).

629





24


embedded image



1H-NMR (CDCl3, 400 MHz) δ 8.89 (s, 1H), 8.55 (d, J = 8.8 Hz, 1H), 8.39 (d, J = 3.6 Hz, 1H), 7.95 (s, 1H), 7.64-7.72 (m, 2H), 7.44 (d, J = 8.0 Hz, 1H), 7.21 (dd, J = 4.4, 9.2 Hz, 2H), 7.14 (br s., 2H), 6.74~6.81 (m, 1H), 6.11 (d, J = 3.6 Hz, 1H), 4.34 (t, J = 6.4 Hz, 2H), 3.36 (s, 3H), 3.31 (t, J = 6.4 Hz, 2H), 3.02 (d, J = 4.8 Hz, 3H), 2.69 (s, 3H).

636





25


embedded image



1H-NMR (CDCl3, 400 MHz) δ 10.15 (s, 1H), 8.48 (s, 1H), 8.00~8.09 (m, 2H), 7.86 (s, 1H), 7.51 (m, J = 8.4 Hz, 1H), 7.40 (s, 1H), 7.16 (d, J = 8.4 Hz, 1H), 7.07 (s, 1H), 6.95 (s, 1H), 6.84 (s, 1H), 4.40 (t, J = 8.4 Hz, 2H), 2.93 (s, 3H), 2.88 (d, J = 4.8 Hz, 3H), 2.53 (s, 3H), 2.50 (t, J = 8.4 Hz, 2H).

585





26


embedded image



1H-NMR (CDCl3, 400 MHz) δ 8.26 (s, 1H), 7.60 (d, J = 8.0 Hz, 1H), 7.59 (s, 1H), 7.40 (d, J = 8.0 Hz, 1H), 7.20 (s, 1H), 7.09~7.12 (m 2H), 6.70~6.74 (m 1H), 5.76 (br s, 1H), 4.28 (t, J = 6.4 Hz, 2H), 3.37 (s, 3H), 3.25 (t, J = 6.4 Hz, 2H), 2.84 (d, J = 4.8 Hz, 3H), 2.56 (s, 3H), 2.29 (s, 6H).

613





27


embedded image



1H-NMR (DMSO-d6, 400 MHz) δ 8.50 (s, 1H), 8.14 (s, 1H), 7.89 (s, 1H), 7.67 (d, J = 8.0 Hz, 1H), 7.62 (s, 1H), 7.48 (d, J = 8.0 Hz, 1H), 7.31 (s, 1H), 7.14~7.19 (m, 2H), 6.80 (t, J = 8.0 Hz, 1H) 6.20 (br s, 1H), 4.34 (t, J = 6.8 Hz, 2H), 4.25 (dd, J1 = 14.4 Hz, J2 = 7.6 Hz, 2H), 3.35 (s, 3H), 3.31 (t, J = 6.8 Hz, 2H), 3.03 (d, J = 4.8 Hz, 3H), 2.76 (s, 3H), 1.56 (t, J = 7.2 Hz, 3H).

613





28


embedded image



1H-NMR (CDCl3, 400 MHz) δ 8.89 (s, 1H), 7.92 (s, 1H), 7.56~7.65 (m, H), 7.38 (d, J = 8.0 Hz, 1H), 7.16~7.23 (m, 2H), 7.05~7.13 (m, 2H), 6.68~6.75 (m, 1H), 5.90~6.01 (br s, 1H), 4.28 (t, J = 6.4 Hz, 2H), 3.30 (s, 3H), 3.23 (t, J = 6.4 Hz, 2H), 2.91 (d, J = 4.8 Hz, 3H), 2.63 (s, 3H), 1.98~2.10 (m, 1H), 0.98~1.09 (m, 4H).

625









Example 29



embedded image


Step 1—Synthesis of 5-bromo-2-(4-formylphenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a degassed solution of 5-bromo-2-iodo-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (400 mg, 0.82 mmol), (4-formylphenyl) boronic acid (110 mg, 0.74 mmol) and Na2CO3 (174 mg, 1.6 mmol) in DMF (4 mL) was added Pd(dppf)Cl2 (20 mg) under N2, and the mixture was stirred at 100° C. for 2 h. After the solvent was removed, the residue was purified by column chromatography (DCM:EtOAc=10:1) to give the product of 5-bromo-2-(4-formylphenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (200 mg, yield: 52%). 1H-NMR (CDCl3, 400 MHz) δ 10.09 (s, 1H), 8.09˜8.15 (m, 3H), 8.02 (s, 2H), 7.75 (s, 1H), 5.90 (br s., 1H), 3.36 (s, 3H), 3.11 (s, 3H), 3.05 (d, J=4.8 Hz, 3H). MS (M+H)+: 465/467.


Step 2—Synthesis of 5-bromo-2-(4-(difluoromethyl)phenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a solution of 5-bromo-2-(4-formylphenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (50 mg, 0.1 mmol) in DCM (1 mL) was added dropwise DAST (0.2 mL) at −78° C. Then the mixture was stirred at 25° C. for 5 hours. Then MeOH (3 ml) was added, the pH of mixture was adjusted to 7 and extracted with DCM (10 ml). The organic phase was dried over Na2SO4. After concentrated in vacuo, the residue was purified by prep-TLC (DCM:EtOAc=10:1) to give the product of 5-bromo-2-(4-(difluoromethyl)phenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (26 mg, yield: 50%). 1H-NMR (CDCl3, 400 MHz) δ 8.16 (s, 1H), 7.99 (d, J=8.0 Hz, 2H), 7.73 (s, 1H), 7.66 (d, J=8.0 Hz, 2H), 6.56˜6.88 (m, 1H), 5.85 (br s., 1H), 3.35 (s, 3H), 3.10 (s, 3H), 3.02 (d, J=5.2 Hz, 3H). MS (M+H)+: 487/489.


Step 3—Synthesis of 2-(4-(difluoromethyl)phenyl)-N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide



embedded image


To a degassed solution of 5-bromo-2-(4-(difluoromethyl)phenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (50 mg, 0.1 mmol), KOAc (30 mg, 0.3 mmol) and bis(pinacolato)diboron (130 mg, 0.5 mmol) in 1,4-dioxane (2 mL) and H2O (0.1 mL) was added Pd(dppf)Cl2 (5 mg) under N2, and the mixture was stirred at 130° C. for 3 hours. After the solvent was removed, the residue was purified by column chromatography (DCM:EtOAc=10:1) to give the product of 2-(4-(difluoromethyl)phenyl)-N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide (30 mg, yield: 55%). 1H-NMR (CDCl3, 400 MHz) δ 8.16 (s, 1H), 7.99 (d, J=8.0 Hz, 2H), 7.73 (s, 1H), 7.66 (d, J=80 Hz, 2H), 6.56˜6.88 (m, 1H), 5.85 (br s., 1H), 3.35 (s, 3H), 3.10 (s, 3H), 3.02 (d, J=5.2 Hz, 3H), 1.33 (s, 12H). MS (M+H)+: 535.


Step 4—Synthesis of 2-(4-(difluoromethyl)phenyl)-5-(11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a degassed solution of 2-(4-(difluoromethyl)phenyl)-N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide (120 mg, 0.22 mmol), 2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridine (64 mg, 0.22 mmol), Na2CO3 (62 mg, 0.45 mmol), in 1,4-dioxane (3 mL) and H2O (0.1 mL) was added Pd2(dba)3 (5 mg) and X-Phos (5 mg) under N2. Then the mixture was stirred at 100° C. for 2 hours. The reaction mixture was cooled to RT and filtered. The filtrate was washed with H2O and dried over Na2SO4. After concentrated, the residue was purified by prep-HPLC to give the product (30 mg, yield: 21%). 1H-NMR (CDCl3, 400 MHz) δ 8.08 (d, J=8.0 Hz, 2H), 8.03 (s, 1H), 7.74˜7.64 (m, 4H), 7.47 (d, J=7.6 Hz, 1H), 7.30 (s, 1H), 7.22˜7.14 (m, 2H), 6.89˜6.81 (m, 1H), 6.81˜6.73 (m, 1H), 6.00 (d, J=4.4 Hz, 1H), 4.37 (t, J=6.4 Hz, 2H), 3.40 (s, 3H), 3.34 (t, J=6.4 Hz, 2H), 3.02 (d, J=4.8 Hz, 3H), 2.72 (s, 3H). MS (M+H)+: 645.


Example 30



embedded image


Step 1—Synthesis of 6-amino-5-bromo-2-(2,4-difluorophenyl)-N-methylbenzofuran-3-carboxamide



embedded image


To a mixture of (6-amino-5-bromo-3-(methylcarbamoyl)benzofuran-2-yl)boronic acid (4.78 g, 11.46 mmol), K3PO4.3H2O (8.32 g, 31.25 mmol) and 2,4-difluoro-1-iodobenzene (2.50 g, 10.42 mmol) in DMF (25 mL) was added Pd(dppf)Cl2 (380 mg, 0.52 mmol) under N2, and then the mixture was stirred at 15° C. for 3 hours. After the solvent was removed, EtOAc/H2O (10 mL/20 mL) was added to the mixture. After stirring 15 minutes, the resulting solid was filtered to give the product of 6-amino-5-bromo-2-(2,4-difluorophenyl)-N-methylbenzofuran-3-carboxamide (2.80 g, yield: 70%). 1H-NMR (DMSO-d6, 400 MHz) δ 8.06 (d, J=1.6 Hz, 1H), 7.74 (d, J=10.4 Hz, 1H), 7.70 (s, 1H), 7.41 (s, 1H), 7.24 (s, 1H), 7.00 (s, 1H), 5.58 (br s, 2H), 2.76 (s, 3H). MS (M+H)+: 281/283


Step 2—Synthesis of 5-bromo-2-(2,4-difluorophenyl)-N-methyl-6-(methylsulfonamido)benzofuran-3-carboxamide



embedded image


To a mixture of (6-amino-5-bromo-3-(methylcarbamoyl)benzofuran-2-yl)boronic acid (400 mg, 1.01 mmol) and pyridine (830 mg, 10.49 mmol) in DCM (15 mL), MsCl (601 mg, 5.25 mmol) was added dropwise at 0° C. The mixture was allowed to room temperature and stirred overnight. The reaction mixture was quenched with NaHCO3 and extracted with DCM. The organic phase was washed with brine, dried over Na2SO4, and concentrated. The residue was dissolved in THF:H2O=5:1 (15 mL) and LiOH.H2O (800 mg, 0.02 mmol) was added. The mixture was stirred for 1 hour at room temperature. EtOAc (300 mL) was added, and the organic phase washed with NH4Cl(a.q.), brine, dried over Na2SO4, filtrated and concentrated in vacuo. The residue was purified by column chromatography (eluted with DCM/EtOAc=10/1 to 2/1) to give 5-bromo-2-(2,4-difluorophenyl)-N-methyl-6-(methylsulfonamido)benzofuran-3-carboxamide (210 mg, yield: 43%). 1H-NMR (CDCl3, 400 MHz) δ 8.32 (s, 1H), 7.91 (s, 1H), 7.64˜7.90 (m, 1H), 7.06 (d, J=3.0 Hz, 1H), 7.03 (d, J=3.0 Hz, 1H), 6.88 (s, 1H), 3.02 (s, 3H), 2.97 (d, J=4.8 Hz, 3H). MS (M+H)+: 459/461


Step 3—Synthesis of 5-bromo-2-(2,4-difluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a mixture of 5-bromo-2-(2,4-difluorophenyl)-N-methyl-6-(methylsulfonamido)benzofuran-3-carboxamide (653 mg, 1.38 mmol), K2CO3 (406 mg, 2.94 mmol) in DMF (10 mL) was added MeI (519 mg, 3.66 mmol), then the mixture was stirred at 80° C. After 3 hours, the solvent was removed by vacuum, the mixture was washed with H2O (20 mL) and extract with DCM (50 mL×3), dried over Na2SO4 and concentrated to give the desired product of 5-bromo-2-(2,4-difluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (600 mg, yield: 89%). 1H-NMR (CDCl3, 400 MHz) δ 8.24 (s, 1H), 7.69˜7.75 (m, 2H), 7.05˜7.10 (m, 1H), 6.98˜7.03 (m, 1H), 5.64 (d, J=3.0 Hz, 1H), 3.09 (s, 3H), 2.97 (s, 3H), 2.95 (d, J=3.6 Hz, 3H). MS (M+H)+: 473/475.


Step 4—Synthesis of 2-(2,4-difluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide



embedded image


To a degassed mixture of 5-bromo-2-(2,4-difluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (400 mg, 0.85 mmol), bis(pinacolato)diboron (1 g, 4.23 mmol), KOAc (249 mg, 2.54 mmol) in 1,4-Dioxane (5 mL) and H2O (1 mL) was added Pd(dppf)Cl2 (5 mg), then the mixture was stirred at 130° C. After 3 hours, the solvent was removed by vacuum, and the mixture was washed with H2O (20 mL), extract with DCM (50 mL×3), dried over Na2SO4. After concentrated, the residue was purified by column chromatography (PE/EtOAc=2/1) to give the product of 2-(2,4-difluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide (240 mg, yield: 54%). 1H-NMR (CDCl3, 400 MHz) δ 8.26 (s, 1H), 7.73 (d, J=6.8 Hz, 1H), 7.56 (s, 1H), 7.05 (t, J=6.8 Hz, 1H), 6.96 (q, J=6.8 Hz, 1H), 5.96 (s, 1H), 3.33 (s, 3H), 2.97 (s, 3H), 2.93 (d, J=4.8 Hz, 3H), 1.20 (s, 12H). MS (M+H)+: 521.


Step 5—Synthesis of 2-(2,4-difluorophenyl)-5-(11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a solution of 2-(2,4-difluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide (500 mg, 0.96 mmol), 2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridine (240 mg, 0.87 mmol) and K2CO3. (241 mg, 1.75 mmol) in 1,4-dioxane (5 mL) and H2O (5 drop) were added X-Phos (10 mg) and Pd2(dba)3 (10 mg) under N2. The reaction mixture was stirred at 100° C. for 10 hours and concentrated in vacuo to remove 1,4-dioxane. The reaction mixture was extracted with EtOAc. The organic phase was washed with brine, dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by prep-HPLC to give the product (230 mg, yield: 42%). 1H-NMR (CDCl3, 400 MHz) δ 8.08 (s, 1H), 7.7˜57.81 (m, 1H), 7.66˜7.69 (m, 2H), 7.46 (d, J=8.0 Hz, 1H), 7.28 (s, 1H), 7.15˜7.18 (m, 2H), 6.98˜7.09 (m, 2H), 6.79 (t, J=8.4 Hz, 1H), 5.81 (br s, 1H), 4.35 (t, J=6.4 Hz, 2H), 3.39 (s, 3H), 3.32 (t, J=6.4 Hz, 2H), 2.96 (d, J=4.8 Hz, 3H), 2.68 (s, 3H). MS (M+H)+: 631.


Example 31



embedded image


Step 1—Synthesis of 6-chloro-2-(5-fluoro-1H-indol-2-yl)pyridin-3-ol



embedded image


To a degassed solution of 6-chloro-2-iodopyridin-3-ol (1.0 g, 3.91 mmol) and (1-(tert-butoxycarbonyl)-5-fluoro-1H-indol-2-yl)boronic acid (prepared using the method described in Example 1, 1.3 g, 4.66 mmol) in 1,4-dioxane-H2O (20 mL, 10:1), Pd(PPh3)2Cl2 (120 mg) and NaHCO3 (1.0 g, 11.90 mmol) were added under N2. The mixture was heated to 100° C. overnight. The reaction mixture was cooled to room temperature, filtered and washed with EtOAc. The filtrate was washed with H2O, brine, dried over Na2SO4. After concentrated, the residue was purified by column chromatography (PE:EA 5:1) to give the product of 6-chloro-2-(5-fluoro-1H-indol-2-yl)pyridin-3-ol (600 mg, yield: 58.2%). 1H-NMR (DMSO-d6, 400 MHz) δ 11.37 (br s, 1H), 11.03 (s, 1H), 7.51 (dd, J=8.8, 4.8 Hz, 1H), 7.41 (d, J=8.4 Hz, 1H), 7.34 (dd, J=10.0, 2.4 Hz, 1H), 7.30 (d, J=1.2 Hz, 1H), 7.26 (d, J=8.4 Hz, 1H), 6.94˜6.99 (m, 1H). MS (M+H)+: 263/265.


Step 2—Synthesis of 6-chloro-2-(5-fluoro-1H-indol-2-yl)pyridin-3-yl trifluoromethanesulfonate



embedded image


To a degassed solution of 6-chloro-2-(5-fluoro-1H-indol-2-yl)pyridin-3-ol (600 mg, 2.28 mmol) and DIPEA (1.0 g, 7.74 mmol) in DCM (10 mL), Tf2O (1.3 g, 4.61 mmol) were added dropwise under N2 at 0° C. The mixture was stirred at RT for 2 hours. The mixture was diluted with water and extracted with DCM. The organic layers were washed with brine, dried over Na2SO4, and concentrated. The mixture was purified by column chromatography (PE:EA=10:1) to give 6-chloro-2-(5-fluoro-1H-indol-2-yl)pyridin-3-yl trifluoromethanesulfonate (700 mg, yield: 77.6%). 1H-NMR (DMSO-d6, 400 MHz) δ 11.91 (br s, 1H), 8.17 (d, J=8.8 Hz, 1H), 7.66 (d, J=8.8 Hz, 1H), 7.51˜7.58 (m, 1H), 7.42˜7.50 (m, 1H), 7.18 (d, J=1.6 Hz, 1H), 7.06˜7.14 (m, 1H). MS (M+H)+: 395/397.


Step 3—Synthesis of 2-(6-chloro-3-vinylpyridin-2-yl)-5-fluoro-1H-indole



embedded image


To a mixture of 6-chloro-2-(5-fluoro-1H-indol-2-yl)pyridin-3-yl trifluoromethanesulfonate (200 mg, 0.51 mmol), tributyl(vinyl)stannane (170 mg, 0.54 mmol) and anhydrous LiCl (70 mg, 1.65 mmol) in DMF (2 mL), Pd(PPh3)2Cl2 (30 mg, 0.04 mmol) was added under N2 protection. The reaction mixture was stirred at 50° C. for 1 h. Then it was filtered and concentrated in vacuo, the residue was purified by column chromatography (PE: EA=20:1) to give the product of 2-(6-chloro-3-vinylpyridin-2-yl)-5-fluoro-1H-indole (100 mg, yield: 72.4%). 1H-NMR (CDCl3, 400 MHz) δ 11.67 (br s, 1H), 8.05 (d, J=8.4 Hz, 1H), 7.45˜7.55 (m, 2H), 7.38 (d, J=10.0 Hz, 1H), 7.15 (dd, J=17.2, 11.2 Hz, 1H), 7.00˜7.06 (m, 1H), 6.84 (d, J=2.0 Hz, 1H), 5.93 (d, J=17.2 Hz, 1H), 5.59 (d, J=11.2 Hz, 1H). MS (M+H)+: 273/275.


Step 4—Synthesis of 2-(6-chloro-3-vinylpyridin-2-yl)-5-fluoro-1H-indole



embedded image


A mixture of 2-(6-chloro-3-vinylpyridin-2-yl)-5-fluoro-1H-indole (100 mg, 0.36 mmol), DMSO (0.5 mL), TBAC (0.1 mL of 50% a.q. solution) and KOH (1.0 mL of 60% a.q. solution) was stirred under N2 at 100° C. for 4 h. After cooled and extracted with EtOAc, the organic layer was washed with water, brine, dried over Na2SO4, and concentrated. The residue was purified by prep-TLC (PE:EA=10:1) to give the product of 2-chloro-10-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridine (50 mg, yield: 50.0%). 1H-NMR (CDCl3, 400 MHz) δ 7.51 (d, J=8.0 Hz, 1H), 7.25˜7.38 (m, 3H), 7.15 (d, J=8.0 Hz, 1H), 6.98˜7.04 (m, 1H), 4.26 (d, J=6.4 Hz, 2H), 3.22 (d, J=6.4 Hz, 2H). MS (M+H)+: 273/275.


Step 5—Synthesis of 5-(10-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a degassed solution of 2-chloro-10-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridine (50 mg, 0.18 mmol) and 2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide (80 mg, 0.16 mmol) in 1,4-dioxane (5 mL), Pd2(dba)3 (10 mg), X-Phos (10 mg) and K3PO4 (100 mg, 0.38 mmol) were added under N2. After heated to 100° C. for 1 hour, the reaction mixture was cooled to RT, filtered and washed with EtOAc. The filtrate was washed with H2O, brine, dried over Na2SO4. After concentrated, the residue was purified by prep-HPLC to give the product (60 mg, yield: 61.5%). 1H-NMR (CDCl3, 400 MHz) δ 7.99 (s, 1H), 7.92˜7.98 (m, 2H), 7.65˜7.67 (m, 2H), 7.43 (d, J=7.6 Hz, 1H), 7.28˜7.32 (m, 2H), 7.15˜7.24 (m, 3H), 6.98˜7.03 (m, 1H), 6.05 (br s, 1H), 4.31 (t, J=6.4 Hz, 2H), 3.36 (s, 3H), 3.30 (t, J=6.4 Hz, 2H), 2.97 (d, J=4.8 Hz, 3H), 2.68 (s, 3H). MS (M+H)+: 613.


Example 32



embedded image


Step 1—Synthesis of 5-bromo-N-methyl-2-(4-methyl-2-oxopyridin-1 (2H)-yl)-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a mixture of 5-bromo-2-iodo-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (500 mg, 1.03 mmol), 4-methylpyridin-2-ol (168 mg, 1.54 mmol) and K2CO3 (283 mg, 2.05 mmol) in DMF (5 mL) was stirred at 80° C. for 10 hours and concentrated in vacuo to remove DMF. The reaction mixture was extracted with EtOAc, washed with brine, dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by column chromatography (PE:EtOAc=1:1) to give the product of 5-bromo-N-methyl-2-(4-methyl-2-oxopyridin-1 (2H)-yl)-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (400 mg, yield: 83%). 1H-NMR (CDCl3, 400 MHz) δ 8.32 (s, 1H), 7.85 (s, 1H), 7.67 (s, 1H), 7.22 (d, J=7.2 Hz, 1H), 6.52 (s, 1H), 6.28 (d, J=1.6 Hz, 1H), 3.32 (s, 3H), 3.06 (s, 3H), 2.89 (d, J=4.8 Hz, 3H), 2.31 (s, 3H). MS (M+H)+: 468/470.


Step 2—Synthesis of N-methyl-2-(4-methyl-2-oxopyridin-1 (2H)-yl)-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide



embedded image


To a mixture of 5-bromo-N-methyl-2-(4-methyl-2-oxopyridin-1 (2H)-yl)-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (400 mg, 0.85 mmol), bis(pinacolato)diboron (1.08 mg, 4.27 mmol) and KOAc (168 mg, 1.71 mmol) in 1,4-dioxane/H2O (5 mL/0.5 mL) was added Pd(dppf)Cl2 (10 mg) under N2. The reaction mixture was stirred at 120° C. for 5 hours and concentrated in vacuo to remove 1,4-dioxane. The reaction mixture was extracted with EtOAc, washed with brine, dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by column chromatography (PE:EA=1:1) to give the product of N-methyl-2-(4-methyl-2-oxopyridin-1 (2H)-yl)-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide (200 mg, yield: 45%). 1H-NMR (CDCl3, 400 MHz) δ 8.45 (s, 1H), 7.75 (d, J=4.4 Hz, 1H) 7.56 (s, 1H), 7.23 (s, 1H), 6.51 (s, 1H), 6.25 (d, J=7.2 Hz, 1H), 3.34 (s, 3H), 2.96 (s, 3H), 2.91 (d, J=4.8 Hz, 3H), 2.30 (s, 3H), 1.35 (s, 12H). MS (M+H)+: 516.


Step 3—Synthesis of 5-(11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-N-methyl-2-(4-methyl-2-oxopyridin-1 (2H)-yl)-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a mixture of N-methyl-2-(4-methyl-2-oxopyridin-1 (2H)-yl)-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide (100 mg, 0.19 mmol), 2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridine (63 mg, 0.23 mmol) and K2CO3 (54 mg, 0.39 mmol) in 1,4-dioxane/H2O (3 mL/0.1 mL) were added X-Phos (10 mg) and Pd2(dba)3 (10 mg) under N2. The reaction mixture was stirred at 80° C. for 5 hours and concentrated in vacuo to remove 1,4-dioxane. The reaction mixture was extracted with EtOAc, washed with brine, dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified by prep-HPLC to give the product (30 mg, yield: 24%). 1H-NMR (CDCl3, 400 MHz) δ 8.17 (s, 1H), 7.83˜7.85 (m, 1H), 7.66˜7.68 (m, 2H), 7.44 (d, J=8.0 Hz, 1H), 7.26˜7.18 (m, 2H), 7.14˜7.18 (m, 2H), 6.77˜6.81 (m, 1H), 6.53 (s, 1H), 6.29 (d, J=8.0 Hz, 1H), 4.34 (t, J=6.4 Hz, 2H), 3.40 (s, 3H), 3.31 (t, J=6.4 Hz, 2H), 2.90 (d, J=4.8 Hz, 3H), 2.61 (s, 3H), 2.32 (s, 3H). MS (M+H)+: 626.


Examples 33-34

Examples 33 and 34, depicted in the table below, were prepared in accordance with the method described in Example 32.


















MS


Example
Structure
NMR
(M + H)+







33


embedded image



1H-NMR (CDCl3, 400 MHz) δ 9.31 (s, 1H), 8.59 (s, 1H), 8.50 (s, 1H), 8.04 (s, 1H), 7.62 (s, 2H), 7.40 (d, J = 7.6 Hz, 1H), 7.09 (s, 3H), 6.75 (d, J = 5.2 Hz, 1H), 4.29 (d, J = 6.8 Hz, 2H), 3.37 (s, 3H), 3.26 (t, J = 6.8 Hz, 2H), 2.95 (d, J = 4.4 Hz, 3H), 2.53 (s, 3H).

653





34


embedded image



1H-NMR (CDCl3, 400 MHz) δ 8.17 (s, 1H), 7.67 (t, J = 3.6 Hz, 3H), 7.53~7.59 (m, 1H), 7.45 (d, J = 8.0 Hz, 1H), 7.40 (d, J = 5.6 Hz, 1H), 7.24~7.26 (m, 1H), 7.13~7.20 (m, 2H), 6.73~6.83 (m, 2H), 6.45 (t, J = 6.4 Hz, 1H), 4.35 (t, J = 6.4 Hz, 2H), 3.41 (s, 3H), 3.32 (t, J = 6.4 Hz, 2H), 2.91 (d, J = 4.8 Hz, 3H), 2.62 (s, 3H).

612









Example 35



embedded image


Step 1—Synthesis of methyl 5-bromo-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)benzofuran-2-carboxylate



embedded image


To a solution of 5-bromo-2-iodo-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (4.0 g, 8.21 mmol) in MeOH (10 mL) and DMSO (35 mL) was added Pd(PPh3)2Cl2 under Are. The suspension was degassed under vacuum and purged with CO 4 times. The mixture was stirred under CO (50 psi) at 50° C. for 16 h. Then 30 mL MeOH was added to the mixture. The resulting solid was filtered to give the product of methyl 5-bromo-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)benzofuran-2-carboxylate (2.90 g, yield: 84%). 1H-NMR (CDCl3, 400 MHz) δ 9.39 (br s, 1H), 8.94 (s, 1H), 7.74 (s, 1H), 4.08 (s, 3H), 3.34 (s, 3H), 3.08 (s, 3H), 3.04 (d, J=4.8 Hz, 3H). (M+H)+: 419/421


Step 2—Synthesis of 5-bromo-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)benzofuran-2-carboxylic acid



embedded image


To a suspension of methyl 5-bromo-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)benzofuran-2-carboxylate (2.90 g, 6.92 mmol) in 1,4-dioxane/H2O (30 mL/5 mL) was added LiOH.H2O (870 mg, 20.75 mmol). The mixture was stirred at room temperature overnight. Then it was concentrated in vacuo, diluted with water, acidized with HCl (aq. 2 M) and extracted with EtOAc. The organic layer was washed with brine, dried over Na2SO4 and concentrated give the product of 5-bromo-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)benzofuran-2-carboxylic acid (2.50 g, yield: 89%). It was used for the next step without further purification. 1H-NMR (DMSO-d6, 400 MHz) δ 9.16 (br s, 1H), 8.27 (s, 1H), 8.16 (s, 1H), 3.17 (s., 6H), 2.82 (d, J=4.4 Hz, 3H). (M+H)+: 405/407.


Step 3—Synthesis of 5-bromo-N2-methoxy-N2,N3-dimethyl-6-(N-methylmethylsulfonamido)benzofuran-2,3-dicarboxamide



embedded image


A mixture of 5-bromo-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)benzofuran-2-carboxylic acid (1 g, 2.47 mmol), HOBT (500 mg, 3.70 mmol) and EDCI (946 mg, 4.94 mmol) in DMF (20 mL) was stirred at room temperature under N2 for 2 h. Et3N (1.25 g, 12.34 mmol) and N,O-dimethylhydroxylamine hydrochloride (1.03 g, 12.34 mmol) were added, then the mixture was stirred for 18 h. The mixture was concentrated, diluted with water, extracted with EA, the organic layers were washed with Na2CO3 (a.q.), brine, dried over Na2SO4, concentrated. The residue was purified by column chromatography (DCM:EA=20:1) to get product 5-bromo-N2-methoxy-N2,N3-dimethyl-6-(N-methylmethylsulfonamido)benzofuran-2,3-dicarboxamide (0.68 g, yield: 62%). 1H-NMR (CDCl3, 400 MHz) δ 8.83 (s, 1H), 8.74 (s, 1H), 7.72 (s, 1H), 3.89 (s, 3H), 3.42 (s, 3H), 3.34 (s, 3H), 3.08 (s, 3H), 2.98 (d, J=4.8 Hz, 3H). MS (M+H)+: 448/450.


Step 4—Synthesis of ethyl 5-bromo-6-nitrobenzofuran-3-carboxylate



embedded image


To a mixture of 5-bromo-N2-methoxy-N2,N3-dimethyl-6-(N-methylmethylsulfonamido)benzofuran-2,3-dicarboxamide (1 g, 2.3 mmol) in THF (10 mL) was added DIBAL-H (11.5 mL, 11.5 mmol) at −78° C. dropwise. The mixture was stirred at −15° C. for 2 h. Then it was quenched with HCl (a.q.), extracted with CH2Cl2, dried over Na2SO4, concentrated. The residue was crude product of 5-bromo-2-formyl-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (700 mg, yield: 74%). 1H-NMR (CDCl3, 400 MHz) δ 10.07 (s, 1H), 9.14 (br s, 1H), 9.00 (s, 1H), 7.80 (s, 1H), 3.35 (s, 3H), 3.10 (s, 3H), 3.05 (d, J=4.4 Hz, 3H). MS (M+H)+: 389/391.


Step 5—Synthesis of 5-bromo-2-((hydroxyimino)methyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


A mixture of 5-bromo-2-formyl-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (700 mg, 1.8 mmol), hydroxylamine hydrochloride (280 mg, 4.05 mmol) and pyridine (284 mg, 3.6 mmol) in DMSO (5 mL) was stirred at room temperature for 2 h. The mixture was diluted with water, then filtered, the residue was crude product of 5-bromo-2-((hydroxyimino)methyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (650 mg, yield: 75%). 1H-NMR (DMSO-d6, 400 MHz) δ 12.26 (s, 1H), 8.46 (s, 1H), 8.41 (d, J=4.4 Hz, 1H), 8.18 (s, 1H), 8.06 (s, 1H), 3.20 (s, 3H), 3.19 (s, 3H), 2.83 (d, J=4.4 Hz, 3H). MS (M+H)+: 404/406.


Step 6—Synthesis of 5-bromo-N-hydroxy-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)benzofuran-2-carbimidoyl chloride



embedded image


A mixture of 5-bromo-2-((hydroxyimino)methyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (100 mg, 0.25 mmol) and NCS (40 mg, 0.30 mmol) in DMF (2 mL) was stirred at 60° C. for 18 h. The mixture was used for the next step without purification, 5-bromo-N-hydroxy-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)benzofuran-2-carbimidoyl chloride was obtained (90 mg, yield: 65%). 1H-NMR (DMSO-d6, 400 MHz) δ 13.33 (s, 1H), 8.65 (d, J=4 Hz, 1H), 8.13 (s, 1H), 8.05 (s, 1H), 3.20 (s, 6H), 2.80 (d, J=4.8 Hz, 3H). MS (M+H)+: 438/440.


Step 7—Synthesis of 5-bromo-2-(5-ethoxy-4-methyl-4,5-dihydroisoxazol-3-yl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


A mixture of 5-bromo-N-hydroxy-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)benzofuran-2-carbimidoyl chloride (400 mg, 0.91 mmol), 1-ethoxyprop-1-ene (392 mg, 4.56 mmol) and NaHCO3 (153 mg, 1.82 mmol) in DMF (10 mL) was stirred at room temperature for 16 h. It was concentrated, diluted with water, extracted with EtOAc, the organic layers were washed with brine, dried over Na2SO4, and concentrated. The residue was purified by column chromatography (PE:EA=2:1) to get product 5-bromo-2-(5-ethoxy-4-methyl-4,5-dihydroisoxazol-3-yl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (370 mg, yield: 83%). MS (M+H)+: 488/490.


Step 8—Synthesis of 5-bromo-N-methyl-2-(4-methylisoxazol-3-yl)-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


A mixture of 5-bromo-2-(5-ethoxy-4-methyl-4,5-dihydroisoxazol-3-yl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (50 mg, 0.10 mmol) in DCM/TFA (1 mL/2 mL) was stirred at reflux for 16 h. It was washed with water, brine, dried over Na2SO4, and concentrated. The residue was purified by prep-TLC (DCM:MeOH=20:1) to get 5-bromo-N-methyl-2-(4-methylisoxazol-3-yl)-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (40 mg, yield: 88%). 1H-NMR (CDCl3, 400 MHz) δ 9.14 (br s, 1H), 8.84 (s, 1H), 8.39 (s, 1H), 7.76 (s, 1H), 3.35 (s, 3H), 3.10 (s, 3H), 3.04 (d, J=4.8 Hz, 3H), 2.36 (s, 3H). MS (M+H)+: 442/444.


Step 9—Synthesis of N-methyl-2-(4-methylisoxazol-3-yl)-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide



embedded image


To a mixture of 5-bromo-N-methyl-2-(4-methylisoxazol-3-yl)-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (250 mg, 0.57 mmol), bis(pinacolato)diboron (719 mg, 2.83 mmol) and KOAc (169 mg, 1.7 mmol) in dioxane (5 mL) was added Pd(dppf)Cl2 (43 mg, 0.06 mmol) under N2. The mixture was stirred at 85° C. for 3 h, then it was cooled to room temperature, diluted with water, extracted with EA, dried over Na2SO4, and concentrated. The residue was purified by prep-TLC (DCM:MeOH=60:1) to get crude product of N-methyl-2-(4-methylisoxazol-3-yl)-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide (64 mg, yield: 20%). 1H-NMR (CDCl3, 400 MHz) δ 9.01 (br s, 1H), 8.97 (s, 1H), 8.36 (s, 1H), 7.76 (s, 1H), 3.36 (s, 3H), 3.04 (d, J=4.8 Hz, 3H), 2.99 (s, 3H), 2.36 (s, 3H), 1.36 (s, 12H). MS (M+H)+: 490.


Step 10—Synthesis of 5-(11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-N-methyl-2-(4-methylisoxazol-3-yl)-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a mixture of N-methyl-2-(4-methylisoxazol-3-yl)-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide (60 mg, 0.12 mmol), 2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridine (50 mg, 0.18 mmol) and K3PO4.3H2O (98 mg, 0.37 mmol) in dioxane/H2O (1.5 mL/0.5 mL) was added Pd2(dba)3 (8 mg, 0.008 mmol) and X-Phos (8 mg, 0.016 mmol) under N2. The mixture was stirred at 85° C. for 3 h. It was concentrated, diluted with water, extracted with EtOAc, and dried over Na2SO4. The residue was purified by prep-TLC (DCM:MeOH=60:1) to get the product (35 mg, yield: 48%). 1H-NMR (CDCl3, 400 MHz) δ 9.12 (d, J=3.6 Hz, 1H), 8.68 (s, 1H), 8.40 (s, 1H), 7.76 (s, 1H), 7.68 (d, J=7.6 Hz, 1H), 7.48 (d, J=7.6 Hz, 1H), 7.28˜7.26 (m, 1H), 7.21˜7.12 (m, 2H), 6.85˜6.75 (m, 1H), 4.36 (t, J=6.4 Hz, 2H), 3.46 (s, 3H), 3.33 (t, J=6.4 Hz, 2H), 3.03 (d, J=4.8 Hz, 3H), 2.60 (s, 3H), 2.40 (s, 3H). MS (M+H)+: 600.


Example 36



embedded image


Step 1—Synthesis of 5-bromo-N-methyl-6-(N-methylmethylsulfonamido)-2-(2-methylthiazol-5-yl)benzofuran-3-carboxamide



embedded image


To a degassed solution of compound 5-bromo-2-iodo-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (600 mg, 1.2 mmol), 2-methylthiazole (244 mg, 2.5 mmol) and Na2CO3 (261 mg, 2.5 mmol) in DMF (6 mL) was added Pd(dppf)Cl2 (50 mg) under N2, and the mixture was stirred at 80° C. for 12 h. After the solvent was removed, the residue was purified by column chromatography (DCM:EtOAc=2:1) to give the product of 5-bromo-N-methyl-6-(N-methylmethylsulfonamido)-2-(2-methylthiazol-5-yl)benzofuran-3-carboxamide (240 mg, yield: 42%). 1H-NMR (CDCl3, 400 MHz) δ 8.36 (s, 1H), 7.98˜8.01 (m, 1H), 7.68 (s, 1H), 6.06 (br s., 1H), 3.33 (s, 3H), 3.10 (s, 3H), 3.08 (d, J=5.2 Hz, 3H), 2.78 (s, 3H). MS (M+H)+: 458/460.


Step 2—Synthesis of N-methyl-6-(N-methylmethylsulfonamido)-2-(2-methylthiazol-5-yl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide



embedded image


To a degassed solution of 5-bromo-N-methyl-6-(N-methylmethylsulfonamido)-2-(2-methylthiazol-5-yl)benzofuran-3-carboxamide (240 mg, 0.52 mmol), KOAc (154 mg, 1.6 mmol) and dis(pinacolato)diboron (665 mg, 2.6 mmol) in 1,4-dioxane (5 mL) and H2O (0.1 mL) was added Pd(dppf)Cl2 (10 mg) under N2, and the mixture was stirred at 130° C. for 3 hours. After the solvent was removed, the residue was purified by prep-TLC (DCM:EtOAc=2:1) to give the product of N-methyl-6-(N-methylmethylsulfonamido)-2-(2-methylthiazol-5-yl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide (120 mg, yield: 45%). 1H-NMR (CDCl3, 400 MHz) δ 8.39 (s, 1H), 8.08 (s, 1H), 7.55 (s, 1H), 6.34 (br s., 1H), 3.36 (s, 3H), 3.09 (d, J=4.4 Hz, 3H), 2.95 (s, 3H), 2.75 (s, 3H), 1.38 (s, 12H). MS (M+H)+: 506.


Step 3—Synthesis of 5-(11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-N-methyl-6-(N-methylmethylsulfonamido)-2-(2-methylthiazol-5-yl)benzofuran-3-carboxamide



embedded image


To a degassed solution of N-methyl-6-(N-methylmethylsulfonamido)-2-(2-methylthiazol-5-yl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide (200 mg, 0.40 mmol), 2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridine (113 mg, 0.41 mmol), Na2CO3 (84 mg, 0.79 mmol), K2CO3 (109 mg, 0.79 mmol) in 1,4-dioxane (5 mL) and H2O (0.1 mL) were added Pd2(dba)3 (10 mg) and X-Phos (10 mg) under N2. Then the mixture was stirred at 100° C. for 2 hours. The reaction mixture was cooled to RT and filtered. The filtrate was washed with H2O and dried over Na2SO4. After concentrated, the residue was purified by prep-HPLC to give the product (30 mg, yield: 12%). 1H-NMR (CDCl3, 400 MHz) δ 8.41 (s, 1H), 7.89 (s, 1H), 7.62˜7.68 (m, 2H), 7.45 (d, J=7.6 Hz, 1H), 7.24 (s, 1H), 7.12˜7.20 (m, 2H), 6.74˜6.81 (m, 1H), 6.22 (d, J=4.0 Hz, 1H), 4.32 (t, J=6.4 Hz, 2H), 3.32 (s, 3H), 3.27˜3.31 (m, 2H), 3.02 (d, J=4.8 Hz, 3H), 2.75 (s, 3H), 2.74 (s, 3H). MS (M+H)+: 616.


Examples 37 and 38

Examples 37 and 38, depicted in the table below, were prepared in accordance with the method described in Example 36.


















MS


Example
Structure
NMR
(M + H)+







37


embedded image



1H-NMR (CDCl3, 400 MHz) δ 8.93 (s, 1H), 8.72 (s, 1H), 7.93 (s, 1H), 7.73~7.65 (m, 2H), 7.47 (d, J = 7.6 Hz, 1H), 7.29 (s, 1H), 7.22~7.11 (m, 2H), 6.79 (dd, J = 7.2, 9.2 Hz, 1H), 6.25 (br s, 1H), 4.34 (t, J = 6.5 Hz, 2H), 3.38~3.28 (m, 5H), 3.05 (d, J = 4.8 Hz, 3H), 2.77 (s, 3H).

602





38


embedded image



1H-NMR (CDCl3, 400 MHz) δ 8.41 (s, 1H), 7.89 (s, 1H), 7.62~7.66 (m, 2H), 7.43 (d, J = 7.6 Hz, 1H), 7.23 (s, 1H), 7.10~7.15 (m, 2H), 6.76 (dd, J = 7.6, 8.8 Hz, 1H), 6.17 (d, J = 3.6 Hz, 1H), 4.31 (t, J = 6.4 Hz, 2H), 3.31 (s, 3H), 3.26~3.30 (m, 2H), 3.05~3.10 (m, 2H), 3.01 (d, J = 4.8 Hz, 3H), 2.73 (s, 3H), 1.43 (t, J = 7.6 Hz, 3H).

630









Example 39



embedded image


Step 1—Synthesis of 6-chloro-4-(4-fluoro-1H-indol-2-yl)pyridin-3-yl trifluoromethanesulfonate



embedded image


To a solution of 6-chloro-4-(4-fluoro-1H-indol-2-yl)pyridin-3-ol (prepared using similar method described in Example 1, 500 mg, 1.9 mmol), DIPEA (490 mg, 3.8 mmol) and DMAP (5 mg, 0.02 mmol) in DCM (2 mL) was added Tf2O (1.07 g, 3.8 mmol) dropwise at 0° C. under nitrogen. The mixture was stirred at 25° C. for 5 hours. The mixture was diluted with H2O extracted with EtOAc, then the combined organic phase was washed with brine, dried over Na2SO4 and concentrated in vacuo to give the crude product. It was purified by column (PE:EA=5:1) to obtain the product of 6-chloro-4-(4-fluoro-1H-indol-2-yl)pyridin-3-yl trifluoromethanesulfonate (600 mg, yield: 80%). 1H-NMR (CDCl3, 400 MHz) δ 8.71 (s, 1H), 8.39 (s, 1H), 7.64 (s, 1H), 7.18˜7.20 (m, 3H), 6.77˜6.82 (m, 1H). MS (M+H)+: 395/397.


Step 2—Synthesis of 2-(2-chloro-5-vinylpyridin-4-yl)-4-fluoro-1H-indole



embedded image


To a solution of 6-chloro-4-(4-fluoro-1H-indol-2-yl)pyridin-3-yl trifluoromethanesulfonate (50 mg, 0.12 mmol), LiCl (8 mg, 0.19 mmol) in DMF (0.8 mL) was added tributyl(vinyl)stannane (80 mg, 0.25 mmol) dropwise at 25° C. under nitrogen. The mixture was stirred at 65° C. for 10 hours. The mixture was diluted with H2O, extracted with EtOAc, then the combined organic phase was washed with brine, dried over Na2SO4 and concentrated in vacuo to give the crude product. It was purified by column (PE:EA=5:1) to obtain the product of 2-(2-chloro-5-vinylpyridin-4-yl)-4-fluoro-1H-indole (150 mg, yield: 40%). 1H-NMR (CDCl3, 400 MHz) δ 8.45 (s, 1H), 7.41 (s, 1H), 7.15 (d, J=2.4 Hz, 1H), 6.88 (d, J=6.0 Hz, 1H), 6.79 (t, J=6.0 Hz, 1H), 6.35˜6.41 (m, 1H), 6.07˜6.10 (m, 1H), 5.77 (d, J=17.6 Hz, 1H), 5.49˜5.62 (m, 2H). MS (M+H)+: 273/275.


Step 3—Synthesis of 2-chloro-11-fluoro-5,6-dihydroindolo[2,1-a][2,6]naphthyridine



embedded image


A solution of 2-(2-chloro-5-vinylpyridin-4-yl)-4-fluoro-1H-indole (50 mg, 0.18 mmol), Bu4NCl (0.07 mL of 50% a.q. solution), DMSO (0.25 mL) and KOH (0.6 mL of 60% a.q. solution) was stirred at 100° C. for 3 hours. Then the mixture was diluted with H2O, extracted with EtOAc, then the combined organic phase was washed with brine, dried over Na2SO4 and concentrated in vacuo to give the crude product. It was purified by column (PE:EA=3:1) to obtain the product of 2-chloro-11-fluoro-5,6-dihydroindolo[2,1-a][2,6]naphthyridine (11 mg, yield: 20%). 1H-NMR (CDCl3, 400 MHz) δ 8.30 (s, 1H), 7.62 (s, 1H), 7.18˜7.21 (m, 2H), 7.12 (s, 1H), 6.81 (t, J=8.4 Hz, 1H), 4.29 (t, J=6.0 Hz, 2H), 3.20 (t, J=6.0 Hz, 2H). MS (M+H)+: 273/275.


Step 4—Synthesis of 5-(11-fluoro-5,6-dihydroindolo[2,1-a][2,6]naphthyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a solution of compound 2-chloro-11-fluoro-5,6-dihydroindolo[2,1-a][2,6]naphthyridine (70 mg, 0.26 mmol), 2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide (155 mg, 0.31 mmol) and K2CO3 (53 mg, 0.38 mmol) in 1,4-dioxane (2 mL) and water (0.05 mL) were added X-Phos (10 mg) and Pd2(dba)3 (10 mg) under nitrogen. The mixture was heated at 120° C. for 3 hours and filtered through the celite pad. The filtrate was extracted with EtOAc, then the combined organic phase was washed with brine, dried over Na2SO4 and concentrated. The crude product was purified by prep-HPLC to give the desired compound (50 mg, yield: 30%). 1H-NMR (CDCl3, 400 MHz) δ 8.54 (s, 1H), 7.91˜7.98 (m, 4H), 7.62 (s, 1H), 7.09˜7.16 (m, 5H), 6.74 (t, J=8.8 Hz, 1H), 5.95 (s, 1H), 4.29 (t, J=6.0 Hz, 2H), 3.23 (t, J=6.0 Hz, 2H), 3.17 (s, 3H), 2.95 (d, J=4.0 Hz, 3H), 2.83 (s, 3H). MS (M+H)+: 613.


Example 40

Example 40, depicted in the table below, was prepared in accordance with the method described in Example 39.















Compound


MS


ID
Structure
NMR
(M + H)+







40


embedded image



1H-NMR (CDCl3, 400 MHz) δ 8.96 (s, 1H), 8.68 (s, 1H), 8.53 (d, J = 3.6 Hz, 1H), 8.19 (s, 1H), 8.16 (s, 1H), 8.06 (s, 1H), 7.94 (s, 1H), 7.44 (d, J = 6.4 Hz, 2H), 7.25 (s, 1H), 7.19 (t, J = 4.8 Hz, 1H), 6.87 (t, J = 8.0 Hz, 1H), 4.38-4.43 (m, 2H), 3.18-3.52 (m, 2H), 3.22 (m, 3H), 3.05 (s, 3H), 2.81 (d, J = 4.0 Hz, 3H), 2.55 (s, 3H).

610









Examples 41 and 42



embedded image


41 (Enantiomer 1, peak 1 on SFC) 42 (Enantiomer 2, peak 2 on SFC)


Step 1—Synthesis of (Z)-2-(2-chloro-5-(prop-1-en-1-yl)pyridin-4-yl)-4-fluoro-1H-indole



embedded image


To a mixture of 6-chloro-4-(4-fluoro-1H-indol-2-yl)pyridin-3-yl trifluoromethanesulfonate (200 mg, 0.51 mmol), (Z)-tributyl (prop-1-en-1-yl) stannane (170 mg, 0.51 mmol) and anhydrous LiCl (70 mg, 1.65 mmol) in DMF (2 mL), Pd(PPh3)2Cl2 (30 mg, 0.04 mmol) was added under N2 protection. The reaction mixture was stirred at 100° C. for 10 h. Then it was filtered and concentrated in vacuo. The residue was purified by column chromatography (PE:EA=10:1) to give the product of (Z)-2-(2-chloro-5-(prop-1-en-1-yl)pyridin-4-yl)-4-fluoro-1H-indole (90 mg, yield: 61.9%). 1H-NMR (CDCl3, 400 MHz) δ 12.02 (br s, 1H), 8.33 (s, 1H), 7.87 (s, 1H), 7.31 (d, J=7.6 Hz, 1H), 7.16˜7.21 (m, 1H), 6.98 (d, J=1.2 Hz, 1H), 6.85 (dd, J=10.8, 7.6 Hz, 1H), 6.57 (dd, J=11.2, 1.6 Hz, 1H), 6.06˜6.16 (m, 1H), 1.78 (dd, J=6.8, 1.6 Hz, 3H). MS (M+H)+: 287.


Step 2—Synthesis of 2-chloro-11-fluoro-6-methyl-5,6-dihydroindolo[2,1-a][2,6]naphthyridine



embedded image


A mixture of (Z)-2-(2-chloro-5-(prop-1-en-1-yl)pyridin-4-yl)-4-fluoro-1H-indole (90 mg, 0.31 mmol), DMSO (0.5 mL), TBAC (0.1 mL of 50% a.q. solution) and KOH (1.0 mL of 60% aq. solution) was stirred under N2 at 100° C. for 5 h. After cooling and extraction with EtOAc, the organic layer was washed with water, brine, dried over Na2SO4, and concentrated.


The residue was purified by prep-TLC (PE:EA=5:1) to give the product of 2-chloro-11-fluoro-6-methyl-5,6-dihydroindolo[2,1-a][2,6]naphthyridine (30 mg, yield: 33.3%). 1H-NMR (CDCl3, 400 MHz) δ 8.31 (s, 1H), 7.62 (s, 1H), 7.14˜7.22 (m, 2H), 7.11 (s, 1H), 6.81˜6.82 (m, 1H), 4.92˜4.98 (m, 1H), 3.35˜3.41 (m, 1H), 2.99 (d, J=15.2 Hz, 1H), 1.24 (d, J=6.4 Hz, 3H). MS (M+H)+: 287.


Step 3—Synthesis of (R or S)-5-(11-fluoro-6-methyl-5,6-dihydroindolo[2,1-a][2,6]naphthyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide and (S or R)-5-(11-fluoro-6-methyl-5,6-dihydroindolo[2,1-a][2,6]naphthyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a degassed solution of 2-chloro-11-fluoro-6-methyl-5,6-dihydroindolo[2,1-a][2,6]naphthyridine (80 mg, 0.28 mmol) and (2-(4-fluorophenyl)-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)benzofuran-5-yl)boronic acid (130 mg, 0.26 mmol) in 1,4-dioxane (5 mL), Pd2(dba)3 (10 mg), X-Phos (10 mg) and K3PO4 (200 mg, 0.75 mmol) were added under N2. The mixture was heated to 80° C. for 2 hours. The reaction mixture was cooled to RT, filtered and washed with EtOAc. The filtrate was washed with H2O, brine, dried over Na2SO4. After being concentrated, the residue was purified by prep-HPLC and SFC to give two single enantiomers.


Compound 41 (enantiomer 1, peak 1 on SFC, AS-H_S3_5_403ML8MIN15CM, HPLC_RT=4.47 min) (40 mg, yield: 24.7%). 1H-NMR (CDCl3, 400 MHz) δ 8.59 (s, 1H), 8.05 (s, 1H), 7.92˜8.02 (m, 3H), 7.67 (s, 1H), 7.14˜7.24 (m, 5H), 6.78˜6.82 (m, 1H), 6.13 (br s, 1H), 4.99 (q, J=6.4 Hz, 1H), 3.49 (dd, J=15.6, 6.4 Hz, 1H), 3.23 (s, 3H), 2.96˜3.08 (m, 4H), 2.85 (s, 3H), 1.28 (d, J=6.4 Hz, 3H). MS (M+H)+: 628.


Compound 42 (enantiomer 2, peak 2 on SFC, AS-H_S3_5_403ML8MIN15CM, HPLC_RT=5.24 min) (40 mg, yield: 24.7%). 1H-NMR (CDCl3, 400 MHz) δ 8.59 (s, 1H), 8.05 (s, 1H), 7.92˜8.02 (m, 3H), 7.67 (s, 1H), 7.14˜7.24 (m, 5H), 6.78˜6.82 (m, 1H), 6.13 (br s, 1H), 4.99 (q, J=6.4 Hz, 1H), 3.49 (dd, J=15.6, 6.4 Hz, 1H), 3.23 (s, 3H), 2.96˜3.08 (m, 4H), 2.85 (s, 3H), 1.28 (d, J=6.4 Hz, 3H). MS (M+H)+: 628.


Example 43



embedded image


Step 1—Synthesis of 5-bromo-3-(4-fluoro-1H-indol-2-yl)pyridin-2-ol



embedded image


A mixture of 5-bromo-3-iodopyridin-2-ol (5 g, 16.7 mmol), (4-fluoro-1H-indol-2-yl)boronic acid (4.48 g, 25.0 mmol), sodium carbonate (3.53 g, 33.4 mmol) and Pd(dppf)Cl2 (200 mg) in 1,4-dioxane (50 mL) was stirred at 100° C. for 16 hours under nitrogen atmosphere. The mixture was filtered through celite and concentrated, and the residue was diluted with water and EA. After extracted with EA, the combined organic phase was washed with brine, dried over sodium sulfate and concentrated to afford crude 5-bromo-3-(4-fluoro-1H-indol-2-yl)pyridin-2-ol (3.5 g) through the column chromatography (DCM:EA=5:1). Finally the pure 5-bromo-3-(4-fluoro-1H-indol-2-yl)pyridin-2-ol (2.7 g, yield: 52%) was obtained by recrystallized from DCM:MeOH (10:1). 1H-NMR (DMSO-d6, 400 MHz) δ 12.36 (br s, 1H), 11.79 (s, 1H), 8.25 (s, 1H), 7.70 (s, 1H), 7.39 (s, 1H), 7.30 (d, J=8.00 Hz, 1H), 7.04˜7.07 (m, 1H), 6.74˜6.78 (m, 1H). MS (M+H)+: 307/309.


Step 2—Synthesis of 5-bromo-3-(4-fluoro-1H-indol-2-yl)pyridin-2-yl trifluoromethanesulfonate



embedded image


To a mixture of 5-bromo-3-(4-fluoro-1H-indol-2-yl)pyridin-2-ol (1.0 g, 3.26 mmol), DIPEA (1.26 g, 9.77 mmol) and DMAP (80 mg, 0.65 mmol) in 15 mL of DCM was added Tf2O (1.84 g, 6.51 mmol) at 0 dropwise. After stirring at room temperature for 3 hours, the mixture was suspended in water and extracted with DCM. Then the combined organic phase was washed with brine, dried over sodium sulfate and concentrated. After column chromatography (PE:EA=20:1), 5-bromo-3-(4-fluoro-1H-indol-2-yl)pyridin-2-yl trifluoromethanesulfonate (1.0 g, yield: 52%) was obtained. 1H-NMR (DMSO-d6, 400 MHz) δ 12.28 (s, 1H), 8.81 (s, 1H), 8.60 (s, 1H), 7.32 (d, J=8.40 Hz, 1H), 7.18˜7.22 (m, 1H), 7.05 (s, 1H), 6.84˜6.87 (m, 1H). MS (M+H)+: 439/441.


Step 3—Synthesis of 2-(5-bromo-2-vinylpyridin-3-yl)-4-fluoro-1H-indole



embedded image


A mixture of 5-bromo-3-(4-fluoro-1H-indol-2-yl)pyridin-2-yl trifluoromethanesulfonate (700 mg, 1.59 mmol), tributyl(vinyl)stannane (556 mg, 1.75 mmol), LiCl (202 mg, 4.78 mmol) and Pd(PPh3)2Cl2 (100 mg) in DMF (20 mL) was stirred at 60° C. for 3 hours under nitrogen atmosphere. The mixture was filtered through celite and concentrated in vacuo to remove DMF, and then the residue was suspended in water/EA. After extracted with EA, the combined organic phase was washed with brine, dried over sodium sulfate and concentrated to afford 2-(5-bromo-2-vinylpyridin-3-yl)-4-fluoro-1H-indole (400 mg, yield: 79%) through the column chromatography (PE:EA=20:1). 1H-NMR (CDCl3, 400 MHz) δ 8.64 (s, 1H), 8.46 (s, 1H), 7.93 (s, 1H), 7.14˜7.22 (m, 2H), 7.00˜7.07 (m, 1H), 6.82˜6.86 (m, 1H), 6.76 (s, 1H), 6.43˜6.47 (m, 1H), 5.54˜5.58 (m, 1H). MS (M+H)+: 317/319.


Step 4—Synthesis of 2-(5-bromo-2-(2-chloroethyl)pyridin-3-yl)-4-fluoro-1H-indole



embedded image


A solution of 2-(5-bromo-2-vinylpyridin-3-yl)-4-fluoro-1H-indole (800 mg, 2.52 mmol) in 20 mL of HCl/1,4-dioxane (4 mol/L) was heated at 80° C. for 2 hours and then the mixture was concentrated in vacuo to give the crude 2-(5-bromo-2-(2-chloroethyl)pyridin-3-yl)-4-fluoro-1H-indole (800 mg, yield: 90%) which was used directly in the next step without further purification. 1H-NMR (CDCl3, 400 MHz) δ 8.76 (s, 1H), 8.66 (s, 1H), 7.93 (s, 1H), 7.14˜7.22 (m, 2H), 6.73˜6.86 (m, 1H), 6.72 (s, 1H), 4.07˜4.10 (m, 2H), 3.35˜3.36 (m, 2H). MS (M+H)+: 353/355.


Step 5—Synthesis of 2-bromo-11-fluoro-5,6-dihydroindolo[2,1-f][1,6]naphthyridine



embedded image


A solution of 2-(5-bromo-2-(2-chloroethyl)pyridin-3-yl)-4-fluoro-1H-indole (800 mg, 2.26 mmol) and cesium carbonate (1.47 g, 4.52 mmol) in 25 mL of DMF was heated at 80° C. for 1 hour. The mixture was concentrated in vacuo to remove DMF, and then the residue was suspended in water and EA. After extraction with EA, the combined organic phase was washed with brine, dried over sodium sulfate and concentrated to afford crude 2-bromo-11-fluoro-5,6-dihydroindolo[2,1-f][1,6]naphthyridine (600 mg, yield: 83.6%) which was used directly in the next step without further purification. 1H-NMR (CDCl3, 400 MHz) δ 8.48 (s, 1H), 8.11 (s, 1H), 7.11˜7.17 (m, 2H), 6.97 (s, 1H), 6.78˜6.82 (m, 1H), 4.34˜4.37 (m, 2H), 3.34˜3.37 (m, 2H). MS (M+H)+: 317/319.


Step 6—Synthesis of 5-(11-fluoro-5,6-dihydroindolo[2,1-f][1,6]naphthyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


A mixture of 2-bromo-11-fluoro-5,6-dihydroindolo[2,1-f][1,6]naphthyridine (50 mg, 0.16 mmol), 2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide (87 mg, 0.17 mmol), potassium carbonate (43 mg, 0.32 mmol) and Pd(dtbpf)Cl2 (10 mg) in 1,4-dioxane/water (1/0.1 mL) was stirred at 100° C. for 6 hours under nitrogen atmosphere. The mixture was filtered through Celite pad and concentrated, and then the residue was diluted with water and EA. After extraction with EA, the combined organic phase was washed with brine, dried over sodium sulfate and concentrated to afford the desired product (40 mg, yield: 41.7%) through the prep-HPLC. 1H-NMR (CDCl3, 400 MHz) δ 8.50 (s, 1H), 8.19 (s, 1H), 7.93˜7.96 (m, 2H), 7.90 (s, 1H), 7.65 (s, 1H), 7.19˜7.24 (m, 2H), 7.15˜7.17 (m, 2H), 7.02 (s, 1H), 6.77˜6.82 (m, 1H), 5.88 (br s, 1H), 4.41˜4.44 (m, 2H), 3.45˜3.49 (m, 2H), 3.18 (s, 3H), 2.99 (d, J=4.80 Hz, 3H), 2.84 (s, 3H). MS (M+H)+: 613.


Example 44



embedded image


Step 1—Synthesis of 5-(6-(4-fluoro-1H-indol-2-yl)-5-hydroxypyrazin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a degassed solution of 5-bromo-3-(4-fluoro-1H-indol-2-yl)pyrazin-2-ol (prepared using similar method described in Example 1, 1.23 g, 3.98 mmol), 2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide (2 g, 3.98 mmol) and K3PO4.3H2O (2.12 g, 7.96 mmol) in IPA (20 mL) and H2O (2 mL) was added Pd2(dba)3 (50 mg) and X-Phos (50 mg) under nitrogen atmosphere. The reaction mixture was stirred at 100° C. for 16 h, concentrated in vacuo to remove IPA. The residue was diluted with H2O and extracted with DCM. Then the combined organic layers were washed with brine (100 mL), dried over Na2SO4, filtered, concentrated and purified by column chromatography (PE:EA=1:2) to give 5-(6-(4-fluoro-1H-indol-2-yl)-5-hydroxypyrazin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (1 g, yield: 41%). 1H-NMR (CDCl3, 400 MHz) δ 8.52 (d, J=4.0 Hz, 1H), 8.06 (s, 1H), 8.02 (s, 1H), 7.71 (s, 1H), 7.66 (s, 1H), 7.42 (t, J=8.8 Hz, 2H), 7.33˜7.36 (m, 2H), 7.11˜7.18 (m, 2H), 6.80 (dd, J1=10.4 Hz, J2=8.0 Hz, 2H), 3.27 (s, 3H), 3.10 (s, 3H), 2.85 (d, J=4.8 Hz, 3H). MS (M+H)+: 604.


Step 2—Synthesis of 3-(4-fluoro-1H-indol-2-yl)-5-(2-(4-fluorophenyl)-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)benzofuran-5-yl)pyrazin-2-yl trifluoromethanesulfonate



embedded image


To a solution of 5-(6-(4-fluoro-1H-indol-2-yl)-5-hydroxypyrazin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (400 mg, 0.68 mmol) and N-phenyl-O-((trifluoromethyl)sulfonyl)-N-(((trifluoromethyl)sulfonyl)oxy) hydroxylamine (400 mg, 1.03 mmol) in THF (10 mL) was added Et3N (276 mg, 2.73 mmol) under nitrogen atmosphere. The reaction mixture was stirred at room temperature overnight, diluted with H2O and extracted with DCM. The combined organic layers were washed with brine (100 mL), dried over Na2SO4, filtered, concentrated and purified by column chromatography (PE:EA=1:1) to give 3-(4-fluoro-1H-indol-2-yl)-5-(2-(4-fluorophenyl)-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)benzofuran-5-yl)pyrazin-2-yl trifluoromethanesulfonate (150 mg, yield: 36%). 1H-NMR (CDCl3, 400 MHz) δ 8.40 (s, 1H), 8.17 (s, 1H), 7.89˜7.93 (m, 2H), 7.64 (s, 1H), 7.48 (s, 1H), 7.33˜7.36 (m, 2H), 7.13˜7.21 (m, 3H), 6.78 (dd, J=8.8 Hz, 6.8 Hz, 1H), 5.87 (br s, 1H), 3.19 (s, 3H), 3.02 (s, 3H), 2.98 (d, J=4.8 Hz, 3H). MS (M+H)+: 736.


Step 3—Synthesis of 5-(6-(4-fluoro-1H-indol-2-yl)-5-vinylpyrazin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a mixture of 3-(4-fluoro-1H-indol-2-yl)-5-(2-(4-fluorophenyl)-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)benzofuran-5-yl)pyrazin-2-yl trifluoromethanesulfonate (30 mg, 0.04 mmol), tributyl(vinyl)stannane (16 mg, 0.05 mmol) and LiCl (5 mg, 0.12 mmol) in DMF (1 mL) was added Pd(PPh3)2Cl2 (5 mg) under nitrogen atmosphere. The reaction mixture was stirred at 60° C. for 1 h, concentrated and purified by prep-TLC (PE:EA=1:1) to give 5-(6-(4-fluoro-1H-indol-2-yl)-5-vinylpyrazin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (15 mg, yield: 60%). 1H-NMR (CDCl3, 400 MHz) δ 8.66 (s, 1H), 8.10 (s, 1H), 7.86˜7.89 (m, 2H), 7.57˜7.61 (m, 2H), 7.38˜7.44 (m, 3H), 7.04˜7.19 (m, 3H), 6.71 (t, J=8.0 Hz, 1H), 6.50˜6.54 (m, 1H), 5.89 (br s, 1H), 5.71˜5.74 (m, 1H), 3.11 (s, 3H), 2.91 (s, 6H). MS (M+H)+: 614.


Step 4—Synthesis of 5-(11-fluoro-5,6-dihydropyrazino[2′,3′:3,4]pyrido[1,2-a]indol-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


A solution of 5-(6-(4-fluoro-1H-indol-2-yl)-5-vinylpyrazin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (50 mg, 0.08 mmol) in HCl/1,4-dioxane (1 mL) was stirred at 80° C. for 1 h under nitrogen atmosphere. The reaction mixture was concentrated; then the residue was used to the next step reaction without purification. To the residue in DMF, Cs2CO3 was added (53 mg, 0.16 mmol), the reaction mixture was stirred at 80° C. for 1 h under nitrogen atmosphere, concentrated and purified by prep-TLC (PE:EA=1:2) to give the desired product (10 mg, yield: 20%). 1H-NMR (CDCl3, 400 MHz) δ 8.56 (s, 1H), 8.03 (s, 1H), 7.90 (dd, J=9.2 Hz, 5.2 Hz, 2H), 7.64 (s, 1H), 7.29 (s, 1H), 7.09˜7.17 (m, 4H), 6.74˜6.78 (m, 1H), 5.85 (br s, 1H), 4.41 (t, J=6.8 Hz, 2H), 3.47 (t, J=6.8 Hz, 2H), 3.34 (s, 3H), 2.94 (d, J=4.8 Hz, 3H), 2.73 (s, 3H). MS (M+H)+: 614.


Example 45



embedded image


Step 1—Synthesis of (2-(4-fluorophenyl)-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido) furo[2,3-b]pyridin-5-yl)boronic acid



embedded image


To a degassed solution of 2-(4-fluorophenyl)-5-iodo-N-methyl-6-(N-methylmethylsulfonamido)furo[2,3-b]pyridine-3-carboxamide (prepared according to International Patent Application Publication No. WO2011131709, 3.0 g, 6 mmol), hypodiboric acid (1.08 g, 1.2 mmol) and KOAc (1.8 g, 1.8 mmol) in EtOH (60 mL) was added Pd(OAc)2 (50 mg) and Ad2n-BuP (50 mg) under nitrogen atmosphere. The reaction mixture was stirred at 80° C. for 5 h, concentrated in vacuo to remove EtOH. The residue was purified by column chromatography (EA:DCM=1:1˜DCM:MeOH=5:1) to afford the desired product of (2-(4-fluorophenyl)-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)furo[2,3-b]pyridin-5-yl)boronic acid (1 g, yield: 40%). 1H-NMR (DMSO-d6, 400 MHz) δ 8.53 (d, J=4.4 Hz, 1H), 8.24 (s, 1H), 8.00˜8.04 (m, 3H), 7.40 (t, J=8.8 Hz, 2H), 4.08 (br s, 1H), 3.17 (s, 3H), 3.06 (s, 3H), 2.85 (d, J=4.8 Hz, 3H). MS (M+H)+: 422.


Step 2—Synthesis of 5-(11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)furo[2,3-b]pyridine-3-carboxamide



embedded image


To a degassed solution of (2-(4-fluorophenyl)-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)furo[2,3-b]pyridin-5-yl)boronic acid (100 mg, 0.24 mmol), 2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridine (60 mg, 0.21 mmol) and K2CO3 (98 mg, 0.72 mmol) in 1,4-dioxane (3 mL) and H2O (5 drops) was added Pd2(dba)3 (10 mg) and X-Phos (10 mg) under nitrogen atmosphere. The reaction mixture was stirred at 100° C. for 2 h, concentrated in vacuo to remove 1,4-dioxane. The residue was diluted with H2O and extracted with DCM. The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered, concentrated and purified to give the desired product (80 mg, yield: 55%) through the prep-HPLC. 1H-NMR (CDCl3, 400 MHz) δ 8.60 (s, 1H), 8.00˜8.04 (m, 2H), 7.76 (d, J=8.0 Hz, 1H), 7.67 (d, J=8.0 Hz, 1H), 7.35 (s, 1H), 7.21 (t, J=8.8 Hz, 2H), 7.14˜7.16 (m, 2H), 6.75˜6.80 (m, 1H), 5.97 (br s, 1H), 4.33 (t, J=6.4 Hz, 2H), 3.25˜3.32 (m, 5H), 3.18 (s, 3H), 3.00 (d, J=4.8 Hz, 3H). MS (M+H)+: 614.


Example 46



embedded image


Step 1—Synthesis of 4-(benzyloxy)-3-nitropyridine



embedded image


4-chloro-3-nitropyridine (30 g, 0.19 mol) was slowly added to a suspension of phenylmethanol (30 mL, 0.29 mol), K2CO3 (26.2 g, 0.19 mol and KOH (42.5 g, 0.76 mmol) in toluene (2 L) at 0° C. A catalytic amount of tris[2-(2-methoxyethoxyl)ethyl]amine was added to the reaction mixture, which was then stirred at room temperature for 2 hours. After filtration, the resulting filtrate was concentrated to give the crude product, which was crystallized from DCM/PE (1:5) to afford 4-(benzyloxy)-3-nitropyridine (27.3 g, yield: 62.7%). 1H-NMR (CDCl3, 400 MHz) δ 9.00 (s, 1H), 8.56 (d, J=6.0 Hz, 1H), 7.34˜7.43 (m, 5H), 7.03 (d, J=6.0 Hz, 1H), 5.29 (s, 2H). MS (M+H)+: 231.


Step 2—Synthesis of 4-(benzyloxy)pyridin-3-amine



embedded image


To a solution of 4-(benzyloxy)-3-nitropyridine (42.0 g, 0.18 mol), Fe (30.6 g, 0.55 mol) and NH4Cl (48.8 g, 0.91 mol) in MeOH:THF:H2O=(2:2:1, 750 mL) was stirred at reflux for 3 h. After being filtered and concentrated in vacuum, the residue was purified by column chromatography (DCM:MeOH=10:1) to give to give the pure 4-(benzyloxy)pyridin-3-amine (29.5 g, yield: 80.7%). 1H-NMR (CDCl3, 400 MHz) δ 8.25 (s, 1H), 7.88 (s, 1H), 7.20˜7.65 (m, 5H), 6.89 (s, 1H), 5.18 (s, 2H). MS (M+H)+: 201.


Step 3—Synthesis of N-(4-(benzyloxy)pyridin-3-yl)methanesulfonamide



embedded image


To a solution of 4-(benzyloxy)pyridin-3-amine (40 g, 0.2 mol) and Et3N (60.6 g, 0.6 mol) in DCM (600 mL), MsCl (45.8 g, 0.4 mol) was added dropwise at 0° C. The mixture was allowed to warm up to room temperature and stirred overnight. The reaction mixture was quenched with NaHCO3 and extracted with DCM. The organic phase was washed with brine, dried over Na2SO4, and concentrated. The residue was dissolved in THF:H2O=5:1 (500 mL) and LiOH.H2O (16.8 g, 0.4 mmol) was added. The mixture was stirred for 4 hours at room temperature. EtOAc (300 mL) was added, and the organic phase washed with NaHCO3 (sat., a.q.), brine, dried over Na2SO4, filtrated and concentrated to give pure N-(4-(benzyloxy)pyridin-3-yl)methanesulfonamide (30.0 g, yield: 54.0%). 1H-NMR (CDCl3, 400 MHz) δ 8.67 (s, 1H), 8.34 (d, J=5.6 Hz, 1H), 7.35˜7.42 (m, 5H), 6.91 (d, J=5.6 Hz, 1H), 6.61 (br, 1H), 5.15 (s, 2H), 2.96 (s, 3H). MS (M+H)+: 279.


Step 4—Synthesis of N-(4-(benzyloxy)pyridin-3-yl)-N-methylmethanesulfonamide



embedded image


To a suspension of N-(4-(benzyloxy)pyridin-3-yl)methanesulfonamide (30 g, 0.11 mol) and K2CO3 (14.9 g, 0.11 mol) in DMF (300 mL) was added dropwise CH3I (15.3 g, 0.11 mol) at 0° C. under N2, and then the mixture was stirred for 1 hour at this temperature. H2O was added to the mixture and extracted with dichloromethane. The combined organic layer was washed with H2O, brine, dried over Na2SO4, filtrated and concentrated in vacuo to give N-(4-(benzyloxy)pyridin-3-yl)-N-methylmethanesulfonamide (15 g, yield: 50%), which was used for the next step without further purification. 1H-NMR (CDCl3, 400 MHz) δ 8.46 (s, 1H), 8.41 (d, J=6.0 Hz, 1H), 7.37˜7.39 (m, 5H), 6.94 (d, J=6.0 Hz, 1H), 5.15 (s, 2H), 3.22 (s, 3H), 2.79 (s, 3H). MS (M+H)+: 293.


Step 5—Synthesis of methyl 5-(benzyloxy)-2-(4-fluorophenyl)-6-(N-methylmethylsulfonamido)pyrazolo[1,5-a]pyridine-3-carboxylate



embedded image


To a solution of N-(4-(benzyloxy)pyridin-3-yl)methanesulfonamide (9.0 g, 30.0 mmol) in DCM (100 mL) was added (O-(mesitylsulfonyl)hydroxylamine) (20 g, 92 mmol) in DCM (20 mL) dropwise quickly at 0° C. under N2, and stirred for 2 h at room temperature. After concentrated and the residue was dissolved in MeOH, and re-concentrated again. The residue was dissolved in DMF (20 mL), and then added slowly dropwise to a mixture of methyl 3-(4-fluorophenyl)propiolate (6 g, 34 mmol) and K2CO3 (21 g, 154 mmol) in DMF (100 mL) at 0° C. under N2. The mixture was stirred overnight at room temperature. The reaction mixture was then concentrated in vacuo and the resulting residue was suspended in H2O and extracted with DCM. The organic layer was washed with H2O, brine and concentrated in vacuo. The residue obtained was purified using flash column chromatography on silica gel (eluted with PE/EtOAc from 5/1 to 3/1) to give methyl 5-(benzyloxy)-2-(4-fluorophenyl)-6-(N-methylmethylsulfonamido)pyrazolo[1,5-a]pyridine-3-carboxylate (3.5 g, yield: 24%). 1H-NMR (CDCl3, 400 MHz) δ 8.54 (s, 1H), 7.70˜7.74 (m, 2H), 7.68 (s, 1H), 7.42˜7.44 (m, 5H), 7.13 (t, J=8.8 Hz, 2H), 5.24 (s, 2H), 3.80 (s, 3H), 3.29 (s, 3H), 2.81 (s, 3H). MS (M+H)+: 484.


Step 6—Synthesis of 5-(benzyloxy)-2-(4-fluorophenyl)-6-(N-methylmethylsulfonamido) pyrazolo[1,5-a]pyridine-3-carboxylic acid



embedded image


To a solution of methyl 5-(benzyloxy)-2-(4-fluorophenyl)-6-(N-methylmethylsulfonamido)pyrazolo[1,5-a]pyridine-3-carboxylate (3 g, 6.2 mmol) in dioxane/H2O (1/1, 60 mL) was added LiOH.H2O (781 mg, 19 mmol), and the mixture was heated to reflux for 3 hours, and concentrated in vacuo. The residue obtained was dissolved in H2O, 1 N HCl was added until pH reached 3, and the resulting precipitate was collected by filtration, washed with water, dried to afford 5-(benzyloxy)-2-(4-fluorophenyl)-6-(N-methylmethylsulfonamido)pyrazolo[1,5-a]pyridine-3-carboxylic acid (2.8 g, yield: 96%). 1H-NMR (DMSO-d6, 400 MHz) δ 12.4 (s, 1H), 8.97 (s, 1H), 7.74˜7.77 (m, 2H), 7.59 (s, 1H), 7.54 (d, J=7.2 Hz, 2H), 7.29˜7.43 (m, 3H), 7.26 (t, J=8.8 Hz, 2H), 5.32 (s, 2H), 3.54 (s, 3H), 3.20 (s, 3H), 3.00 (s, 3H). MS (M+H)+: 470.


Step 7—Synthesis of 5-(benzyloxy)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido) pyrazolo[1,5-a]pyridine-3-carboxamide



embedded image


5-(benzyloxy)-2-(4-fluorophenyl)-6-(N-methylmethylsulfonamido)pyrazolo[1,5-a]pyridine-3-carboxylic acid (3 g, 6.4 mmol), HOBT (1.3 g, 9.6 mmol) and EDCI (2.5 g, 12.8 mmol) in DMF (50 mL) was allowed to stir at room temperature. After 30 minutes, CH3NH2.HCl (857 mg, 12.8 mmol) and Et3N (1.3 g, 12.8 mmol) were added to the mixture, and the mixture was allowed to stir overnight at room temperature. After the solvent was removed, H2O and NaHCO3 (aq.) were added and the mixture was stirred at room temperature for 1 hour. After filtration, the cake was washed with H2O and dried to give pure 5-(benzyloxy)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)pyrazolo[1,5-a]pyridine-3-carboxamide (3 g, yield: 97%). 1H-NMR (CDCl3, 400 MHz) δ 8.43 (s, 1H), 7.86 (s, 1H), 7.56˜7.59 (m, 2H), 7.35˜7.44 (m, 5H), 7.16 (t, J=8.8 Hz, 2H), 5.41 (br s, 1H), 5.15 (s, 2H), 3.22 (s, 3H), 2.77 (d, J=4.8 Hz, 3H), 2.71 (s, 3H). MS (M+H)+: 483.


Step 8—Synthesis of 2-(4-fluorophenyl)-5-hydroxy-N-methyl-6-(N-methylmethylsulfonamido) pyrazolo[1,5-a]pyridine-3-carboxamide



embedded image


A degassed solution of 5-(benzyloxy)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)pyrazolo[1,5-a]pyridine-3-carboxamide (3.0 g, 6.2 mmol) in THF (50 mL) was charged with 10% palladium on carbon (0.3 g). The reaction mixture was hydrogenated at room temperature under H2 balloon for 4 hours. The reaction mixture was filtered, and washed with DMF twice. The filtrate concentrated in vacuo and dried to afford pure 2-(4-fluorophenyl)-5-hydroxy-N-methyl-6-(N-methylmethylsulfonamido) pyrazolo[1,5-a]pyridine-3-carboxamide (2.3 g, yield: 94%). MS (M+H)+: 393.


Step 9—Synthesis of 2-(4-fluorophenyl)-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido) pyrazolo[1,5-a]pyridin-5-yl trifluoromethanesulfonate



embedded image


To a solution of 2-(4-fluorophenyl)-5-hydroxy-N-methyl-6-(N-methylmethylsulfonamido)pyrazolo[1,5-a]pyridine-3-carboxamide (2.3 g, 5.8 mmol) and 1,1,1-trifluoro-N-phenyl-N-(trifluoromethylsulfonyl)methanesulfonamide (3.1 g, 8.8 mmol) in DCM (70 mL) was added dropwise Et3N (1.2 g, 11.7 mmol) under N2, and the mixture was stirred overnight at room temperature. After dilution with H2O and extraction with DCM, the organic phase was washed with brine, dried over Na2SO4, and concentrated. The residue obtained was purified using flash column chromatography on silica gel (eluted with PE/EtOAc from 10/1 to 5/1) to give 2-(4-fluorophenyl)-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)pyrazolo[1,5-a]pyridin-5-yl trifluoromethanesulfonate (1.5 g, yield: 50%). 1H-NMR (CDCl3, 400 MHz) δ 8.65 (s, 1H), 8.35 (s, 1H), 7.61˜7.65 (m, 2H), 7.21˜7.26 (m, 2H), 5.55 (br s, 1H), 3.34 (s, 3H), 3.14 (s, 3H), 2.83 (d, J=4.8 Hz, 3H). MS (M+H)+: 525.


Step 10—Synthesis of 2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazolo[1,5-a]pyridine-3-carboxamide



embedded image


To a N2 degassed solution of 2-(4-fluorophenyl)-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)pyrazolo[1,5-a]pyridin-5-yltrifluoromethanesulfonate (300 mg, 0.6 mmol), KOAc (168 mg, 1.8 mmol) and bis(pinacolato)diboron (726 mg, 3 mmol) in dioxane (10 mL) was added Pd(dppf)Cl2 under N2, and the mixture was stirred at room temperature for 1 hour, and then put it into 120° C. oil-batch and stirred for 2 hours. After the solvent was removed, the residue was purified by column chromatography (PE:EA=10:1 to 2:1) to give (PE:EA=5:1, DCM:EA=3:1) to give the product of 2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazolo[1,5-a]pyridine-3-carboxamide (100 mg, yield: 30%). 1H-NMR (CDCl3, 400 MHz) δ 8.70 (s, 1H), 8.48 (s, 1H), 7.60˜7.63 (m, 2H), 7.12˜7.16 (m, 2H), 5.51 (br s, 1H), 3.72 (s, 3H), 2.93 (s, 3H), 2.81 (d, J=4.8 Hz, 3H), 1.29 (s, 12H). MS (M+H)+: 503.


Step 11—Synthesis of 5-(11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)pyrazolo[1,5-a]pyridine-3-carboxamide



embedded image


To a solution of 2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazolo[1,5-a]pyridine-3-carboxamide (110 mg, 0.22 mmol), 2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridine (50 mg, 0.18 mmol) and K2CO3 (50 mg, 0.366 mmol) in 1,4-dioxane (2 mL) were added X-Phos (10 mg) and Pd2(dba)3 (10 mg) under N2. The reaction mixture was stirred at 100° C. for 5 hours and concentrated in vacuo to remove 1,4-dioxane. The reaction mixture was extracted with EtOAc, washed with brine, dried over Na2SO4, filtered and evaporated concentrated in vacuo. The residue was purified by prep-HPLC to give the desired product (50 mg, yield: 44%). 1H-NMR (CDCl3, 400 MHz) δ 8.67 (s, 1H), 8.51 (s, 1H), 7.70˜7.73 (m, 3H), 7.52 (d, J=8.0 Hz, 1H), 7.27 (s, 1H), 7.23 (s, 1H), 7.14˜7.20 (m, 3H), 6.80 (t, J=8.0 Hz, 1H), 5.56 (br s, 1H), 4.36 (t, J=6.4 Hz, 2H), 3.51 (s, 3H), 3.34 (t, J=6.4 Hz, 2H), 2.85 (d, J=4.8 Hz, 3H), 2.63 (s, 3H). MS (M+H)+: 613.


Example 47



embedded image


Step 1—Synthesis of 5-bromo-2-nitrobenzaldehyde



embedded image


Fuming nitric acid (200 mL) was added to sulphuric acid (400 mL) at 0° C. and then 3-bromobenzaldehyde (100 g, 0.54 mol) was added dropwise in 15 minutes. After stirring for 10 minutes at the same temperature, the reaction mixture was poured into ice water and filtered to afford the mixture of products which was purified by flash column chromatography eluting with PE/EtOAc (100/1 to 20/1) to give 5-bromo-2-nitrobenzaldehyde as white solid (60 g, 48%). 1H-NMR (CDCl3, 400 MHz) δ 10.41 (s, 1H), 8.02˜8.06 (m, 2H), 7.88 (s, 1H). MS (M+H)+: 230/232.


Step 2—Synthesis of 5-bromo-2,4-dinitrobenzaldehyde



embedded image


To ice cold sulphuric acid (360 mL) was added fuming nitric acid (180 mL). 5-bromo-2-nitrobenzaldehyde (60 g, 0.26 mol) was added to the solution within 15 min. The reaction mixture was stirred for 10 minutes at the same temperature then at RT for 30 min, at 45° C. for 2 h and finally at 50° C. for 3 h, at which time TLC showed complete reaction. The reaction mixture was poured into ice water and extracted with chloroform to give a mixture of products which was separated by flash column chromatography eluting with PE/EtOAc (20/1 to 3/1). The desired 5-bromo-2,4-dinitrobenzaldehyde was isolated as white solid (6.5 g, 9%). 1H-NMR (CDCl3, 400 MHz) δ 10.48 (s, 1H), 8.61 (s, 1H), 8.30 (s, 1H). MS (M+H)+: 275/277.


Step 3—Synthesis of 5-bromo-3-cyano-2-(4-fluorophenyl)-6-nitro-2H-indazole 1-oxide



embedded image


To a stirred suspension of 5-bromo-2,4-dinitrobenzaldehyde (6.5 g, 0.024 mol) in acetic acid (120 mL) was added 4-fluoroaniline (9.28 g, 0.054 mol) and the reaction mixture was heated to 60° C. to get a clear solution. To this was added sodium cyanide portionwise (6.0 g, 0.123 mol) and the reaction mixture was stirred at 60° C. for 5 min. Acetic anhydride (1.9 mL, 0.020 mol) was added and the reaction mixture was stirred for 5 min at the same temperature. Sodium cyanide (6.0 g, 0.123 mol) was then added at the same temperature at which time the product precipitated out. After stirring for 30 min, the mixture was cooled to RT, diluted with water (50 mL), filtered and washed with ethanol to afford crude product, which was purified by flash column chromatography eluting with PE/EtOAc (100/1) to give pure 5-bromo-3-cyano-2-(4-fluorophenyl)-6-nitro-2H-indazole 1-oxide (5.0 g, 56%). 1H-NMR (CDCl3, 400 MHz) δ 8.40 (s, 1H), 8.15 (s, 1H), 7.67˜7.70 (m, 2H), 7.38˜7.42 (m, 2H). MS (M+H)+: 377/379.


Step 4—Synthesis of 5-bromo-2-(4-fluorophenyl)-6-nitro-2H-indazole-3-carbonitrile



embedded image


To a stirred solution of 5-bromo-3-cyano-2-(4-fluorophenyl)-6-nitro-2H-indazole 1-oxide (5.0 g, 0.013 mol) in chloroform (50 mL) was added phosphorus trichloride (5.0 mL, 0.057 mol) dropwise and the reaction mixture was heated to 60° C. for 1 h. Then the reaction mixture was poured into ice water and extracted into DCM (2×80 mL). The organic layer was concentrated, and ethanol was added to the residue. After filtration, 5-bromo-2-(4-fluorophenyl)-6-nitro-2H-indazole-3-carbonitrile was collected as solid (4.3 g, 91%). 1H-NMR (CDCl3, 400 MHz) δ 8.38 (s, 1H), 8.25 (s, 1H), 7.89˜7.91 (m, 2H), 7.33˜7.37 (m, 2H). MS (M+H)+: 361/363.


Step 5—Synthesis of 6-amino-5-bromo-2-(4-fluorophenyl)-2H-indazole-3-carbonitrile



embedded image


To a suspension of 5-bromo-2-(4-fluorophenyl)-6-nitro-2H-indazole-3-carbonitrile (500 mg, 139 mmol) in a mixture of methanol (50 mL), THF (5 mL) and water (2.5 mL) was added ammonium chloride (370 mg, 6.91 mmol) followed by iron powder (386 mg, 6.91 mmol). The mixture was heated to 80° C. and stirred vigorously for 90 min. The resulting suspension was filtered through celite and washed with EtOAc. The filtrate was concentrated, and the residue was suspended in water and extracted with EtOAc. The organic layer was contracted to give 6-amino-5-bromo-2-(4-fluorophenyl)-2H-indazole-3-carbonitrile (310 mg, 68%) as a yellow solid. 1H-NMR (CDCl3, 400 MHz) δ 7.99 (s, 1H), 7.80˜7.82 (m, 2H), 7.23˜7.57 (m, 2H), 7.03 (s, 1H), 4.40 (s, 2H). MS (M+H)+: 331/333.


Step 6—Synthesis of 6-amino-5-bromo-2-(4-fluorophenyl)-2H-indazole-3-carboxylic acid



embedded image


A suspension of 6-amino-5-bromo-2-(4-fluorophenyl)-2H-indazole-3-carbonitrile (50 mg, 0.14 mmol) in 10% NaOH (5 mL) was heated at 100° C. for 48 h. The reaction mixture was cooled to RT and poured into water (10 mL). After pH was adjusted to 4 with 1 N HCl, the mixture was extracted with EtOAc. The combined organic phases were washed with brine, dried over Na2SO4, filtered and evaporated to get 6-amino-5-bromo-2-(4-fluorophenyl)-2H-indazole-3-carboxylic acid (45 mg, yield: 85%). 1H-NMR (DMSO-d6, 400 MHz) δ 8.20 (s, 1H), 7.77˜7.82 (m, 2H), 7.45˜7.52 (m, 2H), 7.03 (s, 1H), 6.61 (s, 1H), 5.60 (s, 2H). MS (M+H)+: 350/352.


Step 7—Synthesis of 6-amino-5-bromo-2-(4-fluorophenyl)-N-methyl-2H-indazole-3-carboxamide



embedded image


To a solution of 6-amino-5-bromo-2-(4-fluorophenyl)-2H-indazole-3-carboxylic acid (1.0 g, 2.8 mmol) in dry DMF (500 mL) were added EDCI (1.1 g, 6.2 mmol) and HOBT (0.5 g, 4.3 mmol). The reaction mixture was stirred at room temperature for 1 h, and then Et3N (5 mL) and MeNH2.HCl (0.9 g, 14.3 mmol) were added to the reaction mixture. After stirring for another 2 hours, the reaction mixture was concentrated in vacuum and 300 mL Na2CO3 (a.q.) was added to the mixture. The resulting solid was filtered to give the crude product, which was purified by column chromatography (PE:EA=3:1) to give 6-amino-5-bromo-2-(4-fluorophenyl)-N-methyl-2H-indazole-3-carboxamide (0.8 g, yield: 74%). 1H-NMR (CDCl3, 400 MHz) δ 8.10 (s, 1H), 7.55˜7.58 (m, 2H), 7.20˜7.26 (m, 2H), 6.98 (s, 1H), 5.75 (s, 1H), 4.32 (s, 2H), 2.98 (d, J=4.8 Hz, 3H). MS (M+H)+: 363/365.


Step 8—Synthesis of 5-bromo-2-(4-fluorophenyl)-N-methyl-6-(methylsulfonamido)-2H-indazole-3-carboxamide



embedded image


To a degassed solution of 6-amino-5-bromo-2-(4-fluorophenyl)-N-methyl-2H-indazole-3-carboxamide (0.75 g, 2.1 mmol) in DCM/Pyridine (10 mL/3 mL) was added MsCl (1.2 g, 10.5 mmol) under N2 at 0° C. for 30 min. The reaction was stirred at 25° C. for 8 hours. The reaction mixture was diluted with water, extracted with EtOAc and washed with brine, and then dried over Na2SO4. After concentration, the residue was purified by column chromatography (PE:EA=2:1) to give crude 5-bromo-2-(4-fluorophenyl)-N-methyl-6-(methylsulfonamido)-2H-indazole-3-carboxamide (0.65 g, yield: 78%). 1H-NMR (CDCl3, 400 MHz) δ 8.30 (s, 1H), 8.03 (s, 1H), 7.56˜7.60 (m, 2H), 7.20˜7.25 (m, 2H), 6.97 (s, 1H), 5.73 (s, 1H), 3.08 (s, 3H), 2.99 (d, J=4.8 Hz, 3H). MS (M+H)+: 441/443.


Step 9—Synthesis of 5-bromo-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)-2H-indazole-3-carboxamide



embedded image


To a suspension of 5-bromo-2-(4-fluorophenyl)-N-methyl-6-(methylsulfonamido)-2H-indazole-3-carboxamide (0.5 g, 1.1 mmol) and K2CO3 (0.5 g, 3.4 mmol) in DMF (10 mL) was added dropwise CH3I (0.8 g, 5.6 mmol) at 0° C. under N2, and then the mixture was stirred at 80° C. for 2 hours. After concentration under vacuum, the residue was suspended in H2O and extracted with DCM. The combined organic layer was washed with H2O, brine, dried over Na2SO4, filtrated and concentrated to give 5-bromo-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)-2H-indazole-3-carboxamide (0.48 g, yield: 93%). 1H-NMR (CDCl3, 400 MHz) δ 8.21 (s, 1H), 8.01 (s, 1H), 7.56˜7.60 (m, 2H), 7.21˜7.27 (m, 2H), 6.97 (s, 1H), 3.228 (s, 3H), 3.07 (d, J=4.8 Hz, 3H), 2.77 (s, 3H). MS (M+H)+: 455/457.


Step 10—Synthesis of 2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2H-indazole-3-carboxamide



embedded image


To a degassed solution of 5-bromo-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)-2H-indazole-3-carboxamide (0.25 g, 0.55 mmol), KOAc (0.16 g, 1.65 mmol) and bis(pinacolato)diboron (0.71 g, 2.75 mmol) in 1,4-dioxane (5 mL), Pd(dppf)Cl2 (0.05 mg) was added. The reaction mixture was stirred at 100° C. for 6 hours, and then filtered through a celite pad. The filtrate was concentrated in vacuo, and the residue was purified by column chromatography (PE:EA=1:1) to give 2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2H-indazole-3-carboxamide (0.15 mg, yield: 50%). 1H-NMR (CDCl3, 400 MHz) δ 8.14 (s, 1H), 7.53˜7.56 (m, 3H), 7.13˜7.17 (m, 2H), 6.37 (s, 1H), 3.26 (s, 3H), 3.16 (d, J=4.8 Hz, 3H), 2.93 (s, 3H), 1.32 (s, 12H). MS (M+H)+: 503.


Step 11—Synthesis of 5-(11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)-2H-indazole-3-carboxamide



embedded image


To a degassed solution of 2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-2H-indazole-3-carboxamide (70 mg, 0.14 mmol), 2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridine (46 mg, 0.17 mmol) and K3PO4.3H2O (112 mg, 0.42 mmol) in 1,4-dioxane (5 mL) was added Pd2(dba)3 (13 mg, 0.014 mmol) and X-Phos (14 mg, 0.028 mmol) under N2. The reaction mixture was heated to 100° C. and stirred for 3 hours, and then filtered through a celite pad. The filtrate was concentrated in vacuo, and the residue was purified by prep-HPLC to give the desired product (20 mg, yield: 24%). 1H-NMR (CDCl3, 400 MHz) δ 8.03 (s, 1H), 7.83 (s, 1H), 7.55˜7.60 (m, 1H), 7.52˜7.54 (m, 2H), 7.45 (s, 1H), 7.09˜7.19 (m, 5H), 6.73 (s, 1H), 6.30 (s, 1H), 6.20 (s, 1H), 4.28 (t, J=6.0 Hz, 2H), 3.32 (s, 3H), 3.25 (t, J=6.0 Hz, 2H), 2.92 (d, J=4.8 Hz, 3H), 2.76 (s, 3H). MS (M+H)+: 613.


Example 48



embedded image


Step 1—Synthesis of 4-bromo-1,2-dihydropyridazine-3,6-dione



embedded image


To a solution of NH2NH2.H2SO4 (362 mg, 2.8 mmol) in H2O (5 mL) was added dropwise 3-bromofuran-2,5-dione (500 mg, 2.8 mmol), and the mixture was stirred at 90° C. for 4 hours. After the reaction, the mixture was filtered to get 4-bromo-1,2-dihydropyridazine-3,6-dione (300 mg, yield: 56%). 1H-NMR (DMSO-d6, 400 MHz) δ 12.47 (s, 1H), 11.17 (s, 1H), 7.62 (s, 1H). MS (M+H)+:191/193.


Step 2—Synthesis of 4-(4-fluoro-1H-indol-2-yl)-1,2-dihydropyridazine-3,6-dione



embedded image


To a degassed solution of 4-bromo-1,2-dihydropyridazine-3,6-dione (100 mg, 0.5 mmol), (4-fluoro-1H-indol-2-yl)boronic acid (131 mg, 0.7 mmol) and K3PO4 (279 mg, 1.1 mmol) in DMF (2 mL) and H2O (0.2 mL) was added Pd(dppf)Cl2 (5 mg) under N2. The mixture was stirred at 80° C. for 12 h. After the solvent was removed, the residue was purified by prep-TLC (DCM:EtOAc=10:1) to give the product of 4-(4-fluoro-1H-indol-2-yl)-1,2-dihydropyridazine-3,6-dione (50 mg, yield: 39%). 1H-NMR (DMSO-d6, 400 MHz) δ 11.95 (br. s, 1H), 7.47˜7.68 (m, 3H), 7.35 (d, J=8.0 Hz, 1H), 7.11˜7.19 (m, 1H), 6.81 (dd, J=8.0, 10.4 Hz, 1H). MS (M+H)+: 246.


Step 3—Synthesis of 2-(3,6-dichloropyridazin-4-yl)-4-fluoro-1H-indole



embedded image


A solution of 4-(4-fluoro-1H-indol-2-yl)-1,2-dihydropyridazine-3,6-dione (60 mg, 0.24 mmol) in POCl3 (5 mL) was stirred at 100° C. for 3 hours. After the reaction, it was concentrated and the residue was purified by column chromatography (PE:EA=5:1) to get the product of 2-(3,6-dichloropyridazin-4-yl)-4-fluoro-1H-indole (30 mg, yield: 43%). 1H-NMR (CDCl3, 400 MHz) δ 7.85 (s, 1H), 7.50 (s, 1H), 7.12˜7.21 (m, 2H), 6.70˜6.77 (m, 1H). MS (M+H)+: 282/284.


Step 4—Synthesis of 2-(6-chloro-3-vinylpyridazin-4-yl)-4-fluoro-1H-indole



embedded image


To a degassed solution of 2-(3,6-dichloropyridazin-4-yl)-4-fluoro-1H-indole (500 mg, 1.7 mmol), tributyl(vinyl)stannane (560 mg, 1.7 mmol) and LiCl (375 mg, 8.9 mmol) in DMF (5 mL) was added Pd(PPh3)2Cl2 (30 mg) under N2, and the mixture was stirred at 80° C. for 12 h. After the solvent was removed, the residue was purified by prep-TLC (PE:EtOAc=2:1) to give the product of 2-(6-chloro-3-vinylpyridazin-4-yl)-4-fluoro-1H-indole (140 mg, yield: 29%). 1H-NMR (CDCl3, 400 MHz) δ 8.71 (s, 1H), 7.61 (s, 1H), 7.16 (dd, J=10.8, 16.8 Hz, 2H), 7.03 (s, 1H), 6.87 (d, J=6.8 Hz, 2H), 6.68 (s, 1H), 5.79 (s, 1H). MS (M+H)+: 274/276.


Step 5—Synthesis of 2-(6-chloro-3-(2-chloroethyl)pyridazin-4-yl)-4-fluoro-1H-indole



embedded image


A solution of 2-(6-chloro-3-vinylpyridazin-4-yl)-4-fluoro-1H-indole (80 mg, 0.29 mmol) in HCl-dioxane (5 mL) was stirred at 50° C. After 15 minutes, NaHCO3 (a.q) was added. The mixture was extracted with DCM. The organic phase was dried over Na2SO4. After being concentrated in vacuo, the residue was purified by prep-TLC (DCM) to give the product of 2-(6-chloro-3-(2-chloroethyl)pyridazin-4-yl)-4-fluoro-1H-indole (60 mg, yield: 66%). 1H-NMR (CDCl3, 400 MHz) δ 9.04 (s, 1H), 7.63 (s, 1H), 7.28˜7.31 (m, 1H), 7.22˜7.26 (m, 1H), 6.99 (d, J=1.6 Hz, 1H), 6.84˜6.93 (m, 1H), 4.24 (t, J=6.4 Hz, 2H), 3.66 (t, J=6.5 Hz, 2H). MS (M+H)+: 310/312.


Step 6—Synthesis of 2-chloro-11-fluoro-5,6-dihydropyridazino[4′,3′:3,4]pyrido[1,2-a]indole



embedded image


To a suspension of 2-(6-chloro-3-(2-chloroethyl)pyridazin-4-yl)-4-fluoro-1H-indole (60 mg, 0.17 mmol) in DMF (1 mL) was added Cs2CO3 (124 mg, 0.34 mmol), and then the mixture was stirred for 20 minutes at 60° C. After being concentrated in vacuo, the resulting residue was suspended in H2O and extracted with DCM. The combined organic layer was washed with H2O and dried over Na2SO4. After concentration, the residue was purified by prep-TLC (DCM) to give the product of 2-chloro-11-fluoro-5,6-dihydropyridazino[4′,3′:3,4]pyrido[1,2-a]indole (40 mg, yield: 75%). 1H-NMR (CDCl3, 400 MHz) δ 8.40 (s, 1H), 7.63 (s, 1H), 7.47 (d, J=8.22 Hz, 1H), 7.24˜7.33 (m, 1H), 6.87˜6.97 (m, 1H), 4.52 (t, J=6.4 Hz, 2H), 3.53 (t, J=6.4 Hz, 2H). MS (M+H)+: 274/276.


Step 7—Synthesis of 5-(11-fluoro-5,6-dihydropyridazino[4′,3′: 3,4]pyrido[1,2-a]indol-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a degassed solution of 2-chloro-11-fluoro-5,6-dihydropyridazino[4′,3′:3,4]pyrido[1,2-a]indole (100 mg, 0.37 mmol), (2-(4-fluorophenyl)-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)benzofuran-5-yl)boronic acid (169 mg, 0.4 mmol) and K3PO4 (155 mg, 0.73 mmol) in DMF (3 mL) was added Pd(dtbpf)Cl2 (5 mg) under N2. Then the mixture was stirred at 100° C. for 3 hours. The reaction mixture was cooled to RT and filtered. The filtrate was washed with H2O and dried over Na2SO4. After being concentrated, the residue was purified by prep-HPLC to give the product of 5-(11-fluoro-5,6-dihydropyridazino[4′,3′:3,4]pyrido[1,2-a]indol-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (30 mg, yield: 13%). 1H-NMR (CDCl3, 400 MHz) δ 8.11 (d, J=9.0 Hz, 2H), 8.00 (dd, J=5.6, 8.8 Hz, 2H), 7.66 (s, 1H), 7.25 (s, 1H), 7.15˜7.23 (m, 4H), 6.81 (dd, J=7.6, 9.6 Hz, 1H), 6.17 (d, J=4.0 Hz, 1H), 4.47 (t, J=6.4 Hz, 2H), 3.67 (t, J=6.4 Hz, 2H), 3.21 (s, 3H), 3.00 (d, J=4.8 Hz, 3H), 2.97 (s, 3H). MS (M+H)+: 614.


Examples 49-51



embedded image


Step 1—Synthesis of 2-(3-allyl-6-chloropyridin-2-yl)-4-fluoro-1H-indole



embedded image


To a mixture of 6-chloro-2-(4-fluoro-1H-indol-2-yl)pyridin-3-yl trifluoromethanesulfonate (2 g, 5.1 mmol), allyltributylstannane (2 g, 6.1 mmol) and anhydrous LiCl (256 mg, 6.1 mmol) in THF (30 mL), Pd(PPh3)4 (588 mg, 0.51 mmol) was added under N2 protection. The reaction mixture was stirred at 60° C. overnight. After it was concentrated in vacuo, the residue was purified by column chromatography (PE:EA=400:1˜100:1) to give the product of 2-(3-allyl-6-chloropyridin-2-yl)-4-fluoro-1H-indole (1 g, yield: 68%). 1H-NMR (CDCl3, 400 MHz) δ 9.72 (s, 1H), 7.57 (d, J=8.0 Hz, 1H), 7.12˜7.24 (m, 3H), 7.02 (s, 1H), 6.77 (t, J=8.0 Hz, 1H), 6.04˜6.12 (m, 1H), 5.25 (d, J=10.0 Hz, 1H), 5.13 (d, J=17.2 Hz, 1H), 3.74 (d, J=6.0 Hz, 2H). MS (M+H)+: 287/289.


Step 2—Synthesis of 3-(6-chloro-2-(4-fluoro-1H-indol-2-yl)pyridin-3-yl)propane-1,2-diol



embedded image


A mixture of 2-(3-allyl-6-chloropyridin-2-yl)-4-fluoro-1H-indole (100 mg, 0.35 mmol) and NMO (124 mg, 1.05 mmol) in THF (5 mL) and H2O (2.5 mL) was stirred at 0° C. Then OsO4 (20 mg) was added, and the mixture was stirred at RT overnight. After cooling to 0° C., a.q. Na2S2O3 was added and extracted with CH2Cl2. Then the organic layer was washed with water, brine, dried over Na2SO4 and concentrated on a rotary evaporator. The residue was purified by column chromatography (PE:EA=2:1˜1:1) to give the product of 3-(6-chloro-2-(4-fluoro-1H-indol-2-yl)pyridin-3-yl)propane-1,2-diol (100 mg, yield: 89%). 1H-NMR (CDCl3, 400 MHz) δ 9.88 (s, 1H), 7.65 (d, J=8.4 Hz, 1H), 7.12˜7.18 (m, 3H), 7.02 (s, 1H), 6.76 (t, J=8.0 Hz, 1H), 4.12˜4.16 (m, 1H), 3.81˜3.85 (m, 1H), 3.64˜3.67 (m, 1H), 3.07˜3.15 (m, 2H). MS (M+H)+: 321/323.


Step 3—Synthesis of 3-(6-chloro-2-(4-fluoro-1H-indol-2-yl)pyridin-3-yl)-2-hydroxypropyl methanesulfonate



embedded image


A mixture of 3-(6-chloro-2-(4-fluoro-1H-indol-2-yl)pyridin-3-yl)propane-1,2-diol (100 mg, 0.31 mmol) and TEA (94 mg, 0.93 mmol) in THF (6 mL) was stirred under N2 at 0° C., MsCl (36 mg, 0.31 mmol) was added dropwise. The reaction was stirred at 0° C. for 1 h and RT overnight. Water was added and after extraction with EA, the organic layer was washed with water, brine, dried over Na2SO4 and concentrated on a rotary evaporator. The residue was purified by column chromatography (PE:EA=2:1˜1:1) to give the product of 3-(6-chloro-2-(4-fluoro-1H-indol-2-yl)pyridin-3-yl)-2-hydroxypropyl methanesulfonate (60 mg, yield: 48%). 1H-NMR (MeOD, 400 MHz) δ 7.80 (d, J=7.6 Hz, 1H), 7.26˜7.32 (m, 2H), 7.09˜7.11 (m, 1H), 7.04 (s, 1H), 6.70 (t, J=7.6 Hz, 1H), 4.20˜4.29 (m, 3H), 3.09˜3.13 (m, 5H). MS (M+H)+: 399/401.


Step 4—Synthesis of (2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-6-yl) methanol



embedded image


A solution of 3-(6-chloro-2-(4-fluoro-1H-indol-2-yl)pyridin-3-yl)-2-hydroxypropyl methanesulfonate (100 mg, 0.25 mmol) in DMF (5 mL) was stirred under N2 at 0° C. After NaH (30 mg, 60% in mineral oil, 0.75 mmol) was added, the mixture was stirred at 0° C. for 0.5 h and RT for 2 h. After cooling to 0° C., water was added and the mixture extracted with EtOAc. Then the organic layer was washed with water, brine, dried over Na2SO4 and concentrated on a rotary evaporator. The residue was purified by column chromatography (PE:EA=2:1˜1:1) to give the product of (2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-6-yl)methanol (70 mg, yield: 92%). 1H-NMR (CDCl3, 400 MHz) δ 7.50 (d, J=7.6 Hz, 1H), 7.37 (s, 1H), 7.13˜7.19 (m, 3H), 6.78 (t, J=7.6 Hz, 1H), 4.79˜4.84 (m, 1H), 3.69˜3.73 (m, 1H), 3.60˜3.62 (m, 1H), 3.38˜3.42 (m, 1H), 3.24 (d, J=16.4 Hz, 1H). MS (M+H)+: 303/305.


Step 5—Synthesis of 2-(2,4-difluorophenyl)-5-(11-fluoro-6-(hydroxymethyl)-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide, (R or S)-2-(2,4-difluorophenyl)-5-(1.1-fluoro-6-(hydroxymethyl)-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide and (S or R)-2-(2,4-difluorophenyl)-5-(11-fluoro-6-(hydroxymethyl)-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (Compound 48, Compound 49 and Compound 50)



embedded image


To a solution of (2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-6-yl)methanol (200 mg, 0.66 mmol), 2-(2,4-difluorophenyl)-N-methyl-6-(N-methylmethyl sulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide (346 mg, 0.66 mmol), K3PO4 (420 mg, 1.98 mmol), Pd2(dba)3 (63 mg) and X-Phos (63 mg) in dioxane/H2O (9 mL/2 mL) was stirred at 100° C. for 3 h. After the reaction, the mixture was filtered to remove the solid and then the mixture was diluted with water and extracted with EA. The organic layer was washed with brine, dried over aq. Na2SO4 and concentrated. The residue was purified by column chromatography (PE:EA=1:1) to give 2-(2,4-difluorophenyl)-5-(11-fluoro-6-(hydroxymethyl)-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (Compound 49, 350 mg, yield: 87%). 1H-NMR (CDCl3, 400 MHz) δ 8.09 (s, 1H), 7.63˜7.73 (m, 3H), 7.43 (d, J=8.0 Hz, 1H), 7.1˜57.25 (m, 3H), 6.90˜7.03 (m, 2H), 6.78 (d, J=8.0 Hz, 1H), 5.79˜5.81 (m, 1H), 4.85˜4.87 (m, 1H), 3.51˜3.76 (m, 3H), 3.38 (s, 3H), 3.28˜3.32 (m, 1H), 2.92 (d, J=5.2 Hz, 3H), 2.64 (s, 3H). MS (M+H)+: 661.


2-(2,4-difluorophenyl)-5-(11-fluoro-6-(hydroxymethyl)-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide was separated by SFC to afford two single enantiomers.


Compound 50 (enantiomer 1, peak 1 on SFC, AD-H3UM45404ML3 MIN, HPLC_RT=2.193 min HPLC_RT=2.193 min) (120 mg). 1H-NMR (CDCl3, 400 MHz) δ 8.09 (s, 1H), 7.63˜7.73 (m, 3H), 7.43 (d, J=8.0 Hz, 1H), 7.15˜7.25 (m, 3H), 6.90˜7.03 (m, 2H), 6.78 (d, J=8.0 Hz, 1H), 5.79˜5.81 (m, 1H), 4.85˜4.87 (m, 1H), 3.51˜3.76 (m, 3H), 3.38 (s, 3H), 3.2˜83.32 (m, 1H), 2.92 (d, J=5.2 Hz, 3H), 2.64 (s, 3H). MS (M+H)+: 661.


Compound 51 (enantiomer 2, peak 2 on SFC, AD-H3UM45404ML3 MIN, HPLC_RT=2.193 min HPLC_RT=2.555 min) (120 mg). 1H-NMR (CDCl3, 400 MHz) δ 8.09 (s, 1H), 7.63˜7.73 (m, 3H), 7.43 (d, J=8.0 Hz, 1H), 7.15˜7.25 (m, 3H), 6.90˜7.03 (m, 2H), 6.78 (d, J=8.0 Hz, 1H), 5.79˜5.81 (m, 1H), 4.85˜4.87 (m, 1H), 3.51˜3.76 (m, 3H), 3.38 (s, 3H), 3.2˜83.32 (m, 1H), 2.92 (d, J=5.2 Hz, 3H), 2.64 (s, 3H). MS (M+H)+: 661.


Examples 52-54

Examples 52-54, depicted in the table below, were prepared in accordance with the method described for Examples 49-51.














Example
Structure
MS (M + H)+







52


embedded image


643





53


embedded image


643





54


embedded image


643





55


embedded image


629









Examples 56-57



embedded image


Step 1—Synthesis of (2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-6-yl)methyl methanesulfonate



embedded image


A mixture of (2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-6-yl)methanol (200 mg, 0.66 mmol) and TEA (200 mg, 1.98 mmol) in THF (8 mL) was stirred at 0° C. and MsCl (114 mg, 0.99 mmol) was added dropwise. The reaction was stirred at RT for 8 h. The reaction was washed with 1N HCl and extracted with DCM, dried with Na2SO4 and concentrated to give (2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-6-yl)methyl methanesulfonate (200 mg, yield: 80%). 1H-NMR (CDCl3, 400 MHz) δ 7.54 (d, J=8.0 Hz, 1H), 7.41 (s, 1H), 7.11˜7.20 (m, 3H), 6.79˜6.83 (m, 1H), 5.03˜5.08 (m, 1H), 4.06˜4.21 (m, 2H), 3.46˜3.52 (m, 1H), 3.24˜3.28 (m, 1H), 2.69 (s, 3H). MS (M+H)+: 381/383.


Step 2—Synthesis of (2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-6-yl)methyl methanesulfonate



embedded image


To a degassed solution of (2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-6-yl)methyl methanesulfonate (600 mg, 1.6 mmol) in DMF (6 mL) was added CH3SNa (100 mg, 1.6 mmol) at 0° C., and the mixture was stirred at 0° C. for 3 h. Then the mixture was diluted with water (10 mL) and extracted with EtOAc (10 mL*3). The combined organic layer was washed with brine (30 mL), dried over Na2SO4 and concentrated. The residue was purified by column chromatography (PE:EA=5:1) to afford the desired product of 2-chloro-11-fluoro-6-((methylthio)methyl)-5,6-dihydroindolo[1,2-h][1,7]naphthyridine (350 mg, yield: 66.8%). 1H-NMR (CDCl3, 400 MHz) δ 7.61 (d, J=7.6 Hz, 1H), 7.45 (s, 1H), 7.18˜7.26 (m, 3H), 6.87 (dd, J=8.0, 9.6 Hz, 1H), 4.84˜4.92 (m, 1H), 3.42˜3.55 (m, 2H), 2.65˜2.72 (m, 1H), 2.52˜2.60 (m, 1H), 2.15 (s, 3H). MS (M+H)+: 333/335.


Step 3—Synthesis of 2-chloro-11-fluoro-6-((methylsulfonyl)methyl)-5,6-dihydroindolo[1,2-h][1,7]naphthyridine



embedded image


To a degassed solution of 2-chloro-11-fluoro-6-((methylthio)methyl)-5,6-dihydroindolo[1,2-h][1,7]naphthyridine (100 mg, 0.3 mmol) in MeOH (2 mL) was added a solution of Oxone (185 mg, 0.3 mmol) in H2O (2 mL) at 0° C., and the mixture was stirred at 25° C. for 24 h. Then the mixture was diluted with water (5 mL) and extracted with EtOAc (5 mL×3). The combined organic layer was washed with brine (10 mL), dried over Na2SO4 and concentrated. The residue was purified by column chromatography (DCM:EA=10:1) to afford the desired product of 2-chloro-11-fluoro-6-((methylsulfonyl)methyl)-5,6-dihydroindolo[1,2-h][1,7]naphthyridine (70 mg, yield: 64%). 1H-NMR (CDCl3, 400 MHz) δ 7.62 (d, J=7.6 Hz, 1H), 7.43 (s, 1H), 7.22˜7.29 (m, 3H), 6.83˜6.89 (m, 1H), 5.45˜5.51 (m, 1H), 3.51˜3.62 (m, 2H), 3.14˜3.21 (m, 1H), 2.97˜3.05 (m, 1H), 2.84 (s, 3H). MS (M+H)+: 365/367.


Step 4—Synthesis of (R or S)-5-(11-fluoro-6-((methylsulfonyl)methyl)-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide and (S or R)-5-(11-fluoro-6-((methylsulfonyl)methyl)-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


56 (Enantiomer 1, peak 1 on SFC) 57 (Enantiomer 2, peak 2 on SFC)


To a degassed solution of 2-chloro-11-fluoro-6-((methylsulfonyl)methyl)-5,6-dihydroindolo[1,2-h][1,7]naphthyridine (220 mg, 0.6 mmol), 2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide (333 mg, 0.66 mmol) and Na2CO3 (128 mg, 1.2 mmol) in dioxane/water (5 mL/1 ml) was added Pd(dtbpf)Cl2 (22 mg) under N2. The mixture was stirred at 100° C. for 3 h. After the solvent was removed, the residue was purified by prep-TLC (DCM:EtOAc=3:1) and SFC to give two single enantiomers.


Compound 56 (enantiomer 1, peak 1 on SFC, AD-H3UM5404ML8 MIN, HPLC_RT=3.116 min) (100 mg, yield: 23.5%). 1H-NMR (CDCl3, 400 MHz) δ 8.06 (s, 1H), 7.97 (dd, J=5.6, 8.0 Hz, 2H), 7.77 (d, J=7.6 Hz, 1H), 7.66 (s, 1H), 7.52 (d, J=7.6 Hz, 1H), 7.3˜17.35 (m, 2H), 7.19˜7.25 (m, 3H), 6.85 (t, J=8.8 Hz, 1H), 5.89 (d, J=4.4 Hz, 1H), 5.52 (br. s, 1H), 3.60˜3.68 (m, 2H), 3.42 (s, 3H), 3.26 (dd, J=9.6, 13.6 Hz, 1H), 3.08 (d, J=13.6 Hz, 1H), 3.00 (d, J=4.8 Hz, 3H), 2.83 (s, 3H), 2.77 (s, 3H). MS (M+H)+: 705.


Compound 57 (enantiomer 2, peak 2 on SFC, AD-H3UM5404ML8 MIN, HPLC_RT=4.637 min) (100 mg, yield: 23.5%). 1H-NMR (CDCl3, 400 MHz) δ 8.06 (s, 1H), 7.97 (dd, J=5.6, 8.0 Hz, 2H), 7.77 (d, J=7.6 Hz, 1H), 7.66 (s, 1H), 7.52 (d, J=7.6 Hz, 1H), 7.31˜7.35 (m, 2H), 7.19˜7.25 (m, 3H), 6.85 (t, J=8.8 Hz, 1H), 5.89 (d, J=4.4 Hz, 1H), 5.52 (br. s., 1H), 3.60˜3.68 (m, 2H), 3.42 (s, 3H), 3.26 (dd, J=9.6, 13.6 Hz, 1H), 3.08 (d, J=13.6 Hz, 1H), 3.00 (d, J=4.8 Hz, 3H), 2.83 (s, 3H), 2.77 (s, 3H). MS (M+H)+: 705.


Examples 58-59

Compounds 58 and 59, depicted in the table below, were prepared in accordance with the method described for Examples 56-57.















Compound


MS


ID
Structure
NMR
(M + H)+







58


embedded image



1H-NMR (CDCl3, 400 MHz) δ 8.14 (s, 1H), 7.78 (d, J = 7.6 Hz, 2H), 7.68 (s, 1H), 7.52 (d, J = 7.6 Hz, 1H), 7.31~7.35 (m, 2H), 7.21~7.25 (m, 1H), 7.09 (t, J = 7.6 Hz, 1H), 7.02 (t, J = 8.8 Hz, 1H), 6.85 (t, J = 1H), 5.75 (br s, 1H), 5.52 (br s, 1H), 3.62~3.69 (m, 2H), 3.42 (s, 3H), 3.23~3.29 (m, 1H), 3.08 (d, J = 12.4 Hz, 1H), 2.97 (d, J = 4.8 Hz, 3H), 2.84 (s, 3H), 2.77 (s, 3H).

723





59


embedded image



1H-NMR (CDCl3, 400 MHz) δ 8.14 (s, 1H), 7.78 (d, J = 7.6 Hz, 2H), 7.68 (s, 1H), 7.52 (d, J = 7.6 Hz, 1H), 7.31~7.35 (m, 2H), 7.21~7.25 (m, 1H), 7.09 (t, J = 7.6 Hz, 1H), 7.02 (t, J = 8.8 Hz, 1H), 6.85 (t, J = 8.8 Hz, 1H), 5.75 (br s, 1H), 5.52 (br s, 1H), 3.62~3.69 (m, 2H), 3.42 (s, 3H), 3.23~3.29 (m, 1H), 3.08 (d, J = 12.4 Hz, 1H), 2.97 (d, J = 4.8 Hz, 3H), 2.84 (s, 3H), 2.77 (s, 3H).

723









Examples 60-61



embedded image


Step 1—Synthesis of 2-(2-chloro-5-(prop-1-en-2-yl)pyridin-4-yl)-4-fluoro-1H-indole



embedded image


To a suspension of 6-chloro-4-(4-fluoro-1H-indol-2-yl)pyridin-3-yl trifluoromethanesulfonate (100 mg, 0.25 mmol) and tributyl(prop-1-en-2-yl)stannane (100 mg, 0.28 mmol) in DMF (2 mL) was added Pd(PPh3)4 (10 mg, 0.02 mmol) at 100° C. under N2, and the mixture was stirred for 5 hours. After concentrated in vacuum, the residue was suspended in H2O and extracted with EA. The combined organic layer was washed with H2O, brine, dried over Na2SO4, filtrated and concentrated to give 2-(2-chloro-5-(prop-1-en-2-yl)pyridin-4-yl)-4-fluoro-1H-indole (50 mg, yield: 70%). 1H-NMR (CDCl3, 400 MHz) δ 8.96 (s, 1H), 8.14 (s, 1H), 7.47 (s, 1H), 7.08˜7.13 (m, 2H), 6.95 (s, 1H), 6.72˜6.76 (m, 1H), 5.39 (s, 1H), 5.23 (s, 1H), 1.77 (s, 3H). MS (M+H)+: 287/289.


Step 2—Synthesis of 2-(6-chloro-4-(4-fluoro-1H-indol-2-yl)pyridin-3-yl)propan-1-ol



embedded image


To a degassed solution of 2-(2-chloro-5-(prop-1-en-2-yl)pyridin-4-yl)-4-fluoro-1H-indole (50 mg, 0.17 mmol) was added BH3-DMS (0.5 mL) under N2 protection at 0° C., and the mixture was stirred for 1 hour. Then NaBO3.4H2O (60 mg, 0.34 mmol) and H2O2 (0.2 mL) was added under N2 protection at 0° C. for 0.5 hours, and the reaction mixture was stirred at RT for 2 hours. After diluted with H2O and extracted with EtOAc, the combined organic layer was washed with H2O, brine, dried over Na2SO4, and the residue was purified by prep-HPLC to give 2-(6-chloro-4-(4-fluoro-1H-indol-2-yl)pyridin-3-yl)propan-1-ol (20 mg, yield: 36%). 1H-NMR (CDCl3, 400 MHz) δ 10.86 (s, 1H), 8.17 (s, 1H), 7.33 (s, 1H), 7.18 (s, 1H), 7.06˜7.10 (m, 1H), 6.71˜7.76 (m, 2H), 4.10 (s, 1H), 3.95 (s, 1H), 3.75 (t, J=12.0 Hz 1H), 3.42˜3.51 (m, 1H), 1.14 (d, J=8.0 Hz, 3H). MS (M+H)+: 305/307.


Step 3—Synthesis of 2-chloro-11-fluoro-5-methyl-5,6-dihydroindolo[2,1-a][2,6]naphthyridine



embedded image


To a suspension of 2-(6-chloro-4-(4-fluoro-1H-indol-2-yl)pyridin-3-yl)propan-1-ol (60 mg, 0.21 mmol) and DEAD (111 mg, 0.63 mmol) in THF (4 mL) was added PPh3 (156 mg, 0.63 mmol) at 0° C. under N2, and the mixture was stirred for 12 hours. Then the mixture was diluted with H2O and extracted with EA. The combined organic layer was washed with H2O, brine, dried over Na2SO4, filtrated and concentrated to give 2-chloro-11-fluoro-5-methyl-5,6-dihydroindolo[2,1-a][2,6]naphthyridine (30 mg, yield: 56%). 1H-NMR (CDCl3, 400 MHz) δ 8.26 (s, 1H), 7.54 (s, 1H), 7.05˜7.15 (m, 3H), 6.73˜6.77 (m, 1H), 4.16˜4.20 (m, 1H), 4.00˜4.04 (m, 1H), 3.31˜3.35 (m, 1H), 1.30 (d, J=8.0 Hz, 3H). MS (M+H)+: 287/289.


Step 4—Synthesis of (R or S)-5-(11-fluoro-5-methyl-5,6-dihydroindolo[2,1-a][2,6]naphthyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide and (S or R)-5-(11-fluoro-5-methyl-5,6-dihydroindolo[2,1-a][2,6]naphthyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a stirring mixture of 2-chloro-11-fluoro-5-methyl-5,6-dihydroindolo[2,1-a][2,6]naphthyridine (100 mg, 0.34 mmol), 2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide (191 mg, 0.38 mmol) and K3PO4.3H2O (79 mg, 1.00 mmol) in Dioxane/H2O (2 mL/0.2 mL) was added X-Phos (20 mg) and Pd2(dba)3 (20 mg) under N2 protection. The mixture was stirred at a pre-heated oil-bath at 100° C. for 2 h. The reaction mixture was concentrated in vacuo and it was extracted with EtOAc. It was washed with H2O, brine and dried over Na2SO4. After concentration, the residue was purified by prep-HPLC and SFC to give two single enantiomers.


Compound 60 (enantiomer 1, peak 1 on SFC, AD-H3UM5603ML5MIN, HPLC_RT=0.810 min) (40 mg, yield: 19.8%). 1H-NMR (CDCl3, 400 MHz) δ 8.65 (s, 1H), 7.98˜8.05 (m, 4H), 7.69 (s, 1H), 7.16˜7.23 (m, 5H), 6.80˜6.84 (m, 1H), 6.09 (s, 1H), 4.31˜4.35 (m, 1H), 4.12˜4.16 (m, 1H), 3.47˜3.52 (m, 1H), 3.24 (s, 3H), 3.01 (d, J=4.8 Hz, 3H), 2.88 (s, 3H), 1.30 (d, J=8.0 Hz, 3H). MS (M+H)+: 627.


Compound 61 (enantiomer 2, peak 2 on SFC, AD-H3UM5603ML5MIN, HPLC_RT=2.422 min) (40 mg, yield: 19.8%). 1H-NMR (CDCl3, 400 MHz) δ 8.56 (s, 1H), 7.90˜8.95 (m, 4H), 7.59 (s, 1H), 7.16˜7.23 (m, 5H), 6.80˜6.84 (m, 1H), 6.09 (s, 1H), 4.22˜4.26 (m, 1H), 4.04˜4.07 (m, 1H), 3.39˜3.41 (m, 1H), 3.16 (s, 3H), 2.93 (d, J=4.8 Hz, 3H), 2.80 (s, 3H), 1.37 (d, J=8.0 Hz, 3H). MS (M+H)+: 627.


Example 62

Example 62, depicted in the table below, were prepared in accordance with the method described for Examples 60-61.


















MS


Example
Structure
NMR
(M + H)+







62


embedded image



1H-NMR (CDCl3, 400 MHz) δ 7.95~8.00 (m, 3H), 7.71 (d, J = 8.0 Hz, 1H), 7.67 (s, 1H), 7.50 (d, J = 8.0 Hz, 1H), 7.29 (s, 1H), 7.17~7.22 (m, 4H), 6.80~6.83 (m, 1H), 6.06 (s, 1H), 4.33~4.37 (m, 1H), 4.07~4.11 (m, 1H), 3.46~3.49 (m, 1H), 3.44 (s, 3H), 2.98 (d, J = 4.8 Hz, 3H), 2.66 (s, 3H), 1.45 (d, J = 8.0 Hz, 3H).

627









Example 63



embedded image


Step 1—Synthesis of 2-chloro-4-(4-fluoro-1H-indol-2-yl)pyrimidin-5-yl trifluoromethanesulfonate



embedded image


DIPEA (1.5 g, 11.61 mmol) was added to a suspension of 2-chloro-4-(4-fluoro-1H-indol-2-yl)pyrimidin-5-ol (1.0 g, 3.79 mmol) in anhydrous DCM (20 mL) under N2. The reaction mixture was cooled to 0° C., and then trifluoromethanesulfonic anhydride (2.2 g, 7.80 mmol) was added dropwise. After stirring at 0° C. for 30 minutes, the mixture was diluted with water and extracted with CH2Cl2. The organic layers were washed with brine, dried over Na2SO4, and concentrated. The residue was purified by column chromatography (PE:EA=10:1) to give the product of 2-chloro-4-(4-fluoro-1H-indol-2-yl)pyrimidin-5-yl trifluoromethanesulfonate (1.0 g, yield: 66.7%). 1H-NMR (DMSO-d6, 400 MHz) δ 12.47 (s, 1H), 9.09 (s, 1H), 7.41˜7.46 (m, 2H), 7.28˜7.34 (m, 1H), 6.89˜6.94 (m, 1H). MS (M+H)+: 396/398.


Step 2—Synthesis of 2-(2-chloro-5-vinylpyrimidin-4-yl)-4-fluoro-1H-indole



embedded image


To a mixture of 2-chloro-4-(4-fluoro-1H-indol-2-yl)pyrimidin-5-yl trifluoromethanesulfonate (500 mg, 1.26 mmol), tributyl(vinyl)stannane (400 mg, 1.26 mmol) and anhydrous LiCl (150 mg, 3.54 mmol) in DMF (5 mL), Pd(PPh3)2Cl2 (50 mg, 0.07 mmol) was added under N2 protection. The reaction mixture was stirred at 60° C. for 1 h. Then it was concentrated in vacuo, and the residue was purified by column chromatography (PE:EA=10:1) to give the product of 2-(2-chloro-5-vinylpyrimidin-4-yl)-4-fluoro-1H-indole (250 g, yield: 72.3%). 1H-NMR (DMSO-d6, 400 MHz) δ 12.21 (br s, 1H), 8.85 (s, 1H), 7.41 (d, J=7.6 Hz, 1H), 7.21˜7.27 (m, 1H), 7.08˜7.20 (m, 2H), 6.87 (dd, J=10.4, 7.6 Hz, 1H), 6.01 (dd, J=17.6, 1.2 Hz, 1H), 5.72 (dd, J=11.2, 1.2 Hz, 1H). MS (M+H)+: 274/276.


Step 3—Synthesis of 5-(4-(4-fluoro-1H-indol-2-yl)-5-vinylpyrimidin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a degassed solution of 2-(2-chloro-5-vinylpyrimidin-4-yl)-4-fluoro-1H-indole (100 mg, 0.36 mmol) and (2-(4-fluorophenyl)-3-(methylcarbamoyl)-6-(N-methylmethyl sulfonamido)benzofuran-5-yl)boronic acid (150 mg, 0.36 mmol) in 1,4-dioxane (5 mL), Pd2(dba)3 (20 mg), X-Phos (20 mg) and K3PO4 (200 mg, 0.56 mmol) were added under N2. The mixture was heated to 80° C. for 2 hours. The reaction mixture was cooled to RT, filtered and washed with EtOAc. The filtrate was washed with H2O, brine, and dried over Na2SO4. After concentration, the residue was purified by column chromatography (DCM:MeOH 40:1) to give the product of 5-(4-(4-fluoro-1H-indol-2-yl)-5-vinylpyrimidin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (150 mg, yield: 68.4%). 1H-NMR (CDCl3, 400 MHz) δ 10.76 (br s, 1H), 8.88 (s, 1H), 8.63 (s, 1H), 7.98˜8.05 (m, 2H), 7.64 (s, 1H), 7.59 (s, 1H), 7.15˜7.25 (m, 6H), 6.74˜6.84 (m, 1H), 5.91 (d, J=18.0 Hz, 1H), 5.69 (d, J=11.2 Hz, 1H), 3.39 (s, 3H), 3.03 (d, J=4.8 Hz, 3H), 2.87 (s, 3H). MS (M+H)+: 614


Step 4—Synthesis of 5-(11-fluoro-5,6-dihydropyrimido[4′,5′:3,4]pyrido[1,2-a]indol-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a solution of 5-(4-(4-fluoro-1H-indol-2-yl)-5-vinylpyrimidin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (50 mg, 0.08 mmol) in DMAc (1 mL), K3PO4 (100 mg, 0.37 mmol) was added under N2 protection. The reaction mixture was stirred at 90° C. overnight. Then the mixture was diluted with water and extracted with EtOAc. The organic layers were washed with brine, dried over Na2SO4, and concentrated. The residue was purified by prep-HPLC to give the product of 5-(11-fluoro-5,6-dihydropyrimido[4′,5′:3,4]pyrido[1,2-a]indol-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (10 mg, yield: 20.0%). 1H-NMR (CDCl3, 400 MHz) δ 8.75 (s, 1H), 8.36 (s, 1H), 8.00˜8.10 (m, 2H), 7.75 (s, 1H), 7.53 (s, 1H), 7.15˜7.25 (m, 4H), 6.84 (t, J=8.8 Hz, 1H), 6.01 (br s, 1H), 4.41 (t, J=6.4 Hz, 2H), 3.50 (s, 3H), 3.34 (t, J=6.4 Hz, 2H), 3.05 (d, J=4.8 Hz, 3H), 2.83 (s, 3H). MS (M+H)+: 614


Example 64



embedded image


Step 1—Synthesis of 1-(6-chloro-4-(4-fluoro-1H-indol-2-yl)pyridin-3-yl)ethane-1,2-diol



embedded image


To a solution of 2-(2-chloro-5-vinylpyridin-4-yl)-4-fluoro-1H-indole (2 g, 7.35 mmol) in THF—H2O (26 mL-13 mL), NMO (2.15 g, 18.38 mmol) and OsO4 (56 mg, 0.22 mmol) were added. The reaction mixture was stirred at room temperature for 2 hours. Then Na2SO3 and water were added. The mixture was stirred at room temperature for 20 min and filtered. The filtrate was extracted with EA. The organic layer was washed with brine, dried over Na2SO4 and concentrated. The residue was purified by column chromatography (DCM:MeOH=10:1) to give 1-(6-chloro-4-(4-fluoro-1H-indol-2-yl)pyridin-3-yl)ethane-1,2-diol (1.9 g, yield: 84.4%). 1H-NMR (Methanol-d4, 400 MHz) 8.61 (s, 1H), 7.63 (s, 1H), 7.24 (d, J=8.0 Hz, 1H), 7.08˜7.19 (m, 1H), 6.90 (s, 1H), 6.75 (dd, J=7.6, 10.0 Hz, 1H), 5.19 (t, J=6.0 Hz, 1H), 3.75˜3.82 (m, 1H), 3.67˜3.74 (m, 1H). MS (M+H)+: 307/309.


Step 2—Synthesis of 6-chloro-4-(4-fluoro-1H-indol-2-yl)nicotinaldehyde



embedded image


To a solution of 1-(6-chloro-4-(4-fluoro-1H-indol-2-yl)pyridin-3-yl)ethane-1,2-diol (1.9 g, 6.21 mmol) in THF—H2O (26 mL-13 mL), NaIO4 (1.94 g, 9.31 mmol) was added, the reaction mixture was stirred at room temperature for 2 hours. Then Na2SO3 and water were added and the mixture was stirred at room temperature for 30 min. The mixture was extracted with EA. The organic layer was washed with brine, dried over Na2SO4 and concentrated. The residue was purified by column chromatography (DCM:MeOH=20:1) to afford the desired product of 6-chloro-4-(4-fluoro-1H-indol-2-yl)nicotinaldehyde (1.4 g, yield: 82.4%). 1H-NMR (Methanol-d4, 400 MHz) δ 8.48 (s, 1H), 7.79 (s, 1H), 7.40 (d, J=8.4 Hz, 1H), 7.17˜7.25 (m, 1H), 6.95 (s, 1H), 6.79 (dd, J=8.0, 10.0 Hz, 1H), 6.70 (s, 1H). MS (M+H)+: 275/277.


Step 3—Synthesis of 2-(2-chloro-5-(2-methylprop-1-en-1-yl)pyridin-4-yl)-4-fluoro-1H-indole



embedded image


n-BuLi (3.15 mL, 7.88 mmol, 2.5 M in hexane) was added dropwise to a mixture of isopropyltriphenylphosphonium iodide (3.55 g, 8.21 mmol) in THF (30 mL) at −78° C. under N2 atmosphere. The mixture was stirred at −78° C. for 1 hour. Then a solution of 6-chloro-4-(4-fluoro-1H-indol-2-yl)nicotinaldehyde (900 mg, 3.28 mmol) in THF (10 mL) was added dropwise to the mixture at −78° C. The mixture was stirred at room temperature overnight. The mixture was then diluted with water (100 mL) and extracted with EA (50 mL*3). The organic layer was washed with brine (50 mL), dried over Na2SO4 and concentrated. The residue was purified by column chromatography (PE:EA=4:1) to give the product of 2-(2-chloro-5-(2-methylprop-1-en-1-yl)pyridin-4-yl)-4-fluoro-1H-indole (500 mg, yield: 50.6%). 1H-NMR (CDCl3, 400 MHz) δ 8.66 (br s, 1H), 8.17 (s, 1H), 7.51 (s, 1H), 7.12 (br s, 2H), 6.96 (d, J=1.6 Hz, 1H), 6.70˜6.81 (m, 1H), 6.22 (br s, 1H), 1.97 (s, 3H), 1.68˜1.70 (m, 1H), 1.74 (s, 3H). MS (M+H)+: 301/303.


Step 4—Synthesis of 5-(4-(4-fluoro-1H-indol-2-yl)-5-(2-methylprop-1-en-1-yl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a degassed solution of 2-(2-chloro-5-(2-methylprop-1-en-1-yl)pyridin-4-yl)-4-fluoro-1H-indole (230 mg, 0.767 mmol) and 2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide (322 mg, 0.767 mmol) in 1,4-dioxane/H2O (4 mL/1 mL), Pd2(dba)3 (30 mg, 0.038 mmol), X-Phos (36 mg, 0.076 mmol) and K3PO4 (612 mg, 2.301 mmol) were added under N2 atmosphere. The mixture was stirred at 90° C. for 3 hours under N2 atmosphere. The reaction mixture was then diluted with water (100 mL) and extracted with EA (50 mL×3). The organic layer was washed with brine (10 mL), dried over Na2SO4. After concentrated, the residue was purified by column chromatography (PE:EA=2:1) to give the product of 5-(4-(4-fluoro-1H-indol-2-yl)-5-(2-methylprop-1-en-1-yl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (400 mg, yield: 81.5%). 1H-NMR (CDCl3, 400 MHz) δ 9.67 (br s, 1H), 8.61 (s, 1H), 8.20 (s, 1H), 8.09 (s, 1H), 8.00˜8.06 (m, 2H), 7.54 (s, 1H), 7.11˜7.24 (m, 4H), 6.99 (s, 1H), 6.75˜6.83 (m, 1H), 6.39 (br s, 1H), 6.01 (br s, 1H), 3.22 (s, 3H), 2.99˜3.06 (m, 6H), 2.07 (s, 3H), 1.93 (s, 3H). MS (M+H)+: 641.


Step 5—Synthesis of 5-(11-fluoro-6,6-dimethyl-5,6-dihydroindolo[2,1-a][2,6]naphthyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


A mixture of 5-(4-(4-fluoro-1H-indol-2-yl)-5-(2-methylprop-1-en-1-yl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (100 mg, 0.156 mmol) and K3PO4 (166 mg, 0.781 mmol) in DMAc (0.8 mL) was stirred at 100° C. overnight. The mixture was then diluted with water (40 mL) and extracted with EA (20 mL*3). The organic layer was washed with brine (30 mL*3), dried over Na2SO4 and concentrated. The residue was purified by prep-TLC (DCM:EA=1:1) to afford the product of 5-(11-fluoro-6,6-dimethyl-5,6-dihydroindolo[2,1-a][2,6]naphthyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (40 mg, yield: 40.0%). 1H-NMR (CDCl3, 400 MHz) δ 8.49 (s, 1H), 7.86˜8.00 (m, 4H), 7.61 (s, 1H), 7.45 (d, J=8.0 Hz, 1H), 7.00˜7.18 (m, 4H), 6.72 (t, J=8.4 Hz, 1H), 6.01 (br s, 1H), 3.16 (s, 3H), 3.08 (br s, 2H), 2.93 (d, J=4.4 Hz, 3H), 2.77 (s, 3H), 1.69 (br s, 6H). MS (M+H)+: 641.


Example 65



embedded image


Step 1—Synthesis of 4-(tributylstannyl)but-3-en-1-ol



embedded image


A stirred mixture of but-3-yn-1-ol (7.00 g, 99.87 mmol) and AIBN (0.50 g, 2.81 mmol), contained in large round-bottomed flask equipped with a reflux condenser, was degassed with N2 at 25° C. and treated with Bu3SnH (40.4 mL, 150 mmol). The mixture was slowly heated to 90° C. and stirred at 100° C. for 16 hours. The solution was cooled and the crude product was subjected to chromatography on Al2O3 gel with a 1-10% gradient of EtOAc in PE. The mixture of E-isomer and Z-isomer were obtained as colorless oil (15 g, 40.9%).


Step 2—Synthesis of 4-(6-chloro-2-(4-fluoro-1H-indol-2-yl)pyridin-3-yl)but-3-en-1-ol



embedded image


To a N2 degassed solution of 4-(tributylstannyl)but-3-en-1-ol (1.2 g, 3.32 mmol), 6-chloro-2-(4-fluoro-1H-indol-2-yl)pyridin-3-yltrifluoromethanesulfonate (1.19 g, 3.02 mmol) and LiCl (128 mg, 3.02 mmol) in THF (10 mL) was added Pd(PPh3)4 (100 mg) under N2. The reaction mixture was stirred at 60° C. for 15 hours. To the mixture, H2O was added and the mixture was extracted with EtOAc. The combined organic phase was washed with brine, dried over Na2SO4. The crude product was purified by column (PE:EA=10:1 to 4:1) to give the mixture of (Z and E)-4-(6-chloro-2-(4-fluoro-1H-indol-2-yl)pyridin-3-yl)but-3-en-1-ol (530 mg, yield: 50%).


Step 3—Synthesis of (E)-5-(6-(4-fluoro-1H-indol-2-yl)-5-(4-hydroxybut-1-en-1-yl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


A solution of 4-(6-chloro-2-(4-fluoro-1H-indol-2-yl)pyridin-3-yl)but-3-en-1-ol (500 mg, 1.58 mmol), (2-(4-fluorophenyl)-3-(methylcarbamoyl)-6-(N-methylmethyl sulfonamido)benzofuran-5-yl)boronic acid (800 mg, 1.89 mmol) and Na2CO3 (340 mg, 3.15 mmol) in dioxane/DMF/H2O (5 mL/1 mL/0.2 mL) was added Pd2(dba)3 (10 mg) and X-Phos (10 mg) under N2. It was put into a pre-heated oil-bath at 110° C. for 8 hours. The mixture was concentrated and it was extracted with EtOAc. The combined organic phase was washed with brine, and dried over Na2SO4. The crude product was purified by column (PE:EA=1:1) to give the pure product of (Z and E)-5-(6-(4-fluoro-1H-indol-2-yl)-5-(4-hydroxybut-1-en-1-yl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (570 mg, yield: 52%).


Step 4—Synthesis of 5-(11-fluoro-6-(2-hydroxyethyl)-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


A solution of (Z and E)-5-(6-(4-fluoro-1H-indol-2-yl)-5-(4-hydroxybut-1-en-1-yl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (300 mg, 0.457 mmol) in DMAc (9 mL) was added K3PO4.3H2O (606 mg, 2.28 mmol) under N2. It was put into a pre-heated oil-bath at 110° C. for 3 hours. The mixture was diluted with H2O and it was extracted with EtOAc (500 mL×5). The combined organic phase was washed with brine, and dried over Na2SO4. The crude product was purified with Prep-HPLC to give the pure product of 5-(11-fluoro-6-(2-hydroxyethyl)-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (100 mg, yield: 30%). 1H-NMR (CDCl3, 400 MHz) δ 7.97 (s, 1H), 7.88˜7.92 (m, 2H), 7.63 (s, 1H), 7.60 (s, 1H), 7.40 (d, J=8.0 Hz, 1H), 7.09˜7.22 (m, 5H), 6.72 (t, J=8.4 Hz, 1H), 5.83 (s, 1H), 5.02 (d, J=6.4 Hz, 1H), 3.43˜3.55 (m, 4H), 3.35 (s, 3H), 3.10 (d, J=24.0 Hz, 1H), 2.94 (d, J=4.4 Hz, 3H), 2.61 (s, 3H), 1.78 (d, J=5.2 Hz, 2H). MS (M+H)+: 657.


Example 66



embedded image


Step 1—Synthesis of 2-(2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-6-yl)acetonitrile



embedded image


A mixture of (2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-6-yl)methyl methanesulfonate (1 g, 2.52 mmol) and KCN (491 mg, 7.55 mmol) in DMF (15 mL) was stirred at 80° C. for 15 hours. The mixture was diluted with water (30 mL) and extracted with EA (20 mL×3). The organic layer was washed with brine (30 mL), dried over Na2SO4 and concentrated. The residue was purified by column (PE:EA=1:1) to afford the desired product of 2-(2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-6-yl)acetonitrile (600 mg, yield: 73%). 1H-NMR (CDCl3, 400 MHz) δ 7.49 (d, J=8.0 Hz, 1H), 7.35 (s, 1H), 7.18 (d, J=8.0 Hz 2H), 7.09 (d, J=8.4 Hz, 1H), 6.77˜6.79 (m, 1H), 5.00˜5.06 (m, 1H), 3.48˜3.54 (m, 1H), 3.27 (d, J=16.4 Hz, 1H), 2.54˜2.59 (m, 1H), 2.37˜2.44 (m, 1H). MS (M+H)+: 312/314.


Step 2—Synthesis of 2-(2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-6-yl)acetaldehyde



embedded image


A solution of 2-(2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-6-yl)acetonitrile (300 mg, 0.78 mmol) in DCM (6 mL) was stirred at −78° C. for 10 minutes. Then DIBAL-H (1 ml, 1 mmol) was added slowly to the reaction mixture at −78° C. Then the mixture was heated at −78° C. for 2 hours. The mixture was diluted with water (30 mL) and extracted with EA (20 mL×3). The organic layer was washed with brine (30 mL), dried over Na2SO4 and concentrated. The residue was purified by column (PE:EA=3:1) to afford the desired product of 2-(2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-6-yl)acetaldehyde (200 mg, yield: 66%). 1H-NMR (CDCl3, 400 MHz) δ 9.63 (s, 1H), 7.44 (d, J=7.8 Hz, 1H), 7.34 (d, J=9.8 Hz, 1H), 7.11˜7.13 (m, 2H), 7.04 (d, J=8.4 Hz, 1H), 6.75 (t, J=8.0 Hz, 1H), 3.42 (s, 1H), 3.03 (d, J=8.4 Hz, 1H), 2.61˜2.70 (m, 2H). MS (M+H)+: 315/317.


Step 3—Synthesis of 2-chloro-11-fluoro-6-(2-(3-fluoroazetidin-1-yl)ethyl)-5,6-dihydroindolo[1,2-h][1,7]naphthyridine



embedded image


A mixture of 2-(2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-6-yl)acetaldehyde (200 mg, 0.617 mmol) and 3-fluoroazetidine (95 mg, 1.23 mmol) in DCM (4 mL) was stirred at room temperature for 30 minutes under N2. Then Na(OAc)3BH (50 mg) was added to the mixture, and the mixture was stirred at room temperature for 1 hour. After dilution with water (30 mL) and extraction with EA (20 mL×3), the organic layer was washed with brine (30 mL*3), dried over Na2SO4 and concentrated. The residue was purified by column chromatography (PE:EA=1:1) to afford the desired product of 2-chloro-11-fluoro-6-(2-(3-fluoroazetidin-1-yl)ethyl)-5,6-dihydroindolo[1,2-h][1,7]naphthyridine (100 mg, yield: 43%). 1H-NMR (CDCl3, 400 MHz) δ 7.44 (d, J=8.0 Hz, 1H), 7.3 (s, 1H), 7.16 (d, J=8.0 Hz, 1H), 7.10 (t, J=4.8 Hz, J=2.7 Hz, 2H), 6.69˜6.74 (m, 1H), 5.11˜5.14 (m, 0.5H), 4.97˜4.50 (m, 0.5H), 4.87 (t, J=4.8 Hz, 1H), 3.57 (d, J=6.8 Hz, 1H), 3.33˜3.39 (m, 1H), 2.95˜3.06 (m, 2H), 2.2˜72.38 (m, 1H). MS (M+H)+: 374/376.


Step 4—Synthesis of 5-(11-fluoro-6-(2-(3-fluoroazetidin-1-yl)ethyl)-5,6-dihydroindolo[1,2-][1,7]naphthyridin-2-yl)-N-methyl-6-(N-methylmethylsulfonamido)-2-(6-methylpyridin-3-yl)benzofuran-3-carboxamide



embedded image


A mixture of 2-chloro-11-fluoro-6-(2-(3-fluoroazetidin-1-yl)ethyl)-5,6-dihydroindolo[1,2-h][1,7]naphthyridine (100 mg, 0.269 mmol), N-methyl-6-(N-methylmethylsulfonamido)-2-(6-methylpyridin-3-yl)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide (150 mg, 0.295 mmol), K2CO3 (75 mg, 0.537 mmol), Pd2(dba)3 (15 mg, 0.027 mmol) and X-Phos (10 mg, 0.054 mmol) in dioxane/H2O (4 mL/10 d) was stirred at 110° C. for 4 hours under N2 atmosphere. The mixture was then filtered through Celite and concentrated. The residue was purified by column chromatography (PE:EA=2:1) to afford the desired product of 5-(11-fluoro-6-(2-(3-fluoroazetidin-1-yl)ethyl)-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-N-methyl-6-(N-methylmethylsulfonamido)-2-(6-methylpyridin-3-yl)benzofuran-3-carboxamide (85 mg, yield: 44.9%). 1H-NMR (CDCl3, 400 MHz) δ 8.97 (s, 1H), 8.17 (t, J=6.0, 2.0 Hz, 1H), 8.00 (s, 1H), 7.61 (t, J=8.0 Hz, 2H), 7.39 (d, J=8.0 Hz, 1H), 7.24 (d, J=6.8 Hz, 2H), 7.12 (s, 2H), 6.73˜6.77 (m, 1H), 5.97 (d, J=4.8 Hz, 1H), 4.97˜5.14 (m, 1H), 4.95 (d, J=6.0 Hz, 1H), 3.49˜3.56 (m, 4H), 3.33 (s, 3H), 3.00˜3.06 (m, 3H), 2.95 (d, J=4.8 Hz, 3H), 2.62 (s, 3H), 2.58 (s, 3H), 2.33˜2.36 (m, 1H), 1.59˜1.56 (m, 2H). MS (M+H)+: 711.


Examples 67-71

Examples 67 to 71, depicted in the table below, were prepared in accordance with the method described in Example 66.


















MS


Example
Structure
NMR
(M + H)+







67


embedded image


Same as compound 66
711





68


embedded image


Same as compound 66
711





69


embedded image



1H-NMR (CDCl3, 400 MHz) δ 8.02 (s, 1H), 7.96 (dd, J =5.6, 8.4 Hz, 2H), 7.63~7.69 (m, 2H), 7.53 (d, J = 7.6 Hz, 1H), 7.14~7.23 (m, 4H), 7.08 (s, 1H), 6.74~6.82 (m, 1H), 6.07~6.08 (m, 1H), 4.62~4.65 (m, 1H), 4.08~4.17 (m, 2H), 3.31 (s, 3H), 3.06 (dd, J = 6.0, 13.6 Hz, 1H), 2.97 (d, J = 4.8 Hz, 3H), 2.78 (s, 3H), 2.70 (dd, J = 6.8, 13.6 Hz, 1H).

643





70


embedded image



1H-NMR (CDCl3, 400 MHz) δ 8.98 (s, 1H), 8.18 (d, J = 8.4 Hz, 1H), 7.95 (s, 1H), 7.63 (d, J = 9.6 Hz, 2H), 7.50 (d, J = 7.6 Hz, 1H), 7.23 (d, J = 8.0 Hz, 1H), 7.12~7.14 (m, 1H), 7.05 (d, J = 8.0 Hz, 1H), 7.00 (s, 1H), 6.73 (t, J = 10.0 Hz, 1H), 6.12 (br d, J = 4.8 Hz, 1H), 5.20 (d, J = 5.2 Hz, 0.5H), 5.06 (t, J = 5.2 Hz, 0.5H), 3.46~3.89 (m, 2H), 3.70 (s, 3H), 3.56 (s, 3H), 3.25~3.68 (m, 5H), 2.94 (d, J = 4.4 Hz, 3H), 2.44~2.62 (m, 5H).

697





71


embedded image


Same as compound 71
697









Example 72



embedded image


Step 1—Synthesis of (E)-ethyl 3-(2,6-dichloropyridin-3-yl)acrylate



embedded image


To a solution of 2,6-dichloro-3-iodopyridine (3.0 g, 10.95 mmol), (E)-ethyl 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)acrylate (2.476 g, 10.95 mmol), and Dichloro[1,1′-bis(di-tert-butylphosphino)ferrocene]palladium (II) (0.357 g, 0.548 mmol) in THF (81 mL) was added 1M (aq) K2CO3 (54.8 mL, 54.8 mmol). A steady stream of N2 was bubbled through the resultant mixture for 5 minutes. Stirring was continued at ambient temperature for 2 h then the mixture was diluted with EtOAc and the organic layer washed successively with water (1×) and brine (1×). The organic layer was dried over MgSO4, filtered, and concentrated in vacuo. The residue was purified by silica gel column chromatography using a gradient of 0-30% EtOAc/Hexanes as eluent to give the title compound as a tan solid. 1H NMR δ (ppm) (CHCl3-d): 7.95 (1H, d, J=16 Hz), 7.90 (1H, d, J=8.1 Hz), 7.35 (1H, d, J=8.1 Hz), 6.47 (1H, d, J=16 Hz), 4.32 (2H, q, J=7.15 Hz), 1.38 (3H, t, J=7.14 Hz). MS (M+H)+: 245.


Step 2—Synthesis of ethyl 3-(2,6-dichloropyridin-3-yl)propanoate



embedded image


To a solution of (E)-ethyl 3-(2,6-dichloropyridin-3-yl)acrylate (500 mg, 2.032 mmol) in Ethanol (15 mL) was added 10% platinum on carbon (96 mg, 0.049 mmol). The flask was alternately evacuated and filled with hydrogen five times. The solution was stirred under 1 ATM (balloon) of hydrogen for 1 h, then filtered through celite, while washing well with MeOH and EtOAc. The filtrate was concentrated in vacuo to provide the title compound which was used without further purification. 1H NMR δ (ppm) (CHCl3-d): 7.62 (1H, d, J=7.92 Hz), 7.24 (1H, d, J=7.8), 4.15 (2H, q, J=7.15 Hz), 3.05 (2H, t, J=7.35 Hz), 2.69 (2H, t, J=7.35 Hz), 1.31˜1.24 (3H, m). MS (M+H)+: 247.


Step 3—Synthesis of ethyl 3-(2,6-dichloropyridin-3-yl)propan-1-ol



embedded image


To a solution of ethyl 3-(2,6-dichloropyridin-3-yl)propanoate in THF (12 mL) was added LiBH4 (88 mg, 4.03 mmol). The resultant mixture was stirred at ambient temperature for 2.5 h then was cooled to 0° C. and carefully quenched by addition of saturated NH4Cl(aq) solution. The reaction mixture was diluted with EtOAc and the organic layer washed successively with H2O (1×) and brine (1×). The organic extract was dried over MgSO4, filtered, and concentrated in vacuo. Purification by silica gel column chromatography using a gradient of 0-60% EtOAc/Hexanes as eluent provided the title compound. 1H NMR δ (ppm) (CHCl3-d): 7.58 (1H, d, J=7.92 Hz), 7.25 (1H, d, J=7.80), 3.73 (2H, q, J=5.48 Hz), 2.85 (2H, t, J=7.76 Hz), 1.95˜1.88 (2H, m), 1.42 (1H, m). MS (M+H)+: 205.


Step 4—Synthesis of 5-(6-chloro-5-(3-hydroxypropyl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a mixture of ethyl 3-(2,6-dichloropyridin-3-yl)propan-1-ol (180 mg, 0.873 mmol), 2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide (439 mg, 0.873 mmol), XantPhos Biphenyl precatalyst (78 mg, 0.087 mmol), and cesium carbonate (854 mg, 2.62 mmol) was added 1,4-Dioxane (7.3 mL) and water (1.5 mL). A steady stream of N2 was bubbled through the reaction mixture for 5 minutes then the mixture was heated to 70° C. for 4 h. The mixture was cooled to ambient temperature, diluted with EtOAc, and washed successively with H2O (1×) and brine (1×). The organic layer was dried over MgSO4, filtered, and concentrated in vacuo. The residue was purified by silica gel column chromatography using a gradient of 0-100% EtOAc/CH2Cl2 as eluent. Further purification by preparative TLC using 50% EtOAc/CH2Cl2 as eluent provided the title compound. 1H NMR δ (ppm) (CHCl3-d): 8.01˜7.96 (3H, m), 7.72 (1H, d, J=7.68 Hz), 7.68 (1H, s), 7.56 (1H, d, J=7.70 Hz), 7.23 (2H, t, J=8.6 Hz), 5.95 (1H, br s), 3.78 (2H, q, J=5.76 Hz), 3.24 (3H, s), 3.03 (3H, d, J=4.88 Hz), 2.93 (2H, t, J=7.76 Hz), 2.88 (3H, s), 2.03˜1.97 (2H, m). MS (M+H)+: 545.


Step 5—Synthesis of 5-(6-(4-fluoro-1H-indol-2-yl)-5-(3-hydroxypropyl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a mixture of 5-(6-chloro-5-(3-hydroxypropyl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (180 mg, 0.330 mmol), 4-fluoroindole-2-boronic acid pinacol ester (129 mg, 0.494 mmol), cesium carbonate (215 mg, 0.659 mmol) and dichloro[1,1′-bis(di-tert-butylphosphino)ferrocene]palladium (II) (21.49 mg, 0.033 mmol) was added 1,4-dioxane (3 mL) and H2O (0.3 mL). A steady stream of N2 was bubbled through the reaction mixture for 5 minutes then the mixture was heated to 90° C. for 3 h. The mixture was cooled to ambient temperature, diluted with EtOAc, and washed successively with H2O (1×) and brine (1×). The organic layer was dried over MgSO4, filtered, and concentrated in vacuo. The residue was purified by silica gel column chromatography using a gradient of 0-100% EtOAc/CH2Cl2 as eluent. The residue was triturated with CH2Cl2 to provide the title compound as a white solid. 1H NMR δ (ppm) (CHCl3-d): 10.30 (1H, br s), 8.07 (1H, s), 8.00 (2H, dd, J=8.56, 5.31 Hz), 7.78 (1H, d, J=7.88 Hz), 7.69 (1H, s), 7.50 (1H, d, J=7.85 Hz), 7.27˜7.23 (3H, m), 7.17˜7.13 (2H, m), 6.81˜6.78 (1H, m), 5.93˜5.87 (1H, m), 3.89 (2H, d, J=5.37 Hz), 3.24 (2H, t, J=7.86 Hz), 3.16 (3H, s), 3.02 (3H, d, J=4.90 Hz), 2.92 (3H, s), 2.13 (2H, t, J=7.43 Hz). MS (M+H)+: 645.


Step 6—Synthesis of 5-(12-fluoro-6,7-dihydro-5H-pyrido[2′,3′:3,4]azepino[1,2-a]indol-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a solution of 5-(6-(4-fluoro-1H-indol-2-yl)-5-(3-hydroxypropyl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (153 mg, 0.237 mmol) and triphenylphosphine (62.2 mg, 0.237 mmol) in THF was added diisopropyl azodicarboxylate (0.046 mL, 0.237 mmol). The resultant mixture was stirred at ambient temperature for 10 h then was concentrated in vacuo and the residue purified by silica gel column chromatography using a step gradient of 0-50-100% EtOAc/Hexanes as eluent. 1H NMR δ (ppm) (CHCl3-d): 8.07 (1H, s), 8.02 (2H, dd, J=8.61, 5.32 Hz), 7.75 (1H, d, J=7.78 Hz), 7.70 (1H, s), 7.60 (1H, d, J=7.77 Hz), 7.26˜7.0 (4H, m), 7.09 (1H, s), 6.85˜6.81 (1H, m), 5.98 (1H, br s), 4.23 (2H, t, J=6.66 Hz), 3.35 (3H, s), 3.03 (3H, d, J=4.90 Hz), 2.85 (2H, t, J=7.00 Hz), 2.80 (3H, s), 2.47˜2.42 (2H, m), MS (M+H)+: 627.


Example 73

Example 73, depicted in the table below, were prepared in accordance with the method described in Example 72.















Compound


MS


ID
Structure
NMR
(M + H)+







73


embedded image



1NMR δ (ppm)(CHCl3-d): 8.09 (1 H, s), 8.04-8.00 (2H, m), 7.77 (1H, d, J = 7.83 Hz), 7.68 (1H, s), 7.65 (2H, d, J = 7.90 Hz), 7.52 (1H, d, J = 7.34 Hz), 7.34-7.30 (1H, m), 7.25- 7.21 (3H, m) 5.96 (1H, br s) 4.27 (2H, t, J = 6.64 Hz), 3.36 (3H, s), 3.04 (3H, d, J = 4.91 Hz), 2.89-2.82 (5H, m), 2.50-2.43 (2H, m).

634









Example 74



embedded image


Step 1—Synthesis of ethyl 3-(2-chloro-6-(2-(4-fluorophenyl)-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)benzofuran-5-yl)pyridin-3-yl)propanoate



embedded image


To a mixture of ethyl 3-(2,6-dichloropyridin-3-yl)propanoate (170 mg, 0.685 mmol), 2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide (344 mg, 0.685 mmol), XantPhos Biphenyl precatalyst (60.9 mg, 0.069 mmol), and cesium carbonate (670 mg, 2.056 mmol) was added 1,4-dioxane (5.7 mL) and H2O (1.2 mL). A steady stream of N2 was bubbled through the reaction mixture for 5 minutes then the mixture was heated to 70° C. for 16 h. The mixture was cooled to ambient temperature, diluted with EtOAc, and washed successively with H2O (1×) and brine (1×). The organic layer was dried over MgSO4, filtered, and concentrated in vacuo. The residue was purified by silica gel column chromatography using a gradient of 0-100% EtOAc/Hexanes as eluent. 1H NMR δ (ppm) (CHCl3-d): 8.01˜7.96 (3H, m), 7.75 (1H, t, J=7.74 Hz), 7.69 (1H, s), 7.55 (1H, d, J=7.70 Hz), 7.27˜7.21 (2H, m) 5.91 (1H, br s), 4.19 (2H, q, J=7.12 Hz), 3.27˜3.20 (3H, m), 3.18˜3.12 (2H, m), 3.03 (3H, d, J=4.92 Hz), 2.90˜2.80 (3H, m), 2.77 (2H, t, J=7.51 Hz), 1.30 (3H, t, J=7.15 Hz). MS (M+H)+: 588.


Step 2—Synthesis of ethyl 3-(2-(4-fluoro-1H-indol-2-yl)-6-(2-(4-fluorophenyl)-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)benzofuran-5-yl)pyridin-3-yl)propanoate



embedded image


To a mixture of ethyl 3-(2-chloro-6-(2-(4-fluorophenyl)-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)benzofuran-5-yl)pyridin-3-yl)propanoate (120 mg, 0.204 mmol), 4-fluoro-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indole (80 mg, 0.306 mmol), cesium carbonate (133 mg, 0.408 mmol) and dichloro[1,1′-bis(di-t-butylphosphino)ferrocene]palladium (II) (13.3 mg, 0.02 mmol) was added 1,4-dioxane (1.86 mL) and H2O (0.186 mL). A steady stream of N2 was bubbled through the reaction mixture for 5 minutes then the mixture was heated to 90° C. for 9 h. The mixture was cooled to ambient temperature, diluted with EtOAc, and washed successively with H2O (1×) and brine (1×). The organic layer was dried over MgSO4, filtered, and concentrated in vacuo. The residue was purified by silica gel column chromatography using a gradient of 0-100% EtOAc/Hexanes as eluent and further purified by silica gel column chromatography using a step gradient of 0-20-80% EtOAc/CH2Cl2 as eluent. MS (M+H)+: 687.


Step 3—Synthesis of 3-(2-(4-fluoro-1H-indol-2-yl)-6-(2-(4-fluorophenyl)-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)benzofuran-5-yl)pyridin-3-yl)propanoic acid



embedded image


A mixture of ethyl 3-(2-(4-fluoro-1H-indol-2-yl)-6-(2-(4-fluorophenyl)-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)benzofuran-5-yl)pyridin-3-yl)propanoate (91 mg, 0.133 mmol) and lithium hydroxide monohydrate (27.8 mg, 0.663 mmol) were suspended in THF (0.53 mL), MeOH (0.27 mL), and H2O (0.2 mL). The resultant mixture was stirred at ambient temperature overnight then cooled in an ice bath and acidified to pH 2 with 1 N HCl. The mixture was then diluted with EtOAc and H2O, separated into layers and the aqueous layer extracted with EtOAc (2×). The combined organic layers were washed with brine (4×) until last brine wash was pH 7 then were dried over MgSO4, filtered, and concentrated in vacuo. The resulting product was azeotroped from CH3CN (4×) and used as a tan solid without further purification. MS (M+H)+: 659.


Step 4—Synthesis of 5-(12-fluoro-7-oxo-6,7-dihydro-5H-pyrido[2′,3′:3,4]azepino[1,2-a]indol-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a mixture of 3-(2-(4-fluoro-1H-indol-2-yl)-6-(2-(4-fluorophenyl)-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)benzofuran-5-yl)pyridin-3-yl)propanoic acid (88 mg, 0.133 mmol) and HATU (126 mg, 0.333 mmol) in DMF (6650 μl) was added DIEA (116 μl, 0.665 mmol). The resultant mixture was stirred for 1 h at ambient temperature then was diluted with EtOAc and washed successively with H2O (3×) and brine (1×). The organic layer was dried over MgSO4, filtered, concentrated in vacuo, and the residue purified by silica gel column chromatography using a gradient of 0-30% EtOAc/CH2Cl2 as eluent. 1H NMR δ (ppm) (CHCl3-d): 8.38 (1H, d, J=8.46 Hz), 8.12 (1H, s), 8.02 (2H, dd, J=8.55, 5.32 Hz), 7.69 (3H, d, J=9.74 Hz), 7.60 (1H, d, J=7.80 Hz), 7.36 (1H, td, J=8.24, 5.56 Hz), 7.25 (2H, t, J=8.60 Hz), 7.02 (1H, t, J=8.68 Hz), 5.95 (1H, br s), 3.35 (3H, s), 3.27˜3.22 (2H, m), 3.20˜3.15 (2H, m), 3.05 (3H, d, J=4.90 Hz), 2.85 (3H, s). MS (M+H)+: 641.


Example 75

Example 75, depicted in the table below, were prepared in accordance with the method described in Example 74.


















MS


Example
Structure
NMR
(M + H)+







75


embedded image



1H NMR δ (ppm)(CHCl3-d): 8.82 (1 H, d, J = 8.52 Hz), 8.12 (1 H, s), 8.05-7.99 (2 H, m), 7.82 (1 H, s), 7.72 (1 H, d, J = 7.87 Hz), 7.68-7.62 (3 H, m), 7.47 (1 H, t, J = 8.02 Hz), 7.24 (2 H, t, J = 8.65 Hz), 6.00 (1 H, br s), 3.40 (3 H, s), 3.31-3.25 (2 H, m), 3.23-3.17 (2 H, m), 3.05 (3 H, dd, J = 9.71, 4.93 Hz), 2.90-2.88 (3 H, m).

648









Example 76



embedded image


Step 1—Synthesis of (E)-6-bromo-2-chloro-3-(2-ethoxyvinyl)pyridine



embedded image


To a mixture of 6-bromo-2-chloro-3-iodopyridine (8.0 g, 25.1 mmol), (E)-2-(2-ethoxyvinyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (4.98 g, 25.1 mmol), cesium carbonate (16.38 g, 50.3 mmol), and [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium(II) (2.052, 2.51 mmol) was added 1,4-dioxane (100 mL) and H2O (10 mL). A steady stream of N2 was bubbled through the reaction mixture for 15 minutes then the mixture was heated to 73° C. for 20 h. The mixture was cooled to ambient temperature, diluted with EtOAc, and washed successively with H2O (2×) and brine (1×). The organic layer was dried over MgSO4, filtered, and concentrated in vacuo. The residue was purified by silica gel column chromatography using a step gradient of 0-10-60% EtOAc/Hexanes as eluent. MS (M+H): 261.


Step 2—Synthesis of (E)-5-(6-chloro-5-(2-ethoxyvinyl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a mixture of (E)-6-bromo-2-chloro-3-(2-ethoxyvinyl)pyridine (3.37 g, 12.84 mmol), 2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide (6.45 g, 12.84 mmol), cesium carbonate (10.46 g, 32.1 mmol), and 1,1′ Bis(diphenylphosphino)ferrocene-palladium(II)dichloride dichloromethane complex (0.524 g, 0.642 mmol) was added 1,4-Dioxane (83 mL) followed by H2O (8.34 mL). A steady stream of N2 was bubbled through the reaction mixture for 7 minutes then the mixture was heated to 60° C. for 16 h. The mixture was cooled to ambient temperature, diluted with EtOAc, and washed successively with H2O (1×) and brine (1×). The organic layer was dried over MgSO4, filtered, and concentrated in vacuo. The solid residue was triturated with EtOAc and further purified by silica gel column chromatography using a gradient of 0-30% EtOAc/CH2Cl2 as eluent. Further purification by silica gel column chromatography with a gradient of 0-100% EtOAc/Hexanes as eluent provided the title compound as a tan solid. 1H NMR δ (ppm) (CHCl3-d): 8.03˜7.97 (3H, m), 7.79 (1H, d, J=7.96 Hz), 7.69 (1H, s), 7.52 (1H, d, J=7.50 Hz), 7.28˜7.00 (2H, m), 7.11 (1H, d, J=13.00 Hz), 6.12 (1H, d, J=13.00 Hz), 5.91 (1H, br s), 4.04 (2H, q, J=7.04 Hz), 3.24 (3H, s), 3.03 (3H, d, J=4.87 Hz), 2.89 (3H, s), 1.44 (3H, t, J=7.00). MS (M+H): 557.


Step 3-Synthesis of (E)-tert-butyl 2-(3-(2-ethoxyvinyl)-6-(2-(4-fluorophenyl)-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)benzofuran-5-yl)pyridin-2-yl)-3-methyl-1H-indole-1-carboxylate



embedded image


To a mixture of (E)-5-(6-chloro-5-(2-ethoxyvinyl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (150 mg, 0.269 mmol), tert-butyl 3-methyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indole-1-carboxylate (144 mg, 0.403 mmol), cesium carbonate (175 mg, 0.538 mmol), and 1,1′-bis(di-tert-butylphosphino)ferrocene palladium dichloride (17.5 mg, 0.027 mmol) was added 1,4-dioxane (2.44 mL) and H2O (0.244 mL). A steady stream of N2 was bubbled through the reaction mixture for 5 minutes then the mixture was heated to 90° C. for 10 h. The mixture was cooled to ambient temperature, diluted with EtOAc, and washed successively with H2O (1×) and brine (1×). The organic layer was dried over MgSO4, filtered, and concentrated in vacuo. The residue was purified by silica gel column chromatography using a gradient of 0-100% EtOAc/Hexanes as eluent. MS (M+H): 753.


Step 4-Synthesis of tert-butyl 2-(6-(2-(4-fluorophenyl)-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)benzofuran-5-yl)-3-(2-hydroxyethyl)pyridin-2-yl)-3-methyl-1H-indole-1-carboxylate



embedded image


To a solution of (E)-tert-butyl 2-(3-(2-ethoxyvinyl)-6-(2-(4-fluorophenyl)-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)benzofuran-5-yl)pyridin-2-yl)-3-methyl-1H-indole-1-carboxylate (72 mg, 0.096 mmol) in THF (0.350 mL) at 0° C. was added Mercuric acetate (36.6 mg, 0.115 mmol) in H2O (0.350 mL). After stirring the resultant mixture at 0° C. for 30 min, NaBH4 (14.47 mg, 0.383 mmol) in 0.580 mL of saturated (aq) K2CO3 solution was added and the reaction mixture was warmed to ambient temperature. After 1.5 h at ambient temperature, the reaction mixture was diluted with EtOAc and washed successively with H2O (2×) and brine (1×). The organic layer was dried over MgSO4, filtered, and concentrated in vacuo. The residue was purified by silica gel column chromatography using a gradient of 0-90% EtOAc/CH2Cl2 as eluent. 1H NMR δ (ppm) (CHCl3-d): 8.16 (1H, d, J=8.33 Hz), 8.03˜7.99 (3H, m), 7.87 (1H, d, J=7.99 Hz), 7.69˜7.65 (2H, m), 7.57 (1H, d, J=7.77 Hz), 7.39 (1H, t, J=7.73 Hz), 7.33 (1H, t, J=7.42 Hz), 7.22 (2H, t, J=8.58 Hz), 5.95 (1H, br s), 3.81 (2H, m), 3.19 (3H, s), 3.00 (3H, d, J=4.88 Hz), 2.98˜2.83 (2H, m), 2.78 (3H, s), 2.11 (3H, s), 1.45 (6H, s), 1.27 (3H, s). MS (M+H): 727.


Step 5—Synthesis of 2-(4-fluorophenyl)-5-(5-(2-hydroxyethyl)-6-(3-methyl-1H-indol-2-yl)pyridin-2-yl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a solution of tert-butyl 2-(6-(2-(4-fluorophenyl)-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)benzofuran-5-yl)-3-(2-hydroxyethyl)pyridin-2-yl)-3-methyl-1H-indole-1-carboxylate (50 mg, 0.069 mmol) in CH2Cl2 (1.5 mL) was added TFA (1.5 mL). The resultant mixture was stirred at ambient temperature for 1.5 h then was concentrated in vacuo, diluted with EtOAc and washed successively with saturated NaHCO3 (aq) solution (1×) and brine (1×). The organic layer was dried over MgSO4, filtered, and concentrated in vacuo to give the title compound, which was used without further purification. MS (M+H): 627.


Step 6—Synthesis of 2-(4-fluorophenyl)-N-methyl-5-(12-methyl-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a mixture of 2-(4-fluorophenyl)-5-(5-(2-hydroxyethyl)-6-(3-methyl-1H-indol-2-yl)pyridin-2-yl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (43 mg, 0.069 mmol) and triphenylphosphine (90 mg, 0.343 mmol) in THF (4 mL) was diisopropyl azodicarboxylate (0.067 mL, 0.343 mmol). The resultant mixture was stirred at ambient temperature for 16 h, diluted with EtOAc and washed successively with H2O (2×) and brine (1×). The organic layer was dried over MgSO4, filtered, and concentrated in vacuo. The residue was purified by silica gel column chromatography using a step gradient of 0-40-90% EtOAc/Hexanes as eluent. Further purification by silica gel column chromatography with a gradient of 0-10% EtOAc/CH2Cl2 as eluent, followed by lyophilization from CH3CN/H2O provided the title compound. 1H NMR δ (ppm) (CHCl3-d): 8.16 (1H, s), 8.07˜8.03 (2H, m), 7.70˜7.66 (3H, m), 7.47 (1H, d, J=7.77 Hz), 7.37 (1H, d, J=8.24 Hz), 7.32 (1H, D, J=7.1 Hz), 7.26˜7.22 (2H, m), 7.16 (1H, t, J=7.40 Hz), 5.95 (1H, br s), 4.34 (2H, t, J=6.37 Hz), 3.32 (3H, s), 3.28 (2H, t, J=6.4 Hz), 3.05 (3H, d, J=4.91 Hz), 2.83 (3H, s), 2.75 (3H, s). MS (M+H): 609.


Examples 77 and 78

Examples 77 and 78, depicted in the table below, were prepared in accordance with the methods described above for Example 76.















Ex-





am-


MS


ple
Structure
NMR
(M + H)+







77


embedded image



1H NMR δ (ppm)(DMSO-d6): 8.58 (1 H, q, J = 4.63 Hz), 8.37 (1 H, dd, J = 4.55, 1.38 Hz), 8.06-8.01 (3 H, m), 7.99 (1 H, d, J = 8.34 Hz), 7.90 (1 H, d, J = 8.0 Hz), 7.86 (1 H, s), 7.58 (1 H, d, J = 7.88 Hz), 7.42 (2 H, t, J = 8.79 Hz), 7.22-7.18 (1 H, m), 7.16 (1 H, s), 4.42 (2 H, t, J = 6.62 Hz), 3.36-3.31 (5 H, m), 2.94 (3 H, s), 2.83 (3 H, d, J = 4.58 Hz)

596





78


embedded image



1H NMR δ (ppm)(DMSO-d6): 8.58-8.54 (1 H, m), 8.06-8.01 (3 H, m), 7.95 (1 H, d, J = 8.86 Hz), 7.87- 7.52 (3 H, m), 7.42 (2 H, t, J = 8.77 Hz), 7.00 (1 H, s), 6.67 (1 H, d, J = 8.82 Hz), 4.37 (2 H, t, J = 6.66 Hz), 3.87 (3 H, s), 3.33-3.29 (5 H, m), 2.92 (3 H, s), 2.83 (3 H, d, J = 4.58 Hz).

626









Example 79



embedded image


Step 1—Synthesis of 2-(4-fluorophenyl)-N-methyl-5-(12-methylindolo[1,2-h][1,7]naphthyridin-2-yl)-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a solution of (E)-tert-butyl 2-(3-(2-ethoxyvinyl)-6-(2-(4-fluorophenyl)-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)benzofuran-5-yl)pyridin-2-yl)-3-methyl-1H-indole-1-carboxylate (60 mg, 0.080 mmol) in 1,4-Dioxane (2 mL) was added 4 M HCl in dioxane (1 mL, 4.00 mmol). The resultant bright orange mixture was stirred at ambient temperature for 45 minutes then was heated at 60° C. for 1.75 h. The mixture was concentrated in vacuo, azeotroping from CH3CN, and the residue was diluted with saturated NaHCO3 (aq) solution and extracted into EtOAc. The organic layer was washed with brine (1×), dried over MgSO4, concentrated in vacuo, and purified by silica gel column chromatography using a gradient of 0-70% EtOAc/Hexanes as eluent. Lyophilization from CH3CN/H2O provided the title compound. 1H NMR δ (ppm) (CHCl3-d): 8.27 (1H, s), 8.14 (1H, d, J=7.34 Hz), 8.09˜8.05 (2H, m), 7.91˜7.84 (3H, m), 7.71˜7.68 (2H, m), 7.46˜7.43 (2H, m), 7.24 (2H, t, J=8.65 Hz), 6.61 (1H, d, J=7.33 Hz), 5.96 (1H, br s), 3.35 (3H, s), 3.06 (6H, m), 2.73 (3H, s). MS (M+H): 607.


Examples 80 and 81

Examples 80 and 81, depicted in the table below, were prepared in accordance with the methods described in Example 79.


















MS


Example
Structure
NMR
(M + H)+







80


embedded image



1H NMR δ (ppm)(DMSO-d6): 8.77 (1 H, d, J = 7.28 Hz), 8.61-8.57 (2 H, m), 8.26 (1 H, d, J = 8.17 Hz), 8.11 (1 H, s), 8.05 (2 H, dd, J = 8.48, 5.31 Hz), 7.95 (1 H, s), 7.81 (1 H, d, J = 8.18 Hz), 7.43 (2 H, t, J = 8.69 Hz), 7.37 (1 H, s), 7.10 (1 H, d, J = 7.30 Hz), 6.84 (1 H, d, J = 8.90 Hz), 3.96 (3 H, s), 3.40 (3 H, s), 2.90 (3 H, s), 2.84 (3 H, s)

624





81


embedded image



1H NMR δ (ppm)(DMSO-d6): 11.75 (1 H, br s), 8.66 (1 H, d, J = 7.53 Hz), 8.60 (1 H, d, J = 5.72 Hz), 8.38 (1 H, d, J = 9.72 Hz), 8.25 (1 H, d, J = 8.31 Hz), 8.09-7.96 (3 H, m), 7.79 (1 H, d, J = 8.36 Hz), 7.43 (3 H, t, J = 8.68 Hz), 7.14 (1 H, d, J = 7.33 Hz), 6.96 (1 H, s), 6.24 (1 H, d, J = 9.83 Hz), 2.97-2.91 (3 H, m), 2.87-2.80 (3 H, m)

610









Examples 82 and 82′



embedded image


Step 1—Synthesis of (E)-2,6-dichloro-3-(2-ethoxyvinyl)pyridine



embedded image


To a degassed solution of 2,6-dichloro-3-iodopyridine (3.0 g, 11 mmol) and (E)-2-(2-ethoxyvinyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane (2.2 g, 11 mmol) in 1,4-dioxane (20 mL) and water (1.0 mL) was added CS2CO3 (7.1 g, 22 mmol) and 1,1′-bis(di-tert-butylphosphino)ferrocene palladium chloride (357 mg, 0.54 mmol) under N2 protection. The resulting mixture was heated to 70° C. and stirred at this temperature overnight. The reaction was cooled, filtered through a pad of the celite and washed with ethyl acetate. The combined filtrate was evaporated in vacuo. The resulting residue was purified using column chromatography (eluted with 0-20% EtOAc/DCM) to provide (E)-2,6-dichloro-3-(2-ethoxyvinyl)pyridine (1.96 mg, yield: 86%). MS (M+H)+: 218.


Step 2—Synthesis of (E)-5-(6-chloro-5-(2-ethoxyvinyl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a degassed solution of (E)-2,6-dichloro-3-(2-ethoxyvinyl)pyridine (500 mg, 2.29 mmol) and 2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide (1.15 g, 2.29 mmol) in 1,4-dioxane (8 mL) and water (200 μL) was added CS2CO3 (1.49 g, 4.59 mmol) and 1,1′-bis(di-tert-butylphosphino)ferrocene palladium chloride (120 mg, 0.18 mmol) under N2 protection. The resulting mixture was heated to 90° C. and stirred at this temperature overnight. The reaction was cooled, filtered through a pad of the celite and washed with ethyl acetate. The combined filtrate was evaporated in vacuo. The resulting residue was purified using column chromatography (eluted with 0-30% EtOAc/DCM) to provide (E)-5-(6-chloro-5-(2-ethoxyvinyl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (200 mg, yield: 16%). MS (M+H)+: 558.


Step 3—Synthesis of (E)-5-(5-(2-ethoxyvinyl)-6-(4-fluoro-1H-indol-2-yl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a degassed solution of (E)-5-(6-chloro-5-(2-ethoxyvinyl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (200 mg, 0.36 mmol) and 4-fluoro-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indole (140 mg, 0.54 mmol) in 1,4-dioxane (3 mL) and water (200 μl) was added CS2CO3 (234 mg, 0.72 mmol) and 1,1′-bis(di-tert-butylphosphino)ferrocene palladium chloride (24 mg, 0.04 mmol) under N2 protection. The resulting mixture was heated to 90° C. and stirred at this temperature for 3 hours. The reaction was cooled, filtered through a pad of the celite and washed with ethyl acetate. The combined filtrate was evaporated in vacuo. Preparative TLC (eluted with 0-30% acetone/hexane) provided (E)-5-(5-(2-ethoxyvinyl)-6-(4-fluoro-1H-indol-2-yl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (65 mg, yield: 28%) MS (M+H)+: 657.


Step 4—Synthesis of compound 82 and 82′



embedded image


To a solution of (E)-5-(5-(2-ethoxyvinyl)-6-(4-fluoro-1H-indol-2-yl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (70 mg, 0.11 mmol) in acetonitrile (4 ml) was added NaI (80 mg, 0.53 mmol) and TMSCl (58 mg, 0.53 mmol). The resulting mixture was stirred at RT for 3 hours, then treated with NaHCO3 and extracted with EtOAc. The organic layer was washed with brine, dried (Na2SO4), filtered and concentrated in vacuo. Preparative TLC gave 5-(11-fluoroindolo[1,2-h][1,7]naphthyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (compound 82, 22 mg, yield: 34%) and 5-(7-fluoro-11H-pyrido[2,3-a]carbazol-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (compound 82′, 10 mg, yield: 15%).


Examples 83 and 84

Examples 83 and 84, depicted in the table below, were prepared in accordance with the methods described above for Example 82.














Example
Structure
MS M + H)+







83


embedded image


617





84


embedded image


628









Example 85



embedded image


Step 1—Synthesis of 2-(2,6-dichloropyridin-3-yl)acetaldehyde



embedded image


To a solution of (E)-2,6-dichloro-3-(2-ethoxyvinyl)pyridine (890 mg, 4.08 mmol) in acetonitrile (10 ml) was added NaI (1.84 g, 12.2 mmol) and TMSCl (887 mg, 8.16 mmol). The resulting mixture was stirred at RT for 4 hours, then treated with NaHCO3 and extracted with EtOAc. The organic layer was washed with brine, dried (Na2SO4), filtered and concentrated in vacuo to give the yellow oil 2-(2,6-dichloropyridin-3-yl)acetaldehyde (760 mg, yield: 98%). MS (M+H)+: 190.


Step 2—Synthesis of 2-(2,6-dichloropyridin-3-yl)ethanol



embedded image


NaBH4 (80 mg, 2.11 mmol) was added to a solution of 2-(2,6-dichloropyridin-3-yl)acetaldehyde (200 mg, 1.05 mmol) in THF (1 ml) and MeOH (1 ml) and stirred at RT for 1 hour. The resulting mixture was treated with NH4Cl and extracted with EtOAc. The organic layer was washed with brine, dried (Na2SO4), filtered and concentrated in vacuo to give the yellow oil 2-(2,6-dichloropyridin-3-yl)ethanol (200 mg, yield: 99%)


Step 3—Synthesis of 2-(6-chloro-2-(4-fluoro-1H-indol-2-yl)pyridin-3-yl)ethanol



embedded image


To a degassed solution of 2-(2,6-dichloropyridin-3-yl)ethanol (200 mg, 1.04 mmol) and 4-fluoro-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indole (272 mg, 1.04 mmol) in 1,4-dioxane (8 mL) and water (200 μl) was added CS2CO3 (679 mg, 2.08 mmol) and 1,1′-bis(di-tert-butylphosphino)ferrocene palladium chloride (68 mg, 0.10 mmol) under N2 protection. The resulting mixture was heated to 90° C. and stirred at this temperature overnight. The reaction was cooled, filtered through a pad of the celite and washed with ethyl acetate. The combined filtrate was evaporated in vacuo. Preparative TLC (eluted with 20% EtOAc/DCM) to provide 2-(6-chloro-2-(4-fluoro-1H-indol-2-yl)pyridin-3-yl)ethanol (60 mg, yield: 19%) MS (M+H)+: 291.


Step 4—Synthesis of 2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridine



embedded image


2-(6-chloro-2-(4-fluoro-1H-indol-2-yl)pyridin-3-yl)ethanol (60 mg, 0.21 mmol) was dissolved in THF (4 ml). To the resulting mixture was added diisopropyl azodicarboxylate (42 mg, 0.21 mmol) and PPh3 (54 mg, 0.21 mmol). The resulting mixture was stirred at RT overnight. Preparative TLC gave 2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridine (50 mg, yield: 89%) MS (M+H)+: 273.


Step 5—Synthesis of 5-(11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a degassed solution of 2-chloro-11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridine (50 mg, 0.18 mmol) and 2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide (120 mg, 0.24 mmol) in 1,4-dioxane (8 mL) and water (200 μl) was added CS2CO3 (119 mg, 0.37 mmol) and 1,1′-bis(di-tert-butylphosphino)ferrocene palladium chloride (20 mg, 0.03 mmol) under N2 protection. The resulting mixture was heated to 90° C. and stirred for 4 hours. The reaction was cooled, filtered through a pad of the celite and washed with ethyl acetate. The combined filtrate was evaporated in vacuo. Preparative TLC (eluted with 30% EtOAc/DCM) provide 5-(11-fluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (90 mg, yield: 80%) MS (M+H)+: 613.


Example 86

Example 86, depicted in the table below, was prepared in accordance with the methods described above for Example 85.














Compound ID
Structure
MS M + H)+







86


embedded image


619









Example 87



embedded image


Step 1—Synthesis of 5-(6-chloro-5-formylpyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a degassed solution of 2,6-dichloronicotinaldehyde (315 mg, 1.79 mmol) and 2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide (600 mg, 1.19 mmol) in 1,4-dioxane (8 mL) and water (300 μL) was added K2CO3 (330 mg, 2.40 mmol) and Pd(dppf)Cl2 (175 mg, 0.24 mmol) under N2 protection. The resulting mixture was heated to 80° C. and stirred at this temperature overnight. The reaction was cooled, filtered through a pad of celite and washed with ethyl acetate. The combined filtrate was evaporated in vacuo. The resulting residue was purified using column chromatography (eluted with 0-30% EtOAc/DCM) to provide 5-(6-chloro-5-formylpyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (530 mg, yield: 86%) MS (M+H)+: 516.


Step 3—Synthesis of 5-(10-fluoro-5-hydroxy-5H-pyrido[2′,3′:3,4]pyrrolo[1,2-a]indol-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a degassed solution of 5-(6-chloro-5-formylpyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (150 mg, 0.29 mmol) and 4-fluoro-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indole (94 mg, 0.35 mmol) in 1,4-dioxane (3 mL) and water (200 μL) was added CS2CO3 (189 mg, 0.58 mmol) and 1,1′-bis(di-tert-butylphosphino)ferrocene palladium chloride (30 mg, 0.05 mmol) under N2 protection. The resulting mixture was heated to 90° C. and stirred for 3 hours. The reaction was cooled, filtered through a pad of celite and washed with ethyl acetate. The combined filtrate was evaporated in vacuo. Preparative TLC (eluted with (30% ethyl acetate/DCM) to provide 5-(10-fluoro-5-hydroxy-5H-pyrido[2′,3′:3,4]pyrrolo[1,2-a]indol-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (40 mg, yield: 22%) MS (M+H)+: 615.


Example 88



embedded image


Step 1—Synthesis of 5-(6-chloro-5-(hydroxymethyl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a solution of 5-(6-chloro-5-formylpyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (300 mg, 0.58 mmol) in THF (4 ml) and MeOH (1 ml) was added NaBH4 (66 mg, 1.74 mmol). The resulting mixture was stirred at RT for 3 hours then treated with NH4Cl and extracted with EtOAc. The organic layer was washed with brine, dried (Na2SO4), filtered and concentrated in vacuo to give 5-(6-chloro-5-(hydroxymethyl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (300 mg, yield: 100%). MS (M+H)+: 518.


Step 2—Synthesis of 5-(6-(4-fluoro-1H-indol-2-yl)-5-(hydroxymethyl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a degassed solution of 5-(6-chloro-5-(hydroxymethyl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (300 mg, 0.58 mmol) and 4-fluoro-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indole (197 mg, 0.75 mmol) in 1,4-dioxane (5 mL) and water (300 μL) was added Cs2CO3 (377 mg, 1.16 mmol) and 1,1′-bis(di-tert-butylphosphino)ferrocene palladium chloride (38 mg, 0.06 mmol) under N2 protection. The resulting mixture was heated to 80° C. and stirred at this temperature overnight. The reaction was cooled, filtered through a pad of celite and washed with ethyl acetate. The combined filtrate was evaporated in vacuo. Preparative TLC (eluted with 0-30% acetone/hexane) to provide 5-(6-(4-fluoro-1H-indol-2-yl)-5-(2-hydroxyethyl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (280 mg, yield: 78%) MS (M+H)+: 617.


Step 3—Synthesis of 5-(10-fluoro-5H-pyrido[2′,3′:3,4]pyrrolo[1,2-a]indol-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


5-(6-(4-fluoro-1H-indol-2-yl)-5-(hydroxymethyl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (280 mg, 0.45 mmol) was dissolved in THF (8 mL). To the resulting mixture was added diisopropyl azodicarboxylate (184 mg, 0.91 mmol) and PPh3 (238 mg, 0.91 mmol). The resulting mixture was stirred at RT overnight. Preparative TLC gave 5-(10-fluoro-5H-pyrido[2′,3′:3,4]pyrrolo[1,2-a]indol-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (230 mg, yield: 85%) MS (M+H)+: 599.


Example 89



embedded image


Step 1—Synthesis of 2-(benzyloxy)-1-(6-bromo-2-chloropyridin-3-yl)ethanol



embedded image


6-Bromo-2-chloro-3-iodopyridine (1.50 g, 4.71 mmol) was dissolved in THF (15 mL) and cooled to −78° C., followed by addition of isopropylmagnesium chloride-lithium chloride complex (3.99 mL, 5.18 mmol). The mixture was stirred at −78° C. for 1 h. 2-(benzyloxy)acetaldehyde (0.849 g, 5.65 mmol) was added and the resulting mixture warmed to 0° C. and stirred for 2 h. The mixture was treated with saturated NH4Cl and extracted with ethyl acetate. The organic layer was washed with brine, dried (Na2SO4), filtered and concentrated in vacuo to gave 2-(benzyloxy)-1-(6-bromo-2-chloropyridin-3-yl)ethanol (1.54 g, yield: 95%). MS (M+H)+: 344.


Step 2—Synthesis of 3-(2-(benzyloxy)-1-fluoroethyl)-6-bromo-2-chloropyridine



embedded image


To a solution of 2-(benzyloxy)-1-(6-bromo-2-chloropyridin-3-yl)ethanol (1.54 g, 4.49 mmol) in DCM (10 mL) at 0° C. was added diethylaminosulfur trifluoride (0.594 ml, 4.49 mmol). The resulting mixture was stirred at 0° C. for 30 min, then warmed to RT and stirred for 1 h. The mixture was concentrated in vacuo and the resulting residue purified using column chromatography (eluted with 0-20% ethyl acetate/hexane) to provide 3-(2-(benzyloxy)-1-fluoroethyl)-6-bromo-2-chloropyridine (490 mg, yield: 32%)


Step 3—Synthesis of 5-(5-(2-(benzyloxy)-1-fluoroethyl)-6-chloropyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a degassed solution of 2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzofuran-3-carboxamide (714 mg, 1.422 mmol) and 3-(2-(benzyloxy)-1-fluoroethyl)-6-bromo-2-chloropyridine (490 mg, 1.422 mmol) in 1,4-Dioxane (15 ml) and water (2 mL) was added K2CO3 (393 mg, 2.84 mmol) and Pd(dppf)Cl2 (100 mg, 0.137 mmol) under N2 protection. The resulting mixture was heated to 85° C. and stirred for 6 h. The reaction was cooled, filtered through a pad of celite and washed with ethyl acetate. The combined filtrate was evaporated in vacuo. The resulting residue was purified using column chromatography (eluted with 0-20% ethyl acetate/DCM) to provide 5-(5-(2-(benzyloxy)-1-fluoroethyl)-6-chloropyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (660 mg, yield: 67%). MS (M+H)+: 641.


Step 4—Synthesis of 5-(5-(2-(benzyloxy)-1-fluoroethyl)-6-chloropyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a degassed solution of 5-(5-(2-(benzyloxy)-1-fluoroethyl)-6-chloropyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (610 mg, 0.953 mmol) and 4-fluoro-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indole (373 mg, 1.429 mmol) in 1,4-Dioxane (8 mL) and water (300 μL) was added Cs2CO3 (621 mg, 1.906 mmol) and DTBPF PdCl2 (60 mg, 0.092 mmol) under N2 protection. The resulting mixture was heated to 85° C. and stirred overnight. The reaction was cooled, filtered through a pad of celite and washed with ethyl acetate. The combined filtrate was evaporated in vacuo and the resulting residue was purified using column chromatography (eluted with 0-20% ethyl acetate/DCM) to provide 5-(5-(2-(benzyloxy)-1-fluoroethyl)-6-chloropyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (530 mg, yield: 75%). MS (M+H)+: 739.


Step 5—Synthesis of 5-(6-(4-fluoro-1H-indol-2-yl)-5-(1-fluoro-2-hydroxyethyl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a solution of 5-(5-(2-(benzyloxy)-1-fluoroethyl)-6-(4-fluoro-1H-indol-2-yl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (530 mg, 0.717 mmol) in THF (10 mL) was added palladium hydroxide (101 mg, 0.717 mmol). The mixture was hydrogenated under a balloon for 2 h. Filtration through a pad of celite removed the solid. After washing with ethyl acetate, the combined filtrate was concentrated in vacuo to give 5-(6-(4-fluoro-1H-indol-2-yl)-5-(1-fluoro-2-hydroxyethyl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (465 mg, yield: 100%). MS (M+H)+: 649.


Step 6—Synthesis of 5-(5,11-difluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a solution of 5-(6-(4-fluoro-1H-indol-2-yl)-5-(1-fluoro-2-hydroxyethyl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (465 mg, 0.717 mmol) and triphenylphosphine (282 mg, 1.075 mmol) in DCM (15 mL) was added diisopropyl azodicarboxylate (217 mg, 1.075 mmol). The resulting mixture was stirred at RT overnight and concentrated in vacuo. The resulting residue was purified using column chromatography (eluted with 20-50% ethyl acetate/hexane) to provide 5-(5,11-difluoro-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (250 mg, yield: 55%). MS (M+H)+: 631.


Example 90



embedded image




embedded image


To a solution of 5-(11-fluoro-6-(hydroxymethyl)-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (180 mg, 0.280 mmol) in DCM (8 mL) at 0° C. was added trifluoromethanesulfonic anhydride in DCM (0.560 mL, 0.560 mmol). The mixture was stirred at 0° C. for 2 h, then at RT for 1 h. The mixture was evaporated in vacuo to remove the volatiles, redissolved in DMF (8 mL), followed by addition of N,N-diisopropyl ethylamine (362 mg, 2.80 mmol) and 3-fluoroazetidine hydrochloride (125 mg, 1.120 mmol). The mixture was heated to 60° C. and stirred for 3 h. 20 ml H2O was added and the mixture stirred for 10 min. Filtration collected the yellow solid. After washing with additional H2O, the yellow solid was purified by chromatography, and eluted with 0-70% ethyl acetate in hexane to gave 5-(11-fluoro-6-((3-fluoroazetidin-1-yl)methyl)-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (42 mg, yield: 21%). MS (M+H)+: 700.


Examples 91-95

Examples 91-95, depicted in the table below, were prepared in accordance with the methods described above for Example 90.














Example
Structure
MS M+ H)+







91


embedded image


717





92


embedded image


681





93


embedded image


713





94


embedded image


713





95


embedded image


718









Example 96



embedded image




embedded image


5-(11-fluoro-6-(hydroxymethyl)-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (40 mg, 0.062 mmol) was dissolved in DCM (2 ml) and cooled to 0° C., followed by addition of DAST (0.025 ml, 0.187 mmol). The mixture was stirred at 0° C. for 1 h, then at RT for 30 min and then concentrated in vacuo. Preparative TLC (50% DCM in ethyl acetate) gave 5-(11-fluoro-6-(fluoromethyl)-5,6-dihydroindolo[1,2-h][1,7]naphthyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (14 mg, yield: 35%). MS (M+H)+: 645.


Example 97



embedded image


Step 1—Synthesis of (6-bromo-2-chloropyridin-3-yl)methanol



embedded image


To a solution of 6-bromo-2-chloronicotinaldehyde (5.40 g, 24.50 mmol) in THF (30 ml) and MeOH (10.0 ml) at 0° C. was added NaBH4 (0.927 g, 24.50 mmol). The mixture was stirred at 0° C. for 2 h. Saturated NH4Cl (50 ml) was added and the mixture extracted with ethyl acetate, dried over (Na2SO4), filtered and concentrated in vacuo to give (6-bromo-2-chloropyridin-3-yl)methanol (5.50 mg, yield: 101%). MS (M+H)+: 224.


Step 2—Synthesis of 2-(6-bromo-2-chloropyridin-3-yl)acetonitrile



embedded image


To a solution of (6-bromo-2-chloropyridin-3-yl)methanol (5.45 g, 24.50 mmol) and Et3N (5.12 mL, 36.7 mmol) at 0° C. was added MsCl (2.081 mL, 26.9 mmol). The resulting mixture was stirred at 0° C. for 2 h. 30 ml diethyl ether was added and the mixture stirred for 20 min. Filtration removed the yellow solid. After washing with 2×20 ml diethyl ether, the combined filtrate was concentrated in vacuo to give the yellow solid.


The above yellow solid was dissolved in 20 ml DMSO, followed by addition of NaCN (3.72 g, 36.7 mmol). The mixture was stirred at RT for 20 hours. 100 ml H2O was added and the mixture extracted with ethyl acetate, dried over (Na2SO4), filtered and concentrated in vacuo. The resulting residue was purified using column chromatography (eluted with 0-20% ethyl acetate in hexane) gave 2-(6-bromo-2-chloropyridin-3-yl)acetonitrile (3.8 g, yield: 67%)


Step 3—Synthesis of 1-(6-bromo-2-chloropyridin-3-yl)cyclopropanecarbonitrile



embedded image


To a solution of NaOH (2.59 g, 64.8 mmol) in water (6 mL) was added 2-(6-bromo-2-chloropyridin-3-yl)acetonitrile (1.5 g, 6.48 mmol), tetrabutyl ammonium bromide (0.418 g, 1.296 mmol) and 1-bromo-2-chloroethane (1.079 mL, 12.96 mmol). The resulting mixture was heated to 50° C. and stirred for 2 h, extracted with ethyl acetate (2×30 mL), dried over Na2SO4, filtered and concentrated in vacuo. The residue was purified using column chromtography (eluted with 0-20% ethyl acetate in hexane) to give 1-(6-bromo-2-chloropyridin-3-yl)cyclopropanecarbonitrile (850 mg, yield: 51%) MS (M+H)+: 258.


Step 4—Synthesis of (1-(6-bromo-2-chloropyridin-3-yl)cyclopropyl)methanol



embedded image


To a solution of 1-(6-bromo-2-chloropyridin-3-yl)cyclopropane carbonitrile (850 mg, 3.30 mmol) in THF (15 mL) at RT was added DIBAL-H (5.08 mL, 6.60 mmol) and stirred for 1 h. 50 ml saturated NH4Cl was and the mixture extracted with ethyl acetate, dried over Na2SO4, filtered and concentrated in vacuo.


The above residue was dissolved in 5 ml THF/MeOH (1/1) followed by addition of NaBH4 (250 mg, 6.60 mmol). The mixture was stirred at RT for 1 h. H2O was added and the mixture extracted with ethyl acetate, dried over Na2SO4, filtered and concentrated in vacuo. Preparative TLC gave the yellow solid 100 mg. to give (1-(6-bromo-2-chloropyridin-3-yl)cyclopropyl)methanol (100 mg, yield: 12%) MS (M+H)+: 263.


Step 5—Synthesis of 5-(6-chloro-5-(1-(hydroxymethyl)cyclopropyl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a solution of (1-(6-bromo-2-chloropyridin-3-yl)cyclopropyl)methanol (100 mg, 0.381 mmol) and (2-(4-fluorophenyl)-3-(methylcarbamoyl)-6-(N-methylmethylsulfonamido)benzofuran-5-yl)boronic acid (160 mg, 0.381 mmol) in 1,4-Dioxane (2 mL) and water (20 μL) was added K2CO3 (105 mg, 0.762 mmol) and Pd(dppf)Cl2 (55 mg, 0.075 mmol). The mixture was heated to 70° C. and stirred overnight. Filtration through a pad of celite removed the solid. After washing with ethyl acetate, the combined filtrate was concentrated in vacuo. Preparative TLC gave 5-(6-chloro-5-(1-(hydroxymethyl)cyclopropyl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (80 mg, yield: 37.6%) MS (M+H)+: 559.


Step 6—Synthesis of 5-(11′-fluoro-6′H-spiro[cyclopropane-1,5′-indolo[1,2-h][1,7]naphthyridin]-2′-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a solution of 5-(6-chloro-5-(1-(hydroxymethyl)cyclopropyl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (80 mg, 0.143 mmol) and 4-fluoro-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indole (56.1 mg, 0.215 mmol) in 1,4-Dioxane (2 mL) and water (20 μL) was added Cs2CO3 (93 mg, 0.287 mmol) and DTBPF PdCl2 (21 mg, 0.032 mmol). The mixture was heated to 80° C., stirred overnight, concentrated in vacuo and dried. The residue was dissolved in DCM (2.00 mL), followed by addition of triphenylphosphine (94 mg, 0.358 mmol) and DIAD (0.070 mL, 0.358 mmol). The mixture was stirred at RT for 4 h. Preparative TLC (eluted with 20% ethyl acetate in DCM) gave 5-(11′-fluoro-6′H-spiro[cyclopropane-1,5′-indolo[1,2-h][1,7]naphthyridin]-2′-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (4.5 mg, yield: 5%) MS (M+H)+: 639.


Example 98



embedded image


Step 1—Synthesis of 5-(5-allyl-6-chloropyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a degassed solution of chloride (250 mg, 1.08 mmol), boronic ester (551 mg, 1.08 mmol) and Cs2CO3 (701 mg, 2.15 mmol) in 1,4-dioxane (5 mL) and H2O (0.25 mL) was added 1,1′-bis(di-tert-butylphosphino)ferrocene palladium dichloride (70 mg, 0.11 mmol) under nitrogen atmosphere. The reaction mixture was stirred at 80° C. for 2.5 h, then cooled, filtered to remove inorganic solids and concentrated. The residue was purified on silica gel eluting with 0-100% acetone/hexane to give 5-(5-allyl-6-chloropyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (510 mg, 90% yield). MS (M+H)+: 527.


Step 2—Synthesis of 1-allyl-4-fluoro-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indole



embedded image


The indole (500 mg, 1.92 mmol) was added to a slurry of NaH (115 mg, 2.87 mmol, 60%) in DMF (6 mL) and cooled to 7° C. Allyl bromide (1.65 mL, 19.15) was added and the grey slurry was aged in ice bath for 1 hr. The mixture was warmed to room temperature and then another 0.5 mL allyl bromide and 188 mg NaH were added. The reaction was quenched with water and EtOAc, then a few drops 2N HCl was added until the solution was pH=10. The organic was washed with brine, dried over MgSO4 and concentrated to oil. The oil was purified on silica gel eluting with 0-40% acetone/hexane and isolated 1-allyl-4-fluoro-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indole (576 mg, 10% yield).


Step 3—Synthesis of 5-(5-allyl-6-(1-allyl-4-fluoro-1H-indol-3-yl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a degassed solution of chloride (268 mg, 0.51 mmol), boronic ester (153 mg, 0.51 mmol) and Cs2CO3 (331 mg, 1.02 mmol) in 1,4-dioxane (5 mL) and H2O (0.25 mL) was added 1,1′-bis(di-tert-butylphosphino)ferrocene palladium dichloride (33 mg, 0.051 mmol) under nitrogen atmosphere. The reaction mixture was stirred at 70° C. for 2 h, then cooled and concentrated. The mixture was redissolved in DCM, washed with water and purified on silica gel eluting with 0-40% acetone/hexane to give 5-(5-allyl-6-(1-allyl-4-fluoro-1H-indol-3-yl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (180 mg, 53% yield). MS (M+H)+: 666.


Step 4—Synthesis of 5-(12-fluoro-7H-pyrido[2′,3′:3,4]azepino[1,2-a]indol-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


Benzylidene-bis-(tricyclohexylphosphino)dichlororuthenium (1.2 mg, 1.5 umol) was dissolved in 1 mL DCM, degassed with subsurface N2 sparge and added by syringe to a similarly degassed 1 mL DCM solution of the diallyl compound and aged for 16 hours. The reaction was purified on a prep plate with 60% EtOAc/hexane to obtain 5-(12-fluoro-7H-pyrido[2′,3′:3,4]azepino[1,2-a]indol-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (7 mg, 37% yield). MS (M+H)+: 624.


Example 99



embedded image


Step 1—Synthesis of (E)-3-(4-((tert-butyldimethylsilyl)oxy)but-1-en-1-yl)-2,6-dichloropyridine



embedded image


To a degassed solution of iodide (2.0 g, 7.30 mmol), boronic ester (2.70 mL, 7.30 mmol) and 1,1′-bis(di-tert-butylphosphino)ferrocene palladium dichloride (238 mg, 0.365 mmol) was added degassed aq. K3PO4 (22 mL, 22.0 mmol, 1M). The reaction mixture was stirred at room temperature for 16 h. The mixture was concentrated, diluted with EtOAc and the aqueous layer was cut. The organic layer was dried over Mg2SO4 and concentrated to a black oil. The mixture was purified on silica with DCM to obtain (E)-3-(4-((tert-butyldimethylsilyl)oxy)but-1-en-1-yl)-2,6-dichloropyridine (2.5 g, 100% yield). MS (M+H)+: 331.


Step 2—Synthesis of 4-(2,6-dichloropyridin-3-yl)butan-1-ol



embedded image


The alkene (100 mg, 0.301 mmol) was dissolved in MeOH (5 mL). The flask was evacuated and backfilled with N2 three times. Pt/C (12 mg, 10 wt %) was added and the flask was evacuated, backfilled with N2 three times, then evacuated and backfilled with H2 from a balloon and aged for 2 hours. The mixture was filtered over celite, washed with methanol, and then most of the methanol was removed. HCl (1 mL, 4M in IPA) was added and the yellow solution was aged 10 minutes and then concentrated to give 4-(2,6-dichloropyridin-3-yl)butan-1-ol (45 mg, 68% yield). MS (M+H)+: 219.


Step 3—Synthesis of 5-(6-chloro-5-(4-hydroxybutyl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


A degassed solution of chloride (45 mg, 0.175 mmol), boronic ester (106 mg, 0.201 mmol) Cs2CO3 (229 mg, 0.702 mmol) and Xphos pre-catalyst (16 mg, 0.018 mmol) in 1,4-dioxane (2 mL) and H2O (0.25 mL) was heated at 70° C. for 16 h. Upon cooling the aqueous portion was removed and the organic portion was concentrated and purified on silica gel eluting with 0-10% MeOH/DCM to give 5-(6-chloro-5-(4-hydroxybutyl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (79 mg, 80% yield). MS (M+H)+: 559.


Step 4—Synthesis of 5-(6-(4-fluoro-1H-indol-2-yl)-5-(4-hydroxybutyl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


To a degassed solution of chloride (74 mg, 0.13 mmol), boronic ester (35 mg, 0.13 mmol) and Cs2CO3 (86 mg, 0.26 mmol) in 1,4-dioxane (5 mL) and H2O (0.25 mL) was added 1,1′-bis(di-tert-butylphosphino)ferrocene palladium dichloride (9 mg, 0.013 mmol) under nitrogen atmosphere. The reaction mixture was stirred at 70° C. for 2 h, at which point another 8 mg catalyst and 34 mg boronate was added and the reaction aged at 70° C. for 16 h. The mixture was cooled, diluted with EtOAc, washed with water, dried over MgSO4 and purified on silica gel eluting with 0-5% MeOH/DCM to give 5-(6-(4-fluoro-1H-indol-2-yl)-5-(4-hydroxybutyl)pyridin-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (45 mg, 52% yield). MS (M+H)+: 658.


Step 5—Synthesis of 5-(13-fluoro-5,6,7,8-tetrahydropyrido[2′,3′:3,4]azocino[1,2-a]indol-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide



embedded image


The alcohol (44 mg, 0.067 mmol) was dissolved in THF (2 mL) and then PPh3 (18 mg, 0.067) and DIAD (14 mg, 0.067 mmol) were added and the mixture heated to 50° C. for 16 hours. Additional PPh3 (18 mg, 0.067) and DIAD (28 mg, 0.134 mmol) were added in two more increments to get complete conversion. The reaction was then concentrated to oil and purified on silica eluting with 0-5-15% EtOAc/DCM, followed by a prep plate run in 25% acetone/hexane to obtain 5-(13-fluoro-5,6,7,8-tetrahydropyrido[2′,3′:3,4]azocino[1,2-a]indol-2-yl)-2-(4-fluorophenyl)-N-methyl-6-(N-methylmethylsulfonamido)benzofuran-3-carboxamide (20 mg, 46% yield). MS (M+H)+: 640.


Example 100
Measuring Compound Inhibitory Potency

Measurement of inhibition by compounds was performed using the HCV replicon system. Several different replicons encoding different HCV genotypes or mutations were used. In addition, potency measurements were made using different formats of the replicon assay, including different ways of measurements and different plating formats. See Jan M. Vrolijk et al., A replicons-based bioassay for the measurement of interferons in patients with chronic hepatitis C, 110 J. VIROLOGICAL METHODS 201 (2003); Steven S. Carroll et al., Inhibition of Hepatitis C Virus RNA Replication by 2′-Modified Nucleoside Analogs, 278 (14) J. BIOLOGICAL CHEMISTRY 11979 (2003). However, the underlying principles are common to all of these determinations, and are outlined below.


Stable neomycin phosphotransferase encoding replicons-harboring cell lines were used, so all cell lines were maintained under G418 selection prior to the assay. Potency was deteremined using a cell ELISA assay with an antibody to the replicons encoded NS3/4a protease. See Caterina Trozzi et al., In Vitro Selection and Characterization of Hepatitis C Virus Serine Protease Variants Resistant to an Active-Site Peptide Inhibitor, 77 (6) J. Virol. 3669 (2003). To initiate an assay, replicon cells were plated in the presence of a dilution series of test compound in the absence of G418. Typically, the assays were performed in a 96-well plate formate for manual operation, or a 384-well plate format for automated assay. Replicon cells and compound were incubated for 96 hours. At the end of the assay, cells were washed free of media and compound, and the cells were then lysed. RNA was quantified indirectly through detection of replicon-encoded NS3/4A protein levels, through an ELISA-based assay with an antibody specific for NS3/4A. IC50 determinations were calculated as a percentage of a DMSO control by fitting the data to a four-parameter fit function and the data obtained for genotypes 1a and 1b using this method is provided in the table below.











TABLE 1





Compound
1a
1b


No.
IC50 (nM)
IC50 (nM)

















1
3.784
4.674


2
2.093
3.902


3
7.944
6.694


4
6.989
7.297


5
3.859
2.299


6
16.8
10.16


7
17.61
6.785


8
580.5
121.6


9
38.18
31.24


10
9.437
10.66


11
2121
661.1


12
4.59
10.64


13
17.27
16.5


14
26.39
24.33


15
3.409
5.087


16
27.02
14.6


17
372.6
197.4


18
5479
296


19
56.94
52.33


20
680.5
129.3


21
149.2
34.33


22
1454
162.7


23
212.3
83.2


24
19.97
18.86


25
287.2
99.41


26
69.35
84.39


27
9.129
11.1


29
13.54
15.49


30
2.489
3.805


31
4.534
4.642


32
10.55
3.952


33
170.5
23.61


34
5.385
4.657


35
3.28
5.194


36
10.33
10.25


37
9.827
14.23


38
21.82
17.03


39
2.71
2.83


40
16.56
17.07


41
19.27
6.325


42
3.101
5.474


43
1.735
3.701


44
1.765
1.867


45
4.288
8.718


46
2.232
2.401


47
14.54
8.465


48
1.716
2.642


50
1.154
3.36


51
4.49
3.424


52
3.346
2.319


53
8.069
4.028


54
1.56
2.25


55
0.9708
1.806


56
3.404
1.997


57
8.158
3.392


58
1.52
1.706


59
5.446
2.904


60
5
10.11


61
2.275
7.801


62
2.687
2.548


63
4.336
3.281


64
23.21
9.086


65
3.622
3.957


66
13.02
11.44


67
12.48
16.38


68
70.9
60.62


69
2.298
2.949


70
55.35
79.95


71
41.69
42.2


72
3.71
4.295


73
2.014
2.777


74
8.198
11.45


75
46.66
43.17


76
999.6
193.6


77
27.94
4.025


78
206.3
22.05


79
93.78
77.28


80
27.71
9.773


81
196.3
33.86


82
2.885
5.542


83
1.488
3.439


84
4.283
6.407


85
2.993
3.287


86
1.907
3.053


87
7.689
7.286


88
8.728
9.433


89
2.12
5.486


90
7.234
2.787


91
27.551
9.23


92
46.471
2.55


94
34
10.84


95
8.911
7.525


96
7.893
3.179


97
10.73
9.982


98
4.173
8.646


99
6.498
7.168









It will be appreciated that various of the above-discussed and other features and functions, or alternatives thereof, may be desirably combined into many other different systems or applications. Also that various presently unforeseen or unanticipated alternatives, modifications, variations or improvements therein may be subsequently made by those skilled in the art which are also intended to be encompassed by the following claims.

Claims
  • 1. A compound having structural formula I:
  • 2. The compound of claim 1, wherein R2 and R4 are C1-C6 alkyl.
  • 3. The compound of claim 1 or 2, wherein R2, R3 and R4 are methyl.
  • 4. The compound of any one of claims 1 to 3, wherein one of Ra and R8 is hydrogen.
  • 5. The compound of any one of claims 1 to 4, wherein halo is F.
  • 6. The compound of claim 1, or a pharmaceutically acceptable salt thereof, having the formula:
  • 7. The compound of claim 6 wherein R5 is hydrogen or methyl;R6 is hydrogen, methyl, —CH2OH, —CH2CH2OH, —CH2SO2CH3,
  • 8. The compound of any one of claims 1 to 7, wherein A is C3-C6 cycloalkyl, or a 5-6 membered aromatic monocyclic ring with 0, 1, 2 or 3 heteroatom ring atoms selected from N and S, optionally substituted with 1 or 2 substituents independently selected from C1-C6 alkyl, C1-C6 alkoxy, C1-C6 haloalkyl, oxo, halo, and —O—C1-C6 haloalkyl.
  • 9. The compound of any one of claims 1 to 7, wherein A is cyclopropyl,
  • 10. The compound of claim 9, wherein A is
  • 11. The compound of claim 1 which is any one of
  • 12. The compound of claim 1 which is any one of
  • 13. A pharmaceutical composition comprising (i) a pharmaceutically acceptable carrier and (ii) an effective amount of the compound of any one of claims 1-12 or a pharmaceutically acceptable salt thereof.
  • 14. The pharmaceutical composition of claim 13, further comprising a second therapeutic agent selected from the group consisting of HCV antiviral agents, immunomodulators, and anti-infective agents.
  • 15. The pharmaceutical composition of claim 14, wherein the second therapeutic agent is selected from the group consisting of HCV NS3 and NS3/4A protease inhibitors, HCV NS5A inhibitors and HCV NS5B polymerase inhibitors.
  • 16. A method of treating a patient infected with HCV, the method comprising administering to the patient the compound of any one of claims 1 to 12, or a pharmaceutically acceptable salt thereof, in an amount effective to treat infection by HCV in the patient.
  • 17. The method of claim 16, further comprising administering to said patient an effective amount of at least one second therapeutic agent selected from the group consisting of HCV NS3 and NS3/4A protease inhibitors, HCV NS5A inhibitors and HCV NS5B polymerase inhibitors.
Priority Claims (1)
Number Date Country Kind
PCT/CN2013/000128 Feb 2013 CN national
PCT Information
Filing Document Filing Date Country Kind
PCT/US2014/014361 2/3/2014 WO 00
Publishing Document Publishing Date Country Kind
WO2014/123793 8/14/2014 WO A
US Referenced Citations (14)
Number Name Date Kind
6777392 Maurya et al. Aug 2004 B2
7105499 Carroll et al. Sep 2006 B2
7112600 Hashimoto et al. Sep 2006 B1
7125855 Bhat et al. Oct 2006 B2
7202224 Eldrup et al. Apr 2007 B2
20030203948 Fujishita et al. Oct 2003 A1
20040067901 Bhat et al. Apr 2004 A1
20040110717 Carroll et al. Jun 2004 A1
20060100262 Conte et al. May 2006 A1
20080045498 Griffith et al. Feb 2008 A1
20090048239 Conte et al. Feb 2009 A1
20100093694 Yeung et al. Apr 2010 A1
20110256099 Yeung et al. Oct 2011 A1
20120328569 McComas et al. Dec 2012 A1
Foreign Referenced Citations (25)
Number Date Country
WO0147883 Jul 2001 WO
WO0177091 Oct 2001 WO
WO0204425 Jan 2002 WO
WO0206246 Jan 2002 WO
WO0220497 Mar 2002 WO
WO0257287 Jul 2002 WO
WO0257425 Jul 2002 WO
WO03068244 Aug 2003 WO
WO2004000858 Dec 2003 WO
WO2004003138 Jan 2004 WO
WO2004007512 Jan 2004 WO
WO2004041201 May 2004 WO
WO2005003147 Jan 2005 WO
WO2005016927 Feb 2005 WO
WO2006020082 Feb 2006 WO
WO2006066079 Jun 2006 WO
WO2006066080 Jun 2006 WO
WO2008075103 Jun 2008 WO
WO2009010783 Jan 2009 WO
WO2009010785 Jan 2009 WO
WO2011106992 Sep 2011 WO
WO2013033971 Mar 2013 WO
WO2013034048 Mar 2013 WO
WO2014123794 Aug 2014 WO
WO2014123795 Aug 2014 WO
Non-Patent Literature Citations (1)
Entry
Carroll et al., Inhibition of Hepatitis C Virus RNA Replication by 2′-Modified Nucleoside Analogs, J. Biol. Chem., 2003, 11979-11984, 278(14).
Related Publications (1)
Number Date Country
20150361101 A1 Dec 2015 US